T. Rice, D. H. Lachance, A. M. Molinaro, J. E. Eckel-passow, K. M. Walsh et al., Understanding inherited genetic risk of adult glioma-a review, Neuro-oncology practice, vol.3, issue.1, pp.10-16, 2016.

D. A. Abrams, J. A. Hanson, J. M. Brown, F. P. Hsu, J. B. Delashaw et al., Timing of surgery and bevacizumab therapy in neurosurgical patients with recurrent high grade glioma, Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, vol.22, issue.1, pp.35-44, 2015.

T. A. Wilson, M. A. Karajannis, and D. H. Harter, Glioblastoma multiforme: State of the art and future therapeutics, Surgical neurology international, vol.5, p.64, 2014.

H. Ohgaki and P. Kleihues, Clinical cancer research : an official journal of the American Association for Cancer Research, vol.19, pp.764-72, 2013.

B. Costa, S. Bendinelli, P. Gabelloni, D. Pozzo, E. et al., Human glioblastoma multiforme: p53 reactivation by a novel MDM2 inhibitor, PloS one, vol.8, issue.8, p.72281, 2013.

R. Desai, C. M. Suryadevara, K. A. Batich, S. H. Farber, L. Sanchez-perez et al., Emerging immunotherapies for glioblastoma, Expert opinion on emerging drugs, vol.21, issue.2, pp.133-178, 2016.

T. Ozawa, C. W. Brennan, L. Wang, M. Squatrito, T. Sasayama et al., PDGFRA gene rearrangements are frequent genetic events in PDGFRA-amplified glioblastomas, Genes & development, vol.24, pp.2205-2223, 2010.

F. Han, R. Hu, H. Yang, J. Liu, J. Sui et al., Comprehensive genomic characterization defines human glioblastoma genes and core pathways, OncoTargets and therapy, vol.9, issue.7216, pp.1061-1069, 2008.

M. G. Waugh, Chromosomal Instability and Phosphoinositide Pathway Gene Signatures in Glioblastoma Multiforme, Molecular neurobiology, vol.53, issue.1, pp.621-651, 2016.

D. N. 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.131, issue.6, pp.803-823, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01479018

R. G. Verhaak, K. A. 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, pp.98-110, 2010.

M. M. Inda, R. Bonavia, and J. Seoane, Glioblastoma multiforme: a look inside its heterogeneous nature, Cancers, vol.6, issue.1, pp.226-265, 2014.

M. Ammirati, S. Chotai, H. Newton, T. Lamki, L. Wei et al., Hypofractionated intensity modulated radiotherapy with temozolomide in newly diagnosed glioblastoma multiforme, Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, vol.21, issue.4, pp.633-640, 2014.

L. Cheng, Q. Wu, O. A. Guryanova, Z. Huang, Q. Huang et al., Elevated invasive potential of glioblastoma stem cells. Biochemical and biophysical research communications, vol.406, pp.643-651, 2011.

L. Yang, C. Lin, L. Wang, H. Guo, and X. Wang, Hypoxia and hypoxia-inducible factors in glioblastoma multiforme progression and therapeutic implications, Experimental cell research, vol.318, pp.2417-2443, 2012.

K. Urbanska, J. Sokolowska, M. Szmidt, and P. Sysa, Glioblastoma multiforme -an overview, Contemporary oncology, vol.18, pp.307-319, 2014.

T. Mammoto, A. Jiang, E. Jiang, D. Panigrahy, M. W. Kieran et al., Role of collagen matrix in tumor angiogenesis and glioblastoma multiforme progression. The American journal of pathology, vol.183, pp.1293-305, 2013.

A. Sottoriva, I. Spiteri, S. G. Piccirillo, A. Touloumis, V. P. Collins et al., Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics, Proceedings of the National Academy of Sciences of the United States of America, vol.110, issue.10, pp.4009-4023, 2013.

P. D. Robbins and A. E. Morelli, Regulation of immune responses by extracellular vesicles, Nature reviews Immunology, vol.14, issue.3, pp.195-208, 2014.

P. D. Robbins, A. Dorronsoro, and C. N. Booker, Regulation of chronic inflammatory and immune processes by extracellular vesicles, The Journal of clinical investigation, vol.126, issue.4, pp.1173-80, 2016.

C. Grange and G. Camussi, Immunosuppressive role of extracellular vesicles: HLA-G, an important player. Annals of translational medicine, vol.5, p.223, 2017.

D. Todorova, S. Simoncini, R. Lacroix, F. Sabatier, and F. Dignat-george, Extracellular Vesicles in Angiogenesis, Circulation research, vol.120, issue.10, pp.1658-73, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01777799

V. Zappulli, K. P. Friis, Z. Fitzpatrick, C. A. Maguire, and X. O. Breakefield, Extracellular vesicles and intercellular communication within the nervous system, The Journal of clinical investigation, vol.126, issue.4, pp.1198-207, 2016.

M. Tkach and C. Thery, Communication by Extracellular Vesicles: Where We Are and Where We Need to Go, Cell, vol.164, issue.6, pp.1226-1258, 2016.

N. O. Yamada, Extracellular vesicles: emerging mediators of intercellular communication and tumor angiogenesis, vol.5, p.59, 2017.

G. Raposo and W. Stoorvogel, Extracellular vesicles: exosomes, microvesicles, and friends, The Journal of cell biology, vol.200, issue.4, pp.373-83, 2013.

L. A. Ban, N. A. Shackel, and S. V. Mclennan, Extracellular Vesicles: A New Frontier in Biomarker Discovery for Non-Alcoholic Fatty Liver Disease, International journal of molecular sciences, vol.17, issue.3, p.376, 2016.

M. Kalamvoki and T. Deschamps, Extracellular vesicles during Herpes Simplex Virus type 1 infection: an inquire, Virology journal, vol.13, p.63, 2016.

R. Xu, D. W. Greening, H. J. Zhu, N. Takahashi, and R. J. Simpson, Extracellular vesicle isolation and characterization: toward clinical application, The Journal of clinical investigation, vol.126, issue.4, pp.1152-62, 2016.

B. T. Pan and R. M. Johnstone, Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: selective externalization of the receptor, Cell, vol.33, issue.3, pp.967-78, 1983.

A. V. Vlassov, S. Magdaleno, R. Setterquist, and R. Conrad, Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials, Biochimica et biophysica acta, vol.1820, issue.7, pp.940-948, 2012.

E. R. Abels, X. O. Breakefield, M. Yanez-mo, P. R. Siljander, Z. Andreu et al., Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cellular and molecular neurobiology, Journal of extracellular vesicles, vol.36, issue.3, p.27066, 2015.

K. Miyado, K. Yoshida, K. Yamagata, K. Sakakibara, M. Okabe et al., The fusing ability of sperm is bestowed by CD9-containing vesicles released from eggs in mice, Proceedings of the National Academy of Sciences of the United States of America, vol.105, issue.35, pp.12921-12927, 2008.

L. Corrigan, S. Redhai, A. Leiblich, S. Fan, S. M. Perera et al., BMP-regulated exosomes from Drosophila male reproductive glands reprogram female behavior, The Journal of cell biology, vol.206, issue.5, pp.671-88, 2014.

C. Fruhbeis, D. Frohlich, W. P. Kuo, J. Amphornrat, S. Thilemann et al.,

, Neurotransmitter-triggered transfer of exosomes mediates oligodendrocyte-neuron communication, PLoS biology, vol.11, issue.7, p.1001604, 2013.

D. Frohlich, W. P. Kuo, C. Fruhbeis, J. J. Sun, C. M. Zehendner et al., Multifaceted effects of oligodendroglial exosomes on neurons: impact on neuronal firing rate, signal transduction and gene regulation, Philosophical transactions of the Royal Society of London Series B, Biological sciences, p.369, 1652.

B. D. Grant and J. G. Donaldson, Pathways and mechanisms of endocytic recycling, Nature reviews Molecular cell biology, vol.10, issue.9, pp.597-608, 2009.

D. B. Iaea and F. R. Maxfield, Membrane order in the plasma membrane and endocytic recycling compartment. PloS one, vol.12, p.188041, 2017.

T. Wollert and J. H. Hurley, Molecular mechanism of multivesicular body biogenesis by ESCRT complexes, Nature, vol.464, issue.7290, pp.864-873, 2010.

M. Colombo, C. Moita, G. Van-niel, J. Kowal, J. Vigneron et al., Analysis of ESCRT functions in exosome biogenesis, composition and secretion highlights the heterogeneity of extracellular vesicles, Journal of cell science, vol.126, pp.5553-65, 2013.

M. F. Baietti, Z. Zhang, E. Mortier, A. Melchior, G. Degeest et al., Syndecan-syntenin-ALIX regulates the biogenesis of exosomes, Nature cell biology, vol.14, issue.7, pp.677-85, 2012.

S. Stuffers, S. Wegner, C. Stenmark, H. Brech, and A. , Multivesicular endosome biogenesis in the absence of ESCRTs, Traffic, vol.10, issue.7, pp.925-962, 2009.

K. Laulagnier, D. Grand, A. Dujardin, S. Hamdi, H. Vincent-schneider et al., PLD2 is enriched on exosomes and its activity is correlated to the release of exosomes, FEBS letters, vol.572, issue.1-3, pp.11-15, 2004.

H. Stenmark, Rab GTPases as coordinators of vesicle traffic, Nature reviews Molecular cell biology, vol.10, issue.8, pp.513-538, 2009.

M. Ostrowski, N. B. Carmo, S. Krumeich, I. Fanget, G. Raposo et al., Rab27a and Rab27b control different steps of the exosome secretion pathway, Nature cell biology, vol.12, issue.1, pp.1-13, 2010.

R. Alonso, C. Mazzeo, I. Merida, and M. Izquierdo, A new role of diacylglycerol kinase alpha on the secretion of lethal exosomes bearing Fas ligand during activation-induced cell death of T lymphocytes, Biochimie, vol.89, issue.2, pp.213-234, 2007.

C. M. Fader, D. G. Sanchez, M. B. Mestre, and M. I. Colombo, TI-VAMP/VAMP7 and VAMP3/cellubrevin: two v-SNARE proteins involved in specific steps of the autophagy/multivesicular body pathways, Biochimica et biophysica acta, vol.1793, issue.12, pp.1901-1917, 2009.

N. Puri and P. A. Roche, Mast cells possess distinct secretory granule subsets whose exocytosis is regulated by different SNARE isoforms, Proceedings of the National Academy of Sciences of the United States of America, vol.105, issue.7, pp.2580-2585, 2008.

N. Tiwari, C. C. Wang, C. Brochetta, G. Ke, F. Vita et al., VAMP-8 segregates mast cellpreformed mediator exocytosis from cytokine trafficking pathways, Blood, vol.111, issue.7, pp.3665-74, 2008.

L. A. Mulcahy, R. C. Pink, and D. R. Carter, Routes and mechanisms of extracellular vesicle uptake, Journal of extracellular vesicles, vol.3, 2014.

K. J. Svensson, H. C. Christianson, A. Wittrup, E. Bourseau-guilmain, E. Lindqvist et al., Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid Raft-mediated endocytosis negatively regulated by caveolin-1, The Journal of biological chemistry, vol.288, issue.24, pp.17713-17737, 2013.

N. A. Atai, L. Balaj, H. Van-veen, X. O. Breakefield, P. A. Jarzyna et al., Heparin blocks transfer of extracellular vesicles between donor and recipient cells, Journal of neuro-oncology, vol.115, issue.3, pp.343-51, 2013.

M. P. Plebanek, R. K. Mutharasan, O. Volpert, A. Matov, J. C. Gatlin et al., Nanoparticle targeting and cholesterol flux through scavenger receptor type B-1 inhibits cellular exosome uptake, Scientific reports, vol.5, p.15724, 2015.

I. Parolini, C. Federici, C. Raggi, L. Lugini, S. Palleschi et al., Microenvironmental pH is a key factor for exosome traffic in tumor cells, The Journal of biological chemistry, vol.284, issue.49, pp.34211-34233, 2009.

I. Nakase and S. Futaki, Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes, Scientific reports, vol.5, p.10112, 2015.

M. Hr, E. Bayraktar, G. Kh, M. F. Abd-ellah, P. Amero et al., Exosomes: From Garbage Bins to Promising Therapeutic Targets. International journal of molecular sciences, vol.18, 2017.

L. Cicero, A. Stahl, P. D. Raposo, and G. , Extracellular vesicles shuffling intercellular messages: for good or for bad. Current opinion in cell biology, vol.35, pp.69-77, 2015.

Q. Feng, C. Zhang, D. Lum, J. E. Druso, B. Blank et al., A class of extracellular vesicles from breast cancer cells activates VEGF receptors and tumour angiogenesis, Nature communications, vol.8, p.14450, 2017.

L. Treps, S. Edmond, E. Harford-wright, E. M. Galan-moya, A. Schmitt et al., Extracellular vesicle-transported Semaphorin3A promotes vascular permeability in glioblastoma, Oncogene, vol.35, issue.20, pp.2615-2638, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01406334

L. R. Languino, A. Singh, M. Prisco, G. J. Inman, A. Luginbuhl et al., Exosome-mediated transfer from the tumor microenvironment increases TGFbeta signaling in squamous cell carcinoma, American journal of translational research, vol.8, issue.5, pp.2432-2439, 2016.

S. V. Manda, Y. Kataria, B. R. Tatireddy, B. Ramakrishnan, B. G. Ratnam et al., Exosomes as a biomarker platform for detecting epidermal growth factor receptor-positive high-grade gliomas, Journal of neurosurgery, vol.128, issue.4, pp.1091-101, 2018.

C. Subra, D. Grand, K. Laulagnier, A. Stella, G. Lambeau et al., Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins, Journal of lipid research, vol.51, issue.8, pp.2105-2125, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00497229

T. Skotland, K. Sandvig, and A. Llorente, Lipids in exosomes: Current knowledge and the way forward, Progress in lipid research, vol.66, pp.30-41, 2017.

A. O. Batagov and I. V. Kurochkin, Exosomes secreted by human cells transport largely mRNA fragments that are enriched in the 3'-untranslated regions, Biology direct, vol.8, p.12, 2013.

R. Crescitelli, C. Lasser, T. G. Szabo, A. Kittel, M. Eldh et al., Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes, Journal of extracellular vesicles, vol.2, 2013.

L. Cheng, X. Sun, B. J. Scicluna, B. M. Coleman, and A. F. Hill, Characterization and deep sequencing analysis of exosomal and non-exosomal miRNA in human urine, Kidney international, vol.86, issue.2, pp.433-477, 2014.

X. Huang, T. Yuan, M. Tschannen, Z. Sun, H. Jacob et al., Characterization of human plasma-derived exosomal RNAs by deep sequencing, BMC genomics, vol.14, p.319, 2013.

Y. Ogawa, Y. Taketomi, M. Murakami, M. Tsujimoto, and R. Yanoshita, Small RNA transcriptomes of two types of exosomes in human whole saliva determined by next generation sequencing, Biological & pharmaceutical bulletin, vol.36, issue.1, pp.66-75, 2013.

A. Waldenstrom, N. Genneback, U. Hellman, and G. Ronquist, Cardiomyocyte microvesicles contain DNA/RNA and convey biological messages to target cells, PloS one, vol.7, issue.4, p.34653, 2012.

L. Balaj, R. Lessard, L. Dai, Y. J. Cho, S. L. Pomeroy et al., Tumour microvesicles contain retrotransposon elements and amplified oncogene sequences, Nature communications, vol.2, p.180, 2011.

V. Budnik, C. Ruiz-canada, and F. Wendler, Extracellular vesicles round off communication in the nervous system, Nature reviews Neuroscience, vol.17, issue.3, pp.160-72, 2016.

Z. M. Liu, Y. B. Wang, and X. H. Yuan, Exosomes from murine-derived GL26 cells promote glioblastoma tumor growth by reducing number and function of CD8+T cells, Asian Pacific journal of cancer prevention : APJCP, vol.14, issue.1, pp.309-323, 2013.

C. Kahlert and R. Kalluri, Exosomes in tumor microenvironment influence cancer progression and metastasis, Journal of molecular medicine, vol.91, issue.4, pp.431-438, 2013.

C. C. Li, S. A. Eaton, P. E. Young, M. Lee, R. Shuttleworth et al., Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells, RNA biology, vol.10, issue.8, pp.1333-1377, 2013.

D. M. O'rourke, M. P. Nasrallah, A. Desai, J. J. Melenhorst, K. Mansfield et al., A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma, Science translational medicine, vol.9, issue.399, 2017.

G. Schiera, D. Liegro, C. M. Saladino, P. Pitti, R. Savettieri et al., Oligodendroglioma cells synthesize the differentiation-specific linker histone H1 and release it into the extracellular environment through shed vesicles, International journal of oncology, vol.43, issue.6, pp.1771-1777, 2013.

U. Putz, J. Howitt, A. Doan, C. P. Goh, L. H. Low et al., The tumor suppressor PTEN is exported in exosomes and has phosphatase activity in recipient cells, Science signaling, vol.5, issue.243, p.70, 2012.

W. T. Arscott, A. T. Tandle, S. Zhao, J. E. Shabason, I. K. Gordon et al., Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration, Translational oncology, vol.6, issue.6, pp.638-686, 2013.

G. L. Semenza, Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy, Trends in pharmacological sciences, vol.33, issue.4, pp.207-221, 2012.

P. Kucharzewska, H. C. Christianson, J. E. Welch, K. J. Svensson, E. Fredlund et al., Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development, Proceedings of the National Academy of Sciences, 2013.

G. Zhang, Y. Zhang, S. Cheng, Z. Wu, F. Liu et al., CD133 positive U87 glioblastoma cellsderived exosomal microRNAs in hypoxia-versus normoxia-microenviroment, Journal of neurooncology, vol.135, issue.1, pp.37-46, 2017.

H. Paz, N. Pathak, and J. Yang, Invading one step at a time: the role of invadopodia in tumor metastasis, Oncogene, vol.33, issue.33, pp.4193-202, 2014.

H. S. Leong, A. E. Robertson, K. Stoletov, S. J. Leith, C. A. Chin et al., Invadopodia are required for cancer cell extravasation and are a therapeutic target for metastasis, Cell reports, vol.8, issue.5, pp.1558-70, 2014.

S. S. Stylli, I. St, A. H. Kaye, and P. Lock, Prognostic significance of Tks5 expression in gliomas, Journal of clinical neuroscience : official journal of the Neurosurgical Society of Australasia, vol.19, issue.3, pp.436-478, 2012.

D. Hoshino, K. C. Kirkbride, K. Costello, E. S. Clark, S. Sinha et al., Exosome secretion is enhanced by invadopodia and drives invasive behavior, Cell reports, vol.5, issue.5, pp.1159-68, 2013.

D. M. Mallawaaratchy, S. Hallal, B. Russell, L. Ly, S. Ebrahimkhani et al., Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease, Journal of neuro-oncology, vol.131, issue.2, pp.233-277, 2017.

F. Ricklefs, M. Mineo, A. K. Rooj, I. Nakano, A. Charest et al., Extracellular Vesicles from High-Grade Glioma Exchange Diverse Pro-oncogenic Signals That Maintain Intratumoral Heterogeneity, Cancer research, vol.76, issue.10, pp.2876-81, 2016.

A. Bronisz, Y. Wang, M. O. Nowicki, P. Peruzzi, K. Ansari et al., Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1, Cancer research, vol.74, issue.3, pp.738-50, 2014.

X. Sun, X. Ma, J. Wang, Y. Zhao, Y. Wang et al., Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal, Oncotarget, vol.8, issue.22, pp.36137-36185, 2017.

J. De-vrij, S. L. Maas, K. M. Kwappenberg, R. Schnoor, A. Kleijn et al., Glioblastomaderived extracellular vesicles modify the phenotype of monocytic cells, International journal of cancer, vol.137, issue.7, pp.1630-1672, 2015.

K. E. Van-der-vos, E. R. Abels, X. Zhang, C. Lai, E. Carrizosa et al., Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain, Neuro-oncology, vol.18, issue.1, pp.58-69, 2016.

R. Sosnowski, M. Zagrodzka, and T. Borkowski, The limitations of multiparametric magnetic resonance imaging also must be borne in mind, Central European journal of urology, vol.69, issue.1, pp.22-25, 2016.

C. Peca, R. Pacelli, A. Elefante, D. Caro, M. L. Vergara et al., Early clinical and neuroradiological worsening after radiotherapy and concomitant temozolomide in patients with glioblastoma: tumour progression or radionecrosis?, Clinical neurology and neurosurgery, vol.111, issue.4, pp.331-335, 2009.

M. R. Neagu, R. Y. Huang, D. A. Reardon, and P. Y. Wen, How treatment monitoring is influencing treatment decisions in glioblastomas. Current treatment options in neurology, vol.17, p.343, 2015.

M. Ilie and P. Hofman, Pros: Can tissue biopsy be replaced by liquid biopsy? Translational lung cancer research, vol.5, pp.420-423, 2016.

A. P. Patel, I. Tirosh, J. J. Trombetta, A. K. Shalek, S. M. Gillespie et al., Single-cell RNAseq highlights intratumoral heterogeneity in primary glioblastoma, Science, vol.344, issue.6190, pp.1396-401, 2014.

S. Yip, J. Miao, D. P. Cahill, A. J. Iafrate, K. Aldape et al., MSH6 mutations arise in glioblastomas during temozolomide therapy and mediate temozolomide resistance. Clinical cancer research : an official journal of the American Association for Cancer Research, vol.15, pp.4622-4631, 2009.

H. Valadi, K. Ekstrom, A. Bossios, M. Sjostrand, J. J. Lee et al., Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nature cell biology, vol.9, issue.6, pp.654-663, 2007.

M. Morishita, Y. Takahashi, M. Nishikawa, K. Sano, K. Kato et al., Quantitative analysis of tissue distribution of the B16BL6-derived exosomes using a streptavidin-lactadherin fusion protein and iodine-125-labeled biotin derivative after intravenous injection in mice, Journal of pharmaceutical sciences, vol.104, issue.2, pp.705-718, 2015.

J. Skog, T. Wurdinger, S. Van-rijn, D. H. Meijer, L. Gainche et al., Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers, Nature cell biology, vol.10, issue.12, pp.1470-1476, 2008.

M. Noerholm, L. Balaj, T. Limperg, A. Salehi, L. D. Zhu et al., RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls, BMC cancer, vol.12, p.22, 2012.

F. Lan, Q. Qing, Q. Pan, M. Hu, H. Yu et al., Serum exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma, Cellular oncology (Dordrecht), vol.41, issue.1, pp.25-33, 2018.

W. W. Chen, L. Balaj, L. M. Liau, M. L. Samuels, S. K. Kotsopoulos et al., BEAMing and Droplet Digital PCR Analysis of Mutant IDH1 mRNA in Glioma Patient Serum and Cerebrospinal Fluid Extracellular Vesicles, Molecular therapy Nucleic acids, vol.2, p.109, 2013.

J. C. Akers, W. Hua, H. Li, V. Ramakrishnan, Z. Yang et al., A cerebrospinal fluid microRNA signature as biomarker for glioblastoma, Oncotarget, vol.8, issue.40, pp.68769-79, 2017.

J. Figueroa, L. M. Phillips, T. Shahar, A. Hossain, J. Gumin et al., Exosomes from GliomaAssociated Mesenchymal Stem Cells Increase the Tumorigenicity of Glioma Stem-like Cells via Transfer of miR-1587. Cancer research, vol.77, pp.5808-5827, 2017.

H. Shao, J. Chung, K. Lee, L. Balaj, C. Min et al., Chip-based analysis of exosomal mRNA mediating drug resistance in glioblastoma, Nature communications, vol.6, p.6999, 2015.

V. Ramakrishnan, D. Kushwaha, D. C. Koay, H. Reddy, Y. Mao et al., Post-transcriptional regulation of O(6)-methylguanine-DNA methyltransferase MGMT in glioblastomas. Cancer biomarkers : section A of Disease markers, vol.10, pp.185-93, 2011.

D. Kushwaha, V. Ramakrishnan, K. Ng, T. Steed, T. Nguyen et al., A genome-wide miRNA screen revealed miR-603 as a MGMT-regulating miRNA in glioblastomas, Oncotarget, vol.5, issue.12, pp.4026-4065, 2014.

J. K. Yang, J. P. Yang, J. Tong, S. Y. Jing, B. Fan et al., Exosomal miR-221 targets DNM3 to induce tumor progression and temozolomide resistance in glioma, Journal of neuro-oncology, vol.131, issue.2, pp.255-65, 2017.

S. Pinet, B. Bessette, N. Vedrenne, A. Lacroix, L. Richard et al., TrkB-containing exosomes promote the transfer of glioblastoma aggressiveness to YKL-40-inactivated glioblastoma cells, Oncotarget, vol.7, issue.31, pp.50349-64, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01819184

R. Stupp, W. P. Mason, M. J. Van-den-bent, M. Weller, B. Fisher et al., Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. The New England journal of medicine, vol.352, pp.987-96, 2005.

G. Andre-gregoire, N. Bidere, and J. Gavard, Temozolomide affects Extracellular Vesicles Released by Glioblastoma Cells, Biochimie, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01715956

A. Zeng, Z. Wei, W. Yan, J. Yin, X. Huang et al., Exosomal transfer of miR-151a enhances chemosensitivity to temozolomide in drug-resistant glioblastoma, Cancer letters, vol.436, pp.10-21, 2018.

D. Garnier, B. Meehan, T. Kislinger, P. Daniel, A. Sinha et al., Divergent evolution of temozolomide resistance in glioblastoma stem cells is reflected in extracellular vesicles and coupled with radiosensitization, Neuro-oncology, vol.20, issue.2, pp.236-284, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01814308

D. Uribe, A. Torres, J. D. Rocha, I. Niechi, C. Oyarzun et al., Multidrug resistance in glioblastoma stem-like cells: Role of the hypoxic microenvironment and adenosine signaling. Molecular aspects of medicine, vol.55, pp.140-51, 2017.

J. S. Redzic, T. H. Ung, and M. W. Graner, Glioblastoma extracellular vesicles: reservoirs of potential biomarkers. Pharmacogenomics and personalized medicine, vol.7, pp.65-77, 2014.

J. M. Figueroa, J. Skog, J. Akers, H. Li, R. Komotar et al., Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients, Neuro-oncology, vol.19, issue.11, pp.1494-502, 2017.

J. C. Akers, V. Ramakrishnan, R. Kim, S. Phillips, V. Kaimal et al., miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients, Journal of neuro-oncology, vol.123, issue.2, pp.205-221, 2015.

V. Vlaeminck-guillem, Extracellular Vesicles in Prostate Cancer Carcinogenesis, Diagnosis, and Management. Frontiers in oncology, vol.8, p.222, 2018.

H. Zheng, Y. Zhan, S. Liu, J. Lu, J. Luo et al., The roles of tumor-derived exosomes in nonsmall cell lung cancer and their clinical implications, Journal of experimental & clinical cancer research : CR, vol.37, issue.1, p.226, 2018.

Y. G. Zhang, M. W. Zhou, L. Bai, R. Y. Han, K. Lv et al., Extracellular vesicles promote esophageal cancer progression by delivering lncZEB1-AS1 between cells. European review for medical and pharmacological sciences, vol.22, pp.2662-70, 2018.

M. X. Liu, J. Liao, M. Xie, Z. K. Gao, X. H. Wang et al., miR-93-5p Transferred by Exosomes Promotes the Proliferation of Esophageal Cancer Cells via Intercellular Communication by Targeting PTEN, Biomedical and environmental sciences : BES, vol.31, issue.3, pp.171-85, 2018.

S. Sharma, F. Zuniga, G. E. Rice, L. C. Perrin, J. D. Hooper et al., Tumor-derived exosomes in ovarian cancer -liquid biopsies for early detection and real-time monitoring of cancer progression, Oncotarget, vol.8, issue.61, pp.104687-703, 2017.

E. A. Armstrong, E. W. Beal, J. Chakedis, A. Z. Paredes, D. Moris et al., Exosomes in Pancreatic Cancer: from Early Detection to Treatment, Journal of gastrointestinal surgery : official journal of the Society for Surgery of the Alimentary Tract, vol.22, issue.4, pp.737-50, 2018.

C. Zhao, F. Gao, S. Weng, and Q. Liu, Cancer biomarkers : section A of Disease markers, vol.20, pp.357-67, 2017.

Y. Yan, G. Fu, and M. L. , Role of exosomes in pancreatic cancer, Oncology letters, vol.15, issue.5, pp.7479-88, 2018.

T. Kitagawa, K. Taniuchi, M. Tsuboi, M. Sakaguchi, T. Kohsaki et al., Circulating pancreatic cancer exosomal RNAs for detection of pancreatic cancer. Molecular oncology, 2018.

B. J. Tauro, D. W. Greening, R. A. Mathias, H. Ji, S. Mathivanan et al., Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes, Methods, vol.56, issue.2, pp.293-304, 2012.

E. Van-der-pol, A. N. Boing, P. Harrison, A. Sturk, and R. Nieuwland, Classification, functions, and clinical relevance of extracellular vesicles, Pharmacological reviews, vol.64, issue.3, pp.676-705, 2012.

H. G. Zhang and W. E. Grizzle, Exosomes: a novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. The American journal of pathology, vol.184, pp.28-41, 2014.

J. Webber and A. Clayton, How pure are your vesicles, Journal of extracellular vesicles, vol.2, 2013.

O. E. Bryzgunova, M. M. Zaripov, T. E. Skvortsova, E. A. Lekchnov, A. E. Grigor'eva et al., Comparative Study of Extracellular Vesicles from the Urine of Healthy Individuals and Prostate Cancer Patients, PloS one, vol.11, issue.6, p.157566, 2016.

J. A. Saugstad, T. A. Lusardi, K. R. Van-keuren-jensen, J. I. Phillips, B. Lind et al., Analysis of extracellular RNA in cerebrospinal fluid, Journal of extracellular vesicles, vol.6, issue.1, p.1317577, 2017.

M. Li, E. Zeringer, T. Barta, J. Schageman, A. Cheng et al., Analysis of the RNA content of the exosomes derived from blood serum and urine and its potential as biomarkers, Philosophical transactions of the Royal Society of London Series B, Biological sciences, p.369, 1652.

I. Moret, D. Sanchez-izquierdo, M. Iborra, L. Tortosa, A. Navarro-puche et al., Assessing an improved protocol for plasma microRNA extraction, PloS one, vol.8, issue.12, p.82753, 2013.

W. L. Dean, M. J. Lee, T. D. Cummins, D. J. Schultz, and D. W. Powell, Proteomic and functional characterisation of platelet microparticle size classes, Thrombosis and haemostasis, vol.102, issue.4, pp.711-719, 2009.

K. C. Vickers, B. T. Palmisano, B. M. Shoucri, R. D. Shamburek, and A. T. Remaley, MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins, Nature cell biology, vol.13, issue.4, pp.423-456, 2011.

B. W. Sodar, A. Kittel, K. Paloczi, K. V. Vukman, X. Osteikoetxea et al., Low-density lipoprotein mimics blood plasma-derived exosomes and microvesicles during isolation and detection, Sci Rep, vol.6, p.24316, 2016.

N. M. Teplyuk, B. Mollenhauer, G. Gabriely, A. Giese, E. Kim et al., MicroRNAs in cerebrospinal fluid identify glioblastoma and metastatic brain cancers and reflect disease activity, Neuro-oncology, vol.14, issue.6, pp.689-700, 2012.

J. H. Kang, B. Mollenhauer, C. S. Coffey, J. B. Toledo, D. Weintraub et al., CSF biomarkers associated with disease heterogeneity in early Parkinson's disease: the Parkinson's Progression Markers Initiative study, Acta neuropathologica, vol.131, issue.6, pp.935-984, 2016.

C. L. Weston, M. J. Glantz, and J. R. Connor, Detection of cancer cells in the cerebrospinal fluid: current methods and future directions, Fluids and barriers of the CNS, vol.8, issue.1, p.14, 2011.

E. Mora, S. Álvarez-cubela, and E. Oltra, Biobanking of exosomes in the era of precision medicine: are we there yet? International journal of molecular sciences, vol.17, p.13, 2015.

L. A. Madden, R. V. Vince, M. E. Sandstrom, L. Taylor, L. Mcnaughton et al., Microparticleassociated vascular adhesion molecule-1 and tissue factor follow a circadian rhythm in healthy human subjects, Thrombosis and haemostasis, vol.99, issue.5, pp.909-924, 2008.

C. Fruhbeis, S. Helmig, S. Tug, P. Simon, and E. M. Kramer-albers, Physical exercise induces rapid release of small extracellular vesicles into the circulation, Journal of extracellular vesicles, vol.4, p.28239, 2015.

B. Gyorgy, K. Paloczi, A. Kovacs, E. Barabas, G. Beko et al., Improved circulating microparticle analysis in acid-citrate dextrose (ACD) anticoagulant tube, Thrombosis research, vol.133, issue.2, pp.285-92, 2014.

F. M. Lang, A. Hossain, J. Gumin, E. N. Momin, Y. Shimizu et al., Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas, Neurooncology, vol.20, issue.3, pp.380-90, 2018.

M. S. Pavlyukov, H. Yu, S. Bastola, M. Minata, V. O. Shender et al., Apoptotic Cell-Derived Extracellular Vesicles Promote Malignancy of Glioblastoma Via Intercellular Transfer of Splicing Factors, Cancer cell, vol.34, issue.1, pp.119-154, 2018.

R. A. Kore and E. C. Abraham, Inflammatory cytokines, interleukin-1 beta and tumor necrosis factoralpha, upregulated in glioblastoma multiforme, raise the levels of CRYAB in exosomes secreted by U373 glioma cells. Biochemical and biophysical research communications, vol.453, pp.326-357, 2014.

L. Manterola, E. Guruceaga, G. Perez-larraya, J. Gonzalez-huarriz, M. Jauregui et al., A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool, Neuro-oncology, vol.16, issue.4, pp.520-527, 2014.

Q. Cai, A. Zhu, and L. Gong, Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1, Bulletin du cancer, vol.105, issue.7-8, pp.643-51, 2018.

K. Huang, C. Fang, K. Yi, X. Liu, H. Qi et al., The role of PTRF/Cavin1 as a biomarker in both glioma and serum exosomes, Theranostics, vol.8, issue.6, pp.1540-57, 2018.

R. Shi, P. Y. Wang, X. Y. Li, J. X. Chen, Y. Li et al., Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients, Oncotarget, vol.6, issue.29, pp.26971-81, 2015.

J. Zheng, X. Liu, Y. Xue, W. Gong, J. Ma et al., TTBK2 circular RNA promotes glioma malignancy by regulating miR-217/HNF1?/Derlin-1 pathway, Journal of hematology & oncology, vol.10, issue.1, p.52, 2017.