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

The early career researcher's toolkit: translating tissue engineering, regenerative medicine and cell therapy products

    Qasim A Rafiq

    *Author for correspondence:

    E-mail Address: q.rafiq@aston.ac.uk

    Centre for Biological Engineering, Wolfson School of Mechanical & Manufacturing Engineering, Loughborough University, Leicestershire, LE11 3TU, UK

    Aston Medical Research Institute, School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Ilida Ortega

    Bioengineering & Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, S10 2TA, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Stuart I Jenkins

    Institute for Science & Technology in Medicine, Keele University, Staffordshire, ST5 5BG, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Samantha L Wilson

    Academic Ophthalmology, Division of Clincial Neuroscience, Queen's Medical Centre Campus, University of Nottingham, NG7 2UH, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Asha K Patel

    Wolfson Centre for Stem Cells, Tissue Engineering & Modeling, University of Nottingham, Nottingham, NG7 2RD, UK

    David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA

    All authors contributed equally.

    Search for more papers by this author

    ,
    Amanda L Barnes

    Department of Biology, University of York, YO10 5DD, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Christopher F Adams

    Institute for Science & Technology in Medicine, Keele University, Staffordshire, ST5 5BG, UK

    All authors contributed equally.

    Search for more papers by this author

    ,
    Derfogail Delcassian

    Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK

    Wolfson Centre for Stem Cells, Centre for Biological Sciences, School of Pharmacy, University of Nottingham, NG7 2RD, UK

    All authors contributed equally.

    Search for more papers by this author

    &
    David Smith

    Centre for Biological Engineering, Wolfson School of Mechanical & Manufacturing Engineering, Loughborough University, Leicestershire, LE11 3TU, UK

    PCT, a Caladrius company, 4 Pearl Court, Suite C, Allendale, NJ 07401, USA

    All authors contributed equally.

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/rme.15.56

    Although the importance of translation for the development of tissue engineering, regenerative medicine and cell-based therapies is widely recognized, the process of translation is less well understood. This is particularly the case among some early career researchers who may not appreciate the intricacies of translational research or make decisions early in development which later hinders effective translation. Based on our own research and experiences as early career researchers involved in tissue engineering and regenerative medicine translation, we discuss common pitfalls associated with translational research, providing practical solutions and important considerations which will aid process and product development. Suggestions range from effective project management, consideration of key manufacturing, clinical and regulatory matters and means of exploiting research for successful commercialization.

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

    References

    • 1 Cyranoski D. Collateral damage: how a case of misconduct brought a leading Japanese biology institute to its knees. Nature 520(7549), 600–603 (2015).
    • 2 Cyranoski D. Whistle-blower breaks his silence. Nature 505(7485), 593–594 (2014).
    • 3 Collins FS, Tabak LA. Policy: NIH plans to enhance reproducibility. Nature 505(7485), 612–613 (2014).
    • 4 Freedman LP, Cockburn IM, Simcoe TS. The economics of reproducibility in preclinical research. PLoS Biol. 13(6), e1002165 (2015).
    • 5 Fogler HS. Experimental planning. In: Essentials of Chemical Reaction Engineering. Fogler HS (Ed.). Pearson Education, Inc., NJ, USA, 271–272 (2011).
    • 6 Kirouac DC, Zandstra PW. The systematic production of cells for cell therapies. Cell Stem Cell 3(4), 369–381 (2008).
    • 7 Placzek MR, Chung IM, Macedo HM et al. Stem cell bioprocessing: fundamentals and principles. J. R. Soc. Interface 6(32), 209–232 (2009).
    • 8 Williams DJ, Thomas RJ, Hourd PC et al. Precision manufacturing for clinical–quality regenerative medicines. Philos. Trans. A. Math. Phys. Eng. Sci. 370(1973), 3924–3949 (2012).•• An overview of the importance of precision manufacturing in the tissue engineering and regenerative medicine field with a description of quality engineering techniques and their application for successful translation
    • 9 Ratcliffe E, Thomas RJ, Williams DJ. Current understanding and challenges in bioprocessing of stem cell-based therapies for regenerative medicine. Br. Med. Bull. 100(1), 137–155 (2011).
    • 10 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 15(1), 9–19 (2013).• A comprehensive review of the requirements for defining and measuring the quality of cellular products, a prerequisite for successful translation.
    • 11 Burger SR, Juliano L, Wang W. Cellular raw material collection in cell therapy: critical determinant of product quality. Drug Discov. World 15(3), 29–34 (2014).
    • 12 Lionberger RA, Lee SL, Lee L, Raw A, Yu LX. Quality by design: concepts for andas. AAPS J. 10(2), 268–276 (2008).
    • 13 Zimmerman RL. 10 best practices of good laboratories. ASTM Stand. News 38(6), 24–31 (2010).
    • 14 Brosnan K, Want A, Coopman K, Hewitt CJ. Multiparameter flow cytometry for the characterization of human embryonic stem cells. Biotechnol. Lett. 35(1), 55–65 (2013).
    • 15 Shapiro HM. Practical Flow Cytometry. John Wiley & Sons, Hoboken, NJ, USA (2005).
    • 16 Plagnol AC, Rowley E, Martin P, Livesey F. Industry perceptions of barriers to commercialization of regenerative medicine products in the UK. Regen. Med. 4(4), 549–559 (2009).
    • 17 Couto DS, Perez-Breva L, Cooney CL. Regenerative medicine: learning from past examples. Tissue Eng. Part A 18(21–22), 2386–2393 (2012).
    • 18 Cai M, Yang Y. Targeted genome editing tools for disease modeling and gene therapy. Curr. Gene Ther. 14(1), 2–9 (2014).
    • 19 Mason C. Regenerative medicine 2.0. Regen. Med. 2(1), 11–18 (2007).
    • 20 Bubela T, Mccabe C. Value-engineered translation for regenerative medicine: meeting the needs of health systems. Stem Cells Dev. 22(Suppl. 1), 89–93 (2013).
    • 21 Bertram TA, Tentoff E, Johnson PC, Tawil B, Van Dyke M, Hellman KB. Hurdles in tissue engineering/regenerative medicine product commercialization: a pilot survey of governmental funding agencies and the financial industry. Tissue Eng. Part A 18(21–22), 2187–2194 (2012).
    • 22 Weinstein MC, Torrance G, McGuire A. QALYs: the basics. Value Health 12, S5–S9 (2009).
    • 23 Wouters S, Van Exel NJA, Rohde KIM, Brouwer WBF. Are all health gains equally important? An exploration of acceptable health as a reference point in health care priority setting. Health Qual. Life Outcomes 13(1), 1–10 (2015).
    • 24 Brinker M, O'connor D. Stakeholders in outcome measures: review from a clinical perspective. Clin. Orthop. Relat. Res. 471(11), 3426–3436 (2013).
    • 25 Mcateer H, Cosh E, Freeman G, Pandit A, Wood P, Lilford R. Cost-effectiveness analysis at the development phase of a potential health technology: examples based on tissue engineering of bladder and urethra. J. Tissue Eng. Regen. Med. 1(5), 343–349 (2007).
    • 26 Trounson A, Baum E, Gibbons D, Tekamp-Olson P. Developing a case study model for successful translation of stem cell therapies. Cell Stem Cell 6(6), 513–516 (2010).
    • 27 Rose JB, May M, Williams DJ. Determinants of clinician adoption of regenerative therapies in the UK and Canada: an ophthalmology perspective. Regen. Med. 10(4), 1–13 (2015).
    • 28 Champkin J. George E.P. Box, 1919–2013. Significance 10(3), 32–34 (2013).
    • 29 Box GEP. Robustness in the strategy of scientific model building. In: Robustness in Statistics. Launer RL, Wilkinson GN (Eds). Academic Press, NY, USA, 201–236 (1979).
    • 30 Leist M, Hartung T. Inflammatory findings on species extrapolations: humans are definitely no 70-kg mice. Arch. Toxicol. 87(4), 563–567 (2013).
    • 31 Lemmon VP, Ferguson AR, Popovich PG et al. Minimum information about a spinal cord injury experiment: a proposed reporting standard for spinal cord injury experiments. J. Neurotrauma 31(15), 1354–1361 (2014).
    • 32 Lowenstein PR, Castro MG. Uncertainty in the translation of preclinical experiments to clinical trials. Why do most Phase III clinical trials fail? Curr. Gene Ther. 9(5), 368–374 (2009).• An easily digestible survey of why clinical failures occur with an important rallying call for ‘preclinical robustness’.
    • 33 Cook N, Jodrell DI, Tuveson DA. Predictive in vivo animal models and translation to clinical trials. Drug Discov. Today 17(5–6), 253–260 (2012).
    • 34 Andersen ME, Betts K, Dragan Y et al. Developing microphysiological systems for use as regulatory tools – challenges and opportunities. Altex 31(3), 364–367 (2014).
    • 35 Balls M. The origins and early days of the three Rs concept. Altern. Lab. Anim. 37(3), 255–265 (2009).
    • 36 Liebsch M, Grune B, Seiler A et al. Alternatives to animal testing: current status and future perspectives. Arch. Toxicol. 85(8), 841–858 (2011).
    • 37 Varga OE, Hansen AK, Sandoe P, Olsson IA. Validating animal models for preclinical research: a scientific and ethical discussion. Altern. Lab. Anim. 38(3), 245–248 (2010).• A comprehensive overview of the urgent need for, and a proposed route toward, preclinical model validation.
    • 38 US FDA. Guidance for industry and FDA staff: classification of products as drugs and devices and additional product classification issues (2011). www.fda.gov/downloads/RegulatoryInformation/Guidances/UCM258957.pdf.
    • 39 Lee JK, Responte DJ, Cissell DD, Hu JC, Nolta JA, Athanasiou KA. Clinical translation of stem cells: insight for cartilage therapies. Crit. Rev. Biotechnol. 34(1), 89–100 (2014).
    • 40 FDA. Definition of primary mode of action of a combination product. Final rule. Fed. Regist. 70(164), 49848–49862 (2005).
    • 41 Kramer DB, Xu S, Kesselheim AS. Regulation of medical devices in the United States and European Union. N. Engl. J. Med. 366(9), 848–855 (2012).
    • 42 Pashuck ET, Stevens MM. Designing regenerative biomaterial therapies for the clinic. Sci. Transl Med. 4(160), 160sr164 (2012).
    • 43 European Medicines Agency, Committee for Advanced Therapies (CAT). Reflection paper on classification of advanced therapy medicinal products (ema/cat/600280/2010) (2012). www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2015/06/WC500187744.pd.
    • 44 Baum E, Littman N, Ruffin M, Ward S, Aschheim K. White paper: key tools and technology hurdles in advancing stem-cell therapies (2013). www.cirm.ca.gov/files/files/funding_page/Key-Tools-Tech-Hurdles-in-Advancing-Stem-Cell-Therapies.pdf.
    • 45 Halme DG, Kessler DA. Fda regulation of stem-cell-based therapies. N. Engl. J. Med. 355(16), 1730–1735 (2006).
    • 46 Prestwich GD, Bhatia S, Breuer CK et al. What is the greatest regulatory challenge in the translation of biomaterials to the clinic? Sci. Transl Med. 4(160), 160cm114 (2012).
    • 47 FDA. Medical devices containing materials derived from animal sources (except for in vitro diagnostic devices) 2014. www.fda.gov/downloads/MedicalDevices/DeviceRegulationandGuidance/GuidanceDocuments/UCM381491.pdf.
    • 48 Kemp P. History of regenerative medicine: looking backwards to move forwards. Regen. Med. 1(5), 653–669 (2006).
    • 49 US FDA. Summary of safety and effectiveness data for Apligraf™ (1998). www.accessdata.fda.gov/cdrh_docs/pdf/P950032b.pdf.
    • 50 Azuma K. Regulatory landscape of regenerative medicine in Japan. Curr. Stem Cell Rep. 1(2), 118–128 (2015).•• A brief but important paper providing an overview of the regulatory, reimbursement and other strategic changes in Japan with respect to regenerative medicine.
    • 51 Cyranoski D. Japan to offer fast-track approval path for stem cell therapies. Nat. Med. 19(5), 510–510 (2013).
    • 52 Office for Life Sciences, Department for Business Innovation & Skills. Taking stock of regenerative medicine in the United Kingdom (2011). www.gov.uk/government/uploads/system/uploads/attachment_data/file/32459/11-1056-taking-stock-of-regenerative-medicine.pdf.
    • 53 Narkilahti S, Rajala K, Pihlajamaki H, Suuronen R, Hovatta O, Skottman H. Monitoring and analysis of dynamic growth of human embryonic stem cells: comparison of automated instrumentation and conventional culturing methods. Biomed. Eng. Online doi:10.1186/1475-925X-6-11 (2007) (Epub ahead of print).
    • 54 Health Canada. Summary Basis of Decision (SBD) for PROCHYMAL® (2012). www.hc-sc.gc.ca/dhp-mps/prodpharma/sbd-smd/drug-med/sbd_smd_2012_prochymal_150026-eng.php.
    • 55 Thurman-Newell JA, Petzing JN, Williams DJ. Quantification of biological variation in blood-based therapy – a summary of a meta-analysis to inform manufacturing in the clinic. Vox Sang. 6, 11 (2015).
    • 56 Rafiq QA, Hewitt CJ. Cell therapies: why scale matters. Pharm. Bioprocess. 3(2), 97–99 (2015).• A perspective on the importance of understanding the significance and impact of scale for therapeutic bioprocess development.
    • 57 Pollard D. Are automated disposable small-scale reactors set to dominate the future of pharmaceutical bioprocess development? Pharm. Bioprocess. 2(1), 9–12 (2014).
    • 58 Fierabracci A. Recents patents for isolating, delivering and tracking adult stem cells in regenerative medicine. Recent Pat. Drug Deliv. Formul. 4(2), 105–113 (2010).
    • 59 Gov UK. Intellectual property and your work (2014). www.gov.uk/intellectual-property-an-overview/what-ip-is.
    • 60 Willemse L, Lyall D, Rudnicki M. Catalyzing stem cell research. Regen. Med. 3(5), 761–764 (2008).
    • 61 Munisi HI, Xie Z, Sengoku S. Exploring innovation in stem cell and regenerative medicine in Japan: the power of the consortium-based approach. Regen. Med. 9(4), 467–477 (2014).
    • 62 Research Councils UK. Material world: knowledge economy (2004). www.rcuk.ac.uk/Publications/archive/MaterialWorld.
    • 63 British Standards Institute (BSI). PAS83:2012 developing human cells for clinical applications in the European Union and the United States of America (2012). www.cambridge-brc.org.uk/sites/default/files/PAS83-2012%5B1%5D.pdf.