Stem cell therapy: a revolutionary cure or a pandora’s box
- Eva Sykova and Serhiy Forostyak
Marei Stem Cell Research & Therapy (2025) 16:255
Abstract
This review article examines how stem cell therapies can cure various diseases and injuries while also discussing
the difficulties and moral conundrums that come with their application. The article focuses on the revolutionary
developments in stem cell research, especially the introduction of gene editing tools like CRISPR-Cas9, which can
potentially improve the safety and effectiveness of stem cell-based treatments. To guarantee the responsible use
of stem cells in clinical applications, it is also argued that standardizing clinical procedures and fortifying ethical
and regulatory frameworks are essential first steps. The assessment also highlights the substantial obstacles that still
need to be addressed, such as the moral dilemmas raised by the use of embryonic stem cells, the dangers of unlicensed
stem cell clinics, and the difficulties in obtaining and paying for care for patients. The study emphasizes
how critical it is to address these problems to stop exploitation, guarantee patient safety, and increase the accessibility
of stem cell therapy. The review also addresses the significance of thorough clinical trials, public education, and policy
development to guarantee that stem cell research may fulfill its full potential. The review concludes by describing
stem cell research as a promising but complicated topic that necessitates a thorough evaluation of both the hazards
and the benefits. To overcome the ethical, legal, and accessibility obstacles and eventually guarantee that stem cell
treatments may be safely and fairly included in conventional healthcare, it urges cooperation between the scientific
community, legislators, and the general public.
Keywords Stem cell research, Regenerative medicine, Induced pluripotent stem cells (iPSCs), Embryonic stem cells
(ESCs), Mesenchymal stem cells (MSCs), Hematopoietic stem cells (HSCs), Organoids, Disease modeling, Gene editing,
CRISPR-Cas9, Neurodegenerative disorders, Alzheimer’s disease, Parkinson’s disease, Spinal cord injury, Diabetes
treatment, Cardiovascular regeneration, Tissue engineering, Organ transplantation, Ethical concerns, Regulatory
challenges, Commercialization, Biotech investments, Unregulated stem cell clinics, Reproducibility issues, Clinical
applications, Tumor formation risk, Immune rejection, Standardization of protocols, Future of stem cell therapies
article original link: click here
References
1. Ja T. Embryonic stem cell lines, derived from human blastocysts. Science.
1998;282:1145–7.
2. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from
mouse embryonic and adult fibroblast cultures by defined factors. Cell.
2006;126(4):663–76.
3. Thompson E. James Till and Ernest McCulloch: The Team that Discovered
Stem Cells. The Rosen Publishing Group, Inc; 2020.
4. Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical
development success rates for investigational drugs. Nat Biotechnol.
2014;32(1):40–51.
5. Passweg JR, Halter J, Bucher C, Gerull S, Heim D, Rovó A, et al. Hematopoietic
stem cell transplantation: a review and recommendations
for follow-up care for the general practitioner. Swiss Med Wkly.
2012;142(4142):w13696–w13696.
6. Liang L, Sheha H, Li J, Tseng S. Limbal stem cell transplantation: new
progress and challenges. Eye. 2009;23(10):1946–53.
7. Rezania A, Bruin JE, Arora P, Rubin A, Batushansky I, Asadi A, et al. Reversal
of diabetes with insulin-producing cells derived in vitro from human
pluripotent stem cells. Nat Biotechnol. 2014;32(11):1121–33.
8. Takahashi K, Yamanaka S. Induced pluripotent stem cells in medicine and
biology. Development. 2013;140(12):2457–61.
9. Moy AB, Kamath A, Ternes S, Kamath J. The challenges to advancing
induced pluripotent stem cell-dependent cell replacement therapy. Med
Res Arch. 2023;11(11).
10. Sun C, Serra C, Kalicharan BH, Harding J, Rao M. Challenges and considerations
of preclinical development for iPSC-based myogenic cell therapy.
Cells. 2024;13(7):596.
11. Sato T, Vries RG, Snippert HJ, Van De Wetering M, Barker N, Stange DE,
et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a
mesenchymal niche. Nature. 2009;459(7244):262–5.
12. Clevers H. Modeling development and disease with organoids. Cell.
2016;165(7):1586–97.
13. Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME,
et al. Cerebral organoids model human brain development and microcephaly.
Nature. 2013;501(7467):373–9.
14. Schwank G, Koo BK, Sasselli V, Dekkers JF, Heo I, Demircan T, et al. Functional
repair of CFTR by CRISPR/Cas9 in intestinal stem cell organoids of
cystic fibrosis patients. Cell Stem Cell. 2013;13(6):653–8.
15. Fatehullah A, Tan SH, Barker N. Organoids as an in vitro human development
and disease model. Nat Cell Biol. 2016;18(3):246–54.
16. Camp JG, Sekine K, Gerber T, Loeffler-Wirth H, Binder H, Gac M, et al.
Multilineage communication regulates human liver bud development
from pluripotency. Nature. 2017;546(7659):533–8.
17. Han Y, Yang L, Lacko LA, Chen S. Human organoid models to study SARSCoV-
2 infection. Nat Methods. 2022;19(4):418–28.
18. Van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A,
et al. Prospective derivation of a living organoid biobank of colorectal
cancer patients. Cell. 2015;161(4):933–45.
19. Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, et al.
Vascularized and functional human liver from an iPSC-derived organ bud
transplant. Nature. 2013;499(7459):481–4.
20. Huch M, Koo BK. Modeling mouse and human development using organoid
cultures. Development. 2015;142(18):3113–25.
21. Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable
dual-–RNA–guided DNA endonuclease in adaptive bacterial
immunity. Science. 2012;337(6096):816–21.
22. Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, et al. Multiplex genome
engineering using CRISPR/Cas systems. Science. 2013;339(6121):819–23.
23. Hsu PD, Lander ES, Zhang F. Development and applications of CRISPRCas9
for genome engineering. Cell. 2014;157(6):1262–78.
24. Tsai SQ, Joung JK. Defining and improving the genome-wide specificities
of CRISPR–Cas9 nucleases. Nat Rev Genet. 2016;17(5):300–12.
25. Kleinstiver BP, Pattanayak V, Prew MS, Tsai SQ, Nguyen NT, Zheng Z, et al.
High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide
off-target effects. Nature. 2016;529(7587):490–5.
26. Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, et al.
Search-and-replace genome editing without double-strand breaks or
donor DNA. Nature. 2019;576(7785):149–57.
27. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al.
Repurposing CRISPR as an RNA-guided platform for sequence-specific
control of gene expression. Cell. 2013;152(5):1173–83.
28. Long C, Amoasii L, Mireault AA, McAnally JR, Li H, Sanchez-Ortiz E, et al.
Postnatal genome editing partially restores dystrophin expression in a
mouse model of muscular dystrophy. Science. 2016;351(6271):400–3.
29. DeWitt MA, Magis W, Bray NL, Wang T, Berman JR, Urbinati F, et al. Selection-
free genome editing of the sickle mutation in human adult hematopoietic
stem/progenitor cells. Sci Transl Med. 2016;8(360):360ra–134.
30. Frangoul H, Altshuler D, Cappellini MD, Chen YS, Domm J, Eustace BK,
et al. CRISPR-Cas9 gene editing for sickle cell disease and β-thalassemia.
N Engl J Med. 2021;384(3):252–60.
31. Dyer O., A Chinese researcher who made CRISPR babies, is sentenced to
three years in prison. BMJ Br Med J Online. 2020;36811.
32. Tyumentseva M, Tyumentsev A, Akimkin V. CRISPR/Cas9 landscape: Current
state and future perspectives. Int J Mol Sci. 2023;24(22):16077.
33. Angrist M, Barrangou R, Baylis F, Brokowski C, Burgio G, Caplan A, et al.
Reactions to the National Academies/Royal Society report on heritable
human genome editing. CRISPR J. 2020;3(5):332–49.
34. World Health Organization. Human genome editing: Recommendations.
In: Human genome editing: recommendations. 2021.
35. Kriks S, Shim JW, Piao J, Ganat YM, Wakeman DR, Xie Z, et al. Dopamine
neurons derived from human ES cells efficiently engraft in animal models
of Parkinson’s disease. Nature. 2011;480(7378):547–51.
36. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T, et al. Modeling
familial Alzheimer’s disease with induced pluripotent stem cells. Hum
Mol Genet. 2011;20(23):4530–9.
37. Marei HE, Althani A, Afifi N, Michetti F, Pescatori M, Pallini R, et al.
Gene expression profiling of embryonic human neural stem cells and
dopaminergic neurons from adult human substantia nigra. PLoS ONE.
2011;6(12): e28420.
38. Marei HE, Ahmed AE, Michetti F, Pescatori M, Pallini R, Casalbore P,
et al. The gene expression profile of adult human olfactory bulbs and
embryonic neural stem cells suggests distinct signaling pathways and
epigenetic control. PLoS ONE. 2012;7(4): e33542.
39. Marei HE, Farag A, Althani A, Afifi N, Abd-Elmaksoud A, Lashen S, et al.
Human olfactory bulb neural stem cells expressing hNGF restore
cognitive deficit in Alzheimer’s disease rat model. J Cell Physiol.
2015;230(1):116–30.
40. Marei HE, Lashen S, Farag A, Althani A, Afifi N, Rezk S, et al. Human olfactory
bulb neural stem cells mitigate movement disorders in a rat model
of Parkinson’s disease. J Cell Physiol. 2015;230(7):1614–29.
41. Chen MY, Chi CY, Zheng CW, Wang CH, Chiu IM. Coordinated actions of
neurogenesis and gliogenesis in nerve injury repair and neuroregeneration.
Int J Transl Med. 2024;4(4):810–30.
42. Chen Z, Zhao G. First-in-human transplantation of autologous induced
neural stem cell-derived dopaminergic precursors to treat Parkinson’s
disease. Sci Bull. 2023;68(22):2700–3.
43. Lindvall O, Kokaia Z. Stem cells in human neurodegenerative disorders—
time for clinical translation? J Clin Investig. 2010;120(1):29–40.
44. Wang Z. Assessing tumorigenicity in stem cell-derived therapeutic products:
a critical step in safeguarding regenerative medicine. Bioengineering.
2023;10(7):857.
45. Petrus-Reurer S, Romano M, Howlett S, Jones JL, Lombardi G, Saeb-Parsy
K. Immunological considerations and challenges for regenerative cellular
therapies. Commun Biol. 2021;4(1):798.
46. Pushp P, Nogueira DE, Rodrigues CA, Ferreira FC, Cabral JM, Gupta MK.
A concise review on induced pluripotent stem cell-derived cardiomyocytes
for personalized regenerative medicine. Stem Cell Rev Rep.
2021;17:748–76.
47. Hasan A, Deeb G, Rahal R, Atwi K, Mondello S, Marei HE, et al. Mesenchymal
stem cells in the treatment of traumatic brain injury. Front Neurol.
2017;8:28.
48. Müller P, Lemcke H, David R. Stem cell therapy in heart diseases–cell
types, mechanisms and improvement strategies. Cell Physiol Biochem.
2018;48(6):2607–55.
49. Garbern JC, Lee RT. Cardiac stem cell therapy and the promise of heart
regeneration. Cell Stem Cell. 2013;12(6):689–98.
50. Correia CD, Ferreira A, Fernandes MT, Silva BM, Esteves F, Leitão HS,
et al. Are we on the road to success for human stem cells for cardiac
disease modeling and preclinical and clinical applications? Cells.
2023;12(13):1727.
51. Bartunek J, Behfar A, Dolatabadi D, Vanderheyden M, Ostojic M, Dens J,
et al. Cardiopoietic stem cell therapy in heart failure: the C-CURE (Cardiopoietic
stem Cell therapy in heart failure) multicenter randomized trial
with lineage-specified biologics. J Am Coll Cardiol. 2013;61(23):2329–38.
52. Menasché P. Cell therapy trials for heart regeneration—lessons learned
and future directions. Nat Rev Cardiol. 2018;15(11):659–71.
53. Makkar RR, Kereiakes DJ, Aguirre F, Kowalchuk G, Chakravarty T, Malliaras
K, et al. Intracoronary ALLogeneic heart STem cells to Achieve Myocardial
Regeneration (ALLSTAR): a randomized, placebo-controlled, doubleblinded
trial. Eur Heart J. 2020;41(36):3451–8.
54. Der Sarkissian S, Lévesque T, Noiseux N. Optimizing stem cells for cardiac
repair: current status and new frontiers in regenerative cardiology. World
J Stem Cells. 2017;9(1):9.
55. Li ZH, Wang J, Xu JP, Wang J, Yang X. Recent advances in CRISPR-based
genome editing technology and its applications in cardiovascular
research. Mil Med Res. 2023;10(1):12.
56. Duan B. State-of-the-art review of 3D bioprinting for cardiovascular tissue
engineering. Ann Biomed Eng. 2017;45:195–209.
57. Sun R, Li X, Liu M, Zeng Y, Chen S, Zhang P. Advances in stem cell therapy
for cardiovascular disease. Int J Mol Med. 2016;38(1):23–9.
58. Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, et al. Mesenchymal
stem cells modified with Akt prevent remodeling and restore the
performance of infarcted hearts. Nat Med. 2003;9(9):1195–201.
59. Habiba UE, Khan N, Greene DL, Ahmad K, Shamim S, Umer A. Meta-analysis
shows that mesenchymal stem cell therapy can be a possible diabetes
treatment. Front Endocrinol. 2024;15:1380443.
60. Zhang Y, Chen W, Feng B, Cao H. The clinical efficacy and safety of stem
cell therapy for diabetes mellitus: a systematic review and meta-analysis.
Aging Dis. 2020;11(1):141.
61. Luo Y, Yu P, Liu J. The efficiency of stem cell differentiation into functional
beta cells for treating insulin-requiring diabetes: Recent Advances and
current challenges. Endocrine. 2024;1–14.
62. Carlsson PO, Espes D, Sisay S, Davies LC, Smith CE, Svahn MG. Umbilical
cord-derived mesenchymal stromal cells preserve endogenous insulin
production in type 1 diabetes: a phase I/II randomized double-blind
placebo-controlled trial. Diabetologia. 2023;66(8):1431–41.
63. Zhao Y, Knight CM, Jiang Z, Delgado E, Van Hoven AM, Ghanny S, et al.
Stem Cell Educator therapy in type 1 diabetes: from the bench to clinical
trials. Autoimmun Rev. 2022;21(5): 103058.
64. Kaur A, Midha S, Giri S, Mohanty S. Functional skin grafts: where biomaterials
meet stem cells. Stem Cells Int. 2019;2019(1):1286054.
65. Hu C, Li L. In vitro culture of isolated primary hepatocytes and stem
cell-derived hepatocyte-like cells for liver regeneration. Protein Cell.
2015;6(8):562–74.
66. Skolasinski SD, Panoskaltsis-Mortari A. Lung tissue bioengineering
for chronic obstructive pulmonary disease: overcoming the need for
lung transplantation from human donors. Expert Rev Respir Med.
2019;13(7):665–78.
67. Ghosh D, Mehta N, Patil A, Sengupta J. Ethical issues in human embryonic
stem cells (hESCs) biomedical use. J Reprod Health Med. 2016;2:S37-47.
68. Park SJ, Kim YY, Han JY, Kim SW, Kim H, Ku SY. Advancements in human
embryonic stem cell research: clinical applications and ethical issues.
Tissue Eng Regen Med. 2024;21(3):379–94.
69. Lo B, Parham L. Ethical issues in stem cell research. Endocr Rev.
2009;30(3):204–13.
70. Michler RE. Stem cell therapy for heart failure. Cardiol Rev.
2014;22(3):105–16.
71. Laflamme MA, Murry CE. Regenerating the heart. Nat Biotechnol.
2005;23(7):845–56.
72. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, et al.
Induction of pluripotent stem cells from adult human fibroblasts by
defined factors. Cell. 2007;131(5):861–72.
73. Herberts CA, Kwa MS, Hermsen HP. Risk factors in the development of
stem cell therapy. J Transl Med. 2011;9:1–14.
74. King NM, Perrin J. Ethical issues in stem cell research and therapy. Stem
Cell Res Ther. 2014;5:1–6.
75. Langford L, Foong P. Unproven stem cell therapies: evaluating patients’
capacity to give informed consent. Griffith Law Rev. 2024;33(1):58–88.
76. Fink DW Jr. FDA regulation of stem cell-based products. Science.
2009;324(5935):1662–3.
77. Turner L. US stem cell clinics, patient safety, and the FDA. Trends Mol
Med. 2015;21(5):271–3.
78. Volpato V, Smith J, Sandor C, Ried JS, Baud A, Handel A, et al. Reproducibility
of molecular phenotypes after long-term differentiation to
human iPSC-derived neurons: a multi-site omics study. Stem cell Rep.
2018;11(4):897–911.
79. Lyu Z, Xin M, Oyston DR, Xue T, Kang H, Wang X, et al. Cause and consequence
of heterogeneity in human mesenchymal stem cells: challenges
in clinical application. Pathol Res Pract. 2024;260: 155354.
80. Mummery CL, Zhang J, Ng ES, Elliott DA, Elefanty AG, Kamp TJ. Differentiation
of human embryonic stem cells and induced pluripotent stem cells
to cardiomyocytes: a methods overview. Circ Res. 2012;111(3):344–58.
81. Halme DG, Kessler DA. FDA regulation of stem-cell-based therapies. N
Engl J Med. 2006;355(16):1730.
82. Seet WT, Mat Afandi MA, Shamsuddin SA, Lokanathan Y, Ng MH, Maarof
M. Current good manufacturing practice (cGMP) Facility and stem cell
production. In: Stem Cell Production: Processes, Practices and Regulations.
Springer; 2022. p. 37–68.
83. Aly RM. The current state of stem cell-based therapies: an overview. Stem
Cell Investig. 2020;7.
84. Lee H, Cho HJ, Han Y, Lee SH. Mid-to long-term efficacy and safety of
stem cell therapy for acute myocardial infarction: a systematic review and
meta-analysis. Stem Cell Res Ther. 2024;15(1):290.
85. Bahari M, Mokhtari H, Yeganeh F. Stem cell therapy, the market, the
opportunities and the threat. Int J Mol Cell Med. 2023;12(3):310.
86. Kamenova K, Caulfield T. Stem cell hype: media portrayal of therapy translation.
Sci Transl Med. 2015;7(278):278ps4.
87. Takashima K, Minari J, Chan S, Muto K. Hope for the best, but prepare for
the worst: Social media posted by participants in stem cell clinical trials.
Regen Ther. 2023;24:294–7.
88. BS S, Chua K, Tan B. Ensuring Safety in Stem Cell Therapies: A Review of
Tumorigenic Risks and Mitigation Strategies. 2024.
89. Mezey É. Human mesenchymal stem/stromal cells in immune regulation
and therapy. Stem Cells Transl Med. 2022;11(2):114–34.
90. Sessa G, Adami R, Bottai D. iPSC preparation and epigenetic memory:
Does the tissue origin matter? Cells. 2021;10(6):1470.
91. Haworth R, Sharpe M. Accept or reject: the role of immune tolerance in
developing stem cell therapies and possible future approaches. Toxicol
Pathol. 2021;49(7):1308–16.
92. Singh B. Cutting Edges in Human Germline Editing Reconciling Scientific
Progress With Rogues and Legal Framework: Global Observatory Its
Inherent Conundrums. In: Ethical Dimensions of AI Development. IGI
Global; 2025. p. 227–50.
93. Doxzen K, Halpern J. Focusing on Human Rights: a framework for CRISPR
germline genome editing ethics and regulation. Perspect Biol Med.
2020;63(1):44–53.
