S. Silvente-poirot and M. Poirot, Cholesterol metabolism and cancer: the good, the bad and the ugly, Curr. Opin. Pharmacol, vol.12, pp.673-676, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00822284

C. Zerbinati and L. Iuliano, Cholesterol and related sterols autoxidation, Free Radic, Biol. Med, vol.111, pp.151-155, 2017.

G. J. Schroepfer, Oxysterols: modulators of cholesterol metabolism and other processes, Physiol. Rev, vol.80, pp.361-554, 2000.

M. Poirot and S. Silvente-poirot, Cholesterol-5,6-epoxides: chemistry, biochemistry, metabolic fate and cancer, Biochimie, vol.95, pp.622-631, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00743461

M. R. Paillasse, N. Saffon, H. Gornitzka, S. Silvente-poirot, M. Poirot et al., Surprising unreactivity of cholesterol-5,6-epoxides towards nucleophiles, J. Lipid Res, vol.53, pp.718-725, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00743457

J. S. Min, M. I. Khan, S. O. Lee, D. G. Yim, K. H. Seol et al., Impact of cooking, storage, and reheating conditions on the formation of cholesterol oxidation products in Pork Loin, Korean J. Food Sci. Anim. Resour, vol.36, pp.23-28, 2016.

M. I. Khan, J. S. Min, S. O. Lee, D. G. Yim, K. H. Seol et al., Cooking, storage, and reheating effect on the formation of cholesterol oxidation products in processed meat products, Lipids Health Dis, vol.14, p.89, 2015.

J. Ahn, I. Jeong, B. Kwak, D. Leem, T. Yoon et al., Rapid determination of cholesterol in milk containing emulsified foods, Food Chem, vol.135, pp.2411-2417, 2012.

H. Lee, J. Chien, and B. I. Chen, Formation of cholesterol oxidation products in marinated foods during heating, J. Agric. Food Chem, vol.54, pp.4873-4879, 2006.

G. P. Savage, P. C. Dutta, and M. T. Rodriguez-estrada, Cholesterol oxides: their occurrence and methods to prevent their generation in foods, Asia Pac. J. Clin. Nutr, vol.11, pp.72-78, 2002.

V. K. Rao, B. N. Kowale, N. P. Babu, and G. S. Bisht, Effect of cooking and storage on lipid oxidation and development of cholesterol oxidation products in water buffalo meat, Meat Sci, vol.43, pp.179-185, 1996.

B. N. Kowale, V. K. Rao, N. P. Babu, N. Sharma, and G. S. Bisht, Lipid oxidation and cholesterol oxidation in mutton during cooking and storage, Meat Sci, vol.43, pp.195-202, 1996.

J. E. Pie, K. Spahis, and C. Seillan, Evaluation of oxidative degradation of cholesterol in food and food ingredients: identification and quantification of cholesterol oxides, J. Agric. Food Chem, vol.38, pp.973-979, 1990.

J. Bascoul, N. Domergue, M. Olle, and A. , Crastes de Paulet, Autoxidation of cholesterol in tallows heated under deep frying conditions: evaluation of oxysterols by GLC and TLC-FID, Lipids, vol.21, pp.383-387, 1986.

P. B. Addis, Occurrence of lipid oxidation products in foods, Food Chem. Toxicol, vol.24, pp.1021-1030, 1986.

S. Silvente-poirot and M. Poirot, Cholesterol epoxide hydrolase and cancer, Curr. Opin. Pharmacol, vol.12, pp.696-703, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00743463

P. Medina, M. R. Paillasse, G. Segala, M. Poirot, and S. Silvente-poirot, Identification and pharmacological characterization of cholesterol-5,6-epoxide hydrolase as a target for tamoxifen and AEBS ligands, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.13520-13525, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00519159

M. Voisin, P. Medina, A. Mallinger, F. Dalenc, E. Huc-claustre et al., Identification of a tumor-promoter cholesterol metabolite in human breast cancers acting through the glucocorticoid receptor, Proc. Natl. Acad. Sci. U. S. A, vol.114, pp.9346-9355, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02316171

S. Silvente-poirot, G. Segala, M. C. Poirot, and M. Poirot, Ligand-dependent transcriptional induction of lethal autophagy: a new perspective for cancer treatment, Autophagy, pp.555-557, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02380583

M. Poirot and S. Silvente-poirot, The tumor-suppressor cholesterol metabolite, dendrogenin A, is a new class of LXR modulator activating lethal autophagy in cancers, Biochem. Pharmacol, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-02380575

G. Segala, M. David, P. Medina, M. C. Poirot, N. Serhan et al., Nat. Commun, vol.8, p.1903, 2017.

F. Dalenc, M. Poirot, and S. Silvente-poirot, Dendrogenin a: a mammalian metabolite of cholesterol with tumor suppressor and neurostimulating properties, Curr. Med. Chem, vol.22, pp.3533-3549, 2015.

, Scheme describing the OCDO metabolic and tumor promoter pathway

S. Silvente-poirot and M. Poirot, the balance, vol.343, pp.1445-1446, 2014.
URL : https://hal.archives-ouvertes.fr/inserm-00968924

J. A. Salvador, J. F. Carvalho, M. A. Neves, S. M. Silvestre, A. J. Leitao et al., Anticancer steroids: linking natural and semi-synthetic compounds, Nat. Prod. Rep, vol.30, pp.324-374, 2013.

P. Medina, M. R. Paillasse, G. Segala, M. Voisin, L. Mhamdi et al., Dendrogenin A arises from cholesterol and histamine metabolism and shows cell differentiation and anti-tumour properties, Nat. Commun, vol.4, p.1840, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00823974

S. Silvente-poirot, P. Medina, M. Record, and M. Poirot, From tamoxifen to dendrogenin A: the discovery of a mammalian tumor suppressor and cholesterol metabolite, Biochimie, vol.130, pp.109-114, 2016.

M. Poirot and S. Silvente-poirot, When cholesterol meets histamine, it gives rise to dendrogenin A: a tumour suppressor metabolite, Biochem. Soc. Trans, vol.44, pp.631-637, 2016.

E. R. Nelson, The significance of cholesterol and its metabolite, 27-hydroxycholesterol in breast cancer, Mol. Cell. Endocrinol, vol.466, pp.73-80, 2018.

S. He and E. R. Nelson, 27-Hydroxycholesterol, an endogenous selective estrogen receptor modulator, Maturitas, vol.104, pp.29-35, 2017.

L. L. Smith, Review of progress in sterol oxidations, Lipids, vol.31, pp.453-487, 1987.

R. E. Marker, E. Rohman, and S. L. , Cholesterol and sitosterol derivatives, J. Am. Chem. Soc, vol.62, pp.516-517, 1940.

M. Fieser, A. Quilico, A. Nickon, W. E. Rosen, E. J. Tarlton et al., Permanganate oxidation of ergosterol, J. Am. Chem. Soc, vol.75, pp.4066-4071, 1953.

L. F. Fieser and S. Rajagopalan, Selective oxidation with N-bromosuccinimide. II. cholestena-3b,5a,6b-triol, J. Am. Chem. Soc, vol.71, pp.3938-3941, 1949.

A. Rosado-abon, M. Romero-avila, and M. A. , Iglesis-Arteaga, A rapid and simple one-pot procedure for the synthesis of 3b-acetoxy-5a-hydroxy-6-oxo steroids, Arch. Org. Chem, vol.10, pp.110-115, 2010.

J. F. Carvalho, M. M. Silva, J. N. Moreira, S. Simoes, and M. L. Sa-e-melo, Sterols as anticancer agents: synthesis of ring-B oxygenated steroids, cytotoxic profile, and comprehensive SAR analysis, J. Med. Chem, vol.53, pp.7632-7638, 2010.

M. Frigerio, M. Santagostino, and S. Sputore, Process for the Oxidation of Primary and Secondary Alcohols to Aldehydes and Ketones and for the Oxidation of 1,2-diols to Alpha-ketols and Alpha-diketones, Patent, EP0658533, pp.1-12, 1994.

R. P. Graber, J. C. Snoddy, H. B. Arnold, and N. L. Wendler, Notes -the oxidation of cholesterol by periodic acid, J. Org. Chem, vol.21, pp.1517-1518, 1956.

M. Voisin, S. Silvente-poirot, and M. Poirot, One step synthesis of 6-oxo-cholestan-3beta,5alpha-diol, Biochem. Biophys. Res. Commun, vol.446, pp.782-785, 2014.

R. Soules, E. Noguer, L. Iuliano, C. Zerbinati, J. Leignadier et al., Improvement of 5,6alpha-epoxycholesterol, 5,6beta-epoxycholesterol, cholestane-3beta,5alpha,6beta-triol and 6-oxo-cholestan-3beta,5alpha-diol recovery for quantification by GC/ MS, Chem. Phys. Lipids, vol.207, pp.92-98, 2017.

F. Dalenc, L. Iuliano, T. Filleron, C. Zerbinati, M. Voisin et al., Circulating oxysterol metabolites as potential new surrogate markers in patients with hormone receptor-positive breast cancer: results of the OXYTAM study, J. Steroid Biochem. Mol. Biol, vol.169, pp.210-218, 2017.

L. Iuliano, P. J. Crick, C. Zerbinati, L. Tritapepe, J. Abdel-khalik et al., Cholesterol metabolites exported from human brain, Steroids, vol.99, pp.189-193, 2015.

P. Medina, M. R. Paillasse, B. Payre, S. Silvente-poirot, and M. Poirot, Synthesis of new alkylaminooxysterols with potent cell differentiating activities: identification of leads for the treatment of cancer and neurodegenerative diseases, J. Med. Chem, vol.52, pp.7765-7777, 2009.
URL : https://hal.archives-ouvertes.fr/inserm-00519151

W. J. Szczepek, Reactions of 5-hydroxy-6-oxosteroids. VI. Acid-catalyzed isomerization of some 5a-hydroxy-6-oxosteroids, Acta Chim. Hung, vol.123, pp.69-76, 1986.

W. J. Szczepek, Reactions of 5-hydroxy-6-oxo steroids. VII. New reagent for the Westphalen-type rearrangement of 5a-hydroxy-6-oxo steroids, Acta Chim. Hung, vol.123, pp.77-82, 1986.

W. J. Szczepek, Tribromic acid as brominating agent, Pol. J. Chem, vol.55, pp.709-711, 1981.

W. J. Szczepek, J. Gumulka, and J. J. Jagodzinski, Reactions of 5-hydroxy-6-oxo steroids. II. New aromatization reactions of steroids, Bull. Acad. Pol. Sci. Ser. Sci. Chim, vol.28, pp.165-172, 1980.

M. K. Pulfer, C. Taube, E. Gelfand, and R. C. Murphy, Ozone exposure in vivo and formation of biologically active oxysterols in the lung, J. Pharmacol. Exp. Therapeut, vol.312, pp.256-264, 2005.

Z. Xiangdong, F. Bo, and T. Linghui, The dendritic aminosterol compounds, their preparation method and application as tumor immunotherapy agent for cancer treatment, Patent, CN 102344480, pp.1-15, 2012.

F. Bo, T. Linghui, Z. Xiangdong, and Z. Chenghe, Synthesis of 6b-butylaminocholestan-3b, 5a-diol, an analogue of anti-tumoral dendrogenin, Acta Acad. Med. Mil. Tertiae, vol.32, pp.1051-1054, 2010.

X. Jiang, R. Sidhu, L. Mydock-mcgrane, F. Hsu, D. F. Covey et al., Development of a bile acidebased newborn screen for Niemann-Pick disease type C, Sci. Transl. Med, vol.8, pp.337-363, 2016.

H. G. Roscoe, R. Goldstein, and M. J. Fahrenbach, Metabolism of cholestane-3-beta,5-alpha,6-beta-triol. I. The fate of cholestanetriol in the rat, Biochem. Pharmacol, vol.17, pp.1189-1196, 1968.

S. Kikuchi, Y. Imai, Z. Suzuoki, T. Matsu, and S. Noguchi, Biologic studies of cholestane-3-beta,5-alpha,6-beta-triol and its derivatives. 3. The metabolic fate and metabolites of cholestane-3-beta,5-alpha,6-beta-triol in animals, J. Pharmacol. Exp. Therapeut, vol.159, pp.399-408, 1968.

H. G. Roscoe and M. J. Fahrenbach, Metabolism of cholestane-3 beta,5 alpha,6 beta-triol. II. Identification of two major neutral metabolites in the rat, J. Lipid Res, vol.12, pp.17-23, 1971.

M. Yamaguchi, Y. Endo, M. Shimizu, K. Yamamoto, S. Yamada et al., 3 beta,5 alpha-Dihydroxycholestan-6-one exists in human blood, Biol. Pharm. Bull, vol.20, pp.1044-1046, 1997.

M. K. Pulfer and R. C. Murphy, Formation of biologically active oxysterols during ozonolysis of cholesterol present in lung surfactant, J. Biol. Chem, vol.279, pp.26331-26338, 2004.

R. Lathe and Y. Kotelevtsev, Steroid signaling: ligand-binding promiscuity, molecular symmetry, and the need for gating, Steroids, vol.82, pp.14-22, 2014.

A. Odermatt and P. Klusonova, 11b-Hydroxysteroid dehydrogenase 1: regeneration of active glucocorticoids is only part of the story, J. Steroid Biochem. Mol. Biol, vol.151, pp.85-92, 2015.

T. Mitic, S. Shave, N. Semjonous, I. Mcnae, D. F. Cobice et al., 11beta-Hydroxysteroid dehydrogenase type 1 contributes to the balance between 7-keto-and 7-hydroxy-oxysterols in vivo, vol.86, pp.146-153, 2013.

M. Wamil, R. Andrew, K. E. Chapman, J. Street, N. M. Morton et al., 7-oxysterols modulate glucocorticoid activity in adipocytes through competition for 11beta-hydroxysteroid dehydrogenase type, Endocrinology, vol.149, pp.5909-5918, 2008.

R. A. Schweizer, M. Zurcher, Z. Balazs, B. Dick, and A. Odermatt, Rapid hepatic metabolism of 7-ketocholesterol by 11beta-hydroxysteroid dehydrogenase type 1: species-specific differences between the rat, human, and hamster enzyme, J. Biol. Chem, vol.279, pp.18415-18424, 2004.

K. Chapman, M. Holmes, and J. , Seckl, 11beta-hydroxysteroid dehydrogenases: intracellular gate-keepers of tissue glucocorticoid action, Physiol. Rev, vol.93, pp.1139-1206, 2013.

G. Segala, P. Medina, L. Iuliano, C. Zerbinati, M. R. Paillasse et al., -Epoxy-cholesterols contribute to the anticancer pharmacology of tamoxifen in breast cancer cells, Biochem. Pharmacol, vol.5, issue.6, pp.175-189, 2013.

B. Rubis, S. Grodecka-gazdecka, R. Lecybyl, M. Ociepa, Z. Krozowski et al., Contribution of protein kinase A and protein kinase C signalling pathways to the regulation of HSD11B2 expression and proliferation of MCF-7 cells, Acta Biochim. Pol, vol.51, pp.919-924, 2004.

A. R. Nawrocki, C. E. Goldring, R. M. Kostadinova, F. J. Frey, and B. M. Frey, In vivo footprinting of the human 11b-hydroxysteroid dehydrogenase type 2 promoter: evidence for cell-specific regulation by Sp1 and Sp3, J. Biol. Chem, vol.277, pp.14647-14656, 2002.

S. Hundertmark, H. Buhler, M. Rudolf, H. Weitzel, and V. Ragosch, Inhibition of 11 beta-hydroxysteroid dehydrogenase activity enhances the antiproliferative effect of glucocorticosteroids on MCF-7 and ZR-75-1 breast cancer cells, J. Endocrinol, vol.155, pp.171-180, 1997.

T. Mitic, R. Andrew, B. R. Walker, and P. W. , Hadoke, 11beta-Hydroxysteroid dehydrogenase type 1 contributes to the regulation of 7-oxysterol levels in the arterial wall through the inter-conversion of 7-ketocholesterol and 7beta-hydroxycholesterol, Biochimie, vol.95, pp.548-555, 2013.

Z. Balazs, L. G. Nashev, C. Chandsawangbhuwana, M. E. Baker, and A. Odermatt, Hexose-6-phosphate dehydrogenase modulates the effect of inhibitors and alternative substrates of 11beta-hydroxysteroid dehydrogenase 1, Mol. Cell. Endocrinol, vol.301, pp.117-122, 2009.

M. Hult, B. Elleby, N. Shafqat, S. Svensson, A. Rane et al., Human and rodent type 1 11beta-hydroxysteroid dehydrogenases are 7beta-hydroxycholesterol dehydrogenases involved in oxysterol metabolism, Cell. Mol. Life Sci, vol.61, pp.992-999, 2004.

C. Frick, A. G. Atanasov, P. Arnold, J. Ozols, and A. Odermatt, Appropriate function of 11beta-hydroxysteroid dehydrogenase type 1 in the endoplasmic reticulum lumen is dependent on its N-terminal region sharing similar topological determinants with 50-kDa esterase, J. Biol. Chem, vol.279, pp.31131-31138, 2004.

H. Fuda, N. B. Javitt, K. Mitamura, S. Ikegawa, and C. A. Strott, Oxysterols are substrates for cholesterol sulfotransferase, J. Lipid Res, vol.48, pp.1343-1352, 2007.

A. A. Kandutsch, H. W. Chen, and H. J. Heiniger, Biological activity of some oxygenated sterols, Science, vol.201, pp.498-501, 1978.

A. A. Kandutsch and H. W. Chen, Inhibition of cholesterol synthesis by oxygenated sterols, Lipids, vol.13, pp.704-707, 1978.

W. K. Cavenee, G. F. Gibbons, H. W. Chen, and A. A. Kandutsch, Effects of various oxygenated sterols on cellular sterol biosynthesis in Chinese hamster lung cells resistant to 25-hydroxycholesterol, Biochim. Biophys. Acta, vol.575, pp.255-265, 1979.

D. T. Witiak, R. A. Parker, D. R. Brann, M. E. Dempsey, M. C. Ritter et al., Biological evaluation in vivo and in vitro of selected 5-alpha-cholestane-3-beta, 5-alpha, 6-beta-triol analogs as hypocholesterolemic agents, J. Med. Chem, vol.14, pp.216-222, 1971.

B. , J. Hora, V. Landa, V. Cerný, and F. Sorm, On steroids CXIII. Sterilizing effect of some 6-ketosteroids on housefly/Musca domestica L, Steroids, vol.11, pp.475-496, 1968.

S. Yachnin, R. A. Streuli, L. I. Gordon, and R. C. Hsu, Alteration of peripheral blood cell membrane function and morphology by oxygenated sterols; a membrane insertion hypothesis, Curr. Top. Hematol, vol.2, pp.245-271, 1979.

R. A. Streuli, J. Chung, A. M. Scanu, and S. Yachnin, Inhibition of human lymphocyte E-rosette formation by oxygenated sterols, J. Immunol, vol.123, pp.2897-2902, 1979.

L. I. Gordon, J. Bass, and S. Yachnin, Inhibition of human polymorphonuclear leukocyte chemotaxis by oxygenated sterol compounds, Proc. Natl. Acad. Sci. U. S. A, vol.77, pp.4313-4316, 1980.

O. Kucuk, J. Stoner-picking, S. Yachnin, L. I. Gordon, R. M. Williams et al., Inhibition of NK cell-mediated cytotoxicity by oxysterols, Cell. Immunol, vol.139, pp.541-549, 1992.

O. Kucuk, J. Stoner-picking, S. Yachnin, L. I. Gordon, R. M. Williams et al., Inhibition of cytolytic T lymphocyte activity by oxysterols, Lipids, vol.29, pp.657-660, 1994.

G. A. Ansari, R. D. Walker, V. B. Smart, and L. L. Smith, Further investigations of mutagenic cholesterol preparations, Food Chem. Toxicol, vol.20, pp.35-41, 1982.

J. F. Carvalho, M. M. Silva, J. N. Moreira, S. Simoes, M. L. Sa et al., Selective cytotoxicity of oxysterols through structural modulation on rings A and B. Synthesis, in vitro evaluation, and SAR, J. Med. Chem, vol.54, pp.6375-6393, 2011.

B. Kosmider, J. E. Loader, R. C. Murphy, and R. J. Mason, Apoptosis induced by ozone and oxysterols in human alveolar epithelial cells, Free Radic, Biol. Med, vol.48, pp.1513-1524, 2010.

Y. Endo, H. Fukasawa, Y. Hashimoto, and K. Shudo, Synthesis of oxygenated cholesterols as structural mimics of phorbol ester-type tumor promoters, Chem. Pharm. Bull. (Tokyo), pp.462-469, 1994.

Y. Endo, Y. Hashimoto, H. Fukasawa, and K. Shudo, Oxygenated cholesterols as ligands for cytosolic-nuclear tumor promoter binding protein: yakkasteroids, Biochem. Biophys. Res. Commun, vol.194, pp.1529-1535, 1993.

J. F. Carvalho, M. M. Silva, J. N. Moreira, S. Simoes, M. L. Sa et al., Efficient chemoenzymatic synthesis, cytotoxic evaluation, and SAR of epoxysterols, J. Med. Chem, vol.52, pp.4007-4019, 2009.

A. Vejux, L. Malvitte, and G. Lizard, Side effects of oxysterols: cytotoxicity, oxidation, inflammation, and phospholipidosis, Braz, J. Med. Biol. Res, vol.41, pp.545-556, 2008.

T. Wielkoszynski, K. Gawron, J. Strzelczyk, P. Bodzek, M. Zalewska-ziob et al., Cellular toxicity of oxycholesterols, vol.28, pp.387-398, 2006.

A. Berthier, S. Lemaire-ewing, C. Prunet, T. Montange, A. Vejux et al., 7-Ketocholesterol-induced apoptosis. Involvement of several pro-apoptotic but also anti-apoptotic calcium-dependent transduction pathways, FEBS J, vol.272, pp.3093-3104, 2005.

G. J. Bishop and C. Koncz, Brassinosteroids and plant steroid hormone signaling, Plant Cell, vol.14, pp.97-110, 2002.

J. Liu, D. Zhang, X. Sun, T. Ding, B. Lei et al., Structure-activity relationship of brassinosteroids and their agricultural practical usages, vol.124, pp.1-17, 2017.

A. Rosado-abon, G. De-dios-bravo, R. Rodriguez-sotres, and M. A. Iglesias-arteaga, Synthesis and plant growth promoting activity of dinorcholanic lactones bearing the 5alpha-hydroxy-6-oxo moiety, J. Steroid Biochem. Mol. Biol, vol.134, pp.45-50, 2013.

A. Rosado-abon, G. De-dios-bravo, R. Rodriguez-sotres, and M. A. Iglesias-arteaga, Synthesis and plant growth promoting activity of polyhydroxylated ketones bearing the 5alpha-hydroxy-6-oxo moiety and cholestane side chain, Steroids, vol.77, pp.461-466, 2012.

M. Romero-avila, G. De-dios-bravo, J. M. Mendez-stivalet, R. Rodriguez-sotres, and M. A. Iglesias-arteaga, Synthesis and biological activity of furostanic analogues of brassinosteroids bearing the 5alpha-hydroxy-6-oxo moiety, vol.72, pp.955-959, 2007.

C. Brosa, L. Soca, E. Terricabras, J. C. Ferrer, and A. Alsina, New synthetic brassinosteroids: a 5a-hydroxy-6-ketone analog with strong plant growth promoting activity, Tetrahedron, vol.54, pp.12337-12348, 1998.

S. D. Clouse, Brassino steroid signal transduction: from receptor kinase activation to transcriptional networks regulating plant development, Plant Cell, vol.23, pp.1219-1230, 2011.

M. Poirot, Four decades of discovery in breast cancer research and treatment -an interview with V. Craig Jordan, Int. J. Dev. Biol, vol.55, pp.703-712, 2011.

S. Chumsri, T. Howes, T. Bao, G. Sabnis, and A. Brodie, Aromatase, aromatase inhibitors, and breast cancer, J. Steroid Biochem. Mol. Biol, vol.125, pp.13-22, 2011.

V. C. Jordan and A. M. Brodie, Development and evolution of therapies targeted to the estrogen receptor for the treatment and prevention of breast cancer, Steroids, vol.72, pp.7-25, 2007.

J. Leignadier, F. Dalenc, M. Poirot, and S. Silvente-poirot, Improving the efficacy of hormone therapy in breast cancer: the role of cholesterol metabolism in SERM-mediated autophagy, cell differentiation and death, Biochem. Pharmacol, vol.144, pp.18-28, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-02380608

D. V. Kratschmar, A. Vuorinen, T. Cunha, G. Wolber, D. Classen-houben et al., Characterization of activity and binding mode of glycyrrhetinic acid derivatives inhibiting 11beta-hydroxysteroid dehydrogenase type 2, J. Steroid Biochem. Mol. Biol, vol.125, pp.129-142, 2011.

C. Zhou, F. Ye, H. Wu, H. Ye, and Q. Chen, Recent advances in the study of 11beta-Hydroxysteroid dehydrogenase type 2 (11beta-HSD2)Inhibitors, Environ. Toxicol. Pharmacol, vol.52, pp.47-53, 2017.

K. R. Beck, M. Bachler, A. Vuorinen, S. Wagner, M. Akram et al., Inhibition of 11beta-hydroxysteroid dehydrogenase 2 by the fungicides itraconazole and posaconazole, Biochem. Pharmacol, vol.130, pp.93-103, 2017.

N. Sundahl, J. Bridelance, C. Libert, K. De, I. M. Bosscher et al., Selective glucocorticoid receptor modulation: new directions with non-steroidal scaffolds, vol.152, pp.28-41, 2015.

N. Shapira, The potential contribution of dietary factors to breast cancer prevention, Eur. J. Cancer Prev, vol.26, pp.385-395, 2017.

R. T. Chlebowski, A. K. Aragaki, G. L. Anderson, C. A. Thomson, J. E. Manson et al., Low-fat dietary pattern and breast cancer mortality in the Women's health initiative randomized controlled trial, J. Clin. Oncol, vol.35, pp.2919-2926, 2017.

J. H. Parada, S. E. Steck, P. T. Bradshaw, L. S. Engel, K. Conway et al., Grilled, barbecued, and smoked meat intake and survival following breast cancer, J. Natl. Cancer Inst, vol.109, pp.299-299, 2017.

T. Fiolet, B. Srour, L. Sellem, E. Kesse-guyot, B. Es et al., Consumption of ultraprocessed foods and cancer risk: results from NutriNet-Sant e prospective cohort, vol.360, p.322, 2018.