438
Views
16
CrossRef citations to date
0
Altmetric
Review

Optimising use of thiopurines in inflammatory bowel disease

&
Pages 877-888 | Received 21 Feb 2017, Accepted 03 Jul 2017, Published online: 12 Jul 2017

References

  • Edwards FC, Truelove SC. The course and prognosis of ulcerative colitis. Gut. 1963;4:299–315.
  • Gomollon F, Dignass A, Annese V, et al. 3rd European evidence-based consensus on the diagnosis and management of Crohn’s disease 2016: part 1: diagnosis and medical management. J Crohn’s Colitis. 2017;11:3–25.
  • Dignass A, Lindsay JO, Sturm A, et al. Second European evidence-based consensus on the diagnosis and management of ulcerative colitis part 2: current management. J Crohn’s Colitis. 2012;6:991–1030.
  • D’Haens GR, Fedorak R, Lemann M, et al. Endpoints for clinical trials evaluating disease modification and structural damage in adults with Crohn’s disease. Inflamm Bowel Dis. 2009;15:1599–1604.
  • Loftus EV Jr. Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences. Gastroenterology. 2004;126:1504–1517.
  • Gisbert JP, Panes J. Loss of response and requirement of infliximab dose intensification in Crohn’s disease: a review. Am J Gastroenterol. 2009;104:760–767.
  • Elion G. The purine path to chemotherapy. Science. 1989;244:41–47.
  • Bean RH. The treatment of chronic ulcerative colitis with 6-mercaptopurine. Med J Aust. 1962;49(2):592–593.
  • Present DH, Korelitz BI, Wisch N, et al. Treatment of Crohn’s disease with 6-mercaptopurine. New England J Med. 1980;302:981–987.
  • Jewell DP, Truelove SC Azathioprine in ulcerative colitis: final report on controlled therapeutic trial. British Medical J. 1974;4:627–630.
  • Rutgeerts PJ. An historical overview of the treatment of Crohn’s disease: why do we need biological therapies? Rev Gastroenterol Disord. 2004;4(Suppl 3):S3–S9.
  • Timmer A, Patton PH, Chande N, et al. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev. 2016;18:CD000478.
  • Chande N, Townsend CM, Parker CE, et al. Azathioprine or 6-mercaptopurine for induction of remission in Crohn’s disease. Cochrane Database Syst Rev. 2016;10:CD000545.
  • Panaccione R, Ghosh S, Middleton S, et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology. 2014;146:392–400.
  • Mantzaris GJ, Christidou A, Sfakianakis M, et al. Azathioprine is superior to budesonide in achieving and maintaining mucosal healing and histologic remission in steroid-dependent Crohn’s disease. Inflamm Bowel Dis. 2009;15:375–382.
  • Colombel JF, Sandborn WJ, Reinisch W, et al. Infliximab, azathioprine, or combination therapy for Crohn’s Disease. New England J Med. 2010;362:1383–1395.
  • Mao EJ, Hazlewood GS, Kaplan GG, et al. Systematic review with meta-analysis: comparative efficacy of immunosuppressants and biologics for reducing hospitalisation and surgery in Crohn’s disease and ulcerative colitis. Aliment Pharmacol Ther. 2017;45:3–13.
  • Chhaya V, Pollok RC, Cecil E, et al. Impact of early thiopurines on surgery in 2770 children and young people diagnosed with inflammatory bowel disease: a national population-based study. Aliment Pharmacol Ther. 2015;42:990–999.
  • Ramadas AV, Gunesh S, Thomas GA, et al. Natural history of Crohn’s disease in a population-based cohort from Cardiff (1986-2003): a study of changes in medical treatment and surgical resection rates. Gut. 2010;59:1200–1206.
  • Fasanmade AA, Adedokun OJ, Ford J, et al. Population pharmacokinetic analysis of infliximab in patients with ulcerative colitis. Eur J Clin Pharmacol. 2009;65:1211–1228.
  • Roblin X, Williet N, Peyrin-Biroulet L. Thiopurine metabolism in the era of combotherapy. Inflamm Bowel Dis. 2016;22:1496–1501.
  • Matsumoto T, Motoya S, Watanabe K, et al. Adalimumab monotherapy and a combination with azathioprine for Crohn’s disease: a prospective, randomized trial. J Crohn’s Colitis. 2016;10:1259–1266.
  • Panes J, Lopez-Sanroman A, Bermejo F, et al. Early azathioprine therapy is no more effective than placebo for newly diagnosed Crohn’s disease. Gastroenterology. 2013;145:766–774.
  • Cosnes J, Bourrier A, Laharie D, et al. Early administration of azathioprine vs conventional management of Crohn’s disease: a randomized controlled trial. Gastroenterology. 2013;145:758–765.
  • Markowitz J, Grancher K, Kohn N, et al. A multicenter trial of 6-mercaptopurine and prednisone in children with newly diagnosed Crohn’s disease. Gastroenterology. 2000;119:895–902.
  • Lakatos PL, Golovics PA, David G, et al. Has there been a change in the natural history of Crohn’s disease? Surgical rates and medical management in a population-based inception cohort from Western Hungary between 1977-2009. Am J Gastroenterol. 2012;107:579–588.
  • Chatu S, Subramanian V, Saxena S, et al. The role of thiopurines in reducing the need for surgical resection in Crohn’s disease: a systematic review and meta-analysis. Am J Gastroenterol. 2014;109:23–34.
  • Chatu S, Saxena S, Subramanian V, et al. The impact of timing and duration of thiopurine treatment on first intestinal resection in Crohn’s disease: national UK population-based study 1989-2010. Am J Gastroenterol. 2014;109:409–416.
  • Kariyawasam VC, Selinger CP, Katelaris PH, et al. Early use of thiopurines or methotrexate reduces major abdominal and perianal surgery in Crohn’s disease. Inflamm Bowel Dis. 2014;20:1382–1390.
  • Burisch J, Pedersen N, Cukovic-Cavka S, et al. Initial disease course and treatment in an inflammatory bowel disease inception cohort in Europe: the ECCO-EpiCom cohort. Inflamm Bowel Dis. 2014;20:36–46.
  • Blaker PA, Arenas-Hernandez M, Marinaki AM, et al. The pharmacogenetic basis of individual variation in thiopurine metabolism. Per Med. 2012;9:707–725.
  • McGovern DP, Travis SP, Duley J, et al. Azathioprine intolerance in patients with IBD may be imidazole-related and is independent of TPMT activity. Gastroenterology. 2002;122:838–839.
  • Irving PM. Can we get more from our current treatments? Best Pract Res Clin Gastroenterol. 2014;28:451–463.
  • Kostic AD, Xavier RJ, Gevers D. The microbiome in inflammatory bowel disease: current status and the future ahead. Gastroenterology. 2014;146:1489–1499.
  • Oancea I, Movva R, Das I, et al. Colonic microbiota can promote rapid local improvement of murine colitis by thioguanine independently of T lymphocytes and host metabolism. Gut. 2017;66:59–69.
  • Fairchild CR, Maybaum J, Kennedy KA. Concurrent unilateral chromatid damage and DNA strand breakage in response to 6-thioguanine treatment. Biochem Pharmacol. 1986;35:3533–3541.
  • Tiede I, Fritz G, Strand S, et al. CD28-dependent Rac1 activation is the molecular target of azathioprine in primary human CD4+ T lymphocytes. J Clin Invest. 2003;111:1133–1145.
  • Marinkovic G, Hamers AA, de Vries CJ, et al. 6-mercaptopurine reduces macrophage activation and gut epithelium proliferation through inhibition of GTPase Rac1. Inflamm Bowel Dis. 2014;20:1487–1495.
  • Dubinsky MC, Lamothe S, Yang HY, et al. Pharmacogenomics and metabolite measurement for 6-mercaptopurine therapy in inflammatory bowel disease. Gastroenterology. 2000;118:705–713.
  • Osterman MT, Kundu R, Lichtenstein GR, et al. Association of 6-thioguanine nucleotide levels and inflammatory bowel disease activity: a meta-analysis. Gastroenterology. 2006;130:1047–1053.
  • Moreau AC, Paul S, Del Tedesco E, et al. Association between 6-thioguanine nucleotides levels and clinical remission in inflammatory disease: a meta-analysis. Inflamm Bowel Dis. 2014;20:464–471.
  • Smith M, Blaker P, Patel C, et al. The impact of introducing thioguanine nucleotide monitoring into an inflammatory bowel disease clinic. Int J Clin Pract. 2013;67:161–169.
  • Wright S, Sanders DS, Lobo AJ, et al. Clinical significance of azathioprine active metabolite concentrations in inflammatory bowel disease. Gut. 2004;53:1123–1128.
  • Hindorf U, Lyrenas E, Nilsson A, et al. Monitoring of long-term thiopurine therapy among adults with inflammatory bowel disease. Scand J Gastroenterol. 2004;39:1105–1112.
  • Gupta P, Gokhale R, Kirschner BS. 6-mercaptopurine metabolite levels in children with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2001;33:450–454.
  • Roblin X, Serre-Debeauvais F, Phelip JM, et al. 6-tioguanine monitoring in steroid-dependent patients with inflammatory bowel diseases receiving azathioprine. Aliment Pharmacol Ther. 2005;21:829–839.
  • Goldberg R, Moore G, Cunningham G, et al. Thiopurine metabolite testing in inflammatory bowel disease. J Gastroenterol Hepatol. 2016;31:553–560. Epub 2015/10/30.
  • Kennedy NA, Asser TL, Mountifield RE, et al. Thiopurine metabolite measurement leads to changes in management of inflammatory bowel disease. Intern Med J. 2013;43:278–286.
  • Reinshagen M, Schutz E, Armstrong VW, et al. 6-thioguanine nucleotide-adapted azathioprine therapy does not lead to higher remission rates than standard therapy in chronic active Crohn disease: results from a randomized, controlled, open trial. Clin Chem. 2007;53:1306–1314.
  • Dassopoulos T, Dubinsky MC, Bentsen JL, et al. Randomised clinical trial: individualised vs. weight-based dosing of azathioprine in Crohn’s disease. Aliment Pharmacol Ther. 2014;39:163–175.
  • Thomas CW Jr., Lowry PW, Franklin CL, et al. Erythrocyte mean corpuscular volume as a surrogate marker for 6-thioguanine nucleotide concentration monitoring in patients with inflammatory bowel disease treated with azathioprine or 6-mercaptopurine. Inflamm Bowel Dis. 2003;9:237–245.
  • Decaux G, Prospert F, Horsmans Y, et al. Relationship between red cell mean corpuscular volume and 6-thioguanine nucleotides in patients treated with azathioprine. J Lab Clin Med. 2000;135:256–262.
  • Bouguen G, Sninsky C, Tang KL, et al. Change in erythrocyte mean corpuscular volume during combination therapy with azathioprine and infliximab is associated with mucosal healing: a post hoc analysis from SONIC. Inflamm Bowel Dis. 2015;21:606–614.
  • Yarur AJ, Kubiliun MJ, Czul F, et al. Concentrations of 6-thioguanine nucleotide correlate with trough levels of infliximab in patients with inflammatory bowel disease on combination therapy. Clinical Gastroenterol Hepatol. 2015;13(1118–24):e3.
  • Kariyawasam VC, Ward MG, Blaker PA, et al. 343 azathioprine decreases the risk of adalimumab primary non-response and secondary loss of response but only if adequately dosed. Gastroenterology. 2014;146:S–79.
  • Roblin X, Boschetti G, Williet N, et al. Azathioprine dose reduction in inflammatory bowel disease patients on combination therapy: an open-label, prospective and randomised clinical trial. Aliment Pharmacol Ther. 2017;46:142–149.
  • Nasuno M, Motoya S, Sugiyama K, et al. Low-dose azathioprine improves long-term efficacy of infliximab maintenance treatment in Japanese patients with Crohn’s disease. Gastroenterology. 2017;152:S408–S9.
  • Goodhand JR, Kamperidis N, Sirwan B, et al. Factors associated with thiopurine non-adherence in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2013;38:1097–1108.
  • Gearry RB, Barclay ML. Azathioprine and 6-mercaptopurine pharmacogenetics and metabolite monitoring in inflammatory bowel disease. J Gastroenterol Hepatol. 2005;20:1149–1157.
  • Smith MA, Blaker P, Marinaki AM, et al. Optimising outcome on thiopurines in inflammatory bowel disease by co-prescription of allopurinol. J Crohn’s Colitis. 2012;6:905–912.
  • Blaker PA, Arenas-Hernandez M, Smith MA, et al. Mechanism of allopurinol induced TPMT inhibition. Biochem Pharmacol. 2013;86:539–547.
  • Appell ML, Wagner A, Hindorf U. A skewed thiopurine metabolism is a common clinical phenomenon that can be successfully managed with a combination of low-dose azathioprine and allopurinol. J Crohn’s Colitis. 2013;7:510–513.
  • 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;22:441–446.
  • Sparrow MP, Hande SA, Friedman S, et al. Effect of allopurinol on clinical outcomes in inflammatory bowel disease nonresponders to azathioprine or 6-mercaptopurine. Clinical Gastroenterol Hepatol. 2007;5:209–214.
  • Leung Y, Sparrow MP, Schwartz M, et al. Long term efficacy and safety of allopurinol and azathioprine or 6-mercaptopurine in patients with inflammatory bowel disease. J Crohn’s Colitis. 2009;3:162–167.
  • Hoentjen F, Seinen ML, Hanauer SB, et al. Safety and effectiveness of long-term allopurinol-thiopurine maintenance treatment in inflammatory bowel disease. Inflamm Bowel Dis. 2013;19:363–369.
  • Hung S, Chung W, Chen Y. Genetics of severe drug hypersensitivity reactions in Han Chinese. In: Pichler WJ, editor. Drug hypersensitivity. Basel: Karger; 2007. p. 105–114.
  • Gearry RB, Barclay ML, Burt MJ, et al. Thiopurine drug adverse effects in a population of New Zealand patients with inflammatory bowel disease. Pharmacoepidemiol Drug Saf. 2004;13:563–567.
  • Teich N, Mohl W, Bokemeyer B, et al. Azathioprine-induced acute pancreatitis in patients with inflammatory bowel diseases–a prospective study on incidence and severity. J Crohn’s Colitis. 2016;10:61–68.
  • Ansari A, Patel N, Sanderson J, et al. Low-dose azathioprine or mercaptopurine in combination with allopurinol can bypass many adverse drug reactions in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2010;31:640–647.
  • Pavlidis P, Stamoulos P, Abdulrehman A, et al. Long-term safety and efficacy of low-dose azathioprine and allopurinol cotherapy in inflammatory bowel disease: a large observational study. Inflamm Bowel Dis. 2016;22:1639–1646.
  • 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;19:1404–1410.
  • Shih DQ, Nguyen M, Zheng L, et al. Split-dose administration of thiopurine drugs: a novel and effective strategy for managing preferential 6-MMP metabolism. Aliment Pharmacol Ther. 2012;36:449–458.
  • Egan LJ, Derijks LJJ, Hommes DW. Pharmacogenomics in inflammatory bowel disease. Clin Gastroenterol Hepatol. 2006;4:21–28.
  • Nagy F, Molnár T, Szepes Z, et al. Efficacy of 6-mercaptopurine treatment after azathioprine hypersensitivity in inflammatory bowel disease. World J Gastroenterol. 2008;14:4342–4346.
  • Amin J, Huang B, Yoon J, et al. Update 2014: advances to optimize 6-mercaptopurine and azathioprine to reduce toxicity and improve efficacy in the management of IBD. Inflamm Bowel Dis. 2015;21:445–452.
  • Hindorf U, Johansson M, Eriksson A, et al. Mercaptopurine treatment should be considered in azathioprine intolerant patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2009;29:654–661.
  • Miller NA, Farrow EG, Gibson M, et al. A 26-hour system of highly sensitive whole genome sequencing for emergency management of genetic diseases. Genome Med. 2015;7:100.
  • Appell ML, Berg J, Duley J, et al. Nomenclature for alleles of the thiopurine methyltransferase gene. Pharmacogenet Genomics. 2013;23:242–248.
  • Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics: monogenic inheritance of erythrocyte thiopurine methyltransferase activity. Am J Hum Genet. 1980;32:651–662.
  • Schaeffeler E, Fischer C, Brockmeier D, et al. Comprehensive analysis of thiopurine S-methyltransferase phenotype-genotype correlation in a large population of German-Caucasians and identification of novel TPMT variants. Pharmacogenetics. 2004;14:407–417.
  • Lennard L, Van Loon JA, Lilleyman JS, et al. Thiopurine pharmacogenetics in leukemia: correlation of erythrocyte thiopurine methyltransferase activity and 6-thioguanine nucleotide concentrations. Clin Pharmacol Ther. 1987;41:18–25.
  • Higgs JE, Payne K, Roberts C, et al. Are patients with intermediate TPMT activity at increased risk of myelosuppression when taking thiopurine medications? Pharmacogenomics. 2010;11:177–188.
  • Lennard L, Cartwright CS, Wade R, et al. Thiopurine methyltransferase genotype-phenotype discordance and thiopurine active metabolite formation in childhood acute lymphoblastic leukaemia. Br J Clin Pharmacol. 2013;76:125–136.
  • Hindorf U, Appell ML. Genotyping should be considered the primary choice for pre-treatment evaluation of thiopurine methyltransferase function. J Crohn’s Colitis. 2012;6:655–659.
  • Colombel JF, Ferrari N, Debuysere H, et al. Genotypic analysis of thiopurine S-methyltransferase in patients with Crohn’s disease and severe myelosuppression during azathioprine therapy. Gastroenterology. 2000;118:1025–1030.
  • Ansari A, Hassan C, Duley J, et al. Thiopurine methyltransferase activity and the use of azathioprine in inflammatory bowel disease. Aliment Pharmacol Ther. 2002;16:1743–1750.
  • Yang S-K, Hong M, Baek J, et al. A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet. 2014;46:1017–1020.
  • Hindorf U, Lindqvist M, Peterson C, et al. Pharmacogenetics during standardised initiation of thiopurine treatment in inflammatory bowel disease. Gut. 2006;55:1423–1431.
  • Seinen ML, van Asseldonk DP, Mulder CJ, et al. Dosing 6-thioguanine in inflammatory bowel disease: expert-based guidelines for daily practice. Journal Gastrointestinal Liver Diseases. 2010;19:291–294.
  • Kaskas BA, Louis E, Hindorf U, et al. Safe treatment of thiopurine S-methyltransferase deficient Crohn’s disease patients with azathioprine. Gut. 2003;52:140–142.
  • Relling MV, Gardner EE, Sandborn WJ, et al. Clinical pharmacogenetics implementation consortium guidelines for thiopurine methyltransferase genotype and thiopurine dosing. Clin Pharmacol Ther. 2011;89:387–391.
  • Fong SC, Blaker PA, Arenas-Hernandez M, et al. Getting the best out of thiopurine therapy: thiopurine S-methyltransferase and beyond. Biomark Med. 2015;9:51–65.
  • 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;24:331–342.
  • Rahier JF, Magro F, Abreu C, et al. Second European evidence-based consensus on the prevention, diagnosis and management of opportunistic infections in inflammatory bowel disease. J Crohn’s Colitis. 2014;8:443–468.
  • Candy S, Wright J, Gerber M, et al. A controlled double blind study of azathioprine in the management of Crohn’s disease. Gut. 1995;37:674–678.
  • Ardizzone S, Maconi G, Sampietro GM, et al. Azathioprine and mesalamine for prevention of relapse after conservative surgery for Crohn’s disease. Gastroenterology. 2004;127:730–740.
  • Jostins L, Ripke S, Weersma RK, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491:119–124.
  • Heap GA, Weedon MN, Bewshea CM, et al. HLA-DQA1-HLA-DRB1 variants confer susceptibility to pancreatitis induced by thiopurine immunosuppressants. Nat Genet. 2014;46:1131–1134.
  • de Boer NK, Derijks LJ, Gilissen LP, et al. On tolerability and safety of a maintenance treatment with 6-thioguanine in azathioprine or 6-mercaptopurine intolerant IBD patients. World J Gastroenterol. 2005;11:5540–5544.
  • Dubinsky MC, Vasiliauskas EA, Singh H, et al. 6-thioguanine can cause serious liver injury in inflammatory bowel disease patients. Gastroenterology. 2003;125:298–303.
  • Musumba CO. Review article: the association between nodular regenerative hyperplasia, inflammatory bowel disease and thiopurine therapy. Aliment Pharmacol Ther. 2013;38:1025–1037.
  • van Asseldonk DP, Jharap B, Verheij J, et al. The prevalence of nodular regenerative hyperplasia in inflammatory bowel disease patients treated with thioguanine is not associated with clinically significant liver disease. Inflamm Bowel Dis. 2016;22:2112–2120.
  • Pavlidis P, Ansari A, Duley J, et al. Splitting a therapeutic dose of thioguanine may avoid liver toxicity and be an efficacious treatment for severe inflammatory bowel disease: a 2-center observational cohort study. Inflamm Bowel Dis. 2014;20:2239–2246.
  • Zhong Z, Sanchez-Lopez E, Karin M. Autophagy, inflammation, and immunity: a troika governing cancer and its treatment. Cell. 2016;166:288–298.
  • Ariyaratnam J, Subramanian V. Association between thiopurine use and nonmelanoma skin cancers in patients with inflammatory bowel disease: a meta-analysis. Am J Gastroenterol. 2014;109:163–169.
  • 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;141:1621-28.e1-5.
  • Setshedi M, Epstein D, Winter TA, et al. Use of thiopurines in the treatment of inflammatory bowel disease is associated with an increased risk of non-melanoma skin cancer in an at-risk population: a cohort study. J Gastroenterol Hepatol. 2012;27:385–389.
  • Annese V, Beaugerie L, Egan L, et al. European evidence-based consensus: inflammatory bowel disease and malignancies. J Crohn’s Colitis. 2015;9:945–965.
  • Beaugerie L, Brousse N, Bouvier AM, et al. Lymphoproliferative disorders in patients receiving thiopurines for inflammatory bowel disease: a prospective observational cohort study. The Lancet. 2009;374:1617–1625.
  • 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;43:252–261.
  • Beigel F, Steinborn A, Schnitzler F, et al. Risk of malignancies in patients with inflammatory bowel disease treated with thiopurines or anti-TNF alpha antibodies. Pharmacoepidemiol Drug Saf. 2014;23:735–744.
  • Posthuma EF, Westendorp RG, van der Sluys Veer A, et al. Fatal infectious mononucleosis: a severe complication in the treatment of Crohn’s disease with azathioprine. Gut. 1995;36:311–313.
  • Louis E, Irving P, Beaugerie L. Use of azathioprine in IBD: modern aspects of an old drug. Gut. 2014;63:1695–1699.
  • Green J, Kirchgesner J, Beaugerie L, et al. Impact on life expectancy of withdrawing thiopurines in patients with Crohn’s disease in sustained clinical remission: a lifetime risk-benefit analysis. PLoS One. 2016;11:e0157191.
  • Beaugerie L, Carrat F, Colombel JF, et al. Risk of new or recurrent cancer under immunosuppressive therapy in patients with IBD and previous cancer. Gut. 2014;63:1416–1423.
  • Shelton E, Laharie D, Scott FI, et al. Cancer recurrence following immune-suppressive therapies in patients with immune-mediated diseases: a systematic review and meta-analysis. Gastroenterology. 2016;151:97–109.
  • Aberra FN, Lewis JD, Hass D, et al. Corticosteroids and immunomodulators: postoperative infectious complication risk in inflammatory bowel disease patients. Gastroenterology. 2003;125:320–327.
  • Colombel JF, Loftus EV Jr., Tremaine WJ, et al. Early postoperative complications are not increased in patients with Crohn’s disease treated perioperatively with infliximab or immunosuppressive therapy. Am J Gastroenterol. 2004;99:878–883.
  • Myrelid P, Olaison G, Sjodahl R, et al. Thiopurine therapy is associated with postoperative intra-abdominal septic complications in abdominal surgery for Crohn’s disease. Dis Colon Rectum. 2009;52:1387–1394.
  • Patel KV, Darakhshan AA, Griffin N, et al. Patient optimization for surgery relating to Crohn’s disease. Nat Rev Gastroenterol Hepatol. 2016;13:707–719.
  • De Cruz P, Kamm MA, Hamilton AL, et al. Crohn’s disease management after intestinal resection: a randomised trial. Lancet (London, England). 2015;385:1406–1417.
  • OP026. The TOPPIC trial: a randomised, double-blind parallel-group trial of mercaptopurine versus placebo to prevent recurrence of Crohn’s disease following surgical resection in 240 patients. J Crohn’s Colitis. 2016;10:S21–S22.
  • Nguyen GC, Seow CH, Maxwell C, et al. The Toronto consensus statements for the management of inflammatory bowel disease in pregnancy. Gastroenterology. 2016;150:734–757.
  • Angelberger S, Reinisch W, Messerschmidt A, et al. Long-term follow-up of babies exposed to azathioprine in utero and via breastfeeding. J Crohn’s Colitis. 2011;5:95–100.
  • Sheikh M, Nelson-Piercy C, Duley J, et al. Successful pregnancies with thiopurine-allopurinol co-therapy for inflammatory bowel disease. J Crohn’s Colitis. 2015;9:680–684.
  • Wong DR, Coenen MJ, Derijks LJ, et al. Early prediction of thiopurine-induced hepatotoxicity in inflammatory bowel disease. Aliment Pharmacol Ther. 2016;45:391–402.
  • Wong DR, Coenen MJ, Vermeulen SH, et al. Early assessment of thiopurine metabolites identifies patients at risk of thiopurine-induced leukopenia in inflammatory bowel disease. J Crohn’s Colitis. 2016;11:175–184.
  • Kennedy NA, Kalla R, Warner B, et al. Thiopurine withdrawal during sustained clinical remission in inflammatory bowel disease: relapse and recapture rates, with predictive factors in 237 patients. Aliment Pharmacol Ther. 2014;40:1313–1323.
  • Lemann M, Mary JY, Colombel JF, et al. A randomized, double-blind, controlled withdrawal trial in Crohn’s disease patients in long-term remission on azathioprine. Gastroenterology. 2005;128:1812–1818.
  • Treton X, Bouhnik Y, Mary JY, et al. Azathioprine withdrawal in patients with Crohn’s disease maintained on prolonged remission: a high risk of relapse. Clinical Gastroenterol Hepatol. 2009;7:80–85.
  • Rosario M, Dirks NL, Gastonguay MR, et al. Population pharmacokinetics-pharmacodynamics of vedolizumab in patients with ulcerative colitis and Crohn’s disease. Aliment Pharmacol Ther. 2015;42:188–202.

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.