1
|
Daley MC, Moreau M, Bronk P, Fisher J, Kofron CM, Mende U, McMullen P, Choi BR, Coulombe K. In vitro to in vivo extrapolation from 3D hiPSC-derived cardiac microtissues and physiologically based pharmacokinetic modeling to inform next-generation arrhythmia risk assessment. Toxicol Sci 2024; 201:145-157. [PMID: 38897660 PMCID: PMC11347779 DOI: 10.1093/toxsci/kfae079] [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] [Indexed: 06/21/2024] Open
Abstract
Proarrhythmic cardiotoxicity remains a substantial barrier to drug development as well as a major global health challenge. In vitro human pluripotent stem cell-based new approach methodologies have been increasingly proposed and employed as alternatives to existing in vitro and in vivo models that do not accurately recapitulate human cardiac electrophysiology or cardiotoxicity risk. In this study, we expanded the capacity of our previously established 3D human cardiac microtissue model to perform quantitative risk assessment by combining it with a physiologically based pharmacokinetic model, allowing a direct comparison of potentially harmful concentrations predicted in vitro to in vivo therapeutic levels. This approach enabled the measurement of concentration responses and margins of exposure for 2 physiologically relevant metrics of proarrhythmic risk (i.e. action potential duration and triangulation assessed by optical mapping) across concentrations spanning 3 orders of magnitude. The combination of both metrics enabled accurate proarrhythmic risk assessment of 4 compounds with a range of known proarrhythmic risk profiles (i.e. quinidine, cisapride, ranolazine, and verapamil) and demonstrated close agreement with their known clinical effects. Action potential triangulation was found to be a more sensitive metric for predicting proarrhythmic risk associated with the primary mechanism of concern for pharmaceutical-induced fatal ventricular arrhythmias, delayed cardiac repolarization due to inhibition of the rapid delayed rectifier potassium channel, or hERG channel. This study advances human-induced pluripotent stem cell-based 3D cardiac tissue models as new approach methodologies that enable in vitro proarrhythmic risk assessment with high precision of quantitative metrics for understanding clinically relevant cardiotoxicity.
Collapse
Affiliation(s)
- Mark C Daley
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| | | | - Peter Bronk
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | | | - Celinda M Kofron
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| | - Ulrike Mende
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | | | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Kareen Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, United States
| |
Collapse
|
2
|
Satsuka A, Ribeiro AJS, Kawagishi H, Yanagida S, Hirata N, Yoshinaga T, Kurokawa J, Sugiyama A, Strauss DG, Kanda Y. Contractility assessment using aligned human iPSC-derived cardiomyocytes. J Pharmacol Toxicol Methods 2024; 128:107530. [PMID: 38917571 DOI: 10.1016/j.vascn.2024.107530] [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: 02/08/2024] [Revised: 05/17/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Cardiac safety assessment, such as lethal arrhythmias and contractility dysfunction, is critical during drug development. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been shown to be useful in predicting drug-induced proarrhythmic risk through international validation studies. Although cardiac contractility is another key function, fit-for-purpose hiPSC-CMs in evaluating drug-induced contractile dysfunction remain poorly understood. In this study, we investigated whether alignment of hiPSC-CMs on nanopatterned culture plates can assess drug-induced contractile changes more efficiently than non-aligned monolayer culture. METHODS Aligned hiPSC-CMs were obtained by culturing on 96-well culture plates with a ridge-groove-ridge nanopattern on the bottom surface, while non-aligned hiPSC-CMs were cultured on regular 96-well plates. Next-generation sequencing and qPCR experiments were performed for gene expression analysis. Contractility of the hiPSC-CMs was assessed using an imaging-based motion analysis system. RESULTS When cultured on nanopatterned plates, hiPSC-CMs exhibited an aligned morphology and enhanced expression of genes encoding proteins that regulate contractility, including myosin heavy chain, calcium channel, and ryanodine receptor. Compared to cultures on regular plates, the aligned hiPSC-CMs also showed both enhanced contraction and relaxation velocity. In addition, the aligned hiPSC-CMs showed a more physiological response to positive and negative inotropic agents, such as isoproterenol and verapamil. DISCUSSION Taken together, the aligned hiPSC-CMs exhibited enhanced structural and functional properties, leading to an improved capacity for contractility assessment compared to the non-aligned cells. These findings suggest that the aligned hiPSC-CMs can be used to evaluate drug-induced cardiac contractile changes.
Collapse
Affiliation(s)
- Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Silver Spring, MD 20903, USA
| | - Hiroyuki Kawagishi
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Naoya Hirata
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takashi Yoshinaga
- Advanced Biosignal Safety Assessment, Eisai Co., Ltd, 5-1-3 Tokodai, Tsukuba, Ibaraki 300-2635, Japan
| | - Junko Kurokawa
- Department of Bio-Informational Pharmacology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka-shi, Shizuoka 422-8526, Japan
| | - Atsushi Sugiyama
- Department of Pharmacology, Faculty of Medicine, Toho University, 5-21-16 Omori-nishi, Ota-ku, Tokyo 143-8540, Japan
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan.
| |
Collapse
|
3
|
Lickiss B, Hunker J, Bhagwan J, Linder P, Thomas U, Lotay H, Broadbent S, Dragicevic E, Stoelzle-Feix S, Turner J, Gossmann M. Chamber-specific contractile responses of atrial and ventricular hiPSC-cardiomyocytes to GPCR and ion channel targeting compounds: A microphysiological system for cardiac drug development. J Pharmacol Toxicol Methods 2024; 128:107529. [PMID: 38857637 DOI: 10.1016/j.vascn.2024.107529] [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: 02/26/2024] [Revised: 05/15/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) have found utility for conducting in vitro drug screening and disease modelling to gain crucial insights into pharmacology or disease phenotype. However, diseases such as atrial fibrillation, affecting >33 M people worldwide, demonstrate the need for cardiac subtype-specific cells. Here, we sought to investigate the base characteristics and pharmacological differences between commercially available chamber-specific atrial or ventricular hiPSC-CMs seeded onto ultra-thin, flexible PDMS membranes to simultaneously measure contractility in a 96 multi-well format. We investigated the effects of GPCR agonists (acetylcholine and carbachol), a Ca2+ channel agonist (S-Bay K8644), an HCN channel antagonist (ivabradine) and K+ channel antagonists (4-AP and vernakalant). We observed differential effects between atrial and ventricular hiPSC-CMs on contractile properties including beat rate, beat duration, contractile force and evidence of arrhythmias at a range of concentrations. As an excerpt of the compound analysis, S-Bay K8644 treatment showed an induced concentration-dependent transient increase in beat duration of atrial hiPSC-CMs, whereas ventricular cells showed a physiological increase in beat rate over time. Carbachol treatment produced marked effects on atrial cells, such as increased beat duration alongside a decrease in beat rate over time, but only minimal effects on ventricular cardiomyocytes. In the context of this chamber-specific pharmacology, we not only add to contractile characterization of hiPSC-CMs but propose a multi-well platform for medium-throughput early compound screening. Overall, these insights illustrate the key pharmacological differences between chamber-specific cardiomyocytes and their application on a multi-well contractility platform to gain insights for in vitro cardiac liability studies and disease modelling.
Collapse
Affiliation(s)
| | - Jan Hunker
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Jamie Bhagwan
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Peter Linder
- innoVitro GmbH, Artilleriestr 2, 52428 Jülich, Germany
| | - Ulrich Thomas
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | - Hardeep Lotay
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Steven Broadbent
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Elena Dragicevic
- Nanion Technologies GmbH, Ganghoferstr 70A, 80339 Munich, Germany
| | | | - Jan Turner
- Axol Bioscience Ltd, Babraham Research Campus, Cambridge CB22 3AT, UK
| | | |
Collapse
|
4
|
Wu W, Sun J, Zhang J, Zhao H, Qiu S, Li C, Shi C, Xu Y. Phosphoproteomics reveals a novel mechanism underlying the proarrhythmic effects of nilotinib, vandetanib, and mobocertinib. Toxicology 2024; 505:153830. [PMID: 38754619 DOI: 10.1016/j.tox.2024.153830] [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: 03/11/2024] [Revised: 04/30/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
The use of tyrosine kinase inhibitors (TKIs) has resulted in significant occurrence of arrhythmias. However, the precise mechanism of the proarrhythmic effect is not fully understood. In this study, we found that nilotinib (NIL), vandetanib (VAN), and mobocertinib (MOB) induced the development of "cellrhythmia" (arrhythmia-like events) in a concentration-dependent manner in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Continuous administration of NIL, VAN, or MOB in animals significantly prolonged the action potential durations (APD) and increased susceptibility to arrhythmias. Using phosphoproteomic analysis, we identified proteins with altered phosphorylation levels after treatment with 3 μM NIL, VAN, and MOB for 1.5 h. Using these identified proteins as substrates, we performed kinase-substrate enrichment analysis to identify the kinases driving the changes in phosphorylation levels of these proteins. MAPK and WNK were both inhibited by NIL, VAN, and MOB. A selective inhibitor of WNK1, WNK-IN-11, induced concentration- and time-dependent cellrhythmias and prolonged field potential duration (FPD) in hiPSC-CMs in vitro; furthermore, administration in guinea pigs confirmed that WNK-IN-11 prolonged ventricular repolarization and increased susceptibility to arrhythmias. Fingding indicated that WNK1 inhibition had an in vivo and in vitro arrhythmogenic phenotype similar to TKIs. Additionally,three of TKIs reduced hERG and KCNQ1 expression at protein level, not at transcription level. Similarly, the knockdown of WNK1 decreased hERG and KCNQ1 protein expression in hiPSC-CMs. Collectively, our data suggest that the proarrhythmic effects of NIL, VAN, and MOB occur through a kinase inhibition mechanism. NIL, VAN, and MOB inhibit WNK1 kinase, leading to a decrease in hERG and KCNQ1 protein expression, thereby prolonging action potential repolarization and consequently cause arrhythmias.
Collapse
Affiliation(s)
- Wenting Wu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Jinglei Sun
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Jiali Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Haining Zhao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Suhua Qiu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Congxin Li
- Department of Pharmacy, Third Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Chenxia Shi
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
| | - Yanfang Xu
- Department of Pharmacology, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of New Drug Pharmacology and Toxicology, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China.
| |
Collapse
|
5
|
Pierson JB, Berridge B, Blinova K, Brooks MB, Eldridge S, O'Brien CE, Pugsley MK, Schultze AE, Smith G, Stockbridge N, Valentin JP, Vicente J. Collaborative science in action: A 20 year perspective from the Health and Environmental Sciences Institute (HESI) Cardiac Safety Committee. J Pharmacol Toxicol Methods 2024; 127:107511. [PMID: 38710237 DOI: 10.1016/j.vascn.2024.107511] [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: 03/27/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/08/2024]
Abstract
The Health and Environmental Sciences Institute (HESI) is a nonprofit organization dedicated to resolving global health challenges through collaborative scientific efforts across academia, regulatory authorities and the private sector. Collaborative science across non-clinical disciplines offers an important keystone to accelerate the development of safer and more effective medicines. HESI works to address complex challenges by leveraging diverse subject-matter expertise across sectors offering access to resources, data and shared knowledge. In 2008, the HESI Cardiac Safety Committee (CSC) was established to improve public health by reducing unanticipated cardiovascular (CV)-related adverse effects from pharmaceuticals or chemicals. The committee continues to significantly impact the field of CV safety by bringing together experts from across sectors to address challenges of detecting and predicting adverse cardiac outcomes. Committee members have collaborated on the organization, management and publication of prospective studies, retrospective analyses, workshops, and symposia resulting in 38 peer reviewed manuscripts. Without this collaboration these manuscripts would not have been published. Through their work, the CSC is actively addressing challenges and opportunities in detecting potential cardiac failure modes using in vivo, in vitro and in silico models, with the aim of facilitating drug development and improving study design. By examining past successes and future prospects of the CSC, this manuscript sheds light on how the consortium's multifaceted approach not only addresses current challenges in detecting potential cardiac failure modes but also paves the way for enhanced drug development and study design methodologies. Further, exploring future opportunities and challenges will focus on improving the translational predictability of nonclinical evaluations and reducing reliance on animal research in CV safety assessments.
Collapse
Affiliation(s)
| | | | | | - Marjory B Brooks
- Comparative Coagulation Section, Department of Population Medicine and Diagnostic Sciences, Cornell University, Ithaca, NY, USA
| | - Sandy Eldridge
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD, USA
| | - Claire E O'Brien
- Health and Environmental Sciences Institute, Washington, DC, USA.
| | - Michael K Pugsley
- Toxicology & Safety Pharmacology, Cytokinetics, South San Francisco, CA, USA
| | - A Eric Schultze
- Pathology, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Godfrey Smith
- Clyde Biosciences Ltd, Newhouse, UK; University of Glasgow, Scotland, UK
| | | | - Jean-Pierre Valentin
- UCB Biopharma SRL, Development Science, Non-Clinical Safety Evaluation, Braine l'Alleud, Belgium
| | - Jose Vicente
- Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
6
|
Kieda J, Shakeri A, Landau S, Wang EY, Zhao Y, Lai BF, Okhovatian S, Wang Y, Jiang R, Radisic M. Advances in cardiac tissue engineering and heart-on-a-chip. J Biomed Mater Res A 2024; 112:492-511. [PMID: 37909362 PMCID: PMC11213712 DOI: 10.1002/jbm.a.37633] [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: 07/05/2023] [Revised: 09/26/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023]
Abstract
Recent advances in both cardiac tissue engineering and hearts-on-a-chip are grounded in new biomaterial development as well as the employment of innovative fabrication techniques that enable precise control of the mechanical, electrical, and structural properties of the cardiac tissues being modelled. The elongated structure of cardiomyocytes requires tuning of substrate properties and application of biophysical stimuli to drive its mature phenotype. Landmark advances have already been achieved with induced pluripotent stem cell-derived cardiac patches that advanced to human testing. Heart-on-a-chip platforms are now commonly used by a number of pharmaceutical and biotechnology companies. Here, we provide an overview of cardiac physiology in order to better define the requirements for functional tissue recapitulation. We then discuss the biomaterials most commonly used in both cardiac tissue engineering and heart-on-a-chip, followed by the discussion of recent representative studies in both fields. We outline significant challenges common to both fields, specifically: scalable tissue fabrication and platform standardization, improving cellular fidelity through effective tissue vascularization, achieving adult tissue maturation, and ultimately developing cryopreservation protocols so that the tissues are available off the shelf.
Collapse
Affiliation(s)
- Jennifer Kieda
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Amid Shakeri
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Shira Landau
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Erika Yan Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yimu Zhao
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Benjamin Fook Lai
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Sargol Okhovatian
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ying Wang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Richard Jiang
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Milica Radisic
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Yanagida S, Kawagishi H, Kanda Y. [Cardiotoxicity risk assessment of anti-cancer drugs and future perspectives]. Nihon Yakurigaku Zasshi 2024; 159:83-89. [PMID: 38432924 DOI: 10.1254/fpj.23094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Cardiotoxicity is a serious adverse effect of anti-cancer drugs. Anti-cancer drug-induced cardiotoxicity are arrhythmia, cardiac contractile dysfunction, coronary artery disease, and hypertension, which affect to the quality of life in patients with cancer. In particular, cardiac contractile dysfunction is a life-threatening symptom leading to heart failure, suggesting that it is very important to predict the risk of developing the contractile dysfunction by anti-cancer drugs. Recently, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can be used to assess the risk of drug-induced arrhythmias. This prompts us to evaluate other cardiotoxic effects such as contractility dysfunction and structural toxicity with hiPSC-CMs. Since anti-cancer drug-induced contractility dysfunction are considered to be induced by chronic exposure, we have developed a method to assess chronic contractility dysfunction by imaging analysis of hiPSC-CMs. BMS-986094, which failed in clinical trials due to the occurrence of heart failure, was used as a positive compound. We found that chronic exposure to BMS-986094 decreased the contraction and relaxation velocity in hiPSC-CMs. Doxorubicin was observed to decrease cytotoxicity and both contraction and relaxation velocities in hiPSC-CMs. We are currently further evaluating other anti-cancer drugs with different mode-of-actions using hiPSC-CMs and assess the predictivity and utility of contractile assessment using hiPSC-CMs by comparing with real-world data. Here, we introduce our novel method to assess the chronic contractility of hiPSC-CMs by imaging analysis and discuss the future perspectives for assessing the anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
| | | | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
8
|
Yanagida S, Kanda Y. [Prediction of Cardiac Toxicity by Anti-cancer Drugs Using iPSC Cardiomyocytes]. YAKUGAKU ZASSHI 2024; 144:265-271. [PMID: 38432935 DOI: 10.1248/yakushi.23-00164-3] [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] [Indexed: 03/05/2024]
Abstract
Recent advances in cancer therapy have significantly improved the survival rate of patients with cancer. In contrast, anti-cancer drug-induced adverse effects, especially cardiotoxicity, have come to affect patients' prognosis and quality of life. Therefore, there is a growing need to understand the anti-cancer drug-induced cardiotoxicity. Human induced pluripotent stem (iPS) cell-derived cardiomyocytes (hiPSC-CMs) have been used to assess drug-induced cardiotoxicity by improving the predictability of clinical cardiotoxicity and the principles of the 3Rs (replacement, reduction and refinement). To predict the anti-cancer drug-induced cardiotoxicity, we developed a novel method to assess drug-induced proarrhythmia risk using hiPSC-CMs by participating in the international validation. In addition, we established the chronic contractility toxicity assessment by image-based motion analysis. The compound BMS-986094, which was withdrawn from clinical trials, inhibited contractility velocity and relaxation velocity in hiPSC-CMs. Currently, we are trying to investigate the predictability of the contractility assay by comparing the hiPSC-CM data with adverse events reports from real-world database. In this review, we would like to introduce the novel imaging-based contractility method using hiPSC-CMs and future perspectives in anti-cancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences
- Graduate School of Biomedical and Health Sciences (Pharmaceutical Sciences), Hiroshima University
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences
| |
Collapse
|
9
|
Allan A, Creech J, Hausner C, Krajcarski P, Gunawan B, Poulin N, Kozlowski P, Clark CW, Dow R, Saraithong P, Mair DB, Block T, Monteiro da Rocha A, Kim DH, Herron TJ. High-throughput longitudinal electrophysiology screening of mature chamber-specific hiPSC-CMs using optical mapping. iScience 2023; 26:107142. [PMID: 37416454 PMCID: PMC10320609 DOI: 10.1016/j.isci.2023.107142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
hiPSC-CMs are being considered by the Food and Drug Administration and other regulatory agencies for in vitro cardiotoxicity screening to provide human-relevant safety data. Widespread adoption of hiPSC-CMs in regulatory and academic science is limited by the immature, fetal-like phenotype of the cells. Here, to advance the maturation state of hiPSC-CMs, we developed and validated a human perinatal stem cell-derived extracellular matrix coating applied to high-throughput cell culture plates. We also present and validate a cardiac optical mapping device designed for high-throughput functional assessment of mature hiPSC-CM action potentials using voltage-sensitive dye and calcium transients using calcium-sensitive dyes or genetically encoded calcium indicators (GECI, GCaMP6). We utilize the optical mapping device to provide new biological insight into mature chamber-specific hiPSC-CMs, responsiveness to cardioactive drugs, the effect of GCaMP6 genetic variants on electrophysiological function, and the effect of daily β-receptor stimulation on hiPSC-CM monolayer function and SERCA2a expression.
Collapse
Affiliation(s)
- Andrew Allan
- Cairn Research, Graveney Road, Faversham, Kent ME13 8UP UK
| | - Jeffery Creech
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Christian Hausner
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Peyton Krajcarski
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Bianca Gunawan
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Noah Poulin
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Paul Kozlowski
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Christopher Wayne Clark
- University of Michigan, School of Public Health, Department of Environmental Health Sciences, Ann Arbor, MI 48109, USA
| | - Rachel Dow
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
| | - Prakaimuk Saraithong
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Devin B. Mair
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Travis Block
- StemBioSys, Inc, 3463 Magic Drive, Suite 110, San Antonio, TX 78229, USA
| | - Andre Monteiro da Rocha
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Todd J. Herron
- University of Michigan, Frankel Cardiovascular Regeneration Core Laboratory, Ann Arbor, MI 48109, USA
- Michigan Medicine, Internal Medicine-Cardiology, Ann Arbor, MI 48109, USA
- Michigan Medicine, Molecular & Integrative Physiology, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Ma MS, Sundaram S, Lou L, Agarwal A, Chen CS, Bifano TG. High throughput screening system for engineered cardiac tissues. Front Bioeng Biotechnol 2023; 11:1177688. [PMID: 37251575 PMCID: PMC10210164 DOI: 10.3389/fbioe.2023.1177688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction: Three dimensional engineered cardiac tissues (3D ECTs) have become indispensable as in vitro models to assess drug cardiotoxicity, a leading cause of failure in pharmaceutical development. A current bottleneck is the relatively low throughput of assays that measure spontaneous contractile forces exerted by millimeter-scale ECTs typically recorded through precise optical measurement of deflection of the polymer scaffolds that support them. The required resolution and speed limit the field of view to at most a few ECTs at a time using conventional imaging. Methods: To balance the inherent tradeoff among imaging resolution, field of view and speed, an innovative mosaic imaging system was designed, built, and validated to sense contractile force of 3D ECTs seeded on a 96-well plate. Results: The system performance was validated through real-time, parallel contractile force monitoring for up to 3 weeks. Pilot drug testing was conducted using isoproterenol. Discussion: The described tool increases contractile force sensing throughput to 96 samples per measurement; significantly reduces cost, time and labor needed for preclinical cardiotoxicity assay using 3D ECT. More broadly, our mosaicking approach is a general way to scale up image-based screening in multi-well formats.
Collapse
Affiliation(s)
- Marshall S. Ma
- Mechanical Engineering, Boston University, Boston, MA, United States
- Photonics Center, Boston University, Boston, MA, United States
| | | | - Lihua Lou
- Mechanical and Materials Engineering, Florida International University, Miami, FL, United States
| | - Arvind Agarwal
- Mechanical and Materials Engineering, Florida International University, Miami, FL, United States
| | | | - Thomas G. Bifano
- Mechanical Engineering, Boston University, Boston, MA, United States
- Photonics Center, Boston University, Boston, MA, United States
| |
Collapse
|
11
|
Casciola M, Feaster TK, Caiola MJ, Keck D, Blinova K. Human in vitro assay for irreversible electroporation cardiac ablation. Front Physiol 2023; 13:1064168. [PMID: 36699682 PMCID: PMC9869257 DOI: 10.3389/fphys.2022.1064168] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/29/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction: Pulsed electric field (PEF) cardiac ablation has been recently proposed as a technique to treat drug resistant atrial fibrillation by inducing cell death through irreversible electroporation (IRE). Improper PEF dosing can result in thermal damage or reversible electroporation. The lack of comprehensive and systematic studies to select PEF parameters for safe and effective IRE cardiac treatments hinders device development and regulatory decision-making. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been proposed as an alternative to animal models in the evaluation of cardiac electrophysiology safety. Methods: We developed a novel high-throughput in vitro assay to quantify the electric field threshold (EFT) for electroporation (acute effect) and cell death (long-term effect) in hiPSC-CMs. Monolayers of hiPSC-CMs were cultured in high-throughput format and exposed to clinically relevant biphasic PEF treatments. Electroporation and cell death areas were identified using fluorescent probes and confocal microscopy; electroporation and cell death EFTs were quantified by comparison of fluorescent images with electric field numerical simulations. Results: Study results confirmed that PEF induces electroporation and cell death in hiPSC-CMs, dependent on the number of pulses and the amplitude, duration, and repetition frequency. In addition, PEF-induced temperature increase, absorbed dose, and total treatment time for each PEF parameter combination are reported. Discussion: Upon verification of the translatability of the in vitro results presented here to in vivo models, this novel hiPSC-CM-based assay could be used as an alternative to animal or human studies and can assist in early nonclinical device development, as well as inform regulatory decision-making for cardiac ablation medical devices.
Collapse
|
12
|
Yang X, Ribeiro AJS, Pang L, Strauss DG. Use of Human iPSC-CMs in Nonclinical Regulatory Studies for Cardiac Safety Assessment. Toxicol Sci 2022; 190:117-126. [PMID: 36099065 DOI: 10.1093/toxsci/kfac095] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a human-relevant platform for cardiac function assessment. Alternative assays using hiPSC-CMs are increasingly being employed for regulatory decision-making. A retrospective review revealed steady use of hiPSC-CM-based in vitro assays in nonclinical studies of drug-induced cardiotoxicity in regulatory submissions to the U.S. Food and Drug Administration (FDA). Most of the hiPSC-CMs data were obtained in exploratory studies and submitted as supportive evidence in concordance with other nonclinical data. Some of those studies were used to inform clinical trial design. This article provides an overview of the use of hiPSC-CMs in regulatory applications to FDA, with a focus on the integration of human-relevant in vitro data into proarrhythmic and non-proarrhythmic risk assessment. By identifying the regulatory submissions including hiPSC-CMs data, we explore their utility and discuss their limitations for predicting human cardiac safety in clinical trials. An important take-home message is that regulatory acceptance of hiPSC-CMs data is dependent on both the context of use and accurate data interpretation.
Collapse
Affiliation(s)
- Xi Yang
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, & Nephrology, Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| | - Alexandre J S Ribeiro
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arizona 72079, USA
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration , Silver Spring, Maryland 20903, USA
| |
Collapse
|
13
|
Feaster TK, Feric N, Pallotta I, Narkar A, Casciola M, Graziano MP, Aschar-Sobbi R, Blinova K. Acute effects of cardiac contractility modulation stimulation in conventional 2D and 3D human induced pluripotent stem cell-derived cardiomyocyte models. Front Physiol 2022; 13:1023563. [PMID: 36439258 PMCID: PMC9686332 DOI: 10.3389/fphys.2022.1023563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022] Open
Abstract
Cardiac contractility modulation (CCM) is a medical device therapy whereby non-excitatory electrical stimulations are delivered to the myocardium during the absolute refractory period to enhance cardiac function. We previously evaluated the effects of the standard CCM pulse parameters in isolated rabbit ventricular cardiomyocytes and 2D human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) monolayers, on flexible substrate. In the present study, we sought to extend these results to human 3D microphysiological systems to develop a robust model to evaluate various clinical CCM pulse parameters in vitro. HiPSC-CMs were studied in conventional 2D monolayer format, on stiff substrate (i.e., glass), and as 3D human engineered cardiac tissues (ECTs). Cardiac contractile properties were evaluated by video (i.e., pixel) and force-based analysis. CCM pulses were assessed at varying electrical ‘doses’ using a commercial pulse generator. A robust CCM contractile response was observed for 3D ECTs. Under comparable conditions, conventional 2D monolayer hiPSC-CMs, on stiff substrate, displayed no contractile response. 3D ECTs displayed enhanced contractile properties including increased contraction amplitude (i.e., force), and accelerated contraction and relaxation slopes under standard acute CCM stimulation. Moreover, 3D ECTs displayed enhanced contractility in a CCM pulse parameter-dependent manner by adjustment of CCM pulse delay, duration, amplitude, and number relative to baseline. The observed acute effects subsided when the CCM stimulation was stopped and gradually returned to baseline. These data represent the first study of CCM in 3D hiPSC-CM models and provide a nonclinical tool to assess various CCM device signals in 3D human cardiac tissues prior to in vivo animal studies. Moreover, this work provides a foundation to evaluate the effects of additional cardiac medical devices in 3D ECTs.
Collapse
Affiliation(s)
- Tromondae K. Feaster
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Nicole Feric
- Valo Health Inc, Alexandria Center for Life Sciences, New York, NY, United States
| | - Isabella Pallotta
- Valo Health Inc, Alexandria Center for Life Sciences, New York, NY, United States
| | - Akshay Narkar
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Maura Casciola
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Michael P. Graziano
- Valo Health Inc, Alexandria Center for Life Sciences, New York, NY, United States
| | - Roozbeh Aschar-Sobbi
- Valo Health Inc, Alexandria Center for Life Sciences, New York, NY, United States
| | - Ksenia Blinova
- Office of Science and Engineering Laboratories, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, United States
- *Correspondence: Ksenia Blinova,
| |
Collapse
|
14
|
Satsuka A, Hayashi S, Yanagida S, Ono A, Kanda Y. Contractility assessment of human iPSC-derived cardiomyocytes by using a motion vector system and measuring cell impedance. J Pharmacol Toxicol Methods 2022; 118:107227. [PMID: 36243255 DOI: 10.1016/j.vascn.2022.107227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/16/2022] [Accepted: 10/10/2022] [Indexed: 12/31/2022]
Abstract
Predicting drug-induced cardiotoxicity during the non-clinical stage is important to avoid severe consequences in the clinical trials of new drugs. Human iPSC-derived cardiomyocytes (hiPSC-CMs) hold great promise for cardiac safety assessments in drug development. To date, multi-electrode array system (MEA) has been a widely used as a tool for the assessment of proarrhythmic risk with hiPSC-CMs. Recently, new methodologies have been proposed to assess in vitro contractility, such as the force and velocity of cell contraction, using hiPSC-CMs. Herein, we focused on an imaging-based motion vector system (MV) and an electric cell-substrate impedance sensing system (IMP). We compared the output signals of hiPSC-CMs from MV and IMP in detail and observed a clear correlation between the parameters. In addition, we assessed the effects of isoproterenol and verapamil on hiPSC-CM contraction and identified a correlation in the contractile change of parameters obtained with MV and IMP. These results suggest that both assay systems could be used to monitor hiPSC-CM contraction dynamics.
Collapse
Affiliation(s)
- Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kanagawa 210-9501, Japan
| | - Sayo Hayashi
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kanagawa 210-9501, Japan
| | - Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kanagawa 210-9501, Japan; Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-naka, kita-ku, Okayama, Okayama 700-8530, Japan
| | - Atsushi Ono
- Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-naka, kita-ku, Okayama, Okayama 700-8530, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kanagawa 210-9501, Japan; Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 1-1-1, Tsushima-naka, kita-ku, Okayama, Okayama 700-8530, Japan.
| |
Collapse
|
15
|
Visone R, Lozano-Juan F, Marzorati S, Rivolta MW, Pesenti E, Redaelli A, Sassi R, Rasponi M, Occhetta P. Predicting human cardiac QT alterations and pro-arrhythmic effects of compounds with a 3D beating heart-on-chip platform. Toxicol Sci 2022; 191:47-60. [PMID: 36226800 PMCID: PMC9887672 DOI: 10.1093/toxsci/kfac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Determining the potential cardiotoxicity and pro-arrhythmic effects of drug candidates remains one of the most relevant issues in the drug development pipeline (DDP). New methods enabling to perform more representative preclinical in vitro studies by exploiting induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) are under investigation to increase the translational power of the outcomes. Here we present a pharmacological campaign conducted to evaluate the drug-induced QT alterations and arrhythmic events on uHeart, a 3D miniaturized in vitro model of human myocardium encompassing iPSC-CM and dermal fibroblasts embedded in fibrin. uHeart was mechanically trained resulting in synchronously beating cardiac microtissues in 1 week, characterized by a clear field potential (FP) signal that was recorded by means of an integrated electrical system. A drug screening protocol compliant with the new International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines was established and uHeart was employed for testing the effect of 11 compounds acting on single or multiple cardiac ion channels and well-known to elicit QT prolongation or arrhythmic events in clinics. The alterations of uHeart's electrophysiological parameters such as the beating period, the FP duration, the FP amplitude, and the detection of arrhythmic events prior and after drug administration at incremental doses were effectively analyzed through a custom-developed algorithm. Results demonstrated the ability of uHeart to successfully anticipate clinical outcome and to predict the QT prolongation with a sensitivity of 83.3%, a specificity of 100% and an accuracy of 91.6%. Cardiotoxic concentrations of drugs were notably detected in the range of the clinical highest blood drug concentration (Cmax), qualifying uHeart as a fit-to-purpose preclinical tool for cardiotoxicity studies.
Collapse
Affiliation(s)
| | | | | | | | | | - Alberto Redaelli
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | - Roberto Sassi
- Department of Computer Science, Università degli Studi di Milano, Milan, 20133, Italy
| | - Marco Rasponi
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, 20133, Italy
| | | |
Collapse
|
16
|
Trovato C, Mohr M, Schmidt F, Passini E, Rodriguez B. Cross clinical-experimental-computational qualification of in silico drug trials on human cardiac purkinje cells for proarrhythmia risk prediction. FRONTIERS IN TOXICOLOGY 2022; 4:992650. [PMID: 36278026 PMCID: PMC9581132 DOI: 10.3389/ftox.2022.992650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
The preclinical identification of drug-induced cardiotoxicity and its translation into human risk are still major challenges in pharmaceutical drug discovery. The ICH S7B Guideline and Q&A on Clinical and Nonclinical Evaluation of QT/QTc Interval Prolongation and Proarrhythmic Potential promotes human in silico drug trials as a novel tool for proarrhythmia risk assessment. To facilitate the use of in silico data in regulatory submissions, explanatory control compounds should be tested and documented to demonstrate consistency between predictions and the historic validation data. This study aims to quantify drug-induced electrophysiological effects on in silico cardiac human Purkinje cells, to compare them with existing in vitro rabbit data, and to assess their accuracy for clinical pro-arrhythmic risk predictions. The effects of 14 reference compounds were quantified in simulations with a population of in silico human cardiac Purkinje models. For each drug dose, five electrophysiological biomarkers were quantified at three pacing frequencies, and results compared with available in vitro experiments and clinical proarrhythmia reports. Three key results were obtained: 1) In silico, repolarization abnormalities in human Purkinje simulations predicted drug-induced arrhythmia for all risky compounds, showing higher predicted accuracy than rabbit experiments; 2) Drug-induced electrophysiological changes observed in human-based simulations showed a high degree of consistency with in vitro rabbit recordings at all pacing frequencies, and depolarization velocity and action potential duration were the most consistent biomarkers; 3) discrepancies observed for dofetilide, sotalol and terfenadine are mainly caused by species differences between humans and rabbit. Taken together, this study demonstrates higher accuracy of in silico methods compared to in vitro animal models for pro-arrhythmic risk prediction, as well as a high degree of consistency with in vitro experiments commonly used in safety pharmacology, supporting the potential for industrial and regulatory adoption of in silico trials for proarrhythmia prediction.
Collapse
Affiliation(s)
- Cristian Trovato
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Marcel Mohr
- Sanofi-Aventis Deutschland GmbH, R&D Preclinical Safety, Frankfurt, Germany
| | - Friedemann Schmidt
- Sanofi-Aventis Deutschland GmbH, R&D Preclinical Safety, Frankfurt, Germany
| | - Elisa Passini
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| | - Blanca Rodriguez
- Department of Computer Science, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Wei F, Pence L, Woodling K, Bagam P, Beger R, Gamboa da Costa G, Pang L. Effects of Serum and Compound Preparation Methods on Delayed Repolarization Evaluation With Human iPSC-CMs. Toxicol Sci 2022; 188:48-61. [PMID: 35478258 DOI: 10.1093/toxsci/kfac043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2024] Open
Abstract
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been widely used in the Comprehensive in vitro Proarrhythmia Assay (CiPA). The notable difference of the electrophysiological (EP) responses of hiPSC-CMs in serum and serum-free media (SFM) is puzzling and may impact regulatory decision-making on the cardiac safety of candidate drugs in inducing QT prolongation and torsade de pointes (TdP). In this study, we compared the EP responses of hiPSC-CMs to 10 CiPA compounds and moxifloxacin in serum and SFM; explained the potential reason behind the different EP responses—abiotic compound loss to plastic tubes/plates of hydrophobic compounds prepared in SFM; and investigated the impact of compound preparation methods on drug bioavailability in exposure media, which affects the TdP risk prediction of drugs tested in serum-containing and SFM. For assays to be conducted in SFM, awareness of abiotic compound loss of hydrophobic compounds in serum-free preparations is critical for delay repolarization evaluation and data extrapolation from in vitro to in vivo.
Collapse
Affiliation(s)
- Feng Wei
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
- Department of Structural Heart Disease, the First Affiliated Hospital of Xi’an Jiaotong University , Xi’an, Shaanxi 710061, China
| | - Lisa Pence
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Kellie Woodling
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Prathyusha Bagam
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Richard Beger
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Goncalo Gamboa da Costa
- Office of the Director, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| | - Li Pang
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson, Arkansas 72079, USA
| |
Collapse
|
18
|
Downregulation of hERG Channel Expression By Tyrosine Kinase Inhibitors Nilotinib And Vandetanib Predominantly Contributes To Arrhythmogenesis. Toxicol Lett 2022; 365:11-23. [DOI: 10.1016/j.toxlet.2022.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/11/2022] [Accepted: 06/03/2022] [Indexed: 11/20/2022]
|
19
|
Park SJ, Yeon SK, Kim Y, Kim HJ, Kim S, Kim J, Choi JW, Kim B, Lee EH, Kim R, Seo SH, Lee J, Kim JW, Lee HY, Hwang H, Bahn YS, Cheong E, Park JH, Park KD. Discovery of Novel Sphingosine-1-Phosphate-1 Receptor Agonists for the Treatment of Multiple Sclerosis. J Med Chem 2022; 65:3539-3562. [PMID: 35077170 DOI: 10.1021/acs.jmedchem.1c01979] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The sphingosine-1-phosphate-1 (S1P1) receptor agonists have great potential for the treatment of multiple sclerosis (MS) because they can inhibit lymphocyte egress through receptor internalization. We designed and synthesized triazole and isoxazoline derivatives to discover a novel S1P1 agonist for MS treatment. Of the two scaffolds, the isoxazoline derivative was determined to have excellent in vitro efficacy and drug-like properties. Among them, compound 21l was found to have superior drug-like properties as well as excellent in vitro efficacies (EC50 = 7.03 nM in β-arrestin recruitment and EC50 = 11.8 nM in internalization). We also confirmed that 21l effectively inhibited lymphocyte egress in the peripheral lymphocyte count test and significantly improved the clinical score in the experimental autoimmune encephalitis MS mouse model.
Collapse
Affiliation(s)
- Sun Jun Park
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.,Cureverse Co., Ltd., KIST, 1st Floor, H2 Building, Seoul 02792, Republic of Korea
| | - Seul Ki Yeon
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
| | - Yoowon Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyeon Jeong Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Siwon Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jushin Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji Won Choi
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
| | - Byungeun Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Elijah Hwejin Lee
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Rium Kim
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Seon Hee Seo
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea
| | - Jaeick Lee
- Doping Control Center, KIST, Seoul 02792, Republic of Korea
| | - Jun Woo Kim
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Ha-Yeon Lee
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Eunji Cheong
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jong-Hyun Park
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ki Duk Park
- Convergence Research Center for Diagnosis, Treatment & Care System of Dementia, Korea Institute of Science & Technology (KIST), Seoul 02792, Republic of Korea.,Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
20
|
Kilfoil P, Feng SL, Bassyouni A, Lee T, Leishman D, Li D, MacEwan DJ, Sharma P, Watt ED, Jenkinson S. Characterization of a high throughput human stem cell cardiomyocyte assay to predict drug-induced changes in clinical electrocardiogram parameters. Eur J Pharmacol 2021; 912:174584. [PMID: 34678241 DOI: 10.1016/j.ejphar.2021.174584] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 01/12/2023]
Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hIPSC-CM's) play an increasingly important role in the safety profiling of candidate drugs. For such models to have utility a clear understanding of clinical translation is required. In the present study we examined the ability of our hIPSC-CM model to predict the clinically observed effects of a diverse set of compounds on several electrocardiogram endpoints, including changes in QT and QRS intervals. To achieve this, compounds were profiled in a novel high throughput voltage-sensitive dye platform. Measurements were taken acutely (30 min) and chronically (24 h) to ensure that responses from compounds with slow onset kinetics or that affected surface ion channel expression would be captured. In addition, to avoid issues associated with changes in free drug levels due to protein binding, assays were run in serum free conditions. Changes in hIPSC-CM threshold APD90 values correlated with compound plasma exposures that produced a +10 ms change in clinical QTc (Pearson r2 = 0.80). In addition, randomForest modeling showed high predictivity in defining TdP risk (AUROC value = 0.938). Risk associated with QRS prolongation correlated with an increase in action potential rise-time (AUROC value = 0.982). The in-depth understanding of the clinical translatability of our hIPSC-CM model positions this assay to play a key role in defining cardiac risk early in drug development. Moreover, the ability to perform longer term studies enables the detection of compounds that may not be highlighted by more acute assay formats, such as inhibitors of hERG trafficking.
Collapse
Affiliation(s)
- Peter Kilfoil
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Shuyun Lily Feng
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Asser Bassyouni
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Tiffany Lee
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA
| | - Derek Leishman
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | | | - David J MacEwan
- Department of Pharmacology and Therapeutics, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, L69 3GE, UK
| | - Parveen Sharma
- Department of Cardiovascular & Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, L69 3GE, UK
| | | | - Stephen Jenkinson
- Worldwide Research and Development, Pfizer Inc., La Jolla, CA, 92121, USA.
| |
Collapse
|
21
|
Feaster TK, Casciola M, Narkar A, Blinova K. Acute effects of cardiac contractility modulation on human induced pluripotent stem cell-derived cardiomyocytes. Physiol Rep 2021; 9:e15085. [PMID: 34729935 PMCID: PMC8564440 DOI: 10.14814/phy2.15085] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiac contractility modulation (CCM) is an intracardiac therapy whereby nonexcitatory electrical simulations are delivered during the absolute refractory period of the cardiac cycle. We previously evaluated the effects of CCM in isolated adult rabbit ventricular cardiomyocytes and found a transient increase in calcium and contractility. In the present study, we sought to extend these results to human cardiomyocytes using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to develop a robust model to evaluate CCM in vitro. HiPSC-CMs (iCell Cardiomyocytes2 , Fujifilm Cellular Dynamic, Inc.) were studied in monolayer format plated on flexible substrate. Contractility, calcium handling, and electrophysiology were evaluated by fluorescence- and video-based analysis (CellOPTIQ, Clyde Biosciences). CCM pulses were applied using an A-M Systems 4100 pulse generator. Robust hiPSC-CMs response was observed at 14 V/cm (64 mA) for pacing and 28 V/cm (128 mA, phase amplitude) for CCM. Under these conditions, hiPSC-CMs displayed enhanced contractile properties including increased contraction amplitude and faster contraction kinetics. Likewise, calcium transient amplitude increased, and calcium kinetics were faster. Furthermore, electrophysiological properties were altered resulting in shortened action potential duration (APD). The observed effects subsided when the CCM stimulation was stopped. CCM-induced increase in hiPSC-CMs contractility was significantly more pronounced when extracellular calcium concentration was lowered from 2 mM to 0.5 mM. This study provides a comprehensive characterization of CCM effects on hiPSC-CMs. These data represent the first study of CCM in hiPSC-CMs and provide an in vitro model to assess physiologically relevant mechanisms and evaluate safety and effectiveness of future cardiac electrophysiology medical devices.
Collapse
Affiliation(s)
- Tromondae K. Feaster
- Office of Science and Engineering LaboratoriesCenter for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Maura Casciola
- Office of Science and Engineering LaboratoriesCenter for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Akshay Narkar
- Office of Science and Engineering LaboratoriesCenter for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Ksenia Blinova
- Office of Science and Engineering LaboratoriesCenter for Devices and Radiological HealthUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
22
|
Aguayo-Ortiz R, Creech J, Jiménez-Vázquez EN, Guerrero-Serna G, Wang N, da Rocha AM, Herron TJ, Espinoza-Fonseca LM. A multiscale approach for bridging the gap between potency, efficacy, and safety of small molecules directed at membrane proteins. Sci Rep 2021; 11:16580. [PMID: 34400719 PMCID: PMC8368179 DOI: 10.1038/s41598-021-96217-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 08/06/2021] [Indexed: 01/17/2023] Open
Abstract
Membrane proteins constitute a substantial fraction of the human proteome, thus representing a vast source of therapeutic drug targets. Indeed, newly devised technologies now allow targeting "undruggable" regions of membrane proteins to modulate protein function in the cell. Despite the advances in technology, the rapid translation of basic science discoveries into potential drug candidates targeting transmembrane protein domains remains challenging. We address this issue by harmonizing single molecule-based and ensemble-based atomistic simulations of ligand-membrane interactions with patient-derived induced pluripotent stem cell (iPSC)-based experiments to gain insights into drug delivery, cellular efficacy, and safety of molecules directed at membrane proteins. In this study, we interrogated the pharmacological activation of the cardiac Ca2+ pump (Sarcoplasmic reticulum Ca2+-ATPase, SERCA2a) in human iPSC-derived cardiac cells as a proof-of-concept model. The combined computational-experimental approach serves as a platform to explain the differences in the cell-based activity of candidates with similar functional profiles, thus streamlining the identification of drug-like candidates that directly target SERCA2a activation in human cardiac cells. Systematic cell-based studies further showed that a direct SERCA2a activator does not induce cardiotoxic pro-arrhythmogenic events in human cardiac cells, demonstrating that pharmacological stimulation of SERCA2a activity is a safe therapeutic approach targeting the heart. Overall, this novel multiscale platform encompasses organ-specific drug potency, efficacy, and safety, and opens new avenues to accelerate the bench-to-patient research aimed at designing effective therapies directed at membrane protein domains.
Collapse
Affiliation(s)
- Rodrigo Aguayo-Ortiz
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico, Mexico
| | - Jeffery Creech
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eric N Jiménez-Vázquez
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Guadalupe Guerrero-Serna
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nulang Wang
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andre Monteiro da Rocha
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Todd J Herron
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA
- Frankel Cardiovascular Regeneration Core Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
- CARTOX, Inc., 56655 Grand River Ave., PO Box 304, New Hudson, MI, 48165, USA
| | - L Michel Espinoza-Fonseca
- Division of Cardiovascular Medicine, Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Arai K, Kitsuka T, Nakayama K. Scaffold-based and scaffold-free cardiac constructs for drug testing. Biofabrication 2021; 13. [PMID: 34233316 DOI: 10.1088/1758-5090/ac1257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
The safety and therapeutic efficacy of new drugs are tested in experimental animals. However, besides being a laborious, costly process, differences in drug responses between humans and other animals and potential cardiac adverse effects lead to the discontinued development of new drugs. Thus, alternative approaches to animal tests are needed. Cardiotoxicity and responses of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to drugs are conventionally evaluated by cell seeding and two-dimensional (2D) culture, which allows measurements of field potential duration and the action potentials of CMs using multielectrode arrays. However, 2D-cultured hiPSC-CMs lack 3D spatial adhesion, and have fewer intercellular and extracellular matrix interactions, as well as different contractile behavior from CMsin vivo. This issue has been addressed using tissue engineering to fabricate three-dimensional (3D) cardiac constructs from hiPSC-CMs culturedin vitro. Tissue engineering can be categorized as scaffold-based and scaffold-free. In scaffold-based tissue engineering, collagen and fibrin gel scaffolds comprise a 3D culture environment in which seeded cells exhibit cardiac-specific functions and drug responses, whereas 3D cardiac constructs fabricated by tissue engineering without a scaffold have high cell density and form intercellular interactions. This review summarizes the characteristics of scaffold-based and scaffold-free cardiac tissue engineering and discusses the applications of fabricated cardiac constructs to drug screening.
Collapse
Affiliation(s)
- Kenichi Arai
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan.,Department of Clinical Biomaterial Applied Science, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Takahiro Kitsuka
- Department of Cardiovascular Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| |
Collapse
|
24
|
Yanagida S, Satsuka A, Hayashi S, Ono A, Kanda Y. Comprehensive Cardiotoxicity Assessment of COVID-19 Treatments Using Human Induced Pluripotent Stem Cell-derived Cardiomyocytes. Toxicol Sci 2021; 183:227-239. [PMID: 34142159 DOI: 10.1093/toxsci/kfab079] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) continues to spread across the globe, with numerous clinical trials underway seeking to develop and test effective COVID-19 therapies, including remdesivir. Several ongoing studies have reported hydroxychloroquine-induced cardiotoxicity, including development of torsade de pointes (TdP). Meanwhile, human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are expected to serve as a tool for assessing drug-induced cardiotoxicity, such as TdP and contraction impairment. However, the cardiotoxicity of COVID-19 treatments has not been fully assessed using hiPSC-CMs. In the present study, we focused on drug repurposing with various modes of actions and examined the TdP risk associated with COVID-19 treatments using field potential using multi-electrode array (MEA) system and motion analysis with hiPSC-CMs. Hydroxychloroquine induced early after depolarization, while remdesivir, favipiravir, camostat and ivermectin had little effect on field potentials. We then analyzed electromechanical window (EMw), which is defined as the difference between field potential and contraction-relaxation durations. Hydroxychloroquine decreased EMw of hiPSC-CMs in a concentration-dependent manner. In contrast, other drugs have little effect. Our data suggest that hydroxychloroquine has proarrhythmic risk and other drugs have low proarrhythmic risk. Thus, hiPSC-CMs represent a useful tool for assessing the comprehensive cardiotoxicity caused by COVID-19 treatments in non-clinical settings.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kanagawa, Japan.,Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kanagawa, Japan
| | - Sayo Hayashi
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kanagawa, Japan
| | - Atsushi Ono
- Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS), Kanagawa, Japan
| |
Collapse
|
25
|
Ziegler R, Häusermann F, Kirchner S, Polonchuk L. Cardiac Safety of Kinase Inhibitors - Improving Understanding and Prediction of Liabilities in Drug Discovery Using Human Stem Cell-Derived Models. Front Cardiovasc Med 2021; 8:639824. [PMID: 34222360 PMCID: PMC8242589 DOI: 10.3389/fcvm.2021.639824] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/31/2021] [Indexed: 11/13/2022] Open
Abstract
Many small molecule kinase inhibitors (SMKIs) used to fight cancer have been associated with cardiotoxicity in the clinic. Therefore, preventing their failure in clinical development is a priority for preclinical discovery. Our study focused on the integration and concurrent measurement of ATP, apoptosis dynamics and functional cardiac indexes in human stem cell-derived cardiomyocytes (hSC-CMs) to provide further insights into molecular determinants of compromised cardiac function. Ten out of the fourteen tested SMKIs resulted in a biologically relevant decrease in either beating rate or base impedance (cell number index), illustrating cardiotoxicity as one of the major safety liabilities of SMKIs, in particular of those involved in the PI3K–AKT pathway. Pearson's correlation analysis indicated a good correlation between the different read-outs of functional importance. Therefore, measurement of ATP concentrations and apoptosis in vitro could provide important insight into mechanisms of cardiotoxicity. Detailed investigation of the cellular signals facilitated multi-parameter evaluation allowing integrative assessment of cardiomyocyte behavior. The resulting correlation can be used as a tool to highlight changes in cardiac function and potentially to categorize drugs based on their mechanisms of action.
Collapse
Affiliation(s)
- Ricarda Ziegler
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Fabian Häusermann
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Stephan Kirchner
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Liudmila Polonchuk
- Pharmaceutical Sciences, Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
26
|
Deo M, Akwaboah A, Tsevi B, Treat JA, Cordeiro JM. Role of the rapid delayed rectifier K + current in human induced pluripotent stem cells derived cardiomyocytes. ARCHIVES OF STEM CELL AND THERAPY 2021; 1:14-18. [PMID: 33604593 PMCID: PMC7889062 DOI: 10.46439/stemcell.1.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Makarand Deo
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Akwasi Akwaboah
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Bright Tsevi
- Department of Engineering, Norfolk State University, Norfolk, Virginia, USA
| | - Jacqueline A Treat
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| | - Jonathan M Cordeiro
- Department of Experimental Cardiology, Masonic Medical Research Institute, Utica, New York, USA
| |
Collapse
|
27
|
Strauss DG, Wu WW, Li Z, Koerner J, Garnett C. Translational Models and Tools to Reduce Clinical Trials and Improve Regulatory Decision Making for QTc and Proarrhythmia Risk (ICH E14/S7B Updates). Clin Pharmacol Ther 2021; 109:319-333. [PMID: 33332579 PMCID: PMC7898549 DOI: 10.1002/cpt.2137] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 01/06/2023]
Abstract
After multiple drugs were removed from the market secondary to drug-induced torsade de pointes (TdP) risk, the International Council for Harmonisation (ICH) released guidelines in 2005 that focused on the nonclinical (S7B) and clinical (E14) assessment of surrogate biomarkers for TdP. Recently, Vargas et al. published a pharmaceutical-industry perspective making the case that "double-negative" nonclinical data (negative in vitro hERG and in vivo heart-rate corrected QT (QTc) assays) are associated with such low probability of clinical QTc prolongation and TdP that potentially all double-negative drugs would not need detailed clinical QTc evaluation. Subsequently, the ICH released a new E14/S7B Draft Guideline containing Questions and Answers (Q&As) that defined ways that double-negative nonclinical data could be used to reduce the number of "Thorough QT" (TQT) studies and reach a low-risk determination when a TQT or equivalent could not be performed. We review the Vargas et al. proposal in the context of what was contained in the ICH E14/S7B Draft Guideline and what was proposed by the ICH E14/S7B working group for a "stage 2" of updates (potential expanded roles for nonclinical data and details for assessing TdP risk of QTc-prolonging drugs). Although we do not agree with the exact probability statistics in the Vargas et al. paper because of limitations in the underlying datasets, we show how more modest predictive value of individual assays could still result in low probability for TdP with double-negative findings. Furthermore, we expect that the predictive value of the nonclinical assays will improve with implementation of the new ICH E14/S7B Draft Guideline.
Collapse
Affiliation(s)
- David G. Strauss
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Wendy W. Wu
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Zhihua Li
- Division of Applied Regulatory ScienceOffice of Clinical PharmacologyOffice of Translational SciencesCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - John Koerner
- Division of Pharm/Tox for Cardiology, Hematology, Endocrinology and NephrologyOffice of Cardiology, Hematology, Endocrinology and NephrologyOffice of New DrugsCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Christine Garnett
- Division of Cardiology and NephrologyOffice of Cardiology, Hematology, Endocrinology and NephrologyOffice of New DrugsCenter for Drug Evaluation and ResearchUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
28
|
Yanagida S, Satsuka A, Hayashi S, Ono A, Kanda Y. Chronic cardiotoxicity assessment of BMS-986094, a guanosine nucleotide analogue, using human iPS cell-derived cardiomyocytes. J Toxicol Sci 2021; 46:359-369. [PMID: 34334557 DOI: 10.2131/jts.46.359] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Predicting drug-induced side effects in the cardiovascular system is very important because it can lead to the discontinuation of new drugs/candidates or the withdrawal of marketed drugs. Although chronic assessment of cardiac contractility is an important issue in safety pharmacology, an in vitro evaluation system has not been fully developed. We previously developed an imaging-based contractility assay system to detect acute cardiotoxicity using human iPS cell-derived cardiomyocytes (hiPSC-CMs). To extend the system to chronic toxicity assessment, we examined the effects of the anti-hepatitis C virus (HCV) drug candidate BMS-986094, a guanosine nucleotide analogue, which was withdrawn from phase 2 clinical trials because of unexpected contractility toxicities. Additionally, we examined sofosbuvir, another nucleotide analogue inhibitor of HCV that has been approved as an anti-HCV drug. Motion imaging analysis revealed the difference in cardiotoxicity between the cardiotoxic BMS-986094 and the less toxic sofosbuvir in hiPSC-CMs, with a minimum of 4 days of treatment. In addition, we found that BMS-986094-induced contractility impairment was mediated by a decrease in calcium transient. These data suggest that chronic treatment improves the predictive power for the cardiotoxicity of anti-HCV drugs. Thus, hiPSC-CMs can be a useful tool to assess drug-induced chronic cardiotoxicity in non-clinical settings.
Collapse
Affiliation(s)
- Shota Yanagida
- Division of Pharmacology, National Institute of Health Sciences (NIHS).,Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Ayano Satsuka
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Sayo Hayashi
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| | - Atsushi Ono
- Division of Pharmaceutical Sciences, Graduated School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University
| | - Yasunari Kanda
- Division of Pharmacology, National Institute of Health Sciences (NIHS)
| |
Collapse
|