Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Use of human induced pluripotent stem cell–derived cardiomyocytes to assess drug cardiotoxicity

Abstract

Cardiotoxicity has historically been a major cause of drug removal from the pharmaceutical market. Several chemotherapeutic compounds have been noted for their propensities to induce dangerous cardiac-specific side effects such as arrhythmias or cardiomyocyte apoptosis. However, improved preclinical screening methodologies have enabled cardiotoxic compounds to be identified earlier in the drug development pipeline. Human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs) can be used to screen for drug-induced alterations in cardiac cellular contractility, electrophysiology, and viability. We previously established a novel ‘cardiac safety index’ (CSI) as a metric that can evaluate potential cardiotoxic drugs via high-throughput screening of hiPSC-CMs. This metric quantitatively examines drug-induced alterations in CM function, using several in vitro readouts, and normalizes the resulting toxicity values to the in vivo maximum drug blood plasma concentration seen in preclinical or clinical pharmacokinetic models. In this ~1-month-long protocol, we describe how to differentiate hiPSCs into hiPSC-CMs and subsequently implement contractility and cytotoxicity assays that can evaluate drug-induced cardiotoxicity in hiPSC-CMs. We also describe how to carry out the calculations needed to generate the CSI metric from these quantitative toxicity measurements.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Workflow for generating the cardiac safety index for drugs of interest using high-throughput contractility and cytotoxicity analysis of hiPSC-CMs.
Fig. 2: Procedure for chemically defined differentiation of hiPSCs into hiPSC-CMs and downstream replating.
Fig. 3: Setup for drug-induced cytotoxicity and contractility assessment of hiPSC-CMs.
Fig. 4: Equations used to obtain the CSI.
Fig. 5: Mathematical example of CSI calculations for cardiotoxic tyrosine kinase inhibitor sorafenib.

Similar content being viewed by others

Data availability

Source data are provided or are available from the corresponding author upon request.

References

  1. Chang, H. M., Okwuosa, T. M., Scarabelli, T., Moudgil, R. & Yeh, E. T. H. Cardiovascular complications of cancer therapy: best practices in diagnosis, prevention, and management: part 2. J. Am. Coll. Cardiol. 70, 2552–2565 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  2. Chang, H. M., Moudgil, R., Scarabelli, T., Okwuosa, T. M. & Yeh, E. T. H. Cardiovascular complications of cancer therapy: best practices in diagnosis, prevention, and management: part 1. J. Am. Coll. Cardiol. 70, 2536–2551 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  3. Force, T., Krause, D. S. & Van Etten, R. A. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat. Rev. Cancer 7, 332–344 (2007).

    Article  CAS  PubMed  Google Scholar 

  4. Moslehi, J. J. & Deininger, M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J. Clin. Oncol. 33, 4210–4218 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Wu, A. H. Cardiotoxic drugs: clinical monitoring and decision making. Heart 94, 1503–1509 (2008).

    Article  CAS  PubMed  Google Scholar 

  6. Magdy, T., Schuldt, A. J. T., Wu, J. C., Bernstein, D. & Burridge, P. W. Human induced pluripotent stem cell (hiPSC)-derived cells to assess drug cardiotoxicity: opportunities and problems. Annu. Rev. Pharmacol. Toxicol. 58, 83–103 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Ferri, N. et al. Drug attrition during pre-clinical and clinical development: understanding and managing drug-induced cardiotoxicity. Pharmacol. Ther. 138, 470–484 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Stack, J. P., Moslehi, J., Sayed, N. & Wu, J. C. Cancer therapy-induced cardiomyopathy: can human induced pluripotent stem cell modelling help prevent it? Eur. Heart J. https://doi.org/10.1093/eurheartj/ehx811 (2018).

  9. Colatsky, T. et al. The comprehensive in vitro proarrhythmia assay (CiPA) initiative—update on progress. J. Pharmacol. Toxicol. Methods 81, 15–20 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Sharma, A. et al. High-throughput screening of tyrosine kinase inhibitor cardiotoxicity with human induced pluripotent stem cells. Sci. Transl. Med. 9, eaaf2584 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  11. Mercola, M., Colas, A. & Willems, E. Induced pluripotent stem cells in cardiovascular drug discovery. Circ. Res. 112, 534–548 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  12. Guth, B. D. Preclinical cardiovascular risk assessment in modern drug development. Toxicol. Sci. 97, 4–20 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Hoffmann, P. & Warner, B. Are hERG channel inhibition and QT interval prolongation all there is in drug-induced torsadogenesis? A review of emerging trends. J. Pharmacol. Toxicol. Methods 53, 87–105 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Mitcheson, J. S., Hancox, J. C. & Levi, A. J. Cultured adult cardiac myocytes: future applications, culture methods, morphological and electrophysiological properties. Cardiovasc. Res. 39, 280–300 (1998).

    Article  CAS  PubMed  Google Scholar 

  15. Sharma, A., Wu, J. C. & Wu, S. M. Induced pluripotent stem cell-derived cardiomyocytes for cardiovascular disease modeling and drug screening. Stem Cell Res. Ther. 4, 150 (2013).

    Article  PubMed Central  PubMed  Google Scholar 

  16. Garg, P. et al. Genome editing of induced pluripotent stem cells to decipher cardiac channelopathy variant. J. Am. Coll. Cardiol. 72, 62–75 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  17. Ma, N. et al. Determining the pathogenicity of a genomic variant of uncertain significance using CRISPR/Cas9 and human-induced pluripotent stem cells. Circulation https://doi.org/10.1161/CIRCULATIONAHA.117.032273 (2018).

  18. Burridge, P. W., Sharma, A. & Wu, J. C. Genetic and epigenetic regulation of human cardiac reprogramming and differentiation in regenerative medicine. Annu. Rev. Genet. 49, 461–484 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. Burridge, P. W. et al. Chemically defined generation of human cardiomyocytes. Nat. Methods 11, 855–860 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Sharma, A. et al. CRISPR/Cas9-mediated fluorescent tagging of endogenous proteins in human pluripotent stem cells. Curr. Protoc. Hum. Genet. 96, 21.11.1–21.11.20 (2018).

    Article  Google Scholar 

  21. Sharma, A. et al. Human induced pluripotent stem cell-derived cardiomyocytes as an in vitro model for coxsackievirus B3-induced myocarditis and antiviral drug screening platform. Circ. Res. 115, 556–566 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Liang, P. et al. Patient-specific and genome-edited induced pluripotent stem cell-derived cardiomyocytes elucidate single-cell phenotype of Brugada syndrome. J. Am. Coll. Cardiol. 68, 2086–2096 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  23. Itzhaki, I. et al. Modelling the long QT syndrome with induced pluripotent stem cells. Nature 471, 225–229 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Sun, N. et al. Patient-specific induced pluripotent stem cells as a model for familial dilated cardiomyopathy. Sci. Transl. Med. 4, 130ra147 (2012).

    Article  Google Scholar 

  25. Wu, H. et al. Epigenetic regulation of phosphodiesterases 2A and 3A underlies compromised beta-adrenergic signaling in an iPSC model of dilated cardiomyopathy. Cell Stem Cell 17, 89–100 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Lan, F. et al. Abnormal calcium handling properties underlie familial hypertrophic cardiomyopathy pathology in patient-specific induced pluripotent stem cells. Cell Stem Cell 12, 101–113 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Burridge, P. W. et al. Human induced pluripotent stem cell-derived cardiomyocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat. Med. 22, 547–556 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  28. Kodo, K. et al. iPSC-derived cardiomyocytes reveal abnormal TGF-beta signalling in left ventricular non-compaction cardiomyopathy. Nat. Cell Biol. 18, 1031–1042 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Navarrete, E. G. et al. Screening drug-induced arrhythmia [corrected] using human induced pluripotent stem cell-derived cardiomyocytes and low-impedance microelectrode arrays. Circulation 128, S3–S13 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Tohyama, S. et al. Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 12, 127–137 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. McKeithan, W. L. et al. An automated platform for assessment of congenital and drug-induced arrhythmia with hiPSC-derived cardiomyocytes. Front. Physiol. 8, 766 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  32. Millard, D. et al. Cross-site reliability of human induced pluripotent stem-cell derived cardiomyocyte based safety assays using microelectrode arrays: results from a blinded CiPA pilot study. Toxicol. Sci. 164, 550-562 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  33. Ando, H. et al. A new paradigm for drug-induced torsadogenic risk assessment using human iPS cell-derived cardiomyocytes. J. Pharmacol. Toxicol. Methods 84, 111–127 (2017).

    Article  CAS  PubMed  Google Scholar 

  34. Blinova, K. et al. Comprehensive translational assessment of human-induced pluripotent stem cell derived cardiomyocytes for evaluating drug-induced arrhythmias. Toxicol. Sci. 155, 234–247 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Churko, J. M. et al. Transcriptomic and epigenomic differences in human induced pluripotent stem cells generated from six reprogramming methods. Nat. Biomed. Eng. 1, 826–837 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  36. Riss, T. L. et al. in Assay Guidance Manual (eds. Sittampalam, G. S. et al.) https://www.ncbi.nlm.nih.gov/books/NBK53196/ (2004).

  37. Adrian, R. J. & Westerweel, J. Particle Image Velocimetry (Cambridge University Press, Cambridge, UK, 2011).

  38. Del Alamo, J. C. et al. Spatio-temporal analysis of eukaryotic cell motility by improved force cytometry. Proc. Natl. Acad. Sci. USA 104, 13343–13348 (2007).

    Article  PubMed Central  PubMed  Google Scholar 

  39. Serrano, R., Aung, A. & Varghese, S. & del Alamo, J. C. Three-dimensional monolayer stress cytometry. Biophys. J. 112, 271a (2017).

    Article  Google Scholar 

  40. Banerjee, I. et al. Cyclic stretch of embryonic cardiomyocytes increases proliferation, growth, and expression while repressing Tgf-beta signaling. J. Mol. Cell. Cardiol. 79, 133–144 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Del Alamo, J. C. et al. High throughput physiological screening of iPSC-derived cardiomyocytes for drug development. Biochim. Biophys. Acta 1863, 1717–1727 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  42. Taylor, Z. J., Gurka, R., Kopp, G. A. & Liberzon, A. Long-duration time-resolved PIV to study unsteady aerodynamics. IEEE Trans. Instrum. Meas. 59, 3262–3269 (2010).

    Article  Google Scholar 

  43. Smith, D. A., Di, L. & Kerns, E. H. The effect of plasma protein binding on in vivo efficacy: misconceptions in drug discovery. Nat. Rev. Drug Discov. 9, 929–939 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Veerman, C. C. et al. Immaturity of human stem-cell-derived cardiomyocytes in culture: fatal flaw or soluble problem? Stem Cells Dev. 24, 1035–1052 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Churko, J. M., Burridge, P. W. & Wu, J. C. Generation of human iPSCs from human peripheral blood mononuclear cells using non-integrative Sendai virus in chemically defined conditions. Methods Mol. Biol. 1036, 81–88 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  46. Watanabe, K. et al. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol. 25, 681–686 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Pei, F. et al. Chemical-defined and albumin-free generation of human atrial and ventricular myocytes from human pluripotent stem cells. Stem Cell Res. 19, 94–103 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Lian, X. et al. Chemically defined, albumin-free human cardiomyocyte generation. Nat. Methods 12, 595–596 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Huo, J. et al. Evaluation of batch variations in induced pluripotent stem cell-derived human cardiomyocytes from 2 major suppliers. Toxicol. Sci. 156, 25–38 (2017).

    CAS  PubMed  Google Scholar 

  50. Hasinoff, B. B. & Patel, D. Mechanisms of myocyte cytotoxicity induced by the multikinase inhibitor sorafenib. Cardiovasc. Toxicol. 10, 1–8 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Moore, M. et al. Phase I study to determine the safety and pharmacokinetics of the novel Raf kinase and VEGFR inhibitor BAY 43-9006, administered for 28 days on/7 days off in patients with advanced, refractory solid tumors. Ann. Oncol. 16, 1688–1694 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We acknowledge the Stanford High-Throughput Bioscience Center for assistance with high-throughput imaging and plate reader assays. We acknowledge support from an American Heart Association (AHA) Predoctoral Fellowship (13PRE15770000) and an NSF Graduate Research Fellowship (DGE-114747) (A.S.); Burroughs Wellcome Foundation Innovation grant 1015009, AHA grant 17MERIT3361009, and National Institutes of Health (NIH) grants R01 HL132875, R01 HL130020, R01 HL128170, R01 HL123968, and R24 HL117756 (J.C.W.); and NIH grants NIH R01 HL130840, R01 HL128072, and R01 HL132225 (M.M.).

Author information

Authors and Affiliations

Authors

Contributions

A.S. designed, supervised, and performed experiments, and wrote the manuscript with help from all authors. W.L.M. performed high-throughput experiments related to the contractility analysis. R.S. developed analysis platforms for quantitative assessment of hiPSC-CM contractility. T.K. performed experiments related to hiPSC-CM cytotoxicity analysis. P.W.B. provided expertise related to hiPSC-CM platform development. J.C.d.Á. provided mathematical expertise related to hiPSC-CM contractility analysis. M.M. provided advice and proofread the manuscript. J.C.W. provided advice, proofread the manuscript, and provided financial support for experiments.

Corresponding author

Correspondence to Joseph C. Wu.

Ethics declarations

Competing interests

M.M. holds equity in and is on the scientific advisory board for Vala Sciences, a company offering high-content screening services and instrumentation for measuring the electrical and contractile physiology of CMs. J.C.W. is a cofounder of and scientific advisory board member for Khoris, a company using hiPSCs to accelerate drug discovery. The remaining authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related link

Key references using this protocol

Sharma, A. et al. Sci. Transl. Med. 9, eaaf2584 (2017): https://doi.org/10.1126/scitranslmed.aaf2584

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sharma, A., McKeithan, W.L., Serrano, R. et al. Use of human induced pluripotent stem cell–derived cardiomyocytes to assess drug cardiotoxicity. Nat Protoc 13, 3018–3041 (2018). https://doi.org/10.1038/s41596-018-0076-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0076-8

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research