Cargando…

Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling

Abemaciclib, a selective inhibitor of cyclin‐dependent kinases 4 and 6, is metabolized mainly by cytochrome P450 (CYP)3A4. Clinical studies were performed to assess the impact of strong inhibitor (clarithromycin) and inducer (rifampin) on the exposure of abemaciclib and active metabolites. A physiol...

Descripción completa

Detalles Bibliográficos
Autores principales: Posada, Maria M., Morse, Bridget L., Turner, P. Kellie, Kulanthaivel, Palaniappan, Hall, Stephen D., Dickinson, Gemma L.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7318171/
https://www.ncbi.nlm.nih.gov/pubmed/32080863
http://dx.doi.org/10.1002/jcph.1584
_version_ 1783550784670007296
author Posada, Maria M.
Morse, Bridget L.
Turner, P. Kellie
Kulanthaivel, Palaniappan
Hall, Stephen D.
Dickinson, Gemma L.
author_facet Posada, Maria M.
Morse, Bridget L.
Turner, P. Kellie
Kulanthaivel, Palaniappan
Hall, Stephen D.
Dickinson, Gemma L.
author_sort Posada, Maria M.
collection PubMed
description Abemaciclib, a selective inhibitor of cyclin‐dependent kinases 4 and 6, is metabolized mainly by cytochrome P450 (CYP)3A4. Clinical studies were performed to assess the impact of strong inhibitor (clarithromycin) and inducer (rifampin) on the exposure of abemaciclib and active metabolites. A physiologically based pharmacokinetic (PBPK) model incorporating the metabolites was developed to predict the effect of other strong and moderate CYP3A4 inhibitors and inducers. Clarithromycin increased the area under the plasma concentration‐time curve (AUC) of abemaciclib and potency‐adjusted unbound active species 3.4‐fold and 2.5‐fold, respectively. Rifampin decreased corresponding exposures 95% and 77%, respectively. These changes influenced the fraction metabolized via CYP3A4 in the model. An absolute bioavailability study informed the hepatic and gastric availability. In vitro data and a human radiolabel study determined the fraction and rate of formation of the active metabolites as well as absorption‐related parameters. The predicted AUC ratios of potency‐adjusted unbound active species with rifampin and clarithromycin were within 0.7‐ and 1.25‐fold of those observed. The PBPK model predicted 3.78‐ and 7.15‐fold increases in the AUC of the potency‐adjusted unbound active species with strong CYP3A4 inhibitors itraconazole and ketoconazole, respectively; and 1.62‐ and 2.37‐fold increases with the concomitant use of moderate CYP3A4 inhibitors verapamil and diltiazem, respectively. The model predicted modafinil, bosentan, and efavirenz would decrease the AUC of the potency‐adjusted unbound active species by 29%, 42%, and 52%, respectively. The current PBPK model, which considers changes in unbound potency‐adjusted active species, can be used to inform dosing recommendations when abemaciclib is coadministered with CYP3A4 perpetrators.
format Online
Article
Text
id pubmed-7318171
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-73181712020-06-29 Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling Posada, Maria M. Morse, Bridget L. Turner, P. Kellie Kulanthaivel, Palaniappan Hall, Stephen D. Dickinson, Gemma L. J Clin Pharmacol NON COVID ARTICLES Abemaciclib, a selective inhibitor of cyclin‐dependent kinases 4 and 6, is metabolized mainly by cytochrome P450 (CYP)3A4. Clinical studies were performed to assess the impact of strong inhibitor (clarithromycin) and inducer (rifampin) on the exposure of abemaciclib and active metabolites. A physiologically based pharmacokinetic (PBPK) model incorporating the metabolites was developed to predict the effect of other strong and moderate CYP3A4 inhibitors and inducers. Clarithromycin increased the area under the plasma concentration‐time curve (AUC) of abemaciclib and potency‐adjusted unbound active species 3.4‐fold and 2.5‐fold, respectively. Rifampin decreased corresponding exposures 95% and 77%, respectively. These changes influenced the fraction metabolized via CYP3A4 in the model. An absolute bioavailability study informed the hepatic and gastric availability. In vitro data and a human radiolabel study determined the fraction and rate of formation of the active metabolites as well as absorption‐related parameters. The predicted AUC ratios of potency‐adjusted unbound active species with rifampin and clarithromycin were within 0.7‐ and 1.25‐fold of those observed. The PBPK model predicted 3.78‐ and 7.15‐fold increases in the AUC of the potency‐adjusted unbound active species with strong CYP3A4 inhibitors itraconazole and ketoconazole, respectively; and 1.62‐ and 2.37‐fold increases with the concomitant use of moderate CYP3A4 inhibitors verapamil and diltiazem, respectively. The model predicted modafinil, bosentan, and efavirenz would decrease the AUC of the potency‐adjusted unbound active species by 29%, 42%, and 52%, respectively. The current PBPK model, which considers changes in unbound potency‐adjusted active species, can be used to inform dosing recommendations when abemaciclib is coadministered with CYP3A4 perpetrators. John Wiley and Sons Inc. 2020-02-20 2020-07 /pmc/articles/PMC7318171/ /pubmed/32080863 http://dx.doi.org/10.1002/jcph.1584 Text en © 2020 Eli Lilly and Company. The Journal of Clinical Pharmacology published by Wiley Periodicals, Inc. on behalf of American College of Clinical Pharmacolog This is an open access article under the terms of the http://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 NON COVID ARTICLES
Posada, Maria M.
Morse, Bridget L.
Turner, P. Kellie
Kulanthaivel, Palaniappan
Hall, Stephen D.
Dickinson, Gemma L.
Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title_full Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title_fullStr Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title_full_unstemmed Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title_short Predicting Clinical Effects of CYP3A4 Modulators on Abemaciclib and Active Metabolites Exposure Using Physiologically Based Pharmacokinetic Modeling
title_sort predicting clinical effects of cyp3a4 modulators on abemaciclib and active metabolites exposure using physiologically based pharmacokinetic modeling
topic NON COVID ARTICLES
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7318171/
https://www.ncbi.nlm.nih.gov/pubmed/32080863
http://dx.doi.org/10.1002/jcph.1584
work_keys_str_mv AT posadamariam predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling
AT morsebridgetl predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling
AT turnerpkellie predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling
AT kulanthaivelpalaniappan predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling
AT hallstephend predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling
AT dickinsongemmal predictingclinicaleffectsofcyp3a4modulatorsonabemaciclibandactivemetabolitesexposureusingphysiologicallybasedpharmacokineticmodeling