Allogeneic CD20-targeted γδ T cells exhibit innate and adaptive antitumor activities in preclinical B-cell lymphoma models

Kevin P Nishimoto, Taylor Barca, Aruna Azameera, Amani Makkouk, Jason M Romero, Lu Bai, Mary M Brodey, Jackie Kennedy-Wilde, Hui Shao, Stephanie Papaioannou, Amy Doan, Cynthia Masri, Ngoc T Hoang, Hayden Tessman, Vidhya Dhevi Ramanathan, Ana Giner-Rubio, Frank Delfino, Kriti Sharma, Kevin Bray, Matthew Hoopes, Daulet Satpayev, Ranjita Sengupta, Marissa Herrman, Stewart E Abbot, Blake T Aftab, Zili An, Swapna Panuganti, Sandra M Hayes, Kevin P Nishimoto, Taylor Barca, Aruna Azameera, Amani Makkouk, Jason M Romero, Lu Bai, Mary M Brodey, Jackie Kennedy-Wilde, Hui Shao, Stephanie Papaioannou, Amy Doan, Cynthia Masri, Ngoc T Hoang, Hayden Tessman, Vidhya Dhevi Ramanathan, Ana Giner-Rubio, Frank Delfino, Kriti Sharma, Kevin Bray, Matthew Hoopes, Daulet Satpayev, Ranjita Sengupta, Marissa Herrman, Stewart E Abbot, Blake T Aftab, Zili An, Swapna Panuganti, Sandra M Hayes

Abstract

Objectives: Autologous chimeric antigen receptor (CAR) αβ T-cell therapies have demonstrated remarkable antitumor efficacy in patients with haematological malignancies; however, not all eligible cancer patients receive clinical benefit. Emerging strategies to improve patient access and clinical responses include using premanufactured products from healthy donors and alternative cytotoxic effectors possessing intrinsic tumoricidal activity as sources of CAR cell therapies. γδ T cells, which combine innate and adaptive mechanisms to recognise and kill malignant cells, are an attractive candidate platform for allogeneic CAR T-cell therapy. Here, we evaluated the manufacturability and functionality of allogeneic peripheral blood-derived CAR+ Vδ1 γδ T cells expressing a second-generation CAR targeting the B-cell-restricted CD20 antigen.

Methods: Donor-derived Vδ1 γδ T cells from peripheral blood were ex vivo-activated, expanded and engineered to express a novel anti-CD20 CAR. In vitro and in vivo assays were used to evaluate CAR-dependent and CAR-independent antitumor activities of CD20 CAR+ Vδ1 γδ T cells against B-cell tumors.

Results: Anti-CD20 CAR+ Vδ1 γδ T cells exhibited innate and adaptive antitumor activities, such as in vitro tumor cell killing and proinflammatory cytokine production, in addition to in vivo tumor growth inhibition of B-cell lymphoma xenografts in immunodeficient mice. Furthermore, CD20 CAR+ Vδ1 γδ T cells did not induce xenogeneic graft-versus-host disease in immunodeficient mice.

Conclusion: These preclinical data support the clinical evaluation of ADI-001, an allogeneic CD20 CAR+ Vδ1 γδ T cell, and a phase 1 study has been initiated in patients with B-cell malignancies (NCT04735471).

Keywords: B‐cell lymphoma; CD20; adoptive cell therapy; chimeric antigen receptor; γδ T cells.

Conflict of interest statement

KPN, TB, AA, AM, JMR, LB, MMB, JK‐W, HS, SP, AD, CM, NTH, HT, VDR, AG‐R, M Hoopes, DS, RS, M Herrman, SEA, BTA, ZA, SP and SMH are, or were, employees of Adicet Bio, Inc., and FD, KS and KB are employees of Regeneron Pharmaceuticals, Inc.

© 2022 Adicet Bio Inc. Clinical & Translational Immunology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology, Inc.

Figures

Figure 1
Figure 1
Binding properties of the fully human anti‐CD20 mAb clone 3H7. (a) Titration of 3H7 mAb (), therapeutic anti‐CD20 mAb rituximab () and human IgG1 control antibody () binding to parental HEK293 cells, HEK293 cells stably expressing human CD20 (HEK293‐hCD20), and Raji and Daudi human B‐cell lymphoma cell lines. Cell‐surface binding of the antibodies over a range of concentrations (1.69 pm to 100 nm) was performed at 4°C, detected by flow cytometry using APC‐labelled anti‐human IgG antibody and reported as geometric mean fluorescence intensity (geoMFI). For each dose curve, staining with the APC‐labelled anti‐human IgG antibody alone is included in the graph as a continuation of the threefold serial dilution and is represented as the lowest dose. EC50 values of the binding curves for each anti‐CD20 mAb on each CD20+ cell line are shown. Data are representative of two independent experiments. (b) Biochemical off‐rates of 3H7 mAb () and rituximab () were determined by measuring the amount of cell‐bound antibody over the course of a 3‐h time period. Antibodies (2 μg mL−1 each) were incubated with Raji cells for 2 h at room temperature. The cells were then washed, resuspended in 1% serum containing medium and incubated at 37°C. At time 0, 15, 30, 45, 60, 90, 120 and 180 min, an aliquot of cells was removed, washed, stained with PE‐labelled anti‐human Fc antibody and analysed by flow cytometry.
Figure 2
Figure 2
Overview of allogeneic CD20 CAR Vδ1 γδ T‐cell manufacturing process. (a) Flow chart highlighting the key steps in the manufacture of allogeneic CD20 CAR+ Vδ1 γδ T cells. Vδ1 γδ T cells present in healthy donor‐derived PBMCs are ex vivo‐activated and expanded with an agonistic anti‐Vδ1 mAb and then engineered to express the CD20 CAR by transduction with a self‐inactivating, replication‐incompetent gamma‐retroviral vector. After transduction, γδ T cells are further expanded in culture with human IL‐2 and are then enriched by αβ T‐cell depletion prior to formulation and cryopreservation. (b) Schematic diagram of the second‐generation CD20 CAR, which consists of the 3H7 scFv combined with CD8α hinge and transmembrane regions fused to the intracellular signalling domains of 4‐1BB and CD3ζ. CAR expression is regulated by the EF1a promoter. (c) The CD20 CAR+ Vδ1 γδ T‐cell manufacturing process results in a substantial fold expansion of Vδ1 γδ T cells. The mean ± standard deviation (SD) of five manufacturing runs using PBMCs from three different donors (large‐scale) and five different donors (small‐scale) are shown. (d) Average percentage of Vδ1 γδ T cells expressing the CD20 CAR from small‐scale and large‐scale manufacturing runs as measured by flow cytometry. The mean ± SD of five small‐scale manufacturing runs using PBMCs from five different donors and of seven large‐scale manufacturing runs using PBMCs from four different donors are shown. (e) Box plots of % cell composition using flow cytometric analysis of CD20 CAR+ Vδ1 T‐cell products derived from four different donors at Day 0, pre‐depletion and post‐depletion of αβ T‐cell time points (large scale). Box plot horizontal lines represent the mean, minimum and maximum values of each % cell type (Vδ1 T cells, Vδ2 T cells, αβ T cells and NK cells). (f) Cellular composition of final cell product was assessed by flow cytometry. Pie charts represent the composition of three representative donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products. (g) Selective activation and expansion of Vδ1 γδ T cells with an agonistic anti‐Vδ1 mAb results in a diverse, not skewed, TCRγ repertoire. Treemaps of the TCRγ repertoire, which were created using Tableau software and are reported as TRGV (Vγ) usage in each functionally rearranged CDR3 sequence. Two donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products are shown. The size of the square indicates its relative frequency in each sample. Relative frequencies of functionally rearranged TRGV (Vγ) sequences from ImMunoGeneTics (IMGT) are provided as a reference. Statistics were calculated using the paired t‐test; ns, not significant.
Figure 3
Figure 3
Structure and signalling capacity of the CD20 (3H7) CAR. (a) Transduction efficiency of Jurkat‐Lucia™ cells, which carry an NFAT‐inducible luciferase reporter, with the CD20 CAR gamma‐retroviral vector. Percentage of CAR‐transduced Jurkat‐Lucia cells expressing the CD20 CAR on their cell surface was measured by flow cytometry and is representative of three independent experiments. (b) The CD20 CAR, when expressed on the surface of Jurkat‐Lucia cells, does not induce antigen‐independent tonic signalling. CAR‐mediated signal transduction and activation were assessed in the presence or absence of target antigen by culturing parental (untransduced) and CAR‐transduced Jurkat‐Lucia cells either alone or at a 1:1 E:T ratio with CD20+ Raji cells. Stimulation with PMA (phorbol 12‐myristate 13‐acetate) and ionomycin was used as a positive control for NFAT activation. Duplicate samples were tested for each condition. Data are representative of three independent experiments. (c) The 3H7 CAR is expressed as dimers and higher‐order oligomers on the surface of Vδ1 γδ T cells. Untransduced (UT) or CAR‐transduced Vδ1 γδ T cells were surface‐biotinylated using EZ‐Link Sulfo‐NHS‐LC‐Biotin, lysed in Brij® O10 buffer containing a protease inhibitor cocktail and incubated with an anti‐CD20 CAR mAb coupled to Sepharose 4B beads. Immunoprecipitated proteins were resolved by non‐reducing (NR) and reducing SDS‐PAGE analysis and transferred to nitrocellulose membranes. Biotinylated CAR complexes were visualised by IRDye® 800CW Streptavidin and the Odyssey Imager. The positions of CAR dimers and oligomers (NR conditions) and monomers (R conditions) are marked. Data are representative of four independent experiments. (d) CAR‐associated phospho‐CD3ζ is only detected after CAR stimulation. CD20 CAR+ Vδ1 γδ T cells were unstimulated or stimulated on Protein L‐coated 96‐well plates for 10 min and then lysed in Milliplex® MAP Cell Signaling Universal Lysis Buffer. Lysates were resolved by reducing SDS‐PAGE analysis and then transferred to nitrocellulose membranes. Membranes were blocked and then incubated with antibodies against total CD3ζ or phospho‐CD3ζ (Tyr 142) overnight at 4°C on a shaker. CAR‐associated CD3ζ and phospho‐CD3ζ proteins were visualised using anti‐mouse IgG IR800 and the Odyssey Imager, and their positions are marked. In whole‐cell lysates, the CAR resolves as two bands: enzymatic deglycosylation of cell lysates revealed that the upper band is the glycosylated form, and that the lower band is the unglycosylated form (data not shown). Data are representative of three independent experiments.
Figure 4
Figure 4
Phenotypic analysis of CD20 CAR+ Vδ1 γδ T cells. (a, b) Flow cytometric analysis reveals that the majority of CAR+ Vδ1 γδ T cells in three donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products exhibit a naïve‐like T‐cell memory phenotype. Expression levels of CCR7, CD62L, CD95 and CD45RO on gated naïve (CD27+ CD45RA+) CD4+ T cells from healthy donor‐derived PBMCs are shown as naïve T‐cell controls. Memory cell subsets were defined by the expression of CD27 and CD45RA: naïve‐like (CD27+ CD45RA+); central memory or TCM (CD27+ CD45RA−); effector memory or TEM (CD27− CD45RA−); and terminally differentiated effector memory or TEMRA (CD27− CD45RA+). (c) Heatmap showing percentages of CAR+ Vδ1 γδ T cells in three donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products that express chemokine receptors, natural killer (NK) cell receptors and terminal differentiation markers. (d) Heatmap showing percentages of CAR+ Vδ1 γδ T cells in four donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products that co‐express PD‐1 and another co‐inhibitory receptor, that is TIM‐3, LAG‐3 or TIGIT.
Figure 5
Figure 5
Comparison of the cytotoxic potentials of untransduced Vδ1 γδ T cells, CD20 CAR‐transduced Vδ1 γδ T cells and CD20 CAR‐transduced αβ T cells against CD20+ target cells. Cytotoxic potentials of untransduced Vδ1 γδ T cells, CD20 CAR‐transduced Vδ1 γδ T cells (71.5% CAR+) and CD20 CAR‐transduced αβ T cells (68% CAR+), all derived from the same healthy donor, were evaluated against CD20+ target cells in a 48‐h IncuCyte Immune Cell Killing Assay, in which T cells were co‐cultured with NucR‐expressing Raji or Mino target cells at E:T ratios of 10:1, 3:3:1 and 1.1:1. The viability of the NucR‐expressing targets, reported as cytotoxicity index, was monitored every 2 h over the course of 48 h with the IncuCyte Live‐Cell Analysis System. Please note that the lower the cytotoxicity index, the better the cytotoxic potential. Data are shown as mean ± SD of triplicates and are representative of three independent experiments using cell products from two different donors. The killing curves for each cell product were statistically analysed using the mixed model two‐way ANOVA. (****P < 0.0001, **P < 0.01 and *P < 0.05; NS, not significant).
Figure 6
Figure 6
In vitro functional analyses of preclinical large‐scale manufactured CD20 CAR+ Vδ1 γδ T cells. (a) Relative CD20 expression levels on malignant B‐cell lines [Mino, Raji, JVM‐2, WILL‐2, RR‐Raji], normal peripheral blood B cells and the CD20− Jurkat T‐ALL cell line as determined by flow cytometry. Relative CD20 expression is reported as CD20 geoMFI/isotype control geoMFI (MFI ratio). The data are representative of two independent experiments. (b) Cytotoxic potential of CD20 CAR+ Vδ1 γδ T cells against B‐lymphoma cell lines expressing varying levels of CD20 in a short‐term cytotoxicity assay. Cytotoxic potential was assessed by titrating E:T ratios in an 18‐h cytotoxicity assay in which CD20 CAR+ Vδ1 γδ T‐cell effectors were co‐cultured with RFluc‐expressing target cells. Controls included CD20 CAR+ Vδ1 γδ T cells and B‐lymphoma cell lines cultured alone. Data are shown as mean ± SD of triplicates and are representative of two independent experiments using two different donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products. (c) Effect of clinically relevant concentrations of rituximab on the ability of CD20 CAR+ Vδ1 γδ T cells to recognise and kill RFluc‐expressing RR‐Raji target cells. Cytotoxic potential of CD20 CAR+ Vδ1 γδ T cells against RR‐Raji cells at a fixed 3:1 ratio was assessed after pre‐incubation of target cells with increasing concentrations of rituximab (0–400 μg mL−1) in an 18‐h luciferase cytotoxicity assay. Data are shown as mean ± SD of triplicates and are representative of two different rituximab‐resistant B‐lymphoma cell lines [**P ≤ 0.005 (t‐test)]. (d) Cytokine and chemokine production by CD20 CAR+ Vδ1 γδ T‐cell effectors after an 18‐h co‐culture with Raji cells at fixed 1.75:1 ratio. Levels of IFN‐γ, GM‐CSF, TNF‐α, IL‐2, IL‐6, IL‐8, IL‐13, MIP‐1α, MIP‐1β, RANTES, IP‐10 and IL‐17A were measured in culture medium supernatants using the Luminex platform. Data are shown as mean ± SD of triplicates and are representative of two independent experiments using two different donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products. (e) Proliferative potential of CD20 CAR+ Vδ1 γδ T cells following three rounds of target antigen exposure in a 7‐day culture period. Aliquots of cells harvested on Days 2, 5 and 7 were analysed by flow cytometry to assess cellular proliferation by dye dilution. Representative data from three independent experiments using three donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products are shown. (f) Cytotoxic potential of CD20 CAR+ Vδ1 γδ T cells against Raji cells in a long‐term cytotoxicity assay. Cytotoxic potential was assessed at a 1:2 E:T ratio in a 120‐h cytotoxicity assay in which CD20 CAR+ Vδ1 γδ T cells were co‐cultured with NucR‐expressing Raji cells ± 20 IU of human IL‐2. The viability of the NucR‐expressing Raji cells was monitored every 2 h over the course of 120 h with the IncuCyte Live‐Cell Analysis System. Data are shown as mean ± SD of triplicates and represent three donor‐derived CD20 CAR+ Vδ1 γδ T‐cell products.
Figure 7
Figure 7
In vivo and ex vivo analyses of CD20+ tumor‐bearing NSG mice treated with CD20 CAR+ Vδ1 γδ T cells. (a)In vivo efficacy of three different doses (5 × 106, 1 × 107 and 2 × 107) of viable CD20 CAR+ Vδ1 γδ T cells in combination with 13 000 IU IL‐2 in a SC Raji Burkitt lymphoma model in NSG mice (n = 5 per group). The SC xenograft model was chosen because tumor control in this model is dependent on the ability of CAR T cells to traffic to the tumor, to proliferate and to lyse tumor cells. Tumor growth, body weight and animal health were monitored two times per week after tumor implantation. Tumor volume was calculated using the formula: Volume (mm3) = (length × width2)/2. The Kruskal–Wallis test was used to assess statistical significance among the groups on Day 14 (**P = 0.0011). Data are representative of three independent experiments using one donor‐derived CD20 CAR+ Vδ1 γδ T‐cell product. (b)In vivo efficacy of three different doses (2.5 × 106, 5 × 106 and 1 × 107) of viable CD20 CAR+ Vδ1 γδ T cells in combination with 13 000 IU IL‐2 in a SC JVM‐2 mantle cell lymphoma model in NSG mice (n = 5 per group). The Kruskal–Wallis test was used to assess statistical significance among the groups on Day 30 (**P = 0.0062). Data are representative of two independent experiments using one donor‐derived CD20 CAR+ Vδ1 γδ T‐cell product. (c) Proliferative potential of untransduced and CD20 CAR+ Vδ1 γδ T cells in blood, bone marrow and tumor harvested from Raji tumor‐bearing mice on Day 6 post‐treatment. Untransduced and CD20 CAR+ Vδ1 γδ T cells were labelled with CellTrace Violet prior to adoptive transfer at a dose of 8 × 106 cells in combination with IL‐2 (n = 3 per group). Data are representative of two independent experiments using one donor‐derived CD20 CAR+ Vδ1 γδ T‐cell product. (d) Differentiation potential of untransduced and CD20 CAR+ Vδ1 γδ T cells in bone marrow and tumor harvested from Raji tumor‐bearing mice on Day 6 post‐treatment. Memory cell subsets were defined by the expression of CD27 and CD45RA: naïve‐like (CD27+ CD45RA+); TCM (CD27+ CD45RA−); TEM (CD27− CD45RA−); and TEMRA (CD27− CD45RA+). Memory phenotypes of pre‐dose untransduced and CD20 CAR+ Vδ1 γδ T cells are shown for comparison. (e) Venn diagrams of DEGs in Raji tumors treated with 1 × 107 CD20 CAR+ Vδ1 γδ T cells (red circle) from two donors and in JVM‐2 tumors treated with 1 × 107 CD20 CAR+ Vδ1 γδ T cells (blue circle) from two donors in combination with IL‐2. The overlapping area represents DEGs that are shared between the two tumor models. Gene expression levels were quantitated using the nCounter® CAR T Characterization and Human Immunology Panels on the nCounter® SPRINT Profiler according to the manufacturer’s instructions. nSolver™ software was used to normalise the data and to identify DEGs (i.e. adjusted P ≤ 0.05 and log2 fold change ≥ 1) in tumors treated with CD20 CAR+ Vδ1 γδ T cells versus tumors treated with untransduced Vδ1 γδ T cells. Gene ontology analysis was performed using Database for Annotation, Visualization and Integrated Discovery (DAVID, https://david.ncifcrf.gov/) to identify the biological pathways associated with DEGs that are unique to each tumor model and that are shared by the two tumor models.
Figure 8
Figure 8
Phenotypic and functional analyses of clinical‐scale CD20 CAR+ Vδ1 γδ T cells. (a) Dot plot demonstrating that 79% of clinical‐scale CD20 CAR+ Vδ1 γδ T cells exhibit a naïve‐like T‐cell memory phenotype. Data are representative of two independent experiments. (b) Cytotoxic potential of, and IFN‐γ production by, clinical‐scale CD20 CAR+ Vδ1 γδ T cells (49% CAR+) against Raji cells in an 18‐h cytotoxicity assay. Controls included CD20 CAR+ Vδ1 γδ T cells and B‐lymphoma cell lines cultured alone. Levels of IFN‐γ in culture medium supernatants were measured using the Luminex platform. Data are shown as mean ± SD of triplicates and are representative of three independent experiments. (c) Cytotoxic potential of clinical CD20 CAR+ Vδ1 γδ T cells against Raji cells in a long‐term cytotoxicity assay. Cytotoxic potential was assessed at a 1:2 E:T ratio in a 120‐h cytotoxicity assay in which CD20 CAR+ Vδ1 γδ T‐cell effectors were co‐cultured with NucR‐expressing Raji cells in the presence or absence of 20 IU of human IL‐2. The viability of the NucR‐expressing Raji cells was monitored every 2 h over the course of 120 h with the IncuCyte Live‐Cell Analysis System. Data are shown as mean ± SD of triplicates and are representative of three independent experiments. (d)In vivo efficacy of two different doses (5 × 106 and 1 × 107) of viable clinical‐scale CD20 CAR+ Vδ1 γδ T cells in combination with 13 000 IU IL‐2 in a SC Raji Burkitt lymphoma model in NSG mice (n = 7 mice per cell treatment group and n = 10 mice in the tumor‐alone group). Tumor growth, body weight and animal health were monitored two times per week after tumor implantation. The Kruskal–Wallis test was used to assess statistical significance among groups on Day 15 (***P = 0.0001). The data represent one independent experiment.

References

    1. McCreedy BJ, Senyukov VV, Nguyen KT. Off the shelf T cell therapies for hematologic malignancies. Best Pract Res Clin Haematol 2018; 31: 166–175.
    1. Shah NN, Fry TJ. Mechanisms of resistance to CAR T cell therapy. Nat Rev Clin Oncol 2019; 16: 372–385.
    1. Ruella M, Kenderian SS. Next‐generation chimeric antigen receptor T‐cell therapy: going off the shelf. BioDrugs 2017; 31: 473–481.
    1. Graham C, Jozwik A, Pepper A, Benjamin R. Allogeneic CAR‐T cells: more than ease of access? Cells 2018; 7: 155.
    1. Aftab BT, Sasu B, Krishnamurthy J, Gschweng E, Alcazer V, Depil S. Toward “off‐the‐shelf” allogeneic CAR T cells. Adv Cell Gene Ther 2020; 3: 1–11.
    1. Gilham DE, Michaux A, Breman E et al. TCR inhibitory molecule as a promising allogeneic NKG2D CAR‐T cell approach. J Clin Oncol 2018; 36(Suppl 15): Abstract e15042.
    1. Shen RR, Pham CD, Min WUM, Munson DJ, Aftab BT. Functional demonstration of CD19 chimeric antigen receptor (CAR) engineered Epstein‐Barr virus (EBV) specific T cells: an off‐the‐shelf, allogeneic CAR T‐cell immunotherapy platform. Cancer Res 2019; 79(Suppl 13): Abstract 2310.
    1. Gomes AQ, Martins DS, Silva‐Santos B. Targeting γδ T lymphocytes for cancer immunotherapy: from novel mechanistic insight to clinical application. Cancer Res 2010; 70: 10024–10027.
    1. Lamb LS, Musk P, Ye Z et al. Human γδ+ T lymphocytes have in vitro graft vs leukemia activity in the absence of an allogeneic response. Bone Marrow Transplant 2001; 27: 601–606.
    1. Hayday AC, Saito H, Gillies SD et al. Structure, organization, and somatic rearrangement of T cell gamma genes. Cell 1985; 40: 259–269.
    1. Brenner MB, McLean J, Dialynas DP et al. Identification of a putative second T‐cell receptor. Nature 1986; 322: 145–149.
    1. Bonneville M, O’Brien RL, Born WK. γδ T cell effector functions: a blend of innate programming and acquired plasticity. Nat Rev Immunol 2010; 10: 467–478.
    1. Gentles AJ, Newman AM, Liu CL et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med 2015; 21: 938–945.
    1. Wu Y, Kyle‐Cezar F, Woolf RT et al. An innate‐like Vδ1+ γδ T cell compartment in the human breast is associated with remission in triple‐negative breast cancer. Sci Transl Med 2019; 11: eaax9364.
    1. Lo Presti E, Dieli F, Meraviglia S. Tumor‐infiltrating γδ T lymphocytes: pathogenic role, clinical significance, and differential programing in the tumor microenvironment. Front Immunol 2014; 5: 607.
    1. Siegers GM, Lamb LS Jr. Cytotoxic and regulatory properties of circulating Vδ1+ γδ T cells: a new player on the cell therapy field? Mol Ther 2014; 22: 1416–1422.
    1. Deniger DC, Moyes JS, Cooper LJ. Clinical applications of gamma delta T cells with multivalent immunity. Front Immunol 2014; 5: 636.
    1. Perko R, Kang G, Sunkara A, Leung W, Thomas PG, Dallas MH. Gamma delta T cell reconstitution is associated with fewer infections and improved event‐free survival after hematopoietic stem cell transplantation for pediatric leukemia. Biol Blood Marrow Transplant 2015; 21: 130–136.
    1. Bertaina A, Roncarolo MG. Graft engineering and adoptive immunotherapy: new approaches to promote immune tolerance after hematopoietic stem cell transplantation. Front Immunol 2019; 10: 1342.
    1. Hill JA, Li D, Hay KA et al. Infectious complications of CD19‐targeted chimeric antigen receptor‐modified T‐cell immunotherapy. Blood 2018; 131: 121–130.
    1. Siegers GM, Dutta I, Lai R, Postovit LM. Functional plasticity of gamma delta T cells and breast tumor targets in hypoxia. Front Immunol 2018; 9: 1367.
    1. Murphy AJ, Macdonald LE, Stevens S et al. Mice with megabase humanization of their immunoglobulin genes generate antibodies as efficiently as normal mice. Proc Natl Acad Sci USA 2014; 111: 5153–5158.
    1. Smith EJ, Olson K, Haber LJ et al. A novel, native‐format bispecific antibody triggering T‐cell killing of B‐cells is robustly active in mouse tumor models and cynomolgus monkeys. Sci Rep 2015; 5: 17943.
    1. Davey MS, Willcox CR, Joyce SP et al. Clonal selection in the human Vδ1 T cell repertoire indicates γδ TCR‐dependent adaptive immune surveillance. Nat Commun 2017; 8: 14760.
    1. Di Lorenzo B, Simões AE, Caiado F et al. Broad cytotoxic targeting of acute myeloid leukemia by polyclonal delta one T cells. Cancer Immunol Res 2019; 7: 552–558.
    1. Frigault MJ, Lee J, Basil MC et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol Res 2015; 3: 356–367.
    1. Long AH, Haso WM, Shern JF et al. 4–1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med 2015; 21: 581–590.
    1. Gomes‐Silva D, Mukherjee M, Srinivasan M et al. Tonic 4–1BB costimulation in chimeric antigen receptors impedes T cell survival and is vector‐dependent. Cell Rep 2017; 21: 17–26.
    1. Blanco R, Alarcón B. TCR nanoclusters as the framework for transmission of conformational changes and cooperativity. Front Immunol 2012; 3: 115.
    1. Fraietta JA, Lacey SF, Orlando EJ et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med 2018; 24: 563–571.
    1. Sommermeyer D, Hudecek M, Kosasih PL 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.
    1. Locke FL, Rossi J, Neelapu SS et al. Product characteristics associated with in vivo expansion of anti‐CD19 CAR T cells in patients treated with axicabtagene ciloleucel (axi‐cel). J Clin Oncol 2017; 35(Suppl 15): Abstract 3023.
    1. Pizzolato G, Kaminski H, Tosolini M et al. Single‐cell RNA sequencing unveils the shared and distinct cytotoxic hall marks of human TCRVδ1 and TCRVδ2 γδ T lymphocytes. Proc Natl Acad Sci USA 2019; 116: 11906–11915.
    1. Sebestyen Z, Prinz I, Déchanet‐Merville J, Santos‐Silva B, Kuball J. Translating gammadelta (γδ) T cells and their receptors into cancer cell therapies. Nat Rev Drug Discov 2020; 19: 169–184.
    1. Kabelitz D, Wesch D. Features and functions of γδ T lymphocytes: focus on chemokines and their receptors. Crit Rev Immunol 2003; 23: 339–370.
    1. Scott DW, Gascoyne RD. The tumour microenvironment in B cell lymphomas. Nat Rev Cancer 2014; 14: 517–534.
    1. Fowler NH, Cheah CY, Gascoyne RD et al. Role of the tumor microenvironment in mature B‐cell lymphoid malignancies. Haematologica 2016; 101: 531–540.
    1. Sarkar S, Sabhachandani P, Ravi D et al. Dynamic analysis of human natural killer cell response at single‐cell resolution in B‐cell non‐Hodgkin lymphoma. Front Immunol 2017; 8: 1736.
    1. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol 2015; 15: 486–499.
    1. Fisher J, Sharma R, Don DW et al. Engineering γδ T cells limits tonic signaling associated with chimeric antigen receptors. Sci Signal 2019; 12: eaax1872.
    1. Prevodnik VK, Lavrencak J, Horvat M, Novakovic BJ. The predictive significance of CD20 expression in B cell lymphomas. Diagn Pathol 2011; 6: 33.
    1. Sonoki T, Li Y, Miyanishi S et al. Establishment of a novel CD20 negative mature B‐cell line, WILL2, from a CD20 positive diffuse large B‐cell lymphoma patient treated with rituximab. Int J Hematol 2009; 89: 400–402.
    1. Berinstein NL, Grillo‐López AJ, White CA et al. Association of serum rituximab (IDEC‐C2B8) concentration and anti‐tumor response in the treatment of recurrent low‐grade or follicular non‐Hodgkin’s lymphoma. Ann Oncol 1998; 9: 995–1001.
    1. Rougé L, Chiang N, Steffek M et al. Structure of CD20 in complex with the therapeutic monoclonal antibody rituximab. Science 2020; 367: 1224–1230.
    1. Xue Q, Bettini E, Paczkowski P et al. Single‐cell multiplexed cytokine profiling of CD19 CAR‐T cells reveals a diverse landscape of polyfunctional antigen‐specific response. J Immunother Cancer 2017; 5: 85.
    1. Rossi J, Paczkowski P, Shen Y‐W et al. Preinfusion polyfunctional anti‐CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 2018; 132: 804–814.
    1. Shultz LD, Ishikawa F, Greiner DL. Humanized mice in translational biomedical research. Nat Rev Immunol 2007; 7: 118–130.
    1. Neelapu SS. CAR‐T efficacy: is conditioning the key? Blood 2019; 133: 1799–1800.
    1. Almeida AR, Correia DV, Fernandes‐Platzgummer A et al. Delta one T cells for immunotherapy of chronic lymphocytic leukemia: clinical‐grade expansion/differentiation and preclinical proof of concept. Clin Cancer Res 2016; 22: 5795–5804.
    1. Capsomidis A, Benthall G, Van Acker HH et al. Chimeric antigen receptor‐engineered human gamma delta T cells: enhanced cytotoxicity with retention of cross presentation. Mol Ther 2018; 26: 354–365.
    1. Kaartinen T, Luostarinen A, Maliniemi P et al. Low interleukin‐2 concentration favors generation of early memory T cells over effector phenotypes during chimeric antigen receptor T‐cell expansion. Cytotherapy 2017; 19: 689–702.
    1. Xu Y, Zhang M, Ramos CA et al. Closely related T‐memory stem cells correlate with in vivo expansion of CAR.CD19‐T cells and are preserved by IL‐7 and IL‐15. Blood 2014; 123: 3750–3759.
    1. Daydé D, Ternant D, Ohresser M et al. Tumor burden influences exposure and response to rituximab: pharmacokinetic‐pharmacodynamic modeling using a syngeneic bioluminescent murine model expressing human CD20. Blood 2009; 113: 3765–3772.
    1. Fujisaki H, Kakuda H, Shimasaki N et al. Expansion of highly cytotoxic human natural killer cells for cancer cell therapy. Cancer Res 2009; 69: 4010–4017.
    1. Sommer C, Boldajipour B, Kuo TC et al. Preclinical evaluation of allogeneic CAR T cells targeting BCMA for the treatment of multiple myeloma. Mol Ther 2019; 27: 1126–1138.
    1. Liu E, Tong Y, Dotti G et al. Cord blood NK cells engineered to express IL‐15 and a CD19‐targeted CAR show long‐term persistence and potent antitumor activity. Leukemia 2018; 32: 520–531.
    1. Chen Y, Sun C, Landoni E, Metelitsa L, Dotti G, Savoldo B. Eradication of neuroblastoma by T cells redirected with an optimized GD2‐specific chimeric antigen receptor and interleukin‐15. Clin Cancer Res 2019; 25: 2915–2924.
    1. Makkouk A, Yang X, Barca T et al. Off‐the‐shelf Vδ1 gamma delta T cells engineered with glypican‐3 (GPC‐3)‐specific chimeric antigen receptor (CAR) and soluble IL‐15 display robust antitumor efficacy against hepatocellular carcinoma. J Immunother Cancer 2021; 9: e003441.
    1. Abou‐el‐Enein M, Elsallab M, Feldman SA et al. Scalable manufacturing of CAR T cells for cancer immunotherapy. Blood Cancer Discov 2021; 5: 408–422.
    1. Eyles JE, Vessillier S, Jones A, Stacey G, Schneider CK, Price J. Cell therapy products: focus on issues with manufacturing and quality control of chimeric antigen receptor T‐cell therapies. Chem Technol Biotechnol 2019; 94: 1008–1016.
    1. Czuczman MS, Olejniczak D, Gowda A et al. Acquirement of rituximab resistance in lymphoma cell lines is associated with both global CD20 gene and protein down‐regulation regulated at the pretranscriptional and posttranscriptional levels. Clin Cancer Res 2008; 14: 1561–1570.
    1. Zheng Z, Chinnasamy N, Morgan RA. Protein L: a novel reagent for the detection of chimeric antigen receptor (CAR) expression by flow cytometry. J Transl Med 2012; 10: 29.
    1. Wang D, Starr R, Alizadeh D, Yang X, Forman SJ, Brown CE. In vitro tumor cell rechallenge for predictive evaluation of chimeric antigen receptor T cell antitumor function. J vis Exp 2019; (144): e59275.

Source: PubMed

3
Tilaa