Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 1/2011

Open Access 01-01-2011 | Short Communication

Effects of methimazole on the elimination of irinotecan

Authors: Jessica M. van der Bol, Theo J. Visser, Walter J. Loos, Floris A. de Jong, Erik A. C. Wiemer, Maarten O. van Aken, Andre S. Planting, Jan H. Schellens, Jaap Verweij, Ron H. J. Mathijssen

Published in: Cancer Chemotherapy and Pharmacology | Issue 1/2011

Login to get access

Abstract

Purpose

To study the possible pharmacokinetic and pharmacodynamic interactions between irinotecan and methimazole.

Methods

A patient treated for colorectal cancer with single agent irinotecan received methimazole co-medication for Graves’ disease. Irinotecan pharmacokinetics and side effects were followed during a total of four courses (two courses with and two courses without methimazole).

Results

Plasma concentrations of the active irinotecan metabolite SN-38 and its inactive metabolite SN-38-Glucuronide were both higher (a mean increase of 14 and 67%, respectively) with methimazole co-medication, compared to irinotecan monotherapy. As a result, the mean SN-38 glucuronidation rate increased with 47% during concurrent treatment. Other possible confounding factors did not change over time. Specific adverse events due to methimazole co-treatment were not seen.

Conclusions

Additional in vitro experiments suggest that these results can be explained by induction of UGT1A1 by methimazole, leading to higher SN-38G concentrations. The prescribed combination of these drugs may lead to highly toxic intestinal SN-38 levels. We therefore advise physicians to be very careful in combining methimazole with regular irinotecan doses, especially in patients who are prone to irinotecan toxicity.
Appendix
Available only for authorised users
Literature
1.
go back to reference Vanhoefer U, Harstrick A, Achterrath W, Cao S, Seeber S, Rustum YM (2001) Irinotecan in the treatment of colorectal cancer: clinical overview. J Clin Oncol 19:1501–1518PubMed Vanhoefer U, Harstrick A, Achterrath W, Cao S, Seeber S, Rustum YM (2001) Irinotecan in the treatment of colorectal cancer: clinical overview. J Clin Oncol 19:1501–1518PubMed
2.
go back to reference Di Paolo A, Bocci G, Danesi R, Del Tacca M (2006) Clinical pharmacokinetics of irinotecan-based chemotherapy in colorectal cancer patients. Curr Clin Pharmacol 1:311–323CrossRefPubMed Di Paolo A, Bocci G, Danesi R, Del Tacca M (2006) Clinical pharmacokinetics of irinotecan-based chemotherapy in colorectal cancer patients. Curr Clin Pharmacol 1:311–323CrossRefPubMed
3.
go back to reference Innocenti F, Kroetz DL, Schuetz E, Dolan ME, Ramirez J, Relling M, Chen P, Das S, Rosner GL, Ratain MJ (2009) Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J Clin Oncol 27:2604–2614CrossRefPubMed Innocenti F, Kroetz DL, Schuetz E, Dolan ME, Ramirez J, Relling M, Chen P, Das S, Rosner GL, Ratain MJ (2009) Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J Clin Oncol 27:2604–2614CrossRefPubMed
4.
go back to reference de Jong FA, Sparreboom A, Verweij J, Mathijssen RH (2008) Lifestyle habits as a contributor to anti-cancer treatment failure. Eur J Cancer 44:374–382CrossRefPubMed de Jong FA, Sparreboom A, Verweij J, Mathijssen RH (2008) Lifestyle habits as a contributor to anti-cancer treatment failure. Eur J Cancer 44:374–382CrossRefPubMed
5.
go back to reference Mathijssen RH, Verweij J, de Bruijn P, Loos WJ, Sparreboom A (2002) Effects of St. John’s wort on irinotecan metabolism. J Natl Cancer Inst 94:1247–1249PubMed Mathijssen RH, Verweij J, de Bruijn P, Loos WJ, Sparreboom A (2002) Effects of St. John’s wort on irinotecan metabolism. J Natl Cancer Inst 94:1247–1249PubMed
6.
go back to reference van der Bol JM, Mathijssen RH, Loos WJ, Friberg LE, van Schaik RH, de Jonge MJ, Planting AS, Verweij J, Sparreboom A, de Jong FA (2007) Cigarette smoking and irinotecan treatment: pharmacokinetic interaction and effects on neutropenia. J Clin Oncol 25:2719–2726CrossRefPubMed van der Bol JM, Mathijssen RH, Loos WJ, Friberg LE, van Schaik RH, de Jonge MJ, Planting AS, Verweij J, Sparreboom A, de Jong FA (2007) Cigarette smoking and irinotecan treatment: pharmacokinetic interaction and effects on neutropenia. J Clin Oncol 25:2719–2726CrossRefPubMed
7.
go back to reference Guo Z, Raeissi S, White RB, Stevens JC (1997) Orphenadrine and methimazole inhibit multiple cytochrome P450 enzymes in human liver microsomes. Drug Metab Dispos 25:390–393PubMed Guo Z, Raeissi S, White RB, Stevens JC (1997) Orphenadrine and methimazole inhibit multiple cytochrome P450 enzymes in human liver microsomes. Drug Metab Dispos 25:390–393PubMed
8.
go back to reference Visser TJ, Kaptein E, Gijzel A, de Herder WW, Cannon ML, Bonthuis F, de Greef WJ (1996) Effects of thyroid status and thyrostatic drugs on hepatic glucuronidation of lodothyronines and other substrates in rats. Endocrine 4:78–85CrossRef Visser TJ, Kaptein E, Gijzel A, de Herder WW, Cannon ML, Bonthuis F, de Greef WJ (1996) Effects of thyroid status and thyrostatic drugs on hepatic glucuronidation of lodothyronines and other substrates in rats. Endocrine 4:78–85CrossRef
9.
go back to reference de Bruijn P, Willems EW, Loos WJ, Verweij J, Sparreboom A (2004) Indirect determination of the irinotecan metabolite 7-ethyl-10-O-glucuronyl-camptothecin in human samples. Anal Biochem 328:84–86CrossRefPubMed de Bruijn P, Willems EW, Loos WJ, Verweij J, Sparreboom A (2004) Indirect determination of the irinotecan metabolite 7-ethyl-10-O-glucuronyl-camptothecin in human samples. Anal Biochem 328:84–86CrossRefPubMed
10.
go back to reference Sparreboom A, de Bruijn P, de Jonge MJ, Loos WJ, Stoter G, Verweij J, Nooter K (1998) Liquid chromatographic determination of irinotecan and three major metabolites in human plasma, urine and feces. J Chromatogr B Biomed Sci Appl 712:225–235CrossRefPubMed Sparreboom A, de Bruijn P, de Jonge MJ, Loos WJ, Stoter G, Verweij J, Nooter K (1998) Liquid chromatographic determination of irinotecan and three major metabolites in human plasma, urine and feces. J Chromatogr B Biomed Sci Appl 712:225–235CrossRefPubMed
11.
go back to reference Van Steenbergen W, Fevery J, De Vos R, Leyten R, Heirwegh KP, De Groote J (1989) Thyroid hormones and the hepatic handling of bilirubin. I. Effects of hypothyroidism and hyperthyroidism on the hepatic transport of bilirubin mono- and diconjugates in the Wistar rat. Hepatology 9:314–321CrossRefPubMed Van Steenbergen W, Fevery J, De Vos R, Leyten R, Heirwegh KP, De Groote J (1989) Thyroid hormones and the hepatic handling of bilirubin. I. Effects of hypothyroidism and hyperthyroidism on the hepatic transport of bilirubin mono- and diconjugates in the Wistar rat. Hepatology 9:314–321CrossRefPubMed
12.
go back to reference Kehrer DF, Mathijssen RH, Verweij J, de Bruijn P, Sparreboom A (2002) Modulation of irinotecan metabolism by ketoconazole. J Clin Oncol 20:3122–3129CrossRefPubMed Kehrer DF, Mathijssen RH, Verweij J, de Bruijn P, Sparreboom A (2002) Modulation of irinotecan metabolism by ketoconazole. J Clin Oncol 20:3122–3129CrossRefPubMed
13.
go back to reference Tobin PJ, Beale P, Noney L, Liddell S, Rivory LP, Clarke S (2006) A pilot study on the safety of combining chrysin, a non-absorbable inducer of UGT1A1, and irinotecan (CPT-11) to treat metastatic colorectal cancer. Cancer Chemother Pharmacol 57:309–316CrossRefPubMed Tobin PJ, Beale P, Noney L, Liddell S, Rivory LP, Clarke S (2006) A pilot study on the safety of combining chrysin, a non-absorbable inducer of UGT1A1, and irinotecan (CPT-11) to treat metastatic colorectal cancer. Cancer Chemother Pharmacol 57:309–316CrossRefPubMed
14.
go back to reference Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1996) Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res 56:3752–3757PubMed Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1996) Involvement of beta-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res 56:3752–3757PubMed
15.
go back to reference Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1998) Inhibition of intestinal microflora beta-glucuronidase modifies the distribution of the active metabolite of the antitumor agent, irinotecan hydrochloride (CPT-11) in rats. Cancer Chemother Pharmacol 42:280–286CrossRefPubMed Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1998) Inhibition of intestinal microflora beta-glucuronidase modifies the distribution of the active metabolite of the antitumor agent, irinotecan hydrochloride (CPT-11) in rats. Cancer Chemother Pharmacol 42:280–286CrossRefPubMed
16.
go back to reference Xie R, Mathijssen RH, Sparreboom A, Verweij J, Karlsson MO (2002) Clinical pharmacokinetics of irinotecan and its metabolites in relation with diarrhea. Clin Pharmacol Ther 72:265–275CrossRefPubMed Xie R, Mathijssen RH, Sparreboom A, Verweij J, Karlsson MO (2002) Clinical pharmacokinetics of irinotecan and its metabolites in relation with diarrhea. Clin Pharmacol Ther 72:265–275CrossRefPubMed
17.
go back to reference Kehrer DF, Sparreboom A, Verweij J, de Bruijn P, Nierop CA, van de Schraaf J, Ruijgrok EJ, de Jonge MJ (2001) Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res 7:1136–1141PubMed Kehrer DF, Sparreboom A, Verweij J, de Bruijn P, Nierop CA, van de Schraaf J, Ruijgrok EJ, de Jonge MJ (2001) Modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res 7:1136–1141PubMed
18.
go back to reference Slatter JG, Su P, Sams JP, Schaaf LJ, Wienkers LC (1997) Bioactivation of the anticancer agent CPT-11 to SN-38 by human hepatic microsomal carboxylesterases and the in vitro assessment of potential drug interactions. Drug Metab Dispos 25:1157–1164PubMed Slatter JG, Su P, Sams JP, Schaaf LJ, Wienkers LC (1997) Bioactivation of the anticancer agent CPT-11 to SN-38 by human hepatic microsomal carboxylesterases and the in vitro assessment of potential drug interactions. Drug Metab Dispos 25:1157–1164PubMed
19.
go back to reference Yong WP, Ramirez J, Innocenti F, Ratain MJ (2005) Effects of ketoconazole on glucuronidation by UDP-glucuronosyltransferase enzymes. Clin Cancer Res 11:6699–6704CrossRefPubMed Yong WP, Ramirez J, Innocenti F, Ratain MJ (2005) Effects of ketoconazole on glucuronidation by UDP-glucuronosyltransferase enzymes. Clin Cancer Res 11:6699–6704CrossRefPubMed
20.
go back to reference Burger H, van Tol H, Boersma AW, Brok M, Wiemer EA, Stoter G, Nooter K (2004) Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump. Blood 104:2940–2942CrossRefPubMed Burger H, van Tol H, Boersma AW, Brok M, Wiemer EA, Stoter G, Nooter K (2004) Imatinib mesylate (STI571) is a substrate for the breast cancer resistance protein (BCRP)/ABCG2 drug pump. Blood 104:2940–2942CrossRefPubMed
Metadata
Title
Effects of methimazole on the elimination of irinotecan
Authors
Jessica M. van der Bol
Theo J. Visser
Walter J. Loos
Floris A. de Jong
Erik A. C. Wiemer
Maarten O. van Aken
Andre S. Planting
Jan H. Schellens
Jaap Verweij
Ron H. J. Mathijssen
Publication date
01-01-2011
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 1/2011
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-010-1414-x

Other articles of this Issue 1/2011

Cancer Chemotherapy and Pharmacology 1/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine