E. V. Bandera, S. H. Fay, E. Giovannucci, M. F. Leitzmann, R. Marklew et al., World Cancer Research Fund International Continuous Update Project P (2016) The use and interpretation of anthropometric measures in cancer epidemiology: A perspective from the world cancer research fund international continuous update project, Int J Cancer, vol.139, pp.2391-2398

S. P. Bauersfeld, C. S. Kessler, M. Wischnewsky, A. Jaensch, N. Steckhan et al., The effects of short-term fasting on quality of life and tolerance to chemotherapy in patients with breast and ovarian cancer: a randomized cross-over pilot study, BMC Cancer, vol.18, p.476, 2018.

P. Baumeister, S. Luo, W. C. Skarnes, G. Sui, E. Seto et al., Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers, Mol Cell Biol, vol.25, pp.4529-4569, 2005.

H. M. Beere, B. B. Wolf, K. Cain, D. D. Mosser, A. Mahboubi et al.,

A. Ben-zvi, E. A. Miller, and R. I. Morimoto, Collapse of proteostasis represents an early molecular event in Caenorhabditis elegans aging, Proc Natl Acad Sci U S A, vol.106, pp.14914-14923, 2009.

B. Sahra, I. Regazzetti, C. Robert, G. Laurent, K. et al., Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1, Cancer Res, vol.71, pp.4366-72, 2011.

J. L. Benci, B. Xu, Y. Qiu, T. J. Wu, H. Dada et al., Tumor Interferon Signaling Regulates a Multigenic Resistance Program to Immune Checkpoint Blockade, Cell, vol.167, p.12, 2016.

M. Beneteau, B. Zunino, M. A. Jacquin, O. Meynet, J. Chiche et al., Combination of glycolysis inhibition with chemotherapy results in an antitumor immune response, Proc Natl Acad Sci U S A, vol.109, pp.20071-20077, 2012.

R. Besch, H. Poeck, T. Hohenauer, D. Senft, G. Hacker et al., Proapoptotic signaling induced by RIG-I and MDA-5 results in type I interferon-independent apoptosis in human melanoma cells, J Clin Invest, vol.119, pp.2399-411, 2009.

D. E. Biancur, J. A. Paulo, B. Malachowska, Q. Del-rey, M. Sousa et al., Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism, Nat Commun, vol.8, p.15965, 2017.

N. Blazanin, J. Son, A. B. Craig-lucas, C. L. John, K. J. Breech et al., ER stress and distinct outputs of the IRE1alpha RNase control proliferation and senescence in response to oncogenic Ras, Proc Natl Acad Sci U S A, vol.114, pp.9900-9905, 2017.

A. Blees, K. Reichel, S. Trowitzsch, O. Fisette, C. Bock et al., Assembly of the MHC I peptide-loading complex determined by a conserved ionic lock-switch, Sci Rep, vol.5, p.17341, 2015.

W. J. Blot and R. E. Tarone, Doll and Peto's quantitative estimates of cancer risks: holding generally true for 35 years, J Natl Cancer Inst, vol.107, 2015.

M. J. Bonorden, O. P. Rogozina, C. M. Kluczny, M. E. Grossmann, P. L. Grambsch et al., Intermittent calorie restriction delays prostate tumor detection and increases survival time in TRAMP mice, Nutr Cancer, vol.61, p.125, 2009.

L. Booth, J. L. Roberts, A. Poklepovic, J. Kirkwood, and P. Dent, HDAC inhibitors enhance the immunotherapy response of melanoma cells, Oncotarget, vol.8, pp.83155-83170, 2017.

L. Bordone, D. Cohen, A. Robinson, M. C. Motta, E. Van-veen et al., SIRT1 transgenic mice show phenotypes resembling calorie restriction, Aging Cell, vol.6, pp.759-67, 2007.

M. Boutant, S. S. Kulkarni, M. Joffraud, F. Raymond, S. Metairon et al., SIRT1 Gain of Function Does Not Mimic or Enhance the Adaptations to Intermittent Fasting, Cell Rep, vol.14, pp.2068-2075, 2016.

J. W. Brewer, L. M. Hendershot, C. J. Sherr, and J. A. Diehl, Mammalian unfolded protein response inhibits cyclin D1 translation and cell-cycle progression, Proc Natl Acad Sci U S A, vol.96, pp.8505-8515, 1999.

R. G. Bristow and R. P. Hill, Hypoxia and metabolism. Hypoxia, DNA repair and genetic instability, Nat Rev Cancer, vol.8, pp.180-92, 2008.

J. M. Bruey, C. Ducasse, P. Bonniaud, L. Ravagnan, S. A. Susin et al., Hsp27 negatively regulates cell death by interacting with cytochrome c, Nat Cell Biol, vol.2, pp.645-52, 2000.

B. Bujisic, D. Gassart, A. Tallant, R. Demaria, O. Zaffalon et al., Impairment of both IRE1 expression and XBP1 activation is a hallmark of GCB DLBCL and contributes to tumor growth, Blood, vol.129, pp.2420-2428, 2017.

R. Buono and V. D. Longo, Starvation, Stress Resistance, and Cancer, Trends Endocrinol Metab, vol.29, pp.271-280, 2018.

B. C. Burnette, H. Liang, Y. Lee, L. Chlewicki, N. N. Khodarev et al., The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity, Cancer Res, vol.71, pp.2488-96, 2011.

T. C. Campbell, The Past, Present, and Future of Nutrition and Cancer: Part 1-Was A Nutritional Association Acknowledged a Century Ago, Nutr Cancer, vol.69, pp.811-817, 2017.

M. A. Cannarile, M. Weisser, W. Jacob, A. M. Jegg, C. H. Ries et al., Colony-stimulating factor 1 receptor (CSF1R) inhibitors in cancer therapy, J Immunother Cancer, vol.5, p.53, 2017.

H. Cao, G. Wang, L. Meng, H. Shen, Z. Feng et al., Association between circulating levels of IGF-1 and IGFBP-3 and lung cancer risk: a meta-analysis, PLoS One, vol.7, p.49884, 2012.

M. A. Carpio, M. Michaud, W. Zhou, J. K. Fisher, L. D. Walensky et al., BCL-2 family member BOK promotes apoptosis in response to endoplasmic reticulum stress, Proc Natl Acad Sci U S A, vol.112, pp.7201-7207, 2015.

J. H. Cha, W. H. Yang, W. Xia, Y. Wei, L. C. Chan et al., Metformin Promotes Antitumor Immunity via Endoplasmic-Reticulum-Associated Degradation of PD-L1, Mol Cell, vol.71, p.7, 2018.

A. Chalkiadaki and L. Guarente, The multifaceted functions of sirtuins in cancer, Nat Rev Cancer, vol.15, pp.608-632, 2015.

B. Chaneton and E. Gottlieb, PGAMgnam style: a glycolytic switch controls biosynthesis, Cancer Cell, vol.22, pp.565-571, 2012.

C. L. Chang, Y. T. Hsu, C. C. Wu, Y. C. Yang, C. Wang et al., Immune mechanism of the antitumor effects generated by bortezomib, J Immunol, vol.189, pp.3209-3229, 2012.

C. Chaveroux, C. Sarcinelli, V. Barbet, S. Belfeki, A. Barthelaix et al., Nutrient shortage triggers the hexosamine biosynthetic pathway via the GCN2-ATF4 signalling pathway, Sci Rep, vol.6, p.27278, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01595359

X. Chen, D. Iliopoulos, Q. Zhang, Q. Tang, M. B. Greenblatt et al., XBP1 promotes triplenegative breast cancer by controlling the HIF1alpha pathway, Nature, vol.508, pp.103-107, 2014.

J. H. Cheong, E. S. Park, J. Liang, J. B. Dennison, D. Tsavachidou et al., Dual inhibition of tumor energy pathway by 2-deoxyglucose and metformin is effective against a broad spectrum of preclinical cancer models, Mol Cancer Ther, vol.10, pp.2350-62, 2011.

J. A. Cho, A. H. Lee, B. Platzer, B. C. Cross, B. M. Gardner et al., The unfolded protein response element IRE1alpha senses bacterial proteins invading the ER to activate RIG-I and innate immune signaling, Cell Host Microbe, vol.13, pp.558-69, 2013.

K. M. Choi, Y. Y. Kwon, and C. K. Lee, Characterization of global gene expression during assurance of lifespan extension by caloric restriction in budding yeast, Exp Gerontol, vol.48, pp.1455-68, 2013.

H. Y. Cohen, C. Miller, K. J. Bitterman, N. R. Wall, B. Hekking et al., Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase, Science, vol.305, pp.390-392, 2004.

G. A. Colditz, T. A. Sellers, and E. Trapido, Epidemiology -identifying the causes and preventability of cancer?, Nat Rev Cancer, vol.6, pp.75-83, 2006.

G. A. Colditz, K. Y. Wolin, and S. Gehlert, Applying what we know to accelerate cancer prevention, Sci Transl Med, vol.4, pp.127-131, 2012.

, Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128.9 million children, adolescents, and adults, Collaboration, vol.390, pp.2627-2642, 2017.

F. Concha-benavente, R. M. Srivastava, S. Trivedi, Y. Lei, U. Chandran et al., Identification of the Cell-Intrinsic and -Extrinsic Pathways Downstream of EGFR and IFNgamma That Induce PD-L1 Expression in Head and Neck Cancer, Cancer Res, vol.76, pp.1031-1074, 2016.

T. Condamine, G. A. Dominguez, J. I. Youn, A. V. Kossenkov, S. Mony et al., Lectin-type oxidized LDL receptor, 2016.

J. Condeelis and J. W. Pollard, Macrophages: obligate partners for tumor cell migration, invasion, and metastasis, Cell, vol.124, pp.263-269, 2006.

K. L. Cook, D. R. Soto-pantoja, P. A. Clarke, M. I. Cruz, A. Zwart et al., , 2016.

, Endoplasmic Reticulum Stress Protein GRP78 Modulates Lipid Metabolism to Control Drug Sensitivity and Antitumor Immunity in Breast Cancer, Cancer Res, vol.76, pp.5657-5670

D. V. Correia, A. Lopes, and B. Silva-santos, Tumor cell recognition by gammadelta T lymphocytes: T-cell receptor vs. NK-cell receptors, Oncoimmunology, vol.2, p.22892, 2013.

T. G. Cotter, Apoptosis and cancer: the genesis of a research field, Nat Rev Cancer, vol.9, pp.501-508, 2009.

J. Grootjans, A. Kaser, R. J. Kaufman, and R. S. Blumberg, The unfolded protein response in immunity and inflammation, Nat Rev Immunol, vol.16, pp.469-84, 2016.

M. I. Gross, S. D. Demo, J. B. Dennison, L. Chen, T. Chernov-rogan et al., Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer, Mol Cancer Ther, vol.13, pp.890-901, 2014.

L. Guarente, Calorie restriction and sirtuins revisited, Genes Dev, vol.27, pp.2072-85, 2013.

J. Guevara-aguirre, P. Balasubramanian, M. Guevara-aguirre, M. Wei, F. Madia et al., Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans, Sci Transl Med, vol.3, pp.70-83, 2011.

D. M. Gwinn, D. B. Shackelford, D. F. Egan, M. M. Mihaylova, A. Mery et al., AMPK phosphorylation of raptor mediates a metabolic checkpoint, Mol Cell, vol.30, pp.214-240, 2008.

A. Halama, M. Kulinski, S. S. Dib, S. B. Zaghlool, K. S. Siveen et al., Accelerated lipid catabolism and autophagy are cancer survival mechanisms under inhibited glutaminolysis, Cancer Lett, vol.430, pp.133-147, 2018.

K. Halbleib, K. Pesek, R. Covino, H. F. Hofbauer, D. Wunnicke et al., Activation of the Unfolded Protein Response by Lipid Bilayer Stress, Mol Cell, vol.67, p.8, 2017.

D. T. Hall, T. Griss, J. F. Ma, B. J. Sanchez, J. Sadek et al., The AMPK agonist 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), but not metformin, prevents inflammation-associated cachectic muscle wasting, EMBO Mol Med, vol.10, 2018.

J. M. Hall, M. K. Lee, B. Newman, J. E. Morrow, L. A. Anderson et al., Linkage of early-onset familial breast cancer to chromosome 17q21, Science, vol.250, pp.1684-1693, 1990.

D. Han, A. G. Lerner, L. Vande-walle, J. P. Upton, W. Xu et al., IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates, Cell, vol.138, pp.562-75, 2009.

D. Hanahan and R. A. Weinberg, The hallmarks of cancer. Cell, vol.100, pp.57-70, 2000.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-74, 2011.

C. Handschin, Caloric restriction and exercise "mimetics'': Ready for prime time?, Pharmacol Res, vol.103, pp.158-66, 2016.

D. G. Hardie, AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy, Nat Rev Mol Cell Biol, vol.8, pp.774-85, 2007.

L. S. Hart, J. T. Cunningham, T. Datta, S. Dey, F. Tameire et al., ER stress-mediated autophagy promotes Myc-dependent transformation and tumor growth, J Clin Invest, vol.122, pp.4621-4655, 2012.

A. E. Harvey, L. M. Lashinger, G. Otto, N. P. Nunez, and S. D. Hursting, Decreased systemic IGF-1 in response to calorie restriction modulates murine tumor cell growth, nuclear factor-kappaB activation, and inflammation-related gene expression, Mol Carcinog, vol.52, pp.997-1006, 2013.

K. R. Harwood and J. A. Hanover, Nutrient-driven O-GlcNAc cycling -think globally but act locally, J Cell Sci, vol.127, p.132, 2014.

J. Heitman, N. R. Movva, and M. N. Hall, Targets for cell cycle arrest by the immunosuppressant rapamycin in yeast, Science, vol.253, pp.905-914, 1991.

S. Henis-korenblit, P. Zhang, M. Hansen, M. Mccormick, S. J. Lee et al., Insulin/IGF-1 signaling mutants reprogram ER stress response regulators to promote longevity, Proc Natl Acad Sci U S A, vol.107, pp.9730-9735, 2010.

D. Herranz, G. Iglesias, M. Munoz-martin, and M. Serrano, Limited role of Sirt1 in cancer protection by dietary restriction, Cell Cycle, vol.10, pp.2215-2222, 2011.

D. Herranz, M. Munoz-martin, M. Canamero, F. Mulero, B. Martinez-pastor et al., Sirt1 improves healthy ageing and protects from metabolic syndrome-associated cancer, Nat Commun, vol.1, p.3, 2010.

C. Hetz, E. Chevet, and H. P. Harding, Targeting the unfolded protein response in disease, Nat Rev Drug Discov, vol.12, pp.703-722, 2013.

C. Hetz, E. Chevet, and S. A. Oakes, Proteostasis control by the unfolded protein response, Nat Cell Biol, vol.17, pp.829-867, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01175531

H. Horn, B. Bohme, L. Dietrich, and M. Koch, Endocannabinoids in Body Weight Control. Pharmaceuticals, p.11, 2018.

B. L. Horton and T. F. Gajewski, Back from the dead: TIL apoptosis in cancer immune evasion, Br J Cancer, vol.118, pp.309-311, 2018.

B. L. Horton, J. B. Williams, A. Cabanov, S. Spranger, and T. F. Gajewski, Intratumoral CD8(+) T-cell Apoptosis Is a Major Component of T-cell Dysfunction and Impedes Antitumor Immunity, Cancer Immunol Res, vol.6, pp.14-24, 2018.

A. M. Hosios and M. G. Vander-heiden, Acetate metabolism in cancer cells, Cancer Metab, vol.2, p.27, 2014.

J. Hou, T. F. Greten, and Q. Xia, Immunosuppressive cell death in cancer, Nat Rev Immunol, vol.17, p.401, 2017.

J. Hou, Y. Zhou, Y. Zheng, J. Fan, W. Zhou et al., Hepatic RIG-I predicts survival and interferon-alpha therapeutic response in hepatocellular carcinoma, Cancer Cell, vol.25, pp.49-63, 2014.

J. J. Howell, S. J. Ricoult, I. Ben-sahra, and B. D. Manning, A growing role for mTOR in promoting anabolic metabolism, Biochem Soc Trans, vol.41, pp.906-918, 2013.

G. Hoxhaj, J. Hughes-hallett, R. C. Timson, E. Ilagan, M. Yuan et al., , 2017.

, Signaling Network Senses Changes in Cellular Purine Nucleotide Levels, Cell Rep, vol.21, pp.1331-1346

F. Hu, X. Yu, H. Wang, D. Zuo, C. Guo et al., ER stress and its regulator X-box-binding protein-1 enhance polyIC-induced innate immune response in dendritic cells, Eur J Immunol, vol.41, pp.1086-97, 2011.

R. Hu, A. Warri, L. Jin, A. Zwart, R. B. Riggins et al., NF-kappaB signaling is required for XBP1 (unspliced and spliced)-mediated effects on antiestrogen responsiveness and cell fate decisions in breast cancer, Mol Cell Biol, vol.35, pp.379-90, 2015.

J. Huang and B. D. Manning, A complex interplay between Akt, TSC2 and the two mTOR complexes, Biochem Soc Trans, vol.37, pp.217-239, 2009.

K. Y. Hur, J. S. So, V. Ruda, M. Frank-kamenetsky, K. Fitzgerald et al., IRE1alpha activation protects mice against acetaminophen-induced hepatotoxicity, J Exp Med, vol.209, pp.307-325, 2012.

F. Imamura, R. Micha, S. Khatibzadeh, S. Fahimi, P. Shi et al., Global Burden of Diseases N, Chronic Diseases Expert G (2015) Dietary quality among men and women in 187 countries in 1990 and 2010: a systematic assessment, Lancet Glob Health, vol.3, pp.132-174

D. K. Ingram and G. S. Roth, Glycolytic inhibition as a strategy for developing calorie restriction mimetics, Exp Gerontol, vol.46, pp.148-54, 2011.

D. K. Ingram and G. S. Roth, Calorie restriction mimetics: can you have your cake and eat it, too?, Ageing Res Rev, vol.20, pp.46-62, 2015.

S. M. Jeon, N. S. Chandel, and N. Hay, AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress, Nature, vol.485, pp.661-666, 2012.

L. Jin, G. N. Alesi, and S. Kang, Glutaminolysis as a target for cancer therapy, Oncogene, vol.35, pp.3619-3644, 2016.

S. C. Johnson, P. S. Rabinovitch, and M. Kaeberlein, mTOR is a key modulator of ageing and age-related disease, Nature, vol.493, pp.338-383, 2013.

R. B. Jones, R. O'connor, S. Mueller, M. Foley, G. L. Szeto et al., Histone deacetylase inhibitors impair the elimination of HIV-infected cells by cytotoxic T-lymphocytes, PLoS Pathog, vol.10, p.1004287, 2014.

R. G. Jones, T. Bui, C. White, M. Madesh, C. M. Krawczyk et al., The proapoptotic factors Bax and Bak regulate T Cell proliferation through control of endoplasmic reticulum Ca(2+) homeostasis, Immunity, vol.27, pp.268-80, 2007.

J. A. Joyce and D. T. Fearon, T cell exclusion, immune privilege, and the tumor microenvironment, Science, vol.348, pp.74-80, 2015.

V. R. Juneja, K. A. Mcguire, R. T. Manguso, M. W. Lafleur, N. Collins et al., PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity, J Exp Med, vol.214, pp.895-904, 2017.

J. Li, C. X. Zhang, Y. M. Liu, K. L. Chen, and G. Chen, A comparative study of anti-aging properties and mechanism: resveratrol and caloric restriction, Oncotarget, vol.8, pp.65717-65729, 2017.

K. Li, S. Qu, X. Chen, Q. Wu, and M. Shi, Promising Targets for Cancer Immunotherapy: TLRs, RLRs, and STINGMediated Innate Immune Pathways, Int J Mol Sci, vol.18, 2017.

X. X. Li, H. S. Zhang, Y. M. Xu, R. J. Zhang, Y. Chen et al., Knockdown of IRE1alpha inhibits colonic tumorigenesis through decreasing beta-catenin and IRE1alpha targeting suppresses colon cancer cells, Oncogene Liberti MV, vol.41, pp.211-218, 2016.

P. Lichtenstein, N. V. Holm, P. K. Verkasalo, A. Iliadou, J. Kaprio et al., Environmental and heritable factors in the causation of cancer--analyses of cohorts of twins from, N Engl J Med, vol.343, pp.78-85, 2000.

S. Lim, J. B. Phillips, M. Da-silva, L. Zhou, M. Fodstad et al., Interplay between Immune Checkpoint Proteins and Cellular Metabolism, Cancer Res, vol.77, pp.1245-1249, 2017.

C. Lippens, F. V. Duraes, J. Dubrot, D. Brighouse, M. Lacroix et al., IDO-orchestrated crosstalk between pDCs and Tregs inhibits autoimmunity, J Autoimmun, vol.75, pp.39-49, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01438478

Y. Liu, Y. Komohara, N. Domenick, M. Ohno, M. Ikeura et al., Expression of antigen processing and presenting molecules in brain metastasis of breast cancer, Cancer Immunol Immunother, vol.61, pp.789-801, 2012.

F. Llambi, Y. M. Wang, B. Victor, M. Yang, D. M. Schneider et al., BOK Is a Non-canonical BCL-2 Family Effector of Apoptosis Regulated by ER-Associated Degradation, Cell, vol.165, pp.421-454, 2016.

L. Presti, E. Pizzolato, G. Corsale, A. M. Caccamo, N. Sireci et al., ) gammadelta T Cells and Tumor Microenvironment: From Immunosurveillance to Tumor Evasion, Front Immunol, vol.9, p.1395, 2018.

S. E. Logue, E. P. Mcgrath, P. Cleary, S. Greene, K. Mnich et al., Inhibition of IRE1 RNase activity modulates the tumor cell secretome and enhances response to chemotherapy, Nat Commun, vol.9, p.3267, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01879955

E. H. Ma, G. Bantug, T. Griss, S. Condotta, R. M. Johnson et al., Serine Is an Essential Metabolite for Effector T Cell Expansion. Cell Metab, vol.25, pp.345-357, 2017.

Y. Ma and L. M. Hendershot, The role of the unfolded protein response in tumour development: friend or foe?, Nat Rev Cancer, vol.4, pp.966-77, 2004.

O. Maddocks, D. Athineos, E. C. Cheung, P. Lee, T. Zhang et al., Modulating the therapeutic response of tumours to dietary serine and glycine starvation, Nature, vol.544, pp.372-376, 2017.

F. Madeo, F. Pietrocola, T. Eisenberg, and G. Kroemer, Caloric restriction mimetics: towards a molecular definition, Nat Rev Drug Discov, vol.13, p.137, 2014.

Y. D. Mahnke, E. Devevre, P. Baumgaertner, M. Matter, N. Rufer et al., Human melanomaspecific CD8(+) T-cells from metastases are capable of antigen-specific degranulation and cytolysis directly ex vivo, Oncoimmunology, vol.1, pp.467-530, 2012.

V. Mai, L. H. Colbert, D. Berrigan, S. N. Perkins, R. Pfeiffer et al., Calorie restriction and diet composition modulate spontaneous intestinal tumorigenesis in Apc(Min) mice through different mechanisms, Cancer Res, vol.63, pp.1752-1757, 2003.

A. S. Malamas, S. R. Gameiro, K. M. Knudson, and J. W. Hodge, Sublethal exposure to alpha radiation (223Ra dichloride) enhances various carcinomas' sensitivity to lysis by antigen-specific cytotoxic T lymphocytes through calreticulinmediated immunogenic modulation, Oncotarget, vol.7, pp.86937-86947, 2016.

B. D. Manning, Game of TOR -The Target of Rapamycin Rules Four Kingdoms, N Engl J Med, vol.377, pp.1297-1306, 2017.

A. Mantovani and A. Sica, Macrophages, innate immunity and cancer: balance, tolerance, and diversity, Curr Opin Immunol, vol.22, pp.231-238, 2010.

M. G. Marcu, Y. J. Jung, S. Lee, E. J. Chung, M. J. Lee et al., Curcumin is an inhibitor of p300 histone acetylatransferase, Med Chem, vol.2, pp.169-74, 2006.

A. Marcus, B. G. Gowen, T. W. Thompson, A. Iannello, M. Ardolino et al., Recognition of tumors by the innate immune system and natural killer cells, Adv Immunol, vol.122, pp.91-128, 2014.

G. Marino, F. Pietrocola, F. Madeo, and G. Kroemer, Caloric restriction mimetics: natural/physiological pharmacological autophagy inducers, Autophagy, vol.10, pp.1879-82, 2014.

K. A. Mark, K. J. Dumas, D. Bhaumik, B. Schilling, S. Davis et al., Vitamin D Promotes Protein Homeostasis and Longevity via the Stress Response Pathway Genes skn-1, ire-1, and xbp-1, Cell Rep, vol.17, pp.1227-1237, 2016.

A. Martin-montalvo, E. M. Mercken, S. J. Mitchell, H. H. Palacios, P. L. Mote et al., Metformin improves healthspan and lifespan in mice, Nat Commun, vol.4, p.2192, 2013.

F. Martinon, X. Chen, A. H. Lee, and L. H. Glimcher, TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages, Nat Immunol, vol.11, pp.411-419, 2010.

I. Martins, O. Kepp, F. Schlemmer, S. Adjemian, M. Tailler et al., Restoration of the immunogenicity of cisplatin-induced cancer cell death by endoplasmic reticulum stress, Oncogene, vol.30, pp.1147-58, 2011.

S. R. Mattarollo, S. Loi, H. Duret, Y. Ma, L. Zitvogel et al., Pivotal role of innate and adaptive immunity in anthracycline chemotherapy of established tumors, Cancer Res, vol.71, pp.4809-4829, 2011.

M. Maurel, E. Chevet, J. Tavernier, and S. Gerlo, Getting RIDD of RNA: IRE1 in cell fate regulation, Trends Biochem Sci, vol.39, pp.245-54, 2014.

M. F. Mccarty and K. I. Block, Preadministration of high-dose salicylates, suppressors of NF-kappaB activation, may increase the chemosensitivity of many cancers: an example of proapoptotic signal modulation therapy, Integr Cancer Ther, vol.5, pp.252-68, 2006.

C. M. Mccay, M. F. Crowell, and L. A. Maynard, The effect of retarded growth upon the length of life span and upon the ultimate body size, Nutrition, vol.5, pp.155-71, 1935.

J. J. Mcneil, M. R. Nelson, R. L. Woods, J. E. Lockery, R. Wolfe et al., Effect of Aspirin on All-Cause Mortality in the Healthy Elderly, Cardiac glycosides exert anticancer effects by inducing immunogenic cell death. Sci Transl Med, vol.4, pp.143-99, 2012.

O. Meynet, M. Beneteau, M. A. Jacquin, L. A. Pradelli, A. Cornille et al., Glycolysis inhibition targets Mcl-1 to restore sensitivity of lymphoma cells to ABT-737-induced apoptosis, Leukemia, vol.26, pp.1145-1152, 2012.

O. Meynet and J. E. Ricci, Caloric restriction and cancer: molecular mechanisms and clinical implications, Trends Mol Med, vol.20, pp.419-446, 2014.

Y. Miki, J. Swensen, D. Shattuck-eidens, P. A. Futreal, K. Harshman et al., A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science, vol.266, pp.66-71, 1994.

R. K. Minor, D. L. Smith, J. Sossong, A. M. Kaushik, S. Poosala et al., Chronic ingestion of 2-deoxy-D-glucose induces cardiac vacuolization and increases mortality in rats, Toxicol Appl Pharmacol, vol.243, pp.332-341, 2010.

S. J. Mitchell, M. Bernier, J. A. Mattison, M. A. Aon, T. A. Kaiser et al., Daily Fasting Improves Health and Survival in Male Mice Independent of Diet Composition and Calories, Cell Metab, 2018.

D. Mittal, M. M. Gubin, R. D. Schreiber, and M. J. Smyth, New insights into cancer immunoediting and its three component phases--elimination, equilibrium and escape, Curr Opin Immunol, vol.27, pp.16-25, 2014.

J. Monge, M. Kricun, J. Radovcic, D. Radovcic, A. Mann et al., Fibrous dysplasia in a 120,000+ year old Neandertal from Krapina, Croatia. PLoS One, vol.8, p.64539, 2013.

N. R. Monu and A. B. Frey, Myeloid-derived suppressor cells and anti-tumor T cells: a complex relationship, Immunol Invest, vol.41, pp.595-613, 2012.

L. V. Moore, K. W. Dodd, F. E. Thompson, K. A. Grimm, S. A. Kim et al., Using Behavioral Risk Factor Surveillance System Data to Estimate the Percentage of the Population Meeting US Department of Agriculture Food Patterns Fruit and Vegetable Intake Recommendations, Am J Epidemiol, vol.181, pp.979-88, 2015.

J. D. Moyer, J. T. Oliver, and R. E. Handschumacher, Salvage of circulating pyrimidine nucleosides in the rat, Cancer Res, vol.41, pp.3010-3017, 1981.

H. Mujcic, A. Nagelkerke, K. M. Rouschop, S. Chung, N. Chaudary et al., Hypoxic activation of the PERK/eIF2alpha arm of the unfolded protein response promotes metastasis through induction of LAMP3, Clin Cancer Res, vol.19, pp.6126-6163, 2013.

J. P. Munoz, S. Ivanova, J. Sanchez-wandelmer, P. Martinez-cristobal, E. Noguera et al., Mfn2 modulates the UPR and mitochondrial function via repression of PERK, EMBO J, vol.32, pp.2348-61, 2013.

B. Y. Nabet, Y. Qiu, J. E. Shabason, T. J. Wu, T. Yoon et al., Exosome RNA Unshielding Couples Stromal Activation to Pattern Recognition Receptor Signaling in Cancer. Cell, vol.170, pp.352-366, 2017.

N. Naidoo, M. Ferber, M. Master, Y. Zhu, and A. I. Pack, Aging impairs the unfolded protein response to sleep deprivation and leads to proapoptotic signaling, J Neurosci, vol.28, pp.6539-6587, 2008.

Y. Naito, K. Saito, K. Shiiba, A. Ohuchi, K. Saigenji et al., CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer, Cancer Res, vol.58, pp.3491-3495, 1998.

H. Nakagawa, A. Umemura, K. Taniguchi, J. Font-burgada, D. Dhar et al., ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development, Cancer Cell, vol.26, pp.331-343, 2014.

A. Nakamura, T. Nambu, S. Ebara, Y. Hasegawa, K. Toyoshima et al., Inhibition of GCN2 sensitizes ASNS-low cancer cells to asparaginase by disrupting the amino acid response, Proc Natl Acad Sci U S A, vol.115, pp.7776-7785, 2018.

T. Namba, K. Chu, R. Kodama, S. Byun, K. W. Yoon et al., Loss of p53 enhances the function of the endoplasmic reticulum through activation of the IRE1alpha/XBP1 pathway, Oncotarget, vol.6, pp.19990-20001, 2015.

L. Niederreiter, T. M. Fritz, T. E. Adolph, A. M. Krismer, F. A. Offner et al., ER stress transcription factor Xbp1 suppresses intestinal tumorigenesis and directs intestinal stem cells, J Exp Med, vol.210, pp.2041-56, 2013.

S. Nikolai, K. Pallauf, P. Huebbe, and G. Rimbach, Energy restriction and potential energy restriction mimetics, Nutr Res Rev, vol.28, pp.100-120, 2015.

Z. Niu, M. Wang, L. Zhou, L. Yao, Q. Liao et al., Elevated GRP78 expression is associated with poor prognosis in patients with pancreatic, cancer. Sci Rep, vol.5, p.16067, 2015.

C. Norman, K. A. Howell, A. H. Millar, J. M. Whelan, and D. A. Day, Salicylic acid is an uncoupler and inhibitor of mitochondrial electron transport, Plant Physiol, vol.134, pp.492-501, 2004.

R. Nowarski, N. Gagliani, S. Huber, and R. A. Flavell, Innate immune cells in inflammation and cancer, Cancer Immunol Res, vol.1, pp.77-84, 2013.

R. Noy and J. W. Pollard, Tumor-associated macrophages: from mechanisms to therapy, Immunity, vol.41, pp.49-61, 2014.

T. O'sullivan, R. Saddawi-konefka, W. Vermi, C. M. Koebel, C. Arthur et al., Cancer immunoediting by the innate immune system in the absence of adaptive immunity, J Exp Med, vol.209, pp.1869-82, 2012.

J. Obacz, A. T. , L. Reste, P. J. Urra, H. Quillien et al., Endoplasmic reticulum proteostasis in glioblastoma-From molecular mechanisms to therapeutic perspectives, Sci Signal, vol.10, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01502552

M. Obeid, A. Tesniere, F. Ghiringhelli, G. M. Fimia, L. Apetoh et al., Calreticulin exposure dictates the immunogenicity of cancer cell death, Nat Med, vol.13, pp.54-61, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00451702

A. R. Orillion, N. P. Damayanti, L. Shen, R. Adelaiye-ogala, H. C. Affronti et al., Dietary protein restriction reprograms tumor associated macrophages and enhances immunotherapy, Clin Cancer Res Pages F, vol.353, pp.2654-66, 2005.

K. Pallauf and G. Rimbach, Autophagy, polyphenols and healthy ageing, Ageing Res Rev, vol.12, pp.237-52, 2013.

R. Pallavi, M. Giorgio, and P. G. Pelicci, Insights into the beneficial effect of caloric/ dietary restriction for a healthy and prolonged life, Front Physiol, vol.3, p.318, 2012.

T. Panaretakis, O. Kepp, U. Brockmeier, A. Tesniere, A. C. Bjorklund et al., Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death, EMBO J, vol.28, pp.578-90, 2009.

N. E. Papaioannou, O. V. Beniata, P. Vitsos, O. Tsitsilonis, and P. Samara, Harnessing the immune system to improve cancer therapy, Ann Transl Med, vol.4, p.261, 2016.

I. A. Park, S. H. Heo, I. H. Song, Y. A. Kim, H. S. Park et al., Endoplasmic reticulum stress induces secretion of high-mobility group proteins and is associated with tumor-infiltrating lymphocytes in triple-negative breast cancer, Oncotarget, vol.7, pp.59957-59964, 2016.

M. J. Paul-clark, T. Van-cao, N. Moradi-bidhendi, D. Cooper, and D. W. Gilroy, 15-epi-lipoxin A4-mediated induction of nitric oxide explains how aspirin inhibits acute inflammation, J Exp Med, vol.200, pp.69-78, 2004.

N. N. Pavlova and C. B. Thompson, The Emerging Hallmarks of Cancer Metabolism, Cell Metab, vol.23, pp.27-47, 2016.

E. R. Pereira, K. Frudd, W. Awad, and L. M. Hendershot, Endoplasmic reticulum (ER) stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 (HIF-1) transcriptional activity on targets like vascular endothelial growth factor (VEGF), J Biol Chem, vol.289, pp.3352-64, 2014.

J. Perez-escuredo, R. K. Dadhich, S. Dhup, A. Cacace, V. F. Van-hee et al., Lactate promotes glutamine uptake and metabolism in oxidative cancer cells, Cell Cycle, vol.15, pp.72-83, 2016.

I. Pernicova and M. Korbonits, Metformin--mode of action and clinical implications for diabetes and cancer, Nat Rev Endocrinol, vol.10, pp.143-56, 2014.

L. W. Peterson and D. Artis, Intestinal epithelial cells: regulators of barrier function and immune homeostasis, Nat Rev Immunol, vol.14, pp.141-53, 2014.

C. Pfirschke, C. Engblom, S. Rickelt, V. Cortez-retamozo, C. Garris et al., Immunogenic Chemotherapy Sensitizes Tumors to Checkpoint Blockade Therapy. Immunity, vol.44, pp.343-54, 2016.

K. N. Phoenix, F. Vumbaca, M. M. Fox, R. Evans, and K. P. Claffey, Dietary energy availability affects primary and metastatic breast cancer and metformin efficacy, Breast Cancer Res Treat, vol.123, pp.333-377, 2010.

F. Pietrocola, F. Castoldi, M. C. Maiuri, and G. Kroemer, Aspirin-another caloric-restriction mimetic, Autophagy, pp.1-2, 2018.

F. Pietrocola, F. Castoldi, M. Markaki, S. Lachkar, G. Chen et al., Aspirin Recapitulates Features of Caloric Restriction, Cell Rep, vol.22, pp.2395-2407, 2018.

F. Pietrocola, J. Pol, E. Vacchelli, S. Rao, D. P. Enot et al., Caloric Restriction Mimetics Enhance Anticancer Immunosurveillance, Cancer Cell, vol.30, pp.147-160, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01431196

C. Piperi, V. Samaras, G. Levidou, N. Kavantzas, E. Boviatsis et al., Prognostic significance of IL-8-STAT-3 pathway in astrocytomas: correlation with IL-6, VEGF and microvessel morphometry, Cytokine, vol.55, pp.387-95, 2011.

J. M. Ploeger, J. C. Manivel, L. N. Boatner, and D. G. Mashek, Caloric Restriction Prevents Carcinogen-Initiated Liver Tumorigenesis in Mice, Cancer Prev Res (Phila), vol.10, pp.660-670, 2017.

H. Poeck, R. Besch, C. Maihoefer, M. Renn, D. Tormo et al., ) 5'-Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma, Nat Med, vol.14, pp.1256-63, 2008.

H. Puthalakath, L. A. Reilly, P. Gunn, L. Lee, P. N. Kelly et al., ER stress triggers apoptosis by activating BH3-only protein Bim, Cell, vol.129, pp.1337-1386, 2007.

D. Pytel, I. Majsterek, and J. A. Diehl, Tumor progression and the different faces of the PERK kinase, Oncogene, vol.35, pp.1207-1222, 2016.

M. Raghavan, S. J. Wijeyesakere, L. R. Peters, D. Cid, and N. , Calreticulin in the immune system: ins and outs, vol.34, pp.13-21, 2013.

G. Ramadori, G. Konstantinidou, N. Venkateswaran, T. Biscotti, L. Morlock et al., Diet-induced unresolved ER stress hinders KRAS-driven lung tumorigenesis, Cell Metab, vol.21, pp.117-142, 2015.

R. Ramakrishnan, V. A. Tyurin, F. Veglia, T. Condamine, A. Amoscato et al., Oxidized lipids block antigen cross-presentation by dendritic cells in cancer, J Immunol, vol.192, pp.2920-2951, 2014.

A. C. Ranganathan, L. Zhang, A. P. Adam, and J. A. Aguirre-ghiso, Functional coupling of p38-induced up-regulation of BiP and activation of RNA-dependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells, Cancer Res, vol.66, pp.1702-1713, 2006.

G. Rena, D. G. Hardie, and E. R. Pearson, The mechanisms of action of metformin, Diabetologia, vol.60, pp.1577-1585, 2017.

R. Reyes, N. A. Wani, K. Ghoshal, S. T. Jacob, and T. Motiwala, Sorafenib and 2-Deoxyglucose Synergistically Inhibit Proliferation of Both Sorafenib-Sensitive and -Resistant HCC Cells by Inhibiting ATP Production, Gene Expr, vol.17, pp.129-140, 2017.

M. Richter, N. Vidovic, B. Honrath, P. Mahavadi, R. Dodel et al., Activation of SK2 channels preserves ER Ca(2)(+) homeostasis and protects against ER stress-induced cell death, Cell Death Differ, vol.23, pp.814-841, 2016.

C. H. Ries, M. A. Cannarile, S. Hoves, J. Benz, K. Wartha et al., Targeting tumor-associated macrophages with anti-CSF-1R antibody reveals a strategy for cancer therapy, Cancer Cell, vol.25, pp.846-59, 2014.

K. Robien, W. Demark-wahnefried, and C. L. Rock, Evidence-based nutrition guidelines for cancer survivors: current guidelines, knowledge gaps, and future research directions, J Am Diet Assoc, vol.111, pp.368-75, 2011.

P. C. Rodriguez, D. G. Quiceno, J. Zabaleta, B. Ortiz, A. H. Zea et al., Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses, Cancer Res, vol.64, pp.5839-5888, 2004.

B. D. Roebuck, K. J. Baumgartner, and D. L. Macmillan, Caloric restriction and intervention in pancreatic carcinogenesis in the rat, Cancer Res, vol.53, pp.46-52, 1993.

B. M. Rothschild, B. J. Witzke, and I. Hershkovitz, Metastatic cancer in the Jurassic, Lancet, vol.354, p.398, 1999.

C. Rubio-patino, J. P. Bossowski, E. Chevet, and J. E. Ricci, Reshaping the Immune Tumor Microenvironment Through IRE1 Signaling, Trends Mol Med, vol.24, pp.607-614, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01835398

C. Rubio-patino, J. P. Bossowski, D. Donatis, G. M. Mondragon, L. Villa et al., Low-Protein Diet Induces IRE1alpha-Dependent Anticancer Immunosurveillance, Cell Metab, vol.27, p.7, 2018.

C. Rubio-patino, J. P. Bossowski, E. Villa, L. Mondragon, B. Zunino et al., Low carbohydrate diet prevents Mcl-1-mediated resistance to BH3-mimetics, Oncotarget, vol.7, pp.73270-73279, 2016.

G. N. Ruegsegger, J. A. Sevage, T. E. Childs, K. B. Grigsby, and F. W. Booth, 5-Aminoimidazole-4-carboxamide ribonucleotide prevents fat gain following the cessation of voluntary physical activity, Exp Physiol, vol.102, pp.1474-1485, 2017.

B. A. Ruggeri, D. M. Klurfeld, D. Kritchevsky, and R. W. Furlanetto, Caloric restriction and 7,12-dimethylbenz(a)anthracene-induced mammary tumor growth in rats: alterations in circulating insulin, insulin-like growth factors I and II, and epidermal growth factor, Cancer Res, vol.49, pp.4130-4134, 1989.

D. M. Sabatini, Twenty-five years of mTOR: Uncovering the link from nutrients to growth, Proc Natl Acad Sci U S A, vol.114, pp.11818-11825, 2017.

N. Sahu, D. Cruz, D. Gao, M. Sandoval, W. Haverty et al., Proline Starvation Induces Unresolved ER Stress and Hinders mTORC1-Dependent Tumorigenesis, Cell Metab, vol.24, pp.753-761, 2016.

P. Saintigny, E. Massarelli, S. Lin, Y. H. Ahn, Y. Chen et al., CXCR2 expression in tumor cells is a poor prognostic factor and promotes invasion and metastasis in lung adenocarcinoma, Cancer Res, vol.73, pp.571-82, 2013.

T. Sakatani, K. Maemura, N. Hiyama, Y. Amano, K. Watanabe et al., High expression of IRE1 in lung adenocarcinoma is associated with a lower rate of recurrence, Jpn J Clin Oncol, vol.47, pp.543-550, 2017.

Y. Sancak, L. Bar-peled, R. Zoncu, A. L. Markhard, S. Nada et al., Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids, Cell, vol.141, pp.290-303, 2010.

M. F. Sanmamed and L. Chen, Inducible expression of B7-H1 (PD-L1) and its selective role in tumor site immune modulation, Cancer J, vol.20, pp.256-61, 2014.

M. R. Sarker, S. Franks, N. Sumien, N. Thangthaeng, F. Filipetto et al., Curcumin Mimics the Neurocognitive and Anti-Inflammatory Effects of Caloric Restriction in a Mouse Model of Midlife Obesity, PLoS One, vol.10, p.140431, 2015.

V. I. Sayin, S. E. Leboeuf, S. X. Singh, S. M. Davidson, D. Biancur et al., , 2017.

D. M. Schewe and J. A. Aguirre-ghiso, ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo, Proc Natl Acad Sci U S A, vol.105, pp.10519-10543, 2008.

T. Schmelzle and M. N. Hall, TOR, a central controller of cell growth, Cell, vol.103, pp.253-62, 2000.

A. H. Schonthal, Endoplasmic reticulum stress: its role in disease and novel prospects for therapy, Scientifica (Cairo), vol.2012, p.857516, 2012.

M. Scortegagna, H. Kim, J. L. Li, H. Yao, L. M. Brill et al., Fine tuning of the UPR by the ubiquitin ligases Siah1/2, PLoS Genet, vol.10, p.1004348, 2014.

B. Seliger, M. J. Maeurer, and S. Ferrone, Antigen-processing machinery breakdown and tumor growth, Immunol Today, vol.21, pp.455-64, 2000.

L. Senovilla, I. Vitale, I. Martins, M. Tailler, C. Pailleret et al., An immunosurveillance mechanism controls cancer cell ploidy. Science, vol.337, pp.1678-84, 2012.

S. Shalapour and M. Karin, Immunity, inflammation, and cancer: an eternal fight between good and evil, J Clin Invest, vol.125, pp.3347-55, 2015.

N. P. Shanware, K. Bray, C. H. Eng, F. Wang, M. Follettie et al., Glutamine deprivation stimulates mTOR-JNK-dependent chemokine secretion, Nat Commun, vol.5, p.4900, 2014.

Y. Shi, E. Felley-bosco, T. M. Marti, K. Orlowski, M. Pruschy et al., Starvation-induced activation of ATM/Chk2/p53 signaling sensitizes cancer cells to cisplatin, BMC Cancer, vol.12, p.571, 2012.

C. S. Shin, P. Mishra, J. D. Watrous, V. Carelli, D. 'aurelio et al., The glutamate/cystine xCT antiporter antagonizes glutamine metabolism and reduces nutrient flexibility, Nat Commun, vol.8, p.15074, 2017.

S. J. Simpson, L. Couteur, D. G. , J. De, J. George et al., The Geometric Framework for Nutrition as a tool in precision medicine, Nutr Healthy Aging, vol.4, pp.217-226, 2017.

L. V. Sinclair, J. Rolf, E. Emslie, Y. B. Shi, P. M. Taylor et al., Control of amino-acid transport by antigen receptors coordinates the metabolic reprogramming essential for T cell differentiation, Nat Immunol, vol.14, pp.500-508, 2013.

A. Sistigu, T. Yamazaki, E. Vacchelli, K. Chaba, D. P. Enot et al., Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy, Nat Med, vol.20, p.1301, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02047408

D. L. Smith, C. F. Elam, J. Mattison, J. A. Lane, M. A. Roth et al., Metformin supplementation and life span in Fischer-344 rats, J Gerontol A Biol Sci Med Sci, vol.65, pp.468-74, 2010.

J. A. Smith, A new paradigm: innate immune sensing of viruses via the unfolded protein response, Front Microbiol, vol.5, p.222, 2014.

J. A. Smith, M. J. Turner, M. L. Delay, E. I. Klenk, D. P. Sowders et al., Endoplasmic reticulum stress and the unfolded protein response are linked to synergistic IFN-beta induction via X-box binding protein 1, Eur J Immunol, vol.38, pp.1194-203, 2008.

S. M. Solon-biet, A. C. Mcmahon, J. W. Ballard, K. Ruohonen, L. E. Wu et al., The ratio of macronutrients, not caloric intake, dictates cardiometabolic health, aging, and longevity in ad libitum-fed mice, Cell Metab, vol.19, pp.418-448, 2014.

S. M. Solon-biet, S. J. Mitchell, S. C. Coogan, V. C. Cogger, R. Gokarn et al., Dietary Protein to Carbohydrate Ratio and Caloric Restriction: Comparing Metabolic Outcomes in Mice, Cell Rep, vol.11, pp.1529-1563, 2015.

M. Song and E. L. Giovannucci, Cancer risk: many factors contribute, Science, vol.347, pp.728-737, 2015.

M. Song and E. L. Giovannucci, RE: Doll and Peto's Quantitative Estimates of Cancer Risks: Holding Generally True for 35 Years, J Natl Cancer Inst, vol.107, 2015.

J. R. Speakman, S. E. Mitchell, and M. Mazidi, Calories or protein? The effect of dietary restriction on lifespan in rodents is explained by calories alone, Exp Gerontol, vol.86, pp.28-38, 2016.

R. Spisek, A. Charalambous, A. Mazumder, D. H. Vesole, S. Jagannath et al., Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications, Blood, vol.109, pp.4839-4884, 2007.

S. Spranger, Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment, Int Immunol, vol.28, pp.383-91, 2016.

R. Sriburi, H. Bommiasamy, G. L. Buldak, G. R. Robbins, M. Frank et al., Coordinate regulation of phospholipid biosynthesis and secretory pathway gene expression in XBP-1(S)-induced endoplasmic reticulum biogenesis, J Biol Chem, vol.282, pp.7024-7058, 2007.

G. N. Stemmermann, A. M. Nomura, P. H. Chyou, I. Kato, and T. Kuroishi, Cancer incidence in Hawaiian Japanese: migrants from Okinawa compared with those from other prefectures, Jpn J Cancer Res, vol.82, pp.1366-70, 1991.

S. R. Stowell, T. Ju, and R. D. Cummings, Protein glycosylation in cancer, Annu Rev Pathol, vol.10, pp.473-510, 2015.

A. Q. Sukkurwala, I. Martins, Y. Wang, F. Schlemmer, C. Ruckenstuhl et al., Immunogenic calreticulin exposure occurs through a phylogenetically conserved stress pathway involving the chemokine CXCL8, Cell Death Differ, vol.21, pp.59-68, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02109979

M. R. Sullivan and M. G. Vander-heiden, When cancer needs what's non-essential, Nat Cell Biol, vol.19, pp.418-420, 2017.

M. Swamy, S. Pathak, K. M. Grzes, S. Damerow, L. V. Sinclair et al., Glucose and glutamine fuel protein O-GlcNAcylation to control T cell self-renewal and malignancy, Nat Immunol, vol.17, pp.712-732, 2016.

W. R. Swindell, Heat shock proteins in long-lived worms and mice with insulin/insulin-like signaling mutations, Aging (Albany NY), vol.1, pp.573-580, 2009.

S. Talapatra and C. B. Thompson, Growth factor signaling in cell survival: implications for cancer treatment, J Pharmacol Exp Ther, vol.298, pp.873-881, 2001.

A. B. Tam, A. C. Koong, and M. Niwa, Ire1 has distinct catalytic mechanisms for XBP1/HAC1 splicing and RIDD, Cell Rep, vol.9, pp.850-858, 2014.

J. Tang, Y. S. Guo, Y. Zhang, X. L. Yu, L. Li et al., CD147 induces UPR to inhibit apoptosis and chemosensitivity by increasing the transcription of Bip in hepatocellular carcinoma, Cell Death Differ, vol.19, pp.1779-90, 2012.

M. Wang and R. J. Kaufman, The impact of the endoplasmic reticulum protein-folding environment on cancer development, Nat Rev Cancer, vol.14, pp.581-97, 2014.

W. Wang, Q. Xie, X. Zhou, J. Yao, X. Zhu et al., , p.2, 2015.

Y. Wang, M. Swiecki, S. A. Mccartney, and M. Colonna, dsRNA sensors and plasmacytoid dendritic cells in host defense and autoimmunity, Immunol Rev, vol.243, pp.74-90, 2011.

M. Wei, P. Fabrizio, J. Hu, H. Ge, C. Cheng et al., Life span extension by calorie restriction depends on Rim15 and transcription factors downstream of Ras/PKA, Tor, and Sch9, PLoS Genet, vol.4, p.13, 2008.
URL : https://hal.archives-ouvertes.fr/ensl-00708090

S. Weimer, J. Priebs, D. Kuhlow, M. Groth, S. Priebe et al., D-Glucosamine supplementation extends life span of nematodes and of ageing mice, Nat Commun, vol.5, p.3563, 2014.

J. B. Williams, B. L. Horton, Y. Zheng, Y. Duan, J. D. Powell et al., The EGR2 targets LAG-3 and 4-1BB describe and regulate dysfunctional antigen-specific CD8+ T cells in the tumor microenvironment, J Exp Med, vol.214, pp.381-400, 2017.

M. A. Wolfert and G. J. Boons, Adaptive immune activation: glycosylation does matter, Nat Chem Biol, vol.9, pp.776-84, 2013.

D. M. Woods, A. L. Sodre, A. Villagra, A. Sarnaik, E. M. Sotomayor et al., HDAC Inhibition Upregulates PD-1, 2015.

, Ligands in Melanoma and Augments Immunotherapy with PD-1 Blockade, Cancer Immunol Res, vol.3, pp.1375-85

X. Xia, L. Lei, W. Qin, L. Wang, G. Zhang et al., GCN2 controls the cellular checkpoint: potential target for regulating inflammation, Cell Death Discov, vol.4, p.20, 2018.

X. J. Xia, Y. Y. Gao, J. Zhang, L. Wang, S. Zhao et al., Autophagy mediated by arginine depletion activation of the nutrient sensor GCN2 contributes to interferon-gamma-induced malignant transformation of primary bovine mammary epithelial cells, Cell Death Discov, vol.2, p.15065, 2016.

H. Xie, C. H. Tang, J. H. Song, A. Mancuso, D. Valle et al., IRE1alpha RNase-dependent lipid homeostasis promotes survival in Myc-transformed cancers, J Clin Invest, vol.128, pp.1300-1316, 2018.

T. Yamamori, S. Meike, M. Nagane, H. Yasui, and O. Inanami, ER stress suppresses DNA double-strand break repair and sensitizes tumor cells to ionizing radiation by stimulating proteasomal degradation of Rad51, FEBS Lett, vol.587, pp.3348-53, 2013.

H. Yang, Y. Ma, G. Chen, H. Zhou, T. Yamazaki et al., Contribution of RIP3 and MLKL to immunogenic cell death signaling in cancer chemotherapy, Oncoimmunology, vol.5, p.1149673, 2016.

M. Yang, S. A. Kenfield, E. L. Van-blarigan, J. L. Batista, H. D. Sesso et al., Dietary patterns after prostate cancer diagnosis in relation to disease-specific and total mortality, Cancer Prev Res (Phila), vol.8, pp.545-51, 2015.

X. Yang and K. Qian, Protein O-GlcNAcylation: emerging mechanisms and functions, Nat Rev Mol Cell Biol, vol.18, pp.452-465, 2017.

X. Yang, X. Zhang, M. L. Fu, R. R. Weichselbaum, T. F. Gajewski et al., Targeting the tumor microenvironment with interferon-beta bridges innate and adaptive immune responses, Cancer Cell, vol.25, pp.37-48, 2014.

J. Ye, M. Kumanova, L. S. Hart, K. Sloane, H. Zhang et al., The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation, EMBO J, vol.29, pp.2082-96, 2010.

J. L. Yecies and B. D. Manning, Transcriptional control of cellular metabolism by mTOR signaling, Cancer Res, vol.71, pp.2815-2835, 2011.

A. York, Microbiome: Gut microbiota sways response to cancer immunotherapy, Nat Rev Microbiol, vol.16, p.121, 2018.

K. Yoshida, Y. Hirabayashi, F. Watanabe, T. Sado, and T. Inoue, Caloric restriction prevents radiation-induced myeloid leukemia in C3H/HeMs mice and inversely increases incidence of tumor-free death: implications in changes in number of hemopoietic progenitor cells, Exp Hematol, vol.34, pp.274-83, 2006.

K. Yoshida, T. Inoue, Y. Hirabayashi, T. Matsumura, K. Nemoto et al., Radiation-induced myeloid leukemia in mice under calorie restriction, Leukemia, vol.11, pp.410-412, 1997.

Y. H. Youm, K. Y. Nguyen, R. W. Grant, E. L. Goldberg, M. Bodogai et al., The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease, Nat Med, vol.21, pp.263-272, 2015.

A. Yuan, Y. J. Hsiao, H. Y. Chen, H. W. Chen, C. C. Ho et al., Opposite Effects of M1 and M2 Macrophage Subtypes on, Lung Cancer Progression. Sci Rep, vol.5, p.14273, 2015.

L. Zhai, E. Ladomersky, A. Lenzen, B. Nguyen, R. Patel et al., IDO1 in cancer: a Gemini of immune checkpoints, Cell Mol Immunol, vol.15, pp.447-457, 2018.

J. Zhang, N. N. Pavlova, and C. B. Thompson, Cancer cell metabolism: the essential role of the nonessential amino acid, glutamine, EMBO J, vol.36, pp.1302-1315, 2017.

L. Zhang, J. R. Conejo-garcia, D. Katsaros, P. A. Gimotty, M. Massobrio et al., Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer, N Engl J Med, vol.348, pp.203-216, 2003.

L. Zhang, Z. Li, G. Ding, X. La, P. Yang et al., GRP78 plays an integral role in tumor cell inflammation-related migration induced by M2 macrophages, Cell Signal, vol.37, pp.136-148, 2017.

Y. Zhao, T. Wu, S. Shao, B. Shi, and Y. Zhao, Phenotype, development, and biological function of myeloid-derived suppressor cells, Oncoimmunology, vol.5, p.1004983, 2016.

J. Zhou, B. Mao, Q. Zhou, D. Ding, M. Wang et al., Endoplasmic reticulum stress activates telomerase, Aging Cell, vol.13, pp.197-200, 2014.

L. Zhu, K. Ploessl, R. Zhou, D. Mankoff, and H. F. Kung, Metabolic Imaging of Glutamine in Cancer, J Nucl Med, vol.58, pp.533-537, 2017.

X. Zhu, S. Pattenden, and R. Bremner, pRB is required for interferon-gamma-induction of the MHC class II abeta gene, Oncogene, vol.18, pp.4940-4947, 1999.

A. Zink, H. Rohrbach, U. Szeimies, H. G. Hagedorn, C. J. Haas et al., Malignant tumors in an ancient Egyptian population, Anticancer Res, vol.19, pp.4273-4280, 1999.

L. Zitvogel, Y. Ma, D. Raoult, G. Kroemer, and T. F. Gajewski, The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies, Science, vol.359, pp.1366-1370, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01791568

, Anexes 2. Article, vol.2

C. Rubio-patino, J. P. Bossowski, E. Villa, L. Mondragon, B. Zunino et al., Low carbohydrate diet prevents Mcl-1-mediated resistance to BH3-mimetics, Oncotarget, vol.7, pp.73270-73279, 2016.

C. Hetz, Proteostasis control by the unfolded protein response, Nat. Cell Biol, vol.17, pp.829-838, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01175531

M. Aguilera, M. Oliveros, M. Martínez-padró-n, J. A. Barbas, and A. Ferrú-s,

, Ariadne-1: a vital Drosophila gene is required in development and defines a new conserved family of ring-finger proteins, Genetics, vol.155, pp.1231-1244

R. N. Alcalay, L. N. Clark, K. S. Marder, and W. E. Bradley, Lack of association between cancer history and PARKIN genotype: a family based study in PARKIN/Parkinson's families, Genes Chromosomes Cancer, vol.51, pp.1109-1113, 2012.

R. Beroukhim, C. H. Mermel, D. Porter, G. Wei, S. Raychaudhuri et al., The landscape of somatic copy-number alteration across human cancers, Nature, vol.463, pp.899-905, 2010.

Z. Cakir, K. Funk, J. Lauterwasser, F. Todt, R. M. Zerbes et al., Parkin promotes proteasomal degradation of misregulated BAX, J. Cell Sci, 2017.

D. F. Calvisi, S. Ladu, A. Gorden, M. Farina, J. S. Lee et al., Mechanistic and prognostic significance of aberrant methylation in the molecular pathogenesis of human hepatocellular carcinoma, J. Clin. Invest, vol.117, pp.2713-2722, 2007.

R. G. Carroll, E. Hollville, and S. J. Martin, Parkin sensitizes toward apoptosis induced by mitochondrial depolarization through promoting degradation of Mcl-1, Cell Rep, vol.9, pp.1538-1553, 2014.

R. Cesari, E. S. Martin, G. A. Calin, F. Pentimalli, R. Bichi et al., Parkin, a gene implicated in autosomal recessive juvenile parkinsonism, is a candidate tumor suppressor gene on chromosome 6q25-q27, Proc. Natl. Acad. Sci. USA, vol.100, pp.5956-5961, 2003.

Y. Chen and G. W. Dorn, PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria, Science, vol.340, pp.471-475, 2013.

A. H. Chourasia, M. L. Boland, and K. F. Macleod, Mitophagy and cancer, 2015.

I. E. Clark, M. W. Dodson, C. Jiang, J. H. Cao, J. R. Huh et al., Drosophila pink1 is required for mitochondrial function and interacts genetically with parkin, Nature, vol.441, pp.1162-1166, 2006.

F. Elmehdawi, G. Wheway, K. Szymanska, M. Adams, A. S. High et al., Human Homolog of Drosophila Ariadne (HHARI) is a marker of cellular proliferation associated with nuclear bodies, Exp. Cell Res, vol.319, pp.161-172, 2013.

M. Erkan, J. Kleeff, I. Esposito, T. Giese, K. Ketterer et al., Loss of BNIP3 expression is a late event in pancreatic cancer contributing to chemoresistance and worsened prognosis, Oncogene, vol.24, pp.4421-4432, 2005.

Y. Gong, T. I. Zack, L. G. Morris, K. Lin, E. Hukkelhoven et al., Pan-cancer genetic analysis identifies PARK2 as a master regulator of G1/S cyclins, Nat. Genet, vol.46, pp.588-594, 2014.

J. M. Heo, A. Ordureau, J. A. Paulo, J. Rinehart, and J. W. Harper, The PINK1-PARKIN Mitochondrial Ubiquitylation Pathway Drives a Program of OPTN/NDP52 recruitment and TBK1 activation to promote mitophagy, Mol. Cell, vol.60, pp.7-20, 2015.

L. Herhaus and I. Dikic, Expanding the ubiquitin code through posttranslational modification, EMBO Rep, vol.16, pp.1071-1083, 2015.

E. Hollville, R. G. Carroll, S. P. Cullen, and S. J. Martin, Bcl-2 family proteins participate in mitochondrial quality control by regulating Parkin/ PINK1-dependent mitophagy, Mol. Cell, vol.55, pp.451-466, 2014.

B. N. Johnson, A. K. Berger, G. P. Cortese, and M. J. Lavoie, The ubiquitin E3 ligase parkin regulates the proapoptotic function of Bax, Proc. Natl. Acad. Sci. USA, vol.109, pp.6283-6288, 2012.

L. A. Kane, M. Lazarou, A. I. Fogel, Y. Li, K. Yamano et al., PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity, J. Cell Biol, vol.205, pp.143-153, 2014.

H. Katayama, T. Kogure, N. Mizushima, T. Yoshimori, and A. Miyawaki, A sensitive and quantitative technique for detecting autophagic events based on lysosomal delivery, Chem. Biol, vol.18, pp.1042-1052, 2011.

, Clonogenic assay of A549 control cells (siCont) or cells silenced for ARIH1 (siARIH1) and treated with cisplatin (25 mM) for 6 hr. Pictures were taken 5 days after treatment. (F and G) A549 cells silenced for ARIH1 with two different siRNAs were treated with increasing concentrations of cisplatin, Apoptosis was analyzed by immunoblotting for PARP cleavage (F) and DEVDase activity (G)

, A549 cells silenced for ARIH1 were treated for 16 hr with the indicated amounts of CCCP or cisplatin (25 mM), and apoptosis was assessed using propidium iodide staining and flow cytometry measuring sub-G1 DNA fragmentation

C. A549-crispr-ctl and . Pink1, were treated for 48 hr with cisplatin (25 mM) or etoposide (25 mM) or irradiated with UV (40 J/m 2 ). Plasma membrane permeabilization (i.e., cell death) was assessed using DAPI staining and analyzed by flow cytometry

. A549-crispr-ctl, A549 CRISPR ATG7, and A549 CRISPR ATG12 were treated as described in (I)

A. Kazlauskaite, R. J. Martínez-torres, S. Wilkie, A. Kumar, J. Peltier et al., Binding to serine 65-phosphorylated ubiquitin primes Parkin for optimal PINK1-dependent phosphorylation and activation, EMBO Rep, vol.16, pp.939-954, 2015.

O. Kerscher, R. Felberbaum, and M. Hochstrasser, Modification of proteins by ubiquitin and ubiquitin-like proteins, Annu. Rev. Cell Dev. Biol, vol.22, pp.159-180, 2006.

T. Kitada, S. Asakawa, N. Hattori, H. Matsumine, Y. Yamamura et al., , 1998.

F. Koyano, K. Okatsu, H. Kosako, Y. Tamura, E. Go et al., Ubiquitin is phosphorylated by PINK1 to activate parkin, Nature, vol.510, pp.162-166, 2014.

Y. Kulathu and D. Komander, Atypical ubiquitylation: the unexplored world of polyubiquitin beyond Lys48 and Lys63 linkages, Nat. Rev. Mol. Cell Biol, vol.13, pp.508-523, 2012.

M. Lazarou, D. A. Sliter, L. A. Kane, S. A. Sarraf, C. Wang et al., The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy, Nature, vol.524, pp.309-314, 2015.

O. Meynet, M. Bé-né-teau, M. A. Jacquin, L. A. Pradelli, A. Cornille et al., Glycolysis inhibition targets Mcl-1 to restore sensitivity of lymphoma cells to ABT-737-induced apoptosis, Leukemia, vol.26, pp.1145-1147, 2012.

O. Meynet, B. Zunino, L. Happo, L. A. Pradelli, J. Chiche et al., Caloric restriction modulates Mcl-1 expression and sensitizes lymphomas to BH3 mimetic in mice, Blood, vol.122, pp.2402-2411, 2013.

T. P. Moynihan, H. C. Ardley, U. Nuber, S. A. Rose, P. F. Jones et al., The ubiquitin-conjugating enzymes UbcH7 and UbcH8 interact with RING finger/IBR motif-containing domains of HHARI and H7-AP1, J. Biol. Chem, vol.274, pp.30963-30968, 1999.

V. Nagy and I. Dikic, Ubiquitin ligase complexes: from substrate selectivity to conjugational specificity, Biol. Chem, vol.391, pp.163-169, 2010.

D. Narendra, A. Tanaka, D. F. Suen, Y. , and R. J. , , 2008.

D. Narendra, L. A. Kane, D. N. Hauser, I. M. Fearnley, Y. et al., , 2010.

, /SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both, Autophagy, vol.6, pp.1090-1106

K. Okatsu, K. Saisho, M. Shimanuki, K. Nakada, H. Shitara et al., , 2010.

S. S. Parelkar, J. G. Cadena, C. Kim, Z. Wang, R. Sugal et al., , 2012.

J. Park, S. B. Lee, S. Lee, Y. Kim, S. Song et al., Mitochondrial dysfunction in Drosophila PINK1 mutants is complemented by parkin, Nature, vol.441, pp.1157-1161, 2006.

L. A. Pradelli, M. Bé-né-teau, C. Chauvin, M. A. Jacquin, S. Marchetti et al., Glycolysis inhibition sensitizes tumor cells to death receptors-induced apoptosis by AMP kinase activation leading to Mcl-1 block in translation, Oncogene, vol.29, pp.1641-1652, 2010.

B. Richter, D. A. Sliter, L. Herhaus, A. Stolz, C. Wang et al., Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria, Proc. Natl. Acad. Sci. USA, vol.113, pp.4039-4044, 2016.

D. C. Scott, D. Y. Rhee, D. M. Duda, I. R. Kelsall, J. L. Olszewski et al., Two distinct types of E3 ligases work in unison to regulate substrate ubiquitylation, Cell, vol.166, pp.1198-1214, 2016.

H. Shimura, N. Hattori, S. Kubo, Y. Mizuno, S. Asakawa et al., Familial Parkinson disease gene product, parkin, is a ubiquitin-protein ligase, Nat. Genet, vol.25, pp.302-305, 2000.

D. E. Spratt, H. Walden, and G. S. Shaw, RBR E3 ubiquitin ligases: new structures, new insights, new questions, Biochem. J, vol.458, pp.421-437, 2014.

A. M. Taherbhoy, S. W. Tait, S. E. Kaiser, A. H. Williams, A. Deng et al., , 2011.

, Atg8 transfer from Atg7 to Atg3: a distinctive E1-E2 architecture and mechanism in the autophagy pathway, Mol. Cell, vol.44, pp.451-461

N. G. Tan, H. C. Ardley, S. A. Rose, J. P. Leek, A. F. Markham et al., Characterisation of the human and mouse orthologues of the Drosophila ariadne gene, Cytogenet. Cell Genet, vol.90, pp.242-245, 2000.

N. G. Tan, H. C. Ardley, G. B. Scott, S. A. Rose, A. F. Markham et al., Human homologue of ariadne promotes the ubiquitylation of translation initiation factor 4E homologous protein, 4EHP. FEBS Lett, vol.554, pp.501-504, 2003.

S. Veeriah, L. Morris, D. Solit, C. , and T. A. , The familial Parkinson disease gene PARK2 is a multisite tumor suppressor on chromosome 6q25.2-27 that regulates cyclin E, Cell Cycle, vol.9, pp.1451-1452, 2010.

S. Veeriah, B. S. Taylor, S. Meng, F. Fang, E. Yilmaz et al., Somatic mutations of the Parkinson's disease-associated gene PARK2 in glioblastoma and other human malignancies, Nat. Genet, vol.42, pp.77-82, 2010.

E. Villa and J. E. Ricci, How does metabolism affect cell death in cancer?, FEBS J, vol.283, pp.2653-2660, 2016.

L. Von-stechow, D. Typas, J. Carreras-puigvert, L. Oort, R. Siddappa et al., The E3 ubiquitin ligase ARIH1 protects against genotoxic stress by initiating a 4EHP-mediated mRNA translation arrest, Mol. Cell. Biol, vol.35, pp.1254-1268, 2015.

D. C. Wallace, A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine, Annu. Rev. Genet, vol.39, pp.359-407, 2005.

T. Wang, J. J. Wei, D. M. Sabatini, and E. S. Lander, Genetic screens in human cells using the CRISPR-Cas9 system, Science, vol.343, pp.80-84, 2014.

T. Wauer, M. Simicek, A. Schubert, and D. Komander, Mechanism of phospho-ubiquitin-induced PARKIN activation, Nature, vol.524, pp.370-374, 2015.

H. Wei, L. Liu, C. , and Q. , Selective removal of mitochondria via mitophagy: distinct pathways for different mitochondrial stresses, Biochim. Biophys. Acta, vol.1853, issue.10, pp.2784-2790, 2015.

D. M. Wenzel, A. Lissounov, P. S. Brzovic, and R. E. Klevit, UBCH7 reactivity profile reveals parkin and HHARI to be RING/HECT hybrids, Nature, vol.474, pp.105-108, 2011.

L. Xu, D. C. Lin, D. Yin, and H. P. Koeffler, An emerging role of PARK2 in cancer, J. Mol. Med. (Berl.), vol.92, pp.31-42, 2014.

, France 5 Immunogenomics and Inflammation Research Unit EA 4130, pp.1052-5286

, Laboratoire d'Excellence TOUCAN; Programme Hospitalo-Universitaire en Cancérologie CAPTOR

F. Toulouse,

U. Ciri, ;. De-lyon, and . Inserm-u1111;-ens-de-lyon,

;. Université-lyon1 and . Cnrs, 69007 Lyon, France *equal contribution 10 Corresponding authors: Els Verhoeyen (els.verhoeyen@unice.fr/els.verhoeyen@ens-lyon.fr) and Jean-Ehrland Ricci (ricci@unice.fr) 11 Lead contact: Els Verhoeyen

, Phone: +33 4 89 06 43 01; Fax +33 4 89 06 42 21, email: els.verhoeyen@unice.fr/els.verhoeyen@ens-lyon, vol.6

J. Obacz, T. Avril, C. Rubio-patino, J. P. Bossowski, A. Igbaria et al., Regulation of tumor-stroma interactions by the unfolded protein response, FEBS J, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01808307

N. Dejeans, S. Manie, C. Hetz, F. Bard, T. Hupp et al., Addicted to secrete -novel concepts and targets in cancer therapy, Trends Mol Med, vol.20, pp.242-250, 2014.

C. Hetz, E. Chevet, and S. A. Oakes, Proteostasis control by the unfolded protein response, Nat Cell Biol, vol.17, pp.829-838, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01175531

D. Ron and P. Walter, Signal integration in the endoplasmic reticulum unfolded protein response, Nat Rev Mol Cell Biol, vol.8, pp.519-529, 2007.

E. Chevet, C. Hetz, and A. Samali, Endoplasmic reticulum stress-activated cell reprogramming in oncogenesis, Cancer Discov, vol.5, pp.586-597, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01152845

B. Gardner and P. Walter, Unfolded proteins are Ire1-activating ligands that directly induce the unfolded protein response, Science, vol.333, pp.1891-1894, 2011.

G. E. Karagoz, D. Acosta-alvear, H. T. Nguyen, C. P. Lee, F. Chu et al., An unfolded protein-induced conformational switch activates mammalian IRE1, Elife, vol.6, p.30700, 2017.

H. Urra, E. Dufey, A. T. Chevet, E. Hetz, and C. , Endoplasmic reticulum stress and the hallmarks of cancer, Trends Cancer, vol.2, pp.252-262, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01308007

P. J. Le-reste, A. T. Quillien, V. Morandi, X. Chevet, and E. , Signaling the unfolded protein response in primary brain cancers, Brain Res, vol.1642, pp.9-69, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01295652

C. Hetz, The unfolded protein response: controlling cell fate decisions under ER stress and beyond, Nat Rev Mol Cell Biol, vol.13, pp.9-102, 2012.

N. Dejeans, K. Barroso, M. E. Fernandez-zapico, A. Samali, and E. Chevet, Novel roles of the unfolded protein response in the control of tumor development and aggressiveness, Semin Cancer Biol, vol.33, pp.7-73, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01150385

P. Walter and D. Ron, The unfolded protein response: from stress pathway to homeostatic regulation, Science, vol.334, pp.1081-1086, 2011.

C. Hetz, F. Martinon, D. Rodriguez, and L. H. Glimcher, The unfolded protein response: integrating stress signals through the stress sensor IRE1alpha, Physiol Rev, vol.91, pp.1219-1243, 2011.

M. Maurel, E. Chevet, J. Tavernier, and S. Gerlo, Getting RIDD of RNA: IRE1 in cell fate regulation, Trends Biochem Sci, vol.39, pp.245-254, 2014.

F. Urano, X. Wang, A. Bertolotti, Y. Zhang, P. Chung et al., Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1, Science, vol.287, pp.664-666, 2000.

M. Maurel, E. P. Mcgrath, K. Mnich, S. Healy, E. Chevet et al., Controlling the unfolded protein response-mediated life and death decisions in cancer, Semin Cancer Biol, vol.33, pp.7-66, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01163729

M. Sato, V. J. Yao, W. Arap, and R. Pasqualini, GRP78 signaling hub a receptor for targeted tumor therapy, Adv Genet, vol.69, pp.7-114, 2010.

P. Ren, C. Chen, J. Yue, J. Zhang, and Z. Yu, High expression of glucose-regulated protein 78 (GRP78) is associated with metastasis and poor prognosis in patients with esophageal squamous cell carcinoma, Onco Targets Ther, vol.10, pp.617-625, 2017.

L. Yang, S. Yang, J. Liu, X. Wang, J. J. Cao et al., Expression of GRP78 predicts taxane-based therapeutic resistance and recurrence of human gastric cancer, Exp Mol Pathol, vol.96, pp.235-241, 2014.

H. C. Zheng, H. Takahashi, X. H. Li, T. Hara, S. Masuda et al., Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas, Hum Pathol, vol.39, pp.1042-1049, 2008.

S. L. Rosekrans, M. C. Wielenga, M. Van-de-wetering, M. Ferrante, A. S. Lee et al.,

L. Vermeulen and H. J. Snippert, Stem cell dynamics in homeostasis and cancer of the intestine, Nat Rev Cancer, vol.14, pp.468-480, 2014.

L. Niederreiter, T. M. Fritz, T. E. Adolph, A. M. Krismer, F. A. Offner et al., ER stress transcription factor Xbp1 suppresses intestinal tumorigenesis and directs intestinal stem cells, J Exp Med, vol.210, pp.2041-2056, 2013.

L. Vincenz, R. Jager, M. O'dwyer, and A. Samali, Endoplasmic reticulum stress and the unfolded protein response: targeting the Achilles heel of multiple myeloma, Mol Cancer Ther, vol.12, pp.831-843, 2013.

F. Tameire, I. I. Verginadis, and C. Koumenis, Cell intrinsic and extrinsic activators of the unfolded protein response in cancer: mechanisms and targets for therapy, Semin Cancer Biol, vol.33, pp.3-15, 2015.

E. Dufey, H. Urra, and C. Hetz, ER proteostasis addiction in cancer biology: novel concepts, Semin Cancer Biol, vol.33, pp.0-47, 2015.

S. A. Oakes, Endoplasmic reticulum proteostasis: a key checkpoint in cancer, Am J Physiol Cell Physiol, vol.312, pp.93-102, 2017.

X. Chen, D. Iliopoulos, Q. Zhang, Q. Tang, M. B. Greenblatt et al., XBP1 promotes triple-negative breast cancer by controlling the HIF1alpha pathway, Nature, vol.508, pp.103-107, 2014.

X. Hou, Y. Liu, H. Liu, X. Chen, M. Liu et al., PERK silence inhibits glioma cell growth under low glucose stress by blockage of p-AKT and subsequent HK2's mitochondria translocation, Sci Rep, vol.5, p.9065, 2015.

J. Obacz, A. T. , L. Reste, P. J. Urra, H. Quillien et al., Endoplasmic reticulum proteostasis in glioblastoma-From molecular mechanisms to therapeutic perspectives, Sci Signal, vol.10, p.2323, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01502552

Y. Bu and J. A. Diehl, PERK integrates oncogenic signaling and cell survival during cancer development, J Cell Physiol, vol.231, pp.2088-2096, 2016.

M. Blaustein, D. Perez-munizaga, M. A. Sanchez, C. Urrutia, A. Grande et al., Modulation of the Akt pathway reveals a novel link with PERK/eIF2alpha, which is relevant during hypoxia, PLoS One, vol.8, p.69668, 2013.

K. M. Rouschop, L. J. Dubois, T. G. Keulers, T. Van-den-beucken, P. Lambin et al., PERK/ eIF2alpha signaling protects therapy resistant hypoxic cells through induction of glutathione synthesis and protection against ROS, Proc Natl Acad Sci, vol.110, pp.4622-4627, 2013.

P. Freis, J. Bollard, J. Lebeau, P. Massoma, J. Fauvre et al., ) mTOR inhibitors activate PERK signaling and favor viability of gastrointestinal neuroendocrine cell lines, Oncotarget, vol.8, pp.20974-20987, 2017.

M. S. Sosa, P. Bragado, and J. A. Aguirre-ghiso, Mechanisms of disseminated cancer cell dormancy: an awakening field, Nat Rev Cancer, vol.14, pp.611-622, 2014.

D. M. Schewe and J. A. Aguirre-ghiso, ATF6alpha-Rheb-mTOR signaling promotes survival of dormant tumor cells in vivo, Proc Natl Acad Sci, vol.105, pp.10519-10524, 2008.

A. C. Ranganathan, L. Zhang, A. P. Adam, and J. A. Aguirreghiso, Functional coupling of p38-induced up-regulation of BiP and activation of RNAdependent protein kinase-like endoplasmic reticulum kinase to drug resistance of dormant carcinoma cells, Cancer Res, vol.66, pp.1702-1711, 2006.

Y. X. Feng, E. S. Sokol, D. Vecchio, C. A. Sanduja, S. Claessen et al., Epithelial-to-mesenchymal transition activates PERK-eIF2alpha and sensitizes cells to endoplasmic reticulum stress, Cancer Discov, vol.4, pp.702-715, 2014.

S. Dey, C. M. Sayers, I. I. Verginadis, S. L. Lehman, Y. Cheng et al., ATF4-dependent induction of heme oxygenase 1 prevents anoikis and promotes metastasis, J Clin Invest, vol.125, pp.2592-2608, 2015.

H. Mujcic, A. Nagelkerke, K. M. Rouschop, S. Chung, N. Chaudary et al., Hypoxic activation of the PERK/eIF2alpha arm of the unfolded protein response promotes metastasis through induction of LAMP3, Clin Cancer Res, vol.19, pp.6126-6137, 2013.

X. Shen, Y. Xue, Y. Si, Q. Wang, Z. Wang et al., The unfolded protein response potentiates epithelial-to-mesenchymal transition (EMT) of gastric cancer cells under severe hypoxic conditions, 2015.

, Med Oncol, vol.32, p.447

G. Auf, A. Jabouille, S. Guerit, R. Pineau, M. Delugin et al., Inositol-requiring enzyme 1alpha is a key regulator of angiogenesis and invasion in malignant glioma, Proc Natl Acad Sci, vol.107, pp.15553-15558, 2010.

N. Dejeans, O. Pluquet, S. Lhomond, F. Grise, M. Bouchecareilh et al.,

C. A. Lyssiotis and A. C. Kimmelman, Metabolic interactions in the tumor microenvironment, Trends Cell Biol, vol.27, pp.863-875, 2017.

Y. Mezawa and A. Orimo, The roles of tumor-and metastasis-promoting carcinoma-associated fibroblasts in human carcinomas, Cell Tissue Res, vol.365, pp.675-689, 2016.

N. Maishi and K. Hida, Tumor endothelial cells accelerate tumor metastasis, Cancer Sci, vol.108, pp.1921-1926, 2017.

T. L. Whiteside, The tumor microenvironment and its role in promoting tumor growth, Oncogene, vol.27, pp.5904-5912, 2008.

W. H. Fridman, L. Zitvogel, C. Sautes-fridman, and G. Kroemer, The immune contexture in cancer prognosis and treatment, Nat Rev Clin Oncol, vol.14, pp.717-734, 2017.

W. H. Goldmann, Mechanical aspects of cell shape regulation and signaling, Cell Biol Int, vol.26, pp.313-317, 2002.

H. J. Jongsma and R. Wilders, Gap junctions in cardiovascular disease, Circ Res, vol.86, pp.1193-1197, 2000.

T. Huang, Y. Wan, Y. Zhu, X. Fang, N. Hiramatsu et al., Downregulation of gap junction expression and function by endoplasmic reticulum stress, J Cell Biochem, vol.107, pp.973-983, 2009.

H. Yamasaki, M. Mesnil, Y. Omori, N. Mironov, and V. Krutovskikh, Mutat Res, vol.333, pp.181-188, 1995.

S. Yang, Q. Wen, Y. Liu, C. Zhang, M. Wang et al., Increased expression of CX43 on stromal cells promotes leukemia apoptosis, Oncotarget, vol.6, pp.44323-44331, 2015.

H. Yamasaki, Gap junctional intercellular communication and carcinogenesis, Carcinogenesis, vol.11, pp.1051-1058, 1990.

N. M. Kumar and N. B. Gilula, The gap junction communication channel, Cell, vol.84, pp.381-388, 1996.

T. Aasen, M. Mesnil, C. C. Naus, P. Lampe, and D. W. Laird, Gap junctions and cancer: communicating for 50 years, Nat Rev Cancer, vol.17, p.74, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02129361

T. Aasen, M. Mesnil, C. C. Naus, P. Lampe, and D. W. Laird, Gap junctions and cancer: communicating for 50 years, Nat Rev Cancer, vol.16, pp.775-788, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02129361

J. Trosko and C. C. Chang, Mechanism of upregulated gap junctional intercellular communication during chemoprevention and chemotherapy of cancer, 2001.

, Mutat Res, pp.219-229

R. P. Huang, M. Z. Hossain, R. Huang, J. Gano, Y. Fan et al., Connexin 43 (cx43) enhances chemotherapy-induced apoptosis in human glioblastoma cells, Int J Cancer, vol.92, pp.130-138, 2001.

A. Raza, A. Ghoshal, S. Chockalingam, and S. S. Ghosh, Connexin-43 enhances tumor suppressing activity of artesunate via gap junction-dependent as well as independent pathways in human breast cancer cells, Sci Rep, vol.7, p.7580, 2017.

T. J. King and P. D. Lampe, The gap junction protein connexin32 is a mouse lung tumor suppressor, Cancer Res, vol.64, pp.7191-7196, 2004.

L. Koffler, S. Roshong, K. Park, I. Cesen-cummings, K. Thompson et al., Growth inhibition in G(1) and altered expression of cyclin D1 and p27(kip-1)after forced connexin expression in lung and liver carcinoma cells, J Cell Biochem, vol.79, pp.347-354, 2000.

E. Leithe, M. Mesnil, and T. Aasen, The connexin 43 C-terminus: a tail of many tales, Biochim Biophys Acta, vol.1860, pp.4-8, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02129358

X. Y. Ye, Q. H. Jiang, T. Hong, Z. Y. Zhang, R. J. Yang et al., Altered expression of connexin43 and phosphorylation connexin43 in glioma tumors, Int J Clin Exp Pathol, vol.8, pp.4296-4306, 2015.

V. Su and A. F. Lau, Connexins: mechanisms regulating protein levels and intercellular communication, FEBS Lett, vol.588, pp.1212-1220, 2014.

L. S. Musil, A. C. Le, J. K. Vanslyke, and L. M. Roberts, Regulation of connexin degradation as a mechanism to increase gap junction assembly and function, J Biol Chem, vol.275, pp.25207-25215, 2000.

J. K. Vanslyke, S. M. Deschenes, and L. S. Musil, Intracellular transport, assembly, and degradation of wild-type and disease-linked mutant gap junction proteins, Mol Biol Cell, vol.11, pp.1933-1946, 2000.

J. K. Vanslyke and L. S. Musil, Dislocation and degradation from the ER are regulated by cytosolic stress, J Cell Biol, vol.157, pp.381-394, 2002.

J. K. Vanslyke and L. S. Musil, Degradation of connexins from the plasma membrane is regulated by inhibitors of protein synthesis, Cell Commun Adhes, vol.10, pp.329-333, 2003.

J. K. Vanslyke and L. S. Musil, Cytosolic stress reduces degradation of connexin43 internalized from the cell surface and enhances gap junction formation and function, Mol Biol Cell, vol.16, pp.5247-5257, 2005.

K. Denzer, M. J. Kleijmeer, H. F. Heijnen, W. Stoorvogel, and H. J. Geuze, Exosome: from internal vesicle of the multivesicular body to intercellular signaling device, J Cell Sci, vol.113, pp.3365-3374, 2000.

A. Schneider and M. Simons, Exosomes: vesicular carriers for intercellular communication in neurodegenerative disorders, Cell Tissue Res, vol.352, pp.33-47, 2013.

M. Simons and G. Raposo, Exosomes-vesicular carriers for intercellular communication, Curr Opin Cell Biol, vol.21, pp.575-581, 2009.

L. K. Kd-&-mahal, Identification of a conserved glycan signature for microvesicles, J Proteome Res, vol.10, pp.4624-4633, 2011.

J. Zhang, S. Li, L. Li, M. Li, C. Guo et al., Exosome and exosomal microRNA: trafficking, sorting, and function, Genomics Proteomics Bioinformatics, vol.13, pp.7-24, 2015.

D. J. Gibbings, C. Ciaudo, M. Erhardt, and O. Voinnet, Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity, Nat Cell Biol, vol.11, pp.1143-1149, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00423288

A. Ramteke, H. Ting, C. Agarwal, S. Mateen, R. Somasagara et al., Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules, 2015.

, Mol Carcinog, vol.54, pp.554-565

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

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

Y. Wei, X. Lai, S. Yu, S. Chen, Y. Ma et al., Exosomal miR-221/ 222 enhances tamoxifen resistance in recipient ERpositive breast cancer cells, Breast Cancer Res Treat, vol.147, pp.423-431, 2014.

R. Safaei, B. J. Larson, T. C. Cheng, M. A. Gibson, S. Otani et al., Abnormal lysosomal trafficking and enhanced exosomal export of cisplatin in drug-resistant human ovarian carcinoma cells, Mol Cancer Ther, vol.4, pp.1595-1604, 2005.

V. Y. Chen, M. M. Posada, L. L. Blazer, T. Zhao, and G. R. Rosania, The role of the VPS4A-exosome pathway in the intrinsic egress route of a DNA-binding anticancer drug, Pharm Res, vol.23, pp.1687-1695, 2006.

L. M. Epple, R. D. Dodd, A. L. Merz, A. M. Dechkovskaia, M. Herring et al., Induction of the unfolded protein response drives enhanced metabolism and chemoresistance in glioma cells, PLoS One, vol.8, p.73267, 2013.

G. K. Panigrahi and G. Deep, Exosomes-based biomarker discovery for diagnosis and prognosis of prostate cancer, Front Biosci (Landmark Ed), vol.22, pp.1682-1696, 2017.

F. Properzi, M. Logozzi, and S. Fais, Exosomes: the future of biomarkers in medicine, Biomark Med, vol.7, pp.769-778, 2013.

R. Nedaeinia, M. Manian, M. H. Jazayeri, M. Ranjbar, R. Salehi et al., Circulating exosomes and exosomal microRNAs as biomarkers in gastrointestinal cancer, Cancer Gene Ther, vol.24, pp.4-8, 2017.

A. Zlotogorski-hurvitz, D. Dayan, G. Chaushu, J. Korvala, T. Salo et al., Human saliva-derived exosomes: comparing methods of isolation, J Histochem Cytochem, vol.63, pp.181-189, 2015.

V. Palanisamy, S. Sharma, A. Deshpande, H. Zhou, J. Gimzewski et al., Nanostructural and transcriptomic analyses of human saliva derived exosomes, PLoS One, vol.5, p.8577, 2010.

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

N. Almqvist, A. Lonnqvist, S. Hultkrantz, C. Rask, and E. Telemo, Serum-derived exosomes from antigen-fed mice prevent allergic sensitization in a model of allergic asthma, Immunology, vol.125, pp.1-27, 2008.

C. Y. Chen, M. Hogan, and C. J. Ward, Purification of exosome-like vesicles from urine, Methods Enzymol, vol.524, pp.225-241, 2013.

M. F. Peterson, N. Otoc, J. K. Sethi, A. Gupta, and T. J. Antes, Integrated systems for exosome investigation, Methods, vol.87, pp.3-4, 2015.

L. A. Harshyne, B. J. Nasca, L. C. Kenyon, D. W. Andrews, and D. C. Hooper, Serum exosomes and cytokines promote a T-helper cell type 2 environment in the peripheral blood of glioblastoma patients, Neuro Oncol, vol.18, pp.206-215, 2016.

F. Ciregia, A. Urbani, and G. Palmisano, Extracellular vesicles in brain tumors and neurodegenerative diseases, Front Mol Neurosci, vol.10, p.276, 2017.

M. W. Graner, R. I. Cumming, and D. D. Bigner, The heat shock response and chaperones/heat shock proteins in brain tumors: surface expression, release, and possible immune consequences, J Neurosci, vol.27, pp.11214-11227, 2007.

J. C. Akers, V. Ramakrishnan, R. Kim, S. Phillips, V. Kaimal et al., ) miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients, 2015.

, J Neurooncol, vol.123, pp.205-216

L. Saadatpour, E. Fadaee, S. Fadaei, N. Mansour, R. Mohammadi et al., Glioblastoma: exosome and microRNA as novel diagnosis biomarkers, Cancer Gene Ther, vol.23, pp.415-418, 2016.

L. Manterola, E. Guruceaga, G. Perez-larraya, J. Gonzalez-huarriz, M. Jauregui et al., A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool, Neuro Oncol, vol.16, pp.520-527, 2014.

S. Kanemoto, R. Nitani, T. Murakami, M. Kaneko, R. Asada et al., Multivesicular body formation enhancement and exosome release during endoplasmic reticulum stress, Biochem Biophys Res Commun, vol.480, pp.166-172, 2016.

N. R. Mahadevan, V. Anufreichik, J. J. Rodvold, K. T. Chiu, H. Sepulveda et al., Cell-extrinsic effects of tumor ER stress imprint myeloid dendritic cells and impair CD8(+) T cell priming, PLoS One, vol.7, 2012.

N. R. Mahadevan, J. Rodvold, H. Sepulveda, S. Rossi, A. F. Drew et al., Transmission of endoplasmic reticulum stress and pro-inflammation from tumor cells to myeloid cells, Proc Natl Acad Sci, vol.108, pp.6561-6566, 2011.

M. Zanetti, Cell-extrinsic effects of the tumor unfolded protein response on myeloid cells and T cells, 2013.

, Ann N Y Acad Sci, vol.1284, pp.6-11

J. J. Rodvold, K. T. Chiu, N. Hiramatsu, J. K. Nussbacher, V. Galimberti et al., Intercellular transmission of the unfolded protein response promotes survival and drug resistance in cancer cells, Sci Signal, vol.10, p.7177, 2017.

H. Zhang, Y. Yue, T. Sun, X. Wu, and S. Xiong, Transmissible endoplasmic reticulum stress from myocardiocytes to macrophages is pivotal for the pathogenesis of CVB3-induced viral myocarditis, Sci Rep, vol.7, p.42162, 2017.

H. Urra and C. Hetz, A novel ER stressindependent function of the UPR in angiogenesis, Mol Cell, vol.54, pp.542-544, 2014.

E. R. Pereira, N. Liao, G. A. Neale, and L. M. Hendershot, Transcriptional and post-transcriptional regulation of proangiogenic factors by the unfolded protein response, PLoS One, vol.5, p.12521, 2010.

B. Drogat, A. P. Nguyen, D. T. Bouchecareilh, M. Pineau, R. Nalbantoglu et al., IRE1 signaling is essential for ischemia-induced vascular endothelial growth factor-A expression and contributes to angiogenesis and tumor growth in vivo, Cancer Res, vol.67, pp.6700-6707, 2007.

J. M. Wang, Y. Qiu, Z. Q. Yang, L. Li, and K. Zhang, Inositol-requiring enzyme 1 facilitates diabetic wound healing through modulating microRNAs, Diabetes, vol.66, pp.177-192, 2017.

Y. Wang, G. N. Alam, Y. Ning, F. Visioli, Z. Dong et al., The unfolded protein response induces the angiogenic switch in human tumor cells through the PERK/ATF4 pathway, Cancer Res, vol.72, pp.5396-5406, 2012.

J. D. Blais, C. L. Addison, R. Edge, T. Falls, H. Zhao et al., Perk-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress, Mol Cell Biol, vol.26, pp.9517-9532, 2006.

C. Philippe, A. Dubrac, C. Quelen, A. Desquesnes, L. Van-den-berghe et al., PERK mediates the IRES-dependent translational activation of mRNAs encoding angiogenic growth factors after ischemic stress, Sci Signal, vol.9, p.44, 2016.

K. Ozawa, Y. Tsukamoto, O. Hori, Y. Kitao, H. Yanagi et al., Regulation of tumor angiogenesis by oxygen-regulated protein 150, an inducible endoplasmic reticulum chaperone, Cancer Res, vol.61, pp.4206-4213, 2001.

E. Karali, S. Bellou, D. Stellas, A. Klinakis, C. Murphy et al., Signals through ATF6 and PERK to promote endothelial cell survival and angiogenesis in the absence of ER stress, Mol Cell, vol.54, pp.559-572, 2014.

F. Visioli, Y. Wang, G. N. Alam, Y. Ning, P. V. Rados et al., Glucose-regulated protein 78 (Grp78) confers chemoresistance to tumor endothelial cells under acidic stress, PLoS One, vol.9, 2014.

K. S. Han, N. Li, P. A. Raven, L. Fazli, S. Frees et al., Inhibition of endoplasmic reticulum chaperone protein glucose-regulated protein 78 potentiates antiangiogenic therapy in renal cell carcinoma through inactivation of the PERK/eIF2alpha pathway, Oncotarget, vol.6, pp.34818-34830, 2015.

Q. Ruan, S. Han, W. G. Jiang, M. E. Boulton, Z. J. Chen et al., alphaB-crystallin, an effector of unfolded protein response, confers anti-VEGF resistance to breast cancer via maintenance of intracrine VEGF in endothelial cells, Mol Cancer Res, vol.9, pp.1632-1643, 2011.

J. R. Cubillos-ruiz, S. E. Bettigole, and L. H. Glimcher, Tumorigenic and immunosuppressive effects of endoplasmic reticulum stress in cancer, Cell, vol.168, pp.692-706, 2017.

W. Shi, Y. Liao, S. N. Willis, N. Taubenheim, M. Inouye et al., Transcriptional profiling of mouse B cell terminal differentiation defines a signature for antibody-secreting plasma cells, Nat Immunol, vol.16, pp.663-673, 2015.

S. Morita, S. A. Villalta, H. C. Feldman, A. C. Register, W. Rosenthal et al., Targeting ABL, 2017.

B. Bujisic, D. Gassart, A. Tallant, R. Demaria, O. Zaffalon et al., Impairment of both IRE1 expression and XBP1 activation is a hallmark of GCB DLBCL and contributes to tumor growth, Blood, vol.129, pp.2420-2428, 2017.

J. Grootjans, A. Kaser, R. J. Kaufman, and R. S. Blumberg, The unfolded protein response in immunity and inflammation, Nat Rev Immunol, vol.16, pp.469-484, 2016.

F. Osorio, S. J. Tavernier, E. Hoffmann, Y. Saeys, L. Martens et al., The unfolded-protein-response sensor IRE-1alpha regulates the function of CD8alpha+ dendritic cells, Nat Immunol, vol.15, pp.248-257, 2014.

S. J. Tavernier, F. Osorio, L. Vandersarren, J. Vetters, N. Vanlangenakker et al., Regulated IRE1-dependent mRNA decay sets the threshold for dendritic cell survival, Nat Cell Biol, vol.19, pp.698-710, 2017.

J. R. Cubillos-ruiz, P. C. Silberman, M. R. Rutkowski, S. Chopra, A. Perales-puchalt et al., ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis, Cell, vol.161, pp.1527-1538, 2015.

F. Martinon, X. Chen, A. H. Lee, and L. H. Glimcher, TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages, 2010.

, Nat Immunol, vol.11, pp.411-418

P. T. Thevenot, R. A. Sierra, P. L. Raber, A. A. Al-khami, J. Trillo-tinoco et al., The stress-response sensor chop regulates the function and accumulation of myeloid-derived suppressor cells in tumors, Immunity, vol.41, pp.389-401, 2014.

H. Nakagawa, A. Umemura, K. Taniguchi, J. Fontburgada, D. Dhar et al., ER stress cooperates with hypernutrition to trigger TNFdependent spontaneous HCC development, Cancer Cell, vol.26, pp.331-343, 2014.

J. C. Goodall, C. Wu, Y. Zhang, L. Mcneill, L. Ellis et al., Endoplasmic reticulum stress-induced transcription factor, CHOP, is crucial for dendritic cell IL-23 expression, Proc Natl Acad Sci, vol.107, pp.17698-17703, 2010.

S. Kim, Y. Joe, H. J. Kim, Y. S. Kim, S. O. Jeong et al., Endoplasmic reticulum stress-induced IRE1alpha activation mediates cross-talk of GSK-3beta and XBP-1 to regulate inflammatory cytokine production, J Immunol, vol.194, pp.4498-4506, 2015.

N. P. Shanware, K. Bray, C. H. Eng, F. Wang, M. Follettie et al., Glutamine deprivation stimulates mTOR-JNKdependent chemokine secretion, Nat Commun, vol.5, p.4900, 2014.

J. Liu, D. Ibi, K. Taniguchi, J. Lee, H. Herrema et al., Inflammation improves glucose homeostasis through IKKbeta-XBP1s interaction, Cell, vol.167, pp.1052-1066, 2016.

A. Shkoda, P. A. Ruiz, H. Daniel, S. C. Kim, G. Rogler et al., Interleukin-10 blocked endoplasmic reticulum stress in intestinal epithelial cells: impact on chronic inflammation, Gastroenterology, vol.132, pp.190-207, 2007.

K. Zhang, X. Shen, J. Wu, K. Sakaki, T. Saunders et al., Endoplasmic reticulum stress activates cleavage of CREBH to induce a systemic inflammatory response, Cell, vol.124, pp.587-599, 2006.

L. Yang, E. S. Calay, J. Fan, A. Arduini, R. C. Kunz et al., METABOLISM. S-Nitrosylation links obesityassociated inflammation to endoplasmic reticulum dysfunction, Science, vol.349, pp.500-506, 2015.
URL : https://hal.archives-ouvertes.fr/cea-01820387

M. M. Robblee, C. C. Kim, J. Porter-abate, M. Valdearcos, K. L. Sandlund et al., Saturated fatty acids engage an IRE1alpha-dependent pathway to activate the NLRP3 inflammasome in myeloid cells, Cell Rep, vol.14, pp.2611-2623, 2016.

K. Halbleib, K. Pesek, R. Covino, H. F. Hofbauer, D. Wunnicke et al., Activation of the unfolded protein response by lipid bilayer stress, Mol Cell, vol.67, pp.673-684, 2017.

D. N. Bronner, B. H. Abuaita, X. Chen, K. A. Fitzgerald, G. Nunez et al., Endoplasmic reticulum stress activates the inflammasome via NLRP3-and caspase-2-driven mitochondrial damage, Immunity, vol.43, pp.451-462, 2015.

L. Galluzzi, A. Buque, O. Kepp, L. Zitvogel, and G. Kroemer, Immunogenic cell death in cancer and infectious disease, Nat Rev Immunol, vol.17, pp.7-111, 2017.

T. Panaretakis, O. Kepp, U. Brockmeier, A. Tesniere, A. C. Bjorklund et al., Mechanisms of pre-apoptotic calreticulin exposure in immunogenic cell death, EMBO J, vol.28, pp.578-590, 2009.

M. Raghavan, S. J. Wijeyesakere, L. Peters, and N. Cid, Calreticulin in the immune system: ins and outs, Trends Immunol, vol.34, pp.3-21, 2013.

L. Senovilla, I. Vitale, I. Martins, M. Tailler, C. Pailleret et al., An immunosurveillance mechanism controls cancer cell ploidy, Science, vol.337, pp.1678-1684, 2012.

I. A. Park, S. H. Heo, I. H. Song, Y. A. Kim, H. S. Park et al., Endoplasmic reticulum stress induces secretion of high-mobility group proteins and is associated with tumor-infiltrating lymphocytes in triple-negative breast cancer, Oncotarget, vol.7, pp.59957-59964, 2016.

K. L. Cook, D. R. Soto-pantoja, P. A. Clarke, M. I. Cruz, A. Zwart et al., Endoplasmic reticulum stress protein GRP78 modulates lipid metabolism to control drug sensitivity and antitumor immunity in breast cancer, Cancer Res, vol.76, pp.5657-5670, 2016.

J. A. Cho, A. H. Lee, B. Platzer, B. C. Cross, B. M. Gardner et al., The unfolded protein response element IRE1alpha senses bacterial proteins invading the ER to activate RIG-I and innate immune signaling, Cell Host Microbe, vol.13, pp.558-569, 2013.

S. C. Eckard, G. I. Rice, A. Fabre, C. Badens, E. E. Gray et al., The SKIV2L RNA exosome limits activation of the RIG-Ilike receptors, Nat Immunol, vol.15, pp.839-845, 2014.

B. Y. Nabet, Y. Qiu, J. E. Shabason, T. J. Wu, T. Yoon et al., Exosome RNA unshielding couples stromal activation to pattern recognition receptor signaling in cancer, Cell, vol.170, pp.352-366, 2017.

J. Dassler-plenker, K. S. Reiners, J. G. Van-den-boorn, H. P. Hansen, B. Putschli et al., RIG-I activation induces the release of extracellular vesicles with antitumor activity, Oncoimmunology, vol.5, p.1219827, 2016.

H. Poeck, R. Besch, C. Maihoefer, M. Renn, D. Tormo et al., 0 -Triphosphate-siRNA: turning gene silencing and Rig-I activation against melanoma, Nat Med, vol.5, pp.1256-1263, 2008.

J. Hou, Y. Zhou, Y. Zheng, J. Fan, W. Zhou et al., Hepatic RIG-I predicts survival and interferon-alpha therapeutic response in hepatocellular carcinoma, Cancer Cell, vol.25, pp.9-63, 2014.

R. Besch, H. Poeck, T. Hohenauer, D. Senft, G. Hacker et al., Proapoptotic signaling induced by RIG-I and MDA-5 results in type I interferon-independent apoptosis in human melanoma cells, J Clin Invest, vol.119, pp.2399-2411, 2009.

P. Duewell, A. Steger, H. Lohr, H. Bourhis, H. Hoelz et al., RIG-I-like helicases induce immunogenic cell death of pancreatic cancer cells and sensitize tumors toward killing by CD8(+) T cells, Cell Death Differ, vol.21, pp.1825-1837, 2014.

H. Y. Su, R. T. Waldron, R. Gong, V. K. Ramanujan, S. J. Pandol et al., The unfolded protein response plays a predominant homeostatic role in response to mitochondrial stress in pancreatic stellate cells, PLoS One, vol.11, p.148999, 2016.

N. Harris, C. Vennin, J. Conway, K. L. Vine, M. Pinese et al., SerpinB2 regulates stromal remodelling and local invasion in pancreatic cancer, Oncogene, vol.36, pp.4288-4298, 2017.

Y. Buganim, S. Madar, Y. Rais, L. Pomeraniec, E. Harel et al., Transcriptional activity of ATF3 in the stromal compartment of tumors promotes cancer progression, Carcinogenesis, vol.32, pp.1749-1757, 2011.

C. Hetz and S. Saxena, ER stress and the unfolded protein response in neurodegeneration, Nat Rev Neurol, vol.13, pp.477-491, 2017.

A. A. Farooqi, K. T. Li, S. Fayyaz, Y. T. Chang, M. Ismail et al., Anticancer drugs for the modulation of endoplasmic reticulum stress and oxidative stress, Tumour Biol, vol.36, pp.5743-5752, 2015.

D. V. Krysko, A. D. Garg, A. Kaczmarek, O. Krysko, P. Agostinis et al., Immunogenic cell death and DAMPs in cancer therapy, Nat Rev Cancer, vol.12, pp.860-875, 2012.

N. Rufo, A. D. Garg, and P. Agostinis, The unfolded protein response in immunogenic cell death and cancer immunotherapy, Trends Cancer, vol.3, pp.643-658, 2017.

I. C. Salaroglio, E. Panada, E. Moiso, I. Buondonno, P. Provero et al., PERK induces resistance to cell death elicited by endoplasmic reticulum stress and chemotherapy, 2017.

, Mol Cancer, vol.16, p.91

A. Higa, S. Taouji, S. Lhomond, D. Jensen, M. E. Fernandezzapico et al., Endoplasmic reticulum stressactivated transcription factor ATF6alpha requires the disulfide isomerase PDIA5 to modulate chemoresistance, Mol Cell Biol, vol.34, pp.1839-1849, 2014.

L. J. Bu, H. Q. Yu, L. L. Fan, X. Q. Li, F. Wang et al., , 2017.

, Melatonin, a novel selective ATF-6 inhibitor, induces human hepatoma cell apoptosis through COX-2 downregulation, World J Gastroenterol, vol.23, pp.986-998

I. Papandreou, N. C. Denko, M. Olson, H. Van-melckebeke, L. S. Tam et al., Identification of an Ire1alpha endonuclease specific inhibitor with cytotoxic activity against human multiple myeloma, Blood, vol.117, pp.1311-1314, 2011.

J. Ming, S. Ruan, M. Wang, D. Ye, N. Fan et al., A novel chemical, STF-083010, reverses tamoxifen-related drug resistance in breast cancer by inhibiting IRE1/XBP1, Oncotarget, vol.6, pp.40692-40703, 2015.

N. Mimura, M. Fulciniti, G. Gorgun, Y. T. Tai, D. Cirstea et al., Blockade of XBP1 splicing by inhibition of IRE1alpha is a promising therapeutic option in multiple myeloma, Blood, vol.119, pp.5772-5781, 2012.

R. Ghosh, L. Wang, E. S. Wang, B. G. Perera, A. Igbaria et al., Allosteric inhibition of the IRE1alpha RNase preserves cell viability and function during endoplasmic reticulum stress, Cell, vol.158, pp.534-548, 2014.

J. M. Axten, J. R. Medina, Y. Feng, A. Shu, S. P. Romeril et al., Discovery of 7-methyl-5-(1-{[3-(trifluoromethyl)phenyl]acetyl}-2,3-dihydro-1H-indol-5-yl)-7H-p yrrolo[2,3-d]pyrimidin-4-amine (GSK2606414), a potent and selective first-in-class inhibitor of protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), J Med Chem, vol.55, pp.7193-7207, 2012.

C. Atkins, Q. Liu, E. Minthorn, S. Y. Zhang, D. J. Figueroa et al., Characterization of a novel PERK kinase inhibitor with antitumor and antiangiogenic activity, Cancer Res, vol.73, 1993.

L. R. Palam, J. Gore, K. E. Craven, J. L. Wilson, and M. Korc, Integrated stress response is critical for gemcitabine resistance in pancreatic ductal adenocarcinoma, Cell Death Dis, vol.6, 1913.

Y. J. Jeon, J. H. Kim, J. I. Shin, M. Jeong, J. Cho et al., Salubrinal-mediated upregulation of eIF2alpha phosphorylation increases doxorubicin sensitivity in MCF-7/ADR cells, Mol Cells, vol.39, pp.129-135, 2016.