Point-Of-Care CAR T-Cell Production (ARI-0001) Using a Closed Semi-automatic Bioreactor: Experience From an Academic Phase I Clinical Trial

Maria Castella, Miguel Caballero-Baños, Valentín Ortiz-Maldonado, Europa Azucena González-Navarro, Guillermo Suñé, Asier Antoñana-Vidósola, Anna Boronat, Berta Marzal, Lucía Millán, Beatriz Martín-Antonio, Joan Cid, Miquel Lozano, Enric García, Jaime Tabera, Esteve Trias, Unai Perpiña, Josep Ma Canals, Tycho Baumann, Daniel Benítez-Ribas, Elías Campo, Jordi Yagüe, Álvaro Urbano-Ispizua, Susana Rives, Julio Delgado, Manel Juan, Maria Castella, Miguel Caballero-Baños, Valentín Ortiz-Maldonado, Europa Azucena González-Navarro, Guillermo Suñé, Asier Antoñana-Vidósola, Anna Boronat, Berta Marzal, Lucía Millán, Beatriz Martín-Antonio, Joan Cid, Miquel Lozano, Enric García, Jaime Tabera, Esteve Trias, Unai Perpiña, Josep Ma Canals, Tycho Baumann, Daniel Benítez-Ribas, Elías Campo, Jordi Yagüe, Álvaro Urbano-Ispizua, Susana Rives, Julio Delgado, Manel Juan

Abstract

Development of semi-automated devices that can reduce the hands-on time and standardize the production of clinical-grade CAR T-cells, such as CliniMACS Prodigy from Miltenyi, is key to facilitate the development of CAR T-cell therapies, especially in academic institutions. However, the feasibility of manufacturing CAR T-cell products from heavily pre-treated patients with this system has not been demonstrated yet. Here we report and characterize the production of 28 CAR T-cell products in the context of a phase I clinical trial for CD19+ B-cell malignancies (NCT03144583). The system includes CD4-CD8 cell selection, lentiviral transduction and T-cell expansion using IL-7/IL-15. Twenty-seven out of 28 CAR T-cell products manufactured met the full list of specifications and were considered valid products. Ex vivo cell expansion lasted an average of 8.5 days and had a mean transduction rate of 30.6 ± 13.44%. All products obtained presented cytotoxic activity against CD19+ cells and were proficient in the secretion of pro-inflammatory cytokines. Expansion kinetics was slower in patient's cells compared to healthy donor's cells. However, product potency was comparable. CAR T-cell subset phenotype was highly variable among patients and largely determined by the initial product. TCM and TEM were the predominant T-cell phenotypes obtained. 38.7% of CAR T-cells obtained presented a TN or TCM phenotype, in average, which are the subsets capable of establishing a long-lasting T-cell memory in patients. An in-depth analysis to identify individual factors contributing to the optimal T-cell phenotype revealed that ex vivo cell expansion leads to reduced numbers of TN, TSCM, and TEFF cells, while TCM cells increase, both due to cell expansion and CAR-expression. Overall, our results show for the first time that clinical-grade production of CAR T-cells for heavily pre-treated patients using CliniMACS Prodigy system is feasible, and that the obtained products meet the current quality standards of the field. Reduced ex vivo expansion may yield CAR T-cell products with increased persistence in vivo.

Keywords: CAR T-cell production; CD19; CliniMACS Prodigy; chimeric antigen receptor; immunotherapy; leukemia; lymphoma.

Copyright © 2020 Castella, Caballero-Baños, Ortiz-Maldonado, González-Navarro, Suñé, Antoñana-Vidósola, Boronat, Marzal, Millán, Martín-Antonio, Cid, Lozano, García, Tabera, Trias, Perpiña, Canals, Baumann, Benítez-Ribas, Campo, Yagüe, Urbano-Ispizua, Rives, Delgado and Juan.

Figures

Figure 1
Figure 1
ARI-0001-cell expansion in CliniMACS Prodigy. (A) Expansion kinetics of ARI-0001-cell products (Total cell number). Gray points indicate individual products. Black triangles indicate Mean ± SD and adjusting curve. (B) Expansion kinetics of CAR19+ cells (red) and total cell number (black). Mean ± SD is represented. (C) Expansion kinetics of ARI-0001 cells (Total cell number) comparing healthy controls and different types of disease. Mean ± SEM is represented. (D) Percentage of CD3 and CAR19 positive cells as determined by flow cytometry. Mean ± SD is also indicated. Panels on the right show flow cytometry representative image corresponding to CAR19 and CD3 staining in ARI-0001-cell final products and Control T cells (Untransduced).
Figure 2
Figure 2
ARI-0001-cell potency. (A) Cytotoxicity assay after 4 h of ARI-0001 co-culture with NALM6 cells, at the indicated ratios. Mean ± SD of all 27 CAR T cell products is indicated. (#) Dashed line indicates minimum of ARI-0001-cell cytotoxicity level for a product to be considered valid. (B) IFNγ, TNFα, and GranzymeB levels measured in the supernatants of the cytotoxicity assays. E:T ratio 0 indicates no target cells. (*) indicates statistical significance, p < 0.05. (C) Comparison of ARI-0001 cytotoxic potential after 4 h of co-culture with NALM6 cells, at the indicated ratios. Mean ± SD is shown. “n.s.” indicates not statistically significant (Non parametric test). (D) Comparison of IFNγ, TNFα, and GranzymeB levels measured in the supernatants of the cytotoxicity assay at E:T ratio 1:1. “HD” indicates Healthy donors. “n.s.” indicates not statistically significant (Parametric test applied to IFNγ and TNFα and non-parametric test applied to GranzymeB).
Figure 3
Figure 3
ARI-0001 cell subset characterization. (A) CD4/CD8 ratio of apheresis products, after CD4-CD8 cell selection and of the final product. (B) CD4/CD8 ratio variation during cell expansion. Left panel corresponds to products with an initial ratio < 1. Right panel corresponds to products with an initial ratio > 1. (C) CAR19 transduction efficiency in CD4 and CD8 cells. Mean ± SD is shown. (D) Percentage of T-cell subpopulations within initial (CD4-CD8 cell selection) and final products (CAR– and CAR+ cells). (E) Representative flow cytometry plots of three different patients showing T cell populations in initial and final products. (F) Differences in MFI for CD45RA and CCR7 in initial and final products. Lower panel shows paired analysis for CCR7 MFI. (*) indicates statistical significance, p < 0.05. n.s. indicates not statistically significant.

References

    1. Gill S, June CH. Going viral: chimeric antigen receptor T-cell therapy for hematological malignancies. Immunol Rev. (2015) 263:68–89. 10.1111/imr.12243
    1. Barrett DM, Singh N, Porter DL, Grupp SA, June CH. Chimeric antigen receptor therapy for cancer. Annu Rev Med. (2014) 65:333–47. 10.1146/annurev-med-060512-150254
    1. Dai H, Wang Y, Lu X, Han W. Chimeric antigen receptors modified T-cells for cancer therapy. JNCI J Natl Cancer Inst. (2016) 108:djv439. 10.1093/jnci/djv439
    1. Sadelain M. CAR therapy: the CD19 paradigm. J Clin Invest. (2015) 125:3392–400. 10.1172/JCI80010
    1. Jensen MC, Riddell SR. Design and implementation of adoptive therapy with chimeric antigen receptor-modified T cells. Immunol Rev. (2014) 257:127–44. 10.1111/imr.12139
    1. Chang ZL, Chen YY. CARs: synthetic immunoreceptors for cancer therapy and beyond. Trends Mol Med. (2017) 23:430–50. 10.1016/j.molmed.2017.03.002
    1. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, et al. . Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. (2010) 116:4099–102. 10.1182/blood-2010-04-281931
    1. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. (2011) 365:725–33. 10.1056/NEJMoa1103849
    1. Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, et al. . Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL. Blood. (2016) 127:2980–90. 10.1182/blood-2015-12-686725
    1. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. . Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. (2014) 371:1507–17. 10.1056/NEJMoa1407222
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. . T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. (2015) 385:517–28. 10.1016/S0140-6736(14)61403-3
    1. Brentjens RJ, Riviere I, Park JH, Davila ML, Wang X, Stefanski J, et al. . Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood. (2011) 118:4817–28. 10.1182/blood-2011-04-348540
    1. Kochenderfer JN, Dudley ME, Carpenter RO, Kassim SH, Rose JJ, Telford WG, et al. . Donor-derived CD19-targeted T cells cause regression of malignancy persisting after allogeneic hematopoietic stem cell transplantation. Blood. (2013) 122:4129–39. 10.1182/blood-2013-08-519413
    1. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RPT, Carpenter RO, Stetler-Stevenson M, et al. . Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol. (2015) 33:540–9. 10.1200/JCO.2014.56.2025
    1. Brudno JN, Maric I, Hartman SD, Rose JJ, Wang M, Lam N, et al. . T cells genetically modified to express an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma. J Clin Oncol. (2018) 36:2267–80. 10.1200/JCO.2018.77.8084
    1. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, et al. . T cells expressing an anti-B-cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. (2016) 128:1688–700. 10.1182/blood-2016-04-711903
    1. Zhao W-H, Liu J, Wang B-Y, Chen Y-X, Cao X-M, Yang Y, et al. . A phase 1, open-label study of LCAR-B38M, a chimeric antigen receptor T cell therapy directed against B cell maturation antigen, in patients with relapsed or refractory multiple myeloma. J Hematol Oncol. (2018) 11:141. 10.1186/s13045-018-0681-6
    1. Ramos CA, Ballard B, Zhang H, Dakhova O, Gee AP, Mei Z, et al. . Clinical and immunological responses after CD30-specific chimeric antigen receptor-redirected lymphocytes. J Clin Invest. (2017) 127:3462–71. 10.1172/JCI94306
    1. Wang C-M, Wu Z-Q, Wang Y, Guo Y-L, Dai H-R, Wang X-H, et al. . Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res. (2017) 23:1156–66. 10.1158/1078-0432.CCR-16-1365
    1. Fry TJ, Shah NN, Orentas RJ, Stetler-Stevenson M, Yuan CM, Ramakrishna S, et al. . CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med. (2017) 24:20–8. 10.1038/nm.4441
    1. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. . A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res. (2006) 12:6106–15. 10.1158/1078-0432.CCR-06-1183
    1. O'Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, et al. . A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. (2017) 9:eaaa0984. 10.1126/scitranslmed.aaa0984
    1. Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, et al. . Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric antigen receptor-modified T-cell therapy for CEA+ liver metastases. Clin Cancer Res. (2015) 21:3149–59. 10.1158/1078-0432.CCR-14-1421
    1. Beatty GL, O'Hara MH, Lacey SF, Torigian DA, Nazimuddin F, Chen F, et al. . Activity of mesothelin-specific chimeric antigen receptor T cells against pancreatic carcinoma metastases in a phase 1 trial. Gastroenterology. (2018) 155:29–32. 10.1053/j.gastro.2018.03.029
    1. Ahmed N, Brawley VS, Hegde M, Robertson C, Ghazi A, Gerken C, et al. . Human epidermal growth factor receptor 2 (HER2) -specific chimeric antigen receptor-modified T cells for the immunotherapy of HER2-positive sarcoma. J Clin Oncol. (2015) 33:1688–96. 10.1200/JCO.2014.58.0225
    1. Thistlethwaite FC, Gilham DE, Guest RD, Rothwell DG, Pillai M, Burt DJ, et al. . The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol Immunother. (2017) 66:1425–36. 10.1007/s00262-017-2034-7
    1. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. (2010) 18:843–51. 10.1038/mt.2010.24
    1. Lamers CH, Sleijfer S, van Steenbergen S, van Elzakker P, van Krimpen B, Groot C, et al. . Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol Ther. (2013) 21:904–12. 10.1038/mt.2013.17
    1. Lamers CHJ, Sleijfer S, Vulto AG, Kruit WHJ, Kliffen M, Debets R, et al. . Treatment of metastatic renal cell carcinoma with autologous T-lymphocytes genetically retargeted against carbonic anhydrase IX: first clinical experience. J Clin Oncol. (2006) 24:e20–2. 10.1200/JCO.2006.05.9964
    1. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. . Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N Engl J Med. (2016) 375:2561–9. 10.1056/NEJMoa1610497
    1. Knochelmann HM, Smith AS, Dwyer CJ, Wyatt MM, Mehrotra S, Paulos CM. CAR T cells in solid tumors: blueprints for building effective therapies. Front Immunol. (2018) 9:1740. 10.3389/fimmu.2018.01740
    1. Long KB, Young RM, Boesteanu AC, Davis MM, Melenhorst JJ, Lacey SF, et al. . CAR T cell therapy of non-hematopoietic malignancies: detours on the road to clinical success. Front Immunol. (2018) 9:2740. 10.3389/fimmu.2018.02740
    1. Hoyos V, Savoldo B, Quintarelli C, Mahendravada A, Zhang M, Vera J, et al. . Engineering CD19-specific T lymphocytes with interleukin-15 and a suicide gene to enhance their anti-lymphoma/leukemia effects and safety. Leukemia. (2010) 24:1160–70. 10.1038/leu.2010.75
    1. Chmielewski M, Abken H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. (2017) 21:3205–19. 10.1016/j.celrep.2017.11.063
    1. Krenciute G, Prinzing BL, Yi Z, Wu M-F, Liu H, Dotti G, et al. . Gottschalk S. Transgenic expression of IL15 improves antiglioma activity of IL13Rα2-CAR T cells but results in antigen loss variants. Cancer Immunol Res. (2017) 5:571–81. 10.1158/2326-6066.CIR-16-0376
    1. Chmielewski M, Abken H. CAR T cells transform to trucks: chimeric antigen receptor-redirected T cells engineered to deliver inducible IL-12 modulate the tumour stroma to combat cancer. Cancer Immunol Immunother. (2012) 61:1269–77. 10.1007/s00262-012-1202-z
    1. Castella M, Boronat A, Martín-Ibáñez R, Rodríguez V, Suñé G, Caballero 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. (2019) 12:134–44. 10.1016/j.omtm.2018.11.010
    1. Sommermeyer D, Hudecek M, Kosasih PL, Gogishvili T, Maloney DG, Turtle CJ, et al. . Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. (2016) 30:492–500. 10.1038/leu.2015.247
    1. Kwajah MMS, Schwarz H. CD137 ligand signaling induces human monocyte to dendritic cell differentiation. Eur J Immunol. (2010) 40:1938–49. 10.1002/eji.200940105
    1. Kawalekar OU, O'Connor RS, Fraietta JA, Guo L, McGettigan SE, Posey AD, et al. . Distinct signaling of coreceptors regulates specific metabolism pathways and impacts memory development in CAR T cells. Immunity. (2016) 44:380–90. 10.1016/j.immuni.2016.01.021
    1. Aleksandrova K, Leise J, Priesner C, Melk A, Kubaink F, Abken H, et al. . Functionality and cell senescence of CD4/ CD8-selected CD20 CAR T cells manufactured using the automated CliniMACS Prodigy® platform. Transfus Med Hemotherapy. (2019) 46:47–54. 10.1159/000495772
    1. Marín Morales JM, Münch N, Peter K, Freund D, Oelschlägel U, Hölig K, et al. . Automated clinical grade expansion of regulatory T cells in a fully closed system. Front Immunol. (2019) 10:38. 10.3389/fimmu.2019.00038
    1. Wang X, Rivière I. Clinical manufacturing of CAR T cells: foundation of a promising therapy. Mol Ther Oncolytics. (2016) 3:16015. 10.1038/mto.2016.15
    1. Zhang W, Jordan KR, Schulte B, Purev E. Characterization of clinical grade CD19 chimeric antigen receptor T cells produced using automated CliniMACS prodigy system. Drug Des Devel Ther. (2018) 12:3343–56. 10.2147/DDDT.S175113
    1. Priesner C, Aleksandrova K, Esser R, Mockel-Tenbrinck N, Leise J, Drechsel K, et al. . Automated enrichment, transduction, and expansion of clinical-scale CD62L+ T cells for manufacturing of gene therapy medicinal products. Hum Gene Ther. (2016) 27:860–9. 10.1089/hum.2016.091
    1. Mackall CL, Fleisher TA, Brown MR, Andrich MP, Chen CC, Feuerstein IM, et al. . Distinctions between CD8+ and CD4+ T-cell regenerative pathways result in prolonged T-cell subset imbalance after intensive chemotherapy. Blood. (1997) 89:3700–7. 10.1182/blood.V89.10.3700.3700_3700_3707
    1. Mackall C, Fleisher T, Brown M, Magrath I, Shad A, Horowitz M, et al. . Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood. (1994) 84:2221–8. 10.1182/blood.V84.7.2221.2221
    1. Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, et al. . Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. (2016) 127:1117–27. 10.1182/blood-2015-11-679134
    1. Porter DL, Hwang W-T, Frey N V, Lacey SF, Shaw PA, Loren AW, et al. . Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. (2015) 7:303ra139. 10.1126/scitranslmed.aac5415
    1. Zhu F, Shah NN, Xu H, Schneider D, Orentas R, Dropulic B, et al. CAR-T cell production using the Clinimacs® Prodigy system. Blood. (2016) 128:5724 10.1182/blood.V128.22.5724.5724
    1. Mock U, Nickolay L, Philip B, Cheung GWK, Zhan H, Johnston ICD, et al. . Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy. (2016) 18:1002–11. 10.1016/j.jcyt.2016.05.009
    1. Blaeschke F, Stenger D, Kaeuferle T, Willier S, Lotfi R, Kaiser AD, 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. (2018) 67:1053–66. 10.1007/s00262-018-2155-7
    1. Blaeschke F, Kaeuferle T, Feucht J, Weber D, Lotfi R, Kaiser A, et al. Defined central memory and stem memory T cell phenotype of CD4 and CD8 CAR T cells for the treatment of CD19+ acute lymphoblastic leukemia in an automated closed system. Blood. (2016) 128:1053–66. 10.1182/blood.V128.22.4558.4558
    1. Hollyman D, Stefanski J, Przybylowski M, Bartido S, Borquez-Ojeda O, Taylor C, et al. . Manufacturing validation of biologically functional T cells targeted to CD19 antigen for autologous adoptive cell therapy. J Immunother. (2009) 32:169–80. 10.1097/CJI.0b013e318194a6e8
    1. Gomes-Silva D, Mukherjee M, Srinivasan M, Krenciute G, Dakhova O, Zheng Y, et al. . Tonic 4-1BB costimulation in chimeric antigen receptors impedes T cell survival and is vector-dependent. Cell Rep. (2017) 21:17–26. 10.1016/j.celrep.2017.09.015
    1. Gattinoni L, Lugli E, Ji Y, Pos Z, Paulos CM, Quigley MF, et al. . A human memory T cell subset with stem cell-like properties. Nat Med. (2011) 17:1290–7. 10.1038/nm.2446
    1. Hinrichs CS, Borman ZA, Gattinoni L, Yu Z, Burns WR, Huang J, et al. . Human effector CD8+ T cells derived from naive rather than memory subsets possess superior traits for adoptive immunotherapy. Blood. (2011) 117:808–14. 10.1182/blood-2010-05-286286
    1. Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. (2008) 118:294–305. 10.1172/JCI32103
    1. Larbi A, Fulop T. From “truly naïve” to “exhausted senescent” T cells: when markers predict functionality. Cytom Part A. (2014) 85:25–35. 10.1002/cyto.a.22351
    1. Leitenberg D, Novak TJ, Farber D, Smith BR, Bottomly K. The extracellular domain of CD45 controls association with the CD4-T cell receptor complex and the response to antigen-specific stimulation. J Exp Med. (1996) 183:249–59. 10.1084/jem.183.1.249

Source: PubMed

3
Se inscrever