Y. Miki, J. Swensen, D. Shattuck-eidens, P. Futreal, and K. Harshman, A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1, Science, vol.266, issue.5182, pp.66-71, 1994.
DOI : 10.1126/science.7545954

S. Narod and W. Foulkes, BRCA1 and BRCA2: 1994 and beyond, Nature Reviews Cancer, vol.14, issue.9, pp.665-676, 2004.
DOI : 10.1002/1521-1878(200008)22:8<728::AID-BIES6>3.3.CO;2-2

R. Yarden and M. Papa, BRCA1 at the crossroad of multiple cellular pathways: approaches for therapeutic interventions, Molecular Cancer Therapeutics, vol.5, issue.6, pp.1396-1404, 2006.
DOI : 10.1158/1535-7163.MCT-05-0471

T. Xiang, A. Ohashi, Y. Huang, T. Pandita, and T. Ludwig, Negative Regulation of AKT Activation by BRCA1, Cancer Research, vol.68, issue.24, pp.10040-10044, 2008.
DOI : 10.1158/0008-5472.CAN-08-3009

H. Kang, H. Kim, J. Rih, T. Mattson, and K. Kim, BRCA1 Plays a Role in the Hypoxic Response by Regulating HIF-1?? Stability and by Modulating Vascular Endothelial Growth Factor Expression, Journal of Biological Chemistry, vol.62, issue.19, pp.13047-13056, 2006.
DOI : 10.1016/S0046-8177(98)90427-0

P. Van-der-groep, A. Bouter, F. Menko, E. Van-der-wall, and P. Van-diest, High frequency of HIF-1?? overexpression in BRCA1 related breast cancer, Breast Cancer Research and Treatment, vol.8, issue.1, pp.475-480, 2008.
DOI : 10.1093/jnci/djh114

K. Moreau, E. Dizin, H. Ray, C. Luquain, and E. Lefai, BRCA1 Affects Lipid Synthesis through Its Interaction with Acetyl-CoA Carboxylase, Journal of Biological Chemistry, vol.269, issue.6, pp.3172-3181, 2006.
DOI : 10.1111/j.1349-7006.2004.tb02195.x

URL : https://hal.archives-ouvertes.fr/hal-00314886

R. Deberardinis, J. Lum, G. Hatzivassiliou, and C. Thompson, The Biology of Cancer: Metabolic Reprogramming Fuels Cell Growth and Proliferation, Cell Metabolism, vol.7, issue.1, pp.11-20, 2008.
DOI : 10.1016/j.cmet.2007.10.002

M. Vander-heiden, L. Cantley, and C. Thompson, Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation, Science, vol.26, issue.1, pp.1029-1033, 2009.
DOI : 10.1038/nrc2536

R. Cairns, I. Harris, and T. Mak, Regulation of cancer cell metabolism, Nature Reviews Cancer, vol.118, issue.2, pp.85-95, 2011.
DOI : 10.1158/0008-5472.CAN-09-2266

C. Thompson, Metabolic Enzymes as Oncogenes or Tumor Suppressors, New England Journal of Medicine, vol.360, issue.8, pp.813-815, 2009.
DOI : 10.1056/NEJMe0810213

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2848669/pdf

B. Pasini and C. Stratakis, SDH mutations in tumorigenesis and inherited endocrine tumours: lesson from the phaeochromocytoma-paraganglioma syndromes, Journal of Internal Medicine, vol.1073, issue.1, pp.19-42, 2009.
DOI : 10.1016/S0002-9440(10)64547-6

A. King, M. Selak, and E. Gottlieb, Succinate dehydrogenase and fumarate hydratase: linking mitochondrial dysfunction and cancer, Oncogene, vol.25, issue.34, pp.4675-4682, 2006.
DOI : 10.1371/journal.pgen.0010008

URL : http://www.nature.com/onc/journal/v25/n34/pdf/1209594a.pdf

S. Niemann and U. Muller, Mutations in SDHC cause autosomal dominant paraganglioma, type 3, Nat Genet, vol.26, pp.268-270, 2000.

W. Kaelin and . Jr, SDH5 Mutations and Familial Paraganglioma: Somewhere Warburg is Smiling, Cancer Cell, vol.16, issue.3, pp.180-182, 2009.
DOI : 10.1016/j.ccr.2009.08.013

URL : https://doi.org/10.1016/j.ccr.2009.08.013

H. Yan, D. Parsons, J. G. Mclendon, R. Rasheed, and B. , Mutations in Gliomas, New England Journal of Medicine, vol.360, issue.8, pp.765-773, 2009.
DOI : 10.1056/NEJMoa0808710

D. Parsons, S. Jones, X. Zhang, J. Lin, and R. Leary, An Integrated Genomic Analysis of Human Glioblastoma Multiforme, Science, vol.25, issue.13, pp.1807-1812, 2008.
DOI : 10.1002/jcc.20084

D. Morvan and A. Demidem, Metabolomics by Proton Nuclear Magnetic Resonance Spectroscopy of the Response to Chloroethylnitrosourea Reveals Drug Efficacy and Tumor Adaptive Metabolic Pathways, Cancer Research, vol.67, issue.5, pp.2150-2159, 2007.
DOI : 10.1158/0008-5472.CAN-06-2346

M. Bayet-robert and D. Morvan, Metabolomics Reveals Metabolic Targets and Biphasic Responses in Breast Cancer Cells Treated by Curcumin Alone and in Association with Docetaxel, PLoS ONE, vol.287, issue.3, pp.57971-101371, 2013.
DOI : 10.1371/journal.pone.0057971.s001

I. Bae, S. Fan, Q. Meng, J. Rih, and H. Kim, BRCA1 Induces Antioxidant Gene Expression and Resistance to Oxidative Stress, Cancer Research, vol.64, issue.21, pp.7893-7909, 2004.
DOI : 10.1158/0008-5472.CAN-04-1119

URL : http://cancerres.aacrjournals.org/content/canres/64/21/7893.full.pdf

L. Cao, X. Xu, L. Cao, R. Wang, and X. Coumoul, Absence of full-length Brca1 sensitizes mice to oxidative stress and carcinogen-induced tumorigenesis in the esophagus and forestomach, Carcinogenesis, vol.28, issue.7, pp.1401-1407, 2007.
DOI : 10.1093/carcin/bgm060

H. Nam, B. Chung, Y. Kim, K. Lee, and D. Lee, Combining tissue transcriptomics and urine metabolomics for breast cancer biomarker identification, Bioinformatics, vol.25, issue.23, pp.3151-3157, 2009.
DOI : 10.1093/bioinformatics/btp558

URL : https://academic.oup.com/bioinformatics/article-pdf/25/23/3151/16890667/btp558.pdf

J. Ippolito, J. Xu, S. Jain, K. Moulder, and S. Mennerick, An integrated functional genomics and metabolomics approach for defining poor prognosis in human neuroendocrine cancers, Proceedings of the National Academy of Sciences, vol.6, issue.11, pp.9901-9906, 2005.
DOI : 10.1038/ncb1181

URL : http://www.pnas.org/content/102/28/9901.full.pdf

M. Privat, C. Aubel, S. Arnould, Y. Communal, and M. Ferrara, Breast cancer cell response to genistein is conditioned by BRCA1 mutations, Biochemical and Biophysical Research Communications, vol.379, issue.3, pp.785-789, 2009.
DOI : 10.1016/j.bbrc.2008.12.151

M. Bayet-robert, D. Loiseau, P. Rio, A. Demidem, and C. Barthomeuf, Quantitative two-dimensional HRMAS 1H-NMR spectroscopy-based metabolite profiling of human cancer cell lines and response to chemotherapy, Magnetic Resonance in Medicine, vol.74, issue.5, pp.1172-83, 2010.
DOI : 10.4161/cc.6.7.4050

F. Elstrodt, A. Hollestelle, J. Nagel, M. Gorin, and M. Wasielewski, Mutation Analysis of 41 Human Breast Cancer Cell Lines Reveals Three New Deleterious Mutants, Cancer Research, vol.66, issue.1, pp.41-45, 2006.
DOI : 10.1158/0008-5472.CAN-05-2853

E. Gottlieb and I. Tomlinson, Mitochondrial tumour suppressors: a genetic and biochemical update, Nature Reviews Cancer, vol.284, issue.11, pp.857-866, 2005.
DOI : 10.1152/ajplung.00149.2002

Z. Reitman and H. Yan, Isocitrate Dehydrogenase 1 and 2 Mutations in Cancer: Alterations at a Crossroads of Cellular Metabolism, JNCI Journal of the National Cancer Institute, vol.102, issue.13, pp.932-973, 2010.
DOI : 10.1093/jnci/djq187

R. Robey and N. Hay, Is Akt the ???Warburg kinase???????Akt-energy metabolism interactions and oncogenesis, Seminars in Cancer Biology, vol.19, issue.1, pp.25-31, 2009.
DOI : 10.1016/j.semcancer.2008.11.010

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2814453/pdf

B. Xu, S. Kim, and M. Kastan, Involvement of Brca1 in S-Phase and G2-Phase Checkpoints after Ionizing Irradiation, Molecular and Cellular Biology, vol.21, issue.10, pp.3445-3450, 2001.
DOI : 10.1128/MCB.21.10.3445-3450.2001

E. Chautard, G. Loubeau, A. Tchirkov, J. Chassagne, and C. Vermot-desroches, Akt signaling pathway: a target for radiosensitizing human malignant glioma, Neuro Oncol, vol.12, issue.5, pp.434-477, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01636179

S. Mathupala, Y. Ko, and P. Pedersen, Hexokinase-2 bound to mitochondria: Cancer's stygian link to the ???Warburg effect??? and a pivotal target for effective therapy, Seminars in Cancer Biology, vol.19, issue.1, pp.17-24, 2009.
DOI : 10.1016/j.semcancer.2008.11.006

R. Scatena, P. Bottoni, A. Pontoglio, L. Mastrototaro, and B. Giardina, Glycolytic enzyme inhibitors in cancer treatment, Expert Opinion on Investigational Drugs, vol.269, issue.10, pp.1533-1545, 2008.
DOI : 10.1021/jm980334n

M. Vander-heiden, Targeting cancer metabolism: a therapeutic window opens, Nature Reviews Drug Discovery, vol.25, issue.9, pp.671-684, 2011.
DOI : 10.1101/gad.1987211

M. Seo, J. Kim, D. Neau, I. Sehgal, and Y. Lee, Structure-Based Development of Small Molecule PFKFB3 Inhibitors: A Framework for Potential Cancer Therapeutic Agents Targeting the Warburg Effect, PLoS ONE, vol.50, issue.9, p.24179, 2011.
DOI : 10.1371/journal.pone.0024179.t003

T. Xiang, Y. Jia, D. Sherris, S. Li, and H. Wang, Targeting the Akt/mTOR pathway in Brca1-deficient cancers, Oncogene, vol.3, issue.21, pp.2443-2450, 2011.
DOI : 10.1054/bjoc.1999.0995

M. Selak, S. Armour, E. Mackenzie, H. Boulahbel, and D. Watson, Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-?? prolyl hydroxylase, Cancer Cell, vol.7, issue.1, pp.77-85, 2005.
DOI : 10.1016/j.ccr.2004.11.022

URL : https://doi.org/10.1016/j.ccr.2004.11.022

P. Pollard, J. Briere, N. Alam, J. Barwell, and E. Barclay, Accumulation of Krebs cycle intermediates and over-expression of HIF1?? in tumours which result from germline FH and SDH mutations, Human Molecular Genetics, vol.14, issue.15, pp.2231-2239, 2005.
DOI : 10.1093/hmg/ddi227

G. Semenza, P. Roth, H. Fang, and G. Wang, Transcriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1, J Biol Chem, vol.269, pp.23757-23763, 1994.

R. Shaw, Glucose metabolism and cancer, Current Opinion in Cell Biology, vol.18, issue.6, pp.598-608, 2006.
DOI : 10.1016/j.ceb.2006.10.005

C. Magnard, R. Bachelier, A. Vincent, M. Jaquinod, and S. Kieffer, BRCA1 interacts with acetyl-CoA carboxylase through its tandem of BRCT domains, Oncogene, vol.285, issue.44, pp.6729-6739, 2002.
DOI : 10.1126/science.285.5428.747

J. Brunet, A. Vazquez-martin, R. Colomer, B. Grana-suarez, and B. Martin-castillo, BRCA1 and acetyl-CoA carboxylase: The metabolic syndrome of breast cancer, Molecular Carcinogenesis, vol.66, issue.2, pp.157-163, 2008.
DOI : 10.1002/mc.20364

E. Delikatny, S. Chawla, D. Leung, and H. Poptani, MR-visible lipids and the tumor microenvironment, NMR in Biomedicine, vol.25, issue.(Suppl 1), pp.592-611, 2011.
DOI : 10.1148/radiology.211.3.r99jn46791

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3640643/pdf

S. Fan, Q. Meng, T. Saha, F. Sarkar, and E. Rosen, Low Concentrations of Diindolylmethane, a Metabolite of Indole-3-Carbinol, Protect against Oxidative Stress in a BRCA1-Dependent Manner, Cancer Research, vol.69, issue.15, pp.6083-6091, 2009.
DOI : 10.1158/0008-5472.CAN-08-3309

O. Warburg, On the Origin of Cancer Cells, Science, vol.123, issue.3191, pp.309-314, 1956.
DOI : 10.1126/science.123.3191.309