Quality control programmes for induced pluripotent stem cell-derived medicinal products
https://doi.org/10.30895/2221-996X-2025-25-2-127-140
Abstract
INTRODUCTION. Currently, there are no harmonised regulatory requirements for the quality control of human somatic cell therapy and tissue-engineered medicinal products that contain differentiated cells derived from induced pluripotent stem cells (iPSCs). This lack of uniform requirements underscores the need for approaches to developing quality control programmes and establishing critical quality attributes for iPSC-derived medicinal products within the Eurasian Economic Union (EAEU) regulatory framework.
AIM. This study aimed to systematise global regulatory experience and EAEU regulatory requirements for the development and justification of quality control programmes for iPSC-derived medicinal products.
DISCUSSION. Medicinal products derived from iPSCs are mainly used in the treatment of neurodegenerative, cardiovascular, and oncological diseases, diabetes, graft-versus-host disease, and eye diseases. Over the past decade, specific recommendations and requirements for the quality of clinical-grade iPSCs have been published by the Chinese Society for Stem Cell Research (CSSCR), the Japanese Ministry of Health, Labour, and Welfare (MHLW), the Global Alliance for iPSC Therapy (GAiT), and the European Bank of iPSC (EBiSC). The EAEU regulatory requirements for the quality of genetically modified cells have been in effect since 2025 (Chapter 32 of Decision No. 89 of the Council of the Eurasian Economic Commission “On Approval of the Rules for Assessment of Biological Medicines in the EAEU” of November 3, 2016). The list of critical quality attributes for clinical-grade iPSCs proposed by the GAiT generally corresponds to the EAEU regulatory framework and can be used in drawing up quality control programmes for iPSC-derived medicinal products in the Russian Federation and the EAEU. Quality control programmes for finished somatic cell therapy or tissue-engineered medicinal products derived from iPSCs should be based on the principle of quality attribute traceability from the starting material onwards. The production of iPSCs is a full-fledged production process that must comply with Good Manufacturing Practice (GMP) requirements for genetically modified cells. Specific quality controls for iPSCs should include tests for residual reprogramming vector DNA, markers of the undifferentiated state, and pluripotency as part of purity characterisation, identification, and potency evaluation, respectively.
CONCLUSIONS. A quality control programme for a finished iPSC-derived medicinal product should correspond to the type of differentiated cells and take into account the indications for use. Critical quality considerations for iPSC characterisation include demonstrating the absence of contaminating undifferentiated cells or cells with new immunogenic epitopes and confirming the identity and genetic stability of iPSCs. The considered quality assessment approaches provide a basis for developing both quality control strategies for iPSC-derived medicinal products and specifications for marketing authorisation according to the EAEU requirements.
Keywords
About the Authors
E. V. MelnikovaRussian Federation
Ekaterina V. Melnikova, Cand. Sci. (Biol.)
8/2, Petrovsky Blvd, Moscow 127051
O. A. Rachinskaya
Russian Federation
Olga A. Rachinskaya, Cand. Sci. (Biol.)
8/2, Petrovsky Blvd, Moscow 127051
I. S. Semenova
Russian Federation
Irina S. Semenova, Cand. Sci. (Biol.)
8/2, Petrovsky Blvd, Moscow 127051
V. A. Merkulov
Russian Federation
Vadim A. Merkulov, Dr. Sci. (Med.), Prof.
8/2, Petrovsky Blvd, Moscow 127051;
8/2 Trubetskaya St., Moscow 119991
References
1. Sullivan S, Stacey GN, Akazawa Ch, Aoyama N, Baptista R, Bedford P, et al. Quality control guidelines for clinical-grade human induced pluripotent stem cell lines. Regen Med. 2018; 13(7):859–66. https://doi.org/10.2217/rme-2018-0095
2. Yoshida S, Kato TM, Sato Y, Umekage M, Ichisaka T, Tsukahara M, et al. A clinical-grade HLA haplobank of human induced pluripotent stem cells matching approximately 40% of the Japanese population. Med. 2023;4(1):51–66.e10. https://doi.org/10.1016/j.medj.2022.10.003
3. Tian P, Elefanty A, Stanley EG, Durnall JC, Thompson LH, Elwood NJ. Creation of GMP-compliant iPSCs from banked umbilical cord blood. Front Cell Dev Biol. 2022;10:835321. https://doi.org/10.3389/fcell.2022.835321
4. Novoa JJ, Westra IM, Steeneveld E, Neves NF, Arendzen ChH, Rajaei B, et al. Good Manufacturing Practice-compliant human induced pluripotent stem cells: From bench to putative clinical products. Cytotherapy. 2024;26(6):556–66. https://doi.org/10.1016/j.jcyt.2024.02.021
5. Novoa J, Westra I, Steeneveld E, Neves NF, Daleman L, Asensio AB, et al. Validating human induced pluripotent stem cell-specific quality control tests for the release of an intermediate drug product in a Good Manufacturing Practice quality system. Cytotherapy. 2024;26(9):1105–17. https://doi.org/10.1016/j.jcyt.2024.04.004
6. 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. 2020;45:101773. https://doi.org/10.1016/j.scr.2020.101773
7. Steeg R, Mueller SC, Mah N, Holst B, Cabrera-Socorro A, Stacey GN, et al. EBiSC best practice: How to ensure optimal generation, qualification, and distribution of iPSC lines. Stem Cell Reports. 2021;16(8):1853–67. https://doi.org/10.1016/j.stemcr.2021.07.009
8. Shi J-X, Zhang K-Z. Advancements in autologous stem cell transplantation for Parkinson’s disease. Curr Stem Cell Res Ther. 2024;19(10):1321–7. https://doi.org/10.2174/1574888x19666230907112413
9. Cerneckis J, Cai H, Shi Y. Induced pluripotent stem cells (iPSCs): Molecular mechanisms of induction and applications. Signal Transduct Target Ther. 2024;9(1):112. https://doi.org/10.1038/s41392-024-01809-0
10. Schweitzer JS, Song B, Herrington TM, Park T-Y, Lee N, Ko S, et al. Personalized iPSC-derived dopamine progenitor cells for Parkinson’s disease. N Engl J Med. 2020;382(20):1926–32. https://doi.org/10.1056/nejmoa1915872
11. Choompoo N, Bartley OJM, Precious SV, Vinh N-N, Schnell Ch, Garcia A, et al. Induced pluripotent stem cells derived from the developing striatum as a potential donor source for cell replacement therapy for Huntington disease. Cytotherapy. 2021;23(2):111–8. https://doi.org/10.1016/j.jcyt.2020.06.001
12. Huang K, Hu S, Cheng K. A new era of cardiac cell therapy: Opportunities and challenges. Adv Healthc Mater. 2019; 8(2):e1801011. https://doi.org/10.1002/adhm.201801011
13. Finck AV, Blanchard T, Roselle CP, Golinelli G, June CH. Engineered cellular immunotherapies in cancer and beyond. Nat Med. 2022;28:678–89. https://doi.org/10.1038/s41591-022-01765-8
14. Cuesta-Gomez N, Verhoeff K, Jasra IT, Pawlick R, Dadheech N, Shapiro AMJ, et al. Characterization of stem-cell-derived islets during differentiation and after implantation. Cell Rep. 2022;40(8):111238. https://doi.org/10.1016/j.celrep.2022.111238
15. Ramzy A, Thompson DM, Ward-Hartstonge KA, Ivison S, Cook L, Garcia RV, et al. Implanted pluripotent stem-cell-derived pancreatic endoderm cells secrete glucose-responsive C-peptide in patients with type 1 diabetes. Cell Stem Cell. 2021;28(12):2047–61. https://doi.org/10.1016/j.stem.2021.10.003
16. Kelly K, Bloor AJC, Griffin JE, Radia R, Yeung DT, Rasko JEJ, et al. Two-year safety outcomes of iPS cell-derived mesenchymal stromal cells in acute steroid-resistant graft-versus-host disease. Nat Med. 2024;30(6):1556–8. https://doi.org/10.1038/s41591-024-02990-z
17. Kim JY, Nam Y, Rim YA, Ju JH. Review of the current trends in clinical trials involving induced pluripotent stem cells. Stem Cell Rev Rep. 2022;18(1):142–54. https://doi.org/10.1007/s12015-021-10262-3
18. Melnikova EV, Merkulov VA, Merkulova OV. Regulation for the translation of gene and cell therapy into medical practice in East Asian Countries. Bulletin of the Scientiffc Centre for Expert Evaluation of Medicinal Products. Regulatory Research and Medicine Evaluation. 2024;14(1):29-41 (In Russ.). https://doi.org/10.30895/1991-2919-2024-14-1-29-41
19. Zhang Y, Wei J, Cao J, Zhang K, Peng Y, Deng H, et al. Requirements for human-induced pluripotent stem cells. Cell Prolif. 2022;55(4):e13182. https://doi.org/10.1111/cpr.13182
20. Sullivanan S, Ginty P, McMahon S, May M, Solomon SL, Kurtz A, et al. The Global Alliance for iPSC Therapies (GAiT). Stem Cell Res. 2020;49:102036. https://doi.org/10.1016/j.scr.2020.102036
21. Wei W, Gaffney DJ, Chinnery PF. Cell reprogramming shapes the mitochondrial DNA landscape. Nat Commun. 2021; 12(1):5241. https://doi.org/10.1038/s41467-021-25482-x
22. Yamanaka S. Pluripotent stem cell-based cell therapy — promise and challenges. Cell Stem Cell. 2020;27(4):523–31. https://doi.org/10.1016/j.stem.2020.09.014
23. Wu J, Li T, Guo M, Ji J, Meng X, Fu T, et al. Treating a type 2 diabetic patient with impaired pancreatic islet function by personalized endoderm stem cell-derived islet tissue. Cell Discov. 2024;10(1):45. https://doi.org/10.1038/s41421-024-00662-3
24. Maruyama Y. Regulatory update of regenerative medicine in Japan. 7th India-Japan Medical Products Regulatory Symposium. New Delhi, India; 2024.
25. Khorolsky MD, Semenova IS, Melnikova EV, Olefir YuV. The use of short tandem repeat analysis for cell line authentication. BIOpreparations. Prevention, Diagnosis, Treatment. 2019;19(4):251–60 (In Russ.). https://doi.org/10.30895/2221-996X-2019-19-4-251-260
26. Melnikova EV, Rachinskaya OA, Trusov GA, Khorolsky MD, Semenova IS, Tereshkina NV, et al. Justification of methodological approaches to identification testing of biomedical cell products. BIOpreparations. Prevention, Diagnosis, Treatment. 2019;19(1):28–38 (In Russ.). https://doi.org/10.30895/2221-996X-2019-19-1-28-38
27. Pokrovsky NS, Vodyakova MA, Merkulov VA, Melnikova EV. Specific aspects and key parameters of validation for methods of phenotyping of cell lines included in cell therapy products. Immunologiya. 2024;45(3):343–54 (In Russ.). https://doi.org/10.33029/1816-2134-2024-45-3-343-354
28. Pokrovsky NS, Vodyakova MA, Merkulov VA, Melnikova EV. Recommendations for creating a validation algorithm of cell lines phenotyping methods. Immunologiya. 2024;45(4):505–17 (In Russ.). https://doi.org/10.33029/1816-2134-2024-45-4-505-517
29. Chaplenko AA, Merkulova OV, Semenova IS, Saifutdinova AR, Melnikova EV, Merkulov VA. Detection of mycoplasmas in eukaryotic cell lines by real-time PCR using various methods of sample concentration. Tsitologya. 2020;62(1):56–63 (In Russ.). https://doi.org/10.31857/S0041377120010034
30. Richards S, Aziz N, Bale Sh, Bick D, Das S, Gastier-Foster J, et al. Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24. https://doi.org/10.1038/gim.2015.30
31. Beckenkamp LR, da Silva CG, Hohendorff MLI, Ogliari KS. Manufacturing parameters for the creation of clinical-grade human-induced pluripotent stem cell lines from umbilical cord mesenchymal stromal cells. Stem Cells Transl Med. 2024;13(5):454–61. https://doi.org/10.1093/stcltm/szae010
32. Poetsch MS, Strano A, Guan K. Human induced pluripotent stem cells: From cell origin, genomic stability, and epigenetic memory to translational medicine. Stem Cells. 2022; 40(6):546–55. https://doi.org/10.1093/stmcls/sxac020
33. Scesa G, Adami R, Bottai D. iPSC preparation and epigenetic memory: Does the tissue origin matter? Cells. 2021;10(6):1470. https://doi.org/10.3390/cells10061470
34. Hargus G, Ehrlich M, Hallmann AL, Kuhlmann T. Human stem cell models of neurodegeneration: A novel approach to study mechanisms of disease development. Acta Neuropathol. 2014; 127(2):151–73. https://doi.org/10.1007/s00401-013-1222-6
35. Chlebanowska P, Sulkowski M, Skrzypek K, Tejchman A, Muszyńska A, Noroozi R, et al. Origin of the induced pluripotent stem cells affects their differentiation into dopaminergic neurons. Int J Mol Sci. 2020;21(16):5705. https://doi.org/10.3390/ijms21165705
Supplementary files
![]() |
1. Table S1. Examples of clinical trials of cell based medicinal products derived from induced pluripotent stem cells (iPSCs) | |
Subject | ||
Type | Исследовательские инструменты | |
Download
(393KB)
|
Indexing metadata ▾ |
![]() |
2. Table S2. Critical quality attributes for clinical-grade induced pluripotent stem cells (iPSCs) | |
Subject | ||
Type | Исследовательские инструменты | |
Download
(763KB)
|
Indexing metadata ▾ |
![]() |
3. Fig. S1. A quality control programme for obtaining pancreatic islets from induced pluripotent stem cells. PBMC, peripheral blood mononuclear cells; AFP, alpha-fetoprotein. | |
Subject | ||
Type | Исследовательские инструменты | |
Download
(414KB)
|
Indexing metadata ▾ |
Review
For citations:
Melnikova E.V., Rachinskaya O.A., Semenova I.S., Merkulov V.A. Quality control programmes for induced pluripotent stem cell-derived medicinal products. Biological Products. Prevention, Diagnosis, Treatment. 2025;25(2):127-140. (In Russ.) https://doi.org/10.30895/2221-996X-2025-25-2-127-140