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
Research ArticleOpen Accesscc iconby icon

Institutional and infrastructure challenges for hospitals producing advanced therapies in the UK: the concept of ‘point-of-care manufacturing readiness’

    Edison Bicudo

    *Author for correspondence:

    E-mail Address: e.bicudo@ucl.ac.uk

    Department of Science, Technology, Engineering, & Public Policy, University College London, Shropshire House (4th Floor), 11–20 Capper Street, London, WC1E 6JA, UK

    &
    Irina Brass

    Department of Science, Technology, Engineering, & Public Policy, University College London, Shropshire House (4th Floor), 11–20 Capper Street, London, WC1E 6JA, UK

    Published Online:https://doi.org/10.2217/rme-2022-0064

    Aim: To propose the concept of point-of-care manufacturing readiness for analyzing the capacity that a country, a health system or an institution has developed to manufacture therapies in clinical settings (point-of-care manufacture). The focus is on advanced therapies (cell, gene and tissue engineering therapies) in the UK. Materials & methods: Literature review, analysis of quantitative data, and qualitative interviews with professionals and practitioners developing and administering advanced therapies. Results: Three components of point-of-care manufacturing readiness are analyzed staff and institutional procedures, infrastructure, and relations between hospitals and service providers. Conclusion: The technical and regulatory experience that has been gained through manufacturing advanced therapies at small scale in hospitals qualifies the UK for more complex and larger-scale production of therapies in the future.

    Graphical abstract

    Point-of-care manufacturing readiness, its components and its challenges

    Plain language summary

    Point-of-care manufacture is the production of therapies in hospitals, carried out when there is no time for storing the medicine, which is delivered to the patient with no delays. Such procedures can be useful for advanced therapies derived from techniques such as gene editing, cell manipulation and tissue engineering. Over the last decades, UK hospitals have produced advanced therapies in small quantities. In the future, this production will likely be more thoroughly integrated into clinical routines. In this way, the technical and regulatory experience that hospitals have accumulated so far underpins the more frequent and larger-scale work expected for the future. This accumulation of expertise, infrastructure and institutional contacts provides the foundation for what we call ‘point-of-care manufacturing readiness’.

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

    References

    • 1. Sadin SR, Povinelli FP, Rosen R. The NASA technology push towards future space mission systems. Acta Astronautica 20, 73–77 (1989).
    • 2. Wu C, Wang B, Zhang C, Wysk RA, Chen Y-W. Bioprinting: an assessment based on manufacturing readiness levels. Crit. Rev. Biotechnol. 37(3), 333–354 (2017).
    • 3. US Department of Defense. Manufacturing readiness level (MRL) deskbook (2018). www.dodmrl.com/MRL_Deskbook_2018.pdf
    • 4. Dye E, Sturgess A, Maheshwari G et al. Examining manufacturing readiness for breakthrough drug development. AAPS PharmSciTech 17(3), 529–538 (2015).
    • 5. Creelman B, Frivold C, Jessup S, Saxon G, Jarrahian C. Manufacturing readiness assessment for evaluation of the microneedle array patch industry: an exploration of barriers to full-scale manufacturing. Drug Deliv. Transl. Res. 12(2), 368–375 (2021).
    • 6. Hourd P, Williams DJ. Scanning the horizon for high value-add manufacturing science: accelerating manufacturing readiness for the next generation of disruptive, high-value curative cell therapeutics. Cytotherapy 20(5), 759–767 (2018).
    • 7. De Wilde S, Guchelaar H-J, Zandvliet ML, Meij P. Clinical development of gene- and cell-based therapies: overview of the European landscape. Mol. Ther. Methods Clin. Dev. 3, 1–7 (2016).
    • 8. Pearce KF, Hildebrandt M, Greinix H et al. Regulation of advanced therapy medicinal products in Europe and the role of academia. Cytotherapy 16(3), 289–297 (2014).
    • 9. Advanced Therapy Treatment Centres. Example risk assessment proforma for proposed activities involving Gene Therapy Medicinal Products (GTMPs) or Gene Therapy Investigational Medicinal Products (GTIMPs) (2022). www.theattcnetwork.co.uk/wp-content/uploads/2021/08/WB-GTMP-RISK-ASSESSMENT-07.21.pdf
    • 10. Kaddis JS, Hanson MS, Cravens J et al. Standardized transportation of human islets: an islet cell resource center study of more than 2000 shipments. Cell Transplant. 22(7), 1101–1111 (2013).
    • 11. Robertson K, Roberts L, Plummer D, Ede J. Complexity and workload of cytotoxic manufacturing at a regional hospital: a comparative study. J. Pharm. Pract. Res. 42(3), 193–195 (2012).
    • 12. Bicudo E, Brass I, Carmichael P, Farid S. The UK’s emerging regulatory framework for point-of-care manufacture: insights from a workshop on advanced therapies. Cell & Gene Therapy Insights 7(9), 1005–1015 (2021).
    • 13. Bicudo E, Brass I, Carmichael P. Point-of-care manufacture of advanced therapies: readiness measures for hospitals, companies, and regulatory agencies. (2022). Accessed in August 2022. Available at: www.ucl.ac.uk/steapp/sites/steapp/files/point-of-care_manufacture_of_advanced_therapies_fthm_policy_brief_jul_2022.pdf
    • 14. Harrison RP, Rafiq QA, Medcalf N. Automating decentralized manufacturing of cell and gene therapy products. Cell Gene Ther. Insights 2(4), 489–497 (2016).
    • 15. Iancu EM, Kandalaft LE. Challenges and advantages of cell therapy manufacturing under good manufacturing practices within the hospital setting. Curr. Opin. Biotechnol. 65, 233–241 (2020). •• An overview of the challenges of GMP advanced therapy medicinal products manufacture in hospitals.
    • 16. Sheba Online. On-site manufacture of CAR-T cells enables faster effective treatments. Sheba Online (2022). www.shebaonline.org/on-site-manufacture-of-car-t-cell-products/
    • 17. Dimitropoulos G, Jafari P, De Buys Roessingh A, Hirt-Burri N, Raffoul W, Applegate LA. Burn patient care lost in good manufacturing practices? Ann. Burns Fire Disasters 29(2), 111–115 (2016).
    • 18. Buchholz CJ, Sanzenbacher R, Schüle S. The European hospital exemption clause: new option for gene therapy? Hum. Gene Ther. 23, 7–12 (2012).
    • 19. Coppens DG, Gardarsdottir H, De Bruin ML, Meij P, Leufkens HGM, Hoekman J. Regulating advanced therapy medicinal products through the Hospital Exemption: an analysis of regulatory approaches in nine EU countries. Regen. Med. 15(8), 2015–2028 (2020). • Examples of how the hospital exemption has been used in the EU.
    • 20. Hills A, Awigena-Cook J, Genenz K et al. An assessment of the hospital exemption landscape across European member states: regulatory frameworks, use and impact. Cytotherapy 22, 772–779 (2020).
    • 21. Medicines and Healthcare Products Regulatory Agency. Regulation (EC) n. 1394/2007 on Advanced Therapy Medicinal Products (‘the ATMP regulation’): guidance on the UK’s arrangements under the hospital exemption scheme. (1), 1–10 (2010). https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/397738/Guidance_on_the_UK_s_arrangements_under_the_hospital_exemption_scheme.pdf
    • 22. Thomson Reuters. Unlicensed medicinal products in the UK. 1–10 (2017). www.arnoldporter.com/~/media/files/perspectives/publications/2017/02/unlicensed-medicinal-products-in-the-uk.pdf • An overview of the UK’s Specials manufacture regulatory scheme.
    • 23. Thrasher AJ. ‘Special exemptions’: should they be put on trial? Mol. Ther. 21(2), 261–262 (2013).
    • 24. Viganò M, Giordano R, Lazzari L. Challenges of running a GMP facility for regenerative medicine in a public hospital. Regen. Med. 12(7), 803–813 (2017).
    • 25. Kaiser AD, Assenmacher M, Schröder B et al. Towards a commercial process for the manufacture of genetically modified T cells for therapy. Cancer Gene Ther. 22, 72–78 (2015).
    • 26. González SL, Jendres CD, Samur EA. Evaluación de buenas prácticas de manufactura en la elaboración de fórmulas enterales en hospitales públicos de Santiago, Chile. Nutricion Hospitalaria 28(6), 2021–2026 (2013).
    • 27. Girma M, Desale A, Hassen F, Sisay A, Tsegaye A. Survey-defined and interview-elicited challenges that faced Ethiopian government hospital laboratories as they applied ISO 15189 accreditation standards in resource-constrained settings in 2017. Am. J. Clin. Pathol. 150(4), 303–309 (2018).
    • 28. Bicudo E. Pharmaceutical research, democracy and conspiracy: international clinical trials in local medical institutions. Routledge, London (2014).
    • 29. Gardner J, Webster A, Barry J. Anticipating the clinical adoption of regenerative medicine: building institutional readiness in the UK. Regen. Med. 13(1), 29–39 (2018). •• The concept of institutional readiness.
    • 30. Hodder I. The interpretation of documents and material culture. In: Handbook of Qualitative Research. Denzin NKLincoln YS (Eds). Sage, Thousand Oaks, CA, USA, 393–402 (1994).
    • 31. European Parliament and Council of the EU. Regulation EC 1394/2007 of the European Parliament and the Council (2007). https://eur-lex.europa.eu/LexUriServ/LexUriServ.do?uri=OJ:L:2007:324:0121:0137:EN:PDF
    • 32. Medicines and Healthcare Products Regulatory Agency. The supply of unlicensed medicinal products (‘specials’). Guidance Note 14 (2014). https://assets.publishing.service.gov.uk/government/uploads/system/uploads/attachment_data/file/373505/The_supply_of_unlicensed_medicinal_products__specials_.pdf
    • 33. Medicines and Healthcare Products Regulatory Agency. Consultation on point of care manufacturing. www.gov.uk/government/consultations/point-of-care-consultation/consultation-on-point-of-care-manufacturing •• The Medicines and Healthcare Products Regulatory Agency’s regulatory framework proposal.
    • 34. Medicines and Healthcare Products Regulatory Agency. Register of licensed manufacturing sites (2021). www.gov.uk/government/publications/human-and-veterinary-medicines-register-of-licensed-manufacturing-sites
    • 35. Human Tissue Authority. Find an establishment (2022). www.hta.gov.uk/professional/establishments
    • 36. R Core Team. R: a language and environment for statistical computing (2019). www.R-project.org/
    • 37. Newcastle University. First patient receives NHS stem cell treatment for rare eye condition (2019). www.ncl.ac.uk/press/articles/archive/2019/12/sight-savingtreatment/
    • 38. Advanced Therapy Treatment Centres. Advanced Therapies NHS readiness toolkit (2022). www.theattcnetwork.co.uk/nhs-readiness-toolkit-results?document_type=201&treatment_type=&sort=&search=&Search=Search • Practical measures for enhancing readiness in advanced therapy medicinal products in hospitals.
    • 39. Black A, Caulfield D. The role of a qualified person within advanced therapy medicinal products (ATMPs). The ATTC Network (2021). https://attc-143fd.kxcdn.com/wp-content/uploads/2021/11/NAAATC_The-Role-of-the-QP.pdf
    • 40. Wick J. ISO certifications in the medical field: the must-have standards. ISO Update (2020). https://isoupdate.com/general/iso-certifications-in-the-medical-field-the-must-have-standards/
    • 41. Green AD, Kavanagh-Wright L, Lee GR. Investigation of the long-term yield of auditing for conformity with the ISO 15189:2012 quality standard in a hospital pathology laboratory. Pract. Lab. Med. 20(e00159), 1–6 (2020).
    • 42. O’Connor L, Malkin A, Carroll B. Evaluating the impact of ISO 15189 on an Irish histopathology laboratory. Biomed. Scientist September 2016, 476–482 (2016).
    • 43. Seddon J. Ten arguments against ISO 9000. Managing Service Quality 7(4), 162–168 (1997).
    • 44. Wilson IG, Smye M, Wallace JC. Meta-audit of laboratory ISO accreditation inspections: measuring the old emperor's clothes. MicrobiologyOpen 51(1), 95–105 (2016).
    • 45. Fact-Jacie. International standards for hematopoietic cellular therapy: product collection, processing, and administration (8th edition). (2021). www.ebmt.org/8th-edition-fact-jacie-standards
    • 46. Gannon PO, Harari A, Auger A et al. Development of an optimized closed and semi-automatic protocol for good manufacturing practice manufacturing of tumor-infiltrating lymphocytes in a hospital environment. Cytotherapy 22(12), 780–791 (2020).
    • 47. Petrenko Y, Chudickova M, Vackova I et al. Clinically relevant solution for the hypothermic storage and transportation of human multipotent mesenchymal stromal cells. Stem Cells Int. 11, 1–11 (2019).
    • 48. Medcalf N. Centralized or decentralized manufacturing? Key business model considerations for cell therapies. Cell Gene Ther. Insights 2(1), 95–109 (2016). • An analysis of the possible models for therapy manufacture in decentralized ways.
    • 49. Cell and Gene Therapy Catapult. Cell and gene therapy GMP manufacturing in the UK: capability and capacity analysis (2021). https://ct.catapult.org.uk/sites/default/files/publication/2021%20GMP%20Manufacturing%20Report.pdf
    • 50. Bersenev A, Fesnak A. Place of academic GMP facilities in modern cell therapy. In: Cell Reprogramming for Immunotherapy: Methods and Protocols. Katz SGRabinovich PM (Eds). Humana Press, New York, NY, USA, 329–339 (2020).
    • 51. Smethurst PA, Jolley J, Braund R et al. Rejuvenation of RBCs: validation of a manufacturing method suitable for clinical use. Transfusion 59(9), 2952–2963 (2019).
    • 52. Veronesi E, Murgia A, Caselli A et al. Transportation conditions for prompt use of ex vivo expanded and freshly harvested clinical-grade bone marrow mesenchymal stromal/stem cells for bone regeneration. Tissue Eng. Part C Methods 20(3), 239–251 (2014).
    • 53. Whiteside TL, Griffin DL, Stanson J et al. Shipping of therapeutic somatic cell products. Cytotherapy 13(2), 201–213 (2011).
    • 54. Yu N-H, Chun SY, Ha Y-S et al. Optimal stem cell transporting conditions to maintain cell viability and characteristics. Tissue Eng. Regen. Med. 15(5), 639–647 (2018).
    • 55. Howard A, Chitphakdithai P, Waller EK et al. Evaluation of peripheral blood stem cell quality in products transported by traditional courier or commercial overnight shipping services. Transfusion 54(6), 1501–1507 (2014).
    • 56. Dyson PG, Tocchetti R, Hiwase S. Validation of an alternative transport system for cryopreserved haemopoietic progenitor cells. Cytotherapy 16(4), S56 (2014).
    • 57. Oie Y, Nozaki T, Takayanagi H et al. Development of a cell sheet transportation technique for regenerative medicine. Tissue Eng. Part C Methods 20(5), 373–382 (2014).
    • 58. Alliance for Regenerative Medicine. Position on hospital exemption (2017). https://alliancerm.org/sites/default/files/ARM_position_on_HE_final.pdf
    • 59. Nakano LA, Cançado ELR, Chaves CE et al. A randomized crossover trial to assess therapeutic efficacy and cost reduction of acid ursodeoxycholic manufactured by the university hospital for the treatment of primary biliary cholangitis. BMC Gastroenterol. 20(1), 1–8 (2020).
    • 60. Singh H, Figliola MJ, Dawson MJ et al. Manufacture of clinical-grade CD19-specific T cells stably expressing chimeric antigen receptor using Sleeping Beauty system and artificial antigen presenting cells. PLoS ONE 8(5), e64138 (2013).
    • 61. Van Der Loo JCM, Swaney WP, Grassman E et al. Scale-up and manufacturing of clinical-grade self-inactivating gamma-retroviral vectors by transient transfection. Gene Ther. 19(3), 246–254 (2012).
    • 62. Juan M, Delgado J, Calvo G, Trias E, Urbano-Ispizua Á. Is hospital exemption an alternative or a bridge to European Medicines Agency for developing academic chimeric antigen receptor T-cell in Europe? Our experience with ARI-0001. Hum. Gene Ther. 32, 1004–1007 (2021).
    • 63. Gafni A. Alternatives to the QALY measure for economic evaluations. Support. Care Cancer 5, 105–111 (1997).
    • 64. Digiusto DL, Melsop K, Srivastava R, Tran C-aT. Proceedings of the first academic symposium on developing, qualifying and operating a cell and gene therapy manufacturing facility. Cytotherapy 20, 1486–1494 (2018).
    • 65. Lanzarone E, Marconi S, Conti M et al. Hospital factory for manufacturing customised, patient-specific 3D anatomo-functional models and prostheses. In: Factories of the Future: the Italian Flagship Initiative. Tolio TCopani GTerkaj W (Eds).Springer, Cham, Switzerland, 233–254 (2019).
    • 66. Webster A, Gardner J. Aligning technology and institutional readiness: the adoption of innovation. Technol. Anal. Strategic Manag. 31(10), 1229–1241 (2019).
    • 67. E Bicudo, A Faulkner, P Li. Digital readinessin 3D bioprinting: software, governance and hospitals’ proto-clinical interfaces. Reg.Med. 16(3), 237–252 (2022).