1
|
Begovic M, Schneider L, Zhou X, Hamdani N, Akin I, El-Battrawy I. The Role of Human-Induced Pluripotent Stem Cells in Studying Cardiac Channelopathies. Int J Mol Sci 2024; 25:12034. [PMID: 39596103 PMCID: PMC11593457 DOI: 10.3390/ijms252212034] [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: 08/23/2024] [Revised: 10/22/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiac channelopathies are inherited diseases that increase the risk of sudden cardiac death. While different genes have been associated with inherited channelopathies, there are still subtypes, e.g., catecholaminergic polymorphic ventricular tachycardia and Brugada syndrome, where the genetic cause remains unknown. Various models, including animal models, heterologous expression systems, and the human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSCs-CMs) model, have been used to study the pathophysiological mechanisms of channelopathies. Recently, researchers have focused on using hiPSCs-CMs to understand the genotype-phenotype correlation and screen drugs. By combining innovative techniques such as Clustered Regularly Interspaced Short Palindromic Repeats/Clustered Regularly Interspaced Short Palindromic Repeats associated protein 9 (CRISPR/Cas9)-mediated genome editing, and three-dimensional (3D) engineered heart tissues, we can gain new insights into the pathophysiological mechanisms of channelopathies. This approach holds promise for improving personalized drug treatment. This review highlights the role of hiPSCs-CMs in understanding the pathomechanism of Brugada syndrome and catecholaminergic polymorphic ventricular tachycardia and how these models can be utilized for drug screening.
Collapse
Affiliation(s)
- Merima Begovic
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Luca Schneider
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Xiaobo Zhou
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China
| | - Nazha Hamdani
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Physiology, Cardiovascular Research Institute, University Maastricht, 6229HX Maastricht, The Netherlands
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Intézet címe Semmelweis University, 1089 Budapest, Hungary
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Ibrahim El-Battrawy
- Institute of Physiology, Department of Cellular and Translational Physiology, Ruhr-University Bochum, 44801 Bochum, Germany; (M.B.); (L.S.); (N.H.)
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
- Department of Cardiology and Rhythmology, St. Josef Hospital, Ruhr University, 44791 Bochum, Germany
| |
Collapse
|
2
|
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
|
3
|
Tsai HHD, Ford LC, Burnett SD, Dickey AN, Wright FA, Chiu WA, Rusyn I. Informing Hazard Identification and Risk Characterization of Environmental Chemicals by Combining Transcriptomic and Functional Data from Human-Induced Pluripotent Stem-Cell-Derived Cardiomyocytes. Chem Res Toxicol 2024; 37:1428-1444. [PMID: 39046974 DOI: 10.1021/acs.chemrestox.4c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Environmental chemicals may contribute to the global burden of cardiovascular disease, but experimental data are lacking to determine which substances pose the greatest risk. Human-induced pluripotent stem cell (iPSC)-derived cardiomyocytes are a high-throughput cardiotoxicity model that is widely used to test drugs and chemicals; however, most studies focus on exploring electro-physiological readouts. Gene expression data may provide additional molecular insights to be used for both mechanistic interpretation and dose-response analyses. Therefore, we hypothesized that both transcriptomic and functional data in human iPSC-derived cardiomyocytes may be used as a comprehensive screening tool to identify potential cardiotoxicity hazards and risks of the chemicals. To test this hypothesis, we performed concentration-response analysis of 464 chemicals from 12 classes, including both pharmaceuticals and nonpharmaceutical substances. Functional effects (beat frequency, QT prolongation, and asystole), cytotoxicity, and whole transcriptome response were evaluated. Points of departure were derived from phenotypic and transcriptomic data, and risk characterization was performed. Overall, 244 (53%) substances were active in at least one phenotype; as expected, pharmaceuticals with known cardiac liabilities were the most active. Positive chronotropy was the functional phenotype activated by the largest number of tested chemicals. No chemical class was particularly prone to pose a potential hazard to cardiomyocytes; a varying proportion (10-44%) of substances in each class had effects on cardiomyocytes. Transcriptomic data showed that 69 (15%) substances elicited significant gene expression changes; most perturbed pathways were highly relevant to known key characteristics of human cardiotoxicants. The bioactivity-to-exposure ratios showed that phenotypic- and transcriptomic-based POD led to similar results for risk characterization. Overall, our findings demonstrate how the integrative use of in vitro transcriptomic and phenotypic data from iPSC-derived cardiomyocytes not only offers a complementary approach for hazard and risk prioritization, but also enables mechanistic interpretation of the in vitro test results to increase confidence in decision-making.
Collapse
Affiliation(s)
- Han-Hsuan D Tsai
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Lucie C Ford
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Sarah D Burnett
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Allison N Dickey
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27603, United States
| | - Fred A Wright
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27603, United States
- Department of Statistics and Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina 27603, United States
| | - Weihsueh A Chiu
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| | - Ivan Rusyn
- Interdisciplinary Faculty of Toxicology, Texas A&M University, College Station, Texas 77843, United States
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
4
|
Wakatsuki T, Daily N, Hisada S, Nunomura K, Lin B, Zushida K, Honda Y, Asyama M, Takasuna K. Bayesian approach enabled objective comparison of multiple human iPSC-derived Cardiomyocytes' Proarrhythmia sensitivities. J Pharmacol Toxicol Methods 2024; 128:107531. [PMID: 38852688 DOI: 10.1016/j.vascn.2024.107531] [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: 11/27/2023] [Revised: 05/07/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
The one-size-fits-all approach has been the mainstream in medicine, and the well-defined standards support the development of safe and effective therapies for many years. Advancing technologies, however, enabled precision medicine to treat a targeted patient population (e.g., HER2+ cancer). In safety pharmacology, computational population modeling has been successfully applied in virtual clinical trials to predict drug-induced proarrhythmia risks against a wide range of pseudo cohorts. In the meantime, population modeling in safety pharmacology experiments has been challenging. Here, we used five commercially available human iPSC-derived cardiomyocytes growing in 384-well plates and analyzed the effects of ten potential proarrhythmic compounds with four concentrations on their calcium transients (CaTs). All the cell lines exhibited an expected elongation or shortening of calcium transient duration with various degrees. Depending on compounds inhibiting several ion channels, such as hERG, peak and late sodium and L-type calcium or IKs channels, some of the cell lines exhibited irregular, discontinuous beating that was not predicted by computational simulations. To analyze the shapes of CaTs and irregularities of beat patterns comprehensively, we defined six parameters to characterize compound-induced CaT waveform changes, successfully visualizing the similarities and differences in compound-induced proarrhythmic sensitivities of different cell lines. We applied Bayesian statistics to predict sample populations based on experimental data to overcome the limited number of experimental replicates in high-throughput assays. This process facilitated the principal component analysis to classify compound-induced sensitivities of cell lines objectively. Finally, the association of sensitivities in compound-induced changes between phenotypic parameters and ion channel inhibitions measured using patch clamp recording was analyzed. Successful ranking of compound-induced sensitivity of cell lines was in lined with visual inspection of raw data.
Collapse
Affiliation(s)
- Tetsuro Wakatsuki
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; InvivoSciences, Inc., Madison, WI 53719, USA.
| | - Neil Daily
- InvivoSciences, Inc., Madison, WI 53719, USA
| | - Sunao Hisada
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Hamamatsu Photonics, K.K. Systems Division, Shizuoka 431-3196, Japan
| | - Kazuto Nunomura
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Osaka 565-0871, Japan
| | - Bangzhong Lin
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Center for Supporting Drug Discovery and Life Science Research, Graduate School of Pharmaceutical Science, Osaka University, Osaka 565-0871, Japan
| | - Ko Zushida
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; FUJIFILM Wako Pure Chemical Corporation, Osaka 540-8605, Japan
| | - Yayoi Honda
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Sumika Chemical Analysis Service, Ltd. (SCAS), Osaka 554-0022, Japan
| | - Mahoko Asyama
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Mitsubishi Tanabe Pharma Corporation, Kanagawa 251-8555, Japan
| | - Kiyoshi Takasuna
- Consortium for Safety Assessment Using Human iPS Cells (CSAHi), HEART Team, Tokyo, Japan; Daiichi Sankyo RD Novare Co., Ltd., Tokyo 134-8630, Japan; Axcelead Drug Discovery Partners, Inc., Kanagawa 251-0012, Japan
| |
Collapse
|
5
|
Shi X, Rao R, Xu M, Dong M, Feng S, Huang Y, Zhou B. Methylcellulose improves dissociation quality of adult human primary cardiomyocytes. Heliyon 2024; 10:e31653. [PMID: 38841456 PMCID: PMC11152705 DOI: 10.1016/j.heliyon.2024.e31653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
Obtaining high-quality adult human primary cardiomyocytes (hPCM) have been technically challenging due to isolation-induced biochemical and mechanical stress. Building upon a previous tissue slicing-assisted digestion method, we introduced polymers into the digestion solution to reduce mechanical damage to cells. We found that low-viscosity methylcellulose (MC) significantly improved hPCM viability and yield. Mechanistically, it protected cells from membrane damage, which led to decreased apoptosis and mitochondrial reactive oxygen species production. MC also improved the electrophysiological properties of hPCMs by maintaining the density of sodium channels. The effects on cell viability and cell yield effects were not recapitulated by MC of larger viscosities, other cellulose derivatives, nor shear protectants polyethylene glycol and polyvinyl alcohol. Finally, MC also enhanced the isolation efficiency and the culture quality of hPCMs from diseased ventricular myocardium, expanding its potential applications. Our findings showed that the isolation quality of hPCMs can be further improved through the addition of a polymer, rendering hPCMs a more reliable cellular model for cardiac research.
Collapse
Affiliation(s)
- Xun Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Rongjia Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Miaomiao Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Mengqi Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Shanshan Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Science, Shenzhen, Shenzhen, China
| |
Collapse
|
6
|
Daskalopoulos EP, Deceuninck P, Whelan M, Gribaldo L. Transition to innovative, human-relevant pre-clinical cardiovascular research: a perspective. Cardiovasc Res 2024; 120:e26-e29. [PMID: 38717065 PMCID: PMC11135634 DOI: 10.1093/cvr/cvae080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/12/2024] [Accepted: 02/14/2024] [Indexed: 05/30/2024] Open
Affiliation(s)
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027 Ispra (VA), Italy
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027 Ispra (VA), Italy
| | - Laura Gribaldo
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027 Ispra (VA), Italy
| |
Collapse
|
7
|
Raniga K, Nasir A, Vo NTN, Vaidyanathan R, Dickerson S, Hilcove S, Mosqueira D, Mirams GR, Clements P, Hicks R, Pointon A, Stebbeds W, Francis J, Denning C. Strengthening cardiac therapy pipelines using human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 2024; 31:292-311. [PMID: 38366587 DOI: 10.1016/j.stem.2024.01.007] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 01/19/2024] [Indexed: 02/18/2024]
Abstract
Advances in hiPSC isolation and reprogramming and hPSC-CM differentiation have prompted their therapeutic application and utilization for evaluating potential cardiovascular safety liabilities. In this perspective, we showcase key efforts toward the large-scale production of hiPSC-CMs, implementation of hiPSC-CMs in industry settings, and recent clinical applications of this technology. The key observations are a need for traceable gender and ethnically diverse hiPSC lines, approaches to reduce cost of scale-up, accessible clinical trial datasets, and transparent guidelines surrounding the safety and efficacy of hiPSC-based therapies.
Collapse
Affiliation(s)
- Kavita Raniga
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK; Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK.
| | - Aishah Nasir
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Nguyen T N Vo
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | | | | | | | - Diogo Mosqueira
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Gary R Mirams
- Centre for Mathematical Medicine & Biology, School of Mathematical Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | - Peter Clements
- Pathology, Non-Clinical Safety, GlaxoSmithKline R&D, Stevenage SG1 2NY, UK
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy Department, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, London WC2R 2LS, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | | | - Jo Francis
- Mechanstic Biology and Profiling, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Chris Denning
- The Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
8
|
Seal S, Spjuth O, Hosseini-Gerami L, García-Ortegón M, Singh S, Bender A, Carpenter AE. Insights into Drug Cardiotoxicity from Biological and Chemical Data: The First Public Classifiers for FDA Drug-Induced Cardiotoxicity Rank. J Chem Inf Model 2024; 64:1172-1186. [PMID: 38300851 PMCID: PMC10900289 DOI: 10.1021/acs.jcim.3c01834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
Drug-induced cardiotoxicity (DICT) is a major concern in drug development, accounting for 10-14% of postmarket withdrawals. In this study, we explored the capabilities of chemical and biological data to predict cardiotoxicity, using the recently released DICTrank data set from the United States FDA. We found that such data, including protein targets, especially those related to ion channels (e.g., hERG), physicochemical properties (e.g., electrotopological state), and peak concentration in plasma offer strong predictive ability for DICT. Compounds annotated with mechanisms of action such as cyclooxygenase inhibition could distinguish between most-concern and no-concern DICT. Cell Painting features for ER stress discerned most-concern cardiotoxic from nontoxic compounds. Models based on physicochemical properties provided substantial predictive accuracy (AUCPR = 0.93). With the availability of omics data in the future, using biological data promises enhanced predictability and deeper mechanistic insights, paving the way for safer drug development. All models from this study are available at https://broad.io/DICTrank_Predictor.
Collapse
Affiliation(s)
- Srijit Seal
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Ola Spjuth
- Department
of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box
591, SE-75124 Uppsala, Sweden
| | - Layla Hosseini-Gerami
- Ignota
Labs, The Bradfield Centre, Cambridge Science Park, County Hall, Westminster Bridge Road, Cambridge CB4 0GA, U.K.
| | - Miguel García-Ortegón
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Shantanu Singh
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Andreas Bender
- Yusuf
Hamied Department of Chemistry, University
of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K.
| | - Anne E. Carpenter
- Imaging
Platform, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
9
|
Mozneb M, Jenkins A, Sances S, Pohlman S, Workman MJ, West D, Ondatje B, El-Ghazawi K, Woodbury A, Garcia VJ, Patel S, Arzt M, Dezem F, Laperle AH, Moser VA, Ho R, Yucer N, Plummer J, Barrett RJ, Svendsen CN, Sharma A. Multi-lineage heart-chip models drug cardiotoxicity and enhances maturation of human stem cell-derived cardiovascular cells. LAB ON A CHIP 2024; 24:869-881. [PMID: 38252454 DOI: 10.1039/d3lc00745f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Cardiovascular toxicity causes adverse drug reactions and may lead to drug removal from the pharmaceutical market. Cancer therapies can induce life-threatening cardiovascular side effects such as arrhythmias, muscle cell death, or vascular dysfunction. New technologies have enabled cardiotoxic compounds to be identified earlier in drug development. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes (CMs) and vascular endothelial cells (ECs) can screen for drug-induced alterations in cardiovascular cell function and survival. However, most existing hiPSC models for cardiovascular drug toxicity utilize two-dimensional, immature cells grown in static culture. Improved in vitro models to mechanistically interrogate cardiotoxicity would utilize more adult-like, mature hiPSC-derived cells in an integrated system whereby toxic drugs and protective agents can flow between hiPSC-ECs that represent systemic vasculature and hiPSC-CMs that represent heart muscle (myocardium). Such models would be useful for testing the multi-lineage cardiotoxicities of chemotherapeutic drugs such as VEGFR2/PDGFR-inhibiting tyrosine kinase inhibitors (VPTKIs). Here, we develop a multi-lineage, fully-integrated, cardiovascular organ-chip that can enhance hiPSC-EC and hiPSC-CM functional and genetic maturity, model endothelial barrier permeability, and demonstrate long-term functional stability. This microfluidic organ-chip harbors hiPSC-CMs and hiPSC-ECs on separate channels that can be subjected to active fluid flow and rhythmic biomechanical stretch. We demonstrate the utility of this cardiovascular organ-chip as a predictive platform for evaluating multi-lineage VPTKI toxicity. This study may lead to the development of new modalities for the evaluation and prevention of cancer therapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Maedeh Mozneb
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Amelia Jenkins
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Stephany Pohlman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Michael J Workman
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Dylan West
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Briana Ondatje
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Kareem El-Ghazawi
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Amanda Woodbury
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Shachi Patel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Madelyn Arzt
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Felipe Dezem
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alex H Laperle
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - V Alexandra Moser
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Nur Yucer
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
| | - Jasmine Plummer
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Robert J Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Arun Sharma
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Pavilion, Room 8405, Los Angeles, CA 90048, USA.
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
10
|
Sang L, Zhou Z, Luo S, Zhang Y, Qian H, Zhou Y, He H, Hao K. An In Silico Platform to Predict Cardiotoxicity Risk of Anti-tumor Drug Combination with hiPSC-CMs Based In Vitro Study. Pharm Res 2024; 41:247-262. [PMID: 38148384 PMCID: PMC10879352 DOI: 10.1007/s11095-023-03644-4] [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: 07/18/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
OBJECTIVE Antineoplastic agent-induced systolic dysfunction is a major reason for interruption of anticancer treatment. Although targeted anticancer agents infrequently cause systolic dysfunction, their combinations with chemotherapies remarkably increase the incidence. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) provide a potent in vitro model to assess cardiovascular safety. However, quantitatively predicting the reduction of ejection fraction based on hiPSC-CMs is challenging due to the absence of the body's regulatory response to cardiomyocyte injury. METHODS Here, we developed and validated an in vitro-in vivo translational platform to assess the reduction of ejection fraction induced by antineoplastic drugs based on hiPSC-CMs. The translational platform integrates drug exposure, drug-cardiomyocyte interaction, and systemic response. The drug-cardiomyocyte interaction was implemented as a mechanism-based toxicodynamic (TD) model, which was then integrated into a quantitative system pharmacology-physiological-based pharmacokinetics (QSP-PBPK) model to form a complete translational platform. The platform was validated by comparing the model-predicted and clinically observed incidence of doxorubicin and trastuzumab-induced systolic dysfunction. RESULTS A total of 33,418 virtual patients were incorporated to receive doxorubicin and trastuzumab alone or in combination. For doxorubicin, the QSP-PBPK-TD model successfully captured the overall trend of systolic dysfunction incidences against the cumulative doses. For trastuzumab, the predicted incidence interval was 0.31-2.7% for single-agent treatment and 0.15-10% for trastuzumab-doxorubicin sequential treatment, covering the observations in clinical reports (0.50-1.0% and 1.5-8.3%, respectively). CONCLUSIONS In conclusion, the in vitro-in vivo translational platform is capable of predicting systolic dysfunction incidence almost merely depend on hiPSC-CMs, which could facilitate optimizing the treatment protocol of antineoplastic agents.
Collapse
Affiliation(s)
- Lan Sang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhengying Zhou
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Shizheng Luo
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yicui Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjie Qian
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kun Hao
- State Key Laboratory of Natural Medicines, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Tang X, Liu H, Rao R, Huang Y, Dong M, Xu M, Feng S, Shi X, Wang L, Wang Z, Zhou B. Modeling drug-induced mitochondrial toxicity with human primary cardiomyocytes. SCIENCE CHINA. LIFE SCIENCES 2024; 67:301-319. [PMID: 37864082 DOI: 10.1007/s11427-023-2369-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 05/16/2023] [Indexed: 10/22/2023]
Abstract
Mitochondrial toxicity induced by therapeutic drugs is a major contributor for cardiotoxicity, posing a serious threat to pharmaceutical industries and patients' lives. However, mitochondrial toxicity testing is not incorporated into routine cardiac safety screening procedures. To accurately model native human cardiomyocytes, we comprehensively evaluated mitochondrial responses of adult human primary cardiomyocytes (hPCMs) to a nucleoside analog, remdesivir (RDV). Comparison of their response to human pluripotent stem cell-derived cardiomyocytes revealed that the latter utilized a mitophagy-based mitochondrial recovery response that was absent in hPCMs. Accordingly, action potential duration was elongated in hPCMs, reflecting clinical incidences of RDV-induced QT prolongation. In a screen for mitochondrial protectants, we identified mitochondrial ROS as a primary mediator of RDV-induced cardiotoxicity. Our study demonstrates the utility of hPCMs in the detection of clinically relevant cardiac toxicities, and offers a framework for hPCM-based high-throughput screening of cardioprotective agents.
Collapse
Affiliation(s)
- Xiaoli Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Hong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Rongjia Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Mengqi Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Miaomiao Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Shanshan Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Xun Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China
| | - Zengwu Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
- Department of Epidemiology, Cardiovascular Institute and Fuwai Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China.
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, 518020, China.
| |
Collapse
|
12
|
Qu Y, Li T, Liu Z, Li D, Tong W. DICTrank: The largest reference list of 1318 human drugs ranked by risk of drug-induced cardiotoxicity using FDA labeling. Drug Discov Today 2023; 28:103770. [PMID: 37714406 DOI: 10.1016/j.drudis.2023.103770] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Drug-induced cardiotoxicity (DICT) is a leading cause of drug trial failure and discontinuation. Current drug annotations for cardiotoxicity largely focus on individual outcomes or mechanisms. Considering the broad spectrum of adverse cardiac events, we developed Drug-Induced Cardiotoxicity Rank (DICTrank) using FDA labeling and comprehensively classified 1318 human drugs into four categories: Most-DICT-Concern (n = 341), Less-DICT-Concern (n = 528), No-DICT-Concern (n = 343), and Ambiguous-DICT-Concern (n = 106). Notably, DICTrank covers diverse therapeutic categories, of which several were enriched with Most-DICT-Concern drugs, such as antineoplastic agents, sex hormones, anti-inflammatory drugs, beta-blockers, and cardiac therapy. DICTrank currently presents the largest drug list of DICT annotation, and it could contribute to the development of new approach methods, including AI models for early identification of DICT risk during drug development and beyond.
Collapse
Affiliation(s)
- Yanyan Qu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA; University of Arkansas at Little Rock and University of Arkansas for Medical Sciences Joint Bioinformatics Program, Little Rock, AR, USA
| | - Ting Li
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Zhichao Liu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA
| | - Dongying Li
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA.
| | - Weida Tong
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, USA.
| |
Collapse
|
13
|
Seal S, Spjuth O, Hosseini-Gerami L, García-Ortegón M, Singh S, Bender A, Carpenter AE. Insights into Drug Cardiotoxicity from Biological and Chemical Data: The First Public Classifiers for FDA DICTrank. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.15.562398. [PMID: 37905146 PMCID: PMC10614794 DOI: 10.1101/2023.10.15.562398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Drug-induced cardiotoxicity (DICT) is a major concern in drug development, accounting for 10-14% of postmarket withdrawals. In this study, we explored the capabilities of various chemical and biological data to predict cardiotoxicity, using the recently released Drug-Induced Cardiotoxicity Rank (DICTrank) dataset from the United States FDA. We analyzed a diverse set of data sources, including physicochemical properties, annotated mechanisms of action (MOA), Cell Painting, Gene Expression, and more, to identify indications of cardiotoxicity. We found that such data, including protein targets, especially those related to ion channels (such as hERG), physicochemical properties (such as electrotopological state) as well as peak concentration in plasma offer strong predictive ability as well as valuable insights into DICT. We also found compounds annotated with particular mechanisms of action, such as cyclooxygenase inhibition, could distinguish between most-concern and no-concern DICT compounds. Cell Painting features related to ER stress discern the most-concern cardiotoxic compounds from non-toxic compounds. While models based on physicochemical properties currently provide substantial predictive accuracy (AUCPR = 0.93), this study also underscores the potential benefits of incorporating more comprehensive biological data in future DICT predictive models. With the availability of - omics data in the future, using biological data promises enhanced predictability and delivers deeper mechanistic insights, paving the way for safer therapeutic drug development. All models and data used in this study are publicly released at https://broad.io/DICTrank_Predictor.
Collapse
Affiliation(s)
- Srijit Seal
- Imaging Platform, Broad Institute of MIT and Harvard, US
| | - Ola Spjuth
- Department of Pharmaceutical Biosciences, Uppsala University, Sweden
| | | | | | - Shantanu Singh
- Imaging Platform, Broad Institute of MIT and Harvard, US
| | | | | |
Collapse
|
14
|
Valentin JP, Sibony A, Rosseels ML, Delaunois A. "Appraisal of state-of-the-art" The 2021 Distinguished Service Award of the Safety Pharmacology Society: Reflecting on the past to tackle challenges ahead. J Pharmacol Toxicol Methods 2023; 123:107269. [PMID: 37149063 DOI: 10.1016/j.vascn.2023.107269] [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: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
This appraisal of state-of-the-art manuscript highlights and expands upon the thoughts conveyed in the lecture of Dr. Jean-Pierre Valentin, recipient of the 2021 Distinguished Service Award of the Safety Pharmacology Society, given on the 2nd December 2021. The article reflects on the strengths, weaknesses, opportunities, and threats that surrounded the evolution of safety and secondary pharmacology over the last 3 decades with a particular emphasis on pharmaceutical drug development delivery, scientific and technological innovation, complexities of regulatory framework and people leadership and development. The article further built on learnings from past experiences to tackle constantly emerging issues and evolving landscape whilst being cognizant of the challenges facing these disciplines in the broader drug development and societal context.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium.
| | - Alicia Sibony
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| | - Marie-Luce Rosseels
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| | - Annie Delaunois
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| |
Collapse
|
15
|
Parra-Lucares A, Villa E, Romero-Hernández E, Méndez-Valdés G, Retamal C, Vizcarra G, Henríquez I, Maldonado-Morales EAJ, Grant-Palza JH, Ruíz-Tagle S, Estrada-Bobadilla V, Toro L. Tic-Tac: A Translational Approach in Mechanisms Associated with Irregular Heartbeat and Sinus Rhythm Restoration in Atrial Fibrillation Patients. Int J Mol Sci 2023; 24:12859. [PMID: 37629037 PMCID: PMC10454641 DOI: 10.3390/ijms241612859] [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: 06/01/2023] [Revised: 08/08/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Atrial fibrillation (AF) is a prevalent cardiac condition predominantly affecting older adults, characterized by irregular heartbeat rhythm. The condition often leads to significant disability and increased mortality rates. Traditionally, two therapeutic strategies have been employed for its treatment: heart rate control and rhythm control. Recent clinical studies have emphasized the critical role of early restoration of sinus rhythm in improving patient outcomes. The persistence of the irregular rhythm allows for the progression and structural remodeling of the atria, eventually leading to irreversible stages, as observed clinically when AF becomes permanent. Cardioversion to sinus rhythm alters this progression pattern through mechanisms that are still being studied. In this review, we provide an in-depth analysis of the pathophysiological mechanisms responsible for maintaining AF and how they are modified during sinus rhythm restoration using existing therapeutic strategies at different stages of clinical investigation. Moreover, we explore potential future therapeutic approaches, including the promising prospect of gene therapy.
Collapse
Affiliation(s)
- Alfredo Parra-Lucares
- Critical Care Unit, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Cardiovascular Department, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Eduardo Villa
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Gabriel Méndez-Valdés
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Catalina Retamal
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Geovana Vizcarra
- Division of Internal Medicine, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
| | - Ignacio Henríquez
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Juan H. Grant-Palza
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | - Sofía Ruíz-Tagle
- School of Medicine, Faculty of Medicine, Universidad de Chile, Santiago 8380420, Chile
| | | | - Luis Toro
- Division of Nephrology, Department of Medicine, Hospital Clínico Universidad de Chile, Santiago 8380420, Chile
- Centro de Investigación Clínica Avanzada, Hospital Clínico, Universidad de Chile, Santiago 8380420, Chile
| |
Collapse
|
16
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the Morphological and Biological Functions of Vascularized Microphysiological Systems with Supervised Machine Learning. Ann Biomed Eng 2023; 51:1723-1737. [PMID: 36913087 DOI: 10.1007/s10439-023-03177-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function-oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
Affiliation(s)
- James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Christopher P Chaftari
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, 101 Bizzell Street, College Station, TX, 77843, USA.
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX, USA.
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, USA.
| |
Collapse
|
17
|
Vuorenpää H, Björninen M, Välimäki H, Ahola A, Kroon M, Honkamäki L, Koivumäki JT, Pekkanen-Mattila M. Building blocks of microphysiological system to model physiology and pathophysiology of human heart. Front Physiol 2023; 14:1213959. [PMID: 37485060 PMCID: PMC10358860 DOI: 10.3389/fphys.2023.1213959] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
Microphysiological systems (MPS) are drawing increasing interest from academia and from biomedical industry due to their improved capability to capture human physiology. MPS offer an advanced in vitro platform that can be used to study human organ and tissue level functions in health and in diseased states more accurately than traditional single cell cultures or even animal models. Key features in MPS include microenvironmental control and monitoring as well as high biological complexity of the target tissue. To reach these qualities, cross-disciplinary collaboration from multiple fields of science is required to build MPS. Here, we review different areas of expertise and describe essential building blocks of heart MPS including relevant cardiac cell types, supporting matrix, mechanical stimulation, functional measurements, and computational modelling. The review presents current methods in cardiac MPS and provides insights for future MPS development with improved recapitulation of human physiology.
Collapse
Affiliation(s)
- Hanna Vuorenpää
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Miina Björninen
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hannu Välimäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Micro- and Nanosystems Research Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Antti Ahola
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mart Kroon
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Biomaterials and Tissue Engineering Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Laura Honkamäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Neuro Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jussi T. Koivumäki
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Computational Biophysics and Imaging Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Mari Pekkanen-Mattila
- Centre of Excellence in Body-on-Chip Research (CoEBoC), BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Heart Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
18
|
Arefin A, Mendoza M, Dame K, Garcia MI, Strauss DG, Ribeiro AJS. Reproducibility of drug-induced effects on the contractility of an engineered heart tissue derived from human pluripotent stem cells. Front Pharmacol 2023; 14:1212092. [PMID: 37469866 PMCID: PMC10352809 DOI: 10.3389/fphar.2023.1212092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/14/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction: Engineered heart tissues (EHTs) are three-dimensional culture platforms with cardiomyocytes differentiated from human pluripotent stem cells (hPSCs) and were designed for assaying cardiac contractility. For drug development applications, EHTs must have a stable function and provide reproducible results. We investigated these properties with EHTs made with different tissue casting batches and lines of differentiated hPSC-cardiomyocytes and analyzed them at different times after being fabricated. Methods: A video-optical assay was used for measuring EHT contractile outputs, and these results were compared with results from motion traction analysis of beating hPSC-cardiomyocytes cultured as monolayers in two-dimensional cultures. The reproducibility of induced contractile variations was tested using compounds with known mechanistic cardiac effects (isoproterenol, EMD-57033, omecamtiv mecarbil, verapamil, ranolazine, and mavacamten), or known to be clinically cardiotoxic (doxorubicin, sunitinib). These drug-induced variations were characterized at different electrical pacing rates and variations in intracellular calcium transients were also assessed in EHTs. Results: To ensure reproducibility in experiments, we established EHT quality control criteria based on excitation-contraction coupling and contractile sensitivity to extracellular calcium concentration. In summary, a baseline contractile force of 0.2 mN and excitation-contraction coupling of EHTs were used as quality control criteria to select suitable EHTs for analysis. Overall, drug-induced contractile responses were similar between monolayers and EHTs, where a close relationship was observed between contractile output and calcium kinetics. Contractile variations at multiple time points after adding cardiotoxic compounds were also detectable in EHTs. Discussion: Reproducibility of drug-induced effects in EHTs between experiments and relative to published work on these cellular models was generally observed. Future applications for EHTs may require additional mechanistic criteria related to drug effects and cardiac functional outputs to be measured in regard to specific contexts of use.
Collapse
Affiliation(s)
- Ayesha Arefin
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, United States
| | - Melissa Mendoza
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - M. Iveth Garcia
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - David G. Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Alexandre J. S. Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
19
|
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: 3.0] [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
|
20
|
Mummery C. Getting to the heart of stem cell research: an interview with Christine Mummery. Dis Model Mech 2023; 16:dmm050270. [PMID: 37218469 PMCID: PMC10233713 DOI: 10.1242/dmm.050270] [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: 05/24/2023] Open
Abstract
Professor Christine Mummery has pioneered the use of pluripotent stem cell models to investigate heart development and disease, and pushed the boundaries of what can be achieved with these versatile cells. In 2008, she became Chair of Developmental Biology at Leiden University Medical Centre, where she has refined and advanced in vitro models of the heart and now harnesses their clinical potential to screen drugs and personalise treatments for patients with various types of heart disease. Christine has become integral to the stem cell community by promoting cross-disciplinary research and serving on several Ethical Councils, Scientific Advisory Boards and Editorial Boards. Her influence in the field of stem cell research led her to become the president of International Society for Stem Cell Research in 2020, and she has won numerous awards, including the Hans Bloemendal Medal 2014 for innovative interdisciplinary research, with Gordon Keller, the prestigious Lefoulon-Delalande Prize 2021 and the International Society for Stem Cell Research (ISSCR) Public Service Award 2023. In this interview, Christine shares her career trajectory, how disease modelling is shifting towards advanced in vitro systems and what challenges the field has yet to overcome.
Collapse
Affiliation(s)
- Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Centre, 2333 BE Leiden, The Netherlands
| |
Collapse
|
21
|
Rivera‐Arbeláez JM, Keekstra D, Cofiño‐Fabres C, Boonen T, Dostanic M, ten Den SA, Vermeul K, Mastrangeli M, van den Berg A, Segerink LI, Ribeiro MC, Strisciuglio N, Passier R. Automated assessment of human engineered heart tissues using deep learning and template matching for segmentation and tracking. Bioeng Transl Med 2023; 8:e10513. [PMID: 37206226 PMCID: PMC10189437 DOI: 10.1002/btm2.10513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 05/21/2023] Open
Abstract
The high rate of drug withdrawal from the market due to cardiovascular toxicity or lack of efficacy, the economic burden, and extremely long time before a compound reaches the market, have increased the relevance of human in vitro models like human (patient-derived) pluripotent stem cell (hPSC)-derived engineered heart tissues (EHTs) for the evaluation of the efficacy and toxicity of compounds at the early phase in the drug development pipeline. Consequently, the EHT contractile properties are highly relevant parameters for the analysis of cardiotoxicity, disease phenotype, and longitudinal measurements of cardiac function over time. In this study, we developed and validated the software HAARTA (Highly Accurate, Automatic and Robust Tracking Algorithm), which automatically analyzes contractile properties of EHTs by segmenting and tracking brightfield videos, using deep learning and template matching with sub-pixel precision. We demonstrate the robustness, accuracy, and computational efficiency of the software by comparing it to the state-of-the-art method (MUSCLEMOTION), and by testing it with a data set of EHTs from three different hPSC lines. HAARTA will facilitate standardized analysis of contractile properties of EHTs, which will be beneficial for in vitro drug screening and longitudinal measurements of cardiac function.
Collapse
Affiliation(s)
- José M. Rivera‐Arbeláez
- Department of Applied Stem Cell Technologies, TechMed CentreUniversity of TwenteEnschedethe Netherlands
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, TechMed Centre, Max Planck Institute for Complex Fluid DynamicsUniversity of TwenteEnschedethe Netherlands
| | - Danjel Keekstra
- Data Management & Biometrics (DMB) GroupUniversity of TwenteEnschedethe Netherlands
| | - Carla Cofiño‐Fabres
- Department of Applied Stem Cell Technologies, TechMed CentreUniversity of TwenteEnschedethe Netherlands
| | | | | | - Simone A. ten Den
- Department of Applied Stem Cell Technologies, TechMed CentreUniversity of TwenteEnschedethe Netherlands
| | - Kim Vermeul
- Department of Applied Stem Cell Technologies, TechMed CentreUniversity of TwenteEnschedethe Netherlands
| | | | - Albert van den Berg
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, TechMed Centre, Max Planck Institute for Complex Fluid DynamicsUniversity of TwenteEnschedethe Netherlands
| | - Loes I. Segerink
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, TechMed Centre, Max Planck Institute for Complex Fluid DynamicsUniversity of TwenteEnschedethe Netherlands
| | | | - Nicola Strisciuglio
- Data Management & Biometrics (DMB) GroupUniversity of TwenteEnschedethe Netherlands
| | - Robert Passier
- Department of Applied Stem Cell Technologies, TechMed CentreUniversity of TwenteEnschedethe Netherlands
- Department of Anatomy and EmbryologyLeiden University Medical CentreLeidenthe Netherlands
| |
Collapse
|
22
|
Tronolone JJ, Mathur T, Chaftari CP, Jain A. Evaluation of the morphological and biological functions of vascularized microphysiological systems with supervised machine learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523755. [PMID: 36711458 PMCID: PMC9882172 DOI: 10.1101/2023.01.12.523755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Vascularized microphysiological systems and organoids are contemporary preclinical experimental platforms representing human tissue or organ function in health and disease. While vascularization is emerging as a necessary physiological organ-level feature required in most such systems, there is no standard tool or morphological metric to measure the performance or biological function of vascularized networks within these models. Further, the commonly reported morphological metrics may not correlate to the network's biological function - oxygen transport. Here, a large library of vascular network images was analyzed by the measure of each sample's morphology and oxygen transport potential. The oxygen transport quantification is computationally expensive and user-dependent, so machine learning techniques were examined to generate regression models relating morphology to function. Principal component and factor analyses were applied to reduce dimensionality of the multivariate dataset, followed by multiple linear regression and tree-based regression analyses. These examinations reveal that while several morphological data relate poorly to the biological function, some machine learning models possess a relatively improved, but still moderate predictive potential. Overall, random forest regression model correlates to the biological function of vascular networks with relatively higher accuracy than other regression models.
Collapse
|
23
|
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: 12.0] [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
|
24
|
Zhou B, Shi X, Tang X, Zhao Q, Wang L, Yao F, Hou Y, Wang X, Feng W, Wang L, Sun X, Wang L, Hu S. Functional isolation, culture and cryopreservation of adult human primary cardiomyocytes. Signal Transduct Target Ther 2022; 7:254. [PMID: 35882831 PMCID: PMC9325714 DOI: 10.1038/s41392-022-01044-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 04/11/2022] [Accepted: 05/26/2022] [Indexed: 12/27/2022] Open
Abstract
Cardiovascular diseases are the most common cause of death globally. Accurately modeling cardiac homeostasis, dysfunction, and drug response lies at the heart of cardiac research. Adult human primary cardiomyocytes (hPCMs) are a promising cellular model, but unstable isolation efficiency and quality, rapid cell death in culture, and unknown response to cryopreservation prevent them from becoming a reliable and flexible in vitro cardiac model. Combing the use of a reversible inhibitor of myosin II ATPase, (-)-blebbistatin (Bleb), and multiple optimization steps of the isolation procedure, we achieved a 2.74-fold increase in cell viability over traditional methods, accompanied by better cellular morphology, minimally perturbed gene expression, intact electrophysiology, and normal neurohormonal signaling. Further optimization of culture conditions established a method that was capable of maintaining optimal cell viability, morphology, and mitochondrial respiration for at least 7 days. Most importantly, we successfully cryopreserved hPCMs, which were structurally, molecularly, and functionally intact after undergoing the freeze-thaw cycle. hPCMs demonstrated greater sensitivity towards a set of cardiotoxic drugs, compared to human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Further dissection of cardiomyocyte drug response at both the population and single-cell transcriptomic level revealed that hPCM responses were more pronouncedly enriched in cardiac function, whereas hiPSC-CMs responses reflected cardiac development. Together, we established a full set of methodologies for the efficient isolation and prolonged maintenance of functional primary adult human cardiomyocytes in vitro, unlocking their potential as a cellular model for cardiovascular research, drug discovery, and safety pharmacology.
Collapse
Affiliation(s)
- Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| | - Xun Shi
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoli Tang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Quanyi Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| | - Le Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongfeng Hou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,18 Jinma Industrial Park, Fangshan District, Beijing, China
| | - Xianqiang Wang
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Feng
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liqing Wang
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaogang Sun
- Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China
| | - Shengshou Hu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China. .,Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, Shenzhen, China. .,Department of Cardiac Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
25
|
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: 1.5] [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
|
26
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
27
|
Huang CY, Nicholson MW, Wang JY, Ting CY, Tsai MH, Cheng YC, Liu CL, Chan DZH, Lee YC, Hsu CC, Hsu YH, Yang CF, Chang CMC, Ruan SC, Lin PJ, Lin JH, Chen LL, Hsieh ML, Cheng YY, Hsu WT, Lin YL, Chen CH, Hsu YH, Wu YT, Hacker TA, Wu JC, Kamp TJ, Hsieh PCH. Population-based high-throughput toxicity screen of human iPSC-derived cardiomyocytes and neurons. Cell Rep 2022; 39:110643. [PMID: 35385754 DOI: 10.1016/j.celrep.2022.110643] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 01/13/2022] [Accepted: 03/16/2022] [Indexed: 12/21/2022] Open
Abstract
In this study, we establish a population-based human induced pluripotent stem cell (hiPSC) drug screening platform for toxicity assessment. After recruiting 1,000 healthy donors and screening for high-frequency human leukocyte antigen (HLA) haplotypes, we identify 13 HLA-homozygous "super donors" to represent the population. These "super donors" are also expected to represent at least 477,611,135 of the global population. By differentiating these representative hiPSCs into cardiomyocytes and neurons we show their utility in a high-throughput toxicity screen. To validate hit compounds, we demonstrate dose-dependent toxicity of the hit compounds and assess functional modulation. We also show reproducible in vivo drug toxicity results using mouse models with select hit compounds. This study shows the feasibility of using a population-based hiPSC drug screening platform to assess cytotoxicity, which can be used as an innovative tool to study inter-population differences in drug toxicity and adverse drug reactions in drug discovery applications.
Collapse
Affiliation(s)
- Ching Ying Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | | | - Jyun Yuan Wang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Chien Yu Ting
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Ming Heng Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yu Che Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chun Lin Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Darien Z H Chan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yi Chan Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Ching Chuan Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yu Hung Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chiou Fong Yang
- Institute of Applied Mechanics, National Taiwan University, Taipei 106, Taiwan
| | - Cindy M C Chang
- Cardiovascular Physiology Core Facility, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Shu Chian Ruan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Po Ju Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Jen Hao Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Li Lun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Marvin L Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Cardiovascular Physiology Core Facility, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yuan Yuan Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wan Tseng Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yi Ling Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Chien Hsiun Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Yu Hsiang Hsu
- Institute of Applied Mechanics, National Taiwan University, Taipei 106, Taiwan
| | - Ying Ta Wu
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Timothy A Hacker
- Cardiovascular Physiology Core Facility, Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Timothy J Kamp
- Department of Medicine and Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Patrick C H Hsieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan; Department of Medicine and Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, WI 53705, USA; Institute of Medical Genomics and Proteomics and Institute of Clinical Medicine, National Taiwan University, Taipei 106, Taiwan.
| |
Collapse
|
28
|
Cooper BL, Posnack NG. Characteristics of Bisphenol Cardiotoxicity: Impaired Excitability, Contractility, and Relaxation. Cardiovasc Toxicol 2022; 22:273-280. [PMID: 35143014 PMCID: PMC9204785 DOI: 10.1007/s12012-022-09719-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
Bisphenol a (BPA) is a high production volume chemical that is frequently used to manufacture epoxy resins and polycarbonate plastics. BPA-containing products are now pervasive, and as a result, biomonitoring studies report widespread exposure in > 90% of adults, adolescents, and children. Both epidemiological and experimental studies have reported associations between BPA exposure and adverse cardiovascular health outcomes. With increasing concerns regarding BPA exposure, a few structurally similar bisphenol chemicals have been introduced as replacements, including bisphenol s (BPS) and bisphenol f (BPF). In accordance with the recently established "Key characteristics of cardiovascular toxicants", we reviewed the literature to highlight the immediate effects of bisphenol chemicals on (1) cardiac excitability, and (2) contractility and relaxation. BPA inhibits key cardiac ion channels, impairs cardiac excitability, and acts as a more potent inhibitor as compared to BPF and BPS. Through the inhibition of calcium current, some studies report that bisphenol chemicals can act as negative inotropic agents. Yet, others suggest that low dose exposures may increase contractility and precipitate triggered arrhythmias via the phosphorylation of key calcium handling proteins. Accordingly, we propose additional considerations for future work to comprehensively address the cardiac safety profile of BPA, as compared to replacement chemicals.
Collapse
Affiliation(s)
- Blake L. Cooper
- Children’s National Heart Institute, Children’s National Hospital, Washington, DC 20010, USA,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Hospital, Washington, DC 20010, USA,Department of Pharmacology & Physiology, George Washington University, Washington, DC 20037, USA
| | - Nikki Gillum Posnack
- Children's National Heart Institute, Children's National Hospital, Washington, DC, 20010, USA. .,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Hospital, Washington, DC, 20010, USA. .,Department of Pediatrics, George Washington University, Washington, DC, 20037, USA. .,Department of Pharmacology & Physiology, George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
29
|
Simms L, Yu F, Palmer J, Rudd K, Sticken ET, Wieczorek R, Chapman F, Czekala L, Stevenson M, O’Connell G. Use of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes to Predict the Cardiotoxicity Potential of Next Generation Nicotine Products. FRONTIERS IN TOXICOLOGY 2022; 4:747508. [PMID: 35295225 PMCID: PMC8915889 DOI: 10.3389/ftox.2022.747508] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/20/2022] [Indexed: 12/20/2022] Open
Abstract
Combustible cigarette smoking is an established risk factor for cardiovascular disease. By contrast, the cardiotoxicity potential of non-combustible next generation nicotine products (NGPs), which includes heated tobacco products (HTPs) and electronic vaping products (EVPs), and how this compares relative to combustible cigarettes is currently an area of scientific exploration. As such, there is a need for a rapid screening assay to assess this endpoint. The Cardio quickPredict is a metabolomics biomarker-based assay that uses human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) to screen for potential structural and functional cardiac toxicants based on the changes of four metabolites, lactic acid, arachidonic acid, thymidine, and 2'-deoxycytidine. The study aims were to investigate the cardiotoxicity potential of NGPs compared to cigarettes, in addition to nicotine. To accomplish this, hiPSC-CM were exposed to smoke or aerosol bubbled PBS samples: reference cigarette (1R6F); three variants of HTP; and three EVP variants. The 1R6F bPBS was the most active, having cardiotoxic potential at 0.3-0.6% bPBS (0.4-0.9 μg/mL nicotine), followed by HTP, which displayed cardiotoxic potential at a 10 times higher concentration, 3.3% bPBS (4.1 μg/mL nicotine). Both 1R6F and HTP bPBS (at 10-fold higher concentration than 1R6F) affected all four predictive metabolites, whereas none of the EVP bPBS samples were active in the assay up to the maximal concentration tested (10% bPBS). Nicotine tested on its own was predicted to have cardiotoxic potential at concentrations greater than 80 μg/mL, which is higher than expected physiological levels associated with combustible cigarette smoking. The application of this rapid screening assay to NGP research and the associated findings adds to the weight-of-evidence indicating that NGPs have a tobacco harm reduction potential when compared to combustible cigarettes. Additionally, this technique was shown to be sensitive and robust for the assessment of different NGPs and may be considered as part of a larger overall scientific framework for NGP assessments.
Collapse
Affiliation(s)
- Liam Simms
- Imperial Brands PLC, Bristol, United Kingdom
| | - Fan Yu
- Imperial Brands PLC, Bristol, United Kingdom
| | - Jessica Palmer
- Stemina Biomarker Discovery Inc., Madison, WI, United States
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Soepriatna AH, Kim TY, Daley MC, Song E, Choi BR, Coulombe KLK. Human Atrial Cardiac Microtissues for Chamber-Specific Arrhythmic Risk Assessment. Cell Mol Bioeng 2021; 14:441-457. [PMID: 34777603 DOI: 10.1007/s12195-021-00703-x] [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: 02/22/2021] [Accepted: 09/02/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction Although atrial fibrillation is the most prevalent disorder of electrical conduction, the mechanisms behind atrial arrhythmias remain elusive. To address this challenge, we developed a robust in vitro model of 3D atrial microtissue from human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and evaluated chamber-specific chemical responses experimentally and computationally. Methods We differentiated atrial and ventricular cardiomyocytes (aCMs/vCMs) from GCaMP6f-expressing hiPSCs and assessed spontaneous AP activity using fluorescence imaging. Self-assembling 3D microtissues were formed with lactate purified CMs and 5% human cardiac fibroblasts and electrically stimulated for one week before high resolution action potential (AP) optical mapping. AP responses to the atrial-specific potassium repolarizing current I Kur-blocker 4-Aminopyridine (4-AP) and funny current I f-blocker Ivabradine were characterized within their therapeutic window. Finally, we expanded upon a published hiPSC-CM computational model by incorporating the atrial-specific I Kur current, modifying ion channel conductances to match the AP waveforms of our microtissues, and employing the updated model to reinforce our experimental findings. Results High purity CMs (> 75% cTnT+) demonstrated subtype specification by MLC2v expression. Spontaneous beating rates significantly decreased following 3D microtissue formation, with atrial microtissues characterized by their faster spontaneous beating rate, slower AP rise time, and shorter AP duration (APD) compared to ventricular microtissues. We measured atrial-specific responses, including dose-dependent APD prolongation with 4-AP treatment and dose-dependent reduction in spontaneous activity post-Ivabradine treatment. Conclusion The presented in vitro platform for screening atrial-specific responses is both robust and sensitive, with high throughput, enabling studies focused at elucidating the mechanisms underlying atrial arrhythmias. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00703-x.
Collapse
Affiliation(s)
- Arvin H Soepriatna
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Tae Yun Kim
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI USA
| | - Mark C Daley
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Elena Song
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| | - Bum-Rak Choi
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI USA
| | - Kareen L K Coulombe
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI USA
| |
Collapse
|
31
|
Chen L, He Y, Wang X, Ge J, Li H. Ventricular voltage-gated ion channels: Detection, characteristics, mechanisms, and drug safety evaluation. Clin Transl Med 2021; 11:e530. [PMID: 34709746 PMCID: PMC8516344 DOI: 10.1002/ctm2.530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiac voltage-gated ion channels (VGICs) play critical roles in mediating cardiac electrophysiological signals, such as action potentials, to maintain normal heart excitability and contraction. Inherited or acquired alterations in the structure, expression, or function of VGICs, as well as VGIC-related side effects of pharmaceutical drug delivery can result in abnormal cellular electrophysiological processes that induce life-threatening cardiac arrhythmias or even sudden cardiac death. Hence, to reduce possible heart-related risks, VGICs must be acknowledged as important targets in drug discovery and safety studies related to cardiac disease. In this review, we first summarize the development and application of electrophysiological techniques that are employed in cardiac VGIC studies alone or in combination with other techniques such as cryoelectron microscopy, optical imaging and optogenetics. Subsequently, we describe the characteristics, structure, mechanisms, and functions of various well-studied VGICs in ventricular myocytes and analyze their roles in and contributions to both physiological cardiac excitability and inherited cardiac diseases. Finally, we address the implications of the structure and function of ventricular VGICs for drug safety evaluation. In summary, multidisciplinary studies on VGICs help researchers discover potential targets of VGICs and novel VGICs in heart, enrich their knowledge of the properties and functions, determine the operation mechanisms of pathological VGICs, and introduce groundbreaking trends in drug therapy strategies, and drug safety evaluation.
Collapse
Affiliation(s)
- Lulan Chen
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yue He
- Department of CardiologyShanghai Xuhui District Central Hospital & Zhongshan‐xuhui HospitalShanghaiChina
| | - Xiangdong Wang
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Junbo Ge
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| | - Hua Li
- Department of Cardiology, Shanghai Institute of Cardiovascular DiseasesShanghai Xuhui District Central Hospital & Zhongshan‐xuhui Hospital, Zhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
32
|
Burnett SD, Blanchette AD, Chiu WA, Rusyn I. Cardiotoxicity Hazard and Risk Characterization of ToxCast Chemicals Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes from Multiple Donors. Chem Res Toxicol 2021; 34:2110-2124. [PMID: 34448577 PMCID: PMC8762671 DOI: 10.1021/acs.chemrestox.1c00203] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Heart disease remains a significant human health burden worldwide with a significant fraction of morbidity attributable to environmental exposures. However, the extent to which the thousands of chemicals in commerce and the environment may contribute to heart disease morbidity is largely unknown, because in contrast to pharmaceuticals, environmental chemicals are seldom tested for potential cardiotoxicity. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes have become an informative in vitro model for cardiotoxicity testing of drugs with the availability of cells from multiple individuals allowing in vitro testing of population variability. In this study, we hypothesized that a panel of iPSC-derived cardiomyocytes from healthy human donors can be used to screen for the potential cardiotoxicity hazard and risk of environmental chemicals. We conducted concentration-response testing of 1029 chemicals (drugs, pesticides, flame retardants, polycyclic aromatic hydrocarbons (PAHs), plasticizers, industrial chemicals, food/flavor/fragrance agents, etc.) in iPSC-derived cardiomyocytes from 5 donors. We used kinetic calcium flux and high-content imaging to derive quantitative measures as inputs into Bayesian population concentration-response modeling of the effects of each chemical. We found that many environmental chemicals pose a hazard to human cardiomyocytes in vitro with more than half of all chemicals eliciting positive or negative chronotropic or arrhythmogenic effects. However, most of the tested environmental chemicals for which human exposure and high-throughput toxicokinetics data were available had wide margins of exposure and, thus, do not appear to pose a significant human health risk in a general population. Still, relatively narrow margins of exposure (<100) were estimated for some perfuoroalkyl substances and phthalates, raising concerns that cumulative exposures may pose a cardiotoxicity risk. Collectively, this study demonstrated the value of using a population-based human in vitro model for rapid, high-throughput hazard and risk characterization of chemicals for which little to no cardiotoxicity data are available from guideline studies in animals.
Collapse
Affiliation(s)
- Sarah D. Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Alexander D. Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Weihsueh A. Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843-4458, USA
| |
Collapse
|
33
|
Comparison of 10 Control hPSC Lines for Drug Screening in an Engineered Heart Tissue Format. Stem Cell Reports 2021; 15:983-998. [PMID: 33053362 PMCID: PMC7561618 DOI: 10.1016/j.stemcr.2020.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/19/2022] Open
Abstract
Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) are commercially available, and cardiac differentiation established routine. Systematic evaluation of several control hiPSC-CM is lacking. We investigated 10 different control hiPSC-CM lines and analyzed function and suitability for drug screening. Five commercial and 5 academic hPSC-CM lines were casted in engineered heart tissue (EHT) format. Spontaneous and stimulated EHT contractions were analyzed, and 7 inotropic indicator compounds investigated on 8 cell lines. Baseline contractile force, kinetics, and rate varied widely among the different lines (e.g., relaxation time range: 118-471 ms). In contrast, the qualitative correctness of responses to BayK-8644, nifedipine, EMD-57033, isoprenaline, and digoxin in terms of force and kinetics varied only between 80% and 93%. Large baseline differences between control cell lines support the request for isogenic controls in disease modeling. Variability appears less relevant for drug screening but needs to be considered, arguing for studies with more than one line.
Collapse
|
34
|
Ton AT, Nguyen W, Sweat K, Miron Y, Hernandez E, Wong T, Geft V, Macias A, Espinoza A, Truong K, Rasoul L, Stafford A, Cotta T, Mai C, Indersmitten T, Page G, Miller PE, Ghetti A, Abi-Gerges N. Arrhythmogenic and antiarrhythmic actions of late sustained sodium current in the adult human heart. Sci Rep 2021; 11:12014. [PMID: 34103608 PMCID: PMC8187365 DOI: 10.1038/s41598-021-91528-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022] Open
Abstract
Late sodium current (late INa) inhibition has been proposed to suppress the incidence of arrhythmias generated by pathological states or induced by drugs. However, the role of late INa in the human heart is still poorly understood. We therefore investigated the role of this conductance in arrhythmias using adult primary cardiomyocytes and tissues from donor hearts. Potentiation of late INa with ATX-II (anemonia sulcata toxin II) and E-4031 (selective blocker of the hERG channel) slowed the kinetics of action potential repolarization, impaired Ca2+ homeostasis, increased contractility, and increased the manifestation of arrhythmia markers. These effects could be reversed by late INa inhibitors, ranolazine and GS-967. We also report that atrial tissues from donor hearts affected by atrial fibrillation exhibit arrhythmia markers in the absence of drug treatment and inhibition of late INa with GS-967 leads to a significant reduction in arrhythmic behaviour. These findings reveal a critical role for the late INa in cardiac arrhythmias and suggest that inhibition of this conductance could provide an effective therapeutic strategy. Finally, this study highlights the utility of human ex-vivo heart models for advancing cardiac translational sciences.
Collapse
Affiliation(s)
- Anh Tuan Ton
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - William Nguyen
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Katrina Sweat
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Yannick Miron
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Eduardo Hernandez
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tiara Wong
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Valentyna Geft
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Andrew Macias
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Ana Espinoza
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Ky Truong
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Lana Rasoul
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Alexa Stafford
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tamara Cotta
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Christina Mai
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tim Indersmitten
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Guy Page
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA.
| |
Collapse
|
35
|
Gnecchi M, Sala L, Schwartz PJ. Precision Medicine and cardiac channelopathies: when dreams meet reality. Eur Heart J 2021; 42:1661-1675. [PMID: 33686390 PMCID: PMC8088342 DOI: 10.1093/eurheartj/ehab007] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/10/2020] [Accepted: 01/12/2021] [Indexed: 12/17/2022] Open
Abstract
Precision Medicine (PM) is an innovative approach that, by relying on large populations’ datasets, patients’ genetics and characteristics, and advanced technologies, aims at improving risk stratification and at identifying patient-specific management through targeted diagnostic and therapeutic strategies. Cardiac channelopathies are being progressively involved in the evolution brought by PM and some of them are benefiting from these novel approaches, especially the long QT syndrome. Here, we have explored the main layers that should be considered when developing a PM approach for cardiac channelopathies, with a focus on modern in vitro strategies based on patient-specific human-induced pluripotent stem cells and on in silico models. PM is where scientists and clinicians must meet and integrate their expertise to improve medical care in an innovative way but without losing common sense. We have indeed tried to provide the cardiologist’s point of view by comparing state-of-the-art techniques and approaches, including revolutionary discoveries, to current practice. This point matters because the new approaches may, or may not, exceed the efficacy and safety of established therapies. Thus, our own eagerness to implement the most recent translational strategies for cardiac channelopathies must be tempered by an objective assessment to verify whether the PM approaches are indeed making a difference for the patients. We believe that PM may shape the diagnosis and treatment of cardiac channelopathies for years to come. Nonetheless, its potential superiority over standard therapies should be constantly monitored and assessed before translating intellectually rewarding new discoveries into clinical practice.
Collapse
Affiliation(s)
- Massimiliano Gnecchi
- Department of Cardiothoracic and Vascular Sciences-Coronary Care Unit and Laboratory of Clinical and Experimental Cardiology, Fondazione IRCCS Policlinico San Matteo, Viale Golgi 19, 27100 Pavia, Italy.,Department of Molecular Medicine, Unit of Cardiology, University of Pavia, Viale Golgi 19, 27100 Pavia, Italy.,Department of Medicine, University of Cape Town, J-Floor, Old Main Building, Groote Schuur Hospital, Observatory, 7925 Cape Town, South Africa
| | - Luca Sala
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Via Pier Lombardo 22 - 20135 Milan, Italy
| | - Peter J Schwartz
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Via Pier Lombardo 22 - 20135 Milan, Italy
| |
Collapse
|
36
|
Jordaan P, Dumotier B, Traebert M, Miller PE, Ghetti A, Urban L, Abi-Gerges N. Cardiotoxic Potential of Hydroxychloroquine, Chloroquine and Azithromycin in Adult Human Primary Cardiomyocytes. Toxicol Sci 2021; 180:356-368. [PMID: 33483756 PMCID: PMC7928616 DOI: 10.1093/toxsci/kfaa194] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Substantial efforts have been recently committed to develop coronavirus disease-2019 (COVID-19) medications, and Hydroxychloroquine alone or in combination with Azithromycin has been promoted as a repurposed treatment. Although these drugs may increase cardiac toxicity risk, cardiomyocyte mechanisms underlying this risk remain poorly understood in humans. Therefore, we evaluated the proarrhythmia risk and inotropic effects of these drugs in the cardiomyocyte contractility-based model of the human heart. We found Hydroxychloroquine to have a low proarrhythmia risk, whereas Chloroquine and Azithromycin were associated with high risk. Hydroxychloroquine proarrhythmia risk changed to high with low level of K+, whereas high level of Mg2+ protected against proarrhythmic effect of high Hydroxychloroquine concentrations. Moreover, therapeutic concentration of Hydroxychloroquine caused no enhancement of elevated temperature-induced proarrhythmia. Polytherapy of Hydroxychloroquine plus Azithromycin and sequential application of these drugs were also found to influence proarrhythmia risk categorization. Hydroxychloroquine proarrhythmia risk changed to high when combined with Azithromycin at therapeutic concentration. However, Hydroxychloroquine at therapeutic concentration impacted the cardiac safety profile of Azithromycin and its proarrhythmia risk only at concentrations above therapeutic level. We also report that Hydroxychloroquine and Chloroquine, but not Azithromycin, decreased contractility while exhibiting multi-ion channel block features, and Hydroxychloroquine's contractility effect was abolished by Azithromycin. Thus, this study has the potential to inform clinical studies evaluating repurposed therapies, including those in the COVID-19 context. Additionally, it demonstrates the translational value of the human cardiomyocyte contractility-based model as a key early discovery path to inform decisions on novel therapies for COVID-19, malaria, and inflammatory diseases.
Collapse
Affiliation(s)
- Pierre Jordaan
- Chief Medical Officer and Patient Safety, Novartis AG, Basel, Switzerland
| | - Bérengère Dumotier
- Novartis Institutes for Biomedical Research, Preclinical Safety, Basel, Switzerland
| | - Martin Traebert
- Novartis Institutes for Biomedical Research, Preclinical Safety, Basel, Switzerland
| | - Paul E Miller
- AnaBios Corporation, San Diego, California 92109, USA
| | - Andre Ghetti
- AnaBios Corporation, San Diego, California 92109, USA
| | - Laszlo Urban
- Novartis Institutes for Biomedical Research, Preclinical Secondary Pharmacology, Cambridge, Massachusetts, USA
| | | |
Collapse
|
37
|
Bozza WP, Takeda K, Alterovitz WL, Chou CK, Shen RF, Zhang B. Anthracycline-Induced Cardiotoxicity: Molecular Insights Obtained from Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs). AAPS JOURNAL 2021; 23:44. [PMID: 33719006 PMCID: PMC7956936 DOI: 10.1208/s12248-021-00576-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/24/2021] [Indexed: 01/18/2023]
Abstract
Anthracyclines are a class of chemotherapy drugs that are highly effective for the treatment of human cancers, but their clinical use is limited by associated dose-dependent cardiotoxicity. The precise mechanisms by which individual anthracycline induces cardiotoxicity are not fully understood. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are emerging as a physiologically relevant model to assess drugs cardiotoxicity. Here, we describe an assay platform by coupling hiPSC-CMs and impedance measurement, which allows real-time monitoring of cardiomyocyte cellular index, beating amplitude, and beating rate. Using this approach, we have performed comparative studies on a panel of four anthracycline drugs (doxorubicin, epirubicin, idarubicin, and daunorubicin) which share a high degree of structural similarity but are associated with distinct cardiotoxicity profiles and maximum cumulative dose limits. Notably, results from our hiPSC-CMs impedance model (dose-dependent responses and EC50 values) agree well with the recommended clinical dose limits for these drugs. Using time-lapse imaging and RNAseq, we found that the differences in anthracycline cardiotoxicity are closely linked to extent of cardiomyocyte uptake and magnitude of activation/inhibition of several cellular pathways such as death receptor signaling, ROS production, and dysregulation of calcium signaling. The results provide molecular insights into anthracycline cardiac interactions and offer a novel assay system to more robustly assess potential cardiotoxicity during drug development.
Collapse
Affiliation(s)
- William P Bozza
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Kazuyo Takeda
- Microscopy and Imaging Core Facility, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Wei-Lun Alterovitz
- High-performance Integrated Virtual Environment, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Chao-Kai Chou
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Rong-Fong Shen
- Facility for Biotechnology Resources, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Baolin Zhang
- Office of Biotechnology Products, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| |
Collapse
|
38
|
Tu C, Cunningham NJ, Zhang M, Wu JC. Human Induced Pluripotent Stem Cells as a Screening Platform for Drug-Induced Vascular Toxicity. Front Pharmacol 2021; 12:613837. [PMID: 33790786 PMCID: PMC8006367 DOI: 10.3389/fphar.2021.613837] [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: 10/03/2020] [Accepted: 01/22/2021] [Indexed: 01/02/2023] Open
Abstract
Evaluation of potential vascular injury is an essential part of the safety study during pharmaceutical development. Vascular liability issues are important causes of drug termination during preclinical investigations. Currently, preclinical assessment of vascular toxicity primarily relies on the use of animal models. However, accumulating evidence indicates a significant discrepancy between animal toxicity and human toxicity, casting doubt on the clinical relevance of animal models for such safety studies. While the causes of this discrepancy are expected to be multifactorial, species differences are likely a key factor. Consequently, a human-based model is a desirable solution to this problem, which has been made possible by the advent of human induced pluripotent stem cells (iPSCs). In particular, recent advances in the field now allow the efficient generation of a variety of vascular cells (e.g., endothelial cells, smooth muscle cells, and pericytes) from iPSCs. Using these cells, different vascular models have been established, ranging from simple 2D cultures to highly sophisticated vascular organoids and microfluidic devices. Toxicity testing using these models can recapitulate key aspects of vascular pathology on molecular (e.g., secretion of proinflammatory cytokines), cellular (e.g., cell apoptosis), and in some cases, tissue (e.g., endothelium barrier dysfunction) levels. These encouraging data provide the rationale for continuing efforts in the exploration, optimization, and validation of the iPSC technology in vascular toxicology.
Collapse
Affiliation(s)
- Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Nathan J Cunningham
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Mao Zhang
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States.,Department of Medicine, Stanford University, Stanford, CA, United States.,Department of Radiology, Stanford University, Stanford, CA, United States
| |
Collapse
|
39
|
Burnett SD, Blanchette AD, Chiu WA, Rusyn I. Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes as an in vitro model in toxicology: strengths and weaknesses for hazard identification and risk characterization. Expert Opin Drug Metab Toxicol 2021; 17:887-902. [PMID: 33612039 DOI: 10.1080/17425255.2021.1894122] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Human induced pluripotent stem cell (iPSC)-derived cardiomyocytes is one of the most widely used cell-based models that resulted from the discovery of how non-embryonic stem cells can be differentiated into multiple cell types. In just one decade, iPSC-derived cardiomyocytes went from a research lab to widespread use in biomedical research and preclinical safety evaluation for drugs and other chemicals. AREAS COVERED This manuscript reviews data on toxicology applications of human iPSC-derived cardiomyocytes. We detail the outcome of a systematic literature search on their use (i) in hazard assessment for cardiotoxicity liabilities, (ii) for risk characterization, (iii) as models for population variability, and (iv) in studies of personalized medicine and disease. EXPERT OPINION iPSC-derived cardiomyocytes are useful to increase the accuracy, precision, and efficiency of cardiotoxicity hazard identification for both drugs and non-pharmaceuticals, with recent efforts beginning to demonstrate their utility for risk characterization. Notable limitations include the needs to improve the maturation of cells in culture, to better understand their potential use identifying structural cardiotoxicity, and for additional case studies involving population-wide and disease-specific risk characterization. Ultimately, the greatest future benefits are likely for non-pharmaceutical chemicals, filling a critical gap where no routine testing for cardiotoxicity is currently performed.
Collapse
Affiliation(s)
- Sarah D Burnett
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Alexander D Blanchette
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Weihsueh A Chiu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
40
|
Choi DK, Kim YK, HoonYu J, Min SH, Park SW. Genome editing of hPSCs: Recent progress in hPSC-based disease modeling for understanding disease mechanisms. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 181:271-287. [PMID: 34127196 DOI: 10.1016/bs.pmbts.2021.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Generation of proper models for studying human genetic diseases has been hindered until recently by the scarcity of primary cell samples from genetic disease patients and inefficient genetic modification tools. However, recent advances in clustered, regularly interspaced short palindromic repeats (CRISPR)/Cas9 technology and human induced pluripotent stem cells (hiPSCs) have provided an opportunity to explore the function of pathogenic variants and obtain gene-corrected cells for autologous cell therapy. In this chapter, we address recent applications of CRISPR/Cas9 to hiPSCs in genetic diseases, including neurodegenerative, cardiovascular, and rare diseases.
Collapse
Affiliation(s)
- Dong-Kyu Choi
- New Drug Development Center, DGMIF, Daegu, Republic of Korea
| | - Yong-Kyu Kim
- New Drug Development Center, DGMIF, Daegu, Republic of Korea
| | - Ji HoonYu
- New Drug Development Center, DGMIF, Daegu, Republic of Korea
| | - Sang-Hyun Min
- New Drug Development Center, DGMIF, Daegu, Republic of Korea
| | - Sang-Wook Park
- Department of Oral Biochemistry, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
41
|
Berg EL. The future of phenotypic drug discovery. Cell Chem Biol 2021; 28:424-430. [PMID: 33529582 DOI: 10.1016/j.chembiol.2021.01.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/30/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Phenotypic drug discovery (PDD) uses biological systems directly for new drug screening. While PDD has proved effective in the discovery of drugs with novel mechanisms, for broader adoption, key challenges need resolution: progression of poorly qualified leads and overloaded pipelines due to lack of effective tools to process and prioritize hits; and advancement of leads with undesirable mechanisms that fail at more expensive stages of discovery. Here I discuss how human-based phenotypic platforms are being applied throughout the discovery process for hit triage and prioritization, for elimination of hits with unsuitable mechanisms, and for supporting clinical strategies through pathway-based decision frameworks. Harnessing the data generated in these platforms can also fuel a deeper understanding of drug efficacy and toxicity mechanisms. As these approaches increase in use, they will gain in power for driving better decisions, generating better leads faster and in turn promoting greater adoption of PDD.
Collapse
|
42
|
Mohamed TMA, Conklin DJ. Emerging technology and platforms for cardiotoxicity testing. Toxicol Appl Pharmacol 2020; 408:115262. [PMID: 33017593 PMCID: PMC7747951 DOI: 10.1016/j.taap.2020.115262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Tamer M A Mohamed
- Institute of Molecular Cardiology, University of Louisville, Louisville, KY 40292, United States; Diabetes and Obesity Center, University of Louisville, Louisville, KY 40292, United States; Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40292, United States; Department of Medicine, University of Louisville, Louisville, KY 40292, United States
| | - Daniel J Conklin
- Diabetes and Obesity Center, University of Louisville, Louisville, KY 40292, United States; Christina Lee Brown Envirome Institute, University of Louisville, Louisville, KY 40292, United States; Department of Medicine, University of Louisville, Louisville, KY 40292, United States.
| |
Collapse
|
43
|
Pang L, Liu Z, Wei F, Cai C, Yang X. Improving cardiotoxicity prediction in cancer treatment: integration of conventional circulating biomarkers and novel exploratory tools. Arch Toxicol 2020; 95:791-805. [PMID: 33219404 DOI: 10.1007/s00204-020-02952-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/05/2020] [Indexed: 12/31/2022]
Abstract
Early detection strategies and improvements in cancer treatment have dramatically reduced the cancer mortality rate in the United States (US). However, cardiovascular (CV) side effects of cancer therapy are frequent among the 17 million cancer survivors in the US today, and cardiovascular disease (CVD) has become the second leading cause of morbidity and mortality among cancer survivors. Circulating biomarkers are ideal for detecting and monitoring CV side effects of cancer therapy. Here, we summarize the current state of clinical studies on conventional serum and plasma CVD biomarkers to detect and prevent cardiac injury during cancer treatment. We also review how novel exploratory tools such as genetic testing, human stem cell-derived cardiomyocytes, Omics technologies, and artificial intelligence can elucidate underlying molecular and genetic mechanisms of CV injury and to improve predicting cancer therapy-related cardiotoxicity (CTRC). Current regulatory requirements for biomarker qualifications are also addressed. We present generally applicable lessons learned from published studies, particularly on how to improve reproducibility. The combination of conventional circulating biomarkers and novel exploratory tools will pave the way for precision medicine and improve the clinical practice of prediction, detection, and management of CTRC.
Collapse
Affiliation(s)
- Li Pang
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Zhichao Liu
- Division of Bioinformation and Biostatistics, National Center for Toxicological Research, US. Food and Drug Administration, Jefferson, AR, USA
| | - Feng Wei
- Department of Structural Heart Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chengzhong Cai
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Xi Yang
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, & Nephrology, Office of New Drug, Center for Drug Evaluation and Research, US. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
44
|
Liu R, Li D, Sun F, Rampoldi A, Maxwell JT, Wu R, Fischbach P, Castellino SM, Du Y, Fu H, Mandawat A, Xu C. Melphalan induces cardiotoxicity through oxidative stress in cardiomyocytes derived from human induced pluripotent stem cells. Stem Cell Res Ther 2020; 11:470. [PMID: 33153480 PMCID: PMC7643439 DOI: 10.1186/s13287-020-01984-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/20/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment-induced cardiotoxicity is a leading noncancer-related cause of acute and late onset morbidity and mortality in cancer patients on antineoplastic drugs such as melphalan-increasing clinical case reports have documented that it could induce cardiotoxicity including severe arrhythmias and heart failure. As the mechanism by which melphalan impairs cardiac cells remains poorly understood, here, we aimed to use cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs) to investigate the cellular and molecular mechanisms of melphalan-induced cardiotoxicity. METHODS hiPSC-CMs were generated and treated with clinically relevant doses of melphalan. To characterize melphalan-induced cardiotoxicity, cell viability and apoptosis were quantified at various treatment durations. Ca2+ transient and contractility analyses were used to examine the alterations of hiPSC-CM function. Proteomic analysis, reactive oxygen species detection, and RNA-Sequencing were conducted to investigate underlying mechanisms. RESULTS Melphalan treatment of hiPSC-CMs induced oxidative stress, caused Ca2+ handling defects and dysfunctional contractility, altered global transcriptomic and proteomic profiles, and resulted in apoptosis and cell death. The antioxidant N-acetyl-L-cysteine attenuated these genomic, cellular, and functional alterations. In addition, several other signaling pathways including the p53 and transforming growth factor-β signaling pathways were also implicated in melphalan-induced cardiotoxicity according to the proteomic and transcriptomic analyses. CONCLUSIONS Melphalan induces cardiotoxicity through the oxidative stress pathway. This study provides a unique resource of the global transcriptomic and proteomic datasets for melphalan-induced cardiotoxicity and can potentially open up new clinical mechanism-based targets to prevent and treat melphalan-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rui Liu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
- Department of Pediatrics, The Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Dong Li
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Fangxu Sun
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Antonio Rampoldi
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Joshua T Maxwell
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Peter Fischbach
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA
| | - Yuhong Du
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haian Fu
- Emory Chemical Biology Discovery Center and the Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Anant Mandawat
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Cardio-Oncology Program, Winship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - Chunhui Xu
- Department of Pediatrics, Emory University School of Medicine and Children's Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA, 30322, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
45
|
Pang L. Toxicity testing in the era of induced pluripotent stem cells: A perspective regarding the use of patient-specific induced pluripotent stem cell–derived cardiomyocytes for cardiac safety evaluation. CURRENT OPINION IN TOXICOLOGY 2020. [DOI: 10.1016/j.cotox.2020.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Assessment of Cardiotoxicity With Stem Cell-based Strategies. Clin Ther 2020; 42:1892-1910. [PMID: 32938533 DOI: 10.1016/j.clinthera.2020.08.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/20/2020] [Accepted: 08/25/2020] [Indexed: 02/08/2023]
Abstract
PURPOSE Adverse cardiovascular drug effects pose a substantial medical risk and represent a common cause of drug withdrawal from the market. Thus, current in vitro assays and in vivo animal models still have shortcomings in assessing cardiotoxicity. A human model for more accurate preclinical cardiotoxicity assessment is highly desirable. Current differentiation protocols allow for the generation of human pluripotent stem cell-derived cardiomyocytes in basically unlimited numbers and offer the opportunity to study drug effects on human cardiomyocytes. The purpose of this review is to provide a brief overview of the current approaches to translate studies with pluripotent stem cell-derived cardiomyocytes from basic science to preclinical risk assessment. METHODS A review of the literature was performed to gather data on the pathophysiology of cardiotoxicity, the current cardiotoxicity screening assays, stem cell-derived cardiomyocytes, and their application in cardiotoxicity screening. FINDINGS There is increasing evidence that stem cell-derived cardiomyocytes predict arrhythmogenicity with high accuracy. Cardiomyocyte immaturity represents the major limitation so far. However, strategies are being developed to overcome this hurdle, such as tissue engineering. In addition, stem cell-based strategies offer the possibility to assess structural drug toxicity (eg, by anticancer drugs) on complex models that more closely mirror the structure of the heart and contain endothelial cells and fibroblasts. IMPLICATIONS Pluripotent stem cell-derived cardiomyocytes have the potential to substantially change how preclinical cardiotoxicity screening is performed. To which extent they will replace or complement current approaches is being evaluated.
Collapse
|
47
|
Miller JM, Meki MH, Ou Q, George SA, Gams A, Abouleisa RRE, Tang XL, Ahern BM, Giridharan GA, El-Baz A, Hill BG, Satin J, Conklin DJ, Moslehi J, Bolli R, Ribeiro AJS, Efimov IR, Mohamed TMA. Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity. Toxicol Appl Pharmacol 2020; 406:115213. [PMID: 32877659 DOI: 10.1016/j.taap.2020.115213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 μm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 μM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.
Collapse
Affiliation(s)
- Jessica M Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Moustafa H Meki
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Anna Gams
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Riham R E Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Brooke M Ahern
- Department of Physiology, University of Kentucky, KY, USA
| | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, KY, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, USA
| | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Javid Moslehi
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Alexandre J S Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, Division of Applied Regulatory Science, Silver Spring, MD, USA.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA; Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, KY, USA; Institute of Cardiovascular Sciences, University of Manchester, UK; Faculty of Pharmacy, Zagazig University, Egypt.
| |
Collapse
|
48
|
Guns PJD, Guth BD, Braam S, Kosmidis G, Matsa E, Delaunois A, Gryshkova V, Bernasconi S, Knot HJ, Shemesh Y, Chen A, Markert M, Fernández MA, Lombardi D, Grandmont C, Cillero-Pastor B, Heeren RMA, Martinet W, Woolard J, Skinner M, Segers VFM, Franssen C, Van Craenenbroeck EM, Volders PGA, Pauwelyn T, Braeken D, Yanez P, Correll K, Yang X, Prior H, Kismihók G, De Meyer GRY, Valentin JP. INSPIRE: A European training network to foster research and training in cardiovascular safety pharmacology. J Pharmacol Toxicol Methods 2020; 105:106889. [PMID: 32565326 DOI: 10.1016/j.vascn.2020.106889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 02/05/2023]
Abstract
Safety pharmacology is an essential part of drug development aiming to identify, evaluate and investigate undesirable pharmacodynamic properties of a drug primarily prior to clinical trials. In particular, cardiovascular adverse drug reactions (ADR) have halted many drug development programs. Safety pharmacology has successfully implemented a screening strategy to detect cardiovascular liabilities, but there is room for further refinement. In this setting, we present the INSPIRE project, a European Training Network in safety pharmacology for Early Stage Researchers (ESRs), funded by the European Commission's H2020-MSCA-ITN programme. INSPIRE has recruited 15 ESR fellows that will conduct an individual PhD-research project for a period of 36 months. INSPIRE aims to be complementary to ongoing research initiatives. With this as a goal, an inventory of collaborative research initiatives in safety pharmacology was created and the ESR projects have been designed to be complementary to this roadmap. Overall, INSPIRE aims to improve cardiovascular safety evaluation, either by investigating technological innovations or by adding mechanistic insight in emerging safety concerns, as observed in the field of cardio-oncology. Finally, in addition to its hands-on research pillar, INSPIRE will organize a number of summer schools and workshops that will be open to the wider community as well. In summary, INSPIRE aims to foster both research and training in safety pharmacology and hopes to inspire the future generation of safety scientists.
Collapse
Affiliation(s)
- Pieter-Jan D Guns
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Brian D Guth
- Boehringer Ingelheim Pharma GmbH & Co KG, Drug Discovery Sciences, Biberach an der Riss, Germany
| | | | | | | | - Annie Delaunois
- UCB Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine-l'Alleud, Belgium
| | - Vitalina Gryshkova
- UCB Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine-l'Alleud, Belgium
| | | | | | - Yair Shemesh
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Markert
- Boehringer Ingelheim Pharma GmbH & Co KG, Drug Discovery Sciences, Biberach an der Riss, Germany
| | | | | | | | - Berta Cillero-Pastor
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, the Netherlands
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging Institute (M4I), Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, the Netherlands
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, Centre of Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, United Kingdom
| | - Matt Skinner
- Vivonics Preclinical Ltd, BioCity, Nottingham, United Kingdom
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Constantijn Franssen
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Paul G A Volders
- Department of Cardiology, CARIM, Maastricht University Medical Center+, Maastricht, the Netherlands
| | | | | | - Paz Yanez
- Department of Research Affairs & Innovation, University of Antwerp, Antwerp, Belgium
| | - Krystle Correll
- Safety Pharmacology Society, Reston, Virginia, United States
| | - Xi Yang
- Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Helen Prior
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK
| | - Gábor Kismihók
- Leibniz Information Centre for Science and Technology, Hannover, Germany; Marie Curie Alumni Association, Brussels, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Jean-Pierre Valentin
- UCB Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine-l'Alleud, Belgium
| |
Collapse
|
49
|
Shi M, Tien NT, de Haan L, Louisse J, Rietjens IMCM, Bouwmeester H. Evaluation of in vitro models of stem cell-derived cardiomyocytes to screen for potential cardiotoxicity of chemicals. Toxicol In Vitro 2020; 67:104891. [PMID: 32446838 DOI: 10.1016/j.tiv.2020.104891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/14/2020] [Accepted: 05/16/2020] [Indexed: 12/12/2022]
Abstract
Cardiotoxicity is an important toxicological endpoint for chemical and drug safety assessment. The present study aims to evaluate two stemcell-based in vitro models for cardiotoxicity screening of chemicals. Eleven model compounds were used to evaluate responses of mouse embryonic stem cell-derived cardiomyocytes (mESC-CMs) using beating arrest as a readout and the analysis of electrophysiological parameters measured with a multi-electrode array (MEA) platform of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Results revealed that the hiPSC-CM MEA assay responded to all compounds. The mESC-CM beating arrest assay was not responsive to potassium channel blockers and showed a lower sensitivity to sodium channel blockers and Na+/K+ ATPase inhibitors compared to the hiPSC-CM MEA assay. Calcium channel blockers and a β-adrenergic receptor agonist showed comparable potencies in both models. The in vitro response concentrations from hiPSC-CMs were highly concordant with human effective serum concentrations of potassium and sodium channel blockers. It is concluded that both in vitro models enable the cardiotoxicity screening with different applicability domains. The mESC-CM beating arrest assay may be used as a first step in a tiered approach while the hiPSC-CM MEA assay may be the best starting point for quantitative in vitro to in vivo extrapolations.
Collapse
Affiliation(s)
- Miaoying Shi
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands.
| | - Nguyen T Tien
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands
| | - Laura de Haan
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands.
| | - Jochem Louisse
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands.
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands.
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700, EA, Wageningen, the Netherlands.
| |
Collapse
|
50
|
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: 7] [Impact Index Per Article: 1.8] [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
|