J. R. Passweg, Is the use of unrelated donor transplantation leveling off in Europe? The, 2016.

, European Society for Blood and Marrow Transplant activity survey report, Bone Marrow Transplant, vol.53, pp.1139-1148, 2018.

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

E. Hatzimichael and M. Tuthill, Hematopoietic stem cell transplantation, Stem Cells Cloning, vol.3, pp.105-117, 2010.

J. Tiercy, How to select the best available related or unrelated donor of hematopoietic stem cells?, Haematologica, vol.101, pp.680-687, 2016.

R. L. Powles, Mismatched family donors for bone-marrow transplantation as treatment for acute leukaemia, Lancet, vol.1, pp.612-615, 1983.

X. Huang, Current status of haploidentical stem cell transplantation for leukemia, J Hematol Oncol, vol.1, p.27, 2008.

J. M. Rowe and M. S. Tallman, How I treat acute myeloid leukemia, Blood, vol.116, pp.3147-3156, 2010.

H. M. Vriesendorp, Aims of conditioning, Exp. Hematol, vol.31, pp.844-854, 2003.

E. P. Alyea, Impact of conditioning regimen intensity on outcome of allogeneic hematopoietic cell transplantation for advanced acute myelogenous leukemia and myelodysplastic syndrome, Biol. Blood Marrow Transplant, vol.12, pp.1047-1055, 2006.

R. Diaconescu, Morbidity and mortality with nonmyeloablative compared with myeloablative conditioning before hematopoietic cell transplantation from HLA-matched related donors, Blood, vol.104, pp.1550-1558, 2004.

M. Mielcarek, Graft-versus-host disease after nonmyeloablative versus conventional hematopoietic stem cell transplantation, Blood, vol.102, pp.756-762, 2003.

J. L. Ferrara, J. E. Levine, P. Reddy, and E. Holler, Graft-versus-host disease, Lancet, vol.373, pp.1550-1561, 2009.

S. Ghimire, Pathophysiology of GvHD and Other HSCT-Related Major Complications. Front Immunol, vol.8, p.79, 2017.

A. J. Barrett and M. Battiwalla, Relapse after allogeneic stem cell transplantation, Expert Rev Hematol, vol.3, pp.429-441, 2010.

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.

R. Glas, Recruitment and activation of natural killer (NK) cells in vivo determined by the target cell phenotype. An adaptive component of NK cell-mediated responses, J. Exp. Med, vol.191, pp.129-138, 2000.

Y. T. Bryceson, Functional analysis of human NK cells by flow cytometry, Methods Mol. Biol, vol.612, pp.335-352, 2010.

R. Jacobs, CD56bright cells differ in their KIR repertoire and cytotoxic features from CD56dim NK cells, Eur. J. Immunol, vol.31, pp.3121-3127, 2001.

M. A. Cooper, T. A. Fehniger, and M. A. Caligiuri, The biology of human natural killer-cell subsets, Trends Immunol, vol.22, pp.633-640, 2001.

E. Vivier, What is natural in natural killer cells?, Immunol. Lett, vol.107, pp.1-7, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00165603

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

M. E. Matos, Expression of a functional c-kit receptor on a subset of natural killer cells, J. Exp. Med, vol.178, pp.1079-1084, 1993.

A. C. Jaleco, Fetal liver contains committed NK progenitors, but is not a site for development of CD34+ cells into T cells, J. Immunol, vol.159, pp.694-702, 1997.

C. Vitale, Analysis of the activating receptors and cytolytic function of human natural killer cells undergoing in vivo differentiation after allogeneic bone marrow transplantation, Eur. J. Immunol, vol.34, pp.455-460, 2004.

E. Montaldo, Human NK cell receptors/markers: a tool to analyze NK cell development, subsets and function, Cytometry A, vol.83, pp.702-713, 2013.

A. Marçais, Regulation of mouse NK cell development and function by cytokines, Front Immunol, vol.4, p.450, 2013.

M. J. Smyth, Activation of NK cell cytotoxicity, Mol. Immunol, vol.42, pp.501-510, 2005.

E. Vivier, J. A. Nunès, and F. Vély, Natural killer cell signaling pathways, Science, vol.306, pp.1517-1519, 2004.
URL : https://hal.archives-ouvertes.fr/hal-00080580

F. Fang, W. Xiao, and . Tian, Z. NK cell-based immunotherapy for cancer. Semin. Immunol, vol.31, pp.37-54, 2017.

L. Chávez-galán, M. C. Angel, E. Zenteno, R. Chávez, and R. Lascurain, Cell death mechanisms induced by cytotoxic lymphocytes, Cell. Mol. Immunol, vol.6, pp.15-25, 2009.

G. Alter, J. M. Malenfant, and M. Altfeld, CD107a as a functional marker for the identification of natural killer cell activity, J. Immunol. Methods, vol.294, pp.15-22, 2004.

C. Fauriat, E. O. Long, H. 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.

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

R. Wang, J. J. Jaw, N. C. Stutzman, Z. Zou, and P. D. Sun, Natural killer cell-produced IFN-? and TNF-? induce target cell cytolysis through up-regulation of ICAM-1, J. Leukoc. Biol, vol.91, pp.299-309, 2012.

L. Wang, F. Du, and X. Wang, TNF-alpha induces two distinct caspase-8 activation pathways, Cell, vol.133, pp.693-703, 2008.

L. Chiossone, M. Vienne, Y. M. Kerdiles, and E. Vivier, Natural killer cell immunotherapies against cancer: checkpoint inhibitors and more, Seminars in Immunology, vol.31, pp.55-63, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01764667

L. L. Lanier, Natural killer cell receptor signaling, Curr. Opin. Immunol, vol.15, pp.308-314, 2003.

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.

D. H. Raulet, Roles of the NKG2D immunoreceptor and its ligands, Nat. Rev. Immunol, vol.3, pp.781-790, 2003.

S. Bauer, Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA, Science, vol.285, pp.727-729, 1999.

C. S. Brandt, The B7 family member B7-H6 is a tumor cell ligand for the activating natural killer cell receptor NKp30 in humans, J. Exp. Med, vol.206, pp.1495-1503, 2009.

E. Pogge-von-strandmann, Human leukocyte antigen-B-associated transcript 3 is released from tumor cells and engages the NKp30 receptor on natural killer cells, Immunity, vol.27, pp.965-974, 2007.

B. Rosental, Proliferating cell nuclear antigen is a novel inhibitory ligand for the natural cytotoxicity receptor NKp44, J. Immunol, vol.187, pp.5693-5702, 2011.

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

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.

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

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, p.1307491, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01789566

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.

C. Bottino, Identification of PVR (CD155) and Nectin-2 (CD112) as cell surface ligands for the human DNAM-1 (CD226) activating molecule, J. Exp. Med, vol.198, pp.557-567, 2003.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

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.

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, 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

R. Castriconi, Transforming growth factor beta 1 inhibits expression of NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells, Proc. Natl. Acad. Sci. U.S.A, vol.100, pp.4120-4125, 2003.

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.

A. Chretien, NKp30 expression is a prognostic immune biomarker for stratification of patients with intermediate-risk acute myeloid leukemia, Oncotarget, vol.8, pp.49548-49563, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01568368

F. Aversa, M. F. Martelli, and A. Velardi, Haploidentical hematopoietic stem cell transplantation with a megadose T-cell-depleted graft: harnessing natural and adaptive immunity, Semin. Oncol, vol.39, pp.643-652, 2012.

K. C. Hsu, Improved outcome in HLA-identical sibling hematopoietic stem-cell transplantation for acute myelogenous leukemia predicted by KIR and HLA genotypes, Blood, vol.105, pp.4878-4884, 2005.

L. Ruggeri, Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants, Science, vol.295, pp.2097-2100, 2002.

J. M. Venstrom, Donor activating KIR2DS1 in leukemia, N. Engl. J. Med, vol.371, p.2042, 2014.

J. M. Venstrom, Donor activating KIR3DS1 is associated with decreased acute GVHD in unrelated allogeneic hematopoietic stem cell transplantation, Blood, vol.115, pp.3162-3165, 2010.

J. Storek, Reconstitution of the immune system after hematopoietic stem cell transplantation in humans, Semin Immunopathol, vol.30, pp.425-437, 2008.

L. Ruggeri, Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants, Science, vol.295, issue.5562, pp.2097-100, 2002.

M. A. Ullah, Functional Reconstitution of Natural Killer Cells in Allogeneic Hematopoietic Stem Cell Transplantation, Front Immunology, vol.7, p.144, 2016.

C. Pical-izard, Reconstitution of natural killer cells in HLA-matched HSCT after reducedintensity conditioning: impact on clinical outcome, Biol Blood Marrow Transplant, pp.429-468, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01108575

J. M. Palmer, Clinical relevance of natural killer cells following hematopoietic stem cell transplantation, Journal of Cancer, vol.4, issue.1, pp.25-35, 2013.

U. Koehl, Clinical grade purification and expansion of NK cell products for an optimized manufacturing protocol, Front Oncol, vol.3, p.118, 2013.

R. W. Childs and M. Carlsten, Therapeutic approaches to enhance natural killer cell cytotoxicity against cancer: The force awakens, Nature Reviews Drug Discovery, vol.14, pp.487-498, 2015.

R. Ghasemi, Selective targeting of IL-2 to NKG2D bearing cells for improved immunotherapy, Nat Commun, vol.7, p.12878, 2016.

T. Floros and A. A. Tarhini, Anticancer Cytokines: Biology and Clinical Effects of Interferon-?2, Interleukin (IL)-2, IL-15, IL-21, and IL-12, Semin. Oncol, vol.42, pp.539-548, 2015.

C. De-rham, The proinflammatory cytokines IL-2, IL-15 and IL-21 modulate the repertoire of mature human natural killer cell receptors, Arthritis Research and Therapy, vol.9, pp.1-15, 2007.

A. Prica and M. Crump, Improving CD20 antibody therapy: obinutuzumab in lymphoproliferative disorders, Leuk. Lymphoma, vol.1, p.10, 2019.

S. O. Ciurea, Phase 1 clinical trial using mbIL21 ex vivo-expanded donor-derived NK cells after haploidentical transplantation, Blood, vol.130, pp.1857-1868, 2017.

D. A. Knorr, V. Bachanova, M. R. Verneris, and J. Miller, Clinical utility of natural killer cells in cancer therapy and transplantation, Semin. Immunol, vol.26, pp.161-172, 2014.

S. A. Rosenberg, Observations on the systemic administration of autologous lymphokineactivated killer cells and recombinant interleukin-2 to patients with metastatic cancer, N. Engl. J. Med, vol.313, pp.1485-1492, 1985.

J. S. Miller, Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer, Blood, vol.105, pp.3051-3057, 2005.

R. S. Mehta, E. J. Shpall, and K. Rezvani, Cord Blood as a Source of Natural Killer Cells, Front Med (Lausanne), vol.2, p.93, 2015.

C. Eguizabal, Natural killer cells for cancer immunotherapy: pluripotent stem cells-derived NK cells as an immunotherapeutic perspective, Front Immunol, vol.5, p.439, 2014.

Y. Li, D. L. Hermanson, B. S. Moriarity, and D. S. Kaufman, Human iPSC-Derived Natural Killer Cells Engineered with Chimeric Antigen Receptors Enhance Anti-tumor Activity, Cell Stem Cell, vol.23, pp.181-192, 2018.

M. Carlsten, Clinical-Grade mRNA Electroporation of NK Cells: A Novel and Highly Efficient Method to Genetically Reprogram Human NK Cells for Cancer Immunotherapy, Blood, vol.124, p.2153, 2014.

B. C. Shaffer, Phase II Study of Haploidentical Natural Killer Cell Infusion for Treatment of Relapsed or Persistent Myeloid Malignancies Following Allogeneic Hematopoietic Cell Transplantation, Biol. Blood Marrow Transplant, vol.22, pp.705-709, 2016.

A. Curti, Successful transfer of alloreactive haploidentical KIR ligand-mismatched natural killer cells after infusion in elderly high risk acute myeloid leukemia patients, Blood, vol.118, pp.3273-3279, 2011.

E. K. Kim, Ex vivo activation and expansion of natural killer cells from patients with advanced cancer with feeder cells from healthy volunteers, Cytotherapy, vol.15, pp.231-241, 2013.

A. M. Levin, Exploiting a natural conformational switch to engineer an interleukin-2 'superkine', Nature, vol.484, pp.529-533, 2012.

A. Lundqvist, M. Berg, A. Smith, and R. W. Childs, Bortezomib treatment to potentiate the antitumor immunity of ex-vivo expanded adoptively infused autologous natural killer cells, Journal of Cancer, vol.2, pp.383-385, 2011.

R. W. Childs and M. Berg, Bringing natural killer cells to the clinic: ex vivo manipulation

, Hematology, pp.234-246, 2013.

. Sharma, The Future of Immune Checkpoint 4, vol.348, 2015.

F. Romagné, Preclinical characterization of 1-7F9, a novel human anti-KIR receptor therapeutic antibody that augments natural killer-mediated killing of tumor cells, Blood, vol.114, pp.2667-2677, 2009.

V. M. Braud, HLA-E binds to natural killer cell receptors CD94/NKG2A, B and C, Nature, vol.391, pp.795-799, 1998.

M. López-botet, M. Llano, and M. Ortega, Human cytomegalovirus and natural killer-mediated surveillance of HLA class I expression: a paradigm of host-pathogen adaptation, Immunol. Rev, vol.181, pp.193-202, 2001.

S. Nguyen, HLA-E upregulation on IFN-?-activated AML blasts impairs CD94/ NKG2A-dependent NK cytolysis after haplo-mismatched hematopoietic SCT, Bone Marrow Transplantation, vol.43, pp.693-699, 2009.

T. Yazdi and M. , The positive prognostic effect of stromal CD8+ tumor-infiltrating T cells is restrained by the expression of HLA-E in non-small cell lung carcinoma, Oncotarget, vol.7, pp.3477-3488, 2016.

S. Pesce, Identification of a subset of human natural killer cells expressing high levels of programmed death 1: A phenotypic and functional characterization, Journal of Allergy and Clinical Immunology, vol.139, pp.335-346, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01788010

R. Das, Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo, J. Immunol, vol.194, pp.950-959, 2015.

A. C. Anderson, Tim-3: an emerging target in the cancer immunotherapy landscape, Cancer Immunol Res, vol.2, pp.393-398, 2014.

Q. Zhou, Coexpression of Tim-3 and PD-1 identifies a CD8 ? T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia, Blood, vol.117, pp.4501-4510, 2013.

C. Fauriat, Estimation of the Size of the Alloreactive NK Cell Repertoire: Studies in Individuals Homozygous for the Group A KIR Haplotype, The Journal of Immunology, vol.181, pp.6010-6019, 2008.

A. Roberto, The early expansion of anergic NKG2Apos/CD56dim/CD16neg natural killer cells represents a therapeutic target in haploidentical haematopoietic stem cell transplantation. Haematologica haematol, p.186619, 2017.

C. Pical-izard, Reconstitution of Natural Killer Cells in HLA-Matched HSCT after Reduced-Intensity Conditioning: Impact on Clinical Outcome, Biology of Blood and Marrow Transplantation, vol.21, pp.429-439, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01108575

S. Nguyen, NK-cell reconstitution after haploidentical hematopoietic stem-cell transplantations: Immaturity of NK cells and inhibitory effect of NKG2A override GvL effect, Blood, vol.105, pp.4135-4142, 2005.

C. Veuillen, Primary B-CLL resistance to NK cell cytotoxicity can be overcome in vitro and in vivo by priming NK cells and monoclonal antibody therapy, Journal of Clinical Immunology, vol.32, pp.632-646, 2012.