Next Article in Journal
Characterisation of Endogenous Peptides Present in Virgin Olive Oil
Next Article in Special Issue
Pancreatic Ductal Cell-Derived Extracellular Vesicles Are Effective Drug Carriers to Enhance Paclitaxel’s Efficacy in Pancreatic Cancer Cells through Clathrin-Mediated Endocytosis
Previous Article in Journal
Biotechnological Applications and Health-Promoting Properties of Flavonols: An Updated View
Previous Article in Special Issue
Indomethacin and Diclofenac Hybrids with Oleanolic Acid Oximes Modulate Key Signaling Pathways in Pancreatic Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Delivering Therapeutics to Glioblastoma: Overcoming Biological Constraints

Department of Neurological Surgery, University of Massachusetts Medical School, 55Lake Avenue North, Worcester, MA 01655, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(3), 1711; https://doi.org/10.3390/ijms23031711
Submission received: 30 December 2021 / Revised: 30 January 2022 / Accepted: 31 January 2022 / Published: 2 February 2022

Abstract

:
Glioblastoma multiforme is the most lethal intrinsic brain tumor. Even with the existing treatment regimen of surgery, radiation, and chemotherapy, the median survival time is only 15–23 months. The invasive nature of this tumor makes its complete removal very difficult, leading to a high recurrence rate of over 90%. Drug delivery to glioblastoma is challenging because of the molecular and cellular heterogeneity of the tumor, its infiltrative nature, and the blood–brain barrier. Understanding the critical characteristics that restrict drug delivery to the tumor is necessary to develop platforms for the enhanced delivery of effective treatments. In this review, we address the impact of tumor invasion, the molecular and cellular heterogeneity of the tumor, and the blood–brain barrier on the delivery and distribution of drugs using potential therapeutic delivery options such as convection-enhanced delivery, controlled release systems, nanomaterial systems, peptide-based systems, and focused ultrasound.

1. Introduction

Glioblastoma multiforme (GBM) is the most common primary brain malignancy in adults, accounting for more than 50% of intrinsic brain tumors [1]. According to the World Health Organization (WHO) classification, GBM is a grade IV glioma, highly invasive with a five-year survival rate of less than 5% [2,3]. This highly aggressive disease presents a very poor prognosis. The median survival time from diagnosis of the tumor is approximately 15–23 months. The incidence of GBM is 3.19 per 100,000 persons in the United States with a median age of 64 years [4,5,6,7,8].
Widely recognized risk factors associated with GBM occurrence are exposure to high-dose ionizing radiation and certain genetic syndromes, including neurofibromatosis type 1 and Li-Fraumeni syndrome [2,9,10]. The occurrence of GBM is higher in males and among individuals 50 years of age and older [5].
GBM is characterized by rapid cell proliferation and extensive invasion of tumor cells into the surrounding brain, making complete removal of the tumor impossible [11]. These features lead to a high recurrence rate, even with the current treatment regimen of maximum safe surgical removal, radiation therapy and temozolomide chemotherapy. Intratumoral molecular heterogeneity, presence of the blood–brain barrier, and tumor immune evasion via local immunosuppression limits the success of existing therapies [12]. Numerous drug therapies directed against GBM have shown promising results in in vitro assays, but all have had limited success in vivo. This is due in part to the diffuse, heterogeneous nature of the tumor, and the blood–brain barrier that limits the ability of many drugs to enter the brain parenchyma. Understanding the critical factors that restrict drug delivery to the brain is necessary to develop platforms for the enhanced delivery of effective drugs.
In this review, we discuss the impact that molecular and cellular tumor heterogeneity, tumor dispersion and the blood–brain barrier have on the delivery and distribution of pharmacological agents to GBM. The currently available chemotherapeutic delivery systems designed to overcome these constraints are also discussed (Figure 1). We will focus on the use of convection-enhanced delivery, controlled release systems, nanomaterial systems, peptide-based therapeutics and focused ultrasound (FUS) for the treatment of this aggressive tumor.

1.1. Molecular Heterogeneity

GBM is characterized by molecular heterogeneity within a single tumor in addition to inter-patient heterogeneity. Molecular heterogeneity underlies the cellular heterogeneity in GBM, i.e., the differences between cell types within a tumor [13]. This molecular heterogeneity may be responsible for differences in individual patient responses to therapy and prognosis as well as the failure of targeted therapies [14,15]. Common gene mutations include IDH1/2 mutations, O6-methylguanine DNA methyltransferase (MGMT) promoter methylation, co-deletion of 1p and 19q, and EGFR amplification/truncation. Primary GBM frequently exhibits epidermal growth factor receptor (EGFR) over expression, PTEN (MMC-I) mutation or deletion, CDKN2A (p16) deletion, and MDM2 amplification. Secondary GBM predominantly has IDH1, ATRX and TP53 mutations [16]. Based on differences in genetic alterations and the expression of EGFR, NF1, PDGFRA/IDH1, PI3K and other key genes, primary GBM can be classified into four subtypes: proneural, neural, classical, and mesenchymal, with each subtype varying in its gene expression signature [17]. Secondary GBMs predominantly have IDH1 (or less commonly IDH2) mutations that have a proneural gene expression signature and a better prognosis than GBMs with wild-type IDH1/IDH2.
In addition to the molecular heterogeneity described above, GBM cells are morphologically and functionally heterogeneous. Single-cell RNA-Seq studies of individual tumor cells have revealed that GBM tumor cells exist in multiple stages of differentiation [18]. Other studies have shown that astrocyte-like GBM cells can transdifferentiate to become endothelial-like cells [19,20]. As a result of this extraordinary cellular heterogeneity, individual GBM cells may be more or less replicative, invasive, or sensitive to radiation or chemotherapy [21].

1.2. Blood–Brain Barrier

The blood–brain barrier is composed of a highly specialized circuit of blood vessels that are lined by brain microvascular endothelial cells (BMEC), the cell–cell junctions between which restrict the entry of potentially harmful substances (including systemically administered therapeutics) into the brain. The BMECs are surrounded by pericytes, astrocytes and the basal membrane [22,23]. This is indeed a protective layer safeguarding the brain from damaging agents in the systemic circulation and keeping CNS homeostasis to allow proper neuronal function [24]. The two major features of the blood–brain barrier are: (1) the presence of tight junctions limiting paracellular transport, and (2) reduced fenestrations and transport vesicles limiting transcellular transport. Small molecules need to be less than 400 Da in size and lipid soluble in order to cross the barrier [25].
Disruption of the blood–brain barrier (BBB) is observed in glioma tumor regions. Unfortunately, this degree of BBB disruption is not sufficient to allow for the ability of therapeutics to reach this diffusely infiltrating tumor [26,27]. Biochemical and physical methods can be used to further increase the permeability of this biological barrier. Ion channel activators such as ATP-sensitive and calcium-activated potassium channel activators, phosphodiesterase 5 (PDE5) inhibitors, bradykinin type 2 receptor activators, adenosine 2A receptor (A2AR) agonists, papaverine, and certain microRNAs represent biochemical approaches to biochemical modulation of the BBB. PDE5 inhibitors decrease cGMP degradation and increase vesicular transport, thereby increasing BBB permeability within the tumor region. Potassium channel activators downregulate tight junction protein expression and increase the formation of pinocytotic vesicles. Bradykinin activators increase transcytosis and modulate tight junction protein expression and cGMP synthesis. A2AR agonists and papaverine downregulate tight junction protein expression. MicroRNAs such as miR-132-3p increase blood–brain barrier permeability by increasing transcytosis. Mannitol, an osmotic agent, is widely used to disrupt the blood–brain barrier by vasodilatation and shrinkage of endothelial cells [28,29]. Physical strategies for modulating BBB permeability include the application of electromagnetic pulses, laser-induced thermal therapy, radiotherapy, and focused ultrasound (FUS). All these methods downregulate the expression of tight junction proteins. FUS is also involved in increasing transcytosis [22,30].

2. Methods of Drug Delivery to Glioblastoma

In this review, we will discuss the application of controlled release systems, convection-enhanced delivery, nanomaterial systems, peptide-based therapeutics, and focused ultrasound for drug delivery to glioblastoma, overcoming the obstacles posed by GBM molecular and cellular heterogeneity, GBM cell invasion/dispersal and the blood–brain barrier.

2.1. Controlled-Release Systems

Implantable drug release systems enable the direct delivery of therapeutic agents to the tumor site, circumventing the need to cross the blood–brain barrier. Drug-loaded biocompatible materials such as drug-impregnated gels can be designed so that they release low doses of drugs at the tumor site over a prolonged period of time.
Implantable controlled-release delivery systems can be constructed with degradable or non-degradable polymers. Between these two options, biodegradable polymers are more commonly used [31]. One such biodegradable polymer delivery system that has been used clinically for GBM is the Gliadel® wafer. This is a biodegradable polymer wafer loaded with the chemotherapeutic drug BCNU. It is the only FDA-approved drug delivery implant for treating GBM [11]. Gliadel was approved by the FDA in 1996 for recurrent GBM and later in 2003 for upfront treatment of malignant glioma [31,32,33]. In another study, biodegradable wafers were created for the combined delivery of temozolomide and carmustine in a rat glioma model. This approach increased the median survival of the animals significantly, with 25% of the animals living long term > 120 days [34]. Biodegradable polymer implants releasing rapamycin were found to increase survival both in the presence and absence of radiation therapy in a rat malignant glioma model [35]. The rigid structure of these systems has limited drug loading capacity and they can be dislodged from the original site of implantation. In addition, the wafer systems also led to the occurrence of seizures, intracranial hypertension, meningitis, cerebral edema, and impaired wound healing in neurosurgical patients [11].
A pH-responsive carboxymethylcellulose biopolymer system was used to deliver the chemotherapeutic drug, rhodamine B encapsulated in multiple emulsions to glioblastoma cells in vitro [36]. In a mouse GBM tumor resection model, hydrogel-based co-delivery of the chemotherapeutic agents paclitaxel and temozolomide enhanced survival [37]. Hydrogels can adapt their shape while nevertheless retaining sufficient drug capacity [11]. The use of multiple emulsions and hydrogels avoided the limitations seen with the rigid structure of wafer implants. However, the intrinsic hydrophilic nature of hydrogels does not allow for the effective delivery of hydrophobic therapeutic agents [38].
With sustainable release of therapeutic agents over time, implantable controlled-release systems limit the local GBM recurrence after resection by interacting with the tumor cells at the resection site. However, these systems are not only limited by a variety of side effects as listed above, but also by poor drug distribution to distant tumor cells that have migrated in the normal brain parenchyma. Consequently, these implantable local delivery systems have limited ability to affect infiltrating GBM cells distant to the site of implantation.

2.2. Convection-Enhanced Delivery

Convection-enhanced delivery (CED) is a catheter-based drug delivery method that depends on pressure gradients rather than diffusion to deliver therapeutic agents into brain tumors. This technique involves the stereotactic insertion of one or more catheters into the tumor. The catheters are connected to a syringe/drug delivery pump that maintains a positive pressure gradient that promotes the distribution of higher volumes of drug over a larger area. Ensuring direct intratumoral drug delivery independent of drug concentration or diffusivity of the infusate, CED allows the use of large volumes of the drug at lower and less-toxic doses [39]. The ratio between the volume of distribution and the volume of infusion is a key factor determining the optimum flow rate and success of this technique.
CED of temozolomide, the standard cytostatic drug for the treatment of GBM, combined with subcutaneous immunizations with irradiated cancer cells had a synergistic effect on tumor growth and overall survival in a mouse-GL261 glioma model. However, the same protocol did not display synergy in the KR158 mouse glioma model, which is resistant to radiation and chemotherapy. In the latter model, CED of temozolomide increased survival [40]. By initiating antitumor immune response, irradiated cells may help to overcome intratumoral immunosuppression and enhance the antitumor activity of temozolomide.
In a phase I clinical trial, CED was used to infuse carboplatin to patients with WHO grade III astrocytomas or grade IV gliomas (GBM). This approach increased median overall survival without any systemic toxicity [41]. This was the first clinical trial to demonstrate that CED of carboplatin into the brain is safe. Oral or intravenous administration of platinated drugs does not produce effective concentrations in the brain and has been associated with systemic toxicity [42].
MRI-guided CED of iron oxide nanoparticles (IONP) conjugated to epidermal growth factor receptor deletion mutant III antibody (EGFRVIIIAb) showed significant increase in survival in a mouse GBM model [43]. Binding of EGFRvIIIAb-IONP conjugate to target cells was evaluated by changes in the MRI signal. IONP is a theranostic nanoparticle which has imaging properties as well as anticancer activity. EGFRvIIIAb is a tumor-specific cell surface protein. The experimental animal groups treated with EGFRvIIIAb-IONP conjugate as well as EGFRvIIIAb alone had a single survivor after 120 days. This study demonstrated the feasibility of conjugating biological agents or ligands that bind to tumor-specific proteins along with concomitant imaging to ensure targeted distribution of the therapeutic agent.
Direct delivery of anticancer agents in large volumes and at less toxic doses makes CED an attractive drug delivery method for GBM. Implantable catheters can be used to repeat CED at required intervals, but the efficacy of this approach may be limited by the highly invasive nature of the tumor with GBM cells dispersed to distant sites [44]. Challenges in maintaining the ratio between the volumes of distribution and infusion, flow rate, formation of air bubbles, infusate reflux along the catheter, and the variability in the tumor tissue composition can limit the establishment of the required pressure gradient, thereby limiting the efficacy of CED [45].

2.3. Nanomaterial Systems

Nanomedicine is an emerging candidate for cancer therapy that uses a variety of different types of nanocarriers such as lipid-based, polymer-based, inorganic, viral, and drug-conjugated nanoparticles [46]. Small size, high surface-to-volume ratio and other physico-chemical parameters make nanoparticles unique [47]. Accentuated drug delivery along with potential applications in diagnosis and imaging makes nanomedicine an appealing approach to cancer therapeutics [48].
Nanomaterial systems for drug delivery improve GBM tumor targeting mainly by promoting drug diffusion through the blood–brain barrier and by the enhanced permeability and retention (EPR) effect. The EPR effect is dependent on the highly angiogenic nature of GBM where leaky vasculature is commonly present [49]. Because of this phenomenon, nanoparticles can passively modulate the biodistribution of loaded molecules and increase their accumulation in cancer tissues with leaky vasculature [50,51,52]. The efficacy of antitumor drug delivery via nanoparticles is also enhanced by several additional mechanisms, including increased cellular internalization, activation of immune cells, and reactive oxygen species (ROS) production [53]. Surface functionalization of a nanoparticle with an active principle, a targeting agent, and a compound for detection will increase the functional range of the nanomaterial drug delivery targeting biomolecules, thereby enabling both drug delivery and disease diagnosis [54]. The long half-life and improved cellular uptake and biodistribution increase the appeal of nanomaterial systems for tumor therapy [49].
The delivery of tumor antigens and adjuvants using nanoparticles as a vehicle has been shown to increase the efficacy of immune therapy against GBM [55,56,57]. Different types of nanomaterials, including polymeric nanoparticles or lipid-based nanoparticles, may also be used to deliver nucleic acids intended for tumor therapy [49]. A poly(ε-caprolactone) (PCL)-based nanoparticle system was used to deliver the natural growth modulating tripeptide GHK (glycyl-L-histidyl-L-lysine) to human GBM cells in vitro, thereby reducing GBM cell viability to nearly 65%. However, this approach did not show anticancer activity at concentrations less than 20 mg/mL [58]. GHK is a natural tripeptide with a varied array of biological activities including anticancer, antioxidant, anti-inflammatory, skin remodeling, wound healing, etc. [59]. The anti-GBM activity of GHK demonstrated in this study in vitro requires further validation in vivo.
A nanobubble-based theranostic system consisting of intravenously administered iron-platinum nanoparticles loaded with doxorubicin and surface-functionalized with transferrin (to allow for tumor targeting) reduced glioma growth in a mouse model by almost 70%. The nanobubbles were burst by exposing to high intensity focused ultrasound (HIFU) to bypass the blood–brain barrier by generating a cavitation effect. This single nanocomposite, combining nanomaterials, chemotherapeutic agents and MRI contrast agents all together for the first time, was able to cross the blood–brain barrier, target the tumor cells and allow imaging of the brain tumor. This multimodal system is an example of combining several strategies to enhance the success rate of therapy [60].
The physicochemical properties of nanomaterials enable them to deliver therapeutic agents to the brain via drug encapsulation or by surface modification. The clinical use of the nanomaterial drug delivery systems is limited by the poorly controlled accumulation and distribution of particles in and around the specific target site [61,62]. The use of multiple therapeutic strategies in combination with nanocarriers may improve the success rate of targeted tumor therapies. However, the potential toxic effects of nanoparticle systems due to aggregation upon introduction into biological systems remains poorly understood. The effect of protein corona formation around nanoparticles and its relevance regarding their biological activity is also being extensively investigated [63].

2.4. Peptide Based Therapeutics

There are three main types of peptide-based therapeutics—tumor homing peptides, peptides targeting aberrant cellular signaling pathways and cell-penetrating peptides [64]. GBM cells have increased expression of membrane proteins that are responsible for cellular function and maintenance, protein synthesis, intercellular signaling, cell movement, and antigen presentation [8,65]. Tumor homing peptides can bind to specific molecular targets on the surface of GBM cells and will be taken up by the cells by endocytosis [66]. The binding process of these peptides is faster than antibodies. These can also be used for in vivo tumor imaging. Binding of some of these peptides can also enhance or antagonize signal transduction pathways in cancer cells or tumor tissues. Peptides and their derivatives targeting aberrant cellular signaling pathways can improve the efficacy of tumor therapy by increased selectivity in their interaction with the oncogenic pathways [64]. Cell-penetrating peptides (CPPs) are small, basic, positively charged peptide derivatives that can pass through the cell membrane [64,67].
Highly selective tumor-targeting peptides obtained using a biopanning phage display library directed against GBM cells were able to cross the BBB and deliver the oncolytic virus VSVΔM51 to GBM in a mouse model in vivo [68]. These peptides, when delivered in combination with gadolinium, also enabled the visualization of the tumors via MRI. The use of peptides directed against multiple targets provides a mechanism for addressing the heterogeneity of the tumor while nevertheless allowing for tumor specific oncolytic virus delivery.
Self-assembled spherical nanoparticles containing a peptide probe (Cy5.5-SAPD-99mTc) that combines tumor homing ability with mitochondria targeting was found to have promising theranostic possibilities owing to the enhanced apoptosis in GBM cells coupled with imaging functionality [69]. Incorporating both tumor-homing and mitochondria-targeting components helps to increase the specificity of drug delivery.
Peptide derivatives of rabies virus glycoproteins, RVG29 and RVG15-liposome, were used to deliver anticancer chemotherapeutic docetaxel nanoparticles and paclitaxel-cholesterol to glioma-bearing ICR mice with a positive effect on animal survival [70,71]. The RVG peptides target the nicotinic acetylcholine receptor, the increased expression of which is noted in the hypoxic and ischemic conditions within the tumor microenvironment. Administration of RVG peptides thus aids in tumor-specific chemotherapeutic delivery.
WSW (also called PhrCACET1) is a tumor-targeting peptide (derived from Clostridium acetobutylicum) that was fused to paclitaxel nanosuspensions and used to target GBM cell membranes in a glioma mouse model. The use of WSW induced apoptosis and prolonged survival of the animals [72]. By combining BBB penetration with tumor targeting, this biomimetic drug delivery system has enhanced tumor-targeting specificity.
The use of polydopamine (PDA)-coated zein-curcumin nanoparticles functionalized with the peptide G23 inhibited cell proliferation and migration in glioma cells in vitro [73]. Here, the peptide G23 binds to ganglioside GM-1 and enables crossing of the BBB. The anti-inflammatory, antimicrobial, and anticancer activities of curcumin have been widely demonstrated [74,75,76].
A dual peptide nanocomplex created by combining SynB3 (a cell penetration peptide) with PVGLIG (an MMP-2 sensitive peptide) and paclitaxel inhibited cell migration and invasion in multiple GBM cell lines, suppressed GBM tumor growth in vivo, and increased overall survival in a mouse model of GBM [77]. The aberrant expression of matrix metalloproteinases (MMPs) has been widely reported in tumors, and the addition of an MMP-sensitive peptide increased the tumor specificity of the drug cargo in this system.
To enhance the membrane permeability of peptides, membrane receptors such as low-density lipoprotein receptor, IL-4 receptor, and transferrin receptor which are abundantly expressed on GBM cells have been used to direct the delivery of tumor-homing peptides to brain malignancies, utilizing receptor-mediated transcytosis [78,79,80]. Peptide-mediated drug delivery is limited by the poor in vivo stability due to the proteolytic degradation of peptides in the circulation when administered systemically. In addition, the short half-life of peptides results in limited bioavailability. This can be overcome by chemical modification or conjugation with macromolecules or nanocarriers with greater target specificity [64]. Identifying additional GBM-targeting peptides is needed to further exploit the benefits of this mode of drug delivery.

2.5. Focused Ultrasound

Focused ultrasound (FUS) is an image-guided, noninvasive method to transiently open the blood barrier, thereby enhancing the efficacy of therapeutic delivery to GBM. FUS can be used to reversibly disrupt the BBB without irreversible tissue damage [81]. The use of FUS in combination with circulating microbubbles works by creating mechanical cavitation effects that transiently disrupt the BBB [82]. In addition to the cavitation effects and thermal ablation, FUS may also work via immunomodulation [83]. Future concurrent application of noninvasive FUS along with other modes of therapeutic drug delivery to GBM is promising.
By increasing BBB permeability, FUS can enhance the delivery of varied therapeutic agents to the tumor. For example, FUS-induced disruption of the BBB enhanced the local delivery of temozolomide to tumors and increased the overall survival of rats harboring experimentally induced gliomas [84]. FUS application followed by BCNU administration resulted in a reduced rate of tumor growth and improved survival in rats [85]. In a mouse model bearing temozolomide-resistant glioma, low-intensity fluorescent ultrasound (LIFU) was used to deliver a liposomal O6-(4-bromothenyl)guanine (O6BTG) derivative that inactivates MGMT [86]. Because MGMT enhances DNA repair in tumor cells, MGMT silencing has been associated with more favorable outcomes after temozolomide treatment [87].
The use of imaging techniques along with FUS increases the rate of drug delivery to specifically identified tumor tissues. MRI guided FUS (MRgFUS) was used to achieve a higher tissue delivery of liposome-encapsulated doxorubicin in rats, temozolomide in mice, and cisplatin-conjugated gold nanoparticles in mice [84,88,89]. In an additional study, MRgFUS was used to deliver the intravenously administered monoclonal antibody, trastzumab, to Her2-positive intracranial metastases in breast cancer patients without any adverse events [90].
Using this noninvasive technique, relatively low systemic doses of therapeutic agents can be used, thereby reducing systemic toxic effects [91]. Transient application of FUS only results in a transient opening of the BBB and does not lead to long-term BBB defects [25]. FUS has the potential to enhance therapeutic drug efficacy against GBM because it is noninvasive and provides reproducible enhancements in drug delivery in early investigational studies. Clinical trials using FUS in GBM are currently underway [81,92,93]. Nevertheless, FUS is not immune to side effects which include edema, intracerebral hemorrhage, and uncontrolled thermal injury in brain [94].

3. Conclusions

Molecular and cellular heterogeneity, GBM cell dispersal and the BBB are critical constraints limiting the efficacy of anti-GBM drug therapy. Applications of CED, controlled-release systems, nanomaterial systems, peptide-based therapeutics and focused ultrasound for drug delivery to tumor enhance survival with reduced toxicity in animal studies (Table 1). Despite currently available treatments, the highly invasive GBM continues to be a deadly disease without cure in patients. Therefore, clinical trials that combine currently available therapies with the novel drug delivery approaches discussed here may enhance the effectiveness of molecular therapeutics in GBM.

Author Contributions

E.N.M. prepared the manuscript. B.C.B., H.W.Y., R.S.C. and M.D.J. critically reviewed the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

The Maroun Semaan Professorship and the University of Massachusetts Chan Medical School.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

GBMGlioblastoma multiforme
WHOWorld Health Organization
IDHIsocitrate dehydrogenase
EGFREpidermal growth factor receptor
MGMTO6-methylguanine DNA methyltransferase
PTNPleiotrophin
CDKN2ACyclin Dependent Kinase Inhibitor 2A
MDM2Mouse double minute 2 homolog
NF1Neurofibromatosis type 1
PDGFRAPlatelet Derived Growth Factor Receptor Alpha
GSCGlioblastoma stem cells
NSCNeural stem cell
BEMCBrain microvascular endothelial cells
ATPAdenosine triphosphate
PDE5Phosphodiesterase 5
FUSFocused ultrasound
CEDConvection enhanced delivery
EPREnhanced permeability and retention
ROSReactive oxygen species
PCLPoly(ε-caprolactone)
GHKglycyl-L-histidyl-L-lysine
CPPCell penetrating peptide
PDAPolydopamine
MMPMatrix Metalloproteinase
MRIMagnetic resonance imaging
MRgFUSMagnetic resonance guided focused ultrasound
LIFULow intensity fluorescent ultrasound
HIFUHigh intensity fluorescent ultrasound

References

  1. Schiffer, D.; Annovazzi, L.; Casalone, C.; Corona, C.; Mellai, M. Glioblastoma: Microenvironment and Niche Concept. Cancers 2018, 11, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hanif, F.; Muzaffar, K.; Perveen, K.; Malhi, S.M.; Simjee, S.U. Glioblastoma Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical Presentation and Treatment. Asian Pac. J. Cancer Prev. 2017, 18, 3–9. [Google Scholar] [CrossRef] [PubMed]
  3. Louis, D.N.; Perry, A.; Reifenberger, G.; von Deimling, A.; Cavenee, W.K.; Ohgaki, H.; Wiestler O., D.; Kleihues, P.; Ellison, D.W. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 2016, 131, 803–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. SEER*Explorer Application. Available online: https://seer.cancer.gov/explorer/application.html?site=661&data_type=4&graph_type=5&compareBy=sex&chk_sex_3=3&chk_sex_2=2&series=9&race=1&age_range=1&stage=101&advopt_precision=1&advopt_show_ci=on&advopt_display=2 (accessed on 13 October 2021).
  5. Tamimi, A.F.; Juweid, M. Epidemiology and Outcome of Glioblastoma. In Glioblastoma; De Vleeschouwer, S., Ed.; Codon Publications: Brisbane, Australia, 2017. Available online: http://www.ncbi.nlm.nih.gov/books/NBK470003/ (accessed on 29 April 2021).
  6. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and Molecular Prognostic Review of Glioblastoma. Cancer Epidemiol. Prev Biomark. 2014, 23, 1985–1996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Koshy, M.; Villano, J.L.; Dolecek, T.A.; Howard, A.; Mahmood, U.; Chmura, S.J.; Weichselbaum, R.R.; McCarthy, B.J. Improved survival time trends for glioblastoma using the SEER 17 population-based registries. J. Neurooncol. 2012, 107, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Shergalis, A.; Bankhead, A.; Luesakul, U.; Muangsin, N.; Neamati, N. Current Challenges and Opportunities in Treating Glioblastoma. Pharm. Rev. 2018, 70, 412–445. [Google Scholar] [CrossRef] [Green Version]
  9. Orr, B.A.; Clay, M.R.; Pinto, E.M.; Kesserwan, C. An update on the central nervous system manifestations of Li–Fraumeni syndrome. Acta Neuropathol 2020, 139, 669–687. [Google Scholar] [CrossRef]
  10. Han, S.; Liu, Y.; Cai, S.J.; Qian, M.; Ding, J.; Larion, M.; Gilbert, M.R.; Yang, C. IDH mutation in glioma: Molecular mechanisms and potential therapeutic targets. Br. J. Cancer 2020, 122, 1580–1589. [Google Scholar] [CrossRef]
  11. Bastiancich, C.; Danhier, P.; Préat, V.; Danhier, F. Anticancer drug-loaded hydrogels as drug delivery systems for the local treatment of glioblastoma. J. Control. Release 2016, 243, 29–42. [Google Scholar] [CrossRef]
  12. Janjua, T.I.; Rewatkar, P.; Ahmed-Cox, A.; Saeed, I.; Mansfeld, F.M.; Kulshreshtha, R.; Kumeria, T.; Ziegler, D.S.; Kavallaris, M.; Mazzieri, R.; et al. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv. Drug Deliv Rev. 2021, 171, 108–138. [Google Scholar] [CrossRef]
  13. Piraino, S.W.; Thomas, V.; O’Donovan, P.; Furney, S.J. Mutations: Driver Versus Passenger. In Encyclopedia of Cancer, 3rd ed.; Boffetta, P., Hainaut, P., Eds.; Academic Press: Salt Lake City, UT, USA, 2019; pp. 551–562. [Google Scholar] [CrossRef]
  14. Friedmann-Morvinski, D. Glioblastoma Heterogeneity and Cancer Cell Plasticity. Crit Rev. Oncog. 2014, 19, 327–336. [Google Scholar] [CrossRef] [PubMed]
  15. Aum, D.J.; Kim, D.H.; Beaumont, T.L.; Leuthardt, E.C.; Dunn, G.P.; Kim, A.H. Molecular and cellular heterogeneity: The hallmark of glioblastoma. Neurosurg. Focus 2014, 37, E11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Kleihues, P.; Ohgaki, H. Primary and secondary glioblastomas: From concept to clinical diagnosis. Neuro-Oncology 1999, 1, 44–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Verhaak, R.G.W.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. An integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR and NF1. Cancer Cell. 2010, 17, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Couturier, C.P.; Ayyadhury, S.; Le, P.U.; Nadaf, J.; Monlong, J.; Riva, G.; Allache, R.; Baig, S.; Yan, X.; Bourgey, M.; et al. Single-cell RNA-seq reveals that glioblastoma recapitulates a normal neurodevelopmental hierarchy. Nat. Commun. 2020, 11, 3406. [Google Scholar] [CrossRef]
  19. Guelfi, S.; Duffau, H.; Bauchet, L.; Rothhut, B.; Hugnot, J.P. Vascular Transdifferentiation in the CNS: A Focus on Neural and Glioblastoma Stem-Like Cells. Stem Cells Int. 2016, 2016, e2759403. [Google Scholar] [CrossRef] [Green Version]
  20. Matarredona, E.R.; Pastor, A.M. Neural Stem Cells of the Subventricular Zone as the Origin of Human Glioblastoma Stem Cells. Therapeutic Implications. Front. Oncol. 2019, 9, 779. [Google Scholar] [CrossRef]
  21. Auffinger, B.; Spencer, D.; Pytel, P.; Ahmed, A.U.; Lesniak, M.S. The role of glioma stem cells in chemotherapy resistance and glioblastoma multiforme recurrence. Expert Rev. Neurother. 2015, 15, 741–752. [Google Scholar] [CrossRef] [Green Version]
  22. Luo, H.; Shusta, E.V. Blood-Brain Barrier Modulation to Improve Glioma Drug Delivery. Pharmaceutics 2020, 12, 1085. [Google Scholar] [CrossRef]
  23. Dong, X. Current Strategies for Brain Drug Delivery. Theranostics 2018, 8, 1481–1493. [Google Scholar] [CrossRef]
  24. Daneman, R.; Prat, A. The Blood–Brain Barrier. Cold Spring Harb. Perspect Biol. 2015, 7, a020412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Burgess, A.; Shah, K.; Hough, O.; Hynynen, K. Focused ultrasound-mediated drug delivery through the blood-brain barrier. Expert Rev. Neurother. 2015, 15, 477–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Cao, Y.; Sundgren, P.C.; Tsien, C.I.; Chenevert, T.T.; Junck, L. Physiologic and metabolic magnetic resonance imaging in gliomas. J. Clin. Oncol. 2006, 24, 1228–1235. [Google Scholar] [CrossRef]
  27. Abbott, N.J.; Patabendige, A.A.K.; Dolman, D.E.M.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  28. Li, J.; Zheng, M.; Shimoni, O.; Banks, W.A.; Bush, A.I.; Gamble, J.R.; Shi, B. Development of Novel Therapeutics Targeting the Blood–Brain Barrier: From Barrier to Carrier. Adv. Sci. 2021, 8, 2101090. [Google Scholar] [CrossRef]
  29. Rapoport, S.I. Osmotic opening of the blood-brain barrier: Principles, mechanism, and therapeutic applications. Cell. Mol. Neurobiol. 2000, 20, 217–230. [Google Scholar] [CrossRef] [PubMed]
  30. Wala, K.; Szlasa, W.; Saczko, J.; Rudno-Rudzińska, J.; Kulbacka, J. Modulation of Blood–Brain Barrier Permeability by Activating Adenosine A2 Receptors in Oncological Treatment. Biomolecules 2021, 11, 633. [Google Scholar] [CrossRef]
  31. Sawyer, A.J.; Piepmeier, J.M.; Saltzman, W.M. New Methods for Direct Delivery of Chemotherapy for Treating Brain Tumors. Yale J. Biol. Med. 2006, 79, 141–152. [Google Scholar]
  32. Zhou, J.; Atsina, K.B.; Himes, B.T.; Strohbehn, G.W.; Saltzman, W.M. Novel Delivery Strategies for Glioblastoma. Cancer J. Sudbury Mass. 2012, 18, 89–99. [Google Scholar] [CrossRef] [Green Version]
  33. Yeini, E.; Ofek, P.; Albeck, N.; Ajamil, D.R.; Neufeld, L.; Eldar-Boock, A.; Kleiner, R.; Vaskovich, D.; Koshrovski-Michael, S.; Dangoor, S.I.; et al. Targeting Glioblastoma: Advances in Drug Delivery and Novel Therapeutic Approaches. Adv. Ther. 2021, 4, 2000124. [Google Scholar] [CrossRef]
  34. Shapira-Furman, T.; Serra, R.; Gorelick, N.; Doglioli, M.; Tagliaferri, V.; Cecia, A.; Peters, M.; Kumar, A.; Rottenberg, Y.; Langer, R.; et al. Biodegradable wafers releasing Temozolomide and Carmustine for the treatment of brain cancer. J. Control. Release 2019, 295, 93–101. [Google Scholar] [CrossRef] [PubMed]
  35. Tyler, B.; Wadsworth, S.; Recinos, V.; Mehta, V.; Vellimana, A.; Li, K.; Rosenblatt, J.; Do, H.; Gallia, G.L.; Siu, I.M.; et al. Local delivery of rapamycin: A toxicity and efficacy study in an experimental malignant glioma model in rats. Neuro-Oncology 2011, 13, 700–709. [Google Scholar] [CrossRef] [PubMed]
  36. Dluska, E.; Markowska-Radomska, A.; Metera, A.; Ordak, M. Multiple Emulsions as a Biomaterial-Based Delivery System for the Controlled Release of an Anti-cancer Drug. J. Phys. Conf. Ser. 2020, 1681, 012021. [Google Scholar] [CrossRef]
  37. Zhao, M.; Bozzato, E.; Joudiou, N.; Ghiassinejad, S.; Danhier, F.; Gallez, B.; Préat, V. Codelivery of paclitaxel and temozolomide through a photopolymerizable hydrogel prevents glioblastoma recurrence after surgical resection. J. Control. Release 2019, 309, 72–81. [Google Scholar] [CrossRef]
  38. Basso, J.; Miranda, A.; Nunes, S.; Cova, T.; Sousa, J.; Vitorino, C.; Pais, A. Hydrogel-Based Drug Delivery Nanosystems for the Treatment of Brain Tumors. Gels 2018, 4, 62. [Google Scholar] [CrossRef] [Green Version]
  39. Jahangiri, A.; Chin, A.T.; Flanigan, P.M.; Chen, R.; Bankiewicz, K.; Aghi, M.K. Convection-enhanced delivery in glioblastoma: A review of preclinical and clinical studies. J. Neurosurg. 2017, 126, 191–200. [Google Scholar] [CrossRef] [Green Version]
  40. Enríquez Pérez, J.; Kopecky, J.; Visse, E.; Darabi, A.; Siesjö, P. Convection-enhanced delivery of temozolomide and whole cell tumor immunizations in GL261 and KR158 experimental mouse gliomas. BMC Cancer 2020, 20, 7. [Google Scholar] [CrossRef] [Green Version]
  41. Wang, J.L.; Barth, R.F.; Cavaliere, R.; Puduvalli, V.K.; Giglio, P.; Lonser, R.R.; Elder, J.B. Phase I trial of intracerebral convection-enhanced delivery of carboplatin for treatment of recurrent high-grade gliomas. PLoS ONE 2020, 15, e0244383. [Google Scholar] [CrossRef]
  42. Muldoon, L.L.; Soussain, C.; Jahnke, K.; Johanson, C.; Siegal, T.; Smith, Q.R.; Hall, W.A.; Hynynen, K.; Senter, P.D.; Peereboom, D.M.; et al. Chemotherapy Delivery Issues in Central Nervous System Malignancy: A Reality Check. J. Clin. Oncol. 2007, 25, 2295–2305. [Google Scholar] [CrossRef] [Green Version]
  43. Hadjipanayis, C.G.; Machaidze, R.; Kaluzova, M.; Wang, L.; Schuette, A.J.; Chen, H.; Wu, X.; Mao, H. EGFRvIII Antibody–Conjugated Iron Oxide Nanoparticles for Magnetic Resonance Imaging–Guided Convection-Enhanced Delivery and Targeted Therapy of Glioblastoma. Cancer Res. 2010, 70, 6303–6312. [Google Scholar] [CrossRef] [Green Version]
  44. Nwagwu, C.D.; Immidisetti, A.V.; Jiang, M.Y.; Adeagbo, O.; Adamson, D.C.; Carbonell, A.M. Convection Enhanced Delivery in the Setting of High-Grade Gliomas. Pharmaceutics 2021, 13, 561. [Google Scholar] [CrossRef] [PubMed]
  45. Mehta, A.M.; Sonabend, A.M.; Bruce, J.N. Convection-Enhanced Delivery. Neurotherapeutics 2017, 14, 358–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Tran, S.; DeGiovanni, P.J.; Piel, B.; Rai, P. Cancer nanomedicine: A review of recent success in drug delivery. Clin. Transl. Med. 2017, 6, e44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Wicki, A.; Witzigmann, D.; Balasubramanian, V.; Huwyler, J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J. Control. Release 2015, 200, 138–157. [Google Scholar] [CrossRef]
  48. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
  49. Michael, J.S.; Lee, B.S.; Zhang, M.; Yu, J.S. Nanotechnology for Treatment of Glioblastoma Multiforme. J. Transl. Intern. Med. 2018, 6, 128–133. [Google Scholar] [CrossRef] [Green Version]
  50. Kalyane, D.; Raval, N.; Maheshwari, R.; Tambe, V.; Kalia, K.; Tekade, R.K. Employment of enhanced permeability and retention effect (EPR): Nanoparticle-based precision tools for targeting of therapeutic and diagnostic agent in cancer. Mater. Sci. Eng. C. 2019, 98, 1252–1276. [Google Scholar] [CrossRef]
  51. Greish, K. Enhanced Permeability and Retention (EPR) Effect for Anticancer Nanomedicine Drug Targeting. In Cancer Nanotechnology: Methods and Protocols. Methods in Molecular Biology; Grobmyer, S.R., Moudgil, B.M., Eds.; Humana Press: New York, NY, USA, 2010; pp. 25–37. [Google Scholar] [CrossRef]
  52. Brannon-Peppas, L.; Blanchette, J.O. Nanoparticle and targeted systems for cancer therapy. Adv. Drug Deliv. Rev. 2004, 56, 1649–1659. [Google Scholar] [CrossRef]
  53. Alphandéry, E. Nano-Therapies for Glioblastoma Treatment. Cancers 2020, 12, 242. [Google Scholar] [CrossRef] [Green Version]
  54. Mout, R.; Moyano, D.F.; Rana, S.; Rotello, V.M. Surface functionalization of nanoparticles for nanomedicine. Chem. Soc. Rev. 2012, 41, 2539–2544. [Google Scholar] [CrossRef]
  55. Taiarol, L.; Formicola, B.; Magro, R.D.; Sesana, S.; Re, F. An update of nanoparticle-based approaches for glioblastoma multiforme immunotherapy. Nanomedicine 2020, 15, 1861–1871. [Google Scholar] [CrossRef] [PubMed]
  56. Ngobili, T.A.; Daniele, M.A. Nanoparticles and direct immunosuppression. Exp. Biol. Med. 2016, 241, 1064–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Blank, F.; Gerber, P.; Rothen-Rutishauser, B.; Sakulkhu, U.; Salaklang, J.; De Peyer, K.; Gehr, P.; Nicod, L.P.; Hofmann, H.; Geiser, T.; et al. Biomedical nanoparticles modulate specific CD4+ T cell stimulation by inhibition of antigen processing in dendritic cells. Nanotoxicology 2011, 5, 606–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Budama-Kilinc, Y.; Kecel-Gunduz, S.; Cakir-Koc, R.; Aslan, B.; Bicak, B.; Kokcu, Y.; Ozel, A.E.; Akyuz, S. Structural Characterization and Drug Delivery System of Natural Growth-Modulating Peptide Against Glioblastoma Cancer. Int. J. Pept. Res. Ther. 2021, 27, 2015–2028. [Google Scholar] [CrossRef]
  59. Pickart, L.; Vasquez-Soltero, J.M.; Margolina, A. The Effect of the Human Peptide GHK on Gene Expression Relevant to Nervous System Function and Cognitive Decline. Brain Sci. 2017, 7, 20. [Google Scholar] [CrossRef] [Green Version]
  60. Chan, M.H.; Chen, W.; Li, C.H.; Fang, C.Y.; Chang, Y.C.; Wei, D.H.; Liu, R.S.; Hsiao, M. An Advanced In Situ Magnetic Resonance Imaging and Ultrasonic Theranostics Nanocomposite Platform: Crossing the Blood–Brain Barrier and Improving the Suppression of Glioblastoma Using Iron-Platinum Nanoparticles in Nanobubbles. ACS Appl. Mater. Interfaces 2021, 13, 26759–26769. [Google Scholar] [CrossRef]
  61. Pasut, G. Grand Challenges in Nano-Based Drug Delivery. Front. Med. Technol. 2019, 1, 1. Available online: https://www.frontiersin.org/article/10.3389/fmedt.2019.00001 (accessed on 28 January 2022). [CrossRef] [Green Version]
  62. Sharma, D.; Sharma, N.; Pathak, M.; Agrawala, P.K.; Basu, M.; Ojha, H. Chapter 2—Nanotechnology-based drug delivery systems: Challenges and opportunities. In Drug Targeting and Stimuli Sensitive Drug Delivery Systems; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2018; pp. 39–79. [Google Scholar] [CrossRef]
  63. Rampado, R.; Crotti, S.; Caliceti, P.; Pucciarelli, S.; Agostini, M. Recent Advances in Understanding the Protein Corona of Nanoparticles and in the Formulation of “Stealthy” Nanomaterials. Front. Bioeng. Biotechnol. 2020, 8, 166. Available online: https://www.frontiersin.org/article/10.3389/fbioe.2020.00166 (accessed on 26 January 2022). [CrossRef]
  64. Raucher, D. Tumor targeting peptides: Novel therapeutic strategies in glioblastoma. Curr. Opin. Pharmacol. 2019, 47, 14–19. [Google Scholar] [CrossRef]
  65. Polisetty, R.V.; Gautam, P.; Sharma, R.; Harsha, H.C.; Nair, S.C.; Gupta, M.K.; Uppin, M.S.; Challa, S.; Puligopu, A.K.; Ankathi, P.; et al. LC-MS/MS analysis of differentially expressed glioblastoma membrane proteome reveals altered calcium signaling and other protein groups of regulatory functions. Mol. Cell. Proteom. 2012, 11, M111.013565. [Google Scholar] [CrossRef] [Green Version]
  66. Kondo, E.; Iioka, H.; Saito, K. Tumor-homing peptide and its utility for advanced cancer medicine. Cancer Sci. 2021, 112, 2118–2125. [Google Scholar] [CrossRef] [PubMed]
  67. Audrey, G.; Claire, L.C.; Joel, E. Effect of the NFL-TBS.40-63 peptide on canine glioblastoma cells. Int. J. Pharm. 2021, 605, 120811. [Google Scholar] [CrossRef] [PubMed]
  68. Rahn, J.J.; Lun, X.; Jorch, S.K.; Hao, X.; Venugopal, C.; Vora, P.; Ahn, B.Y.; Babes, L.; Alshehri, M.M.; Cairncross, J.; et al. Development of a peptide-based delivery platform for targeting malignant brain tumors. Biomaterials 2020, 252, 120105. [Google Scholar] [CrossRef] [PubMed]
  69. Rizvi, S.F.A.; Shahid, S.; Mu, S.; Zhang, H. Hybridization of tumor homing and mitochondria-targeting peptide domains to design novel dual-imaging self-assembled peptide nanoparticles for theranostic applications. Drug Deliv. Transl. Res. 2021, 1–12. [Google Scholar] [CrossRef] [PubMed]
  70. Hua, H.; Zhang, X.; Mu, H.; Meng, Q.; Jiang, Y.; Wang, Y.; Lu, X.; Wang, A.; Liu, S.; Zhang, Y.; et al. RVG29-modified docetaxel-loaded nanoparticles for brain-targeted glioma therapy. Int. J. Pharm. 2018, 543, 179–189. [Google Scholar] [CrossRef] [PubMed]
  71. Xin, X.; Liu, W.; Zhang, Z.A.; Han, Y.; Qi, L.-L.; Zhang, Y.-Y.; Zhang, X.-T.; Duan, H.-X.; Chen, L.-Q.; Jin, M.-J.; et al. Efficient Anti-Glioma Therapy Through the Brain-Targeted RVG15-Modified Liposomes Loading Paclitaxel-Cholesterol Complex. Int. J. Nanomed. 2021, 16, 5755–5776. [Google Scholar] [CrossRef]
  72. Fan, Y.; Cui, Y.; Hao, W.; Chen, M.; Liu, Q.; Wang, Y.; Yang, M.; Li, Z.; Gong, W.; Song, S.; et al. Carrier-free highly drug-loaded biomimetic nanosuspensions encapsulated by cancer cell membrane based on homology and active targeting for the treatment of glioma. Bioact. Mater. 2021, 6, 4402–4414. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, H.; van Os, W.L.; Tian, X.; Zu, G.; Ribovski, L.; Bron, R.; Bussmann, J.; Kros, A.; Liu, Y.S.; Zuhorn, I. Development of curcumin-loaded zein nanoparticles for transport across the blood–brain barrier and inhibition of glioblastoma cell growth. Biomater. Sci. 2021, 9, 7092–7103. [Google Scholar] [CrossRef]
  74. Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the golden nutraceutical: Multitargeting for multiple chronic diseases. Br. J. Pharmacol. 2017, 174, 1325–1348. [Google Scholar] [CrossRef] [Green Version]
  75. Shabaninejad, Z.; Pourhanifeh, M.H.; Movahedpour, A.; Mottaghi, R.; Nickdasti, A.; Mortezapour, E.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; Sadeghian, M.; et al. Therapeutic potentials of curcumin in the treatment of glioblstoma. Eur. J. Med. Chem. 2020, 188, 112040. [Google Scholar] [CrossRef]
  76. Zorofchian Moghadamtousi, S.; Abdul Kadir, H.; Hassandarvish, P.; Tajik, H.; Abubakar, S.; Zandi, K. A Review on Antibacterial, Antiviral, and Antifungal Activity of Curcumin. BioMed Res. Int. 2014, 2014, e186864. [Google Scholar] [CrossRef] [PubMed]
  77. Hua, D.; Tang, L.; Wang, W.; Tang, S.; Yu, L.; Zhou, X.; Wang, Q.; Sun, C.; Shi, C.; Luo, W.; et al. Improved Antiglioblastoma Activity and BBB Permeability by Conjugation of Paclitaxel to a Cell-Penetrative MMP-2-Cleavable Peptide. Adv. Sci. 2021, 8, 2001960. [Google Scholar] [CrossRef] [PubMed]
  78. Ayo, A.; Laakkonen, P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021, 13, 481. [Google Scholar] [CrossRef] [PubMed]
  79. Demeule, M.; Currie, J.C.; Bertrand, Y.; Ché, C.; Nguyen, T.; Régina, A.; Gabathuler, R.; Castaigne, J.P.; Béliveau, R. Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector angiopep-2. J. Neurochem. 2008, 106, 1534–1544. [Google Scholar] [CrossRef]
  80. Hong, H.-Y.; Lee, H.Y.; Kwak, W.; Yoo, J.; Na, M.-H.; So, I.S.; Kwon, T.-H.; Park, H.-S.; Huh, S.; Oh, G.T.; et al. Phage display selection of peptides that home to atherosclerotic plaques: IL-4 receptor as a candidate target in atherosclerosis. J. Cell. Mol. Med. 2008, 12, 2003–2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Bunevicius, A.; McDannold, N.J.; Golby, A.J. Focused Ultrasound Strategies for Brain Tumor Therapy. Oper. Neurosurg. 2020, 19, 9–18. [Google Scholar] [CrossRef]
  82. Wu, S.K.; Tsai, C.L.; Huang, Y.; Hynynen, K. Focused Ultrasound and Microbubbles-Mediated Drug Delivery to Brain Tumor. Pharmaceutics 2020, 13, 15. [Google Scholar] [CrossRef]
  83. Cohen-Inbar, O.; Xu, Z.; Sheehan, J.P. Focused ultrasound-aided immunomodulation in glioblastoma multiforme: A therapeutic concept. J. Ther. Ultrasound. 2016, 4, 2. [Google Scholar] [CrossRef] [Green Version]
  84. Wei, K.C.; Chu, P.C.; Wang, H.Y.J.; Huang, C.Y.; Chen, P.Y.; Tsai, H.C.; Lu, Y.J.; Lee, P.I.; Tseng, I.C.; Feng, L.Y.; et al. Focused ultrasound-induced blood-brain barrier opening to enhance temozolomide delivery for glioblastoma treatment: A preclinical study. PLoS ONE 2013, 8, e58995. [Google Scholar] [CrossRef] [Green Version]
  85. Liu, H.L.; Hua, M.Y.; Chen, P.Y.; Chu, P.C.; Pan, C.H.; Yang, H.W.; Huang, C.Y.; Wang, J.J.; Yen, T.C.; Wei, K.C. Blood-Brain Barrier Disruption with Focused Ultrasound Enhances Delivery of Chemotherapeutic Drugs for Glioblastoma Treatment. Radiology 2010, 255, 415–425. [Google Scholar] [CrossRef]
  86. Papachristodoulou, A.; Signorell, R.D.; Werner, B.; Brambilla, D.; Luciani, P.; Cavusoglu, M.; Grandjean, J.; Silginer, M.; Rudin, M.; Martin, E.; et al. Chemotherapy sensitization of glioblastoma by focused ultrasound-mediated delivery of therapeutic liposomes. J. Control. Release 2019, 295, 130–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hegi, M.E.; Diserens, A.C.; Gorlia, T.; Hamou, M.F.; de Tribolet, N.; Weller, M.; Kros, J.M.; Hainfellner, J.A.; Mason, W.; Mariani, L.; et al. MGMT Gene Silencing and Benefit from Temozolomide in Glioblastoma. N. Engl. J. Med. 2005, 352, 997–1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Treat, L.H.; McDannold, N.; Vykhodtseva, N.; Zhang, Y.; Tam, K.; Hynynen, K. Targeted delivery of doxorubicin to the rat brain at therapeutic levels using MRI-guided focused ultrasound. Int. J. Cancer 2007, 121, 901–907. [Google Scholar] [CrossRef] [PubMed]
  89. Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M.; et al. Enhancing glioblastoma treatment using cisplatin-gold-nanoparticle conjugates and targeted delivery with magnetic resonance-guided focused ultrasound. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 1137–1148. [Google Scholar] [CrossRef]
  90. Meng, Y.; Reilly, R.M.; Pezo, R.C.; Trudeau, M.; Sahgal, A.; Singnurkar, A.; Perry, J.; Myrehaug, S.; People, C.B.; Davidson, B.; et al. MR-guided focused ultrasound enhances delivery of trastuzumab to Her2-positive brain metastases. Sci. Transl. Med. 2021, 13, eabj4011. [Google Scholar] [CrossRef]
  91. Timbie, K.F.; Mead, B.P.; Price, R.J. Drug and gene delivery across the blood–brain barrier with focused ultrasound. J. Control. Release 2015, 219, 61–75. [Google Scholar] [CrossRef] [Green Version]
  92. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US).. Assessment of Safety and Feasibility of ExAblate Blood-Brain Barrier Disruption for the Treatment of High Grade Glioma in Patients Undergoing Standard Chemotherapy. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03551249 (accessed on 6 December 2021).
  93. ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US).. A Study to Evaluate the Safety and Feasibility of Exablate Model 4000 Type-2 to Temporarily Mediate Blood-Brain Barrier Disruption (BBBD) in Patients with Suspected Infiltrating Glioma in the Setting of Planned Surgical Interventions. 2021. Available online: https://clinicaltrials.gov/ct2/show/NCT03322813 (accessed on 6 December 2021).
  94. Wang, D.; Wang, C.; Wang, L.; Chen, Y. A comprehensive review in improving delivery of small-molecule chemotherapeutic agents overcoming the blood-brain/brain tumor barriers for glioblastoma treatment. Drug Deliv. 2019, 26, 551–565. [Google Scholar] [CrossRef]
Figure 1. Challenges to delivery of therapeutics to glioblastoma and the currently available drug delivery techniques.
Figure 1. Challenges to delivery of therapeutics to glioblastoma and the currently available drug delivery techniques.
Ijms 23 01711 g001
Table 1. Summary of the methods of drug delivery to GBM with the examples discussed in the text.
Table 1. Summary of the methods of drug delivery to GBM with the examples discussed in the text.
Method of Drug DeliverySpecific Examples
Controlled release systemsGliadel [11,31,32,33]
Biodegradable wafers for the combined delivery of temozolomide and carmustine [34]
Biodegradable polymer implants releasing rapamycin [35]
Carboxymethylcellulose biopolymer system delivering rhodamine B [36]
Hydrogel based co-delivery of paclitaxel and temozolomide [37]
Convection enhanced deliveryTemozolomide [40]
Carboplatin [41]
Iron oxide nanoparticles conjugated to epidermal growth factor receptor deletion mutant III antibody (EG-FRVIIIAb)/MRI-guided [43]
Nanomaterial SystemsPoly(ε-caprolactone) (PCL) based nanoparticle system to deliver the natural growth modulating tripeptide GHK (glycyl-L-histidyl-L-lysine) [58]
Nanobubble-based theranostic system consisting of intravenously administered iron-platinum nanoparticles loaded with doxorubicin and surface-functionalized with transferrin [60]
Peptide based therapeuticsTumor targeting peptides delivering deliver the oncolytic virus VSVΔM51, in combination with gadolinium [68]
Self-assembled spherical nanoparticles containing a peptide probe (Cy5.5-SAPD-99mTc) with mitochondria targeting [69]
Peptide derivatives of rabies virus glycoproteins, RVG29 and RVG15-liposome, delivering anticancer chemotherapeutic docetaxel nanoparticles and paclitaxel-cholesterol [70,71]
WSW (also called PhrCACET1) peptide fused to paclitaxel nanosuspensions [72]
Use of polydopamine (PDA)-coated zein-curcumin nanoparticles functionalized with the peptide G23 [73]
Dual peptide nanocomplex created by combining SynB3 (a cell penetration peptide) with PVGLIG (an MMP-2 sensitive peptide) and paclitaxel [77]
Focused ultrasoundTemozolomide [84]
BCNU [85]
Liposomal O6-(4-bromothenyl)guanine (O6BTG) [86]
Liposome-encapsulated doxorubicin [88]
Cisplatin conjugated gold nanoparticles [89]
Trastzumab [90]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mathew, E.N.; Berry, B.C.; Yang, H.W.; Carroll, R.S.; Johnson, M.D. Delivering Therapeutics to Glioblastoma: Overcoming Biological Constraints. Int. J. Mol. Sci. 2022, 23, 1711. https://doi.org/10.3390/ijms23031711

AMA Style

Mathew EN, Berry BC, Yang HW, Carroll RS, Johnson MD. Delivering Therapeutics to Glioblastoma: Overcoming Biological Constraints. International Journal of Molecular Sciences. 2022; 23(3):1711. https://doi.org/10.3390/ijms23031711

Chicago/Turabian Style

Mathew, Elza N., Bethany C. Berry, Hong Wei Yang, Rona S. Carroll, and Mark D. Johnson. 2022. "Delivering Therapeutics to Glioblastoma: Overcoming Biological Constraints" International Journal of Molecular Sciences 23, no. 3: 1711. https://doi.org/10.3390/ijms23031711

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop