S. Kitada, I. Pedersen, A. Schimmer, and J. Reed, Dysregulation of apoptosis genes in hematopoietic malignancies, Oncogene, vol.21, issue.21, pp.3459-3474, 2002.
DOI : 10.1200/JCO.1999.17.9.2941

N. Chiorazzi, K. Rai, and M. Ferrarini, Chronic Lymphocytic Leukemia, New England Journal of Medicine, vol.352, issue.8, pp.804-815, 2005.
DOI : 10.1056/NEJMra041720

URL : https://hal.archives-ouvertes.fr/hal-01375537

R. Dorsam and J. Gutkind, G-protein-coupled receptors and cancer, Nature Reviews Cancer, vol.341, issue.2, pp.79-94, 2007.
DOI : 10.4049/jimmunol.172.5.2853

J. Mazella, N. Zsürger, V. Navarro, J. Chabry, M. Kaghad et al., The 100-kDa Neurotensin Receptor Is gp95/Sortilin, A Non-G-Protein-coupled Receptor, Journal of Biological Chemistry, vol.63, issue.41, pp.26273-26276, 1998.
DOI : 10.1074/jbc.273.6.3582

URL : http://www.jbc.org/content/273/41/26273.full.pdf

S. Somaï, A. Gompel, W. Rostène, and P. Forgez, Neurotensin counteracts apoptosis in breast cancer cells, Biochemical and Biophysical Research Communications, vol.295, issue.2, pp.482-488, 2002.
DOI : 10.1016/S0006-291X(02)00703-9

M. Younes, Z. Wu, S. Dupouy, A. Lupo, N. Mourra et al., Neurotensin (NTS) and its receptor (NTSR1) causes EGFR, HER2 and HER3 over-expression and their autocrine/paracrine activation in lung tumors, confirming responsiveness to erlotinib, Oncotarget, vol.5, issue.18, pp.8252-8269, 2014.
DOI : 10.18632/oncotarget.1633

URL : http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=1633&path%5B%5D=4619

S. Dupouy, N. Mourra, V. Doan, A. Gompel, M. Alifano et al., The potential use of the neurotensin high affinity receptor 1 as a biomarker for cancer progression and as a component of personalized medicine in selective cancers, Biochimie, vol.93, issue.9, pp.1369-1378, 2011.
DOI : 10.1016/j.biochi.2011.04.024

S. Saada, P. Marget, A. Fauchais, L. M. Chemin, G. Sindou et al., Differential Expression of Neurotensin and Specific Receptors, NTSR1 and NTSR2, in Normal and Malignant Human B Lymphocytes, The Journal of Immunology, vol.189, issue.11, pp.5293-5303, 2012.
DOI : 10.4049/jimmunol.1102937

URL : https://hal.archives-ouvertes.fr/hal-00872646

S. Bamford, E. Dawson, S. Forbes, J. Clements, R. Pettett et al., The COSMIC (Catalogue of Somatic Mutations in Cancer) database and website, British Journal of Cancer, vol.32, issue.2, pp.355-358, 2004.
DOI : 10.1093/nar/gkh073

J. Frade, A. Rodríguez-tébar, and Y. Barde, Induction of cell death by endogenous nerve growth factor through its p75 receptor, Nature, vol.383, issue.6596, pp.166-168, 1996.
DOI : 10.1038/383166a0

S. Dupouy, V. Doan, Z. Wu, N. Mourra, J. Liu et al., Activation of EGFR, HER2 and HER3 by neurotensin/neurotensin receptor 1 renders breast tumors aggressive yet highly responsive to lapatinib and metformin in mice, Oncotarget, vol.5, issue.18, pp.8235-8251, 2014.
DOI : 10.18632/oncotarget.1632

URL : http://www.impactjournals.com/oncotarget/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=1632&path%5B%5D=3046

C. Wilson, T. Naves, F. Vincent, B. Melloni, F. Bonnaud et al., Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase receptors, Journal of Cell Science, vol.127, issue.18, pp.3983-3997, 2014.
DOI : 10.1242/jcs.149336

URL : https://hal.archives-ouvertes.fr/hal-01059696

C. Wilson, T. Naves, S. Saada, S. Pinet, F. Vincent et al., The Implications of Sortilin/Vps10p Domain Receptors in Neurological and Human Diseases, CNS & Neurological Disorders - Drug Targets, vol.13, issue.8, pp.1354-1365, 2014.
DOI : 10.2174/1871527313666141023151642

R. Dielschneider, X. W. Yoon, J. Noh, E. Banerji, V. Li et al., Gefitinib targets ZAP-70-expressing chronic lymphocytic leukemia cells and inhibits B-cell receptor signaling, Cell Death & Disease, vol.9, issue.10, p.1439, 2014.
DOI : 10.1038/sj.mt.6300184

URL : http://www.nature.com/cddis/journal/v5/n10/pdf/cddis2014391a.pdf

A. Fauchais, F. Lalloué, M. Lise, A. Boumediene, J. Preud-'homme et al., Role of Endogenous Brain-Derived Neurotrophic Factor and Sortilin in B Cell Survival, The Journal of Immunology, vol.181, issue.5, pp.3027-3038, 2008.
DOI : 10.4049/jimmunol.181.5.3027

URL : https://hal.archives-ouvertes.fr/hal-00453223

C. Thiele, Z. Li, and A. Mckee, On Trk--The TrkB Signal Transduction Pathway Is an Increasingly Important Target in Cancer Biology, Clinical Cancer Research, vol.15, issue.19, pp.5962-5967, 2009.
DOI : 10.1158/1078-0432.CCR-08-0651

URL : http://clincancerres.aacrjournals.org/content/clincanres/15/19/5962.full.pdf

R. Pearse, S. Swendeman, Y. Li, D. Rafii, and B. Hempstead, A neurotrophin axis in myeloma: TrkB and BDNF promote tumor-cell survival, Blood, vol.105, issue.11, pp.4429-4436, 2005.
DOI : 10.1182/blood-2004-08-3096

URL : http://www.bloodjournal.org/content/bloodjournal/105/11/4429.full.pdf

L. Ai, C. Sun, Y. Wang, L. Zhang, Z. Chu et al., Gene silencing of the BDNF/TrkB axis in multiple myeloma blocks bone destruction and tumor burden in vitro and in vivo, International Journal of Cancer, vol.96, issue.5, pp.1074-1084, 2013.
DOI : 10.1182/blood-2003-06-1992

S. Martin, J. Vincent, and J. Mazella, Recycling ability of the mouse and the human neurotensin type 2 receptors depends on a single tyrosine residue, J Cell Sci, vol.115, pp.165-173, 2002.

R. 26-raijmakers, K. Kraiczek, A. De-jong, S. Mohammed, and A. Heck, ???Reversed-Phase High-Performance Liquid Chromatography Phosphochip Coupled to a Quadrupole Time-of-Flight Mass Spectrometer, Analytical Chemistry, vol.82, issue.3, pp.824-832, 2010.
DOI : 10.1021/ac901764g

S. Kitada, J. Andersen, S. Akar, J. Zapata, S. Takayama et al., Expression of apoptosis-regulating proteins in chronic lymphocytic leukemia: correlations with In vitro and In vivo chemoresponses, Blood, vol.91, pp.3379-3389, 1998.

P. Longo, L. Laurenti, S. Gobessi, A. Petlickovski, M. Pelosi et al., The Akt signaling pathway determines the different proliferative capacity of chronic lymphocytic leukemia B-cells from patients with progressive and stable disease, Leukemia, vol.72, issue.1, pp.110-120, 2007.
DOI : 10.1182/blood-2002-11-3569

P. Longo, L. Laurenti, S. Gobessi, S. Sica, G. Leone et al., The Akt/Mcl-1 pathway plays a prominent role in mediating antiapoptotic signals downstream of the B-cell receptor in chronic lymphocytic leukemia B cells, Blood, vol.111, issue.2, pp.846-855, 2008.
DOI : 10.1182/blood-2007-05-089037

J. Woyach, A. Johnson, and J. Byrd, The B-cell receptor signaling pathway as a therapeutic target in CLL, Blood, vol.120, issue.6, pp.1175-1184, 2012.
DOI : 10.1182/blood-2012-02-362624

A. Ghosh and N. Kay, Critical Signal Transduction Pathways in CLL, Adv Exp Med Biol, vol.792, pp.215-239, 2013.
DOI : 10.1007/978-1-4614-8051-8_10

URL : http://europepmc.org/articles/pmc3918736?pdf=render

E. Rosati, R. Sabatini, G. Rampino, A. Tabilio, D. Ianni et al., Constitutively activated Notch signaling is involved in survival and apoptosis resistance of B-CLL cells, Blood, vol.113, issue.4, pp.856-865, 2009.
DOI : 10.1182/blood-2008-02-139725

URL : http://www.bloodjournal.org/content/bloodjournal/113/4/856.full.pdf

K. Saiya-cork, R. Collins, B. Parkin, P. Ouillette, E. Kuizon et al., A Pathobiological Role of the Insulin Receptor in Chronic Lymphocytic Leukemia, Clinical Cancer Research, vol.17, issue.9, pp.2679-2692, 2011.
DOI : 10.1158/1078-0432.CCR-10-2058

N. Yaktapour, R. Übelhart, J. Schüler, K. Aumann, C. Dierks et al., Insulin-like growth factor-1 receptor (IGF1R) as a novel target in chronic lymphocytic leukemia, Blood, vol.122, issue.9, pp.1621-1633, 2013.
DOI : 10.1182/blood-2013-02-484386

URL : http://www.bloodjournal.org/content/bloodjournal/122/9/1621.full.pdf

J. Burger, M. Burger, and T. Kipps, Chronic lymphocytic leukemia B cells express functional CXCR4 chemokine receptors that mediate spontaneous migration beneath bone marrow stromal cells, Blood, vol.94, pp.3658-3667, 1999.

R. Möhle, C. Failenschmid, F. Bautz, and L. Kanz, Overexpression of the chemokine receptor CXCR4 in B cell chronic lymphocytic leukemia is associated with increased functional response to stromal cell-derived factor-1 (SDF-1), Leukemia, vol.13, issue.12, pp.1954-1959, 1999.
DOI : 10.1038/sj.leu.2401602

M. Ticchioni, M. Essafi, P. Jeandel, F. Davi, J. Cassuto et al., Homeostatic chemokines increase survival of B-chronic lymphocytic leukemia cells through inactivation of transcription factor FOXO3a, Oncogene, vol.343, issue.50, pp.7081-7091, 2007.
DOI : 10.4049/jimmunol.167.6.3064

P. Schröttner, M. Leick, and M. Burger, The role of chemokines in B cell chronic lymphocytic leukaemia: pathophysiological aspects and clinical impact, Annals of Hematology, vol.81, issue.5, pp.437-446, 2010.
DOI : 10.4049/jimmunol.179.7.4335

A. Drost, G. Seitz, A. Boehmler, M. Funk, K. Norz et al., The G protein-coupled receptor CysLT1 mediates chemokine-like effects and prolongs survival in chronic lymphocytic leukemia, Leukemia & Lymphoma, vol.18, issue.4, pp.665-673, 2012.
DOI : 10.1111/j.1365-2141.2007.06590.x

K. Breitschopf, J. Haendeler, P. Malchow, A. Zeiher, and S. Dimmeler, Posttranslational Modification of Bcl-2 Facilitates Its Proteasome-Dependent Degradation: Molecular Characterization of the Involved Signaling Pathway, Molecular and Cellular Biology, vol.20, issue.5, pp.1886-1896, 2000.
DOI : 10.1128/MCB.20.5.1886-1896.2000

R. Rooswinkel, B. Van-de-kooij, E. De-vries, M. Paauwe, R. Braster et al., Antiapoptotic potency of Bcl-2 proteins primarily relies on their stability, not binding selectivity, Blood, vol.123, issue.18, pp.2806-2815, 2014.
DOI : 10.1182/blood-2013-08-519470

M. Tomasi, M. Ryoo, K. Ramani, I. Tomasi, P. Giordano et al., Methionine adenosyltransferase ?2 sumoylation positively regulate Bcl-2 expression in human colon and liver cancer cells, Oncotarget, vol.6, pp.37706-37723, 2015.
DOI : 10.18632/oncotarget.5342

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=5342&path%5B%5D=14881

Z. Wu, A. Galmiche, J. Liu, N. Stadler, D. Wendum et al., Neurotensin regulation induces overexpression and activation of EGFR in HCC and restores response to erlotinib and sorafenib, Cancer Letters, vol.388, pp.73-84, 2017.
DOI : 10.1016/j.canlet.2016.11.032

M. Magazin, E. Poszepczynska-guigné, M. Bagot, L. Boumsell, C. Pruvost et al., Sezary Syndrome Cells Unlike Normal Circulating T Lymphocytes Fail to Migrate Following Engagement of NT1 Receptor, Journal of Investigative Dermatology, vol.122, issue.1, pp.111-118, 2004.
DOI : 10.1046/j.0022-202X.2003.22131.x

URL : https://doi.org/10.1046/j.0022-202x.2003.22131.x

S. Choi, H. Chae, T. Park, H. Ha, and K. Kim, Characterization of high affinity neurotensin receptor NTR1 in HL-60 cells and its down regulation during granulocytic differentiation, British Journal of Pharmacology, vol.46, issue.4, pp.1050-1056, 1999.
DOI : 10.1139/y89-247

S. Swift, J. Burns, and N. Maitland, Altered Expression of Neurotensin Receptors Is Associated with the Differentiation State of Prostate Cancer, Cancer Research, vol.70, issue.1, pp.347-356, 2010.
DOI : 10.1158/0008-5472.CAN-09-1252

L. Ohman, L. Franzén, U. Rudolph, L. Birnbaumer, and E. Hörnquist, Regression of Peyer's patches in Galphai2 deficient mice prior to colitis is associated with reduced expression of Bcl-2 and increased apoptosis, Gut, vol.51, issue.3, pp.392-397, 2002.
DOI : 10.1136/gut.51.3.392

H. Dalwadi, B. Wei, M. Schrage, K. Spicher, T. Su et al., B Cell Developmental Requirement for the G??i2 Gene, The Journal of Immunology, vol.170, issue.4, pp.1707-1715, 2003.
DOI : 10.4049/jimmunol.170.4.1707

URL : http://www.jimmunol.org/content/jimmunol/170/4/1707.full.pdf

F. Cattaneo, G. Guerra, M. Parisi, D. Marinis, M. Tafuri et al., Cell-Surface Receptors Transactivation Mediated by G Protein-Coupled Receptors, International Journal of Molecular Sciences, vol.47, issue.11, pp.19700-19728, 2014.
DOI : 10.1016/j.advenzreg.2006.12.011

H. El-shewy, K. Johnson, M. Lee, A. Jaffa, L. Obeid et al., Insulin-like Growth Factors Mediate Heterotrimeric G Protein-dependent ERK1/2 Activation by Transactivating Sphingosine 1-Phosphate Receptors, Journal of Biological Chemistry, vol.18, issue.42, pp.31399-31407, 2006.
DOI : 10.1523/JNEUROSCI.2730-05.2006

URL : http://www.jbc.org/content/281/42/31399.full.pdf

N. Delcourt, J. Bockaert, and P. Marin, GPCR-jacking: from a new route in RTK signalling to a new concept in GPCR activation, Trends in Pharmacological Sciences, vol.28, issue.12, pp.602-607, 2007.
DOI : 10.1016/j.tips.2007.09.007

E. Mira, R. Lacalle, M. González, C. Gómez-moutón, J. Abad et al., A role for chemokine receptor transactivation in growth factor signaling, EMBO reports, vol.163, issue.2, pp.151-156, 2001.
DOI : 10.1016/0167-0115(95)00159-X

URL : http://embor.embopress.org/content/embor/2/2/151.full.pdf

J. Hobson, H. Rosenfeldt, L. Barak, A. Olivera, S. Poulton et al., Role of the Sphingosine-1-Phosphate Receptor EDG-1 in PDGF-Induced Cell Motility, Science, vol.291, issue.5509, pp.1800-1803, 2001.
DOI : 10.1126/science.1057559

N. Moughal, C. Waters, B. Sambi, S. Pyne, and N. Pyne, Nerve growth factor signaling involves interaction between the Trk A receptor and lysophosphatidate receptor 1 systems: nuclear translocation of the lysophosphatidate receptor 1 and Trk A receptors in pheochromocytoma 12 cells, Cellular Signalling, vol.16, issue.1, pp.127-136, 2004.
DOI : 10.1016/j.cellsig.2003.08.004

N. Moughal, C. Waters, W. Valentine, M. Connell, J. Richardson et al., Protean agonism of the lysophosphatidic acid receptor-1 with Ki16425 reduces nerve growth factor-induced neurite outgrowth in pheochromocytoma 12 cells, Journal of Neurochemistry, vol.392, issue.6, pp.1920-1929, 2006.
DOI : 10.1016/S1388-1981(02)00330-X

N. Vita, F. Oury-donat, P. Chalon, M. Guillemot, M. Kaghad et al., Neurotensin is an antagonist of the human neurotensin NT2 receptor expressed in Chinese hamster ovary cells, European Journal of Pharmacology, vol.360, issue.2-3, pp.265-272, 1998.
DOI : 10.1016/S0014-2999(98)00678-5

A. Bürkle, M. Niedermeier, A. Schmitt-gräff, W. Wierda, M. Keating et al., Overexpression of the CXCR5 chemokine receptor, and its ligand, CXCL13 in B-cell chronic lymphocytic leukemia, Blood, vol.110, issue.9, pp.3316-3325, 2007.
DOI : 10.1182/blood-2007-05-089409

M. De-la-cruz-morcillo, J. Berger, R. Sánchez-prieto, S. Saada, T. Naves et al., p75 neurotrophin receptor and pro-BDNF promote cell survival and migration in clear cell renal cell carcinoma, Oncotarget, vol.7, issue.23, pp.75-34480, 2016.
DOI : 10.18632/oncotarget.8911

C. Chu, Z. She, X. Zhang, L. Chen, L. Ai et al., Brain-derived neurotrophic factor is a potential osteoclast stimulating factor in multiple myeloma, Int J Cancer, vol.130, pp.827-836, 2012.

M. Cazorla, J. Prémont, A. Mann, N. Girard, C. Kellendonk et al., Identification of a low???molecular weight TrkB antagonist with anxiolytic and antidepressant activity in mice, Journal of Clinical Investigation, vol.121, issue.5, pp.1846-1857, 2011.
DOI : 10.1172/JCI43992DS1

W. Strober, Trypan Blue Exclusion Test of Cell Viability, Curr Protoc Immunol A3.B, vol.111, pp.1-3, 2015.
DOI : 10.1002/0471142735.ima03bs111