A. Monnereau, Estimation nationale de l'incidence des cancers en France entre 1980 et 2012

J. M. Bennett, Proposals for the Classification of the Acute Leukaemias FrenchAmerican-British (FAB) Co-operative Group, British journal of haematology, vol.33, pp.451-458, 1976.

D. A. Arber, The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood, vol.127, pp.2391-2405, 2016.

N. Howlader, Cancer Statistics Review, 1975.

H. Döhner, Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel, Blood, vol.129, pp.424-447, 2017.

W. Li, High-dose cytarabine in acute myeloid leukemia treatment: a systematic review and meta-analysis, PLoS ONE, vol.9, p.110153, 2014.

A. Gratwohl, Cause of death after allogeneic haematopoietic stem cell transplantation (HSCT) in early leukaemias: an EBMT analysis of lethal infectious complications and changes over calendar time, Bone Marrow Transplant, vol.36, pp.757-769, 2005.

B. O'sullivan, The TNM classification of malignant tumours-towards common understanding and reasonable expectations, Lancet Oncol, vol.18, pp.849-851, 2017.

J. Fourcade, CD8(+) T cells specific for tumor antigens can be rendered dysfunctional by the tumor microenvironment through upregulation of the inhibitory receptors BTLA and PD-1, Cancer Res, vol.72, pp.887-896, 2012.

J. Galon, Type, density, and location of immune cells within human colorectal tumors predict clinical outcome, Science, vol.313, pp.1960-1964, 2006.

J. Galon, Immunoscore and Immunoprofiling in cancer: an update from the melanoma and immunotherapy bridge 2015, J Transl Med, vol.14, p.273, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388016

L. Fend, Prognostic impact of the expression of NCR1 and NCR3 NK cell receptors and PD-L1 on advanced non-small cell lung cancer, Oncoimmunology, vol.6, p.1163456, 2017.

A. J. Gentles, The prognostic landscape of genes and infiltrating immune cells across human cancers, Nat Med, vol.21, pp.938-945, 2015.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, p.646

R. D. Schreiber, L. J. Old, and M. J. Smyth, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion, Science, vol.331, pp.1565-1570, 2011.

G. Dranoff, Cytokines in cancer pathogenesis and cancer therapy, Nat. Rev. Cancer, vol.4, pp.11-22, 2004.

A. M. Dickinson, Graft-versus-Leukemia Effect Following Hematopoietic Stem Cell Transplantation for Leukemia, Front Immunol, vol.8, 2017.

A. Cossarizza, Guidelines for the use of flow cytometry and cell sorting in immunological studies, Eur. J. Immunol, vol.47, pp.1584-1797, 2017.
URL : https://hal.archives-ouvertes.fr/pasteur-01619848

H. Kared, S. Martelli, T. P. Ng, S. L. Pender, and A. Larbi, CD57 in human natural killer cells and T-lymphocytes, vol.65, pp.441-452, 2016.

E. Vivier, S. Ugolini, D. Blaise, C. Chabannon, and L. Brossay, Targeting natural killer cells and natural killer T cells in cancer, Nature Reviews Immunology, vol.12, pp.239-252, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00685473

M. G. Morvan and L. L. Lanier, NK cells and cancer: you can teach innate cells new tricks, Nat. Rev. Cancer, vol.16, pp.7-19, 2016.

L. Moretta, Cellular and molecular basis of natural killer and natural killer-like activity, Immunol. Lett, vol.88, pp.89-93, 2003.

S. Sivori, p46, a Novel Natural Killer Cell-specific Surface Molecule That Mediates Cell Activation, J Exp Med, vol.186, pp.1129-1136, 1997.

D. Pende, Identification and molecular characterization of NKp30, a novel triggering receptor involved in natural cytotoxicity mediated by human natural killer cells, J. Exp

. Med, , vol.190, pp.1505-1516, 1999.

M. Vitale, NKp44, a novel triggering surface molecule specifically expressed by activated natural killer cells, is involved in non-major histocompatibility complex-restricted tumor cell lysis, J. Exp. Med, vol.187, pp.2065-2072, 1998.

P. H. Kruse, J. Matta, S. Ugolini, and E. Vivier, Natural cytotoxicity receptors and their ligands, Immunol. Cell Biol, vol.92, pp.221-229, 2014.

J. Matta, Induction of B7-H6, a ligand for the natural killer cell-activating receptor NKp30, in inflammatory conditions, Blood, vol.122, pp.394-404, 2013.

B. Rosental, U. Hadad, M. Brusilovsky, K. S. Campbell, and A. Porgador, A novel mechanism for cancer cells to evade immune attack by NK cells: The interaction between NKp44 and proliferating cell nuclear antigen, Oncoimmunology, vol.1, pp.572-574, 2012.

F. Baychelier, Identification of a cellular ligand for the natural cytotoxicity receptor NKp44, Blood, vol.122, pp.2935-2942, 2013.

T. Walzer, Identification, activation, and selective in vivo ablation of mouse NK cells via NKp46, Proc Natl Acad Sci U S A, vol.104, pp.3384-3389, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00165486

L. Chiche, The role of natural killer cells in sepsis, J. Biomed. Biotechnol, p.986491, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00611407

K. Imai, S. Matsuyama, S. Miyake, K. Suga, and K. Nakachi, Natural cytotoxic activity of peripheral-blood lymphocytes and cancer incidence: an 11-year follow-up study of a general population, The Lancet, vol.356, pp.1795-1799, 2000.

E. Mamessier, Human breast cancer cells enhance self tolerance by promoting evasion from NK cell antitumor immunity, J. Clin. Invest, vol.121, pp.3609-3622, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01765654

C. Pasero, Inherent and Tumor-Driven Immune Tolerance in the Prostate Microenvironment Impairs Natural Killer Cell Antitumor Activity, Cancer Res, vol.76, pp.2153-2165, 2016.

C. Pasero, Highly effective NK cells are associated with good prognosis in patients with metastatic prostate cancer, Oncotarget, vol.6, pp.14360-14373, 2015.

R. T. Costello, Defective expression and function of natural killer cell-triggering receptors in patients with acute myeloid leukemia, Blood, vol.99, pp.3661-3667, 2002.

S. Sivori, NKp46 is the major triggering receptor involved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation between surface density of NKp46 and natural cytotoxicity against autologous, allogeneic or xenogeneic target cells, Eur. J. Immunol, vol.29, pp.1656-1666, 1999.

C. Fauriat, Deficient expression of NCR in NK cells from acute myeloid leukemia: Evolution during leukemia treatment and impact of leukemia cells in NCRdull phenotype induction, Blood, vol.109, pp.323-330, 2007.

A. Chretien, NKp30 expression is a prognostic immune biomarker for stratification of patients with intermediate-risk acute myeloid leukemia, Oncotarget, 2017.

A. Chretien, NKp46 expression on NK cells as a prognostic and predictive biomarker for response to allo-SCT in patients with AML, Oncoimmunology, vol.6, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01789566

A. Chretien, Increased NK Cell Maturation in Patients with, Acute Myeloid Leukemia. Front Immunol, vol.6, p.564, 2015.

A. Chretien, Natural Killer Defective Maturation Is Associated with Adverse Clinical Outcome in Patients with, Acute Myeloid Leukemia. Front Immunol, vol.8, p.573, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01568363

L. Chen and D. B. Flies, Molecular mechanisms of T cell co-stimulation and co-inhibition

, Nat. Rev. Immunol, vol.13, pp.227-242, 2013.

S. M. Kaech and W. Cui, Transcriptional control of effector and memory CD8+ T cell differentiation, Nat Rev Immunol, vol.12, pp.749-761, 2012.

T. Willinger, T. Freeman, H. Hasegawa, A. J. Mcmichael, and M. F. Callan, Molecular signatures distinguish human central memory from effector memory CD8 T cell subsets, J. Immunol, vol.175, pp.5895-5903, 2005.

S. Jiang and C. Dong, A complex issue on CD4(+) T-cell subsets, Immunol. Rev, vol.252, pp.5-11, 2013.

D. Mougiakakos, A. Choudhury, A. Lladser, R. Kiessling, and C. C. Johansson, Regulatory T cells in cancer, Adv. Cancer Res, vol.107, pp.57-117, 2010.

G. J. Bates, Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse, J. Clin. Oncol, vol.24, pp.5373-5380, 2006.

Z. Shen, Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer, J. Cancer Res. Clin. Oncol, vol.136, pp.1585-1595, 2010.

D. Wolf, The expression of the regulatory T cell-specific forkhead box transcription factor FoxP3 is associated with poor prognosis in ovarian cancer, Clin. Cancer Res, vol.11, pp.8326-8331, 2005.

H. Tao, Prognostic potential of FOXP3 expression in non-small cell lung cancer cells combined with tumor-infiltrating regulatory T cells, Lung Cancer, vol.75, pp.95-101, 2012.

J. Jang, Crosstalk between Regulatory T Cells and Tumor-Associated Dendritic Cells Negates Anti-tumor Immunity in Pancreatic Cancer, Cell Rep, vol.20, pp.558-571, 2017.

C. Ustun, J. S. Miller, D. H. Munn, D. J. Weisdorf, and B. R. Blazar, Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation?, Blood, vol.118, pp.5084-5095, 2011.

Z. Shenghui, Elevated frequencies of CD4 + CD25 + CD127lo regulatory T cells is associated to poor prognosis in patients with acute myeloid leukemia, Int. J. Cancer, vol.129, pp.1373-1381, 2011.

F. Benchetrit, Les lymphocytes T CD8 : rôle dans l'immunosurveillance et l'immunothérapie antitumorale, Bulletin du Cancer, vol.90, pp.677-685, 2003.

V. Catros, Lymphocytes T?? en cancérologie-Des lymphocytes tueurs non conventionnels, Med Sci, vol.26, pp.185-192
DOI : 10.1051/medsci/2010262185

URL : https://www.medecinesciences.org/articles/medsci/pdf/2010/03/medsci2010262p185.pdf

D. Montagna, Emergence of antitumor cytolytic T cells is associated with maintenance of hematologic remission in children with acute myeloid leukemia, Blood, vol.108, pp.3843-3850, 2006.

D. Ozkazanc, D. Yoyen-ermis, E. Tavukcuoglu, Y. Buyukasik, and G. Esendagli, Functional exhaustion of CD4(+) T cells induced by co-stimulatory signals from myeloid leukaemia cells, Immunology, vol.149, pp.460-471, 2016.

D. R. Bandura, Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry

, Chem, vol.81, pp.6813-6822, 2009.

S. C. Bendall, Single-Cell Mass Cytometry of Differential Immune and Drug Responses Across a Human Hematopoietic Continuum, Science, vol.332, pp.687-696, 2011.

K. I. Fread, W. D. Strickland, G. P. Nolan, and E. R. Zunder, An updated debarcoding tool for mass cytometry with cell type-specific and cell sample-specific stringency adjustment

, Pac Symp Biocomput, vol.22, pp.588-598, 2016.

R. Finck, Normalization of mass cytometry data with bead standards, Cytometry A, vol.83, pp.483-494, 2013.

A. Muntasell, Targeting NK-cell checkpoints for cancer immunotherapy, Curr. Opin. Immunol, vol.45, pp.73-81, 2017.

E. J. Wherry, T cell exhaustion, Nat. Immunol, vol.12, pp.492-499, 2011.

M. Hashimoto, CD8 T Cell Exhaustion in Chronic Infection and Cancer: Opportunities for Interventions, Annu. Rev. Med, vol.69, pp.301-318, 2018.

S. L. Topalian, Safety, activity, and immune correlates of anti-PD-1 antibody in cancer, N. Engl. J. Med, vol.366, pp.2443-2454, 2012.

S. M. Ansell, PD-1 blockade with nivolumab in relapsed or refractory Hodgkin's lymphoma, N. Engl. J. Med, vol.372, pp.311-319, 2015.

G. M. Mason, Phenotypic Complexity of the Human Regulatory T Cell Compartment Revealed by Mass Cytometry, J. Immunol, vol.195, pp.2030-2037, 2015.

S. Kordasti, Deep phenotyping of Tregs identifies an immune signature for idiopathic aplastic anemia and predicts response to treatment, Blood, vol.128, pp.1193-1205, 2016.

, Annexe 2 : Stratégie de fenêtrage en cytométrie de flux (BD FACSCanto II®)

, On sélectionne les lymphocytes en fonction de leur taille et de leur granulosité sur le graphe SSC/FSC qui n'existe pas en cytométrie de masse, puis on sélectionne les CD45 + , puis les NK sont définis par l'expression de CD56 et l'absence de CD3. On peut ensuite étudier l'expression de NKp46, comme en cytométrie de masse