1932

Abstract

In recent decades, drug development costs have increased by approximately a hundredfold, and yet about 1 in 7 licensed drugs are withdrawn from the market, often due to cardiotoxicity. This review considers whether technologies using human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) could complement existing assays to improve discovery and safety while reducing socioeconomic costs and assisting with regulatory guidelines on cardiac safety assessments. We draw on lessons from our own work to suggest a panel of 12 drugs that will be useful in testing the suitability of hiPSC-CM platforms to evaluate contractility. We review issues, including maturity versus complexity, consistency, quality, and cost, while considering a potential need to incorporate auxiliary approaches to compensate for limitations in hiPSC-CM technology. We give examples on how coupling hiPSC-CM technologies with Cas9/CRISPR genome engineering is starting to be used to personalize diagnosis, stratify risk, provide mechanistic insights, and identify new pathogenic variants for cardiovascular disease.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010919-023309
2020-01-06
2024-04-18
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/60/1/annurev-pharmtox-010919-023309.html?itemId=/content/journals/10.1146/annurev-pharmtox-010919-023309&mimeType=html&fmt=ahah

Literature Cited

  1. 1. 
    Moffat JG, Vincent F, Lee JA, Eder J, Prunotto M 2017. Opportunities and challenges in phenotypic drug discovery: an industry perspective. Nat. Rev. Drug Discov. 16:531–43
    [Google Scholar]
  2. 2. 
    Denning C, Borgdorff V, Crutchley J, Firth KSA, George V et al. 2016. Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform. Biochim. Biophys. Acta 1863:1728–48
    [Google Scholar]
  3. 3. 
    Int. Fed. Pharm. Manuf. Assoc 2017. The Pharmaceutical Industry and Global Health: Facts and Figures 2017 Int. Fed. Pharm. Manuf. Assoc. https://www.ifpma.org/wp-content/uploads/2017/02/IFPMA-Facts-And-Figures-2017.pdf
  4. 4. 
    Catapult Med. Disc 2018. State of the discovery nation 2018 and the role of the Medicines Discovery Catapult: joint report by the Bioindustry Association and the Medicines Discovery Catapult Rep., Catapult Med. Disc., Alderley Edge UK: https://md.catapult.org.uk/resources/report-state-of-the-discovery-nation-2018/
  5. 5. 
    Sakai S, Miyauchi T, Kobayashi M, Yamaguchi I, Goto K, Sugishita Y 1996. Inhibition of myocardial endothelin pathway improves long-term survival in heart failure. Nature 384:353–55
    [Google Scholar]
  6. 6. 
    Spinale FG, Walker JD, Mukherjee R, Iannini JP, Keever AT, Gallagher KP 1997. Concomitant endothelin receptor subtype-A blockade during the progression of pacing-induced congestive heart failure in rabbits: beneficial effects on left ventricular and myocyte function. Circulation 95:1918–29
    [Google Scholar]
  7. 7. 
    Packer M, McMurray J, Massie BM, Caspi A, Charlon V et al. 2005. Clinical effects of endothelin receptor antagonism with bosentan in patients with severe chronic heart failure: results of a pilot study. J. Card. Fail. 11:12–20
    [Google Scholar]
  8. 8. 
    Dixon JA, Spinale FG. 2009. Large animal models of heart failure a critical link in the translation of basic science to clinical practice. Circ. Heart Fail. 2:262–71
    [Google Scholar]
  9. 9. 
    Chen YC, Yu JH, Niu YY, Qin DD, Liu HL et al. 2017. Modeling Rett syndrome using TALEN-edited MECP2 mutant cynomolgus monkeys. Cell 169:945–55.e10
    [Google Scholar]
  10. 10. 
    Wang Y-K, Zhu W-W, Wu M-H, Wu Y-H, Liu Z-X et al. 2018. Human clinical-grade parthenogenetic ESC-derived dopaminergic neurons recover locomotive defects of nonhuman primate models of Parkinson's disease. Stem Cell Rep 11:171–82
    [Google Scholar]
  11. 11. 
    Qiu J, Zhou Q. 2017. Qi Zhou: the coming of age in China's stem-cell research and regenerative medicine. Natl. Sci. Rev. 4:550–52
    [Google Scholar]
  12. 12. 
    Bowes J, Brown AJ, Hamon J, Jarolimek W, Sridhar A et al. 2012. Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat. Rev. Drug Discov. 11:909–22
    [Google Scholar]
  13. 13. 
    Laverty HG, Benson C, Cartwright EJ, Cross MJ, Garland C et al. 2011. How can we improve our understanding of cardiovascular safety liabilities to develop safer medicines?. Br. J. Pharmacol. 163:675–93
    [Google Scholar]
  14. 14. 
    Onakpoya IJ, Heneghan CJ, Aronson JK 2016. Post-marketing withdrawal of 462 medicinal products because of adverse drug reactions: a systematic review of the world literature. BMC Med 14:10
    [Google Scholar]
  15. 15. 
    Qureshi ZP, Seoane-Vazquez E, Rodriguez-Monguio R, Stevenson KB, Szeinbach SL 2011. Market withdrawal of new molecular entities approved in the United States from 1980 to 2009. Pharmacoepidemiol. Drug Saf. 20:772–77
    [Google Scholar]
  16. 16. 
    Bhattacharya S. 2005. Up to 140,000 heart attacks linked to Vioxx. NewScientist Jan. 25. https://www.newscientist.com/article/dn6918-up-to-140000-heart-attacks-linked-to-vioxx/
    [Google Scholar]
  17. 17. 
    Fuentes AV, Pineda MD, Venkata KCN 2018. Comprehension of top 200 prescribed drugs in the US as a resource for pharmacy teaching, training and practice. Pharmacy 6:43
    [Google Scholar]
  18. 18. 
    Schachter AD, Ramoni MF. 2007. Clinical forecasting in drug development. Nat. Rev. Drug Discov. 6:107–8
    [Google Scholar]
  19. 19. 
    Raab S, Klingenstein M, Liebau S, Linta L 2014. A comparative view on human somatic cell sources for iPSC generation. Stem Cells Int 2014:768391
    [Google Scholar]
  20. 20. 
    Xue Y, Cai X, Wang L, Liao B, Zhang H et al. 2013. Generating a non-integrating human induced pluripotent stem cell bank from urine-derived cells. PLOS ONE 8:e70573
    [Google Scholar]
  21. 21. 
    Smith JGW, Owen T, Bhagwan JR, Mosqueira D, Scott E et al. 2018. Isogenic pairs of hiPSC-CMs with hypertrophic cardiomyopathy/LVNC-associated ACTCI E99K mutation unveil differential functional deficits. Stem Cell Rep 11:1226–43
    [Google Scholar]
  22. 22. 
    Mosqueira D, Mannhardt I, Bhagwan JR, Lis-Slimak K, Katili P et al. 2018. CRISPR/Cas9 editing in human pluripotent stem cell–cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. Eur. Heart J. 39:3879–92
    [Google Scholar]
  23. 23. 
    Devalla HD, Schwach V, Ford JW, Milnes JT, El-Haou S et al. 2015. Atrial-like cardiomyocytes from human pluripotent stem cells are a robust preclinical model for assessing atrial-selective pharmacology. EMBO Mol. Med. 7:394–410
    [Google Scholar]
  24. 24. 
    Lee JH, Protze SI, Laksman Z, Backx PH, Keller GM 2017. Human pluripotent stem cell-derived atrial and ventricular cardiomyocytes develop from distinct mesoderm populations. Cell Stem Cell 21:179–94.e4
    [Google Scholar]
  25. 25. 
    Lemme M, Ulmer BM, Lemoine MD, Zech ATL, Flenner F et al. 2018. Atrial-like engineered heart tissue: an in vitro model of the human atrium. Stem Cell Rep 11:1378–90
    [Google Scholar]
  26. 26. 
    Eschenhagen T, Mummery C, Knollmann BC 2015. Modelling sarcomeric cardiomyopathies in the dish: from human heart samples to iPSC cardiomyocytes. Cardiovasc. Res. 105:424–38
    [Google Scholar]
  27. 27. 
    Soares FAC, Sheldon M, Rao M, Mummery C, Vallier L 2014. International coordination of large-scale human induced pluripotent stem cell initiatives: Wellcome Trust and ISSCR workshops white paper. Stem Cell Rep 3:931–39
    [Google Scholar]
  28. 28. 
    Eschenhagen T, Carrier L. 2018. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes-a systematic review. Pflugers Arch 471:755–68
    [Google Scholar]
  29. 29. 
    Hockemeyer D, Jaenisch R. 2016. Induced pluripotent stem cells meet genome editing. Cell Stem Cell 18:573–86
    [Google Scholar]
  30. 30. 
    Doench JG. 2018. Am I ready for CRISPR? A user's guide to genetic screens. Nat. Rev. Genet. 19:67–80
    [Google Scholar]
  31. 31. 
    Sala L, Bellin M, Mummery CL 2017. Integrating cardiomyocytes from human pluripotent stem cells in safety pharmacology: Has the time come?. Br. J. Pharmacol. 174:3749–65
    [Google Scholar]
  32. 32. 
    Kramer J, Obejero-Paz CA, Myatt G, Kuryshev YA, Bruening-Wright A et al. 2013. MICE models: superior to the HERG model in predicting Torsade de Pointes. Sci. Rep. 3:2100
    [Google Scholar]
  33. 33. 
    Kanda Y, Yamazaki D, Osada T, Yoshinaga T, Sawada K 2018. Development of torsadogenic risk assessment using human induced pluripotent stem cell-derived cardiomyocytes: Japan iPS Cardiac Safety Assessment (JiCSA) update. J. Pharmacol. Sci. 138:233–39
    [Google Scholar]
  34. 34. 
    Sager PT, Gintant G, Turner JR, Pettit S, Stockbridge N 2014. Rechanneling the cardiac proarrhythmia safety paradigm: a meeting report from the Cardiac Safety Research Consortium. Am. Heart J. 167:292–300
    [Google Scholar]
  35. 35. 
    Fermini B, Hancox JC, Abi-Gerges N, Bridgland-Taylor M, Chaudhary KW et al. 2016. A new perspective in the field of cardiac safety testing through the comprehensive in vitro proarrhythmia assay paradigm. J. Biomol. Screen. 21:1–11
    [Google Scholar]
  36. 36. 
    ICH (Int. Counc. Harmon. Tech. Requir. Pharm. Hum. Use) 2005. The non-clinical evaluation of the potential for delayed ventricular repolarization (QT interval prolongation) by human pharmaceuticals Guideline S7B ICH, Geneva:
  37. 37. 
    Lu HR, Vlaminckx E, Van Ammel K, De Clerck F 2002. Drug-induced long QT in isolated rabbit Purkinje fibers: importance of action potential duration, triangulation and early afterdepolarizations. Eur. J. Pharmacol. 452:183–92
    [Google Scholar]
  38. 38. 
    Pugsley MK, Authier S, Koerner JE, Redfern WS, Markgraf CG et al. 2018. An overview of the safety pharmacology society strategic plan. J. Pharmacol. Toxicol. Methods 93:35–45
    [Google Scholar]
  39. 39. 
    Blinova K, Dang Q, Millard D, Smith G, Pierson J et al. 2018. International multisite study of human-induced pluripotent stem cell-derived cardiomyocytes for drug proarrhythmic potential assessment. Cell Rep 24:3582–92
    [Google Scholar]
  40. 40. 
    Kopljar I, Lu HR, Van Ammel K, Otava M, Tekle F et al. 2018. Development of a human iPSC cardiomyocyte–based scoring system for cardiac hazard identification in early drug safety de-risking. Stem Cell Rep 11:1365–77
    [Google Scholar]
  41. 41. 
    Lu Z, Wu C-YC, Jiang Y-P, Ballou LM, Clausen C et al. 2012. Suppression of phosphoinositide 3-kinase signaling and alteration of multiple ion currents in drug-induced long QT syndrome. Sci. Transl. Med. 4:131ra50
    [Google Scholar]
  42. 42. 
    Ando H, Yoshinaga T, Yamamotoa W, Asakura K, Uda T et al. 2017. A new paradigm for drug-induced torsadogenic risk assessment using human iPS cell–derived cardiomyocytes. J. Pharmacol. Toxicol. Methods 84:111–27
    [Google Scholar]
  43. 43. 
    Yamamoto W, Asakura K, Ando H, Taniguchi T, Ojima A et al. 2016. Electrophysiological characteristics of human iPSC–derived cardiomyocytes for the assessment of drug-induced proarrhythmic potential. PLOS ONE 11:e0167348
    [Google Scholar]
  44. 44. 
    Strauss D. 2018. Questions & answers: clinical and non-clinical evaluation of QT/QTc interval prolongation and proarryhythmic potential Doc. E14/S7B, Int. Counc. Harmon. Tech. Requir. Pharm Hum. Use, Geneva: https://www.ich.org/products/guidelines/efficacy/article/efficacy-guidelines.html#13-3
  45. 45. 
    ICH (Int. Counc. Harmon. Tech. Requir. Pharm. Hum. Use) 2018. ICH S7B and E14 Q&A. Final Concept Pap., ICH, Geneva. https://www.ich.org/fileadmin/Public_Web_Site/ICH_Products/Guidelines/Efficacy/E14/E14S7BIWG_ConceptPaper_Final_2018_1122.pdf
  46. 46. 
    ICH (Int. Counc. Harmon. Tech. Requir. Pharm. Hum. Use) 2019. ICH E14/S7B IWG work plan. Work Plan, ICH, Geneva. https://database.ich.org/sites/default/files/E14-S7BIWG_WorkPlan_2019_0731.pdf
  47. 47. 
    Costa VM, Carvalho F, Duarte JA, de Lourdes Bastos M, Remiao F 2013. The heart as a target for xenobiotic toxicity: the cardiac susceptibility to oxidative stress. Chem. Res. Toxicol. 26:1285–311
    [Google Scholar]
  48. 48. 
    Berridge BR, Mowat V, Nagai H, Nyska A, Okazaki Y et al. 2016. Non-proliferative and proliferative lesions of the cardiovascular system of the rat and mouse. J. Toxicol. Pathol. 29:1S–47S
    [Google Scholar]
  49. 49. 
    Marwick TH. 2018. Ejection fraction pros and cons: JACC state-of-the-art review. J. Am. Coll. Cardiol. 72:2360–79
    [Google Scholar]
  50. 50. 
    Guo L, Dong Z, Guthrie H 2009. Validation of a guinea pig Langendorff heart model for assessing potential cardiovascular liability of drug candidates. J. Pharmacol. Toxicol. Methods 60:130–51
    [Google Scholar]
  51. 51. 
    Pointon A, Pilling J, Dorval T, Wang Y, Archer C, Pollard C 2017. High-throughput imaging of cardiac microtissues for the assessment of cardiac contraction during drug. Toxicol. Sci. 155:444–57
    [Google Scholar]
  52. 52. 
    Pointon A, Harmer AR, Dale IL, Abi-Gerges N, Bowes J et al. 2015. Assessment of cardiomyocyte contraction in human-induced pluripotent stem cell-derived cardiomyocytes. Toxicol. Sci. 144:227–37
    [Google Scholar]
  53. 53. 
    Archer CR, Sargeant R, Basak J, Pilling J, Barnes JR, Pointon A 2018. Characterization and validation of a human 3D cardiac microtissue for the assessment of changes in cardiac pathology. Sci. Rep. 8:10160
    [Google Scholar]
  54. 54. 
    Oleaga C, Riu A, Rothemund S, Lavado A, McAleer CW et al. 2018. Investigation of the effect of hepatic metabolism on off-target cardiotoxicity in a multi-organ human-on-a-chip system. Biomaterials 182:176–90
    [Google Scholar]
  55. 55. 
    Sala L, van Meer BJ, Tertoolen LGJ, Bakkers J, Bellin M et al. 2018. MUSCLEMOTION: a versatile open software tool to quantify cardiomyocyte and cardiac muscle contraction in vitro and in vivo. Circ. Res. 122:e5–16
    [Google Scholar]
  56. 56. 
    Duncan G, Firth K, George V, Hoang MD, Staniforth A et al. 2017. Drug-mediated shortening of action potentials in LQTS2 human induced pluripotent stem cell–derived cardiomyocytes. Stem Cells Dev 26:1695–705
    [Google Scholar]
  57. 57. 
    Mannhardt I, Breckwoldt K, Letuffe-Breniere D, Schaaf S, Schulz H et al. 2016. Human engineered heart tissue: analysis of contractile force. Stem Cell Rep 7:29–42
    [Google Scholar]
  58. 58. 
    Butler L, Cros C, Oldman KL, Harmer AR, Pointon A et al. 2015. Enhanced characterization of contractility in cardiomyocytes during early drug safety assessment. Toxicol. Sci. 145:396–406
    [Google Scholar]
  59. 59. 
    Lemoine MD, Mannhardt I, Breckwoldt K, Prondzynski M, Flenner F et al. 2017. Human iPSC-derived cardiomyocytes cultured in 3D engineered heart tissue show physiological upstroke velocity and sodium current density. Sci. Rep. 7:5464
    [Google Scholar]
  60. 60. 
    Lemoine MD, Krause T, Koivumaeki JT, Prondzynski M, Schulze ML et al. 2018. Human induced pluripotent stem cell-derived engineered heart tissue as a sensitive test system for QT prolongation and arrhythmic triggers. Circ. Arrhythm. Electrophysiol. 11:e006035
    [Google Scholar]
  61. 61. 
    Mannhardt I, Eder A, Dumotier B, Prondzynski M, Kraemer E et al. 2017. Blinded contractility analysis in hiPSC-cardiomyocytes in engineered heart tissue format: comparison with human atrial trabeculae. Toxicol. Sci. 158:164–75
    [Google Scholar]
  62. 62. 
    Scott CW, Zhang X, Abi-Gerges N, Lamore SD, Abassi YA, Peters MF 2014. An impedance-based cellular assay using human iPSC-derived cardiomyocytes to quantify modulators of cardiac contractility. Toxicol. Sci. 142:331–38
    [Google Scholar]
  63. 63. 
    Shakur Y, Fong M, Hensley J, Cone J, Movsesian MA et al. 2002. Comparison of the effects of cilostazol and milrinone on cAMP-PDE activity, intracellular cAMP and calcium in the heart. Cardiovasc. Drugs Ther. 16:417–27
    [Google Scholar]
  64. 64. 
    Lee Y-K, Ng K-M, Lai W-H, Chan Y-C, Lau Y-M et al. 2011. Calcium homeostasis in human induced pluripotent stem cell-derived cardiomyocytes. Stem Cell Rev. Rep. 7:976–86
    [Google Scholar]
  65. 65. 
    Ronaldson-Bouchard K, Ma SP, Yeager K, Chen T, Song L et al. 2018. Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556:239–43
    [Google Scholar]
  66. 66. 
    MacQueen LA, Sheehy SP, Chantre CO, Zimmerman JF, Pasqualini FS et al. 2018. A tissue-engineered scale model of the heart ventricle. Nat. Biomed. Eng. 2:930–41
    [Google Scholar]
  67. 67. 
    Li RA, Keung W, Cashman TJ, Backeris PC, Johnson BV et al. 2018. Bioengineering an electro-mechanically functional miniature ventricular heart chamber from human pluripotent stem cells. Biomaterials 163:116–27
    [Google Scholar]
  68. 68. 
    Zhao Y, Rafatian N, Feric NT, Cox BJ, Aschar-Sobbi R et al. 2019. A platform for generation of chamber-specific cardiac tissues and disease modeling. Cell 176:4913–27.e18
    [Google Scholar]
  69. 69. 
    Hortigon-Vinagre MP, Zamora V, Burton FL, Green J, Gintant GA, Smith GL 2016. The use of ratiometric fluorescence measurements of the voltage sensitive dye di-4-ANEPPS to examine action potential characteristics and drug effects on human induced pluripotent stem cell–derived cardiomyocytes. Toxicol. Sci. 154:320–31
    [Google Scholar]
  70. 70. 
    Thomas SHL, Behr ER. 2016. Pharmacological treatment of acquired QT prolongation and Torsades de Pointes. Br. J. Clin. Pharmacol. 81:420–27
    [Google Scholar]
  71. 71. 
    Pinski SL, Eguia LE, Trohman RG 2002. What is the minimal pacing rate that prevents Torsades de Pointes? Insights from patients with permanent pacemakers. Pacing Clin. Electrophysiol. 25:1612–15
    [Google Scholar]
  72. 72. 
    Giacomelli E, Bellin M, Sala L, Van Meer BJ, Tertoolen LGJ et al. 2017. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development 144:1008–17
    [Google Scholar]
  73. 73. 
    Mills RJ, Titmarsh DM, Koenig X, Parker BL, Ryall JG et al. 2017. Functional screening in human cardiac organoids reveals a metabolic mechanism for cardiomyocyte cell cycle arrest. PNAS 114:E8372–81
    [Google Scholar]
  74. 74. 
    Krijger PHL, Di Stefano B, de Wit E, Limone F, van Oevelen C et al. 2016. Cell-of-origin-specific 3D genome structure acquired during somatic cell reprogramming. Cell Stem Cell 18:597–610
    [Google Scholar]
  75. 75. 
    Birket MJ, Ribeiro MC, Verkerk AO, Ward D, Leitoguinho AR et al. 2015. Expansion and patterning of cardiovascular progenitors derived from human pluripotent stem cells. Nat. Biotechnol. 33:970–79
    [Google Scholar]
  76. 76. 
    Woo LA, Tkachenko S, Ding M, Plowright AT, Engkvist O et al. 2019. High-content phenotypic assay for proliferation of human iPSC-derived cardiomyocytes identifies L-type calcium channels as targets. J. Mol. Cell. Cardiol. 127:204–14
    [Google Scholar]
  77. 77. 
    Zhang X, Guo L, Zeng H, White SL, Furniss M et al. 2016. Multi-parametric assessment of cardiomyocyte excitation-contraction coupling using impedance and field potential recording: a tool for cardiac safety assessment. J. Pharmacol. Toxicol. Methods 81:201–16
    [Google Scholar]
  78. 78. 
    Hortigon-Vinagre MP, Zamora V, Burton FL, Butler P, Smith GL 2018. Measurements of voltage, intracellular Ca2+ and contraction from spontaneous iPSC-derived cardiomyocytes using CellOPTIQ® platform reveals complex effects from drugs that parallel in vivo cardiac effects. J. Pharmacol. Toxicol. Methods 93:119–20
    [Google Scholar]
  79. 79. 
    Conant G, Ahadian S, Zhao Y, Radisic M 2017. Kinase inhibitor screening using artificial neural networks and engineered cardiac biowires. Sci. Rep. 7:11807
    [Google Scholar]
  80. 80. 
    Gong JQX, Sobie EA. 2018. Population-based mechanistic modeling allows for quantitative predictions of drug responses across cell types. NPJ Syst. Biol. Appl. 4:11
    [Google Scholar]
  81. 81. 
    Reynolds JG, Geretti E, Hendriks BS, Lee H, Leonard SC et al. 2012. HER2-targeted liposomal doxorubicin displays enhanced anti-tumorigenic effects without associated cardiotoxicity. Toxicol. Appl. Pharmacol. 262:1–10
    [Google Scholar]
  82. 82. 
    Terrenoire C, Wang K, Tung KWC, Chung WK, Pass RH et al. 2013. Induced pluripotent stem cells used to reveal drug actions in a long QT syndrome family with complex genetics. J. Gen. Physiol. 141:61–72
    [Google Scholar]
  83. 83. 
    Chaudhari U, Nemade H, Sureshkumar P, Vinken M, Ates G et al. 2018. Functional cardiotoxicity assessment of cosmetic compounds using human-induced pluripotent stem cell–derived cardiomyocytes. Arch. Toxicol. 92:371–81
    [Google Scholar]
  84. 84. 
    Casartelli A, Lanzoni A, Comelli R, Crivellente F, Defazio R et al. 2011. A novel and integrated approach for the identification and characterization of drug-induced cardiac toxicity in the dog. Toxicol. Pathol. 39:361–71
    [Google Scholar]
  85. 85. 
    Wang H, Sheehan RP, Palmer AC, Everley RA, Boswell SA et al. 2019. Adaptation of human iPSC-derived cardiomyocytes to tyrosine kinase inhibitors reduces acute cardiotoxicity via metabolic reprogramming. bioRxiv 365841. https://doi.org/10.1101/365841
    [Crossref]
  86. 86. 
    Ma N, Zhang JZ, Itzhaki I, Zhang SL, Chen HD et al. 2018. Determining the pathogenicity of a genomic variant of uncertain significance using CRISPR/Cas9 and human-induced pluripotent stem cells. Circulation 138:2666–81
    [Google Scholar]
  87. 87. 
    Burridge PW, Li YF, Matsa E, Wu H, Ong S-G et al. 2016. Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat. Med. 22:547–56
    [Google Scholar]
  88. 88. 
    Matsa E, Burridge PW, Yu K-H, Ahrens JH, Termglinchan V et al. 2016. Transcriptome profiling of patient-specific human iPSC-cardiomyocytes predicts individual drug safety and efficacy responses in vitro. Cell Stem Cell 19:311–25
    [Google Scholar]
  89. 89. 
    Bassez G, Audureau E, Hogrel J-Y, Arrouasse R, Baghdoyan S et al. 2018. Improved mobility with metformin in patients with myotonic dystrophy type 1: a randomized controlled trial. Brain 141:2855–65
    [Google Scholar]
  90. 90. 
    Sankar PL, Parker LS. 2017. The Precision Medicine Initiative's All of Us Research Program: an agenda for research on its ethical, legal, and social issues. Genet. Med. 19:743–50
    [Google Scholar]
  91. 91. 
    Marian AJ, Roberts R. 2001. The molecular genetic basis for hypertrophic cardiomyopathy. J. Mol. Cell. Cardiol. 33:655–70
    [Google Scholar]
  92. 92. 
    Garg P, Oikonomopoulos A, Chen H, Li Y, Lam CK et al. 2018. Genome editing of induced pluripotent stem cells to decipher cardiac channelopathy variant. J. Am. Coll. Cardiol. 72:62–75
    [Google Scholar]
  93. 93. 
    Fan M. 2017. Induced pluripotent stem cells: global markets Rep., BCC Research Wellesley, MA:
  94. 94. 
    Bargehr J, Ong LP, Colzani M, Davaapil H, Hofsteen Pet al. 2019. Epicardial cells derived from human embryonic stem cells augment cardiomyocyte-driven heart regeneration. Nat. Biotechnol 378895906
    [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010919-023309
Loading
/content/journals/10.1146/annurev-pharmtox-010919-023309
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error