1932

Abstract

Billions of US dollars are invested every year by the pharmaceutical industry in drug development, with the aim of introducing new drugs that are effective and have minimal side effects. Thirty percent of in-pipeline drugs are excluded in an early phase of preclinical and clinical screening owing to cardiovascular safety concerns, and several lead molecules that pass the early safety screening make it to market but are later withdrawn owing to severe cardiac side effects. Although the current drug safety screening methodologies can identify some cardiotoxic drug candidates, they cannot accurately represent the human heart in many aspects, including genomics, transcriptomics, and patient- or population-specific cardiotoxicity. Despite some limitations, human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) are a powerful and evolving technology that has been shown to recapitulate many attributes of human cardiomyocytes and their drug responses. In this review, we discuss the potential impact of the inclusion of the hiPSC-CM platform in premarket candidate drug screening

Loading

Article metrics loading...

/content/journals/10.1146/annurev-pharmtox-010617-053110
2018-01-06
2024-04-19
Loading full text...

Full text loading...

/deliver/fulltext/pharmtox/58/1/annurev-pharmtox-010617-053110.html?itemId=/content/journals/10.1146/annurev-pharmtox-010617-053110&mimeType=html&fmt=ahah

Literature Cited

  1. Arrowsmith J, Miller P. 1.  2013. Trial watch: phase II and phase III attrition rates 2011–2012. Nat. Rev. Drug Discov. 12:569 [Google Scholar]
  2. 2. US Food Drug Admin. 2015. Reports received and reports entered into FAERS by year. In FDA Adverse Event Reporting System (FAERS) Silver Spring, MD: US Food Drug Admin https://www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Surveillance/AdverseDrugEffects/ucm070434.htm [Google Scholar]
  3. Kuhn M, Letunic I, Jensen LJ, Bork P. 3.  2016. The SIDER database of drugs and side effects. Nucleic Acids Res 44:D1075–79 [Google Scholar]
  4. Magdy T, Burmeister BT, Burridge PW. 4.  2016. Validating the pharmacogenomics of chemotherapy-induced cardiotoxicity: What is missing?. Pharmacol. Ther. 168:113–25 [Google Scholar]
  5. van der Hooft CS, Heeringa J, Brusselle GG, Hofman A, Witteman JC. 5.  et al. 2006. Corticosteroids and the risk of atrial fibrillation. Arch. Intern. Med. 166:1016–20 [Google Scholar]
  6. Wilkinson GS, Baillargeon J, Kuo YF, Freeman JL, Goodwin JS. 6.  2010. Atrial fibrillation and stroke associated with intravenous bisphosphonate therapy in older patients with cancer. J. Clin. Oncol. 28:4898–905 [Google Scholar]
  7. Ray WA, Meredith S, Thapa PB, Hall K, Murray KT. 7.  2004. Cyclic antidepressants and the risk of sudden cardiac death. Clin. Pharmacol. Ther. 75:234–41 [Google Scholar]
  8. Ray WA, Murray KT, Meredith S, Narasimhulu SS, Hall K, Stein CM. 8.  2004. Oral erythromycin and the risk of sudden death from cardiac causes. N. Engl. J. Med. 351:1089–96 [Google Scholar]
  9. Onakpoya IJ, Heneghan CJ, Aronson JK. 9.  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]
  10. Bond CA, Raehl CL. 10.  2006. Adverse drug reactions in United States hospitals. Pharmacotherapy 26:601–8 [Google Scholar]
  11. Gintant G. 11.  2011. An evaluation of hERG current assay performance: translating preclinical safety studies to clinical QT prolongation. Pharmacol. Ther. 129:109–19 [Google Scholar]
  12. Wallis RM. 12.  2010. Integrated risk assessment and predictive value to humans of non-clinical repolarization assays. Br. J. Pharmacol. 159:115–21 [Google Scholar]
  13. Gintant G, Sager PT, Stockbridge N. 13.  2016. Evolution of strategies to improve preclinical cardiac safety testing. Nat. Rev. Drug Discov. 15:457–71 [Google Scholar]
  14. Kramer J, Obejero-Paz CA, Myatt G, Kuryshev YA, Bruening-Wright A. 14.  et al. 2013. MICE models: superior to the HERG model in predicting Torsade de Pointes. Sci. Rep. 3:2100 [Google Scholar]
  15. Mirams GR, Davies MR, Brough SJ, Bridgland-Taylor MH, Cui Y. 15.  et al. 2014. Prediction of Thorough QT study results using action potential simulations based on ion channel screens. J. Pharmacol. Toxicol. Methods 70:246–54 [Google Scholar]
  16. van Opstal JM, Schoenmakers M, Verduyn SC, de Groot SHM, Leunissen JDM. 16.  et al. 2001. Chronic amiodarone evokes no torsade de pointes arrhythmias despite QT lengthening in an animal model of acquired long-QT syndrome. Circulation 104:2722–27 [Google Scholar]
  17. Colatsky T, Fermini B, Gintant G, Pierson JB, Sager P. 17.  et al. 2016. The Comprehensive in Vitro Proarrhythmia Assay (CiPA) initiative—update on progress. J. Pharmacol. Toxicol. Methods 81:15–20 [Google Scholar]
  18. Handy DE, Castro R, Loscalzo J. 18.  2011. Epigenetic modifications: basic mechanisms and role in cardiovascular disease. Circulation 123:2145–56 [Google Scholar]
  19. Loscalzo J, Handy DE. 19.  2014. Epigenetic modifications: basic mechanisms and role in cardiovascular disease (2013 Grover Conference series). Pulmonary Circ 4:169–74 [Google Scholar]
  20. Gallagher MM, Magliano G, Yap YG, Padula M, Morgia V. 20.  et al. 2006. Distribution and prognostic significance of QT intervals in the lowest half centile in 12,012 apparently healthy persons. Am. J. Cardiol. 98:933–35 [Google Scholar]
  21. Ackerman MJ, Mohler PJ. 21.  2010. Defining a new paradigm for human arrhythmia syndromes: phenotypic manifestations of gene mutations in ion channel– and transporter-associated proteins. Circ. Res. 107:457–65 [Google Scholar]
  22. Chen IY, Matsa E, Wu JC. 22.  2016. Induced pluripotent stem cells: at the heart of cardiovascular precision medicine. Nat. Rev. Cardiol. 13:333–49 [Google Scholar]
  23. Arking DE, Pulit SL, Crotti L, van der Harst P, Munroe PB. 23.  et al. 2014. Genetic association study of QT interval highlights role for calcium signaling pathways in myocardial repolarization. Nat. Genet. 46:826–36 [Google Scholar]
  24. Omran J, Firwana B, Koerber S, Bostick B, Alpert MA. 24.  2016. Effect of obesity and weight loss on ventricular repolarization: a systematic review and meta-analysis. Obes. Rev. 17:520–30 [Google Scholar]
  25. Rabkin SW. 25.  2015. Impact of age and sex on QT prolongation in patients receiving psychotropics. Can. J. Psychiatry 60:206–14 [Google Scholar]
  26. McNamara DM, Taylor AL, Tam SW, Worcel M, Yancy CW. 26.  et al. 2014. G-protein beta-3 subunit genotype predicts enhanced benefit of fixed-dose isosorbide dinitrate and hydralazine: results of A-HeFT. JACC Heart Failure 2:551–57 [Google Scholar]
  27. Burridge PW, Matsa E, Shukla P, Lin ZC, Churko JM. 27.  et al. 2014. Chemically defined generation of human cardiomyocytes. Nat. Methods 11:855–60 [Google Scholar]
  28. Mordwinkin NM, Burridge PW, Wu JC. 28.  2013. A review of human pluripotent stem cell-derived cardiomyocytes for high-throughput drug discovery, cardiotoxicity screening, and publication standards. J. Cardiovasc. Transl. Res. 6:22–30 [Google Scholar]
  29. van den Berg CW, Elliott DA, Braam SR, Mummery CL, Davis RP. 29.  2016. Differentiation of human pluripotent stem cells to cardiomyocytes under defined conditions. Methods Mol. Biol. 1353:163–80 [Google Scholar]
  30. Aguilar JS, Begum AN, Alvarez J, Zhang XB, Hong Y, Hao J. 30.  2015. Directed cardiomyogenesis of human pluripotent stem cells by modulating Wnt/β-catenin and BMP signalling with small molecules. Biochem. J. 469:235–41 [Google Scholar]
  31. Burridge PW, Holmström A, Wu JC. 31.  2015. Chemically defined culture and cardiomyocyte differentiation of human pluripotent stem cells. Curr. Protocols Hum. Genet. 87:21.3.1–15 [Google Scholar]
  32. Minami I, Yamada K, Otsuji TG, Yamamoto T, Shen Y. 32.  et al. 2012. A small molecule that promotes cardiac differentiation of human pluripotent stem cells under defined, cytokine- and xeno-free conditions. Cell Rep 2:1448–60 [Google Scholar]
  33. Willems E, Cabral-Teixeira J, Schade D, Cai W, Reeves P. 33.  et al. 2012. Small molecule-mediated TGF-β type II receptor degradation promotes cardiomyogenesis in embryonic stem cells. Cell Stem Cell 11:242–52 [Google Scholar]
  34. Zhang M, Schulte JS, Heinick A, Piccini I, Rao J. 34.  et al. 2015. Universal cardiac induction of human pluripotent stem cells in two and three-dimensional formats: implications for in vitro maturation. Stem Cells 33:1456–69 [Google Scholar]
  35. Kehat I, Kenyagin-Karsenti D, Snir M, Segev H, Amit M. 35.  et al. 2001. Human embryonic stem cells can differentiate into myocytes with structural and functional properties of cardiomyocytes. J. Clin. Investig. 108:407–14 [Google Scholar]
  36. Yao S, Chen S, Clark J, Hao E, Beattie GM. 36.  et al. 2006. Long-term self-renewal and directed differentiation of human embryonic stem cells in chemically defined conditions. PNAS 103:6907–12 [Google Scholar]
  37. Burridge PW, Anderson D, Priddle H, Barbadillo Muñoz MD, Chamberlain S. 37.  et al. 2007. Improved human embryonic stem cell embryoid body homogeneity and cardiomyocyte differentiation from a novel V-96 plate aggregation system highlights interline variability. Stem Cells 25:929–38 [Google Scholar]
  38. Yang L, Soonpaa MH, Adler ED, Roepke TK, Kattman SJ. 38.  et al. 2008. Human cardiovascular progenitor cells develop from a KDR+ embryonic-stem-cell-derived population. Nature 453:524–28 [Google Scholar]
  39. Laflamme MA, Chen KY, Naumova AV, Muskheli V, Fugate JA. 39.  et al. 2007. Cardiomyocytes derived from human embryonic stem cells in pro-survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25:1015–24 [Google Scholar]
  40. Uosaki H, Fukushima H, Takeuchi A, Matsuoka S, Nakatsuji N. 40.  et al. 2011. Efficient and scalable purification of cardiomyocytes from human embryonic and induced pluripotent stem cells by VCAM1 surface expression. PLOS ONE 6:e23657 [Google Scholar]
  41. Gonzalez R, Lee JW, Schultz PG. 41.  2011. Stepwise chemically induced cardiomyocyte specification of human embryonic stem cells. Angew. Chem. Int. Ed. Engl. 50:11181–85 [Google Scholar]
  42. Lian X, Hsiao C, Wilson G, Zhu K, Hazeltine LB. 42.  et al. 2012. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. PNAS 109:E1848–57 [Google Scholar]
  43. Xu XQ, Graichen R, Soo SY, Balakrishnan T, Rahmat SN. 43.  et al. 2008. Chemically defined medium supporting cardiomyocyte differentiation of human embryonic stem cells. Differentiation 76:958–70 [Google Scholar]
  44. Burridge PW, Thompson S, Millrod MA, Weinberg S, Yuan X. 44.  et al. 2011. A universal system for highly efficient cardiac differentiation of human induced pluripotent stem cells that eliminates interline variability. PLOS ONE 6:e18293 [Google Scholar]
  45. Chong JJ, Yang X, Don CW, Minami E, Liu YW. 45.  et al. 2014. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510:273–77 [Google Scholar]
  46. Chen G, Gulbranson DR, Hou Z, Bolin JM, Ruotti V. 46.  et al. 2011. Chemically defined conditions for human iPSC derivation and culture. Nat. Methods 8:424–29 [Google Scholar]
  47. Kempf H, Andree B, Zweigerdt R. 47.  2016. Large-scale production of human pluripotent stem cell derived cardiomyocytes. Adv. Drug Deliv. Rev. 96:18–30 [Google Scholar]
  48. Lam AT, Chen AK, Ting SQ, Reuveny S, Oh SK. 48.  2016. Integrated processes for expansion and differentiation of human pluripotent stem cells in suspended microcarriers cultures. Biochem. Biophys. Res. Commun. 473:764–68 [Google Scholar]
  49. Lecina M, Ting S, Choo A, Reuveny S, Oh S. 49.  2010. Scalable platform for human embryonic stem cell differentiation to cardiomyocytes in suspended microcarrier cultures. Tissue Eng. Part C Methods 16:1609–19 [Google Scholar]
  50. Bizy A, Guerrero-Serna G, Hu B, Ponce-Balbuena D, Willis BC. 50.  et al. 2013. Myosin light chain 2-based selection of human iPSC-derived early ventricular cardiac myocytes. Stem Cell Res 11:1335–47 [Google Scholar]
  51. Josowitz R, Lu J, Falce C, D'Souza SL, Wu M. 51.  et al. 2014. Identification and purification of human induced pluripotent stem cell-derived atrial-like cardiomyocytes based on sarcolipin expression. PLOS ONE 9:e101316 [Google Scholar]
  52. Zhu WZ, Xie Y, Moyes KW, Gold JD, Askari B, Laflamme MA. 52.  2010. Neuregulin/ErbB signaling regulates cardiac subtype specification in differentiating human embryonic stem cells. Circ. Res. 107:776–86 [Google Scholar]
  53. Zhang Q, Jiang J, Han P, Yuan Q, Zhang J. 53.  et al. 2011. Direct differentiation of atrial and ventricular myocytes from human embryonic stem cells by alternating retinoid signals. Cell Res 21:579–87 [Google Scholar]
  54. Devalla HD, Schwach V, Ford JW, Milnes JT, El-Haou S. 54.  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]
  55. Smith AS, Macadangdang J, Leung W, Laflamme MA, Kim DH. 55.  2017. Human iPSC-derived cardiomyocytes and tissue engineering strategies for disease modeling and drug screening. Biotechnol. Adv. 35:77–94 [Google Scholar]
  56. Hwang HS, Kryshtal DO, Feaster TK, Sanchez-Freire V, Zhang J. 56.  et al. 2015. Human induced pluripotent stem cell (hiPSC) derived cardiomyocytes to understand and test cardiac calcium handling: a glass half full. J. Mol. Cell. Cardiol. 89:379–80 [Google Scholar]
  57. Yang X, Pabon L, Murry CE. 57.  2014. Engineering adolescence: maturation of human pluripotent stem cell–derived cardiomyocytes. Circ. Res. 114:511–23 [Google Scholar]
  58. Denning C, Borgdorff V, Crutchley J, Firth KS, George V. 58.  et al. 2016. Cardiomyocytes from human pluripotent stem cells: from laboratory curiosity to industrial biomedical platform. Biochim. Biophys. Acta 1863:1728–48 [Google Scholar]
  59. Zhu R, Blazeski A, Poon E, Costa KD, Tung L, Boheler KR. 59.  2014. Physical developmental cues for the maturation of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res. Ther. 5:117 [Google Scholar]
  60. Veerman CC, Kosmidis G, Mummery CL, Casini S, Verkerk AO, Bellin M. 60.  2015. Immaturity of human stem-cell-derived cardiomyocytes in culture: fatal flaw or soluble problem?. Stem Cells Dev 24:1035–52 [Google Scholar]
  61. Robertson C, Tran DD, George SC. 61.  2013. Concise review: maturation phases of human pluripotent stem cell-derived cardiomyocytes. Stem Cells 31:829–37 [Google Scholar]
  62. Keung W, Boheler KR, Li RA. 62.  2014. Developmental cues for the maturation of metabolic, electrophysiological and calcium handling properties of human pluripotent stem cell-derived cardiomyocytes. Stem Cell Res. Ther. 5:17 [Google Scholar]
  63. Bedada FB, Wheelwright M, Metzger JM. 63.  2016. Maturation status of sarcomere structure and function in human iPSC-derived cardiac myocytes. Biochim. Biophys. Acta 1863:1829–38 [Google Scholar]
  64. Bedada FB, Chan SSK, Metzger SK, Zhang L, Zhang J. 64.  et al. 2014. Acquisition of a quantitative, stoichiometrically conserved ratiometric marker of maturation status in stem cell-derived cardiac myocytes. Stem Cell Rep 3:594–605 [Google Scholar]
  65. Liang P, Lan F, Lee AS, Gong T, Sanchez-Freire V. 65.  et al. 2013. Drug screening using a library of human induced pluripotent stem cell-derived cardiomyocytes reveals disease-specific patterns of cardiotoxicity. Circulation 127:1677–91 [Google Scholar]
  66. Lieu DK, Fu JD, Chiamvimonvat N, Tung KC, McNerney GP. 66.  et al. 2013. Mechanism-based facilitated maturation of human pluripotent stem cell-derived cardiomyocytes. Circ. Arrhythmia Electrophysiol. 6:191–201 [Google Scholar]
  67. Rao C, Prodromakis T, Kolker L, Chaudhry UA, Trantidou T. 67.  et al. 2013. The effect of microgrooved culture substrates on calcium cycling of cardiac myocytes derived from human induced pluripotent stem cells. Biomaterials 34:2399–411 [Google Scholar]
  68. Germanguz I, Sedan O, Zeevi-Levin N, Shtrichman R, Barak E. 68.  et al. 2011. Molecular characterization and functional properties of cardiomyocytes derived from human inducible pluripotent stem cells. J. Cell. Mol. Med. 15:38–51 [Google Scholar]
  69. Lundy SD, Zhu WZ, Regnier M, Laflamme MA. 69.  2013. Structural and functional maturation of cardiomyocytes derived from human pluripotent stem cells. Stem Cells Dev 22:1991–2002 [Google Scholar]
  70. Jung G, Fajardo G, Ribeiro AJ, Kooiker KB, Coronado M. 70.  et al. 2016. Time-dependent evolution of functional versus remodeling signaling in induced pluripotent stem cell-derived cardiomyocytes and induced maturation with biomechanical stimulation. FASEB J 30:1464–79 [Google Scholar]
  71. Cho GS, Lee DI, Tampakakis E, Murphy S, Andersen P. 71.  et al. 2017. Neonatal transplantation confers maturation of PSC-derived cardiomyocytes conducive to modeling cardiomyopathy. Cell Rep 18:571–82 [Google Scholar]
  72. Kadota S, Pabon L, Reinecke H, Murry CE. 72.  2017. In vivo maturation of human induced pluripotent stem cell-derived cardiomyocytes in neonatal and adult rat hearts. Stem Cell Rep 8:278–89 [Google Scholar]
  73. Nunes SS, Miklas JW, Liu J, Aschar-Sobbi R, Xiao Y. 73.  et al. 2013. Biowire: a platform for maturation of human pluripotent stem cell–derived cardiomyocytes. Nat. Methods 10:781–87 [Google Scholar]
  74. Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M. 74.  et al. 2011. Growth of engineered human myocardium with mechanical loading and vascular coculture. Circ. Res. 109:47–59 [Google Scholar]
  75. Ruan JL, Tulloch NL, Saiget M, Paige SL, Razumova MV. 75.  et al. 2015. Mechanical stress promotes maturation of human myocardium from pluripotent stem cell-derived progenitors. Stem Cells 33:2148–57 [Google Scholar]
  76. Carson D, Hnilova M, Yang X, Nemeth CL, Tsui JH. 76.  et al. 2016. Nanotopography-induced structural anisotropy and sarcomere development in human cardiomyocytes derived from induced pluripotent stem cells. ACS Appl. Mater. Interfaces 8:21923–32 [Google Scholar]
  77. Ribeiro AJ, Ang YS, Fu JD, Rivas RN, Mohamed TM. 77.  et al. 2015. Contractility of single cardiomyocytes differentiated from pluripotent stem cells depends on physiological shape and substrate stiffness. PNAS 112:12705–10 [Google Scholar]
  78. Tiburcy M, Hudson JE, Balfanz P, Schlick SF, Meyer T. 78.  et al. 2017. Defined engineered human myocardium with advanced maturation for applications in heart failure modelling and repair. Circulation 135:1832–47 [Google Scholar]
  79. Kim C, Wong J, Wen J, Wang S, Wang C. 79.  et al. 2013. Studying arrhythmogenic right ventricular dysplasia with patient-specific iPSCs. Nature 494:105–10 [Google Scholar]
  80. Yang X, Rodriguez M, Pabon L, Fischer KA, Reinecke H. 80.  et al. 2014. Tri-iodo-l-thyronine promotes the maturation of human cardiomyocytes-derived from induced pluripotent stem cells. J. Mol. Cell. Cardiol. 72:296–304 [Google Scholar]
  81. Ivashchenko CY, Pipes GC, Lozinskaya IM, Lin Z, Xiaoping X. 81.  et al. 2013. Human-induced pluripotent stem cell-derived cardiomyocytes exhibit temporal changes in phenotype. Am. J. Physiol. Heart Circ. Physiol. 305:H913–22 [Google Scholar]
  82. Kosmidis G, Bellin M, Ribeiro MC, van Meer B, Ward-van Oostwaard D. 82.  et al. 2015. Altered calcium handling and increased contraction force in human embryonic stem cell derived cardiomyocytes following short term dexamethasone exposure. Biochem. Biophys. Res. Commun. 467:998–1005 [Google Scholar]
  83. Wen JY, Wei CY, Shah K, Wong J, Wang C, Chen HS. 83.  2015. Maturation-based model of arrhythmogenic right ventricular dysplasia using patient-specific induced pluripotent stem cells. Circ. J. 79:1402–8 [Google Scholar]
  84. Birket MJ, Ribeiro MC, Kosmidis G, Ward D, Leitoguinho AR. 84.  et al. 2015. Contractile defect caused by mutation in MYBPC3 revealed under conditions optimized for human PSC-cardiomyocyte function. Cell Rep 13:733–45 [Google Scholar]
  85. Xu C, He JQ, Kamp TJ, Police S, Hao X. 85.  et al. 2006. Human embryonic stem cell-derived cardiomyocytes can be maintained in defined medium without serum. Stem Cells Dev 15:931–41 [Google Scholar]
  86. Földes G, Matsa E, Kriston-Vizi J, Leja T, Amisten S. 86.  et al. 2014. Aberrant α-adrenergic hypertrophic response in cardiomyocytes from human induced pluripotent cells. Stem Cell Rep 3:905–14 [Google Scholar]
  87. Fu JD, Rushing SN, Lieu DK, Chan CW, Kong CW. 87.  et al. 2011. Distinct roles of microRNA-1 and -499 in ventricular specification and functional maturation of human embryonic stem cell-derived cardiomyocytes. PLOS ONE 6:e27417 [Google Scholar]
  88. Kuppusamy KT, Jones DC, Sperber H, Madan A, Fischer KA. 88.  et al. 2015. Let-7 family of microRNA is required for maturation and adult-like metabolism in stem cell-derived cardiomyocytes. PNAS 112:E2785–94 [Google Scholar]
  89. Rana P, Anson B, Engle S, Will Y. 89.  2012. Characterization of human-induced pluripotent stem cell–derived cardiomyocytes: bioenergetics and utilization in safety screening. Toxicol. Sci. 130:117–31 [Google Scholar]
  90. Wang G, McCain ML, Yang L, He A, Pasqualini FS. 90.  et al. 2014. Modeling the mitochondrial cardiomyopathy of Barth syndrome with induced pluripotent stem cell and heart-on-chip technologies. Nat. Med. 20:616–23 [Google Scholar]
  91. Poon E, Keung W, Liang Y, Ramalingam R, Yan B. 91.  et al. 2015. Proteomic analysis of human pluripotent stem cell-derived, fetal, and adult ventricular cardiomyocytes reveals pathways crucial for cardiac metabolism and maturation. Circ. Cardiovasc. Genet. 8:427–36 [Google Scholar]
  92. Marroquin LD, Hynes J, Dykens JA, Jamieson JD, Will Y. 92.  2007. Circumventing the Crabtree effect: replacing media glucose with galactose increases susceptibility of HepG2 cells to mitochondrial toxicants. Toxicol. Sci. 97:539–47 [Google Scholar]
  93. Itzhaki I, Maizels L, Huber I, Zwi-Dantsis L, Caspi O. 93.  et al. 2011. Modelling the long QT syndrome with induced pluripotent stem cells. Nature 471:225–29 [Google Scholar]
  94. Lan F, Lee AS, Liang P, Sanchez-Freire V, Nguyen PK. 94.  et al. 2013. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 12:101–13 [Google Scholar]
  95. Sun N, Yazawa M, Liu J, Han L, Sanchez-Freire V. 95.  et al. 2012. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci. Transl. Med. 4:130ra47 [Google Scholar]
  96. Sharma A, Burridge PW, McKeithan WL, Serrano R, Shukla P. 96.  et al. 2017. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci. Transl. Med. 9:eaaf2584 [Google Scholar]
  97. Burridge PW, Li YF, Matsa E, Wu H, Ong SG. 97.  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]
  98. Crivat G, Taraska JW. 98.  2012. Imaging proteins inside cells with fluorescent tags. Trends Biotechnol 30:8–16 [Google Scholar]
  99. Rajamohan D, Kalra S, Duc Hoang M, George V, Staniforth A. 99.  et al. 2016. Automated electrophysiological and pharmacological evaluation of human pluripotent stem cell-derived cardiomyocytes. Stem Cells Dev 25:439–52 [Google Scholar]
  100. Navarrete EG, Liang P, Lan F, Sanchez-Freire V, Simmons C. 100.  et al. 2013. Screening drug-induced arrhythmia using human induced pluripotent stem cell–derived cardiomyocytes and low-impedance microelectrode arrays. Circulation 128:S3–13 [Google Scholar]
  101. Helassa N, Zhang XH, Conte I, Scaringi J, Esposito E. 101.  et al. 2015. Fast-response calmodulin-based fluorescent indicators reveal rapid intracellular calcium dynamics. Sci. Rep. 5:15978 [Google Scholar]
  102. Chen TW, Wardill TJ, Sun Y, Pulver SR, Renninger SL. 102.  et al. 2013. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499:295–300 [Google Scholar]
  103. Cerignoli F, Charlot D, Whittaker R, Ingermanson R, Gehalot P. 103.  et al. 2012. High throughput measurement of Ca2+ dynamics for drug risk assessment in human stem cell-derived cardiomyocytes by kinetic image cytometry. J. Pharmacol. Toxicol. Methods 66:246–56 [Google Scholar]
  104. Roden DM. 104.  2008. Cellular basis of drug-induced torsades de pointes. Br. J. Pharmacol. 154:1502–7 [Google Scholar]
  105. Lester RM, Olbertz J. 105.  2016. Early drug development: assessment of proarrhythmic risk and cardiovascular safety. Expert Rev. Clin. Pharmacol. 9:1611–18 [Google Scholar]
  106. Kramer DB, Zimetbaum PJ. 106.  2011. Long-QT syndrome. Cardiol. Rev. 19:217–25 [Google Scholar]
  107. Cross MJ, Berridge BR, Clements PJ, Cove-Smith L, Force TL. 107.  et al. 2015. Physiological, pharmacological and toxicological considerations of drug-induced structural cardiac injury. Br. J. Pharmacol. 172:957–74 [Google Scholar]
  108. Varga ZV, Ferdinandy P, Liaudet L, Pacher P. 108.  2015. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am. J. Physiol. Heart Circ. Physiol. 309:H1453–67 [Google Scholar]
  109. Mercurio V, Pirozzi F, Lazzarini E, Marone G, Rizzo P. 109.  et al. 2016. Models of heart failure based on the cardiotoxicity of anticancer drugs. J. Card. Fail. 22:449–58 [Google Scholar]
  110. Hanna AD, Lam A, Tham S, Dulhunty AF, Beard NA. 110.  2014. Adverse effects of doxorubicin and its metabolic product on cardiac RyR2 and SERCA2A. Mol. Pharmacol. 86:438–49 [Google Scholar]
  111. Sag CM, Kohler AC, Anderson ME, Backs J, Maier LS. 111.  2011. CaMKII-dependent SR Ca leak contributes to doxorubicin-induced impaired Ca handling in isolated cardiac myocytes. J. Mol. Cell. Cardiol. 51:749–59 [Google Scholar]
  112. Kim SY, Kim SJ, Kim BJ, Rah SY, Chung SM. 112.  et al. 2006. Doxorubicin-induced reactive oxygen species generation and intracellular Ca2+ increase are reciprocally modulated in rat cardiomyocytes. Exp. Mol. Med. 38:535–45 [Google Scholar]
  113. Wang GX, Wang YX, Zhou XB, Korth M. 113.  2001. Effects of doxorubicinol on excitation–contraction coupling in guinea pig ventricular myocytes. Eur. J. Pharmacol. 423:99–107 [Google Scholar]
  114. Rainer PP, Doleschal B, Kirk JA, Sivakumaran V, Saad Z. 114.  et al. 2012. Sunitinib causes dose-dependent negative functional effects on myocardium and cardiomyocytes. BJU Int 110:1455–62 [Google Scholar]
  115. Octavia Y, Tocchetti CG, Gabrielson KL, Janssens S, Crijns HJ, Moens AL. 115.  2012. Doxorubicin-induced cardiomyopathy: from molecular mechanisms to therapeutic strategies. J. Mol. Cell Cardiol. 52:1213–25 [Google Scholar]
  116. Doherty KR, Wappel RL, Talbert DR, Trusk PB, Moran DM. 116.  et al. 2013. Multi-parameter in vitro toxicity testing of crizotinib, sunitinib, erlotinib, and nilotinib in human cardiomyocytes. Toxicol. Appl. Pharmacol. 272:245–55 [Google Scholar]
  117. Ueno M, Kakinuma Y, Yuhki K, Murakoshi N, Iemitsu M. 117.  et al. 2006. Doxorubicin induces apoptosis by activation of caspase-3 in cultured cardiomyocytes in vitro and rat cardiac ventricles in vivo. J. Pharmacol. Sci. 101:151–58 [Google Scholar]
  118. Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC. 118.  et al. 2007. Topoisomerase IIβ–mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res 67:8839–46 [Google Scholar]
  119. Anwar A, Anwar IJ, Delafontaine P. 119.  2015. Elevation of cardiovascular risk by non-steroidal anti-inflammatory drugs. Trends Cardiovasc. Med. 25:726–35 [Google Scholar]
  120. Li Z, Hu S, Ghosh Z, Han Z, Wu JC. 120.  2011. Functional characterization and expression profiling of human induced pluripotent stem cell- and embryonic stem cell-derived endothelial cells. Stem Cells Dev 20:1701–10 [Google Scholar]
  121. White MP, Rufaihah AJ, Liu L, Ghebremariam YT, Ivey KN. 121.  et al. 2013. Limited gene expression variation in human embryonic stem cell and induced pluripotent stem cell-derived endothelial cells. Stem Cells 31:92–103 [Google Scholar]
  122. Bhattacharyya S, Schapira AH, Mikhailidis DP, Davar J. 122.  2009. Drug-induced fibrotic valvular heart disease. Lancet 374:577–85 [Google Scholar]
  123. Cosyns B, Droogmans S, Rosenhek R, Lancellotti P. 123.  2013. Drug-induced valvular heart disease. Heart 99:7–12 [Google Scholar]
  124. Fitzgerald LW, Burn TC, Brown BS, Patterson JP, Corjay MH. 124.  et al. 2000. Possible role of valvular serotonin 5-HT2B receptors in the cardiopathy associated with fenfluramine. Mol. Pharmacol. 57:75–81 [Google Scholar]
  125. Nebigil CG, Launay JM, Hickel P, Tournois C, Maroteaux L. 125.  2000. 5-Hydroxytryptamine 2B receptor regulates cell-cycle progression: cross-talk with tyrosine kinase pathways. PNAS 97:2591–96 [Google Scholar]
  126. Rothman RB, Baumann MH, Savage JE, Rauser L, McBride A. 126.  et al. 2000. Evidence for possible involvement of 5-HT2B receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medications. Circulation 102:2836–41 [Google Scholar]
  127. Babiarz JE, Ravon M, Sridhar S, Ravindran P, Swanson B. 127.  et al. 2012. Determination of the human cardiomyocyte mRNA and miRNA differentiation network by fine-scale profiling. Stem Cells Dev 21:1956–65 [Google Scholar]
  128. Ma J, Guo L, Fiene SJ, Anson BD, Thomson JA. 128.  et al. 2011. High purity human-induced pluripotent stem cell-derived cardiomyocytes: electrophysiological properties of action potentials and ionic currents. Am. J. Physiol. Heart Circ. Physiol. 301:H2006–17 [Google Scholar]
  129. Puppala D, Collis LP, Sun SZ, Bonato V, Chen X. 129.  et al. 2013. Comparative gene expression profiling in human-induced pluripotent stem cell–derived cardiocytes and human and cynomolgus heart tissue. Toxicol. Sci. 131:292–301 [Google Scholar]
  130. Malan D, Zhang M, Stallmeyer B, Müller J, Fleischmann BK. 130.  et al. 2016. Human iPS cell model of type 3 long QT syndrome recapitulates drug-based phenotype correction. Basic Res. Cardiol. 111:14 [Google Scholar]
  131. Carvajal-Vergara X, Sevilla A, D'Souza SL, Ang Y-S, Schaniel C. 131.  et al. 2010. Patient-specific induced pluripotent stem cell derived models of LEOPARD syndrome. Nature 465:808–12 [Google Scholar]
  132. Yazawa M, Hsueh B, Jia X, Pasca AM, Bernstein JA. 132.  et al. 2011. Using iPS cells to investigate cardiac phenotypes in patients with Timothy Syndrome. Nature 471:230–34 [Google Scholar]
  133. Drawnel FM, Boccardo S, Prummer M, Delobel F, Graff A. 133.  et al. 2014. Disease modeling and phenotypic drug screening for diabetic cardiomyopathy using human induced pluripotent stem cells. Cell Rep 9:810–21 [Google Scholar]
/content/journals/10.1146/annurev-pharmtox-010617-053110
Loading
/content/journals/10.1146/annurev-pharmtox-010617-053110
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