R. B. Herberman, M. E. Nunn, H. T. Holden, and D. H. Lavrin, Natural cytotoxic reactivity of mouse lymphoid cells against syngeneic and allogeneic tumors. II. Characterization of effector cells, Int. J. Cancer, vol.16, pp.230-239, 1975.

R. Kiessling, E. Klein, and H. Wigzell, Natural" killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype, Eur. J. Immunol, vol.5, pp.112-117, 1975.

E. Vivier, E. Tomasello, M. Baratin, T. Walzer, and S. Ugolini, Functions of natural killer cells, Nat. Immunol, vol.9, pp.503-510, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00294184

N. D. Huntington, H. Tabarias, K. Fairfax, J. Brady, Y. Hayakawa et al., NK cell maturation and peripheral homeostasis is associated with KLRG1 up-regulation, J. Immunol, vol.178, pp.4764-4770, 2007.

M. A. Cooper, J. M. Elliott, P. A. Keyel, L. Yang, J. A. Carrero et al., Cytokine-induced memory-like natural killer cells, Proc. Natl. Acad. Sci, vol.106, pp.1915-1919, 2009.

J. C. Sun, J. N. Beilke, and L. L. Lanier, Adaptive immune features of natural killer cells, Nature, vol.457, pp.557-561, 2009.

G. Ferlazzo, M. Pack, D. Thomas, C. Paludan, D. Schmid et al., Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs, Proc. Natl. Acad. Sci, vol.101, pp.16606-16611, 2004.

J. Parrish-novak, S. R. Dillon, A. Nelson, A. Hammond, C. Sprecher et al., Interleukin 21 and its receptor are involved in NK cell expansion and regulation of lymphocyte function, Nature, vol.408, pp.57-63, 2000.

H. Suzuki, G. S. Duncan, H. Takimoto, and T. W. Mak, Abnormal development of intestinal intraepithelial lymphocytes and peripheral natural killer cells in mice lacking the IL-2 receptor beta chain, J. Exp. Med, vol.185, pp.499-505, 1997.

J. P. Lodolce, D. L. Boone, S. Chai, R. E. Swain, T. Dassopoulos et al., IL-15 receptor maintains lymphoid homeostasis by supporting lymphocyte homing and proliferation, Immunity, vol.9, pp.669-676, 1998.

M. K. Kennedy, M. Glaccum, S. N. Brown, E. A. Butz, J. L. Viney et al., Reversible defects in natural killer and memory CD8 T cell lineages in interleukin 15-deficient mice, J. Exp. Med, vol.191, pp.771-780, 2000.

E. Eckelhart, W. Warsch, E. Zebedin, O. Simma, D. Stoiber et al., A novel Ncr1-Cre mouse reveals the essential role of STAT5 for NK-cell survival and development, Blood, vol.117, pp.1565-1573, 2011.

T. A. Fehniger, S. F. Cai, X. Cao, A. J. Bredemeyer, R. M. Presti et al., Acquisition of murine NK cell cytotoxicity requires the translation of a pre-existing pool of granzyme B and perforin mRNAs, Immunity, vol.26, pp.798-811, 2007.

M. Lucas, W. Schachterle, K. Oberle, P. Aichele, and A. Diefenbach, Dendritic cells prime natural killer cells by trans-presenting interleukin 15, Immunity, vol.26, pp.503-517, 2007.

E. Mortier, T. Woo, R. Advincula, S. Gozalo, and A. Ma, IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation, J. Exp. Med, vol.205, pp.1213-1225, 2008.

R. Romee, S. E. Schneider, J. W. Leong, J. M. Chase, C. R. Keppel et al., Cytokine activation induces human memory-like NK cells, Blood, vol.120, pp.4751-4760, 2012.

R. Paolini, G. Bernardini, R. Molfetta, and A. Santoni, NK cells and interferons, Cytokine Growth Factor Rev, vol.26, pp.113-120, 2015.
URL : https://hal.archives-ouvertes.fr/pasteur-01182761

F. Mattei, G. Schiavoni, F. Belardelli, and D. F. Tough, IL-15 is expressed by dendritic cells in response to type I IFN, double-stranded RNA, or lipopolysaccharide and promotes dendritic cell activation, J. Immunol, vol.167, pp.1179-1187, 2001.

S. L. Colpitts, T. A. Stoklasek, C. R. Plumlee, J. J. Obar, C. Guo et al., Cutting edge: The role of IFN-alpha receptor and MyD88 signaling in induction of IL-15 expression in vivo, J. Immunol, vol.188, pp.2483-2487, 2012.

L. L. Lanier, Up on the tightrope: Natural killer cell activation and inhibition, Nat. Immunol, vol.9, pp.495-502, 2008.

E. Vivier, D. H. Raulet, A. Moretta, M. A. Caligiuri, L. Zitvogel et al., Innate or adaptive immunity? The example of natural killer cells, Science, vol.331, pp.44-49, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00611585

A. Moretta, C. Bottino, M. Vitale, D. Pende, R. Biassoni et al., Receptors for HLA class-I molecules in human natural killer cells, Annu. Rev. Immunol, vol.14, pp.619-648, 1996.

N. Kadri, T. L. Thanh, and P. Hoglund, Selection, tuning, and adaptation in mouse NK cell education, Immunol. Rev, vol.267, pp.167-177, 2015.

K. Karre, Natural killer cell recognition of missing self, Nat. Immunol, vol.9, pp.477-480, 2008.

E. O. Long, H. S. Kim, D. Liu, M. E. Peterson, and S. Rajagopalan, Controlling natural killer cell responses: Integration of signals for activation and inhibition, Annu. Rev. Immunol, vol.31, pp.227-258, 2013.

A. D. Barrow, C. J. Martin, and M. Colonna, The Natural Cytotoxicity Receptors in Health and Disease, Front. Immunol, vol.10, 2019.

S. H. Lee, T. Miyagi, and C. A. Biron, Keeping NK cells in highly regulated antiviral warfare, Trends Immunol, vol.28, pp.252-259, 2007.

M. J. Smyth, E. Cretney, K. Takeda, R. H. Wiltrout, L. M. Sedger et al., Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) contributes to interferon gamma-dependent natural killer cell protection from tumor metastasis, J. Exp. Med, vol.193, pp.661-670, 2001.

M. C. Cuturi, I. Anegon, F. Sherman, R. Loudon, S. C. Clark et al., Production of hematopoietic colony-stimulating factors by human natural killer cells, J. Exp. Med, vol.169, pp.569-583, 1989.

C. Fauriat, E. O. Long, H. G. Ljunggren, and Y. T. Bryceson, Regulation of human NK-cell cytokine and chemokine production by target cell recognition, Blood, vol.115, pp.2167-2176, 2010.

L. L. Lanier, G. Yu, and J. H. Phillips, Co-association of CD3 zeta with a receptor (CD16) for IgG Fc on human natural killer cells, Nature, vol.342, pp.803-805, 1989.

E. Vivier, A. J. Da-silva, M. Ackerly, H. Levine, C. E. Rudd et al., Association of a 70-kDa tyrosine phosphoprotein with the CD16: Zeta: Gamma complex expressed in human natural killer cells, Eur. J. Immunol, vol.23, pp.1872-1876, 1993.

C. Chester, K. Fritsch, and H. E. Kohrt, Natural Killer Cell Immunomodulation: Targeting Activating, Inhibitory, and Co-stimulatory Receptor Signaling for, Cancer Immunotherapy. Front. Immunol, vol.6, 2015.

C. Guillerey, N. D. Huntington, and M. J. Smyth, Targeting natural killer cells in cancer immunotherapy, Nat. Immunol, vol.17, pp.1025-1036, 2016.

A. D. Barrow and M. Colonna, Exploiting NK Cell Surveillance Pathways for Cancer Therapy, Cancers, vol.11, 2019.

J. Crespo, H. Sun, T. H. Welling, Z. Tian, and W. Zou, T cell anergy, exhaustion, senescence, and stemness in the tumor microenvironment, Curr. Opin. Immunol, vol.25, pp.214-221, 2013.

J. Bi and . Tian, Z. NK Cell Exhaustion. Front. Immunol, vol.8, p.760, 2017.

R. T. Costello, S. Sivori, E. Marcenaro, M. Lafage-pochitaloff, M. J. Mozziconacci et al., Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia, Blood, vol.99, pp.3661-3667, 2002.

M. Hasmim, Y. Messai, L. Ziani, J. Thiery, J. H. Bouhris et al., Critical Role of Tumor Microenvironment in Shaping NK Cell Functions: Implication of Hypoxic Stress, Front. Immunol, vol.6, 2015.

A. Horowitz, D. M. Strauss-albee, M. Leipold, J. Kubo, N. Nemat-gorgani et al., Genetic and environmental determinants of human NK cell diversity revealed by mass cytometry, Sci. Transl. Med, 2013.

J. M. Elliott, J. A. Wahle, and W. M. Yokoyama, MHC class I-deficient natural killer cells acquire a licensed phenotype after transfer into an MHC class I-sufficient environment, J. Exp. Med, vol.207, pp.2073-2079, 2010.

J. P. Goodridge, B. Onfelt, and K. J. Malmberg, Newtonian cell interactions shape natural killer cell education, Immunol. Rev, vol.267, pp.197-213, 2015.

N. C. Fernandez, E. Treiner, R. E. Vance, A. M. Jamieson, S. Lemieux et al., A subset of natural killer cells achieves self-tolerance without expressing inhibitory receptors specific for self-MHC molecules, Blood, vol.105, pp.4416-4423, 2005.

S. Kim, J. Poursine-laurent, S. M. Truscott, L. Lybarger, Y. J. Song et al., Licensing of natural killer cells by host major histocompatibility complex class I molecules, Nature, vol.436, pp.709-713, 2005.

L. L. Lanier, NK cell recognition, Annu. Rev. Immunol, vol.23, pp.225-274, 2005.

C. A. Stewart, F. Laugier-anfossi, F. Vely, X. Saulquin, J. Riedmuller et al., Recognition of peptide-MHC class I complexes by activating killer immunoglobulin-like receptors, Proc. Natl. Acad. Sci, vol.102, pp.13224-13229, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00079731

D. H. Raulet and R. E. Vance, Self-tolerance of natural killer cells, Nat. Rev. Immunol, vol.6, pp.520-531, 2006.

W. M. Yokoyama and S. Kim, How do natural killer cells find self to achieve tolerance? Immunity, vol.24, pp.249-257, 2006.

M. F. Wu and D. H. Raulet, Class I-deficient hemopoietic cells and nonhemopoietic cells dominantly induce unresponsiveness of natural killer cells to class I-deficient bone marrow cell grafts, J. Immunol, vol.158, pp.1628-1633, 1997.

N. T. Joncker, N. C. Fernandez, E. Treiner, E. Vivier, and D. H. Raulet, NK cell responsiveness is tuned commensurate with the number of inhibitory receptors for self-MHC class I: The rheostat model, J. Immunol, vol.182, pp.4572-4580, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00408430

H. G. Ljunggren and K. Karre, Host resistance directed selectively against H-2-deficient lymphoma variants. Analysis of the mechanism, J. Exp. Med, vol.162, pp.1745-1759, 1985.

W. T. Nash, J. Teoh, H. Wei, A. Gamache, and M. G. Brown, Know Thyself: NK-Cell Inhibitory Receptors Prompt Self-Tolerance, Education, and Viral Control, Front. Immunol, vol.5, 2014.

K. J. Malmberg, M. Carlsten, A. Bjorklund, E. Sohlberg, Y. T. Bryceson et al., Natural killer cell-mediated immunosurveillance of human cancer, Semin. Immunol, vol.31, pp.20-29, 2017.

N. Anfossi, P. Andre, S. Guia, C. S. Falk, S. Roetynck et al., Human NK cell education by inhibitory receptors for MHC class I, Immunity, vol.25, pp.331-342, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00091535

J. Hasenkamp, A. Borgerding, M. Uhrberg, C. Falk, B. Chapuy et al., Self-tolerance of human natural killer cells lacking self-HLA-specific inhibitory receptors, Scand. J. Immunol, vol.67, pp.218-229, 2008.

M. Bix, N. S. Liao, M. Zijlstra, J. Loring, R. Jaenisch et al., Rejection of class I MHC-deficient haemopoietic cells by irradiated MHC-matched mice, Nature, vol.349, pp.329-331, 1991.

P. Hoglund, C. Ohlen, E. Carbone, L. Franksson, H. G. Ljunggren et al., Recognition of beta 2-microglobulin-negative (beta 2m-) T-cell blasts by natural killer cells from normal but not from beta 2m-mice: Nonresponsiveness controlled by beta 2m-bone marrow in chimeric mice, Proc. Natl. Acad. Sci, vol.88, pp.10332-10336, 1991.

N. S. Liao, M. Bix, M. Zijlstra, R. Jaenisch, and D. Raulet, MHC class I deficiency: Susceptibility to natural killer (NK) cells and impaired NK activity, Science, vol.253, pp.199-202, 1991.

J. M. Elliott and W. M. Yokoyama, Unifying concepts of MHC-dependent natural killer cell education, Trends Immunol, vol.32, pp.364-372, 2011.

N. Kadri, A. K. Wagner, S. Ganesan, K. Karre, S. Wickstrom et al., Dynamic Regulation of NK Cell Responsiveness, Curr. Top. Microbiol. Immunol, vol.395, pp.95-114, 2016.

D. E. Oppenheim, S. J. Roberts, S. L. Clarke, R. Filler, J. M. Lewis et al., Sustained localized expression of ligand for the activating NKG2D receptor impairs natural cytotoxicity in vivo and reduces tumor immunosurveillance, Nat. Immunol, vol.6, pp.928-937, 2005.

K. Wiemann, H. W. Mittrucker, U. Feger, S. A. Welte, W. M. Yokoyama et al., Systemic NKG2D down-regulation impairs NK and CD8 T cell responses in vivo, J. Immunol, vol.175, pp.720-729, 2005.

S. K. Tripathy, P. A. Keyel, L. Yang, J. T. Pingel, T. P. Cheng et al., Continuous engagement of a self-specific activation receptor induces NK cell tolerance, J. Exp. Med, vol.205, pp.1829-1841, 2008.

C. Koch, Y. Kim, T. Zoller, C. Born, and A. Steinle, Chronic NKG2D Engagement In Vivo Differentially Impacts NK Cell Responsiveness by Activating NK Receptors, Front. Immunol, vol.8, 1466.

V. Groh, J. Wu, C. Yee, and T. Spies, Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation, Nature, vol.419, pp.734-738, 2002.

J. C. Sun and L. L. Lanier, Tolerance of NK cells encountering their viral ligand during development, J. Exp. Med, vol.205, pp.1819-1828, 2008.

F. D. Bolanos and S. K. Tripathy, Activation receptor-induced tolerance of mature NK cells in vivo requires signaling through the receptor and is reversible, J. Immunol, vol.186, pp.2765-2771, 2011.

B. Mazumdar, F. D. Bolanos, and S. K. Tripathy, Viral infection transiently reverses activation receptor-mediated NK cell hyporesponsiveness in an MHC class I-independent mechanism, Eur. J. Immunol, vol.43, pp.1345-1355, 2013.

C. A. Miner, T. K. Giri, C. E. Meyer, M. Shabsovich, and S. K. Tripathy, Acquisition of activation receptor ligand by trogocytosis renders NK cells hyporesponsive, J. Immunol, vol.194, pp.1945-1953, 2015.

S. Guia, B. N. Jaeger, S. Piatek, S. Mailfert, T. Trombik et al., Confinement of activating receptors at the plasma membrane controls natural killer cell tolerance, Sci. Signal, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00609667

E. Staaf, P. N. Hedde, S. Singh, J. Piguet, E. Gratton et al., Educated natural killer cells show dynamic movement of the activating receptor NKp46 and confinement of the inhibitory receptor Ly49A, Sci. Signal, vol.11, 2018.

P. N. Key, J. Germino, L. Yang, S. J. Piersma, and S. K. Tripathy, Chronic Ly49H Receptor Engagement in vivo Decreases NK Cell Response to Stimulation Through ITAM-Dependent and Independent Pathways Both in vitro and in vivo, Front. Immunol, vol.10, p.1692, 2019.

G. P. Dunn, A. T. Bruce, H. Ikeda, L. J. Old, and R. D. Schreiber, Cancer immunoediting: From immunosurveillance to tumor escape, Nat. Immunol, vol.3, pp.991-998, 2002.

Z. Brahmi, R. A. Bray, and S. I. Abrams, Evidence for an early calcium-independent event in the activation of the human natural killer cell cytolytic mechanism, J. Immunol, vol.135, pp.4108-4113, 1985.

A. Bajpai, B. S. Kwon, and Z. Brahmi, Rapid loss of perforin and serine protease RNA in cytotoxic lymphocytes exposed to sensitive targets, Immunology, vol.74, pp.258-263, 1991.

A. M. Shenoy and Z. Brahmi, Inhibition of the calpain-mediated proteolysis of protein kinase C enhances lytic activity in human NK cells, Cell. Immunol, vol.138, pp.24-34, 1991.

J. J. Gibboney, A. M. Shenoy, X. Jin, and Z. Brahmi, Signal transduction in activated natural killer cells and natural killer cells inactivated with sensitive targets, Nat. Immun, vol.11, pp.57-68, 1992.

S. Bauer, V. Groh, J. Wu, A. Steinle, J. H. Phillips et al., Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA, Science, vol.285, pp.727-729, 1999.

B. Sanchez-correa, S. Morgado, I. Gayoso, J. M. Bergua, J. G. Casado et al., Human NK cells in acute myeloid leukaemia patients: Analysis of NK cell-activating receptors and their ligands, Cancer Immunol. Immunother, vol.60, pp.1195-1205, 2011.

J. D. Coudert, J. Zimmer, E. Tomasello, M. Cebecauer, M. Colonna et al., Altered NKG2D function in NK cells induced by chronic exposure to NKG2D ligand-expressing tumor cells, Blood, vol.106, pp.1711-1717, 2005.
URL : https://hal.archives-ouvertes.fr/hal-00079028

N. Dulphy, A. S. Chretien, Z. Khaznadar, C. Fauriat, A. Nanbakhsh et al., Underground Adaptation to a Hostile Environment: Acute Myeloid Leukemia vs, Natural Killer Cells. Front. Immunol, vol.7, p.94, 2016.

B. Sanchez-correa, I. Gayoso, J. M. Bergua, J. G. Casado, S. Morgado et al., Decreased expression of DNAM-1 on NK cells from acute myeloid leukemia patients, Immunol. Cell. Biol, vol.90, pp.109-115, 2012.

H. Naora and M. L. Gougeon, Enhanced survival and potent expansion of the natural killer cell population of HIV-infected individuals by exogenous interleukin-15, Immunol. Lett, vol.68, pp.359-367, 1999.

A. S. Fauci, D. Mavilio, and S. Kottilil, NK cells in HIV infection: Paradigm for protection or targets for ambush, Nat. Rev. Immunol, vol.5, pp.835-843, 2005.

U. C. Meier, R. E. Owen, E. Taylor, A. Worth, N. Naoumov et al., Shared alterations in NK cell frequency, phenotype, and function in chronic human immunodeficiency virus and hepatitis C virus infections, J. Virol, vol.79, pp.12365-12374, 2005.

J. Mikulak, F. Oriolo, E. Zaghi, C. Di-vito, and D. Mavilio, Natural killer cells in HIV-1 infection and therapy, vol.31, pp.2317-2330, 2017.

S. R. Ostrowski, H. Ullum, B. K. Pedersen, J. Gerstoft, and T. L. Katzenstein, 2B4 expression on natural killer cells increases in HIV-1 infected patients followed prospectively during highly active antiretroviral therapy, Clin. Exp. Immunol, vol.141, pp.526-533, 2005.

A. De-maria, M. Fogli, P. Costa, G. Murdaca, F. Puppo et al., 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, vol.33, pp.2410-2418, 2003.

D. Mavilio, J. Benjamin, M. Daucher, G. Lombardo, S. Kottilil et al., Natural killer cells in HIV-1 infection: Dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates, Proc. Natl. Acad. Sci, vol.100, pp.15011-15016, 2003.

D. Mavilio, G. Lombardo, J. Benjamin, D. Kim, D. Follman et al., Characterization of CD56-/CD16+ natural killer (NK) cells: A highly dysfunctional NK subset expanded in HIV-infected viremic individuals, Proc. Natl. Acad. Sci, vol.102, pp.2886-2891, 2005.

M. C. Sirianni, I. Mezzaroma, F. Aiuti, and A. Moretta, Analysis of the cytolytic activity mediated by natural killer cells from acquired immunodeficiency syndrome patients in response to phytohemagglutinin or anti-CD16 monoclonal antibody, Eur. J. Immunol, vol.24, pp.1874-1878, 1994.

S. K. Kyei, H. Ogbomo, S. Li, M. Timm-mccann, R. F. Xiang et al., Mechanisms by Which Interleukin-12 Corrects Defective NK Cell Anticryptococcal Activity in HIV-Infected Patients, MBio, vol.7, pp.878-894, 2016.

M. Leszczyszyn-pynka, Natural cytotoxicity of peripheral blood mononuclear cells in Herpes simplex and Varicella-zoster virus infections, Acta Haematol. Pol, vol.26, pp.393-402, 1995.

V. V. Murugin, I. N. Zuikova, N. E. Murugina, A. E. Shulzhenko, B. V. Pinegin et al., Reduced degranulation of NK cells in patients with frequently recurring herpes, Clin. Vaccine Immunol, vol.18, pp.1410-1415, 2011.

D. L. Confer, G. M. Vercellotti, D. Kotasek, J. L. Goodman, A. Ochoa et al., Herpes simplex virus-infected cells disarm killer lymphocytes, Proc. Natl. Acad. Sci, vol.87, pp.3609-3613, 1990.

T. P. Welliver, R. P. Garofalo, Y. Hosakote, K. H. Hintz, L. Avendano et al., Severe human lower respiratory tract illness caused by respiratory syncytial virus and influenza virus is characterized by the absence of pulmonary cytotoxic lymphocyte responses, J. Infect. Dis, vol.195, pp.1126-1136, 2007.

M. L. Heltzer, S. E. Coffin, K. Maurer, A. Bagashev, Z. Zhang et al., Immune dysregulation in severe influenza, J. Leukoc. Biol, vol.85, pp.1036-1043, 2009.

V. Del-gobbo, N. Villani, S. Marini, E. Balestra, and R. Calio, Suppressor cells induced by influenza virus inhibit interleukin-2 production in mice, Immunology, vol.69, pp.454-459, 1990.

S. Nogusa, B. W. Ritz, S. H. Kassim, S. R. Jennings, and E. M. Gardner, Characterization of age-related changes in natural killer cells during primary influenza infection in mice, Mech. Ageing Dev, vol.129, pp.223-230, 2008.

H. Guo, P. Kumar, T. M. Moran, A. Garcia-sastre, Y. Zhou et al., The functional impairment of natural killer cells during influenza virus infection, Immunol. Cell Biol, vol.87, pp.579-589, 2009.

H. Mao, W. Tu, G. Qin, H. K. Law, S. F. Sia et al., Influenza virus directly infects human natural killer cells and induces cell apoptosis, J. Virol, vol.83, pp.9215-9222, 2009.

J. C. Yoon, J. B. Lim, J. H. Park, and J. M. Lee, Cell-to-cell contact with hepatitis C virus-infected cells reduces functional capacity of natural killer cells, J. Virol, vol.85, pp.12557-12569, 2011.

C. M. Yang, J. C. Yoon, J. H. Park, and J. M. Lee, Hepatitis C virus impairs natural killer cell activity via viral serine protease NS3, PLoS ONE, vol.12, 2017.

H. Arase, E. S. Mocarski, A. E. Campbell, A. B. Hill, and L. L. Lanier, Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors, Science, vol.296, pp.1323-1326, 2002.

H. R. Smith, J. W. Heusel, I. K. Mehta, S. Kim, B. G. Dorner et al., Recognition of a virus-encoded ligand by a natural killer cell activation receptor, Proc. Natl. Acad. Sci, vol.99, pp.8826-8831, 2002.

A. Beldi-ferchiou, M. Lambert, S. Dogniaux, F. Vely, E. Vivier et al., PD-1 mediates functional exhaustion of activated NK cells in patients with Kaposi sarcoma, Oncotarget, vol.7, pp.72961-72977, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01438166

S. Dupuy, M. Lambert, D. Zucman, S. P. Choukem, S. Tognarelli et al., Human Herpesvirus 8 (HHV8) sequentially shapes the NK cell repertoire during the course of asymptomatic infection and Kaposi sarcoma, PLoS Pathog, vol.8, 2012.

J. Zhang, S. Jin, X. Li, L. Liu, L. Xi et al., Human Papillomavirus Type 16 Disables the Increased Natural Killer Cells in Early Lesions of the Cervix, J. Immunol. Res, 2019.

U. A. Hasan, E. Bates, F. Takeshita, A. Biliato, R. Accardi et al., TLR9 expression and function is abolished by the cervical cancer-associated human papillomavirus type 16, J. Immunol, vol.178, pp.3186-3197, 2007.

N. A. Arreygue-garcia, A. Daneri-navarro, A. Toro-arreola, A. Cid-arregui, O. Gonzalez-ramella et al., Augmented serum level of major histocompatibility complex class I-related chain A (MICA) protein and reduced NKG2D expression on NK and T cells in patients with cervical cancer and precursor lesions, BMC Cancer, vol.8, p.16, 2008.

T. Garcia-iglesias, A. Toro-arreola, B. Albarran-somoza, S. Toro-arreola, P. E. Sanchez-hernandez et al., Low NKp30, NKp46 and NKG2D expression and reduced cytotoxic activity on NK cells in cervical cancer and precursor lesions, BMC Cancer, vol.9, p.186, 2009.

F. Souza-fonseca-guimaraes, J. Cursons, and N. D. Huntington, The Emergence of Natural Killer Cells as a Major Target in Cancer Immunotherapy, Trends Immunol, vol.40, pp.142-158, 2019.

A. D. Jacobs, R. E. Champlin, and D. W. Golde, Recombinant alpha-2-interferon for hairy cell leukemia, Blood, vol.65, pp.1017-1020, 1985.

S. A. Rosenberg, M. T. Lotze, L. M. Muul, S. Leitman, A. E. Chang et al., Observations on the systemic administration of autologous lymphokine-activated killer cells and recombinant interleukin-2 to patients with metastatic cancer, N. Engl. J. Med, vol.313, pp.1485-1492, 1985.

Y. J. Liu and . Ipc, Professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors, Annu. Rev. Immunol, vol.23, pp.275-306, 2005.

T. R. Malek, The biology of interleukin-2, Annu. Rev. Immunol, vol.26, pp.453-479, 2008.

R. M. Hansen and E. C. Borden, Current status of interferons in the treatment of cancer, Oncol. Williston Park NY, vol.6, pp.19-24, 1992.

S. Terawaki, S. Chikuma, S. Shibayama, T. Hayashi, T. Yoshida et al., IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity, J. Immunol, vol.186, pp.2772-2779, 2011.

T. Jiang, C. Zhou, and S. Ren, Role of IL-2 in cancer immunotherapy, Oncoimmunology, vol.5, 2016.

S. I. Abrams and Z. Brahmi, The functional loss of human natural killer cell activity induced by K562 is reversible via an interleukin-2-dependent mechanism, Cell. Immunol, vol.101, pp.558-570, 1986.

C. Heuser, M. Ganser, A. Hombach, H. Brand, G. Denton et al., An anti-MUC1-antibody-interleukin-2 fusion protein that activates resting NK cells to lysis of MUC1-positive tumour cells, Br. J. Cancer, vol.89, pp.1130-1139, 2003.

A. V. Lefever and A. Funahashi, Phenotype and function of natural killer cells in patients with bronchogenic carcinoma, Cancer Res, vol.51, pp.5596-5601, 1991.

G. Konjevic, K. Mirjacic-martinovic, A. Vuletic, V. Jovic, V. Jurisic et al., Low expression of CD161 and NKG2D activating NK receptor is associated with impaired NK cell cytotoxicity in metastatic melanoma patients, Clin. Exp. Metastasis, vol.24, pp.1-11, 2007.

M. R. Parkhurst, J. P. Riley, M. E. Dudley, and S. A. Rosenberg, Adoptive transfer of autologous natural killer cells leads to high levels of circulating natural killer cells but does not mediate tumor regression, Clin. Cancer Res, vol.17, pp.6287-6297, 2011.

S. Platonova, J. Cherfils-vicini, D. Damotte, L. Crozet, V. Vieillard et al., Profound coordinated alterations of intratumoral NK cell phenotype and function in lung carcinoma, Cancer Res, vol.71, pp.5412-5422, 2011.

K. S. Reiners, J. Kessler, M. Sauer, A. Rothe, H. P. Hansen et al., Rescue of impaired NK cell activity in hodgkin lymphoma with bispecific antibodies in vitro and in patients, Mol. Ther, vol.21, pp.895-903, 2013.

J. J. Kiladjian, E. Bourgeois, I. Lobe, T. Braun, G. Visentin et al., Cytolytic function and survival of natural killer cells are severely altered in myelodysplastic syndromes, Leukemia, vol.20, pp.463-470, 2006.

M. Ardolino, C. S. Azimi, A. Iannello, T. N. Trevino, L. Horan et al., Cytokine therapy reverses NK cell anergy in MHC-deficient tumors, J. Clin. Investig, vol.124, pp.4781-4794, 2014.

M. Kobayashi, L. Fitz, M. Ryan, R. M. Hewick, S. C. Clark et al., Identification and purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple biologic effects on human lymphocytes, J. Exp. Med, vol.170, pp.827-845, 1989.

J. M. Weiss, J. J. Subleski, J. M. Wigginton, and R. H. Wiltrout, Immunotherapy of cancer by IL-12-based cytokine combinations, Expert Opin. Biol. Ther, vol.7, pp.1705-1721, 2007.

J. W. Leong, J. M. Chase, R. Romee, S. E. Schneider, R. P. Sullivan et al., Preactivation with IL-12, IL-15, and IL-18 induces CD25 and a functional high-affinity IL-2 receptor on human cytokine-induced memory-like natural killer cells, Biol. Blood Marrow. Transplant, vol.20, pp.463-473, 2014.

J. Marks-konczalik, S. Dubois, J. M. Losi, H. Sabzevari, N. Yamada et al., IL-2-induced activation-induced cell death is inhibited in IL-15 transgenic mice, Proc. Natl. Acad. Sci, vol.97, pp.11445-11450, 2000.

T. A. Waldmann, The biology of interleukin-2 and interleukin-15: Implications for cancer therapy and vaccine design, Nat. Rev. Immunol, vol.6, pp.595-601, 2006.

M. A. Cheever, Twelve immunotherapy drugs that could cure cancers, Immunol. Rev, vol.222, pp.357-368, 2008.

D. H. Verhoeven, A. S. De-hooge, E. C. Mooiman, S. J. Santos, M. M. Ten-dam et al., NK cells recognize and lyse Ewing sarcoma cells through NKG2D and DNAM-1 receptor dependent pathways, Mol. Immunol, vol.45, pp.3917-3925, 2008.

M. J. Szczepanski, M. Szajnik, A. Welsh, K. A. Foon, T. L. Whiteside et al., Interleukin-15 enhances natural killer cell cytotoxicity in patients with acute myeloid leukemia by upregulating the activating NK cell receptors, Cancer Immunol. Immunother, vol.59, pp.73-79, 2010.

M. Balsamo, W. Vermi, M. Parodi, G. Pietra, C. Manzini et al., Melanoma cells become resistant to NK-cell-mediated killing when exposed to NK-cell numbers compatible with NK-cell infiltration in the tumor, Eur. J. Immunol, vol.42, pp.1833-1842, 2012.

I. Nakamura, Y. Furuichi, and K. Sugimoto, Restoration of natural killer cell activity by interferon-free direct-acting antiviral combination therapy in chronic hepatitis C patients, Hepatol. Res, vol.48, pp.855-861, 2018.

S. J. Lin, R. L. Roberts, B. J. Ank, Q. H. Nguyen, E. K. Thomas et al., Effect of interleukin (IL)-12 and IL-15 on activated natural killer (ANK) and antibody-dependent cellular cytotoxicity (ADCC) in HIV infection, J. Clin. Immunol, vol.18, pp.335-345, 1998.

B. J. Park, K. A. Wannemuehler, B. J. Marston, N. Govender, P. G. Pappas et al., Estimation of the current global burden of cryptococcal meningitis among persons living with HIV/AIDS, AIDS, vol.23, pp.525-530, 2009.

C. A. Horn and R. G. Washburn, Anticryptococcal activity of NK cell-enriched peripheral blood lymphocytes from human immunodeficiency virus-infected subjects: Responses to interleukin-2, interferon-gamma, and interleukin-12, J. Infect. Dis, vol.172, pp.1023-1027, 1995.

T. Saito, R. Ruffman, R. D. Welker, R. B. Herberman, and M. A. Chirigos, Development of hyporesponsiveness of natural killer cells to augmentation of activity after multiple treatments with biological response modifiers, Cancer Immunol. Immunother, vol.19, pp.130-135, 1985.

T. Saito, R. D. Welker, H. Fukui, R. B. Herberman, and M. A. Chirigos, Development of hyporesponsiveness to augmentation of natural killer cell activity after multiple doses of maleic anhydride divinyl ether: Association with decreased numbers of large granular lymphocytes, Cell. Immunol, vol.90, pp.577-589, 1985.

J. E. Talmadge, R. B. Herberman, M. A. Chirigos, A. E. Maluish, M. A. Schneider et al., Hyporesponsiveness to augmentation of murine natural killer cell activity in different anatomical compartments by multiple injections of various immunomodulators including recombinant interferons and interleukin 2, J. Immunol, vol.135, pp.2483-2491, 1985.

W. E. Fogler, K. Volker, M. Watanabe, J. M. Wigginton, P. Roessler et al., Recruitment of hepatic NK cells by IL-12 is dependent on IFN-gamma and VCAM-1 and is rapidly down-regulated by a mechanism involving T cells and expression of Fas, J. Immunol, vol.161, pp.6014-6021, 1998.

K. G. Elpek, M. P. Rubinstein, A. Bellemare-pelletier, A. W. Goldrath, and S. J. Turley, Mature natural killer cells with phenotypic and functional alterations accumulate upon sustained stimulation with IL-15/IL-15Ralpha complexes, Proc. Natl. Acad. Sci, vol.107, pp.21647-21652, 2010.

G. Romera-cardenas, L. M. Thomas, S. Lopez-cobo, E. M. Garcia-cuesta, E. O. Long et al., Ionomycin Treatment Renders NK Cells Hyporesponsive, PLoS ONE, vol.11, 2016.

M. Frutoso, S. Morisseau, F. Tamzalit, A. Quemener, D. Meghnem et al., Emergence of NK Cell Hyporesponsiveness after Two IL-15 Stimulation Cycles, J. Immunol, vol.201, pp.493-506, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01805185

E. Mortier, A. Quemener, P. Vusio, I. Lorenzen, Y. Boublik et al., Soluble interleukin-15 receptor alpha (IL-15R alpha)-sushi as a selective and potent agonist of IL-15 action through IL-15R beta/gamma. Hyperagonist IL-15 x IL-15R alpha fusion proteins, J. Biol. Chem, vol.281, pp.1612-1619, 2006.

M. Epardaud, K. G. Elpek, M. P. Rubinstein, A. R. Yonekura, A. Bellemare-pelletier et al., Interleukin-15/interleukin-15R alpha complexes promote destruction of established tumors by reviving tumor-resident CD8+ T cells, Cancer Res, vol.68, pp.2972-2983, 2008.
URL : https://hal.archives-ouvertes.fr/hal-01602340

K. P. Han, X. Zhu, B. Liu, E. Jeng, L. Kong et al., IL-15:IL-15 receptor alpha superagonist complex: High-level co-expression in recombinant mammalian cells, purification and characterization, Cytokine, vol.56, pp.804-810, 2011.

E. Chertova, C. Bergamaschi, O. Chertov, R. Sowder, J. Bear et al., Characterization and favorable in vivo properties of heterodimeric soluble IL-15.IL-15Ralpha cytokine compared to IL-15 monomer, J. Biol. Chem, vol.288, pp.18093-18103, 2013.

J. R. Huddlestone, T. C. Merigan, . Jr, and M. B. Oldstone, Induction and kinetics of natural killer cells in humans following interferon therapy, Nature, vol.282, pp.417-419, 1979.

A. E. Maluish, J. R. Ortaldo, J. C. Conlon, S. A. Sherwin, R. Leavitt et al., Depression of natural killer cytotoxicity after in vivo administration of recombinant leukocyte interferon, J. Immunol, vol.131, pp.503-507, 1983.

C. A. Spina, J. L. Fahey, D. Durkos-smith, F. Dorey, and G. Sarna, Suppression of natural killer cell cytotoxicity in the peripheral blood of patients receiving interferon therapy, J. Biol. Response Mod, vol.2, pp.458-469, 1983.

J. E. Portielje, C. H. Lamers, W. H. Kruit, A. Sparreboom, R. L. Bolhuis et al., Repeated administrations of interleukin (IL)-12 are associated with persistently elevated plasma levels of IL-10 and declining IFN-gamma, tumor necrosis factor-alpha, IL-6, and IL-8 responses, Clin. Cancer Res, vol.9, pp.76-83, 2003.

J. S. Miller, C. Morishima, D. G. Mcneel, M. R. Patel, H. E. Kohrt et al., A First-in-Human Phase I Study of Subcutaneous Outpatient Recombinant Human IL15 (rhIL15) in Adults with Advanced Solid Tumors, Clin. Cancer Res, vol.24, pp.1525-1535, 2018.

M. C. Sneller, W. C. Kopp, K. J. Engelke, J. L. Yovandich, S. P. Creekmore et al., IL-15 administered by continuous infusion to rhesus macaques induces massive expansion of CD8+ T effector memory population in peripheral blood, Blood, vol.118, pp.6845-6848, 2011.

A. L. Ellis-connell, A. J. Balgeman, K. R. Zarbock, G. Barry, A. Weiler et al., ALT-803 Transiently Reduces Simian Immunodeficiency Virus Replication in the Absence of Antiretroviral Treatment, J. Virol, vol.92, pp.1748-1765, 2018.

M. Felices, A. J. Lenvik, R. Mcelmurry, S. Chu, P. Hinderlie et al., Continuous treatment with IL-15 exhausts human NK cells via a metabolic defect, JCI Insight, vol.3, 2018.

, This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license, © 2019 by the authors. Licensee MDPI