D. Louis, A. Perry, G. Reifenberger, A. Von-deimling, D. Figarella-branger et al., The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary, Acta Neuropathologica, vol.45, issue.6, pp.803-820, 2016.
DOI : 10.1007/s00401-016-1545-1

URL : https://hal.archives-ouvertes.fr/hal-01479018

R. Stupp, M. Hegi, W. Mason, M. Van-den-bent, M. Taphoorn et al., Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, The Lancet Oncology, vol.10, issue.5, pp.459-466, 2009.
DOI : 10.1016/S1470-2045(09)70025-7

M. Teo, S. Martin, K. Owusu-agyemang, S. Nowicki, C. B. Mackinnon et al., A survival analysis of GBM patients in the West of Scotland pre- and post-introduction of the Stupp regime, British Journal of Neurosurgery, vol.75, issue.3, pp.351-355, 2014.
DOI : 10.3171/2011.2.JNS10998

R. Stupp, S. Taillibert, A. Kanner, S. Kesari, D. Steinberg et al., Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma, JAMA, vol.314, issue.23, pp.2535-2543, 2015.
DOI : 10.1001/jama.2015.16669

J. Lathia, S. Mack, E. Mulkearns-hubert, C. Valentim, and J. Rich, Cancer stem cells in glioblastoma, Genes & Development, vol.29, issue.12, pp.1203-1217, 2015.
DOI : 10.1101/gad.261982.115

R. Verhaak, K. Hoadley, E. Purdom, V. Wang, Y. Qi et al., Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1, Cancer Cell, vol.17, issue.1, pp.98-110, 2010.
DOI : 10.1016/j.ccr.2009.12.020

E. Claus, K. Walsh, J. Wiencke, A. Molinaro, J. Wiemels et al., Survival and low-grade glioma: the emergence of genetic information, Neurosurgical Focus, vol.38, issue.1, p.6, 2015.
DOI : 10.3171/2014.10.FOCUS12367

S. Ilkanizadeh, J. Lau, M. Huang, D. Foster, R. Wong et al., Glial Progenitors as Targets for Transformation in Glioma, Adv Cancer Res, vol.121, pp.1-65, 2014.
DOI : 10.1016/B978-0-12-800249-0.00001-9

H. Phillips, S. Kharbanda, R. Chen, W. Forrest, R. Soriano et al., Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis, Cancer Cell, vol.9, issue.3, pp.157-173, 2006.
DOI : 10.1016/j.ccr.2006.02.019

J. Hale, M. Sinyuk, J. Rich, and J. Lathia, Decoding the cancer stem cell hypothesis in glioblastoma, CNS Oncology, vol.2, issue.4, pp.319-330, 2013.
DOI : 10.2217/cns.13.23

J. Chen, Y. Li, T. Yu, R. Mckay, D. Burns et al., A restricted cell population propagates glioblastoma growth after chemotherapy, Nature, vol.2, issue.7412, pp.522-526, 2012.
DOI : 10.1038/nature11287

C. Morshead and D. Van-der-kooy, Disguising adult neural stem cells, Current Opinion in Neurobiology, vol.14, issue.1, pp.125-131, 2004.
DOI : 10.1016/j.conb.2004.01.008

N. Sanai, A. Alvarez-buylla, and M. Berger, Neural Stem Cells and the Origin of Gliomas, New England Journal of Medicine, vol.353, issue.8, pp.811-822, 2005.
DOI : 10.1056/NEJMra043666

N. Goffart, J. Kroonen, and B. Rogister, Glioblastoma-Initiating Cells: Relationship with Neural Stem Cells and the Micro-Environment, Cancers, vol.5, issue.3, pp.1049-1071, 2013.
DOI : 10.3390/cancers5031049

S. Bao, Q. Wu, R. Mclendon, Y. Hao, Q. Shi et al., Glioma stem cells promote radioresistance by preferential activation of the DNA damage response, Nature, vol.8, issue.7120, pp.756-760, 2006.
DOI : 10.1038/nature05236

G. Kao, Z. Jiang, A. Fernandes, A. Gupta, and A. Maity, Inhibition of Phosphatidylinositol-3-OH Kinase/Akt Signaling Impairs DNA Repair in Glioblastoma Cells following Ionizing Radiation, Journal of Biological Chemistry, vol.282, issue.29, pp.21206-21212, 2007.
DOI : 10.1074/jbc.M703042200

B. Campos, Z. Gal, A. Baader, T. Schneider, C. Sliwinski et al., Aberrant self-renewal and quiescence contribute to??the??aggressiveness of glioblastoma, The Journal of Pathology, vol.4, issue.1, pp.23-33, 2014.
DOI : 10.1002/path.4366

A. Murat, E. Migliavacca, T. Gorlia, W. Lambiv, T. Shay et al., Stem Cell???Related ???Self-Renewal??? Signature and High Epidermal Growth Factor Receptor Expression Associated With Resistance to Concomitant Chemoradiotherapy in Glioblastoma, Journal of Clinical Oncology, vol.26, issue.18, pp.3015-3024, 2008.
DOI : 10.1200/JCO.2007.15.7164

G. Chen, J. Kong, C. Tucker-burden, M. Anand, Y. Rong et al., Human Brat Ortholog TRIM3 Is a Tumor Suppressor That Regulates Asymmetric Cell Division in Glioblastoma, Cancer Research, vol.74, issue.16, pp.4536-4548, 2014.
DOI : 10.1158/0008-5472.CAN-13-3703

K. Lewis and C. Petritsch, Abstract, Translational Neuroscience, vol.4, issue.4, pp.484-503, 2013.
DOI : 10.2478/s13380-013-0148-8

URL : https://hal.archives-ouvertes.fr/hal-01336771

C. Patru, L. Romao, P. Varlet, L. Coulombel, E. Raponi et al., CD133, CD15/SSEA-1, CD34 or side populations do not resume tumor-initiating properties of long-term cultured cancer stem cells from human malignant glio-neuronal tumors, BMC Cancer, vol.4, issue.5, p.66, 2010.
DOI : 10.1016/j.stem.2009.03.003

URL : https://hal.archives-ouvertes.fr/inserm-00663773

C. Thirant, E. Galan-moya, L. Dubois, S. Pinte, P. Chafey et al., Differential Proteomic Analysis of Human Glioblastoma and Neural Stem Cells Reveals HDGF as a Novel Angiogenic Secreted Factor, STEM CELLS, vol.30, issue.Pt 1, pp.845-853, 2012.
DOI : 10.1002/stem.1062

E. Galan-moya, L. Guelte, A. , L. Fernandes, E. Thirant et al., Secreted factors from brain endothelial cells maintain glioblastoma stem-like cell expansion through the mTOR pathway. EMBO reports, pp.470-476, 2011.

L. Guelte, A. Galan-moya, E. Dwyer, J. Treps, L. Kettler et al., Semaphorin 3A elevates endothelial cell permeability through PP2A inactivation, Journal of Cell Science, vol.125, issue.17, pp.4137-4146, 2012.
DOI : 10.1242/jcs.108282

L. Treps, S. Edmond, E. Harford-wright, E. Galan-moya, A. Schmitt et al., Extracellular vesicle-transported Semaphorin3A promotes vascular permeability in glioblastoma, Oncogene, vol.119, issue.20, 2015.
DOI : 10.1038/ncomms7999

URL : https://hal.archives-ouvertes.fr/inserm-01406334

S. Azzi, L. Treps, H. Leclair, H. Ngo, E. Harford-wright et al., Desert Hedgehog/Patch2 Axis Contributes to Vascular Permeability and Angiogenesis in Glioblastoma, Frontiers in Pharmacology, vol.44, issue.Pt 17, p.281, 2015.
DOI : 10.1097/MPA.0000000000000532

URL : https://hal.archives-ouvertes.fr/inserm-01247505

E. Galan-moya, L. Treps, L. Oliver, H. Chneiweiss, F. Vallette et al., Endothelial Secreted Factors Suppress Mitogen Deprivation-Induced Autophagy and Apoptosis in Glioblastoma Stem-Like Cells, PLoS ONE, vol.13, issue.3, p.93505, 2014.
DOI : 10.1371/journal.pone.0093505.g004

URL : https://hal.archives-ouvertes.fr/inserm-01075054

K. Tjioe, . Tod, and J. Gavard, Luteolin impacts on the DNAdamage pathway in oral squamous cell carcinoma, Nutr Cancer, 2016.

Z. Zhang, J. Gao, X. Cai, Y. Zhao, Y. Wang et al., Escin Sodium Induces Apoptosis of Human Acute Leukemia Jurkat T Cells, Phytotherapy Research, vol.75, issue.12, pp.1747-1755, 2011.
DOI : 10.1002/ptr.3457

Y. Zhang, S. Wang, J. Ma, H. Li, J. Ye et al., EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition, Journal of Neuro-Oncology, vol.1542, issue.1, pp.41-52, 2015.
DOI : 10.1007/s11060-014-1604-1

C. Sirtori, Aescin: pharmacology, pharmacokinetics and therapeutic profile, Pharmacological Research, vol.44, issue.3, pp.183-193, 2001.
DOI : 10.1006/phrs.2001.0847

O. Carrasco, A. Ranero, E. Hong, and H. Vidrio, Endothelial Function Impairment in Chronic Venous Insufficiency: Effect of Some Cardiovascular Protectant Agents, Angiology, vol.60, issue.6, pp.763-771, 2009.
DOI : 10.1177/0003319709332108

Y. Zhou, X. Hui, N. Li, W. Zhuang, G. Liu et al., Saponins from Chinese Buckeye Seed Reduce Cerebral Edema: Metaanalysis of Randomized Controlled Trials, Planta Medica, vol.71, issue.11, pp.993-998, 2005.
DOI : 10.1055/s-2006-946616

B. Ottillinger and K. Greeske, Rational therapy of chronic venous insufficiency ??? chances and limits of the therapeutic use of horse-chestnut seeds extract, BMC Cardiovascular Disorders, vol.4, issue.2, p.5, 2001.
DOI : 10.1016/S0014-2999(96)00645-0

M. Li, C. Lu, L. Zhang, J. Zhang, Y. Du et al., Oral Administration of Escin Inhibits Acute Inflammation and Reduces Intestinal Mucosal Injury in Animal Models, Evidence-Based Complementary and Alternative Medicine, vol.9, issue.3, p.503617, 2015.
DOI : 10.1152/ajpgi.00095.2013

D. Wetzel, W. Menke, R. Dieter, V. Smasal, B. Giannetti et al., Escin/diethylammonium salicylate/heparin combination gels for the topical treatment of acute impact injuries: a randomised, double blind, placebo controlled, multicentre study, British Journal of Sports Medicine, vol.36, issue.3, pp.183-188, 2002.
DOI : 10.1136/bjsm.36.3.183

A. Rimmon, A. Vexler, L. Berkovich, G. Earon, R. I. Lev-ari et al., Escin Chemosensitizes Human Pancreatic Cancer Cells and Inhibits the Nuclear Factor-kappaB Signaling Pathway, Biochemistry Research International, vol.11, issue.11, p.251752, 2013.
DOI : 10.1111/j.1749-6632.2010.05852.x

J. Patlolla, L. Qian, L. Biddick, Y. Zhang, D. Desai et al., ??-Escin Inhibits NNK-Induced Lung Adenocarcinoma and ALDH1A1 and RhoA/Rock Expression in A/J Mice and Growth of H460 Human Lung Cancer Cells, Cancer Prevention Research, vol.6, issue.10, pp.1140-1149, 2013.
DOI : 10.1158/1940-6207.CAPR-13-0216

G. Huang, D. Shen, C. Cai, Q. Zhang, H. Ren et al., ??-escin reverses multidrug resistance through inhibition of the GSK3??/??-catenin pathway in cholangiocarcinoma, World Journal of Gastroenterology, vol.21, issue.4, pp.1148-1157, 2015.
DOI : 10.3748/wjg.v21.i4.1148

D. Shen, J. Kang, W. Song, W. Zhang, W. Li et al., Apoptosis of Human Cholangiocarcinoma Cell Lines induced by ??-Escin through Mitochondrial Caspase-dependent Pathway, Phytotherapy Research, vol.75, issue.10, pp.1519-1526, 2011.
DOI : 10.1002/ptr.3435

H. Lee, J. Hong, E. Kim, and S. Kim, Escin Suppresses Migration and Invasion Involving the Alteration of CXCL16/CXCR6 Axis in Human Gastric Adenocarcinoma AGS Cells, Nutrition and Cancer, vol.33, issue.6, pp.938-945, 2014.
DOI : 10.1007/s10495-006-4065-y

Y. Wang, X. Xu, P. Zhao, B. Tong, Z. Wei et al., Escin Ia suppresses the metastasis of triple-negative breast cancer by inhibiting epithelial-mesenchymal transition via downregulating LOXL2 expression, Oncotarget, 2016.

M. Montopoli, G. Froldi, M. Comelli, M. Prosdocimi, and L. Caparrotta, Aescin Protection of Human Vascular Endothelial Cells Exposed to Cobalt Chloride Mimicked Hypoxia and Inflammatory Stimuli, Planta Medica, vol.73, issue.3, pp.285-288, 2007.
DOI : 10.1055/s-2007-967118

A. Ishii, T. Kimura, H. Sadahiro, H. Kawano, K. Takubo et al., Histological Characterization of the Tumorigenic ???Peri-Necrotic Niche??? Harboring Quiescent Stem-Like Tumor Cells in Glioblastoma, PLOS ONE, vol.11, issue.1, p.147366, 2016.
DOI : 10.1371/journal.pone.0147366.s005

M. Zeniou, M. Feve, S. Mameri, J. Dong, C. Salome et al., Chemical Library Screening and Structure-Function Relationship Studies Identify Bisacodyl as a Potent and Selective Cytotoxic Agent Towards Quiescent Human Glioblastoma Tumor Stem-Like Cells, PLOS ONE, vol.138, issue.4, p.134793, 2015.
DOI : 10.1371/journal.pone.0134793.s015

URL : https://hal.archives-ouvertes.fr/hal-01223842

E. Maeno, T. Tsubata, and Y. Okada, Apoptotic Volume Decrease (AVD) Is Independent of Mitochondrial Dysfunction and Initiator Caspase Activation, Cells, vol.1, issue.4, pp.1156-1167, 2012.
DOI : 10.3390/cells1041156

C. Bortner and J. Cidlowski, Caspase Independent/Dependent Regulation of K+, Cell Shrinkage, and Mitochondrial Membrane Potential during Lymphocyte Apoptosis, Journal of Biological Chemistry, vol.274, issue.31, pp.21953-21962, 1999.
DOI : 10.1074/jbc.274.31.21953

C. Vu, C. Bortner, and J. Cidlowski, Differential Involvement of Initiator Caspases in Apoptotic Volume Decrease and Potassium Efflux during Fas- and UV-induced Cell Death, Journal of Biological Chemistry, vol.276, issue.40, pp.37602-37611, 2001.
DOI : 10.1074/jbc.M104810200

I. Ma and A. Allan, The Role of Human Aldehyde Dehydrogenase in Normal and Cancer Stem Cells, Stem Cell Reviews and Reports, vol.36, issue.144, pp.292-306, 2011.
DOI : 10.1007/s12015-010-9208-4

A. Schafer, J. Teufel, F. Ringel, M. Bettstetter, I. Hoepner et al., Aldehyde dehydrogenase 1A1--a new mediator of resistance to temozolomide in glioblastoma, Neuro-Oncology, vol.14, issue.12, pp.1452-1464, 2012.
DOI : 10.1093/neuonc/nos270

P. Mao, K. Joshi, J. Li, S. Kim, P. Li et al., Mesenchymal glioma stem cells are maintained by activated glycolytic metabolism involving aldehyde dehydrogenase 1A3, Proceedings of the National Academy of Sciences, vol.110, issue.21, pp.8644-8649, 2013.
DOI : 10.1073/pnas.1221478110

H. Sun, S. Du, G. Liao, X. Xie, C. Ren et al., Do Glioma Patients Derive Any Therapeutic Benefit From Taking a Higher Cumulative Dose of Temozolomide Regimens?, Medicine, vol.94, issue.20, p.827, 2015.
DOI : 10.1097/MD.0000000000000827

N. Weiss, C. Deboux, N. Chaverot, F. Miller, A. Baron-van-evercooren et al., IL8 and CXCL13 are potent chemokines for the recruitment of human neural precursor cells across brain endothelial cells, Journal of Neuroimmunology, vol.223, issue.1-2, pp.131-134, 2010.
DOI : 10.1016/j.jneuroim.2010.03.009

R. Mccloy, S. Rogers, C. Caldon, T. Lorca, A. Castro et al., phase overrides the SAC and decouples mitotic events, Cell Cycle, vol.109, issue.9, pp.1400-1412, 2014.
DOI : 10.1371/journal.pone.0045726