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

Experimental and computational approach to establish fit-for-purpose cell viability assays

    Laura Pierce

    Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD 20899, USA

    ,
    Hidayah Anderson

    Division of Cellular & Gene Therapies, CBER, FDA, Silver Spring, MD 20993, USA

    ,
    Swarnavo Sarkar

    Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD 20899, USA

    ,
    Steven R Bauer

    *Authors for correspondence:

    E-mail Address: sumona.sarkar@nist.gov

    and

    E-mail Address: srbauer@wakehealth.edu

    Division of Cellular & Gene Therapies, CBER, FDA, Silver Spring, MD 20993, USA

    &
    Sumona Sarkar

    *Authors for correspondence:

    E-mail Address: sumona.sarkar@nist.gov

    and

    E-mail Address: srbauer@wakehealth.edu

    Biosystems & Biomaterials Division, National Institute of Standards & Technology, Gaithersburg, MD 20899, USA

    Published Online:https://doi.org/10.2217/rme-2023-0154

    Aim: Cell viability assays are critical for cell-based products. Here, we demonstrate a combined experimental and computational approach to identify fit-for-purpose cell assays that can predict changes in cell proliferation, a critical biological response in cell expansion. Materials & methods: Jurkat cells were systematically injured using heat (45 ± 1°C). Cell viability was measured at 0 h and 24 h after treatment using assays for membrane integrity, metabolic function and apoptosis. Proliferation kinetics for longer term cultures were modeled using the Gompertz distribution to establish predictive models between cell viability results and proliferation. Results & conclusion: We demonstrate an approach for ranking these assays as predictors of cell proliferation and for setting cell viability specifications when a particular proliferation response is required.

    Plain language summary

    In recent years, there has been a surge in the amount of cellular therapy products which have been engineered to treat patients with severe diseases. These cellular products use living cells to treat the disease, and the quality of these cell products is critical for ensuring product safety and effectiveness. Throughout the process of engineering and manufacturing these cell products, many cells can die or be in the process of dying, and the amount of dead cells in the product can impact product yield and quality. In any given cell product at any given time during the manufacturing process, cells are exposed to stresses, and these stresses can injure the cells through several mechanisms, leading to a range of cell death events that can follow different timelines. There are many existing assays which evaluate the health of the cells, known as cell viability assays, and these assays can be based on many different cell features that indicate a cell has been injured (i.e., cell membrane permeability, changes in cell metabolism, molecular markers for cell death). These cell viability assays provide different insights into the state of cell health/injury based on what cell features are being evaluated and the timing at which the viability measurements are taken, and some viability assays may be more appropriate than others for specific applications. Therefore, a method is needed to appropriately select cell viability assays that are designed to evaluate injuries to cells that occur in specific bioprocess. In this series of studies, we used a range of analytical methods to study the number of living and dead cells in a series of cell populations that we treated to induce damage to the cells, reducing their ability to grow. We then used mathematical models to determine the relationship between cell viability measurements and cell growth over time, and used the results to determine the sensitivity of the viability assays to changes in cell growth. We used a specific cell line in this example, but this technique can be applied to any cell line or cell sample population and different types of injuries can be applied to the cells. This approach can be used by manufacturers of cell-based products and therapies to identify cell viability assays that are meaningful for monitoring the production of cells and characterizing product quality.

    Graphical abstract

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

    References

    • 1. Mueller H, Kassack MU, Wiese M. Comparison of the usefulness of the MTT, ATP, and calcein assays to predict the potency of cytotoxic agents in various human cancer cell lines. J. Biomol. Screen 9(6), 506–515 (2004).
    • 2. Petty RD, Sutherland LA, Hunter EM, Cree IA. Comparison of Mtt and Atp-Based Assays for the Measurement of Viable Cell Number. J. Biolum. Chemilum. 10(1), 29–34 (1995).
    • 3. Weisenthal LM, Dill PL, Kurnick NB, Lippman ME. Comparison of Dye Exclusion Assays with a Clonogenic-Assay in the Determination of Drug-Induced Cyto-Toxicity. Cancer Res. 43(1), 258–264 (1983).
    • 4. Adan A, Kiraz Y, Baran Y. Cell Proliferation and Cytotoxicity Assays. Curr. Pharm. Biotechnol. 17(14), 1213–1221 (2016).
    • 5. International Organization for Standardization. ISO 20391-1:2018 Biotechnology – Cell counting – Part 1: general guidance on cell counting methods. ISO/TC 276 Biotechnology/WG3 5–9 (2018).
    • 6. Lin-Gibson S, Sarkar S, Ito Y. Defining quality attributes to enable measurement assurance for cell therapy products. Cytotherapy 18(10), 1241–1244 (2016). •• Proposes a generalized framework for establishing critical quality attributes of cell therapy products. It highlights the need to establish well-designed, fit-for-purpose measurements as well as the use of appropriate statistical methods.
    • 7. Bauer SR. Stem Cell-based Products in Medicine: FDA Regulatory Considerations. In: Handbook of Stem Cells. Lanza R (Ed.). Elsevier, MA, USA, 805–814 (2004).
    • 8. Hoogendoorn KH, Crommelin DJA, Jiskoot W. Formulation of Cell-Based Medicinal Products: A Question of Life or Death? J. Pharm. Sci. 110(5), 1885–1894 (2021).
    • 9. Chan LL-Y, Kuksin D, Laverty DJ, Saldi S, Qiu J. Morphological observation and analysis using automated image cytometry for the comparison of trypan blue and fluorescence-based viability detection method. Cytotech. 461–473 doi: 10.1007/s10616-014-9704-5(67) (2015). • Compares two commonly used cell viability methods and demonstrates the impact of exposure to the chemicals on the resulting measurement. It emphasizes the concept of identifying an appropriate assay type which will reflect the sample's properties without properties of the dye confounding the measurement.
    • 10. Cadena-Herrera D, Esparza-De Lara JE, Ramírez-Ibañez ND et al. Validation of three viable-cell counting methods: manual, semi-automated, and automated. Biotechnol. Rep. 7, 9–16 (2015).
    • 11. Cummings BS, Schnellmann RG. Measurement of Cell Death in Mammalian Cells. Curr. Protocols Pharmacol. 25(1), 1–30 (2004).
    • 12. Mascotti K, Mccullough J, Burger SR. HPC viability measurement: trypan blue versus acridine orange and propidium iodide. Transfusion 40, 693–696 (2000).
    • 13. Chong EA, Levine BL, Grupp SA et al. CAR T cell viability release testing and clinical outcomes: is there a lower limit? Blood 134(21), 1873–1875 (2019). •• Evaluates efficacy of the cell therapy product tisagenlecleucel, comparing viability measurements to patient outcome and finding no correlation between those administered product >80% viability and those administered product of lower than 80% viability. This evidence was used to argue for a lower (70%) viability release criterion and also calls for a closer study of release criteria for CAR-T products, driven by patient response.
    • 14. Pierce L, Sarkar S, Chan LL-Y, Lin B, Qiu J. Outcomes from a cell viability workshop: fit-for-purpose considerations for cell viability measurements for cellular therapeutic products. Cell and Gene Therapy Insights 7, 551–569 (2021). •• This review of a cell viability workshop reports the results of a survey of cell therapy industry needs for viability measurements and highlights the concept of fit-for-purpose measurement needs. It demonstrates the concept of identifying and selecting a fit-for-purpose method through evaluating measurement method considerations, biological considerations, and sample considerations.
    • 15. Milleron RS, Bratton SB. Heat Shock Induces Apoptosis Independently of Any Known Initiator Caspase-activating Complex. J. Biol. Chem. 281(25), 16991–17000 (2006).
    • 16. Velichko AK, Markova EN, Petrova NV, Razin SV, Kantidze OL. Mechanisms of heat shock response in mammals. Cell. Mol. Life Sci. 70(22), 4229–4241 (2013).
    • 17. Richter K, Haslbeck M, Buchner J. The Heat Shock Response: life on the Verge of Death. Molecular Cell 40(2), 253–266 (2010).
    • 18. Tran SEF, Meinander A, Holmström TH et al. Heat stress downregulates FLIP and sensitizes cells to Fas receptor-mediated apoptosis. Cell Death & Differentiation 10(10), 1137–1147 (2003).
    • 19. International Organization for Standardization. ISO 20391-2 2019: Biotechnology – Cell counting – Part 2: Experimental design and statistical analysis to quantify counting method performance. ISO/TC 276 Biotechnology/WG3 8–22 (2019).
    • 20. Vaghi C, Rodallec A, Fanciullino R et al. Population modeling of tumor growth curves and the reduced Gompertz model improve prediction of the age of experimental tumors. PLOS Computational Biology 16(2), e1007178 (2020). • Explains the modeling of proliferation kinetics using the Gompertz model and reduced Gompertz model and explains the key differences in parameters for each. It explains how the Gompertz model provides a superior fit to populations data as compared to the logistic and exponential models.
    • 21. Winsor CP. The Gompertz curve as a growth curve. Proc. Natl Acad. Sci. USA 18, 1–8 (1932).
    • 22. Chatterjee T, Chatterjee BK, Majumdar D, Chakrabarti P. Antibacterial effect of silver nanoparticles and the modeling of bacterial growth kinetics using a modified Gompertz model. Bba-Gen Subjects 1850(2), 299–306 (2015).
    • 23. Horowitz J, Normand MD, Corradini MG, Peleg M. Probabilistic Model of Microbial Cell Growth, Division, and Mortality. Appl. Environ. Microbiol. 76(1), 230–242 (2010).
    • 24. Van Engeland M, Nieland LJ, Ramaekers FC, Schutte B, Reutelingsperger CP. Annexin V-affinity assay: a review on an apoptosis detection system based on phosphatidylserine exposure. Cytometry 31(1), 1–9 (1998).
    • 25. Mandrekar J. Receiver operating characteristic curve in diagnostic test assessment. J. Thoracic Oncol. 5(9), 1315–1316 (2010). • Explains the utility of using a receiver operating characteristic curve in biomarker analysis applications.
    • 26. Riss T, Niles A, Moravec R, Karassina N, Vidugiriene J. Cytotoxicity assays: in vitro methods to measure dead cells. In: Assay Guidance Manual [Internet]. MD, USA, 1–16 (2019).
    • 27. Green D, Evan G. A matter of life and death. Cancer Cell 1(1), 19–30 (2002).
    • 28. Guo M, Hay B. Cell proliferation and apoptosis. Curr. Opin. Cell Biol. 11(6), 745–752 (1999).
    • 29. Bersenev A, Kili S. Management of ‘out of specification’ commercial autologous CAR-T cell products. Cell and Gene Therapy Insights 4(11), 1051–1058 (2018). • This paper gives several recent examples of cell therapy products which fell out of specification, highlights the idea that CAR-T products have inherent manufacturing variabilities, and stresses that these variabilities may not necessarily be reflective of product outcome. It calls for the development of an alternate approach to setting CAR-T manufacturing specifications.
    • 30. Özkaya A, Geyik C. From viability to cell death: claims with insufficient evidence in high-impact cell culture studies. PLOS ONE 17(2), e0250754 1–11 (2022).
    • 31. Komatsu H, Qi M, Gonzalez N et al. A Multiparametric Assessment of Human Islets Predicts Transplant Outcomes in Diabetic Mice. Cell Transplant. 30, 9636897211052291 (2021). • This paper gives an example of the value of using multiparameter approaches to determining a biological functional outcome from bioassay measurements.
    • 32. Altman N, Krzywinski M. Predicting with confidence and tolerance. Nat. Methods 15(11), 843–845 (2018).