Y. T. Bryceson, S. C. Chiang, S. Darmanin, C. Fauriat, H. Schlums et al., Molecular mechanisms of natural killer cell activation, J Innate Immun, vol.3, pp.216-242, 2011.

E. Montaldo, D. Zotto, G. , D. Chiesa, M. Mingari et al., Human NK cell receptors/markers: a tool to analyze NK cell development, subsets and function, Cytom A, vol.83, pp.702-715, 2013.

S. Mahapatra, E. M. Mace, C. G. Minard, L. R. Forbes, A. Vargas-hernandez et al., High-resolution phenotyping identifies NK cell subsets that distinguish healthy children from adults, PloS One, vol.12, p.181134, 2017.

D. M. Strauss-albee and C. A. Blish, Human NK Cell Diversity in Viral Infection: Ramifications of Ramification, Front Immunol, vol.7, p.66, 2016.

D. Sanchez-martinez, A. Vega, N. Orecchioni, S. Talarico, G. Cornillon et al., Expansion Of Allogeneic NK Cells With Efficient Antibody-Dependent, Cell Cytotoxicity Against Multiple Tumor Cells. Theranostic, vol.8, pp.3856-3869, 2018.

M. Shevtsov and G. Multhoff, Immunological and Translational Aspects of NK Cell-Based Antitumor Immunotherapies, Front Immunol, vol.7, p.492, 2016.

Y. Liu, H. Wu, M. A. Sheard, R. Sposto, S. S. Somanchi et al., Growth and activation of natural killer cells ex vivo from children with neuroblastoma for adoptive cell therapy, Clin Cancer Res Off J Am Assoc Cancer Res, vol.19, pp.2132-2143, 2013.

D. Boer, R. J. Homann, D. Perelson, and A. S. , Different dynamics of CD4+ and CD8+ T cell responses during and after acute lymphocytic choriomeningitis virus infection, J Immunol Baltim Md, vol.1950, pp.3928-3935, 2003.

A. Anel, J. I. Aguilo, E. Catalan, J. Garaude, M. G. Rathore et al.,

C. Kinase, PKC-theta) in Natural Killer Cell Function and Anti-Tumor Immunity, Front Immunol, vol.3, p.187, 2012.

A. Sarvaria, D. Jawdat, J. A. Madrigal, and A. Saudemont, Umbilical Cord Blood Natural Killer Cells, Their Characteristics, and Potential Clinical Applications, Front Immunol, vol.8, p.329, 2017.

P. D. Kottaridis, J. North, M. Tsirogianni, C. Marden, E. R. Samuel et al., Two-Stage Priming of Allogeneic Natural Killer Cells for the Treatment of Patients with Acute Myeloid Leukemia: A Phase I Trial, PLoS One, vol.10, p.123416, 2015.

L. Moretta, G. Pietra, P. Vacca, D. Pende, F. Moretta et al., All-trans retinoic acid (ATRA) induces miR-23a expression, decreases CTSC expression and granzyme B activity leading to impaired NK cell cytotoxicity, Int J Biochem Cell Biol, vol.178, pp.42-52, 2014.

N. Vey, J. H. Bourhis, N. Boissel, D. Bordessoule, T. Prebet et al., A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission, Blood, vol.120, pp.4317-4340, 2012.

P. André, C. Denis, C. Soulas, C. Bourbon-caillet, J. Lopez et al., Anti-NKG2A mAb Is a Checkpoint Inhibitor that Promotes Anti-tumor Immunity by Unleashing Both T and NK Cells, Cell, vol.175, pp.1731-1743, 2018.

. Hu-z, Overcome the Impairment of NK Cells for Icon and Antibody Immunotherapy of Cancer, J Immune Based Ther Vaccines Antimicrob, vol.2, pp.1-8, 2013.

M. Carlsten, N. Korde, R. Kotecha, R. Reger, S. Bor et al., Checkpoint Inhibition of KIR2D with the Monoclonal Antibody IPH2101 Induces Contraction and Hyporesponsiveness of NK Cells in Patients with Myeloma, Clin Cancer Res, vol.22, pp.5211-5222, 2016.

M. Giuliani, B. Janji, and G. Berchem, Activation of NK cells and disruption of PD-L1/PD-1 axis: two different ways for lenalidomide to block myeloma progression, Oncotarget, vol.8, pp.24031-24044, 2017.

D. N. Vo, A. C. Allende-vega, N. Morschhauser, F. Houot, R. Menard et al., NK cell activation and recovery of NK cell subsets in lymphoma patients after obinutuzumab and lenalidomide treatment, Oncoimmunology, vol.7, p.1409322, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01780378

K. Lagrue, A. Carisey, D. J. Morgan, R. Chopra, and D. M. Davis, Lenalidomide augments actin remodeling and lowers NK-cell activation thresholds, Blood, vol.126, pp.50-60, 2015.

E. Krzywinska, N. Allende-vega, A. Cornillon, D. N. Vo, L. Cayrefourcq et al., Identification of anti tumor cells carrying natural killer (NK) cell antigens in patients with hematological cancers, EBioMedicine, vol.2, pp.1364-1376, 2015.
URL : https://hal.archives-ouvertes.fr/inserm-01867114

E. Krzywinska, A. Cornillon, N. Allende-vega, D. N. Vo, R. C. Lu et al., CD45 Isoform Profile Identifies Natural Killer (NK) Subsets with Differential Activity, PLoS One, vol.11, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01867110

M. Villalba, N. Lopez-royuela, E. Krzywinska, M. G. Rathore, R. A. Hipskind et al., Chemical metabolic inhibitors for the treatment of blood-borne cancers, Anticancer Agents Med Chem, vol.14, pp.223-255, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-01867009

M. Villalba, M. G. Rathore, N. Lopez-royuela, E. Krzywinska, J. Garaude et al., From tumor cell metabolism to tumor immune escape, Int J Biochem Cell Biol, vol.45, pp.106-119, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00726717

C. Baier, A. Fino, C. Sanchez, L. Farnault, P. Rihet et al., Natural Killer Cells Modulation in Hematological Malignancies, Front Immunol, vol.4, p.459, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01455668

D. H. Mckenna, D. Sumstad, N. Bostrom, D. M. Kadidlo, S. Fautsch et al., Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience, Transfusion, vol.47, pp.520-528, 2007.

J. S. Miller, Y. Soignier, A. Panoskaltsis-mortari, S. A. Mcnearney, G. H. Yun et al., Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer, Blood, vol.105, pp.3051-3058, 2005.

L. Ruggeri, A. Mancusi, E. Burchielli, F. Aversa, M. F. Martelli et al., Natural killer cell alloreactivity in allogeneic hematopoietic transplantation, Curr Opin Oncol, vol.19, pp.142-149, 2007.

J. E. Rubnitz, H. Inaba, R. C. Ribeiro, S. Pounds, B. Rooney et al., NKAML: a pilot study to determine the safety and feasibility of haploidentical natural killer cell transplantation in childhood acute myeloid leukemia, J Clin Oncol, vol.28, pp.955-964, 2010.

D. H. Mckenna, D. Sumstad, N. Bostrom, D. M. Kadidlo, S. Fautsch et al.,

R. Dewaard, P. B. Mcglave, D. J. Weisdorf, and J. E. Wagner, Good manufacturing practices production of natural killer cells for immunotherapy: a six-year single-institution experience, Transfusion, vol.47, pp.520-528, 2007.

H. N. Haspels, M. A. Rahman, J. V. Joseph, G. Navarro, A. Chekenya et al., Glioblastoma Stem-Like Cells Are More Susceptible Than Differentiated Cells to Natural Killer Cell Lysis Mediated Through Killer Immunoglobulin-Like Receptors-Human Leukocyte Antigen Ligand Mismatch and Activation Receptor-Ligand Interactions, Int J Biochem Cell Biol, vol.9, pp.42-52, 2014.

D. Sanchez-martinez, G. Azaceta, A. Muntasell, N. Aguilo, D. Nunez et al., Human NK cells activated by EBV lymphoblastoid cells overcome anti-apoptotic mechanisms of drug resistance in haematological cancer cells, Activated Allogeneic NK Cells Preferentially Kill Poor Prognosis B-Cell Chronic Lymphocytic Leukemia Cells. Front Immunol, vol.4, p.454, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01835574

C. Chabannon, B. Mfarrej, S. Guia, S. Ugolini, R. Devillier et al., Manufacturing Natural Killer Cells as Medicinal Products, Front Immunol, vol.7, p.504, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01438165

J. P. Veluchamy, N. Kok, H. J. Van-der-vliet, H. Verheul, T. D. De-gruijl et al., The Rise of Allogeneic Natural Killer Cells As a Platform for Cancer Immunotherapy: Recent Innovations and Future Developments, Front Immunol, vol.8, p.631, 2017.

N. Shimasaki, E. Coustan-smith, T. Kamiya, and D. Campana, Expanded and armed natural killer cells for cancer treatment, Cytotherapy, vol.18, pp.1422-1434, 2016.

Y. Wu, Z. Tian, and H. Wei, Developmental and Functional Control of Natural Killer Cells by, Cytokines. Front Immunol, vol.8, p.930, 2017.

M. Imamura, D. Shook, T. Kamiya, N. Shimasaki, S. Chai et al., Autonomous growth and increased cytotoxicity of natural killer cells expressing membrane-bound interleukin-15, Blood, vol.124, pp.1081-1088, 2014.

J. L. Oyer, V. Pandey, R. Y. Igarashi, S. S. Somanchi, A. Zakari et al., Natural killer cells stimulated with PM21 particles expand and biodistribute in vivo: Clinical implications for cancer treatment, Cytotherapy, vol.18, pp.653-663, 2016.

C. J. Denman, V. V. Senyukov, S. S. Somanchi, P. V. Phatarpekar, L. M. Kopp et al., Membrane-Bound IL-21 Promotes Sustained Ex Vivo Proliferation of Human Natural Killer Cells, Chambers AM, Lupo KB, Matosevic S. Tumor Microenvironment-Induced Immunometabolic Reprogramming of Natural Killer Cells. Front Immunol, vol.7, p.30264, 2012.

V. Huber, C. Camisaschi, A. Berzi, S. Ferro, L. Lugini et al., Cancer acidity: An ultimate frontier of tumor immune escape and a novel target of immunomodulation, Semin Cancer Biol, vol.43, pp.74-89, 2017.

S. Charni, G. De-bettignies, M. G. Rathore, J. I. Aguilo, P. J. Van-den-elsen et al., Oxidative phosphorylation induces de novo expression of the MHC class I in tumor cells through the ERK5 pathway, J Immunol, vol.185, pp.3498-503, 2010.
URL : https://hal.archives-ouvertes.fr/hal-02193457

E. Catalán, S. Charni, P. Jaime, J. Aguiló, J. Enríquez et al., MHC-I modulation due to metabolic changes regulates tumor sensitivity to CTL and NK cells, Oncoimmunology, vol.4, p.985924, 2015.

M. Grudzien and A. Rapak, Effect of Natural Compounds on NK Cell Activation, J Immunol Res, p.4868417, 2018.

C. H. Kim, Control of Innate and Adaptive Lymphocytes by the RAR-Retinoic Acid Axis, Immune Netw, vol.18, p.1, 2018.

X. Fan, P. Wang, C. Zhang, Y. Zhang, Y. Fu et al., All-trans retinoic acid enhances cytotoxicity of CIK cells against human lung adenocarcinoma by upregulating MICA and IL-2 secretion, Sci Rep, vol.7, p.16481, 2017.

M. Oliveira-l-de, F. Teixeira, and M. N. Sato, Impact of Retinoic Acid on Immune Cells and Inflammatory Diseases, Mediators Inflamm, 2018.

C. Lo-nigro, M. Macagno, D. Sangiolo, L. Bertolaccini, M. Aglietta et al., NKmediated antibody-dependent cell-mediated cytotoxicity in solid tumors: biological evidence and clinical perspectives, Ann Transl Med, vol.7, p.105, 2019.

H. Michaud, J. Eliaou, V. Lafont, N. Bonnefoy, and L. Gros, Tumor antigen-targeting monoclonal antibody-based immunotherapy: Orchestrating combined strategies for the development of long-term antitumor immunity, Oncoimmunology, vol.3, p.955684, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02181343

G. Cartron, L. Dacheux, G. Salles, P. Solal-celigny, P. Bardos et al., Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene, Blood, vol.99, pp.754-762, 2002.

J. D. Mellor, M. P. Brown, H. R. Irving, J. R. Zalcberg, and A. Dobrovic, A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer, J Hematol OncolJ Hematol Oncol, vol.6, p.1, 2013.

G. Cartron and H. Watier, Obinutuzumab: what is there to learn from clinical trials?, Blood, vol.130, pp.581-589, 2017.

X. Song, S. Hong, W. T. Kwon, L. M. Bailey, P. Basse et al.,

T. Secretory, Armed Natural Killer Cell-Based Therapy: In Vitro and In Vivo Colorectal Peritoneal Carcinomatosis Xenograft, Mol Cancer Ther, vol.15, pp.1591-1601, 2016.

L. A. Fogel, W. M. Yokoyama, and A. R. French, Natural killer cells in human autoimmune disorders, Arthritis Res Ther, vol.15, p.216, 2013.

N. Dalbeth, M. Callan, R. Yamin, O. Berhani, H. Peleg et al., High percentages and activity of synovial fluid NK cells present in patients with advanced stage active Rheumatoid Arthritis, Pathogenic Role of Immune Cells in Rheumatoid Arthritis: Implications in Clinical Treatment and Biomarker Development. Cells, vol.46, p.1351, 2002.

K. Takahashi, S. Miyake, T. Kondo, K. Terao, M. Hatakenaka et al.,

T. , Natural killer type 2 bias in remission of multiple sclerosis, J Clin Invest, vol.107, pp.23-29, 2001.

I. Hamann, J. Dörr, R. Glumm, C. Chanvillard, A. Janssen et al., Characterization of natural killer cells in paired CSF and blood samples during neuroinflammation, J Neuroimmunol, vol.254, pp.165-169, 2013.

B. Bielekova, M. Catalfamo, S. Reichert-scrivner, A. Packer, M. Cerna et al., Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis, Proc Natl Acad Sci, vol.103, pp.5941-5946, 2006.

D. Schepis, I. Gunnarsson, M. Eloranta, J. Lampa, S. H. Jacobson et al., Tumor-Primed Human Natural Killer Cells Lyse NK-Resistant Tumor Targets: Evidence of a Two-Stage Process in Resting NK Cell Activation, J Rheumatol, vol.126, pp.605-619, 1996.

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.991-999, 2002.

M. Lopez-botet, C. Vilches, D. Redondo-pachon, A. Muntasell, A. Pupuleku et al., Dual Role of Natural Killer Cells on Graft Rejection and Control of Cytomegalovirus Infection in Renal Transplantation, Front Immunol, vol.8, p.166, 2017.

M. Gumá, M. Budt, A. Sáez, T. Brckalo, H. Hengel et al., Expansion of CD94/NKG2C+ NK cells in response to human cytomegalovirus-infected fibroblasts, Blood, vol.107, pp.3624-3631, 2006.

A. Rölle, J. Pollmann, E. Ewen, V. Le, A. Halenius et al., IL-12-producing monocytes and HLA-E control HCMV-driven NKG2C+ NK cell expansion, J Clin Invest, vol.124, pp.5305-5316, 2014.

H. Peng, Z. Tian, J. Pahl, A. Cerwenka, and J. Ni, Memory-Like NK Cells: Remembering a Previous Activation by Cytokines and NK Cell Receptors, Front Immunol, vol.8, p.1143, 2017.

C. Capuano, S. Battella, C. Pighi, L. Franchitti, O. Turriziani et al., Tumor-Targeting Anti-CD20 Antibodies Mediate In Vitro Expansion of Memory Natural Killer Cells: Impact of CD16 Affinity Ligation Conditions and In Vivo Priming, Front Immunol, vol.9, 2018.

H. Klingemann, L. Boissel, and F. Toneguzzo, Natural Killer Cells for Immunotherapy -Advantages of the NK-92 Cell Line over, Blood NK Cells. Front Immunol, vol.7, 2016.

Y. Yan, P. Steinherz, H. G. Klingemann, D. Dennig, B. H. Childs et al.,

, Antileukemia activity of a natural killer cell line against human leukemias, Clin Cancer Res, vol.4, pp.2859-2868, 1998.

B. Clémenceau, R. Vivien, C. Pellat, M. Foss, G. Thibault et al., The human natural killer cytotoxic cell line NK-92, once armed with a murine CD16 receptor, represents a convenient cellular tool for the screening of mouse mAbs according to their ADCC potential, mAbs, vol.5, pp.587-594, 2013.

B. A. Williams, A. D. Law, B. Routy, N. Denhollander, V. Gupta et al., A phase I trial of NK-92 cells for refractory hematological malignancies relapsing after autologous hematopoietic cell transplantation shows safety and evidence of efficacy, Oncotarget, vol.8, pp.89256-89268, 2017.