The Role of Human γδ T Cells in Anti-Tumor Immunity and Their Potential for Cancer Immunotherapy
Abstract
:1. Introduction
2. Diversity of Human γδ T Cell Subsets
3. The Tumor Cell Recognition of γδ T Cells
3.1. Tumor Cell Recognition of Vγ9Vδ2 T Cells
3.2. Tumor Cell Recognition of Vδ1 T Cells
3.3. Tumor Cell Recognition of Vδ1−Vδ2− T Cells
4. Anti-Tumor Mechanism of γδ T Cells
5. γδ T Cell-Based Cancer Immunotherapy
Cell Type | Interventions | Cancers | Phase | Status | References or Study Registration |
---|---|---|---|---|---|
Vγ9Vδ2 T | BrHPP + IL-2 (in vitro) | RCC | I | Finished | [100] |
Vγ9Vδ2 T | 2M3B1PP + IL-2 (in vitro) | RCC | Pilot study | Finished | [106] |
Vγ9Vδ2 T | 2M3B1PP, ZOL + IL-2 (in vitro) | RCC | I | Finished | [104] |
Vγ9Vδ2 T | ZOL + IL-2 (in vitro) | NSCLC | I | Finished | [101] |
Vγ9Vδ2 T | ZOL + IL-2 (in vitro) | NSCLC | I | Finished | [102] |
Vγ9Vδ2 T | ZOL + IL-2 (in vitro) | MM | I | Finished | [105] |
Vγ9Vδ2 T | ZOL + IL-2 (in vitro) | Gastric cancer | Pilot study | Finished | [98] |
Vγ9Vδ2 T | ZOL + IL-2 (in vitro) | Metastatic solid tumors | I | Finished | [103] |
Vγ9Vδ2 T | Pamidronate + IL-2 (in vivo) | MM and NHL | Pilot study | Finished | [107] |
Vγ9Vδ2 T | ZOL + IL-2 (in vivo) | RCC, AML and melanoma | I/II | Finished | [108] |
Vγ9Vδ2 T | BrHPP + IL-2 (in vivo) | RCC, CRC and breast cancer | I | Finished | [99] |
Vγ9Vδ2 T | ZOL + IL-2 (in vivo) | RCC | Pilot study | Finished | [109] |
Vγ9Vδ2 T | ZOL + IL-2 (in vivo) | Breast cancer | I | Finished | [25] |
Vγ9Vδ2 T | ZOL + IL-2 (in vivo) | Neuroblastoma | I | Finished | [110] |
γδ T | In vitro expanded | Lymphoma and leukemia | Early phase 1 | Recruiting | NCT04028440 |
γδ T | In vitro expanded | Acute myeloid leukemia | I | Recruiting | NCT04008381 |
γδ T | Combined with surgery (in vitro expanded) | Pancreatic cancer | I/II | Finished | NCT03180437 |
γδ T | Combined with DC-CIK cells (in vitro expanded) | Breast cancer | I/II | Finished | NCT02418481 |
γδ T | Combined with surgery (in vitro expanded) | Breast cancer | I/II | Finished | NCT03183206 |
γδ T | Combined with DC-CIK cells (in vitro expanded) | Liver cancer | I/II | Finished | NCT02425735 |
γδ T | Combined with surgery (in vitro expanded) | Liver cancer | I/II | Finished | NCT03183219 |
γδ T | Combined with DC-CIK cells (in vitro expanded) | Lung cancer | I/II | Finished | NCT02425748 |
γδ T | Combined with surgery (in vitro expanded) | Lung cancer | I/II | Finished | NCT03183232 |
γδ T | In vitro expanded | HCC | I | Not yet recruiting | NCT04032392 |
γδ T | Anti-CD19-CAR | Leukemia and lymphoma | I | Not yet recruiting | NCT02656147 |
γδ T | NKG2DL-targeting CAR | Relapsed or refractory solid tumors | I | Not yet recruiting | NCT04107142 |
γδ T | TEG001 | AML and MM | I | Recruiting | NTR6541 |
6. New Strategies to Improve γδ T Cell-Based Cancer Immunotherapy
7. Conclusions and Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Carding, S.R.; Egan, P.J. Gammadelta T cells: Functional plasticity and heterogeneity. Nat. Rev. Immunol. 2002, 2, 336–345. [Google Scholar] [CrossRef] [PubMed]
- Chien, Y.H.; Meyer, C.; Bonneville, M. gammadelta T cells: First line of defense and beyond. Annu. Rev. Immunol. 2014, 32, 121–155. [Google Scholar] [CrossRef] [PubMed]
- Poggi, A.; Zocchi, M.R. γδ T Lymphocytes as a First Line of Immune Defense: Old and New Ways of Antigen Recognition and Implications for Cancer Immunotherapy. Front. Immunol. 2014, 5, 575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holtmeier, W.; Kabelitz, D. gammadelta T cells link innate and adaptive immune responses. Chem. Immunol. Allergy 2005, 86, 151–183. [Google Scholar] [PubMed]
- Bonneville, M.; O’Brien, R.L.; Born, W.K. Gammadelta T cell effector functions: A blend of innate programming and acquired plasticity. Nat. Rev. Immunol. 2010, 10, 467–478. [Google Scholar] [CrossRef]
- Chitadze, G.; Oberg, H.-H.; Wesch, D.; Kabelitz, D. The Ambiguous Role of γδ T Lymphocytes in Antitumor Immunity. Trends Immunol. 2017, 38, 668–678. [Google Scholar] [CrossRef]
- Wu, Y.; Kyle-Cezar, F.; Woolf, R.T.; Naceur-Lombardelli, C.; Owen, J.; Biswas, D.; Lorenc, A.; Vantourout, P.; Gazinska, P.; Grigoriadis, A.; 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. [Google Scholar] [CrossRef]
- Devaud, C.; Rousseau, B.; Netzer, S.; Pitard, V.; Paroissin, C.; Khairallah, C.; Costet, P.; Moreau, J.-F.; Couillaud, F.; Dechanet-Merville, J.; et al. Anti-metastatic potential of human Vδ1(+) γδ T cells in an orthotopic mouse xenograft model of colon carcinoma. Cancer Immunol. Immunother. 2013, 62, 1199–1210. [Google Scholar] [CrossRef]
- Kakimi, K.; Matsushita, H.; Murakawa, T.; Nakajima, J. γδ T cell therapy for the treatment of non-small cell lung cancer. Transl. Lung Cancer Res. 2014, 3, 23–33. [Google Scholar]
- Correia, D.V.; Lopes, A.; Silva-Santos, B. Tumor cell recognition by γδ T lymphocytes: T-cell receptor vs. NK-cell receptors. Oncoimmunology 2013, 2, e22892. [Google Scholar] [CrossRef] [Green Version]
- Lafont, V.; Sanchez, F.; Laprevotte, E.; Michaud, H.-A.; Gros, L.; Eliaou, J.-F.; Bonnefoy, N. Plasticity of γδ T Cells: Impact on the Anti-Tumor Response. Front. Immunol. 2014, 5, 622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silva-Santos, B.; Mensurado, S.; Coffelt, S.B. gammadelta T cells: Pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 2019, 19, 392–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sebestyen, Z.; Prinz, I.; Déchanet-Merville, J.; Silva-Santos, B.; Kuball, J. Translating gammadelta (γδ) T cells and their receptors into cancer cell therapies. Nat. Rev. Drug Discov. 2020, 19, 169–184. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.W.; Chung, Y.S.; Kim, T.J. Heterogeneity of Human γδ T Cells and Their Role in Cancer Immunity. Immune Netw. 2020, 20, e5. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, L.C.; Shaw, E.M.; Stankovic, S.; Snell, G.I.; Brooks, A.G.; Westall, G.P. The complex existence of γδ T cells following transplantation: The good, the bad and the simply confusing. Clin. Transl. Immunol. 2019, 8, e1078. [Google Scholar] [CrossRef]
- Fichtner, A.S.; Ravens, S.; Prinz, I. Human γδ TCR Repertoires in Health and Disease. Cells 2020, 9, 800. [Google Scholar] [CrossRef] [Green Version]
- Pang, D.J.; Neves, J.F.; Sumaria, N.; Pennington, D.J. Understanding the complexity of γδ T-cell subsets in mouse and human. Immunology 2012, 136, 283–290. [Google Scholar] [CrossRef]
- Mikulak, J.; Oriolo, F.; Bruni, E.; Roberto, A.; Colombo, F.S.; Villa, A.; Bosticardo, M.; Bortolomai, I.; Lo Presti, E.; Meraviglia, S.; et al. NKp46-expressing human gut-resident intraepithelial Vδ1 T cell subpopulation exhibits high antitumor activity against colorectal cancer. Jci Insight 2019, 4. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Niu, C.; Cui, J. Gamma-delta (gammadelta) T cells: Friend or foe in cancer development? J. Transl. Med. 2018, 16, 3. [Google Scholar] [CrossRef] [Green Version]
- Sant, S.; Jenkins, M.R.; Dash, P.; Watson, K.A.; Wang, Z.; Pizzolla, A.; Koutsakos, M.; Nguyen, T.H.; Lappas, M.; Crowe, J.; et al. Human γδ T-cell receptor repertoire is shaped by influenza viruses, age and tissue compartmentalisation. Clin. Transl. Immunol. 2019, 8, e1079. [Google Scholar] [CrossRef] [Green Version]
- Siegers, G.M.; Dhamko, H.; Wang, X.-H.; Mathieson, A.M.; Kosaka, Y.; Felizardo, T.C.; Medin, J.A.; Tohda, S.; Schueler, J.; Fisch, P.; et al. Human Vδ1 γδ T cells expanded from peripheral blood exhibit specific cytotoxicity against B-cell chronic lymphocytic leukemia-derived cells. Cytotherapy 2011, 13, 753–764. [Google Scholar] [CrossRef] [PubMed]
- Knight, A.; Mackinnon, S.; Lowdell, M.W. Human Vdelta1 gamma-delta T cells exert potent specific cytotoxicity against primary multiple myeloma cells. Cytotherapy 2012, 14, 1110–1118. [Google Scholar] [CrossRef] [PubMed]
- Alnaggar, M.; Xu, Y.; Li, J.; He, J.; Chen, J.; Li, M.; Wu, Q.; Lin, L.; Liang, Y.; Wang, X.; et al. Allogenic Vγ9Vδ2 T cell as new potential immunotherapy drug for solid tumor: A case study for cholangiocarcinoma. J. Immunother. Cancer 2019, 7, 36. [Google Scholar] [CrossRef] [PubMed]
- Joalland, N.; Chauvin, C.; Oliver, L.; Vallette, F.M.; Pecqueur, C.; Jarry, U.; Scotet, E. IL-21 Increases the Reactivity of Allogeneic Human Vγ9Vδ2 T Cells Against Primary Glioblastoma Tumors. J. Immunother. 2018, 41, 224–231. [Google Scholar] [CrossRef]
- Meraviglia, S.; Eberl, M.; Vermijlen, D.; Todaro, M.; Buccheri, S.; Cicero, G.; La Mendola, C.; Guggino, G.; D’Asaro, M.; Orlando, V.; et al. In vivo manipulation of Vgamma9Vdelta2 T cells with zoledronate and low-dose interleukin-2 for immunotherapy of advanced breast cancer patients. Clin. Exp. Immunol. 2010, 161, 290–297. [Google Scholar]
- Di Carlo, E.; Bocca, P.; Emionite, L.; Cilli, M.; Cipollone, G.; Morandi, F.; Raffaghello, L.; Pistoia, V.; Prigione, I. Mechanisms of the antitumor activity of human Vγ9Vδ2 T cells in combination with zoledronic acid in a preclinical model of neuroblastoma. Mol. Ther. 2013, 21, 1034–1043. [Google Scholar] [CrossRef] [Green Version]
- Davey, M.S.; Willcox, C.R.; Hunter, S.; Kasatskaya, S.A.; Remmerswaal, E.B.M.; Salim, M.; Mohammed, F.; Bemelman, F.J.; Chudakov, D.M.; Oo, Y.H.; et al. The human Vδ2 T-cell compartment comprises distinct innate-like Vγ9 and adaptive Vγ9 subsets. Nat. Commun. 2018, 9, 1760. [Google Scholar] [CrossRef] [PubMed]
- Mangan, B.A.; Dunne, M.R.; O’Reilly, V.P.; Dunne, P.J.; Exley, M.A.; O’Shea, D.; Scotet, E.; Hogan, A.E.; Doherty, D.G. Cutting edge: CD1d restriction and Th1/Th2/Th17 cytokine secretion by human Vδ3 T cells. J. Immunother. 2013, 191, 30–34. [Google Scholar] [CrossRef]
- Willcox, C.R.; Pitard, V.; Netzer, S.; Couzi, L.; Salim, M.; Silberzahn, T.; Moreau, J.-F.; Hayday, A.C.; Willcox, B.E.; Déchanet-Merville, J. Cytomegalovirus and tumor stress surveillance by binding of a human γδ T cell antigen receptor to endothelial protein C receptor. Nat. Immunol. 2012, 13, 872–879. [Google Scholar] [CrossRef]
- Qin, G.; Liu, Y.; Zheng, J.; Xiang, Z.; Ng, I.H.Y.; Malik Peiris, J.S.; Lau, Y.-L.; Tu, W. Phenotypic and functional characterization of human γδ T-cell subsets in response to influenza A viruses. J. Infect. Dis. 2012, 205, 1646–1653. [Google Scholar] [CrossRef] [Green Version]
- Chen, H.; He, W. Human regulatory γδT cells and their functional plasticity in the tumor microenvironment. Cell. Mol. Immunol. 2018, 15, 411–413. [Google Scholar] [CrossRef]
- Wesch, D.; Glatzel, A.; Kabelitz, D. Differentiation of resting human peripheral blood gamma delta T cells toward Th1- or Th2-phenotype. Cell. Immunol. 2001, 212, 110–117. [Google Scholar] [CrossRef]
- McGeachy, M.J.; Cua, D.J.; Gaffen, S.L. The IL-17 Family of Cytokines in Health and Disease. Immunity 2019, 50, 892–906. [Google Scholar] [CrossRef]
- Caccamo, N.; La Mendola, C.; Orlando, V.; Meraviglia, S.; Todaro, M.; Stassi, G.; Sireci, G.; Fournié, J.J.; Dieli, F. Differentiation, phenotype, and function of interleukin-17-producing human Vγ9Vδ2 T cells. Blood 2011, 118, 129–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ness-Schwickerath, K.J.; Jin, C.; Morita, C.T. Cytokine requirements for the differentiation and expansion of IL-17A- and IL-22-producing human Vgamma2Vdelta2 T cells. J. Immunother. 2010, 184, 7268–7280. [Google Scholar]
- Mou, W.; Han, W.; Ma, X.; Wang, X.; Qin, H.; Zhao, W.; Ren, X.; Chen, X.; Yang, W.; Cheng, H.; et al. γδTFH cells promote B cell maturation and antibody production in neuroblastoma. BMC Immunol. 2017, 18, 36. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ye, J.; Ma, C.; Hsueh, E.C.; Eickhoff, C.S.; Zhang, Y.; Varvares, M.A.; Hoft, D.F.; Peng, G. Tumor-derived γδ regulatory T cells suppress innate and adaptive immunity through the induction of immunosenescence. J. Immunother. 2013, 190, 2403–2414. [Google Scholar] [CrossRef] [PubMed]
- Peng, G.; Wang, H.Y.; Peng, W.; Kiniwa, Y.; Seo, K.H.; Wang, R.-F. Tumor-infiltrating gammadelta T cells suppress T and dendritic cell function via mechanisms controlled by a unique toll-like receptor signaling pathway. Immunity 2007, 27, 334–348. [Google Scholar] [CrossRef] [Green Version]
- Teirlinck, A.C.; McCall, M.B.B.; Roestenberg, M.; Scholzen, A.; Woestenenk, R.; de Mast, Q.; van der Ven, A.J.A.M.; Hermsen, C.C.; Luty, A.J.F.; Sauerwein, R.W. Longevity and composition of cellular immune responses following experimental Plasmodium falciparum malaria infection in humans. PLoS Pathog. 2011, 7, e1002389. [Google Scholar] [CrossRef]
- Hoft, D.F.; Brown, R.M.; Roodman, S.T. Bacille Calmette-Guerin vaccination enhances human gamma delta T cell responsiveness to mycobacteria suggestive of a memory-like phenotype. J. Immunol. 1998, 161, 1045–1054. [Google Scholar]
- Eberl, M.; Engel, R.; Beck, E.; Jomaa, H. Differentiation of human gamma-delta T cells towards distinct memory phenotypes. Cell. Immunol. 2002, 218, 1–6. [Google Scholar] [CrossRef]
- Silva-Santos, B.; Serre, K.; Norell, H. γδ T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef] [PubMed]
- Mullen, P.J.; Yu, R.; Longo, J.; Archer, M.C.; Penn, L.Z. The interplay between cell signalling and the mevalonate pathway in cancer. Nat. Rev. Cancer 2016, 16, 718–731. [Google Scholar] [CrossRef] [PubMed]
- Scotet, E.; Martinez, L.O.; Grant, E.; Barbaras, R.; Jenö, P.; Guiraud, M.; Monsarrat, B.; Saulquin, X.; Maillet, S.; Estève, J.-P.; et al. Tumor recognition following Vgamma9Vdelta2 T cell receptor interactions with a surface F1-ATPase-related structure and apolipoprotein A-I. Immunity 2005, 22, 71–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vavassori, S.; Kumar, A.; Wan, G.S.; Ramanjaneyulu, G.S.; Cavallari, M.; El Daker, S.; Beddoe, T.; Theodossis, A.; Williams, N.K.; Gostick, E.; et al. Butyrophilin 3A1 binds phosphorylated antigens and stimulates human γδ T cells. Nat. Immunol. 2013, 14, 908–916. [Google Scholar] [CrossRef]
- Kabelitz, D. CD277 takes the lead in human γδ T-cell activation. Blood 2012, 120, 2159–2161. [Google Scholar] [CrossRef] [Green Version]
- Sandstrom, A.; Peigné, C.-M.; Léger, A.; Crooks, J.E.; Konczak, F.; Gesnel, M.-C.; Breathnach, R.; Bonneville, M.; Scotet, E.; Adams, E.J. The intracellular B30.2 domain of butyrophilin 3A1 binds phosphoantigens to mediate activation of human Vγ9Vδ2 T cells. Immunity 2014, 40, 490–500. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, K.; Li, J.; Puthenveetil, R.; Lin, X.; Poe, M.M.; Hsiao, C.-H.C.; Vinogradova, O.; Wiemer, A.J. The butyrophilin 3A1 intracellular domain undergoes a conformational change involving the juxtamembrane region. FASEB J. 2017, 31, 4697–4706. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Li, L.; Yuan, L.; Zhou, X.; Duan, J.; Xiao, H.; Cai, N.; Han, S.; Ma, X.; Liu, W.; et al. A Structural Change in Butyrophilin upon Phosphoantigen Binding Underlies Phosphoantigen-Mediated Vγ9Vδ2 T Cell Activation. Immunity 2019, 50. [Google Scholar] [CrossRef] [Green Version]
- Peigné, C.-M.; Léger, A.; Gesnel, M.-C.; Konczak, F.; Olive, D.; Bonneville, M.; Breathnach, R.; Scotet, E. The Juxtamembrane Domain of Butyrophilin BTN3A1 Controls Phosphoantigen-Mediated Activation of Human Vγ9Vδ2 T Cells. J. Immunother. 2017, 198, 4228–4234. [Google Scholar] [CrossRef] [Green Version]
- Gu, S.; Sachleben, J.R.; Boughter, C.T.; Nawrocka, W.I.; Borowska, M.T.; Tarrasch, J.T.; Skiniotis, G.; Roux, B.; Adams, E.J. Phosphoantigen-induced conformational change of butyrophilin 3A1 (BTN3A1) and its implication on Vγ9Vδ2 T cell activation. Proc. Natl. Acad. Sci. USA 2017, 114, E7311–E7320. [Google Scholar] [CrossRef] [Green Version]
- Karunakaran, M.M.; Willcox, C.R.; Salim, M.; Paletta, D.; Fichtner, A.S.; Noll, A.; Starick, L.; Nöhren, A.; Begley, C.R.; Berwick, K.A.; et al. Butyrophilin-2A1 Directly Binds Germline-Encoded Regions of the Vγ9Vδ2 TCR and Is Essential for Phosphoantigen Sensing. Immunity 2020, 52. [Google Scholar] [CrossRef] [PubMed]
- Rigau, M.; Ostrouska, S.; Fulford, T.S.; Johnson, D.N.; Woods, K.; Ruan, Z.; McWilliam, H.E.G.; Hudson, C.; Tutuka, C.; Wheatley, A.K.; et al. Butyrophilin 2A1 is essential for phosphoantigen reactivity by γδ T cells. Science 2020, 367. [Google Scholar] [CrossRef] [PubMed]
- Dhar, P.; Wu, J.D. NKG2D and its ligands in cancer. Curr. Opin. Immunol. 2018, 51, 55–61. [Google Scholar] [CrossRef] [PubMed]
- Toutirais, O.; Cabillic, F.; Le Friec, G.; Salot, S.; Loyer, P.; Le Gallo, M.; Desille, M.; de La Pintière, C.T.; Daniel, P.; Bouet, F.; et al. DNAX accessory molecule-1 (CD226) promotes human hepatocellular carcinoma cell lysis by Vgamma9Vdelta2 T cells. Eur. J. Immunol. 2009, 39, 1361–1368. [Google Scholar] [CrossRef]
- Gertner-Dardenne, J.; Castellano, R.; Mamessier, E.; Garbit, S.; Kochbati, E.; Etienne, A.; Charbonnier, A.; Collette, Y.; Vey, N.; Olive, D. Human Vγ9Vδ2 T cells specifically recognize and kill acute myeloid leukemic blasts. J. Immunother. 2012, 188, 4701–4708. [Google Scholar] [CrossRef]
- Luoma, A.M.; Castro, C.D.; Adams, E.J. γδ T cell surveillance via CD1 molecules. Trends Immunol. 2014, 35, 613–621. [Google Scholar] [CrossRef]
- Bai, L.; Picard, D.; Anderson, B.; Chaudhary, V.; Luoma, A.; Jabri, B.; Adams, E.J.; Savage, P.B.; Bendelac, A. The majority of CD1d-sulfatide-specific T cells in human blood use a semiinvariant Vδ1 TCR. Eur. J. Immunol. 2012, 42, 2505–2510. [Google Scholar] [CrossRef]
- Luoma, A.M.; Castro, C.D.; Mayassi, T.; Bembinster, L.A.; Bai, L.; Picard, D.; Anderson, B.; Scharf, L.; Kung, J.E.; Sibener, L.V.; et al. Crystal structure of Vδ1 T cell receptor in complex with CD1d-sulfatide shows MHC-like recognition of a self-lipid by human γδ T cells. Immunity 2013, 39, 1032–1042. [Google Scholar] [CrossRef] [Green Version]
- Dhodapkar, M.V.; Geller, M.D.; Chang, D.H.; Shimizu, K.; Fujii, S.-I.; Dhodapkar, K.M.; Krasovsky, J. A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma. J. Exp. Med. 2003, 197, 1667–1676. [Google Scholar] [CrossRef]
- Dhodapkar, M.V.; Richter, J. Harnessing natural killer T (NKT) cells in human myeloma: Progress and challenges. Clin. Immunol. 2011, 140, 160–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, D.; Song, L.; Brawley, V.S.; Robison, N.; Wei, J.; Gao, X.; Tian, G.; Margol, A.; Ahmed, N.; Asgharzadeh, S.; et al. Medulloblastoma expresses CD1d and can be targeted for immunotherapy with NKT cells. Clin. Immunol. 2013, 149, 55–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nowak, M.; Arredouani, M.S.; Tun-Kyi, A.; Schmidt-Wolf, I.; Sanda, M.G.; Balk, S.P.; Exley, M.A. Defective NKT cell activation by CD1d+ TRAMP prostate tumor cells is corrected by interleukin-12 with α-galactosylceramide. PLoS ONE 2010, 5, e11311. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Chen, Y.; Momin, A.; Shaner, R.; Wang, E.; Bowen, N.J.; Matyunina, L.V.; Walker, L.D.; McDonald, J.F.; Sullards, M.C.; et al. Elevation of sulfatides in ovarian cancer: An integrated transcriptomic and lipidomic analysis including tissue-imaging mass spectrometry. Mol. Cancer 2010, 9, 186. [Google Scholar] [CrossRef] [Green Version]
- Xu, B.; Pizarro, J.C.; Holmes, M.A.; McBeth, C.; Groh, V.; Spies, T.; Strong, R.K. Crystal structure of a gammadelta T-cell receptor specific for the human MHC class I homolog MICA. Proc. Natl. Acad. Sci. USA 2011, 108, 2414–2419. [Google Scholar] [CrossRef] [Green Version]
- Poggi, A.; Venturino, C.; Catellani, S.; Clavio, M.; Miglino, M.; Gobbi, M.; Steinle, A.; Ghia, P.; Stella, S.; Caligaris-Cappio, F.; et al. Vdelta1 T lymphocytes from B-CLL patients recognize ULBP3 expressed on leukemic B cells and up-regulated by trans-retinoic acid. Cancer Res. 2004, 64, 9172–9179. [Google Scholar] [CrossRef] [Green Version]
- Groh, V.; Steinle, A.; Bauer, S.; Spies, T. Recognition of stress-induced MHC molecules by intestinal epithelial gammadelta T cells. Science 1998, 279, 1737–1740. [Google Scholar] [CrossRef]
- Satoh-Takayama, N.; Vosshenrich, C.A.J.; Lesjean-Pottier, S.; Sawa, S.; Lochner, M.; Rattis, F.; Mention, J.-J.; Thiam, K.; Cerf-Bensussan, N.; Mandelboim, O.; et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 2008, 29, 958–970. [Google Scholar] [CrossRef]
- Almeida, A.R.; Correia, D.V.; Fernandes-Platzgummer, A.; da Silva, C.L.; da Silva, M.G.; Anjos, D.R.; Silva-Santos, B. 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. [Google Scholar] [CrossRef] [Green Version]
- Correia, D.V.; Fogli, M.; Hudspeth, K.; da Silva, M.G.; Mavilio, D.; Silva-Santos, B. Differentiation of human peripheral blood Vδ1+ T cells expressing the natural cytotoxicity receptor NKp30 for recognition of lymphoid leukemia cells. Blood 2011, 118. [Google Scholar] [CrossRef] [Green Version]
- Silva-Santos, B.; Strid, J. Working in “NK Mode”: Natural Killer Group 2 Member D and Natural Cytotoxicity Receptors in Stress-Surveillance by γδ T Cells. Front. Immunol. 2018, 9, 851. [Google Scholar] [CrossRef] [Green Version]
- Hudspeth, K.; Silva-Santos, B.; Mavilio, D. Natural cytotoxicity receptors: Broader expression patterns and functions in innate and adaptive immune cells. Front. Immunol. 2013, 4, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Lorenzo, B.; Simões, A.E.; Caiado, F.; Tieppo, P.; Correia, D.V.; Carvalho, T.; da Silva, M.G.; Déchanet-Merville, J.; Schumacher, T.N.; Prinz, I.; et al. Broad Cytotoxic Targeting of Acute Myeloid Leukemia by Polyclonal Delta One T Cells. Cancer Immunol. Res. 2019, 7, 552–558. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Witherden, D.A.; Havran, W.L. EPCR: A stress trigger for γδ T cells. Nat. Immunol. 2012, 13, 812–814. [Google Scholar] [CrossRef] [PubMed]
- Stam, A.; de Bruin, R.; Roovers, R.; van Bergen en Henegouwen, P.; Verheul, H.; de Gruijl, T.D.; van der Vliet, H. Specific tumor targeting and activation of Vγ9Vδ2 T cells by bi-specific nanobodies. J. Immunother. Cancer 2014, 2, 127. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Peng, H.; Xu, Q.; Ye, Z. Sensitization of human osteosarcoma cells to Vγ9Vδ2 T-cell-mediated cytotoxicity by zoledronate. J. Orthop. Res. 2012, 30, 824–830. [Google Scholar] [CrossRef]
- D’Asaro, M.; La Mendola, C.; Di Liberto, D.; Orlando, V.; Todaro, M.; Spina, M.; Guggino, G.; Meraviglia, S.; Caccamo, N.; Messina, A.; et al. V gamma 9V delta 2 T lymphocytes efficiently recognize and kill zoledronate-sensitized, imatinib-sensitive, and imatinib-resistant chronic myelogenous leukemia cells. J. Immunother. 2010, 184, 3260–3268. [Google Scholar]
- Wu, D.; Wu, P.; Wu, X.; Ye, J.; Wang, Z.; Zhao, S.; Ni, C.; Hu, G.; Xu, J.; Han, Y.; et al. expanded human circulating Vδ1 γδT cells exhibit favorable therapeutic potential for colon cancer. Oncoimmunology 2015, 4, e992749. [Google Scholar] [CrossRef] [Green Version]
- Halary, F.; Pitard, V.; Dlubek, D.; Krzysiek, R.; de la Salle, H.; Merville, P.; Dromer, C.; Emilie, D.; Moreau, J.-F.; Déchanet-Merville, J. Shared reactivity of Vδ2neg γδ T cells against cytomegalovirus-infected cells and tumor intestinal epithelial cells. J. Exp. Med. 2005, 201, 1567–1578. [Google Scholar] [CrossRef] [Green Version]
- Tokuyama, H.; Hagi, T.; Mattarollo, S.R.; Morley, J.; Wang, Q.; So, H.-F.; Fai-So, H.; Moriyasu, F.; Nieda, M.; Nicol, A.J. V gamma 9 V delta 2 T cell cytotoxicity against tumor cells is enhanced by monoclonal antibody drugs--rituximab and trastuzumab. Int. J. Cancer 2008, 122, 2526–2534. [Google Scholar] [CrossRef]
- Gertner-Dardenne, J.; Bonnafous, C.; Bezombes, C.; Capietto, A.-H.; Scaglione, V.; Ingoure, S.; Cendron, D.; Gross, E.; Lepage, J.-F.; Quillet-Mary, A.; et al. Bromohydrin pyrophosphate enhances antibody-dependent cell-mediated cytotoxicity induced by therapeutic antibodies. Blood 2009, 113, 4875–4884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Capietto, A.-H.; Martinet, L.; Fournié, J.-J. Stimulated γδ T cells increase the in vivo efficacy of trastuzumab in HER-2+ breast cancer. J. Immunother. 2011, 187, 1031–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caccamo, N.; Battistini, L.; Bonneville, M.; Poccia, F.; Fournié, J.J.; Meraviglia, S.; Borsellino, G.; Kroczek, R.A.; La Mendola, C.; Scotet, E.; et al. CXCR5 identifies a subset of Vgamma9Vdelta2 T cells which secrete IL-4 and IL-10 and help B cells for antibody production. J. Immunother. 2006, 177, 5290–5295. [Google Scholar]
- Bansal, R.R.; Mackay, C.R.; Moser, B.; Eberl, M. IL-21 enhances the potential of human γδ T cells to provide B-cell help. Eur. J. Immunol. 2012, 42, 110–119. [Google Scholar] [CrossRef]
- Crawford, G.; Hayes, M.D.; Seoane, R.C.; Ward, S.; Dalessandri, T.; Lai, C.; Healy, E.; Kipling, D.; Proby, C.; Moyes, C.; et al. Epithelial damage and tissue γδ T cells promote a unique tumor-protective IgE response. Nat. Immunol. 2018, 19, 859–870. [Google Scholar] [CrossRef]
- Rezende, R.M.; Lanser, A.J.; Rubino, S.; Kuhn, C.; Skillin, N.; Moreira, T.G.; Liu, S.; Gabriely, G.; David, B.A.; Menezes, G.B.; et al. γδ T cells control humoral immune response by inducing T follicular helper cell differentiation. Nat. Commun. 2018, 9, 3151. [Google Scholar] [CrossRef] [Green Version]
- Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef]
- Mao, C.; Mou, X.; Zhou, Y.; Yuan, G.; Xu, C.; Liu, H.; Zheng, T.; Tong, J.; Wang, S.; Chen, D. Tumor-activated TCRγδ+ T cells from gastric cancer patients induce the antitumor immune response of TCRαβ+ T cells via their antigen-presenting cell-like effects. J. Immunol. Res. 2014, 2014, 593562. [Google Scholar] [CrossRef] [Green Version]
- Muto, M.; Baghdadi, M.; Maekawa, R.; Wada, H.; Seino, K.-I. Myeloid molecular characteristics of human γδ T cells support their acquisition of tumor antigen-presenting capacity. Cancer Immunol. Immunother. 2015, 64, 941–949. [Google Scholar] [CrossRef] [Green Version]
- Maniar, A.; Zhang, X.; Lin, W.; Gastman, B.R.; Pauza, C.D.; Strome, S.E.; Chapoval, A.I. Human gammadelta T lymphocytes induce robust NK cell-mediated antitumor cytotoxicity through CD137 engagement. Blood 2010, 116, 1726–1733. [Google Scholar] [CrossRef]
- Van Acker, H.H.; Anguille, S.; Van Tendeloo, V.F.; Lion, E. Empowering gamma delta T cells with antitumor immunity by dendritic cell-based immunotherapy. Oncoimmunology 2015, 4, e1021538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conti, L.; Casetti, R.; Cardone, M.; Varano, B.; Martino, A.; Belardelli, F.; Poccia, F.; Gessani, S. Reciprocal activating interaction between dendritic cells and pamidronate-stimulated gammadelta T cells: Role of CD86 and inflammatory cytokines. J. Immunother. 2005, 174, 252–260. [Google Scholar]
- Zou, W. Mechanistic insights into cancer immunity and immunotherapy. Cell. Mol. Immunol. 2018, 15, 419–420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, L.; Xu, H.; Peng, G. TLR-mediated metabolic reprogramming in the tumor microenvironment: Potential novel strategies for cancer immunotherapy. Cell. Mol. Immunol. 2018, 15, 428–437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, Y.-L.; Ding, Y.-P.; Tanaka, Y.; Shen, L.-W.; Wei, C.-H.; Minato, N.; Zhang, W. γδ T cells and their potential for immunotherapy. Int. J. Biol. Sci. 2014, 10, 119–135. [Google Scholar] [CrossRef] [Green Version]
- Lo Presti, E.; Pizzolato, G.; Gulotta, E.; Cocorullo, G.; Gulotta, G.; Dieli, F.; Meraviglia, S. Current Advances in gammadelta T Cell-Based Tumor Immunotherapy. Front. Immunol. 2017, 8, 1401. [Google Scholar] [CrossRef] [Green Version]
- Zysk, A.; DeNichilo, M.O.; Panagopoulos, V.; Zinonos, I.; Liapis, V.; Hay, S.; Ingman, W.; Ponomarev, V.; Atkins, G.; Findlay, D.; et al. Adoptive transfer of ex vivo expanded Vγ9Vδ2 T cells in combination with zoledronic acid inhibits cancer growth and limits osteolysis in a murine model of osteolytic breast cancer. Cancer Lett. 2017, 386, 141–150. [Google Scholar] [CrossRef] [Green Version]
- Wada, I.; Matsushita, H.; Noji, S.; Mori, K.; Yamashita, H.; Nomura, S.; Shimizu, N.; Seto, Y.; Kakimi, K. Intraperitoneal injection of in vitro expanded Vγ9Vδ2 T cells together with zoledronate for the treatment of malignant ascites due to gastric cancer. Cancer Med. 2014, 3, 362–375. [Google Scholar] [CrossRef]
- Bennouna, J.; Levy, V.; Sicard, H.; Senellart, H.; Audrain, M.; Hiret, S.; Rolland, F.; Bruzzoni-Giovanelli, H.; Rimbert, M.; Galéa, C.; et al. Phase I study of bromohydrin pyrophosphate (BrHPP, IPH 1101), a Vgamma9Vdelta2 T lymphocyte agonist in patients with solid tumors. Cancer Immunol. Immunother. 2010, 59, 1521–1530. [Google Scholar] [CrossRef]
- Bennouna, J.; Bompas, E.; Neidhardt, E.M.; Rolland, F.; Philip, I.; Galéa, C.; Salot, S.; Saiagh, S.; Audrain, M.; Rimbert, M.; et al. Phase-I study of Innacell gammadelta, an autologous cell-therapy product highly enriched in gamma9delta2 T lymphocytes, in combination with IL-2, in patients with metastatic renal cell carcinoma. Cancer Immunol. Immunother. 2008, 57, 1599–1609. [Google Scholar] [CrossRef]
- Nakajima, J.; Murakawa, T.; Fukami, T.; Goto, S.; Kaneko, T.; Yoshida, Y.; Takamoto, S.; Kakimi, K. A phase I study of adoptive immunotherapy for recurrent non-small-cell lung cancer patients with autologous gammadelta T cells. Eur. J. Cardio-Thorac. Surg. 2010, 37, 1191–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sakamoto, M.; Nakajima, J.; Murakawa, T.; Fukami, T.; Yoshida, Y.; Murayama, T.; Takamoto, S.; Matsushita, H.; Kakimi, K. Adoptive immunotherapy for advanced non-small cell lung cancer using zoledronate-expanded γδTcells: A phase I clinical study. J. Immunother. 2011, 34, 202–211. [Google Scholar] [CrossRef] [PubMed]
- Nicol, A.J.; Tokuyama, H.; Mattarollo, S.R.; Hagi, T.; Suzuki, K.; Yokokawa, K.; Nieda, M. Clinical evaluation of autologous gamma delta T cell-based immunotherapy for metastatic solid tumours. Br. J. Cancer 2011, 105, 778–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kobayashi, H.; Tanaka, Y.; Yagi, J.; Minato, N.; Tanabe, K. Phase I/II study of adoptive transfer of γδ T cells in combination with zoledronic acid and IL-2 to patients with advanced renal cell carcinoma. Cancer Immunol. Immunother. 2011, 60, 1075–1084. [Google Scholar] [CrossRef]
- Abe, Y.; Muto, M.; Nieda, M.; Nakagawa, Y.; Nicol, A.; Kaneko, T.; Goto, S.; Yokokawa, K.; Suzuki, K. Clinical and immunological evaluation of zoledronate-activated Vgamma9gammadelta T-cell-based immunotherapy for patients with multiple myeloma. Exp. Hematol. 2009, 37, 956–968. [Google Scholar] [CrossRef]
- Kobayashi, H.; Tanaka, Y.; Yagi, J.; Osaka, Y.; Nakazawa, H.; Uchiyama, T.; Minato, N.; Toma, H. Safety profile and anti-tumor effects of adoptive immunotherapy using gamma-delta T cells against advanced renal cell carcinoma: A pilot study. Cancer Immunol. Immunother. 2007, 56, 469–476. [Google Scholar] [CrossRef]
- Wilhelm, M.; Kunzmann, V.; Eckstein, S.; Reimer, P.; Weissinger, F.; Ruediger, T.; Tony, H.-P. Gammadelta T cells for immune therapy of patients with lymphoid malignancies. Blood 2003, 102, 200–206. [Google Scholar] [CrossRef] [Green Version]
- Kunzmann, V.; Smetak, M.; Kimmel, B.; Weigang-Koehler, K.; Goebeler, M.; Birkmann, J.; Becker, J.; Schmidt-Wolf, I.G.H.; Einsele, H.; Wilhelm, M. Tumor-promoting versus tumor-antagonizing roles of γδ T cells in cancer immunotherapy: Results from a prospective phase I/II trial. J. Immunother. 2012, 35, 205–213. [Google Scholar] [CrossRef]
- Lang, J.M.; Kaikobad, M.R.; Wallace, M.; Staab, M.J.; Horvath, D.L.; Wilding, G.; Liu, G.; Eickhoff, J.C.; McNeel, D.G.; Malkovsky, M. Pilot trial of interleukin-2 and zoledronic acid to augment γδ T cells as treatment for patients with refractory renal cell carcinoma. Cancer Immunol. Immunother. 2011, 60, 1447–1460. [Google Scholar] [CrossRef] [Green Version]
- Pressey, J.G.; Adams, J.; Harkins, L.; Kelly, D.; You, Z.; Lamb, L.S. In vivo expansion and activation of γδ T cells as immunotherapy for refractory neuroblastoma: A phase 1 study. Medicine 2016, 95, e4909. [Google Scholar] [CrossRef]
- Kouakanou, L.; Xu, Y.; Peters, C.; He, J.; Wu, Y.; Yin, Z.; Kabelitz, D. Vitamin C promotes the proliferation and effector functions of human γδ T cells. Cell. Mol. Immunol. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Siegers, G.M.; Lamb, L.S. Cytotoxic and regulatory properties of circulating Vδ1+ γδ T cells: A new player on the cell therapy field? Mol. Ther. 2014, 22, 1416–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meraviglia, S.; Dieli, F. Clonal expansion shapes the human Vδ1T cell receptor repertoire. Cell. Mol. Immunol. 2018, 15, 96–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oberg, H.-H.; Peipp, M.; Kellner, C.; Sebens, S.; Krause, S.; Petrick, D.; Adam-Klages, S.; Röcken, C.; Becker, T.; Vogel, I.; et al. Novel bispecific antibodies increase γδ T-cell cytotoxicity against pancreatic cancer cells. Cancer Res. 2014, 74, 1349–1360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oberg, H.-H.; Kellner, C.; Gonnermann, D.; Peipp, M.; Peters, C.; Sebens, S.; Kabelitz, D.; Wesch, D. γδ T cell activation by bispecific antibodies. Cell. Immunol. 2015, 296, 41–49. [Google Scholar] [CrossRef]
- de Bruin, R.C.G.; Veluchamy, J.P.; Lougheed, S.M.; Schneiders, F.L.; Lopez-Lastra, S.; Lameris, R.; Stam, A.G.; Sebestyen, Z.; Kuball, J.; Molthoff, C.F.M.; et al. A bispecific nanobody approach to leverage the potent and widely applicable tumor cytolytic capacity of Vγ9Vδ2-T cells. Oncoimmunology 2017, 7, e1375641. [Google Scholar] [CrossRef]
- Oberg, H.H.; Kellner, C.; Gonnermann, D.; Sebens, S.; Bauerschlag, D.; Gramatzki, M.; Kabelitz, D.; Peipp, M.; Wesch, D. Tribody [(HER2)xCD16] Is More Effective Than Trastuzumab in Enhancing γδ T Cell and Natural Killer Cell Cytotoxicity Against HER2-Expressing Cancer Cells. Front. Immunol. 2018, 9, 814. [Google Scholar] [CrossRef]
- Johnson, L.A.; June, C.H. Driving gene-engineered T cell immunotherapy of cancer. Cell Res. 2017, 27, 38–58. [Google Scholar] [CrossRef] [Green Version]
- Shah, N.N.; Fry, T.J. Mechanisms of resistance to CAR T cell therapy. Nat. Rev. Clin. Oncol. 2019, 16, 372–385. [Google Scholar] [CrossRef]
- Deniger, D.C.; Moyes, J.S.; Cooper, L.J.N. Clinical applications of gamma delta T cells with multivalent immunity. Front. Immunol. 2014, 5, 636. [Google Scholar] [CrossRef] [Green Version]
- Deniger, D.C.; Switzer, K.; Mi, T.; Maiti, S.; Hurton, L.; Singh, H.; Huls, H.; Olivares, S.; Lee, D.A.; Champlin, R.E.; et al. Bispecific T-cells expressing polyclonal repertoire of endogenous γδ T-cell receptors and introduced CD19-specific chimeric antigen receptor. Mol. Ther. 2013, 21, 638–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fisher, J.; Abramowski, P.; Wisidagamage Don, N.D.; Flutter, B.; Capsomidis, A.; Cheung, G.W.-K.; Gustafsson, K.; Anderson, J. Avoidance of On-Target Off-Tumor Activation Using a Co-stimulation-Only Chimeric Antigen Receptor. Mol. Ther. 2017, 25, 1234–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Capsomidis, A.; Benthall, G.; Van Acker, H.H.; Fisher, J.; Kramer, A.M.; Abeln, Z.; Majani, Y.; Gileadi, T.; Wallace, R.; Gustafsson, K.; et al. Chimeric Antigen Receptor-Engineered Human Gamma Delta T Cells: Enhanced Cytotoxicity with Retention of Cross Presentation. Mol. Ther. 2018, 26, 354–365. [Google Scholar] [CrossRef] [Green Version]
- Gründer, C.; van Dorp, S.; Hol, S.; Drent, E.; Straetemans, T.; Heijhuurs, S.; Scholten, K.; Scheper, W.; Sebestyen, Z.; Martens, A.; et al. γ9 and δ2CDR3 domains regulate functional avidity of T cells harboring γ9δ2TCRs. Blood 2012, 120, 5153–5162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marcu-Malina, V.; Heijhuurs, S.; van Buuren, M.; Hartkamp, L.; Strand, S.; Sebestyen, Z.; Scholten, K.; Martens, A.; Kuball, J. Redirecting αβ T cells against cancer cells by transfer of a broadly tumor-reactive γδT-cell receptor. Blood 2011, 118, 50–59. [Google Scholar] [CrossRef] [PubMed]
- Braham, M.V.J.; Minnema, M.C.; Aarts, T.; Sebestyen, Z.; Straetemans, T.; Vyborova, A.; Kuball, J.; Öner, F.C.; Robin, C.; Alblas, J. Cellular immunotherapy on primary multiple myeloma expanded in a 3D bone marrow niche model. Oncoimmunology 2018, 7, e1434465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Straetemans, T.; Kierkels, G.J.J.; Doorn, R.; Jansen, K.; Heijhuurs, S.; Dos Santos, J.M.; van Muyden, A.D.D.; Vie, H.; Clemenceau, B.; Raymakers, R.; et al. GMP-Grade Manufacturing of T Cells Engineered to Express a Defined γδTCR. Front. Immunol. 2018, 9, 1062. [Google Scholar] [CrossRef]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Cheng, C.; Hou, J.; Qi, X.; Wang, X.; Han, P.; Yang, X. Distinct contribution of PD-L1 suppression by spatial expression of PD-L1 on tumor and non-tumor cells. Cell. Mol. Immunol. 2019, 16, 392–400. [Google Scholar] [CrossRef]
- Yu, G.; Wu, Y.; Wang, W.; Xu, J.; Lv, X.; Cao, X.; Wan, T. Low-dose decitabine enhances the effect of PD-1 blockade in colorectal cancer with microsatellite stability by re-modulating the tumor microenvironment. Cell. Mol. Immunol. 2019, 16, 401–409. [Google Scholar] [CrossRef]
- Li, X.; Song, W.; Shao, C.; Shi, Y.; Han, W. Emerging predictors of the response to the blockade of immune checkpoints in cancer therapy. Cell. Mol. Immunol. 2019, 16, 28–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Lu, H.; Gu, Y.; Zhang, X.; Zhang, G.; Shi, T.; Chen, W. Tim-3 suppresses the killing effect of Vγ9Vδ2 T cells on colon cancer cells by reducing perforin and granzyme B expression. Exp. Cell Res. 2020, 386, 111719. [Google Scholar] [CrossRef]
- Rossi, C.; Gravelle, P.; Decaup, E.; Bordenave, J.; Poupot, M.; Tosolini, M.; Franchini, D.-M.; Laurent, C.; Morin, R.; Lagarde, J.-M.; et al. Boosting γδ T cell-mediated antibody-dependent cellular cytotoxicity by PD-1 blockade in follicular lymphoma. Oncoimmunology 2019, 8, 1554175. [Google Scholar] [CrossRef] [PubMed]
- Mittal, D.; Gubin, M.M.; Schreiber, R.D.; Smyth, M.J. New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape. Curr. Opin. Immunol. 2014, 27, 16–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Acker, H.H.; Anguille, S.; Willemen, Y.; Van den Bergh, J.M.; Berneman, Z.N.; Lion, E.; Smits, E.L.; Van Tendeloo, V.F. Interleukin-15 enhances the proliferation, stimulatory phenotype, and antitumor effector functions of human gamma delta T cells. J. Hematol. Oncol. 2016, 9, 101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cairo, C.; Sagnia, B.; Cappelli, G.; Colizzi, V.; Leke, R.G.F.; Leke, R.J.; Pauza, C.D. Human cord blood γδ T cells expressing public Vγ2 chains dominate the response to bisphosphonate plus interleukin-15. Immunology 2013, 138, 346–360. [Google Scholar] [CrossRef]
Subset | Common Vγ Pairs | Tissue Distribution | Anti-Tumor Effector Molecules |
---|---|---|---|
Vδ1 | Vγ2, Vγ3, Vγ4, Vγ5, Vγ8, Vγ10 | Skin, small intestine, liver, spleen | IFN-γ, TNF-α, Perforin, Granzyme, TRAIL, FASL |
Vδ2 | Vγ9 | Peripheral blood | IFN-γ, TNF-α, Perforin, Granzyme, TRAIL, FASL |
Vδ3 | Not defined | Liver, small intestine | Not defined |
Vδ5 | Vγ4 | Peripheral blood | Not defined |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
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. https://doi.org/10.3390/cells9051206
Liu Y, Zhang C. The Role of Human γδ T Cells in Anti-Tumor Immunity and Their Potential for Cancer Immunotherapy. Cells. 2020; 9(5):1206. https://doi.org/10.3390/cells9051206
Chicago/Turabian StyleLiu, Yuxia, and Cai Zhang. 2020. "The Role of Human γδ T Cells in Anti-Tumor Immunity and Their Potential for Cancer Immunotherapy" Cells 9, no. 5: 1206. https://doi.org/10.3390/cells9051206
APA StyleLiu, Y., & Zhang, C. (2020). The Role of Human γδ T Cells in Anti-Tumor Immunity and Their Potential for Cancer Immunotherapy. Cells, 9(5), 1206. https://doi.org/10.3390/cells9051206