Cargando…

Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels

Introduction: As the third generation of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), osimertinib has demonstrated more significant cardiotoxicity than previous generations of EGFR-TKIs. Investigating the mechanism of osimertinib cardiotoxicity can provide a reference for a...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Peiwen, Tian, Xiaohui, Wang, Gongxin, Jiang, Enshe, Li, Yanming, Hao, Guoliang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10267729/
https://www.ncbi.nlm.nih.gov/pubmed/37324483
http://dx.doi.org/10.3389/fphar.2023.1177003
_version_ 1785058986658103296
author Li, Peiwen
Tian, Xiaohui
Wang, Gongxin
Jiang, Enshe
Li, Yanming
Hao, Guoliang
author_facet Li, Peiwen
Tian, Xiaohui
Wang, Gongxin
Jiang, Enshe
Li, Yanming
Hao, Guoliang
author_sort Li, Peiwen
collection PubMed
description Introduction: As the third generation of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), osimertinib has demonstrated more significant cardiotoxicity than previous generations of EGFR-TKIs. Investigating the mechanism of osimertinib cardiotoxicity can provide a reference for a comprehensive understanding of osimertinib-induced cardiotoxicity and the safety of the usage of this drug in clinical practice. Methods: Multichannel electrical mapping with synchronous ECG recording was used to investigate the effects of varying osimertinib concentrations on electrophysiological indicators in isolated Langendorff-perfused hearts of guinea pigs. Additionally, a whole-cell patch clamp was used to detect the impact of osimertinib on the currents of hERG channels transfected into HEK293 cells and the Nav1.5 channel transfected into Chinese hamster ovary cells and acute isolated ventricular myocytes from SD rats. Results: Acute exposure to varying osimertinib concentrations produced prolongation in the PR interval, QT interval, and QRS complex in isolated hearts of guinea pigs. Meanwhile, this exposure could concentration-dependently increase the conduction time in the left atrium, left ventricle, and atrioventricular without affecting the left ventricle conduction velocity. Osimertinib inhibited the hERG channel in a concentration-dependent manner, with an IC(50) of 2.21 ± 1.29 μM. Osimertinib also inhibited the Nav1.5 channel in a concentration-dependent manner, with IC(50) values in the absence of inactivation, 20% inactivation, and 50% inactivation of 15.58 ± 0.83 μM, 3.24 ± 0.09 μM, and 2.03 ± 0.57 μM, respectively. Osimertinib slightly inhibited the currents of L-type Ca(2+) channels in a concentration-dependent manner in acutely isolated rat ventricular myocytes. Discussion: Osimertinib could prolong the QT interval; PR interval; QRS complex; left atrium, left ventricle, and atrioventricular conduction time in isolated guinea pig hearts. Furthermore, osimertinib could block the hERG, Nav1.5, and L-type Ca(2+) channels in concentration-dependent manners. Therefore, these findings might be the leading cause of the cardiotoxicity effects, such as QT prolongation and decreased left ventricular ejection fraction.
format Online
Article
Text
id pubmed-10267729
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-102677292023-06-15 Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels Li, Peiwen Tian, Xiaohui Wang, Gongxin Jiang, Enshe Li, Yanming Hao, Guoliang Front Pharmacol Pharmacology Introduction: As the third generation of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), osimertinib has demonstrated more significant cardiotoxicity than previous generations of EGFR-TKIs. Investigating the mechanism of osimertinib cardiotoxicity can provide a reference for a comprehensive understanding of osimertinib-induced cardiotoxicity and the safety of the usage of this drug in clinical practice. Methods: Multichannel electrical mapping with synchronous ECG recording was used to investigate the effects of varying osimertinib concentrations on electrophysiological indicators in isolated Langendorff-perfused hearts of guinea pigs. Additionally, a whole-cell patch clamp was used to detect the impact of osimertinib on the currents of hERG channels transfected into HEK293 cells and the Nav1.5 channel transfected into Chinese hamster ovary cells and acute isolated ventricular myocytes from SD rats. Results: Acute exposure to varying osimertinib concentrations produced prolongation in the PR interval, QT interval, and QRS complex in isolated hearts of guinea pigs. Meanwhile, this exposure could concentration-dependently increase the conduction time in the left atrium, left ventricle, and atrioventricular without affecting the left ventricle conduction velocity. Osimertinib inhibited the hERG channel in a concentration-dependent manner, with an IC(50) of 2.21 ± 1.29 μM. Osimertinib also inhibited the Nav1.5 channel in a concentration-dependent manner, with IC(50) values in the absence of inactivation, 20% inactivation, and 50% inactivation of 15.58 ± 0.83 μM, 3.24 ± 0.09 μM, and 2.03 ± 0.57 μM, respectively. Osimertinib slightly inhibited the currents of L-type Ca(2+) channels in a concentration-dependent manner in acutely isolated rat ventricular myocytes. Discussion: Osimertinib could prolong the QT interval; PR interval; QRS complex; left atrium, left ventricle, and atrioventricular conduction time in isolated guinea pig hearts. Furthermore, osimertinib could block the hERG, Nav1.5, and L-type Ca(2+) channels in concentration-dependent manners. Therefore, these findings might be the leading cause of the cardiotoxicity effects, such as QT prolongation and decreased left ventricular ejection fraction. Frontiers Media S.A. 2023-05-30 /pmc/articles/PMC10267729/ /pubmed/37324483 http://dx.doi.org/10.3389/fphar.2023.1177003 Text en Copyright © 2023 Li, Tian, Wang, Jiang, Li and Hao. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Li, Peiwen
Tian, Xiaohui
Wang, Gongxin
Jiang, Enshe
Li, Yanming
Hao, Guoliang
Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title_full Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title_fullStr Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title_full_unstemmed Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title_short Acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
title_sort acute osimertinib exposure induces electrocardiac changes by synchronously inhibiting the currents of cardiac ion channels
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10267729/
https://www.ncbi.nlm.nih.gov/pubmed/37324483
http://dx.doi.org/10.3389/fphar.2023.1177003
work_keys_str_mv AT lipeiwen acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels
AT tianxiaohui acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels
AT wanggongxin acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels
AT jiangenshe acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels
AT liyanming acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels
AT haoguoliang acuteosimertinibexposureinduceselectrocardiacchangesbysynchronouslyinhibitingthecurrentsofcardiacionchannels