Skip to main content
Top
Published in: Cancer Chemotherapy and Pharmacology 4/2008

01-04-2008 | Original Article

Involvement of UDP-glucuronosyltransferase activity in irinotecan-induced delayed-onset diarrhea in rats

Authors: Masaharu Onoue, Akinobu Kurita, Shoichi Kado, Tsuneo Matsumoto, Norimasa Kaneda, Kazumi Uchida, Ikuo Kato, Teruo Yokokura

Published in: Cancer Chemotherapy and Pharmacology | Issue 4/2008

Login to get access

Abstract

We assessed the involvement of UDP-glucuronosyltransferase (UGT) activity in episodes of irinotecan hydrochloride (CPT-11)-induced delayed-onset diarrhea using a mutant rat strain with an inherited deficiency of UGT1A (Gunn rats). Gunn rats exhibited severe diarrhea after the intravenous administration of CPT-11 at a dose of 20 mg/kg, whereas Wistar rats did not. In the epithelium of the small intestine and cecum in Gunn rats, the shortening of villi, degeneration of crypts, and destruction of the nucleus were observed. The AUC, MRT, and t 1/2 of CPT-11, and the AUC of 7-ethyl-10-hydroxycamptothecin (SN-38) in plasma were, respectively, 1.6-fold, 1.5-fold, 1.7-fold, and 6.5-fold higher, and the cumulative biliary excretion rate of SN-38 was 2.3-fold higher, in Gunn rats than Wistar rats. SN-38 glucuronide excreted via bile in Wistar rats was not de-conjugated in the small intestinal lumen. The SN-38 AUC values in small intestinal tissues were also 5.0 to 5.8-fold higher in Gunn rats than Wistar rats. In conclusion, Gunn rats developed severe delayed-onset diarrhea after i.v. administration of CPT-11 at a much lower dose. Severe intestinal impairments would be induced in Gunn rats through exposure to SN-38 at high levels for a long period mainly via the intestinal lumen and partially via the bloodstream. These results clarified that the deficiency of UGT activity contributed greatly to the induction of the CPT-11-induced delayed-onset diarrhea and epithelial impairment in the intestine. In the clinic, great care is needed when using chemotherapy with CPT-11 in patients with poor UGT activity.
Literature
1.
go back to reference Abigerges D, Armand J-P, Chabot GG, da Costa L, Fadel E, Cote C, Hérait P, Gandia D (1994) Irinotecan (CPT-11) high-dose escalation using intensive high-dose loperamide to control diarrhea. J Natl Cancer Inst 86:446–449PubMedCrossRef Abigerges D, Armand J-P, Chabot GG, da Costa L, Fadel E, Cote C, Hérait P, Gandia D (1994) Irinotecan (CPT-11) high-dose escalation using intensive high-dose loperamide to control diarrhea. J Natl Cancer Inst 86:446–449PubMedCrossRef
2.
go back to reference Ando Y, Saka H, Ando M, Sawa T, Muro K, Ueoka H, Yokoyama A, Saitoh S, Shimokata K, Hasegawa Y (2000) Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res 60:6921–6926PubMed Ando Y, Saka H, Ando M, Sawa T, Muro K, Ueoka H, Yokoyama A, Saitoh S, Shimokata K, Hasegawa Y (2000) Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. Cancer Res 60:6921–6926PubMed
3.
go back to reference Atsumi R, Suzuki W, Hakusui H (1991) Identification of metabolites of irinotecan, a new derivative of camptothecin, in the rat bile and its biliary excretion. Xenobiotica 21:1159–1169PubMed Atsumi R, Suzuki W, Hakusui H (1991) Identification of metabolites of irinotecan, a new derivative of camptothecin, in the rat bile and its biliary excretion. Xenobiotica 21:1159–1169PubMed
4.
go back to reference Chowdhury JR, Kondapalli R, Chowdhury NR (1993) Gunn rat: a model for inherited deficiency of bilirubin glucuronidation. Adv Vet Sci Comp Med 37:149–173PubMed Chowdhury JR, Kondapalli R, Chowdhury NR (1993) Gunn rat: a model for inherited deficiency of bilirubin glucuronidation. Adv Vet Sci Comp Med 37:149–173PubMed
5.
go back to reference Fukuoka M, Niitani H, Suzuki A, Motomiya M, Hasegawa K, Nishiwaki Y, Kuriyama T, Ariyoshi Y, Negoro S, Masuda N, Nakajima S, Taguchi T (1992) A phase II study of CPT-11, a new derivative of camptothecin, for previously untreated non-small-cell lung cancer (see comment citation in Medline). J Clin Oncol 10:16–20PubMed Fukuoka M, Niitani H, Suzuki A, Motomiya M, Hasegawa K, Nishiwaki Y, Kuriyama T, Ariyoshi Y, Negoro S, Masuda N, Nakajima S, Taguchi T (1992) A phase II study of CPT-11, a new derivative of camptothecin, for previously untreated non-small-cell lung cancer (see comment citation in Medline). J Clin Oncol 10:16–20PubMed
6.
go back to reference Furuta T, Yokokura T, Mutai M (1998) Antitumor activity of CPT-11 against rat Walker 256 carcinoma (in Japanese). Jpn J Cancer Chemother 15:2757–2760 Furuta T, Yokokura T, Mutai M (1998) Antitumor activity of CPT-11 against rat Walker 256 carcinoma (in Japanese). Jpn J Cancer Chemother 15:2757–2760
7.
go back to reference Gandia D, Abigerges D, Armand J-P, Chabot GG, da Costa L, de Forni M (1993) CPT-11-induced cholinergic effects in cancer patients. J Clin Oncol 11:196–197PubMed Gandia D, Abigerges D, Armand J-P, Chabot GG, da Costa L, de Forni M (1993) CPT-11-induced cholinergic effects in cancer patients. J Clin Oncol 11:196–197PubMed
8.
go back to reference Gange JF, Montminy V, Belanger P, Journault K, Guillemette C (2002) Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroycamptothecin (SN-38). Mol Pharmacol 62:608–617CrossRef Gange JF, Montminy V, Belanger P, Journault K, Guillemette C (2002) Common human UGT1A polymorphisms and the altered metabolism of irinotecan active metabolite 7-ethyl-10-hydroycamptothecin (SN-38). Mol Pharmacol 62:608–617CrossRef
9.
go back to reference Gibson RJ, Bowen JM, Inglis MRB, Cummins AG, Keefe DMK. (2003) Irinotecan causes severe small intestinal damage, as well as colonic damage, in the rat with implanted breast cancer. J Gastroenterol Hepatol 18:1095–1100PubMedCrossRef Gibson RJ, Bowen JM, Inglis MRB, Cummins AG, Keefe DMK. (2003) Irinotecan causes severe small intestinal damage, as well as colonic damage, in the rat with implanted breast cancer. J Gastroenterol Hepatol 18:1095–1100PubMedCrossRef
10.
go back to reference Goncalves E, da Costa L, Abigerges D, Armand J-P (1995) A new enkephalinase inhibitor as an alternative to loperamide in the prevention of diarrhea induced by CPT-11. J Clin Oncol 13:2144–2146PubMed Goncalves E, da Costa L, Abigerges D, Armand J-P (1995) A new enkephalinase inhibitor as an alternative to loperamide in the prevention of diarrhea induced by CPT-11. J Clin Oncol 13:2144–2146PubMed
11.
go back to reference Gupta E, Lestingi TM, Mick R, Ramirez J, Vokes EE, Ratain MJ (1994) Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res 54:3723–3725PubMed Gupta E, Lestingi TM, Mick R, Ramirez J, Vokes EE, Ratain MJ (1994) Metabolic fate of irinotecan in humans: correlation of glucuronidation with diarrhea. Cancer Res 54:3723–3725PubMed
12.
go back to reference Gupta E, Wang X, Ramirez J, Ratain MJ (1997) Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother Pharmacol 39:40–444CrossRef Gupta E, Wang X, Ramirez J, Ratain MJ (1997) Modulation of glucuronidation of SN-38, the active metabolite of irinotecan, by valproic acid and phenobarbital. Cancer Chemother Pharmacol 39:40–444CrossRef
13.
go back to reference Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M, Karrison T, Janisch L, Ramírez J, Rudin CM, Vokes EE, Ratain MJ (2004) Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 22:1382–1388PubMedCrossRef Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M, Karrison T, Janisch L, Ramírez J, Rudin CM, Vokes EE, Ratain MJ (2004) Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol 22:1382–1388PubMedCrossRef
14.
go back to reference Kaneda N, Yokokura T (1990) Nonlinear pharmacokinetics of CPT-11 in rats. Cancer Res 50:1721–1725PubMed Kaneda N, Yokokura T (1990) Nonlinear pharmacokinetics of CPT-11 in rats. Cancer Res 50:1721–1725PubMed
15.
go back to reference Kawato Y, Tsumori T, Akahane K, Sekiguchi M, Sato K (1990) Inhibitory effect of CPT-11, a derivative of camptothecin, on acetylcholinesterase, and its binding ability to acetylcholine receptors (in Japanese). Clin Rep 24:7407–7412 Kawato Y, Tsumori T, Akahane K, Sekiguchi M, Sato K (1990) Inhibitory effect of CPT-11, a derivative of camptothecin, on acetylcholinesterase, and its binding ability to acetylcholine receptors (in Japanese). Clin Rep 24:7407–7412
16.
go back to reference Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K (1991) Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res 51:4187–4191PubMed Kawato Y, Aonuma M, Hirota Y, Kuga H, Sato K (1991) Intracellular roles of SN-38, a metabolite of the camptothecin derivative CPT-11, in the antitumor effect of CPT-11. Cancer Res 51:4187–4191PubMed
17.
go back to reference Kitagawa C, Ando M, Ando Y, Sekido Y, Wakai K, Imaizumi K, Shimokata K, Hasegawa Y (2005) Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Pharmacogenet Genomics 15:35–41PubMedCrossRef Kitagawa C, Ando M, Ando Y, Sekido Y, Wakai K, Imaizumi K, Shimokata K, Hasegawa Y (2005) Genetic polymorphism in the phenobarbital-responsive enhancer module of the UDP-glucuronosyltransferase 1A1 gene and irinotecan toxicity. Pharmacogenet Genomics 15:35–41PubMedCrossRef
18.
go back to reference Kojima A, Shinkai T, Saijo N (1993) Cytogenetic effects of CPT-11 and its active metabolite, SN-38 on human lymphocytes. Jpn J Clin Oncol 23:116–122PubMed Kojima A, Shinkai T, Saijo N (1993) Cytogenetic effects of CPT-11 and its active metabolite, SN-38 on human lymphocytes. Jpn J Clin Oncol 23:116–122PubMed
19.
go back to reference Komatsu Y, Takei M, Yuki S, Fuse N, Furukawa S, Kato T, Takeda H, Kato M, Asaka M (2005) Treatment of a Gilbert’s syndrome patient with irinotecan, leucovorin and 5-fluorouracil. J Chemother 17:111–114PubMed Komatsu Y, Takei M, Yuki S, Fuse N, Furukawa S, Kato T, Takeda H, Kato M, Asaka M (2005) Treatment of a Gilbert’s syndrome patient with irinotecan, leucovorin and 5-fluorouracil. J Chemother 17:111–114PubMed
20.
go back to reference Kudoh S, Fukuoka M, Masuda N, Yoshikawa A, Kusunoki Y, Matsui K, Negoro S, Takifuji N, Nakagawa K, Hirashima T (1995) Relationship between the pharmacokinetics of irinotecan and diarrhea during combination chemotherapy with cisplatin. Jpn J Cancer Res 86:406–413PubMed Kudoh S, Fukuoka M, Masuda N, Yoshikawa A, Kusunoki Y, Matsui K, Negoro S, Takifuji N, Nakagawa K, Hirashima T (1995) Relationship between the pharmacokinetics of irinotecan and diarrhea during combination chemotherapy with cisplatin. Jpn J Cancer Res 86:406–413PubMed
21.
go back to reference Kunimoto T, Nitta K, Tanaka T, Uehara M, Baba H, Takeuchi M, Yokokura T, Sawada S, Miyasaka T, Mutai M (1987) Antitumor activity of 7-ethyl-10-(4-(1-piperidino)-1-piperidino) carbonyloxycamptothecin, a novel water-soluble derivative of camptothecin, against murine tumors. Cancer Res 47:5944–5947PubMed Kunimoto T, Nitta K, Tanaka T, Uehara M, Baba H, Takeuchi M, Yokokura T, Sawada S, Miyasaka T, Mutai M (1987) Antitumor activity of 7-ethyl-10-(4-(1-piperidino)-1-piperidino) carbonyloxycamptothecin, a novel water-soluble derivative of camptothecin, against murine tumors. Cancer Res 47:5944–5947PubMed
22.
go back to reference Kurita A, Kaneda N (1999) High-performance liquid chromatographic method for the simultaneous determination of the camptothecin derivative irinotecan hydrochloride, CPT-11, and its metabolites SN-38 and SN-38 glucuronide in rat plasma with a fully automated on-line solid phase extraction system, PROSPEKT. J Chromatogr B 724:335–344CrossRef Kurita A, Kaneda N (1999) High-performance liquid chromatographic method for the simultaneous determination of the camptothecin derivative irinotecan hydrochloride, CPT-11, and its metabolites SN-38 and SN-38 glucuronide in rat plasma with a fully automated on-line solid phase extraction system, PROSPEKT. J Chromatogr B 724:335–344CrossRef
23.
go back to reference Kurita K, Kado S, Kaneda N, Onoue M, Hashimoto S, Yokokura T (2000) Modified irinotecan hydrochloride (CPT-11) administration schedule improves induction of delayed-onset diarrhea in rats. Cancer Chemother Pharmacol 46:211–220PubMedCrossRef Kurita K, Kado S, Kaneda N, Onoue M, Hashimoto S, Yokokura T (2000) Modified irinotecan hydrochloride (CPT-11) administration schedule improves induction of delayed-onset diarrhea in rats. Cancer Chemother Pharmacol 46:211–220PubMedCrossRef
24.
go back to reference Mehra R, Murren J, Chung G, Smith B, Psyrri A (2005) Severe irinotacan-induced toxicies in a patient with uridine diphosphate glucuronosyltransferase 1A1 polymorphism. Clin Colorectal Cancer 5:61–64PubMedCrossRef Mehra R, Murren J, Chung G, Smith B, Psyrri A (2005) Severe irinotacan-induced toxicies in a patient with uridine diphosphate glucuronosyltransferase 1A1 polymorphism. Clin Colorectal Cancer 5:61–64PubMedCrossRef
25.
go back to reference Mehta VK, Cho C, Ford JM, Jambalos C, Poen J, Koong A, Lin A, Bastidas JA, Young H, Dunphy EP, Fisher G (2003) Phase II trial of preoperative 3D conformal radiotherapy, protracted venous infusion 5-fluorouracil, and weekly CPT-11, followed by surgery for ultrasound-staged T3 rectal cancer. Int J Radiat Oncol Biol Phys 55:132–137PubMedCrossRef Mehta VK, Cho C, Ford JM, Jambalos C, Poen J, Koong A, Lin A, Bastidas JA, Young H, Dunphy EP, Fisher G (2003) Phase II trial of preoperative 3D conformal radiotherapy, protracted venous infusion 5-fluorouracil, and weekly CPT-11, followed by surgery for ultrasound-staged T3 rectal cancer. Int J Radiat Oncol Biol Phys 55:132–137PubMedCrossRef
26.
go back to reference Ohe Y, Sakai Y, Shinkai T, Eguchi K, Tamura T, Kojima A, Kunikane H, Okamoto H, Karato A, Ohmatsu H, Kanzawa F, Saijo N (1993) Phase I study and pharmacokinetics of CPT-11 with 5-day continuous infusion. J Natl Cancer Inst 84:972–974CrossRef Ohe Y, Sakai Y, Shinkai T, Eguchi K, Tamura T, Kojima A, Kunikane H, Okamoto H, Karato A, Ohmatsu H, Kanzawa F, Saijo N (1993) Phase I study and pharmacokinetics of CPT-11 with 5-day continuous infusion. J Natl Cancer Inst 84:972–974CrossRef
27.
go back to reference Ohno R, Okada K, Masaoka T, Kuramoto A, Arima T, Yoshida Y, Ariyoshi H, Ichimaru M, Sakai Y, Oguro M, Ito Y, Morishima Y, Yokomaru S, Ohta K (1990) An early II phase study of CPT-11: a new derivative of camptothecin, for the treatment of leukemia and lymphoma. J Clin Oncol 8:1907–1912PubMed Ohno R, Okada K, Masaoka T, Kuramoto A, Arima T, Yoshida Y, Ariyoshi H, Ichimaru M, Sakai Y, Oguro M, Ito Y, Morishima Y, Yokomaru S, Ohta K (1990) An early II phase study of CPT-11: a new derivative of camptothecin, for the treatment of leukemia and lymphoma. J Clin Oncol 8:1907–1912PubMed
28.
go back to reference Paoluzzi L, Singh AS, Price DK, Danesi R, Mathijssen RH, Verweij J, Figg WD, Sparreboom A (2004) Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. J Clin Pharmacol 44:854–860PubMedCrossRef Paoluzzi L, Singh AS, Price DK, Danesi R, Mathijssen RH, Verweij J, Figg WD, Sparreboom A (2004) Influence of genetic variants in UGT1A1 and UGT1A9 on the in vivo glucuronidation of SN-38. J Clin Pharmacol 44:854–860PubMedCrossRef
29.
go back to reference Rothenberg ML, Kuhn JG, Burris HA, Morales MT, Nelson J, Eckardt JR, Rock MK, Terada K, von Hoff DD (1992) Phase I and pharmacokinetic trial of CPT-11 in patients with refractory solid tumors. Proc Am Soc Clin Oncol 11:2194–2204 Rothenberg ML, Kuhn JG, Burris HA, Morales MT, Nelson J, Eckardt JR, Rock MK, Terada K, von Hoff DD (1992) Phase I and pharmacokinetic trial of CPT-11 in patients with refractory solid tumors. Proc Am Soc Clin Oncol 11:2194–2204
30.
go back to reference Rouits E, Boisdron-Celle M, Dumont A, Guerin O, Morel A, Gamelin E (2004) Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res 10:5151–5159PubMedCrossRef Rouits E, Boisdron-Celle M, Dumont A, Guerin O, Morel A, Gamelin E (2004) Relevance of different UGT1A1 polymorphisms in irinotecan-induced toxicity: a molecular and clinical study of 75 patients. Clin Cancer Res 10:5151–5159PubMedCrossRef
31.
go back to reference Saliba F, Hagipantelli R, Misset J-L, Bastian G, Vassal G, Bonnay M, Herait P, Cote C, Mahjoubi M, Mignard D, Cvitkovic E (1998) Pathophysiology and therapy of irinotecan-induced delayed-onset diarrhea in patients with advanced colorectal cancer: a prospective assessment. J Clin Oncol 16:2745–2751PubMed Saliba F, Hagipantelli R, Misset J-L, Bastian G, Vassal G, Bonnay M, Herait P, Cote C, Mahjoubi M, Mignard D, Cvitkovic E (1998) Pathophysiology and therapy of irinotecan-induced delayed-onset diarrhea in patients with advanced colorectal cancer: a prospective assessment. J Clin Oncol 16:2745–2751PubMed
32.
go back to reference Sasaki Y, Hakusui H, Mizuno S, Morita M, Miya T, Eguchi K, Shinkai T, Tamura T, Ohe Y, Saijo N (1995) A pharmacokinetic and pharmacodynamic analysis of CPT-11 and its metabolite SN-38. Jpn J Cancer Res 86:101–110PubMed Sasaki Y, Hakusui H, Mizuno S, Morita M, Miya T, Eguchi K, Shinkai T, Tamura T, Ohe Y, Saijo N (1995) A pharmacokinetic and pharmacodynamic analysis of CPT-11 and its metabolite SN-38. Jpn J Cancer Res 86:101–110PubMed
33.
go back to reference Shimada Y, Yoshino M, Wakui A, Nakao I, Futatsuki K, Sakata Y, Kambe M, Taguchi T, Ogawa N (1993) Phase II study of CPT-11, a new camptothecin derivative, in metastatic colorectal cancer. CPT-11 Gastrointestinal Cancer Study Group. J Clin Oncol 11:909–913PubMed Shimada Y, Yoshino M, Wakui A, Nakao I, Futatsuki K, Sakata Y, Kambe M, Taguchi T, Ogawa N (1993) Phase II study of CPT-11, a new camptothecin derivative, in metastatic colorectal cancer. CPT-11 Gastrointestinal Cancer Study Group. J Clin Oncol 11:909–913PubMed
34.
go back to reference Shirao K, Shimada Y, Kondo H, Saito D, Yamao T, Ono H, Yokoyama T, Fukuda H, Oka M, Watanabe Y, Ohtsu A, Boku N, Fujii T, Oda Y, Muro K, Yoshida S (1997) Phase I–II study of irinotecan hydrochloride combined with cisplatin in patients with advanced gastric cancer. J Clin Oncol 15:921–927PubMed Shirao K, Shimada Y, Kondo H, Saito D, Yamao T, Ono H, Yokoyama T, Fukuda H, Oka M, Watanabe Y, Ohtsu A, Boku N, Fujii T, Oda Y, Muro K, Yoshida S (1997) Phase I–II study of irinotecan hydrochloride combined with cisplatin in patients with advanced gastric cancer. J Clin Oncol 15:921–927PubMed
35.
go back to reference Takasuna K, Kasai Y, Kitano Y, Mori K, Kobayashi R, Hagiwara T, Kakihata K, Hirohashi M, Nomura M, Nagai E, Kamataki T (1995) Protective effects of Kampo medicine and baicalin against intestinal toxicity of a new anticancer camptothecin derivative, irinotecan hydrochloride (CPT-11), in rats. Jpn J Cancer Res 86:978–984PubMed Takasuna K, Kasai Y, Kitano Y, Mori K, Kobayashi R, Hagiwara T, Kakihata K, Hirohashi M, Nomura M, Nagai E, Kamataki T (1995) Protective effects of Kampo medicine and baicalin against intestinal toxicity of a new anticancer camptothecin derivative, irinotecan hydrochloride (CPT-11), in rats. Jpn J Cancer Res 86:978–984PubMed
36.
go back to reference Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1996) Involvement of β-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride. Cancer Res 56:3752–3757PubMed Takasuna K, Hagiwara T, Hirohashi M, Kato M, Nomura M, Nagai E, Yokoi T, Kamataki T (1996) Involvement of β-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride. Cancer Res 56:3752–3757PubMed
37.
go back to reference Takeuchi S, Dobashi K, Fujimoto S, Tanaka K, Suzuki M, Terashima Y, Hasumi K, Akiya K, Negishi Y, Tamiya T, Tanizawa O, Sugawa T, Umesaki N, Sekiba K, Aono T, Nakano H, Noda K, Shirota M, Yakushiji M, Sugiyama T, Hashimoto M, Yajima A, Takamizawa H, Sonoda T, Takeda Y, Tomoda Y, Ohta M, Ozaki M, Hirabayashi K, Hiura M, Hatae M, Nishigaki K, Taguchi T (1991) A late phase II study of CPT-11 on uterine cervical cancer and ovarian cancer. Research groups of CPT-11 in Gynecologic Cancers (in Japanese). Jpn J Cancer Chemother 18:1681–1689 Takeuchi S, Dobashi K, Fujimoto S, Tanaka K, Suzuki M, Terashima Y, Hasumi K, Akiya K, Negishi Y, Tamiya T, Tanizawa O, Sugawa T, Umesaki N, Sekiba K, Aono T, Nakano H, Noda K, Shirota M, Yakushiji M, Sugiyama T, Hashimoto M, Yajima A, Takamizawa H, Sonoda T, Takeda Y, Tomoda Y, Ohta M, Ozaki M, Hirabayashi K, Hiura M, Hatae M, Nishigaki K, Taguchi T (1991) A late phase II study of CPT-11 on uterine cervical cancer and ovarian cancer. Research groups of CPT-11 in Gynecologic Cancers (in Japanese). Jpn J Cancer Chemother 18:1681–1689
38.
go back to reference Tallman MN, Ritter JK, Smith PC (2005) Differential rates of glucuronidation for 7-ethyl-10-hydroxycamptothecin (SN-38) lactone and carboxylate in human and rat microsomes and recombinant UDP-glucuronosyltransferase isoforms. Drug Metab Dispos 33:977–983PubMedCrossRef Tallman MN, Ritter JK, Smith PC (2005) Differential rates of glucuronidation for 7-ethyl-10-hydroxycamptothecin (SN-38) lactone and carboxylate in human and rat microsomes and recombinant UDP-glucuronosyltransferase isoforms. Drug Metab Dispos 33:977–983PubMedCrossRef
39.
go back to reference Tallman MN, Miles KK, Kessler FK, Nielsen JN, Tian X, Ritter JK, Smith PC (2007) The contribution of intestinal UDP-glucuronosyltransferases in modulating 7-ethyl-10-hydroxy-camptothecin (SN-38)-induced gastrointestinal toxicity in rats. J Pharmacol Exp Ther 320:29–37PubMedCrossRef Tallman MN, Miles KK, Kessler FK, Nielsen JN, Tian X, Ritter JK, Smith PC (2007) The contribution of intestinal UDP-glucuronosyltransferases in modulating 7-ethyl-10-hydroxy-camptothecin (SN-38)-induced gastrointestinal toxicity in rats. J Pharmacol Exp Ther 320:29–37PubMedCrossRef
Metadata
Title
Involvement of UDP-glucuronosyltransferase activity in irinotecan-induced delayed-onset diarrhea in rats
Authors
Masaharu Onoue
Akinobu Kurita
Shoichi Kado
Tsuneo Matsumoto
Norimasa Kaneda
Kazumi Uchida
Ikuo Kato
Teruo Yokokura
Publication date
01-04-2008
Publisher
Springer-Verlag
Published in
Cancer Chemotherapy and Pharmacology / Issue 4/2008
Print ISSN: 0344-5704
Electronic ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-007-0512-x

Other articles of this Issue 4/2008

Cancer Chemotherapy and Pharmacology 4/2008 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