1
|
Rahmani K, Yang Y, Foster EP, Tsai CT, Meganathan DP, Alvarez DD, Gupta A, Cui B, Santoro F, Bloodgood BL, Yu R, Forro C, Jahed Z. Intelligent in-cell electrophysiology: Reconstructing intracellular action potentials using a physics-informed deep learning model trained on nanoelectrode array recordings. Nat Commun 2025; 16:657. [PMID: 39809732 PMCID: PMC11733287 DOI: 10.1038/s41467-024-55571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Intracellular electrophysiology is essential in neuroscience, cardiology, and pharmacology for studying cells' electrical properties. Traditional methods like patch-clamp are precise but low-throughput and invasive. Nanoelectrode Arrays (NEAs) offer a promising alternative by enabling simultaneous intracellular and extracellular action potential (iAP and eAP) recordings with high throughput. However, accessing intracellular potentials with NEAs remains challenging. This study presents an AI-supported technique that leverages thousands of synchronous eAP and iAP pairs from stem-cell-derived cardiomyocytes on NEAs. Our analysis revealed strong correlations between specific eAP and iAP features, such as amplitude and spiking velocity, indicating that extracellular signals could be reliable indicators of intracellular activity. We developed a physics-informed deep learning model to reconstruct iAP waveforms from extracellular recordings recorded from NEAs and Microelectrode arrays (MEAs), demonstrating its potential for non-invasive, long-term, high-throughput drug cardiotoxicity assessments. This AI-based model paves the way for future electrophysiology research across various cell types and drug interactions.
Collapse
Affiliation(s)
- Keivan Rahmani
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, USA
| | - Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Ethan Paul Foster
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
| | - Dhivya Pushpa Meganathan
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, USA
| | - Diego D Alvarez
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Aayush Gupta
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy
- Neuroelectronic Interfaces, Faculty of Electrical Engineering and IT, RWTH Aachen, Aachen, Germany
- Institute of Biological Information Processing-Bioelectronics, IBI-3, Forschungszentrum, Juelich, Germany
| | - Brenda L Bloodgood
- Department of Neurobiology, School of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rose Yu
- Department of Computer Science and Engineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Csaba Forro
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Wu-Tsai Neuroscience Institute and ChEM-H Institute, Stanford University, Stanford, CA, USA.
- Center for Advanced Biomaterials for Healthcare, Istituto Italiano di Tecnologia, Naples, Italy.
- Neuroelectronic Interfaces, Faculty of Electrical Engineering and IT, RWTH Aachen, Aachen, Germany.
- Institute of Biological Information Processing-Bioelectronics, IBI-3, Forschungszentrum, Juelich, Germany.
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA.
| | - Zeinab Jahed
- Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, USA.
- Shu Chien-Gene Lay Department of Bioengineering, Jacobs School of Engineering, University of California, San Diego, CA, USA.
| |
Collapse
|
2
|
Heitmann S, Vandenberg JI, Hill AP. Assessing drug safety by identifying the axis of arrhythmia in cardiomyocyte electrophysiology. eLife 2023; 12:RP90027. [PMID: 38079357 PMCID: PMC10712948 DOI: 10.7554/elife.90027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Many classes of drugs can induce fatal cardiac arrhythmias by disrupting the electrophysiology of cardiomyocytes. Safety guidelines thus require all new drugs to be assessed for pro-arrhythmic risk prior to conducting human trials. The standard safety protocols primarily focus on drug blockade of the delayed-rectifier potassium current (IKr). Yet the risk is better assessed using four key ion currents (IKr, ICaL, INaL, IKs). We simulated 100,000 phenotypically diverse cardiomyocytes to identify the underlying relationship between the blockade of those currents and the emergence of ectopic beats in the action potential. We call that relationship the axis of arrhythmia. It serves as a yardstick for quantifying the arrhythmogenic risk of any drug from its profile of multi-channel block alone. We tested it on 109 drugs and found that it predicted the clinical risk labels with an accuracy of 88.1-90.8%. Pharmacologists can use our method to assess the safety of novel drugs without resorting to animal testing or unwieldy computer simulations.
Collapse
Affiliation(s)
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research InstituteDarlinghurstAustralia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South WalesSydneyAustralia
| | - Adam P Hill
- Victor Chang Cardiac Research InstituteDarlinghurstAustralia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South WalesSydneyAustralia
- Victor Chang Cardiac Research Institute Innovation CentreDarlinghurstAustralia
| |
Collapse
|
3
|
Kostrubsky V, Liu Y, Muste C, Gu C, Kirkland M, Nishimura N, Hasegawa K, Hasumi K, Yuan L. Preclinical safety, toxicokinetics and metabolism of BIIB131, a novel prothrombolytic agent for acute stroke. Regul Toxicol Pharmacol 2023; 145:105498. [PMID: 37778433 DOI: 10.1016/j.yrtph.2023.105498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/23/2023] [Accepted: 09/28/2023] [Indexed: 10/03/2023]
Abstract
BIIB131, a small molecule, is currently in Phase 2 for the treatment of acute ischemic stroke. Safety and metabolism of BIIB131 were evaluated following intravenous administration to rats and monkeys. Exposure increased dose-proportionally in rats up to 60 mg/kg and more than dose-proportionally in monkeys at greater than 10 mg/kg accompanied by prolonged half-life and safety findings. The BIIB131 was poorly metabolized in microsomes with no inhibition of CYPs. BIIB131-glucuronide, formed by UGT1A1, accounted for 21.5% metabolism in human hepatocytes and 28-40% in rat bile. In rats, excretion was primarily via the bile. BIIB131 inhibited the hERG and Nav1.5 cardiac channels by 39% but showed no effect on cardiovascular parameters in monkeys. Toxicology findings were limited to reversable hematuria, changes in urinary parameters and local effects. A MTD of 30 mg/kg was established in monkeys, the most sensitive species, at total plasma Cmax and AUC of 6- and 14-fold, respectively, greater than the NOAEL. The Phase 1 study started with intravenous 0.05 mg/kg and ascended to 6.0 mg/kg which corresponded to safety margins of 147- to 0.9-fold (for Cmax) within the linear drug exposure. Thus, the preclinical profile of BIIB131 has been appropriately characterized and supports its further clinical development.
Collapse
Affiliation(s)
- Vick Kostrubsky
- Nonclinical Safety Science, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA.
| | - Ying Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Cathy Muste
- Drug Metabolism and Pharmacokinetics, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Chungang Gu
- Drug Metabolism and Pharmacokinetics, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Melissa Kirkland
- Nonclinical Safety Science, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| | - Naoko Nishimura
- Division of Research and Development, TMS Co., Ltd., Tokyo, Japan
| | - Keiko Hasegawa
- Division of Research and Development, TMS Co., Ltd., Tokyo, Japan
| | - Keiji Hasumi
- Division of Research and Development, TMS Co., Ltd., Tokyo, Japan; Department of Applied Biological Science, Tokyo University of Agriculture and Technology, Tokyo, 183-8509, Japan
| | - Long Yuan
- Drug Metabolism and Pharmacokinetics, Biogen, Inc., 225 Binney Street, Cambridge, MA, 02142, USA
| |
Collapse
|
4
|
Furutani K. Facilitation of hERG Activation by Its Blocker: A Mechanism to Reduce Drug-Induced Proarrhythmic Risk. Int J Mol Sci 2023; 24:16261. [PMID: 38003453 PMCID: PMC10671758 DOI: 10.3390/ijms242216261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Modulation of the human Ether-à-go-go-Related Gene (hERG) channel, a crucial voltage-gated potassium channel in the repolarization of action potentials in ventricular myocytes of the heart, has significant implications on cardiac electrophysiology and can be either antiarrhythmic or proarrhythmic. For example, hERG channel blockade is a leading cause of long QT syndrome and potentially life-threatening arrhythmias, such as torsades de pointes. Conversely, hERG channel blockade is the mechanism of action of Class III antiarrhythmic agents in terminating ventricular tachycardia and fibrillation. In recent years, it has been recognized that less proarrhythmic hERG blockers with clinical potential or Class III antiarrhythmic agents exhibit, in addition to their hERG-blocking activity, a second action that facilitates the voltage-dependent activation of the hERG channel. This facilitation is believed to reduce the proarrhythmic potential by supporting the final repolarizing of action potentials. This review covers the pharmacological characteristics of hERG blockers/facilitators, the molecular mechanisms underlying facilitation, and their clinical significance, as well as unresolved issues and requirements for research in the fields of ion channel pharmacology and drug-induced arrhythmias.
Collapse
Affiliation(s)
- Kazuharu Furutani
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Tokushima Bunri University, 180 Nishihama-Boji, Yamashiro-cho, Tokushima 770-8514, Japan
| |
Collapse
|
5
|
MacRae CA, Peterson RT. Zebrafish as a Mainstream Model for In Vivo Systems Pharmacology and Toxicology. Annu Rev Pharmacol Toxicol 2023; 63:43-64. [PMID: 36151053 DOI: 10.1146/annurev-pharmtox-051421-105617] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Pharmacology and toxicology are part of a much broader effort to understand the relationship between chemistry and biology. While biomedicine has necessarily focused on specific cases, typically of direct human relevance, there are real advantages in pursuing more systematic approaches to characterizing how health and disease are influenced by small molecules and other interventions. In this context, the zebrafish is now established as the representative screenable vertebrate and, through ongoing advances in the available scale of genome editing and automated phenotyping, is beginning to address systems-level solutions to some biomedical problems. The addition of broader efforts to integrate information content across preclinical model organisms and the incorporation of rigorous analytics, including closed-loop deep learning, will facilitate efforts to create systems pharmacology and toxicology with the ability to continuously optimize chemical biological interactions around societal needs. In this review, we outline progress toward this goal.
Collapse
Affiliation(s)
- Calum A MacRae
- Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA;
| | | |
Collapse
|
6
|
Zhou H, Zhang Z, Zhu L, Li P, Hong S, Liu L, Liu X. Prediction of drug pro-arrhythmic cardiotoxicity using a semi-physiologically based pharmacokinetic model linked to cardiac ionic currents inhibition. Toxicol Appl Pharmacol 2022; 457:116312. [PMID: 36343672 DOI: 10.1016/j.taap.2022.116312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 10/23/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Drug-induced torsades de pointes (TdP) risks are responsible for the withdrawal of many drugs from the market. Nowadays, assessments of drug-induced TdP risks are mainly based on maximum effective free therapeutic plasma concentration (EFTPCmax) and cardiac ionic current inhibitions using the human ventricular myocytes model (Tor-ORd model). Myocytes are targets of drug-induced TdP. The TdP risks may be directly linked to myocyte drug concentrations. We aimed to develop a semi-physiologically based pharmacokinetic (Semi-PBPK) model linked to cardiac ionic current inhibition (pharmacodynamics, PD) (Semi-PBPK-PD) to simultaneously predict myocyte drug concentrations and their TdP risks in humans. Alterations in action potential duration (ΔAPD90) were simulated using the Tor-ORd model and ionic current inhibition parameters based on myocyte or plasma drug concentrations. The predicted ΔAPD90 values were translated into in vivo alterations in QT interval(ΔQTc) induced by moxifloxacin, dofetilide, or sotalol. Myocyte drug concentrations of moxifloxacin, dofetilide, and sotalol gave better predictions of ΔQTc than plasma. Following validating the developed semi-PBPK-PD model, TdP risks of 37 drugs were assessed using ΔAPD90 and early afterdepolarization occurrence, which were estimated based on 10 × EFTPCmax and 10 × EFTMCmax (maximum effective free therapeutic myocyte concentration). 10 × EFTMCmax gave more sensitive and accurate predictions of pro-arrhythmic cardiotoxicity and the predicted TdP risks were also closer to clinic practice than 10 × EFTPCmax. In conclusion, pharmacokinetics and TdP risks of 37 drugs were successfully predicted using the semi-PBPK-PD model. Myocyte drug concentrations gave better predictions of ΔQTc and TdP risks than plasma.
Collapse
Affiliation(s)
- Han Zhou
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zexin Zhang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ping Li
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shijin Hong
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
Yang Y, Liu A, Tsai CT, Liu C, Wu JC, Cui B. Cardiotoxicity drug screening based on whole-panel intracellular recording. Biosens Bioelectron 2022; 216:114617. [PMID: 36027802 PMCID: PMC9930661 DOI: 10.1016/j.bios.2022.114617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 07/18/2022] [Accepted: 07/31/2022] [Indexed: 01/11/2023]
Abstract
Unintended binding of small-molecule drugs to ion channels affects electrophysiological properties of cardiomyocytes and potentially leads to arrhythmia and heart failure. The waveforms of intracellular action potentials reflect the coordinated activities of cardiac ion channels and serve as a reliable means for assessing drug toxicity, but the implementation is limited by the low throughput of patch clamp for intracellular recording measurements. In the last decade, several new technologies are being developed to address this challenge. We recently developed the nanocrown electrode array (NcEA) technology that allows robust, parallel, and long-duration recording of intracellular action potentials (iAPs). Here, we demonstrate that NcEAs allow comparison of iAP waveforms before and after drug treatment from the same cell. This self-referencing comparison not only shows distinct drug effects of sodium, potassium, and calcium blockers, but also reveals subtle differences among three subclasses of sodium channel blockers with sub-millisecond accuracy. Furthermore, self-referencing comparison unveils heterogeneous drug responses among different cells. In our study, whole-panel simultaneous intracellular recording can be reliably achieved with ∼94% success rate. The average duration of intracellular recording is ∼30 min and some last longer than 2 h. With its high reliability, long recording duration, and easy-to-use nature, NcEA would be useful for iAP-based preclinical drug screening.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Aofei Liu
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Ching-Ting Tsai
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA,Department of Medicine, Stanford University, Stanford, CA, USA,Department of Radiology, Stanford University, Stanford, CA, USA
| | - Bianxiao Cui
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA; Wu-Tsai Neuroscience Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Shi M, Dong Y, Bouwmeester H, Rietjens IMCM, Strikwold M. In vitro-in silico-based prediction of inter-individual and inter-ethnic variations in the dose-dependent cardiotoxicity of R- and S-methadone in humans. Arch Toxicol 2022; 96:2361-2380. [PMID: 35604418 PMCID: PMC9217890 DOI: 10.1007/s00204-022-03309-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 04/27/2022] [Indexed: 12/02/2022]
Abstract
New approach methodologies predicting human cardiotoxicity are of interest to support or even replace in vivo-based drug safety testing. The present study presents an in vitro–in silico approach to predict the effect of inter-individual and inter-ethnic kinetic variations in the cardiotoxicity of R- and S-methadone in the Caucasian and the Chinese population. In vitro cardiotoxicity data, and metabolic data obtained from two approaches, using either individual human liver microsomes or recombinant cytochrome P450 enzymes (rCYPs), were integrated with physiologically based kinetic (PBK) models and Monte Carlo simulations to predict inter-individual and inter-ethnic variations in methadone-induced cardiotoxicity. Chemical specific adjustment factors were defined and used to derive dose–response curves for the sensitive individuals. Our simulations indicated that Chinese are more sensitive towards methadone-induced cardiotoxicity with Margin of Safety values being generally two-fold lower than those for Caucasians for both methadone enantiomers. Individual PBK models using microsomes and PBK models using rCYPs combined with Monte Carlo simulations predicted similar inter-individual and inter-ethnic variations in methadone-induced cardiotoxicity. The present study illustrates how inter-individual and inter-ethnic variations in cardiotoxicity can be predicted by combining in vitro toxicity and metabolic data, PBK modelling and Monte Carlo simulations. The novel methodology can be used to enhance cardiac safety evaluations and risk assessment of chemicals.
Collapse
Affiliation(s)
- Miaoying Shi
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands. .,NHC Key Laboratory of Food Safety Risk Assessment, Chinese Academy of Medical Sciences Research Unit (No. 2019RU014), China National Center for Food Safety Risk Assessment, Beijing, 100021, China.
| | - Yumeng Dong
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Marije Strikwold
- Van Hall Larenstein University of Applied Sciences, 8901 BV, Leeuwarden, The Netherlands
| |
Collapse
|
9
|
Wang M, Ma Y, Shen Z, Jiang L, Zhang X, Wei X, Han Z, Liu H, Yang T. Mapping the Knowledge of Antipsychotics-Induced Sudden Cardiac Death: A Scientometric Analysis in CiteSpace and VOSviewer. Front Psychiatry 2022; 13:925583. [PMID: 35873271 PMCID: PMC9300900 DOI: 10.3389/fpsyt.2022.925583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/09/2022] [Indexed: 11/30/2022] Open
Abstract
The drugs on the market for schizophrenia are first-generation and second-generation antipsychotics. Some of the first-generation drugs have more side effects than the other drugs, so they are gradually no longer being applied clinically. Years of research have shown that the risk of sudden cardiac death in psychotic patients is associated with drug use, and antipsychotic drugs have certain cardiotoxicity and can induce arrhythmias. The mechanism of antipsychotic-induced sudden cardiac death is complicated. Highly cited papers are among the most commonly used indicators for measuring scientific excellence. This article presents a high-level analysis of highly cited papers using Web of Science core collection databases, scientometrics methods, and thematic clusters. Temporal dynamics of focus topics are identified using a collaborative network (author, institution, thematic clusters, and temporal dynamics of focus topics are identified), keyword co-occurrence analysis, co-citation clustering, and keyword evolution. The primary purpose of this study is to discuss the visual results, summarize the research progress, and predict the future research trends by bibliometric methods of CiteSpace and VOSviewer. This study showed that a research hotspot is that the mechanisms of cardiotoxicity, the safety monitoring, and the assessment of the risk-benefit during clinical use of some newer antipsychotics, clozapine and olanzapine. We discussed relevant key articles briefly and provided ideas for future research directions for more researchers to conduct related research.
Collapse
Affiliation(s)
- Min Wang
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Yixun Ma
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Zefang Shen
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Lufang Jiang
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Xiaoyuan Zhang
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Xuan Wei
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| | - Zhengqi Han
- Institute for Digital Technology and Law, China University of Political Science and Law, Beijing, China.,The CUPL Scientometrics and Evaluation Center of Rule of Law, China University of Political Science and Law, Beijing, China
| | - Hongxia Liu
- Institute for Digital Technology and Law, China University of Political Science and Law, Beijing, China.,The CUPL Scientometrics and Evaluation Center of Rule of Law, China University of Political Science and Law, Beijing, China
| | - Tiantong Yang
- Key Laboratory of Evidence Science, Institute of Evidence Law and Forensic Science, Ministry of Education, China University of Political Science and Law, Beijing, China.,Collaborative Innovation Center of Judicial Civilization, Beijing, China
| |
Collapse
|
10
|
Mokrov GV. Linked biaromatic compounds as cardioprotective agents. Arch Pharm (Weinheim) 2021; 355:e2100428. [PMID: 34967027 DOI: 10.1002/ardp.202100428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 11/08/2022]
Abstract
Cardiovascular diseases (CVDs) are widespread in the modern world, and their number is constantly growing. For a long time, CVDs have been the leading cause of morbidity and mortality worldwide. Drugs for the treatment of CVD have been developed almost since the beginning of the 20th century, and a large number of effective cardioprotective agents of various classes have been created. Nevertheless, the need for the design and development of new safe drugs for the treatment of CVD remains. Literature data indicate that a huge number of cardioprotective agents of various generations and mechanisms correspond to a single generalized pharmacophore model containing two aromatic nuclei linked by a linear linker. In this regard, we put forward a concept for the design of a new generation of cardioprotective agents with a multitarget mechanism of action within the indicated pharmacophore model. This review is devoted to a generalization of the currently known compounds with cardioprotective properties and corresponding to the pharmacophore model of biaromatic compounds linked by a linear linker. Particular attention is paid to the history of the creation of these drugs, approaches to their design, and analysis of the structure-action relationship within each class.
Collapse
Affiliation(s)
- Grigory V Mokrov
- Department of Medicinal Chemistry, FSBI "Zakusov Institute of Pharmacology", Moscow, Russia
| |
Collapse
|
11
|
Pesti K, Földi MC, Zboray K, Toth AV, Lukacs P, Mike A. Characterization of Compound-Specific, Concentration-Independent Biophysical Properties of Sodium Channel Inhibitor Mechanism of Action Using Automated Patch-Clamp Electrophysiology. Front Pharmacol 2021; 12:738460. [PMID: 34497526 PMCID: PMC8419314 DOI: 10.3389/fphar.2021.738460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 08/10/2021] [Indexed: 01/15/2023] Open
Abstract
We have developed an automated patch-clamp protocol that allows high information content screening of sodium channel inhibitor compounds. We have observed that individual compounds had their specific signature patterns of inhibition, which were manifested irrespective of the concentration. Our aim in this study was to quantify these properties. Primary biophysical data, such as onset rate, the shift of the half inactivation voltage, or the delay of recovery from inactivation, are concentration-dependent. We wanted to derive compound-specific properties, therefore, we had to neutralize the effect of concentration. This study describes how this is done, and shows how compound-specific properties reflect the mechanism of action, including binding dynamics, cooperativity, and interaction with the membrane phase. We illustrate the method using four well-known sodium channel inhibitor compounds, riluzole, lidocaine, benzocaine, and bupivacaine. Compound-specific biophysical properties may also serve as a basis for deriving parameters for kinetic modeling of drug action. We discuss how knowledge about the mechanism of action may help to predict the frequency-dependence of individual compounds, as well as their potential persistent current component selectivity. The analysis method described in this study, together with the experimental protocol described in the accompanying paper, allows screening for inhibitor compounds with specific kinetic properties, or with specific mechanisms of inhibition.
Collapse
Affiliation(s)
- Krisztina Pesti
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- School of Ph.D. Studies, Semmelweis University, Budapest, Hungary
| | - Mátyás C. Földi
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Katalin Zboray
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Adam V. Toth
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Peter Lukacs
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| | - Arpad Mike
- Department of Biochemistry, ELTE Eötvös Loránd University, Budapest, Hungary
- Plant Protection Institute, Centre for Agricultural Research, Martonvásár, Hungary
| |
Collapse
|
12
|
Ellermann C, Wolfes J, Eckardt L, Frommeyer G. Role of the rabbit whole-heart model for electrophysiologic safety pharmacology of non-cardiovascular drugs. Europace 2021; 23:828-836. [PMID: 33200170 DOI: 10.1093/europace/euaa288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/27/2020] [Indexed: 11/13/2022] Open
Abstract
Plenty of non-cardiovascular drugs alter cardiac electrophysiology and may ultimately lead to life-threatening arrhythmias. In clinical practice, measuring the QT interval as a marker for the repolarization period is the most common tool to assess the electrophysiologic safety of drugs. However, the sole measurement of the QT interval may be insufficient to determine the proarrhythmic risk of non-cardiovascular agents. Several other markers are considered in pre-clinical safety testing to determine potential harm on cardiac electrophysiology. Besides measuring typical electrophysiologic parameters such as repolarization duration, whole-heart models allow the determination of potential predictors for proarrhythmia. Spatial and temporal heterogeneity as well as changes of shape of the action potential can be easily assessed. In addition, provocation manoeuvers (either by electrolyte imbalances or programmed pacing protocols) may induce sustained arrhythmias and thereby determine ventricular vulnerability to arrhythmias. Compared with the human heart, the rabbit heart possesses a similar distribution of ion currents that govern cardiac repolarization, resulting in a rectangular action potential configuration in both species. In addition, similar biophysical properties of rabbit and human cardiac ion channels lead to a comparable pharmacologic response in human and rabbit hearts. Of note, arrhythmia patterns resemble in both species due to the similar effective size of human and rabbit hearts. Thus, the rabbit heart is particularly suitable for testing the electrophysiologic safety of drugs. Several experimental setups have been developed for studying cardiac electrophysiology in rabbits, ranging from single cell to tissue preparations, whole-heart setups, and in vivo models.
Collapse
Affiliation(s)
- Christian Ellermann
- Department of Cardiology II (Electrophysiology), University Hospital Münster, Albert-Schweitzer-Campus 1, Münster D-48149, Germany
| | - Julian Wolfes
- Department of Cardiology II (Electrophysiology), University Hospital Münster, Albert-Schweitzer-Campus 1, Münster D-48149, Germany
| | - Lars Eckardt
- Department of Cardiology II (Electrophysiology), University Hospital Münster, Albert-Schweitzer-Campus 1, Münster D-48149, Germany
| | - Gerrit Frommeyer
- Department of Cardiology II (Electrophysiology), University Hospital Münster, Albert-Schweitzer-Campus 1, Münster D-48149, Germany
| |
Collapse
|
13
|
Midei MG, Darpo B, Ayers G, Brown R, Couderc JP, Daly W, Ferber G, Sager PT, Camm AJ. Electrophysiological and ECG Effects of Perhexiline, a Mixed Cardiac Ion Channel Inhibitor, Evaluated in Nonclinical Assays and in Healthy Subjects. J Clin Pharmacol 2021; 61:1606-1617. [PMID: 34214210 DOI: 10.1002/jcph.1934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/28/2021] [Indexed: 11/10/2022]
Abstract
Perhexiline has been used to treat hypertrophic cardiomyopathy. In addition to its effect on carnitine-palmitoyltransferase-1, it has mixed ion channel effects through inhibition of several cardiac ion currents. Effects on cardiac ion channels expressed in mammalian cells were assayed using a manual patch-clamp technique, action potential duration (APD) was measured in ventricular trabeculae of human donor hearts, and electrocardiogram effects were evaluated in healthy subjects in a thorough QT (TQT) study. Perhexiline blocked several cardiac ion currents at concentrations within the therapeutic range (150-600 ng/mL) with IC50 for hCav1.2 ∼ hERG < late hNav1.5. A significant APD shortening was observed in perhexiline-treated cardiomyocytes. The TQT study was conducted with a pilot part in 9 subjects to evaluate a dosing schedule that would achieve therapeutic and supratherapeutic perhexiline plasma concentrations on days 4 and 6, respectively. Guided by the results from the pilot, 104 subjects were enrolled in a parallel-designed part with a nested crossover comparison for the positive control. Perhexiline caused QTc prolongation, with the largest effect on ΔΔQTcF, 14.7 milliseconds at therapeutic concentrations and 25.6 milliseconds at supratherapeutic concentrations and a positive and statistically significant slope of the concentration-ΔΔQTcF relationship (0.018 milliseconds per ng/mL; 90%CI, 0.0119-0.0237 milliseconds per ng/mL). In contrast, the JTpeak interval was shortened with a negative concentration-JTpeak relationship, a pattern consistent with multichannel block. Further studies are needed to evaluate whether this results in a low proarrhythmic risk.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Georg Ferber
- Statistik Georg Ferber GmbH, Riehen, Switzerland
| | - Philip T Sager
- Department of Medicine, Cardiovascular Research Institute, Stanford University, Palo Alto, California, USA
| | - A John Camm
- Division of Clinical Sciences, Cardiovascular and Cell Sciences Research Institute, St George's University of London, London, UK
| |
Collapse
|
14
|
Kowalska M, Nowaczyk J, Nowaczyk A. K V11.1, Na V1.5, and Ca V1.2 Transporter Proteins as Antitarget for Drug Cardiotoxicity. Int J Mol Sci 2020; 21:E8099. [PMID: 33143033 PMCID: PMC7663169 DOI: 10.3390/ijms21218099] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Safety assessment of pharmaceuticals is a rapidly developing area of pharmacy and medicine. The new advanced guidelines for testing the toxicity of compounds require specialized tools that provide information on the tested drug in a quick and reliable way. Ion channels represent the third-largest target. As mentioned in the literature, ion channels are an indispensable part of the heart's work. In this paper the most important information concerning the guidelines for cardiotoxicity testing and the way the tests are conducted has been collected. Attention has been focused on the role of selected ion channels in this process.
Collapse
Affiliation(s)
- Magdalena Kowalska
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland;
| | - Jacek Nowaczyk
- Faculty of Chemistry, Nicolaus Copernicus University, 87-100 Toruń, Poland;
| | - Alicja Nowaczyk
- Department of Organic Chemistry, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, 85-094 Bydgoszcz, Poland;
| |
Collapse
|
15
|
Bystricky W, Maier C, Gintant G, Bergau D, Carter D. Identification of Drug-Induced Multichannel Block and Proarrhythmic Risk in Humans Using Continuous T Vector Velocity Effect Profiles Derived From Surface Electrocardiograms. Front Physiol 2020; 11:567383. [PMID: 33071822 PMCID: PMC7530300 DOI: 10.3389/fphys.2020.567383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/27/2020] [Indexed: 01/07/2023] Open
Abstract
We present continuous T vector velocity (TVV) effect profiles as a new method for identifying drug effects on cardiac ventricular repolarization. TVV measures the temporal change in the myocardial action potential distribution during repolarization. The T vector dynamics were measured as the time required to reach p percent of the total T vector trajectory length, denoted as Tr(p), with p in {1, …, 100%}. The Tr(p) values were individually corrected for heart rate at each trajectory length percentage p. Drug effects were measured by evaluating the placebo corrected changes from baseline of Tr(p)c jointly for all p using functional mixed effects models. The p-dependent model parameters were implemented as cubic splines, providing continuous drug effect profiles along the entire ventricular repolarization process. The effect profile distributions were approximated by bootstrap simulations. We applied this TVV-based analysis approach to ECGs available from three published studies that were conducted in the CiPA context. These studies assessed the effect of 10 drugs and drug combinations with different ion channel blocking properties on myocardial repolarization in a total of 104 healthy volunteers. TVV analysis revealed that blockade of outward potassium currents alone presents an effect profile signature of continuous accumulation of delay throughout the entire repolarization interval. In contrast, block of inward sodium or calcium currents involves acceleration, which accumulates during early repolarization. The balance of blocking inward versus outward currents was reflected in the percentage pzero of the T vector trajectory length where accelerated repolarization transitioned to delayed repolarization. Binary classification using a threshold pzero = 43% separated predominant hERG channel blocking drugs with potentially higher proarrhythmic risk (moxifloxacin, dofetilide, quinidine, chloroquine) from multichannel blocking drugs with low proarrhythmic risk (ranolazine, verapamil, lopinavir/ritonavir) with sensitivity 0.99 and specificity 0.97. The TVV-based effect profile provides a detailed view of drug effects throughout the entire ventricular repolarization interval. It enables the evaluation of drug-induced blocks of multiple cardiac repolarization currents from clinical ECGs. The proposed pzero parameter enhances identification of the proarrhythmic risk of a drug beyond QT prolongation, and therefore constitutes an important tool for cardiac arrhythmia risk assessment.
Collapse
Affiliation(s)
- Werner Bystricky
- Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, IL, United States
| | - Christoph Maier
- Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, IL, United States
- Department of Medical Informatics, Heilbronn University, Heilbronn, Germany
| | - Gary Gintant
- Integrated Sciences and Technology, AbbVie, Inc., North Chicago, IL, United States
| | - Dennis Bergau
- Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, IL, United States
| | - David Carter
- Clinical Pharmacology and Pharmacometrics, AbbVie, Inc., North Chicago, IL, United States
| |
Collapse
|
16
|
Song L, Zhang ZF, Hu LK, Zhang PH, Cao ZZ, Liu ZP, Zhang PP, Ma JH. Curcumin, a Multi-Ion Channel Blocker That Preferentially Blocks Late Na + Current and Prevents I/R-Induced Arrhythmias. Front Physiol 2020; 11:978. [PMID: 32973546 PMCID: PMC7472421 DOI: 10.3389/fphys.2020.00978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Increasing evidence shows that Curcumin (Cur) has a protective effect against cardiovascular diseases. However, the role of Cur in the electrophysiology of cardiomyocytes is currently not entirely understood. Therefore, the present study was conducted to investigate the effects of Cur on the action potential and transmembrane ion currents in rabbit ventricular myocytes to explore its antiarrhythmic property. The whole-cell patch clamp was used to record the action potential and ion currents, while the multichannel acquisition and analysis system was used to synchronously record the electrocardiogram and monophasic action potential. The results showed that 30 μmol/L Cur shortened the 50 and 90% repolarization of action potential by 17 and 7%, respectively. In addition, Cur concentration dependently inhibited the Late-sodium current (I Na.L), Transient-sodium current (I Na.T), L-type calcium current (I Ca.L), and Rapidly delayed rectifying potassium current (I Kr), with IC50 values of 7.53, 398.88, 16.66, and 9.96 μmol/L, respectively. Importantly, the inhibitory effect of Cur on I Na.L was 52.97-fold higher than that of I Na.T. Moreover, Cur decreased ATX II-prolonged APD, suppressed the ATX II-induced early afterdepolarization (EAD) and Ca2+-induced delayed afterdepolarization (DAD) in ventricular myocytes, and reduced the occurrence and average duration of ventricular tachycardias and ventricular fibrillations induced by ischemia-reperfusion injury. In conclusion, Cur inhibited I Na.L, I Na.T, I Ca.L, and I Kr; shortened APD; significantly suppressed EAD and DAD-like arrhythmogenic activities at the cellular level; and exhibited antiarrhythmic effect at the organ level. It is first revealed that Cur is a multi-ion channel blocker that preferentially blocks I Na.L and may have potential antiarrhythmic property.
Collapse
Affiliation(s)
- Lv Song
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Ze-Fu Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Liang-Kun Hu
- Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Pei-Hua Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhen-Zhen Cao
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Zhi-Pei Liu
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Pei-Pei Zhang
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | - Ji-Hua Ma
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China.,College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College of Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Shi M, Bouwmeester H, Rietjens IMCM, Strikwold M. Integrating in vitro data and physiologically based kinetic modeling-facilitated reverse dosimetry to predict human cardiotoxicity of methadone. Arch Toxicol 2020; 94:2809-2827. [PMID: 32367273 PMCID: PMC7395048 DOI: 10.1007/s00204-020-02766-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 04/22/2020] [Indexed: 12/23/2022]
Abstract
Development of novel testing strategies to detect adverse human health effects is of interest to replace in vivo-based drug and chemical safety testing. The aim of the present study was to investigate whether physiologically based kinetic (PBK) modeling-facilitated conversion of in vitro toxicity data is an adequate approach to predict in vivo cardiotoxicity in humans. To enable evaluation of predictions made, methadone was selected as the model compound, being a compound for which data on both kinetics and cardiotoxicity in humans are available. A PBK model for methadone in humans was developed and evaluated against available kinetic data presenting an adequate match. Use of the developed PBK model to convert concentration–response curves for the effect of methadone on human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) in the so-called multi electrode array (MEA) assay resulted in predictions for in vivo dose–response curves for methadone-induced cardiotoxicity that matched the available in vivo data. The results also revealed differences in protein plasma binding of methadone to be a potential factor underlying variation between individuals with respect to sensitivity towards the cardiotoxic effects of methadone. The present study provides a proof-of-principle of using PBK modeling-based reverse dosimetry of in vitro data for the prediction of cardiotoxicity in humans, providing a novel testing strategy in cardiac safety studies.
Collapse
Affiliation(s)
- Miaoying Shi
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Marije Strikwold
- Van Hall Larenstein University of Applied Sciences, 8901 BV, Leeuwarden, The Netherlands
| |
Collapse
|
18
|
Orvos P, Kohajda Z, Szlovák J, Gazdag P, Árpádffy-Lovas T, Tóth D, Geramipour A, Tálosi L, Jost N, Varró A, Virág L. Evaluation of Possible Proarrhythmic Potency: Comparison of the Effect of Dofetilide, Cisapride, Sotalol, Terfenadine, and Verapamil on hERG and Native IKr Currents and on Cardiac Action Potential. Toxicol Sci 2020; 168:365-380. [PMID: 30561737 DOI: 10.1093/toxsci/kfy299] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The proarrhythmic potency of drugs is usually attributed to the IKr current block. During safety pharmacology testing analysis of IKr in cardiomyocytes was replaced by human ether-a-go-go-related gene (hERG) test using automated patch-clamp systems in stable transfected cell lines. Aim of this study was to compare the effect of proarrhythmic compounds on hERG and IKr currents and on cardiac action potential. The hERG current was measured by using both automated and manual patch-clamp methods on HEK293 cells. The native ion currents (IKr, INaL, ICaL) were recorded from rabbit ventricular myocytes by manual patch-clamp technique. Action potentials in rabbit ventricular muscle and undiseased human donor hearts were studied by conventional microelectrode technique. Dofetilide, cisapride, sotalol, terfenadine, and verapamil blocked hERG channels at 37°C with an IC50 of 7 nM, 18 nM, 343 μM, 165 nM, and 214 nM, respectively. Using manual patch-clamp, the IC50 values of sotalol and terfenadine were 78 µM and 31 nM, respectively. The IC50 values calculated from IKr measurements at 37°C were 13 nM, 26 nM, 52 μM, 54 nM, and 268 nM, respectively. Cisapride, dofetilide, and sotalol excessively lengthened, terfenadine, and verapamil did not influence the action potential duration. Terfenadine significantly inhibited INaL and moderately ICaL, verapamil blocked only ICaL. Automated hERG assays may over/underestimate proarrhythmic risk. Manual patch-clamp has substantially higher sensitivity to certain drugs. Action potential studies are also required to analyze complex multichannel effects. Therefore, manual patch-clamp and action potential experiments should be a part of preclinical safety tests.
Collapse
Affiliation(s)
- Péter Orvos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,Department of Ophthalmology, University of Szeged, Szeged H-6720, Hungary
| | - Zsófia Kohajda
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary
| | - Jozefina Szlovák
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | - Péter Gazdag
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | | | - Dániel Tóth
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | - Amir Geramipour
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine
| | | | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| | - László Virág
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine.,MTA-SZTE Research Group for Cardiovascular Pharmacology, Hungarian Academy of Sciences, Szeged H-6720, Hungary.,Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, Szeged H-6720, Hungary
| |
Collapse
|
19
|
Zhou X, Qu Y, Passini E, Bueno-Orovio A, Liu Y, Vargas HM, Rodriguez B. Blinded In Silico Drug Trial Reveals the Minimum Set of Ion Channels for Torsades de Pointes Risk Assessment. Front Pharmacol 2020; 10:1643. [PMID: 32082155 PMCID: PMC7003137 DOI: 10.3389/fphar.2019.01643] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Torsades de Pointes (TdP) is a type of ventricular arrhythmia which could be observed as an unwanted drug-induced cardiac side effect, and it is associated with repolarization abnormalities in single cells. The pharmacological evaluations of TdP risk in previous years mainly focused on the hERG channel due to its vital role in the repolarization of cardiomyocytes. However, only considering drug effects on hERG led to false positive predictions since the drug action on other ion channels can also have crucial regulatory effects on repolarization. To address the limitation of only evaluating hERG, the Comprehensive in Vitro Proarrhythmia Assay initiative has proposed to systematically integrate drug effects on multiple ion channels into in silico drug trial to improve TdP risk assessment. It is not clear how many ion channels are sufficient for reliable TdP risk predictions, and whether differences in IC50 and Hill coefficient values from independent sources can lead to divergent in silico prediction outcomes. The rationale of this work is to investigate the above two questions using a computationally efficient population of human ventricular cells optimized to favor repolarization abnormality. Our blinded results based on two independent data sources confirm that simulations with the optimized population of human ventricular cell models enable efficient in silico drug screening, and also provide direct observation and mechanistic analysis of repolarization abnormality. Our results show that 1) the minimum set of ion channels required for reliable TdP risk predictions are Nav1.5 (peak), Cav1.2, and hERG; 2) for drugs with multiple ion channel blockage effects, moderate IC50 variations combined with variable Hill coefficients can affect the accuracy of in silico predictions.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Yusheng Qu
- SPARC, Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Elisa Passini
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Alfonso Bueno-Orovio
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| | - Yang Liu
- GAU, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Hugo M Vargas
- SPARC, Amgen Research, Amgen Inc., Thousand Oaks, CA, United States
| | - Blanca Rodriguez
- Department of Computer Science, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
20
|
Vicente J, Strauss DG, Upreti VV, Fossler MJ, Sager PT, Noveck R. The Potential Role of the J‐T
peak
Interval in Proarrhythmic Cardiac Safety: Current State of the Science From the American College of Clinical Pharmacology and the Cardiac Safety Research Consortium. J Clin Pharmacol 2019; 59:909-914. [DOI: 10.1002/jcph.1411] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Jose Vicente
- Division of Cardiovascular and Renal Products, Office of Drug Evaluation I, Office of New Drugs, Center for Drug Evaluation and ResearchUS Food and Drug Administration Silver Spring MD USA
| | - David G. Strauss
- Division of Applied Regulatory ScienceUS Food and Drug Administration Silver Spring MD USA
| | - Vijay V. Upreti
- Clinical Pharmacology Oncology Therapeutic Area HeadClinical Pharmacology Modeling and SimulationAmgen South San Francisco CA USA
| | - Michael J. Fossler
- Clinical Operations & Quantitative SciencesTrevena, Inc. Chesterbrook PA USA
| | | | - Robert Noveck
- Duke Early Phase 1 Clinical Research Unit Durham NC USA
| |
Collapse
|
21
|
Vicente J, Zusterzeel R, Johannesen L, Ochoa-Jimenez R, Mason JW, Sanabria C, Kemp S, Sager PT, Patel V, Matta MK, Liu J, Florian J, Garnett C, Stockbridge N, Strauss DG. Assessment of Multi-Ion Channel Block in a Phase I Randomized Study Design: Results of the CiPA Phase I ECG Biomarker Validation Study. Clin Pharmacol Ther 2019; 105:943-953. [PMID: 30447156 PMCID: PMC6654598 DOI: 10.1002/cpt.1303] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/20/2018] [Indexed: 11/24/2022]
Abstract
Balanced multi‐ion channel‐blocking drugs have low torsade risk because they block inward currents. The Comprehensive In Vitro Proarrhythmia Assay (CiPA) initiative proposes to use an in silico cardiomyocyte model to determine the presence of balanced block, and absence of heart rate corrected J‐Tpeak (J‐Tpeakc) prolongation would be expected for balanced blockers. This study included three balanced blockers in a 10‐subject‐per‐drug parallel design; lopinavir/ritonavir and verapamil met the primary end point of ΔΔJ‐Tpeakc upper bound < 10 ms, whereas ranolazine did not (upper bounds of 8.8, 6.1, and 12.0 ms, respectively). Chloroquine, a predominant blocker of the potassium channel encoded by the ether‐à‐go‐go related gene (hERG), prolonged ΔΔQTc and ΔΔJ‐Tpeakc by ≥ 10 ms. In a separate crossover design, diltiazem (calcium block) did not shorten dofetilide‐induced ΔQTc prolongation, but shortened ΔJ‐Tpeakc and prolonged ΔTpeak‐Tend. Absence of J‐Tpeakc prolongation seems consistent with balanced block; however, small sample size (10 subjects) may be insufficient to characterize concentration‐response in some cases.
Collapse
Affiliation(s)
- Jose Vicente
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Robbert Zusterzeel
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Lars Johannesen
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Roberto Ochoa-Jimenez
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jay W Mason
- Department of Medicine, Division of Cardiology, University of Utah, Salt Lake City, Utah, USA.,Spaulding Clinical Research, West Bend, Wisconsin, USA
| | | | - Sarah Kemp
- Spaulding Clinical Research, West Bend, Wisconsin, USA
| | | | - Vikram Patel
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Murali K Matta
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jiang Liu
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Jeffry Florian
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Christine Garnett
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Norman Stockbridge
- Office of New Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - David G Strauss
- Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
22
|
Abstract
More than four decades passed since sigma receptors were first mentioned. Since then, existence of at least two receptor subtypes and their tissue distributions have been proposed. Nowadays, it is clear, that sigma receptors are unique ubiquitous proteins with pluripotent function, which can interact with so many different classes of proteins. As the endoplasmic resident proteins, they work as molecular chaperones - accompany various proteins during their folding, ensure trafficking of the maturated proteins between cellular organelles and regulate their functions. In the heart, sigma receptor type 1 is more dominant. Cardiac sigma 1 receptors regulate response to endoplasmic reticulum stress, modulates calcium signaling in cardiomyocyte and can affect function of voltage-gated ion channels. They contributed in pathophysiology of cardiac hypertrophy, heart failure and many other cardiovascular disorders. Therefore, sigma receptors are potential novel targets for specific treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- T Stracina
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | |
Collapse
|
23
|
Windley MJ, Lee W, Vandenberg JI, Hill AP. The Temperature Dependence of Kinetics Associated with Drug Block of hERG Channels Is Compound-Specific and an Important Factor for Proarrhythmic Risk Prediction. Mol Pharmacol 2018; 94:760-769. [PMID: 29728448 DOI: 10.1124/mol.117.111534] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/02/2018] [Indexed: 02/14/2025] Open
Abstract
Current mandated preclinical tests for drug-induced proarrhythmia are very sensitive, but not sufficiently specific. This has led to concern that there is a high attrition rate of potentially safe drugs that could have been beneficial to patients. The comprehensive in vitro proarrhythmia initiative has proposed new metrics based around in silico risk predictions, which are informed, among other things, by measures of human ether-à-go-go-related gene channel (hERG) block kinetics. However, high-throughput patch-clamp systems set to collect these data largely operate at ambient temperature, whereas the simulations for risk prediction are carried out at physiologic temperature. The aims of this study were to: 1) determine to what degree kinetics of drug block of hERG are temperature-dependent, 2) assess the impact of any temperature dependence of drug binding kinetics on repolarization in silico, and 3) identify whether a common set of Q10 scalars can be used to extrapolate kinetic data gathered at ambient to physiologic temperatures for use in in silico proarrhythmic risk prediction. We show that, for a range of drugs, kinetics of block are temperature-dependent and, furthermore, that the degree of temperature dependence is different for each drug. As a result, no common set of Q10 scalars could describe the observed range of temperature dependencies. These results suggest that if accurate physiologic temperature models of the kinetics of drug binding are important for in silico risk prediction, the in vitro data should be acquired at physiologic temperature.
Collapse
Affiliation(s)
- Monique J Windley
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - William Lee
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - Jamie I Vandenberg
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| | - Adam P Hill
- Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia and St Vincent's Clinical School, University of NSW, Darlinghurst, New South Wales, Australia
| |
Collapse
|
24
|
Patel N, Hatley O, Berg A, Romero K, Wisniowska B, Hanna D, Hermann D, Polak S. Towards Bridging Translational Gap in Cardiotoxicity Prediction: an Application of Progressive Cardiac Risk Assessment Strategy in TdP Risk Assessment of Moxifloxacin. AAPS JOURNAL 2018. [DOI: 10.1208/s12248-018-0199-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
25
|
Park E, Gintant GA, Bi D, Kozeli D, Pettit SD, Pierson JB, Skinner M, Willard J, Wisialowski T, Koerner J, Valentin JP. Can non-clinical repolarization assays predict the results of clinical thorough QT studies? Results from a research consortium. Br J Pharmacol 2018; 175:606-617. [PMID: 29181850 PMCID: PMC5786459 DOI: 10.1111/bph.14101] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/12/2017] [Accepted: 11/13/2017] [Indexed: 01/06/2023] Open
Abstract
Background and Purpose Translation of non‐clinical markers of delayed ventricular repolarization to clinical prolongation of the QT interval corrected for heart rate (QTc) (a biomarker for torsades de pointes proarrhythmia) remains an issue in drug discovery and regulatory evaluations. We retrospectively analysed 150 drug applications in a US Food and Drug Administration database to determine the utility of established non‐clinical in vitro IKr current human ether‐à‐go‐go‐related gene (hERG), action potential duration (APD) and in vivo (QTc) repolarization assays to detect and predict clinical QTc prolongation. Experimental Approach The predictive performance of three non‐clinical assays was compared with clinical thorough QT study outcomes based on free clinical plasma drug concentrations using sensitivity and specificity, receiver operating characteristic (ROC) curves, positive (PPVs) and negative predictive values (NPVs) and likelihood ratios (LRs). Key Results Non‐clinical assays demonstrated robust specificity (high true negative rate) but poor sensitivity (low true positive rate) for clinical QTc prolongation at low‐intermediate (1×–30×) clinical exposure multiples. The QTc assay provided the most robust PPVs and NPVs (ability to predict clinical QTc prolongation). ROC curves (overall test accuracy) and LRs (ability to influence post‐test probabilities) demonstrated overall marginal performance for hERG and QTc assays (best at 30× exposures), while the APD assay demonstrated minimal value. Conclusions and Implications The predictive value of hERG, APD and QTc assays varies, with drug concentrations strongly affecting translational performance. While useful in guiding preclinical candidates without clinical QT prolongation, hERG and QTc repolarization assays provide greater value compared with the APD assay.
Collapse
Affiliation(s)
- Eunjung Park
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | - Gary A Gintant
- Department of Integrative Pharmacology, AbbVie, North Chicago, IL, USA
| | - Daoqin Bi
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | - Devi Kozeli
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | | | | - Matthew Skinner
- Drug Safety and Metabolism, AstraZeneca, Macclesfield, Cheshire, UK
| | - James Willard
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | | - John Koerner
- Center for Drug Evaluation and Research, US FDA, Silver Spring, MD, USA
| | | |
Collapse
|
26
|
Sharifi M, Buzatu D, Harris S, Wilkes J. Development of models for predicting Torsade de Pointes cardiac arrhythmias using perceptron neural networks. BMC Bioinformatics 2017; 18:497. [PMID: 29297274 PMCID: PMC5751783 DOI: 10.1186/s12859-017-1895-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Blockage of some ion channels and in particular, the hERG (human Ether-a’-go-go-Related Gene) cardiac potassium channel delays cardiac repolarization and can induce arrhythmia. In some cases it leads to a potentially life-threatening arrhythmia known as Torsade de Pointes (TdP). Therefore recognizing drugs with TdP risk is essential. Candidate drugs that are determined not to cause cardiac ion channel blockage are more likely to pass successfully through clinical phases II and III trials (and preclinical work) and not be withdrawn even later from the marketplace due to cardiotoxic effects. The objective of the present study is to develop an SAR (Structure-Activity Relationship) model that can be used as an early screen for torsadogenic (causing TdP arrhythmias) potential in drug candidates. The method is performed using descriptors comprised of atomic NMR chemical shifts (13C and 15N NMR) and corresponding interatomic distances which are combined into a 3D abstract space matrix. The method is called 3D-SDAR (3-dimensional spectral data-activity relationship) and can be interrogated to identify molecular features responsible for the activity, which can in turn yield simplified hERG toxicophores. A dataset of 55 hERG potassium channel inhibitors collected from Kramer et al. consisting of 32 drugs with TdP risk and 23 with no TdP risk was used for training the 3D-SDAR model. Results An artificial neural network (ANN) with multilayer perceptron was used to define collinearities among the independent 3D-SDAR features. A composite model from 200 random iterations with 25% of the molecules in each case yielded the following figures of merit: training, 99.2%; internal test sets, 66.7%; external (blind validation) test set, 68.4%. In the external test set, 70.3% of positive TdP drugs were correctly predicted. Moreover, toxicophores were generated from TdP drugs. Conclusion A 3D-SDAR was successfully used to build a predictive model for drug-induced torsadogenic and non-torsadogenic drugs based on 55 compounds. The model was tested in 38 external drugs. Electronic supplementary material The online version of this article (10.1186/s12859-017-1895-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mohsen Sharifi
- Division of Systems Biology, FDA's National Center for Toxicological Research, Jefferson, AR, 72079, USA
| | - Dan Buzatu
- Division of Systems Biology, FDA's National Center for Toxicological Research, Jefferson, AR, 72079, USA.
| | - Stephen Harris
- Division of Systems Biology, FDA's National Center for Toxicological Research, Jefferson, AR, 72079, USA
| | - Jon Wilkes
- Division of Systems Biology, FDA's National Center for Toxicological Research, Jefferson, AR, 72079, USA
| |
Collapse
|
27
|
Takeda M, Miyagawa S, Fukushima S, Saito A, Ito E, Harada A, Matsuura R, Iseoka H, Sougawa N, Mochizuki-Oda N, Matsusaki M, Akashi M, Sawa Y. Development of In Vitro Drug-Induced Cardiotoxicity Assay by Using Three-Dimensional Cardiac Tissues Derived from Human Induced Pluripotent Stem Cells. Tissue Eng Part C Methods 2017; 24:56-67. [PMID: 28967302 PMCID: PMC5757089 DOI: 10.1089/ten.tec.2017.0247] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
An in vitro drug-induced cardiotoxicity assay is a critical step in drug discovery for clinical use. The use of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) is promising for this purpose. However, single hiPSC-CMs are limited in their ability to mimic native cardiac tissue structurally and functionally, and the generation of artificial cardiac tissue using hiPSC-CMs is an ongoing challenging. We therefore developed a new method of constructing three-dimensional (3D) artificial tissues in a short time by coating extracellular matrix (ECM) components on cell surfaces. We hypothesized that 3D cardiac tissues derived from hiPSC-CMs (3D-hiPSC-CT) could be used for an in vitro drug-induced cardiotoxicity assay. 3D-hiPSC-CT were generated by fibronectin and gelatin nanofilm coated single hiPSC-CMs. Histologically, 3D-hiPSC-CT exhibited a sarcomere structure in the myocytes and ECM proteins, such as fibronectin, collagen type I/III, and laminin. The administration of cytotoxic doxorubicin at 5.0 μM induced the release of lactate dehydrogenase, while that at 2.0 μM reduced the cell viability. E-4031, human ether-a-go-go related gene (hERG)-type potassium channel blocker, and isoproterenol induced significant changes both in the Ca transient parameters and contractile parameters in a dose-dependent manner. The 3D-hiPSC-CT exhibited doxorubicin-sensitive cytotoxicity and hERG channel blocker/isoproterenol-sensitive electrical activity in vitro, indicating its usefulness for drug-induced cardiotoxicity assays or drug screening systems for drug discovery.
Collapse
Affiliation(s)
- Maki Takeda
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Shigeru Miyagawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Satsuki Fukushima
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Atsuhiro Saito
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Emiko Ito
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Akima Harada
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Ryohei Matsuura
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Hiroko Iseoka
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Nagako Sougawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Noriko Mochizuki-Oda
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| | - Michiya Matsusaki
- 2 Department of Applied Chemistry, Osaka University Graduate School of Engineering , Osaka, Japan
| | - Mitsuru Akashi
- 3 Building Block Science Joint Research Chair, Graduate School of Frontier Biosciences, Osaka University , Suita, Japan
| | - Yoshiki Sawa
- 1 Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine , Suita, Osaka, Japan
| |
Collapse
|
28
|
Hazell L, Raschi E, De Ponti F, Thomas SHL, Salvo F, Ahlberg Helgee E, Boyer S, Sturkenboom M, Shakir S. Evidence for the hERG Liability of Antihistamines, Antipsychotics, and Anti-Infective Agents: A Systematic Literature Review From the ARITMO Project. J Clin Pharmacol 2017; 57:558-572. [PMID: 28019033 DOI: 10.1002/jcph.838] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 10/08/2016] [Indexed: 04/21/2025]
Abstract
A systematic review was performed to categorize the hERG (human ether-a-go-go-related gene) liability of antihistamines, antipsychotics, and anti-infectives and to compare it with current clinical risk of torsade de pointes (TdP). Eligible studies were hERG assays reporting half-minimal inhibitory concentrations (IC50). A "hERG safety margin" was calculated from the IC50 divided by the peak human plasma concentration (free Cmax ). A margin below 30 defined hERG liability. Each drug was assigned an "uncertainty score" based on volume, consistency, precision, and internal and external validity of evidence. The hERG liability was compared to existing knowledge on TdP risk (www.credibledrugs.org). Of 1828 studies, 82 were eligible, allowing calculation of safety margins for 61 drugs. Thirty-one drugs (51%) had evidence of hERG liability including 6 with no previous mention of TdP risk (eg, desloratadine, lopinavir). Conversely, 16 drugs (26%) had no evidence of hERG liability including 6 with known, or at least conditional or possible, TdP risk (eg, chlorpromazine, sulpiride). The main sources of uncertainty were the validity of the experimental conditions used (antihistamines and antipsychotics) and nonuse of reference compounds (anti-infectives). In summary, hERG liability was categorized for 3 widely used drug classes, incorporating a qualitative assessment of the strength of available evidence. Some concordance with TdP risk was observed, although several drugs had hERG liability without evidence of clinical risk and vice versa. This may be due to gaps in clinical evidence, limitations of hERG/Cmax data, or other patient/drug-specific factors that contribute to real-life TdP risk.
Collapse
Affiliation(s)
- Lorna Hazell
- Drug Safety Research Unit, Southampton, United Kingdom
| | - Emanuel Raschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabrizio De Ponti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simon H L Thomas
- Institute of Cellular Medicine, Faculty of Medicine, Newcastle University, Newcastle, United Kingdom
| | - Francesco Salvo
- University of Bordeaux U657, CHU de Bordeaux, Bordeaux, France
| | - Ernst Ahlberg Helgee
- Drug Safety and Metabolism, AstraZeneca Innovative Medicines and Early Development, Mölndal, Sweden
| | - Scott Boyer
- Computational Toxicology, Swedish Toxicology Sciences Research Center, Södertälje, Sweden
| | | | - Saad Shakir
- Drug Safety Research Unit, Southampton, United Kingdom
| |
Collapse
|
29
|
Maturation of human pluripotent stem cell derived cardiomyocytes is improved in cardiovascular construct. Cytotechnology 2017; 69:785-800. [PMID: 28397099 PMCID: PMC5595750 DOI: 10.1007/s10616-017-0088-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 03/17/2017] [Indexed: 11/21/2022] Open
Abstract
In order to translate preclinical data into the clinical studies, relevant in vitro models with structure and key functional properties similar to native human tissue should be used. In vitro cardiac models with vascular structures mimic the highly vascularized myocardium and provide interactions between endothelial cells, stromal cells and cardiomyocytes. Currently, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been shown to present immature morphology and fetal-like electrophysiological properties that may limit their use as physiological test platform. The aim of this study was to develop multicellular in vitro cardiovascular construct modeling human heart tissue. In the cardiovascular construct, hPSC-CMs were cultured with a vascular-like network formed by human foreskin fibroblasts and human umbilical vein endothelial cells that served as a platform in the construct. Cardiomyocyte orientation, maturation, electrophysiological properties and drug responses of the cardiovascular construct were characterized and compared to CM monoculture. hPSC-CMs in cardiovascular construct showed elongated morphology and aligned with the vascular-like network. Electrophysiological properties and calcium metabolism of hPSC-CMs as well as response to E-4031 and adrenaline demonstrated normal physiological behavior. Increased expression of cardiac structural proteins and ion channels in cardiovascular construct compared to CM monoculture were detected. In conclusion, vascular-like network supports the structural and functional maturation of hPSC-CMs. Our results suggest that cardiovascular construct presents more mature in vitro cardiac model compared to CM monoculture and could therefore serve as an advanced test system for cardiac safety and efficacy assessment as well as a model system for biomedical research.
Collapse
|
30
|
Sube R, Ertel EA. Cardiomyocytes Derived from Human Induced Pluripotent Stem Cells: An In-Vitro Model to Predict Cardiac Effects of Drugs. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jbise.2017.1011040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
31
|
Gintant GA, Su Z, Martin RL, Cox BF. Utility of hERG Assays as Surrogate Markers of Delayed Cardiac Repolarization and QT Safety. Toxicol Pathol 2016; 34:81-90. [PMID: 16507548 DOI: 10.1080/01926230500431376] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
HERG (human-ether-a-go-go-related gene) encodes for a cardiac potassium channel that plays a critical role in defining ventricular repolarization. Noncardiovascular drugs associated with a rare but potentially lethal ventricular arrhythmia (Torsades de Pointes) have been linked to delayed cardiac repolarization and block of hERG current. This brief overview will discuss the role of hERG current in cardiac electrophysiology, its involvement in drug-induced delayed repolarization, and approaches used to define drug effects on hERG current. In addition, examples of hERG blocking drugs acting differently (i.e., overt and covert hERG blockade due to multichannel block) together with the utility and limitations of hERG assays as tools to predict the risk of delayed repolarization and proarrhythmia are discussed.
Collapse
Affiliation(s)
- Gary A Gintant
- Deptartment of Integrative Pharmacology, Abbott Laboratories, Abbott Park, Illinois 60064-6119, USA.
| | | | | | | |
Collapse
|
32
|
Onal B, Hund TJ. Integrative approaches for prediction of cardiotoxic drug effects and mitigation strategies. J Mol Cell Cardiol 2016; 102:1-2. [PMID: 27894864 DOI: 10.1016/j.yjmcc.2016.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/10/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Birce Onal
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center and The Ohio State University College of Engineering, USA; Department of Biomedical Engineering, The Ohio State University Wexner Medical Center and The Ohio State University College of Engineering, USA
| | - Thomas J Hund
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center and The Ohio State University College of Engineering, USA; Department of Biomedical Engineering, The Ohio State University Wexner Medical Center and The Ohio State University College of Engineering, USA; Department of Internal Medicine, The Ohio State University Wexner Medical Center and The Ohio State University College of Engineering, USA.
| |
Collapse
|
33
|
Abstract
The prediction of molecular targets is highly beneficial during the drug discovery process, be it for off-target elucidation or deconvolution of phenotypic screens. Here, we present OCEAN, a target prediction tool exclusively utilizing publically available ChEMBL data. OCEAN uses a heuristics approach based on a validation set containing almost 1000 drug ← → target relationships. New ChEMBL data (ChEMBL20 as well as ChEMBL21) released after the validation was used for a prospective OCEAN performance check. The success rates of OCEAN to predict correctly the targets within the TOP10 ranks are 77% for recently marketed drugs and 62% for all new ChEMBL20 compounds and 51% for all new ChEMBL21 compounds. OCEAN is also capable of identifying polypharmacological compounds; the success rate for molecules simultaneously hitting at least two targets is 64% to be correctly predicted within the TOP10 ranks. The source code of OCEAN can be found at http://www.github.com/rdkit/OCEAN.
Collapse
Affiliation(s)
- Paul Czodrowski
- Discovery Technologies, Merck Serono Research, Merck KGaA , Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Wolf-Guido Bolick
- Discovery Technologies, Merck Serono Research, Merck KGaA , Frankfurter Straße 250, 64293 Darmstadt, Germany
| |
Collapse
|
34
|
Wiśniowska B, Polak S. Virtual Clinical Trial Toward Polytherapy Safety Assessment: Combination of Physiologically Based Pharmacokinetic/Pharmacodynamic-Based Modeling and Simulation Approach With Drug-Drug Interactions Involving Terfenadine as an Example. J Pharm Sci 2016; 105:3415-3424. [PMID: 27640752 DOI: 10.1016/j.xphs.2016.08.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 10/21/2022]
Abstract
A Quantitative Systems Pharmacology approach was utilized to predict the cardiac consequences of drug-drug interaction (DDI) at the population level. The Simcyp in vitro-in vivo correlation and physiologically based pharmacokinetic platform was used to predict the pharmacokinetic profile of terfenadine following co-administration of the drug. Electrophysiological effects were simulated using the Cardiac Safety Simulator. The modulation of ion channel activity was dependent on the inhibitory potential of drugs on the main cardiac ion channels and a simulated free heart tissue concentration. ten Tusscher's human ventricular cardiomyocyte model was used to simulate the pseudo-ECG traces and further predict the pharmacodynamic consequences of DDI. Consistent with clinical observations, predicted plasma concentration profiles of terfenadine show considerable intra-subject variability with recorded Cmax values below 5 ng/mL for most virtual subjects. The pharmacokinetic and pharmacodynamic effects of inhibitors were predicted with reasonable accuracy. In all cases, a combination of the physiologically based pharmacokinetic and physiology-based pharmacodynamic models was able to differentiate between the terfenadine alone and terfenadine + inhibitor scenario. The range of QT prolongation was comparable in the clinical and virtual studies. The results indicate that mechanistic in vitro-in vivo correlation can be applied to predict the clinical effects of DDI even without comprehensive knowledge on all mechanisms contributing to the interaction.
Collapse
Affiliation(s)
- Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, Kraków 30-688, Poland.
| | - Sebastian Polak
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, Kraków 30-688, Poland; Simcyp (part of Certara), Blades Enterprise Centre, John Street, Sheffield S2 4SU, UK
| |
Collapse
|
35
|
Qiu XS, Chauveau S, Anyukhovsky EP, Rahim T, Jiang YP, Harleton E, Feinmark SJ, Lin RZ, Coronel R, Janse MJ, Opthof T, Rosen TS, Cohen IS, Rosen MR. Increased Late Sodium Current Contributes to the Electrophysiological Effects of Chronic, but Not Acute, Dofetilide Administration. Circ Arrhythm Electrophysiol 2016; 9:e003655. [PMID: 27071826 DOI: 10.1161/circep.115.003655] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 03/01/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Drugs are screened for delayed rectifier potassium current (IKr) blockade to predict long QT syndrome prolongation and arrhythmogenesis. However, single-cell studies have shown that chronic (hours) exposure to some IKr blockers (eg, dofetilide) prolongs repolarization additionally by increasing late sodium current (INa-L) via inhibition of phosphoinositide 3-kinase. We hypothesized that chronic dofetilide administration to intact dogs prolongs repolarization by blocking IKr and increasing INa-L. METHODS AND RESULTS We continuously infused dofetilide (6-9 μg/kg bolus+6-9 μg/kg per hour IV infusion) into anesthetized dogs for 7 hours, maintaining plasma levels within the therapeutic range. In separate experiments, myocardial biopsies were taken before and during 6-hour intravenous dofetide infusion, and the level of phospho-Akt was determined. Acute and chronic dofetilide effects on action potential duration (APD) were studied in canine left ventricular subendocardial slabs using microelectrode techniques. Dofetilide monotonically increased QTc and APD throughout 6.5-hour exposure. Dofetilide infusion during ≥210 minutes inhibited Akt phosphorylation. INa-L block with lidocaine shortened QTc and APD more at 6.5 hours than at 50 minutes (QTc) or 30 minutes (APD) dofetilide administration. In comparison, moxifloxacin, an IKr blocker with no effects on phosphoinositide 3-kinase and INa-L prolonged APD acutely but no additional prolongation occurred on chronic superfusion. Lidocaine shortened APD equally during acute and chronic moxifloxacin superfusion. CONCLUSIONS Increased INa-L contributes to chronic dofetilide effects in vivo. These data emphasize the need to include time and INa-L in evaluating the phosphoinositide 3-kinase inhibition-derived proarrhythmic potential of drugs and provide a mechanism for benefit from lidocaine administration in clinical acquired long QT syndrome.
Collapse
Affiliation(s)
- Xiaoliang S Qiu
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Samuel Chauveau
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Evgeny P Anyukhovsky
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Tania Rahim
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Ya-Ping Jiang
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Erin Harleton
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Steven J Feinmark
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Richard Z Lin
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Ruben Coronel
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Michiel J Janse
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Tobias Opthof
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Tove S Rosen
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| | - Ira S Cohen
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.).
| | - Michael R Rosen
- From the Department of Physiology and Biophysics, Stony Brook University, NY (X.S.Q., S.C., E.P.A., T.R., Y.-P.J., R.Z.L., I.S.C.); Departments of Pharmacology (E.H., S.J.F., M.R.R.) and Pediatrics (T.S.R., M.R.R.), College of Physician and Surgeons of Columbia University, New York, NY; Medical Service, Northport VA Medical Center, NY (R.Z.L.); Department of Clinical and Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands (R.C., M.J.J., T.O.); L'Institut de RYthmologie et de modélisation Cardiaque (LIRYC), Université Bordeaux Segalen, Bordeaux, France (R.C.); and Department of Medical Physiology, University Medical Center Utrecht, The Netherlands (T.O.)
| |
Collapse
|
36
|
|
37
|
Johannesen L, Vicente J, Mason JW, Erato C, Sanabria C, Waite-Labott K, Hong M, Lin J, Guo P, Mutlib A, Wang J, Crumb WJ, Blinova K, Chan D, Stohlman J, Florian J, Ugander M, Stockbridge N, Strauss DG. Late sodium current block for drug-induced long QT syndrome: Results from a prospective clinical trial. Clin Pharmacol Ther 2016; 99:214-23. [PMID: 26259627 PMCID: PMC5421403 DOI: 10.1002/cpt.205] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 08/05/2015] [Indexed: 12/19/2022]
Abstract
Drug-induced long QT syndrome has resulted in many drugs being withdrawn from the market. At the same time, the current regulatory paradigm for screening new drugs causing long QT syndrome is preventing drugs from reaching the market, sometimes inappropriately. In this study, we report the results of a first-of-a-kind clinical trial studying late sodium (mexiletine and lidocaine) and calcium (diltiazem) current blocking drugs to counteract the effects of hERG potassium channel blocking drugs (dofetilide and moxifloxacin). We demonstrate that both mexiletine and lidocaine substantially reduce heart-rate corrected QT (QTc) prolongation from dofetilide by 20 ms. Furthermore, all QTc shortening occurs in the heart-rate corrected J-Tpeak (J-Tpeak c) interval, the biomarker we identified as a sign of late sodium current block. This clinical trial demonstrates that late sodium blocking drugs can substantially reduce QTc prolongation from hERG potassium channel block and assessment of J-Tpeak c may add value beyond only assessing QTc.
Collapse
Affiliation(s)
- L Johannesen
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| | - J Vicente
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
- BSICoS Group, Aragón Institute for Engineering Research (I3A), IIS
Aragón, University of Zaragoza, Zaragoza, Spain
| | - JW Mason
- Spaulding Clinical, West Bend, Wisconsin, USA
- University of Utah, Salt Lake City, Utah, USA
| | - C Erato
- Spaulding Clinical, West Bend, Wisconsin, USA
| | - C Sanabria
- Spaulding Clinical, West Bend, Wisconsin, USA
| | | | - M Hong
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - J Lin
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - P Guo
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - A Mutlib
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - J Wang
- Frontage Laboratories, Exton, Pennsylvania, USA
| | - WJ Crumb
- Zenas Technologies, Metairie, Louisiana, USA
| | - K Blinova
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - D Chan
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - J Stohlman
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - J Florian
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - M Ugander
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| | - N Stockbridge
- Center for Drug Evaluation and Research, US Food and Drug Administration,
Silver Spring, Maryland, USA
| | - DG Strauss
- Center for Devices and Radiological Health, US Food and Drug Administration,
Silver Spring, Maryland, USA
- Department of Clinical Physiology, Karolinska Institutet and Karolinska
University Hospital, Stockholm, Sweden
| |
Collapse
|
38
|
Eder A, Vollert I, Hansen A, Eschenhagen T. Human engineered heart tissue as a model system for drug testing. Adv Drug Deliv Rev 2016; 96:214-24. [PMID: 26026976 DOI: 10.1016/j.addr.2015.05.010] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/11/2015] [Accepted: 05/21/2015] [Indexed: 12/29/2022]
Abstract
Drug development is time- and cost-intensive and, despite extensive efforts, still hampered by the limited value of current preclinical test systems to predict side effects, including proarrhythmic and cardiotoxic effects in clinical practice. Part of the problem may be related to species-dependent differences in cardiomyocyte biology. Therefore, the event of readily available human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CM) has raised hopes that this human test bed could improve preclinical safety pharmacology as well as drug discovery approaches. However, hiPSC-CM are immature and exhibit peculiarities in terms of ion channel function, gene expression, structural organization and functional responses to drugs that limit their present usefulness. Current efforts are thus directed towards improving hiPSC-CM maturity and high-content readouts. Culturing hiPSC-CM as 3-dimensional engineered heart tissue (EHT) improves CM maturity and anisotropy and, in a 24-well format using silicone racks, enables automated, multiplexed high content readout of contractile function. This review summarizes the principal technology and focuses on advantages and disadvantages of this technology and its potential for preclinical drug screening.
Collapse
|
39
|
Abstract
Attrition due to nonclinical safety represents a major issue for the productivity of pharmaceutical research and development (R&D) organizations, especially during the compound optimization stages of drug discovery and the early stages of clinical development. Focusing on decreasing nonclinical safety-related attrition is not a new concept, and various approaches have been experimented with over the last two decades. Front-loading testing funnels in Discovery with in vitro toxicity assays designed to rapidly identify unfavorable molecules was the approach adopted by most pharmaceutical R&D organizations a few years ago. However, this approach has also a non-negligible opportunity cost. Hence, significant refinements to the "fail early, fail often" paradigm have been proposed recently to reflect the complexity of accurately categorizing compounds with early data points without taking into account other important contextual aspects, in particular efficacious systemic and tissue exposures. This review provides an overview of toxicology approaches and models that can be used in pharmaceutical Discovery at the series/lead identification and lead optimization stages to guide and inform chemistry efforts, as well as a personal view on how to best use them to meet nonclinical safety-related attrition objectives consistent with a sustainable pharmaceutical R&D model. The scope of this review is limited to small molecules, as large molecules are associated with challenges that are quite different. Finally, a perspective on how several emerging technologies may impact toxicity evaluation is also provided.
Collapse
Affiliation(s)
- Eric A G Blomme
- Global Preclinical Safety, AbbVie Inc. , 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Yvonne Will
- Drug Safety Research and Development, Pfizer , Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
40
|
Yuan Y, Bai X, Luo C, Wang K, Zhang H. The virtual heart as a platform for screening drug cardiotoxicity. Br J Pharmacol 2015; 172:5531-5547. [PMID: 25363597 PMCID: PMC4667856 DOI: 10.1111/bph.12996] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 10/23/2014] [Accepted: 10/28/2014] [Indexed: 01/01/2023] Open
Abstract
To predict the safety of a drug at an early stage in its development is a major challenge as there is a lack of in vitro heart models that correlate data from preclinical toxicity screening assays with clinical results. A biophysically detailed computer model of the heart, the virtual heart, provides a powerful tool for simulating drug-ion channel interactions and cardiac functions during normal and disease conditions and, therefore, provides a powerful platform for drug cardiotoxicity screening. In this article, we first review recent progress in the development of theory on drug-ion channel interactions and mathematical modelling. Then we propose a family of biomarkers that can quantitatively characterize the actions of a drug on the electrical activity of the heart at multi-physical scales including cellular and tissue levels. We also conducted some simulations to demonstrate the application of the virtual heart to assess the pro-arrhythmic effects of cisapride and amiodarone. Using the model we investigated the mechanisms responsible for the differences between the two drugs on pro-arrhythmogenesis, even though both prolong the QT interval of ECGs. Several challenges for further development of a virtual heart as a platform for screening drug cardiotoxicity are discussed.
Collapse
Affiliation(s)
- Yongfeng Yuan
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Xiangyun Bai
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Cunjin Luo
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Kuanquan Wang
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
| | - Henggui Zhang
- School of Computer Science and TechnologyHarbin Institute of TechnologyHarbinChina
- Biological Physics GroupSchool of Physics and AstronomyThe University of ManchesterManchesterUK
| |
Collapse
|
41
|
Salvo F, Pariente A, Shakir S, Robinson P, Arnaud M, Thomas SHL, Raschi E, Fourrier-Réglat A, Moore N, Sturkenboom M, Hazell on behalf of Investigators o L. Sudden cardiac and sudden unexpected death related to antipsychotics: A meta-analysis of observational studies. Clin Pharmacol Ther 2015; 99:306-14. [DOI: 10.1002/cpt.250] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/09/2015] [Indexed: 01/11/2023]
Affiliation(s)
- F Salvo
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
| | - A Pariente
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
- CIC Bordeaux CIC1401; Bordeaux France
| | - S Shakir
- Drug Safety Research Unit; Southampton Hampshire UK
| | - P Robinson
- CIC Bordeaux CIC1401; Bordeaux France
- ADERA; Pessac France
| | - M Arnaud
- University of Bordeaux, INSERM U657; Bordeaux France
| | - SHL Thomas
- Medical Toxicology Centre, Institute of Cellular Medicine; Newcastle University; Newcastle UK
| | - E Raschi
- Department of Medical and Surgical Sciences; University of Bologna; Bologna Italy
| | - A Fourrier-Réglat
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
| | - N Moore
- University of Bordeaux, INSERM U657; Bordeaux France
- CHU Bordeaux; Bordeaux France
- CIC Bordeaux CIC1401; Bordeaux France
| | - M Sturkenboom
- Department of Medical Informatics; Erasmus University Medical Centre; Rotterdam Netherlands
| | | | | |
Collapse
|
42
|
Asakura K, Hayashi S, Ojima A, Taniguchi T, Miyamoto N, Nakamori C, Nagasawa C, Kitamura T, Osada T, Honda Y, Kasai C, Ando H, Kanda Y, Sekino Y, Sawada K. Improvement of acquisition and analysis methods in multi-electrode array experiments with iPS cell-derived cardiomyocytes. J Pharmacol Toxicol Methods 2015; 75:17-26. [DOI: 10.1016/j.vascn.2015.04.002] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/30/2015] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
|
43
|
Computational investigations of hERG channel blockers: New insights and current predictive models. Adv Drug Deliv Rev 2015; 86:72-82. [PMID: 25770776 DOI: 10.1016/j.addr.2015.03.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/13/2015] [Accepted: 03/04/2015] [Indexed: 01/08/2023]
Abstract
Identification of potential human Ether-a-go-go Related-Gene (hERG) potassium channel blockers is an essential part of the drug development and drug safety process in pharmaceutical industries or academic drug discovery centers, as they may lead to drug-induced QT prolongation, arrhythmia and Torsade de Pointes. Recent reports also suggest starting to address such issues at the hit selection stage. In order to prioritize molecules during the early drug discovery phase and to reduce the risk of drug attrition due to cardiotoxicity during pre-clinical and clinical stages, computational approaches have been developed to predict the potential hERG blockage of new drug candidates. In this review, we will describe the current in silico methods developed and applied to predict and to understand the mechanism of actions of hERG blockers, including ligand-based and structure-based approaches. We then discuss ongoing research on other ion channels and hERG polymorphism susceptible to be involved in LQTS and how systemic approaches can help in the drug safety decision.
Collapse
|
44
|
Gintant G. In Vitro Early Safety Pharmacology Screening: Perspectives Related to Cardiovascular Safety. Handb Exp Pharmacol 2015; 229:47-64. [PMID: 26091635 DOI: 10.1007/978-3-662-46943-9_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vitro screening for cardiovascular safety liabilities of novel drug candidates presents a challenge for the pharmaceutical industry. Such approaches rely on detecting pharmacologic effects on key components of complex integrated system early in drug discovery to define potential safety liabilities. Key to such studies are the concepts of hazard identification vs. risk assessment, drug specificity vs. selectivity, and an appreciation of the challenges faced when attempting to translate in vitro findings to preclinical in vivo as well as clinical effects. This chapter defines some key aspects of early safety pharmacology screening for cardiovascular liabilities, citing studies of two key depolarizing cardiac currents (fast sodium current and L-type calcium current) as examples linked to effects on cardiac conduction and repolarization.
Collapse
|
45
|
Polak S, Pugsley MK, Stockbridge N, Garnett C, Wiśniowska B. Early Drug Discovery Prediction of Proarrhythmia Potential and Its Covariates. AAPS JOURNAL 2015; 17:1025-32. [PMID: 25940083 PMCID: PMC4476985 DOI: 10.1208/s12248-015-9773-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/16/2015] [Indexed: 12/26/2022]
|
46
|
Trenor B, Gomis-Tena J, Cardona K, Romero L, Rajamani S, Belardinelli L, Giles WR, Saiz J. In silico assessment of drug safety in human heart applied to late sodium current blockers. Channels (Austin) 2015; 7:249-62. [PMID: 23696033 DOI: 10.4161/chan.24905] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drug-induced action potential (AP) prolongation leading to Torsade de Pointes is a major concern for the development of anti-arrhythmic drugs. Nevertheless the development of improved anti-arrhythmic agents, some of which may block different channels, remains an important opportunity. Partial block of the late sodium current (I(NaL)) has emerged as a novel anti-arrhythmic mechanism. It can be effective in the settings of free radical challenge or hypoxia. In addition, this approach can attenuate pro-arrhythmic effects of blocking the rapid delayed rectifying K(+) current (I(Kr)). The main goal of our computational work was to develop an in-silico tool for preclinical anti-arrhythmic drug safety assessment, by illustrating the impact of I(Kr)/I(NaL) ratio of steady-state block of drug candidates on "torsadogenic" biomarkers. The O'Hara et al. AP model for human ventricular myocytes was used. Biomarkers for arrhythmic risk, i.e., AP duration, triangulation, reverse rate-dependence, transmural dispersion of repolarization and electrocardiogram QT intervals, were calculated using single myocyte and one-dimensional strand simulations. Predetermined amounts of block of I(NaL) and I(Kr) were evaluated. "Safety plots" were developed to illustrate the value of the specific biomarker for selected combinations of IC(50)s for I(Kr) and I(NaL) of potential drugs. The reference biomarkers at baseline changed depending on the "drug" specificity for these two ion channel targets. Ranolazine and GS967 (a novel potent inhibitor of I(NaL)) yielded a biomarker data set that is considered safe by standard regulatory criteria. This novel in-silico approach is useful for evaluating pro-arrhythmic potential of drugs and drug candidates in the human ventricle.
Collapse
|
47
|
Gunaruwan P, Howes LG. Assessing the Arrhythmogenic Potential of New Drugs: A Guide for the Pharmaceutical Physician. Pharmaceut Med 2015. [DOI: 10.1007/s40290-015-0082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
48
|
Wiśniowska B, Mendyk A, Fijorek K, Polak S. Computer-based prediction of the drug proarrhythmic effect: problems, issues, known and suspected challenges. Europace 2015; 16:724-35. [PMID: 24798962 DOI: 10.1093/europace/euu009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
It is likely that computer modelling and simulations will become an element of comprehensive cardiac safety testing. Their role would be primarily the integration and the interpretation of previously gathered data. There are still unanswered questions and issues which we list and describe below. They include sources of data used for the development of the models as well as data utilized as input information, which can come from the in vitro studies and the quantitative structure-activity relationship models. The pharmacokinetics of the drugs in question play a crucial role as their active concentration should be considered, yet the question remains where is the right place to assess it. The pharmacodynamic angle includes complications coming from multiple drugs (i.e. active metabolites) acting in parallel as well as the type of interaction with (potentially) multiple affected channels. Once established, the model and the methodology of its use should be further validated, optimistically against individual data reported at the clinical level as the physiological, anatomical, and genetic parameters play a crucial role in the drug-triggered arrhythmia induction. All the abovementioned issues should be at least considered and-hopefully-resolved, to properly utilize the mathematical models for a cardiac safety assessment.
Collapse
Affiliation(s)
- Barbara Wiśniowska
- Unit of Pharmacoepidemiology and Pharmacoeconomics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9 Street, 30-688 Kraków, Poland
| | | | | | | |
Collapse
|
49
|
On-chip in vitro cell-network pre-clinical cardiac toxicity using spatiotemporal human cardiomyocyte measurement on a chip. Sci Rep 2014; 4:4670. [PMID: 24751527 PMCID: PMC5381194 DOI: 10.1038/srep04670] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 03/24/2014] [Indexed: 11/09/2022] Open
Abstract
To overcome the limitations and misjudgments of conventional prediction of arrhythmic cardiotoxicity, we have developed an on-chip in vitro predictive cardiotoxicity assay using cardiomyocytes derived from human stem cells employing a constructive spatiotemporal two step measurement of fluctuation (short-term variability; STV) of cell's repolarization and cell-to-cell conduction time, representing two origins of lethal arrhythmia. Temporal STV of field potential duration (FPD) showed a potential to predict the risks of lethal arrhythmia originated from repolarization dispersion for false negative compounds, which was not correctly predicted by conventional measurements using animal cells, even for non-QT prolonging clinical positive compounds. Spatial STV of conduction time delay also unveiled the proarrhythmic risk of asynchronous propagation in cell networks, whose risk cannot be correctly predicted by single-cell-based measurements, indicating the importance of the spatiotemporal fluctuation viewpoint of in vitro cell networks for precise prediction of lethal arrhythmia reaching clinical assessment such as thorough QT assay.
Collapse
|
50
|
Nilsson MF, Webster WS. Effects of Macrolide Antibiotics on Rat Embryonic Heart Function In Vitro. ACTA ACUST UNITED AC 2014; 101:189-98. [DOI: 10.1002/bdrb.21107] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 02/15/2014] [Accepted: 02/16/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Mats F. Nilsson
- Drug Safety and Toxicology; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
| | - William S. Webster
- Drug Safety and Toxicology; Department of Pharmaceutical Biosciences; Uppsala University; Uppsala Sweden
- Department of Anatomy and Histology; School of Medical Sciences; University of Sydney; Sydney Australia
| |
Collapse
|