3,185
Views
2
CrossRef citations to date
0
Altmetric
Research Paper

The importance of cell culture parameter standardization: an assessment of the robustness of the 2102Ep reference cell line

, ORCID Icon, , , ORCID Icon & ORCID Icon
Pages 341-357 | Received 09 Nov 2020, Accepted 24 Dec 2020, Published online: 11 Jan 2021

References

  • Kirkeby A, Nolbrant S, Tiklova K, et al. Predictive markers guide differentiation to improve graft outcome in clinical translation of hESC-based therapy for parkinson’s disease. Cell Stem Cell. 2017;20(1):135–148.
  • Sundberg M, Bogetofte H, Lawson T, et al. Improved cell therapy protocols for Parkinson’s disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells. 2013;31(8):1548–1562.
  • Grealish S, Diguet E, Kirkeby A, 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. 2014;15(5):653–665.
  • French A, Bravery C, Smith J, et al. Enabling consistency in pluripotent stem Cell-Derived products for research and development and clinical applications through material standards. Stem Cells Transl Med. 2015;4(3):217–223.
  • Carmen J, Burger SR, McCaman M. Developing assays to address identity, potency, purity and safety: cell characterization in cell therapy process development. Regen Med. 2012;7(1):85–100.
  • Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv. 2013;31:1600.
  • Deasy BM, Jankowski RJ, Payne TR, et al. Modeling stem cell population growth: incorporating terms for proliferative heterogeneity. Stem Cells. 2003;21(5):536–545.
  • Salmikangas P, Menezes-Ferreira M, Reischl I, et al. Manufacturing, characterization and control of cell-based medicinal products: challenging paradigms toward commercial use. Regen Med. 2015;10(1):65–78.
  • Lipsitz YY, Timmins NE, Zandstra PW. Quality cell therapy manufacturing by design. Nat Biotechnol. 2016;34(4):393–400.
  • Kusena JW, Thomas RJ, McCall MJ, et al. From protocol to product: ventral midbrain dopaminergic neuron differentiation for the treatment of Parkinson’s disease. Regen Med. 2019;14(11):1057–1069.
  • Vatsan RS, Bross PF, Liu K, et al. Regulation of immunotherapeutic products for cancer and FDA’s role in product development and clinical evaluation. J Immunother Cancer. 2013;1(1):1–5.
  • Bubela T, McCabe C, Archibald P, et al. Bringing regenerative medicines to the clinic: the future for regulation and reimbursement. Regen Med. 2015;10(7):897–911.
  • Rowley J, Abraham E, Campbell A, et al. Meeting lot-size challenges of manufacturing adherent cells for therapy. Bioprocess Int. 2013;10:16–22.
  • Bravery CA, Carmen J, Fong T, et al. Potency assay development for cellular therapy products: an ISCT* review of the requirements and experiences in the industry. Cytotherapy. 2013;15(1):9–19.e9.
  • U.S. Department of health and human services, food and drug administration C for BE and R. Draft guidance for industry: potency tests for cellular and gene therapy products. Biotechnol Law Rep. 2011;27:568–577.
  • Huang J, Kaul G, Cai C, et al. Quality by design case study: an integrated multivariate approach to drug product and process development. Int J Pharm. 2009;382(1–2):23–32.
  • Williams DJ, Archer R, Archibald P, et al. Comparability: manufacturing, characterization and controls, report of a UK regenerative medicine platform pluripotent stem cell platform workshop, Trinity Hall, Cambridge, 14-15 September 2015. Regen Med. 2016;11(5):483–492.
  • Brindley DA, French AL, Baptista R, et al. Cell therapy bioprocessing technologies and indicators of technological convergence. Bioprocess Int. 2014;12:14–21.
  • Salmikangas P. Dossier Requirements for ATMPs: scientific requirements for cell therapy and tissue engineered products - Quality Aspects [Internet]. Workshop on Advanced Therapy Medicinal Products, European Medicines Agency (EMA) London; 2009. //www.ema.europa.eu/en/documents/presentation/quality-requirements-cell-based-medicinal-products-paula-salmikangas_en.pdf
  • European Medicines Agency. Guideline on human cell-based medicinal products. Doc Ref EMEA/CHMP 2008; EMA:EMEA/CHMP/410869/2006.
  • Doi D, Samata B, Katsukawa M, et al. Isolation of human induced pluripotent stem cell-derived dopaminergic progenitors by cell sorting for successful transplantation. Stem Cell Reports. 2014;2(3):337–350.
  • European Medicines Agency. ICH Topic Q5D quality of biotechnological products : derivation and characterisation of cell substrates used for production of biotechnological/biological products. 1997.
  • Jones BS, Lamb LS, Goldman F, et al. Improving the safety of cell therapy products by suicide gene transfer. Front Pharmacol. 2014;5:254.
  • Yagyu S, Hoyos V, Del Bufalo F, et al. An inducible caspase-9 suicide gene to improve the safety of therapy using human induced pluripotent stem cells. Mol Ther. 2015;23:1475–1485.
  • Josephson R, Ording CJ, Liu Y, et al. Qualification of embryonal carcinoma 2102Ep as a reference for human embryonic stem cell research. Stem Cells. 2007;25:437–446.
  • Andrews PW. Human embryonal carcinoma cells in culture do not synthesize fibronectin until they differentiate. Int J Cancer. 1982;30:567–571.
  • Pamies D, Bal-Price A, Simeonov A, et al. Good cell culture practice for stem cells & stem-cell-derived models. Altex. 2017;34(1)95–132.
  • Allegrucci C, Young LE. Differences between human embryonic stem cell lines. Hum Reprod Update. 2007;13(2):103–120.
  • Geraghty RJ, Capes-Davis A, Davis JM, et al. Guidelines for the use of cell lines in biomedical research. Br J Cancer. 2014;111(6):1021–1046.
  • Plaia TW, Josephson R, Liu Y, et al. Characterization of a new NIH-registered variant human embryonic stem cell line, BG01V: a tool for human embryonic stem cell research. Stem Cells. 2006;24(3):531–546.
  • Yin Z, Ker DF, Junkers S, et al. Data-driven prediction of stem cell expansion cultures. In: 2011 Annual International Conference of the IEEE Engineering in Medicine and Biology Society. Boston, Massachusetts, USA:IEEE; 2011. 3577–3580.
  • Ker DFE, Weiss LE, Junkers SN, et al. An engineered approach to stem cell culture: automating the decision process for real-time adaptive subculture of stem cells. PLoS One. 2011;6(11):e27672.
  • Phelan K, May KM. Mammalian cell tissue culture techniques. Curr Protoc Mol Biol. 2017;117(1):A.3F.1-A.3F.23.
  • Freshney RI. Culture of animal cells. Hoboken, NJ, USA: John Wiley & Sons, Inc; 2010.
  • Phelan MC. Techniques for mammalian cell tissue culture. Curr Protoc Hum Genet. 2006;3:A.3F.1–A.3F.18.
  • Phelan K, May KM. Basic techniques in mammalian cell tissue culture. Curr Protoc Cell Biol. 2015;2015:1.1.1–1.1.22.
  • Senger RS, Karim MN. Optimization of fed-batch parameters and harvest time of CHO cell cultures for a glycosylated product with multiple mechanisms of inactivation. Biotechnol Bioeng. 2007;98(2):378–390.
  • Kotidis P, Jedrzejewski P, Sou SN, et al. Model-based optimization of antibody galactosylation in CHO cell culture. Biotechnol Bioeng. 2019;116:1612–1626.
  • ISO International Organization for Standardization. ISO 9001:2015 - Quality management systems — Requirements [Internet]. 2015 [cited 2020 Dec 16]. Available from: https://www.iso.org/standard/62085.html
  • Heathman TRJ, Glyn VAM, Picken A, et al. Expansion, harvest and cryopreservation of human mesenchymal stem cells in a serum-free microcarrier process. Biotechnol Bioeng. 2015;112(8):1696–1707.
  • Schinzel RT, Ahfeldt T, Lau FH, et al. Efficient culturing and genetic manipulation of human pluripotent stem cells. PLoS One. 2011;6(12):e27495.
  • Shariatzadeh M, Chandra A, Wilson SL, et al. Distributed automated manufacturing of pluripotent stem cell products. Int J Adv Manuf Technol. 2020;106(3–4):1085–1103.
  • Reuveny S, Velez D, Macmillan JD, et al. Factors affecting cell growth and monoclonal antibody production in stirred reactors. J Immunol Methods. 1986;86:53–59.
  • Hunsberger JG, Shupe T, Atala A. An industry-driven roadmap for manufacturing in regenerative medicine. Stem Cells Transl Med. 2018;7(8):564–568.
  • Hourd P, Chandra A, Alvey D, et al. Qualification of academic facilities for small-scale automated manufacture of autologous cell-based products. Regen Med. 2014;9(6):799–815.
  • Andrews PW, Goodfellow PN, Shevinsky LH, et al. Cell‐surface antigens of a clonal human embryonal carcinoma cell line: morphological and antigenic differentiation in culture. Int J Cancer. 1982;29(5):523–531.
  • Tewary M, Shakiba N, Zandstra PW. Stem cell bioengineering: building from stem cell biology. Nat Rev Genet. 2018;19(10):595–614.
  • Cooper S. Distinguishing between linear and exponential cell growth during the division cycle: single-cell studies, cell-culture studies, and the object of cell-cycle research. Theor Biol Med Model. 2006;3(1):10.
  • Gage BK, Webber TD, Kieffer TJ. Initial cell seeding density influences pancreatic endocrine development during in vitro differentiation of human embryonic stem cells. PLoS One. 2013;8(12):8.
  • Jacobs K, Zambelli F, Mertzanidou A, et al. Higher-density culture in human embryonic stem cells results in DNA damage and genome instability. Stem Cell Reports. 2016;6(3):330–341.
  • Kempf H, Olmer R, Haase A, et al. Bulk cell density and Wnt/TGFbeta signalling regulate mesendodermal patterning of human pluripotent stem cells. Nat Commun. 2016;7(1):13602.
  • Bulusu V, Aulehla A. Metabolic control of cellular differentiation. Dev Cell. 2016;39:286–287.
  • Martano G, Borroni EM, Lopci E, et al. Metabolism of stem and progenitor cells: proper methods to answer specific questions. Front Mol Neurosci. 2019;12:151.
  • Shyh-Chang N, Ng HH. The metabolic programming of stem cells. Genes Dev. 2017;31(4):336–346.
  • Thomson M, Liu SJ, Zou LN, et al. Pluripotency factors in embryonic stem cells regulate differentiation into germ layers. Cell. 2011;145(6):875–889.
  • Wattanapanitch M, Klincumhom N, Potirat P, et al. Dual small-molecule targeting of SMAD signaling stimulates human induced pluripotent stem cells toward neural lineages. PLoS One. 2014;9(9):e106952.
  • Kirkeby A, Nelander J, Parmar M. Generating regionalized neuronal cells from pluripotency, a step-by-step protocol. Front Cell Neurosci. 2013;6:1–4.
  • Wilson HK, Canfield SG, Hjortness MK, et al. Exploring the effects of cell seeding density on the differentiation of human pluripotent stem cells to brain microvascular endothelial cells. Fluids Barriers CNS. 2015;12(1):1–12.
  • Singh SJ, Turner W, Glaser DE, et al. Metabolic shift in density-dependent stem cell differentiation. Cell Commun Signal. 2017;15(1):44.
  • Horiguchi I, Urabe Y, Kimura K, et al. Effects of glucose, lactate and basic FGF as limiting factors on the expansion of human induced pluripotent stem cells. J Biosci Bioeng. 2018;125:111–115.
  • Palm W, Thompson CB. Nutrient acquisition strategies of mammalian cells. Nature. 2017;546:234–242.
  • O’Brien CM, Mulukutla BC, Mashek DG, et al. Regulation of metabolic homeostasis in cell culture bioprocesses. Trends Biotechnol. 2020;38(10):1113–1127.
  • Wall IB, Davie N. Quality control in cell and tissue engineering. Standardisat Cell Tissue Eng. 2013;148–65.
  • Coecke S, Balls M, Bowe G, et al. Guidance on good cell culture practice. Cell Technol Cell Prod. 2007;page 313–5.
  • Knight LA, Cree IA. Quality assurance and good laboratory practice. Methods Mol Biol. 2011;731:115–124. .
  • NICE. NICE recommends another revolutionary CAR T-cell therapy for adults with lymphoma | News and features | News | NICE [Internet]. NICE 2019; //www.nice.org.uk/news/article/nice-recommends-another-revolutionary-car-t-cell-therapy-for-adults-with-lymphoma
  • Kuehn BM. The promise and challenges of CAR-T gene therapy. J Am Med Assoc. 2017;318(22):2167–2169.