The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma
Abstract
:Simple Summary
Abstract
1. Introduction
2. Functional γδ T-Cell Subsets in Mice and Humans
3. Anti-Tumour Functions of γδ T Cells
3.1. γδ T Cells in Solid Cancers
3.2. γδ T Cells in Haematological Malignancies
3.3. γδ T Cells as Tools for Immunotherapies
4. Pro-Tumour Functions of γδ T Cells
Regulation of γδ T-Cell Cancer Immunity
5. γδ T-Cell Lymphoma/Leukaemia
5.1. Development of γδ T-Cell Lymphoma/Leukaemia
5.2. Common Genetic Aberrations
Disease Subtype | Median Survival | Disease Site | Chromosomal Lesions | Dysregulated Pathways | Genes Frequently Affected | Ref. |
---|---|---|---|---|---|---|
HSTL | 13 months | Spleen, liver | Isochromosome 7q, trisomy 8 | Epigenetic modifiers | SETD2, INO80, TET3, SMARCA2 | [214] [215] [268] |
JAK-STAT | STAT5B, STAT3 | |||||
AKT-mTOR | PIK3CD | |||||
MEITL | 7 months | Intestine | Gain of 8q24 (MYC), 1q, 7q or 9q; loss of 8p, 16q, 11q or 9p21.3 (CDKN2A/B) * | JAK-STAT | STAT5B, JAK1, JAK3 * | [222] [229] [269] [255] |
RAS-MAPK | KRAS, NRAS, BRAF * | |||||
Epigenetic modifiers | SETD2, TET2, YLPM1, CREBBP * | |||||
PCGDTL | 15–31 months | Skin | Gain of 1q, 15q or 7q; loss of 9p or 18q | RAS-MAPK | KRAS, NRAS, MAPK1 | [217] [218] [251] |
JAK-STAT | STAT5B, SOCS1, JAK3, STAT3 | |||||
Epigenetic modifiers | ARID1A, TRRAP, TET2, KMT2D | |||||
Cell cycle | CDKN2A | |||||
γδ T-LGLL | 62–114 months | Blood | Rare; gain of 7p21 and loss of Chr Y or Chr 6 reported | JAK-STAT | STAT3, STAT5B * | [233] [232] [234] [244] |
EATL | 7 months | Intestine | Gain of 1q, 7q or 9q; loss of 9p or 17p12- 13.2 (TP53) * | JAK-STAT | SOCS1, JAK1, STAT3, JAK3, STAT5B * | [222] [219] [269] |
Epigenetic modifiers | TET2, SETD2 * | |||||
Survival | DAPK3 * | |||||
γδ T-ALL | 5-year OS: 67% | Thymus, blood | Complex cytogenetic abnormalities; loss of 6q13- 23 or 12p11-13; translocations t(11;14) or t(10;11) | - | Gene fusions: SET-NUP214, CALM-AF10 | [237] [256] [257] [270] [271] |
JAK-STAT | IL7R, JAK3, STAT5B, JAK1 * | |||||
AKT-mTOR (via CK2) | - |
5.3. Current Treatment Options and Promising Therapeutic Directions
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Hayday, A.C. γδ T Cell Update: Adaptate Orchestrators of Immune Surveillance. J. Immunol. 2019, 203, 311–320. [Google Scholar] [CrossRef] [PubMed]
- Ribot, J.C.; Lopes, N.; Silva-Santos, B. γδ T cells in tissue physiology and surveillance. Nat. Rev. Immunol. 2021, 21, 221–232. [Google Scholar] [CrossRef]
- Kohlgruber, A.C.; Gal-Oz, S.T.; LaMarche, N.M.; Shimazaki, M.; Duquette, D.; Koay, H.F.; Nguyen, H.N.; Mina, A.I.; Paras, T.; Tavakkoli, A.; et al. γδ T cells producing interleukin-17A regulate adipose regulatory T cell homeostasis and thermogenesis. Nat. Immunol. 2018, 19, 464–474. [Google Scholar] [CrossRef] [PubMed]
- Nielsen, M.M.; Witherden, D.A.; Havran, W.L. γδ T cells in homeostasis and host defence of epithelial barrier tissues. Nat. Rev. Immunol. 2017, 17, 733–745. [Google Scholar] [CrossRef] [PubMed]
- Papotto, P.H.; Yilmaz, B.; Silva-Santos, B. Crosstalk between γδ T cells and the microbiota. Nat. Microbiol. 2021, 6, 1110–1117. [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]
- Hayday, A.C.; Vantourout, P. The Innate Biologies of Adaptive Antigen Receptors. Annu. Rev. Immunol. 2020, 38, 487–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hayday, A.C. [gamma][delta] cells: A right time and a right place for a conserved third way of protection. Annu Rev. Immunol. 2000, 18, 975–1026. [Google Scholar] [CrossRef]
- Baum, T.P.; Hierle, V.; Pasqual, N.; Bellahcene, F.; Chaume, D.; Lefranc, M.P.; Jouvin-Marche, E.; Marche, P.N.; Demongeot, J. IMGT/GeneInfo: T cell receptor gamma TRG and delta TRD genes in database give access to all TR potential V(D)J recombinations. BMC Bioinformatics 2006, 7, 224. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Heilig, J.S.; Tonegawa, S. Diversity of murine gamma genes and expression in fetal and adult T lymphocytes. Nature 1986, 322, 836–840. [Google Scholar] [CrossRef]
- LeFranc, M.P.; Forster, A.; Baer, R.; Stinson, M.A.; Rabbitts, T.H. Diversity and rearrangement of the human T cell rearranging gamma genes: Nine germ-line variable genes belonging to two subgroups. Cell 1986, 45, 237–246. [Google Scholar] [CrossRef]
- Vantourout, P.; Hayday, A. Six-of-the-best: Unique contributions of γδ T cells to immunology. Nat. Rev. Immunol. 2013, 13, 88–100. [Google Scholar] [CrossRef] [Green Version]
- Patel, S.; Burga, R.A.; Powell, A.B.; Chorvinsky, E.A.; Hoq, N.; McCormack, S.E.; Van Pelt, S.N.; Hanley, P.J.; Cruz, C.R.Y. Beyond CAR T Cells: Other Cell-Based Immunotherapeutic Strategies Against Cancer. Front. Oncol. 2019, 9, 196. [Google Scholar] [CrossRef] [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] [PubMed] [Green Version]
- Silva-Santos, B.; Mensurado, S.; Coffelt, S.B. γδ T cells: Pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 2019, 19, 392–404. [Google Scholar] [CrossRef] [Green Version]
- Godfrey, D.I.; Le Nours, J.; Andrews, D.M.; Uldrich, A.P.; Rossjohn, J. Unconventional T Cell Targets for Cancer Immunotherapy. Immunity 2018, 48, 453–473. [Google Scholar] [CrossRef] [Green Version]
- Castella, B.; Kopecka, J.; Sciancalepore, P.; Mandili, G.; Foglietta, M.; Mitro, N.; Caruso, D.; Novelli, F.; Riganti, C.; Massaia, M. The ATP-binding cassette transporter A1 regulates phosphoantigen release and Vγ9Vδ2 T cell activation by dendritic cells. Nat. Commun. 2017, 8, 15663. [Google Scholar] [CrossRef] [PubMed]
- Zocchi, M.R.; Costa, D.; Venè, R.; Tosetti, F.; Ferrari, N.; Minghelli, S.; Benelli, R.; Scabini, S.; Romairone, E.; Catellani, S.; et al. Zoledronate can induce colorectal cancer microenvironment expressing BTN3A1 to stimulate effector γδ T cells with antitumor activity. Oncoimmunology 2017, 6, e1278099. [Google Scholar] [CrossRef]
- Blazquez, J.L.; Benyamine, A.; Pasero, C.; Olive, D. New Insights into the Regulation of γδ T Cells by BTN3A and Other BTN/BTNL in Tumor Immunity. Front. Immunol. 2018, 9, 1601. [Google Scholar] [CrossRef] [Green Version]
- Payne, K.K.; Mine, J.A.; Biswas, S.; Chaurio, R.A.; Perales-Puchalt, A.; Anadon, C.M.; Costich, T.L.; Harro, C.M.; Walrath, J.; Ming, Q.; et al. BTN3A1 governs antitumor responses by coordinating αβ and γδ T cells. Science 2020, 369, 942–949. [Google Scholar] [CrossRef]
- Sawaisorn, P.; Tangchaikeeree, T.; Chan-On, W.; Leepiyasakulchai, C.; Udomsangpetch, R.; Hongeng, S.; Jangpatarapongsa, K. Antigen-Presenting Cell Characteristics of Human γδ T Lymphocytes in Chronic Myeloid Leukemia. Immunol. Investig. 2019, 48, 11–26. [Google Scholar] [CrossRef]
- Holmen Olofsson, G.; Idorn, M.; Carnaz Simões, A.M.; Aehnlich, P.; Skadborg, S.K.; Noessner, E.; Debets, R.; Moser, B.; Met, Ö.; Thor Straten, P. Vγ9Vδ2 T Cells Concurrently Kill Cancer Cells and Cross-Present Tumor Antigens. Front. Immunol. 2021, 12, 645131. [Google Scholar] [CrossRef]
- Rampoldi, F.; Ullrich, L.; Prinz, I. Revisiting the Interaction of γδ T-Cells and B-Cells. Cells 2020, 9, 743. [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]
- 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] [PubMed]
- Tawfik, D.; Groth, C.; Gundlach, J.P.; Peipp, M.; Kabelitz, D.; Becker, T.; Oberg, H.H.; Trauzold, A.; Wesch, D. TRAIL-Receptor 4 Modulates γδ T Cell-Cytotoxicity Toward Cancer Cells. Front. Immunol. 2019, 10, 2044. [Google Scholar] [CrossRef]
- Deseke, M.; Prinz, I. Ligand recognition by the γδ TCR and discrimination between homeostasis and stress conditions. Cell Mol. Immunol. 2020, 17, 914–924. [Google Scholar] [CrossRef]
- Nussbaumer, O.; Thurnher, M. Functional Phenotypes of Human Vγ9Vδ2 T Cells in Lymphoid Stress Surveillance. Cells 2020, 9, 772. [Google Scholar] [CrossRef] [Green Version]
- Maggi, L.; Santarlasci, V.; Capone, M.; Peired, A.; Frosali, F.; Crome, S.Q.; Querci, V.; Fambrini, M.; Liotta, F.; Levings, M.K.; et al. CD161 is a marker of all human IL-17-producing T-cell subsets and is induced by RORC. Eur J. Immunol. 2010, 40, 2174–2181. [Google Scholar] [CrossRef]
- Hunter, S.; Willcox, C.R.; Davey, M.S.; Kasatskaya, S.A.; Jeffery, H.C.; Chudakov, D.M.; Oo, Y.H.; Willcox, B.E. Human liver infiltrating γδ T cells are composed of clonally expanded circulating and tissue-resident populations. J. Hepatol. 2018, 69, 654–665. [Google Scholar] [CrossRef]
- Dieli, F.; Poccia, F.; Lipp, M.; Sireci, G.; Caccamo, N.; Di Sano, C.; Salerno, A. Differentiation of effector/memory Vdelta2 T cells and migratory routes in lymph nodes or inflammatory sites. J. Exp. Med. 2003, 198, 391–397. [Google Scholar] [CrossRef] [Green Version]
- Kadekar, D.; Agerholm, R.; Rizk, J.; Neubauer, H.A.; Suske, T.; Maurer, B.; Viñals, M.T.; Comelli, E.M.; Taibi, A.; Moriggl, R.; et al. The neonatal microenvironment programs innate γδ T cells through the transcription factor STAT5. J. Clin. Investig. 2020, 130, 2496–2508. [Google Scholar] [CrossRef]
- Lopes, N.; McIntyre, C.; Martin, S.; Raverdeau, M.; Sumaria, N.; Kohlgruber, A.C.; Fiala, G.J.; Agudelo, L.Z.; Dyck, L.; Kane, H.; et al. Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments. Nat. Immunol. 2021, 22, 179–192. [Google Scholar] [CrossRef]
- Ribeiro, S.T.; Ribot, J.C.; Silva-Santos, B. Five Layers of Receptor Signaling in γδ T-Cell Differentiation and Activation. Front. Immunol. 2015, 6, 15. [Google Scholar] [CrossRef] [PubMed] [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]
- Chen, X.; Shang, W.; Xu, R.; Wu, M.; Zhang, X.; Huang, P.; Wang, F.; Pan, S. Distribution and functions of γδ T cells infiltrated in the ovarian cancer microenvironment. J. Transl. Med. 2019, 17, 144. [Google Scholar] [CrossRef] [Green Version]
- Lo Presti, E.; Pizzolato, G.; Corsale, A.M.; Caccamo, N.; Sireci, G.; Dieli, F.; Meraviglia, S. γδ T Cells and Tumor Microenvironment: From Immunosurveillance to Tumor Evasion. Front. Immunol. 2018, 9, 1395. [Google Scholar] [CrossRef]
- Wu, P.; Wu, D.; Ni, C.; Ye, J.; Chen, W.; Hu, G.; Wang, Z.; Wang, C.; Zhang, Z.; Xia, W.; et al. γδT17 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]
- Sacchi, A.; Tumino, N.; Sabatini, A.; Cimini, E.; Casetti, R.; Bordoni, V.; Grassi, G.; Agrati, C. Myeloid-Derived Suppressor Cells Specifically Suppress IFN-γ Production and Antitumor Cytotoxic Activity of Vδ2 T Cells. Front. Immunol. 2018, 9, 1271. [Google Scholar] [CrossRef] [Green Version]
- Tomogane, M.; Sano, Y.; Shimizu, D.; Shimizu, T.; Miyashita, M.; Toda, Y.; Hosogi, S.; Tanaka, Y.; Kimura, S.; Ashihara, E. Human Vγ9Vδ2 T cells exert anti-tumor activity independently of PD-L1 expression in tumor cells. Biochem. Biophys. Res. Commun. 2021, 573, 132–139. [Google Scholar] [CrossRef]
- Sureshbabu, S.K.; Chaukar, D.; Chiplunkar, S.V. Hypoxia regulates the differentiation and anti-tumor effector functions of γδT cells in oral cancer. Clin. Exp. Immunol. 2020, 201, 40–57. [Google Scholar] [CrossRef]
- Agerholm, R.; Bekiaris, V. Evolved to protect, designed to destroy: IL-17-producing γδ T cells in infection, inflammation, and cancer. Eur. J. Immunol. 2021. [Google Scholar] [CrossRef]
- Haas, J.D.; Ravens, S.; Düber, S.; Sandrock, I.; Oberdörfer, L.; Kashani, E.; Chennupati, V.; Föhse, L.; Naumann, R.; Weiss, S.; et al. Development of interleukin-17-producing γδ T cells is restricted to a functional embryonic wave. Immunity 2012, 37, 48–59. [Google Scholar] [CrossRef] [Green Version]
- Buus, T.B.; Ødum, N.; Geisler, C.; Lauritsen, J.P.H. Three distinct developmental pathways for adaptive and two IFN-γ-producing γδ T subsets in adult thymus. Nat. Commun. 2017, 8, 1911. [Google Scholar] [CrossRef]
- Sandrock, I.; Reinhardt, A.; Ravens, S.; Binz, C.; Wilharm, A.; Martins, J.; Oberdörfer, L.; Tan, L.; Lienenklaus, S.; Zhang, B.; et al. Genetic models reveal origin, persistence and non-redundant functions of IL-17-producing γδ T cells. J. Exp. Med. 2018, 215, 3006–3018. [Google Scholar] [CrossRef] [Green Version]
- Muñoz-Ruiz, M.; Sumaria, N.; Pennington, D.J.; Silva-Santos, B. Thymic Determinants of γδ T Cell Differentiation. Trends Immunol. 2017, 38, 336–344. [Google Scholar] [CrossRef]
- Yin, Z.; Chen, C.; Szabo, S.J.; Glimcher, L.H.; Ray, A.; Craft, J. T-Bet expression and failure of GATA-3 cross-regulation lead to default production of IFN-gamma by gammadelta T cells. J. Immunol. 2002, 168, 1566–1571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sumaria, N.; Grandjean, C.L.; Silva-Santos, B.; Pennington, D.J. Strong TCRγδ Signaling Prohibits Thymic Development of IL-17A-Secreting γδ T Cells. Cell Rep. 2017, 19, 2469–2476. [Google Scholar] [CrossRef] [Green Version]
- Zarin, P.; In, T.S.; Chen, E.L.; Singh, J.; Wong, G.W.; Mohtashami, M.; Wiest, D.L.; Anderson, M.K.; Zúñiga-Pflücker, J.C. Integration of T-cell receptor, Notch and cytokine signals programs mouse γδ T-cell effector differentiation. Immunol. Cell Biol. 2018, 96, 994–1007. [Google Scholar] [CrossRef] [Green Version]
- Deusch, K.; Lüling, F.; Reich, K.; Classen, M.; Wagner, H.; Pfeffer, K. A major fraction of human intraepithelial lymphocytes simultaneously expresses the gamma/delta T cell receptor, the CD8 accessory molecule and preferentially uses the V delta 1 gene segment. Eur J. Immunol. 1991, 21, 1053–1059. [Google Scholar] [CrossRef] [PubMed]
- Peng, M.Y.; Wang, Z.H.; Yao, C.Y.; Jiang, L.N.; Jin, Q.L.; Wang, J.; Li, B.Q. Interleukin 17-producing gamma delta T cells increased in patients with active pulmonary tuberculosis. Cell Mol. Immunol. 2008, 5, 203–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fenoglio, D.; Poggi, A.; Catellani, S.; Battaglia, F.; Ferrera, A.; Setti, M.; Murdaca, G.; Zocchi, M.R. Vdelta1 T lymphocytes producing IFN-gamma and IL-17 are expanded in HIV-1-infected patients and respond to Candida albicans. Blood 2009, 113, 6611–6618. [Google Scholar] [CrossRef] [PubMed]
- 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. Immunol. 2010, 184, 7268–7280. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Ryan, P.L.; Sumaria, N.; Holland, C.J.; Bradford, C.M.; Izotova, N.; Grandjean, C.L.; Jawad, A.S.; Bergmeier, L.A.; Pennington, D.J. Heterogeneous yet stable Vδ2(+) T-cell profiles define distinct cytotoxic effector potentials in healthy human individuals. Proc. Natl. Acad. Sci. USA 2016, 113, 14378–14383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, L.; Fichtner, A.S.; Bruni, E.; Odak, I.; Sandrock, I.; Bubke, A.; Borchers, A.; Schultze-Florey, C.; Koenecke, C.; Förster, R.; et al. A fetal wave of human type 3 effector γδ cells with restricted TCR diversity persists into adulthood. Sci. Immunol. 2021, 6, eabf0125. [Google Scholar] [CrossRef] [PubMed]
- Giri, S.; Lal, G. Differentiation and functional plasticity of gamma-delta (γδ) T cells under homeostatic and disease conditions. Mol. Immunol. 2021, 136, 138–149. [Google Scholar] [CrossRef]
- McKenzie, D.R.; Kara, E.E.; Bastow, C.R.; Tyllis, T.S.; Fenix, K.A.; Gregor, C.E.; Wilson, J.J.; Babb, R.; Paton, J.C.; Kallies, A.; et al. IL-17-producing γδ T cells switch migratory patterns between resting and activated states. Nat. Commun. 2017, 8, 15632. [Google Scholar] [CrossRef]
- Casetti, R.; Martino, A. The plasticity of gamma delta T cells: Innate immunity, antigen presentation and new immunotherapy. Cell Mol. Immunol. 2008, 5, 161–170. [Google Scholar] [CrossRef]
- 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]
- 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] [PubMed] [Green Version]
- Casetti, R.; Agrati, C.; Wallace, M.; Sacchi, A.; Martini, F.; Martino, A.; Rinaldi, A.; Malkovsky, M. Cutting edge: TGF-beta1 and IL-15 Induce FOXP3+ gammadelta regulatory T cells in the presence of antigen stimulation. J. Immunol. 2009, 183, 3574–3577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Park, J.H.; Kim, H.J.; Kim, C.W.; Kim, H.C.; Jung, Y.; Lee, H.S.; Lee, Y.; Ju, Y.S.; Oh, J.E.; Park, S.H.; et al. Tumor hypoxia represses γδ T cell-mediated antitumor immunity against brain tumors. Nat. Immunol. 2021, 22, 336–346. [Google Scholar] [CrossRef]
- Li, L.; Cao, B.; Liang, X.; Lu, S.; Luo, H.; Wang, Z.; Wang, S.; Jiang, J.; Lang, J.; Zhu, G. Microenvironmental oxygen pressure orchestrates an anti- and pro-tumoral γδ T cell equilibrium via tumor-derived exosomes. Oncogene 2019, 38, 2830–2843. [Google Scholar] [CrossRef] [PubMed]
- Siegers, G.M.; Dutta, I.; Lai, R.; Postovit, L.M. Functional Plasticity of Gamma Delta T Cells and Breast Tumor Targets in Hypoxia. Front. Immunol. 2018, 9, 1367. [Google Scholar] [CrossRef] [PubMed]
- Douguet, L.; Bod, L.; Lengagne, R.; Labarthe, L.; Kato, M.; Avril, M.F.; Prévost-Blondel, A. Nitric oxide synthase 2 is involved in the pro-tumorigenic potential of γδ17 T cells in melanoma. Oncoimmunology 2016, 5, e1208878. [Google Scholar] [CrossRef] [Green Version]
- Zhao, N.; Dang, H.; Ma, L.; Martin, S.P.; Forgues, M.; Ylaya, K.; Hewitt, S.M.; Wang, X.W. Intratumoral γδ 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]
- Pietschmann, K.; Beetz, S.; Welte, S.; Martens, I.; Gruen, J.; Oberg, H.H.; Wesch, D.; Kabelitz, D. Toll-like receptor expression and function in subsets of human gammadelta T lymphocytes. Scand. J. Immunol. 2009, 70, 245–255. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- 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]
- Sperling, A.I.; Linsley, P.S.; Barrett, T.A.; Bluestone, J.A. CD28-mediated costimulation is necessary for the activation of T cell receptor-gamma delta + T lymphocytes. J. Immunol. 1993, 151, 6043–6050. [Google Scholar] [PubMed]
- Stojanovic, A.; Correia, M.P.; Cerwenka, A. The NKG2D/NKG2DL Axis in the Crosstalk Between Lymphoid and Myeloid Cells in Health and Disease. Front. Immunol. 2018, 9, 827. [Google Scholar] [CrossRef] [PubMed] [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]
- Das, H.; Groh, V.; Kuijl, C.; Sugita, M.; Morita, C.T.; Spies, T.; Bukowski, J.F. MICA engagement by human Vgamma2Vdelta2 T cells enhances their antigen-dependent effector function. Immunity 2001, 15, 83–93. [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, 992–1001. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- von Lilienfeld-Toal, M.; Nattermann, J.; Feldmann, G.; Sievers, E.; Frank, S.; Strehl, J.; Schmidt-Wolf, I.G. Activated gammadelta T cells express the natural cytotoxicity receptor natural killer p 44 and show cytotoxic activity against myeloma cells. Clin. Exp. Immunol. 2006, 144, 528–533. [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 Vδ1 T cell subpopulation exhibits high antitumor activity against colorectal cancer. JCI Insight 2019, 4, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Constant, P.; Davodeau, F.; Peyrat, M.A.; Poquet, Y.; Puzo, G.; Bonneville, M.; Fournié, J.J. Stimulation of human gamma delta T cells by nonpeptidic mycobacterial ligands. Science 1994, 264, 267–270. [Google Scholar] [CrossRef]
- Thompson, K.; Roelofs, A.J.; Jauhiainen, M.; Mönkkönen, H.; Mönkkönen, J.; Rogers, M.J. Activation of γδ T cells by bisphosphonates. Adv. Exp. Med. Biol. 2010, 658, 11–20. [Google Scholar] [CrossRef]
- Bukowski, J.F.; Morita, C.T.; Brenner, M.B. Human gamma delta T cells recognize alkylamines derived from microbes, edible plants, and tea: Implications for innate immunity. Immunity 1999, 11, 57–65. [Google Scholar] [CrossRef] [Green Version]
- Thompson, K.; Rojas-Navea, J.; Rogers, M.J. Alkylamines cause Vgamma9Vdelta2 T-cell activation and proliferation by inhibiting the mevalonate pathway. Blood 2006, 107, 651–654. [Google Scholar] [CrossRef] [PubMed]
- Gober, H.J.; Kistowska, M.; Angman, L.; Jenö, 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]
- 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] [PubMed] [Green Version]
- Laad, A.D.; Thomas, M.L.; Fakih, A.R.; Chiplunkar, S.V. Human gamma delta T cells recognize heat shock protein-60 on oral tumor cells. Int. J. Cancer 1999, 80, 709–714. [Google Scholar] [CrossRef]
- Eagle, R.A.; Traherne, J.A.; Hair, J.R.; Jafferji, I.; Trowsdale, J. ULBP6/RAET1L is an additional human NKG2D ligand. Eur. J. Immunol. 2009, 39, 3207–3216. [Google Scholar] [CrossRef]
- Eagle, R.A.; Trowsdale, J. Promiscuity and the single receptor: NKG2D. Nat. Rev. Immunol. 2007, 7, 737–744. [Google Scholar] [CrossRef] [PubMed]
- Fu, Y.X.; Vollmer, M.; Kalataradi, H.; Heyborne, K.; Reardon, C.; Miles, C.; O’Brien, R.; Born, W. Structural requirements for peptides that stimulate a subset of gamma delta T cells. J. Immunol. 1994, 152, 1578–1588. [Google Scholar]
- Uldrich, A.P.; Le Nours, J.; Pellicci, D.G.; Gherardin, N.A.; McPherson, K.G.; Lim, R.T.; Patel, O.; Beddoe, T.; Gras, S.; Rossjohn, J.; et al. CD1d-lipid antigen recognition by the γδ TCR. Nat. Immunol. 2013, 14, 1137–1145. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- Chauvin, C.; Joalland, N.; Perroteau, J.; Jarry, U.; Lafrance, L.; Willem, C.; Retière, C.; Oliver, L.; Gratas, C.; Gautreau-Rolland, L.; et al. NKG2D Controls Natural Reactivity of Vγ9Vδ2 T Lymphocytes against Mesenchymal Glioblastoma Cells. Clin. Cancer Res. 2019, 25, 7218–7228. [Google Scholar] [CrossRef] [Green Version]
- Nedellec, S.; Sabourin, C.; Bonneville, M.; Scotet, E. NKG2D costimulates human V gamma 9V delta 2 T cell antitumor cytotoxicity through protein kinase C theta-dependent modulation of early TCR-induced calcium and transduction signals. J. Immunol. 2010, 185, 55–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Neill, K.; Pastar, I.; Tomic-Canic, M.; Strbo, N. Perforins Expression by Cutaneous Gamma Delta T Cells. Front. Immunol. 2020, 11, 1839. [Google Scholar] [CrossRef] [PubMed]
- Uchida, R.; Ashihara, E.; Sato, K.; Kimura, S.; Kuroda, J.; Takeuchi, M.; Kawata, E.; Taniguchi, K.; Okamoto, M.; Shimura, K.; et al. Gamma delta T cells kill myeloma cells by sensing mevalonate metabolites and ICAM-1 molecules on cell surface. Biochem. Biophys. Res. Commun. 2007, 354, 613–618. [Google Scholar] [CrossRef] [PubMed]
- Harly, C.; Guillaume, Y.; Nedellec, S.; Peigné, C.M.; Mönkkönen, H.; Mönkkönen, J.; Li, J.; Kuball, J.; Adams, E.J.; Netzer, S.; et al. Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human γδ T-cell subset. Blood 2012, 120, 2269–2279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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, eaay5516. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Nussbaumer, O.; Koslowski, M. The emerging role of γδ T cells in cancer immunotherapy. Immuno-Oncol. Technol. 2019, 1, 3–10. [Google Scholar] [CrossRef] [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]
- Godfrey, D.I.; Uldrich, A.P.; McCluskey, J.; Rossjohn, J.; Moody, D.B. The burgeoning family of unconventional T cells. Nat. Immunol. 2015, 16, 1114–1123. [Google Scholar] [CrossRef] [PubMed]
- Todaro, M.; D’Asaro, M.; Caccamo, N.; Iovino, F.; Francipane, M.G.; Meraviglia, S.; Orlando, V.; La Mendola, C.; Gulotta, G.; Salerno, A.; et al. Efficient killing of human colon cancer stem cells by gammadelta T lymphocytes. J. Immunol. 2009, 182, 7287–7296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, Z.; Liu, Y.; Zheng, J.; Liu, M.; Lv, A.; Gao, Y.; Hu, H.; Lam, K.T.; Chan, G.C.; Yang, Y.; et al. Targeted activation of human Vγ9Vδ2-T cells controls epstein-barr virus-induced B cell lymphoproliferative disease. Cancer Cell 2014, 26, 565–576. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Braza, M.S.; Klein, B.; Fiol, G.; Rossi, J.F. γδ T-cell killing of primary follicular lymphoma cells is dramatically potentiated by GA101, a type II glycoengineered anti-CD20 monoclonal antibody. Haematologica 2011, 96, 400–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mizoguchi, A.; Mizoguchi, E.; de Jong, Y.P.; Takedatsu, H.; Preffer, F.I.; Terhorst, C.; Bhan, A.K. Role of the CD5 molecule on TCR gammadelta T cell-mediated immune functions: Development of germinal centers and chronic intestinal inflammation. Int. Immunol. 2003, 15, 97–108. [Google Scholar] [CrossRef]
- Huang, Y.; Getahun, A.; Heiser, R.A.; Detanico, T.O.; Aviszus, K.; Kirchenbaum, G.A.; Casper, T.L.; Huang, C.; Aydintug, M.K.; Carding, S.R.; et al. γδ T Cells Shape Preimmune Peripheral B Cell Populations. J. Immunol. 2016, 196, 217–231. [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. Immunol. 2006, 177, 5290–5295. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed]
- Petrasca, A.; Melo, A.M.; Breen, E.P.; Doherty, D.G. Human Vδ3(+) γδ T cells induce maturation and IgM secretion by B cells. Immunol. Lett. 2018, 196, 126–134. [Google Scholar] [CrossRef]
- Brandes, M.; Willimann, K.; Moser, B. Professional antigen-presentation function by human gammadelta T Cells. Science 2005, 309, 264–268. [Google Scholar] [CrossRef] [PubMed]
- Gruenbacher, G.; Thurnher, M. Mevalonate metabolism governs cancer immune surveillance. OncoImmunology 2017, 6, e1342917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muto, M.; Baghdadi, M.; Maekawa, R.; Wada, H.; Seino, K. 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] [PubMed] [Green Version]
- Khan, M.W.A.; Curbishley, S.M.; Chen, H.-C.; Thomas, A.D.; Pircher, H.; Mavilio, D.; Steven, N.M.; Eberl, M.; Moser, B. Expanded Human Blood-Derived γδT Cells Display Potent Antigen-Presentation Functions. Front. Immunol. 2014, 5, 344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jouen-Beades, F.; Paris, E.; Dieulois, C.; Lemeland, J.F.; Barre-Dezelus, V.; Marret, S.; Humbert, G.; Leroy, J.; Tron, F. In vivo and in vitro activation and expansion of gammadelta T cells during Listeria monocytogenes infection in humans. Infect. Immunol. 1997, 65, 4267–4272. [Google Scholar] [CrossRef] [Green Version]
- Devilder, M.C.; Maillet, S.; Bouyge-Moreau, I.; Donnadieu, E.; Bonneville, M.; Scotet, E. Potentiation of antigen-stimulated V gamma 9V delta 2 T cell cytokine production by immature dendritic cells (DC) and reciprocal effect on DC maturation. J. Immunol. 2006, 176, 1386–1393. [Google Scholar] [CrossRef] [Green Version]
- Münz, C.; Steinman, R.M.; Fujii, S. Dendritic cell maturation by innate lymphocytes: Coordinated stimulation of innate and adaptive immunity. J. Exp. Med. 2005, 202, 203–207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leslie, D.S.; Vincent, M.S.; Spada, F.M.; Das, H.; Sugita, M.; Morita, C.T.; Brenner, M.B. CD1-mediated gamma/delta T cell maturation of dendritic cells. J. Exp. Med. 2002, 196, 1575–1584. [Google Scholar] [CrossRef] [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. Immunol. 2005, 174, 252–260. [Google Scholar] [CrossRef]
- Girard, P.; Ponsard, B.; Charles, J.; Chaperot, L.; Aspord, C. Potent Bidirectional Cross-Talk Between Plasmacytoid Dendritic Cells and γδT Cells Through BTN3A, Type I/II IFNs and Immune Checkpoints. Front. Immunol. 2020, 11, 11. [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. Immunol. 2013, 191, 30–34. [Google Scholar] [CrossRef]
- Wang, J.; Lin, C.; Li, H.; Li, R.; Wu, Y.; Liu, H.; Zhang, H.; He, H.; Zhang, W.; Xu, J. Tumor-infiltrating γδT cells predict prognosis and adjuvant chemotherapeutic benefit in patients with gastric cancer. Oncoimmunology 2017, 6, e1353858. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Tang, F.; Li, Z.; Cui, L.; He, W. Critical role of γ4 chain in the expression of functional Vγ4Vδ1 T cell receptor of gastric tumour-infiltrating γδT lymphocytes. Scand. J. Immunol. 2012, 75, 102–108. [Google Scholar] [CrossRef]
- Foord, E.; Arruda, L.C.M.; Gaballa, A.; Klynning, C.; Uhlin, M. Characterization of ascites- and tumor-infiltrating γδ T cells reveals distinct repertoires and a beneficial role in ovarian cancer. Sci. Transl. Med. 2021, 13, 13. [Google Scholar] [CrossRef] [PubMed]
- Andreu-Ballester, J.C.; Galindo-Regal, L.; Hidalgo-Coloma, J.; Cuéllar, C.; García-Ballesteros, C.; Hurtado, C.; Uribe, N.; Del Carmen Martín, M.; Jiménez, A.I.; López-Chuliá, F.; et al. Differences in circulating γδ T cells in patients with primary colon cancer and relation with prognostic factors. PLoS ONE 2020, 15, e0243545. [Google Scholar] [CrossRef]
- Narayanan, S.; Kawaguchi, T.; Yan, L.; Peng, X.; Qi, Q.; Takabe, K. Cytolytic Activity Score to Assess Anticancer Immunity in Colorectal Cancer. Ann. Surg. Oncol. 2018, 25, 2323–2331. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Zhang, E.; Long, J.; Hu, Z.; Peng, J.; Liu, L.; Tang, F.; Li, L.; Ouyang, Y.; Zeng, Z. Immune infiltration in renal cell carcinoma. Cancer Sci. 2019, 110, 1564–1572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.W.; Park, C.; Joung, J.G.; Kang, M.; Chung, Y.S.; Oh, W.J.; Yeom, S.Y.; Park, W.Y.; Kim, T.J.; Seo, S.I. Renal Cell Carcinoma-Infiltrating CD3(low) Vγ9Vδ1 T Cells Represent Potentially Novel Anti-Tumor Immune Players. Curr. Issues Mol. Biol. 2021, 43, 226–239. [Google Scholar] [CrossRef] [PubMed]
- Bryant, N.L.; Suarez-Cuervo, C.; Gillespie, G.Y.; Markert, J.M.; Nabors, L.B.; Meleth, S.; Lopez, R.D.; Lamb, L.S., Jr. Characterization and immunotherapeutic potential of gammadelta T-cells in patients with glioblastoma. Neuro. Oncol. 2009, 11, 357–367. [Google Scholar] [CrossRef] [PubMed]
- Boissière-Michot, F.; Chabab, G.; Mollevi, C.; Guiu, S.; Lopez-Crapez, E.; Ramos, J.; Bonnefoy, N.; Lafont, V.; Jacot, W. Clinicopathological Correlates of γδ T Cell Infiltration in Triple-Negative Breast Cancer. Cancers 2021, 13, 765. [Google Scholar] [CrossRef] [PubMed]
- Hidalgo, J.V.; Bronsert, P.; Orlowska-Volk, M.; Díaz, L.B.; Stickeler, E.; Werner, M.; Schmitt-Graeff, A.; Kayser, G.; Malkovsky, M.; Fisch, P. Histological Analysis of γδ T Lymphocytes Infiltrating Human Triple-Negative Breast Carcinomas. Front. Immunol. 2014, 5, 632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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, eaax9364. [Google Scholar] [CrossRef] [PubMed]
- Lo Presti, E.; Mocciaro, F.; Mitri, R.D.; Corsale, A.M.; Di Simone, M.; Vieni, S.; Scibetta, N.; Unti, E.; Dieli, F.; Meraviglia, S. Analysis of colon-infiltrating γδ T cells in chronic inflammatory bowel disease and in colitis-associated cancer. J. Leukoc. Biol. 2020, 108, 749–760. [Google Scholar] [CrossRef] [PubMed]
- Chabab, G.; Barjon, C.; Abdellaoui, N.; Salvador-Prince, L.; Dejou, C.; Michaud, H.A.; Boissière-Michot, F.; Lopez-Crapez, E.; Jacot, W.; Pourquier, D.; et al. Identification of a regulatory Vδ1 gamma delta T cell subpopulation expressing CD73 in human breast cancer. J. Leukoc. Biol. 2020, 107, 1057–1067. [Google Scholar] [CrossRef] [PubMed]
- Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef]
- Lopes, N.; Silva-Santos, B. Functional and metabolic dichotomy of murine γδ T cell subsets in cancer immunity. Eur. J. Immunol. 2021, 51, 17–26. [Google Scholar] [CrossRef]
- 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 γδ T Cells Producing either IL17 or IFNγ Depending on the Tumor Stage. Cancer Immunol. Res. 2017, 5, 397–407. [Google Scholar] [CrossRef] [Green Version]
- 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.; 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]
- Castella, B.; Vitale, C.; Coscia, M.; Massaia, M. Vγ9Vδ2 T cell-based immunotherapy in hematological malignancies: From bench to bedside. Cell Mol. Life Sci. 2011, 68, 2419–2432. [Google Scholar] [CrossRef]
- Dolstra, H.; Fredrix, H.; van der Meer, A.; de Witte, T.; Figdor, C.; van de Wiel-van Kemenade, E. TCR gamma delta cytotoxic T lymphocytes expressing the killer cell-inhibitory receptor p58.2 (CD158b) selectively lyse acute myeloid leukemia cells. Bone Marrow Transplant. 2001, 27, 1087–1093. [Google Scholar] [CrossRef] [Green Version]
- 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. Immunol. 2012, 188, 4701–4708. [Google Scholar] [CrossRef]
- Gomes, A.Q.; Correia, D.V.; Grosso, A.R.; Lança, T.; Ferreira, C.; Lacerda, J.F.; Barata, J.T.; Silva, M.G.; Silva-Santos, B. Identification of a panel of ten cell surface protein antigens associated with immunotargeting of leukemias and lymphomas by peripheral blood gammadelta T cells. Haematologica 2010, 95, 1397–1404. [Google Scholar] [CrossRef] [PubMed]
- 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]
- 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]
- 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]
- 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] [PubMed] [Green Version]
- Sicard, H.; Al Saati, T.; Delsol, G.; Fournié, J.J. Synthetic phosphoantigens enhance human Vgamma9Vdelta2 T lymphocytes killing of non-Hodgkin’s B lymphoma. Mol. Med. 2001, 7, 711–722. [Google Scholar] [CrossRef] [Green Version]
- Street, S.E.; Hayakawa, Y.; Zhan, Y.; Lew, A.M.; MacGregor, D.; Jamieson, A.M.; Diefenbach, A.; Yagita, H.; Godfrey, D.I.; Smyth, M.J. Innate immune surveillance of spontaneous B cell lymphomas by natural killer cells and gammadelta T cells. J. Exp. Med. 2004, 199, 879–884. [Google Scholar] [CrossRef]
- Zumwalde, N.A.; Sharma, A.; Xu, X.; Ma, S.; Schneider, C.L.; Romero-Masters, J.C.; Hudson, A.W.; Gendron-Fitzpatrick, A.; Kenney, S.C.; Gumperz, J.E. Adoptively transferred Vγ9Vδ2 T cells show potent antitumor effects in a preclinical B cell lymphomagenesis model. JCI Insight 2017, 2, 13. [Google Scholar] [CrossRef] [Green Version]
- Gundermann, S.; Klinker, E.; Kimmel, B.; Flierl, U.; Wilhelm, M.; Einsele, H.; Kunzmann, V. A comprehensive analysis of primary acute myeloid leukemia identifies biomarkers predicting susceptibility to human allogeneic Vγ9Vδ2 T cells. J. Immunother. 2014, 37, 321–330. [Google Scholar] [CrossRef]
- Catellani, S.; Poggi, A.; Bruzzone, A.; Dadati, P.; Ravetti, J.L.; Gobbi, M.; Zocchi, M.R. Expansion of Vdelta1 T lymphocytes producing IL-4 in low-grade non-Hodgkin lymphomas expressing UL-16-binding proteins. Blood 2007, 109, 2078–2085. [Google Scholar] [CrossRef] [PubMed]
- Federmann, B.; Bornhauser, M.; Meisner, C.; Kordelas, L.; Beelen, D.W.; Stuhler, G.; Stelljes, M.; Schwerdtfeger, R.; Christopeit, M.; Behre, G.; et al. Haploidentical allogeneic hematopoietic cell transplantation in adults using CD3/CD19 depletion and reduced intensity conditioning: A phase II study. Haematologica 2012, 97, 1523–1531. [Google Scholar] [CrossRef] [Green Version]
- Barfield, R.C.; Otto, M.; Houston, J.; Holladay, M.; Geiger, T.; Martin, J.; Leimig, T.; Gordon, P.; Chen, X.; Handgretinger, R. A one-step large-scale method for T- and B-cell depletion of mobilized PBSC for allogeneic transplantation. Cytotherapy 2004, 6, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Li Pira, G.; Malaspina, D.; Girolami, E.; Biagini, S.; Cicchetti, E.; Conflitti, G.; Broglia, M.; Ceccarelli, S.; Lazzaro, S.; Pagliara, D.; et al. Selective Depletion of αβ T Cells and B Cells for Human Leukocyte Antigen-Haploidentical Hematopoietic Stem Cell Transplantation. A Three-Year Follow-Up of Procedure Efficiency. Biol Blood Marrow Transplant. 2016, 22, 2056–2064. [Google Scholar] [CrossRef] [Green Version]
- Locatelli, F.; Merli, P.; Pagliara, D.; Li Pira, G.; Falco, M.; Pende, D.; Rondelli, R.; Lucarelli, B.; Brescia, L.P.; Masetti, R.; et al. Outcome of children with acute leukemia given HLA-haploidentical HSCT after αβ T-cell and B-cell depletion. Blood 2017, 130, 677–685. [Google Scholar] [CrossRef]
- Airoldi, I.; Bertaina, A.; Prigione, I.; Zorzoli, A.; Pagliara, D.; Cocco, C.; Meazza, R.; Loiacono, F.; Lucarelli, B.; Bernardo, M.E.; et al. γδ T-cell reconstitution after HLA-haploidentical hematopoietic transplantation depleted of TCR-αβ+/CD19+ lymphocytes. Blood 2015, 125, 2349–2358. [Google Scholar] [CrossRef]
- Godder, K.T.; Henslee-Downey, P.J.; Mehta, J.; Park, B.S.; Chiang, K.Y.; Abhyankar, S.; Lamb, L.S. Long term disease-free survival in acute leukemia patients recovering with increased gammadelta T cells after partially mismatched related donor bone marrow transplantation. Bone Marrow Transplant. 2007, 39, 751–757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, C.M.; McCarthy, P.L.; Wallace, P.K.; Zhang, Y.; Fora, A.; Mellors, P.; Tario, J.D.; McCarthy, B.L.S.; Chen, G.L.; Holstein, S.A.; et al. Immune signatures associated with improved progression-free and overall survival for myeloma patients treated with AHSCT. Blood Adv. 2017, 1, 1056–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klyuchnikov, E.; Badbaran, A.; Massoud, R.; Fritsche-Friedland, U.; Janson, D.; Ayuk, F.; Wolschke, C.; Bacher, U.; Kröger, N. Enhanced Immune Reconstitution of γδ T Cells after Allogeneic Stem Cell Transplantation Overcomes the Negative Impact of Pretransplantation Minimal Residual Disease-Positive Status in Patients with Acute Myelogenous Leukemia. Transplant. Cell Ther. 2021, 27, 841–850. [Google Scholar] [CrossRef]
- Lança, T.; Correia, D.V.; Moita, C.F.; Raquel, H.; Neves-Costa, A.; Ferreira, C.; Ramalho, J.S.; Barata, J.T.; Moita, L.F.; Gomes, A.Q.; et al. The MHC class Ib protein ULBP1 is a nonredundant determinant of leukemia/lymphoma susceptibility to gammadelta T-cell cytotoxicity. Blood 2010, 115, 2407–2411. [Google Scholar] [CrossRef] [Green Version]
- Paczulla, A.M.; Rothfelder, K.; Raffel, S.; Konantz, M.; Steinbacher, J.; Wang, H.; Tandler, C.; Mbarga, M.; Schaefer, T.; Falcone, M.; et al. Absence of NKG2D ligands defines leukaemia stem cells and mediates their immune evasion. Nature 2019, 572, 254–259. [Google Scholar] [CrossRef] [PubMed]
- Wakita, D.; Sumida, K.; Iwakura, Y.; Nishikawa, H.; Ohkuri, T.; Chamoto, K.; Kitamura, H.; Nishimura, T. Tumor-infiltrating IL-17-producing gammadelta T cells support the progression of tumor by promoting angiogenesis. Eur. J. Immunol. 2010, 40, 1927–1937. [Google Scholar] [CrossRef]
- Kabelitz, D.; Serrano, R.; Kouakanou, L.; Peters, C.; Kalyan, S. Cancer immunotherapy with γδ T cells: Many paths ahead of us. Cell Mol. Immunol. 2020, 17, 925–939. [Google Scholar] [CrossRef] [PubMed]
- Park, J.H.; Lee, H.K. Function of γδ T cells in tumor immunology and their application to cancer therapy. Exp. Mol. Med. 2021, 53, 318–327. [Google Scholar] [CrossRef]
- Hoeres, T.; Smetak, M.; Pretscher, D.; Wilhelm, M. Improving the Efficiency of Vγ9Vδ2 T-Cell Immunotherapy in Cancer. Front. Immunol. 2018, 9, 800. [Google Scholar] [CrossRef] [PubMed]
- De Weerdt, I.; Hofland, T.; Lameris, R.; Endstra, S.; Jongejan, A.; Moerland, P.D.; de Bruin, R.C.G.; Remmerswaal, E.B.M.; Ten Berge, I.J.M.; Liu, N.; et al. Improving CLL Vγ9Vδ2-T-cell fitness for cellular therapy by ex vivo activation and ibrutinib. Blood 2018, 132, 2260–2272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Argentati, K.; Re, F.; Serresi, S.; Tucci, M.G.; Bartozzi, B.; Bernardini, G.; Provinciali, M. Reduced number and impaired function of circulating gamma delta T cells in patients with cutaneous primary melanoma. J. Investig. Dermatol. 2003, 120, 829–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kabelitz, D.; Pechhold, K.; Bender, A.; Wesselborg, S.; Wesch, D.; Friese, K.; Janssen, O. Activation and activation-driven death of human gamma/delta T cells. Immunol. Rev. 1991, 120, 71–88. [Google Scholar] [CrossRef] [PubMed]
- Siegers, G.M.; Lamb, L.S., Jr. Cytotoxic and regulatory properties of circulating Vδ1+ γδ T cells: A new player on the cell therapy field? Mol. Ther. 2014, 22, 1416–1422. [Google Scholar] [CrossRef] [Green Version]
- Alves, J.J.P.; De Medeiros Fernandes, T.A.A.; De Araújo, J.M.G.; Cobucci, R.N.O.; Lanza, D.C.F.; Bezerra, F.L.; Andrade, V.S.; Fernandes, J.V. Th17 response in patients with cervical cancer. Oncol. Lett. 2018, 16, 6215–6227. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.; Zhang, X.; Xu, R.; Shang, W.; Ming, W.; Wang, F.; Wang, J. Implication of IL-17 producing ɑβT and γδT cells in patients with ovarian cancer. Hum. Immunol. 2020, 81, 244–248. [Google Scholar] [CrossRef]
- Ma, S.; Cheng, Q.; Cai, Y.; Gong, H.; Wu, Y.; Yu, X.; Shi, L.; Wu, D.; Dong, C.; Liu, H. IL-17A produced by γδ T cells promotes tumor growth in hepatocellular carcinoma. Cancer Res. 2014, 74, 1969–1982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patil, R.S.; Shah, S.U.; Shrikhande, S.V.; Goel, M.; Dikshit, R.P.; Chiplunkar, S.V. IL17 producing γδT cells induce angiogenesis and are associated with poor survival in gallbladder cancer patients. Int. J. Cancer 2016, 139, 869–881. [Google Scholar] [CrossRef]
- Carmi, Y.; Rinott, G.; Dotan, S.; Elkabets, M.; Rider, P.; Voronov, E.; Apte, R.N. Microenvironment-derived IL-1 and IL-17 interact in the control of lung metastasis. J. Immunol. 2011, 186, 3462–3471. [Google Scholar] [CrossRef] [Green Version]
- Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via γδ T Cells. Cell 2019, 176, 998–1013.e16. [Google Scholar] [CrossRef] [Green Version]
- Zhang, H.; Chai, W.; Yang, W.; Han, W.; Mou, W.; Xi, Y.; Chen, X.; Wang, H.; Wang, W.; Qin, H.; et al. The increased IL-17-producing γδT cells promote tumor cell proliferation and migration in neuroblastoma. Clin. Immunol. 2020, 211, 108343. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Kang, N.; Tang, L.; Li, X.; Wu, D.; Li, W.; Chen, X.; Cui, L.; Ba, D.; He, W. Identification and characterization of Foxp3(+) gammadelta T cells in mouse and human. Immunol. Lett. 2009, 125, 105–113. [Google Scholar] [CrossRef]
- Wesch, D.; Peters, C.; Siegers, G.M. Human gamma delta T regulatory cells in cancer: Fact or fiction? Front. Immunol. 2014, 5, 598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hua, F.; Kang, N.; Gao, Y.A.; Cui, L.X.; Ba, D.N.; He, W. Potential regulatory role of in vitro-expanded Vδ1 T cells from human peripheral blood. Immunol. Res. 2013, 56, 172–180. [Google Scholar] [CrossRef]
- Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic inflammation and cytokines in the tumor microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef] [Green Version]
- Schlecker, E.; Stojanovic, A.; Eisen, C.; Quack, C.; Falk, C.S.; Umansky, V.; Cerwenka, A. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J. Immunol. 2012, 189, 5602–5611. [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 γδ T cells promoting tumor formation. Proc. Natl. Acad. Sci. USA 2017, 114, E9056–E9065. [Google Scholar] [CrossRef] [Green Version]
- Rei, M.; Gonçalves-Sousa, N.; Lança, T.; Thompson, R.G.; Mensurado, S.; Balkwill, F.R.; Kulbe, H.; Pennington, D.J.; Silva-Santos, B. Murine CD27(-) Vγ6(+) γδ T cells producing IL-17A promote ovarian cancer growth via mobilization of protumor small peritoneal macrophages. Proc. Natl. Acad. Sci. USA 2014, 111, 3562–3570. [Google Scholar] [CrossRef] [Green Version]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef]
- Chang, C.H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [Green Version]
- Blank, C.; Brown, I.; Marks, R.; Nishimura, H.; Honjo, T.; Gajewski, T.F. Absence of programmed death receptor 1 alters thymic development and enhances generation of CD4/CD8 double-negative TCR-transgenic T cells. J. Immunol. 2003, 171, 4574–4581. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.; Crabill, G.A.; Pritchard, T.S.; McMiller, T.L.; Wei, P.; Pardoll, D.M.; Pan, F.; Topalian, S.L. Mechanisms regulating PD-L1 expression on tumor and immune cells. J. Immunother. Cancer 2019, 7, 305. [Google Scholar] [CrossRef] [PubMed]
- Wu, K.; Feng, J.; Xiu, Y.; Li, Z.; Lin, Z.; Zhao, H.; Zeng, H.; Xia, W.; Yu, L.; Xu, B. Vδ2 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]
- 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] [Green Version]
- Tanaka, Y. Cancer immunotherapy harnessing γδ T cells and programmed death-1. Immunol. Rev. 2020, 298, 237–253. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Yu, H.; Rozeboom, L.; Rivard, C.J.; Ellison, K.; Dziadziuszko, R.; Suda, K.; Ren, S.; Wu, C.; Hou, L.; et al. LAG-3 Protein Expression in Non-Small Cell Lung Cancer and Its Relationship with PD-1/PD-L1 and Tumor-Infiltrating Lymphocytes. J. Thorac. Oncol. 2017, 12, 814–823. [Google Scholar] [CrossRef] [Green Version]
- Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef]
- Bhat, S.A.; Vedpathak, D.M.; Chiplunkar, S.V. Checkpoint Blockade Rescues the Repressive Effect of Histone Deacetylases Inhibitors on γδ T Cell Function. Front. Immunol. 2018, 9, 1615. [Google Scholar] [CrossRef] [Green Version]
- Catafal-Tardos, E.; Baglioni, M.V.; Bekiaris, V. Inhibiting the Unconventionals: Importance of Immune Checkpoint Receptors in γδ T, MAIT, and NKT Cells. Cancers 2021, 13, 4647. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Flippot, R.; Escudier, B.; Albiges, L. Immune Checkpoint Inhibitors: Toward New Paradigms in Renal Cell Carcinoma. Drugs 2018, 78, 1443–1457. [Google Scholar] [CrossRef] [PubMed]
- Iwasaki, M.; Tanaka, Y.; Kobayashi, H.; Murata-Hirai, K.; Miyabe, H.; Sugie, T.; Toi, M.; Minato, N. Expression and function of PD-1 in human γδ T cells that recognize phosphoantigens. Eur. J. Immunol. 2011, 41, 345–355. [Google Scholar] [CrossRef] [Green Version]
- Hoeres, T.; Holzmann, E.; Smetak, M.; Birkmann, J.; Wilhelm, M. PD-1 signaling modulates interferon-γ production by Gamma Delta (γδ) T-Cells in response to leukemia. Oncoimmunology 2019, 8, 1550618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jonescheit, H.; Oberg, H.-H.; Gonnermann, D.; Hermes, M.; Sulaj, V.; Peters, C.; Kabelitz, D.; Wesch, D. Influence of Indoleamine-2,3-Dioxygenase and Its Metabolite Kynurenine on γδ T Cell Cytotoxicity against Ductal Pancreatic Adenocarcinoma Cells. Cells 2020, 9, 1140. [Google Scholar] [CrossRef] [PubMed]
- Li, P.; Wu, R.; Li, K.; Yuan, W.; Zeng, C.; Zhang, Y.; Wang, X.; Zhu, X.; Zhou, J.; Li, P.; et al. IDO Inhibition Facilitates Antitumor Immunity of Vγ9Vδ2 T Cells in Triple-Negative Breast Cancer. Front. Oncol. 2021, 11, 679517. [Google Scholar] [CrossRef]
- Nanda, R.; Chow, L.Q.; Dees, E.C.; Berger, R.; Gupta, S.; Geva, R.; Pusztai, L.; Pathiraja, K.; Aktan, G.; Cheng, J.D.; et al. Pembrolizumab in Patients with Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J. Clin. Oncol. 2016, 34, 2460–2467. [Google Scholar] [CrossRef]
- Le Naour, J.; Galluzzi, L.; Zitvogel, L.; Kroemer, G.; Vacchelli, E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2020, 9, 1777625. [Google Scholar] [CrossRef] [PubMed]
- Sabbione, F.; Gabelloni, M.L.; Ernst, G.; Gori, M.S.; Salamone, G.; Oleastro, M.; Trevani, A.; Geffner, J.; Jancic, C.C. Neutrophils suppress γδ T-cell function. Eur. J. Immunol. 2014, 44, 819–830. [Google Scholar] [CrossRef] [Green Version]
- Kalyan, S.; Chandrasekaran, V.; Quabius, E.S.; Lindhorst, T.K.; Kabelitz, D. Neutrophil uptake of nitrogen-bisphosphonates leads to the suppression of human peripheral blood γδ T cells. Cell Mol. Life Sci. 2014, 71, 2335–2346. [Google Scholar] [CrossRef] [PubMed]
- Mensurado, S.; Rei, M.; Lança, T.; Ioannou, M.; Gonçalves-Sousa, N.; Kubo, H.; Malissen, M.; Papayannopoulos, V.; Serre, K.; Silva-Santos, B. Tumor-associated neutrophils suppress pro-tumoral IL-17+ γδ T cells through induction of oxidative stress. PLoS Biol. 2018, 16, e2004990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yi, Y.; He, H.W.; Wang, J.X.; Cai, X.Y.; Li, Y.W.; Zhou, J.; Cheng, Y.F.; Jin, J.J.; Fan, J.; Qiu, S.J. The functional impairment of HCC-infiltrating γδ T cells, partially mediated by regulatory T cells in a TGFβ- and IL-10-dependent manner. J. Hepatol. 2013, 58, 977–983. [Google Scholar] [CrossRef] [PubMed]
- Cheng, M.; Qian, L.; Shen, G.; Bian, G.; Xu, T.; Xu, W.; Shen, G.; Hu, S. Microbiota modulate tumoral immune surveillance in lung through a γδT17 immune cell-dependent mechanism. Cancer Res. 2014, 74, 4030–4041. [Google Scholar] [CrossRef] [Green Version]
- Pro, B.; Allen, P.; Behdad, A. Hepatosplenic T-cell lymphoma: A rare but challenging entity. Blood 2020, 136, 2018–2026. [Google Scholar] [CrossRef] [PubMed]
- Yabe, M.; Miranda, R.N.; Medeiros, L.J. Hepatosplenic T-cell Lymphoma: A review of clinicopathologic features, pathogenesis, and prognostic factors. Hum. Pathol. 2018, 74, 5–16. [Google Scholar] [CrossRef]
- Matutes, E. The 2017 WHO update on mature T- and natural killer (NK) cell neoplasms. Int. J. Lab. Hematol. 2018, 40, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Macon, W.R.; Levy, N.B.; Kurtin, P.J.; Salhany, K.E.; Elkhalifa, M.Y.; Casey, T.T.; Craig, F.E.; Vnencak-Jones, C.L.; Gulley, M.L.; Park, J.P.; et al. Hepatosplenic alphabeta T-cell lymphomas: A report of 14 cases and comparison with hepatosplenic gammadelta T-cell lymphomas. Am. J. Surg. Pathol. 2001, 25, 285–296. [Google Scholar] [CrossRef]
- Foss, F.M.; Horwitz, S.M.; Civallero, M.; Bellei, M.; Marcheselli, L.; Kim, W.S.; Cabrera, M.E.; Dlouhy, I.; Nagler, A.; Advani, R.H.; et al. Incidence and outcomes of rare T cell lymphomas from the T Cell Project: Hepatosplenic, enteropathy associated and peripheral gamma delta T cell lymphomas. Am. J. Hematol. 2020, 95, 151–155. [Google Scholar] [CrossRef] [PubMed]
- McKinney, M.; Moffitt, A.B.; Gaulard, P.; Travert, M.; De Leval, L.; Nicolae, A.; Raffeld, M.; Jaffe, E.S.; Pittaluga, S.; Xi, L.; et al. The Genetic Basis of Hepatosplenic T-cell Lymphoma. Cancer Discov. 2017, 7, 369–379. [Google Scholar] [CrossRef] [Green Version]
- Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Harris, N.L.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 revision of the World Health Organization classification of lymphoid neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
- Guitart, J.; Weisenburger, D.D.; Subtil, A.; Kim, E.; Wood, G.; Duvic, M.; Olsen, E.; Junkins-Hopkins, J.; Rosen, S.; Sundram, U.; et al. Cutaneous γδ T-cell lymphomas: A spectrum of presentations with overlap with other cytotoxic lymphomas. Am. J. Surg. Pathol. 2012, 36, 1656–1665. [Google Scholar] [CrossRef]
- Toro, J.R.; Liewehr, D.J.; Pabby, N.; Sorbara, L.; Raffeld, M.; Steinberg, S.M.; Jaffe, E.S. Gamma-delta T-cell phenotype is associated with significantly decreased survival in cutaneous T-cell lymphoma. Blood 2003, 101, 3407–3412. [Google Scholar] [CrossRef] [Green Version]
- Al Somali, Z.; Hamadani, M.; Kharfan-Dabaja, M.; Sureda, A.; El Fakih, R.; Aljurf, M. Enteropathy-Associated T cell Lymphoma. Curr Hematol. Malig. Rep. 2021, 16, 140–147. [Google Scholar] [CrossRef]
- van Vliet, C.; Spagnolo, D.V. T- and NK-cell lymphoproliferative disorders of the gastrointestinal tract: Review and update. Pathology 2020, 52, 128–141. [Google Scholar] [CrossRef] [Green Version]
- van Wanrooij, R.L.; de Jong, D.; Langerak, A.W.; Ylstra, B.; van Essen, H.F.; Heideman, D.A.; Bontkes, H.J.; Mulder, C.J.; Bouma, G. Novel variant of EATL evolving from mucosal γδ-T-cells in a patient with type I RCD. BMJ Open Gastroenterol. 2015, 2, e000026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicolae, A.; Xi, L.; Pham, T.H.; Pham, T.A.; Navarro, W.; Meeker, H.G.; Pittaluga, S.; Jaffe, E.S.; Raffeld, M. Mutations in the JAK/STAT and RAS signaling pathways are common in intestinal T-cell lymphomas. Leukemia 2016, 30, 2245–2247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tse, E.; Gill, H.; Loong, F.; Kim, S.J.; Ng, S.B.; Tang, T.; Ko, Y.H.; Chng, W.J.; Lim, S.T.; Kim, W.S.; et al. Type II enteropathy-associated T-cell lymphoma: A multicenter analysis from the Asia Lymphoma Study Group. Am. J. Hematol. 2012, 87, 663–668. [Google Scholar] [CrossRef] [PubMed]
- Tan, S.Y.; Chuang, S.S.; Tang, T.; Tan, L.; Ko, Y.H.; Chuah, K.L.; Ng, S.B.; Chng, W.J.; Gatter, K.; Loong, F.; et al. Type II EATL (epitheliotropic intestinal T-cell lymphoma): A neoplasm of intra-epithelial T-cells with predominant CD8αα phenotype. Leukemia 2013, 27, 1688–1696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, J.K.; Chan, A.C.; Cheuk, W.; Wan, S.K.; Lee, W.K.; Lui, Y.H.; Chan, W.K. Type II enteropathy-associated T-cell lymphoma: A distinct aggressive lymphoma with frequent γδ T-cell receptor expression. Am. J. Surg. Pathol. 2011, 35, 1557–1569. [Google Scholar] [CrossRef] [PubMed]
- Sieniawski, M.; Angamuthu, N.; Boyd, K.; Chasty, R.; Davies, J.; Forsyth, P.; Jack, F.; Lyons, S.; Mounter, P.; Revell, P.; et al. Evaluation of enteropathy-associated T-cell lymphoma comparing standard therapies with a novel regimen including autologous stem cell transplantation. Blood 2010, 115, 3664–3670. [Google Scholar] [CrossRef] [Green Version]
- Delabie, J.; Holte, H.; Vose, J.M.; Ullrich, F.; Jaffe, E.S.; Savage, K.J.; Connors, J.M.; Rimsza, L.; Harris, N.L.; Müller-Hermelink, K.; et al. Enteropathy-associated T-cell lymphoma: Clinical and histological findings from the international peripheral T-cell lymphoma project. Blood 2011, 118, 148–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Verbeek, W.H.; Van De Water, J.M.; Al-Toma, A.; Oudejans, J.J.; Mulder, C.J.; Coupé, V.M. Incidence of enteropathy--associated T-cell lymphoma: A nation-wide study of a population-based registry in The Netherlands. Scand. J. Gastroenterol. 2008, 43, 1322–1328. [Google Scholar] [CrossRef]
- Yi, J.H.; Lee, G.W.; Do, Y.R.; Jung, H.R.; Hong, J.Y.; Yoon, D.H.; Suh, C.; Choi, Y.S.; Yi, S.Y.; Sohn, B.S.; et al. Multicenter retrospective analysis of the clinicopathologic features of monomorphic epitheliotropic intestinal T-cell lymphoma. Ann. Hematol. 2019, 98, 2541–2550. [Google Scholar] [CrossRef] [PubMed]
- Malamut, G.; Chandesris, O.; Verkarre, V.; Meresse, B.; Callens, C.; Macintyre, E.; Bouhnik, Y.; Gornet, J.M.; Allez, M.; Jian, R.; et al. Enteropathy associated T cell lymphoma in celiac disease: A large retrospective study. Dig. Liver. Dis. 2013, 45, 377–384. [Google Scholar] [CrossRef] [PubMed]
- Cheon, H.; Dziewulska, K.H.; Moosic, K.B.; Olson, K.C.; Gru, A.A.; Feith, D.J.; Loughran, T.P. Advances in the Diagnosis and Treatment of Large Granular Lymphocytic Leukemia. Curr. Hematol. Malig. Rep. 2020, 15, 103–112. [Google Scholar] [CrossRef] [PubMed]
- Sandberg, Y.; Almeida, J.; Gonzalez, M.; Lima, M.; Bárcena, P.; Szczepañski, T.; van Gastel-Mol, E.J.; Wind, H.; Balanzategui, A.; van Dongen, J.J.; et al. TCRgammadelta + large granular lymphocyte leukemias reflect the spectrum of normal antigen-selected TCRgammadelta+ T-cells. Leukemia 2006, 20, 505–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bourgault-Rouxel, A.S.; Loughran, T.P., Jr.; Zambello, R.; Epling-Burnette, P.K.; Semenzato, G.; Donadieu, J.; Amiot, L.; Fest, T.; Lamy, T. Clinical spectrum of gammadelta+ T cell LGL leukemia: Analysis of 20 cases. Leuk. Res. 2008, 32, 45–48. [Google Scholar] [CrossRef] [PubMed]
- Yabe, M.; Medeiros, L.J.; Wang, S.A.; Konoplev, S.; Ok, C.Y.; Loghavi, S.; Lu, G.; Flores, L.; Khoury, J.D.; Cason, R.C.; et al. Clinicopathologic, Immunophenotypic, Cytogenetic, and Molecular Features of γδ T-Cell Large Granular Lymphocytic Leukemia: An Analysis of 14 Patients Suggests Biologic Differences With αβ T-Cell Large Granular Lymphocytic Leukemia. Am. J. Clin. Pathol. 2015, 144, 607–619. [Google Scholar] [CrossRef]
- Barilà, G.; Teramo, A.; Calabretto, G.; Vicenzetto, C.; Gasparini, V.R.; Pavan, L.; Leoncin, M.; Vedovato, S.; Frigo, A.C.; Facco, M.; et al. Stat3 mutations impact on overall survival in large granular lymphocyte leukemia: A single-center experience of 205 patients. Leukemia 2020, 34, 1116–1124. [Google Scholar] [CrossRef] [PubMed]
- Matos, D.M.; Rizzatti, E.G.; Fernandes, M.; Buccheri, V.; Falcão, R.P. Gammadelta and alphabeta T-cell acute lymphoblastic leukemia: Comparison of their clinical and immunophenotypic features. Haematologica 2005, 90, 264–266. [Google Scholar]
- Pui, C.H.; Pei, D.; Cheng, C.; Tomchuck, S.L.; Evans, S.N.; Inaba, H.; Jeha, S.; Raimondi, S.C.; Choi, J.K.; Thomas, P.G.; et al. Treatment response and outcome of children with T-cell acute lymphoblastic leukemia expressing the gamma-delta T-cell receptor. Oncoimmunology 2019, 8, 1599637. [Google Scholar] [CrossRef] [Green Version]
- Herling, M.; Rengstl, B.; Scholtysik, R.; Hartmann, S.; Küppers, R.; Hansmann, M.-L.; Diebner, H.H.; Roeder, I.; Abken, H.; Newrzela, S.; et al. Concepts in mature T-cell lymphomas—Highlights from an international joint symposium on T-cell immunology and oncology. Leuk. Lymphoma 2017, 58, 788–796. [Google Scholar] [CrossRef] [PubMed]
- Gowda, L.; Foss, F. Hepatosplenic T-Cell Lymphomas. Cancer Treat. Res. 2019, 176, 185–193. [Google Scholar] [CrossRef]
- Yabe, M.; Medeiros, L.J.; Daneshbod, Y.; Davanlou, M.; Bueso-Ramos, C.E.; Moran, E.J.; Young, K.H.; Miranda, R.N. Hepatosplenic T-cell lymphoma arising in patients with immunodysregulatory disorders: A study of 7 patients who did not receive tumor necrosis factor-α inhibitor therapy and literature review. Ann. Diagn. Pathol. 2017, 26, 16–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kotlyar, D.S.; Osterman, M.T.; Diamond, R.H.; Porter, D.; Blonski, W.C.; Wasik, M.; Sampat, S.; Mendizabal, M.; Lin, M.V.; Lichtenstein, G.R. A systematic review of factors that contribute to hepatosplenic T-cell lymphoma in patients with inflammatory bowel disease. Clin. Gastroenterol. Hepatol. 2011, 9, 36–41.e1. [Google Scholar] [CrossRef] [PubMed]
- Lamy, T.; Moignet, A.; Loughran, T.P. LGL leukemia: From pathogenesis to treatment. Blood 2017, 129, 1082–1094. [Google Scholar] [CrossRef] [PubMed]
- Savola, P.; Kelkka, T.; Rajala, H.L.; Kuuliala, A.; Kuuliala, K.; Eldfors, S.; Ellonen, P.; Lagström, S.; Lepistö, M.; Hannunen, T.; et al. Somatic mutations in clonally expanded cytotoxic T lymphocytes in patients with newly diagnosed rheumatoid arthritis. Nat. Commun. 2017, 8, 15869. [Google Scholar] [CrossRef]
- Koskela, H.L.; Eldfors, S.; Ellonen, P.; van Adrichem, A.J.; Kuusanmäki, H.; Andersson, E.I.; Lagström, S.; Clemente, M.J.; Olson, T.; Jalkanen, S.E.; et al. Somatic STAT3 mutations in large granular lymphocytic leukemia. N. Engl. J. Med. 2012, 366, 1905–1913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, D.; Shen, M.; Li, X.; Sun, L. Roles of γδ T cells in the pathogenesis of autoimmune diseases. Clin. Dev. Immunol. 2013, 2013, 985753. [Google Scholar] [CrossRef] [Green Version]
- Langerak, A.W.; Wolvers-Tettero, I.L.; van den Beemd, M.W.; van Wering, E.R.; Ludwig, W.D.; Hählen, K.; Necker, A.; van Dongen, J.J. Immunophenotypic and immunogenotypic characteristics of TCRgammadelta+ T cell acute lymphoblastic leukemia. Leukemia 1999, 13, 206–214. [Google Scholar] [CrossRef] [PubMed]
- Falini, B.; Flenghi, L.; Pileri, S.; Pelicci, P.; Fagioli, M.; Martelli, M.F.; Moretta, L.; Ciccone, E. Distribution of T cells bearing different forms of the T cell receptor gamma/delta in normal and pathological human tissues. J. Immunol. 1989, 143, 2480–2488. [Google Scholar] [PubMed]
- Casorati, G.; De Libero, G.; Lanzavecchia, A.; Migone, N. Molecular analysis of human gamma/delta + clones from thymus and peripheral blood. J. Exp. Med. 1989, 170, 1521–1535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, A.L.; Swerdlow, S.H.; Przybylski, G.K.; Surti, U.; Choi, J.K.; Campo, E.; Trucco, M.M.; Van Oss, S.B.; Felgar, R.E. Intestinal γδ T-cell lymphomas are most frequently of type II enteropathy-associated T-cell type. Hum. Pathol. 2013, 44, 1131–1145. [Google Scholar] [CrossRef] [Green Version]
- Przybylski, G.K.; Wu, H.; Macon, W.R.; Finan, J.; Leonard, D.G.; Felgar, R.E.; DiGiuseppe, J.A.; Nowell, P.C.; Swerdlow, S.H.; Kadin, M.E.; et al. Hepatosplenic and subcutaneous panniculitis-like gamma/delta T cell lymphomas are derived from different Vdelta subsets of gamma/delta T lymphocytes. J. Mol. Diagn. 2000, 2, 11–19. [Google Scholar] [CrossRef]
- Daniels, J.; Doukas, P.G.; Escala, M.E.M.; Ringbloom, K.G.; Shih, D.J.H.; Yang, J.; Tegtmeyer, K.; Park, J.; Thomas, J.J.; Selli, M.E.; et al. Cellular origins and genetic landscape of cutaneous gamma delta T cell lymphomas. Nat. Commun. 2020, 11, 1806. [Google Scholar] [CrossRef] [PubMed]
- Arnulf, B.; Copie-Bergman, C.; Delfau-Larue, M.H.; Lavergne-Slove, A.; Bosq, J.; Wechsler, J.; Wassef, M.; Matuchansky, C.; Epardeau, B.; Stern, M.; et al. Nonhepatosplenic gammadelta T-cell lymphoma: A subset of cytotoxic lymphomas with mucosal or skin localization. Blood 1998, 91, 1723–1731. [Google Scholar] [PubMed]
- Belhadj, K.; Reyes, F.; Farcet, J.P.; Tilly, H.; Bastard, C.; Angonin, R.; Deconinck, E.; Charlotte, F.; Leblond, V.; Labouyrie, E.; et al. Hepatosplenic gammadelta T-cell lymphoma is a rare clinicopathologic entity with poor outcome: Report on a series of 21 patients. Blood 2003, 102, 4261–4269. [Google Scholar] [CrossRef] [Green Version]
- Finalet Ferreiro, J.; Rouhigharabaei, L.; Urbankova, H.; van der Krogt, J.A.; Michaux, L.; Shetty, S.; Krenacs, L.; Tousseyn, T.; De Paepe, P.; Uyttebroeck, A.; et al. Integrative genomic and transcriptomic analysis identified candidate genes implicated in the pathogenesis of hepatosplenic T-cell lymphoma. PLoS ONE 2014, 9, e102977. [Google Scholar] [CrossRef] [Green Version]
- Roberti, A.; Dobay, M.P.; Bisig, B.; Vallois, D.; Boéchat, C.; Lanitis, E.; Bouchindhomme, B.; Parrens, M.C.; Bossard, C.; Quintanilla-Martinez, L.; et al. Type II enteropathy-associated T-cell lymphoma features a unique genomic profile with highly recurrent SETD2 alterations. Nat. Commun. 2016, 7, 12602. [Google Scholar] [CrossRef]
- Ben Abdelali, R.; Roggy, A.; Leguay, T.; Cieslak, A.; Renneville, A.; Touzart, A.; Banos, A.; Randriamalala, E.; Caillot, D.; Lioure, B.; et al. SET-NUP214 is a recurrent γδ lineage-specific fusion transcript associated with corticosteroid/chemotherapy resistance in adult T-ALL. Blood 2014, 123, 1860–1863. [Google Scholar] [CrossRef] [Green Version]
- Asnafi, V.; Radford-Weiss, I.; Dastugue, N.; Bayle, C.; Leboeuf, D.; Charrin, C.; Garand, R.; Lafage-Pochitaloff, M.; Delabesse, E.; Buzyn, A.; et al. CALM-AF10 is a common fusion transcript in T-ALL and is specific to the TCRgammadelta lineage. Blood 2003, 102, 1000–1006. [Google Scholar] [CrossRef] [PubMed]
- Schrader, A.; Crispatzu, G.; Oberbeck, S.; Mayer, P.; Pützer, S.; von Jan, J.; Vasyutina, E.; Warner, K.; Weit, N.; Pflug, N.; et al. Actionable perturbations of damage responses by TCL1/ATM and epigenetic lesions form the basis of T-PLL. Nat. Commun. 2018, 9, 697. [Google Scholar] [CrossRef]
- Chiarle, R.; Voena, C.; Ambrogio, C.; Piva, R.; Inghirami, G. The anaplastic lymphoma kinase in the pathogenesis of cancer. Nat. Rev. Cancer 2008, 8, 11–23. [Google Scholar] [CrossRef]
- Muro, R.; Takayanagi, H.; Nitta, T. T cell receptor signaling for γδT cell development. Inflamm Regen 2019, 39, 6. [Google Scholar] [CrossRef]
- Fiala, G.J.; Gomes, A.Q.; Silva-Santos, B. From thymus to periphery: Molecular basis of effector γδ-T cell differentiation. Immunol. Rev. 2020, 298, 47–60. [Google Scholar] [CrossRef] [PubMed]
- Hoelbl, A.; Kovacic, B.; Kerenyi, M.A.; Simma, O.; Warsch, W.; Cui, Y.; Beug, H.; Hennighausen, L.; Moriggl, R.; Sexl, V. Clarifying the role of Stat5 in lymphoid development and Abelson-induced transformation. Blood 2006, 107, 4898–4906. [Google Scholar] [CrossRef] [Green Version]
- Orlova, A.; Wingelhofer, B.; Neubauer, H.A.; Maurer, B.; Berger-Becvar, A.; Keserű, G.M.; Gunning, P.T.; Valent, P.; Moriggl, R. Emerging therapeutic targets in myeloproliferative neoplasms and peripheral T-cell leukemia and lymphomas. Expert Opin. Ther. Targets 2018, 22, 45–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wingelhofer, B.; Neubauer, H.A.; Valent, P.; Han, X.; Constantinescu, S.N.; Gunning, P.T.; Müller, M.; Moriggl, R. Implications of STAT3 and STAT5 signaling on gene regulation and chromatin remodeling in hematopoietic cancer. Leukemia 2018, 32, 1713–1726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brachet-Botineau, M.; Polomski, M.; Neubauer, H.A.; Juen, L.; Hédou, D.; Viaud-Massuard, M.-C.; Prié, G.; Gouilleux, F. Pharmacological Inhibition of Oncogenic STAT3 and STAT5 Signaling in Hematopoietic Cancers. Cancers 2020, 12, 240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pham, H.T.T.; Maurer, B.; Prchal-Murphy, M.; Grausenburger, R.; Grundschober, E.; Javaheri, T.; Nivarthi, H.; Boersma, A.; Kolbe, T.; Elabd, M.; et al. STAT5BN642H is a driver mutation for T cell neoplasia. J. Clin. Investig. 2018, 128, 387–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Araujo, E.D.; Erdogan, F.; Neubauer, H.A.; Meneksedag-Erol, D.; Manaswiyoungkul, P.; Eram, M.S.; Seo, H.S.; Qadree, A.K.; Israelian, J.; Orlova, A.; et al. Structural and functional consequences of the STAT5B. Nat. Commun. 2019, 10, 2517. [Google Scholar] [CrossRef] [Green Version]
- Nicolae, A.; Xi, L.; Pittaluga, S.; Abdullaev, Z.; Pack, S.D.; Chen, J.; Waldmann, T.A.; Jaffe, E.S.; Raffeld, M. Frequent STAT5B mutations in γδ hepatosplenic T-cell lymphomas. Leukemia 2014, 28, 2244–2248. [Google Scholar] [CrossRef] [PubMed]
- Moffitt, A.B.; Ondrejka, S.L.; McKinney, M.; Rempel, R.E.; Goodlad, J.R.; Teh, C.H.; Leppa, S.; Mannisto, S.; Kovanen, P.E.; Tse, E.; et al. Enteropathy-associated T cell lymphoma subtypes are characterized by loss of function of SETD2. J. Exp. Med. 2017, 214, 1371–1386. [Google Scholar] [CrossRef] [PubMed]
- Ribeiro, S.T.; Tesio, M.; Ribot, J.C.; Macintyre, E.; Barata, J.T.; Silva-Santos, B. Casein kinase 2 controls the survival of normal thymic and leukemic γδ T cells via promotion of AKT signaling. Leukemia 2017, 31, 1603–1610. [Google Scholar] [CrossRef] [PubMed]
- Kim, R.; Boissel, N.; Touzart, A.; Leguay, T.; Thonier, F.; Thomas, X.; Raffoux, E.; Huguet, F.; Villarese, P.; Fourrage, C.; et al. Adult T-cell acute lymphoblastic leukemias with IL7R pathway mutations are slow-responders who do not benefit from allogeneic stem-cell transplantation. Leukemia 2020, 34, 1730–1740. [Google Scholar] [CrossRef]
- Santana, V.M.; Abromowitch, M.; Sandlund, J.T.; Behm, F.G.; Ayers, G.D.; Roberson, P.K.; Pui, C.H. MACOP-B treatment in children and adolescents with advanced diffuse large-cell non-Hodgkin’s lymphoma. Leukemia 1993, 7, 187–191. [Google Scholar]
- Zinzani, P.L.; Tura, S.; Cajozzo, A.; Leone, G.; Papa, G.; Gentilini, P.; Rossi, G.; Aitini, E.; Mandelli, F. Anthracycline containing regimens in intermediate grade lymphoma. Italian Cooperative Study Group on Intermediate Grade Malignant Lymphoma. Leuk. Lymphoma 1993, 10, 39–41. [Google Scholar] [CrossRef]
- Bezwoda, W.R.; Rastogi, R.B. A randomized comparative study of cyclophosphamide, vincristine, doxorubicin, and prednisolone and cyclophosphamide, vincristine, mitoxantrone, and prednisolone regimens in the treatment of intermediate- and high-grade lymphoma with 8 years’ follow-up. Semin. Hematol. 1994, 31, 3. [Google Scholar] [PubMed]
- Köppler, H.; Pflüger, K.H.; Eschenbach, I.; Pfab, R.; Birkmann, J.; Zeller, W.; Holle, R.; Steinhauer, U.E.; Gropp, C.; Oehl, S.; et al. Randomised comparison of CHOEP versus alternating hCHOP/IVEP for high-grade non-Hodgkin’s lymphomas: Treatment results and prognostic factor analysis in a multi-centre trial. Ann. Oncol. 1994, 5, 49–55. [Google Scholar] [CrossRef]
- Kesavan, M.; Eyre, T.A.; Collins, G.P. Front-Line Treatment of High Grade B Cell Non-Hodgkin Lymphoma. Curr. Hematol. Malig. Rep. 2019, 14, 207–218. [Google Scholar] [CrossRef] [Green Version]
- Schmitz, N.; Nickelsen, M.; Ziepert, M.; Haenel, M.; Borchmann, P.; Schmidt, C.; Viardot, A.; Bentz, M.; Peter, N.; Ehninger, G.; et al. Conventional chemotherapy (CHOEP-14) with rituximab or high-dose chemotherapy (MegaCHOEP) with rituximab for young, high-risk patients with aggressive B-cell lymphoma: An open-label, randomised, phase 3 trial (DSHNHL 2002-1). Lancet Oncol. 2012, 13, 1250–1259. [Google Scholar] [CrossRef]
- Cunningham, D.; Hawkes, E.A.; Jack, A.; Qian, W.; Smith, P.; Mouncey, P.; Pocock, C.; Ardeshna, K.M.; Radford, J.A.; McMillan, A.; et al. Rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisolone in patients with newly diagnosed diffuse large B-cell non-Hodgkin lymphoma: A phase 3 comparison of dose intensification with 14-day versus 21-day cycles. Lancet 2013, 381, 1817–1826. [Google Scholar] [CrossRef] [Green Version]
- Molina, T.J.; Canioni, D.; Copie-Bergman, C.; Recher, C.; Brière, J.; Haioun, C.; Berger, F.; Fermé, C.; Copin, M.C.; Casasnovas, O.; et al. Young patients with non-germinal center B-cell-like diffuse large B-cell lymphoma benefit from intensified chemotherapy with ACVBP plus rituximab compared with CHOP plus rituximab: Analysis of data from the Groupe d’Etudes des Lymphomes de l’Adulte/lymphoma study association phase III trial LNH 03-2B. J. Clin. Oncol. 2014, 32, 3996–4003. [Google Scholar] [CrossRef] [PubMed]
- Dinçol, D.; Akbulut, H.; Içli, F.; Samur, M.; Karaoğuz, H.; Demirkazik, A.; Cay, F. Results of the cyclophosphamide, doxorubicin, vincristine, prednisolone (CHOP) +/- bleomycin treatment and evaluation of prognostic factors in aggressive lymphomas in Turkey. Oncology 1997, 54, 376–379. [Google Scholar] [CrossRef] [PubMed]
- Niitsu, N.; Okamoto, M.; Kuraishi, Y.; Nakamura, S.; Kodama, F.; Hirano, M. CyclOBEAP (cyclophosphamide, vincristine, bleomycin, etoposide, doxorubicin, prednisolone) regimen with granulocyte colony-stimulating factor (G-CSF) for patients with aggressive non-Hodgkin’s lymphoma: A pilot study. The Adult Lymphoma Treatment Study Group (ALTSG). Eur. J. Haematol. 2000, 65, 188–194. [Google Scholar] [CrossRef]
- Meerwaldt, J.H.; Carde, P.; Somers, R.; Thomas, J.; Kluin-Nelemans, J.C.; Bron, D.; Noordijk, E.M.; Cosset, J.M.; Bijnens, L.; Teodorovic, I.; et al. Persistent improved results after adding vincristine and bleomycin to a cyclophosphamide/hydroxorubicin/Vm-26/prednisone combination (CHVmP) in stage III-IV intermediate- and high-grade non-Hodgkin’s lymphoma. The EORTC Lymphoma Cooperative Group. Ann. Oncol. 1997, 8, 67–70. [Google Scholar] [CrossRef] [PubMed]
- Wilder, D.D.; Ogden, J.L.; Jain, V.K. Efficacy of fludarabine/mitoxantrone/dexamethasone alternating with CHOP in bulky follicular non-Hodgkin’s lymphoma. Clin. Lymphoma 2002, 2, 229–237. [Google Scholar] [CrossRef] [PubMed]
- Sakai, R.; Fujisawa, S.; Fujimaki, K.; Kanamori, H.; Ishigatsubo, Y. Long-term remission in a patient with hepatosplenic gammadelta T cell lymphoma after cord blood stem cell transplantation following autologous peripheral blood stem cell transplantation. Bone Marrow Transplant. 2006, 37, 537–538. [Google Scholar] [CrossRef] [PubMed]
- Gassas, A.; Kirby, M.; Weitzman, S.; Ngan, B.; Abla, O.; Doyle, J.J. Hepatosplenic gammadelta T-cell lymphoma in a 10-year-old boy successfully treated with hematopoietic stem cell transplantation. Am. J. Hematol. 2004, 75, 113–114. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Jiang, Y.; Zhu, Q.; Duan, Y.; Chen, X.; Xu, T.; Jin, Z.; Li, C.; Wu, D.; Huang, H. Clinical features and treatment outcomes of 14 patients with hepatosplenic γ δ T-cell lymphoma. J. Cancer Res. Clin. Oncol. 2021, 147, 3441–3445. [Google Scholar] [CrossRef] [PubMed]
- Carlo-Stella, C.; Delarue, R.; Scarfo, L.; Barde, P.J.; Nair, A.; Locatelli, S.L.; Morello, L.; Magagnoli, M.; Vakkalanka, S.; Viswanadha, S.; et al. A First-in-human Study of Tenalisib (RP6530), a Dual PI3K δ/γ Inhibitor, in Patients with Relapsed/Refractory Hematologic Malignancies: Results from the European Study. Clin. Lymphoma Myeloma. Leuk. 2020, 20, 78–86. [Google Scholar] [CrossRef] [PubMed]
- Laribi, K.; Alani, M.; Truong, C.; Baugier de Materre, A. Recent Advances in the Treatment of Peripheral T-Cell Lymphoma. Oncologist 2018, 23, 1039–1053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.T.; Guo, W.; Sun, M.; Han, W.; Du, Z.H.; Wang, X.X.; Du, B.B.; Bai, O. Effect of chidamide on treating hepatosplenic T-cell lymphoma: A case report. World J. Clin. Cases 2020, 8, 3122–3129. [Google Scholar] [CrossRef] [PubMed]
- Aldinucci, D.; Poletto, D.; Zagonel, V.; Rupolo, M.; Degan, M.; Nanni, P.; Gattei, V.; Pinto, A. In vitro and in vivo effects of 2'-deoxycoformycin (Pentostatin) on tumour cells from human gammadelta+ T-cell malignancies. Br. J. Haematol. 2000, 110, 188–196. [Google Scholar] [CrossRef] [PubMed]
- Grigg, A.P. 2′-Deoxycoformycin for hepatosplenic gammadelta T-cell lymphoma. Leuk. Lymphoma 2001, 42, 797–799. [Google Scholar] [CrossRef] [PubMed]
- Iannitto, E.; Barbera, V.; Quintini, G.; Cirrincione, S.; Leone, M. Hepatosplenic gammadelta T-cell lymphoma: Complete response induced by treatment with pentostatin. Br. J. Haematol. 2002, 117, 995–996. [Google Scholar] [CrossRef] [PubMed]
- Bennett, M.; Matutes, E.; Gaulard, P. Hepatosplenic T cell lymphoma responsive to 2'-deoxycoformycin therapy. Am. J. Hematol. 2010, 85, 727–729. [Google Scholar] [CrossRef] [PubMed]
- Bergmann, A.K.; Fataccioli, V.; Castellano, G.; Martin-Garcia, N.; Pelletier, L.; Ammerpohl, O.; Bergmann, J.; Bhat, J.; Pau, E.C.S.; Martín-Subero, J.I.; et al. DNA methylation profiling of hepatosplenic T-cell lymphoma. Haematologica 2019, 104, e104–e107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Schönefeldt, S.; Wais, T.; Herling, M.; Mustjoki, S.; Bekiaris, V.; Moriggl, R.; Neubauer, H.A. The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma. Cancers 2021, 13, 6212. https://doi.org/10.3390/cancers13246212
Schönefeldt S, Wais T, Herling M, Mustjoki S, Bekiaris V, Moriggl R, Neubauer HA. The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma. Cancers. 2021; 13(24):6212. https://doi.org/10.3390/cancers13246212
Chicago/Turabian StyleSchönefeldt, Susann, Tamara Wais, Marco Herling, Satu Mustjoki, Vasileios Bekiaris, Richard Moriggl, and Heidi A. Neubauer. 2021. "The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma" Cancers 13, no. 24: 6212. https://doi.org/10.3390/cancers13246212
APA StyleSchönefeldt, S., Wais, T., Herling, M., Mustjoki, S., Bekiaris, V., Moriggl, R., & Neubauer, H. A. (2021). The Diverse Roles of γδ T Cells in Cancer: From Rapid Immunity to Aggressive Lymphoma. Cancers, 13(24), 6212. https://doi.org/10.3390/cancers13246212