Cargando…

Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors

PURPOSE: Etirinotecan pegol (EP), a long-acting topoisomerase-1 inhibitor, is a polyethylene glycol conjugate of irinotecan, with an intended indication for treatment of breast cancer with brain metastases. The objective of this study was to develop a population pharmacokinetic (popPK) model of EP a...

Descripción completa

Detalles Bibliográficos
Autores principales: Sy, Sherwin K. B., Chia, Yen Lin, Gordi, Toufigh, Hoch, Ute, Eldon, Michael A.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer Berlin Heidelberg 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5907632/
https://www.ncbi.nlm.nih.gov/pubmed/29564497
http://dx.doi.org/10.1007/s00280-018-3562-3
_version_ 1783315571900678144
author Sy, Sherwin K. B.
Chia, Yen Lin
Gordi, Toufigh
Hoch, Ute
Eldon, Michael A.
author_facet Sy, Sherwin K. B.
Chia, Yen Lin
Gordi, Toufigh
Hoch, Ute
Eldon, Michael A.
author_sort Sy, Sherwin K. B.
collection PubMed
description PURPOSE: Etirinotecan pegol (EP), a long-acting topoisomerase-1 inhibitor, is a polyethylene glycol conjugate of irinotecan, with an intended indication for treatment of breast cancer with brain metastases. The objective of this study was to develop a population pharmacokinetic (popPK) model of EP and four of its metabolites (irinotecan, SN38, SN38-glucuronide, and APC) and determine covariates affecting their pharmacokinetics. METHODS: Data from 83 cancer patients enrolled in phase 1 studies were used. The model was developed in two stages: (1) concentration–time data were analyzed with a 3-analyte model for EP, irinotecan, and SN38; and (2) a 5-analyte model developed based on expansion of 3-analyte model to include concentration–time data for SN38 glucuronide and APC with parameter values from 3-analyte model fixed. Covariate relationships with parameters were selected based on Wald’s test within the Wald’s Approximation Method approach, first for the 3-analyte model then the 5-analyte model. RESULTS: The final integrated popPK model for the five analytes was a two-compartment per analyte model that followed the metabolic cascade of EP to irinotecan, followed by metabolism of irinotecan to the previously known metabolites, but with altered exposures as compared to administration of irinotecan. With the model developed based on total dose of EP, the population estimates of EP clearance and central volume were 0.237 L/h and 5.5 L, respectively. Patient age, body surface area (BSA), and estimated glomerular filtration rate were found to correlate with EP clearance and BSA with EP central volume. Individuals who were homozygous for UGT1A1*28 genotype had modestly reduced elimination capacity of SN38 compared to heterozygous and wild-type genotypes. Simulations evaluating the clinical importance of significant covariates indicated minimal change in areas under the curve and peak concentrations of EP and SN38. CONCLUSIONS: The pharmacokinetics of EP and four metabolites including the active metabolite SN38 were described by an integrated popPK model. Other than BSA, which was already accounted by a BSA-based dosing scheme, no other covariates were deemed to have clinical implications. No EP starting dose adjustment based on patient demographics and other covariates was deemed necessary. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s00280-018-3562-3) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-5907632
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Springer Berlin Heidelberg
record_format MEDLINE/PubMed
spelling pubmed-59076322018-04-20 Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors Sy, Sherwin K. B. Chia, Yen Lin Gordi, Toufigh Hoch, Ute Eldon, Michael A. Cancer Chemother Pharmacol Original Article PURPOSE: Etirinotecan pegol (EP), a long-acting topoisomerase-1 inhibitor, is a polyethylene glycol conjugate of irinotecan, with an intended indication for treatment of breast cancer with brain metastases. The objective of this study was to develop a population pharmacokinetic (popPK) model of EP and four of its metabolites (irinotecan, SN38, SN38-glucuronide, and APC) and determine covariates affecting their pharmacokinetics. METHODS: Data from 83 cancer patients enrolled in phase 1 studies were used. The model was developed in two stages: (1) concentration–time data were analyzed with a 3-analyte model for EP, irinotecan, and SN38; and (2) a 5-analyte model developed based on expansion of 3-analyte model to include concentration–time data for SN38 glucuronide and APC with parameter values from 3-analyte model fixed. Covariate relationships with parameters were selected based on Wald’s test within the Wald’s Approximation Method approach, first for the 3-analyte model then the 5-analyte model. RESULTS: The final integrated popPK model for the five analytes was a two-compartment per analyte model that followed the metabolic cascade of EP to irinotecan, followed by metabolism of irinotecan to the previously known metabolites, but with altered exposures as compared to administration of irinotecan. With the model developed based on total dose of EP, the population estimates of EP clearance and central volume were 0.237 L/h and 5.5 L, respectively. Patient age, body surface area (BSA), and estimated glomerular filtration rate were found to correlate with EP clearance and BSA with EP central volume. Individuals who were homozygous for UGT1A1*28 genotype had modestly reduced elimination capacity of SN38 compared to heterozygous and wild-type genotypes. Simulations evaluating the clinical importance of significant covariates indicated minimal change in areas under the curve and peak concentrations of EP and SN38. CONCLUSIONS: The pharmacokinetics of EP and four metabolites including the active metabolite SN38 were described by an integrated popPK model. Other than BSA, which was already accounted by a BSA-based dosing scheme, no other covariates were deemed to have clinical implications. No EP starting dose adjustment based on patient demographics and other covariates was deemed necessary. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1007/s00280-018-3562-3) contains supplementary material, which is available to authorized users. Springer Berlin Heidelberg 2018-03-21 2018 /pmc/articles/PMC5907632/ /pubmed/29564497 http://dx.doi.org/10.1007/s00280-018-3562-3 Text en © The Author(s) 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
spellingShingle Original Article
Sy, Sherwin K. B.
Chia, Yen Lin
Gordi, Toufigh
Hoch, Ute
Eldon, Michael A.
Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title_full Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title_fullStr Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title_full_unstemmed Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title_short Integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
title_sort integrated population pharmacokinetics of etirinotecan pegol and its four metabolites in cancer patients with solid tumors
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5907632/
https://www.ncbi.nlm.nih.gov/pubmed/29564497
http://dx.doi.org/10.1007/s00280-018-3562-3
work_keys_str_mv AT sysherwinkb integratedpopulationpharmacokineticsofetirinotecanpegolanditsfourmetabolitesincancerpatientswithsolidtumors
AT chiayenlin integratedpopulationpharmacokineticsofetirinotecanpegolanditsfourmetabolitesincancerpatientswithsolidtumors
AT gorditoufigh integratedpopulationpharmacokineticsofetirinotecanpegolanditsfourmetabolitesincancerpatientswithsolidtumors
AT hochute integratedpopulationpharmacokineticsofetirinotecanpegolanditsfourmetabolitesincancerpatientswithsolidtumors
AT eldonmichaela integratedpopulationpharmacokineticsofetirinotecanpegolanditsfourmetabolitesincancerpatientswithsolidtumors