
Nanoparticle‑based delivery systems for targeted therapy in brain tumors: Progress, challenges and perspectives (Review)
- Authors:
- Published online on: August 19, 2025 https://doi.org/10.3892/ijo.2025.5789
- Article Number: 83
-
Copyright: © Si et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Brain tumors, especially those originating in the central nervous system (CNS) such as gliomas, rank among the most aggressive and lethal forms of cancer. Gliomas are typically derived from neural stem cells, progenitor cells or dedifferentiated mature neural cells, and are characterized by a poor 5-year survival rate of <30% (1,2). The poor survival rate associated with brain tumors can be ascribed to the difficulty of an early diagnosis (3) and the lack of standard treatment regimens (4).
A major obstacle in brain tumor therapy is the presence of physiological barriers, most notably, the blood-brain barrier (BBB) and the blood-brain tumor barrier, that severely limit the delivery of therapeutic agents to intracranial tumors (5,6). These barriers restrict the permeability of most chemotherapeutic agents and are compounded by active efflux mechanisms and insufficient tumor specificity (7,8). As a result, conventional treatment strategies often fail to deliver adequate drug concentrations to the tumor site, diminishing therapeutic efficacy and increasing the risk of systemic toxicity (8). To address these challenges, increasing attention has been directed toward the development of drug delivery systems capable of traversing the BBB and achieving site-specific accumulation within tumor tissues. In this context, nanomedicine has emerged as a particularly promising approach. Among its various applications, nanoparticle (NP)-based delivery systems stand out for their ability to enhance the pharmacokinetic and pharmacodynamic profiles of therapeutic agents (9,10). Rapid progress in nanotechnology has led to the design and optimization of various nanocarriers, such as liposomes, solid lipid NPs (SLNs), dendrimers, polymeric micelles and inorganic NPs, that are engineered with specific physicochemical properties, including particle size, surface charge and ligand functionalization, to enhance biological compatibility and targeting efficiency (9). These tailored systems have demonstrated the ability to across the BBB, accumulate selectively in tumor tissues and facilitate efficient cellular uptake; thereby, enabling localized and sustained therapeutic effects. In neuro-oncology, NP-based platforms have shown strong potential for selective tumor accumulation and localized drug release, reducing off-target effects and enhancing treatment efficacy (10).
Unlike a number of existing reviews that separately discuss drug (8,9,11) or gene (12,13) delivery systems, the present review systematically integrates both therapeutic modalities. This comprehensive synthesis provides a holistic view of how different nanocarriers, ranging from liposomes to dendrimers and inorganic systems, are being engineered to deliver not only chemotherapeutics but also genetic payloads [such as small interfering RNA (siRNA), short hairpin RNA (shRNA) and micro RNA (miRNA)] across the BBB. Furthermore, the present review also adopts a material-based classification (lipid-based, dendritic, polymeric and inorganic nanocarriers) rather than a payload-based or mechanism-based classification. This structure allows readers to directly compare and contrast the physicochemical properties, targeting strategies and translational potential of each nanocarrier platform. To address the gap between bench and bedside, a new section titled 'Clinical challenges in translating NP-based therapies for brain tumors' is also introduced, where the limitations in immunogenicity, scalability, model relevance and human BBB heterogeneity (critical considerations that are often overlooked in reviews focused solely on laboratory advancements) are discussed (14-18). To support conceptual understanding, new schematic diagrams illustrating the BBB structure, transport mechanisms and NP-cell interactions are presented, which distinguish the present review from more text-heavy reviews. We consider that these additions and the integrative approach significantly enhance the value of the present review and differentiate it from prior literature in the field.
Structural features and transport mechanisms of the BBB
The BBB is a dynamic and highly selective interface that separates the systemic circulation from the CNS. The BBB plays a pivotal role in maintaining neural homeostasis by strictly regulating the entry of ions, nutrients and xenobiotics into the brain parenchyma (5). However, this protective function also presents a challenge for the delivery of therapeutics to brain tumors, including glioblastoma multiforme (GBM), which requires sustained and targeted drug accumulation in the tumor microenvironment (6).
Structural and functional composition of the BBB
The BBB is composed primarily of a monolayer of non-fenestrated endothelial cells that are tightly sealed by continuous tight junctions (TJs). These endothelial cells function within the context of the neurovascular unit, which includes pericytes embedded within the basement membrane, astrocyte end-foot processes that ensheath >90% of the vascular surface and the surrounding extracellular matrix components, such as collagen and laminin (Fig. 1A) (19). Together, these elements ensure integrity, selectivity and homeostatic regulation of the brain microenvironment.
BBB endothelial cells possess a unique set of functional characteristics that distinguish them from cells in the peripheral vasculature. A key feature of BBB endothelial cells is their exceptionally high transendothelial electrical resistance, often exceeding 1,000-2,000 Ω cm2 in vivo, which effectively limits paracellular diffusion of water-soluble and charged molecules (20,21). Moreover, these cells exhibit markedly low rates of pinocytosis and transcytosis; thereby, restricting non-specific vesicular transport and preserving the selectivity of the barrier (22). Notably, the BBB possesses an active drug efflux system, predominantly mediated by adenosine triphosphate-binding cassette transporters, which effectively transport a wide variety of endogenous metabolites and exogenous compounds, such as a number of chemotherapeutic agents, back into the bloodstream (23).
At the molecular level, the restrictive nature of the BBB is governed by the TJ complexes located at the apical lateral membrane between adjacent endothelial cells. These complexes are composed of integral membrane proteins, such as claudins (particularly claudin-5), occludin and junctional adhesion molecules. These proteins are anchored intracellularly by cytoplasmic scaffold proteins that include zonula occludens-1 (ZO-1), ZO-2 and cingulin (Fig. 1B) (24). These structures not only maintain the structural cohesion of the endothelial monolayer but also dynamically regulate paracellular permeability in response to physiological or pathological stimuli.
Collectively, these structural and functional features make the BBB an extraordinarily effective yet formidable obstacle to therapeutic delivery, especially in the treatment of brain tumors. Therefore, any strategy that enhances drug penetration into the brain must be designed with a comprehensive understanding of this intricate barrier system.
Main transport pathways across the BBB
Despite its restrictive nature, the BBB allows specific translocation of molecules through several tightly regulated pathways (Fig. 1C) (19). Among the various mechanisms, the paracellular route refers to the movement of hydrophilic molecules between adjacent endothelial cells. However, this pathway is effectively sealed in the BBB due to the presence of TJs, which consist of transmembrane proteins (such as claudins, occludin and junctional adhesion molecules) connected to the cytoskeleton via adaptor proteins, such as ZO-1 and ZO-2 (25). Unless TJ modulators or novel nanotechnologies are used, most drugs fail to achieve effective brain exposure via paracellular diffusion (26). However, even when TJs are transiently loosened through pharmacological or physical means to enhance permeability, the risks of drug-induced toxicity and barrier disruption persist and limit the clinical applicability of the paracellular pathway (27,28). Therefore, the paracellular aqueous pathway contributes minimally to drug penetration and remains a formidable hurdle in brain tumor therapy.
Unlike the paracellular pathway, the transcellular lipophilic route allows small, lipid-soluble compounds to diffuse directly through endothelial cell membranes. This route is exploited by endogenous molecules, such as oxygen, carbon dioxide and certain hormones, as well as small-molecule drugs, such as some anesthetics. However, successful diffusion requires favorable physicochemical characteristics; typically, low molecular weight, low hydrogen-bonding potential and a relatively high logP value for the lipid-water partition coefficient (29-31). Numerous anticancer drugs and alkylating agents, including doxorubicin (DOX) or temozolomide (32) and monoclonal antibodies (33), do not meet these criteria, rendering passive diffusion across the BBB inefficient. Moreover, even lipophilic drugs are often subject to rapid efflux via transporters, such as P-glycoprotein, further limiting their accumulation in the brain (34).
Receptor-mediated transcytosis (RMT) is one of the most promising routes for NP-based delivery across the BBB due to its high specificity, affinity and potential for receptor recycling. This mechanism involves ligand binding to specific receptors on the luminal surface of endothelial cells, followed by clathrin-mediated endocytosis and exocytosis into the brain parenchyma (11,35). Numerous preclinical studies have shown that NPs conjugated with ligands, such as transferrin, lactoferrin, angiopep-2 or monoclonal antibodies targeting the transferrin receptor or the insulin receptor, achieve significantly enhanced brain penetration (36-38). These ligands not only facilitate BBB transcytosis but also enable subsequent tumor targeting, making RMT an attractive platform for theranostic applications in glioblastoma and other CNS malignancies.
Adsorptive-mediated transcytosis (AMT) relies on electrostatic interactions between the negatively-charged luminal surface of endothelial cells and positively-charged ligands or carriers (39). This non-specific mechanism is particularly useful to enhance the transport of cationic proteins (40), peptides (41) and NPs (42) across the BBB. Cell-penetrating peptides, such as trans-activator of transcription (TAT), penetratin and rabies virus glycoprotein 29, have been widely used to facilitate AMT-mediated delivery of both small molecules and macromolecules (43-45). Although AMT is less selective than RMT, it has the advantage of delivering diverse payloads independent of specific receptor expression, which expands its applicability across different brain tumor subtypes. However, the use of strong cationic charges must be balanced against the risk of cytotoxicity and non-specific distribution.
Solute carrier transporters constitute a broad family of influx proteins at the BBB that enable the uptake of essential nutrients, metabolites and ions into the brain (46). These transporters include glucose transporter 1, which facilitates glucose entry (47), L-type amino acid transporter 1, which is responsible for transporting large neutral amino acids (48), and monocarboxylate transporters, which mediate the influx of lactate and other monocarboxylates (49). Notably, these transporters can be harnessed for brain-targeted drug delivery by chemically modifying drugs or nanocarriers to resemble endogenous substrates. For example, glucose-conjugated or amino acid-decorated NPs have shown promise in facilitating carrier-mediated transport across the BBB (50). Such approaches offer a non-invasive, receptor-independent route to enhance the delivery of small-molecule therapeutics or genetic materials into brain tumor tissues, making solute carrier-mediated influx an attractive mechanism for developing effective brain-targeted therapies.
An in-depth understanding of these transport routes is critical for designing effective nanocarrier platforms for brain tumor therapy. Functionalization with targeting ligands (such as angiopep-2 and transferrin) enables RMT-based entry, while surface charge modulation enhances AMT. Dual-targeting strategies that combine BBB penetration with tumor specificity are also emerging as effective approaches for glioma therapy.
NP-based drug delivery in brain tumors
Advancements in nanotechnology have opened new avenues to overcome the notable challenges posed by the BBB and the complex microenvironment of brain tumors. In particular, NP-based delivery systems offer versatile platforms capable of transporting a wide array of therapeutic agents across the BBB, including small-molecule drugs, nucleic acids and gene-editing constructs, with enhanced precision, stability and efficacy. Over the past decade, a diverse spectrum of nanocarriers has been engineered to meet the distinct physicochemical and biological requirements of CNS drug delivery. These include lipid-based carriers, such as liposomes and SLNs (51,52), polymeric NPs (50) constructed from biodegradable materials, such as poly(lactic-co-glycolic acid) (PLGA) (53) or polyethylene glycol (PEG) (50), dendrimers with highly-branched architectures (54) and inorganic systems, such as gold NPs (AuNPs) (55), graphene oxide (GO) (56) and magnetic iron oxide NPs (IONPs) (57). These platforms possess unique advantages in drug loading capacity, targeting capabilities, biocompatibility and multifunctionality, making them suitable not only for conventional chemotherapy but also for gene silencing, messenger RNA replacement and immunomodulation. Table I (50-69) summarizes representative NP-based delivery systems applied to brain tumor therapy, highlighting their physicochemical characteristics (such as particle size and zeta potential), payloads, targeting strategies and therapeutic outcomes. Furthermore, given the formidable barrier posed by the BBB, a variety of surface modification approaches have been developed to enhance nanocarrier penetration and targeting efficiency. These approaches, including ligand conjugation, cell-penetrating peptides and stimuli-responsive coatings, are summarized in Table II (7,70-86).
The present review also provides a holistic view of how different nanocarriers, ranging from liposomes to dendrimers and inorganic systems, are being engineered to deliver not only chemotherapeutics but also genetic payloads (such as siRNA, shRNA and miRNA) across the BBB. In the following sections, these nanocarrier classes and their applications in delivering both chemical and genetic therapeutics to brain tumors are reviewed, with an emphasis on structural design, delivery mechanisms and preclinical outcomes.
Lipid-based nanocarriers
Lipid-based nanocarriers, including liposomes, SLNs and lipid-polymer hybrid NPs (LPHNs), represent some of the most well-established and clinically relevant platforms for brain-targeted drug and gene delivery. These carriers are biocompatible, structurally versatile and easily modifiable, making them ideal for encapsulating hydrophilic and lipophilic drugs, as well as genetic materials, such as siRNA, miRNA and plasmid DNA (51,62).
Liposomes
Liposomes are spherical vesicles composed of lipid bilayers that can be modified with targeting ligands to enhance BBB penetration and tumor specificity. Human interleukin-13 (IL-13) is a cytokine released by activated T cells and can induce proinflammatory and anti-inflammatory immune responses (87). Reports have indicated that IL-13 receptor α2 (IL-13Rα2) is found at high levels in brain tumors, such as GBM and pilocytic astrocytomas, but not in normal tissues (88,89). Therefore, IL-13Rα2 is a promising target to deliver cytotoxic drugs to various aggressive brain tumors. Madhankumar et al (90) developed IL-13-conjugated liposomes to deliver DOX specifically to brain tumors, which led to less cytotoxicity and greater accumulation and retention of DOX in the glioma cells compared with therapy without these liposomes. Mice receiving intraperitoneal injections of IL-13-conjugated liposomes carrying DOX showed a significant decrease in U251 glioma growth (90). In addition to IL-13, numerous other ligands promote the brain tumor targeting ability of liposomes. Zong et al (63) designed a dual-targeting liposomal system modified with TAT (AYGRKKRRQRRR) and T7 (HAIYPRH) peptides. The specific ligand, T7, targets the BBB and brain tumors, while the non-specific ligand, TAT, enhances passage through the BBB and increases tumor penetration. Intravenous administration of dual-targeted DOX liposomes led to enhanced delivery of DOX to brain glioma tissue in a C6 glioma mouse model. Furthermore, Cen et al (58) developed a DOX-loaded liposomal system modified with the SS31 peptide, which exhibits dual-targeting capabilities by facilitating both BBB penetration and mitochondrial targeting within glioma cells. Liposomal system-DOX liposomes accumulated effectively in tumor tissues, significantly inhibiting tumor growth and extending survival rates without notable toxicity. This dual-targeting approach offers a promising strategy to enhance the therapeutic efficacy of liposomal DOX in glioma treatment.
In gene delivery, cationic liposomes have enabled the systemic administration of siRNAs and miRNAs to brain tumors. For instance, Wei et al (91) performed a layer-by-layer assembly of protamine/chondroitin sulfate/siRNA/cationic liposomes followed by T7 peptide modification to create a targeted siRNA-epidermal growth factor receptor (EGFR) delivery system. This modified NP specifically accumulated anti-EGFR siRNA in brain tumor tissues upon intravenous administration, leading to downregulation of EGFR expression and high survival rates in a U87 mouse glioma model.
Cyclophilin A (CypA) is a tumor marker that is elevated in various cancer types, including liver, brain and lung cancer (92-94). CypA plays a crucial role in epithelial to mesenchymal transition and cancer metastasis (94) and can be used as a target for tumor therapy. Saw et al (62) developed a liposome-based extra domain B targeting nanoplatform for CypA siRNA delivery and glioblastoma therapy. After systemic administration, these new siRNA delivery NPs targeted glioma cells and effectively inhibited glioblastoma tumor growth by silencing CypA, which plays a crucial role in the malignant transformation of brain cancer and the maintenance of glioma cell stemness (62). These findings indicate that this RNA interference NP platform is a potentially effective tool for targeted brain tumor therapy.
The biopharmaceutical challenges related to the therapeutic use of non-coding RNAs could potentially be addressed by lipid NPs (95). The therapeutic efficacy of non-coding RNAs delivered by lipid NPs has been confirmed in different forms of cancer (96).
SLNs
SLNs are submicron colloidal carriers composed of biocompatible solid lipids stabilized by surfactants. Compared with conventional liposomes, SLNs offer superior physicochemical stability, lower burst release and improved control over drug release kinetics (97). SLNs are capable of encapsulating both hydrophilic and lipophilic agents and are amenable to various administration routes, including intravenous and oral delivery (52,54).
SLNs have shown notable success in enhancing brain delivery of chemotherapeutics. For example, edelfosine-loaded SLNs administered orally to glioma-bearing mice crossed the BBB efficiently and accumulated within tumor tissues, leading to a significant reduction in tumor volume and prolonged survival compared with non-SLN therapy (52). Beyond synthetic drugs, SLNs have been used to deliver poorly-soluble natural products with anticancer properties. Rutin, a flavonol glycoside with pro-apoptotic and anti-proliferative effects, has low water solubility and poor oral bioavailability (98). Encapsulation within SLNs improves the BBB permeability and bioavailability of rutin, resulting in enhanced brain accumulation and tumor suppression in preclinical models (54,99). Further innovations include surface-modified SLNs with targeting ligands, such as transferrin or apolipoprotein E (ApoE) mimetics, which exploit RMT pathways to improve brain specificity (100). Additionally, SLNs have been used for co-delivery systems, such as the simultaneous encapsulation of paclitaxel (PTX) and P-glycoprotein inhibitors, to overcome drug efflux mechanisms in glioma cells, with less cytotoxicity and enhanced therapeutic outcomes (101).
LPHNs
LPHNs combine the structural stability of polymeric cores with the membrane fluidity and biocompatibility of lipid shells; thereby, bridging the functional advantages of both systems. These hybrid carriers are particularly well-suited for gene delivery due to their high encapsulation efficiency, low cytotoxicity and the ability to promote endosomal escape and intracellular release of nucleic acid cargos (102).
One of the most promising hybrid platforms is the self-assembled lipid-coated calcium phosphate NP (SANP). These carriers typically consist of a calcium phosphate core complexed with genetic material that is coated with PEGylated cationic lipids, such as N-[1-(2,3-dioleoyloxy) propyl]-N,N,N-trimethylammonium and modified lipids, such as ceramide-PEG2000. In glioma models, SANPs have demonstrated high miRNA and shRNA delivery efficiency, with significant accumulation in tumor tissue and potent gene silencing effects in vivo (51,103). As with miRNA therapy, SANPs have been used for co-delivery of nucleic acids and chemotherapeutics, such as bisphosphonates or DOX (104). The lipid shell allows prolonged systemic circulation and BBB traversal, while the calcium phosphate core promotes endosomal rupture via a 'proton sponge' effect, enhancing cytoplasmic release. For instance, a recent study incorporated miRNA-124 and PTX into SANPs to simultaneously silence oncogenic pathways and induce apoptosis, achieving synergistic antitumor efficacy with minimal systemic toxicity (105).
Further optimization strategies include the use of targeted peptides (such as arginyl-glycyl-aspartic peptides and T7) on the NP surface to increase glioma-specific uptake, as well as pH-responsive lipids that enable controlled gene release in the acidic tumor microenvironment (63).
Dendrimers
Dendrimers are highly-branched, monodisperse and nanoscale macromolecules that offer exceptional versatility for drug and gene delivery in brain tumors. Their well-defined architecture, multivalent surface and internal cavities allow for the simultaneous loading of therapeutic agents and targeting ligands (106). Among dendrimers, poly(amidoamine) (PAMAM) dendrimers have been the most extensively studied due to their biocompatibility, tunable size and ease of functionalization. These features enable dendrimers to serve as flexible nanocarriers for drug molecules, genetic materials and multifunctional payloads (107).
In drug delivery, dendrimers function either as nanocapsules to encapsulate hydrophobic agents or as nanoconjugates bearing covalently attached drugs via cleavable linkers. PAMAM dendrimers have shown particular promise in brain tumor models when engineered with dual-targeting moieties. For instance, dendrimers co-modified with angiopep-2 and an EGFR-binding peptide exhibited enhanced BBB permeability and glioma-targeting ability via low-density lipoprotein receptor-related protein 1 (LRP1)- and EGFR-mediated transcytosis. This platform significantly improved the delivery of DOX, resulting in prolonged survival and reduced systemic toxicity in glioma-bearing mice compared with therapy without this platform (61). Similarly, dendrimers functionalized with transferrin and wheat germ agglutinin achieved dual-site targeting, facilitating enhanced uptake into glioma cells and accumulation within tumor tissues; thereby, improving therapeutic outcomes after systemic administration of DOX (59).
Dendrimers also provide a promising scaffold for gene delivery owing to their cationic surface, which facilitates complexation with nucleic acids. PAMAM dendrimers have been used to deliver a range of genetic payloads, including siRNA, shRNA and plasmid DNA, for gene silencing and transcriptional modulation in brain tumors (108). Surface modification with PEG or β-cyclodextrin improves stability and biocompatibility while reducing immunogenicity (109). By tuning the surface charge and size, these dendrimer complexes can traverse the BBB and achieve transgene expression within tumor cells. Notably, dendrimers support intracellular delivery via endosomal escape mechanisms and avoid rapid degradation of nucleic acids in circulation (110).
Beyond direct tumor cell killing, dendrimers have demonstrated potential in modulating the tumor immune microenvironment. Hydroxyl-terminated PAMAM dendrimers have been shown to selectively accumulate in tumor-associated macrophages and activated microglia, particularly in glioma tissues with compromised BBB integrity (60,111-113). A notable example is the targeted delivery of triptolide, a potent immunomodulatory agent, via dendrimer conjugation. This formulation not only enhanced tumor localization but also reprogrammed tumor-associated macrophages from an M2-like immunosuppressive phenotype to an M1-like pro-inflammatory state, resulting in both reduced tumor burden and minimized systemic toxicity in glioblastoma (65).
The multivalency of dendrimers allows them to be co-loaded with chemotherapeutic agents and nucleic acids, facilitating combination therapies in a single nanoplatform. For example, hybrid systems have been developed to simultaneously deliver DOX and siRNA, thereby achieving synergistic effects through cytotoxicity and gene silencing (114). Such designs enable coordinated regulation of tumor proliferation and resistance mechanisms, providing a rational strategy for multifaceted glioma therapy.
Polymeric NPs
Polymeric NPs have garnered notable interest for both drug and gene delivery in brain tumor therapy due to their excellent biocompatibility, structural flexibility and tunable physicochemical properties (115). Typically synthesized from US Food and Drug Administration-approved polymers, such as PLGA, PEG, polycaprolactone and polyethylenimine (PEI), these systems allow for fine control of particle size, surface charge, release kinetics and functional modification (50,56,67).
PLGA-based NPs are widely used to deliver chemotherapeutic agents across the BBB. For example, PTX-loaded PLGA NPs coated with poloxamer 188 significantly increased BBB penetration and PTX accumulation in glioblastoma tissues, outperforming free drug administration in a rat GBM model (56). Co-delivery systems based on PLGA and chitosan have also been developed to encapsulate carmustine and O6-benzylguanine. This combination synergistically depleted O6-methylguanine-DNA methyltransferase, sensitizing tumors to alkylating agents and improving survival in F98 glioma-bearing rats (116). Targeted polymeric micelles further enhance brain accumulation. For instance, 2-deoxy-D-glucose-modified poly(ethylene glycol)-co-poly(trimethylene carbonate) PTX NPs were developed as a potential dual-target drug delivery system that can enhance BBB penetration through glucose transporter (GLUT)-mediated cross-cellular action and improve drug accumulation in gliomas through GLUT-mediated endocytosis. These glucose-decorated NPs increased drug accumulation in the brain and led to improved survival rates in an RG2 mouse glioma model, compared with plain NPs and PTX (50). Similarly, transferrin receptor-targeted micelles carrying PTX showed prolonged tumor retention and increased therapeutic efficacy in U87 MG intracranial gliomas (117). Other advanced polymeric formulations include DOX-loaded polybutylcyanoacrylate NPs and methoxy-PE G-poly(ε-caprolactone) micelles, which have demonstrated superior tumor accumulation, reduced systemic toxicity and enhanced survival in C6 glioma models (69,118).
Polymeric nanocarriers also support efficient gene transfer across the BBB. One example is cationic albumin-conjugated PEG NPs loaded with plasmid-encoding tumor necrosis factor-related apoptosis-inducing ligand. These particles have been shown to cross the BBB via AMT and induce selective apoptosis in glioma cells following systemic administration, with strong antitumor effects in vivo (66). DNA-loaded NPs formulated with an inclusion of densely PEGylated PEI (that is, brain-penetrating NPs) were recently reported to provide widespread transgene expression in the rat brain while exhibiting good in vivo safety profiles (119). Additionally, Negron et al (67) developed polymer-based DNA-loaded NPs with small particle diameters (~50 nm) and non-adhesive surface PEG coatings, which efficiently penetrated both brain tumor tissue and healthy brain parenchyma. These brain-penetrating NPs facilitated widespread transgene expression in heathy rodent striatum and aggressive brain tumor tissue through intracranial administration. These systems efficiently delivered DNA and siRNA while minimizing neuroinflammation and cytotoxicity.
In summary, polymeric NPs offer an adaptable platform to deliver a broad spectrum of therapeutic agents, including cytotoxic drugs, plasmid DNA and siRNA, across the BBB. Their ability to incorporate targeting ligands, modulate release kinetics and achieve sustained transgene expression makes polymeric NPs invaluable in both standalone and combination therapy strategies for malignant brain tumors.
Inorganic NPs
Inorganic NPs offer distinct physicochemical advantages, such as precise size control, surface reactivity, magnetic or optical responsiveness and structural rigidity, which makes them highly valuable in brain tumor therapy (120). Unlike organic materials, inorganic carriers can integrate diagnostic and therapeutic functions within a single platform (that is, theranostics) (121). This section focuses on three prominent classes: AuNPs, graphene-based nanocarriers and IONPs, detailing their respective applications in both drug and gene delivery.
AuNPs
AuNPs have been used to enhance drug stability, targeting and controlled release. For example, self-assembled gold nanospheres (~80 nm) have been designed to remain stable in circulation but disassemble in response to acidic and reductive conditions in the glioma microenvironment. This responsiveness improves intratumoral accumulation while minimizing off-target toxicity through renal clearance of dissociated particles (64). In targeted chemotherapy, antibody-conjugated gold-silica nanoshells directed against IL-13Rα2, which is upregulated in glioblastoma, enabled precise tumor cell recognition and thermal ablation. Upon near-infrared irradiation, these nanoshells induced cell death in U373 and U87 glioma models (122). Moreover, Feng et al (64) described a practical method for assembling AuNPs into ~80 nm nanospheres for use as a drug delivery platform. This platform not only enhanced drug retention in brain tumors but was able to dynamically switch to the single formulation for excretion. These self-assembled AuNPs can target brain tumor cells and respond to tumor microenvironmental parameters, including high vascular permeability and acidic and reductive conditions, suggesting that gold nanoassemblies may be an effective targeting strategy for brain tumor treatment (123).
AuNPs are also efficient vehicles for genetic payloads due to their high surface area and ease of nucleic acid conjugation. For instance, β-cyclodextrin-modified dendrimer-entrapped AuNPs have been used to co-deliver siRNAs targeting Bcl-2 and vascular endothelial growth factor. These carriers achieved robust gene silencing in glioma cells with minimal cytotoxicity and good serum stability (124). Another notable approach involves spherical nucleic acids, in which oligonucleotides are densely arranged on AuNP cores. When encapsulated in ApoE-modified liposomes, these spherical nucleic acids accumulate in glioma tissues and facilitate efficient miRNA modulation, opening avenues for post-transcriptional gene regulation (53).
Graphene-based nanocarriers
GO sheets, with their large surface area and π-π interaction capability, are excellent carriers for chemotherapeutics. Angiopep-2-modified GO loaded with DOX was shown to enhance uptake by LRP1-upregulated glioma cells, achieving greater cytotoxicity than that with free DOX or unmodified GO formulations (57). In a dual-modality platform, mesoporous silica-coated GO nanosheets (GSPI) were functionalized with targeting peptide and loaded with DOX (GPSID). The resulting GSPID system combined chemo- and photothermal therapy. Upon near-infrared laser exposure, the GSPID system induced significant glioma cell death through thermal enhancement of drug activity (68).
Although limited compared with drug delivery, GO has also been used to deliver nucleic acids. The surface of GO enables electrostatic complexation with negatively-charged RNA or DNA (125). Modified GO-PEI hybrids can deliver shRNA into glioma cells with good transfection efficiency, especially when further engineered for nuclear targeting or stimuli-responsive release. For example, co-delivery of irinotecan and shRNA targeting stomatin-like protein 2 (SLP2) using magnetic GO hybrid nanocarriers improved glioma treatment efficacy by downregulating oncogenic pathways and enhancing chemotherapeutic synergy (126).
IONPs
IONPs exhibit superparamagnetic properties, allowing external magnetic fields to enhance tumor-specific delivery across the BBB. For example, DOX-loaded trimethoxysilylpropyl-ethylenediamine triacetic acid-stabilized IONPs, when co-administered with a cadherin-binding peptide and magnetically-guided, significantly enhanced toxicity in glioma cells by temporarily loosening TJs at the BBB (127). Targeting was further improved using linTT1 peptide-functionalized IONPs, which bind to p32/gC1qR proteins upregulated in the glioma vasculature. These IONPs boosted glioma-specific uptake and promoted co-delivery of other NPs, such as albumin-PTX and silver nanocarriers (128). Another innovative application involved salinomycin-loaded IONPs, modified with PEG and PEI to improve circulation and BBB transit. These NPs selectively induced apoptosis in GBM cells via the generation of reactive oxygen species while sparing healthy tissue, making them a promising candidate for magnetically-targeted chemotherapy (129).
Although still at a preclinical stage, IONPs have been adapted for magnetofection, which is a technique that uses magnetic force to accelerate and direct nucleic acid delivery (130). PEI-coated IONPs complexed with plasmids or siRNA have been shown to transfect glioma cells both in vitro and in vivo under magnetic guidance (131). Co-delivery systems integrating siRNA and small molecules (such as cisplatin) are also under exploration, aiming to synergize gene silencing and DNA damage mechanisms.
Delivery strategies based on the protein corona (PC) in brain tumors
Although NPs as drug carriers hold great promise for brain tumor therapy, numerous challenges remain in clinical translation (132). The PC poses a significant biological challenge for the translation of targeted drug delivery systems from the laboratory to clinical use, greatly impacting the targeting accuracy and biodistribution (133). NPs entering a biological environment experience surface modification due to dynamic physicochemical interactions with proteins and other biomolecules. The surface becomes coated with plasma proteins, lipids and other molecules, forming a PC (134). The PC modifies the size and surface properties of NPs (135), which makes it difficult for NPs to arrive at target sites, including brain tumors (Fig. 2) (136,137). Therefore, PC-mediated targeting by maintaining the function of target plasma proteins on the NP surface may offer a new approach for specific drug delivery. The two most important types of proteins in the PC are opsonins and de-opsonins (138). Mononuclear macrophages can easily identify and clear NPs with surface opsonins, which greatly reduces drug delivery (139). De-opsonins show the opposite effect to that of opsonins as they prevent NP uptake by macrophages and prolong the NP circulation time. Albumin, the most abundant plasma protein, is a typical de-opsonin. Coating NPs with albumin significantly inhibits the adsorption of opsonin, reduces macrophage phagocytosis, decreases NP cytotoxicity and prolongs NP circulation in the blood (140).
PC-mediated targeting provides a novel approach to precise drug delivery by accurately controlling the interaction modes of functional plasma proteins on the surface of NPs. Zhang et al (141) developed bioinspired liposomes (SP-sLips) by modifying the liposomal surface with a short non-toxic peptide derived from amyloid β-protein (Aβ)1-42. This peptide specifically interacts with the lipid-binding domain of exchangeable apolipoproteins. SP-sLips absorb plasma apolipoproteins A1, E and J, and achieve brain-targeted delivery by exposing the receptor-binding domain of apolipoproteins. DOX-loaded SP-sLips have demonstrated a significant improvement in brain distribution and anti-brain cancer effects compared with DOX-loaded plain liposomes. Another study (142) fabricated PEG-polyethylene-polylactice acid nanomicelles (PMs) decorated by Aβ25-35 with an amide bond (Aβ-CN) to load PTX. Aβ-CN peptide was used to bind the C-terminal domain of ApoE when exposed to physiological conditions and specifically bound to the lipid-binding domain of ApoE in vivo, forming an ApoE-enriched PC surrounding Aβ-CN-PMs. This formation demonstrated enhanced anticancer effects and a favorable tissue distribution profile, with rapid accumulation in brain tumor tissues.
Clinical challenges in translating NP-based therapies for brain tumors
Despite the notable preclinical progress achieved using nanocarriers for drug and gene delivery in brain tumors, translating these systems into clinical practice remains a formidable task. Numerous NP platforms, ranging from liposomes and dendrimers to inorganic and hybrid systems, have demonstrated efficient BBB penetration, tumor targeting and therapeutic efficacy in animal models. However, only a handful have advanced to clinical trials (14,17) and none have become standard-of-care for glioma treatment. Several key clinical challenges hinder this transition: i) Safety, immunogenicity and long-term biocompatibility. Nanocarriers often exhibit unpredictable interactions with the immune system. For example, cationic surfaces can activate complement cascades or induce cytokine release, leading to hypersensitivity or off-target effects (18). Even biocompatible materials, such as PEG, may elicit anti-PEG antibodies after repeat administration (16), undermining therapeutic efficacy. Moreover, long-term accumulation of inorganic NPs (such as gold or iron oxide) in non-target tissues raises concerns about potential chronic toxicity. ii) Physiological complexity of the human BBB and tumor microenvironment. While most nanocarrier studies rely on rodent models, the human BBB is markedly more selective and the glioblastoma vasculature is heterogeneous and dynamically remodeled during disease progression (15). As a result, targeting strategies that succeed in preclinical models may underperform or fail in clinical settings. Additionally, the tumor microenvironment, including hypoxia, immune suppression and abnormal interstitial pressure, can impair nanocarrier penetration and distribution within brain tumors (143). iii) Reproducibility, scale-up and manufacturing constraints. The structural complexity of nanocarriers presents a major hurdle for consistent large-scale production. Batch-to-batch variability in size, surface charge and drug loading efficiency can affect pharmacokinetics and therapeutic outcomes. Moreover, regulatory agencies require detailed physicochemical characterization, stability profiling and Good Manufacturing Practice compliance, all of which remain difficult to achieve for sophisticated multifunctional systems. iv) Lack of predictive models for human translation. Currently, to the best of our knowledge, there is a lack of robust in vitro or in silico models that accurately predict nanocarrier behavior in the human brain. This limits the ability to assess clinical feasibility early in the developmental pipeline, resulting in high attrition rates during translation.
To bridge the gap between bench and bedside, future efforts must focus not only on optimizing the design and targeting efficiency of nanocarriers, but also on improving their biological stability, minimizing immunogenicity and enhancing reproducibility on a clinical scale. Among the most critical biological factors influencing in vivo behavior is the formation of a PC, a dynamic biomolecular layer that adsorbs onto NPs upon contact with biological fluids.
Conclusions and new perspectives
Innovative local chemotherapy using gene therapy-based NPs has been proposed as a treatment for patients with brain tumors. Zhang et al (130) modified the porous structure of IONPs by attaching carboxyl groups to enable the codelivery of siRNA targeting glutathione peroxidase 4 (si-GPX4) and cisplatin with high drug loading efficiencies. This nanoformulation exerted notable effects on glioblastoma cells, but the effects on normal human astrocytes were limited. During intracellular degradation, IONPs significantly increased iron (Fe2+ and Fe3+) levels, while cisplatin destroyed nuclear DNA and mitochondrial DNA, resulting in apoptosis. Additionally, the co-released si-GPX4 enhanced the therapeutic efficacy by promoting ferroptosis, suggesting that si-GPX4 may be a safe and effective inducer of ferroptosis and apoptosis for combined brain tumor therapy. Chuang et al (126) developed a co-delivery strategy for the chemotherapeutic drug, irinotecan (CPT-11), and SLP2 shRNA using dual targeting NPs by combining magnetic targeting with ligand-mediated active targeting for the treatment of brain tumors. The magnetic GO (mGO) nanocarrier was conjugated with chitosan and urocanic acid (mGOCU) for shRNA complexation and endosomal escape. After PEGylation by interaction of the hydrophobic tails of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[maleimide(polyethylene glycol)-2000] with the mGO surface followed by binding gastrin releasing peptide to PEG (mGOCUG), CPT-11 was loaded via π-π interaction and the SLP2 shRNA was complexed to form mGOCUG/CPT-11/shRNA. Intravenous administration of mGOCUG/CPT-11/shRNA exhibited excellent antitumor efficacy in orthotopic brain tumors, which presents a versatile mGO-based drug and gene delivery system for effective combination therapy in brain tumors.
Notably, efforts to incorporate multiple organic and inorganic nanomaterials as potential therapeutic agents have also been made. A theranostic liposome incorporating superparamagnetic IONPs, quantum dots and cilengitide has been developed for optical imaging and as a therapeutic drug delivery system (144). Wu et al (145) have also created an aqueous, surfactant-free ferrofluid composed of superparamagnetic IONPs coated with silicate mesolayers and carbon shells. It was found that this NP decreased the viability of brain tumors more than that of the respective primary analogues of the NP. Conjugated polymer NPs have been designed and synthesized by integrating a metal oxide magnetic core (Fe3O4 and NiFe2O4 NPs; 5 nm) into their matrix during the nanoprecipitation process (146). This incorporation may be used for simultaneous magnetic resonance and fluorescence imaging in brain tumors.
The effectiveness of NP-based delivery systems indicates the potential to develop more effective and personalized combination therapies for patients with brain tumors. However, despite these advancements, drug carriers with controllable size, integrated targeting function, appropriate structure and the ability to cross biological barriers must be developed and further optimized in the future.
Availability of data and materials
Not applicable.
Authors' contributions
JXS reviewed the literature and wrote the first draft. ZCL produced the figures. FG and XJ collected part of the data. YYM supervised and revised the manuscript. JXS, ZCL, FG, XJ and YYM were involved in writing the paper. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Acknowledgements
Not applicable.
Funding
This work was supported by the Medical Science and Technology Project of Zhejiang Province (grant nos. 2023KY465 and 2021KY529).
References
Miller KD, Ostrom QT, Kruchko C, Patil N, Tihan T, Cioffi G, Fuchs HE, Waite KA, Jemal A, Siegel RL and Barnholtz-Sloan JS: Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 71:381–406. 2021.PubMed/NCBI | |
Ostrom QT, Patil N, Cioffi G, Waite K, Kruchko C and Barnholtz-Sloan JS: CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the united states in 2013-2017. Neuro Oncol. 22(12 Suppl 2): iv1–iv96. 2020. View Article : Google Scholar : PubMed/NCBI | |
Sadad T, Rehman A, Munir A, Saba T, Tariq U, Ayesha N and Abbasi R: Brain tumor detection and multi-classification using advanced deep learning techniques. Microsc Res Tech. 84:1296–1308. 2021. View Article : Google Scholar : PubMed/NCBI | |
Tan AC, Ashley DM, Lopez GY, Malinzak M, Friedman HS and Khasraw M: Management of glioblastoma: State of the art and future directions. CA Cancer J Clin. 70:299–312. 2020.PubMed/NCBI | |
Arvanitis CD, Ferraro GB and Jain RK: The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 20:26–41. 2020. View Article : Google Scholar | |
Da Ros M, De Gregorio V, Iorio AL, Giunti L, Guidi M, de Martino M, Genitori L and Sardi I: Glioblastoma chemoresistance: The double play by microenvironment and Blood-brain barrier. Int J Mol Sci. 19:28792018. View Article : Google Scholar : PubMed/NCBI | |
Gao H, Yang Z, Zhang S, Cao S, Pang Z, Yang X and Jiang X: Glioma-homing peptide with a cell-penetrating effect for targeting delivery with enhanced glioma localization, penetration and suppression of glioma growth. J Control Release. 172:921–928. 2013. View Article : Google Scholar : PubMed/NCBI | |
Liu Y and Lu W: Recent advances in brain tumor-targeted nano-drug delivery systems. Expert Opin Drug Deliv. 9:671–686. 2012. View Article : Google Scholar : PubMed/NCBI | |
Han L and Jiang C: Evolution of blood-brain barrier in brain diseases and related systemic nanoscale brain-targeting drug delivery strategies. Acta Pharm Sin B. 11:2306–2325. 2021. View Article : Google Scholar : PubMed/NCBI | |
Jena L, McErlean E and McCarthy H: Delivery across the blood-brain barrier: Nanomedicine for glioblastoma multiforme. Drug Deliv Transl Res. 10:304–318. 2020. View Article : Google Scholar : | |
Hersh AM, Alomari S and Tyler BM: Crossing the blood-brain barrier: Advances in nanoparticle technology for drug delivery in Neuro-oncology. Int J Mol Sci. 23:41532022. View Article : Google Scholar : PubMed/NCBI | |
Chen J, Guo Z, Tian H and Chen X: Production and clinical development of nanoparticles for gene delivery. Mol Ther Methods Clin Dev. 3:160232016. View Article : Google Scholar : PubMed/NCBI | |
Huh MS, Lee EJ, Koo H, Yhee JY, Oh KS, Son S, Lee S, Kim SH, Kwon IC and Kim K: Polysaccharide-based nanoparticles for gene delivery. Top Curr Chem (Cham). 375:312017. View Article : Google Scholar : PubMed/NCBI | |
Arami H, Patel CB, Madsen SJ, Dickinson PJ, Davis RM, Zeng Y, Sturges BK, Woolard KD, Habte FG, Akin D, et al: Nanomedicine for spontaneous brain tumors: A companion clinical trial. ACS Nano. 13:2858–2869. 2019. View Article : Google Scholar : PubMed/NCBI | |
Hahn A, Bode J, Kruwel T, Solecki G, Heiland S, Bendszus M, Tews B, Winkler F, Breckwoldt MO and Kurz FT: Glioblastoma multiforme restructures the topological connectivity of cerebrovascular networks. Sci Rep. 9:117572019. View Article : Google Scholar : PubMed/NCBI | |
Moreno A, Pitoc GA, Ganson NJ, Layzer JM, Hershfield MS, Tarantal AF and Sullenger BA: Anti-PEG Antibodies inhibit the anticoagulant activity of PEgylated aptamers. Cell Chem Biol. 26:634–644.e3. 2019. View Article : Google Scholar : PubMed/NCBI | |
Verry C, Dufort S, Villa J, Gavard M, Iriart C, Grand S, Charles J, Chovelon B, Cracowski JL, Quesada JL, et al: Theranostic AGuIX nanoparticles as radiosensitizer: A phase I, dose-escalation study in patients with multiple brain metastases (NANO-RAD trial). Radiother Oncol. 160:159–165. 2021. View Article : Google Scholar : PubMed/NCBI | |
Zhanataev AK, Anisina EA, Kulakova AV, Shilovskiy IP, Lisitsyn AA, Koloskova OO, Khaitov MR and Durnev AD: Genotoxicity of cationic lipopeptide nanoparticles. Toxicol Lett. 328:1–6. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lalatsa A and Butt AM: Physiology of the Blood-brain barrier and mechanisms of transport across the BBB. Nanotechnology-based Targeted Drug Delivery Systems for Brain Tumors. Elsevier Inc. 49–74. 2018. | |
Crone C and Olesen SP: Electrical resistance of brain microvascular endothelium. Brain Res. 241:49–55. 1982. View Article : Google Scholar : PubMed/NCBI | |
Kadry H, Noorani B and Cucullo L: A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS. 17:692020. View Article : Google Scholar : PubMed/NCBI | |
Andreone BJ, Chow BW, Tata A, Lacoste B, Ben-Zvi A, Bullock K, Deik AA, Ginty DD, Clish CB and Gu C: Blood-brain barrier permeability is regulated by lipid transport-dependent suppression of caveolae-mediated transcytosis. Neuron. 94:581–594.e5. 2017. View Article : Google Scholar : PubMed/NCBI | |
Eng ME, Imperio GE, Bloise E and Matthews SG: ATP-binding cassette (ABC) drug transporters in the developing blood-brain barrier: Role in fetal brain protection. Cell Mol Life Sci. 79:4152022. View Article : Google Scholar : PubMed/NCBI | |
Lochhead JJ, Yang J, Ronaldson PT and Davis TP: Structure, function, and regulation of the blood-brain barrier tight junction in central nervous system disorders. Front Physiol. 11:9142020. View Article : Google Scholar : PubMed/NCBI | |
Salama NN, Eddington ND and Fasano A: Tight junction modulation and its relationship to drug delivery. Adv Drug Deliv Rev. 58:15–28. 2006. View Article : Google Scholar : PubMed/NCBI | |
Lemmer HJ and Hamman JH: Paracellular drug absorption enhancement through tight junction modulation. Expert Opin Drug Deliv. 10:103–114. 2013. View Article : Google Scholar | |
Lin Y, Gan L, Ren L, Ma C, Dai M, Qian K, Ye Q and Lin X: Acupuncture with specific mode electro-stimulation effectively and transiently opens the BBB through Shh signaling pathway. Neuroreport. 34:873–886. 2023. View Article : Google Scholar : PubMed/NCBI | |
Yang J, Betterton RD, Williams EI, Stanton JA, Reddell ES, Ogbonnaya CE, Dorn E, Davis TP, Lochhead JJ and Ronaldson PT: High-dose acetaminophen alters the integrity of the blood-brain barrier and leads to increased CNS uptake of codeine in rats. Pharmaceutics. 14:9492022. View Article : Google Scholar : PubMed/NCBI | |
Han S, Mei L, Quach T, Porter C and Trevaskis N: Lipophilic conjugates of drugs: A tool to improve drug pharmacokinetic and therapeutic profiles. Pharm Res. 38:1497–1518. 2021. View Article : Google Scholar : PubMed/NCBI | |
Muller J, Martins A, Csabi J, Fenyvesi F, Konczol A, Hunyadi A and Balogh GT: BBB penetration-targeting physicochemical lead selection: Ecdysteroids as chemo-sensitizers against CNS tumors. Eur J Pharm Sci. 96:571–577. 2017. View Article : Google Scholar | |
Rankovic Z: CNS drug design: Balancing physicochemical properties for optimal brain exposure. J Med Chem. 58:2584–2608. 2015. View Article : Google Scholar | |
van Tellingen O, Yetkin-Arik B, de Gooijer MC, Wesseling P, Wurdinger T and de Vries HE: Overcoming the blood-brain tumor barrier for effective glioblastoma treatment. Drug Resist Updat. 19:1–12. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Liu Z, Gao C, Bian H, Ma Y, Jing F and Zhao X: Role of rituximab in treatment of patients with primary central nervous system lymphoma (PCNSL): A systematic review and Meta-analysis. Clin Lymphoma Myeloma Leuk. 23:733–741. 2023. View Article : Google Scholar : PubMed/NCBI | |
Paul PR, Mishra MK, Bora S, Kukal S, Singh A, Kukreti S and Kukreti R: The Impact of P-Glycoprotein on CNS drug efflux and variability in response. J Biochem Mol Toxicol. 39:e701902025. View Article : Google Scholar : PubMed/NCBI | |
Zensi A, Begley D, Pontikis C, Legros C, Mihoreanu L, Wagner S, Buchel C, von Briesen H and Kreuter J: Albumin nanoparticles targeted with Apo E enter the CNS by transcytosis and are delivered to neurones. J Control Release. 137:78–86. 2009. View Article : Google Scholar : PubMed/NCBI | |
Demeule M, Regina A, Che C, Poirier J, Nguyen T, Gabathuler R, Castaigne JP and Beliveau R: Identification and design of peptides as a new drug delivery system for the brain. J Pharmacol Exp Ther. 324:1064–1072. 2008. View Article : Google Scholar | |
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL and Moos T: Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol. 181:1016652019. View Article : Google Scholar : PubMed/NCBI | |
Xin H, Jiang X, Gu J, Sha X, Chen L, Law K, Chen Y, Wang X, Jiang Y and Fang X: Angiopep-conjugated poly(ethylene glycol)-co-poly(epsilon-caprolactone) nanoparticles as dual-targeting drug delivery system for brain glioma. Biomaterials. 32:4293–4305. 2011. View Article : Google Scholar : PubMed/NCBI | |
Gonatas NK, Stieber A, Hickey WF, Herbert SH and Gonatas JO: Endosomes and Golgi vesicles in adsorptive and fluid phase endocytosis. J Cell Biol. 99:1379–1390. 1984. View Article : Google Scholar : PubMed/NCBI | |
Szecsko A, Meszaros M, Simoes B, Cavaco M, Chaparro C, Porkolab G, Castanho M, Deli MA, Neves V and Veszelka S: PepH3-modified nanocarriers for delivery of therapeutics across the blood-brain barrier. Fluids Barriers CNS. 22:312025. View Article : Google Scholar : PubMed/NCBI | |
Li Y, Zheng X, Gong M and Zhang J: Delivery of a peptide-drug conjugate targeting the blood brain barrier improved the efficacy of paclitaxel against glioma. Oncotarget. 7:79401–79407. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang ZH, Wang ZY, Sun CS, Wang CY, Jiang TY and Wang SL: Trimethylated chitosan-conjugated PLGA nanoparticles for the delivery of drugs to the brain. Biomaterials. 31:908–915. 2010. View Article : Google Scholar | |
Dowaidar M: Cell-penetrating peptides with nanoparticles hybrid delivery vectors and their uptake pathways. Mitochondrion. 78:1019062024. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Huang R, Han L, Ke W, Shao K, Ye L, Lou J and Jiang C: Brain-targeting gene delivery and cellular internalization mechanisms for modified rabies virus glycoprotein RVG29 nanoparticles. Biomaterials. 30:4195–4202. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wu MC, Wang EY and Lai TW: TAT peptide at treatment-level concentrations crossed brain endothelial cell monolayer independent of receptor-mediated endocytosis or peptide-inflicted barrier disruption. PLoS One. 18:e02926812023. View Article : Google Scholar : PubMed/NCBI | |
Hu C, Tao L, Cao X and Chen L: The solute carrier transporters and the brain: Physiological and pharmacological implications. Asian J Pharm Sci. 15:131–144. 2020. View Article : Google Scholar : PubMed/NCBI | |
Al-Ahmad AJ: Comparative study of expression and activity of glucose transporters between stem cell-derived brain microvascular endothelial cells and hCMEC/D3 cells. Am J Physiol Cell Physiol. 313:C421–C429. 2017. View Article : Google Scholar : PubMed/NCBI | |
Singh N and Ecker GF: Insights into the structure, function, and ligand discovery of the large neutral amino acid transporter 1, LAT1. Int J Mol Sci. 19:12782018. View Article : Google Scholar : PubMed/NCBI | |
Vijay N and Morris ME: Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des. 20:1487–1498. 2014. View Article : Google Scholar : | |
Jiang X, Xin H, Ren Q, Gu J, Zhu L, Du F, Feng C, Xie Y, Sha X and Fang X: Nanoparticles of 2-deoxy-D-glucose functionalized poly(ethylene glycol)-co-poly(trimethylene carbonate) for dual-targeted drug delivery in glioma treatment. Biomaterials. 35:518–529. 2014. View Article : Google Scholar | |
Campani V, Zappavigna S, Scotti L, Abate M, Porru M, Leonetti C, Caraglia M and De Rosa G: Hybrid lipid self-assembling nanoparticles for brain delivery of microRNA. Int J Pharm. 588:1196932020. View Article : Google Scholar : PubMed/NCBI | |
Estella-Hermoso de Mendoza A, Preat V, Mollinedo F and Blanco-Prieto MJ: In vitro and in vivo efficacy of edelfosine-loaded lipid nanoparticles against glioma. J Control Release. 156:421–426. 2011. View Article : Google Scholar : PubMed/NCBI | |
Grafals-Ruiz N, Rios-Vicil CI, Lozada-Delgado EL, Quinones-Diaz BI, Noriega-Rivera RA, Martinez-Zayas G, Santana-Rivera Y, Santiago-Sanchez GS, Valiyeva F and Vivas-Mejia PE: Brain targeted gold liposomes improve RNAi delivery for glioblastoma. Int J Nanomedicine. 15:2809–2828. 2020. View Article : Google Scholar : PubMed/NCBI | |
Pandian SRK, Pavadai P, Vellaisamy S, Ravishankar V, Palanisamy P, Sundar LM, Chandramohan V, Sankaranarayanan M, Panneerselvam T and Kunjiappan S: Formulation and evaluation of Rutin-loaded solid lipid nanoparticles for the treatment of brain tumor. Naunyn Schmiedebergs Arch Pharmacol. 394:735–749. 2021. View Article : Google Scholar | |
Norouzi M, Yathindranath V, Thliveris JA, Kopec BM, Siahaan TJ and Miller DW: Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy: A combinational approach for enhanced delivery of nanoparticles. Sci Rep. 10:112922020. View Article : Google Scholar : PubMed/NCBI | |
Wohlfart S, Khalansky AS, Gelperina S, Maksimenko O, Bernreuther C, Glatzel M and Kreuter J: Efficient chemotherapy of rat glioblastoma using doxorubicin-loaded PLGA nanoparticles with different stabilizers. PLoS One. 6:e191212011. View Article : Google Scholar : PubMed/NCBI | |
Zhao Y, Yin H and Zhang X: Modification of graphene oxide by angiopep-2 enhances anti-glioma efficiency of the nanoscaled delivery system for doxorubicin. Aging (Albany NY). 12:10506–10516. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cen J, Dai X, Zhao H, Li X, Hu X, Wu J and Duan S: Doxorubicin-loaded liposome with the function of 'Killing two birds with one stone' against Glioma. ACS Appl Mater Interfaces. 15:46697–46709. 2023. View Article : Google Scholar : PubMed/NCBI | |
He H, Li Y, Jia XR, Du J, Ying X, Lu WL, Lou JN and Wei Y: PEGylated Poly(amidoamine) dendrimer-based dual-targeting carrier for treating brain tumors. Biomaterials. 32:478–487. 2011. View Article : Google Scholar | |
Khoury ES, Sharma A, Ramireddy RR, Thomas AG, Alt J, Fowler A, Rais R, Tsukamoto T, Blue ME, Slusher B, et al: Dendrimer-conjugated glutaminase inhibitor selectively targets microglial glutaminase in a mouse model of Rett syndrome. Theranostics. 10:5736–5748. 2020. View Article : Google Scholar : PubMed/NCBI | |
Liu C, Zhao Z, Gao H, Rostami I, You Q, Jia X, Wang C, Zhu L and Yang Y: Enhanced blood-brain-barrier penetrability and tumor-targeting efficiency by peptide-functionalized poly(amidoamine) dendrimer for the therapy of gliomas. Nanotheranostics. 3:311–330. 2019. View Article : Google Scholar : PubMed/NCBI | |
Saw PE, Zhang A, Nie Y, Zhang L, Xu Y and Xu X: Tumor-associated fibronectin targeted liposomal nanoplatform for cyclophilin a siRNA delivery and targeted malignant glioblastoma therapy. Front Pharmacol. 9:11942018. View Article : Google Scholar : PubMed/NCBI | |
Zong T, Mei L, Gao H, Shi K, Chen J, Wang Y, Zhang Q, Yang Y and He Q: Enhanced glioma targeting and penetration by dual-targeting liposome co-modified with T7 and TAT. J Pharm Sci. 103:3891–3901. 2014. View Article : Google Scholar : PubMed/NCBI | |
Feng Q, Shen Y, Fu Y, Muroski ME, Zhang P, Wang Q, Xu C, Lesniak MS, Li G and Cheng Y: Self-assembly of gold nanoparticles shows microenvironment-mediated dynamic switching and enhanced brain tumor targeting. Theranostics. 7:1875–1889. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liaw K, Sharma R, Sharma A, Salazar S, Appiani La Rosa S and Kannan RM: Systemic dendrimer delivery of triptolide to tumor-associated macrophages improves Anti-tumor efficacy and reduces systemic toxicity in glioblastoma. J Control Release. 329:434–444. 2021. View Article : Google Scholar : | |
Lu W, Sun Q, Wan J, She Z and Jiang XG: Cationic albumin-conjugated pegylated nanoparticles allow gene delivery into brain tumors via intravenous administration. Cancer Res. 66:11878–11887. 2006. View Article : Google Scholar : PubMed/NCBI | |
Negron K, Khalasawi N, Lu B, Ho CY, Lee J, Shenoy S, Mao HQ, Wang TH, Hanes J and Suk JS: Widespread gene transfer to malignant gliomas with In vitro-to-In vivo correlation. J Control Release. 303:1–11. 2019. View Article : Google Scholar : PubMed/NCBI | |
Wang Y, Wang K, Zhao J, Liu X, Bu J, Yan X and Huang R: Multifunctional mesoporous Silica-coated graphene nanosheet used for chemo-photothermal synergistic targeted therapy of glioma. J Am Chem Soc. 135:4799–4804. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Yu J, Zhang L, Cai J, Cai D and Lv C: Enhanced anti-tumor effects of doxorubicin on glioma by entrapping in polybutylcyanoacrylate nanoparticles. Tumour Biol. 37:2703–2708. 2016. View Article : Google Scholar | |
Dal Magro R, Ornaghi F, Cambianica I, Beretta S, Re F, Musicanti C, Rigolio R, Donzelli E, Canta A, Ballarini E, et al: ApoE-modified solid lipid nanoparticles: A feasible strategy to cross the blood-brain barrier. J Control Release. 249:103–110. 2017. View Article : Google Scholar : PubMed/NCBI | |
Demeule M, Currie JC, Bertrand Y, Che C, Nguyen T, Regina A, Gabathuler R, Castaigne JP and Beliveau R: Involvement of the low-density lipoprotein receptor-related protein in the transcytosis of the brain delivery vector angiopep-2. J Neurochem. 106:1534–1544. 2008. View Article : Google Scholar : PubMed/NCBI | |
Hultqvist G, Syvanen S, Fang XT, Lannfelt L and Sehlin D: Bivalent brain shuttle increases antibody uptake by monovalent binding to the transferrin receptor. Theranostics. 7:308–318. 2017. View Article : Google Scholar : PubMed/NCBI | |
Molino Y, David M, Varini K, Jabes F, Gaudin N, Fortoul A, Bakloul K, Masse M, Bernard A, Drobecq L, et al: Use of LDL receptor-targeting peptide vectors for in vitro and in vivo cargo transport across the blood-brain barrier. FASEB J. 31:1807–1827. 2017. View Article : Google Scholar : PubMed/NCBI | |
Pardridge WM, Kang YS, Buciak JL and Yang J: Human insulin receptor monoclonal antibody undergoes high affinity binding to human brain capillaries in vitro and rapid transcytosis through the blood-brain barrier in vivo in the primate. Pharm Res. 12:807–816. 1995. View Article : Google Scholar : PubMed/NCBI | |
Ribecco-Lutkiewicz M, Sodja C, Haukenfrers J, Haqqani AS, Ly D, Zachar P, Baumann E, Ball M, Huang J, Rukhlova M, et al: A novel human induced pluripotent stem cell blood-brain barrier model: Applicability to study antibody-triggered receptor-mediated transcytosis. Sci Rep. 8:18732018. View Article : Google Scholar : PubMed/NCBI | |
Wu LP, Ahmadvand D, Su J, Hall A, Tan X, Farhangrazi ZS and Moghimi SM: Crossing the blood-brain-barrier with nanoligand drug carriers self-assembled from a phage display peptide. Nat Commun. 10:46352019. View Article : Google Scholar : PubMed/NCBI | |
Zhan C, Li B, Hu L, Wei X, Feng L, Fu W and Lu W: Micelle-based Brain-targeted drug delivery enabled by a nicotine acetylcholine receptor ligand. Angew Chem Int Ed Engl. 50:5482–5485. 2011. View Article : Google Scholar : PubMed/NCBI | |
Bera S, Kar RK, Mondal S, Pahan K and Bhunia A: Structural elucidation of the Cell-penetrating penetratin peptide in model membranes at the atomic level: Probing hydrophobic interactions in the Blood-brain barrier. Biochemistry. 55:4982–4996. 2016. View Article : Google Scholar : PubMed/NCBI | |
Bertrand Y, Currie JC, Poirier J, Demeule M, Abulrob A, Fatehi D, Stanimirovic D, Sartelet H, Castaigne JP and Beliveau R: Influence of glioma tumour microenvironment on the transport of ANG1005 via low-density lipoprotein receptor-related protein 1. Br J Cancer. 105:1697–1707. 2011. View Article : Google Scholar : PubMed/NCBI | |
Du D, Chang N, Sun S, Li M, Yu H, Liu M, Liu X, Wang G, Li H, Liu X, et al: The role of glucose transporters in the distribution of p-aminophenyl-α-d-mannopyranoside modified liposomes within mice brain. J Control Release. 182:99–110. 2014. View Article : Google Scholar : PubMed/NCBI | |
Joshi S, Cooke JRN, Ellis JA, Emala CW and Bruce JN: Targeting brain tumors by intra-arterial delivery of cell-penetrating peptides: A novel approach for primary and metastatic brain malignancy. J Neurooncol. 135:497–506. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Li J, Shao K, Huang R, Ye L, Lou J and Jiang C: A leptin derived 30-amino-acid peptide modified pegylated poly-L-lysine dendrigraft for brain targeted gene delivery. Biomaterials. 31:5246–5257. 2010. View Article : Google Scholar : PubMed/NCBI | |
Molina-Arcas M, Casado FJ and Pastor-Anglada M: Nucleoside transporter proteins. Curr Vasc Pharmacol. 7:426–434. 2009. View Article : Google Scholar : PubMed/NCBI | |
Romano A, Koczwara JB, Gallelli CA, Vergara D, Micioni Di Bonaventura MV, Gaetani S and Giudett AM: Fats for thoughts: An update on brain fatty acid metabolism. Int J Biochem Cell Biol. 84:40–45. 2017. View Article : Google Scholar : PubMed/NCBI | |
Strickland M and Stoll EA: Metabolic reprogramming in glioma. Front Cell Dev Biol. 5:432017. View Article : Google Scholar : PubMed/NCBI | |
Yang Y, Yang Y, Xie X, Cai X, Zhang H, Gong W, Wang Z and Mei X: PEGylated liposomes with NGR ligand and heat-activable cell-penetrating peptide-doxorubicin conjugate for tumor-specific therapy. Biomaterials. 35:4368–4381. 2014. View Article : Google Scholar : PubMed/NCBI | |
Minty A, Chalon P, Derocq JM, Dumont X, Guillemot JC, Kaghad M, Labit C, Leplatois P, Liauzun P, Miloux B, et al: Interleukin-13 is a new human lymphokine regulating inflammatory and immune responses. Nature. 362:248–250. 1993. View Article : Google Scholar : PubMed/NCBI | |
Debinski W and Gibo DM: Molecular expression analysis of restrictive receptor for interleukin 13, a brain tumor-associated cancer/testis antigen. Mol Med. 6:440–449. 2000. View Article : Google Scholar : PubMed/NCBI | |
Kawakami M, Kawakami K, Takahashi S, Abe M and Puri RK: Analysis of interleukin-13 receptor alpha2 expression in human pediatric brain tumors. Cancer. 101:1036–1042. 2004. View Article : Google Scholar : PubMed/NCBI | |
Madhankumar AB, Slagle-Webb B, Mintz A, Sheehan JM and Connor JR: Interleukin-13 receptor-targeted nanovesicles are a potential therapy for glioblastoma multiforme. Mol Cancer Ther. 5:3162–3169. 2006. View Article : Google Scholar : PubMed/NCBI | |
Wei L, Guo XY, Yang T, Yu MZ, Chen DW and Wang JC: Brain tumor-targeted therapy by systemic delivery of siRNA with Transferrin receptor-mediated core-shell nanoparticles. Int J Pharm. 510:394–405. 2016. View Article : Google Scholar : PubMed/NCBI | |
Gong Z, Chi C, Huang X, Chu H, Wang J, Du F, Jiang L and Chen J: Cyclophilin a is overexpressed in hepatocellular carcinoma and is associated with the cell cycle. Anticancer Res. 37:4443–4447. 2017.PubMed/NCBI | |
Jain R, Atak A, Yeola A and Srivastava S: Proteomic level changes associated with S3I201 treated U87 glioma cells. J Proteomics. 150:341–350. 2017. View Article : Google Scholar | |
Yang H, Chen J, Yang J, Qiao S, Zhao S and Yu L: Cyclophilin A is upregulated in small cell lung cancer and activates ERK1/2 signal. Biochem Biophys Res Commun. 361:763–767. 2007. View Article : Google Scholar : PubMed/NCBI | |
Campani V, De Rosa G, Misso G, Zarone MR and Grimaldi A: Lipid nanoparticles to deliver miRNA in cancer. Curr Pharm Biotechnol. 17:741–749. 2016. View Article : Google Scholar : PubMed/NCBI | |
Singh A, Trivedi P and Jain NK: Advances in siRNA delivery in cancer therapy. Artif Cells Nanomed Biotechnol. 46:274–283. 2018. View Article : Google Scholar | |
Pink DL, Loruthai O, Ziolek RM, Wasutrasawat P, Terry AE, Lawrence MJ and Lorenz CD: On the structure of solid lipid nanoparticles. Small. 15:e19031562019. View Article : Google Scholar : PubMed/NCBI | |
Negahdari R, Bohlouli S, Sharifi S, Maleki Dizaj S, Rahbar Saadat Y, Khezri K, Jafari S, Ahmadian E, Gorbani Jahandizi N and Raeesi S: Therapeutic benefits of rutin and its nanoformulations. Phytother Res. 35:1719–1738. 2021. View Article : Google Scholar | |
Ishak RAH, Mostafa NM and Kamel AO: Stealth lipid polymer hybrid nanoparticles loaded with rutin for effective brain delivery-comparative study with the gold standard (Tween 80): Optimization, characterization and biodistribution. Drug Deliv. 24:1874–1890. 2017. View Article : Google Scholar : PubMed/NCBI | |
Neves AR, Queiroz JF, Lima SAC and Reis S: Apo E-Functionalization of solid lipid nanoparticles enhances brain drug delivery: Uptake mechanism and transport pathways. Bioconjug Chem. 28:995–1004. 2017. View Article : Google Scholar : PubMed/NCBI | |
Banerjee I, De K, Mukherjee D, Dey G, Chattopadhyay S, Mukherjee M, Mandal M, Bandyopadhyay AK, Gupta A, Ganguly S and Misra M: Paclitaxel-loaded solid lipid nanoparticles modified with Tyr-3-octreotide for enhanced anti-angiogenic and anti-glioma therapy. Acta Biomater. 38:69–81. 2016. View Article : Google Scholar : PubMed/NCBI | |
Mukherjee A, Waters AK, Kalyan P, Achrol AS, Kesari S and Yenugonda VM: Lipid-polymer hybrid nanoparticles as a next-generation drug delivery platform: State of the art, emerging technologies, and perspectives. Int J Nanomedicine. 14:1937–1952. 2019. View Article : Google Scholar : PubMed/NCBI | |
Salzano G, Zappavigna S, Luce A, D'Onofrio N, Balestrieri ML, Grimaldi A, Lusa S, Ingrosso D, Artuso S, Porru M, et al: Transferrin-targeted nanoparticles containing zoledronic acid as a potential tool to inhibit glioblastoma growth. J Biomed Nanotechnol. 12:811–830. 2016. View Article : Google Scholar : PubMed/NCBI | |
Aili T, Zong JB, Zhou YF, Liu YX, Yang XL, Hu B and Wu JH: Recent advances of self-assembled nanoparticles in the diagnosis and treatment of atherosclerosis. Theranostics. 14:7505–7533. 2024. View Article : Google Scholar : PubMed/NCBI | |
Chen C, Shen M, Liao H, Guo Q, Fu H, Yu J and Duan Y: A paclitaxel and microRNA-124 coloaded stepped cleavable nanosystem against triple negative breast cancer. J Nanobiotechnology. 19:552021. View Article : Google Scholar : PubMed/NCBI | |
Srinageshwar B, Peruzzaro S, Andrews M, Johnson K, Hietpas A, Clark B, McGuire C, Petersen E, Kippe J, Stewart A, et al: PAMAM dendrimers cross the Blood-brain barrier when administered through the carotid artery in C57BL/6J mice. Int J Mol Sci. 18:6282017. View Article : Google Scholar : PubMed/NCBI | |
Li J, Liang H, Liu J and Wang Z: Poly (amidoamine) (PAMAM) dendrimer mediated delivery of drug and pDNA/siRNA for cancer therapy. Int J Pharm. 546:215–225. 2018. View Article : Google Scholar : PubMed/NCBI | |
Fana M, Gallien J, Srinageshwar B, Dunbar GL and Rossignol J: PAMAM dendrimer nanomolecules utilized as drug delivery systems for potential treatment of glioblastoma: A systematic review. Int J Nanomedicine. 15:2789–2808. 2020. View Article : Google Scholar : PubMed/NCBI | |
Saraswathy M, Knight GT, Pilla S, Ashton RS and Gong S: Multifunctional drug nanocarriers formed by cRGD-conjugated betaCD-PAMAM-PEG for targeted cancer therapy. Colloids Surf B Biointerfaces. 126:590–597. 2015. View Article : Google Scholar : PubMed/NCBI | |
Tambe V, Thakkar S, Raval N, Sharma D, Kalia K and Tekade RK: Surface engineered dendrimers in siRNA delivery and gene silencing. Curr Pharm Des. 23:2952–2975. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liaw K, Zhang F, Mangraviti A, Kannan S, Tyler B and Kannan RM: Dendrimer size effects on the selective brain tumor targeting in orthotopic tumor models upon systemic administration. Bioeng Transl Med. 5:e101602020. View Article : Google Scholar : PubMed/NCBI | |
Sharma A, Liaw K, Sharma R, Zhang Z, Kannan S and Kannan RM: Targeting mitochondrial dysfunction and oxidative stress in activated microglia using Dendrimer-based therapeutics. Theranostics. 8:5529–5547. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sharma R, Kim SY, Sharma A, Zhang Z, Kambhampati SP, Kannan S and Kannan RM: Activated microglia targeting Dendrimer-minocycline conjugate as therapeutics for neuroinflammation. Bioconjug Chem. 28:2874–2886. 2017. View Article : Google Scholar : PubMed/NCBI | |
Hu Q, Yao J, Wang X, Wang Y, Fu X, Ma J, Lin H, Xu J, Shen L and Yu X: Combinational chemoimmunotherapy for breast cancer by codelivery of doxorubicin and PD-L1 siRNA using a PAMAM-incorporated liposomal nanoplatform. ACS Appl Mater Interfaces. 14:8782–8792. 2022. View Article : Google Scholar : PubMed/NCBI | |
Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N and Tyler BM: Polymeric nanoparticles in brain cancer therapy: A review of current approaches. Polymers (Basel). 14:29632022. View Article : Google Scholar : PubMed/NCBI | |
Qian L, Zheng J, Wang K, Tang Y, Zhang X, Zhang H, Huang F, Pei Y and Jiang Y: Cationic core-shell nanoparticles with carmustine contained within O6-benzylguanine shell for glioma therapy. Biomaterials. 34:8968–8978. 2013. View Article : Google Scholar : PubMed/NCBI | |
Zhang P, Hu L, Yin Q, Feng L and Li Y: Transferrin-modified c[RGDfK]-paclitaxel loaded hybrid micelle for sequential blood-brain barrier penetration and glioma targeting therapy. Mol Pharm. 9:1590–1598. 2012. View Article : Google Scholar : PubMed/NCBI | |
Xin H, Chen L, Gu J, Ren X, Wei Z, Luo J, Chen Y, Jiang X, Sha X and Fang X: Enhanced anti-glioblastoma efficacy by PTX-loaded PEGylated poly(varepsilon-caprolactone) nanoparticles: In vitro and in vivo evaluation. Int J Pharm. 402:238–247. 2010. View Article : Google Scholar : PubMed/NCBI | |
Izquierdo M: Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Ther. 12:217–227. 2005. View Article : Google Scholar | |
Razavi ZS, Razavi FS and Alizadeh SS: Inorganic nanoparticles and blood-brain barrier modulation: Advancing targeted neurological therapies. Eur J Med Chem. 287:1173572025. View Article : Google Scholar : PubMed/NCBI | |
Liu Y, Bhattarai P, Dai Z and Chen X: Photothermal therapy and photoacoustic imaging via nanotheranostics in fighting cancer. Chem Soc Rev. 48:2053–2108. 2019. View Article : Google Scholar : | |
Bernardi RJ, Lowery AR, Thompson PA, Blaney SM and West JL: Immunonanoshells for targeted photothermal ablation in medulloblastoma and glioma: An in vitro evaluation using human cell lines. J Neurooncol. 86:165–172. 2008. View Article : Google Scholar | |
Ruan S, Xie R, Qin L, Yu M, Xiao W, Hu C, Yu W, Qian Z, Ouyang L, He Q and Gao H: Aggregable Nanoparticles-enabled chemotherapy and autophagy inhibition combined with Anti-PD-L1 antibody for improved glioma treatment. Nano Lett. 19:8318–8332. 2019. View Article : Google Scholar : PubMed/NCBI | |
Qiu J, Kong L, Cao X, Li A, Wei P, Wang L, Mignani S, Caminade AM, Majoral JP and Shi X: Enhanced delivery of therapeutic siRNA into glioblastoma cells using Dendrimer-entrapped gold nanoparticles conjugated with β-cyclodextrin. Nanomaterials (Basel). 8:1312018. View Article : Google Scholar | |
Maleki P, Dinari A, Jahangiri B and Raheb J: In vitro assessments of nanoplexes of polyethylenimine-coated graphene oxide-plasmid through various cancer cell lines and primary mesenchymal stem cells. PLoS One. 18:e02958222023. View Article : Google Scholar : PubMed/NCBI | |
Chuang CC, Lan YH, Lu YJ, Weng YL and Chen JP: Targeted delivery of irinotecan and SLP2 shRNA with GRP-conjugated magnetic graphene oxide for glioblastoma treatment. Biomater Sci. 10:3201–3222. 2022. View Article : Google Scholar : PubMed/NCBI | |
Norouzi M, Yathindranath V, Thliveris JA and Miller DW: Salinomycin-loaded iron oxide nanoparticles for glioblastoma therapy. Nanomaterials (Basel). 10:4772020. View Article : Google Scholar : PubMed/NCBI | |
Saalik P, Lingasamy P, Toome K, Mastandrea I, Rousso-Noori L, Tobi A, Simon-Gracia L, Hunt H, Paiste P, Kotamraju VR, et al: Peptide-guided nanoparticles for glioblastoma targeting. J Control Release. 308:109–118. 2019. View Article : Google Scholar : PubMed/NCBI | |
Manago A, Leanza L, Carraretto L, Sassi N, Grancara S, Quintana-Cabrera R, Trimarco V, Toninello A, Scorrano L, Trentin L, et al: Early effects of the antineoplastic agent salinomycin on mitochondrial function. Cell Death Dis. 6:e19302015. View Article : Google Scholar : PubMed/NCBI | |
Zhang Y, Fu X, Jia J, Wikerholmen T, Xi K, Kong Y, Wang J, Chen H, Ma Y, Li Z, et al: Glioblastoma therapy using codelivery of cisplatin and glutathione peroxidase targeting siRNA from iron oxide nanoparticles. ACS Appl Mater Interfaces. 12:43408–43421. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang X, Ruifang L, Zhu Y, Wang Z, Zhang H, Cui L, Duan S and Guo Y: Active targeting co-delivery of therapeutic Sur siRNA and an antineoplastic drug via epidermal growth factor receptor-mediated magnetic nanoparticles for synergistic programmed cell death in glioblastoma stem cells. Materials Chemistry Front. 4:574–588. 2019. View Article : Google Scholar | |
Nichols JW and Bae YH: EPR: Evidence and fallacy. J Control Release. 190:451–464. 2014. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Yu M, Ning X, Zhou C, Yang S and Zheng J: PEGylation and zwitterionization: Pros and cons in the renal clearance and tumor targeting of near-IR-emitting gold nanoparticles. Angew Chem Int Ed Engl. 52:12572–12576. 2013. View Article : Google Scholar : PubMed/NCBI | |
Corbo C, Molinaro R, Parodi A, Toledano Furman NE, Salvatore F and Tasciotti E: The impact of nanoparticle protein corona on cytotoxicity, immunotoxicity and target drug delivery. Nanomedicine (Lond). 11:81–100. 2016. View Article : Google Scholar | |
Zhang TX, Zhu GY, Lu BY, Zhang CL and Peng Q: Concentration-dependent protein adsorption at the nano-bio interfaces of polymeric nanoparticles and serum proteins. Nanomedicine (Lond). 12:2757–2769. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu J, Dong J, Zhang T and Peng Q: Graphene-based nanomaterials and their potentials in advanced drug delivery and cancer therapy. J Control Release. 286:64–73. 2018. View Article : Google Scholar : PubMed/NCBI | |
Xiao W, Wang Y, Zhang H, Liu Y, Xie R, He X, Zhou Y, Liang L and Gao H: The protein corona hampers the transcytosis of Transferrin-modified nanoparticles through blood-brain barrier and attenuates their targeting ability to brain tumor. Biomaterials. 274:1208882021. View Article : Google Scholar : PubMed/NCBI | |
Schottler S, Landfester K and Mailander V: Controlling the stealth effect of nanocarriers through understanding the protein corona. Angew Chem Int Ed Engl. 55:8806–8815. 2016. View Article : Google Scholar : PubMed/NCBI | |
Gao H and He Q: The interaction of nanoparticles with plasma proteins and the consequent influence on nanoparticles behavior. Expert Opin Drug Deliv. 11:409–420. 2014. View Article : Google Scholar : PubMed/NCBI | |
Peng Q, Wei XQ, Yang Q, Zhang S, Zhang T, Shao XR, Cai XX, Zhang ZR and Lin YF: Enhanced biostability of nanoparticle-based drug delivery systems by albumin corona. Nanomedicine (Lond). 10:205–214. 2015. View Article : Google Scholar : PubMed/NCBI | |
Zhang Z, Guan J, Jiang Z, Yang Y, Liu J, Hua W, Mao Y, Li C, Lu W, Qian J and Zhan C: Brain-targeted drug delivery by manipulating protein corona functions. Nat Commun. 10:35612019. View Article : Google Scholar : PubMed/NCBI | |
Zhang ZA, Xin X, Liu C, Liu YH, Duan HX, Qi LL, Zhang YY, Zhao HM, Chen LQ, Jin MJ, et al: Novel brain-targeted nanomicelles for anti-glioma therapy mediated by the ApoE-enriched protein corona in vivo. J Nanobiotechnology. 19:4532021. View Article : Google Scholar : PubMed/NCBI | |
Cheng Z, Li M, Dey R and Chen Y: Nanomaterials for cancer therapy: Current progress and perspectives. J Hematol Oncol. 14:852021. View Article : Google Scholar : PubMed/NCBI | |
Xu HL, Yang JJ, ZhuGe DL, Lin MT, Zhu QY, Jin BH, Tong MQ, Shen BX, Xiao J and Zhao YZ: Glioma-targeted delivery of a theranostic liposome integrated with quantum dots, superparamagnetic iron oxide, and cilengitide for Dual-imaging guiding cancer surgery. Adv Healthc Mater. 7:e17011302018. View Article : Google Scholar : PubMed/NCBI | |
Wu VM, Huynh E, Tang S and Uskokovic V: Brain and bone cancer targeting by a ferrofluid composed of superparamagnetic iron-oxide/silica/carbon nanoparticles (earthicles). Acta Biomater. 88:422–447. 2019. View Article : Google Scholar : PubMed/NCBI | |
Arias-Ramos N, Ibarra LE, Serrano-Torres M, Yague B, Caverzan MD, Chesta CA, Palacios RE and Lopez-Larrubia P: Iron oxide incorporated conjugated polymer nanoparticles for simultaneous use in magnetic resonance and fluorescent imaging of brain tumors. Pharmaceutics. 13:12582021. View Article : Google Scholar : PubMed/NCBI |