S. Jawhari, B. Bessette, S. Hombourger, K. Durand, A. Lacroix et al., Autophagy and TrkC/NT-3 signaling joined forces boost the hypoxic glioblastoma cell survival, Carcinogenesis, vol.38, pp.592-603, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01873444

J. Xiong, L. Zhou, M. Yang, Y. Lim, Y. Zhu et al., ProBDNF and its receptors are upregulated in glioma and inhibit the growth of glioma cells in vitro, Neuro-Oncol, vol.15, pp.990-1007, 2013.

S. Lawn, N. Krishna, A. Pisklakova, X. Qu, D. A. Fenstermacher et al., Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells, J Biol Chem, vol.290, pp.3814-3824, 2015.

S. Giraud, E. Loum, B. Bessette, M. Mathonnet, and F. Lalloué, P75 neurotrophin receptor is sequestered in the Golgi apparatus of the U-87 MG human glioblastoma cell line, Int J Oncol, vol.38, pp.391-399, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00597714

K. Batista, I. F. Vega, S. A. De-eulate-beramendi, J. Morales, A. Kurbanov et al., Prognostic significance of the markers IDH1 and YKL40 related to the subventricular zone, Folia Neuropathol, vol.1, pp.52-59, 2015.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.96, 2018.

, Licence CC BY

H. R. Haynes, P. White, K. M. Hares, J. Redondo, K. C. Kemp et al., The transcription factor PPAR? is overexpressed and is associated with a favourable prognosis in IDH-wildtype primary glioblastoma, Histopathology, vol.70, pp.1030-1043, 2017.

R. Verhaak, K. A. Hoadley, E. Purdom, V. Wang, Y. Qi et al., An integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR and NF1, Cancer Cell, vol.17, p.98, 2010.

C. Jungk, A. Mock, J. Exner, C. Geisenberger, R. Warta et al., Spatial transcriptome analysis reveals Notch pathway-associated prognostic markers in IDH1 wild-type glioblastoma involving the subventricular zone, BMC Med, vol.14, p.170, 2016.

H. Wang, K. Tang, T. Liang, W. Zhang, J. Li et al., The comparison of clinical and biological characteristics between IDH1 and IDH2 mutations in gliomas, J Exp Clin Cancer Res CR, vol.35, p.86, 2016.

L. Mesturoux, K. Durand, I. Pommepuy, S. Robert, F. Caire et al., Molecular Analysis of Tumor Cell Components in Pilocytic Astrocytomas, Gangliogliomas, and Oligodendrogliomas, Appl Immunohistochem Mol Morphol AIMM, vol.24, pp.496-500, 2016.

J. Jeuken, S. Cornelissen, S. Boots-sprenger, S. Gijsen, and P. Wesseling, Multiplex ligationdependent probe amplification: a diagnostic tool for simultaneous identification of different genetic markers in glial tumors, J Mol Diagn JMD, vol.8, pp.433-443, 2006.

M. Ermonval, D. Petit, L. Duc, A. Kellermann, O. Gallet et al., Glycosylation-related genes are variably expressed depending on the differentiation state of a bioaminergic neuronal cell line: implication for the cellular prion protein, Glycoconj J, vol.26, pp.477-493, 2009.
URL : https://hal.archives-ouvertes.fr/hal-01211889

T. D. Schmittgen and K. J. Livak, Analyzing real-time PCR data by the comparative CT method, Nat Protoc, vol.3, p.1101, 2008.

F. Husson, J. Josse, and S. Le, FactoMineR: An R Package for Multivariate Analysis, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00359835

Z. Gu, R. Eils, and M. Schlesner, Complex heatmaps reveal patterns and correlations in multidimensional genomic data, Bioinforma Oxf Engl, vol.32, pp.2847-2849, 2016.

D. N. Louis, A. Perry, P. Burger, D. W. Ellison, G. Reifenberger et al., International Society Of Neuropathology--Haarlem consensus guidelines for nervous system tumor classification and grading, Brain Pathol Zurich Switz, vol.24, pp.429-435, 2014.

K. Gao, X. Chen, J. Zhang, Y. Wang, W. Yan et al., A pseudogene-signature in glioma predicts survival, J Exp Clin Cancer Res CR, vol.34, 2015.

S. Li, J. Shi, H. Gao, Y. Yuan, Q. Chen et al., Identification of a gene signature associated with radiotherapy and prognosis in gliomas, Oncotarget, vol.5, pp.88974-88987, 2014.

C. L. Nutt, D. R. Mani, R. A. Betensky, P. Tamayo, J. G. Cairncross et al., Gene expressionbased classification of malignant gliomas correlates better with survival than histological classification, Cancer Res, vol.63, pp.1602-1607, 2003.

M. Shirahata, K. Iwao-koizumi, S. Saito, N. Ueno, M. Oda et al., Gene expression-based molecular diagnostic system for malignant gliomas is superior to histological diagnosis, Clin Cancer Res Off J Am Assoc Cancer Res, vol.13, pp.7341-7356, 2007.

L. A. Gravendeel, M. C. Kouwenhoven, O. Gevaert, J. J. De-rooi, A. P. Stubbs et al., Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology, Cancer Res, vol.69, pp.9065-9072, 2009.

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

M. Ceccarelli, F. P. Barthel, T. M. Malta, T. S. Sabedot, S. R. Salama et al., Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma, Cell, vol.164, pp.550-563, 2016.

M. Palani, R. Arunkumar, and A. J. Vanisree, Methylation and Expression Patterns of Tropomyosin-Related Kinase Genes in Different Grades of Glioma, NeuroMolecular Med, vol.16, pp.529-539, 2014.

S. Rutkowski, A. Von-bueren, V. Hoff, K. Hartmann, W. Shalaby et al., Prognostic relevance of clinical and biological risk factors in childhood medulloblastoma: results of patients treated in the prospective multicenter trial HIT'91, Clin Cancer Res Off J Am Assoc Cancer Res, vol.13, pp.2651-2657, 2007.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.97, 2018.

, Licence CC BY

M. Assimakopoulou, M. Kondyli, G. Gatzounis, T. Maraziotis, and J. Varakis, Neurotrophin receptors expression and JNK pathway activation in human astrocytomas, BMC Cancer, vol.7, p.202, 2007.

J. Dewitt, V. Ochoa, J. Urschitz, M. Elston, S. Moisyadi et al., Constitutively active TrkB confers an aggressive transformed phenotype to a neural crest derived cell line, Oncogene, vol.33, pp.977-985, 2014.

S. Wadhwa, T. C. Nag, A. Jindal, R. Kushwaha, A. K. Mahapatra et al., Expression of the neurotrophin receptors Trk A and Trk B in adult human astrocytoma and glioblastoma, J Biosci, vol.28, pp.181-188, 2003.

G. Steponaitis, D. Skiriut?, A. Kazlauskas, I. Golubickait?, R. Stakaitis et al.,

, High CHI3L1 expression is associated with glioma patient survival, Diagn Pathol, vol.11, 2016.

G. Qin, X. Li, Z. Chen, G. Liao, Y. Su et al., Prognostic Value of YKL-40 in Patients with Glioblastoma: a Systematic Review and Meta-analysis, Mol Neurobiol, vol.54, pp.3264-3270, 2017.

M. Thuy, J. Kam, G. Lee, P. L. Tao, D. Q. Ling et al., A novel literature-based approach to identify genetic and molecular predictors of survival in glioblastoma multiforme: Analysis of 14,678 patients using systematic review and meta-analytical tools, J Clin Neurosci, vol.22, pp.785-799, 2015.

J. E. Eckel-passow, D. H. Lachance, A. M. Molinaro, K. M. Walsh, P. A. Decker et al.,

, Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors, N Engl J Med, vol.372, pp.2499-2508, 2015.

T. Kessler, F. Sahm, A. Sadik, D. Stichel, A. Hertenstein et al., Molecular differences in IDH wildtype glioblastoma according to MGMT promoter methylation, Neuro-Oncol, vol.20, pp.367-379, 2018.

S. Turcan, D. Rohle, A. Goenka, L. A. Walsh, F. Fang et al., IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype, Nature, vol.483, pp.479-483, 2012.

H. Noushmehr, D. J. Weisenberger, K. Diefes, H. S. Phillips, K. Pujara et al., Identification of a CpG Island Methylator Phenotype that Defines a Distinct Subgroup of Glioma, Cancer Cell, vol.17, pp.510-522, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01201573

D. H. Heiland, R. Ferrarese, C. R. Dai, F. Masilamani, A. P. Kling et al., Jun-N-terminal phosphorylation regulates DNMT1 expression and genome wide methylation in gliomas, Oncotarget, vol.8, pp.6940-6954, 2016.

M. Faibish, R. Francescone, B. Bentley, W. Yan, and R. Shao, A YKL-40-neutralizing antibody blocks tumor angiogenesis and progression: a potential therapeutic agent in cancers, Mol Cancer Ther, vol.10, pp.742-751, 2011.

Y. Akiyama, T. Ashizawa, M. Komiyama, H. Miyata, C. Oshita et al., YKL-40 downregulation is a key factor to overcome temozolomide resistance in a glioblastoma cell line, Oncol Rep, vol.32, pp.159-175, 2014.

S. Wislet, G. Vandervelden, B. Rogister, S. Pramanik, Y. A. Sulistio et al., From Neural Crest Development to Cancer and Vice Versa: How p75NTR and (Pro)neurotrophins Could Act on Cell Migration and Invasion? Front Mol Neurosci, Mol Neurobiol, vol.11, issue.9, pp.7401-59, 2017.

A. L. Johnston, X. Lun, J. J. Rahn, A. Liacini, L. Wang et al., The p75 neurotrophin receptor is a central regulator of glioma invasion, e212. 16. Alshehri MM, Robbins SM, Senger DL. The Role of Neurotrophin Signaling in Gliomagenesis: A Focus on the p75 Neurotrophin Receptor (p75NTR/CD271), vol.5, pp.367-404, 2007.

S. Pinet, B. Bessette, N. Vedrenne, A. Lacroix, L. Richard et al., TrkBcontaining exosomes promote the transfer of glioblastoma aggressiveness to YKL-40-Elise DELUCHE-MOURICOUT| Thèse de doctorat | Université de Limoges, vol.21, p.119, 2018.

, Licence CC BY

, Oncotarget, vol.7, issue.31, pp.50349-64, 2016.

S. Giraud, E. Loum, B. Bessette, M. Mathonnet, and F. Lalloué, P75 neurotrophin receptor is sequestered in the Golgi apparatus of the U-87 MG human glioblastoma cell line, Int J Oncol, vol.38, issue.2, pp.391-399, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00597714

J. L. Croucher, R. Iyer, N. Li, V. Molteni, L. J. Gordon et al., TrkB Inhibition by GNF-4256 Slows Growth and Enhances Chemotherapeutic Efficacy in Neuroblastoma Xenografts, Cancer Chemother Pharmacol, vol.75, issue.1, pp.131-172, 2015.

A. E. Evans, K. D. Kisselbach, X. Liu, A. Eggert, N. Ikegaki et al., Effect of CEP-751 (KT-6587) on neuroblastoma xenografts expressing TrkB, Med Pediatr Oncol

F. R. Hirsch, M. Varella-garcia, P. A. Bunn, D. Maria, M. V. Veve et al.,

, Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis, J Clin Oncol Off J Am Soc Clin Oncol, vol.21, issue.20, pp.3798-807, 2003.

K. J. Livak and T. D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods San Diego Calif, vol.25, pp.402-410, 2001.

S. Lawn, N. Krishna, A. Pisklakova, X. Qu, D. A. Fenstermacher et al., Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells, J Biol Chem, vol.290, issue.6, pp.3814-3838, 2015.

T. Wang, S. Luo, C. Lin, P. Chang, and M. Chen, Modulation of p75 neurotrophin receptor under hypoxic conditions induces migration and invasion of C6 glioma cells, Clin Exp Metastasis, vol.32, issue.1, pp.73-81, 2015.

L. Wang, J. J. Rahn, X. Lun, B. Sun, J. Kelly et al., Gamma-Secretase Represents a Therapeutic Target for the Treatment of Invasive Glioma Mediated by the p75 Neurotrophin Receptor, PLOS Biol, vol.6, issue.11, p.289, 2008.

B. Y. Ahn, R. Saldanha-gama, J. J. Rahn, X. Hao, J. Zhang et al., Glioma invasion mediated by the p75 neurotrophin receptor (p75NTR/CD271) requires regulated interaction with PDLIM1, Oncogene, vol.35, issue.11, p.1411, 2016.

P. A. Forsyth, N. Krishna, S. Lawn, J. G. Valadez, X. Qu et al., p75 neurotrophin receptor cleavage by ?-and ?-secretases is required for neurotrophin-mediated proliferation of brain tumor-initiating cells, J Biol Chem, vol.289, issue.12, pp.8067-8085, 2014.

A. Thomaz, M. Jaeger, M. Buendia, V. Bambini-junior, L. J. Gregianin et al., BDNF/TrkB Signaling as a Potential Novel Target in Pediatric Brain Tumors: Anticancer Activity of Selective TrkB Inhibition in Medulloblastoma Cells, J Mol Neurosci MN, vol.59, issue.3, pp.326-359, 2016.

T. E. Heinen, D. Santos, R. P. Da-rocha, A. , D. Santos et al., Trk inhibition reduces cell proliferation and potentiates the effects of chemotherapeutic agents in Ewing sarcoma, Oncotarget, vol.7, issue.23, pp.34860-80, 2016.

K. V. Pinheiro, C. Alves, M. Buendia, M. S. Gil, A. Thomaz et al., Targeting tyrosine receptor kinase B in gliomas. Neuro-Oncol, vol.19, pp.138-147, 2017.

G. Chen, T. G. Gharib, C. Huang, J. Taylor, D. E. Misek et al., Discordant Protein and mRNA Expression in Lung Adenocarcinomas, Mol Cell Proteomics, vol.1, issue.4, pp.304-317, 2002.

A. Modelska, A. Quattrone, and A. Re, Molecular portraits: the evolution of the concept of transcriptome-based cancer signatures. Brief Bioinform, vol.16, pp.1000-1007, 2015.

B. Zhang, J. Wang, X. Wang, J. Zhu, Q. Liu et al., Proteogenomic characterization of human colon and rectal cancer, Nature, vol.513, issue.7518, pp.382-389, 2014.

. Références-bibliographiques-1,

K. Abou-el-ardat, M. Seifert, K. Becker, S. Eisenreich, M. Lehmann et al., Comprehensive molecular characterization of multifocal glioblastoma proves its monoclonal origin and reveals novel insights into clonal evolution and heterogeneity of glioblastomas, Neuro-Oncol, vol.19, pp.546-557, 2017.

B. Y. Ahn, R. F. Saldanha-gama, J. J. Rahn, X. Hao, J. Zhang et al., Glioma invasion mediated by the p75 neurotrophin receptor (p75NTR/CD271) requires regulated interaction with PDLIM1, p.35, 1411.

J. C. Akers, V. Ramakrishnan, R. Kim, J. Skog, I. Nakano et al., MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development, PloS One, vol.8, p.78115, 2013.

Y. Akiyama, T. Ashizawa, M. Komiyama, H. Miyata, C. Oshita et al., YKL-40 downregulation is a key factor to overcome temozolomide resistance in a glioblastoma cell line, Oncol. Rep, vol.32, pp.159-166, 2014.

K. Al-nedawi, B. Meehan, J. Micallef, V. Lhotak, L. May et al.,

, Intercellular transfer of the oncogenic receptor EGFRvIII by microvesicles derived from tumour cells, Nat. Cell Biol, vol.10, pp.619-624

A. Amatu, A. Sartore-bianchi, and S. Siena, NTRK gene fusions as novel targets of cancer therapy across multiple tumour types, ESMO Open, vol.1, p.23, 2016.

C. L. Andersen, J. L. Jensen, and T. F. Ørntoft, Normalization of Real-Time Quantitative Reverse Transcription-PCR Data: A Model-Based Variance Estimation Approach to Identify Genes Suited for Normalization, Applied to Bladder and Colon Cancer Data Sets, Cancer Res, vol.64, pp.5245-5250, 2004.

E. Ardini, M. Menichincheri, P. Banfi, R. Bosotti, C. De-ponti et al., Entrectinib, a Pan-TRK, ROS1, and ALK Inhibitor with Activity in Multiple Molecularly Defined Cancer Indications, Mol. Cancer Ther, vol.15, pp.628-639, 2016.

M. Assimakopoulou, M. Kondyli, G. Gatzounis, T. Maraziotis, and J. Varakis, Neurotrophin receptors expression and JNK pathway activation in human astrocytomas, BMC Cancer, vol.7, p.202, 2007.

P. Bailey and P. C. Bucy, Oligodendrogliomas of the brain, J. Pathol. Bacteriol, vol.32, pp.735-751, 1929.

P. Bailey and H. Cushing, A classification of the tumours of the glioma group on a histogenetic basis, with a correlated study of prognosis, Br. J. Surg, vol.14, pp.554-555, 1927.

Y. A. Barde, D. Edgar, and H. Thoenen, Purification of a new neurotrophic factor from mammalian brain, EMBO J, vol.1, pp.549-553, 1982.

P. A. Barker, C. Lomen-hoerth, E. M. Gensch, S. O. Meakin, D. J. Glass et al., Tissue-specific alternative splicing generates two isoforms of the trkA receptor, J. Biol. Chem, vol.268, pp.15150-15157, 1993.

L. Bauchet, S. Zouaoui, A. Darlix, N. Menjot-de-champfleur, E. Ferreira et al., Assessment and treatment relevance in elderly glioblastoma patients, Neuro-Oncol, vol.16, pp.1459-1468, 2014.

B. G. Baumert, M. E. Hegi, M. J. Van-den-bent, A. Von-deimling, T. Gorlia et al., Temozolomide chemotherapy versus radiotherapy in high-risk low-grade glioma (EORTC 22033-26033): a randomised, open-label, phase 3 intergroup study, Lancet Oncol, vol.17, pp.1521-1532, 2016.

M. J. Bent, . Van-den, B. Baumert, S. C. Erridge, M. A. Vogelbaum et al., , vol.21, p.142, 2018.

, Licence CC BY

M. Son, A. A. Brandes, P. M. Clement, J. F. Baurain, and W. P. Mason, Interim results from the CATNON trial (EORTC study 26053-22054) of treatment with concurrent and adjuvant temozolomide for 1p/19q non-co-deleted anaplastic glioma: a phase 3, randomised, open-label intergroup study, 18. van den Bent, vol.390, pp.344-350, 2010.

C. Bettegowda, N. Agrawal, Y. Jiao, M. Sausen, L. D. Wood et al., Mutations in CIC and FUBP1 Contribute to Human Oligodendroglioma, vol.333, pp.1453-1455, 2011.

G. Bleau, F. Massicotte, Y. Merlen, and C. Boisvert, Mammalian chitinaselike proteins, EXS, vol.87, pp.211-221, 1999.

K. L. Boeshore, C. N. Luckey, R. E. Zigmond, and T. H. Large, TrkB isoforms with distinct neurotrophin specificities are expressed in predominantly nonoverlapping populations of avian dorsal root ganglion neurons, J. Neurosci. Off. J. Soc. Neurosci, vol.19, pp.4739-4747, 1999.

M. K. Boisen, C. B. Holst, N. Consalvo, O. L. Chinot, and J. S. Johansen, , 2017.

, Plasma YKL-40 as a biomarker for bevacizumab efficacy in patients with newly diagnosed glioblastoma in the phase 3 randomized AVAglio trial, Oncotarget, vol.9, pp.6752-6762

D. Bonneh-barkay, S. J. Bissel, J. Kofler, A. Starkey, G. Wang et al., Astrocyte and macrophage regulation of YKL-40 expression and cellular response in neuroinflammation, Brain Pathol. Zurich Switz, vol.22, pp.530-546, 2012.

M. Z. Braganza, C. M. Kitahara, A. Berrington-de-gonzález, P. D. Inskip, K. J. Johnson et al., Ionizing radiation and the risk of brain and central nervous system tumors: a systematic review, Neuro-Oncol, vol.14, pp.1316-1324, 2012.

C. W. Brennan, R. G. Verhaak, A. Mckenna, B. Campos, H. Noushmehr et al., The somatic genomic landscape of glioblastoma, Cell, vol.155, pp.462-477, 2013.

G. M. Brodeur, Neuroblastoma: biological insights into a clinical enigma, Nat. Rev. Cancer, vol.3, pp.203-216, 2003.

G. M. Brodeur, Spontaneous regression of neuroblastoma, Cell Tissue Res, vol.372, pp.277-286, 2018.

J. C. Buckner, S. L. Pugh, E. G. Shaw, M. R. Gilbert, G. Barger et al., Phase III study of radiation therapy (RT) with or without procarbazine, CCNU, and vincristine (PCV) in low-grade glioma: RTOG 9802 with Alliance, ECOG, and SWOG, J. Clin. Oncol, vol.32, 2000.

A. P. Bussink, D. Speijer, J. M. Aerts, and R. G. Boot, Evolution of mammalian chitinase(-like) members of family 18 glycosyl hydrolases, Genetics, vol.177, pp.959-970, 2007.

G. Cairncross, M. Wang, E. Shaw, R. Jenkins, D. Brachman et al., Phase III trial of chemoradiotherapy for anaplastic oligodendroglioma: long-term results of RTOG 9402, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.31, pp.337-343, 2013.

R. A. Cairns, I. S. Harris, and T. W. Mak, Regulation of cancer cell metabolism, Nat. Rev. Cancer, vol.11, pp.85-95, 2011.

C. Network, D. J. Brat, R. G. Verhaak, K. D. Aldape, W. K. Yung et al., Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas, N. Engl. J. Med, vol.372, pp.2481-2498, 2015.

D. Capper, H. Zentgraf, J. Balss, C. Hartmann, and A. Deimling, Mono-Licence CC BY-NC-ND 3.0 clonal antibody specific for IDH1 R132H mutation, Acta Neuropathol. (Berl.), vol.118, pp.599-601, 2009.

M. Ceccarelli, F. P. Barthel, T. M. Malta, T. S. Sabedot, S. R. Salama et al., Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma, Cell, vol.164, pp.550-563, 2016.

K. L. Chaichana, I. Jusue-torres, R. Navarro-ramirez, S. M. Raza, M. Pascual-gallego et al., Establishing percent resection and residual volume thresholds affecting survival and recurrence for patients with newly diagnosed intracranial glioblastoma, Neuro-Oncol, vol.16, pp.113-122, 2014.

M. C. Chamberlain and D. Born, Prognostic significance of relative 1p/19q codeletion in oligodendroglial tumors, J. Neurooncol, vol.125, pp.249-251, 2015.

M. V. Chao, M. A. Bothwell, A. H. Ross, H. Koprowski, A. A. Lanahan et al., Gene transfer and molecular cloning of the human NGF receptor, Science, vol.232, pp.518-521, 1986.

G. Chen, T. G. Gharib, C. Huang, J. M. Taylor, D. E. Misek et al., Discordant Protein and mRNA Expression in Lung Adenocarcinomas, Mol. Cell. Proteomics, vol.1, pp.304-313, 2002.

M. Cheray, D. Petit, L. Forestier, L. Karayan-tapon, A. Maftah et al., Glycosylation-related gene expression is linked to differentiation status in glioblastomas undifferentiated cells, Cancer Lett, vol.312, pp.24-32, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00873130

M. Cheray, B. Bessette, A. Lacroix, C. Mélin, S. Jawhari et al., KLRC3, a Natural Killer receptor gene, is a key factor involved in glioblastoma tumourigenesis and aggressiveness, J. Cell. Mol. Med, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01819164

O. L. Chinot, W. Wick, W. Mason, R. Henriksson, F. Saran et al., Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma, N. Engl. J. Med, vol.370, pp.709-722, 2014.

D. O. Clary and L. F. Reichardt, An alternatively spliced form of the nerve growth factor receptor TrkA confers an enhanced response to neurotrophin 3, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.11133-11137, 1994.

M. Colombo, G. Raposo, and C. Théry, Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles, Annu. Rev. Cell Dev. Biol, vol.30, pp.255-289, 2014.

C. Coutton, G. Vieville, V. Satre, F. Devillard, A. et al., Multiplex Ligation-dependent Probe Amplification (MLPA) et sondes « à façon » entièrement synthétiques. Guide pratique, recommandations et expérience au CHU de Grenoble, IRBM, vol.33, pp.227-235, 2012.

C. Daumas-duport and P. Varlet, , 2003.

, Rev. Neurol, vol.159, pp.622-636

L. Decoster, K. Van-puyvelde, S. Mohile, U. Wedding, U. Basso et al., Screening tools for multidimensional health problems warranting a geriatric assessment in older cancer patients: an update on SIOG recommendations ?, Ann. Oncol. Off. J. Eur. Soc. Med. Oncol, vol.26, pp.288-300, 2015.

E. Deluche, S. Leobon, F. Lamarche, and N. Tubiana-mathieu, First validation of the G-8 geriatric screening tool in older patients with glioblastoma, J. Geriatr. Oncol, p.0, 2018.

H. J. Dubbink, P. N. Atmodimedjo, J. M. Kros, P. J. French, M. Sanson et al., Molecular classification of anaplastic oligodendroglioma using next-generation sequencing: a report of the prospective randomized EORTC Brain Tumor Group 26951 phase III trial, Neuro-Oncol, vol.18, pp.388-400, 2016.

, Licence CC BY

F. Ducray, A. Idbaih, A. De-reyniès, I. Bièche, J. Thillet et al., Anaplastic oligodendrogliomas with 1p19q codeletion have a proneural gene expression profile, Mol. Cancer, vol.7, p.41, 2008.

H. Duffau, The reliability of asleep-awake-asleep protocol for intraoperative functional mapping and cognitive monitoring in glioma surgery, Acta Neurochir. (Wien), vol.155, pp.1803-1804, 2013.

H. Duffau, M. Gallot, A. Spittler, and M. Gardien, La chirurgie éveillée des tumeurs cérébrales, 2012.

J. E. Eckel-passow, D. H. Lachance, A. M. Molinaro, K. M. Walsh, P. A. Decker et al., Glioma Groups Based on 1p/19q, IDH, and TERT Promoter Mutations in Tumors, vol.372, pp.2499-2508, 2015.

R. Eid, M. Demattei, H. Episkopou, C. Augé-gouillou, A. Decottignies et al., Genetic Inactivation of ATRX Leads to a Decrease in the Amount of Telomeric Cohesin and Level of Telomere Transcription in Human Glioma Cells, Mol. Cell. Biol, vol.35, pp.2818-2830, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01224268

P. Ernfors, K. Lee, J. , and R. , Mice lacking brain-derived neurotrophic factor develop with sensory deficits, Nature, vol.368, pp.147-150, 1994.

P. Ernfors, K. F. Lee, J. Kucera, J. , and R. , Lack of neurotrophin-3 leads to deficiencies in the peripheral nervous system and loss of limb proprioceptive afferents, Cell, vol.77, pp.503-512, 1994.

A. F. Farago, L. P. Le, Z. Zheng, A. Muzikansky, A. Drilon et al., Durable Clinical Response to Entrectinib in NTRK1-Rearranged Non-Small Cell Lung Cancer, J. Thorac. Oncol. Off. Publ. Int. Assoc. Study Lung Cancer, vol.10, pp.1670-1674, 2015.

D. Figarella-branger, C. Colin, B. Coulibaly, B. Quilichini, A. M. Paula et al., Référentiel gliomes diffus de l'adulte de grade OMS II, III et IV : anatomie pathologique et biologie, Ann. Pathol, vol.58, pp.318-327, 2008.

P. A. Forsyth, N. Krishna, S. Lawn, J. G. Valadez, X. Qu et al., p75 neurotrophin receptor cleavage by ?-and ?-secretases is required for neurotrophin-mediated proliferation of brain tumor-initiating cells, J. Biol. Chem, vol.289, pp.8067-8085, 2014.

R. A. Francescone, S. Scully, M. Faibish, S. L. Taylor, D. Oh et al., Role of YKL-40 in the angiogenesis, radioresistance, and progression of glioblastoma, J. Biol. Chem, vol.286, pp.15332-15343, 2011.

M. M. Fuster and J. D. Esko, The sweet and sour of cancer: glycans as novel therapeutic targets, Nat. Rev. Cancer, vol.5, pp.526-542, 2005.

C. Giannini, B. W. Scheithauer, A. L. Weaver, P. C. Burger, J. M. Kros et al., Oligodendrogliomas: Reproducibility and Prognostic Value of Histologic Diagnosis and Grading, J. Neuropathol. Exp. Neurol, vol.60, pp.248-262, 2001.

R. J. Gibbons, D. J. Picketts, L. Villard, and D. R. Higgs, Mutations in a putative global transcriptional regulator cause X-linked mental retardation with alphathalassemia (ATR-X syndrome), Cell, vol.80, pp.837-845, 1995.

M. R. Gilbert, J. J. Dignam, T. S. Armstrong, J. S. Wefel, D. T. Blumenthal et al., A randomized trial of bevacizumab for newly diagnosed glioblastoma, N. Engl. J. Med, vol.370, pp.699-708, 2014.

E. Gillet, A. Alentorn, B. Doukouré, E. Mundwiller, H. F. Van-thuijl et al., TP53 and p53 statuses and their clinical impact in diffuse low grade gliomas, J. Neurooncol, vol.118, pp.131-139, 2014.

S. Giraud, E. Loum, B. Bessette, M. Mathonnet, and F. Lalloué, , 2011.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.145, 2018.

, Licence CC BY

, trophin receptor is sequestered in the Golgi apparatus of the U-87 MG human glioblastoma cell line, Int. J. Oncol, vol.38, pp.391-399

D. D. Gonda, J. C. Akers, R. Kim, S. N. Kalkanis, F. H. Hochberg et al., Neuro-oncologic applications of exosomes, microvesicles, and other nano-sized extracellular particles, Neurosurgery, vol.72, pp.501-510, 2013.

A. M. Goodman, S. Kato, L. Bazhenova, S. P. Patel, G. M. Frampton et al., Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers, Mol. Cancer Ther. molcanther, p.386, 2017.

J. Gourlay, A. P. Morokoff, R. B. Luwor, H. Zhu, A. H. Kaye et al., The emergent role of exosomes in glioma, J. Clin. Neurosci. Off. J. Neurosurg. Soc. Australas, vol.35, pp.13-23, 2017.

C. Gozé, M. Blonski, G. Le-maistre, L. Bauchet, E. Dezamis et al., Imaging growth and isocitrate dehydrogenase 1 mutation are independent predictors for diffuse low-grade gliomas, Neuro-Oncol, vol.16, pp.1100-1109, 2014.

L. A. Gravendeel, M. C. Kouwenhoven, O. Gevaert, J. J. De-rooi, A. P. Stubbs et al., Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology, Cancer Res, vol.69, pp.9065-9072, 2009.

M. A. Grotzer, A. J. Janss, P. C. Phillips, and J. Q. Trojanowski, Neurotrophin receptor TrkC predicts good clinical outcome in medulloblastoma and other primitive neuroectodermal brain tumors, Klin. Padiatr, vol.212, pp.196-199, 2000.

C. Gzell, M. Back, H. Wheeler, D. Bailey, and M. Foote, Radiotherapy in Glioblastoma: the Past, the Present and the Future, Clin. Oncol. R. Coll. Radiol. G. B, vol.29, pp.15-25, 2017.

S. Hakomori, Glycosylation defining cancer malignancy: New wine in an old bottle, Proc. Natl. Acad. Sci. U. S. A, vol.99, pp.10231-10233, 2002.

S. Hakomori and R. Kannagi, Glycosphingolipids as tumor-associated and differentiation markers, J. Natl. Cancer Inst, vol.71, pp.231-251, 1983.

F. Hallböök, Evolution of the vertebrate neurotrophin and Trk receptor gene families, Curr. Opin. Neurobiol, vol.9, pp.616-621, 1999.

F. Hallböök, C. F. Ibáñez, and H. Persson, Evolutionary studies of the nerve growth factor family reveal a novel member abundantly expressed in Xenopus ovary, Neuron, vol.6, pp.845-858, 1991.

K. Hansen, B. Wagner, W. Hamel, M. Schweizer, F. Haag et al., Autophagic cell death induced by TrkA receptor activation in human glioblastoma cells, J. Neurochem, vol.103, pp.259-275, 2007.

S. Harada, L. B. Henderson, J. R. Eshleman, C. D. Gocke, P. Burger et al., Genomic Changes in Gliomas Detected Using Single Nucleotide Polymorphism Array in Formalin-Fixed, Paraffin-Embedded Tissue: Superior Results Compared with Microsatellite Analysis, J. Mol. Diagn, vol.13, pp.541-548, 2011.

C. Hartmann, J. Meyer, J. Balss, D. Capper, W. Mueller et al., Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: a study of 1,010 diffuse gliomas, Acta Neuropathol. (Berl.), vol.118, pp.469-474, 2009.

D. H. Heiland, R. Ferrarese, R. Claus, F. Dai, A. P. Masilamani et al., c-Jun-N-terminal phosphorylation regulates DNMT1 expression and genome wide methylation in gliomas, Oncotarget, vol.8, pp.6940-6954, 2016.

J. D. Henson, J. A. Hannay, S. W. Mccarthy, J. A. Royds, T. R. Yeager et al., A robust assay for alternative lengthening of telomeres in tumors shows the significance of alternative leng, 2005.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.146, 2018.

, Licence CC BY

, thening of telomeres in sarcomas and astrocytomas, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.11, pp.217-225

K. Herrup, E. M. Shooter, F. R. Hirsch, M. Varella-garcia, P. A. Bunn et al., Epidermal growth factor receptor in non-small-cell lung carcinomas: correlation between gene copy number and protein expression and impact on prognosis, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.70, pp.3798-3807, 1973.

R. Ho, A. Eggert, T. Hishiki, J. E. Minturn, N. Ikegaki et al., Resistance to chemotherapy mediated by TrkB in neuroblastomas, Cancer Res, vol.62, pp.6462-6466, 2002.

A. Hohn, J. Leibrock, K. Bailey, and Y. A. Barde, Identification and characterization of a novel member of the nerve growth factor/brain-derived neurotrophic factor family, Nature, vol.344, pp.339-341, 1990.

C. Horbinski, G. Wang, and C. A. Wiley, YKL-40 is directly produced by tumor cells and is inversely linked to EGFR in glioblastomas, Int. J. Clin. Exp. Pathol, vol.3, pp.226-237, 2010.

A. Hormigo, B. Gu, S. Karimi, E. Riedel, K. S. Panageas et al., YKL-40 and matrix metalloproteinase-9 as potential serum biomarkers for patients with high-grade gliomas, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.12, pp.5698-5704, 2006.

E. J. Huang and L. F. Reichardt, Trk receptors: roles in neuronal signal transduction, Annu. Rev. Biochem, vol.72, pp.609-642, 2003.

T. Ius, M. Isola, R. Budai, G. Pauletto, B. Tomasino et al., Low-grade glioma surgery in eloquent areas: volumetric analysis of extent of resection and its impact on overall survival. A single-institution experience in 190 patients: clinical article, J. Neurosurg, vol.117, pp.1039-1052, 2012.

F. M. Iwamoto, A. F. Hottinger, S. Karimi, E. Riedel, J. Dantis et al., Serum YKL-40 is a marker of prognosis and disease status in high-grade gliomas, Neuro-Oncol, vol.13, pp.1244-1251, 2011.

R. Iyer, L. Wehrmann, R. L. Golden, K. Naraparaju, J. L. Croucher et al., Entrectinib is a potent inhibitor of Trk-driven neuroblastomas in a xenograft mouse model, Cancer Lett, vol.372, pp.179-186, 2016.

S. Jawhari, B. Bessette, S. Hombourger, K. Durand, A. Lacroix et al., Autophagy and TrkC/NT-3 signaling joined forces boost the hypoxic glioblastoma cell survival, Carcinogenesis, vol.38, pp.592-603, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01873444

R. B. Jenkins, H. Blair, K. V. Ballman, C. Giannini, R. M. Arusell et al., A t(1;19)(q10;p10) Mediates the Combined Deletions of 1p and 19q and Predicts a Better Prognosis of Patients with Oligodendroglioma, Cancer Res, vol.66, pp.9852-9861, 2006.

J. Jeuken, S. Cornelissen, S. Boots-sprenger, S. Gijsen, and P. Wesseling, , 2006.

, Multiplex ligation-dependent probe amplification: a diagnostic tool for simultaneous identification of different genetic markers in glial tumors, J. Mol. Diagn. JMD, vol.8, pp.433-443

Y. Jiao, P. J. Killela, Z. J. Reitman, B. A. Rasheed, C. M. Heaphy et al., Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas, Oncotarget, vol.3, pp.709-722, 2012.

J. S. Johansen, M. K. Williamson, J. S. Rice, and P. A. Price, Identification of proteins secreted by human osteoblastic cells in culture, J. Bone Miner. Res, vol.7, pp.501-512, 1992.

J. S. Johansen, B. V. Jensen, A. Roslind, D. Nielsen, and P. A. Price, Serum YKL-40, a new prognostic biomarker in cancer patients?, Cancer Epidemiol. Biomark. Prev, 2006.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.147, 2018.

, Licence CC BY

, Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol, vol.15, pp.194-202

J. S. Johansen, T. Lottenburger, H. J. Nielsen, J. E. Jensen, M. N. Svendsen et al., Diurnal, weekly, and long-time variation in serum concentrations of YKL-40 in healthy subjects, Cancer Epidemiol. Biomark. Prev. Publ. Am. Assoc. Cancer Res. Cosponsored Am. Soc. Prev. Oncol, vol.17, pp.2603-2608, 2008.

J. S. Johansen, N. A. Schultz, and B. V. Jensen, Plasma YKL-40: a potential new cancer biomarker?, Future Oncol. Lond. Engl, vol.5, pp.1065-1082, 2009.

B. E. Johnson, T. Mazor, C. Hong, M. Barnes, K. Aihara et al., Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma, Science, vol.343, pp.189-193, 2014.

A. L. Johnston, X. Lun, J. J. Rahn, A. Liacini, L. Wang et al., The p75 neurotrophin receptor is a central regulator of glioma invasion, PLoS Biol, vol.5, p.212, 2007.

S. V. Kalinichenko, E. P. Kopantzev, E. V. Korobko, I. V. Palgova, L. E. Zavalishina et al., Pdcd4 protein and mRNA level alterations do not correlate in human lung tumors, Lung Cancer, vol.62, pp.173-180, 2008.

A. Kamoun, A. Idbaih, C. Dehais, N. Elarouci, C. Carpentier et al., Integrated multi-omics analysis of oligodendroglial tumours identifies three subgroups of 1p/19q co-deleted gliomas, Nat. Commun, vol.7, p.11263, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01320022

D. R. Kaplan and F. D. Miller, Signal transduction by the neurotrophin receptors, Curr. Opin. Cell Biol, vol.9, pp.213-221, 1997.

C. Kenis, L. Decoster, K. Van-puyvelde, J. De-grève, G. Conings et al., Performance of two geriatric screening tools in older patients with cancer, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.32, pp.19-26, 2014.

J. W. Kernohan and R. F. Mabon, A simplified classification of the gliomas, Proc. Staff Meet. Mayo Clin, vol.24, pp.71-75, 1949.

Y. B. Khotskaya, V. R. Holla, A. F. Farago, K. R. Shaw, F. Meric-bernstam et al., Targeting TRK family proteins in cancer, Pharmacol. Ther, vol.173, pp.58-66, 2017.

P. J. Killela, C. J. Pirozzi, P. Healy, Z. J. Reitman, E. Lipp et al., Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas, Oncotarget, vol.5, pp.1515-1525, 2014.

S. Kimura, A. Yoshino, Y. Katayama, T. Watanabe, and T. Fukushima, , 2002.

, Growth control of C6 glioma in vivo by nerve growth factor, J. Neurooncol, vol.59, pp.199-205

P. Kleihues and L. H. Sobin, World Health Organization classification of tumors, Cancer, vol.88, p.2887, 2000.
URL : https://hal.archives-ouvertes.fr/hal-01479018

P. Kleihues, P. C. Burger, and B. W. Scheithauer, Definitions and explanatory notes, Histological Typing of Tumours of the Central Nervous System, pp.11-51, 1993.

R. Klein, V. Nanduri, S. A. Jing, F. Lamballe, P. Tapley et al., The trkB tyrosine protein kinase is a receptor for brain-derived neurotrophic factor and neurotrophin-3, Cell, vol.66, pp.395-403, 1991.

R. Klein, R. J. Smeyne, W. Wurst, L. K. Long, B. A. Auerbach et al., Targeted disruption of the trkB neurotrophin receptor gene results in nervous system lesions and neonatal death, Cell, vol.75, pp.113-122, 1993.

J. B. De-kok, R. W. Roelofs, B. A. Giesendorf, J. L. Pennings, E. T. Waas et al., Normalization of gene expression measurements in tumor tissues: comparison of 13 endogenous control genes, Lab. Investig. J. Tech. Methods Pathol, vol.85, pp.154-159, 2005.

A. Koussounadis, S. P. Langdon, I. H. Um, D. J. Harrison, and V. A. Smith, Relationship between differentially expressed mRNA and mRNA-protein correlations in a, 2015.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.148, 2018.

, Licence CC BY-NC-ND 3.0 xenograft model system, Sci. Rep, vol.5

K. Kramer, N. V. Cheung, W. L. Gerald, M. Laquaglia, B. H. Kushner et al., Correlation of MYCN amplification, Trk-A and CD44 expression with clinical stage in 250 patients with neuroblastoma, Eur. J. Cancer, vol.33, pp.2098-2100, 1997.

R. A. Kroes, G. Dawson, and J. R. Moskal, Focused microarray analysis of glyco-gene expression in human glioblastomas, J. Neurochem, vol.103, issue.1, pp.14-24, 2007.

J. M. Kros, T. Gorlia, M. C. Kouwenhoven, P. Zheng, V. P. Collins et al., Panel review of anaplastic oligodendroglioma from European Organization For Research and Treatment of Cancer Trial 26951: assessment of consensus in diagnosis, influence of 1p/19q loss, and correlations with outcome, J. Neuropathol. Exp. Neurol, vol.66, pp.545-551, 2007.

B. M. Ku, Y. K. Lee, J. Ryu, J. Y. Jeong, J. Choi et al., CHI3L1 (YKL-40) is expressed in human gliomas and regulates the invasion, growth and survival of glioma cells, Int. J. Cancer, vol.128, pp.1316-1326, 2011.

, Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.7312-7317

F. Lamballe, R. Klein, and M. Barbacid, trkC, a new member of the trk family of tyrosine protein kinases, is a receptor for neurotrophin-3, Cell, vol.66, pp.967-979, 1991.

A. M. Lange and H. Lo, Inhibiting TRK Proteins in Clinical Cancer Therapy, Cancers, vol.10, 2018.

N. Laperriere, L. Zuraw, and G. Cairncross, and Cancer Care Ontario Practice Guidelines Initiative Neuro-Oncology Disease Site Group, Radiother. Oncol. J. Eur. Soc. Ther. Radiol. Oncol, vol.64, pp.259-273, 2002.

S. Lawn, N. Krishna, A. Pisklakova, X. Qu, D. A. Fenstermacher et al., Neurotrophin signaling via TrkB and TrkC receptors promotes the growth of brain tumor-initiating cells, J. Biol. Chem, vol.290, pp.3814-3824, 2015.

K. F. Lee, E. Li, L. J. Huber, S. C. Landis, A. H. Sharpe et al., Targeted mutation of the gene encoding the low affinity NGF receptor p75 leads to deficits in the peripheral sensory nervous system, Cell, vol.69, pp.737-749, 1992.

R. Lee, P. Kermani, K. K. Teng, and B. L. Hempstead, Regulation of cell survival by secreted proneurotrophins, Science, vol.294, pp.1945-1948, 2001.

H. E. Leeper, A. A. Caron, P. A. Decker, R. B. Jenkins, D. H. Lachance et al., IDH mutation, 1p19q codeletion and ATRX loss in WHO grade II gliomas, Oncotarget, vol.6, pp.30295-30305, 2015.

V. Lessmann, K. Gottmann, and M. Malcangio, Neurotrophin secretion: current facts and future prospects, Prog. Neurobiol, vol.69, pp.341-374, 2003.

R. Levi-montalcini, Effects of mouse tumor transplantation on the nervous system, Ann. N. Y. Acad. Sci, vol.55, pp.330-344, 1952.

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

S. Li, J. Shi, H. Gao, Y. Yuan, Q. Chen et al., Identification of a gene signature associated with radiotherapy and prognosis in gliomas, Oncotarget, vol.5, pp.88974-88987, 2014.

A. L. Lin and L. M. Deangelis, Reappraising the 2016 WHO classification for diffuse glioma, Neuro-Oncol, vol.19, pp.609-610, 2017.

M. E. Van-linde, J. C. Van-der-mijn, T. V. Pham, J. C. Knol, L. E. Wedekind et al., , vol.21, p.149, 2018.

, Licence CC BY

K. E. Aliaga, E. S. Buter, J. Jimenez, C. R. Reijneveld, and J. C. , Evaluation of potential circulating biomarkers for prediction of response to chemoradiation in patients with glioblastoma, J. Neurooncol, vol.129, pp.221-230, 2016.

D. Liu, M. Offin, S. Harnicar, B. T. Li, and A. Drilon, Entrectinib: an orally available, selective tyrosine kinase inhibitor for the treatment of NTRK, ROS1, and ALK fusion-positive solid tumors, Ther. Clin. Risk Manag, vol.14, pp.1247-1252, 2018.

X. Liu, P. Ernfors, H. Wu, and R. Jaenisch, Sensory but not motor neuron deficits in mice lacking NT4 and BDNF, Nature, vol.375, pp.238-241, 1995.

X. Liu, N. Gerges, A. Korshunov, N. Sabha, D. Khuong-quang et al., Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations, Acta Neuropathol. (Berl.), vol.124, pp.615-625, 2012.

E. Liuu, F. Canouï-poitrine, C. Tournigand, M. Laurent, M. Chaubet et al., External validation of the G-8 geriatric screening tool to identify vulnerable elderly cancer patients: The ELCAPA-02 study, J. Geriatr. Oncol, vol.3, pp.45-46, 2012.

E. Liuu, F. Canouï-poitrine, C. Tournigand, M. Laurent, P. Caillet et al., Accuracy of the G-8 geriatric-oncology screening tool for identifying vulnerable elderly patients with cancer according to tumour site: The ELCAPA-02 study, J. Geriatr. Oncol, vol.5, pp.11-19, 2014.

K. J. Livak and T. D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods San Diego Calif, vol.25, pp.402-408, 2001.

H. Loiseau, A. Huchet, M. Rué, A. Cowppli-bony, and I. Baldi, Épidémiologie des tumeurs cérébrales primitives, Rev. Neurol, vol.165, pp.650-670, 2009.

D. N. Louis, E. C. Holland, and J. G. Cairncross, Glioma classification: a molecular reappraisal, Am. J. Pathol, vol.159, pp.779-786, 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 Neuropathol. (Berl.), vol.114, pp.97-109, 2007.

D. N. Louis, A. Perry, P. Burger, D. W. Ellison, G. Reifenberger et al., International Society Of Neuropathology--Haarlem consensus guidelines for nervous system tumor classification and grading, Brain Pathol. Zurich Switz, vol.24, pp.429-435, 2014.

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 Neuropathol. (Berl.), vol.131, pp.803-820, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01479018

D. Loussouarn, A. Le-loupp, J. Frenel, F. Leclair, A. Von-deimling et al., Comparison of immunohistochemistry, DNA sequencing and allele-specific PCR for the detection of IDH1 mutations in gliomas, Int. J. Oncol, vol.40, pp.2058-2062, 2012.

C. Lu, P. S. Ward, G. S. Kapoor, D. Rohle, S. Turcan et al., IDH mutation impairs histone demethylation and results in a block to cell differentiation, Nature, vol.483, pp.474-478, 2012.

K. Luberg, J. Wong, C. S. Weickert, and T. Timmusk, Human TrkB gene: novel alternative transcripts, protein isoforms and expression pattern in the prefrontal cerebral cortex during postnatal development, J. Neurochem, vol.113, pp.952-964, 2010.

A. Malmström, B. H. Grønberg, C. Marosi, R. Stupp, D. Frappaz et al., Temozolomide versus standard 6-week radiotherapy versus hypofractionated radiotherapy in patients older than 60 years with glioblastoma: the Nordic randomised, Lancet Oncol, vol.13, pp.916-926, 2012.

C. Martinez-tapia, E. Paillaud, E. Liuu, C. Tournigand, R. Ibrahim et al., , vol.21, p.150, 2018.

, Licence CC BY

V. Culine, S. Canoui-poitrine, F. Audureau, and E. , Prognostic value of the G8 and modified-G8 screening tools for multidimensional health problems in older patients with cancer, Eur. J. Cancer, vol.83, pp.211-219, 2017.

D. Martin-zanca, S. H. Hughes, and M. Barbacid, A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences, Nature, vol.319, pp.743-748, 1986.

S. A. Melo, H. Sugimoto, J. T. O'connell, N. Kato, A. Villanueva et al., Cancer exosomes perform cellindependent microRNA biogenesis and promote tumorigenesis, Cancer Cell, vol.26, pp.707-721, 2014.

L. Mesturoux, K. Durand, I. Pommepuy, S. Robert, F. Caire et al., Molecular Analysis of Tumor Cell Components in Pilocytic Astrocytomas, Gangliogliomas, and Oligodendrogliomas, Appl. Immunohistochem. Mol. Morphol. AIMM, vol.24, pp.496-500, 2016.

P. Metellus, B. Coulibaly, C. Colin, A. M. De-paula, A. Vasiljevic et al., Absence of IDH mutation identifies a novel radiologic and molecular subtype of WHO grade II gliomas with dismal prognosis, Acta Neuropathol. (Berl.), vol.120, pp.719-729, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01848106

A. Modelska, A. Quattrone, and A. Re, Molecular portraits: the evolution of the concept of transcriptome-based cancer signatures, Brief. Bioinform, vol.16, pp.1000-1007, 2015.

K. W. Moremen, M. Tiemeyer, and A. V. Nairn, Vertebrate protein glycosylation: diversity, synthesis and function, Nat. Rev. Mol. Cell Biol, vol.13, p.448, 2012.

K. V. Moser, M. Reindl, I. Blasig, and C. Humpel, Brain capillary endothelial cells proliferate in response to NGF, express NGF receptors and secrete NGF after inflammation, Brain Res, vol.1017, pp.53-60, 2004.

P. A. Muller and K. H. Vousden, p53 mutations in cancer, Nat. Cell Biol, vol.15, pp.2-8, 2013.

P. A. Muller and K. H. Vousden, Mutant p53 in Cancer: New Functions and Therapeutic Opportunities, Cancer Cell, vol.25, pp.304-317, 2014.

A. Nakagawara, Trk receptor tyrosine kinases: a bridge between cancer and neural development, Cancer Lett, vol.169, pp.107-114, 2001.

A. Nakagawara, M. Arima, C. G. Azar, N. J. Scavarda, and G. M. Brodeur, Inverse relationship between trk expression and N-myc amplification in human neuroblastomas, Cancer Res, vol.52, pp.1364-1368, 1992.

A. Nakagawara, C. G. Azar, N. J. Scavarda, and G. M. Brodeur, Expression and function of TRK-B and BDNF in human neuroblastomas, Mol. Cell. Biol, vol.14, pp.759-767, 1994.

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, The Role of ATRX in Glioma Biology. Front. Oncol. 7, vol.22, pp.1715-1719, 2001.

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.

H. Noushmehr, D. J. Weisenberger, K. Diefes, H. S. Phillips, K. Pujara et al., Identification of a CpG Island Methylator Phenotype that Defines a Distinct Subgroup of Glioma, Cancer Cell, vol.17, pp.510-522, 2010.
URL : https://hal.archives-ouvertes.fr/hal-01201573

C. L. Nutt, D. R. Mani, R. A. Betensky, P. Tamayo, J. G. Cairncross et al., Gene expression-based classification of malignant gliomas correlates better with survival than histological classification, Cancer Res, vol.63, pp.1602-1607, 2003.

C. L. Nutt, R. A. Betensky, M. A. Brower, T. T. Batchelor, D. N. Louis et al.,

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.151, 2018.

, Licence CC BY

A. O. Rachamimov, YKL-40 is a differential diagnostic marker for histologic subtypes of high-grade gliomas, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.11, pp.2258-2264, 2005.

H. Ohgaki and P. Kleihues, Population-based studies on incidence, survival rates, and genetic alterations in astrocytic and oligodendroglial gliomas, J. Neuropathol. Exp. Neurol, vol.64, pp.479-489, 2005.

H. Ohgaki and P. Kleihues, The definition of primary and secondary glioblastoma, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.19, pp.764-772, 2013.

Y. Okamoto, P. Di-patre, C. Burkhard, S. Horstmann, B. Jourde et al., Populationbased study on incidence, survival rates, and genetic alterations of low-grade diffuse astrocytomas and oligodendrogliomas, Acta Neuropathol. (Berl.), vol.108, pp.49-56, 2004.

A. Olar, K. M. Wani, K. D. Alfaro-munoz, L. E. Heathcock, H. F. Van-thuijl et al., IDH mutation status and role of WHO grade and mitotic index in overall survival in grade II-III diffuse gliomas, Acta Neuropathol. (Berl.), vol.129, pp.585-596, 2015.

Q. T. Ostrom, L. Bauchet, F. G. Davis, I. Deltour, J. L. Fisher et al., The epidemiology of glioma in adults: a "state of the science, review. Neuro-Oncol, vol.16, pp.896-913, 2014.

Q. T. Ostrom, H. Gittleman, J. Fulop, M. Liu, R. Blanda et al., CBTRUS Statistical Report: Primary Brain and Central Nervous System Tumors Diagnosed in the United States, Neuro-Oncol, vol.17, pp.1-62, 2008.

J. Pallud, E. Mandonnet, H. Duffau, M. Kujas, R. Guillevin et al., Prognostic value of initial magnetic resonance imaging growth rates for World Health Organization grade II gliomas, Ann. Neurol, vol.60, pp.380-383, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00163977

D. W. Parsons, S. Jones, X. Zhang, J. Lin, R. J. Leary et al., An integrated genomic analysis of human glioblastoma multiforme, Science, vol.321, pp.1807-1812, 2008.

C. E. Pelloski, A. Mahajan, M. Maor, E. L. Chang, S. Woo et al., YKL-40 expression is associated with poorer response to radiation and shorter overall survival in glioblastoma, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.11, pp.3326-3334, 2005.

J. R. Perry, N. Laperriere, C. J. O'callaghan, A. A. Brandes, J. Menten et al., Short-course radiation plus temozolomide in elderly patients with glioblastoma, N. Engl. J. Med, vol.376, pp.1027-1037, 2017.

B. R. Pflug, A. M. Colangelo, C. Tornatore, and I. Mocchetti, TrkA induces differentiation but not apoptosis in C6-2B glioma cells, J. Neurosci. Res, vol.64, pp.636-645

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

F. Pignatti, M. Van-den-bent, D. Curran, C. Debruyne, R. Sylvester et al., Prognostic factors for survival in adult patients with cerebral low-grade glioma, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.20, pp.2076-2084, 2002.

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, pp.50349-50364, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01819184

S. S. Pinho and C. A. Reis, Glycosylation in cancer: mechanisms and clinical implications, Nat. Rev. Cancer, vol.15, pp.540-555, 2015.

G. Qin, X. Li, Z. Chen, G. Liao, Y. Su et al., Prognostic Value of YKL-40 in Patients with Glioblastoma: a Systematic Review and Meta-analysis, 2017.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.152, 2018.

, Licence CC BY

, Mol. Neurobiol, vol.54, pp.3264-3270

V. Quillien, A. Lavenu, F. Ducray, M. Joly, O. Chinot et al., Validation of the highperformance of pyrosequencing for clinical MGMT testing on a cohort of glioblastoma patients from a prospective dedicated multicentric trial, Oncotarget, vol.7, pp.61916-61929, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01358205

M. Rehli, S. W. Krause, R. Andreesen, G. Reifenberger, H. Wirsching et al., Molecular characterization of the gene for human cartilage gp-39 (CHI3L1), a member of the chitinase protein family and marker for late stages of macrophage differentiation, Genomics, vol.43, pp.221-225, 1997.

, Advances in the molecular genetics of gliomas -implications for classification and therapy, Nat. Rev. Clin. Oncol, vol.14, pp.434-452

G. H. Renkema, R. G. Boot, F. L. Au, W. E. Donker-koopman, A. Strijland et al., Chitotriosidase, a chitinase, and the 39-kDa human cartilage glycoprotein, a chitin-binding lectin, are homologues of family 18 glycosyl hydrolases secreted by human macrophages, Eur. J. Biochem, vol.251, pp.504-509, 1998.

D. E. Reuss, Y. Mamatjan, D. Schrimpf, D. Capper, V. Hovestadt et al., IDH mutant diffuse and anaplastic astrocytomas have similar age at presentation and little difference in survival: a grading problem for WHO, Acta Neuropathol. (Berl.), vol.129, pp.867-873, 2015.

V. Rigau, S. Zouaoui, H. Mathieu-daudé, A. Darlix, A. Maran et al., French brain tumor database: 5-year histological results on 25 756 cases, Brain Pathol. Zurich Switz, vol.21, pp.633-644, 2011.

M. Ringsholt, E. V. Høgdall, J. S. Johansen, P. A. Price, and L. H. Christensen, YKL-40 protein expression in normal adult human tissues--an immunohistochemical study, J. Mol. Histol, vol.38, pp.33-43, 2007.

A. Rodríguez-tébar, G. Dechant, R. Götz, and Y. A. Barde, Binding of neurotrophin-3 to its neuronal receptors and interactions with nerve growth factor and brainderived neurotrophic factor, EMBO J, vol.11, pp.917-922, 1992.

P. P. Roux and P. A. Barker, Neurotrophin signaling through the p75 neurotrophin receptor, Prog. Neurobiol, vol.67, pp.203-233, 2002.

M. V. Ruiz-pérez, A. B. Henley, and M. Arsenian-henriksson, The MYCN Protein in Health and Disease, Genes, vol.8, 2017.

A. Saidi, S. Javerzat, A. Bellahcène, J. De-vos, L. Bello et al., Experimental anti-angiogenesis causes upregulation of genes associated with poor survival in glioblastoma, Int. J. Cancer, vol.122, pp.2187-2198, 2008.

N. Sanai, M. Polley, M. W. Mcdermott, A. T. Parsa, M. S. Berger et al., Abstract LB-302: Potential role of larotrectinib (LOXO-101), a selective pan-TRK inhibitor, in NTRK fusion-positive recurrent glioblastoma, J. Neurosurg, vol.115, pp.3-8, 2011.

A. Schroeder, O. Mueller, S. Stocker, R. Salowsky, M. Leiber et al., The RIN: an RNA integrity number for assigning integrity values to RNA measurements, BMC Mol. Biol, vol.7, p.3, 2006.

R. A. Segal, L. C. Goumnerova, Y. K. Kwon, C. D. Stiles, and S. L. Pomeroy, Expression of the neurotrophin receptor TrkC is linked to a favorable outcome in medulloblastoma, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.12867-12871, 1994.

Z. Shajani-yi, F. B. De-abreu, J. D. Peterson, and G. J. Tsongalis, Frequency of Somatic TP53 Mutations in Combination with Known Pathogenic Mutations in Colon Adenocarcinoma, Non-Small Cell Lung Carcinoma, and Gliomas as Identified by Next-Generation Sequencing, vol.20, pp.256-262, 2018.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.153, 2018.

C. Licence and R. Shao, YKL-40 acts as an angiogenic factor to promote tumor angiogenesis, Front. Physiol, vol.4, 2013.

R. Shao, R. Francescone, N. Ngernyuang, B. Bentley, S. L. Taylor et al., Anti-YKL-40 antibody and ionizing irradiation synergistically inhibit tumor vascularization and malignancy in glioblastoma, Carcinogenesis, vol.35, pp.373-382, 2014.

G. Shin, T. Kang, S. Yang, S. Baek, Y. Jeong et al., GENT: gene expression database of normal and tumor tissues, Cancer Inform, vol.10, pp.149-157, 2011.

M. Shirahata, K. Iwao-koizumi, S. Saito, N. Ueno, M. Oda et al., Gene expression-based molecular diagnostic system for malignant gliomas is superior to histological diagnosis, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.13, pp.7341-7356, 2007.

K. Shostak, V. Labunskyy, V. Dmitrenko, T. Malisheva, M. Shamayev et al., HC gp-39 gene is upregulated in glioblastomas, Cancer Lett, vol.198, pp.203-210, 2003.

J. Skog, T. Würdinger, S. Van-rijn, D. H. Meijer, L. Gainche et al., Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers, Nat. Cell Biol, vol.10, pp.1470-1476, 2008.

R. J. Smeyne, R. Klein, A. Schnapp, L. K. Long, S. Bryant et al., Severe sensory and sympathetic neuropathies in mice carrying a disrupted Trk/NGF receptor gene, Nature, vol.368, pp.246-249, 1994.

A. Sottoriva, I. Spiteri, S. G. Piccirillo, A. Touloumis, V. P. Collins et al., Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.4009-4014, 2013.

P. Soubeyran, M. Fonck, C. Blanc-bisson, J. Blanc, J. Ceccaldi et al., Predictors of early death risk in older patients treated with first-line chemotherapy for cancer, J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol, vol.30, pp.1829-1834, 2012.

P. Soubeyran, C. Bellera, J. Goyard, D. Heitz, H. Curé et al., Screening for Vulnerability in Older Cancer Patients: The ONCODAGE Prospective Multicenter Cohort Study, PLoS ONE, vol.9, 2014.

C. F. De-souza, T. S. Sabedot, T. M. Malta, L. Stetson, O. Morozova et al., A Distinct DNA Methylation Shift in a Subset of Glioma CpG Island Methylator Phenotypes during Tumor Recurrence, Cell Rep, vol.23, pp.637-651, 2018.

G. Steponaitis, D. Skiriut?, A. Kazlauskas, I. Golubickait?, R. Stakaitis et al., Analysis of the human TrkB gene genomic organization reveals novel TrkB isoforms, unusual gene length, and splicing mechanism, Biochem. Biophys. Res. Commun, vol.11, pp.1054-1065, 2002.

J. M. Stokoe, J. Pearce, R. Sinha, and A. Ring, G8 and VES-13 scores predict chemotherapy toxicity in older patients with cancer, J. Geriatr. Oncol, vol.3, p.81, 2012.

R. Stupp, W. P. Mason, M. J. Van-den-bent, M. Weller, B. Fisher et al., Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma, N. Engl. J. Med, vol.352, pp.987-996, 2005.

R. Stupp, M. E. Hegi, T. Gorlia, S. C. Erridge, J. Perry et al., Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): a multicentre, randomised, open-label, Lancet Oncol, vol.15, pp.1100-1108, 2014.

H. Suzuki, K. Aoki, K. Chiba, Y. Sato, Y. Shiozawa et al.,

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.154, 2018.

, Licence CC BY

A. Niida, K. Motomura, and F. Ohka, Mutational landscape and clonal architecture in grade II and III gliomas, Nat. Genet, vol.47, pp.458-468, 2015.

E. Tabouret, A. T. Nguyen, C. Dehais, C. Carpentier, F. Ducray et al., Prognostic impact of the 2016 WHO classification of diffuse gliomas in the French POLA cohort, Acta Neuropathol. (Berl.), vol.132, pp.625-634, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01480146

A. Tacconelli, A. R. Farina, L. Cappabianca, A. Gulino, and A. R. Mackay, , 2005.

, Alternative TrkAIII splicing: a potential regulated tumor-promoting switch and therapeutic target in neuroblastoma, Future Oncol. Lond. Engl, vol.1, pp.689-698

M. K. Tanwar, M. R. Gilbert, and E. C. Holland, Gene expression microarray analysis reveals YKL-40 to be a potential serum marker for malignant character in human glioma, Cancer Res, vol.62, pp.4364-4368, 2002.

S. Tauszig-delamasure, L. Yu, J. R. Cabrera, J. Bouzas-rodriguez, C. Mermet-bouvier et al., The TrkC receptor induces apoptosis when the dependence receptor notion meets the neurotrophin paradigm, Proc. Natl. Acad. Sci. U. S. A, vol.104, pp.13361-13366, 2007.

K. K. Teng and B. L. Hempstead, Neurotrophins and their receptors: signaling trios in complex biological systems, Cell. Mol. Life Sci. CMLS, vol.61, pp.35-48, 2004.

H. K. Teng, K. K. Teng, R. Lee, S. Wright, S. Tevar et al., ProBDNF Induces Neuronal Apoptosis via Activation of a Receptor Complex of p75NTR and Sortilin, J. Neurosci, vol.25, pp.5455-5463, 2005.

C. Théry, S. Amigorena, G. Raposo, C. , and A. , Isolation and characterization of exosomes from cell culture supernatants and biological fluids, Curr. Protoc. Cell Biol. Chapter, 2006.

E. G. Trams, C. J. Lauter, N. Salem, and U. Heine, Exfoliation of membrane ecto-enzymes in the form of micro-vesicles, Biochim. Biophys. Acta, vol.645, pp.63-70, 1981.

L. Treps, R. Perret, S. Edmond, D. Ricard, and J. Gavard, Glioblastoma stem-like cells secrete the pro-angiogenic VEGF-A factor in extracellular vesicles, J. Extracell, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01573268

S. Turcan, D. Rohle, A. Goenka, L. A. Walsh, F. Fang et al., IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype, Nature, vol.483, pp.479-483, 2012.

E. G. Van-meir, C. G. Hadjipanayis, A. D. Norden, H. Shu, P. Y. Wen et al., Exciting New Advances in Neuro-Oncology, CA. Cancer J. Clin, vol.60, pp.166-193, 2010.

A. Varki, R. Kannagi, B. Toole, P. Stanley, R. D. Varki et al., Glycosylation Changes in Cancer, Essentials of Glycobiology, 2015.

R. G. Verhaak, K. A. Hoadley, E. Purdom, V. Wang, Y. Qi et al., An integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR and NF1, Cancer Cell, vol.17, p.98, 2010.

F. Vincent, T. Naves, C. M. Wilson, F. Lalloué, M. O. Jauberteau et al., Sortilin mediates the release and transfer of exosomes in human lung cancer A549 cells, Rev. Mal. Respir, vol.32, p.324, 2015.

A. P. Vogazianou, R. Chan, L. M. Bäcklund, D. M. Pearson, L. Liu et al., Distinct patterns of 1p and 19q alterations identify subtypes of human gliomas that have different prognoses, Neuro-Oncol, vol.12, pp.664-678, 2010.

B. Vogelstein, N. Papadopoulos, V. E. Velculescu, S. Zhou, L. A. Diaz et al., Cancer Genome Landscapes. Science, vol.339, pp.1546-1558, 2013.

J. De-vrij, S. L. Maas, K. M. Kwappenberg, R. Schnoor, A. Kleijn et al., Glioblastoma, 2015.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.155, 2018.

C. C. Licence and . By-nc-nd-3, 0 derived extracellular vesicles modify the phenotype of monocytic cells, Int. J. Cancer, vol.137, pp.1630-1642

S. Wadhwa, T. C. Nag, A. Jindal, R. Kushwaha, A. K. Mahapatra et al., Expression of the neurotrophin receptors Trk A and Trk B in adult human astrocytoma and glioblastoma, J. Biosci, vol.28, pp.181-188, 2003.

H. Wang, K. Tang, T. Liang, W. Zhang, J. Li et al., The comparison of clinical and biological characteristics between IDH1 and IDH2 mutations in gliomas, J. Exp. Clin. Cancer Res. CR, vol.35, p.86, 2016.

L. Wang, J. J. Rahn, X. Lun, B. Sun, J. J. Kelly et al., Gamma-Secretase Represents a Therapeutic Target for the Treatment of Invasive Glioma Mediated by the p75 Neurotrophin Receptor, PLOS Biol, vol.6, p.289, 2008.

T. Wang, S. Luo, C. Lin, P. Chang, C. et al., Modulation of p75 neurotrophin receptor under hypoxic conditions induces migration and invasion of C6 glioma cells, Clin. Exp. Metastasis, vol.32, pp.73-81, 2015.

Y. Wang, C. Hagel, W. Hamel, S. Müller, L. Kluwe et al., Trk A, B, and C are commonly expressed in human astrocytes and astrocytic gliomas but not by human oligodendrocytes and oligodendroglioma, Acta Neuropathol. (Berl.), vol.96, pp.357-364, 1998.

M. Weller, R. Stupp, M. E. Hegi, M. Van-den-bent, J. C. Tonn et al., Personalized care in neuro-oncology coming of age: why we need MGMT and 1p/19q testing for malignant glioma patients in clinical practice, Neuro-Oncol, vol.14, pp.100-108, 2012.

M. Weller, M. Van-den-bent, J. C. Tonn, R. Stupp, M. Preusser et al., European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas, Lancet Oncol, vol.18, pp.315-329, 2017.

P. Wesseling, M. Van-den-bent, P. , and A. , Oligodendroglioma: pathology, molecular mechanisms and markers, Acta Neuropathol. (Berl.), vol.129, pp.809-827, 2015.

B. Wiestler, D. Capper, T. Holland-letz, A. Korshunov, . Deimling et al., ATRX loss refines the classification of anaplastic gliomas and identifies a subgroup of IDH mutant astrocytic tumors with better prognosis, Acta Neuropathol. (Berl.), vol.126, pp.443-451, 2013.

C. M. Wilson, T. Naves, F. Vincent, B. Melloni, F. Bonnaud et al., Sortilin mediates the release and transfer of exosomes in concert with two tyrosine kinase receptors, J. Cell Sci, vol.127, pp.3983-3997, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01059696

L. Xia, B. Wu, Z. Fu, F. Feng, E. Qiao et al., Prognostic role of IDH mutations in gliomas: a meta-analysis of 55 observational studies, Oncotarget, vol.6, pp.17354-17365, 2015.

Z. X. Xiao, R. Q. Chen, D. X. Hu, X. Q. Xie, S. B. Yu et al., Identification of repaglinide as a therapeutic drug for glioblastoma multiforme, Biochem. Biophys. Res. Commun, vol.488, pp.33-39, 2017.

J. Xiong, L. Zhou, Y. Lim, M. Yang, Y. Zhu et al., Mature BDNF promotes the growth of glioma cells in vitro, Oncol. Rep, vol.30, pp.2719-2724, 2013.

J. Xiong, L. Zhou, M. Yang, Y. Lim, Y. Zhu et al., ProBDNF and its receptors are upregulated in glioma and inhibit the growth of glioma cells in vitro, Neuro-Oncol, vol.15, pp.990-1007, 2013.

J. Xiong, L. I. Zhou, Y. Lim, M. Yang, Y. Zhu et al., Mature brain-derived neurotrophic factor and its receptor TrkB are upregulated in human glioma tissues, Oncol. Lett, vol.10, pp.223-227, 2015.

D. J. Yamashiro, A. Nakagawara, N. Ikegaki, X. G. Liu, and G. M. Brodeur, Expression of TrkC in favorable human neuroblastomas, Oncogene, vol.12, pp.37-41, 1996.

D. Elise and . De-doctorat-|-université-de-limoges, , vol.21, p.156, 2018.

, Licence CC BY

H. Yan, D. W. Parsons, G. Jin, R. Mclendon, B. A. Rasheed et al., IDH1 and IDH2 Mutations in Gliomas, N. Engl. J. Med, vol.360, pp.765-773, 2009.

B. Zhang, J. Wang, X. Wang, J. Zhu, Q. Liu et al., Proteogenomic characterization of human colon and rectal cancer, Nature, vol.513, pp.382-387, 2014.

L. Zhang, Z. Zhang, Y. Liu, Y. Xue, and I. Parney, Programmed deathligand 1 (PD-L1) may play a role in malignant glioma infiltration, Med. Hypotheses, vol.85, pp.127-129, 2015.

W. Zhang, M. Kawanishi, K. Miyake, M. Kagawa, N. Kawai et al., Association between YKL-40 and adult primary astrocytoma, Cancer, vol.116, pp.2688-2697, 2010.

S. Zouaoui, A. Darlix, P. Fabbro-peray, H. Mathieu-daudé, V. Rigau et al., Oncological patterns of care and outcomes for 265 elderly patients with newly diagnosed glioblastoma in France, Neurosurg. Rev, vol.37, pp.415-423, 2014.