Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer
Abstract
Simple Summary
Abstract
1. Introduction
2. Regulation of NK Cell Activation and Cytotoxic Activity
3. Inhibitory Receptors Binding to MHC-I Molecules
4. Inhibitory Receptors Binding to Nectin and Nectin-Like Molecules
4.1. TIGIT
4.2. CD96
4.3. CD112R
5. T Cell-Associated Immune Checkpoints
5.1. Members of the B7-CD28 Superfamily
5.1.1. PD-1
5.1.2. CTLA-4
5.1.3. BTLA (CD272)
5.1.4. Receptor(s) to B7-H3
5.1.5. VISTA
5.2. LAG-3
5.3. TIM-3
6. Other Inhibitory Pathways
6.1. Adenosinergic Signaling
6.2. Interleukin-1 Receptor 8 (IL-1R8)
6.3. KIR3DL3
6.4. CD161
6.5. Siglec Receptors
6.6. CD300a
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Guillerey, C. Roles of cytotoxic and helper innate lymphoid cells in cancer. Mamm. Genome 2018, 29, 777–789. [Google Scholar] [CrossRef]
- Guillerey, C. NK Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1273, 69–90. [Google Scholar] [CrossRef]
- Prager, I.; Liesche, C.; van Ooijen, H.; Urlaub, D.; Verron, Q.; Sandstrom, N.; Fasbender, F.; Claus, M.; Eils, R.; Beaudouin, J.; et al. NK cells switch from granzyme B to death receptor-mediated cytotoxicity during serial killing. J. Exp. Med. 2019, 216, 2113–2127. [Google Scholar] [CrossRef]
- Glasner, A.; Levi, A.; Enk, J.; Isaacson, B.; Viukov, S.; Orlanski, S.; Scope, A.; Neuman, T.; Enk, C.D.; Hanna, J.H.; et al. NKp46 Receptor-Mediated Interferon-gamma Production by Natural Killer Cells Increases Fibronectin 1 to Alter Tumor Architecture and Control Metastasis. Immunity 2018, 48, 396–398. [Google Scholar] [CrossRef]
- Correia, A.L.; Guimaraes, J.C.; Auf der Maur, P.; De Silva, D.; Trefny, M.P.; Okamoto, R.; Bruno, S.; Schmidt, A.; Mertz, K.; Volkmann, K.; et al. Hepatic stellate cells suppress NK cell-sustained breast cancer dormancy. Nature 2021. [Google Scholar] [CrossRef]
- Bald, T.; Pedde, A.M.; Corvino, D.; Bottcher, J.P. The role of NK cell as central communicators in cancer immunity. Adv. Immunol. 2020, 147, 61–88. [Google Scholar] [CrossRef]
- Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef] [PubMed]
- Bottcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e1014. [Google Scholar] [CrossRef] [PubMed]
- Guillerey, C.; Smyth, M.J. NK Cells and Cancer Immunoediting. Curr Top. Microbiol. Immunol. 2016, 395, 115–145. [Google Scholar] [CrossRef]
- Huntington, N.D.; Cursons, J.; Rautela, J. The cancer-natural killer cell immunity cycle. Nat. Rev. Cancer 2020, 20, 437–454. [Google Scholar] [CrossRef]
- Duault, C.; Kumar, A.; Taghi Khani, A.; Lee, S.J.; Yang, L.; Huang, M.; Hurtz, C.; Manning, B.; Ghoda, L.Y.; McDonald, T.; et al. Activated Natural Killer Cells Predict Poor Clinical Prognosis in High-risk B- and T- cell Acute Lymphoblastic Leukemia. Blood 2021. [Google Scholar] [CrossRef]
- Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef] [PubMed]
- Myers, J.A.; Miller, J.S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18, 85–100. [Google Scholar] [CrossRef]
- Huang, R.S.; Lai, M.C.; Shih, H.A.; Lin, S. A robust platform for expansion and genome editing of primary human natural killer cells. J. Exp. Med. 2021, 218. [Google Scholar] [CrossRef]
- Daher, M.; Basar, R.; Gokdemir, E.; Baran, N.; Uprety, N.; Nunez Cortes, A.K.; Mendt, M.; Kerbauy, L.N.; Banerjee, P.P.; Shanley, M.; et al. Targeting a cytokine checkpoint enhances the fitness of armored cord blood CAR-NK cells. Blood 2021, 137, 624–636. [Google Scholar] [CrossRef]
- Delconte, R.B.; Kolesnik, T.B.; Dagley, L.F.; Rautela, J.; Shi, W.; Putz, E.M.; Stannard, K.; Zhang, J.G.; Teh, C.; Firth, M.; et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat. Immunol. 2016, 17, 816–824. [Google Scholar] [CrossRef]
- Viel, S.; Marcais, A.; Guimaraes, F.S.; Loftus, R.; Rabilloud, J.; Grau, M.; Degouve, S.; Djebali, S.; Sanlaville, A.; Charrier, E.; et al. TGF-beta inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci. Signal. 2016, 9, ra19. [Google Scholar] [CrossRef] [PubMed]
- Rautela, J.; Dagley, L.F.; de Oliveira, C.C.; Schuster, I.S.; Hediyeh-Zadeh, S.; Delconte, R.B.; Cursons, J.; Hennessy, R.; Hutchinson, D.S.; Harrison, C.; et al. Therapeutic blockade of activin-A improves NK cell function and antitumor immunity. Sci. Signal. 2019, 12. [Google Scholar] [CrossRef]
- Lanuza, P.M.; Pesini, C.; Arias, M.A.; Calvo, C.; Ramirez-Labrada, A.; Pardo, J. Recalling the Biological Significance of Immune Checkpoints on NK Cells: A Chance to Overcome LAG3, PD1, and CTLA4 Inhibitory Pathways by Adoptive NK Cell Transfer? Front. Immunol. 2019, 10, 3010. [Google Scholar] [CrossRef]
- Cooper, M.A.; Fehniger, T.A.; Caligiuri, M.A. The biology of human natural killer-cell subsets. Trends Immunol. 2001, 22, 633–640. [Google Scholar] [CrossRef]
- Freud, A.G.; Mundy-Bosse, B.L.; Yu, J.; Caligiuri, M.A. The Broad Spectrum of Human Natural Killer Cell Diversity. Immunity 2017, 47, 820–833. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Ng, S.S.; De Labastida Rivera, F.; Yan, J.; Corvino, D.; Das, I.; Zhang, P.; Kuns, R.; Chauhan, S.B.; Hou, J.; Li, X.Y.; et al. The NK cell granule protein NKG7 regulates cytotoxic granule exocytosis and inflammation. Nat. Immunol. 2020, 21, 1205–1218. [Google Scholar] [CrossRef] [PubMed]
- Verron, Q.; Forslund, E.; Brandt, L.; Leino, M.; Frisk, T.W.; Olofsson, P.E.; Onfelt, B. NK cells integrate signals over large areas when building immune synapses but require local stimuli for degranulation. Sci. Signal. 2021, 14. [Google Scholar] [CrossRef] [PubMed]
- Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
- Quatrini, L.; Della Chiesa, M.; Sivori, S.; Mingari, M.C.; Pende, D.; Moretta, L. Human NK cells, their receptors and function. Eur. J. Immunol. 2021, 51, 1566–1579. [Google Scholar] [CrossRef] [PubMed]
- Vivier, E.; Nunes, J.A.; Vely, F. Natural killer cell signaling pathways. Science 2004, 306, 1517–1519. [Google Scholar] [CrossRef]
- Sivori, S.; Della Chiesa, M.; Carlomagno, S.; Quatrini, L.; Munari, E.; Vacca, P.; Tumino, N.; Mariotti, F.R.; Mingari, M.C.; Pende, D.; et al. Inhibitory Receptors and Checkpoints in Human NK Cells, Implications for the Immunotherapy of Cancer. Front. Immunol. 2020, 11, 2156. [Google Scholar] [CrossRef]
- Moretta, A.; Bottino, C.; Vitale, M.; Pende, D.; Biassoni, R.; Mingari, M.C.; Moretta, L. Receptors for HLA class-I molecules in human natural killer cells. Annu. Rev. Immunol. 1996, 14, 619–648. [Google Scholar] [CrossRef]
- 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]
- Colonna, M.; Navarro, F.; Bellon, T.; Llano, M.; Garcia, P.; Samaridis, J.; Angman, L.; Cella, M.; Lopez-Botet, M. A common inhibitory receptor for major histocompatibility complex class I molecules on human lymphoid and myelomonocytic cells. J. Exp. Med. 1997, 186, 1809–1818. [Google Scholar] [CrossRef] [PubMed]
- Long, E.O. Negative signaling by inhibitory receptors: The NK cell paradigm. Immunol. Rev. 2008, 224, 70–84. [Google Scholar] [CrossRef] [PubMed]
- Elliott, J.M.; Yokoyama, W.M. Unifying concepts of MHC-dependent natural killer cell education. Trends Immunol. 2011, 32, 364–372. [Google Scholar] [CrossRef]
- van Hall, T.; André, 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]
- Carlsten, M.; Korde, N.; Kotecha, R.; Reger, R.; Bor, S.; Kazandjian, D.; Landgren, O.; Childs, R.W. Checkpoint Inhibition of KIR2D with the Monoclonal Antibody IPH2101 Induces Contraction and Hyporesponsiveness of NK Cells in Patients with Myeloma. Clin. Cancer Res. 2016, 22, 5211–5222. [Google Scholar] [CrossRef]
- 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]
- van Montfoort, N.; Borst, L.; Korrer, M.J.; Sluijter, M.; Marijt, K.A.; Santegoets, S.J.; van Ham, V.J.; Ehsan, I.; Charoentong, P.; André, P.; et al. NKG2A Blockade Potentiates CD8 T Cell Immunity Induced by Cancer Vaccines. Cell 2018, 175, 1744–1755.e1715. [Google Scholar] [CrossRef] [PubMed]
- André, P.; Denis, C.; Soulas, C.; Bourbon-Caillet, C.; Lopez, J.; Arnoux, T.; Bléry, 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.e1713. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Correa, B.; Valhondo, I.; Hassouneh, F.; Lopez-Sejas, N.; Pera, A.; Bergua, J.M.; Arcos, M.J.; Banas, H.; Casas-Aviles, I.; Duran, E.; et al. DNAM-1 and the TIGIT/PVRIG/TACTILE Axis: Novel Immune Checkpoints for Natural Killer Cell-Based Cancer Immunotherapy. Cancers 2019, 11, 877. [Google Scholar] [CrossRef]
- Harjunpaa, H.; Guillerey, C. TIGIT as an emerging immune checkpoint. Clin. Exp. Immunol. 2020, 200, 108–119. [Google Scholar] [CrossRef]
- Xu, F.; Sunderland, A.; Zhou, Y.; Schulick, R.D.; Edil, B.H.; Zhu, Y. Blockade of CD112R and TIGIT signaling sensitizes human natural killer cell functions. Cancer Immunol. Immunother. 2017, 66, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
- Du, W.; Yang, M.; Turner, A.; Xu, C.; Ferris, R.L.; Huang, J.; Kane, L.P.; Lu, B. TIM-3 as a Target for Cancer Immunotherapy and Mechanisms of Action. Int. J. Mol. Sci. 2017, 18, 645. [Google Scholar] [CrossRef] [PubMed]
- Chiang, E.Y.; de Almeida, P.E.; de Almeida Nagata, D.E.; Bowles, K.H.; Du, X.; Chitre, A.S.; Banta, K.L.; Kwon, Y.; McKenzie, B.; Mittman, S.; et al. CD96 functions as a co-stimulatory receptor to enhance CD8(+) T cell activation and effector responses. Eur J. Immunol. 2020, 50, 891–902. [Google Scholar] [CrossRef] [PubMed]
- Chan, C.J.; Martinet, L.; Gilfillan, S.; Souza-Fonseca-Guimaraes, F.; Chow, M.T.; Town, L.; Ritchie, D.S.; Colonna, M.; Andrews, D.M.; Smyth, M.J. The receptors CD96 and CD226 oppose each other in the regulation of natural killer cell functions. Nat. Immunol. 2014, 15, 431–438. [Google Scholar] [CrossRef]
- Takai, Y.; Miyoshi, J.; Ikeda, W.; Ogita, H. Nectins and nectin-like molecules: Roles in contact inhibition of cell movement and proliferation. Nat. Rev. Mol. Cell Biol. 2008, 9, 603–615. [Google Scholar] [CrossRef]
- Fuchs, A.; Colonna, M. The role of NK cell recognition of nectin and nectin-like proteins in tumor immunosurveillance. Semin Cancer Biol 2006, 16, 359–366. [Google Scholar] [CrossRef]
- Whelan, S.; Ophir, E.; Kotturi, M.F.; Levy, O.; Ganguly, S.; Leung, L.; Vaknin, I.; Kumar, S.; Dassa, L.; Hansen, K.; et al. PVRIG and PVRL2 Are Induced in Cancer and Inhibit CD8(+) T-cell Function. Cancer Immunol. Res. 2019, 7, 257–268. [Google Scholar] [CrossRef] [PubMed]
- Sarhan, D.; Cichocki, F.; Zhang, B.; Yingst, A.; Spellman, S.R.; Cooley, S.; Verneris, M.R.; Blazar, B.R.; Miller, J.S. Adaptive NK Cells with Low TIGIT Expression Are Inherently Resistant to Myeloid-Derived Suppressor Cells. Cancer Res. 2016, 76, 5696–5706. [Google Scholar] [CrossRef]
- Yin, X.; Liu, T.; Wang, Z.; Ma, M.; Lei, J.; Zhang, Z.; Fu, S.; Fu, Y.; Hu, Q.; Ding, H.; et al. Expression of the Inhibitory Receptor TIGIT Is Up-Regulated Specifically on NK Cells With CD226 Activating Receptor From HIV-Infected Individuals. Front. Immunol. 2018, 9, 2341. [Google Scholar] [CrossRef]
- Wang, F.; Hou, H.; Wu, S.; Tang, Q.; Liu, W.; Huang, M.; Yin, B.; Huang, J.; Mao, L.; Lu, Y.; et al. TIGIT expression levels on human NK cells correlate with functional heterogeneity among healthy individuals. Eur J. Immunol. 2015, 45, 2886–2897. [Google Scholar] [CrossRef]
- Stannard, K.A.; Lemoine, S.; Waterhouse, N.J.; Vari, F.; Chatenoud, L.; Gandhi, M.K.; Martinet, L.; Smyth, M.J.; Guillerey, C. Human peripheral blood DNAM-1(neg) NK cells are a terminally differentiated subset with limited effector functions. Blood Adv. 2019, 3, 1681–1694. [Google Scholar] [CrossRef] [PubMed]
- He, Y.; Peng, H.; Sun, R.; Wei, H.; Ljunggren, H.G.; Yokoyama, W.M.; Tian, Z. Contribution of inhibitory receptor TIGIT to NK cell education. J. Autoimmun. 2017, 81, 1–12. [Google Scholar] [CrossRef]
- Maas, R.J.; Hoogstad-van Evert, J.S.; Van der Meer, J.M.; Mekers, V.; Rezaeifard, S.; Korman, A.J.; de Jonge, P.K.; Cany, J.; Woestenenk, R.; Schaap, N.P.; et al. TIGIT blockade enhances functionality of peritoneal NK cells with altered expression of DNAM-1/TIGIT/CD96 checkpoint molecules in ovarian cancer. Oncoimmunology 2020, 9, 1843247. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef] [PubMed]
- Chauvin, J.M.; Ka, M.; Pagliano, O.; Menna, C.; Ding, Q.; DeBlasio, R.; Sanders, C.; Hou, J.; Li, X.Y.; Ferrone, S.; et al. IL15 Stimulation with TIGIT Blockade Reverses CD155-mediated NK-Cell Dysfunction in Melanoma. Clin. Cancer Res. 2020, 26, 5520–5533. [Google Scholar] [CrossRef] [PubMed]
- Sun, H.; Huang, Q.; Huang, M.; Wen, H.; Lin, R.; Zheng, M.; Qu, K.; Li, K.; Wei, H.; Xiao, W.; et al. Human CD96 Correlates to Natural Killer Cell Exhaustion and Predicts the Prognosis of Human Hepatocellular Carcinoma. Hepatology 2019, 70, 168–183. [Google Scholar] [CrossRef]
- Zheng, Q.; Wang, B.; Gao, J.; Xin, N.; Wang, W.; Song, X.; Shao, Y.; Zhao, C. CD155 knockdown promotes apoptosis via AKT/Bcl-2/Bax in colon cancer cells. J. Cell Mol. Med. 2018, 22, 131–140. [Google Scholar] [CrossRef]
- Stanietsky, N.; Simic, H.; Arapovic, J.; Toporik, A.; Levy, O.; Novik, A.; Levine, Z.; Beiman, M.; Dassa, L.; Achdout, H.; et al. The interaction of TIGIT with PVR and PVRL2 inhibits human NK cell cytotoxicity. Proc. Natl. Acad. Sci. USA 2009, 106, 17858–17863. [Google Scholar] [CrossRef]
- Liu, S.; Zhang, H.; Li, M.; Hu, D.; Li, C.; Ge, B.; Jin, B.; Fan, Z. Recruitment of Grb2 and SHIP1 by the ITT-like motif of TIGIT suppresses granule polarization and cytotoxicity of NK cells. Cell Death Differ. 2013, 20, 456–464. [Google Scholar] [CrossRef]
- Li, M.; Xia, P.; Du, Y.; Liu, S.; Huang, G.; Chen, J.; Zhang, H.; Hou, N.; Cheng, X.; Zhou, L.; et al. T-cell immunoglobulin and ITIM domain (TIGIT) receptor/poliovirus receptor (PVR) ligand engagement suppresses interferon-gamma production of natural killer cells via beta-arrestin 2-mediated negative signaling. J. Biol. Chem. 2014, 289, 17647–17657. [Google Scholar] [CrossRef]
- Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 2009, 10, 48–57. [Google Scholar] [CrossRef]
- Carlsten, M.; Bjorkstrom, N.K.; Norell, H.; Bryceson, Y.; van Hall, T.; Baumann, B.C.; Hanson, M.; Schedvins, K.; Kiessling, R.; Ljunggren, H.G.; et al. DNAX accessory molecule-1 mediated recognition of freshly isolated ovarian carcinoma by resting natural killer cells. Cancer Res. 2007, 67, 1317–1325. [Google Scholar] [CrossRef] [PubMed]
- Cluxton, C.D.; Spillane, C.; O’Toole, S.A.; Sheils, O.; Gardiner, C.M.; O’Leary, J.J. Suppression of Natural Killer cell NKG2D and CD226 anti-tumour cascades by platelet cloaked cancer cells: Implications for the metastatic cascade. PLoS ONE 2019, 14, e0211538. [Google Scholar] [CrossRef] [PubMed]
- Peng, Y.P.; Xi, C.H.; Zhu, Y.; Yin, L.D.; Wei, J.S.; Zhang, J.J.; Liu, X.C.; Guo, S.; Fu, Y.; Miao, Y. Altered expression of CD226 and CD96 on natural killer cells in patients with pancreatic cancer. Oncotarget 2016, 7, 66586–66594. [Google Scholar] [CrossRef] [PubMed]
- Martinet, L.; Smyth, M.J. Balancing natural killer cell activation through paired receptors. Nat. Rev. Immunol. 2015, 15, 243–254. [Google Scholar] [CrossRef] [PubMed]
- Georgiev, H.; Ravens, I.; Papadogianni, G.; Bernhardt, G. Coming of Age: CD96 Emerges as Modulator of Immune Responses. Front. Immunol. 2018, 9, 1072. [Google Scholar] [CrossRef]
- Chambers, C.A. The expanding world of co-stimulation: The two-signal model revisited. Trends Immunol. 2001, 22, 217–223. [Google Scholar] [CrossRef]
- Fuchs, A.; Cella, M.; Giurisato, E.; Shaw, A.S.; Colonna, M. Cutting edge: CD96 (tactile) promotes NK cell-target cell adhesion by interacting with the poliovirus receptor (CD155). J. Immunol. 2004, 172, 3994–3998. [Google Scholar] [CrossRef] [PubMed]
- Blake, S.J.; Dougall, W.C.; Miles, J.J.; Teng, M.W.; Smyth, M.J. Molecular Pathways: Targeting CD96 and TIGIT for Cancer Immunotherapy. Clin. Cancer Res. 2016, 22, 5183–5188. [Google Scholar] [CrossRef]
- Roman Aguilera, A.; Lutzky, V.P.; Mittal, D.; Li, X.Y.; Stannard, K.; Takeda, K.; Bernhardt, G.; Teng, M.W.L.; Dougall, W.C.; Smyth, M.J. CD96 targeted antibodies need not block CD96-CD155 interactions to promote NK cell anti-metastatic activity. Oncoimmunology 2018, 7, e1424677. [Google Scholar] [CrossRef]
- Hanamatsu, Y.; Saigo, C.; Kito, Y.; Takeuchi, T. An obstructive role of NK cells on metastatic growth of clear-cell sarcoma cells in a xenoplant murine model. Mol. Clin. Oncol. 2021, 14, 9. [Google Scholar] [CrossRef]
- Zhu, Y.; Paniccia, A.; Schulick, A.C.; Chen, W.; Koenig, M.R.; Byers, J.T.; Yao, S.; Bevers, S.; Edil, B.H. Identification of CD112R as a novel checkpoint for human T cells. J. Exp. Med. 2016, 213, 167–176. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, Y.; Cao, G.; Zheng, X.; Sun, C.; Wei, H.; Tian, Z.; Xiao, W.; Sun, R.; Sun, H. Blockade of checkpoint receptor PVRIG unleashes anti-tumor immunity of NK cells in murine and human solid tumors. J. Hematol. Oncol. 2021, 14, 100. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; He, Y.; He, W.; Wu, G.; Zhou, X.; Sheng, Q.; Zhong, W.; Lu, Y.; Ding, Y.; Lu, Q.; et al. Exhausted CD8+T Cells in the Tumor Immune Microenvironment: New Pathways to Therapy. Front. Immunol. 2020, 11, 622509. [Google Scholar] [CrossRef]
- Sharpe, A.H.; Pauken, K.E. The diverse functions of the PD1 inhibitory pathway. Nat. Rev. Immunol. 2018, 18, 153–167. [Google Scholar] [CrossRef] [PubMed]
- Fife, B.T.; Bluestone, J.A. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Immunol. Rev. 2008, 224, 166–182. [Google Scholar] [CrossRef]
- Hsu, J.; Hodgins, J.J.; Marathe, M.; Nicolai, C.J.; Bourgeois-Daigneault, M.C.; Trevino, T.N.; Azimi, C.S.; Scheer, A.K.; Randolph, H.E.; Thompson, T.W.; et al. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J. Clin. Invest. 2018, 128, 4654–4668. [Google Scholar] [CrossRef] [PubMed]
- Judge, S.J.; Dunai, C.; Aguilar, E.G.; Vick, S.C.; Sturgill, I.R.; Khuat, L.T.; Stoffel, K.M.; Van Dyke, J.; Longo, D.L.; Darrow, M.A.; et al. Minimal PD-1 expression in mouse and human NK cells under diverse conditions. J. Clin. Investig. 2020, 130, 3051–3068. [Google Scholar] [CrossRef] [PubMed]
- Quatrini, L.; Wieduwild, E.; Escaliere, B.; Filtjens, J.; Chasson, L.; Laprie, C.; Vivier, E.; Ugolini, S. Endogenous glucocorticoids control host resistance to viral infection through the tissue-specific regulation of PD-1 expression on NK cells. Nat. Immunol. 2018, 19, 954–962. [Google Scholar] [CrossRef]
- Pesce, S.; Greppi, M.; Tabellini, G.; Rampinelli, F.; Parolini, S.; Olive, D.; Moretta, L.; Moretta, A.; Marcenaro, E. Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization. J. Allergy Clin. Immunol. 2017, 139, 335–346. [Google Scholar] [CrossRef]
- Beldi-Ferchiou, A.; Lambert, M.; Dogniaux, S.; Vély, F.; Vivier, E.; Olive, D.; Dupuy, S.; Levasseur, F.; Zucman, D.; Lebbé, C.; et al. PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma. Oncotarget 2016, 7, 72961–72977. [Google Scholar] [CrossRef]
- MacFarlane, A.W.; Jillab, M.; Plimack, E.R.; Hudes, G.R.; Uzzo, R.G.; Litwin, S.; Dulaimi, E.; Al-Saleem, T.; Campbell, K.S. PD-1 expression on peripheral blood cells increases with stage in renal cell carcinoma patients and is rapidly reduced after surgical tumor resection. Cancer Immunol. Res. 2014, 2, 320–331. [Google Scholar] [CrossRef] [PubMed]
- Vari, F.; Arpon, D.; Keane, C.; Hertzberg, M.S.; Talaulikar, D.; Jain, S.; Cui, Q.; Han, E.; Tobin, J.; Bird, R.; et al. Immune evasion via PD-1/PD-L1 on NK cells and monocyte/macrophages is more prominent in Hodgkin lymphoma than DLBCL. Blood 2018, 131, 1809–1819. [Google Scholar] [CrossRef]
- Benson, D.M., Jr.; Bakan, C.E.; Mishra, A.; Hofmeister, C.C.; Efebera, Y.; Becknell, B.; Baiocchi, R.A.; Zhang, J.; Yu, J.; Smith, M.K.; et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: A therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood 2010, 116, 2286–2294. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Cheng, Y.; Xu, Y.; Wang, Z.; Du, X.; Li, C.; Peng, J.; Gao, L.; Liang, X.; Ma, C. Increased expression of programmed cell death protein 1 on NK cells inhibits NK-cell-mediated anti-tumor function and indicates poor prognosis in digestive cancers. Oncogene 2017, 36, 6143–6153. [Google Scholar] [CrossRef] [PubMed]
- Quatrini, L.; Vacca, P.; Tumino, N.; Besi, F.; Di Pace, A.L.; Scordamaglia, F.; Martini, S.; Munari, E.; Mingari, M.C.; Ugolini, S.; et al. Glucocorticoids and the cytokines IL-12, IL-15, and IL-18 present in the tumor microenvironment induce PD-1 expression on human natural killer cells. J. Allergy Clin. Immunol. 2021, 147, 349–360. [Google Scholar] [CrossRef] [PubMed]
- Mariotti, F.R.; Petrini, S.; Ingegnere, T.; Tumino, N.; Besi, F.; Scordamaglia, F.; Munari, E.; Pesce, S.; Marcenaro, E.; Moretta, A.; et al. PD-1 in human NK cells: Evidence of cytoplasmic mRNA and protein expression. Oncoimmunology 2019, 8, 1557030. [Google Scholar] [CrossRef] [PubMed]
- Hasim, N.A.; Amin, L.; Mahadi, Z.; Yusof, N.A.M.; Ngah, A.C.; Yaacob, M.; Olesen, A.P.; Aziz, A.A. The Integration and Harmonisation of Secular and Islamic Ethical Principles in Formulating Acceptable Ethical Guidelines for Modern Biotechnology in Malaysia. Sci. Eng. Ethics 2020, 26, 1797–1825. [Google Scholar] [CrossRef]
- Wherry, E.J.; Ha, S.J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef]
- Della Chiesa, M.; Pesce, S.; Muccio, L.; Carlomagno, S.; Sivori, S.; Moretta, A.; Marcenaro, E. Features of Memory-Like and PD-1(+) Human NK Cell Subsets. Front. Immunol. 2016, 7, 351. [Google Scholar] [CrossRef] [PubMed]
- Tumino, N.; Martini, S.; Munari, E.; Scordamaglia, F.; Besi, F.; Mariotti, F.R.; Bogina, G.; Mingari, M.C.; Vacca, P.; Moretta, L. Presence of innate lymphoid cells in pleural effusions of primary and metastatic tumors: Functional analysis and expression of PD-1 receptor. Int. J. Cancer 2019, 145, 1660–1668. [Google Scholar] [CrossRef]
- Wiesmayr, S.; Webber, S.A.; Macedo, C.; Popescu, I.; Smith, L.; Luce, J.; Metes, D. Decreased NKp46 and NKG2D and elevated PD-1 are associated with altered NK-cell function in pediatric transplant patients with PTLD. Eur. J. Immunol. 2012, 42, 541–550. [Google Scholar] [CrossRef]
- Huang, B.Y.; Zhan, Y.P.; Zong, W.J.; Yu, C.J.; Li, J.F.; Qu, Y.M.; Han, S. The PD-1/B7-H1 pathway modulates the natural killer cells versus mouse glioma stem cells. PLoS ONE 2015, 10, e0134715. [Google Scholar] [CrossRef] [PubMed]
- Rowshanravan, B.; Halliday, N.; Sansom, D.M. CTLA-4: A moving target in immunotherapy. Blood 2018, 131, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Stojanovic, A.; Fiegler, N.; Brunner-Weinzierl, M.; Cerwenka, A. CTLA-4 Is Expressed by Activated Mouse NK Cells and Inhibits NK Cell IFN-γ Production in Response to Mature Dendritic Cells. J. Immunol. 2014, 192, 4184–4191. [Google Scholar] [CrossRef]
- Lougaris, V.; Lanzi, G.; Baronio, M.; Gazzurelli, L.; Vairo, D.; Lorenzini, T.; Badolato, R.; Notarangelo, L.D.; Boschi, A.; Moratto, D.; et al. Progressive severe B cell and NK cell deficiency with T cell senescence in adult CD40L deficiency. Clin. Immunol. 2018, 190, 11–14. [Google Scholar] [CrossRef]
- Lang, S.; Vujanovic, N.L.; Wollenberg, B.; Whiteside, T.L. Absence of B7.1-CD28/CTLA-4-mediated co-stimulation in human NK cells. Eur. J. Immunol. 1998, 28, 780–786. [Google Scholar] [CrossRef]
- Davis-Marcisak, E.F.; Fitzgerald, A.A.; Kessler, M.D.; Danilova, L.; Jaffee, E.M.; Zaidi, N.; Weiner, L.M.; Fertig, E.J. Transfer learning between preclinical models and human tumors identifies conserved NK cell activation signature in anti-CTLA-4 responsive tumors. bioRxiv 2021. [Google Scholar] [CrossRef]
- Caudana, P.; Nunez, N.G.; De La Rochere, P.; Pinto, A.; Denizeau, J.; Alonso, R.; Niborski, L.L.; Lantz, O.; Sedlik, C.; Piaggio, E. IL2/Anti-IL2 Complex Combined with CTLA-4, But Not PD-1, Blockade Rescues Antitumor NK Cell Function by Regulatory T-cell Modulation. Cancer Immunol. Res. 2019, 7, 443–457. [Google Scholar] [CrossRef]
- Pires Da Silva, I.E.D.; Gallois, A.; Lui, K.P.; Shapiro, R.L.; Pavlick, A.C.; Bhardwaj, N.; Osman, I. The effect of ipilimumab on natural killer cells identifies the subset of advanced melanoma patients with clinical response. J. Clin. Oncol. 2015, 33, 9065. [Google Scholar] [CrossRef]
- Tallerico, R.; Cristiani, C.M.; Staaf, E.; Garofalo, C.; Sottile, R.; Capone, M.; Pico de Coaña, Y.; Madonna, G.; Palella, E.; Wolodarski, M.; et al. IL-15, TIM-3 and NK cells subsets predict responsiveness to anti-CTLA-4 treatment in melanoma patients. OncoImmunology 2017, 6, e1261242. [Google Scholar] [CrossRef]
- Romano, E.; Kusio-Kobialka, M.; Foukas, P.G.; Baumgaertner, P.; Meyer, C.; Ballabeni, P.; Michielin, O.; Weide, B.; Romero, P.; Speiser, D.E. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc. Natl. Acad Sci. USA 2015, 112, 6140–6145. [Google Scholar] [CrossRef] [PubMed]
- Krummel, M.F.; Allison, J.P. CTLA-4 engagement inhibits IL-2 accumulation and cell cycle progression upon activation of resting T cells. J. Exp. Med. 1996, 183, 2533–2540. [Google Scholar] [CrossRef]
- Kerdiles, Y.; Ugolini, S.; Vivier, E. T cell regulation of natural killer cells. J. Exp. Med. 2013, 210, 1065–1068. [Google Scholar] [CrossRef] [PubMed]
- Hannani, D.; Vétizou, M.; Enot, D.; Rusakiewicz, S.; Chaput, N.; Klatzmann, D.; Desbois, M.; Jacquelot, N.; Vimond, N.; Chouaib, S.; et al. Anticancer immunotherapy by CTLA-4 blockade: Obligatory contribution of IL-2 receptors and negative prognostic impact of soluble CD25. Cell Res. 2015, 25, 208–224. [Google Scholar] [CrossRef] [PubMed]
- Ghiringhelli, F.; Ménard, C.; Martin, F.; Zitvogel, L. The role of regulatory T cells in the control of natural killer cells: Relevance during tumor progression. Immunol. Rev. 2006, 214, 229–238. [Google Scholar] [CrossRef]
- Sottile, R.; Tannazi, M.; Johansson, M.H.; Cristiani, C.M.; Calabro, L.; Ventura, V.; Cutaia, O.; Chiarucci, C.; Covre, A.; Garofalo, C.; et al. NK- and T-cell subsets in malignant mesothelioma patients: Baseline pattern and changes in the context of anti-CTLA-4 therapy. Int. J. Cancer 2019, 145, 2238–2248. [Google Scholar] [CrossRef]
- Khan, S.; Burt, D.J.; Ralph, C.; Thistlethwaite, F.C.; Hawkins, R.E.; Elkord, E. Tremelimumab (anti-CTLA4) mediates immune responses mainly by direct activation of T effector cells rather than by affecting T regulatory cells. Clin. Immunol. 2011, 138, 85–96. [Google Scholar] [CrossRef]
- Watanabe, N.; Gavrieli, M.; Sedy, J.R.; Yang, J.; Fallarino, F.; Loftin, S.K.; Hurchla, M.A.; Zimmerman, N.; Sim, J.; Zang, X.; et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat. Immunol. 2003, 4, 670–679. [Google Scholar] [CrossRef] [PubMed]
- Cai, G.; Freeman, G.J. The CD160, BTLA, LIGHT/HVEM pathway: A bidirectional switch regulating T-cell activation. Immunol. Rev. 2009, 229, 244–258. [Google Scholar] [CrossRef]
- Šedý, J.R.; Bjordahl, R.L.; Bekiaris, V.; Macauley, M.G.; Ware, B.C.; Norris, P.S.; Lurain, N.S.; Benedict, C.A.; Ware, C.F. CD160 Activation by Herpesvirus Entry Mediator Augments Inflammatory Cytokine Production and Cytolytic Function by NK Cells. J. Immunol. 2013, 191, 828–836. [Google Scholar] [CrossRef]
- Sordo-Bahamonde, C.; Lorenzo-Herrero, S.; Gonzalez-Rodriguez, A.P.R.; Payer, Á.; González-García, E.; López-Soto, A.; Gonzalez, S. BTLA/HVEM Axis Induces NK Cell Immunosuppression and Poor Outcome in Chronic Lymphocytic Leukemia. Cancers 2021, 13, 1766. [Google Scholar] [CrossRef] [PubMed]
- Chapoval, A.I.; Ni, J.; Lau, J.S.; Wilcox, R.A.; Flies, D.B.; Liu, D.; Dong, H.; Sica, G.L.; Zhu, G.; Tamada, K.; et al. B7-H3: A costimulatory molecule for T cell activation and IFN-γ production. Nat. Immunol. 2001, 2, 269–274. [Google Scholar] [CrossRef]
- Zhang, G.; Wang, J.; Kelly, J.; Gu, G.; Hou, J.; Zhou, Y.; Redmond, H.P.; Wang, J.H.; Zhang, X. B7-H3 augments the inflammatory response and is associated with human sepsis. J. Immunol. 2010, 185, 3677–3684. [Google Scholar] [CrossRef]
- Hashiguchi, M.; Kobori, H.; Ritprajak, P.; Kamimura, Y.; Kozono, H.; Azuma, M. Triggering receptor expressed on myeloid cell-like transcript 2 (TLT-2) is a counter-receptor for B7-H3 and enhances T cell responses. Proc. Natl. Acad Sci. USA 2008, 105, 10495–10500. [Google Scholar] [CrossRef] [PubMed]
- Leitner, J.; Klauser, C.; Pickl, W.F.; Stöckl, J.; Majdic, O.; Bardet, A.F.; Kreil, D.P.; Dong, C.; Yamazaki, T.; Zlabinger, G.; et al. B7-H3 is a potent inhibitor of human T-cell activation: No evidence for B7-H3 and TREML2 interaction. Eur. J. Immunol. 2009, 39, 1754–1764. [Google Scholar] [CrossRef] [PubMed]
- Vigdorovich, V.; Ramagopal, U.A.; Lázár-Molnár, E.; Sylvestre, E.; Lee, J.S.; Hofmeyer, K.A.; Zang, X.; Nathenson, S.G.; Almo, S.C. Structure and T Cell Inhibition Properties of B7 Family Member, B7-H3. Structure 2013, 21, 707–717. [Google Scholar] [CrossRef]
- Husain, B.; Ramani, S.R.; Chiang, E.; Lehoux, I.; Paduchuri, S.; Arena, T.A.; Patel, A.; Wilson, B.; Chan, P.; Franke, Y.; et al. A Platform for Extracellular Interactome Discovery Identifies Novel Functional Binding Partners for the Immune Receptors B7-H3/CD276 and PVR/CD155. Mol. Cell. Proteom. 2019, 18, 2310–2323. [Google Scholar] [CrossRef]
- Miyazaki, A.; Hanafusa, T.; Yamada, K.; Miyagawa, J.; Fujino-Kurihara, H.; Nakajima, H.; Nonaka, K.; Tarui, S. Predominance of T lymphocytes in pancreatic islets and spleen of pre-diabetic non-obese diabetic (NOD) mice: A longitudinal study. Clin. Exp. Immunol. 1985, 60, 622–630. [Google Scholar]
- Cocco, C.; Morandi, F.; Airoldi, I. Immune Checkpoints in Pediatric Solid Tumors: Targetable Pathways for Advanced Therapeutic Purposes. Cells 2021, 10, 927. [Google Scholar] [CrossRef] [PubMed]
- Steinberger, P.; Majdic, O.; Derdak, S.V.; Pfistershammer, K.; Kirchberger, S.; Klauser, C.; Zlabinger, G.; Pickl, W.F.; Stöckl, J.; Knapp, W. Molecular Characterization of Human 4Ig-B7-H3, a Member of the B7 Family with Four Ig-Like Domains. J. Immunol. 2004, 172, 2352–2359. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.-h.; Martin-Orozco, N.; Zheng, P.; Li, J.; Zhang, P.; Tan, H.; Park, H.J.; Jeong, M.; Chang, S.H.; Kim, B.-S.; et al. Inhibition of the B7-H3 immune checkpoint limits tumor growth by enhancing cytotoxic lymphocyte function. Cell Res. 2017, 27, 1034–1045. [Google Scholar] [CrossRef] [PubMed]
- Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014, 74, 1924–1932. [Google Scholar] [CrossRef]
- Flies, D.B.; Wang, S.; Xu, H.; Chen, L. Cutting Edge: A Monoclonal Antibody Specific for the Programmed Death-1 Homolog Prevents Graft-versus-Host Disease in Mouse Models. J. Immunol. 2011, 187, 1537–1541. [Google Scholar] [CrossRef]
- Wang, L.; Rubinstein, R.; Lines, J.L.; Wasiuk, A.; Ahonen, C.; Guo, Y.; Lu, L.-F.; Gondek, D.; Wang, Y.; Fava, R.A.; et al. VISTA, a novel mouse Ig superfamily ligand that negatively regulates T cell responses. J. Exp. Med. 2011, 208, 577–592. [Google Scholar] [CrossRef]
- Triebel, F.; Jitsukawa, S.; Baixeras, E.; Roman-Roman, S.; Genevee, C.; Viegas-Pequignot, E.; Hercend, T. LAG-3, a novel lymphocyte activation gene closely related to CD4. J. Exp. Med. 1990, 171, 1393–1405. [Google Scholar] [CrossRef]
- Merino, A.; Zhang, B.; Dougherty, P.; Luo, X.; Wang, J.; Blazar, B.R.; Miller, J.S.; Cichocki, F. Chronic stimulation drives human NK cell dysfunction and epigenetic reprograming. J. Clin. Investig. 2019, 129, 3770–3785. [Google Scholar] [CrossRef]
- Narayanan, S.; Ahl, P.J.; Bijin, V.A.; Kaliaperumal, N.; Lim, S.G.; Wang, C.-I.; Fairhurst, A.-M.; Connolly, J.E. LAG3 is a Central Regulator of NK Cell Cytokine Production. bioRxiv 2020. [Google Scholar] [CrossRef]
- Kritikou, J.S.; Dahlberg, C.I.; Baptista, M.A.; Wagner, A.K.; Banerjee, P.P.; Gwalani, L.A.; Poli, C.; Panda, S.K.; Kärre, K.; Kaech, S.M.; et al. IL-2 in the tumor microenvironment is necessary for Wiskott-Aldrich syndrome protein deficient NK cells to respond to tumors in vivo. Sci. Rep. 2016, 6, 30636. [Google Scholar] [CrossRef]
- Miyazaki, T.; Dierich, A.; Benoist, C.; Mathis, D. Independent modes of natural killing distinguished in mice lacking Lag3. Science 1996, 272, 405–408. [Google Scholar] [CrossRef]
- Jin, J.; Ahn, Y.-O.; Kim, T.M.; Keam, B.; Kim, D.-W.; Heo, D.S. The CD56bright CD62L+ NKG2A+ immature cell subset is dominantly expanded in human cytokine-induced memory-like NK cells. bioRxiv 2018. [Google Scholar] [CrossRef]
- Huard, B.; Tournier, M.; Triebel, F. LAG-3 does not define a specific mode of natural killing in human. Immunol. Lett 1998, 61, 109–112. [Google Scholar] [CrossRef]
- Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature 2002, 415, 536–541. [Google Scholar] [CrossRef] [PubMed]
- Liang, D.; Shao, H.; Born, W.K.; O’Brien, R.L.; Kaplan, H.J.; Sun, D. High level expression of A2ARs is required for the enhancing function, but not for the inhibiting function, of γδ T cells in the autoimmune responses of EAU. Plos ONE 2018, 13, e0199601. [Google Scholar] [CrossRef]
- Yu, X.; Zheng, Y.; Mao, R.; Su, Z.; Zhang, J. BTLA/HVEM Signaling: Milestones in Research and Role in Chronic Hepatitis B Virus Infection. Front. Immunol. 2019, 10. [Google Scholar] [CrossRef] [PubMed]
- Llibre, A.; Klenerman, P.; Willberg, C.B. Multi-functional lectin-like transcript-1: A new player in human immune regulation. Immunol. Lett. 2016, 177, 62–69. [Google Scholar] [CrossRef]
- Seth, S.; Maier, M.K.; Qiu, Q.; Ravens, I.; Kremmer, E.; Förster, R.; Bernhardt, G. The murine pan T cell marker CD96 is an adhesion receptor for CD155 and nectin-1. Biochem. Biophys Res. Commun. 2007, 364, 959–965. [Google Scholar] [CrossRef]
- Molgora, M.; Bonavita, E.; Ponzetta, A.; Riva, F.; Barbagallo, M.; Jaillon, S.; Popovic, B.; Bernardini, G.; Magrini, E.; Gianni, F.; et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature 2017, 551, 110–114. [Google Scholar] [CrossRef]
- Molgora, M.; Supino, D.; Mantovani, A.; Garlanda, C. Tuning inflammation and immunity by the negative regulators IL-1R2 and IL-1R8. Immunol. Rev. 2018, 281, 233–247. [Google Scholar] [CrossRef] [PubMed]
- Molgora, M.; Barajon, I.; Mantovani, A.; Garlanda, C. Regulatory Role of IL-1R8 in Immunity and Disease. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
- Dimitrova, M.; Zenarruzabeitia, O.; Borrego, F.; Simhadri, V.R. CD300c is uniquely expressed on CD56bright Natural Killer Cells and differs from CD300a upon ligand recognition. Sci. Rep. 2016, 6, 23942. [Google Scholar] [CrossRef] [PubMed]
- Bachelet, I.; Munitz, A.; Moretta, A.; Moretta, L.; Levi-Schaffer, F. The Inhibitory Receptor IRp60 (CD300a) Is Expressed and Functional on Human Mast Cells. J. Immunol. 2005, 175, 7989–7995. [Google Scholar] [CrossRef]
- Littera, R.; Piredda, G.; Argiolas, D.; Lai, S.; Congeddu, E.; Ragatzu, P.; Melis, M.; Carta, E.; Michittu, M.B.; Valentini, D.; et al. KIR and their HLA Class I ligands: Two more pieces towards completing the puzzle of chronic rejection and graft loss in kidney transplantation. Plos ONE 2017, 12, e0180831. [Google Scholar] [CrossRef]
- Workman, C.J.; Vignali, D.A. The CD4-related molecule, LAG-3 (CD223), regulates the expansion of activated T cells. Eur J. Immunol. 2003, 33, 970–979. [Google Scholar] [CrossRef] [PubMed]
- Kouo, T.; Huang, L.; Pucsek, A.B.; Cao, M.; Solt, S.; Armstrong, T.; Jaffee, E. Galectin-3 Shapes Antitumor Immune Responses by Suppressing CD8+ T Cells via LAG-3 and Inhibiting Expansion of Plasmacytoid Dendritic Cells. Cancer Immunol. Res. 2015, 3, 412–423. [Google Scholar] [CrossRef]
- Wang, J.; Sanmamed, M.F.; Datar, I.; Su, T.T.; Ji, L.; Sun, J.; Chen, L.; Chen, Y.; Zhu, G.; Yin, W.; et al. Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell 2019, 176, 334–347.e312. [Google Scholar] [CrossRef]
- Xu, F.; Liu, J.; Liu, D.; Liu, B.; Wang, M.; Hu, Z.; Du, X.; Tang, L.; He, F. LSECtin expressed on melanoma cells promotes tumor progression by inhibiting antitumor T-cell responses. Cancer Res. 2014, 74, 3418–3428. [Google Scholar] [CrossRef]
- Lee, N.; Llano, M.; Carretero, M.; Ishitani, A.; Navarro, F.; López-Botet, M.; Geraghty, D.E. HLA-E is a major ligand for the natural killer inhibitory receptor CD94/NKG2A. Proc. Natl. Acad Sci. USA 1998, 95, 5199–5204. [Google Scholar] [CrossRef]
- Kawasaki, Y.; Ito, A.; Withers, D.A.; Taima, T.; Kakoi, N.; Saito, S.; Arai, Y. Ganglioside DSGb5, preferred ligand for Siglec-7, inhibits NK cell cytotoxicity against renal cell carcinoma cells. Glycobiology 2010, 20, 1373–1379. [Google Scholar] [CrossRef]
- Nicoll, G.; Avril, T.; Lock, K.; Furukawa, K.; Bovin, N.; Crocker, P.R. Ganglioside GD3 expression on target cells can modulate NK cell cytotoxicity via siglec-7-dependent and -independent mechanisms. Eur J. Immunol. 2003, 33, 1642–1648. [Google Scholar] [CrossRef] [PubMed]
- Shao, J.Y.; Yin, W.W.; Zhang, Q.F.; Liu, Q.; Peng, M.L.; Hu, H.D.; Hu, P.; Ren, H.; Zhang, D.Z. Siglec-7 Defines a Highly Functional Natural Killer Cell Subset and Inhibits Cell-Mediated Activities. Scand. J. Immunol. 2016, 84, 182–190. [Google Scholar] [CrossRef] [PubMed]
- Belisle, J.A.; Horibata, S.; Jennifer, G.A.; Petrie, S.; Kapur, A.; André, S.; Gabius, H.J.; Rancourt, C.; Connor, J.; Paulson, J.C.; et al. Identification of Siglec-9 as the receptor for MUC16 on human NK cells, B cells, and monocytes. Mol. Cancer 2010, 9, 118. [Google Scholar] [CrossRef]
- Zhao, D.; Jiang, X.; Xu, Y.; Yang, H.; Gao, D.; Li, X.; Gao, L.; Ma, C.; Liang, X. Decreased Siglec-9 Expression on Natural Killer Cell Subset Associated With Persistent HBV Replication. Front. Immunol. 2018, 9, 1124. [Google Scholar] [CrossRef]
- Huang, Y.H.; Zhu, C.; Kondo, Y.; Anderson, A.C.; Gandhi, A.; Russell, A.; Dougan, S.K.; Petersen, B.S.; Melum, E.; Pertel, T.; et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 2015, 517, 386–390. [Google Scholar] [CrossRef] [PubMed]
- Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid–mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
- Johnston, R.J.; Su, L.J.; Pinckney, J.; Critton, D.; Boyer, E.; Krishnakumar, A.; Corbett, M.; Rankin, A.L.; Dibella, R.; Campbell, L.; et al. VISTA is an acidic pH-selective ligand for PSGL-1. Nature 2019, 574, 565–570. [Google Scholar] [CrossRef]
- Laszik, Z.; Jansen, P.J.; Cummings, R.D.; Tedder, T.F.; McEver, R.P.; Moore, K.L. P-selectin glycoprotein ligand-1 is broadly expressed in cells of myeloid, lymphoid, and dendritic lineage and in some nonhematopoietic cells. Blood 1996, 88, 3010–3021. [Google Scholar] [CrossRef]
- Wang, J.; Wu, G.; Manick, B.; Hernandez, V.; Renelt, M.; Erickson, C.; Guan, J.; Singh, R.; Rollins, S.; Solorz, A.; et al. VSIG-3 as a ligand of VISTA inhibits human T-cell function. Immunology 2019, 156, 74–85. [Google Scholar] [CrossRef]
- Ndhlovu, L.C.; Lopez-Vergès, S.; Barbour, J.D.; Jones, R.B.; Jha, A.R.; Long, B.R.; Schoeffler, E.C.; Fujita, T.; Nixon, D.F.; Lanier, L.L. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood 2012, 119, 3734–3743. [Google Scholar] [CrossRef]
- Gleason, M.K.; Lenvik, T.R.; McCullar, V.; Felices, M.; O’Brien, M.S.; Cooley, S.A.; Verneris, M.R.; Cichocki, F.; Holman, C.J.; Panoskaltsis-Mortari, A.; et al. Tim-3 is an inducible human natural killer cell receptor that enhances interferon gamma production in response to galectin-9. Blood 2012, 119, 3064–3072. [Google Scholar] [CrossRef]
- Xu, L.; Huang, Y.; Tan, L.; Yu, W.; Chen, D.; Lu, C.; He, J.; Wu, G.; Liu, X.; Zhang, Y. Increased Tim-3 expression in peripheral NK cells predicts a poorer prognosis and Tim-3 blockade improves NK cell-mediated cytotoxicity in human lung adenocarcinoma. Int. Immunopharmacol 2015, 29, 635–641. [Google Scholar] [CrossRef] [PubMed]
- da Silva, I.P.; Gallois, A.; Jimenez-Baranda, S.; Khan, S.; Anderson, A.C.; Kuchroo, V.K.; Osman, I.; Bhardwaj, N. Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol. Res. 2014, 2, 410–422. [Google Scholar] [CrossRef]
- Wang, Z.; Zhu, J.; Gu, H.; Yuan, Y.; Zhang, B.; Zhu, D.; Zhou, J.; Zhu, Y.; Chen, W. The Clinical Significance of Abnormal Tim-3 Expression on NK Cells from Patients with Gastric Cancer. Immunol. Invest. 2015, 44, 578–589. [Google Scholar] [CrossRef] [PubMed]
- Farkas, A.M.; Audenet, F.; Anastos, H.; Galsky, M.; Sfakianos, J.; Bhardwaj, N. Tim-3 and TIGIT mark Natural Killer cells susceptible to effector dysfunction in human bladder cancer. J. Immunol. 2018. [Google Scholar] [CrossRef]
- Xu, L.Y.; Chen, D.D.; He, J.Y.; Lu, C.C.; Liu, X.G.; Le, H.B.; Wang, C.Y.; Zhang, Y.K. Tim-3 expression by peripheral natural killer cells and natural killer T cells increases in patients with lung cancer--reduction after surgical resection. Asian Pac. J. Cancer Prev. 2014, 15, 9945–9948. [Google Scholar] [CrossRef][Green Version]
- Wolf, Y.; Anderson, A.C.; Kuchroo, V.K. TIM3 comes of age as an inhibitory receptor. Nat. Rev. Immunol. 2020, 20, 173–185. [Google Scholar] [CrossRef] [PubMed]
- So, E.C.; Khaladj-Ghom, A.; Ji, Y.; Amin, J.; Song, Y.; Burch, E.; Zhou, H.; Sun, H.; Chen, S.; Bentzen, S.; et al. NK cell expression of Tim-3: First impressions matter. Immunobiology 2019, 224, 362–370. [Google Scholar] [CrossRef] [PubMed]
- Golden-Mason, L.; Waasdorp Hurtado, C.E.; Cheng, L.; Rosen, H.R. Hepatitis C viral infection is associated with activated cytolytic natural killer cells expressing high levels of T cell immunoglobulin- and mucin-domain-containing molecule-3. Clin. Immunol. 2015, 158, 114–125. [Google Scholar] [CrossRef]
- Zheng, Y.; Li, Y.; Lian, J.; Yang, H.; Li, F.; Zhao, S.; Qi, Y.; Zhang, Y.; Huang, L. TNF-α-induced Tim-3 expression marks the dysfunction of infiltrating natural killer cells in human esophageal cancer. J. Transl. Med. 2019, 17, 165. [Google Scholar] [CrossRef]
- Young, A.; Mittal, D.; Stagg, J.; Smyth, M.J. Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov. 2014, 4, 879–888. [Google Scholar] [CrossRef] [PubMed]
- Young, A.; Ngiow, S.F.; Gao, Y.; Patch, A.M.; Barkauskas, D.S.; Messaoudene, M.; Lin, G.; Coudert, J.D.; Stannard, K.A.; Zitvogel, L.; et al. A2AR Adenosine Signaling Suppresses Natural Killer Cell Maturation in the Tumor Microenvironment. Cancer Res. 2018, 78, 1003–1016. [Google Scholar] [CrossRef]
- Nakamura, K.; Casey, M.; Oey, H.; Vari, F.; Stagg, J.; Gandhi, M.K.; Smyth, M.J. Targeting an adenosine-mediated "don’t eat me signal" augments anti-lymphoma immunity by anti-CD20 monoclonal antibody. Leukemia 2020, 34, 2708–2721. [Google Scholar] [CrossRef]
- Chambers, A.M.; Wang, J.; Lupo, K.B.; Yu, H.; Atallah Lanman, N.M.; Matosevic, S. Adenosinergic Signaling Alters Natural Killer Cell Functional Responses. Front. Immunol. 2018, 9, 2533. [Google Scholar] [CrossRef]
- Souza-Fonseca-Guimaraes, F.; Huntington, N.D. A new checkpoint for Natural Killer cell activation. Immunol. Cell Biol. 2018, 96, 5–7. [Google Scholar] [CrossRef]
- Campbell, K.S. Mystery Checkpoint Revealed: KIR3DL3 Finally Found a Ligand in HHLA2. Cancer Immunol. Res. 2021, 9, 128. [Google Scholar] [CrossRef] [PubMed]
- Wei, Y.; Ren, X.; Galbo, P.M., Jr.; Moerdler, S.; Wang, H.; Sica, R.A.; Etemad-Gilbertson, B.; Shi, L.; Zhu, L.; Tang, X.; et al. KIR3DL3-HHLA2 is a human immunosuppressive pathway and a therapeutic target. Sci. Immunol. 2021, 6. [Google Scholar] [CrossRef]
- Bhatt, R.S.; Berjis, A.; Konge, J.C.; Mahoney, K.M.; Klee, A.N.; Freeman, S.S.; Chen, C.H.; Jegede, O.A.; Catalano, P.J.; Pignon, J.C.; et al. KIR3DL3 Is an Inhibitory Receptor for HHLA2 that Mediates an Alternative Immunoinhibitory Pathway to PD1. Cancer Immunol. Res. 2021, 9, 156–169. [Google Scholar] [CrossRef] [PubMed]
- Rosen, D.B.; Bettadapura, J.; Alsharifi, M.; Mathew, P.A.; Warren, H.S.; Lanier, L.L. Cutting edge: Lectin-like transcript-1 is a ligand for the inhibitory human NKR-P1A receptor. J. Immunol. 2005, 175, 7796–7799. [Google Scholar] [CrossRef] [PubMed]
- Rosen, D.B.; Cao, W.; Avery, D.T.; Tangye, S.G.; Liu, Y.J.; Houchins, J.P.; Lanier, L.L. Functional consequences of interactions between human NKR-P1A and its ligand LLT1 expressed on activated dendritic cells and B cells. J. Immunol. 2008, 180, 6508–6517. [Google Scholar] [CrossRef]
- Bialoszewska, A.; Malejczyk, J. Biological and Clinical Significance of Human NKRP1A/LLT1 Receptor/Ligand Interactions. Crit Rev. Immunol. 2018, 38, 479–489. [Google Scholar] [CrossRef] [PubMed]
- Aldemir, H.; Prod’homme, V.; Dumaurier, M.J.; Retiere, C.; Poupon, G.; Cazareth, J.; Bihl, F.; Braud, V.M. Cutting edge: Lectin-like transcript 1 is a ligand for the CD161 receptor. J. Immunol. 2005, 175, 7791–7795. [Google Scholar] [CrossRef] [PubMed]
- Roth, P.; Mittelbronn, M.; Wick, W.; Meyermann, R.; Tatagiba, M.; Weller, M. Malignant glioma cells counteract antitumor immune responses through expression of lectin-like transcript-1. Cancer Res. 2007, 67, 3540–3544. [Google Scholar] [CrossRef]
- Marrufo, A.M.; Mathew, S.O.; Chaudhary, P.; Malaer, J.D.; Vishwanatha, J.K.; Mathew, P.A. Blocking LLT1 (CLEC2D, OCIL)-NKRP1A (CD161) interaction enhances natural killer cell-mediated lysis of triple-negative breast cancer cells. Am. J. Cancer Res. 2018, 8, 1050–1063. [Google Scholar] [PubMed]
- Rahim, M.M.; Chen, P.; Mottashed, A.N.; Mahmoud, A.B.; Thomas, M.J.; Zhu, Q.; Brooks, C.G.; Kartsogiannis, V.; Gillespie, M.T.; Carlyle, J.R.; et al. The mouse NKR-P1B:Clr-b recognition system is a negative regulator of innate immune responses. Blood 2015, 125, 2217–2227. [Google Scholar] [CrossRef]
- Tanaka, M.; Fine, J.H.; Kirkham, C.L.; Aguilar, O.A.; Belcheva, A.; Martin, A.; Ketela, T.; Moffat, J.; Allan, D.S.J.; Carlyle, J.R. The Inhibitory NKR-P1B:Clr-b Recognition Axis Facilitates Detection of Oncogenic Transformation and Cancer Immunosurveillance. Cancer Res. 2018, 78, 3589–3603. [Google Scholar] [CrossRef]
- Braud, V.M.; Biton, J.; Becht, E.; Knockaert, S.; Mansuet-Lupo, A.; Cosson, E.; Damotte, D.; Alifano, M.; Validire, P.; Anjuère, F.; et al. Expression of LLT1 and its receptor CD161 in lung cancer is associated with better clinical outcome. Oncoimmunology 2018, 7, e1423184. [Google Scholar] [CrossRef]
- Mathew, P.A.; Chuang, S.S.; Vaidya, S.V.; Kumaresan, P.R.; Boles, K.S.; Pham, H.T. The LLT1 receptor induces IFN-gamma production by human natural killer cells. Mol. Immunol. 2004, 40, 1157–1163. [Google Scholar] [CrossRef]
- Bambard, N.D.; Mathew, S.O.; Mathew, P.A. LLT1-mediated activation of IFN-gamma production in human natural killer cells involves ERK signalling pathway. Scand. J. Immunol. 2010, 71, 210–219. [Google Scholar] [CrossRef]
- Daly, J.; Carlsten, M.; O’Dwyer, M. Sugar Free: Novel Immunotherapeutic Approaches Targeting Siglecs and Sialic Acids to Enhance Natural Killer Cell Cytotoxicity Against Cancer. Front. Immunol. 2019, 10, 1047. [Google Scholar] [CrossRef]
- Falco, M.; Biassoni, R.; Bottino, C.; Vitale, M.; Sivori, S.; Augugliaro, R.; Moretta, L.; Moretta, A. Identification and molecular cloning of p75/AIRM1, a novel member of the sialoadhesin family that functions as an inhibitory receptor in human natural killer cells. J. Exp. Med. 1999, 190, 793–802. [Google Scholar] [CrossRef] [PubMed]
- Jandus, C.; Boligan, K.F.; Chijioke, O.; Liu, H.; Dahlhaus, M.; Démoulins, T.; Schneider, C.; Wehrli, M.; Hunger, R.E.; Baerlocher, G.M.; et al. Interactions between Siglec-7/9 receptors and ligands influence NK cell–dependent tumor immunosurveillance. J. Clin. Investig. 2014, 124, 1810–1820. [Google Scholar] [CrossRef] [PubMed]
- Prescher, H.; Frank, M.; Gütgemann, S.; Kuhfeldt, E.; Schweizer, A.; Nitschke, L.; Watzl, C.; Brossmer, R. Design, Synthesis, and Biological Evaluation of Small, High-Affinity Siglec-7 Ligands: Toward Novel Inhibitors of Cancer Immune Evasion. J. Med. Chem 2017, 60, 941–956. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, S.; Yoshimura, A.; Yasuda, Y.; Mori, A.; Tanaka, H.; Takahashi, T.; Kitajima, K.; Sato, C. Chemical Synthesis and Evaluation of a Disialic Acid-Containing Dextran Polymer as an Inhibitor for the Interaction between Siglec 7 and Its Ligand. ChemBioChem 2017, 18, 1194–1203. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Sejas, N.; Campos, C.; Hassouneh, F.; Sanchez-Correa, B.; Tarazona, R.; Pera, A.; Solana, R. Effect of CMV and Aging on the Differential Expression of CD300a, CD161, T-bet, and Eomes on NK Cell Subsets. Front. Immunol. 2016, 7. [Google Scholar] [CrossRef] [PubMed]
- Lankry, D.; Simic, H.; Klieger, Y.; Levi-Schaffer, F.; Jonjic, S.; Mandelboim, O. Expression and Function of CD300 in NK Cells. J. Immunol. 2010, 185, 2877–2886. [Google Scholar] [CrossRef]
- Cantoni, C.; Bottino, C.; Augugliaro, R.; Morelli, L.; Marcenaro, E.; Castriconi, R.; Vitale, M.; Pende, D.; Sivori, S.; Millo, R.; et al. Molecular and functional characterization of IRp60, a member of the immunoglobulin superfamily that functions as an inhibitory receptor in human NK cells. Eur J. Immunol. 1999, 29, 3148–3159. [Google Scholar] [CrossRef]
- Lankry, D.; Rovis, T.L.; Jonjic, S.; Mandelboim, O. The interaction between CD300a and phosphatidylserine inhibits tumor cell killing by NK cells. Eur J. Immunol. 2013, 43, 2151–2161. [Google Scholar] [CrossRef] [PubMed]
- Vitallé, J.; Terrén, I.; Orrantia, A.; Pérez-Garay, R.; Vidal, F.; Iribarren, J.A.; Rodríguez, C.; Lirola, A.M.L.; Bernal, E.; Zenarruzabeitia, O.; et al. CD300a inhibits CD16-mediated NK cell effector functions in HIV-1-infected patients. Cell Mol. Immunol. 2019, 16, 940–942. [Google Scholar] [CrossRef]
- Sen, D.R.; Kaminski, J.; Barnitz, R.A.; Kurachi, M.; Gerdemann, U.; Yates, K.B.; Tsao, H.W.; Godec, J.; LaFleur, M.W.; Brown, F.D.; et al. The epigenetic landscape of T cell exhaustion. Science 2016, 354, 1165–1169. [Google Scholar] [CrossRef] [PubMed]
Receptor | Expression | Ligands | Refs |
---|---|---|---|
A2AR | T, NK cells | Adenosine | [136] |
Unknown receptor to B7-H3 | B7-H3 | [124] | |
BTLA | T, B, NK cells, DCs and myeloid cells | HVEM | [112,137] |
CD112R | T, NK cells | CD112 | [72] |
CD161 | T, NK cells | Lectin-like transcript 1 (LLT1, also called CLEC2D or OCIL) | [138] |
CD96 | T, NK, NKT cells | Human: CD155 (PVR, Necl-5) Mouse: CD155 and Nectin-1 | [68,139] |
CTLA-4 | Regulatory T cells (Tregs), activated T and B cells, activated murine NK cells, human NK cells? | CD80 (B7-1), CD86 (B7-2) | [96] |
IL-1R8 | Monocytes, B, T, NK cells, DCs | IL-37 | [140,141,142] |
Irp60 | Mast cells, NK cells | Phosphatidylethanolamine (PE) and phosphatidylserine (PtdSer) | [143,144] |
KIRs | NK cells, CD8+ T cells | HLA-ABC | [145] |
LAG-3 | Regulatory T cells (Tregs), activated T and NK cells, B cells, NKT cells, pDCs | MHC II; Galectin-3 (Gal-3); LSECtin fibrinogen-like protein 1 (FGL1); | [146,147,148,149] |
NKG2A | NK cells, CD8+ T cells | HLA-E | [150] |
PD-1 | Activated T and B cells, NK cells, NKT cells, ILC-2 cells, myeloid cells | PD-L1 (B7-H1); PD-L2 (B7-DC) | [78] |
Siglec-7 | Monocytes, CD8+ T and NK cells | DSGb5, GD3 | [151,152,153] |
Siglec-9 | T, B, NK cells (CD56dim), granulocytes and monocytes | MUC16 | [154,155] |
TIGIT | T, NK, NKT cells | CD155, CD112, CD113 | [58,61] |
TIM-3 | T, NK, NKT cells and myeloid cells | Galectin-9 (Gal-9) Phosphatidylserine (PtdSer); High mobility group protein B1- HMGB1; Carcinoembryonic antigen cell adhesion molecule 1 CEACAM-1 | [156,157] |
VISTA | Myeloid cells, TCRγδ T cells, naïve CD4+ and CD8+ TCRαβ T cells, Tregs | PSGL-1, VSIG3 | [158,159,160] |
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
Buckle, I.; Guillerey, C. Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer. Cancers 2021, 13, 4263. https://doi.org/10.3390/cancers13174263
Buckle I, Guillerey C. Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer. Cancers. 2021; 13(17):4263. https://doi.org/10.3390/cancers13174263
Chicago/Turabian StyleBuckle, Irina, and Camille Guillerey. 2021. "Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer" Cancers 13, no. 17: 4263. https://doi.org/10.3390/cancers13174263
APA StyleBuckle, I., & Guillerey, C. (2021). Inhibitory Receptors and Immune Checkpoints Regulating Natural Killer Cell Responses to Cancer. Cancers, 13(17), 4263. https://doi.org/10.3390/cancers13174263