Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Quality cell therapy manufacturing by design

Abstract

Transplantation of live cells as therapeutic agents is poised to offer new treatment options for a wide range of acute and chronic diseases. However, the biological complexity of cells has hampered the translation of laboratory-scale experiments into industrial processes for reliable, cost-effective manufacturing of cell-based therapies. We argue here that a solution to this challenge is to design cell manufacturing processes according to quality-by-design (QbD) principles. QbD integrates scientific knowledge and risk analysis into manufacturing process development and is already being adopted by the biopharmaceutical industry. Many opportunities to incorporate QbD into cell therapy manufacturing exist, although further technology development is required for full implementation. Linking measurable molecular and cellular characteristics of a cell population to final product quality through QbD is a crucial step in realizing the potential for cell therapies to transform healthcare.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Modular bioreactor design facilitates QbD.
Figure 2: The QbD process.
Figure 3: Design space development for CAR-T therapies.

Similar content being viewed by others

References

  1. Kirouac, D.C. & Zandstra, P.W. The systematic production of cells for cell therapies. Cell Stem Cell 3, 369–381 (2008).

    Article  CAS  Google Scholar 

  2. US Food and Drug Administration. Guidance for Industry: Q8(R2) pharmaceutical development. http://www.fda.gov/downloads/Drugs/.../Guidances/ucm073507.pdf (International Conference on Harmonization, 2009).

  3. Rathore, A.S. & Winkle, H. Quality by design for biopharmaceuticals. Nat. Biotechnol. 27, 26–34 (2009).

    Article  CAS  Google Scholar 

  4. Alliance for Regenerative Medicine. Annual Data Report on Gene and Cellular Therapies and the Regenerative Medicine Sector. http://alliancerm.org/sites/default/files/ARM_Annual_Report_2015_Web_Version_FINAL.pdf (2015).

  5. Selen, A. et al. Meeting report: applied biopharmaceutics and quality by design for dissolution/release specification setting: product quality for patient benefit. AAPS J. 12, 465–472 (2010).

    Article  CAS  Google Scholar 

  6. Lund, T.C., Boitano, A.E., Delaney, C.S., Shpall, E.J. & Wagner, J.E. Advances in umbilical cord blood manipulation-from niche to bedside. Nat. Rev. Clin. Oncol. 12, 163–174 (2015).

    Article  Google Scholar 

  7. Dominici, M. et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8, 315–317 (2006).

    Article  CAS  Google Scholar 

  8. Krampera, M., Galipeau, J., Shi, Y., Tarte, K. & Sensebe, L. ; MSC Committee of the International Society for Cellular Therapy (ISCT). Immunological characterization of multipotent mesenchymal stromal cells—The International Society for Cellular Therapy (ISCT) working proposal. Cytotherapy 15, 1054–1061 (2013).

    Article  Google Scholar 

  9. Hollyman, D. et al. Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J. Immunother. 32, 169–180 (2009).

    Article  CAS  Google Scholar 

  10. Bravery, C.A. et al. Potency assay development for cellular therapy products: an ISCT review of the requirements and experiences in the industry. Cytotherapy 15, 9–19 (2013).

    Article  Google Scholar 

  11. Xu, C. et al. Efficient generation and cryopreservation of cardiomyocytes derived from human embryonic stem cells. Regen. Med. 6, 53–66 (2011).

    Article  CAS  Google Scholar 

  12. Leobon, B. et al. Myoblasts transplanted into rat infarcted myocardium are functionally isolated from their host. Proc. Natl. Acad. Sci. USA 100, 7808–7811 (2003).

    Article  CAS  Google Scholar 

  13. Hentze, H. et al. Teratoma formation by human embryonic stem cells: evaluation of essential parameters for future safety studies. Stem Cell Res. 2, 198–210 (2009).

    Article  Google Scholar 

  14. Tohyama, S. et al. Distinct metabolic flow enables large-scale purification of mouse and human pluripotent stem cell-derived cardiomyocytes. Cell Stem Cell 12, 127–137 (2012).

    Article  Google Scholar 

  15. Ben-David, U. et al. Selective elimination of human pluripotent stem cells by an oleate synthesis inhibitor discovered in a high-throughput screen. Cell Stem Cell 12, 167–179 (2013).

    Article  CAS  Google Scholar 

  16. Sarugaser, R., Hanoun, L., Keating, A., Stanford, W.L. & Davies, J.E. Human mesenchymal stem cells self-renew and differentiate according to a deterministic hierarchy. PLoS One 4, e6498 (2009).

    Article  Google Scholar 

  17. Clarke, D. et al. Managing particulates in cellular therapy. Cytotherapy 14, 1032–1040 (2012).

    Article  CAS  Google Scholar 

  18. The United States Pharmacopeia and The National Formulary. Ancillary Materials for Cell, Gene, and Tissue-Engineered Products. USP 29 General Chapter <1043> NF 24 (USP, 2006).

  19. Atouf, F., Provost, N.M. & Rosenthal, F.M. Standards for ancillary materials used in cell- and tissue-based therapies. Bioprocess Int. 11, 12–22 (2013).

    Google Scholar 

  20. Chen, X. et al. Investigations into the metabolism of two-dimensional colony and suspended microcarrier cultures of human embryonic stem cells in serum-free media. Stem Cells Dev. 19, 1781–1792 (2010).

    Article  CAS  Google Scholar 

  21. Csaszar, E. et al. Rapid expansion of human hematopoietic stem cells by automated control of inhibitory feedback signaling. Cell Stem Cell 10, 218–229 (2012).

    Article  CAS  Google Scholar 

  22. Kempf, H. et al. Controlling expansion and cardiomyogenic differentiation of human pluripotent stem cells in scalable suspension culture. Stem Cell Reports 3, 1132–1146 (2014).

    Article  CAS  Google Scholar 

  23. Csaszar, E., Chen, K., Caldwell, J., Chan, W. & Zandstra, P.W. Real-time monitoring and control of soluble signaling factors enables enhanced progenitor cell outputs from human cord blood stem cell cultures. Biotechnol. Bioeng. 111, 1258–1264 (2013).

    Article  Google Scholar 

  24. Nazareth, E.J. et al. High-throughput fingerprinting of human pluripotent stem cell fate responses and lineage bias. Nat. Methods 10, 1225–1231 (2013).

    Article  CAS  Google Scholar 

  25. June, C.H., Riddell, S.R. & Schumacher, T.N. Adoptive cellular therapy: a race to the finish line. Sci. Transl. Med. 7, 280ps7 (2015).

    Article  Google Scholar 

  26. Thomas, R.J., Hourd, P.C. & Williams, D.J. Application of process quality engineering techniques to improve the understanding of the in vitro processing of stem cells for therapeutic use. J. Biotechnol. 136, 148–155 (2008).

    Article  CAS  Google Scholar 

  27. Hunt, M., Meng, G., Rancourt, D., Gates, I.D. & Kallos, M.S. Factorial experimental design for the culture of human embryonic stem cells as aggregates in stirred suspension bioreactors reveals the potential for interaction effects between bioprocess parameters. Tissue Eng. Part C Methods 20, 76–89 (2013).

    Article  Google Scholar 

  28. Ratcliffe, E. et al. Application of response surface methodology to maximize the productivity of scalable automated human embryonic stem cell manufacture. Regen. Med. 8, 39–48 (2013).

    Article  CAS  Google Scholar 

  29. Zandstra, P.W., Eaves, C.J. & Piret, J.M. Expansion of hematopoietic progenitor cell populations in stirred suspension bioreactors of normal human bone marrow cells. Biotechnology (NY) 12, 909–914 (1994).

    CAS  Google Scholar 

  30. Dahlberg, A., Delaney, C. & Bernstein, I.D. Ex vivo expansion of human hematopoietic stem and progenitor cells. Blood 117, 6083–6090 (2011).

    Article  CAS  Google Scholar 

  31. Dexter, T.M., Allen, T.D. & Lajtha, L.G. Conditions controlling the proliferation of haemopoietic stem cells in vitro. J. Cell. Physiol. 91, 335–344 (1977).

    Article  CAS  Google Scholar 

  32. Marturana, F., Timmins, N.E. & Nielsen, L.K. Short-term exposure of umbilical cord blood CD34+ cells to granulocyte-macrophage colony-stimulating factor early in culture improves ex vivo expansion of neutrophils. Cytotherapy 13, 366–377 (2011).

    Article  CAS  Google Scholar 

  33. Hewitt, C.J. et al. Expansion of human mesenchymal stem cells on microcarriers. Biotechnol. Lett. 33, 2325–2335 (2011).

    Article  CAS  Google Scholar 

  34. Selekman, J.A., Das, A., Grundl, N.J. & Palecek, S.P. Improving efficiency of human pluripotent stem cell differentiation platforms using an integrated experimental and computational approach. Biotechnol. Bioeng. 110, 3024–3037 (2013).

    Article  CAS  Google Scholar 

  35. Datta, P., Linhardt, R.J. & Sharfstein, S.T. An 'omics approach towards CHO cell engineering. Biotechnol. Bioeng. 110, 1255–1271 (2013).

    Article  CAS  Google Scholar 

  36. Shyh-Chang, N. et al. Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 339, 222–226 (2013).

    Article  Google Scholar 

  37. Wang, J. et al. Dependence of mouse embryonic stem cells on threonine catabolism. Science 325, 435–439 (2009).

    Article  CAS  Google Scholar 

  38. Turner, J. et al. Metabolic profiling and flux analysis of MEL-2 human embryonic stem cells during exponential growth at physiological and atmospheric oxygen concentrations. PLoS One 9, e112757 (2014).

    Article  Google Scholar 

  39. Qiao, W. et al. PERT: a method for expression deconvolution of human blood samples from varied microenvironmental and developmental conditions. PLOS Comput. Biol. 8, e1002838 (2012).

    Article  CAS  Google Scholar 

  40. Dunn, S.-J., Martello, G., Yordanov, B., Emmott, S. & Smith, A.G. Defining an essential transcription factor program for naïve pluripotency. Science 344, 1156–1160 (2014).

    Article  CAS  Google Scholar 

  41. US Food and Drug Administration. Guidance for Industry. PAT: a Framework for Innovative Pharmaceutical Development, Manufacturing and Quality Assurance. http://www.fda.gov/downloads/Drugs/.../Guidances/ucm070305.pdf (Pharmaceutical CGMPs, 2004).

  42. Quintás-Cardama, A. et al. Multifactorial optimization of gammaretroviral gene transfer into human T lymphocytes for clinical application. Hum. Gene Ther. 18, 1253–1260 (2007).

    Article  Google Scholar 

  43. Csaszar, E., Cohen, S. & Zandstra, P.W. Blood stem cell products: toward sustainable benchmarks for clinical translation. BioEssays 35, 201–210 (2013).

    Article  CAS  Google Scholar 

  44. Molodecky, N.A. et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 142, 46–54.e42 (2012).

    Article  Google Scholar 

  45. Health Canada. Summary basis of decision for Prochymal. http://www.hc-sc.gc.ca/dhp-mps/prodpharma/sbd-smd/drug-med/sbd_smd_2012_prochymal_150026-eng.php (2012).

  46. Prowse, A.B.J. et al. Transforming the promise of pluripotent stem cell-derived cardiomyocytes to a therapy: challenges and solutions for clinical trials. Can. J. Cardiol. 30, 1335–1349 (2014).

    Article  Google Scholar 

  47. Mendicino, M., Bailey, A.M., Wonnacott, K., Puri, R.K. & Bauer, S.R. MSC-based product characterization for clinical trials: an FDA perspective. Cell Stem Cell 14, 141–145 (2014).

    Article  CAS  Google Scholar 

  48. Ratcliffe, E., Glen, K.E., Workman, V.L., Stacey, A.J. & Thomas, R.J. A novel automated bioreactor for scalable process optimisation of haematopoietic stem cell culture. J. Biotechnol. 161, 387–390 (2012).

    Article  CAS  Google Scholar 

  49. Lipsitz, Y.Y. & Zandstra, P.W. Human pluripotent stem cell process parameter optimization in a small scale suspension bioreactor. BMC Proceedings 9 (Suppl 9), O10 (2015).

    Article  Google Scholar 

Download references

Acknowledgements

We thank members of the P.Z. laboratory as well as D. Baksh, M. Curtis, E. Csaszar, J. Lebkowski, A. Lipsitz and R. Preti for constructive comments and apologize to colleagues whose research we were unable to cite due to space constraints. F. Agbanyo, chief of Blood, Cells, Tissues and Organs Division, Health Products and Food Branch Health Canada, Government of Canada is gratefully acknowledged for her careful review and suggested revisions of this manuscript. For clarity, the perspectives expressed in this paper do not necessarily represent the opinion of Health Canada. Y.Y.L. is supported by an NSERC Alexander Graham Bell Canada Graduate Scholarship, and P.W.Z. is supported as the Canada Research Chair in Stem Cell Bioengineering.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Peter W Zandstra.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lipsitz, Y., Timmins, N. & Zandstra, P. Quality cell therapy manufacturing by design. Nat Biotechnol 34, 393–400 (2016). https://doi.org/10.1038/nbt.3525

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.3525

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research