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:

Expanding access to CAR T cell therapies through local manufacturing

Abstract

Chimeric antigen receptor (CAR) T cells are changing the therapeutic landscape for hematological malignancies. To date, all six CAR T cell products approved by the US Food and Drug Administration (FDA) are autologous and centrally manufactured. As the numbers of approved products and indications continue to grow, new strategies to increase cell-manufacturing capacity are urgently needed to ensure patient access. Distributed manufacturing at the point of care or at other local manufacturing sites would go a long way toward meeting the rising demand. To ensure successful implementation, it is imperative to harness novel technologies to achieve uniform product quality across geographically dispersed facilities. This includes the use of automated cell-production systems, in-line sensors and process simulation for enhanced quality control and efficient supply chain management. A comprehensive effort to understand the critical quality attributes of CAR T cells would enable better definition of widely attainable release criteria. To supplement oversight by national regulatory agencies, we recommend expansion of the role of accreditation bodies. Moreover, regulatory standards may need to be amended to accommodate the unique characteristics of distributed manufacturing models.

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

Access options

Buy this article

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

Fig. 1: Distributed manufacturing models.
Fig. 2: Proposed centralized approvals of distributed models.
Fig. 3: Pathways for exemption and exclusion of products from market approval requirements.
Fig. 4: Distribution of FACT-compliant organizations in the United States.

Similar content being viewed by others

References

  1. Schuster, S. J. et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N. Engl. J. Med. 380, 45–56 (2019).

    CAS  PubMed  Google Scholar 

  2. Grupp, S. A., DiNofia, A. M. & Si Lim, S. J. Tisagenlecleucel for treatment of children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia. Pediatr. Blood Cancer 68, e29123 (2021).

    Google Scholar 

  3. Neelapu, S. S. et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N. Engl. J. Med. 377, 2531–2544 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Abramson, J. S. et al. Lisocabtagene maraleucel for patients with relapsed or refractory large B-cell lymphomas (TRANSCEND NHL 001): a multicentre seamless design study. Lancet 396, 839–852 (2020).

    PubMed  Google Scholar 

  5. Brentjens, R. J. et al. Eradication of systemic B-cell tumors by genetically targeted human T lymphocytes co-stimulated by CD80 and interleukin-15. Nat. Med. 9, 279–286 (2003).

    CAS  PubMed  Google Scholar 

  6. Kalos, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    CAS  PubMed  Google Scholar 

  9. Finck, A. V., Blanchard, T., Roselle, C. P., Golinelli, G. & June, C. H. Engineered cellular immunotherapies in cancer and beyond. Nat. Med. 28, 678–689 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu, E. et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N. Engl. J. Med. 382, 545–553 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Rohaan, M. W. et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N. Engl. J. Med. 387, 2113–2125 (2022).

    CAS  PubMed  Google Scholar 

  13. Center for Biologics Evaluation and Research (CBER). Approval Letter—Kymriah. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/106989/download (2017).

  14. Center for Biologics Evaluation and Research (CBER). Approval Letter—Yescarta. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/108458/download (2017).

  15. Center for Biologics Evaluation and Research (CBER). Approval Letter—Tecartus. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/140415/download (2020).

  16. Center for Biologics Evaluation and Research (CBER). Approval Letter—Breyanzi. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/145712/download (2021).

  17. Center for Biologics Evaluation and Research (CBER). Approval Letter—CARVYKTI. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/156572/download (2022).

  18. Center for Biologics Evaluation and Research (CBER). Approval Letter—ABECMA. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/147062/download (2021).

  19. Center for Biologics Evaluation and Research (CBER). Approval Letter—Breyanzi. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/159473/download (2022).

  20. Center for Biologics Evaluation and Research (CBER). Approval letter—Yescarta. U.S. Food and Drug Administration (FDA) https://www.fda.gov/media/157539/download (2022).

  21. Kourelis, T. et al. Ethical challenges with CAR T slot allocation with idecabtagene vicleucel manufacturing access. J. Clin. Oncol. 40, e20021 (2022).

    Google Scholar 

  22. Al Hadidi, S. et al. Clinical outcome of patients with relapsed refractory multiple myeloma listed for BCMA directed commercial CAR-T therapy. Bone Marrow Transplant. 58, 443–445 (2023).

  23. Ahmed, N. et al. ‘Waitlist mortality’ is high for myeloma patients with limited access to BCMA therapy. Front. Oncol. 13, 1206715 (2023).

    PubMed  PubMed Central  Google Scholar 

  24. Locke, F. L. et al. Real-world impact of time from leukapheresis to infusion (vein-to-vein time) in patients with relapsed or refractory (r/r) large B-cell lymphoma (LBCL) treated with axicabtagene ciloleucel. Blood 140, 7512–7515 (2022).

    Google Scholar 

  25. Wang, K. et al. A multiscale simulation framework for the manufacturing facility and supply chain of autologous cell therapies. Cytotherapy 21, 1081–1093 (2019).

    CAS  PubMed  Google Scholar 

  26. Srai, J. S., Badman, C., Krumme, M., Futran, M. & Johnston, C. Future supply chains enabled by continuous processing—opportunities and challenges. May 20–21 2014 Continuous Manufacturing Symposium. J. Pharm. Sci. 104, 840–849 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Abou-el-Enein, M. et al. Scalable manufacturing of CAR T cells for cancer immunotherapy. Blood Cancer Discov. 2, 408–422 (2021).

    CAS  Google Scholar 

  28. Levine, B. L., Miskin, J., Wonnacott, K. & Keir, C. Global manufacturing of CAR T cell therapy. Mol. Ther. Methods Clin. Dev. 4, 92–101 (2017).

    CAS  PubMed  Google Scholar 

  29. Myles, L. & Church, T. D. An industry survey of implementation strategies for clinical supply chain management of cell and gene therapies. Cytotherapy 24, 344–355 (2022).

    CAS  PubMed  Google Scholar 

  30. Papathanasiou, M. M. et al. Autologous CAR T-cell therapies supply chain: challenges and opportunities? Cancer Gene Ther. 27, 799–809 (2020).

    CAS  Google Scholar 

  31. Harrison, R. P., Ruck, S., Medcalf, N. & Rafiq, Q. A. Decentralized manufacturing of cell and gene therapies: overcoming challenges and identifying opportunities. Cytotherapy 19, 1140–1151 (2017).

    PubMed  Google Scholar 

  32. Alqazaqi, R. et al. Geographic and racial disparities in access to chimeric antigen receptor-T cells and bispecific antibodies trials for multiple myeloma. JAMA Netw. Open 5, e2228877 (2022).

    PubMed  PubMed Central  Google Scholar 

  33. Palani, H. K. et al. Decentralized manufacturing of anti CD19 CAR-T cells using CliniMACS Prodigy®: real-world experience and cost analysis in India. Bone Marrow Transplant. 58, 160–167 (2022).

  34. Iancu, E. M. & Kandalaft, L. E. Challenges and advantages of cell therapy manufacturing under good manufacturing practices within the hospital setting. Curr. Opin. Biotechnol. 65, 233–241 (2020).

    CAS  PubMed  Google Scholar 

  35. Benjamin, R. et al. UCART19, a first-in-class allogeneic anti-CD19 chimeric antigen receptor T-cell therapy for adults with relapsed or refractory B-cell acute lymphoblastic leukaemia (CALM): a phase 1, dose-escalation trial. Lancet Haematol. 9, e833–e843 (2022).

    CAS  PubMed  Google Scholar 

  36. Benjamin, R. et al. Genome-edited, donor-derived allogeneic anti-CD19 chimeric antigen receptor T cells in paediatric and adult B-cell acute lymphoblastic leukaemia: results of two phase 1 studies. Lancet 396, 1885–1894 (2020).

    CAS  PubMed  Google Scholar 

  37. Mailankody, S. et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat. Med. 29, 422–429 (2023).

    CAS  PubMed  Google Scholar 

  38. Neelapu, S. S. et al. First-in-human data of ALLO-501 and ALLO-647 in relapsed/refractory large B-cell or follicular lymphoma (R/R LBCL/FL): ALPHA study. J. Clin. Oncol. 38, 8002 (2020).

    Google Scholar 

  39. Ghassemi, S. et al. Reducing ex vivo culture improves the antileukemic activity of chimeric antigen receptor (CAR) T cells. Cancer Immunol. Res. 6, 1100–1109 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Ottaviano, G. et al. Phase 1 clinical trial of CRISPR-engineered CAR19 universal T cells for treatment of children with refractory B cell leukemia. Sci. Transl. Med. 14, eabq3010 (2022).

    CAS  PubMed  Google Scholar 

  41. Chiesa, R. et al. Base-edited CAR7 T cells for relapsed T-cell acute lymphoblastic leukemia. N. Engl. J. Med. 389, 899–910 (2023).

  42. Hu, Y. et al. Genetically modified CD7-targeting allogeneic CAR-T cell therapy with enhanced efficacy for relapsed/refractory CD7-positive hematological malignancies: a phase I clinical study. Cell Res. 32, 995–1007 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Lam, C., Meinert, E., Yang, A. & Cui, Z. Comparison between centralized and decentralized supply chains of autologous chimeric antigen receptor T-cell therapies: a UK case study based on discrete event simulation. Cytotherapy 23, 433–451 (2021).

    CAS  PubMed  Google Scholar 

  44. Ran, T., Eichmüller, S. B., Schmidt, P. & Schlander, M. Cost of decentralized CAR T‐cell production in an academic nonprofit setting. Int. J. Cancer 147, 3438–3445 (2020).

    CAS  PubMed  Google Scholar 

  45. Bersenev, A. & Fesnak, A. Place of academic GMP facilities in modern cell therapy. In Cell Reprogramming for Immunotherapy (eds Katz, S. G. & Rabinovich, P. M.) Vol. 2097, 329–339 (Springer, 2020).

  46. Zhu, F. et al. Closed-system manufacturing of CD19 and dual-targeted CD20/19 chimeric antigen receptor T cells using the CliniMACS Prodigy device at an academic medical center. Cytotherapy 20, 394–406 (2018).

    CAS  PubMed  Google Scholar 

  47. Mock, U. et al. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS Prodigy. Cytotherapy 18, 1002–1011 (2016).

    CAS  PubMed  Google Scholar 

  48. Lock, D. et al. Automated, scaled, transposon-based production of CAR T cells. J. Immunother. Cancer 10, e005189 (2022).

    PubMed  PubMed Central  Google Scholar 

  49. Castella, M. et al. Point-of-care CAR T-cell production (ARI-0001) using a closed semi-automatic bioreactor: experience from an academic phase I clinical trial. Front. Immunol. 11, 482 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Jackson, Z. et al. Automated manufacture of autologous CD19 CAR-T cells for treatment of non-Hodgkin lymphoma. Front. Immunol. 11, 1941 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Shah, N. N. et al. Bispecific anti-CD20, anti-CD19 CAR T cells for relapsed B cell malignancies: a phase 1 dose escalation and expansion trial. Nat. Med. 26, 1569–1575 (2020).

    CAS  PubMed  Google Scholar 

  52. Harrison, R. P., Ruck, S., Rafiq, Q. A. & Medcalf, N. Decentralised manufacturing of cell and gene therapy products: learning from other healthcare sectors. Biotechnol. Adv. 36, 345–357 (2018).

    PubMed  Google Scholar 

  53. Center for Drug Evaluation and Research (CDER). Distributed Manufacturing and Point-of-Care Manufacturing of Drugs https://www.fda.gov/about-fda/reports-budgets-cder/distributed-manufacturing-and-point-care-manufacturing-drugs-discussion-paper (2022).

  54. Chalasani, R., Hershey, T. B. & Gellad, W. F. Cost and access implications of defining CAR-T therapy as a drug. JAMA Health Forum 1, e200868 (2020).

    PubMed  Google Scholar 

  55. U.S. Food & Drug Administration (FDA). FDA Regulation of Human Cells, Tissues, and Cellular and Tissue-Based Products (HCT/P’s) Product List https://public4.pagefreezer.com/browse/FDA/06-02-2023T10:15/https://www.fda.gov/vaccines-blood-biologics/tissue-tissue-products/fda-regulation-human-cells-tissues-and-cellular-and-tissue-based-products-hctps-product-list (2018).

  56. U.S. Food & Drug Administration (FDA). Regulatory Considerations for Human Cells, Tissues, and Cellular and Tissue-Based Products: Minimal Manipulation and Homologous Use https://www.fda.gov/media/109176/download (2020).

  57. U.S. Food & Drug Administration (FDA). Same Surgical Procedure Exception under 21 CFR 1271.15(b): Questions and Answers Regarding the Scope of the Exception https://www.fda.gov/regulatory-information/search-fda-guidance-documents/same-surgical-procedure-exception-under-21-cfr-127115b-questions-and-answers-regarding-scope (2017).

  58. Nagai, S. Regulatory hurdles for CAR T-cell therapy in Japan. Lancet Haematol. 8, e686–e687 (2021).

    CAS  PubMed  Google Scholar 

  59. U.S. Food & Drug Administration (FDA). Identification of Manufacturing Establishments in Applications Submitted to CBER and CDER Questions and Answers https://www.fda.gov/media/131911/download (2019).

  60. U.S. Food & Drug Administration (FDA). Demonstration of Comparability of Human Biological Products, Including Therapeutic Biotechnology-Derived Products https://www.fda.gov/regulatory-information/search-fda-guidance-documents/demonstration-comparability-human-biological-products-including-therapeutic-biotechnology-derived (1996).

  61. Better, M., Chiruvolu, V. & Sabatino, M. Overcoming challenges for engineered autologous T cell therapies. Cell Gene Ther. Insights 4, 173–186 (2018).

    Google Scholar 

  62. U.S. Food & Drug Administration (FDA). Considerations for the Development of Chimeric Antigen Receptor (CAR) T Cell Products https://www.fda.gov/media/156896/download (2022).

  63. Marks, P. & Gottlieb, S. Balancing safety and innovation for cell-based regenerative medicine. N. Engl. J. Med. 378, 954–959 (2018).

    PubMed  Google Scholar 

  64. Marks, P. & Gottlieb, S. Statement from FDA Commissioner Scott Gottlieb, M.D. and Peter Marks, M.D., Ph.D., Director of the Center for Biologics Evaluation and Research on New Policies to Advance Development of Safe and Effective Cell and Gene Therapies https://www.fda.gov/news-events/press-announcements/statement-fda-commissioner-scott-gottlieb-md-and-peter-marks-md-phd-director-center-biologics (2019).

  65. Medicines & Healthcare products Regulatory Agency (MHRA). Consultation on Point of Care Manufacturing https://www.gov.uk/government/consultations/point-of-care-consultation/consultation-on-point-of-care-manufacturing (2021).

  66. Nizzi, F. Redefining the role of blood establishments as raw material suppliers, manufacturers, and distributors for new cell therapies: the Blood Systems experience. Transfusion 56, 29S–31S (2016).

    PubMed  Google Scholar 

  67. Coppens, D. G. et al. Regulating advanced therapy medicinal products through the Hospital Exemption: an analysis of regulatory approaches in nine EU countries. Regen. Med. 15, 2015–2028 (2020).

    CAS  PubMed  Google Scholar 

  68. Coppens, D. G. M. et al. Advanced therapy medicinal product manufacturing under the hospital exemption and other exemption pathways in seven European Union countries. Cytotherapy 22, 592–600 (2020).

    PubMed  Google Scholar 

  69. Trias, E., Juan, M., Urbano-Ispizua, A. & Calvo, G. The hospital exemption pathway for the approval of advanced therapy medicinal products: an underused opportunity? The case of the CAR-T ARI-0001. Bone Marrow Transplant. 57, 156–159 (2022).

    PubMed  PubMed Central  Google Scholar 

  70. Ortíz-Maldonado, V. et al. CART19-BE-01: a multicenter trial of ARI-0001 cell therapy in patients with CD19+ relapsed/refractory malignancies. Mol. Ther. 29, 636–644 (2021).

    PubMed  Google Scholar 

  71. Therapeutic Goods Administration. Autologous Human Cells and Tissues Products Regulation https://www.tga.gov.au/resources/resource/guidance/autologous-human-cells-and-tissues-products-regulation (2019).

  72. Ivaskiene, T., Mauricas, M. & Ivaska, J. Hospital exemption for advanced therapy medicinal products: issue in application in the European Union Member States. Curr. Stem Cell Res. Ther. 12, 45–51 (2016).

    Google Scholar 

  73. Cuende, N. et al. The puzzling situation of hospital exemption for advanced therapy medicinal products in Europe and stakeholders’ concerns. Cytotherapy 16, 1597–1600 (2014).

    PubMed  Google Scholar 

  74. The Alliance for Regenerative Medicine. Recommendations for the Use of Hospital Exemption http://alliancerm.org/wp-content/uploads/2020/10/ARM-position-on-HE-final-Oct-2020.pdf (2020).

  75. EFPIA & EBE. Hospital exemption for advanced therapy medicinal products (ATMPs): greater transparency needed in order to improve patient safety and access to ATMPs. https://www.efpia.eu/news-events/the-efpia-view/statements-press-releases/10102017-ebe-and-efpia-call-on-the-eu-commission-and-member-states-to-improve-transparency-on-hospital-exemptions-for-advanced-therapies/ (2017).

  76. Cuende, N. et al. Patient access to and ethical considerations of the application of the European Union hospital exemption rule for advanced therapy medicinal products. Cytotherapy 24, 686–690 (2022).

    PubMed  Google Scholar 

  77. Warkentin, P. I. Voluntary accreditation of cellular therapies: Foundation for the Accreditation of Cellular Therapy (FACT). Cytotherapy 5, 299–305 (2003).

    CAS  PubMed  Google Scholar 

  78. Maus, M. V. & Nikiforow, S. The why, what, and how of the new FACT standards for immune effector cells. J. Immunother. Cancer 5, 36 (2017).

    PubMed  PubMed Central  Google Scholar 

  79. Hort, S. et al. Toward rapid, widely available autologous CAR-T cell therapy—artificial intelligence and automation enabling the smart manufacturing hospital. Front. Med. 9, 913287 (2022).

    Google Scholar 

  80. Blache, U., Popp, G., Dünkel, A., Koehl, U. & Fricke, S. Potential solutions for manufacture of CAR T cells in cancer immunotherapy. Nat. Commun. 13, 5225 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Soler, M. & Lechuga, L. Boosting cancer immunotherapies with optical biosensor nanotechnologies. Eur. Med. J. 4, 124–132 (2019).

    Google Scholar 

  82. Oh, B.-R. et al. Integrated nanoplasmonic sensing for cellular functional immunoanalysis using human blood. ACS Nano 8, 2667–2676 (2014).

    CAS  Google Scholar 

  83. Oh, B.-R. et al. Multiplexed nanoplasmonic temporal profiling of T-cell response under immunomodulatory agent exposure. ACS Sens. 1, 941–948 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Raphael, M. P., Christodoulides, J. A., Delehanty, J. B., Long, J. P. & Byers, J. M. Quantitative imaging of protein secretions from single cells in real time. Biophys. J. 105, 602–608 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Kurucz, I. et al. Label-free optical biosensor for on-line monitoring the integrated response of human B cells upon the engagement of stimulatory and inhibitory immune receptors. Sens. Actuators B Chem. 240, 528–535 (2017).

    CAS  Google Scholar 

  86. Soler, M. et al. Two-dimensional label-free affinity analysis of tumor-specific CD8 T cells with a biomimetic plasmonic sensor. ACS Sens. 3, 2286–2295 (2018).

    CAS  PubMed  Google Scholar 

  87. Walsh, A. J. et al. Classification of T-cell activation via autofluorescence lifetime imaging. Nat. Biomed. Eng. 5, 77–88 (2020).

    PubMed  PubMed Central  Google Scholar 

  88. Rossoff, J. et al. Out-of-specification tisagenlecleucel does not compromise safety or efficacy in pediatric acute lymphoblastic leukemia. Blood 138, 2138–2142 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Jacobson, C. A. et al. Outcomes of patients (Pts) in ZUMA-9, a multicenter, open-label study of axicabtagene ciloleucel (axi-cel) in relapsed/refractory large B cell lymphoma (R/R LBCL) for expanded access and commercial out-of-specification (OOS) product. Blood 136, 2–3 (2020).

    Google Scholar 

  90. Chong, E. A. et al. CAR T cell viability release testing and clinical outcomes: is there a lower limit? Blood 134, 1873–1875 (2019).

    PubMed  PubMed Central  Google Scholar 

  91. U.S. Food & Drug Administration (FDA). Approval Order—CliniMACS CD34 Reagent System https://wayback.archive-it.org/7993/20190208123839/https://www.fda.gov/BiologicsBloodVaccines/BloodBloodProducts/ApprovedProducts/PremarketApprovalsPMAs/ucm382986.htm (2014).

  92. U.S. Food & Drug Administration (FDA). Getting a Humanitarian Use Device to Market https://www.fda.gov/medical-devices/humanitarian-device-exemption/getting-humanitarian-use-device-market (2022).

  93. Priesner, C. et al. Automated enrichment, transduction, and expansion of clinical-scale CD62L+ T cells for manufacturing of gene therapy medicinal products. Hum. Gene Ther. 27, 860–869 (2016).

    CAS  Google Scholar 

  94. Lock, D. et al. Automated manufacturing of potent CD20-directed chimeric antigen receptor T cells for clinical use. Hum. Gene Ther. 28, 914–925 (2017).

    CAS  Google Scholar 

  95. Blaeschke, F. et al. Induction of a central memory and stem cell memory phenotype in functionally active CD4+ and CD8+ CAR T cells produced in an automated good manufacturing practice system for the treatment of CD19+ acute lymphoblastic leukemia. Cancer Immunol. Immunother. 67, 1053–1066 (2018).

    CAS  PubMed  Google Scholar 

  96. Castella, M. et al. Development of a novel anti-CD19 chimeric antigen receptor: a paradigm for an affordable CAR T cell production at academic institutions. Mol. Ther. Methods Clin. Dev. 12, 134–144 (2019).

    CAS  PubMed  Google Scholar 

  97. Zhang, W., Jordan, K. R., Schulte, B. & Purev, E. Characterization of clinical grade CD19 chimeric antigen receptor T cells produced using automated CliniMACS Prodigy system. Drug Des. Devel. Ther. 12, 3343–3356 (2018).

    CAS  Google Scholar 

  98. Aleksandrova, K. et al. Functionality and cell senescence of CD4/ CD8-selected CD20 CAR T cells manufactured using the automated CliniMACS Prodigy® platform. Transfus. Med. Hemother. 46, 47–54 (2019).

    Google Scholar 

  99. Fernández, L. et al. GMP-compliant manufacturing of NKG2D CAR memory T cells using CliniMACS Prodigy. Front. Immunol. 10, 2361 (2019).

    PubMed  PubMed Central  Google Scholar 

  100. Vedvyas, Y. et al. Manufacturing and preclinical validation of CAR T cells targeting ICAM-1 for advanced thyroid cancer therapy. Sci. Rep. 9, 10634 (2019).

    PubMed  PubMed Central  Google Scholar 

  101. Arcangeli, S. et al. Next-generation manufacturing protocols enriching TSCM CAR T cells can overcome disease-specific T cell defects in cancer patients. Front. Immunol. 11, 1217 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Bozza, M. et al. A nonviral, nonintegrating DNA nanovector platform for the safe, rapid, and persistent manufacture of recombinant T cells. Sci. Adv. 7, eabf1333 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Palen, K., Zurko, J., Johnson, B. D., Hari, P. & Shah, N. N. Manufacturing chimeric antigen receptor T cells from cryopreserved peripheral blood cells: time for a collect-and-freeze model? Cytotherapy 23, 985–990 (2021).

    CAS  PubMed  Google Scholar 

  104. Glienke, W. et al. GMP-compliant manufacturing of TRUCKs: CAR T cells targeting GD2 and releasing inducible IL-18. Front. Immunol. 13, 839783 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Joedicke, J. J. et al. Accelerating clinical-scale production of BCMA CAR T cells with defined maturation stages. Mol. Ther. Methods Clin. Dev. 24, 181–198 (2022).

    CAS  PubMed  Google Scholar 

  106. Nicod, C. et al. CAR-T cells targeting IL-1RAP produced in a closed semiautomatic system are ready for the first phase I clinical investigation in humans. Curr. Res. Transl. Med. 71, 103385 (2023).

    CAS  PubMed  Google Scholar 

  107. Maschan, M. et al. Multiple site place-of-care manufactured anti-CD19 CAR-T cells induce high remission rates in B-cell malignancy patients. Nat. Commun. 12, 7200 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Marcela V. Maus.

Ethics declarations

Competing interests

M.V.M. is an inventor on patents related to adoptive cell therapies, held by Massachusetts General Hospital (some licensed to ProMab) and the University of Pennsylvania (some licensed to Novartis). M.V.M. holds equity in 2Seventy Bio, Century Therapeutics, NexImmune, Oncternal and TCR2 and has served as a consultant for multiple companies involved in cell therapies. M.V.M. is on the board of directors of 2Seventy Bio. M.V.M. has received grant and/or research support from CRISPR Therapeutics, Kite Pharma, Servier and Novartis. M.E. declares no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Mark Lowdell, Ulrike Koehl and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elsallab, M., Maus, M.V. Expanding access to CAR T cell therapies through local manufacturing. Nat Biotechnol 41, 1698–1708 (2023). https://doi.org/10.1038/s41587-023-01981-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-023-01981-8

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