Standardisation is the key to the sustained, rapid and healthy development of stem cell-based therapy

Jing Zhang , Moran Suo , Jinzuo Wang , Xin Liu , Huagui Huang , Kaizhong Wang , Xiangyan Liu , Tianze Sun , Zhonghai Li , Jing Liu

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1646

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1646 DOI: 10.1002/ctm2.1646
REVIEW

Standardisation is the key to the sustained, rapid and healthy development of stem cell-based therapy

Author information +
History +
PDF

Abstract

Background: Stem cell-based therapy (SCT) is an important component of regenerative therapy that brings hope to many patients. After decades of development, SCT has made significant progress in the research of various diseases, and the market size has also expanded significantly. The transition of SCT from small-scale, customized experiments to routine clinical practice requires the assistance of standards. Many countries and international organizations around the world have developed corresponding SCT standards, which have effectively promoted the further development of the SCT industry.

Methods: We conducted a comprehensive literature review to introduce the clinical application progress of SCT and focus on the development status of SCT standardization.

Results: We first briefly introduced the types and characteristics of stem cells, and summarized the current clinical application and market development of SCT. Subsequently, we focused on the development status of SCT-related standards as of now from three levels: the International Organization for Standardization (ISO), important international organizations, and national organizations. Finally, we provided perspectives and conclusions on the significance and challenges of SCT standardization.

Conclusions: Standardization plays an important role in the sustained, rapid and healthy development of SCT.

Keywords

clinical application / regenerative medicine / standardisation / stem cell therapy

Cite this article

Download citation ▾
Jing Zhang, Moran Suo, Jinzuo Wang, Xin Liu, Huagui Huang, Kaizhong Wang, Xiangyan Liu, Tianze Sun, Zhonghai Li, Jing Liu. Standardisation is the key to the sustained, rapid and healthy development of stem cell-based therapy. Clinical and Translational Medicine, 2024, 14(4): e1646 DOI:10.1002/ctm2.1646

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Zakrzewski W, Dobrzyński M, Szymonowicz M, Rybak Z. Stem cells: past, present, and future. Stem Cell Res Ther. 2019;10(1):68.

[2]

Bacakova L, Zarubova J, Travnickova M, et al. Stem cells: their source, potency and use in regenerative therapies with focus on adipose-derived stem cells—a review. Biotechnol Adv. 2018;36(4):1111-1126.

[3]

Ntege EH, Sunami H, Shimizu Y. Advances in regenerative therapy: a review of the literature and future directions. Regenerat Therap. 2020;14:136-153.

[4]

Yamanaka S. Pluripotent stem cell-based cell therapy-promise and challenges. Cell Stem Cell. 2020;27(4):523-531.

[5]

Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663-676.

[6]

Ratajczak MZ, Ratajczak J, Kucia M. Very small embryonic-like stem cells (VSELs). Circ Res. 2019;124(2):208-210.

[7]

Suszynska M, Zuba-Surma EK, Maj M, et al. The proper criteria for identification and sorting of very small embryonic-like stem cells, and some nomenclature issues. Stem Cells Dev. 2014;23(7):702-713.

[8]

Lan T, Luo M, Wei X. Mesenchymal stem/stromal cells in cancer therapy. J Hematol Oncol. 2021;14(1):195.

[9]

Naji A, Eitoku M, Favier B, Deschaseaux F, Rouas-Freiss N, Suganuma N. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol Life Sci. 2019;76(17):3323-3348.

[10]

Drissen R, Thongjuea S, Theilgaard-Mönch K, Nerlov C. Identification of two distinct pathways of human myelopoiesis. Sci Immunol. 2019;4(35):eaau7148.

[11]

Kowalski K, Dos Santos M, Maire P, Ciemerych MA, Brzoska E. Induction of bone marrow-derived cells myogenic identity by their interactions with the satellite cell niche. Stem Cell Res Ther. 2018;9(1):258.

[12]

Dulak J, Szade K, Szade A, Nowak W, Józkowicz A. Adult stem cells: hopes and hypes of regenerative medicine. Acta Biochim Pol. 2015;62(3):329-337.

[13]

Shekkeris AS, Jaiswal PK, Khan WS. Clinical applications of mesenchymal stem cells in the treatment of fracture non-union and bone defects. Curr Stem Cell Res Ther. 2012;7(2):127-133.

[14]

Han Y, Yang J, Fang J, et al. The secretion profile of mesenchymal stem cells and potential applications in treating human diseases. Signal Transduct Targeted Therap. 2022;7(1):92.

[15]

Al-Ghadban S, Bunnell BA. Adipose tissue-derived stem cells: immunomodulatory effects and therapeutic potential. Physiology (Bethesda, Md). 2020;35(2):125-133.

[16]

Thomas ED, Lochte HL, Lu WC, Ferrebee JW. Intravenous infusion of bone marrow in patients receiving radiation and chemotherapy. N Engl J Med. 1957;257(11):491-496.

[17]

Thomas ED, Lochte HL, Cannon JH, Sahler OD, Ferrebee JW. Supralethal whole body irradiation and isologous marrow transplantation in man. J Clin Invest. 1959;38:1709-1716. 10 Pt 1-2.

[18]

Thomas ED, Storb R, Fefer A, et al. Aplastic anaemia treated by marrow transplantation. Lancet (London, England). 1972;1(7745):284-289.

[19]

Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292(5819):154-156.

[20]

Thomson JA, Itskovitz-Eldor J, Shapiro SS, et al. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145-1147.

[21]

Bretzner F, Gilbert F, Baylis F, Brownstone RM. Target populations for first-in-human embryonic stem cell research in spinal cord injury. Cell Stem Cell. 2011;8(5):468-475.

[22]

Schwartz SD, Hubschman JP, Heilwell G, et al. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet (London, England). 2012;379(9817):713-720.

[23]

Menasché P, Vanneaux V, Hagège A, et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report. Eur Heart J. 2015;36(30):2011-2017.

[24]

Mazid MA, Ward C, Luo Z, et al. Rolling back human pluripotent stem cells to an eight-cell embryo-like stage. Nature. 2022;605(7909):315-324.

[25]

Wei W, Luo J. Thoughts on chemistry, manufacturing, and control of cell therapy products for clinical application. Hum Gene Ther. 2019;30(2):119-126.

[26]

Fang WH. Governmental regulations and increasing food and drug administration oversight of regenerative medicine products: what's new in 2020?Arthroscopy. 2020;36(10):2765-2770.

[27]

Cossu G, Birchall M, Brown T, et al. Lancet Commission: stem cells and regenerative medicine. Lancet (London, England). 2018;391(10123):883-910.

[28]

Lovell-Badge R, Anthony E, Barker RA, et al. ISSCR Guidelines for Stem Cell Research and Clinical Translation: the 2021 update. Stem Cell Rep. 2021;16(6):1398-1408.

[29]

Turner L. ISSCR's Guidelines for Stem Cell Research and Clinical Translation: supporting development of safe and efficacious stem cell-based interventions. Stem Cell Rep. 2021;16(6):1394-1397.

[30]

Arcidiacono JA, Bauer SR, Kaplan DS, Allocca CM, Sarkar S, Lin-Gibson S. FDA and NIST collaboration on standards development activities supporting innovation and translation of regenerative medicine products. Cytotherapy. 2018;20(6):779-784.

[31]

Liu G, David BT, Trawczynski M, Fessler RG. Advances in pluripotent stem cells: history, mechanisms, technologies, and applications. Stem Cell Rev Rep. 2020;16(1):3-32.

[32]

Wobus AM, Boheler KR. Embryonic stem cells: prospects for developmental biology and cell therapy. Physiol Rev. 2005;85(2):635-678.

[33]

Riveiro AR, Brickman JM. From pluripotency to totipotency: an experimentalist's guide to cellular potency. Development. 2020;147(16):dev189845.

[34]

Denker HW. Autonomy in the development of stem cell-derived embryoids: sprouting blastocyst-like cysts, and ethical implications. Cells. 2021;10(6):1461.

[35]

Pereira Daoud AM, Popovic M, Dondorp WJ, et al. Modelling human embryogenesis: embryo-like structures spark ethical and policy debate. Hum Reprod Update. 2020;26(6):779-798.

[36]

Swijnenburg RJ, Schrepfer S, Govaert JA, et al. Immunosuppressive therapy mitigates immunological rejection of human embryonic stem cell xenografts. Proc Nat Acad Sci USA. 2008;105(35):12991-12996.

[37]

Liu C, Moten A, Ma Z, Lin HK. The foundational framework of tumors: gametogenesis, p53, and cancer. Semin Cancer Biol. 2022;81:193-205.

[38]

Grinnemo KH, Sylvén C, Hovatta O, Dellgren G, Corbascio M. Immunogenicity of human embryonic stem cells. Cell Tissue Res. 2008;331(1):67-78.

[39]

Araki R, Uda M, Hoki Y, et al. Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells. Nature. 2013;494(7435):100-104.

[40]

Guha P, Morgan JW, Mostoslavsky G, Rodrigues NP, Boyd AS. Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells. Cell Stem Cell. 2013;12(4):407-412.

[41]

Pan G, Mu Y, Hou L, Liu J. Examining the therapeutic potential of various stem cell sources for differentiation into insulin-producing cells to treat diabetes. Annales d'endocrinologie. 2019;80(1):47-53.

[42]

Assessment of established techniques to determine developmental and malignant potential of human pluripotent stem cells. Nat Commun. 2018;9(1):1925.

[43]

Khan FA, Almohazey D, Alomari M, Almofty SA. Isolation, culture, and functional characterization of human embryonic stem cells: current trends and challenges. Stem Cells Int. 2018;2018:1429351.

[44]

Gao X, Sprando RL, Yourick JJ. A rapid and highly efficient method for the isolation, purification, and passaging of human-induced pluripotent stem cells. Cellular Reprogr. 2018;20(5):282-288.

[45]

Cao S, Loh K, Pei Y, Zhang W, Han J. Overcoming barriers to the clinical utilization of iPSCs: reprogramming efficiency, safety and quality. Protein Cell. 2012;3(11):834-845.

[46]

Guerin CL, Blandinières A, Planquette B, et al. Very small embryonic-like stem cells are mobilized in human peripheral blood during hypoxemic COPD exacerbations and pulmonary hypertension. Stem Cell Rev Rep. 2017;13(4):561-566.

[47]

Lahlil R, Scrofani M, Barbet R, Tancredi C, Aries A, Hénon P. VSELs maintain their pluripotency and competence to differentiate after enhanced ex vivo expansion. Stem Cell Rev Rep. 2018;14(4):510-524.

[48]

Filidou E, Kandilogiannakis L, Tarapatzi G, et al. A simplified and effective approach for the isolation of small pluripotent stem cells derived from human peripheral blood. Biomedicines. 2023;11(3).

[49]

Liu DD, He JQ, Sinha R, et al. Purification and characterization of human neural stem and progenitor cells. Cell. 2023;186(6):1179-1194. e1115.

[50]

Sousa BR, Parreira RC, Fonseca EA, et al. Human adult stem cells from diverse origins: an overview from multiparametric immunophenotyping to clinical applications. Cytometry A. 2014;85(1):43-77.

[51]

Yu H, Huang Y, Yang L. Research progress in the use of mesenchymal stem cells and their derived exosomes in the treatment of osteoarthritis. Ageing Res Rev. 2022;80:101684.

[52]

Mizukami A, Swiech K. Mesenchymal stromal cells: from discovery to manufacturing and commercialization. Stem Cells Int. 2018;2018:4083921.

[53]

Beane OS, Darling EM. Isolation, characterization, and differentiation of stem cells for cartilage regeneration. Ann Biomed Eng. 2012;40(10):2079-2097.

[54]

Clevers H, Watt FM. Defining adult stem cells by function, not by phenotype. Annu Rev Biochem. 2018;87(1):1015-1027.

[55]

Furlani D, Ugurlucan M, Ong L, et al. Is the intravascular administration of mesenchymal stem cells safe? Mesenchymal stem cells and intravital microscopy. Microvasc Res. 2009;77(3):370-376.

[56]

Sanchez-Diaz M, Quiñones-Vico MI, Sanabria de la Torre R, et al. Biodistribution of mesenchymal stromal cells after administration in animal models and humans: a systematic review. J Clin Med. 2021;10(13).

[57]

Gholamrezanezhad A, Mirpour S, Bagheri M, et al. In vivo tracking of 111In-oxine labeled mesenchymal stem cells following infusion in patients with advanced cirrhosis. Nucl Med Biol. 2011;38(7):961-967.

[58]

Koç ON, Gerson SL, Cooper BW, et al. Rapid hematopoietic recovery after coinfusion of autologous-blood stem cells and culture-expanded marrow mesenchymal stem cells in advanced breast cancer patients receiving high-dose chemotherapy. J Clin Oncol. 2000;18(2):307-316.

[59]

Sokal EM, Lombard CA, Roelants V, et al. Biodistribution of liver-derived mesenchymal stem cells after peripheral injection in a hemophilia A patient. Transplantation. 2017;101(8):1845-1851.

[60]

Sood V, Mittal BR, Bhansali A, et al. Biodistribution of 18F-FDG-labeled autologous bone marrow-derived stem cells in patients with type 2 diabetes mellitus: exploring targeted and intravenous routes of delivery. Clin Nucl Med. 2015;40(9):697-700.

[61]

Lezaic L, Socan A, Peitl PK, et al. Imaging and 1-day kinetics of intracoronary stem cell transplantation in patients with idiopathic dilated cardiomyopathy. Nucl Med Biol. 2016;43(7):410-414.

[62]

Penicka M, Lang O, Widimsky P, et al. One-day kinetics of myocardial engraftment after intracoronary injection of bone marrow mononuclear cells in patients with acute and chronic myocardial infarction. Heart. 2007;93(7):837-841.

[63]

Simari RD, Pepine CJ, Traverse JH, et al. Bone marrow mononuclear cell therapy for acute myocardial infarction: a perspective from the cardiovascular cell therapy research network. Circ Res. 2014;114(10):1564-1568.

[64]

Laird DJ, von Andrian UH, Wagers AJ. Stem cell trafficking in tissue development, growth, and disease. Cell. 2008;132(4):612-630.

[65]

Magnon C, Lucas D, Frenette PS. Trafficking of stem cells. Methods Mol Biol. 2011;750:3-24.

[66]

Henriksson HB, Papadimitriou N, Hingert D, Baranto A, Lindahl A, Brisby H. The traceability of mesenchymal stromal cells after injection into degenerated discs in patients with low back pain. Stem Cells Dev. 2019;28(17):1203-1211.

[67]

Huerta CT, Voza FA, Ortiz YY, Liu ZJ, Velazquez OC. Targeted cell delivery of mesenchymal stem cell therapy for cardiovascular disease applications: a review of preclinical advancements. Front Cardiovasc Med. 2023;10:1236345.

[68]

Sobacchi C, Palagano E, Villa A, Menale C. Soluble factors on stage to direct mesenchymal stem cells fate. Front Bioeng Biotechnol. 2017;5:32.

[69]

Reinhardt M, Bader A, Giri S. Devices for stem cell isolation and delivery: current need for drug discovery and cell therapy. Expert Rev Med Devices. 2015;12(3):353-364.

[70]

Duckers HJ, Houtgraaf J, Hehrlein C, et al. Final results of a phase IIa, randomised, open-label trial to evaluate the percutaneous intramyocardial transplantation of autologous skeletal myoblasts in congestive heart failure patients: the SEISMIC trial. EuroIntervention. 2011;6(7):805-812.

[71]

Ng NN, Thakor AS. Locoregional delivery of stem cell-based therapies. Sci Transl Med. 2020;12(547):eaba4564.

[72]

Qayyum AA, Mouridsen M, Nilsson B, et al. Danish phase II trial using adipose tissue derived mesenchymal stromal cells for patients with ischaemic heart failure. ESC Heart Fail. 2023;10(2):1170-1183.

[73]

Raval AN, Johnston PV, Duckers HJ, et al. Point of care, bone marrow mononuclear cell therapy in ischemic heart failure patients personalized for cell potency: 12-month feasibility results from CardiAMP heart failure roll-in cohort. Int J Cardiol. 2021;326:131-138.

[74]

Bartunek J, Terzic A. Optimized catheter system demonstrates utility for endomyocardial delivery of cardiopoietic stem cells in target patients with heart failure. Tex Heart Inst J. 2023;50(5):e238247.

[75]

Konstanty-Kalandyk J, Piątek J, Miszalski-Jamka T, et al. The combined use of transmyocardial laser revascularisation and intramyocardial injection of bone-marrow derived stem cells in patients with end-stage coronary artery disease: one year follow-up. Kardiol Pol. 2013;71(5):485-492.

[76]

Moreira Rde C, Haddad AF, Silva SA, et al. Intracoronary stem-cell injection after myocardial infarction: microcirculation sub-study. Arq Bras Cardiol. 2011;97(5):420-426.

[77]

Medicetty S, Wiktor D, Lehman N, et al. Percutaneous adventitial delivery of allogeneic bone marrow-derived stem cells via infarct-related artery improves long-term ventricular function in acute myocardial infarction. Cell Transplant. 2012;21(6):1109-1120.

[78]

Siminiak T, Fiszer D, Jerzykowska O, et al. Percutaneous trans-coronary-venous transplantation of autologous skeletal myoblasts in the treatment of post-infarction myocardial contractility impairment: the POZNAN trial. Eur Heart J. 2005;26(12):1188-1195.

[79]

Brecknell JE, Fawcett JW. A device for the implantation of multiple cellular deposits into a large volume of brain from a single cannula site. Exp Neurol. 1996;138(2):338-344.

[80]

Xue J, Wu Y, Bao Y, et al. Clinical considerations in Parkinson's disease cell therapy. Ageing Res Rev. 2023;83:101792.

[81]

Silvestrini MT, Yin D, Coppes VG, et al. Radially branched deployment for more efficient cell transplantation at the scale of the human brain. Stereotact Funct Neurosurg. 2013;91(2):92-103.

[82]

Riley J, Glass J, Feldman EL, et al. Intraspinal stem cell transplantation in amyotrophic lateral sclerosis: a phase I trial, cervical microinjection, and final surgical safety outcomes. Neurosurgery. 2014;74(1):77-87.

[83]

Riley JP, Raore B, Taub JS, Federici T, Boulis NM. Platform and cannula design improvements for spinal cord therapeutics delivery. Neurosurgery. 2011;69:ons147-154.

[84]

Stemcell Clinical Research DB.

[85]

Schwartz SD, Regillo CD, Lam BL, 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 (London, England). 2015;385(9967):509-516.

[86]

Song WK, Park KM, Kim HJ, et al. Treatment of macular degeneration using embryonic stem cell-derived retinal pigment epithelium: preliminary results in Asian patients. Stem Cell Rep. 2015;4(5):860-872.

[87]

da Cruz L, Fynes K, Georgiadis O, et al. Phase 1 clinical study of an embryonic stem cell-derived retinal pigment epithelium patch in age-related macular degeneration. Nat Biotechnol. 2018;36(4):328-337.

[88]

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. 2018;4(1):50.

[89]

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. 2021;54(9):e13100.

[90]

Limnios IJ, Chau YQ, Skabo SJ, Surrao DC, O'Neill HC. Efficient differentiation of human embryonic stem cells to retinal pigment epithelium under defined conditions. Stem Cell Res Ther. 2021;12(1):248.

[91]

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. 2021;28(12):2047-2061. e2045.

[92]

Huang L, Zhang S, Wu J, et al. Immunity-and-matrix-regulatory cells enhance cartilage regeneration for meniscus injuries: a phase I dose-escalation trial. Signal Transduct Target Therap. 2023;8(1):417.

[93]

Shin JH, Ryu CM, Yu HY, et al. Safety of human embryonic stem cell-derived mesenchymal stem cells for treating interstitial cystitis: a Phase I study. Stem Cells Transl Med. 2022;11(10):1010-1020.

[94]

Mandai M, Watanabe A, Kurimoto Y, et al. Autologous induced stem-cell-derived retinal cells for macular degeneration. N Engl J Med. 2017;376(11):1038-1046.

[95]

Bloor AJC, Patel A, Griffin JE, et al. Production, safety and efficacy of iPSC-derived mesenchymal stromal cells in acute steroid-resistant graft versus host disease: a phase I, multicenter, open-label, dose-escalation study. Nat Med. 2020;26(11):1720-1725.

[96]

Du H, Huo Z, Chen Y, et al. Induced pluripotent stem cells and their applications in amyotrophic lateral sclerosis. Cells. 2023;12(6):971.

[97]

Morimoto S, Takahashi S, Ito D, et al. Phase 1/2a clinical trial in ALS with ropinirole, a drug candidate identified by iPSC drug discovery. Cell Stem Cell. 2023;30(6):766-780. e769.

[98]

Gurusamy N, Alsayari A, Rajasingh S, Rajasingh J. Chapter one—adult stem cells for regenerative therapy. In: Teplow DB, ed. Progress in Molecular Biology and Translational Science. Academic Press; 2018:1-22.

[99]

Bhartiya D, Jha N, Tripathi A, Tripathi A. Very small embryonic-like stem cells have the potential to win the three-front war on tissue damage, cancer, and aging. Front Cell Dev Biol. 2022;10:1061022.

[100]

Guerin CL, Loyer X, Vilar J, et al. Bone-marrow-derived very small embryonic-like stem cells in patients with critical leg ischaemia: evidence of vasculogenic potential. Thromb Haemostasis. 2015;113(5):1084-1094.

[101]

Jokura Y, Yano K, Yamato M. Comparison of the new Japanese legislation for expedited approval of regenerative medicine products with the existing systems in the USA and European Union. J Tissue Eng Regen Med. 2018;12(2):e1056-e1062.

[102]

Globle Market Insight, Inc. Stem Cell Therapy Market Size By Type (Allogenic Stem Cell Therapy, Autologous Stem Cell Therapy), By Therapeutic Area (Oncology, Orthopedic, Cardiovascular, Neurology), End-use (Hospitals, Clinics), COVID-19 Impact Analysis, Regional Outlook, Technology Potential, Competitive Market Share & Forecast, 2021-2027. 2021.

[103]

MarketsandMarkets, Inc. Stem cell therapy market by type (allogeneic, autologous), cell source (adipose tissue-derived msc, bonemarrow, placenta/umbilical cord), therapeutic application (musculoskeletal, wounds, surgeries, cardiovascular, neurological)—global forecast to 2027. 2022.

[104]

Malchenko S, Xie J, de Fatima, Bonaldo M, et al. Onset of rosette formation during spontaneous neural differentiation of hESC and hiPSC colonies. Gene. 2014;534(2):400-407.

[105]

Hayashi Y, Ohnuma K, Furue MK. Pluripotent stem cell heterogeneity. Adv Exp Med Biol. 2019;1123:71-94.

[106]

Zhou T, Yuan Z, Weng J, et al. Challenges and advances in clinical applications of mesenchymal stromal cells. J Hematol Oncol. 2021;14(1):24.

[107]

Galipeau J, Sensébé L. Mesenchymal stromal cells: clinical challenges and therapeutic opportunities. Cell Stem Cell. 2018;22(6):824-833.

[108]

ISO/TC 276. ISO 24651:2022 Biotechnology—Biobanking—Requirements for human mesenchymal stromal cells derived from bone marrow.

[109]

ISO/TC 276. ISO 24603:2022 Biotechnology—Biobanking—Requirements for human and mouse pluripotent stem cells.

[110]

ISO/TC 276. ISO/TS 22859:2022 Biotechnology—Biobanking—Requirements for human mesenchymal stromal cells derived from umbilical cord tissue.

[111]

ISO/TC 276. ISO 21973:2020 Biotechnology—General requirements for transportation of cells for therapeutic use.

[112]

ISO/TC 276. ISO 20391-1:2018 Biotechnology—Cell counting—Part 1: General guidance on cell counting methods.

[113]

ISO/TC 276. ISO 20391-2:2019 Biotechnology—Cell counting—Part 2: Experimental design and statistical analysis to quantify counting method performance.

[114]

ISO/TC 276. ISO 23033:2021 Biotechnology—Analytical methods—General requirements and considerations for the testing and characterization of cellular therapeutic products.

[115]

ISO/TC 276. ISO/TS 23565:2021 Biotechnology—Bioprocessing—General requirements and considerations for equipment systems used in the manufacturing of cells for therapeutic use.

[116]

ISO/TC 276. ISO 20399:2022 Biotechnology—Ancillary materials present during the production of cellular therapeutic products and gene therapy products.

[117]

ISO/TC 276. ISO/TR 22758:2020 Biotechnology—Biobanking—Implementation guide for ISO 20387.

[118]

ISO/TC 276. ISO 21899:2020 Biotechnology—Biobanking—General requirements for the validation and verification of processing methods for biological material in biobanks.

[119]

ISO/TC 276. ISO 20387:2018 Biotechnology—Biobanking—General requirements for biobanking.

[120]

ISO/TC 276. ISO/WD 18162 Biotechnology—Biobanking—Requirements for human neural stem cells derived from pluripotent stem cells.

[121]

ISO/TC 276. ISO/CD 24479 Biotechnology—Minimum requirements for cellular morphological analysis—Image capture, image processing, and morphometry.

[122]

ISO/TC 276. ISO/FDIS 20404 Biotechnology—Bioprocessing—General requirements for the design of packaging to contain cells for therapeutic use.

[123]

ISO/TC 276. ISO/AWI 8934 Biotechnology—General considerations and requirements for cell viability analytical methods—Part 1: Mammalian cells.

[124]

ISO/TC 276. ISO/CD 8472-1 Biotechnology—Data interoperability for stem cell data—Part 1: Framework.

[125]

Association for the Advancement of Blood and Biotherapies. Standards for Cellular Therapy Services, 10th ed. 2021.

[126]

Foundation for the Accreditation of Cellular Therapy. <Eighth Edition FACT-JACIE International Standards for Hematopoietic Cellular Therapy Product Collection, Processing and Administration, 8.1-UpdatedLinks (533_1).pdf>. 2021.

[127]

Foundation for the Accreditation of Cellular Therapy. <FACT-JACIE International Standards for Hematopoietic Cellular Therapy Product Collection, Processing, and Administration, Seventh Edition_ Effective May 30, 2018 (236_1).pdf>. 2018.

[128]

Foundation for the Accreditation of Cellular Therapy. <NetCord-FACT International Standards for Cord Blood Collection, Banking, and Release for Administration, Seventh Edition_ Effective January 15, 2020 (229_3).pdf>. 2019.

[129]

Foundation for the Accreditation of Cellular Therapy. Second Edition FACT Common Standards for Cellular Therapies. 2019.

[130]

Dominici M, Le Blanc K, Mueller I, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8(4):315-317.

[131]

Krampera M, Galipeau J, Shi Y, Tarte K, Sensebe L, Therapy MSCCotISfC. Immunological characterization of multipotent mesenchymal stromal cells—The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy. 2013;15(9):1054-1061.

[132]

Guidelines for the conduct of human embryonic stem cell research. Curr Prot Stem Cell Biol. 2007;Appendix 1:Appendix 1A.

[133]

Hyun I, Lindvall O, Ahrlund-Richter L, et al. New ISSCR guidelines underscore major principles for responsible translational stem cell research. Cell Stem Cell. 2008;3(6):607-609.

[134]

Daley GQ, Hyun I, Apperley JF, et al. setting global standards for stem cell research and clinical translation: the 2016 ISSCR guidelines. Stem Cell Rep. 2016;6(6):787-797.

[135]

International Society for Stem Cell Research. <isscr-guidelines-for-stem-cell-research-and-clinical-translation-2021.pdf>. 2021.

[136]

European Directorate for the Quality of Medicines and HealthCare. <Contents of the 11th Edition.pdf>. 2021.

[137]

European Directorate for the Quality of Medicines and HealthCare. <Contents of the 10th Edition.pdf>. 2019.

[138]

European Directorate for the Quality of Medicines and HealthCare. Guide to the Quality and Safety of Tissues and Cells for Human Application, 5th ed. 2022.

[139]

International Council for Harmonisation. <M10_Guideline_Step4_2022_0524.pdf>. 2022.

[140]

International Council for Harmonisation. <S6_R1_Guideline_0.pdf>. 2011.

[141]

International Council for Harmonisation. <Q5A(R1) Guideline_0.pdf>. 1999.

[142]

International Council for Harmonisation. <Q5D Guideline.pdf>. 1997.

[143]

British Standards Institution. PAS 157 Evaluation of materials of biological origin used in the production of cell-based medicinal products—guide. 2015.

[144]

British Standards Institution. PAS 84:2012 Cell therapy and regenerative medicine—glossary. 2012.

[145]

British Standards Institution. PAS 83:2012 Developing human cells for clinical applications in the European Union and the United States of America—guide. 2012.

[146]

British Standards Institution. PAS 93: 2011 Characterization of human cells for clinical applications—guide. 2011.

[147]

German Institute for Standardization. DIN 13279 Biotechnology—requirements for sample containers for storing biological materials in biobanks. 2022.

[148]

Association Francaise de Normalisation. NF X42-000 Biotechnology—Vocabulary—General Terms. 1986.

[149]

Association Francaise de Normalisation. NF U47-200 Animal health analysis methods—Good practice guide for cell cultures. 2016.

[150]

National Cell Manufacturing Consortium. <Cell-Manufacturing-Roadmap-to-2030_ForWeb_110819.pdf>. 2019.

[151]

National Cell Manufacturing Consortium. <NCMC-Roadmap-Update_07-28-2017.pdf>. 2017.

[152]

National Cell Manufacturing Consortium. <NCMC_Roadmap_021816_high_res-2.pdf>. 2016.

[153]

American Society for Testing and Materials. F3106-22 standard guide for in vitro osteoblast differentiation assays. 2022.

[154]

American Society for Testing and Materials. F3088-22 standard practice for use of a centrifugation method to quantify/study cell-material adhesive interactions. 2022.

[155]

American Society for Testing and Materials. F2998-14 guide for using fluorescence microscopy to quantify the spread area of fixed cells. 2021.

[156]

American Society for Testing and Materials. F2997-21 standard practice for quantification of calcium deposits in osteogenic culture of progenitor cells using fluorescent image analysis. 2021.

[157]

American Society for Testing and Materials. F2131-21 standard test method for in vitro biological activity of recombinant human bone morphogenetic protein-2 (rhBMP-2) Using the W-20 mouse stromal cell line. 2021.

[158]

American Society for Testing and Materials. F2944-20 standard practice for automated colony forming unit (cfu) assays—image acquisition and analysis method for enumerating and characterizing cells and colonies in culture. 2020.

[159]

American Society for Testing and Materials. F3368-19 standard guide for cell potency assays for cell therapy and tissue engineered products. 2019.

[160]

American Society for Testing and Materials. F3294-18 standard guide for performing quantitative fluorescence intensity measurements in cell-based assays with widefield epifluorescence microscopy. 2018.

[161]

American Society for Testing and Materials. F3206-17 standard guide for assessing medical device cytocompatibility with delivered cellular therapies. 2017.

[162]

Clinical and Laboratory Standards Institute. H42-A2 Enumeration of Immunologically Defined Cell Populations by Flow Cytometry; Approved Guideline-Second Edition. 2007.

[163]

Clinical and Laboratory Standards Institute. I/LA29-A Detection of HLA-Specific Alloantibody by Flow Cytometry and Solid Phase Assays; Approved Guideline. 2008.

[164]

Clinical and Laboratory Standards Institute. I/LA26-A2 Performance of Single Cell Immune Response Assays; Approved Guideline—Second Edition. 2013.

[165]

Parenteral Drug Association. TR 81-2019 Cell-Based Therapy Control Strategy. 2019.

[166]

Japanese Industrial Standards Committee. K3600 biotechnology−vocabulary. 2020.

[167]

Japanese Industrial Standards Committee. K3603 plastic vials for frozen storage and ultra low-temperature preservation. 2021.

[168]

Korean Agency for Technology and Standards. KS P 1600 General guideline of safety test for the cell based therapeutic substances. 2018.

[169]

Standardization Administration of China. GB/T 40365-2021 General guide for cell sterility testing. 2021.

[170]

Standardization Administration of China. GB/T 40172-2021 General guidance on detection methods of mammalian cell cross-contamination. 2021.

[171]

Standardization Administration of China. GB/T 39730-2020 General requirements for cell counting—Flow cytometry. 2020.

[172]

Standardization Administration of China. GB/T 39729-2020 General requirements for measurement of cell purity—Flow cytometry. 2020.

[173]

Standardization Administration of China. GB/T 38788-2020 Technical specification for establishment of porcine pluripotent stem cells. 2020.

[174]

Standardization Administration of China. GB/T 35520-2017 chemicals—embryotoxicity—embryonic stem cell test. 2017.

RIGHTS & PERMISSIONS

2024 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

143

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/