, de l'incidence et de la mortalité par cancer en France entre 1980 et 2012: étude à partir des registres des cancers du réseau Francim, 2013.

, Survie des personnes atteintes de cancer en France métropolitaine 1989-2013 -Partie 2 : hémopathies malignes / 2016 / Maladies chroniques et traumatismes / Rapports et synthèses / Publications et outils / Accueil [Internet

A. R. Derolf, S. Y. Kristinsson, T. Andersson, O. Landgren, P. W. Dickman et al., Improved patient survival for acute myeloid leukemia: a population-based study of 9729 patients diagnosed in Sweden between 1973 and, Blood. 16 avr, vol.113, issue.16, pp.3666-72, 2005.

R. Liersch, C. Müller-tidow, W. E. Berdel, and U. Krug, Prognostic factors for acute myeloid leukaemia in adults -biological significance and clinical use, Br J Haematol. avr, vol.165, issue.1, pp.17-38, 2014.

F. R. Appelbaum, H. Gundacker, D. R. Head, M. L. Slovak, C. L. Willman et al., Age and acute myeloid leukemia, Blood. 1 mai, vol.107, issue.9, pp.3481-3486, 2006.

C. Schoch, W. Kern, S. Schnittger, T. Büchner, W. Hiddemann et al., The influence of age on prognosis of de novo acute myeloid leukemia differs according to cytogenetic subgroups, Haematologica. sept, vol.89, issue.9, pp.1082-90, 2004.

U. Krug, C. Röllig, A. Koschmieder, A. Heinecke, M. C. Sauerland et al., Complete remission and early death after intensive chemotherapy in patients aged 60 years or older with acute myeloid leukaemia: a web-based application for prediction of outcomes, Lancet Lond Engl. 11 déc, vol.376, issue.9757, pp.2000-2008, 2010.

J. Menzin, K. Lang, C. C. Earle, D. Kerney, and R. Mallick, The outcomes and costs of acute myeloid leukemia among the elderly, Arch Intern Med. 22 juill, vol.162, issue.14, pp.1597-603, 2002.

F. J. Giles, G. Borthakur, F. Ravandi, S. Faderl, S. Verstovsek et al., The haematopoietic cell transplantation comorbidity index score is predictive of early death and survival in patients over 60 years of age receiving induction therapy for acute myeloid leukaemia, Br J Haematol. févr, vol.136, issue.4, pp.624-631, 2007.

B. Deschler, G. Ihorst, U. Platzbecker, U. Germing, E. März et al., Parameters detected by geriatric and quality of life assessment in 195 older patients with myelodysplastic syndromes and acute myeloid leukemia are highly predictive for outcome, Haematologica. févr, vol.98, issue.2, pp.208-224, 2013.

M. L. Sorror, M. B. Maris, R. Storb, F. Baron, B. M. Sandmaier et al., Hematopoietic cell transplantation (HCT)-specific comorbidity index: a new tool for risk assessment before allogeneic HCT, Blood, vol.15, issue.8, pp.2912-2921, 2005.

G. Juliusson, P. Antunovic, A. Derolf, S. Lehmann, L. Möllgård et al., Age and acute myeloid leukemia: real world data on decision to treat and outcomes from the Swedish Acute Leukemia Registry. Blood. 30 avr, vol.113, pp.4179-87, 2009.

G. Østgård, L. S. Medeiros, B. C. Sengeløv, H. Nørgaard, M. Andersen et al., Epidemiology and Clinical Significance of Secondary and Therapy-Related Acute Myeloid Leukemia: A National Population-Based Cohort Study, J Clin Oncol Off J Am Soc Clin Oncol, vol.33, issue.31, pp.3641-3650, 2015.

E. Hulegårdh, C. Nilsson, V. Lazarevic, H. Garelius, P. Antunovic et al., Characterization and prognostic features of secondary acute myeloid leukemia in a population-based setting: a report from the Swedish Acute Leukemia Registry, Am J Hematol. mars, vol.90, issue.3, pp.208-222, 2015.

S. Kayser, K. Döhner, J. Krauter, C. Köhne, H. A. Horst et al., The impact of therapyrelated acute myeloid leukemia (AML) on outcome in 2853 adult patients with newly diagnosed AML, Blood. 17 févr, vol.117, issue.7, pp.2137-2182, 2011.

G. Borthakur, E. Lin, N. Jain, E. E. Estey, J. E. Cortes et al., Survival is poorer in patients with secondary core-binding factor acute myelogenous leukemia compared with de novo corebinding factor leukemia, Cancer. 15 juill, vol.115, issue.14, pp.3217-3238, 2009.

E. Cheung, A. J. Perissinotti, D. L. Bixby, P. W. Burke, K. M. Pettit et al., The leukemia strikes back: a review of pathogenesis and treatment of secondary AML, Ann Hematol. mars, vol.98, issue.3, pp.541-59, 2019.

W. Kern, T. Haferlach, C. Schoch, H. Loffler, W. Gassmann et al., Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia: data from the German AML Cooperative Group (AMLCG), Trial. Blood. 1 janv, vol.101, issue.1, pp.64-70, 1992.

P. J. Sung and S. M. Luger, Minimal Residual Disease in Acute Myeloid Leukemia, Curr Treat Options Oncol, vol.18, issue.1, p.1, 2017.

F. Buccisano, L. Maurillo, G. J. Schuurhuis, D. Principe, M. I. et al., The emerging role of measurable residual disease detection in AML in morphologic remission, Semin Hematol. avr, vol.56, issue.2, pp.125-155, 2019.

J. M. Bennett, D. Catovsky, M. T. Daniel, G. Flandrin, D. A. Galton et al., Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group, Br J Haematol. août, vol.33, issue.4, pp.451-459, 1976.

J. M. Bennett, D. Catovsky, M. T. Daniel, G. Flandrin, D. A. Galton et al., Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-AmericanBritish Cooperative Group, Ann Intern Med, vol.103, issue.4, pp.620-625, 1985.

N. L. Harris, E. S. Jaffe, J. Diebold, G. Flandrin, H. K. Muller-hermelink et al., The World Health Organization Classification of Neoplastic Diseases of the Hematopoietic and Lymphoid Tissues, Ann Oncol. déc, vol.10, issue.12, pp.1419-1451, 1999.

J. W. Vardiman, N. L. Harris, and R. D. Brunning, The World Health Organization (WHO) classification of the myeloid neoplasms, Blood, vol.100, issue.7, pp.2292-302, 2002.

J. W. Vardiman, J. Thiele, D. A. Arber, R. D. Brunning, M. J. Borowitz et al., The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes, Blood. 30 juill, vol.114, issue.5, pp.937-51, 2009.

D. A. Arber, A. Orazi, R. Hasserjian, J. Thiele, M. J. Borowitz et al., The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia, Blood. 19 mai, vol.127, issue.20, pp.2391-405, 2016.

D. Grimwade, R. K. Hills, A. V. 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. 22 juill, vol.116, issue.3, pp.354-65, 2010.

M. L. Slovak, K. J. Kopecky, P. A. Cassileth, D. H. Harrington, K. S. Theil et al., Karyotypic analysis predicts outcome of preremission and postremission therapy in adult acute myeloid leukemia: a Southwest Oncology Group/Eastern Cooperative Oncology Group Study, Blood. 15 déc, vol.96, issue.13, pp.4075-83, 2000.

J. C. Byrd, K. Mrózek, R. K. Dodge, A. J. Carroll, C. G. Edwards et al., Pretreatment cytogenetic abnormalities are predictive of induction success, cumulative incidence of relapse, and overall survival in adult patients with de novo acute myeloid leukemia: results from Cancer and Leukemia Group B (CALGB 8461), Blood. 15 déc, vol.100, issue.13, pp.4325-4361, 2002.

K. Mrózek, N. A. Heerema, and C. D. Bloomfield, Cytogenetics in acute leukemia, Blood Rev. juin, vol.18, issue.2, pp.115-151, 2004.

H. Döhner, E. H. Estey, S. Amadori, F. R. Appelbaum, T. Büchner et al., Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet, Blood. 21 janv, vol.115, issue.3, pp.453-74, 2010.

H. Döhner, E. Estey, D. Grimwade, S. Amadori, F. R. Appelbaum et al., Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel, Blood, vol.26, issue.4, pp.424-471, 2017.

R. F. Schlenk, K. Döhner, J. Krauter, S. Fröhling, A. Corbacioglu et al., Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia, N Engl J Med. 1 mai, vol.358, issue.18, pp.1909-1927, 2008.

H. Dombret and C. Gardin, An update of current treatments for adult acute myeloid leukemia, Blood. 7 janv, vol.127, issue.1, pp.53-61, 2016.

H. Döhner, D. J. Weisdorf, and C. D. Bloomfield, Acute Myeloid Leukemia. Longo DL, éditeur, N Engl J Med. 17 sept, vol.373, issue.12, pp.1136-52, 2015.

H. F. Fernandez, Z. Sun, X. Yao, M. R. Litzow, S. M. Luger et al., Anthracycline dose intensification in acute myeloid leukemia, N Engl J Med. 24 sept, vol.361, issue.13, pp.1249-59, 2009.

J. P. Patel, M. Gönen, M. E. Figueroa, H. Fernandez, Z. Sun et al., Prognostic relevance of integrated genetic profiling in acute myeloid leukemia, N Engl J Med. 22 mars, vol.366, issue.12, pp.1079-89, 2012.

M. G. Inc, RANDOMIZED COMPARISON OF 90 MG VERSUS 60 MG DAUNORUBICIN IN 7+3

D. Sur,

E. Berman, V. Raymond, A. Daghestani, Z. A. Arlin, T. S. Gee et al., 4-demethoxydaunorubicin (idarubicin) in combination with 1-beta-D-arabinofuranosylcytosine in the treatment of relapsed or refractory acute leukemia, Cancer Res. 15 janv, vol.49, issue.2, pp.477-81, 1989.

E. Berman and M. Mcbride, Comparative cellular pharmacology of daunorubicin and idarubicin in human multidrug-resistant leukemia cells, Blood. 15 juin, vol.79, issue.12, pp.3267-73, 1992.

F. Mandelli, M. Vignetti, S. Suciu, R. Stasi, M. Petti et al., Daunorubicin versus mitoxantrone versus idarubicin as induction and consolidation chemotherapy for adults with acute myeloid leukemia: the EORTC and GIMEMA Groups Study AML-10, J Clin Oncol Off J Am Soc Clin Oncol, vol.27, issue.32, pp.5397-403, 2009.

B. Lioure, M. C. Bene, A. Pigneux, A. Huynh, P. Chevallier et al., Early matched sibling hematopoietic cell transplantation for adult AML in first remission using an age-adapted strategy: long-term results of a prospective GOELAMS study, Blood. 22 mars, vol.119, issue.12, pp.2943-2951, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00849702

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, J Clin Oncol Off J Am Soc Clin Oncol. 20 janv, vol.31, issue.3, pp.321-328, 2013.

C. Récher, M. C. Béné, B. Lioure, A. Pigneux, N. Vey et al., Long-term results of a randomized phase 3 trial comparing idarubicin and daunorubicin in younger patients with acute myeloid leukaemia, Leukemia. févr, vol.28, issue.2, pp.440-443, 2014.

H. Liu, R. Fu, L. Li, G. Wang, J. Song et al., Comparison of Reduced-Intensity Idarubicin and Daunorubicin Plus Cytarabine as Induction Chemotherapy for Elderly Patients with Newly Diagnosed Acute Myeloid Leukemia, Clin Drug Investig. févr, vol.37, issue.2, pp.167-74, 2017.

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, Lancet Lond Engl. 21 avr, vol.379, issue.9825, pp.1508-1524, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01056520

A. K. Burnett, R. K. 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, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.29, issue.4, pp.369-77, 2011.

J. E. Lancet, G. L. Uy, J. E. Cortes, L. F. Newell, T. L. Lin et al., Final results of a phase III randomized trial of CPX-351 versus 7+3 in older patients with newly diagnosed high risk (secondary) AML, J Clin Oncol. 20 mai, vol.34, pp.7000-7000, 2016.

R. M. Stone, S. J. Mandrekar, B. L. Sanford, K. Laumann, S. Geyer et al., Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation, N Engl J Med. 3 août, vol.377, issue.5, pp.454-64, 2017.

R. J. Mayer, R. B. Davis, C. A. Schiffer, D. T. Berg, B. L. Powell et al., Intensive postremission chemotherapy in adults with acute myeloid leukemia. Cancer and Leukemia Group B, N Engl J Med, vol.331, issue.14, pp.896-903, 1994.

B. Löwenberg, T. Pabst, E. Vellenga, W. Van-putten, H. C. Schouten et al., Cytarabine dose for acute myeloid leukemia, N Engl J Med. 17 mars, vol.364, issue.11, pp.1027-1063, 2011.

A. K. Burnett, R. K. Hills, D. W. Milligan, A. H. Goldstone, A. G. Prentice et al., Attempts to optimize induction and consolidation treatment in acute myeloid leukemia: results of the MRC AML12 trial, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.28, issue.4, pp.586-95, 2010.

A. K. Burnett, N. H. Russell, R. K. Hills, A. E. Hunter, L. Kjeldsen et al., Optimization of chemotherapy for younger patients with acute myeloid leukemia: results of the medical research council AML15 trial, J Clin Oncol Off J Am Soc Clin Oncol. 20 sept, vol.31, issue.27, pp.3360-3368, 2013.

S. Miyawaki, S. Ohtake, S. Fujisawa, H. Kiyoi, K. Shinagawa et al., A randomized comparison of 4 courses of standard-dose multiagent chemotherapy versus 3 courses of high-dose cytarabine alone in postremission therapy for acute myeloid leukemia in adults: the JALSG AML201 Study, Blood. 24 févr, vol.117, issue.8, pp.2366-72, 2011.

X. Thomas, M. Elhamri, E. Raffoux, A. Renneville, C. Pautas et al., Comparison of high-dose cytarabine and timed-sequential chemotherapy as consolidation for younger adults with AML in first remission: the ALFA-9802 study, Blood. 18 août, vol.118, issue.7, pp.1754-62, 2011.

J. Koreth, R. Schlenk, K. J. Kopecky, S. Honda, J. Sierra et al., Allogeneic stem cell transplantation for acute myeloid leukemia in first complete remission: systematic review and metaanalysis of prospective clinical trials, JAMA. 10 juin, vol.301, issue.22, pp.2349-61, 2009.

J. J. Cornelissen, D. Breems, W. Van-putten, A. A. Gratwohl, J. R. Passweg et al.,

, Comparative analysis of the value of allogeneic hematopoietic stem-cell transplantation in acute myeloid leukemia with monosomal karyotype versus other cytogenetic risk categories, J Clin Oncol Off J Am Soc Clin Oncol. 10 juin, vol.30, issue.17, pp.2140-2146, 2012.

A. Brands-nijenhuis, M. Labopin, H. C. Schouten, L. Volin, G. Socié et al.,

, Monosomal karyotype as an adverse prognostic factor in patients with acute myeloid leukemia treated with allogeneic hematopoietic stem-cell transplantation in first complete remission: a retrospective survey on behalf of the ALWP of the EBMT, Haematologica. févr, vol.101, issue.2, pp.248-55, 2016.

J. A. Webster and K. W. Pratz, Acute myeloid leukemia in the elderly: therapeutic options and choice, Leuk Lymphoma, vol.59, issue.2, pp.274-87, 2018.

F. Ravandi, E. K. Ritchie, H. Sayar, J. E. Lancet, M. D. Craig et al., Vosaroxin plus cytarabine versus placebo plus cytarabine in patients with first relapsed or refractory acute myeloid leukaemia (VALOR): a randomised, controlled, double-blind, multinational, phase 3 study, Lancet Oncol. sept, vol.16, issue.9, pp.1025-1061, 2015.

. Ii-p, Final safety and efficacy data from the phase III QuANTUM-R study -oral abstract from ASH, 2018.

D. Sur,

D. G. Gilliland, C. T. Jordan, and C. A. Felix, The molecular basis of leukemia, Hematol Am Soc Hematol Educ Program, pp.80-97, 2004.

, Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia, N Engl J Med. 30 mai, vol.368, issue.22, pp.2059-74, 2013.

D. G. Gilliland and J. D. Griffin, The roles of FLT3 in hematopoiesis and leukemia, Blood. 1 sept, vol.100, issue.5, pp.1532-1574, 2002.

M. Nakao, S. Yokota, T. Iwai, H. Kaneko, S. Horiike et al., Internal tandem duplication of the flt3 gene found in acute myeloid leukemia, Leukemia. déc, vol.10, issue.12, pp.1911-1919, 1996.

C. Thiede, C. Steudel, B. Mohr, M. Schaich, U. Schäkel et al., Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis, Blood. 15 juin, vol.99, issue.12, pp.4326-4361, 2002.

P. D. Kottaridis, R. E. Gale, M. E. Frew, G. Harrison, S. E. Langabeer et al., The presence of a FLT3 internal tandem duplication in patients with acute myeloid leukemia (AML) adds important prognostic information to cytogenetic risk group and response to the first cycle of chemotherapy: analysis of 854 patients from the United Kingdom Medical Research Council AML 10 and 12 trials, Blood. 15 sept, vol.98, issue.6, pp.1752-1761, 2001.

Y. Yamamoto, H. Kiyoi, Y. Nakano, R. Suzuki, Y. Kodera et al., Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies, Blood. 15 avr, vol.97, issue.8, pp.2434-2443, 2001.

M. Yanada, K. Matsuo, T. Suzuki, H. Kiyoi, and T. Naoe, Prognostic significance of FLT3 internal tandem duplication and tyrosine kinase domain mutations for acute myeloid leukemia: a metaanalysis, Leukemia. août, vol.19, issue.8, pp.1345-1354, 2005.

R. E. Gale, C. Green, C. Allen, A. J. Mead, A. K. Burnett et al., The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia, Blood. 1 mars, vol.111, issue.5, pp.2776-84, 2008.

S. Liu, H. Dong, X. Bao, Q. Qiu, H. Li et al., Impact of FLT3-ITD length on prognosis of acute myeloid leukemia, Haematologica. 3 août, 2018.

S. Kayser, R. F. Schlenk, M. C. Londono, F. Breitenbuecher, K. Wittke et al., Insertion of FLT3 internal tandem duplication in the tyrosine kinase domain-1 is associated with resistance to chemotherapy and inferior outcome, Blood. 17 sept, vol.114, issue.12, pp.2386-92, 2009.

M. Beran, R. Luthra, H. Kantarjian, and E. Estey, FLT3 mutation and response to intensive chemotherapy in young adult and elderly patients with normal karyotype, Leuk Res. juin, vol.28, issue.6, pp.547-50, 2004.

A. Renneville, C. Roumier, V. Biggio, O. Nibourel, N. Boissel et al., Cooperating gene mutations in acute myeloid leukemia: a review of the literature, Leukemia. mai, vol.22, issue.5, pp.915-946, 2008.

X. Liu, Q. Ye, X. Zhao, P. Zhang, S. Li et al., RAS mutations in acute myeloid leukaemia patients: A review and meta-analysis, Clin Chim Acta. févr, vol.489, pp.254-60, 2019.

J. E. Boissel, N. Chevret, S. Delabesse, E. Renneville, A. Cornillet et al., Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia, Blood. 21 mars, vol.121, issue.12, pp.2213-2236, 2013.

M. Ko, Y. Huang, A. M. Jankowska, U. J. Pape, M. Tahiliani et al., Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2, Nature. 9 déc, vol.468, issue.7325, pp.839-882, 2010.

, Mutation in TET2 in myeloid cancers

D. Sur,

W. Chou, S. Chou, C. Liu, C. Chen, H. Hou et al., TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics, Blood, vol.118, issue.14, pp.3803-3813, 2011.

T. Zhang, J. Zhou, Y. Wang, Y. Wen, X. Guo et al., TET2 expression is a potential prognostic and predictive biomarker in cytogenetically normal acute myeloid leukemia, J Cell Physiol. août, vol.233, issue.8, pp.5838-5884, 2018.

, TET2 mutations improve the new European LeukemiaNet risk classification of acute myeloid leukemia: a Cancer and Leukemia Group B study. -PubMed -NCBI

D. Sur,

M. T. Cedena, I. Rapado, A. Santos-lozano, R. Ayala, E. Onecha et al., Mutations in the DNA methylation pathway and number of driver mutations predict response to azacitidine in myelodysplastic syndromes, Oncotarget, vol.8, issue.63, pp.106948-61, 2017.

X. Yan, J. Xu, Z. Gu, C. Pan, G. Lu et al., Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia, Nat Genet. 13 mars, vol.43, issue.4, pp.309-324, 2011.

F. Thol, F. Damm, A. Lüdeking, C. Winschel, K. Wagner et al., Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia, J Clin Oncol Off J Am Soc Clin Oncol. 20 juill, vol.29, issue.21, pp.2889-96, 2011.

, Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation

D. Sur,

R. J. Molenaar, S. Thota, Y. Nagata, B. Patel, M. Clemente et al., Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms, Leukemia. nov, vol.29, issue.11, pp.2134-2176, 2015.

J. Mondesir, C. Willekens, M. Touat, S. Botton, and . De, IDH1 and IDH2 mutations as novel therapeutic targets: current perspectives, J Blood Med, vol.7, p.171, 2016.

M. L. Amaya and D. A. Pollyea, Targeting the IDH2 Pathway in Acute Myeloid Leukemia, Clin Cancer Res. 16 mai, 2018.

C. L. Green, C. M. Evans, L. Zhao, R. K. Hills, A. K. Burnett et al., The prognostic significance of IDH2 mutations in AML depends on the location of the mutation, Blood. 14 juill, vol.118, issue.2, pp.409-421, 2011.

M. Janin, E. Mylonas, V. Saada, J. Micol, A. Renneville et al., Serum 2-hydroxyglutarate production in IDH1-and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute Leukemia French Association group, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.32, issue.4, pp.297-305, 2014.

D. G. Gilliland, Molecular genetics of human leukemias: new insights into therapy. Semin Hematol, vol.39, pp.6-11, 2002.

M. Osato, N. Asou, E. Abdalla, K. Hoshino, H. Yamasaki et al., Biallelic and heterozygous point mutations in the runt domain of the AML1/PEBP2alphaB gene associated with myeloblastic leukemias, Blood. 15 mars, vol.93, issue.6, pp.1817-1841, 1999.

C. Nerlov, EBPalpha mutations in acute myeloid leukaemias, Nat Rev Cancer. mai, vol.4, issue.5, pp.394-400, 2004.

A. Dufour, F. Schneider, K. H. Metzeler, E. Hoster, S. Schneider et al., Acute myeloid leukemia with biallelic CEBPA gene mutations and normal karyotype represents a distinct genetic entity associated with a favorable clinical outcome, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.28, issue.4, pp.570-577, 2010.

, Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that, PubMed -NCBI

U. Bacher, W. Kern, S. Schnittger, W. Hiddemann, T. Haferlach et al., Population-based agespecific incidences of cytogenetic subgroups of acute myeloid leukemia. Haematologica, vol.90, pp.1502-1512, 2005.

V. Gelsi-boyer, V. Trouplin, J. Adélaïde, J. Bonansea, N. Cervera et al., Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia, Br J Haematol. juin, vol.145, issue.6, pp.788-800, 2009.

A. Adli, M. Lafave, L. M. Gao, J. Hricik, T. Shih et al., ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression, Cancer Cell. 14 août, vol.22, issue.2, pp.180-93, 2012.

D. Inoue, J. Kitaura, K. Togami, K. Nishimura, Y. Enomoto et al., Myelodysplastic syndromes are induced by histone methylation-altering ASXL1 mutations, J Clin Invest, vol.123, issue.11, pp.4627-4667, 2013.

B. Falini, I. Nicoletti, M. F. Martelli, and C. Mecucci, Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features, Blood. 1 févr, vol.109, issue.3, pp.874-85, 2007.

, Prevalence, clinical profile, and prognosis of NPM mutations in AML with normal karyotype

D. Sur,

K. Döhner, R. F. Schlenk, M. Habdank, C. Scholl, F. G. Rücker et al., Mutant nucleophosmin (NPM1) predicts favorable prognosis in younger adults with acute myeloid leukemia and normal cytogenetics: interaction with other gene mutations, Blood. 1 déc, vol.106, issue.12, pp.3740-3746, 2005.

, Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): association with other gene abnormalities and previously established gene expres, PubMed -NCBI

, Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype

D. Sur,

, Nucleophosmin mutations in de novo acute myeloid leukemia: the age-dependent incidences and the stability during disease evolution

M. J. Burke, Minimal Residual Disease in NPM1-Mutated AML, N Engl J Med. 4 févr, vol.374, issue.5, pp.481-483, 2016.

A. J. Levine, J. Momand, and C. A. Finlay, The p53 tumour suppressor gene, Nature. 6 juin, vol.351, issue.6326, pp.453-459, 1991.

D. Bowen, M. J. Groves, A. K. Burnett, Y. Patel, C. Allen et al., TP53 gene mutation is frequent in patients with acute myeloid leukemia and complex karyotype, and is associated with very poor prognosis, Leukemia. janv, vol.23, issue.1, pp.203-209, 2009.

, TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal, PubMed -NCBI

A. Letai, Functional precision cancer medicine-moving beyond pure genomics, Nat Med. 8 sept, vol.23, issue.9, pp.1028-1063, 2017.

G. Barbany, M. Höglund, B. Simonsson, C. Swedish, and . Group, Complete molecular remission in chronic myelogenous leukemia after imatinib therapy, N Engl J Med. 15 août, vol.347, issue.7, pp.539-579, 2002.

F. Montemurro, G. Valabrega, and M. Aglietta, Trastuzumab treatment in breast cancer, N Engl J Med. 18 mai, vol.354, p.2186, 1920.

J. A. Incorvati, S. Shah, Y. Mu, and J. Lu, Targeted therapy for HER2 positive breast cancer, J Hematol OncolJ Hematol Oncol. 3 juin, vol.6, p.38, 2013.

C. Robert, B. Karaszewska, J. Schachter, P. Rutkowski, A. Mackiewicz et al., Improved overall survival in melanoma with combined dabrafenib and trametinib, N Engl J Med. 1 janv, vol.372, issue.1, pp.30-39, 2015.

C. L. O'bryant, S. D. Wenger, M. Kim, and L. A. Thompson, Crizotinib: a new treatment option for ALKpositive non-small cell lung cancer, Ann Pharmacother. févr, vol.47, issue.2, pp.189-97, 2013.

A. Buzyn, J. Blay, N. Hoog-labouret, M. Jimenez, F. Nowak et al., Equal access to innovative therapies and precision cancer care, Nat Rev Clin Oncol. 22 mars, vol.13, p.385, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01792383

H. West and . Jack, Novel Precision Medicine Trial Designs: Umbrellas and Baskets, JAMA Oncol. 1 mars, vol.3, issue.3, p.423, 2017.

V. Hoff, D. D. Stephenson, J. J. Rosen, P. Loesch, D. M. Borad et al., Pilot study using molecular profiling of patients' tumors to find potential targets and select treatments for their refractory cancers, J Clin Oncol Off J Am Soc Clin Oncol, vol.28, issue.33, pp.4877-83, 2010.

A. Tsimberidou, S. Wen, D. S. Hong, J. J. Wheler, G. S. Falchook et al., Personalized Medicine for Patients with Advanced Cancer in the Phase I Program at MD Anderson: Validation and Landmark Analyses, Clin Cancer Res. 15 sept, vol.20, issue.18, pp.4827-4863, 2014.

C. Massard, S. Michiels, C. Ferté, L. Deley, M. Lacroix et al., High-Throughput Genomics and Clinical Outcome in Hard-to-Treat Advanced Cancers

, Trial. Cancer Discov, vol.7, issue.6, pp.586-95, 2017.

M. Schwaederle, M. Zhao, J. J. Lee, A. M. Eggermont, R. L. Schilsky et al., Impact of Precision Medicine in Diverse Cancers: A Meta-Analysis of Phase II Clinical Trials, J Clin Oncol, vol.33, issue.32, pp.3817-3842, 2015.

D. L. Jardim, F. Jardim, D. L. Schwaederle, M. Wei, C. Lee et al., Impact of a Biomarker-Based Strategy on Oncology Drug Development: A Meta-analysis of Clinical Trials Leading to FDA Approval, J Natl Cancer Inst, vol.107, issue.11, 2015.

L. Tourneau, C. Delord, J. Gonçalves, A. Gavoille, C. Dubot et al., Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): a multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial, Lancet Oncol, vol.16, issue.13, pp.1324-1358, 2015.

A. M. Tsimberidou and R. Kurzrock, Precision medicine: lessons learned from the SHIVA trial, Lancet Oncol. déc, vol.16, issue.16, pp.579-80, 2015.

V. Prasad and R. P. Gale, Precision medicine in acute myeloid leukemia: Hope, hype or both?, Leuk Res, vol.48, pp.73-80, 2016.

, Thérapies ciblées : modes d'action -Thérapies ciblées et immunothérapie spécifique

D. Sur,

M. E. Huang, Y. C. Ye, S. R. Chen, J. R. Chai, J. X. Lu et al., Use of all-trans retinoic acid in the treatment of acute promyelocytic leukemia, Blood. août, vol.72, issue.2, pp.567-72, 1988.

L. Degos, H. Dombret, C. Chomienne, M. T. Daniel, J. M. Micléa et al., All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia, Blood. 15 mai, vol.85, issue.10, pp.2643-53, 1995.

S. R. Bohl, L. Bullinger, and F. G. Rücker, New Targeted Agents in Acute Myeloid Leukemia: New Hope on the Rise, Int J Mol Sci. 23 avr, vol.20, issue.8, 2019.

P. P. Zarrinkar, R. N. Gunawardane, M. D. Cramer, M. F. Gardner, D. Brigham et al., AC220 is a uniquely potent and selective inhibitor of FLT3 for the treatment of acute myeloid leukemia (AML), Blood, vol.114, issue.14, pp.2984-92, 2009.

M. Wu, C. Li, and X. Zhu, FLT3 inhibitors in acute myeloid leukemia, J Hematol OncolJ Hematol Oncol, vol.4, issue.2018

Y. Alvarado, H. M. Kantarjian, R. Luthra, F. Ravandi, G. Borthakur et al., Treatment with FLT3 inhibitor in patients with FLT3 -mutated acute myeloid leukemia is associated with development of secondary FLT3 -tyrosine kinase domain mutations: FLT3 Inhibitor Treatment in Mutated AML, Cancer. 15 juill, vol.120, issue.14, pp.2142-2151, 2014.

S. D. Baker, E. I. Zimmerman, Y. Wang, S. Orwick, D. S. Zatechka et al., Emergence of Polyclonal FLT3 Tyrosine Kinase Domain Mutations during Sequential Therapy with Sorafenib and Sunitinib in FLT3-ITD-Positive Acute Myeloid Leukemia, Clin Cancer Res, vol.19, issue.20, pp.5758-68, 2013.

J. Hasskarl and . Sorafenib, Recent Results Cancer Res Fortschritte Krebsforsch Progres Dans Rech Sur Cancer, vol.184, pp.61-70, 2010.

O. Abrams, T. J. Yuen, H. A. Ngai, T. J. Louie, S. G. Yee et al., SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo, Blood. 1 mai, vol.101, issue.9, pp.3597-605, 2003.

W. Zhang, M. Konopleva, Y. Shi, T. Mcqueen, D. Harris et al., Mutant FLT3: a direct target of sorafenib in acute myelogenous leukemia, J Natl Cancer Inst. 6 févr, vol.100, issue.3, 2008.

M. Crump, D. Hedley, S. Kamel-reid, B. Leber, R. Wells et al., A randomized phase I clinical and biologic study of two schedules of sorafenib in patients with myelodysplastic syndrome or acute myeloid leukemia: a NCIC (National Cancer Institute of Canada, Clinical Trials Group Study. Leuk Lymphoma. févr, vol.51, issue.2, pp.252-60, 2010.

F. Ravandi, J. E. Cortes, D. Jones, S. Faderl, G. Garcia-manero et al., Phase I/II study of combination therapy with sorafenib, idarubicin, and cytarabine in younger patients with acute myeloid leukemia, J Clin Oncol Off J Am Soc Clin Oncol. 10 avr, vol.28, issue.11, pp.1856-62, 2010.

G. L. Uy, S. J. Mandrekar, K. Laumann, G. Marcucci, W. Zhao et al., A phase 2 study incorporating sorafenib into the chemotherapy for older adults with FLT3 -mutated acute myeloid leukemia: CALGB 11001, Blood Adv. 24 janv, vol.1, issue.5, pp.331-371, 2017.

C. Röllig, H. Serve, A. Hüttmann, R. Noppeney, C. Müller-tidow et al., Addition of sorafenib versus placebo to standard therapy in patients aged 60 years or younger with newly diagnosed acute myeloid leukaemia (SORAML): a multicentre, phase 2, randomised controlled trial, Lancet Oncol. déc, vol.16, issue.16, pp.1691-1700, 2015.

A. Tschan-plessl, J. P. Halter, D. Heim, M. Medinger, J. R. Passweg et al., Synergistic effect of sorafenib and cGvHD in patients with high-risk FLT3-ITD+AML allows long-term disease control after allogeneic transplantation, Ann Hematol, vol.94, issue.11, pp.1899-905, 2015.

L. Xuan, Y. Wang, F. Huang, E. Jiang, L. Deng et al., Effect of sorafenib on the outcomes of patients with FLT3-ITD acute myeloid leukemia undergoing allogeneic hematopoietic stem cell transplantation, Cancer. 1 mai, vol.124, issue.9, pp.1954-63, 2018.

A. M. Brunner, S. Li, A. T. Fathi, M. Wadleigh, V. T. Ho et al., Haematopoietic cell transplantation with and without sorafenib maintenance for patients with FLT3 -ITD acute myeloid leukaemia in first complete remission, Br J Haematol, vol.175, issue.3, pp.496-504, 2016.

. Ii-p, SORMAIN study of maintenance therapy with sorafenib in patients with FLT3-ITD acute myeloid leukemia -oral abstract from ASH, 2018.

D. Sur,

Y. Zhang, X. L. Fan, Z. Huang, F. Jiang, Q. Xu et al., Sorafenib as salvage therapy in refractory relapsed acute myeloid leukemia with positive FLT3 mutation

, Zhonghua Xue Ye Xue Za Zhi Zhonghua Xueyexue Zazhi. avr, vol.37, issue.4, pp.292-298, 2016.

S. K. Metzelder, T. Schroeder, M. Lübbert, M. Ditschkowski, K. Götze et al., Long-term survival of sorafenib-treated FLT3-ITD-positive acute myeloid leukaemia patients relapsing after allogeneic stem cell transplantation, Eur J Cancer, vol.86, pp.233-242, 2017.

M. R. Muppidi, S. Portwood, E. A. Griffiths, J. E. Thompson, L. A. Ford et al., Decitabine and Sorafenib Therapy in FLT-3 ITD-Mutant Acute Myeloid Leukemia, Clin Lymphoma Myeloma Leuk. juin, vol.15, pp.73-82, 2015.

F. Ravandi, M. L. Alattar, M. R. Grunwald, M. A. Rudek, T. Rajkhowa et al., Phase 2 study of azacytidine plus sorafenib in patients with acute myeloid leukemia and FLT-3 internal tandem duplication mutation, Blood. 6 juin, vol.121, issue.23, pp.4655-62, 2013.

B. W. Cooper, T. L. Kindwall-keller, M. D. Craig, R. J. Creger, M. Hamadani et al., A phase I study of midostaurin and azacitidine in relapsed and elderly AML patients, Clin Lymphoma Myeloma Leuk. juill, vol.15, issue.7, pp.428-432, 2015.

P. Strati, H. Kantarjian, F. Ravandi, A. Nazha, G. Borthakur et al., Phase I/II trial of the combination of midostaurin (PKC412) and 5-azacytidine for patients with acute myeloid leukemia and myelodysplastic syndrome, Am J Hematol. avr, vol.90, issue.4, pp.276-81, 2015.

C. B. Williams, S. Kambhampati, W. Fiskus, J. Wick, C. Dutreix et al., Preclinical and phase I results of decitabine in combination with midostaurin (PKC412) for newly diagnosed elderly or relapsed/refractory adult patients with acute myeloid leukemia, Pharmacotherapy. déc, vol.33, issue.12, pp.1341-52, 2013.

D. B. Mendel, A. D. Laird, X. Xin, S. G. Louie, J. G. Christensen et al., In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and plateletderived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship, Clin Cancer Res Off J Am Assoc Cancer Res. janv, vol.9, issue.1, pp.327-364, 2003.

W. Fiedler, A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease, Blood. 28 sept, vol.105, issue.3, pp.986-93, 2004.

W. Fiedler, S. Kayser, M. Kebenko, M. Janning, J. Krauter et al., A phase I/II study of sunitinib and intensive chemotherapy in patients over 60 years of age with acute myeloid leukaemia and activating FLT3 mutations, Br J Haematol. juin, vol.169, issue.5, pp.694-700, 2015.

K. M. Kampa-schittenhelm, M. C. Heinrich, F. Akmut, H. Döhner, K. Döhner et al.,

. Quizartinib, AC220) is a potent second generation class III tyrosine kinase inhibitor that displays a distinct inhibition profile against mutant-FLT3, -PDGFRA and -KIT isoforms, Mol Cancer, vol.12, issue.1, p.19, 2013.

J. E. Cortes, A. E. Perl, H. Dombret, H. Dohner, B. Steffen et al., Response rate and bridging to hematopoietic stem cell transplantation (HSCT) with quizartinib (AC220) in patients with FLT3-ITD positive or negative relapsed/refractory AML after second-line chemotherapy or previous bone marrow transplant, J Clin Oncol. 20 mai, vol.31, pp.7012-7012, 2013.

A. E. Perl, H. Dohner, P. H. Rousselot, J. Marie, G. Martinelli et al., Efficacy and safety of quizartinib (AC220) in patients age ? 70 years with FLT3-ITD positive or negative relapsed/refractory acute myeloid leukemia (AML), J Clin Oncol. 20 mai, vol.31, pp.7023-7023, 2013.

, Final results of a randomized phase 2 study showing the clinical benefit of quizartinib (AC220) in patients with FLT3-ITD positive relapsed or refractory acute myeloid leukemia, | Journal of Clinical Oncology

, 160. Laboratory and Clinical Investigations to Identify the Optimal Dosing Strategy for Quizartinib (AC220) Monotherapy in FLT3-Itd-Positive (+) Relapsed/Refractory (R/R) Acute Myeloid Leukemia (AML) | Blood Journal

D. Sur,

, The benefit of treatment with quizartinib and subsequent bridging to HSCT for FLT3-ITD(+) patients with AML, | Journal of Clinical Oncology

D. Sur,

J. K. Altman, J. M. Foran, K. W. Pratz, D. Trone, J. E. Cortes et al., Phase 1 study of quizartinib in combination with induction and consolidation chemotherapy in patients with newly diagnosed acute myeloid leukemia, Am J Hematol. févr, vol.93, issue.2, pp.213-234, 2018.

, The Combination of Quizartinib with Azacitidine or Low Dose Cytarabine Is Highly Active in Patients (Pts) with FLT3-ITD Mutated Myeloid Leukemias: Interim Report of a Phase I/II Trial | Blood Journal

K. M. Kampa-schittenhelm, J. Frey, L. A. Haeusser, B. Illing, A. A. Pavlovsky et al., Crenolanib is a type I tyrosine kinase inhibitor that inhibits mutant KIT D816 isoforms prevalent in systemic mastocytosis and core binding factor leukemia, Oncotarget, vol.8, issue.10, 2017.

E. I. Zimmerman, D. C. Turner, J. Buaboonnam, S. Hu, S. Orwick et al., Crenolanib is active against models of drug-resistant FLT3-ITD-positive acute myeloid leukemia, Blood, vol.21, issue.22, pp.3607-3622, 2013.

, Low Relapse Rate in Younger Patients ? 60 Years Old with Newly Diagnosed FLT3-Mutated Acute Myeloid Leukemia (AML) Treated with Crenolanib and Cytarabine/Anthracycline Chemotherapy | Blood Journal

. Crenolanib and . Tki, Can be Safely Combined with Cytarabine and Anthracycline Induction Chemotherapy and Results in High Response Rates in Patients with Newly Diagnosed FLT3 Mutant Acute Myeloid Leukemia (AML) | Blood Journal

D. Sur,

, Safety Study of Salvage Chemotherapy High-Dose Ara-C/Mitoxantrone (HAM) and Type I FLT3-TKI Crenolanib in First Relapsed/Primary Refractory AML | Blood Journal

D. Sur,

M. Ohanian, H. M. Kantarjian, G. Borthakur, T. M. Kadia, M. Konopleva et al., Efficacy of a Type I FLT3 Inhibitor, Crenolanib, with Idarubicin and High-Dose Ara-C in Multiply Relapsed/Refractory FLT3+ AML, Blood. 2 déc, vol.128, issue.22, pp.2744-2744, 2016.

J. K. Randhawa, H. M. Kantarjian, G. Borthakur, P. A. Thompson, M. Konopleva et al., Results of a Phase II Study of Crenolanib in Relapsed/Refractory Acute Myeloid Leukemia Patients (Pts) with Activating FLT3 Mutations, Blood. 6 déc, vol.124, issue.21, pp.389-389, 2014.

H. Jetani, I. Garcia-cadenas, T. Nerreter, S. Thomas, J. Rydzek et al., CAR T-cells targeting FLT3 have potent activity against FLT3?ITD+ AML and act synergistically with the FLT3-inhibitor crenolanib, Leukemia. mai, vol.32, issue.5, pp.1168-79, 2018.

M. Mori, N. Kaneko, Y. Ueno, M. Yamada, R. Tanaka et al., Gilteritinib, a FLT3/AXL inhibitor, shows antileukemic activity in mouse models of FLT3 mutated acute myeloid leukemia, Invest New Drugs, vol.35, issue.5, pp.556-65, 2017.

L. Y. Lee, D. Hernandez, T. Rajkhowa, S. C. Smith, J. R. Raman et al., Preclinical studies of gilteritinib, a next-generation FLT3 inhibitor, Blood, vol.12, issue.2, pp.257-60, 2017.

. Ii-p, Combination of gilteritinib with induction and consolidation chemotherapy for the treatment of patients with newly diagnosed acute myeloid leukemia

D. Sur,

S. Dhillon and . Gilteritinib, First Global Approval. Drugs. févr, vol.79, issue.3, pp.331-340, 2019.

B. K. Ragon and C. D. Dinardo, Targeting IDH1 and IDH2 Mutations in Acute Myeloid Leukemia, Curr Hematol Malig Rep. déc, vol.12, issue.6, pp.537-583, 2017.

C. D. Dinardo, E. M. Stein, S. De-botton, G. J. Roboz, J. K. Altman et al., Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML, N Engl J Med. 21 juin, vol.378, issue.25, pp.2386-98, 2018.

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. 3 mai, vol.340, issue.6132, pp.622-628, 2013.

K. Yen, J. Travins, F. Wang, M. D. David, E. Artin et al., AG-221, a First-in-Class Therapy Targeting Acute Myeloid Leukemia Harboring Oncogenic IDH2 Mutations, Cancer Discov. mai, vol.7, issue.5, pp.478-93, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01568357

M. D. Amatangelo, L. Quek, A. Shih, E. M. Stein, M. Roshal et al., Enasidenib induces acute myeloid leukemia cell differentiation to promote clinical response, Blood, vol.10, issue.6, pp.732-773, 2017.

E. M. Stein, C. D. Dinardo, D. A. Pollyea, A. T. Fathi, G. J. Roboz et al., Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia, Blood, vol.10, issue.6, pp.722-753, 2017.

. Ii-p, Ivosidenib or enasidenib in combination with induction and consolidation therapy in newly diagnosed acute myeloid leukemia with IDH or IDH2 mutation -oral abstract presented at ASH, 2018.

. Ii-p, Enasidenib in previously untreated IDH2-mutated acute myeloid leukemia -results from the Beat AML Master Trial -oral abstract from ASH, 2018.

D. Sur, idh2-mutated-acute-myeloid-leukemia-results-from-the-beat-aml-master-trial?_&___prerender 184. Combination of Enasidenib and Venetoclax Shows Superior Anti-Leukemic Activity Against IDH2 Mutated AML in Patient-Derived Xenograft Models | Blood Journal

A. M. Disponible-sur-;-intlekofer, A. H. Shih, B. Wang, A. Nazir, A. S. Rustenburg et al., Acquired resistance to IDH inhibition through trans or cis dimer-interface mutations, Nature, vol.559, issue.7712, pp.125-134, 2018.

D. Hanahan and R. A. Weinberg, The hallmarks of cancer, Cell. 7 janv, vol.100, issue.1, pp.57-70, 2000.

A. Mcbride, S. Houtmann, L. Wilde, C. Vigil, C. M. Eischen et al., The Role of Inhibition of Apoptosis in Acute Leukemias and Myelodysplastic Syndrome, Front Oncol, vol.9, p.192, 2019.

U. Testa and R. Riccioni, Deregulation of apoptosis in acute myeloid leukemia, Haematologica. janv, vol.92, issue.1, pp.81-94, 2007.

S. Ogilvy, D. Metcalf, C. G. Print, M. L. Bath, A. W. Harris et al., Constitutive Bcl-2 expression throughout the hematopoietic compartment affects multiple lineages and enhances progenitor cell survival, Proc Natl Acad Sci, vol.96, issue.26, pp.14943-14951, 1999.

C. D. Dinardo, K. Pratz, V. Pullarkat, B. A. Jonas, M. Arellano et al., Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia, Blood. 3 janv, vol.133, issue.1, pp.7-17, 2019.

, Phase 1/2 Study of Venetoclax with Low-Dose Cytarabine in Treatment-Naive, Elderly Patients with Acute Myeloid Leukemia Unfit for Intensive Chemotherapy: 1-Year Outcomes | Blood Journal

. Ii-p, Venetoclax in combination with chemotherapy in elderly patients with acute myeloid leukemia -CAVEAT study -Oral abstract from ASH, 2018.

D. Sur,

I. Aldoss, D. Yang, A. Aribi, H. Ali, K. Sandhu et al., Efficacy of the combination of venetoclax and hypomethylating agents in relapsed/refractory acute myeloid leukemia, Haematologica. sept, vol.103, issue.9, pp.404-411, 2018.

C. D. Dinardo, C. R. Rausch, C. Benton, T. Kadia, N. Jain et al., Clinical experience with the BCL2-inhibitor venetoclax in combination therapy for relapsed and refractory acute myeloid leukemia and related myeloid malignancies, Am J Hematol. mars, vol.93, issue.3, pp.401-408, 2018.

M. Konopleva and A. Letai, BCL-2 inhibition in AML: an unexpected bonus? Blood. 6 sept, vol.132, pp.1007-1019, 2018.

M. Konopleva, D. A. Pollyea, J. Potluri, B. Chyla, L. Hogdal et al., Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia, Cancer Discov, vol.6, issue.10, pp.1106-1123, 2016.

T. Teh, N. Nguyen, D. M. Moujalled, D. Segal, G. Pomilio et al., Enhancing venetoclax activity in acute myeloid leukemia by co-targeting MCL1, Leukemia. févr, vol.32, issue.2, pp.303-315, 2018.

D. M. Moujalled, G. Pomilio, C. Ghiurau, A. Ivey, J. Salmon et al., Combining BH3-mimetics to target both BCL-2 and MCL1 has potent activity in pre-clinical models of acute myeloid leukemia, Leukemia. avr, vol.33, issue.4, pp.905-922, 2019.

K. Kojima, J. Ishizawa, and M. Andreeff, Pharmacological activation of wild-type p53 in the therapy of leukemia, Exp Hematol. sept, vol.44, issue.9, pp.791-799, 2016.

M. G. Inc, PHASE 1B/2 COMBINATION STUDY OF APR-246 AND AZACITIDINE (AZA) IN... by David Sallman

D. Sur,

P. A. Cassier, M. Castets, A. Belhabri, and N. Vey, Targeting apoptosis in acute myeloid leukaemia, Br J Cancer, vol.117, issue.8, pp.1089-98, 2017.

Y. Sun, B. Chen, and A. Deshpande, Epigenetic Regulators in the Development, Maintenance, and Therapeutic Targeting of, Acute Myeloid Leukemia. Front Oncol, vol.8, p.41, 2018.

M. Stahl, S. D. Gore, N. Vey, and T. Prebet, Lost in translation? Ten years of development of histone deacetylase inhibitors in acute myeloid leukemia and myelodysplastic syndromes, Expert Opin Investig Drugs, vol.25, issue.3, pp.307-324, 2016.

J. P. Bewersdorf, R. Shallis, M. Stahl, and A. M. Zeidan, Epigenetic therapy combinations in acute myeloid leukemia: what are the options?, Ther Adv Hematol, vol.10, p.2040620718816698, 2019.

M. Cerrano and R. Itzykson, New Treatment Options for Acute Myeloid Leukemia in 2019, Curr Oncol Rep. 4 févr, vol.21, issue.2, p.16, 2019.

K. Agrawal, V. Das, P. Vyas, and M. Hajdúch, Nucleosidic DNA demethylating epigenetic drugs -A comprehensive review from discovery to clinic, Pharmacol Ther, vol.188, pp.45-79, 2018.

P. Fenaux, G. J. Mufti, E. Hellström-lindberg, V. Santini, N. Gattermann et al., Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia, J Clin Oncol Off J Am Soc Clin Oncol. 1 févr, vol.28, issue.4, pp.562-571, 2010.

P. Fenaux, G. J. Mufti, E. Hellstrom-lindberg, V. Santini, C. Finelli et al., Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study, Lancet Oncol. mars, vol.10, issue.3, pp.223-255, 2009.

H. Dombret, J. F. Seymour, A. Butrym, A. Wierzbowska, D. Selleslag et al., International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts, Blood. 16 juill, vol.126, issue.3, pp.291-300, 2015.

L. Pleyer, S. Burgstaller, M. Girschikofsky, W. Linkesch, R. Stauder et al.,

, Azacitidine in 302 patients with WHO-defined acute myeloid leukemia: results from the Austrian Azacitidine Registry of the AGMT-Study Group, Ann Hematol, vol.93, issue.11, pp.1825-1863, 2014.

S. Thépot, R. Itzykson, V. Seegers, C. Recher, E. Raffoux et al., Azacitidine in untreated acute myeloid leukemia: a report on 149 patients, Am J Hematol. avr, vol.89, issue.4, pp.410-416, 2014.

R. Itzykson, S. Thépot, C. Berthon, J. Delaunay, D. Bouscary et al., Azacitidine for the treatment of relapsed and refractory AML in older patients, Leuk Res. févr, vol.39, issue.2, pp.124-154, 2015.

, Azacytidine treatment for relapsed or refractory acute myeloid leukemia after intensive chemotherapy

L. H. Van-der-helm, E. Scheepers, N. Veeger, S. Daenen, A. B. Mulder et al., Azacitidine might be beneficial in a subgroup of older AML patients compared to intensive chemotherapy: a single centre retrospective study of 227 consecutive patients, J Hematol OncolJ Hematol Oncol. 16 avr, vol.6, p.29, 2013.

Z. Lao, R. Yiu, G. C. Wong, and A. Ho, Treatment of elderly patients with acute myeloid leukemia with azacitidine results in fewer hospitalization days and infective complications but similar survival compared with intensive chemotherapy, Asia Pac J Clin Oncol. mars, vol.11, issue.1, pp.54-61, 2015.

F. Traina, V. Visconte, P. Elson, A. Tabarroki, A. M. Jankowska et al., Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms, Leukemia. janv, vol.28, issue.1, pp.78-87, 2014.

G. Garcia-manero, S. D. Gore, S. Kambhampati, B. Scott, A. Tefferi et al., Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes, Leukemia. avr, vol.30, issue.4, pp.889-96, 2016.

H. M. Kantarjian, X. G. Thomas, A. Dmoszynska, A. Wierzbowska, G. Mazur et al.,

. Multicenter, 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, J Clin Oncol Off J Am Soc Clin Oncol. 20 juill, vol.30, issue.21, pp.2670-2677, 2012.

G. Huls, S. Suciu, P. Wijermans, M. Kicinski, and M. Lübbert, 10-day vs 5-day decitabine: equivalence cannot be concluded, Lancet Haematol, vol.6, issue.4, p.177, 2019.

H. M. Kantarjian, G. J. Roboz, P. L. Kropf, K. Yee, C. L. O'connell et al.,

, in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial, Lancet Oncol, vol.18, issue.10, pp.1317-1343, 2017.

, Phase III ASTRAL-1 Study of Guadecitabine in Patients With Treatment-Naive AML Ineligible to Receive Intensive Induction Chemotherapy -The ASCO Post

D. Sur,

P. A. Marks, Histone deacetylase inhibitors: a chemical genetics approach to understanding cellular functions, Biochim Biophys Acta. déc, vol.1799, pp.717-742, 2010.

J. C. Byrd, G. Marcucci, M. R. Parthun, J. J. Xiao, R. B. Klisovic et al., A phase 1 and pharmacodynamic study of depsipeptide (FK228) in chronic lymphocytic leukemia and acute myeloid leukemia, Blood. 1 févr, vol.105, issue.3, pp.959-67, 2005.

V. M. Klimek, S. Fircanis, P. Maslak, I. Guernah, M. Baum et al., Tolerability, pharmacodynamics, and pharmacokinetics studies of depsipeptide (romidepsin) in patients with acute myelogenous leukemia or advanced myelodysplastic syndromes, Clin Cancer Res Off J Am Assoc Cancer Res. 1 févr, vol.14, issue.3, pp.826-858, 2008.

I. Gojo, M. Tan, H. Fang, M. Sadowska, R. Lapidus et al., Translational phase I trial of vorinostat (suberoylanilide hydroxamic acid) combined with cytarabine and etoposide in patients with relapsed, refractory, or high-risk acute myeloid leukemia, Clin Cancer Res Off J Am Assoc Cancer Res. 1 avr, vol.19, issue.7, pp.1838-51, 2013.

D. J. Deangelo, A. Spencer, K. N. Bhalla, H. M. Prince, T. Fischer et al., Phase Ia/II, twoarm, open-label, dose-escalation study of oral panobinostat administered via two dosing schedules in patients with advanced hematologic malignancies, Leukemia. août, vol.27, issue.8, pp.1628-1664, 2013.

J. Zuber, J. Shi, E. Wang, A. R. Rappaport, H. Herrmann et al., RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia, Nature. 3 août, vol.478, issue.7370, pp.524-532, 2011.

A. Alqahtani, K. Choucair, M. Ashraf, D. M. Hammouda, A. Alloghbi et al., Bromodomain and extra-terminal motif inhibitors: a review of preclinical and clinical advances in cancer therapy, Future Sci OA. mars, vol.5, issue.3, p.372, 2019.

T. Braun and C. Gardin, Investigational BET bromodomain protein inhibitors in early stage clinical trials for acute myelogenous leukemia (AML), Expert Opin Investig Drugs. 3 juill, vol.26, issue.7, pp.803-814, 2017.

C. Berthon, E. Raffoux, X. Thomas, N. Vey, C. Gomez-roca et al., Bromodomain inhibitor OTX015 in patients with acute leukaemia: a dose-escalation, phase 1 study, Lancet Haematol. avr, vol.3, issue.4, pp.186-195, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01791279

, a pan-inhibitor of bromodomain and extra terminal (BET) proteins, in patients (pts) with relapsed/refractory (RR) acute myeloid leukemia (AML): Preliminary data, -human study of ABBV-075 (mivebresib)

D. Sur,

L. M. Drusbosky, R. Vidva, S. Gera, A. V. Lakshminarayana, V. P. Shyamasundar et al., Predicting response to BET inhibitors using computational modeling: A BEAT AML project study, Leuk Res. févr, vol.77, pp.42-50, 2019.

Y. Okada, Q. Feng, Y. Lin, Q. Jiang, Y. Li et al., hDOT1L links histone methylation to leukemogenesis, Cell. 22 avr, vol.121, issue.2, pp.167-78, 2005.

M. Kühn, M. J. Hadler, S. R. Daigle, R. P. Koche, A. V. Krivtsov et al., MLL partial tandem duplication leukemia cells are sensitive to small molecule DOT1L inhibition, Haematologica. mai, vol.100, issue.5, pp.190-193, 2015.

E. M. Stein, G. Garcia-manero, D. A. Rizzieri, R. Tibes, J. G. Berdeja et al., The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia, Blood. 14 juin, vol.131, issue.24, pp.2661-2670, 2018.

R. E. Rau, B. A. Rodriguez, M. Luo, M. Jeong, A. Rosen et al., DOT1L as a therapeutic target for the treatment of DNMT3A-mutant acute myeloid leukemia, Blood, vol.18, issue.7, pp.971-81, 2016.

M. Kühn, E. Song, Z. Feng, A. Sinha, C. Chen et al., Targeting Chromatin Regulators Inhibits Leukemogenic Gene Expression in NPM1 Mutant Leukemia, Cancer Discov, vol.6, issue.10, pp.1166-81, 2016.

C. R. Klaus, D. Iwanowicz, D. Johnston, C. A. Campbell, J. J. Smith et al., DOT1L inhibitor EPZ-5676 displays synergistic antiproliferative activity in combination with standard of care drugs and hypomethylating agents in MLL-rearranged leukemia cells, J Pharmacol Exp Ther. sept, vol.350, issue.3, pp.646-56, 2014.

J. Wang, K. Saijo, D. Skola, J. C. Ma, Q. Merkurjev et al., Histone demethylase LSD1 regulates hematopoietic stem cells homeostasis and protects from death by endotoxic shock, Proc Natl Acad Sci, vol.9, issue.2, pp.244-52, 2018.

W. Fiskus, S. Sharma, B. Shah, B. P. Portier, S. Devaraj et al., Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells, Leukemia. nov, vol.28, issue.11, pp.2155-64, 2014.

G. Castelli, E. Pelosi, and U. Testa, Targeting histone methyltransferase and demethylase in acute myeloid leukemia therapy, OncoTargets Ther. 28 déc, vol.11, pp.131-55, 2017.

T. Prebet, Z. Sun, M. E. Figueroa, R. Ketterling, A. Melnick et al., Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: results of the US Leukemia Intergroup trial E1905, J Clin Oncol Off J Am Soc Clin Oncol. 20 avr, vol.32, issue.12, pp.1242-1250, 2014.

M. A. Sekeres, M. Othus, A. F. List, O. Odenike, R. M. Stone et al., Randomized Phase II Study of Azacitidine Alone or in Combination With Lenalidomide or With Vorinostat in Higher-Risk Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia: North American Intergroup Study SWOG S1117, J Clin Oncol Off J Am Soc Clin Oncol. 20 août, vol.35, issue.24, pp.2745-53, 2017.

G. Garcia-manero, Y. Abaza, K. Takahashi, B. C. Medeiros, M. Arellano et al., Pracinostat plus azacitidine in older patients with newly diagnosed acute myeloid leukemia: results of a phase 2 study, Blood Adv. 26 févr, vol.3, issue.4, pp.508-526, 2019.

G. Garcia-manero, G. Montalban-bravo, J. G. Berdeja, Y. Abaza, E. Jabbour et al., Phase 2, randomized, double-blind study of pracinostat in combination with azacitidine in patients with untreated, higher-risk myelodysplastic syndromes, Cancer. 15 mai, vol.123, issue.6, pp.994-1002, 2017.

J. Issa, G. Garcia-manero, X. Huang, J. Cortes, F. Ravandi et al., Results of phase 2 randomized study of low-dose decitabine with or without valproic acid in patients with myelodysplastic syndrome and acute myelogenous leukemia, Cancer. 15 févr, vol.121, issue.4, pp.556-61, 2015.

R. L. Momparler, S. Côté, L. F. Momparler, and Y. Idaghdour, Inhibition of DNA and Histone Methylation by 5-Aza-2'-Deoxycytidine (Decitabine) and 3-Deazaneplanocin-A on Antineoplastic Action and Gene Expression in, Myeloid Leukemic Cells. Front Oncol, vol.7, p.19, 2017.

W. Fiskus, Y. Wang, A. Sreekumar, K. M. Buckley, H. Shi et al., Combined epigenetic therapy with the histone methyltransferase EZH2 inhibitor 3-deazaneplanocin A and the histone deacetylase inhibitor panobinostat against human AML cells, Blood. 24 sept, vol.114, issue.13, pp.2733-2776, 2009.

V. I. Avramis, R. A. Mecum, J. Nyce, D. A. Steele, and J. S. Holcenberg, Pharmacodynamic and DNA methylation studies of high-dose 1-beta-D-arabinofuranosyl cytosine before and after in vivo 5-azacytidine treatment in pediatric patients with refractory acute lymphocytic leukemia, Cancer Chemother Pharmacol, vol.24, issue.4, pp.203-213, 1989.

X. Ye, X. Zhou, J. Wei, G. Xu, Y. Li et al., Epigenetic priming with decitabine followed by low-dose idarubicin/cytarabine has an increased anti-leukemic effect compared to traditional chemotherapy in high-risk myeloid neoplasms, Leuk Lymphoma, vol.57, issue.6, pp.1311-1319, 2016.

J. M. Scandura, G. J. Roboz, M. Moh, E. Morawa, F. Brenet et al., Phase 1 study of epigenetic priming with decitabine prior to standard induction chemotherapy for patients with AML, Blood. 11 août, vol.118, issue.6, pp.1472-80, 2011.

C. Müller-tidow, P. Tschanter, C. Röllig, C. Thiede, A. Koschmieder et al., Azacitidine in combination with intensive induction chemotherapy in older patients with acute myeloid leukemia: The AML-AZA trial of the Study Alliance Leukemia, Leukemia. mars, vol.30, issue.3, pp.555-61, 2016.

J. Zhang, Y. Cao, and J. Li, Efficacy and Safety of Decitabine Combined with CAG (Cytarabine, Aclarubicin, G-CSF) for Patients with Intermediate or High Risk Myelodysplastic Syndrome and Acute Myeloid Leukemia

, Zhongguo Shi Yan Xue Ye Xue Za Zhi. avr, vol.27, issue.2, pp.494-503, 2019.

R. Rosato, S. Hock, P. Dent, Y. Dai, and S. Grant, LBH-589 (panobinostat) potentiates fludarabine antileukemic activity through a JNK-and XIAP-dependent mechanism, Leuk Res. avr, vol.36, issue.4, pp.491-499, 2012.

G. Garcia-manero, F. P. Tambaro, N. B. Bekele, H. Yang, F. Ravandi et al., Phase II trial of vorinostat with idarubicin and cytarabine for patients with newly diagnosed acute myelogenous leukemia or myelodysplastic syndrome, J Clin Oncol Off J Am Soc Clin Oncol. 20 juin, vol.30, issue.18, pp.2204-2214, 2012.

, SWOG S1203: A Randomized Phase III Study of Standard Cytarabine Plus Daunorubicin (7+3) Therapy Versus Idarubicin with High Dose Cytarabine (IA) with or without Vorinostat

, Younger Patients with Previously Untreated Acute Myeloid Leukemia (AML) | Blood Journal

D. Sur,

E. M. Ocio, P. Herrera, M. Olave, N. Castro, J. A. Pérez-simón et al., Panobinostat as part of induction and maintenance for elderly patients with newly diagnosed acute myeloid leukemia: phase Ib/II panobidara study, Haematologica. oct, vol.100, issue.10, pp.1294-300, 2015.

M. Almstedt, N. Blagitko-dorfs, J. Duque-afonso, J. Karbach, D. Pfeifer et al., The DNA demethylating agent 5-aza-2'-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells, Leuk Res. juill, vol.34, issue.7, pp.899-905, 2010.

O. B. Gbolahan, A. M. Zeidan, M. Stahl, A. Zaid, M. Farag et al.,

, Immunotherapeutic Concepts to Target Acute Myeloid Leukemia: Focusing on the Role of Monoclonal Antibodies, Hypomethylating Agents and the Leukemic Microenvironment, Int J Mol Sci. 31 juill, vol.18, issue.8, 2017.

N. Daver, P. Boddu, G. Garcia-manero, S. S. Yadav, P. Sharma et al., Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes, Leukemia, vol.32, issue.5, pp.1094-105, 2018.

A. D. Ørskov, M. B. Treppendahl, A. Skovbo, M. S. Holm, L. S. Friis et al., Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation, Oncotarget. 20 avr, vol.6, issue.11, pp.9612-9638, 2015.

K. B. Chiappinelli, P. L. Strissel, A. Desrichard, H. Li, C. Henke et al., Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses, Cell. 27 août, vol.162, issue.5, pp.974-86, 2015.

, BMS-936558) in Combination With 5-Azacytidine (Vidaza) or Nivolumab With Ipilimumab in Combination With 5-Azacytidine for the Treatment of Patients With Refractory/ Relapsed Acute Myeloid Leukemia and Newly Diagnosed Older AML, An Open-Label Phase II Study of Nivolumab

, Years) Patients -AdisInsight

T. Terao and Y. Minami, Targeting Hedgehog (Hh) Pathway for the Acute Myeloid Leukemia Treatment, Cells. 3 avr, vol.8, issue.4, 2019.

D. Bixby, R. Noppeney, T. L. Lin, J. Cortes, J. Krauter et al., Safety and efficacy of vismodegib in relapsed/refractory acute myeloid leukaemia: results of a phase Ib trial, Br J Haematol. mai, vol.185, issue.3, pp.595-603, 2019.

J. E. Cortes, F. H. Heidel, A. Hellmann, W. Fiedler, B. D. Smith et al., Randomized comparison of low dose cytarabine with or without glasdegib in patients with newly diagnosed acute myeloid leukemia or high-risk myelodysplastic syndrome, Leukemia. févr, vol.33, issue.2, pp.379-89, 2019.

J. E. Cortes, D. Smith, B. Wang, E. S. Merchant, A. Oehler et al., Glasdegib in combination with cytarabine and daunorubicin in patients with AML or high-risk MDS: Phase 2 study results, Am J Hematol, vol.93, issue.11, pp.1301-1311, 2018.

, Paper: Phase I/IB Study of Azacitidine and Hedgehog Pathway Inhibition in Myeloid Malignancies

E. Latuske, H. Stamm, M. Klokow, G. Vohwinkel, J. Muschhammer et al., Combined inhibition of GLI and FLT3 signaling leads to effective anti-leukemic effects in human acute myeloid leukemia, Oncotarget. 25 avr, vol.8, issue.17, pp.29187-201, 2017.

M. H. Kirschbaum, T. Synold, A. S. Stein, J. Tuscano, J. M. Zain et al., A phase 1 trial dose-escalation study of tipifarnib on a week-on, week-off schedule in relapsed, refractory or highrisk myeloid leukemia, Leukemia, vol.25, issue.10, pp.1543-1550, 2011.

J. E. Lancet, I. Gojo, J. Gotlib, E. J. Feldman, J. Greer et al., A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia, Blood. 15 févr, vol.109, issue.4, pp.1387-94, 2007.

J. Harousseau, J. E. Lancet, J. Reiffers, B. Lowenberg, X. Thomas et al., A phase 2 study of the oral farnesyltransferase inhibitor tipifarnib in patients with refractory or relapsed acute myeloid leukemia, Blood. 15 juin, vol.109, issue.12, pp.5151-5157, 2007.

H. P. Erba, M. Othus, R. B. Walter, M. H. Kirschbaum, M. S. Tallman et al., Four different regimens of farnesyltransferase inhibitor tipifarnib in older, untreated acute myeloid leukemia patients: North American Intergroup Phase II study SWOG S0432, Leuk Res. mars, vol.38, issue.3, pp.329-362, 2014.

J. Downward, Targeting RAS signalling pathways in cancer therapy, Nat Rev Cancer. janv, vol.3, issue.1, pp.11-22, 2003.

M. Eskandarpour, S. Kiaii, C. Zhu, J. Castro, A. J. Sakko et al., Suppression of oncogenic NRAS by RNA interference induces apoptosis of human melanoma cells, Int J Cancer. 20 mai, vol.115, issue.1, pp.65-73, 2005.

X. Huang, S. Schwind, R. Santhanam, A. Eisfeld, C. Chiang et al., Targeting the RAS/MAPK pathway with miR-181a in acute myeloid leukemia, Oncotarget. 13 sept, vol.7, issue.37, pp.59273-86, 2016.

J. O. Lauchle, D. Kim, D. T. Le, K. Akagi, M. Crone et al., Response and resistance to MEK inhibition in leukaemias initiated by hyperactive Ras, Nature. 17 sept, vol.461, issue.7262, pp.411-415, 2009.

N. Jain, E. Curran, N. M. Iyengar, E. Diaz-flores, R. Kunnavakkam et al., Phase II study of the oral MEK inhibitor selumetinib in advanced acute myelogenous leukemia: a University of Chicago phase II consortium trial, Clin Cancer Res Off J Am Assoc Cancer Res. 15 janv, vol.20, issue.2, pp.490-498, 2014.

G. Borthakur, L. Popplewell, M. Boyiadzis, J. Foran, U. Platzbecker et al., Activity of the oral mitogen-activated protein kinase kinase inhibitor trametinib in RAS-mutant relapsed or refractory myeloid malignancies, Cancer, vol.15, issue.12, pp.1871-1880, 2016.

A. Maiti, K. Naqvi, T. M. Kadia, G. Borthakur, K. Takahashi et al., Phase II Trial of MEK Inhibitor Binimetinib (MEK162) in RAS-mutant Acute Myeloid Leukemia, Clin Lymphoma Myeloma Leuk. mars, vol.19, issue.3, pp.142-148, 2019.

Q. Xu, S. Simpson, T. J. Scialla, A. Bagg, and M. Carroll, Survival of acute myeloid leukemia cells requires PI3 kinase activation, Blood. 1 août, vol.102, issue.3, pp.972-80, 2003.

J. Bertacchini, N. Heidari, L. Mediani, S. Capitani, M. Shahjahani et al., Targeting PI3K/AKT/mTOR network for treatment of leukemia, Cell Mol Life Sci CMLS. juin, vol.72, issue.12, pp.2337-2384, 2015.

B. K. Ragon, H. Kantarjian, E. Jabbour, F. Ravandi, J. Cortes et al., Buparlisib, a PI3K inhibitor, demonstrates acceptable tolerability and preliminary activity in a phase I trial of patients with advanced leukemias, Am J Hematol. janv, vol.92, issue.1, pp.7-11, 2017.

A. E. Perl, M. T. Kasner, D. E. Tsai, D. T. Vogl, A. W. Loren et al., A phase I study of the mammalian target of rapamycin inhibitor sirolimus and MEC chemotherapy in relapsed and refractory acute myelogenous leukemia, Clin Cancer Res Off J Am Assoc Cancer Res, vol.15, issue.21, pp.6732-6741, 2009.

S. Amadori, R. Stasi, A. M. Martelli, A. Venditti, G. Meloni et al., Temsirolimus, an mTOR inhibitor, in combination with lower-dose clofarabine as salvage therapy for older patients with acute myeloid leukaemia: results of a phase II GIMEMA study (AML-1107), Br J Haematol. janv, vol.156, issue.2, pp.205-217, 2012.

A. Rashidi and J. F. Dipersio, Targeting the leukemia-stroma interaction in acute myeloid leukemia: rationale and latest evidence, Ther Adv Hematol. févr, vol.7, issue.1, pp.40-51, 2016.

G. L. Uy, M. P. Rettig, I. H. Motabi, K. Mcfarland, K. M. Trinkaus et al., A phase 1/2 study of chemosensitization with the CXCR4 antagonist plerixafor in relapsed or refractory acute myeloid leukemia, Blood. 26 avr, vol.119, issue.17, pp.3917-3941, 2012.

G. J. Roboz, E. K. Ritchie, Y. Dault, L. Lam, D. C. Marshall et al., Phase I trial of plerixafor combined with decitabine in newly diagnosed older patients with acute myeloid leukemia, Haematologica, vol.103, issue.8, pp.1308-1324, 2018.

A. M. Gruszka, D. Valli, C. Restelli, and M. Alcalay, Adhesion Deregulation in Acute Myeloid Leukaemia, Cells. 17 janv, vol.8, issue.1, 2019.

B. Cho, H. Kim, and M. Konopleva, Targeting the CXCL12/CXCR4 axis in acute myeloid leukemia: from bench to bedside, Korean J Intern Med. mars, vol.32, issue.2, pp.248-57, 2017.

I. F. Tannock and J. A. Hickman, Limits to Personalized Cancer Medicine, N Engl J Med. 29 sept, vol.375, issue.13, pp.1289-94, 2016.

F. Perez, N. G. El-chakhtoura, K. M. Papp-wallace, B. M. Wilson, and R. A. Bonomo, Treatment options for infections caused by carbapenem-resistant Enterobacteriaceae : can we apply "precision medicine" to antimicrobial chemotherapy?, Expert Opin Pharmacother. 12 avr, vol.17, issue.6, pp.761-81, 2016.

A. Gustavsson and T. Olofsson, Prediction of response to chemotherapy in acute leukemia by in vitro drug sensitivity testing on leukemic stem cells, Cancer Res, vol.44, issue.10, pp.4648-52, 1984.

J. M. Sargent and C. G. Taylor, Appraisal of the MTT assay as a rapid test of chemosensitivity in acute myeloid leukaemia, Br J Cancer. août, vol.60, issue.2, pp.206-216, 1989.

E. Klumper, G. J. Ossenkoppele, R. Pieters, D. R. Huismans, A. H. Loonen et al., In vitro resistance to cytosine arabinoside, not to daunorubicin, is associated with the risk of relapse in de novo acute myeloid leukaemia, Br J Haematol. juin, vol.93, issue.4, pp.903-913, 1996.

B. Snijder, G. I. Vladimer, N. Krall, K. Miura, A. Schmolke et al., Image-based ex-vivo drug screening for patients with aggressive haematological malignancies: interim results from a single-arm, open-label, pilot study, Lancet Haematol. déc, vol.4, issue.12, pp.595-606, 2017.

R. T. Swords, D. Azzam, A. , H. Lohse, I. Volmar et al., Ex-vivo sensitivity profiling to guide clinical decision making in acute myeloid leukemia: A pilot study, Leuk Res. janv, vol.64, pp.34-41, 2018.

V. Lavallée, J. Krosl, S. Lemieux, G. Boucher, P. Gendron et al., Chemo-genomic interrogation of CEBPA mutated AML reveals recurrent CSF3R mutations and subgroup sensitivity to JAK inhibitors, Blood, vol.16, issue.24, pp.3054-61, 2016.

T. Pemovska, M. Kontro, B. Yadav, H. Edgren, S. Eldfors et al., Individualized Systems Medicine Strategy to Tailor Treatments for Patients with Chemorefractory Acute Myeloid Leukemia, Cancer Discov. 1 déc, vol.3, issue.12, pp.1416-1445, 2013.

J. W. Tyner, C. E. Tognon, D. Bottomly, B. Wilmot, S. E. Kurtz et al., Functional genomic landscape of acute myeloid leukaemia, Nature. oct, vol.562, issue.7728, pp.526-557, 2018.

Y. Collette, T. Prébet, A. Goubard, J. Adélaïde, R. Castellano et al., Drug response profiling can predict response to ponatinib in a patient with t(1;9)(q24;q34)-associated B-cell acute lymphoblastic leukemia, Blood Cancer J. 13 mars, vol.5, issue.3, p.292, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01218497

N. Cervera, N. Carbuccia, S. Garnier, A. Guille, J. Adélaïde et al., Molecular characterization of acute erythroid leukemia (M6-AML) using targeted next-generation sequencing, Leukemia. avr, vol.30, issue.4, pp.966-70, 2016.

H. Li and R. Durbin, Fast and accurate long-read alignment with Burrows-Wheeler transform, Bioinforma Oxf Engl. 1 mars, vol.26, issue.5, pp.589-95, 2010.

M. A. Depristo, E. Banks, R. Poplin, K. V. Garimella, J. R. Maguire et al., A framework for variation discovery and genotyping using next-generation DNA sequencing data, Nat Genet. mai, vol.43, issue.5, pp.491-499, 2011.

D. C. Koboldt, Q. Zhang, D. E. Larson, D. Shen, M. D. Mclellan et al., VarScan 2: Somatic mutation and copy number alteration discovery in cancer by exome sequencing, Genome Res. 1 mars, vol.22, issue.3, pp.568-76, 2012.

L. T. Fang, P. T. Afshar, A. Chhibber, M. Mohiyuddin, Y. Fan et al., An ensemble approach to accurately detect somatic mutations using SomaticSeq, Genome Biol. 17 sept, vol.16, p.197, 2015.

K. Wang, M. Li, and H. Hakonarson, ANNOVAR: functional annotation of genetic variants from highthroughput sequencing data, Nucleic Acids Res. sept, vol.38, issue.16, p.164, 2010.

J. T. Robinson, H. Thorvaldsdóttir, W. Winckler, M. Guttman, E. S. Lander et al., Integrative genomics viewer, Nat Biotechnol. janv, vol.29, issue.1, pp.24-30, 2011.

H. Thorvaldsdóttir, J. T. Robinson, and J. P. Mesirov, Integrative Genomics Viewer (IGV): highperformance genomics data visualization and exploration, Brief Bioinform. mars, vol.14, issue.2, pp.178-92, 2013.

J. Perera-bel, B. Hutter, C. Heining, A. Bleckmann, M. Fröhlich et al., From somatic variants towards precision oncology: Evidence-driven reporting of treatment options in molecular tumor boards, Genome Med, vol.15, issue.1, p.18, 2018.

H. E. Haarberg, K. Paraiso, E. Wood, V. W. Rebecca, V. K. Sondak et al., Inhibition of Wee1, AKT, and CDK4 underlies the efficacy of the HSP90 inhibitor XL888 in an in vivo model of NRAS-mutant melanoma, Mol Cancer Ther. juin, vol.12, issue.6, pp.901-913, 2013.

E. Stein, J. Chromik, D. J. Deangelo, M. Chatterjee, R. Noppeney et al., Abstract CT152: Phase I dose-and regimen-finding study of NVP-HDM201 in pts with advanced TP53 wt acute leukemias, Cancer Res. 1 juill, vol.77, pp.152-152, 2013.

A. J. Patel, C. Liao, Z. Chen, C. Liu, Y. Wang et al., BET bromodomain inhibition triggers apoptosis of NF1-associated malignant peripheral nerve sheath tumors through Bim induction, Cell Rep. 16 janv, vol.6, issue.1, pp.81-92, 2014.

C. Rubio-perez, D. Tamborero, M. P. Schroeder, A. A. Antolín, J. Deu-pons et al., Silico Prescription of Anticancer Drugs to Cohorts of 28 Tumor Types Reveals Targeting Opportunities, Cancer Cell. mars, vol.27, issue.3, pp.382-96, 2015.

R. Dienstmann, I. S. Jang, B. Bot, S. Friend, and J. Guinney, Database of genomic biomarkers for cancer drugs and clinical targetability in solid tumors, Cancer Discov. févr, vol.5, issue.2, pp.118-141, 2015.

C. Eifert and R. S. Powers, From cancer genomes to oncogenic drivers, tumour dependencies and therapeutic targets, Nat Rev Cancer, vol.12, issue.8, pp.572-580, 2012.

F. Andre, E. Mardis, M. Salm, J. Soria, L. L. Siu et al., Prioritizing targets for precision cancer medicine, Ann Oncol. 1 déc, vol.25, issue.12, pp.2295-303, 2014.

J. Mateo, D. Chakravarty, R. Dienstmann, S. Jezdic, A. Gonzalez-perez et al., A framework to rank genomic alterations as targets for cancer precision medicine: the ESMO Scale for Clinical Actionability of molecular Targets (ESCAT), Ann Oncol. 1 sept, vol.29, issue.9, pp.1895-902, 2018.

R. Bejar, A. Lord, K. Stevenson, M. Bar-natan, A. Pérez-ladaga et al., TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients, Blood, vol.124, issue.17, pp.2705-2717, 2014.

S. E. Kurtz, C. A. Eide, A. Kaempf, V. Khanna, S. L. Savage et al., Molecularly targeted drug combinations demonstrate selective effectiveness for myeloid-and lymphoid-derived hematologic malignancies, Proc Natl Acad Sci, vol.114, issue.36, pp.7554-63, 2017.

M. Cruijsen, M. Lübbert, P. Wijermans, and G. Huls, Clinical Results of Hypomethylating Agents in AML Treatment, J Clin Med. 25 déc, vol.4, issue.1, pp.1-17, 2014.

C. Karantanou, P. S. Godavarthy, and D. S. Krause, Targeting the bone marrow microenvironment in acute leukemia, Leuk Lymphoma. 12 févr, pp.1-11, 2018.

B. Yadav, T. Pemovska, A. Szwajda, E. Kulesskiy, M. Kontro et al., Quantitative scoring of differential drug sensitivity for individually optimized anticancer therapies, 2015.

, Disponible sur

C. Brideau, B. Gunter, B. Pikounis, and A. Liaw, Improved statistical methods for hit selection in highthroughput screening, J Biomol Screen. déc, vol.8, issue.6, pp.634-681, 2003.

N. A. , C. S. , and C. T. , Data Analysis Approaches in High Throughput Screening, El-Shemy H, éditeur. Drug Discovery, 2013.

N. Malo, J. A. Hanley, S. Cerquozzi, J. Pelletier, and R. Nadon, Statistical practice in high-throughput screening data analysis, Nat Biotechnol. févr, vol.24, issue.2, pp.167-75, 2006.

R. Itzykson, O. Kosmider, T. Cluzeau, V. Mansat-de-mas, F. Dreyfus et al., Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias, Leukemia. juill, vol.25, issue.7, pp.1147-52, 2011.

K. H. Metzeler, A. Walker, S. Geyer, R. Garzon, R. B. Klisovic et al., DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia, Leukemia. mai, vol.26, issue.5, pp.1106-1113, 2012.

H. Nahi, G. Selivanova, S. Lehmann, L. Möllgård, S. Bengtzen et al., Mutated and nonmutated TP53 as targets in the treatment of leukaemia, Br J Haematol. mai, vol.141, issue.4, pp.445-53, 2008.

J. S. Welch, A. A. Petti, C. A. Miller, C. C. Fronick, M. O'laughlin et al., TP53 and Decitabine in Acute Myeloid Leukemia and Myelodysplastic Syndromes, N Engl J Med, vol.24, issue.21, pp.2023-2059, 2016.

G. Lozanski, N. A. Heerema, I. W. Flinn, L. Smith, J. Harbison et al., Alemtuzumab is an effective therapy for chronic lymphocytic leukemia with p53 mutations and deletions, Blood. 1 mai, vol.103, issue.9, pp.3278-81, 2004.

E. M. Alexandrova, A. R. Yallowitz, D. Li, S. Xu, R. Schulz et al., Improving survival by exploiting tumour dependence on stabilized mutant p53 for treatment, Nature. 16 juill, vol.523, issue.7560, pp.352-358, 2015.

A. Venkatanarayan, P. Raulji, W. Norton, D. Chakravarti, C. Coarfa et al., IAPP-driven metabolic reprogramming induces regression of p53-deficient tumours in vivo, Nature. 29 janv, vol.517, issue.7536, pp.626-656, 2015.

S. Lee, H. Dvinge, E. Kim, H. Cho, J. Micol et al., Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins, Nat Med, vol.22, issue.6, pp.672-680, 2016.

P. A. Ascierto, D. Schadendorf, C. Berking, S. S. Agarwala, C. M. Van-herpen et al., MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a nonrandomised, open-label phase 2 study, Lancet Oncol. mars, vol.14, issue.3, pp.249-56, 2013.

A. A. Adjei, R. B. Cohen, W. Franklin, C. Morris, D. Wilson et al., Phase I pharmacokinetic and pharmacodynamic study of the oral, small-molecule mitogen-activated protein kinase kinase 1/2 inhibitor AZD6244 (ARRY-142886) in patients with advanced cancers, J Clin Oncol Off J Am Soc Clin Oncol. 1 mai, vol.26, issue.13, pp.2139-2185, 2008.

K. Ohashi, L. V. Sequist, M. E. Arcila, C. M. Lovly, X. Chen et al., Characteristics of lung cancers harboring NRAS mutations, Clin Cancer Res Off J Am Assoc Cancer Res. 1 mai, vol.19, issue.9, pp.2584-91, 2013.

T. Steinbrunn, T. Stühmer, C. Sayehli, M. Chatterjee, H. Einsele et al., Combined targeting of MEK/MAPK and PI3K/Akt signalling in multiple myeloma, Br J Haematol, vol.159, issue.4, pp.430-470, 2012.

M. Martinez-garcia, U. Banerji, J. Albanell, R. Bahleda, S. Dolly et al., First-inhuman, phase I dose-escalation study of the safety, pharmacokinetics, and pharmacodynamics of RO5126766, a first-in-class dual MEK/RAF inhibitor in patients with solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 1 sept, vol.18, issue.17, pp.4806-4825, 2012.

X. Wu, G. Xu, X. Li, W. Xu, Q. Li et al., Small Molecule Inhibitor that Stabilizes the Autoinhibited Conformation of the Oncogenic Tyrosine Phosphatase SHP2, J Med Chem. 14 févr, vol.62, issue.3, pp.1125-1162, 2019.

R. J. Chan and G. Feng, PTPN11 is the first identified proto-oncogene that encodes a tyrosine phosphatase, Blood. 1 févr, vol.109, issue.3, pp.862-869, 2007.

A. Prahallad, G. Heynen, G. Germano, S. M. Willems, B. Evers et al., PTPN11 Is a Central Node in Intrinsic and Acquired Resistance to Targeted Cancer Drugs, Cell Rep. 29 sept, vol.12, issue.12, pp.1978-85, 2015.

K. A. Robertson, G. Nalepa, F. Yang, D. C. Bowers, C. Y. Ho et al., Imatinib mesylate for plexiform neurofibromas in patients with neurofibromatosis type 1: a phase 2 trial, Lancet Oncol. déc, vol.13, issue.12, pp.1218-1242, 2012.

G. Johansson, Y. Y. Mahller, M. H. Collins, M. Kim, T. Nobukuni et al., Effective in vivo targeting of the mammalian target of rapamycin pathway in malignant peripheral nerve sheath tumors, Mol Cancer Ther. mai, vol.7, issue.5, pp.1237-1282, 2008.

F. Iorio, T. A. Knijnenburg, D. J. Vis, G. R. Bignell, M. P. Menden et al., Landscape of Pharmacogenomic Interactions in Cancer. Cell. 28 juill, vol.166, issue.3, pp.740-54, 2016.

R. Tremblay-lemay, N. Rastgoo, M. Pourabdollah, and H. Chang, EZH2 as a therapeutic target for multiple myeloma and other haematological malignancies, Biomark Res, vol.7, issue.2018

H. D. Nguyen, W. Y. Leong, W. Li, P. Reddy, J. D. Sullivan et al., Spliceosome Mutations Induce R Loop-Associated Sensitivity to ATR Inhibition in Myelodysplastic Syndromes, Cancer Res. 15 sept, vol.78, issue.18, pp.5363-74, 2018.

S. L. Maguire, A. Leonidou, P. Wai, C. Marchiò, C. K. Ng et al., SF3B1 mutations constitute a novel therapeutic target in breast cancer, J Pathol. mars, vol.235, issue.4, pp.571-80, 2015.

C. N. Harrison, A. M. Vannucchi, J. Kiladjian, A. Hk, H. Gisslinger et al., Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis, Leukemia, vol.30, issue.8, pp.1701-1708, 2016.

M. Levis, Midostaurin approved for FLT3-mutated AML, Blood, vol.29, issue.26, pp.3403-3409, 2017.

R. Ferrarotto, Y. Mitani, L. Diao, I. Guijarro, J. Wang et al., Activating NOTCH1 Mutations Define a Distinct Subgroup of Patients With Adenoid Cystic Carcinoma Who Have Poor Prognosis, Propensity to Bone and Liver Metastasis, and Potential Responsiveness to Notch1 Inhibitors, J Clin Oncol Off J Am Soc Clin Oncol. 20 janv, vol.35, issue.3, pp.352-60, 2017.

Q. Ye, J. Jiang, G. Zhan, W. Yan, L. Huang et al., Small molecule activation of NOTCH signaling inhibits acute myeloid leukemia, Sci Rep, vol.23, p.26510, 2016.

M. L. Bailey, N. J. O'neil, D. M. Van-pel, D. A. Solomon, T. Waldman et al., Glioblastoma cells containing mutations in the cohesin component STAG2 are sensitive to PARP inhibition, Mol Cancer Ther. mars, vol.13, issue.3, pp.724-756, 2014.

J. Waaler, O. Machon, L. Tumova, H. Dinh, V. Korinek et al., A novel tankyrase inhibitor decreases canonical Wnt signaling in colon carcinoma cells and reduces tumor growth in conditional APC mutant mice, Cancer Res. 1 juin, vol.72, issue.11, pp.2822-2854, 2012.

M. Javadi, T. D. Richmond, K. Huang, and D. L. Barber, CBL linker region and RING finger mutations lead to enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling via elevated levels of JAK2 and LYN, J Biol Chem. 5 juill, vol.288, issue.27, pp.19459-70, 2013.

C. Polprasert, I. Schulze, M. A. Sekeres, H. Makishima, B. Przychodzen et al., Inherited and Somatic Defects in DDX41 in Myeloid Neoplasms, Cancer Cell. 11 mai, vol.27, issue.5, pp.658-70, 2015.

P. M. Lorusso, C. M. Rudin, J. C. Reddy, R. Tibes, G. J. Weiss et al., Phase I trial of hedgehog pathway inhibitor vismodegib (GDC-0449) in patients with refractory, locally advanced or metastatic solid tumors, Clin Cancer Res Off J Am Assoc Cancer Res. 15 avr, vol.17, issue.8, pp.2502-2513, 2011.

A. Sekulic, M. R. Migden, A. E. Oro, L. Dirix, K. D. Lewis et al., Efficacy and safety of vismodegib in advanced basal-cell carcinoma, N Engl J Med. 7 juin, vol.366, issue.23, pp.2171-2180, 2012.

J. Mateo, S. Carreira, S. Sandhu, S. Miranda, H. Mossop et al., DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer, N Engl J Med, vol.373, issue.18, pp.1697-708, 2015.

E. R. Plimack, R. L. Dunbrack, T. A. Brennan, M. D. Andrake, Y. Zhou et al., Defects in DNA Repair Genes Predict Response to Neoadjuvant Cisplatin-based Chemotherapy in Muscleinvasive Bladder Cancer, Eur Urol. déc, vol.68, issue.6, pp.959-67, 2015.

M. Eich, W. P. Roos, T. Nikolova, and B. Kaina, Contribution of ATM and ATR to the resistance of glioblastoma and malignant melanoma cells to the methylating anticancer drug temozolomide, Mol Cancer Ther, vol.12, issue.11, pp.2529-2569, 2013.

, JAK inhibitor in CALR-mutant myelofibrosis. -PubMed -NCBI

D. Sur,

C. C. Wong, I. Martincorena, A. G. Rust, M. Rashid, C. Alifrangis et al., Inactivating CUX1 mutations promote tumorigenesis, Nat Genet. janv, vol.46, issue.1, pp.33-41, 2014.

Z. M. Ramdzan and A. Nepveu, CUX1, a haploinsufficient tumour suppressor gene overexpressed in advanced cancers, Nat Rev Cancer, vol.14, issue.10, pp.673-82, 2014.

S. P. Treon, L. Xu, and Z. Hunter, MYD88 Mutations and Response to Ibrutinib in Waldenström's Macroglobulinemia, N Engl J Med. 6 août, vol.373, issue.6, pp.584-590, 2015.

S. T. Kim, J. Lee, S. H. Park, J. O. Park, Y. S. Park et al., Prospective phase II trial of everolimus in PIK3CA amplification/mutation and/or PTEN loss patients with advanced solid tumors refractory to standard therapy, BMC Cancer, vol.23, issue.1, p.211, 2017.

T. A. Yap, L. Yan, A. Patnaik, I. Fearen, D. Olmos et al., First-in-man clinical trial of the oral pan-AKT inhibitor MK-2206 in patients with advanced solid tumors, J Clin Oncol Off J Am Soc Clin Oncol. 10 déc, vol.29, issue.35, pp.4688-95, 2011.

A. J. Templeton, V. Dutoit, R. Cathomas, C. Rothermundt, D. Bärtschi et al., Phase 2 trial of single-agent everolimus in chemotherapy-naive patients with castration-resistant prostate cancer (SAKK 08/08), Eur Urol. juill, vol.64, issue.1, pp.150-158, 2013.

, PTEN activity could be a predictive marker of trastuzumab efficacy in the treatment of ErbB2-overexpressing breast cancer

D. Sur,

N. D. Peyser, M. Freilino, L. Wang, Y. Zeng, H. Li et al., Frequent promoter hypermethylation of PTPRT increases STAT3 activation and sensitivity to STAT3 inhibition in head and neck cancer, Oncogene. 3 mars, vol.35, issue.9, pp.1163-1172, 2016.

A. H. Berger, M. Imielinski, F. Duke, J. Wala, N. Kaplan et al., Oncogenic RIT1 mutations in lung adenocarcinoma, Oncogene. 28 août, vol.33, issue.35, pp.4418-4441, 2014.