Skip to main content

TCR γ4δ1-Engineered αβT Cells Exhibit Effective Antitumor Activity

Abstract

T cell engineering with T cell receptors (TCRs) specific for tumors plays an important role in adoptive T cell transfer (ATC) therapy for cancer. Here, we present a novel strategy to redirect peripheral blood-derived αβT cells against tumors via TCRγ4δ1 gene transduction. The broad-spectrum antitumor activity of TCRδ1 cells in innate immunity is dependent on CDR3δ1. TCRγ4δ1-engineered αβT cells were prepared by lentiviral transduction and characterized by analyzing in vitro and in vivo cytotoxicity to tumors, ability of proliferation and cytokine production, and potential role in autoimmunity. Results show that TCRγ4δ1 genes were transduced to approximately 36% of polyclonal αβT cells. TCRγ4δ1-engineered αβT cells exhibited effective in vitro TCRγδ-dependent cytotoxicity against various tumor cells via the perforin-granzyme pathway. They also showed a strong proliferative capacity and robust cytokine production. TCRγ4δ1-engineered αβT cells neither expressed mixed TCR dimers nor bound/killed normal cells in vitro. More important, adoptive transfer of TCRγ4δ1-engineered αβT cells into nude mice bearing a human HepG2 cell line significantly suppressed tumor growth. Our results demonstrate a novel role for TCRγ4δ1 in gene therapy and ATC for cancer.

Introduction

Adoptive T cell transfer (ACT) is a promising immunotherapy strategy for cancer. Chimeric antigen receptor (CAR)-modified T cells have shown promise in the treatment of B cell malignancy. (1) However, ACT immunotherapy for solid tumors faces the challenge of specificity when targeting tumors. To date, T cells have frequently been tested as effector cells for ACT, including tumor infiltrating lymphocytes (TILs) for metastatic melanoma (2,3) and T cell receptor (TCR) gene-engineered T cells for other tumor types. (4,5) Although Rosenberg’s laboratory found that a mutation in erbb2 interacting protein triggered CD4 + TH1 cell activation and demonstrated a cure efficacy in ACT, the method is not feasible, given the high cost and complicated process. (6) Therefore, our laboratory and other groups have pursued TCRγδ gene therapy as an alternative approach.

Compared with TCRαβ, which is highly specific for its antigen, TCRγδ displays characteristics of innate immunity, directly recognizing many stress-induced antigens in an MHC-independent manner in the early stages of inflammation and tumorigenesis. (7) Human γδT cells are grouped into 2 major subsets, Vδ1 and Vδ2 T cells. Vδ1 T cells are common in mucosa, especially the submucosal areas of the gastrointestinal, respiratory and genitourinary tracts. They recognize MHC class I-related molecules A and B (MICA and MICB) and UL-16-binding proteins (ULBPs) expressed at variable levels on epithelial tumor cells and some leukemias and lymphomas. Vδ2 T cells belong to a minor subset of the total T cell pool in the peripheral blood, responding mainly to aminobisphosphonates/synthetic phosphoantigen. (8) Due to broad-spectrum tumor recognition of TCRγδ, TCRγδ gene transduction into effector T cells, such as αβT cells, may be an attractive therapeutic approach. It appears to resolve the fundamental problem of tumor targeting not found in TCRαβ. Previous studies by other groups and our laboratory have confirmed that TCRγ9δ2-transduced αβT cells, or TCRγ9δ2-modified peripheral blood mononuclear cells (PBMCs), mediate cytotoxicity against a broad range of tumor cell lines in vitro and suppress tumor growth in Daudi or SKOV3 tumor cell-bearing mice models. (9,10)

Preparing a large number of tumor-reactive T cells in a short time is a major challenge for ACT in cancer patients. Transduction of tumor antigen-reactive TCR into T cells is one strategy to acquire sufficient T cells. Antigen-specific TCRαβ-modified CD8 + αβT cells display significant antitumor activity (11) However, a potential disadvantage of TCRαβ-modified αβT cells is the formation of new mixed TCRs. Specifically, the introduced TCRα or TCRβ chains can pair with the endogenous TCRα or TCRβ chains to generate mixed TCR dimers. These mixed TCR dimers do not experience thymic negative selection during T cell development and elicit an autoimmune response. Bendle et al. first noted that OT-I TCRαβ-transduced CD8 + T cells triggered in vivo mispairing-mediated autoimmunity in C57BL/6 mice. (12) To prevent this, multiple approaches were used, including murinized TCR and cysteine-modified TCR, with γδT cells as recipient cells transduced with exogenous TCRαβ. (13,14) TCRαβ-engineered γδT cells exhibited no evidence of TCR mispairing; TCRδ or TCRγ in γδT cells did not exchange chains with TCRαβ. (15,16) Other approaches, except for complete knockdown of TCRα and TCRβ chains, reduced but did not completely prevent mispairing. Nevertheless, other major drawbacks of TCRαβs as genetic donors include MHC restriction and single antigen-peptide recognition, restricting the application to fewer patients. (17)

Our laboratory previously identified complementary determining region 3 (CDR3) at a high frequency in TCRδ1 chain from TILs in human gastric cancer. This high-frequency CDR3, termed GTM, is a critical region for tumor antigen recognition; its binding to gastric tumors reached 88.89%. The full-length TCRδ1 with GTM and TCRγ4 chains were amplified and paired to form TCRγ4δ1 receptor. We confirmed TCRγ4δ1-Fc fusion proteins bind to a large panel of tumor cell lines and tissues through their TCRs, including gastric carcinoma BGC823, kidney cancer G401, lung adenocarcinoma GLC-82, colonic carcinoma HT29 and ovarian cancer SKOV3. These fusion proteins strongly bind MICA and ULBP5 in enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR) assay. (18) MICA is a stress-inducible cell surface antigen expressed on epithelial tumors in lung, colon, prostate, kidney, breast and ovary carcinomas. (19) Our previous data demonstrated that TCRγ4δ1 has broad-spectrum tumor recognition. Due to the high prevalence and diversity of TCRγ4δ1 ligands on malignant cells, we reasoned that TCRγ4δ1 targeting various ligands as genetic donors to modify effector cells is a more promising approach compared with specific single-chain antibody fragments (scFv) or TCRαβ targeting only single antigens. In addition, it prevented tumor immune escape resulting from loss or deficiency of single antigens during tumor progression. Thus, we propose a promising strategy to redirect peripheral blood-derived αβT cells against various tumors via a broadly tumor-reactive TCRγ4δ1 gene transfer method.

Here we show for the first time that TCRγ4δ1-engineered αβT cells exhibit significant antitumor activity in vitro and in vivo. Moreover, TCRγ4δ1-engineered αβT cells neither expressed mixed TCR dimers nor bound/killed normal cells in vitro. These results indicate that TCRγ4δ1-engineered αβT cells are a promising effector in ACT for cancer therapy.

Materials and Methods

Cells

Human tumor cell lines (HepG2, BGC-803, K562, Raji, fetal liver cells ccc-HEL-1, adrenal cortical reticular epithelial cells 1308.1.86, renal epithelial cell line 293T) were obtained from the American Type Culture Collection (ATCC). HepG2, BGC-803, ccc-HEL-1, 1308.1.86 and 293T cells were cultured in Dulbecco’s modified Eagle’s medium (Gibco, USA) supplemented with 10% fetal bovine serum (FCS), 100 U/mL penicillin-streptomycin, 1 mmol/L L-glutamine and 1 mmol/L sodium pyruvate. K562 and Raji cells were maintained in RPMI-1640 medium (Gibco) supplemented with 10% FCS, 100 U/mL penicillin-streptomycin, 1 mmol/L L-glutamine and 1 mmol/L sodium pyruvate. Human PBMCs, from healthy donors who provided informed consent, were isolated by density gradient centrifugation on Ficoll-Hypaque (TianjinHaoyang, China) and cultured in RPMI-1640 medium supplemented with 10% FCS and 200 IU/mL recombinant human interleukin-2 (IL-2).

Construction of Lentiviral Vectors and Production of Lentiviral Supernatant

The full-length sequence of TCRγ4 and TCRδ1 chains with a tumor antigen-specific CDR3 region identified from TILs of gastric carcinoma tissues was been previously described. (18) Briefly, the V region of TCRδ1 was amplified using cDNA from the TILs of gastric carcinoma tissues as a template with Vδ1- and Cδ1-specific primers, and subcloned into a pGEM-T easy vector for sequence analysis. We identified one high-frequency CDR3δ1-specific binding to gastric carcinoma tissues as a critical region (termed GTM). Finally, full-length TCRδ1 with GTM and TCRγ4 chains were amplified from cDNA of gastric tumor-derived γδTILs by PCR using full-length TCRδ1 and TCRγ4 primers directed at 5′-end region and 3′-end region, then cloned into pREP9 and pREP7 vectors for sequence analysis. After sequence was identified, especially for GTM, the TCRδ1 and TCRγ4 genes were cloned individually and co-cloned into pCDH vectors containing the marker genes copGFP to obtain pCDH-TCRδ1, pCDH-TCRγ4 and pCDH-TCRγ4δ1. These recombinant lentiviral vectors and pCDH vector were co-transfected into 293T cells with psPAX2 and PMD2.G vectors, respectively, using Lipofectamine 2000 (Invitrogen, USA). The lentiviral supernatant was collected, concentrated and stored at −80°C.

Generation of αβT Cells and Lentiviral Transfer

The γδT cells of PBMCs were removed by positive selection using FITC-labeled γδT-cell-specific immunomagnetic beads (Miltenyi, Germany). PBMCs without γδT cells were stimulated for 72 h with anti-CD3 and anti-CD28 in the presence of 200 IU/mL IL-2. The αβT cells were transfected with TCRγ4δ1-lentivirus, TCRδ1-lentivirus, TCRγ4-lentivirus and mock-lentivirus, using 10 ug/mL polybrene (Sigma, USA) and then cultured in RPMI-1640 with 10% FCS plus 400 IU/mL IL-2 for 7 d. TCRγ4δ1, TCRδ1 and TCRγ4 surface molecule staining was performed separately with anti-human TCRαβ-PE and anti-human TCRγδ-APC antibodies (Biolegend, USA), anti-human Vδ1-FITC antibody (Thermo, USA), and rabbit anti-human TCRγ primary antibody (Santa Cruz, USA) and goat anti-rabbit IgG-FITC secondary antibody (ZSGB-bio, China). Immunofluorescence was measured with an Accuri 6 Flow cytometer and analyzed by BD Accuri C6 software.

Binding of TCRγ4δ1-Fc to Tested Cells

TCRγ4δ1-Fc was constructed by fusing the complete extracellular domain of TCRγ4δ1 to the constant region of human IgG1 (18) and expressed by Sino Biological Inc. For flow cytometry: To examine the binding of TCRγ4δ1-Fc with various cells, including HepG2, BGC-803, K562, Raji, PBMCs, ccc-HEL-1, 1308.1.86 and 293T cells, the tested cells were individually incubated with TCRγ4δ1-Fc for 60 min at 4°C. FITC-conjugated goat anti-human IgG antibody (ZSGB-bio, China) was added and incubated for 30 min at 4°C; human IgG-Fc was used as a control. Immunofluorescence was measured by an Accuri 6 Flow cytometer (BD, USA) and analyzed using BD Accuri C6 software. Confocal microscopy: Tumor cells including HepG2, BGC-803, K562 and Raji were plated on slides with or without polylysine overnight and fixed with 4% cold paraformaldehyde. Fixed cells were incubated with TCRγ4δ1-Fc protein for 30 min at 4°C. FITC-conjugated goat anti-human IgG antibody was then added and incubated for 30 min at 4°C, human IgG-Fc was used as the control. Slides were examined with a confocal laser microscope (Leica, USA).

Cytokine Production and Proliferation Capacity

Cytokine production: Flow cytometry was used to determine the expression of IL-2 and IFN-γ in TCRγ4δ1-engineered αβT cells against target cells including HepG2, PBMCs, 1308.1.86, ccc-HEL-1 and 293T cells. Effector cells comprised TCRγ4δ1-engineered αβT cells, mock-engineered αβT cells, TCRγ4δ1-engineered αβT cells with resting treatment and mock-engineered αβT cells with resting treatment. Resting effector cells were cultured in RPMI-1640 medium with 5% FCS for 24 h. PBMCs as target cells were labeled by carboxyfluorescein diacetate N-succinimidyl ester (CFSE). Effector cells were incubated with target cells at an effector-to-target ratio of 3:1 in the presence of Brefeldin A (BFA) (Biosciences, USA) for 8 h. Cells were fixed and permeabilized with 1 × cytoFix/cytoPerm buffer (BD, USA) for 30 min, and then stained with anti-human IL-2-PE or anti-human IFN-γ-APC (Biolegend, USA) for 30 min in the dark. Cells were washed with 1X permeabilization/wash buffer and analyzed on an Accuri 6 flow cytometer. Flow cytometry data were based on gating at the effector cell population. Data analysis was performed with BD Accuri C6 software. Proliferation: Forty-eight-well plates were treated with 1 µg/mL anti-TCRγδ antibody for 2 h at 37°C. After washing, 5 X 105 mock- and TCRγ4δ1-engineered αβT cells were plated separately in RPMI-1640 medium with 200 IU/mL IL-2 for 8 h. Cells were then harvested and stained with anti-human Ki67 PEcy5 (BD, USA) according to the intracellular cytokine staining protocol and analyzed by flow cytometry.

Cytotoxicity

HepG2, BGC-803, K562, Raji, PBMCs, 1308.1.86, ccc-HEL-1 and 293T as target cells were added to 96-well plates at a density of 1 × 104 per well. Transfected effector cells including mock- and TCRγ4δ1-engineered αβT cells were incubated with target cells at an effector-to-target ratio of 10:1 for 6 h; each condition was plated in triplicate. The three controls were background group, spontaneous release group and maximal release group. We detected the cytotoxicity by a lactate dehydrogenase (LDH) assay, using the Cytotox 96 (Promega, USA) nonradioactive cytotoxicity assay reagent kit instructions. Cytotoxicity was calculated using the following formula:

$$\% \;{\rm{of}}\;{\rm{cytotoxicity}}\;{\rm{ = }}\;{{({\rm{experimental}}\;{\rm{release }} - {\rm{ spontaneous}}\;{\rm{release)}}} \over {({\rm{total}}\;{\rm{release }} - {\rm{ spontaneous release)}}}} \times \;100$$

Antitumor activity of TCRγ4δ1-engineered αβT cells in vivo. Mice: Animal experiments were performed according to the Animal Use Committee guidelines of the Experiment Animal Center, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences. Athymic BALB/c nu/nu mice (female, 4–6 wks) were obtained from the Laboratory Animal Center of the Chinese National Institute for the Control of Pharmaceutical and Biological Products, and housed under specific pathogen-free condition. Animal tumor model: To investigate the antitumor activity of TCRγ4δ1-engineered αβT cells in vivo, approximately 2 × 106 HepG2 cells in 100 µL of PBS was subcutaneously injected into the rear right flank of BALB/c nu/nu mice. After the tumor grew to approximately 100 mm3, mice bearing HepG2 tumors were randomly divided into 2 groups and intratumorally injected with TCRγ4δ1-engineered αβT cells (1 X 106, n = 8) or mock-engineered αβT cells (1 X 106, n = 8) every 3 d a total of 4 times. Tumors were measured with an electronic caliper every 3 d, and the tumor volumes were calculated using the following formula:

Tumor volume (mm3) = (long diameter, mm) × (short diameter, mm)2 ÷ 2

The tumor growth inhibition ratio (D%) was estimated according to the following formula: D% = (Vc − Vt) / Vc × 100%

Vc and Vt represent the means of tumor volumes of the control and treatment mice, respectively. On the last day of the experiment, the animals were euthanized by the cervical vertebra exarticulation method, and the tumors were excised and photographed.

Statistical Analysis

Data from the in vitro experiments are presented as the mean ± standard deviation (SD). Analysis of variance and independent samples t-test were used to analyze data. For in vivo experiments, the tumor volume was assessed by analysis of variance and paired t-test, and the data are presented as the mean ± SEM. p value < 0.05 is regarded as significant.

Results

The Lentiviral Vector Efficiently Transduced TCRγ4δ1 into Peripheral Blood-Derived αβT Cells

We previously identified a high-frequency CDR3 dominant sequence (CDR3δ1: CAFLPHADKLIFGKG), termed GTM, in TCRδ1 chain from TILs in human gastric cancer through RT-PCR and analysis of a large number of CDR3δ1 sequences. We confirmed that the CDR3δ1 peptide played a crucial role in tumor antigen recognition and bound to a wide variety of tumor cell lines and tissues similar to intact TCRγ4δ1. (18) The full-length TCRδ1 with GTM and TCRγ4 chains were amplified from cDNA of a gastric tumor-derived γδTILs by PCR and paired to form TCRγ4δ1 (18) (Figure 1A). Using the amplified TCRδ1 and TCRγ4 DNAs from pREP9-TCRδ1 with GTM and pREP7-TCRγ4, we constructed pCDH-TCRγ4δ1, a lentiviral vector harboring the TCRδ1 and TCRγ4 chain DNAs. The TCRγ4 gene was controlled by the cytomegalovirus (CMV) promoter (Figure 1B).

Figure 1
figure 1

The lentiviral vector efficiently transduced TCRγ4δ1 into peripheral blood-derived αβT cells. (A) Schematic diagram of TCRγ4δ1 receptor. The V region of TCRδ1 was amplified using RNA from the γδTILs of gastric carcinoma tissues by RT-PCR. A high-frequency CDR3δ1 sequence, termed GTM, was identified by sequence analysis. The full-length TCRδ1 with GTM and TCRγ4 chains were amplified from cDNA of one case of gastric tumor-derived γδTILs by PCR and paired to form TCRγ4δ1. (B) Schematic diagram of the lentiviral vector containing the TCRδ1 and TCRγ4 genes. The TCRδ1 and TCRγ4 genes were amplified by PCR and co-cloned into pCDH lentiviral vector. (LTR, long terminal repeats; CMV, cytomegalovirus promoter.) (C) The cell-surface expression of TCRγ4δ1 in αβT cells. Peripheral blood-derived αβT cells were lentivirally transduced with either mock lentiviral vectors or lentiviral vectors with TCRγ4δ1, then these cells were separately stained with APC-conjugated anti-TCRγδ and PE-conjugated anti-TCRαβ mAbs and analyzed by flow cytometry. The percentage of positive cells for each quadrant is indicated. (TCR, T-cell receptor; CDR, complementary determining region; TIL, tumor-infiltrating lymphocyte.)

To examine whether peripheral blood-derived αβT cells can be lentivirally engineered with TCRγ4δ1, a lentiviral vector with TCRγ4δ1 was transduced into pre-activated peripheral blood-derived αβT cells. TCRγ4δ1 expression was determined by staining with specific TCRγδ and TCRαβ antibodies. Approximately 36% of polyclonally activated αβT cells expressed TCRγ4δ1 (Figure 1C), indicating that TCRγ4δ1s were efficiently transduced.

TCRγ4δ1-engineered αβT cells were cytotoxic to several tumor cells in vitro. To assess binding of TCRγ4δ1 to tumor cells in vitro, we determined the binding capacity of TCRγ4δ1-Fc fusion proteins to various tumor cells by flow cytometry and confocal microscopy. Results show significant binding activity with HepG2, BGC-803 and K562 tumor cells, with binding rates of 94.23%, 91.46% and 72.05%, respectively (Figure 2A and 2B). No binding was observed in Raji tumor cells.

Figure 2
figure 2

Cytotoxicity of TCRγ4δ1-engineered αβT cells to tumor cells in vitro. (A) Flow cytometry profiles of the binding of TCRγ4δ1-Fc fusion protein to the tumor cell lines HepG2, BGC-803, K562 and Raji. After incubation with TCRγ4δ1-Fc, tested cells were stained with FITC-conjugated goat anti-human IgG and assessed by flow cytometry. (B) Confocal images of the binding of the TCRγ4δ1-Fc fusion protein to the tumor cell lines HepG2, BGC-803, K562 and Raji. After incubation with TCRγ4δ1-Fc, tested cells were stained with FITC-conjugated goat anti-human IgG and detected by laser scanning confocal microscopy. (C) Cytolytic activity of TCRγ4δ1-engineered αβT cells against tumor cells. TCRγ4δ1-engineered αβT cells were incubated with HepG2, BGC-803, K562 and Raji target cells at an effector-to-target ratio of 10:1 for 6 h. Cytotoxicity was analyzed by the LDH cytotoxicity detection kit. The data are representative of 3 independent experiments and expressed as the mean ± SD. *, P < 0.05; **, P < 0.01 (ns, no significance). (D) Expression of FasL and granzyme B in TCRγ4δ1-engineered αβT cells was determined by flow cytometry. Mock- and TCRγ4δ1-engineered αβT cells were cultured with HepG2 at an effector-to-target ratio of 10:1 for 6 h, and the cellular surface molecule FasL and intracellular granzyme B were analyzed by flow cytometry. A representative FCM analysis of cytotoxic molecules is shown. (LDH, lactate dehydrogenase.)

LDH assay was performed to assess the cytolytic capacities of TCRγ4δ1-engineered αβT cells to tumor cells. We found significant cytotoxicity to HepG2 (55.78 ± 10.39%) and BGC-803 (67.36 ± 5.30%) tumor cells, and less activity against K562 tumor cells (41.33 ± 1.28%) at an effector-to-target ratio of 10:1. In comparison, mock-engineered αβT cells had significantly less cytotoxicity against HepG2 22.26 ± 12.56% (p = 0.024), BGC-803 32.56 ± 15.01% (p = 0.019) and K562 19.29 ± 5.25% (p = 0.002) (Figure 2C). Raji cells were not lysed and served as a negative control. We found that the cytotoxicity of TCRγ4δ1-engineered αβT cells was proportionate to the binding ability of TCRγ4δ1-Fc to tumor cells. This indicates that TCRγ4δ1-engineered αβT cells exert cytolytic activity against HepG2, BGC-803 and K562 tumor cells in a TCRγ4δ1-dependent manner.

Cytotoxicity of TCRγ4δ1-engineered αβT cells to tumor cells is related to perforin-granzyme. Previous reports showed that T cells kill tumors through 2 main mechanisms, Fas-FasL and perforin-granzyme pathways. Fas-FasL is involved in the transmission of cell death signaling via binding of Fas molecules on the tumor cell surface with ligands on T cells. Perforin-granzyme involves synergy between perforin and granzyme molecules released by T cells. Granzyme B is more abundant, with higher enzyme activity, compared with other types of granzymes. (20,21) Thus, we measured the expression of FasL and granzyme B by flow cytometry on mock- and TCRγ4δ1-engineered αβT cells following incubation with HepG2 cells for 6 h. We found no significant difference in FasL expression between the 2 groups. Results show that the percentage of TCRγ4δ1-engineered αβT cells expressing granzyme B was 40.66%, significantly higher than the control group (25.66%; Figure 2D). These data suggest that TCRγ4δ1-engineered αβT cells kill tumor cells via the perforin-granzyme pathway.

TCRγ4δ1-engineered αβT cells produced cytokines IL-2 and IFN-γ when stimulated by HepG2 tumor cells. The cytokines IL-2 and IFN-γ play important roles in antitumor activity. (22,23) We analyzed the IL-2 and IFN-γ expression of resting and nonresting TCRγ4δ1-engineered αβT cells and mock-engineered αβT cells by flow cytometry after incubation with HepG2 tumor cells and treatment with BFA for 6 h. Results show that the percentage of restingTCRγ4δ1-engineered αβT cells expressing IL-2 and IFN-γ was 30.38% and 31.56%, respectively, higher than control groups (9.49% and 10.43%, respectively; Figure 3A). The percentage of nonresting TCRγ4δ1-engineered αβT cells producing IL-2 and IFN-γ was 33.66% and 31.16%, respectively, higher than the control group (14.63% and 10.78%, respectively; Figure 3B). Two different statuses of TCRγ4δ1-engineered αβT cells produced similar amounts of cytokines. In addition, TCRγ4δ1-engineered αβT cells expressed Ki67, a proliferation marker, significantly higher than control after stimulation with anti-γδTCR antibody (Figure 3C). These findings indicate that TCRγ4δ1-engineered αβT cells produce cytokines and proliferate similar to γδT cells.

Figure 3
figure 3

Cytokine production and proliferative capacity of TCRγ4δ1-engineered αβT cells. (A) Expression of IL-2 and IFN-γ in resting TCRγ4δ1-engineered αβT cells. Resting mock- and TCRγ4δ1-engineered αβT cells were incubated with HepG2 at an effector-to-target ratio of 3:1 for 6 h with BFA, and the expression of intracellular IL-2 and IFN-γ was determined by flow cytometry. (B) Expression of IL-2 and IFN-γ in the nonresting TCRγ4δ1-engineered αβT cells. The nonresting mock- and TCRγ4δ1-engineered αβT cells were incubated with HepG2 at an effector-to-target ratio of 3:1 for 6 h with BFA, and expression of intracellular IL-2 and IFN-γ was determined by flow cytometry. (C) Expression of intracellular Ki67 in TCRγ4δ1-engineered αβT cells. Mock- and TCRγ4δ1-engineered αβT cells were stimulated with plate-bound anti-TCRγδ for 6 h, and the expression of Ki67 was examined by flow cytometry. Shown is a representative FCM analysis of cytokines and Ki67. (IL, interleukin; IFN, interferon; BFA, brefeldin A.)

TCRγ4δ1-engineered αβT cells neither expressed mixed TCR dimers nor bound/killed normal cells in vitro. Previous studies have shown that the TCR receptor is transported to the surface of T cells after the formation of complete TCR dimers-CD3 complex. (24) Thus, we transduced TCRγ4, or TCRδ1 chains alone, into αβT cells and analyzed surface expression. TCRγ4δ1-engineered αβT cells served as a positive control. We found no TCRγ4 or TCRδ1 expression on the surface of TCRγ4- or TCRδ1-engineered αβT cells. In contrast, TCRγ4δ1 was expressed on the surface of TCRγ4δ1-engineered αβT cells (Figure 4A). These data suggest that the introduced TCRγ4 or TCRδ1 chains did not pair with the endogenous TCRα or TCRβ chains to generate mixed TCR dimers. This feature would be beneficial in reducing the risk of autoimmunity. We also assessed the binding activity of TCRγ4δ1-Fc and the cytotoxicity of TCRγ4δ1-engineered αβT cells to some normal cells by flow cytometry and LDH assay. We found that TCRγ4δ1-Fc did not bind to PBMCs, 1308.1.86, ccc-HEL-1 or 293T cells (Figure 4B). The cytotoxicity of TCRγ4δ1-engineered αβT cells toward PBMCs, 1308.1.86, ccc-HEL-1 and 293T cells was 2.79% ± 2.04%, 8.11% ± 3.52%, 11.57% ± 1.89% and 15.53% ± 0.68% at an effector-to-target ratio of 10:1, respectively, compared with 4.04% ± 0.54% (p = 0.365), 9.10% ± 6.14% (p = 0.821), 8.13% ± 3.17% (p = 0.181) and 15.44% ± 0.30% (p = 0.850) of mock-engineered αβT cells, respectively (Figure 4C). The cytotoxicity of TCRγ4δ1-engineered αβT cells was in accordance with the binding ability of TCRγ4δ1-Fc to normal cells. We further evaluated the ability of mock- or TCRγ4δ1-engineered αβT cells to produce IFN-γ in response to normal cells by flow cytometry. No significant difference was observed between the 2 groups (Figure 4D). Taken together, these results indicate that TCRγ4δ1-engineered αβT cells lack an autoimmune response to several normal cells in vitro.

Figure 4
figure 4

Determination of TCRγ4 or δ1 expression and binding/activity to normal cells of TCRγ4δ1-engineered αβT cells. (A) Cell surface expression of TCRδ1 and TCRγ4 chains on the TCRδ1- or TCRγ4-engineered αβT cells. TCRδ1- and TCRγ4-engineered αβT cells were separately stained with anti-human Vδ1-FITC antibody, and with rabbit anti-human TCRγ primary antibody and goat anti-rabbit IgG-FITC secondary antibody, and then assessed by flow cytometry. TCRγ4δ1 expression on the surface of TCRγ4δ1-engineered αβT cells served as a positive control. (B) Binding of TCRγ4δ1-Fc to normal cells including PBMCs, 1308.1.86, ccc-HEL-1 and 293T cells. After incubation with TCRγ4δ1-Fc, these normal cells were stained with FITC-conjugated goat anti-human IgG and then evaluated by flow cytometry. (C) Cytotoxicity of TCRγ4δ1-engineered αβT cells to PBMCs, 1308.1.86, ccc-HEL-1 and 293T. TCRγ4δ1-engineered αβT cells were incubated with these normal cells at an effector-to-target ratio of 10:1 for 6 h, and the cytolytic activity was detected by LDH assay. The data are representative of 3 independent experiments and expressed as the mean ± SD (ns, no significance). (D) Expression of IFN-γ in the TCRγ4δ1-engineered αβT cells. Mock- and TCRγ4δ1-engineered αβT cells were individually incubated with PBMCs, 1308.1.86, ccc-HEL-1 and 293T at an effector-to-target ratio of 3:1 for 6 h with BFA, and expression of intracellular IFN-γ was determined by flow cytometry. Shown is a representative FCM analysis of the IFN-γ and TCR chains. (PBMC, peripheral blood mononuclear cell.)

TCRγδ1-engineered αβT cells displayed antitumor activity in vivo. To assess the antitumor effect of TCRγ4δ1-engineered αβT cells in vivo, mice were subcutaneously injected with 2 × 106 HepG2 cells in the rear right flank. After the tumor grew to approximately 100 mm3, mock- and TCRγ4δ1-engineered αβT cells were intratumorally injected into nude mice every 3 d a total of 4 times. Tumors were measured by a caliper every 3 d, starting on d 0. The tumor growth curve and the excised tumors show that TCRγ4δ1-engineered αβT cells had significantly higher tumor inhibiting effect compared with the control group (P = 0.001; Figures 5A and C). The tumor growth inhibition ratio was calculated, and its curve is illustrated in Figure 5B. The tumor growth inhibition ratio was above 60% after d 9 and reached 73.26% at d 27. TCRγ4δ1-engineered αβT cells significantly retarded the tumor growth of human HepG2 tumor cells in a nude mouse model.

Figure 5
figure 5

Antitumor activity of TCRγ4δ1-engineered αβT cells in vivo. Mice were subcutaneously injected with 2 × 106 HepG2 cells in the rear right flank. When tumor volume reached approximately 100 mm3, mock- and TCRγ4δ1-engineered αβT cells were injected intratumorally every 3 d a total of 4 times, and the tumor volume in the right flank of each mouse was measured every 3 d until 30 d. (A) Mice treated with TCRγ4δ1-engineered αβT cells exhibited significant suppression of tumor volume compared with mice treated with mock-engineered αβT cells (P = 0.001). Black arrows indicate the treatment time; each point represents the mean ± SEM, **, P < 0.01. (B) The tumor growth inhibition ratio was estimated. (C) Image of the excised tumors in the experiment evaluating the influences of TCRγ4δ1-engineered αβT cells on antitumor activity in HepG2 tumor-bearing nude mice.

Discussion

In this study, αβT cells were redirected by lentiviral transduction of the TCRγ4δ1 gene to generate effective tumor-reactive T cells. To date, genetically engineered T cells have been developed via several tools to deliver genes into T cells, including the viral transfection system, electroporation and the transposase system. (25) Viral methods are common for gene delivery. We chose the lentiviral transduction system to avoid the drawbacks of a retroviral transfection method. First, lentiviruses mediate the integration of their genome into the DNA of the host dividing and nondividing cells, whereas retroviruses only infect dividing cells. (2628) We found that lentiviral vector efficiently transduced TCRγ4δ1 genes into αβT cells. Approximately 36% of polyclonally activated αβT cells were lentivirally transduced with TCRγ4δ1. Second, lentiviruses have a lower risk of insertional mutagenesis compared with retroviruses. Lentiviruses preferentially concentrate in high gene density regions and integrate into transcriptional units, while retroviruses prefer to insert in the neighborhood of transcription start sites of active genes. (2931) Third, lentiviral vectors are less prone to activate adjacent genes compared with retroviral vectors. (32,33)

We used peripheral blood-derived αβT cells as targets for transduction of TCRγ4δ1 based on several considerations. First, the efficiency of TCRγ4δ1-surface expression is controlled by formation of the TCR-CD3 complex, and recognition signaling of TCRγ4δ1 and tumor antigens is delivered to intracellular structures in a CD3-dependent manner. CD3 molecules are naturally expressed in the αβT cells. Thus, it is not necessary to redirect TCRγ4δ1-engineered αβT cells with the CD3 gene. (24) Second, transfer of TCRγδ to αβT cells, or transfer of TCRαβ to γδT cells, generates a large quantity of tumor-reactive T cells without the expression of mixed TCR dimers, avoiding an autoimmune response. (9,17) Therefore, TCRγ4δ1-engineered αβT cells avoid the expression of potentially harmful mixed TCR dimers. Formation of TCR dimers-CD3 complex is required for TCR transport to the surface of T cells. Here, we found that TCRδ1 and TCRγ4 chains were not expressed on the surface of TCRδ1- and TCRγ4-transduced αβT cells, indicating that the introduced TCRδ1 and TCRγ4 chains did not pair with endogenous TCRα or TCRβ chains, which could reduce the risk of autoimmunity. Third, the main aim of the construction of TCR-modified T cells was to obtain a sufficient number of tumor-reactive T cells for cancer immunotherapy. The αβT cells, as genetic recipients, comprise 42% to 69% of PBMCs and rapidly expand upon stimulation of anti-CD3 and anti-CD28 antibodies. We observed that TCRγ4δ1-engineered αβT cells significantly proliferated upon stimulation of anti-γδTCR antibody. These data support our design of using αβT cells as the targets of TCRγ4δ1 transduction.

T cells mediate antitumor reactivity in a TCR-dependent manner, driven by the binding of TCR with tumor antigen. Therefore, it is of utmost importance to screen for T cell receptors with a high affinity for tumor antigen and a broad spectrum of tumor recognition. Previously, we identified a high-frequency CDR3 sequence in the TCRδ1 chain from TILs in human gastric cancer and found that this CDR3 was crucial to antigen recognition. We also found that TCRγ4δ1-Fc fusion proteins had a strong binding ability to the tumor-associated antigens MICA and UL-16 binding protein 5 (ULBP5) via ELISA and SPR assays. (18) These results suggest that TCRγ4δ1 possess a high affinity to antigen recognition. In addition, multiple tumor-expressed antigens, such as MICA, MHC class I chain-related molecules B (MICB) and various ULBPs, are recognized by the TCR of human Vδ1 T cells and are constitutively expressed at variable levels on many epithelial tumor cells and some leukemias and lymphomas. (19,34,35) We therefore explored the binding ability of TCRγ4δ1 and a variety of tumors, particularly epithelial tumors. Our previous study reported that TCRγ4δ1 displayed strong binding to BGC-823, G401, GLC-82, HT29 and SKOV3 cells and tissues. (18) In this study, we found consistent results showing that TCRγ4δ1s also significantly bound other tumor cells, including HepG2, BGC-803, K562 (Figures 2A and B). Our results support the notion that TCRγ4δ1 recognizes a large panel of tumors, especially epithelium-derived tumors. In summary, TCRγ4δ1 has a broad spectrum of tumor recognition.

Tumor-infiltrating Vδ1 T cells and peripheral blood-derived Vδ1 T cells exerted remarkable cytotoxicity against epithelium-derived tumors and some blood-derived tumors, such as renal carcinoma, colorectal cancer, pancreatic cancer and lymphoid leukemia. (34,36,37) However, the proliferative capacity and function of γδ T cells are significantly impaired in cancer patients. (38) Thus, TCRγ4δ1-engineered αβT cells might be an attractive alternative strategy to kill tumors. Here, we found that TCRγ4δ1-engineered αβT cells displayed striking cytolytic activity against HepG2, BGC-803 and K562 in a TCRγδ-dependent manner. Importantly, adoptive transfer of TCRγ4δ1-engineered αβT cells into nude mice bearing a human HepG2 cell line significantly suppressed tumor growth. These findings display a similar effect when compared with previous reports showing that TCRγ9δ2-modified αβT cells/PBMCs exerted cytotoxicity against a variety of tumors. (9,10) However, Vδ1 γδT cells reside preferentially in the intestine and in the skin epithelium, and their Vδ1 TCRs recognize protein antigens, notably MICA and ULBP3, which are generally expressed on epithelial tumor cells. In contrast, Tγ9δ2 cells mainly appear in the blood and their TCRγ9δ2 recognizes phosphoantigens. (39) This suggests that TCRγ4δ1-engineered αβT cells preferentially kill epithelial tumors compared with TCRγ9δ2-engineered αβT cells/PBMCs.

Furthermore, we showed that the transduced TCRγ4 or TCRδ1 chain gene did not pair with the endogenous TCRα or TCRβ chains to generate new mixed TCR heterodimers. This is advantageous in reducing the risk of autoimmunity. TCRγ4δ1 primarily recognizes stress-induced molecules, such as MICA and ULBPs. The expression of these stress-induced molecules is upregulated under heat shock, viral or mycobacterial infection, or oxidative stress. (4042) Expression of MICA and MICB on epithelial cells can be stress-induced to regulate protective responses through the Vδ1 γδ T cells. (43) Thus, cancer patients with severe inflammation should avoid adoptive TCRγ4δ1-engineered αβT cell transfer therapy unless inflammation is eliminated. These stress-induced molecules are rarely expressed on normal cells under physiological conditions. Most normal human tissues do not express MICA, but glandular and gastric epithelial cells do, which occurred in the intracellular location and was not transported to the cell surface. (44,45) Here, we show that PBMCs, ccc-HEL-1, 1308.1.86 and 293T normal cells did not induce any activation of TCRγ4δ1-engineered αβT cells in a TCRγδ-dependent manner, measured by IFN-γ expression and cytotoxicity. These analyses support the hypothesis that TCRγ4δ1-engineered αβT cells do not attack normal cells through TCRγ4δ1 recognition, implying that TCRγ4δ1 possesses a high specificity for tumors. Although we cannot preclude autoimmunity resulting from allologous TCRαβ recognition or complicated human physiological conditions in vivo, TCRγ4δ1-engineered αβT cells are a relatively safe strategy for cancer therapy. Our further research will focus on knockdown of allologous TCRαβ receptors and in vivo autoimmunity.

TCRγ4δ1-engineered αβT cells are effective killer cells that express much IL-2 and IFN-γ upon incubation with HepG2 cells. IL-2 displays pleiotropic effects on the immune system, promoting T cell proliferation and differentiation, downregulation of IL-2-induced expansion of regulatory T cells, and inhibition of tumor growth. (46,47) IFN-γ secretion enhances the immunogenicity of tumors by increasing MHC expression on tumor cells and TCR recognition of tumor cells, and inhibits the induction of CD4 + CD25 + regulatory T cells, providing a positive feedback loop for antitumor reactivity. Importantly, IFN-γ is beneficial for the suppression of tumor cells in vivo. (23,48,49)

The main challenge of this study was to improve the transduction rate and expression level of TCRγ4δ1 in αβT cells. Previous reports suggested that the creation of high-avidity TCR-engineered T cells was determined by the TCR cell-surface makeup of T cells, and that transport of the TCR to the plasma membrane depended on complete assembly of the TCR/CD3 complex. (24,50) In TCRγ4δ1-engineered αβT cells, competition between TCRγ4δ1 and TCRαβ for association with CD3 could restrict their expression levels on the cell surface. Thus, the upregulation of TCRγ4δ1 cell surface expression is advantageous in reducing the surface expression of endogenous TCRαβ. In summary, the increased transduction rate and expression level of TCRγ4δ1 in αβT cells will enhance the efficacy of TCRγ4δ1-engineered αβT cells and decrease the risk of autoimmunity resulting from allologous TCRαβ recognition.

Conclusion

Our results highlight a novel TCRγδ-engineering αβT cell-effector ACT therapy for solid tumors, with safety and strong antitumor efficacy, and with the clinical potential to treat cancer patients.

Disclosure

The authors declare that they have no competing interests as defined by Molecular Medicine, or other interests that might be perceived to influence the results and discussion reported in this paper.

References

  1. Maude SL, et al. (2014) Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371:1507–17.

    Article  Google Scholar 

  2. Rosenberg S, Spiess P, Lafreniere R. (1986) A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. Science. 233:1318–21.

    Article  CAS  Google Scholar 

  3. Rosenberg SA, et al. (2011) Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 17:4550–7.

    Article  CAS  Google Scholar 

  4. Ho WY, Blattman JN, Dossett ML, Yee C, Greenberg PD. (2003) Adoptive immunotherapy: engineering T cell responses as biologic weapons for tumor mass destruction. Cancer Cell 3:431–7.

    Article  CAS  Google Scholar 

  5. Sadelain M, Rivière I, Brentjens, R. (2003) Targeting tumours with genetically enhanced T lymphocytes. Nat. Rev. Cancer. 3:35–45.

    Article  CAS  Google Scholar 

  6. Tran E, et al. (2014) Cancer immunotherapy based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344:641–5.

    Article  CAS  Google Scholar 

  7. Cao W, He W. (2014) The recognition pattern of gamma/delta T cells. Forties in Biosciences. 10:2676–700.

    Article  Google Scholar 

  8. Kabelitz D, Wesch D, He W. (2007) Perspectives of γδT Cells in Tumor Immunology. Cancer Res. 67:5–8.

    Article  CAS  Google Scholar 

  9. Marcu-Malina V, et al. (2011) Redirecting alphabeta T cells against cancer cells by transfer of a broadly tumor-reactive gammadelta T-cell receptor. Blood. 118:50–9.

    Article  CAS  Google Scholar 

  10. Zhao H, Xi X, Cui L, He W. (2012) CDR3δ-grafted γ9δ2T cells mediate effective antitumor reactivity. Cell Mol. Immunol. 9:147–54.

    Article  CAS  Google Scholar 

  11. Hiasa A, et al. (2008) Long-term phenotypic, functional and genetic stability of cancer-specific T-cell receptor (TCR) αβ genes transduced to CD8+ T cells. Gene Ther. 15:695–99.

    Article  CAS  Google Scholar 

  12. Bendle GM, et al. (2010). Lethal graft-versus-host disease in mouse models of T cell receptor gene therapy. Nat. Med. 16(5): 565–70, 561p following 570.

    Article  CAS  Google Scholar 

  13. Daniel-Meshulam I, Ya’akobi S, Ankri C, Cohen CJ. (2012). How (specific) would like your T-cells today? Generating T-cell therapeutic function through TCR-gene transfer. Front/Immunol. 3:186.

    Google Scholar 

  14. Govers C, Sebestyen Z, Coccoris M, Willemsen RA, Debets R. (2010). T cell receptor gene therapy: strategies for optimizing transgenic TCR pairing. Trends Mol. Med. 16(2): 77–87.

    Article  CAS  Google Scholar 

  15. Koning F, Maloy WL. (1987) Independent association of T cell receptor beta and gamma chains with CD3 in the same cell. J. Exp. Med. 166:595–600.

    Article  CAS  Google Scholar 

  16. Saito T, Hochstenbach F. (1988) Surface expression of only gamma delta and/or alpha beta T cell receptor heterodimers by cells with four (alpha, beta, gamma, delta) functional receptor chains. J. Exp. Med. 168:1003–20.

    Article  CAS  Google Scholar 

  17. Hiasa A, et al. (2009) Rapid αβ TCR-mediated responses in γδ T cells transduced with cancer-specific TCR genes. Gene Ther. 16:620–8.

    Article  CAS  Google Scholar 

  18. Jiang Y, Guo Y, Xi X, Cui L, He W. (2011) Flanking V and J sequences of complementary determining region 3 of T cell receptor (TCR) delta1 (CDR3delta1) determine the structure and function of TCRgamma4delta1. J Biol. Chem. 286:25611–19.

    Article  CAS  Google Scholar 

  19. Groh V, et al. (1999) Broad tumor-associated expression and recognition by tumor-derived gamma delta T cells of MICA and MICB. Proc. Natl. Acad. Sci. U.S.A. 96:6879–84.

    Article  CAS  Google Scholar 

  20. Vignaux F, Golstein P. (1994) Fas-based lymphocyte-mediated cytotoxicity against syngeneic activated lymphocytes: a regulatory pathway? Eur. J. Immunol. 24:923–7.

    Article  CAS  Google Scholar 

  21. Trapani JA. (1995) Target cell apoptosis induced by cytotoxic T cells and natural killer cells involves synergy between the pore-forming protein, perforin, and the serine protease, granzyme B. Aust. N.Z. J. Med. 25:793–9.

    Article  CAS  Google Scholar 

  22. Rosenberg SA. (2014) IL-2: the first effective immunotherapy for human cancer. J. Immunol. 192:5451–8.

    Article  CAS  Google Scholar 

  23. Gao Y, et al. (2003) Gamma delta T cells provide an early source of interferon gamma in tumor immunity. J. Exp. Med. 198:433–42.

    Article  CAS  Google Scholar 

  24. Heemskerk MHM, et al. (2007) Efficiency of T-cell receptor expression in dual-specific T cells is controlled by the intrinsic qualities of the TCR chains within the TCR-CD3 complex. Blood. 109:235–43.

    Article  CAS  Google Scholar 

  25. Jena B., G. Dotti, Cooper L. J. (2010). Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood. 116(7): 1035–44.

    Article  CAS  Google Scholar 

  26. Kafri T, Blömer U, Peterson DA, Gage FH, Verma IM. (1997) Sustained expression of genes delivered directly into liver and muscle by lentiviral vectors. Nat. Genet. 17:314–17.

    Article  CAS  Google Scholar 

  27. Naldini L, et al. (1996) In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science. 272:263–7.

    Article  CAS  Google Scholar 

  28. Nguyen T, Oberholzer J. (2002) Highly efficient lentiviral vector-mediated transduction of nondividing, fully reimplantable primary hepatocytes. Mol. Ther. 6:199–209.

    Article  CAS  Google Scholar 

  29. Bobisse S, Zanovello P, Rosato A. (2007) T-cell receptor gene transfer by lentiviral vectors in adoptive cell therapy. Expert Opin. Biol. Ther. 7:893–906.

    Article  CAS  Google Scholar 

  30. Aiuti A, et al. (2013) Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science. 341:1233151.

    Article  Google Scholar 

  31. Montini E, et al. (2006) Hematopoietic stem cell gene transfer in a tumor-prone mouse model uncovers low genotoxicity of lentiviral vector integration. Nat. Biotechnol. 24:687–96.

    Article  CAS  Google Scholar 

  32. Hematti P, et al. (2004) Distinct genomic integration of MLV and SIV vectors in primate hematopoietic stem and progenitor cells. PLOS Biol. 2:e423.

    Article  Google Scholar 

  33. Mitchell RS, et al. (2004) Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLOS Biol. 2:E234.

    Article  Google Scholar 

  34. Maeurer MJ, Martin D. (1996) Human intestinal Vdelta1+ lymphocytes recognize tumor cells of epithelial origin. J. Exp. Med. 183:1681–96.

    Article  CAS  Google Scholar 

  35. Poggi A, Venturino C. (2004) Vdelta1 T lymphocytes from B-CLL patients recognize ULBP3 expressed on leukemic B cells and up-regulated by trans-retinoic acid. Cancer Res. 64:9172–9.

    Article  CAS  Google Scholar 

  36. Correia DV, et al. (2011) Differentiation of human peripheral blood Vdelta1+ T cells expressing the natural cytotoxicity receptor NKp30 for recognition of lymphoid leukemia cells. Blood. 118:992–1001.

    Article  CAS  Google Scholar 

  37. Choudhary A, et al. (1995) Selective lysis of autologous tumor cells by recurrent gamma delta tumor-infiltrating lymphocytes from renal carcinoma. J. Immunol. 154:3932–40.

    CAS  PubMed  Google Scholar 

  38. Wilhelm M, Kunzmann V. (2003) Gammadelta T cells for immune therapy of patients with lymphoid malignancies. Blood. 102:200–06.

    Article  CAS  Google Scholar 

  39. Kabelitz D, Kalyan S, Oberg H, Wesch D. (2013) Human Vδ2 versus non-Vδ2 γδ T cells in antitumor immunity. Oncoimmunology. 2:e23304.

    Article  Google Scholar 

  40. Groh V, et al. (1996) Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc. Natl. Acad. Sci. U.S.A. 93:12445–50.

    Article  CAS  Google Scholar 

  41. Qi J, Zhang J, Zhang S, Cui L, He W. (2003) Immobilized MICA could expand human Vdelta1 gammadelta T cells in vitro that displayed major histocompatibility complex class I chain-related A-dependent cytotoxicity to human epithelial carcinomas. Scand. J. Immunol. 58:211–20.

    Article  CAS  Google Scholar 

  42. Yamamoto K, Fujiyama Y. (2001) Oxidative stress increases MICA and MICB gene expression in the human colon carcinoma cell line (CaCo-2). Biochim. Biophys. Acta. 1526:10–12.

    Article  CAS  Google Scholar 

  43. Groh V, Steinle A. (1998) Recognition of stress-induced MHC molecules by intestinal epithelial gammadelta T cells. Science. 279:1737–40.

    Article  CAS  Google Scholar 

  44. Groh V, et al. (1996) Cell stress-regulated human major histocompatibility complex class I gene expressed in gastrointestinal epithelium. Proc. Natl. Acad. Sci. U.S.A. 93:12445–50.

    Article  CAS  Google Scholar 

  45. Hüe S, et al. (2004) A direct role for NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity. 21:367–77.

    Article  Google Scholar 

  46. Gong G, et al. (2009) Phosphoantigen-activated V gamma 2V delta 2 T cells antagonize IL-2-induced CD4+CD25+Foxp3+ T regulatory cells in mycobacterial infection. Blood. 113:837–45.

    Article  CAS  Google Scholar 

  47. Skrombolas D, Frelinger JG. (2014) Challenges and developing solutions for increasing the benefits of IL-2 treatment in tumor therapy. Expert Rev Clin Immunol. 10:207–17.

    Article  CAS  Google Scholar 

  48. Seki N, et al. (2002) Tumor-specific CTL kill murine renal cancer cells using both perforin and Fas ligand-mediated lysis in vitro, but cause tumor regression in vivo in the absence of perforin. J. Immunol. 168:3484–92.

    Article  CAS  Google Scholar 

  49. Nishikawa H, et al. (2005) IFN-gamma controls the generation/activation of CD4+ CD25+ regulatory T cells in antitumor immune response. J. Immunol. 175:4433–40.

    Article  CAS  Google Scholar 

  50. Clevers H, Alarcon B, Wileman T, Terhorst C. (1988) The T cell receptor/CD3 complex: a dynamic protein ensemble. Annu. Rev. Immunol. 6:629–62.

    Article  CAS  Google Scholar 

Download references

Acknowledgments

We greatly appreciate Dr Tingting Sun and Dr Yan Jiang for providing the lentiviral package system and TCRγ4δ1 genes, respectively, and Dr Chunyun Sun for help with the TCRγ4δ1-Fc fusion protein expression. This study was supported by the National Program for Key Basic Research Projects (2013CB530503), National Natural Science Foundation of China (81471574), the Mega-Projects of National Science Research for the 12th Five-Year Plan (2012ZX10001006) and the graduate innovation fund of Peking Union Medical College, China (2013100116). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. We thank Dr. Austin Cape at ASJ Editors for edits and comments.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wei He.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, and provide a link to the Creative Commons license. You do not have permission under this license to share adapted material derived from this article or parts of it.

The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this license, visit (http://creativecommons.org/licenses/by-nc-nd/4.0/)

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, K., You, H., Li, Y. et al. TCR γ4δ1-Engineered αβT Cells Exhibit Effective Antitumor Activity. Mol Med 22, 519–529 (2016). https://doi.org/10.2119/molmed.2016.00023

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2119/molmed.2016.00023

Keywords