Next Article in Journal
Short-Term Starvation Weakens the Efficacy of Cell Cycle Specific Chemotherapy Drugs through G1 Arrest
Next Article in Special Issue
Analysis of the Electron Density of a Water Molecule Encapsulated by Two Cholic Acid Residues
Previous Article in Journal
Polypyrrole-Stabilized Polypeptide for Eco-Friendly Supercapacitors
Previous Article in Special Issue
High-Performance and Stable Perovskite Solar Cells Using Carbon Quantum Dots and Upconversion Nanoparticles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances on Surface-Modified GBM Targeted Nanoparticles: Targeting Strategies and Surface Characterization

1
Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
2
IRCCS Fondazione Don Carlo Gnocchi ONLUS, 20148 Milan, Italy
3
Nanotech Lab, TE.FAR.T.I., Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2496; https://doi.org/10.3390/ijms24032496
Submission received: 26 December 2022 / Revised: 20 January 2023 / Accepted: 24 January 2023 / Published: 27 January 2023
(This article belongs to the Special Issue Nano-Materials and Methods 4.0)

Abstract

:
Glioblastoma multiforme (GBM) is the most common malignant brain tumor, associated with low long-term survival. Nanoparticles (NPs) developed against GBM are a promising strategy to improve current therapies, by enhancing the brain delivery of active molecules and reducing off-target effects. In particular, NPs hold high potential for the targeted delivery of chemotherapeutics both across the blood–brain barrier (BBB) and specifically to GBM cell receptors, pathways, or the tumor microenvironment (TME). In this review, the most recent strategies to deliver drugs to GBM are explored. The main focus is on how surface functionalizations are essential for BBB crossing and for tumor specific targeting. We give a critical analysis of the various ligand-based approaches that have been used to target specific cancer cell receptors and the TME, or to interfere with the signaling pathways of GBM. Despite the increasing application of NPs in the clinical setting, new methods for ligand and surface characterization are needed to optimize the synthesis, as well as to predict their in vivo behavior. An expert opinion is given on the future of this research and what is still missing to create and characterize a functional NP system for improved GBM targeting.

1. Introduction

Brain tumors are a wide and heterogenous class of neoplasms that differentiate in epidemiology, prognosis, and histological, molecular, and clinic characteristics [1,2]. Among them, glioblastoma multiforme (GBM) is the most aggressive and common primary brain tumor. GBM is mainly diagnosed in males older than 60 years, and the incidence rate varies according to geographical area [3]. Despite the low percentage of new cases each year and advances in molecular profiling and in chemotherapy, GBM represents a major clinical issue in both the medical and the economical setting. Unfortunately, the standard of care and current treatments rarely lead to the successful eradication of the tumor, resulting in a median overall survival of less than 2 years from diagnosis [4,5,6]. The failure of GBM treatment is linked to several factors [7,8,9]. First, GBM has an infiltrative nature and is characterized by intra- and inter-tumor heterogeneity. Moreover, the blood–brain barrier (BBB) is considered one of the main obstacles for the delivery of chemotherapeutics to GBM; due to its protective nature, it prevents the delivery of drugs to the tumor site, minimizing the concentration of therapeutics and decreasing their efficacy. In addition, efflux transporters coupled with GBM stem cells can induce resistance to drugs, such as temozolomide, which is considered the gold standard of chemotherapeutic therapies against GBM. All these factors lead to a poor prognosis and tumor recurrence. Therefore, advanced therapeutic strategies have been proposed and studied to improve patient outcomes. Nanoparticles (NPs) are nanometric systems that have led to groundbreaking discoveries across the board from electronics to imaging, art, etc. In this framework, the application of NPs in the medical field, known as nanomedicine, has opened new opportunities for the treatment of hard-to-treat diseases, e.g., neurodegenerative or genetic diseases, especially GBM [10,11]. Several NPs have been tested to encapsulate traditional drugs in order to enhance drug solubility, encapsulation, and protection in the biological environment, reduce off-target toxicity, and enable passage through the BBB and/or the specific delivery of drugs to GBM via targeting, as well as controlled release at the target site [12,13,14,15,16,17,18,19,20,21,22,23]. The possibility of engineering the surface of NPs with molecules that can promote specific accumulation in the brain or in GBM cells has opened the way toward highly effective treatment options. Unfortunately, the formulation of surface-modified NPs is often lacking a thorough characterization of their surface composition, primarily due to the technical difficulty or high cost of the techniques necessary for this aim, leading to a poor possibility of optimizing the formulation of these NPs.
In this review, we highlight the recent research performed to revolutionize NP targeting to improve brain penetration, by specifically targeting the BBB, GBM cells, or the tumor microenvironment (TME) (summarized in Figure 1). Moreover, we look at current methods used to characterize the presence and quantity of targeting moieties on the NP surface, including an expert opinion on where we are and what still needs to be improved to increase our capacity to combat such a frightening and deadly disease.

2. Targeting the BBB

The BBB is a strictly selective cellular barrier located between the blood compartment and the brain. On the one hand, it regulates the permeability of small molecules and ions to ensure brain nutrition and an appropriate neuronal function; on the other hand, it prevents unwanted cells and substances from entering the brain. The presence of this highly selective barrier significantly hinders the delivery of chemotherapeutics to the brain and, thus, the treatment of brain cancer. The organization and functioning of the BBB can be altered under pathological conditions; in the case of brain tumors like GBM, a distinct form of the BBB exists and is known as the blood–brain tumor barrier (BBTB). It is well known that the BBTB of GBM patients is disrupted and leaky, which could facilitate the passage of chemotherapeutics to the brain; however, it must be said that this disruption is generally not able to allow the permeation of significant amounts of drugs [24]. Thus, it is necessary to investigate the mechanisms underlying the passage of solutes across the BBB to create BBB-permeable drug delivery systems.
Over the last decades, numerous strategies to improve the delivery of therapeutics to the brain for treating GBM have been investigated, including the use of BBB and BBTB-targeted NPs. These strategies enabled the crossing of these barriers and the penetration of the drug delivery system inside the brain, where it could release its payload to have a therapeutic effect. All these approaches utilized transport mechanisms such as passive diffusion, which depends on physicochemical properties (e.g., lipophilicity/hydrophilicity, charge, and molecular weight), or active transport, with a carrier-mediated transport or a receptor-mediated transport. In this context, receptors and transporters overexpressed on the BBB, such as transferrin (Tf) [25], insulin [26], low-density lipoprotein [27], lactoferrin (Lf) [28], nicotinic receptors [29], and glucose [30] and choline transporters [31], have been extensively studied and exploited to improve the targeting and crossing of the BBB by NPs, with the aim of GBM treatment; however, the recent literature on BBB-permeable systems evidences how targeting single receptors and transporters has limited efficacy. On the contrary, recent studies tended to focus on the use of dual or multiple ligands to maximize the possibility to cross the BBB. For example, Han and coworkers developed a formulation of nanocapsules consisting of a cleavable crosslinked peptide and 2-methacryloyloxyethyl phosphorylcholine (MPC), a molecule that contains a choline and an acetylcholine analogue. The aim was to enhance the delivery across the BBB or the BBTB of a therapeutic monoclonal antibody via choline transporter- or acetylcholine nicotinic receptor-mediated transport, in order to suppress the tumor proliferation. Results showed that nimotuzumab could be effectively delivered to the central nervous system in orthotopic U87-EGFRwt glioma xenograft mouse models, thanks to the presence of the dual targeting moiety [32]. Formicola et al. designed and developed liposomes composed of cholesterol, sphingomyelin, and DSPE–PEG–maleimide, functionalized with a modified apolipoprotein E-derived peptide (CWG-LRKLRKRLLR; mApoE) and a neurotoxin, chlorotoxin, demonstrating their synergistic effects in improving the permeability of doxorubicin-loaded liposomes across a human cell-based BBB model. Furthermore, they demonstrated that targeted doxorubicin-loaded liposomes were able to reduce the viability of GBM U87 cells seeded in the basolateral compartment to the same extent as free doxorubicin (−76.6%), but without damaging the endothelial monolayer [33]. Notwithstanding these interesting results, this study was limited to in vitro experiments, demonstrating that still much work needs to be conducted to translate those liposomes to a clinic setting. A slightly different approach was investigated by Galstyan and others. Here, the authors developed nano-immunoconjugates (NIC) consisting of a poly(β-l-malic acid) (PMLA) backbone, to which checkpoint inhibitor antibodies were covalently attached, namely, cytotoxic T-lymphocyte-associated antigen 4 (a-CTLA-4) and programmed cell death-1 (PD-1) antibodies. These two antibodies can help activate the local antitumor immune response in the brain; however, their direct administration is unsuccessful due to their inability to cross the BBB. For this reason, the antibodies were covalently linked to the PMLA backbone, which was also modified by conjugation with an anti-mouse Tf receptor antibody to enable BBB crossing using Tf receptor-mediated transport. NIC treatment of mice bearing intracranial GL261 GBM resulted in an increase in CD8+ T cells, NK cells, and macrophages with a corresponding decrease of regulatory T cells in the brain tumor area [34]. This study is a successful example of delivery across the BBB of polymer-conjugated antibodies for immunotherapy of brain tumors, which could lead to an effective GBM treatment via activation of the local brain tumor immune response.

3. Targeting GBM Cell Receptors

Active targeting to promote BBB crossing, although successful, is often insufficient to ensure the penetration of therapeutic molecules into GBM cells, resulting in off-target effects on healthy tissues in the brain that are not intended to receive chemotherapy. For this purpose, various ligands have been investigated to be conjugated on the surface of NPs to improve GBM specificity by recognizing and binding specific or overexpressed features of the GBM cell surface [35,36,37,38]. The most investigated ligands include small molecules such as folic acid (FA), Tf, and Lf, which bind to their respective receptors, and the RGD peptide that targets the αVβ3 integrin. It is noteworthy that, in recent years, most researchers did not focus on the development of novel GBM targeting ligands, but they devoted efforts toward optimizing the combination of already known targeting moieties, NP types, and drugs. This trend probably stems from the repurposing of known ligands and drugs, with well-known toxicity and mechanisms, which can boost the translation of nanosystems from bench to bedside. At the same time, this highlights the intricate co-dependency of NP type, drug used, and ligands to have a positive therapeutic outcome.
There are a few recent papers in which new ligands are proposed for specific GBM targeting, most of them being antibodies or fractions thereof. Guo et al. formulated doxorubicin-loaded PEGylated liposomes (DOPC:DSPE–PEG–COOH) decorated with the ITGA2 antibody for the selective targeting of GBM cells. ITGA2 was proposed as a novel and appealing therapeutic target since it was found to be considerably upregulated in both human GBM and other cancer cells compared to normal brain tissues. The in vitro results confirmed the selectivity of targeted liposomes toward the tumor, being able to cross a leaky BBTB but not a healthy BBB, and exhibiting an IC50 value four times smaller than the nontargeted control. The ITGA2 antibody also possessed another potential therapeutic effect by blocking GBM cell migration, further enhancing the antitumoral activity of doxorubicin [39]. Another group engineered biodegradable polymeric poly(lactic-co-glycolic) acid (PLGA) NPs with a cell-surface vimentin antibody M08. M08-NPs showed increased specificity for GBM along with an ability to interfere with tumor cell numbers while promoting healthy cell adhesion when tested in coculture assays. Moreover, a major apoptotic effect and cell death were observed for paclitaxel-loaded M08-NPs compared to nontargeted NPs or free paclitaxel. These results demonstrated the potential of this ligand to be a novel targeting candidate that could be conjugated onto a drug delivery platform for the treatment of GBM [40]. A major limitation to both the articles by Guo et al. and Duskey et al. is that all assays were only performed in vitro, and further in vivo analyses are needed to corroborate these results.
In the framework of tumor-homing peptides, a new targeting ligand (Ser–Ile–Trp–Val/SIWV), derived from an isoform of annexin A3, was discovered by Huh et al. SIWV was coupled with porous silica NPs (SIWV-NPs) in order to selectively deliver an anticancer drug to GBM. Biodistribution analysis in BALB/c mice with xenograft implantation showed successful brain accumulation with a higher presence of silica SIWV NPs at the site of the GBM xenograft. This resulted in a prolonged survival rate and a significant reduction in tumor mass, probably thanks to the caveolin overexpression at the BBB level [41]; however, it is important to note that the GBM tropism was way more evident in vitro compared to the in vivo results, where NPs were also found in other brain tissues [42]. In another study published in 2021, the same team demonstrated that the silica SIWV-NPs could penetrate a human GBM tumoroid more efficiently compared to the same nanoformulation functionalized with the RGD peptide or PEGylation [43]. Similarly to the previous work, specificity resulted particularly enhanced in vitro, but this behavior was reduced in vivo, where silica SIWV-NPs demonstrated preferred accumulation in the GBM xenograft, but they were abundantly found in other brain tissues. While this innovative system holds very high potential for GBM treatment, it is crucial to find novel ligands with significant GBM specificity in order to reduce side-effects on healthy tissues.
Another potential anti-GBM therapy based on peptide-targeted NPs was developed by Hsieh and coworkers. The peptide CTCE9908 was previously used for specific drug delivery in different types of cancers due to its behavior as agonist for the chemokine receptor CXCR4, but never for GBM [44]. In this study, the CTCE9908 was conjugated on lipid calcium phosphate nanocarriers loaded with siRNA, demonstrating effective and efficient delivery of the siRNA both in GBM cell cultures and in vivo in orthotopic GBM mice. This increased uptake resulted in PD-1 ligand gene silencing; however, although there was improved delivery of CTCE9908-NPs in the tumor area compared to nontargeted nanocarriers, a notable biodistribution was found in the kidney and in the liver, which is synonymous to a rapid clearance from circulation and might underline poor GBM specificity [45].

4. Dual Targeting of the BBB and GBM Cells

As evidenced by the results presented in the previous section, GBM targeting moieties are crucial to allow for GBM specificity, but their efficacy is often limited in vivo due to the complexity of the biological system, as nanocarriers need to both cross the BBB and be specific toward GBM. Thus, in recent years, two trends have emerged. The first one is the combination of two known ligands, one for BBB crossing and the other for GBM. The second one, instead, involves the functionalization of NPs with a novel single ligand that realizes both BBB crossing and tumor targeting, relying on the expression of common receptors on endothelial and tumoral cells. Here, we discuss some of the work published after 2019.
A sequential targeting approach was proposed by Kuo’s group by developing PEGylated poly(ethylene glycol)–poly(ε-caprolactone) polymeric NPs engineered with wheat germ agglutinin and FA to target the BBB and GBM, respectively (WF-NPs), and loaded with the anticancer drugs etoposide, carmustine, or doxorubicin. The authors demonstrated that the double-targeted WF-NPs showed increased BBB penetration and GBM internalization in vitro, highlighting a synergistic effect of the two ligands that resulted in increased accumulation compared to the single ligands and the nontargeted NPs [46]. While interesting, this study was only limited to in vitro experiments, and an in-depth analysis on the in vivo biodistribution of these NPs is necessary to assess the potential of this strategy to overcome the BBB and specifically target GBM cells.
Wu et al. carried out an interesting study on the surface modification of cabazitaxel-loaded PEGylated nanocrystal liposomes composed of hydrogenated soy phosphatidylcholine and mPEG2000-DSPE. The D-peptide (VAP), agonist of a receptor which is overexpressed both at the BBTB and on GBM cells, and p-hydroxybenzoic acid (p-HA), known to be BBB-permeable, were linked to the surface of liposomes, generating a “Y-shaped” targeting moiety. The double-targeted liposomes resulted in enhanced BBB crossing and GBM spheroid accumulation in vitro compared to both the free drug and the monofunctionalized NPs. When dosed to orthotopic GBM mice, double-targeted NPs were able to significantly cross the BBB, exerting antiangiogenic and apoptotic effects after drug release; however, the authors did not specifically address the GBM specificity, as they did not quantify the amount of nanocarrier or drug in the tumor, but only in the whole brain [47]. This is a crucial point that must be addressed in these types of studies, as it is fundamental to determine the actual specificity of the proposed system.
The popular RGD peptide was used by Hu et al. to formulate tumor-targeted self-assembling nanocarriers. The team synthesized a pH-sensitive doxorubicin prodrug and a derivative of the RGD peptide, which were able to spontaneously form nanocarriers when combined. Specifically, the combination of these two molecules in different ratios led to the selective formation of vesicles or micelles, both showing selective biodistribution to GBM in vivo, with improved anticancer therapy following the release of doxorubicin [48]. More recently, Qi and coworkers combined the targeting ability of the popular RGD peptide and Lf into a drug-loaded DSPE-PEG liposomal nanosystem, finding an optimal RGD:Lf ratio of 3.75:1. The aim was to enhance the performance of the well-established single-ligand-decorated NPs thanks to the ability to bind both the Lf receptor and integrin αVβ3. The expected synergistic effect of RGD and Lf was confirmed by improved transport across an in vitro BBB model and deeper penetration into tumor spheroids compared to the formulations with the single ligands. The brain- and tumor-targeting ability was also demonstrated in vivo, where RGD-Lf liposome administration resulted in an increased chemotherapeutic efficacy of docetaxel, inducing a reduction in the tumor mass, as well as increases in apoptosis and the survival rate of mice [49].
While dual-targeting systems using two known ligands show great promise, the possibility of using a single ligand for dual targeting has also been investigated. With the aim of exploiting the upregulation of LRP1 receptor in both BBB and GBM cells, a novel targeting stapled peptide was synthesized and conjugated onto PEGylated polylactic acid polymeric micelles by Ruan et al. The peptide ST-RAP12, derived from the RAP protein, had a higher LRP1-binding affinity and uptake in endothelial and GBM cells than the non-stapled form. While ST-RAP12 micelles could not reach the brain statistically more than RAP12 micelles, both surface-modified nanocarriers demonstrated a significant increase in brain and GBM accumulation compared to nontargeted micelles. The potential clinical application of this new peptide-modified NPs was supported by an increased survival rate, apoptosis, and angiogenesis inhibition after administration of ST-RAP12 micelles loaded with paclitaxel in glioma-bearing mice [50]. The possibility to use a single ligand for dual targeting represents a great advantage from a technological point of view, as it allows the use of a simpler system, with fewer formulation steps and, consequently, easier characterization and cost-effective production.
Along the same lines, Li et al. synthetized DSPE–mPEG micelles decorated with a peptidic ligand (DATP) to interact with the neuropeptide Y receptor Y1. DATP was demonstrated to not only promote concomitantly BBB transcytosis and glioma targeting of micelles, but also to improve these processes compared to the functionalization with ATP in the L configuration (LATP). The in vitro data highlighted an increased BBB crossing compared to Angiopep-2- and RVG29-modified micelles, two well-known targeting moieties. Furthermore, the favorable properties of DATP-micelles improved the application of photothermal therapy in GBM mice, resulting in a greater therapeutic effect [51]. Interestingly, the authors claimed that ligand density led to different amounts of uptake in GBM cells; however, the ligand density was not quantified, and they only referred to the amount of ligand used for different formulations. This is a crucial point, as discussed in detail in Section 6, that demonstrates the importance of a thorough characterization of the surface of targeted NPs, as well as the lack of techniques available to succeed in this aim.
Starting from a viral glycoprotein, a derivative peptide of 15 amino acids called RVG15 was recently demonstrated to be a novel and efficient targeting peptide for BBB crossing, since it is able to bind the nicotinic acetylcholine receptor [52]. This peptide was conjugated onto the surface of soybean phosphatidylcholine, cholesterol, DSPE–PEG liposomes loaded with cholesterol-conjugated paclitaxel, providing BBB permeability and selectivity toward GBM cells [53]. Indeed, RVG15 liposomes improved paclitaxel delivery and efficacy compared to nontargeted nanosystems; not only could RVG15 liposomes accumulate in the brain 1 h after administration in vivo, but they were also abundantly present in tumors. The interference with GBM progression was ascertained through the inhibition of tumor growth and metastases formation. Unfortunately, the authors did not perform statistical measurements to assess the prevalence of liposomes in the tumor tissues compared to surrounding healthy tissues in the brain, which is a critical evaluation to determine the GBM specificity of this interesting dual ligand.
Another ligand synthesized for two-in-one targeting purposes was the tetrapeptide mnRwr (mn), which should interact with the integrin αVβ3. In order to evaluate its performance, the authors compared mn with the well-studied c(RGDyK) peptide. BBTB transcytosis and tumor spheroid penetration were comparable for the two ligands. In contrast, mn-liposomes exhibited improved biodistribution and pharmacokinetics in vivo, resulting in a greater accumulation in the tumor site of xenograft-injected mice, as well as increased therapeutic efficacy against GBM [54].
Another innovative strategy was proposed by Huo et al. Exploiting the high Tf concentration in the blood and the upregulation of Tf receptors in both brain endothelial and GBM cells, covalent organic framework nanospheres, composed of PEI-coated mesoporous silica nanospheres, were decorated with the peptide T10, which has a high affinity to Tf. After systemic administration, a Tf-based corona on the surface of NPs was generated in the bloodstream due to the strong Tf-T10 association, allowing for the specific endogenous interaction of Tf-coated NPs with Tf receptors. The BBB- and GBM-targeting ability was demonstrated after in vivo administration, corroborated by a decrease in IC50 value and a prolonged mice survival rate compared to the free drug, the nonmodified formulation, and the commercial liposomal doxorubicin Caelyx® (Baxter Holding B.V. Utrecht, The Netherlands). Interestingly, T10-modified NPs presented a prolonged release profile for doxorubicin and, most importantly, did not involve the action of the efflux pumps [55]. This study represents an interesting and innovative approach toward tumor targeting, as it exploits endogenous mechanisms by directing the formation of the protein corona to allow for a more specific and nonimmunogenic brain and GBM targeting.
These studies displayed groundbreaking results in the field by simultaneously overcoming what are considered two of the major barriers to GBM delivery and treatment. Table 1 summarizes the selected nanosystems. While showing improved results, the characterization and in vivo analyses are still critical missing steps in their path toward a true GBM treatment, which becomes even more complicated in animal models with the biological differences in the tumor vicinity, as discussed in the next section.

5. Targeting the Tumor Microenvironment (TME)

So far in this review, novel strategies to target the BBB or GBM cells have been discussed; however, there is another option that has recently entered into play to try to combat cancer. The TME has taken the spotlight in recent years as a promising target as it plays a pivotal role in tumor growth and in the formation of metastasis. Indeed, it concentrates solutes and cells that can trigger the epithelial-to-mesenchymal transition of epithelial cells, which can ultimately lead to the formation of metastasis [62]. All cancers host a TME which could vary between patients and cancer types/progression, but the study of the differences and intricacies between each different TME is still in its infancy and not known well enough to make distinctions. Therefore, targeting the TME cannot be considered GBM-specific, but will surely play a critical role in the improved treatment of GBM, as well as other cancer types. Moreover, in recent years, the possibility of studying TME characteristics has been supported by the big improvements in microfluidic-based techniques; with this technique, it is possible to regulate the interstitial flow, rheological properties, and nutrient and oxygen concentration, finely mimicking the physiological characteristics of the TME [63]. This allows us to have more accurate models with faster and more accurate analyses that will translate into more effective systems. In order to exploit the characteristics of the TME to reduce tumor growth, NPs have been shown to be particularly efficient, thanks to their specific targeting potential [64]. Over the last decade, the number of studies on the TME has exploded; in 2021, the number of review articles on this topic seemed to outnumber the research articles, allowing readers to look up and understand the idea of the TME and how it can be taken advantage of for enhanced chemotherapy. Therefore, in this section, we only give an update on the most recent studies (2022) regarding TME targeting and how they are advancing potential therapeutic strategies.
One of the oldest, most investigated, and well-known ways to target tumors linked to the TME characteristics is the enhanced permeation and retention effect (EPR). The EPR effect was first observed in the 1990s, and it has led the production of nanomedicines with a size around 200–300 nm to facilitate extravasation and accumulation at tumor sites [65]; however, recent considerations that the EPR effect is minimized in humans compared to rodents, as well as its heterogeneity from patient to patient or disease to disease, have made it more evident that the EPR effect alone cannot ensure the specific accumulation of NPs in tumors or successful translation to a clinical setting [66,67,68,69,70]. For this reason, NPs have been further engineered to enhance their efficacy [71]. Targeting moieties toward TME components can be used to enhance the efficacy of NPs that are already present and trapped near the cancer cells due to the EPR effect. For example, to further facilitate the penetration of NPs in the matrix of solid tumors, their surface can be decorated with ligands that target cells of the extracellular matrix (ECM), such as pericytes, endothelial cells, or cancer associated fibroblasts [72,73,74]. To target the ECM, several studies have investigated the use of collagenase or hyaluronic acid to coat chemotherapeutic-loaded NPs. While the use of hyaluronic acid can promote extravasation following interaction with the CD44 receptor [75], the use of collagenase was recently demonstrated by Shukla et al. to improve ECM penetration and accumulation at the tumor site of biocompatible albumin NPs loaded with gemcitabine. The efficacy of these coated NPs was tested in tumor spheroids, demonstrating more efficient delivery of gemcitabine to cancer cells compared to NPs without collagenase [56].
The high concentration of growth factors that occurs at tumor sites has also been investigated for NP targeting. In particular, the epidermal growth factor (EGF) is one of the most researched moieties with which to decorate NPs for cancer therapy. While most of the previous strategies to target the EGF receptor were based on the use of monoclonal antibodies, such as the anti-EGF receptor monoclonal antibody cetuximab, on the market as Erbitux® (Merck Healthcare KGaA, Darmstadt, Germany), the endogenous EGF molecule (6 kDa) was recently proposed as a smaller and easier molecule to use as a targeting ligand to be conjugated on the surface of NPs [76,77]. Skóra et al. confirmed the possibility of selectively delivering silver NPs to cancer cells by loading them into EGF-decorated liposomes that were able to induce toxicity in cancer cells in vitro [57]. Growth factors and cytokines are also the target of the research published by Zhang et al. in June 2022; here, the authors validated the use of gold NPs to inhibit the tumorigenic phenotype and tumor growth both in vitro and in vivo, highlighting the crucial role of soluble hormones in the TME in tumor progression [78]. Another hormone that has been used to target cancer cells is the vascular endothelial growth factor (VEGF). VEGF receptors are highly overexpressed in most solid tumors to allow for a fast growth of cancer cells. Thus, using VEGF as a targeting moiety for the TME has represented one of the most promising targeting strategies for a long time [79]. Unfortunately, it has been evidenced that tumors are often insensitive to antiangiogenic treatments with anti-VEGF therapies. This phenomenon has been mostly observed in highly innervated tumors, revealing the crucial role of tumor-associated nerves (TANs) in the efficacy of anticancer treatments [80,81]. Treating tumors with VEGF-targeted NPs could induce a response from TANs with production of neurosignals that might restore the angiogenic behavior of cancer cells, canceling the efficacy of the treatment [82]. This crosstalk highlights the complexity of the TME and the need for more in-depth studies of the conditions that can occur in tumor sites, together with the need of novel systems that can address more than one target with a single administration.
Once in the TME through the EPR effect or active targeting, NPs can also be designed to take advantage of some of the characteristics of the TME by passive targeting. The TME is characterized by acidic, hypoxic, and oxidative conditions, which have been widely addressed as targets for stimulus-responsive NPs [83,84,85,86,87]. In January 2022, Wei et al. proposed a system for diagnostic purposes where a pH-sensitive moiety was conjugated on the surface of Fe3O4-based probes. This moiety was demonstrated to be able to interact with cancer cell membrane at acidic pH, highlighting the presence of cancer cells by PET/MR imaging [88]. Koo et al. optimized a system based on copper–iron peroxide NPs which can degrade in acidic conditions. This system, designed for chemodynamic therapy of solid tumors, produced oxygen, relieving the hypoxic state, and released Fe ions, which improved MR imaging as a contrast enhancement [89]. It is also important to underline that inorganic NPs, while demonstrating impressive results in the treatment and possibility for more precise or early-stage diagnostics, are also known to have issues with toxicity or the inability to be cleared from the body, which might represent a limiting step in the transition toward the clinic and that must be taken into consideration when designing novel NP-based strategies [90,91]. Overall, the pH or hypoxic reactive lipidic or polymeric NPs were unfortunately less published this year compared to previous years, probably due to the lower specificity of this type of targeting than receptor-mediated targeting.
As previously mentioned, activating the immune system to recognize tumors became a novel line of research against tumor cells in the last decade; however, this approach often resulted ineffective, as the TME can block immune recognition [92]. Over the last year, several studies still tried to overcome this barrier by using NPs to modulate the immune system and activate it against tumoral cells [93]. For example, NPs have been used to deliver siRNA or mRNA to cancer cells to modulate the expression of specific proteins, such as Her2. Downregulation of Her2 recently resulted highly effective in promoting cell death in breast cancer models, exhibiting a synergistic effect with taxane [94]; however, Her2 reduction leads to the production of cytokines that might promote tumor recurrence [95]. Moreover, cancer cells can overexpress antiphagocytic molecules such as CD47, ultimately inhibiting the inflammatory response and facilitating immune escape of cancer cells [96]. Therefore, one of the most recent and innovative approaches is to act on the TME in order to block the inhibition of the immune response, to finally allow the activation of immune cells towards tumors. This strategy, called immune checkpoint blockade (ICB) therapy, is now on the rise, with several reviews highlighting its potential; however, most of them focused on the lack of research studies, due to the inherent difficulty and complexity of this kind of approach [72,73,97,98]. Modulation of the TME for immunotherapy and ICB therapy can be performed by addressing soluble cytokines, enzymes, and other proteins e.g., CTLA-4 protein and PD-1 receptor, as well as cells such as T-cells, cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs).
Macrophages in the TME usually have an M2 phenotype, meaning that they have an inherent anti-inflammatory activity. Therapies targeted to TAMs can include the repolarization of anti-inflammatory M2 macrophages to the proinflammatory M1 phenotype and inhibition or elimination of activated TAMs. Feng et al. recently proposed a drug delivery system by engineering an albumin-based NP with scavenger receptor A and SPARC protein which targeted both TAMs and tumoral cells, respectively. The group demonstrated that these targeted NPs were able to eliminate TAMs from the TME, leading to an increase in inflammatory cytokines that would ultimately promote immune recognition of tumoral cells [58]. Wang et al. recently developed ultrasmall Cu2−xSe inorganic NPs coated with a biomimetic cell membrane for ICB therapy of GBM. These NPs were able to polarize the macrophages to an M1 phenotype and decrease the expression of the PD-1 ligand. Moreover, after treatment with their NPs, they noticed an increase in memory T cells in the spleen, which could possibly prevent GBM recurrence through the protection by the immune response [59]. Similar results were obtained by Alghamri et al. with synthetic protein NPs modified with a transcytotic peptide and loaded with an inhibitor of the CXCL12/CXCR4 signaling pathway, which is associated with tumor growth and progression. This NP drug delivery system, when coupled with radiation therapy, was able to block the signaling cascade, resulting in induced immunological cell death and the proliferation of memory cells, lowering the risk of recurrences in a GBM mouse model [99].
Another interesting approach is to target regulatory B cells (Bregs), which have been recently proposed to be responsible for the attenuation of the antitumoral immune response, as they can upregulate the production of interleukin 10 and the PD-1 ligand [100]. This strategy was investigated by Shen et al. using lipid-protamine NPs to load plasmid DNA encoding a CXCL13 trap to reduce the differentiation of Bregs. The DOBP- and cholesterol-based lipid–protamine–DNA (LPD) NPs developed by the team successfully reduced tumor growth, showing the pivotal role of Bregs in tumor progression [101].
To enhance the efficacy of any treatment targeted to the TME, some interesting approaches were published this year using dual systems that can exploit two of the cited targeting strategies with a single nanosystem. To this end, Ding et al. proposed the formulation of an alginate hydrogel loaded with two separate nanosystems: (1) porphyrin-modified Fe3O4 NPs, sensitive to near infrared light for photodynamic therapy and able to produce reactive oxygen species (ROS); (2) albumin-based NPs, conjugated to an antibody against the PD-1 ligand via a ROS-sensitive linker that could be cleaved in the TME containing high levels of ROS. This complex system was demonstrated to have high therapeutic efficacy by combining the positive effects of photodynamic therapy, ROS induction, and immunomodulation [60]. Another dual strategy system was presented by Sun et al., where platinum NPs stabilized with phenylboronic acid were co-assembled with dextran-coated NPs loaded with sotuletinib. The linkage between the two was a pH-sensitive borate ester, which allowed the system to disassemble at the acidic pH of the TME. After disassembly, platinum NPs were able to penetrate in the most internal layers of the tumors and induce cell death due to the release of Pt2+, while sotuletinib was able to induce the depletion of TAMs, reversing the immunosuppressive microenvironment [61].
The TME has become a hot topic in the research field, and targeting its variable components has become a huge area of research which holds great promise for understanding and better controlling the way in which a tumor survives, grows, and re-emerges after treatment. While it is present in all tumors, a better understanding the TME will play an even more critical role in treating the more difficult-to-reach tumors, such as GBM. With the help of nanotechnologies, utilizing these differences around the tumor masses could help improve early-stage diagnostics and late-stage treatments; however, we are still very naïve about the interplay and complexity of the TME and how to take advantage of it to its full potential. Reviews continue to describe the ideas and key points of the TME, but a huge amount of research is needed in order to optimize a system that combines its numerous components for a complete and therapeutically relevant system.

6. Surface Characterization

As discussed in previous sections, surface functionalizations of NPs are key to achieve targeted delivery of chemotherapeutics, especially for GBM and brain tumors. Unfortunately, the characterization of the surface of NPs is often overlooked, due to the lack or high cost of the techniques used, as well as the difficulty of specifically looking at only the surface and not the whole nanosystem; however, surface characterization is a crucial step of the development of a nano-drug delivery system. From a biological point of view, the surface is responsible for the interaction with the target, and its characteristics can influence the composition of the protein corona, the clearance time, and the toxicity of the system. From a technological perspective, it is important to understand the surface composition of NPs in order to maximize the efficiency of the engineering process, while also ensuring the formulation of reproducible NPs that can be translated into an industrial and clinical setting.
Paramount parameters to describe NPs are size, surface charge, and shape, along with porosity, surface chemistry, crystalline structure, purity, drug loading, hydrophobicity, etc. Currently, several technologies are available for the characterization of NPs [102,103], with the choice of the method depending on the sample characteristics and the final application [104]. More often than not, it is difficult to achieve a thorough characterization of a NP system due to several challenges, the most neglected aspect being the surface characterization [105]. The lack of accurate characterization of NP surface could be one of the reasons for undesired biodistribution in off-target organs and/or rapid clearance. Indeed, the fate of the NPs in the complex biological environment after administration both in vitro and in vivo is determined by the interaction of the surface with the various biological components. It is well known that surface chemistry has implications in solubility, stability, biocompatibility, and pharmacokinetics of NPs, along with influencing the internalization and the ultimate efficacy of the nanosystem [106]. One of the most important aspects to be determined when characterizing an NP system is the ligand density. Depending on ligand density, two trends of cellular uptake can be identified for a single ligand: (1) optimum density with a plateau, and (2) optimum density with a maximum [107]. The identification of a behavior over the other would lead to a more conscious interpretation of biological results. Moreover, this would be pivotal information that could greatly improve the design and optimization of NPs with the optimal density of ligand to exert its targeting activity, as well as reduce the waste of materials during the formulation process.
Unfortunately, many papers on NPs for GBM treatment lack an adequate surface analysis before moving on to animal studies. If present, this could help prevent therapeutic failure, or it would allow higher NP accumulation at the tumor site. The surface characterization can be carried out using both classical and advanced tools that, however, do not allow a single-level analysis, but only in bulk [108]. Because of the complexity of NPs and the information obtained from each technology, the preferable method to analyze the surface of NPs would be a multidisciplinary approach. Different techniques should be used to extract complementary information with the purpose of having a snapshot of the system that is as detailed as possible before proceeding with further in vitro and in vivo tests. An overview of analytical tools used to obtain information about the surface features of NPs formulated for GBM therapy is shown in Figure 2.

6.1. Electrophoretic Light Scattering

One of the two most commonly studied NP parameters throughout the literature is the surface charge (the other being hydrodynamic diameter). By exploiting the electrophoretic light scattering, the zeta potential can be measured in an easy and fast way. This value not only provides information on the surface charge of NPs in suspension, but can also help predict the stability of the formulation, as colloids with a zeta potential close to neutrality often tend to form aggregates during storage [109,110]. Moreover, the zeta potential is a crucial parameter when considering the toxicity of nanosystems, as it is well known that cationic NPs tend to be quickly cleared from the bloodstream and induce toxicity after administration [111]. In several studies, surface charge measurements at different steps of the formulation are used as a control for the effective functionalization of NPs; however, this parameter often does not give an indication of the amount of ligand. In fact, a shift in the zeta potential is generally considered significant only for huge changes, and its use as an indicator for successful modification is often limited to the case of coated NPs [112,113]. On the contrary, ligands such as peptides, small molecules, or antibodies hardly ever induce a significant shift in the zeta potential, due to the small amount added to the formulations. Moreover, shifts in the zeta potential are only qualitative, and they do not provide information about the amount of ligand or coating molecules present, nor whether they are chemically conjugated or nonspecifically adsorbed on the surface, or whether they are present in the correct 3D orientation, which is a critical aspect of antibody-based targeting strategies. Unfortunately, quantification of the targeting ligands on the surface of NPs is often a long and difficult task that requires multiple techniques.

6.2. Fourier-Transform Infrared Spectroscopy

One of the most widely used techniques for assessing the functionalization of anti-GBM NPs is Fourier-transform infrared spectroscopy (FT-IR). FT-IR is a vibrational, nondestructive method that provides structural and chemical information of a sample by using radiation in the infrared field. The positions and the intensity of the bands in the IR spectrum allow a qualitative analysis, indicating the presence of the ligand interacting with the NP components. Over the years, FT-IR has proven to be a versatile and powerful tool for material characterization [114,115,116,117]. In [118,119,120,121,122,123], the effective coupling of the targeting moiety was evaluated through the direct spectral comparison among the whole NP formulation, the nontargeted one, and/or the ligand. The attribution of specific peaks made it possible to identify the contribution of the ligand in the IR spectrum of NPs. Unfortunately, other components in the NP or the drug itself might interfere with the signals coming from the ligand; thus, this technique does not give quantitative information about the NP surface, but only qualitative. Another major limitation of this technique for surface characterization lies in the fact that the IR light might penetrate into the matrix of the NP [124]. This is a crucial aspect especially for pre-conjugated ligands that might end up being entrapped in the matrix but not displayed on the surface, e.g., in the case of pre-modified lipids that during the formation of a liposome could be faced either in the internal phase or onto the surface. In this case, FT-IR would give positive results, but it would not take into account the real 3D disposition of the ligand.

6.3. Raman Spectroscopy

Complementary to IR, Raman spectroscopy (RS) relies upon the inelastic scattering of light and provides a molecular fingerprint of the analyzed target. Thanks to the possibility of obtaining qualitative and quantitative information with a low amount of sample in a few minutes through label-free analysis, RS and surface-enhanced Raman spectroscopy have found application in the pharmaceutical and biological field [125,126,127]. Although RS offers many advantages, its application for NP characterization is not widely exploited. For example, Sahli’s group verified the formation of a complex among PEGylated gold NPs, temozolomide, and gemcitabine via RS, where the decrease in the peak intensity at 1292 cm−1 in the presence of the drugs denoted the reaction between COO and NH3+ groups indicating successful conjugation [128]. Similarly, RS was also used to assess the bond of cisplatin to gold nanopeanuts developed as a dual therapeutic platform against GBM [129]. Difficulties in the use of this technique lie in the complexity of Raman spectra interpretation since the technique gives an overview of the presence, concentration, and interaction of all the molecules present in the sample; however, the comparison between formulations that differ in only one component can help identify the contribution of the molecule of interest in the Raman spectrum. Lastly, similar to FT-IR, when NPs are functionalized prior to formulation, the possible penetration of the laser in depth of the NPs can generate signals not specifically related to the ligand present on the surface, but also inside the matrix.

6.4. Nuclear Magnetic Resonance Spectroscopy

Nuclear magnetic resonance spectroscopy, also called NMR, is a robust and informative platform to obtain structural information about molecules. Taking advantage of nuclear spin variation, NMR is able to identify the type and the abundance of isotopes in both liquid and solid samples. Therefore NMR can provide insight into surface features and modifications [130,131,132]. For example, NMR spectra were used to confirm the successful conjugation of Angiopep-2 on polymers for NP formulation either by the lack of the N-hydroxysuccinimide peak or by the presence of Angiopep-2 peaks on the polymer spectrum [133,134]. Another approach that can be followed is the attribution of the chemical shift. Similarly, Minaeia et al. ascertained the binding of FA to polymeric NPs through the identification of protons related to the grafted molecule, while Vilella et al. confirmed the conjugation of a BBB-crossing peptide to PLGA to be used to formulate brain-targeted NPs [135,136]. NMR spectroscopy can be used to quantify the conjugation yield of a ligand, indicating to which extent the material to be formulated into NPs has been linked to the targeting moiety. However, similarly to the techniques previously discussed, this information is not indicative of the actual presence of the targeting ligand on the surface of the system.

6.5. Surface Plasmon Resonance Spectroscopy

In addition to the quantification of the ligand present on the surface of a nanosystem, it is crucial to verify whether it still retains the ability to bind its receptor after coupling with NPs; to this aim, the nanoformulation can be analyzed by surface plasmon resonance spectroscopy (SPR). SPR is a highly sensitive optical technique used to study the binding affinity and kinetics between an analyte in solution and ligands spotted on a gold chip in real time, without the need for any labels. Since the evanescent wave propagates for about 200 nm from the chip surface, SPR is a suitable tool for the analysis of nanosystems [137,138,139]. Using SPR, Gries et al. compared the binding kinetics between the free peptide KDKPPR and the peptide-based NPs against GBM, not only demonstrating that the coupling did not impair the activity of the ligand, but also finding a higher KD after the coupling of the targeting moiety with NPs [140]. The same rationale was used in another study, in which the Tf binding efficiency after coupling to chitosan NPs was also quantified through inductively coupled plasma optical emission spectroscopy [141]; however, the authors did not exploit the multiplexing characteristic of SPR. Indeed, no receptor apart from the Tf receptor was immobilized on the chip in order to evaluate the specificity of the binding, resulting in incomplete information, which might be useful to predict the efficacy of this nanosystem. Notwithstanding the useful information that can be obtained from sensorgrams about the optimization of NP and their potential in vitro and in vivo efficacy, SPR is not widely present in laboratories. Limitations may be due to the cost of gold chips on which ligands are spotted, along with the optimization of the surface chemistry of the chip for ligand immobilization, necessary to obtain reproducible substrates with desired characteristics.

6.6. X-ray Photoelectron Spectroscopy

Among the available techniques, X-ray photoelectron spectroscopy (XPS) represents a valuable tool for surface characterization. According to the photoelectron emission process, XPS reveals the elemental composition of the outermost layer (within 10 nm) of the material after the irradiation of X-rays; however, a possible limitation is the ultrahigh-vacuum condition necessary for the analysis, which might be incompatible with the material used for the formulations. Over the years, this sensitive electron spectroscopy has been used for the surface characterization of almost every material [142]. Since all elements except H and He can be detected, it is possible to follow the variation in NP surface composition before and after the functionalization procedure. For example, the variation of C concentration and its signal reflected the formation of the metal–organic framework layer on Fe3O4 NPs [143]. With the same aim, Venditti et al. analyzed Au-NPs coated with two different thiol-bearing moieties that may serve as linkers for future ligands. The XPS analysis was able to distinguish the signal of the gold atoms belonging to the NP core, from that relative to the surface atoms. This allowed measuring that around 20% of the gold atoms on the NP surface were covalently bonded with sulfur of the two moieties [144]. One of the major limitations to the use of this technique for the characterization of surface-modified NPs is that this is particularly efficient in the determination of metal ions; in general, the more the material of the NP and the ligand are chemically different, the more this technique is useful and efficient. Unfortunately, organic NPs are often functionalized with organic moieties, e.g., liposomes, PLGA, or chitosan NPs, conjugated with peptides, antibodies, or small molecules. In these cases, the use of XPS is greatly limited by the high elemental similarity of the components, which hampers the possibility of using this method for surface characterization.

6.7. Thermogravimetric Analysis

Thermogravimetric analysis (TGA) is a technique that correlates the mass of a sample with the increasing temperature, and it is a commonly used technique for a quantitative measurement of a sample with application in the study of nanomaterial structure [145,146,147], as changes in the specimen mass indicate the loss or transformation of one or more components, particularly related to the adsorption or desorption of coating layers; however, no indication of the nature of the material lost is given. Exploiting TGA, Świętek et al. studied the formation of multiple coatings on the surface of silica–Fe2O3 NPs, using tannic acid and chitosan. While this technique allowed authors to measure the amount of water present in the sample, which could be related to the number of hydrophilic components in the coating layers, it was necessary to couple this analysis with other techniques to have an overview of the sample. The formation of the layers was evaluated in parallel with FT-IR and zeta potential, and the presence and intensity of specific IR peaks and the variation in surface charge were considered as markers of the successful coating procedure [148]. The analysis of the coating formation was also verified by Shahein et al. after the formulation of mesoporous silica NPs covered with two different shells: one made of chitosan–stearic acid (CS) and the other made of whey protein–gum Arabic (WA). In this study, the amount of CS and WA forming the coating was deducted from the mass variation between the nanoformulation with and without the coating layer [149]. While interesting, this approach requires the instrument to have a very high resolution, so as to measure small variations in the case of NPs where the amount of material used for the coating is small.

6.8. Scanning Electron Microscopy and Transmission Electron Microscopy

Scanning electron microscopy (SEM) and Transmission electron microscopy (TEM) are well-established tools for imaging in the field of material characterization. SEM creates a 3D image by detecting the reflected electrons, whereas TEM uses the transmitted electrons to provide a 2D image. As a result, SEM can help yield more detailed information about the sample surface while TEM can provide information about the internal structure. For both techniques, the high resolution enables visualizing specimens at the nanometer level; hence, they have found application in NP characterization by allowing the study of shape and size [150,151,152]. Numerous studies have used SEM and TEM images to better characterize and understand the influence of surface functionalization on morphology, porosity, and surface area of nanocarriers. Ghaferi et al. carried out SEM analysis at different stages of the synthesis and formulation of drug-loaded PEGylated liposomes. Images, however, did not provide evidence of surface changes, but only of the retention of the spherical shape and the monodispersity after the decoration with PEG molecules [153]. Similar information was obtained by Pulvirenti and coworkers analyzing Fe3O4 NPs grafted with a metal–organic framework layer (Figure 3a) [143]. Indeed, even if SEM is considered a valuable technique for evaluating the features of material surfaces, no differences in texture following the modification were observed in the studies mentioned above. On the other hand, TEM images of biomimetic NPs developed by Zhang and coworkers highlighted a core–shell architecture after coating an siRNA-based complex with melanoma cell membrane (Figure 3b) [154]. In another example, differences in the TEM images of hyaluronic acid NPs revealed a structural modification after conjugation of Angiopep-2 on their surface (Figure 3c) [155]. While both of these techniques are fundamental and critical for demonstrating a full and intricate characterization of reproducible and stable NPs, authors often miss the description of the differences pre and post modification. This major flaw can be noted not only when commenting on coated NPs, but even more when discussing the characterization of NPs modified with small ligands, where the characterization is even more difficult. A general idea of the effects during the modification process can be determined, but the specific features in terms of location, quantity, or distribution of the ligands are still outside of the resolution range of the general techniques, and they must be improved to have a true knowledge of how these variables could affect the biological and pharmaceutical effects of NPs.

6.9. Other Methods

A broadly used technique to characterize NPs is UV/Vis spectroscopy. When applied to surface-modified NPs, this method can help understand if the functionalization has been successfully achieved, by evaluating the absorbance change or band shifting occurring after surface modification. These results are particularly evident in the case of inorganic NPs. In this view, Wang et al. developed maleimide PEG-modified CuS NPs with the aim of combining hyperthermia with immunotherapy for cancer therapy. The absorption spectra of PEG-coated CuS NPs were analyzed using a UV/Vis/NIR spectrophotometer, and the spectra of PEG-functionalized NPs showed a different intensity of absorbance compared to the nonfunctionalized NPs, indicating that the surface functionalization was successful [156]. Similarly, Lesiak and coworkers performed a surface study of iron oxide NPs functionalized with different biocompatible molecules adsorbed on the surface, such as succinic acid, oxalic acid, L-arginine, citric acid, and glutamic acid. They demonstrated that the surface adsorption of these organic molecules had an impact on the UV/Vis spectra of iron oxide NPs, producing a change in both the intensity and the wavelength of absorbance of the modified NPs compared to the nonmodified ones [157]. Those two studies, as well as others in the recent literature, indicate that the UV/Vis characterization of surface-modified NPs is a valid tool in the characterization of inorganic NPs [158,159,160]; however, many researchers demonstrated that organic surface-modified NPs can also be characterized using this technique. This is possible by exploiting the ability of some organic molecules to react with UV/Vis-detectable substances. For example, the amounts of protein- and peptide-based ligands can sometimes be assessed by specific colorimetric assays. Due to the low sample volume required and the simplicity, the BCA assay is the most widely used. This is an indirect ligand quantification which involves measuring the free ligand at the wavelength of 562 nm, meaning that the separation of formed NPs from nonconjugated ligand is required [46,161,162,163]. The purification is often a limiting step during formulation of NPs, and it requires thorough optimization to ensure that the method chosen does not cause NP destabilization and ligand detachment, which would interfere with ligand quantification. Unfortunately, in addition to the purification, the BCA assay is only applicable to a limited class of ligands, and must be properly optimized for each molecule.
An interesting strategy is the combination of size-exclusion chromatography and ultrahigh-performance liquid-chromatography as a novel single-step methodology for ligand separation and quantification. With this strategy, Gazaille et al. could separate lipid nanocapsules decorated with a tubulin-binding peptide from the unconjugated ligand, thanks to this size-exclusion chromatography step. Liquid chromatography was consequently used to quantify the amount of ligand on the surface [164]. Although this approach results very attractive by combining a purification and quantification method, it is important to note that this type of separation is often only possible for water-soluble ligands, as size exclusion gels are often incompatible with elution in solvents, limiting the number of ligands that can be analyzed. Moreover, analysis via liquid chromatography often relies on UV or fluorescence detectors, and some ligands might need an additional derivatization step to be visualized, thus complicating the quantification protocol.

7. Conclusions

To date, the multidisciplinary therapeutic approach for GBM has not led to the expected effects needed for a functional treatment, with only slight improvements in the prognosis and life expectancy of the patients, which remains very poor. Thanks to several advantages, NPs have taken the spotlight as novel and more elegant methods to create therapeutics to fulfill the unmet clinical needs against hard-to-treat diseases. With these successes, scientists have exploited these advantages to try to improve GBM treatments as well; however, the fast growth rate, invasiveness, shift in biological microenvironment, resistance to drugs, and vast variability of GBM from patient to patient have greatly hampered their effectiveness to deliver chemotherapeutics to the tumor site and create a system with potential clinical applications. For this reason, more advanced targeted NP systems and early detection options are being designed and researched each year.
Currently, the most studied method to improve GBM treatments using NP delivery systems is conjugating ligands onto the surface of the NPs in order to exploit ligand–receptor interactions to improve specific drug delivery to the GBM tumor and limit off-target effects. Over the years several types of ligands that act on different targets have been involved for this purpose; however, oftentimes, as in the case of Tf and its receptor, they are ubiquitous, and the true selectivity is not up to par. Novel and improved ligands are being sought out to be more specific, even at the early stages of disease progression.
In the case of GBM, the BBB is still considered one of the major barriers to successful chemotherapeutic delivery. Therefore, many new GBM-targeting studies do not actually look at true GBM targeting, but attempt to improve the rate of BBB crossing in order to improve GBM treatment. While this is critical for a therapeutically relevant NP against GBM, it still lacks specificity within the central nervous system and can be subject to toxicity to the local healthy cells. Therefore, other ligands that specifically target the GBM cells over healthy cells are required. These ligands would improve selectivity and safety profiles, but these studies are often limited to in vitro studies, or the potential GBM ligands lack the ability to cross the BBB. To this end, the major portion of recent research articles have explored co-targeting with both BBB and GBM ligands incorporated into a single NP system. While this will drastically improve both delivery amounts and specific toxicity against GBM while maintaining better safety profiles for healthy cells, the complexity of simultaneously formulating, characterizing, and testing the interaction of two ligands is a huge logistical barrier that needs improved methodologies and assays to properly understand and utilize the results. On the other hand, some researchers have proposed novel ligands that might be able to simultaneously promote BBB crossing and display GBM specificity; these ligands would greatly improve the therapeutic efficacy of targeted NPs by ensuring targeting with a single moiety. However, this type of investigation is still in its infancy due to the difficulty of finding GBM specific ligands that allow BBB crossing, as well as optimizing and analyzing both effects contemporaneously.
Actively targeting the BBB and tumor cells with ligands has always been the staple of novel research, but another option has recently presented itself. Researchers have discovered that many targeted systems do not work due to the ability of the tumor cells to modify their TME in order to block the immune response or degrade NP systems due to factors such as acidic pH, high ROS levels, or the presence of solutes and cells that hamper immune recognition. Therefore, instead of directly targeting the cells, the possibility to utilize these variations in the tumor environment to activate the NPs, control drug release, block tumor metastasis, and minimize the immune system inhibition is being researched. Huge numbers of reviews are being published on this topic each year, but our true experimental understanding of the TME for each type of cancer and its variability from patient to patient is still lacking. This will require vast research and comparisons between systems in order to elucidate the true nature and importance of these factors in combating GBM.
NPs targeting the BBB, tumor cells, or TME are being designed and researched in abundance for their ability to combat GBM in highly advanced in vitro or in vivo models; however, a critical component of the research is oftentimes overlooked. While aiming to combat or cure GBM, many times, the characterization of the NP vehicle is overlooked, performing the bare minimum of size, polydispersity, and surface charge characterizations. In order to improve these targeted systems, it is critical to standardize a higher level of surface characterization using various intricate techniques to determine the amount, position, distribution, and availability of these targeting ligands on the surface of the NPs, as well as to correlate those results with how these variables affect their biological and therapeutic outcomes. Unfortunately, most of the techniques used to characterize the surface of NPs do not give complete information, and their use is often limited to some classes of ligands, hampering their use for different types of ligands or nanosystems. Increasing the characterization of these NP systems will allow scientists to better compare different systems, reducing the variability in results and allowing them to determine which surface characteristics and conjugation methods are more suitable and effective to yield more efficacious and promising drug delivery systems.
GBM is an incredibly deadly disease, increased by its complexity and difficulty in procuring new effective treatments, even when using advanced NP systems. These studies demonstrate the cutting-edge research to overcome these difficulties. While each small discovery, involving new ligands and improved methodologies that are being researched each day, is critical, a combined effort will be needed to simultaneously cross the BBB express GBM cell-specific targeting, taking advantage of or inhibiting the changes in the TME in order to have a multifunctional system that will be more adept at early-stage diagnosis and more efficacious in late-stage treatments. At the same time, it is pivotal to standardize techniques that can allow for a more detailed characterization of the surface of NPs, in order to have a thorough understanding of the nanosystems being used, with the aim of improving not only therapeutic efficacy, but also reproducibility and production. All the above will require future research to improve the design, formulation, and characterization technologies in order to maximize the curative effect, as well as minimize cancer resistance and recurrence. Altogether, researchers are paving the way to help treat and improve the life expectancy and provide a ray of hope for the future patients that will be diagnosed with this horrible disease.

Author Contributions

Conceptualization, F.R., I.O. and J.T.D.; methodology, F.R. and R.C.; investigation, F.R., R.C., I.O., S.P. and J.T.D.; writing—original draft preparation, F.R., R.C., I.O. and S.P.; writing—review and editing, F.R., I.O., S.P., M.B., G.T. and J.T.D.; visualization, F.R., B.R. and R.C.; supervision, J.T.D., M.B., M.A.V. and B.R.; project administration, M.B., J.T.D., B.R. and G.T.; funding acquisition, G.T., M.A.V., M.B., B.R. and J.T.D. All authors read and agreed to the published version of the manuscript.

Funding

This research was partially funded by the Telethon GGP19113 project “Pigment epithelium-derived factor (PEDF) peptides as therapeutic agents for inherited retinal degeneration”, an IMI EU Grant “Investigating mechanisms and models predictive of accessibility of therapeutics (IM2PACT) into the brain” IMI2—Call 12, GA n. 807015 (http://im2pact.org), a Ministero degli Esteri e della Cooperazione Internazionale MAECI grant, Progetti di ricerca scientifica e tecnologica di grande rilevanza by Ministero degli Esteri, Progetti Italy–USA “Nanomedicine for blood–brain barrier (BBB) crossing in CNS oncologic pathologies”, Prot. nr. MAE00691612020-06-26, FAR Unimore Fondazione di Modena “Nano-immuno targeting per il trattamento del glioblastoma multiforme” NIT Project, GLIOSILK Project (Reference Number: EURONANOMED2019-075), and the Creutzfeldt–Jakob disease Foundation (CJDF). This study was also supported by Fondazione Regionale per la Ricerca Biomedica, research project “New frontiers of engineered nanovectors to improve treatment efficacy and safety in neurological disorders—NEVERMIND Project” (project number CP2_16/2018).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to acknowledge Chiara Lucchi and Giuseppe Biagini from the Laboratory of Experimental Epilepsy, Department of Biomedical, Metabolic, and Neural Sciences, and Chiara Chiavelli and Massimo Dominici from the Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children and Adults at the University of Modena and Reggio Emilia for their help during the revision process of this manuscript and their expertise in the field of GBM therapy.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Miller, K.D.; Ostrom, Q.T.; Kruchko, C.; Patil, N.; Tihan, T.; Cioffi, G.; Fuchs, H.E.; Waite, K.A.; Jemal, A.; Siegel, R.L.; et al. Brain and Other Central Nervous System Tumor Statistics, 2021. CA A Cancer J. Clin. 2021, 71, 381–406. [Google Scholar] [CrossRef] [PubMed]
  2. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A Summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  3. Molinaro, A.M.; Taylor, J.W.; Wiencke, J.K.; Wrensch, M.R. Genetic and Molecular Epidemiology of Adult Diffuse Glioma. Nat. Rev. Neurol. 2019, 15, 405–417. [Google Scholar] [CrossRef] [PubMed]
  4. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Tan, A.C.; Ashley, D.M.; López, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of Glioblastoma: State of the Art and Future Directions. CA A Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef]
  6. Delgado-López, P.D.; Corrales-García, E.M. Survival in Glioblastoma: A Review on the Impact of Treatment Modalities. Clin. Transl. Oncol. 2016, 18, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
  7. Wu, W.; Klockow, J.L.; Zhang, M.; Lafortune, F.; Chang, E.; Jin, L.; Wu, Y.; Daldrup-Link, H.E. Glioblastoma Multiforme (GBM): An Overview of Current Therapies and Mechanisms of Resistance. Pharmacol. Res. 2021, 171, 105780. [Google Scholar] [CrossRef] [PubMed]
  8. Noch, E.K.; Ramakrishna, R.; Magge, R. Challenges in the Treatment of Glioblastoma: Multisystem Mechanisms of Therapeutic Resistance. World Neurosurg. 2018, 116, 505–517. [Google Scholar] [CrossRef]
  9. Perrin, S.L.; Samuel, M.S.; Koszyca, B.; Brown, M.P.; Ebert, L.M.; Oksdath, M.; Gomez, G.A. Glioblastoma Heterogeneity and the Tumour Microenvironment: Implications for Preclinical Research and Development of New Treatments. Biochem. Soc. Trans. 2019, 47, 625–638. [Google Scholar] [CrossRef]
  10. Birolini, G.; Valenza, M.; Ottonelli, I.; Passoni, A.; Favagrossa, M.; Duskey, J.T.; Bombaci, M.; Vandelli, M.A.; Colombo, L.; Bagnati, R.; et al. Insights into Kinetics, Release, and Behavioral Effects of Brain-Targeted Hybrid Nanoparticles for Cholesterol Delivery in Huntington’s Disease. J. Control. Release 2021, 330, 587–598. [Google Scholar] [CrossRef]
  11. Pederzoli, F.; Ruozi, B.; Duskey, J.; Hagmeyer, S.; Sauer, A.K.; Grabrucker, S.; Coelho, R.; Oddone, N.; Ottonelli, I.; Daini, E.; et al. Nanomedicine Against Aβ Aggregation by β–Sheet Breaker Peptide Delivery: In Vitro Evidence. Pharmaceutics 2019, 11, 572. [Google Scholar] [CrossRef] [Green Version]
  12. Duskey, J.T.; da Ros, F.; Ottonelli, I.; Zambelli, B.; Vandelli, M.A.; Tosi, G.; Ruozi, B. Enzyme Stability in Nanoparticle Preparations Part 1: Bovine Serum Albumin Improves Enzyme Function. Molecules 2020, 25, 4593. [Google Scholar] [CrossRef]
  13. Duskey, J.T.; Ottonelli, I.; Rinaldi, A.; Parmeggiani, I.; Zambelli, B.; Wang, L.Z.; Prud’homme, R.K.; Vandelli, M.A.; Tosi, G.; Ruozi, B. Tween® Preserves Enzyme Activity and Stability in PLGA Nanoparticles. Nanomaterials 2021, 11, 2946. [Google Scholar] [CrossRef]
  14. Mulvihill, J.J.; Cunnane, E.M.; Ross, A.M.; Duskey, J.T.; Tosi, G.; Grabrucker, A.M. Drug Delivery across the Blood–Brain Barrier: Recent Advances in the Use of Nanocarriers. Nanomedicine 2020, 15, 205–214. [Google Scholar] [CrossRef] [PubMed]
  15. Rigon, L.; Salvalaio, M.; Pederzoli, F.; Legnini, E.; Duskey, J.T.; D’Avanzo, F.; De Filippis, C.; Ruozi, B.; Marin, O.; Vandelli, M.A.; et al. Targeting Brain Disease in MPSII: Preclinical Evaluation of IDS-Loaded PLGA Nanoparticles. Int. J. Mol. Sci. 2019, 20, 2014. [Google Scholar] [CrossRef] [Green Version]
  16. Liu, Z.; Ji, X.; He, D.; Zhang, R.; Liu, Q.; Xin, T. Nanoscale Drug Delivery Systems in Glioblastoma. Nanoscale Res. Lett. 2022, 17, 27. [Google Scholar] [CrossRef]
  17. Aparicio-Blanco, J.; Sanz-Arriazu, L.; Lorenzoni, R.; Blanco-Prieto, M.J. Glioblastoma Chemotherapeutic Agents Used in the Clinical Setting and in Clinical Trials: Nanomedicine Approaches to Improve Their Efficacy. Int. J. Pharm. 2020, 581, 119283. [Google Scholar] [CrossRef]
  18. Shabani, L.; Abbasi, M.; Amini, M.; Amani, A.M.; Vaez, A. The Brilliance of Nanoscience over Cancer Therapy: Novel Promising Nanotechnology-Based Methods for Eradicating Glioblastoma. J. Neurol. Sci. 2022, 440, 120316. [Google Scholar] [CrossRef] [PubMed]
  19. Di Filippo, L.D.; Duarte, J.L.; Luiz, M.T.; de Araújo, J.T.C.; Chorilli, M. Drug Delivery Nanosystems in Glioblastoma Multiforme Treatment: Current State of the Art. Curr. Neuropharmacol. 2021, 19, 787–812. [Google Scholar] [CrossRef]
  20. Amaral, M.; Cruz, N.; Rosa, A.; Nogueira, B.; Costa, D.; Santos, F.; Brazão, M.; Policarpo, P.; Mateus, R.; Kobozev, Y.; et al. An Update of Advanced Nanoplatforms for Glioblastoma Multiforme Management. EXCLI J. 2021, 20, 1544. [Google Scholar] [CrossRef]
  21. Li, J.; Zhao, J.; Tan, T.; Liu, M.; Zeng, Z.; Zeng, Y.; Zhang, L.; Fu, C.; Chen, D.; Xie, T. Nanoparticle Drug Delivery System for Glioma and Its Efficacy Improvement Strategies: A Comprehensive Review. Int. J. Nanomed. 2020, 15, 2563–2582. [Google Scholar] [CrossRef] [Green Version]
  22. Anselmo, A.C.; Mitragotri, S. Nanoparticles in the Clinic. Bioeng. Transl. Med. 2016, 1, 10–29. [Google Scholar] [CrossRef] [PubMed]
  23. Dang, Y.; Guan, J. Nanoparticle-Based Drug Delivery Systems for Cancer Therapy. Smart Mater. Med. 2020, 1, 10–19. [Google Scholar] [CrossRef] [PubMed]
  24. van Tellingen, O.; Yetkin-Arik, B.; de Gooijer, M.C.; Wesseling, P.; Wurdinger, T.; de Vries, H.E. Overcoming the Blood-Brain Tumor Barrier for Effective Glioblastoma Treatment. Drug Resist. Updat. 2015, 19, 1–12. [Google Scholar] [CrossRef] [PubMed]
  25. Dixit, S.; Novak, T.; Miller, K.; Zhu, Y.; Kenney, M.E.; Broome, A.-M. Transferrin Receptor-Targeted Theranostic Gold Nanoparticles for Photosensitizer Delivery in Brain Tumors. Nanoscale 2015, 7, 1782–1790. [Google Scholar] [CrossRef] [PubMed]
  26. Ulbrich, K.; Knobloch, T.; Kreuter, J. Targeting the Insulin Receptor: Nanoparticles for Drug Delivery across the Blood–Brain Barrier (BBB). J. Drug Target. 2011, 19, 125–132. [Google Scholar] [CrossRef]
  27. He, C.; Li, J.; Cai, P.; Ahmed, T.; Henderson, J.T.; Foltz, W.D.; Bendayan, R.; Rauth, A.M.; Wu, X.Y. Two-Step Targeted Hybrid Nanoconstructs Increase Brain Penetration and Efficacy of the Therapeutic Antibody Trastuzumab against Brain Metastasis of HER2-Positive Breast Cancer. Adv. Funct. Mater. 2018, 28, 1705668. [Google Scholar] [CrossRef]
  28. Qiao, R.; Jia, Q.; Hüwel, S.; Xia, R.; Liu, T.; Gao, F.; Galla, H.-J.; Gao, M. Receptor-Mediated Delivery of Magnetic Nanoparticles across the Blood–Brain Barrier. ACS Nano 2012, 6, 3304–3310. [Google Scholar] [CrossRef]
  29. Wei, X.; Zhan, C.; Shen, Q.; Fu, W.; Xie, C.; Gao, J.; Peng, C.; Zheng, P.; Lu, W. A D-Peptide Ligand of Nicotine Acetylcholine Receptors for Brain-Targeted Drug Delivery. Angew. Chem. Int. Ed. 2015, 54, 3023–3027. [Google Scholar] [CrossRef]
  30. Jiang, X.; Xin, H.; Ren, Q.; Gu, J.; Zhu, L.; Du, F.; Feng, C.; Xie, Y.; Sha, X.; 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 2014, 35, 518–529. [Google Scholar] [CrossRef]
  31. Li, J.; Guo, Y.; Kuang, Y.; An, S.; Ma, H.; Jiang, C. Choline Transporter-Targeting and Co-Delivery System for Glioma Therapy. Biomaterials 2013, 34, 9142–9148. [Google Scholar] [CrossRef]
  32. Han, L.; Liu, C.; Qi, H.; Zhou, J.; Wen, J.; Wu, D.; Xu, D.; Qin, M.; Ren, J.; Wang, Q.; et al. Systemic Delivery of Monoclonal Antibodies to the Central Nervous System for Brain Tumor Therapy. Adv. Mater. 2019, 31, 1805697. [Google Scholar] [CrossRef] [PubMed]
  33. Formicola, B.; Dal Magro, R.; Montefusco-Pereira, C.V.; Lehr, C.-M.; Koch, M.; Russo, L.; Grasso, G.; Deriu, M.A.; Danani, A.; Bourdoulous, S.; et al. The Synergistic Effect of Chlorotoxin-MApoE in Boosting Drug-Loaded Liposomes across the BBB. J. Nanobiotechnol. 2019, 17, 115. [Google Scholar] [CrossRef]
  34. Galstyan, A.; Markman, J.L.; Shatalova, E.S.; Chiechi, A.; Korman, A.J.; Patil, R.; Klymyshyn, D.; Tourtellotte, W.G.; Israel, L.L.; Braubach, O.; et al. Blood–Brain Barrier Permeable Nano Immunoconjugates Induce Local Immune Responses for Glioma Therapy. Nat. Commun. 2019, 10, 3850. [Google Scholar] [CrossRef] [Green Version]
  35. Jnaidi, R.; Almeida, A.J.; Gonçalves, L.M. Solid Lipid Nanoparticles and Nanostructured Lipid Carriers as Smart Drug Delivery Systems in the Treatment of Glioblastoma Multiforme. Pharmaceutics 2020, 12, 860. [Google Scholar] [CrossRef]
  36. Wadajkar, A.S.; Dancy, J.G.; Hersh, D.S.; Anastasiadis, P.; Tran, N.L.; Woodworth, G.F.; Winkles, J.A.; Kim, A.J. Tumor-targeted Nanotherapeutics: Overcoming Treatment Barriers for Glioblastoma. WIREs Nanomed. Nanobiotechnol. 2017, 9, e1439. [Google Scholar] [CrossRef]
  37. Luiz, M.T.; Delello Di Filippo, L.; Tofani, L.B.; de Araújo, J.T.C.; Dutra, J.A.P.; Marchetti, J.M.; Chorilli, M. Highlights in Targeted Nanoparticles as a Delivery Strategy for Glioma Treatment. Int. J. Pharm. 2021, 604, 120758. [Google Scholar] [CrossRef]
  38. Raucher, D. Tumor Targeting Peptides: Novel Therapeutic Strategies in Glioblastoma. Curr. Opin. Pharmacol. 2019, 47, 14–19. [Google Scholar] [CrossRef]
  39. Guo, P.; Moses-Gardner, A.; Huang, J.; Smith, E.R.; Moses, M.A. ITGA2 as a Potential Nanotherapeutic Target for Glioblastoma. Sci. Rep. 2019, 9, 6195. [Google Scholar] [CrossRef] [Green Version]
  40. Duskey, J.T.; Rinaldi, A.; Ottonelli, I.; Caraffi, R.; De Benedictis, C.A.; Sauer, A.K.; Tosi, G.; Vandelli, M.A.; Ruozi, B.; Grabrucker, A.M. Glioblastoma Multiforme Selective Nanomedicines for Improved Anti-Cancer Treatments. Pharmaceutics 2022, 14, 1450. [Google Scholar] [CrossRef]
  41. Zhao, Y.-L.; Song, J.-N.; Zhang, M. Role of Caveolin-1 in the Biology of the Blood-Brain Barrier. Rev. Neurosci. 2014, 25, 247–254. [Google Scholar] [CrossRef]
  42. Kang, R.H.; Jang, J.-E.; Huh, E.; Kang, S.J.; Ahn, D.-R.; Kang, J.S.; Sailor, M.J.; Yeo, S.G.; Oh, M.S.; Kim, D.; et al. A Brain Tumor-Homing Tetra-Peptide Delivers a Nano-Therapeutic for More Effective Treatment of a Mouse Model of Glioblastoma. Nanoscale Horiz. 2020, 5, 1213–1225. [Google Scholar] [CrossRef] [PubMed]
  43. Kang, R.H.; Park, J.; Kim, J.; Chowdhury, T.; Oh, J.H.; Kim, J.; Shin, J.; Kim, M.; Park, C.-K.; Lee, S.; et al. A Deep Dive: SIWV Tetra-Peptide Enhancing the Penetration of Nanotherapeutics into the Glioblastoma. ACS Biomater. Sci. Eng. 2022, 8, 4163–4174. [Google Scholar] [CrossRef] [PubMed]
  44. Zhao, R.; Liu, J.; Li, Z.; Zhang, W.; Wang, F.; Zhang, B. Recent Advances in CXCL12/CXCR4 Antagonists and Nano-Based Drug Delivery Systems for Cancer Therapy. Pharmaceutics 2022, 14, 1541. [Google Scholar] [CrossRef] [PubMed]
  45. Hsieh, H.-T.; Huang, H.-C.; Chung, C.-W.; Chiang, C.-C.; Hsia, T.; Wu, H.-F.; Huang, R.-L.; Chiang, C.-S.; Wang, J.; Lu, T.-T.; et al. CXCR4-Targeted Nitric Oxide Nanoparticles Deliver PD-L1 SiRNA for Immunotherapy against Glioblastoma. J. Control. Release 2022, 352, 920–930. [Google Scholar] [CrossRef] [PubMed]
  46. Kuo, Y.-C.; Chang, Y.-H.; Rajesh, R. Targeted Delivery of Etoposide, Carmustine and Doxorubicin to Human Glioblastoma Cells Using Methoxy Poly(Ethylene Glycol)-poly(Ε-caprolactone) Nanoparticles Conjugated with Wheat Germ Agglutinin and Folic Acid. Mater. Sci. Eng. C 2019, 96, 114–128. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, S.; Lu, L.; Zhou, J.; Ran, D.; Wang, S.; Xu, Q.; Xu, W.; Wang, J.; Liu, Y.; Xie, C.; et al. All-Stage Targeted Therapy for Glioblastoma Based on Lipid Membrane Coated Cabazitaxel Nanocrystals. J. Control. Release 2022, 345, 685–695. [Google Scholar] [CrossRef] [PubMed]
  48. Hu, X.-Y.; Gao, L.; Mosel, S.; Ehlers, M.; Zellermann, E.; Jiang, H.; Knauer, S.K.; Wang, L.; Schmuck, C. From Supramolecular Vesicles to Micelles: Controllable Construction of Tumor-Targeting Nanocarriers Based on Host–Guest Interaction between a Pillar[5]Arene-Based Prodrug and a RGD-Sulfonate Guest. Small 2018, 14, 1803952. [Google Scholar] [CrossRef]
  49. Qi, N.; Zhang, S.; Zhou, X.; Duan, W.; Gao, D.; Feng, J.; Li, A. Combined Integrin Avβ3 and Lactoferrin Receptor Targeted Docetaxel Liposomes Enhance the Brain Targeting Effect and Anti-Glioma Effect. J. Nanobiotechnol. 2021, 19, 446. [Google Scholar] [CrossRef] [PubMed]
  50. Ruan, H.; Yao, S.; Wang, S.; Wang, R.; Xie, C.; Guo, H.; Lu, W. Stapled RAP12 Peptide Ligand of LRP1 for Micelles-Based Multifunctional Glioma-Targeted Drug Delivery. J. Chem. Eng. 2021, 403, 126296. [Google Scholar] [CrossRef]
  51. Li, Y.; Pan, Y.; Wang, Y.; Jiang, Z.; Akakuru, O.U.; Li, M.; Zhang, X.; Yuan, B.; Xing, J.; Luo, L.; et al. A D-Peptide Ligand of Neuropeptide Y Receptor Y1 Serves as Nanocarrier Traversing of the Blood Brain Barrier and Targets Glioma. Nano Today 2022, 44, 101465. [Google Scholar] [CrossRef]
  52. Han, M.; Xing, H.; Chen, L.; Cui, M.; Zhang, Y.; Qi, L.; Jin, M.; Yang, Y.; Gao, C.; Gao, Z.; et al. Efficient Antiglioblastoma Therapy in Mice through Doxorubicin-Loaded Nanomicelles Modified Using a Novel Brain-Targeted RVG-15 Peptide. J. Drug Target 2021, 29, 1016–1028. [Google Scholar] [CrossRef]
  53. Xin, X.; Liu, W.; Zhang, Z.-A.; Han, Y.; Qi, L.-L.; Zhang, Y.-Y.; Zhang, X.-T.; Duan, H.-X.; Chen, L.-Q.; Jin, M.-J.; et al. Efficient Anti-Glioma Therapy Through the Brain-Targeted RVG15-Modified Liposomes Loading Paclitaxel-Cholesterol Complex. Int. J. Nanomed. 2021, 16, 5755–5776. [Google Scholar] [CrossRef] [PubMed]
  54. Li, J.; Chai, Z.; Lu, J.; Xie, C.; Ran, D.; Wang, S.; Zhou, J.; Lu, W. αvβ3-Targeted Liposomal Drug Delivery System with Attenuated Immunogenicity Enabled by Linear Pentapeptide for Glioma Therapy. J. Control. Release 2020, 322, 542–554. [Google Scholar] [CrossRef] [PubMed]
  55. Huo, T.; Yang, Y.; Qian, M.; Jiang, H.; Du, Y.; Zhang, X.; Xie, Y.; Huang, R. Versatile Hollow COF Nanospheres via Manipulating Transferrin Corona for Precise Glioma-Targeted Drug Delivery. Biomaterials 2020, 260, 120305. [Google Scholar] [CrossRef] [PubMed]
  56. Shukla, M.K.; Behera, C.; Chakraborty, S.; Sandha, K.K.; Goswami, A.; Gupta, P.N. Tumor Micro-Environment Targeted Collagenase-Modified Albumin Nanoparticles for Improved Drug Delivery. J. Drug Deliv. Sci. Technol. 2022, 71, 103366. [Google Scholar] [CrossRef]
  57. Skóra, B.; Piechowiak, T.; Szychowski, K.A. Epidermal Growth Factor-Labeled Liposomes as a Way to Target the Toxicity of Silver Nanoparticles into EGFR-Overexpressing Cancer Cells in Vitro. Toxicol. Appl. Pharmacol. 2022, 443, 116009. [Google Scholar] [CrossRef] [PubMed]
  58. Feng, J.; Xiang, L.; Fang, C.; Tan, Y.; Li, Y.; Gong, T.; Wu, Q.; Gong, T.; Zhang, Z. Dual-Targeting of Tumor Cells and Tumor-Associated Macrophages by Palmitic Acid Modified Albumin Nanoparticles for Antitumor and Antimetastasis Therapy. ACS Appl. Mater. Interfaces 2022, 14, 14887–14902. [Google Scholar] [CrossRef]
  59. Wang, T.; Zhang, H.; Qiu, W.; Han, Y.; Liu, H.; Li, Z. Biomimetic Nanoparticles Directly Remodel Immunosuppressive Microenvironment for Boosting Glioblastoma Immunotherapy. Bioact. Mater. 2022, 16, 418–432. [Google Scholar] [CrossRef] [PubMed]
  60. Ding, M.; Fan, Y.; Lv, Y.; Liu, J.; Yu, N.; Kong, D.; Sun, H.; Li, J. A Prodrug Hydrogel with Tumor Microenvironment and Near-Infrared Light Dual-Responsive Action for Synergistic Cancer Immunotherapy. Acta Biomater. 2022, 149, 334–346. [Google Scholar] [CrossRef]
  61. Sun, X.; Zhang, J.; Zhao, X.; Yang, C.; Shi, M.; Zhang, B.; Hu, H.; Qiao, M.; Chen, D.; Zhao, X. Binary Regulation of the Tumor Microenvironment by a PH-Responsive Reversible Shielding Nanoplatform for Improved Tumor Chemo-Immunotherapy. Acta Biomater. 2022, 138, 505–517. [Google Scholar] [CrossRef]
  62. Izraely, S.; Witz, I.P. Site-Specific Metastasis: A Cooperation between Cancer Cells and the Metastatic Microenvironment. Int. J. Cancer 2021, 148, 1308–1322. [Google Scholar] [CrossRef] [PubMed]
  63. Oh, H.J.; Kim, J.; Kim, H.; Choi, N.; Chung, S. Microfluidic Reconstitution of Tumor Microenvironment for Nanomedical Applications. Adv. Healthc. Mater. 2021, 10, 2002122. [Google Scholar] [CrossRef]
  64. Wang, W.; Jin, Y.; Liu, X.; Chen, F.; Zheng, X.; Liu, T.; Yang, Y.; Yu, H. Endogenous Stimuli-Activatable Nanomedicine for Immune Theranostics for Cancer. Adv. Funct. Mater. 2021, 31, 2100386. [Google Scholar] [CrossRef]
  65. 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, 7921–7924. [Google Scholar] [CrossRef] [PubMed]
  66. Leroux, J.-C. Editorial: Drug Delivery: Too Much Complexity, Not Enough Reproducibility. Angew. Chem. Int. Ed. 2017, 56, 15170–15171. [Google Scholar] [CrossRef] [Green Version]
  67. Danhier, F. To Exploit the Tumor Microenvironment: Since the EPR Effect Fails in the Clinic, What Is the Future of Nanomedicine? J. Control. Release 2016, 244, 108–121. [Google Scholar] [CrossRef]
  68. Nakamura, Y.; Mochida, A.; Choyke, P.L.; Kobayashi, H. Nanodrug Delivery: Is the Enhanced Permeability and Retention Effect Sufficient for Curing Cancer? Bioconjug. Chem. 2016, 27, 2225–2238. [Google Scholar] [CrossRef] [PubMed]
  69. de Lázaro, I.; Mooney, D.J. A Nanoparticle’s Pathway into Tumours. Nat. Mater. 2020, 19, 486–487. [Google Scholar] [CrossRef]
  70. Golombek, S.K.; May, J.-N.; Theek, B.; Appold, L.; Drude, N.; Kiessling, F.; Lammers, T. Tumor Targeting via EPR: Strategies to Enhance Patient Responses. Adv. Drug Deliv. Rev. 2018, 130, 17–38. [Google Scholar] [CrossRef]
  71. Park, J.; Choi, Y.; Chang, H.; Um, W.; Ryu, J.H.; Kwon, I.C. Alliance with EPR Effect: Combined Strategies to Improve the EPR Effect in the Tumor Microenvironment. Theranostics 2019, 9, 8073–8090. [Google Scholar] [CrossRef]
  72. Kim, J.; Hong, J.; Lee, J.; Fakhraei Lahiji, S.; Kim, Y.-H. Recent Advances in Tumor Microenvironment-Targeted Nanomedicine Delivery Approaches to Overcome Limitations of Immune Checkpoint Blockade-Based Immunotherapy. J. Control. Release 2021, 332, 109–126. [Google Scholar] [CrossRef]
  73. Li, J.; Burgess, D.J. Nanomedicine-Based Drug Delivery towards Tumor Biological and Immunological Microenvironment. Acta Pharm. Sin. B 2020, 10, 2110–2124. [Google Scholar] [CrossRef]
  74. Fu, S.; Xu, X.; Ma, Y.; Zhang, S.; Zhang, S. RGD Peptide-Based Non-Viral Gene Delivery Vectors Targeting Integrin Avβ3 for Cancer Therapy. J. Drug Target 2019, 27, 1–11. [Google Scholar] [CrossRef]
  75. Lv, Y.; Xu, C.; Zhao, X.; Lin, C.; Yang, X.; Xin, X.; Zhang, L.; Qin, C.; Han, X.; Yang, L.; et al. Nanoplatform Assembled from a CD44-Targeted Prodrug and Smart Liposomes for Dual Targeting of Tumor Microenvironment and Cancer Cells. ACS Nano 2018, 12, 1519–1536. [Google Scholar] [CrossRef] [PubMed]
  76. Ryu, J.H.; Shin, M.; Kim, S.A.; Lee, S.; Kim, H.; Koo, H.; Kim, B.-S.; Song, H.K.; Kim, S.H.; Choi, K.; et al. In Vivo Fluorescence Imaging for Cancer Diagnosis Using Receptor-Targeted Epidermal Growth Factor-Based Nanoprobe. Biomaterials 2013, 34, 9149–9159. [Google Scholar] [CrossRef] [PubMed]
  77. Zalba, S.; Contreras, A.M.; Merino, M.; Navarro, I.; de Ilarduya, C.T.; Trocóniz, I.F.; Koning, G.; Garrido, M.J. EGF-Liposomes Promote Efficient EGFR Targeting in Xenograft Colocarcinoma Model. Nanomedicine 2016, 11, 465–477. [Google Scholar] [CrossRef]
  78. Zhang, Y.; Elechalawar, C.K.; Yang, W.; Frickenstein, A.N.; Asfa, S.; Fung, K.-M.; Murphy, B.N.; Dwivedi, S.K.; Rao, G.; Dey, A.; et al. Disabling Partners in Crime: Gold Nanoparticles Disrupt Multicellular Communications within the Tumor Microenvironment to Inhibit Ovarian Tumor Aggressiveness. Mater. Today 2022, 56, 79–95. [Google Scholar] [CrossRef]
  79. Ambasta, R.K.; Sharma, A.; Kumar, P. Nanoparticle Mediated Targeting of VEGFR and Cancer Stem Cells for Cancer Therapy. Vasc. Cell 2011, 3, 26. [Google Scholar] [CrossRef] [Green Version]
  80. Gysler, S.M.; Drapkin, R. Tumor Innervation: Peripheral Nerves Take Control of the Tumor Microenvironment. J. Clin. Investig. 2021, 131, e147276. [Google Scholar] [CrossRef]
  81. Hernandez, S.; Serrano, A.G.; Solis Soto, L.M. The Role of Nerve Fibers in the Tumor Immune Microenvironment of Solid Tumors. Adv. Biol. 2022, 6, 2200046. [Google Scholar] [CrossRef] [PubMed]
  82. Zahalka, A.H.; Arnal-Estapé, A.; Maryanovich, M.; Nakahara, F.; Cruz, C.D.; Finley, L.W.S.; Frenette, P.S. Adrenergic Nerves Activate an Angio-Metabolic Switch in Prostate Cancer. Science 2017, 358, 321–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Shen, J.; Sun, C.; Wang, Z.; Chu, Z.; Liu, C.; Xu, X.; Xia, M.; Zhao, M.; Wang, C. Sequential Receptor–Mediated Mixed-Charge Nanomedicine to Target Pancreatic Cancer, Inducing Immunogenic Cell Death and Reshaping the Tumor Microenvironment. Int. J. Pharm. 2021, 601, 120553. [Google Scholar] [CrossRef] [PubMed]
  84. Zeng, W.; Liu, P.; Pan, W.; Singh, S.R.; Wei, Y. Hypoxia and Hypoxia Inducible Factors in Tumor Metabolism. Cancer Lett. 2015, 356, 263–267. [Google Scholar] [CrossRef]
  85. Rinaldi, A.; Caraffi, R.; Grazioli, M.V.; Oddone, N.; Giardino, L.; Tosi, G.; Vandelli, M.A.; Calzà, L.; Ruozi, B.; Duskey, J.T. Applications of the ROS-Responsive Thioketal Linker for the Production of Smart Nanomedicines. Polymers 2022, 14, 687. [Google Scholar] [CrossRef]
  86. Oddone, N.; Pederzoli, F.; Duskey, J.T.; De Benedictis, C.A.; Grabrucker, A.M.; Forni, F.; Angela Vandelli, M.; Ruozi, B.; Tosi, G. ROS-Responsive “Smart” Polymeric Conjugate: Synthesis, Characterization and Proof-of-Concept Study. Int. J. Pharm. 2019, 570, 118655. [Google Scholar] [CrossRef]
  87. Oddone, N.; Boury, F.; Garcion, E.; Grabrucker, A.M.; Martinez, M.C.; Da Ros, F.; Janaszewska, A.; Forni, F.; Vandelli, M.A.; Tosi, G.; et al. Synthesis, Characterization, and In Vitro Studies of an Reactive Oxygen Species (ROS)-Responsive Methoxy Polyethylene Glycol-Thioketal-Melphalan Prodrug for Glioblastoma Treatment. Front. Pharmacol. 2020, 11, 574. [Google Scholar] [CrossRef]
  88. Wei, X.; Zhao, H.; Huang, G.; Liu, J.; He, W.; Huang, Q. ES-MION-Based Dual-Modality PET/MRI Probes for Acidic Tumor Microenvironment Imaging. ACS Omega 2022, 7, 3442–3451. [Google Scholar] [CrossRef]
  89. Koo, S.; Park, O.K.; Kim, J.; Han, S.I.; Yoo, T.Y.; Lee, N.; Kim, Y.G.; Kim, H.; Lim, C.; Bae, J.-S.; et al. Enhanced Chemodynamic Therapy by Cu–Fe Peroxide Nanoparticles: Tumor Microenvironment-Mediated Synergistic Fenton Reaction. ACS Nano 2022, 16, 2535–2545. [Google Scholar] [CrossRef] [PubMed]
  90. Malhotra, N.; Lee, J.-S.; Liman, R.A.D.; Ruallo, J.M.S.; Villaflores, O.B.; Ger, T.-R.; Hsiao, C.-D. Potential Toxicity of Iron Oxide Magnetic Nanoparticles: A Review. Molecules 2020, 25, 3159. [Google Scholar] [CrossRef] [PubMed]
  91. Feng, Q.; Liu, Y.; Huang, J.; Chen, K.; Huang, J.; Xiao, K. Uptake, Distribution, Clearance, and Toxicity of Iron Oxide Nanoparticles with Different Sizes and Coatings. Sci. Rep. 2018, 8, 2082. [Google Scholar] [CrossRef] [Green Version]
  92. Kalbasi, A.; Ribas, A. Tumour-Intrinsic Resistance to Immune Checkpoint Blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef] [PubMed]
  93. Gao, S.; Yang, X.; Xu, J.; Qiu, N.; Zhai, G. Nanotechnology for Boosting Cancer Immunotherapy and Remodeling Tumor Microenvironment: The Horizons in Cancer Treatment. ACS Nano 2021, 15, 12567–12603. [Google Scholar] [CrossRef] [PubMed]
  94. Ngamcherdtrakul, W.; Bejan, D.S.; Cruz-Muñoz, W.; Reda, M.; Zaidan, H.Y.; Siriwon, N.; Marshall, S.; Wang, R.; Nelson, M.A.; Rehwaldt, J.P.C.; et al. Targeted Nanoparticle for Co-Delivery of HER2 SiRNA and a Taxane to Mirror the Standard Treatment of HER2+ Breast Cancer: Efficacy in Breast Tumor and Brain Metastasis. Small 2022, 18, 2107550. [Google Scholar] [CrossRef]
  95. Walens, A.; DiMarco, A.V.; Lupo, R.; Kroger, B.R.; Damrauer, J.S.; Alvarez, J.V. CCL5 Promotes Breast Cancer Recurrence through Macrophage Recruitment in Residual Tumors. eLife 2019, 8, e43653. [Google Scholar] [CrossRef] [PubMed]
  96. Zhou, X.; Liu, X.; Huang, L. Macrophage-Mediated Tumor Cell Phagocytosis: Opportunity for Nanomedicine Intervention. Adv. Funct. Mater. 2021, 31, 2006220. [Google Scholar] [CrossRef]
  97. Morad, G.; Helmink, B.A.; Sharma, P.; Wargo, J.A. Hallmarks of Response, Resistance, and Toxicity to Immune Checkpoint Blockade. Cell 2021, 184, 5309–5337. [Google Scholar] [CrossRef]
  98. Kubli, S.P.; Berger, T.; Araujo, D.V.; Siu, L.L.; Mak, T.W. Beyond Immune Checkpoint Blockade: Emerging Immunological Strategies. Nat. Rev. Drug Discov. 2021, 20, 899–919. [Google Scholar] [CrossRef] [PubMed]
  99. Alghamri, M.S.; Banerjee, K.; Mujeeb, A.A.; Mauser, A.; Taher, A.; Thalla, R.; McClellan, B.L.; Varela, M.L.; Stamatovic, S.M.; Martinez-Revollar, G.; et al. Systemic Delivery of an Adjuvant CXCR4–CXCL12 Signaling Inhibitor Encapsulated in Synthetic Protein Nanoparticles for Glioma Immunotherapy. ACS Nano 2022, 16, 8729–8750. [Google Scholar] [CrossRef] [PubMed]
  100. Sarvaria, A.; Madrigal, J.A.; Saudemont, A. B Cell Regulation in Cancer and Anti-Tumor Immunity. Cell Mol. Immunol. 2017, 14, 662–674. [Google Scholar] [CrossRef]
  101. Shen, L.; Li, J.; Liu, Q.; Das, M.; Song, W.; Zhang, X.; Tiruthani, K.; Dorosheva, O.; Hu, H.; Lai, S.K.; et al. Nano-Trapping CXCL13 Reduces Regulatory B Cells in Tumor Microenvironment and Inhibits Tumor Growth. J. Control. Release 2022, 343, 303–313. [Google Scholar] [CrossRef] [PubMed]
  102. Mourdikoudis, S.; Pallares, R.M.; Thanh, N.T.K. Characterization Techniques for Nanoparticles: Comparison and Complementarity upon Studying Nanoparticle Properties. Nanoscale 2018, 10, 12871–12934. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Kaliva, M.; Vamvakaki, M. Nanomaterials Characterization. In Polymer Science and Nanotechnology; Elsevier: Amsterdam, The Netherlands, 2020; pp. 401–433. ISBN 978-0-12-816806-6. [Google Scholar]
  104. Modena, M.M.; Rühle, B.; Burg, T.P.; Wuttke, S. Nanoparticle Characterization: What to Measure? Adv. Mater. 2019, 31, 1901556. [Google Scholar] [CrossRef] [PubMed]
  105. Baer, D.R.; Engelhard, M.H.; Johnson, G.E.; Laskin, J.; Lai, J.; Mueller, K.; Munusamy, P.; Thevuthasan, S.; Wang, H.; Washton, N.; et al. Surface Characterization of Nanomaterials and Nanoparticles: Important Needs and Challenging Opportunities. J. Vac. Sci. Technol. 2013, 31, 050820. [Google Scholar] [CrossRef] [Green Version]
  106. Verma, A.; Stellacci, F. Effect of Surface Properties on Nanoparticle–Cell Interactions. Small 2010, 6, 12–21. [Google Scholar] [CrossRef]
  107. Smith, A.M.; Johnston, K.A.; Crawford, S.E.; Marbella, L.E.; Millstone, J.E. Ligand Density Quantification on Colloidal Inorganic Nanoparticles. Analyst 2017, 142, 11–29. [Google Scholar] [CrossRef]
  108. Jayawardena, H.S.N.; Liyanage, S.H.; Rathnayake, K.; Patel, U.; Yan, M. Analytical Methods for Characterization of Nanomaterial Surfaces. Anal. Chem. 2021, 93, 1889–1911. [Google Scholar] [CrossRef]
  109. Kumar, A.; Dixit, C.K. Methods for Characterization of Nanoparticles. In Advances in Nanomedicine for the Delivery of Therapeutic Nucleic Acids; Elsevier: Amsterdam, The Netherlands, 2017; pp. 43–58. ISBN 978-0-08-100557-6. [Google Scholar]
  110. Bhattacharjee, S. DLS and Zeta Potential—What They Are and What They Are Not? J. Control. Release 2016, 235, 337–351. [Google Scholar] [CrossRef]
  111. Silva, A.M.; Martins-Gomes, C.; Coutinho, T.E.; Fangueiro, J.F.; Sanchez-Lopez, E.; Pashirova, T.N.; Andreani, T.; Souto, E.B. Soft Cationic Nanoparticles for Drug Delivery: Production and Cytotoxicity of Solid Lipid Nanoparticles (SLNs). Appl. Sci. 2019, 9, 4438. [Google Scholar] [CrossRef] [Green Version]
  112. Ottonelli, I.; Bighinati, A.; Adani, E.; Loll, F.; Caraffi, R.; Vandelli, M.A.; Boury, F.; Tosi, G.; Duskey, J.T.; Marigo, V.; et al. Optimization of an Injectable Hydrogel Depot System for the Controlled Release of Retinal-Targeted Hybrid Nanoparticles. Pharmaceutics 2023, 15, 25. [Google Scholar] [CrossRef]
  113. Martens, T.F.; Remaut, K.; Deschout, H.; Engbersen, J.F.J.; Hennink, W.E.; van Steenbergen, M.J.; Demeester, J.; De Smedt, S.C.; Braeckmans, K. Coating Nanocarriers with Hyaluronic Acid Facilitates Intravitreal Drug Delivery for Retinal Gene Therapy. J. Control. Release 2015, 202, 83–92. [Google Scholar] [CrossRef] [PubMed]
  114. Mester, L.; Govyadinov, A.A.; Chen, S.; Goikoetxea, M.; Hillenbrand, R. Subsurface Chemical Nanoidentification by Nano-FTIR Spectroscopy. Nat. Commun. 2020, 11, 3359. [Google Scholar] [CrossRef]
  115. Ţucureanu, V.; Matei, A.; Avram, A.M. FTIR Spectroscopy for Carbon Family Study. Crit. Rev. Anal. Chem. 2016, 46, 502–520. [Google Scholar] [CrossRef] [PubMed]
  116. Riaz, T.; Zeeshan, R.; Zarif, F.; Ilyas, K.; Muhammad, N.; Safi, S.Z.; Rahim, A.; Rizvi, S.A.A.; Rehman, I.U. FTIR Analysis of Natural and Synthetic Collagen. Appl. Spectrosc. Rev. 2018, 53, 703–746. [Google Scholar] [CrossRef]
  117. Guerrero-Pérez, M.O.; Patience, G.S. Experimental Methods in Chemical Engineering: Fourier Transform Infrared Spectroscopy—FTIR. Can. J. Chem. Eng. 2020, 98, 25–33. [Google Scholar] [CrossRef]
  118. Liu, Y.; Wang, X.; Li, J.; Tang, J.; Li, B.; Zhang, Y.; Gu, N.; Yang, F. Sphingosine 1-Phosphate Liposomes for Targeted Nitric Oxide Delivery to Mediate Anticancer Effects against Brain Glioma Tumors. Adv. Mater. 2021, 33, 2101701. [Google Scholar] [CrossRef] [PubMed]
  119. Zhang, H.; van Os, W.L.; Tian, X.; Zu, G.; Ribovski, L.; Bron, R.; Bussmann, J.; Kros, A.; Liu, Y.; Zuhorn, I.S. Development of Curcumin-Loaded Zein Nanoparticles for Transport across the Blood–Brain Barrier and Inhibition of Glioblastoma Cell Growth. Biomater. Sci. 2021, 9, 7092–7103. [Google Scholar] [CrossRef] [PubMed]
  120. Luque-Michel, E.; Sebastian, V.; Larrea, A.; Marquina, C.; Blanco-Prieto, M.J. Co-Encapsulation of Superparamagnetic Nanoparticles and Doxorubicin in PLGA Nanocarriers: Development, Characterization and in Vitro Antitumor Efficacy in Glioma Cells. Eur. J. Pharm. Biopharm. 2019, 145, 65–75. [Google Scholar] [CrossRef]
  121. Budama-Kilinc, Y.; Kecel-Gunduz, S.; Cakir-Koc, R.; Aslan, B.; Bicak, B.; Kokcu, Y.; Ozel, A.E.; Akyuz, S. Structural Characterization and Drug Delivery System of Natural Growth-Modulating Peptide Against Glioblastoma Cancer. Int. J. Pept. Res. Ther. 2021, 27, 2015–2028. [Google Scholar] [CrossRef]
  122. Mazarei, M.; Mohammadi Arvejeh, P.; Mozafari, M.R.; Khosravian, P.; Ghasemi, S. Anticancer Potential of Temozolomide-Loaded Eudragit-Chitosan Coated Selenium Nanoparticles: In Vitro Evaluation of Cytotoxicity, Apoptosis and Gene Regulation. Nanomaterials 2021, 11, 1704. [Google Scholar] [CrossRef]
  123. Ortiz-Islas, E.; Sosa-Arróniz, A.; Manríquez-Ramírez, M.E.; Rodríguez-Pérez, C.E.; Tzompantzi, F.; Padilla, J.M. Mesoporous Silica Nanoparticles Functionalized with Folic Acid for Targeted Release Cis-Pt to Glioblastoma Cells. Rev. Adv. Mater. Sci. 2021, 60, 25–37. [Google Scholar] [CrossRef]
  124. Carissimi, G.; Montalbán, M.G.; Víllora, G.; Barth, A. Direct Quantification of Drug Loading Content in Polymeric Nanoparticles by Infrared Spectroscopy. Pharmaceutics 2020, 12, 912. [Google Scholar] [CrossRef] [PubMed]
  125. Paudel, A.; Raijada, D.; Rantanen, J. Raman Spectroscopy in Pharmaceutical Product Design. Adv. Drug Deliv. Rev. 2015, 89, 3–20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Gualerzi, A.; Picciolini, S.; Carlomagno, C.; Terenzi, F.; Ramat, S.; Sorbi, S.; Bedoni, M. Raman Profiling of Circulating Extracellular Vesicles for the Stratification of Parkinson’s Patients. Nanomedicine 2019, 22, 102097. [Google Scholar] [CrossRef] [PubMed]
  127. Huh, Y.S.; Chung, A.J.; Erickson, D. Surface Enhanced Raman Spectroscopy and Its Application to Molecular and Cellular Analysis. Microfluid Nanofluid 2009, 6, 285–297. [Google Scholar] [CrossRef]
  128. Sahli, F.; Courcelle, M.; Palama, T.; Djaker, N.; Savarin, P.; Spadavecchia, J. Temozolomide, Gemcitabine, and Decitabine Hybrid Nanoconjugates: From Design to Proof-of-Concept (PoC) of Synergies toward the Understanding of Drug Impact on Human Glioblastoma Cells. J. Med. Chem. 2020, 63, 7410–7421. [Google Scholar] [CrossRef]
  129. Depciuch, J.; Miszczyk, J.; Maximenko, A.; Zielinski, P.M.; Rawojć, K.; Panek, A.; Olko, P.; Parlinska-Wojtan, M. Gold Nanopeanuts as Prospective Support for Cisplatin in Glioblastoma Nano-Chemo-Radiotherapy. Int. J. Mol. Sci. 2020, 21, 9082. [Google Scholar] [CrossRef]
  130. Marbella, L.E.; Millstone, J.E. NMR Techniques for Noble Metal Nanoparticles. Chem. Mater. 2015, 27, 2721–2739. [Google Scholar] [CrossRef] [Green Version]
  131. Garcia-Fuentes, M.; Torres, D.; Martín-Pastor, M.; Alonso, M.J. Application of NMR Spectroscopy to the Characterization of PEG-Stabilized Lipid Nanoparticles. Langmuir 2004, 20, 8839–8845. [Google Scholar] [CrossRef]
  132. Marchetti, A.; Chen, J.; Pang, Z.; Li, S.; Ling, D.; Deng, F.; Kong, X. Understanding Surface and Interfacial Chemistry in Functional Nanomaterials via Solid-State NMR. Adv. Mater. 2017, 29, 1605895. [Google Scholar] [CrossRef]
  133. Pucci, C.; De Pasquale, D.; Marino, A.; Martinelli, C.; Lauciello, S.; Ciofani, G. Hybrid Magnetic Nanovectors Promote Selective Glioblastoma Cell Death through a Combined Effect of Lysosomal Membrane Permeabilization and Chemotherapy. ACS Appl. Mater. Interfaces 2020, 12, 29037–29055. [Google Scholar] [CrossRef] [PubMed]
  134. Hoyos-Ceballos, G.P.; Ruozi, B.; Ottonelli, I.; Da Ros, F.; Vandelli, M.A.; Forni, F.; Daini, E.; Vilella, A.; Zoli, M.; Tosi, G.; et al. PLGA-PEG-ANG-2 Nanoparticles for Blood-Brain Barrier Crossing: Proof-of-Concept Study. Pharmaceutics 2020, 12, 72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Emamgholizadeh Minaei, S.; Khoei, S.; Khoee, S.; Karimi, M.R. Tri-Block Copolymer Nanoparticles Modified with Folic Acid for Temozolomide Delivery in Glioblastoma. Int. J. Biochem. Cell Biol. 2019, 108, 72–83. [Google Scholar] [CrossRef] [PubMed]
  136. Vilella, A.; Tosi, G.; Grabrucker, A.M.; Ruozi, B.; Belletti, D.; Vandelli, M.A.; Boeckers, T.M.; Forni, F.; Zoli, M. Insight on the Fate of CNS-Targeted Nanoparticles. Part I: Rab5-Dependent Cell-Specific Uptake and Distribution. J. Control. Release 2014, 174, 195–201. [Google Scholar] [CrossRef] [PubMed]
  137. Picciolini, S.; Gualerzi, A.; Carlomagno, C.; Cabinio, M.; Sorrentino, S.; Baglio, F.; Bedoni, M. An SPRi-Based Biosensor Pilot Study: Analysis of Multiple Circulating Extracellular Vesicles and Hippocampal Volume in Alzheimer’s Disease. J. Pharm. Biomed. Anal. 2021, 192, 113649. [Google Scholar] [CrossRef]
  138. Crielaard, B.J.; Yousefi, A.; Schillemans, J.P.; Vermehren, C.; Buyens, K.; Braeckmans, K.; Lammers, T.; Storm, G. An in Vitro Assay Based on Surface Plasmon Resonance to Predict the in Vivo Circulation Kinetics of Liposomes. J. Control. Release 2011, 156, 307–314. [Google Scholar] [CrossRef]
  139. Gregori, M.; Taylor, M.; Salvati, E.; Re, F.; Mancini, S.; Balducci, C.; Forloni, G.; Zambelli, V.; Sesana, S.; Michael, M.; et al. Retro-Inverso Peptide Inhibitor Nanoparticles as Potent Inhibitors of Aggregation of the Alzheimer’s Aβ Peptide. Nanomedicine 2017, 13, 723–732. [Google Scholar] [CrossRef] [Green Version]
  140. Gries, M.; Thomas, N.; Daouk, J.; Rocchi, P.; Choulier, L.; Jubréaux, J.; Pierson, J.; Reinhard, A.; Jouan-Hureaux, V.; Chateau, A.; et al. Multiscale Selectivity and in Vivo Biodistribution of NRP-1-Targeted Theranostic AGuIX Nanoparticles for PDT of Glioblastoma. Int. J. Nanomed. 2020, 15, 8739–8758. [Google Scholar] [CrossRef]
  141. Gabold, B.; Adams, F.; Brameyer, S.; Jung, K.; Ried, C.L.; Merdan, T.; Merkel, O.M. Transferrin-Modified Chitosan Nanoparticles for Targeted Nose-to-Brain Delivery of Proteins. Drug Deliv. Transl. Res. 2022. [Google Scholar] [CrossRef]
  142. Krishna, D.N.G.; Philip, J. Review on Surface-Characterization Applications of X-Ray Photoelectron Spectroscopy (XPS): Recent Developments and Challenges. Appl. Surf. Sci. Adv. 2022, 12, 100332. [Google Scholar] [CrossRef]
  143. Pulvirenti, L.; Monforte, F.; Lo Presti, F.; Li Volti, G.; Carota, G.; Sinatra, F.; Bongiorno, C.; Mannino, G.; Cambria, M.T.; Condorelli, G.G. Synthesis of MIL-Modified Fe3O4 Magnetic Nanoparticles for Enhancing Uptake and Efficiency of Temozolomide in Glioblastoma Treatment. Int. J. Mol. Sci. 2022, 23, 2874. [Google Scholar] [CrossRef]
  144. Venditti, I.; Cartoni, A.; Cerra, S.; Fioravanti, R.; Salamone, T.A.; Sciubba, F.; Tabocchini, M.A.; Dini, V.; Battocchio, C.; Iucci, G.; et al. Hydrophilic Gold Nanoparticles as Anti-PD-L1 Antibody Carriers: Synthesis and Interface Properties. Part. Part. Syst. Charact. 2022, 39, 2100282. [Google Scholar] [CrossRef]
  145. Saadatkhah, N.; Carillo Garcia, A.; Ackermann, S.; Leclerc, P.; Latifi, M.; Samih, S.; Patience, G.S.; Chaouki, J. Experimental Methods in Chemical Engineering: Thermogravimetric Analysis—TGA. Can. J. Chem. Eng. 2020, 98, 34–43. [Google Scholar] [CrossRef]
  146. Mansfield, E.; Tyner, K.M.; Poling, C.M.; Blacklock, J.L. Determination of Nanoparticle Surface Coatings and Nanoparticle Purity Using Microscale Thermogravimetric Analysis. Anal. Chem. 2014, 86, 1478–1484. [Google Scholar] [CrossRef]
  147. Bannov, A.G.; Popov, M.V.; Kurmashov, P.B. Thermal Analysis of Carbon Nanomaterials: Advantages and Problems of Interpretation. J. Therm. Anal. Calorim. 2020, 142, 349–370. [Google Scholar] [CrossRef]
  148. Świętek, M.; Ma, Y.-H.; Wu, N.-P.; Paruzel, A.; Tokarz, W.; Horák, D. Tannic Acid Coating Augments Glioblastoma Cellular Uptake of Magnetic Nanoparticles with Antioxidant Effects. Nanomaterials 2022, 12, 1310. [Google Scholar] [CrossRef] [PubMed]
  149. Shahein, S.A.; Aboul-Enein, A.M.; Higazy, I.M.; Abou-Elella, F.; Lojkowski, W.; Ahmed, E.R.; Mousa, S.A.; AbouAitah, K. Targeted Anticancer Potential against Glioma Cells of Thymoquinone Delivered by Mesoporous Silica Core-Shell Nanoformulations with PH-Dependent Release. Int. J. Nanomed. 2019, 14, 5503–5526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Smith, D.J. Chapter 1. Characterization of Nanomaterials Using Transmission Electron Microscopy. In Nanoscience & Nanotechnology Series; Kirkland, A.I., Haigh, S.J., Eds.; Royal Society of Chemistry: Cambridge, UK, 2015; pp. 1–29. ISBN 978-1-84973-805-7. [Google Scholar]
  151. Michen, B.; Geers, C.; Vanhecke, D.; Endes, C.; Rothen-Rutishauser, B.; Balog, S.; Petri-Fink, A. Avoiding Drying-Artifacts in Transmission Electron Microscopy: Characterizing the Size and Colloidal State of Nanoparticles. Sci. Rep. 2015, 5, 9793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Eaton, P.; Quaresma, P.; Soares, C.; Neves, C.; de Almeida, M.P.; Pereira, E.; West, P. A Direct Comparison of Experimental Methods to Measure Dimensions of Synthetic Nanoparticles. Ultramicroscopy 2017, 182, 179–190. [Google Scholar] [CrossRef] [PubMed]
  153. Ghaferi, M.; Raza, A.; Koohi, M.; Zahra, W.; Akbarzadeh, A.; Ebrahimi Shahmabadi, H.; Alavi, S.E. Impact of PEGylated Liposomal Doxorubicin and Carboplatin Combination on Glioblastoma. Pharmaceutics 2022, 14, 2183. [Google Scholar] [CrossRef]
  154. Zhang, D.; Sun, Y.; Wang, S.; Zou, Y.; Zheng, M.; Shi, B. Brain-Targeting Metastatic Tumor Cell Membrane Cloaked Biomimetic Nanomedicines Mediate Potent Chemodynamic and RNAi Combinational Therapy of Glioblastoma. Adv. Funct. Mater. 2022, 32, 2209239. [Google Scholar] [CrossRef]
  155. Costagliola di Polidoro, A.; Zambito, G.; Haeck, J.; Mezzanotte, L.; Lamfers, M.; Netti, P.A.; Torino, E. Theranostic Design of Angiopep-2 Conjugated Hyaluronic Acid Nanoparticles (Thera-ANG-CHANPs) for Dual Targeting and Boosted Imaging of Glioma Cells. Cancers 2021, 13, 503. [Google Scholar] [CrossRef] [PubMed]
  156. Wang, R.; He, Z.; Cai, P.; Zhao, Y.; Gao, L.; Yang, W.; Zhao, Y.; Gao, X.; Gao, F. Surface-Functionalized Modified Copper Sulfide Nanoparticles Enhance Checkpoint Blockade Tumor Immunotherapy by Photothermal Therapy and Antigen Capturing. ACS Appl. Mater. Interfaces 2019, 11, 13964–13972. [Google Scholar] [CrossRef] [PubMed]
  157. Lesiak, B.; Rangam, N.; Jiricek, P.; Gordeev, I.; Tóth, J.; Kövér, L.; Mohai, M.; Borowicz, P. Surface Study of Fe3O4 Nanoparticles Functionalized With Biocompatible Adsorbed Molecules. Front. Chem. 2019, 7, 642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Borowik, A.; Butowska, K.; Konkel, K.; Banasiuk, R.; Derewonko, N.; Wyrzykowski, D.; Davydenko, M.; Cherepanov, V.; Styopkin, V.; Prylutskyy, Y.; et al. The Impact of Surface Functionalization on the Biophysical Properties of Silver Nanoparticles. Nanomaterials 2019, 9, 973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. León Félix, L.; Sanz, B.; Sebastián, V.; Torres, T.E.; Sousa, M.H.; Coaquira, J.a.H.; Ibarra, M.R.; Goya, G.F. Gold-Decorated Magnetic Nanoparticles Design for Hyperthermia Applications and as a Potential Platform for Their Surface-Functionalization. Sci. Rep. 2019, 9, 4185. [Google Scholar] [CrossRef] [Green Version]
  160. Wang, G.; Yan, C.; Gao, S.; Liu, Y. Surface Chemistry of Gold Nanoparticles Determines Interactions with Bovine Serum Albumin. Mater. Sci. Eng. C 2019, 103, 109856. [Google Scholar] [CrossRef]
  161. Ramalho, M.J.; Bravo, M.; Loureiro, J.A.; Lima, J.; Pereira, M.C. Transferrin-Modified Nanoparticles for Targeted Delivery of Asiatic Acid to Glioblastoma Cells. Life Sci. 2022, 296, 120435. [Google Scholar] [CrossRef]
  162. Su, X.; Zhang, D.; Zhang, H.; Zhao, K.; Hou, W. Preparation and Characterization of Angiopep-2 Functionalized Ginsenoside-Rg3 Loaded Nanoparticles and the Effect on C6 Glioma Cells. Pharm. Dev. Technol. 2020, 25, 385–395. [Google Scholar] [CrossRef]
  163. He, W.; Li, X.; Morsch, M.; Ismail, M.; Liu, Y.; Rehman, F.U.; Zhang, D.; Wang, Y.; Zheng, M.; Chung, R.; et al. Brain-Targeted Codelivery of Bcl-2/Bcl-Xl and Mcl-1 Inhibitors by Biomimetic Nanoparticles for Orthotopic Glioblastoma Therapy. ACS Nano 2022, 16, 6293–6308. [Google Scholar] [CrossRef]
  164. Gazaille, C.; Sicot, M.; Akiki, M.; Lautram, N.; Dupont, A.; Saulnier, P.; Eyer, J.; Bastiat, G. Characterization of Biological Material Adsorption to the Surface of Nanoparticles without a Prior Separation Step: A Case Study of Glioblastoma-Targeting Peptide and Lipid Nanocapsules. Pharm. Res. 2021, 38, 681–691. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of novel molecules investigated as targeting moieties for the development of targeted NPs, as well as those targeting the TME for GBM therapy. Abbreviations: BBB—blood-brain barrier; EGF—epidermal growth factor; FA—folic acid; GBM—glioblastoma multiforme; Lf—lactoferrin; MPC—2-methacryloyloxyethyl phosphorylcholine; NPs—nanoparticles; p-HA—p-hydroxybenzoic acid; Tf—transferrin; TME—tumor microenvironment; VEGF—vascular endothelial growth factor.
Figure 1. Schematic representation of novel molecules investigated as targeting moieties for the development of targeted NPs, as well as those targeting the TME for GBM therapy. Abbreviations: BBB—blood-brain barrier; EGF—epidermal growth factor; FA—folic acid; GBM—glioblastoma multiforme; Lf—lactoferrin; MPC—2-methacryloyloxyethyl phosphorylcholine; NPs—nanoparticles; p-HA—p-hydroxybenzoic acid; Tf—transferrin; TME—tumor microenvironment; VEGF—vascular endothelial growth factor.
Ijms 24 02496 g001
Figure 2. Techniques used for surface characterization of NPs for GBM therapy. Abbreviations: GBM—glioblastoma multiforme; NPs—nanoparticles; UV-Vis—ultraviolet-visible.
Figure 2. Techniques used for surface characterization of NPs for GBM therapy. Abbreviations: GBM—glioblastoma multiforme; NPs—nanoparticles; UV-Vis—ultraviolet-visible.
Ijms 24 02496 g002
Figure 3. Morphological characterization of surface-modified NPs. (a) Fe3O4 magnetic NPs covered with a metal–organic framework. SEM images of bare (left) and decorated (right) NPs. Adapted with permission from Pulvirenti et al. [143] (IJMS, MDPI, 2022). (b) TEM analysis of biomimetic NPs made up of siRNA complexed polyethyleneimine xanthate before (left) and after the cell membrane coating (right). Adapted with permission from Zhang et al. [154] (Adv. Funct. Mater., Wiley, 2022). (c) TEM images of hyaluronic acid NPs pre (left) and post (right) modification with Angiopep-2. Adapted with permission from Costagliola di Polidoro et al. [155] (Cancers, MDPI, 2021). Abbreviations: NPs—nanoparticles; SEM—scanning electron microscopy; TEM—transmission electron microscopy.
Figure 3. Morphological characterization of surface-modified NPs. (a) Fe3O4 magnetic NPs covered with a metal–organic framework. SEM images of bare (left) and decorated (right) NPs. Adapted with permission from Pulvirenti et al. [143] (IJMS, MDPI, 2022). (b) TEM analysis of biomimetic NPs made up of siRNA complexed polyethyleneimine xanthate before (left) and after the cell membrane coating (right). Adapted with permission from Zhang et al. [154] (Adv. Funct. Mater., Wiley, 2022). (c) TEM images of hyaluronic acid NPs pre (left) and post (right) modification with Angiopep-2. Adapted with permission from Costagliola di Polidoro et al. [155] (Cancers, MDPI, 2021). Abbreviations: NPs—nanoparticles; SEM—scanning electron microscopy; TEM—transmission electron microscopy.
Ijms 24 02496 g003
Table 1. Summary of the presented nanosystems for GBM targeting.
Table 1. Summary of the presented nanosystems for GBM targeting.
NP TypeTargeting MoietyTargetKey ResultsRef.
Crosslinked
peptide
MPCAcetylcholine
transporter
Successful delivery of nimotuzumab in orthotopic glioma xenograft mice[32]
LiposomesApoE derived
peptide + chlorotoxin
Lipid transportDoxorubicin loaded into the liposomes produced reduced viability of GBM U87 cells and did not affect endothelial cells in vitro[33]
Nano-immune conjugateTf Receptor antibodyTf receptorPMLA backbone conjugated to Tf
Receptor antibody for targeting and CTLA-4 and PD-1 antibodies. The system was able to induce antitumor immune
response in GBM mice
[34]
PEGylated
liposomes
ITGA2 antibodyITGA2Doxorubicin loaded liposomes were able to cross the BBTB but not the healthy BBB. ITGA2 blocked GBM cell migration[39]
PLGA NPsM08 antibodyCell surface
vimentin
NPs loaded with paclitaxel showed increased apoptosis in GBM cells compared to healthy astrocytes[40]
Porous silica NPsSIWV peptideCaveolin-mediated transportAccumulation of NPs in the brain of mice with GBM xenografts, resulting in prolonged survival, with higher GBM selectivity in vitro than in vivo[42,43]
Lipid-CaP NPsCTCE9908 peptideCXCR4Efficient delivery of siRNA in GBM cultures and GBM mice, resulting in silencing of the PD-1 gene ligand[45]
PEG–PCL NPsWGA + FASialic acid + FA receptorNPs were loaded with different anticancer drugs, and the double-ligand strategy showed improved targeting efficacy compared to the single moieties in vitro[46]
PEGylated
liposomes
VAP + p-HAGRP78 protein + dopamine receptorsEnhanced BBB crossing and GBM accumulation in spheroids
Apoptotic and antiangiogenic effect in orthotopic GBM mice
[47]
PEGylated
liposomes
RGD + LfIntegrin αvβ3 + Lf
receptor
Improved BBB crossing and GBM accumulation in spheroids
Improved efficacy of docetaxel in vivo compared to nontargeted liposomes
[49]
Polymeric
micelles
ST-RAP12 peptideLRP1 receptorThe peptide improved GBM specificity of paclitaxel-loaded micelles, with increased survival rate and inhibited angiogenesis in vivo[50]
DSPE–PEG
micelles
DATPNeuropeptide Y
receptor Y1
Increased BBB crossing in vitro compared to other known ligands
The ATP peptide improved photothermal therapy in vivo
[51]
PEGylated
liposomes
RVG15 peptideNicotinic
acetylcholine
receptor
Improved delivery of paclitaxel across the BBB and accumulation in GBM cells in vivo
Inhibition of tumor growth and metastases formation
[53]
LiposomesmnRwr peptideIntegrin αvβ3Increased penetration in tumor spheroids compared to RGD peptide, and increased accumulation in GBM mice[54]
PEI-coated
silica NPs
T10 peptideTf receptorInduced formation of a Tf corona on the surface of NPs to target the Tf Receptor
Efficient BBB crossing and GBM targeting in vivo with prolonged release of doxorubicin
[55]
Albumin NPsCollagenaseExtracellular matrixEfficient delivery of gemcitabine in tumor spheroids[56]
LiposomesEGFEGF receptorIncreased delivery of silver NPs loaded into liposomes, specifically to GBM cells in vitro[57]
Albumin NPsScavenger receptor A + SPARC proteinTAMs in TMEImproved ICB therapy with elimination of TAMs from the TME[58]
Copper–selenium NPsBiomimetic cell membraneTMEShift of TAMs to an M1 phenotype, decreased expression of the PD-1 ligand, and increase in memory T cells[59]
Albumin NPsROS-sensitive linker + PD-1 ligand
antibody
ROS in TMESystem enclosed in a hydrogel together with iron oxide NPs for combined photodynamic therapy and immunomodulation[60]
Platinum NPs + dextran NPsLinkage via pH
-sensitive borate ester
Acidic pH in TMEDisassembly of the two NPs improved penetration into GBM and release of loaded sotuletinib to eliminate TAMs[61]
Abbreviations: BBB—blood-brain barrier; BBTB—blood-brain tumor barrier; CaP—calcium phosphate; EGF—epidermal growth factor; FA—folic acid; GBM—glioblastoma multiforme; ICB—immune checkpoint blockade; Lf—lactoferrin; MPC—2-methacryloyloxyethyl phosphorylcholine; NPs—nanoparticles; PD-1—programmed cell death 1- (ligand, antibody, etc); PEG-PCL—poly(ethylene glycol)–poly(ε-caprolactone); PEI—polyethyleneimine; p-HA—p-hydroxybenzoic acid; PMLA—poly(β-L-malic acid); ROS—reactive oxygen species; TAMs—tumor-associated macrophages; Tf—transferrin; TME—tumor microenvironment; VEGF—vascular endothelial growth factor; WGA—wheat germ agglutinin.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rodà, F.; Caraffi, R.; Picciolini, S.; Tosi, G.; Vandelli, M.A.; Ruozi, B.; Bedoni, M.; Ottonelli, I.; Duskey, J.T. Recent Advances on Surface-Modified GBM Targeted Nanoparticles: Targeting Strategies and Surface Characterization. Int. J. Mol. Sci. 2023, 24, 2496. https://doi.org/10.3390/ijms24032496

AMA Style

Rodà F, Caraffi R, Picciolini S, Tosi G, Vandelli MA, Ruozi B, Bedoni M, Ottonelli I, Duskey JT. Recent Advances on Surface-Modified GBM Targeted Nanoparticles: Targeting Strategies and Surface Characterization. International Journal of Molecular Sciences. 2023; 24(3):2496. https://doi.org/10.3390/ijms24032496

Chicago/Turabian Style

Rodà, Francesca, Riccardo Caraffi, Silvia Picciolini, Giovanni Tosi, Maria Angela Vandelli, Barbara Ruozi, Marzia Bedoni, Ilaria Ottonelli, and Jason Thomas Duskey. 2023. "Recent Advances on Surface-Modified GBM Targeted Nanoparticles: Targeting Strategies and Surface Characterization" International Journal of Molecular Sciences 24, no. 3: 2496. https://doi.org/10.3390/ijms24032496

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

Article Metrics

Back to TopTop