Optimal Sampling Strategies for Irinotecan (CPT-11) and its Active Metabolite (SN-38) in Cancer Patients

Irinotecan (CPT-11) is an anticancer agent widely used in the treatment of a variety of adult solid tumors. The objective of this study was to develop an optimal sampling strategy model that accurately estimates pharmacokinetic parameters of CPT-11 and its active metabolite, SN-38. This study included 221 patients with advanced solid tumors or lymphoma receiving CPT-11 single or combination therapy with 5-fluorouracil (5-FU)/leucovorin (LV) (FOLFIRI) plus bevacizumab from 4 separate clinical trials. Population pharmacokinetic analysis of CPT-11 and SN-38 was performed by non-linear mixed effects modeling. The optimal sampling strategy model was developed using D-optimality with expected distribution approach. The pharmacokinetic profiles of CPT-11 and SN-38 were best described by a 3- and 2-compartment model, respectively, with first-order elimination. Body surface area and co-administration with 5-FU/LV plus bevacizumab were significant covariates (p < 0.01) for volumes of the central compartment of CPT-11 and SN-38, and clearance of CPT-11. Pre-treatment total bilirubin and co-administration with 5-FU/LV and bevacizumab were significant covariates (p < 0.01) for clearance of SN-38. Accurate and precise predictive performance (r2 > 0.99, -2 < bias (%ME) < 0, precision (% RMSE) < 12) of both CPT-11 and SN-38 was achieved using: (i) 6 fixed sampling times collected at 1.5, 3.5, 4, 5.75, 22, 23.5 hours post-infusion; or (ii) 1 fixed time and 2 sampling windows collected at 1.5, [3-5.75], [22-23.5] hours post-infusion. The present study demonstrates that an optimal sampling design with three blood samples achieves accurate and precise pharmacokinetic parameter estimates for both CPT-11 and SN-38.

[1]  R. Mathijssen,et al.  Individualization of Irinotecan Treatment: A Review of Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics , 2018, Clinical Pharmacokinetics.

[2]  F. Innocenti,et al.  Genotype-Guided Dosing Study of FOLFIRI plus Bevacizumab in Patients with Metastatic Colorectal Cancer , 2016, Clinical Cancer Research.

[3]  Soma Das,et al.  Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[4]  Andrew C. Hooker,et al.  PopED: An extended, parallelized, nonlinear mixed effects models optimal design tool , 2012, Comput. Methods Programs Biomed..

[5]  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.

[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]  E. Wiemer,et al.  A CYP3A4 Phenotype–Based Dosing Algorithm for Individualized Treatment of Irinotecan , 2010, Clinical Cancer Research.

[8]  Soma Das,et al.  Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[9]  Leon Aarons,et al.  An Effective Approach for Obtaining Optimal Sampling Windows for Population Pharmacokinetic Experiments , 2008, Journal of biopharmaceutical statistics.

[10]  M. Ratain,et al.  Pharmacogenetic Testing for Uridine Diphosphate Glucuronosyltransferase 1A1 Polymorphisms: Are We There Yet? , 2008, Pharmacotherapy.

[11]  F. Pinguet,et al.  A limited sampling strategy to estimate the pharmacokinetic parameters of irinotecan and its active metabolite, SN-38, in patients with metastatic digestive cancer receiving the FOLFIRI regimen. , 2007, Oncology reports.

[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]  J. Verweij,et al.  Prophylaxis of irinotecan-induced diarrhea with neomycin and potential role for UGT1A1*28 genotype screening: a double-blind, randomized, placebo-controlled study. , 2006, The oncologist.

[14]  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.

[15]  R. Goldberg,et al.  Phase 1 and Pharmacokinetic Study of Intravenous Irinotecan in Refractory Solid Tumor Patients with Hepatic Dysfunction , 2006, Clinical Cancer Research.

[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]  David Z. D'Argenio,et al.  Optimal sampling times for pharmacokinetic experiments , 1981, Journal of Pharmacokinetics and Biopharmaceutics.

[18]  J. Verweij,et al.  Prediction of irinotecan pharmacokinetics by use of cytochrome P450 3A4 phenotyping probes. , 2004, Journal of the National Cancer Institute.

[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. Foracchia,et al.  POPED, a software for optimal experiment design in population kinetics , 2004, Comput. Methods Programs Biomed..

[21]  Stuart L. Beal,et al.  Ways to Fit a PK Model with Some Data Below the Quantification Limit , 2001, Journal of Pharmacokinetics and Pharmacodynamics.

[22]  J. Robert,et al.  Identification and kinetics of a β-glucuronide metabolite of SN-38 in human plasma after administration of the camptothecin derivative irinotecan , 2004, Cancer Chemotherapy and Pharmacology.

[23]  P. Adamson,et al.  The importance of pharmacokinetic limited sampling models for childhood cancer drug development. , 2003, Clinical cancer research : an official journal of the American Association for Cancer Research.

[24]  C. Fuchs,et al.  Phase III comparison of two irinotecan dosing regimens in second-line therapy of metastatic colorectal cancer. , 2003, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  Mark J Ratain,et al.  Population pharmacokinetic model for irinotecan and two of its metabolites, SN‐38 and SN‐38 glucuronide , 2002, Clinical pharmacology and therapeutics.

[26]  J. Verweij,et al.  Clinical pharmacokinetics of irinotecan and its metabolites in relation with diarrhea , 2002, Clinical pharmacology and therapy.

[27]  J. Verweij,et al.  Clinical pharmacokinetics of irinotecan and its metabolites: a population analysis. , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  J. Verweij,et al.  Irinotecan pharmacokinetics-pharmacodynamics: the clinical relevance of prolonged exposure to SN-38 , 2002, British Journal of Cancer.

[29]  M. Ratain Irinotecan dosing: does the CPT in CPT-11 stand for "Can't Predict Toxicity"? , 2002, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

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

[31]  R. Goldberg,et al.  Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. , 2001, The New England journal of medicine.

[32]  H. Saka,et al.  Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: a pharmacogenetic analysis. , 2000, Cancer research.

[33]  L. Saltz,et al.  Irinotecan plus fluorouracil and leucovorin for metastatic colorectal cancer. Irinotecan Study Group. , 2000, The New England journal of medicine.

[34]  C. V. Pesheck,et al.  Pharmacokinetics, metabolism, and excretion of irinotecan (CPT-11) following I.V. infusion of [(14)C]CPT-11 in cancer patients. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[35]  J. Verweij,et al.  Sparse-data set analysis for irinotecan and SN-38 pharmacokinetics in cancer patients co-treated with cisplatin. , 1999, Anti-cancer drugs.

[36]  J. Verweij,et al.  Irinotecan (CPT-11) metabolism and disposition in cancer patients. , 1998, Clinical cancer research : an official journal of the American Association for Cancer Research.

[37]  N. Kemeny,et al.  Phase I clinical and pharmacokinetic study of irinotecan, fluorouracil, and leucovorin in patients with advanced solid tumors. , 1996, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[38]  P. Hérait,et al.  Pharmacokinetics and pharmacodynamics of irinotecan during a phase II clinical trial in colorectal cancer. Pharmacology and Molecular Mechanisms Group of the European Organization for Research and Treatment of Cancer. , 1996, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[39]  M. Ratain,et al.  Limited-sampling models for irinotecan pharmacokinetics-pharmacodynamics: prediction of biliary index and intestinal toxicity. , 1996, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[40]  H. Ohmatsu,et al.  Efficient Sampling Strategies for Forecasting Pharmacokinetic Parameters of Irinotecan (CPT‐11): Implication for Area Under the Concentration‐Time Curve Monitoring , 1995, Therapeutic drug monitoring.

[41]  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.

[42]  N. Saijo,et al.  A Limited Sampling Model for Estimating Pharmacokinetics of CPT‐11 and Its Metabolite SN‐38 , 1995, Japanese journal of cancer research : Gann.

[43]  K. Eguchi,et al.  A Pharmacokinetic and Pharmacodynamic Analysis of CPT‐11 and Its Active Metabolite SN‐38 , 1995, Japanese journal of cancer research : Gann.

[44]  P. Hérait,et al.  Phase I and pharmacologic studies of the camptothecin analog irinotecan administered every 3 weeks in cancer patients. , 1995, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[45]  S. Culine,et al.  Phase I and pharmacokinetic study of the camptothecin derivative irinotecan, administered on a weekly schedule in cancer patients. , 1994, Cancer research.

[46]  P. Bosma,et al.  Bilirubin UDP-glucuronosyltransferase 1 is the only relevant bilirubin glucuronidating isoform in man. , 1994, The Journal of biological chemistry.

[47]  L. Grochow,et al.  Phase I and pharmacological study of the novel topoisomerase I inhibitor 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11) administered as a ninety-minute infusion every 3 weeks. , 1994, Cancer research.

[48]  G. Weiss,et al.  Phase I and pharmacokinetic trial of weekly CPT-11. , 1993, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[49]  J. Boehm,et al.  Synthesis of water-soluble (aminoalkyl)camptothecin analogs: inhibition of topoisomerase I and antitumor activity , 1991 .

[50]  D Z D'Argenio,et al.  Incorporating prior parameter uncertainty in the design of sampling schedules for pharmacokinetic parameter estimation experiments. , 1990, Mathematical biosciences.

[51]  G. Schwarz Estimating the Dimension of a Model , 1978 .

[52]  H. Akaike A new look at the statistical model identification , 1974 .