J. Yao, M. Hassan, A. Phan, C. Dagohoy, C. Leary et al., One Hundred Years After ???Carcinoid???: Epidemiology of and Prognostic Factors for Neuroendocrine Tumors in 35,825 Cases in the United States, Journal of Clinical Oncology, vol.26, issue.18, pp.3063-72, 2008.
DOI : 10.1200/JCO.2007.15.4377

K. Young, R. Iyer, D. Morganstein, I. Chau, D. Cunningham et al., Pancreatic neuroendocrine tumors: a review, Future Oncology, vol.28, issue.7, pp.853-64, 2015.
DOI : 10.1097/COC.0b013e31815e4557

D. Metz and R. Jensen, Gastrointestinal Neuroendocrine Tumors: Pancreatic Endocrine Tumors, Gastroenterology, vol.135, issue.5, pp.1469-92, 2008.
DOI : 10.1053/j.gastro.2008.05.047

URL : http://europepmc.org/articles/pmc2612755?pdf=render

A. Mahjoub, O. Reilly, and E. , Emerging therapies for pancreas neuroendocrine cancers, Chinese Clin Oncol, vol.2, p.23, 2013.

K. Oberg, L. Kvols, M. Caplin, D. Fave, G. De-herder et al., Consensus report on the use of somatostatin analogs for the management of neuroendocrine tumors of the gastroenteropancreatic system, Annals of Oncology, vol.15, issue.6, pp.966-73, 2004.
DOI : 10.1093/annonc/mdh216

M. Caplin, M. Pavel, J. ?wik?a, A. Phan, M. Raderer et al., Lanreotide in Metastatic Enteropancreatic Neuroendocrine Tumors, New England Journal of Medicine, vol.371, issue.3, pp.224-257, 2014.
DOI : 10.1056/NEJMoa1316158

A. Rinke, H. Muller, C. Schade-brittinger, K. Klose, P. Barth et al., Placebo-Controlled, Double-Blind, Prospective, Randomized Study on the Effect of Octreotide LAR in the Control of Tumor Growth in Patients With Metastatic Neuroendocrine Midgut Tumors: A Report From the PROMID Study Group, Journal of Clinical Oncology, vol.27, issue.28, pp.4656-63, 2009.
DOI : 10.1200/JCO.2009.22.8510

Y. Jiao, C. Shi, B. Edil, R. De-wilde, D. Klimstra et al., DAXX/ATRX, MEN1, and mTOR Pathway Genes Are Frequently Altered in Pancreatic Neuroendocrine Tumors, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors, pp.1199-203, 2011.
DOI : 10.1056/NEJMoa1001671

URL : http://europepmc.org/articles/pmc3144496?pdf=render

E. Missiaglia, I. Dalai, S. Barbi, S. Beghelli, M. Falconi et al., Pancreatic Endocrine Tumors: Expression Profiling Evidences a Role for AKT-mTOR Pathway, Journal of Clinical Oncology, vol.28, issue.2, pp.245-55, 2010.
DOI : 10.1200/JCO.2008.21.5988

M. Ghayouri, D. Boulware, A. Nasir, J. Strosberg, L. Kvols et al., Activation of the serine/theronine protein kinase Akt in enteropancreatic neuroendocrine tumors, Anticancer Res, vol.30, pp.5063-5070, 2010.

C. Zhou, J. J. Yuan, F. Shi, M. Zhang, J. Liu et al., mTOR Activation in Well Differentiated Pancreatic Neuroendocrine Tumors: A Retrospective Study on 34 Cases, Hepatogastroenterology, vol.58, issue.112, pp.2140-2143, 2011.
DOI : 10.5754/hge11212

X. Han, J. Y. Zhao, J. Xu, X. Lou, and W. , Expression of PTEN and mTOR in pancreatic neuroendocrine tumors, Tumor Biology, vol.256, issue.5, pp.2871-2880, 2013.
DOI : 10.1097/SLA.0b013e31824e6108

K. Zitzmann, D. Toni, E. Brand, S. Göke, B. Meinecke et al., The Novel mTOR Inhibitor RAD001 (Everolimus) Induces Antiproliferative Effects in Human Pancreatic Neuroendocrine Tumor Cells, Neuroendocrinology, vol.25, issue.1, pp.54-60, 2007.
DOI : 10.1074/jbc.273.23.14424

J. Yao, M. Shah, T. Ito, C. Bohas, E. Wolin et al., Everolimus for Advanced Pancreatic Neuroendocrine Tumors, New England Journal of Medicine, vol.364, issue.6, pp.514-537, 2011.
DOI : 10.1056/NEJMoa1009290

O. Reilly, K. Rojo, F. She, Q. Solit, D. Mills et al., mTOR Inhibition Induces Upstream Receptor Tyrosine Kinase Signaling and Activates Akt, Cancer Research, vol.66, issue.3, pp.1500-1508, 2006.
DOI : 10.1158/0008-5472.CAN-05-2925

A. Carracedo, L. Ma, J. Teruya-feldstein, F. Rojo, L. Salmena et al., Inhibition of mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in human cancer, Journal of Clinical Investigation, vol.118, pp.3065-74, 2008.
DOI : 10.1172/JCI34739DS1

C. Bousquet, J. Guillermet-guibert, N. Saint-laurent, E. Archer-lahlou, F. Lopez et al., Direct binding of p85 to sst2 somatostatin receptor reveals a novel mechanism for inhibiting PI3K pathway, The EMBO Journal, vol.62, issue.17, pp.3943-54, 2006.
DOI : 10.1128/MCB.22.12.4062-4072.2002

M. Theodoropoulou, J. Zhang, S. Laupheimer, M. Paez-pereda, C. Erneux et al., Octreotide, a Somatostatin Analogue, Mediates Its Antiproliferative Action in Pituitary Tumor Cells by Altering Phosphatidylinositol 3-Kinase Signaling and Inducing Zac1 Expression, Cancer Research, vol.66, issue.3, pp.1576-82, 2006.
DOI : 10.1158/0008-5472.CAN-05-1189

J. Yao, C. Lombard-bohas, E. Baudin, L. Kvols, P. Rougier et al., Daily Oral Everolimus Activity in Patients With Metastatic Pancreatic Neuroendocrine Tumors After Failure of Cytotoxic Chemotherapy: A Phase II Trial, Journal of Clinical Oncology, vol.28, issue.1, pp.69-76, 2010.
DOI : 10.1200/JCO.2009.24.2669

A. Mohamed, M. Blanchard, M. Albertelli, F. Barbieri, T. Brue et al., Pasireotide and octreotide antiproliferative effects and sst2 trafficking in human pancreatic neuroendocrine tumor cultures, Endocrine Related Cancer, vol.364, issue.6, pp.691-704, 2014.
DOI : 10.1056/NEJMoa1009290

URL : https://hal.archives-ouvertes.fr/hal-01176951

A. Singhi, T. Liu, J. Roncaioli, D. Cao, H. Zeh et al., Alternative Lengthening of Telomeres and Loss of DAXX/ATRX Expression Predicts Metastatic Disease and Poor Survival in Patients with Pancreatic Neuroendocrine Tumors, Clinical Cancer Research, vol.23, issue.2, pp.600-609, 2017.
DOI : 10.1158/1078-0432.CCR-16-1113

F. Meric-bernstam, A. Akcakanat, H. Chen, K. Do, T. Sangai et al., PIK3CA/PTEN Mutations and Akt Activation As Markers of Sensitivity to Allosteric mTOR Inhibitors, Clinical Cancer Research, vol.18, issue.6, pp.1777-89, 2012.
DOI : 10.1158/1078-0432.CCR-11-2123

A. Thiery-vuillemin, G. Mouillet, T. Nguyen-tan-hon, P. Montcuquet, T. Maurina et al., Impact of everolimus blood concentration on its anti-cancer activity in patients with metastatic renal cell carcinoma, Cancer Chemotherapy and Pharmacology, vol.378, issue.3, pp.999-1007, 2014.
DOI : 10.1016/S0140-6736(11)61613-9

K. Thudium, J. Gallo, E. Bouillaud, C. Sachs, S. Eddy et al., Bioavailability of everolimus administered as a single 5 mg tablet versus five 1 mg tablets: a randomized, openlabel , two-way crossover study of healthy volunteers, Clin Pharmacol, vol.7, pp.11-18, 2015.

E. Woltering, V. Salvo, O. Dorisio, T. Lyons, J. Li et al., Clinical Value of Monitoring Plasma Octreotide Levels During Chronic Octreotide Long-Acting Repeatable Therapy in Carcinoid Patients, Pancreas, vol.37, issue.1, pp.94-100, 2008.
DOI : 10.1097/MPA.0b013e31816907ab

E. Wolin, K. Hu, G. Hughes, E. Bouillaud, V. Giannone et al., Safety, tolerability, pharmacokinetics, and pharmacodynamics of a long-acting release (LAR) formulation of pasireotide (SOM230) in patients with gastroenteropancreatic neuroendocrine tumors: results from a randomized, multicenter, open-label, phase I study, Cancer Chemotherapy and Pharmacology, vol.29, issue.Suppl 1, pp.387-95, 2013.
DOI : 10.1200/JCO.2010.33.2056

M. Pertuit, D. Romano, C. Zeiller, A. Barlier, A. Enjalbert et al., The gsp Oncogene Disrupts Ras/ERK-Dependent Prolactin Gene Regulation in gsp Inducible Somatotroph Cell Line, Endocrinology, vol.152, issue.4, pp.1234-1277, 2011.
DOI : 10.1210/en.2010-1077

URL : https://hal.archives-ouvertes.fr/hal-00625462

A. Gorshtein, H. Rubinfeld, E. Kendler, M. Theodoropoulou, V. Cerovac et al., Mammalian target of rapamycin inhibitors rapamycin and RAD001 (everolimus) induce anti-proliferative effects in GH-secreting pituitary tumor cells in vitro, Endocrine Related Cancer, vol.16, issue.3, pp.1017-1044, 2009.
DOI : 10.1677/ERC-08-0269

M. Zatelli, M. Minoia, C. Filieri, F. Tagliati, M. Buratto et al., Effect of Everolimus on Cell Viability in Nonfunctioning Pituitary Adenomas, The Journal of Clinical Endocrinology & Metabolism, vol.95, issue.2, pp.968-76, 2010.
DOI : 10.1210/jc.2009-1641

V. Cerovac, J. Monteserin-garcia, H. Rubinfeld, M. Buchfelder, M. Losa et al., The Somatostatin Analogue Octreotide Confers Sensitivity to Rapamycin Treatment on Pituitary Tumor Cells, Cancer Research, vol.70, issue.2, pp.666-74, 2010.
DOI : 10.1158/0008-5472.CAN-09-2951

S. Grozinsky-glasberg, G. Franchi, M. Teng, C. Leontiou, R. De-oliveira et al., Octreotide and the mTOR Inhibitor RAD001 (Everolimus) Block Proliferation and Interact with the Akt-mTOR-p70S6K Pathway in a Neuro-Endocrine Tumour Cell Line, Neuroendocrinology, vol.86, issue.3, pp.168-81, 2008.
DOI : 10.1073/pnas.91.6.2315

A. Moreno, A. Akcakanat, M. Munsell, A. Soni, J. Yao et al., Antitumor activity of rapamycin and octreotide as single agents or in combination in neuroendocrine tumors, Endocrine Related Cancer, vol.15, issue.1, pp.257-66, 2008.
DOI : 10.1677/ERC-07-0202

T. Ito, L. Lee, and R. Jensen, Treatment of symptomatic neuroendocrine tumor syndromes: recent advances and controversies, Expert Opinion on Pharmacotherapy, vol.11, issue.7, pp.2191-205, 2016.
DOI : 10.1007/s40265-016-0539-6

M. Kulke, E. Bergsland, and J. Yao, Glycemic Control in Patients with Insulinoma Treated with Everolimus, New England Journal of Medicine, vol.360, issue.2, pp.195-202, 2009.
DOI : 10.1056/NEJMc0806740

H. Fiebrich, E. Siemerink, A. Brouwers, T. Links, W. Remkes et al., Everolimus Induces Rapid Plasma Glucose Normalization in Insulinoma Patients by Effects on Tumor As Well As Normal Tissues, The Oncologist, vol.16, issue.6, pp.783-790, 2011.
DOI : 10.1634/theoncologist.2010-0222

V. Bernard, C. Lombard-bohas, M. Taquet, F. Caroli-bosc, P. Ruszniewski et al., Efficacy of everolimus in patients with metastatic insulinoma and refractory hypoglycemia, European Journal of Endocrinology, vol.23, issue.8, pp.665-74, 2013.
DOI : 10.1093/annonc/mds115

M. Asayama, T. Yamada-murano, H. Hara, A. Ooki, M. Kurosumi et al., Everolimus Dramatically Improves Glycemic Control in Unresectable Metastatic Insulinoma: A Case Report, Japanese Journal of Clinical Oncology, vol.335, issue.6076, pp.186-90, 2014.
DOI : 10.1126/science.1215135

H. Bainbridge, E. Larbi, and G. Middleton, Symptomatic Control of Neuroendocrine Tumours with Everolimus, Hormones and Cancer, vol.22, issue.17, pp.254-263, 2015.
DOI : 10.1200/JCO.2004.03.025

J. Li, J. Song, M. Cassidy, P. Rychahou, M. Starr et al., PI3K p110??/Akt Signaling Negatively Regulates Secretion of the Intestinal Peptide Neurotensin Through Interference of Granule Transport, Molecular Endocrinology, vol.26, issue.8, pp.1380-93, 2012.
DOI : 10.1210/me.2012-1024

J. Valentino, J. Li, Y. Zaytseva, W. Mustain, V. Elliott et al., Cotargeting the PI3K and RAS Pathways for the Treatment of Neuroendocrine Tumors, Clinical Cancer Research, vol.20, issue.5, pp.1212-1234, 2014.
DOI : 10.1158/1078-0432.CCR-13-1897

F. Giovinazzo, S. Schimmack, B. Svejda, D. Alaimo, R. Pfragner et al., Chromogranin A and Its Fragments as Regulators of Small Intestinal Neuroendocrine Neoplasm Proliferation, PLoS ONE, vol.246, issue.11, p.81111, 2013.
DOI : 10.1371/journal.pone.0081111.t001

S. Ortolani, C. Ciccarese, S. Cingarlini, G. Tortora, and F. Massari, Suppression of mTOR pathway in solid tumors: lessons learned from clinical experience in renal cell carcinoma and neuroendocrine tumors and new perspectives, Future Oncology, vol.2, issue.11, pp.1809-1837, 2015.
DOI : 10.1200/JCO.2011.40.9631

S. Datta, A. Brunet, and M. Greenberg, Cellular survival: a play in three Akts, Genes & Development, vol.13, issue.22, pp.2905-2932, 1999.
DOI : 10.1101/gad.13.22.2905

T. Fenton and I. Gout, Functions and regulation of the 70kDa ribosomal S6 kinases, The International Journal of Biochemistry & Cell Biology, vol.43, issue.1, pp.47-59, 2011.
DOI : 10.1016/j.biocel.2010.09.018

K. Zitzmann, J. Rüden, . Von, S. Brand, B. Göke et al., Compensatory activation of Akt in response to mTOR and Raf inhibitors ??? A rationale for dual-targeted therapy approaches in neuroendocrine tumor disease, Cancer Letters, vol.295, issue.1, pp.100-109, 2010.
DOI : 10.1016/j.canlet.2010.02.018

S. Iida, Y. Miki, K. Ono, J. Akahira, Y. Nakamura et al., Synergistic anti-tumor effects of RAD001 with MEK inhibitors in neuroendocrine tumors: A potential mechanism of therapeutic limitation of mTOR inhibitor, Molecular and Cellular Endocrinology, vol.350, issue.1, pp.99-106, 2012.
DOI : 10.1016/j.mce.2011.11.024

L. Kvols, K. Oberg, O. Dorisio, T. Mohideen, P. De-herder et al., Pasireotide (SOM230) shows efficacy and tolerability in the treatment of patients with advanced neuroendocrine tumors refractory or resistant to octreotide LAR: results from a phase II study, Endocrine Related Cancer, vol.92, issue.3, pp.657-66, 2012.
DOI : 10.1093/jnci/92.3.205

M. Zatelli, M. Minoia, C. Martini, F. Tagliati, M. Ambrosio et al., Everolimus as a new potential antiproliferative agent in aggressive human bronchial carcinoids, Endocrine Related Cancer, vol.17, issue.3, pp.719-748, 2010.
DOI : 10.1677/ERC-10-0097

J. Yao, A. Phan, D. Chang, R. Wolff, K. Hess et al., Efficacy of RAD001 (Everolimus) and Octreotide LAR in Advanced Low- to Intermediate-Grade Neuroendocrine Tumors: Results of a Phase II Study, Journal of Clinical Oncology, vol.26, issue.26, pp.4311-4319, 2008.
DOI : 10.1200/JCO.2008.16.7858

J. Scoazec, Angiogenesis in Neuroendocrine Tumors: Therapeutic Applications, Neuroendocrinology, vol.250, issue.1, pp.45-56, 2013.
DOI : 10.1148/radiol.2501080291

M. Fjällskog, M. Lejonklou, K. Oberg, B. Eriksson, and E. Janson, Expression of molecular targets for tyrosine kinase receptor antagonists in malignant endocrine pancreatic tumors, Clin Cancer Res, vol.9, pp.1469-73, 2003.

B. Terris, J. Scoazec, L. Rubbia, L. Bregeaud, M. Pepper et al., Expression of vascular endothelial growth factor in digestive neuroendocrine tumours, Histopathology, vol.10, issue.2, pp.133-141, 1998.
DOI : 10.1016/0167-4889(94)90016-7

J. Zhang, Z. Jia, Q. Li, L. Wang, A. Rashid et al., Elevated expression of vascular endothelial growth factor correlates with increased angiogenesis and decreased progression-free survival among patients with low-grade neuroendocrine tumors, Cancer, vol.23, issue.8, pp.1478-86, 2007.
DOI : 10.1016/S0002-9440(10)65289-3

S. Cascinu, D. Ferro, E. Ligi, M. Staccioli, M. Giordani et al., Inhibition of Vascular Endothelial Growth Factor by Octreotide in Colorectal Cancer Patients, Cancer Investigation, vol.18, issue.1, pp.8-12, 2001.
DOI : 10.1016/0167-0115(93)90356-D

R. Mentlein, O. Eichler, F. Forstreuter, and J. Held-feindt, Somatostatin inhibits the production of vascular endothelial growth factor in human glioma cells, International Journal of Cancer, vol.5, issue.4, pp.545-50, 2001.
DOI : 10.1074/jbc.273.12.7099

M. Guba, P. Von-breitenbuch, M. Steinbauer, G. Koehl, S. Flegel et al., Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: involvement of vascular endothelial growth factor, Nature Medicine, vol.11, issue.2, pp.128-163, 2002.
DOI : 10.1006/cyto.1998.0426

M. Zatelli, D. Piccin, C. Vignali, F. Tagliati, M. Ambrosio et al., Pasireotide, a multiple somatostatin receptor subtypes ligand, reduces cell viability in non-functioning pituitary adenomas by inhibiting vascular endothelial growth factor secretion, Endocrine Related Cancer, vol.14, issue.1, pp.91-102, 2007.
DOI : 10.1677/ERC-06-0026

K. Villaume, M. Blanc, G. Gouysse, T. Walter, C. Couderc et al., VEGF Secretion by Neuroendocrine Tumor Cells Is Inhibited by Octreotide and by Inhibitors of the PI3K/AKT/mTOR Pathway, Neuroendocrinology, vol.91, issue.3, pp.268-78, 2010.
DOI : 10.1159/000289569

URL : https://hal.archives-ouvertes.fr/hal-00475083

E. Woltering, Development of Targeted Somatostatin-Based Antiangiogenic Therapy: A Review and Future Perspectives, Cancer Biotherapy & Radiopharmaceuticals, vol.18, issue.4, pp.601-610, 2003.
DOI : 10.1089/108497803322287691

T. Phung, K. Ziv, D. Dabydeen, G. Eyiah-mensah, M. Riveros et al., Pathological angiogenesis is induced by sustained Akt signaling and inhibited by rapamycin, Cancer Cell, vol.10, issue.2, pp.159-70, 2006.
DOI : 10.1016/j.ccr.2006.07.003

URL : https://doi.org/10.1016/j.ccr.2006.07.003

T. Walter, J. Hommell-fontaine, G. Gouysse, C. Pourreyron, M. Nejjari et al., Effects of Somatostatin and Octreotide on the Interactions between Neoplastic Gastroenteropancreatic Endocrine Cells and Endothelial Cells: A Comparison between in vitro and in vivo Properties, Neuroendocrinology, vol.15, issue.3, pp.200-208, 2011.
DOI : 10.1158/1078-0432.CCR-08-2346

URL : https://hal.archives-ouvertes.fr/hal-00710320

E. Raymond, L. Dahan, J. Raoul, Y. Bang, I. Borbath et al., Sunitinib Malate for the Treatment of Pancreatic Neuroendocrine Tumors, New England Journal of Medicine, vol.364, issue.6, pp.501-514, 2011.
DOI : 10.1056/NEJMoa1003825

S. Falletta, S. Partelli, C. Rubini, D. Nann, A. Doria et al., mTOR inhibitors response and mTOR pathway in pancreatic neuroendocrine tumors, Endocrine-Related Cancer, vol.60, issue.11, pp.883-91, 2016.
DOI : 10.1530/ERC-15-0413

I. Modlin, L. Bodei, and M. Kidd, Neuroendocrine tumor biomarkers: From monoanalytes to transcripts and algorithms, Best Practice & Research Clinical Endocrinology & Metabolism, vol.30, issue.1, pp.59-77, 2016.
DOI : 10.1016/j.beem.2016.01.002

M. Höpfner, V. Baradari, A. Huether, C. Schöfl, and H. Scherübl, The insulin-like growth factor receptor 1 is a promising target for novel treatment approaches in neuroendocrine gastrointestinal tumours, Endocrine Related Cancer, vol.13, issue.1, pp.135-184, 2006.
DOI : 10.1677/erc.1.01090

D. Karhoff, S. Sauer, J. Schrader, R. Arnold, V. Fendrich et al., Rap1/B-Raf Signaling Is Activated in Neuroendocrine Tumors of the Digestive Tract and Raf Kinase Inhibition Constitutes a Putative Therapeutic Target, Neuroendocrinology, vol.4, issue.1, pp.45-53, 2007.
DOI : 10.1210/me.15.9.1559

I. Passacantilli, G. Capurso, L. Archibugi, S. Calabretta, S. Caldarola et al., Combined therapy with RAD001 e BEZ235 overcomes resistance of PET immortalized cell lines to mTOR inhibition, Oncotarget, vol.5, issue.14, pp.5381-91, 2014.
DOI : 10.18632/oncotarget.2111

P. Jaquet, G. Gunz, and F. Grisoli, Hormonal Regulation of Prolactin Release by Human Prolactinoma Cells Cultured in Serum-Free Conditions, Hormone Research, vol.22, issue.3, pp.153-63, 1985.
DOI : 10.1159/000180089

T. Cuny, M. Pertuit, M. Sahnoun-fathallah, A. Daly, G. Occhi et al., Genetic analysis in young patients with sporadic pituitary macroadenomas: besides AIP don't forget MEN1 genetic analysis, European Journal of Endocrinology, vol.20, issue.1, pp.533-574, 2013.
DOI : 10.1002/humu.10092

URL : https://hal.archives-ouvertes.fr/hal-00933803

A. Saveanu, G. Gunz, H. Dufour, P. Caron, F. Fina et al., Bim-23244, a somatostatin receptor subtype 2-and 5-selective analog with enhanced efficacy in suppressing growth hormone (GH) from octreotide-resistant human GH-secreting adenomas, J Clin Endocrinol Metab, vol.86, pp.140-145, 2001.