H. Akil, A. Perraud, C. Mélin, M. O. Jauberteau, and M. Mathonnet, Fine-tuning roles of endogenous brain-derived neurotrophic factor, TrkB and sortilin in colorectal cancer cell survival, PLoS One, vol.6, p.25097, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00873136

A. A. Alizadeh, M. B. Eisen, R. E. Davis, C. Ma, I. S. Lossos et al., Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling, Nature, vol.403, pp.503-511, 2000.

E. Arranz, M. Robledo, B. Martínez, J. Gallego, A. Román et al., Incidence of homogeneously staining regions in non-Hodgkin lymphomas, Cancer Genet Cytogenet, vol.87, p.1, 1996.

A. S. Baldwin, Control of oncogenesis and cancer therapy resistance by the transcription factor NF-kappaB, J Clin Invest, vol.107, pp.241-246, 2001.

P. A. Barker, High Affinity Not in the Vicinity?, Neuron, vol.53, pp.1-4, 2007.

C. Bellanger, L. Dubanet, M. C. Lise, A. L. Fauchais, D. Bordessoule et al., Endogenous neurotrophins and Trk signalling in diffuse large B cell lymphoma cell lines are involved in sensitivity to rituximab-induced apoptosis, PLoS One, vol.6, p.27213, 2011.

G. V. Chaitanya, A. J. Steven, and P. P. Babu, PARP-1 cleavage fragments: signatures of cell-death proteases in neurodegeneration, Cell Commun Signal, vol.8, p.31, 2010.

R. E. Davis, K. D. Brown, U. Siebenlist, and L. M. Staudt, Constitutive nuclear factor B ? B activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells, J Exp Med, vol.194, pp.1861-1874, 2001.

D. Santi, L. Annunziata, P. Sessa, E. Bramanti, and P. , Brain-derived neurotrophic factor and TrkB receptor in experimental autoimmune encephalomyelitis and multiple sclerosis, J Neurol Sci, vol.287, pp.17-26, 2009.

A. L. Fauchais, F. Lalloué, M. C. Lise, A. Boumediene, J. L. Preud'homme et al., Role of endogenous brain-derived neurotrophic factor and sortilin in B cell survival, J Immunol, vol.181, pp.3027-3038, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00453223

Q. Qiao, Y. Nozaki, K. Sakoe, N. Komatsu, and K. Kirito, NF-?B mediates aberrant activation of HIF-1 in malignant lymphoma, Exp Hematol, vol.38, pp.1199-1208, 2010.

B. Reichardt, Neurotrophin-regulated signalling pathways, Philos Trans R Soc Lond B Biol Sci, vol.361, pp.1545-1564, 2006.

A. R. Rezvani and D. G. Maloney, Rituximab resistance, Best Pract Res Clin Haematol, vol.24, pp.203-216, 2011.

A. Rosenwald, G. Wright, W. C. Chan, J. M. Connors, E. Campo et al.,

H. , K. P. Stokke, T. Staudt, and L. M. , The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. Lymphoma/Leukemia Molecular Profiling Project, N Engl J Med, vol.346, pp.1937-1947, 2002.

J. F. Seymour, M. Pfreundschuh, M. Trn?ný, L. H. Sehn, J. Catalano et al., R-CHOP with or without bevacizumab in patients with previously untreated diffuse large B-cell lymphoma: final MAIN study outcomes, Haematologica, vol.99, pp.1343-1349, 2014.

L. F. Sniderhan, T. M. Garcia-bates, M. Burgart, S. H. Bernstein, R. P. Phipps et al., Neurotrophin signalling through tropomyosin receptor kinases contributes to survival and proliferation of non-Hodgkin lymphoma, Exp Hematol, vol.37, pp.1295-1309, 2009.

E. Suzuki, K. Umezawa, and B. Bonavida, Rituximab inhibits the constitutively activated PI3KAkt pathway in B-NHL cell lines: involvement in chemosensitization to drug-induced apoptosis, Oncogene, vol.26, pp.6184-6193, 2007.

S. H. Swerdlow, E. Campo, N. L. Harris, E. S. Jaffe, S. A. Pileri et al., WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 2008.

K. K. Teng, S. Felice, T. Kim, and B. L. Hempstead, Understanding proneurotrophin actions: Recent advances and challenges, Dev Neurobiol, vol.70, pp.350-359, 2010.

C. J. Thiele, Z. Li, and A. E. Mckee, On Trk-the TrkB signal transduction pathway is an increasingly important target in cancer biology, Clin Cancer Res, vol.15, pp.5962-5967, 2009.

D. Troutaud, B. Petit, C. Bellanger, B. Marin, M. P. Gourin-chaury et al., Prognostic significance of BAD and AIF apoptotic pathways in diffuse large B-cell lymphoma, Clin Lymphoma Myeloma Leuk, vol.10, issue.3, pp.118-124, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00597660

E. Vanhecke, E. Adriaenssens, S. Verbeke, S. Meignan, E. Germain et al., , 2011.

, are expressed in breast cancer and can be targeted to inhibit tumor cell survival, Clin Cancer Res, vol.17, pp.1741-1752

J. A. Vega, O. García-suárez, J. Hannestad, M. Pérez-pérez, and A. Germanà, Neurotrophins and the immune system, J Anat, vol.203, pp.1-19, 2003.

Y. Wang, D. Fei, M. Vanderlaan, and A. Song, Biological activity of bevacizumab, a humanized anti-VEGF antibody in vitro, Angiogenesis, vol.7, pp.335-345, 2004.

T. Wehrman, X. He, B. Raab, A. Dukipatti, H. Blau et al., Structural and mechanistic insights into nerve growth factor interactions with the TrkA and p75 receptors, Neuron, vol.53, pp.25-38, 2007.

G. J. Weiner, Rituximab: mechanism of action, Semin Hematol, vol.47, pp.115-123, 2010.

G. Wright, B. Tan, A. Rosenwald, E. H. Hurt, A. Wiestner et al., A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma, Proc Natl Acad Sci, vol.100, pp.9991-9996, 2003.

R. Alizadeh, A. A. Eisen, M. B. Davis, and R. E. , Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling, Nature, vol.403, pp.503-511, 2000.
DOI : 10.1038/35000501

G. Andreola, L. Rivoltini, and C. Castelli, Induction of lymphocyte apoptosis by tumor cell secretion of FasL-bearing microvesicles, J Exp Med, vol.195, pp.1303-1319, 2002.

J. C. Arévalo and S. H. Wu, Neurotrophin Signaling: Many Exciting Surprises!, vol.63, pp.1523-1560, 2006.

T. Aung, B. Chapuy, and D. Vogel, Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3, Proc Natl Acad Sci, vol.108, issue.37, pp.15336-15377, 2011.

C. Bellanger, L. Dubanet, and M. C. Lise, Endogenous neurotrophins and Trk signalling in diffuse large B cell lymphoma cell lines are involved in sensitivity to rituximab-induced apoptosis, PLoS One, vol.6, p.27213, 2011.
DOI : 10.1371/journal.pone.0027213

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0027213&type=printable

A. Bobrie and C. Théry, Exosomes and communication between tumours and the immune system: are all exosomes equal?, Biochem Soc Trans, vol.41, issue.1, pp.263-270, 2013.
DOI : 10.1042/bst20120245

B. Bonavida, Postulated mechanisms of resistance of B-cell non-Hodgkin lymphoma to rituximab treatment regimens: strategies to overcome resistance, Semin Oncol, vol.41, issue.5, pp.667-77, 2014.

B. Coiffier, Rituximab therapy in malignant lymphoma, Oncogene, vol.26, issue.25, pp.3603-3616, 2007.
DOI : 10.1038/sj.onc.1210376

URL : https://www.nature.com/articles/1210376.pdf

M. S. Czuczman, S. Olejniczak, and A. Gowda, Acquirement of rituximab resistance in lymphoma cell lines is associated with both global CD20 gene and protein downregulation regulated at the pretranscriptional and posttranscriptional levels, Clin. Cancer Res, vol.14, issue.5, pp.1561-70, 2008.

R. E. Davis, K. D. Brown, U. Siebenlist, and L. M. Staudt, Constitutive nuclear factor B ? B activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells, J Exp Med, vol.194, pp.1861-74, 2001.

L. Dubanet, H. Bentayeb, and B. Petit, Anti-apoptotic role and clinical relevance of neurotrophins in diffuse large B-cell lymphomas, Br J Cancer, vol.113, issue.6, pp.934-978, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01819218

C. Gutzeit, N. Nagy, and M. Gentile, Exosomes derived from Burkitt's lymphoma cell lines induce proliferation, differentiation, and class-switch recombination in B cells, J Immunol, vol.192, issue.12, pp.5852-62, 2014.
DOI : 10.4049/jimmunol.1302068

URL : http://www.jimmunol.org/content/192/12/5852.full.pdf

B. N. Hannafon and W. Q. Ding, Intercellular communication by exosome-derived microRNAs in cancer, Int J Mol Sci, vol.14, issue.7, pp.14240-69, 2013.
DOI : 10.3390/ijms140714240

URL : https://www.mdpi.com/1422-0067/14/7/14240/pdf

J. Hiraga, A. Tomita, and . Sugimoto, Down-regulation of CD20 expression in B-cell lymphoma cells after treatment with rituximab-containing combination chemotherapies: its prevalence and clinical significance, Blood, vol.113, pp.4885-93, 2009.

S. W. Jang, X. Liu, and M. Yepes, A selective TrkB agonist with potent neurotrophic activities by 7,8-dihydroxyflavone, Proc Natl Acad Sci U S A, vol.107, issue.6, pp.2687-92, 2010.
DOI : 10.1073/pnas.0913572107

URL : http://www.pnas.org/content/107/6/2687.full.pdf

N. A. Johnson, M. Boyle, and A. Bashashati, Diffuse large B-cell lymphoma: reduced CD20 expression is associated with an inferior survival, Blood, vol.113, issue.16, pp.3773-80, 2009.
DOI : 10.1182/blood-2008-09-177469

URL : http://www.bloodjournal.org/content/113/16/3773.full.pdf

C. Kahlert and R. Kalluri, Exosomes in tumor microenvironment influence cancer progression and metastasis, J Mol Med, vol.91, issue.4, pp.431-438, 2013.
DOI : 10.1007/s00109-013-1020-6

URL : http://europepmc.org/articles/pmc4073669?pdf=render

R. Koch, T. Aung, D. Vogel, and B. Chapuy, Nuclear Trapping through Inhibition of Exosomal Export by Indomethacin Increases Cytostatic Efficacy of Doxorubicin and Pixantrone, Clin Cancer Res, vol.22, issue.2, pp.395-404, 2016.

L. T. Lam, R. E. Davis, and J. Pierce, Small molecule inhibitors of I?B kinase are selectively toxic for subgroups of diffuse large B-cell lymphoma defined by gene expression profiling, Clin Cancer Res, vol.11, pp.28-40, 2005.

C. Liu, S. Yu, and K. Zinn, Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function, J Immunol, vol.176, pp.1375-85, 2006.
DOI : 10.4049/jimmunol.176.3.1375

URL : http://www.jimmunol.org/content/176/3/1375.full.pdf

G. Lenz and L. M. Staudt, Aggressive lymphomas, N Engl J Med, vol.362, issue.15, pp.1417-1446, 2010.

H. Miyoshi, F. Arakawa, and K. Sato, Comparison of CD20 expression in B-cell lymphoma between newly diagnosed, untreated cases and those after rituximab treatment, Cancer Sci, vol.103, issue.8, pp.1567-73, 2012.

S. H. Olejniczak, C. C. Stewart, K. Donohue, and M. S. Czuczman, A quantitative exploration of surface antigen expression in common B-cell malignancies using flow cytometry, Immunol Invest, vol.35, pp.93-114, 2006.

G. Raposo, H. W. Nijman, and . Stoorvogelw, B lymphocytes secrete antigen-presenting vesicles, J Exp Med, vol.183, pp.1161-72, 1996.

A. R. Rezvani and D. G. Maloney, Rituximab resistance, Best Pract Res Clin Haematol, vol.24, pp.203-219, 2011.

Y. Suzuki, T. Yoshida, and G. Wang, Association of CD20 levels with clinicopathological parameters and its prognostic significance for patients with DLBCL, Ann Hematol, vol.91, issue.7, pp.997-1002, 2012.

C. Théry, S. Amigorena, G. Raposo, and A. Clayton, Isolation and characterization of exosomes from cell culture supernatants and biological fluids, Curr Protoc Cell Biol, 2006.

M. Winiarska, K. Bojarczuk, and B. Pyrzynska, Inhibitors of SRC kinases impair antitumor activity of anti-CD20 monoclonal antibodies, MAbs, vol.6, issue.5, pp.1300-1313, 2014.

W. Wojciechowski, H. Li, and S. Marshall, Enhanced expression of CD20 in human tumor B cells is controlled through ERK-dependent mechanisms, J Immunol, vol.174, issue.12, pp.7859-68, 2005.

G. Wright, B. Tan, and A. Rosenwald, A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma, Proc Natl Acad Sci, vol.100, pp.9991-9997, 2003.

X. Zhang, X. Yuan, and H. Shi, Exosomes in cancer: small particle, big player, J Hematol Oncol, vol.8, p.83, 2015.

, Article 3: Evidence for the biological and therapeutic relevance of the prohibitin and EIF4A ligand, FL3, in non­Hodgkin B cell lymphoma

, Marie-Odile Jauberteau, vol.7

, Running title: anticancer mechanisms of flavaglines in DLBCL Funding: This research was supported by Cancéropôle GSO (Grand Sud Ouest) and the Ligue Contre le Cancer, 5 Beaujon University Hospital (AP-HP-PRES Paris 7 Diderot), 100 boulevard du Général Leclerc, vol.7200, pp.435-967

E. , danielle.troutaud@unilim.fr Text: 4054 words-Abstract: 240 words Number of Figures: 7 (and 1 supplementary figure, p.52

L. H. Sehn and R. D. Gascoyne, Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity, Blood, vol.125, issue.1, pp.22-32, 2015.

L. Pasqualucci, V. Trifonov, and G. Fabbri, Analysis of the coding genome of diffuse large B-cell lymphoma, Nat Genet, vol.43, issue.9, pp.830-837, 2011.

E. Horvilleur, T. Sbarrato, and K. Hill, A role for eukaryotic initiation factor 4B overexpression in the pathogenesis of diffuse large B-cell lymphoma, Leukemia, vol.28, issue.5, pp.1092-1102, 2014.

D. Gratzinger, S. Zhao, R. J. Tibshirani, and E. D. Hsi, Prognostic significance of VEGF, VEGF receptors, and microvessel density in diffuse large B cell lymphoma treated with anthracycline-based chemotherapy, Lab Invest, vol.88, pp.38-47, 2008.

P. R. Hagner, A. Schneider, and R. B. Gartenhaus, Targeting the translational machinery as a novel treatment strategy for hematologic malignancies, Blood, vol.115, issue.11, pp.2127-2135, 2010.

R. Vaidya and T. E. Witzig, Prognostic factors for diffuse large B-cell lymphoma in the R(X)CHOP era, Ann Oncol, vol.25, issue.11, pp.2124-2133, 2014.

L. H. Sehn, Paramount prognostic factors that guide therapeutic strategies in diffuse large Bcell lymphoma, Hematology Am Soc Hematol Educ Program, vol.2012, pp.402-409

B. Coiffier, Rituximab therapy in malignant lymphoma, Oncogene, vol.26, issue.25, pp.3603-3613, 2007.

M. Pfreundschuh, How I treat elderly patients with diffuse large B-cell lymphoma, Blood, vol.116, issue.24, pp.5103-5110, 2010.

C. Basmadjian, F. Thuaud, N. Ribeiro, and L. Désaubry, Flavaglines: potent anticancer drugs that target prohibitins and the helicase eIF4A, Future Med Chem, vol.5, pp.2185-2197, 2013.

B. Hausott, H. Greger, and M. B. , Flavaglines: a group of efficient growth inhibitors block cell cycle progression and induce apoptosis in colorectal cancer cells, Int J Cancer, vol.109, issue.6, pp.933-940, 2004.

J. Y. Zhu, I. N. Lavrik, and U. Mahlknecht, The traditional Chinese herbal compound rocaglamide preferentially induces apoptosis in leukemia cells by modulation of mitogenactivated protein kinase activities, Int J Cancer, vol.121, pp.1839-1846, 2007.

D. M. Lucas, R. B. Edwards, and G. Lozanski, The novel plant-derived agent silvestrol has Bcell selective activity in chronic lymphocytic leukemia and acute lymphoblastic leukemia in vitro and in vivo, Blood, vol.113, pp.4656-4666, 2009.

N. Ribeiro, F. Thuaud, C. Nebigil, and L. Désaubry, Recent advances in the biology and chemistry of the flavaglines, Bioorg Med Chem, vol.20, issue.6, pp.1857-1864, 2012.

M. E. Bordeleau, F. Robert, and B. Gerard, Therapeutic suppression of translation initiation modulates chemosensitivity in a mouse lymphoma model, J Clin Invest, vol.118, issue.7, pp.2651-2660, 2008.

G. Polier, J. Neumann, and F. Thuaud, The natural anticancer compounds rocaglamides inhibit the Raf-MEK-ERK pathway by targeting prohibitin 1 and 2, Chem Biol, vol.19, issue.9, pp.1093-1104, 2012.

J. M. Chambers, L. M. Lindqvist, and A. Webb, Synthesis of biotinylated episilvestrol: highly selective targeting of the translation factors eIF4AI/II, Org Lett, vol.15, issue.6, pp.1406-1409, 2013.

M. Li-weber, Molecular mechanisms and anti-cancer aspects of the medicinal phytochemicals rocaglamides (=flavaglines), Int J Cancer, vol.137, issue.8, pp.1791-1799, 2015.

M. Bhat, N. Robichaud, and L. Hulea, Targeting the translation machinery in cancer, Nat Rev Drug Discov, vol.14, issue.4, pp.261-278, 2015.

S. Koushyar, W. G. Jiang, and D. A. Dart, Unveiling the potential of prohibitin in cancer, Cancer Lett, vol.369, issue.2, pp.316-322, 2015.

R. Cencic, M. Carrier, and G. Galicia-vázquez, Antitumor activity and mechanism of action of the cyclopenta[b]benzofuran, silvestrol, PLoS One, vol.4, issue.4, p.5223, 2009.

K. Rajalingam and T. Rudel, Ras-Raf signaling needs prohibitin, Cell Cycle, vol.4, issue.11, pp.1503-1505, 2005.

Y. T. Peng, P. Chen, R. Y. Ouyang, and L. Song, Multifaceted role of prohibitin in cell survival and apoptosis, Apoptosis, vol.20, issue.9, pp.1135-1149, 2015.

L. Dubanet, H. Bentayeb, and B. Petit, Anti-apoptotic role and clinical relevance of neurotrophins in diffuse large B-cell lymphomas, Br J Cancer, vol.113, issue.6, pp.934-944, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01819218

S. H. Swerdlow, E. Campo, and N. L. Harris, WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th edn, 2008.

C. P. Hans, D. D. Weisenburger, and T. C. Greiner, Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray, Blood, vol.103, pp.275-282, 2004.

F. Thuaud, Y. Bernard, and G. Türkeri, Synthetic analogue of rocaglaol displays a potent and selective cytotoxicity in cancer cells: involvement of apoptosis inducing factor and caspase-12, J Med Chem, vol.52, issue.16, pp.5176-5187, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00467437

C. Bellanger, L. Dubanet, and M. C. Lise, Endogenous neurotrophins and Trk signaling in diffuse large B cell lymphoma cell lines are involved in sensitivity to rituximab-induced apoptosis, PLoS One, vol.6, issue.11, p.27213, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00873135

D. Troutaud, B. Petit, and C. Bellanger, Prognostic significance of Bad and AIF apoptotic pathways in diffuse large B-cell lymphoma, Clin. Lymphoma Myeloma Leuk, vol.10, issue.2, pp.118-124, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00597660

L. Boussemart, H. Malka-mahieu, and I. Girault, eIF4F is a nexus of resistance to antiBRAF and anti-MEK cancer therapies, Nature, vol.513, issue.7516, pp.105-109, 2014.

R. E. Davis, K. D. Brown, U. Siebenlist, and L. M. Staudt, Constitutive nuclear factor B ? B activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells, J Exp Med, vol.194, pp.1861-1874, 2001.

L. T. Lam, R. E. Davis, and J. Pierce, Small molecule inhibitors of I?B kinase are selectively toxic for subgroups of diffuse large B-cell lymphoma defined by gene expression profiling, Clin Cancer Res, vol.11, pp.28-40, 2005.

B. Baumann, F. Bohnenstengel, and D. Siegmund, Rocaglamide derivatives are potent inhibitors of NF-kappa B activation in T-cells, J. Biol. Chem, vol.277, pp.44791-44800, 2002.

J. C. Reed and M. Pellecchia, Apoptosis-based therapies for hematologic malignancies, Blood, vol.106, pp.408-418, 2005.

L. Delavallée, L. Cabon, and P. Galán-malo, AIF-mediated caspase-independent necroptosis: a new chance for targeted therapeutics, IUBMB Life, vol.63, issue.4, pp.221-232, 2011.

R. Cencic, F. Robert, and G. Galicia-vázquez, Modifying chemotherapy response by targeted inhibition of eukaryotic initiation factor 4A, Blood Cancer J, vol.3, p.128, 2013.

A. L. Wolfe, K. Singh, and Y. Zhong, RNA G-quadruplexes cause eIF4A-dependent oncogene translation in cancer, Nature, vol.513, issue.7516, pp.65-70, 2014.

C. J. Lin, Z. Nasr, and P. K. Premsrirut, Targeting synthetic lethal interactions between Myc and the eIF4F complex impedes tumorigenesis, Cell Rep, vol.1, issue.4, pp.325-333, 2012.

S. Iwasaki, S. N. Floor, and N. T. Ingolia, Rocaglates convert DEAD-box protein eIF4A into a sequence-selective translational repressor, Nature, 2016.

J. J. Steinhardt, R. J. Peroutka, and K. Mazan-mamczarz, Inhibiting CARD11 translation during BCR activation by targeting the eIF4A RNA helicase, Blood, vol.124, issue.25, pp.3758-3767, 2014.

A. L. Landon, P. A. Muniandy, and A. C. Shetty, MNKs act as a regulatory switch for eIF4E1 and eIF4E3 driven mRNA translation in DLBCL, Nat Commun, vol.5, p.5413, 2014.

A. R. Jazirehi, M. I. Vega, and D. Chatterjee, Inhibition of the Raf-MEK1/2-ERK1/2 signaling pathway, Bcl-xL down-regulation, and chemosensitization of nonHodgkin's lymphoma B cells by Rituximab, Cancer Res, vol.64, pp.7117-7126, 2004.

F. A. Scholl, P. A. Dumesic, and P. A. Khavari, Effects of active MEK1 expression in vivo, Cancer Lett, vol.230, issue.1, pp.1-5, 2005.

B. Dai, X. F. Zhao, and P. Hagner, Extracellular signal-regulated kinase positively regulates the oncogenic activity of MCT-1 in diffuse large B-cell lymphoma, Cancer Res, vol.69, pp.7835-7843, 2009.

S. Bhalla, A. M. Evens, and B. Dai, The novel anti-MEK small molecule AZD6244 induces BIM-dependent and AKT-independent apoptosis in diffuse large B-cell lymphoma, Blood, vol.118, issue.4, pp.1052-1061, 2011.

C. G. Proud, Mnks, eIF4E phosphorylation and cancer, Biochim Biophys Acta, vol.1849, issue.7, pp.766-773, 2015.

H. G. Wendel, R. L. Silva, and A. Malina, Dissecting eIF4E action in tumorigenesis, Genes Dev, vol.21, issue.24, pp.3232-3237, 2007.

O. Riesterer, D. Zingg, and J. Hummerjohann, Degradation of PKB/Akt protein by inhibition of the VEGF receptor/mTOR pathway in endothelial cells, Oncogene, vol.23, issue.26, pp.4624-4635, 2004.

L. G. Nijtmans, S. M. Artal, L. A. Grivell, and P. J. Coates, The mitochondrial PHB complex: roles in mitochondrial respiratory complex assembly, ageing and degenerative disease, Cell Mol Life Sci, vol.59, pp.143-155, 2002.

W. Song, L. Tian, and S. S. Li, The aberrant expression and localization of prohibitin during apoptosis of human cholangiocarcinoma Mz-ChA-1 cells, FEBS Lett, vol.588, issue.3, pp.422-428, 2014.

L. Jiang, P. Dong, and Z. Zhang, Akt phosphorylates Prohibitin 1 to mediate its mitochondrial localization and promote proliferation of bladder cancer cells, Cell Death Dis, vol.6, p.1660, 2015.

A. L. Theiss and S. V. Sitaraman, The role and therapeutic potential of prohibitin in disease, Biochim Biophys Acta, vol.1813, issue.6, pp.1137-1143, 2011.

, REFERENCES BIBLIOGRAPHIQUES

C. Admyre, S. M. Johansson, K. R. Qazi, J. Filén, R. Lahesmaa et al., Exosomes with immune modulatory features are present in human breast milk, J. Immunol. Baltim. Md, vol.179, pp.1969-1978, 1950.

H. Akil, A. Perraud, C. Mélin, M. Jauberteau, and M. Mathonnet, Fine-tuning roles of endogenous brain-derived neurotrophic factor, TrkB and sortilin in colorectal cancer cell survival, PloS One, vol.6, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00873136

R. F. Alderson, R. Curtis, A. L. Alterman, R. M. Lindsay, and P. S. Distefano, Truncated TrkB mediates the endocytosis and release of BDNF and neurotrophin-4/5 by rat astrocytes and schwann cells in vitro, Brain Res, vol.871, pp.210-222, 2000.

D. D. Alexander, P. J. Mink, H. Adami, E. T. Chang, P. Cole et al., The non-Hodgkin lymphomas: a review of the epidemiologic literature, Int. J. Cancer, vol.120, pp.1-39, 2007.

A. A. Alizadeh, M. B. Eisen, R. E. Davis, C. Ma, I. S. Lossos et al., Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling, Nature, vol.403, pp.503-511, 2000.

A. Anastasia, P. A. Barker, M. V. Chao, and B. L. Hempstead, Detection of p75NTR Trimers: Implications for Receptor Stoichiometry and Activation, J. Neurosci. Off. J. Soc. Neurosci, vol.35, pp.11911-11920, 2015.

E. Ardini, M. Menichincheri, P. Banfi, R. Bosotti, C. De-ponti et al., Entrectinib, a Pan-TRK, ROS1, and ALK Inhibitor with Activity in Multiple Molecularly Defined Cancer Indications, Mol. Cancer Ther, vol.15, pp.628-639, 2016.

J. C. Arévalo and S. H. Wu, Neurotrophin signaling: many exciting surprises!, Cell. Mol. Life Sci. CMLS, vol.63, pp.1523-1537, 2006.

J. C. Arévalo, H. Yano, K. K. Teng, and M. V. Chao, A unique pathway for sustained neurotrophin signaling through an ankyrin-rich membrane-spanning protein, EMBO J, vol.23, pp.2358-2368, 2004.

J. K. Atwal, B. Massie, F. D. Miller, and D. R. Kaplan, The TrkB-Shc site signals neuronal survival and local axon growth via MEK and P13-kinase, Neuron, vol.27, pp.265-277, 2000.

T. Aung, B. Chapuy, D. Vogel, D. Wenzel, M. Oppermann et al., Exosomal evasion of humoral immunotherapy in aggressive B-cell lymphoma modulated by ATP-binding cassette transporter A3, Proc. Natl. Acad. Sci, vol.108, pp.15336-15341, 2011.

D. W. Banner, A. Arcy, W. Janes, R. Gentz, H. J. Schoenfeld et al., Crystal structure of the soluble human 55 kd TNF receptor-human TNF beta complex: implications for TNF receptor activation, Cell, vol.73, pp.431-445, 1993.

A. A. Bapat, R. M. Munoz, V. Hoff, D. D. , H. et al., Blocking Nerve Growth Factor Signaling Reduces the Neural Invasion Potential of Pancreatic Cancer Cells, PloS One, vol.11, 2016.

Y. A. Barde, D. Edgar, and H. Thoenen, Purification of a new neurotrophic factor from mammalian brain, EMBO J, vol.1, pp.549-553, 1982.

P. A. Barker, High affinity not in the vicinity?, Neuron, vol.53, pp.1-4, 2007.

P. A. Barker and E. M. Shooter, Disruption of NGF binding to the low affinity neurotrophin receptor p75LNTR reduces NGF binding to TrkA on PC12 cells, Neuron, vol.13, pp.203-215, 1994.

P. A. Barker, F. D. Miller, T. H. Large, M. , and R. A. , Generation of the truncated form of the nerve growth factor receptor by rat Schwann cells. Evidence for post-translational processing, J. Biol. Chem, vol.266, pp.19113-19119, 1991.

P. A. Barker, C. Lomen-hoerth, E. M. Gensch, S. O. Meakin, D. J. Glass et al., , 1993.

, Tissue-specific alternative splicing generates two isoforms of the trkA receptor, J. Biol. Chem, vol.268, pp.15150-15157

C. Basmadjian, F. Thuaud, N. Ribeiro, and L. Désaubry, Flavaglines: potent anticancer drugs that target prohibitins and the helicase eIF4A, Future Med. Chem, vol.5, pp.2185-2197, 2013.

C. E. Bazenet, M. A. Mota, R. , and L. L. , The small GTP-binding protein Cdc42 is required for nerve growth factor withdrawal-induced neuronal death, Proc. Natl. Acad. Sci. U. S. A, vol.95, pp.3984-3989, 1998.

E. B. Becker, J. Howell, Y. Kodama, P. A. Barker, B. et al., Characterization of the cJun N-terminal kinase-BimEL signaling pathway in neuronal apoptosis, J. Neurosci. Off. J. Soc. Neurosci, vol.24, pp.8762-8770, 2004.

S. A. Beers, C. H. Chan, R. R. French, M. S. Cragg, G. et al., CD20 as a target for therapeutic type I and II monoclonal antibodies, Semin. Hematol, vol.47, pp.107-114, 2010.

W. Béguelin, R. Popovic, M. Teater, Y. Jiang, K. L. Bunting et al., EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation, Cancer Cell, vol.23, pp.677-692, 2013.

W. Béguelin, S. Sawh, N. Chambwe, F. C. Chan, Y. Jiang et al., IL10 receptor is a novel therapeutic target in DLBCLs. Leukemia, vol.29, pp.1684-1694, 2015.

C. Bellanger, L. Dubanet, M. Lise, A. Fauchais, D. Bordessoule et al., Endogenous neurotrophins and Trk signaling in diffuse large B cell lymphoma cell lines are involved in sensitivity to rituximab-induced apoptosis, PloS One, vol.6, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00873135

B. Bellosillo, N. Villamor, A. López-guillermo, S. Marcé, J. Esteve et al., Complement-mediated cell death induced by rituximab in B-cell lymphoproliferative disorders is mediated in vitro by a caspase-independent mechanism involving the generation of reactive oxygen species, Blood, vol.98, pp.2771-2777, 2001.

B. Berninger, D. E. García, N. Inagaki, C. Hahnel, and D. Lindholm, BDNF and NT-3 induce intracellular Ca2+ elevation in hippocampal neurones, Neuroreport, vol.4, pp.1303-1306, 1993.

A. L. Bhakar, J. L. Howell, C. E. Paul, A. H. Salehi, E. B. Becker et al., Apoptosis induced by p75NTR overexpression requires Jun kinase-dependent phosphorylation of Bad, J. Neurosci. Off. J. Soc. Neurosci, vol.23, pp.11373-11381, 2003.

J. Bil, M. Winiarska, D. Nowis, K. Bojarczuk, A. Dabrowska-iwanicka et al., Bortezomib modulates surface CD20 in B-cell malignancies and affects rituximab-mediated complement-dependent cytotoxicity, Blood, vol.115, pp.3745-3755, 2010.

T. R. Bilderback, V. R. Gazula, M. P. Lisanti, and R. T. Dobrowsky, Caveolin interacts with Trk A and p75(NTR) and regulates neurotrophin signaling pathways, J. Biol. Chem, vol.274, pp.257-263, 1999.

A. Bobrie and C. Théry, Exosomes and communication between tumours and the immune system: are all exosomes equal?, Biochem. Soc. Trans, vol.41, pp.263-267, 2013.

B. Bonavida, Postulated mechanisms of resistance of B-cell non-Hodgkin lymphoma to rituximab treatment regimens: strategies to overcome resistance, Semin. Oncol, vol.41, pp.667-677, 2014.

M. Bordeleau, F. Robert, B. Gerard, L. Lindqvist, S. M. Chen et al., Therapeutic suppression of translation initiation modulates chemosensitivity in a mouse lymphoma model, J. Clin. Invest, vol.118, pp.2651-2660, 2008.

L. Boussemart, H. Malka-mahieu, I. Girault, D. Allard, O. Hemmingsson et al., eIF4F is a nexus of resistance to antiBRAF and anti-MEK cancer therapies, Nature, vol.513, pp.105-109, 2014.
DOI : 10.1038/nature13572

A. B. Brann, M. Tcherpakov, I. M. Williams, A. H. Futerman, and M. Fainzilber, Nerve growth factor-induced p75-mediated death of cultured hippocampal neurons is age-dependent and transduced through ceramide generated by neutral sphingomyelinase, J. Biol. Chem, vol.277, pp.9812-9818, 2002.
DOI : 10.1074/jbc.m109862200

URL : http://www.jbc.org/content/277/12/9812.full.pdf

P. A. Bresnahan, R. Leduc, L. Thomas, J. Thorner, H. L. Gibson et al., Human fur gene encodes a yeast KEX2-like endoprotease that cleaves pro-beta-NGF in vivo, J. Cell Biol, vol.111, pp.2851-2859, 1990.
DOI : 10.1083/jcb.111.6.2851

URL : http://jcb.rupress.org/content/111/6/2851.full.pdf

L. Breuza, M. Garcia, M. Delgrossi, L. Bivic, and A. , Role of the membrane-proximal Oglycosylation site in sorting of the human receptor for neurotrophins to the apical membrane of MDCK cells, Exp. Cell Res, vol.273, pp.178-186, 2002.
URL : https://hal.archives-ouvertes.fr/hal-00306518

G. M. Brodeur, J. E. Minturn, R. Ho, A. M. Simpson, R. Iyer et al., Trk receptor expression and inhibition in neuroblastomas, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.15, pp.3244-3250, 2009.
DOI : 10.1158/1078-0432.ccr-08-1815

URL : http://clincancerres.aacrjournals.org/content/15/10/3244.full.pdf

C. Brodie and E. W. Gelfand, Functional nerve growth factor receptors on human B lymphocytes. Interaction with IL-2, J. Immunol. Baltim. Md, vol.148, pp.3492-3497, 1950.

M. S. Brown, J. Ye, R. B. Rawson, and J. L. Goldstein, Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans, Cell, vol.100, pp.391-398, 2000.

A. Brunet, A. Bonni, M. J. Zigmond, M. Z. Lin, P. Juo et al., Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor, Cell, vol.96, pp.857-868, 1999.
DOI : 10.1016/s0092-8674(00)80595-4

URL : https://doi.org/10.1016/s0092-8674(00)80595-4

J. K. Bubien, L. J. Zhou, P. D. Bell, R. A. Frizzell, and T. F. Tedder, Transfection of the CD20 cell surface molecule into ectopic cell types generates a Ca2+ conductance found constitutively in B lymphocytes, J. Cell Biol, vol.121, pp.1121-1132, 1993.

N. T. Bui, H. König, C. Culmsee, E. Bauerbach, M. Poppe et al., p75 neurotrophin receptor is required for constitutive and NGF-induced survival signalling in PC12 cells and rat hippocampal neurones, J. Neurochem, vol.81, pp.594-605, 2002.
DOI : 10.1046/j.1471-4159.2002.00841.x

M. Caby, D. Lankar, C. Vincendeau-scherrer, G. Raposo, and C. Bonnerot, Exosomal-like vesicles are present in human blood plasma, Int. Immunol, vol.17, pp.879-887, 2005.
DOI : 10.1093/intimm/dxh267

URL : https://academic.oup.com/intimm/article-pdf/17/7/879/8201897/dxh267.pdf

R. Camicia, H. C. Winkler, and P. O. Hassa, Novel drug targets for personalized precision medicine in relapsed/refractory diffuse large B-cell lymphoma: a comprehensive review, Mol. Cancer, vol.14, 2015.

E. Campo, S. H. Swerdlow, N. L. Harris, S. Pileri, H. Stein et al., The 2008 WHO classification of lymphoid neoplasms and beyond: evolving concepts and practical applications, Blood, vol.117, pp.5019-5032, 2011.

S. Cang, N. Mukhi, K. Wang, and D. Liu, Novel CD20 monoclonal antibodies for lymphoma therapy, J. Hematol. Oncol.J Hematol Oncol, vol.5, p.64, 2012.
DOI : 10.1186/1756-8722-5-64

URL : https://jhoonline.biomedcentral.com/track/pdf/10.1186/1756-8722-5-64

M. Canuel, N. Bhattacharyya, A. Balbis, L. Yuan, and C. R. Morales, Sortilin and prosaposin localize to detergent-resistant membrane microdomains, Exp. Cell Res, vol.315, pp.240-247, 2009.
DOI : 10.1016/j.yexcr.2008.10.009

G. Cartron, L. Dacheux, G. Salles, P. Solal-celigny, P. Bardos et al., Therapeutic activity of humanized anti-CD20 monoclonal antibody and polymorphism in IgG Fc receptor FcgammaRIIIa gene, Blood, vol.99, pp.754-758, 2002.
DOI : 10.1182/blood.v99.3.754

URL : http://www.bloodjournal.org/content/99/3/754.full.pdf

G. Cartron, H. Watier, J. Golay, and P. Celigny, From the bench to the bedside: ways to improve rituximab efficacy, Blood, vol.104, pp.2635-2642, 2004.
DOI : 10.1182/blood-2004-03-1110

URL : http://www.bloodjournal.org/content/104/9/2635.full.pdf

M. Cazorla, A. Jouvenceau, C. Rose, J. Guilloux, C. Pilon et al., , 2010.

-. Cyclotraxin and . First, Highly Potent and Selective TrkB Inhibitor, Has Anxiolytic Properties in Mice, PLoS ONE, vol.5

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, J. Clin. Invest, vol.121, pp.1846-1857, 2011.

R. Chakravarthy, K. Mnich, and A. M. Gorman, Nerve growth factor (NGF)-mediated regulation of p75(NTR) expression contributes to chemotherapeutic resistance in triple negative breast cancer cells, Biochem. Biophys. Res. Commun, vol.478, pp.1541-1547, 2016.

M. Chang, J. C. Arevalo, and M. V. Chao, Ternary complex with Trk, p75, and an ankyrinrich membrane spanning protein, J. Neurosci. Res, vol.78, pp.186-192, 2004.

M. V. Chao and M. Bothwell, Neurotrophins: to cleave or not to cleave, Neuron, vol.33, pp.9-12, 2002.

M. V. Chao and B. L. Hempstead, p75 and Trk: a two-receptor system, Trends Neurosci, vol.18, pp.321-326, 1995.

M. V. Chao, M. A. Bothwell, A. H. Ross, H. Koprowski, A. A. Lanahan et al., Gene transfer and molecular cloning of the human NGF receptor, Science, vol.232, pp.518-521, 1986.

B. Chapuy, R. Koch, U. Radunski, S. Corsham, N. Cheong et al., Intracellular ABC transporter A3 confers multidrug resistance in leukemia cells by lysosomal drug sequestration, Leukemia, vol.22, pp.1576-1586, 2008.

W. Chen, X. Liu, M. Lv, L. Chen, J. Zhao et al., Exosomes from drug-resistant breast cancer cells transmit chemoresistance by a horizontal transfer of microRNAs, PloS One, vol.9, p.95240, 2014.

A. Chiaramello, K. Neuman, K. Palm, M. Metsis, and T. Neuman, Helix-loop-helix transcription factors mediate activation and repression of the p75LNGFR gene, Mol. Cell. Biol, vol.15, pp.6036-6044, 1995.

L. S. Chin, S. F. Murray, K. M. Zitnay, R. , and B. , K252a inhibits proliferation of glioma cells by blocking platelet-derived growth factor signal transduction, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.3, pp.771-776, 1997.

N. Chitranshi, V. Gupta, Y. Dheer, V. Gupta, R. Vander-wall et al., Molecular determinants and interaction data of cyclic peptide inhibitor with the extracellular domain of TrkB receptor, Data Brief, vol.6, pp.776-782, 2016.

W. W. Choi, D. D. Weisenburger, T. C. Greiner, M. A. Piris, A. H. Banham et al., A new immunostain algorithm classifies diffuse large B-cell lymphoma into molecular subtypes with high accuracy, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.15, pp.5494-5502, 2009.

V. Chopin, C. Lagadec, R. Toillon, L. Bourhis, and X. , Neurotrophin signaling in cancer stem cells, Cell. Mol. Life Sci. CMLS, vol.73, pp.1859-1870, 2016.

V. Ciravolo, V. Huber, G. C. Ghedini, E. Venturelli, F. Bianchi et al., Potential role of HER2-overexpressing exosomes in countering trastuzumab-based therapy, J. Cell. Physiol, vol.227, pp.658-667, 2012.

D. O. Clary and L. F. Reichardt, An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.11133-11137, 1994.

R. A. Clynes, T. L. Towers, L. G. Presta, and J. V. Ravetch, Inhibitory Fc receptors modulate in vivo cytotoxicity against tumor targets, Nat. Med, vol.6, pp.443-446, 2000.

S. Cohen, R. Levi-montalcini, and V. Hamburger, A NERVE GROWTH-STIMULATING FACTOR ISOLATED FROM SARCOM AS 37 AND 180, Proc. Natl. Acad. Sci. U. S. A, vol.40, pp.1014-1018, 1954.

B. Coiffier, State-of-the-art therapeutics: diffuse large B-cell lymphoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.23, pp.6387-6393, 2005.

B. Coiffier, Rituximab therapy in malignant lymphoma, Oncogene, vol.26, pp.3603-3613, 2007.

B. Coiffier, E. Lepage, J. Briere, R. Herbrecht, H. Tilly et al., CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma, N. Engl. J. Med, vol.346, pp.235-242, 2002.

B. Coiffier, P. Feugier, N. Mounier, P. Franchi-rezgui, E. Van-den-neste et al., Long-term results of the GELA study comparing R-CHOP and CHOP chemotherapy in older patients with diffuse large B-cell lymphoma show good survival in poor-risk patients, ASCO Meet. Abstr, vol.25, p.8009, 2007.

K. C. Corbit, D. A. Foster, and M. R. Rosner, Protein kinase Cdelta mediates neurogenic but not mitogenic activation of mitogen-activated protein kinase in neuronal cells, Mol. Cell. Biol, vol.19, pp.4209-4218, 1999.

C. Corcoran, S. Rani, K. O'brien, A. O'neill, M. Prencipe et al., Docetaxel-resistance in prostate cancer: evaluating associated phenotypic changes and potential for resistance transfer via exosomes, PloS One, vol.7, p.50999, 2012.

E. J. Coulson, K. Reid, M. Baca, K. A. Shipham, S. M. Hulett et al., Chopper, a new death domain of the p75 neurotrophin receptor that mediates rapid neuronal cell death, J. Biol. Chem, vol.275, pp.30537-30545, 2000.

S. Covaceuszach, P. V. Konarev, A. Cassetta, F. Paoletti, D. I. Svergun et al., The conundrum of the high-affinity NGF binding site formation unveiled?, Biophys. J, vol.108, pp.687-697, 2015.

M. S. Cragg, S. M. Morgan, H. T. Chan, B. P. Morgan, A. V. Filatov et al., Complement-mediated lysis by anti-CD20 mAb correlates with segregation into lipid rafts, Blood, vol.101, pp.1045-1052, 2003.

J. L. Croucher, R. Iyer, N. Li, V. Molteni, J. Loren et al., TrkB inhibition by GNF-4256 slows growth and enhances chemotherapeutic efficacy in neuroblastoma xenografts, Cancer Chemother. Pharmacol, vol.75, pp.131-141, 2015.

J. L. Cultrera, D. , and S. M. , Diffuse large B-cell lymphoma: current strategies and future directions, Cancer Control J. Moffitt Cancer Cent, vol.19, pp.204-213, 2012.

M. S. Czuczman, S. Olejniczak, A. Gowda, A. Kotowski, A. Binder et al., Acquirement of rituximab resistance in lymphoma cell lines is associated with both global CD20 gene and protein down-regulation regulated at the pretranscriptional and posttranscriptional levels, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.14, pp.1561-1570, 2008.

R. E. Davis, K. D. Brown, U. Siebenlist, and L. M. Staudt, Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells, J. Exp. Med, vol.194, pp.1861-1874, 2001.

F. G. De-braud, M. Niger, S. Damian, B. Bardazza, A. Martinetti et al., Alka-372-001: First-in-human, phase I study of entrectinib-an oral pan-trk, ROS1, and ALK inhibitor-in patients with advanced solid tumors with relevant molecular alterations, J Clin Oncol, vol.33, 2015.

P. De-paepe, D. Wolf-peeters, and C. , Diffuse large B-cell lymphoma: a heterogeneous group of non-Hodgkin lymphomas comprising several distinct clinicopathological entities, Leukemia, vol.21, pp.37-43, 2007.

P. De-paepe, R. Achten, G. Verhoef, I. Wlodarska, M. Stul et al., Large cleaved and immunoblastic lymphoma may represent two distinct clinicopathologic entities within the group of diffuse large B-cell lymphomas, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.23, pp.7060-7068, 2005.

J. P. Deans, H. Li, and M. J. Polyak, CD20-mediated apoptosis: signalling through lipid rafts, Immunology, vol.107, pp.176-182, 2002.

G. Dechant, B. , and Y. A. , Signalling through the neurotrophin receptor p75NTR, Curr. Opin. Neurobiol, vol.7, pp.413-418, 1997.

M. F. Defreitas, P. S. Mcquillen, and C. J. Shatz, A novel p75NTR signaling pathway promotes survival, not death, of immunopurified neocortical subplate neurons, J. Neurosci. Off. J. Soc. Neurosci, vol.21, pp.5121-5129, 2001.

C. Demosthenous, J. J. Han, M. J. Stenson, M. J. Maurer, L. E. Wellik et al., Translation initiation complex eIF4F is a therapeutic target for dual mTOR kinase inhibitors in non-Hodgkin lymphoma, Oncotarget, vol.6, pp.9488-9501, 2015.

S. Descamps, R. A. Toillon, E. Adriaenssens, V. Pawlowski, S. M. Cool et al., Nerve growth factor stimulates proliferation and survival of human breast cancer cells through two distinct signaling pathways, J. Biol. Chem, vol.276, pp.17864-17870, 2001.

B. B. Ding, J. J. Yu, R. Y. Yu, L. M. Mendez, R. Shaknovich et al., Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large B-cell lymphomas, Blood, vol.111, pp.1515-1523, 2008.

R. T. Dobrowsky, M. H. Werner, A. M. Castellino, M. V. Chao, and Y. A. Hannun, Activation of the sphingomyelin cycle through the low-affinity neurotrophin receptor, Science, vol.265, pp.1596-1599, 1994.

R. C. Doebele, L. E. Davis, A. Vaishnavi, A. T. Le, A. Estrada-bernal et al., An Oncogenic NTRK Fusion in a Patient with Soft-Tissue Sarcoma with Response to the Tropomyosin-Related Kinase Inhibitor LOXO-101, Cancer Discov, vol.5, pp.1049-1057, 2015.

L. Dollé, I. El-yazidi-belkoura, E. Adriaenssens, V. Nurcombe, and H. Hondermarck, Nerve growth factor overexpression and autocrine loop in breast cancer cells, Oncogene, vol.22, pp.5592-5601, 2003.

D. Onofrio, M. De-grazia, U. Morrone, S. Cuomo, L. Spinsanti et al., Expression of neurotrophin receptors in normal and malignant B lymphocytes, Eur. Cytokine Netw, vol.11, pp.283-291, 2000.

D. A. Doyle, A. Lee, J. Lewis, E. Kim, M. Sheng et al., Crystal structures of a complexed and peptide-free membrane protein-binding domain: molecular basis of peptide recognition by PDZ, Cell, vol.85, pp.1067-1076, 1996.

F. Drouet, X. Cahu, Y. Pointreau, F. Denis, and M. Mahé, , 2010.

, Cancer Radiothérapie J. Société Fr. Radiothérapie Oncol, vol.14, issue.1, pp.210-229

L. Dubanet, H. Bentayeb, B. Petit, A. Olivrie, S. Saada et al., Anti-apoptotic role and clinical relevance of neurotrophins in diffuse large B-cell lymphomas, Br. J. Cancer, vol.113, pp.934-944, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01500853

H. Dudek, S. R. Datta, T. F. Franke, M. J. Birnbaum, R. Yao et al., Regulation of neuronal survival by the serine-threonine protein kinase, Akt. Science, vol.275, pp.661-665, 1997.

A. E. Edling, T. Nanavati, J. M. Johnson, and V. K. Tuohy, Human and murine lymphocyte neurotrophin expression is confined to B cells, J. Neurosci. Res, vol.77, pp.709-717, 2004.

D. A. Einfeld, J. P. Brown, M. A. Valentine, E. A. Clark, and J. A. Ledbetter, Molecular cloning of the human B cell CD20 receptor predicts a hydrophobic protein with multiple transmembrane domains, EMBO J, vol.7, pp.711-717, 1988.

W. R. Epa, K. Markovska, and G. L. Barrett, The p75 neurotrophin receptor enhances TrkA signalling by binding to Shc and augmenting its phosphorylation, J. Neurochem, vol.89, pp.344-353, 2004.

A. F. Farago, L. P. Le, Z. Zheng, A. Muzikansky, A. Drilon et al., Durable Clinical Response to Entrectinib in NTRK1Rearranged Non-Small Cell Lung Cancer, J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer, vol.10, pp.1670-1674, 2015.

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, J. Immunol. Baltim. Md, vol.181, pp.3027-3038, 1950.
URL : https://hal.archives-ouvertes.fr/hal-00453223

D. Feng, T. Kim, E. Özkan, M. Light, R. Torkin et al., Molecular and Structural Insight into proNGF Engagement of p75NTR and Sortilin, J. Mol. Biol, vol.396, pp.967-984, 2010.

B. M. Fenner, Truncated TrkB: beyond a dominant negative receptor, Cytokine Growth Factor Rev, vol.23, pp.15-24, 2012.

P. Feugier, A. Van-hoof, C. Sebban, P. Solal-celigny, R. Bouabdallah et al., Long-term results of the R-CHOP study in the treatment of elderly patients with diffuse large B-cell lymphoma: a study by the Groupe d'Etude des Lymphomes de l'Adulte, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.23, pp.4117-4126, 2005.

C. R. Flowers, R. Sinha, and J. M. Vose, Improving Outcomes for Patients With Diffuse Large B-Cell Lymphoma, CA. Cancer J. Clin, 2010.

L. Fontán and A. Melnick, Molecular pathways: targeting MALT1 paracaspase activity in lymphoma, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.19, pp.6662-6668, 2013.

). France and C. , Médicaments utilisés en cancérologie: évaluation thérapeutique (CNHIM), 2001.

R. A. Franklin, C. Brodie, I. Melamed, N. Terada, J. J. Lucas et al., Nerve growth factor induces activation of MAP-kinase and p90rsk in human B lymphocytes, J. Immunol. Baltim. Md, vol.154, pp.4965-4972, 1950.

R. D. Gascoyne, Pathologic prognostic factors in diffuse aggressive non-Hodgkin's lymphoma, Hematol. Oncol. Clin. North Am, vol.11, pp.847-862, 1997.

A. De-gassart, C. Geminard, B. Fevrier, G. Raposo, and M. Vidal, Lipid raft-associated protein sorting in exosomes, Blood, vol.102, pp.4336-4344, 2003.

K. Gatter and F. Pezzella, Diffuse large B-cell lymphoma, Diagn. Histopathol, vol.16, pp.69-81, 2010.

R. Ge, E. Tan, S. Sharghi-namini, and H. H. Asada, Exosomes in Cancer Microenvironment and Beyond: have we Overlooked these Extracellular Messengers? Cancer Microenviron, vol.5, pp.323-332, 2012.

J. J. Gentry, N. J. Rutkoski, T. L. Burke, and B. D. Carter, A functional interaction between the p75 neurotrophin receptor interacting factors, TRAF6 and NRIF, J. Biol. Chem, vol.279, pp.16646-16656, 2004.

D. J. Glass, S. H. Nye, P. Hantzopoulos, M. J. Macchi, S. P. Squinto et al., TrkB mediates BDNF/NT-3-dependent survival and proliferation in fibroblasts lacking the low affinity NGF receptor, Cell, vol.66, pp.405-413, 1991.

W. L. Gluck, D. Hurst, A. Yuen, A. M. Levine, M. A. Dayton et al., Phase I studies of interleukin (IL)-2 and rituximab in Bcell non-hodgkin's lymphoma: IL-2 mediated natural killer cell expansion correlations with clinical response, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.10, pp.2253-2264, 2004.

J. Golay, M. Lazzari, V. Facchinetti, S. Bernasconi, G. Borleri et al., CD20 levels determine the in vitro susceptibility to rituximab and complement of Bcell chronic lymphocytic leukemia: further regulation by CD55 and CD59, Blood, vol.98, pp.3383-3389, 2001.

P. A. Gonzales, T. Pisitkun, J. D. Hoffert, D. Tchapyjnikov, R. A. Star et al., Large-scale proteomics and phosphoproteomics of urinary exosomes, J. Am. Soc. Nephrol. JASN, vol.20, pp.363-379, 2009.

J. R. Graff, B. W. Konicek, J. H. Carter, and E. G. Marcusson, Targeting the eukaryotic translation initiation factor 4E for cancer therapy, Cancer Res, vol.68, pp.631-634, 2008.

P. M. Grob, A. H. Ross, H. Koprowski, and M. Bothwell, Characterization of the human melanoma nerve growth factor receptor, J. Biol. Chem, vol.260, pp.8044-8049, 1985.

M. Guiton, F. J. Gunn-moore, D. J. Glass, D. R. Geis, G. D. Yancopoulos et al., Naturally occurring tyrosine kinase inserts block high affinity binding of phospholipase C gamma and Shc to TrkC and neurotrophin-3 signaling, J. Biol. Chem, vol.270, pp.20384-20390, 1995.

T. M. Habermann, E. A. Weller, V. A. Morrison, R. D. Gascoyne, P. A. Cassileth et al., Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.24, pp.3121-3127, 2006.

P. R. Hagner, A. Schneider, and R. B. Gartenhaus, Targeting the translational machinery as a novel treatment strategy for hematologic malignancies, Blood, vol.115, pp.2127-2135, 2010.

F. Hallböök, Evolution of the vertebrate neurotrophin and Trk receptor gene families, Curr. Opin. Neurobiol, vol.9, pp.616-621, 1999.

F. Hallböök, C. F. Ibáñez, P. , and H. , Evolutionary studies of the nerve growth factor family reveal a novel member abundantly expressed in Xenopus ovary, Neuron, vol.6, pp.845-858, 1991.

W. Hampe, M. Rezgaoui, I. Hermans-borgmeyer, and C. H. Schaller, The genes for the human VPS10 domain-containing receptors are large and contain many small exons, Hum. Genet, vol.108, pp.529-536, 2001.

B. N. Hannafon and W. Ding, Intercellular communication by exosome-derived microRNAs in cancer, Int. J. Mol. Sci, vol.14, pp.14240-14269, 2013.

C. P. Hans, D. D. Weisenburger, T. C. Greiner, R. D. Gascoyne, J. Delabie et al., Confirmation of the molecular classification of diffuse large B-cell lymphoma by immunohistochemistry using a tissue microarray, Blood, vol.103, pp.275-282, 2004.

A. Harjunpää, T. Wiklund, J. Collan, R. Janes, J. Rosenberg et al., Complement activation in circulation and central nervous system after rituximab (antiCD20) treatment of B-cell lymphoma, Leuk. Lymphoma, vol.42, pp.731-738, 2001.

A. W. Harrington, J. Y. Kim, and S. O. Yoon, Activation of Rac GTPase by p75 is necessary for c-jun N-terminal kinase-mediated apoptosis, J. Neurosci. Off. J. Soc. Neurosci, vol.22, pp.156-166, 2002.

N. L. Harris, E. S. Jaffe, H. Stein, P. M. Banks, J. K. Chan et al., A revised European-American classification of lymphoid neoplasms: a proposal from the International Lymphoma Study Group, Blood, vol.84, pp.1361-1392, 1994.

B. L. Hempstead, D. Martin-zanca, D. R. Kaplan, L. F. Parada, and M. V. Chao, High-affinity NGF binding requires coexpression of the trk proto-oncogene and the low-affinity NGF receptor, Nature, vol.350, pp.678-683, 1991.

C. Henry, M. Deschamps, P. Rohrlich, J. Pallandre, J. Rémy-martin et al., Identification of an alternative CD20 transcript variant in B-cell malignancies coding for a novel protein associated to rituximab resistance, Blood, vol.115, pp.2420-2429, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00484312

K. Herrup and E. M. Shooter, Properties of the beta nerve growth factor receptor of avian dorsal root ganglia, Proc. Natl. Acad. Sci. U. S. A, vol.70, pp.3884-3888, 1973.

N. P. Hessvik, K. Sandvig, L. , and A. , Exosomal miRNAs as Biomarkers for Prostate Cancer, p.36, 2013.

H. Heyn and M. Esteller, EZH2: an epigenetic gatekeeper promoting lymphomagenesis, Cancer Cell, vol.23, pp.563-565, 2013.

J. Hillis, M. O'dwyer, and A. M. Gorman, Neurotrophins and B-cell malignancies, Cell. Mol. Life Sci. CMLS, vol.73, pp.41-56, 2016.

J. Hiraga, A. Tomita, T. Sugimoto, K. Shimada, M. Ito et al., Down-regulation of CD20 expression in B-cell lymphoma cells after treatment with rituximab-containing combination chemotherapies: its prevalence and clinical significance, Blood, vol.113, pp.4885-4893, 2009.

S. Hirai, M. Izawa, S. Osada, G. Spyrou, and S. Ohno, Activation of the JNK pathway by distantly related protein kinases, MEKK and MUK, Oncogene, vol.12, pp.641-650, 1996.

J. K. Hofmeister, D. Cooney, and K. M. Coggeshall, Clustered CD20 induced apoptosis: srcfamily kinase, the proximal regulator of tyrosine phosphorylation, calcium influx, and caspase 3-dependent apoptosis, Blood Cells. Mol. Dis, vol.26, pp.133-143, 2000.

A. Hohn, J. Leibrock, K. Bailey, B. , and Y. A. , Identification and characterization of a novel member of the nerve growth factor/brain-derived neurotrophic factor family, Nature, vol.344, pp.339-341, 1990.

P. H. Holden, V. Asopa, A. G. Robertson, A. R. Clarke, S. Tyler et al., Immunoglobulin-like domains define the nerve growth factor binding site of the TrkA receptor, Nat. Biotechnol, vol.15, pp.668-672, 1997.

M. Holgado-madruga, D. K. Moscatello, D. R. Emlet, R. Dieterich, and A. J. Wong, Grb2associated binder-1 mediates phosphatidylinositol 3-kinase activation and the promotion of cell survival by nerve growth factor, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.12419-12424, 1997.

H. Hondermarck, Neurotrophins and their receptors in breast cancer, Cytokine Growth Factor Rev, vol.23, pp.357-365, 2012.

E. Horvilleur, T. Sbarrato, K. Hill, R. V. Spriggs, M. Screen et al., A role for eukaryotic initiation factor 4B overexpression in the pathogenesis of diffuse large B-cell lymphoma, Leukemia, vol.28, pp.1092-1102, 2014.

S. Hu, Z. Y. Xu-monette, A. Tzankov, T. Green, L. Wu et al., MYC/BCL2 protein coexpression contributes to the inferior survival of activated B-cell subtype of diffuse large B-cell lymphoma and demonstrates highrisk gene expression signatures: a report from The International DLBCL Rituximab-CHOP Consortium Program, Blood, vol.121, pp.4021-4031, 2013.

M. Hummel, S. Bentink, H. Berger, W. Klapper, S. Wessendorf et al., A biologic definition of Burkitt's lymphoma from transcriptional and genomic profiling, N. Engl. J. Med, vol.354, pp.2419-2430, 2006.

J. Iqbal, P. N. Meyer, L. M. Smith, N. A. Johnson, J. M. Vose et al., BCL2 Predicts Survival in Germinal Center B-celllike Diffuse Large B-cell Lymphoma Treated with CHOP-like Therapy and Rituximab, Clin. Cancer Res, vol.17, pp.7785-7795, 2011.

K. Ishibashi, M. Suzuki, S. Sasaki, and M. Imai, Identification of a new multigene fourtransmembrane family (MS4A) related to CD20, HTm4 and beta subunit of the high-affinity IgE receptor, Gene, vol.264, pp.87-93, 2001.

R. Iyer, A. E. Evans, X. Qi, R. Ho, J. E. Minturn et al., Lestaurtinib enhances the antitumor efficacy of chemotherapy in murine xenograft models of neuroblastoma, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.16, pp.1478-1485, 2010.

R. Iyer, C. R. Varela, J. E. Minturn, R. Ho, A. M. Simpson et al., AZ64 inhibits TrkB and enhances the efficacy of chemotherapy and local radiation in neuroblastoma xenografts, Cancer Chemother. Pharmacol, vol.70, pp.477-486, 2012.

R. Iyer, L. Wehrmann, R. L. Golden, K. Naraparaju, J. L. Croucher et al., Entrectinib is a potent inhibitor of Trk-driven neuroblastomas in a xenograft mouse model, Cancer Lett, vol.372, pp.179-186, 2016.

R. Janknecht, W. H. Ernst, V. Pingoud, and A. Nordheim, Activation of ternary complex factor Elk-1 by MAP kinases, EMBO J, vol.12, pp.5097-5104, 1993.

P. Jansen, K. Giehl, J. R. Nyengaard, K. Teng, O. Lioubinski et al., Roles for the pro-neurotrophin receptor sortilin in neuronal development, aging and brain injury, Nat. Neurosci, vol.10, pp.1449-1457, 2007.

A. R. Jazirehi and B. Bonavida, Development of rituximab-resistant B-NHL clones: an in vitro model for studying tumor resistance to monoclonal antibody-mediated immunotherapy, Methods Mol. Biol. Clifton NJ, vol.731, pp.407-419, 2011.

A. R. Jazirehi, M. I. Vega, and B. Bonavida, Development of rituximab-resistant lymphoma clones with altered cell signaling and cross-resistance to chemotherapy, Cancer Res, vol.67, pp.1270-1281, 2007.

I. Jilani, S. O'brien, T. Manshuri, D. A. Thomas, V. A. Thomazy et al., Transient down-modulation of CD20 by rituximab in patients with chronic lymphocytic leukemia, Blood, vol.102, pp.3514-3520, 2003.

H. Jin, Y. Pan, L. He, H. Zhai, X. Li et al., p75 neurotrophin receptor inhibits invasion and metastasis of gastric cancer, Mol. Cancer Res. MCR, vol.5, pp.423-433, 2007.
DOI : 10.1158/1541-7786.mcr-06-0407

URL : http://mcr.aacrjournals.org/content/5/5/423.full.pdf

D. Johnson, A. Lanahan, C. R. Buck, A. Sehgal, C. Morgan et al., Expression and structure of the human NGF receptor, Cell, vol.47, pp.545-554, 1986.

N. A. Johnson, M. Boyle, A. Bashashati, S. Leach, A. Brooks-wilson et al., Diffuse large B-cell lymphoma: reduced CD20 expression is associated with an inferior survival, Blood, vol.113, pp.3773-3780, 2009.
DOI : 10.1182/blood-2008-09-177469

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2943836/pdf

R. M. Johnstone, Revisiting the road to the discovery of exosomes, Blood Cells. Mol. Dis, vol.34, pp.214-219, 2005.

K. R. Jones and L. F. Reichardt, Molecular cloning of a human gene that is a member of the nerve growth factor family, Proc. Natl. Acad. Sci. U. S. A, vol.87, pp.8060-8064, 1990.

P. Kahle, P. A. Barker, E. M. Shooter, and C. Hertel, p75 nerve growth factor receptor modulates p140trkA kinase activity, but not ligand internalization, in PC12 cells, J. Neurosci. Res, vol.38, pp.599-606, 1994.
DOI : 10.1002/jnr.490380512

C. Kahlert and R. Kalluri, Exosomes in tumor microenvironment influence cancer progression and metastasis, J. Mol. Med. Berl. Ger, vol.91, pp.431-437, 2013.
DOI : 10.1007/s00109-013-1020-6

URL : http://europepmc.org/articles/pmc4073669?pdf=render

H. Kalra, R. J. Simpson, H. Ji, E. Aikawa, P. Altevogt et al., Vesiclepedia: a compendium for extracellular vesicles with continuous community annotation, PLoS Biol, vol.10, p.1001450, 2012.
DOI : 10.1371/journal.pbio.1001450

URL : https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.1001450&type=printable

D. Kang, S. Oh, S. Ahn, B. Lee, and M. H. Moon, Proteomic analysis of exosomes from human neural stem cells by flow field-flow fractionation and nanoflow liquid chromatography-tandem mass spectrometry, J. Proteome Res, vol.7, pp.3475-3480, 2008.

D. R. Kaplan and F. D. Miller, Neurobiology: A move to sort life from death, Nature, vol.427, pp.798-799, 2004.
DOI : 10.1038/427798a

N. Kawamura, K. Kawamura, M. Manabe, and T. Tanaka, Inhibition of brain-derived neurotrophic factor/tyrosine kinase B signaling suppresses choriocarcinoma cell growth, Endocrinology, vol.151, pp.3006-3014, 2010.
DOI : 10.1210/en.2009-1378

URL : https://academic.oup.com/endo/article-pdf/151/7/3006/7411513/endo3006.pdf

J. H. Kehrl, A. Riva, G. L. Wilson, and C. Thévenin, Molecular mechanisms regulating CD19, CD20 and CD22 gene expression, Immunol. Today, vol.15, pp.432-436, 1994.

S. Keller, M. P. Sanderson, A. Stoeck, A. , and P. , Exosomes: from biogenesis and secretion to biological function, Immunol. Lett, vol.107, pp.102-108, 2006.

R. S. Kenchappa, N. Zampieri, M. V. Chao, P. A. Barker, H. K. Teng et al., Ligand-dependent cleavage of the P75 neurotrophin receptor is necessary for NRIF nuclear translocation and apoptosis in sympathetic neurons, Neuron, vol.50, pp.219-232, 2006.

M. Kerschensteiner, E. Gallmeier, L. Behrens, V. V. Leal, T. Misgeld et al., Activated human T cells, B cells, and monocytes produce brain-derived neurotrophic factor in vitro and in inflammatory brain lesions: a neuroprotective role of inflammation?, J. Exp. Med, vol.189, pp.865-870, 1999.

P. Kharaziha, S. Ceder, Q. Li, P. , and T. , Tumor cell-derived exosomes: a message in a bottle, Biochim. Biophys. Acta, vol.1826, pp.103-111, 2012.

G. Khursigara, J. Bertin, H. Yano, H. Moffett, P. S. Distefano et al., A prosurvival function for the p75 receptor death domain mediated via the caspase recruitment domain receptor-interacting protein 2, J. Neurosci. Off. J. Soc. Neurosci, vol.21, pp.5854-5863, 2001.

F. S. Khwaja, E. J. Quann, N. Pattabiraman, S. Wynne, and D. Djakiew, Carprofen induction of p75NTR-dependent apoptosis via the p38 mitogen-activated protein kinase pathway in prostate cancer cells, Mol. Cancer Ther, vol.7, pp.3539-3545, 2008.

T. Kim and B. L. Hempstead, NRH2 is a trafficking switch to regulate sortilin localization and permit proneurotrophin-induced cell death, EMBO J, vol.28, pp.1612-1623, 2009.

D. Kim, B. Kang, O. Y. Kim, D. Choi, J. Lee et al., EVpedia: an integrated database of high-throughput data for systemic analyses of extracellular vesicles, J. Extracell. Vesicles, vol.2, 2013.

S. Kim, A. A. Salim, S. M. Swanson, and A. D. Kinghorn, Potential of cyclopenta[b]benzofurans from Aglaia species in cancer chemotherapy, Anticancer Agents Med. Chem, vol.6, pp.319-345, 2006.

H. Kimata, A. Yoshida, C. Ishioka, T. Kusunoki, S. Hosoi et al., Nerve growth factor specifically induces human IgG4 production, Eur. J. Immunol, vol.21, pp.137-141, 1991.

M. L. King, C. Chiang, H. Ling, E. Fujita, M. Ochiai et al., X-Ray crystal structure of rocaglamide, a novel antileulemic 1H-cyclopenta[b]benzofuran from Aglaia elliptifolia, J. Chem. Soc., Chem. Commun, pp.1150-1151, 1982.

U. Klein and R. Dalla-favera, Germinal centres: role in B-cell physiology and malignancy, Nat. Rev. Immunol, vol.8, pp.22-33, 2008.

R. Klein, L. F. Parada, F. Coulier, and M. Barbacid, trkB, a novel tyrosine protein kinase receptor expressed during mouse neural development, EMBO J, vol.8, pp.3701-3709, 1989.

R. Klein, S. Q. Jing, V. Nanduri, E. O'rourke, and M. Barbacid, The trk proto-oncogene encodes a receptor for nerve growth factor, Cell, vol.65, pp.189-197, 1991.

R. Klein, V. Nanduri, S. A. Jing, F. Lamballe, P. Tapley et al., The trkB tyrosine protein kinase is a receptor for brain-derived neurotrophic factor and neurotrophin-3, Cell, vol.66, pp.395-403, 1991.

S. Knapper, A. K. Burnett, T. Littlewood, W. J. Kell, S. Agrawal et al., A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy, Blood, vol.108, pp.3262-3270, 2006.

R. Koch, T. Aung, D. Vogel, B. Chapuy, D. Wenzel et al., Nuclear Trapping through Inhibition of Exosomal Export by Indomethacin Increases Cytostatic Efficacy of Doxorubicin and Pixantrone, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.22, pp.395-404, 2016.

H. R. Koene, M. Kleijer, J. Algra, D. Roos, A. E. Von-dem-borne et al., Fc gammaRIIIa-158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa-48L/R/H phenotype, Blood, vol.90, pp.1109-1114, 1997.

H. E. Kohrt, I. Sagiv-barfi, S. Rafiq, S. E. Herman, J. P. Butchar et al., Ibrutinib antagonizes rituximab-dependent NK cellmediated cytotoxicity, Blood, vol.123, pp.1957-1960, 2014.

L. E. Van-der-kolk, A. J. Grillo-lópez, J. W. Baars, C. E. Hack, and M. H. Van-oers, Complement activation plays a key role in the side-effects of rituximab treatment, Br. J. Haematol, vol.115, pp.807-811, 2001.

D. Krappmann, Attacking MALT1 for ABC-DLBCL therapy, Oncotarget, vol.3, pp.1489-1490, 2012.
DOI : 10.18632/oncotarget.794

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=download&path%5B%5D=794&path%5B%5D=1126

E. Krock, J. B. Currie, M. H. Weber, J. A. Ouellet, L. S. Stone et al., Nerve Growth Factor Is Regulated by Toll-Like Receptor 2 in Human Intervertebral Discs, J. Biol. Chem, vol.291, pp.3541-3551, 2016.

I. Kronfeld, G. Kazimirsky, E. W. Gelfand, B. , and C. , NGF rescues human B lymphocytes from anti-IgM induced apoptosis by activation of PKCzeta, Eur. J. Immunol, vol.32, pp.136-143, 2002.

E. Labouyrie, M. Parrens, A. De-mascarel, B. Bloch, and J. P. Merlio, Distribution of NGF receptors in normal and pathologic human lymphoid tissues, J. Neuroimmunol, vol.77, pp.161-173, 1997.

E. Labouyrie, P. Dubus, A. Groppi, F. X. Mahon, J. Ferrer et al., Expression of neurotrophins and their receptors in human bone marrow, Am. J. Pathol, vol.154, pp.405-415, 1999.

L. T. Lam, R. E. Davis, J. Pierce, M. Hepperle, Y. Xu et al., Small molecule inhibitors of IkappaB kinase are selectively toxic for subgroups of diffuse large B-cell lymphoma defined by gene expression profiling, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.11, pp.28-40, 2005.

L. T. Lam, G. Wright, R. E. Davis, G. Lenz, P. Farinha et al., Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-{kappa}B pathways in subtypes of diffuse large B-cell lymphoma, Blood, vol.111, pp.3701-3713, 2008.

F. Lamballe, R. Klein, and M. Barbacid, trkC, a new member of the trk family of tyrosine protein kinases, is a receptor for neurotrophin-3, Cell, vol.66, pp.967-979, 1991.

A. Lambiase, L. Bracci-laudiero, S. Bonini, S. Bonini, G. Starace et al., Human CD4+ T cell clones produce and release nerve growth factor and express high-affinity nerve growth factor receptors, J. Allergy Clin. Immunol, vol.100, pp.408-414, 1997.
DOI : 10.1016/s0091-6749(97)70256-2

H. G. Lamparski, A. Metha-damani, J. Yao, S. Patel, D. Hsu et al., Production and characterization of clinical grade exosomes derived from dendritic cells, J. Immunol. Methods, vol.270, pp.211-226, 2002.

S. Lawn, N. Krishna, A. Pisklakova, X. Qu, D. A. Fenstermacher et al., Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells, J. Biol. Chem, vol.290, pp.3814-3824, 2015.

R. Lee, P. Kermani, K. K. Teng, and B. L. Hempstead, Regulation of cell survival by secreted proneurotrophins, Science, vol.294, pp.1945-1948, 2001.

S. J. Lee, G. G. Li, S. T. Kim, M. E. Hong, J. Jang et al., NTRK1 rearrangement in colorectal cancer patients: evidence for actionable target using patient-derived tumor cell line, Oncotarget, vol.6, pp.39028-39035, 2015.

J. Leibrock, F. Lottspeich, A. Hohn, M. Hofer, B. Hengerer et al., Molecular cloning and expression of brain-derived neurotrophic factor, Nature, vol.341, pp.149-152, 1989.

G. Lenz and L. M. Staudt, Aggressive lymphomas, N. Engl. J. Med, vol.362, pp.1417-1429, 2010.
DOI : 10.1056/nejmra0807082

G. Lenz, G. Wright, S. S. Dave, W. Xiao, J. Powell et al., Stromal gene signatures in large-B-cell lymphomas, N. Engl. J. Med, vol.359, pp.2313-2323, 2008.

G. Lenz, G. W. Wright, N. C. Emre, H. Kohlhammer, S. S. Dave et al., Molecular subtypes of diffuse large B-cell lymphoma arise by distinct genetic pathways, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.13520-13525, 2008.

G. Lenz, G. Wright, S. S. Dave, W. Xiao, J. Powell et al., Stromal gene signatures in large-B-cell lymphomas, N. Engl. J. Med, vol.359, pp.2313-2323, 2008.
DOI : 10.1056/nejmoa0802885

URL : https://digitalcommons.unmc.edu/cgi/viewcontent.cgi?article=1031&context=com_pathmicro_articles

V. Lessmann, K. Gottmann, and M. Malcangio, Neurotrophin secretion: current facts and future prospects, Prog. Neurobiol, vol.69, pp.341-374, 2003.
DOI : 10.1016/s0301-0082(03)00019-4

C. Léveillé, R. Al-daccak, M. , and W. , CD20 is physically and functionally coupled to MHC class II and CD40 on human B cell lines, Eur. J. Immunol, vol.29, pp.65-74, 1999.

H. Li, L. M. Ayer, J. Lytton, and J. P. Deans, Store-operated cation entry mediated by CD20 in membrane rafts, J. Biol. Chem, vol.278, pp.42427-42434, 2003.

H. Li, L. M. Ayer, M. J. Polyak, C. M. Mutch, R. J. Petrie et al., The CD20 calcium channel is localized to microvilli and constitutively associated with membrane rafts: antibody binding increases the affinity of the association through an epitope-dependent cross-linking-independent mechanism, J. Biol. Chem, vol.279, pp.19893-19901, 2004.

J. Li, C. A. Sherman-baust, M. Tsai-turton, R. E. Bristow, R. B. Roden et al., , 2009.

, Claudin-containing exosomes in the peripheral circulation of women with ovarian cancer, BMC Cancer, vol.9, p.244

Z. Li, Y. Ma, J. Wang, X. Zeng, R. Li et al., Exosomal microRNA141 is upregulated in the serum of prostate cancer patients, OncoTargets Ther, vol.9, pp.139-148, 2016.

E. Liepinsh, L. L. Ilag, G. Otting, and C. F. Ibáñez, NMR structure of the death domain of the p75 neurotrophin receptor, EMBO J, vol.16, pp.4999-5005, 1997.

S. H. Lim, L. , and R. , Translational medicine in action: anti-CD20 therapy in lymphoma, J. Immunol. Baltim. Md, pp.1519-1524, 2014.

Z. Lin, J. Y. Tann, E. T. Goh, C. Kelly, K. B. Lim et al., Structural basis of death domain signaling in the p75 neurotrophin receptor, vol.4, p.11692, 2015.

M. Li-weber, Molecular mechanisms and anti-cancer aspects of the medicinal phytochemicals rocaglamides (=flavaglines), Int. J. Cancer, vol.137, pp.1791-1799, 2015.

A. Lo-cicero, P. D. Stahl, R. , and G. , Extracellular vesicles shuffling intercellular messages: for good or for bad, Curr. Opin. Cell Biol, vol.35, pp.69-77, 2015.

I. S. Lossos, Molecular pathogenesis of diffuse large B-cell lymphoma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.23, pp.6351-6357, 2005.

K. Luberg, J. Wong, C. S. Weickert, and T. Timmusk, Human TrkB gene: novel alternative transcripts, protein isoforms and expression pattern in the prefrontal cerebral cortex during postnatal development, J. Neurochem, vol.113, pp.952-964, 2010.

K. Luberg, R. Park, E. Aleksejeva, and T. Timmusk, Novel transcripts reveal a complex structure of the human TRKA gene and imply the presence of multiple protein isoforms, BMC Neurosci, vol.16, p.78, 2015.

D. M. Lucas, R. B. Edwards, G. Lozanski, D. A. West, J. D. Shin et al., The novel plant-derived agent silvestrol has B-cell selective activity in chronic lymphocytic leukemia and acute lymphoblastic leukemia in vitro and in vivo, Blood, vol.113, pp.4656-4666, 2009.

S. Lynch, S. G. Santos, E. C. Campbell, A. M. Nimmo, C. Botting et al., Novel MHC class I structures on exosomes, J. Immunol. Baltim. Md, vol.183, pp.1884-1891, 1950.

P. Macor, C. Tripodo, S. Zorzet, E. Piovan, F. Bossi et al., In vivo targeting of human neutralizing antibodies against CD55 and CD59 to lymphoma cells increases the antitumor activity of rituximab, Cancer Res, vol.67, pp.10556-10563, 2007.

P. C. Maisonpierre, L. Belluscio, S. Squinto, N. Y. Ip, M. E. Furth et al., Neurotrophin-3: a neurotrophic factor related to NGF and BDNF, Science, vol.247, pp.1446-1451, 1990.

D. G. Maloney, A. J. Grillo-lópez, C. A. White, D. Bodkin, R. J. Schilder et al., IDEC-C2B8 (Rituximab) anti-CD20 monoclonal antibody therapy in patients with relapsed low-grade non-Hodgkin's lymphoma, Blood, vol.90, pp.2188-2195, 1997.

J. Mandelbaum, G. Bhagat, H. Tang, T. Mo, M. Brahmachary et al., BLIMP1 is a tumor suppressor gene frequently disrupted in activated B cell-like diffuse large B cell lymphoma, Cancer Cell, vol.18, pp.568-579, 2010.

T. Manshouri, K. Do, X. Wang, F. J. Giles, S. M. O'brien et al., Circulating CD20 is detectable in the plasma of patients with chronic lymphocytic leukemia and is of prognostic significance, Blood, vol.101, pp.2507-2513, 2003.

D. Marchetti, R. Aucoin, J. Blust, B. Murry, and A. Greiter-wilke, p75 neurotrophin receptor functions as a survival receptor in brain-metastatic melanoma cells, J. Cell. Biochem, vol.91, pp.206-215, 2004.

M. Martelli, A. J. Ferreri, C. Agostinelli, A. Di-rocco, M. Pfreundschuh et al., Diffuse large B-cell lymphoma, Crit. Rev. Oncol. Hematol, vol.87, pp.146-171, 2013.

D. Martin-zanca, G. Mitra, L. K. Long, and M. Barbacid, Molecular characterization of the human trk oncogene, Cold Spring Harb. Symp. Quant. Biol. 51 Pt, vol.2, pp.983-992, 1986.

I. E. Mazzoni, F. A. Saïd, R. Aloyz, F. D. Miller, and D. Kaplan, Ras regulates sympathetic neuron survival by suppressing the p53-mediated cell death pathway, J. Neurosci. Off. J. Soc. Neurosci, vol.19, pp.9716-9727, 1999.

L. M. Mcgregor, B. K. Mccune, J. R. Graff, P. R. Mcdowell, K. E. Romans et al., Roles of trk family neurotrophin receptors in medullary thyroid carcinoma development and progression, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.4540-4545, 1999.

I. Melamed, H. Patel, C. Brodie, and E. W. Gelfand, Activation of Vav and Ras through the nerve growth factor and B cell receptors by different kinases, Cell. Immunol, vol.191, pp.83-89, 1999.

S. A. Melo, H. Sugimoto, J. T. O'connell, N. Kato, A. Villanueva et al., Cancer Exosomes Perform Cell-Independent MicroRNA Biogenesis and Promote Tumorigenesis, Cancer Cell, vol.26, pp.707-721, 2014.

M. Metsis, T. Timmusk, R. Allikmets, M. Saarma, P. et al., Regulatory elements and transcriptional regulation by testosterone and retinoic acid of the rat nerve growth factor receptor promoter, Gene, vol.121, pp.247-254, 1992.

D. S. Middlemas, R. A. Lindberg, and T. Hunter, trkB, a neural receptor protein-tyrosine kinase: evidence for a full-length and two truncated receptors, Mol. Cell. Biol, vol.11, pp.143-153, 1991.

Z. K. Mirnics, C. Yan, C. Portugal, T. Kim, H. U. Saragovi et al., P75 neurotrophin receptor regulates expression of neural cell adhesion molecule 1, Neurobiol. Dis, vol.20, pp.969-985, 2005.

H. Miyoshi, F. Arakawa, K. Sato, Y. Kimura, J. Kiyasu et al., Comparison of CD20 expression in B-cell lymphoma between newly diagnosed, untreated cases and those after rituximab treatment, Cancer Sci, vol.103, pp.1567-1573, 2012.

R. M. Mohammad, A. Aboukameel, S. Nabha, D. Ibrahim, A. et al., Rituximab, Cyclophosphamide, Dexamethasone (RCD) regimen induces cure in WSU-WM xenograft model and a partial remission in previously treated Waldenstrom's macroglobulinemia patient, J. Drug Target, vol.10, pp.405-410, 2002.

A. Monnereau, L. Remontet, M. Maynadié, F. Binder-foucard, A. Belot et al., Estimation nationale de l'incidence des cancers en France entre 1980 et 2012. Partie 2Hémopathies malignes

S. A. Moodie, B. M. Willumsen, M. J. Weber, and A. Wolfman, Complexes of Ras.GTP with Raf-1 and mitogen-activated protein kinase kinase, Science, vol.260, pp.1658-1661, 1993.

G. Morgan, M. Vornanen, J. Puitinen, A. Naukkarinen, H. Brincker et al., Changing trends in the incidence of nonHodgkin's lymphoma in Europe, Biomed Study Group. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, vol.8, issue.2, pp.49-54, 1997.

R. D. Morin, N. A. Johnson, T. M. Severson, A. J. Mungall, J. An et al., Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin, Nat. Genet, vol.42, pp.181-185, 2010.

A. Morotti, S. Mila, P. Accornero, E. Tagliabue, P. et al., K252a inhibits the oncogenic properties of Met, the HGF receptor, Oncogene, vol.21, pp.4885-4893, 2002.

J. J. Muris, C. J. Meijer, W. Vos, J. H. Van-krieken, N. M. Jiwa et al., Immunohistochemical profiling based on Bcl-2, CD10 and MUM1 expression improves risk stratification in patients with primary nodal diffuse large B cell lymphoma, J. Pathol, vol.208, pp.714-723, 2006.

C. Nabhan, K. , and N. E. , The emerging role of ofatumumab in the treatment of chronic lymphocytic leukemia, Clin. Med. Insights Oncol, vol.5, pp.45-53, 2011.

K. D. Nadezhdin, I. García-carpio, S. A. Goncharuk, K. S. Mineev, A. S. Arseniev et al., Structural Basis of p75 Transmembrane Domain Dimerization, J. Biol. Chem, vol.291, pp.12346-12357, 2016.

L. M. Nadler, S. J. Korsmeyer, K. C. Anderson, A. W. Boyd, B. Slaughenhoupt et al., B cell origin of non-T cell acute lymphoblastic leukemia. A model for discrete stages of neoplastic and normal pre-B cell differentiation, J. Clin. Invest, vol.74, pp.332-340, 1984.

R. Nagasubramanian, J. Wei, P. Gordon, J. C. Rastatter, M. C. Cox et al., Infantile Fibrosarcoma With NTRK3-ETV6 Fusion Successfully Treated With the Tropomyosin-Related Kinase Inhibitor LOXO-101. Pediatr, Blood Cancer, vol.63, pp.1468-1470, 2016.

A. Nakagawara, X. G. Liu, N. Ikegaki, P. S. White, D. J. Yamashiro et al., Cloning and chromosomal localization of the human TRK-B tyrosine kinase receptor gene (NTRK2), Genomics, vol.25, pp.538-546, 1995.

Y. Natkunam, P. Farinha, E. D. Hsi, C. P. Hans, R. Tibshirani et al., LMO2 protein expression predicts survival in patients with diffuse large B-cell lymphoma treated with anthracycline-based chemotherapy with and without rituximab, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.26, pp.447-454, 2008.

P. Naveilhan, I. Neveu, C. Baudet, H. Funakoshi, D. Wion et al., 1,25Dihydroxyvitamin D3 regulates the expression of the low-affinity neurotrophin receptor, Brain Res. Mol. Brain Res, vol.41, pp.259-268, 1996.

M. S. Nielsen, The sortilin cytoplasmic tail conveys Golgi-endosome transport and binds the VHS domain of the GGA2 sorting protein, EMBO J, vol.20, pp.2180-2190, 2001.

S. K. Nilsson, S. Christensen, M. K. Raarup, R. O. Ryan, M. S. Nielsen et al., Endocytosis of apolipoprotein A-V by members of the low density lipoprotein receptor and the VPS10p domain receptor families, J. Biol. Chem, vol.283, pp.25920-25927, 2008.

E. N. Nolte-'t-hoen, E. J. Van-der-vlist, M. Aalberts, H. C. Mertens, B. J. Bosch et al., Quantitative and qualitative flow cytometric analysis of nanosized cell-derived membrane vesicles, Nanomedicine Nanotechnol. Biol. Med, vol.8, pp.712-720, 2012.

A. C. Nyborg, T. B. Ladd, C. W. Zwizinski, J. J. Lah, and T. E. Golde, Sortilin, SorCS1b, and SorLA Vps10p sorting receptors, are novel ?-secretase substrates, Mol. Neurodegener, vol.1, p.3, 2006.

S. H. Nye, S. P. Squinto, D. J. Glass, T. N. Stitt, P. Hantzopoulos et al., K-252a and staurosporine selectively block autophosphorylation of neurotrophin receptors and neurotrophin-mediated responses, Mol. Biol. Cell, vol.3, pp.677-686, 1992.

A. Nykjaer and T. E. Willnow, Sortilin: a receptor to regulate neuronal viability and function, Trends Neurosci, vol.35, pp.261-270, 2012.

A. Nykjaer, R. Lee, K. K. Teng, P. Jansen, P. Madsen et al., Sortilin is essential for proNGF-induced neuronal cell death, Nature, vol.427, pp.843-848, 2004.

A. Nykjaer, T. E. Willnow, and C. M. Petersen, p75NTR-live or let die, Curr. Opin. Neurobiol, vol.15, pp.49-57, 2005.

S. Odate, H. Onishi, K. Nakamura, M. Kojima, A. Uchiyama et al., Tropomyosin-related kinase B inhibitor has potential for tumor regression and relapse prevention in pulmonary large cell neuroendocrine carcinoma, Anticancer Res, vol.33, pp.3699-3703, 2013.

E. Oflazoglu and L. P. Audoly, Evolution of anti-CD20 monoclonal antibody therapeutics in oncology, vol.2, pp.14-19, 2010.

Y. Ogawa, M. Kanai-azuma, Y. Akimoto, H. Kawakami, Y. et al., Exosome-like vesicles with dipeptidyl peptidase IV in human saliva, Biol. Pharm. Bull, vol.31, pp.1059-1062, 2008.

T. Okamoto, A. Schlegel, P. E. Scherer, and M. P. Lisanti, Caveolins, a family of scaffolding proteins for organizing "preassembled signaling complexes" at the plasma membrane, J. Biol. Chem, vol.273, pp.5419-5422, 1998.

Y. Okugawa, K. Tanaka, Y. Inoue, M. Kawamura, A. Kawamoto et al., Brain-derived neurotrophic factor/tropomyosin-related kinase B pathway in gastric cancer, Br. J. Cancer, vol.108, pp.121-130, 2013.

M. E. Palko, V. Coppola, and L. Tessarollo, Evidence for a role of truncated trkC receptor isoforms in mouse development, J. Neurosci. Off. J. Soc. Neurosci, vol.19, pp.775-782, 1999.

P. T. Pang, H. K. Teng, E. Zaitsev, N. T. Woo, K. Sakata et al., Cleavage of proBDNF by tPA/plasmin is essential for long-term hippocampal plasticity, Science, vol.306, pp.487-491, 2004.

Y. H. Park, S. K. Sohn, J. G. Kim, M. Lee, H. S. Song et al., Interaction between BCL2 and interleukin-10 gene polymorphisms alter outcomes of diffuse large B-cell lymphoma following rituximab plus CHOP chemotherapy, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.15, pp.2107-2115, 2009.

L. Pasqualucci, M. Compagno, J. Houldsworth, S. Monti, A. Grunn et al., Inactivation of the PRDM1/BLIMP1 gene in diffuse large B cell lymphoma, J. Exp. Med, vol.203, pp.311-317, 2006.

F. Passiglia, R. Caparica, E. Giovannetti, M. Giallombardo, A. Listi et al., The potential of neurotrophic tyrosine kinase (NTRK) inhibitors for treating lung cancer, Expert Opin. Investig. Drugs, vol.25, pp.385-392, 2016.

M. R. Patel, T. M. Bauer, S. V. Liu, A. Drilon, J. Wheler et al., STARTRK-1: Phase 1/2a study of entrectinib, an oral Pan-Trk, ROS1, and ALK inhibitor, in patients with advanced solid tumors with relevant molecular alterations, Age, vol.1, p.10, 2015.

R. N. Pearse, S. L. Swendeman, Y. Li, D. Rafii, and B. L. Hempstead, A neurotrophin axis in myeloma: TrkB and BDNF promote tumor-cell survival, Blood, vol.105, pp.4429-4436, 2005.

R. J. Petrie and J. P. Deans, Colocalization of the B cell receptor and CD20 followed by activation-dependent dissociation in distinct lipid rafts, J. Immunol. Baltim. Md, vol.169, pp.2886-2891, 1950.

M. Pfeifer, M. Grau, D. Lenze, S. Wenzel, A. Wolf et al., PTEN loss defines a PI3K/AKT pathway-dependent germinal center subtype of diffuse large B-cell lymphoma, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.12420-12425, 2013.

M. Pfreundschuh, L. Trümper, A. Osterborg, R. Pettengell, M. Trneny et al., CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group, Lancet Oncol, vol.7, pp.379-391, 2006.

T. Pisitkun, R. Shen, and M. A. Knepper, Identification and proteomic profiling of exosomes in human urine, Proc. Natl. Acad. Sci. U. S. A, vol.101, pp.13368-13373, 2004.

G. Polier, J. Neumann, F. Thuaud, N. Ribeiro, C. Gelhaus et al., The natural anticancer compounds rocaglamides inhibit the Raf-MEK-ERK pathway by targeting prohibitin 1 and 2, Chem. Biol, vol.19, pp.1093-1104, 2012.

M. J. Polyak, S. H. Tailor, and J. P. Deans, Identification of a cytoplasmic region of CD20 required for its redistribution to a detergent-insoluble membrane compartment, J. Immunol. Baltim. Md, vol.161, pp.3242-3248, 1950.

M. J. Polyak, H. Li, N. Shariat, and J. P. Deans, CD20 homo-oligomers physically associate with the B cell antigen receptor. Dissociation upon receptor engagement and recruitment of phosphoproteins and calmodulin-binding proteins, J. Biol. Chem, vol.283, pp.18545-18552, 2008.

J. C. Powell, C. Twomey, R. Jain, and J. V. Mccarthy, Association between Presenilin-1 and TRAF6 modulates regulated intramembrane proteolysis of the p75NTR neurotrophin receptor, J. Neurochem, vol.108, pp.216-230, 2009.

C. G. Proud, Mnks, eIF4E phosphorylation and cancer, Biochim. Biophys. Acta, vol.1849, pp.766-773, 2015.

G. Rabinowits, C. Gerçel-taylor, J. M. Day, D. D. Taylor, and G. H. Kloecker, Exosomal microRNA: a diagnostic marker for lung cancer, Clin. Lung Cancer, vol.10, pp.42-46, 2009.

M. J. Radeke, T. P. Misko, C. Hsu, L. A. Herzenberg, and E. M. Shooter, Gene transfer and molecular cloning of the rat nerve growth factor receptor, Nature, vol.325, pp.593-597, 1987.

R. Rahmé, E. Benayoun, C. Pautas, C. Cordonnier, O. Wagner-ballon et al., Treatment with 5-azacytidin upregulates the expression of CD20 in CD20-negative B cell acute lymphoblastic leukemia: a case report, Exp. Hematol, vol.41, pp.505-507, 2013.

K. Rajalingam, C. Wunder, V. Brinkmann, Y. Churin, M. Hekman et al., Prohibitin is required for Ras-induced Raf-MEK-ERK activation and epithelial cell migration, Nat. Cell Biol, vol.7, pp.837-843, 2005.

G. Raposo, H. W. Nijman, W. Stoorvogel, R. Liejendekker, C. V. Harding et al., B lymphocytes secrete antigen-presenting vesicles, J. Exp. Med, vol.183, pp.1161-1172, 1996.

M. Record, C. Subra, S. Silvente-poirot, and M. Poirot, Exosomes as intercellular signalosomes and pharmacological effectors, Biochem. Pharmacol, vol.81, pp.1171-1182, 2011.
DOI : 10.1016/j.bcp.2011.02.011

URL : https://hal.archives-ouvertes.fr/inserm-00743451

M. Record, K. Carayon, M. Poirot, and S. Silvente-poirot, Exosomes as new vesicular lipid transporters involved in cell-cell communication and various pathophysiologies, Biochim. Biophys. Acta, vol.1841, pp.108-120, 2014.
DOI : 10.1016/j.bbalip.2013.10.004

URL : https://hal.archives-ouvertes.fr/inserm-00968935

L. F. Reichardt, Neurotrophin-regulated signalling pathways, Philos. Trans. R. Soc. Lond. B. Biol. Sci, vol.361, pp.1545-1564, 2006.
DOI : 10.1098/rstb.2006.1894

URL : http://europepmc.org/articles/pmc1664664?pdf=render

Y. Renaudineau, V. Devauchelle-pensec, C. Hanrotel, J. Pers, A. Saraux et al., Monoclonal anti-CD20 antibodies: mechanisms of action and monitoring of biological effects, Jt. Bone Spine Rev. Rhum, vol.76, pp.458-463, 2009.
DOI : 10.1016/j.jbspin.2009.03.010

A. R. Rezvani and D. G. Maloney, Rituximab resistance, Best Pract. Res. Clin. Haematol, vol.24, pp.203-216, 2011.

N. Ribeiro, F. Thuaud, C. Nebigil, and L. Désaubry, Recent advances in the biology and chemistry of the flavaglines, Bioorg. Med. Chem, vol.20, pp.1857-1864, 2012.

A. Riccio, S. Ahn, C. M. Davenport, J. A. Blendy, and D. D. Ginty, Mediation by a CREB family transcription factor of NGF-dependent survival of sympathetic neurons, Science, vol.286, pp.2358-2361, 1999.

P. D. Robbins and A. E. Morelli, Regulation of immune responses by extracellular vesicles, Nat. Rev. Immunol, vol.14, pp.195-208, 2014.

F. Robert, M. Carrier, S. Rawe, S. Chen, S. Lowe et al., Altering Chemosensitivity by Modulating Translation Elongation, PLoS ONE, vol.4, 2009.
DOI : 10.1371/journal.pone.0005428

URL : https://doi.org/10.1371/journal.pone.0005428

A. Rodriguez-tébar, G. Dechant, B. , and Y. A. , Binding of brain-derived neurotrophic factor to the nerve growth factor receptor, Neuron, vol.4, pp.487-492, 1990.

A. Rodríguez-tébar, G. Dechant, R. Götz, B. , and Y. A. , Binding of neurotrophin-3 to its neuronal receptors and interactions with nerve growth factor and brain-derived neurotrophic factor, EMBO J, vol.11, pp.917-922, 1992.

A. Rosenwald, G. Wright, W. C. Chan, J. M. Connors, E. Campo et al., The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma, N. Engl. J. Med, vol.346, pp.1937-1947, 2002.

P. Rosini, G. De-chiara, M. Lucibello, E. Garaci, F. Cozzolino et al., NGF withdrawal induces apoptosis in CESS B cell line through p38 MAPK activation and Bcl-2 phosphorylation, Biochem. Biophys. Res. Commun, vol.278, pp.753-759, 2000.
DOI : 10.1006/bbrc.2000.3871

URL : https://flore.unifi.it/bitstream/2158/206720/1/206720.pdf

P. P. Roux and P. A. Barker, Neurotrophin signaling through the p75 neurotrophin receptor, Prog. Neurobiol, vol.67, pp.203-233, 2002.
DOI : 10.1016/s0301-0082(02)00016-3

M. Rozakis-adcock, J. Mcglade, G. Mbamalu, G. Pelicci, R. Daly et al., Association of the Shc and Grb2/Sem5 SH2-containing proteins is implicated in activation of the Ras pathway by tyrosine kinases, Nature, vol.360, pp.689-692, 1992.

R. Safaei, Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drugresistant human ovarian carcinoma cells, Mol. Cancer Ther, vol.4, pp.1595-1604, 2005.
DOI : 10.1158/1535-7163.mct-05-0102

URL : http://mct.aacrjournals.org/content/4/10/1595.full.pdf

A. H. Salehi, S. Xanthoudakis, and P. A. Barker, NRAGE, a p75 neurotrophin receptorinteracting protein, induces caspase activation and cell death through a JNK-dependent mitochondrial pathway, J. Biol. Chem, vol.277, pp.48043-48050, 2002.
DOI : 10.1074/jbc.m205324200

URL : http://www.jbc.org/content/277/50/48043.full.pdf

D. Schack, E. Casademunt, R. Schweigreiter, M. Meyer, M. Bibel et al., Complete ablation of the neurotrophin receptor p75NTR causes defects both in the nervous and the vascular system, Nat. Neurosci, vol.4, pp.977-978, 2001.

A. Schenone, J. S. Gill, D. A. Zacharias, and A. J. Windebank, Expression of high-and lowaffinity neurotrophin receptors on human transformed B lymphocytes, J. Neuroimmunol, vol.64, pp.141-149, 1996.
DOI : 10.1016/0165-5728(95)00162-x

C. Schliemann, A. Palumbo, K. Zuberbühler, A. Villa, M. Kaspar et al., Complete eradication of human B-cell lymphoma xenografts using rituximab in combination with the immunocytokine L19-IL2, Blood, vol.113, pp.2275-2283, 2009.

B. Schuhmann, A. Dietrich, S. Sel, C. Hahn, M. Klingenspor et al., A role for brain-derived neurotrophic factor in B cell development, J. Neuroimmunol, vol.163, pp.15-23, 2005.

A. Scuto, M. Kujawski, C. Kowolik, L. Krymskaya, L. Wang et al., STAT3 inhibition is a therapeutic strategy for ABC-like diffuse large B-cell lymphoma, Cancer Res, vol.71, pp.3182-3188, 2011.

A. Sehgal, N. Patil, and M. Chao, A constitutive promoter directs expression of the nerve growth factor receptor gene, Mol. Cell. Biol, vol.8, pp.3160-3167, 1988.

L. H. Sehn, Paramount prognostic factors that guide therapeutic strategies in diffuse large B-cell lymphoma, Hematol. Educ. Program Am. Soc. Hematol. Am. Soc. Hematol. Educ. Program, pp.402-409, 2012.

L. H. Sehn and R. D. Gascoyne, Diffuse large B-cell lymphoma: optimizing outcome in the context of clinical and biologic heterogeneity, Blood, vol.125, pp.22-32, 2015.

N. G. Seidah, S. Benjannet, S. Pareek, D. Savaria, J. Hamelin et al., Cellular processing of the nerve growth factor precursor by the mammalian pro-protein convertases, Biochem. J, vol.314, pp.951-960, 1996.

D. Shan, J. A. Ledbetter, and O. W. Press, Signaling events involved in anti-CD20-induced apoptosis of malignant human B cells, Cancer Immunol. Immunother. CII, vol.48, pp.673-683, 2000.

D. L. Shelton, J. Sutherland, J. Gripp, T. Camerato, M. P. Armanini et al., Human trks: molecular cloning, tissue distribution, and expression of extracellular domain immunoadhesins, J. Neurosci. Off. J. Soc. Neurosci, vol.15, pp.477-491, 1995.

M. Sheng and C. Sala, PDZ domains and the organization of supramolecular complexes, Annu. Rev. Neurosci, vol.24, pp.1-29, 2001.

M. A. Shipp, K. N. Ross, P. Tamayo, A. P. Weng, J. L. Kutok et al., Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning, Nat. Med, vol.8, pp.68-74, 2002.

S. Siberil, C. Dutertre, C. Boix, and J. Teillaud, , 2005.

, Transfus. Clin. Biol. J. Soc. Francaise Transfus. Sang, vol.12, pp.114-122

V. Singh, D. Gupta, A. , and A. , Development of Novel Anti-Cd20 Monoclonal Antibodies and Modulation in Cd20 Levels on Cell Surface: Looking to Improve Immunotherapy Response, J. Cancer Sci. Ther, vol.7, pp.347-358, 2015.

S. Skeldal, D. Matusica, A. Nykjaer, and E. J. Coulson, Proteolytic processing of the p75 neurotrophin receptor: A prerequisite for signalling?: Neuronal life, growth and death signalling are crucially regulated by intra-membrane proteolysis and trafficking of p75(NTR), BioEssays News Rev. Mol. Cell. Dev. Biol, vol.33, pp.614-625, 2011.

S. Skeldal, A. M. Sykes, S. Glerup, D. Matusica, N. Palstra et al., Mapping of the Interaction Site between Sortilin and the p75 Neurotrophin Receptor Reveals a Regulatory Role for the Sortilin Intracellular Domain in p75 Neurotrophin Receptor Shedding and Apoptosis, J. Biol. Chem, vol.287, pp.43798-43809, 2012.

M. S?abicki, K. S. Lee, A. Jethwa, L. Sellner, F. Sacco et al., Dissection of CD20 regulation in lymphoma using RNAi. Leukemia, 2016.

M. R. Smith, Rituximab (monoclonal anti-CD20 antibody): mechanisms of action and resistance, Oncogene, vol.22, pp.7359-7368, 2003.

L. F. Sniderhan, T. M. Garcia-bates, M. Burgart, S. H. Bernstein, R. P. Phipps et al., Neurotrophin signaling through tropomyosin receptor kinases contributes to survival and proliferation of non-Hodgkin lymphoma, Exp. Hematol, vol.37, pp.1295-1309, 2009.

P. Sondermann and D. E. Szymkowski, Harnessing Fc receptor biology in the design of therapeutic antibodies, Curr. Opin. Immunol, vol.40, pp.78-87, 2016.

L. M. Staudt and R. B. Gartenhaus, Promising personalized therapeutic options for diffuse large B-cell Lymphoma Subtypes with oncogene addictions, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.18, pp.4538-4548, 2010.

S. F. De-stgroth and D. Scheidegger, Production of monoclonal antibodies: strategy and tactics, J. Immunol. Methods, vol.35, pp.1-21, 1980.

P. Stoilov, E. Castren, and S. Stamm, Analysis of the human TrkB gene genomic organization reveals novel TrkB isoforms, unusual gene length, and splicing mechanism, Biochem. Biophys. Res. Commun, vol.290, pp.1054-1065, 2002.

J. M. Street, P. E. Barran, C. L. Mackay, S. Weidt, C. Balmforth et al., Identification and proteomic profiling of exosomes in human cerebrospinal fluid, J. Transl. Med, vol.10, p.5, 2012.

S. Stuffers, C. Sem-wegner, H. Stenmark, and A. Brech, Multivesicular endosome biogenesis in the absence of ESCRTs, Traffic Cph. Den, vol.10, pp.925-937, 2009.

C. Subra, K. Laulagnier, B. Perret, and M. Record, Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies, Biochimie, vol.89, pp.205-212, 2007.

T. Sugimoto, A. Tomita, J. Hiraga, K. Shimada, H. Kiyoi et al., Escape mechanisms from antibody therapy to lymphoma cells: downregulation of CD20 mRNA by recruitment of the HDAC complex and not by DNA methylation, Biochem. Biophys. Res. Commun, vol.390, pp.48-53, 2009.

E. Suzuki, K. Umezawa, and B. Bonavida, Rituximab inhibits the constitutively activated PI3K-Akt pathway in B-NHL cell lines: involvement in chemosensitization to drug-induced apoptosis, Oncogene, vol.26, pp.6184-6193, 2007.

S. H. Swerdlow, E. Campo, and N. L. Harris, WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues, 2008.

S. H. Swerdlow, E. Campo, S. A. Pileri, N. L. Harris, H. Stein et al., The 2016 revision of the World Health Organization classification of lymphoid neoplasms, Blood, vol.127, pp.2375-2390, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01800015

A. Tacconelli, A. R. Farina, L. Cappabianca, G. Desantis, A. Tessitore et al., TrkA alternative splicing: a regulated tumorpromoting switch in human neuroblastoma, Cancer Cell, vol.6, pp.347-360, 2004.

M. Takizawa, H. Tolarová, Z. Li, W. Dubois, S. Lim et al., AID expression levels determine the extent of cMyc oncogenic translocations and the incidence of B cell tumor development, J. Exp. Med, vol.205, pp.1949-1957, 2008.

W. Tam, M. Gomez, A. Chadburn, J. W. Lee, W. C. Chan et al., Mutational analysis of PRDM1 indicates a tumor-suppressor role in diffuse large B-cell lymphomas, Blood, vol.107, pp.4090-4100, 2006.

M. Taniuchi, E. M. Johnson, P. J. Roach, L. , and J. C. , Phosphorylation of nerve growth factor receptor proteins in sympathetic neurons and PC12 cells. In vitro phosphorylation by the cAMP-independent protein kinase FA/GSK-3, J. Biol. Chem, vol.261, pp.13342-13349, 1986.

P. Tapley, F. Lamballe, and M. Barbacid, K252a is a selective inhibitor of the tyrosine protein kinase activity of the trk family of oncogenes and neurotrophin receptors, Oncogene, vol.7, pp.371-381, 1992.

T. F. Tedder and P. Engel, CD20: a regulator of cell-cycle progression of B lymphocytes, Immunol. Today, vol.15, pp.450-454, 1994.

T. F. Tedder, G. Klejman, S. F. Schlossman, and H. Saito, Structure of the gene encoding the human B lymphocyte differentiation antigen CD20 (B1), J. Immunol. Baltim. Md, vol.142, pp.2560-2568, 1950.

J. L. Teeling, W. J. Mackus, L. J. Wiegman, J. H. Van-den-brakel, S. A. Beers et al., The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20, J. Immunol. Baltim. Md, vol.177, pp.362-371, 1950.

K. K. Teng and B. L. Hempstead, Neurotrophins and their receptors: signaling trios in complex biological systems, Cell. Mol. Life Sci. CMLS, vol.61, pp.35-48, 2004.

H. K. Teng, K. K. Teng, R. Lee, S. Wright, S. Tevar et al., ProBDNF induces neuronal apoptosis via activation of a receptor complex of p75NTR and sortilin, J. Neurosci. Off. J. Soc. Neurosci, vol.25, pp.5455-5463, 2005.

L. Tessarollo, Pleiotropic functions of neurotrophins in development, Cytokine Growth Factor Rev, vol.9, pp.125-137, 1998.

M. Testoni, E. Zucca, K. H. Young, and F. Bertoni, Genetic lesions in diffuse large B-cell lymphomas, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.26, pp.1069-1080, 2015.

C. Théry, L. Duban, E. Segura, P. Véron, O. Lantz et al., Indirect activation of naïve CD4+ T cells by dendritic cell-derived exosomes, Nat. Immunol, vol.3, pp.1156-1162, 2002.

C. Théry, M. Ostrowski, and E. Segura, Membrane vesicles as conveyors of immune responses, Nat. Rev. Immunol, vol.9, pp.581-593, 2009.

C. J. Thiele, Z. Li, and A. E. Mckee, On Trk-the TrkB signal transduction pathway is an increasingly important target in cancer biology, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.15, pp.5962-5967, 2009.

C. A. Thompson, A. Purushothaman, V. C. Ramani, I. Vlodavsky, and R. D. Sanderson, Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes, J. Biol. Chem, vol.288, pp.10093-10099, 2013.

L. W. Thorpe, K. Werrbach-perez, and J. R. Perez-polo, Effects of nerve growth factor on the expression of interleukin-2 receptors on cultured human lymphocytes, Ann. N. Y. Acad. Sci, vol.496, pp.310-311, 1987.

F. Thuaud, Y. Bernard, G. Türkeri, R. Dirr, G. Aubert et al., Synthetic analogue of rocaglaol displays a potent and selective cytotoxicity in cancer cells: involvement of apoptosis inducing factor and caspase-12, J. Med. Chem, vol.52, pp.5176-5187, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00467437

H. Tilly, U. Vitolo, J. Walewski, M. G. Da-silva, O. Shpilberg et al., Diffuse large B-cell lymphoma (DLBCL): ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO, vol.23, pp.78-82, 2012.

M. Torcia, L. Bracci-laudiero, M. Lucibello, L. Nencioni, D. Labardi et al., Nerve growth factor is an autocrine survival factor for memory B lymphocytes, Cell, vol.85, pp.345-356, 1996.

K. Trajkovic, C. Hsu, S. Chiantia, L. Rajendran, D. Wenzel et al., Ceramide triggers budding of exosome vesicles into multivesicular endosomes, Science, vol.319, pp.1244-1247, 2008.

E. G. Trams, C. J. Lauter, N. Salem, and U. Heine, Exfoliation of membrane ecto-enzymes in the form of micro-vesicles, Biochim. Biophys. Acta, vol.645, pp.63-70, 1981.

G. Trinchieri, Interleukin-12 and the regulation of innate resistance and adaptive immunity, Nat. Rev. Immunol, vol.3, pp.133-146, 2003.

Y. Tsutsumi, H. Ohigashi, S. Ito, S. Shiratori, and T. Teshima, 5-Azacytidine partially restores CD20 expression in follicular lymphoma that lost CD20 expression after rituximab treatment: a case report, J. Med. Case Reports, vol.10, p.27, 2016.

J. Uchida, Y. Lee, M. Hasegawa, Y. Liang, A. Bradney et al., Mouse CD20 expression and function, Int. Immunol, vol.16, pp.119-129, 2004.

C. K. Underwood, K. Reid, L. M. May, P. F. Bartlett, and E. J. Coulson, Palmitoylation of the C-terminal fragment of p75(NTR) regulates death signaling and is required for subsequent cleavage by gamma-secretase, Mol. Cell. Neurosci, vol.37, pp.346-358, 2008.

T. L. Unruh, H. Li, C. M. Mutch, N. Shariat, L. Grigoriou et al., Cholesterol depletion inhibits src family kinase-dependent calcium mobilization and apoptosis induced by rituximab crosslinking, Immunology, vol.116, pp.223-232, 2005.

A. R. Vaillant, I. Mazzoni, C. Tudan, M. Boudreau, D. R. Kaplan et al., Depolarization and neurotrophins converge on the phosphatidylinositol 3-kinase-Akt pathway to synergistically regulate neuronal survival, J. Cell Biol, vol.146, pp.955-966, 1999.

A. Valent, G. Danglot, and A. Bernheim, Mapping of the tyrosine kinase receptors trkA (NTRK1), trkB (NTRK2) and trkC(NTRK3) to human chromosomes 1q22, 9q22 and 15q25 by fluorescence in situ hybridization, Eur. J. Hum. Genet. EJHG, vol.5, pp.102-104, 1997.

M. A. Valentine, T. Cotner, L. Gaur, R. Torres, C. et al., Expression of the human Bcell surface protein CD20: alteration by phorbol 12-myristate 13-acetate, Proc. Natl. Acad. Sci. U. S. A, vol.84, pp.8085-8089, 1987.

E. Vanhecke, E. Adriaenssens, S. Verbeke, S. Meignan, E. Germain et al., Brain-derived neurotrophic factor and neurotrophin-4/5 are expressed in breast cancer and can be targeted to inhibit tumor cell survival, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.17, pp.1741-1752, 2011.

P. Venugopal, S. Sivaraman, X. K. Huang, J. Nayini, S. A. Gregory et al., Effects of cytokines on CD20 antigen expression on tumor cells from patients with chronic lymphocytic leukemia, Leuk. Res, vol.24, pp.411-415, 2000.

P. Viatour, M. Bentires-alj, A. Chariot, V. Deregowski, L. De-leval et al., NF-kappa B2/p100 induces Bcl-2 expression, Leukemia, vol.17, pp.1349-1356, 2003.

M. Vilar, I. Charalampopoulos, R. S. Kenchappa, A. Reversi, J. M. Klos-applequist et al., Ligand-independent signaling by disulfide-crosslinked dimers of the p75 neurotrophin receptor, J. Cell Sci, vol.122, pp.3351-3357, 2009.

M. Volosin, W. Song, R. D. Almeida, D. R. Kaplan, B. L. Hempstead et al., Interaction of survival and death signaling in basal forebrain neurons: roles of neurotrophins and proneurotrophins, J. Neurosci. Off. J. Soc. Neurosci, vol.26, pp.7756-7766, 2006.

C. A. Walshe, S. A. Beers, R. R. French, C. H. Chan, P. W. Johnson et al., Induction of cytosolic calcium flux by CD20 is dependent upon B Cell antigen receptor signaling, J. Biol. Chem, vol.283, pp.16971-16984, 2008.

M. Walton, A. M. Woodgate, A. Muravlev, R. Xu, M. J. During et al., CREB phosphorylation promotes nerve cell survival, J. Neurochem, vol.73, pp.1836-1842, 1999.

W. Wang and M. T. Lotze, Good things come in small packages: exosomes, immunity and cancer, Cancer Gene Ther, vol.21, pp.139-141, 2014.

L. Wang, J. J. Rahn, X. Lun, B. Sun, J. J. Kelly et al., Gamma-secretase represents a therapeutic target for the treatment of invasive glioma mediated by the p75 neurotrophin receptor, PLoS Biol, vol.6, p.289, 2008.

W. Wang, J. Chen, G. , and X. , The role of nerve growth factor and its receptors in tumorigenesis and cancer pain, Biosci. Trends, vol.8, pp.68-74, 2014.

Y. M. Wang, M. L. Seibenhener, M. L. Vandenplas, and M. W. Wooten, Atypical PKC zeta is activated by ceramide, resulting in coactivation of NF-kappaB/JNK kinase and cell survival, J. Neurosci. Res, vol.55, pp.293-302, 1999.

A. T. Weeraratna, S. L. Dalrymple, J. C. Lamb, S. R. Denmeade, S. Miknyoczki et al., Pan-trk inhibition decreases metastasis and enhances host survival in experimental models as a result of its selective induction of apoptosis of prostate cancer cells, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.7, pp.2237-2245, 2001.

G. J. Weiner, Rituximab: mechanism of action, Semin. Hematol, vol.47, pp.115-123, 2010.

L. M. Weiner, R. Surana, W. , and S. , Monoclonal antibodies: versatile platforms for cancer immunotherapy, Nat. Rev. Immunol, vol.10, pp.317-327, 2010.

U. B. Westergaard, E. S. Sørensen, G. Hermey, M. S. Nielsen, A. Nykjaer et al., Functional organization of the sortilin Vps10p domain, J. Biol. Chem, vol.279, pp.50221-50229, 2004.

J. Whitfield, S. J. Neame, L. Paquet, O. Bernard, and J. Ham, Dominant-negative c-Jun promotes neuronal survival by reducing BIM expression and inhibiting mitochondrial cytochrome c release, Neuron, vol.29, pp.629-643, 2001.

E. J. Wing, D. M. Magee, T. L. Whiteside, S. S. Kaplan, and R. K. Shadduck, Recombinant human granulocyte/macrophage colony-stimulating factor enhances monocyte cytotoxicity and secretion of tumor necrosis factor alpha and interferon in cancer patients, Blood, vol.73, pp.643-646, 1989.

M. Winiarska, K. Bojarczuk, B. Pyrzynska, J. Bil, M. Siernicka et al., Inhibitors of SRC kinases impair antitumor activity of anti-CD20 monoclonal antibodies, vol.6, pp.1300-1313, 2014.

W. Wojciechowski, H. Li, S. Marshall, C. Dell'agnola, and I. Espinoza-delgado, Enhanced expression of CD20 in human tumor B cells is controlled through ERK-dependent mechanisms, J. Immunol. Baltim. Md, vol.174, pp.7859-7868, 1950.

J. Wong and B. Garner, Evidence that truncated TrkB isoform, TrkB-Shc can regulate phosphorylated TrkB protein levels, Biochem. Biophys. Res. Commun, vol.420, pp.331-335, 2012.

M. W. Wooten, G. Zhou, M. L. Seibenhener, C. , and E. S. , A role for zeta protein kinase C in nerve growth factor-induced differentiation of PC12 cells, Cell Growth Differ. Mol. Biol. J. Am. Assoc. Cancer Res, vol.5, pp.395-403, 1994.

M. W. Wooten, M. L. Seibenhener, K. B. Neidigh, and M. L. Vandenplas, Mapping of atypical protein kinase C within the nerve growth factor signaling cascade: relationship to differentiation and survival of PC12 cells, Mol. Cell. Biol, vol.20, pp.4494-4504, 2000.

M. W. Wooten, M. L. Vandenplas, M. L. Seibenhener, T. Geetha, and M. T. Diaz-meco, Nerve growth factor stimulates multisite tyrosine phosphorylation and activation of the atypical protein kinase C's via a src kinase pathway, Mol. Cell. Biol, vol.21, pp.8414-8427, 2001.

G. Wright, B. Tan, A. Rosenwald, E. H. Hurt, A. Wiestner et al., A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma, Proc. Natl. Acad. Sci. U. S. A, vol.100, pp.9991-9996, 2003.

R. Wubbolts, R. S. Leckie, P. T. Veenhuizen, G. Schwarzmann, W. Möbius et al., Proteomic and biochemical analyses of human B cell-derived exosomes. Potential implications for their function and multivesicular body formation, J. Biol. Chem, vol.278, pp.10963-10972, 2003.

D. Xia, W. Li, L. Zhang, H. Qian, S. Yao et al., RNA interference-mediated knockdown of brain-derived neurotrophic factor (BDNF) promotes cell cycle arrest and apoptosis in B-cell lymphoma cells, Neoplasma, vol.61, pp.523-532, 2014.

M. Yamada, H. Ohnishi, S. Sano, A. Nakatani, T. Ikeuchi et al., Insulin receptor substrate (IRS)-1 and IRS-2 are tyrosine-phosphorylated and associated with phosphatidylinositol 3-kinase in response to brain-derived neurotrophic factor in cultured cerebral cortical neurons, J. Biol. Chem, vol.272, pp.30334-30339, 1997.

H. Yan and M. V. Chao, Disruption of cysteine-rich repeats of the p75 nerve growth factor receptor leads to loss of ligand binding, J. Biol. Chem, vol.266, pp.12099-12104, 1991.

X. Yang, T. A. Martin, and W. G. Jiang, Biological influence of brain-derived neurotrophic factor (BDNF) on colon cancer cells, Exp. Ther. Med, vol.6, pp.1475-1481, 2013.

B. H. Ye, G. Cattoretti, Q. Shen, J. Zhang, N. Hawe et al., The BCL-6 proto-oncogene controls germinal-centre formation and Th2-type inflammation, Nat. Genet, vol.16, pp.161-170, 1997.

X. Ye, P. Mehlen, S. Rabizadeh, T. Vanarsdale, H. Zhang et al., TRAF family proteins interact with the common neurotrophin receptor and modulate apoptosis induction, J. Biol. Chem, vol.274, pp.30202-30208, 1999.

K. H. Young, K. Leroy, M. B. Møller, G. W. Colleoni, M. Sánchez-beato et al., Structural profiles of TP53 gene mutations predict clinical outcome in diffuse large B-cell lymphoma: an international collaborative study, Blood, vol.112, pp.3088-3098, 2008.

P. E. Zage, T. C. Graham, L. Zeng, W. Fang, C. Pien et al., The selective Trk inhibitor AZ623 inhibits brain-derived neurotrophic factor-mediated neuroblastoma cell proliferation and signaling and is synergistic with topotecan, Cancer, vol.117, pp.1321-1391, 2011.

J. Zhang, V. Grubor, C. L. Love, A. Banerjee, K. L. Richards et al., Genetic heterogeneity of diffuse large B-cell lymphoma, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.1398-1403, 2013.

S. Zhang, D. Guo, W. Luo, Q. Zhang, Y. Zhang et al., TrkB is highly expressed in NSCLC and mediates BDNF-induced the activation of Pyk2 signaling and the invasion of A549 cells, BMC Cancer, vol.10, p.43, 2010.

X. Zhang, X. Yuan, H. Shi, L. Wu, H. Qian et al., Exosomes in cancer: small particle, big player, J. Hematol. Oncol.J Hematol Oncol, vol.8, p.83, 2015.

X. Zhou, W. Hu, and X. Qin, The role of complement in the mechanism of action of rituximab for B-cell lymphoma: implications for therapy, The Oncologist, vol.13, pp.954-966, 2008.

X. Zhu, Y. Xu, L. M. Solis, W. Tao, L. Wang et al., Long-circulating siRNA nanoparticles for validating Prohibitin1-targeted non-small cell lung cancer treatment, Proc. Natl. Acad. Sci. U. S. A, vol.112, pp.7779-7784, 2015.

L. Zitvogel, A. Regnault, A. Lozier, J. Wolfers, C. Flament et al., Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes, Nat. Med, vol.4, pp.594-600, 1998.

A. A. Zupan, P. A. Osborne, C. E. Smith, N. R. Siegel, R. M. Leimgruber et al., Identification, purification, and characterization of truncated forms of the human nerve growth factor receptor, J. Biol. Chem, vol.264, pp.11714-11720, 1989.