We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Clinical applications of pluripotent stem cells and their derivatives: current status and future perspectives

    Shalmali Pendse

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    ,
    Anuradha Vaidya

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    &
    Vaijayanti Kale

    *Author for correspondence: Tel.: +91 20 2811 6315 6393;

    E-mail Address: vaijayanti.kale@ssbs.edu.in

    Symbiosis Centre for Stem Cell Research, Symbiosis International (Deemed University), Pune, 412115, India

    Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Pune, 412115, India

    Published Online:https://doi.org/10.2217/rme-2022-0045

    Pluripotent stem cells (PSCs) can differentiate into specific cell types and thus hold great promise in regenerative medicine to treat certain diseases. Hence, several studies have been performed harnessing their salutary properties in regenerative medicine. Despite several challenges associated with the clinical applications of PSCs, worldwide efforts are harnessing their potential in the regeneration of damaged tissues. Several clinical trials have been performed using PSCs or their derivatives. However, the delay in publishing the data obtained in the trials has led to a lack of awareness about their outcomes, resulting in apprehension about cellular therapies. Here, the authors review the published papers containing data from recent clinical trials done with PSCs. PSC-derived extracellular vesicles hold great potential in regenerative therapy. Since published papers containing the data obtained in clinical trials on PSC-derived extracellular vesicles are not available yet, the authors have reviewed some of the pre-clinical work done with them.

    Plain language summary

    Embryonic stem cells (ESCs) can make all types of cells in the body. Likewise, induced pluripotent stem cells (iPSCs), which are laboratory-generated counterparts of ESCs, possess similar properties. ESCs and iPSCs have immense application in regenerative medicine, as they can be the only cure for certain diseases and conditions that are incurable with currently available treatments; however, several challenges remain. Notably, many clinical trials using these cells or their products are going on globally. However, due to the extensive time frame required to complete the clinical trials and publish the data obtained, the outcomes of these trials do not reach the general population. This delay in information flow to the public domain creates apprehension about cellular therapy. Here, the authors have reviewed recent publications documenting the results obtained in the clinical trials done with ESCs and iPSCs (together referred to as pluripotent stem cells). The vesicles (called extracellular vesicles) secreted by pluripotent stem cells also have great regenerative potential. Since published papers containing the results obtained in clinical trials done with these vesicles are not available yet, the authors have reviewed some pre-clinical work done on them.

    Graphical abstract

    Papers of special note have been highlighted as: • of interest

    References

    • 1. Barati M, Akhondi M, Mousavi NS et al. Pluripotent stem cells: cancer study, therapy, and vaccination. Stem. Cell Rev. Rep. 17(6), 1975–1992 (2021).
    • 2. Chan SW, Rizwan M, Yim EKF. Emerging methods for enhancing pluripotent stem cell expansion. Front. Cell Dev. Biol. 8, 70 (2020).
    • 3. Herberts CA, Kwa MSG, Hermsen HPH. Risk factors in the development of stem cell therapy. J. Transl. Med. 9, 29 (2011).
    • 4. Yamanaka S. Pluripotent stem cell-based cell therapy – promise and challenges. Cell Stem Cell 27(4), 523–531 (2020).
    • 5. Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res. Ther. 10(1), 68 (2019).
    • 6. Eguizabal C, Aran B, Chuva de Sousa Lopes SM et al. Two decades of embryonic stem cells: a historical overview. Hum. Reprod. Open 2019(1), hoy024 (2019).
    • 7. Thompson JA, Itskovitz-Eldor J, Shapiro SS et al. Embryonic stem cell lines derived from human blastocysts. Science 282(5391), 1145–1147 (1998).
    • 8. Mahla RS. Stem cells applications in regenerative medicine and disease therapeutics. Int. J. Cell Biol. 2016, 6940283 (2016).
    • 9. Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod. Biol. Endocrinol. 13, 9 (2015).
    • 10. Murry CE, Keller G. Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132(4), 661–680 (2008).
    • 11. Carpenedo RL, McDevitt TC. Chapter II.1.7 – Stem cells: key concepts. In: Biomaterials Science (3rd Edition). Ratner BDHoffman ASSchoen FJLemons JE (Eds). Academic Press, MA, USA, 487–495 (2013).
    • 12. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126(4), 663–676 (2006). • Paper demonstrating the creation of induced pluripotent stem cells for the first time
    • 13. Omole AE, Fakoya AOJ. Ten years of progress and promise of induced pluripotent stem cells: historical origins, characteristics, mechanisms, limitations, and potential applications. PeerJ 2018(May), e4370 (2018).
    • 14. Moradi S, Mahdizadeh H, Šarić T et al. Research and therapy with induced pluripotent stem cells (iPSCs): social, legal, and ethical considerations. Stem Cell Res. Ther. 10(1), 341 (2019).
    • 15. Papp B, Plath K. Reprogramming to pluripotency: stepwise resetting of the epigenetic landscape. Cell Res. 21(3), 486–501 (2011).
    • 16. Cyranoski D. The cells that sparked a revolution. Nature 555(7697), 428–430 (2018).
    • 17. Rowe RG, Daley GQ. Induced pluripotent stem cells in disease modelling and drug discovery. Nat. Rev. Genet. 20(7), 377–388 (2019).
    • 18. Yagi T, Ito D, Okada Y et al. Modeling familial Alzheimer's disease with induced pluripotent stem cells. Hum. Mol. Genet. 20(23), 4530–4539 (2011).
    • 19. Chamberlain SJ. Disease modelling using human iPSCss. Hum. Mol. Genet. 25(R2), R173–R181 (2016).
    • 20. Okuno H, Okano H. Modeling human congenital disorders with neural crest developmental defects using patient-derived induced pluripotent stem cells, Japanese Society of Regenerative Medicine. Regen. Ther. 18, 275–280 (2021).
    • 21. Volarevic V, Markovic BS, Gazdic M et al. Ethical and safety issues of stem cell-based therapyr. Int. J. Med. Sci. 15(1), 36–45 (2018).
    • 22. Lee SY, Chung SK. Integrating gene correction in the reprogramming and transdifferentiation processes: a one-step strategy to overcome stem cell-based gene therapy limitations. Stem Cell Int. 2016, 2725670 (2016).
    • 23. Malik N, Rao MS. A review of the methods for human iPSC derivation. Methods Mol. Biol. 997, 23–33 (2013).
    • 24. Yue W, Li Y, Zhang T et al. ESC-derived basal forebrain cholinergic neurons ameliorate the cognitive symptoms associated with Alzheimer's disease in mouse models. Stem Cell Rep. 5(5), 776–790 (2015).
    • 25. Eckert A, Huang L, Gonzalez R, Kim H-S, Hamblin MH, Lee J-P. Bystander effect fuels human induced pluripotent stem cell-derived neural stem cells to quickly attenuate early stage neurological deficits after stroke. Stem Cells Transl. Med. 4(7), 841–851 (2015).
    • 26. Cha M-Y, Kwon Y-W, Ahn H-S et al. Protein-induced pluripotent stem cells ameliorate cognitive dysfunction and reduce Aβ deposition in a mouse model of Alzheimer's disease. Stem Cells Transl. Med. 6(1), 293–305 (2017).
    • 27. Caldas HC, Lojudice FH, Dias C et al. Induced pluripotent stem cells reduce progression of experimental chronic kidney disease but develop Wilms' tumors. Stem Cells Intl 2017, 7428316 (2017).
    • 28. Yuan L, Liu X, Zhang L et al. A chimeric humanized mouse model by engrafting the human induced pluripotent stem cell-derived hepatocyte-like cell for the chronic hepatitis B virus infection. Front. Microbiol. 9, 908 (2018).
    • 29. Doi D, Magotani H, Kikuchi T et al. Pre-clinical study of induced pluripotent stem cell-derived dopaminergic progenitor cells for Parkinson's disease. Nat. Commun. 11(1), 3369 (2020).
    • 30. de Carvalho Ribeiro P, Lojudice FH, Fernandes-Charpiot IMM et al. Therapeutic potential of human induced pluripotent stem cells and renal progenitor cells in experimental chronic kidney disease. Stem Cell Res. Ther. 11(1), 530 (2020).
    • 31. Wu YR, Hashiguchi T, Sho J, Chiou SH, Takahashi M, Mandai M. Transplanted mouse embryonic stem cell–derived retinal ganglion cells integrate and form synapses in a retinal ganglion cell-depleted mouse model. Invest. Ophthal. Vis. Sci. 62(13), 26 (2021).
    • 32. Oswald J, Kegeles E, Minelli T, Volchkov P, Baranov P. Transplantation of miPSC/mESC-derived retinal ganglion cells into healthy and glaucomatous retinas. Mol. Ther .Methods Clin. Dev. 21, 180–198 (2021).
    • 33. Deuse T, Tediashvili G, Hu X et al. Hypoimmune induced pluripotent stem cell-derived cell therapeutics treat cardiovascular and pulmonary diseases in immunocompetent allogeneic mice. Proc. Natl Acad. Sci. USA 118(28), e2022091118 (2021).
    • 34. Kimbrel EA, Lanza R. Pluripotent stem cells: the last 10 years. Regen. Med. 11(8), 831–847 (2016).
    • 35. Schwartz SD, Tan G, Hosseini H, Nagiel A. Subretinal transplantation of embryonic stem cell–derived retinal pigment epithelium for the treatment of macular degeneration: an assessment at 4 years. Invest. Ophthalmol. Vis. Sci. 57(5), ORSFc1–ORSFc9 (2016).
    • 36. Liu Y, Xu HW, Wang L et al. Human embryonic stem cell-derived retinal pigment epithelium transplants as a potential treatment for wet age-related macular degeneration. Cell Discov. 4, 50 (2018).
    • 37. Smith EN, D'Antonio-Chronowska A, Greenwald WW et al. Human iPSC-derived retinal pigment epithelium: a model system for prioritizing and functionally characterizing causal variants at AMD risk loci. Stem Cell Rep. 12(6), 1342–1353 (2019).
    • 38. Geng Z, Walsh PJ, Truong V et al. Generation of retinal pigmented epithelium from iPSCs derived from the conjunctiva of donors with and without age related macular degeneration. PLoS One 12(3), e0173575 (2017).
    • 39. Ye K, Takemoto Y, Ito A et al. Reproducible production and image-based quality evaluation of retinal pigment epithelium sheets from human induced pluripotent stem cells. Sci. Rep. 10(1), 14387 (2020).
    • 40. Gong J, Cai H, Noggle S et al. Stem cell-derived retinal pigment epithelium from patients with age-related macular degeneration exhibit reduced metabolism and matrix interactions. Stem Cell Transl. Med. 9(3), 364–376 (2020).
    • 41. Hazim RA, Karumbayaram S, Jiang M et al. Differentiation of RPE cells from integration-free iPS cells and their cell biological characterization. Stem Cell Res. Ther. 8(1), 217 (2017).
    • 42. Zhu D, Xie M, Gademann F et al. Protective effects of human iPS-derived retinal pigmented epithelial cells on retinal degenerative disease. Stem Cell Res. Ther. 11(1), 98 (2020).
    • 43. Qiu TG. Transplantation of human embryonic stem cell-derived retinal pigment epithelial cells (MA09-hRPE) in macular degeneration. NPJ Regen. Med. 4, 19 (2019).
    • 44. Wang Y, Tang Z, Gu P. Stem/progenitor cell-based transplantation for retinal degeneration: a review of clinical trials. Cell Death Dis. 11(9), 793 (2020).
    • 45. Li SY, Liu Y, Wang L et al. A phase I clinical trial of human embryonic stem cell-derived retinal pigment epithelial cells for early-stage Stargardt macular degeneration: 5-years' follow-up. Cell Prolif. 54(9), e13100 (2021).
    • 46. Mandai M, Watanabe A, Kurimoto Y et al. Autologous induced stem-cell–derived retinal cells for macular degeneration. N. Engl. J. Med. 376(11), 1038–1046 (2017).
    • 47. Takagi S, Mandai M, Gocho K et al. Evaluation of transplanted autologous induced pluripotent stem cell-derived retinal pigment epithelium in exudative age-related macular degeneration. Ophthalmol. Retina 3(10), 850–859 (2019).
    • 48. Chen S, Du K, Zou C. Current progress in stem cell therapy for Type 1 diabetes mellitus. Stem Cell Res. Ther. 11(1), 275 (2020).
    • 49. Corrêa-Giannella ML, Raposo do Amaral AS. Pancreatic islet transplantation. Diabetol. Metab. Syndr. 1(1), 9 (2009).
    • 50. Cayabyab F, Nih LR, Yoshihara E. Advances in pancreatic islet transplantation sites for the treatment of diabetes. Front. Endocrinol. 12, 732431 (2021).
    • 51. Bellin MD, Dunn TB. Transplant strategies for Type 1 diabetes: whole pancreas, islet and porcine beta cell therapies. Diabetologica 63(10), 2049–2056 (2020).
    • 52. Kondo Y, Toyoda T, Inagaki N, Osafune K. iPSC technology-based regenerative therapy for diabetes. J. Diabetes Investig. 9(2), 234–243 (2018).
    • 53. Ramzy A, Thompson DM, Ward-Hartstonge KA et al. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with Type 1 diabetes. Cell Stem Cell 28(12), 2047–2061.e5 (2021).• Breakthrough clinical study demonstrating the use of pluripotent stem cells in cardiovascular disease
    • 54. Shapiro AMJ, Thompson D, Donner TW et al. Insulin expression and C-peptide in Type 1 diabetes subjects implanted with stem cell-derived pancreatic endoderm cells in an encapsulation device. Cell Rep. Med. 2(12), 100466 (2021).• Associating clinical study demonstrating the use of pluripotent stem cells in cardiovascular disease.
    • 55. Silver SE, Barrs RW, Mei Y. Transplantation of human pluripotent stem cell-derived cardiomyocytes for cardiac regenerative therapy. Front. Cardiovasc. Med. 8, 707890 (2021).
    • 56. Turner D, Rieger AC, Balkan W, Hare JM. Clinical-based cell therapies for heart disease – current and future state. Rambam Maimonides Med. J. 11(2), e0015 (2020).
    • 57. Menasché P, Vanneaux V, Hagège A et al. Transplantation of human embryonic stem cell-derived cardiovascular progenitors for severe ischemic left ventricular dysfunction. J. Am. Coll. Cardiol. 71(4), 429–438 (2018).
    • 58. Sivandzade F, Cucullo L. Regenerative stem cell therapy for neurodegenerative diseases: an overview. Int. J. Mol. Sci. 22(4), 2153 (2021).
    • 59. Sugai K, Sumida M, Shofuda T et al. First-in-human clinical trial of transplantation of iPSC-derived NS/PCs in subacute complete spinal cord injury: study protocol. Regen. Ther. 18, 321–333 (2021).
    • 60. Takahashi J. iPS cell-based therapy for Parkinson's disease: a Kyoto trial. Regen. Ther. 13, 18–22 (2020). • An important study underscoring the efficacy of induced pluripotent stem cells in neurodegenerative diseases at a pre-clinical level. The clinical study was initiated on the basis of this study.
    • 61. Schweitzer JS, Song B, Herrington TM et al. Personalized iPSC-derived dopamine progenitor cells for Parkinson's disease. N. Engl. J. Med. 382(20), 1926–1932 (2020).
    • 62. Armijo E, Edwards G, Flores A et al. Induced pluripotent stem cell-derived neural precursors improve memory, synaptic and pathological abnormalities in a mouse model of Alzheimer's disease. Cells 10(7), 1802 (2021).
    • 63. Bhartiya D. Clinical translation of stem cells for regenerative medicine: a comprehensive analysis. Circ. Res.. 124(6), 840–842 (2019).
    • 64. Solomon S, Pitossi F, Rao MS. Banking on iPSC – is it doable and is it worthwhile. Stem Cell Rev. Rep. 11(1), 1–10 (2015).
    • 65. Jeske R, Bejoy J, Marzano M, Li Y. Human pluripotent stem cell-derived extracellular vesicles: characteristics and applications, mary ann liebert inc. Tissue Eng. Part B Rev. 26(2), 129–144 (2020).
    • 66. Lener T, Gimona M, Aigner L et al. Applying extracellular vesicles based therapeutics in clinical trials – an ISEV position paper. J. Extracell. Vesicles 4, 30087 (2015).
    • 67. He F, Li L, Fan R, Wang X, Chen X, Xu Y. Extracellular vesicles: an emerging regenerative treatment for oral disease. Front. Cell Dev. Biol. 9, 669011 (2021).
    • 68. Loyer X, Vion AC, Tedgui A, Boulanger CM. Microvesicles as cell–cell messengers in cardiovascular diseases. Circ. Res. 114(2), 345–353 (2014).
    • 69. Todorova D, Simoncini S, Lacroix R, Sabatier F, Dignat-George F. Extracellular vesicles in angiogenesis. Circ. Res. 120(10), 1658–1673 (2017).
    • 70. Collino F, Lopes JA, Tapparo M et al. Extracellular vesicles derived from induced pluripotent stem cells promote renoprotection in acute kidney injury model. Cells 9(2), 453 (2020).
    • 71. Ratajczak J, Miekus K, Kucia M et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia 20(5), 847–856 (2006).
    • 72. Yuan Y, Du W, Liu J et al. Stem cell-derived exosome in cardiovascular diseases: macro roles of micro particles. Front. Pharmocol. 9, 547 (2018).
    • 73. Wu Q, Wang J, Tan WLW et al. Extracellular vesicles from human embryonic stem cell-derived cardiovascular progenitor cells promote cardiac infarct healing through reducing cardiomyocyte death and promoting angiogenesis. Cell Death Dis. 11(5), 354 (2020).
    • 74. Khan M, Nickoloff E, Abramova T et al. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ. Res. 117(1), 52–64 (2015).
    • 75. Pang Y, Ma M, Wang D et al. Embryonic stem cell-derived exosomes attenuatetransverse aortic constriction induced heart failure by increasing angiogenesis. Front. Cardiovasc. Med. 8, 638771 (2021).
    • 76. el Harane N, Kervadec A, Bellamy V et al. Acellular therapeutic approach for heart failure: in vitro production of extracellular vesicles from human cardiovascular progenitors. Eur. Heart J. 39(20), 1835–1847 (2018).
    • 77. Correa BL, el Harane N, Desgres M et al. Extracellular vesicles fail to trigger the generation of new cardiomyocytes in chronically infarcted hearts. Theranostics 11(20), 10114–10124 (2021).
    • 78. Santoso MR, Ikeda G, Tada Y et al. Exosomes from induced pluripotent stem cell–derived cardiomyocytes promote autophagy for myocardial repair. J. Am. Heart Assoc. 9(6), e014345 (2020).
    • 79. Liu M, Qiu Y, Xue Z et al. Small extracellular vesicles derived from embryonic stem cells restore ovarian function of premature ovarian failure through PI3K/AKT signaling pathway. Stem Cell Res. Ther. 11(1), 3 (2020).
    • 80. Yu L, Liu S, Wang C et al. Embryonic stem cell-derived extracellular vesicles promote the recovery of kidney injury. Stem Cell Res. Ther. 12(1), 379 (2021).
    • 81. Saint-Pol J, Gosselet F, Duban-Deweer S, Pottiez G, Karamanos Y. Targeting and crossing the blood–brain barrier with extracellular vesicles. Cells 9(4), 851 (2020).
    • 82. Zhu Q, Ling X, Yang Y et al. Embryonic stem cells-derived exosomes endowed with targeting properties as chemotherapeutics delivery vehicles for glioblastoma therapy. Adv. Sci. 6(6), 1801899 (2019).
    • 83. Fan C, Zhang E, Joshi J, Yang J, Zhang J, Zhu W. Utilization of human induced pluripotent stem cells for cardiac repair. Front. Cell Dev. Biol. 8, 36 (2020).
    • 84. Adamiak M, Cheng G, Bobis-Wozowicz S et al. Induced pluripotent stem cell (iPSC)-derived extracellular vesicles are safer and more effective for cardiac repair than iPSCs. Circ. Res. 122(2), 296–309 (2018).
    • 85. Zhu Y, Wang Y, Zhao B et al. Comparison of exosomes secreted by induced pluripotent stem cell-derived mesenchymal stem cells and synovial membrane-derived mesenchymal stem cells for the treatment of osteoarthritis. Stem Cell Res. Ther. 8(1), 64 (2017).
    • 86. Han Z, Liu S, Pei Y et al. Highly efficient magnetic labelling allows MRI tracking of the homing of stem cell-derived extracellular vesicles following systemic delivery. J. Extracell. Vesicles 10(3), e12054 (2021).
    • 87. Kulkarni R, Bajaj M, Ghode S, Jalnapurkar S, Limaye L, Kale VP. Intercellular transfer of microvesicles from young mesenchymal stromal cells rejuvenates aged murine hematopoietic stem cells. Stem Cells 36(3), 420–433 (2018).
    • 88. Jalnapurkar S, Moirangthem RD, Singh S, Limaye L, Kale V. Microvesicles secreted by nitric oxide-primed mesenchymal stromal cells boost the engraftment potential of hematopoietic stem cells. Stem Cells 37(1), 128–138 (2019).
    • 89. Gorecka J, Kostiuk V, Fereydooni A et al. The potential and limitations of induced pluripotent stem cells to achieve wound healing. Stem Cell Res. Ther. 10(1), 1–10 (2019).
    • 90. Baldari S, Di Rocco G, Piccoli M, Pozzobon M, Muraca M, Toietta G. Challenges and strategies for improving the regenerative effects of mesenchymal stromal cell-based therapies. Int. J. Mol. Sci. 18(10), 2087 (2017).
    • 91. Simeon M, Dangwal S, Sachinidis A, Doss MX. Application of the pluripotent stem cells and genomics in cardiovascular research – what we have learnt and not learnt until now. Cells 10(11), 3112 (2021).
    • 92. O'Shea O, Steeg R, Chapman C, Mackintosh P, Stacey GN. Development and implementation of large-scale quality control for the European bank for induced pluripotent stem cells. Stem Cell Res. 45, 101773 (2020).
    • 93. Steeg R, Neubauer JC, Müller SC, Ebneth A, Zimmermann H. The EBiSC iPSC bank for disease studies. Stem Cell Res. 49, 102034 (2020).
    • 94. Huang CY, Liu CL, Ting CY et al. Human iPSC banking: barriers and opportunities. J. Biomed. Sci. 26(1), 87 (2019).
    • 95. Kawase E, Takada K, Suemori H. Kyoto hESC cell resource for regenerative medicine. Stem Cell Res. 49, 102020 (2020).
    • 96. Umekage M, Sato Y, Takasu N. Overview: an iPS cell stock at CiRA. Inflamm. Regen. 39, 17 (2019).
    • 97. Flahou C, Morishima T, Takizawa H, Sugimoto N. Fit-for-all iPSC-derived cell therapies and their evaluation in humanized mice with NK cell immunity. Front. Immunol. 12, 662360 (2021).
    • 98. Sackett SD, Brown ME, Tremmel DM, Ellis T, Burlingham WJ, Odorico JS. Modulation of human allogeneic and syngeneic pluripotent stem cells and immunological implications for transplantation. Transpl. Revs (Orlando, Fla.) 30(2), 61–70 (2016).
    • 99. Karagiannis P, Nakauchi A, Yamanaka S. Bringing induced pluripotent stem cell technology to the bedside. JMA J. 1(1), 6–14 (2018).
    • 100. Koga K, Wang B, Kaneko S. Current status and future perspectives of HLA-edited induced pluripotent stem cells. Inflamm. Regen. 40, 23 (2020).
    • 101. Jang Y, Choi J, Park N et al. Development of immunocompatible pluripotent stem cells via CRISPR-based human leukocyte antigen engineering. Exp. Mol. Med. 51(1), 1–11 (2019).
    • 102. Xu H, Wang B, Ono M et al. Targeted disruption of HLA genes via CRISPR-Cas9 generates iPSCs with enhanced immune compatibility. Cell Stem Cell 24(4), 566–578.e7 (2019).
    • 103. Han X, Wang M, Duan S et al. Generation of hypoimmunogenic human pluripotent stem cells. Proc. Natl Acad. Sci. USA 116(21), 10441–10446 (2019).