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.

  • Science and Society
  • Published:

Pluripotent stem cells progressing to the clinic

Abstract

Basic experimental stem cell research has opened up the possibility of many diverse clinical applications; however, translation to clinical trials has been restricted to only a few diseases. To broaden this clinical scope, pluripotent stem cell derivatives provide a uniquely scalable source of functional differentiated cells that can potentially repair damaged or diseased tissues to treat a wide spectrum of diseases and injuries. However, gathering sound data on their distribution, longevity, function and mechanisms of action in host tissues is imperative to realizing their clinical benefit. The large-scale availability of treatments involving pluripotent stem cells remains some years away, because of the long and demanding regulatory pathway that is needed to ensure their safety.

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

Relevant articles

Open Access articles citing this article.

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: The leading applications for pluripotent stem cell derivatives.
Figure 2: The bench to bedside pathway.
Figure 3: The translation pathway for product development and interactions with regulatory agencies.

References

  1. Trounson, A. & McDonald, C. Stem cell therapies in clinical trials: progress and challenges. Cell Stem Cell 17, 11–22 (2015).

    Article  CAS  Google Scholar 

  2. Trounson, A., Thakar, R. G., Lomax, G. & Gibbons, D. Clinical trials for stem cell therapies. BMC Med. 9, 52 (2011).

    Article  Google Scholar 

  3. Ratcliffe, E., Glen, K. E., Naing, M. W. & Williams, D. J. Current status and perspectives on stem cell-based therapies undergoing clinical trials for regenerative medicine: case studies. Br. Med. Bull. 108, 73–94 (2013).

    Article  Google Scholar 

  4. Glenn, J. D. & Whartenby, K. A. Mesenchymal stem cells: emerging mechanisms of immunomodulation and therapy. World J. Stem Cells 6, 526–539 (2014).

    Article  Google Scholar 

  5. Sarukhan, A., Zanotti, L. & Viola, A. Mesenchymal stem cells: myths and reality. Swiss Med. Wkly 145, w14229 (2015).

    PubMed  Google Scholar 

  6. Eggenhofer, E., Luk, F., Dahlke, M. H. & Hoogduijn, M. J. The life and fate of mesenchymal stem cells. Front. Immunol. 5, 148 (2014).

    Article  Google Scholar 

  7. von der Haar, K., Lavrentieva, A., Stahl, F., Scheper, T. & Blume, C. Lost signature: progress and failures in in vivo tracking of implanted stem cells. Appl. Microbiol. Biotechnol. 99, 9907–9922 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Reubinoff, B. E., Pera, M. F., Fong, C. Y., Trounson, A. & Bongso, A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat. Biotechnol. 18, 399–404 (2000).

    Article  CAS  Google Scholar 

  10. 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 

  11. Takahashi, K. et al. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131, 861–872 (2007).

    CAS  Google Scholar 

  12. Takahashi. K. & Yamanaka, S. A decade of transcription factor-mediated reprogramming to pluripotency. Nat. Rev. Mol. Cell Biol. http://dx.doi.org/10.1038/nrm.2016.8 (2016).

  13. Kimbrel, E. A. & Lanza, R. Current status of pluripotent stem cells: moving the first therapies to the clinic. Nat. Rev. Drug Discov. 14, 681–692 (2015).

    Article  CAS  Google Scholar 

  14. Dimmeler, S., Ding, S., Rando, T. A. & Trounson, A. Translational strategies and challenges in regenerative medicine. Nat. Med. 20, 814–821 (2014).

    Article  CAS  Google Scholar 

  15. Mascetti, V. L. & Pedersen, R. A. Human-mouse chimerism validates human stem cell pluripotency. Cell Stem Cell 18, 67–72 (2016).

    Article  CAS  Google Scholar 

  16. Mascetti, V. L. & Pedersen, R. A. Naivete of the human pluripotent stem cell. Nat. Biotechnol. 32, 68–70 (2014).

    Article  CAS  Google Scholar 

  17. Kehinde, E. O. They see a rat, we seek a cure for diseases: the current status of animal experimentation in medical practice. Med. Princ. Pract. 22 (Suppl. 1), 52–61 (2013).

    Article  Google Scholar 

  18. Kastner, A. & Gauthier, P. Are rodents an appropriate pre-clinical model for treating spinal cord injury? Examples from the respiratory system. Exp. Neurol. 213, 249–256 (2008).

    Article  CAS  Google Scholar 

  19. Schroeder, M. A. & DiPersio, J. F. Mouse models of graft-versus-host disease: advances and limitations. Dis. Model. Mech. 4, 318–333 (2011).

    Article  CAS  Google Scholar 

  20. Thomas, K. E. & Moon, L. D. Will stem cell therapies be safe and effective for treating spinal cord injuries? Br. Med. Bull. 98, 127–142 (2011).

    Article  Google Scholar 

  21. Tetzlaff, W. et al. A systematic review of cellular transplantation therapies for spinal cord injury. J. Neurotrauma 28, 1611–1682 (2011).

    Article  Google Scholar 

  22. Priest, C. A. et al. Preclinical safety of human embryonic stem cell-derived oligodendrocyte progenitors supporting clinical trials in spinal cord injury. Regen. Med. 10, 939–958 (2015).

    Article  CAS  Google Scholar 

  23. Chapman, A. R. & Scala, C. C. Evaluating the first-in-human clinical trial of a human embryonic stem cell-based therapy. Kennedy Inst. Eth. J. 22, 243–261 (2012).

    Article  Google Scholar 

  24. Barker, R. A. et al. The long-term safety and efficacy of bilateral transplantation of human fetal striatal tissue in patients with mild to moderate Huntington's disease. J. Neurol. Neurosurg. Psychiatry 84, 657–665 (2013).

    Article  Google Scholar 

  25. Cicchetti, F. et al. Neural transplants in patients with Huntington's disease undergo disease-like neuronal degeneration. Proc. Natl Acad. Sci. USA 106, 12483–12488 (2009).

    Article  CAS  Google Scholar 

  26. Steinbeck, J. A. & Studer, L. Moving stem cells to the clinic: potential and limitations for brain repair. Neuron 86, 187–206 (2015).

    Article  CAS  Google Scholar 

  27. Reddington, A. E., Rosser, A. E. & Dunnett, S. B. Differentiation of pluripotent stem cells into striatal projection neurons: a pure MSN fate may not be sufficient. Front. Cell. Neurosci. 8, 398 (2014).

    Article  Google Scholar 

  28. Grealish, S. et al. Human ESC-derived dopamine neurons show similar preclinical efficacy and potency to fetal neurons when grafted in a rat model of Parkinson's disease. Cell Stem Cell 15, 653–665 (2014).

    Article  CAS  Google Scholar 

  29. Ganat, Y. M. et al. Identification of embryonic stem cell-derived midbrain dopaminergic neurons for engraftment. J. Clin. Invest. 122, 2928–2939 (2012).

    Article  CAS  Google Scholar 

  30. Brevini, T. A., Pennarossa, G., Vanelli, A., Maffei, S. & Gandolfi, F. Parthenogenesis in non-rodent species: developmental competence and differentiation plasticity. Theriogenology 77, 766–772 (2012).

    Article  CAS  Google Scholar 

  31. Gonzalez, R. et al. Proof of concept studies exploring the safety and functional activity of human parthenogenetic-derived neural stem cells for the treatment of Parkinson's disease. Cell Transplant 24, 681–690 (2015).

    Article  Google Scholar 

  32. Parkinson's stem cell therapy OK'd for testing. Bloomberg [online], (2015).

  33. Politis, M. et al. Serotonin neuron loss and nonmotor symptoms continue in Parkinson's patients treated with dopamine grafts. Sci. Transl. Med. 4, 128ra141 (2012).

    Article  Google Scholar 

  34. Vazin, T. et al. The effect of multivalent Sonic hedgehog on differentiation of human embryonic stem cells into dopaminergic and GABAergic neurons. Biomaterials 35, 941–948 (2014).

    Article  CAS  Google Scholar 

  35. Martinez, C. et al. Proliferation of murine midbrain neural stem cells depends upon an endogenous sonic hedgehog (Shh) source. PLoS ONE 8, e65818 (2013).

    Article  CAS  Google Scholar 

  36. L'Episcopo, F. et al. Wnt/β-catenin signaling is required to rescue midbrain dopaminergic progenitors and promote neurorepair in ageing mouse model of Parkinson's disease. Stem Cells 32, 2147–2163 (2014).

    Article  Google Scholar 

  37. Rowland, N. C. et al. Combining cell transplants or gene therapy with deep brain stimulation for Parkinson's disease. Mov. Disord. 30, 190–195 (2015).

    Article  Google Scholar 

  38. Novel cellular therapeutic approach for ALS gets FDA clearance for first-in-human trials. Q Therapeutics [online], (2015).

  39. Gowing, G. et al. Glial cell line-derived neurotrophic factor-secreting human neural progenitors show long-term survival, maturation into astrocytes, and no tumor formation following transplantation into the spinal cord of immunocompromised rats. Neuroreport 25, 367–372 (2014).

    Article  CAS  Google Scholar 

  40. Kaur, S. J., McKeown, S. R. & Rashid, S. Mutant SOD1 mediated pathogenesis of amyotrophic lateral sclerosis. Gene 577, 109–118 (2016).

    Article  CAS  Google Scholar 

  41. Contestabile, A. Amyotrophic lateral sclerosis: from research to therapeutic attempts and therapeutic perspectives. Curr. Med. Chem. 18, 5655–5665 (2011).

    Article  CAS  Google Scholar 

  42. Garcia, J. M. et al. Stem cell therapy for retinal diseases. World J. Stem Cells 7, 160–164 (2015).

    Article  Google Scholar 

  43. Forest, D. L., Johnson, L. V. & Clegg, D. O. Cellular models and therapies for age-related macular degeneration. Dis. Model. Mech. 8, 421–427 (2015).

    Article  CAS  Google Scholar 

  44. Song, W. K. et al. Treatment of macular degeneration using embryonic stem cell-derived retinal pigment epithelium: preliminary results in Asian patients. Stem Cell Rep. 4, 860–872 (2015).

    Article  CAS  Google Scholar 

  45. Krohne, T. U. et al. Generation of retinal pigment epithelial cells from small molecules and OCT4 reprogrammed human induced pluripotent stem cells. Stem Cells Transl. Med. 1, 96–109 (2012).

    Article  CAS  Google Scholar 

  46. Pennington, B. O., Clegg, D. O., Melkoumian, Z. K. & Hikita, S. T. Defined culture of human embryonic stem cells and xeno-free derivation of retinal pigmented epithelial cells on a novel, synthetic substrate. Stem Cells Transl. Med. 4, 165–177 (2015).

    Article  CAS  Google Scholar 

  47. Hu, Y. et al. A novel approach for subretinal implantation of ultrathin substrates containing stem cell-derived retinal pigment epithelium monolayer. Ophthalm. Res. 48, 186–191 (2012).

    Article  Google Scholar 

  48. Nazari, H. et al. Stem cell based therapies for age-related macular degeneration: the promises and the challenges. Prog. Retin. Eye Res. 48, 1–39 (2015).

    Article  CAS  Google Scholar 

  49. Carr, A. J. et al. Development of human embryonic stem cell therapies for age-related macular degeneration. Trends Neurosci. 36, 385–395 (2013).

    Article  CAS  Google Scholar 

  50. Schwartz, S. D. et al. Human embryonic stem cell-derived retinal pigment epithelium in patients with age-related macular degeneration and Stargardt's macular dystrophy: follow-up of two open-label phase 1/2 studies. Lancet 385, 509–516 (2015).

    Article  Google Scholar 

  51. Dang, Y., Zhang, C. & Zhu, Y. Stem cell therapies for age-related macular degeneration: the past, present, and future. Clin. Interv. Aging 10, 255–264 (2015).

    Article  Google Scholar 

  52. Zhao, T. et al. Humanized mice reveal differential immunogenicity of cells derived from autologous induced pluripotent stem cells. Cell Stem Cell 17, 353–359 (2015).

    Article  CAS  Google Scholar 

  53. First iPS cell transplant patient makes progress one year on. Japan Times [online], (2015).

  54. Schulz, T. C. et al. A scalable system for production of functional pancreatic progenitors from human embryonic stem cells. PLoS ONE 7, e37004 (2012).

    Article  CAS  Google Scholar 

  55. Rezania, A. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat. Biotechnol. 32, 1121–1133 (2014).

    Article  CAS  Google Scholar 

  56. Pagliuca, F. W. et al. Generation of functional human pancreatic β cells in vitro. Cell 159, 428–439 (2014).

    Article  CAS  Google Scholar 

  57. Lee, S. H. et al. Human β-cell precursors mature into functional insulin-producing cells in an immunoisolation device: implications for diabetes cell therapies. Transplantation 87, 983–991 (2009).

    Article  Google Scholar 

  58. Kumar, S. S. et al. Recent developments in β-cell differentiation of pluripotent stem cells induced by small and large molecules. Int. J. Mol. Sci. 15, 23418–23447 (2014).

    Article  Google Scholar 

  59. Schulz, T. C. Concise review: manufacturing of pancreatic endoderm cells for clinical trials in type 1 diabetes. Stem Cells Transl. Med. 4, 927–931 (2015).

    Article  CAS  Google Scholar 

  60. Sornberger, J. A crucial moment in time for stem cell R&D. Biotechnology Focus [online], (2015).

    Google Scholar 

  61. Gouadon, E. et al. Concise review: pluripotent stem cell-derived cardiac cells, a promising cell source for therapy of heart failure: where do we stand? Stem Cells 34, 34–43 (2015).

    Article  Google Scholar 

  62. Wehman, B., Siddiqui, O. T., Mishra, R., Sharma, S. & Kaushal, S. Stem cell therapy for CHD: towards translation. Cardiol. Young 25 (Suppl. 2), 58–66 (2015).

    Article  Google Scholar 

  63. Fernandes, S. et al. Comparison of human embryonic stem cell-derived cardiomyocytes, cardiovascular progenitors, and bone marrow mononuclear cells for cardiac repair. Stem Cell Rep. 5, 753–762 (2015).

    Article  CAS  Google Scholar 

  64. Chong, J. J. et al. Human embryonic-stem-cell-derived cardiomyocytes regenerate non-human primate hearts. Nature 510, 273–277 (2014).

    Article  CAS  Google Scholar 

  65. Menasche, P. et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report. Eur. Heart J. 36, 2011–2017 (2015).

    Article  Google Scholar 

  66. Menasche, P. et al. Towards a clinical use of human embryonic stem cell-derived cardiac progenitors: a translational experience. Eur. Heart J. 36, 743–750 (2015).

    Article  CAS  Google Scholar 

  67. Knoepfler, P. S. From bench to FDA to bedside: US regulatory trends for new stem cell therapies. Adv. Drug Deliv. Rev. 82–83, 192–196 (2015).

    Article  Google Scholar 

  68. Baghbaderani, B. A. et al. cGMP-manufactured human induced pluripotent stem cells are available for pre-clinical and clinical applications. Stem Cell Rep. 5, 647–659 (2015).

    Article  CAS  Google Scholar 

  69. Simonson, O. E., Domogatskaya, A., Volchkov, P. & Rodin, S. The safety of human pluripotent stem cells in clinical treatment. Ann. Med. 47, 370–380 (2015).

    Article  Google Scholar 

  70. Ma, H. et al. Abnormalities in human pluripotent cells due to reprogramming mechanisms. Nature 511, 177–183 (2014).

    Article  CAS  Google Scholar 

  71. Campbell, A. et al. Concise review: process development considerations for cell therapy. Stem Cells Transl. Med. 4, 1155–1163 (2015).

    Article  Google Scholar 

  72. Shaw, G. Unregulated stem cell clinics proliferate in the US, despite 2013 court ruling. Neurology Today 15, 1,8–9 (2015).

    Google Scholar 

  73. Kyoto University's potential iPS cell therapy for Parkinson's may be delayed. Japan Times [online], (2015).

  74. International Society for Stem Cell Research DRAFT Guidelines for Stem Cell Science and Clinical Translation. ISSCR [online], (2015).

  75. Institute of Medicine Stem cell therapies: opportunities for ensuring the quality and safety of clinical offerings — summary of a joint workshop. IOM [online], (2014).

  76. Ghosh, S., Thrasher, A. J. & Gaspar, H. B. Gene therapy for monogenic disorders of the bone marrow. Br. J. Haematol. 171, 55–170 (2015).

    Article  Google Scholar 

  77. Xu, P. et al. Both TALENs and CRISPR/Cas9 directly target the HBB IVS2–654 (C > T) mutation in β-thalassemia-derived iPSCs. Sci. Rep. 5, 12065 (2015).

    Article  Google Scholar 

  78. Drake, M. J. & Bates, P. Application of gene-editing technologies to HIV-1. Curr. Opin. HIV AIDS 10, 123–127 (2015).

    Article  CAS  Google Scholar 

  79. Gu, W. G. & Chen, X. Q. Targeting CCR5 for anti-HIV research. Eur. J. Clin. Microbiol. Infect. Dis. 33, 1881–1887 (2014).

    Article  CAS  Google Scholar 

  80. Tasian, S. K. & Gardner, R. A. CD19-redirected chimeric antigen receptor-modified T cells: a promising immunotherapy for children and adults with B-cell acute lymphoblastic leukemia (ALL). Ther. Adv. Hematol. 6, 228–241 (2015).

    Article  CAS  Google Scholar 

  81. Perales, M. A., Sauter, C. S. & Armand, P. Fast cars and no brakes: autologous stem cell transplantation as a platform for novel immunotherapies. Biol. Blood Marrow Transplant. 22, 17–22 (2015).

    Article  Google Scholar 

  82. June, C. H. & Levine, B. L. T cell engineering as therapy for cancer and HIV: our synthetic future. Philos. Trans. R Soc. Lond. Biol. Sci. 370, 20140374 (2015).

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alan Trounson.

Ethics declarations

Competing interests

A.T. declares competing financial interests as a Board Director of Stem Cells Inc., California, USA, and Cartherics Pty Ltd., Australia.

PowerPoint slides

Glossary

Allogeneic therapies

Transplants using cells or tissues derived from a different person.

Autologous therapies

Immunologically compatible transplants using the patient's own cells or tissues.

A9 dopaminergic neurons

Mature dopamine-producing neurons of the midbrain substantia nigra, which are lost in Parkinson disease.

Choroid

Vascular layer of the eye between the retina and the sclera.

Cell-mediated immune rejection

Activation of the innate immune system in response to cells recognized as foreign, which can constitute a major barrier to successful organ transplantation and allogeneic cell therapies.

Dyskinesia

Repetitive, involuntary and purposeless movements.

Expandable (immortal) in culture

Capable of continuous cell replication and passage in vitro without senescence.

Graft-versus-host disease

(GVHD). When the immune cells of a foreign transplant attack the recipient's own tissues, causing serious life-threatening loss of organ or tissue function.

Housekeeping genes

Constitutive genes that are responsible for the maintenance of basic cell function and are normally expressed in all cells.

Humanized mice

Genetically modified mice with human immune cells that are tolerant to human grafts.

Immune-competent humans

Humans with natural uncompromised immunity.

Immune-incompetent mice

Mice that are genetically modified to prevent the development of an adaptive immune system.

Infarct

An area of tissue death or necrosis caused by lack of local oxygen due to blockage of blood supply.

Knock-in gene mutants

Cells or organisms with gene and regulatory DNA elements introduced into their genomes to enable expression of an introduced mutant gene.

Macula

An oval-shaped area near the centre of the retina of the eye, specialized for high-acuity vision.

Oligodendrocyte

Glial cell of the central nervous system (CNS) that is responsible for creating the myelin sheath surrounding the axons of CNS neurons, enabling transmission of electronic impulses.

Optogenetic

Light-directed identification and control of individual neurons, in living tissue, that have been genetically modified to express light-sensitive ion channels.

Pharmacogenetic

Drug-directed control of individual neurons that have been genetically modified to respond to specific drugs.

Teratomas

Tumours with organ and tissue components that are derivatives of more than one germ layer, typically formed in vivo from pluripotent stem cells.

Tissue stem cell

Undifferentiated stem cell in a tissue or organ that can self-renew and give rise to the major cell types of that tissue or organ. Tissue stem cells are also referred to as adult stem cells or somatic stem cells.

Ventricular arrhythmias

Severely abnormal heart rhythms that cause the majority of sudden deaths due to heart problems.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Trounson, A., DeWitt, N. Pluripotent stem cells progressing to the clinic. Nat Rev Mol Cell Biol 17, 194–200 (2016). https://doi.org/10.1038/nrm.2016.10

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm.2016.10

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