A. Omuro and L. M. Deangelis, Glioblastoma and other malignant gliomas: a clinical review, pp.1842-1850, 2013.

T. A. Ulrich, E. M. De-juan-pardo, and S. Kumar, The mechanical rigidity of the extracellular matrix regulates the structure, motility, and proliferation of glioma cells, Cancer Res, vol.69, pp.4167-74, 2009.

I. Fischer, J. P. Gagner, M. Law, E. W. Newcomb, and D. Zagzag, Angiogenesis in gliomas: biology and molecular pathophysiology, vol.15, pp.297-310, 2005.

N. A. Bush, S. M. Chang, and M. S. Berger, Current and future strategies for treatment of glioma, Neurosurg Rev, vol.40, pp.1-14, 2017.

F. B. Furnari, T. Fenton, R. M. Bachoo, A. Mukasa, J. M. Stommel et al., Malignant astrocytic glioma: genetics, biology, and paths to treatment, Genes Dev, vol.21, pp.2683-2710, 2007.

S. A. Grossman and J. F. Batara, Current management of glioblastoma multiforme, Semin Oncol, pp.635-644, 2004.

S. Tsigkos, S. Mariz, J. Llinares, L. Fregonese, S. Aarum et al., Establishing medical plausibility in the context of orphan medicines designation in the European Union, Orphanet J Rare Dis, vol.9, p.175, 2014.

J. Bianco, C. Bastiancich, A. Jankovski, A. Rieux, V. Preat et al., On glioblastoma and the search for a cure: where do we stand?, Cell Mol Life Sci, vol.74, pp.2451-2466, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076749

T. R. Jue and K. L. Mcdonald, The challenges associated with molecular targeted therapies for glioblastoma, J Neurooncol, vol.127, pp.427-434, 2016.

N. R. Parker, A. L. Hudson, P. Khong, J. F. Parkinson, T. Dwight et al., Intratumoral heterogeneity identified at the epigenetic, genetic and transcriptional level in glioblastoma, Sci Rep, vol.6, p.22477, 2016.

T. Yamahara, Y. Numa, T. Oishi, T. Kawaguchi, T. Seno et al., Morphological and flow cytometric analysis of cell infiltration in glioblastoma: a comparison of autopsy brain and neuroimaging, Brain Tumor Pathol, vol.27, pp.81-87, 2010.

S. M. Robbins and D. L. Senger, Assessing Mechanisms of Glioblastoma Invasion, pp.275-298, 2013.

P. G. Fisher and P. A. Buffler, Malignant gliomas in 2005: where to GO from here?, JAMA, vol.293, pp.615-617, 2005.

T. Denysenko, L. Gennero, M. A. Roos, A. Melcarne, C. Juenemann et al., Glioblastoma cancer stem cells: heterogeneity, microenvironment and related therapeutic strategies, Cell Biochem Funct, vol.28, pp.343-351, 2010.
DOI : 10.1002/cbf.1666

T. Smets, T. M. Lawson, C. Grandin, A. Jankovski, and C. Raftopoulos, Immediate post-operative MRI suggestive of the site and timing of glioblastoma recurrence after gross total resection: a retrospective longitudinal preliminary study, Eur Radiol, vol.23, pp.1467-1477, 2013.

M. C. Mabray, R. F. Barajas, and S. Cha, Modern brain tumor imaging, Brain Tumor Res Treat, vol.3, pp.8-23, 2015.
DOI : 10.14791/btrt.2015.3.1.8

URL : http://synapse.koreamed.org/Synapse/Data/PDFData/0212BTRT/btrt-3-8.pdf

V. Cuccarini, A. Erbetta, M. Farinotti, L. Cuppini, F. Ghielmetti et al., Advanced MRI may complement histological diagnosis of lower grade gliomas and help in predicting survival, J Neurooncol, vol.126, pp.279-88, 2016.

H. W. Kao, S. W. Chiang, H. W. Chung, F. Y. Tsai, and C. Y. Chen, Advanced MR imaging of gliomas: an update, Biomed Res Int, p.970586, 2013.

T. Kazda, M. Bulik, P. Pospisil, R. Lakomy, M. Smrcka et al., Advanced MRI increases the diagnostic accuracy of recurrent glioblastoma: Single institution thresholds and validation of MR spectroscopy and diffusion weighted MR imaging, Neuroimage Clin, vol.11, pp.316-321, 2016.

D. Treister, S. Kingston, K. E. Hoque, M. Law, and M. S. Shiroishi, Multimodal magnetic resonance imaging evaluation of primary brain tumors, Semin Oncol, pp.478-495, 2014.

R. Altieri, F. Zenga, M. M. Fontanella, F. Cofano, A. Agnoletti et al., Glioma Surgery: Technological Advances to Achieve a Maximal Safe Resection, vol.27, pp.297-302, 2015.

J. Buckner, P. Brown, B. O'neill, F. Meyer, C. Wetmore et al., Central nervous system tumors, Mayo Clin Proc, vol.82, pp.1271-1286, 2007.

R. Stupp, M. Brada, M. J. Van-den, J. C. Bent, G. Tonn et al., High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.25, issue.3, pp.93-101, 2014.
DOI : 10.1093/annonc/mdu050

URL : https://academic.oup.com/annonc/article-pdf/25/suppl_3/iii93/6674203/mdu050.pdf

P. D. Delgado-lopez and E. M. Corrales-garcia, Survival in glioblastoma: a review on the impact of treatment modalities, vol.18, pp.1062-1071, 2016.

M. Jansen, S. Yip, and D. N. Louis, Molecular pathology in adult neuro-oncology: an update on diagnostic, prognostic and predictive markers, Lancet Neurol, vol.9, pp.717-726, 2010.

K. L. Chaichana, M. J. Mcgirt, J. Frazier, F. Attenello, H. Guerrero-cazares et al., Relationship of glioblastoma multiforme to the lateral ventricles predicts survival following tumor resection, J Neurooncol, vol.89, pp.219-224, 2008.

K. L. Chaichana, L. Pinheiro, and H. Brem, Delivery of local therapeutics to the brain: working toward advancing treatment for malignant gliomas, Ther Deliv, vol.6, pp.353-369, 2015.

A. F. Hottinger, K. J. Abdullah, and R. Stupp, Current standard of care in glioblastoma therapy, 2017.

N. R. Smoll, K. Schaller, and O. P. Gautschi, Long-term survival of patients with glioblastoma multiforme (GBM), J Clin Neurosci, vol.20, pp.670-675, 2013.

J. G. Wolbers, Novel strategies in glioblastoma surgery aim at safe, supra-maximum resection in conjunction with local therapies, Chin J Cancer, vol.33, pp.8-15, 2014.

K. R. Yabroff, L. Harlan, C. Zeruto, J. Abrams, and B. Mann, Patterns of care and survival for patients with glioblastoma multiforme diagnosed during, Neuro Oncol, vol.14, pp.351-359, 2006.

W. Dandy, Removal of right cerebral hemisphere for certain tumors with hemiplegia: Preliminary report, J Amer Med Assoc, vol.90, pp.823-825, 1928.

J. Wright, J. Chugh, C. Wright, F. Alonso, A. Hdeib et al., Laser interstitial thermal therapy followed by minimal-access transsulcal resection for the treatment of large and difficult to access brain tumors, Neurosurg Focus, p.14, 2016.

S. L. Hervey-jumper and M. S. Berger, Maximizing safe resection of low-and high-grade glioma, J Neurooncol, vol.130, pp.269-282, 2016.

R. L. Yong and R. R. Lonser, Surgery for glioblastoma multiforme: striking a balance, World neurosurgery, vol.76, pp.528-530, 2011.

R. Young, A. Jamshidi, G. Davis, and J. Sherman, Current trends in the surgical management and treatment of adult glioblastoma, Ann Transl Med, vol.3, p.121, 2015.

M. Lacroix, D. Abi-said, D. R. Fourney, Z. L. Gokaslan, W. Shi et al., A multivariate analysis of 416 patients with glioblastoma multiforme: prognosis, extent of resection, and survival, J Neurosurg, vol.95, pp.190-198, 2001.

D. M. Patel, N. Agarwal, K. L. Tomei, D. R. Hansberry, and I. M. Goldstein, Optimal Timing of Whole-Brain Radiation Therapy Following Craniotomy for Cerebral Malignancies, World Neurosurg, vol.84, pp.412-419, 2015.

R. Mirimanoff, T. Gorlia, W. Mason, M. Van-den, R. Bent et al., Radiotherapy and temozolomide for newly diagnosed glioblastoma: recursive partitioning analysis of the EORTC 26981/22981-NCIC CE3 phase III randomized trial, J Clin Oncol, vol.24, pp.2563-2569, 2006.

D. Patel, N. Agarwal, K. Tomei, D. Hansberry, and I. Goldstein, Optimal timing of whole-brain radiation therapy following craniotomy for cerebral malignancies, World Neurosurg, vol.84, pp.412-419, 2015.

R. Stupp, M. Brada, M. Van-den, J. Bent, G. Tonn et al., High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.25, pp.93-101, 2014.

C. Corso and R. Bindra, Success and failures of combined modalities in glioblastoma multiforme: Old problems and new directions, Semin Radiat Oncol, vol.26, pp.281-298, 2016.

P. Ehrlich, Address in pathology on chemotherapeutics: Scientific principles, methods, and results, vol.2, pp.445-451, 1913.

E. Krumbhaar and H. Krunbhaar, The blood and bone barrow in yelloe cross gas (mustard gas) poisoning, J Med Res, vol.40, pp.497-508, 1919.

E. Krumbhaar, Rôle of the blood and bone marrow in certain forms of gas poisoning: I. Peripheral blood changes and their significance, J Amer Med Assoc, vol.72, pp.39-41, 1919.

J. Fenn and R. Udelsman, First use of intravenous chemotherapy cancer treatment: rectifying the record, J Am Coll Surg, vol.212, pp.413-417, 2011.

R. Thomas, L. Recht, and S. , Advances in the management of glioblastoma: the role of temozolomide and MGMT testing, Clin Pharmacol, vol.5, pp.1-9, 2013.

T. Connors, Anticancer drug development: The way forward, Oncologist, vol.1, pp.180-181, 1996.

G. F. Woodworth, G. P. Dunn, E. A. Nance, J. Hanes, and H. Brem, Emerging insights into barriers to effective brain tumor therapeutics, Front Oncol, vol.4, p.126, 2014.

O. Van-tellingen, B. Yetkin-arik, M. C. De-gooijer, P. Wesseling, T. Wurdinger et al., Overcoming the blood-brain tumor barrier for effective glioblastoma treatment, Drug Resist Updat, vol.19, pp.1-12, 2015.

P. Rubin, D. Gash, J. Hansen, D. Nelson, and J. Williams, Disruption of the blood-brain barrier as the primary effect of CNS irradiation, Radiother Oncol, pp.51-60, 1994.

E. Peereboom and . Neuwelt, Chemotherapy delivery issues in central nervous system malignancy: a reality check, J Clin Oncol, vol.25, pp.2295-2305, 2007.

M. Cohen, Y. Shen, P. Keegan, and R. Pazdur, FDA drug approval summary: bevacizumab (Avastin) as treatment of recurrent glioblastoma multiforme, Oncologist, vol.14, pp.1131-1138, 2009.

L. Ashby, K. Smith, and B. Stea, Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: a systematic literature review, World J Surg Oncol, vol.14, p.225, 2016.

M. Westphal, D. Hilt, E. Bortey, P. Delavault, R. Olivares et al., A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma, Neuro Oncol, vol.5, pp.79-88, 2003.

H. Bock, M. Puchner, F. Lohmann, M. Schütze, S. Koll et al., First-line treatment of malignant glioma with carmustine implants followed by concomitant radiochemotherapy: a multicenter experience, Neurosurg Rev, vol.33, pp.441-449, 2010.

H. Brem, S. Piantadosi, P. Burger, M. Walker, R. Selker et al., Placebo-controlled trial of safety and efficacy of intraoperative controlled delivery by biodegradable polymers of chemotherapy for recurrent gliomas. The Polymer-brain Tumor Treatment Group, Lancet, vol.345, pp.1008-1012, 1995.

M. Borner, W. Scheithauer, C. Twelves, J. Maroun, and H. Wilke, Answering patients' needs: oral alternatives to intravenous therapy, Oncologist, vol.6, pp.12-16, 2001.
DOI : 10.1634/theoncologist.6-2004-12

URL : http://theoncologist.alphamedpress.org/content/6/suppl_4/12.full.pdf

G. Liu, E. Franssen, M. Fitch, and E. Warner, Patient preferences for oral versus intravenous palliative chemotherapy, J Clin Oncol, vol.15, pp.110-115, 1997.

M. Brada, S. Stenning, R. Gabe, L. Thompson, D. Levy et al., Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma, J Clin Oncol, vol.28, pp.4601-4608, 2010.

J. Silber, M. Bobola, A. Blank, and M. Chamberlain, O(6)-methylguanine-DNA methyltransferase in glioma therapy: promise and problems, Biochim Biophys Acta, pp.71-82, 2012.

A. Davies, U. Weinberg, and Y. Palti, Tumor treating fields: a new frontier in cancer therapy, Ann N Y Acad Sci, pp.86-95, 2013.

S. Taillibert, E. Le-rhun, and M. Chamberlain, Tumor treating fields: a new standard treatment for glioblastoma?, Curr Opin Neurol, vol.28, pp.659-664, 2015.

E. Domingo-musibay and E. Galanis, What next for newly diagnosed glioblastoma?, Future Oncol, vol.11, pp.3273-3283, 2015.

R. Stupp, E. Wong, A. Kanner, D. Steinberg, H. Engelhard et al., NovoTTF-100A versus physician's choice chemotherapy in recurrent glioblastoma: a randomised phase III trial of a novel treatment modality, Eur J Cancer, vol.48, pp.2192-2202, 2012.

M. D. Prados, S. A. Byron, N. L. Tran, J. J. Phillips, A. M. Molinaro et al., Neur Oncol, vol.17, pp.1051-1063, 2015.

M. Brada, S. Stenning, R. Gabe, L. C. Thompson, D. Levy et al., Temozolomide versus procarbazine, lomustine, and vincristine in recurrent high-grade glioma, J Clin Oncol, vol.28, pp.4601-4608, 2010.

M. Weller, R. Stupp, G. Reifenberger, A. A. Brandes, M. J. Van-den et al., MGMT promoter methylation in malignant gliomas: ready for personalized medicine?, Nat Rev Neurol, vol.6, pp.39-51, 2010.

J. N. Sarkaria, G. J. Kitange, C. D. James, R. Plummer, H. Calvert et al., Mechanisms of chemoresistance to alkylating agents in malignant glioma, Clin Cancer Res, vol.14, pp.2900-2908, 2008.

F. Danhier, K. Messaoudi, L. Lemaire, J. P. Benoit, and F. Lagarce, Combined anti-Galectin-1 and anti-EGFR siRNA-loaded chitosan-lipid nanocapsules decrease temozolomide resistance in glioblastoma: in vivo evaluation, Int J Pharm, vol.481, pp.154-161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392431

J. J. Vredenburgh, A. Desjardins, J. E. Herndon, J. Marcello, D. A. Reardon et al., Bevacizumab plus irinotecan in recurrent glioblastoma multiforme, J Clin Oncol, vol.25, pp.4722-4729, 2007.
DOI : 10.1200/jco.2007.12.2440

H. S. Friedman, M. D. Prados, P. Y. Wen, T. Mikkelsen, D. Schiff et al., Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma, J Clin Oncol, vol.27, pp.4733-4740, 2009.

P. Fu, Y. S. He, Q. Huang, T. Ding, Y. C. Cen et al., Bevacizumab treatment for newly diagnosed glioblastoma: Systematic review and meta-analysis of clinical trials, Mol Clin Oncol, vol.4, pp.833-838, 2016.

A. Narayana, P. Kelly, J. Golfinos, E. Parker, G. Johnson et al., Antiangiogenic therapy using bevacizumab in recurrent high-grade glioma: impact on local control and patient survival, J Neurosurg, vol.110, pp.173-180, 2009.

D. R. Johnson, H. E. Leeper, and J. H. Uhm, Glioblastoma survival in the United States improved after Food and Drug Administration approval of bevacizumab: a population-based analysis, Cancer, vol.119, pp.3489-3495, 2013.

W. Wick, M. Weller, M. Van-den, R. Bent, and . Stupp, Bevacizumab and recurrent malignant gliomas: a European perspective, J Clin Oncol, vol.28, pp.188-189, 2010.

A. Desjardins, Neuro-oncology: What is the optimal use of bevacizumab in glioblastoma?, Nat Rev Neurol, vol.11, pp.429-430, 2015.

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

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

W. Wick, O. Chinot, M. Bendszus, W. Mason, R. Henriksson et al., Evaluation of pseudoprogression rates and tumor progression patterns in a phase III trial of bevacizumab plus radiotherapy/temozolomide for newly diagnosed glioblastoma, Neuro Oncol, vol.18, pp.1434-1441, 2016.

O. Chinot, R. Nishikawa, W. Mason, R. Henriksson, F. Saran et al., Upfront bevacizumab may extend survival for glioblastoma patients who do not receive secondline therapy: an exploratory analysis of AVAglio, Neuro Oncol, vol.18, pp.1313-1318, 2016.

S. Rose, FDA pulls approval for Avastin in breast cancer, Cancer Discov, vol.1, pp.1-2, 2011.

B. Kovic and F. Xie, Economic evaluation of bevacizumab for the first-line treatment of newly diagnosed glioblastoma multiforme, J Clin Oncol, vol.33, pp.2296-2302, 2015.

B. Campos, L. Olsen, T. Urup, and H. Poulsen, A comprehensive profile of recurrent glioblastoma, vol.35, pp.5819-5825, 2016.

H. Sun, S. Du, G. Liao, X. Xie, C. Ren et al., Do glioma patients derive any therapeutic benefit from taking a higher cumulative dose of temozolomide regimens?: a meta-analysis, Medicine, vol.94, p.827, 2015.

A. Tosoni, E. Franceschi, R. Poggi, and A. Brandes, Relapsed glioblastoma: treatment strategies for initial and subsequent recurrences, Curr Treat Options Oncol, vol.17, p.49, 2016.

A. Yin, J. Cheng, X. Zhang, and B. Liu, The treatment of glioblastomas: a systematic update on clinical Phase III trials, Crit Rev Oncol Hematol, vol.87, pp.265-282, 2013.

E. C. Holland, Glioblastoma multiforme: the terminator, Proc Natl Acad Sci U S A, vol.97, pp.6242-6244, 2000.

L. Tavano and R. Muzzalupo, Multi-functional vesicles for cancer therapy: The ultimate magic bullet, Colloids Surf B Biointerfaces, pp.161-171, 2016.

G. Brada?, U. Büll, R. Fahlbusch, T. Grumme, E. Kazner et al., Computed Tomography in Intracranial Tumors: Differential Diagnosis and Clinical Aspects, 1982.

R. Stupp, M. Hegi, W. Mason, M. Van-den, M. Bent et al.,

T. Eisenhauer, M. Gorlia, D. Weller, J. Lacombe, R. Cairncross et al., Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, Lancet Oncol, vol.10, pp.459-466, 2009.

J. Scott, L. Bauchet, T. Fraum, L. Nayak, A. Cooper et al., Recursive partitioning analysis of prognostic factors for glioblastoma patients aged 70 years or older, Cancer, vol.118, pp.5595-5600, 2012.

M. Neagu and D. Reardon, An update on the role of immunotherapy and vaccine strategies for primary brain tumors, Curr Treat Options Oncol, vol.16, p.54, 2015.

J. Sampson and D. Mitchell, Vaccination strategies for neuro-oncology, Neuro Oncol, vol.17, pp.15-25, 2015.
DOI : 10.1093/neuonc/nov159

URL : https://academic.oup.com/neuro-oncology/article-pdf/17/suppl_7/vii15/7498347/nov159.pdf

H. Ardon, S. Van-gool, I. Lopes, W. Maes, R. Sciot et al., Integration of autologous dendritic cell-based immunotherapy in the primary treatment for patients with newly diagnosed glioblastoma multiforme: a pilot study, J Neurooncol, pp.261-272, 2010.

V. Schijns, C. Pretto, L. Devillers, D. Pierre, F. Hofman et al., First clinical results of a personalized immunotherapeutic vaccine against recurrent, incompletely resected, treatment-resistant glioblastoma multiforme (GBM) tumors, based on combined allo-and auto-immune tumor reactivity, Vaccine, pp.269-2696, 2015.

J. Reijneveld, E. Voest, and M. Taphoorn, Angiogenesis in malignant primary and metastatic brain tumors, J Neurol, vol.247, pp.597-608, 2000.
DOI : 10.1007/s004150070128

C. Scaringi, R. Enrici, and G. Minniti, Combining molecular targeted agents with radiation therapy for malignant gliomas, Onco Targets Ther, vol.6, pp.1079-1095, 2013.

M. Mcgee, J. Hamner, R. Williams, S. Rosati, T. Sims et al., Improved intratumoral oxygenation through vascular normalization increases glioma sensitivity to ionizing radiation, Int J Radiat Oncol Biol Phys, vol.76, pp.1537-1545, 2010.

V. Gupta, N. Jaskowiak, M. Beckett, H. Mauceri, J. Grunstein et al., Vascular endothelial growth factor enhances endothelial cell survival and tumor radioresistance, Cancer J, vol.8, pp.47-54, 2002.

E. Bolderson, D. Richard, B. Zhou, and K. Khanna, Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair, Clin Cancer Res, vol.15, pp.6314-6320, 2009.

M. Rouleau, A. Patel, M. Hendzel, S. Kaufmann, and G. Poirier, PARP inhibition: PARP1 and beyond, vol.10, pp.293-301, 2010.
DOI : 10.1038/nrc2812

URL : http://europepmc.org/articles/pmc2910902?pdf=render

H. Bryant, N. Schultz, H. Thomas, K. Parker, D. Flower et al., Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase, Nature, vol.434, pp.913-917, 2005.

H. Farmer, N. Mccabe, C. Lord, A. Tutt, D. Johnson et al., Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy, Nature, vol.434, pp.917-921, 2005.

C. Powell, C. Mikropoulos, S. Kaye, C. Nutting, S. Bhide et al., Pre-clinical and clinical evaluation of PARP inhibitors as tumour-specific radiosensitisers, Cancer Treat Rev, vol.36, pp.566-575, 2010.

J. Senra, B. Telfer, K. Cherry, C. Mccrudden, D. Hirst et al., Inhibition of PARP-1 by olaparib (AZD2281) increases the radiosensitivity of a lung tumor xenograft, Mol Cancer Ther, vol.10, pp.1949-1958, 2011.

D. Bonnet and J. Dick, Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell, Nature Med, vol.3, pp.730-737, 1997.

R. Galli, E. Binda, U. Orfanelli, B. Cipelletti, A. Gritti et al., Isolation and characterization of tumorigenic, stem-like neural precursors from human glioblastoma, Cancer Res, vol.64, pp.7011-7021, 2004.

D. Beier, S. Röhrl, D. Pillai, S. Schwarz, L. Kunz-schughart et al., Temozolomide preferentially depletes cancer stem cells in glioblastoma, Cancer Res, vol.68, pp.5706-5715, 2008.

M. Hegi, A. Diserens, T. Gorlia, M. Hamou, N. Tribolet et al., MGMT gene silencing and benefit from temozolomide in glioblastoma, N Engl J Med, vol.352, pp.997-1003, 2005.

J. Lathia, S. Mack, E. Mulkearns-hubert, C. Valentim, and J. Rich, Cancer stem cells in glioblastoma, Genes Dev, vol.29, pp.1203-1217, 2015.

P. Gupta, T. Onder, G. Jiang, K. Tao, C. Kuperwasser et al., Identification of selective inhibitors of cancer stem cells by high-throughput screening, Cell, vol.138, pp.645-659, 2009.

L. Qin, P. Jia, Z. Zhang, and S. Zhang, ROS-p53-cyclophilin-D signaling mediates salinomycin-induced glioma cell necrosis, J Exp Clin Canc Res, vol.34, p.57, 2015.
DOI : 10.1186/s13046-015-0174-1

URL : https://jeccr.biomedcentral.com/track/pdf/10.1186/s13046-015-0174-1

A. Calzolari, E. Saulle, M. De-angelis, L. Pasquini, A. Boe et al., Salinomycin potentiates the cytotoxic effects of TRAIL on glioblastoma cell lines, PLoS One, vol.9, p.94438, 2014.

W. Pardridge, Blood-brain barrier delivery, Drug Discov Today, vol.12, pp.54-61, 2007.

M. Vogelbaum and M. Aghi, Convection-enhanced delivery for the treatment of glioblastoma, Neuro Oncol, vol.17, pp.3-8, 2015.

K. Hynynen, N. Mcdannold, N. Vykhodtseva, and F. Jolesz, Non-invasive opening of BBB by focused ultrasound, Acta Neurochir Suppl, vol.86, pp.555-558, 2003.
DOI : 10.1007/978-3-7091-0651-8_113

J. Choi, J. Feshitan, B. Baseri, S. Wang, Y. Tung et al., Microbubble-size dependence of focused ultrasound-induced blood-brain barrier opening in mice in vivo, IEEE Trans Biomed Eng, vol.57, pp.145-154, 2010.

H. Liu, P. Hsu, C. Lin, C. Huang, W. Chai et al., Focused ultrasound enhances central nervous system delivery of bevacizumab for malignant glioma treatment, Radiology, pp.99-108, 2016.

A. Carpentier, M. Canney, A. Vignot, V. Reina, K. Beccaria et al., Clinical trial of blood-brain barrier disruption by pulsed ultrasound, Sci Transl Med, vol.8, pp.343-342, 2016.

G. Sarkar, G. Curran, J. Sarkaria, V. Lowe, and R. Jenkins, Peptide carrier-mediated non-covalent delivery of unmodified cisplatin, methotrexate and other agents via intravenous route to the brain, PLoS One, vol.9, p.97655, 2014.

T. Allen and P. Cullis, Drug delivery systems: entering the mainstream, Science, pp.1818-1822, 2004.
DOI : 10.1126/science.1095833

D. Peer, J. Karp, S. Hong, O. Farokhzad, R. Margalit et al., Nanocarriers as an emerging platform for cancer therapy, Nat Nanotech, vol.2, pp.751-760, 2007.
DOI : 10.1038/nnano.2007.387

A. Gutkin, Z. Cohen, and D. Peer, Harnessing nanomedicine for therapeutic intervention in glioblastoma, Expert Opin Drug Deliv, vol.13, pp.1573-1582, 2016.
DOI : 10.1080/17425247.2016.1200557

S. Kim, J. Harford, K. Pirollo, and E. Chang, Effective treatment of glioblastoma requires crossing the bloodbrain barrier and targeting tumors including cancer stem cells: The promise of nanomedicine, Biochem Biophys Res Commun, vol.468, pp.485-489, 2015.

F. Danhier, K. Messaoudi, L. Lemaire, J. Benoit, and F. Lagarce, Combined anti-Galectin-1 and anti-EGFR siRNA-loaded chitosan-lipid nanocapsules decrease temozolomide resistance in glioblastoma: in vivo evaluation, Int J Pharm, vol.481, pp.154-161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392431

C. Bastiancich, K. Vanvarenberg, B. Ucakar, M. Pitorre, G. Bastiat et al., Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of glioblastoma, J Control Release, vol.225, pp.283-293, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388804

T. Hoare and K. Ds, Hydrogels in drug delivery: Progress and challenges, vol.49, pp.1993-2007, 2008.

C. Bastiancich, P. Danhier, V. Préat, and F. Danhier, Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma, J Control Release, vol.243, pp.29-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02076746

A. Vellimana, V. Recinos, L. Hwang, K. Fowers, K. Li et al., Combination of paclitaxel thermal gel depot with temozolomide and radiotherapy significantly prolongs survival in an experimental rodent glioma model, J Neurooncol, vol.111, pp.229-236, 2013.

T. Fourniols, L. Randolph, A. Staub, K. Vanvarenberg, J. Leprince et al., Temozolomide-loaded photopolymerizable PEG-DMA-based hydrogel for the treatment of glioblastoma, J Control Release, vol.210, pp.95-104, 2015.

F. Marchesi, M. Turriziani, G. Tortorelli, G. Avvisati, F. Torino et al., Triazene compounds: mechanism of action and related DNA repair systems, Pharmacol Res, vol.56, pp.275-287, 2007.

G. Perazzoli, J. Prados, R. Ortiz, O. Caba, L. Cabeza et al., Temozolomide resistance in glioblastoma cell lines: Implication of MGMT, MMR, P-glycoprotein and CD133 expression, PLoS One, vol.10, p.140131, 2015.

M. Weller, R. Stupp, G. Reifenberger, A. Brandes, M. Van-den et al., MGMT promoter methylation in malignant gliomas: ready for personalized medicine?, Nat Rev Neurol, vol.6, pp.39-51, 2010.

N. Parker, P. Khong, J. Parkinson, V. Howell, and H. Wheeler, Molecular heterogeneity in glioblastoma: potential clinical implications, Front Oncol, vol.5, p.55, 2015.

K. Messaoudi, C. Clavreul, F. Danhier, P. Saulnier, J. Benoît et al., Combined silencing expression of MGMT with EGFR or galectin-1 enhances the sensitivity of glioblastoma to temozolomide, Eur J Nanomed, vol.7, pp.97-107, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01153701

B. Kaina, G. Margison, and M. Christmann, Targeting O?-methylguanine-DNA methyltransferase with specific inhibitors as a strategy in cancer therapy, Cell Mol Life Sci, vol.67, pp.3663-3681, 2010.

Y. Ramirez, A. Mladek, R. Phillips, M. Gynther, J. Rautio et al., Evaluation of novel imidazotetrazine analogues designed to overcome temozolomide resistance and glioblastoma regrowth, Mol Cancer Ther, vol.14, pp.111-119, 2015.

J. Rich and D. Bigner, Development of novel targeted therapies in the treatment of malignant glioma, Nat Rev Drug Discov, vol.3, pp.430-446, 2004.

T. Verschuere, J. Toelen, W. Maes, F. Poirier, L. Boon et al., Glioma-derived galectin-1 regulates innate and adaptive antitumor immunity, Int J Cancer, vol.134, pp.873-884, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00951339

M. L. Mercier, S. Fortin, V. Mathieu, R. Kiss, and F. Lefranc, Galectins and gliomas, vol.20, pp.17-27, 2010.

I. Camby, C. Decaestecker, F. Lefranc, H. Kaltner, H. Gabius et al., Galectin-1 knocking down in human U87 glioblastoma cells alters their gene expression pattern, Biochem Biophys Res Commun, vol.335, pp.27-35, 2005.

A. Burgess, K. Shah, O. Hough, and K. Hynynen, Focused ultrasound-mediated drug delivery through the blood-brain barrier, Expert Rev Neurother, vol.15, pp.477-491, 2015.

R. Karim, C. Palazzo, B. Evrard, and G. Piel, Nanocarriers for the treatment of glioblastoma multiforme: Current state-of-the-art, J Control Release, vol.227, pp.23-37, 2016.

F. Danhier, O. Feron, and V. Preat, To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery, J Control Release, vol.148, pp.135-146, 2010.

M. Singh, S. Kundu, M. A. Reddy, V. Sreekanth, R. K. Motiani et al., Injectable small molecule hydrogel as a potential nanocarrier for localized and sustained in vivo delivery of doxorubicin, Nanoscale, vol.6, pp.12849-12855, 2014.

M. Westphal, D. C. Hilt, E. Bortey, P. Delavault, R. Olivares et al., A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma, Neuro Oncol, vol.5, pp.79-88, 2003.

B. K. Hendricks, A. A. Cohen-gadol, and J. C. Miller, Novel delivery methods bypassing the blood-brain and blood-tumor barriers, Neurosurgical focus, vol.38, p.10, 2015.

T. Garg, S. Bhandari, G. Rath, and A. K. Goyal, Current strategies for targeted delivery of bio-active drug molecules in the treatment of brain tumor, J Drug Targ, vol.23, pp.865-887, 2015.

V. Varenika, P. Dickinson, J. Bringas, R. Lecouteur, R. Higgins et al., Detection of infusate leakage in the brain using real-time imaging of convection-enhanced delivery, J Neurosurg, vol.109, pp.874-880, 2008.

J. B. Wolinsky, Y. L. Colson, and M. W. Grinstaff, Local drug delivery strategies for cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers, J Control Release, vol.159, pp.14-26, 2012.

T. A. Juratli, G. Schackert, and D. Krex, Current status of local therapy in malignant gliomas--a clinical review of three selected approaches, Pharmacol Ther, vol.139, pp.341-358, 2013.

K. G. Abdullah and J. Burdick, Local Drug Delivery in the Treatment of Glioblastoma, 2017.

D. A. Bota, A. Desjardins, J. A. Quinn, M. L. Affronti, and H. S. Friedman, Interstitial chemotherapy with biodegradable BCNU (Gliadel) wafers in the treatment of malignant gliomas, Ther Clin Risk Manag, vol.3, pp.707-715, 2007.

, Food and Drug administration webpage, 2013.

J. Perry, A. Chambers, K. Spithoff, and N. Laperriere, Gliadel wafers in the treatment of malignant glioma: a systematic review, Curr Oncol, vol.14, pp.189-194, 2007.

S. A. Grossman, C. Reinhard, O. M. Colvin, M. Chasin, R. Brundrett et al., The intracerebral distribution of BCNU delivered by surgically implanted biodegradable polymers, J Neurosurg, vol.76, pp.640-647, 1992.

L. K. Fung, M. G. Ewend, A. Sills, E. P. Sipos, R. Thompson et al., Pharmacokinetics of interstitial delivery of carmustine, 4-hydroperoxycyclophosphamide, and paclitaxel from a biodegradable polymer implant in the monkey brain, Cancer Res, vol.58, pp.672-684, 1998.

D. F. Emerich, S. R. Winn, P. Snodgrass, D. Lafreniere, M. Agostino et al., Injectable chemotherapeutic microspheres and glioma II: enhanced survival following implantation into deep inoperable tumors, Pharm Res, vol.17, pp.776-781, 2000.

Y. Y. Tseng, Y. C. Wang, C. H. Su, T. C. Yang, T. M. Chang et al., Concurrent delivery of carmustine, irinotecan, and cisplatin to the cerebral cavity using biodegradable nanofibers: In vitro and in vivo studies, Colloids Surf B Biointerfaces, pp.254-261, 2015.

J. A. Floyd, A. Galperin, and B. D. Ratner, Drug encapsulated polymeric microspheres for intracranial tumor therapy: A review of the literature, Adv Drug Deliv Rev, vol.91, pp.23-37, 2015.

P. Menei, M. Boisdron-celle, A. Croue, G. Guy, and J. P. Benoit, Effect of stereotactic implantation of biodegradable 5-fluorouracil-loaded microspheres in healthy and C6 glioma-bearing rats, Neurosurgery, vol.39, pp.123-114, 1996.

L. Lemaire, V. G. Roullin, F. Franconi, M. C. Venier-julienne, P. Menei et al., Therapeutic efficacy of 5-fluorouracil-loaded microspheres on rat glioma: a magnetic resonance imaging study, vol.14, pp.360-366, 2001.

D. F. Emerich, S. R. Winn, Y. Hu, J. Marsh, P. Snodgrass et al., Injectable chemotherapeutic microspheres and glioma I: enhanced survival following implantation into the cavity wall of debulked tumors, Pharm Res, vol.17, pp.767-775, 2000.

P. Menei, E. Jadaud, N. Faisant, M. Boisdron-celle, S. Michalak et al., Stereotaxic implantation of 5-fluorouracil-releasing microspheres in malignant glioma, Cancer, vol.100, pp.405-410, 2004.

Y. H. Zhang, H. Zhang, J. M. Liu, and Z. J. Yue, Temozolomide/PLGA microparticles: a new protocol for treatment of glioma in rats, Med oncol, vol.28, pp.901-906, 2011.

X. Zhang, S. Yao, C. Liu, and Y. Jiang, Tumor tropic delivery of doxorubicin-polymer conjugates using mesenchymal stem cells for glioma therapy, Biomaterials, pp.269-281, 2015.

Y. Y. Tseng, Y. C. Kau, and S. J. Liu, Advanced interstitial chemotherapy for treating malignant glioma, Expert Opin Drug Deliv, vol.13, pp.1533-1544, 2016.

A. Mangraviti, D. Gullotti, B. Tyler, and H. Brem, Nanobiotechnology-based delivery strategies: New frontiers in brain tumor targeted therapies, J Control Release, vol.240, pp.443-453, 2016.

A. Mangraviti, B. Tyler, and H. Brem, Interstitial chemotherapy for malignant glioma: Future prospects in the era of multimodal therapy, Surg Neurol Int, vol.6, pp.78-84, 2015.

G. Orive, O. A. Ali, E. Anitua, J. L. Pedraz, and D. F. Emerich, Biomaterial-based technologies for brain anticancer therapeutics and imaging, Biochim Biophys Acta, pp.96-107, 2010.

N. A. Peppas, P. Bures, W. Leobandung, and H. Ichikawa, Hydrogels in pharmaceutical formulations, Eur J Pharm Biopharm, vol.50, pp.27-46, 2000.

N. A. Peppas and R. Langer, New challenges in biomaterials, vol.263, pp.1715-1720, 1994.
DOI : 10.1126/science.8134835

B. D. Ratner and A. S. , Synthetic hydrogels for biomedical applications, Hydrogels for Medical and Related Applications, pp.1-36, 1976.

C. C. Lin and A. T. Metters, Hydrogels in controlled release formulations: network design and mathematical modeling, Adv Drug Deliv Rev, vol.58, pp.1379-1408, 2006.
DOI : 10.1016/j.addr.2006.09.004

S. R. Van-tomme, G. Storm, and W. E. Hennink, In situ gelling hydrogels for pharmaceutical and biomedical applications, Int J Pharm, vol.355, pp.1-18, 2008.

C. Wang, X. Tong, and F. Yang, Bioengineered 3D brain tumor model to elucidate the effects of matrix stiffness on glioblastoma cell behavior using PEG-based hydrogels, Mol Pharm, vol.11, pp.2115-2125, 2014.

S. S. Rao, J. Dejesus, A. R. Short, J. J. Otero, A. Sarkar et al., Glioblastoma behaviors in threedimensional collagen-hyaluronan composite hydrogels, vol.5, pp.9276-9284, 2013.
DOI : 10.1021/am402097j

URL : http://europepmc.org/articles/pmc4333346?pdf=render

C. Jiglaire, N. Baeza-kallee, E. Denicolai, D. Barets, P. Metellus et al., Ex vivo cultures of glioblastoma in three-dimensional hydrogel maintain the original tumor growth behavior and are suitable for preclinical drug and radiation sensitivity screening, Exp Cell Res, vol.321, pp.99-108, 2014.

U. Akbar, T. Jones, J. Winestone, M. Michael, A. Shukla et al., Delivery of temozolomide to the tumor bed via biodegradable gel matrices in a novel model of intracranial glioma with resection, J Neurooncol, vol.94, pp.203-212, 2009.

A. K. Vellimana, V. R. Recinos, L. Hwang, K. D. Fowers, K. W. Li et al., Combination of paclitaxel thermal gel depot with temozolomide and radiotherapy significantly prolongs survival in an experimental rodent glioma model, J Neurooncol, vol.111, pp.229-236, 2013.

B. Tyler, K. D. Fowers, K. W. Li, V. R. Recinos, J. M. Caplan et al., A thermal gel depot for local delivery of paclitaxel to treat experimental brain tumors in rats, J Neurosurg, vol.113, pp.210-217, 2010.

C. V. Rahman, S. J. Smith, P. S. Morgan, K. A. Langmack, P. A. Clarke et al., Adjuvant chemotherapy for brain tumors delivered via a novel intra-cavity moldable polymer matrix, PloS one, vol.8, p.77435, 2013.
DOI : 10.1371/journal.pone.0077435

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0077435&type=printable

T. Fourniols, L. D. Randolph, A. Staub, K. Vanvarenberg, J. G. Leprince et al., Temozolomide-loaded photopolymerizable PEG-DMA-based hydrogel for the treatment of glioblastoma, J Control Release, vol.210, pp.95-104, 2015.

J. I. Kim, B. Kim, C. Chun, S. H. Lee, and S. C. Song, MRI-monitored long-term therapeutic hydrogel system for brain tumors without surgical resection, Biomaterials, pp.4836-4842, 2012.
DOI : 10.1016/j.biomaterials.2012.03.048

T. Arai, T. Joki, M. Akiyama, M. Agawa, Y. Mori et al., Novel drug delivery system using thermoreversible gelation polymer for malignant glioma, J Neurooncol, vol.77, pp.9-15, 2006.
DOI : 10.1007/s11060-005-9001-4

T. Arai, O. Benny, T. Joki, L. G. Menon, M. Machluf et al., Novel local drug delivery system using thermoreversible gel in combination with polymeric microspheres or liposomes, vol.30, pp.1057-1064, 2010.

T. Ozeki, D. Kaneko, K. Hashizawa, Y. Imai, T. Tagami et al., Combination therapy of surgical tumor resection with implantation of a hydrogel containing camptothecin-loaded poly(lactic-co-glycolic acid) microspheres in a C6 rat glioma model, Biol Pharm Bull, vol.35, pp.545-550, 2012.

T. Ozeki, K. Hashizawa, D. Kaneko, Y. Imai, and H. Okada, Treatment of rat brain tumors using sustainedrelease of camptothecin from poly(lactic-co-glycolic acid) microspheres in a thermoreversible hydrogel, Chem Pharm Bull, vol.58, pp.1142-1147, 2010.

S. Vinchon-petit, D. Jarnet, S. Michalak, A. Lewis, J. P. Benoit et al., Local implantation of doxorubicin drug eluting beads in rat glioma, Int J Pharm, vol.402, pp.184-189, 2010.

S. H. Ranganath, I. Kee, W. B. Krantz, P. K. Chow, and C. H. Wang, Hydrogel matrix entrapping PLGA-paclitaxel microspheres: drug delivery with near zero-order release and implantability advantages for malignant brain tumour chemotherapy, Pharm Res, vol.26, pp.2101-2114, 2009.

S. H. Ranganath, Y. Fu, D. Y. Arifin, I. Kee, L. Zheng et al., The use of submicron/nanoscale PLGA implants to deliver paclitaxel with enhanced pharmacokinetics and therapeutic efficacy in intracranial glioblastoma in mice, Biomaterials, pp.5199-5207, 2010.

N. Qi, C. Cai, W. Zhang, Y. Niu, J. Yang et al., Sustained delivery of cytarabine-loaded vesicular phospholipid gels for treatment of xenografted glioma, Int J Pharm, vol.472, pp.48-55, 2014.

T. Chen, T. Gong, T. Zhao, X. Liu, Y. Fu et al., Paclitaxel loaded phospholipid-based gel as a drug delivery system for local treatment of glioma, Int J Pharm, vol.528, pp.127-132, 2017.

S. Kim, S. K. Nishimoto, J. D. Bumgardner, W. O. Haggard, M. W. Gaber et al., A chitosan/betaglycerophosphate thermo-sensitive gel for the delivery of ellagic acid for the treatment of brain cancer, Biomaterials, pp.4157-4166, 2010.

C. T. Tsao, F. M. Kievit, A. Ravanpay, A. E. Erickson, M. C. Jensen et al., Thermoreversible poly(ethylene glycol)-g-chitosan hydrogel as a therapeutic T lymphocyte depot for localized glioblastoma immunotherapy, Biomacromolecules, vol.15, pp.2656-2662, 2014.

J. R. Tauro and R. A. Gemeinhart, Matrix metalloprotease triggered delivery of cancer chemotherapeutics from hydrogel matrixes, Bioconj Chem, vol.16, pp.1133-1139, 2005.

S. A. Meenach, J. M. Shapiro, J. Z. Hilt, and K. W. Anderson, Characterization of PEG-iron oxide hydrogel nanocomposites for dual hyperthermia and paclitaxel delivery, J Biomat Sci. Polymer edition, pp.1112-1126, 2013.

Y. Xu, M. Shen, Y. Sun, P. Gao, and Y. Duan, Polymer Nanocomposites Based Thermo-Sensitive Gel for Paclitaxel and Temozolomide Co-Delivery to Glioblastoma Cells, J Nanosci Nanotech, vol.15, pp.9777-9787, 2015.
DOI : 10.1166/jnn.2015.12338

F. Danhier, E. Ansorena, J. M. Silva, R. Coco, A. L. Breton et al., PLGA-based nanoparticles: an overview of biomedical applications, J Control Release, pp.505-522, 2012.

A. Alexander, J. Ajazuddin, S. Khan, S. Saraf, and . Saraf, Poly(ethylene glycol)-poly(lactic-co-glycolic acid) based thermosensitive injectable hydrogels for biomedical applications, J Control Release, vol.172, pp.715-729, 2013.

N. L. Elstad and K. D. Fowers, OncoGel (ReGel/paclitaxel)--clinical applications for a novel paclitaxel delivery system, Adv Drug Deliv Rev, pp.785-794, 2009.
DOI : 10.1016/j.addr.2009.04.010

B. Baroli, Photopolymerization of biomaterials: issues and potentialities in drug delivery, tissue engineering, and cell encapsulation applications, J Chem Tech Biotech, vol.81, pp.491-499, 2006.

K. T. Nguyen and J. L. West, Photopolymerizable hydrogels for tissue engineering applications, Biomaterials, vol.23, pp.4307-4314, 2002.
DOI : 10.1016/s0142-9612(02)00175-8

N. Schleich, F. Danhier, and V. Preat, Iron oxide-loaded nanotheranostics: major obstacles to in vivo studies and clinical translation, J Control Release, vol.198, pp.35-54, 2015.

L. Sun, D. Y. Joh, A. Al-zaki, M. Stangl, S. Murty et al., Theranostic Application of Mixed Gold and Superparamagnetic Iron Oxide Nanoparticle Micelles in Glioblastoma Multiforme, J Biomed Nanotech, vol.12, pp.347-356, 2016.

J. I. Kim, C. Chun, B. Kim, J. M. Hong, J. K. Cho et al., Thermosensitive/magnetic poly(organophosphazene) hydrogel as a long-term magnetic resonance contrast platform, Biomaterials, pp.218-224, 2012.

L. Jiang, Q. Zhou, K. Mu, H. Xie, Y. Zhu et al., pH/temperature sensitive magnetic nanogels conjugated with Cy5.5-labled lactoferrin for MR and fluorescence imaging of glioma in rats, Biomaterials, vol.34, pp.7418-7428, 2013.
DOI : 10.1016/j.biomaterials.2013.05.078

H. Xie, Y. Zhu, W. Jiang, Q. Zhou, H. Yang et al., Lactoferrin-conjugated superparamagnetic iron oxide nanoparticles as a specific MRI contrast agent for detection of brain glioma in vivo, Biomaterials, vol.32, pp.495-502, 2011.

N. Qi, X. Tang, X. Lin, P. Gu, C. Cai et al., Sterilization stability of vesicular phospholipid gels loaded with cytarabine for brain implant, Int J Pharm, vol.427, pp.234-241, 2012.

S. Zhang, M. A. Anderson, Y. Ao, B. S. Khakh, J. Fan et al., Tunable diblock copolypeptide hydrogel depots for local delivery of hydrophobic molecules in healthy and injured central nervous system, Biomaterials, vol.35, 1989.

K. Park, The drug delivery field at the inflection point: Time to fight its way out of the egg, Journal of Controlled Release, vol.267, pp.2-14, 2017.

K. Riehemann, S. W. Schneider, T. A. Luger, B. Godin, M. Ferrari et al., Nanomedicine -challenge and perspectives, Angew Chem Int Ed Engl, vol.48, pp.872-897, 2009.
DOI : 10.1002/anie.200802585

URL : http://europepmc.org/articles/pmc4175737?pdf=render

A. Miranda, M. J. Blanco-prieto, J. Sousa, A. Pais, and C. Vitorino, Breaching barriers in glioblastoma. Part II: Targeted drug delivery and lipid nanoparticles, Int J Pharm, vol.531, pp.389-410, 2017.

N. T. Huynh, M. Morille, J. Bejaud, P. Legras, A. Vessieres et al., Treatment of 9L gliosarcoma in rats by ferrociphenol-loaded lipid nanocapsules based on a passive targeting strategy via the EPR effect, vol.28, pp.3189-3198, 2011.

M. I. Koukourakis, S. Koukouraki, I. Fezoulidis, N. Kelekis, G. Kyrias et al., High intratumoural accumulation of stealth liposomal doxorubicin (Caelyx) in glioblastomas and in metastatic brain tumours, Brit J Cancer, vol.83, pp.1281-1286, 2000.

T. Siegal, A. Horowitz, and A. Gabizon, Doxorubicin encapsulated in sterically stabilized liposomes for the treatment of a brain tumor model: biodistribution and therapeutic efficacy, J Neurosurg, vol.83, pp.1029-1037, 1995.

A. Bernardi, E. Braganhol, E. Jager, F. Figueiro, M. I. Edelweiss et al., Indomethacin-loaded nanocapsules treatment reduces in vivo glioblastoma growth in a rat glioma model, vol.281, pp.53-63, 2009.

X. Liu, W. Cui, B. Li, and Z. Hong, Targeted therapy for glioma using cyclic RGD-entrapped polyionic complex nanomicelles, Int J Nanomed, vol.7, pp.2853-2862, 2012.

C. Zhan, B. Gu, C. Xie, J. Li, Y. Liu et al., Cyclic RGD conjugated poly(ethylene glycol)-co-poly(lactic acid) micelle enhances paclitaxel anti-glioblastoma effect, J Control Release, vol.143, pp.136-142, 2010.
DOI : 10.1016/j.jconrel.2009.12.020

J. Du, W. L. Lu, X. Ying, Y. Liu, P. Du et al., Dual-targeting topotecan liposomes modified with tamoxifen and wheat germ agglutinin significantly improve drug transport across the blood-brain barrier and survival of brain tumor-bearing animals, Mol Pharm, vol.6, pp.905-917, 2009.

M. Ghosh and R. O. Ryan, ApoE enhances nanodisk-mediated curcumin delivery to glioblastoma multiforme cells, Nanomedicine, vol.9, pp.763-771, 2014.
DOI : 10.2217/nnm.13.35

URL : http://europepmc.org/articles/pmc3904962?pdf=render

J. Chang, A. Paillard, C. Passirani, M. Morille, J. P. Benoit et al., Transferrin adsorption onto PLGA nanoparticles governs their interaction with biological systems from blood circulation to brain cancer cells, Pharm Res, vol.29, pp.1495-1505, 2012.

W. H. Ren, J. Chang, C. H. Yan, X. M. Qian, L. X. Long et al., Development of transferrin functionalized poly(ethylene glycol)/poly(lactic acid) amphiphilic block copolymeric micelles as a potential delivery system targeting brain glioma, J Mater Sci Mater Med, vol.21, pp.2673-2681, 2010.

E. Locatelli, M. Naddaka, C. Uboldi, G. Loudos, E. Fragogeorgi et al., Targeted delivery of silver nanoparticles and alisertib: in vitro and in vivo synergistic effect against glioblastoma, Nanomedicine, vol.9, pp.839-849, 2014.

C. Fang, K. Wang, Z. R. Stephen, Q. Mu, F. M. Kievit et al., Temozolomide nanoparticles for targeted glioblastoma therapy, ACS Appl Mater Interfaces, vol.7, pp.6674-6682, 2015.
DOI : 10.1021/am5092165

URL : http://europepmc.org/articles/pmc4637162?pdf=render

G. Gu, H. Xia, Q. Hu, Z. Liu, M. Jiang et al., PEG-co-PCL nanoparticles modified with MMP-2/9 activatable low molecular weight protamine for enhanced targeted glioblastoma therapy, Biomaterials, vol.34, pp.196-208, 2013.

J. Guo, X. Gao, L. Su, H. Xia, G. Gu et al., Aptamer-functionalized PEG-PLGA nanoparticles for enhanced anti-glioma drug delivery, Biomaterials, vol.32, pp.8010-8020, 2011.

H. Gao, Z. Yang, S. Cao, Y. Xiong, S. Zhang et al., Tumor cells and neovasculature dual targeting delivery for glioblastoma treatment, Biomaterials, vol.35, pp.2374-2382, 2014.
DOI : 10.1016/j.biomaterials.2013.11.076

C. S. Schneider, J. G. Perez, E. Cheng, C. Zhang, P. Mastorakos et al., Minimizing the non-specific binding of nanoparticles to the brain enables active targeting of Fn14-positive glioblastoma cells, Biomaterials, vol.42, pp.42-51, 2015.

A. S. Wadajkar, J. G. Dancy, N. B. Roberts, N. P. Connolly, D. K. Strickland et al., Decreased non-specific adhesivity, receptor targeted (DART) nanoparticles exhibit improved dispersion, cellular uptake, and tumor retention in invasive gliomas, J Control Release, vol.267, pp.144-153, 2017.

A. Beduneau, F. Hindre, A. Clavreul, J. C. Leroux, P. Saulnier et al., Brain targeting using novel lipid nanovectors, J Control Release, vol.126, pp.44-49, 2008.
URL : https://hal.archives-ouvertes.fr/inserm-00258097

A. Beduneau, P. Saulnier, F. Hindre, A. Clavreul, J. C. Leroux et al., Design of targeted lipid nanocapsules by conjugation of whole antibodies and antibody Fab' fragments, Biomaterials, vol.28, pp.4978-4990, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00198713

J. Balzeau, M. Pinier, R. Berges, P. Saulnier, J. Benoit et al., The effect of functionalizing lipid nanocapsules with NFL-TBS.40-63 peptide on their uptake by glioblastoma cells, Biomaterials, vol.34, pp.3381-3389, 2013.

A. Laine, N. T. Huynh, A. Clavreul, J. Balzeau, J. Béjaud et al., Brain tumour targeting strategies via coated ferrociphenol lipid nanocapsules, Eur J Pharm Biopharm, vol.81, pp.690-693, 2012.
DOI : 10.1016/j.ejpb.2012.04.012

URL : https://hal.archives-ouvertes.fr/hal-01877954

Y. C. Kuo and C. T. Liang, Inhibition of human brain malignant glioblastoma cells using carmustine-loaded catanionic solid lipid nanoparticles with surface anti-epithelial growth factor receptor, Biomaterials, vol.32, pp.3340-3350, 2011.

Y. C. Kuo and C. H. Lee, Inhibition against growth of glioblastoma multiforme in vitro using etoposide-loaded solid lipid nanoparticles with p-aminophenyl-alpha-D-manno-pyranoside and folic acid, J Pharm Sci, vol.104, pp.1804-1814, 2015.

M. Nikanjam, A. R. Gibbs, C. A. Hunt, T. F. Budinger, and T. M. Forte, Synthetic nano-LDL with paclitaxel oleate as a targeted drug delivery vehicle for glioblastoma multiforme, J Control Release, vol.124, pp.163-171, 2007.

H. He, Y. Li, X. R. Jia, J. Du, X. Ying et al., PEGylated Poly(amidoamine) dendrimerbased dual-targeting carrier for treating brain tumors, Biomaterials, vol.32, pp.478-487, 2011.

P. Jativa and V. Cena, Use of nanoparticles for glioblastoma treatment: a new approach, Nanomedicine (London), vol.12, pp.2533-2554, 2017.

J. Aparicio-blanco and A. I. Torres-suarez, Glioblastoma Multiforme and Lipid Nanocapsules: A Review, vol.11, pp.1283-1311, 2015.
DOI : 10.1166/jbn.2015.2084

A. S. Wadajkar, J. G. Dancy, D. S. Hersh, P. Anastasiadis, N. L. Tran et al., Tumor-targeted nanotherapeutics: overcoming treatment barriers for glioblastoma, Wiley Interdiscip Rev Nanomed Nanobiotechnol, vol.9, p.4, 2017.

P. A. Chiarelli, F. M. Kievit, M. Zhang, and R. G. Ellenbogen, Bionanotechnology and the future of glioma, Surg Neurol Int, vol.6, pp.45-58, 2015.

N. T. Huynh, C. Passirani, P. Saulnier, and J. P. Benoit, Lipid nanocapsules: a new platform for nanomedicine, Int J Pharm, vol.379, pp.201-209, 2009.

E. Moysan, Y. Gonzalez-fernandez, N. Lautram, J. Bejaud, G. Bastiat et al., An innovative hydrogel of gemcitabine-loaded lipid nanocapsules: when the drug is a key player of the nanomedicine structure, Soft matter, vol.10, pp.1767-1777, 2014.

E. S. Newlands, M. F. Stevens, S. R. Wedge, R. T. Wheelhouse, and C. Brock, Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials, Cancer Treatment Rev, vol.23, pp.35-61, 1997.

H. S. Friedman, T. Kerby, and H. Calvert, Temozolomide and treatment of malignant glioma, Clin Cancer Res, vol.6, pp.2585-2597, 2000.

R. Stupp, M. E. Hegi, W. P. Mason, M. J. Van-den, M. J. Bent et al., Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, vol.10, pp.459-466, 2009.

R. D. Kortmann, B. Jeremic, M. Weller, L. Plasswilm, and M. Bamberg, Radiochemotherapy of malignant glioma in adults. Clinical experiences, Strahlenther Onkol, vol.179, pp.219-232, 2003.

C. Bastiancich, P. Danhier, V. Preat, and F. Danhier, Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma, J Control Release, vol.243, pp.29-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02076746

K. Messaoudi, A. Clavreul, and F. Lagarce, Toward an effective strategy in glioblastoma treatment. Part I: resistance mechanisms and strategies to overcome resistance of glioblastoma to temozolomide, Drug Discov Today, vol.20, pp.899-905, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392441

W. Wick, M. Weller, M. Van-den, M. Bent, M. Sanson et al., MGMT testing--the challenges for biomarker-based glioma treatment, Nat Rev Neurol, vol.10, pp.372-385, 2014.

J. Bianco, C. Bastiancich, A. Jankovski, A. Rieux, V. Preat et al., On glioblastoma and the search for a cure: where do we stand?, Cell Mol Life Sci, vol.74, pp.2451-2466, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076749

E. C. Jung, . Chao, and . St;,

E. Murphy and J. H. Suh, Principles and Tenets of Radiation Treatment in Glioblastoma, 2017.

E. Moysan, G. Bastiat, and J. P. Benoit, Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications, Mol Pharm, vol.10, pp.430-444, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00787112

P. Huang and W. Plunkett, Induction of apoptosis by gemcitabine, Seminars in oncology, vol.22, pp.19-25, 1995.

H. J. Guchelaar, D. J. Richel, and A. Van-knapen, Clinical, toxicological and pharmacological aspects of gemcitabine, Cancer Treat Rev, vol.22, pp.15-31, 1996.

L. J. Sinyuk and M. , Connexins: Bridging the gap between cancer cell communication in glioblastoma, Cancer, 2015.

S. Cottin, K. Ghani, P. O. De-campos-lima, and M. Caruso, Gemcitabine intercellular diffusion mediated by gap junctions: new implications for cancer therapy, Mol Cancer, vol.9, p.141, 2010.

D. S. Shewach and T. S. Lawrence, Gemcitabine and radiosensitization in human tumor cells, Invest New Drugs, vol.14, pp.257-263, 1996.

T. S. Lawrence, E. Y. Chang, T. M. Hahn, and D. S. Shewach, Delayed radiosensitization of human colon carcinoma cells after a brief exposure to 2',2'-difluoro-2'-deoxycytidine (Gemcitabine), Clin Cancer Res, vol.3, pp.777-782, 1997.

A. M. Bergman, H. M. Pinedo, and G. J. Peters, Determinants of resistance to 2',2'-difluorodeoxycytidine (gemcitabine), Drug Resist Updat, vol.5, pp.19-33, 2002.

B. Pauwels, A. E. Korst, H. A. Lambrechts, G. G. Pattyn, C. M. De-pooter et al., The radiosensitising effect of difluorodeoxyuridine, a metabolite of gemcitabine, in vitro, Cancer Chemother Pharmacol, vol.58, pp.219-228, 2006.

T. S. Lawrence, A. W. Blackstock, and C. Mcginn, The mechanism of action of radiosensitization of conventional chemotherapeutic agents, Sem Radiat Oncol, vol.13, pp.13-21, 2003.

B. Pauwels, A. E. Korst, G. G. Pattyn, H. A. Lambrechts, D. R. Van-bockstaele et al., Cell cycle effect of gemcitabine and its role in the radiosensitizing mechanism in vitro, Int J Radiat Oncol Biol Phys, vol.57, pp.1075-1083, 2003.

G. Carpinelli, B. Bucci, I. D'agnano, R. Canese, F. Caroli et al., Gemcitabine treatment of experimental C6 glioma: the effects on cell cycle and apoptotic rate, vol.26, pp.3017-3024, 2006.

L. J. Ostruszka and D. S. Shewach, The role of cell cycle progression in radiosensitization by 2',2'-difluoro-2'-deoxycytidine, Cancer Res, vol.60, pp.6080-6088, 2000.

D. Latz, K. Fleckenstein, M. Eble, J. Blatter, M. Wannenmacher et al., Radiosensitizing potential of gemcitabine (2',2'-difluoro-2'-deoxycytidine) within the cell cycle in vitro, Int J Radiat Oncol Biol Phys, vol.41, pp.875-882, 1998.

A. K. Nowak, B. W. Robinson, and R. A. Lake, Gemcitabine exerts a selective effect on the humoral immune response: implications for combination chemo-immunotherapy, Cancer Res, vol.62, pp.2353-2358, 2002.

E. Suzuki, V. Kapoor, A. S. Jassar, L. R. Kaiser, and S. M. Albelda, Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity, Clin Cancer Res, vol.11, pp.6713-6721, 2005.

E. Suzuki, J. Sun, V. Kapoor, A. S. Jassar, and S. M. Albelda, Gemcitabine has significant immunomodulatory activity in murine tumor models independent of its cytotoxic effects, Cancer Biol Ther, vol.6, pp.880-885, 2007.

I. Shevchenko, S. Karakhanova, S. Soltek, J. Link, J. Bayry et al., Low-dose gemcitabine depletes regulatory T cells and improves survival in the orthotopic Panc02 model of pancreatic cancer, Int J Cancer, vol.133, pp.98-107, 2013.

A. A. Thomas, M. S. Ernstoff, and C. E. Fadul, Immunotherapy for the Treatment of Glioblastoma, Cancer J, vol.18, pp.59-68, 2012.

J. Rieger, S. Durka, J. Streffer, J. Dichgans, and M. Weller, Gemcitabine cytotoxicity of human malignant glioma cells: modulation by antioxidants, BCL-2 and dexamethasone, Eur J Pharm, vol.365, pp.301-308, 1999.

M. Weller, J. Streffer, W. Wick, R. D. Kortmann, E. Heiss et al., Preirradiation gemcitabine chemotherapy for newly diagnosed glioblastoma. A phase II study, Cancer, pp.423-427, 2001.

S. Z. Gertler, D. Macdonald, M. Goodyear, P. Forsyth, D. J. Stewart et al., NCIC-CTG phase II study of gemcitabine in patients with malignant glioma (IND.94), vol.11, pp.315-318, 2000.

W. Wick, M. Hermisson, R. D. Kortmann, W. M. Kuker, F. Duffner et al., Neoadjuvant gemcitabine/treosulfan chemotherapy for newly diagnosed glioblastoma: a phase II study, J Neurooncol, vol.59, pp.151-155, 2002.

H. Szelenyi, E. Thiel, N. Niederle, and U. Keilholz, A Phase I trial of gemcitabine and treosulfan (GeT) to overcome multidrug resistance in patients with advanced solid tumors, Proc Am Soc Clin Oncol, p.228, 1999.

M. Genc, N. Castro-kreder, A. Barten-van-rijbroek, L. J. Stalpers, and J. Haveman, Enhancement of effects of irradiation by gemcitabine in a glioblastoma cell line and cell line spheroids, J Cancer Res Clin Oncol, vol.130, pp.45-51, 2004.

F. Fehlauer, M. Muench, E. J. Smid, B. Slotman, E. Richter et al., Combined modality therapy of gemcitabine and irradiation on human glioma spheroids derived from cell lines and biopsy tissue, Oncol Rep, vol.15, pp.97-105, 2006.

Y. Esumi, K. Mitsugi, A. Takao, H. Seki, and M. Kawai, Disposition of gemcitabine in rat and dog after single and multiple dosings, vol.24, pp.805-817, 1994.

J. Z. Kerr, S. L. Berg, R. Dauser, J. Nuchtern, M. J. Egorin et al., Plasma and cerebrospinal fluid pharmacokinetics of gemcitabine after intravenous administration in nonhuman primates, Cancer Chemother Pharmacol, vol.47, pp.411-414, 2001.

S. K. Apparaju, G. A. Gudelsky, and P. B. Desai, Pharmacokinetics of gemcitabine in tumor and non-tumor extracellular fluid of brain: an in vivo assessment in rats employing intracerebral microdialysis, Cancer Chemother Pharmacol, pp.223-229, 2008.

J. Sigmond, R. J. Honeywell, T. J. Postma, C. M. Dirven, S. M. De-lange et al., Gemcitabine uptake in glioblastoma multiforme: potential as a radiosensitizer, vol.20, pp.182-187, 2009.

T. S. Lawrence, A. Eisbruch, and D. S. Shewach, Gemcitabine-mediated radiosensitization, Semin Oncol, vol.24, pp.7-24, 1997.

A. Maraveyas, J. Sgouros, S. Upadhyay, A. H. Abdel-hamid, M. Holmes et al., Gemcitabine twice weekly as a radiosensitiser for the treatment of brain metastases in patients with carcinoma: a phase I study, Br J Cancer, vol.92, pp.815-819, 2005.

A. Fabi, A. Mirri, A. Felici, A. Vidiri, A. Pace et al., Fixed dose-rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma: a dose-finding study, J Neurooncol, vol.87, pp.79-84, 2008.

G. Metro, A. Fabi, M. A. Mirri, A. Vidiri, A. Pace et al., Phase II study of fixed dose rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma multiforme, Cancer Chemother Pharmacol, vol.65, pp.391-397, 2010.

M. M. Kim, S. Camelo-piragua, M. Schipper, Y. Tao, D. Normolle et al., Gemcitabine Plus Radiation Therapy for High-Grade

K. A. Batich and J. H. Sampson, Standard of care and future pharmacological treatment options for malignant glioma: an urgent need for screening and identification of novel tumor-specific antigens, Expert Opin Pharmacother, vol.15, pp.2047-2061, 2014.

S. Tsigkos, S. Mariz, J. Llinares, L. Fregonese, S. Aarum et al., Establishing medical plausibility in the context of orphan medicines designation in the European Union, Orphanet J Rare Dis, vol.9, p.175, 2014.

Q. T. Ostrom, H. Gittleman, P. Farah, A. Ondracek, Y. Chen et al., CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States, vol.15, pp.1-56, 2006.

S. A. Grossman, X. Ye, S. Piantadosi, S. Desideri, L. B. Nabors et al., 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.2443-2449, 2010.

R. D. Kortmann, B. Jeremic, M. Weller, L. Plasswilm, and M. Bamberg, Radiochemotherapy of malignant glioma in adults. Clinical experiences, Strahlenther Onkol, vol.179, pp.219-232, 2003.

R. Stupp, M. Brada, M. J. Van-den, J. C. Bent, G. Tonn et al., High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.25, issue.3, pp.93-101, 2014.

P. P. Wang, J. Frazier, and H. Brem, Local drug delivery to the brain, Adv Drug Deliv Rev, vol.54, pp.987-1013, 2002.

S. A. Grossman and J. F. Batara, Current management of glioblastoma multiforme, Semin Oncol, pp.635-644, 2004.

Q. Pan, X. J. Yang, H. M. Wang, X. T. Dong, W. Wang et al., Chemoresistance to temozolomide in human glioma cell line U251 is associated with increased activity of O6-methylguanine-DNA methyltransferase and can be overcome by metronomic temozolomide regimen, Cell Biochem Biophys, vol.62, pp.185-191, 2012.

J. N. Sarkaria, G. J. Kitange, C. D. James, R. Plummer, H. Calvert et al., Mechanisms of chemoresistance to alkylating agents in malignant glioma, Clin Cancer Res, vol.14, pp.2900-2908, 2008.

F. Danhier, K. Messaoudi, L. Lemaire, J. P. Benoit, and F. Lagarce, Combined anti-Galectin-1 and anti-EGFR siRNA-loaded chitosan-lipid nanocapsules decrease temozolomide resistance in glioblastoma: in vivo evaluation, Int J Pharm, vol.481, pp.154-161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392431

K. Messaoudi, A. Clavreul, and F. Lagarce, Toward an effective strategy in glioblastoma treatment. Part I: resistance mechanisms and strategies to overcome resistance of glioblastoma to temozolomide, Drug Discov Today, vol.20, pp.899-905, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392441

F. Danhier, O. Feron, and V. Preat, To exploit the tumor microenvironment: Passive and active tumor targeting of nanocarriers for anti-cancer drug delivery, J Control Release, vol.148, pp.135-146, 2010.

F. Danhier, A. L. Breton, and V. Preat, RGD-based strategies to target alpha(v) beta(3) integrin in cancer therapy and diagnosis, Mol Pharm, vol.9, pp.2961-2973, 2012.

N. T. Huynh, C. Passirani, P. Saulnier, and J. P. Benoit, Lipid nanocapsules: a new platform for nanomedicine, Int J Pharm, vol.379, pp.201-209, 2009.

J. Aparicio-blanco and A. I. Torres-suarez, Glioblastoma Multiforme and Lipid Nanocapsules: A Review, vol.11, pp.1283-1311, 2015.

F. H. Hochberg and A. Pruitt, Assumptions in the radiotherapy of glioblastoma, vol.30, pp.907-911, 1980.

M. Westphal, D. C. Hilt, E. Bortey, P. Delavault, R. Olivares et al., A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma, Neuro Oncol, vol.5, pp.79-88, 2003.

H. H. Engelhard, The role of interstitial BCNU chemotherapy in the treatment of malignant glioma, Surg Neurol, vol.53, pp.458-464, 2000.

D. A. Bota, A. Desjardins, J. A. Quinn, M. L. Affronti, and H. S. Friedman, Interstitial chemotherapy with biodegradable BCNU (Gliadel) wafers in the treatment of malignant gliomas, Ther Clin Risk Manag, vol.3, pp.707-715, 2007.

T. A. Juratli, G. Schackert, and D. Krex, Current status of local therapy in malignant gliomas--a clinical review of three selected approaches, Pharmacol Ther, vol.139, pp.341-358, 2013.

S. Brem, B. Tyler, K. Li, G. Pradilla, F. Legnani et al., Local delivery of temozolomide by biodegradable polymers is superior to oral administration in a rodent glioma model, Cancer Chemother Pharmacol, vol.60, pp.643-650, 2007.

B. Tyler, K. D. Fowers, K. W. Li, V. R. Recinos, J. M. Caplan et al., A thermal gel depot for local delivery of paclitaxel to treat experimental brain tumors in rats, J Neurosurg, vol.113, pp.210-217, 2010.

T. Ozeki, D. Kaneko, K. Hashizawa, Y. Imai, T. Tagami et al., Combination therapy of surgical tumor resection with implantation of a hydrogel containing camptothecin-loaded poly(lactic-co-glycolic acid) microspheres in a C6 rat glioma model, Biol Pharm Bull, vol.35, pp.545-550, 2012.

B. Y. Ong, S. H. Ranganath, L. Y. Lee, F. Lu, H. S. Lee et al., Paclitaxel delivery from PLGA foams for controlled release in post-surgical chemotherapy against glioblastoma multiforme, Biomaterials, vol.30, pp.3189-3196, 2009.

A. K. Vellimana, V. R. Recinos, L. Hwang, K. D. Fowers, K. W. Li et al., Combination of paclitaxel thermal gel depot with temozolomide and radiotherapy significantly prolongs survival in an experimental rodent glioma model, J Neurooncol, vol.111, pp.229-236, 2013.

P. Menei, L. Capelle, J. Guyotat, S. Fuentes, R. Assaker et al., Local and sustained delivery of 5-fluorouracil from biodegradable microspheres for the radiosensitization of malignant glioma: a randomized phase II trial, Neurosurgery, vol.56, pp.242-248, 2005.

T. Fourniols, L. D. Randolph, A. Staub, K. Vanvarenberg, J. G. Leprince et al., Temozolomide-loaded photopolymerizable PEG-DMA-based hydrogel for the treatment of glioblastoma, J Control Release, vol.210, pp.95-104, 2015.

J. Sigmond, R. J. Honeywell, T. J. Postma, C. M. Dirven, S. M. De-lange et al., Gemcitabine uptake in glioblastoma multiforme: potential as a radiosensitizer, vol.20, pp.182-187, 2009.

E. Moysan, G. Bastiat, and J. P. Benoit, Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications, Mol Pharm, vol.10, pp.430-444, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00787112

M. L. Immordino, P. Brusa, F. Rocco, S. Arpicco, M. Ceruti et al., Cattel, Preparation, characterization, cytotoxicity and pharmacokinetics of liposomes containing lipophilic gemcitabine prodrugs, J Control Release, vol.100, pp.331-346, 2004.

E. Moysan, Y. Gonzalez-fernandez, N. Lautram, J. Bejaud, G. Bastiat et al., An innovative hydrogel of gemcitabine-loaded lipid nanocapsules: when the drug is a key player of the nanomedicine structure, Soft Matter, vol.10, pp.1767-1777, 2014.

S. Galban, B. Lemasson, T. M. Williams, F. Li, K. A. Heist et al., DW-MRI as a biomarker to compare therapeutic outcomes in radiotherapy regimens incorporating temozolomide or gemcitabine in glioblastoma, PLoS One, vol.7, p.35857, 2012.

S. P. Heurtault, B. Pech, B. Proust, J. E. Richard, J. Benoit et al., Lipidic nanocapsules, formulation process and use as a drug delivery system, 2001.

B. Heurtault, P. Saulnier, B. Pech, J. E. Proust, and J. P. Benoit, A novel phase inversion-based process for the preparation of lipid nanocarriers, Pharm Res, vol.19, pp.875-880, 2002.

S. Arpicco, C. Lerda, E. Pozza, C. Costanzo, N. Tsapis et al., Hyaluronic acid-coated liposomes for active targeting of gemcitabine, Eur J Pharm Biopharm, vol.85, pp.373-380, 2013.

A. Beloqui, M. A. Solinis, A. R. Gascon, A. Pozo-rodriguez, A. Rieux et al., Mechanism of transport of saquinavir-loaded nanostructured lipid carriers across the intestinal barrier, J Control Release, vol.166, pp.115-123, 2013.

N. Hajos and I. Mody, Establishing a physiological environment for visualized in vitro brain slice recordings by increasing oxygen supply and modifying aCSF content, J Neurosci Methods, vol.183, pp.107-113, 2009.

K. K. Chereddy, A. Lopes, S. Koussoroplis, V. Payen, C. Moia et al., Combined effects of PLGA and vascular endothelial growth factor promote the healing of non-diabetic and diabetic wounds, Nanomedicine, vol.11, pp.1975-1984, 2015.

H. Meng, M. Wang, H. Liu, X. Liu, A. Situ et al., Use of a lipid-coated mesoporous silica nanoparticle platform for synergistic gemcitabine and paclitaxel delivery to human pancreatic cancer in mice, ACS nano, vol.9, pp.3540-3557, 2015.

X. Yu, Y. Di, C. Xie, Y. Song, H. He et al., An in vitro and in vivo study of gemcitabine-loaded albumin nanoparticles in a pancreatic cancer cell line, Int J Nanomedicine, vol.10, pp.6825-6834, 2015.

D. Cosco, D. Paolino, F. De-angelis, F. Cilurzo, C. Celia et al., Aqueous-core PEG-coated PLA nanocapsules for an efficient entrapment of water soluble anticancer drugs and a smart therapeutic response, Eur J Pharm Biopharm, vol.89, pp.30-39, 2015.

T. Manabe, H. Okino, R. Maeyama, K. Mizumoto, M. Tanaka et al., New infusion device for transtissue, sustained local delivery of anticancer agent to surgically resected tissue: potential use for suppression of local recurrence of pancreatic cancer, J Biomed Mater Res B Appl Biomater, vol.73, pp.203-207, 2005.

S. Bobone, E. Miele, B. Cerroni, D. Roversi, A. Bocedi et al., Liposome-Templated Hydrogel Nanoparticles as Vehicles for Enzyme-Based Therapies, pp.7572-7580, 2015.

J. Hao, X. Wang, Y. Bi, Y. Teng, J. Wang et al., Fabrication of a composite system combining solid lipid nanoparticles and thermosensitive hydrogel for challenging ophthalmic drug delivery, Colloids Surf B Biointerfaces, vol.114, pp.111-120, 2014.

A. C. Santos-akkari, E. V. Ramos-campos, A. F. Keppler, L. F. Fraceto, E. De-paula et al., Budesonide-hydroxypropyl-beta-cyclodextrin inclusion complex in binary poloxamer 407/403 system for ulcerative colitis treatment: A physico-chemical study from micelles to hydrogels, Colloids Surf B Biointerfaces, pp.138-147, 2016.

S. Chatelin, A. Constantinesco, and R. Willinger, Fifty years of brain tissue mechanical testing: from in vitro to in vivo investigations, Biorheology, pp.255-276, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00591614

J. J. Iliff, M. Wang, Y. Liao, B. A. Plogg, W. Peng et al., A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta, vol.4, pp.147-111, 2012.

P. Menei, E. Jadaud, N. Faisant, M. Boisdron-celle, S. Michalak et al., Stereotaxic implantation of 5-fluorouracil-releasing microspheres in malignant glioma, Cancer, vol.100, pp.405-410, 2004.

J. Rieger, S. Durka, J. Streffer, J. Dichgans, and M. Weller, Gemcitabine cytotoxicity of human malignant glioma cells: modulation by antioxidants, BCL-2 and dexamethasone, Eur J Pharmacol, vol.365, pp.301-308, 1999.

L. J. Ostruszka and D. S. Shewach, The role of cell cycle progression in radiosensitization by 2',2'-difluoro-2'-deoxycytidine, Cancer Res, vol.60, pp.6080-6088, 2000.

P. Guo, J. Ma, S. Li, Z. Guo, A. L. Adams et al., Targeted delivery of a peripheral benzodiazepine receptor ligand-gemcitabine conjugate to brain tumors in a xenograft model, Cancer Chemother Pharmacol, vol.48, pp.169-176, 2001.

M. Genc, N. Castro-kreder, A. Barten-van-rijbroek, L. J. Stalpers, and J. Haveman, Enhancement of effects of irradiation by gemcitabine in a glioblastoma cell line and cell line spheroids, J Cancer Res Clin Oncol, vol.130, pp.45-51, 2004.

G. Carpinelli, B. Bucci, I. D'agnano, R. Canese, F. Caroli et al., Gemcitabine treatment of experimental C6 glioma: the effects on cell cycle and apoptotic rate, vol.26, pp.3017-3024, 2006.

C. X. Wang, L. S. Huang, L. B. Hou, L. Jiang, Z. T. Yan et al., Antitumor effects of polysorbate-80 coated gemcitabine polybutylcyanoacrylate nanoparticles in vitro and its pharmacodynamics in vivo on C6 glioma cells of a brain tumor model, Brain Res, pp.91-99, 2009.

P. Brusa, M. L. Immordino, F. Rocco, and L. Cattel, Antitumor activity and pharmacokinetics of liposomes containing lipophilic gemcitabine prodrugs, Anticancer Res, vol.27, pp.195-199, 2007.

W. G. Chung, M. A. Sandoval, B. R. Sloat, P. D. Lansakara, and Z. Cui, Stearoyl gemcitabine nanoparticles overcome resistance related to the over-expression of ribonucleotide reductase subunit M1, J Control Release, vol.157, pp.132-140, 2012.

B. R. Sloat, M. A. Sandoval, D. Li, W. G. Chung, P. D. Lansakara et al., In vitro and in vivo anti-tumor activities of a gemcitabine derivative carried by nanoparticles, Int J Pharm, vol.409, pp.278-288, 2011.

N. Wauthoz, G. Bastiat, E. Moysan, A. Cieslak, K. Kondo et al., Safe lipid nanocapsule-based gel technology to target lymph nodes and combat mediastinal metastases from an orthotopic non-small-cell lung cancer model in SCID-CB17 mice, Nanomedicine, vol.11, pp.1237-1245, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392447

E. Garcion, A. Lamprecht, B. Heurtault, A. Paillard, A. Aubert-pouessel et al., A new generation of anticancer, drug-loaded, colloidal vectors reverses multidrug resistance in glioma and reduces tumor progression in rats, Molecular cancer therapeutics, issue.5, pp.1710-1722, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00129865

A. Paillard, F. Hindre, C. Vignes-colombeix, J. P. Benoit, and E. Garcion, The importance of endo-lysosomal escape with lipid nanocapsules for drug subcellular bioavailability, Biomaterials, pp.7542-7554, 2010.

H. Okino, R. Maeyama, T. Manabe, T. Matsuda, and M. Tanaka, Trans-tissue, sustained release of gemcitabine from photocured gelatin gel inhibits the growth of heterotopic human pancreatic tumor in nude mice, Clin Cancer Res, vol.9, pp.5786-5793, 2003.

O. Benny, S. K. Kim, K. Gvili, I. S. Radzishevsky, A. Mor et al., In vivo fate and therapeutic efficacy of PF-4/CTF microspheres in an orthotopic human glioblastoma model, FASEB J, vol.22, pp.488-499, 2008.

J. T. Dilworth, S. A. Krueger, M. Dabjan, I. S. Grills, J. Torma et al., Pulsed low-dose irradiation of orthotopic glioblastoma multiforme (GBM) in a pre-clinical model: Effects on vascularization and tumor control, Radiotherapy and Oncology, vol.108, pp.149-154, 2013.

Q. Mu, G. Lin, V. K. Patton, K. Wang, O. W. Press et al., Gemcitabine and chlorotoxin conjugated iron oxide nanoparticles for glioblastoma therapy, J Mater Chem B Mater Biol Med, vol.4, pp.32-36, 2016.

D. H. Shin, S. J. Lee, J. S. Kim, J. H. Ryu, and J. S. Kim, Synergistic Effect of Immunoliposomal Gemcitabine and Bevacizumab in Glioblastoma Stem Cell-Targeted Therapy, J Biomed Nanotechnol, vol.11, pp.1989-2002, 2015.

S. Z. Gertler, D. Macdonald, M. Goodyear, P. Forsyth, D. J. Stewart et al., NCIC-CTG phase II study of gemcitabine in patients with malignant glioma (IND.94), vol.11, pp.315-318, 2000.

M. Weller, J. Streffer, W. Wick, R. D. Kortmann, E. Heiss et al., Preirradiation gemcitabine chemotherapy for newly diagnosed glioblastoma. A phase II study, Cancer, pp.423-427, 2001.

A. Fabi, A. Mirri, A. Felici, A. Vidiri, A. Pace et al., Fixed dose-rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma: a dose-finding study, J Neurooncol, vol.87, pp.79-84, 2008.

G. Metro, A. Fabi, M. A. Mirri, A. Vidiri, A. Pace et al., Phase II study of fixed dose rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma multiforme, Cancer Chemother Pharmacol, vol.65, pp.391-397, 2010.

W. Wick, M. Hermisson, R. D. Kortmann, W. M. Kuker, F. Duffner et al., Neoadjuvant gemcitabine/treosulfan chemotherapy for newly diagnosed glioblastoma: a phase II study, J Neurooncol, vol.59, pp.151-155, 2002.

. Shimizu, Routes of administration, The Laboratory Mouse (Handbook of Experimental Animals, pp.527-541, 2004.

E. Fournier, C. Passirani, C. N. Montero-menei, and J. P. Benoit, Biocompatibility of implantable synthetic polymeric drug carriers: focus on brain biocompatibility, pp.3311-3331, 2003.

D. Ito, Y. Imai, K. Ohsawa, K. Nakajima, Y. Fukuuchi et al., Microglia-specific localisation of a novel calcium binding protein, Iba1, Brain Res Mol Brain Res, vol.57, pp.1-9, 1998.

K. B. Bjugstad, K. Lampe, D. S. Kern, and M. Mahoney, Biocompatibility of poly(ethylene glycol)-based hydrogels in the brain: an analysis of the glial response across space and time, J Biomed Mater Res A, vol.95, pp.79-91, 2010.

M. T. Giordana, A. Attanasio, P. Cavalla, A. Migheli, M. C. Vigliani et al., Reactive cell proliferation and microglia following injury to the rat brain, Neuropathology and applied neurobiology, vol.20, p.163, 1994.

A. Bernardi, E. Braganhol, E. Jäger, F. Figueiró, M. Edelweiss et al., Indomethacin-loaded nanocapsules treatment reduces in vivo glioblastoma growth in a rat glioma model, Cancer Lett, vol.281, pp.53-63, 2009.

R. Stupp, W. Mason, M. Van-den, M. Bent, B. Weller et al., Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, vol.352, pp.987-996, 2005.

F. Lefranc, N. Sadeghi, I. Camby, T. Metens, O. Dewitte et al., Present and potential future issues in glioblastoma treatmen, Expert Rev Anticancer Ther, issue.6, pp.719-732, 2006.

C. Bastiancich, P. Danhier, V. Préat, and F. Danhier, Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma, J Control Release, vol.243, pp.29-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02076746

H. Bock, M. Puchner, F. Lohmann, M. Schütze, S. Koll et al., First-line treatment of malignant glioma with carmustine implants followed by concomitant radiochemotherapy: a multicenter experience, Neurosurg Rev, vol.33, pp.441-449, 2010.

L. Ashby, K. Smith, and B. Stea, Gliadel wafer implantation combined with standard radiotherapy and concurrent followed by adjuvant temozolomide for treatment of newly diagnosed high-grade glioma: a systematic literature review, World J Surg Oncol, vol.14, p.225, 2016.

T. Kauer, J. Figueiredo, S. Hingtgen, and K. Shah, Encapsulated therapeutic stem cells implanted in the tumor resection cavity induce cell death in gliomas, Nat Neurosci, vol.15, pp.197-214, 2011.

U. Akbar, T. Jones, J. Winestone, M. Michael, A. Shukla et al., Delivery of temozolomide to the tumor bed via biodegradable gel matrices in a novel model of intracranial glioma with resection, J Neurooncol, vol.94, pp.203-212, 2009.

J. Denbo, R. Williams, W. Orr, T. Sims, C. Ng et al., Continuous local delivery of interferon-? stabilizes tumor vasculature in an orthotopic glioblastoma xenograft resection model, Surgery, vol.150, pp.497-504, 2011.

T. Ozeki, D. Kaneko, K. Hashizawa, Y. Imai, T. Tagami et al., Combination therapy of surgical tumor resection with implantation of a hydrogel containing camptothecin-loaded poly(lactic-co-glycolic acid) microspheres in a C6 rat glioma model, Biol Pharm Bull, vol.35, pp.545-550, 2012.

F. Danhier, K. Messaoudi, L. Lemaire, J. Benoit, and F. Lagarce, Combined anti-Galectin-1 and anti-EGFR siRNA-loaded chitosan-lipid nanocapsules decrease temozolomide resistance in glioblastoma: in vivo evaluation, Int J Pharm, vol.481, pp.154-161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392431

V. Jacobs, P. Valdes, W. Hickey, and J. D. Leo, Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model, ASN Neuro, vol.3, p.63, 2011.

K. Chaichana, I. Jusue-torres, R. Navarro-ramirez, S. 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.

A. Fukui, Y. Muragaki, T. Saito, T. Maruyama, M. Nitta et al., Volumetric analysis using low-field intraoperative magnetic resonance imaging for 168 newly diagnosed supratentorial glioblastomas: effects of extent of resection and residual tumor volume on survival and recurrence, World Neurosurg, vol.98, pp.73-80, 2016.

N. Sanai, M. Polley, M. Mcdermott, A. Parsa, and M. Berger, An extent of resection threshold for newly diagnosed glioblastomas, J Neurosurg, vol.115, pp.3-8, 2011.

T. Hoare and D. Kohane, Hydrogels in drug delivery: Progress and challenges, vol.49, pp.1993-2007, 2008.

F. Hochberg and A. Pruitt, Assumptions in the radiotherapy of glioblastoma, vol.30, pp.907-911, 1980.

E. C. Holland, Glioblastoma multiforme: the terminator, Proc Natl Acad Sci U S A, vol.97, pp.6242-6244, 2000.
DOI : 10.1073/pnas.97.12.6242

URL : http://www.pnas.org/content/97/12/6242.full.pdf

D. N. Louis, A. Perry, G. Reifenberger, A. Deimling, D. Figarella-branger et al., The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary, Acta Neuropathol, vol.131, pp.803-820, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01479018

R. L. Yong and R. R. Lonser, Surgery for glioblastoma multiforme: striking a balance, World neurosurgery, vol.76, pp.528-530, 2011.

K. R. Yabroff, L. Harlan, C. Zeruto, J. Abrams, and B. Mann, Patterns of care and survival for patients with glioblastoma multiforme diagnosed during, Neuro-oncology, vol.14, pp.351-359, 2006.

R. Stupp, M. Brada, M. J. Van-den, J. C. Bent, G. Tonn et al., High-grade glioma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up, Ann Oncol, vol.25, issue.3, pp.93-101, 2014.

F. H. Hochberg and A. Pruitt, Assumptions in the radiotherapy of glioblastoma, vol.30, pp.907-911, 1980.

S. Roy, D. Lahiri, T. Maji, and J. Biswas, Recurrent Glioblastoma: Where we stand, South Asian J Cancer, vol.4, pp.163-173, 2015.
DOI : 10.4103/2278-330x.175953

URL : https://doi.org/10.4103/2278-330x.175953

R. Stupp, M. E. Hegi, W. P. Mason, M. J. Van-den, M. J. Bent et al., Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase, vol.10, pp.459-466, 2009.

N. R. Smoll, K. Schaller, and O. P. Gautschi, Long-term survival of patients with glioblastoma multiforme (GBM), J Clin Neurosci, vol.20, pp.670-675, 2013.

T. Yamahara, Y. Numa, T. Oishi, T. Kawaguchi, T. Seno et al., Morphological and flow cytometric analysis of cell infiltration in glioblastoma: a comparison of autopsy brain and neuroimaging, Brain Tumor Pathol, vol.27, pp.81-87, 2010.

T. Denysenko, L. Gennero, M. A. Roos, A. Melcarne, C. Juenemann et al., Glioblastoma cancer stem cells: heterogeneity, microenvironment and related therapeutic strategies, Cell biochemistry and function, vol.28, pp.343-351, 2010.
DOI : 10.1002/cbf.1666

D. Treister, S. Kingston, K. E. Hoque, M. Law, and M. S. Shiroishi, Multimodal magnetic resonance imaging evaluation of primary brain tumors, Semin Oncol, pp.478-495, 2014.

S. Osuka and E. G. Van-meir, Overcoming therapeutic resistance in glioblastoma: the way forward, J Clin Invest, vol.127, pp.415-426, 2017.

D. M. Patel, N. Agarwal, K. L. Tomei, D. R. Hansberry, and I. M. Goldstein, Optimal Timing of Whole-Brain Radiation Therapy Following Craniotomy for Cerebral Malignancies, World neurosurgery, vol.84, pp.412-419, 2015.

J. R. Silber, M. S. Bobola, A. Blank, and M. C. Chamberlain, O(6)-methylguanine-DNA methyltransferase in glioma therapy: promise and problems, Biochim Biophys Acta, pp.71-82, 2012.

N. R. Parker, A. L. Hudson, P. Khong, J. F. Parkinson, T. Dwight et al., Intratumoral heterogeneity identified at the epigenetic, genetic and transcriptional level in glioblastoma, Sci rep, vol.6, p.22477, 2016.

H. Okura, C. A. Smith, and J. T. Rutka, Gene therapy for malignant glioma, Mol Cell Ther, vol.2, p.21, 2014.

A. Tivnan, T. Heilinger, E. C. Lavelle, and J. H. Prehn, Advances in immunotherapy for the treatment of glioblastoma, J Neurooncol, pp.1-9, 2017.

A. Mangraviti, D. Gullotti, B. Tyler, and H. Brem, Nanobiotechnology-based delivery strategies: New frontiers in brain tumor targeted therapies, J Control Release, vol.240, pp.443-453, 2016.

R. Karim, C. Palazzo, B. Evrard, and G. Piel, Nanocarriers for the treatment of glioblastoma multiforme: Current state-of-the-art, J Control Release, vol.227, pp.23-37, 2016.

A. Burgess, K. Shah, O. Hough, and K. Hynynen, Focused ultrasound-mediated drug delivery through the blood-brain barrier, Expert Rev Neurother, vol.15, pp.477-491, 2015.
DOI : 10.1586/14737175.2015.1028369

URL : http://europepmc.org/articles/pmc4702264?pdf=render

J. Bianco, C. Bastiancich, A. Jankovski, A. Rieux, V. Preat et al., On glioblastoma and the search for a cure: where do we stand?, Cell Mol Life Sci, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076749

M. Westphal, Z. Ram, V. Riddle, D. Hilt, and E. Bortey, Gliadel wafer in initial surgery for malignant glioma: long-term follow-up of a multicenter controlled trial, Acta Neurochir, pp.269-275, 2006.

P. Menei, L. Capelle, J. Guyotat, S. Fuentes, R. Assaker et al., Local and sustained delivery of 5-fluorouracil from biodegradable microspheres for the radiosensitization of malignant glioma: a randomized phase II trial, Neurosurgery, vol.56, pp.242-248, 2005.

C. Bastiancich, P. Danhier, V. Preat, and F. Danhier, Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma, J Control Release, vol.243, pp.29-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02076746

D. A. Bota, A. Desjardins, J. A. Quinn, M. L. Affronti, and H. S. Friedman, Interstitial chemotherapy with biodegradable BCNU (Gliadel) wafers in the treatment of malignant gliomas, Ther Clin Risk Manag, vol.3, pp.707-715, 2007.

M. Westphal, D. C. Hilt, E. Bortey, P. Delavault, R. Olivares et al., A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma, Neuro Oncol, vol.5, pp.79-88, 2003.

P. C. Mcgovern, E. Lautenbach, P. J. Brennan, R. A. Lustig, and N. O. Fishman, Risk factors for postcraniotomy surgical site infection after 1,3-bis (2-chloroethyl)-1-nitrosourea (Gliadel) wafer placement, Clin Infect Dis, vol.36, pp.759-765, 2003.

C. Bastiancich, K. Vanvarenberg, B. Ucakar, M. Pitorre, G. Bastiat et al., Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of glioblastoma, J Control Release, vol.225, pp.283-293, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388804

T. Fourniols, L. D. Randolph, A. Staub, K. Vanvarenberg, J. G. Leprince et al., Temozolomide-loaded photopolymerizable PEG-DMA-based hydrogel for the treatment of glioblastoma, J Control Release, vol.210, pp.95-104, 2015.

J. B. Wolinsky, Y. L. Colson, and M. W. Grinstaff, Local drug delivery strategies for cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers, J Control Release, vol.159, pp.14-26, 2012.
DOI : 10.1016/j.jconrel.2011.11.031

URL : http://europepmc.org/articles/pmc3878823?pdf=render

B. Heurtault, P. Saulnier, B. Pech, J. E. Proust, and J. P. Benoit, A novel phase inversion-based process for the preparation of lipid nanocarriers, Pharm res, vol.19, pp.875-880, 2002.

E. Moysan, Y. Gonzalez-fernandez, N. Lautram, J. Bejaud, G. Bastiat et al., An innovative hydrogel of gemcitabine-loaded lipid nanocapsules: when the drug is a key player of the nanomedicine structure, Soft Matter, vol.10, pp.1767-1777, 2014.

N. Wauthoz, G. Bastiat, E. Moysan, A. Cieslak, K. Kondo et al., Safe lipid nanocapsule-based gel technology to target lymph nodes and combat mediastinal metastases from an orthotopic non-small-cell lung cancer model in SCID-CB17 mice, Nanomedicine, vol.11, pp.1237-1245, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392447

R. L. Alexander, B. T. Greene, S. V. Torti, and G. L. Kucera, A novel phospholipid gemcitabine conjugate is able to bypass three drug-resistance mechanisms, Cancer Chemother Pharmacol, vol.56, pp.15-21, 2005.

F. Danhier, K. Messaoudi, L. Lemaire, J. P. Benoit, and F. Lagarce, Combined anti-Galectin-1 and anti-EGFR siRNA-loaded chitosan-lipid nanocapsules decrease temozolomide resistance in glioblastoma: in vivo evaluation, Int J Pharm, vol.481, pp.154-161, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392431

J. Bianco, C. Bastiancich, N. Joudiou, B. Gallez, A. Rieux et al., Novel model of orthotopic U-87 MG glioblastoma resection in athymic nude mice, J Neurosci Methods, vol.284, pp.96-102, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076529

J. R. Mackey, R. S. Mani, M. Selner, D. Mowles, J. D. Young et al., Functional nucleoside transporters are required for gemcitabine influx and manifestation of toxicity in cancer cell lines, Cancer Res, vol.58, pp.4349-4357, 1998.

J. Sun, V. L. Damaraju, C. E. Cass, and M. B. Sawyer, Inhibition of nucleoside transporters by tyrosine kinase inhibitors and its effects on chemotherapy efficacy, Cancer Cell & Microenvironment, vol.1, p.389, 2014.

J. Rieger, S. Durka, J. Streffer, J. Dichgans, and M. Weller, Gemcitabine cytotoxicity of human malignant glioma cells: modulation by antioxidants, BCL-2 and dexamethasone, Eur J Pharmacol, vol.365, pp.301-308, 1999.

S. Y. Cho, J. Polster, J. M. Engles, J. Hilton, E. H. Abraham et al., In vitro evaluation of adenosine 5'-monophosphate as an imaging agent of tumor metabolism, J Nucl Med, vol.47, pp.837-845, 2006.

X. M. Tao, J. C. Wang, J. B. Wang, Q. Feng, S. Y. Gao et al., Enhanced anticancer activity of gemcitabine coupling with conjugated linoleic acid against human breast cancer in vitro and in vivo, Eur J Pharm Biopharm, vol.82, pp.401-409, 2012.

P. D. Lansakara, B. L. Rodriguez, and Z. Cui, Synthesis and in vitro evaluation of novel lipophilic monophosphorylated gemcitabine derivatives and their nanoparticles, Int J Pharm, vol.429, pp.123-134, 2012.

R. D. Dubey, A. Saneja, P. K. Gupta, and P. N. Gupta, Recent advances in drug delivery strategies for improved therapeutic efficacy of gemcitabine, Eur J Pharm Sci, vol.93, pp.147-162, 2016.

D. S. Gesto, N. M. Cerqueira, P. A. Fernandes, and M. J. Ramos, Gemcitabine: a critical nucleoside for cancer therapy, Curr Med Chem, vol.19, pp.1076-1087, 2012.

E. Garcion, A. Lamprecht, B. Heurtault, A. Paillard, A. Aubert-pouessel et al., A new generation of anticancer, drug-loaded, colloidal vectors reverses multidrug resistance in glioma and reduces tumor progression in rats, Mol Cancer Ther, issue.5, pp.1710-1722, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00129865

A. Paillard, F. Hindre, C. Vignes-colombeix, J. P. Benoit, and E. Garcion, The importance of endo-lysosomal escape with lipid nanocapsules for drug subcellular bioavailability, Biomaterials, pp.7542-7554, 2010.

K. Sugano, M. Kansy, P. Artursson, A. Avdeef, S. Bendels et al., Coexistence of passive and carrier-mediated processes in drug transport, Nat Rev Drug Discov, vol.9, pp.597-614, 2010.

G. Bastiat, C. O. Pritz, C. Roider, F. Fouchet, E. Lignieres et al., A new tool to ensure the fluorescent dye labeling stability of nanocarriers: a real challenge for fluorescence imaging, J Control Release, vol.170, pp.334-342, 2013.

K. B. Bjugstad, K. Lampe, D. S. Kern, and M. Mahoney, Biocompatibility of poly(ethylene glycol)-based hydrogels in the brain: an analysis of the glial response across space and time, J Biomed Mater Res A, vol.95, pp.79-91, 2010.

E. Fournier, C. Passirani, N. Colin, S. Sagodira, P. Menei et al., The brain tissue response to biodegradable poly(methylidene malonate 2.1.2)-based microspheres in the rat, Biomaterials, vol.27, pp.4963-4974, 2006.

V. L. Jacobs, P. A. Valdes, W. F. Hickey, and J. A. Leo, Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model, ASN neuro, vol.3, p.63, 2011.

L. Qiang, Y. Yang, Y. J. Ma, F. H. Chen, L. B. Zhang et al., Isolation and characterization of cancer stem like cells in human glioblastoma cell lines, Cancer letters, vol.279, pp.13-21, 2009.

A. Gaudin, E. Song, A. R. King, J. K. Saucier-sawyer, R. Bindra et al., PEGylated squalenoyl-gemcitabine nanoparticles for the treatment of glioblastoma, Biomaterials, pp.136-144, 2016.

O. Okolie, J. R. Bago, R. S. Schmid, D. M. Irvin, R. E. Bash et al., Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model, Neuro Oncol, vol.18, pp.1622-1633, 2016.

L. Zitvogel, A. Tesniere, L. Apetoh, F. Ghiringhelli, and G. Kroemer, Bull Acad Natl Med, vol.192, pp.1469-1489, 2008.

I. Szadvari, O. Krizanova, and P. Babula, Athymic nude mice as an experimental model for cancer treatment, Physiol Res, vol.65, pp.441-453, 2016.

M. S. Sasso, G. Lollo, M. Pitorre, S. Solito, L. Pinton et al., Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy, Biomaterials, pp.47-62, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01392469

T. Wurdinger, K. Deumelandt, H. J. Van-der, P. Vliet, T. D. Wesseling et al., Mechanisms of intimate and long-distance cross-talk between glioma and myeloid cells: how to break a vicious cycle, Biochim Biophys Acta, pp.560-575, 2014.

A. Tuettenberg, K. Steinbrink, and D. Schuppan, Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy, Nanomedicine (Lond), vol.11, pp.2735-2751, 2016.

B. Raychaudhuri, P. Rayman, J. Ireland, J. Ko, B. Rini et al., Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma, Neuro Oncol, vol.13, pp.591-599, 2011.

. Evaluation and . .. Time, ANTI-TUMOR EFFICACY OF GEMC12-LNC AFTER PERISURGICAL ADMINISTRATION IN THE RESECTION CAVITY OF C6 TUMOR-BEARING RATS

A. .. Rats,

I. N. Vitro and . .. Cytotoxicity-studies,

C. Bastiancich, G. Bastiat, and F. Lagarce, Gemcitabine and glioblastoma: challenges and current perspectives, Drug Discov Today, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076519

J. Sigmond, R. J. Honeywell, T. J. Postma, C. M. Dirven, S. M. De-lange et al., Gemcitabine uptake in glioblastoma multiforme: potential as a radiosensitizer, vol.20, pp.182-187, 2009.

G. Metro, A. Fabi, M. A. Mirri, A. Vidiri, A. Pace et al., Phase II study of fixed dose rate gemcitabine as radiosensitizer for newly diagnosed glioblastoma multiforme, Cancer Chemother Pharmacol, vol.65, pp.391-397, 2010.

R. Stupp, W. P. Mason, M. J. Van-den, M. Bent, B. Weller et al., Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma, vol.352, pp.987-996, 2005.

J. Bianco, C. Bastiancich, A. Jankovski, A. Rieux, V. Preat et al., On glioblastoma and the search for a cure: where do we stand?, Cell Mol Life Sci, vol.74, pp.2451-2466, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076749

B. Auffinger, D. Spencer, P. Pytel, A. U. Ahmed, and M. S. Lesniak, The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence, Exp Rev Neurother, vol.15, pp.741-752, 2015.

R. Rahman, Biomaterial-based local drug delivery to brain tumors, Ther Deliv, vol.5, pp.1243-1245, 2014.

L. Kleinberg, 85% carmustine slow-release wafer in malignant glioma: evidence for role in era of standard adjuvant temozolomide, Polifeprosan, vol.20, pp.115-130, 2012.

D. A. Bota, A. Desjardins, J. A. Quinn, M. L. Affronti, and H. S. Friedman, Interstitial chemotherapy with biodegradable BCNU (Gliadel) wafers in the treatment of malignant gliomas, Ther Clin Risk Manag, vol.3, pp.707-715, 2007.

M. Westphal, D. C. Hilt, E. Bortey, P. Delavault, R. Olivares et al., A phase 3 trial of local chemotherapy with biodegradable carmustine (BCNU) wafers (Gliadel wafers) in patients with primary malignant glioma, Neuro Oncol, vol.5, pp.79-88, 2003.

P. Miglierini, M. Bouchekoua, B. Rousseau, P. Hieu, J. Malhaire et al., Impact of the peroperatory application of GLIADEL wafers (BCNU, carmustine) in combination with temozolomide and radiotherapy in patients with glioblastoma multiforme: Efficacy and toxicity, Clin Neurol Neurosurg, vol.114, pp.1222-1225, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01721429

J. N. Sarkaria, G. J. Kitange, C. D. James, R. Plummer, H. Calvert et al., Mechanisms of chemoresistance to alkylating agents in malignant glioma, Clin Cancer Res, vol.14, pp.2900-2908, 2008.

C. Bastiancich, J. Bianco, K. Vanvarenberg, B. Ucakar, N. Joudiou et al., Injectable nanomedicine hydrogel for local chemotherapy of glioblastoma after surgical resection, J Control Release, vol.264, pp.45-54, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076524

C. Bastiancich, K. Vanvarenberg, B. Ucakar, M. Pitorre, G. Bastiat et al., Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of glioblastoma, J Control Release, vol.225, pp.283-293, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388804

E. Moysan, Y. Gonzalez-fernandez, N. Lautram, J. Bejaud, G. Bastiat et al., An innovative hydrogel of gemcitabine-loaded lipid nanocapsules: when the drug is a key player of the nanomedicine structure, Soft Matter, vol.10, pp.1767-1777, 2014.

A. Gaudin, E. Song, A. R. King, J. K. Saucier-sawyer, R. Bindra et al., PEGylated squalenoyl-gemcitabine nanoparticles for the treatment of glioblastoma, Biomaterials, pp.136-144, 2016.

U. Akbar, T. Jones, J. Winestone, M. Michael, A. Shukla et al., Delivery of temozolomide to the tumor bed via biodegradable gel matrices in a novel model of intracranial glioma with resection, J Neurooncol, vol.94, pp.203-212, 2009.

J. W. Denbo, R. F. Williams, W. S. Orr, T. L. Sims, C. Y. Ng et al., Continuous local delivery of interferon-beta stabilizes tumor vasculature in an orthotopic glioblastoma xenograft resection model, vol.150, pp.497-504, 2011.

T. M. Kauer, J. L. Figueiredo, S. Hingtgen, and K. Shah, Encapsulated therapeutic stem cells implanted in the tumor resection cavity induce cell death in gliomas, Nat Neurosci, vol.15, pp.197-204, 2012.

K. J. Sweeney, M. A. Jarzabek, P. Dicker, D. F. O'brien, J. J. Callanan et al., Validation of an imageable surgical resection animal model of Glioblastoma (GBM), J Neurosci Methods, vol.233, pp.99-104, 2014.

J. Bianco, C. Bastiancich, N. Joudiou, B. Gallez, A. Rieux et al., Novel model of orthotopic U-87 MG glioblastoma resection in athymic nude mice, J Neurosci Methods, vol.284, pp.96-102, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076529

M. E. Sughrue, I. Yang, A. J. Kane, M. J. Rutkowski, S. Fang et al., Immunological considerations of modern animal models of malignant primary brain tumors, J Transl Med, vol.7, p.84, 2009.

S. E. Gould, M. R. Junttila, and F. J. De-sauvage, Translational value of mouse models in oncology drug development, Nat Med, vol.21, pp.431-439, 2015.

R. F. Barth and B. Kaur, Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas, J Neurooncol, vol.94, pp.299-312, 2009.

B. Heurtault, P. Saulnier, B. Pech, J. E. Proust, and J. P. Benoit, A novel phase inversion-based process for the preparation of lipid nanocarriers, Pharm Res, vol.19, pp.875-880, 2002.

C. Simonsson, G. Bastiat, M. Pitorre, A. S. Klymchenko, J. Bejaud et al., Inter-nanocarrier and nanocarrier-to-cell transfer assays demonstrate the risk of an immediate unloading of dye from labeled lipid nanocapsules, Eur J Pharm Biopharm, vol.98, pp.47-56, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388767

S. Wang and J. Zhou, Diffusion tensor MR imaging of rat glioma models: A correlation study of MR imaging and histology, J Comput Assist Tomogr, vol.36, pp.739-744, 2012.

D. Jeong, C. Malalis, J. A. Arrington, A. S. Field, J. W. Choi et al., Mean apparent diffusion coefficient values in defining radiotherapy planning target volumes in glioblastoma, Quant Imaging Med Surg, vol.5, pp.835-845, 2015.

I. Tkac, Z. Starcuk, I. Y. Choi, and R. Gruetter, In vivo 1H NMR spectroscopy of rat brain at 1 ms echo time, Magn Reson Med, vol.41, pp.649-656, 1999.

A. Horska and P. B. Barker, Imaging of brain tumors: MR spectroscopy and metabolic imaging, Neuroimaging Clin N Am, vol.20, pp.293-310, 2010.

W. M. Reid, A. Rolfe, D. Register, J. E. Levasseur, S. B. Churn et al., Strain-related differences after experimental traumatic brain injury in rats, J Neurotrauma, vol.27, pp.1243-1253, 2010.

M. Candolfi, J. F. Curtin, W. S. Nichols, A. G. Muhammad, G. D. King et al., Intracranial glioblastoma models in preclinical neuro-oncology: neuropathological characterization and tumor progression, J neurooncol, vol.85, pp.133-148, 2007.

E. Roger, J. C. Gimel, C. Bensley, A. S. Klymchenko, and J. P. Benoit, Lipid nanocapsules maintain full integrity after crossing a human intestinal epithelium model, J Control Release, vol.253, pp.11-18, 2017.

A. C. Groo, P. Saulnier, J. C. Gimel, J. Gravier, C. Ailhas et al., Fate of paclitaxel lipid nanocapsules in intestinal mucus in view of their oral delivery, Int J Nanomedicine, vol.8, pp.4291-4302, 2013.

V. L. Jacobs, P. A. Valdes, W. F. Hickey, and J. A. Leo, Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model, ASN neuro, vol.3, p.63, 2011.

L. A. Lampson, M. A. Lampson, and A. D. Dunne, Exploiting the lacZ reporter gene for quantitative analysis of disseminated tumor growth within the brain: use of the lacZ gene product as a tumor antigen, for evaluation of antigenic modulation, and to facilitate image analysis of tumor growth in situ, Cancer Res, vol.53, pp.176-182, 1993.

G. H. Vince, M. Bendszus, T. Schweitzer, R. H. Goldbrunner, S. Hildebrandt et al., Spontaneous regression of experimental gliomas--an immunohistochemical and MRI study of the C6 glioma spheroid implantation model, Exp Neurol, vol.190, pp.478-485, 2004.

A. Vonarbourg, A. Sapin, L. Lemaire, F. Franconi, P. Menei et al., Characterization and detection of experimental rat gliomas using magnetic resonance imaging, Magma, pp.133-139, 2004.

S. Doblas, T. He, D. Saunders, J. Hoyle, N. Smith et al., In vivo characterization of several rodent glioma models by 1H MRS, NMR in biomedicine, vol.25, pp.685-694, 2012.

S. Doblas, T. He, D. Saunders, J. Pearson, J. Hoyle et al., Glioma morphology and tumor-induced vascular alterations revealed in seven rodent glioma models by in vivo magnetic resonance imaging and angiography, J Magn Reson Imaging, vol.32, pp.267-275, 2010.

. .. Main-contributions,

. In, . Vitro, and . .. Studies,

. Animal/tumor-models:-anti-tumor and . .. Studies,

I. N. Vivo-studies:-the and . .. Resection-model,

. The, . Of-gemc12-lnc, and . .. Hydrogel,

T. Brain, . Of, . Drug, and . .. Site,

. .. Perspectives,

C. V. Rahman, S. J. Smith, P. S. Morgan, K. A. Langmack, P. A. Clarke et al., Adjuvant chemotherapy for brain tumors delivered via a novel intra-cavity moldable polymer matrix, PloS one, vol.8, p.77435, 2013.

C. Bastiancich, P. Danhier, V. Preat, and F. Danhier, Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma, J Control Release, vol.243, pp.29-42, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02076746

J. J. Olson, L. Nayak, D. R. Ormond, P. Y. Wen, and S. N. Kalkanis, The role of cytotoxic chemotherapy in the management of progressive glioblastoma, J Neurooncol, vol.118, pp.501-555, 2014.

J. Zhou, K. B. Atsina, B. T. Himes, G. W. Strohbehn, and W. M. Saltzman, Novel delivery strategies for glioblastoma, Cancer J, vol.18, pp.89-99, 2012.

C. Bastiancich, G. Bastiat, and F. Lagarce, Gemcitabine and glioblastoma: challenges and current perspectives, Drug Discov Today, 2017.
DOI : 10.1016/j.drudis.2017.10.010

URL : https://hal.archives-ouvertes.fr/hal-02076519

E. Moysan, G. Bastiat, and J. P. Benoit, Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications, Molecular Pharm, vol.10, pp.430-444, 2013.
URL : https://hal.archives-ouvertes.fr/inserm-00787112

N. T. Huynh, C. Passirani, P. Saulnier, and J. P. Benoit, Lipid nanocapsules: a new platform for nanomedicine, Int J Pharm, vol.379, pp.201-209, 2009.

E. Moysan, Y. Gonzalez-fernandez, N. Lautram, J. Bejaud, G. Bastiat et al., An innovative hydrogel of gemcitabine-loaded lipid nanocapsules: when the drug is a key player of the nanomedicine structure, Soft Matter, vol.10, pp.1767-1777, 2014.

Y. R. Lawrence, D. T. Blumenthal, D. Matceyevsky, A. A. Kanner, F. Bokstein et al., Delayed initiation of radiotherapy for glioblastoma: how important is it to push to the front (or the back) of the line?, J Neurooncol, vol.105, pp.1-7, 2011.

C. Irwin, M. Hunn, G. Purdie, and D. Hamilton, Delay in radiotherapy shortens survival in patients with high grade glioma, J Neurooncol, vol.85, pp.339-382, 2007.

E. Fournier, C. Passirani, C. N. Montero-menei, and J. P. Benoit, Biocompatibility of implantable synthetic polymeric drug carriers: focus on brain biocompatibility, pp.3311-3331, 2003.
DOI : 10.1016/s0142-9612(03)00161-3

J. Rieger, S. Durka, J. Streffer, J. Dichgans, and M. Weller, Gemcitabine cytotoxicity of human malignant glioma cells: modulation by antioxidants, BCL-2 and dexamethasone, Eur J Pharmacol, vol.365, pp.301-308, 1999.

L. J. Ostruszka and D. S. Shewach, The role of cell cycle progression in radiosensitization by 2',2'-difluoro-2'-deoxycytidine, Cancer Res, vol.60, pp.6080-6088, 2000.

M. Genc, N. Castro-kreder, A. Barten-van-rijbroek, L. J. Stalpers, and J. Haveman, Enhancement of effects of irradiation by gemcitabine in a glioblastoma cell line and cell line spheroids, J Cancer Res Clin Oncol, vol.130, pp.45-51, 2004.

G. Carpinelli, B. Bucci, I. D'agnano, R. Canese, F. Caroli et al., Gemcitabine treatment of experimental C6 glioma: the effects on cell cycle and apoptotic rate, vol.26, pp.3017-3024, 2006.

M. L. Immordino, P. Brusa, F. Rocco, S. Arpicco, M. Ceruti et al., Cattel, Preparation, characterization, cytotoxicity and pharmacokinetics of liposomes containing lipophilic gemcitabine prodrugs, J Control Release, vol.100, pp.331-346, 2004.

A. Gaudin, E. Song, A. R. King, J. K. Saucier-sawyer, R. Bindra et al., PEGylated squalenoyl-gemcitabine nanoparticles for the treatment of glioblastoma, Biomaterials, pp.136-144, 2016.

P. Brusa, M. L. Immordino, F. Rocco, and L. Cattel, Antitumor activity and pharmacokinetics of liposomes containing lipophilic gemcitabine prodrugs, Anticancer Res, vol.27, pp.195-199, 2007.

W. G. Chung, M. A. Sandoval, B. R. Sloat, P. D. Lansakara, and Z. Cui, Stearoyl gemcitabine nanoparticles overcome resistance related to the over-expression of ribonucleotide reductase subunit M1, J Control Rel, vol.157, pp.132-140, 2012.

B. R. Sloat, M. A. Sandoval, D. Li, W. G. Chung, P. D. Lansakara et al., In vitro and in vivo anti-tumor activities of a gemcitabine derivative carried by nanoparticles, Int J Pharm, vol.409, pp.278-288, 2011.

N. Wauthoz, G. Bastiat, E. Moysan, A. Cieslak, K. Kondo et al., Safe lipid nanocapsule-based gel technology to target lymph nodes and combat mediastinal metastases from an orthotopic non-small-cell lung cancer model in SCID-CB17 mice, Nanomedicine, vol.11, pp.1237-1245, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392447

C. Bastiancich, K. Vanvarenberg, B. Ucakar, M. Pitorre, G. Bastiat et al., Lauroyl-gemcitabine-loaded lipid nanocapsule hydrogel for the treatment of glioblastoma, J Control release, vol.225, pp.283-293, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01388804

X. M. Tao, J. C. Wang, J. B. Wang, Q. Feng, S. Y. Gao et al., Enhanced anticancer activity of gemcitabine coupling with conjugated linoleic acid against human breast cancer in vitro and in vivo, Eur J Pharm Biopharm, vol.82, pp.401-409, 2012.

P. D. Lansakara, B. L. Rodriguez, and Z. Cui, Synthesis and in vitro evaluation of novel lipophilic monophosphorylated gemcitabine derivatives and their nanoparticles, Int J Pharm, vol.429, pp.123-134, 2012.

R. D. Dubey, A. Saneja, P. K. Gupta, and P. N. Gupta, Recent advances in drug delivery strategies for improved therapeutic efficacy of gemcitabine, Eur J Pharm Sci, vol.93, pp.147-162, 2016.

D. P. Ivanov, B. Coyle, D. A. Walker, and A. M. Grabowska, In vitro models of medulloblastoma: Choosing the right tool for the job, J Biotechnol, vol.236, pp.10-25, 2016.

D. P. Ivanov, A. J. Al-rubai, A. M. Grabowska, and M. K. Pratten, Separating chemotherapy-related developmental neurotoxicity from cytotoxicity in monolayer and neurosphere cultures of human fetal brain cells, Toxicol In Vitro, vol.37, pp.88-96, 2016.

V. L. Jacobs, P. A. Valdes, W. F. Hickey, and J. A. Leo, Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model, ASN neuro, vol.3, p.63, 2011.

J. Bianco, C. Bastiancich, N. Joudiou, B. Gallez, A. Rieux et al., Novel model of orthotopic U-87 MG glioblastoma resection in athymic nude mice, J Neurosci Met, vol.284, pp.96-102, 2017.
URL : https://hal.archives-ouvertes.fr/hal-02076529

L. Qiang, Y. Yang, Y. J. Ma, F. H. Chen, L. B. Zhang et al., Isolation and characterization of cancer stem like cells in human glioblastoma cell lines, Cancer Lett, vol.279, pp.13-21, 2009.

M. Allen, M. Bjerke, H. Edlund, S. Nelander, and B. Westermark, Origin of the U87MG glioma cell line: Good news and bad news, Sci Transl Med, vol.8, pp.354-357, 2016.

O. Okolie, J. R. Bago, R. S. Schmid, D. M. Irvin, R. E. Bash et al., Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model, Neuro Oncol, vol.18, pp.1622-1633, 2016.

L. Zitvogel, A. Tesniere, L. Apetoh, F. Ghiringhelli, and G. Kroemer, Immunological aspects of anticancer chemotherapy, Bull Acad Natl Med, vol.192, pp.1469-1489, 2008.

I. Szadvari, O. Krizanova, and P. Babula, Athymic nude mice as an experimental model for cancer treatment, Physiol Res, vol.65, pp.441-453, 2016.

M. S. Sasso, G. Lollo, M. Pitorre, S. Solito, L. Pinton et al., Low dose gemcitabine-loaded lipid nanocapsules target monocytic myeloid-derived suppressor cells and potentiate cancer immunotherapy, Biomaterials, pp.47-62, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01392469

T. Wurdinger, K. Deumelandt, H. J. Van-der, P. Vliet, T. D. Wesseling et al., Mechanisms of intimate and long-distance cross-talk between glioma and myeloid cells: how to break a vicious cycle, Biochim Biophys Acta, pp.560-575, 2014.

A. Tuettenberg, K. Steinbrink, and D. Schuppan, Myeloid cells as orchestrators of the tumor microenvironment: novel targets for nanoparticular cancer therapy, Nanomedicine (Lond), vol.11, pp.2735-2751, 2016.

B. Raychaudhuri, P. Rayman, J. Ireland, J. Ko, B. Rini et al., Myeloid-derived suppressor cell accumulation and function in patients with newly diagnosed glioblastoma, Neuro-Oncol, vol.13, pp.591-599, 2011.

M. Candolfi, J. F. Curtin, W. S. Nichols, A. G. Muhammad, G. D. King et al., Intracranial glioblastoma models in preclinical neuro-oncology: neuropathological characterization and tumor progression, J Neurooncol, vol.85, pp.133-148, 2007.

L. A. Lampson, M. A. Lampson, and A. D. Dunne, Exploiting the lacZ reporter gene for quantitative analysis of disseminated tumor growth within the brain: use of the lacZ gene product as a tumor antigen, for evaluation of antigenic modulation, and to facilitate image analysis of tumor growth in situ, Cancer Res, vol.53, pp.176-182, 1993.

R. F. Barth and B. Kaur, Rat brain tumor models in experimental neuro-oncology: the C6, 9L, T9, RG2, F98, BT4C, RT-2 and CNS-1 gliomas, J Neurooncol, vol.94, pp.299-312, 2009.

A. Vonarbourg, A. Sapin, L. Lemaire, F. Franconi, P. Menei et al., Characterization and detection of experimental rat gliomas using magnetic resonance imaging, Magma, pp.133-139, 2004.

P. C. Huszthy, I. Daphu, S. P. Niclou, D. Stieber, J. M. Nigro et al., vivo models of primary brain tumors: pitfalls and perspectives, vol.14, pp.979-993, 2012.

S. E. Gould, M. R. Junttila, and F. J. De-sauvage, Translational value of mouse models in oncology drug development, Nat Med, vol.21, pp.431-439, 2015.

L. Janbazian, J. Karamchandani, and S. Das, Mouse models of glioblastoma: lessons learned and questions to be answered, J Neurooncol, vol.118, pp.1-8, 2014.

M. E. Sughrue, I. Yang, A. J. Kane, M. J. Rutkowski, S. Fang et al., Immunological considerations of modern animal models of malignant primary brain tumors, J Transl Med, vol.7, p.84, 2009.

E. I. Fomchenko and E. C. Holland, Mouse models of brain tumors and their applications in preclinical trials, Clin Cancer Res, vol.12, pp.5288-5297, 2006.

M. A. Vetten, C. S. Yah, T. Singh, and M. Gulumian, Challenges facing sterilization and depyrogenation of nanoparticles: Effects on structural stability and biomedical applications in preclinical trials Nanomedicine, vol.10, pp.1391-1399, 2014.

K. , A. Da, and S. Aquino, Sterilization by Gamma Irradiation, Gamma Radiation, 2012.

Y. Deng, A. Ediriwickrema, F. Yang, J. Lewis, M. Girardi et al., A sunblock based on bioadhesive nanoparticles, vol.14, pp.1278-1285, 2015.

S. Zhang, J. Ermann, M. D. Succi, A. Zhou, M. J. Hamilton et al., An inflammation-targeting hydrogel for local drug delivery in inflammatory bowel disease, Sci Transl Med, vol.7, pp.300-128, 2015.

T. Furst, M. Piette, A. Lechanteur, B. Evrard, and G. Piel, Mucoadhesive cellulosic derivative sponges as drug delivery system for vaginal application, Eur J Pharm Biopharm, vol.95, pp.128-163, 2015.

C. Zhang, P. Mastorakos, M. Sobral, S. Berry, E. Song et al., Strategies to enhance the distribution of nanotherapeutics in the brain, J Control Release, 2017.

U. M. Upadhyay, B. Tyler, Y. Patta, R. Wicks, K. Spencer et al., Intracranial microcapsule chemotherapy delivery for the localized treatment of rodent metastatic breast adenocarcinoma in the brain, Proc Natl Acad Sci U S A, vol.111, pp.16071-16076, 2014.

V. G. Roullin, J. R. Deverre, L. Lemaire, F. Hindre, M. C. Venier-julienne et al., Anti-cancer drug diffusion within living rat brain tissue: an experimental study using [3H](6)-5-fluorouracil-loaded PLGA microspheres, Eur J Pharm Biopharm, vol.53, pp.293-299, 2002.

J. Zhou, T. R. Patel, R. W. Sirianni, G. Strohbehn, M. Q. Zheng et al., Highly penetrative, drugloaded nanocarriers improve treatment of glioblastoma, Proc Natl Acad Sci U S A, vol.110, pp.11751-11756, 2013.

S. O'reilly, N. R. Hartman, S. A. Grossman, J. M. Strong, R. F. Struck et al., Tissue and tumor distribution of C-penclomedine in rats, Clin Cancer Res, vol.2, pp.541-548, 1996.

E. Nance, C. Zhang, T. Y. Shih, Q. Xu, B. S. Schuster et al., Brain-penetrating nanoparticles improve paclitaxel efficacy in malignant glioma following local administration, ACS nano, vol.8, pp.10655-10664, 2014.

P. Mastorakos, C. Zhang, E. Song, Y. E. Kim, H. W. Park et al., Biodegradable brain-penetrating DNA nanocomplexes and their use to treat malignant brain tumors, J Control Release, vol.262, pp.37-46, 2017.

E. A. Nance, G. F. Woodworth, K. A. Sailor, T. Y. Shih, Q. Xu et al., A dense poly(ethylene glycol) coating improves penetration of large polymeric nanoparticles within brain tissue, Sci Transl Med, vol.4, pp.149-119, 2012.

R. Ramachandran, V. R. Junnuthula, G. S. Gowd, A. Ashokan, J. Thomas et al., Theranostic 3-Dimensional nano brain-implant for prolonged and localized treatment of recurrent glioma, Sci Rep, vol.7, p.43271, 2017.

J. L. Mcfaline-figueroa, C. J. Braun, M. Stanciu, Z. D. Nagel, P. Mazzucato et al., Minor changes in expression of the mismatch repair protein MSH2 exert a major impact on glioblastoma response to temozolomide, Cancer Res, 2015.

J. Felsberg, N. Thon, S. Eigenbrod, B. Hentschel, M. C. Sabel et al., Promoter methylation and expression of MGMT and the DNA mismatch repair genes MLH1, MSH2, MSH6 and PMS2 in paired primary and recurrent glioblastomas, Int J Cancer, vol.129, pp.659-670, 2011.

A. D. Adema, K. Van-der-born, R. J. Honeywell, and G. J. Peters, Cell cycle effects and increased adduct formation by temozolomide enhance the effect of cytotoxic and targeted agents in lung cancer cell lines, J Chemother, vol.21, pp.338-384, 2009.

B. L. Ebert, E. Niemierko, K. Shaffer, and R. Salgia, Use of temozolomide with other cytotoxic chemotherapy in the treatment of patients with recurrent brain metastases from lung cancer, Oncologist, vol.8, pp.69-75, 2003.

S. Peltier, J. M. Oger, F. Lagarce, W. Couet, and J. P. Benoit, Enhanced oral paclitaxel bioavailability after administration of paclitaxel-loaded lipid nanocapsules, Pharm Res, vol.23, pp.1243-1250, 2006.

J. Hureaux, F. Lagarce, F. Gagnadoux, L. Vecellio, A. Clavreul et al., Lipid nanocapsules: ready-to-use nanovectors for the aerosol delivery of paclitaxel, Eur J Pharm Biopharm, vol.73, pp.239-246, 2009.

E. Garcion, A. Lamprecht, B. Heurtault, A. Paillard, A. Aubert-pouessel et al., A new generation of anticancer, drug-loaded, colloidal vectors reverses multidrug resistance in glioma and reduces tumor progression in rats, Mol Cancer Ther, issue.5, pp.1710-1722, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00129865

P. B. Gupta, T. T. Onder, G. Jiang, K. Tao, C. Kuperwasser et al., Identification of selective inhibitors of cancer stem cells by high-throughput screening, Cell, vol.138, pp.645-659, 2009.

R. S. Tigli-aydin, G. Kaynak, and M. Gumusderelioglu, Salinomycin encapsulated nanoparticles as a targeting vehicle for glioblastoma cells, J Biomed Mater Res A, vol.104, pp.455-464, 2016.

E. Xipell, T. Aragon, N. Martinez-velez, B. Vera, M. A. Idoate et al., Endoplasmic reticulum stress-inducing drugs sensitize glioma cells to temozolomide through downregulation of MGMT, MPG, and Rad51, Neuro Oncol, vol.18, pp.1109-1119, 2016.

E. Xipell, M. Gonzalez-huarriz, J. J. Martinez-de-irujo, A. Garcia-garzon, F. F. Lang et al., Salinomycin induced ROS results in abortive autophagy and leads to regulated necrosis in glioblastoma, vol.7, pp.30626-30641, 2016.

G. N. Zhang, Y. Liang, L. J. Zhou, S. P. Chen, G. Chen et al., Combination of salinomycin and gemcitabine eliminates pancreatic cancer cells, Cancer letters, vol.313, pp.137-144, 2011.

Z. Daman, H. Montazeri, M. Azizi, F. Rezaie, S. N. Ostad et al., Polymeric Micelles of PEG-PLA Copolymer as a Carrier for Salinomycin Against Gemcitabine-Resistant Pancreatic Cancer, Pharm Res, vol.32, pp.3756-3767, 2015.

A. Huczynski, Salinomycin: a new cancer drug candidate, Chem Biol Drug Des, vol.79, pp.235-238, 2012.

J. Balzeau, M. Pinier, R. Berges, P. Saulnier, J. Benoit et al., The effect of functionalizing lipid nanocapsules with NFL-TBS.40-63 peptide on their uptake by glioblastoma cells, Biomaterials, vol.34, pp.3381-3389, 2013.

R. Berges, J. Balzeau, A. C. Peterson, and J. Eyer, A tubulin binding peptide targets glioma cells disrupting their microtubules, blocking migration, and inducing apoptosis, Mol Ther, vol.20, pp.1367-1377, 2012.

A. Tivnan, T. Heilinger, E. C. Lavelle, and J. H. Prehn, Advances in immunotherapy for the treatment of glioblastoma, J Neurooncol, pp.1-9, 2017.

J. H. Sampson, M. V. Maus, and C. H. June, Immunotherapy for Brain Tumors, J Clin Oncol, vol.35, pp.2450-2456, 2017.
DOI : 10.1200/jco.2017.72.8089

P. Perng and M. Lim, Immunosuppressive Mechanisms of Malignant Gliomas: Parallels at Non-CNS Sites, Front Oncol, vol.5, p.153, 2015.

G. Sciumè, A. Santoni, and G. Bernardini, Chemokines and glioma: invasion and more, J Neuroimmunol, vol.224, pp.8-12, 2010.

E. Suzuki, J. Sun, V. Kapoor, A. S. Jassar, and S. M. Albelda, Gemcitabine has significant immunomodulatory activity in murine tumor models independent of its cytotoxic effects, Cancer Biol Ther, vol.6, pp.880-885, 2007.

L. Galluzzi, L. Senovilla, L. Zitvogel, and G. Kroemer, The secret ally: immunostimulation by anticancer drugs, Nat Rev Drug Discov, vol.11, pp.215-248, 2012.

O. Draghiciu, J. Lubbers, H. W. Nijman, and T. Daemen, Myeloid derived suppressor cells-An overview of combat strategies to increase immunotherapy efficacy, Oncoimmunology, vol.4, p.954829, 2015.

D. Mathios, J. E. Kim, A. Mangraviti, J. Phallen, C. K. Park et al.,

. Lim, Anti-PD-1 antitumor immunity is enhanced by local and abrogated by systemic chemotherapy in GBM, Sci Transl Med, vol.8, pp.370-180, 2016.

K. L. Chaichana, E. E. Cabrera-aldana, I. Jusue-torres, O. Wijesekera, A. Olivi et al., When Gross Total Resection of a Glioblastoma Is Possible, How Much Resection Should Be Achieved?, World neurosurgery, vol.82, pp.257-265, 2014.

J. K. Shah, M. B. Potts, P. K. Sneed, M. K. Aghi, and M. W. Mcdermott, Surgical Cavity Constriction and Local Progression Between Resection and Adjuvant Radiosurgery for Brain Metastases, Cureus, vol.8, p.575, 2016.
DOI : 10.7759/cureus.575

URL : https://doi.org/10.7759/cureus.575

P. Ferroli, M. Broggi, A. Franzini, E. Maccagnano, M. Lamperti et al., Surgifoam and mitoxantrone in the glioblastoma multiforme postresection cavity: the first step of locoregional chemotherapy through an ad hoc-placed catheter: technical note, Neurosurgery, vol.59, pp.433-434, 2006.

K. O. Learned, S. Mohan, I. Z. Hyder, L. J. Bagley, S. Wang et al., Imaging features of a gelatin-thrombin matrix hemostatic agent in the intracranial surgical bed: a unique space-occupying pseudomass, AJNR Am J Neuroradiol, vol.35, pp.686-690, 2014.