123
Views
14
CrossRef citations to date
0
Altmetric
Drug Profile

Azathioprine in inflammatory bowel disease: improved molecular insights and resulting clinical implications

&
Pages 23-34 | Published online: 10 Jan 2014

References

  • Glickmann RM. Inflammatory bowel disease.In: Ulcerative Colitis and Crohn’s disease. Fauci AS, Braunwald E, Isselbacher KJ et al. (Eds). McGraw–Hill, NY, USA, 1969–1974 (1991).
  • Buhner S, Buning C, Genschel J et al. Genetic basis for increased intestinal permeability in families with Crohn’s disease: role of CARD15 3020insC mutation? Gut55, 342–347 (2006).
  • Hugot JP. CARD15/NOD2 mutations in Crohn’s disease. Ann. NY Acad. Sci.1072, 9–18 (2006).
  • Koutroubakis I, Manousos ON, Meuwissen SG et al. Environmental risk factors in inflammatory bowel disease. Hepatogastroenterology43, 381–393 (1996).
  • Neurath MF, Schurmann G. Immunopathogenesis of inflammatory bowel diseases. Chirurg71, 30–40 (2000).
  • Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J. Clin. Invest.117, 514–521 (2007).
  • Carter MJ, Lobo AJ, Travis SP. Guidelines for the management of inflammatory bowel disease in adults. Gut53(Suppl. 5), V1–V16 (2004).
  • Targan SR, Deem RL, Liu M et al. Definition of a lamina propria T cell responsive state. Enhanced cytokine responsiveness of T cells stimulated through the CD2 pathway. J. Immunol.154, 664–675 (1995).
  • Allez M, Mayer L. Regulatory T cells: peace keepers in the gut. Inflamm. Bowel Dis.10, 666–676 (2004).
  • Braunstein J, Qiao L, Autschbach F et al. T cells of the human intestinal lamina propria are high producers of interleukin-10. Gut41, 215–220 (1997).
  • Neurath MF, Finotto S, Fuss I et al. Regulation of T-cell apoptosis in inflammatory bowel disease: to die or not to die, that is the mucosal question. Trends Immunol.22, 21–26 (2001).
  • Faubion WA Jr, Loftus EV Jr, Harmsen WS et al. The natural history of corticosteroid therapy for inflammatory bowel disease: a population-based study. Gastroenterology121, 255–260 (2001).
  • Nguyen GC, Harris ML, Dassopoulos T. Insights in immunomodulatory therapies for ulcerative colitis and Crohn’s disease. Curr. Gastroenterol. Rep.8, 499–505 (2006).
  • Sandborn WJ. Azathioprine: state of the art in inflammatory bowel disease. Scand. J. Gastroenterol.225, 92–99 (1998).
  • Bean RH. The treatment of chronic ulcerative colitis with 6-mercaptopurine. Med. J. Aust.49(2), 592–593 (1962).
  • Ewe K, Press AG, Singe CC et al. Azathioprine combined with prednisolone or monotherapy with prednisolone in active Crohn’s disease. Gastroenterology105, 367–372 (1993).
  • Hawthorne AB, Logan RF, Hawkey CJ et al. Randomised controlled trial of azathioprine withdrawal in ulcerative colitis. BMJ305, 20–22 (1992).
  • Kirk AP, Lennard-Jones JE. Controlled trial of azathioprine in chronic ulcerative colitis. Br. Med. J. (Clin. Res. Ed.)284, 1291–1292 (1982).
  • O’Donoghue DP, Dawson A, Powell-Tuck J et al. Double-blind withdrawal trial of azathioprine as maintenance treatment for Crohn’s disease. Lancet2, 955–957 (1978).
  • Pearson DC, May GR, Fick GH et al. Azathioprine and 6-mercaptopurine in Crohn disease. A meta-analysis. Ann. Intern. Med.123, 132–142 (1995).
  • Sandborn W, Sutherland L, Pearson D et al. Azathioprine or 6-mercaptopurine for inducing remission of Crohn’s disease. Cochrane Database Syst. Rev. CD000545 (2000).
  • Tanis AA. Azathioprine in inflammatory bowel disease, a safe alternative? >Mediators Inflamm.7, 141–144 (1998).
  • van Hogezand RA. Medical management of patients with difficult-to-treat inflammatory bowel disease. Neth. J. Med.45, 55–59 (1994).
  • Holtmann MH, Neurath MF. From immunogenic mechanisms to novel therapeutic approaches in inflammatory bowel disease. Adv. Exp. Med. Biol.579, 227–242 (2006).
  • Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. Lancet369, 1641–1657 (2007).
  • Sandborn WJ, Hanauer S, Loftus EV Jr et al. An open-label study of the human anti-TNF monoclonal antibody adalimumab in subjects with prior loss of response or intolerance to infliximab for Crohn’s disease. Am. J. Gastroenterol.99, 1984–1989 (2004).
  • Targan SR, Hanauer SB, van Deventer SJ et al. A short-term study of chimeric monoclonal antibody cA2 to tumor necrosis factor α for Crohn’s disease.Crohn’s Disease cA2 Study Group. N. Engl. J. Med.337, 1029–1035 (1997).
  • Atreya R, Mudter J, Finotto S et al. Blockade of interleukin 6 trans signaling suppresses T-cell resistance against apoptosis in chronic intestinal inflammation: evidence in crohn disease and experimental colitis in vivo. Nat. Med.6, 583–588 (2000).
  • Ito H, Takazoe M, Fukuda Y et al. A pilot randomized trial of a human anti-interleukin-6 receptor monoclonal antibody in active Crohn’s disease. Gastroenterology126, 989–996 (2004).
  • 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.12, 558–565 (2006).
  • Mannon PJ, Fuss IJ, Mayer L et al. Anti-interleukin-12 antibody for active Crohn’s disease. N. Engl. J. Med.351, 2069–2079 (2004).
  • Ghosh S, Goldin E, Gordon FH et al. Natalizumab for active Crohn’s disease. N. Engl. J. Med.348, 24–32 (2003).
  • Nakamura K, Honda K, Mizutani T et al. Novel strategies for the treatment of inflammatory bowel disease: selective inhibition of cytokines and adhesion molecules. World J. Gastroenterol.12, 4628–4635 (2006).
  • Vermeire S, Noman M, Van Assche G et al. Effectiveness of concomitant immunosuppressive therapy in suppressing the formation of antibodies to infliximab in Crohn’s disease. Gut56, 1226–1231 (2007).
  • Lemann M, Mary JY, Duclos B et al. Infliximab plus azathioprine for steroid-dependent Crohn’s disease patients: a randomized placebo-controlled trial. Gastroenterology130, 1054–1061 (2006).
  • Hanauer SB. Risks and benefits of combining immunosuppressives and biological agents in inflammatory bowel disease: is the synergy worth the risk? Gut56, 1181–1183 (2007).
  • Mackey AC, Green L, Liang LC et al. Hepatosplenic T cell lymphoma associated with infliximab use in young patients treated for inflammatory bowel disease. J. Pediatr. Gastroenterol. Nutr.44, 265–267 (2007).
  • Poppe D, Tiede I, Fritz G et al. Azathioprine suppresses ezrin-radixin-moesin-dependent T cell–APC conjugation through inhibition of Vav guanosine exchange activity on Rac proteins. J. Immunol.176, 640–651 (2006).
  • 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.111, 1133–1145 (2003).
  • Mudter J, Neurath MF. Apoptosis of T cells and the control of inflammatory bowel disease: therapeutic implications. Gut56, 293–303 (2007).
  • Boirivant M, Marini M, Di Felice G et al. Lamina propria T cells in Crohn’s disease and other gastrointestinal inflammation show defective CD2 pathway-induced apoptosis. Gastroenterology116, 557–565 (1999).
  • Ina K, Itoh J, Fukushima K et al. Resistance of Crohn’s disease T cells to multiple apoptotic signals is associated with a Bcl-2/Bax mucosal imbalance. J. Immunol.163, 1081–1090 (1999).
  • Ito H, Hirotani T, Yamamoto M et al. Anti-IL-6 receptor monoclonal antibody inhibits leukocyte recruitment and promotes T-cell apoptosis in a murine model of Crohn’s disease. J. Gastroenterol.37(Suppl. 14), 56–61 (2002).
  • Fuss IJ, Marth T, Neurath MF et al. Anti-interleukin 12 treatment regulates apoptosis of Th1 T cells in experimental colitis in mice. Gastroenterology117, 1078–1088 (1999).
  • Van den Brande JM, Koehler TC, Zelinkova Z et al. Prediction of antitumour necrosis factor clinical efficacy by real-time visualisation of apoptosis in patients with Crohn’s disease. Gut56, 509–517 (2007).
  • Elion GB. The George Hitchings and Gertrude Elion Lecture.The pharmacology of azathioprine. Ann. NY Acad. Sci.685, 400–407 (1993).
  • Bohon J, de los Santos CR. Structural effect of the anticancer agent 6-thioguanine on duplex DNA. Nucleic Acids Res.31, 1331–1338 (2003).
  • Somerville L, Krynetski EY, Krynetskaia NF et al. Structure and dynamics of thioguanine-modified duplex DNA. J. Biol. Chem.278, 1005–1011 (2003).
  • Karran P. Thiopurines, DNA damage, DNA repair and therapy-related cancer. Br. Med. Bull.79–80, 153–170 (2006).
  • Maltzman JS, Koretzky GA. Azathioprine: old drug, new actions. J. Clin. Invest.111, 1122–1124 (2003).
  • Dayton JS, Turka LA, Thompson CB et al. Comparison of the effects of mizoribine with those of azathioprine, 6-mercaptopurine, and mycophenolic acid on T lymphocyte proliferation and purine ribonucleotide metabolism. Mol. Pharmacol.41, 671–676 (1992).
  • Quemeneur L, Gerland LM, Flacher M et al. Differential control of cell cycle, proliferation, and survival of primary T lymphocytes by purine and pyrimidine nucleotides. J. Immunol.170, 4986–4995 (2003).
  • Thomas CW, Myhre GM, Tschumper R et al. Selective inhibition of inflammatory gene expression in activated T lymphocytes: a mechanism of immune suppression by thiopurines. J. Pharmacol. Exp. Ther.312, 537–545 (2005).
  • Neurath MF, Kiesslich R, Teichgraber U et al. 6-thioguanosine diphosphate and triphosphate levels in red blood cells and response to azathioprine therapy in Crohn’s disease. Clin. Gastroenterol. Hepatol.3, 1007–1014 (2005).
  • Cantrell D. Lymphocyte signalling: a coordinating role for Vav? Curr.Biol.8, R535–R538 (1998).
  • Bustelo XR. Vav proteins, adaptors and cell signaling. Oncogene20, 6372–6381 (2001).
  • Khoshnan A, Tindell C, Laux I et al. The NF-κB cascade is important in Bcl-xL expression and for the anti-apoptotic effects of the CD28 receptor in primary human CD4+ lymphocytes. J. Immunol.165, 1743–1754 (2000).
  • Cantrell DA. GTPases and T cell activation. Immunol. Rev.192, 122–130 (2003).
  • Crespo P, Schuebel KE, Ostrom AA et al. Phosphotyrosine-dependent activation of Rac-1 GDP/GTP exchange by the Vav proto-oncogene product. Nature385, 169–172 (1997).
  • Darnell JE Jr. STATs and gene regulation. Science277, 1630–1635 (1997).
  • Fanger GR, Johnson NL, Johnson GL. MEK kinases are regulated by EGF and selectively interact with Rac/Cdc42. Embo J.16, 4961–4972 (1997).
  • Grad JM, Zeng XR, Boise LH. Regulation of Bcl-xL: a little bit of this and a little bit of STAT. Curr. Opin. Oncol.12, 543–549 (2000).
  • Bretscher A. Regulation of cortical structure by the ezrin–radixin–moesin protein family. Curr. Opin. Cell. Biol.11, 109–116 (1999).
  • Faure S, Salazar-Fontana LI, Semichon M et al. ERM proteins regulate cytoskeleton relaxation promoting T cell-APC conjugation. Nat. Immunol.5, 272–279 (2004).
  • Holtmann MH, Krummenauer F, Claas C et al. Long-term effectiveness of azathioprine in IBD beyond 4 years: a European multicenter study in 1176 patients. Dig. Dis. Sci.51, 1516–1524 (2006).
  • Dubinsky MC, Yang H, Hassard PV et al. 6-MP metabolite profiles provide a biochemical explanation for 6-MP resistance in patients with inflammatory bowel disease. Gastroenterology122, 904–915 (2002).
  • Stocco G, Martelossi S, Decorti G et al. Thiopurine-S-methyltransferase genotype and the response to azathioprine in inflammatory bowel disease. Aliment. Pharmacol. Ther.26, 1083–1084 (2007).
  • Winter JW, Gaffney D, Shapiro D et al. Assessment of thiopurine methyltransferase enzyme activity is superior to genotype in predicting myelosuppression following azathioprine therapy in patients with inflammatory bowel disease. Aliment. Pharmacol. Ther.25, 1069–1077 (2007).
  • Lamers CB, Griffioen G, van Hogezand RA et al. Azathioprine: an update on clinical efficacy and safety in inflammatory bowel disease. Scand. J. Gastroenterol.230(Suppl.), 111–115 (1999).
  • Fraser AG, Orchard TR, Jewell DP. The efficacy of azathioprine for the treatment of inflammatory bowel disease: a 30 year review. Gut50, 485–489 (2002).
  • Present DH, Meltzer SJ, Krumholz MP et al. 6-mercaptopurine in the management of inflammatory bowel disease: short- and long-term toxicity. Ann. Intern. Med.111, 641–649 (1989).
  • Chang FW, Reinhart S, Fraser NM. Effect of 30% sodium sulfacetamide on corneal sensitivity. Am. J. Optom. Physiol. Opt.61, 318–320 (1984).
  • Kinlen LJ. Incidence of cancer in rheumatoid arthritis and other disorders after immunosuppressive treatment. Am. J. Med.78, 44–49 (1985).
  • Opelz G, Henderson R. Incidence of non-Hodgkin lymphoma in kidney and heart transplant recipients. Lancet342, 1514–1516 (1993).
  • Silman AJ, Petrie J, Hazleman B et al. Lymphoproliferative cancer and other malignancy in patients with rheumatoid arthritis treated with azathioprine: a 20 year follow up study. Ann. Rheum. Dis.47, 988–992 (1988).
  • Garnier JL, Lebranchu Y, Dantal J et al. Hodgkin’s disease after transplantation. Transplantation61, 71–76 (1996).
  • Kwon JH, Farrell RJ. The risk of lymphoma in the treatment of inflammatory bowel disease with immunosuppressive agents. Crit. Rev. Oncol. Hematol.56, 169–178 (2005).
  • Lewis JD, Schwartz JS, Lichtenstein GR. Azathioprine for maintenance of remission in Crohn’s disease: benefits outweigh the risk of lymphoma. Gastroenterology118, 1018–1024 (2000).
  • de Jong DJ, Derijks LJ, Naber AH et al. Safety of thiopurines in the treatment of inflammatory bowel disease. Scand. J. Gastroenterol.239(Suppl.) 69–72 (2003).
  • Osterman MT, Kundu R, Lichtenstein GR et al. Association of 6-thioguanine nucleotide levels and inflammatory bowel disease activity: a meta-analysis. Gastroenterology130, 1047–1053 (2006).
  • Teml A, Schaeffeler E, Herrlinger KR et al. Thiopurine treatment in inflammatory bowel disease: clinical pharmacology and implication of pharmacogenetically guided dosing. Clin. Pharmacokinet.46, 187–208 (2007).
  • Dilger K, Schaeffeler E, Lukas M et al. Monitoring of thiopurine methyltransferase activity in postsurgical patients with Crohn’s disease during 1 year of treatment with azathioprine or mesalazine. Ther. Drug Monit.29, 1–5 (2007).
  • Lennard L, Lilleyman JS, Van Loon J et al. Genetic variation in response to 6-mercaptopurine for childhood acute lymphoblastic leukaemia. Lancet336, 225–229 (1990).
  • Schaeffeler E, Eichelbaum M, Reinisch W et al. Three novel thiopurine S-methyltransferase allelic variants (TPMT*20, *21, *22) – association with decreased enzyme function. Hum. Mutat.27, 976– (2006).
  • Lennard L. TPMT in the treatment of Crohn’s disease with azathioprine. Gut51, 143–146 (2002).
  • Szumlanski CL, Weinshilboum RM. Sulphasalazine inhibition of thiopurine methyltransferase: possible mechanism for interaction with 6-mercaptopurine and azathioprine. Br. J. Clin. Pharmacol.39, 456–459 (1995).
  • Xin HW, Fischer C, Schwab M et al. Thiopurine S-methyltransferase as a target for drug interactions. Eur. J. Clin. Pharmacol.61,395–398 (2005).
  • Marinaki AM, Duley JA, Arenas M et al. Mutation in the ITPA gene predicts intolerance to azathioprine. Nucleosides Nucleotides Nucleic Acids23, 1393–1397 (2004).
  • Stocco G, Martelossi S, Barabino A et al. Glutathione-S-transferase genotypes and the adverse effects of azathioprine in young patients with inflammatory bowel disease. Inflamm. Bowel Dis.13, 57–64 (2007).
  • Shipkova M, Lorenz K, Oellerich M et al. Measurement of erythrocyte inosine triphosphate pyrophosphohydrolase (ITPA) activity by HPLC and correlation of ITPA genotype-phenotype in a Caucasian population. Clin. Chem.52, 240–247 (2006).
  • Eklund BI, Moberg M, Bergquist J et al. Divergent activities of human glutathione transferases in the bioactivation of azathioprine. Mol. Pharmacol.70, 747–754 (2006).
  • Hobara N, Watanabe A. Impaired metabolism of azathioprine in carbon tetrachloride-injured rats. Hepatogastroenterology28, 192–194 (1981).
  • Herrlinger KR, Kreisel W, Schwab M et al. 6-thioguanine – efficacy and safety in chronic active Crohn’s disease. Aliment. Pharmacol. Ther.17, 503–508 (2003).
  • Palmieri O, Latiano A, Bossa F et al. Sequential evaluation of thiopurine methyltransferase, inosine triphosphate pyrophosphatase, and HPRT1 genes polymorphisms to explain thiopurines’ toxicity and efficacy. Aliment. Pharmacol. Ther.26, 737–745 (2007).
  • 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. Gastroenterology118, 1025–1030 (2000).
  • Winter J, Walker A, Shapiro D et al. Cost–effectiveness of thiopurine methyltransferase genotype screening in patients about to commence azathioprine therapy for treatment of inflammatory bowel disease. Aliment. Pharmacol. Ther.20, 593–599 (2004).
  • Ansari A, Hassan C, Duley J et al. Thiopurine methyltransferase activity and the use of azathioprine in inflammatory bowel disease. Aliment. Pharmacol. Ther.16, 1743–1750 (2002).
  • Campbell S, Kingstone K, Ghosh S. Relevance of thiopurine methyltransferase activity in inflammatory bowel disease patients maintained on low-dose azathioprine. Aliment. Pharmacol. Ther.16, 389–398 (2002).
  • Cuffari C, Dassopoulos T, Turnbough L et al. Thiopurine methyltransferase activity influences clinical response to azathioprine in inflammatory bowel disease. Clin. Gastroenterol. Hepatol.2, 410–417 (2004).
  • Reuther LO, Sonne J, Larsen NE et al. Pharmacological monitoring of azathioprine therapy. Scand. J. Gastroenterol.38, 972–977 (2003).
  • Cuffari C, Theoret Y, Latour S et al. 6-Mercaptopurine metabolism in Crohn’s disease: correlation with efficacy and toxicity. Gut39, 401–406 (1996).
  • Wusk B, Kullak-Ublick GA, Rammert C et al. Therapeutic drug monitoring of thiopurine drugs in patients with inflammatory bowel disease or autoimmune hepatitis. Eur. J. Gastroenterol. Hepatol.16, 1407–1413 (2004).
  • Herrlinger KR, Fellermann K, Fischer C et al. Thioguanine-nucleotides do not predict efficacy of tioguanine in Crohn’s disease. Aliment. Pharmacol. Ther.19, 1269–1276 (2004).
  • Lowry PW, Franklin CL, Weaver AL et al. Measurement of thiopurine methyltransferase activity and azathioprine metabolites in patients with inflammatory bowel disease. Gut49, 665–670 (2001).
  • Cuffari C, Hunt S, Bayless T. Utilisation of erythrocyte 6-thioguanine metabolite levels to optimise azathioprine therapy in patients with inflammatory bowel disease. Gut48, 642–646 (2001).

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.