Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer.

PURPOSE The risk of severe neutropenia from treatment with irinotecan is related in part to UGT1A1*28, a variant that reduces the elimination of SN-38, the active metabolite of irinotecan. We aimed to identify the maximum-tolerated dose (MTD) and dose-limiting toxicity (DLT) of irinotecan in patients with advanced solid tumors stratified by the *1/*1, *1/*28, and *28/*28 genotypes. PATIENTS AND METHODS Sixty-eight patients received an intravenous flat dose of irinotecan every 3 weeks. Forty-six percent of the patients had the *1/*1 genotype, 41% had the *1/*28 genotype, and 13% had the *28/*28 genotype. The starting dose of irinotecan was 700 mg in patients with the *1/*1 and *1/*28 genotypes and 500 mg in patients with the *28/*28 genotype. Pharmacokinetic evaluation was performed at cycle 1. RESULTS In patients with the *1/*1 genotype, the MTD was 850 mg (four DLTs per 16 patients), and 1,000 mg was not tolerated (two DLTs per six patients). In patients with the *1/*28 genotype, the MTD was 700 mg (five DLTs per 22 patients), and 850 mg was not tolerated (four DLTs per six patients). In patients with the *28/*28 genotype, the MTD was 400 mg (one DLT per six patients), and 500 mg was not tolerated (three DLTs per three patients). The DLTs were mainly myelosuppression and diarrhea. Irinotecan clearance followed linear kinetics. At the MTD for each genotype, dosing by genotype resulted in similar SN-38 areas under the curve (AUCs; r(2) = 0.0003; P = .97), but the irinotecan AUC was correlated with the actual dose (r(2) = 0.39; P < .001). Four of 48 patients with disease known to be responsive to irinotecan achieved partial response. CONCLUSION The UGT1A1*28 genotype can be used to individualize dosing of irinotecan. Additional studies should evaluate the effect of genotype-guided dosing on efficacy in patients receiving irinotecan.

[1]  F. Innocenti,et al.  Clinical Implementation of Germ Line Cancer Pharmacogenetic Variants During the Next‐Generation Sequencing Era , 2014, Clinical pharmacology and therapeutics.

[2]  Joshua M. Stuart,et al.  The Cancer Genome Atlas Pan-Cancer analysis project , 2013, Nature Genetics.

[3]  Jan Bogaerts,et al.  Designing transformative clinical trials in the cancer genome era. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  M. Baiget,et al.  A genotype-directed phase I–IV dose-finding study of irinotecan in combination with fluorouracil/leucovorin as first-line treatment in advanced colorectal cancer , 2011, British Journal of Cancer.

[5]  Jeffrey D Sachs,et al.  Expansion of cancer care and control in countries of low and middle income: a call to action , 2010, The Lancet.

[6]  F. Innocenti,et al.  Genotype-driven phase I study of irinotecan administered in combination with fluorouracil/leucovorin in patients with metastatic colorectal cancer. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[7]  Z. Kmietowicz Task force hopes to deliver affordable cancer drugs to developing countries , 2009, BMJ : British Medical Journal.

[8]  R. Schilsky,et al.  Translating the cancer genome into clinically useful tools and strategies , 2009, Disease Models & Mechanisms.

[9]  Wei Zhang,et al.  Hepatocyte nuclear factor-1 alpha is associated with UGT1A1, UGT1A9 and UGT2B7 mRNA expression in human liver , 2008, The Pharmacogenomics Journal.

[10]  W. Humphreys,et al.  Characterization of the UDP Glucuronosyltransferase Activity of Human Liver Microsomes Genotyped for the UGT1A1*28 Polymorphism , 2007, Drug Metabolism and Disposition.

[11]  Joseph G Ibrahim,et al.  UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. , 2007, Journal of the National Cancer Institute.

[12]  Teruhiko Yoshida,et al.  Irinotecan pharmacokinetics/pharmacodynamics and UGT1A genetic polymorphisms in Japanese: roles of UGT1A1*6 and *28 , 2007, Pharmacogenetics and genomics.

[13]  M. Ratain,et al.  The Role of SN‐38 Exposure, UGT1A1*28 Polymorphism, and Baseline Bilirubin Level in Predicting Severe Irinotecan Toxicity , 2007, Journal of clinical pharmacology.

[14]  M. Ratain,et al.  Pharmacogenetics of irinotecan: clinical perspectives on the utility of genotyping. , 2006, Pharmacogenomics.

[15]  Giuseppe Toffoli,et al.  The role of UGT1A1*28 polymorphism in the pharmacodynamics and pharmacokinetics of irinotecan in patients with metastatic colorectal cancer. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[16]  E. Shin,et al.  Comprehensive analysis of UGT1A polymorphisms predictive for pharmacokinetics and treatment outcome in patients with non-small-cell lung cancer treated with irinotecan and cisplatin. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[17]  E. Eisenhauer,et al.  RECIST revisited: a review of validation studies on tumour assessment. , 2006, European journal of cancer.

[18]  J. Meyerhardt,et al.  Systemic therapy for colorectal cancer. , 2005, The New England journal of medicine.

[19]  Soma Das,et al.  Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. , 2004, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  M. Egorin Horseshoes, hand grenades, and body-surface area-based dosing: aiming for a target. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  A. Di Rienzo,et al.  Haplotype structure of the UDP-glucuronosyltransferase 1A1 promoter in different ethnic groups. , 2002, Pharmacogenetics.

[22]  J. Verweij,et al.  Impact of body-size measures on irinotecan clearance: alternative dosing recommendations. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[23]  R. Schilsky,et al.  UGT1A1*28 polymorphism as a determinant of irinotecan disposition and toxicity , 2002, The Pharmacogenomics Journal.

[24]  A. Di Rienzo,et al.  Variability at the uridine diphosphate glucuronosyltransferase 1A1 promoter in human populations and primates. , 1999, Pharmacogenetics.

[25]  A. Di Rienzo,et al.  Phenotype‐genotype correlation of in vitro SN‐38 (active metabolite of irinotecan) and bilirubin glucuronidation in human liver tissue with UGT1A1 promoter polymorphism , 1999, Clinical pharmacology and therapeutics.

[26]  P. Hérait,et al.  High dose-intensity of irinotecan administered every 3 weeks in advanced cancer patients: a feasibility study. , 1997, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[27]  S. Culine,et al.  Population pharmacokinetics and pharmacodynamics of irinotecan (CPT-11) and active metabolite SN-38 during phase I trials. , 1995, Annals of oncology : official journal of the European Society for Medical Oncology.