A. Jemal, F. Bray, and M. Center, Global cancer statistics, CA: A Cancer Journal for Clinicians, vol.82, issue.19 suppl, pp.69-90, 2011.
DOI : 10.1097/00002030-199912240-00012

F. Obrocea, M. Sajin, and E. Marinescu, Colorectal cancer and the 7th revision of the TNM staging system: review of changes and suggestions for uniform pathologic reporting, Romanian J Morphol Embryol Rev Roum Morphol Embryol, vol.52, pp.537-581, 2011.

G. Housman, S. Byler, and S. Heerboth, Drug Resistance in Cancer: An Overview, Cancers, vol.6, issue.3, pp.1769-92, 2014.
DOI : 10.4137/GEG.S12270

URL : http://www.mdpi.com/2072-6694/6/3/1769/pdf

R. Levi-montalcini and S. Cohen, IN VITRO AND IN VIVO EFFECTS OF A NERVE GROWTH-STIMULATING AGENT ISOLATED FROM SNAKE VENOM, Proceedings of the National Academy of Sciences, vol.42, issue.9, pp.695-704, 1956.
DOI : 10.1073/pnas.42.9.695

A. Patapoutian and L. Reichardt, Trk receptors: mediators of neurotrophin action, Current Opinion in Neurobiology, vol.11, issue.3, pp.272-80, 2001.
DOI : 10.1016/S0959-4388(00)00208-7

W. Friedman and L. Greene, Neurotrophin Signaling via Trks and p75, Experimental Cell Research, vol.253, issue.1, pp.131-173, 1999.
DOI : 10.1006/excr.1999.4705

H. Akil, A. Perraud, and C. , Fine-Tuning Roles of Endogenous Brain-Derived Neurotrophic Factor, TrkB and Sortilin in Colorectal Cancer Cell Survival, PLoS ONE, vol.124, issue.9, p.25097, 2011.
DOI : 10.1371/journal.pone.0025097.t003

URL : https://hal.archives-ouvertes.fr/hal-00873136

C. Au, M. Siu, and X. Liao, Tyrosine kinase B receptor and BDNF expression in ovarian cancers ??? Effect on cell migration, angiogenesis and clinical outcome, Cancer Letters, vol.281, issue.2, pp.151-61, 2009.
DOI : 10.1016/j.canlet.2009.02.025

H. Bai, H. Li, and W. Li, The PI3K/AKT/mTOR pathway is a potential predictor of distinct invasive and migratory capacities in human ovarian cancer cell lines, Oncotarget, vol.6, issue.28, pp.25520-25552, 2015.
DOI : 10.18632/oncotarget.4550

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, pp.934-978, 2015.

S. Jia, W. Wang, and Z. Hu, BDNF mediated TrkB activation contributes to the EMT progression and the poor prognosis in human salivary adenoid cystic carcinoma, Oral Oncology, vol.51, issue.1, pp.64-70, 2015.
DOI : 10.1016/j.oraloncology.2014.10.008

M. Kim, W. Lee, and J. Jeong, Induction of metastatic potential by TrkB via activation of IL6/JAK2/STAT3 and PI3K/AKT signaling in breast cancer, Oncotarget, vol.6, issue.37, pp.40158-71, 2015.
DOI : 10.18632/oncotarget.5522

G. Sclabas, S. Fujioka, and C. Schmidt, Overexpression of tropomysin-related kinase B in metastatic human pancreatic cancer cells, Clin Cancer Res Off J Am Assoc Cancer Res, vol.11, pp.440-449, 2005.

P. Zhang, Z. Xing, and X. Li, Tyrosine receptor kinase B silencing inhibits anoikis-resistance and improves anticancer efficiency of sorafenib in human renal cancer cells, International Journal of Oncology, vol.48, issue.4, pp.1417-1442, 2016.
DOI : 10.3892/ijo.2016.3356

C. Brunetto-de-farias, D. Rosemberg, and T. Heinen, BDNF/TrkB Content and Interaction with Gastrin-Releasing Peptide Receptor Blockade in Colorectal Cancer, Oncology, vol.62, issue.5-6, pp.430-439, 2010.
DOI : 10.2174/092986609788490177

H. Akil, A. Perraud, and M. Jauberteau, Tropomyosin-related kinase B/brain derived-neurotrophic factor signaling pathway as a potential therapeutic target for colorectal cancer, World Journal of Gastroenterology, vol.22, issue.2, pp.490-500, 2016.
DOI : 10.1002/jso.20785

R. Roesler, C. De-farias, and A. Abujamra, BDNF/TrkB signaling as an anti-tumor target, Expert Review of Anticancer Therapy, vol.17, issue.10, pp.1473-1478, 2011.
DOI : 10.1158/1078-0432.CCR-10-2802

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.371-81, 1992.

T. Heinen, D. Santos, R. Da-rocha, and A. , Trk inhibition reduces cell proliferation and potentiates the effects of chemotherapeutic agents in Ewing sarcoma, Oncotarget, vol.7, issue.23, pp.34860-80, 2016.
DOI : 10.18632/oncotarget.8992

N. Mizushima, The pleiotropic role of autophagy: from protein metabolism to bactericide, Cell Death and Differentiation, vol.34, pp.1535-1576, 2005.
DOI : 10.1016/0005-2787(77)90005-3

URL : http://www.nature.com/cdd/journal/v12/n2s/pdf/4401728a.pdf

K. Han, J. Kim, and M. Choi, Autophagy mediates phase transitions from cell death to life, Heliyon, vol.1, issue.1, p.27, 2015.
DOI : 10.1016/j.heliyon.2015.e00027

URL : https://doi.org/10.1016/j.heliyon.2015.e00027

G. Kroemer, M. ~. , G. Levine, and B. , Autophagy and the Integrated Stress Response, Molecular Cell, vol.40, issue.2, pp.280-93, 2010.
DOI : 10.1016/j.molcel.2010.09.023

URL : https://doi.org/10.1016/j.molcel.2010.09.023

S. Lorin, P. Codogno, and M. Djavaheri-mergny, Autophagy: a new concept in cancer research, Bull Cancer (Paris), vol.95, pp.43-50, 2008.

Y. Kabeya, N. Mizushima, and T. Ueno, LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing, The EMBO Journal, vol.19, issue.21, pp.5720-5728, 2000.
DOI : 10.1093/emboj/19.21.5720

URL : http://emboj.embopress.org/content/embojnl/19/21/5720.full.pdf

D. Klionsky, K. Abdelmohsen, and A. Abe, Guidelines for the use and interpretation of assays for monitoring autophagy, Autophagy, vol.4, issue.4, pp.1-222, 2016.
DOI : 10.4161/auto.6.4.12244

URL : https://hal.archives-ouvertes.fr/hal-01343085

L. Galluzzi, F. Pietrocola, . Bravo-san, and J. Pedro, Autophagy in malignant transformation and cancer progression, The EMBO Journal, vol.34, issue.7, pp.856-80, 2015.
DOI : 10.15252/embj.201490784

URL : http://emboj.embopress.org/content/embojnl/34/7/856.full.pdf

F. Burada, E. Nicoli, and M. Ciurea, Autophagy in colorectal cancer: An important switch from physiology to pathology, World Journal of Gastrointestinal Oncology, vol.7, issue.11, pp.271-84, 2015.
DOI : 10.4251/wjgo.v7.i11.271

URL : http://europepmc.org/articles/pmc4644850?pdf=render

K. Sakitani, Y. Hirata, and Y. Hikiba, Inhibition of autophagy exerts anti-colon cancer effects via apoptosis induced by p53 activation and ER stress, BMC Cancer, vol.456, issue.7219, p.795, 2015.
DOI : 10.1038/nature07383

URL : https://bmccancer.biomedcentral.com/track/pdf/10.1186/s12885-015-1789-5?site=bmccancer.biomedcentral.com

K. Sasaki, N. Tsuno, and E. Sunami, Chloroquine potentiates the anti-cancer effect of 5-fluorouracil on colon cancer cells, BMC Cancer, vol.374, issue.2, p.370, 2010.
DOI : 10.1016/j.bbrc.2008.07.031

URL : https://bmccancer.biomedcentral.com/track/pdf/10.1186/1471-2407-10-370?site=bmccancer.biomedcentral.com

J. Wang, X. Tan, and Q. Yang, Inhibition of autophagy promotes apoptosis and enhances anticancer efficacy of adriamycin via augmented ROS generation in prostate cancer cells, The International Journal of Biochemistry & Cell Biology, vol.77, pp.80-90, 2016.
DOI : 10.1016/j.biocel.2016.05.020

L. Evy, J. Cacheux, W. Bara, and M. , Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth, Nat Cell Biol, vol.17, pp.1062-73, 2015.

, Journal of Cellular and Molecular Medicine, p.2621

, J. Cell. Mol. Med, vol.21, issue.10, p.2017

N. Kawamura, K. Kawamura, and M. Manabe, Inhibition of Brain-Derived Neurotrophic Factor/Tyrosine Kinase B Signaling Suppresses Choriocarcinoma Cell Growth, Endocrinology, vol.151, issue.7, pp.3006-3020, 2010.
DOI : 10.1210/en.2009-1378

S. Lefort, C. Joffre, and Y. Kieffer, Inhibition of autophagy as a new means of improving chemotherapy efficiency in high-LC3B triple-negative breast cancers, Autophagy, vol.61, issue.12, pp.2122-2164, 2015.
DOI : 10.4161/auto.4451

M. Elin, C. Perraud, A. Christou, and N. , New ex-ovo colorectal-cancer models from different SdFFF-sorted tumour-initiating cells, Anal Bioanal Chem, vol.407, pp.8433-8476, 2015.

A. Perraud, H. Akil, and M. Nouaille, Implications of cleaved caspase 3 and AIF expression in colorectal cancer based on patient age, Oncology Reports, vol.27, pp.1787-93, 2012.
DOI : 10.3892/or.2012.1737

URL : https://hal.archives-ouvertes.fr/hal-00873114

A. Fauchais, F. Lallou-e, and M. Lise, Role of Endogenous Brain-Derived Neurotrophic Factor and Sortilin in B Cell Survival, The Journal of Immunology, vol.181, issue.5, pp.3027-3065, 1950.
DOI : 10.4049/jimmunol.181.5.3027

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

E. Vanhecke, E. Adriaenssens, and S. Verbeke, Brain-Derived Neurotrophic Factor and Neurotrophin-4/5 Are Expressed in Breast Cancer and Can Be Targeted to Inhibit Tumor Cell Survival, Clinical Cancer Research, vol.17, issue.7, pp.1741-52, 2011.
DOI : 10.1158/1078-0432.CCR-10-1890

K. Tanaka, Y. Okugawa, and Y. Toiyama, Brain-Derived Neurotrophic Factor (BDNF)-Induced Tropomyosin-Related Kinase B (Trk B) Signaling Is a Potential Therapeutic Target for Peritoneal Carcinomatosis Arising from Colorectal Cancer, PLoS ONE, vol.56, issue.79, p.96410, 2014.
DOI : 10.1371/journal.pone.0096410.t001

URL : https://doi.org/10.1371/journal.pone.0096410

S. Nye, S. Squinto, and D. Glass, K-252a and staurosporine selectively block autophosphorylation of neurotrophin receptors and neurotrophin-mediated responses., Molecular Biology of the Cell, vol.3, issue.6
DOI : 10.1091/mbc.3.6.677

URL : http://europepmc.org/articles/pmc275622?pdf=render

, Mol Biol Cell, vol.3, pp.677-86, 1992.

K. Sato, K. Tsuchihara, and S. Fujii, Autophagy Is Activated in Colorectal Cancer Cells and Contributes to the Tolerance to Nutrient Deprivation, Cancer Research, vol.67, issue.20, pp.9677-84, 2007.
DOI : 10.1158/0008-5472.CAN-07-1462

Y. Yang, L. Hu, and H. Zheng, Application and interpretation of current autophagy inhibitors and activators, Acta Pharmacologica Sinica, vol.17, issue.5, pp.625-660, 2013.
DOI : 10.1093/hmg/ddm294

URL : https://www.nature.com/articles/aps20135.pdf

Y. Li, S. Lam, and J. Mak, Erlotinib-induced autophagy in epidermal growth factor receptor mutated non-small cell lung cancer, Lung Cancer, vol.81, issue.3, pp.354-61, 2013.
DOI : 10.1016/j.lungcan.2013.05.012

Z. Chen, S. Gao, and D. Wang, Colorectal cancer cells are resistant to anti-EGFR monoclonal antibody through adapted autophagy, Am J Transl Res, vol.8, pp.1190-1196, 2016.

F. Janku, D. Mcconkey, and D. Hong, Autophagy as a target for anticancer therapy, Nature Reviews Clinical Oncology, vol.4, issue.9, pp.528-567, 2011.
DOI : 10.4161/auto.5338

J. Cui, Y. Hu, and X. Feng, EGFR inhibitors and autophagy in cancer treatment, Tumor Biology, vol.17, issue.43, pp.11701-11710, 2014.
DOI : 10.3748/wjg.v17.i43.4779

S. Pascolo, Time to use a dose of Chloroquine as an adjuvant to anti-cancer chemotherapies, European Journal of Pharmacology, vol.771, pp.139-183, 2016.
DOI : 10.1016/j.ejphar.2015.12.017

S. Aveic and G. Tonini, Resistance to receptor tyrosine kinase inhibitors in solid tumors: can we improve the cancer fighting strategy by blocking autophagy?, Cancer Cell International, vol.8, issue.1, p.62, 2016.
DOI : 10.1038/nrc2442

R. Hewitt, A. Mcmarlin, and D. Kleiner, Validation of a model of colon cancer progression, The Journal of Pathology, vol.192, issue.4, pp.446-54, 2000.
DOI : 10.1038/bjc.1995.55

H. Fujikawa, K. Tanaka, and Y. Toiyama, High TrkB expression levels are associated with poor prognosis and EMT induction in colorectal cancer cells, Journal of Gastroenterology, vol.12, issue.7, pp.775-84, 2012.
DOI : 10.1158/1078-0432.CCR-06-0038

S. Diaz-cano, Tumor Heterogeneity: Mechanisms and Bases for a Reliable Application of Molecular Marker Design, International Journal of Molecular Sciences, vol.19, issue.2, pp.1951-2011, 2012.
DOI : 10.1093/annonc/mdn021

M. Fan, J. Sun, and W. Wang, Tropomyosin-related kinase B promotes distant metastasis of colorectal cancer through protein kinase B-mediated anoikis suppression and correlates with poor prognosis, Apoptosis, vol.17, issue.5, pp.860-70, 2014.
DOI : 10.1038/sj.onc.1203598

A. Giatromanolaki, M. Koukourakis, and A. Harris, Prognostic relevance of light chain 3 (LC3A) autophagy patterns in colorectal adenocarcinomas, Journal of Clinical Pathology, vol.63, issue.10, pp.867-72, 2010.
DOI : 10.1136/jcp.2010.079525

, Journal of Cellular and Molecular Medicine