I. Bibliographie-1, T. Reya, S. Morrison, M. Clarke, and I. Weissman, Stem cells, cancer, and cancer stem cells, Nature, 2001.

O. Beyne-rauzy, Tumor necrosis factor-?? inhibits hTERT gene expression in human myeloid normal and leukemic cells, Blood, vol.106, issue.9, pp.3200-3205, 2005.
DOI : 10.1182/blood-2005-04-1386

F. Rosenbauer and D. Tenen, Transcription factors in myeloid development: balancing differentiation with transformation, Nature Reviews Immunology, vol.14, issue.2, pp.105-122, 2007.
DOI : 10.1038/nri2024

B. Huntly and D. Gilliland, Timeline: Leukaemia stem cells and the evolution of cancer-stem-cell research, Nature Reviews Cancer, vol.91, issue.4, pp.311-332, 2005.
DOI : 10.1038/nm1296-1329

S. Orkin and L. Zon, SnapShot: hematopoiesis. Cell, Feb, vol.22132, issue.4, p.712, 2008.
DOI : 10.1016/j.cell.2008.02.013

URL : http://doi.org/10.1016/j.cell.2008.02.013

N. Nardi and Z. Alfonso, The hematopoietic stroma, Brazilian Journal of Medical and Biological Research, vol.32, issue.5, pp.601-610, 1999.
DOI : 10.1590/S0100-879X1999000500014

T. Lapidot, C. Sirard, J. Vormoor, B. Murdoch, T. Hoang et al., A cell initiating human acute myeloid leukaemia after transplantation into SCID mice, Nature, vol.367, issue.6464, pp.645-653, 1994.
DOI : 10.1038/367645a0

D. Bonnet and J. Dick, Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell, Nature Medicine, vol.6, issue.7, pp.730-737, 1997.
DOI : 10.1038/367645a0

D. Figueiredo-pontes, L. Pintão, M. Oliveira, L. Dalmazzo, L. Jácomo et al., Determination of P-glycoprotein, MDR-related protein 1, breast cancer resistance protein, and lung-resistance protein expression in leukemic stem cells of acute myeloid leukemia, Cytometry Part B: Clinical Cytometry, vol.128, issue.3, 2008.
DOI : 10.1002/cyto.b.20403

C. Grove and G. Vassiliou, Acute myeloid leukaemia: a paradigm for the clonal evolution of cancer? Dis Model Mech, pp.941-51, 2014.

M. Lawrence, P. Stojanov, P. Polak, G. Kryukov, K. Cibulskis et al., Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature The Cancer Genome Atlas Research Network. Genomic and Epigenomic Landscapes of Adult De Novo Acute Myeloid Leukemia, N Engl J Med, vol.499368, issue.745722, pp.214-222, 2013.

D. Gilliland and J. Griffin, The roles of FLT3 in hematopoiesis and leukemia, Blood, vol.100, issue.5, pp.1532-1574, 2002.
DOI : 10.1182/blood-2002-02-0492

L. Ding, T. Ley, D. Larson, C. Miller, D. Koboldt et al., Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing, Nature, vol.115, issue.7382, pp.506-516, 2012.
DOI : 10.1038/nature10738

Z. Reitman and H. Yan, Isocitrate Dehydrogenase 1 and 2 Mutations in Cancer: Alterations at a Crossroads of Cellular Metabolism, JNCI Journal of the National Cancer Institute, vol.102, issue.13, pp.932-973, 2010.
DOI : 10.1093/jnci/djq187

K. Yen, M. Bittinger, S. Su, and V. Fantin, Cancer-associated IDH mutations: biomarker and therapeutic opportunities, Oncogene, vol.324, issue.49, pp.6409-6426, 2009.
DOI : 10.1038/onc.2010.444

L. Dang, J. S. Su, and S. , IDH mutations in glioma and acute myeloid leukemia, Trends in Molecular Medicine, vol.16, issue.9, 2010.
DOI : 10.1016/j.molmed.2010.07.002

T. Sjöblom, S. Jones, L. Wood, D. Parsons, J. Lin et al., The consensus coding sequences of human breast and colorectal cancers. Science An integrated genomic analysis of human glioblastoma multiforme. Science, pp.268-741807, 2006.

E. Mardis, L. Ding, D. Dooling, D. Larson, M. Mclellan et al., Recurring Mutations Found by Sequencing an Acute Myeloid Leukemia Genome, New England Journal of Medicine, vol.361, issue.11, pp.1058-66, 2009.
DOI : 10.1056/NEJMoa0903840

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3201812

S. Gross, R. Cairns, M. Minden, E. Driggers, M. Bittinger et al., Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations, The Journal of Experimental Medicine, vol.64, issue.2
DOI : 10.1126/science.1170944

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2822606

G. Marcucci, K. Maharry, Y. Wu, M. Radmacher, K. Mrózek et al., Gene Mutations Identify Novel Molecular Subsets Within De Novo Cytogenetically Normal Acute Myeloid Leukemia: A Cancer and Leukemia Group B Study, Journal of Clinical Oncology, vol.28, issue.14, pp.2348-55, 2010.
DOI : 10.1200/JCO.2009.27.3730

P. Ward, J. Cross, C. Lu, O. Weigert, O. Abel-wahab et al., Identification of additional IDH mutations associated with oncometabolite R(???)-2-hydroxyglutarate production, Oncogene, vol.31, issue.19, pp.312491-312499, 2012.
DOI : 10.1016/j.bbrc.2010.10.038

M. Kranendijk, E. Struys, E. Van-schaftingen, K. Gibson, W. Kanhai et al., IDH2 Mutations in Patients with D-2-Hydroxyglutaric Aciduria. Science, pp.336-336, 2010.

P. Ward, J. Patel, D. Wise, O. Abdel-wahab, B. Bennett et al., The Common Feature of Leukemia-Associated IDH1 and IDH2 Mutations Is a Neomorphic Enzyme Activity Converting ?-Ketoglutarate to 2-Hydroxyglutarate. Cancer Cell, pp.225-259, 2010.

H. Yan, D. Parsons, J. G. Mclendon, R. Rasheed, B. Yuan et al., IDH1 and IDH2 mutations in gliomas, N Engl J Med Feb, vol.19360, issue.8, pp.765-73, 2009.

S. Zhao, Y. Lin, W. Xu, W. Jiang, Z. Zha et al., Glioma-Derived Mutations in IDH1 Dominantly Inhibit IDH1 Catalytic Activity and Induce HIF-1. Science, pp.261-266, 2009.

L. Dang, D. White, S. Gross, B. Bennett, M. Bittinger et al., Cancer-associated IDH1 mutations produce 2-hydroxyglutarate, Nature, vol.50, issue.7274, pp.739-783, 2009.
DOI : 10.1038/nature08617

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2818760

B. Pietrak, H. Zhao, H. Qi, C. Quinn, E. Gao et al., A Tale of Two Subunits: How the Neomorphic R132H IDH1 Mutation Enhances Production of ??HG, Biochemistry, vol.50, issue.21, pp.4804-4816, 2011.
DOI : 10.1021/bi200499m

G. Jin, Z. Reitman, C. Duncan, I. Spasojevic, D. Gooden et al., Disruption of wild-type IDH1 suppresses D-2-hydroxyglutarate production in IDH1-mutated gliomas. Cancer Res, pp.496-501, 2013.

G. Lindahl, G. Lindstedt, and S. Lindstedt, Metabolism of 2-amino-5-hydroxyadipic acid in the rat, Archives of Biochemistry and Biophysics, vol.119, issue.1, pp.347-52, 1967.
DOI : 10.1016/0003-9861(67)90463-8

R. Chalmers, A. Lawson, R. Watts, A. Tavill, J. Kamerling et al., D-2-hydroxyglutaric aciduria: Case report and biochemical studies, Journal of Inherited Metabolic Disease, vol.31, issue.1, pp.11-16, 1980.
DOI : 10.1007/BF02312516

W. Xu, H. Yang, Y. Liu, Y. Yang, P. Wang et al., Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of ?-ketoglutarate-dependent dioxygenases. Cancer Cell, pp.17-30, 2011.

C. Loenarz and C. Schofield, Expanding chemical biology of 2-oxoglutarate oxygenases, Nature Chemical Biology, vol.4, issue.3, pp.152-158, 2008.
DOI : 10.1038/nature06145

D. Krell, P. Mulholland, A. Frampton, J. Krell, J. Stebbing et al., IDH mutations in tumorigenesis and their potential role as novel therapeutic targets, Future Oncology, vol.9, issue.12, pp.1923-1958
DOI : 10.2217/fon.13.143

M. Tahiliani, K. Koh, Y. Shen, W. Pastor, H. Bandukwala et al., Conversion of 5-Methylcytosine to 5-Hydroxymethylcytosine in Mammalian DNA by MLL Partner TET1, Science, vol.324, issue.5929, pp.930-935, 2009.
DOI : 10.1126/science.1170116

J. Guo, Y. Su, C. Zhong, M. G. Song, and H. , Emerging roles of TET proteins and 5-hydroxymethylcytosines in active DNA demethylation and beyond. Cell Cycle Georget Tex, pp.2662-2670, 2011.

S. Ito, D. 'alessio, A. Taranova, O. Hong, K. Sowers et al., Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification, Nature, vol.22, issue.7310, pp.1129-1162, 2010.
DOI : 10.1038/nature09303

S. Jin, Y. Jiang, R. Qiu, T. Rauch, Y. Wang et al., 5-Hydroxymethylcytosine is strongly depleted in human cancers but its levels do not correlate with IDH1 mutations. Cancer Res, pp.7360-7365, 2011.

H. Noushmehr, D. Weisenberger, K. Diefes, H. Phillips, K. Pujara et al., Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell, pp.510-532, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01201573

J. Laffaire, S. Everhard, A. Idbaih, E. Crinière, Y. Marie et al., Methylation profiling identifies 2 groups of gliomas according to their tumorigenesis. Neuro-Oncol, pp.84-98, 2011.

B. Christensen, A. Smith, S. Zheng, D. Koestler, E. Houseman et al., DNA Methylation, Isocitrate Dehydrogenase Mutation, and Survival in Glioma, JNCI Journal of the National Cancer Institute, vol.103, issue.2, pp.143-53, 2011.
DOI : 10.1093/jnci/djq497

T. Watanabe, S. Nobusawa, P. Kleihues, and H. Ohgaki, IDH1 Mutations Are Early Events in the Development of Astrocytomas and Oligodendrogliomas, The American Journal of Pathology, vol.174, issue.4, pp.1149-53, 2009.
DOI : 10.2353/ajpath.2009.080958

C. Lu, P. Ward, G. Kapoor, D. Rohle, S. Turcan et al., IDH mutation impairs histone demethylation and results in a block to cell differentiation, Nature, vol.249, issue.7390, pp.474-482, 2012.
DOI : 10.1038/nature10860

S. Turcan, D. Rohle, A. Goenka, L. Walsh, F. Fang et al., IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell, pp.479-83, 2010.

D. Pollyea, H. Kohrt, B. Zhang, J. Zehnder, D. Schenkein et al., mutant acute myeloid leukemia: predicting patient responses, minimal residual disease and correlations with methylcytosine and hydroxymethylcytosine levels, Leukemia & Lymphoma, vol.54, issue.2, pp.408-418, 2013.
DOI : 10.1016/j.ccr.2010.11.015

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3688062

M. Sasaki, C. Knobbe, J. Munger, E. Lind, D. Brenner et al., IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics, Nature, vol.29, issue.7413, pp.656-665, 2012.
DOI : 10.1038/nature11323

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4005896

P. Metellus, C. Colin, D. Taieb, E. Guedj, I. Nanni-metellus et al., IDH mutation status impact on in vivo hypoxia biomarkers expression: new insights from a clinical, nuclear imaging and immunohistochemical study in 33 glioma patients, Journal of Neuro-Oncology, vol.13, issue.3, pp.591-600, 2011.
DOI : 10.1007/s11060-011-0625-2

A. Weljie, A. Bondareva, P. Zang, and F. Jirik, (1)H NMR metabolomics identification of markers of hypoxiainduced metabolic shifts in a breast cancer model system, J Biomol NMR, vol.49, pp.3-4185, 2011.

A. Majmundar, W. Wong, and M. Simon, Hypoxia-Inducible Factors and the Response to Hypoxic Stress, Molecular Cell, vol.40, issue.2
DOI : 10.1016/j.molcel.2010.09.022

M. Sasaki, C. Knobbe, M. Itsumi, A. Elia, I. Harris et al., D-2-hydroxyglutarate produced by mutant IDH1 perturbs collagen maturation and basement membrane function, Genes & Development, vol.26, issue.18, pp.2038-2087, 2012.
DOI : 10.1101/gad.198200.112

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3444730

R. Chowdhury, K. Yeoh, Y. Tian, L. Hillringhaus, E. Bagg et al., The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases, EMBO reports, vol.1815, issue.5, pp.463-472, 2011.
DOI : 10.1126/science.1170944

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3090014

P. Koivunen, S. Lee, C. Duncan, G. Lopez, G. Lu et al., Transformation by the (R)-enantiomer of 2-hydroxyglutarate linked to EGLN activation, Nature, vol.37, issue.7390, pp.484-492, 2012.
DOI : 10.1038/nature10898

J. Losman, R. Looper, P. Koivunen, S. Lee, R. Schneider et al., (R)-2-Hydroxyglutarate Is Sufficient to Promote Leukemogenesis and Its Effects Are Reversible, Science, vol.339, issue.6127, pp.1621-1626, 2013.
DOI : 10.1126/science.1231677

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3836459

S. Williams, M. Karajannis, L. Chiriboga, J. Golfinos, A. Von-deimling et al., R132H-mutation of isocitrate dehydrogenase-1 is not sufficient for HIF-1?? upregulation in adult glioma, Acta Neuropathologica, vol.324, issue.2
DOI : 10.1007/s00401-010-0790-y

C. Bardella, P. Pollard, and I. Tomlinson, SDH mutations in cancer, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1807, issue.11, 2011.
DOI : 10.1016/j.bbabio.2011.07.003

URL : http://doi.org/10.1016/j.bbabio.2011.07.003

O. Reilly, M. Boehm, T. Shing, Y. Fukai, N. Vasios et al., Endostatin: An Endogenous Inhibitor of Angiogenesis and Tumor Growth, Cell, vol.88, issue.2, pp.277-85, 1997.
DOI : 10.1016/S0092-8674(00)81848-6

D. Hanahan and R. Weinberg, Hallmarks of cancer: the next generation. Cell, pp.646-74, 2011.

P. Pollard, N. Wortham, E. Barclay, A. Alam, G. Elia et al., Evidence of increased microvessel density and activation of the hypoxia pathway in tumours from the hereditary leiomyomatosis and renal cell cancer syndrome Succinate inhibition of alpha-ketoglutarate-dependent enzymes in a yeast model of paraganglioma, J Pathol. Hum Mol Genet, vol.20516, issue.124, pp.41-50, 2005.

Z. Reitman, J. G. Karoly, E. Spasojevic, I. Yang, J. Kinzler et al., Profiling the effects of isocitrate dehydrogenase 1 and 2 mutations on the cellular metabolome, Proceedings of the National Academy of Sciences, vol.108, issue.8, pp.3270-3275, 2011.
DOI : 10.1073/pnas.1019393108

A. Latini, C. Da-silva, G. Ferreira, P. Schuck, K. Scussiato et al., Mitochondrial energy metabolism is markedly impaired by d-2-hydroxyglutaric acid in rat tissues, Molecular Genetics and Metabolism, vol.86, issue.1-2, pp.188-99, 2005.
DOI : 10.1016/j.ymgme.2005.05.002

S. Kölker, V. Pawlak, B. Ahlemeyer, J. Okun, F. Hörster et al., -2-hydroxyglutaric aciduria, European Journal of Neuroscience, vol.16, issue.Suppl. 1
DOI : 10.1002/1531-8249(199901)45:1<111::aid-art17>3.0.co;2-n

K. Wellen, G. Hatzivassiliou, U. Sachdeva, T. Bui, J. Cross et al., ATP-citrate lyase links cellular metabolism to histone acetylation. Science, pp.1076-80, 2009.
DOI : 10.1126/science.1164097

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2746744

H. Luksch, O. Uckermann, A. Stepulak, S. Hendruschk, J. Marzahn et al., Silencing of selected glutamate receptor subunits modulates cancer growth, Anticancer Res, vol.31, issue.10, pp.3181-92, 2011.

Y. Piao, L. Lu, and J. De-groot, AMPA receptors promote perivascular glioma invasion via beta1 integrindependent adhesion to the extracellular matrix. Neuro-Oncol, pp.260-73, 2009.

S. Lyons, W. Chung, A. Weaver, T. Ogunrinu, and H. Sontheimer, Autocrine glutamate signaling promotes glioma cell invasion. Cancer Res, pp.9463-71, 2007.
DOI : 10.1158/0008-5472.can-07-2034

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2045073

M. Seltzer, B. Bennett, A. Joshi, P. Gao, A. Thomas et al., Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res, pp.8981-8988, 2010.

M. Mcfarland and R. Cripps, Diabetes Mellitus and Increased Risk of Cancer: Focus on Metformin and the Insulin Analogs, Pharmacotherapy, vol.31, issue.11, pp.1159-78, 2010.
DOI : 10.2337/db09-1371

S. Ronnebaum, O. Ilkayeva, S. Burgess, J. Joseph, D. Lu et al., A Pyruvate Cycling Pathway Involving Cytosolic NADP-dependent Isocitrate Dehydrogenase Regulates Glucose-stimulated Insulin Secretion, Journal of Biological Chemistry, vol.281, issue.41, pp.30593-602, 2006.
DOI : 10.1074/jbc.M511908200

A. Latini, K. Scussiato, R. Rosa, S. Llesuy, A. Belló-klein et al., D-2-hydroxyglutaric acid induces oxidative stress in cerebral cortex of young rats, European Journal of Neuroscience, vol.77, issue.Suppl. 1, pp.2017-2039, 2003.
DOI : 10.1046/j.1460-9568.2003.02639.x

D. Rakheja, S. Konoplev, L. Medeiros, and W. Chen, IDH mutations in acute myeloid leukemia, Human Pathology, vol.43, issue.10, pp.1541-51
DOI : 10.1016/j.humpath.2012.05.003

H. Yan, D. Parsons, J. G. Mclendon, R. Rasheed, B. Yuan et al., Mutations in Gliomas, New England Journal of Medicine, vol.360, issue.8, pp.765-73, 2009.
DOI : 10.1056/NEJMoa0808710

M. Patnaik, C. Hanson, J. Hodnefield, T. Lasho, C. Finke et al., Differential prognostic effect of IDH1 versus IDH2 mutations in myelodysplastic syndromes: a Mayo Clinic Study of 277 patients, Leukemia, vol.26, issue.1, pp.101-106, 2012.
DOI : 10.1002/ajh.21984

A. Tefferi, T. Lasho, O. Abdel-wahab, P. Guglielmelli, J. Patel et al., IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis, Leukemia, vol.114, issue.7, pp.1302-1311, 2010.
DOI : 10.1182/blood-2008-12-194548

Y. Zhang, H. Wei, K. Tang, D. Lin, C. Zhang et al., Mutation Analysis of Isocitrate Dehydrogenase in Acute Lymphoblastic Leukemia, Genetic Testing and Molecular Biomarkers, vol.16, issue.8, pp.991-996, 2012.
DOI : 10.1089/gtmb.2011.0323

P. Paschka, R. Schlenk, V. Gaidzik, M. Habdank, J. Krönke et al., Internal Tandem Duplication, Journal of Clinical Oncology, vol.28, issue.22, pp.3636-3679, 2010.
DOI : 10.1200/JCO.2010.28.3762

S. Abbas, S. Lugthart, F. Kavelaars, A. Schelen, J. Koenders et al., Acquired mutations in the genes encoding IDH1 and IDH2 both are recurrent aberrations in acute myeloid leukemia: prevalence and prognostic value, Blood, vol.116, issue.12, pp.2122-2128, 2010.
DOI : 10.1182/blood-2009-11-250878

K. Patel, F. Ravandi, D. Ma, A. Paladugu, B. Barkoh et al., Mutation, American Journal of Clinical Pathology, vol.135, issue.1, pp.35-45, 2011.
DOI : 10.1309/AJCPD7NR2RMNQDVF

URL : https://hal.archives-ouvertes.fr/hal-00552549

F. Damm, F. Thol, I. Hollink, M. Zimmermann, K. Reinhardt et al., Prevalence and prognostic value of IDH1 and IDH2 mutations in childhood AML: a study of the AML???BFM and DCOG study groups, Leukemia, vol.114, issue.11, pp.1704-1714, 2011.
DOI : 10.1038/leu.2010.251

C. Green, C. Evans, R. Hills, A. Burnett, D. Linch et al., The prognostic significance of IDH1 mutations in younger adult patients with acute myeloid leukemia is dependent on FLT3/ITD status, Blood, vol.116, issue.15, 2010.
DOI : 10.1182/blood-2010-02-270926

D. Rakheja, S. Konoplev, L. Medeiros, and W. Chen, IDH mutations in acute myeloid leukemia, Human Pathology, vol.43, issue.10, pp.1541-51
DOI : 10.1016/j.humpath.2012.05.003

E. Estey, S. Amadori, F. Appelbaum, T. Buchner, and A. Burnett, étude à parxr des registres des cancers du réseau Francim. Saint-Maurice: Institut de veille sanitaire; 2013 Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet, Blood, vol.115, issue.3, pp.453-74, 1980.

D. Arber, A. Orazi, R. Hasserjian, J. Thiele, M. Borowitz et al., The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia

B. Löwenberg, J. Downing, A. Burnett, M. Smith, R. Hills et al., Acute myeloid leukemia Independent prognostic variables in acute myeloid leukaemia, N Engl J Med Blood Rev, vol.34125, issue.141, pp.1051-62, 1999.

S. Kayser, K. Dohner, J. Krauter, C. Kohne, H. Horst et al., The impact of therapy-related acute myeloid leukemia (AML) on outcome in 2853 adult patients with newly diagnosed AML. Blood, Feb, vol.17117, issue.7, pp.2137-2182, 2011.

O. Weinberg, M. Seetharam, L. Ren, K. Seo, L. Ma et al., Clinical characterization of acute myeloid leukemia with myelodysplasia-related changes as defined by the 2008 WHO classification system, Blood, vol.113, issue.9, pp.1906-1914, 2009.
DOI : 10.1182/blood-2008-10-182782

M. Miesner, C. Haferlach, U. Bacher, T. Weiss, K. Macijewski et al., Multilineage dysplasia (MLD) in acute myeloid leukemia (AML) correlates with MDS-related cytogenetic abnormalities and a prior history of MDS or MDS/MPN but has no independent prognostic relevance: a comparison of 408 cases classified as "AML not otherwise specified" (AML-NOS) or "AML with myelodysplasia-related changes" (AML-MRC), Blood, vol.116, issue.15, pp.2742-51, 2010.
DOI : 10.1182/blood-2010-04-279794

D. Grimwade, R. Hills, A. Moorman, H. Walker, S. Chatters et al., Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials, Blood, vol.116, issue.3, pp.354-65, 2010.
DOI : 10.1182/blood-2009-11-254441

S. Kayser, M. Zucknick, K. Dohner, J. Krauter, C. Kohne et al., Monosomal karyotype in adult acute myeloid leukemia: prognostic impact and outcome after different treatment strategies, Blood, vol.119, issue.2, pp.551-559, 2012.
DOI : 10.1182/blood-2011-07-367508

URL : http://mediatum.ub.tum.de/doc/1181634/document.pdf

B. Wouters, B. Lowenberg, C. Erpelinck-verschueren, W. Van-putten, P. Valk et al., Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome, Blood, vol.113, issue.13, pp.3088-91, 2009.
DOI : 10.1182/blood-2008-09-179895

V. Grossmann, S. Schnittger, A. Kohlmann, C. Eder, A. Roller et al., A novel hierarchical prognostic model of AML solely based on molecular mutations, Blood, vol.120, issue.15, pp.2963-72, 2012.
DOI : 10.1182/blood-2012-03-419622

J. Patel, M. Gönen, M. Figueroa, H. Fernandez, Z. Sun et al., Prognostic Relevance of Integrated Genetic Profiling in Acute Myeloid Leukemia, New England Journal of Medicine, vol.366, issue.12, pp.1079-89, 2012.
DOI : 10.1056/NEJMoa1112304

C. Sloan, M. Luskin, A. Boccuti, A. Sehgal, J. Zhao et al., A Modified Integrated Genetic Model for Risk Prediction in Younger Patients with Acute Myeloid Leukemia, PLOS ONE, 2016.
DOI : 10.1371/journal.pone.0153016.s002

A. Mead, D. Linch, R. Hills, K. Wheatley, A. Burnett et al., FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood, pp.1262-70, 2007.

N. Boissel, O. Nibourel, A. Renneville, C. Gardin, O. Reman et al., Prognostic Impact of Isocitrate Dehydrogenase Enzyme Isoforms 1 and 2 Mutations in Acute Myeloid Leukemia: A Study by the Acute Leukemia French Association Group, Journal of Clinical Oncology, vol.28, issue.23, pp.3717-3740, 2010.
DOI : 10.1200/JCO.2010.28.2285

C. Green, C. Evans, L. Zhao, R. Hills, A. Burnett et al., The prognostic significance of IDH2 mutations in AML depends on the location of the mutation, Blood, vol.118, issue.2, pp.409-421, 2011.
DOI : 10.1182/blood-2010-12-322479

F. Thol, F. Damm, K. Wagner, G. Göhring, B. Schlegelberger et al., Prognostic impact of IDH2 mutations in cytogenetically normal acute myeloid leukemia, Blood, vol.116, issue.4, pp.614-620, 2010.
DOI : 10.1182/blood-2010-03-272146

K. Wagner, F. Damm, G. Göhring, K. Görlich, M. Heuser et al., Single Nucleotide Polymorphism in Cytogenetically Normal Acute Myeloid Leukemia: SNP rs11554137 Is an Adverse Prognostic Factor, Journal of Clinical Oncology, vol.28, issue.14, pp.2356-64, 2010.
DOI : 10.1200/JCO.2009.27.6899

W. Chou, H. Hou, C. Chen, J. Tang, M. Yao et al., Distinct clinical and biologic characteristics in adult acute myeloid leukemia bearing the isocitrate dehydrogenase 1 mutation, Blood, vol.115, issue.14, 2010.
DOI : 10.1182/blood-2009-11-253070

W. Chou, W. Lei, B. Ko, H. Hou, C. Chen et al., The prognostic impact and stability of Isocitrate dehydrogenase 2 mutation in adult patients with acute myeloid leukemia, Leukemia, vol.268, issue.2, 2011.
DOI : 10.1038/ncponc0097

D. Rakheja, S. Konoplev, M. Su, D. Wheeler, D. Muzny et al., High incidence of IDH mutations in acute myeloid leukaemia with cuplike nuclei, British Journal of Haematology, vol.360, issue.1, pp.125-133, 2011.
DOI : 10.1111/j.1365-2141.2011.08646.x

A. Andersson, D. Miller, J. Lynch, A. Lemoff, Z. Cai et al., IDH1 and IDH2 mutations in pediatric acute leukemia, Leukemia, vol.92, issue.10, pp.1570-1577, 2011.
DOI : 10.1182/blood.V99.12.4326

M. Pigazzi, G. Ferrari, R. Masetti, B. Falini, F. Martinolli et al., Low prevalence of IDH1 gene mutation in childhood AML in Italy, Leukemia, vol.25, issue.1, pp.173-177, 2011.
DOI : 10.1182/blood-2009-11-253070

M. Sekeres, P. Elson, M. Kalaycio, A. Advani, E. Copelan et al., Time from diagnosis to treatment initiation predicts survival in younger, but not older, acute myeloid leukemia patients, Blood, vol.113, issue.1, 2009.
DOI : 10.1182/blood-2008-05-157065

F. Lacombe, C. Arnoulet, M. Maynadié, E. Lippert, I. Luquet et al., Early clearance of peripheral blasts measured by flow cytometry during the first week of AML induction therapy as a new independent prognostic factor: a GOELAMS study, Leukemia, vol.22, issue.2, pp.350-357, 2009.
DOI : 10.1038/leu.2008.296

M. Elliott, M. Litzow, L. Letendre, R. Wolf, C. Hanson et al., Early peripheral blood blast clearance during induction chemotherapy for acute myeloid leukemia predicts superior relapse-free survival, Blood, vol.110, issue.13, pp.4172-4176, 2007.
DOI : 10.1182/blood-2007-07-104091

H. Dombret and C. Gardin, An update of current treatments for adult acute myeloid leukemia, Blood, vol.127, issue.1, p.2016
DOI : 10.1182/blood-2015-08-604520

H. Fernandez, Z. Sun, X. Yao, M. Litzow, S. Luger et al., Anthracycline Dose Intensification in Acute Myeloid Leukemia, New England Journal of Medicine, vol.361, issue.13, pp.1249-59, 2009.
DOI : 10.1056/NEJMoa0904544

M. Luskin, J. Lee, H. Fernandez, H. Lazarus, J. Rowe et al., High Dose Daunorubicin Improves Survival in AML up to Age 60, Across All Cytogenetic Risk Groups Including Patients with Unfavorable Cytogenetic Risk, and FLT3-ITD Mutant AML: Updated Analyses from Eastern Cooperative Oncology Trial E1900, Blood, vol.124, issue.21, pp.373-373, 2014.

B. Löwenberg, G. Ossenkoppele, W. Van-putten, H. Schouten, C. Graux et al., High-Dose Daunorubicin in Older Patients with Acute Myeloid Leukemia, New England Journal of Medicine, vol.361, issue.13, pp.1235-1283, 2009.
DOI : 10.1056/NEJMoa0901409

J. Lee, Y. Joo, H. Kim, S. Bae, M. Kim et al., A randomized trial comparing standard versus high-dose daunorubicin induction in patients with acute myeloid leukemia, Blood, vol.118, issue.14, pp.3832-3873, 2011.
DOI : 10.1182/blood-2011-06-361410

A. Burnett, N. Russell, R. Hills, J. Kell, J. Cavenagh et al., A randomized comparison of daunorubicin 90 mg/m2 vs 60 mg/m2 in AML induction: results from the UK NCRI AML17 trial in 1206 patients, Blood, vol.125, issue.25, pp.3878-85, 2015.
DOI : 10.1182/blood-2015-01-623447

C. Pautas, F. Merabet, X. Thomas, E. Raffoux, C. Gardin et al., Randomized Study of Intensified Anthracycline Doses for Induction and Recombinant Interleukin-2 for Maintenance in Patients With Acute Myeloid Leukemia Age 50 to 70 Years: Results of the ALFA-9801 Study, Journal of Clinical Oncology, vol.28, issue.5, pp.808-822, 2010.
DOI : 10.1200/JCO.2009.23.2652

URL : https://hal.archives-ouvertes.fr/hal-00453228

S. Ohtake, S. Miyawaki, H. Fujita, H. Kiyoi, K. Shinagawa et al., Randomized study of induction therapy comparing standard-dose idarubicin with high-dose daunorubicin in adult patients with previously untreated acute myeloid leukemia: the JALSG AML201 Study, Blood, vol.117, issue.8, pp.2358-65, 2011.
DOI : 10.1182/blood-2010-03-273243

J. Weick, K. Kopecky, F. Appelbaum, D. Head, L. Kingsbury et al., A randomized investigation of high-dose versus standard-dose cytosine arabinoside with daunorubicin in patients with previously untreated acute myeloid leukemia: a Southwest Oncology Group study, Blood, vol.88, issue.8, pp.2841-51, 1996.

J. Bishop, J. Matthews, G. Young, J. Szer, A. Gillett et al., A randomized study of high-dose cytarabine in induction in acute myeloid leukemia, Blood, vol.87, issue.5, pp.1710-1717, 1996.

B. Löwenberg, T. Pabst, E. Vellenga, W. Van-putten, H. Schouten et al., Cytarabine Dose for Acute Myeloid Leukemia, New England Journal of Medicine, vol.364, issue.11, pp.1027-1063, 2011.
DOI : 10.1056/NEJMoa1010222

R. Willemze, S. Suciu, G. Meloni, B. Labar, M. J. Halkes et al., High-Dose Cytarabine in Induction Treatment Improves the Outcome of Adult Patients Younger Than Age 46 Years With Acute Myeloid Leukemia: Results of the EORTC-GIMEMA AML-12 Trial, Journal of Clinical Oncology, vol.32, issue.3, pp.219-247, 2014.
DOI : 10.1200/JCO.2013.51.8571

S. Petersdorf, K. Kopecky, M. Slovak, C. Willman, T. Nevill et al., A phase 3 study of gemtuzumab ozogamicin during induction and postconsolidation therapy in younger patients with acute myeloid leukemia, Blood, vol.121, issue.24, pp.4854-60, 2013.
DOI : 10.1182/blood-2013-01-466706

A. Burnett, R. Hills, D. Milligan, L. Kjeldsen, J. Kell et al., Identification of Patients With Acute Myeloblastic Leukemia Who Benefit From the Addition of Gemtuzumab Ozogamicin: Results of the MRC AML15 Trial, Journal of Clinical Oncology, vol.29, issue.4, pp.369-77, 2011.
DOI : 10.1200/JCO.2010.31.4310

S. Castaigne, C. Pautas, C. Terré, E. Raffoux, D. Bordessoule et al., Effect of gemtuzumab ozogamicin on survival of adult patients with de-novo acute myeloid leukaemia (ALFA-0701): a randomised, open-label, phase 3 study, The Lancet, vol.379, issue.9825, pp.1508-1524, 2012.
DOI : 10.1016/S0140-6736(12)60485-1

URL : https://hal.archives-ouvertes.fr/hal-01056520

A. Burnett, N. Russell, R. Hills, J. Kell, S. Freeman et al., Addition of Gemtuzumab Ozogamicin to Induction Chemotherapy Improves Survival in Older Patients With Acute Myeloid Leukemia, Journal of Clinical Oncology, vol.30, issue.32, pp.3924-3955, 2012.
DOI : 10.1200/JCO.2012.42.2964

R. Hills, S. Castaigne, F. Appelbaum, J. Delaunay, S. Petersdorf et al., Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials, The Lancet Oncology, vol.15, issue.9, pp.986-96, 2014.
DOI : 10.1016/S1470-2045(14)70281-5

B. Löwenberg, J. Beck, and C. Graux, Gemtuzumab ozogamicin as postremission treatment in AML at 60 years of age or more: results of a multicenter phase 3 study, Blood, vol.115, issue.13, pp.2586-2591, 2010.
DOI : 10.1182/blood-2009-10-246470

J. Holowiecki, S. Grosicki, S. Giebel, T. Robak, S. Kyrcz-krzemien et al., Cladribine, But Not Fludarabine, Added to Daunorubicin and Cytarabine During Induction Prolongs Survival of Patients With Acute Myeloid Leukemia: A Multicenter, Randomized Phase III Study, Journal of Clinical Oncology, vol.30, issue.20, pp.2441-2449, 2012.
DOI : 10.1200/JCO.2011.37.1286

S. Faderl, S. Verstovsek, J. Cortes, F. Ravandi, M. Beran et al., Clofarabine and cytarabine combination as induction therapy for acute myeloid leukemia (AML) in patients 50 years of age or older, Blood, vol.108, issue.1, pp.45-51, 2006.
DOI : 10.1182/blood-2005-08-3294

H. Kantarjian, H. Erba, D. Claxton, M. Arellano, R. Lyons et al., Phase II Study of Clofarabine Monotherapy in Previously Untreated Older Adults With Acute Myeloid Leukemia and Unfavorable Prognostic Factors, Journal of Clinical Oncology, vol.28, issue.4, pp.549-55, 2010.
DOI : 10.1200/JCO.2009.23.3130

R. Willemze, S. Suciu, P. Muus, C. Halkes, G. Meloni et al., Clofarabine in combination with a standard remission induction regimen (cytosine arabinoside and idarubicin) in patients with previously untreated intermediate and bad-risk acute myelogenous leukemia (AML) or high-risk myelodysplastic syndrome (HR-MDS): phase I results of an ongoing phase I/II study of the leukemia groups of EORTC and GIMEMA (EORTC GIMEMA 06061/AML-14A trial), Annals of Hematology, vol.21, issue.5, 2014.
DOI : 10.1007/s00277-014-2056-6

A. Burnett, N. Russell, R. Hills, A. Hunter, L. Kjeldsen et al., Optimization of Chemotherapy for Younger Patients With Acute Myeloid Leukemia: Results of the Medical Research Council AML15 Trial, Journal of Clinical Oncology, vol.31, issue.27, pp.3360-3368, 2013.
DOI : 10.1200/JCO.2012.47.4874

H. Serve, U. Krug, R. Wagner, M. Sauerland, A. Heinecke et al., Sorafenib in Combination With Intensive Chemotherapy in Elderly Patients With Acute Myeloid Leukemia: Results From a Randomized, Placebo-Controlled Trial, Journal of Clinical Oncology, vol.31, issue.25, pp.313110-313118, 2013.
DOI : 10.1200/JCO.2012.46.4990

J. Cornelissen, W. Van-putten, L. Verdonck, M. Theobald, J. E. Daenen et al., Results of a HOVON/SAKK donor versus no-donor analysis of myeloablative HLA-identical sibling stem cell transplantation in first remission acute myeloid leukemia in young and middle-aged adults: benefits for whom?, Blood, vol.109, issue.9, 2007.
DOI : 10.1182/blood-2006-06-025627

J. Koreth, R. Schlenk, K. Kopecky, S. Honda, J. Sierra et al., Allogeneic Stem Cell Transplantation for Acute Myeloid Leukemia in First Complete Remission, JAMA, vol.301, issue.22, pp.1909-1927, 2008.
DOI : 10.1001/jama.2009.813

N. Russell, L. Kjeldsen, C. Craddock, A. Pagliuca, J. Yin et al., A comparative assessment of the curative potential of reduced intensity allografts in acute myeloid leukaemia, Leukemia, vol.92, issue.7, pp.1478-84
DOI : 10.1182/blood-2004-02-0545

J. Cornelissen, J. Versluis, J. Passweg, W. Van-putten, M. Manz et al., Comparative therapeutic value of post-remission approaches in patients with acute myeloid leukemia aged 40???60 years, Leukemia, vol.1, issue.5, pp.1041-50, 2015.
DOI : 10.1002/bimj.4710280508

J. Passweg, M. Labopin, J. Cornelissen, L. Volin, G. Socié et al., Conditioning intensity in middleaged patients with AML in first CR: no advantage for myeloablative regimens irrespective of the risk group?an observational analysis by the Acute Leukemia Working Party of the EBMT, Bone Marrow Transplant, 2015.

N. Boissel, A. Renneville, T. Leguay, P. Lefebvre, C. Recher et al., Dasatinib in high-risk core binding factor acute myeloid leukemia in first complete remission: a French Acute Myeloid Leukemia Intergroup trial, Haematologica, vol.100, issue.6, pp.780-785, 2015.
DOI : 10.3324/haematol.2014.114884

P. Paschka, R. Schlenk, and D. Weber, Dasatinib (DAS) in combination with chemotherapy and as maintenance in core-binding factor (CBF) acute myeloid leukemia (AML): a phase Ib/IIa study of the German- Austrian AML Study Group (AMLSG) [abstract], Haematologica, vol.100, issue.s1, 2015.

G. Marcucci, S. Geyer, and W. Zhao, Adding KIT inhibitor dasatinib (DAS) to chemotherapy overcomes the negative impact of KIT mutation/over-expression in core binding factor (CBF) acute myeloid leukemia (AML): results from CALGB 10801 (Alliance) [abstract] Results from a randomized trial of salvage chemotherapy followed by lestaurtinib for patients with FLT3 mutant AML in first relapse, Blood Blood, vol.124, issue.21, 2011.

S. Knapper, R. Hills, and J. Cavenagh, A randomised comparison of the sequential addition of the FLT3 inhibitor lestaurtinib (CEP701) to standard first line chemotherapy for FLT3-mutated acute myeloid leukemia: The UK experience [abstract] Secondary mutations as mediators of resistance to targeted therapy in leukemia, Blood Blood, vol.124125, issue.2121, pp.3236-3281, 2014.

A. Burnett, R. Hills, C. Green, S. Jenkinson, K. Koo et al., The impact on outcome of the addition of all-trans retinoic acid to intensive chemotherapy in younger patients with nonacute promyelocytic acute myeloid leukemia: overall results and results in genotypic subgroups defined by mutations in NPM1, FLT3, and CEBPA, Blood, vol.115, issue.5, pp.948-56, 2010.
DOI : 10.1182/blood-2009-08-236588

R. Schlenk, K. Dohner, M. Kneba, K. Gotze, F. Hartmann et al., Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG Trial AML HD98B, Haematologica, vol.94, issue.1, pp.54-60, 2009.
DOI : 10.3324/haematol.13378

E. Hajj, H. Dassouki, Z. Berthier, C. Raffoux, E. Ades et al., Retinoic acid and arsenic trioxide trigger degradation of mutated NPM1, resulting in apoptosis of AML cells. Blood, pp.3447-54, 2015.

M. Martelli, I. Gionfriddo, F. Mezzasoma, F. Milano, S. Pierangeli et al., Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells, Blood, vol.125, issue.22, pp.3455-65, 2015.
DOI : 10.1182/blood-2014-11-611459

F. Wang, J. Travins, B. Delabarre, V. Penard-lacronique, S. Schalm et al., Targeted Inhibition of Mutant IDH2 in Leukemia Cells Induces Cellular Differentiation, Science, vol.340, issue.6132, pp.622-628, 2013.
DOI : 10.1126/science.1234769

D. Rohle, J. Popovici-muller, N. Palaskas, S. Turcan, C. Grommes et al., An Inhibitor of Mutant IDH1 Delays Growth and Promotes Differentiation of Glioma Cells, Science, vol.340, issue.6132, pp.626-656, 2013.
DOI : 10.1126/science.1236062

A. Kernytsky, F. Wang, E. Hansen, S. Schalm, K. Straley et al., IDH2 mutation-induced histone and DNA hypermethylation is progressively reversed by small-molecule inhibition. Blood, pp.296-303, 2015.
DOI : 10.1182/blood-2013-10-533604

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4295919

C. Dinardo, E. Stein, and J. Altman, AG-221, an oral, selective, fi rst-in-class,potent inhibitor of the IDH2 mutant enzyme, induced durable responses in a phaseI study of IDH2 mutation-positive advanced hematologic malignancies, EuropeanHematology Association Learning Center 13, 2015.

C. Dinardo, S. De-botton, and D. Pollyea, Molecular profiling and relationship with clinical response in patients with IDH1 mutation-positive hematologic malignancies receiving AG-120, a first-in-class potent inhibitor of mutant IDH1, in addition to data from the completed dose escalation portion of the phase 1 study, Blood, pp.126-1306, 2015.

B. Kc and C. Dinardo, Evidence for Clinical Differentiation and Differentiation Syndrome in Patients With

A. Emadi, R. Faramand, B. Carter-cooper, S. Tolu, L. Ford et al., AG-120 Clin Lymphoma Myeloma Leuk Presence of isocitrate dehydrogenase mutations may predict clinical response to hypomethylating agents in patients with acute myeloid leukemia, Acute Myeloid Leukemia and IDH1 Mutations Treated With the Targeted Mutant IDH1 Inhibitor, pp.77-86, 2015.

K. Metzeler, A. Walker, S. Geyer, R. Garzon, R. Klisovic et al., DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia, Leukemia, vol.468, issue.5, 2012.
DOI : 10.1038/leu.2011.342

C. Dinardo, K. Patel, G. Garcia-manero, R. Luthra, S. Pierce et al., mutations with outcome in older patients with acute myeloid leukemia treated with hypomethylating agents, Leukemia & Lymphoma, vol.55, issue.8, pp.1925-1934, 2014.
DOI : 10.1182/blood-2011-07-367961

T. Schumacher, L. Bunse, S. Pusch, F. Sahm, B. Wiestler et al., A vaccine targeting mutant IDH1 induces antitumour immunity, Nature, vol.124, issue.7514, pp.324-331, 2014.
DOI : 10.1038/nature13387

S. Pellegatta, L. Valletta, C. Corbetta, M. Patanè, I. Zucca et al., Effective immuno-targeting of the IDH1 mutation R132H in a murine model of intracranial glioma, Acta Neuropathologica Communications, vol.210, issue.1, p.4, 2015.
DOI : 10.1186/s40478-014-0180-0

L. Dang, Y. K. Attar, and E. , IDH mutations in cancer and progress toward development of targeted therapeutics, Annals of Oncology, vol.27, issue.4, pp.599-608
DOI : 10.1093/annonc/mdw013

M. Jaglal, V. Duong, C. Bello, A. Ali, N. Padron et al., Cladribine, cytarabine, filgrastim, and mitoxantrone (CLAG-M) compared to standard induction in acute myeloid leukemia from myelodysplastic syndrome after azanucleoside failure, Leukemia Research, vol.38, issue.4, pp.443-449
DOI : 10.1016/j.leukres.2013.12.010

H. Becker, G. Marcucci, K. Maharry, M. Radmacher, K. Mrozek et al., Mutations in Older Patients With Cytogenetically Normal De Novo Acute Myeloid Leukemia and Associated Gene- and MicroRNA-Expression Signatures: A Cancer and Leukemia Group B Study, Journal of Clinical Oncology, vol.28, issue.4, pp.596-604, 2010.
DOI : 10.1200/JCO.2009.25.1496

C. Gardin, S. Chevret, C. Pautas, P. Turlure, E. Raffoux et al., Superior Long-Term Outcome With Idarubicin Compared With High-Dose Daunorubicin in Patients With Acute Myeloid Leukemia Age 50 Years and Older, Journal of Clinical Oncology, vol.31, issue.3, pp.321-328, 2013.
DOI : 10.1200/JCO.2011.40.3642

A. Burnett, D. Milligan, A. Prentice, A. Goldstone, M. Mcmullin et al., A comparison of lowdose cytarabine and hydroxyurea with or without all-trans retinoic acid for acute myeloid leukemia and highrisk myelodysplastic syndrome in patients not considered fit for intensive treatment. Cancer, pp.1114-1138, 2007.

H. Kantarjian, X. Thomas, A. Dmoszynska, A. Wierzbowska, G. Mazur et al., Multicenter, Randomized, Open-Label, Phase III Trial of Decitabine Versus Patient Choice, With Physician Advice, of Either Supportive Care or Low-Dose Cytarabine for the Treatment of Older Patients With Newly Diagnosed Acute Myeloid Leukemia, Journal of Clinical Oncology, vol.30, issue.21, pp.2670-2677, 2012.
DOI : 10.1200/JCO.2011.38.9429

H. Dohner, M. Lubbert, W. Fiedler, L. Fouillard, A. Haaland et al., Randomized, phase 2 trial of low-dose cytarabine with or without volasertib in AML patients not suitable for induction therapy, Blood, vol.124, issue.9, pp.1426-1459, 2014.
DOI : 10.1182/blood-2014-03-560557

D. Naval, H. Kantarjian, and G. Garcia-manero, Phase I/II study of vosaroxin and decitabine in newly diagnosed older patients (pts) with acute myeloid leukemia (AML) and high risk myelodysplastic syndrome (MDS) [abstract] Curability of Patients With Acute Myeloid Leukemia Who Did Not Undergo Transplantation in First Remission, Blood J Clin Oncol, vol.124, issue.2110, pp.311293-301, 2013.

E. Freireich, E. Gehan, D. Sulman, D. Boggs, and E. Frei, The effect of chemotherapy on acute leukemia in the human, Journal of Chronic Diseases, vol.14, issue.6, pp.593-608, 1961.
DOI : 10.1016/0021-9681(61)90118-7

D. Campana and C. Pui, Detection of minimal residual disease in acute leukemia: methodologic advances and clinical significance, Blood. Mar, vol.1585, issue.6, pp.1416-1450, 1995.

S. Schnittger, M. Weisser, C. Schoch, W. Hiddemann, T. Haferlach et al., New score predicting for prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid leukemia based on quantification of fusion transcripts, Blood, vol.102, issue.8, pp.2746-55, 2003.
DOI : 10.1182/blood-2003-03-0880

J. Stentoft, P. Hokland, M. Ostergaard, H. Hasle, and C. Nyvold, Minimal residual core binding factor AMLs by real time quantitative PCR???Initial response to chemotherapy predicts event free survival and close monitoring of peripheral blood unravels the kinetics of relapse, Leukemia Research, vol.30, issue.4, pp.389-95, 2006.
DOI : 10.1016/j.leukres.2005.08.030

H. Ommen, C. Nyvold, K. Braendstrup, B. Andersen, I. Ommen et al., Relapse prediction in acute myeloid leukaemia patients in complete remission using WT1 as a molecular marker: development of a mathematical model to predict time from molecular to clinical relapse and define optimal sampling intervals, British Journal of Haematology, vol.47, issue.6, pp.782-91, 2008.
DOI : 10.1038/sj.leu.2403809

D. Cilloni, F. Messa, F. Arruga, I. Defilippi, E. Gottardi et al., Early prediction of treatment outcome in acute myeloid leukemia by measurement of WT1 transcript levels in peripheral blood samples collected after chemotherapy, Haematologica, vol.93, issue.6, pp.921-925, 2008.
DOI : 10.3324/haematol.12165

D. Cilloni, A. Renneville, F. Hermitte, R. Hills, S. Daly et al., Assay to Enhance Risk Stratification in Acute Myeloid Leukemia: A European LeukemiaNet Study, Journal of Clinical Oncology, vol.27, issue.31, pp.5195-201, 2009.
DOI : 10.1200/JCO.2009.22.4865

S. Freeman, P. Virgo, S. Couzens, D. Grimwade, N. Russell et al., Prognostic Relevance of Treatment Response Measured by Flow Cytometric Residual Disease Detection in Older Patients With Acute Myeloid Leukemia, Journal of Clinical Oncology, vol.31, issue.32, pp.314123-314154, 2013.
DOI : 10.1200/JCO.2013.49.1753

M. Terwijn, W. Van-putten, A. Kelder, V. Van-der-velden, R. Brooimans et al., High Prognostic Impact of Flow Cytometric Minimal Residual Disease Detection in Acute Myeloid Leukemia: Data From the HOVON/SAKK AML 42A Study, Journal of Clinical Oncology, vol.31, issue.31, pp.313889-97, 2013.
DOI : 10.1200/JCO.2012.45.9628

J. Krönke, R. Schlenk, K. Jensen, F. Tschürtz, A. Corbacioglu et al., -Mutated Acute Myeloid Leukemia: A Study From the German-Austrian Acute Myeloid Leukemia Study Group, Journal of Clinical Oncology, vol.29, issue.19, pp.2709-2725, 2011.
DOI : 10.1200/JCO.2011.35.0371

H. Ommen, S. Schnittger, J. Jovanovic, I. Ommen, H. Hasle et al., Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1, and CBFB-MYH11 acute myeloid leukemias, Blood, vol.115, issue.2, pp.198-205, 2010.
DOI : 10.1182/blood-2009-04-212530

J. Yin, O. Brien, M. Hills, R. Daly, S. Wheatley et al., Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse: results of the United Kingdom MRC AML-15 trial, Blood, vol.120, issue.14, pp.2826-2861, 2012.
DOI : 10.1182/blood-2012-06-435669

H. Ommen, P. Hokland, T. Haferlach, L. Abildgaard, T. Alpermann et al., Relapse kinetics in acute myeloid leukaemias with MLL translocations or partial tandem duplications within the MLL gene, Br J Haematol. 2014, vol.165, issue.5, pp.618-646

L. Abildgaard, H. Ommen, B. Lausen, H. Hasle, and C. Nyvold, fusion transcript in acute myeloid leukaemia, European Journal of Haematology, vol.158, issue.5, pp.394-402, 2013.
DOI : 10.1111/ejh.12156

H. Ommen, A. Touzart, E. Macintyre, W. Kern, T. Haferlach et al., -positive acute myeloid leukemia patients, European Journal of Haematology, vol.36, issue.5, pp.436-477, 2015.
DOI : 10.1111/ejh.12511

URL : https://hal.archives-ouvertes.fr/hal-00509049

M. Weisser, W. Kern, S. Rauhut, C. Schoch, W. Hiddemann et al., Prognostic impact of RT-PCR-based quantification of WT1 gene expression during MRD monitoring of acute myeloid leukemia, Leukemia, vol.25, issue.8, pp.1416-1439, 2005.
DOI : 10.1038/sj.leu.2403809

D. Jacobsohn, W. Tse, S. Chaleff, A. Rademaker, R. Duerst et al., gene expression before haematopoietic stem cell transplant in children with acute myeloid leukaemia predicts poor event-free survival, British Journal of Haematology, vol.97, issue.6, pp.669-74, 2009.
DOI : 10.1111/j.1365-2141.2009.07770.x

R. Walter, T. Gooley, B. Wood, F. Milano, M. Fang et al., Impact of Pretransplantation Minimal Residual Disease, As Detected by Multiparametric Flow Cytometry, on Outcome of Myeloablative Hematopoietic Cell Transplantation for Acute Myeloid Leukemia, Journal of Clinical Oncology, vol.29, issue.9, pp.1190-1197, 2011.
DOI : 10.1200/JCO.2010.31.8121

G. Rossi, A. Carella, M. Minervini, F. Di-nardo, C. Waure et al., Optimal time-points for minimal residual disease monitoring change on the basis of the method used in patients with acute myeloid leukemia who underwent allogeneic stem cell transplantation: A comparison between multiparameter flow cytometry and Wilms??? tumor 1 expression, Leukemia Research, vol.39, issue.2, pp.138-181, 2015.
DOI : 10.1016/j.leukres.2014.11.011

J. Gabert, E. Beillard, V. Van-der-velden, W. Bi, D. Grimwade et al., Standardization and quality control studies of ???real-time??? quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia ??? A Europe Against Cancer Program, Leukemia, vol.17, issue.12, 2003.
DOI : 10.1038/sj.leu.2403135

M. Østergaard, J. Stentoft, and P. Hokland, A real-time quantitative RT-PCR assay for monitoring DEK-CAN fusion transcripts arising from translocation t(6;9) in acute myeloid leukemia, Leukemia Research, vol.28, issue.11, pp.1213-1218, 2004.
DOI : 10.1016/j.leukres.2004.03.011

K. Tobal, L. Frost, and J. Liu-yin, Quantification of DEK-CAN fusion transcript by real-time reverse transcription polymerase reaction in patients with t(6;9) acute myeloid leukemia, Haematologica, 2004.

C. Willekens, O. Blanchet, A. Renneville, P. Cornillet-lefebvre, C. Pautas et al., Prospective long-term minimal residual disease monitoring using RQ-PCR in RUNX1-RUNX1T1-positive acute myeloid leukemia: results of the French CBF-2006 trial, Haematologica, vol.101, issue.3, pp.328-363, 2016.
DOI : 10.3324/haematol.2015.131946

D. Grimwade and S. Freeman, Defining minimal residual disease in acute myeloid leukemia: which platforms are ready for " prime time " ? Blood, pp.3345-55, 2014.
DOI : 10.1182/asheducation-2014.1.222

P. Gorello, G. Cazzaniga, F. Alberti, M. Dell-'oro, E. Gottardi et al., Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations, Leukemia, vol.106, issue.6, pp.1103-1111, 2006.
DOI : 10.1038/sj.leu.2404149

F. Buccisano, L. Maurillo, D. Principe, M. , D. Poeta et al., Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood, pp.332-373, 2012.

C. Dinardo and S. Luger, Beyond morphology, Current Opinion in Hematology, vol.19, issue.2, pp.82-90
DOI : 10.1097/MOH.0b013e3283501325

W. Kern, U. Bacher, C. Haferlach, S. Schnittger, and T. Haferlach, The role of multiparameter flow cytometry for disease monitoring in AML, Best Practice & Research Clinical Haematology, vol.23, issue.3, pp.379-90, 2010.
DOI : 10.1016/j.beha.2010.06.007

H. Larsen, A. Roug, T. Just, G. Brown, and P. Hokland, Expression of the hMICL in acute myeloid leukemiaa highly reliable disease marker at diagnosis and during follow-up, Cytometry B Clin Cytom. 2012, vol.82, issue.1, pp.3-8

A. Van-rhenen, B. Moshaver, A. Kelder, N. Feller, A. Nieuwint et al., Aberrant marker expression patterns on the CD34+CD38??? stem cell compartment in acute myeloid leukemia allows to distinguish the malignant from the normal stem cell compartment both at diagnosis and in remission, Leukemia, vol.18, issue.8, 2007.
DOI : 10.1038/sj.leu.2404754

A. Roug, H. Larsen, L. Nederby, T. Just, G. Brown et al., hMICL and CD123 in combination with a CD45/CD34/CD117 backbone - a universal marker combination for the detection of minimal residual disease in acute myeloid leukaemia, British Journal of Haematology, vol.62, issue.2, pp.212-234, 2014.
DOI : 10.1111/bjh.12614

C. Nyvold, J. Stentoft, K. Braendstrup, D. Melsvik, S. Moestrup et al., Wilms' tumor 1 mutation accumulated during therapy in acute myeloid leukemia: biological and clinical implications, Leukemia, vol.91, issue.11, pp.2051-2055, 2006.
DOI : 10.1182/blood-2005-04-1656

C. Bachas, G. Schuurhuis, I. Hollink, Z. Kwidama, B. Goemans et al., High-frequency type I/II mutational shifts between diagnosis and relapse are associated with outcome in pediatric AML: implications for personalized medicine, Blood, vol.116, issue.15, pp.2752-2760, 2010.
DOI : 10.1182/blood-2010-03-276519

J. Bibault, M. Figeac, N. Hélevaut, C. Rodriguez, S. Quief et al., Next-generation sequencing of FLT3 internal tandem duplications for minimal residual disease monitoring in acute myeloid leukemia, Oncotarget, vol.6, issue.26, pp.22812-22833, 2015.
DOI : 10.18632/oncotarget.4333

W. Chou, H. Hou, C. Liu, C. Chen, L. Lin et al., Sensitive measurement of quantity dynamics of FLT3 internal tandem duplication at early time points provides prognostic information, Annals of Oncology, vol.22, issue.3, pp.696-704, 2011.
DOI : 10.1093/annonc/mdq402

E. Abdelhamid, C. Preudhomme, N. Helevaut, O. Nibourel, C. Gardin et al., Minimal residual disease monitoring based on FLT3 internal tandem duplication in adult acute myeloid leukemia, Leukemia Research, vol.36, issue.3, 2012.
DOI : 10.1016/j.leukres.2011.11.002

N. Feller, M. Van-der-pol, A. Van-stijn, G. Weijers, A. Westra et al., MRD parameters using immunophenotypic detection methods are highly reliable in predicting survival in acute myeloid leukaemia, Leukemia, vol.18, issue.8, pp.1380-90, 2004.
DOI : 10.1038/sj.leu.2403405

D. Voskova, C. Schoch, S. Schnittger, W. Hiddemann, T. Haferlach et al., Stability of leukemiaassociated aberrant immunophenotypes in patients with acute myeloid leukemia between diagnosis and relapse: comparison with cytomorphologic, cytogenetic, and molecular genetic findings, Cytometry B Clin Cytom, vol.62, issue.1, pp.25-38, 2004.

S. Jaiswal, P. Fontanillas, J. Flannick, A. Manning, P. Grauman et al., Age-Related Clonal Hematopoiesis Associated with Adverse Outcomes, New England Journal of Medicine, vol.371, issue.26, pp.2488-98, 2014.
DOI : 10.1056/NEJMoa1408617

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4306669

G. Genovese, A. Kähler, R. Handsaker, J. Lindberg, S. Rose et al., Clonal Hematopoiesis and Blood-Cancer Risk Inferred from Blood DNA Sequence, New England Journal of Medicine, vol.371, issue.26, pp.2477-87, 2014.
DOI : 10.1056/NEJMoa1409405

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4290021

G. Pløen, L. Nederby, P. Guldberg, M. Hansen, L. Ebbesen et al., mutations at long-term remission in adult patients with AML, British Journal of Haematology, vol.43, issue.4, pp.478-86, 2014.
DOI : 10.1111/bjh.13062

H. Debarri, D. Lebon, C. Roumier, M. Cheok, A. Marceau-renaut et al., IDH1/2 but not DNMT3A mutations are suitable targets for minimal residual disease monitoring in acute myeloid leukemia patients: a study by the Acute Leukemia French Association, Oncotarget, vol.6, issue.39, pp.42345-53, 2015.

H. Ommen, Monitoring minimal residual disease in acute myeloid leukaemia: a review of the current evolving strategies, Therapeutic Advances in Hematology, vol.121, issue.1, pp.3-16
DOI : 10.1111/bjh.13062

J. Rubnitz, H. Inaba, G. Dahl, R. Ribeiro, W. Bowman et al., Minimal residual disease-directed therapy for childhood acute myeloid leukaemia: results of the AML02 multicentre trial, The Lancet Oncology, vol.11, issue.6, 2010.
DOI : 10.1016/S1470-2045(10)70090-5

H. Zhu, X. Zhang, Y. Qin, D. Liu, H. Jiang et al., MRD-directed risk stratification treatment may improve outcomes of t(8;21) AML in the first complete remission: results from the AML05 multicenter trial, Blood, vol.121, issue.20, pp.4056-62, 2013.
DOI : 10.1182/blood-2012-11-468348

S. Pozzi, S. Geroldi, E. Tedone, S. Luchetti, R. Grasso et al., expression, British Journal of Haematology, vol.83, issue.4, 2013.
DOI : 10.1111/bjh.12181

K. Sockel, M. Wermke, J. Radke, A. Kiani, M. Schaich et al., Minimal residual disease-directed preemptive treatment with azacitidine in patients with NPM1-mutant acute myeloid leukemia and molecular relapse, Haematologica, vol.96, issue.10, pp.1568-70, 2011.
DOI : 10.3324/haematol.2011.044388

URL : http://doi.org/10.3324/haematol.2011.044388

U. Platzbecker, M. Wermke, J. Radke, U. Oelschlaegel, F. Seltmann et al., Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial, Leukemia, vol.116, issue.3, pp.381-390, 2012.
DOI : 10.1038/leu.2011.234

W. Chou, K. Peng, W. Lei, B. Ko, W. Tsay et al., Persistence of mutant isocitrate dehydrogenase in patients with acute myeloid leukemia in remission, Leukemia, vol.330, issue.3, pp.527-536, 2012.
DOI : 10.1038/leu.2011.215

I. Jeziskova, F. Razga, M. Toskova, D. Dvorakova, S. Timilsina et al., gene, Leukemia & Lymphoma, vol.47, issue.4, pp.867-70
DOI : 10.1038/onc.2010.444

M. Janin, E. Mylonas, V. Saada, J. Micol, A. Renneville et al., -Mutated De Novo Acute Myeloid Leukemia: A Study by the Acute Leukemia French Association Group, Journal of Clinical Oncology, vol.32, issue.4, pp.297-305
DOI : 10.1200/JCO.2013.50.2047

URL : https://hal.archives-ouvertes.fr/halshs-00159466

X. Thomas, S. De-botton, and S. Chevret, Clofarabine-based consolidation improves relapse-free survival of younger adults with non-favorable acute myeloid leukemia (AML) in first remission: results of the randomized ALFA-0702/CLARA study, Blood, vol.126, issue.23, 2015.

D. Tholen, Protocols for determination of limits of detection and limits of quantitation: approved guideline, 2004.

K. Heinonen, K. Mrózek, D. Lawrence, D. Arthur, M. Pettenati et al., Clinical characteristics of patients with de novo acute myeloid leukaemia and isolated trisomy 11: a Cancer and Leukemia Group B study, British Journal of Haematology, vol.305, issue.3, pp.513-533, 1998.
DOI : 10.1002/(SICI)1096-8652(199705)55:1<41::AID-AJH8>3.0.CO;2-3

S. Chotirat, W. Thongnoppakhun, O. Promsuwicha, C. Boonthimat, and C. Auewarakul, Molecular alterations of isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) metabolic genes and additional genetic mutations in newly diagnosed acute myeloid leukemia patients, Journal of Hematology & Oncology, vol.5, issue.1, pp.5-230, 2012.
DOI : 10.1038/leu.2010.113

S. Whitman, The MLL partial tandem duplication: evidence for recessive gain-of-function in acute myeloid leukemia identifies a novel patient subgroup for molecular-targeted therapy, Blood, vol.106, issue.1, pp.345-52, 2005.
DOI : 10.1182/blood-2005-01-0204