DOI QR코드

DOI QR Code

New in vitro multiple cardiac ion channel screening system for preclinical Torsades de Pointes risk prediction under the Comprehensive in vitro Proarrhythmia Assay concepta

  • Jin Ryeol An (Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety) ;
  • Seo-Yeong Mun (Department of Physiology, Kangwon National University School of Medicine) ;
  • In Kyo Jung (Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety) ;
  • Kwan Soo Kim (Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety) ;
  • Chan Hyeok Kwon (Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety) ;
  • Sun Ok Choi (Pharmacological Research Division, Toxicological Evaluation and Research Department, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety) ;
  • Won Sun Park (Department of Physiology, Kangwon National University School of Medicine)
  • Received : 2023.02.15
  • Accepted : 2023.03.29
  • Published : 2023.05.01

Abstract

Cardiotoxicity, particularly drug-induced Torsades de Pointes (TdP), is a concern in drug safety assessment. The recent establishment of human induced pluripotent stem cell-derived cardiomyocytes (human iPSC-CMs) has become an attractive human-based platform for predicting cardiotoxicity. Moreover, electrophysiological assessment of multiple cardiac ion channel blocks is emerging as an important parameter to recapitulate proarrhythmic cardiotoxicity. Therefore, we aimed to establish a novel in vitro multiple cardiac ion channel screening-based method using human iPSC-CMs to predict the drug-induced arrhythmogenic risk. To explain the cellular mechanisms underlying the cardiotoxicity of three representative TdP high- (sotalol), intermediate- (chlorpromazine), and low-risk (mexiletine) drugs, and their effects on the cardiac action potential (AP) waveform and voltage-gated ion channels were explored using human iPSC-CMs. In a proof-of-principle experiment, we investigated the effects of cardioactive channel inhibitors on the electrophysiological profile of human iPSC-CMs before evaluating the cardiotoxicity of these drugs. In human iPSC-CMs, sotalol prolonged the AP duration and reduced the total amplitude (TA) via selective inhibition of IKr and INa currents, which are associated with an increased risk of ventricular tachycardia TdP. In contrast, chlorpromazine did not affect the TA; however, it slightly increased AP duration via balanced inhibition of IKr and ICa currents. Moreover, mexiletine did not affect the TA, yet slightly reduced the AP duration via dominant inhibition of ICa currents, which are associated with a decreased risk of ventricular tachycardia TdP. Based on these results, we suggest that human iPSC-CMs can be extended to other preclinical protocols and can supplement drug safety assessments.

Keywords

Acknowledgement

This study was supported by a grant (21181MFDS276) from the Ministry of Food and Drug Safety, Republic of Korea from 2021-2022.

References

  1. Kannankeril PJ, Roden DM. Drug-induced long QT and torsade de pointes: recent advances. Curr Opin Cardiol. 2007;22:39-43.  https://doi.org/10.1097/HCO.0b013e32801129eb
  2. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3:711-715.  https://doi.org/10.1038/nrd1470
  3. Food and Drug Administration, HHS. International Conference on Harmonisation; guidance on E14 Clinical Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential for Non-Antiarrhythmic Drugs; availability. Notice. Fed Regist. 2005;70:61134-61135. 
  4. Food and Drug Administration, HHS. International Conference on Harmonisation; guidance on S7B Nonclinical Evaluation of the Potential for Delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceuticals; availability. Notice. Fed Regist. 2005;70:61133-61134. 
  5. Guo L, Coyle L, Abrams RM, Kemper R, Chiao ET, Kolaja KL. Refining the human iPSC-cardiomyocyte arrhythmic risk assessment model. Toxicol Sci. 2013;136:581-594.  https://doi.org/10.1093/toxsci/kft205
  6. Cavero I, Holzgrefe H. Comprehensive in vitro Proarrhythmia Assay, a novel in vitro/in silico paradigm to detect ventricular proarrhythmic liability: a visionary 21st century initiative. Expert Opin Drug Saf. 2014;13:745-758. 
  7. Sager PT, Gintant G, Turner JR, Pettit S, Stockbridge N. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. Am Heart J. 2014;167:292-300.  https://doi.org/10.1016/j.ahj.2013.11.004
  8. Magyar J, Iost N, Kortvely A, Banyasz T, Virag L, Szigligeti P, Varro A, Opincariu M, Szecsi J, Papp JG, Nanasi PP. Effects of endothelin-1 on calcium and potassium currents in undiseased human ventricular myocytes. Pflugers Arch. 2000;441:144-149.  https://doi.org/10.1007/s004240000400
  9. Ando H, Yoshinaga T, Yamamoto W, Asakura K, Uda T, Taniguchi T, Ojima A, Shinkyo R, Kikuchi K, Osada T, Hayashi S, Kasai C, Miyamoto N, Tashibu H, Yamazaki D, Sugiyama A, Kanda Y, Sawada K, Sekino Y. A new paradigm for drug-induced torsadogenic risk assessment using human iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Methods. 2017;84:111-127.  https://doi.org/10.1016/j.vascn.2016.12.003
  10. Cavero I, Crumb W. ICH S7B draft guideline on the non-clinical strategy for testing delayed cardiac repolarisation risk of drugs: a critical analysis. Expert Opin Drug Saf. 2005;4:509-530.  https://doi.org/10.1517/14740338.4.3.509
  11. Giorgi MA, Bolanos R, Gonzalez CD, Di Girolamo G. QT interval prolongation: preclinical and clinical testing arrhythmogenesis in drugs and regulatory implications. Curr Drug Saf. 2010;5:54-57.  https://doi.org/10.2174/157488610789869148
  12. Lu HR, Marien R, Saels A, De Clerck F. Species plays an important role in drug-induced prolongation of action potential duration and early afterdepolarizations in isolated Purkinje fibers. J Cardiovasc Electrophysiol. 2001;12:93-102.  https://doi.org/10.1046/j.1540-8167.2001.00093.x
  13. Bussek A, Wettwer E, Christ T, Lohmann H, Camelliti P, Ravens U. Tissue slices from adult mammalian hearts as a model for pharmacological drug testing. Cell Physiol Biochem. 2009;24:527-536.  https://doi.org/10.1159/000257528
  14. Thomas D, Wu K, Kathofer S, Katus HA, Schoels W, Kiehn J, Karle CA. The antipsychotic drug chlorpromazine inhibits HERG potassium channels. Br J Pharmacol. 2003;139:567-574.  https://doi.org/10.1038/sj.bjp.0705283
  15. Gualdani R, Tadini-Buoninsegni F, Roselli M, Defrenza I, Contino M, Colabufo NA, Lentini G. Inhibition of hERG potassium channel by the antiarrhythmic agent mexiletine and its metabolite mhydroxymexiletine. Pharmacol Res Perspect. 2015;3:e00160. 
  16. Crumb WJ Jr, Vicente J, Johannesen L, Strauss DG. An evaluation of 30 clinical drugs against the comprehensive in vitro proarrhythmia assay (CiPA) proposed ion channel panel. J Pharmacol Toxicol Methods. 2016;81:251-262.  https://doi.org/10.1016/j.vascn.2016.03.009
  17. Katchman AN, Koerner J, Tosaka T, Woosley RL, Ebert SN. Comparative evaluation of HERG currents and QT intervals following challenge with suspected torsadogenic and nontorsadogenic drugs. J Pharmacol Exp Ther. 2006;316:1098-1106.  https://doi.org/10.1124/jpet.105.093393
  18. Gintant GA, Su Z, Martin RL, Cox BF. Utility of hERG assays as surrogate markers of delayed cardiac repolarization and QT safety. Toxicol Pathol. 2006;34:81-90.  https://doi.org/10.1080/01926230500431376
  19. Belardinelli L, Liu G, Smith-Maxwell C, Wang WQ, El-Bizri N, Hirakawa R, Karpinski S, Li CH, Hu L, Li XJ, Crumb W, Wu L, Koltun D, Zablocki J, Yao L, Dhalla AK, Rajamani S, Shryock JC. A novel, potent, and selective inhibitor of cardiac late sodium current suppresses experimental arrhythmias. J Pharmacol Exp Ther. 2013;344:23-32.  https://doi.org/10.1124/jpet.112.198887
  20. Johannesen L, Vicente J, Mason JW, Sanabria C, Waite-Labott K, Hong M, Guo P, Lin J, Sorensen JS, Galeotti L, Florian J, Ugander M, Stockbridge N, Strauss DG. Differentiating drug-induced multichannel block on the electrocardiogram: randomized study of dofetilide, quinidine, ranolazine, and verapamil. Clin Pharmacol Ther. 2014;96:549-558.  https://doi.org/10.1038/clpt.2014.155
  21. Fermini B, Hancox JC, Abi-Gerges N, Bridgland-Taylor M, Chaudhary KW, Colatsky T, Correll K, Crumb W, Damiano B, Erdemli G, Gintant G, Imredy J, Koerner J, Kramer J, Levesque P, Li Z, Lindqvist A, Obejero-Paz CA, Rampe D, Sawada K, et al. A new perspective in the field of cardiac safety testing through the comprehensive in vitro Proarrhythmia Assay paradigm. J Biomol Screen. 2016;21:1-11. https://doi.org/10.1177/1087057115594589