, ISCIII PI13-00139); and the European Union's Horizon 2020 Research and Innovation Programme under the Marie Sk?odowska-Curie Grant Agreements, TRAINERS), vol.675448

L. B. Sullivan, Supporting aspartate biosynthesis is an essential function of respiration in proliferating cells, Cell, vol.162, pp.552-563, 2015.

G. Jiménez-valerio and O. Casanovas, Angiogenesis and metabolism: Entwined for therapy resistance, Trends Cancer, vol.3, pp.10-18, 2017.

Y. Wang, 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.

A. Longchamp, Amino acid restriction triggers angiogenesis via GCN2/ATF4 regulation of VEGF and H 2 S production, Cell, vol.173, pp.117-129, 2018.

S. D. Prabhu and N. G. Frangogiannis, The biological basis for cardiac repair after myocardial infarction: From inflammation to fibrosis, Circ. Res, vol.119, pp.91-112, 2016.

M. Lenski and M. A. Scherer, Analysis of synovial inflammatory markers to differ infectious from gouty arthritis, Clin. Biochem, vol.47, pp.49-55, 2014.

N. P. Shanware, Glutamine deprivation stimulates mTOR-JNK-dependent chemokine secretion, Nat. Commun, vol.5, p.4900, 2014.

P. A. Gameiro and K. Struhl, Nutrient deprivation elicits a transcriptional and translational inflammatory response coupled to decreased protein synthesis, Cell Rep, vol.24, pp.1415-1424, 2018.

S. Yoon, NF-?B and STAT3 cooperatively induce IL6 in starved cancer cells, Oncogene, vol.31, pp.3467-3481, 2012.

N. E. Mjiyad, A. Caro-maldonado, S. Ramírez-peinado, and C. Muñoz-pinedo, Sugar-free approaches to cancer cell killing, Oncogene, vol.30, pp.253-264, 2011.

C. Alfaro, Interleukin-8 in cancer pathogenesis, treatment and follow-up, Cancer Treat. Rev, vol.60, pp.24-31, 2017.

H. Xi, M. Kurtoglu, and T. J. Lampidis, The wonders of 2-deoxy-D-glucose, IUBMB Life, vol.66, pp.110-121, 2014.

A. Levy and J. Doyen, Metformin for non-small cell lung cancer patients: Opportunities and pitfalls, Crit. Rev. Oncol. Hematol, vol.125, pp.41-47, 2018.

M. Bergamino, Fasting plasma glucose is an independent predictor of survival in patients with locally advanced non-small cell lung cancer treated with concurrent chemoradiotherapy, BMC Cancer, vol.19, p.165, 2019.

S. Ramírez-peinado, 2-deoxyglucose induces Noxa-dependent apoptosis in alveolar rhabdomyosarcoma, Cancer Res, vol.71, pp.6796-6806, 2011.

X. H. Lowman, The proapoptotic function of Noxa in human leukemia cells is regulated by the kinase Cdk5 and by glucose, Mol. Cell, vol.40, pp.823-833, 2010.

H. Y. Lee, Ca 2+ -dependent demethylation of phosphatase PP2Ac promotes glucose deprivation-induced cell death independently of inhibiting glycolysis, Sci. Signal, vol.11, p.7893, 2018.

C. L. León-annicchiarico, ATF4 mediates necrosis induced by glucose deprivation and apoptosis induced by 2-deoxyglucose in the same cells, FEBS J, vol.282, pp.3647-3658, 2015.

P. S. Gonzalez, Mannose impairs tumour growth and enhances chemotherapy, Nature, vol.563, pp.719-723, 2018.

C. C. Dibble and B. D. Manning, Signal integration by mTORC1 coordinates nutrient input with biosynthetic output, Nat. Cell Biol, vol.15, pp.555-564, 2013.

H. Muaddi, Phosphorylation of eIF2? at serine 51 is an important determinant of cell survival and adaptation to glucose deficiency, Mol. Biol. Cell, vol.21, pp.3220-3231, 2010.

S. Shin, ERK2 mediates metabolic stress response to regulate cell fate, Mol. Cell, vol.59, pp.382-398, 2015.

C. Chaveroux, 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-02379652

J. Obacz, Regulation of tumor-stroma interactions by the unfolded protein response, FEBS J, vol.286, pp.279-296, 2019.
URL : https://hal.archives-ouvertes.fr/hal-01808307

C. Rubio-patiño, Low-protein diet induces IRE1?-dependent anticancer immunosurveillance, Cell Metab, vol.27, pp.828-842, 2018.

J. Obacz, IRE1-UBE2D3 signaling controls the recruitment of myeloid cells to glioblastoma, p.31, 2019.

S. E. Logue, 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

Y. Iwasaki, Activating transcription factor 4 links metabolic stress to interleukin-6 expression in macrophages, Diabetes, vol.63, pp.152-161, 2014.

K. Zhang and R. J. Kaufman, From endoplasmic-reticulum stress to the inflammatory response, Nature, vol.454, pp.455-462, 2008.

K. L. Chong, Human p68 kinase exhibits growth suppression in yeast and homology to the translational regulator GCN2, EMBO J, vol.11, pp.1553-1562, 1992.

C. García-jiménez and C. R. Goding, Starvation and pseudo-starvation as drivers of cancer metastasis through translation reprogramming, Cell Metab, vol.29, pp.254-267, 2019.

N. Sunaga, Oncogenic KRAS-induced interleukin-8 overexpression promotes cell growth and migration and contributes to aggressive phenotypes of non-small cell lung cancer, Int. J. Cancer, vol.130, pp.1733-1744, 2012.

D. E. Johnson, R. A. O'keefe, and J. R. Grandis, Targeting the IL-6/JAK/STAT3 signalling axis in cancer, Nat. Rev. Clin. Oncol, vol.15, pp.234-248, 2018.

K. P. Malabanan, P. Kanellakis, A. Bobik, and L. M. Khachigian, Activation transcription factor-4 induced by fibroblast growth factor-2 regulates vascular endothelial growth factor-A transcription in vascular smooth muscle cells and mediates intimal thickening in rat arteries following balloon injury, Circ. Res, vol.103, pp.378-387, 2008.

V. Bezzerri, Mapping the transcriptional machinery of the IL-8 gene in human bronchial epithelial cells, J. Immunol, vol.187, pp.6069-6081, 2011.

M. A. Reid, Z. Dai, and J. W. Locasale, The impact of cellular metabolism on chromatin dynamics and epigenetics, Nat. Cell Biol, vol.19, pp.1298-1306, 2017.

C. Ecker and J. L. Riley, Translating in vitro T cell metabolic findings to in vivo tumor models of nutrient competition, Cell Metab, vol.28, pp.190-195, 2018.

C. H. Chang, Metabolic competition in the tumor microenvironment is a driver of cancer progression, Cell, vol.162, pp.1229-1241, 2015.

P. Ho, Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses, Cell, vol.162, pp.1217-1228, 2015.

A. Henze and M. Mazzone, The impact of hypoxia on tumor-associated macrophages, J. Clin. Invest, vol.126, pp.3672-3679, 2016.

W. Li, Aerobic glycolysis controls myeloid-derived suppressor cells and tumor immunity via a specific CEBPB isoform in triple-negative breast cancer, Cell Metab, vol.28, pp.87-103, 2018.

J. Li, Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy, Immunity, vol.49, pp.178-193, 2018.