M. Certo, D. G. Moore, V. Nishino, M. Wei, G. Korsmeyer et al., Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members, Cancer Cell, vol.9, issue.5, pp.351-65, 2006.
DOI : 10.1016/j.ccr.2006.03.027

X. Chi, J. Kale, B. Leber, and D. Andrews, Regulating cell death at, on, and in membranes, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1843, issue.9, pp.2100-2113, 2014.
DOI : 10.1016/j.bbamcr.2014.06.002

F. Llambi, Y. Wang, B. Victor, M. Yang, D. Schneider et al., BOK Is a Non-canonical BCL-2 Family Effector of Apoptosis Regulated by ER-Associated Degradation, Cell, vol.165, issue.2, pp.421-454, 2016.
DOI : 10.1016/j.cell.2016.02.026

Y. Tsujimoto, L. Finger, J. Yunis, P. Nowell, and C. Croce, Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation, Science, vol.226, issue.4678, pp.1097-1106, 1984.
DOI : 10.1126/science.6093263

M. Cleary, S. Smith, and J. Sklar, Cloning and structural analysis of cDNAs for bcl-2 and a hybrid bcl-2/immunoglobulin transcript resulting from the t(14;18) translocation, Cell, vol.47, issue.1, pp.19-28, 1986.
DOI : 10.1016/0092-8674(86)90362-4

D. Vaux, C. S. Adams, and J. , Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells, Nature, vol.335, issue.6189, pp.440-442, 1988.
DOI : 10.1038/335440a0

T. Mcdonnell, N. Deane, F. Platt, G. Nunez, U. Jaeger et al., bcl-2-Immunoglobulin transgenic mice demonstrate extended B cell survival and follicular lymphoproliferation, Cell, vol.57, issue.1, pp.79-88, 1989.
DOI : 10.1016/0092-8674(89)90174-8

D. Hockenbery, G. Nuñez, C. Milliman, R. Schreiber, and S. Korsmeyer, Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death, Nature, vol.348, issue.6299, pp.334-340, 1990.
DOI : 10.1038/348334a0

J. Reed and M. Pellecchia, Apoptosis-based therapies for hematologic malignancies, Blood, vol.106, issue.2, pp.408-426, 2005.
DOI : 10.1182/blood-2004-07-2761

J. Huang, W. Fairbrother, and J. Reed, Therapeutic targeting of Bcl-2 family for treatment of B-cell malignancies, Expert Review of Hematology, vol.14, issue.3, pp.283-97, 2015.
DOI : 10.1038/sj.cdd.4402178

G. Lessene, P. Czabotar, and P. Colman, BCL-2 family antagonists for cancer therapy, Nature Reviews Drug Discovery, vol.35, issue.12, pp.989-1000, 2008.
DOI : 10.1093/nar/28.1.235

G. Bird, F. Bernal, K. Pitter, and L. Walensky, Chapter 22 Synthesis and Biophysical Characterization of Stabilized ?????Helices of BCL???2 Domains, Methods Enzymol, vol.446, pp.369-86, 2008.
DOI : 10.1016/S0076-6879(08)01622-4

A. Ashkenazi, W. Fairbrother, J. Leverson, and A. Souers, From basic apoptosis discoveries to advanced selective BCL-2 family inhibitors, Nature Reviews Drug Discovery, vol.2, issue.4, pp.273-84, 2017.
DOI : 10.1002/prot.24816

M. Hengartner, R. Ellis, and H. Horvitz, Caenorhabditis elegans gene ced-9 protects cells from programmed cell death, Nature, vol.356, issue.6369, pp.494-503, 1992.
DOI : 10.1038/356494a0

M. Hengartner and H. Horvitz, C. elegans cell survival gene ced-9 encodes a functional homolog of the mammalian proto-oncogene bcl-2, Cell, vol.76, issue.4, pp.665-76, 1994.
DOI : 10.1016/0092-8674(94)90506-1

URL : https://hal.archives-ouvertes.fr/pasteur-00456477

J. Reed, M. Cuddy, T. Slabiak, C. Croce, and P. Nowell, Oncogenic potential of bcl-2 demonstrated by gene transfer, Nature, vol.336, issue.6196, pp.259-61, 1988.
DOI : 10.1038/336259a0

R. Bissonnette, F. Echeverri, A. Mahboubi, and D. Green, Apoptotic cell death induced by c-myc is inhibited by bcl-2, Nature, vol.359, issue.6395, pp.552-556, 1992.
DOI : 10.1038/359552a0

F. Abdallah, E. Harrington, and G. Evan, Cooperative interaction between c-myc and bcl-2 proto-oncogenes, Nature, vol.359, pp.554-560, 1992.

R. Soderquist and A. Eastman, BCL2 Inhibitors as Anticancer Drugs: A Plethora of Misleading BH3 Mimetics, Molecular Cancer Therapeutics, vol.15, issue.9, pp.2011-2018, 2016.
DOI : 10.1158/1535-7163.MCT-16-0031

T. Oltersdorf, S. Elmore, A. Shoemaker, R. Armstrong, D. Augeri et al., An inhibitor of Bcl-2 family proteins induces regression of solid tumours, Nature, vol.360, issue.7042, pp.677-81, 2005.
DOI : 10.1016/0014-5793(95)00062-E

D. Gaizo-moore, V. Brown, J. Certo, M. Love, T. Novina et al., Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737, Journal of Clinical Investigation, vol.117, issue.1, pp.112-133, 2007.
DOI : 10.1172/JCI28281DS1

T. Tsao, Y. Shi, S. Kornblau, H. Lu, S. Konoplev et al., Concomitant inhibition of DNA methyltransferase and BCL-2 protein function synergistically induce mitochondrial apoptosis in acute myelogenous leukemia cells, Annals of Hematology, vol.119, issue.12, pp.1861-70, 2012.
DOI : 10.1182/blood-2011-08-375758

C. Tse, A. Shoemaker, J. Adickes, M. Anderson, J. Chen et al., ABT-263: A Potent and Orally Bioavailable Bcl-2 Family Inhibitor, Cancer Research, vol.68, issue.9, pp.3421-3429, 2008.
DOI : 10.1158/0008-5472.CAN-07-5836

K. Wagner, C. E. Rucker, E. Riedlinger, G. Broussard, C. Schwartzberg et al., Conditional deletion of the Bcl-x gene from erythroid cells results in hemolytic anemia and profound splenomegaly, Development, vol.127, pp.4949-58, 2000.

K. Mason, M. Carpinelli, J. Fletcher, J. Collinge, A. Hilton et al., Programmed Anuclear Cell Death Delimits Platelet Life Span, Cell, vol.128, issue.6, pp.1173-86, 2007.
DOI : 10.1016/j.cell.2007.01.037

H. Zhang, P. Nimmer, S. Tahir, J. Chen, R. Fryer et al., Bcl-2 family proteins are essential for platelet survival, Cell Death and Differentiation, vol.53, pp.943-51, 2007.
DOI : 10.1097/01.fjc.0000171755.28317.85

E. Lee, S. Grabow, S. Chappaz, G. Dewson, C. Hockings et al., Physiological restraint of Bak by Bcl-xL is essential for cell survival, Genes Dev, vol.30, pp.1240-50, 2016.

D. Veis, C. Sorenson, J. Shutter, and S. Korsmeyer, Bcl-2-deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented hair, Cell, vol.75, issue.2, pp.229-269, 1993.
DOI : 10.1016/0092-8674(93)80065-M

K. Nakayama, K. Nakayama, I. Negishi, K. Kuida, H. Sawa et al., Targeted disruption of Bcl-2 alpha beta in mice: occurrence of gray hair, polycystic kidney disease, and lymphocytopenia., Proceedings of the National Academy of Sciences, vol.91, issue.9, pp.3700-3704, 1994.
DOI : 10.1073/pnas.91.9.3700

A. Roberts, J. Seymour, J. Brown, W. Wierda, T. Kipps et al., Substantial Susceptibility of Chronic Lymphocytic Leukemia to BCL2 Inhibition: Results of a Phase I Study of Navitoclax in Patients With Relapsed or Refractory Disease, Journal of Clinical Oncology, vol.30, issue.5, pp.488-96, 2012.
DOI : 10.1200/JCO.2011.34.7898

W. Wilson, O. Connor, O. Czuczman, M. Lacasce, A. Gerecitano et al., Navitoclax, a targeted high-affinity inhibitor of BCL-2, in lymphoid malignancies: a phase 1 dose-escalation study of safety, pharmacokinetics, pharmacodynamics, and antitumour activity, The Lancet Oncology, vol.11, issue.12, pp.1149-59, 2010.
DOI : 10.1016/S1470-2045(10)70261-8

A. Roberts, R. Advani, B. Kahl, D. Persky, J. Sweetenham et al., lymphoid malignancies, British Journal of Haematology, vol.96, issue.5, pp.669-78, 2015.
DOI : 10.1038/sj.bjc.6603762

A. Souers, J. Leverson, E. Boghaert, S. Ackler, N. Catron et al., ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets, Nature Medicine, vol.559, issue.2, pp.202-210, 2013.
DOI : 10.1038/leu.2009.151

C. Tam, A. Roberts, M. Anderson, S. Dawson, R. Hicks et al., COMBINATION IBRUTINIB (IBR) AND VENETOCLAX (VEN) FOR THE TREATMENT OF MANTLE CELL LYMPHOMA (MCL): PRIMARY ENDPOINT ASSESSMENT OF THE PHASE 2 AIM STUDY, Presented at: International Conference on Malignant Lymphoma (ICML), 2017.
DOI : 10.1002/hon.2437_134

D. Chiron, C. Dousset, C. Brosseau, C. Touzeau, S. Maïga et al., Biological rational for sequential targeting of Bruton tyrosine kinase and Bcl-2 to overcome CD40-induced ABT-199 resistance in mantle cell lymphoma, Oncotarget, vol.6, issue.11, pp.8750-8759, 2015.
DOI : 10.18632/oncotarget.3275

B. Sun, B. Shah, W. Fiskus, J. Qi, K. Rajapakshe et al., Synergistic activity of BET protein antagonist-based combinations in mantle cell lymphoma cells sensitive or resistant to ibrutinib, Blood, vol.126, issue.13, pp.1565-74, 2015.
DOI : 10.1182/blood-2015-04-639542

J. Qin, J. Ziffra, L. Stennett, B. Bodner, B. Bonish et al., Proteasome Inhibitors Trigger NOXA-Mediated Apoptosis in Melanoma and Myeloma Cells, Cancer Research, vol.65, issue.14, pp.6282-93, 2005.
DOI : 10.1158/0008-5472.CAN-05-0676

P. Gomez-bougie, S. Wuillème-toumi, E. Ménoret, V. Trichet, N. Robillard et al., Noxa Up-regulation and Mcl-1 Cleavage Are Associated to Apoptosis Induction by Bortezomib in Multiple Myeloma, Cancer Research, vol.67, issue.11, pp.5418-5442, 2007.
DOI : 10.1158/0008-5472.CAN-06-4322

P. Gomez-bougie, E. Ménoret, P. Juin, C. Dousset, C. Pellat-deceunynck et al., Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction, Biochemical and Biophysical Research Communications, vol.413, issue.3, pp.460-464, 2011.
DOI : 10.1016/j.bbrc.2011.08.118

E. Punnoose, J. Leverson, F. Peale, E. Boghaert, L. Belmont et al., Expression Profile of BCL-2, BCL-XL, and MCL-1 Predicts Pharmacological Response to the BCL-2 Selective Antagonist Venetoclax in Multiple Myeloma Models, Molecular Cancer Therapeutics, vol.15, issue.5, pp.1132-1176, 2016.
DOI : 10.1158/1535-7163.MCT-15-0730

P. Moreau, A. Chanan-khan, A. Roberts, A. Agarwal, T. Facon et al., Promising efficacy and acceptable safety of venetoclax plus bortezomib and dexamethasone in relapsed/refractory MM, Blood, vol.130, issue.22, 2017.
DOI : 10.1182/blood-2017-06-788323

S. Chen, Y. Dai, X. Pei, and S. Grant, Bim Upregulation by Histone Deacetylase Inhibitors Mediates Interactions with the Bcl-2 Antagonist ABT-737: Evidence for Distinct Roles for Bcl-2, Bcl-xL, and Mcl-1, Molecular and Cellular Biology, vol.29, issue.23
DOI : 10.1128/MCB.01481-08

S. Chen, Y. Zhang, L. Zhou, Y. Leng, H. Lin et al., A Bim-targeting strategy overcomes adaptive bortezomib resistance in myeloma through a novel link between autophagy and apoptosis, Blood, vol.124, issue.17, pp.2687-97, 2014.
DOI : 10.1182/blood-2014-03-564534

L. Bieghs, S. Lub, K. Fostier, K. Maes, V. Valckenborgh et al., The IGF-1 receptor inhibitor picropodophyllin potentiates the anti-myeloma activity of a BH3-mimetic, Oncotarget, vol.5, issue.22, pp.11193-208, 2014.
DOI : 10.18632/oncotarget.1933

P. Gomez-bougie and M. Amiot, Apoptotic Machinery Diversity in Multiple Myeloma Molecular Subtypes, Frontiers in Immunology, vol.4, p.467, 2013.
DOI : 10.3389/fimmu.2013.00467

L. Bodet, P. Gomez-bougie, C. Touzeau, C. Dousset, G. Descamps et al., ABT-737 is highly effective against molecular subgroups of multiple myeloma, Blood, vol.118, issue.14, pp.3901-3911, 2011.
DOI : 10.1182/blood-2010-11-317438

C. Touzeau, C. Dousset, L. Gouill, S. Sampath, D. Leverson et al., The Bcl-2 specific BH3 mimetic ABT-199: a promising targeted therapy for t(11;14) multiple myeloma, Leukemia, vol.21, issue.1, pp.210-212, 2014.
DOI : 10.1038/leu.2011.176

S. Kumar, J. Kaufman, C. Gasparetto, J. Mikhael, R. Vij et al., Efficacy of venetoclax as targeted therapy for relapsed/refractory t(11;14) multiple myeloma, Blood, vol.130, issue.22, 2017.
DOI : 10.1182/blood-2017-06-788786

URL : https://hal.archives-ouvertes.fr/inserm-01631468

C. Touzeau, J. Ryan, J. Guerriero, P. Moreau, T. Chonghaile et al., BH3 profiling identifies heterogeneous dependency on Bcl-2 family members in multiple myeloma and predicts sensitivity to BH3 mimetics, Leukemia, vol.120, issue.3, pp.761-765, 2016.
DOI : 10.1016/j.ymeth.2013.04.006

URL : https://hal.archives-ouvertes.fr/inserm-01411102

J. Gong, T. Khong, D. Segal, Y. Yao, C. Riffkin et al., Hierarchy for targeting prosurvival BCL2 family proteins in multiple myeloma: pivotal role of MCL1, Blood, vol.128, issue.14, pp.1834-1878, 2016.
DOI : 10.1182/blood-2016-03-704908

F. Ravandi, J. Cortes, S. Faderl, O. Brien, S. Garcia-manero et al., Characteristics and outcome of patients with acute myeloid leukemia refractory to 1 cycle of high-dose cytarabine-based induction chemotherapy, Blood, vol.116, issue.26, pp.5818-5841, 2010.
DOI : 10.1182/blood-2010-07-296392

A. Burnett, M. Wetzler, and B. Löwenberg, Therapeutic Advances in Acute Myeloid Leukemia, Journal of Clinical Oncology, vol.29, issue.5, pp.487-94, 2011.
DOI : 10.1200/JCO.2010.30.1820

M. Konopleva, R. Contractor, T. Tsao, I. Samudio, P. Ruvolo et al., Mechanisms of apoptosis sensitivity and resistance to the BH3 mimetic ABT-737 in acute myeloid leukemia, Cancer Cell, vol.10, issue.5, pp.375-88, 2006.
DOI : 10.1016/j.ccr.2006.10.006

T. Vo, J. Ryan, R. Carrasco, D. Neuberg, D. Rossi et al., Relative Mitochondrial Priming of Myeloblasts and Normal HSCs Determines Chemotherapeutic Success in AML, Cell, vol.151, issue.2, pp.344-55, 2012.
DOI : 10.1016/j.cell.2012.08.038

X. Niu, G. Wang, Y. Wang, J. Caldwell, H. Edwards et al., Acute myeloid leukemia cells harboring MLL fusion genes or with the acute promyelocytic leukemia phenotype are sensitive to the Bcl-2-selective inhibitor ABT-199, Leukemia, vol.4, issue.7, pp.1557-60, 2014.
DOI : 10.1038/leu.2013.216

R. Pan, L. Hogdal, J. Benito, D. Bucci, L. Han et al., Selective BCL-2 Inhibition by ABT-199 Causes On-Target Cell Death in Acute Myeloid Leukemia, Cancer Discovery, vol.4, issue.3, pp.362-75, 2014.
DOI : 10.1158/2159-8290.CD-13-0609

J. Ishizawa, K. Kojima, T. Mcqueen, V. Ruvolo, D. Chachad et al., Mitochondrial Profiling of Acute Myeloid Leukemia in the Assessment of Response to Apoptosis Modulating Drugs, PLOS ONE, vol.5, issue.23, p.138377, 2015.
DOI : 10.1371/journal.pone.0138377.s002

S. Chan, D. Thomas, M. Corces-zimmerman, S. Xavy, S. Rastogi et al., Isocitrate dehydrogenase 1 and 2 mutations induce BCL-2 dependence in acute myeloid leukemia, Nature Medicine, vol.17, issue.2, pp.178-84, 2015.
DOI : 10.1016/j.ccr.2010.01.020

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, pp.339-383, 2010.
DOI : 10.1126/science.1170944

M. Kontro, A. Kumar, M. Majumder, S. Eldfors, A. Parsons et al., HOX gene expression predicts response to BCL-2 inhibition in acute myeloid leukemia, Leukemia, vol.9, issue.2, pp.301-310, 2017.
DOI : 10.1007/s11899-014-0198-1

M. Konopleva, D. 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 Discovery, vol.6, issue.10, pp.1106-1123, 2016.
DOI : 10.1158/2159-8290.CD-16-0313

J. Bogenberger, S. Kornblau, W. Pierceall, R. Lena, D. Chow et al., BCL-2 family proteins as 5-Azacytidine-sensitizing targets and determinants of response in myeloid malignancies, Leukemia, vol.70, issue.8, pp.1657-65, 2014.
DOI : 10.1158/1535-7163.MCT-07-0010

J. Bogenberger, D. Delman, N. Hansen, R. Valdez, V. Fauble et al., activity of BCL-2 family inhibitors ABT-199 and ABT-737 combined with 5-azacytidine in myeloid malignancies, Leukemia & Lymphoma, vol.31, issue.1, pp.226-235, 2015.
DOI : 10.1016/j.stem.2012.12.013

K. Pratz, D. Pollyea, J. B. Pullarkat, V. Wei, A. Arellano et al., Safety and efficacy of venetoclax in combination with decitabine or azacitidine in treatment-naïve, elderly patients ?65 years with acute myeloid leukemia, 2017.

A. Wei, S. Strickland, G. Roboz, J. Hou, W. Fiedler et al., Safety and efficacy of venetoclax plus low-dose cytarabine in treatment-naive patients aged ?65 years with acute myeloid leukemia, Blood, vol.128, p.102, 2016.

K. Kojima, M. Konopleva, I. Samudio, W. Schober, W. Bornmann et al., Concomitant Inhibition of MDM2 and Bcl-2 Protein Function Synergistically Induce Mitochondrial Apoptosis in AML, Cell Cycle, vol.5, issue.23, pp.2778-86, 2006.
DOI : 10.4161/cc.5.23.3520

L. Han, Q. Zhang, C. Shi, A. Cavazos, V. Ruvolo et al., Targeting MAPK Signaling Pathway with Cobimetinib (GDC-0973) Enhances Anti-Leukemia Efficacy of Venetoclax (ABT-199/GDC-0199) in Acute Myeloid Leukemia Models, Clinical Lymphoma Myeloma and Leukemia, vol.17, p.97, 2016.
DOI : 10.1016/j.clml.2017.07.064

M. Konopleva, M. Milella, P. Ruvolo, J. Watts, M. Ricciardi et al., MEK inhibition enhances ABT-737-induced leukemia cell apoptosis via prevention of ERK-activated MCL-1 induction and modulation of MCL-1/BIM complex, Leukemia, vol.118, issue.4, pp.778-87, 2012.
DOI : 10.1172/JCI35437

L. Jin, Y. Tabe, K. Kojima, M. Shikami, J. Benito et al., PI3K inhibitor GDC-0941 enhances apoptotic effects of BH-3 mimetic ABT-737 in AML cells in the hypoxic bone marrow microenvironment, Journal of Molecular Medicine, vol.16, issue.12, pp.1383-97, 2013.
DOI : 10.1038/sj.leu.2402608

M. Rahmani, M. Aust, E. Hawkins, R. Parker, M. Ross et al., Co-administration of the mTORC1/TORC2 inhibitor INK128 and the Bcl-2/Bcl-xL antagonist ABT-737 kills human myeloid leukemia cells through Mcl-1 down-regulation and AKT inactivation, Haematologica, vol.100, issue.12, pp.1553-63, 2015.
DOI : 10.3324/haematol.2015.130351

K. Airiau, V. Prouzet-mauléon, B. Rousseau, A. Pigneux, M. Jeanneteau et al., Synergistic cooperation between ABT-263 and MEK1/2 inhibitor: effect on apoptosis and proliferation of acute myeloid leukemia cells, Oncotarget, vol.7, issue.1, pp.845-59, 2016.
DOI : 10.18632/oncotarget.6417

W. Zhang, V. Ruvolo, C. Gao, L. Zhou, W. Bornmann et al., Evaluation of Apoptosis Induction by Concomitant Inhibition of MEK, mTOR, and Bcl-2 in Human Acute Myelogenous Leukemia Cells, Molecular Cancer Therapeutics, vol.13, issue.7, pp.1848-59, 2014.
DOI : 10.1158/1535-7163.MCT-13-0576

K. Knorr, P. Schneider, X. Meng, H. Dai, B. Smith et al., MLN4924 induces Noxa upregulation in acute myelogenous leukemia and synergizes with Bcl-2 inhibitors, Cell Death and Differentiation, vol.49, issue.12, pp.2133-2175, 2015.
DOI : 10.1016/0003-2697(85)90442-7

C. Lehmann, T. Friess, F. Birzele, A. Kiialainen, and M. Dangl, Superior anti-tumor activity of the MDM2 antagonist idasanutlin and the Bcl-2 inhibitor venetoclax in p53 wild-type acute myeloid leukemia models, Journal of Hematology & Oncology, vol.7, issue.2, p.50, 2016.
DOI : 10.1186/s13045-014-0070-8

R. Pan, V. Ruvolo, H. Mu, J. Leverson, G. Nichols et al., Synthetic lethality of combined Bcl-2 inhibition and p53 activation in AML: novel mechanisms and superior antileukemic efficacy, Cancer Cell, 2017.

S. Beurlet, N. Omidvar, P. Gorombei, P. Krief, L. Pogam et al., BCL-2 inhibition with ABT-737 prolongs survival in an NRAS/BCL-2 mouse model of AML by targeting primitive LSK and progenitor cells, Blood, vol.122, issue.16, pp.2864-76, 2013.
DOI : 10.1182/blood-2012-07-445635

S. Jilg, V. Reidel, C. Müller-thomas, J. König, J. Schauwecker et al., Blockade of BCL-2 proteins efficiently induces apoptosis in progenitor cells of high-risk myelodysplastic syndromes patients, Leukemia, vol.115, issue.1, pp.112-135, 2016.
DOI : 10.3324/haematol.2014.104760

S. Suryani, H. Carol, T. Chonghaile, V. Frismantas, C. Sarmah et al., Cell and Molecular Determinants of In Vivo Efficacy of the BH3 Mimetic ABT-263 against Pediatric Acute Lymphoblastic Leukemia Xenografts, Clinical Cancer Research, vol.20, issue.17, pp.4520-4551, 2014.
DOI : 10.1158/1078-0432.CCR-14-0259

S. Alford, A. Kothari, F. Loeff, J. Eichhorn, N. Sakurikar et al., BH3 Inhibitor Sensitivity and Bcl-2 Dependence in Primary Acute Lymphoblastic Leukemia Cells, Cancer Research, vol.75, issue.7, pp.1366-75, 2015.
DOI : 10.1158/0008-5472.CAN-14-1849

T. Chonghaile, J. Roderick, C. Glenfield, J. Ryan, S. Sallan et al., Maturation Stage of T-cell Acute Lymphoblastic Leukemia Determines BCL-2 versus BCL-XL Dependence and Sensitivity to ABT-199, Cancer Discovery, vol.4, issue.9, pp.1074-87, 2014.
DOI : 10.1158/2159-8290.CD-14-0353

N. Anderson, I. Harrold, M. Mansour, T. Sanda, M. Mckeown et al., BCL2-specific inhibitor ABT-199 synergizes strongly with cytarabine against the early immature LOUCY cell line but not more-differentiated T-ALL cell lines, Leukemia, vol.8, issue.5, pp.1145-1153, 2014.
DOI : 10.1038/nm.3048

S. Peirs, F. Matthijssens, S. Goossens, I. Van-de-walle, K. Ruggero et al., ABT-199 mediated inhibition of BCL-2 as a novel therapeutic strategy in T-cell acute lymphoblastic leukemia, Blood, vol.124, issue.25, pp.3738-3785, 2014.
DOI : 10.1182/blood-2014-05-574566

J. Benito, L. Godfrey, K. Kojima, L. Hogdal, M. Wunderlich et al., MLL-Rearranged Acute Lymphoblastic Leukemias Activate BCL-2 through H3K79 Methylation and Are Sensitive to the BCL-2-Specific Antagonist ABT-199, Cell Reports, vol.13, issue.12, pp.2715-2742, 2015.
DOI : 10.1016/j.celrep.2015.12.003

S. Khaw, S. Suryani, K. Evans, R. J. Robbins, A. Kurmasheva et al., Venetoclax responses of pediatric ALL xenografts reveal sensitivity of MLL-rearranged leukemia, Blood, vol.128, issue.10, pp.1382-95, 2016.
DOI : 10.1182/blood-2016-03-707414

T. Ko, C. Chuah, J. Huang, K. Ng, and S. Ong, The BCL2 inhibitor ABT-199 significantly enhances imatinib-induced cell death in chronic myeloid leukemia progenitors, Oncotarget, vol.5, issue.19, pp.9033-9041, 2014.
DOI : 10.18632/oncotarget.1925

B. Carter, P. Mak, H. Mu, H. Zhou, D. Mak et al., Combined targeting of BCL-2 and BCR?ABL tyrosine kinase eradicates chronic myeloid leukemia stem cells, Sci Transl Med, vol.8, pp.355-117, 2016.

E. Carrington, J. Zhang, R. Sutherland, I. Vikstrom, J. Brady et al., Prosurvival Bcl-2 family members reveal a distinct apoptotic identity between conventional and plasmacytoid dendritic cells, Proceedings of the National Academy of Sciences, vol.14, issue.1, pp.4044-4053, 2015.
DOI : 10.1182/blood-2013-01-475855

Y. Zhan, E. Carrington, H. Ko, I. Vikstrom, S. Oon et al., Bcl-2 Antagonists Kill Plasmacytoid Dendritic Cells From Lupus-Prone Mice and Dampen Interferon-?? Production, Arthritis & Rheumatology, vol.8, issue.3, pp.797-808, 2015.
DOI : 10.2215/CJN.05870612

J. Montero, J. Stephansky, T. Cai, G. Griffin, L. Cabal-hierro et al., Blastic Plasmacytoid Dendritic Cell Neoplasm Is Dependent on BCL2 and Sensitive to Venetoclax, Cancer Discovery, vol.7, issue.2, pp.156-64, 2017.
DOI : 10.1158/2159-8290.CD-16-0999

D. Cittelly, P. Das, V. Salvo, J. Fonseca, M. Burow et al., Oncogenic HER2??16 suppresses miR-15a/16 and deregulates BCL-2 to promote endocrine resistance of breast tumors, Carcinogenesis, vol.31, issue.12, pp.2049-57, 2010.
DOI : 10.1093/carcin/bgq192

F. Vaillant, D. Merino, L. Lee, K. Breslin, B. Pal et al., Targeting BCL-2 with the BH3 Mimetic ABT-199 in Estrogen Receptor-Positive Breast Cancer, Cancer Cell, vol.24, issue.1, pp.120-129, 2013.
DOI : 10.1016/j.ccr.2013.06.002

A. Stone, M. Cowley, F. Valdes-mora, R. Mccloy, C. Sergio et al., BCL-2 Hypermethylation Is a Potential Biomarker of Sensitivity to Antimitotic Chemotherapy in Endocrine-Resistant Breast Cancer, Molecular Cancer Therapeutics, vol.12, issue.9, pp.1874-85, 2013.
DOI : 10.1158/1535-7163.MCT-13-0012

L. Zheng, Y. W. Zhang, C. Ding, W. Zhu, H. Lin et al., GDC-0941 sensitizes breast cancer to ABT-737 in vitro and in vivo through promoting the degradation of Mcl-1, Cancer Letters, vol.309, issue.1, pp.27-36, 2011.
DOI : 10.1016/j.canlet.2011.05.011

T. Muranen, L. Selfors, D. Worster, M. Iwanicki, L. Song et al., Inhibition of PI3K/mTOR Leads to Adaptive Resistance in Matrix-Attached Cancer Cells, Cancer Cell, vol.21, issue.2, pp.227-266, 2012.
DOI : 10.1016/j.ccr.2011.12.024

C. Séveno, D. Loussouarn, S. Bréchet, M. Campone, and P. Juin, ??-Secretase inhibition promotes cell death, Noxa upregulation, and sensitization to BH3 mimetic ABT-737 in human breast cancer cells, Breast Cancer Research, vol.435, issue.Suppl 1, p.96, 2012.
DOI : 10.1038/nature03659

S. Tahir, X. Yang, M. Anderson, S. Morgan-lappe, A. Sarthy et al., Influence of Bcl-2 Family Members on the Cellular Response of Small-Cell Lung Cancer Cell Lines to ABT-737, Cancer Research, vol.67, issue.3, pp.1176-83, 2007.
DOI : 10.1158/0008-5472.CAN-06-2203

C. Hann, V. Daniel, E. Sugar, I. Dobromilskaya, S. Murphy et al., Therapeutic Efficacy of ABT-737, a Selective Inhibitor of BCL-2, in Small Cell Lung Cancer, Cancer Research, vol.68, issue.7, pp.2321-2329, 2008.
DOI : 10.1158/0008-5472.CAN-07-5031

A. Shoemaker, M. Mitten, J. Adickes, S. Ackler, M. Refici et al., Activity of the Bcl-2 Family Inhibitor ABT-263 in a Panel of Small Cell Lung Cancer Xenograft Models, Clinical Cancer Research, vol.14, issue.11, pp.3268-77, 2008.
DOI : 10.1158/1078-0432.CCR-07-4622

S. Tahir, J. Wass, M. Joseph, V. Devanarayan, P. Hessler et al., Identification of Expression Signatures Predictive of Sensitivity to the Bcl-2 Family Member Inhibitor ABT-263 in Small Cell Lung Carcinoma and Leukemia/Lymphoma Cell Lines, Molecular Cancer Therapeutics, vol.9, issue.3, pp.545-57, 2010.
DOI : 10.1158/1535-7163.MCT-09-0651

L. Gandhi, D. Camidge, R. De-oliveira, M. Bonomi, P. Gandara et al., Phase I Study of Navitoclax (ABT-263), a Novel Bcl-2 Family Inhibitor, in Patients With Small-Cell Lung Cancer and Other Solid Tumors, Journal of Clinical Oncology, vol.29, issue.7, pp.909-925, 2011.
DOI : 10.1200/JCO.2010.31.6208

C. Rudin, C. Hann, E. Garon, R. De-oliveira, M. Bonomi et al., Phase II Study of Single-Agent Navitoclax (ABT-263) and Biomarker Correlates in Patients with Relapsed Small Cell Lung Cancer, Clinical Cancer Research, vol.18, issue.11, pp.3163-3172, 2012.
DOI : 10.1158/1078-0432.CCR-11-3090

A. Mattoo and D. Fitzgerald, Combination treatments with ABT-263 and an immunotoxin produce synergistic killing of ABT-263-resistant small cell lung cancer cell lines, International Journal of Cancer, vol.6, issue.Suppl 2, pp.978-87, 2013.
DOI : 10.1371/journal.pone.0024012

E. Gardner, N. Connis, J. Poirier, L. Cope, I. Dobromilskaya et al., Rapamycin Rescues ABT-737 Efficacy in Small Cell Lung Cancer, Cancer Research, vol.74, issue.10, pp.2846-56, 2014.
DOI : 10.1158/0008-5472.CAN-13-3460

W. Nakajima, K. Sharma, M. Hicks, N. Le, R. Brown et al., Combination with vorinostat overcomes ABT-263 (navitoclax) resistance of small cell lung cancer, Cancer Biology & Therapy, vol.54, issue.1, pp.27-35, 2016.
DOI : 10.1016/S1470-2045(10)70261-8

D. Potter, M. Galvin, S. Brown, A. Lallo, C. Hodgkinson et al., Inhibition of PI3K/BMX Cell Survival Pathway Sensitizes to BH3 Mimetics in SCLC, Molecular Cancer Therapeutics, vol.15, issue.6, pp.1248-60, 2016.
DOI : 10.1158/1535-7163.MCT-15-0885

K. Ko, J. Wang, S. Perper, Y. Jiang, D. Yanez et al., Bcl-2 as a Therapeutic Target in Human Tubulointerstitial Inflammation, Arthritis & Rheumatology, vol.18, issue.4, pp.2740-51, 2016.
DOI : 10.1681/ASN.2006090992

M. Smith, B. Chyla, E. Mckeegan, and S. Tahir, Development of a flow cytometric method for quantification of BCL-2 family members in chronic lymphocytic leukemia and correlation with sensitivity to BCL-2 family inhibitors, Cytometry Part B: Clinical Cytometry, vol.363, issue.5, 2016.
DOI : 10.1016/j.jim.2010.09.036

R. Thijssen, E. Slinger, K. Weller, C. Geest, T. Beaumont et al., Resistance to ABT-199 induced by microenvironmental signals in chronic lymphocytic leukemia can be counteracted by CD20 antibodies or kinase inhibitors, Haematologica, vol.100, pp.302-308, 2015.
DOI : 10.3324/haematol.2015.124560

T. Song, G. Chai, Y. Liu, X. Yu, Z. Wang et al., Bcl-2 phosphorylation confers resistance on chronic lymphocytic leukaemia cells to the BH3 mimetics ABT-737, ABT-263 and ABT-199 by impeding direct binding, British Journal of Pharmacology, vol.108, issue.Database Issue, pp.471-83, 2016.
DOI : 10.1038/bjc.2013.152

V. Fresquet, M. Rieger, C. Carolis, M. García-barchino, and J. Martinez-climent, Acquired mutations in BCL2 family proteins conferring resistance to the BH3 mimetic ABT-199 in lymphoma, Blood, vol.123, issue.26, pp.4111-4120, 2014.
DOI : 10.1182/blood-2014-03-560284

K. Bojarczuk, B. Sasi, S. Gobessi, I. Innocenti, G. Pozzato et al., BCR signaling inhibitors differ in their ability to overcome Mcl-1-mediated resistance of CLL B cells to ABT-199, Blood, vol.127, issue.25, pp.3192-201, 2016.
DOI : 10.1182/blood-2015-10-675009

J. Deng, E. Isik, S. Fernandes, J. Brown, A. Letai et al., Bruton???s tyrosine kinase inhibition increases BCL-2 dependence and enhances sensitivity to venetoclax in chronic lymphocytic leukemia, Leukemia, vol.374, issue.10, 2017.
DOI : 10.1182/blood-2013-12-543322

P. Hillmen, A. Rawstron, T. Munir, K. Brock, M. Vincente et al., The initial report of the Bloodwise TAP CLARITY study combining ibrutinib and venetoclax in relapsed, refractory CLL shows acceptable safety and promising early indications of efficacy, Presented at: European Hematology Association (EHA) Conference, 2017.

P. Zinzani, M. Topp, S. Yuen, C. Rusconi, I. Fleury et al., Phase 2 study of venetoclax plus rituximab or randomized venetoclax plus bendamustine + rituximab (BR) versus BR in patients with relapsed/refractory follicular lymphoma: interim data, Blood, vol.128, p.617, 2016.

S. De-vos, L. Swinnen, M. Kozloff, D. Wang, E. Reid et al., A dose-escalation study of venetoclax (ABT 199/GDC 0199) in combination with bendamustine and rituximab in patients with relapsed or refractory non-Hodgkin's lymphoma, Blood, vol.126, p.255, 2015.

A. Zelenetz, G. Salles, K. Mason, C. Casulo, L. Gouill et al., Phase 1B study of venetoclax plus R-or G-CHOP in patients with B-cell non-Hodgkin lymphoma, Blood, vol.128, p.3032, 2016.

X. Niu, J. Zhao, J. Ma, C. Xie, H. Edwards et al., Binding of Released Bim to Mcl-1 is a Mechanism of Intrinsic Resistance to ABT-199 which can be Overcome by Combination with Daunorubicin or Cytarabine in AML Cells, Clinical Cancer Research, vol.22, issue.17, pp.4440-51, 2016.
DOI : 10.1158/1078-0432.CCR-15-3057

T. Teh, N. Nguyen, D. Moujalled, D. Segal, G. Pomilio et al., Enhancing venetoclax activity in acute myeloid leukemia by co-targeting MCL1, Leukemia, vol.43, 2017.
DOI : 10.1038/nature08822

C. Touzeau, L. Gouill, S. Mahé, B. Boudreault, J. Gastinne et al., Deep and sustained response after venetoclax therapy in a patient with very advanced refractory myeloma with translocation t(11;14), Haematologica, vol.102, issue.3, pp.112-116, 2017.
DOI : 10.3324/haematol.2016.160408

URL : https://hal.archives-ouvertes.fr/inserm-01445538

S. Matulis, V. Gupta, A. Nooka, H. Hollen, J. Kaufman et al., Dexamethasone treatment promotes Bcl-2 dependence in multiple myeloma resulting in sensitivity to venetoclax, Leukemia, vol.33, issue.5, pp.1086-93, 2016.
DOI : 10.1016/j.cell.2015.01.042

Y. Cao, G. Yang, Z. Hunter, X. Liu, L. Xu et al., mutated Waldenstrom macroglobulinaemia cells, British Journal of Haematology, vol.122, issue.Suppl. 5, pp.134-142, 2015.
DOI : 10.1182/blood-2012-12-475111

G. Lindeman, S. Lok, A. Bergin, J. Whittle, K. Shackleton et al., Safety and efficacy of the BCL2 inhibitor venetoclax in estrogen receptor (ER) and BCL2-positive metastatic breast cancer: the mBEP study, J Clin Oncol, vol.35, p.1044, 2017.

R. Tanos, D. Karmali, S. Nalluri, and K. Goldsmith, Select Bcl-2 antagonism restores chemotherapy sensitivity in high-risk neuroblastoma, BMC Cancer, vol.160, issue.5, p.97, 2016.
DOI : 10.1016/j.cell.2015.01.042

L. Bate-eya, I. Hartog, I. Van-der-ploeg, L. Schild, J. Koster et al., High efficacy of the BCL-2 inhibitor ABT199 (venetoclax) in BCL-2 high-expressing neuroblastoma cell lines and xenografts and rational for combination with MCL-1 inhibition, Oncotarget, vol.7, issue.19, pp.27946-58, 2016.
DOI : 10.18632/oncotarget.8547

M. Cragg, E. Jansen, M. Cook, C. Harris, A. Strasser et al., Treatment of B-RAF mutant human tumor cells with a MEK inhibitor requires Bim and is enhanced by a BH3 mimetic, Journal of Clinical Investigation, vol.118, issue.11, pp.3651-3660, 2008.
DOI : 10.1172/JCI35437DS1

R. Corcoran, K. Cheng, A. Hata, A. Faber, H. Ebi et al., Synthetic Lethal Interaction of Combined BCL-XL and MEK Inhibition Promotes Tumor Regressions in KRAS Mutant Cancer Models, Cancer Cell, vol.23, issue.1, pp.121-129, 2013.
DOI : 10.1016/j.ccr.2012.11.007

Z. Tao, L. Hasvold, L. Wang, X. Wang, A. Petros et al., Activity, ACS Medicinal Chemistry Letters, vol.5, issue.10, pp.1088-93, 2014.
DOI : 10.1021/ml5001867

URL : https://hal.archives-ouvertes.fr/in2p3-00007672

G. Lessene, P. Czabotar, B. Sleebs, K. Zobel, K. Lowes et al., Structure-guided design of a selective BCL-XL inhibitor, Nature Chemical Biology, vol.60, issue.6, pp.390-397, 2013.
DOI : 10.1107/S0907444904019158

A. Kotschy, Z. Szlavik, J. Murray, J. Davidson, A. Maragno et al., The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models, Nature, vol.318, issue.7626, pp.477-82, 2016.
DOI : 10.1038/318533a0

A. Faber, A. Farago, C. Costa, A. Dastur, M. Gomez-caraballo et al., Assessment of ABT-263 activity across a cancer cell line collection leads to a potent combination therapy for small-cell lung cancer, Proceedings of the National Academy of Sciences, vol.112, issue.11, pp.1288-96, 2015.
DOI : 10.1038/nature11003