Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

General Review Article

Current Developments in Targeted Drug Delivery Systems for Glioma

Author(s): Dhrumi Patel, Sarika Wairkar* and Mayur C. Yergeri

Volume 26, Issue 32, 2020

Page: [3973 - 3984] Pages: 12

DOI: 10.2174/1381612826666200424161929

Price: $65

Abstract

Background: Glioma is one of the most commonly observed tumours, representing about 75% of brain tumours in the adult population. Generally, glioma treatment includes surgical resection followed by radiotherapy and chemotherapy. The current chemotherapy for glioma involves the use of temozolomide, doxorubicin, monoclonal antibodies, etc. however, the clinical outcomes in patients are not satisfactory. Primarily, the blood-brain barrier hinders these drugs from reaching the target leading to the recurrence of glioma post-surgery. In addition, these drugs are not target-specific and affect the healthy cells of the body. Therefore, glioma-targeted drug delivery is essential to reduce the rate of recurrence and treat the condition with more reliable alternatives.

Methods: A literature search was conducted to understand glioma pathophysiology, its current therapeutic approaches for targeted delivery using databases like Pub Med, Web of Science, Scopus, and Google Scholar, etc.

Results: This review gives an insight to challenges associated with current treatments, factors influencing drug delivery in glioma, and recent advancements in targeted drug delivery.

Conclusion: The promising results could be seen with nanotechnology-based approaches, like polymeric, lipidbased, and hybrid nanoparticles in the treatment of glioma. Biotechnological developments, such as carrier peptides and gene therapy, are future prospects in glioma therapy. Therefore, these targeted delivery systems will be beneficial in clinical practices for glioma treatment.

Keywords: Carrier peptides, drug targeting, gene therapy, glioma, nanoparticles, therapeutic.

[1]
Dirks PB. Brain tumor stem cells: Bringing order to the chaos of brain cancer. J Clin Oncol 2008; 26(17): 2916-24.
[http://dx.doi.org/10.1200/JCO.2008.17.6792] [PMID: 18539973]
[2]
DeAngelis LM. Brain tumors. N Engl J Med 2001; 344(2): 114-23.
[http://dx.doi.org/10.1056/NEJM200101113440207] [PMID: 11150363]
[3]
Arvold ND, Lee EQ, Mehta MP, et al. Updates in the management of brain metastases. Neuro-oncol 2016; 18(8): 1043-65.
[http://dx.doi.org/10.1093/neuonc/now127] [PMID: 27382120]
[4]
Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2011-2015 Neuro-oncol 2018; 20(suppl_4): iv1-86.
[http://dx.doi.org/10.1093/neuonc/noy131]
[5]
Patchell RA. The management of brain metastases. Cancer Treat Rev 2003; 29(6): 533-40.
[http://dx.doi.org/10.1016/S0305-7372(03)00105-1] [PMID: 14585263]
[6]
Dirks PB. Cancer: stem cells and brain tumours. Nature 2006; 444(7120): 687-8.
[http://dx.doi.org/10.1038/444687a] [PMID: 17151644]
[7]
Diwanji TP, Engelman A, Snider JW, Mohindra P. Epidemiology, diagnosis, and optimal management of glioma in adolescents and young adults. Adolesc Health Med Ther 2017; 8: 99-113.
[http://dx.doi.org/10.2147/AHMT.S53391] [PMID: 28989289]
[8]
Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet 2018; 392(10145): 432-46.
[http://dx.doi.org/10.1016/S0140-6736(18)30990-5]
[9]
Persaud-Sharma D, Burns J, Trangle J, Moulik S. Disparities in brain cancer in the United States: A literature review of gliomas. Med Sci (Basel) 2017; 5(3): 16.
[http://dx.doi.org/10.3390/medsci5030016] [PMID: 29099032]
[10]
Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007; 114(2): 97-109.
[http://dx.doi.org/10.1007/s00401-007-0243-4] [PMID: 17618441]
[11]
Heron M. Deaths: Leading causes for 2015. National Vital Statistics Reports 2015; 66. Available from: https://www.cdc
[12]
Chen Y, Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev 2012; 64(7): 640-65.
[http://dx.doi.org/10.1016/j.addr.2011.11.010]
[13]
Stupp R, Hegi ME. Targeting brain-tumor stem cells. Nat Biotechnol 2007; 25(2): 193-4.
[http://dx.doi.org/10.1038/nbt0207-193] [PMID: 17287755]
[14]
Ricard D, Idbaih A, Ducray F, Lahutte M, Hoang-Xuan K, Delattre JY. Primary brain tumours in adults. Lancet 2012; 379(9830): 1984-96.
[http://dx.doi.org/10.1016/S0140-6736(11)61346-9] [PMID: 22510398]
[15]
Gladson CL, Prayson RA, Liu WM. The pathobiology of glioma tumors. Annu Rev Pathol 2010; 5(1): 33-50.
[http://dx.doi.org/10.1146/annurev-pathol-121808-102109] [PMID: 19737106]
[16]
Posti JP, Bori M, Kauko T, et al. Presenting symptoms of glioma in adults. Acta Neurol Scand 2015; 131(2): 88-93.
[http://dx.doi.org/10.1111/ane.12285] [PMID: 25263022]
[17]
McKinney PA. Brain tumours: incidence, survival, and aetiology. J Neurol Neurosurg Psychiatry 2004; 75(2)(Suppl. 2): ii12-7.
[PMID: 15146034]
[18]
Johung T, Monje M. Neuronal activity in the glioma microenvironment. Curr Opin Neurobiol 2017; 47: 156-61.
[http://dx.doi.org/10.1016/j.conb.2017.10.009]
[19]
Venkatesh HS, Johung TB, Caretti V, et al. Neuronal activity promotes glioma growth through neuroligin-3 secretion. Cell 2015; 161(4): 803-16.
[http://dx.doi.org/10.1016/j.cell.2015.04.012] [PMID: 25913192]
[20]
Li G, Qin Z, Chen Z, Xie L, Wang R, Zhao H. Tumor microenvironment in treatment of glioma. Open Med (Wars) 2017; 12(1): 247-51.
[http://dx.doi.org/10.1515/med-2017-0035] [PMID: 28828406]
[21]
Irvin DM, McNeill RS, Bash RE, Miller CR. Intrinsic astrocyte heterogeneity influences tumor growth in glioma mouse models. Brain Pathol 2017; 27(1): 36-50.
[http://dx.doi.org/10.1111/bpa.12348] [PMID: 26762242]
[22]
Lin Q, Liu Z, Ling F, Xu G. Astrocytes protect glioma cells from chemotherapy and upregulate survival genes via gap junctional communication. Mol Med Rep 2016; 13(2): 1329-35.
[http://dx.doi.org/10.3892/mmr.2015.4680] [PMID: 26676970]
[23]
Chen W, Wang D, Du X, et al. Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Med Oncol 2015; 32(3): 43.
[http://dx.doi.org/10.1007/s12032-015-0487-0] [PMID: 25631631]
[24]
Chen W, Xia T, Wang D, et al. Human astrocytes secrete IL-6 to promote glioma migration and invasion through upregulation of cytomembrane MMP14. Oncotarget 2016; 7(38): 62425-38.
[http://dx.doi.org/10.18632/oncotarget.11515] [PMID: 27613828]
[25]
Kamran N, Kadiyala P, Saxena M, et al. Immunosuppressive myeloid cells’ blockade in the glioma microenvironment enhances the efficacy of immune-stimulatory gene therapy. Mol Ther 2017; 25(1): 232-48.
[http://dx.doi.org/10.1016/j.ymthe.2016.10.003] [PMID: 28129117]
[26]
Kundu S, Xiong A, Spyrou A, et al. Heparanase promotes glioma progression and is inversely correlated with patient survival. Mol Cancer Res 1397; 14(12): 43-53>.
[http://dx.doi.org/10.1158/1541-7786.MCR-16-0223]
[27]
Yao Y, Ye H, Qi Z, et al. B7-H4(B7x)-mediated cross-talk between glioma-initiating cells and macrophages via the IL6/JAK/STAT3 pathway lead to poor prognosis in glioma patients. Clin Cancer Res 2016; 22(11): 2778-90.
[http://dx.doi.org/10.1158/1078-0432.CCR-15-0858] [PMID: 27001312]
[28]
Xia S, Lal B, Tung B, Wang S, Goodwin CR, Laterra J. Tumor microenvironment tenascin-C promotes glioblastoma invasion and negatively regulates tumor proliferation. Neuro-oncol 2016; 18(4): 507-17.
[http://dx.doi.org/10.1093/neuonc/nov171] [PMID: 26320116]
[29]
Grimaldi A, D’Alessandro G, Golia MT, et al. KCa3.1 inhibition switches the phenotype of glioma-infiltrating microglia/macrophages. Cell Death Dis 2016; 7e2174
[http://dx.doi.org/10.1038/cddis.2016.73] [PMID: 27054329]
[30]
He Q, Zou X, Duan D, Liu Y, Xu Q. Malignant transformation of bone marrow stromal cells induced by the brain glioma niche in rats. Mol Cell Biochem 2016; 412(1-2): 1-10.
[http://dx.doi.org/10.1007/s11010-015-2602-0] [PMID: 26590986]
[31]
Zhai M, Wang Y, Zhang L, et al. Glioma targeting peptide modified apoferritin nanocage. Drug Deliv 2018; 25(1): 1013-24.
[http://dx.doi.org/10.1080/10717544.2018.1464082] [PMID: 29726297]
[32]
Nam L, Coll C, Erthal LCS, et al. Drug delivery nanosystems for the localized treatment of glioblastoma multiforme. Materials (Basel) 2018; 11(5): 779.
[http://dx.doi.org/10.3390/ma11050779] [PMID: 29751640]
[33]
Pardridge WM. The blood-brain barrier: bottleneck in brain drug development. NeuroRx 2005; 2(1): 3-14.
[http://dx.doi.org/10.1602/neurorx.2.1.3] [PMID: 15717053]
[34]
Pardridge WM. Molecular Trojan horses for blood-brain barrier drug delivery. Curr Opin Pharmacol 2006; 6(5): 494-500.
[http://dx.doi.org/10.1016/j.coph.2006.06.001] [PMID: 16839816]
[35]
Pardridge WM. Blood-brain barrier delivery. Drug Discov Today 2007; 12(1-2): 54-61.
[http://dx.doi.org/10.1016/j.drudis.2006.10.013] [PMID: 17198973]
[36]
Gloor SM, Wachtel M, Bolliger MF, Ishihara H, Landmann R, Frei K. Molecular and cellular permeability control at the blood-brain barrier. Brain Res Brain Res Rev 2001; 36(2-3): 258-64.
[http://dx.doi.org/10.1016/S0165-0173(01)00102-3] [PMID: 11690623]
[37]
El-Habashy SE, Nazief AM, Adkins CE, et al. Novel treatment strategies for brain tumors and metastases. Pharm Pat Anal 2014; 3(3): 279-96.
[http://dx.doi.org/10.4155/ppa.14.19] [PMID: 24998288]
[38]
Torchilin VP. Passive and active drug targeting: drug delivery to tumors as an example. Handb Exp Pharmacol 2010; (197): 3-53.http://link.springer.com/10.1007/978-3-642-00477-3
[http://dx.doi.org/10.1007/978-3-642-00477-3_1] [PMID: 20217525]
[39]
Alibolandi M, Charbgoo F, Taghdisi SM, Abnous K, Ramezani M. Active targeted nanoscale delivery systems for brain tumor therapeutics. Nanotechnology-based targeted drug delivery systems for brain tumors. Elsevier Inc. 2018; 75-110.
[http://dx.doi.org/10.1016/B978-0-12-812218-1.00004-X]
[40]
Pinto MP, Arce M, Yameen B, Vilos C. Targeted brain delivery nanoparticles for malignant gliomas. Nanomedicine (Lond) 2017; 12(1): 59-72.
[http://dx.doi.org/10.2217/nnm-2016-0307] [PMID: 27876436]
[41]
Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem Soc Rev 2012; 41(7): 2971-3010.
[http://dx.doi.org/10.1039/c2cs15344k] [PMID: 22388185]
[42]
Xu X, Ho W, Zhang X, Bertrand N, Farokhzad O. Cancer nanomedicine: From targeted delivery to combination therapy. Trends Mol Med 2015; 21(4): 223-32.
[http://dx.doi.org/10.1016/j.molmed.2015.01.001] [PMID: 25656384]
[43]
Öcal H, Arica-Yegin B, Vural I, Goracinova K, Caliş S. 5-Fluorouracil-loaded PLA/PLGA PEG-PPG-PEG polymeric nanoparticles: formulation, in vitro characterization and cell culture studies. Drug Dev Ind Pharm 2014; 40(4): 560-7.
[http://dx.doi.org/10.3109/03639045.2013.775581] [PMID: 23596973]
[44]
Karlsson J, Vaughan HJ, Green JJ. Biodegradable polymeric nanoparticles for therapeutic cancer treatments. Annu Rev Chem Biomol Eng 2018; 9: 105-27.
[http://dx.doi.org/10.1146/annurev-chembioeng-060817-084055] [PMID: 29579402]
[45]
Stephen ZR, Kievit FM, Veiseh O, et al. Redox-responsive magnetic nanoparticle for targeted convection-enhanced delivery of O6-benzylguanine to brain tumors. ACS Nano 2014; 8(10): 10383-95.
[http://dx.doi.org/10.1021/nn503735w] [PMID: 25247850]
[46]
Qian L, Zheng J, Wang K, et al. Cationic core-shell nanoparticles with carmustine contained within O6-benzylguanine shell for glioma therapy. Biomaterials 2013; 34(35): 8968-78.
[http://dx.doi.org/10.1016/j.biomaterials.2013.07.097] [PMID: 23953782]
[47]
Yin Y, Fu C, Li M, et al. A pH-sensitive hyaluronic acid prodrug modified with lactoferrin for glioma dual-targeted treatment. Mater Sci Eng C 2016; 67: 159-69.
[http://dx.doi.org/10.1016/j.msec.2016.05.012] [PMID: 27287110]
[48]
Müller RH, Mäder K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery - a review of the state of the art. Eur J Pharm Biopharm 2000; 50(1): 161-77.
[http://dx.doi.org/10.1016/S0939-6411(00)00087-4] [PMID: 10840199]
[49]
Saupe A, Gordon KC, Rades T. Structural investigations on nanoemulsions, solid lipid nanoparticles and nanostructured lipid carriers by cryo-field emission scanning electron microscopy and Raman spectroscopy. Int J Pharm 2006; 314(1): 56-62.
[http://dx.doi.org/10.1016/j.ijpharm.2006.01.022] [PMID: 16574354]
[50]
Brioschi A, Zenga F, Zara GP, Gasco MR, Ducati A, Mauro A. Solid lipid nanoparticles: could they help to improve the efficacy of pharmacologic treatments for brain tumors? Neurol Res 2007; 29(3): 324-30.
[http://dx.doi.org/10.1179/016164107X187017] [PMID: 17509234]
[51]
Banerjee I, De K, Mukherjee D, et al. Paclitaxel-loaded solid lipid nanoparticles modified with Tyr-3-octreotide for enhanced anti-angiogenic and anti-glioma therapy. Acta Biomater 2016; 38: 69-81.
[http://dx.doi.org/10.1016/j.actbio.2016.04.026] [PMID: 27109765]
[52]
Vijayakumar MR, Kumari L, Patel KK, et al. Intravenous administration of trans -resveratrol-loaded TPGS-coated solid lipid nanoparticles for prolonged systemic circulation, passive brain targeting and improved in vitro cytotoxicity against C6 glioma cell lines. RSC Advances 2016; 6(55): 50336-48.
[http://dx.doi.org/10.1039/C6RA10777J]
[53]
Mukherjee A, Waters AK, Kalyan P, Achrol AS, Kesari S, Yenugonda VM. Lipid-polymer hybrid nanoparticles as a next-generation drug delivery platform: state of the art, emerging technologies, and perspectives. Int J Nanomedicine 2019; 14: 1937-52.
[http://dx.doi.org/10.2147/IJN.S198353] [PMID: 30936695]
[54]
Zhang L, Chan JM, Gu FX, et al. Self-assembled lipid--polymer hybrid nanoparticles: A robust drug delivery platform. ACS Nano 2008; 2(8): 1696-702.
[http://dx.doi.org/10.1021/nn800275r] [PMID: 19206374]
[55]
Wakaskar RR. General overview of lipid-polymer hybrid nanoparticles, dendrimers, micelles, liposomes, spongosomes and cubosomes. J Drug Target 2018; 26(4): 311-8.
[http://dx.doi.org/10.1080/1061186X.2017.1367006] [PMID: 28797169]
[56]
Sekerdag E, Lüle S, Bozdağ Pehlivan S, et al. A potential non-invasive glioblastoma treatment: Nose-to-brain delivery of farnesylthiosalicylic acid incorporated hybrid nanoparticles. J Control Release 2017; 261: 187-98.
[http://dx.doi.org/10.1016/j.jconrel.2017.06.032] [PMID: 28684169]
[57]
Yao C, Wu M, Zhang C, et al. Photoresponsive lipid-polymer hybrid nanoparticles for controlled doxorubicin release. Nanotechnology 2017; 28(25)255101
[http://dx.doi.org/10.1088/1361-6528/aa702a] [PMID: 28561013]
[58]
Zhang F, Xu CL, Liu CM. Drug delivery strategies to enhance the permeability of the blood-brain barrier for treatment of glioma. Drug Des Devel Ther 2015; 9: 2089-100.
[http://dx.doi.org/10.2147/DDDT.S79592] [PMID: 25926719]
[59]
Wanjale MV, Kumar GSV. Peptides as a therapeutic avenue for nanocarrier-aided targeting of glioma. Expert Opin Drug Deliv 2017; 14(6): 811-24.
[http://dx.doi.org/10.1080/17425247.2017.1242574] [PMID: 27690671]
[60]
Bellis SL. Advantages of RGD peptides for directing cell association with biomaterials. Biomaterials 2011; 32(18): 4205-10.
[http://dx.doi.org/10.1016/j.biomaterials.2011.02.029] [PMID: 21515168]
[61]
Zeng L, Zou L, Yu H, et al. Treatment of malignant brain tumor by tumor-triggered programmed wormlike micelles with precise targeting and deep penetration. Adv Funct Mater 2016; 26(23): 4201-12.
[http://dx.doi.org/10.1002/adfm.201600642]
[62]
Shi K, Zhou J, Zhang Q, et al. Arginine-glycine-aspartic acid-modified lipid-polymer hybrid nanoparticles for docetaxel delivery in glioblastoma multiforme. J Biomed Nanotechnol 2015; 11(3): 382-91.
[http://dx.doi.org/10.1166/jbn.2015.1965] [PMID: 26307822]
[63]
Taylor TE, Furnari FB, Cavenee WK. Targeting EGFR for treatment of glioblastoma: molecular basis to overcome resistance. Curr Cancer Drug Targets 2012; 12(3): 197-209.
[http://dx.doi.org/10.2174/156800912799277557] [PMID: 22268382]
[64]
Dixit S, Miller K, Zhu Y, et al. Dual receptor-targeted theranostic nanoparticles for localized delivery and activation of photodynamic therapy drug in glioblastomas. Mol Pharm 2015; 12(9): 3250-60.
[http://dx.doi.org/10.1021/acs.molpharmaceut.5b00216] [PMID: 26198693]
[65]
Wei X, Zhan C, Chen X, Hou J, Xie C, Lu W. Retro-inverso isomer of Angiopep-2: A stable d-peptide ligand inspires brain-targeted drug delivery. Mol Pharm 2014; 11(10): 3261-8.
[http://dx.doi.org/10.1021/mp500086e] [PMID: 24673510]
[66]
Lu F, Pang Z, Zhao J, et al. Angiopep-2-conjugated poly(ethylene glycol)-co- poly(ε-caprolactone) polymersomes for dual-targeting drug delivery to glioma in rats. Int J Nanomedicine 2017; 12: 2117-27.
[http://dx.doi.org/10.2147/IJN.S123422] [PMID: 28356732]
[67]
Ren J, Shen S, Wang D, et al. The targeted delivery of anticancer drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with angiopep-2 Biomaterials 2012; 33(11): 3324-33.
[http://dx.doi.org/10.1016/j.biomaterials.2012.01.025]
[68]
Dardevet L, Rani D, Aziz TA, et al. Chlorotoxin: a helpful natural scorpion peptide to diagnose glioma and fight tumor invasion. Toxins (Basel) 2015; 7(4): 1079-101.
[http://dx.doi.org/10.3390/toxins7041079] [PMID: 25826056]
[69]
Fang C, Wang K, Stephen ZR, et al. Temozolomide nanoparticles for targeted glioblastoma therapy. ACS Appl Mater Interfaces 2015; 7(12): 6674-82.
[http://dx.doi.org/10.1021/am5092165] [PMID: 25751368]
[70]
Feng X, Gao X, Kang T, et al. Mammary-derived growth inhibitor targeting peptide-modified PEG-PLA nanoparticles for enhanced targeted glioblastoma therapy. Bioconjug Chem 2015; 26(8): 1850-61.
[http://dx.doi.org/10.1021/acs.bioconjchem.5b00379] [PMID: 26222392]
[71]
Tobias A, Ahmed A, Moon KS, Lesniak MS. The art of gene therapy for glioma: a review of the challenging road to the bedside. J Neurol Neurosurg Psychiatry 2013; 84(2): 213-22.
[http://dx.doi.org/10.1136/jnnp-2012-302946] [PMID: 22993449]
[72]
Chiarelli PA, Kievit FM, Zhang M, Ellenbogen RG. Bionanotechnology and the future of glioma. Surg Neurol Int 2015; 6(2)(Suppl. 1): S45-58.
[http://dx.doi.org/10.4103/2152-7806.151334] [PMID: 25722933]
[73]
Kwiatkowska A, Nandhu MS, Behera P, Chiocca EA, Viapiano MS. Strategies in gene therapy for glioblastoma. Cancers (Basel) 2013; 5(4): 1271-305.
[http://dx.doi.org/10.3390/cancers5041271] [PMID: 24202446]
[74]
Kuang Y, An S, Guo Y, et al. T7 peptide-functionalized nanoparticles utilizing RNA interference for glioma dual targeting. Int J Pharm 2013; 454(11): 11-20.
[http://dx.doi.org/10.1016/j.ijpharm.2013.07.019]
[75]
Son S, Hwang DW, Singha K, et al. RVG peptide tethered bioreducible polyethylenimine for gene delivery to brain. J Control Release 2011; 155(1): 18-25.
[http://dx.doi.org/10.1016/j.jconrel.2010.08.011] [PMID: 20800628]
[76]
Chen Y, Huang H, Yao C, et al. Antitumor activity of combined endostatin and thymidine kinase gene therapy in C6 glioma models. Cancer Med 2016; 5(9): 2477-86.
[http://dx.doi.org/10.1002/cam4.798] [PMID: 27366865]
[77]
Portnow J, Synold TW, Badie B, et al. Neural stem cell-based anticancer gene therapy: A first-in-human study in recurrent high-grade glioma patients. Clin Cancer Res 2017; 23(12): 2951-60.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-1518] [PMID: 27979915]
[78]
Bush NAO, Chang SM, Berger MS. Current and future strategies for treatment of glioma. Neurosurg Rev 2017; 40(1): 1-14.
[http://dx.doi.org/10.1007/s10143-016-0709-8] [PMID: 27085859]
[79]
Robins HI, Lassman AB, Khuntia D. Therapeutic advances in malignant glioma: Current status and future prospects. Neuroimaging Clin N Am 2009; 19(4): 647-56.
[http://dx.doi.org/10.1016/j.nic.2009.08.015] [PMID: 19959010]
[80]
NCT00734682. Bethesda (MD): National Library of Medicine (US). A phase I trial of nanoliposomal CPT-11 (NL CPT-11) in patients with recurrent high-grade gliomas 2015. Avaialble at:. https://www.clinicaltrials.gov/ct2/show/NCT00734682
[81]
NCT00004041. Bethesda (MD): National Library of Medicine (US). June 27, 2018, Gene therapy in treating patients with recurrent malignant gliomas 2018. Available at: . https://clinicaltrials.gov/ct2/show/NCT00004041
[82]
NCT02340156. Bethesda (MD): National Library of Medicine (US), Phase II study of combined temozolomide and sgt-53 for treatment of recurrent glioblastoma. 2019. Available at: . https://www.clinicaltrials.gov/ct2/show/NCT02340156
[83]
NCT03463265. Bethesda (MD): National Library of Medicine (US). ABI-009 (Nab-Rapamycin) in recurrent high grade glioma and newly diagnosed glioblastoma 2019. Available at: . https://clinicaltrials.gov/ct2/show/NCT03463265
[84]
NCT03566199. Bethesda (MD): National Library of Medicine (US). MTX110 by convection-enhanced delivery in treating participants with newly-diagnosed diffuse intrinsic pontine glioma (PNOC015) 2020. Available at: . https://clinicaltrials.gov/ct2/ show/NCT03566199
[85]
NCT04264143. Bethesda (MD): National Library of Medicine (US). CED of MTX110 newly diagnosed diffuse midline gliomas 2020. Avaialble at: . https://clinicaltrials.gov/ ct2/ show/NCT04264143
[86]
NCT04099797. Bethesda (MD): National Library of Medicine (US), C7R-GD2CAR t cells for patients with gd2-expressing brain tumors (GAIL-B). 2020. Avaialble from:. https://clinicaltrials.gov/ ct2/ show/ NCT04099797
[87]
Kamran N, Alghamri MS, Nunez FJ, et al. Current state and future prospects of immunotherapy for glioma. Immunotherapy 2018; 10(4): 317-39.
[http://dx.doi.org/10.2217/imt-2017-0122] [PMID: 29421984]
[88]
Freedman LP, Gibson MC, Ethier SP, Soule HR, Neve RM, Reid YA. Reproducibility: Changing the policies and culture of cell line authentication. Nat Methods 2015; 12(6): 493-7.
[http://dx.doi.org/10.1038/nmeth.3403] [PMID: 26020501]
[89]
Lenting K, Verhaak R, Ter Laan M, Wesseling P, Leenders W. Glioma: Experimental models and reality. Acta Neuropathol 2017; 133(2): 263-82.
[http://dx.doi.org/10.1007/s00401-017-1671-4] [PMID: 28074274]
[90]
Reardon DA, Wen PY. Glioma in 2014: unravelling tumour heterogeneity-implications for therapy. Nat Rev Clin Oncol 2015; 12(2): 69-70.
[http://dx.doi.org/10.1038/nrclinonc.2014.223] [PMID: 25560529]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy