Cargando…

Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters

Renal clearance of many drugs is mediated by renal organic anion transporters OAT1/3 and inhibition of these transporters may lead to drug‐drug interactions (DDIs). Pyridoxic acid (PDA) and homovanillic acid (HVA) were indicated as potential biomarkers of OAT1/3. The objective of this study was to d...

Descripción completa

Detalles Bibliográficos
Autores principales: Ahmad, Amais, Ogungbenro, Kayode, Kunze, Annett, Jacobs, Frank, Snoeys, Jan, Rostami‐Hodjegan, Amin, Galetin, Aleksandra
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8129719/
https://www.ncbi.nlm.nih.gov/pubmed/33704919
http://dx.doi.org/10.1002/psp4.12610
_version_ 1783694361626673152
author Ahmad, Amais
Ogungbenro, Kayode
Kunze, Annett
Jacobs, Frank
Snoeys, Jan
Rostami‐Hodjegan, Amin
Galetin, Aleksandra
author_facet Ahmad, Amais
Ogungbenro, Kayode
Kunze, Annett
Jacobs, Frank
Snoeys, Jan
Rostami‐Hodjegan, Amin
Galetin, Aleksandra
author_sort Ahmad, Amais
collection PubMed
description Renal clearance of many drugs is mediated by renal organic anion transporters OAT1/3 and inhibition of these transporters may lead to drug‐drug interactions (DDIs). Pyridoxic acid (PDA) and homovanillic acid (HVA) were indicated as potential biomarkers of OAT1/3. The objective of this study was to develop a population pharmacokinetic model for PDA and HVA to support biomarker qualification. Simultaneous fitting of biomarker plasma and urine data in the presence and absence of potent OAT1/3 inhibitor (probenecid, 500 mg every 6 h) was performed. The impact of study design (multiple vs. single dose of OAT1/3 inhibitor) and ability to detect interactions in the presence of weak/moderate OAT1/3 inhibitors was investigated, together with corresponding power calculations. The population models developed successfully described biomarker baseline and PDA/HVA OAT1/3‐mediated interaction data. No prominent effect of circadian rhythm on PDA and HVA individual baseline levels was evident. Renal elimination contributed greater than 80% to total clearance of both endogenous biomarkers investigated. Estimated probenecid unbound in vivo OAT inhibitory constant was up to 6.4‐fold lower than in vitro values obtained with PDA as a probe. The PDA model was successfully verified against independent literature reported datasets. No significant difference in power of DDI detection was found between multiple and single dose study design when using the same total daily dose of 2000 mg probenecid. Model‐based simulations and power calculations confirmed sensitivity and robustness of plasma PDA data to identify weak, moderate, and strong OAT1/3 inhibitors in an adequately powered clinical study to support optimal design of prospective clinical OAT1/3 interaction studies.
format Online
Article
Text
id pubmed-8129719
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-81297192021-05-21 Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters Ahmad, Amais Ogungbenro, Kayode Kunze, Annett Jacobs, Frank Snoeys, Jan Rostami‐Hodjegan, Amin Galetin, Aleksandra CPT Pharmacometrics Syst Pharmacol Research Renal clearance of many drugs is mediated by renal organic anion transporters OAT1/3 and inhibition of these transporters may lead to drug‐drug interactions (DDIs). Pyridoxic acid (PDA) and homovanillic acid (HVA) were indicated as potential biomarkers of OAT1/3. The objective of this study was to develop a population pharmacokinetic model for PDA and HVA to support biomarker qualification. Simultaneous fitting of biomarker plasma and urine data in the presence and absence of potent OAT1/3 inhibitor (probenecid, 500 mg every 6 h) was performed. The impact of study design (multiple vs. single dose of OAT1/3 inhibitor) and ability to detect interactions in the presence of weak/moderate OAT1/3 inhibitors was investigated, together with corresponding power calculations. The population models developed successfully described biomarker baseline and PDA/HVA OAT1/3‐mediated interaction data. No prominent effect of circadian rhythm on PDA and HVA individual baseline levels was evident. Renal elimination contributed greater than 80% to total clearance of both endogenous biomarkers investigated. Estimated probenecid unbound in vivo OAT inhibitory constant was up to 6.4‐fold lower than in vitro values obtained with PDA as a probe. The PDA model was successfully verified against independent literature reported datasets. No significant difference in power of DDI detection was found between multiple and single dose study design when using the same total daily dose of 2000 mg probenecid. Model‐based simulations and power calculations confirmed sensitivity and robustness of plasma PDA data to identify weak, moderate, and strong OAT1/3 inhibitors in an adequately powered clinical study to support optimal design of prospective clinical OAT1/3 interaction studies. John Wiley and Sons Inc. 2021-05-01 2021-05 /pmc/articles/PMC8129719/ /pubmed/33704919 http://dx.doi.org/10.1002/psp4.12610 Text en © 2021 The Authors. CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics. https://creativecommons.org/licenses/by-nc/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ (https://creativecommons.org/licenses/by-nc/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.
spellingShingle Research
Ahmad, Amais
Ogungbenro, Kayode
Kunze, Annett
Jacobs, Frank
Snoeys, Jan
Rostami‐Hodjegan, Amin
Galetin, Aleksandra
Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title_full Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title_fullStr Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title_full_unstemmed Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title_short Population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of OAT1/3 renal transporters
title_sort population pharmacokinetic modeling and simulation to support qualification of pyridoxic acid as endogenous biomarker of oat1/3 renal transporters
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8129719/
https://www.ncbi.nlm.nih.gov/pubmed/33704919
http://dx.doi.org/10.1002/psp4.12610
work_keys_str_mv AT ahmadamais populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT ogungbenrokayode populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT kunzeannett populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT jacobsfrank populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT snoeysjan populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT rostamihodjeganamin populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters
AT galetinaleksandra populationpharmacokineticmodelingandsimulationtosupportqualificationofpyridoxicacidasendogenousbiomarkerofoat13renaltransporters