Skip to main content
Erschienen in: Cancer Chemotherapy and Pharmacology 4/2012

01.04.2012 | Short Communication

Ugt1a is required for the protective effect of selenium against irinotecan-induced toxicity

verfasst von: Shousong Cao, Farukh A. Durrani, Youcef M. Rustum, Y. Eugene Yu

Erschienen in: Cancer Chemotherapy and Pharmacology | Ausgabe 4/2012

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Irinotecan (CPT-11) is widely used for the treatment of patients with colorectal cancer. However, the adverse effects associated with the treatment have hindered the efficacies of irinotecan. We have shown that organic selenium compounds could significantly attenuate irinotecan-associated toxicity and enhance antitumor activity in xenograft tumor models. The objective of this study is to determine the role of a specific group of uridine diphosphate glucuronosyltransferases, which is coded by UGT1A, in detoxification process of irinotecan as well as selenium-associated protective effect against irinotecan-induced toxicity.

Methods

In this study, the toxicities of irinotecan, docetaxel and cisplatin in the Ugta1 mutant rats and their wild-type controls were compared. The plasma concentrations of irinotecan and SN-38 were measured. The modulatory effect of a selenium compound on irinotecan-induced toxicity was analyzed in these rats.

Results

We demonstrated that the maximum tolerated doses of irinotecan in the homozygous mutant rats were significantly lower than those in wild-type rats, 25 mg/kg × 1 versus 200 mg/kg × 1 and 3 mg/kg/day × 3 versus 100 mg/kg/day × 3, respectively. The enhanced sensitivity was specific to irinotecan and was not observed with other chemotherapeutic agents, such as docetaxel and cisplatin, where Ugt1a is not required for their metabolism. Our results also showed that selective protection against irinotecan-induced toxicity by 5-methylselenocysteine was achieved in the wild-type rats but not in the Ugt1a null rats.

Conclusion

These data support the hypothesis that expression of UGT1A is critical for 5-methylselenocysteine to exert its protective effect against irinotecan-induced toxicity.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat Ma MK, McLeod HL (2003) Lessons learned from the irinotecan metabolic pathway. Curr Med Chem 10:41–49PubMedCrossRef Ma MK, McLeod HL (2003) Lessons learned from the irinotecan metabolic pathway. Curr Med Chem 10:41–49PubMedCrossRef
2.
Zurück zum Zitat Rivory LP (1996) Irinotecan (CPT-11): a brief overview. Clin Exp Pharmacol Physiol 23:1000–1004PubMedCrossRef Rivory LP (1996) Irinotecan (CPT-11): a brief overview. Clin Exp Pharmacol Physiol 23:1000–1004PubMedCrossRef
3.
Zurück zum Zitat Lavelle F, Bissery MC, Andre S, Roquet F, Riou JF (1996) Preclinical evaluation of CPT-11 and its active metabolite SN-38. Semin Oncol 23:11–20PubMed Lavelle F, Bissery MC, Andre S, Roquet F, Riou JF (1996) Preclinical evaluation of CPT-11 and its active metabolite SN-38. Semin Oncol 23:11–20PubMed
4.
Zurück zum Zitat Rivory LP, Bowles MR, Robert J, Pond SM (1996) Conversion of irinotecan (CPT-11) to its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38), by human liver carboxylesterase. Biochem Pharmacol 52:1103–1111PubMedCrossRef Rivory LP, Bowles MR, Robert J, Pond SM (1996) Conversion of irinotecan (CPT-11) to its active metabolite, 7-ethyl-10-hydroxycamptothecin (SN-38), by human liver carboxylesterase. Biochem Pharmacol 52:1103–1111PubMedCrossRef
5.
Zurück zum Zitat Wiseman LR, Markham A (1996) Irinotecan. A review of its pharmacological properties and clinical efficacy in the management of advanced colorectal cancer. Drugs 52:606–623PubMedCrossRef Wiseman LR, Markham A (1996) Irinotecan. A review of its pharmacological properties and clinical efficacy in the management of advanced colorectal cancer. Drugs 52:606–623PubMedCrossRef
6.
Zurück zum Zitat Satoh T, Hosokawa M, Atsumi R, Suzuki W, Hakusui H, Nagai E (1994) Metabolic activation of CPT-11, 7-ethyl-10-[4-(1-piperidino)-1- piperidino]carbonyloxycamptothecin, a novel antitumor agent, by carboxylesterase. Biol Pharm Bull 17:662–664PubMedCrossRef Satoh T, Hosokawa M, Atsumi R, Suzuki W, Hakusui H, Nagai E (1994) Metabolic activation of CPT-11, 7-ethyl-10-[4-(1-piperidino)-1- piperidino]carbonyloxycamptothecin, a novel antitumor agent, by carboxylesterase. Biol Pharm Bull 17:662–664PubMedCrossRef
7.
Zurück zum Zitat Saltz LB, Cox JV, Blanke C, Rosen LS, Fehrenbacher L, Moore MJ, Maroun JA, Ackland SP, Locker PK, Pirotta N, Elfring GL, Miller LL (2000) Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan study group. N Engl J Med 343:905–914PubMedCrossRef Saltz LB, Cox JV, Blanke C, Rosen LS, Fehrenbacher L, Moore MJ, Maroun JA, Ackland SP, Locker PK, Pirotta N, Elfring GL, Miller LL (2000) Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan study group. N Engl J Med 343:905–914PubMedCrossRef
8.
Zurück zum Zitat Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P, Jandik P, Iveson T, Carmichael J, Alakl M, Gruia G, Awad L, Rougier P (2000) Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised trial. Lancet 355:1041–1047PubMedCrossRef Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P, Jandik P, Iveson T, Carmichael J, Alakl M, Gruia G, Awad L, Rougier P (2000) Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised trial. Lancet 355:1041–1047PubMedCrossRef
9.
Zurück zum Zitat 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
10.
Zurück zum Zitat 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 39:440–444PubMedCrossRef 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 39:440–444PubMedCrossRef
11.
Zurück zum Zitat 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
12.
Zurück zum Zitat Atsumi R, Suzuki W, Hakusui H (1991) Identification of the metabolites of irinotecan, a new derivative of camptothecin, in rat bile and its biliary excretion. Xenobiotica 21:1159–1169PubMedCrossRef Atsumi R, Suzuki W, Hakusui H (1991) Identification of the metabolites of irinotecan, a new derivative of camptothecin, in rat bile and its biliary excretion. Xenobiotica 21:1159–1169PubMedCrossRef
13.
Zurück zum Zitat 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
14.
Zurück zum Zitat Rivory LP, Riou JF, Haaz MC, Sable S, Vuilhorgne M, Commercon A, Pond SM, Robert J (1996) Identification and properties of a major plasma metabolite of irinotecan (CPT-11) isolated from the plasma of patients. Cancer Res 56:3689–3694PubMed Rivory LP, Riou JF, Haaz MC, Sable S, Vuilhorgne M, Commercon A, Pond SM, Robert J (1996) Identification and properties of a major plasma metabolite of irinotecan (CPT-11) isolated from the plasma of patients. Cancer Res 56:3689–3694PubMed
15.
Zurück zum Zitat Cao S, Durrani FA, Rustum YM (2004) Selective modulation of the therapeutic efficacy of anticancer drugs by selenium containing compounds against human tumor xenografts. Clin Cancer Res 10:2561–2569PubMedCrossRef Cao S, Durrani FA, Rustum YM (2004) Selective modulation of the therapeutic efficacy of anticancer drugs by selenium containing compounds against human tumor xenografts. Clin Cancer Res 10:2561–2569PubMedCrossRef
16.
Zurück zum Zitat Fakih M, Cao S, Durrani FA, Rustum YM (2005) Selenium protects against toxicity induced by anticancer drugs and augments antitumor activity: a highly selective, new, and novel approach for the treatment of solid tumors. Clin Colorectal Cancer 5:132–135PubMedCrossRef Fakih M, Cao S, Durrani FA, Rustum YM (2005) Selenium protects against toxicity induced by anticancer drugs and augments antitumor activity: a highly selective, new, and novel approach for the treatment of solid tumors. Clin Colorectal Cancer 5:132–135PubMedCrossRef
17.
Zurück zum Zitat Iyanagi T (1991) Molecular basis of multiple UDP-glucuronosyltransferase isoenzyme deficiencies in the hyperbilirubinemic rat (Gunn rat). J Biol Chem 266:24048–24052PubMed Iyanagi T (1991) Molecular basis of multiple UDP-glucuronosyltransferase isoenzyme deficiencies in the hyperbilirubinemic rat (Gunn rat). J Biol Chem 266:24048–24052PubMed
18.
Zurück zum Zitat Roy-Chowdhury J, Huang TJ, Kesari K, Lederstein M, Arias IM, Roy-Chowdhury N (1991) Molecular basis for the lack of bilirubin-specific and 3-methylcholanthrene-inducible UDP-glucuronosyltransferase activities in Gunn rats. The two isoforms are encoded by distinct mRNA species that share an identical single base deletion. J Biol Chem 266:18294–18298PubMed Roy-Chowdhury J, Huang TJ, Kesari K, Lederstein M, Arias IM, Roy-Chowdhury N (1991) Molecular basis for the lack of bilirubin-specific and 3-methylcholanthrene-inducible UDP-glucuronosyltransferase activities in Gunn rats. The two isoforms are encoded by distinct mRNA species that share an identical single base deletion. J Biol Chem 266:18294–18298PubMed
19.
Zurück zum Zitat 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
20.
Zurück zum Zitat Warner DL, Burke TG (1997) Simple and versatile high-performance liquid chromatographic method for the simultaneous quantitation of the lactone and carboxylate forms of camptothecin anticancer drugs. J Chromatogr B Biomed Sci Appl 691:161–171PubMedCrossRef Warner DL, Burke TG (1997) Simple and versatile high-performance liquid chromatographic method for the simultaneous quantitation of the lactone and carboxylate forms of camptothecin anticancer drugs. J Chromatogr B Biomed Sci Appl 691:161–171PubMedCrossRef
21.
Zurück zum Zitat Ando Y, Saka H, Asai G, Sugiura S, Shimokata K, Kamataki T (1998) UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Ann Oncol 9:845–847PubMedCrossRef Ando Y, Saka H, Asai G, Sugiura S, Shimokata K, Kamataki T (1998) UGT1A1 genotypes and glucuronidation of SN-38, the active metabolite of irinotecan. Ann Oncol 9:845–847PubMedCrossRef
22.
Zurück zum Zitat 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
23.
Zurück zum Zitat Toffoli G, Cecchin E (2004) Uridine diphosphoglucuronosyl transferase and methylenetetrahydrofolate reductase polymorphisms as genomic predictors of toxicity and response to irinotecan-, antifolate- and fluoropyrimidine-based chemotherapy. J Chemother 16(Suppl 4):31–35PubMed Toffoli G, Cecchin E (2004) Uridine diphosphoglucuronosyl transferase and methylenetetrahydrofolate reductase polymorphisms as genomic predictors of toxicity and response to irinotecan-, antifolate- and fluoropyrimidine-based chemotherapy. J Chemother 16(Suppl 4):31–35PubMed
24.
Zurück zum Zitat Santos A, Zanetta S, Cresteil T, Deroussent A, Pein F, Raymond E, Vernillet L, Risse ML, Boige V, Gouyette A, Vassal G (2000) Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Clin Cancer Res 6:2012–2020PubMed Santos A, Zanetta S, Cresteil T, Deroussent A, Pein F, Raymond E, Vernillet L, Risse ML, Boige V, Gouyette A, Vassal G (2000) Metabolism of irinotecan (CPT-11) by CYP3A4 and CYP3A5 in humans. Clin Cancer Res 6:2012–2020PubMed
25.
Zurück zum Zitat Azrak RG, Yu J, Pendyala L, Smith PF, Cao S, Li X, Shannon WD, Durrani FA, McLeod HL, Rustum YM (2005) Irinotecan pharmacokinetic and pharmacogenomic alterations induced by methylselenocysteine in human head and neck xenograft tumors. Mol Cancer Ther 4:843–854PubMedCrossRef Azrak RG, Yu J, Pendyala L, Smith PF, Cao S, Li X, Shannon WD, Durrani FA, McLeod HL, Rustum YM (2005) Irinotecan pharmacokinetic and pharmacogenomic alterations induced by methylselenocysteine in human head and neck xenograft tumors. Mol Cancer Ther 4:843–854PubMedCrossRef
26.
Zurück zum Zitat Ip C, Lisk DJ (1997) Modulation of phase I and phase II xenobiotic-metabolizing enzymes by selenium-enriched garlic in rats. Nutr Cancer 28:184–188PubMedCrossRef Ip C, Lisk DJ (1997) Modulation of phase I and phase II xenobiotic-metabolizing enzymes by selenium-enriched garlic in rats. Nutr Cancer 28:184–188PubMedCrossRef
27.
Zurück zum Zitat Obol’skii OL, Kravchenko LV, Avren’eva LI, Tutel’ian VA (1998) Effect of dietary selenium on the activity of UDP-glucuronosyltransferases and metabolism of mycotoxin deoxynivalenol in rats. Vopr Pitan 67:18–23 Obol’skii OL, Kravchenko LV, Avren’eva LI, Tutel’ian VA (1998) Effect of dietary selenium on the activity of UDP-glucuronosyltransferases and metabolism of mycotoxin deoxynivalenol in rats. Vopr Pitan 67:18–23
Metadaten
Titel
Ugt1a is required for the protective effect of selenium against irinotecan-induced toxicity
verfasst von
Shousong Cao
Farukh A. Durrani
Youcef M. Rustum
Y. Eugene Yu
Publikationsdatum
01.04.2012
Verlag
Springer-Verlag
Erschienen in
Cancer Chemotherapy and Pharmacology / Ausgabe 4/2012
Print ISSN: 0344-5704
Elektronische ISSN: 1432-0843
DOI
https://doi.org/10.1007/s00280-011-1820-8

Weitere Artikel der Ausgabe 4/2012

Cancer Chemotherapy and Pharmacology 4/2012 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.