Aktuelle Rheumatologie 2008; 33(4): 211-216
DOI: 10.1055/s-2008-1027578
Übersichtsarbeit

© Georg Thieme Verlag KG Stuttgart · New York

Toxizität von Azathioprin

Azathioprine – Its Toxicity and Side EffectsG. Riemekasten1 , und die Kommission Pharmakotherapie der Deutschen Gesellschaft für Rheumatologie
  • 1Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité-Universitätsmedizin Berlin
Further Information

Publication History

Publication Date:
21 July 2008 (online)

Zusammenfassung

In dieser Literaturanalyse werden die Arbeiten zur Kurz- und Langzeittoxizität von Azathioprin zusammengefasst, wobei die Häufigkeit und das Vorkommen im zeitlichen Verlauf aufgezeigt werden. 4 – 6 % der Patienten erleiden innerhalb weniger Tage eine starke Übelkeit, die zum Absetzen zwingt. Das Azathioprin-induzierte Fieber kommt bei ca. 1 – 2 % der Patienten vor. Eine Knochenmarksdepression lässt sich in einer Häufigkeit von 2 – 12 % nachweisen. In 3 % der Patienten kommt es zu einer Lebertoxizität, die teilweise dosisabhängig wie auch -unabhängig auftreten kann. Die Nebenwirkungen lassen sich nicht ausreichend durch die Bestimmung des Genotyps der Thiopurin-Methyltransferase (TPMT) vorhersagen. Die Verträglichkeit von Azathioprin ist abhängig von der zugrunde liegenden Erkrankung und ist bei Kollagenosen und Vaskulitiden höher (7 % Abbruchrate wegen Nebenwirkungen) als bei der rheumatoiden Arthritis (25 % Therapieabbruch). Die Erhöhung des Lymphomrisikos durch Azathioprin ist beim Menschen nicht belegt. Ebenso gibt es kein erhöhtes Risiko für Karzinome. Schwangerschaft und Entwicklung des Kindes sind eher von der Grunderkrankung beeinflusst als von der Azathioprin-Therapie, Fehlbildungen kommen etwa wie in der Normalbevölkerung in 3 – 5 % der Schwangerschaften vor. Insgesamt gehört Azathioprin derzeit zu den sichersten Medikamenten.

Abstract

This review summarises literature data about the prevalence of azathioprine-induced short- and long-term toxicity available from different diseases. Within a few days, about 4 – 6 % of the patients suffer from vomiting requiring discontinuation of therapy. Azathioprine-induced fever occurs in about 1 – 2 % of the patients. The prevalence of bone marrow toxicity is between 2 – 12 % and is dose-dependent. Liver toxicity is both dose-dependent and dose-independent and occurs in about 3 % of the patients. Unfortunately, the detection of the thiopurine methyltransferase (TPMT) cannot predict side effects. Tolerance to azathioprine depends on the existing disease and is higher in patients with vasculitis and connective tissue diseases compared to those with rheumatoid arthritis. There are no convincing data showing an increased risk of malignant diseases induced by azathioprine. The outcome of pregnancies is more dependent on the underlying disease of the patients, malformations of the infants are not significantly increased compared to the normal population and occur in about 3 – 5 % of the pregnancies. In conclusion, at present, azathioprine belongs to the safest drugs in rheumatology.

Literatur

  • 1 Agarwal S K, Kalra V, Dinda A. et al . Fibrosing cholestatic hepatitis in renal transplant recipient with CMV infection: a case report.  Int Urol Nephrol. 2004;  36 433-435
  • 2 Alstead E M, Ritchie J K, Lennard-Jones J E. et al . Safety of azathioprine in pregnancy in inflammatory bowel disease.  Gastroenterology. 1990;  99 443-446
  • 3 Barabino A, Torrente F, Ventura A. et al . Azathioprine in paediatric inflammatory bowel disease: an Italian multicentre survey.  Aliment Pharmacol Ther. 2002;  16 1125-1130
  • 4 Bergan S, Rugstad H E, Klemetsdal B. et al . Possibilities for therapeutic drug monitoring of azathioprine: 6-thioguanine nucleotide concentrations and thiopurine methyltransferase activity in red blood cells.  Ther Drug Monit. 1997;  19 318-326
  • 5 Caillard S, Dharnidharka V, Agodoa L. et al . Posttransplant lymphoproliferative disorders after renal transplantation in the United States in era of modern immunosuppression.  Transplantation. 2005;  80 1233-1243
  • 6 Cohen S M, Erturk E, Skibba J L. et al . Azathioprine induction of lymphomas and squamous cell carcinomas in rats.  Cancer Res. 1983;  43 2768-2772
  • 7 Connell W, Miller A. Treating inflammatory bowel disease during pregnancy: risks and safety of drug therapy.  Drug Saf. 1999;  21 311-323. Erratum in: Drug Saf 1999; 21: 456
  • 8 Connell W R, Kamm M A, Dickson M. et al . Long-term neoplasia risk after azathioprine treatment in inflammatory bowel disease.  Lancet. 1994;  343 1249-1252
  • 9 Connell W R, Kamm M A, Ritchie J K. et al . Bone marrow toxicity caused by azathioprine in inflammatory bowel disease: 27 years of experience.  Gut. 1993;  34 1081-1085
  • 10 Danesi R, Del Tacca M. Hematologic toxicity of immunosuppressive treatment.  Transplant Proc. 2004;  36 703-704
  • 11 Danesi R, Del Tacca M. Teratogenesis and immunosuppressive treatment.  Transplant Proc. 2004;  36 705-707
  • 12 De Boer N K, Derijks L J, Gilissen L P. 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
  • 13 DeLeve L D, Wang X, Kuhlenkamp J F. et al . Toxicity of azathioprine and monocrotaline in murine sinusoidal endothelial cells and hepatocytes: the role of glutathione and relevance to hepatic venoocclusive disease.  Hepatology. 1996;  23 589-599
  • 14 el-Gamel A, Evans C, Keevil B. et al . Effect of allopurinol on the metabolism of azathioprine in heart transplant patients.  Transplant Proc. 1998;  30 1127-1129
  • 15 Fraser G M, Grammoustianos K, Reddy J. et al . Long-term immunosuppression without corticosteroids after orthotopic liver transplantation: a positive therapeutic aim.  Liver Transpl Surg. 1996;  2 411-417
  • 16 Garrido Serrano A, Perez Martin F, Guerrero Igea F J. et al . Fatal infectious mononucleosis during azathioprine treatment in Crohn’s disease.  Gastroenterol Hepatol. 2000;  23 7-8
  • 17 Golby M. Fertility after renal transplantation.  Transplantation. 1970;  10 201-207
  • 18 Gombar V K, Enslein K, Blake B W. Carcinogenicity of azathioprine: an S-AR investigation.  Mutat Res. 1993;  302 7-12. Erratum in: Mutat Res 1993; 302: 221
  • 19 Heckmann J M, Lambson E M, Little F. et al . Thiopurine methyltransferase (TPMT) heterozygosity and enzyme activity as predictive tests for the development of azathioprine-related adverse events.  J Neurol Sci. 2005;  231 71-80
  • 20 Holme S A, Duley J A, Sanderson J. et al . Erythrocyte thiopurine methyl transferase assessment prior to azathioprine use in the UK.  QJM. 2002;  95 439-44
  • 21 Jun J B, Cho D Y, Kang C. et al . Thiopurine S-methyltransferase polymorphisms and the relationship between the mutant alleles and the adverse effects in systemic lupus erythematosus patients taking azathioprine.  Clin Exp Rheumatol. 2005;  23 873-876
  • 22 Kader H A, Wenner W J, Telega G W. et al . Normal thiopurine methyltransferase levels do not eliminate 6-mercaptopurine or azathioprine toxicity in children with inflammatory bowel disease.  J Clin Gastroenterol. 2000;  30 409-413
  • 23 Kelly G E, Meikle W D, Moore D E. Enhancement of UV-induced skin carcinogenesis by azathioprine: role of photochemical sensitisation.  Photochem Photobiol. 1989;  49 59-65
  • 24 Kennedy D T, Hayney M S, Lake K D. Azathioprine and allopurinol: the price of an avoidable drug interaction.  Ann Pharmacother. 1996;  30 951-954
  • 25 Lacha Jr J, Jirka J, Nouza M. et al . Kidney transplantation and tumors.  Cas Lek Cesk. 1994;  133 562-565
  • 26 Lewis J D, Schwartz J S, Lichtenstein G R. Azathioprine for maintenance of remission in Crohn’s disease: benefits outweigh the risk of lymphoma.  Gastroenterology. 2000;  118 1018-1024
  • 27 Lopez-Sanroman A, Bermejo F, Carrera E. et al . Efficacy and safety of thiopurinic immunomodulators (azathioprine and mercaptopurine) in steroid-dependent ulcerative colitis.  Aliment Pharmacol Ther. 2004;  20 161-166
  • 28 Markowitz J, Grancher K, Mandel F. et al . Immunosuppressive therapy in pediatric inflammatory bowel disease: results of a survey of the North American Society for Pediatric Gastroenterology and Nutrition. Subcommittee on Immunosuppressive Use of the Pediatric IBD Collaborative Research Forum.  Am J Gastroenterol. 1993;  88 44-48
  • 29 Markowitz J F. Therapeutic efficacy and safety of 6-mercaptopurine and azathioprine in patients with Crohn’s disease.  Rev Gastroenterol Disord. 2003;  3 (Suppl 1) 23-29
  • 30 Marra C A, Esdaile J M, Anis A H. Practical pharmacogenetics: the cost effectiveness of screening for thiopurine s-methyltransferase polymorphisms in patients with rheumatological conditions treated with azathioprine.  J Rheumatol. 2002;  29 2507-2512
  • 31 Marshall E. Clinical trials. In critical condition.  Science. 2003;  300 1225-1226
  • 32 Martin S R, Russo P, Dubois J. et al . Centrilobular fibrosis in long-term follow-up of pediatric liver transplant recipients.  Transplantation. 2002;  74 828-836
  • 33 Martinez F, Nos P, Pastor M. et al . Adverse effects of azathioprine in the treatment of inflammatory bowel disease.  Rev Esp Enferm Dig. 2001;  93 769-778
  • 34 McGovern D P, Jewell D P. Risks and benefits of azathioprine therapy.  Gut. 2005;  54 1055-1059
  • 35 Miller L W. Cardiovascular toxicities of immunosuppressive agents.  Am J Transplant. 2002;  2 807-818
  • 36 Mitrou P S, Fischer M, Mitrou G. et al . The oncogenic effect of immunosuppressive (cytotoxic) agents in (NZB × NZW) mice. II. Emergence of tumors in young animals treated with azathioprine and ifosfamide, including a histologic assessment of the neoplasms.  Arzneimittelforschung. 1979;  29 662-667
  • 37 Naughton M A, Battaglia E, O’Brien S. et al . Identification of thiopurine methyltransferase (TPMT) polymorphisms cannot predict myelosuppression in systemic lupus erythematosus patients taking azathioprine.  Rheumatology. 1999;  38 640-644
  • 38 Oka K, Shimodaira H, Hirano T. et al . Comparison of adrenal functions in kidney transplant recipients with different long-term immunosuppressive treatments – prednisolone and azathioprine versus prednisolone and cyclosporine.  Transplantation. 1993;  56 603-609
  • 39 Polifka J E, Friedman J M. Teratogen update: azathioprine and 6-mercaptopurine.  Teratology. 2002;  65 240-261
  • 40 Present D H, Meltzer S J, Krumholz M P. et al . 6-Mercaptopurine in the management of inflammatory bowel disease: short- and long-term toxicity.  Ann Intern Med. 1989;  111 641-649
  • 41 Reinhold-Keller E, Schmitt W H, Gross W L. Azathioprine toxicity mimicking a relapse of Wegener’s granulomatosis.  Rheumatology. 2001;  40 831-832
  • 42 Silman A J, 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. 1988;  47 988-992
  • 43 Tavadia S M, Mydlarski P R, Reis M D. et al . Screening for azathioprine toxicity: a pharmacoeconomic analysis based on a target case.  J Am Acad Dermatol. 2000;  42 628-632
  • 44 Voogd C E. Azathioprine, a genotoxic agent to be considered non-genotoxic in man.  Mutat Res. 1989;  221 133-152
  • 45 Walker R W, Brochstein J A. Neurologic complications of immunosuppressive agents.  Neurol Clin. 1988;  6 261-278
  • 46 Weersma R K, Peters F T, Oostenbrug L E. et al . Increased incidence of azathioprine-induced pancreatitis in Crohn’s disease compared with other diseases.  Aliment Pharmacol Ther. 2004;  20 843-850
  • 47 Wilkinson A H, Smith J L, Hunsicker L G. et al . Increased frequency of posttransplant lymphomas in patients treated with cyclosporine, azathioprine, and prednisone.  Transplantation. 1989;  47 293-296
  • 48 Williamson R A, Karp L E. Azathioprine teratogenicity: review of the literature and case report.  Obstet Gynecol. 1981;  58 247-250
  • 49 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. 2004;  20 593-599
  • 50 Zukiene J, Zalgeviciene V, Rizgeliene R. The influence of azathioprine on the osteogenesis of the limbs.  Medicina. 2003;  39 584-588

PD Dr. Gabriela Riemekasten

Medizinische Klinik mit Schwerpunkt Rheumatologie und Klinische Immunologie, Charité Universitätsmedizin Berlin

Charitéplatz 1

10117 Berlin

Email: gabriela.riemekasten@charite.de

    >