Control of NK Cell Activation by Immune Checkpoint Molecules
Abstract
:1. Introduction
2. Programmed Cell Death-1
3. Cytotoxic T Lymphocyte-Associated Antigen 4 (CTLA-4)
4. T-Cell Immunoglobulin and Mucin Domain 3 (Tim-3)
5. T Cell Immunoglobulin and ITIM Domain (TIGIT)
6. Lymphocyte Activation Gene-3 (LAG-3)
7. Killer Cell Immunoglobulin-Like Receptors (KIRs)
8. Natural Killer Cell Receptor Group 2 Member A (NKG2A)
9. Concluding Remarks
Acknowledgments
Author Contributions
Conflicts of Interest
References
- 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]
- Lodoen, M.B.; Lanier, L.L. Natural killer cells as an initial defense against pathogens. Curr. Opin. Immunol. 2006, 18, 391–398. [Google Scholar] [CrossRef] [PubMed]
- Cerwenka, A.; Lanier, L.L. Natural killer cells, viruses and cancer. Nat. Rev. Immunol. 2001, 1, 41–49. [Google Scholar] [CrossRef] [PubMed]
- Sivori, S.; Carlomagno, S.; Pesce, S.; Moretta, A.; Vitale, M.; Marcenaro, E. TLR/NCR/KIR: Which one to use and when? Front. Immunol. 2014, 5. [Google Scholar] [CrossRef] [PubMed]
- Raulet, D.H.; Guerra, N. Oncogenic stress sensed by the immune system: Role of natural killer cell receptors. Nat. Rev. Immunol. 2009, 9, 568–580. [Google Scholar] [CrossRef] [PubMed]
- Del Zotto, G.; Marcenaro, E.; Vacca, P.; Sivori, S.; Pende, D.; Chiesa, M.D.; Moretta, F.; Ingegnere, T.; Mingari, M.C.; Moretta, A.; et al. Markers and function of human NK cells in normal and pathological conditions. Cytom. Part B Clin. Cytom. 2017, 92, 100–114. [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] [PubMed]
- Orr, M.T.; Lanier, L.L. Natural killer cell education and tolerance. Cell 2010, 142, 847–856. [Google Scholar] [CrossRef] [PubMed]
- Narni-Mancinelli, E.; Ugolini, S.; Vivier, E. Tuning the threshold of natural killer cell responses. Curr. Opin. Immunol. 2013, 25, 53–58. [Google Scholar] [CrossRef] [PubMed]
- Raulet, D.H.; Vance, R.E. Self-tolerance of natural killer cells. Nat. Rev. Immunol. 2006, 6, 520–531. [Google Scholar] [CrossRef] [PubMed]
- Raulet, D.H. Missing self recognition and self tolerance of natural killer (NK) cells. Semin. Immunol. 2006, 18, 145–150. [Google Scholar] [CrossRef] [PubMed]
- Gasser, S.; Raulet, D.H. Activation and self-tolerance of natural killer cells. Immunol. Rev. 2006, 214, 130–142. [Google Scholar] [CrossRef] [PubMed]
- Brodin, P.; Karre, K.; Hoglund, P. NK cell education: Not an on-off switch but a tunable rheostat. Trends Immunol. 2009, 30, 143–149. [Google Scholar] [CrossRef] [PubMed]
- Yokoyama, W.M.; Kim, S. How do natural killer cells find self to achieve tolerance? Immunity 2006, 24, 249–257. [Google Scholar] [CrossRef] [PubMed]
- Brusilovsky, M.; Rosental, B.; Shemesh, A.; Appel, M.Y.; Porgador, A. Human NK cell recognition of target cells in the prism of natural cytotoxicity receptors and their ligands. J. Immunotoxicol. 2012, 9, 267–274. [Google Scholar] [CrossRef] [PubMed]
- Moretta, A.; Bottino, C.; Vitale, M.; Pende, D.; Cantoni, C.; Mingari, M.C.; Biassoni, R.; Moretta, L. Activating receptors and coreceptors involved in human natural killer cell-mediated cytolysis. Annu. Rev. Immunol. 2001, 19, 197–223. [Google Scholar] [CrossRef] [PubMed]
- Fauriat, C.; Long, E.O.; Ljunggren, H.G.; Bryceson, Y.T. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood 2010, 115, 2167–2176. [Google Scholar] [CrossRef] [PubMed]
- De Maria, A.; Bozzano, F.; Cantoni, C.; Moretta, L. Revisiting human natural killer cell subset function revealed cytolytic CD56(dim)CD16+ NK cells as rapid producers of abundant Ifn-gamma on activation. Proc. Natl. Acad. Sci. USA 2011, 108, 728–732. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Deniz, G.; Erten, G.; Kucuksezer, U.C.; Kocacik, D.; Karagiannidis, C.; Aktas, E.; Akdis, C.A.; Akdis, M. Regulatory NK cells suppress antigen-specific T cell responses. J. Immunol. 2008, 180, 850–857. [Google Scholar] [CrossRef] [PubMed]
- Morandi, B.; Mortara, L.; Carrega, P.; Cantoni, C.; Costa, G.; Accolla, R.S.; Mingari, M.C.; Ferrini, S.; Moretta, L.; Ferlazzo, G. NK cells provide helper signal for CD8+ T cells by inducing the expression of membrane-bound IL-15 on DCS. Int. Immunol. 2009, 21, 599–606. [Google Scholar] [CrossRef] [PubMed]
- Vitale, M.; Della Chiesa, M.; Carlomagno, S.; Pende, D.; Arico, M.; Moretta, L.; Moretta, A. NK-dependent DC maturation is mediated by TNF-α and IFN-γ released upon engagement of the NKP30 triggering receptor. Blood 2005, 106, 566–571. [Google Scholar] [CrossRef] [PubMed]
- Awasthi, A.; Ayello, J.; van de Ven, C.; Elmacken, M.; Sabulski, A.; Barth, M.J.; Czuczman, M.S.; Islam, H.; Klein, C.; Cairo, M.S. Obinutuzumab (GA101) compared to rituximab significantly enhances cell death and antibody-dependent cytotoxicity and improves overall survival against CD20(+) rituximab-sensitive/-resistant burkitt lymphoma (Bl) and precursor B-acute lymphoblastic leukaemia (pre-B-all): Potential targeted therapy in patients with poor risk CD20(+) Bl and pre-B-all. Br. J. Haematol. 2015, 171, 763–775. [Google Scholar] [PubMed]
- Ruggeri, L.; Capanni, M.; Urbani, E.; Perruccio, K.; Shlomchik, W.D.; Tosti, A.; Posati, S.; Rogaia, D.; Frassoni, F.; Aversa, F.; et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002, 295, 2097–2100. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.; Adams, M.; Carter, T.; Chen, R.; Muller, G.; Stirling, D.; Schafer, P.; Bartlett, J.B. Lenalidomide enhances natural killer cell and monocyte-mediated antibody-dependent cellular cytotoxicity of rituximab-treated CD20+ tumor cells. Clin. Cancer Res. 2008, 14, 4650–4657. [Google Scholar] [CrossRef] [PubMed]
- Orange, J.S. Human natural killer cell deficiencies. Curr. Opin. Allergy Clin. Immunol. 2006, 6, 399–409. [Google Scholar] [CrossRef] [PubMed]
- Orange, J.S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 2013, 132, 515–526. [Google Scholar] [CrossRef] [PubMed]
- Plonquet, A.; Haioun, C.; Jais, J.P.; Debard, A.L.; Salles, G.; Bene, M.C.; Feugier, P.; Rabian, C.; Casasnovas, O.; Labalette, M.; et al. Peripheral blood natural killer cell count is associated with clinical outcome in patients with AAIPI 2–3 diffuse large B-cell lymphoma. Ann. Oncol. 2007, 18, 1209–1215. [Google Scholar] [CrossRef] [PubMed]
- Dupuy, S.; Lambert, M.; Zucman, D.; Choukem, S.P.; Tognarelli, S.; Pages, C.; Lebbe, C.; Caillat-Zucman, S. Human herpesvirus 8 (HHV8) sequentially shapes the NK cell repertoire during the course of asymptomatic infection and kaposi sarcoma. PLoS Pathog. 2012, 8, e1002486. [Google Scholar] [CrossRef] [PubMed]
- Beldi-Ferchiou, A.; Lambert, M.; Dogniaux, S.; Vely, F.; Vivier, E.; Olive, D.; Dupuy, S.; Levasseur, F.; Zucman, D.; Lebbe, 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] [PubMed]
- Sene, D.; Levasseur, F.; Abel, M.; Lambert, M.; Camous, X.; Hernandez, C.; Pene, V.; Rosenberg, A.R.; Jouvin-Marche, E.; Marche, P.N.; et al. Hepatitis c virus (HCV) evades NKG2D-dependent NK cell responses through NS5A-mediated imbalance of inflammatory cytokines. PLoS Pathog. 2010, 6, e1001184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cerboni, C.; Neri, F.; Casartelli, N.; Zingoni, A.; Cosman, D.; Rossi, P.; Santoni, A.; Doria, M. Human immunodeficiency virus 1 NEF protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J. Gen. Virol. 2007, 88, 242–250. [Google Scholar] [CrossRef] [PubMed]
- De Maria, A.; Fogli, M.; Costa, P.; Murdaca, G.; Puppo, F.; Mavilio, D.; Moretta, A.; Moretta, L. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKP46, NKP30 and NKP44). Eur. J. Immunol. 2003, 33, 2410–2418. [Google Scholar] [CrossRef] [PubMed]
- Chretien, A.S.; Le Roy, A.; Vey, N.; Prebet, T.; Blaise, D.; Fauriat, C.; Olive, D. Cancer-induced alterations of NK-mediated target recognition: Current and investigational pharmacological strategies aiming at restoring NK-mediated anti-tumor activity. Front. Immunol. 2014, 5, 122. [Google Scholar] [CrossRef] [PubMed]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef] [PubMed]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus ipilimumab in advanced melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [Google Scholar] [CrossRef] [PubMed]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed]
- Agata, Y.; Kawasaki, A.; Nishimura, H.; Ishida, Y.; Tsubata, T.; Yagita, H.; Honjo, T. Expression of the PD-1 antigen on the surface of stimulated mouse t and b lymphocytes. Int. Immunol. 1996, 8, 765–772. [Google Scholar] [CrossRef] [PubMed]
- Bennett, F.; Luxenberg, D.; Ling, V.; Wang, I.M.; Marquette, K.; Lowe, D.; Khan, N.; Veldman, G.; Jacobs, K.A.; Valge-Archer, V.E.; et al. Program death-1 engagement upon TCR activation has distinct effects on costimulation and cytokine-driven proliferation: Attenuation of icos, IL-4, and IL-21, but not CD28, IL-7, and IL-15 responses. J. Immunol. 2003, 170, 711–718. [Google Scholar] [CrossRef] [PubMed]
- Thibult, M.L.; Mamessier, E.; Gertner-Dardenne, J.; Pastor, S.; Just-Landi, S.; Xerri, L.; Chetaille, B.; Olive, D. PD-1 is a novel regulator of human B-cell activation. J. Immunol. 2013, 25, 129–137. [Google Scholar] [CrossRef] [PubMed]
- Nishimura, H.; Okazaki, T.; Tanaka, Y.; Nakatani, K.; Hara, M.; Matsumori, A.; Sasayama, S.; Mizoguchi, A.; Hiai, H.; Minato, N.; et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291, 319–322. [Google Scholar] [CrossRef] [PubMed]
- Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an itim motif-carrying immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef]
- Prokunina, L.; Gunnarsson, I.; Sturfelt, G.; Truedsson, L.; Seligman, V.A.; Olson, J.L.; Seldin, M.F.; Criswell, L.A.; Alarcon-Riquelme, M.E. The systemic lupus erythematosus-associated PDCD1 polymorphism PD1.3A in lupus nephritis. Arthritis Rheum. 2004, 50, 327–328. [Google Scholar] [CrossRef] [PubMed]
- Kong, E.K.; Prokunina-Olsson, L.; Wong, W.H.; Lau, C.S.; Chan, T.M.; Alarcon-Riquelme, M.; Lau, Y.L. A new haplotype of PDCD1 is associated with rheumatoid arthritis in Hong Kong chinese. Arthritis Rheum. 2005, 52, 1058–1062. [Google Scholar] [CrossRef] [PubMed]
- Kroner, A.; Mehling, M.; Hemmer, B.; Rieckmann, P.; Toyka, K.V.; Maurer, M.; Wiendl, H. A PD-1 polymorphism is associated with disease progression in multiple sclerosis. Ann. Neurol. 2005, 58, 50–57. [Google Scholar] [CrossRef] [PubMed]
- Abdel-Wahab, N.; Shah, M.; Suarez-Almazor, M.E. Adverse events associated with immune checkpoint blockade in patients with cancer: A systematic review of case reports. PLoS ONE 2016, 11, e0160221. [Google Scholar] [CrossRef] [PubMed]
- Cappelli, L.C.; Gutierrez, A.K.; Baer, A.N.; Albayda, J.; Manno, R.L.; Haque, U.; Lipson, E.J.; Bleich, K.B.; Shah, A.A.; Naidoo, J.; et al. Inflammatory arthritis and sicca syndrome induced by nivolumab and ipilimumab. Ann. Rheum. Dis. 2017, 76, 43–50. [Google Scholar] [CrossRef] [PubMed]
- Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [PubMed]
- Chemnitz, J.M.; Parry, R.V.; Nichols, K.E.; June, C.H.; Riley, J.L. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents t cell activation. J. Immunol. 2004, 173, 945–954. [Google Scholar] [CrossRef] [PubMed]
- Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217. [Google Scholar] [CrossRef] [PubMed]
- Sheppard, K.A.; Fitz, L.J.; Lee, J.M.; Benander, C.; George, J.A.; Wooters, J.; Qiu, Y.; Jussif, J.M.; Carter, L.L.; Wood, C.R.; et al. PD-1 inhibits t-cell receptor induced phosphorylation of the ZAP70/CD3zeta signalosome and downstream signaling to PKCtheta. FEBS Lett. 2004, 574, 37–41. [Google Scholar] [CrossRef] [PubMed]
- Ahmadzadeh, M.; Johnson, L.A.; Heemskerk, B.; Wunderlich, J.R.; Dudley, M.E.; White, D.E.; Rosenberg, S.A. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009, 114, 1537–1544. [Google Scholar] [CrossRef] [PubMed]
- Okazaki, T.; Honjo, T. Rejuvenating exhausted t cells during chronic viral infection. Cell 2006, 124, 459–461. [Google Scholar] [CrossRef] [PubMed]
- Lipson, E.J.; Sharfman, W.H.; Drake, C.G.; Wollner, I.; Taube, J.M.; Anders, R.A.; Xu, H.; Yao, S.; Pons, A.; Chen, L.; et al. Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin. Cancer Res. 2013, 19, 462–468. [Google Scholar] [CrossRef] [PubMed]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-l1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef] [PubMed]
- Beaver, J.A.; Theoret, M.R.; Mushti, S.; He, K.; Libeg, M.; Goldberg, K.; Sridhara, R.; McKee, A.E.; Keegan, P.; Pazdur, R. FDA approval of nivolumab for the first-line treatment of patients with BRAFV600 wild-type unresectable or metastatic melanoma. Clin. Cancer Res. 2017, 23, 3479–3483. [Google Scholar] [CrossRef] [PubMed]
- Chang, W.S.; Kim, J.Y.; Kim, Y.J.; Kim, Y.S.; Lee, J.M.; Azuma, M.; Yagita, H.; Kang, C.Y. Cutting edge: Programmed death-1/programmed death ligand 1 interaction regulates the induction and maintenance of invariant NKT cell anergy. J. Immunol. 2008, 181, 6707–6710. [Google Scholar] [CrossRef] [PubMed]
- Taylor, S.; Huang, Y.; Mallett, G.; Stathopoulou, C.; Felizardo, T.C.; Sun, M.A.; Martin, E.L.; Zhu, N.; Woodward, E.L.; Elias, M.S.; et al. PD-1 regulates KLRG1+ group 2 innate lymphoid cells. J. Exp. Med. 2017, 214, 1663–1678. [Google Scholar] [CrossRef] [PubMed]
- MacFarlane, A.W.T.; 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]
- 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] [PubMed]
- 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]
- Alvarez, I.B.; Pasquinelli, V.; Jurado, J.O.; Abbate, E.; Musella, R.M.; de la Barrera, S.S.; Garcia, V.E. Role played by the programmed death-1-programmed death ligand pathway during innate immunity against mycobacterium tuberculosis. J. Infect. Dis. 2010, 202, 524–532. [Google Scholar] [CrossRef] [PubMed]
- Golden-Mason, L.; Klarquist, J.; Wahed, A.S.; Rosen, H.R. Cutting edge: Programmed death-1 expression is increased on immunocytes in chronic hepatitis c virus and predicts failure of response to antiviral therapy: Race-dependent differences. J. Immunol. 2008, 180, 3637–3641. [Google Scholar] [CrossRef] [PubMed]
- Norris, S.; Coleman, A.; Kuri-Cervantes, L.; Bower, M.; Nelson, M.; Goodier, M.R. PD-1 expression on natural killer cells and CD8(+) T cells during chronic HIV-1 infection. Viral Immunol. 2012, 25, 329–332. [Google Scholar] [CrossRef] [PubMed]
- 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.e3. [Google Scholar] [CrossRef] [PubMed]
- Seo, H.; Jeon, I.; Kim, B.S.; Park, M.; Bae, E.A.; Song, B.; Koh, C.H.; Shin, K.S.; Kim, I.K.; Choi, K.; et al. IL-21-mediated reversal of NK cell exhaustion facilitates anti-tumour immunity in MHC class I-deficient tumours. Nat. Commun. 2017, 8, 15776. [Google Scholar] [CrossRef] [PubMed]
- Ardolino, M.; Azimi, C.S.; Iannello, A.; Trevino, T.N.; Horan, L.; Zhang, L.; Deng, W.; Ring, A.M.; Fischer, S.; Garcia, K.C.; et al. Cytokine therapy reverses NK cell anergy in MHC-deficient tumors. J. Clin. Investig. 2014, 124, 4781–4794. [Google Scholar] [CrossRef] [PubMed]
- Rudd, C.E.; Taylor, A.; Schneider, H. CD28 and CTLA-4 coreceptor expression and signal transduction. Immunol. Rev. 2009, 229, 12–26. [Google Scholar] [CrossRef] [PubMed]
- Bour-Jordan, H.; Esensten, J.H.; Martinez-Llordella, M.; Penaranda, C.; Stumpf, M.; Bluestone, J.A. Intrinsic and extrinsic control of peripheral T-cell tolerance by costimulatory molecules of the CD28/B7 family. Immunol. Rev. 2011, 241, 180–205. [Google Scholar] [CrossRef] [PubMed]
- Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef]
- Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative disorders with early lethality in mice deficient in CTLA-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef] [PubMed]
- Cho, H.; Kang, H.; Lee, H.H.; Kim, C.W. Programmed cell death 1 (PD-1) and cytotoxic t lymphocyte-associated antigen 4 (CTLA-4) in viral hepatitis. Int. J. Mol. Sci. 2017, 18, 1517. [Google Scholar] [CrossRef] [PubMed]
- Montler, R.; Bell, R.B.; Thalhofer, C.; Leidner, R.; Feng, Z.; Fox, B.A.; Cheng, A.C.; Bui, T.G.; Tucker, C.; Hoen, H.; et al. OX40, PD-1 and CTLA-4 are selectively expressed on tumor-infiltrating T cells in head and neck cancer. Clin. Transl. Immunol. 2016, 5, e70. [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] [PubMed]
- Terme, M.; Ullrich, E.; Aymeric, L.; Meinhardt, K.; Coudert, J.D.; Desbois, M.; Ghiringhelli, F.; Viaud, S.; Ryffel, B.; Yagita, H.; et al. Cancer-induced immunosuppression: IL-18-elicited immunoablative NK cells. Cancer Res. 2012, 72, 2757–2767. [Google Scholar] [CrossRef] [PubMed]
- Jie, H.B.; Schuler, P.J.; Lee, S.C.; Srivastava, R.M.; Argiris, A.; Ferrone, S.; Whiteside, T.L.; Ferris, R.L. CTLA-4(+) regulatory T cells increased in cetuximab-treated head and neck cancer patients suppress NK cell cytotoxicity and correlate with poor prognosis. Cancer Res. 2015, 75, 2200–2210. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Hannani, D.; Vetizou, 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]
- Kerdiles, Y.; Ugolini, S.; Vivier, E. T cell regulation of natural killer cells. J. Exp. Med. 2013, 210, 1065–1068. [Google Scholar] [CrossRef] [PubMed]
- Zhu, C.; Anderson, A.C.; Schubart, A.; Xiong, H.; Imitola, J.; Khoury, S.J.; Zheng, X.X.; Strom, T.B.; Kuchroo, V.K. The TIM-3 ligand galectin-9 negatively regulates t helper type 1 immunity. Nat. Immunol. 2005, 6, 1245–1252. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Nakayama, M.; Akiba, H.; Takeda, K.; Kojima, Y.; Hashiguchi, M.; Azuma, M.; Yagita, H.; Okumura, K. TIM-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood 2009, 113, 3821–3830. [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] [PubMed]
- 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]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. LAG-3, TIM-3, and TIGIT: Co-inhibitory receptors with specialized functions in immune regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef] [PubMed]
- Sun, H.; Gao, W.; Pan, W.; Zhang, Q.; Wang, G.; Feng, D.; Geng, X.; Yan, X.; Li, S. TIM3+ FOXP3+ treg cells are potent inhibitors of effector T cells and are suppressed in rheumatoid arthritis. Inflammation 2017, 40, 1342–1350. [Google Scholar] [CrossRef] [PubMed]
- Linedale, R.; Schmidt, C.; King, B.T.; Ganko, A.G.; Simpson, F.; Panizza, B.J.; Leggatt, G.R. Elevated frequencies of CD8 T cells expressing PD-1, CTLA-4 and TIM-3 within tumour from perineural squamous cell carcinoma patients. PLoS ONE 2017, 12, e0175755. [Google Scholar] [CrossRef] [PubMed]
- Jin, H.T.; Anderson, A.C.; Tan, W.G.; West, E.E.; Ha, S.J.; Araki, K.; Freeman, G.J.; Kuchroo, V.K.; Ahmed, R. Cooperation of TIM-3 and PD-1 in CD8 T-cell exhaustion during chronic viral infection. Proc. Natl. Acad. Sci. USA 2010, 107, 14733–14738. [Google Scholar] [CrossRef] [PubMed]
- Fourcade, J.; Sun, Z.; Benallaoua, M.; Guillaume, P.; Luescher, I.F.; Sander, C.; Kirkwood, J.M.; Kuchroo, V.; Zarour, H.M. Upregulation of TIM-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010, 207, 2175–2186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Zhang, S.; Hu, Y.; Yang, Z.; Li, J.; Liu, X.; Deng, L.; Wang, Y.; Zhang, X.; Jiang, T.; et al. Targeting PD-1 and TIM-3 pathways to reverse CD8 T-cell exhaustion and enhance ex vivo T-cell responses to autologous dendritic/tumor vaccines. J. Immunother. 2016, 39, 171–180. [Google Scholar] [CrossRef] [PubMed]
- Sakuishi, K.; Apetoh, L.; Sullivan, J.M.; Blazar, B.R.; Kuchroo, V.K.; Anderson, A.C. Targeting TIM-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 2010, 207, 2187–2194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ndhlovu, L.C.; Lopez-Verges, 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] [PubMed]
- 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] [PubMed]
- Gallois, A.; Silva, I.; Osman, I.; Bhardwaj, N. Reversal of natural killer cell exhaustion by TIM-3 blockade. Oncoimmunology 2014, 3, e946365. [Google Scholar] [CrossRef] [PubMed]
- Van Audenaerde, J.R.M.; de Waele, J.; Marcq, E.; van Loenhout, J.; Lion, E.; van den Bergh, J.M.J.; Jesenofsky, R.; Masamune, A.; Roeyen, G.; Pauwels, P.; et al. Interleukin-15 stimulates natural killer cell-mediated killing of both human pancreatic cancer and stellate cells. Oncotarget 2017, 8, 56968–56979. [Google Scholar] [CrossRef] [PubMed]
- Ju, Y.; Hou, N.; Meng, J.; Wang, X.; Zhang, X.; Zhao, D.; Liu, Y.; Zhu, F.; Zhang, L.; Sun, W.; et al. T cell immunoglobulin- and mucin-domain-containing molecule-3 (TIM-3) mediates natural killer cell suppression in chronic hepatitis B. J. Hepatol. 2010, 52, 322–329. [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] [PubMed]
- 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]
- 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] [PubMed]
- 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] [PubMed]
- Stanietsky, N.; Rovis, T.L.; Glasner, A.; Seidel, E.; Tsukerman, P.; Yamin, R.; Enk, J.; Jonjic, S.; Mandelboim, O. Mouse tigit inhibits NK-cell cytotoxicity upon interaction with PVR. Eur. J. Immunol. 2013, 43, 2138–2150. [Google Scholar] [CrossRef] [PubMed]
- Bottino, C.; Castriconi, R.; Pende, D.; Rivera, P.; Nanni, M.; Carnemolla, B.; Cantoni, C.; Grassi, J.; Marcenaro, S.; Reymond, N.; et al. Identification of PVR (CD155) and nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule. J. Exp. Med. 2003, 198, 557–567. [Google Scholar] [CrossRef] [PubMed]
- Iguchi-Manaka, A.; Kai, H.; Yamashita, Y.; Shibata, K.; Tahara-Hanaoka, S.; Honda, S.; Yasui, T.; Kikutani, H.; Shibuya, K.; Shibuya, A. Accelerated tumor growth in mice deficient in DNAM-1 receptor. J. Exp. Med. 2008, 205, 2959–2964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. CII 2017. [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] [PubMed]
- 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] [PubMed]
- Huard, B.; Prigent, P.; Tournier, M.; Bruniquel, D.; Triebel, F. CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-ig fusion proteins. Eur. J. Immunol. 1995, 25, 2718–2721. [Google Scholar] [CrossRef] [PubMed]
- Baixeras, E.; Huard, B.; Miossec, C.; Jitsukawa, S.; Martin, M.; Hercend, T.; Auffray, C.; Triebel, F.; Piatier-Tonneau, D. Characterization of the lymphocyte activation gene 3-encoded protein. A new ligand for human leukocyte antigen class ii antigens. J. Exp. Med. 1992, 176, 327–337. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Workman, C.J.; Wang, Y.; El Kasmi, K.C.; Pardoll, D.M.; Murray, P.J.; Drake, C.G.; Vignali, D.A. LAG-3 regulates plasmacytoid dendritic cell homeostasis. J. Immunol. 2009, 182, 1885–1891. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in regulatory T cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef] [PubMed]
- Juno, J.A.; Stalker, A.T.; Waruk, J.L.; Oyugi, J.; Kimani, M.; Plummer, F.A.; Kimani, J.; Fowke, K.R. Elevated expression of LAG-3, but not PD-1, is associated with impaired INKT cytokine production during chronic HIV-1 infection and treatment. Retrovirology 2015, 12. [Google Scholar] [CrossRef] [PubMed]
- Kisielow, M.; Kisielow, J.; Capoferri-Sollami, G.; Karjalainen, K. Expression of lymphocyte activation gene 3 (LAG-3) on B cells is induced by T cells. Eur. J. Immunol. 2005, 35, 2081–2088. [Google Scholar] [CrossRef] [PubMed]
- Hannier, S.; Triebel, F. The MHC class II ligand lymphocyte activation gene-3 is co-distributed with CD8 and CD3-TCR molecules after their engagement by MAB or peptide-MHC class I complexes. Int. Immunol. 1999, 11, 1745–1752. [Google Scholar] [CrossRef] [PubMed]
- Workman, C.J.; Dugger, K.J.; Vignali, D.A. Cutting edge: Molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J. Immunol. 2002, 169, 5392–5395. [Google Scholar] [CrossRef] [PubMed]
- Sierro, S.; Romero, P.; Speiser, D.E. The CD4-like molecule LAG-3, biology and therapeutic applications. Expert Opin. Ther. Targets 2011, 15, 91–101. [Google Scholar] [CrossRef] [PubMed]
- Camisaschi, C.; Casati, C.; Rini, F.; Perego, M.; de Filippo, A.; Triebel, F.; Parmiani, G.; Belli, F.; Rivoltini, L.; Castelli, C. LAG-3 expression defines a subset of CD4(+)CD25(high)FOXP3(+) regulatory T cells that are expanded at tumor sites. J. Immunol. 2010, 184, 6545–6551. [Google Scholar] [CrossRef] [PubMed]
- Ye, B.; Li, X.; Dong, Y.; Wang, Y.; Tian, L.; Lin, S.; Liu, X.; Kong, H.; Chen, Y. Increasing LAG-3 expression suppresses T-cell function in chronic hepatitis B: A balance between immunity strength and liver injury extent. Medicine 2017, 96, e5275. [Google Scholar] [CrossRef] [PubMed]
- Matsuzaki, J.; Gnjatic, S.; Mhawech-Fauceglia, P.; Beck, A.; Miller, A.; Tsuji, T.; Eppolito, C.; Qian, F.; Lele, S.; Shrikant, P.; et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 2010, 107, 7875–7880. [Google Scholar] [CrossRef] [PubMed]
- Macon-Lemaitre, L.; Triebel, F. The negative regulatory function of the lymphocyte-activation gene-3 co-receptor (CD223) on human T cells. Immunology 2005, 115, 170–178. [Google Scholar] [CrossRef] [PubMed]
- Blackburn, S.D.; Shin, H.; Haining, W.N.; Zou, T.; Workman, C.J.; Polley, A.; Betts, M.R.; Freeman, G.J.; Vignali, D.A.; Wherry, E.J. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 2009, 10, 29–37. [Google Scholar] [CrossRef] [PubMed]
- Freeman, G.J.; Sharpe, A.H. A new therapeutic strategy for malaria: Targeting T cell exhaustion. Nat. Immunol. 2012, 13, 113–115. [Google Scholar] [CrossRef] [PubMed]
- Woo, S.R.; Turnis, M.E.; Goldberg, M.V.; Bankoti, J.; Selby, M.; Nirschl, C.J.; Bettini, M.L.; Gravano, D.M.; Vogel, P.; Liu, C.L.; et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012, 72, 917–927. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- 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]
- Taborda, N.A.; Hernandez, J.C.; Lajoie, J.; Juno, J.A.; Kimani, J.; Rugeles, M.T.; Fowke, K.R. Short communication: Low expression of activation and inhibitory molecules on NK cells and CD4(+) T cells is associated with viral control. AIDS Res. Hum. Retrovir. 2015, 31, 636–640. [Google Scholar] [CrossRef] [PubMed]
- Long, E.O.; Barber, D.F.; Burshtyn, D.N.; Faure, M.; Peterson, M.; Rajagopalan, S.; Renard, V.; Sandusky, M.; Stebbins, C.C.; Wagtmann, N.; et al. Inhibition of natural killer cell activation signals by killer cell immunoglobulin-like receptors (CD158). Immunol. Rev. 2001, 181, 223–233. [Google Scholar] [CrossRef] [PubMed]
- Yusa, S.; Campbell, K.S. SRC homology region 2-containing protein tyrosine phosphatase-2 (SHP-2) can play a direct role in the inhibitory function of killer cell IG-like receptors in human NK cells. J. Immunol. 2003, 170, 4539–4547. [Google Scholar] [CrossRef] [PubMed]
- Karre, K. Natural killer cell recognition of missing self. Nat. Immunol. 2008, 9, 477–480. [Google Scholar] [CrossRef] [PubMed]
- Velardi, A.; Ruggeri, L.; Alessandro; Moretta; Moretta, L. NK cells: A lesson from mismatched hematopoietic transplantation. Trends Immunol. 2002, 23, 438–444. [Google Scholar] [CrossRef]
- Shi, J.; Tricot, G.; Szmania, S.; Rosen, N.; Garg, T.K.; Malaviarachchi, P.A.; Moreno, A.; Dupont, B.; Hsu, K.C.; Baxter-Lowe, L.A.; et al. Infusion of haplo-identical killer immunoglobulin-like receptor ligand mismatched NK cells for relapsed myeloma in the setting of autologous stem cell transplantation. Br. J. Haematol. 2008, 143, 641–653. [Google Scholar] [CrossRef] [PubMed]
- Cartron, G.; Dacheux, L.; Salles, G.; Solal-Celigny, P.; Bardos, P.; Colombat, P.; Watier, H. Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IGG FC receptor FCγRIIIA gene. Blood 2002, 99, 754–758. [Google Scholar] [CrossRef] [PubMed]
- Weng, W.K.; Levy, R. Two immunoglobulin g fragment c receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J. Clin. Oncol. 2003, 21, 3940–3947. [Google Scholar] [CrossRef] [PubMed]
- Kohrt, H.E.; Thielens, A.; Marabelle, A.; Sagiv-Barfi, I.; Sola, C.; Chanuc, F.; Fuseri, N.; Bonnafous, C.; Czerwinski, D.; Rajapaksa, A.; et al. Anti-KIR antibody enhancement of anti-lymphoma activity of natural killer cells as monotherapy and in combination with anti-CD20 antibodies. Blood 2014, 123, 678–686. [Google Scholar] [CrossRef] [PubMed]
- Benson, D.M., Jr.; Cohen, A.D.; Jagannath, S.; Munshi, N.C.; Spitzer, G.; Hofmeister, C.C.; Efebera, Y.A.; Andre, P.; Zerbib, R.; Caligiuri, M.A. A phase I trial of the anti-KIR antibody IPH2101 and lenalidomide in patients with relapsed/refractory multiple myeloma. Clin. Cancer Res. 2015, 21, 4055–4061. [Google Scholar] [CrossRef] [PubMed]
- Benson, D.M., Jr.; Hofmeister, C.C.; Padmanabhan, S.; Suvannasankha, A.; Jagannath, S.; Abonour, R.; Bakan, C.; Andre, P.; Efebera, Y.; Tiollier, J.; et al. A phase 1 trial of the anti-KIR antibody IPH2101 in patients with relapsed/refractory multiple myeloma. Blood 2012, 120, 4324–4333. [Google Scholar] [CrossRef] [PubMed]
- Korde, N.; Carlsten, M.; Lee, M.J.; Minter, A.; Tan, E.; Kwok, M.; Manasanch, E.; Bhutani, M.; Tageja, N.; Roschewski, M.; et al. A phase Ii trial of pan-KIR2D blockade with IPH2101 in smoldering multiple myeloma. Haematologica 2014, 99, e81–e83. [Google Scholar] [CrossRef] [PubMed]
- Borrego, F.; Ulbrecht, M.; Weiss, E.H.; Coligan, J.E.; Brooks, A.G. Recognition of human histocompatibility leukocyte antigen (HLA)-e complexed with hla class I signal sequence-derived peptides by CD94/NKG2 confers protection from natural killer cell-mediated lysis. J. Exp. Med. 1998, 187, 813–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, N.; Llano, M.; Carretero, M.; Ishitani, A.; Navarro, F.; Lopez-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] [PubMed]
- Llano, M.; Lee, N.; Navarro, F.; Garcia, P.; Albar, J.P.; Geraghty, D.E.; Lopez-Botet, M. HLA-E-bound peptides influence recognition by inhibitory and triggering CD94/NKG2 receptors: Preferential response to an HLA-G-derived nonamer. Eur. J. Immunol. 1998, 28, 2854–2863. [Google Scholar] [CrossRef]
- Braud, V.; Jones, E.Y.; McMichael, A. The human major histocompatibility complex class IB molecule HLA-E binds signal sequence-derived peptides with primary anchor residues at positions 2 and 9. Eur. J. Immunol. 1997, 27, 1164–1169. [Google Scholar] [CrossRef] [PubMed]
- Lee, N.; Goodlett, D.R.; Ishitani, A.; Marquardt, H.; Geraghty, D.E. HLA-E surface expression depends on binding of tap-dependent peptides derived from certain HLA class I signal sequences. J. Immunol. 1998, 160, 4951–4960. [Google Scholar] [PubMed]
- Costa Arantes, D.A.; Goncalves, A.S.; Jham, B.C.; Duarte, E.C.B.; de Paula, E.C.; de Paula, H.M.; Mendonca, E.F.; Batista, A.C. Evaluation of HLA-G, HLA-E, and PD-l1 proteins in oral osteosarcomas. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2017, 123, e188–e196. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Zhang, D.; Li, F.; Xiao, Z.; Wu, M.; Shi, D.; Xiang, P.; Bao, Z. Loss of fas expression and high expression of HLA-E promoting the immune escape of early colorectal cancer cells. Oncol. Lett. 2017, 13, 3379–3386. [Google Scholar] [CrossRef] [PubMed]
- McWilliams, E.M.; Mele, J.M.; Cheney, C.; Timmerman, E.A.; Fiazuddin, F.; Strattan, E.J.; Mo, X.; Byrd, J.C.; Muthusamy, N.; Awan, F.T. Therapeutic CD94/NKG2A blockade improves natural killer cell dysfunction in chronic lymphocytic leukemia. Oncoimmunology 2016, 5, e1226720. [Google Scholar] [CrossRef] [PubMed]
Inhibitory Immune Checkpoint | Expression | Ligands | Direct Impact on NK Cell Functions |
---|---|---|---|
PD-1 | Activated T and B cells, NK cells, natural killer T (NKT) cells, ILC-2 cells and myeloid cells. | PD-L1 (B7-H1) and PD-L2 (B7-DC) | Inhibition of NK cell cytolytic activity and cytokine production. |
CTLA-4 | Treg cells, Activated T and B cells, Activated mouse NK cells, Human NK cells? | B7-1 and B7-2 | Inhibition of mouse NK-cell IFN-γ production. Direct effect on human NK cells not documented. |
TIM-3 | NK, T, NKT and myeloid cells. | Galectin-9 HMGB1 CEACAM1 | Dual role (inhibition/activation of NK cell functions) depending on the experimental or clinical setting. |
T Cell Immunoglobulin and ITIM Domain (TIGIT) | NK cells, T cells. | CD112 (PVRL2) CD155 (PVR) | Inhibition of NK cell functions. |
LAG-3 | Activated T and NK cells, Treg, B cells, plasmacytoid dendritic cell (DC), NKT. | MHC class II molecules | The impact of LAG-3 on NK cell functions is controversial and not well documented. |
Inhibitory KIRs | NK cells, CD8 T cells. | MHC class I molecules | Inhibition of NK cell functions. |
NKG2A | NK cells, CD8 T cells. | HLA-E (non-classical MHC class I molecule) | Inhibition of NK cell functions. |
© 2017 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Beldi-Ferchiou, A.; Caillat-Zucman, S. Control of NK Cell Activation by Immune Checkpoint Molecules. Int. J. Mol. Sci. 2017, 18, 2129. https://doi.org/10.3390/ijms18102129
Beldi-Ferchiou A, Caillat-Zucman S. Control of NK Cell Activation by Immune Checkpoint Molecules. International Journal of Molecular Sciences. 2017; 18(10):2129. https://doi.org/10.3390/ijms18102129
Chicago/Turabian StyleBeldi-Ferchiou, Asma, and Sophie Caillat-Zucman. 2017. "Control of NK Cell Activation by Immune Checkpoint Molecules" International Journal of Molecular Sciences 18, no. 10: 2129. https://doi.org/10.3390/ijms18102129
APA StyleBeldi-Ferchiou, A., & Caillat-Zucman, S. (2017). Control of NK Cell Activation by Immune Checkpoint Molecules. International Journal of Molecular Sciences, 18(10), 2129. https://doi.org/10.3390/ijms18102129