Potent ex vivo armed T cells using recombinant bispecific antibodies for adoptive immunotherapy with reduced cytokine release

Jeong A Park, Brian H Santich, Hong Xu, Lawrence G Lum, Nai-Kong V Cheung, Jeong A Park, Brian H Santich, Hong Xu, Lawrence G Lum, Nai-Kong V Cheung

Abstract

Background: T cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative.

Methods: We generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells.

Results: The antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αβ T cell-EATs.

Conclusions: EATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.

Trial registration: ClinicalTrials.gov NCT02173093 NCT00027807 NCT04137536.

Keywords: adoptive; immunotherapy; investigational; neuroblastoma; sarcoma; therapies; translational medical research.

Conflict of interest statement

Competing interests: Both MSK and N-KVC have financial interest in Y-mAbs, Abpro-Labs and Eureka Therapeutics. N-KVC reports receiving commercial research grants from Y-mAbs Therapeutics and Abpro-Labs. N-KVC was named as inventor on multiple patents filed by MSK, including those licensed to Y-mAbs Therapeutics, Biotec Pharmacon, and Abpro-Labs. N-KVC is a SAB member for Abpro-Labs and Eureka Therapeutics. LL is cofounder of Transtarget and is a SAB member for Rapa Therapeutics. JAP has no disclosures to report.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Bispecific antibody platform has profound effects on the antitumor activity of bispecific antibody armed T cell immunotherapy. (A) BsAb structural platform. (B) BsAb armed T cells were stained with anti-idiotype antibody, and surface BsAb densities were measured as MFI by flow cytometry. (C) Antibody-dependent T cell-mediated cytotoxicity assay of GD2-BsAb armed T cells at increasing BsAb arming doses. The effector to target cell ratio (E:T ratio) was 10:1. (D) 2×107 of T cells were armed with 2 µg of each BsAb and administered intravenously two doses per week for 2–3 weeks in GD2(+) osteosarcoma PDX and GD2(+) neuroblastoma PDX mouse models. In vivo antitumor responses were compared among groups. (E) Immunohistochemical staining of CD3(+) tumor infiltrating lymphocytes in neuroblastoma PDX tumors treated with a variety of GD2-BsAb armed T cells (on day 10 after the beginning of treatment). (F) Surface BsAb density (MFI) of HER2-BsAb armed T cells. (G) HER2-BsAb armed T cell induced cytotoxicity against osteosarcoma cell line U-2 OS at an E:T ratio of 10:1. (H) HER2-BsAb armed T cells (2 µg of HER2-BsAb/2×107 of T cells) were administered intravenously twice per week for 3 weeks in HER2(+) osteosarcoma PDX-bearing mice. In vivo antitumor effect was compared. **P<0.01, ***P<0.001, ****P<0.0001. BiTE, Bi-specific T-cell engagers; BsAb, bispecific antibodies; GD2, disialoganglioside; HER2, human epidermal growth factor receptor 2; MFI, mean fluorescence intensity; PDX, patient-derived tumor xenograft; scFv, single-chain variable fragment.
Figure 2
Figure 2
Ex vivo arming of T cells with IgG-[L]-scFv-platformed BsAb. (A) Antibody-dependent T cell-mediated cytotoxicity assay of GD2-EATs and HER2-EATs at increasing effector to target ratios (E:T ratios) and at increasing BsAb doses. (B) Surface BsAb density on EAT was analyzed using anti-human IgG Fc-specific antibody and anti-mouse quantum beads. Geometric MFIs of GD2-EATs and HER2-EATs were measured with increasing arming dose of either GD2-BsAb or HER2-BsAb, and BsAb density (MFI) on EAT was referenced to antibody binding capacity (ABC). (C) Comparison of in vitro cytotoxicity between EATs and soluble BsAb with identical E:T ratios, for both anti-GD2 and anti-HER2 systems. BsAb, bispecific antibodies; EATs, ex vivo armed T cells; GD2, disialoganglioside; HER2, human epidermal growth factor receptor 2; MFI, mean fluorescence intensities.
Figure 3
Figure 3
Ex vivo arming of T cells could significantly reduce cytokine release. (A) Th1 cell cytokines (IL-2, IL-6, IL-10, IFN-γ, and TNF-α) released by T cells were measured in each step of arming: (a) in the supernatants after 20 min of incubation with GD2-BsAb (prewash), (b) after the second washing step (postwash), and (c and d) after incubation with target cells (GD2(+) M14 melanoma cell line) at 37℃ for 4 hours. (B) Cytokine release was compared between prewash supernatants (a) and postwash supernatants (b). (C) Cytokine release was compared between GD2-BsAb plus T cells (c), GD2-EATs (d) and unarmed T cells after exposure to target antigen. (D) After intravenous injection of GD2-EATs (armed with 10 µg of GD2-BsAb/2×107 cells) or GD2-BsAb (10 µg) plus unarmed T cells (2×107 cells) into osteosarcoma PDX-bearing mice, serum Th1 cytokine levels were measured at different time points. *P<0.05, **P<0.01, ***P<0.001, and ****P<0.0001. BsAb, bispecific antibody; EATs, ex vivo armed T cells; GD2, disialoganglioside; IFN, interferon; IL, interleukin; PBS, phosphate-buffered saline; PDX, patient-derived tumor xenograft; Th1, T helper 1; TNF, tumor necrosis factor.
Figure 4
Figure 4
EATs showed faster tumor homing kinetics than BsAb-directed unarmed T cells, rapidly bypassing lung sequestration. Luciferase transduced T cells were expanded and armed with BsAb (Luc(+) GD2-EATs). Luc(+) GD2-EATs (10 µg of GD2-BsAb/2×107 T cells) or Luc(+) unarmed T cells (2×107 cells) with or without GD2-BsAb (10 µg) were administered intravenously into GD2(+) neuroblastoma PDX mice when the average tumor volume reached 100 mm3. (A) Bioluminescence images of GD2-EATs trafficking into tumors over days. (B) Bioluminescence images of GD2-BsAb directed unarmed T cell trafficking into tumors over days. (C) Quantitation of T cell infiltration into tumors over time measured by bioluminescence (n=5 mice/group) expressed as total flux or radiance (photons/s) per pixel integrated over the tumor contour (ROI). (D) Tumor growth curves of individual mice treated with GD2-EATs, GD2-BsAb plus unarmed T cells, or unarmed T cells. To test the in vivo persistence of target antigen-specific EATs, Luc(+) GD2-EATs (armed with 10 µg of GD2-BsAb/2×107 T cells) or Luc(+) HER2-EATs (10 µg of HER2-BsAb/2×107 T cells) were intravenously administered into osteosarcoma TEOS1C PDX-bearing mice. Additional two doses of non-luciferase transduced GD2-EATs or HER2-EATs were administered on day 7 and day 14. (E) Quantitation of bioluminescence of Luc(+) EATs in tumors post-treatment. (F) Tumor growth curves of individual mice treated with GD2-EATs, HER2-EATs, or unarmed T cells, or no treatment. (G) Bioluminescence images of Luc(+) GD2-EATs (upper) or Luc(+) HER2-EATs (lower) in tumors over time. Bioluminescence of Luc(+) EATs was detected over 28 days postinjection. BsAb, bispecific antibodies; EATs, ex vivo armed T cells; GD2, disialoganglioside; HER2, human epidermal growth factor receptor 2; PDX, patient-derived tumor xenograft; ROI, region of interest.
Figure 5
Figure 5
Factors determining the in vivo efficacy of EATs. (A) The effect of BsAb dose on in vivo efficacy of EATs. 2×107 of T cells were armed with increasing dose of GD2-BsAb or HER2-BsAb and their antitumor effects were compared. (B) The effect of EAT cell number on the in vivo efficacy of EATs. At a fixed arming dose of 0.5 µg of BsAb/1×106 cells, increasing numbers of GD2-EAT or HER2-EAT (5×106 cells, 1×107 cells, and 2×107 cells) were administered. (C) The effect of EAT treatment schedule on the in vivo efficacy of EATs. GD2-EATs (2 µg of GD2-BsAb/2×107 cells) were administered as low intensity (1 dose/week in arm 1), standard (2 doses/week in arm 2), or dose-dense (3 doses/week in arm 3) and their in vivo antitumor response and survival were compared. (D) The effect of total number of doses of EATs on long-term tumor control and survival. Arm 1, GD2-EATs (10 µg of GD2-BsAb/2×107 cells) were administered twice weekly for 1 week, and GD2-BsAb (10 µg/dose) were followed twice weekly for 3 weeks; arm 2, four doses of GD2-EATs followed by four doses of GD2-BsAb; arm 3, eight doses of GD2-EATs. *P<0.05, **P<0.01, ***P<0.001, and ****P<0.0001. BsAb, bispecific antibody; EATs, ex vivo armed T cells; GD2, disialoganglioside; HER2, human epidermal growth factor receptor 2; PDX, patient-derived tumor xenograft; sc, subcutaneous.
Figure 6
Figure 6
Cryopreserved EATs retained target antigen-specific cytotoxicity and exerted a comparable antitumor activity. (A) Geometric mean fluorescence intensities of BsAb on GD2-EATs (0.5 µg of GD2-BsAb/106 cells) and HER2-EATs (0.5 µg of HER2-BsAb/106 cells) before and after cryopreservation. (B) Antibody-dependent T cell-mediated cytotoxicity assay of GD2-EATs and HER2-EATs against GD2(+) and HER2(+) osteosarcoma cell lines before and after cryopreservation (n=6). (C) In vivo antitumor response of thawed GD2-EATs. T cells were first armed with GD2-BsAb (10 µg of BsAb/2×107 cells) before cryopreservation. These cells were then thawed and administered into mice bearing GD2(+) osteosarcoma PDX to compare their in vivo antitumor potency with freshly armed GD2-EATs derived from the same donor. Four to six doses of cryopreserved (thawed) GD2-EATs or freshly armed (fresh) GD2-EATs were administered intravenously twice per week. (D) Relative body weights over time after treatment with fresh or thawed GD2-EATs. (E) Flow cytometry analyses of peripheral blood (PB) T cells in the mice treated with fresh or thawed GD2-EATs on day 35 after treatment. (F) In vivo antitumor response of thawed GD2-EATs or HER2-EATs against telangiectatic osteosarcoma PDXs. T cells were first armed with GD2-BsAb or HER2-BsAb (10 µg of BsAb/2×107 cells) before cryopreservation. Six doses of GD2-EATs or HER2-EATs were given twice a week for 3 weeks with SC interleukin-2. Both thawed EATs significantly suppressed tumor growth without weight loss, improving survival (p<0.0001). ***P<0.001, ****P<0.0001. ATC, activated T cells; EATs, ex vivo armed T cells; E:T, effector to target cell ratio; GD2, disialoganglioside; HER2, human epidermal growth factor receptor 2; PDX, patient-derived tumor xenograft.
Figure 7
Figure 7
EAT as a versatile platform to harness γδ T cells. (A) Flow cytometry of GD2-BsAb or HER2-BsAb on γδ T cells (γδTs) or unselected polyclonal T cells (mostly αβ T cells (αβTs)) after arming with GD2-BsAb or HER2-BsAb. (B) ADTC assays of γδ-GD2-EATs and γδ-HER2-EATs compared with αβ-GD2-EATs and αβ-HER2-EATs. Non-specific tumor cell killing by unarmed γδ T cells and unarmed αβ T cells (background) were subtracted. (C) ADTC assay of γδ-GD2-EATs and γδ-HER2-EATs at increasing effector to target ratios (E:T ratios) and at increasing BsAb arming doses. (D) In vitro cytotoxicity against breast cancer cell line HCC1954 was compared between fresh γδ-HER2-EATs and thawed γδ-HER2-EATs at increasing E:T ratios and at increasing BsAb arming doses. (E) Flow cytometry analyses of peripheral blood T cells after second dose of γδ-GD2-EATs plus zoledronate and supplementary IL-2 or IL-15. (F) Three doses of γδ-GD2-EATs (10 µg of GD2-BsAb/2×107 cells) were administered with supplementary IL-2 or IL-15 to treat osteosarcoma PDXs and the antitumor response was compared among groups. *P<0.05, ***P<0.001. ADTC, antibody-dependent T cell-mediated cytotoxicity; BsAb, bispecific antibodies; EATs, ex vivo armed T cells; γδ T cells, gamma delta T cells; IL, interleukin; MFI, mean fluorescence intensity; PDX, patient-derived tumor xenograft.

References

    1. Park JH, Rivière I, Gonen M, et al. . Long-Term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018;378:449–59. 10.1056/NEJMoa1709919
    1. Topalian SL, Hodi FS, Brahmer JR, et al. . Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443–54. 10.1056/NEJMoa1200690
    1. Topp MS, Gökbuget N, Zugmaier G, et al. . Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol 2014;32:4134–40. 10.1200/JCO.2014.56.3247
    1. Velasquez MP, Bonifant CL, Gottschalk S. Redirecting T cells to hematological malignancies with bispecific antibodies. Blood 2018;131:30–8. 10.1182/blood-2017-06-741058
    1. Beatty GL, O'Hara M. Chimeric antigen receptor-modified T cells for the treatment of solid tumors: defining the challenges and next steps. Pharmacol Ther 2016;166:30–9. 10.1016/j.pharmthera.2016.06.010
    1. Gajewski TF. Failure at the effector phase: immune barriers at the level of the melanoma tumor microenvironment. Clin Cancer Res 2007;13:5256–61. 10.1158/1078-0432.CCR-07-0892
    1. Moon EK, Wang L-C, Dolfi DV, et al. . Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin Cancer Res 2014;20:4262–73. 10.1158/1078-0432.CCR-13-2627
    1. Gust J, Hay KA, Hanafi L-A, et al. . Endothelial activation and blood-brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR-T cells. Cancer Discov 2017;7:1404–19. 10.1158/-17-0698
    1. Lum LG, Thakur A, Al-Kadhimi Z, et al. . Targeted T-cell therapy in stage IV breast cancer: a phase I clinical trial. Clin Cancer Res 2015;21:2305–14. 10.1158/1078-0432.CCR-14-2280
    1. Yankelevich M, Kondadasula SV, Thakur A, et al. . Anti-CD3 × anti-GD2 bispecific antibody redirects T-cell cytolytic activity to neuroblastoma targets. Pediatr Blood Cancer 2012;59:1198–205. 10.1002/pbc.24237
    1. Reusch U, Sundaram M, Davol PA, et al. . Anti-Cd3 X anti-epidermal growth factor receptor (EGFR) bispecific antibody redirects T-cell cytolytic activity to EGFR-positive cancers in vitro and in an animal model. Clin Cancer Res 2006;12:183–90. 10.1158/1078-0432.CCR-05-1855
    1. Vaishampayan U, Thakur A, Rathore R, et al. . Phase I study of anti-CD3 X anti-HER2 bispecific antibody in metastatic castrate resistant prostate cancer patients. Prostate Cancer 2015;2015:1–10. 10.1155/2015/285193
    1. Lum LG, Thakur A, Pray C, et al. . Multiple infusions of CD20-targeted T cells and low-dose IL-2 after SCT for high-risk non-Hodgkin's lymphoma: a pilot study. Bone Marrow Transplant 2014;49:73–9. 10.1038/bmt.2013.133
    1. Santich BH, Park JA, Tran H, et al. . Interdomain spacing and spatial configuration drive the potency of IgG-[L]-scFv T cell bispecific antibodies. Sci Transl Med 2020;12. 10.1126/scitranslmed.aax1315. [Epub ahead of print: 11 03 2020].
    1. Galon J, Angell HK, Bedognetti D, et al. . The continuum of cancer immunosurveillance: prognostic, predictive, and mechanistic signatures. Immunity 2013;39:11–26. 10.1016/j.immuni.2013.07.008
    1. Thakur A, Huang M, Lum LG. Bispecific antibody based therapeutics: strengths and challenges. Blood Rev 2018;32:339–47. 10.1016/j.blre.2018.02.004
    1. Xu H, Buhtoiarov IN, Guo H, et al. . A novel multimeric IL15/IL15Rα-Fc complex to enhance cancer immunotherapy. Oncoimmunology 2021;10:1893500. 10.1080/2162402X.2021.1893500
    1. Xu H, Cheng M, Guo H, et al. . Retargeting T cells to GD2 pentasaccharide on human tumors using bispecific humanized antibody. Cancer Immunol Res 2015;3:266–77. 10.1158/2326-6066.CIR-14-0230-T
    1. Lopez-Albaitero A, Xu H, Guo H, et al. . Overcoming resistance to HER2-targeted therapy with a novel HER2/CD3 bispecific antibody. Oncoimmunology 2017;6:e1267891. 10.1080/2162402X.2016.1267891
    1. Sen M, Wankowski DM, Garlie NK, et al. . Use of anti-CD3 X anti-HER2/neu bispecific antibody for redirecting cytotoxicity of activated T cells toward HER2/neu+ tumors. J Hematother Stem Cell Res 2001;10:247–60. 10.1089/15258160151134944
    1. Hoseini SS, Guo H, Wu Z, et al. . A potent tetravalent T-cell-engaging bispecific antibody against CD33 in acute myeloid leukemia. Blood Adv 2018;2:1250–8. 10.1182/bloodadvances.2017014373
    1. Wu Z, Guo H-F, Xu H, et al. . Development of a tetravalent Anti-GPA33/Anti-CD3 bispecific antibody for colorectal cancers. Mol Cancer Ther 2018;17:2164–75. 10.1158/1535-7163.MCT-18-0026
    1. Lopez-Albaitero A, Xu H, Guo H, et al. . Overcoming resistance to HER2-targeted therapy with a novel HER2/CD3 bispecific antibody. Oncoimmunology 2017;6:e1267891. 10.1080/2162402X.2016.1267891
    1. Andrade D, Redecha PB, Vukelic M, et al. . Engraftment of peripheral blood mononuclear cells from systemic lupus erythematosus and antiphospholipid syndrome patient donors into BALB-RAG-2-/- IL-2Rγ-/- mice: a promising model for studying human disease. Arthritis Rheum 2011;63:2764–73. 10.1002/art.30424
    1. Ishiguro T, Sano Y, Komatsu S-I, et al. . An anti-glypican 3/CD3 bispecific T cell-redirecting antibody for treatment of solid tumors. Sci Transl Med 2017;9. 10.1126/scitranslmed.aal4291. [Epub ahead of print: 04 Oct 2017].
    1. Park JA, Cheung N-KV. Gd2 or HER2 targeting T cell engaging bispecific antibodies to treat osteosarcoma. J Hematol Oncol 2020;13:172. 10.1186/s13045-020-01012-y
    1. Dobrenkov K, Ostrovnaya I, Gu J, et al. . Oncotargets GD2 and GD3 are highly expressed in sarcomas of children, adolescents, and young adults. Pediatr Blood Cancer 2016;63:1780–5. 10.1002/pbc.26097
    1. Yankelevich M, Modak S, Chu R, et al. . Phase I study of OKT3 X hu3F8 bispecific antibody (GD2Bi) armed T cells (GD2BATs) in GD2-positive tumors. Journal of Clinical Oncology 2019;37:2533. 10.1200/JCO.2019.37.15_suppl.2533
    1. Stone JD, Artyomov MN, Chervin AS, et al. . Interaction of streptavidin-based peptide-MHC oligomers (tetramers) with cell-surface TCRs. J Immunol 2011;187:6281–90. 10.4049/jimmunol.1101734
    1. Ehx G, Somja J, Warnatz H-J, et al. . Xenogeneic graft-versus-host disease in humanized NSG and NSG-HLA-A2/HHD mice. Front Immunol 2018;9:9. 10.3389/fimmu.2018.01943
    1. Morton JJ, Bird G, Refaeli Y, et al. . Humanized mouse xenograft models: narrowing the Tumor-Microenvironment gap. Cancer Res 2016;76:6153–8. 10.1158/0008-5472.CAN-16-1260
    1. Fisher J, Anderson J. Engineering approaches in human gamma delta T cells for cancer immunotherapy. Front Immunol 2018;9:1409. 10.3389/fimmu.2018.01409
    1. Ménétrier-Caux C, Ray-Coquard I, Blay J-Y, et al. . Lymphopenia in cancer patients and its effects on response to immunotherapy: an opportunity for combination with cytokines? J Immunother Cancer 2019;7:85. 10.1186/s40425-019-0549-5
    1. Velasquez MP, Torres D, Iwahori K, et al. . T cells expressing CD19-specific Engager molecules for the immunotherapy of CD19-positive malignancies. Sci Rep 2016;6:27130. 10.1038/srep27130
    1. Choi BD, Yu X, Castano AP, et al. . Car-T cells secreting bites circumvent antigen escape without detectable toxicity. Nat Biotechnol 2019;37:1049–58. 10.1038/s41587-019-0192-1
    1. Klinger M, Brandl C, Zugmaier G, et al. . Immunopharmacologic response of patients with B-lineage acute lymphoblastic leukemia to continuous infusion of T cell-engaging CD19/CD3-bispecific bite antibody blinatumomab. Blood 2012;119:6226–33. 10.1182/blood-2012-01-400515
    1. Staflin K, Zuch de Zafra CL, Schutt LK, et al. . Target arm affinities determine preclinical efficacy and safety of anti-HER2/CD3 bispecific antibody. JCI Insight 2020;5. 10.1172/jci.insight.133757. [Epub ahead of print: 09 Apr 2020].
    1. Bluemel C, Hausmann S, Fluhr P, et al. . Epitope distance to the target cell membrane and antigen size determine the potency of T cell-mediated lysis by bite antibodies specific for a large melanoma surface antigen. Cancer Immunol Immunother 2010;59:1197–209. 10.1007/s00262-010-0844-y
    1. Cuesta Ángel M., Sainz-Pastor N, Bonet J, et al. . Multivalent antibodies: when design surpasses evolution. Trends Biotechnol 2010;28:355–62. 10.1016/j.tibtech.2010.03.007
    1. Ellerman D. Bispecific T-cell engagers: towards understanding variables influencing the in vitro potency and tumor selectivity and their modulation to enhance their efficacy and safety. Methods 2019;154:102–17. 10.1016/j.ymeth.2018.10.026
    1. Mandikian D, Takahashi N, Lo AA, et al. . Relative target affinities of T-cell-dependent bispecific antibodies determine biodistribution in a solid tumor mouse model. Mol Cancer Ther 2018;17:776–85. 10.1158/1535-7163.MCT-17-0657
    1. Lee DW, Gardner R, Porter DL, et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188–95. 10.1182/blood-2014-05-552729
    1. Grupp SA, Kalos M, Barrett D, et al. . Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 2013;368:1509–18. 10.1056/NEJMoa1215134
    1. Kochenderfer JN, Dudley ME, Feldman SA, et al. . B-Cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood 2012;119:2709–20. 10.1182/blood-2011-10-384388
    1. Suntharalingam G, Perry MR, Ward S, et al. . Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 2006;355:1018–28. 10.1056/NEJMoa063842
    1. Li J, Piskol R, Ybarra R, et al. . Cd3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity. Sci Transl Med 2019;11. 10.1126/scitranslmed.aax8861. [Epub ahead of print: 04 Sep 2019].
    1. Lum LG, Thakur A, Choi M, et al. . Clinical and immune responses to anti-CD3 X anti-EGFR bispecific antibody armed activated T cells (EGFR bats) in pancreatic cancer patients. Oncoimmunology 2020;9:1773201. 10.1080/2162402X.2020.1773201
    1. Owen RE, Sinclair E, Emu B, et al. . Loss of T cell responses following long-term cryopreservation. J Immunol Methods 2007;326:93–115. 10.1016/j.jim.2007.07.012
    1. Weiner J, Duran-Struuck R, Zitsman J, et al. . Restimulation after cryopreservation and thawing preserves the phenotype and function of expanded baboon regulatory T cells. Transplant Direct 2015;1:1–7. 10.1097/TXD.0000000000000511
    1. Ford T, Wenden C, Mbekeani A, et al. . Cryopreservation-related loss of antigen-specific IFNγ producing CD4. Vaccine 2017;35:1898–906.
    1. Brudno JN, Somerville RPT, Shi V, et al. . Allogeneic T cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-cell malignancies that progress after allogeneic hematopoietic stem-cell transplantation without causing graft-versus-host disease. J Clin Oncol 2016;34:1112–21. 10.1200/JCO.2015.64.5929
    1. Lum LG, Ramesh M, Thakur A, et al. . Targeting cytomegalovirus-infected cells using T cells armed with anti-CD3 × anti-CMV bispecific antibody. Biol Blood Marrow Transplant 2012;18:1012–22. 10.1016/j.bbmt.2012.01.022
    1. Dutour A, Marin V, Pizzitola I, et al. . In vitro and in vivo antitumor effect of Anti-CD33 chimeric receptor-expressing EBV-CTL against CD33 acute myeloid leukemia. Adv Hematol 2012;2012:683065. 10.1155/2012/683065
    1. Pilipow K, Roberto A, Roederer M, et al. . Il15 and T-cell stemness in T-cell-based cancer immunotherapy. Cancer Res 2015;75:5187–93. 10.1158/0008-5472.CAN-15-1498
    1. Hurton LV, Singh H, Najjar AM, et al. . Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A 2016;113:E7788–97. 10.1073/pnas.1610544113
    1. Van Acker HH, Anguille S, Willemen Y, et al. . Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J Hematol Oncol 2016;9:101. 10.1186/s13045-016-0329-3
    1. Zhao Y, Niu C, Cui J. Gamma-delta (γδ) T cells: friend or foe in cancer development? J Transl Med 2018;16:3. 10.1186/s12967-017-1378-2
    1. Liu Y, Zhang C. The role of human γδ T cells in anti-tumor immunity and their potential for cancer immunotherapy. Cells 2020;9:1206. 10.3390/cells9051206
    1. Richman SA, Nunez-Cruz S, Moghimi B, et al. . High-Affinity GD2-Specific CAR T cells induce fatal encephalitis in a preclinical neuroblastoma model. Cancer Immunol Res 2018;6:36–46. 10.1158/2326-6066.CIR-17-0211
    1. Hoseini SS, Dobrenkov K, Pankov D, et al. . Bispecific antibody does not induce T-cell death mediated by chimeric antigen receptor against disialoganglioside GD2. Oncoimmunology 2017;6:e1320625. 10.1080/2162402X.2017.1320625
    1. Santich B, Hoseini S, Suzuki M. 41BB or CD28 driven disialoganglioside (GD2)-specific CAR-T, but not T-cell engaging bispecific antibody, induces fatal neurotoxicity in mice. In: Advances in Neuroblastoma Research 2021 Annual Meeting; Virtual 2021.
    1. Grabert RC, Cousens LP, Smith JA, et al. . Human T cells armed with HER2/neu bispecific antibodies divide, are cytotoxic, and secrete cytokines with repeated stimulation. Clin Cancer Res 2006;12:569–76. 10.1158/1078-0432.CCR-05-2005

Source: PubMed

3
Abonnere