Review

Nanoparticles drug-delivery systems and antiangiogenic approaches in the treatment of gliomas

Caffo, Maria1,; Cardali, Salvatore Massimo1; Fazzari, Elena1; Barresi, Valeria2; Caruso, Gerardo1

Author Information
Glioma 1(6):p 183-188, Nov–Dec 2018. | DOI: 10.4103/glioma.glioma_43_18
  • Open

Abstract

The prognosis of patients with cerebral gliomas remains noticeably poor. Total surgical resection is almost unachievable due to considerable infiltrative ability of glial cells. Furthermore, adjuvant treatments are burdened by considerable limitations. Angiogenesis is the mechanism by which new blood vessels are formed from preexisting ones, thus supporting neoplasm progression. Gliomas are characterized by extensive microvascular proliferation. The extent of neovascularization in brain tumor correlates directly with the biological aggressiveness, degree of malignancy, and clinical recurrence of the tumor. Although a plethora of molecules can act as inducers of angiogenesis, the major growth factors include members of the vascular endothelium growth factor family. The new therapeutic approaches envisage the identification of specific biomarkers involved in this process and try to inhibit them, thus slowing down the neoplastic progression. Nanoparticles (NPs) show the ability to pass the blood–brain barrier, and moreover, when suitably modified, they can bind to specific overexpressed receptors in the glial cells. As carriers, they are able to protect the therapeutic agent and allow their sustained release. In this review, we describe some NP delivery systems which _target specific biomarkers to intervene in the process of angiogenesis.

INTRODUCTION

Gliomas are the most frequent primary malignant brain tumors in adults. Their incidence is approximately 81% of all malignant brain tumors and 45% of all primary central nervous system (CNS) tumors.[1] The World Health Organization classification divides brain tumors according to the constituent cells of the CNS, such as astrocytes, oligodendrocytes, and ependymal cells, and further sub-classifies according to the presumed level of differentiation, which is determined based on morphological abnormalities. Recently, genetic/epigenetic features that can identify numerous tumor histotypes have also been included.[2] Glioblastoma multiforme (GBM), the most malignant occurring type of glioma, shows an incidence of around 60% of all brain tumors in adults.[3] Patients show a median survival of 14–15 months from the diagnosis.[4] The choice of optimal treatment depends on several factors, such as size of the lesion, patient's age, and neurological status.[5] The standard treatment of gliomas is multimodal that includes surgical resection, radiotherapy, and chemotherapy. The extent of resection is considered a prognostic factor. However, radical resection is not always achievable, both due to the extensive infiltration of the tumor and as an attempt to preserve the functional areas. Several techniques have been designed to improve tumor detection and to increase the possibility of total tumor resection, such as neuronavigation, use of 5-aminolevulinic acid, and intraoperative magnetic resonance imaging (MRI). There is evidence that the combined use of these techniques improves the success rate of complete resection of tumors to 96.2%.[6] Radiotherapy and chemotherapy are related to important side effects, such as postradiation leukoencephalopathy, nerve damage, hair loss, vomiting, infertility, and skin rash. Moreover, the effectiveness of chemotherapy is limited by various factors such as toxic effects on healthy cells, tumor cell chemoresistance, and poor selectivity of anticancer drugs. Moreover, the blood–brain barrier (BBB) limits the delivery of many chemotherapeutic agents.

Angiogenesis, the proliferation of new blood vessels from preexisting vessels, is an index of malignancy.[7] This process consists of various steps including endothelial cell proliferation, stimulation of endothelial cells by endogenous growth factors, cell migration, and capillary tube formation. Tumor progression could be slowed down by hindering the formation of blood vessels inside the tumor. The major advantage of _targeting angiogenesis is better accessibility of a drug into the tumor vasculature. This event can decrease tumor drug resistance, and at the same time, it increases the efficacy of the drug. However, this approach showed significant limitations associated with the physicochemical properties of the compounds and related toxicity, systemic degradation, and optimal concentrations.

Nanotechnology is considered as an emerging field with potential application in cancer research and therapy. In particular, nanoparticles (NPs) allow entry of drugs within the BBB reducing doses administered, side effects, and resistance.[8] Their nanometric size, electrostatic charge, and lipophilic characteristics allow them to freely penetrate into the brain tissue [Figure 1]. NPs can be structured to carry therapeutic drugs and imaging agents, which are loaded on or within the nanocarriers, by chemical conjugation or encapsulation. This review shows recent studies concerning NPs drug-delivery systems (DDSs) which utilized novel _targeted biomarkers in the treatment of gliomas.

F1-1
Figure 1:
Nanoparticle properties

NANOTECHNOLOGY STRATEGIES TO OVERCOME THE BLOOD–BRAIN BARRIER

BBB is a physical barrier which protects the brain and the spinal cord from passage of drugs, neurotoxins, and invading organisms and regulates the movement of nutrients between the systemic circulation and the brain tissue.[8,9] Novel drugs require effective delivery technologies that are suitable to minimize side effects, leading to better patient compliance. Efficacious doses are obtained with high concentrations and administered alone; these drugs lack specificity and cause significant damage to noncancerous tissues. Moreover, most chemotherapeutic agents have poor solubility and are mixed with toxic solvents.[10] NP-based DDSs provide better penetration of therapeutic and diagnostic agents enabling achievement of efficacy with reduced doses and systemic concentration of drugs. Nanostructure-mediated drug delivery shows the possibility to enhance drug bioavailability, improve time of release of drug molecules, and enable precision drug _targeting without altering the structural integrity of the BBB.[11] Surface property modifications allow a more accurate tumor _targeting, conferring advantageous properties to the particle.[12]

NPs may be delivered to specific sites by size-dependent passive _targeting or by active _targeting. Passive _targeting consists of chemical modifications of the NPs to increase permeability or stability. The most common surface modification of different NPs is the insertion of ethylene oxide polymers, known as poly (ethylene glycol) (PEG). PEG is able to mask the NP charge and hide _targeting ligands, directly attached to NPs. PEG increases half-life of nanocarrier DDSs by decreasing their uptake by macrophages due to steric repulsion effects and inhibition of plasma-protein adsorption. PEGylation has been successfully applied in the majority of DDSs, including lipid, polymeric NPs, and inorganic NPs.

Active _targeting is usually achieved by incorporation of a receptor-specific ligand that can promote _targeting of the drug-containing NP toward specific cells. The term “active _targeting” refers to the use of peripherally conjugated _targeting moieties for enhanced delivery of NP systems. This method has been performed to obtain a high degree of selectivity to specific tissues and to enhance the uptake of NPs into cancer cells and angiogenic microcapillaries.[12]

Absorptive-mediated transcytosis is to deliver drugs by NP system, functionalized with cell-penetrating peptides or cationic proteins via electrostatic interactions. However, because it is a nonspecific process, the adsorptive process also occurs in the blood vessels and also in other non_target organs. This evidence results in a reduced concentration of the drug in the brain. Cell-penetrating peptides (CPPs) and cationic proteins are investigated to enhance drug delivery to the brain via adsorptive-mediated transcytosis. A large variety of cargo molecules/materials have been effectively delivered into cells via CPPs, including small molecules, proteins, peptides, fragments of DNA, and NPs. The transcription factor transactivator protein (TAT), involved in the replication cycle of human immunodeficiency virus, was demonstrated to penetrate cells.[13] One of the most interesting demonstrations of the effectiveness of TAT-shuttled nanocarriers across the BBB was accomplished by TAT-conjugated CdS: Mn/ZnS quantum dots.[14] Histological data showed that TAT-Q dots migrated beyond endothelial cells and reached the brain parenchyma.

Transporters for the nutritive substances into the brain are overexpressed on the BBB and can be used for brain-_targeted delivery.[15] The glutathione transporter is highly expressed on the BBB. Systemic administration of glycosyl cholesterol-derived liposomes containing coumarin-6 displayed reduced cytotoxicity and 3.3-fold higher Cmax compared to control liposomes _targeting glucose transporters on BBB.[16]

Receptor-mediated transcytosis across the BBB shows a high specificity. Molecules necessary for the normal function of the brain are delivered to the brain by specific receptors expressed on the BBB endothelial cells. Therapeutic compounds are able to cross the BBB after association/conjugation to these specific ligands. Receptor-mediated transcytosis has been demonstrated for transferrin, insulin, insulin-like growth factors (IGF-1 and IGF-2), and the low-density lipoprotein receptor-related protein (LRP).[17] Transferrin receptor (TR) is a transmembrane glycoprotein overexpressed in GBM cells. Drug _targeting to the TR can be achieved using the endogenous ligand transferrin or using antibodies directed against the TR. Doxorubicin-loaded TR-NPs showed antitumor effect in brain tumor-bearing rats, who survived 70% longer than those treated with the doxorubicin solution.[18] The endogenous ligands may bind with the receptors hindering the binding efficiency of ligand-modified NPs. To avoid this, antibodies against these receptors were developed. Ulbrich et al.[19] developed human serum albumin NPs, coupled to transferrin or TR-mAbs, (OX26) for delivery of loperamide and showed efficiency in transporting the drug to the brain using mice, in which OX26 was conjugated to HAS-NPs. OX26 mAb can avoid competition with endogenous transferrin in the circulation system because it binds to an extracellular domain of TR.[19] LRPs 1 and 2 (LRP1 and LRP2) are multifunctional receptors. Polysorbate 80, a nonionic surfactant, could adsorb ApoE in serum when it was conjugated on to NPs, and polysorbate 80-coated NPs have been also evaluated as a brain-_targeting delivery system by many groups.[20,21] Several drugs that do not cross the BBB, including tubocurarine, loperamide, dalargin, and doxorubicin, show higher concentrations in the brain when associated with polysorbate 80-coated NPs.

ANTIANGIOGENIC STRATEGY IN THE TREATMENT OF GLIOMAS

Angiogenesis is a physiological process that plays an important role during embryonic development, tumor growth, organ growth and repair, and wound healing. The microvascular proliferation of gliomas is structurally and functionally abnormal, and it correlates to vasogenic edema, increased interstitial pressure, and heterogeneous delivery of oxygen and drugs.[22] The blood vessels formed by tumor cells are leaky and enlarged with morphologically aberrant endothelial cells and an incomplete basement membrane. These abnormalities lead to an abnormal tumor microenvironment characterized by interstitial hypertension, hypoxia, and acidosis.[23] Vascular endothelial growth factor (VEGF) and angiopoietin families are predominant angiogenic factors in glioblastoma.[24] The control of the various phases of the angiogenesis can represent a valid approach in tumor therapy. It can prevent the development of tumors and can act as a complementary therapy in cancer treatment. The antiangiogenic approach can be directed to a specific biomarker so as to inhibit or alter a specific pathway, where it can extrinsecate through a link with specific membrane receptors overexpressed by glial cells, thus allowing the _targeted release of the pharmacological compound.

VEGF is expressed in gliomas and directly correlates to tumor grading, vascularity, and clinical behavior while inversely correlates to prognosis.[25] Bevacizumab (Avastin®; Hoffman-La Roche Ltd., Basel, Switzerland) is a humanized monoclonal antibody _targeting VEGF and approved in the US for GBM in 2009. Bevacizumab interferes with the VEGF pathway, thus counteracting the angiogenic processes, reducing vascular permeability and cerebral tributary blood flow of the tumor. However, clinical studies conducted on human subjects have yielded little encouraging results. The phase III “Avaglio” trial, conducted on 921 patients with newly diagnosed GBM, resulted in no overall survival benefit in bevacizumab-treated versus placebo-treated patients (median overall survival of 16.8 months for bevacizumab-treated patients and 16.7 months for placebo-treated patients).[26] A second phase III trial, the RTOG 0825 trial, produced similar results.[27] Recently, a new technology called coacervation has been developed for solid-lipid NPs preparation.[28] Through this technique, different hydrophilic drugs have been loaded into solid-lipid NPs using the hydrophobic ion-pairing technique. A recent experimental study showed an increase in bevacizumab activity when combined with solid-lipid NPs structured by the coacervation technology. The activity of bevacizumab, evaluated using four different in vitro tests on human umbilical vein endothelial cells (HUVEC) cells, increased by 100-fold to 200-fold when delivered in solid-lipid NPs.[29] In a recent study on rat glioma model, the authors describe the possibility to downregulate the expression of VEGF by a compound formed by lipid NPs carrying VEGF antisense oligonucleotides, both in vitro and in vivo.[30] In another study, the authors evaluated a liposomal DDS combined with cisplatin analog, cis-diamminedinitratoplatinum (II), and antibodies against the native form of VEGF or VEGFR2. The results evidenced sustained drug release profile, high affinity to antigens, and increased uptake by glioma C6 and U87MG cells.[31] Cerium oxide NP treatment caused decreased expression levels of VEGF in the human astrocytoma cell line (MOG-G-CCM) associated with reduced motility and capacity of endothelial cells to form new capillaries.[32] Chwalibog et al.[33] reported the action of diamond-NPs on VEGF-A and VEGF-receptor in human U87 glioblastoma cells. After diamond-NPs treatment, the authors demonstrated a reduced fibroblast growth factor 2 and VEGF expression, and a decreased development of blood vessels in GBM. An interesting study evaluated the action of magnetic NPs as (MRI) agents for in vivo visualization of gliomas.[34] Monoclonal antibodies against VEGF were conjugated to ferric oxide-bovine serum albumin (Fe3O4-BSA) through a PEG linker. The results showed that these _targeted NPs are effective in visualization of gliomas under MRI.

Overexpression of epidermal growth factor receptor (EGFR) on blood–brain–tumor barrier (BBTB) allows a selective _targeting of EGFR. EFGR is highly expressed in cancer cells, in more than 40% of GBM cases, and the mutated form EGFRvIII is expressed in more than 40% of GBM cases. A compound structured by PEGylated immunoliposomes (ILs) conjugated with anti-human EGFR antibodies cetuximab (α-hEGFR-ILs) was developed.[35] The affinity of the α-hEGFR-ILs for the EGFR was evaluated in vitro using U87MG and U251MG cells and in vivo using an intracranial U87MG xenograft model. The in vitro studies revealed significantly higher binding of α-hEGFR-ILs, while the in vivo study evidenced a more efficacious collect of α-hEGFR-ILs inside the tumor. A clinical study on animals showed the effectiveness of anti-EGF nanomedicine in GBM.[36] The therapeutic agent used was cetuximab (that binds both EGFR and EGFRvIII deletion mutant) conjugated with iron oxide NPs (IONPs). The result was an increase in survival. The use of an interesting pharmacological compound formed by transferrin-conjugated lipopolyplex nanoparticles (Tf-NPs) causes inhibition of migration and expression of EGFR;[37] these NPs can act both on cancer stem cells (CSCs) and on differentiated cancer cells. In an interesting study, the authors describe the phase I results of EnGeneIC delivery vehicle minicells coupled with doxorubicin, in patients with recurrent GBM.[38] They utilized the anti-EGFR monoclonal antibody Vectibix to _target the EGFR protein on cancer cells, thus releasing doxorubicin. The compound was well tolerated with no dose-limiting toxicity.

Integrins play prominent roles in tumor invasion and angiogenesis. In GBM, αvβ3 and αvβ5 are overexpressed on brain tumor cells and on brain tumor neovessels, favoring interaction between glial cells and the extracellular matrix. These integrins could be specifically _targeted with arginine-glycine-aspartic acid (RGD) peptide. Cyclic RGD and peptide-22 dual-modified liposomes loaded with doxorubicin showed to overcome BBB/BBTB, inhibiting the growth of GBM.[39] The compound paclitaxel (PTX)-loaded c (RGDyK)-PEG -block-poly (lactic acid) micelle (c [RGDyK]-PEG-PLA-PTX) was tested in U87MG cells.[40] Cilengitide (cyclo [RGDfV]) is a cyclic peptide that selectively binds αv integrins. c (RGDyK)-PEG-PLA micelle when loaded with PTX demonstrated the strongest tumor growth inhibition among the studied PTX formulations. The compound increased the overall median survival time of treated mice (48 days). In recent research, a pharmacological compound characterized by a platinum anticancer drug-incorporating polymeric micelle with cyclic RGD showed a high permeability and storage capacity into the tumor tissue.[41]

Glioma cells show an upregulation of expression of interleukin-13 (IL-13) receptor α2 (IL-13rα2) on their surface cells. A novel compound structured with Pep-1 and CREKA peptides GBM _targeting nano-DDS (PC-NP) was designed.[42] Pep-1 was used to overcome the BBTB and interfere with glioma cells via IL-13rα2-mediated endocytosis, and CREKA was utilized to bind to fibrin-fibronectin complexes expressed in tumor microenvironment. The results showed that the cellular uptake of PC-NPs by U87MG cells was significantly enhanced as compared to non_targeting NPs. In accordance with the increased cellular uptake, PC-NPs remarkably increased the cytotoxicity of its payload PTX against U87MG cells. In a recent study, Madhankumar et al.[43] showed the improvement of internalization of doxorubicin-loaded nanoliposomes, _targeted with conjugated IL-13, and cytotoxicity in U251 glioma cells. Bernardi et al.[44] conjugated gold-silica nanoshells to an antibody specific to IL-13rα2 demonstrating that these immunonanoshells are capable of inducing cell death in U373MG and U87MG cell lines. A significant therapeutic effect was found after convection-enhanced delivery (CED) of both IONPs and EGFRvIIIAb-IONPs in mice.[45] Use of bioconjugated magnetic NPs may permit the advancement of CED in the treatment of malignant gliomas due to their sensitive imaging qualities on standard T2 weighted MRI and therapeutic effects.

Curcumin is a polyphenolic compound derived from the Indian spice turmeric. It presents proapoptotic, antiangiogenic, anti-inflammatory, immunomodulatory, and antimitogenic effects.[46] NanoCurc™, a polymeric-NP formulation of curcumin, was used to treat medulloblastoma and glioblastoma cells. This compound caused a dose-dependent decrease in growth of multiple brain tumor cell cultures, including the embryonal tumor-derived lines DAOY and D283Med, the glioblastoma neurosphere lines HSR-GBM1 and the JHH-GBM14 cell lines.[47] The effect was associated with a combination of G2/M arrest and apoptotic induction. Furthermore, curcumin has been utilized in a spherical core-shell nanostructure, formed by amphiphilic methoxypolyethylene glycol-poly (caprolactone) (mPEG-PCL) block copolymers, delivered into C6 glioma cells through endocytosis. In vitro studies proved that the cytotoxicity of these nanoconjugates would be the result of a pro-apoptotic effect against rat C6 glioma cell line, in a dose-dependent manner.[48]

The combination of silver NPs (AgNPs) with magnetic NP hyperthermia (MNPH) treatment has been used as treatment in glioma model. AgNPs had significant effect on enhancing thermoinduced killing in vitro. In the glioma-bearing rat model, AgNPs combined with MNPH enhance Bax (Bcl-2-associated X protein) expression in cancer cells, which was correlated with cell apoptosis induction. The mechanism of thermosensitization by AgNPs might be related to the release of Ag+ cation from the silver nanostructures inside cells.[49]

p53 is currently established in multiple clinical trials. Some authors have developed a nanocomplex structured by a cationic liposome with the surface modified with an anti-TR encapsulated with wtp53 plasmid DNA.[50] The compound, named SGT-53, is designed to _target tumor cells via receptor-mediated endocytosis. As a result, temozolomide-resistant GBM would become more sensitive, resulting in a significant apoptosis of CD133-positive CSCs, and an increase in median survival time.[50]

CONCLUSIONS

Recent studies have shown that nanotechnology could be used to both improve the treatment efficacy and to reduce the adverse side effects. Nanotechnology-based approaches to _targeted delivery of therapeutic compounds may potentially be engineered to carry out specific functions as needed. New NPs DDSs are providing innovative, controlled, and _targeted techniques, resulting in a considerable decrease in therapy-associated side effects while increasing antitumor efficacy. It is also possible to take novel selective contrast enhancement molecules into the brain and in particular into neoplastic brain tissue, to visualize brain tumor and to study in vivo almost all of its aspects, such as cellular proliferation, angiogenesis, necrosis, tumor-safe tissue interface, and edema. This approach could permit the use of a lower total dose of drug, a selective drug delivery to _target brain tumor cells, both into the central core of tumor and into the distal foci of tumor cells within areas often characterized from integrity of BBB. This last aspect is very interesting in the future challenges of multimodal brain tumor diagnosis and treatment, such as precocious diagnosis, preoperative histological and grade diagnosis, preoperative treatment planning, follow-up, and recurrence diagnosis and treatment. Notwithstanding, there are potential risks related to these novel approaches. Some cancer cells could develop drug resistance making the drugs released from the _targeted NPs ineffective. Moreover, NPs might change stability, solubility, and pharmacokinetic properties of the carried drugs. In addition, some materials used to create NPs possess low toxicity but quickly degrade and do not circulate in tissues long enough to warrant a sustained drug/gene delivery. Objects of debate are the long-term effects of interactions between NPs and coating of molecules and _target cells.

Cerebral gliomas represent, to date, a still unresolved challenge for the medical community. There are several factors underlying the disappointing results in brain cancer therapeutics, including limited cell drug uptake, intracellular drug metabolism, inherent tumor sensitivity to chemotherapy, and cellular mechanisms of resistance. Prognosis remains poor and the mortality is still high. New biomolecular studies have allowed identifying novel biomarkers and pathways of various phases of tumor progression. Therefore, new treatments have been proposed with the aim of interacting, in a specific way, with such _targets. The molecules that participate in the angiogenesis, a mechanism that allow and promote neoplastic progression, appear to be of interest. The main limitation of this approach is linked to the identification of the most suitable _target. Furthermore, due to the numerous pathways activated during tumor progression, hypothetical _target may be subject to cyclic re-activation, making the treatment practically ineffective. Identification of multiple _targets could be a more viable option and so is to _target the activated molecules in the early stages of tumor progression.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.

REFERENCES

1. Ostrom QT, Bauchet L, Davis FG, Deltour I, Fisher JL, Langer CE, et al The epidemiology of glioma in adults: A “state of the science” review Neuro Oncol. 2014;16:896–913
2. Banan R, Hartmann C. The new WHO 2016 classification of brain tumors-what neurosurgeons need to know Acta Neurochir (Wien). 2017;159:403–18
3. Rock K, McArdle O, Forde P, Dunne M, Fitzpatrick D, O'Neill B, et al Aclinical review of treatment outcomes in glioblastoma multiforme – The validation in a non-trial population of the results of a randomised phase III clinical trial: Has a more radical approach improved survival? Br J Radiol. 2012;85:e729–33
4. Hanif F, Muzaffar K, Perveen K, Malhi SM, Simjee SH. Glioblastoma multiforme: A review of its epidemiology and pathogenesis through clinical presentation and treatment Asian Pac J Cancer Prev. 2017;18:3–9
5. Caruso G, Merlo L, Tot E, Pignataro C, Caffo MGrumezescu AM. Nanotechnology and the new frontiers of drug delivery in cerebral gliomas Nano- and Microscale Drug Delivery Systems: Design and Fabrication. 2017 Amsterdam Elsevier:95–112
6. Schucht P, Beck J, Abu-Isa J, Andereggen L, Murek M, Seidel K, et al Gross total resection rates in contemporary glioblastoma surgery: Results of an institutional protocol combining 5-aminolevulinic acid intraoperative fluorescence imaging and brain mapping Neurosurgery. 2012;71:927–35
7. Xu Y, Yuan FE, Chen QX, Liu BH. Molecular mechanisms involved in angiogenesis and potential _target of antiangiogenesis in human glioblastomas Glioma. 2018;1:35–42
8. Caruso G, Caffo M, Alafaci C, Raudino G, Cafarella D, Lucerna S, et al Could nanoparticle systems have a role in the treatment of cerebral gliomas? Nanomedicine. 2011;7:744–52
9. Agarwal S, Sane R, Oberoi R, Ohlfest JR, Elmquist WF. Delivery of molecularly _targeted therapy to malignant glioma, a disease of the whole brain Expert Rev Mol Med. 2011;13:e17
10. Kwon GS. Polymeric micelles for delivery of poorly water-soluble compounds Crit Rev Ther Drug Carrier Syst. 2003;20:357–403
11. Dubin CH. Special delivery: Pharmaceutical companies aim to _target their drugs with nano precision Mech Eng Nanotechnol. 2004;126:10–2
12. Praetorius NP, Mandal TK. Engineered nanoparticles in cancer therapy Recent Pat Drug Deliv Formul. 2007;1:37–51
13. Rapoport N, Marin AP, Timoshin AA. Effect of a polymeric surfactant on electron transport in HL-60 cells Arch Biochem Biophys. 2000;384:100–8
14. Santra S, Yang H, Holloway PH, Stanley JT, Mericle RA. Synthesis of water-dispersible fluorescent, radio-opaque, and paramagnetic CdS: Mn/ZnS quantum dots: A multifunctional probe for bioimaging J Am Chem Soc. 2005;127:1656–7
15. Wei X, Chen X, Ying M, Lu W. Brain tumor-_targeted drug delivery strategies Acta Pharm Sin B. 2014;4:193–201
16. Qin Y, Fan W, Chen H, Yao N, Tang W, Tang J, et al In vitro and in vivo investigation of glucose-mediated brain-_targeting liposomes J Drug _target. 2010;18:536–49
17. Zhang TT, Li W, Meng G, Wang P, Liao W. Strategies for transporting nanoparticles across the blood-brain barrier Biomater Sci. 2016;4:219–29
18. Kumar P, Wu H, McBride JL, Jung KE, Kim MH, Davidson BL, et al Transvascular delivery of small interfering RNA to the central nervous system Nature. 2007;448:39–43
19. Ulbrich K, Hekmatara T, Herbert E, Kreuter J. Transferrin- and transferrin-receptor-antibody-modified nanoparticles enable drug delivery across the blood-brain barrier (BBB) Eur J Pharm Biopharm. 2009;71:251–6
20. Martins SM, Sarmento B, Nunes C, Lúcio M, Reis S, Ferreira DC. Brain _targeting effect of camptothecin-loaded solid lipid nanoparticles in rat after intravenous administration Eur J Pharm Biopharm. 2013;85:488–502
21. Wilson B, Lavanya Y, Priyadarshini SR, Ramasamy M, Jenita JL. Albumin nanoparticles for the delivery of gabapentin: Preparation, characterization and pharmacodynamic studies Int J Pharm. 2014;473:73–9
22. Jain RK, di Tomaso E, Duda DG, Loeffler JS, Sorensen AG, Batchelor TT. Angiogenesis in brain tumours Nat Rev Neurosci. 2007;8:610–22
23. Chen F, Zhuang X, Lin L, Yu P, Wang Y, Shi Y, et al New horizons in tumor microenvironment biology: Challenges and opportunities BMC Med. 2015;13:45
24. Caffo M, Barresi V, Caruso G, La Fata G, Pino MA, Raudino G, et alLichtor T Gliomas biology: Angiogenesis and invasion Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications. 2013 Rijeka IntechOpen:37–103
25. Rong Y, Durden DL, Van Meir EG, Brat DJ. 'Pseudopalisading' necrosis in glioblastoma: A familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis J Neuropathol Exp Neurol. 2006;65:529–39
26. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al Arandomized trial of bevacizumab for newly diagnosed glioblastoma N Engl J Med. 2014;370:699–708
27. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma N Engl J Med. 2014;370:709–22
28. Battaglia L, Gallarate M, Cavalli R, Trotta M. Solid lipid nanoparticles produced through a coacervation method J Microencapsul. 2010;27:78–85
29. Battaglia L, Gallarate M, Peira E, Chirio D, Solazzi I, Giordano SM, et al Bevacizumab loaded solid lipid nanoparticles prepared by the coacervation technique: Preliminary in vitro studies Nanotechnology. 2015;26:255102
30. Brioschi AM, Calderoni S, Pradotto LG, Guido M, Strada A, Zenga F, et al Solid lipid nanoparticles carrying oligonucleotides inhibit vascular endothelial growth factor expression in rat glioma models J Nanoneurosci. 2009;1:65–74
31. Shein SA, Kuznetsov II, Abakumova TO, Chelushkin PS, Melnikov PA, Korchagina AA, et al VEGF- and VEGFR2-_targeted liposomes for cisplatin delivery to glioma cells Mol Pharm. 2016;13:3712–23
32. Sack-Zschauer M, Bader S, Brenneisen P. Cerium oxide nanoparticles as novel tool in glioma treatment: An in vitro study J Nanomed Nanotechnol. 2017;8:474
33. Chwalibog A, Grodzik M, Hotowy A, Jaworski S, Prasek M, Sawosz E, et al VEGF-dependent mechanism of anti-angiogenic action of diamond nanoparticles in glioblastoma multiforme tumor TechConnect Briefs. 2012;3:218–21
34. Abakumov MA, Nukolova NV, Sokolsky-Papkov M, Shein SA, Sandalova TO, Vishwasrao HM, et al VEGF-_targeted magnetic nanoparticles for MRI visualization of brain tumor Nanomedicine. 2015;11:825–33
35. Mortensen JH, Jeppesen M, Pilgaard L, Agger R, Duroux M, Zachar V, et al _targeted antiepidermal growth factor receptor (cetuximab) immunoliposomes enhance cellular uptake in vitro and exhibit increased accumulation in an intracranial model of glioblastoma multiforme J Drug Deliv. 2013;2013:209205
36. Freeman AC, Platt SR, Holmes S, Kent M, Robinson K, Howerth E, et al Convection-enhanced delivery of cetuximab conjugated iron-oxide nanoparticles for treatment of spontaneous canine intracranial gliomas J Neurooncol. 2018;137:653–63
37. Wang X, Huang X, Yang Z, Gallego-Perez D, Ma J, Zhao X, et al _targeted delivery of tumor suppressor MicroRNA-1 by transferrin-conjugated lipopolyplex nanoparticles to patient-derived glioblastoma stem cells Curr Pharm Biotechnol. 2014;15:839–46
38. Whittle JR, Lickliter JD, Gan HK, Scott AM, Simes J, Solomon BJ, et al First in human nanotechnology doxorubicin delivery system to _target epidermal growth factor receptors in recurrent glioblastoma J Clin Neurosci. 2015;22:1889–94
39. Chen C, Duan Z, Yuan Y, Li R, Pang L, Liang J, et al Peptide-22 and cyclic RGD functionalized liposomes for glioma _targeting drug delivery overcoming BBB and BBTB ACS Appl Mater Interfaces. 2017;9:5864–73
40. Zhan C, Gu B, Xie C, Li J, Liu Y, Lu W, et al Cyclic RGD conjugated poly (ethylene glycol)-co-poly (lactic acid) micelle enhances paclitaxel anti-glioblastoma effect J Control Release. 2010;143:136–42
41. Miura Y, Takenaka T, Toh K, Wu S, Nishihara H, Kano MR, et al Cyclic RGD-linked polymeric micelles for _targeted delivery of platinum anticancer drugs to glioblastoma through the blood-brain tumor barrier ACS Nano. 2013;7:8583–92
42. Wang X, Zhang Q, Lv L, Fu J, Jiang Y, Xin H, et al Glioma and microenvironment dual _targeted nanocarrier for improved antiglioblastoma efficacy Drug Deliv. 2017;24:1401–9
43. Madhankumar AB, Slagle-Webb B, Mintz A, Sheehan JM, Connor JR. Interleukin-13 receptor-_targeted nanovesicles are a potential therapy for glioblastoma multiforme Mol Cancer Ther. 2006;5:3162–9
44. Bernardi RJ, Lowery AR, Thompson PA, Blaney SM, West JL. Immunonanoshells for _targeted photothermal ablation in medulloblastoma and glioma: An in vitro evaluation using human cell lines J Neurooncol. 2008;86:165–72
45. Hadjipanayis CG, Machaidze R, Kaluzova M, Wang L, Schuette AJ, Chen H, et al EGFRvIII antibody-conjugated iron oxide nanoparticles for magnetic resonance imaging-guided convection-enhanced delivery and _targeted therapy of glioblastoma Cancer Res. 2010;70:6303–12
46. Hatcher H, Planalp R, Cho J, Torti FM, Torti SV. Curcumin: From ancient medicine to current clinical trials Cell Mol Life Sci. 2008;65:1631–52
47. Lim KJ, Bisht S, Bar EE, Maitra A, Eberhart CG. A polymeric nanoparticle formulation of curcumin inhibits growth, clonogenicity and stem-like fraction in malignant brain tumors Cancer Biol Ther. 2011;11:464–73
48. Shao J, Zheng D, Jiang Z, Xu H, Hu Y, Li X, et al Curcumin delivery by methoxy polyethylene glycol-poly (caprolactone) nanoparticles inhibits the growth of C6 glioma cells Acta Biochim Biophys Sin (Shanghai). 2011;43:267–74
49. Liu L, Ni F, Zhang J, Jiang X, Lu X, Guo Z, et al Silver nanocrystals sensitize magnetic-nanoparticle-mediated thermo-induced killing of cancer cells Acta Biochim Biophys Sin (Shanghai). 2011;43:316–23
50. Kim SS, Rait A, Kim E, Pirollo KF, Nishida M, Farkas N, et al Ananoparticle carrying the p53 gene _targets tumors including cancer stem cells, sensitizes glioblastoma to chemotherapy and improves survival ACS Nano. 2014;8:5494–514
Keywords:

Angiogenesis; blood–brain barrier; glioblastoma multiforme; glioma; nanoparticles; nanotechnologies

© 2018 Glioma | Published by Wolters Kluwer – Medknow