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.

  • Perspective
  • Published:

Regenerating the field of cardiovascular cell therapy

Abstract

The retraction of >30 falsified studies by Anversa et al. has had a disheartening impact on the cardiac cell therapeutics field. The premise of heart muscle regeneration by the transdifferentiation of bone marrow cells or putative adult resident cardiac progenitors has been largely disproven. Over the past 18 years, a generation of physicians and scientists has lost years chasing these studies, and patients have been placed at risk with little scientific grounding. Funding agencies invested hundreds of millions of dollars in irreproducible work, and both academic institutions and the scientific community ignored troubling signals over a decade of questionable work. Our collective retrospective analysis identifies preventable problems at the level of the editorial and peer-review process, funding agencies and academic institutions. This Perspective provides a chronology of the forces that led to this scientific debacle, integrating direct knowledge of the process. We suggest a science-driven path forward that includes multiple novel approaches to the problem of heart muscle regeneration.

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: Timeline of fundamental findings and preclinical outcomes supporting or contradicting the notion of cardiac stem/progenitor cell therapy.

Similar content being viewed by others

References

  1. Oransky, I. & Marcus, A. Harvard and the Brigham call for more than 30 retractions of cardiac stem cell research. STAT https://www.statnews.com/2018/10/14/harvard-brigham-retractions-stem-cell/ (2018).

  2. Wilmut, I., Schnieke, A. E., McWhir, J., Kind, A. J. & Campbell, K. H. Viable offspring derived from fetal and adult mammalian cells. Nature 385, 810–813 (1997).

    Article  CAS  Google Scholar 

  3. Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).

    Article  CAS  Google Scholar 

  4. Okita, K., Ichisaka, T. & Yamanaka, S. Generation of germline-competent induced pluripotent stem cells. Nature 448, 313–317 (2007).

    Article  CAS  Google Scholar 

  5. Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145–1147 (1998).

    Article  CAS  Google Scholar 

  6. Krause, D. S. et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell 105, 369–377 (2001).

    Article  CAS  Google Scholar 

  7. Priller, J. et al. Neogenesis of cerebellar Purkinje neurons from gene-marked bone marrow cells in vivo. J. Cell Biol. 155, 733–738 (2001).

    Article  CAS  Google Scholar 

  8. Corbel, S. Y. et al. Contribution of hematopoietic stem cells to skeletal muscle. Nat. Med. 9, 1528–1532 (2003).

    Article  CAS  Google Scholar 

  9. Lagasse, E. et al. Purified hematopoietic stem cells can differentiate into hepatocytes in vivo. Nat. Med. 6, 1229–1234 (2000).

    Article  CAS  Google Scholar 

  10. LaBarge, M. A. & Blau, H. M. Biological progression from adult bone marrow to mononucleate muscle stem cell to multinucleate muscle fiber in response to injury. Cell 111, 589–601 (2002).

    Article  CAS  Google Scholar 

  11. Alvarez-Dolado, M. et al. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature 425, 968–973 (2003).

    Article  CAS  Google Scholar 

  12. Weimann, J. M., Johansson, C. B., Trejo, A. & Blau, H. M. Stable reprogrammed heterokaryons form spontaneously in Purkinje neurons after bone marrow transplant. Nat. Cell Biol. 5, 959–966 (2003).

    Article  CAS  Google Scholar 

  13. Morrison, S. J., Uchida, N. & Weissman, I. L. The biology of hematopoietic stem cells. Annu. Rev. Cell Dev. Biol. 11, 35–71 (1995).

    Article  CAS  Google Scholar 

  14. Massengale, M., Wagers, A. J., Vogel, H. & Weissman, I. L. Hematopoietic cells maintain hematopoietic fates upon entering the brain. J. Exp. Med. 201, 1579–1589 (2005).

    Article  CAS  Google Scholar 

  15. Wagers, A. J., Sherwood, R. I., Christensen, J. L. & Weissman, I. L. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science 297, 2256–2259 (2002).

    Article  CAS  Google Scholar 

  16. Weimann, J. M., Charlton, C. A., Brazelton, T. R., Hackman, R. C. & Blau, H. M. Contribution of transplanted bone marrow cells to Purkinje neurons in human adult brains. Proc. Natl Acad. Sci. USA 100, 2088–2093 (2003).

    Article  CAS  Google Scholar 

  17. Bjornson, C. R., Rietze, R. L., Reynolds, B. A., Magli, M. C. & Vescovi, A. L. Turning brain into blood: a hematopoietic fate adopted by adult neural stem cells in vivo. Science 283, 534–537 (1999).

    Article  CAS  Google Scholar 

  18. Beltrami, A. P. et al. Evidence that human cardiac myocytes divide after myocardial infarction. N. Engl. J. Med. 344, 1750–1757 (2001).

    Article  CAS  Google Scholar 

  19. Leri, A. et al. Telomerase expression and activity are coupled with myocyte proliferation and preservation of telomeric length in the failing heart. Proc. Natl Acad. Sci. USA 98, 8626–8631 (2001).

    Article  CAS  Google Scholar 

  20. Quaini, F. et al. Chimerism of the transplanted heart. N. Engl. J. Med. 346, 5–15 (2002).

    Article  Google Scholar 

  21. Orlic, D. et al. Mobilized bone marrow cells repair the infarcted heart, improving function and survival. Proc. Natl Acad. Sci. USA 98, 10344–10349 (2001).

    Article  CAS  Google Scholar 

  22. Orlic, D. et al. Bone marrow cells regenerate infarcted myocardium. Nature 410, 701–705 (2001).

    Article  CAS  Google Scholar 

  23. Strauer, B. E. et al. Repair of infarcted myocardium by autologous intracoronary mononuclear bone marrow cell transplantation in humans. Circulation 106, 1913–1918 (2002).

    Article  Google Scholar 

  24. Strauer, B. E. et al. [Intracoronary, human autologous stem cell transplantation for myocardial regeneration following myocardial infarction]. Dtsch Med. Wochenschr. 126, 932–938 (2001).

    Article  CAS  Google Scholar 

  25. Wollert, K. C. et al. Intracoronary autologous bone-marrow cell transfer after myocardial infarction: the BOOST randomised controlled clinical trial. Lancet 364, 141–148 (2004).

    Article  Google Scholar 

  26. Assmus, B. et al. Transcoronary transplantation of progenitor cells after myocardial infarction. N. Engl. J. Med. 355, 1222–1232 (2006).

    Article  CAS  Google Scholar 

  27. Murry, C. E. et al. Haematopoietic stem cells do not transdifferentiate into cardiac myocytes in myocardial infarcts. Nature 428, 664–668 (2004).

    Article  CAS  Google Scholar 

  28. Balsam, L. B. et al. Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocardium. Nature 428, 668–673 (2004).

    Article  CAS  Google Scholar 

  29. Nygren, J. M. et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat. Med. 10, 494–501 (2004).

    Article  CAS  Google Scholar 

  30. Chien, K. R. Stem cells: lost in translation. Nature 428, 607–608 (2004).

    Article  CAS  Google Scholar 

  31. Laugwitz, K. L. et al. Postnatal isl1+ cardioblasts enter fully differentiated cardiomyocyte lineages. Nature 433, 647–653 (2005).

    Article  CAS  Google Scholar 

  32. Moretti, A. et al. Multipotent embryonic Isl1 + progenitor cells lead to cardiac, smooth muscle, and endothelial cell diversification. Cell 127, 1151–1165 (2006).

    Article  CAS  Google Scholar 

  33. Zhou, B. et al. Epicardial progenitors contribute to the cardiomyocyte lineage in the developing heart. Nature 454, 109–113 (2008).

    Article  CAS  Google Scholar 

  34. Bu, L. et al. Human ISL1 heart progenitors generate diverse multipotent cardiovascular cell lineages. Nature 460, 113–117 (2009).

    Article  CAS  Google Scholar 

  35. Beltrami, A. P. et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 114, 763–776 (2003).

    Article  CAS  Google Scholar 

  36. Linke, A. et al. Stem cells in the dog heart are self-renewing, clonogenic, and multipotent and regenerate infarcted myocardium, improving cardiac function. Proc. Natl Acad. Sci. USA 102, 8966–8971 (2005).

    Article  CAS  Google Scholar 

  37. Dawn, B. et al. Cardiac stem cells delivered intravascularly traverse the vessel barrier, regenerate infarcted myocardium, and improve cardiac function. Proc. Natl Acad. Sci. USA 102, 3766–3771 (2005).

    Article  CAS  Google Scholar 

  38. Hatzistergos, K. E. et al. Bone marrow mesenchymal stem cells stimulate cardiac stem cell proliferation and differentiation. Circ. Res. 107, 913–922 (2010).

    Article  CAS  Google Scholar 

  39. Fischer, K. M. et al. Enhancement of myocardial regeneration through genetic engineering of cardiac progenitor cells expressing Pim-1 kinase. Circulation 120, 2077–2087 (2009).

    Article  CAS  Google Scholar 

  40. Williams, A. R. et al. Enhanced effect of combining human cardiac stem cells and bone marrow mesenchymal stem cells to reduce infarct size and to restore cardiac function after myocardial infarction. Circulation 127, 213–223 (2013).

    Article  Google Scholar 

  41. Li, Q. et al. Intracoronary administration of cardiac stem cells in mice: a new, improved technique for cell therapy in murine models. Basic Res. Cardiol. 106, 849–864 (2011).

    Article  Google Scholar 

  42. Oh, H. et al. Cardiac muscle plasticity in adult and embryo by heart-derived progenitor cells. Ann. NY Acad. Sci. 1015, 182–189 (2004).

    Article  Google Scholar 

  43. Smith, R. R. et al. Regenerative potential of cardiosphere-derived cells expanded from percutaneous endomyocardial biopsy specimens. Circulation 115, 896–908 (2007).

    Article  Google Scholar 

  44. Bolli, R. et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): initial results of a randomised phase 1 trial. Lancet 378, 1847–1857 (2011).

    Article  Google Scholar 

  45. Makkar, R. R. et al. Intracoronary cardiosphere-derived cells for heart regeneration after myocardial infarction (CADUCEUS): a prospective, randomised phase 1 trial. Lancet 379, 895–904 (2012).

    Article  Google Scholar 

  46. Pouly, J. et al. Cardiac stem cells in the real world. J. Thorac. Cardiovasc. Surg. 135, 673–678 (2008).

    Article  Google Scholar 

  47. Senyo, S. E. et al. Mammalian heart renewal by pre-existing cardiomyocytes. Nature 493, 433–436 (2013).

    Article  CAS  Google Scholar 

  48. Soonpaa, M. H. & Field, L. J. Assessment of cardiomyocyte DNA synthesis in normal and injured adult mouse hearts. Am. J. Physiol. 272, H220–H226 (1997).

    CAS  PubMed  Google Scholar 

  49. Rumyantsev, P. P. DNA synthesis and nuclear division in embryonal and postnatal histogenesis of myocardium (autoradiographic study). Fed. Proc. Transl. Suppl. 24, 899–902 (1965).

    CAS  PubMed  Google Scholar 

  50. van Berlo, J. H. et al. c-kit+ cells minimally contribute cardiomyocytes to the heart. Nature 509, 337–341 (2014).

    Article  Google Scholar 

  51. Moccetti, T., Leri, A. & Anversa, P. Controversy in myocardial regeneration. Regen. Med. 10, 921–924 (2015).

    Article  CAS  Google Scholar 

  52. Sultana, N. et al. Resident c-kit(+) cells in the heart are not cardiac stem cells. Nat. Commun. 6, 8701 (2015).

    Article  CAS  Google Scholar 

  53. Vagnozzi, R. J. et al. Genetic lineage tracing of Sca-1+ cells reveals endothelial but not myogenic contribution to the murine heart. Circulation 138, 2931–2939 (2018).

    Article  CAS  Google Scholar 

  54. Neidig, L. E. et al. Evidence for minimal cardiogenic potential of stem cell antigen 1-positive cells in the adult mouse heart. Circulation 138, 2960–2962 (2018).

    Article  Google Scholar 

  55. Bergmann, O. et al. Evidence for cardiomyocyte renewal in humans. Science 324, 98–102 (2009).

    Article  CAS  Google Scholar 

  56. Kajstura, J. et al. Cardiomyogenesis in the aging and failing human heart. Circulation 126, 1869–1881 (2012); retraction 129, e466 (2014).

  57. Wollert, K. C. et al. Intracoronary autologous bone marrow cell transfer after myocardial infarction: the BOOST-2 randomised placebo-controlled clinical trial. Eur. Heart J. 38, 2936–2943 (2017).

    Article  CAS  Google Scholar 

  58. Janssens, S. et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet 367, 113–121 (2006).

    Article  Google Scholar 

  59. Nowbar, A. N. et al. Discrepancies in autologous bone marrow stem cell trials and enhancement of ejection fraction (DAMASCENE): weighted regression and meta-analysis. Br. Med. J. 348, g2688 (2014).

    Article  Google Scholar 

  60. Morgan, P. et al. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat. Rev. Drug Discov. 17, 167–181 (2018).

    Article  CAS  Google Scholar 

  61. Lian, X. et al. Robust cardiomyocyte differentiation from human pluripotent stem cells via temporal modulation of canonical Wnt signaling. Proc. Natl Acad. Sci. USA 109, E1848–E1857 (2012).

    Article  CAS  Google Scholar 

  62. Birket, M. J. et al. Expansion and patterning of cardiovascular progenitors derived from human pluripotent stem cells. Nat. Biotechnol. 33, 970–979 (2015).

    Article  CAS  Google Scholar 

  63. Lee, J. H., Protze, S. I., Laksman, Z., Backx, P. H. & Keller, G. M. Human pluripotent stem cell-derived atrial and ventricular cardiomyocytes develop from distinct mesoderm populations. Cell Stem Cell 21, 179–194.e4 (2017).

    Article  CAS  Google Scholar 

  64. Foo, K. S. et al. Human ISL1+ ventricular progenitors self-assemble into an in vivo functional heart patch and preserve cardiac function post infarction. Mol. Ther. 26, 1644–1659 (2018).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  66. Liu, Y. W. et al. Human embryonic stem cell-derived cardiomyocytes restore function in infarcted hearts of non-human primates. Nat. Biotechnol. 36, 597–605 (2018).

    Article  CAS  Google Scholar 

  67. Menasché, P. et al. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 71, 429–438 (2018).

    Article  Google Scholar 

  68. Sahara, M., Santoro, F. & Chien, K. R. Programming and reprogramming a human heart cell. EMBO J. 34, 710–738 (2015).

    Article  CAS  Google Scholar 

  69. Qian, L. et al. In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes. Nature 485, 593–598 (2012).

    Article  CAS  Google Scholar 

  70. Mohamed, T. M. A. et al. Regulation of cell cycle to stimulate adult cardiomyocyte proliferation and cardiac regeneration. Cell 173, 104–116.e12 (2018).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors gratefully acknowledge the careful editing and review of the manuscript by M. Sahara of the Karolinska Institutet.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Kenneth R. Chien, Jonas Frisén, Regina Fritsche-Danielson, Douglas A. Melton, Charles E. Murry or Irving L. Weissman.

Ethics declarations

Competing interests

K.R.C. is a scientific founder and equity holder in Moderna Therapeutics and Procella Therapeutics, and chair of the External Science Panel for AstraZeneca. R.F.-D. is an employee of AstraZeneca. J.F. is an advisor to 10XGenomics. D.A.M. is cofounder of Semma Therapeutics. C.E.M. is a scientific founder and equity holder in Cytocardia. I.L.W. is a cofounder of, director of, stockholder in and consultant to Forty Seven Inc, a company currently devoted to cancer immunotherapies with antibodies to macrophage checkpoint inhibitors.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chien, K.R., Frisén, J., Fritsche-Danielson, R. et al. Regenerating the field of cardiovascular cell therapy. Nat Biotechnol 37, 232–237 (2019). https://doi.org/10.1038/s41587-019-0042-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-019-0042-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing