Q. T. Ostrom, H. Gittleman, and P. Liao, CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010-2014, Neuro-Oncol, vol.19, issue.S5, pp.1-88, 2017.

D. A. Reardon and P. Y. Wen, Glioma in 2014: unravelling tumour heterogeneity-implications for therapy, Nat Rev Clin Oncol, vol.12, pp.69-70, 2015.

G. Riddick and H. A. Fine, Integration and analysis of genome-scale data from gliomas, Nat Rev Neurol, vol.7, pp.439-50, 2011.

D. Sturm, H. Witt, and V. Hovestadt, Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma, Cancer Cell, vol.22, pp.425-462, 2012.

C. W. Brennan, R. G. Verhaak, and A. Mckenna, The somatic genomic landscape of glioblastoma, Cell, vol.155, pp.462-77, 2013.

D. N. Louis, A. Perry, and G. Reifenberger, The 2016 world health organization classification of tumors of the central nervous system: a summary, Acta Neuropathol, vol.131, pp.803-823, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01479018

A. P. Patel, I. Tirosh, and J. J. Trombetta, Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma, Science, vol.344, pp.1396-401, 2014.

A. Sottoriva, I. Spiteri, and S. G. Piccirillo, Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics, Proc Natl Acad Sci U S A, vol.110, pp.4009-4023, 2013.

N. Sanai, A. Alvarez-buylla, and M. S. Berger, Neural stem cells and the origin of gliomas, N Engl J Med, vol.353, pp.811-833, 2005.

E. C. Holland, Progenitor cells and glioma formation, Curr Opin Neurol, vol.14, pp.683-691, 2001.

F. Lefranc, Cancer Treatment Reviews, vol.68, pp.145-154, 2018.

E. Gould, How widespread is adult neurogenesis in mammals?, Nature Rev Neurosci, vol.8, pp.481-489, 2007.

M. C. Nunes, N. S. Roy, and H. M. Keyoung, Identification and isolation of multipotential neural progenitor cells from the subcortical white matter of the adult human brain, Nature Med, vol.9, pp.439-486, 2003.

P. Canoll and J. E. Goldman, The interface between glial progenitors and gliomas, Acta Neuropathol, vol.116, pp.465-77, 2008.

C. Calabrese, H. Poppleton, and M. Kocak, A perivascular niche for brain tumor stem cells, Cancer Cell, vol.11, pp.69-82, 2007.

G. B. Pierce, Neoplasms, differentiations and mutations, Am J Pathol, vol.77, pp.103-121, 1974.

E. L. Jackson, J. M. Garcia-verdugo, and S. Gil-perotin, PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling, Neuron, vol.51, pp.187-99, 2006.

N. Sanai, A. D. Tramontin, and A. Quiñones-hinojosa, Unique astrocyte ribbon in adult human brain contains neural stem cells but lacks chain migration, Nature, vol.427, pp.740-744, 2004.

F. T. Merkle, A. D. Tramontin, and J. M. García-verdugo, Radial glia give rise to adult neural stem cells in the subventricular zone, Proc Natl Acad Sci U S A, vol.101, pp.17528-17560, 2004.

M. Westphal and K. Lamszus, The neurobiology of gliomas: from cell biology to the development of therapeutic approaches, Nat Rev Neurosci, vol.12, pp.495-508, 2011.

R. Chen, M. C. Nishimura, and S. M. Bumbaca, A hierarchy of self-renewing tumorinitiating cell types in glioblastoma, Cancer Cell, vol.17, pp.362-75, 2010.

C. E. Griguer, C. R. Oliva, and E. Gobin, CD133 is a marker of bioenergetic stress in human glioma, PLoS One, vol.3, p.3655, 2008.

D. T. Scadden, The stem-cell niche as an entity of action, Nature, vol.441, pp.1075-1084, 2006.

S. K. Singh, C. Hawkins, and I. D. Clarke, Identification of human brain tumour initiating cells, Nature, vol.432, pp.396-401, 2004.

A. Sharma and A. Shiras, Cancer stem cell-vascular endothelial cell interactions in glioblastoma, Biochem Biophys Res Commun, vol.473, pp.688-92, 2016.

A. A. Thomas, C. W. Brennan, and L. M. Deangelis, Emerging therapies for glioblastoma, JAMA Neurol, vol.71, pp.1437-1481, 2014.

I. Paw, R. C. Carpenter, and K. Watabe, Mechanisms regulating glioma invasion, Cancer Lett, vol.362, pp.1-7, 2015.

Y. Lee, J. K. Lee, and S. H. Ahn, WNT signaling in glioblastoma and therapeutic opportunities, Lab Invest, vol.96, pp.137-50, 2016.

M. Weller, W. Wick, and K. Aldape, Nat Rev Dis Primers, vol.1, p.15017, 2015.

M. Weller, M. Van-den-bent, and J. C. Tonn, European Association for Neuro-Oncology (EANO) Task Force on Gliomas. European Association for Neuro-Oncology (EANO) guideline on the diagnosis and treatment of adult astrocytic and oligodendroglial gliomas, Lancet Oncol, vol.18, pp.315-344, 2017.

R. Stupp, M. E. Hegi, and W. P. Mason, European Organisation for Research and Treatment of Cancer Brain Tumour and Radiation Oncology Groups; National Cancer Institute of Canada Clinical Trials Group. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, Lancet Oncol, vol.10, pp.459-66, 2009.

F. Lefranc, J. Brotchi, and R. Kiss, Possible future issues in the treatment of glioblastomas: special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis, J Clin Oncol, vol.23, pp.2411-2433, 2005.

Z. C. Ye and H. Sontheimer, Glioma cells release excitotoxic concentrations of glutamate, Cancer Res, vol.59, pp.4383-91, 1999.

S. A. Lyons, W. J. Chung, and A. K. Weaver, Autocrine glutamate signaling promotes glioma cell invasion, Cancer Res, vol.67, pp.9463-71, 2007.

S. M. Robert, S. C. Buckingham, and S. L. Campbell, SLC7A11 expression is associated with seizures and predicts poor survival in patients with malignant glioma, Sci Transl Med, vol.7, pp.289-86, 2015.

T. Takano, J. H. Lin, and G. Arcuino, Glutamate release promotes growth of malignant gliomas, Nat Med, vol.7, pp.1010-1015, 2001.

Z. C. Ye, J. D. Rothstein, and H. Sontheimer, Compromised glutamate transport in human glioma cells: reduction-mislocalization of sodium-dependent glutamate transporters and enhanced activity of cystine-glutamate exchange, J Neurosci, vol.19, pp.10767-77, 1999.

W. J. Chung, S. A. Lyons, and G. M. Nelson, Inhibition of cystine uptake disrupts the growth of primary brain tumors, J Neurosci, vol.25, pp.7101-7111, 2005.

S. M. Robert and H. Sontheimer, Glutamate transporters in the biology of malignant gliomas, Cell Mol Life Sci, vol.71, pp.1839-54, 2014.

J. F. De-groot, T. J. Liu, and G. Fuller, The excitatory amino acid transporter-2 induces apoptosis and decreases glioma growth in vitro and in vivo, Cancer Res, vol.65, pp.1934-1974, 2005.

S. Ishiuchi, K. Tsuzuki, and Y. Yoshida, Blockage of Ca(2+)-permeable AMPA receptors suppresses migration and induces apoptosis in human glioblastoma cells, Nat Med, vol.8, pp.971-979, 2002.

D. G. Van-vuurden, M. Yazdani, and I. Bosma, Attenuated AMPA receptor expression allows glioblastoma cell survival in glutamate-rich environment, PLoS One, vol.4, p.5953, 2009.

Y. Piao, L. Lu, and J. De-groot, AMPA receptors promote perivascular glioma invasion via beta1 integrin-dependent adhesion to the extracellular matrix, Neuro Oncol, vol.11, pp.260-73, 2009.

S. M. Robert, T. Ogunrinu-babarinde, and K. T. Holt, Role of glutamate transporters in redox homeostasis of the brain, Neurochem Int, vol.73, pp.181-91, 2014.

C. R. Oliva, D. R. Moellering, and G. Y. Gillespie, Acquisition of chemoresistance in gliomas is associated with increased mitochondrial coupling and decreased ROS production, PLoS One, vol.6, p.24665, 2011.

A. Schwab, A. Fabian, and P. J. Hanley, Role of ion channels and transporters in cell migration, Physiol Rev, vol.92, pp.1865-913, 2012.

V. A. Cuddapah and H. Sontheimer, Ion channels and transporters [corrected] in cancer. 2. Ion channels and the control of cancer cell migration, Am J Physiol Cell Physiol, vol.301, pp.541-550, 2011.

V. A. Cuddapah, S. Robel, and S. Watkins, A neurocentric perspective on glioma invasion, Nat Rev Neurosci, vol.15, pp.455-65, 2014.

L. A. Pardo and W. Stühmer, The roles of K(+) channels in cancer, Nat Rev Cancer, vol.14, pp.39-48, 2014.

J. C. Hervé, Membrane channels and transporters in cancers, BBA, vol.1848, pp.2473-2479, 2015.

N. Comes, A. Serrano-albarrás, and J. Capera, Involvement of potassium channels in the progression of cancer to a more malignant phenotype, BBA, vol.1848, pp.2477-92, 2015.

S. Herrmann, M. Ninkovic, and T. Kohl, Cortactin controls surface expression of the voltage-gated potassium channel K(V)10.1, J Biol Chem, vol.287, pp.44151-63, 2012.

K. Preussat, C. Beetz, and M. Schrey, Expression of voltage-gated potassium channels Kv1.3 and Kv1.5 in human gliomas, Neurosci Lett, vol.346, pp.33-39, 2003.

F. Lefranc, H. B. Pouleau, and M. Rynkowski, Voltage-dependent K+ channels as oncotargets in malignant gliomas, Oncotarget, vol.3, pp.516-523, 2012.

S. Arvind, A. Arivazhagan, and V. Santosh, Differential expression of a novel voltage gated potassium channel-Kv 1.5 in astrocytomas and its impact on prognosis in glioblastoma, Br J Neurosurg, vol.26, pp.16-20, 2012.

Y. Bai, H. Liao, and T. Liu, MiR-296-3p regulates cell growth and multi-drug resistance of human glioblastoma by targeting ether-à-go-go (EAG1), Eur J Cancer, vol.49, pp.710-734, 2013.

L. A. Pardo and W. Stühmer, Eag1: an emerging oncological target, Cancer Res, vol.68, pp.1611-1614, 2008.

R. Martínez, W. Stühmer, and S. Martin, Analysis of the expression of Kv10.1 potassium channel in patients with brain metastases and glioblastoma multiforme: impact on survival, BMC Cancer, vol.15, p.839, 2015.

A. Masi, A. Becchetti, and R. Restano-cassulini, hERG1 channels are overexpressed in glioblastoma multiforme and modulate VEGF secretion in glioblastoma cell lines, Br J Cancer, vol.93, pp.781-92, 2005.

C. B. Ransom, X. Liu, and H. Sontheimer, BK channels in human glioma cells have enhanced calcium sensitivity, Glia, vol.38, pp.281-91, 2002.

A. K. Weaver, V. C. Bomben, and H. Sontheimer, Expression and function of calcium-activated potassium channels in human glioma cells, Glia, vol.54, pp.223-256, 2006.

P. Ruggieri, G. Mangino, and B. Fioretti, The inhibition of KCa3.1 channels activity reduces cell motility in glioblastoma derived cancer stem cells, PLoS One, vol.7, p.47825, 2012.

H. Lin, Z. Li, and C. Chen, Transcriptional and post-transcriptional mechanisms for oncogenic overexpression of ether à go-go K+ channel, PLoS One, vol.6, p.20362, 2011.

B. R. Downie, A. Sánchez, and H. Knötgen, Eag1 expression interferes with hypoxia homeostasis and induces angiogenesis in tumors, J Biol Chem, vol.283, pp.36234-36274, 2008.

C. Toral, M. E. Mendoza-garrido, and E. Azorín, Effect of extracellular matrix on adhesion, viability, actin cytoskeleton and K+ currents of cells expressing human ether à go-go channels, Life Sci, vol.81, pp.255-65, 2007.

L. Bianchi, B. Wible, and A. Arcangeli, herg encodes a K+ current highly conserved in tumors of different histogenesis: a selective advantage for cancer cells?, Cancer Res, vol.58, pp.815-837, 1998.

O. Crociani, L. Guasti, and M. Balzi, Cell cycle-dependent expression of HERG1 and HERG1B isoforms in tumor cells, J Biol Chem, vol.278, pp.2947-55, 2003.

A. Becchetti, S. Crescioli, and F. Zanieri, The conformational state of hERG1 channels determines integrin association, downstream signaling, and cancer progression, Sci Signal, vol.10, 2017.

I. Staudacher, J. Jehle, and K. Staudacher, HERG K+ channel-dependent apoptosis and cell cycle arrest in human glioblastoma cells, PLoS One, vol.9, p.88164, 2014.

A. Felipe, J. Bielanska, and N. Comes, Targeting the voltage-dependent K(+) channels Kv1.3 and Kv1.5 as tumor biomarkers for cancer detection and prevention, Curr Med Chem, vol.19, pp.661-74, 2012.

K. L. Turner, A. Honasoge, and S. M. Robert, A Proinvasive Role for the Ca2+-Activated K+ Channel KCa3.1 in Malignant Glioma, Glia, vol.62, pp.971-81, 2014.

G. D'alessandro, M. Catalano, and M. Sciaccaluga, 1 channels are involved in the infiltrative behavior of glioblastoma in vivo, Cell Death Disease, vol.4, p.773, 2013.

C. Wild-bode, M. Weller, and A. Rimner, Sublethal irradiation promotes migration and invasiveness of glioma cells: implications for radiotherapy of human glioblastoma, Cancer Res, vol.61, pp.2744-50, 2001.

M. Steinle, D. Palme, and M. Misovic, Ionizing radiation induces migration of glioblastoma cells by activating BK K channels, Radiother Oncol, vol.101, pp.122-128, 2011.

L. Soroceanu, T. J. Manning, and H. Sontheimer, Modulation of glioma cell migration and invasion using Cl(-) and K(+) ion channel blockers, J Neurosci, vol.19, pp.5942-54, 1999.

C. B. Ransom, J. T. O'neal, and H. Sontheimer, Volume-activated chloride currents contribute to the resting conductance and invasive migration of human glioma cells, J Neurosci, vol.21, pp.7674-83, 2001.

C. W. Habela, N. J. Ernest, and A. F. Swindall, Chloride accumulation drives volume dynamics underlying cell proliferation and migration, J Neurophysiol, vol.101, pp.750-757, 2009.

F. B. Morrone, M. P. Gehring, and N. F. Nicoletti, Calcium channels and associated receptors in malignant brain tumor therapy, Mol Pharmacol, vol.90, pp.403-412, 2016.

M. Osswald, E. Jung, and F. Sahm, Brain tumour cells interconnect to a functional and resistant network, Nature, vol.528, pp.93-101, 2015.

S. Lin and R. I. Gregory, MicroRNA biogenesis pathways in cancer, Nat Rev Cancer, vol.15, pp.321-354, 2015.

J. Fu, M. Rodova, and R. Nanta, NPV-LDE-225 (Erismodegib) inhibits epithelial mesenchymal transition and self-renewal of glioblastoma initiating cells by F. Lefranc et al, Cancer Treatment Reviews, vol.68, pp.145-154, 2018.

, regulating miR-21, miR-128, and miR-200, Neuro Oncol, vol.15, pp.691-706, 2013.

S. A. Ciafrè, S. Galardi, and A. Mangiola, Extensive modulation of a set of microRNAs in primary glioblastoma, Biochem Biophys Res Commun, vol.334, pp.1351-1359, 2005.

J. A. Chan, A. M. Krichevsky, and K. S. Kosik, MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells, Cancer Res, vol.65, pp.6029-6062, 2005.

J. Sana, M. Hajduch, and J. Michalek, MicroRNAs and glioblastoma: roles in core signalling pathways and potential clinical implications, J Cell Mol Med, vol.15, pp.1636-1680, 2011.

M. Karsy, E. Arslan, and F. Moy, Current progress on understanding MicroRNAs in glioblastoma multiforme, Genes Cancer, vol.3, pp.3-15, 2012.

H. G. Møller, A. P. Rasmussen, and H. H. Andersen, A systematic review of microRNA in glioblastoma multiforme: micro-modulators in the mesenchymal mode of migration and invasion, Mol Neurobiol, vol.47, pp.131-175, 2013.

P. M. Costa, A. L. Cardoso, and M. Mano, MicroRNAs in glioblastoma: role in pathogenesis and opportunities for targeted therapies, CNS Neurol Disord: Drug Targets, vol.14, pp.222-260, 2015.

D. Pal, D. Mukhopadhyay, and M. J. Ramaiah, Regulation of cell proliferation and migration by miR-203 via GAS41/miR-10b axis in human glioblastoma cells, PLoS One, vol.11, p.159092, 2016.

J. Godlewski, A. M. Krichevsky, and M. D. Johnson, Belonging to a network-microRNAs, extracellular vesicles, and the glioblastoma microenvironment, Neuro Oncol, vol.17, pp.652-62, 2015.

N. Stojcheva, G. Schechtmann, and S. Sass, MicroRNA-138 promotes acquired alkylator resistance in glioblastoma by targeting the Bcl-2-interacting mediator BIM, Oncotarget, vol.7, pp.12937-50, 2016.

F. Fanini and M. Fabbri, Cancer-derived exosomic microRNAs shape the immune system within the tumor microenvironment: state of the art, Semin Cell Dev Biol, vol.67, pp.23-31, 2017.

Y. Naito, Y. Yoshioka, and Y. Yamamoto, How cancer cells dictate their microenvironment: present roles of extracellular vesicles, Cell Mol Life Sci, vol.74, pp.697-713, 2017.

A. K. Rooj, M. Mineo, and J. Godlewski, MicroRNA and extracellular vesicles in glioblastoma -Small but powerful, Brain Tumor Pathol, vol.33, pp.77-88, 2016.

A. Bronisz, J. Godlewski, and E. A. Chiocca, Extracellular vesicles and MicroRNAs: their role in tumorigenicity and therapy for brain tumors, Cell Mol Neurobiol, vol.36, pp.361-76, 2016.

C. C. Li, S. A. Eaton, and P. E. Young, Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells, RNA Biol, vol.10, pp.1333-1377, 2013.

A. Hoshino, B. Costa-silva, and T. L. Shen, Tumour exosome integrins determine organotropic metastasis, Nature, vol.527, pp.329-364, 2015.

J. C. Akers, W. Hua, and H. Li, A cerebrospinal fluid microRNA signature as biomarker for glioblastoma, Oncotarget, vol.8, pp.68769-79, 2017.

W. T. Arscott, A. T. Tandle, and S. Zhao, Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration, Transl Oncol, vol.6, pp.638-686, 2013.

X. Li, C. Wu, and N. Chen, PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma, Oncotarget, vol.7, pp.33440-50, 2016.

H. F. Zhao, J. Wang, and W. Shao, Recent advances in the use of PI3K inhibitors for glioblastoma multiforme: current preclinical and clinical development, Mol Cancer, vol.16, p.100, 2017.

M. B. Mcferrin, K. L. Turner, and V. A. Cuddapah, Differential role of IK and BK potassium channels as mediators of intrinsic and extrinsic apoptotic cell death, Am J Physiol Cell Physiol, vol.303, pp.1070-1078, 2012.

L. Leanza, B. Henry, and N. Sassi, Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bak-independent death of cancer cells, EMBO Mol Med, vol.4, pp.577-93, 2012.

V. Checchetto, M. Azzolini, and R. Peruzzo, Mitochondrial potassium channels in cell death, Biochem Biophys Res Commun, issue.17, pp.31226-31229, 2017.

S. Bonnet, S. L. Archer, and J. Allalunis-turner, A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth, Cancer Cell, vol.11, pp.37-51, 2007.

E. D. Burg, C. V. Remillard, and J. X. Yuan, K+ channels in apoptosis, J Membr Biol, vol.209, pp.3-20, 2006.

S. Weil, M. Osswald, and G. Solecki, Tumor microtubes convey resistance to surgical lesions and chemotherapy in gliomas, Neuro Oncol, vol.19, pp.1316-1342, 2017.

J. De-groot and H. Sontheimer, Glutamate and the biology of gliomas, Glia, vol.59, pp.1181-1190, 2011.

S. Blecic, M. Rynkowski, D. Witte, and O. , Glutamate and malignant gliomas, from epilepsia to biological aggressiveness: therapeutic implications, Bull Cancer, vol.100, pp.829-864, 2013.

M. Hermisson and M. Weller, NF-kB-independent actions of sulfasalazine dissociate the CD95L-and Apo2L/TRAIL-dependent death signalling pathways in human malignant glioma cells, Cell Death Differ, vol.10, pp.1078-89, 2003.

P. A. Robe, D. H. Martin, and M. T. Nguyen-khac, Early termination of ISRCTN45828668, a phase 1/2 prospective, randomized study of sulfasalazine for the treatment of progressing malignant gliomas in adults, BMC Cancer, vol.9, p.372, 2009.

G. Huberfeld and C. J. Vecht, Seizures and gliomas-towards a single therapeutic approach, Nat Rev Neurol, vol.12, pp.204-220, 2016.

C. Happold, T. Gorlia, and O. Chinot, Does valproic acid improve survival in glioblastoma? A meta-analysis of randomized trials in newly diagnosed glioblastoma, J Clin Oncol, vol.34, pp.731-740, 2016.

F. M. Iwamoto, T. N. Kreisl, and L. Kim, Phase 2 trial of talampanel, a glutamate receptor inhibitor, for adults with recurrent malignant gliomas, Cancer, vol.116, pp.1776-82, 2010.

S. A. Grossman, X. Ye, and M. Chamberlain, Talampanel with standard radiation and temozolomide in patients with newly diagnosed glioblastoma: a multicenter phase II trial, J Clin Oncol, vol.27, pp.4155-61, 2009.

S. A. Grossman, X. Ye, and S. Piantadosi, Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States, Clin Cancer Res, vol.16, pp.2443-2452, 2010.

M. Weller, N. Butowski, and D. D. Tran, ACT IV trial investigators: Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): a randomised, double-blind, international phase 3 trial, Lancet Oncol, vol.18, pp.1373-85, 2017.

K. Yohay, B. Tyler, and K. D. Weaver, Efficacy of local polymer-based and systemic delivery of the anti-glutamatergic agents riluzole and memantine in rat glioma models, J Neurosurg, vol.120, pp.854-63, 2014.

H. S. Chen and S. A. Lipton, The chemical biology of clinically tolerated NMDA receptor antagonists, J Neurochem, vol.97, pp.1611-1637, 2006.

D. Catarzi, V. Colotta, and F. Varano, Competitive AMPA receptor antagonists, Med Res Rev, vol.27, pp.239-78, 2007.

J. A. Debin, J. E. Maggio, and G. R. Strichartz, Purification and characterization of chlorotoxin, a chloride channel ligand from the venom of the scorpion, Am J Physiol, vol.264, pp.361-370, 1993.

A. N. Mamelak, S. Rosenfeld, and R. Bucholz, Phase I single-dose study of intracavitary-administered iodine-131-TM-601 in adults with recurrent high-grade glioma, J Clin Oncol, vol.24, pp.3644-50, 2006.

A. N. Mamelak and D. B. Jacoby, Targeted delivery of antitumoral therapy to glioma and other malignancies with synthetic chlorotoxin (TM-601), Expert Opin Drug Deliv, vol.4, pp.175-86, 2007.

D. C. Hockaday, S. Shen, and J. Fiveash, Imaging glioma extent with 131I-TM-601, J Nucl Med, vol.46, pp.580-586, 2005.

O. Veiseh, F. M. Kievit, and C. Fang, Chlorotoxin bound magnetic nanovector tailored for cancer cell targeting, imaging, and siRNA delivery, Biomaterials, vol.31, pp.8032-8074, 2010.

R. Huang, W. Ke, and L. Han, Targeted delivery of chlorotoxin-modified DNAloaded nanoparticles to glioma via intravenous administration, Biomaterials, vol.32, pp.2399-406, 2011.

J. Deshane, C. C. Garner, and H. Sontheimer, Chlorotoxin inhibits glioma cell invasion via matrix metalloproteinase-2, J Biol Chem, vol.278, pp.4135-4179, 2003.

H. Wulff, N. A. Castle, and L. A. Pardo, Voltage-gated potassium channels as therapeutic targets, Nat Rev Drug Discov, vol.8, pp.982-1001, 2009.

J. García-quiroz and J. Camacho, Astemizole: an old anti-histamine as a new promising anti-cancer drug, Anticancer Agents Med Chem, vol.11, pp.307-321, 2011.

L. A. Pardo, D. Gómez-varela, and F. Major, Approaches targeting K(V)10.1 open a novel window for cancer diagnosis and therapy, Curr Med Chem, vol.19, pp.675-82, 2012.

J. Ma, L. N. Hou, and Z. X. Rong, Antidepressant desipramine leads to C6 glioma cell autophagy: implication for the adjuvant therapy of cancer, Anticancer Agents Med Chem, vol.13, pp.254-60, 2013.

S. H. Jeon, S. H. Kim, and Y. Kim, The tricyclic antidepressant imipramine induces autophagic cell death in U-87MG glioma cells, Biochem Biophys Res Commun, vol.413, pp.311-318, 2011.

T. T. Sales, F. F. Resende, and N. L. Chaves, Suppression of the Eag1 potassium channel sensitizes glioblastoma cells to injury caused by temozolomide, Oncol Lett, vol.12, pp.2581-2590, 2016.

S. Kalyaanamoorthy and K. H. Barakat, Development of Safe Drugs: The hERG Challenge, Med Res Rev, 2017.

S. Gottlieb, Antihistamine drug withdrawn by manufacturer, BMJ, vol.319, p.7, 1999.

J. Napp, L. A. Pardo, and F. Hartung, In vivo imaging of tumour xenografts with an antibody targeting the potassium channel Kv10.1, Eur Biophys J, vol.45, pp.721-754, 2016.

S. B. Ganapathi, M. Kester, and K. S. Elmslie, State-dependent block of HERG potassium channels by R-roscovitine: implications for cancer therapy, Am J Physiol Cell Physiol, vol.296, pp.701-711, 2009.

G. D'alessandro, A. Grimaldi, and G. Chece, 1 channel inhibition sensitizes malignant gliomas to temozolomide treatment, Oncotarget, vol.7, pp.30781-96, 2016.

L. Edalat, B. Stegen, and L. Klumpp, BK K+ channel blockade inhibits radiationinduced migration/brain infiltration of glioblastoma cells, Oncotarget, vol.7, pp.14259-78, 2016.

M. Bury, A. Girault, and V. Mégalizzi, Ophiobolin A induces paraptosis-like cell death in human glioblastoma cells by decreasing BKCa channel activity, Cell Death Dis, vol.4, p.561, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00848910