E. Chatelais, Mort d'un cancer il y a 3 200 ans, 2014.

N. Papavramidou, T. Papavramidis, and T. Demetriou, Ancient greek and grecoroman methods in modern surgical treatment of cancer, Annals of Surgical Oncology, vol.17, pp.665-667, 2010.

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

L. S. Goodman and M. M. Wintrobe, Nitrogen mustard therapy ; use of methyl-bis (beta-chloroethyl) amine hydrochloride and tris (beta-chloroethyl) amine hydrochloride for Hodgkin's disease, lymphosarcoma, leukemia and certain allied and miscellaneous disorders, Journal of the American Medical Association, vol.132, pp.126-132, 1946.

A. Lujambio and S. W. Lowe, The microcosmos of cancer, Nature, vol.482, pp.347-355, 2012.

A. L. Oom, B. A. Humphries, and C. Yang, MicroRNAs : novel players in cancer diagnosis and therapies, BioMed Research International, vol.2014, p.959461, 2014.

J. Ferlay, E. Steliarova-foucher, J. Lortet-tieulent, S. Rosso, J. W. Coebergh et al., Cancer incidence and mortality patterns in Europe : estimates for 40 countries in 2012, European Journal of Cancer, vol.49, pp.1374-1403, 1990.

D. S. Kaufman, W. U. Shipley, and A. S. Feldman, Bladder cancer, Lancet, vol.374, pp.239-249, 2009.

M. Burger, J. W. Catto, G. Dalbagni, H. B. Grossman, H. Herr et al., Epidemiology and risk factors of urothelial bladder cancer, European Urology, vol.63, pp.234-241, 2013.

T. S. Scott, The incidence of bladder tumours in a dyestuffs factory, British Journal of Industrial Medicine, vol.9, pp.127-132, 1952.

Y. Hong, K. Song, and J. Chung, Health effects of chronic arsenic exposure, Journal of Preventive Medicine and Public Health = Yebang Uihakhoe Chi, vol.47, pp.245-252, 2014.

L. E. Moore, A. H. Smith, C. Eng, D. Kalman, S. Devries et al., Arsenic-related chromosomal alterations in bladder cancer, Journal of the National Cancer Institute, vol.94, pp.1688-1696, 2002.

R. J. Sylvester, A. P. Van-der-meijden, W. Oosterlinck, J. A. Witjes, C. Bouffioux et al., Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables : a combined analysis of 2596 patients from seven EORTC trials, European Urology, vol.49, pp.466-465, 2006.

S. D. Lokeshwar, R. Ruiz-cordero, M. C. Hupe, M. Jorda, and M. S. Soloway, Impact of 2004 ISUP/WHO classification on bladder cancer grading, World Journal of Urology, 2015.

F. Pellucchi, M. Freschi, M. Moschini, L. Rocchini, C. Maccagnano et al., Oncological predictive value of the 2004 World Health Organisation grading classification in primary T1 non-muscleinvasive bladder cancer. A step forward or back ?, BJU international, vol.115, pp.267-273, 2015.

P. Sharma, H. Kini, R. R. Pai, K. K. Sahu, and J. Kini, Study of the reproducibility of the 2004 World Health Organization classification of urothelial neoplasms, Indian Journal of Pathology & Microbiology, vol.58, pp.59-61, 2015.

B. W. Van-rhijn, M. Musquera, L. Liu, A. N. Vis, T. C. Zuiverloon et al., Molecular and clinical support for a four-tiered grading system for bladder cancer based on the WHO 1973 and 2004 classifications, Modern Pathology : An Official Journal of the United States and Canadian Academy of Pathology, Inc, vol.28, pp.695-705, 2015.

C. Premo, A. B. Apolo, P. K. Agarwal, and D. E. Citrin, Trimodality Therapy in Bladder Cancer : Who, What, and When ?, The Urologic Clinics of North America, vol.42, pp.169-180, 2015.

A. Morales, D. Eidinger, and A. W. Bruce, Intracavitary Bacillus Calmette-Guerin in the treatment of superficial bladder tumors, The Journal of Urology, vol.116, pp.180-183, 1976.

K. Kawai, J. Miyazaki, A. Joraku, H. Nishiyama, and H. Akaza, Bacillus CalmetteGuerin (BCG) immunotherapy for bladder cancer : current understanding and perspectives on engineered BCG vaccine, Cancer Science, vol.104, pp.22-27, 2013.

L. Topazio, R. Miano, V. Maurelli, G. Gaziev, M. Gacci et al., Could hyaluronic acid (HA) reduce Bacillus Calmette-Guerin (BCG) local side effects ? Results of a pilot study, BMC urology, vol.14, p.64, 2014.

J. Hsu, P. Yin, R. Wood, J. Messing, E. Messing et al., 1 alpha, 25-dihydroxylvitamin D3 promotes Bacillus Calmette-Guerin immunotherapy of bladder cancer, Oncotarget, vol.4, pp.2397-2406, 2013.

C. N. Sternberg, S. M. Donat, J. Bellmunt, R. E. Millikan, W. Stadler et al., Chemotherapy for bladder cancer : treatment guidelines for neoadjuvant chemotherapy, bladder preservation, adjuvant chemotherapy, and metastatic cancer, Urology, vol.69, pp.62-79, 2007.

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

J. M. Ruppert, K. Tokino, and D. Sidransky, Evidence for two bladder cancer suppressor loci on human chromosome 9, Cancer Research, vol.53, pp.5093-5095, 1993.

P. Cairns, M. E. Shaw, and M. A. Knowles, Preliminary mapping of the deleted region of chromosome 9 in bladder cancer, Cancer Research, vol.53, pp.1230-1232, 1993.

J. Gu, Y. Horikawa, M. Chen, C. P. Dinney, and X. Wu, Benzo(a)pyrene diol epoxide-induced chromosome 9p21 aberrations are associated with increased risk of bladder cancer, Biomarkers & Prevention : A Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology, vol.17, pp.2445-2450, 2008.

X. Wu, Urothelial tumorigenesis : a tale of divergent pathways, Nature Reviews. Cancer, vol.5, pp.713-725, 2005.

M. A. Knowles and C. D. Hurst, Molecular biology of bladder cancer : new insights into pathogenesis and clinical diversity, Nature Reviews. Cancer, vol.15, pp.25-41, 2015.

F. Massari, C. Ciccarese, M. Santoni, M. Brunelli, A. Conti et al., The route to personalized medicine in bladder cancer : where do we stand ?, Targeted Oncology, 2015.

D. R. Feldman, G. Iyer, L. Van-alstine, S. Patil, H. Al-ahmadie et al., Presence of somatic mutations within PIK3ca, AKT, RAS, and FGFR3 but not BRAF in cisplatin-resistant germ cell tumors, Clinical Cancer Research : An Official Journal of the American Association for Cancer Research, vol.20, pp.3712-3720, 2014.

D. R. Morais, S. T. Reis, N. Viana, C. B. Piantino, C. Massoco et al., The involvement of miR-100 in bladder urothelial carcinogenesis changing the expression levels of mRNA and proteins of genes related to cell proliferation, survival, apoptosis and chromosomal stability, Cancer Cell International, vol.14, issue.1, p.119, 2014.

Z. Li, X. Li, C. Yu, M. Wang, F. Peng et al., MicroRNA-100 regulates pancreatic cancer cells growth and sensitivity to chemotherapy through targeting FGFR3, Tumour Biology : The Journal of the International Society for Oncodevelopmental Biology and Medicine, vol.35, pp.11751-11759, 2014.

C. Braicu, R. Cojocneanu-petric, S. Chira, A. Truta, A. Floares et al., Clinical and pathological implications of miRNA in bladder cancer, International Journal of Nanomedicine, vol.10, pp.791-800, 2015.

T. Shimizu, H. Suzuki, M. Nojima, H. Kitamura, E. Yamamoto et al., Methylation of a panel of microRNA genes is a novel biomarker for detection of bladder cancer, European Urology, vol.63, pp.1091-1100, 2013.

D. Xie, C. Shang, H. Zhang, Y. Guo, and X. Tong, Up-regulation of miR-9 target CBX7 to regulate invasion ability of bladder transitional cell carcinoma, Medical Science Monitor : International Medical Journal of Experimental and Clinical Research, vol.21, pp.225-230, 2015.

G. Pignot, G. Cizeron-clairac, S. Vacher, A. Susini, S. Tozlu et al., microRNA expression profile in a large series of bladder tumors : identification of a 3-miRNA signature associated with aggressiveness of muscle-invasive bladder cancer, International Journal of Cancer. Journal International Du Cancer, vol.132, pp.2479-2491, 2013.

D. Ricard, A. Idbaih, F. Ducray, M. Lahutte, K. Hoang-xuan et al., Primary brain tumours in adults, Lancet, vol.379, pp.1984-1996, 2012.

L. De-angelis, Brain Tumors, N Engl J Med, issue.344, 2001.

D. N. Louis, H. Ohgaki, O. D. Wiestler, W. K. Cavenee, P. C. Burger et al., The 2007 WHO classification of tumours of the central nervous system, Acta Neuropathologica, vol.114, pp.97-109, 2007.

A. Behin, K. Hoang-xuan, A. F. Carpentier, and J. Delattre, Primary brain tumours in adults, Lancet, vol.361, pp.323-331, 2003.

M. C. Mabray, R. F. Barajas, and S. Cha, Modern brain tumor imaging, Brain Tumor Research and Treatment, vol.3, pp.8-23, 2015.

N. Martin-duverneuil, R. Guillevin, and J. Chiras, Imagerie des gliomes, Cancer/Radiotherapie, vol.12, pp.669-675, 2008.

S. Lonn, A. Ahlbom, P. Hall, M. Feychting, and S. Group, Long-term mobile phone use and brain tumor risk, American Journal of Epidemiology, vol.161, pp.526-535, 2005.

S. Larjavaara, J. Schuz, A. Swerdlow, M. Feychting, C. Johansen et al., Location of gliomas in relation to mobile telephone use : a case-case and case-specular analysis, American Journal of Epidemiology, vol.174, pp.2-11, 2011.

L. Hardell, M. Carlberg, F. Soderqvist, and K. H. Mild, Case-control study of the association between malignant brain tumours diagnosed between 2007 and 2009 and mobile and cordless phone use, International Journal of Oncology, vol.43, pp.1833-1845, 2013.

E. Cardis, B. K. Armstrong, J. D. Bowman, G. G. Giles, M. Hours et al., Risk of brain tumours in relation to estimated RF dose from mobile phones : results from five Interphone countries, Occupational and Environmental Medicine, vol.68, pp.631-640, 2011.
URL : https://hal.archives-ouvertes.fr/pasteur-00722973

S. Vida, L. Richardson, E. Cardis, D. Krewski, M. Mcbride et al., Brain tumours and cigarette smoking : analysis of the INTERPHONE Canada case-control study, Environmental Health : A Global Access Science Source, vol.13, p.55, 2014.
URL : https://hal.archives-ouvertes.fr/pasteur-01137309

C. M. Kitahara, M. S. Linet, A. V. Brenner, S. S. Wang, B. S. Melin et al.,

P. Chanock, P. Hartge, and . Rajaraman, Personal history of diabetes, genetic susceptibility to diabetes, and risk of brain glioma : a pooled analysis of observational studies, Biomarkers & Prevention : A Publication of the American Association for Cancer Research, Cosponsored by the American Society of Preventive Oncology, vol.23, pp.47-54, 2014.

C. Daumas-duport, F. Beuvon, P. Varlet, and C. Fallet-bianco, Gliomas :WHO and Sainte-Anne Hospital classifications, Ann Pathol, vol.20, pp.413-428, 2000.

M. A. Mittler, B. C. Walters, and E. G. Stopa, Observer reliability in histological grading of astrocytoma stereotactic biopsies, Journal of Neurosurgery, vol.85, pp.1091-1094, 1996.

D. Figarella-branger and C. Bouvier, Histological classification of human gliomas : state of art and controversies, Bulletin Du Cancer, vol.92, pp.301-309, 2005.

C. Daumas-duport, P. Varlet, M. L. Tucker, F. Beuvon, P. Cervera et al., Oligodendrogliomas. Part I : Patterns of growth, histological diagnosis, clinical and imaging correlations : a study of 153 cases, Journal of Neuro-Oncology, vol.34, pp.37-59, 1997.

C. Daumas-duport, M. L. Tucker, H. Kolles, P. Cervera, F. Beuvon et al., Oligodendrogliomas. Part II : A new grading system based on morphological and imaging criteria, vol.34, pp.61-78, 1997.

C. Daumas-duport, F. Beuvon, P. Varlet, and C. Fallet-bianco, Gliomas :WHO and Sainte-Anne Hospital classifications, Ann Pathol, vol.20, pp.413-428, 2000.

S. Hofer and A. Merlo, Options therapeutiques pour les gliomes malins de degre III et IV OMS, 2002.

R. Stupp, W. P. Mason, M. J. Van-den, M. Bent, B. Weller et al., European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups, and National Cancer Institute of Canada Clinical Trials Group, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, vol.352, pp.987-996, 2005.

H. Ohgaki and P. Kleihues, Genetic pathways to primary and secondary glioblastoma, The American Journal of Pathology, vol.170, pp.1445-1453, 2007.

A. Idbaih, E. Criniere, K. L. Ligon, O. Delattre, and J. Delattre, Array-based genomics in glioma research, Brain Pathology, vol.20, pp.28-38, 2010.

J. E. Bromberg, M. J. Van-den, and . Bent, Oligodendrogliomas : molecular biology and treatment, The Oncologist, vol.14, pp.155-163, 2009.

M. Nakamura, T. Watanabe, Y. Yonekawa, P. Kleihues, and H. Ohgaki, Promoter methylation of the DNA repair gene MGMT in astrocytomas is frequently associated with G :C -> A :T mutations of the TP53 tumor suppressor gene, Carcinogenesis, vol.22, pp.1715-1719, 2001.

S. A. Ciafre, S. Galardi, A. Mangiola, M. Ferracin, C. Liu et al., Extensive modulation of a set of microRNAs in primary glioblastoma, Biochemical and Biophysical Research Communications, vol.334, pp.1351-1358, 2005.

J. Silber, D. A. Lim, C. Petritsch, A. I. Persson, A. K. Maunakea et al.,

D. G. Vandenberg, C. D. Ginzinger, J. F. James, G. Costello, W. A. Bergers et al., miR-124 and miR-137 inhibit proliferation of glioblastoma multiforme cells and induce differentiation of brain tumor stem cells, BMC medicine, vol.6, p.14, 2008.

B. Malzkorn, M. Wolter, F. Liesenberg, M. Grzendowski, K. Stuhler et al., Identification and functional characterization of microRNAs involved in the malignant progression of gliomas, Brain Pathology (Zurich, Switzerland), vol.20, pp.539-550, 2010.

E. Lages, A. Guttin, M. E. Atifi, C. Ramus, H. Ipas et al., MicroRNA and target protein patterns reveal physiopathological features of glioma subtypes, PloS One, vol.6, issue.5, p.20600, 2011.
URL : https://hal.archives-ouvertes.fr/inserm-00734099

D. S. Kennedy, A. J. Bester, and S. M. Heywood, The regulation of protein synthesis by translation control RNA, Biochemical and Biophysical Research Communications, vol.61, pp.415-423, 1974.

C. Napoli, C. Lemieux, and R. Jorgensen, Introduction of a Chimeric Chalcone Synthase Gene into Petunia Results in Reversible Co-Suppression of Homologous Genes in trans, The Plant cell, vol.2, pp.279-289, 1990.

E. De-paoli, A. Dorantes-acosta, J. Zhai, M. Accerbi, D. Jeong et al.,

R. A. Meyers, P. J. Jorgensen, and . Green, Distinct extremely abundant siRNAs associated with cosuppression in petunia, RNA, vol.15, 1965.

M. Lagos-quintana, Identification of Novel Genes Coding for Small Expressed RNAs, Science, vol.294, pp.853-858, 2001.

S. Baskerville and D. P. Bartel, Microarray profiling of microRNAs reveals frequent coexpression with neighboring miRNAs and host genes, RNA, vol.11, pp.241-247, 2005.

V. N. Kim, MicroRNA biogenesis : coordinated cropping and dicing, Nat Rev Mol Cell Biol, vol.6, pp.376-385, 2005.

Y. Lee, M. Kim, J. Han, K. Yeom, S. Lee et al., MicroRNA genes are transcribed by RNA polymerase II, The EMBO Journal, vol.23, pp.4051-4060, 2004.

V. N. Kim, J. Han, and M. C. Siomi, Biogenesis of small RNAs in animals, Nature Reviews Molecular Cell Biology, vol.10, pp.126-139, 2009.

J. Winter, S. Jung, S. Keller, R. I. Gregory, and S. Diederichs, Many roads to maturity : microRNA biogenesis pathways and their regulation, Nature Cell Biology, vol.11, pp.228-234, 2009.

S. Griffiths-jones, R. J. Grocock, S. Van-dongen, A. Bateman, and A. J. Enright, miRBase : microRNA sequences, targets and gene nomenclature, Nucleic Acids Research, vol.34, pp.140-144, 2006.

S. Griffiths-jones, H. K. Saini, S. Van-dongen, and A. J. Enright, miRBase : tools for microRNA genomics, Nucleic Acids Research, vol.36, pp.154-158, 2008.

L. He and G. J. Hannon, MicroRNAs : small RNAs with a big role in gene regulation, Nature Reviews. Genetics, vol.5, pp.522-531, 2004.

G. A. Calin, C. Sevignani, C. D. Dumitru, T. Hyslop, E. Noch et al., Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers, Proceedings of the National Academy of Sciences of the United States of America, vol.101, pp.2999-3004, 2004.

E. A. Chiocca and S. E. Lawler, The many functions of microRNAs in glioblastoma, World Neurosurgery, vol.73, pp.598-601, 2010.

J. Banzhaf-strathmann and D. Edbauer, Good guy or bad guy : the opposing roles of microRNA 125b in cancer, Cell communication and signaling : CCS, vol.12, p.30, 2014.

S. Costinean, N. Zanesi, Y. Pekarsky, E. Tili, S. Volinia et al., Pre-B cell proliferation and lymphoblastic leukemia/high-grade lymphoma in E(mu)-miR155 transgenic mice, Proceedings of the National Academy of Sciences of the United States of America, vol.103, pp.7024-7029, 2006.

S. D. Selcuklu, M. T. Donoghue, and C. Spillane, miR-21 as a key regulator of oncogenic processes, Biochemical Society Transactions, vol.37, pp.918-925, 2009.

G. A. Calin, C. D. Dumitru, M. Shimizu, R. Bichi, S. Zupo et al., Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.15524-15529, 2002.

A. Cimmino, G. A. Calin, M. Fabbri, M. V. Iorio, M. Ferracin et al.,

R. I. Wojcik, S. Aqeilan, M. Zupo, L. Dono, H. Rassenti et al.,

G. Liu, T. J. Kipps, M. Negrini, and C. M. Croce, miR-15 and miR-16 induce apoptosis by targeting BCL2, Proceedings of the National Academy of Sciences of the United States of America, vol.102, pp.13944-13949, 2005.

C. S. Fuziwara and E. T. Kimura, Insights into Regulation of the miR-17-92 Cluster of miRNAs in Cancer, Frontiers in Medicine, vol.2, p.64, 2015.

E. Lages, H. Ipas, A. Guttin, H. Nesr, F. Berger et al., MicroRNAs : molecular features and role in cancer, Frontiers in Bioscience (Landmark Edition), vol.17, pp.2508-2540, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00733975

, Micro-arn impliques dans la degenerescence maculaire

S. Wang, K. M. Koster, Y. He, and Q. Zhou, miRNAs as potential therapeutic targets for age-related macular degeneration, Future Medicinal Chemistry, vol.4, pp.277-287, 2012.

C. Jopling, Liver-specific microRNA-122 : Biogenesis and function, RNA biology, vol.9, pp.137-142, 2012.

R. E. Lanford, E. S. Hildebrandt-eriksen, A. Petri, R. Persson, M. Lindow et al., Therapeutic Silencing of MicroRNA-122 in Primates with Chronic Hepatitis C Virus Infection, Science, vol.327, pp.198-201, 2010.

R. Kanasty, J. R. Dorkin, A. Vegas, and D. Anderson, Delivery materials for siRNA therapeutics, Nature Materials, vol.12, pp.967-977, 2013.

J. Krutzfeldt, S. Kuwajima, R. Braich, K. G. Rajeev, J. Pena et al., Specificity, duplex degradation and subcellular localization of antagomirs, Nucleic Acids Research, vol.35, issue.9, pp.2885-2892, 2007.

A. Griveau, J. Bejaud, S. Anthiya, S. Avril, D. Autret et al., Silencing of miR-21 by locked nucleic acid-lipid nanocapsule complexes sensitize human glioblastoma cells to radiation-induced cell death, International Journal of Pharmaceutics, vol.454, pp.765-774, 2013.

V. Sanna, N. Pala, and M. Sechi, Targeted therapy using nanotechnology : focus on cancer, International Journal of Nanomedicine, vol.9, pp.467-483, 2014.

N. Tominaga, Y. Yoshioka, and T. Ochiya, A novel platform for cancer therapy using extracellular vesicles, Advanced Drug Delivery Reviews, vol.95, pp.50-55, 2015.

Y. Zhao, Y. Li, L. Wang, H. Yang, Q. Wang et al., microRNA response elements-regulated TRAIL expression shows specific survival-suppressing activity on bladder cancer, Journal of experimental & clinical cancer research : CR, vol.32, p.10, 2013.

M. Chalopin, A. Tesse, M. C. Martinez, D. Rognan, J. Arnal et al., Estrogen Receptor Alpha as a Key Target of Red Wine Polyphenols Action on the Endothelium, PLoS ONE, vol.5, p.8554, 2010.
URL : https://hal.archives-ouvertes.fr/inserm-00504826

M. Hasan, Biological Activities Importance of Chalcone Derivatives, International Journal of Chemical and Biomolecular Science, vol.1, pp.107-112, 2015.

B. Orlikova, D. Tasdemir, F. Golais, M. Dicato, and M. Diederich, Dietary chalcones with chemopreventive and chemotherapeutic potential, Genes & Nutrition, vol.6, pp.125-147, 2011.

D. I. Batovska and I. T. Todorova, Trends in utilization of the pharmacological potential of chalcones, Current Clinical Pharmacology, vol.5, pp.1-29, 2010.

A. Boumendjel, X. Ronot, and J. Boutonnat, Chalcones derivatives acting as cell cycle blockers : potential anti cancer drugs ?, Current drug targets, vol.10, pp.363-371, 2009.

A. Boumendjel, J. Boccard, P. Carrupt, E. Nicolle, M. Blanc et al., Antimitotic and antiproliferative activities of chalcones : forward structure-activity relationship, Journal of Medicinal Chemistry, vol.51, pp.2307-2310, 2008.

E. Zhang, R. Wang, S. Guo, and B. Liu, An update on antitumor activity of naturally occurring chalcones, Evidence-Based Complementary and Alternative Medicine : eCAM, vol.2013, p.815621, 2013.

T. Ji, C. Lin, L. S. Krill, R. Eskander, Y. Guo et al., Flavokawain B, a kava chalcone, inhibits growth of human osteosarcoma cells through G2/M cell cycle arrest and apoptosis, Molecular Cancer, vol.12, p.55, 2013.

W. Wu, H. Ye, L. Wan, X. Han, G. Wang et al., Millepachine, a novel chalcone, induces G2/M arrest by inhibiting CDK1 activity and causing apoptosis via ROS-mitochondrial apoptotic pathway in human hepatocarcinoma cells in vitro and in vivo, Carcinogenesis, vol.34, pp.1636-1643, 2013.

R. N. Eskander, L. M. Randall, T. Sakai, Y. Guo, B. Hoang et al., Flavokawain B, a novel, naturally occurring chalcone, exhibits robust apoptotic effects and induces G2/M arrest of a uterine leiomyosarcoma cell line, The Journal of Obstetrics and Gynaecology Research, vol.38, pp.1086-1094, 2012.

A. Bouchet, A. Boumendjel, E. Khalil, R. Serduc, E. Brauer et al., Chalcone JAI-51 improves efficacy of synchrotron microbeam radiation therapy of brain tumors, Journal of Synchrotron Radiation, vol.19, pp.478-482, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00856984

K. Shen, J. Chang, Y. Hsu, and P. Kuo, Chalcone arrests cell cycle progression and induces apoptosis through induction of mitochondrial pathway and inhibition of nuclear factor kappa B signalling in human bladder cancer cells, Basic & Clinical Pharmacology & Toxicology, vol.101, pp.254-261, 2007.

K. A. Youdim, M. Z. Qaiser, D. J. Begley, C. A. Rice-evans, and N. J. Abbott, Flavonoid permeability across an in situ model of the blood-brain barrier, Free Radical Biology & Medicine, vol.36, pp.592-604, 2004.

P. Bedford, S. A. Shellard, M. C. Walker, R. D. Whelan, J. R. Masters et al.,

. Hill, Differential expression of collateral sensitivity or resistance to cisplatin in human bladder carcinoma cell lines pre-exposed in vitro to either X-irradiation or cisplatin, International Journal of Cancer. Journal International Du Cancer, vol.40, pp.681-686, 1987.

H. Phillips, S. Kharbanda, R. Chen, W. Forrest, R. Soriano et al., Molecular subclasses of high-grade glioma predict prognosis, delineata a pattern of disease progression, and resemble stages in neurogenesis, Cancer Cell, vol.9, pp.157-173, 2006.

A. Li, J. Walling, S. Ahn, Y. Kotliarov, Q. Su et al., Unsupervised analysis of transcriptomic profiles reveals six glioma subtypes, Cancer research, vol.69, pp.2091-2099, 2009.

R. G. Verhaak, K. A. Hoadley, E. Purdom, V. Wang, Y. Qi et al.,

L. Miller, T. Ding, J. P. Golub, and . Mesirov, Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1, Cancer Cell, vol.17, pp.98-110, 2010.

J. R. Perkins, J. M. Dawes, S. B. Mcmahon, D. L. Bennett, C. Orengo et al., ReadqPCR and NormqPCR : R packages for the reading, quality checking and normalisation of RT-qPCR quantification cycle (Cq) data, BMC genomics, vol.13, p.296, 2012.

P. J. Sykes, S. H. Neoh, M. J. Brisco, E. Hughes, J. Condon et al., Quantitation of targets for PCR by use of limiting dilution, BioTechniques, vol.13, pp.444-449, 1992.

T. Mosmann, Rapid colorimetric assay for cellular growth and survival : application to proliferation and cytotoxicity assays, Journal of Immunological Methods, vol.65, pp.55-63, 1983.

R. A. Irizarry, B. M. Bolstad, F. Collin, L. M. Cope, B. Hobbs et al., Summaries of Affymetrix GeneChip probe level data, Nucleic Acids Research, vol.31, p.15, 2003.

V. G. Tusher, R. Tibshirani, and G. Chu, Significance analysis of microarrays applied to the ionizing radiation response, Proceedings of the National Academy of Sciences of the United States of America, vol.98, pp.5116-5121, 2001.

J. Vandesompele, K. De, F. Preter, B. Pattyn, N. Poppe et al., Accurate normalization of real-time quantitative RT-PCR data by geometric averaging of multiple internal control genes, Genome Biology, vol.3, p.34, 2002.

J. S. Mcdonald, D. Milosevic, H. V. Reddi, S. K. Grebe, and A. Algecirasschimnich, Analysis of circulating microRNA : preanalytical and analytical challenges, Clinical Chemistry, vol.57, pp.833-840, 2011.

J. Jarry, D. Schadendorf, C. Greenwood, A. Spatz, and L. C. Van-kempen, The validity of circulating microRNAs in oncology : five years of challenges and contradictions, Molecular Oncology, vol.8, pp.819-829, 2014.

M. B. Eisen, P. T. Spellman, P. O. Brown, and D. Botstein, Cluster analysis and display of genome-wide expression patterns, Proceedings of the National Academy of Sciences of the United States of America, vol.95, pp.14863-14868, 1998.

D. Hua, F. Mo, D. Ding, L. Li, X. Han et al., A catalogue of glioblastoma and brain MicroRNAs identified by deep sequencing, Omics : A Journal of Integrative Biology, vol.16, pp.690-699, 2012.

P. Guo, Q. Nie, J. Lan, J. Ge, Y. Qiu et al., C-Myc negatively controls the tumor suppressor PTEN by upregulating miR-26a in glioblastoma multiforme cells, Biochemical and Biophysical Research Communications, vol.441, pp.186-190, 2013.

B. Liu, X. Wu, B. Liu, C. Wang, Y. Liu et al., MiR-26a enhances metastasis potential of lung cancer cells via AKT pathway by targeting PTEN, Biochimica Et Biophysica Acta, vol.1822, pp.1692-1704, 2012.

J. Chen, X. Fu, Y. Wan, Z. Wang, D. Jiang et al., miR-125b inhibitor enhance the chemosensitivity of glioblastoma stem cells to temozolomide by targeting Bak1, Tumour biology : the journal of the International Society for Oncodevelopmental Biology and Medicine, 2014.

G. Tezcan, B. Tunca, A. Bekar, M. Preusser, A. S. Berghoff et al., microRNA Expression Pattern Modulates Temozolomide Response in GBM Tumors with Cancer Stem Cells, Cellular and molecular neurobiology, 2014.

M. Zhou, Z. Liu, Y. Zhao, Y. Ding, H. Liu et al., MicroRNA-125b confers the resistance of breast cancer cells to paclitaxel through suppression of pro-apoptotic Bcl-2 antagonist killer 1 (Bak1) expression, The Journal of Biological Chemistry, vol.285, pp.21496-21507, 2010.

S. Liu, X. Pan, Q. Yang, L. Wen, Y. Jiang et al., MicroRNA18a enhances the radiosensitivity of cervical cancer cells by promoting radiationinduced apoptosis, Oncology Reports, vol.33, pp.2853-2862, 2015.

Y. Song, P. Wang, W. Zhao, Y. Yao, X. Liu et al., MiR-18a regulates the proliferation, migration and invasion of human glioblastoma cell by targeting neogenin, Experimental Cell Research, vol.324, pp.54-64, 2014.

S. Haemmig, U. Baumgartner, A. Gluck, S. Zbinden, M. P. Tschan et al., miR-125b controls apoptosis and temozolomide resistance by targeting TNFAIP3 and NKIRAS2 in glioblastomas, Cell Death & Disease, vol.5, p.1279, 2014.

A. Boumendjel, A. Mcleer-florin, P. Champelovier, D. Allegro, D. Muhammad et al., A novel chalcone derivative which acts as a microtubule depolymerising agent and an inhibitor of P-gp and BCRP in in-vitro and in-vivo glioblastoma models, BMC Cancer, vol.9, issue.1, p.242, 2009.

A. Boumendjel, X. Ronot, and J. Boutonnat, Chalcones derivatives acting as cell cycle blockers : potential anti cancer drugs ?, Current drug targets, vol.10, pp.363-371, 2009.

V. P. Androutsopoulos, A. Papakyriakou, D. Vourloumis, and D. A. Spandidos, Comparative CYP1a1 and CYP1b1 substrate and inhibitor profile of dietary flavonoids, Bioorganic & Medicinal Chemistry, vol.19, pp.2842-2849, 2011.

J. Lu, G. Getz, E. A. Miska, E. Alvarez-saavedra, J. Lamb et al.,

T. R. Horvitz and . Golub, MicroRNA expression profiles classify human cancers, Nature, vol.435, pp.834-838, 2005.

M. Srinivasan, D. Sedmak, and S. Jewell, Effect of Fixatives and Tissue Processing on the Content and Integrity of Nucleic Acids, The American Journal of Pathology, vol.161, 1961.

J. Song, Z. Bai, W. Han, J. Zhang, H. Meng et al., Identification of suitable reference genes for qPCR analysis of serum microRNA in gastric cancer patients, Digestive Diseases and Sciences, vol.57, pp.897-904, 2012.

N. Yang, S. Kaur, S. Volinia, J. Greshock, H. Lassus et al., MicroRNA microarray identifies Let-7i as a novel biomarker and therapeutic target in human epithelial ovarian cancer, Cancer Research, vol.68, pp.10307-10314, 2008.

K. Kelnar, H. J. Peltier, N. Leatherbury, J. Stoudemire, and A. G. Bader, Quantification of Therapeutic miRNA Mimics in Whole Blood from Nonhuman Primates, Analytical Chemistry, vol.86, pp.1534-1542, 2014.

E. Van-rooij and S. Kauppinen, Development of microRNA therapeutics is coming of age, EMBO Molecular Medicine, vol.6, pp.851-864, 2014.

N. Ratert, H. Meyer, M. Jung, P. Lioudmer, H. Mollenkopf et al., miRNA Profiling Identifies Candidate miRNAs for Bladder Cancer Diagnosis and Clinical Outcome, The Journal of Molecular Diagnostics, vol.15, pp.695-705, 2013.

B. John, A. J. Enright, A. Aravin, T. Tuschl, C. Sander et al., Human MicroRNA targets, PLoS biology, vol.2, p.363, 2004.

A. J. Enright, B. John, U. Gaul, T. Tuschl, C. Sander et al., MicroRNA targets in Drosophila, Genome Biology, vol.5, issue.1, p.1, 2003.

V. Agarwal, G. W. Bell, J. Nam, and D. P. Bartel, Predicting effective microRNA target sites in mammalian mRNAs, vol.4, 2015.

M. D. Paraskevopoulou, G. Georgakilas, N. Kostoulas, I. S. Vlachos, T. Vergoulis et al., DIANA-microT web server v5.0 : service integration into miRNA functional analysis workflows, Nucleic Acids Research, vol.41, pp.169-173, 2013.

A. Krek, D. Grun, M. N. Poy, R. Wolf, L. Rosenberg et al., Combinatorial microRNA target predictions, Nature Genetics, vol.37, pp.495-500, 2005.

J. C. Huang, T. Babak, T. W. Corson, G. Chua, S. Khan et al., Using expression profiling data to identify human microRNA targets, Nature Methods, vol.4, pp.1045-1049, 2007.

N. Wong and X. Wang, miRDB : an online resource for microRNA target prediction and functional annotations, Nucleic Acids Research, vol.43, pp.146-152, 2015.

C. Coronnello and P. V. Benos, ComiR : combinatorial microRNA target prediction tool, Nucleic Acids Research, vol.41, pp.159-164, 2013.

Y. Friedman, G. Naamati, and M. Linial, MiRror : a combinatorial analysis web tool for ensembles of microRNAs and their targets, Bioinformatics, vol.26, pp.1920-1921, 2010.

H. Dweep and N. Gretz, miRWalk2.0 : a comprehensive atlas of microRNA-target interactions, vol.12, p.697, 2015.

F. Xiao, Z. Zuo, G. Cai, S. Kang, X. Gao et al., miRecords : an integrated resource for microRNA-target interactions, Nucleic Acids Research, vol.37, pp.105-110, 2009.

R. S. Geary, Antisense oligonucleotide pharmacokinetics and metabolism, Expert Opinion on Drug Metabolism & Toxicology, vol.5, pp.381-391, 2009.

M. S. Ebert, J. R. Neilson, and P. A. Sharp, MicroRNA sponges : competitive inhibitors of small RNAs in mammalian cells, Nature Methods, vol.4, pp.721-726, 2007.

F. Moshiri, E. Callegari, L. D'abundo, F. Corra, L. Lupini et al., Inhibiting the oncogenic mir-221 by microRNA sponge : toward microRNAbased therapeutics for hepatocellular carcinoma, Gastroenterology and Hepatology from Bed to Bench, vol.7, issue.1, pp.43-54, 2014.

Z. Wang, The principles of MiRNA-masking antisense oligonucleotides technology, Methods in Molecular Biology, vol.676, pp.43-49, 2011.

B. John, A. J. Enright, A. Aravin, T. Tuschl, C. Sander et al., Human MicroRNA targets, PLoS biology, vol.2, p.363, 2004.

D. P. Bartel, MicroRNAs : Target Recognition and Regulatory Functions, Cell, vol.136, pp.215-233, 2009.

S. Obad, C. O. Santos, A. Petri, M. Heidenblad, O. Broom et al.,

S. Hannon and . Kauppinen, Silencing of microRNA families by seed-targeting tiny LNAs, Nature Genetics, vol.43, pp.371-378, 2011.

D. Zheng, Y. Dai, S. Wang, and X. Xing, MicroRNA-299-3p promotes the sensibility of lung cancer to doxorubicin through directly targeting ABCE1, International Journal of Clinical and Experimental Pathology, vol.8, issue.9, pp.10072-10081, 2015.

P. Zhang, L. Wang, C. Rodriguez-aguayo, Y. Yuan, B. G. Debeb et al., miR-205 acts as a tumour radiosensitizer by targeting ZEB1 and Ubc13, vol.5, p.5671, 2014.

X. Shi, M. Zou, J. He, H. Xie, and X. Li, Studies on the identification of constituents in ethanol extract of Radix Glycyrrhizae and their anticancer activity, AJTCAM / African Networks on Ethnomedicines, vol.11, issue.2, pp.334-338, 2014.

J. P. Schroder-van-der-elst, D. Van-der-heide, H. Rokos, G. Morreale-de-escobar, and J. Kohrle, Synthetic flavonoids cross the placenta in the rat and are found in fetal brain, The American Journal of Physiology, vol.274, pp.253-256, 1998.

D. Kavvadias, P. Sand, K. A. Youdim, M. Z. Qaiser, C. Rice-evans et al., The flavone hispidulin, a benzodiazepine receptor ligand with positive allosteric properties, traverses the blood-brain barrier and exhibits anticonvulsive effects, British Journal of Pharmacology, vol.142, pp.811-820, 2004.

M. Vidak, D. Rozman, and R. Komel, Effects of Flavonoids from Food and Dietary Supplements on Glial and Glioblastoma Multiforme Cells, Molecules, vol.20, issue.10, pp.19406-19432, 2015.

A. I. Loaiza-perez, S. Kenney, J. Boswell, M. Hollingshead, M. C. Alley et al., Aryl hydrocarbon receptor activation of an antitumor aminoflavone : basis of selective toxicity for MCF-7 breast tumor cells, Molecular Cancer Therapeutics, vol.3, pp.715-725, 2004.

M. Callero, G. Suarez, G. Luzzani, B. Itkin, B. Nguyen et al., Aryl hydrocarbon receptor activation by aminoflavone : New molecular target for renal cancer treatment, International Journal of Oncology, 2012.

V. P. Androutsopoulos, A. M. Tsatsakis, and D. A. Spandidos, Cytochrome P450 CYP1a1 : wider roles in cancer progression and prevention, BMC Cancer, vol.9, issue.1, p.187, 2009.

T. Shimada, K. Tanaka, S. Takenaka, N. Murayama, M. V. Martin et al., Structure-function relationships of inhibition of human cytochromes P450 1a1, 1a2, 1b1, 2c9, and 3a4 by 33 flavonoid derivatives, Chemical Research in Toxicology, vol.23, pp.1921-1935, 2010.

L. Mclean, U. Soto, K. Agama, J. Francis, R. Jimenez et al., Aminoflavone induces oxidative DNA damage and reactive oxidative species-mediated apoptosis in breast cancer cells, International Journal of Cancer. Journal International Du Cancer, vol.122, pp.1665-1674, 2008.

A. Joiakim, P. A. Mathieu, A. A. Elliott, and J. J. Reiners, Superinduction of CYP1a1 in MCF10a cultures by cycloheximide, anisomycin, and puromycin : a process independent of effects on protein translation and unrelated to suppression of aryl hydrocarbon receptor proteolysis by the proteasome, Molecular Pharmacology, vol.66, pp.936-947, 2004.

C. Weiss, D. Faust, H. Durk, S. K. Kolluri, A. Pelzer et al., TCDD induces c-jun expression via a novel ah (dioxin) receptor-mediated p38-MAPK-dependent pathway, Oncogene, vol.24, pp.4975-4983, 2005.

Z. Zhang, S. Chen, H. Mei, J. Xuan, X. Guo et al., Ginkgo biloba leaf extract induces DNA damage by inhibiting topoisomerase II activity in human hepatic cells, Scientific Reports, vol.5, p.14633, 2015.

N. Yamashita and S. Kawanishi, Distinct mechanisms of DNA damage in apoptosis induced by quercetin and luteolin, Free Radical Research, vol.33, pp.623-633, 2000.

M. Chakrabarti and S. K. Ray, Anti-tumor activities of luteolin and silibinin in glioblastoma cells : overexpression of miR-7-1-3p augmented luteolin and silibinin to inhibit autophagy and induce apoptosis in glioblastoma in vivo, Apoptosis : An International Journal on Programmed Cell Death, 2015.

R. J. Tallarida and R. B. Raffa, The application of drug dose equivalence in the quantitative analysis of receptor occupation and drug combinations, Pharmacology & Therapeutics, vol.127, pp.165-174, 2010.

C. Mackellar, D. Graham, D. W. Will, S. Burgess, and T. Brown, Synthesis and physical properties of anti-HIV antisense oligonucleotides bearing terminal lipophilic groups, Nucleic Acids Research, vol.20, pp.3411-3417, 1992.

J. Krutzfeldt, N. Rajewsky, R. Braich, K. G. Rajeev, T. Tuschl et al., Silencing of microRNAs in vivo with 'antagomirs, Nature, vol.438, pp.685-689, 2005.

G. Leriche, L. Chisholm, and A. Wagner, Cleavable linkers in chemical biology, Bioorganic & Medicinal Chemistry, vol.20, pp.571-582, 2012.

T. Fukami and T. Yokoi, The emerging role of human esterases, Drug Metabolism and Pharmacokinetics, vol.27, issue.5, pp.466-477, 2012.

H. Yang, C. Huang, C. Lin, H. Liu, C. Huang et al., Gadolinium-functionalized nanographene oxide for combined drug and microRNA delivery and magnetic resonance imaging, Biomaterials, vol.35, pp.6534-6542, 2014.

H. Ipas, A. Guttin, A. Guttin, C. Zenga, F. Berger et al., Additional Clues for a Protective Role of Vitamin D in Neurodegenerative Diseases : 1,25-Dihydroxyvitamin D3 Triggers an Anti-Inflammatory Response in Brain Pericyte, Production Scientifique Publications, vol.1, 2014.

M. F. Nissou, M. El-atifi, A. Guttin, C. Godfraind, C. Salon et al., Hypoxia-induced expression of VE-cadherin and filamin B in glioma cell cultures and pseudopalisade structures, J Neurooncol, vol.16, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00851136

M. F. Nissou, J. Brocard, M. El-atifi, A. Guttin, A. Andrieux et al., The transcriptomic response of mixed neuron-glial cell cultures to 1,25-dihydroxyvitamin d3 includes genes limiting the progression of neurodegenerative diseases, PMID : 23455988. PMCID : PMC3962683. 5 : Dupont PY, vol.113, p.3477019, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00838410

E. Lages, H. Ipas, A. Guttin, H. Nesr, F. Berger et al., MicroRNAs : molecular features and role in cancer Front Biosci (Landmark Ed), vol.17, p.3815439, 2012.

E. Lages, A. Guttin, M. El-atifi, C. Ramus, H. Ipas et al.,

C. , D. F. Dhobb, M. Pelletier, L. Wion, D. Gay et al., The heterogeneity of meningioma revealed by multiparameter analysis : infiltrative and non-infiltrative clinical phenotypes, Int J Oncol, vol.6, issue.5, p.21318223, 2011.

. Brevets,

A. Guttin, H. Ipas, M. El-atifi, F. Berger, J. Issartel et al., Développement d'une thé-rapie dépendante des microARN pour les tumeurs solides, Méthodes de diagnostic in vitro intra/inter tissulaire Inventeurs : Lages E