Skip to main content
Technology in Cancer Research & Treatment logoLink to Technology in Cancer Research & Treatment
. 2022 Mar 7;21:15330338221080974. doi: 10.1177/15330338221080974

Nanocarriers Call the Last Shot in the Treatment of Brain Cancers

Amin Mehrabian 1,2,3,4, Mohammad Mashreghi 2,3, Saba Dadpour 2,5, Ali Badiee 2,3, Leila Arabi 2,3, Seyedeh Hoda Alavizadeh 2,3, Seyedeh Alia Moosavian 2,3,, Mahmoud Reza Jaafari 1,2,3,
PMCID: PMC8905056  PMID: 35253549

Abstract

Our brain is protected by physio-biological barriers. The blood–brain barrier (BBB) main mechanism of protection relates to the abundance of tight junctions (TJs) and efflux pumps. Although BBB is crucial for healthy brain protection against toxins, it also leads to failure in a devastating disease like brain cancer. Recently, nanocarriers have been shown to pass through the BBB and improve patients’ survival rates, thus becoming promising treatment strategies. Among nanocarriers, inorganic nanocarriers, solid lipid nanoparticles, liposomes, polymers, micelles, and dendrimers have reached clinical trials after delivering promising results in preclinical investigations. The size of these nanocarriers is between 10 and 1000nm and is modified by surface attachment of proteins, peptides, antibodies, or surfactants. Multiple research groups have reported transcellular entrance as the main mechanism allowing for these nanocarriers to cross BBB. Transport proteins and transcellular lipophilic pathways exist in BBB for small and lipophilic molecules. Nanocarriers cannot enter via the paracellular route, which is limited to water-soluble agents due to the TJs and their small pore size. There are currently several nanocarriers in clinical trials for the treatment of brain cancer. This article reviews challenges as well as fitting attributes of nanocarriers for brain tumor treatment in preclinical and clinical studies.

Keywords: nanocarrier, brain drug delivery, brain cancer, blood–brain barrier, nanomedicine

Introduction

Structural or functional impairments to the brain or spinal cord including trauma, infection, inflammation, tumors, degeneration, and autoimmune disorders are classified as central nervous system (CNS) disorders.1,2 These conditions can lead to serious cognition and physiological impairments and may prove fatal in certain cases. 3 Brain tumors have a high fatality rate and can seriously affect and devastate lives. Despite many improvements in treatment protocols, drug delivery remains a major challenge, and treatment options are limited. The most malignant brain tumors begin with genetic mutations that impair and dysregulate cell function and division.46 Brain tumors are classified as malignant and benign, and further subcategorized as primary and secondary. Whereas primary tumors are caused by the division of brain cells only, secondary tumors develop from the metastasis of other organs to the brain and are also hardly treatable. Primary tumors unlike secondary types start and involve the brain cells. The glioblastoma multiform (GBM), with a treatment-refractory disorder with a patients’ short lifespan, cognitive impairments, and high mortality rate, is one of the most common primary brain tumors.711 The patient’s survival percentage with grade IV glioblastoma is reported to be 4.5% for 5 years by World Health Organization (WHO). Metastatic brain tumor as the secondary tumor type has almost 6 months overall survival. The median survival was at its highest with surgery which was followed by radiation therapy.1214

The surgical resection plus radiotherapy, as well as adjuvant chemotherapy frequently, has been comprised by the treatment guidelines (Figure 1).1517 With the lack of clear tumor margins, precise anatomical location, and biases that exist during the surgery, chemotherapy has been one of the most effective approaches. Despite recent progress in the development of effective chemotherapy agents and efforts on improved drug formulations, the tumor’s molecular heterogeneity has made the treatment process and the resulting outcomes more complicated. 18 The delivery of chemotherapy agents to the brain tissue faces the challenge of crossing the anatomical and physiological barrier, the blood–brain barrier (BBB) being the most important one. The BBB is a highly selective barrier (hardly permeable) that protects the brain cells against blood-circulating agents such as pathogens and toxins. Many approaches to overcoming BBB have been suggested in the literature. One of the promising applied approaches lies in the nanotechnology.1921

Figure 1.

Figure 1.

Current methods in the treatment of brain tumors. These methods are including surgical resection plus radiotherapy, as well as adjuvant chemotherapy.

Nanotechnology, particularly nano-drug delivery systems, is emerging as promising tools in the cancer diagnostics and therapeutics.22,23 Based on the drug-loading methods and nanoparticle surface, size, and zeta potential modifications, the effector molecules can be encapsulated, adsorbed, or attached to the nano-drug delivery system. To date, the most successful nanocarriers based on clinical trials are inorganic nanoparticles (such as metal, metal oxide, carbon, and silica particles), liposomes, micelles, dendrimers, and polymers. Assuming that blood capillaries and cells range between 6 to 9 and 10 to 20 μm, respectively, almost all types of nano-drug delivery systems can reach and deliver therapeutics into organs and cells. Specifically targeting the BBB receptors by surface-functionalized nano-drug delivery systems makes them ideal candidates for the brain cancer drug delivery.2427

This study aims to discuss the challenges regarding brain tumor treatment and to review the current application of nanotechnology in preclinical and clinical studies for this disease category. With careful examination of the literature, the physiological and biological aspects of the BBB were summarized along with the nano-drug delivery strategies that have been reviewed in detail based on the nanostructures. Finally, the majority of all ongoing nano-drug delivery systems which have made their way through the clinical trials were reviewed as the realistic perspective of nanomedicine for brain cancer drug delivery systems.

Blood-Brain Barrier

In 1885, Ehrlich 28 and his colleagues demonstrated the presence of barriers between CNS and the periphery using brain parenchyma staining via intravenous injection. One of Ehrlich’s students, Edwin Goldmann completed Ehrlich’s dye experiments by directly administrating trypan blue directly into the cerebrospinal fluid (CSF). 29 The term BBB generally refers to the distinct characteristics of continuous nonfenestrated brain microvasculature. 30 This unique feature is the consequence of physical barrier properties, molecular barrier properties, as well as specific transporters.3134 The cells, molecules, and ions entrance are suppressed by the protective role of the BBB. Furthermore, it is almost an impermeable barrier for drugs. Even though the BBB makes uniform coverage between the brain parenchyma and blood vessels interface, the circumventricular organs link the CNS and peripheral blood vessels. 35 Blood vessels are made up of 2 cell types: endothelial cells (ECs) that build up blood vessels, and mural cells including vascular smooth muscle cells and pericytes (PCs), which are placed on the outer layer of the ECs. While ECs are primarily responsible for the BBB characteristics, the function and maintenance of the BBB depend on interactions between ECs, PCs, and astrocytes (Figure 2).

Figure 2.

Figure 2.

Schematic representation of the BBB. Endothelial cells are made which are tightly attached via tight junctions. Blood vessels, and mural cells including vascular smooth muscle cells and PCs, are placed on the outer layer. The PCs have incompletely covered the abluminal microvascular side that is attached to the vascular basement membrane. Astrocytes are polarized glial cell types, cover the entire vessel’s tube.

Abbreviations: BBB, blood–brain barrier; PCs, pericytes.

Simple squamous epithelial cells are the bricks of the blood vessel walls. CNS microvascular ECs are 39% thinner than muscle ECs and also there is a 200 nm diameter between the luminal and abluminal surface. 36 The main role of ECs is to limit the entry of cells, molecules, and ions to the brain. The EC tight junctions not only block the paracellular pathway but also vesicle-mediated transcellular flux too.3639 As a result of paracellular and transcellular restrictions, blood–brain transportation is highly controlled.4042 Higher amounts of mitochondria and very low levels of leukocyte adhesion molecules are other specific limiting features.

The PCs incompletely cover the abluminal microvascular side that is attached to the vascular basement membrane. 43 PC cells with their contractile proteins control the capillary diameter. 44 In comparison with other tissues like muscles, in the CNS PCs provide the most coverage. The EC-to-PC content ratio is between 1:1 and 1:3 in CNS microvascular versus 100:1 in muscles. 45

Astrocytes are polarized glial cells that cover the entire vessel’s tube. 46 They not only connect neuronal cells with the blood vessels but also reflect the neuronal signals on the microvessels’ blood flow. This includes contraction/dilation regulation of the vascular smooth muscle cells next to capillaries and arterioles, respectively. 32

The main CNS endothelial cell transporters are efflux and nutrient types. 34 Efflux transporters, including MRPs, Mdr1, and BCRP take advantage of ATP hydrolysis to actively transport different biological membranes. Nutrient transporters facilitate the transportation of nutrients against their concentration gradient. Most of these belong to the family of solute carrier transporters, including slc16a1 (lactate, pyruvate), slc2a1 (glucose), slc7a5 (neutral amino acids, L-DOPA), and slc7a1 (cationic amino acids).4749

Nanotechnology Approaches to Overcome Brain Drug Delivery Challenges

Most of the small lipophilic molecules, less than 400 to 500 Da, can pass through the BBB. 50 Of the greater than 7000 drugs for insomnia, depression, and schizophrenia that were assessed in comprehensive medicinal chemistry (CMC) database study, 51 only 5% reached the CNS and averaged 357 Da. In another study, 12% of drugs were activated upon entering the CNS while only 1% of them were of use for nonpsychotic disorders (neurosis). Antibiotics, antineoplastics, and neuropeptides are common examples of compounds with limited transfer rates through the BBB. 52 To that end, the objective for many brain drug delivery systems is concerned with targeting the BBB receptors.35,53

Generally, invasive and noninvasive methods are 2 major interventions to allow passage through BBB. The invasive procedures occur with transient disruption of BBB by chemical, biological, and physical stimuli. These methods are expensive and have proven to be highly risky. As a result, they are not preferable for the brain drug delivery enhancement.5457 In contrast, noninvasive methods have proven more effective at providing a relatively harmless drug-to-brain delivery system. Furthermore, a blood-to-brain strategy that improves the BBB permeability and facilitates drug-carrier conjugation minimizes the mentioned drawbacks.58,59 It has been demonstrated that the transcytosis mechanism by the BBB cells supports active drug transportation due to the sizable presence of mitochondria. Various nanocarriers have been thought to be able to overcome the BBB, potentiating drug delivery to ischemic lesions and various tumors in CNS. Many research and review papers have aimed to comprehensively examine the effectiveness of brain drug delivery systems.6062

Generally, the paracellular pathway is considered the common route of entry for small hydrophilic molecules, and the transcellular pathway is the preferable route for the transport of small nutritional or therapeutic compounds.6365 Unfortunately, due to the physiologic limitations of the BBB, both pathways apply to a small selection of compounds. Other more feasible and commonly used modes of transportation include a carrier or receptor-mediated transcytosis. Briefly, the endosomal formation following carrier conformational change due to the concentration gradient elucidates pathways. These established pathways to get through the BBB are illustrated in Figure 3.6671

Figure 3.

Figure 3.

Nanocarrier and brain delivery. Various types of nanocarriers including inorganic nanoparticles, dendrimers, SLNs, polymeric nanoparticles, micelles, exosomes, minicells, and liposomes encounter 4 types of transport mechanisms including transcellular, receptor-mediated, paracellular, and carrier-mediated transport to pass through BBB.

Abbreviations: BBB, blood–brain barrier; SLNs, solid lipid nanoparticles.

Nanotechnology Platforms for Brain Cancer Drug Delivery

Inorganic Nanomaterials

These types of nanomaterials based on silica, carbon, metal, and metal oxide particles are widely used in imaging techniques. Stabilized size and monodispersed formation in the bloodstream, high surface area, and for this reason, ease of functionalizing are just a few positive aspects of inorganic nanomaterials.7275 Silica mesoporous nanoparticles, carbon nanotubes with an ultrahigh surface area, gold and iron oxide nanoparticles, especially superparamagnetic iron oxide nanoparticles (SPIONs), are typical inorganic nanoparticle examples (Table 1). In addition to chemical modification by PEG, lactoferrin, cationic serum albumin, poly(isobutylene-alt-maleic anhydride [PMA])s73,76,77 can be used as chemical modifications. These improve the hydrophilicity and decrease both blood aggregations and the reticuloendothelial system (RES) clearance. Additionally, physical approaches like magnetism as a novel drug delivery mechanism have been used to facilitate passing through BBB. 73 Furthermore, it has been demonstrated that external magnetic forces can effectively cross SPIONs through the BBB.73,78

Table 1.

Preclinical Studies. Nanocarriers That Were Used in Preclinical Studies for Brain Delivery.

Name and materials Advantages Limitations References
Inorganic and are based on silica, carbon, metal, and metal oxide, for example, silica mesoporous nanoparticles, carbon nanotubes, gold nanoparticles, iron oxide nanoparticles especially SPIONs
  • Stabilize size,

  • Monodispersed formulation

  • High surface area,

  • Ease of functionalizing,

  • Physical drug delivery systems like magnetism

  • Low hydrophilicity

  • High blood clearance by RES

  • Hard degradability

  • Undesirable drug delivery

293
Solid lipid nanoparticles (SLN) and are made of lipids and stabilized by surfactants
  • 10-1000 nm size

  • Biocompatible

  • Biodegradable

  • High loading efficiency

  • Functionalized by targeting

  • Not suitable for hydrophilic drugs

  • High clearance by RES

294,295
Polymeric nanoparticles made from natural or synthetic polymers
for example, poly(alky cyanoacrylates), poly(lactic acid), human serum albumin (HAS), and chitosan
  • 1-1000 nm

  • Stable

  • Biodegradable

  • Controlled degradation rate

  • Functionalized by targeting

  • Catabolites and degradation rate should be examined before clinical use because of adverse immunological responses

296,297
Dendrimers mainly are based on PAMAM, PPI, or PLL
  • Structural functional groups

  • Many reaction sites

  • Dual targeting

  • Cationic dendrimers (gene delivery)

  • Endosome destruction (Gene delivery)

  • Uniform size distribution

  • High drug loading capacity

  • Complexity

  • Multi-step synthesis

  • Toxicities and safety

  • High clearance by RES

132,298,299
Micelles are based on amphiphilic block copolymers, a hydrophobic core, and a hydrophilic surface
  • 10-100nm

  • Drug delivery of both lipophilic and hydrophilic compounds

  • Stability and long blood circulation time

  • EPR mechanism

  • Easy and reproducible formulation

  • Sterilization by simple filtration

  • Evading the RES

  • Low stability

  • Premature drug release

  • Immunogenicity

  • Dissociate below CMC

300,301
Exosomes are natural extracellular nanovesicles
  • 30-100 nm

  • Natural biocompatibility

  • Stability

  • Controllable intercellular interactions

  • Not immunogenic

  • No toxicity

  • Lack of standardized exosome separation and purification criteria

  • Uncertain mechanism in cancer

  • Heterogeneity

  • Release modifications

171,172,175177
Minicells are bacterially derived nanoparticles
  • 100-300 nm

  • Multiple targeting ligands for targeting

  • Biocompatible

  • Increased encapsulation efficiency

  • Less drug leakage

  • Stability

  • Release profile

  • Immunogenicity

  • Organ toxicity

  • Further evaluations

184,188,189,198200
Liposomes are based on phospholipids
  • 25-1000 nm

  • Delivery of various molecules: MLVs for extended drug release, LUVs for vaccine and gene delivery, SUV for drug delivery through the endothelial cell layer

  • Targeted drug delivery

  • Both hydrophilic and hydrophobic drug delivery

  • High encapsulation efficiency

  • Biocompatible

  • Biodegradable

  • pH-sensitive formulations

  • Thermosensitive formulations

  • Dual targeting

  • EPR mechanism

  • Low circulation time without surface modification

  • Difficulties in sterilization

  • Poor reproducibility in terms of size

  • Limited control over drug release

  • A small variety of surface functional groups

  • Poor stability

208,230,237,302

Although numerous investigations have found inorganic nanoparticles to be efficient enough to pass through the BBB, hard degradability and its following toxicities, and undesirable drug delivery, have excluded them from tangible clinical studies. 79

Solid Lipid Nanoparticles (SLNs)

SLN refers to nanodispersions ranging from 10 to 1000 nm of biocompatible lipids including fatty acids (eg, stearic acid), triglycerides (eg, tristearin), waxes (eg, cetyl palmitate), and steroids (eg, cholesterol) that are stabilized with surfactants8082 (Table 1). A combination of surfactants with the hydrophilic–lipophilic balance (HLB) values less than 12, such as Poloxamer 188 and Pluronic®F68, are used in the SLN structure.81,83 The core of SLN is made of solid lipids, which makes them ideal for hydrophobic drug loading. SLNs have attracted growing attention as potential nano-based anticancer drug delivery formulations for gliomas and glioblastoma.84,85 Similar to other nanocarriers, the surface of SLNs is modified and functionalized via the attachment of various targeting ligands including proteins, peptides, small molecules, and antibodies. This results in increased antitumor activities and reduced adverse effects by targeting specific receptor-mediated endocytosis.8588 Along with the aforementioned advantages, prolonged retention time in the serum and brain can be increased by improving the hydrophilicity of SLNs via PEGylation.

Biocompatibility, biodegradability, and surface modifications are the main advantages of SLNs. However, they can be easily eliminated by the RES from the blood due to their lipophilicity, which can present a potential challenge. 89

Polymeric Nanoparticles

Polymeric nanoparticles including nanospheres and nanocapsules are thermodynamically stable structures made of natural or synthetic polymers, with a range of sizes between 1 and 1000 nm. 90 As a result of considering an optimum nanoformulation that is biodegradable for over a few days, the nondegradable formulations are excluded including quantum dots, nonorganic nanocarriers (silica and metal particles), and needle-shaped carbon nanotubes.91,92 Consequently, 3 types of nanostructured materials are considered including polylactic acid (PLA) or its copolymer poly lactide-co-glycolide acid (PLGA), poly butyl cyanoacrylate acid (PBCA) or poly iso hexyl cyanoacrylate acid (PIHCA), and human serum albumin (HSA). 93 It has been reported that 80% of PBCAs are degraded 24 h after IV injection. Therefore, poly alkyl cyanoacrylates with approximately 2000 to 3000 Da molecular weights have the fastest degradability among polymers. Higher poly alkyl cyanoacrylates toxicity rates were observed with the slowest or fastest degradability rates. The intermediate rates showed lower toxicities.94,95 Interestingly, a formulation of PBCA has achieved clinical trial phase III and now is purchased by the trade name of Livatag® (doxorubicin Transdrug). Livatag® is the product of PBCA and Dox HCl attachment, which increases the PBCA’s molecular weight and substantially improves hepatocarcinoma drug delivery.92,96 The enzymatic cleavage by lipases and esterases is thought to be the prevailing degradation mechanism for PLGA and cyanoacrylates, respectively.9799 Furthermore, albumin nanoparticles are degraded within 3 days in macrophages. 100 Natural polymers are prospective candidates for brain drug delivery over synthetics due to the lessened toxicity, improved biodegradability, and lowered costs. Chitosan with a linear structure and randomly distributed N-acetyl-D-glucosamine (acetylated unit) and β-(1→4)-linked D-glucosamine (deacetylated unit) units are the product of chitin extraction from shrimp shells. As a stabilized, biodegradable, and biocompatible formulation with lower toxicity among natural polymers, it can be prepared by simple techniques.101,102 As the hydrophobic structures have increased permeability, chitosan-based nanoformulations have been modified to enhance their hydrophobicity and thus their ability to penetrate the BBB. Wang et al 103 have shown that trimethylated chitosan loading on the surface of PLGA has enhanced brain uptake. Some polymeric surfactants have been used as the BBB’s permeation enhancer such as polysorbate 20, 40, 60, and 80104,105 and also poloxamer 188 in contrast with the polymers such as poloxamine 908, Cremophor®, and Brij®35.106108 Moreover, PEGylated or the so-called stealth nanoparticles are characterized by lower liver uptake and better blood circulation time and tissue distribution.109,110 The elevated brain concentrations of nanopolymers in tumor-bearing animals versus nontumoral animals indicate that diseases such as brain cancer considerably increase the delivery of nanoformulations to target sites (Table 1).92,111 Although PEGylation has improved many aspects of nano-drug delivery, it is not sufficient for an optimum brain delivery system. 92

Several studies have revealed the substantial effect of the adsorbed drug on the nanoparticle’s charge. In some, tumor accumulation reduction has been referred to as ionic interactions especially the therapeutic agents’ positive charge which causes higher reticuloendothelial system accumulation.112114 The polymeric nanoparticles are penetration enhancers with the ability to link with a peptide as a targeting ligand which improves brain drug delivery. 115 Apolipoprotein targeting ligands have been used on the surface of polymer-based nanocarriers to target the low-density lipoprotein receptor and scavenger receptors on the BBB. 116 Additionally, a few studies have investigated targeting the transferrin receptors with anti-transferrin or transferrin antibodies (OX26 or R17217). 117 Likewise, the insulin receptors have been targeted with just 100 μm amounts of insulin receptor antibody which improves nanopolymer’s drug delivery. 118

Optimistically, polymeric nanocarriers can reach the clinic for brain cancer treatment only if they and their catabolites are biodegradable, and less toxic and immunogenic. 89 Unfortunately, as none of the BBB’s receptors are specific or ubiquitously expressed, the adverse effects might be a limiting factor. 92

Dendrimers

Dendrimers are monodisperse, symmetric, and spherical compounds with a chemical core. They are classified based on their molecular weight. Their properties are mostly determined by their surface functional groups.119121 The particle size growth starts from a nucleation site in the center of dendrimers, from where many consecutive branches develop. Consequently, hundreds of reaction sites are available on the surface of the particles. Poly-amidoamine (PAMAM), poly-L-lysine (PLL), and polypropylene imine (PPI) are the most common types of dendrimers that have been applied for drug delivery. They can be loaded by treatment agents and targeted as novel drug delivery systems.122,123 Another advantage of these types of formulations is dual targeting by different agents. 124

In one study, modified dendrimers via Serine–Arginine–Leucine (SRL) peptide were used as gene delivery systems for the brain. It was shown that SRL peptide was bound to endothelial cell membrane receptors on the BBB and lipoprotein receptor-related protein (LRP). Consequently, this enhanced the dendrimers uptake by brain parenchyma tissue. 125 Furthermore, transferrin and wheat-germ agglutinin (WGA) dual targeting on the PEGylated 7 to 10 nm dendrimers can serve as brain tumor therapeutic agents because of well penetration and accumulation. 126

Cationic dendrimers electrostatically bind to the negatively charged genes for gene delivery applications. The reason why gene delivery through the dendrimers seems to be so promising is this mechanism in which they destroy endosome storage in the cytoplasmic environment.127,128 Furthermore, exogenous genes, microRNA (mRNA), and small interfering RNA (siRNA) delivery to tumor sites have been shown in a few studies. 129

In conclusion, dendrimers have specific advantages such as uniform size distribution, high drug loading capacity, multiple targeting ligand conjugations, and high stability.130,131 In contrast, complex synthesis and formulation development, toxicity, and safety issues (especially the amino-functional groups) have restricted the clinical application of dendrimers (Table 1). Furthermore, their positive amino groups can interact with the blood cells which are negatively charged and structurally disrupt and erode cells. This can lead to hematologic toxicities and nano-drug eliminations. Even though the dendrimer cationic groups cause toxicity, their chemical modifications generally minimize these effects. 132

Micelles

Spheroidal nanomicelles are made by the aggregation of amphiphilic block copolymers in an aqueous environment. In micelles, the hydrophobic core and hydrophilic surface have provided low soluble drug loading, modification, and conjugation for targeted brain drug delivery. 133 The hydrophilic outer layer prolongs both blood circulation and stability. Micelles range between 10 and 100 nm, which is ideal for the enhanced permeation and retention (EPR) mechanism at the tumor site. 130 These nano-drug delivery systems are prioritized over other types of novel drug delivery approaches due to simple, stable, and reproducible formulations, as well as simple filtration techniques for sterilization. Their small size and hydrophilic shell help them evade the RES and consequently have a longer circulation time. Therefore, nanomicelles are the potential candidates for the treatment of brain cancer. However, the potential of immunogenicity, premature drug release, poor stability, and lack of appropriate methods for formulation scale-up have limited their application. In addition to the aforementioned disadvantages, the dissociation of micelles at concentrations lower than the critical micelle concentration (CMC) is one of the most significant challenges.134136

Exosomes

Natural nanovesicles with a diameter of 30 to 100 nm have gotten a lot of attention for potential drug delivery applications because of their ability to carry targeted ligands on their surface. They can escape the immune system entrapments due to their production from body cells, resulting in better blood circulation, biodegradability, and biocompatibility.65,137,138 They are also considered intriguing nano-drug delivery systems due to their inherent aptitude for passing across biological membranes and barriers including the BBB without affecting its integrity. Additionally, some investigations have reported glioma-secreted exosomes circulating in the blood, which indicates their capacity to cross the BBB.139143 The abovementioned characteristics have made them promising candidates for delivery for brain cancer treatment. Many studies have utilized exosomes for the delivery of nucleic acids, proteins, and small molecules.144,145 Many studies have been carried out to take advantage of cutting-edge exosome research, including RNA therapeutics. Exosomes containing mRNA inhibited and reduced vasculogenic mimicry, migration, and angiogenesis, leading to glioma tumor suppression.146,147 The administration of brain endothelial cell-derived exosomes that were loaded with siRNA was used to treat brain cancer in another investigation. Despite their limited cell absorption, siRNAs have shown intriguing therapeutic promise. The nanosized exosomes were successful in delivering siRNAs for the treatment of brain diseases.148150 Interestingly, the main mechanism by which exosomes acquired the ability to cross through the BBB was unraveled in vitro. The findings suggested active transcytosis under the impression of inflammation factors (eg, TNF-α).151153 For simultaneous glioma imaging and treatment study, the neuropilin-1-targeted exosomes were co-loaded with SPIONs and curcumin by electroporation method. Natural nanostructures were given in vitro and in vivo, and their therapeutic and diagnostic benefits were greatly improved. Potent synergistic anticancer effects were attributed to the effect of SPION-mediated magnetic flow hyperthermia and Curcumin-mediated treatments. 154 Exosomes have also been employed to decrease brain malignancies in some innovative and exciting studies. Chaperone-rich cell lysates (CRCLs) in particular may play a key role in the development of antitumor vaccinations. Dendritic cells (DCs) are activated by tumor-derived CRCLs, resulting in potential anti-tumor efficacy. DC-derived exosomes were produced in this study using DCs loaded with CRCLs obtained from GL261 glioma cells. They made antitumor T cell immune responses more robust and effective.155159 The notion that brain endothelial cell-derived exosomes can transfer anticancer medicine across the BBB for the treatment of brain cancer in a zebrafish model has been tested in new findings on anticancer drug delivery. The findings show that exosomes supplied to tumors reduced tumor growth markers in a significant manner. As a result, brain endothelial-derived exosomes could be a promising new nano-drug delivery system that could be investigated further in the clinical development of brain cancer therapy.160,161 In a 3D glioblastoma model, exosomes generated from human endometrial stem cells harboring the apoptotic drug atorvastatin, which inhibits cancer growth through a variety of mechanisms, dramatically reduced tumor growth.162164 A number of researchers have looked into the possibility and mechanism of exosomal surface changes. Surface adjustments are implemented to exosomal surface proteins and functional groups. The presence of exosomal membrane proteins such as cytoskeletal components (actin, tubulin, etc), intracellular fusion proteins (annexin and RAB), and heat shock proteins have been decoded by proteomic research. MHC class I and II, CD86 proteins, integrins, as well as other proteins were also listed.165167 In conclusion, covalent and noncovalent alterations, as well as genetic engineering, are being investigated as techniques to improve exosome efficacy and reduce their drawbacks in the targeted drug delivery.154,165,168170

Considering all the facts, exosomes have natural biocompatibility, greater chemical stability, and the ability to control intercellular interactions when compared to synthesized nanoparticles. They are also regarded as nonimmunogenic, nontoxic, and nonspecific nanocarriers.171173 However, there are challenges to exosome use, such as a lack of standardized exosome separation and purification criteria, an uncertain mechanism for exosome uses in cancer treatment, heterogeneity, and difficulty maintaining exosomes. They must be analyzed and adjusted to get a more desirable and controllable release and scalation in clinical scenarios.174177

Minicells

Minicells are anucleated, nano-sized (100-300 nm), neither alive nor dividing cells due to mutation in genes involved in the normal bacterial cell cycle. Ribosomes, peptidoglycans, plasmids, RNA, and bacterial proteins are all kept intact. As a result, they are metabolically active and capable of carrying out cell processes such as mRNA translation, transcription, and translational activities, as well as ATP synthesis.178180 Therefore, various minicell-producing bacterial strains (such as E coli or others) have been brought into attention or are being researched for possible adoption.181,182 Chemotherapeutics, si/shRNA, drugs, and chemotherapeutics can all be administered to malignant tissue with pinpoint accuracy. Additionally, multiple targeting ligands, such as bi-specific antibodies, can modify their surface to improve their clinical applications.183187 The biocompatibility and biodegradability of these nanocarriers are equivalent to that of other nanocarriers. Furthermore, they appear to be one of the most unique and appealing nano-drug delivery approaches due to increased encapsulation efficiency, overcoming drug leakage, enhancing targeting specificity, and improving treatment agents’ therapeutic index.184,188,189 Many studies are being conducted in order to develop optimal minicell-based drug delivery systems for therapeutic purposes. Minicells that administered the miR-34a greatly increased the temozolomide effects in vivo in a study as adjuvant therapy. MiR-34a regulates signaling pathways implicated in intratumoral heterogeneity and, as a result, temozolomide resistance.190192 In another experiment, late-stage brain cancer dogs were treated with EGFR-targeted minicells carrying doxorubicin and coated with BsAbs. The nanoparticles were found in the brain tumor’s center and had a high median survival rate. There have been no reports of particular toxicity. A Phase 1 clinical trial employing EGFR-targeted, doxorubicin-loaded minicells for the management of patients with relapsed glioblastoma was designed on this premise.193197

Despite the benefits and advantages of minicell structures, more research is needed to interpret them in the clinic, notably on their stability and release profile, as well as their release mechanism in the tumor microenvironment and intratumoral. According to the experiments that have been reported so far, they had no significant detrimental impacts. Before they are extensively employed, however, their immunogenicity due to bacterial sources must be thoroughly investigated. Additionally, the risk of organ damage from long-term organ accumulations adds to the concerns regarding these novel nanoformulations.198200

Liposomes

Liposomes are attractive vesicles for the delivery of various drugs and compounds such as antibiotics, therapeutic proteins, antineoplastics, and peptides.201204 These vesicles, consisting of phospholipids, are spontaneously formed and comprise of single or multiple layers. Liposomes are categorized based on the following classes: (Ι) multi-lamellar vesicles (MLVs), (ΙΙ) large unilamellar vesicles (LUVs), and (ΙΙΙ) small unilamellar vesicles (SUVs). The sizes of MLVs, LUVs, and SUVs are approximately 500, 100 to 500, and 25 to 100 nm, respectively. MLVs are commonly used for extended drug release objectives, while SUVs are optimal for drug delivery through the endothelial cells lining the blood vessels and tissue epithelium. LUVs are medium-sized structures for vaccine and gene delivery purposes.205207

Due to the recent improvements in encapsulation efficiency, drug loading, stabilized formulation preparations, decoration for the molecules targeting delivery, and co-delivery, liposomes are promising drug delivery systems.201,208

However, some limitations affect the liposomal formula application and restrict its utilization for intended purposes. These particulate systems have high clearance and low blood circulation and are cleared by the RES. The blood circulation time can be improved by decreasing the particle size to less than 100 nm, and liposomal surface PEGylation.209,210 Additionally, active targeting is accomplished by ligand attachment for a specific receptor (like monoclonal antibody) on the liposomes, preferentially to the end of the PEG moieties as the targeted liposomes have been demonstrated to be much more effective if they are sterically stabilized.211,212

Many attempts have been made to reach an optimal liposomal formulation.213215 Improving liposomal transportation to the tumor site has been the main objective of several previous studies. Most of these studies examined either cationic or PEGylated liposomes.216,217 Based on recent studies, the main mechanism for liposomal entrance into the brain remains unclear, however, it has been suggested that tight junction disruption may be the most probable mechanism. 218 Some pH-sensitive cell-penetrating peptides such as TR peptides are surface conjugated to the drug-loaded liposomes and were developed and utilized for glioma treatment. 214 This type of nanoformulation was shown to enhance the drug efficacy toward gliomas, both in vitro and in vivo. Moreover, adriamycin (ADM)-encapsulated thermosensitive liposomes enhanced Dox delivery to the brain and prolonged the survival of glioma-bearing mice. 219

Receptor-mediated endocytosis is a general mechanism of cells to import particles. The endocytosis mechanism has been targeted by nanosystems especially liposomes. For instance, the transferrin receptor is over-expressed on the brain capillary endothelial cells.220222 Therefore, the efficacy of dual-targeted doxorubicin (Dox) liposomes conjugated concomitantly to folate and transferrin was investigated in a study. This dual-targeting Dox liposome enhanced the therapeutic efficacy of Dox toward gliomas and reduced the off-target side effects as compared to Dox solution. 223 In another study, the TF-specific targeting ligand and TAT, a nonspecific cell-penetrating peptide (a small positive charged variant derived from trans-activating transcription activator peptide of HIV) were attached to the paclitaxel and Dox containing liposomes (TF/TAT-LP). They exhibited an effective antitumor activity against gliomas and enhanced the median survival time of glioma-bearing mice. 213

Moreover, targeting is an approach that enhances the liposomal formulations’ therapeutic and antitumor effects. It is ascribed to their median particle size and an extent to the homogeny particle size distribution. The EPR effect is the main mechanism for liposomal accumulation and accordingly, 100 nm liposomes can merely reach specific regions.224228 In one study, the noninvasive focused ultrasound method accompanied bubble liposome (with 55-299 nm diameter range) injection into the circulatory system. The results showed that smaller liposomes serve as more effective drug delivery systems than larger ones.229,230 In another study, cationic liposomes were used to transfect tumor cells with an interferon gene-expressing vector (plasmid), which resulted in tumor regression.231,232 Likewise, antisense epidermal growth factor was entrapped into cationic liposomes and tested in human malignant glioma cell lines. 233 The possible cationic liposomal cell uptake mechanism can be explained by negatively charged phospholipid head group interactions with the positively charged liposomes (absorption) and therefore is called adsorptive mediated transcytosis.234,235 In this regard, cationic bovine serum albumin as a positive ligand has shown increased cell absorption and enhanced drug delivery through the aforementioned mechanism.224,236

The difficulties in surface conjugation and small number of functional groups can be the main liposomal drawbacks (Table 1). Moreover, other disadvantages exist, such as poor and low reproducibility of nanoparticle size, stability, and insoluble agent loading. Optimum sterilization and uncontrolled drug release are other challenges associated with the liposomes.89,130,237,238

Nanotechnology Clinical Approaches for Brain Cancer Drug Delivery

ONZEALD™

Sponsored by Lawrence Recht, Nektar Therapeutics has designed a polymeric version of irinotecan as the first long-acting topoisomerase I inhibitor. Irinotecan molecules are attached via an ester bond to the PEG polymer. Carboxylesterase and other enzymes react with the ester bond and cause the release of irinotecan and consequently, producing 7-ethyl-10-hydroxy-camptothecin or SN38 as the active metabolite. SN38 attacks DNA and causes its damage through topoisomerase inhibition. The main objective of the etirinotecan pegol (NKTR-102 or Onzeald) design is to eliminate or attenuate the irinotecan side effects and also improve its efficacy by drug distribution modifications. In preclinical studies, it demonstrated a 300-fold tumor concentration in comparison with a first-generation topo I-inhibitor. The NKTR is larger than normal vessel pores, helping it reach the tumor microenvironment by enhanced permeation and retention. Onzeald efficacy has been assessed in ovarian, breast, brain, colorectal, and lung cancer. The pharmacokinetic characteristics of the ONZEALD metabolite (SN38) differ significantly from others by providing maximal tumor exposure of the active drug (Table 2). For example, the Cmax has been reduced 5 to 10 times and also the half-life has been increased to almost 50 days. The dose of 145 mg/m2 ONZEALD in phase II clinical trial almost equals the dose of 350 mg/m2 irinotecan. The protracted exposure between continuous dosing cycles and lower Cmax has improved the therapeutic effects of the treatments and clinical outcomes. Its open-label, single-arm 2016 phase II clinical trial had completed the ONZEALD’s efficacy in bevacizumab-resistant high-grade gliomas.239241

Table 2.

Nanocarrier-based Clinical Trials.

Name Nanocarrier Properties Clinical phase References
Ozeald (NKTR-102) PEGylated polymeric irinotecan, etirinotecan pegol
  • Limited side effects of irinotecan

  • Improved efficacy, a 300-fold increase in tumor concentration

  • EPR mechanism

  • Evaluated in breast, ovarian, colorectal, brain, and lung cancer

  • Constant exposure of the tumor to the active drug due to reduced Cmax and increased half-life

Phase 2
for Anaplastic Astrocytomas Anaplastic Oligodendrogliomas
Glioblastomas (GBM) is completed
239241
NU-0129 Spherical nucleic acid (SNA) gold nanoparticle formulation composed of small interfering RNAs (siRNAs) targeting BCL2L12 gene
  • Inhibiting the expression of BCL2L12 by NU-0129 induces tumor cell apoptosis

Early Phase 1
for gliosarcoma and
Recurrent glioblastoma is active
243,251
186RNL 186Rhenium Nanoliposomes
  • 100 nm

  • The half-life of 90 h

  • Limited penetration which limits toxicity of other forms of radiation

  • The liposomal formulation helps to retain within the tumor

  • EPR mechanism

Phase 1
Phase 2
for glioma which is recruiting
252255
2B3-101 Glutathione PEGylated liposomal Dox
  • ∼110 nm

  • Targeted drug delivery of Dox

  • Optimal distribution to the brain

  • Targeting glutathione transporters on the surface of BBB

Phase 1
Phase 2
for brain metastases
Lung cancer
Breast cancer
Melanoma
Malignant glioma is completed and
Phase 2
for meningeal carcinomatosis is unknown
262,263
Doxil® PEGylated Liposomal Dox
  • ∼100-110 nm

  • Improved tissue and CSF concentrations

Phase 1
Phase 2
for glioblastoma is completed
264
EGFR (V)-EDV-Dox BsAb-targeted, payload-packaged EDV nanocells
  • 400 nm

  • EPR mechanism

  • BsAb binds to the tumor cell-surface receptor which causes the release of Dox within the cancer cell

  • EDVs are derived from bacteria and cause immunostimulating

  • Bypass the immunosuppression caused by the tumor

Phase 1
for glioblastoma
astrocytoma, Grade IV which is recruiting
266,267

NU-0129

NU-0129 is a class of gene regulation spherical nucleic acid (SNA) nanostructures having well-orientated siRNA oligonucleotides finely designed and synthesized at their surface. Such SNAs are made up of siRNA oligonucleotides constructed on gold (Au) nanoparticle centers with oligo ethylene glycol (OEG) or PEG (OEG/PEG) on their surface to improve stability and circulatory half-life.242,243 Based on the earlier investigations, using Au as the SNA heart allows for accurate spatial analysis of Au distribution in cells and tumors using inductively coupled plasma mass spectrometry (ICP-MS), X-ray fluorescence microscopy (XFM), and silver histopathology staining.244247 For the treatment of glioblastoma, the siRNA gold nanoparticles target the Bcl-2-like protein 12 (BCL2L12) domains. The tumor cells die as a result of the inhibition of BCL2L12 expression. In glioblastoma, BCL2L12 is hypothesized to be upregulated in a tumor-promoting direction. It inhibits the activation of effector caspase-3 and caspase-7. It can also bind wild-type p53 and its mutants, destabilizing them and preventing p53 from attaching to target gene promoters.246,248250 In order to test safety, pharmacokinetics, and intratumoral SNA nanoconjugate accumulation, the very first phase 0 clinical trial involving the systemic microdose delivery of NU-0129 in adults with recurrent glioblastoma was done. As a consequence, it was discovered to be safe and well tolerated by endothelial, immunological, and tumor cells, and it was linked to lower target protein levels in patients. 243 251

Liposomal Rhenium-186 (186Re)

Radiation is an essential part of brain cancer treatment, but due to the toxicity of high doses, its usage is limited. Rhenium-186 (186Re) is a diagnostic imaging rhenium isotope that is chemically similar to technetium-99m (99mTc) and is a reactor-produced isotope with great potential for medical therapy only after successful delivery (Table 2). It has a 90-hour half-life with a 2 mm tissue path length. 252 Its low tissue penetration has provided higher administration doses with the least toxicity. Localized radiation at the tumor site is achievable through the 100 nm liposomal formulations of the 186Re. Its applicability in the failed glioblastoma treatment procedures and accumulation at the tumor site is because of the EPR effect.253255

2B3-101

The phase I/IIa clinical study for 2B3-101 or glutathione (GSH) PEGylated liposomal Dox in patients with glioma and breast cancer brain metastases has concluded. The G-technology employed to create this ideal nanostructure is established on the GSH identified transporter on BBB endothelial (Km of 6 mM). It is generally considered safe and utilizes micromolar glutathione targeting molecules on PEGylated nanoliposomal dosage forms.256259 The brain-to-blood ratio of doxorubicin was 4.8 times greater upon injection of 2B3-101 than generic PEGylated nanoliposomal Dox in discoveries. As a result, the brain’s doxorubicin concentration rises without compromising the BBB’s integrity.260,261 It greatly slowed tumor growth when compared to nontargeted PEGylated liposomal Dox.262,263

PEGylated Liposomal Dox (Doxil®)

Dox refers to a wide range of effective chemotherapeutics for the most aggressive malignancies such as glioblastoma. However, its effectiveness in vivo is under question due to the poor penetration as the result of the BBB. Its CSF and brain tissue concentrations have dramatically increased in tumor models after being sterically stabilized. A PEGylated liposomal Dox formulation with or without another chemotherapeutic agent like temozolomide not only has enhanced drug delivery to the brain but also case series and two-phase II studies concerning recurrent glioblastoma have demonstrated modest promising results. 264

EGFR (V)-EDV-Dox

The EnGeneIC EDVTM technology-based EGFR (V)-EDV-Dox is a 400 nm Dox-loaded bacterial minicell that utilizes bispecific antibodies to function as a targeted therapy in cancer treatment (Table 2). In pigs and dogs, they were well tolerated with modest and temporary toxicity and inflammation, according to earlier investigations.193,265 The minicells go from the bloodstream to the tumor microenvironment, where they assault the tumor cells’ surface and release Dox. Furthermore, remnant EDV bodies in the tumor microenvironment that were unable to infiltrate malignant cells signal the immune system to the tumor site, counteracting the tumor’s immunosuppression. Overall, these microcells polarize M1 macrophages and engage NK cells at the same time, resulting in a Th1 cytokine response with powerful anticancer activity. Upon that, dendritic cell maturation and antigen presentation proceeds, leading to tumor-specific CD8+ T cells and durable tumor remission.193,266269

Discussion

Brain drug delivery systems have significantly advanced over the past few years with current research progressing the field every further. The latest advanced biological and physicochemical properties of the nanocarriers have taken them to higher levels. Furthermore, they have enhanced blood circulation and bioavailability. Not only do they provide a productive functionalized surface for a variety of molecules, but also they can be modified for controlled release over time. However, translating brain tumor treatment into clinical trials encounter unique barriers, largely in part due to the CNS biological barriers such as the BBB. Reduced tumor accumulation seems to be another obstacle for such delivery systems to reach the clinic. Optimizing the physicochemical parameters may overcome the disadvantages of novel nanoformulation. Shape, size, functionality, and surface charge have been modified in a variety of studies in the field of brain drug delivery research.270273

Designing a triggered release, as well as stimuli-responsive formulations, are among the most novel fields of drug delivery studies.130,274,275 Nonetheless, only a handful of studies have attempted for cancer treatment examining stimuli-responsive systems. Temperature, pH, magnetic field, enzymes, oxidative stress, etc are triggering signals that have been frequently used for cancer treatment. The advantages of employing intrinsic environmental features in the tumor site in comparison with normal tissues for improved and efficient stimuli-responsive systems have been comprehensively discussed in the literature. 130 Furthermore, external stimuli such as light, heat, and magnetic fields are other options for controlled release. Liu et al 276 have obtained physicochemical sensitive (pH, temperature, etc) nanopolymers with both anticancer effects and brain tumor magnetic resonance imaging. Their application for both diagnosis and treatment procedures has been positively approved. The concept of these multifunctional nanosystems demonstrates the future of nanocarriers with vast room for growth and advancements in the field.277,278 Nonetheless, the aforementioned nanoparticles’ potential systemic toxicity and neurotoxicity in the clinic should be considered.

Despite numerous efforts to successfully improve the use of nanocarriers in different areas of clinical research, there remain challenges that need to be addressed. Investigating immortalized brain endothelial cell models for the BBB penetration assessment and testing the nanocarriers’ efficiency to get through the BBB with the minimum cellular damaging effects are considered as examples of challenging aspects of this area. The high cost of these cell lines, such as hCMEC/D3, bEND3, and RBE4, limited availability and accessibility, and susceptibility to media and cultural contaminations have widely affected in vitro studies.279,280 However, they have been used more frequently in comparison with astrocyte- or PC-derived cell lines in BBB studies.279,281 Preclinical in vivo studies in the field of brain cancer is often hindered by the difficulty of modeling the biological heterogeneity that is observed between humans and mouse models, which are additional challenges for brain cancer.282284 Through advancements in pathological imaging methods and factoring in higher animal-to-human translational success rates, the in vivo complications may be reduced. The design of clinical trials is faced with difficulty in group classifications given the heterogeneity of tumor and the affected cell types. Furthermore, additional prognostic factors such as the low number of participants in each group may lead to a lack of statistical power to detect significant differences between control and therapeutic arms.285,286

Although more research and development are needed for effective nanocarriers with optimal clinical kinetics, they show promise as a suitable method of delivery for brain cancer treatment based on recent clinical studies.

Concluding Remarks, and Future Perspectives

Nanomedicine provides innovative opportunities in the development of tissue-specific targeted therapeutics, and imaging agents for brain tumor management. We have reviewed the challenges and advantages of several development efforts using nanocarriers for the treatment of brain tumors. Here we highlight the importance of further investigations for the development of effective treatments using nanotechnology for monitoring and treatment. This will facilitate the extremely effective development of nanomedicine in brain cancer disease. Taken together, the main goals of the upcoming brain cancer researches should be not only about having a higher survival rate, but also the patient’s quality of life and especially the burden of treatment morbidities. Therefore, other challenges in brain cancer development may take the nanomedicine drug development under, which a few will be discussed.

First, we found an unmet need in coordinating a multifaceted team of specialists like researchers, neurologists, surgeons, neuropathologists, and other health professionals that will cause less suffering for those who burden the disease. Moreover, clarifying, organizing, and improving the fund and support in cancer investigations and research is required to make small communities of brain cancer research and investigations more developed.

Second, considering prioritized molecular and genetic tumor detection can lead to precise diagnosis and patient stratification and consequently move us toward specific and efficient drug development that more than likely will promote and facilitate current challenging brain cancer medications. This is in line with broadening tailored individualized therapy areas.

Finally, a clinical trial center in this field mostly cannot by itself recruit a considerable statistically powerful number of patients to run the study. Exploiting and extending more clinical trial centers can be a solution plus donating more grants for the brain cancer research portfolio.287292

Thus, even though nanomedicine is a crucial milestone in brain cancer treatment but it requires a better understanding of other essential key elements for further reliable advancements.

Footnotes

Declaration of Conflicting Interests: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: This paper is part of the PhD thesis of Amin Mehrabian (grant number: 960833) supported by Nanotechnology Research Center, Mashhad University of Medical Sciences (MUMS), Mashhad, Iran.

ORCID iD: Amin Mehrabian https://orcid.org/0000-0002-8879-7565

References

  • 1.Kerzarea D, Khedekar P. Indole derivatives acting on central nervous system—review. J Pharm Sci Bioscientific. 2016;6(1):144-156. [Google Scholar]
  • 2.Cacabelos R, Torrellas C, Fernández-Novoa L, López-Muñoz F. Histamine and immune biomarkers in CNS disorders. Mediators Inflamm. 2016;2016:1-10. 10.1155/2016/1924603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hall JE. Guyton and Hall Textbook of Medical Physiology E-Book. Elsevier Health Sciences; 2015. [Google Scholar]
  • 4.Alyautdin R, Khalin I, Nafeeza MI, Haron MH, Kuznetsov D. Nanoscale drug delivery systems and the blood–brain barrier. Int J Nanomed. 2014;9(795):795–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Christ J, Parvathi R. Brain tumors: an engineering perspective. Int J Comput Sci Issues (IJCSI). 2012;9(4):392‐396. [Google Scholar]
  • 6.Fidler IJ. The biology of brain metastasis: challenges for therapy. Cancer J. 2015;21(4):284‐293. [DOI] [PubMed] [Google Scholar]
  • 7.Zhao M, van Straten D, Broekman MLD, Préat V, Schiffelers RM. Nanocarrier-based drug combination therapy for glioblastoma. Theranostics. 2020;10(3):1355‐1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Boire A, Brastianos PK, Garzia L, Valiente M. Brain metastasis. Nat Rev Cancer. 2020;20(1):4‐11. [DOI] [PubMed] [Google Scholar]
  • 9.Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet. 2018;392(10145):432‐446. [DOI] [PubMed] [Google Scholar]
  • 10.Vargo MM. Brain tumors and metastases. Phys Med Rehabil Clin N Am. 2017;28(1):115‐141. [DOI] [PubMed] [Google Scholar]
  • 11.Chen R, Smith-Cohn M, Cohen AL, Colman H. Glioma subclassifications and their clinical significance. Neurotherapeutics. 2017;14(2):284‐297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rastogi K, Bhaskar S, Gupta S, Jain S, Singh D, Kumar P. Palliation of brain metastases: analysis of prognostic factors affecting overall survival. Indian J Palliat Care. 2018;24(3):308‐312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Batash R, Asna N, Schaffer P, Francis N, Schaffer M. Glioblastoma multiforme, diagnosis and treatment; recent literature review. Curr Med Chem. 2017;24(27):3002‐3009. [DOI] [PubMed] [Google Scholar]
  • 14.Soomro SH, Ting LR, Qing YY, Ren M. Molecular biology of glioblastoma: classification and mutational locations. J Pak Med Assoc. 2017;67(9):1410‐1414. [PubMed] [Google Scholar]
  • 15.Gallego O. Nonsurgical treatment of recurrent glioblastoma. Curr Oncol. 2015;22(4):e273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA. 2013;310(17):1842‐1850. [DOI] [PubMed] [Google Scholar]
  • 17.Alifieris C, Trafalis DT. Glioblastoma multiforme: pathogenesis and treatment. Pharmacol Ther. 2015;152:63‐82. [DOI] [PubMed] [Google Scholar]
  • 18.Zorzan M, Giordan E, Redaelli M, Caretta A, Mucignat-Caretta C. Molecular targets in glioblastoma. Future Oncol. 2015;11(9):1407‐1420. [DOI] [PubMed] [Google Scholar]
  • 19.Tang W, Fan W, Lau J, Deng L, Shen Z, Chen X. Emerging blood-brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem Soc Rev. 2019;48(11):2967‐3014. [DOI] [PubMed] [Google Scholar]
  • 20.Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20(1):26‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Liebner S, Dijkhuizen RM, Reiss Y, Plate KH, Agalliu D, Constantin G. Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol. 2018;135(3):311‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Patra JK, Das G, Fraceto LF, Campos EVR, del Pilar Rodriguez-Torres M, Acosta-Torres LS, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnol. 2018;16(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chaturvedi VK, Singh A, Singh VK, Singh MP. Cancer nanotechnology: a new revolution for cancer diagnosis and therapy. Curr Drug Metab. 2019;20(6):416‐429. [DOI] [PubMed] [Google Scholar]
  • 24.Zhang Z, Guan J, Jiang Z, Yang Y, Liu J, Hua W, et al. Brain-targeted drug delivery by manipulating protein corona functions. Nat Commun. 2019;10(1):1‐11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fan K, Jia X, Zhou M, Wang K, Conde J, He J, et al. Ferritin nanocarrier traverses the blood brain barrier and kills glioma. ACS Nano. 2018;12(5):4105‐4115. [DOI] [PubMed] [Google Scholar]
  • 26.Ruan S, Xie R, Qin L, Yu M, Xiao W, Hu C, et al. Aggregable nanoparticles-enabled chemotherapy and autophagy inhibition combined with anti-PD-L1 antibody for improved glioma treatment. Nano Lett. 2019;19(11):8318‐8332. [DOI] [PubMed] [Google Scholar]
  • 27.Chai Z, Ran D, Lu L, Zhan C, Ruan H, Hu X, et al. Ligand-modified cell membrane enables the targeted delivery of drug nanocrystals to glioma. ACS Nano. 2019;13(5):5591‐5601. [DOI] [PubMed] [Google Scholar]
  • 28.Ehrlich P. Eine farbenanalytische Studie. Berlin: Hirschwald. 1885. [Google Scholar]
  • 29.Goldmann EE. Vitalfarbung am zentralnervensystem. Abhandl Konigl preuss Akad Wiss. 1913;1:1‐60. [Google Scholar]
  • 30.Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57(2):178‐201. [DOI] [PubMed] [Google Scholar]
  • 31.Majesky MW. Developmental basis of vascular smooth muscle diversity. Arterioscler Thromb Vasc Biol. 2007;27(6):1248‐1258. [DOI] [PubMed] [Google Scholar]
  • 32.Janzer RC, Raff MC. Astrocytes induce blood–brain barrier properties in endothelial cells. Nature. 1987;325(6101):253‐257. [DOI] [PubMed] [Google Scholar]
  • 33.Gaillard PJ, van der Sandt ICJ, Voorwinden LH, Vu D, Nielsen JL, de Boer AG, et al. Astrocytes increase the functional expression of P-glycoprotein in an in vitro model of the blood-brain barrier. Pharm Res. 2000;17(10):1198‐1205. [DOI] [PubMed] [Google Scholar]
  • 34.Daneman R, Prat A. The blood–brain barrier. Cold Spring Harb Perspect Biol. 2015;7(1):a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Begley DJ. Delivery of therapeutic agents to the central nervous system: the problems and the possibilities. Pharmacol Ther. 2004;104(1):29‐45. [DOI] [PubMed] [Google Scholar]
  • 36.Coomber B, Stewart P. Morphometric analysis of CNS microvascular endothelium. Microvasc Res. 1985;30(1):99‐115. [DOI] [PubMed] [Google Scholar]
  • 37.Reese T, Karnovsky MJ. Fine structural localization of a blood-brain barrier to exogenous peroxidase. J Cell Biol. 1967;34(1):207‐217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Brightman M, Reese T. Junctions between intimately apposed cell membranes in the vertebrate brain. J Cell Biol. 1969;40(3):648‐677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Brightman MW, Hori M, Rapoport SI, Reese TS, Westergaard E. Osmotic opening of tight junctions in cerebral endothelium. J Comp Neurol. 1973;152(4):317‐325. [DOI] [PubMed] [Google Scholar]
  • 40.Villaseñor R, Lampe J, Schwaninger M, Collin L. Intracellular transport and regulation of transcytosis across the blood-brain barrier. Cell Mol Life Sci. 2019;76(6):1081‐1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Riabinska A, Zille M, Terzi MY, Cordell R, Nieminen-Kelhä M, Klohs J, et al. Pigment epithelium-derived factor improves paracellular blood-brain barrier integrity in the normal and ischemic mouse brain. Cell Mol Neurobiol. 2020;40(5):751‐764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gawdi R, Emmady PD. Physiology, Blood Brain Barrier. StatPearls Publishing Copyright © 2021, StatPearls Publishing LLC.; 2021. [Google Scholar]
  • 43.Armulik A, Genové G, Betsholtz C. Pericytes: developmental, physiological, and pathological perspectives, problems, and promises. Dev Cell. 2011;21(2):193‐215. [DOI] [PubMed] [Google Scholar]
  • 44.Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature. 2006;443(7112):700‐704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shepro D, Morel N. Pericyte physiology. FASEB J. 1993;7(11):1031‐1038. [DOI] [PubMed] [Google Scholar]
  • 46.Liebner S, Czupalla CJ, Wolburg H. Current concepts of blood-brain barrier development. Int J Dev Biol. 2011;55(4-5):467‐476. [DOI] [PubMed] [Google Scholar]
  • 47.Abdullahi W, Tripathi D, Ronaldson PT. Blood-brain barrier dysfunction in ischemic stroke: targeting tight junctions and transporters for vascular protection. Am J Physiol Cell Physiol. 2018;315(3):C343‐C356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Langen UH, Ayloo S, Gu C. Development and cell biology of the blood-brain barrier. Annu Rev Cell Dev Biol. 2019;35:591‐613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Felmlee MA, Jones RS, Rodriguez-Cruz V, Follman KE, Morris ME. Monocarboxylate transporters (SLC16): function, regulation, and role in health and disease. Pharmacol Rev. 2020;72(2):466‐485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bradbury M, Begley D, Kreuter J. The blood-brain barrier and drug delivery to the CNS: Informa Health Care; 2000.
  • 51.Ghose AK, Viswanadhan VN, Wendoloski JJ. A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. J Comb Chem. 1999;1(1):55‐68. [DOI] [PubMed] [Google Scholar]
  • 52.Jones AR, Shusta EV. Blood-brain barrier transport of therapeutics via receptor-mediation. Pharm Res. 2007;24(9):1759‐1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Liu X, Chen C. Strategies to optimize brain penetration in drug discovery. Curr Opin Drug Discov Dev. 2005;8(4):505‐512. [PubMed] [Google Scholar]
  • 54.Li X, Tsibouklis J, Weng T, Zhang B, Yin G, Feng G, et al. Nano carriers for drug transport across the blood brain barrier. J Drug Targeting. 2016:1‐39. [DOI] [PubMed] [Google Scholar]
  • 55.Alli S, Figueiredo CA, Golbourn B, Sabha N, Wu MY, Bondoc A, et al. Brainstem blood brain barrier disruption using focused ultrasound: a demonstration of feasibility and enhanced doxorubicin delivery. J Control Release. 2018;281:29‐41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Idbaih A, Canney M, Belin L, Desseaux C, Vignot A, Bouchoux G, et al. Safety and feasibility of repeated and transient blood-brain barrier disruption by pulsed ultrasound in patients with recurrent glioblastoma. Clin Cancer Res. 2019;25(13):3793‐3801. [DOI] [PubMed] [Google Scholar]
  • 57.Karmur BS, Philteos J, Abbasian A, Zacharia BE, Lipsman N, Levin V, et al. Blood-brain barrier disruption in neuro-oncology: strategies, failures, and challenges to overcome. Front Oncol. 2020;10:563‐840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Masserini M. Nanoparticles for brain drug delivery. ISRN Biochem. 2013;2013:238‐428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Carthy DJM, Malhotra M, O'Mahony AM, Cryan JF, O'Driscoll CM. Nanoparticles and the blood-brain barrier: advancing from in-vitro models towards therapeutic significance. Pharm Res. 2015;32(4):1161‐1185. [DOI] [PubMed] [Google Scholar]
  • 60.Ayloo S, Gu C. Transcytosis at the blood-brain barrier. Curr Opin Neurobiol. 2019;57:32‐38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Matias DMLD, Battaglia G. The role of BAR proteins and the glycocalyx in brain endothelium transcytosis. Cells. 2020;9(12):1-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Zhang W, Liu QY, Haqqani AS, Leclerc S, Liu Z, Fauteux F, et al. Differential expression of receptors mediating receptor-mediated transcytosis (RMT) in brain microvessels, brain parenchyma and peripheral tissues of the mouse and the human. Fluids Barriers CNS. 2020;17(1):47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chen Y, Liu L. Modern methods for delivery of drugs across the blood-brain barrier. Adv Drug Deliv Rev. 2012;64(7):640‐665. [DOI] [PubMed] [Google Scholar]
  • 64.Pandit R, Chen L, Götz J. The blood-brain barrier: physiology and strategies for drug delivery. Adv Drug Deliv Rev. 2020;165-166:1-14. [DOI] [PubMed] [Google Scholar]
  • 65.Azarmi M, Maleki H, Nikkam N, Malekinejad H. Transcellular brain drug delivery: a review on recent advancements. Int J Pharm. 2020;586:119‐582. [DOI] [PubMed] [Google Scholar]
  • 66.Razzak RA, Florence GJ, Gunn-Moore FJ. Approaches to CNS drug delivery with a focus on transporter-mediated transcytosis. Int J Mol Sci. 2019;20(12):1-43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Kaya M, Ahishali B. Basic physiology of the blood-brain barrier in health and disease: a brief overview. Tissue Barr. 2020;1840913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bai X, Moraes TF, Reithmeier RAF. Structural biology of solute carrier (SLC) membrane transport proteins. Mol Membr Biol. 2017;34(1-2):1‐32. [DOI] [PubMed] [Google Scholar]
  • 69.Tuma P, Hubbard AL. Transcytosis: crossing cellular barriers. Physiol Rev. 2003;83(3):871‐932. [DOI] [PubMed] [Google Scholar]
  • 70.D'Souza A, Dave KM, Stetler RA DSM. Targeting the blood-brain barrier for the delivery of stroke therapies. Adv Drug Deliv Rev. 2021;171:332‐351. [DOI] [PubMed] [Google Scholar]
  • 71.Sun P, Xiao Y, Di Q, Ma W, Ma X, Wang Q, et al. Transferrin receptor-targeted PEG-PLA polymeric micelles for chemotherapy against glioblastoma multiforme. Int J Nanomed. 2020;15:6673‐6688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yang H. Nanoparticle-mediated brain-specific drug delivery, imaging, and diagnosis. Pharm Res. 2010;27(9):1759‐1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ku S, Yan F, Wang Y, Sun Y, Yang N, Ye L. The blood–brain barrier penetration and distribution of PEGylated fluorescein-doped magnetic silica nanoparticles in rat brain. Biochem Biophys Res Commun. 2010;394(4):871‐876. [DOI] [PubMed] [Google Scholar]
  • 74.Dos Reis SRR, Pinto SR, de Menezes FD, Martinez-Manez R, Ricci-Junior E, Alencar LMR, et al. Senescence and the impact on biodistribution of different nanosystems: the discrepancy on tissue deposition of graphene quantum dots, polycaprolactone nanoparticle and magnetic mesoporous silica nanoparticles in young and elder animals. Pharm Res. 2020;37(3):1-12. [DOI] [PubMed] [Google Scholar]
  • 75.Hettiarachchi SD, Graham RM, Mintz KJ, Zhou Y, Vanni S, Peng Z, et al. Triple conjugated carbon dots as a nano-drug delivery model for glioblastoma brain tumors. Nanoscale. 2019;11(13):6192‐6205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ren J, Shen S, Wang D, Xi Z, Guo L, Pang Z, 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):3324‐3333. [DOI] [PubMed] [Google Scholar]
  • 77.Qiao R, Jia Q, Hüwel S, Xia R, Liu T, Gao F, et al. Receptor-mediated delivery of magnetic nanoparticles across the blood–brain barrier. Acs Nano. 2012;6(4):3304‐3310. [DOI] [PubMed] [Google Scholar]
  • 78.Thomsen LB, Linemann T, Pondman KM, Lichota J, Kim KS, Pieters RJ, et al. Uptake and transport of superparamagnetic iron oxide nanoparticles through human brain capillary endothelial cells. ACS Chem Neurosci. 2013;4(10):1352‐1360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yang Z, Zhang Y, Yang Y, Sun L, Han D, Li H, et al. Pharmacological and toxicological target organelles and safe use of single-walled carbon nanotubes as drug carriers in treating Alzheimer disease. Nanomed: Nanotechnol Biol Med. 2010;6(3):427‐441. [DOI] [PubMed] [Google Scholar]
  • 80.Khameneh B, Halimi V, Jaafari MR, Golmohammadzadeh S. Safranal-loaded solid lipid nanoparticles: evaluation of sunscreen and moisturizing potential for topical applications. Iran J Basic Med Sci. 2015;18(1):58‐63. [PMC free article] [PubMed] [Google Scholar]
  • 81.Mehnert W, Mäder K. Solid lipid nanoparticles: production, characterization and applications. Adv Drug Deliv Rev. 2001;47(2):165‐196. [DOI] [PubMed] [Google Scholar]
  • 82.Blasi P, Giovagnoli S, Schoubben A, Ricci M, Rossi C. Solid lipid nanoparticles for targeted brain drug delivery. Adv Drug Deliv Rev. 2007;59(6):454‐477. [DOI] [PubMed] [Google Scholar]
  • 83.Small DM. Physical chemistry of lipids. Plenum Press; 1986. [Google Scholar]
  • 84.Gastaldi L, Battaglia L, Peira E, Chirio D, Muntoni E, Solazzi I, et al. Solid lipid nanoparticles as vehicles of drugs to the brain: current state of the art. Eur J Pharm Biopharm. 2014;87(3):433‐444. [DOI] [PubMed] [Google Scholar]
  • 85.Tapeinos C, Battaglini M, Ciofani G. Advances in the design of solid lipid nanoparticles and nanostructured lipid carriers for targeting brain diseases. J Control Release. 2017;264:306‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Anand A, Sugumaran A, Narayanasamy D. Brain targeted delivery of anticancer drugs: prospective approach using solid lipid nanoparticles. IET Nanobiotechnol. 2019;13(4):353‐362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Pandian SRK, Pavadai P, Vellaisamy S, Ravishankar V, Palanisamy P, Sundar LM, et al. Formulation and evaluation of rutin-loaded solid lipid nanoparticles for the treatment of brain tumor. Naunyn Schmiedebergs Arch Pharmacol. 2021;394(4):735‐749. [DOI] [PubMed] [Google Scholar]
  • 88.Ak G, Ünal A, Karakayalı T, Özel B, Selvi Günel N, Hamarat Şanlıer Ş. Brain-targeted, drug-loaded solid lipid nanoparticles against glioblastoma cells in culture. Colloids Surf B Biointerfaces. 2021;206:111‐946. [DOI] [PubMed] [Google Scholar]
  • 89.Li X, Tsibouklis J, Weng T, Zhang B, Yin G, Feng G, et al. Nano carriers for drug transport across the blood–brain barrier. J Drug Target. 2017;25(1):17‐28. [DOI] [PubMed] [Google Scholar]
  • 90.Patel T, Zhou J, Piepmeier JM, Saltzman WM. Polymeric nanoparticles for drug delivery to the central nervous system. Adv Drug Deliv Rev. 2012;64(7):701‐705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Borchardt G, Brandriss S, Kreuter J, Margel S. Body distribution of 75Se-radiolabeled silica nanoparticles covalently coated with co-functionalized surfactants after intravenous injection in rats. J Drug Target. 1994;2(1):61‐77. [DOI] [PubMed] [Google Scholar]
  • 92.Kreuter J. Drug delivery to the central nervous system by polymeric nanoparticles: what do we know? Adv Drug Deliv Rev. 2014;71:2‐14. [DOI] [PubMed] [Google Scholar]
  • 93.Wohlfart S, Gelperina S, Kreuter J. Transport of drugs across the blood–brain barrier by nanoparticles. J Control Release. 2012;161(2):264‐273. [DOI] [PubMed] [Google Scholar]
  • 94.Sulheim E, Iversen TG, To Nakstad V, Klinkenberg G, Sletta H, Schmid R, et al. Cytotoxicity of poly(alkyl cyanoacrylate) nanoparticles. Int J Mol Sci. 2017;18(11):1-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Sulheim E, Baghirov H, von Haartman E, Bøe A, Åslund AK, Mørch Y, et al. Cellular uptake and intracellular degradation of poly(alkyl cyanoacrylate) nanoparticles. J Nanobiotechnol. 2016;14(1):1-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Alonso MJ, Couvreur P. Historical view of the design and development of nanocarriers for overcoming biological barriers. In: Nanostructured Biomaterials for Overcoming Biological Barriers: The Royal Society of Chemistry. 2012:3‐36. [Google Scholar]
  • 97.Lenaerts V, Couvreur P, Christiaens-Leyh D, Joiris E, Roland M, Rollman B, et al. Degradation of poly (isobutyl cyanoacrylate) nanoparticles. Biomaterials. 1984;5(2):65‐68. [DOI] [PubMed] [Google Scholar]
  • 98.Scherer D, Robinson J, Kreuter J. Influence of enzymes on the stability of polybutylcyanoacrylate nanoparticles. Int J Pharm. 1994;101(1-2):165‐168. [Google Scholar]
  • 99.Landry F, Bazile D, Spenlehauer G, Veillard M, Kreuter J. Degradation of poly (D, L-lactic acid) nanoparticles coated with albumin in model digestive fluids (USP XXII). Biomaterials. 1996;17(7):715‐723. [DOI] [PubMed] [Google Scholar]
  • 100.Kreuter J. Nanoparticles and nanocapsules-new dosage forms in the nanometer size range. Pharm Acta Helv. 1978;53:33‐39. [PubMed] [Google Scholar]
  • 101.Nagpal K, Singh SK, Mishra DN. Chitosan nanoparticles: a promising system in novel drug delivery. Chem Pharm Bull. 2010;58(11):1423‐1430. [DOI] [PubMed] [Google Scholar]
  • 102.Karatas H, Aktas Y, Gursoy-Ozdemir Y, Bodur E, Yemisci M, Caban S, et al. A nanomedicine transports a peptide caspase-3 inhibitor across the blood–brain barrier and provides neuroprotection. J Neurosci. 2009;29(44):13761‐13769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang ZH, Wang ZY, Sun CS, Wang CY, Jiang TY, Wang SL. Trimethylated chitosan-conjugated PLGA nanoparticles for the delivery of drugs to the brain. Biomaterials. 2010;31(5):908‐915. [DOI] [PubMed] [Google Scholar]
  • 104.Kreuter J, Petrov V, Kharkevich D, Alyautdin R. Influence of the type of surfactant on the analgesic effects induced by the peptide dalargin after its delivery across the blood–brain barrier using surfactant-coated nanoparticles. J Control Release. 1997;49(1):81‐87. [Google Scholar]
  • 105.Hekmatara T, Bernreuther C, Khalansky A, Theisen A, Weissenberger J, Matschke J, et al. Efficient systemic therapy of rat glioblastoma by nanoparticle-bound doxorubicin is due to antiangiogenic effects. Clin Neuropathol. 2008;28(3):153‐164. [DOI] [PubMed] [Google Scholar]
  • 106.Gelperina S, Maksimenko O, Khalansky A, Vanchugova L, Shipulo E, Abbasova K, et al. Drug delivery to the brain using surfactant-coated poly (lactide-co-glycolide) nanoparticles: influence of the formulation parameters. Eur J Pharm Biopharm. 2010;74(2):157‐163. [DOI] [PubMed] [Google Scholar]
  • 107.Wohlfart S, Khalansky AS, Gelperina S, Maksimenko O, Bernreuther C, Glatzel M, et al. Efficient chemotherapy of rat glioblastoma using doxorubicin-loaded PLGA nanoparticles with different stabilizers. PloS One. 2011;6(5):e19121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Calvo P, Gouritin B, Chacun H, Desmaële D, D'angelo J, Noel J-P, et al. Long-circulating PEGylated polycyanoacrylate nanoparticles as new drug carrier for brain delivery. Pharm Res. 2001;18(8):1157‐1166. [DOI] [PubMed] [Google Scholar]
  • 109.Gref R, Minamitake Y, Peracchia MT, Trebetskoy V, Torchilin V, Langer R. Biodegradable long-circulating polymeric nanospheres. Science. 1994;263(5153):1600‐1604. [DOI] [PubMed] [Google Scholar]
  • 110.Bazile D, Prud'homme C, Bassoullet MT, Marlard M, Spenlehauer G, Veillard M. Stealth Me. PEG–PLA nanoparticles avoid uptake by the mononuclear phagocytes system. J Pharm Sci. 1995;84(4):493‐498. [DOI] [PubMed] [Google Scholar]
  • 111.Kreuter J, Gelperina S. Use of nanoparticles for cerebral cancer. Tumori. 2008;94(2):271. [DOI] [PubMed] [Google Scholar]
  • 112.Ambruosi A, Yamamoto H, Kreuter J. Body distribution of polysorbate–80 and doxorubicin-loaded [14C] poly (butyl cyanoacrylate) nanoparticles after IV administration in rats. J Drug Target. 2005;13(10):535‐542. [DOI] [PubMed] [Google Scholar]
  • 113.Ambruosi A, Khalansky AS, Yamamoto H, Gelperina SE, Begley DJ, Kreuter J. Biodistribution of polysorbate 80-coated doxorubicin-loaded [14C]-poly (butyl cyanoacrylate) nanoparticles after intravenous administration to glioblastoma-bearing rats. J Drug Target. 2006;14(2):97‐105. [DOI] [PubMed] [Google Scholar]
  • 114.Brigger I, Morizet J, Laudani L, Aubert G, Appel M, Velasco V, et al. Negative preclinical results with stealth® nanospheres-encapsulated doxorubicin in an orthotopic murine brain tumor model. J Control Release. 2004;100(1):29‐40. [DOI] [PubMed] [Google Scholar]
  • 115.O'donnell A, Moollan A, Baneham S, Ozgul M, Pabari RM, Cox D, et al. Intranasal and intravenous administration of octa-arginine modified poly (lactic-co-glycolic acid) nanoparticles facilitates central nervous system delivery of loperamide. J Pharm Pharmacol. 2015;67(4):525‐536. [DOI] [PubMed] [Google Scholar]
  • 116.Wagner S, Zensi A, Wien SL, Tschickardt SE, Maier W, Vogel T, et al. Uptake mechanism of ApoE-modified nanoparticles on brain capillary endothelial cells as a blood-brain barrier model. PloS One. 2012;7(3):e32568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Pehlivan SB. Nanotechnology-based drug delivery systems for targeting, imaging and diagnosis of neurodegenerative diseases. Pharm Res. 2013;30(10):2499‐2511. [DOI] [PubMed] [Google Scholar]
  • 118.Ulbrich K, Knobloch T, Kreuter J. Targeting the insulin receptor: nanoparticles for drug delivery across the blood–brain barrier (BBB). J Drug Targeting. 2011;19(2):125‐132. [DOI] [PubMed] [Google Scholar]
  • 119.Liu C, Zhao Z, Gao H, Rostami I, You Q, Jia X, et al. Enhanced blood-brain-barrier penetrability and tumor-targeting efficiency by peptide-functionalized poly(amidoamine) dendrimer for the therapy of gliomas. Nanotheranostics. 2019;3(4):311‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhu Y, Liu C, Pang Z. Dendrimer-based drug delivery systems for brain targeting. Biomolecules. 2019;9(12):1-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Liaw K, Zhang F, Mangraviti A, Kannan S, Tyler B, Kannan RM. Dendrimer size effects on the selective brain tumor targeting in orthotopic tumor models upon systemic administration. Bioeng Transl Med. 2020;5(2):e10160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kaanumalle LS, Ramesh R, Murthy Maddipatla VS, Nithyanandhan J, Jayaraman N, Ramamurthy V. Dendrimers as photochemical reaction media. Photochemical behavior of unimolecular and bimolecular reactions in water-soluble dendrimers. J Org Chem. 2005;70(13):5062‐5069. [DOI] [PubMed] [Google Scholar]
  • 123.Newkome GR, Yao Z, Baker GR, Gupta VK. Micelles. Part 1. Cascade molecules: a new approach to micelles. A [27]-arborol. J Org Chem. 1985;50(11):2003‐2004. [Google Scholar]
  • 124.Hermanson G. Bioconjugate Techniques. 2nd edn. Academic Press; 2008. [Google Scholar]
  • 125.Zarebkohan A, Najafi F, Moghimi HR, Hemmati M, Deevband MR, Kazemi B. Synthesis and characterization of a PAMAM dendrimer nanocarrier functionalized by SRL peptide for targeted gene delivery to the brain. Eur J Pharm Sci: Official J Eur Federation Pharm Sci. 2015;78:19‐30. [DOI] [PubMed] [Google Scholar]
  • 126.He H, Li Y, Jia XR, Du J, Ying X, Lu WL, et al. PEGylated poly(amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials. 2011;32(2):478‐487. [DOI] [PubMed] [Google Scholar]
  • 127.Huang RQ, Qu YH, Ke WL, Zhu JH, Pei YY, Jiang C. Efficient gene delivery targeted to the brain using a transferrin-conjugated polyethyleneglycol-modified polyamidoamine dendrimer. FASEB J. 2007;21(4):1117‐1125. [DOI] [PubMed] [Google Scholar]
  • 128.O'Mahony AM, Godinho BM, Cryan JF, O'Driscoll CM. Non-viral nanosystems for gene and small interfering RNA delivery to the central nervous system: formulating the solution. J Pharm Sci. 2013;102(10):3469‐3484. [DOI] [PubMed] [Google Scholar]
  • 129.Ofek P, Fischer W, Calderon M, Haag R, Satchi-Fainaro R. In vivo delivery of small interfering RNA to tumors and their vasculature by novel dendritic nanocarriers. FASEB J. 2010;24(9):3122‐3134. [DOI] [PubMed] [Google Scholar]
  • 130.Meng J, Agrahari V, Youm I. Advances in targeted drug delivery approaches for the central nervous system tumors: the inspiration of nanobiotechnology. J Neuroimm Pharmacol. 2017;12(1):84‐98. [DOI] [PubMed] [Google Scholar]
  • 131.Kesharwani P, Amin MCIM, Giri N, Jain A, Gajbhiye V. Dendrimers in targeting and delivery of drugs. Nanotechnol-Based Approaches Target Deliv Drugs Genes. 2017;363. [Google Scholar]
  • 132.Jain K, Kesharwani P, Gupta U, Jain NK. Dendrimer toxicity: let’s meet the challenge. Int J Pharm. 2010;394(1-2):122‐142. [DOI] [PubMed] [Google Scholar]
  • 133.Zhang P, Hu L, Yin Q, Feng L, Li Y. Transferrin-modified c [RGDfK]-paclitaxel loaded hybrid micelle for sequential blood-brain barrier penetration and glioma targeting therapy. Mol Pharm. 2012;9(6):1590‐1598. [DOI] [PubMed] [Google Scholar]
  • 134.Lu L, Zhao X, Fu T, Li K, He Y, Luo Z, et al. An iRGD-conjugated prodrug micelle with blood-brain-barrier penetrability for anti-glioma therapy. Biomaterials. 2020;230(119666):1–58. [DOI] [PubMed] [Google Scholar]
  • 135.Thotakura N, Parashar P, Raza K. Assessing the pharmacokinetics and toxicology of polymeric micelle conjugated therapeutics. Expert Opin Drug Metab Toxicol. 2021;17(3):323‐332. [DOI] [PubMed] [Google Scholar]
  • 136.Patel MM, Patel BM. Crossing the blood-brain barrier: recent advances in drug delivery to the brain. CNS Drugs. 2017;31(2):109‐133. [DOI] [PubMed] [Google Scholar]
  • 137.Romano E, Netti PA, Torino E. Exosomes in gliomas: biogenesis, isolation, and preliminary applications in nanomedicine. Pharm (Basel). 2020;13(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Rufino-Ramos D, Albuquerque PR, Carmona V, Perfeito R, Nobre RJ, de Almeida L P. Extracellular vesicles: novel promising delivery systems for therapy of brain diseases. J Control Release. 2017;262:247‐258. [DOI] [PubMed] [Google Scholar]
  • 139.Elliott RO, He M. Unlocking the power of exosomes for crossing biological barriers in drug delivery. Pharmaceutics. 2021;13(1):1-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Hu CM, Zhang L, Aryal S, Cheung C, Fang RH, Zhang L. Erythrocyte membrane-camouflaged polymeric nanoparticles as a biomimetic delivery platform. Proc Natl Acad Sci U S A. 2011;108(27):10980‐10985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kalani A, Kamat PK, Chaturvedi P, Tyagi SC, Tyagi N. Curcumin-primed exosomes mitigate endothelial cell dysfunction during hyperhomocysteinemia. Life Sci. 2014;107(1-2):1‐7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Fernandes M, Lopes I, Magalhães L, Sárria MP, Machado R, Sousa JC, et al. Novel concept of exosome-like liposomes for the treatment of Alzheimer’s disease. J Control Release. 2021;336:130‐143. [DOI] [PubMed] [Google Scholar]
  • 143.Morad G, Carman CV, Hagedorn EJ, Perlin JR, Zon LI, Mustafaoglu N, et al. Tumor-derived extracellular vesicles breach the intact blood-brain barrier via transcytosis. ACS Nano. 2019;13(12):13853‐13865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hadla M, Palazzolo S, Corona G, Caligiuri I, Canzonieri V, Toffoli G, et al. Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse models. Nanomedicine (Lond). 2016;11(18):2431‐2441. [DOI] [PubMed] [Google Scholar]
  • 145.Alvarez-Erviti L, Seow Y, Yin H, Betts C, Lakhal S, Wood MJA. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol. 2011;29(4):341‐345. [DOI] [PubMed] [Google Scholar]
  • 146.Zhang Z, Guo X, Guo X, Yu R, Qian M, Wang S, et al. MicroRNA-29a-3p delivery via exosomes derived from engineered human mesenchymal stem cells exerts tumour suppressive effects by inhibiting migration and vasculogenic mimicry in glioma. Aging (Albany NY). 2021;13(4):5055‐5068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Jiang J, Lu J, Wang X, Sun B, Liu X, Ding Y, et al. Glioma stem cell-derived exosomal miR-944 reduces glioma growth and angiogenesis by inhibiting AKT/ERK signaling. Aging (Albany NY). 2021;13(15):19243‐19259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Yang T, Fogarty B, LaForge B, Aziz S, Pham T, Lai L, et al. Delivery of small interfering RNA to inhibit vascular endothelial growth factor in zebrafish using natural brain endothelia cell-secreted exosome nanovesicles for the treatment of brain cancer. AAPS J. 2017;19(2):475‐486. [DOI] [PubMed] [Google Scholar]
  • 149.Borna H, Imani S, Iman M, Azimzadeh Jamalkandi S. Therapeutic face of RNAi: in vivo challenges. Expert Opin Biol Ther. 2015;15(2):269‐285. [DOI] [PubMed] [Google Scholar]
  • 150.Busatto S, Morad G, Guo P, Moses MA. The role of extracellular vesicles in the physiological and pathological regulation of the blood-brain barrier. FASEB Bioadv. 2021;3(9):665‐675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Chen CC, Liu L, Ma F, Wong CW, Guo XE, Chacko JV, et al. Elucidation of exosome migration across the blood-brain barrier model in vitro. Cell Mol Bioeng. 2016;9(4):509‐529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Xin H, Li Y, Cui Y, Yang JJ, Zhang ZG, Chopp M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab. 2013;33(11):1711‐1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Zhang Y, Chopp M, Meng Y, Katakowski M, Xin H, Mahmood A, et al. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J Neurosurg. 2015;122(4):856‐867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Jia G, Han Y, An Y, Ding Y, He C, Wang X, et al. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials. 2018;178:302‐316. [DOI] [PubMed] [Google Scholar]
  • 155.Katakowski M, Chopp M. Exosomes as tools to suppress primary brain tumor. Cell Mol Neurobiol. 2016;36(3):343‐352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Bu N, Wu H, Zhang G, Zhan S, Zhang R, Sun H, et al. Exosomes from dendritic cells loaded with chaperone-rich cell lysates elicit a potent T cell immune response against intracranial glioma in mice. J Mol Neurosci. 2015;56(3):631‐643. [DOI] [PubMed] [Google Scholar]
  • 157.Stoorvogel W, Kleijmeer MJ, Geuze HJ, Raposo G. The biogenesis and functions of exosomes. Traffic. 2002;3(5):321‐330. [DOI] [PubMed] [Google Scholar]
  • 158.Hao S, Bai O, Li F, Yuan J, Laferte S, Xiang J. Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology. 2007;120(1):90‐102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Zeng Y, Feng H, Graner MW, Katsanis E. Tumor-derived, chaperone-rich cell lysate activates dendritic cells and elicits potent antitumor immunity. Blood. 2003;101(11):4485‐4491. [DOI] [PubMed] [Google Scholar]
  • 160.Mulvihill JJ, Cunnane EM, Ross AM, Duskey JT, Tosi G, Grabrucker AM. Drug delivery across the blood-brain barrier: recent advances in the use of nanocarriers. Nanomedicine (Lond). 2020;15(2):205‐214. [DOI] [PubMed] [Google Scholar]
  • 161.Yang T, Martin P, Fogarty B, Brown A, Schurman K, Phipps R, et al. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio Rerio. Pharm Res. 2015;32(6):2003‐2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Nooshabadi VT, Khanmohammadi M, Shafei S, Banafshe HR, Malekshahi ZV, Ebrahimi-Barough S, et al. Impact of atorvastatin loaded exosome as an anti-glioblastoma carrier to induce apoptosis of U87 cancer cells in 3D culture model. Biochem Biophys Rep. 2020;23(100792):1-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Follet J, Rémy L, Hesry V, Simon B, Gillet D, Auvray P, et al. Adaptation to statins restricts human tumour growth in Nude mice. BMC Cancer. 2011;11(491):1-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Vallianou NG, Kostantinou A, Kougias M, Kazazis C. Statins and cancer. Anticancer Agents Med Chem. 2014;14(5):706-712. [DOI] [PubMed] [Google Scholar]
  • 165.Salunkhe S, Basak M, Chitkara D, Mittal A. Surface functionalization of exosomes for target-specific delivery and in vivo imaging & tracking: Strategies and significance. J Control Release. 2020;326:599‐614. [DOI] [PubMed] [Google Scholar]
  • 166.Théry C, Zitvogel L, Amigorena S. Exosomes: composition, biogenesis and function. Nat Rev Immunol. 2002;2(8):569‐579. [DOI] [PubMed] [Google Scholar]
  • 167.Vogt S, Stadlmayr G, Grillari J, Ruker F, Wozniak-Knopp G. Engineering of surface proteins in extracellular vesicles for tissue-specific targeting. Curr Top Biochem Eng. 2019:1-21. [Google Scholar]
  • 168.Tian Y, Li S, Song J, Ji T, Zhu M, Anderson GJ, et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35(7):2383‐2390. [DOI] [PubMed] [Google Scholar]
  • 169.Mentkowski KI, Snitzer JD, Rusnak S, Lang JK. Therapeutic potential of engineered extracellular vesicles. AAPS J. 2018;20(3):50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Smyth T, Petrova K, Payton NM, Persaud I, Redzic JS, Graner MW, et al. Surface functionalization of exosomes using click chemistry. Bioconjugate Chem. 2014;25(10):1777‐1784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Aryani A, Denecke B. Exosomes as a nanodelivery system: a key to the future of neuromedicine? Mol Neurobiol. 2016;53(2):818‐834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Cheng Z, Li M, Dey R, Chen Y. Nanomaterials for cancer therapy: current progress and perspectives. J Hematol Oncol. 2021;14(1):85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Haney MJ, Klyachko NL, Zhao Y, Gupta R, Plotnikova EG, He Z, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18‐30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Pegtel DM, Gould SJ. Exosomes. Ann Rev Biochem. 2019;88:487‐514. [DOI] [PubMed] [Google Scholar]
  • 175.Wei W, Ao Q, Wang X, Cao Y, Liu Y, Zheng SG, et al. Mesenchymal stem cell-derived exosomes: a promising biological tool in nanomedicine. Front Pharmacol. 2020;11:590470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Kooijmans SA, Vader P, van Dommelen SM, van Solinge WW, Schiffelers RM. Exosome mimetics: a novel class of drug delivery systems. Int J Nanomed. 2012;7:1525‐1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Silverman JM, Reiner NE. Exosomes and other microvesicles in infection biology: organelles with unanticipated phenotypes. Cell Microbiol. 2011;13(1):1‐9. [DOI] [PubMed] [Google Scholar]
  • 178.MacDiarmid JA, Brahmbhatt H. Minicells: versatile vectors for targeted drug or si/shRNA cancer therapy. Curr Opin Biotechnol. 2011;22(6):909‐916. [DOI] [PubMed] [Google Scholar]
  • 179.Parti RP, Biswas D, Wang M, Liao M, Dillon JA. A minD mutant of enterohemorrhagic E. coli O157:H7 has reduced adherence to human epithelial cells. Microb Pathog. 2011;51(5):378‐383. [DOI] [PubMed] [Google Scholar]
  • 180.Farley MM, Hu B, Margolin W, Minicells LJ. Back in fashion. J Bacteriol. 2016;198(8):1186‐1195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Jivrajani M, Shrivastava N, Nivsarkar M. A combination approach for rapid and high yielding purification of bacterial minicells. J Microbiol Methods. 2013;92(3):340‐343. [DOI] [PubMed] [Google Scholar]
  • 182.Lee JY, Choy HE, Lee JH, Kim GJ. Generation of minicells from an endotoxin-free Gram-positive strain Corynebacterium glutamicum. J Microbiol Biotechnol. 2015;25(4):554‐558. [DOI] [PubMed] [Google Scholar]
  • 183.van der Meel R, Vehmeijer LJ, Kok RJ, Storm G, van Gaal EV. Ligand-targeted particulate nanomedicines undergoing clinical evaluation: current status. Adv Drug Deliv Rev. 2013;65(10):1284-1298. [DOI] [PubMed] [Google Scholar]
  • 184.Ali MK, Liu Q, Liang K, Li P, Kong Q. Bacteria-derived minicells for cancer therapy. Cancer Lett. 2020;491:11‐21. [DOI] [PubMed] [Google Scholar]
  • 185.MacDiarmid JA, Mugridge NB, Weiss JC, Phillips L, Burn AL, Paulin RP, et al. Bacterially derived 400 nm particles for encapsulation and cancer cell targeting of chemotherapeutics. Cancer Cell. 2007;11(5):431‐445. [DOI] [PubMed] [Google Scholar]
  • 186.Karagiannis ED, Anderson DG. Minicells overcome tumor drug-resistance. Nat Biotechnol. 2009;27(7):620‐621. [DOI] [PubMed] [Google Scholar]
  • 187.Kwan K, Schneider JR, Kobets A, Boockvar JA. Targeting epidermal growth factor receptors in recurrent glioblastoma via a novel epithelial growth factor receptor-conjugated nanocell doxorubicin delivery system. Neurosurgery. 2018;82(3):N23‐N24. [DOI] [PubMed] [Google Scholar]
  • 188.Camilla U R, Proenca A, Buetz C, Shi C, Chao L, Bowman Grant R. Minicells as a damage disposal mechanism in Escherichia coli. mSphere. 3(5):e00428‐e00518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.De Jong WH, Borm PJ. Drug delivery and nanoparticles: applications and hazards. Int J Nanomed. 2008;3(2):133‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Khan MB, Ruggieri R, Jamil E, Tran NL, Gonzalez C, Mugridge N, et al. Nanocell-mediated delivery of miR-34a counteracts temozolomide resistance in glioblastoma. Mol Med. 2021;27(1):28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Akgül S, Patch AM, D'Souza RCJ, Mukhopadhyay P, Nones K, Kempe S, et al. Intratumoural heterogeneity underlies distinct therapy responses and treatment resistance in glioblastoma. Cancers (Basel). 2019;11(2):1-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Karagkouni D, Paraskevopoulou MD, Chatzopoulos S, Vlachos IS, Tastsoglou S, Kanellos I, et al. DIANA-TarBase v8: a decade-long collection of experimentally supported miRNA-gene interactions. Nucl Acids Res. 2018;46(D1):D239‐D245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.MacDiarmid JA, Langova V, Bailey D, Pattison ST, Pattison SL, Christensen N, et al. Targeted doxorubicin delivery to brain tumors via minicells: proof of principle using dogs with spontaneously occurring tumors as a model. PLoS One. 2016;11(4):e0151832. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Eskilsson E, Røsland GV, Solecki G, Wang Q, Harter PN, Graziani G, et al. EGFR heterogeneity and implications for therapeutic intervention in glioblastoma. Neuro Oncol. 2018;20(6):743‐752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.van Zandwijk N, Pavlakis N, Kao SC, Linton A, Boyer MJ, Clarke S, et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 2017;18(10):1386‐1396. [DOI] [PubMed] [Google Scholar]
  • 196.Solomon BJ, Desai J, Rosenthal M, McArthur GA, Pattison ST, Pattison SL, et al. A first-time-in-human phase I clinical trial of bispecific antibody-targeted, paclitaxel-packaged bacterial minicells. PLoS One. 2015;10(12):e0144559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.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(12):1889‐1894. [DOI] [PubMed] [Google Scholar]
  • 198.Wang X, Wang X, Bai X, Yan L, Liu T, Wang M, et al. Nanoparticle ligand exchange and its effects at the nanoparticle-cell membrane interface. Nano Lett. 2019;19(1):8‐18. [DOI] [PubMed] [Google Scholar]
  • 199.Wang X, Cui X, Zhao Y, Chen C. Nano-bio interactions: the implication of size-dependent biological effects of nanomaterials. Sci Chin Life Sci. 2020;63(8):1168‐1182. [DOI] [PubMed] [Google Scholar]
  • 200.Flemming A. Minicells deliver lethal load to tumours. Nat Rev Drug Discov. 2007;6(7):519. [Google Scholar]
  • 201.Khameneh B, Iranshahy M, Ghandadi M, Ghoochi Atashbeyk D, Fazly Bazzaz BS, Iranshahi M. Investigation of the antibacterial activity and efflux pump inhibitory effect of co-loaded piperine and gentamicin nanoliposomes in methicillin-resistant Staphylococcus aureus. Drug Dev Ind Pharm. 2015;41(6):989‐994. [DOI] [PubMed] [Google Scholar]
  • 202.Moghadas-Sharif N, Fazly Bazzaz BS, Khameneh B, Malaekeh-Nikouei B. The effect of nanoliposomal formulations on Staphylococcus epidermidis biofilm. Drug Dev Ind Pharm. 2015;41(3):445‐450. [DOI] [PubMed] [Google Scholar]
  • 203.Zahmatkeshan M, Gheybi F, Rezayat SM, Jaafari MR. Improved drug delivery and therapeutic efficacy of PEgylated liposomal doxorubicin by targeting anti-HER2 peptide in murine breast tumor model. Eur J Pharm Sci: Official J Eur Federation Pharm Sci. 2016;86:125‐135. [DOI] [PubMed] [Google Scholar]
  • 204.Agrawal MA, Tripathi DK, Saraf S, Saraf S, Antimisiaris SG, et al. Recent advancements in liposomes targeting strategies to cross blood-brain barrier (BBB) for the treatment of Alzheimer’s disease. J Control Release. 2017;260:61‐77. [DOI] [PubMed] [Google Scholar]
  • 205.Sheoran R, Khokra SL, Chawla V, Dureja H. Recent patents, formulation techniques, classification and characterization of liposomes. Recent Pat Nanotechnol. 2019;13(1):17‐27. [DOI] [PubMed] [Google Scholar]
  • 206.Papagiannopoulos A, Pippa N, Demetzos C, Pispas S, Radulescu A. Lamellarity and size distributions in mixed DPPC/amphiphilic poly(2-oxazoline) gradient copolymer vesicles and their temperature response. Chem Phys Lipids. 2021;234:105008. [DOI] [PubMed] [Google Scholar]
  • 207.Di Muzio M, Millan-Solsona R, Dols-Perez A, Borrell JH, Fumagalli L, Gomila G. Dielectric properties and lamellarity of single liposomes measured by in-liquid scanning dielectric microscopy. J Nanobiotechnol. 2021;19(1):167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Moosavian SA, Abnous K, Badiee A, Jaafari MR. Improvement in the drug delivery and anti-tumor efficacy of PEGylated liposomal doxorubicin by targeting RNA aptamers in mice bearing breast tumor model. Colloids Surf B Biointerfaces. 2016;139:228‐236. [DOI] [PubMed] [Google Scholar]
  • 209.Alavizadeh SH, Akhtari J, Badiee A, Golmohammadzadeh S, Jaafari MR. Improved therapeutic activity of HER2 affibody-targeted cisplatin liposomes in HER2-expressing breast tumor models. Expert Opin Drug Deliv. 2016;13(3):325‐336. [DOI] [PubMed] [Google Scholar]
  • 210.Gabizon A, Shmeeda H, Barenholz Y. Pharmacokinetics of PEGylated liposomal doxorubicin: review of animal and human studies. Clin Pharmacokinet. 2003;42(5):419‐436. [DOI] [PubMed] [Google Scholar]
  • 211.Garcia-Garcia E, Andrieux K, Gil S, Couvreur P. Colloidal carriers and blood-brain barrier (BBB) translocation: a way to deliver drugs to the brain? Int J Pharm. 2005;298(2):274‐292. [DOI] [PubMed] [Google Scholar]
  • 212.Arabi L, Badiee A, Mosaffa F, Jaafari MR. Targeting CD44 expressing cancer cells with anti-CD44 monoclonal antibody improves cellular uptake and antitumor efficacy of liposomal doxorubicin. J Control Release. 2015;220(Pt A):275‐286. [DOI] [PubMed] [Google Scholar]
  • 213.Chen X, Yuan M, Zhang Q, Ting Yang Y, Gao H, He Q. Synergistic combination of doxorubicin and paclitaxel delivered by blood brain barrier and glioma cells dual targeting liposomes for chemotherapy of brain glioma. Curr Pharm Biotechnol. 2016;17(7):636‐650. [DOI] [PubMed] [Google Scholar]
  • 214.Shi K, Long Y, Xu C, Wang Y, Qiu Y, Yu Q, et al. Liposomes combined an integrin alphavbeta3-specific vector with pH-responsible cell-penetrating property for highly effective antiglioma therapy through the blood-brain barrier. ACS Appl Mater Interfaces. 2015;7(38):21442‐21454. [DOI] [PubMed] [Google Scholar]
  • 215.Du J, Lu WL, Ying X, Liu Y, Du P, Tian W, et al. Dual-targeting topotecan liposomes modified with tamoxifen and wheat germ agglutinin significantly improve drug transport across the blood-brain barrier and survival of brain tumor-bearing animals. Mol Pharm. 2009;6(3):905‐917. [DOI] [PubMed] [Google Scholar]
  • 216.Siegal T, Horowitz A, Gabizon A. Doxorubicin encapsulated in sterically stabilized liposomes for the treatment of a brain tumor model: biodistribution and therapeutic efficacy. J Neurosurg. 1995;83(6):1029‐1037. [DOI] [PubMed] [Google Scholar]
  • 217.Saito R, Bringas JR, McKnight TR, Wendland MF, Mamot C, Drummond DC, et al. Distribution of liposomes into brain and rat brain tumor models by convection-enhanced delivery monitored with magnetic resonance imaging. Cancer Res. 2004;64(7):2572‐2579. [DOI] [PubMed] [Google Scholar]
  • 218.Gilmore JL, Yi X, Quan L, Kabanov AV. Novel nanomaterials for clinical neuroscience. J Neuroimmune Pharmacol. 2008;3(2):83‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Gong W, Wang Z, Liu N, Lin W, Wang X, Xu D, et al. Improving efficiency of adriamycin crossing blood brain barrier by combination of thermosensitive liposomes and hyperthermia. Biol Pharm Bull. 2011;34(7):1058‐1064. [DOI] [PubMed] [Google Scholar]
  • 220.Wang G, Wu B, Li Q, Chen S, Jin X, Liu Y, et al. Active transportation of liposome enhances tumor accumulation, penetration, and therapeutic efficacy. Small. 2020;16(44):e2004172. [DOI] [PubMed] [Google Scholar]
  • 221.Sakurai Y, Kato A, Harashima H. Involvement of caveolin-1-mediated transcytosis in the intratumoral accumulation of liposomes. Biochem Biophys Res Commun. 2020;525(2):313‐318. [DOI] [PubMed] [Google Scholar]
  • 222.Lakkadwala S, Dos Santos Rodrigues B, Sun C, Singh J. Dual functionalized liposomes for efficient co-delivery of anti-cancer chemotherapeutics for the treatment of glioblastoma. J Control Release. 2019;307:247‐260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Gao JQ, Lv Q, Li LM, Tang XJ, Li FZ, Hu YL, et al. Glioma targeting and blood-brain barrier penetration by dual-targeting doxorubincin liposomes. Biomaterials. 2013;34(22):5628‐5639. [DOI] [PubMed] [Google Scholar]
  • 224.Fukuta T, Asai T, Sato A, Namba M, Yanagida Y, Kikuchi T, et al. Neuroprotection against cerebral ischemia/reperfusion injury by intravenous administration of liposomal fasudil. Int J Pharm. 2016;506(1-2):129‐137. [DOI] [PubMed] [Google Scholar]
  • 225.Jain A J. Advances in tumor targeted liposomes. Curr Mol Med. 2018;18(1):44‐57. [DOI] [PubMed] [Google Scholar]
  • 226.Park J, Choi Y, Chang H, Um W, Ryu JH, Kwon IC. Alliance with EPR effect: combined strategies to improve the EPR effect in the tumor microenvironment. Theranostics. 2019;9(26):8073‐8090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Kalyane D, Raval N, Maheshwari R, Tambe V, Kalia K, Tekade RK. 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 Mater Biol Appl. 2019;98:1252‐1276. [DOI] [PubMed] [Google Scholar]
  • 228.Shi Y, van der Meel R, Chen X, Lammers T. The EPR effect and beyond: Strategies to improve tumor targeting and cancer nanomedicine treatment efficacy. Theranostics. 2020;10(17):7921‐7924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.McDannold N, Vykhodtseva N, Hynynen K. Targeted disruption of the blood–brain barrier with focused ultrasound: association with cavitation activity. Phys Med Biol. 2006;51(4):793. [DOI] [PubMed] [Google Scholar]
  • 230.Shen Y, Guo J, Chen G, Chin CT, Chen X, Chen J, et al. Delivery of liposomes with different sizes to mice brain after sonication by focused ultrasound in the presence of microbubbles. Ultrasound Med Biol. 2016;42(7):1499‐1511. [DOI] [PubMed] [Google Scholar]
  • 231.Yoshida J, Mizuno M. Clinical gene therapy for brain tumors. Liposomal delivery of anticancer molecule to glioma. J Neurooncol. 2003;65(3):261‐267. [DOI] [PubMed] [Google Scholar]
  • 232.Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Deliv Rev. 2016;99(Pt A):28‐51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Sugawa N, Ueda S, Nakagawa Y, Nishino H, Nosaka K, Iwashima A, et al. An antisense EGFR oligodeoxynucleotide enveloped in lipofectin induces growth inhibition in human malignant gliomas in vitro. J Neurooncol. 1998;39(3):237‐244. [DOI] [PubMed] [Google Scholar]
  • 234.Lu W, Zhang Y, Tan Y-Z, Hu K-L, Jiang X-G, Fu S-K. Cationic albumin-conjugated PEGylated nanoparticles as novel drug carrier for brain delivery. J Control Release. 2005;107(3):428‐448. [DOI] [PubMed] [Google Scholar]
  • 235.Abbott NJ, Rönnbäck L, Hansson E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat Rev Neurosci. 2006;7(1):41‐53. [DOI] [PubMed] [Google Scholar]
  • 236.Triguero D, Buciak J, Pardridge WM. Capillary depletion method for quantification of blood–brain barrier transport of circulating peptides and plasma proteins. J Neurochem. 1990;54(6):1882‐1888. [DOI] [PubMed] [Google Scholar]
  • 237.Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8(1):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Rastgoo M, Hosseinzadeh H, Alavizadeh H, Abbasi A, Ayati Z, Jaafari MR. Antitumor activity of PEGylated nanoliposomes containing crocin in mice bearing C26 colon carcinoma. Planta Med. 2013;79(6):447‐451. [DOI] [PubMed] [Google Scholar]
  • 239.Nektar. Available from: http://www.nektar.com/pipeline/rd-pipeline/onzeald.
  • 240.Jameson GS, Hamm JT, Weiss GJ, Alemany C, Anthony SP, Basche M, et al. A multicenter, phase I, dose-escalation study to assess the safety, tolerability and pharmacokinetics of Etirinotecan Pegol in patients with refractory solid tumors. Clin Cancer Res. 2012:1201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug?cdrid=586949.
  • 242.Cutler JI, Auyeung E, Mirkin CA. Spherical nucleic acids. J Am Chem Soc. 2012;134(3):1376‐1391. [DOI] [PubMed] [Google Scholar]
  • 243.Kumthekar P, Ko CH, Paunesku T, Dixit K, Sonabend AM, Bloch O, et al. A first-in-human phase 0 clinical study of RNA interference-based spherical nucleic acids in patients with recurrent glioblastoma. Sci Transl Med. 2021;13:584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Jensen SA, Day ES, Ko CH, Hurley LA, Luciano JP, Kouri FM, et al. Spherical nucleic acid nanoparticle conjugates as an RNAi-based therapy for glioblastoma. Sci Transl Med. 2013;5(209):209ra152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Melamed JR, Ioele SA, Hannum AJ, Ullman VM, Day ES. Polyethylenimine-spherical nucleic acid nanoparticles against Gli1 reduce the chemoresistance and stemness of glioblastoma cells. Mol Pharm. 2018;15(11):5135‐5145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Stegh AH, Kim H, Bachoo RM, Forloney KL, Zhang J, Schulze H, et al. Bcl2L12 inhibits post-mitochondrial apoptosis signaling in glioblastoma. Genes Dev. 2007;21(1):98‐111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 247.Scorilas A, Kyriakopoulou L, Yousef GM, Ashworth LK, Kwamie A, Diamandis EP. Molecular cloning, physical mapping, and expression analysis of a novel gene, BCL2L12, encoding a proline-rich protein with a highly conserved BH2 domain of the Bcl-2 family. Genomics. 2001;72(2):217‐221. [DOI] [PubMed] [Google Scholar]
  • 248.Kouri FM, Hurley LA, Daniel WL, Day ES, Hua Y, Hao L, et al. miR-182 integrates apoptosis, growth, and differentiation programs in glioblastoma. Genes Dev. 2015;29(7):732‐745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249.Stegh AH, Kesari S, Mahoney JE, Jenq HT, Forloney KL, Protopopov A, et al. Bcl2L12-mediated inhibition of effector caspase-3 and caspase-7 via distinct mechanisms in glioblastoma. Proc Natl Acad Sci U S A. 2008;105(31):10703‐10708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Stegh AH, Brennan C, Mahoney JA, Forloney KL, Jenq HT, Luciano JP, et al. Glioma oncoprotein Bcl2L12 inhibits the p53 tumor suppressor. Genes Dev. 2010;24(19):2194‐2204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.NU-0129. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug?cdrid=786841.
  • 252.Andrew Brenner M. PhD. Rhenium as a cytotoxic isotope 2015 [Available from: http://www.onclive.com/publications/obtn/2015/april-2015/liposomal-encapsulation-of-radiotherapeutic-holds-promise-in-treating-glioblastoma.
  • 253.Hsu C-W, Chang Y-J, Chang C-H, Chen L-C, Lan K-L, Ting G, et al. Comparative therapeutic efficacy of rhenium-188 radiolabeled-liposome and 5-fluorouracil in LS-174T human colon carcinoma solid tumor xenografts. Cancer Biotherapy Radiopharm. 2012;27(8):481‐489. [DOI] [PubMed] [Google Scholar]
  • 254.Rhenium nanoliposomes.gov. Available from: https://www.cancer.gov/publications/dictionaries/cancer-drug?cdrid=751420.
  • 255.Rhenium liposomal. Available from: https://clinicaltrials.gov/ct2/show/NCT01906385?term=nanoliposome&cond=glioma&rank=1.
  • 256.Gaillard PJ. Case study: to-BBB's G-Technology, getting the best from drug-delivery research with industry-academia partnerships. Ther Deliv. 2011;2(11):1391‐1394. [DOI] [PubMed] [Google Scholar]
  • 257.Gaillard PJ, Appeldoorn CC, Rip J, Dorland R, van der Pol SM, Kooij G, et al. Enhanced brain delivery of liposomal methylprednisolone improved therapeutic efficacy in a model of neuroinflammation. J Control Release. 2012;164(3):364‐369. [DOI] [PubMed] [Google Scholar]
  • 258.Greenwood J, Hammarlund-Udenaes M, Jones HC, Stitt AW, Vandenbroucke RE, Romero IA, et al. Correction to: current research into brain barriers and the delivery of therapeutics for neurological diseases: a report on CNS barrier congress London, UK, 2017. Fluids Barriers CNS. 2018;15(1):3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Hu Y, Gaillard PJ, de Lange ECM, Hammarlund-Udenaes M. Targeted brain delivery of methotrexate by glutathione PEGylated liposomes: How can the formulation make a difference? Eur J Pharm Biopharm. 2019;139:197‐204. [DOI] [PubMed] [Google Scholar]
  • 260.Birngruber T, Raml R, Gladdines W, Gatschelhofer C, Gander E, Ghosh A, et al. Enhanced doxorubicin delivery to the brain administered through glutathione PEGylated liposomal doxorubicin (2B3-101) as compared with generic Caelyx,(®)/Doxil(®)—a cerebral open flow microperfusion pilot study. J Pharm Sci. 2014;103(7):1945‐1948. [DOI] [PubMed] [Google Scholar]
  • 261.Hu Y, Rip J, Gaillard PJ, de Lange ECM, Hammarlund-Udenaes M. The impact of liposomal formulations on the release and brain delivery of methotrexate: an in vivo microdialysis study. J Pharm Sci. 2017;106(9):2606‐2613. [DOI] [PubMed] [Google Scholar]
  • 262.Gaillard PJ, Appeldoorn CC, Dorland R, van Kregten J, Manca F, Vugts DJ, et al. Pharmacokinetics, brain delivery, and efficacy in brain tumor-bearing mice of glutathione PEGylated liposomal doxorubicin (2B3-101). PloS One. 2014;9(1):e82331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Birngruber T, Raml R, Gladdines W, Gatschelhofer C, Gander E, Ghosh A, et al. Enhanced doxorubicin delivery to the brain administered through glutathione PEGylated liposomal doxorubicin (2B3-101) as compared with generic Caelyx,®/Doxil®—a cerebral open flow microperfusion pilot study. J Pharm Sci. 2014;103(7):1945‐1948. [DOI] [PubMed] [Google Scholar]
  • 264.Doxil for glioblastoma. 2009. Available from: https://clinicaltrials.gov/ct2/show/NCT00944801?term=liposome&cond=glioma&draw=1&rank=7.
  • 265.Sagnella SM, Trieu J, Brahmbhatt H, MacDiarmid JA, MacMillan A, Whan RM, et al. Targeted doxorubicin-loaded bacterially derived nano-cells for the treatment of neuroblastoma. Mol Cancer Ther. 2018;17(5):1012‐1023. [DOI] [PubMed] [Google Scholar]
  • 266.Taylor K, Howard CB, Jones ML, Sedliarou I, MacDiarmid J, Brahmbhatt H, et al., editors. Nanocell targeting using engineered bispecific antibodies. MAbs; 2015: Taylor & Francis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.EnGeneIC Delivery Vehicle or EDVTM. Available from: https://clinicaltrials.gov/ct2/show/NCT02766699?term=liposome&cond=glioma&draw=2&rank=14. [Google Scholar]
  • 268.Sagnella SM, Yang L, Stubbs GE, Boslem E, Martino-Echarri E, Smolarczyk K, et al. Cyto-immuno-therapy for cancer: a pathway elicited by tumor-targeted, cytotoxic drug-packaged bacterially derived nanocells. Cancer Cell. 2020;37(3):354‐370.e7. [DOI] [PubMed] [Google Scholar]
  • 269.Emens LA, Ascierto PA, Darcy PK, Demaria S, Eggermont AMM, Redmond WL, et al. Cancer immunotherapy: opportunities and challenges in the rapidly evolving clinical landscape. Eur J Cancer. 2017;81:116‐129. [DOI] [PubMed] [Google Scholar]
  • 270.Agrahari V, Zhang C, Zhang T, Li W, Gounev TK, Oyler NA, et al. Hyaluronidase-sensitive nanoparticle templates for triggered release of HIV/AIDS microbicide in vitro. AAPS J. 2014;16(2):181‐193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Youm I, Agrahari V, Murowchick JB, Youan B-BC. Uptake and cytotoxicity of docetaxel-loaded hyaluronic acid-grafted oily core nanocapsules in MDA-MB 231 cancer cells. Pharm Res. 2014;31(9):2439‐2452. [DOI] [PubMed] [Google Scholar]
  • 272.Zhang T-T, Li W, Meng G, Wang P, Liao W. Strategies for transporting nanoparticles across the blood–brain barrier. Biomater Sci. 2016;4(2):219‐229. [DOI] [PubMed] [Google Scholar]
  • 273.Hsu JF, Chu SM, Liao CC, Wang CJ, Wang YS, Lai MY, et al. Nanotechnology and nanocarrier-based drug delivery as the potential therapeutic strategy for glioblastoma multiforme: an update. Cancers (Basel). 2021;13(2):1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Torchilin VP. Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery. Nat Rev Drug Discov. 2014;13(11):813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Qin C, Zhang C, Zhu F, Xu F, Chen SY, Zhang P, et al. Therapeutic target database update 2014: a resource for targeted therapeutics. Nucl Acids Res. 2013;42(D1):D1118‐D1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Liu R, Liang S, Jiang C, Wang X, Gong Y, Li P, et al. Paramagnetic, pH and temperature-sensitive polymeric particles for anticancer drug delivery and brain tumor magnetic resonance imaging. RSC Adv. 2015;5(106):87512‐87520. [Google Scholar]
  • 277.He H, David A, Chertok B, Cole A, Lee K, Zhang J, et al. Magnetic nanoparticles for tumor imaging and therapy: a so-called theranostic system. Pharm Res. 2013;30(10):2445‐2458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Cheng R, Meng F, Deng C, Klok H-A, Zhong Z. Dual and multi-stimuli responsive polymeric nanoparticles for programmed site-specific drug delivery. Biomaterials. 2013;34(14):3647‐3657. [DOI] [PubMed] [Google Scholar]
  • 279.Wilhelm I, Krizbai IA. In vitro models of the blood-brain barrier for the study of drug delivery to the brain. Mol Pharm. 2014;11(7):1949‐1963. [DOI] [PubMed] [Google Scholar]
  • 280.Ferraris C, Cavalli R, Panciani PP, Battaglia L. Overcoming the blood-brain barrier: successes and challenges in developing nanoparticle-mediated drug delivery systems for the treatment of brain tumours. Int J Nanomed. 2020;15:2999‐3022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Abbott NJ, Rönnbäck L, Hansson E. Astrocyte-endothelial interactions at the blood-brain barrier. Nat Rev Neurosci. 2006;7(1):41‐53. [DOI] [PubMed] [Google Scholar]
  • 282.Peterson JK, Houghton PJ. Integrating pharmacology and in vivo cancer models in preclinical and clinical drug development. Eur J Cancer. 2004;40(6):837‐844. [DOI] [PubMed] [Google Scholar]
  • 283.Huszthy PC, Daphu I, Niclou SP, Stieber D, Nigro JM, Sakariassen P, et al. In vivo models of primary brain tumors: pitfalls and perspectives. Neuro Oncol. 2012;14(8):979‐993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Kerbel RS. What is the optimal rodent model for anti-tumor drug testing? Cancer Metastasis Rev. 1998;17(3):301‐304. [DOI] [PubMed] [Google Scholar]
  • 285.Aldape K, Brindle KM, Chesler L, Chopra R, Gajjar A, Gilbert MR, et al. Challenges to curing primary brain tumours. Nat Rev Clin Oncol. 2019;16(8):509‐520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286.Lichtor T. Molecular Considerations and Evolving Surgical Management Issues in the Treatment of Patients with a Brain Tumor: BoD–Books on Demand; 2015.
  • 287.Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016;131(6):803‐820. [DOI] [PubMed] [Google Scholar]
  • 288.Stupp R, Hegi ME, Gorlia T, Erridge SC, Perry J, Hong YK, et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071-22072 study): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014;15(10):1100‐1108. [DOI] [PubMed] [Google Scholar]
  • 289.Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987‐996. [DOI] [PubMed] [Google Scholar]
  • 290.Kurian KM, Jenkinson MD, Brennan PM, Grant R, Jefferies S, Rooney AG, et al. Brain tumor research in the United Kingdom: current perspective and future challenges. A strategy document from the NCRI Brain Tumor CSG. Neurooncol Pract. 2018;5(1):10‐17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Schmidt-Hansen M, Berendse S, Hamilton W. Symptomatic diagnosis of cancer of the brain and central nervous system in primary care: a systematic review. Fam Pract. 2015;32(6):618‐623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Brown TJ, Brennan MC, Li M, Church EW, Brandmeir NJ, Rakszawski KL, et al. Association of the extent of resection with survival in glioblastoma: a systematic review and meta-analysis. JAMA Oncol. 2016;2(11):1460‐1469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 293.Kim D, Kim J, Park YI, Lee N, Hyeon T. Recent development of inorganic nanoparticles for biomedical imaging. ACS Cent Sci. 2018;4(3):324‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Scioli Montoto S, Muraca G, Ruiz ME. Solid lipid nanoparticles for drug delivery: pharmacological and biopharmaceutical aspects. Front Mol Biosci. 2020;7:587997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Duong VA, Nguyen TT, Maeng HJ. Preparation of solid lipid nanoparticles and nanostructured lipid carriers for drug delivery and the effects of preparation parameters of solvent injection method. Molecules. 2020;25(20):1-36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Essa ML, El-Kemary MA, Ebrahem Saied EM, Leporatti S, Nemany Hanafy NA. Nano targeted therapies made of lipids and polymers have promising strategy for the treatment of lung cancer. Materials (Basel). 2020;13(23):1-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297.Ma Q, Zhao Y, Guan Q, Zhao Y, Zhang H, Ding Z, et al. Amphiphilic block polymer-based self-assembly of high payload nanoparticles for efficient combinatorial chemo-photodynamic therapy. Drug Deliv. 2020;27(1):1656‐1666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Pillay NS, Daniels A, Singh M. Folate-targeted transgenic activity of dendrimer functionalized selenium nanoparticles in vitro. Int J Mol Sci. 2020;21(19):1-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Pooja D, Srinivasa Reddy T, Kulhari H, Kadari A, Adams DJ, Bansal V, et al. N-acetyl-d-glucosamine-conjugated PAMAM dendrimers as dual receptor-targeting nanocarriers for anticancer drug delivery. Eur J Pharm Biopharm. 2020;154:377‐386. [DOI] [PubMed] [Google Scholar]
  • 300.Oerlemans C, Bult W, Bos M, Storm G, Nijsen JF, Hennink WE. Polymeric micelles in anticancer therapy: targeting, imaging and triggered release. Pharm Res. 2010;27(12):2569‐2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Shalek AK, Satija R, Adiconis X, Gertner RS, Gaublomme JT, Raychowdhury R, et al. Single-cell transcriptomics reveals bimodality in expression and splicing in immune cells. Nature. 2013;498(7453):236‐240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302.Darban S A, Nikoofal-Sahlabadi S, Amiri N, Kiamanesh N, Mehrabian A, Zendehbad B, et al. Targeting the leptin receptor: to evaluate therapeutic efficacy and anti-tumor effects of Doxil, in vitro and in vivo in mice bearing C26 colon carcinoma tumor. Colloids Surf B Biointerfaces. 2018;164:107‐115. [DOI] [PubMed] [Google Scholar]

Articles from Technology in Cancer Research & Treatment are provided here courtesy of SAGE Publications

RESOURCES