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

Developing assays to address identity, potency, purity and safety: cell characterization in cell therapy process development

    ,
    Scott R Burger

    Advanced Cell & Gene Therapy, 105 Highgrove Drive, Chapel Hill, NC 27516, USA

    ,
    Michael McCaman

    Therapeutic Cell Solutions Research & Development, Lonza Bioscience, 8830 Biggs Ford Road, Walkersville, MD 21793, USA

    &
    Jon A Rowley

    Therapeutic Cell Solutions Research & Development, Lonza Bioscience, 8830 Biggs Ford Road, Walkersville, MD 21793, USA

    Published Online:https://doi.org/10.2217/rme.11.105

    A major challenge to commercializing cell-based therapies is developing scalable manufacturing processes while maintaining the critical quality parameters (identity, potency, purity, safety) of the final live cell product. Process development activities such as extended passaging and serum reduction/elimination can facilitate the streamlining of cell manufacturing process as long as the biological functions of the product remain intact. Best practices in process development will be dependent on cell characterization; a thorough understanding of the cell-based product. Unique biological properties associated with different types of cell-based products are discussed. Cell characterization may be used as a tool for successful process development activities, which can promote a candidate cell therapy product through clinical development and ultimately to a commercialized product.

    Papers of special note have been highlighted as: ▪ of interest ▪▪ of considerable interest

    References

    • Mason C, Dunnill P. Translational regenerative medicine research: essential to discovery and outcome. Regen. Med.2(3),227–229 (2007).
    • James D. Therapies of tomorrow require more than factories from the past. Bioproc. Int.9,4–11 (2011).
    • Mason C, Hoare M. Regenerative medicine bioprocessing: the need to learn from the experience of other fields. Regen. Med.1(5),615–623 (2006).
    • Bravery C. Regulating interface science healthcare products: myths and uncertainties. J. R. Soc. Interface7,7 (2010).
    • Brandenberger R, Burger S, Campbell A, Fong T, Lapinskas E, Rowley J. Cell therapy bioprocessing integrating process and product development for the next generation of biotherapeutics. Bioproc. Int.9(Suppl. 1),30–37 (2011).
    • Regenerative medicine glossary. Regen. Med.4(Suppl. 4),S1–S88 (2009).▪ The regenerative medicine glossary defines key ideas and concepts associated with the fields of regenerative medicine and/or cell therapy.
    • Rayment EA, Williams DJ. Concise review. Mind the gap: challenges in characterizing and quantifying cell- and tissue-based therapies for clinical translation. Stem Cells28(5),996–1004 (2010).▪▪ Safety is the most important critical quality attribute of a product; this reference addresses safety concerns associated with cell therapy products, more completely than was possible in this manuscript.
    • US FDA. Guidance for FDA Reviewers and Sponsors. Content and Review of Chemistry, Manufacturing, and Control (CMC) Information for Human Somatic Cell Therapy Investigational New Drug Applications (INDs). US Department of Health and Human Services, Rockville, MD, USA, 1,1–39 (2008).▪▪ The CMC guidance for human somatic cell therapy, issued by the US FDA, contains much of the information cell therapy product developers will need in order to develop a clinical-grade cell therapy product.
    • FDA summary for basis of approval: BLA Ref No. 96–0372, Carticel.
    • 10  Mcintyre C, Flyg B, Fong T. Fluorescence-activated cell sorting for CGMP processing of therapeutic cells. Bioproc. Int.8,44–53 (2010).
    • 11  Suni MA, Dunn HS, Orr PL et al. Performance of plate-based cytokine flow cytometry with automated data analysis. BMC Immunol.4,9 (2003).
    • 12  Stroncek DF, Jin P, Wang E, Ren J, Sabatino M, Marincola FM. Global transcriptional analysis for biomarker discovery and validation in cellular therapies. Mol. Diagn. Ther.13(3),181–193 (2009).
    • 13  Kim KS, Kim JS, Lee MR, Jeong HS, Kim J. A study of microRNAs in silico and in vivo: emerging regulators of embryonic stem cells. FEBS J.276(8),2140–2149 (2009).
    • 14  D’amour KA, Agulnick AD, Eliazer S, Kelly OG, Kroon E, Baetge EE. Efficient differentiation of human embryonic stem cells to definitive endoderm. Nat. Biotechnol.23(12),1534–1541 (2005).
    • 15  Caplan AI. Why are MSCs therapeutic? New data: new insight. J. Pathol.217(2),318–324 (2009).
    • 16  Wang M, Crisostomo PR, Herring C, Meldrum KK, Meldrum DR. Human progenitor cells from bone marrow or adipose tissue produce VEGF, HGF, and IGF-I in response to TNF by a p38 MAPK-dependent mechanism. Am. J. Physiol. Regul. Integr. Comp. Physiol.291(4),R880–R884 (2006).
    • 17  US FDA. Final Guidance for Industry. Potency Tests for Cellular and Gene Therapy Products. US Department of Health and Human Services, Rockville, MD, USA, 1,1–19 (2011).▪▪ The FDA guidance for potency tests is a great reference for product developers struggling to determine how to develop a test to measure the biological activity of their product.
    • 18  Liu CH, Hwang SM. Cytokine interactions in mesenchymal stem cells from cord blood. Cytokine32(6),270–279 (2005).
    • 19  Yagi H, Soto-Gutierrez A, Parekkadan B et al. Mesenchymal stem cells: mechanisms of immunomodulation and homing. Cell Transplant.19(6),667–679 (2010).
    • 20  Curti A, Trabanelli S, Salvestrini V, Baccarani M, Lemoli RM. The role of indoleamine 2,3-dioxygenase in the induction of immune tolerance: focus on hematology. Blood113(11),2394–2401 (2009).
    • 21  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. Cytotherapy8(4),315–317 (2006).
    • 22  Debenedette MA, Calderhead DM, Tcherepanova IY, Nicolette CA, Healey DG. Potency of mature CD40L RNA electroporated dendritic cells correlates with IL-12 secretion by tracking multifunctional CD8+/CD28+ cytotoxic T cell responses in vitro. J. Immunother.34(1),45–57 (2011).
    • 23  Kasten P, Beyen I, Egermann M et al. Instant stem cell therapy: characterization and concentration of human mesenchymal stem cells in vitro. Eur. Cell. Mater.16,47–55 (2008).
    • 24  Yang H, Acker JP, Cabuhat M, Letcher B, Larratt L, Mcgann LE. Association of post-thaw viable CD34+ cells and CFU-GM with time to hematopoietic engraftment. Bone Marrow Transplant.35(9),881–887 (2005).
    • 25  Goltry K, Rowley J, Peters M, Burchardt E. Tissue repair cells for the treatment of cardiovascular diseases. Adv. Mol. Med.3(1),9 (2007).
    • 26  Rich IN, Hall KM. Validation and development of a predictive paradigm for hemotoxicology using a multifunctional bioluminescence colony forming proliferation assay. Toxicol. Sci.87(2),427–441 (2005).
    • 27  Zhang YW, Denham J, Thies RS. Oligodendrocyte progenitor cells derived from human embryonic stem cells express neurotrophic factors. Stem Cells Dev.15(6),943–952 (2006).
    • 28  Kroon E, Martinson LA, Kadoya K et al. Pancreatic endoderm derived from human embryonic stem cells generates glucose-responsive insulin-secreting cells in vivo. Nat. Biotechnol.26(4),443–452 (2008).
    • 29  Klimanskaya I, Hipp J, Rezai KA, West M, Atala A, Lanza R. Derivation and comparative assessment of retinal pigment epithelium from human embryonic stem cells using transcriptomics. Cloning Stem Cells6(3),217–245 (2004).
    • 30  Ellington AA, Kullo IJ, Bailey KR, Klee GG. Antibody-based protein multiplex platforms: technical and operational challenges. Clin. Chem.56(2),186–193 (2009).
    • 31  Goldring CE, Duffy PA, Benvenisty N et al. Assessing the safety of stem cell therapeutics. Cell Stem Cell8(6),618–628 (2011).
    • 32  Nelson PN, Carnegie PR, Martin J et al. Demystified. Human endogenous retroviruses. Mol. Pathol.56(1),11–18 (2003).
    • 33  Ryan FP. Human endogenous retroviruses in health and disease: a symbiotic perspective. J. R. Soc. Med.97(12),560–565 (2004).
    • 34  International Conference on Harmonisation. Q5A Viral Safety Evaluation of Biotechnology Products Derived From Cell Lines of Human or Animal Origin. EMEA, Canary Wharf, London, UK, 1,1–33 (2006).
    • 35  Morgan RA, Dudley ME, Rosenberg SA. Adoptive cell therapy: genetic modification to redirect effector cell specificity. Cancer J.16(4),336–341 (2010).
    • 36  US FDA. Guidance for Industry. Guidance for Human Somatic Cell Therapy and Gene Therapy. US Department of Health and Human Services, Rockville, MD, USA, 1,30 (1998).
    • 37  Higano CS, Small EJ, Schellhammer P et al. Sipuleucel-T. Nature Rev. Drug Discov.9(7),513–514 (2010).
    • 38  Carpenter MK, Frey-Vasconcells J, Rao MS. Developing safe therapies from human pluripotent stem cells. Nat. Biotechnol.27(7),606–613 (2009).
    • 39  Strauss S. Geron trial resumes, but standards for stem cell trials remain elusive. Nat. Biotechnol.28(10),989–990 (2010).
    • 40  Sun X, Long X, Yin Y et al. Similar biological characteristics of human embryonic stem cell lines with normal and abnormal karyotypes. Hum. Reprod.23(10),2185–2193 (2008).
    • 41  Gore A, Li Z, Fung HL et al. Somatic coding mutations in human induced pluripotent stem cells. Nature471(7336),63–67 (2011).
    • 42  US FDA. Guidance for Industry. Process Validation: General Principles and Practices. US Department of Health and Human Services, Rockville, MD, USA, 1,1–22 (2011).
    • 43  International Conference on Harmonisation. Q5E. Comparability of Biotechnological/Biological Products Subject to Changes in Their Manufacturing Process. EMEA, Canary Wharf, London, UK, 1,1–11 (2003).
    • 44  International Conference on Harmonisation. Q5C. Guideline for Industry: Quality of Biotechnological Products: Stability Testing of Biotechnological/Biological Products. EMEA, Canary Wharf, London, UK, 1,1–12 (1996).
    • 45  Thomas RJ, Hourd PC, Williams DJ. Application of process quality engineering techniques to improve the understanding of the in vitro processing of stem cells for therapeutic use. J. Biotechnol.136(3–4),148–155 (2008).
    • 46  Julien C, Whitford W. The biopharmaceutical industry’s new operating paradigm. Bioproc. Int.8,6–14 (2008).
    • 47  Rowley J. Developing cell biomanufacturing processes. Chem. Eng. Prog.SBE(Suppl.),6 (2010).
    • 48  Fensten R. Understanding Animal Sera: Considerations for Use in the Production of Biological Therapeutics. John Wiley and Sons Ltd, West Sussex, UK, 45–58 (2007).
    • 49  Dahl JA, Duggal S, Coulston N et al. Genetic and epigenetic instability of human bone marrow mesenchymal stem cells expanded in autologous serum or fetal bovine serum. Int. J. Dev. Biol.52(8),1033–1042 (2008).
    • 50  Shahdadfar A, Fronsdal K, Haug T, Reinholt FP, Brinchmann JE. In vitro expansion of human mesenchymal stem cells: choice of serum is a determinant of cell proliferation, differentiation, gene expression, and transcriptome stability. Stem Cells23(9),1357–1366 (2005).
    • 101  Greenwood J. How do drugs and biologics differ? Biotechnology Industry Organization, Washington, DC, USA (2010). www.bio.org/healthcare/followonbkg/DrugsVBiologics.asp
    • 102  Interactive cell characterization tool (2011). www.advbiols.com/documents/ImportanceofCharacterisation.swf
    • 103  Burger S, Bravery C. ISCT webinar: potency testing. International Society for Cellular Therapy (2011). www.celltherapysociety.org
    • 104  Geron®. www.geron.com
    • 105  ViaCyte®. http://viacyte.com
    • 106  Advanced Cell Technology®. http://advancedcell.com
    • 107  US FDA. 21 CFR 610.15. Code of Federal Regulations, general biological products standards, constituent materials (2002). www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=610.15
    • 108  US FDA. 21 CFR 610.12. Code of Federal Regulations, general biological products standards, sterility (2010). www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=610.12
    • 109  US FDA. 21 CFR 600; 610; 680 (Docket No FDA-2011-N-0080). Code of Federal Regulations, biological products: general; general biological products standards; additional standards for miscellaneous products, amendments to sterility test requirements for biological products. www.accessdata.fda.gov/scripts/oc/ohrms/dailylist.cfm?yr=2011&mn=6&dy=21
    • 110  Geron. About GRNOPC1. Preclinical safety studies: animal toxicology testing of GRNOPC1. www.geron.com/GRNOPC1Trial/grnopc1-sec3.html
    • 111  Clinical Trials registry and database. www.clinicaltrial.gov/