K. L. Abbott, A. V. Nairn, E. M. Hall, M. B. Horton, J. F. Mcdonald et al., Focused glycomic analysis of the N-linked glycan biosynthetic pathway in ovarian cancer, Proteomics, vol.8, issue.16, pp.3210-3220, 2008.

R. L. Aft, F. W. Zhang, and D. Gius, Evaluation of 2-deoxy-D-glucose as a chemotherapeutic agent: Mechanism of cell death, British Journal of Cancer, vol.87, issue.7, pp.805-812, 2002.

L. Agius, E. J. Meredith, and H. S. Sherratt, Stereospecificity of the inhibition by etomoxir of fatty acid and cholesterol synthesis in isolated rat hepatocytes, Biochemical Pharmacology, vol.42, issue.9, pp.1717-1720, 1991.

K. O. Alfarouk, D. Verduzco, C. Rauch, A. K. Muddathir, H. H. Adil et al., Glycolysis, tumor metabolism, cancer growth and dissemination. A new pH-based etiopathogenic perspective and therapeutic approach to an old cancer question, Oncoscience, vol.1, issue.12, pp.777-802, 2014.

K. Alitalo, T. Tammela, and T. V. Petrova, Lymphangiogenesis in development and human disease, Nature, vol.438, issue.7070, pp.946-953, 2005.

H. Allam, K. Aoki, B. B. Benigno, J. F. Mcdonald, S. G. Mackintosh et al., Glycomic analysis of membrane glycoproteins with bisecting glycosylation from ovarian cancer tissues reveals novel structures and functions, Journal of Proteome Research, vol.14, issue.1, pp.434-446, 2015.

H. Allam, B. P. Johnson, M. Zhang, Z. Lu, M. J. Cannon et al., The glycosyltransferase GnT-III activates notch signaling and drives stem cell expansion to promote the growth and invasion of ovarian cancer, The Journal of Biological Chemistry, vol.292, issue.39, pp.16351-16359, 2017.

M. A. Al-rawi and W. G. Jiang, Lymphangiogenesis and cancer metastasis, Front Biosci (Landmark Ed), vol.16, pp.723-739, 2011.

A. Álvarez-aznar, L. Muhl, and K. Gaengel, VEGF receptor tyrosine kinases: Key regulators of vascular function, Current Topics in Developmental Biology, vol.123, pp.433-482, 2017.

R. Andriantsitohaina, L. Duluc, J. C. García-rodríguez, G. Valle, L. Guevara-garcia et al., Systems biology of antioxidants, Clinical Science, vol.123, issue.3, pp.173-192, 2012.

F. Antohe, D. Popov, L. Radulescu, N. Simionescu, T. Börchers et al., Heart microvessels and aortic endothelial cells express the 15 kDa heart-type fatty acid-binding proteins, European Journal of Cell Biology, vol.76, issue.2, pp.102-109, 1998.

X. L. Aranguren, M. Beerens, G. Coppiello, C. Wiese, I. Vandersmissen et al., COUP-TFII orchestrates venous and lymphatic endothelial identity by homoor hetero-dimerisation with PROX1, Journal of Cell Science, vol.126, pp.1164-1175, 2013.

Z. Arany, S. Foo, Y. Ma, J. L. Ruas, A. Bommi-reddy et al., HIF-independent regulation of VEGF and angiogenesis by the transcriptional coactivator PGC-1alpha, Nature, vol.451, issue.7181, pp.1008-1012, 2008.

T. Arao, K. Matsumoto, K. Furuta, K. Kudo, H. Kaneda et al., Acquired drug resistance to vascular endothelial growth factor receptor 2 tyrosine kinase inhibitor in human vascular endothelial cells, Anticancer Research, vol.31, issue.9, pp.2787-2796, 2011.

T. Arcondéguy, E. Lacazette, S. Millevoi, H. Prats, and C. Touriol, VEGF-A mRNA processing, stability and translation: A paradigm for intricate regulation of gene expression at the post-transcriptional level, Nucleic Acids Research, vol.41, issue.17, pp.7997-8010, 2013.

A. W. Armstrong, S. V. Voyles, E. J. Armstrong, E. N. Fuller, and J. C. Rutledge, Angiogenesis and oxidative stress: Common mechanisms linking psoriasis with atherosclerosis, Journal of Dermatological Science, vol.63, issue.1, pp.1-9, 2011.

M. Asada, K. Furukawa, K. Segawa, T. Endo, and A. Kobata, Increased expression of highly branched N-glycans at cell surface is correlated with the malignant phenotypes of mouse tumor cells, Cancer Research, vol.57, issue.6, pp.1073-1080, 1997.

T. Asahara, D. Chen, T. Takahashi, K. Fujikawa, M. Kearney et al., Tie2 receptor ligands, angiopoietin-1 and angiopoietin-2, modulate VEGF-induced postnatal neovascularization, Circulation Research, vol.83, issue.3, pp.233-240, 1998.

A. Aspelund, M. R. Robciuc, S. Karaman, T. Makinen, and K. Alitalo, Lymphatic system in cardiovascular medicine, Circulation Research, vol.118, issue.3, pp.515-530, 2016.

T. Atsumi, J. Chesney, C. Metz, L. Leng, S. Donnelly et al., High expression of inducible 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase, 2002.

, PFKFB3) in human cancers, Cancer Research, vol.62, issue.20, pp.5881-5887

H. G. Augustin, G. Y. Koh, G. Thurston, and K. Alitalo, Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system, Nature Reviews. Molecular Cell Biology, vol.10, issue.3, pp.165-177, 2009.

J. H. Bae, A. R. Hwang, C. Y. Kim, H. G. Yu, H. J. Koh et al., Intravitreal itraconazole inhibits laser-induced choroidal neovascularization in rats, PLoS One, vol.12, issue.6, p.180482, 2017.

P. Baluk, J. Fuxe, H. Hashizume, T. Romano, E. Lashnits et al., Functionally specialized junctions between endothelial cells of lymphatic vessels, The Journal of Experimental Medicine, vol.204, issue.10, pp.2349-2362, 2007.

A. Banerjee, J. Lang, M. Hung, K. Sengupta, S. K. Banerjee et al., Unfolded protein response is required in nu/nu mice microvasculature for treating breast tumor with tunicamycin, The Journal of Biological Chemistry, vol.286, issue.33, pp.29127-29138, 2011.

F. Bard and J. Chia, Cracking the glycome encoder: Signaling, trafficking, and glycosylation, Trends in Cell Biology, vol.26, issue.5, pp.379-388, 2016.

B. Barleon, S. Sozzani, D. Zhou, H. A. Weich, A. Mantovani et al., Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1, Blood, vol.87, issue.8, pp.3336-3343, 1996.

C. Baron-menguy, A. Bocquet, A. Guihot, D. Chappard, M. Amiot et al., Effects of red wine polyphenols on postischemic neovascularization model in rats: Low doses are proangiogenic, high doses anti-angiogenic, The FASEB Journal, vol.21, issue.13, pp.3511-3521, 2007.

E. Barth, G. Stämmler, B. Speiser, and J. Schaper, Ultrastructural quantitation of mitochondria and myofilaments in cardiac muscle from 10 different animal species including man, Journal of Molecular and Cellular Cardiology, vol.24, issue.7, pp.669-681, 1992.

A. Bartolazzi, S. Sciacchitano, and C. &-d'alessandria, Galectin-3: The impact on the clinical management of patients with thyroid nodules and future perspectives, Int J Mol Sci, vol.19, issue.2, 2018.

T. T. Batchelor, E. R. Gerstner, K. E. Emblem, D. G. Duda, J. Kalpathy-cramer et al., Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation, Proc. Natl. Acad. Sci. U.S.A, vol.110, issue.47, pp.19059-19064, 2013.

S. M. Bauer, R. J. Bauer, and O. C. Velazquez, Angiogenesis, vasculogenesis, and induction of healing in chronic wounds, Vascular and Endovascular Surgery, vol.39, issue.4, pp.293-306, 2005.

T. Bäuerle, H. Hilbig, S. Bartling, F. Kiessling, A. Kersten et al., Bevacizumab inhibits breast cancer-induced osteolysis, surrounding soft tissue metastasis, and angiogenesis in rats as visualized by VCT and MRI, Neoplasia, vol.10, issue.5, pp.511-520, 2008.

C. M. Becker and R. J. Amato, Angiogenesis and antiangiogenic therapy in endometriosis, Microvascular Research, vol.74, issue.2-3, pp.121-130, 2007.

R. Benedito, C. Roca, I. Sörensen, S. Adams, A. Gossler et al., The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis, Cell, vol.137, issue.6, pp.1124-1135, 2009.

G. Bergers and D. Hanahan, Modes of resistance to anti-angiogenic therapy, Nature Reviews. Cancer, vol.8, issue.8, pp.592-603, 2008.

C. R. Berkers, O. D. Maddocks, E. C. Cheung, I. Mor, and K. H. Vousden, Metabolic regulation by p53 family members, Cell Metabolism, vol.18, issue.5, pp.617-633, 2013.

M. Berretta, L. Rinaldi, F. Di-benedetto, A. Lleshi, V. De-re et al., Angiogenesis inhibitors for the treatment of hepatocellular carcinoma, Frontiers in Pharmacology, vol.7, p.428, 2016.

S. V. Bhagwat, P. C. Gokhale, A. P. Crew, A. Cooke, Y. Yao et al., Preclinical characterization of OSI-027, a potent and selective inhibitor of mTORC1 and mTORC2: Distinct from rapamycin, Molecular Cancer Therapeutics, vol.10, issue.8, pp.1394-1406, 2011.

M. A. Björndahl, R. Cao, J. B. Burton, E. Brakenhielm, P. Religa et al., Vascular endothelial growth factor-a promotes peritumoral lymphangiogenesis and lymphatic metastasis, Cancer Research, vol.65, issue.20, pp.9261-9268, 2005.

A. Blouin, R. P. Bolender, and E. R. Weibel, Distribution of organelles and membranes between hepatocytes and nonhepatocytes in the rat liver parenchyma. A stereological study, The Journal of Cell Biology, vol.72, issue.2, pp.441-455, 1977.

J. Bonzi, O. Bornet, S. Betzi, B. T. Kasper, L. K. Mahal et al., Pre-B cell receptor binding to galectin-1 modifies galectin-1/carbohydrate affinity to modulate specific galectin-1/glycan lattice interactions, Nature Communications, vol.6, p.6194, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02018118

C. Boscher, J. W. Dennis, and I. R. Nabi, Glycosylation, galectins and cellular signaling, Current Opinion in Cell Biology, vol.23, issue.4, pp.383-392, 2011.

C. Boscher, Y. Z. Zheng, R. Lakshminarayan, L. Johannes, J. W. Dennis et al., Galectin-3 protein regulates mobility of N-cadherin and GM1 ganglioside at cell-cell junctions of mammary carcinoma cells, The Journal of Biological Chemistry, vol.287, issue.39, pp.32940-32952, 2012.

I. Bougioukas, V. Didilis, P. Ypsilantis, A. Giatromanolaki, E. Sivridis et al., Intramyocardial injection of low-dose basic fibroblast growth factor or vascular endothelial growth factor induces angiogenesis in the infarcted rabbit myocardium, Cardiovascular Pathology, vol.16, issue.2, pp.63-68, 2007.

M. Boulton, A role for hepatocyte growth factor in diabetic retinopathy?, The British Journal of Ophthalmology, vol.83, issue.7, pp.763-764, 1999.

S. Brem, H. Brem, J. Folkman, D. Finkelstein, and A. Patz, Prolonged tumor dormancy by prevention of neovascularization in the vitreous, Cancer Research, vol.36, issue.8, pp.2807-2812, 1976.

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, Proceedings of the National Academy of Sciences of the United States of America, vol.96, issue.15, pp.8505-8510, 1999.

C. D. Browne, E. J. Hindmarsh, and J. W. Smith, Inhibition of endothelial cell proliferation and angiogenesis by orlistat, a fatty acid synthase inhibitor, The FASEB Journal, vol.20, issue.12, pp.2027-2035, 2006.

S. Browne and A. Pandit, Engineered systems for therapeutic angiogenesis, Current Opinion in Pharmacology, vol.36, pp.34-43, 2017.

P. H. Burri and M. R. Tarek, A novel mechanism of capillary growth in the rat pulmonary microcirculation, The Anatomical Record, vol.228, issue.1, pp.35-45, 1990.

J. H. Caduff, L. C. Fischer, and P. H. Burri, Scanning electron microscope study of the developing microvasculature in the postnatal rat lung, The Anatomical Record, vol.216, issue.2, pp.154-164, 1986.

L. Cai, Z. Gu, J. Zhong, D. Wen, G. Chen et al., Advances in glycosylationmediated cancer-targeted drug delivery, Drug Discovery Today, pp.30242-30248, 2018.

S. Caja and J. A. Enríquez, Mitochondria in endothelial cells: Sensors and integrators of environmental cues, Redox Biology, vol.12, pp.821-827, 2017.

C. Camaré, M. Pucelle, A. Nègre-salvayre, and R. Salvayre, Angiogenesis in the atherosclerotic plaque, Redox Biology, vol.12, pp.18-34, 2017.

I. Camby, N. Belot, F. Lefranc, N. Sadeghi, Y. De-launoit et al., Galectin-1 modulates human glioblastoma cell migration into the brain through modifications to the actin cytoskeleton and levels of expression of small GTPases, Neuropathol. Exp. Neurol, vol.61, issue.7, pp.585-596, 2002.

I. Camby, N. Belot, S. Rorive, F. Lefranc, C. A. Maurage et al., Galectins are differentially expressed in supratentorial pilocytic astrocytomas, astrocytomas, anaplastic astrocytomas and glioblastomas, and significantly modulate tumor astrocyte migration, Brain Pathology, vol.11, issue.1, pp.12-26, 2001.

I. Camby, M. Le-mercier, F. Lefranc, and R. Kiss, Galectin-1: A small protein with major functions, Glycobiology, vol.16, issue.11, pp.137-157, 2006.

P. A. Campochiaro, R. Sophie, M. Tolentino, D. M. Miller, D. Browning et al., Treatment of diabetic macular edema with an inhibitor of vascular endothelial-protein tyrosine phosphatase that activates Tie2, Ophthalmology, vol.122, issue.3, pp.545-554, 2015.

A. R. Cantelmo, L. Conradi, A. Brajic, J. Goveia, J. Kalucka et al., Inhibition of the glycolytic activator PFKFB3 in endothelium induces tumor vessel normalization, impairs metastasis, and improves chemotherapy, Cancer Cell, vol.30, issue.6, pp.968-985, 2016.

R. Cao, M. A. Björndahl, P. Religa, S. Clasper, S. Garvin et al., PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis, Cancer Cell, vol.6, issue.4, pp.333-345, 2004.

M. Capitão and R. Soares, Angiogenesis and inflammation crosstalk in diabetic retinopathy, Journal of Cellular Biochemistry, vol.117, issue.11, pp.2443-2453, 2016.

P. Carmeliet, Angiogenesis in health and disease, Nature Medicine, vol.9, issue.6, pp.653-660, 2003.

P. Carmeliet and R. K. Jain, Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases, Nature Reviews. Drug Discovery, vol.10, issue.6, pp.417-427, 2011.

S. Cataltepe, M. C. Arikan, X. Liang, T. W. Smith, and O. Cataltepe, Fatty acid binding protein 4 expression in cerebral vascular malformations: Implications for vascular remodelling, Neuropathology and Applied Neurobiology, vol.41, issue.5, pp.646-656, 2015.

M. Caunt, J. Mak, W. Liang, S. Stawicki, Q. Pan et al., Blocking neuropilin-2 function inhibits tumor cell metastasis, Cancer Cell, vol.13, issue.4, pp.331-342, 2008.

J. P. Cerliani, A. G. Blidner, M. A. Toscano, D. O. Croci, and G. A. Rabinovich, Translating the "sugar code" into immune and vascular signaling programs, Trends Biochem. Sci, vol.42, issue.4, pp.255-273, 2017.

K. B. Chandler, D. R. Leon, R. D. Meyer, N. Rahimi, and C. E. Costello, Site-specific Nglycosylation of endothelial cell receptor tyrosine kinase VEGFR-2, Journal of Proteome Research, vol.16, issue.2, pp.677-688, 2017.

. Chao-de-la, J. M. Barca, D. Prunier-mirebeau, P. Amati-bonneau, M. Ferré et al., OPA1-related disorders: Diversity of clinical expression, modes of inheritance and pathophysiology, Neurobiology of Disease, vol.90, pp.20-26, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01392230

J. C. Chappell, S. M. Taylor, N. Ferrara, and V. L. Bautch, Local guidance of emerging vessel sprouts requires soluble Flt-1, Developmental Cell, vol.17, issue.3, pp.377-386, 2009.

S. S. Chaurasia, R. R. Lim, B. H. Parikh, Y. S. Wey, B. B. Tun et al., The NLRP3 inflammasome may contribute to pathologic neovascularization in the advanced stages of diabetic retinopathy, Scientific Reports, vol.8, issue.1, p.2847, 2018.

R. Chebib, L. Verlingue, N. Cozic, M. Faron, P. Burtin et al., Angiogenesis inhibition in the second-line treatment of metastatic colorectal cancer: A systematic review and pooled analysis, Seminars in Oncology, vol.44, issue.2, pp.114-128, 2017.

H. Chen and D. C. Chan, Emerging functions of mammalian mitochondrial fusion and fission, Hum. Mol. Genet, vol.14, issue.2, pp.283-289, 2005.

L. Chen, W. Zhang, N. Fregien, and M. Pierce, The her-2/neu oncogene stimulates the transcription of N-acetylglucosaminyltransferase V and expression of its cell surface oligosaccharide products, Oncogene, vol.17, issue.16, pp.2087-2093, 1998.

W. Chen, Z. Cao, S. Sugaya, M. J. Lopez, V. G. Sendra et al., Pathological lymphangiogenesis is modulated by galectin-8-dependent crosstalk between podoplanin and integrin-associated VEGFR-3, Nature Communications, vol.7, p.11302, 2016.

Z. Chen, M. L. Varney, M. W. Backora, K. Cowan, J. C. Solheim et al., Down-regulation of vascular endothelial cell growth factor-C expression using small interfering RNA vectors in mammary tumors inhibits tumor lymphangiogenesis and spontaneous metastasis and enhances survival, Cancer Research, vol.65, issue.19, pp.9004-9011, 2005.

D. W. Cheng, Y. Jiang, A. Shalev, R. Kowluru, E. D. Crook et al., An analysis of high glucose and glucosamine-induced gene expression and oxidative stress in renal mesangial cells, Archives of Physiology and Biochemistry, vol.112, issue.4-5, pp.189-218, 2006.

J. 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, Molecular Cancer Therapeutics, vol.10, issue.12, pp.2350-2362, 2011.

J. Chesney, R. Mitchell, F. Benigni, M. Bacher, L. Spiegel et al., An inducible gene product for 6-phosphofructo-2-kinase with an AU-rich instability element: Role in tumor cell glycolysis and the Warburg effect, Proceedings of the National Academy of Sciences of the United States of America, vol.96, issue.6, pp.3047-3052, 1999.

J. Chesney, S. Telang, A. Yalcin, A. Clem, N. Wallis et al., Targeted disruption of inducible 6-phosphofructo-2-kinase results in embryonic lethality, Biochemical and Biophysical Research Communications, vol.331, issue.1, pp.139-146, 2005.

C. C. Chua, R. C. Hamdy, and B. H. Chua, Upregulation of vascular endothelial growth factor by H2O2 in rat heart endothelial cells, Free Radical Biology & Medicine, vol.25, issue.8, pp.891-897, 1998.

A. S. Chung and N. Ferrara, Developmental and pathological angiogenesis, Annual Review of Cell and Developmental Biology, vol.27, pp.563-584, 2011.

C. Chung and N. Pherwani, Ziv-aflibercept: A novel angiogenesis inhibitor for the treatment of metastatic colorectal cancer, American Journal of Health-System Pharmacy, vol.70, issue.21, pp.1887-1896, 2013.

H. Clausen and E. P. Bennett, A family of UDP-GalNAc: Polypeptide Nacetylgalactosaminyl-transferases control the initiation of mucin-type O-linked glycosylation, Glycobiology, vol.6, issue.6, pp.635-646, 1996.

B. Clem, S. Telang, A. Clem, A. Yalcin, J. Meier et al., Small-molecule inhibition of 6-phosphofructo-2-kinase activity suppresses glycolytic flux and tumor growth, Molecular Cancer Therapeutics, vol.7, issue.1, pp.110-120, 2008.

B. F. Clem, J. O'neal, A. C. Klarer, S. Telang, and J. Chesney, Clinical development of cancer therapeutics that target metabolism, QJM, vol.6, pp.367-372, 2016.

B. F. Clem, J. O'neal, G. Tapolsky, A. L. Clem, Y. Imbert-fernandez et al., Targeting 6-phosphofructo-2-kinase (PFKFB3) as a therapeutic strategy against cancer, Molecular Cancer Therapeutics, vol.12, issue.8, pp.1461-1470, 2013.

E. Clementi, G. C. Brown, N. Foxwell, and S. Moncada, On the mechanism by which vascular endothelial cells regulate their oxygen consumption, Proc. Natl. Acad. Sci. U.S. A, vol.96, issue.4, pp.1559-1562, 1999.

N. Clere, L. Bermont, S. Fauconnet, I. Lascombe, M. Saunier et al., The human papillomavirus type 18 E6 oncoprotein induces Vascular Endothelial Growth Factor 121 (VEGF121) transcription from the promoter through a p53-independent mechanism, Experimental Cell Research, vol.313, issue.15, pp.3239-3250, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00484927

N. Clere, S. Faure, M. C. Martinez, and R. Andriantsitohaina, Anticancer properties of flavonoids: roles in various stages of carcinogenesis, Cardiovascular & Hematological Agents in Medicinal Chemistry, vol.9, issue.2, pp.62-77, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01390557

C. L. Cole, G. Rushton, G. C. Jayson, and E. Avizienyte, Ovarian cancer cell heparan sulfate 6-O-sulfotransferases regulate an angiogenic program induced by heparinbinding epidermal growth factor (EGF)-like growth factor/EGF receptor signaling, The Journal of Biological Chemistry, vol.289, issue.15, pp.10488-10501, 2014.

E. S. Colombo, G. Menicucci, P. G. Mcguire, and A. Das, Hepatocyte growth factor/ scatter factor promotes retinal angiogenesis through increased urokinase expression, Investigative Ophthalmology & Visual Science, vol.48, issue.4, pp.1793-1800, 2007.

L. C. Conradi, A. Brajic, A. R. Cantelmo, A. Bouché, J. Kalucka et al., Tumor vessel disintegration by maximum tolerable PFKFB3 blockade, Angiogenesis, vol.20, issue.4, pp.599-613, 2017.

G. Coppiello, M. Collantes, M. S. Sirerol-piquer, S. Vandenwijngaert, S. Schoors et al., Meox2/Tcf15 heterodimers program the heart capillary endothelium for cardiac fatty acid uptake, Circulation, vol.131, issue.9, pp.815-826, 2015.

T. Couffinhal, P. Dufourcq, D. Daret, and C. Duplaà, The mechanisms of angiogenesis. Medical and therapeutic applications, La Revue de Médecine Interne, vol.22, issue.11, pp.1064-1082, 2001.

O. Coutelle, H. Hornig-do, A. Witt, M. Andree, L. M. Schiffmann et al., Embelin inhibits endothelial mitochondrial respiration and impairs neoangiogenesis during tumor growth and wound healing, EMBO Molecular Medicine, vol.6, issue.5, pp.624-639, 2014.

D. O. Croci, J. P. Cerliani, T. Dalotto-moreno, S. P. Méndez-huergo, I. D. Mascanfroni et al., Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors, Cell, vol.156, issue.4, pp.744-758, 2014.

D. O. Croci, J. P. Cerliani, N. A. Pinto, L. G. Morosi, and G. A. Rabinovich, Regulatory role of glycans in the control of hypoxia-driven angiogenesis and sensitivity to antiangiogenic treatment, Glycobiology, vol.24, issue.12, pp.1283-1290, 2014.

D. O. Croci, M. Salatino, N. Rubinstein, J. P. Cerliani, L. E. Cavallin et al., Disrupting galectin-1 interactions with N-glycans suppresses hypoxia-driven angiogenesis and tumorigenesis in Kaposi's sarcoma, The Journal of Experimental Medicine, vol.209, issue.11, pp.1985-2000, 2012.

A. Csiszar, N. Labinskyy, J. T. Pinto, P. Ballabh, H. Zhang et al., Resveratrol induces mitochondrial biogenesis in endothelial cells, American Journal of Physiology. Heart and Circulatory Physiology, vol.297, issue.1, pp.13-20, 2009.

L. N. Cueni, L. Chen, H. Zhang, D. Marino, R. Huggenberger et al., Podoplanin-Fc reduces lymphatic vessel formation in vitro and in vivo and causes disseminated intravascular coagulation when transgenically expressed in the skin, Blood, vol.116, issue.20, pp.4376-4384, 2010.

O. Culic, M. L. Gruwel, and J. Schrader, Energy turnover of vascular endothelial cells, The American Journal of Physiology, vol.273, issue.1, pp.205-213, 1997.

C. Cursiefen, L. Chen, L. P. Borges, D. Jackson, J. Cao et al., VEGF-A stimulates lymphangiogenesis and hemangiogenesis in inflammatory neovascularization via macrophage recruitment, The Journal of Clinical Investigation, vol.113, issue.7, pp.1040-1050, 2004.

M. P. Dale, H. E. Ensley, K. Kern, K. A. Sastry, and L. D. Byers, Reversible inhibitors of beta-glucosidase, Biochemistry, vol.24, issue.14, pp.3530-3539, 1985.

S. Das, E. Sarrou, S. Podgrabinska, M. Cassella, S. K. Mungamuri et al., Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses, The Journal of Experimental Medicine, vol.210, issue.8, pp.1509-1528, 2013.

A. Dasgupta, M. Trucco, N. Rainusso, R. J. Bernardi, R. Shuck et al., Metabolic modulation of Ewing sarcoma cells inhibits tumor growth and stem cell properties, Oncotarget, vol.8, issue.44, pp.77292-77308, 2017.

D. Bock, K. Georgiadou, M. Carmeliet, and P. , Role of endothelial cell metabolism in vessel sprouting, Cell Metabolism, vol.18, issue.5, pp.634-647, 2013.

D. Bock, K. Georgiadou, M. Schoors, S. Kuchnio, A. Wong et al., Role of PFKFB3-driven glycolysis in vessel sprouting, Cell, vol.154, issue.3, pp.651-663, 2013.

M. Demetriou, M. Granovsky, S. Quaggin, and J. W. Dennis, Negative regulation of T-cell activation and autoimmunity by Mgat5 N-glycosylation, Nature, vol.409, issue.6821, pp.733-739, 2001.

M. Demetriou, I. R. Nabi, M. Coppolino, S. Dedhar, and J. W. Dennis, Reduced contact-inhibition and substratum adhesion in epithelial cells expressing GlcNAc-transferase V, The Journal of Cell Biology, vol.130, issue.2, pp.383-392, 1995.

D. Demydenko and I. Berest, Expression of galectin-1 in malignant tumors, Experimental Oncology, vol.31, issue.2, pp.74-79, 2009.

Q. Deng, Y. Chen, N. Yin, N. Shan, X. Luo et al., Nacetylglucosaminyltransferase V inhibits the invasion of trophoblast cells by attenuating MMP2/9 activity in early human pregnancy, Placenta, vol.36, issue.11, pp.1291-1299, 2015.

Q. Deng, Y. Chen, N. Yin, N. Shan, X. Luo et al., The role of MGAT5 in human umbilical vein endothelial cells, Reproductive Sciences, vol.24, issue.2, pp.313-323, 2017.

R. M. Denton and J. G. Mccormack, Ca2+ as a second messenger within mitochondria of the heart and other tissues, Annual Review of Physiology, vol.52, pp.451-466, 1990.

N. D'haene, S. Sauvage, C. Maris, I. Adanja, M. Le-mercier et al., VEGFR1 and VEGFR2 involvement in extracellular galectin-1-and galectin-3-induced angiogenesis, Adv Drug Deliv Rev, vol.8, issue.6, pp.148-160, 2013.

S. Dimmeler, I. Fleming, B. Fisslthaler, C. Hermann, R. Busse et al., Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation, Nature, vol.399, issue.6736, pp.601-605, 1999.

S. Dimmeler and A. M. Zeiher, Endothelial cell apoptosis in angiogenesis and vessel regression, Circulation Research, vol.87, issue.6, pp.434-439, 2000.

I. Dimova, G. Popivanov, and V. Djonov, Angiogenesis in cancer -general pathways and their therapeutic implications, Journal of BUON, vol.19, issue.1, pp.15-21, 2014.

R. P. Dings, M. Loren, H. Heun, E. Mcniel, A. W. Griffioen et al., Scheduling of radiation with angiogenesis inhibitors anginex and Avastin improves therapeutic outcome via vessel normalization, Clinical Cancer Research, vol.13, issue.11, pp.3395-3402, 2007.

V. Djonov, O. Baum, and P. H. Burri, Vascular remodeling by intussusceptive angiogenesis, Cell and Tissue Research, vol.314, issue.1, pp.107-117, 2003.

V. Djonov, M. Schmid, S. A. Tschanz, and P. H. Burri, Intussusceptive angiogenesis: Its role in embryonic vascular network formation, Circulation Research, vol.86, issue.3, pp.286-292, 2000.

B. C. Dobner, A. I. Riechardt, A. M. Joussen, S. Englert, and N. E. Bechrakis, Expression of haematogenous and lymphogenous chemokine receptors and their ligands on uveal melanoma in association with liver metastasis, Acta Ophthalmologica, vol.90, issue.8, pp.638-644, 2012.

A. Dobrina and F. Rossi, Metabolic properties of freshly isolated bovine endothelial cells, Biochimica et Biophysica Acta, vol.762, issue.2, pp.295-301, 1983.

A. Dondoni, H. M. Zuurmond, and A. Boscarato, Synthesis of alpha-and beta-D-(1?6)-c-disaccharides by wittig olefination of formyl c-glycosides with glycopyranose 6-phosphoranes, The Journal of Organic Chemistry, vol.62, issue.23, pp.8114-8124, 1997.

D. F. Dong, E. X. Li, J. B. Wang, Y. Y. Wu, F. Shi et al., Anti-angiogenesis and anti-tumor effects of AdNT4-anginex, Cancer Letters, vol.285, issue.2, pp.218-224, 2009.

L. Dong, V. J. Jameson, D. Tilly, J. Cerny, E. Mahdavian et al., Mitochondrial targeting of vitamin E succinate enhances its pro-apoptotic and anti-cancer activity via mitochondrial complex II, The Journal of Biological Chemistry, vol.286, issue.5, pp.3717-3728, 2011.

L. Dong, P. Low, J. C. Dyason, X. Wang, L. Prochazka et al., Alpha-tocopheryl succinate induces apoptosis by targeting ubiquinone-binding sites in mitochondrial respiratory complex II, Oncogene, vol.27, issue.31, pp.4324-4335, 2008.

L. Dong, E. Swettenham, J. Eliasson, X. Wang, M. Gold et al., , 2007.

, Vitamin E analogues inhibit angiogenesis by selective induction of apoptosis in proliferating endothelial cells: The role of oxidative stress, Cancer Research, vol.67, issue.24, pp.11906-11913

D. Santos, S. N. Sheldon, H. Pereira, J. X. Paluch, C. Bridges et al., Galectin-3 acts as an angiogenic switch to induce tumor angiogenesis via Jagged-1/Notch activation, Oncotarget, vol.8, issue.30, pp.49484-49501, 2017.

H. Dosaka-akita, E. Miyoshi, O. Suzuki, T. Itoh, H. Katoh et al., Expression of N-acetylglucosaminyltransferase v is associated with prognosis and histology in non-small cell lung cancers, Clinical Cancer Research, vol.10, issue.5, pp.1773-1779, 2004.

M. A. Doucey, D. Hess, M. J. Blommers, and J. Hofsteenge, Recombinant human interleukin-12 is the second example of a C-mannosylated protein, Glycobiology, vol.9, issue.5, pp.435-441, 1999.

M. A. Doucey, D. Hess, R. Cacan, and J. Hofsteenge, Protein C-mannosylation is enzyme-catalysed and uses dolichyl-phosphate-mannose as a precursor, Molecular Biology of the Cell, vol.9, issue.2, pp.291-300, 1998.

B. P. Dranka, B. G. Hill, and V. M. Darley-usmar, Mitochondrial reserve capacity in endothelial cells: The impact of nitric oxide and reactive oxygen species, Free Radical Biology & Medicine, vol.48, issue.7, pp.905-914, 2010.

N. Draoui, P. De-zeeuw, and P. Carmeliet, Angiogenesis revisited from a metabolic perspective: Role and therapeutic implications of endothelial cell metabolism, Open Biol, vol.7, issue.12, 2017.

L. Duluc, C. Jacques, R. Soleti, R. Andriantsitohaina, and G. Simard, Delphinidin inhibits VEGF induced-mitochondrial biogenesis and Akt activation in endothelial cells, The International Journal of Biochemistry & Cell Biology, vol.53, pp.9-14, 2014.

L. Duluc, C. Jacques, R. Soleti, F. Iacobazzi, G. Simard et al., Modulation of mitochondrial capacity and angiogenesis by red wine polyphenols via estrogen receptor, NADPH oxidase and nitric oxide synthase pathways, The International Journal of Biochemistry & Cell Biology, vol.45, issue.4, pp.783-791, 2013.

J. M. Dunleavey and A. C. Dudley, Vascular mimicry: Concepts and implications for anti-angiogenic therapy, Curr Angiogenes, vol.1, issue.2, pp.133-138, 2012.

T. Duong, P. Koopman, and M. Francois, Tumor lymphangiogenesis as a potential therapeutic target, Journal of Oncology, 2012.

M. Duval, F. Le-boeuf, J. Huot, and J. Gratton, Src-mediated phosphorylation of Hsp90 in response to vascular endothelial growth factor (VEGF) is required for VEGF receptor-2 signaling to endothelial NO synthase, Molecular Biology of the Cell, vol.18, issue.11, pp.4659-4668, 2007.

H. F. Dvorak, Rous-Whipple award lecture. How tumors make bad blood vessels and stroma, The American Journal of Pathology, vol.162, issue.6, pp.1747-1757, 2003.

H. F. Dvorak, Tumor stroma, tumor blood vessels, and antiangiogenesis therapy, Cancer Journal, vol.21, issue.4, pp.237-243, 2015.

J. M. Ebos, G. Bocci, S. Man, P. E. Thorpe, D. J. Hicklin et al., A naturally occurring soluble form of vascular endothelial growth factor receptor 2 detected in mouse and human plasma, Molecular Cancer Research, vol.2, issue.6, pp.315-326, 2004.

G. Eelen, P. De-zeeuw, M. Simons, and P. Carmeliet, Endothelial cell metabolism in normal and diseased vasculature, Circulation Research, vol.116, issue.7, pp.1231-1244, 2015.

G. Eelen, P. De-zeeuw, L. Treps, U. Harjes, B. W. Wong et al., Endothelial cell metabolism, Physiological Reviews, vol.98, issue.1, pp.3-58, 2018.

S. Egginton, A. L. Zhou, M. D. Brown, and O. Hudlická, Unorthodox angiogenesis in skeletal muscle, Cardiovascular Research, vol.49, issue.3, pp.634-646, 2001.

R. Ehehalt, R. Sparla, H. Kulaksiz, T. Herrmann, J. Füllekrug et al., Uptake of long chain fatty acids is regulated by dynamic interaction of FAT/CD36 with cholesterol/sphingolipid enriched microdomains (lipid rafts), BMC Cell Biology, vol.9, p.45, 2008.

H. Elmasri, E. Ghelfi, C. Yu, S. Traphagen, M. Cernadas et al., Endothelial cell-fatty acid binding protein 4 promotes angiogenesis: Role of stem cell factor/c-kit pathway, Angiogenesis, vol.15, issue.3, pp.457-468, 2012.

H. Elmasri, C. Karaaslan, Y. Teper, E. Ghelfi, M. Weng et al., Fatty acid binding protein 4 is a target of VEGF and a regulator of cell proliferation in endothelial cells, The FASEB Journal, vol.23, issue.11, pp.3865-3873, 2009.

M. T. Elola, A. G. Blidner, F. Ferragut, C. Bracalente, and G. A. Rabinovich, Assembly, organization and regulation of cell-surface receptors by lectin-glycan complexes, The Biochemical Journal, vol.469, issue.1, pp.1-16, 2015.

G. Endemann, L. W. Stanton, K. S. Madden, C. M. Bryant, R. T. White et al., CD36 is a receptor for oxidized low density lipoprotein, The Journal of Biological Chemistry, vol.268, issue.16, pp.11811-11816, 1993.

E. Fagiani, P. Lorentz, L. Kopfstein, and G. Christofori, Angiopoietin-1 and -2 exert antagonistic functions in tumor angiogenesis, yet both induce lymphangiogenesis, Cancer Research, vol.71, issue.17, pp.5717-5727, 2011.

G. S. Falchook, S. L. Moulder, J. J. Wheler, Y. Jiang, C. C. Bastida et al., Dual HER2 inhibition in combination with anti-VEGF treatment is active in heavily pretreated HER2-positive breast cancer, Annals of Oncology, vol.24, issue.12, pp.3004-3011, 2013.

B. L. Falcon, S. Barr, P. C. Gokhale, J. Chou, J. Fogarty et al., Reduced VEGF production, angiogenesis, and vascular regrowth contribute to the antitumor properties of dual mTORC1/mTORC2 inhibitors, Cancer Research, vol.71, issue.5, pp.1573-1583, 2011.

R. H. Farnsworth, M. G. Achen, and S. A. Stacker, Lymphatic endothelium: An important interactive surface for malignant cells, Pulmonary Pharmacology & Therapeutics, vol.19, issue.1, pp.51-60, 2006.

L. Favot, S. Martin, T. Keravis, R. Andriantsitohaina, and C. Lugnier, Involvement of cyclin-dependent pathway in the inhibitory effect of delphinidin on angiogenesis, Cardiovascular Research, vol.59, issue.2, pp.479-487, 2003.

N. Ferrara, Vascular endothelial growth factor: Basic science and clinical progress, Endocrine Reviews, vol.25, issue.4, pp.581-611, 2004.

R. Flavin, S. Peluso, P. L. Nguyen, and M. Loda, Fatty acid synthase as a potential therapeutic target in cancer, Future Oncology, vol.6, issue.4, pp.551-562, 2010.

M. François, A. Caprini, B. Hosking, F. Orsenigo, D. Wilhelm et al., Sox18 induces development of the lymphatic vasculature in mice, Nature, vol.456, issue.7222, pp.643-647, 2008.

M. Francois, N. L. Harvey, and B. M. Hogan, The transcriptional control of lymphatic vascular development, Physiology (Bethesda), vol.26, issue.3, pp.146-155, 2011.

J. Fu, H. Gerhardt, J. M. Mcdaniel, B. Xia, X. Liu et al., Endothelial cell O-glycan deficiency causes blood/lymphatic misconnections and consequent fatty liver disease in mice, The Journal of Clinical Investigation, vol.118, issue.11, pp.3725-3737, 2008.

M. Fukasawa, T. Tsuchiya, E. Takayama, N. Shinomiya, K. Uyeda et al., Identification and characterization of the hypoxia-responsive element of the human placental 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase gene, Journal of Biochemistry, vol.136, issue.3, pp.273-277, 2004.

D. Fukumura, S. Kashiwagi, and R. K. Jain, The role of nitric oxide in tumour progression, Nature Reviews. Cancer, vol.6, issue.7, pp.521-534, 2006.

S. Fukunaga, K. Maeda, E. Noda, T. Inoue, K. Wada et al., Association between expression of vascular endothelial growth factor C, chemokine receptor CXCR4 and lymph node metastasis in colorectal cancer, Oncology, vol.71, issue.3-4, pp.204-211, 2006.

D. Fulton, J. P. Gratton, T. J. Mccabe, J. Fontana, Y. Fujio et al., Regulation of endothelium-derived nitric oxide production by the protein kinase Akt, Nature, vol.399, issue.6736, pp.597-601, 1999.

D. Fulton, L. Ruan, S. G. Sood, C. Li, Q. Zhang et al., Agonist-stimulated endothelial nitric oxide synthase activation and vascular relaxation. Role of eNOS phosphorylation at Tyr83, Circulation Research, vol.102, issue.4, pp.497-504, 2008.

T. Funasaka, A. Raz, and P. Nangia-makker, Galectin-3 in angiogenesis and metastasis, Glycobiology, vol.24, issue.10, pp.886-891, 2014.

F. Galland, A. Karamysheva, M. J. Pebusque, J. P. Borg, R. Rottapel et al., The FLT4 gene encodes a transmembrane tyrosine kinase related to the vascular endothelial growth factor receptor, Oncogene, vol.8, issue.5, pp.1233-1240, 1993.

L. S. Gambino, N. G. Wreford, J. F. Bertram, P. Dockery, F. Lederman et al., Angiogenesis occurs by vessel elongation in proliferative phase human endometrium, Human Reproduction, vol.17, issue.5, pp.1199-1206, 2002.

G. Giannone, P. Rondé, M. Gaire, J. Beaudouin, J. Haiech et al., Calcium rises locally trigger focal adhesion disassembly and enhance residency of focal adhesion kinase at focal adhesions, The Journal of Biological Chemistry, vol.279, issue.27, pp.28715-28723, 2004.

J. F. Glatz and J. J. Luiken, From fat to FAT (CD36/SR-B2): Understanding the regulation of cellular fatty acid uptake, Biochimie, vol.136, pp.21-26, 2017.

L. Goldberg, R. Israeli, and Y. Kloog, FTS and 2-DG induce pancreatic cancer cell death and tumor shrinkage in mice, Cell Death & Disease, vol.3, p.284, 2012.

F. G. Gomes, F. Nedel, A. M. Alves, J. E. Nör, and S. B. Tarquinio, Tumor angiogenesis and lymphangiogenesis: Tumor/endothelial crosstalk and cellular/microenvironmental signaling mechanisms, Life Sciences, vol.92, issue.2, pp.101-107, 2013.

R. Gómez, A. Abad, F. Delgado, S. Tamarit, C. Simón et al., Effects of hyperprolactinemia treatment with the dopamine agonist quinagolide on endometriotic lesions in patients with endometriosis-associated hyperprolactinemia, Fertil Steril, vol.95, issue.3, pp.882-888, 2011.

Y. Gong, H. Lan, Z. Yu, M. Wang, S. Wang et al., Blockage of glycolysis by targeting PFKFB3 alleviates sepsis-related acute lung injury via suppressing inflammation and apoptosis of alveolar epithelial cells, Biochemical and Biophysical Research Communications, vol.491, issue.2, pp.522-529, 2017.

C. Granchi and F. Minutolo, Anticancer agents that counteract tumor glycolysis, ChemMedChem, vol.7, issue.8, pp.1318-1350, 2012.

M. Granovsky, J. Fata, J. Pawling, W. J. Muller, R. Khokha et al., Suppression of tumor growth and metastasis in Mgat5-deficient mice, Nature Medicine, vol.6, issue.3, pp.306-312, 2000.

D. R. Green, Apoptotic pathways: The roads to ruin, Cell, vol.94, issue.6, pp.695-698, 1998.

D. E. Greenwalt, K. W. Watt, T. Hasler, R. J. Howard, and S. Patel, Structural, functional, and antigenic differences between bovine heart endothelial CD36 and human platelet CD36, The Journal of Biological Chemistry, vol.265, issue.27, pp.16296-16299, 1990.

A. W. Griffioen and V. L. Thijssen, Galectins in tumor angiogenesis, Ann Transl Med, vol.2, issue.9, p.90, 2014.

J. P. Groningen, A. C. Wenink, and L. H. Testers, Myocardial capillaries: Increase in number by splitting of existing vessels, Anatomy and Embryology, vol.184, issue.1, pp.65-70, 1991.

L. N. Groschner, M. Waldeck-weiermair, R. Malli, and W. F. Graier, Endothelial mitochondria-less respiration, more integration, Pflügers Archiv, vol.464, issue.1, pp.63-76, 2012.

J. Gu and N. Taniguchi, Potential of N-glycan in cell adhesion and migration as either a positive or negative regulator, Cell Adhesion & Migration, vol.2, issue.4, pp.243-245, 2008.

H. Guo, I. Lee, M. Kamar, S. K. Akiyama, and M. Pierce, Aberrant N-glycosylation of beta1 integrin causes reduced alpha5beta1 integrin clustering and stimulates cell migration, Cancer Research, vol.62, issue.23, pp.6837-6845, 2002.

S. Guo and L. A. Dipietro, Factors affecting wound healing, Journal of Dental Research, vol.89, issue.3, pp.219-229, 2010.

S. A. Hafez, T. Caceci, L. E. Freeman, and K. E. Panter, Angiogenesis in the caprine caruncles in non-pregnant and pregnant normal and swainsonine-treated does, Anat Rec (Hoboken), vol.290, issue.7, pp.761-769, 2007.

C. Hagberg, A. Mehlem, A. Falkevall, L. Muhl, and U. Eriksson, Endothelial fatty acid transport: Role of vascular endothelial growth factor B, Physiology (Bethesda), vol.28, issue.2, pp.125-134, 2013.

C. E. Hagberg, A. Falkevall, X. Wang, E. Larsson, J. Huusko et al., Vascular endothelial growth factor B controls endothelial fatty acid uptake, Nature, vol.464, issue.7290, pp.917-921, 2010.

R. Hägerling, C. Pollmann, M. Andreas, C. Schmidt, H. Nurmi et al., A novel multistep mechanism for initial lymphangiogenesis in mouse embryos based on ultramicroscopy, The EMBO Journal, vol.32, issue.5, pp.629-644, 2013.

G. Hajnóczky, G. Csordás, S. Das, C. Garcia-perez, M. Saotome et al., Mitochondrial calcium signalling and cell death: Approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis, Cell Calcium, vol.40, issue.5-6, pp.553-560, 2006.

S. Hakomori, Glycosylation defining cancer malignancy: New wine in an old bottle, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.10231-10233, 2002.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: The next generation, Cell, vol.144, issue.5, pp.646-674, 2011.

T. Hanaya and H. Yamamoto, Synthesis of biopterin and related pterin glycosides, IUBMB Life, vol.65, issue.4, pp.300-309, 2013.

X. Hao, E. A. Silva, A. Månsson-broberg, K. Grinnemo, A. J. Siddiqui et al., Angiogenic effects of sequential release of VEGF-A165 and PDGF-BB with alginate hydrogels after myocardial infarction, Cardiovascular Research, vol.75, issue.1, pp.178-185, 2007.

U. Harjes, E. Bridges, K. M. Gharpure, I. Roxanis, H. Sheldon et al., Antiangiogenic and tumour inhibitory effects of downregulating tumour endothelial FABP4, Oncogene, vol.36, issue.7, pp.912-921, 2017.

U. Harjes, E. Bridges, A. Mcintyre, B. A. Fielding, and A. L. Harris, Fatty acid-binding protein 4, a point of convergence for angiogenic and metabolic signaling pathways in endothelial cells, The Journal of Biological Chemistry, vol.289, issue.33, pp.23168-23176, 2014.

U. Harjes, J. Kalucka, and P. Carmeliet, Targeting fatty acid metabolism in cancer and endothelial cells, Critical Reviews in Oncology/Hematology, vol.97, pp.15-21, 2016.

N. C. Harris, K. Paavonen, N. Davydova, S. Roufail, T. Sato et al., Proteolytic processing of vascular endothelial growth factor-D is essential for its capacity to promote the growth and spread of cancer, The FASEB Journal, vol.25, issue.8, pp.2615-2625, 2011.

J. Hartwich, W. S. Orr, C. Y. Ng, Y. Spence, C. Morton et al., HIF-1? activation mediates resistance to anti-angiogenic therapy in neuroblastoma xenografts, Journal of Pediatric Surgery, vol.48, issue.1, pp.39-46, 2013.

Z. Hassani, A. Saleh, S. Turpault, S. Khiati, W. Morelle et al., Phostine PST3.1a targets MGAT5 and inhibits glioblastoma-initiating cell invasiveness and proliferation, Mol. Cancer Research, vol.15, issue.10, pp.1376-1387, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01610960

I. Häuselmann and L. Borsig, Altered tumor-cell glycosylation promotes metastasis, Frontiers in Oncology, vol.4, p.28, 2014.

P. R. Hess, D. R. Rawnsley, Z. Jakus, Y. Yang, D. T. Sweet et al., Platelets mediate lymphovenous hemostasis to maintain blood-lymphatic separation throughout life, The Journal of Clinical Investigation, vol.124, issue.1, pp.273-284, 2014.

R. Heusschen, I. A. Schulkens, J. Van-beijnum, A. W. Griffioen, and V. L. Thijssen, Endothelial LGALS9 splice variant expression in endothelial cell biology and angiogenesis, Biochimica et Biophysica Acta, vol.1842, issue.2, pp.284-292, 2014.

M. M. Hickey and M. C. Simon, Regulation of angiogenesis by hypoxia and hypoxiainducible factors, Current Topics in Developmental Biology, vol.76, pp.217-257, 2006.

F. Hillen and A. W. Griffioen, Tumour vascularization: Sprouting angiogenesis and beyond, Cancer Metastasis Reviews, vol.26, issue.3-4, pp.489-502, 2007.

S. Hirakawa, From tumor lymphangiogenesis to lymphvascular niche, Cancer Science, vol.100, issue.6, pp.983-989, 2009.

S. Hirakawa, L. F. Brown, S. Kodama, K. Paavonen, K. Alitalo et al., VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites, Blood, vol.109, issue.3, pp.1010-1017, 2007.

S. Hiratsuka, K. Nakao, K. Nakamura, M. Katsuki, Y. Maru et al., Membrane fixation of vascular endothelial growth factor receptor 1 ligand-binding domain is important for vasculogenesis and angiogenesis in mice, Molecular and Cellular Biology, vol.25, issue.1, pp.346-354, 2005.

B. M. Hogan, F. L. Bos, J. Bussmann, M. Witte, N. C. Chi et al., Ccbe1 is required for embryonic lymphangiogenesis and venous sprouting, Nature Genetics, vol.41, issue.4, pp.396-398, 2009.

Y. Hong, N. Harvey, Y. Noh, V. Schacht, S. Hirakawa et al., Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate, Developmental Dynamics, vol.225, issue.3, pp.351-357, 2002.

J. D. Hood, C. J. Meininger, M. Ziche, and H. J. Granger, VEGF upregulates ecNOS message, protein, and NO production in human endothelial cells, The American Journal of Physiology, vol.274, issue.3, pp.1054-1058, 1998.

S. H. Hsieh, N. W. Ying, M. H. Wu, W. F. Chiang, C. L. Hsu et al., Galectin-1, a novel ligand of neuropilin-1, activates VEGFR-2 signaling and modulates the migration of vascular endothelial cells, Oncogene, vol.27, issue.26, pp.3746-3753, 2008.

C. Huang, S. Wang, L. Lin, P. Wang, T. Chen et al., Glycolytic inhibitor 2-deoxyglucose simultaneously targets cancer and endothelial cells to suppress neuroblastoma growth in mice, Disease Models & Mechanisms, vol.8, issue.10, pp.1247-1254, 2015.

J. E. Hudak and C. R. Bertozzi, Glycotherapy: New advances inspire a reemergence of glycans in medicine, Chemistry & Biology, vol.21, issue.1, pp.16-37, 2014.

R. Huggenberger, S. Ullmann, S. T. Proulx, B. Pytowski, K. Alitalo et al., Stimulation of lymphangiogenesis via VEGFR-3 inhibits chronic skin inflammation, The Journal of Experimental Medicine, vol.207, issue.10, pp.2255-2269, 2010.

P. G. Hultin, Bioactive C-glycosides from bacterial secondary metabolism, Current Topics in Medicinal Chemistry, vol.5, issue.14, pp.1299-1331, 2005.

J. Hunter, Essays and observations on natural history, anatomy, physiology, psychology and geology, 1861.

M. M. Hussain, H. Kotz, L. Minasian, A. Premkumar, G. Sarosy et al., , 2003.

, Phase II trial of carboxyamidotriazole in patients with relapsed epithelial ovarian cancer, Journal of Clinical Oncology, vol.21, issue.23, pp.4356-4363

M. Ichikawa and Y. Ichikawa, Facile synthesis of a new type of iminosugar: A nitrogen atom is in the anomeric position, Bioorganic & Medicinal Chemistry, vol.3, issue.2, pp.161-165, 1995.

L. J. Ignarro, Nitric oxide as a unique signaling molecule in the vascular system: A historical overview, Journal of Physiology and Pharmacology, vol.53, issue.4, pp.503-514, 2002.

K. Inamori, J. Gu, M. Ohira, A. Kawasaki, Y. Nakamura et al., High expression of N-acetylglucosaminyltransferase V in favorable neuroblastomas: Involvement of its effect on apoptosis, FEBS Letters, vol.580, issue.2, pp.627-632, 2006.

C. Inampudi, E. Akintoye, T. Ando, and A. Briasoulis, Angiogenesis in peripheral arterial disease, Current Opinion in Pharmacology, vol.39, pp.60-67, 2018.

A. Irrthum, K. Devriendt, D. Chitayat, G. Matthijs, C. Glade et al., Mutations in the transcription factor gene SOX18 underlie recessive and dominant forms of hypotrichosis-lymphedema-telangiectasia, American Journal of Human Genetics, vol.72, issue.6, pp.1470-1478, 2003.

M. Isaji, H. Miyata, Y. Ajisawa, Y. Takehana, and N. Yoshimura, Tranilast inhibits the proliferation, chemotaxis and tube formation of human microvascular endothelial cells in vitro and angiogenesis in vivo, British Journal of Pharmacology, vol.122, issue.6, pp.1061-1066, 1997.

J. S. Isenberg, G. Martin-manso, J. B. Maxhimer, and D. D. Roberts, Regulation of nitric oxide signalling by thrombospondin 1: Implications for anti-angiogenic therapies, Nature Reviews. Cancer, vol.9, issue.3, pp.182-194, 2009.

H. Ishimura, T. Takahashi, H. Nakagawa, S. Nishimura, Y. Arai et al., N-acetylglucosaminyltransferase V and beta1-6 branching N-linked oligosaccharides are associated with good prognosis of patients with bladder cancer, Clinical Cancer Research, vol.12, issue.8, pp.2506-2511, 2006.

K. Ito, S. A. Scott, S. Cutler, L. Dong, J. Neuzil et al., Thiodigalactoside inhibits murine cancers by concurrently blocking effects of galectin-1 on immune dysregulation, angiogenesis and protection against oxidative stress, Angiogenesis, vol.14, issue.3, pp.293-307, 2011.

K. Ito, K. Stannard, E. Gabutero, A. M. Clark, S. Neo et al., Galectin-1 as a potent target for cancer therapy: Role in the tumor microenvironment, Cancer Metastasis Reviews, vol.31, issue.3-4, pp.763-778, 2012.

Y. Ito, E. Miyoshi, M. Sakon, T. Takeda, K. Noda et al., Elevated expression of UDP-N-acetylglucosamine: Alphamannoside beta1,6 Nacetylglucosaminyltransferase is an early event in hepatocarcinogenesis, International Journal of Cancer, vol.91, issue.5, pp.631-637, 2001.

M. Jabs, A. J. Rose, L. H. Lehmann, J. Taylor, I. Moll et al., Inhibition of endothelial notch signaling impairs fatty acid transport and leads to metabolic and vascular remodeling of the adult heart, Circulation, vol.137, issue.24, pp.2592-2608, 2018.

J. Jacobs, Combating cardiovascular disease with angiogenic therapy, Drug Discovery Today, vol.12, pp.1040-1045, 2007.

T. Jain, E. A. Nikolopoulou, Q. Xu, and A. Qu, Hypoxia inducible factor as a therapeutic target for atherosclerosis, Pharmacology & Therapeutics, vol.183, pp.22-33, 2018.

Y. Jang, J. Han, S. J. Kim, J. Kim, M. J. Lee et al., Suppression of mitochondrial respiration with auraptene inhibits the progression of renal cell carcinoma: Involvement of HIF-1? degradation, Oncotarget, vol.6, issue.35, pp.38127-38138, 2015.

R. Ji, Macrophages are important mediators of either tumor-or inflammationinduced lymphangiogenesis, Cellular and Molecular Life Sciences, vol.69, issue.6, pp.897-914, 2012.

R. Ji, Hypoxia and lymphangiogenesis in tumor microenvironment and metastasis, Cancer Letters, vol.346, issue.1, pp.6-16, 2014.

G. Jiménez-valerio and O. Casanovas, Angiogenesis and metabolism: Entwined for therapy resistance, Trends Cancer, vol.3, issue.1, pp.10-18, 2017.

J. L. Johnson, M. B. Jones, S. O. Ryan, and B. A. Cobb, The regulatory power of glycans and their binding partners in immunity, Trends in Immunology, vol.34, issue.6, pp.290-298, 2013.

N. C. Johnson, M. E. Dillard, P. Baluk, D. M. Mcdonald, N. L. Harvey et al., Lymphatic endothelial cell identity is reversible and its maintenance requires Prox1 activity, Genes & Development, vol.22, issue.23, pp.3282-3291, 2008.

S. F. Jones and J. R. Infante, Molecular pathways: Fatty acid synthase, Clinical Cancer Research, vol.21, issue.24, pp.5434-5438, 2015.

T. Ju, K. Brewer, A. Souza, R. D. Cummings, and W. M. Canfield, Cloning and expression of human core 1 beta1,3-galactosyltransferase, The Journal of Biological Chemistry, vol.277, issue.1, pp.178-186, 2002.

S. Julien, M. Bobowski, A. Steenackers, X. Le-bourhis, and P. Delannoy, How do gangliosides regulate RTKs signaling, Cell, vol.2, issue.4, pp.751-767, 2013.
URL : https://hal.archives-ouvertes.fr/hal-00916107

H. J. Jung, J. S. Shim, J. Lee, Y. M. Song, K. C. Park et al., Terpestacin inhibits tumor angiogenesis by targeting UQCRB of mitochondrial complex III and suppressing hypoxia-induced reactive oxygen species production and cellular oxygen sensing, The Journal of Biological Chemistry, vol.285, issue.15, pp.11584-11595, 2010.

L. Jussila and K. Alitalo, Vascular growth factors and lymphangiogenesis, Physiological Reviews, vol.82, issue.3, pp.673-700, 2002.

E. Kaiserling, S. Kröber, and S. Geleff, Lymphatic vessels in the colonic mucosa in ulcerative colitis, Lymphology, vol.36, issue.2, pp.52-61, 2003.

S. Kanda, E. Landgren, M. Ljungström, and L. Claesson-welsh, Fibroblast growth factor receptor 1-induced differentiation of endothelial cell line established from tsA58 large T transgenic mice, Cell Growth & Differentiation, vol.7, issue.3, pp.383-395, 1996.

J. Kang, J. Yoo, S. Lee, W. Tang, B. Aguilar et al., An exquisite cross-control mechanism among endothelial cell fate regulators directs the plasticity and heterogeneity of lymphatic endothelial cells, Blood, vol.116, issue.1, pp.140-150, 2010.

M. J. Karkkainen, R. E. Ferrell, E. C. Lawrence, M. A. Kimak, K. L. Levinson et al., Missense mutations interfere with VEGFR-3 signalling in primary lymphoedema, Nature Genetics, vol.25, issue.2, pp.153-159, 2000.

M. J. Karkkainen, L. Jussila, R. E. Ferrell, D. N. Finegold, and K. Alitalo, Molecular regulation of lymphangiogenesis and targets for tissue oedema, Trends in Molecular Medicine, vol.7, issue.1, pp.18-22, 2001.

T. Karnezis, R. Shayan, C. Caesar, S. Roufail, N. C. Harris et al., VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium, Cancer Cell, vol.21, issue.2, pp.181-195, 2012.

T. Karpanen, M. Wirzenius, T. Mäkinen, T. Veikkola, H. J. Haisma et al., Lymphangiogenic growth factor responsiveness is modulated by postnatal lymphatic vessel maturation, The American Journal of Pathology, vol.169, issue.2, pp.708-718, 2006.

S. Kawakami and M. Hashida, Glycosylation-mediated targeting of carriers, Journal of Controlled Release, vol.190, pp.542-555, 2014.

J. Kazenwadel, G. A. Secker, Y. J. Liu, J. A. Rosenfeld, R. S. Wildin et al., Loss-of-function germline GATA2 mutations in patients with MDS/AML or MonoMAC syndrome and primary lymphedema reveal a key role for GATA2 in the lymphatic vasculature, Blood, vol.119, issue.5, pp.1283-1291, 2012.

R. L. Kendall, G. Wang, and K. A. Thomas, Identification of a natural soluble form of the vascular endothelial growth factor receptor, FLT-1, and its heterodimerization with KDR, Biochemical and Biophysical Research Communications, vol.226, issue.2, pp.324-328, 1996.

T. Keravis, L. Favot, A. A. Abusnina, A. Anton, H. Justiniano et al., Delphinidin inhibits tumor growth by acting on VEGF signalling in endothelial cells, PloS One, vol.10, issue.12, p.145291, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01280466

O. Keunen, M. Johansson, A. Oudin, M. Sanzey, S. A. Rahim et al., Anti-VEGF treatment reduces blood supply and increases tumor cell invasion in glioblastoma, Proceedings of the National Academy of Sciences of the United States of America, vol.108, issue.9, pp.3749-3754, 2011.

H. Kim, V. P. Nguyen, T. V. Petrova, M. Cruz, K. Alitalo et al., Embryonic vascular endothelial cells are malleable to reprogramming via Prox1 to a lymphatic gene signature, BMC Developmental Biology, vol.10, p.72, 2010.

Y. Kim, S. Kim, R. Tatsunami, H. Yamamura, T. Fukai et al., ROS-induced ROS release orchestrated by Nox4, Nox2, and mitochondria in VEGF signaling and angiogenesis, Am. J. Physiol. Cellular Physiology, vol.312, issue.6, pp.749-764, 2017.

S. Kitazume, R. Imamaki, K. Ogawa, Y. Komi, S. Futakawa et al., , 2010.

. Alpha2, 6-sialic acid on platelet endothelial cell adhesion molecule (PECAM) regulates its homophilic interactions and downstream antiapoptotic signaling, The Journal of Biological Chemistry, vol.285, issue.9, pp.6515-6521

H. Kiziltoprak, E. Yetkin, K. Tekin, M. Koc, ?. Akalin et al., Intravitreal aflibercept for the treatment of choroidal neovascularization associated with rubella retinopathy, International Ophthalmology, 2018.

M. A. Kluge, J. L. Fetterman, and J. A. Vita, Mitochondria and endothelial function, Circulation Research, vol.112, issue.8, pp.1171-1188, 2013.

S. Koch, S. Tugues, X. Li, L. Gualandi, and L. Claesson-welsh, Signal transduction by vascular endothelial growth factor receptors, The Biochemical Journal, vol.437, issue.2, pp.169-183, 2011.

N. A. Koonce, R. J. Griffin, and R. P. Dings, Galectin-1 inhibitor OTX008 induces tumor vessel normalization and tumor growth inhibition in human head and neck qquamous cell carcinoma models, International Journal of Molecular Sciences, vol.18, issue.12, 2017.

A. Koziel, A. Woyda-ploszczyca, A. Kicinska, and W. Jarmuszkiewicz, The influence of high glucose on the aerobic metabolism of endothelial EA.hy926 cells, Pflügers Archiv, vol.464, issue.6, pp.657-669, 2012.

J. Krieg, S. Hartmann, A. Vicentini, W. Gläsner, D. Hess et al., Recognition signal for C-mannosylation of Trp-7 in RNase 2 consists of sequence Trp-x-xTrp, Molecular Biology of the Cell, vol.9, issue.2, pp.301-309, 1998.

J. Kroll and J. Waltenberger, A novel function of VEGF receptor-2 (KDR): Rapid release of nitric oxide in response to VEGF-A stimulation in endothelial cells, Biochemical and Biophysical Research Communications, vol.265, issue.3, pp.636-639, 1999.

A. Krützfeldt, R. Spahr, S. Mertens, B. Siegmund, and H. M. Piper, Metabolism of exogenous substrates by coronary endothelial cells in culture, Journal of Molecular and Cellular Cardiology, vol.22, issue.12, pp.1393-1404, 1990.

D. Kumar, S. Haldar, M. Gorain, S. Kumar, F. A. Mulani et al., Epoxyazadiradione suppresses breast tumor growth through mitochondrial depolarization and caspase-dependent apoptosis by targeting PI3K/Akt pathway, BMC Cancer, vol.18, issue.1, p.52, 2018.

K. Kumar, S. Wigfield, H. E. Gee, C. M. Devlin, D. Singleton et al., Dichloroacetate reverses the hypoxic adaptation to bevacizumab and enhances its antitumor effects in mouse xenografts, Journal of Molecular Medicine, vol.91, issue.6, pp.749-758, 2013.

S. Kumar and P. Cieplak, Role of N-glycosylation in activation of proMMP-9. A molecular dynamics simulations study, PloS One, vol.13, issue.1, 2018.

M. Kurtoglu, N. Gao, J. Shang, J. C. Maher, M. A. Lehrman et al., Under normoxia, 2-deoxy-D-glucose elicits cell death in select tumor types not by inhibition of glycolysis but by interfering with N-linked glycosylation, Molecular Cancer Therapeutics, vol.6, issue.11, pp.3049-3058, 2007.

L. Del-puerto-nevado, F. Rojo, S. Zazo, C. Caramés, G. Rubio et al., Active angiogenesis in metastatic renal cell carcinoma predicts clinical benefit to sunitinibbased therapy, British Journal of Cancer, vol.110, issue.11, pp.2700-2707, 2014.

M. Labrie, L. O. De-araujo, L. Communal, A. Mes-masson, Y. St-pierre et al., A unique galectin signature in human prostate cancer progression suggests galectin-1 as a key target for treatment of advanced disease, Scientific Reports, vol.7, issue.1, pp.86-96, 2013.

K. Lashkari, T. Hirose, J. Yazdany, J. W. Mcmeel, A. Kazlauskas et al., Vascular endothelial growth factor and hepatocyte growth factor levels are differentially elevated in patients with advanced retinopathy of prematurity, The American Journal of Pathology, vol.156, issue.4, pp.1337-1344, 2000.

Y. T. Lau and W. C. Ma, Nitric oxide inhibits migration of cultured endothelial cells, Biochemical and Biophysical Research Communications, vol.221, issue.3, pp.670-674, 1996.

A. Lavaud, R. Soleti, A. Hay, P. Richomme, D. Guilet et al., Paradoxical effects of polyphenolic compounds from Clusiaceae on angiogenesis, Biochemical Pharmacology, vol.83, issue.4, pp.514-523, 2012.
URL : https://hal.archives-ouvertes.fr/halsde-00722703

K. Lavrsen, S. Dabelsteen, S. Y. Vakhrushev, A. M. Levann, A. D. Haue et al., De novo expression of human polypeptideN-acetylgalactosaminyltransferase 6 (GalNAc-T6) in colon adenocarcinoma inhibits the differentiation of colonic epithelium, The Journal of Biological Chemistry, vol.293, issue.4, pp.1298-1314, 2018.

L. Mercier, M. Mathieu, V. Haibe-kains, B. Bontempi, G. Mijatovic et al., Knocking down galectin 1 in human hs683 glioblastoma cells impairs both angiogenesis and endoplasmic reticulum stress responses, Journal of Neuropathology and Experimental Neurology, vol.67, issue.5, pp.456-469, 2008.

Q. Le, G. Shi, H. Cao, D. W. Nelson, Y. Wang et al., Galectin-1: A link between tumor hypoxia and tumor immune privilege, Journal of Clinical Oncology, vol.23, issue.35, pp.8932-8941, 2005.

B. Lee, K. H. Kim, H. J. Jung, and H. J. Kwon, Matairesinol inhibits angiogenesis via suppression of mitochondrial reactive oxygen species, Biochemical and Biophysical Research Communications, vol.421, issue.1, pp.76-80, 2012.

K. Legler, R. Rosprim, T. Karius, K. Eylmann, M. Rossberg et al., Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer, Br. J. Cancer, 2018.

P. Lenzi, G. Bocci, and G. Natale, John Hunter and the origin of the term "angiogenesis, Angiogenesis, vol.19, issue.2, pp.255-256, 2016.

H. Li, J. Yang, Z. Liu, W. Wang, Z. Yu et al., Blockage of glycolysis by targeting PFKFB3 suppresses tumor growth and metastasis in head and neck squamous cell carcinoma, J. Exp. Clin. Cancer Res, vol.36, issue.1, p.7, 2017.

J. Li, T. Hampton, J. P. Morgan, and M. Simons, Stretch-induced VEGF expression in the heart, The Journal of Clinical Investigation, vol.100, issue.1, pp.18-24, 1997.

J. Li and A. M. Shah, Endothelial cell superoxide generation: Regulation and relevance for cardiovascular pathophysiology, American Journal of Physiology. Regulatory, Integrative and Comparative Physiology, vol.287, issue.5, pp.1014-1030, 2004.

N. Li, H. Xu, K. Fan, X. Liu, J. Qi et al., Altered ?1,6-GlcNAc branched Nglycans impair TGF-?-mediated epithelial-to-mesenchymal transition through Smad signalling pathway in human lung cancer, Journal of Cellular and Molecular Medicine, vol.18, issue.10, pp.1975-1991, 2014.

X. Li, J. Liu, L. Qian, H. Ke, C. Yao et al., Expression of PFKFB3 and Ki67 in lung adenocarcinomas and targeting PFKFB3 as a therapeutic strategy, Mol. Cell. Biochem, 2018.

S. Liao and P. Y. Weid, Lymphatic system: An active pathway for immune protection, Seminars in Cell & Developmental Biology, vol.38, pp.83-89, 2015.

J. W. Locasale and L. C. Cantley, Metabolic flux and the regulation of mammalian cell growth, Cell Metabolism, vol.14, issue.4, pp.443-451, 2011.

F. Lodola, U. Laforenza, E. Bonetti, D. Lim, S. Dragoni et al., Store-operated Ca2+ entry is remodelled and controls in vitro angiogenesis in endothelial progenitor cells isolated from tumoral patients, PLoS One, vol.7, issue.9, p.42541, 2012.

G. A. Lutty, Effects of diabetes on the eye, Invest. Ophthalmol. Vis. Sci, vol.54, issue.14, pp.81-87, 2013.

C. M. Machado, L. N. Andrade, V. R. Teixeira, F. F. Costa, C. M. Melo et al., Galectin-3 disruption impaired tumoral angiogenesis by reducing VEGF secretion from TGF?1-induced macrophages, Cancer Medicine, vol.3, issue.2, pp.201-214, 2014.

J. L. Magnani and B. Ernst, Glycomimetic drugs-a new source of therapeutic opportunities, Discovery Medicine, vol.8, issue.43, pp.247-252, 2009.

J. C. Maher, M. Wangpaichitr, N. Savaraj, M. Kurtoglu, and T. J. Lampidis, Hypoxiainducible factor-1 confers resistance to the glycolytic inhibitor 2-deoxy-D-glucose, Molecular Cancer Therapeutics, vol.6, issue.2, pp.732-741, 2007.

P. C. Maisonpierre, C. Suri, P. F. Jones, S. Bartunkova, S. J. Wiegand et al., Angiopoietin-2, a natural antagonist for Tie2 that disrupts in vivo angiogenesis, Science, vol.277, issue.5322, pp.55-60, 1997.

T. Mäkinen, T. Veikkola, S. Mustjoki, T. Karpanen, B. Catimel et al., Isolated lymphatic endothelial cells transduce growth, survival and migratory signals via the VEGF-C/D receptor VEGFR-3, The EMBO J, vol.20, issue.17, pp.4762-4773, 2001.

Z. Mallat and A. Tedgui, Apoptosis in the vasculature: Mechanisms and functional importance, British Journal of Pharmacology, vol.130, issue.5, pp.947-962, 2000.

S. W. Malm, N. T. Hanke, A. Gill, L. Carbajal, and A. F. Baker, The anti-tumor efficacy of 2-deoxyglucose and D-allose are enhanced with p38 inhibition in pancreatic and ovarian cell lines, Journal of Experimental & Clinical Cancer Research, vol.34, p.31, 2015.

A. Mantovani, S. Sozzani, M. Locati, P. Allavena, and A. Sica, Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes, Trends in Immunology, vol.23, issue.11, pp.549-555, 2002.

R. Marcu, Y. Zheng, and B. J. Hawkins, Mitochondria and angiogenesis, Adv. Exp. Med. Biol, vol.982, pp.371-406, 2017.

A. I. Markowska, K. C. Jefferies, and N. Panjwani, Galectin-3 protein modulates cell surface expression and activation of vascular endothelial growth factor receptor 2 in human endothelial cells, The Journal of Biological Chemistry, vol.286, issue.34, pp.29913-29921, 2011.

A. I. Markowska, F. Liu, and N. Panjwani, Galectin-3 is an important mediator of VEGF-and bFGF-mediated angiogenic response, The Journal of Experimental Medicine, vol.207, issue.9, pp.1981-1993, 2010.

S. Martin, L. Favot, R. Matz, C. Lugnier, and R. Andriantsitohaina, Delphinidin inhibits endothelial cell proliferation and cell cycle progression through a transient activation of ERK-1/?2, Biochemical Pharmacology, vol.65, issue.4, pp.669-675, 2003.

M. C. Martinez and R. Andriantsitohaina, Microparticles in angiogenesis: Therapeutic potential, Circulation Research, vol.109, issue.1, pp.110-119, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01390661

N. Martinez-bosch, J. Vinaixa, and P. Navarro, Immune evasion in pancreatic cancer: From mechanisms to therapy, Cancer, vol.10, issue.1, 2018.

K. Maruyama, M. Ii, C. Cursiefen, D. G. Jackson, H. Keino et al., Inflammation-induced lymphangiogenesis in the cornea arises from CD11b-positive macrophages, The Journal of Clinical Investigation, vol.115, issue.9, pp.2363-2372, 2005.

D. G. Mashek and R. A. Coleman, Cellular fatty acid uptake: The contribution of metabolism, Current Opinion in Lipidology, vol.17, issue.3, pp.274-278, 2006.

I. Masouyé, G. Hagens, T. H. Van-kuppevelt, P. Madsen, J. H. Saurat et al., Endothelial cells of the human microvasculature express epidermal fatty acid-binding protein, Circulation Research, vol.81, issue.3, pp.297-303, 1997.

H. Matsumoto, T. Hiroe, M. Morimoto, K. Mimura, A. Ito et al., Efficacy of treat-and-extend regimen with aflibercept for pachychoroid neovasculopathy and Type 1 neovascular age-related macular degeneration, Japanese Journal of Ophthalmology, vol.62, issue.2, pp.144-150, 2018.

W. Matthews, C. T. Jordan, G. W. Wiegand, D. Pardoll, and I. R. Lemischka, A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations, Cell, vol.65, issue.7, pp.1143-1152, 1991.

N. Maulik and M. Thirunavukkarasu, Growth factors and cell therapy in myocardial regeneration, Journal of Molecular and Cellular Cardiology, vol.44, issue.2, pp.219-227, 2008.

A. Mcintyre and A. L. Harris, Metabolic and hypoxic adaptation to anti-angiogenic therapy: A target for induced essentiality, EMBO Molecular Medicine, vol.7, issue.4, pp.368-379, 2015.

S. P. Méndez-huergo, A. G. Blidner, and G. A. Rabinovich, Galectins: Emerging regulatory checkpoints linking tumor immunity and angiogenesis, Current Opinion in Immunology, vol.45, pp.8-15, 2017.

J. R. Merchan, K. Kovács, J. W. Railsback, M. Kurtoglu, Y. Jing et al., Antiangiogenic activity of 2-deoxy-D-glucose, PLoS One, vol.5, issue.10, p.13699, 2010.

M. Milkiewicz, M. D. Brown, S. Egginton, and O. Hudlicka, Association between shear stress, angiogenesis, and VEGF in skeletal muscles in vivo, Microcirculation, vol.8, issue.4, pp.229-241, 2001.

R. Missiaen, F. Morales-rodriguez, G. Eelen, and P. Carmeliet, Targeting endothelial metabolism for anti-angiogenesis therapy: A pharmacological perspective, Vascular Pharmacology, vol.90, pp.8-18, 2017.

D. J. Moloney, L. H. Shair, F. M. Lu, J. Xia, R. Locke et al., Mammalian Notch1 is modified with two unusual forms of O-linked glycosylation found on epidermal growth factor-like modules, The Journal of Biological Chemistry, vol.275, issue.13, pp.9604-9611, 2000.

K. W. Moremen, M. Tiemeyer, and A. V. Nairn, Vertebrate protein glycosylation: Diversity, synthesis and function, Nature Reviews. Molecular Cell Biology, vol.13, issue.7, pp.448-462, 2012.

V. Moreno, P. Gonzalo, J. Gómez-escudero, Á. Pollán, R. Acín-pérez et al., An EMMPRIN-?-catenin-Nm23 complex drives ATP production and actomyosin contractility at endothelial junctions, Journal of Cell Science, vol.127, pp.3768-3781, 2014.

F. Morfoisse, F. Tatin, F. Hantelys, A. Adoue, A. Helfer et al., Nucleolin promotes heat shock-associated translation of VEGF-D to promote tumor lymphangiogenesis, Cancer Research, vol.76, issue.15, pp.4394-4405, 2016.

S. Morikawa, P. Baluk, T. Kaidoh, A. Haskell, R. K. Jain et al., Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors, The American Journal of Pathology, vol.160, issue.3, pp.985-1000, 2002.

J. Munkley and D. J. Elliott, Hallmarks of glycosylation in cancer, Oncotarget, vol.7, issue.23, pp.35478-35489, 2016.

M. P. Murphy and R. A. Smith, Drug delivery to mitochondria: The key to mitochondrial medicine, Advanced Drug Delivery Reviews, vol.41, issue.2, pp.235-250, 2000.

J. A. Nagy, E. S. Morgan, K. T. Herzberg, E. J. Manseau, A. M. Dvorak et al., Pathogenesis of ascites tumor growth: Angiogenesis, vascular remodeling, and stroma formation in the peritoneal lining, Cancer Research, vol.55, issue.2, pp.376-385, 1995.

S. Nakahara, T. Saito, N. Kondo, K. Moriwaki, K. Noda et al., A secreted type of beta1,6 N-acetylglucosaminyltransferase V (GnT-V), a novel angiogenesis inducer, is regulated by gamma-secretase, The FASEB Journal, vol.20, issue.14, pp.2451-2459, 2006.

A. W. Nap, G. A. Dunselman, A. W. Griffioen, K. H. Mayo, J. L. Evers et al., Angiostatic agents prevent the development of endometriosis-like lesions in the chicken chorioallantoic membrane, Fertility and Sterility, vol.83, issue.3, pp.793-795, 2005.

G. Nardo, E. Favaro, M. Curtarello, L. Moserle, E. Zulato et al., Glycolytic phenotype and AMP kinase modify the pathologic response of tumor xenografts to VEGF neutralization, Cancer Research, vol.71, issue.12, pp.4214-4225, 2011.

S. Niccoli, D. R. Boreham, C. P. Phenix, and S. J. Lees, Non-radioactive 2-deoxy-2-fluoro-D-glucose inhibits glucose uptake in xenograft tumours and sensitizes HeLa cells to doxorubicin in vitro, PLoS One, vol.12, issue.11, p.187584, 2017.

K. Niessen, G. Zhang, J. B. Ridgway, H. Chen, G. Kolumam et al., The Notch1-Dll4 signaling pathway regulates mouse postnatal lymphatic development, Blood, vol.118, issue.7, pp.1989-1997, 2011.

C. Norrmén, K. I. Ivanov, J. Cheng, N. Zangger, M. Delorenzi et al., FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1, The Journal of Cell Biology, vol.185, issue.3, pp.439-457, 2009.

C. Norrmén, T. Tammela, T. V. Petrova, and K. Alitalo, Biological basis of therapeutic lymphangiogenesis, Circulation, vol.123, issue.12, pp.1335-1351, 2011.

M. Obach, A. Navarro-sabaté, J. Caro, X. Kong, J. Duran et al., 6-Phosphofructo-2-kinase (pfkfb3) gene promoter contains hypoxia-inducible factor-1 binding sites necessary for transactivation in response to hypoxia, The Journal of Biological Chemistry, vol.279, issue.51, pp.53562-53570, 2004.

F. Ogata, K. Fujiu, S. Matsumoto, Y. Nakayama, M. Shibata et al., Excess lymphangiogenesis cooperatively induced by macrophages and CD4(+) T cells drives the pathogenesis of lymphedema, The Journal of Investigative Dermatology, vol.136, issue.3, pp.706-714, 2016.

K. Ohtsubo and J. D. Marth, Glycosylation in cellular mechanisms of health and disease, Cell, vol.126, issue.5, pp.855-867, 2006.

U. A. Okonkwo and L. A. Dipietro, Diabetes and wound angiogenesis, International Journal of Molecular Sciences, vol.18, issue.7, 2017.

W. H. Oldendorf, M. E. Cornford, and W. J. Brown, The large apparent work capability of the blood-brain barrier: A study of the mitochondrial content of capillary endothelial cells in brain and other tissues of the rat, Annals of Neurology, vol.1, issue.5, pp.409-417, 1977.

K. Paavonen, J. Mandelin, T. Partanen, L. Jussila, T. Li et al., Vascular endothelial growth factors C and D and their VEGFR-2 and 3 receptors in blood and lymphatic vessels in healthy and arthritic synovium, The Journal of Rheumatology, vol.29, issue.1, pp.39-45, 2002.

M. Pàez-ribes, E. Allen, J. Hudock, T. Takeda, H. Okuyama et al., Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis, Cancer Cell, vol.15, issue.3, pp.220-231, 2009.

I. Pafumi, M. Festa, F. Papacci, L. Lagostena, C. Giunta et al., Naringenin impairs two-pore channel 2 activity and inhibits VEGF-induced angiogenesis, Scientific Reports, vol.7, issue.1, p.5121, 2017.

K. Pajusola, O. Aprelikova, J. Korhonen, A. Kaipainen, L. Pertovaara et al., FLT4 receptor tyrosine kinase contains seven immunoglobulin-like loops and is expressed in multiple human tissues and cell lines, Cancer Research, vol.52, issue.20, pp.5738-5743, 1992.

E. R. Palmacci and P. H. Seeberger, Synthesis of C-aryl and C-alkyl glycosides using glycosyl phosphates, Organic Letters, vol.3, issue.10, pp.1547-1550, 2001.

Y. Pan, W. Wang, and T. Yago, Transcriptional regulation of podoplanin expression by Prox1 in lymphatic endothelial cells, Microvascular Research, vol.94, pp.96-102, 2014.

M. Pangare and A. Makino, Mitochondrial function in vascular endothelial cell in diabetes, Journal of Smooth Muscle Research, vol.48, issue.1, pp.1-26, 2012.

S. Parangi, M. O'reilly, G. Christofori, L. Holmgren, J. Grosfeld et al., Antiangiogenic therapy of transgenic mice impairs de novo tumor growth, Proceedings of the National Academy of Sciences of the United States of America, vol.93, issue.5, pp.2002-2007, 1996.

D. Park and P. J. Dilda, Mitochondria as targets in angiogenesis inhibition, Molecular Aspects of Medicine, vol.31, issue.1, pp.113-131, 2010.

G. Parra-bonilla, D. F. Alvarez, A. Al-mehdi, M. Alexeyev, and T. Stevens, Critical role for lactate dehydrogenase A in aerobic glycolysis that sustains pulmonary microvascular endothelial cell proliferation, American Journal of Physiology. Lung Cellular and Molecular Physiology, vol.299, issue.4, pp.513-522, 2010.

J. D. Parrish and R. D. Little, Preparation of alpha-C-glycosides from glycals, Organic Letters, vol.4, issue.9, pp.1439-1442, 2002.

E. A. Partridge, C. Le-roy, G. M. Di-guglielmo, J. Pawling, P. Cheung et al., Regulation of cytokine receptors by Golgi N-glycan processing and endocytosis, Science, vol.306, issue.5693, pp.120-124, 2004.

F. Patella, Z. T. Schug, E. Persi, L. J. Neilson, Z. Erami et al., Proteomics-based metabolic modeling reveals that fatty acid oxidation (FAO) controls endothelial cell (EC) permeability, Molecular & Cellular Proteomics, vol.14, issue.3, pp.621-634, 2015.

B. Peck and A. Schulze, Lipid desaturation -the next step in targeting lipogenesis in cancer?, The FEBS Journal, vol.283, issue.15, pp.2767-2778, 2016.

C. Pellet-many, P. Frankel, H. Jia, and I. Zachary, Neuropilins: Structure, function and role in disease, The Biochemical Journal, vol.411, issue.2, pp.211-226, 2008.

J. Peng, K. Hui, C. Hao, Z. Peng, Q. X. Gao et al., Low bone turnover and reduced angiogenesis in streptozotocin-induced osteoporotic mice, Connective Tissue Research, vol.57, issue.4, pp.277-289, 2016.

H. L. Penny, J. L. Sieow, G. Adriani, W. H. Yeap, P. See-chi-ee et al., Warburg metabolism in tumor-conditioned macrophages promotes metastasis in human pancreatic ductal adenocarcinoma, Oncoimmunology, vol.5, issue.8, p.1191731, 2016.

G. Perrone, B. Vincenzi, D. Santini, A. Verzì, G. Tonini et al., Correlation of p53 and bcl-2 expression with vascular endothelial growth factor (VEGF), microvessel density (MVD) and clinico-pathological features in colon cancer, Cancer Letters, vol.208, issue.2, pp.227-234, 2004.

T. V. Petrova, T. Karpanen, C. Norrmén, R. Mellor, T. Tamakoshi et al., Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis, Nature Medicine, vol.10, issue.9, pp.974-981, 2004.

A. Pettersson, J. A. Nagy, L. F. Brown, C. Sundberg, E. Morgan et al., Heterogeneity of the angiogenic response induced in different normal adult tissues by vascular permeability factor/vascular endothelial growth factor, Laboratory Investigation, vol.80, issue.1, pp.99-115, 2000.

S. S. Pinho and C. A. Reis, Glycosylation in cancer: Mechanisms and clinical implications, Nature Reviews. Cancer, vol.15, issue.9, pp.540-555, 2015.

E. Poche?, A. Lity?ska, A. Amoresano, and A. Casbarra, Glycosylation profile of integrin alpha 3 beta 1 changes with melanoma progression, Biochimica et Biophysica Acta, vol.1643, issue.1-3, pp.113-123, 2003.

F. Polet and O. Feron, Endothelial cell metabolism and tumour angiogenesis: Glucose and glutamine as essential fuels and lactate as the driving force, Journal of Internal Medicine, vol.273, issue.2, pp.156-165, 2013.

M. Potente and P. Carmeliet, The link between angiogenesis and endothelial metabolism, Annual Review of Physiology, vol.79, pp.43-66, 2017.

M. Potente, H. Gerhardt, and P. Carmeliet, Basic and therapeutic aspects of angiogenesis, Cell, vol.146, issue.6, pp.873-887, 2011.

C. Prahst, M. Héroult, A. A. Lanahan, N. Uziel, O. Kessler et al., Neuropilin-1-VEGFR-2 complexing requires the PDZ-binding domain of neuropilin-1, The Journal of Biological Chemistry, vol.283, issue.37, pp.25110-25114, 2008.

L. A. Quayle, M. G. Pereira, G. Scheper, T. Wiltshire, R. E. Peake et al., Anti-angiogenic drugs: Direct anti-cancer agents with mitochondrial mechanisms of action, Oncotarget, vol.8, issue.51, pp.88670-88688, 2017.

T. P. Quinn, K. G. Peters, C. De-vries, N. Ferrara, and L. T. Williams, Fetal liver kinase 1 is a receptor for vascular endothelial growth factor and is selectively expressed in vascular endothelium, Proceedings of the National Academy of Sciences of the United States of America, vol.90, issue.16, pp.7533-7537, 1993.

M. Quintero, S. L. Colombo, A. Godfrey, and S. Moncada, Mitochondria as signaling organelles in the vascular endothelium, Proceedings of the National Academy of Sciences of the United States of America, vol.103, issue.14, pp.5379-5384, 2006.

U. Raaz, R. Toh, L. Maegdefessel, M. Adam, F. Nakagami et al., Hemodynamic regulation of reactive oxygen species: Implications for vascular diseases, 2014.

, Antioxidants & Redox Signaling, vol.20, issue.6, pp.914-928

G. A. Rabinovich and J. R. Conejo-garcía, Shaping the immune landscape in cancer by galectin-driven regulatory pathways, Journal of Molecular Biology, vol.428, issue.16, pp.3266-3281, 2016.

A. Ramu, S. Kathiresan, and B. Ahmed, Gramine inhibits angiogenesis and induces apoptosis via modulation of TGF-? signalling in 7,12 dimethylbenz[a]anthracene (DMBA) induced hamster buccal pouch carcinoma, Phytomedicine, vol.33, pp.69-76, 2017.

J. A. Reihill, M. Ewart, D. G. Hardie, and I. P. Salt, AMP-activated protein kinase mediates VEGF-stimulated endothelial NO production, Biochemical and Biophysical Research Communications, vol.354, issue.4, pp.1084-1088, 2007.

K. Reisinger, R. Kaufmann, and J. Gille, Increased Sp1 phosphorylation as a mechanism of hepatocyte growth factor (HGF/SF)-induced vascular endothelial growth factor (VEGF/VPF) transcription, Journal of Cell Science, vol.116, pp.225-238, 2003.

G. Ren, L. H. Michael, M. L. Entman, and N. G. Frangogiannis, Morphological characteristics of the microvasculature in healing myocardial infarcts, The Journal of Histochemistry and Cytochemistry, vol.50, issue.1, pp.71-79, 2002.

S. Rey and G. L. Semenza, Hypoxia-inducible factor-1-dependent mechanisms of vascularization and vascular remodelling, Cardiovascular Research, vol.86, issue.2, pp.236-242, 2010.

N. Roberts, B. Kloos, M. Cassella, S. Podgrabinska, K. Persaud et al., Inhibition of VEGFR-3 activation with the antagonistic antibody more potently suppresses lymph node and distant metastases than inactivation of VEGFR-2, Cancer Research, vol.66, issue.5, pp.2650-2657, 2006.

M. F. Rossier, T channels and steroid biosynthesis: In search of a link with mitochondria, Cell Calcium, vol.40, issue.2, pp.155-164, 2006.

C. Rousseau and O. R. Martin, Stereodirected synthesis of aryl alpha-C-glycosides from 2-O-arylsilyl-glucopyranosides, Organic Letters, vol.5, issue.20, pp.3763-3766, 2003.

A. Sabine, E. Bovay, C. S. Demir, W. Kimura, M. Jaquet et al., FOXC2 and fluid shear stress stabilize postnatal lymphatic vasculature, The Journal of Clinical Investigation, vol.125, issue.10, pp.3861-3877, 2015.

P. Saharinen, M. Bry, and K. Alitalo, How do angiopoietins tie in with vascular endothelial growth factors?, Current Opinion in Hematology, vol.17, issue.3, pp.198-205, 2010.

M. W. Saif, J. A. Knost, E. G. Chiorean, S. R. Kambhampati, D. Yu et al., Phase 1 study of the anti-vascular endothelial growth factor receptor 3 monoclonal antibody LY3022856/IMC-3C5 in patients with advanced and refractory solid tumors and advanced colorectal cancer, Cancer Chemother. Pharmacol, vol.78, issue.4, pp.815-824, 2016.

T. Saito, E. Miyoshi, K. Sasai, N. Nakano, H. Eguchi et al., A secreted type of beta 1,6-N-acetylglucosaminyltransferase V (GnT-V) induces tumor angiogenesis without mediation of glycosylation: A novel function of GnT-V distinct from the original glycosyltransferase activity, The Journal of Biological Chemistry, vol.277, issue.19, pp.17002-17008, 2002.

R. Sakakibara, M. Kato, N. Okamura, T. Nakagawa, Y. Komada et al., Characterization of a human placental fructose-6-phosphate, 2-kinase/fructose-2,6-bisphosphatase, Journal of Biochemistry, vol.122, issue.1, pp.122-128, 1997.

S. Salceda and J. Caro, Hypoxia-inducible factor 1alpha (HIF-1alpha) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes, The Journal of Biological Chemistry, vol.272, issue.36, pp.22642-22647, 1997.

G. Salvato, Quantitative and morphological analysis of the vascular bed in bronchial biopsy specimens from asthmatic and non-asthmatic subjects, Thorax, vol.56, issue.12, pp.902-906, 2001.

P. Salven, S. Mustjoki, R. Alitalo, K. Alitalo, and S. Rafii, VEGFR-3 and CD133 identify a population of CD34+ lymphatic/vascular endothelial precursor cells, Blood, vol.101, issue.1, pp.168-172, 2003.

N. Sawada, A. Jiang, F. Takizawa, A. Safdar, A. Manika et al., Endothelial PGC-1? mediates vascular dysfunction in diabetes, Cell Metabolism, vol.19, issue.2, pp.246-258, 2014.

M. Scartozzi, L. Vincent, M. Chiron, and S. Cascinu, Aflibercept, a new way to target angiogenesis in the second line treatment of metastatic colorectal cancer (mCRC), Targeted Oncology, vol.11, issue.4, pp.489-500, 2016.

H. Schachter, Biosynthetic controls that determine the branching and microheterogeneity of protein-bound oligosaccharides, Biochemistry and Cell Biology, vol.64, issue.3, pp.163-181, 1986.

H. Schachter, The joys of HexNAc. The synthesis and function of N-and O-glycan branches, Glycoconjugate Journal, vol.17, issue.7-9, pp.465-483, 2000.

E. D. Schleicher and C. Weigert, Role of the hexosamine biosynthetic pathway in diabetic nephropathy, Kidney International. Supplement, vol.77, pp.13-18, 2000.

G. W. Schmid-schönbein, Microlymphatics and lymph flow, Physiological Reviews, vol.70, issue.4, pp.987-1028, 1990.

S. Schoors, U. Bruning, R. Missiaen, K. C. Queiroz, G. Borgers et al., Fatty acid carbon is essential for dNTP synthesis in endothelial cells, Nature, vol.520, issue.7546, pp.192-197, 2015.

S. Schoors, K. De-bock, A. R. Cantelmo, M. Georgiadou, B. Ghesquière et al., Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis, Cell Metabolism, vol.19, issue.1, pp.37-48, 2014.

S. F. Schoppmann, P. Birner, J. Stöckl, R. Kalt, R. Ullrich et al., Tumorassociated macrophages express lymphatic endothelial growth factors and are related to peritumoral lymphangiogenesis, The American Journal of Pathology, vol.161, issue.3, pp.947-956, 2002.

T. Schwientek, M. Nomoto, S. B. Levery, G. Merkx, A. G. Van-kessel et al., Control of O-glycan branch formation. Molecular cloning of human cDNA encoding a novel beta1,6-N-acetylglucosaminyltransferase forming core 2 and core 4, The Journal of Biological Chemistry, vol.274, issue.8, pp.4504-4512, 1999.

T. Schwientek, J. C. Yeh, S. B. Levery, B. Keck, G. Merkx et al., Control of O-glycan branch formation. Molecular cloning and characterization of a novel thymus-associated core 2 beta1, 6-n-acetylglucosaminyltransferase, The Journal of Biological Chemistry, vol.275, issue.15, pp.11106-11113, 2000.

G. Seghezzi, S. Patel, C. J. Ren, A. Gualandris, G. Pintucci et al., Fibroblast growth factor-2 (FGF-2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: An autocrine mechanism contributing to angiogenesis, The Journal of Cell Biology, vol.141, issue.7, pp.1659-1673, 1998.

F. Seguin, M. A. Carvalho, D. C. Bastos, M. Agostini, K. G. Zecchin et al., The fatty acid synthase inhibitor orlistat reduces experimental metastases and angiogenesis in B16-F10 melanomas, British Journal of Cancer, vol.107, issue.6, pp.977-987, 2012.

G. L. Semenza, HIF-1: Mediator of physiological and pathophysiological responses to hypoxia, Journal of Applied Physiology, vol.88, issue.4, pp.1474-1480, 2000.

S. Sengupta, E. Gherardi, L. A. Sellers, J. M. Wood, R. Sasisekharan et al., Hepatocyte growth factor/scatter factor can induce angiogenesis independently of vascular endothelial growth factor, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.23, issue.1, pp.69-75, 2003.

B. Sennino and D. M. Mcdonald, Controlling escape from angiogenesis inhibitors, Nature Reviews. Cancer, vol.12, issue.10, pp.699-709, 2012.

W. Shi, B. A. Nacev, B. T. Aftab, S. Head, C. M. Rudin et al., Itraconazole side chain analogues: Structure-activity relationship studies for inhibition of endothelial cell proliferation, vascular endothelial growth factor receptor 2 (VEGFR2) glycosylation, and hedgehog signaling, Journal of Medicinal Chemistry, vol.54, issue.20, pp.7363-7374, 2011.

M. Shibuya, Vascular endothelial growth factor receptor-1 (VEGFR-1/Flt-1): A dual regulator for angiogenesis, Angiogenesis, vol.9, issue.4, pp.225-230, 2006.

M. Shibuya, Vascular endothelial growth factor and its receptor system: Physiological functions in angiogenesis and pathological roles in various diseases, Journal of Biochemistry, vol.153, issue.1, pp.13-19, 2013.

M. Shibuya, VEGF-VEGFR system as a target for suppressing inflammation and other diseases, Endocrine, Metabolic & Immune Disorders Drug Targets, vol.15, issue.2, pp.135-144, 2015.

M. Shibuya, S. Yamaguchi, A. Yamane, T. Ikeda, A. Tojo et al., Nucleotide sequence and expression of a novel human receptor-type tyrosine kinase gene (flt) closely related to the fms family, Oncogene, vol.5, issue.4, pp.519-524, 1990.

J. L. Shifren, J. F. Tseng, C. J. Zaloudek, I. P. Ryan, Y. G. Meng et al., Ovarian steroid regulation of vascular endothelial growth factor in the human endometrium: Implications for angiogenesis during the menstrual cycle and in the pathogenesis of endometriosis, The Journal of Clinical Endocrinology and Metabolism, vol.81, issue.8, pp.3112-3118, 1996.

K. Shirato, K. Nakajima, H. Korekane, S. Takamatsu, C. Gao et al., Hypoxic regulation of glycosylation via the N-acetylglucosamine cycle, Journal of Clinical Biochemistry and Nutrition, vol.48, issue.1, pp.20-25, 2011.

S. K. Shukla, V. Purohit, K. Mehla, V. Gunda, N. V. Chaika et al., MUC1 and HIF-1alpha signaling crosstalk induces anabolic glucose metabolism to impart gemcitabine resistance to pancreatic cancer, Cancer Cell, vol.32, issue.1, pp.71-87, 2017.
DOI : 10.1016/j.ccell.2017.06.004

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5533091

D. Shweiki, A. Itin, D. Soffer, and E. Keshet, Vascular endothelial growth factor induced by hypoxia may mediate hypoxia-initiated angiogenesis, Nature, vol.359, issue.6398, pp.843-845, 1992.
DOI : 10.1038/359843a0

J. Silvestre, D. M. Smadja, and B. I. Lévy, Postischemic revascularization: From cellular and molecular mechanisms to clinical applications, Physiological Reviews, vol.93, issue.4, pp.1743-1802, 2013.
DOI : 10.1152/physrev.00006.2013

B. Sitohy, J. A. Nagy, S. S. Jaminet, and H. F. Dvorak, Tumor-surrogate blood vessel subtypes exhibit differential susceptibility to anti-VEGF therapy, Cancer Research, vol.71, issue.22, pp.7021-7028, 2011.
DOI : 10.1158/0008-5472.can-11-1693

URL : http://cancerres.aacrjournals.org/content/71/22/7021.full.pdf

R. S. Srinivasan, X. Geng, Y. Yang, Y. Wang, S. Mukatira et al., The nuclear hormone receptor Coup-TFII is required for the initiation and early maintenance of Prox1 expression in lymphatic endothelial cells, Genes & Development, vol.24, issue.7, pp.696-707, 2010.

A. Stahl, J. G. Evans, S. Pattel, D. Hirsch, and H. F. Lodish, Insulin causes fatty acid transport protein translocation and enhanced fatty acid uptake in adipocytes, Developmental Cell, vol.2, issue.4, pp.477-488, 2002.
DOI : 10.1016/s1534-5807(02)00143-0

URL : https://doi.org/10.1016/s1534-5807(02)00143-0

P. Stanley, Galectin-1 pulls the strings on VEGFR2, Cell, vol.156, issue.4, pp.625-626, 2014.
DOI : 10.1016/j.cell.2014.01.059

URL : https://doi.org/10.1016/j.cell.2014.01.059

P. Stapor, X. Wang, J. Goveia, S. Moens, and P. Carmeliet, Angiogenesis revisitedrole and therapeutic potential of targeting endothelial metabolism, Journal of Cell Science, vol.127, pp.4331-4341, 2014.
DOI : 10.1242/jcs.153908

URL : http://jcs.biologists.org/content/127/20/4331.full.pdf

S. R. Stowell, T. Ju, and R. D. Cummings, Protein glycosylation in cancer, Annual Review of Pathology, vol.10, pp.473-510, 2015.
DOI : 10.1146/annurev-pathol-012414-040438

URL : http://europepmc.org/articles/pmc4396820?pdf=render

A. Stratmann, W. Risau, and K. H. Plate, Cell type-specific expression of angiopoietin-1 and angiopoietin-2 suggests a role in glioblastoma angiogenesis, Annual Review of Pathology, vol.153, issue.5, pp.1459-1466, 1998.

J. Su, J. Shih, M. Yen, Y. Jeng, C. Chang et al., Cyclooxygenase-2 induces EP1-and HER-2/Neu-dependent vascular endothelial growth factor-C up-regulation: A novel mechanism of lymphangiogenesis in lung adenocarcinoma, Cancer Research, vol.64, issue.2, pp.554-564, 2004.

Y. Sun, J. Wang, H. Li, L. Sun, Y. Wang et al., The effects of budesonide on angiogenesis in a murine asthma model, Archives of Medical Science, vol.9, issue.2, pp.361-367, 2013.

E. Sundlisaeter, A. Dicko, P. Ø. Sakariassen, K. Sondenaa, P. Ø. Enger et al., Lymphangiogenesis in colorectal cancer-prognostic and therapeutic aspects, International Journal of Cancer, vol.121, issue.7, pp.1401-1409, 2007.

S. Taghiloo, E. Allahmoradi, R. Ebadi, M. Tehrani, Z. Hosseini-khah et al., Upregulation of galectin-9 and PD-L1 immune checkpoints molecules in patients with chronic lymphocytic leukemia, Asian Pacific Journal of Cancer Prevention, vol.18, issue.8, pp.2269-2274, 2017.

E. B. Tahara, F. D. Navarete, and A. J. Kowaltowski, Tissue-, substrate-, and site-specific characteristics of mitochondrial reactive oxygen species generation, Free Radical Biology & Medicine, vol.46, issue.9, pp.1283-1297, 2009.

H. Takeuchi and R. S. Haltiwanger, Significance of glycosylation in notch signaling, Biochemical and Biophysical Research Communications, vol.453, issue.2, pp.235-242, 2014.

T. Tammela and K. Alitalo, Lymphangiogenesis: Molecular mechanisms and future promise, Cell, vol.140, issue.4, pp.460-476, 2010.
DOI : 10.1016/j.cell.2010.01.045

URL : https://doi.org/10.1016/j.cell.2010.01.045

K. Taniguchi, R. Kohno, T. Ayada, R. Kato, K. Ichiyama et al., Spreds are essential for embryonic lymphangiogenesis by regulating vascular endothelial growth factor receptor 3 signaling, Molecular and Cellular Biology, vol.27, issue.12, pp.4541-4550, 2007.
DOI : 10.1128/mcb.01600-06

URL : https://mcb.asm.org/content/27/12/4541.full.pdf

R. N. Taylor, J. Yu, P. B. Torres, A. C. Schickedanz, J. K. Park et al., Mechanistic and therapeutic implications of angiogenesis in endometriosis, Reproductive Sciences, vol.16, issue.2, pp.140-146, 2009.

S. Telang, B. F. Clem, A. C. Klarer, A. L. Clem, J. O. Trent et al., Small molecule inhibition of 6-phosphofructo-2-kinase suppresses t cell activation, Journal of Translational Medicine, vol.10, p.95, 2012.

B. I. Terman, M. E. Carrion, E. Kovacs, B. A. Rasmussen, R. L. Eddy et al., Identification of a new endothelial cell growth factor receptor tyrosine kinase, Oncogene, vol.6, issue.9, pp.1677-1683, 1991.

B. I. Terman, S. Jani-sait, M. E. Carrion, and T. B. Shows, The KDR gene maps to human chromosome 4q31.2--q32, a locus which is distinct from locations for other type III growth factor receptor tyrosine kinases, Cytogenet. Cell Genet, vol.60, issue.3-4, pp.214-215, 1992.

L. Teuwen, V. Geldhof, and P. Carmeliet, How glucose, glutamine and fatty acid metabolism shape blood and lymph vessel development, Dev. Biol, issue.17, pp.30495-30500, 2017.
DOI : 10.1016/j.ydbio.2017.12.001

H. Thiagarajan, U. Thiyagamoorthy, I. Shanmugham, G. Dharmalingam-nandagopal, and A. Kaliyaperumal, Angiogenic growth factors in myocardial infarction: A critical appraisal, Heart Failure Reviews, vol.22, issue.6, pp.665-683, 2017.

V. L. Thijssen and A. W. Griffioen, Galectin-1 and -9 in angiogenesis: A sweet couple, Glycobiology, vol.24, issue.10, pp.915-920, 2014.

V. L. Thijssen, G. A. Rabinovich, and A. W. Griffioen, Vascular galectins: Regulators of tumor progression and targets for cancer therapy, Cytokine & Growth Factor Reviews, vol.24, issue.6, pp.547-558, 2013.

V. L. Thijssen, R. Postel, R. J. Brandwijk, R. P. Dings, I. Nesmelova et al., Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy, Proceedings of the National Academy of Sciences of the United States of America, vol.103, issue.43, pp.15975-15980, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00109326

D. D. Thomas, L. A. Ridnour, J. S. Isenberg, W. Flores-santana, C. H. Switzer et al., The chemical biology of nitric oxide: Implications in cellular signaling, Free Radical Biology & Medicine, vol.45, issue.1, pp.18-31, 2008.

G. Thurston, Role of angiopoietins and tie receptor tyrosine kinases in angiogenesis and lymphangiogenesis, Cell and Tissue Research, vol.314, issue.1, pp.61-68, 2003.

Q. Tong, L. Zheng, B. Li, D. Wang, C. Huang et al., Hypoxia-induced mitogenic factor enhances angiogenesis by promoting proliferation and migration of endothelial cells, Experimental Cell Research, vol.312, issue.18, pp.3559-3569, 2006.

G. M. Tozer, V. E. Prise, J. Wilson, M. Cemazar, S. Shan et al., Mechanisms associated with tumor vascular shut-down induced by combretastatin A-4 phosphate: Intravital microscopy and measurement of vascular permeability, Cancer Research, vol.61, issue.17, pp.6413-6422, 2001.

L. Treps, L. Conradi, U. Harjes, and P. Carmeliet, Manipulating angiogenesis by targeting endothelial metabolism: Hitting the engine rather than the drivers-a new perspective?, Pharmacol. Rev, vol.68, issue.3, pp.872-887, 2016.

M. Tunyogi-csapo, T. Koreny, C. Vermes, J. O. Galante, J. J. Jacobs et al., Role of fibroblasts and fibroblast-derived growth factors in periprosthetic angiogenesis, Journal of Orthopaedic Research, vol.25, issue.10, pp.1378-1388, 2007.

T. T. Tuomisto, T. T. Rissanen, I. Vajanto, A. Korkeela, J. Rutanen et al., HIF-VEGF-VEGFR-2, TNF-alpha and IGF pathways are upregulated in critical human skeletal muscle ischemia as studied with DNA array, Atherosclerosis, vol.174, issue.1, pp.111-120, 2004.

R. S. Udan, J. C. Culver, and M. E. Dickinson, Understanding vascular development, Wiley Interdisciplinary Reviews: Developmental Biology, vol.2, issue.3, pp.327-346, 2013.

Y. Urasaki, L. Heath, and C. W. Xu, Coupling of glucose deprivation with impaired histone H2B monoubiquitination in tumors, PLoS One, vol.7, issue.5, p.36775, 2012.

I. Valle, A. Alvarez-barrientos, E. Arza, S. Lamas, and M. Monsalve, PGC-1alpha regulates the mitochondrial antioxidant defense system in vascular endothelial cells, Cardiovascular Research, vol.66, issue.3, pp.562-573, 2005.

G. G. Van-den-eynden, I. Van-der-auwera, S. J. Van-laere, X. B. Trinh, C. G. Colpaert et al., Comparison of molecular determinants of angiogenesis and lymphangiogenesis in lymph node metastases and in primary tumours of patients with breast cancer, The Journal of Pathology, vol.213, issue.1, pp.56-64, 2007.

E. Van-schaftingen, B. Lederer, R. Bartrons, and H. G. Hers, A kinetic study of pyrophosphate: Fructose-6-phosphate phosphotransferase from potato tubers. Application to a microassay of fructose 2,6-bisphosphate, European Journal of Biochemistry, vol.129, issue.1, pp.191-195, 1982.

M. Van-woensel, T. Mathivet, N. Wauthoz, R. Rosière, A. D. Garg et al., Sensitization of glioblastoma tumor micro-environment to chemo-and immunotherapy by Galectin-1 intranasal knock-down strategy, Scientific Reports, vol.7, issue.1, p.1217, 2017.

S. Vandekeere, M. Dewerchin, and P. Carmeliet, Angiogenesis revisited: An overlooked role of endothelial cell metabolism in vessel sprouting, Microcirculation, vol.22, issue.7, pp.509-517, 2015.

M. G. Vander-heiden, L. C. Cantley, and C. B. Thompson, Understanding the Warburg effect: The metabolic requirements of cell proliferation, Science, vol.324, issue.5930, pp.1029-1033, 2009.

D. L. Vaux, Apoptogenic factors released from mitochondria, Biochimica et Biophysica Acta, vol.1813, issue.4, pp.546-550, 2011.

D. Verdegem, S. Moens, P. Stapor, and P. Carmeliet, Endothelial cell metabolism: Parallels and divergences with cancer cell metabolism. Cancer Metab. 2, 19, Oncotarget, vol.9, issue.1, pp.1380-1402, 2014.

C. De-vries, J. A. Escobedo, H. Ueno, K. Houck, N. Ferrara et al., The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor, Science, vol.255, issue.5047, pp.989-991, 1992.

G. L. Wang, B. H. Jiang, E. A. Rue, and G. L. Semenza, Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension, Proceedings of the National Academy of Sciences of the United States of America, vol.92, issue.12, pp.5510-5514, 1995.

J. Wang, D. Ling, L. Tu, P. Van-wijngaarden, G. J. Dusting et al., Gene therapy for diabetic retinopathy: Are we ready to make the leap from bench to bedside?, Pharmacology & Therapeutics, vol.173, pp.1-18, 2017.

M. Wang and R. J. Kaufman, The impact of the endoplasmic reticulum proteinfolding environment on cancer development, Nature Reviews. Cancer, vol.14, issue.9, pp.581-597, 2014.

Q. Wang, B. Liang, N. A. Shirwany, and M. Zou, 2-deoxy-D-glucose treatment of endothelial cells induces autophagy by reactive oxygen species-mediated activation of the AMP-activated protein kinase, PLoS One, vol.6, issue.2, p.17234, 2011.

X. Wang, H. He, H. Zhang, W. Chen, Y. Ji et al., Clinical and prognostic implications of ?1, 6-N-acetylglucosaminyltransferase V in patients with gastric cancer, Cancer Science, vol.104, issue.2, pp.185-193, 2013.

O. Warburg, On the origin of cancer cells, Science, vol.123, issue.3191, pp.309-314, 1956.

A. G. Warren, H. Brorson, L. J. Borud, and S. A. Slavin, Lymphedema: A comprehensive review, Annals of Plastic Surgery, vol.59, issue.4, pp.464-472, 2007.

K. Watanabe, Y. Ohta, K. Toba, Y. Ogawa, H. Hanawa et al., Myocardial CD36 expression and fatty acid accumulation in patients with type I and II CD36 deficiency, Annals of Nuclear Medicine, vol.12, issue.5, pp.261-266, 1998.

K. Watanabe, H. Wakabayashi, J. H. Veerkamp, T. Ono, and T. Suzuki, Immunohistochemical distribution of heart-type fatty acid-binding protein immunoreactivity in normal human tissues and in acute myocardial infarct, The Journal of Pathology, vol.170, issue.1, pp.59-65, 1993.

E. C. Watson, Z. L. Grant, and L. Coultas, Endothelial cell apoptosis in angiogenesis and vessel regression, Cellular and Molecular Life Sciences, vol.74, issue.24, pp.4387-4403, 2017.

K. Wdowiak, T. Francuz, E. Gallego-colon, N. Ruiz-agamez, M. Kubeczko et al., Galectin targeted therapy in oncology: Current knowledge and perspectives, International Journal of Molecular Sciences, vol.19, issue.1, 2018.

X. Wei, J. G. Schneider, S. M. Shenouda, A. Lee, D. A. Towler et al., De novo lipogenesis maintains vascular homeostasis through endothelial nitric-oxide synthase (eNOS) palmitoylation, The Journal of Biological Chemistry, vol.286, issue.4, pp.2933-2945, 2011.

K. E. Wellen and C. B. Thompson, Cellular metabolic stress: Considering how cells respond to nutrient excess, Molecular Cell, vol.40, issue.2, pp.323-332, 2010.

C. Wheeler-jones, R. Abu-ghazaleh, R. Cospedal, R. A. Houliston, J. Martin et al., Vascular endothelial growth factor stimulates prostacyclin production and activation of cytosolic phospholipase A2 in endothelial cells via p42/p44 mitogen-activated protein kinase, FEBS Letters, vol.420, issue.1, pp.28-32, 1997.

W. R. Wilson and M. P. Hay, Targeting hypoxia in cancer therapy, Nature Reviews. Cancer, vol.11, issue.6, pp.393-410, 2011.

J. Wilting, M. Papoutsi, K. Buttler, and J. Becker, Embryonic development of the lymphovascular system and tumor lymphangiogenesis, Cancer Treatment and Research, vol.135, pp.17-24, 2007.

J. Wilting, M. Papoutsi, B. Christ, K. H. Nicolaides, C. S. Von-kaisenberg et al., The transcription factor Prox1 is a marker for lymphatic endothelial cells in normal and diseased human tissues, The FASEB Journal, vol.16, issue.10, pp.1271-1273, 2002.

B. W. Wong, X. Wang, A. Zecchin, B. Thienpont, I. Cornelissen et al., The role of fatty acid ?-oxidation in lymphangiogenesis, Nature, vol.542, issue.7639, pp.49-54, 2017.

G. L. Wright, I. G. Maroulakou, J. Eldridge, T. L. Liby, V. Sridharan et al., VEGF stimulation of mitochondrial biogenesis: Requirement of AKT3 kinase, The FASEB Journal, vol.22, issue.9, pp.3264-3275, 2008.

C. Wu, K. Feng, D. Lu, D. Yan, T. Han et al., Reproductive toxicities caused by swainsonine from locoweed in mice, BioMed Research International, p.6824374, 2016.

J. Wu, L. Shen, L. Qiu, Q. Duan, Z. Luo et al., Reversal effect of GnT-V on the radioresistance of human nasopharyngeal carcinoma cells by alteration ?1, 6-GlcNAc branched N-glycans, International Journal of Clinical and Experimental Pathology, vol.8, issue.9, pp.9901-9911, 2015.

M. Xaymardan, J. Zheng, I. Duignan, A. Chin, J. M. Holm et al., Senescent impairment in synergistic cytokine pathways that provide rapid cardioprotection in the rat heart, The Journal of Experimental Medicine, vol.199, issue.6, pp.797-804, 2004.

D. Xu, M. M. Fuster, R. Lawrence, and J. D. Esko, Heparan sulfate regulates VEGF165-and VEGF121-mediated vascular hyperpermeability, The Journal of Biological Chemistry, vol.286, issue.1, pp.737-745, 2011.

H. Xu, P. Czerwinski, M. Hortmann, H. Sohn, U. Förstermann et al., Protein kinase C alpha promotes angiogenic activity of human endothelial cells via induction of vascular endothelial growth factor, Cardiovascular Research, vol.78, issue.2, pp.349-355, 2008.

H. Xu, J. Edwards, S. Banerji, R. Prevo, D. G. Jackson et al., Distribution of lymphatic vessels in normal and arthritic human synovial tissues, Annals of the Rheumatic Diseases, vol.62, issue.12, pp.1227-1229, 2003.

Y. Xu, X. An, X. Guo, T. G. Habtetsion, Y. Wang et al., Endothelial PFKFB3 plays a critical role in angiogenesis, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.34, issue.6, pp.1231-1239, 2014.

A. Yalcin, B. F. Clem, Y. Imbert-fernandez, S. C. Ozcan, S. Peker et al., 6-Phosphofructo-2-kinase (PFKFB3) promotes cell cycle progression and suppresses apoptosis via Cdk1-mediated phosphorylation of p27, Cell Death & Disease, vol.5, 1337.

H. Yamamoto, J. Swoger, S. Greene, T. Saito, J. Hurh et al., Beta1,6-N-acetylglucosamine-bearing N-glycans in human gliomas: Implications for a role in regulating invasivity, vol.60, pp.134-142, 2000.

C. Yang, K. Matsuura, N. Huang, A. C. Robeson, B. Huang et al., Fatty acid synthase inhibition engages a novel caspase-2 regulatory mechanism to induce ovarian cancer cell death, Oncogene, vol.34, issue.25, pp.3264-3272, 2015.

Y. Yang and B. Yu, Recent advances in the chemical synthesis of C-glycosides, Chemical Reviews, vol.117, issue.19, pp.12281-12356, 2017.

W. Yeh, C. Lin, and W. Fu, Enhancement of glucose transporter expression of brain endothelial cells by vascular endothelial growth factor derived from glioma exposed to hypoxia, Molecular Pharmacology, vol.73, issue.1, pp.170-177, 2008.

S. Ylä-herttuala and K. Alitalo, Gene transfer as a tool to induce therapeutic vascular growth, Nature Medicine, vol.9, issue.6, pp.694-701, 2003.

J. Yoo, E. H. Mashalidis, A. C. Kuk, K. Yamamoto, B. Kaeser et al., GlcNAc-1-P-transferase-tunicamycin complex structure reveals basis for inhibition of N-glycosylation, Nature Structural & Molecular Biology, vol.25, issue.3, pp.217-224, 2018.

H. S. Young, A. M. Summers, M. Bhushan, P. E. Brenchley, and C. E. Griffiths, Single-nucleotide polymorphisms of vascular endothelial growth factor in psoriasis of early onset, The Journal of Investigative Dermatology, vol.122, issue.1, pp.209-215, 2004.

C. Yu, X. Liang, S. Lipsky, C. Karaaslan, H. Kozakewich et al., Dual role of fatty acid-binding protein 5 on endothelial cell fate: A potential link between lipid metabolism and angiogenic responses, Angiogenesis, vol.19, issue.1, pp.95-106, 2016.

W. Yu, N. Zhang, and C. Li, Saccharide modified pharmaceutical nanocarriers for targeted drug and gene delivery, Current Pharmaceutical Design, vol.15, pp.3826-3836, 2009.

I. Zachary, VEGF signalling: Integration and multi-tasking in endothelial cell biology, Biochemical Society Transactions, vol.31, pp.1171-1177, 2003.

J. C. Zampell, S. Elhadad, T. Avraham, E. Weitman, S. Aschen et al., Tolllike receptor deficiency worsens inflammation and lymphedema after lymphatic injury, Am. J. Physiol. Cellular Physiology, vol.302, issue.4, pp.709-719, 2012.
DOI : 10.1152/ajpcell.00284.2011

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3287358/pdf

Y. Y. Zaytseva, V. A. Elliott, P. Rychahou, W. C. Mustain, J. T. Kim et al., Cancer cell-associated fatty acid synthase activates endothelial cells and promotes angiogenesis in colorectal cancer, Carcinogenesis, vol.35, issue.6, pp.1341-1351, 2014.

J. Zhan, Y. Li, J. Yu, Y. Zhao, W. Cao et al., Culture medium of bone marrow-derived human mesenchymal stem cells effects lymphatic endothelial cells and tumor lymph vessel formation, Oncology Letters, vol.9, issue.3, pp.1221-1226, 2015.

D. Zhang, B. Li, J. Shi, L. Zhao, X. Zhang et al., Suppression of tumor growth and metastasis by simultaneously blocking vascular endothelial growth factor (VEGF)-a and VEGF-C with a receptor-immunoglobulin fusion protein, Cancer Research, vol.70, issue.6, pp.2495-2503, 2010.

L. Zhang, Z. Liu, K. Yang, C. Kong, C. Liu et al., Tumor progression of non-small cell lung cancer controlled by albumin and micellar nanoparticles of itraconazole, a multitarget angiogenesis inhibitor, Molecular Pharmaceutics, vol.14, issue.12, pp.4705-4713, 2017.

L. Zhang and K. G. Hagen, Pleiotropic effects of O-glycosylation in colon cancer, The J. Biol. Chem, vol.293, issue.4, pp.1315-1316, 2018.

Y. Zhang, Y. Su, O. V. Volpert, and G. F. Vande-woude, Hepatocyte growth factor/scatter factor mediates angiogenesis through positive VEGF and negative thrombospondin 1 regulation, Proceedings of the National Academy of Sciences of the United States of America, vol.100, issue.22, pp.12718-12723, 2003.

Q. Zhao, X. Shen, W. Zhang, G. Zhu, J. Qi et al., Mice with increased angiogenesis and osteogenesis due to conditional activation of HIF pathway in osteoblasts are protected from ovariectomy induced bone loss, Bone, vol.50, issue.3, pp.763-770, 2012.

Y. Zhao, J. Neuzil, and K. Wu, Vitamin E analogues as mitochondria-targeting compounds: From the bench to the bedside?, Molecular Nutrition & Food Research, vol.53, issue.1, pp.129-139, 2009.

W. Zheng, T. Tammela, M. Yamamoto, A. Anisimov, T. Holopainen et al., Notch restricts lymphatic vessel sprouting induced by vascular endothelial growth factor, Blood, vol.118, issue.4, pp.1154-1162, 2011.

D. Zhong, L. Xiong, T. Liu, X. Liu, X. Liu et al., The glycolytic inhibitor 2-deoxyglucose activates multiple prosurvival pathways through IGF1R, The Journal of Biological Chemistry, vol.284, issue.35, pp.23225-23233, 2009.

W. Zhu, L. Ye, J. Zhang, P. Yu, H. Wang et al., PFK15, a small molecule inhibitor of PFKFB3, induces cell cycle arrest, apoptosis and inhibits invasion in gastric cancer, PLoS One, vol.11, issue.9, p.163768, 2016.

J. E. Ziello, I. S. Jovin, and Y. Huang, Hypoxia-inducible factor (HIF)-1 regulatory pathway and its potential for therapeutic intervention in malignancy and ischemia, The Yale Journal of Biology and Medicine, vol.80, issue.2, pp.51-60, 2007.

Y. Zou, S. Zeng, M. Huang, Q. Qiu, Y. Xiao et al., Inhibition of 6-phosphofructo-2-kinase suppresses fibroblast-like synoviocytes-mediated synovial inflammation and joint destruction in rheumatoid arthritis, British Journal of Pharmacology, vol.174, issue.9, pp.893-908, 2017.

, Phostine 3.1a as a pharmacological compound with anti-angiogenic properties against diseases with excess vascularization

, Raffaella Soleti 1 , Luisa Vergori 1 , Grégory Hilairet 1 , Morgane Le Mao 2 , Naig Gueguen 2 , Salim Khiati 2 , Ludovic Clarion 4 , Norbert Bakalara 5 , Stéphane Germain 3* , Guy Lenaers 2* , Ramaroson Andriantsitohaina 1 ?* 1. INSERM UMR1063, Stress Oxydant et Pathologies Métaboliques

C. Mitolab and . Umr, INSERM U 1083, vol.6015

, SAS, p.34090

. Inserm-u-1051, * Co-last authors ? Corresponding author: Ramaroson Andriantsitohaina, INSERM UMR1063, Stress Oxydant et Pathologies Métaboliques, p.49933

F. Angers, Phone: +33 2 44 68 85 80. E-mail: ramaroson.andriantsitohaina@univ-angers.fr Running Title: Anti-angiogenic properties of PST 3.1a. added at 4 °C overnight to allow antibody interaction with VEGFR2 or Gal-1 protein

, Subsequently, the tubes containing the immunoprecipitates were centrifuged, and the supernatants were eliminated. Pellets were washed two times with 100 µL lysis buffer diluted to 1:5 with PBS

, Gel-loading bugger (15 µL) was then added to the pellets. The tubes were heated for 10 min at 70

, The gel was trans-blotted to perform Western analysis for VEGFR2 and Gal-1 proteins as described above. Data analysis Data are represented as mean ± SEM, °C, after which proteins were separated on 4-12% SDS-PAGE gel

, Statistical analyses were performed either using one-way ANOVA (parametric) for all in vitro angiogenesis experiments, or using Mann-Whitney U-tests (non-parametric) for other experiments, using Prism software package 5.00 (GraphPAD Software, CA)

I. Dimova, G. Popivanov, and V. Djonov, Angiogenesis in cancer -general pathways and their therapeutic implications, J BUON, vol.19, issue.1, pp.15-21, 2014.

G. Eelen, P. De-zeeuw, L. Treps, U. Harjes, B. W. Wong et al., Endothelial Cell Metabolism. Physiol Rev, vol.98, issue.1, pp.3-58, 2018.

L. Treps, L. C. Conradi, U. Harjes, and P. Carmeliet, Manipulating Angiogenesis by Targeting Endothelial Metabolism: Hitting the Engine Rather than the Drivers-A New Perspective?, Pharmacol Rev, vol.68, issue.3, pp.872-887, 2016.

D. Bock, K. Georgiadou, M. Carmeliet, and P. , Role of endothelial cell metabolism in vessel sprouting, Cell Metab, vol.18, issue.5, pp.634-647, 2013.

P. Stapor, X. Wang, J. Goveia, S. Moens, and P. Carmeliet, Angiogenesis revisited -role and therapeutic potential of targeting endothelial metabolism, J Cell Sci, vol.127, pp.4331-4341, 2014.

D. O. Croci, J. P. Cerliani, T. Dalotto-moreno, S. P. Méndez-huergo, I. D. Mascanfroni et al., Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors, Cell, vol.156, issue.4, pp.744-758, 2014.

G. Bergers and D. Hanahan, Modes of resistance to anti-angiogenic therapy, Nat Rev Cancer, vol.8, issue.8, pp.592-603, 2008.

A. Mcintyre and A. L. Harris, Metabolic and hypoxic adaptation to anti-angiogenic therapy: a target for induced essentiality, EMBO Mol Med, vol.7, issue.4, pp.368-379, 2015.

U. Harjes, J. Kalucka, and P. Carmeliet, Targeting fatty acid metabolism in cancer and endothelial cells, Crit Rev Oncol Hematol, vol.97, pp.15-21, 2016.

M. Potente and P. Carmeliet, The Link Between Angiogenesis and Endothelial Metabolism

, Annu Rev Physiol, vol.79, pp.43-66, 2017.

S. Schoors, D. Bock, K. Cantelmo, A. R. Georgiadou, M. Ghesquière et al., Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis, Cell Metab, vol.19, issue.1, pp.37-48, 2014.

A. R. Cantelmo, L. C. Conradi, A. Brajic, J. Goveia, J. Kalucka et al., Inhibition of the Glycolytic Activator PFKFB3 in Endothelium Induces Tumor Vessel Normalization, Impairs Metastasis, and Improves Chemotherapy, Cancer Cell, vol.30, issue.6, pp.968-985, 2016.

R. Benedito, C. Roca, I. Sörensen, S. Adams, A. Gossler et al., The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis, Cell, vol.137, issue.6, pp.1124-1135, 2009.

A. I. Markowska, K. C. Jefferies, and N. Panjwani, Galectin-3 protein modulates cell surface expression and activation of vascular endothelial growth factor receptor 2 in human endothelial cells, J Biol Chem, vol.286, issue.34, pp.29913-29921, 2011.

M. T. Elola, A. G. Blidner, F. Ferragut, C. Bracalente, and G. A. Rabinovich, Assembly, organization and regulation of cell-surface receptors by lectin-glycan complexes

, Biochem J, vol.469, issue.1, pp.1-16, 2015.

Y. Liu, H. Liu, W. Liu, W. Zhang, H. An et al., 6-N-acetylglucosaminyltransferase V predicts recurrence and survival of patients with clear-cell renal cell carcinoma after surgical resection, World J Urol, vol.1, issue.11, pp.1791-1799, 2015.

Z. Hassani, A. Saleh, S. Turpault, S. Khiati, W. Morelle et al., , vol.3, p.1

. Targets, Inhibits Glioblastoma-Initiating Cell Invasiveness and Proliferation, Mol Canc Res, vol.15, issue.10, pp.1376-1387, 2017.

A. W. Griffioen and V. L. Thijssen, Galectins in tumor angiogenesis, Ann Transl Med, vol.2, issue.9, p.90, 2014.

L. Clarion, C. Jacquard, O. Sainte-catherine, S. Loiseau, D. Filippini et al., Oxaphosphinanes: new therapeutic perspectives for glioblastoma, J Med Chem, vol.55, issue.5, pp.2196-2211, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00677707

L. Clarion, C. Jacquard, O. Sainte-catherine, M. Decoux, S. Loiseau et al., Cglycoside mimetics inhibit glioma stem cell proliferation, migration, and invasion, J Med Chem, vol.57, issue.20, pp.8293-8306, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01100620

S. A. Valdés-rives, D. Casique-aguirre, L. Germán-castelán, and M. A. Velasco-velázquez, González-Arenas A. Apoptotic Signaling Pathways in Glioblastoma and Therapeutic Implications, Biomed Res Int, p.7403747, 2017.

A. S. Arbab, M. H. Rashid, K. Angara, T. F. Borin, P. C. Lin et al., Major Challenges and Potential Microenvironment-Targeted Therapies in Glioblastoma, Int J Mol Sci, vol.18, issue.12, 2017.

R. Soleti, T. Benameur, C. Porro, M. A. Panaro, R. Andriantsitohaina et al., Microparticles harboring Sonic Hedgehog promote angiogenesis through the upregulation of adhesion proteins and proangiogenic factors, Carcinogenesis, vol.30, issue.4, pp.580-588, 2009.

M. Bignon, C. Pichol-thievend, J. Hardouin, M. Malbouyres, N. Bréchot et al.,

, Lysyl oxidase-like protein-2 regulates sprouting angiogenesis and type IV collagen assembly in the endothelial basement membrane, Blood, vol.118, issue.14, pp.3979-89, 2011.

A. Yalcin, B. F. Clem, A. Simmons, A. Lane, K. Nelson et al., Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases, J Biol Chem, vol.284, issue.36, pp.24223-24232, 2009.

L. Duluc, C. Jacques, R. Soleti, F. Iacobazzi, G. Simard et al., Modulation of mitochondrial capacity and angiogenesis by red wine polyphenols via estrogen receptor, NADPH oxidase and nitric oxide synthase pathways, Int J Biochem Cell Biol, vol.45, issue.4, pp.783-791, 2013.

A. Yalcin, B. F. Clem, Y. Imbert-fernandez, S. C. Ozcan, S. Peker et al., 6-Phosphofructo-2-kinase (PFKFB3) promotes cell cycle progression and suppresses apoptosis via Cdk1-mediated phosphorylation of p27, Cell Death Dis, vol.5, p.1337, 2014.

J. R. Merchan, K. Kovács, J. W. Railsback, M. Kurtoglu, Y. Jing et al., Antiangiogenic activity of 2-deoxy-D-glucose, PLoS One, vol.5, issue.10, 2010.

P. Stanley, Galectin-1 Pulls the Strings on VEGFR2, Cell, vol.156, issue.4, pp.625-626, 2014.

S. P. Méndez-huergo, A. G. Blidner, and G. A. Rabinovich, Galectins: emerging regulatory checkpoints linking tumor immunity and angiogenesis, Curr Opin Immunol, vol.45, pp.8-15, 2017.

Q. T. Le, G. Shi, H. Cao, D. W. Nelson, Y. Wang et al., Galectin-1: a link between tumor hypoxia and tumor immune privilege, J Clin Oncol, vol.23, issue.35, pp.8932-8941, 2005.

K. Ito, S. A. Scott, S. Cutler, L. F. Dong, J. Neuzil et al., Thiodigalactoside inhibits murine cancers by concurrently blocking effects of galectin-1 on immune dysregulation, angiogenesis and protection against oxidative stress, Angiogenesis, vol.14, issue.3, pp.293-307, 2011.

K. Ito, K. Stannard, E. Gabutero, A. M. Clark, S. Y. Neo et al., Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment, Cancer Metastasis Rev, vol.31, issue.3-4, pp.763-778, 2012.

K. B. Chandler, D. R. Leon, R. D. Meyer, N. Rahimi, and C. E. Costello, Site-Specific NGlycosylation of Endothelial Cell Receptor Tyrosine Kinase VEGFR-2, J Proteome Res, vol.16, issue.2, pp.677-688, 2017.

, Suppl Fig 2: PST 3.1a does not disrupt zebrafish heartbeats

Z. Tg, dsRed) eggs were exposed or not to PST 3.1a at 50, 72 and 100 µM for 72, 96 or 120 hpf. A. Representative pictures showing Zebrafish embryos heartbeats (see Video Suppl Fig. 2 for corresponding videos) in the absence or in the presence of PST 3.1a. B. Quantitative measurement of zebrafish heartbeats was reported as number of heartbeats per min

, Lors de cette étude, nous avons dans un premier temps caractérisé les propriétés anti-angiogéniques de PST 3.1a, in vitro puis in vivo. Nous avons également démontré que PST 3.1a réduit la vascularisation tumorale et la progression du GBM. Dans un second temps, nous nous sommes intéressés aux perturbations métaboliques endothéliales mises en jeu. Nos résultats prouvent que PST 3.1a réduit l'expression de PFKFB3, enzyme clé pour réguler la glycolyse et la différentiation des CEs lors de l'angiogenèse. De plus, Ce travail de thèse a donc porté sur l'implication du glycomimétique PST 3.1a sur l'angiogenèse physiologique et tumorale, ainsi que sur le métabolisme endothélial associé

. Gnt-v-qui-régule-la-n-glycosylation,

. Jain, est une étape essentielle pour la croissance tumorale, l'invasivité et la dissémination métastatique. Bien qu'ils soient principalement utilisés en tant qu'adjuvants à la chimio-et radiothérapie, les traitements anti-angiogéniques représentent pourtant un enjeu majeur dans le cadre des thérapies anti-tumorales. Parmi les tumeurs, le GBM est caractérisé d'un point de vue physiopathologique par son caractère très invasif et hautement angiogénique, L'excès d'angiogenèse est dorénavant bien décrit comme étant impliqué dans la genèse L'angiogenèse

G. Dans-le-cadre-du and . Brandes, 2017) font état d'une activité anti-tumorale in vivo de PST 3.1a et nos résultats le confirment. De façon intéressante, PST 3.1a n'induit pas de mortalité ni de perte de poids chez les souris traitées avec 15 mg/kg pendant 14 jours. La même observation est effectuée à très forte dose chez les souris (900 mg/kg pendant 28 jours, non publié), ainsi que chez le zebrafish aux doses utilisées (100 µM pendant 5 jours). Dans cette étude, nous démontrons en plus que PST 3.1a réduit fortement l'angiogenèse tumorale, de presque 50 %. Même si nos résultats ne représentent pas l'effet de PST 3.1a sur les autres modes de vascularisation du GBM, l'angiogenèse représente le principal moteur de la vascularisation du GBM et PST 3.1a la réduit de manière significative. Ces résultats sont particulièrement intéressants car le microenvironnement vasculaire tumoral est très différent du microenvironnement vasculaire physiologique ; les vaisseaux tumoraux sont décrits comme tortueux, instables, peu perméables, hétérogènes, et les CEs qui les composent possèdent de grandes et lâches jonctions inter-cellulaires, les traitements anti-angiogéniques actuels reposent principalement sur l'utilisation du bevacizumab, mais il provoque une forte toxicité chez les patients, 2011.

. Vandekeere, Une seconde importante limite à l'utilisation du bevacizumab concerne son efficacité nouveau paradigme anti-angiogénique, 2015.

C. Ces, ;. Stratégie-vise-Également-les-cts, and . Verdegem, Ces deux types cellulaires sont décrits comme « brothers in arms, et possèdent plusieurs similitudes métaboliques. Le blocage de ces voies métaboliques communes est stratégique pour les futures thérapies antique l'angiopoïetine like 4, 2006.

. Missiaen, Comme décrit précédemment, un des principaux mécanismes de résistance tumorale vise à augmenter l'interaction VEGFR2 et Gal-1. Afin de contrer les thérapies actuelles, certaines tumeurs mettent en place un autre mécanisme de résistance, en stimulant la sécrétion d'autres facteurs pro-angiogéniques et notamment le FGF, pour favoriser la vascularisation par d'autres voies indépendantes, Une seconde perspective potentielle viserait à étudier l'impact de PST 3.1a sur la voie du FGF, 2004.

. Enfin, une dernière perspective conduirait à étudier l'implication de PST 3.1a sur d'autres pathologies caractérisées par une angiogenèse excessive (Carmeliet, 2003.

. Aterido, En particulier, il a récemment été proposé que la surexpression de GnT-V pourrait être corrélée avec la pathogenèse du psoriasis, 2016.

T. Andre, E. Chastre, L. Kotelevets, J. C. Vaillant, C. Louvet et al., , 1998.

, La Revue De Medecine Interne, vol.19, issue.12, pp.904-913

A. K. Anand, A. R. Chaudhory, H. N. Aggarwal, P. K. Sachdeva, P. S. Negi et al., , 2012.

, Survival outcome and neurotoxicity in patients of high-grade gliomas treated with conformal radiation and temozolamide, J Cancer Res Ther, vol.8, issue.1, pp.50-56

T. Asahara, H. Masuda, T. Takahashi, C. Kalka, C. Pastore et al., Isolation of putative progenitor endothelial cells for angiogenesis, Science, vol.275, issue.5302, pp.964-971, 1997.

A. Aterido, A. Julià, C. Ferrándiz, L. Puig, E. Fonseca et al., GenomeWide Pathway Analysis Identifies Genetic Pathways Associated with Psoriasis, J Invest Dermatol, vol.136, issue.3, pp.593-602, 2016.

R. Babouri, M. Rolland, O. Sainte-catherine, Z. Kabouche, M. Lecouvey et al., , 2015.

, Halogenated oxaphosphinanes: Synthesis, unexpected reactions and evaluation as inhibitors of cancer cell proliferation, Eur J Med Chem, vol.104, pp.33-41

G. Bergers and D. Hanahan, Modes of resistance to anti-angiogenic therapy, Nat Rev Cancer, vol.8, issue.8, pp.592-603, 2008.

A. A. Brandes, M. Bartolotti, A. Tosoni, R. Poggi, and E. Franceschi, Practical Management of Bevacizumab-Related Toxicities in Glioblastoma, Oncologist, vol.20, issue.2, pp.166-175, 2015.

M. Bredel, D. M. Scholtens, A. K. Yadav, A. A. Alvarez, J. J. Renfrow et al., , 2011.

, NFKBIA deletion in glioblastomas, The New England Journal of Medicine, vol.364, issue.7, pp.627-637

S. Brem, R. Cotran, and J. Folkman, Tumor angiogenesis: a quantitative method for histologic grading, J Natl Cancer Inst, vol.48, issue.2, pp.347-56, 1972.

P. C. Burger, T. Shibata, and P. Kleihues, The use of the monoclonal antibody Ki-67 in the identification of proliferating cells: application to surgical neuropathology, Am J Surg Pathol, vol.10, issue.9, pp.611-618, 1986.

P. Carmeliet, Angiogenesis in health and disease, Nature Medicine, vol.9, issue.6, pp.653-660, 2003.

P. Carmeliet and R. K. Jain, Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases, Nature Reviews. Drug Discovery, vol.10, issue.6, pp.417-427, 2011.

A. Carneiro, M. Falcão, I. Azevedo, F. Reis, F. Soares et al., Multiple effects of bevacizumab in angiogenesis: implications for its use in age-related macular degeneration, Acta Ophthalmol, vol.87, issue.5, pp.517-540, 2009.

A. Cazes, A. Galaup, C. Chomel, M. Bignon, N. Bréchot et al., Extracellular MatrixBound Angiopoietin-Like 4 Inhibits Endothelial Cell Adhesion, Migration, and Sprouting and Alters Actin Cytoskeleton, Circ Res, vol.99, issue.11, pp.1207-1222, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00177171

L. Chen, W. Zhang, N. Fregien, and M. Pierce, The her-2/neu oncogene stimulates the transcription of N-acetylglucosaminyltransferase V and expression of its cell surface oligosaccharide products, Oncogene, vol.17, issue.16, pp.2087-2093, 1998.

G. Chimote, J. Sreenivasan, N. Pawar, J. Subramanian, H. Sivaramakrishnan et al., , 2014.

, Comparison of effects of anti-angiogenic agents in the zebrafish efficacy-toxicity model for translational anti-angiogenic drug discovery, Drug Des Devel Ther, vol.8, pp.1107-1130

K. Choi, M. Kennedy, A. Kazarov, J. C. Papadimitriou, and G. Keller, A common precursor for hematopoietic and endothelial cells, Development, vol.125, issue.4, pp.725-732, 1998.

A. S. Chung and N. Ferrara, Developmental and pathological angiogenesis, Annual Review of Cell and Developmental Biology, vol.27, pp.563-584, 2011.

L. Clarion, C. Jacquard, O. Sainte-catherine, M. Decoux, S. Loiseau et al., Cglycoside mimetics inhibit glioma stem cell proliferation, migration, and invasion, Journal of Medicinal Chemistry, vol.57, issue.20, pp.8293-8306, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01100620

L. Clarion, C. Jacquard, O. Sainte-catherine, S. Loiseau, D. Filippini et al., Oxaphosphinanes: new therapeutic perspectives for glioblastoma, Journal of Medicinal Chemistry, vol.55, issue.5, pp.2196-2211, 2012.
URL : https://hal.archives-ouvertes.fr/hal-00677707

L. Conradi, A. Brajic, A. R. Cantelmo, A. Bouché, J. Kalucka et al., Tumor vessel disintegration by maximum tolerable PFKFB3 blockade, Angiogenesis, vol.20, issue.4, pp.599-613, 2017.

T. Couffinhal, P. Dufourcq, D. Daret, and C. Duplaà, , 2001.

, La Revue De Medecine Interne, vol.22, issue.11, pp.1064-1082

D. O. Croci, J. P. Cerliani, T. Dalotto-moreno, S. P. Méndez-huergo, I. D. Mascanfroni et al., Glycosylation-dependent lectin-receptor interactions preserve angiogenesis in anti-VEGF refractory tumors, Cell, vol.156, issue.4, pp.744-758, 2014.

S. Das and P. A. Marsden, Angiogenesis in glioblastoma, N Engl J Med, vol.369, issue.16, pp.1561-1564, 2013.

G. Eelen, P. De-zeeuw, M. Simons, and P. Carmeliet, Endothelial cell metabolism in normal and diseased vasculature, Circulation Research, vol.116, issue.7, pp.1231-1244, 2015.

N. Ferrara and W. J. Henzel, Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells, Biochemical and Biophysical Research Communications, vol.161, issue.2, pp.851-858, 1989.

N. Ferrara, K. J. Hillan, H. P. Gerber, and W. Novotny, Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer, Nat Rev Drug Discov, vol.3, issue.5, pp.391-400, 2004.

J. Folkman, Tumor angiogenesis: therapeutic implications, The New England Journal of Medicine, vol.285, issue.21, pp.1182-1186, 1971.

J. Folkman, Anti-angiogenesis: new concept for therapy of solid tumors, Annals of Surgery, vol.175, issue.3, pp.409-416, 1972.

J. Folkman, Clinical applications of research on angiogenesis, The New England Journal of Medicine, vol.333, issue.26, pp.1757-1763, 1995.

E. Gomez-perdiguero, A. Liabotis-fontugne, M. Durand, C. Faye, S. Ricard-blum et al., J Pathol, vol.240, issue.4, pp.461-471, 2016.

D. Gospodarowicz, Humoral control of cell proliferation: the role of fibroblast growth factor in regeneration, angiogenesis, wound healing, and neoplastic growth, Progress in Clinical and Biological Research, vol.9, pp.1-19, 1976.

P. O. Guichet, I. Bieche, M. Teigell, C. Serguera, B. Rothhut et al., Cell death and neuronal differentiation of glioblastoma stem-like cells induced by neurogenic transcription factors, Glia, issue.2, pp.225-264, 2013.

B. Haas, N. Eckstein, V. Pfeifer, P. Mayer, and M. D. Hass, Efficacy, safety and regulatory status of SGLT2 inhibitors: focus on canagliflozin. Nutrition & Diabetes, 4, e143, 2014.

M. E. Hardee and D. Zagzag, Mechanisms of Glioma-Associated Neovascularization, Am J Pathol, vol.181, issue.4, pp.1126-1167, 2012.

Z. Hassani, A. Saleh, S. Turpault, S. Khiati, W. Morelle et al., , 2017.

. Targets, Inhibits Glioblastoma-Initiating Cell Invasiveness and Proliferation, Molecular Cancer Research: MCR, vol.15, issue.10, pp.1376-1387

F. Hillen and A. W. Griffioen, Tumour vascularization: sprouting angiogenesis and beyond, Cancer Metastasis Reviews, vol.26, issue.3-4, pp.489-502, 2007.

J. E. Hudak and C. R. Bertozzi, Glycotherapy: New advances inspire a reemergence of glycans in medicine, Chemistry & Biology, vol.21, issue.1, pp.16-37, 2014.

S. Isogai, M. Horiguchi, and B. M. Weinstein, The vascular anatomy of the developing zebrafish: An atlas of embryonic and early larval development, Dev Biol, vol.230, issue.2, pp.278-301, 2001.

R. K. Jain, E. Di-tomaso, D. G. Duda, J. S. Loeffler, A. G. Sorensen et al., , 2007.

, Angiogenesis in brain tumours, Nat Rev Neurosci, vol.8, issue.8, pp.610-632

P. J. Killela, Z. J. Reitman, Y. Jiao, C. Bettegowda, N. Agrawal et al., TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal, Proc Natl Acad Sci, vol.110, issue.15, pp.6021-6027, 2013.

A. P. Kyritsis, M. L. Bondy, J. S. Rao, and C. Sioka, Inherited predisposition to glioma, NeuroOncology, vol.12, issue.1, pp.104-113, 2010.

A. Lavaud, R. Soleti, A. Hay, P. Richomme, D. Guilet et al., , 2012.

, Paradoxical effects of polyphenolic compounds from Clusiaceae on angiogenesis, Biochemical Pharmacology, vol.83, issue.4, pp.514-523

A. Lebelt, J. Dziecio?, K. Guzi?ska-ustymowicz, D. Lemancewicz, L. Zimnoch et al., , 2009.

, Angiogenesis in gliomas, Folia Histochem Cytobiol, vol.46, issue.1, pp.69-72

P. Lenzi, G. Bocci, and G. Natale, John Hunter and the origin of the term "angiogenesis, Angiogenesis, vol.19, issue.2, pp.255-256, 2016.

A. Markham, Ertugliflozin: First Global Approval, Drugs, vol.78, issue.4, pp.513-519, 2018.

B. Millauer, S. Wizigmann-voos, H. Schnürch, R. Martinez, N. P. Møller et al., High affinity VEGF binding and developmental expression suggest Flk-1 as a major regulator of vasculogenesis and angiogenesis, Cell, vol.72, issue.6, pp.835-846, 1993.

R. Missiaen, F. Morales-rodriguez, G. Eelen, and P. Carmeliet, Targeting endothelial metabolism for anti-angiogenesis therapy: A pharmacological perspective, Vascular Pharmacology, vol.90, pp.8-18, 2017.

U. Nencha, A. Rahimian, M. Giry, A. Sechi, K. Mokhtari et al., TERT promoter mutations and rs2853669 polymorphism: prognostic impact and interactions with common alterations in glioblastomas, Journal of Neuro-Oncology, vol.126, issue.3, pp.441-446, 2016.

E. A. Partridge, C. Le-roy, G. M. Di-guglielmo, J. Pawling, P. Cheung et al., Regulation of cytokine receptors by Golgi N-glycan processing and endocytosis, Science, vol.306, issue.5693, pp.120-124, 2004.

C. Peghaire, M. L. Bats, R. Sewduth, S. Jeanningros, B. Jaspard et al., Fzd7 (Frizzled-7) Expressed by Endothelial Cells Controls Blood Vessel Formation Through Wnt/?-Catenin Canonical Signaling, Arterioscler Thromb Vasc Biol, vol.36, issue.12, pp.2369-2380, 2016.

M. Potente and P. Carmeliet, The Link Between Angiogenesis and Endothelial Metabolism, Annual Review of Physiology, vol.79, pp.43-66, 2017.

S. M. Raza, G. N. Fuller, C. H. Rhee, S. Huang, K. Hess et al., Identification of necrosis-associated genes in glioblastoma by cDNA microarray analysis, Clin Cancer Res, vol.10, issue.1, pp.212-233, 2004.

D. Ricard, A. Idbaih, F. Ducray, M. Lahutte, K. Hoang-xuan et al., Primary brain tumours in adults, Lancet, issue.9830, pp.1984-1996, 2012.

W. Risau, Mechanisms of angiogenesis, Nature, vol.386, issue.6626, pp.671-674, 1997.

W. Risau and I. Flamme, Vasculogenesis. Annual Review of Cell and Developmental Biology, vol.11, pp.73-91, 1995.

S. Schoors, K. De-bock, A. R. Cantelmo, M. Georgiadou, B. Ghesquière et al., Partial and transient reduction of glycolysis by PFKFB3 blockade reduces pathological angiogenesis, Cell Metabolism, vol.19, issue.1, pp.37-48, 2014.

J. A. Schwartzbaum, J. L. Fisher, K. D. Aldape, and M. Wrensch, Epidemiology and molecular pathology of glioma, Nature Clinical Practice. Neurology, vol.2, issue.9, p.516, 2006.

G. L. Semenza, Vasculogenesis, angiogenesis, and arteriogenesis: mechanisms of blood vessel formation and remodeling, Journal of Cellular Biochemistry, vol.102, issue.4, pp.840-847, 2007.

D. R. Senger, S. J. Galli, A. M. Dvorak, C. A. Perruzzi, V. S. Harvey et al., Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid, Science, vol.219, issue.4587, pp.983-985, 1983.

P. Stanley, Galectin-1 Pulls the Strings on VEGFR2, Cell, vol.156, issue.4, pp.625-631, 2014.

P. Stapor, X. Wang, J. Goveia, S. Moens, and P. Carmeliet, Angiogenesis revisited -role and therapeutic potential of targeting endothelial metabolism, Journal of Cell Science, vol.127, pp.4331-4341, 2014.

S. R. Stowell, T. Ju, and R. D. Cummings, Protein glycosylation in cancer, Annual Review of Pathology, vol.10, pp.473-510, 2015.

S. Telang, B. F. Clem, A. C. Klarer, A. L. Clem, J. O. Trent et al., Small molecule inhibition of 6-phosphofructo-2-kinase suppresses t cell activation, Journal of Translational Medicine, vol.10, p.95, 2012.

L. Treps, L. Conradi, U. Harjes, and P. Carmeliet, Manipulating Angiogenesis by Targeting Endothelial Metabolism: Hitting the Engine Rather than the Drivers-A New Perspective?, Pharmacological Reviews, vol.68, issue.3, pp.872-887, 2016.

T. H. Ung, H. Malone, P. Canoll, and J. N. Bruce, Convection-enhanced delivery for glioblastoma: targeted delivery of antitumor therapeutics, CNS Oncology, vol.4, issue.4, pp.225-234, 2015.

S. Vandekeere, M. Dewerchin, and P. Carmeliet, Angiogenesis Revisited: An Overlooked Role of Endothelial Cell Metabolism in Vessel Sprouting, vol.22, pp.509-517, 1994.

D. Verdegem, S. Moens, P. Stapor, and P. Carmeliet, Endothelial cell metabolism: parallels and divergences with cancer cell metabolism, Cancer & Metabolism, vol.2, p.1, 2014.

A. Yalcin, B. F. Clem, A. Simmons, A. Lane, K. Nelson et al., Nuclear targeting of 6-phosphofructo-2-kinase (PFKFB3) increases proliferation via cyclin-dependent kinases, J Biol Chem, vol.284, issue.36, pp.24223-24232, 2009.

S. Zhao, Y. Lin, W. Xu, W. Jiang, Z. Zha et al., Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha, Science, vol.324, issue.5924, pp.261-266, 2009.