, The "Institut Nationnal du Cancer (INCA), Funding This study was supported by INSERM U1149/The Inflammation Research Center (CRI)

S. Danese and C. Fiocchi, Ulcerative colitis, N. Engl. J. Med, vol.365, pp.1713-1725, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01707812

E. Domènech, M. Mañosa, and E. Cabré, An overview of the natural history of inflammatory bowel diseases, Dig. Dis, vol.32, pp.320-327, 2014.

H. S. Souza and C. Fiocchi, Immunopathogenesis of IBD: current state of the art, Nat Rev Gastroenterol Hepatol, vol.13, pp.13-27, 2016.

P. Hindryckx, V. Jairath, and G. Haens, Acute severe ulcerative colitis: from pathophysiology to clinical management, Nat Rev Gastroenterol Hepatol, vol.13, pp.654-664, 2016.

M. Jo and S. T. Jung, Engineering therapeutic antibodies targeting G-protein-coupled receptors, Exp. Mol. Med, vol.48, p.207, 2016.

T. Saskia and D. C. Baumgart, Targeting leukocyte migration and adhesion in Crohn's disease and ulcerative colitis, Inflammopharmacol, vol.20, pp.1-18, 2012.

R. Cheluvappa, Experimental appendicitis and appendectomy modulate the CCL20-CCR6 axis to limit inflammatory colitis pathology, Int. J. Color. Dis, vol.29, pp.1181-1188, 2014.

M. Zhang, J. D. Venable, and R. L. Thurmond, The histamine H4 receptor in autoimmune disease, Expert Opin. Investig. Drugs, vol.15, pp.1443-1452, 2006.

J. C. Ashton, Cannabinoids for the treatment of inflammation, Curr. Opin. Investig. Drugs, vol.8, pp.373-384, 2007.

A. Wasilewski, M. Storr, M. Zieli, and J. Fichna, Role of G protein-coupled orphan receptors in intestinal inflammation: novel targets in inflammatory bowel diseases, Inflamm. Bowel Dis, vol.21, pp.666-673, 2015.

J. A. Waschek, VIP and PACAP: neuropeptide modulators of CNS inflammation, injury, and repair, Br. J. Pharmacol, vol.169, pp.512-523, 2013.

T. Sakurai, A. Amemiya, and M. Ishii, Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior, Cell, vol.92, pp.573-585, 1998.

K. Ohno, M. Hondo, and T. Sakurai, Cholinergic regulation of orexin/hypocretin neurons through M 3 muscarinic receptor in mice, J. Pharmacol. Sci, vol.106, pp.485-491, 2008.

T. Sakurai, The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness, Nat. Rev. Neurosci, vol.8, pp.171-181, 2007.

M. V. Heinonen, A. K. Purhonen, K. A. Mäkelä, and K. H. Herzig, Functions of orexins in peripheral tissues, Acta Physiol (Oxf), vol.192, pp.471-485, 2008.

T. Voisin, P. Rouet-benzineb, N. Reuter, and M. Laburthe, Orexins and their receptors: structural aspects and role in peripheral tissues, Cell Mol Life Sci CMLS, vol.60, pp.72-87, 2003.

C. M. Duffy, C. Yuan, and L. E. Wisdorf, Role of orexin a signaling in dietary palmitic acid-activated microglial cells, Neurosci. Lett, vol.606, pp.140-144, 2015.

X. Xiong, R. E. White, and L. Xu, Mitigation of murine focal cerebral ischemia by the hypocretin/orexin system is associated with reduced inflammation, Stroke, vol.44, pp.764-770, 2013.

Y. Ogawa, Y. Irukayama-tomobe, and N. Murakoshi, Peripherally administered orexin improves survival of mice with endotoxin shock, p.21055, 2016.

M. Laburthe, T. Voisin, and A. E. Firar, Orexins/hypocretins and orexin receptors in apoptosis: a mini-review, Acta Physiol, vol.198, pp.393-402, 2010.

M. Laburthe and T. Voisin, The orexin receptor OX1R in colon cancer: a promising therapeutic target and a new paradigm in G protein-coupled receptor signalling through ITIMs, Br. J. Pharmacol, vol.165, pp.1678-1687, 2012.

T. Voisin, A. Elfirar, and M. Fasseu, Aberrant expression of OX1 receptors for orexins in colon cancers and liver metastases: an openable gate to apoptosis, Cancer Res, vol.71, pp.3341-3351, 2011.

D. Alexandre, C. Hautot, and M. Mehio, The orexin type 1 receptor is overexpressed in advanced prostate cancer with a neuroendocrine differentiation, and mediates apoptosis, Eur. J. Cancer, vol.50, pp.2126-2133, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01960616

S. Dayot, D. Speisky, and A. Couvelard, In vitro, in vivo and ex vivo demonstration of the antitumoral role of hypocretin-1/orexin-A and almorexant in pancreatic ductal adenocarcinoma, Oncotarget, vol.9, pp.6952-6967, 2018.

C. A. Schneider, W. S. Rasband, and K. W. Eliceiri, NIH image to ImageJ: 25 years of image analysis, Nat. Methods, vol.9, pp.671-675, 2012.

A. E. Firar, T. Voisin, and C. Rouyer-fessard, Discovery of a functional immunoreceptor tyrosine-based switch motif in a 7-transmembrane-spanning receptor: role in the orexin receptor OX1R-driven apoptosis, FASEB J, vol.23, pp.4069-4080, 2009.

M. Ehrström, E. Näslund, and F. Levin, Pharmacokinetic profile of orexin A and effects on plasma insulin and glucagon in the rat, Regul. Pept, vol.119, pp.209-212, 2004.

P. Nicole, P. Couvineau, and N. Jamin, Crucial role of the orexin-B C-terminus in the induction of OX1 receptor-mediated apoptosis: analysis by alanine scanning, molecular modelling and site-directed mutagenesis, Br. J. Pharmacol, vol.172, pp.5211-5223, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01432232

P. M. Cummins, Occludin: one protein, many forms, Mol. Cell. Biol, vol.32, pp.242-250, 2012.

X. Treton, E. Pedruzzi, and C. Guichard, Combined NADPH oxidase 1 and interleukin 10 deficiency induces chronic endoplasmic reticulum stress and causes ulcerative colitis-like disease in mice, PLoS One, vol.9, p.101669, 2014.

W. Blonski, A. M. Buchner, and G. R. Lichtenstein, Treatment of ulcerative colitis, Curr. Opin. Gastroenterol, vol.30, pp.84-96, 2014.

M. N. Ajuebor and M. G. Swain, Role of chemokine and chemokine receptors in the gastrointestinal tract, Immunology, vol.105, pp.137-143, 2002.

J. Torres, R. K. Boyapati, and N. A. Kennedy, Systematic review of effects of withdrawal of immunomodulators or biologic agents from patients with inflammatory bowel disease, Gastroenterology, vol.149, pp.1716-1730, 2015.

S. Agarwal and L. Mayer, Gastrointestinal manifestations in primary immune disorders, Inflamm. Bowel Dis, vol.16, pp.703-711, 2010.

I. Attreya, R. Attreya, and M. F. Neurath, NF-?Bininflammatory bowel disease, J. Intern. Med, vol.263, pp.591-596, 2008.

P. P. Tak and G. S. Firestein, NF-?B: a key role in inflammatory diseases, J. Clin. Invest, vol.107, pp.7-11, 2001.

A. Grinspan and A. Kornbluth, Positioning therapy for ulcerative colitis, Curr Gastroenterol Rep, vol.17, pp.1-8, 2015.

R. P. Macdermott, Chemokines in the inflammatory bowel diseases, J. Clin. Immunol, vol.19, pp.266-272, 1999.

M. S. Maddur, P. Miossec, S. V. Kaveri, and J. Bayry, Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies, Am J Pathol, vol.181, pp.8-18, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-02455555

M. R. Walter, The molecular basis of IL-10 function: from receptor structure to the onset of signaling, Curr. Top. Microbiol. Immunol, vol.380, pp.191-212, 2014.

D. M. Rennick, M. M. Fort, and N. J. Davidson, Studies with IL-10 ? / ? mice: an overview, J. Leukoc. Biol, vol.61, pp.389-396, 1997.

C. S. Leonard and J. P. Kukkonen, Orexin/hypocretin receptor signaling: a functional perspective, Br. J. Pharmacol, vol.171, pp.294-313, 2014.

M. D. Giovangiulio, S. Verheijden, and G. Boesmans, The neuromodulation of the intestinal immune system and its relevance in inflammatory bowel disease, Front. Immunol, vol.6, pp.1-15, 2015.

F. J. Martinez-orozco, J. L. Vicario, and I. Villalibre-valderrey, Narcolepsy with cataplexy and comorbid immunopathological diseases, J. Sleep Res, vol.23, pp.414-419, 2014.

H. Himmerich, P. A. Beitinger, and S. Fulda, Plasma levels of tumor necrosis factor alpha and soluble tumor necrosis factor receptors in patients with narcolepsy, Arch. Intern. Med, vol.166, pp.1739-1743, 2006.

A. Couvineau and M. Laburthe, The family B1 GPCR: structural aspects and interaction with accessory proteins, Curr. Drug Targets, vol.13, pp.103-115, 2012.

K. Newton and V. M. Dixit, Signaling in innate immunity and inflammation, Cold Spring Harb Perspect Biol, vol.4, p.6049, 2016.

C. Boularan and J. H. Kehrl, Implications of non-canonical G-protein signaling for the immune system, Cell. Signal, vol.26, pp.1269-1282, 2014.

C. J. Langmead, J. C. Jerman, and S. J. Brough, Characterization of the binding of [3H]-SB-674042, a novel nonpeptide antagonist, to the human orexin-1 receptor, Br. J. Pharmacol, vol.141, pp.340-346, 2004.

A. L. Kirchgessner and M. Liu, Orexin synthesis and response in the gut, Neuron, vol.24, pp.941-951, 1999.

A. Blais, G. Drouin, and C. Chaumontet, Impact of orexin-A treatment on food intake, energy metabolism and body weight in mice, PLoS One, vol.12, p.169908, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01602768

M. A. Kamm, Review article: maintenance of remission in ulcerative colitis, Aliment. Pharmacol. Ther, vol.16, pp.21-24, 2002.

L. De-lecea, T. S. Kilduff, C. Peyron, X. Gao, P. E. Foye et al., The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity, Proc Natl Acad Sci, vol.95, pp.322-349, 1998.

T. Sakurai, A. Amemiya, M. Ishii, I. Matsuzaki, R. M. Chemelli et al., Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior, Cell, vol.92, pp.80949-80955, 1998.

M. Laburthe and T. Voisin, The orexin receptor OX(1)R in colon cancer: a promising therapeutic target and a new paradigm in G protein-coupled receptor signaling through ITIMs, Br J Pharmacol, vol.165, pp.1678-87, 2012.

L. De-lecea and J. G. Sutcliffe, The hypocretins/orexins: novel hypothalamic neuropeptides involved in different physiological systems, Cell Mol Life Sci, vol.56, pp.473-80, 1999.

M. Mieda and M. Yanagisawa, Sleep, feeding, and neuropeptides: roles of orexins and orexin receptors, Curr Opin Neurobiol, vol.12, pp.339-384, 2002.

L. Lin, J. Faraco, R. Li, H. Kadotani, W. Rogers et al., The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene, Cell, vol.98, pp.365-76, 1999.

R. M. Chemelli, J. T. Willie, C. M. Sinton, J. K. Elmquist, T. Scammell et al., Narcolepsy in orexin knockout mice: molecular genetics of sleep regulation, Cell, vol.98, pp.437-51, 1999.

M. Heinonen, A. Purhonen, K. Mäkelä, and K. H. Herzig, Functions of orexins in peripheral tissues, Acta Physiol, vol.192, pp.471-85, 2008.

T. Voisin, P. Rouet-benzineb, N. Reuter, and M. Laburthe, Orexins and their receptors: structural aspects and role in peripheral tissues, Cell Mol Life Sci, vol.60, pp.72-87, 2003.

A. L. Kirchgessner and M. Liu, Orexin synthesis and response in the gut, Neuron, vol.24, pp.941-51, 1999.

M. Nakabayashi, T. Suzuki, K. Takahashi, K. Totsune, Y. Muramatsu et al., Orexin-A expression in human peripheral tissues, Mol Cell Endocrinol, vol.205, pp.43-50, 2003.

H. S. Randeva, E. Karteris, D. Grammatopoulos, and E. W. Hillhouse, Expression of orexin-A and functional orexin type 2 receptors in the human adult adrenals: implications for adrenal function and energy homeostasis, J Clin Endocrinol Metab, vol.86, pp.4808-4821, 2001.

O. Jöhren, S. J. Neidert, M. Kummer, A. Dendorfer, and P. Dominiak, Preproorexin and orexin receptor mRNAs are differentially expressed in peripheral tissues of male and female rats, Endocrinology, vol.142, pp.3324-3355, 2001.

S. Valiante, G. Liguori, S. Tafuri, R. Campese, R. Monaco et al., Expression of orexin A and its receptor 1 in the human prostate, J Anat, vol.222, pp.473-80, 2013.

Z. Arihara, K. Takahashi, O. Murakami, K. Totsune, M. Sone et al., Immunoreactive orexin-A in human plasma, Peptides, vol.22, pp.139-181, 2001.

S. Sakurai, T. Nishijima, S. Takahashi, K. Yamauchi, Z. Arihara et al., Clinical significance of daytime plasma orexin-A-like immunoreactivity concentrations in patients with obstructive sleep apnea hypopnea syndrome, Respiration, vol.71, pp.380-384, 2004.

P. Nicole, P. Couvineau, N. Jamin, T. Voisin, and A. Couvineau, Crucial role of the orexin-B C-terminus in the induction of OX1 receptor-mediated apoptosis: analysis by alanine scanning, molecular modelling and site-directed mutagenesis, Br J Pharmacol, vol.172, pp.5211-5234, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01432232

M. Laburthe, T. Voisin, E. Firar, and A. , Orexins/hypocretins and orexin receptors in apoptosis: a mini-review, Acta Physiol, vol.198, pp.393-402, 2010.

C. S. Leonard and J. P. Kukkonen, Orexin/hypocretin receptor signaling: a functional perspective, Br J Pharmacol, vol.171, pp.294-307, 2014.

A. Couvineau and M. Laburthe, The family B1 GPCR: structural aspects and interaction with accessory proteins, Curr Drug Targets, vol.13, pp.103-115, 2012.

Y. K. Peterson and L. M. Luttrell, The diverse roles of arrestin scaffolds in G protein-coupled receptor signaling, Pharmacol Rev, vol.69, pp.256-97, 2017.

S. Rajagopal and S. K. Shenoy, GPCR desensitization: acute and prolonged phases, Cell Signal, vol.41, pp.9-16, 2018.

, Frontiers in Endocrinology | www.frontiersin.org, vol.7, p.573, 2018.

N. Gutierrez, A. Mcdonald, and P. H. , GPCRs: Emerging anti-cancer drug targets, Cell Signal, vol.41, pp.65-74, 2018.

T. W. Moody, I. Ramos-alvarez, and R. T. Jensen, Neuropeptide G proteincoupled receptors as oncotargets, Front Endocrinol, vol.9, p.345, 2018.

L. A. Torre, F. Bray, R. L. Siegel, J. Ferlay, and J. Lortet-tieulent, Global cancer statistics 2012, CA Cancer J Clin, vol.65, pp.87-108, 2015.

V. Valentini, R. G. Van-stiphout, G. Lammering, M. A. Gambacorta, M. C. Barba et al., Nomograms for predicting local recurrence, distant metastases, and overall survival for patients with locally advanced rectal cancer on the basis of European randomized clinical trials, J Clin Oncol, vol.29, pp.3163-72, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00719141

R. Siegel, C. Desantis, and A. Jemal, Colorectal cancer statistics 2014, CA Cancer J Clin, vol.64, pp.104-121, 2014.

E. Van-cutsem, J. Tabernero, R. Lakomy, H. Prenen, J. Prausová et al., Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with oxaliplatin-based regimen, J Clin Oncol, vol.30, pp.2499-506, 2012.

A. Grothey, D. Sargent, R. M. Goldberg, and H. J. Schmoll, Survival of patients with advanced colorectal cancer improves with the availability of fluorouracilleucovorin, irinotecan, and oxaliplatin in the course of treatment, J Clin Oncol, vol.22, pp.1209-1223, 2004.

S. D. Markowitz and M. M. Bertagnolli, Molecular origins of cancer: molecular basis of colorectal cancer, N Engl J Med, vol.361, pp.2449-60, 2009.

P. A. Insel, K. Sriram, S. Z. Wiley, A. Wilderman, T. Katakia et al., GPCRomics: GPCR expression in cancer cells and tumors identifies new, potential biomarkers and therapeutic targets, Front Pharmacol, vol.9, p.431, 2018.

M. Laburthe, M. Rousset, C. Boissard, G. Chevalier, A. Zweibaum et al., Vasoactive intestinal peptide: a potent stimulator of adenosine 3':5' cyclic monophosphate accumulation in gut carcinoma cell lines in culture, Proc Natl Acad Sci, vol.75, pp.2772-2777, 1978.

J. J. Maoret, D. Pospaï, C. Rouyer-fessard, A. Couvineau, C. Laboisse et al., Neurotensin receptor and its mRNA are expressed in many human colon cancer cell lines but not in normal colonic epithelium: binding studies and RT-PCR experiments, Biochem Biophys Res Commun, vol.203, pp.465-71, 1994.

P. Singh, B. Dai, H. Wu, and A. Owlia, Role of autocrine and endocrine gastrinlike peptides in colonic carcinogenesis, Curr Opin Gastroenterol, vol.16, pp.68-77, 2000.

D. Darmoul, V. Gratio, H. Devaud, T. Lehy, and M. Laburthe, Aberrant expression and activation of the thrombin receptor protease-activated receptor-1 induces cell proliferation and motility in human colon cancer cells, Am J Pathol, vol.162, issue.10, pp.64283-64289, 2003.

V. Gratio, F. Walker, T. Lehy, M. Laburthe, and D. Darmoul, Aberrant expression of proteinase-activated receptor 4 promotes colon cancer cell proliferation through a persistent signaling that involves Src and ErbB-2 kinase, Int J Cancer, vol.124, pp.1517-1542, 2009.

D. Darmoul, V. Gratio, H. Devaud, F. Peiretti, and M. Laburthe, Activation of proteinase-activated receptor 1 promotes human colon cancer cell proliferation through epidermal growth factor receptor transactivation, Mol Cancer Res, vol.2, pp.514-536, 2004.

M. Yang, W. W. Zhong, N. Srivastava, A. Slavin, J. Yang et al., proteincoupled lysophosphatidic acid receptor stimulate proliferation of colon cancer cells through the {beta}-catenin pathway, Proc Natl Acad Sci, vol.102, pp.6027-6059, 2005.

S. Chell, A. Kaidi, A. C. Williams, and C. Paraskeva, Mediators of PGE2 synthesis and signalling downstream of COX-2 represent potential targets for the prevention/treatment of colorectal cancer, Biochim Biophys Acta, vol.1766, pp.104-123, 2006.

D. Darmoul, V. Gratio, H. Devaud, and M. Laburthe, Protease-activated receptor 2 in colon cancer: trypsin-induced MAPK phosphorylation and cell proliferation are mediated by epidermal growth factor receptor transactivation, J Biol Chem, vol.279, pp.20927-20961, 2004.

R. Lappano and M. Maggiolini, G protein-coupled receptors: novel targets for drug discovery in cancer, Nat Rev Drug Discov, vol.10, pp.47-60, 2011.

P. Rouet-benzineb, C. Rouyer-fessard, A. Jarry, V. Avondo, C. Pouzet et al., Orexins acting at native OX(1) receptor in colon cancer and neuroblastoma cells or at recombinant OX(1) receptor suppress cell growth by inducing apoptosis, J Biol Chem, vol.279, pp.45875-86, 2004.

T. Voisin, A. El-firar, C. Rouyer-fessard, V. Gratio, and M. Laburthe, A hallmark of immunoreceptor, the tyrosine-based inhibitory motif ITIM, is present in the G protein-coupled receptor OX1R for orexins and drives apoptosis: a novel mechanism, FASEB J, vol.22, pp.1993-2002, 2008.

T. Voisin, A. El-firar, M. Fasseu, C. Rouyer-fessard, V. Descatoire et al., Aberrant expression of OX1 receptors for orexins in colon cancers and liver metastases: an openable gate to apoptosis, Cancer Res, vol.71, pp.3341-51, 2011.

T. Lesuffleur, A. Kornowski, C. Luccioni, M. Muleris, A. Barbat et al., Adaptation to 5-fluorouracil of the heterogeneous human colon tumor cell line HT-29 results in the selection of cells committed to differentiation, Int J Cancer, vol.49, pp.721-751, 1991.

S. Dayot, D. Speisky, A. Couvelard, P. Bourgoin, V. Gratio et al., In vitro, in vivo and ex vivo demonstration of the antitumoral role of hypocretin-1/orexin-A and almorexant in pancreatic ductal adenocarcinoma, Oncotarget, vol.9, pp.6952-67, 2018.

N. Chartrel, Y. Anouar, L. Jeandel, D. Alexandre, J. Leprince et al., Methods and Pharmaceutical Compositions Using Orexins (OXA, OXB) for the Treatment of Prostate Cancers. US Patent Application, 2017.

A. El-firar, T. Voisin, C. Rouyer-fessard, M. A. Ostuni, A. Couvineau et al., Discovery of a functional immunoreceptor tyrosine-based switch motif in a 7-transmembrane-spanning receptor: role in the orexin receptor OX1R-driven apoptosis, FASEB J, vol.23, pp.4069-80, 2009.

M. Daëron, S. Jaeger, D. Pasquier, L. Vivier, and E. , Immunoreceptor tyrosinebased inhibition motifs: a quest in the past and future, Immunol Rev, vol.224, pp.11-43, 2008.

D. P. Ryan, T. S. Hong, and N. Bardeesy, Pancreatic adenocarcinoma, N Engl J Med, vol.371, pp.2140-2181, 2014.

C. Neuzillet, A. Tijeras-raballand, P. Bourget, J. Cros, A. Couvelard et al., State of the art and future directions of pancreatic ductal adenocarcinoma therapy, Pharmacol Ther, vol.155, pp.80-104, 2015.

L. Rahib, B. D. Smith, R. Aizenberg, A. B. Rosenzweig, J. M. Fleshman et al., Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States, Cancer Res, vol.74, pp.2913-2934, 2014.

G. Garcea, C. P. Neal, C. J. Pattenden, W. P. Steward, and D. P. Berry, Molecular prognostic markers in pancreatic cancer: a systematic review, Eur J Cancer, vol.41, pp.2213-2249, 2005.

M. A. Shields, S. Dangi-garimella, A. J. Redig, and H. G. Munshi, Biochemical role of the collagen-rich tumour microenvironment in pancreatic cancer progression, Biochem J, vol.441, pp.541-52, 2012.

K. P. Olive, M. A. Jacobetz, C. J. Davidson, A. Gopinathan, D. Mcintyre et al., Inhibition of hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer, Science, vol.324, pp.1457-61, 2009.

N. Messal, N. Fernandez, S. Dayot, V. Gratio, N. P. Prochasson et al., Ectopic expression of OX1R in ulcerative colitis mediates antiinflammatory effect of orexin-A, BBA Mole Basis Dis, vol.1864, pp.3618-3646, 2018.

C. J. Winrow and J. J. Renger, Discovery and development of orexin receptor antagonists as therapeutics for insomnia, Br J Pharmacol, vol.171, pp.283-93, 2014.

A. J. Roecker, C. D. Cox, and P. J. Coleman, Orexin receptor antagonists: new therapeutic agents for the treatment of insomnia, J Med Chem, vol.59, pp.504-534, 2016.

C. Dugovic, J. E. Shelton, L. E. Aluisio, I. C. Fraser, X. Jiang et al., Blockade of orexin-1 receptors attenuates orexin-2 receptor antagonism-induced sleep promotion in the rat, J Frontiers in Endocrinology | www.frontiersin.org, vol.8, p.573, 2018.

T. Pharmacol-exp, , vol.330, pp.142-51, 2009.

D. Smart, C. Sabido-david, S. J. Brough, F. Jewitt, A. Johns et al., SB-334867-A: the first selective orexin-1 receptor antagonist, Br J Pharmacol, vol.132, pp.1179-82, 2001.

P. Bonaventure, J. Shelton, S. Yun, D. Nepomuceno, S. Sutton et al., Characterization of JNJ-42847922, a selective orexin-2 receptor antagonist, as a clinical candidate for the treatment of insomnia, J Pharmacol Exp Ther, vol.354, pp.471-82, 2015.

C. Betschart, S. Hintermann, D. Behnke, S. Cotesta, M. Fendt et al., Identification of a novel series of orexin receptor antagonists with a distinct effect on sleep architecture for the treatment of insomnia, J Med Chem, vol.56, pp.7590-607, 2013.

C. D. Cox, M. J. Breslin, D. B. Whitman, J. D. Schreier, G. B. Mcgaughey et al., Discovery of the dual orexin receptor antagonist [(7R)-4-(5-chloro-1,3-benzoxazol-2-yl)-7-methyl-1,4-diazepan-1-yl][5-methyl-2-(2H?1,2,3-triazol-2-yl)phenyl]methanone (MK-4305) for the treatment of insomnia, J Med Chem, vol.53, pp.5320-5352, 2010.

A. Turku, M. K. Rinne, B. Af, G. Gennäs, H. Xhaard et al., Orexin receptor agonist Yan 7874 is a weak agonist of orexin/hypocretin receptors and shows orexin receptor-independent cytotoxicity, PLoS ONE, vol.12, p.178526, 2017.

Y. Irukayama-tomobe, Y. Ogawa, H. Tominaga, Y. Ishikawa, N. Hosokawa et al., Nonpeptide orexin type-2 receptor agonist ameliorates narcolepsy-cataplexy symptoms in mouse models, Proc Natl Acad Sci, vol.114, pp.5731-5737, 2017.

A. Heifetz, M. J. Bodkin, and P. C. Biggin, Discovery of the first selective, nonpeptidic orexin 2 receptor agonists, J Med Chem, vol.58, pp.7928-7958, 2015.

E. M. Soffin, C. H. Gill, S. J. Brough, J. C. Jerman, and C. H. Davies, Pharmacological characterisation of the orexin receptor subtype mediating postsynaptic excitation in the rat dorsal raphe nucleus, Neuropharmacology, vol.46, pp.1168-76, 2004.

J. S. Smith, R. J. Lefkowitz, and S. Rajagopal, Biased signalling: from simple switches to allosteric microprocessors, Nat Rev Drug Discov, vol.17, pp.243-60, 2018.

R. L. Siegel, K. D. Miller, and A. Jemal, Cancer statistics, CA Cancer J Clin, vol.66, pp.7-30, 2016.

A. Patel and L. Fong, Immunotherapy for prostate cancer: where do we go from here?-PART 2: checkpoint inhibitors, immunotherapy combinations, tumor microenvironment modulation, and cellular therapies, Oncology, vol.32, pp.65-73, 2018.

K. O. Rove and E. D. Crawford, Traditional androgen ablation approaches to advanced prostate cancer: new insights, Can J Urol, vol.21, pp.14-21, 2014.

P. A. Watson, V. K. Arora, and C. L. Sawyers, Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer, Nat Rev Cancer, vol.15, pp.701-712, 2015.

W. Wang, Z. X. Chen, D. Y. Guo, and Y. X. Tao, Regulation of prostate cancer by hormone-responsive G protein-coupled receptors, Pharmacol Ther, 2018.

I. V. Maly and W. A. Hofmann, Calcium and nuclear signaling in prostate cancer, Int J Mol Sci, vol.19, p.1237, 2018.

S. Li, S. Huang, and S. B. Peng, Overexpression of G protein-coupled receptors in cancer cells: involvement in tumor progression, Int J Oncol, vol.27, pp.1329-1368, 2005.

M. Sumitomo, R. Shen, M. Walburg, J. Dai, Y. Geng et al., Neutral endopeptidase inhibits prostate cancer cell migration by blocking focal adhesion kinase signaling, J Clin Invest, vol.106, pp.1399-407, 2000.

B. Collado, M. G. Sanchez, I. Diaz-laviada, J. C. Prieto, and M. J. Carmena, Vasoactive intestinal peptide (VIP) induces c-fos expression in LNCaP prostate cancer cells through a mechanism that involves Ca 2+ signalling. Implications in angiogenesis and neuroendocrine differentiation, Biochim Biophys Acta, vol.1744, pp.224-257, 2005.

D. Farini, A. Puglianiello, C. Mammi, G. Siracusa, and C. Moretti, Dual effect of pituitary adenylate cyclase activating polypeptide on prostate tumor LNCaP cells: short-and long-term exposure affect proliferation and neuroendocrine differentiation, Endocrinology, vol.144, pp.1631-1674, 2003.

M. G. Juarranz, O. Bolanos, I. Gutierrez-canas, E. A. Lerner, P. Robberecht et al., Neuroendocrine differentiation of the LNCaP prostate cancer cell line maintains the expression and function of VIP and PACAP receptors, Cell Signal, vol.13, pp.887-94, 2001.

I. Abasolo, L. M. Montuenga, and A. Calvo, Adrenomedullin prevents apoptosis in prostate cancer cells, Regul Pept, vol.133, pp.115-137, 2006.

M. Zhong, M. L. Boseman, A. C. Millena, and S. A. Khan, Oxytocin induces the migration of prostate cancer cells: involvement of the Gicoupled signaling pathway, Mol Cancer Res, vol.8, pp.1164-72, 2010.

N. E. Bhola and J. E. Grandis, Crosstalk between G-protein-coupled receptors and Epidermal growth factor receptor in cancer, Front. Biosci, vol.13, pp.1857-65, 2008.

D. Alexandre, C. Hautot, M. Mehio, L. Jeandel, M. Courel et al., The orexin type 1 receptor is overexpressed in advanced prostate cancer with a neuroendocrine differentiation, and mediates apoptosis, Eur J Cancer, vol.50, pp.2126-2159, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01960616

W. Malendowicz, M. Szyszka, A. Ziolkowska, M. Rucinski, and Z. Kwias, Elevated expression of orexin receptor 2 (HCRTR2) in benign prostatic hyperplasia is accompanied by lowered serum orexin A concentrations, Int J Mol Med, vol.27, pp.377-83, 2011.

S. Valiante, G. Liguori, S. Tafuri, L. M. Pavone, R. Campese et al., Expression and potential role of the peptide orexin-A in prostate cancer, Biochem Biophys Res Commun, vol.464, pp.1290-1296, 2015.

T. Tsunematsu and A. Yamanaka, The role of orexin/hypocretin in the central nervous system and peripheral tissues, Vitam Horm, vol.89, pp.19-33, 2012.

N. L. Graybill and V. Weissig, A review of orexin's unprecedented potential as a novel, highly-specific treatment for various localized and metastatic cancers, SAGE Open Med, vol.5, pp.1-9, 2017.

J. Wenzel, N. Grabinski, C. A. Knopp, A. Dendorfer, M. Ramanjaneya et al., Hypocretin/orexin increases the expression of steroidogenic enzymes in human adrenocortical NCI H295R cells, Am J Physiol Regul Integr Comp Physiol, vol.297, pp.1601-1610, 2009.

R. Spinazzi, M. Rucinski, G. Neri, L. K. Malendowicz, and G. G. Nussdorfer, Preproorexin and orexin receptors are expressed in cortisol-secreting adrenocortical adenomas, and orexins stimulate in vitro cortisol secretion and growth of tumor cells, J Clin Endocrinol Metab, vol.90, pp.3544-3553, 2005.

Y. Liu, Y. Zhao, and L. Guo, Effects of orexin A on glucose metabolism in human hepatocellular carcinoma in vitro via PI3K/Akt/mTOR-dependent and -independent mechanism, Mol Cell Endocrinol, vol.420, pp.208-224, 2016.

Y. Liu, Y. Zhao, S. Ju, and L. Guo, Orexin A upregulates the protein expression of OX1R and enhances the proliferation of SGC-7901 gastric cancer cells through the ERK signaling pathway, Int J Mol Med, vol.35, pp.539-584, 2015.

J. Wen, Y. Zhao, Y. Shen, and L. Guo, Effect of orexin A on apoptosis in BGC-823 gastric cancer cells via OX1R through the AKT signaling pathway, Mol Med Rep, vol.11, pp.3439-3483, 2015.

B. Bai, X. Chen, R. Zhang, X. Wang, Y. Jiang et al., Dual-agonist occupancy of orexin receptor 1 and cholecystokinin A receptor heterodimers decreases G-protein-dependent signaling and migration in the human colon cancer cell line HT-29, Biochim Biophys Acta, vol.1864, pp.1153-64, 2017.

G. Mazzocchi, L. K. Malendowicz, F. Aragona, P. Rebuffat, L. Gottardo et al., Human pheochromocytomas express orexin receptor type 2 gene and display an in vitro secretory response to orexins A and B, J Clin Endocrinol Metab, vol.86, pp.4818-4839, 2001.

T. Nanmoku, K. Isobe, T. Sakurai, A. Yamanaka, K. Takekoshi et al., Orexins suppress catecholamine synthesis and secretion in cultured PC12 cells, Biochem Biophys Res Commun, vol.274, pp.310-315, 2000.

P. Dehan, C. Canon, G. Trooskens, M. Rehli, C. Munaut et al., Expression of type 2 orexin receptor in human endometrium and its epigenetic silencing in endometrial cancer, J Clin Endocrinol Metab, vol.98, pp.1549-57, 2013.

M. Kantono and B. Guo, Inflammasomes and cancer: the dynamic role of the inflammasome in tumor development, Front Immunol, vol.8, p.1132, 2017.

C. Duffy, C. Yuan, L. Wisdorf, C. J. Billington, C. M. Kotz et al., Role of orexin A signaling in dietary palmitic acid-activated microglial cells, Neurosci Lett, vol.606, pp.140-144, 2015.

X. Xiong, R. E. White, L. Xu, L. Yang, X. Sun et al., Mitigation of murine focal cerebral ischemia by the hypocretin/orexin system is associated with reduced inflammation, Stroke, vol.44, pp.764-70, 2013.

Y. Ogawa, Y. Irukayama-tomobe, N. Murakoshi, M. Kiyama, Y. Ishikawa et al., Peripherally administered orexin improves survival of mice with endotoxin shock, Elife, vol.5, p.21055, 2016.

S. Chieffi, M. Carotenuto, V. Monda, A. Valenzano, I. Villano et al., Orexin system: the key for a healthy life, Front Physiol, vol.8, p.357, 2017.

C. Prochasson, M. Bergere, N. Messal, S. Dayot, V. Rebours et al., The hypothalamic neuropeptide, orexin, prevents chronic pancreatitis in cerulein mice model, Gastroenterology, vol.150, issue.16, pp.33110-33119, 2016.

F. J. Martínez-orozco, J. L. Vicario, I. Villalibre-valderrey, D. Andrés, C. Fernández-arquero et al., Narcolepsy with cataplexy and comorbid immunopathological diseases, J Sleep Res, vol.23, pp.414-423, 2014.

, Références bibliographiques A

A. R. Adamantidis, F. Zhang, A. M. Aravanis, K. Deisseroth, and L. D. Lecea, Neural substrates of awakening probed with optogenetic control of hypocretin neurons, Nature, vol.450, pp.420-424, 2007.

J. M. Adams and S. Cory, Bcl-2-regulated apoptosis: mechanism and therapeutic potential, Current Opinion in Immunology, vol.19, pp.488-496, 2007.

H. Akl, B. M. Badran, N. E. Zein, N. E. Zein, F. Bex et al., HTLV-I infection of WE17/10 CD4+ cell line leads to progressive alteration of Ca2+ influx that eventually results in loss of CD7 expression and activation of an antiapoptotic pathway involving AKT and BAD which paves the way for malignant transformation, Leukemia, vol.21, pp.788-796, 2007.

D. Alexandre, C. Hautot, M. Mehio, L. Jeandel, M. Courel et al., The orexin type 1 receptor is overexpressed in advanced prostate cancer with a neuroendocrine differentiation, and mediates apoptosis, European Journal of Cancer, vol.50, pp.2126-2133, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01960616

M. B. Amin, F. L. Greene, S. B. Edge, C. C. Compton, J. E. Gershenwald et al., The Eighth Edition AJCC Cancer Staging Manual: Continuing to build a bridge from a population-based to a more "personalized" approach to cancer staging, CA: a cancer journal for clinicians, vol.67, pp.93-99, 2017.

S. Ammoun, L. Johansson, M. E. Ekholm, T. Holmqvist, A. S. Danis et al., OX1 orexin receptors activate extracellular signal-regulated kinase in Chinese hamster ovary cells via multiple mechanisms: the role of Ca2+ influx in OX1 receptor signaling, Molecular Endocrinology, vol.20, pp.80-99, 2006.

P. H. Anborgh, J. L. Seachrist, L. B. Dale, and S. S. Ferguson, Receptor/beta-arrestin complex formation and the differential trafficking and resensitization of beta2-adrenergic and angiotensin II type 1A receptors, Molecular Endocrinology, vol.14, pp.2040-2053, 2000.

A. M. Arafat, P. Kaczmarek, M. Skrzypski, E. Pruszy?ska-oszma?ek, P. Ko?odziejski et al., Glucagon regulates orexin A secretion in humans and rodents, Diabetologia, vol.57, pp.2108-2116, 2014.

J. E. Bader, C. M. Deckert, N. Koglin, F. Pluder, K. Mörl et al., From transcription profile to expression: the signaling repertoire of the SK-N-MC neuroepithelioma cell-line, Journal of Receptor and Signal Transduction Research, vol.24, pp.257-282, 2004.

F. R. Balkwill, M. Capasso, and T. Hagemann, The tumor microenvironment at a glance, Journal of Cell Science, vol.125, pp.5591-5596, 2012.

E. J. Balthazar, A. J. Megibow, D. Hulnick, and D. P. Naidich, Carcinoma of the colon: detection and preoperative staging by CT, AJR. American journal of roentgenology, vol.150, pp.301-306, 1988.

M. L. Barreiro, R. Pineda, F. Gaytan, M. Archanco, M. A. Burrell et al., Pattern of orexin expression and direct biological actions of orexin-a in rat testis, Endocrinology, vol.146, pp.5164-5175, 2005.

M. L. Barreiro, R. Pineda, V. M. Navarro, M. Lopez, J. S. Suominen et al., Orexin 1 receptor messenger ribonucleic acid expression and stimulation of testosterone secretion by orexin-A in rat testis, Endocrinology, vol.145, pp.2297-2306, 2004.

R. Bar-shavit, M. Maoz, A. Kancharla, J. K. Nag, D. Agranovich et al., Protein-Coupled Receptors in Cancer, International Journal of Molecular Sciences, p.17, 2016.

M. I. Beig, B. W. Dampney, and P. Carrive, Both Ox1r and Ox2r orexin receptors contribute to the cardiovascular and locomotor components of the novelty stress response in the rat, Neuropharmacology, vol.89, pp.146-156, 2015.

J. Benito, Z. Zeng, M. Konopleva, and W. R. Wilson, Targeting hypoxia in the leukemia microenvironment, International Journal of Hematologic Oncology, vol.2, pp.279-288, 2013.

K. Biega?ska, P. Soko?owska, O. Jöhren, and J. B. Zawilska, Orexin A suppresses the growth of rat C6 glioma cells via a caspase-dependent mechanism, Journal of molecular neuroscience, vol.48, pp.706-712, 2012.

S. Bipat, M. S. Leeuwen, . Van, E. F. Comans, M. E. Pijl et al., Colorectal liver metastases: CT, MR imaging, and PET for diagnosis--meta-analysis, Radiology, vol.237, pp.123-131, 2005.

S. Bipat, S. S. Phoa, O. M. Delden, . Van, P. M. Bossuyt et al., Ultrasonography, computed tomography and magnetic resonance imaging for diagnosis and determining resectability of pancreatic adenocarcinoma: a meta-analysis, Journal of Computer Assisted Tomography, vol.29, pp.438-445, 2005.

A. Blais, G. Drouin, C. Chaumontet, T. Voisin, A. Couvelard et al., Impact of Orexin-A Treatment on Food Intake, Energy Metabolism and Body Weight in Mice, PLoS ONE, p.12, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01602768

M. Blanco, T. García-caballero, M. Fraga, R. Gallego, J. Cuevas et al., Cellular localization of orexin receptors in human adrenal gland, adrenocortical adenomas and pheochromocytomas, Regulatory Peptides, vol.104, pp.161-165, 2002.

M. A. Blasco, Telomeres and human disease: ageing, cancer and beyond, Nature Reviews. Genetics, vol.6, pp.611-622, 2005.

H. Boehmer and C. Daniel, Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer, Nature Reviews. Drug Discovery, vol.12, pp.51-63, 2013.

C. R. Boland and A. Goel, Microsatellite instability in colorectal cancer, Gastroenterology, vol.138, 2010.

S. L. Borgland, S. A. Taha, F. Sarti, H. L. Fields, and A. Bonci, Orexin A in the VTA is critical for the induction of synaptic plasticity and behavioral sensitization to cocaine, Neuron, vol.49, pp.589-601, 2006.

C. Boss and C. Roch, Recent trends in orexin research--2010 to, vol.25, pp.2875-2887, 2015.

M. G. Bourke, S. Salwa, K. J. Harrington, M. J. Kucharczyk, P. F. Forde et al., The emerging role of viruses in the treatment of solid tumours, Cancer Treatment Reviews, vol.37, pp.618-632, 2011.

B. Boutrel, P. J. Kenny, S. E. Specio, R. Martin-fardon, A. Markou et al., Role for hypocretin in mediating stress-induced reinstatement of cocaine-seeking behavior, Proceedings of the National Academy of Sciences, vol.102, pp.19168-19173, 2005.

A. Bouvier, Z. Uhry, V. Jooste, A. Drouillard, L. Remontet et al., Focus on an unusual rise in pancreatic cancer incidence in France, International Journal of Epidemiology, vol.46, pp.1764-1772, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01541663

M. Bouvier, Métaphores, nomenclature et nouveaux paradigmes de signalisation par les récepteurs couplés aux protéines G. médecine/sciences, vol.28, pp.801-803, 2012.

M. Bouvier, W. P. Hausdorff, A. De-blasi, B. F. O'dowd, B. K. Kobilka et al., Removal of phosphorylation sites from the beta 2-adrenergic receptor delays onset of agonist-promoted desensitization, Nature, vol.333, pp.370-373, 1988.

P. Bruhns, B. Iannascoli, P. England, D. A. Mancardi, N. Fernandez et al., Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses, Blood, vol.113, pp.3716-3725, 2009.
URL : https://hal.archives-ouvertes.fr/pasteur-00363931

T. F. Brust, J. M. Conley, and V. J. Watts, G?(i/o)-coupled receptor-mediated sensitization of adenylyl cyclase: 40 years later, European Journal of Pharmacology, vol.763, pp.223-232, 2015.

C. Bucci, P. Thomsen, P. Nicoziani, J. Mccarthy, and B. V. Deurs, Rab7: a key to lysosome biogenesis, Molecular Biology of the Cell, vol.11, pp.467-480, 2000.

D. L. Burkhart and J. Sage, Cellular mechanisms of tumour suppression by the retinoblastoma gene, Nature Reviews. Cancer, vol.8, pp.671-682, 2008.

M. E. Carter, A. Adamantidis, H. Ohtsu, K. Deisseroth, and L. D. Lecea, Sleep homeostasis modulates hypocretin-mediated sleep-to-wake transitions, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.29, pp.10939-10949, 2009.

U. Cavallaro and G. Christofori, Cell adhesion and signalling by cadherins and Ig-CAMs in cancer, Nature Reviews. Cancer, vol.4, pp.118-132, 2004.

B. P. Ceresa and L. E. Limbird, Mutation of an aspartate residue highly conserved among G-protein-coupled receptors results in nonreciprocal disruption of alpha 2-adrenergic receptor-G-protein interactions. A negative charge at amino acid residue 79 forecasts alpha 2A-adrenergic receptor sensitivity to allosteric modulation by monovalent cations and fully effective receptor/G-protein coupling, The Journal of Biological Chemistry, vol.269, pp.29557-29564, 1994.

X. Chang, Y. Zhao, S. Ju, and L. Guo, Orexin-A stimulates 3?-hydroxysteroid dehydrogenase expression and cortisol production in H295R human adrenocortical cells through the AKT pathway, International Journal of Molecular Medicine, vol.34, pp.1523-1528, 2014.

R. M. Chemelli, J. T. Willie, C. M. Sinton, J. K. Elmquist, T. Scammell et al., Narcolepsy in orexin Knockout Mice: Molecular Genetics of Sleep Regulation. Cell, vol.98, pp.437-451, 1999.

L. Chen, Y. Zhao, D. Zheng, S. Ju, Y. Shen et al., Orexin A Affects INS-1 Rat Insulinoma Cell Proliferation via Orexin Receptor 1 and the AKT Signaling Pathway, International Journal of Endocrinology, p.854623, 2013.

V. Cherezov, Lipidic cubic phase technologies for membrane protein structural studies, Current Opinion in Structural Biology, vol.21, pp.559-566, 2011.

S. Chieffi, M. Carotenuto, V. Monda, A. Valenzano, I. Villano et al., Orexin System: The Key for a Healthy Life, Frontiers in Physiology, vol.8, 2017.

L. S. Chin, M. C. Raynor, X. Wei, H. Q. Chen, and L. Li, Hrs interacts with sorting nexin 1 and regulates degradation of epidermal growth factor receptor, The Journal of Biological Chemistry, vol.276, pp.7069-7078, 2001.

E. G. Chiorean, V. Hoff, D. D. Reni, M. Arena, F. P. Infante et al., CA19-9 decrease at 8 weeks as a predictor of overall survival in a randomized phase III trial (MPACT) of weekly nab-paclitaxel plus gemcitabine versus gemcitabine alone in patients with metastatic pancreatic cancer, Annals of Oncology: Official Journal of the European Society for Medical Oncology, vol.27, pp.654-660, 2016.

D. E. Clapham and E. J. Neer, G protein beta gamma subunits, Annual Review of Pharmacology and Toxicology, vol.37, pp.167-203, 1997.

L. Clifford, B. W. Dampney, and P. Carrive, Spontaneously hypertensive rats have more orexin neurons in their medial hypothalamus than normotensive rats, Experimental Physiology, vol.100, pp.388-398, 2015.

P. J. Coleman and J. J. Renger, Orexin receptor antagonists: a review of promising compounds patented since, Expert Opinion on Therapeutic Patents, vol.20, pp.307-324, 2006.

, Gastroenterologie Clinique Et Biologique, vol.22, pp.205-226, 1998.

A. Couvelard, V. Rebours, C. Prochasson, M. Bergere, N. Messal et al., The hypothalamic neuropeptide, orexin, prevents chronic pancreatitis in cerulein mice model, Pancreatology, vol.16, pp.11-12, 2016.

A. Cowppli-bony, Z. Uhry, L. Remontet, N. Voirin, A. Guizard et al., Survival of solid cancer patients in France, 1989-2013: a population-based study. European journal of cancer prevention: the official journal of the European Cancer Prevention Organisation (ECP), vol.26, pp.461-468, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01540069

C. Cremolini, F. Loupakis, C. Antoniotti, C. Lupi, E. Sensi et al., FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first-line treatment of patients with metastatic colorectal cancer: updated overall survival and molecular subgroup analyses of the open-label, phase 3 TRIBE study, The Lancet. Oncology, vol.16, pp.1306-1315, 2015.

M. Curto and A. I. Mcclatchey, Nf2/Merlin: a coordinator of receptor signalling and intercellular contact, British Journal of Cancer, vol.98, pp.256-262, 2008.

A. M. Cypess, S. Lehman, G. Williams, I. Tal, D. Rodman et al., Identification and importance of brown adipose tissue in adult humans, The New England Journal of Medicine, vol.360, pp.1509-1517, 2009.

J. G. Darker, R. A. Porter, D. S. Eggleston, D. Smart, S. J. Brough et al., Structure-activity analysis of truncated orexin-A analogues at the orexin-1 receptor, Bioorganic & Medicinal Chemistry Letters, vol.11, pp.737-740, 2001.

D. Darmoul, V. Gratio, H. Devaud, F. Peiretti, and M. Laburthe, Activation of proteinase-activated receptor 1 promotes human colon cancer cell proliferation through epidermal growth factor receptor transactivation, Molecular cancer research, vol.2, pp.514-522, 2004.

D. Darmoul, J. C. Marie, H. Devaud, V. Gratio, and M. Laburthe, Initiation of human colon cancer cell proliferation by trypsin acting at protease-activated receptor-2, British Journal of Cancer, vol.85, pp.772-779, 2001.

Y. Date, Y. Ueta, H. Yamashita, H. Yamaguchi, S. Matsukura et al., Orexins, orexigenic hypothalamic peptides, interact with autonomic, neuroendocrine and neuroregulatory systems, Proceedings of the National Academy of Sciences of the United States of America, vol.96, pp.748-753, 1999.

C. V. Dayas, T. M. Mcgranahan, R. Martin-fardon, and F. Weiss, Stimuli linked to ethanol availability activate hypothalamic CART and orexin neurons in a reinstatement model of relapse, Biological Psychiatry, vol.63, pp.152-157, 2008.

S. Dayot, D. Speisky, A. Couvelard, P. Bourgoin, V. Gratio et al., In vitro , in vivo and ex vivo demonstration of the antitumoral role of hypocretin-1/orexin-A and almorexant in pancreatic ductal adenocarcinoma, Oncotarget, vol.9, pp.6952-6967, 2018.

C. De-angelis, R. F. Brizzi, and R. Pellicano, Endoscopic ultrasonography for pancreatic cancer: current and future perspectives, Journal of Gastrointestinal Oncology, vol.4, pp.220-230, 2013.

P. Dehan, C. Canon, G. Trooskens, M. Rehli, C. Munaut et al., Expression of type 2 orexin receptor in human endometrium and its epigenetic silencing in endometrial cancer, The Journal of Clinical Endocrinology and Metabolism, vol.98, pp.1549-1557, 2013.

J. R. Delpero, P. Bachellier, N. Regenet, Y. P. Le-treut, F. Paye et al., Pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: a French multicentre prospective evaluation of resection margins in 150 evaluable specimens. HPB: the official journal of the International Hepato Pancreato Biliary Association, vol.16, pp.20-33, 2014.

J. E. Digby, J. Chen, J. Y. Tang, H. Lehnert, R. N. Matthews et al., Orexin receptor expression in human adipose tissue: effects of orexin-A and orexin-B, Journal of Endocrinology, vol.191, pp.129-136, 2006.

D. Duguay, E. Bélanger-nelson, V. Mongrain, A. Beben, A. Khatchadourian et al., Dynein light chain Tctex-type 1 modulates orexin signaling through its interaction with orexin 1 receptor, PloS One, vol.6, p.26430, 2011.

R. Dunn, D. A. Klos, A. S. Adler, and L. Hicke, The C2 domain of the Rsp5 ubiquitin ligase binds membrane phosphoinositides and directs ubiquitination of endosomal cargo, The Journal of Cell Biology, vol.165, pp.135-144, 2004.

M. Ehrström, T. Gustafsson, A. Finn, A. Kirchgessner, P. Grybäck et al., Inhibitory Effect of Exogenous Orexin A on Gastric Emptying, Plasma Leptin, and the Distribution of Orexin and Orexin Receptors in the Gut and Pancreas in Man, The Journal of Clinical Endocrinology & Metabolism, vol.90, pp.2370-2377, 2005.

M. Ehrström, F. Levin, A. L. Kirchgessner, P. T. Schmidt, L. M. Hilsted et al., Stimulatory effect of endogenous orexin A on gastric emptying and acid secretion independent of gastrin, Regulatory Peptides, vol.132, pp.9-16, 2005.

A. El-firar, T. Voisin, C. Rouyer-fessard, M. A. Ostuni, A. Couvineau et al., Discovery of a functional immunoreceptor tyrosine-based switch motif in a 7-transmembrane-spanning receptor: role in the orexin receptor OX1R-driven apoptosis, FASEB journal: official publication of the Federation of American Societies for Experimental Biology, vol.23, pp.4069-4080, 2009.

R. A. España, J. R. Melchior, D. C. Roberts, and S. R. Jones, Hypocretin 1/orexin A in the ventral tegmental area enhances dopamine responses to cocaine and promotes cocaine self-administration, Psychopharmacology, vol.214, pp.415-426, 2011.

I. V. Estabrooke, M. T. Mccarthy, E. Ko, T. C. Chou, R. M. Chemelli et al., Fos Expression in Orexin Neurons Varies with Behavioral State, Journal of Neuroscience, vol.21, pp.1656-1662, 2001.

N. A. Evans, D. A. Groarke, J. Warrack, C. J. Greenwood, K. Dodgson et al., Visualizing differences in ligand-induced beta-arrestin-GFP interactions and trafficking between three recently characterized G protein-coupled receptors, Journal of Neurochemistry, vol.77, pp.476-485, 2001.

E. R. Fearon and B. Vogelstein, A genetic model for colorectal tumorigenesis, Cell, vol.61, pp.759-767, 1990.

J. Ferlay, C. Partensky, and F. Bray, More deaths from pancreatic cancer than breast cancer in the EU by, 2016.

, Acta Oncologica, vol.55, pp.1158-1160

D. Fessart, M. Simaan, B. Zimmerman, J. Comeau, F. F. Hamdan et al., Src-dependent phosphorylation of beta2-adaptin dissociates the beta-arrestin-AP-2 complex, Journal of Cell Science, vol.120, pp.1723-1732, 2007.

S. Fichtner-feigl, R. Kesselring, and W. Strober, Chronic inflammation and the development of malignancy in the GI tract, Trends in Immunology, vol.36, pp.451-459, 2015.

I. J. Fidler, The pathogenesis of cancer metastasis: the "seed and soil" hypothesis revisited, Nature Reviews. Cancer, vol.3, pp.453-458, 2003.

I. Flores, R. Benetti, and M. A. Blasco, Telomerase regulation and stem cell behaviour, Current Opinion in Cell Biology, vol.18, pp.254-260, 2006.

K. Fosgerau and T. Hoffmann, Peptide therapeutics: current status and future directions, Drug Discovery Today, vol.20, pp.122-128, 2015.

A. R. French, D. K. Tadaki, S. K. Niyogi, and D. A. Lauffenburger, Intracellular trafficking of epidermal growth factor family ligands is directly influenced by the pH sensitivity of the receptor/ligand interaction, The Journal of Biological Chemistry, vol.270, pp.4334-4340, 1995.

D. M. Frey, R. A. Droeser, C. T. Viehl, I. Zlobec, A. Lugli et al., High frequency of tumor-infiltrating FOXP3(+) regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients, International Journal of Cancer, vol.126, pp.2635-2643, 2010.

W. H. Fridman, F. Pagès, C. Sautès-fridman, and J. Galon, The immune contexture in human tumours: impact on clinical outcome, Nature Reviews. Cancer, vol.12, pp.298-306, 2012.

N. Fujiki, Y. Yoshida, B. Ripley, K. Honda, E. Mignot et al., Changes in CSF hypocretin-1 (orexin A) levels in rats across 24 hours and in response to food deprivation, NeuroReport, vol.12, p.993, 2001.

H. Funato, A. L. Tsai, J. T. Willie, Y. Kisanuki, S. C. Williams et al., Enhanced orexin receptor-2 signaling prevents diet-induced obesity and improves leptin sensitivity, Cell Metabolism, vol.9, pp.64-76, 2009.

R. M. Gage, K. A. Kim, T. T. Cao, and M. V. Zastrow, A transplantable sorting signal that is sufficient to mediate rapid recycling of G protein-coupled receptors, The Journal of Biological Chemistry, vol.276, pp.44712-44720, 2001.

R. M. Gage, E. A. Matveeva, S. W. Whiteheart, and M. V. Zastrow, Type I PDZ ligands are sufficient to promote rapid recycling of G Protein-coupled receptors independent of binding to N-ethylmaleimide-sensitive factor, The Journal of Biological Chemistry, vol.280, pp.3305-3313, 2005.

T. F. Gajewski, H. Schreiber, and Y. Fu, Innate and adaptive immune cells in the tumor microenvironment, Nature Immunology, vol.14, pp.1014-1022, 2013.

C. Galés and M. Bouvier, New talk between receptor and trimeric G proteins: "an intertwined body dance, 2007.

, Medecine Sciences: M/S, vol.23, pp.1031-1034

G. Gaudriault, D. Nouel, C. Dal-farra, A. Beaudet, and J. P. Vincent, Receptor-induced internalization of selective peptidic mu and delta opioid ligands, The Journal of Biological Chemistry, vol.272, pp.2880-2888, 1997.

K. M. Gautvik, L. Lecea, . De, V. T. Gautvik, P. E. Danielson et al., Overview of the most prevalent hypothalamus-specific mRNAs, as identified by directional tag PCR subtraction, vol.93, pp.8733-8738, 1996.

C. Gentili, R. Boland, and A. Russo-de-boland, Implication of Gbetagamma proteins and c-SRC tyrosine kinase in parathyroid hormone-induced signal transduction in rat enterocytes, The Journal of Endocrinology, vol.188, pp.69-78, 2006.

D. Georgescu, V. Zachariou, M. Barrot, M. Mieda, J. T. Willie et al., Involvement of the lateral hypothalamic peptide orexin in morphine dependence and withdrawal, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.23, pp.3106-3111, 2003.

V. Gocheva, H. Wang, B. B. Gadea, T. Shree, K. E. Hunter et al., IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion, Genes & Development, vol.24, pp.241-255, 2010.

E. Göncz, M. Z. Strowski, C. Grötzinger, K. W. Nowak, P. Kaczmarek et al., Orexin-A inhibits glucagon secretion and gene expression through a Foxo1-dependent pathway, Endocrinology, vol.149, pp.1618-1626, 2008.

A. L. Gotter, A. J. Roecker, R. Hargreaves, P. J. Coleman, C. J. Winrow et al., Orexin receptors as therapeutic drug targets, Progress in Brain Research, vol.198, pp.163-188, 2012.

E. F. Grady, A. M. Garland, P. D. Gamp, M. Lovett, D. G. Payan et al., Delineation of the endocytic pathway of substance P and its seven-transmembrane domain NK1 receptor, Molecular Biology of the Cell, vol.6, pp.509-524, 1995.

W. M. Grady and J. M. Carethers, Genomic and epigenetic instability in colorectal cancer pathogenesis, Gastroenterology, vol.135, pp.1079-1099, 2008.

V. Gratio, F. Walker, T. Lehy, M. Laburthe, and D. Darmoul, Aberrant expression of proteinase-activated receptor 4 promotes colon cancer cell proliferation through a persistent signaling that involves Src and ErbB-2 kinase, International Journal of Cancer, vol.124, pp.1517-1525, 2009.

N. L. Graybill and V. Weissig, A review of orexin's unprecedented potential as a novel, highly-specific treatment for various localized and metastatic cancers, SAGE open medicine, vol.5, 2017.

S. I. Grivennikov, F. R. Greten, and M. Karin, Immunity, inflammation, and cancer. Cell, vol.140, pp.883-899, 2010.

V. V. Gurevich and E. V. Gurevich, Molecular Mechanisms of GPCR Signaling: A Structural Perspective, International Journal of Molecular Sciences, vol.18, 2017.

J. J. Hagan, R. A. Leslie, S. Patel, M. L. Evans, T. A. Wattam et al., Orexin A activates locus coeruleus cell firing and increases arousal in the rat, Proceedings of the National Academy of Sciences of the United States of America, vol.96, pp.10911-10916, 1999.

S. A. Hahn, M. Schutte, A. T. Hoque, C. A. Moskaluk, L. T. Costa et al., DPC4, a candidate tumor suppressor gene at human chromosome 18q21, Science, vol.1, pp.350-353, 1996.

D. Hanahan and L. M. Coussens, Accessories to the crime: functions of cells recruited to the tumor microenvironment, Cancer Cell, vol.21, pp.309-322, 2012.

D. Hanahan and J. Folkman, Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis, Cell, vol.86, pp.353-364, 1996.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

A. C. Hanyaloglu and M. V. Zastrow, Regulation of GPCRs by endocytic membrane trafficking and its potential implications, Annual Review of Pharmacology and Toxicology, vol.48, pp.537-568, 2008.

A. C. Hanyaloglu and M. V. Zastrow, Regulation of GPCRs by endocytic membrane trafficking and its potential implications, Annual Review of Pharmacology and Toxicology, vol.48, pp.537-568, 2008.

J. Hara, C. T. Beuckmann, T. Nambu, J. T. Willie, R. M. Chemelli et al., Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity, Neuron, vol.30, pp.345-354, 2001.

D. M. Harris, V. L. Go, J. R. Reeve, and S. V. Wu, Stimulation of amylase release by Orexin is mediated by Orexin 2 receptor in AR42J cells, Pancreas, vol.25, pp.405-410, 2002.

G. C. Harris, M. Wimmer, and G. Aston-jones, A role for lateral hypothalamic orexin neurons in reward seeking, Nature, vol.437, pp.556-559, 2005.

W. P. Hausdorff, M. Bouvier, B. F. O'dowd, G. P. Irons, M. G. Caron et al., Phosphorylation sites on two domains of the beta 2-adrenergic receptor are involved in distinct pathways of receptor desensitization, The Journal of Biological Chemistry, vol.264, pp.12657-12665, 1989.

A. C. Haynes, B. Jackson, H. Chapman, M. Tadayyon, A. Johns et al., A selective orexin-1 receptor antagonist reduces food consumption in male and female rats, Regulatory Peptides, vol.96, pp.45-51, 2000.

I. Hindmarch, E. Legangneux, N. Stanley, S. Emegbo, and J. Dawson, A double-blind, placebo-controlled investigation of the residual psychomotor and cognitive effects of zolpidem-MR in healthy elderly volunteers, British Journal of Clinical Pharmacology, vol.62, pp.538-545, 2006.

K. P. Hofmann, P. Scheerer, P. W. Hildebrand, H. Choe, J. H. Park et al., A G protein-coupled receptor at work: the rhodopsin model, Trends in Biochemical Sciences, vol.34, pp.540-552, 2009.

J. A. Hollander, Q. Lu, M. D. Cameron, T. M. Kamenecka, and P. J. Kenny, Insular hypocretin transmission regulates nicotine reward, Proceedings of the National Academy of Sciences of the United States of America, vol.105, pp.19480-19485, 2008.

M. Hondo, K. Nagai, K. Ohno, Y. Kisanuki, J. T. Willie et al., Histamine-1 receptor is not required as a downstream effector of orexin-2 receptor in maintenance of basal sleep/wake states, Acta Physiologica, vol.198, pp.287-294, 2010.

Q. Hu, D. Wu, W. Chen, Z. Yan, and Y. Shi, Proteolytic processing of the caspase-9 zymogen is required for apoptosome-mediated activation of caspase-9, The Journal of Biological Chemistry, vol.288, pp.15142-15147, 2013.

S. Huang, Y. E. Dai, Y. Lee, L. Chiou, and L. Hwang, Orexins depolarize rostral ventrolateral medulla neurons and increase arterial pressure and heart rate in rats mainly via orexin 2 receptors, The Journal of Pharmacology and Experimental Therapeutics, vol.334, pp.522-529, 2010.

Z. Huang, W. Qu, W. Li, T. Mochizuki, N. Eguchi et al., Arousal effect of orexin A depends on activation of the histaminergic system, Proceedings of the National Academy of Sciences of the United States of America, vol.98, pp.9965-9970, 2001.

M. J. Huber, Y. Fan, E. Jiang, F. Zhu, R. A. Larson et al., Increased activity of the orexin system in the paraventricular nucleus contributes to salt-sensitive hypertension, American Journal of Physiology. Heart and Circulatory Physiology, vol.313, pp.1075-1086, 2017.

S. Hubert and J. Abastado, Les étapes précoces du processus métastatique. médecine/sciences, vol.30, pp.378-384, 2014.

L. Hui and Y. Chen, Tumor microenvironment: Sanctuary of the devil, Cancer Letters, vol.368, pp.7-13, 2015.

J. H. Hurley and S. D. Emr, The ESCRT complexes: structure and mechanism of a membrane-trafficking network, Annual Review of Biophysics and Biomolecular Structure, vol.35, pp.277-298, 2006.

T. R. Hynes, L. Tang, S. M. Mervine, J. L. Sabo, E. A. Yost et al., Visualization of G protein betagamma dimers using bimolecular fluorescence complementation demonstrates roles for both beta and gamma in subcellular targeting, The Journal of Biological Chemistry, vol.279, pp.30279-30286, 2004.

S. Inman, Novel Combination Shows Promising Responses in Pancreatic Cancer, 2017.

J. Iovanna, M. C. Mallmann, A. Gonçalves, O. Turrini, and J. Dagorn, Current Knowledge on Pancreatic Cancer, Frontiers in Oncology, vol.2, 2012.

Y. Irukayama-tomobe, Y. Ogawa, H. Tominaga, Y. Ishikawa, N. Hosokawa et al., Nonpeptide orexin type-2 receptor agonist ameliorates narcolepsy-cataplexy symptoms in mouse models, Proceedings of the National Academy of Sciences of the United States of America, vol.114, pp.5731-5736, 2017.

, J

K. A. Jacobson and S. Costanzi, New insights for drug design from the X-ray crystallographic structures of Gprotein-coupled receptors, Molecular Pharmacology, vol.82, pp.361-371, 2012.

Y. Jiang, H. B. Mackley, E. T. Kimchi, J. Zhu, N. Gusani et al., Phase I dose escalation study of capecitabine and erlotinib concurrent with radiation in locally advanced pancreatic cancer, Cancer Chemotherapy and Pharmacology, vol.74, pp.205-210, 2014.

O. Jöhren, S. J. Neidert, M. Kummer, A. Dendorfer, and P. Dominiak, Prepro-orexin and orexin receptor mRNAs are differentially expressed in peripheral tissues of male and female rats, Endocrinology, vol.142, pp.3324-3331, 2001.

S. Jones, X. Zhang, D. W. Parsons, J. Lin, R. J. Leary et al., Core signaling pathways in human pancreatic cancers revealed by global genomic analyses, Science, vol.321, pp.1801-1806, 2008.

J. A. Joyce and J. W. Pollard, Microenvironmental regulation of metastasis, Nature Reviews. Cancer, vol.9, pp.239-252, 2009.

B. Jupp, E. Krstew, G. Dezsi, and A. J. Lawrence, Discrete cue-conditioned alcohol-seeking after protracted abstinence: pattern of neural activation and involvement of orexin? receptors, British Journal of Pharmacology, vol.162, pp.880-889, 2011.

S. K. Kamarajah, W. R. Burns, T. L. Frankel, C. S. Cho, and H. Nathan, Validation of the American Joint Commission on Cancer (AJCC) 8th Edition Staging System for Patients with Pancreatic Adenocarcinoma: A Surveillance, Epidemiology and End Results (SEER) Analysis. Annals of Surgical Oncology, vol.24, pp.2023-2030, 2017.

D. Kamato, L. Thach, R. Bernard, V. Chan, W. Zheng et al., Structure, Function, Pharmacology, and Therapeutic Potential of the G Protein, G?/q,11. Frontiers in Cardiovascular Medicine, vol.2, 2015.

L. Karhu, A. Turku, and H. Xhaard, Modeling of the OX1R-orexin-A complex suggests two alternative binding modes, BMC Structural Biology, p.15, 2015.

M. M. Karnani, J. Apergis-schoute, A. Adamantidis, L. T. Jensen, L. Lecea et al., Activation of central orexin/hypocretin neurons by dietary amino acids, Neuron, vol.72, pp.616-629, 2011.

A. E. Karnoub, A. B. Dash, A. P. Vo, A. Sullivan, M. W. Brooks et al., Mesenchymal stem cells within tumour stroma promote breast cancer metastasis, Nature, vol.449, pp.557-563, 2007.

E. Karteris, J. Chen, and H. S. Randeva, Expression of human prepro-orexin and signaling characteristics of orexin receptors in the male reproductive system, The Journal of Clinical Endocrinology and Metabolism, vol.89, pp.1957-1962, 2004.

A. J. Kastin and V. Akerstrom, Orexin A but Not Orexin B Rapidly Enters Brain from Blood by Simple Diffusion, Journal of Pharmacology and Experimental Therapeutics, vol.289, pp.219-223, 1999.

D. Keri and P. Barth, Reprogramming G protein coupled receptor structure and function, Current Opinion in Structural Biology, vol.51, pp.187-194, 2018.

R. C. Kern, D. S. Kang, and J. L. Benovic, Arrestin2/clathrin interaction is regulated by key N-and C-terminal regions in arrestin2, Biochemistry, vol.48, pp.7190-7200, 2009.

K. Kessenbrock, V. Plaks, and Z. Werb, Matrix metalloproteinases: regulators of the tumor microenvironment, Cell, vol.141, pp.52-67, 2010.

E. Khoury, L. Nikolajev, M. Simaan, Y. Namkung, and S. A. Laporte, Differential Regulation of Endosomal GPCR/?-Arrestin Complexes and Trafficking by MAPK, The Journal of Biological Chemistry, vol.289, pp.23302-23317, 2014.

A. L. Kirchgessner and M. Liu, Orexin synthesis and response in the gut, Neuron, vol.24, pp.941-951, 1999.

F. Klemm and J. A. Joyce, Microenvironmental regulation of therapeutic response in cancer, Trends in Cell Biology, vol.25, pp.198-213, 2015.

M. Kobashi, Y. Furudono, R. Matsuo, and T. Yamamoto, Central orexin facilitates gastric relaxation and contractility in rats, Neuroscience Letters, vol.332, pp.171-174, 2002.

L. F. Kolakowski, GCRDb: a G-protein-coupled receptor database, Receptors & Channels, vol.2, pp.1-7, 1994.

V. Konieczny, M. V. Keebler, and C. W. Taylor, Spatial organization of intracellular Ca2+ signals, Seminars in Cell & Developmental Biology, vol.23, pp.172-180, 2012.

Z. K. Krowicki, M. A. Burmeister, H. Berthoud, R. T. Scullion, K. Fuchs et al., Orexins in rat dorsal motor nucleus of the vagus potently stimulate gastric motor function, American Journal of Physiology. Gastrointestinal and Liver Physiology, vol.283, pp.465-472, 2002.

J. P. Kukkonen, Regulation of receptor-coupling to (multiple) G proteins. A challenge for basic research and drug discovery, Receptors & Channels, vol.10, pp.167-183, 2004.

J. P. Kukkonen, A ménage à trois made in heaven: G-protein-coupled receptors, lipids and TRP channels, Cell Calcium, vol.50, pp.9-26, 2011.

J. P. Kukkonen and K. E. Akerman, Orexin receptors couple to Ca2+ channels different from store-operated Ca2+ channels, Neuroreport, vol.12, pp.2017-2020, 2001.

J. P. Kukkonen and C. S. Leonard, Orexin/hypocretin receptor signalling cascades, British Journal of Pharmacology, vol.171, pp.314-331, 2014.

M. Lang, R. M. Söll, F. Dürrenberger, F. M. Dautzenberg, and A. G. Beck-sickinger, Structure-activity studies of orexin a and orexin B at the human orexin 1 and orexin 2 receptors led to orexin 2 receptor selective and orexin 1 receptor preferring ligands, Journal of Medicinal Chemistry, vol.47, pp.1153-1160, 2004.

S. A. Laporte, R. H. Oakley, J. A. Holt, L. S. Barak, and M. G. Caron, The interaction of beta-arrestin with the AP-2 adaptor is required for the clustering of beta 2-adrenergic receptor into clathrin-coated pits, The Journal of Biological Chemistry, vol.275, pp.23120-23126, 2000.

S. A. Laporte, R. H. Oakley, J. Zhang, J. A. Holt, S. S. Ferguson et al., The beta2-adrenergic receptor/betaarrestin complex recruits the clathrin adaptor AP-2 during endocytosis, vol.96, pp.3712-3717, 1999.

J. Larkin, V. Chiarion-sileni, R. Gonzalez, J. J. Grob, C. L. Cowey et al., Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma, The New England Journal of Medicine, vol.373, pp.23-34, 2015.

A. J. Lawrence, M. S. Cowen, H. Yang, F. Chen, and B. Oldfield, The orexin system regulates alcoholseeking in rats, British Journal of Pharmacology, vol.148, pp.752-759, 2006.

L. Lecea, . De, T. S. Kilduff, C. Peyron, X. Gao et al., The hypocretins: Hypothalamus-specific peptides with neuroexcitatory activity, vol.95, pp.322-327, 1998.

J. Lee, E. Bang, K. Chae, J. Kim, D. W. Lee et al., Solution structure of a new hypothalamic neuropeptide, human hypocretin-2/orexin-B, European Journal of Biochemistry, vol.266, pp.831-839, 1999.

Y. Lee, M. Tsai, T. Li, Y. E. Dai, S. Huang et al., Spontaneously hypertensive rats have more orexin neurons in the hypothalamus and enhanced orexinergic input and orexin 2 receptorassociated nitric oxide signalling in the rostral ventrolateral medulla, Experimental Physiology, vol.100, pp.993-1007, 2015.

R. J. Lefkowitz and E. J. Whalen, beta-arrestins: traffic cops of cell signaling, Current Opinion in Cell Biology, vol.16, pp.162-168, 2004.

G. M. Leinninger, D. M. Opland, Y. Jo, M. Faouzi, L. Christensen et al., Leptin action via neurotensin neurons controls orexin, the mesolimbic dopamine system and energy balance, Cell Metabolism, vol.14, pp.313-323, 2011.

M. G. Lesage, J. L. Perry, C. M. Kotz, D. Shelley, and W. A. Corrigall, Nicotine self-administration in the rat: effects of hypocretin antagonists and changes in hypocretin mRNA, Psychopharmacology, vol.209, pp.203-212, 2010.

J. Lewis, L. J. Eiben, D. L. Nelson, J. I. Cohen, K. E. Nichols et al., Distinct interactions of the X-linked lymphoproliferative syndrome gene product SAP with cytoplasmic domains of members of the CD2 receptor family, Clinical Immunology, vol.100, pp.15-23, 2001.

J. Li, Z. Hu, and L. D. Lecea, The hypocretins/orexins: integrators of multiple physiological functions, British Journal of Pharmacology, vol.171, pp.332-350, 2014.

T. Li, J. Y. Chen, S. Huang, Y. E. Dai, and L. Hwang, Cardiovascular pressor effects of orexins in the dorsomedial hypothalamus, European Journal of Pharmacology, vol.818, pp.343-350, 2018.

X. Li, L. Liang, L. Huang, and S. Cai, Standard chemotherapy with cetuximab for treatment of colorectal cancer, World Journal of Gastroenterology, vol.21, pp.7022-7035, 2015.

H. Lin, G-protein-coupled receptors and their (Bio) chemical significance win 2012 Nobel Prize in Chemistry, Biomedical Journal, vol.36, pp.118-124, 2013.

L. Lin, J. Faraco, R. Li, H. Kadotani, W. Rogers et al., The sleep disorder canine narcolepsy is caused by a mutation in the hypocretin (orexin) receptor 2 gene, Cell, vol.98, pp.365-376, 1999.

I. Litosch, Decoding G?q signaling, Life Sciences, vol.152, pp.99-106, 2016.

R. Liu, A. N. Pol, and G. K. Van-den-&-aghajanian, Hypocretins (orexins) regulate serotonin neurons in the dorsal raphe nucleus by excitatory direct and inhibitory indirect actions, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.22, pp.9453-9464, 2002.

Y. Liu, Y. Zhao, and L. Guo, Effects of orexin A on glucose metabolism in human hepatocellular carcinoma in vitro via PI3K/Akt/mTOR-dependent and -independent mechanism, Molecular and Cellular Endocrinology, vol.420, pp.208-216, 2016.

Y. Liu, Y. Zhao, S. Ju, and L. Guo, Orexin A upregulates the protein expression of OX1R and enhances the proliferation of SGC-7901 gastric cancer cells through the ERK signaling pathway, International Journal of Molecular Medicine, vol.35, pp.539-545, 2015.

M. J. Lohse, J. L. Benovic, J. Codina, M. G. Caron, and R. J. Lefkowitz, beta-Arrestin: a protein that regulates beta-adrenergic receptor function, Science, vol.248, pp.1547-1550, 1990.

G. W. Louis, G. M. Leinninger, C. J. Rhodes, and M. G. Myers, Direct innervation and modulation of orexin neurons by lateral hypothalamic LepRb neurons, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.30, pp.11278-11287, 2010.

S. W. Lowe, E. Cepero, and G. Evan, Intrinsic tumour suppression, Nature, vol.432, pp.307-315, 2004.

P. E. Lund, R. Shariatmadari, A. Uustare, M. Detheux, M. Parmentier et al., The orexin OX1 receptor activates a novel Ca2+ influx pathway necessary for coupling to phospholipase C, The Journal of Biological Chemistry, vol.275, pp.30806-30812, 2000.

L. M. Luttrell, T. Biesen, . Van, B. E. Hawes, W. J. Koch et al., G-protein-coupled receptors and their regulation: activation of the MAP kinase signaling pathway by G-protein-coupled receptors, Advances in Second Messenger and Phosphoprotein Research, vol.31, pp.263-277, 1997.

L. M. Luttrell, B. E. Hawes, T. Biesen, . Van, D. K. Luttrell et al., Role of c-Src tyrosine kinase in G protein-coupled receptor-and Gbetagamma subunit-mediated activation of mitogen-activated protein kinases, The Journal of Biological Chemistry, vol.271, pp.19443-19450, 1996.

H. T. Lynch and A. Chapelle, Hereditary colorectal cancer, The New England Journal of Medicine, vol.348, pp.919-932, 2003.

J. Lytton, Na+/Ca2+ exchangers: three mammalian gene families control Ca2+ transport, The Biochemical Journal, vol.406, pp.365-382, 2007.

P. E. Macdonald, J. W. Joseph, and P. Rorsman, Glucose-sensing mechanisms in pancreatic beta-cells, Philosophical Transactions of the Royal Society of London. Series B, Biological Sciences, vol.360, pp.2211-2225, 2005.

J. Magga, G. Bart, C. Oker-blom, J. P. Kukkonen, K. E. Akerman et al., Agonist potency differentiates G protein activation and Ca2+ signalling by the orexin receptor type 1, Biochemical Pharmacology, vol.71, pp.827-836, 2006.

S. V. Mahler, R. J. Smith, D. E. Moorman, G. C. Sartor, and G. Aston-jones, Multiple roles for orexin/hypocretin in addiction, Progress in brain research, vol.198, pp.79-121, 2012.

C. C. Malbon, Frizzleds: new members of the superfamily of G-protein-coupled receptors, Frontiers in Bioscience: A Journal and Virtual Library, vol.9, pp.1048-1058, 2004.

G. Malleo, L. Maggino, C. R. Ferrone, G. Marchegiani, M. Mino-kenudson et al., Number of Examined Lymph Nodes and Nodal Status Assessment in Distal Pancreatectomy for Body/Tail Ductal Adenocarcinoma, Annals of Surgery, 2018.

M. Malumbres and M. Barbacid, Cell cycle, CDKs and cancer: a changing paradigm, Nature Reviews Cancer, vol.9, pp.153-166, 2009.

A. Manglik, T. H. Kim, M. Masureel, C. Altenbach, Z. Yang et al., Structural insights into the dynamic process of ?2-adrenergic receptor signaling, Cell, vol.161, pp.1101-1111, 2015.

J. J. Maoret, Y. Anini, C. Rouyer-fessard, D. Gully, and M. Laburthe, Neurotensin and a non-peptide neurotensin receptor antagonist control human colon cancer cell growth in cell culture and in cells xenografted into nude mice, International Journal of Cancer, vol.80, pp.448-454, 1999.

A. Marchese, M. M. Paing, B. R. Temple, and J. Trejo, Protein-Coupled Receptor Sorting to Endosomes and Lysosomes, Annual Review of Pharmacology and Toxicology, vol.48, pp.601-629, 2008.

I. Mármol, C. Sánchez-de-diego, A. Pradilla-dieste, E. Cerrada, and M. J. Rodriguez-yoldi, Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer, International Journal of Molecular Sciences, p.18, 2017.

E. J. Martin, M. E. Hernandez, and L. F. Hayward, Blockade of orexin receptors attenuates the cardiovascular response to air-jet stress in spontaneously hypertensive rats, Autonomic Neuroscience: Basic & Clinical, vol.201, pp.8-16, 2016.

S. L. Maude, N. Frey, P. A. Shaw, R. Aplenc, D. M. Barrett et al., Chimeric antigen receptor T cells for sustained remissions in leukemia, The New England Journal of Medicine, vol.371, pp.1507-1517, 2014.

S. L. Maude, T. W. Laetsch, J. Buechner, S. Rives, M. Boyer et al., Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia, New England Journal of Medicine, vol.378, pp.439-448, 2018.

M. V. Maus, S. A. Grupp, D. L. Porter, and C. H. June, Antibody-modified T cells: CARs take the front seat for hematologic malignancies, Blood, vol.123, pp.2625-2635, 2014.

G. Mazzocchi, L. K. Malendowicz, F. Aragona, P. Rebuffat, L. Gottardo et al., Human pheochromocytomas express orexin receptor type 2 gene and display an in vitro secretory response to orexins A and B, The Journal of Clinical Endocrinology and Metabolism, vol.86, pp.4818-4821, 2001.

V. A. Medina and E. S. Rivera, Histamine receptors and cancer pharmacology, British Journal of Pharmacology, vol.161, pp.755-767, 2010.

A. Melcher, K. Parato, C. M. Rooney, and J. C. Bell, Thunder and lightning: immunotherapy and oncolytic viruses collide, Molecular Therapy: The Journal of the American Society of Gene Therapy, vol.19, pp.1008-1016, 2011.

N. Messal, N. Fernandez, S. Dayot, V. Gratio, P. Nicole et al., Ectopic expression of OX1R in ulcerative colitis mediates anti-inflammatory effect of orexin-A, Biochimica Et Biophysica Acta. Molecular Basis of Disease, vol.1864, pp.3618-3628, 2018.

M. Mieda, E. Hasegawa, Y. Y. Kisanuki, C. M. Sinton, M. Yanagisawa et al., Differential roles of orexin receptor-1 and -2 in the regulation of non-REM and REM sleep, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.31, pp.6518-6526, 2011.

E. Mignot, G. J. Lammers, B. Ripley, M. Okun, S. Nevsimalova et al., The role of cerebrospinal fluid hypocretin measurement in the diagnosis of narcolepsy and other hypersomnias, Archives of Neurology, vol.59, pp.1553-1562, 2002.

G. Milligan and E. Kostenis, Heterotrimeric G-proteins: a short history, British Journal of Pharmacology, p.147, 2006.

T. Mochizuki, E. Arrigoni, J. N. Marcus, E. L. Clark, M. Yamamoto et al., Orexin receptor 2 expression in the posterior hypothalamus rescues sleepiness in narcoleptic mice, Proceedings of the National Academy of Sciences of the United States of America, vol.108, pp.4471-4476, 2011.

I. Modolell, L. Guarner, and J. R. Malagelada, Vagaries of clinical presentation of pancreatic and biliary tract cancer, Annals of Oncology: Official Journal of the European Society for Medical Oncology, vol.10, pp.82-84, 1999.

T. W. Moody, P. Moreno, and R. T. Jensen, Neuropeptides as lung cancer growth factors, Peptides, vol.72, pp.106-111, 2015.

T. W. Moody, I. Ramos-alvarez, and R. T. Jensen, Neuropeptide G Protein-Coupled Receptors as Oncotargets, Frontiers in Endocrinology, p.9, 2018.

R. H. Moore, E. E. Millman, V. Godines, N. A. Hanania, T. M. Tran et al., Salmeterol stimulation dissociates beta2-adrenergic receptor phosphorylation and internalization, American Journal of Respiratory Cell and Molecular Biology, vol.36, pp.254-261, 2007.

D. E. Moorman and G. Aston-jones, Orexin-1 receptor antagonism decreases ethanol consumption and preference selectively in high-ethanol--preferring Sprague--Dawley rats, Alcohol, vol.43, pp.379-386, 2009.

V. P. Muniz, J. M. Barnes, S. Paliwal, X. Zhang, X. Tang et al., The ARF tumor suppressor inhibits tumor cell colonization independent of p53 in a novel mouse model of pancreatic ductal adenocarcinoma metastasis, Molecular cancer research: MCR, vol.9, pp.867-877, 2011.

J. F. Nabhan, H. Pan, and Q. Lu, Arrestin domain-containing protein 3 recruits the NEDD4 E3 ligase to mediate ubiquitination of the beta2-adrenergic receptor, EMBO reports, vol.11, pp.605-611, 2010.

A. Nagarajan, P. Malvi, and N. Wajapeyee, Oncogene-directed alterations in cancer cell metabolism, Trends in cancer, vol.2, pp.365-377, 2016.

J. A. Nagy, S. Chang, S. Shih, A. M. Dvorak, and H. F. Dvorak, Heterogeneity of the tumor vasculature, Seminars in Thrombosis and Hemostasis, vol.36, pp.321-331, 2010.

M. Nakabayashi, T. Suzuki, K. Takahashi, K. Totsune, Y. Muramatsu et al., Orexin-A expression in human peripheral tissues, Molecular and Cellular Endocrinology, vol.205, pp.43-50, 2003.

T. Nakagawa, T. Sakurai, T. Nishioka, and K. Touhara, , 2005.

, Tanpakushitsu Kakusan Koso. Protein, Nucleic Acid, Enzyme, vol.50, pp.1563-1570

T. Nakamura, K. Uramura, T. Nambu, T. Yada, K. Goto et al., Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system, Brain Research, vol.873, pp.181-187, 2000.

M. Narita, Y. Nagumo, S. Hashimoto, M. Narita, J. Khotib et al., Direct involvement of orexinergic systems in the activation of the mesolimbic dopamine pathway and related behaviors induced by morphine, The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, vol.26, pp.398-405, 2006.

E. Näslund, M. Ehrström, J. Ma, P. M. Hellström, and A. L. Kirchgessner, Localization and effects of orexin on fasting motility in the rat duodenum, American Journal of Physiology. Gastrointestinal and Liver Physiology, vol.282, pp.470-479, 2002.

P. Nicole, P. Couvineau, N. Jamin, T. Voisin, and A. Couvineau, Crucial role of the orexin-B C-terminus in the induction of OX1 receptor-mediated apoptosis: analysis by alanine scanning, molecular modelling and sitedirected mutagenesis, British Journal of Pharmacology, vol.172, pp.5211-5223, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01432232

K. W. Nowak, M. Z. Strowski, M. M. Switonska, P. Kaczmarek, V. Singh et al., Evidence that orexins A and B stimulate insulin secretion from rat pancreatic islets via both receptor subtypes, International Journal of Molecular Medicine, vol.15, pp.969-972, 2005.

R. H. Oakley, S. A. Laporte, J. A. Holt, M. G. Caron, and L. S. Barak, Differential affinities of visual arrestin, beta arrestin1, and beta arrestin2 for G protein-coupled receptors delineate two major classes of receptors, The Journal of Biological Chemistry, vol.275, pp.17201-17210, 2000.

W. M. Oldham and H. E. Hamm, Heterotrimeric G protein activation by G-protein-coupled receptors, Nature Reviews. Molecular Cell Biology, vol.9, pp.60-71, 2008.

R. Ouedraogo, E. Näslund, and A. L. Kirchgessner, Glucose regulates the release of orexin-a from the endocrine pancreas, Diabetes, vol.52, pp.111-117, 2003.

M. J. Overman, R. Mcdermott, J. L. Leach, S. Lonardi, H. Lenz et al., Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): an open-label, multicentre, phase 2 study, The Lancet. Oncology, vol.18, pp.1182-1191, 2017.

K. Palczewski, T. Kumasaka, T. Hori, C. A. Behnke, H. Motoshima et al., Crystal structure of rhodopsin: A G protein-coupled receptor, Science, vol.289, pp.739-745, 2000.

G. Pándy-szekeres, C. Munk, T. M. Tsonkov, S. Mordalski, K. Harpsøe et al., GPCRdb in 2018: adding GPCR structure models and ligands, Nucleic Acids Research, vol.46, pp.440-446, 2018.

J. Park, H. Shim, A. Na, J. Bae, S. Im et al., Orexin A regulates plasma insulin and leptin levels in a time-dependent manner following a glucose load in mice, Diabetologia, vol.58, pp.1542-1550, 2015.

J. I. Partanen, A. I. Nieminen, and J. Klefstrom, 3D view to tumor suppression: Lkb1, polarity and the arrest of oncogenic c-Myc, Cell Cycle, vol.8, pp.716-724, 2009.

R. K. Pasumarthi, L. R. Reznikov, and J. Fadel, Activation of orexin neurons by acute nicotine, European Journal of Pharmacology, vol.535, pp.172-176, 2006.

S. P. Patel and R. Kurzrock, PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy, Molecular Cancer Therapeutics, vol.14, pp.847-856, 2015.

C. E. Perez-leighton, K. Boland, C. J. Billington, and C. M. Kotz, High and low activity rats: elevated intrinsic physical activity drives resistance to diet-induced obesity in non-bred rats, Obesity, vol.21, pp.353-360, 2013.

J. Perroy, S. Pontier, P. G. Charest, M. Aubry, and M. Bouvier, Real-time monitoring of ubiquitination in living cells by BRET, Nature Methods, vol.1, pp.203-208, 2004.

C. Peyron, J. Faraco, W. Rogers, B. Ripley, S. Overeem et al., A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains, Nature Medicine, vol.6, pp.991-997, 2000.

K. L. Pierce and R. J. Lefkowitz, Classical and new roles of ?-arrestins in the regulation of G-PROTEIN-COUPLED receptors, Nature Reviews Neuroscience, vol.2, pp.727-733, 2001.

J. Pin, T. Galvez, and L. Prézeau, Evolution, structure, and activation mechanism of family 3/C G-proteincoupled receptors, Pharmacology & Therapeutics, vol.98, pp.325-354, 2003.

M. F. Pino, A. Divoux, A. V. Simmonds, S. R. Smith, and L. M. Sparks, Investigating the effects of Orexin-A on thermogenesis in human deep neck brown adipose tissue, International Journal of Obesity, vol.41, pp.1646-1653, 2005.

M. S. Pino and D. C. Chung, The chromosomal instability pathway in colon cancer, Gastroenterology, vol.138, pp.2059-2072, 2010.

J. A. Pitcher, N. J. Freedman, and R. J. Lefkowitz, G protein-coupled receptor kinases, Annual Review of Biochemistry, vol.67, pp.653-692, 1998.

A. Plaza-zabala, Á. Flores, R. Maldonado, and F. Berrendero, Hypocretin/orexin signaling in the hypothalamic paraventricular nucleus is essential for the expression of nicotine withdrawal, Biological Psychiatry, vol.71, pp.214-223, 2012.

Y. Prabhu and L. Eichinger, The Dictyostelium repertoire of seven transmembrane domain receptors, European Journal of Cell Biology, vol.85, pp.937-946, 2006.

R. T. Premont and R. R. Gainetdinov, Physiological roles of G protein-coupled receptor kinases and arrestins, Annual Review of Physiology, vol.69, pp.511-534, 2007.

R. T. Premont, J. Inglese, and R. J. Lefkowitz, Protein kinases that phosphorylate activated G protein-coupled receptors, FASEB journal: official publication of the Federation of American Societies for Experimental Biology, vol.9, pp.175-182, 1995.

T. J. Price, M. Peeters, T. W. Kim, J. Li, S. Cascinu et al., Panitumumab versus cetuximab in patients with chemotherapy-refractory wild-type KRAS exon 2 metastatic colorectal cancer (ASPECCT): a randomised, multicentre, open-label, non-inferiority phase 3 study, The Lancet. Oncology, vol.15, pp.569-579, 2014.

J. Putula, P. M. Turunen, L. Johansson, J. Näsman, R. Ra et al., Orexin/hypocretin receptor chimaeras reveal structural features important for orexin peptide distinction, FEBS Letters, vol.585, pp.1368-1374, 2012.

B. Qian and J. W. Pollard, Macrophage diversity enhances tumor progression and metastasis, Cell, vol.141, pp.39-51, 2010.

D. Quail and J. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nature medicine, vol.19, pp.1423-1437, 2013.

D. Quarta, E. Valerio, D. M. Hutcheson, G. Hedou, and C. Heidbreder, The orexin-1 receptor antagonist SB-334867 reduces amphetamine-evoked dopamine outflow in the shell of the nucleus accumbens and decreases the expression of amphetamine sensitization, Neurochemistry International, vol.56, pp.11-15, 2010.

C. Raiborg, K. G. Bache, A. Mehlum, E. Stang, and H. Stenmark, Hrs recruits clathrin to early endosomes, The EMBO journal, vol.20, pp.5008-5021, 2001.

C. Raiborg, B. Bremnes, A. Mehlum, D. J. Gillooly, A. D'arrigo et al., FYVE and coiled-coil domains determine the specific localisation of Hrs to early endosomes, Journal of Cell Science, vol.114, pp.2255-2263, 2001.

M. Raica, A. M. Cimpean, and D. Ribatti, Angiogenesis in pre-malignant conditions, European Journal of Cancer, vol.45, pp.1924-1934, 1990.

S. Rajagopal and S. K. Shenoy, GPCR desensitization: Acute and prolonged phases, Cellular Signalling, vol.41, pp.9-16, 2018.

S. P. Raman, S. Reddy, M. J. Weiss, L. L. Manos, J. L. Cameron et al., Impact of the time interval between MDCT imaging and surgery on the accuracy of identifying metastatic disease in patients with pancreatic cancer, AJR. American journal of roentgenology, vol.204, pp.37-42, 2015.

M. Ramanjaneya, A. C. Conner, J. Chen, P. R. Stanfield, and H. S. Randeva, Orexins stimulate steroidogenic acute regulatory protein expression through multiple signaling pathways in human adrenal H295R cells, Endocrinology, vol.149, pp.4106-4115, 2008.

S. G. Rasmussen, H. Choi, J. J. Fung, E. Pardon, P. Casarosa et al., Structure of a nanobody-stabilized active state of the ?(2) adrenoceptor, Nature, vol.469, pp.175-180, 2011.

R. V. Rebois, M. Robitaille, C. Galés, D. J. Dupré, A. Baragli et al., Heterotrimeric G proteins form stable complexes with adenylyl cyclase and Kir3.1 channels in living cells, Journal of Cell Science, vol.119, pp.2807-2818, 2006.

M. S. Redston, C. Caldas, A. B. Seymour, R. H. Hruban, L. Costa et al., mutations in pancreatic carcinoma and evidence of common involvement of homocopolymer tracts in DNA microdeletions, Cancer Research, vol.54, pp.3025-3033, 1994.

N. P. Restifo, M. E. Dudley, and S. A. Rosenberg, Adoptive immunotherapy for cancer: harnessing the T cell response, Nature Reviews. Immunology, vol.12, pp.269-281, 2012.

J. D. Robishaw and C. H. Berlot, Translating G protein subunit diversity into functional specificity, Current Opinion in Cell Biology, vol.16, pp.206-209, 2004.

C. Rodemer and V. Haucke, Clathrin/AP-2-dependent endocytosis: a novel playground for the pharmacological toolbox?, Handbook of Experimental Pharmacology, pp.105-122, 2008.

T. Roehrs and T. Roth, Insomnia pharmacotherapy, Neurotherapeutics: The Journal of the American Society for Experimental NeuroTherapeutics, vol.9, pp.728-738, 2012.

D. M. Rosenbaum, V. Cherezov, M. A. Hanson, S. G. Rasmussen, F. S. Thian et al., GPCR engineering yields high-resolution structural insights into beta2-adrenergic receptor function, Science, vol.318, pp.1266-1273, 2007.

D. M. Rosenbaum, C. Zhang, J. Lyons, R. Holl, D. Aragao et al., Structure and Function of an Irreversible Agonist-?2 Adrenoceptor complex, Nature, vol.469, pp.236-240, 2011.

P. Rouet-benzineb, C. Rouyer-fessard, A. Jarry, V. Avondo, C. Pouzet et al., Orexins Acting at Native OX1 Receptor in Colon Cancer and Neuroblastoma Cells or at Recombinant OX1 Receptor Suppress Cell Growth by Inducing Apoptosis, Journal of Biological Chemistry, vol.279, pp.45875-45886, 2004.

G. E. Rovati, V. Capra, and R. R. Neubig, The highly conserved DRY motif of class A G protein-coupled receptors: beyond the ground state, Molecular Pharmacology, vol.71, pp.959-964, 2007.

B. Ruffell and L. M. Coussens, Macrophages and therapeutic resistance in cancer, Cancer Cell, vol.27, pp.462-472, 2015.

C. R. Rush, D. L. Armstrong, J. A. Ali, and P. J. Pazzaglia, Benzodiazepine-receptor ligands in humans: acute performance-impairing, subject-rated and observer-rated effects, Journal of Clinical Psychopharmacology, vol.18, pp.154-165, 1998.

Y. Saiki and A. Horii, Molecular pathology of pancreatic cancer, Pathology International, vol.64, pp.10-19, 2014.

T. Sakurai, The role of orexin in motivated behaviours, Nature Reviews. Neuroscience, vol.15, pp.719-731, 2014.

T. Sakurai, A. Amemiya, M. Ishii, I. Matsuzaki, R. M. Chemelli et al., Orexins and Orexin Receptors: A Family of Hypothalamic Neuropeptides and G Protein-Coupled Receptors that Regulate Feeding Behavior, Cell, vol.92, pp.573-585, 1998.

C. B. Saper, P. M. Fuller, N. P. Pedersen, J. Lu, and T. E. Scammell, Sleep state switching, vol.68, pp.1023-1042, 2010.

C. B. Saper, T. E. Scammell, and J. Lu, Hypothalamic regulation of sleep and circadian rhythms, Nature, vol.437, pp.1257-1263, 2005.

A. Schuld, J. Hebebrand, F. Geller, and T. Pollmächer, Increased body-mass index in patients with narcolepsy, Lancet, vol.355, pp.1274-1275, 2000.

J. L. Seachrist, P. H. Anborgh, and S. S. Ferguson, beta 2-adrenergic receptor internalization, endosomal sorting, and plasma membrane recycling are regulated by rab GTPases, The Journal of Biological Chemistry, vol.275, pp.27221-27228, 2000.

J. L. Seachrist and S. S. Ferguson, Regulation of G protein-coupled receptor endocytosis and trafficking by Rab GTPases, Life Sciences, vol.74, pp.225-235, 2003.

D. Sellayah, P. Bharaj, and D. Sikder, Orexin is required for brown adipose tissue development, differentiation, and function, Cell Metabolism, vol.14, pp.478-490, 2011.

D. Sellayah and D. Sikder, Orexin receptor-1 mediates brown fat developmental differentiation, Adipocyte, vol.1, pp.58-63, 2012.

I. Z. Shahid, A. A. Rahman, and P. M. Pilowsky, Intrathecal orexin A increases sympathetic outflow and respiratory drive, enhances baroreflex sensitivity and blocks the somato-sympathetic reflex, British Journal of Pharmacology, vol.162, pp.961-973, 2011.

I. Z. Shahid, A. A. Rahman, and P. M. Pilowsky, Orexin A in rat rostral ventrolateral medulla is pressor, sympatho-excitatory, increases barosensitivity and attenuates the somato-sympathetic reflex, British Journal of Pharmacology, vol.165, pp.2292-2303, 2012.

R. Sharf, D. J. Guarnieri, J. R. Taylor, and R. J. Dileone, Orexin mediates morphine place preference, but not morphine-induced hyperactivity or sensitization, Brain Research, vol.1317, pp.24-32, 2010.

R. Sharf, M. Sarhan, C. E. Brayton, D. J. Guarnieri, J. R. Taylor et al., Orexin signaling via the orexin 1 receptor mediates operant responding for food reinforcement, Biological Psychiatry, vol.67, pp.753-760, 2010.

J. W. Shay and W. E. Wright, The use of telomerized cells for tissue engineering, Nature Biotechnology, vol.18, pp.22-23, 2000.

Y. Shen, Y. Zhao, D. Zheng, X. Chang, S. Ju et al., Effects of orexin A on GLUT4 expression and lipid content via MAPK signaling in 3T3-L1 adipocytes, The Journal of Steroid Biochemistry and Molecular Biology, vol.138, pp.376-383, 2013.

S. K. Shenoy and R. J. Lefkowitz, Trafficking patterns of beta-arrestin and G protein-coupled receptors determined by the kinetics of beta-arrestin deubiquitination, The Journal of Biological Chemistry, vol.278, pp.14498-14506, 2003.

T. Shirasaka, M. Nakazato, S. Matsukura, M. Takasaki, and H. Kannan, Sympathetic and cardiovascular actions of orexins in conscious rats, The American Journal of Physiology, vol.277, pp.1780-1785, 1999.

L. M. Shlapatska, S. V. Mikhalap, A. G. Berdova, O. M. Zelensky, T. J. Yun et al., CD150 association with either the SH2-containing inositol phosphatase or the SH2-containing protein tyrosine phosphatase is regulated by the adaptor protein SH2D1A, Journal of Immunology, vol.166, pp.5480-5487, 1950.

J. R. Shoblock, N. Welty, L. Aluisio, I. Fraser, S. T. Motley et al., Selective blockade of the orexin-2 receptor attenuates ethanol self-administration, place preference, and reinstatement, Psychopharmacology, vol.215, pp.191-203, 2011.

J. M. Siegel, Narcolepsy: A Key Role for Hypocretins (Orexins), Cell, vol.98, pp.409-412, 1999.

M. Sieminski, J. Szypenbejl, and E. Partinen, Sleep, and Blood Pressure, vol.20, p.79, 2018.

M. Skrzypski, M. Billert, K. W. Nowak, and M. Z. Strowski, The role of orexin in controlling the activity of the adipo-pancreatic axis, The Journal of Endocrinology, vol.238, pp.95-108, 2018.

M. Skrzypski, P. Kaczmarek, T. T. Le, T. Wojciechowicz, E. Pruszy?ska-oszmalek et al., Effects of orexin A on proliferation, survival, apoptosis and differentiation of 3T3-L1 preadipocytes into mature adipocytes, FEBS letters, vol.586, pp.4157-4164, 2012.

M. Skrzypski, N. Khajavi, S. Mergler, M. Billert, D. Szczepankiewicz et al., Orexin A modulates INS-1E cell proliferation and insulin secretion via extracellular signal-regulated kinase and transient receptor potential channels, Journal of Physiology and Pharmacology: An Official Journal of the Polish Physiological Society, vol.67, pp.643-652, 2016.

A. Sorkin and M. Von-zastrow, Signal transduction and endocytosis: close encounters of many kinds, Nature Reviews. Molecular Cell Biology, vol.3, pp.600-614, 2002.

M. M. Stanford, J. C. Bell, and M. J. Vähä-koskela, Novel oncolytic viruses: riding high on the next wave?, Cytokine & Growth Factor Reviews, vol.21, pp.177-183, 2010.

H. Stenmark, Rab GTPases as coordinators of vesicle traffic, Nature Reviews. Molecular Cell Biology, vol.10, pp.513-525, 2009.

E. M. Stoffel and F. Kastrinos, Familial colorectal cancer, beyond Lynch syndrome, Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association, vol.12, pp.1059-1068, 2014.

M. A. Swartz, N. Iida, E. W. Roberts, S. Sangaletti, M. H. Wong et al., Tumor microenvironment complexity: emerging roles in cancer therapy, Cancer Research, vol.72, pp.2473-2480, 2012.

M. Szyszka, L. Paschke, M. Tyczewska, M. Rucinski, P. Grabowska et al., Lack of expression of preproorexin and orexin receptors genes in human normal and prostate cancer cell lines, Folia Histochemica Et Cytobiologica, vol.53, pp.333-341, 2015.

T. Tachibana, H. Onodera, T. Tsuruyama, A. Mori, S. Nagayama et al., Increased intratumor Valpha24-positive natural killer T cells: a prognostic factor for primary colorectal carcinomas, Clinical Cancer Research: An Official Journal of the American Association for Cancer Research, vol.11, pp.7322-7327, 2005.

S. Taheri, D. Sunter, C. Dakin, S. Moyes, L. Seal et al., Diurnal variation in orexin A immunoreactivity and prepro-orexin mRNA in the rat central nervous system, Neuroscience Letters, vol.279, pp.109-112, 2000.

N. Takahashi, T. Okumura, H. Yamada, and Y. Kohgo, Stimulation of gastric acid secretion by centrally administered orexin-A in conscious rats, Biochemical and Biophysical Research Communications, vol.254, pp.623-627, 1999.

T. Takai, T. Takaya, M. Nakano, H. Akutsu, A. Nakagawa et al., Orexin-A is composed of a highly conserved C-terminal and a specific, hydrophilic N-terminal region, revealing the structural basis of specific recognition by the orexin-1 receptor, Journal of Peptide Science, vol.12, pp.443-454, 2006.

R. Taximaimaiti, X. Abuliken, M. Maihemuti, D. Abudujilile, and H. Abudulimu, Elevated Expression of Ox2R in Cervical Cancers and Placentas of Uyghur Women in Xinjiang, China. Asian Pacific journal of cancer prevention, vol.17, pp.4959-4963, 2016.

T. C. Thannickal, R. Y. Moore, R. Nienhuis, L. Ramanathan, S. Gulyani et al., Reduced number of hypocretin neurons in human narcolepsy, Neuron, vol.27, pp.469-474, 2000.

X. Tréton, E. Pedruzzi, C. Guichard, Y. Ladeiro, S. Sedghi et al., Combined NADPH oxidase 1 and interleukin 10 deficiency induces chronic endoplasmic reticulum stress and causes ulcerative colitis-like disease in mice, PloS One, vol.9, p.101669, 2014.

P. M. Turunen, M. E. Ekholm, P. Somerharju, and J. P. Kukkonen, Arachidonic acid release mediated by OX1 orexin receptors, British Journal of Pharmacology, vol.159, pp.212-221, 2010.

Z. Uhry, L. Remontet, M. Colonna, A. Belot, P. Grosclaude et al., Cancer incidence estimation at a district level without a national registry: a validation study for 24 cancer sites using French health insurance and registry data, Cancer Epidemiology, vol.37, pp.99-114, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02446485

A. Umar, C. R. Boland, J. P. Terdiman, S. Syngal, A. Chapelle et al., Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability, Journal of the National Cancer Institute, vol.96, pp.261-268, 2004.

P. Valent, D. Bonnet, R. De-maria, T. Lapidot, M. Copland et al., Cancer stem cell definitions and terminology: the devil is in the details, Nature Reviews. Cancer, vol.12, pp.767-775, 2012.

S. Valiante, G. Liguori, S. Tafuri, L. M. Pavone, R. Campese et al., Expression and potential role of the peptide orexin-A in prostate cancer, Biochemical and Biophysical Research Communications, vol.464, pp.1290-1296, 2015.

B. Vogelstein and K. W. Kinzler, Cancer genes and the pathways they control, Nature Medicine, vol.10, pp.789-799, 2004.

T. Voisin, A. E. Firar, M. Fasseu, C. Rouyer-fessard, V. Descatoire et al., Aberrant Expression of OX1 Receptors for Orexins in Colon Cancers and Liver Metastases: an Openable Gate to Apoptosis, Cancer Research, vol.71, pp.3341-3351, 2011.

T. Voisin, A. E. Firar, C. Rouyer-fessard, V. Gratio, and M. Laburthe, A hallmark of immunoreceptor, the tyrosine-based inhibitory motif ITIM, is present in the G protein-coupled receptor OX1R for orexins and drives apoptosis: a novel mechanism, The FASEB Journal, vol.22, 1993.

T. Voisin, P. Rouet-benzineb, N. Reuter, and M. Laburthe, Orexins and their receptors: structural aspects and role in peripheral tissues, Cellular and Molecular Life Sciences (CMLS), vol.60, pp.72-87, 2003.

Z. M. Volovyk, M. J. Wolf, S. V. Prasad, and H. A. Rockman, Agonist-stimulated beta-adrenergic receptor internalization requires dynamic cytoskeletal actin turnover, The Journal of Biological Chemistry, vol.281, pp.9773-9780, 2006.

J. N. Walker, R. Ramracheya, Q. Zhang, P. R. Johnson, M. Braun et al., Regulation of glucagon secretion by glucose: paracrine, intrinsic or both? Diabetes, Obesity & Metabolism, vol.13, issue.1, pp.95-105, 2011.

Y. Wang, Y. Zhou, K. Szabo, C. R. Haft, and J. Trejo, Down-regulation of protease-activated receptor-1 is regulated by sorting nexin 1, Molecular Biology of the Cell, vol.13, pp.1965-1976, 2002.

Y. Wang, F. He, F. Feng, X. Liu, G. Dong et al., Notch signaling determines the M1 versus M2 polarization of macrophages in antitumor immune responses, Cancer Research, vol.70, pp.4840-4849, 2010.

T. Warne, M. J. Serrano-vega, J. G. Baker, R. Moukhametzianov, P. C. Edwards et al., Structure of a beta1-adrenergic G-protein-coupled receptor, Nature, vol.454, pp.486-491, 2008.

D. J. Weisenberger, K. D. Siegmund, M. Campan, J. Young, T. I. Long et al., CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer, Nature Genetics, vol.38, pp.787-793, 2006.

J. Wen, Y. Zhao, and L. Guo, Orexin A induces autophagy in HCT-116 human colon cancer cells through the ERK signaling pathway, International Journal of Molecular Medicine, vol.37, pp.126-132, 2016.

J. Wen, Y. Zhao, Y. Shen, and L. Guo, Effect of orexin A on apoptosis in BGC-823 gastric cancer cells via OX1R through the AKT signaling pathway, Molecular Medicine Reports, vol.11, pp.3439-3444, 2015.

J. Wenzel, N. Grabinski, C. A. Knopp, A. Dendorfer, M. Ramanjaneya et al., , 2009.

, Hypocretin/orexin increases the expression of steroidogenic enzymes in human adrenocortical NCI H295R cells

, American Journal of Physiology. Regulatory, Integrative and Comparative Physiology, vol.297, pp.1601-1609

J. L. Whistler, J. Enquist, A. Marley, J. Fong, F. Gladher et al., Modulation of postendocytic sorting of G protein-coupled receptors, Science, vol.297, pp.615-620, 2002.

G. H. Williams and K. Stoeber, The cell cycle and cancer, The Journal of Pathology, vol.226, pp.352-364, 2012.

R. L. Williams and S. Urbé, The emerging shape of the ESCRT machinery, Nature Reviews. Molecular Cell Biology, vol.8, pp.355-368, 2007.

S. J. Wilson, D. J. Nutt, C. Alford, S. V. Argyropoulos, D. S. Baldwin et al., British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders, Journal of Psychopharmacology, vol.24, pp.1577-1601, 2010.

C. J. Winrow and J. J. Renger, Discovery and development of orexin receptor antagonists as therapeutics for insomnia, British Journal of Pharmacology, vol.171, pp.283-293, 2014.

C. J. Winrow, K. Q. Tanis, D. R. Reiss, A. M. Rigby, J. M. Uslaner et al., Orexin receptor antagonism prevents transcriptional and behavioral plasticity resulting from stimulant exposure, Neuropharmacology, vol.58, pp.185-194, 2010.

T. Wojciechowicz, M. Skrzypski, D. Szczepankiewicz, I. Hertig, P. A. Ko?odziejski et al., Original Research: Orexins A and B stimulate proliferation and differentiation of porcine preadipocytes, Experimental Biology and Medicine, pp.1786-1795, 2016.

J. D. Wolchok, H. Kluger, M. K. Callahan, M. A. Postow, N. A. Rizvi et al., Nivolumab plus ipilimumab in advanced melanoma, The New England Journal of Medicine, vol.369, pp.122-133, 2013.

B. L. Wolfe and J. Trejo, Clathrin-dependent mechanisms of G protein-coupled receptor endocytosis, Traffic, vol.8, pp.462-470, 2007.

S. K. Wong and .. , G protein selectivity is regulated by multiple intracellular regions of GPCRs, Neuro-Signals, vol.12, pp.1-12, 2003.

X. ,

F. Xiao, M. Jiang, D. Du, C. Xia, J. Wang et al., Orexin A regulates cardiovascular responses in stress-induced hypertensive rats, Neuropharmacology, vol.67, pp.16-24, 2013.

F. Xu, H. Wu, V. Katritch, G. W. Han, K. A. Jacobson et al., Structure of an agonist-bound human A2A adenosine receptor, Science, vol.332, pp.322-327, 2011.

S. Xu, T. Furukawa, N. Kanai, M. Sunamura, and A. Horii, Abrogation of DUSP6 by hypermethylation in human pancreatic cancer, Journal of Human Genetics, vol.50, pp.159-167, 2005.

T. Xu, Y. Yang, R. Ward, L. Gao, and Y. Liu, Orexin receptors: Multi-functional therapeutic targets for sleeping disorders, eating disorders, drug addiction, cancers and other physiological disorders, Cellular Signalling, vol.25, pp.2413-2423, 2013.

Z. D. Xu, X. Zhang, and L. Scott, Regulation of G protein-coupled receptor trafficking, Acta Physiologica, vol.190, pp.39-45, 2007.

A. Yamanaka, C. T. Beuckmann, J. T. Willie, J. Hara, N. Tsujino et al., Hypothalamic orexin neurons regulate arousal according to energy balance in mice, Neuron, vol.38, pp.701-713, 2003.

M. Ychou, J. Duffour, A. Kramar, C. Debrigode, S. Gourgou et al., Individual 5-FU dose adaptation in metastatic colorectal cancer: results of a phase II study using a bimonthly pharmacokinetically intensified LV5FU2 regimen, Cancer Chemotherapy and Pharmacology, vol.52, pp.282-290, 2003.

M. Ychou, J. Raoul, J. Douillard, S. Gourgou-bourgade, R. Bugat et al., A phase III randomised trial of LV5FU2 + irinotecan versus LV5FU2 alone in adjuvant high-risk colon cancer (FNCLCC Accord02/FFCD9802), Annals of Oncology: Official Journal of the European Society for Medical Oncology, vol.20, pp.674-680, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00415127

J. Yin, K. Babaoglu, C. A. Brautigam, L. Clark, Z. Shao et al., Structure and ligandbinding mechanism of the human OX1 and OX2 orexin receptors, Nature Structural & Molecular Biology, vol.23, pp.293-299, 2016.

J. Yin, J. C. Mobarec, P. Kolb, and D. M. Rosenbaum, Crystal structure of the human OX2 orexin receptor bound to the insomnia drug suvorexant, Nature, vol.519, pp.247-250, 2015.

B. F. Zamarron and W. Chen, Dual roles of immune cells and their factors in cancer development and progression, International Journal of Biological Sciences, vol.7, pp.651-658, 2011.

A. Zapf-colby and J. M. Olefsky, Nerve growth factor processing and trafficking events following TrkAmediated endocytosis, Endocrinology, vol.139, pp.3232-3240, 1998.

M. V. Zastrow, Mechanisms regulating membrane trafficking of G protein-coupled receptors in the endocytic pathway, Life Sciences, vol.74, pp.217-224, 2003.

M. Zerial and H. Mcbride, Rab proteins as membrane organizers, Nature Reviews. Molecular Cell Biology, vol.2, pp.107-117, 2001.

, Rôle antitumoral de l'orexine A et des ligands biaisés dans les cancers digestifs Impact sur le trafic intracellulaire d'OX1R

, Elles vont interagir avec deux sous-types de récepteurs couplés aux protéines G (RCPG), OX1R et OX2R. Une fois activés, ces deux récepteurs induisent la mobilisation du Ca 2+ intracellulaire via la protéine Gq. Au sein de l'équipe où j'ai effectué ma thèse, il a été clairement montré que le système orexines/OX1Ravait des propriétés anti-tumorales dans certains cancers, Par Stéphanie Dayot Les orexines sont des neuropeptides hypothalamiques qui possèdent deux isoformes, A et B (OxA et OxB, respectivement)

. Voisin, Enfin contre toute attente, j'ai montré que l'almorexant, un antagoniste de type DORA (Dual Orexin Receptor Antagonist) avait des propriétés antitumorales identique à l'OxA, l'agoniste naturel d'OX1R. Les résultats inattendus de l'almorexant vis-à-vis de ses propriétés antitumorales m'ont interpellée et ont ainsi déterminé l'axe de mon deuxième objectif. J'ai donc voulu savoir si cet effet était uniquement lié au PDAC ou s'il était plus largement dans d'autres cancers. Pour cela j'ai étudié l'effet de l'almorexant dans des lignées cellulaires dérivées d'adénocarcinomes coliques humains, les lignées HT-29 et LoVo. De plus, en collaboration avec le groupe de B. Robert (CRCM, INSERM U1194, Montpellier) nous avons développé, par une stratégie de « phage display », un anticorps agonis te dont j'ai montré qu'il mimait les effets de l'OxA sur les mêmes lignées cellulaires. Mon troisième objectif a été 'étudier les phénomènes d'internalisation d'OX1R sous l'action d'OxA et son devenir intracellulaire par des approches de microscopie confocale et d'analyse d'images. En effet, à ce jour peu, pour ne pas dire rien n'est connu. Plusieurs marqueurs de vésicules associées à l'internalisation des protéines ont été utilisés. Bien entendu, à la vue des effets inattendus, de l'almorexant, il me paraissait important d'étudier son impact sur ces phénomènes de régulation. Pour conclure, le récepteur OX1 est une cible potentielle pour le traitement thérapeutique des adénocarcinomes humains du côlon et du pancréas. De plus, la mise en évidence que l'almorexant et l'anticorps C2 miment les effets proapoptotiques et antitumoraux de l'OxA, représente une très bonne alternative au peptide naturel dont les inconvénients en terme de stabilité et d'administration peuvent représenter un frein dans son utilisation thérapeutique éventuelle. De plus, l'expression membranaire du récepteur OX1 au sein de la cellule et son devenir sont différentes en fonction du ligand. Ces données ont donc un intérêt d'un point de vue thérapeutique car l'almorexant comme l'anticorps C2 permettent au récepteur OX1 de rester exprimé à la surface cellulaire et ainsi d'être disponible pour son activité proapoptotique. Orexins are hypothalamic neuropeptides, which have two isoforms, A and B (OxA and OxB, respectively), Il a été montré que l'OxA mais aussi l'OxB induisait une apoptose mitochondriale via OX1R. Ces résultats signifient que le système orexines/OX1R représente une cible potentielle dans le traitement du cancer du côlon. Mon premier objectif de thèse a été d'étudier le rôle des orexines et en particulier de l'OxA sur l'adénocarcinome canalaire du pancréas (PDAC) chez l'Homme. Ces travaux m'ont permis de montrer qu'OX1R était exprimé dans 96% des PDAC testés. De plus, j'ai montré qu'OX1R était exprimé précocement dans les lésions précancéreuses (PanIN). J'ai démontré que la lignée cellulaire humaine dérivée d'un PDAC, la lignée AsPC1-exprimait OX1R et que l'OxA était capable d'induire une apoptose mitochondriale comparable à celle observée dans les cancers du côlon, 2011.