NKG2A Immune Checkpoint in Vδ2 T Cells: Emerging Application in Cancer Immunotherapy
Abstract
:Simple Summary
Abstract
1. Introduction
2. Anti-Tumor Potential of γδ T Cells
3. Vδ2 T Cells as a Source for Cancer Immunotherapies
4. Impact of NKG2A on the Effector Potential of Vδ2 T Cells
5. NKG2A Immune Checkpoint in Vδ2 T Cells
6. NKG2A in Cancer Immunotherapy—The Use of Vδ2 T Cells
7. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
Glossary
ICs | Immune checkpoints |
ICIs | Immune checkpoint inhibitors |
γδ T | Gamma delta T |
αβ T | Alpha beta T |
NK | Natural killer |
TCR | T cell receptor |
MHC | Major histocompatibility complex |
HIV | Human Immunodeficiency virus |
EBV | Epstein–Barr virus |
HBV | Hepatitis B virus |
IFN-g | Interferon-gamma |
TNF-α | Tumor necrosis factor-alpha |
IL | Interleukin |
Ab | Antibody |
PhAg | Phosphoantigen |
ITIMs | Immunoreceptor tyrosine-based inhibition motifs |
HLA | Human leukocyte antigen |
TILs | Tumor infiltrating lymphocytes |
CRC | Colorectal carcinoma |
CLL | Chronic lymphocytic leukemia |
AML | Acute myeloid leukemia |
GBM | Glioblastoma |
CRLM | Liver metastastic colorectal carcinoma |
HCC | Hepatocellular carcinoma |
OC | Ovarian cancer |
IELs | Intraepithelial lymphocytes |
ADCC | Antibody-dependent cellular cytotoxicity |
BTN/BTNs | Butyrophilin family/Butyrophilins |
GvHD | Graft versus Host Disease |
CARs | Chimeric antigen receptors |
BiTEs | Bispecific T cell engagers |
ABP | Aminobisphosphonate |
MM | Multiple myeloma |
NHL | Non-Hodgkin lymphoma |
RCC | Renal cell carcinoma |
ALL | Acute lymphoblastic leukemia |
NSCLC | Non-small cell lung cancer |
TME | Tumor microenvironment |
OS | Overall survival |
HCMV | Human Cytomegalovirus |
mAb | Monoclonal antibody |
EGFR | Epidermal growth factor receptor |
HNSCC | Head and neck squamous cell carcinoma |
References
- Vantourout, P.; Hayday, A. Six-of-the-best: Unique contributions of gammadelta T cells to immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pellicci, D.G.; Koay, H.F.; Berzins, S.P. Thymic development of unconventional T cells: How NKT cells, MAIT cells and gammadelta T cells emerge. Nat. Rev. Immunol. 2020, 20, 756–770. [Google Scholar] [CrossRef] [PubMed]
- Ribot, J.C.; Lopes, N.; Silva-Santos, B. Gammadelta T cells in tissue physiology and surveillance. Nat. Rev. Immunol. 2021, 21, 221–232. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Lin, L.; Xiao, Z.; Li, M.; Wu, X.; Li, W.; Li, X.; Zhao, Q.; Wu, Y.; Zhang, H.; et al. Protective Role of gammadelta T Cells in Different Pathogen Infections and Its Potential Clinical Application. J. Immunol. Res. 2018, 2018, 5081634. [Google Scholar] [CrossRef] [Green Version]
- Silva-Santos, B.; Serre, K.; Norell, H. Gammadelta T cells in cancer. Nat. Rev. Immunol. 2015, 15, 683–691. [Google Scholar] [CrossRef]
- Hudspeth, K.; Fogli, M.; Correia, D.V.; Mikulak, J.; Roberto, A.; Della Bella, S.; Silva-Santos, B.; Mavilio, D. Engagement of NKp30 on Vdelta1 T cells induces the production of CCL3, CCL4, and CCL5 and suppresses HIV-1 replication. Blood 2012, 119, 4013–4016. [Google Scholar] [CrossRef] [Green Version]
- Kuhl, A.A.; Pawlowski, N.N.; Grollich, K.; Blessenohl, M.; Westermann, J.; Zeitz, M.; Loddenkemper, C.; Hoffmann, J.C. Human peripheral gammadelta T cells possess regulatory potential. Immunology 2009, 128, 580–588. [Google Scholar] [CrossRef]
- Zhao, Y.; Niu, C.; Cui, J. Gamma-delta (gammadelta) T cells: Friend or foe in cancer development? J. Transl. Med. 2018, 16, 3, Correction in J. Transl. Med. 2018, 16, 122. [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]
- Rampoldi, F.; Ullrich, L.; Prinz, I. Revisiting the Interaction of gammadelta T-Cells and B-Cells. Cells 2020, 9, 743. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Tian, Q.; Guo, D.; Lin, W.; Xie, X.; Bi, H. Activated gammadelta T Cells Promote Dendritic Cell Maturation and Exacerbate the Development of Experimental Autoimmune Uveitis (EAU) in Mice. Immunol. Invest. 2021, 50, 164–183. [Google Scholar] [CrossRef] [PubMed]
- Deseke, M.; Prinz, I. Ligand recognition by the gammadelta TCR and discrimination between homeostasis and stress conditions. Cell. Mol. Immunol. 2020, 17, 914–924. [Google Scholar] [CrossRef] [PubMed]
- Adams, E.J.; Gu, S.; Luoma, A.M. Human gamma delta T cells: Evolution and ligand recognition. Cell. Immunol. 2015, 296, 31–40. [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 Vdelta2(+) T-cell compartment comprises distinct innate-like Vgamma9(+) and adaptive Vgamma9(-) subsets. Nat. Commun. 2018, 9, 9–1760. [Google Scholar] [CrossRef]
- 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 Vdelta3 T cells. J. Immunol. 2013, 191, 30–34. [Google Scholar] [CrossRef] [Green Version]
- Kenna, T.; Golden-Mason, L.; Norris, S.; Hegarty, J.E.; O’Farrelly, C.; Doherty, D.G. Distinct subpopulations of gamma delta T cells are present in normal and tumor-bearing human liver. Clin. Immunol. 2004, 113, 56–63. [Google Scholar] [CrossRef]
- Bruni, E.; Cimino, M.M.; Donadon, M.; Carriero, R.; Terzoli, S.; Piazza, R.; Ravens, S.; Prinz, I.; Cazzetta, V.; Marzano, P.; et al. Intrahepatic CD69(+)Vdelta1 T cells re-circulate in the blood of patients with metastatic colorectal cancer and limit tumor progression. J. Immunother. Cancer 2022, 10, e004579. [Google Scholar] [CrossRef] [PubMed]
- Marlin, R.; Pappalardo, A.; Kaminski, H.; Willcox, C.R.; Pitard, V.; Netzer, S.; Khairallah, C.; Lomenech, A.M.; Harly, C.; Bonneville, M.; et al. Sensing of cell stress by human gammadelta TCR-dependent recognition of annexin A2. Proc. Natl. Acad. Sci. USA 2017, 114, 3163–3168. [Google Scholar] [CrossRef] [Green Version]
- Wang, L.; Xu, M.; Wang, C.; Zhu, L.; Hu, J.; Chen, S.; Wu, X.; Li, B.; Li, Y. The feature of distribution and clonality of TCR gamma/delta subfamilies T cells in patients with B-cell non-Hodgkin lymphoma. J. Immunol. Res. 2014, 2014, 241246. [Google Scholar] [CrossRef] [Green Version]
- Willcox, C.R.; Pitard, V.; Netzer, S.; Couzi, L.; Salim, M.; Silberzahn, T.; Moreau, J.F.; Hayday, A.C.; Willcox, B.E.; Dechanet-Merville, J. Cytomegalovirus and tumor stress surveillance by binding of a human gammadelta T cell antigen receptor to endothelial protein C receptor. Nat. Immunol. 2012, 13, 872–879. [Google Scholar] [CrossRef]
- Schwartz, J.C.; Gibson, M.S.; Heimeier, D.; Koren, S.; Phillippy, A.M.; Bickhart, D.M.; Smith, T.P.; Medrano, J.F.; Hammond, J.A. The evolution of the natural killer complex; a comparison between mammals using new high-quality genome assemblies and targeted annotation. Immunogenetics 2017, 69, 255–269. [Google Scholar] [CrossRef] [Green Version]
- Huang, R.Y.; Wang, Y.; Jhatakia, A.D.; Deng, A.X.; Bee, C.; Deshpande, S.; Rangan, V.S.; Bezman, N.; Gudmundsson, O.; Chen, G. Higher-Order Structure Characterization of NKG2A/CD94 Protein Complex and Anti-NKG2A Antibody Binding Epitopes by Mass Spectrometry-Based Protein Footprinting Strategies. J. Am. Soc. Mass Spectrom. 2021, 32, 1567–1574. [Google Scholar] [CrossRef]
- Mingari, M.C.; Pietra, G.; Moretta, L. Immune Checkpoint Inhibitors: Anti-NKG2A Antibodies on Board. Trends Immunol. 2019, 40, 83–85. [Google Scholar] [CrossRef] [PubMed]
- Taveirne, S.; Filtjens, J.; Van Ammel, E.; De Colvenaer, V.; Kerre, T.; Taghon, T.; Vandekerckhove, B.; Plum, J.; Held, W.; Leclercq, G. Inhibitory receptors specific for MHC class I educate murine NK cells but not CD8alphaalpha intestinal intraepithelial T lymphocytes. Blood 2011, 118, 339–347. [Google Scholar] [CrossRef] [Green Version]
- Kaiser, B.K.; Pizarro, J.C.; Kerns, J.; Strong, R.K. Structural basis for NKG2A/CD94 recognition of HLA-E. Proc. Natl. Acad. Sci. USA 2008, 105, 6696–6701. [Google Scholar] [CrossRef] [Green Version]
- Long, E.O.; Kim, H.S.; Liu, D.; Peterson, M.E.; Rajagopalan, S. Controlling natural killer cell responses: Integration of signals for activation and inhibition. Annu. Rev. Immunol. 2013, 31, 227–258. [Google Scholar] [CrossRef] [Green Version]
- Joncker, N.T.; Fernandez, N.C.; Treiner, E.; Vivier, E.; Raulet, D.H. NK cell responsiveness is tuned commensurate with the number of inhibitory receptors for self-MHC class I: The rheostat model. J. Immunol. 2009, 182, 4572–4580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ducoin, K.; Oger, R.; Bilonda Mutala, L.; Deleine, C.; Jouand, N.; Desfrancois, J.; Podevin, J.; Duchalais, E.; Cruard, J.; Benlalam, H.; et al. Targeting NKG2A to boost anti-tumor CD8 T-cell responses in human colorectal cancer. Oncoimmunology 2022, 11, 2046931. [Google Scholar] [CrossRef] [PubMed]
- Cazzetta, V.; Bruni, E.; Terzoli, S.; Carenza, C.; Franzese, S.; Piazza, R.; Marzano, P.; Donadon, M.; Torzilli, G.; Cimino, M.; et al. NKG2A expression identifies a subset of human Vdelta2 T cells exerting the highest antitumor effector functions. Cell Rep. 2021, 37, 109871. [Google Scholar] [CrossRef] [PubMed]
- Zaghi, E.; Calvi, M.; Marcenaro, E.; Mavilio, D.; Di Vito, C. Targeting NKG2A to elucidate natural killer cell ontogenesis and to develop novel immune-therapeutic strategies in cancer therapy. J. Leukoc. Biol. 2019, 105, 1243–1251. [Google Scholar] [CrossRef]
- van Montfoort, N.; Borst, L.; Korrer, M.J.; Sluijter, M.; Marijt, K.A.; Santegoets, S.J.; van Ham, V.J.; Ehsan, I.; Charoentong, P.; Andre, P.; et al. NKG2A Blockade Potentiates CD8 T Cell Immunity Induced by Cancer Vaccines. Cell 2018, 175, 1744–1755.e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawamura, T.; Takeda, K.; Kaneda, H.; Matsumoto, H.; Hayakawa, Y.; Raulet, D.H.; Ikarashi, Y.; Kronenberg, M.; Yagita, H.; Kinoshita, K.; et al. NKG2A inhibits invariant NKT cell activation in hepatic injury. J. Immunol. 2009, 182, 250–258. [Google Scholar] [CrossRef] [Green Version]
- Salome, B.; Gomez-Cadena, A.; Loyon, R.; Suffiotti, M.; Salvestrini, V.; Wyss, T.; Vanoni, G.; Ruan, D.F.; Rossi, M.; Tozzo, A.; et al. CD56 as a marker of an ILC1-like population with NK cell properties that is functionally impaired in AML. Blood Adv. 2019, 3, 3674–3687. [Google Scholar] [CrossRef] [PubMed]
- Creelan, B.C.; Antonia, S.J. The NKG2A immune checkpoint—A new direction in cancer immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 277–278. [Google Scholar] [CrossRef] [PubMed]
- Li, D.; Brackenridge, S.; Walters, L.C.; Swanson, O.; Harlos, K.; Rozbesky, D.; Cain, D.W.; Wiehe, K.; Scearce, R.M.; Barr, M.; et al. Mouse and human antibodies bind HLA-E-leader peptide complexes and enhance NK cell cytotoxicity. Commun. Biol. 2022, 5, 271. [Google Scholar] [CrossRef] [PubMed]
- Borst, L.; van der Burg, S.H.; van Hall, T. The NKG2A-HLA-E Axis as a Novel Checkpoint in the Tumor Microenvironment. Clin. Cancer Res. 2020, 26, 5549–5556. [Google Scholar] [CrossRef] [PubMed]
- Haanen, J.B.; Cerundolo, V. NKG2A, a New Kid on the Immune Checkpoint Block. Cell 2018, 175, 1720–1722. [Google Scholar] [CrossRef] [Green Version]
- Gentles, A.J.; Newman, A.M.; Liu, C.L.; Bratman, S.V.; Feng, W.; Kim, D.; Nair, V.S.; Xu, Y.; Khuong, A.; Hoang, C.D.; et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 2015, 21, 938–945. [Google Scholar] [CrossRef] [Green Version]
- Tosolini, M.; Pont, F.; Poupot, M.; Vergez, F.; Nicolau-Travers, M.L.; Vermijlen, D.; Sarry, J.E.; Dieli, F.; Fournie, J.J. Assessment of tumor-infiltrating TCRVgamma9Vdelta2 gammadelta lymphocyte abundance by deconvolution of human cancers microarrays. Oncoimmunology 2017, 6, e1284723. [Google Scholar] [CrossRef]
- Lo Presti, E.; Dieli, F.; Meraviglia, S. Tumor-Infiltrating gammadelta T Lymphocytes: Pathogenic Role, Clinical Significance, and Differential Programing in the Tumor Microenvironment. Front. Immunol. 2014, 5, 607. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Lin, C.; Li, H.; Li, R.; Wu, Y.; Liu, H.; Zhang, H.; He, H.; Zhang, W.; Xu, J. Tumor-infiltrating gammadeltaT cells predict prognosis and adjuvant chemotherapeutic benefit in patients with gastric cancer. Oncoimmunology 2017, 6, e1353858. [Google Scholar] [CrossRef] [PubMed]
- 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 Vdelta1 T cell subpopulation exhibits high antitumor activity against colorectal cancer. JCI Insight 2019, 4, e125884. [Google Scholar] [CrossRef] [Green Version]
- Donia, M.; Ellebaek, E.; Andersen, M.H.; Straten, P.T.; Svane, I.M. Analysis of Vdelta1 T cells in clinical grade melanoma-infiltrating lymphocytes. Oncoimmunology 2012, 1, 1297–1304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, H.; Dai, W.; Guo, J.; Wang, D.; Wen, S.; Yang, L.; Lin, D.; Xie, W.; Wen, L.; Fang, J.; et al. High Abundance of Intratumoral gammadelta T Cells Favors a Better Prognosis in Head and Neck Squamous Cell Carcinoma: A Bioinformatic Analysis. Front. Immunol. 2020, 11, 573920. [Google Scholar] [CrossRef]
- Zhao, N.; Dang, H.; Ma, L.; Martin, S.P.; Forgues, M.; Ylaya, K.; Hewitt, S.M.; Wang, X.W. Intratumoral gammadelta T-Cell Infiltrates, Chemokine (C-C Motif) Ligand 4/Chemokine (C-C Motif) Ligand 5 Protein Expression and Survival in Patients With Hepatocellular Carcinoma. Hepatology 2021, 73, 1045–1060. [Google Scholar] [CrossRef] [PubMed]
- Foord, E.; Arruda, L.C.M.; Gaballa, A.; Klynning, C.; Uhlin, M. Characterization of ascites- and tumor-infiltrating gammadelta T cells reveals distinct repertoires and a beneficial role in ovarian cancer. Sci. Transl. Med. 2021, 13, eabb0192. [Google Scholar] [CrossRef] [PubMed]
- Cordova, A.; Toia, F.; La Mendola, C.; Orlando, V.; Meraviglia, S.; Rinaldi, G.; Todaro, M.; Cicero, G.; Zichichi, L.; Donni, P.L.; et al. Characterization of human gammadelta T lymphocytes infiltrating primary malignant melanomas. PLoS ONE 2012, 7, e49878. [Google Scholar] [CrossRef] [Green Version]
- Tan, L.; Fichtner, A.S.; Bruni, E.; Odak, I.; Sandrock, I.; Bubke, A.; Borchers, A.; Schultze-Florey, C.; Koenecke, C.; Forster, R.; et al. A fetal wave of human type 3 effector gammadelta cells with restricted TCR diversity persists into adulthood. Sci. Immunol. 2021, 6, eabf0125. [Google Scholar] [CrossRef]
- Caccamo, N.; La Mendola, C.; Orlando, V.; Meraviglia, S.; Todaro, M.; Stassi, G.; Sireci, G.; Fournie, J.J.; Dieli, F. Differentiation, phenotype, and function of interleukin-17-producing human Vgamma9Vdelta2 T cells. Blood 2011, 118, 129–138. [Google Scholar] [CrossRef] [Green Version]
- Lo Presti, E.; Toia, F.; Oieni, S.; Buccheri, S.; Turdo, A.; Mangiapane, L.R.; Campisi, G.; Caputo, V.; Todaro, M.; Stassi, G.; et al. Squamous Cell Tumors Recruit gammadelta T Cells Producing either IL17 or IFNgamma Depending on the Tumor Stage. Cancer Immunol. Res. 2017, 5, 397–407. [Google Scholar] [CrossRef] [Green Version]
- Meraviglia, S.; Lo Presti, E.; Tosolini, M.; La Mendola, C.; Orlando, V.; Todaro, M.; Catalano, V.; Stassi, G.; Cicero, G.; Vieni, S.; et al. Distinctive features of tumor-infiltrating gammadelta T lymphocytes in human colorectal cancer. Oncoimmunology 2017, 6, e1347742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, P.; Wu, D.; Ni, C.; Ye, J.; Chen, W.; Hu, G.; Wang, Z.; Wang, C.; Zhang, Z.; Xia, W.; et al. GammadeltaT17 cells promote the accumulation and expansion of myeloid-derived suppressor cells in human colorectal cancer. Immunity 2014, 40, 785–800. [Google Scholar] [CrossRef] [Green Version]
- Van Hede, D.; Polese, B.; Humblet, C.; Wilharm, A.; Renoux, V.; Dortu, E.; de Leval, L.; Delvenne, P.; Desmet, C.J.; Bureau, F.; et al. Human papillomavirus oncoproteins induce a reorganization of epithelial-associated gammadelta T cells promoting tumor formation. Proc. Natl. Acad. Sci. USA 2017, 114, E9056–E9065. [Google Scholar] [CrossRef] [Green Version]
- Patil, R.S.; Shah, S.U.; Shrikhande, S.V.; Goel, M.; Dikshit, R.P.; Chiplunkar, S.V. IL17 producing gammadeltaT cells induce angiogenesis and are associated with poor survival in gallbladder cancer patients. Int. J. Cancer 2016, 139, 869–881. [Google Scholar] [CrossRef]
- Liu, Y.; Zhang, C. The Role of Human gammadelta T Cells in Anti-Tumor Immunity and Their Potential for Cancer Immunotherapy. Cells 2020, 9, 1206. [Google Scholar] [CrossRef]
- Tokuyama, H.; Hagi, T.; Mattarollo, S.R.; Morley, J.; Wang, Q.; So, H.F.; 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]
- Bruni, E.; Cazzetta, V.; Donadon, M.; Cimino, M.; Torzilli, G.; Spata, G.; Leonardi, G.; Dieli, F.; Mikulak, J.; Mavilio, D. Chemotherapy accelerates immune-senescence and functional impairments of Vdelta2(pos) T cells in elderly patients affected by liver metastatic colorectal cancer. J. Immunother. Cancer 2019, 7, 347. [Google Scholar] [CrossRef] [Green Version]
- Li, Z.; Peng, H.; Xu, Q.; Ye, Z. Sensitization of human osteosarcoma cells to Vgamma9Vdelta2 T-cell-mediated cytotoxicity by zoledronate. J. Orthop. Res. 2012, 30, 824–830. [Google Scholar] [CrossRef] [PubMed]
- Wu, D.; Wu, P.; Wu, X.; Ye, J.; Wang, Z.; Zhao, S.; Ni, C.; Hu, G.; Xu, J.; Han, Y.; et al. Ex vivo expanded human circulating Vdelta1 gammadeltaT cells exhibit favorable therapeutic potential for colon cancer. Oncoimmunology 2015, 4, e992749. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halary, F.; Pitard, V.; Dlubek, D.; Krzysiek, R.; de la Salle, H.; Merville, P.; Dromer, C.; Emilie, D.; Moreau, J.F.; Dechanet-Merville, J. Shared reactivity of Vdelta2(neg) gammadelta T cells against cytomegalovirus-infected cells and tumor intestinal epithelial cells. J. Exp. Med. 2005, 201, 1567–1578. [Google Scholar] [CrossRef] [Green Version]
- 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. Immunol. 2010, 184, 3260–3268. [Google Scholar] [CrossRef] [Green Version]
- Wrobel, P.; Shojaei, H.; Schittek, B.; Gieseler, F.; Wollenberg, B.; Kalthoff, H.; Kabelitz, D.; Wesch, D. Lysis of a broad range of epithelial tumour cells by human gamma delta T cells: Involvement of NKG2D ligands and T-cell receptor- versus NKG2D-dependent recognition. Scand. J. Immunol. 2007, 66, 320–328. [Google Scholar] [CrossRef] [PubMed]
- Rincon-Orozco, B.; Kunzmann, V.; Wrobel, P.; Kabelitz, D.; Steinle, A.; Herrmann, T. Activation of V gamma 9V delta 2 T cells by NKG2D. J. Immunol. 2005, 175, 2144–2151. [Google Scholar] [CrossRef] [Green Version]
- Siegers, G.M.; Lamb, L.S., Jr. Cytotoxic and regulatory properties of circulating Vdelta1+ gammadelta T cells: A new player on the cell therapy field? Mol. Ther. 2014, 22, 1416–1422. [Google Scholar] [CrossRef] [Green Version]
- Correia, D.V.; Lopes, A.; Silva-Santos, B. Tumor cell recognition by gammadelta T lymphocytes: T-cell receptor vs. NK-cell receptors. Oncoimmunology 2013, 2, e22892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deniger, D.C.; Maiti, S.N.; Mi, T.; Switzer, K.C.; Ramachandran, V.; Hurton, L.V.; Ang, S.; Olivares, S.; Rabinovich, B.A.; Huls, M.H.; et al. Activating and propagating polyclonal gamma delta T cells with broad specificity for malignancies. Clin. Cancer Res. 2014, 20, 5708–5719. [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 Vdelta1+ T cells expressing the natural cytotoxicity receptor NKp30 for recognition of lymphoid leukemia cells. Blood 2011, 118, 992–1001. [Google Scholar] [CrossRef] [Green Version]
- Fichtner, A.S.; Ravens, S.; Prinz, I. Human gammadelta TCR Repertoires in Health and Disease. Cells 2020, 9, 800. [Google Scholar] [CrossRef] [Green Version]
- Morita, C.T.; Jin, C.; Sarikonda, G.; Wang, H. Nonpeptide antigens, presentation mechanisms, and immunological memory of human Vgamma2Vdelta2 T cells: Discriminating friend from foe through the recognition of prenyl pyrophosphate antigens. Immunol. Rev. 2007, 215, 59–76. [Google Scholar] [CrossRef]
- Gober, H.J.; Kistowska, M.; Angman, L.; Jeno, P.; Mori, L.; De Libero, G. Human T cell receptor gammadelta cells recognize endogenous mevalonate metabolites in tumor cells. J. Exp. Med. 2003, 197, 163–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vantourout, P.; Mookerjee-Basu, J.; Rolland, C.; Pont, F.; Martin, H.; Davrinche, C.; Martinez, L.O.; Perret, B.; Collet, X.; Perigaud, C.; et al. Specific requirements for Vgamma9Vdelta2 T cell stimulation by a natural adenylated phosphoantigen. J. Immunol. 2009, 183, 3848–3857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herrmann, T.; Karunakaran, M.M. Butyrophilins: Gammadelta T Cell Receptor Ligands, Immunomodulators and More. Front. Immunol. 2022, 13, 876493. [Google Scholar] [CrossRef] [PubMed]
- Blazquez, J.L.; Benyamine, A.; Pasero, C.; Olive, D. New Insights Into the Regulation of gammadelta T Cells by BTN3A and Other BTN/BTNL in Tumor Immunity. Front. Immunol. 2018, 9, 1601. [Google Scholar] [CrossRef] [Green Version]
- Handgretinger, R.; Schilbach, K. The potential role of gammadelta T cells after allogeneic HCT for leukemia. Blood 2018, 131, 1063–1072. [Google Scholar] [CrossRef] [Green Version]
- Minculescu, L.; Marquart, H.V.; Ryder, L.P.; Andersen, N.S.; Schjoedt, I.; Friis, L.S.; Kornblit, B.T.; Petersen, S.L.; Haastrup, E.; Fischer-Nielsen, A.; et al. Improved Overall Survival, Relapse-Free-Survival, and Less Graft-vs.-Host-Disease in Patients With High Immune Reconstitution of TCR Gamma Delta Cells 2 Months After Allogeneic Stem Cell Transplantation. Front. Immunol. 2019, 10, 1997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, K.F.; Duarte, J.D.G.; Ostrouska, S.; Behren, A. gammadelta T Cells in the Tumor Microenvironment-Interactions With Other Immune Cells. Front. Immunol. 2022, 13, 894315. [Google Scholar] [CrossRef] [PubMed]
- Sebestyen, Z.; Prinz, I.; Dechanet-Merville, J.; Silva-Santos, B.; Kuball, J. Translating gammadelta (gammadelta) T cells and their receptors into cancer cell therapies. Nat. Rev. Drug Discov. 2020, 19, 169–184. [Google Scholar] [CrossRef] [Green Version]
- Sullivan, L.C.; Shaw, E.M.; Westall, G.P. gammadelta T Cells in Transplantation: Friend and Foe. Transplantation 2018, 102, 1970–1971. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Legut, M.; Cole, D.K.; Sewell, A.K. The promise of gammadelta T cells and the gammadelta T cell receptor for cancer immunotherapy. Cell. Mol. Immunol. 2015, 12, 656–668. [Google Scholar] [CrossRef] [PubMed]
- Park, J.H.; Lee, H.K. Function of gammadelta T cells in tumor immunology and their application to cancer therapy. Exp. Mol. Med. 2021, 53, 318–327. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.; Rosenthal, C.J.; Penn, N.E.; Dunn, Z.S.; Zhou, Y.; Yang, L. Human gammadelta T Cell Subsets and Their Clinical Applications for Cancer Immunotherapy. Cancers 2022, 14, 3005. [Google Scholar] [CrossRef]
- Kabelitz, D.; Serrano, R.; Kouakanou, L.; Peters, C.; Kalyan, S. Cancer immunotherapy with gammadelta T cells: Many paths ahead of us. Cell. Mol. Immunol. 2020, 17, 925–939. [Google Scholar] [CrossRef]
- Miyashita, M.; Shimizu, T.; Ashihara, E.; Ukimura, O. Strategies to Improve the Antitumor Effect of gammadelta T Cell Immunotherapy for Clinical Application. Int. J. Mol. Sci. 2021, 22, 8910. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Peters, C.; Kouakanou, L.; Oberg, H.H.; Wesch, D.; Kabelitz, D. In vitro expansion of Vgamma9Vdelta2 T cells for immunotherapy. Methods Enzymol. 2020, 631, 223–237. [Google Scholar] [CrossRef]
- Kunzmann, V.; Bauer, E.; Feurle, J.; Weissinger, F.; Tony, H.P.; Wilhelm, M. Stimulation of gammadelta T cells by aminobisphosphonates and induction of antiplasma cell activity in multiple myeloma. Blood 2000, 96, 384–392. [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]
- Dieli, F.; Vermijlen, D.; Fulfaro, F.; Caccamo, N.; Meraviglia, S.; Cicero, G.; Roberts, A.; Buccheri, S.; D’Asaro, M.; Gebbia, N.; et al. Targeting human γδ T cells with zoledronate and interleukin-2 for immunotherapy of hormone-refractory prostate cancer. Cancer Res. 2007, 67, 7450–7457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bennouna, J.; Levy, V.; Sicard, H.; Senellart, H.; Audrain, M.; Hiret, S.; Rolland, F.; Bruzzoni-Giovanelli, H.; Rimbert, M.; Galea, 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]
- 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] [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 gammadelta T cells as treatment for patients with refractory renal cell carcinoma. Cancer Immunol. Immunother. 2011, 60, 1447–1460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kunzmann, V.; Smetak, M.; Kimmel, B.; Weigang-Koehler, K.; Goebeler, M.; Birkmann, J.; Becker, J.; Schmidt-Wolf, I.G.; Einsele, H.; Wilhelm, M. Tumor-promoting versus tumor-antagonizing roles of gammadelta T cells in cancer immunotherapy: Results from a prospective phase I/II trial. J. Immunother. 2012, 35, 205–213. [Google Scholar] [CrossRef] [PubMed]
- Pressey, J.G.; Adams, J.; Harkins, L.; Kelly, D.; You, Z.; Lamb, L.S., Jr. In vivo expansion and activation of gammadelta T cells as immunotherapy for refractory neuroblastoma: A phase 1 study. Medicine 2016, 95, e4909. [Google Scholar] [CrossRef]
- Bregeon, D.; Ferron, L.; Chretien, A.; Guillen, F.; Zgonnik, V.; Rivaud, M.; Mazieres, M.R.; Coquerel, G.; Belmant, C.; Plaquevent, J.C. Synthesis of phosphoantigens: Scalable accesses to enantiomers of BrHPP and studies on N-HDMAPP synthesis. Bioorg. Med. Chem. Lett. 2012, 22, 5807–5810. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Bennouna, J.; Bompas, E.; Neidhardt, E.M.; Rolland, F.; Philip, I.; Galea, 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] [PubMed]
- 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. Cardiothorac. Surg. 2010, 37, 1191–1197. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, H.; Tanaka, Y.; Yagi, J.; Minato, N.; Tanabe, K. Phase I/II study of adoptive transfer of gammadelta 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]
- 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] [Green Version]
- Noguchi, A.; Kaneko, T.; Kamigaki, T.; Fujimoto, K.; Ozawa, M.; Saito, M.; Ariyoshi, N.; Goto, S. Zoledronate-activated Vgamma9gammadelta T cell-based immunotherapy is feasible and restores the impairment of gammadelta T cells in patients with solid tumors. Cytotherapy 2011, 13, 92–97. [Google Scholar] [CrossRef]
- 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 gammadeltaTcells: A phase I clinical study. J. Immunother. 2011, 34, 202–211. [Google Scholar] [CrossRef] [PubMed]
- Wada, I.; Matsushita, H.; Noji, S.; Mori, K.; Yamashita, H.; Nomura, S.; Shimizu, N.; Seto, Y.; Kakimi, K. Intraperitoneal injection of in vitro expanded Vgamma9Vdelta2 T cells together with zoledronate for the treatment of malignant ascites due to gastric cancer. Cancer Med. 2014, 3, 362–375. [Google Scholar] [CrossRef]
- Aoki, T.; Matsushita, H.; Hoshikawa, M.; Hasegawa, K.; Kokudo, N.; Kakimi, K. Adjuvant combination therapy with gemcitabine and autologous gammadelta T-cell transfer in patients with curatively resected pancreatic cancer. Cytotherapy 2017, 19, 473–485. [Google Scholar] [CrossRef]
- De Gassart, A.; Le, K.S.; Brune, P.; Agaugue, S.; Sims, J.; Goubard, A.; Castellano, R.; Joalland, N.; Scotet, E.; Collette, Y.; et al. Development of ICT01, a first-in-class, anti-BTN3A antibody for activating Vgamma9Vdelta2 T cell-mediated antitumor immune response. Sci. Transl. Med. 2021, 13, eabj0835. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
- Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Catafal-Tardos, E.; Baglioni, M.V.; Bekiaris, V. Inhibiting the Unconventionals: Importance of Immune Checkpoint Receptors in gammadelta T, MAIT, and NKT Cells. Cancers 2021, 13, 4647. [Google Scholar] [CrossRef] [PubMed]
- Weimer, P.; Wellbrock, J.; Sturmheit, T.; Oliveira-Ferrer, L.; Ding, Y.; Menzel, S.; Witt, M.; Hell, L.; Schmalfeldt, B.; Bokemeyer, C.; et al. Tissue-Specific Expression of TIGIT, PD-1, TIM-3, and CD39 by gammadelta T Cells in Ovarian Cancer. Cells 2022, 11, 964. [Google Scholar] [CrossRef]
- Gertner-Dardenne, J.; Fauriat, C.; Orlanducci, F.; Thibult, M.L.; Pastor, S.; Fitzgibbon, J.; Bouabdallah, R.; Xerri, L.; Olive, D. The co-receptor BTLA negatively regulates human Vgamma9Vdelta2 T-cell proliferation: A potential way of immune escape for lymphoma cells. Blood 2013, 122, 922–931. [Google Scholar] [CrossRef] [Green Version]
- Dondero, A.; Pastorino, F.; Della Chiesa, M.; Corrias, M.V.; Morandi, F.; Pistoia, V.; Olive, D.; Bellora, F.; Locatelli, F.; Castellano, A.; et al. PD-L1 expression in metastatic neuroblastoma as an additional mechanism for limiting immune surveillance. Oncoimmunology 2016, 5, e1064578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hu, G.; Wu, P.; Cheng, P.; Zhang, Z.; Wang, Z.; Yu, X.; Shao, X.; Wu, D.; Ye, J.; Zhang, T.; et al. Tumor-infiltrating CD39(+)gammadeltaTregs are novel immunosuppressive T cells in human colorectal cancer. Oncoimmunology 2017, 6, e1277305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castella, B.; Foglietta, M.; Sciancalepore, P.; Rigoni, M.; Coscia, M.; Griggio, V.; Vitale, C.; Ferracini, R.; Saraci, E.; Omede, P.; et al. Anergic bone marrow Vgamma9Vdelta2 T cells as early and long-lasting markers of PD-1-targetable microenvironment-induced immune suppression in human myeloma. Oncoimmunology 2015, 4, e1047580. [Google Scholar] [CrossRef] [PubMed]
- 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 gammadelta T cell-mediated antibody-dependent cellular cytotoxicity by PD-1 blockade in follicular lymphoma. Oncoimmunology 2019, 8, 1554175. [Google Scholar] [CrossRef]
- Hoeres, T.; Holzmann, E.; Smetak, M.; Birkmann, J.; Wilhelm, M. PD-1 signaling modulates interferon-gamma production by Gamma Delta (gammadelta) T-Cells in response to leukemia. Oncoimmunology 2019, 8, 1550618. [Google Scholar] [CrossRef] [Green Version]
- Girard, P.; Charles, J.; Cluzel, C.; Degeorges, E.; Manches, O.; Plumas, J.; De Fraipont, F.; Leccia, M.T.; Mouret, S.; Chaperot, L.; et al. The features of circulating and tumor-infiltrating gammadelta T cells in melanoma patients display critical perturbations with prognostic impact on clinical outcome. Oncoimmunology 2019, 8, 1601483. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Lu, H.; Gu, Y.; Zhang, X.; Zhang, G.; Shi, T.; Chen, W. Tim-3 suppresses the killing effect of Vgamma9Vdelta2T cells on colon cancer cells by reducing perforin and granzyme B expression. Exp. Cell. Res. 2020, 386, 111719. [Google Scholar] [CrossRef] [PubMed]
- Wu, K.; Feng, J.; Xiu, Y.; Li, Z.; Lin, Z.; Zhao, H.; Zeng, H.; Xia, W.; Yu, L.; Xu, B. Vdelta2 T cell subsets, defined by PD-1 and TIM-3 expression, present varied cytokine responses in acute myeloid leukemia patients. Int. Immunopharmacol. 2020, 80, 106122. [Google Scholar] [CrossRef]
- Wu, K.; Zhao, H.; Xiu, Y.; Li, Z.; Zhao, J.; Xie, S.; Zeng, H.; Zhang, H.; Yu, L.; Xu, B. IL-21-mediated expansion of Vgamma9Vdelta2 T cells is limited by the Tim-3 pathway. Int. Immunopharmacol. 2019, 69, 136–142. [Google Scholar] [CrossRef]
- Halary, F.; Peyrat, M.A.; Champagne, E.; Lopez-Botet, M.; Moretta, A.; Moretta, L.; Vie, H.; Fournie, J.J.; Bonneville, M. Control of self-reactive cytotoxic T lymphocytes expressing gamma delta T cell receptors by natural killer inhibitory receptors. Eur. J. Immunol. 1997, 27, 2812–2821. [Google Scholar] [CrossRef]
- Poccia, F.; Cipriani, B.; Vendetti, S.; Colizzi, V.; Poquet, Y.; Battistini, L.; Lopez-Botet, M.; Fournie, J.J.; Gougeon, M.L. CD94/NKG2 inhibitory receptor complex modulates both anti-viral and anti-tumoral responses of polyclonal phosphoantigen-reactive V gamma 9V delta 2 T lymphocytes. J. Immunol. 1997, 159, 6009–6017. [Google Scholar] [CrossRef] [PubMed]
- Wragg, K.M.; Tan, H.X.; Kristensen, A.B.; Nguyen-Robertson, C.V.; Kelleher, A.D.; Parsons, M.S.; Wheatley, A.K.; Berzins, S.P.; Pellicci, D.G.; Kent, S.J.; et al. High CD26 and Low CD94 Expression Identifies an IL-23 Responsive Vdelta2(+) T Cell Subset with a MAIT Cell-like Transcriptional Profile. Cell Rep. 2020, 31, 107773. [Google Scholar] [CrossRef] [PubMed]
- Pizzolato, G.; Kaminski, H.; Tosolini, M.; Franchini, D.M.; Pont, F.; Martins, F.; Valle, C.; Labourdette, D.; Cadot, S.; Quillet-Mary, A.; et al. Single-cell RNA sequencing unveils the shared and the distinct cytotoxic hallmarks of human TCRVdelta1 and TCRVdelta2 gammadelta T lymphocytes. Proc. Natl. Acad. Sci. USA 2019, 116, 11906–11915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, L.; Fichtner, A.S.; Bubke, A.; Odak, I.; Schultze-Florey, C.; Koenecke, C.; Förster, R.; Jarek, M.; von Kaisenberg, C.; Borchers, A.; et al. A fetal wave of human type-3 γδ T cells with restricted TCR diversity persists into adulthood. bioRxiv 2020. [Google Scholar] [CrossRef] [PubMed]
- Kaiser, B.K.; Barahmand-Pour, F.; Paulsene, W.; Medley, S.; Geraghty, D.E.; Strong, R.K. Interactions between NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J. Immunol. 2005, 174, 2878–2884. [Google Scholar] [CrossRef] [Green Version]
- Di Vito, C.; Mikulak, J.; Mavilio, D. On the Way to Become a Natural Killer Cell. Front. Immunol. 2019, 10, 1812. [Google Scholar] [CrossRef]
- Del Zotto, G.; Marcenaro, E.; Vacca, P.; Sivori, S.; Pende, D.; Della Chiesa, M.; Moretta, F.; Ingegnere, T.; Mingari, M.C.; Moretta, A.; et al. Markers and function of human NK cells in normal and pathological conditions. Cytom. B Clin. Cytom. 2017, 92, 100–114. [Google Scholar] [CrossRef] [Green Version]
- Sanchez Sanchez, G.; Papadopoulou, M.; Azouz, A.; Tafesse, Y.; Mishra, A.; Chan, J.K.Y.; Fan, Y.; Verdebout, I.; Porco, S.; Libert, F.; et al. Identification of distinct functional thymic programming of fetal and pediatric human gammadelta thymocytes via single-cell analysis. Nat. Commun. 2022, 13, 5842. [Google Scholar] [CrossRef]
- Papadopoulou, M.; Dimova, T.; Shey, M.; Briel, L.; Veldtsman, H.; Khomba, N.; Africa, H.; Steyn, M.; Hanekom, W.A.; Scriba, T.J.; et al. Fetal public Vgamma9Vdelta2 T cells expand and gain potent cytotoxic functions early after birth. Proc. Natl. Acad. Sci. USA 2020, 117, 18638–18648. [Google Scholar] [CrossRef]
- McMahon, C.W.; Raulet, D.H. Expression and function of NK cell receptors in CD8+ T cells. Curr. Opin. Immunol. 2001, 13, 465–470. [Google Scholar] [CrossRef]
- He, Y.; Tian, Z. NK cell education via nonclassical MHC and non-MHC ligands. Cell. Mol. Immunol. 2017, 14, 321–330. [Google Scholar] [CrossRef] [Green Version]
- Held, W. Nonclassical NK cell education. Nat. Immunol. 2012, 13, 1135–1137. [Google Scholar] [CrossRef]
- Shreeve, N.; Depierreux, D.; Hawkes, D.; Traherne, J.A.; Sovio, U.; Huhn, O.; Jayaraman, J.; Horowitz, A.; Ghadially, H.; Perry, J.R.B.; et al. The CD94/NKG2A inhibitory receptor educates uterine NK cells to optimize pregnancy outcomes in humans and mice. Immunity 2021, 54, 1231–1244.e4. [Google Scholar] [CrossRef] [PubMed]
- Ivarsson, M.A.; Loh, L.; Marquardt, N.; Kekalainen, E.; Berglin, L.; Bjorkstrom, N.K.; Westgren, M.; Nixon, D.F.; Michaelsson, J. Differentiation and functional regulation of human fetal NK cells. J. Clin. Invest. 2013, 123, 3889–3901. [Google Scholar] [CrossRef] [Green Version]
- Braud, V.M.; Allan, D.S.; O’Callaghan, C.A.; Soderstrom, K.; D’Andrea, A.; Ogg, G.S.; Lazetic, S.; Young, N.T.; Bell, J.I.; Phillips, J.H.; et al. HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C. Nature 1998, 391, 795–799. [Google Scholar] [CrossRef] [PubMed]
- Yunis, E.J.; Romero, V.; Diaz-Giffero, F.; Zuniga, J.; Koka, P. Natural Killer Cell Receptor NKG2A/HLA-E Interaction Dependent Differential Thymopoiesis of Hematopoietic Progenitor Cells Influences the Outcome of HIV Infection. J. Stem Cells 2007, 2, 237–248. [Google Scholar]
- Vales-Gomez, M.; Reyburn, H.T.; Erskine, R.A.; Lopez-Botet, M.; Strominger, J.L. Kinetics and peptide dependency of the binding of the inhibitory NK receptor CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. EMBO J. 1999, 18, 4250–4260. [Google Scholar] [CrossRef] [PubMed]
- Horowitz, A.; Djaoud, Z.; Nemat-Gorgani, N.; Blokhuis, J.; Hilton, H.G.; Beziat, V.; Malmberg, K.J.; Norman, P.J.; Guethlein, L.A.; Parham, P. Class I HLA haplotypes form two schools that educate NK cells in different ways. Sci. Immunol. 2016, 1, eaag1672. [Google Scholar] [CrossRef] [Green Version]
- Hallner, A.; Bernson, E.; Hussein, B.A.; Ewald Sander, F.; Brune, M.; Aurelius, J.; Martner, A.; Hellstrand, K.; Thoren, F.B. The HLA-B -21 dimorphism impacts on NK cell education and clinical outcome of immunotherapy in acute myeloid leukemia. Blood 2019, 133, 1479–1488. [Google Scholar] [CrossRef] [Green Version]
- Petersdorf, E.W.; Carrington, M.; O’HUigin, C.; Bengtsson, M.; De Santis, D.; Dubois, V.; Gooley, T.; Horowitz, M.; Hsu, K.; Madrigal, J.A.; et al. Role of HLA-B exon 1 in graft-versus-host disease after unrelated haemopoietic cell transplantation: A retrospective cohort study. Lancet Haematol. 2020, 7, e50–e60. [Google Scholar] [CrossRef]
- Qu, G.; Wang, S.; Zhou, Z.; Jiang, D.; Liao, A.; Luo, J. Comparing Mouse and Human Tissue-Resident gammadelta T Cells. Front. Immunol. 2022, 13, 891687. [Google Scholar] [CrossRef] [PubMed]
- Angelini, D.F.; Zambello, R.; Galandrini, R.; Diamantini, A.; Placido, R.; Micucci, F.; Poccia, F.; Semenzato, G.; Borsellino, G.; Santoni, A.; et al. NKG2A inhibits NKG2C effector functions of gammadelta T cells: Implications in health and disease. J. Leukoc. Biol. 2011, 89, 75–84. [Google Scholar] [CrossRef] [PubMed]
- Trichet, V.; Benezech, C.; Dousset, C.; Gesnel, M.C.; Bonneville, M.; Breathnach, R. Complex interplay of activating and inhibitory signals received by Vgamma9Vdelta2 T cells revealed by target cell beta2-microglobulin knockdown. J. Immunol. 2006, 177, 6129–6136. [Google Scholar] [CrossRef] [Green Version]
- Bhagat, G.; Naiyer, A.J.; Shah, J.G.; Harper, J.; Jabri, B.; Wang, T.C.; Green, P.H.; Manavalan, J.S. Small intestinal CD8+TCRgammadelta+NKG2A+ intraepithelial lymphocytes have attributes of regulatory cells in patients with celiac disease. J. Clin. Invest. 2008, 118, 281–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Borrego, F.; Ulbrecht, M.; Weiss, E.H.; Coligan, J.E.; Brooks, A.G. Recognition of human histocompatibility leukocyte antigen (HLA)-E complexed with HLA class I signal sequence-derived peptides by CD94/NKG2 confers protection from natural killer cell-mediated lysis. J. Exp. Med. 1998, 187, 813–818. [Google Scholar] [CrossRef] [Green Version]
- Xu, W.; Monaco, G.; Wong, E.H.; Tan, W.L.W.; Kared, H.; Simoni, Y.; Tan, S.W.; How, W.Z.Y.; Tan, C.T.Y.; Lee, B.T.K.; et al. Mapping of gamma/delta T cells reveals Vdelta2+ T cells resistance to senescence. EBioMedicine 2019, 39, 44–58. [Google Scholar] [CrossRef] [Green Version]
- Marin, R.; Ruiz-Cabello, F.; Pedrinaci, S.; Mendez, R.; Jimenez, P.; Geraghty, D.E.; Garrido, F. Analysis of HLA-E expression in human tumors. Immunogenetics 2003, 54, 767–775. [Google Scholar] [CrossRef]
- Allard, M.; Oger, R.; Vignard, V.; Percier, J.M.; Fregni, G.; Perier, A.; Caignard, A.; Charreau, B.; Bernardeau, K.; Khammari, A.; et al. Serum soluble HLA-E in melanoma: A new potential immune-related marker in cancer. PLoS ONE 2011, 6, e21118. [Google Scholar] [CrossRef]
- Levy, E.M.; Bianchini, M.; Von Euw, E.M.; Barrio, M.M.; Bravo, A.I.; Furman, D.; Domenichini, E.; Macagno, C.; Pinsky, V.; Zucchini, C.; et al. Human leukocyte antigen-E protein is overexpressed in primary human colorectal cancer. Int. J. Oncol. 2008, 32, 633–641. [Google Scholar] [CrossRef] [Green Version]
- Zheng, H.; Lu, R.; Xie, S.; Wen, X.; Wang, H.; Gao, X.; Guo, L. Human leukocyte antigen-E alleles and expression in patients with serous ovarian cancer. Cancer Sci. 2015, 106, 522–528. [Google Scholar] [CrossRef] [Green Version]
- Kren, L.; Slaby, O.; Muckova, K.; Lzicarova, E.; Sova, M.; Vybihal, V.; Svoboda, T.; Fadrus, P.; Lakomy, R.; Vanhara, P.; et al. Expression of immune-modulatory molecules HLA-G and HLA-E by tumor cells in glioblastomas: An unexpected prognostic significance? Neuropathology 2011, 31, 129–134. [Google Scholar] [CrossRef]
- Bossard, C.; Bezieau, S.; Matysiak-Budnik, T.; Volteau, C.; Laboisse, C.L.; Jotereau, F.; Mosnier, J.F. HLA-E/beta2 microglobulin overexpression in colorectal cancer is associated with recruitment of inhibitory immune cells and tumor progression. Int. J. Cancer 2012, 131, 855–863. [Google Scholar] [CrossRef]
- Eugene, J.; Jouand, N.; Ducoin, K.; Dansette, D.; Oger, R.; Deleine, C.; Leveque, E.; Meurette, G.; Podevin, J.; Matysiak, T.; et al. The inhibitory receptor CD94/NKG2A on CD8(+) tumor-infiltrating lymphocytes in colorectal cancer: A promising new druggable immune checkpoint in the context of HLAE/beta2m overexpression. Mod. Pathol. 2020, 33, 468–482. [Google Scholar] [CrossRef]
- Seliger, B.; Jasinski-Bergner, S.; Quandt, D.; Stoehr, C.; Bukur, J.; Wach, S.; Legal, W.; Taubert, H.; Wullich, B.; Hartmann, A. HLA-E expression and its clinical relevance in human renal cell carcinoma. Oncotarget 2016, 7, 67360–67372. [Google Scholar] [CrossRef] [Green Version]
- Talebian Yazdi, M.; van Riet, S.; van Schadewijk, A.; Fiocco, M.; van Hall, T.; Taube, C.; Hiemstra, P.S.; van der Burg, S.H. The positive prognostic effect of stromal CD8+ tumor-infiltrating T cells is restrained by the expression of HLA-E in non-small cell lung carcinoma. Oncotarget 2016, 7, 3477–3488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gooden, M.; Lampen, M.; Jordanova, E.S.; Leffers, N.; Trimbos, J.B.; van der Burg, S.H.; Nijman, H.; van Hall, T. HLA-E expression by gynecological cancers restrains tumor-infiltrating CD8(+) T lymphocytes. Proc. Natl. Acad. Sci. USA 2011, 108, 10656–10661. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andersson, E.; Poschke, I.; Villabona, L.; Carlson, J.W.; Lundqvist, A.; Kiessling, R.; Seliger, B.; Masucci, G.V. Non-classical HLA-class I expression in serous ovarian carcinoma: Correlation with the HLA-genotype, tumor infiltrating immune cells and prognosis. Oncoimmunology 2016, 5, e1052213. [Google Scholar] [CrossRef] [Green Version]
- de Kruijf, E.M.; Sajet, A.; van Nes, J.G.; Natanov, R.; Putter, H.; Smit, V.T.; Liefers, G.J.; van den Elsen, P.J.; van de Velde, C.J.; Kuppen, P.J. HLA-E and HLA-G expression in classical HLA class I-negative tumors is of prognostic value for clinical outcome of early breast cancer patients. J. Immunol. 2010, 185, 7452–7459. [Google Scholar] [CrossRef] [Green Version]
- Sun, C.; Xu, J.; Huang, Q.; Huang, M.; Wen, H.; Zhang, C.; Wang, J.; Song, J.; Zheng, M.; Sun, H.; et al. High NKG2A expression contributes to NK cell exhaustion and predicts a poor prognosis of patients with liver cancer. Oncoimmunology 2017, 6, e1264562. [Google Scholar] [CrossRef] [Green Version]
- van Hall, T.; Andre, P.; Horowitz, A.; Ruan, D.F.; Borst, L.; Zerbib, R.; Narni-Mancinelli, E.; van der Burg, S.H.; Vivier, E. Monalizumab: Inhibiting the novel immune checkpoint NKG2A. J. Immunother. Cancer 2019, 7, 263. [Google Scholar] [CrossRef] [PubMed]
- Andre, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Blery, M.; Bonnafous, C.; Gauthier, L.; Morel, A.; et al. Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells. Cell 2018, 175, 1731–1743.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tinker, A.V.; Hirte, H.W.; Provencher, D.; Butler, M.; Ritter, H.; Tu, D.; Azim, H.A., Jr.; Paralejas, P.; Grenier, N.; Hahn, S.A.; et al. Dose-Ranging and Cohort-Expansion Study of Monalizumab (IPH2201) in Patients with Advanced Gynecologic Malignancies: A Trial of the Canadian Cancer Trials Group (CCTG): IND221. Clin. Cancer Res. 2019, 25, 6052–6060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lentz, R.W.; Colton, M.D.; Mitra, S.S.; Messersmith, W.A. Innate Immune Checkpoint Inhibitors: The Next Breakthrough in Medical Oncology? Mol. Cancer Ther. 2021, 20, 961–974. [Google Scholar] [CrossRef] [PubMed]
- Spinosa, P.; Musial-Siwek, M.; Presler, M.; Betts, A.; Rosentrater, E.; Villali, J.; Wille, L.; Zhao, Y.; McCaughtry, T.; Subramanian, K.; et al. Quantitative modeling predicts competitive advantages of a next generation anti-NKG2A monoclonal antibody over monalizumab for the treatment of cancer. CPT Pharmacomet. Syst. Pharmacol. 2021, 10, 220–229. [Google Scholar] [CrossRef]
Feature | γδ T Cells | αβ T Cells | ||
---|---|---|---|---|
Subpopulations | Vδ1 | Vδ2 | CD4 | CD8 |
Development | Thymus, common precursor | |||
TCR | TCRγδCD3ε2γδζ2 | TCRαβCD3ε2γδζ2 | ||
TCR repertoire | Variant Restricted number of γ and δ chains | Semi-invariant Vγ9Vδ2 | Variant | |
Frequency | 25–40% peripheral tissues | 1–10% PBMCs | 25–60% PBMCs | 5–30% PBMCs |
MHC restriction | No MHC restriction | MHC class II | MHC class I | |
TCR ligands | Unprocessed unkown | Unprocessed PhAg | Processed peptides | |
Response | Innate/Adaptive | Adaptive |
Clinical Trial | Phase | Status | Drug | Disease |
---|---|---|---|---|
NCT04307329 | II | Active, not recruiting | Monalizumab + Trastuzumab | Breast cancer |
NCT04590963 | III | Terminated | Monalizumab + Cetuximab | Squamous Cell Carcinoma of the Head and Neck |
NCT05221840 | III | Recruiting | Monalizumab + Durvalumab | Non-Small Cell Lung Cancer |
NCT02921685 | I | Unknown status | Monalizumab | Hematologic Malignancies |
NCT02671435 | I/II | Completed | Monalizumab + Durvalumab | Advanced Solid Tumors |
NCT02557516 | I/II | Terminated | Monalizumab + Ibrutinib | Chronic Lymphocytic Leukemia |
NCT05414032 | II | Not yet recruiting | Monalizumab + Cetuximab | Locoregionally Advanced Head and Neck Squamous Cell Carcinoma |
NCT05061550 | II | Recruiting | Monalizumab + Durvalumab | Non-Small Cell Lung Cancer |
NCT02643550 | I/II | Active, not recruiting | Monalizumab + Cetuximab + anti-PD-L1 | Head and Neck Neoplasms |
NCT04333914 | II | Completed | Monalizumab | Advanced or Metastatic Hematological or Solid Tumor |
NCT03822351 | II | Active, not recruiting | Monalizumab + Durvalumab | Unresectable Stage III Non-Small Cell Lung Cancer |
NCT03088059 | II | Recruiting | Monalizumab | Recurrrent or Metastatic Squamous Cell Carcinoma of the Head and Neck |
NCT03794544 | II | Completed | Monalizumab + Durvalumab | Resectable Early-Stage Non-Small Cell Lung Cancer |
NCT03833440 | II | Recruiting | Monalizumab + Durvalumab | Advanced Non-Small Cell Lung Cancer |
NCT05162755 | I | Recruiting | S095029 ± Sym021 (anti-PD-1) | Advanced Solid Tumor Malignancies |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cazzetta, V.; Depierreux, D.; Colucci, F.; Mikulak, J.; Mavilio, D. NKG2A Immune Checkpoint in Vδ2 T Cells: Emerging Application in Cancer Immunotherapy. Cancers 2023, 15, 1264. https://doi.org/10.3390/cancers15041264
Cazzetta V, Depierreux D, Colucci F, Mikulak J, Mavilio D. NKG2A Immune Checkpoint in Vδ2 T Cells: Emerging Application in Cancer Immunotherapy. Cancers. 2023; 15(4):1264. https://doi.org/10.3390/cancers15041264
Chicago/Turabian StyleCazzetta, Valentina, Delphine Depierreux, Francesco Colucci, Joanna Mikulak, and Domenico Mavilio. 2023. "NKG2A Immune Checkpoint in Vδ2 T Cells: Emerging Application in Cancer Immunotherapy" Cancers 15, no. 4: 1264. https://doi.org/10.3390/cancers15041264
APA StyleCazzetta, V., Depierreux, D., Colucci, F., Mikulak, J., & Mavilio, D. (2023). NKG2A Immune Checkpoint in Vδ2 T Cells: Emerging Application in Cancer Immunotherapy. Cancers, 15(4), 1264. https://doi.org/10.3390/cancers15041264