Cargando…

Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz

PURPOSE: To treat malaria, HIV-infected patients normally receive artemether (80 mg twice daily) concurrently with antiretroviral therapy and drug-drug interactions can potentially occur. Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have...

Descripción completa

Detalles Bibliográficos
Autores principales: Siccardi, Marco, Olagunju, Adeniyi, Seden, Kay, Ebrahimjee, Farid, Rannard, Steve, Back, David, Owen, Andrew
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer-Verlag 2013
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4230487/
https://www.ncbi.nlm.nih.gov/pubmed/25505649
http://dx.doi.org/10.1186/2193-9616-1-4
_version_ 1782344277908520960
author Siccardi, Marco
Olagunju, Adeniyi
Seden, Kay
Ebrahimjee, Farid
Rannard, Steve
Back, David
Owen, Andrew
author_facet Siccardi, Marco
Olagunju, Adeniyi
Seden, Kay
Ebrahimjee, Farid
Rannard, Steve
Back, David
Owen, Andrew
author_sort Siccardi, Marco
collection PubMed
description PURPOSE: To treat malaria, HIV-infected patients normally receive artemether (80 mg twice daily) concurrently with antiretroviral therapy and drug-drug interactions can potentially occur. Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have the potential to reduce artemether pharmacokinetic exposure. The aim of this study was to develop an in vitro in vivo extrapolation (IVIVE) approach to model the interaction between efavirenz and artemether. Artemether dose adjustments were then simulated in order to predict optimal dosing in co-infected patients and inform future interaction study design. METHODS: In vitro data describing the chemical properties, absorption, distribution, metabolism and elimination of efavirenz and artemether were obtained from published literature and included in a physiologically based pharmacokinetic model (PBPK) to predict drug disposition simulating virtual clinical trials. Administration of efavirenz and artemether, alone or in combination, were simulated to mirror previous clinical studies and facilitate validation of the model and realistic interpretation of the simulation. Efavirenz (600 mg once daily) was administered to 50 virtual subjects for 14 days. This was followed by concomitant administration of artemether (80 mg eight hourly) for the first two doses and 80 mg (twice daily) for another two days. RESULTS: Simulated pharmacokinetics and the drug-drug interaction were in concordance with available clinical data. Efavirenz induced first pass metabolism and hepatic clearance, reducing artemether C(max) by 60% and AUC by 80%. Dose increases of artemether, to correct for the interaction, were simulated and a dose of 240 mg was predicted to be sufficient to overcome the interaction and allow therapeutic plasma concentrations of artemether. CONCLUSIONS: The model presented here provides a rational platform to inform the design for a clinical drug interaction study that may save time and resource while the optimal dose is determined empirically. Wider application of IVIVE could help researchers gain a better understanding of the molecular mechanisms underpinning variability in drug disposition.
format Online
Article
Text
id pubmed-4230487
institution National Center for Biotechnology Information
language English
publishDate 2013
publisher Springer-Verlag
record_format MEDLINE/PubMed
spelling pubmed-42304872014-12-11 Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz Siccardi, Marco Olagunju, Adeniyi Seden, Kay Ebrahimjee, Farid Rannard, Steve Back, David Owen, Andrew In Silico Pharmacol Original Research PURPOSE: To treat malaria, HIV-infected patients normally receive artemether (80 mg twice daily) concurrently with antiretroviral therapy and drug-drug interactions can potentially occur. Artemether is a substrate of CYP3A4 and CYP2B6, antiretrovirals such as efavirenz induce these enzymes and have the potential to reduce artemether pharmacokinetic exposure. The aim of this study was to develop an in vitro in vivo extrapolation (IVIVE) approach to model the interaction between efavirenz and artemether. Artemether dose adjustments were then simulated in order to predict optimal dosing in co-infected patients and inform future interaction study design. METHODS: In vitro data describing the chemical properties, absorption, distribution, metabolism and elimination of efavirenz and artemether were obtained from published literature and included in a physiologically based pharmacokinetic model (PBPK) to predict drug disposition simulating virtual clinical trials. Administration of efavirenz and artemether, alone or in combination, were simulated to mirror previous clinical studies and facilitate validation of the model and realistic interpretation of the simulation. Efavirenz (600 mg once daily) was administered to 50 virtual subjects for 14 days. This was followed by concomitant administration of artemether (80 mg eight hourly) for the first two doses and 80 mg (twice daily) for another two days. RESULTS: Simulated pharmacokinetics and the drug-drug interaction were in concordance with available clinical data. Efavirenz induced first pass metabolism and hepatic clearance, reducing artemether C(max) by 60% and AUC by 80%. Dose increases of artemether, to correct for the interaction, were simulated and a dose of 240 mg was predicted to be sufficient to overcome the interaction and allow therapeutic plasma concentrations of artemether. CONCLUSIONS: The model presented here provides a rational platform to inform the design for a clinical drug interaction study that may save time and resource while the optimal dose is determined empirically. Wider application of IVIVE could help researchers gain a better understanding of the molecular mechanisms underpinning variability in drug disposition. Springer-Verlag 2013-03-01 /pmc/articles/PMC4230487/ /pubmed/25505649 http://dx.doi.org/10.1186/2193-9616-1-4 Text en © Siccardi et al.; licensee Springer. 2013 This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Original Research
Siccardi, Marco
Olagunju, Adeniyi
Seden, Kay
Ebrahimjee, Farid
Rannard, Steve
Back, David
Owen, Andrew
Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title_full Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title_fullStr Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title_full_unstemmed Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title_short Use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
title_sort use of a physiologically-based pharmacokinetic model to simulate artemether dose adjustment for overcoming the drug-drug interaction with efavirenz
topic Original Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4230487/
https://www.ncbi.nlm.nih.gov/pubmed/25505649
http://dx.doi.org/10.1186/2193-9616-1-4
work_keys_str_mv AT siccardimarco useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT olagunjuadeniyi useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT sedenkay useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT ebrahimjeefarid useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT rannardsteve useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT backdavid useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz
AT owenandrew useofaphysiologicallybasedpharmacokineticmodeltosimulateartemetherdoseadjustmentforovercomingthedrugdruginteractionwithefavirenz