Next Article in Journal
Revealing the Complexity of Fatigue: A Review of the Persistent Challenges and Promises of Artificial Intelligence
Next Article in Special Issue
Integrative Magnetic Resonance Imaging and Metabolomic Characterization of a Glioblastoma Rat Model
Previous Article in Journal
Vertical Mental Timeline Is Not Influenced by VisuoSpatial Processing
Previous Article in Special Issue
Flavonoid Rutin Presented Anti-Glioblastoma Activity Related to the Modulation of Onco miRNA-125b Expression and STAT3 Signaling and Impact on Microglia Inflammatory Profile
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Update on Emergent Nano-Therapeutic Strategies against Pediatric Brain Tumors

by
Ammu V. V. V. Ravi Kiran
1,
G. Kusuma Kumari
1,
Praveen T. Krishnamurthy
1,
Asha P. Johnson
2,
Madhuchandra Kenchegowda
2,
Riyaz Ali M. Osmani
2,
Amr Selim Abu Lila
3,
Afrasim Moin
3,
H. V. Gangadharappa
2,* and
Syed Mohd Danish Rizvi
3,*
1
Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Rocklands, Ooty 643001, The Nilgiris, Tamil Nadu, India
2
Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, Karnataka, India
3
Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il 81442, Saudi Arabia
*
Authors to whom correspondence should be addressed.
Submission received: 16 December 2023 / Revised: 7 February 2024 / Accepted: 13 February 2024 / Published: 18 February 2024

Abstract

:
Pediatric brain tumors are the major cause of pediatric cancer mortality. They comprise a diverse group of tumors with different developmental origins, genetic profiles, therapeutic options, and outcomes. Despite many technological advancements, the treatment of pediatric brain cancers has remained a challenge. Treatment options for pediatric brain cancers have been ineffective due to non-specificity, inability to cross the blood–brain barrier, and causing off-target side effects. In recent years, nanotechnological advancements in the medical field have proven to be effective in curing challenging cancers like brain tumors. Moreover, nanoparticles have emerged successfully, particularly in carrying larger payloads, as well as their stability, safety, and efficacy monitoring. In the present review, we will emphasize pediatric brain cancers, barriers to treating these cancers, and novel treatment options.

1. Introduction

Childhood/pediatric brain cancers are the second most common pediatric cancers, accounting for about one-fourth of all pediatric cancer cases [1]. The Industrial revolution and advancements in genetic screening and sequencing together ushered in new perspectives (both at the molecular and genetic levels) on these pediatric brain cancers. Mounting studies suggest that mutational burden is much lower in childhood brain cancers compared to adult brain cancers [1,2,3]. Furthermore, crucial targets in adult brain cancers cannot necessarily be exploited in childhood brain cancers due to their unique biology, which differs from adult cancers [1,4,5,6]. Over the past few decades, remarkable progress has been made in the treatment of childhood brain cancers, improving the patient survival rate by at least 5 years. Despite these improvements, many pediatric brain tumors are still incurable with high morbidity rates. Additionally, with the intensification of the therapy, the adverse effects of the chemo- and radiotherapies have become gradually apparent; for example, anthracyclines such as doxorubicin could cause cardiomyopathic problems [1,4].
In recent years, cancer nanomedicine has emerged as an important advancement in improving the therapeutic benefit [7,8]. Different nanoparticles, including organic, inorganic, or lipid-based nanoparticles, have been widely tested in delivering cancer theranostics. Further, these nanoparticles proved to be more advantageous than conventional methods due to their higher payload capacity, stability, and prolonged circulation time, thereby improving safety and efficacy [7,8]. In the present review, we will discuss the various pediatric brain cancers, barriers to treating these cancers, and novel treatment options. For selecting the recent relevant and informative research articles, a focused search using the keywords ‘brain tumors’, ‘childhood cancers’, ‘pediatrics’, ‘nanoparticles’, and ‘nanotheranostics’ was run using the databases such as Scopus, Web of Science, PubMed, ScienceDirect, Directory of Open Access Journals (DOAJ), etc. Articles showing more than 95% content and keyword match were included, and the rest were excluded.

2. Pediatric Brain Cancers: Targets and Mechanisms

Over the past few decades, cancer has been a long-lasting disease due to its heterogeneity [9,10]. In order to develop novel therapeutics, a thorough understanding of the underlying pathophysiological and molecular pathways is essential, especially in distinguishing childhood and adult cancers [11,12]. Unlike adult cancers, pediatric cancers are not triggered by lifestyle changes and are less inherited. Though the chance of risk development increases gradually with age, there are still a few exceptions. For instance, the occurrence of bone, brain, and blood cancers is greater in children than in adults. Further, the kinetic profile and therapeutic outcome differ in children than in adults (e.g., genitourinary pH, intestinal mobility, etc.) [11,13]. Additionally, pediatric tissues are immature and in the continuous growing phase, possessing greater metabolic rates and toxicity issues. Lastly, genetic variations in pediatric cancers, such as acute lymphocytic leukemia, Ewing sarcoma, etc., are greatly driven by fusion oncogenes due to chromosomal translocations. Unlike adult cancers, pediatric cancers possess a lower mutational rate, making their therapeutic targeting more challenging [13,14].
Considering the above-mentioned reasons, there is an imminent need to understand novel therapeutic targets and develop new therapeutic options. In this section, we will be discussing the various pediatric brain tumors and treatable options.

2.1. Medulloblastoma

Medulloblastoma (MB) is highly malignant and is the most common childhood brain cancer formed in the cerebellum [15]. MB is categorized into four molecular subgroups: Sonic hedgehog (SHH), WNT, group 3, and group 4 [15,16]. These subgroups are identified as powerful predictors of therapy outcomes. For instance, patients with WNT tumors have greater survival after therapy than group 3 tumors.

2.1.1. WNT Subgroup

WNT-associated MB is most common in children over the age of three, with a 5-year survival rate, and is seldom metastatic. WNT-MB has no focal somatic copy number aberrations (SCNAs) and typically has chromosome 6 monosomy. Initially, it was recognized that people with Turcot syndrome, a genetic disorder caused by mutations in the adenomatous polyposis coli (APC) gene, a repressor of WNT signaling, had a higher incidence of MB [16]. Later, it was discovered that a subgroup of sporadic MBs had WNT pathway mutations, notably in CTNNB1 (encoding β-catenin). β-catenin enhances WNT target gene transcription by interacting with a number of chromatin modifiers such as histone acetyltransferases, SMARCA4, and CREBBP. Furthermore, whole-genome sequencing (WGS) has revealed that CTNNB1 mutations commonly coincide with missense variations in the DEAD-box RNA helicase DDX3X [17,18]. WNT-MB tumors are thought to form in the dorsal brainstem from progenitor cells in the lower rhombic lip.
Despite WNT-MB’s favorable prognosis, recent clinical trials have focused on lowering chemotherapy or radiation doses in the hopes of reducing off-target implications [16,17]. It is worth noting that WNT signaling has been proposed to play a role in WNT-MB’s exceptional response to standard therapy. Further studies revealed that these tumors release soluble WNT antagonists, which may disrupt the blood–brain barrier and sensitize tumors to chemotherapy.

2.1.2. SHH Subgroup

Patients with SHH-activated MB were considered to have high-risk disease (survival rate of 50–75%), which is lower than WNT patients but higher than patients in group 3. SHH-MB, like WNT-MB, has a relatively even gender distribution. Unlike WNT-MB, most SHH-MB patients are newborns or adults; just a few youngsters have this tumor subtype. SHH-MB’s genome contains substantially more SCNAs than WNT-MB’s genome [13,17,18]. Initially, SHH signaling in MB was discovered in the context of Gorlin syndrome, a hereditary disorder embodied by basal cell carcinomas of the skin, craniofacial abnormalities, and an elevated prevalence of MB. Gorlin patients have germline mutations in PTCH1, a repressor of the SHH pathway. Germline mutations in the gene encoding Suppressor of Fused (SUFU) also predispose to SHH-MB [13,17]. Furthermore, spontaneous MBs in the SHH subgroup exhibit PTCH1 and SUFU loss-of-function mutations, Smoothened (SMO) and SHH activation mutations, GLI2, and MYCN amplifications. SHH pathway gene mutations are discovered in an age-dependent manner: All age groups have PTCH1 mutations; however, infants and adults are more likely to have SUFU mutations, infants are more likely to have SMO mutations, and children under the age of three are more likely to have MYCN and GLI2 amplifications. Mice with Gli2, Smo, Ptch1, and Sufu mutations are also susceptible to MB, indicating the function of these genes as tumorigenesis elicitors. A subset of SHH-MB patients, notably older adolescents and teenagers, have significant “chromothripsis” (chromosome shattering) [17]. Using WGS, Rausch et al. [19] discovered that these individuals typically had germline or somatic TP53 mutations, the former of which is related to Li-Fraumeni syndrome (LFS). Chromothripsis can cause SHH pathway gene amplification, such as GLI2 and MYCN, which increases SHH target gene expression and drives tumor development. For years, scientists have researched the genesis of SHH-MB tumors; however, most recent research indicates that these tumors are caused by granule neuron progenitors (GNPs). Small-molecule SHH pathway antagonists have made it possible to treat this subset of tumors in innovative ways. SMO inhibitors (SMOis), in particular GDC-0449 (vismodegib) and NVP-LDE225 (erismodegib), have been linked in clinical studies to strong (albeit frequently transitory) responses in MB patients [17]. SMO mutations may sometimes prevent long-term therapeutic benefits from occurring; in other circumstances, mutations in downstream components of the SHH pathway (for instance, MYCN or GLI2 amplifications) or in other pathways might render tumor cells resistant to these medications. The antifungal drug itraconazole and the cyclopamine derivative IPI-926 (saridegib) are two examples of second-site SMOis that have shown potential in preclinical research [20,21]. Arsenic trioxide, an inhibitor of downstream components of the SHH pathway, can accelerate GLI2 degradation [13].

2.1.3. Group 3 Subgroup

Group 3-MBs account for about ~20–25% with very little prognosis and occur more in pediatrics than adults. At the time of diagnosis, most group 3-MB patients have the highest metastasis. Most group 3-MB originate in the midline in the proximity of the fourth ventricle of the brainstem [13,17]. The possibility of higher metastasis is due to this location, which facilitates access to the cerebrospinal fluid. Contrastingly, no germline mutations are known for the formation of group 3-MB [18]. The prominence of these tumors is majorly due to the amplification of the MYC oncogene, which fuses with the plasmacytoma variant translocation 1 (PVT1), which stabilizes the MYC proteins. Group 3-MB exhibits orthodenticle homeobox 2 (OTX2) amplification, which upregulates MYC expression, thereby promoting tumor formation. Further, genomic instability in group 3-MB is associated with the loss or gain of chromosomes. One of the key events in group 3-MB that happens in chromosome 17 is the simultaneous loss of 17p and gain of 17q chromosomes [18].
In order to identify novel therapeutics for group 3-MB, a high-throughput screening has been generated. The same study has revealed that gemcitabine and pemetrexed suppressed the group 3-MB in both mouse and human models. Despite improvement in survival having been observed in in vivo models, the involvement of tumor microenvironment has enhanced drug resistance in tumors. Another potential undruggable target in group 3-MBs is the overexpressed MYC gene. Treatment options for MYC can be achieved by bromodomain protein inhibitors such as JQ1, which arrest the G1 phase of the cell cycle, causing apoptosis [18].

2.1.4. Group 4 Subgroup

Group 4-MBs account for more than one-third of all cases, with metastatic hallmark being the most common [13,18]. Unlike adult patients, infants and young adults with group 4-MBs have an intermediate survival rate. Similar to group 3-MBs, group 4-MBs originate adjacent to the fourth ventricle. Most of group-4 MBs possess chromosomal instability (esp. chromosome 17), causing the prevalence of SCNAs resulting from tetraploidization. Further, group 4-MBs possess < 10% mutations in KDM6A, ZMYM3, CTDNEP1, etc. An increase in SCNAs is reported to affect the NF-κB signaling, implying the potential therapeutic target [13]. Although the origin of group 4-MBs is unknown, gene signature has led to the glutamate-secreting neurons, suggesting from glutamatergic progenitors. Further studies revealed that the nuclear transitory zone (NTZ) is the main origin of group 4-MBs [22]. Due to the absence of a suitable animal model that mimics the group 4-MBs, proper therapeutic strategies against these tumors are limited. Few therapeutic options exist for treating group 4-MBs; for tumors that express MYCN and CDK6 amplification, bromodomain inhibitors and cyclin-dependent kinase (CDK) inhibitors along with MYC-destabilizing Aurora kinase A inhibitors may be preferred [23,24].

2.2. Gliomas/High-Grade Gliomas (HGGs)

Pediatric high-grade gliomas (HGGs) are the most common malignant brain tumors, majorly consisting of glioblastoma, astrocytoma, etc. [25]. World Health organization (WHO) has classified glioblastomas as grade IV due to their high proliferation, neovascularization, and necrosis [26,27]. Diffuse intrinsic pontine gliomas (DIPGs) are tumors that resemble gliomas histologically but possess diffusely metastatic growth inside the brainstem. Glioblastomas exhibit higher methylation of O6-methylguanine-DNA methyltransferase (MGMT), which results in impairment of DNA repair with the use of alkylating agents. To date, no therapeutic options have been available for improving survival for HGGs [26,27].

2.3. Neuroblastomas

Neuroblastomas are one of the major causes of death in pediatrics, accounting for about ~10–13% of all pediatric cancer cases [14,28]. Neuroblastomas originate from primordial neural crest cells, which form the adrenal medulla and sympathetic ganglia. Similar to glioblastomas, neuroblastomas also exhibit greater amplification of N-myc, which is associated with the expression of MRP and chromosome 1p deletion [28,29]. Overexpressed MRP on the neuroblastoma surface enhances the chemoresistance potential. Further, amplification of N-myc has downregulated the expression of CD44 receptors, which is a potential marker for aggressive tumor behavior. Additionally, low expression of Trk, a tyrosine kinase receptor, is associated with amplification of N-myc and even advanced stages of neuroblastomas. ALK amplification is another set of somatic mutations, accounting for ~14% of high-risk neuroblastomas [30]. Gain-of-function in ALK could drive the neuroblastoma but requires cooperation from MYCN amplification. Further, ALK upregulates the proto-oncogene RET and RET-driven sympathetic markers of the cholinergic lineage, which offer new therapeutic options, i.e., targeting both ALK and RET [30,31].

2.4. Ependymoma

Ependymoma (EPN) is one of the pediatric brain cancers which can occur in any part of the brain. The most popular originating location is the posterior fossa (cerebellum and brainstem), followed by supratentorial sites (cerebral hemispheres) and spinal cord [32,33]. The only option for treating ependymomas is surgery or radiation, as standard chemotherapy is ineffective. The molecular characteristics of EPNs are heterogeneous, mainly with dysregulation in growth factors such as epidermal growth factor receptor (EGFR), fibroblast growth factor receptor (FGFR), etc. EPNs originating from the posterior fossa (PF-EPN) are classified as PF-EPN-A and -B. PF-EPN-A is reported to be more deadly than PF-EPN-B [34]. Further, treatment with small molecule inhibitors, such as 3-deazaneplanocin A, causes degradation of PRC2 complex or with EZH2 (a GSK343 inhibitor), which competitively binds with S-adenosyl-L-methionine. EPNs originating from supratentorial (ST-EPNs) are reported to harbor fusion between RELA, NF-κB, and C11orf95 [35]. Further studies revealed that RELA fusion proteins alone could initiate the transformation of the neural stem cells [36]. Treatment options against ST-EPNs are mostly common chemotherapeutics such as temozolomide, vincristine, etc. [37] or HDAC inhibitors, including entinostat and vorinostat [38]. Ongoing research using preclinical models of ST-EPNs to evaluate potential druggable targets holds promise for developing treatment of tumors with RELA fusion proteins [32,34].

3. Nano-Based Approaches for Treating Pediatric Brain Cancers

Many research groups have emphasized utilizing nanotechnology to curb tumor progression, especially in pediatrics. In the present section, we will discuss nanotechnological advancements, especially for treating pediatric brain cancer. A schematic outline of diverse nanotherapeutic approached adopted for augmented pediatric brain cancer is depicted in Figure 1.

3.1. Nanotechnology and Blood–Brain Barrier

Due to its highly selective nature, the blood-brain-barrier (BBB) has become the first crucial barrier for many brain therapeutics and diagnostic entities. To develop a novel target-specific, an in-depth understanding of the physiology of BBB and overcoming strategies for nanoparticles is essential [39,40,41].

3.2. Physiology of BBB and Overcoming Strategies

BBB comprises highly specialized cells, which act as a protective barrier around the brain, especially in maintaining brain homeostasis. The cellular architecture of BBB mainly consists of brain capillary endothelial cells (BCECs), astrocytes, and pericytes [42]. Tight junctions between BCECs restrict the cellular diffusion of aqueous moieties. Further, in intact condition, BBB restricts the entry of ~99% of small drug molecules. BBB offers many transport mechanisms internally, i.e., via the transcellular lipophilic pathway, carrier-mediated transport (CMT), or receptor-mediated transport (RMT) [42,43]. However, the delivery of small molecules can be compromised by a large number of efflux pumps (such as adenosine triphosphate binding cassette transporters including multi-drug resistant protein (MRP) and p-glycoprotein (p-gp)), evade the foreign material into the bloodstream. Further, biological compounds, including inflammatory mediators (e.g., bradykinin, prostaglandin, vascular endothelial growth factors (VEGF)), signal receptors to increase BBB permeability [43].
Currently, drug delivery systems that are used in clinics are especially focused on local delivery. However, local delivery has limitations, including high rates of infection and excessive cerebrospinal fluid requirement. A non-invasive with direct delivery of therapeutic agents to the brain is the “intranasal route”, which is currently preferred but requires adjusting parameters such as dosage and positioning [42]. Transient opening of BBB can be achieved by biological (e.g., VEGF) or chemical stimuli (e.g., mannitol, oleic acid, cyclodextrins) but could have non-specific uptake, causing unwanted side effects. Therefore, an ideal approach to disrupting BBB is essential, one that would be controllable, reversible, specific, transient, and selective [43].
One of the best and most convenient approaches for drug delivery is intravenous administration with proper dosing. As discussed earlier, with the proper utilization of transporter proteins, specific receptors could be utilized for active targeting of nanoparticles-based drug deliveries. Further, disruption of BBB using either biological or chemical stimuli or nano-drug delivery systems, using either passive or active targeting, could achieve better BBB transport [41,42]. Moreover, most of the nano-drug delivery systems are lipophilic in nature, which is a crucial feature in bypassing the BBB.
Nanomedicines are small-sized nanocarriers that have been adopted to cure brain illnesses, including brain cancer and Alzheimer’s disease (AD). Functionalized nanoparticles are considered the most useful applicable approach to delivering these recommended drugs to the affected part of the brain. Nanomedicines have a set of unique properties that enable them to deliver anticancer drugs at target sites in the brain. Nanomedicines have the advantages of reduced dimensions and increased biocompatibility that facilitate the easy transport of therapeutic substances into the brain. Small-size nanomedicines can easily interact with the proteins and molecules on the cell surface as well as inside the cell. NP-functionalized nanomedicines have central core structures that ensure the encapsulation or conjugation of drugs and provide protection and prolonged circulation in the blood (Figure 2). Nanomedicines are also specialized to target cells or even an intracellular compartment and thus can deliver the drug at a predetermined dosage directly to the pathological site. Nanomedicines can minimize the dose and frequency and then improve patient compliance. Regardless of some clinical issues, nanomedicines have potential advantages of favorability to the brain, greater stability, biocompatibility and biodegradability, protection from enzymatic degradation, increased half-life, improved bioavailability, and controlled release over other conventional ways of drug delivery to the brain to cure AD (Figure 2) [44].

3.3. Nanoformulations Used for the Treatment of Medulloblastoma

Nanoparticle-based approaches are potential treatment options for pediatric medulloblastoma. NPs-based strategies mainly aim to improve the delivery of drugs by active or passive targeting and improve BBB crossing while reducing the side effects to surrounding healthy tissues.
Recently, herpes simplex virus type I thymidine kinase gene encoded plasmid loaded with poly (beta-amino ester) (PBAE) nanoparticles for gene therapy to medulloblastoma (MB) and atypical teratoid/rhabdoid tumors (AT/RT). The treatment with gene-encapsulated nanoparticles showed controlled apoptosis in transfected cells. In MB and AT/RT implanted mice, the gene therapy exhibited greater overall median survival [45]. An engineered biomimetic nanoparticle with dual targeting was designed to target the cancer stem-like cell population in sonic hedgehog medulloblastoma (SHH-MB). Treatment failure and poor outcomes are the significant struggles associated with the SHH-MB. High-density lipoprotein-mimetic nanoparticles (eHNPs) were used to cross BBB and load SHH inhibitors for the effective treatment of SHH-MB. Multi-component eHNPs were designed using microfluidic technology and are encapsulated with apolipoprotein A1, anti-CD15, and LDE225 (SHH inhibitor). eHNP-A1 improves the stability of the drug and has a therapeutic effect by SR-B1-mediated intracellular cholesterol depletion in tumor cells. These multifunctional nanoparticles exhibited promising effects in SHH-MB treatment and are applicable to other drugs that cannot cross BBB and have low bioavailability [46].
In a study, researchers developed a brain tumor model consisting of DAOY (MB cell lines) aggregates and cerebellum slices for evaluation of a poly(glycerol-adipate) (PGA) nanoparticle drug delivery system. PGA nanoparticles exhibited higher uptake than normal host cells. This novel tumor model suggests the effective evaluation of a drug delivery system between tumor cells and brain cells [47]. Kumar et al. [48] used Hedgehog inhibitor MDB5 and BRD4/PI3K dual inhibitor SF2523 to obtain the synergistic inhibition of medulloblastoma cell lines and to prevent resistance. They designed mPEG-b-PCC-g-DC copolymer-based NPs for effective loading of MDB5 and SF2523. NPs exhibit sustained release of loaded molecules. Targeted NPs were prepared by mixing COG-133-PEG-b-PBC and mPEG-b-PCC-g-DC copolymer and were found to be efficient in the reduction of tumors in orthotopic SHH-MB tumor-bearing NSG mice [48].

3.4. Nanoformulations Used for the Treatment of Glioma

Gliomas are the most common malignancy affecting the central nervous system. The primary treatment barrier for this disease is the difficulty of crossing the BBB by drug molecules. Nanoparticle-based approaches are familiar in overcoming these issues.
In a study, Temozolomide, an anti-glioma drug, was loaded on liposomes using proliposomes. The liposomes showed a slow release of temozolomide compared to the drug solution. The loading of temozolomide in liposomes improves the pharmacokinetic parameters compared to pure drug solution. Liposomes prolong the circulation time and improve the area under the curve (AUC). The biodistribution after IV injection revealed that the drug accumulation in the heart and lung is decreasing, and the concentration of the drug is increasing in the brain [49]. In another study, a targeted drug delivery platform was designed with PAMAM-PEG and transferrin for the encapsulation of temozolomide for the effective targeting of glioma stem cells. Glioma stem cells are responsible for the development of resistance. High cellular uptake and cytotoxicity were observed with transferrin-targeted temozolomide nanoparticles. The nanoparticle effectively crossed the BBB and delivered the drug specifically to the tumor. The PAMAM-PEG-trf nanoparticles induced potent cell apoptosis in drug-resistant glioma stem cells [50].
Gu et al. [51] designed MT1-AF7p peptide-decorated paclitaxel-loaded PEG-PLA nanoparticles for glioma management. MT1-AF7p peptide has high binding to membrane type-1 matrix metalloproteinase (MT1-MMP) overexpressed on glioma cells. To improve the penetration of nanoparticles to glioma cells, the nanoparticles were co-administered with Tumor-homing and penetrating peptide iRGD. In C6 glioma cells, the peptide-decorated NPs showed significant cellular uptake via energy-dependent macropinocytosis and lipid raft-mediated endocytosis compared to non-peptide NPs. The nanoparticle’s extravasation across BBB and accumulation in glioma parenchyma was improved significantly in in vivo imaging and glioma distribution with MT1-AF7p functionalization and iRGD co-administration. Intracranial C6 glioma-bearing nude mice exhibited higher survival time with MT1-AF7p functionalization and iRGD co-administration (Figure 3) [51]. Bhunia et al. [52] have tailored a large amino acid transporter-1 (LAT1) conjugated nanometric liposomal carriers functionalized with amphiphile L-3,4-dihydroxyphenylalanine (L-DOPA) (Amphi-DOPA). Glioma-bearing mice showed higher uptake of NIR-dye labeled Amphi-DOPA to brain tissue. WP1066 labeled Amphi-DOPA enhanced the overall survivability of glioma-bearing C57BL/6J mice by 60% compared to the untreated group [52].
A red blood cell membrane-coated nanoparticle (RBCNP) with a neurotoxin-derived targeting ligand was designed for brain-targeted drug delivery. RBCNP can provide the biological function of natural cell membranes and desirable properties for drug delivery. The targeting moiety CDX peptide derived from candoxin has a high binding affinity to nicotinic acetylcholine receptors overexpressed on brain endothelial cells decorated on RBCNP. In vitro and in vivo results suggest that RBCNP-CDX has promising brain-targeting efficiency. In glioma mouse models, Dox-loaded RBCNP-CDX NPs showed superior therapeutic efficacy with less toxicity [53].
AS1411 aptamer functionalized poly (L-γ-glutamyl-glutamine)-paclitaxel (PGG-PTX) nanoconjugates were designed to achieve active targeting and optimized solubilization of paclitaxel. The tumor uptake of the nanoconjugate was mediated through nucleolin receptors, over-expressed in glioblastoma cells and neovascular endothelial cells. The in vivo fluorescence imaging and biodistribution studies suggest that the AS1411-PGG-PTX has higher tumor accumulation than PGG-PTX. In glioma-bearing mice, this nanoconjugate exhibited prolonged median survival time and most tumor cell apoptosis compared to PGG-PTX [54]. A dual-targeted liposome was designed for the co-delivery of doxorubicin (DOX) and vincristine (VCR) for glioma management. T7 (a ligand of transferrin receptors) and DA7R (ligand of VEGFR 2) peptides were used to target glioma. The dual targeting strategy exhibited higher cellular uptake when compared to the single targeting strategy. The dual targeting and dual drug delivery showed the most favorable anti-glioma effect in vivo [55].
Nanocarrier-based immunotherapy has emerged as a promising approach for the treatment of various cancers, including glioblastoma. Gliomas are highly invasive. They usually infiltrate the normal tissues and make surgical removal of the tissue difficult. Immunotherapy in glioma can target and eliminate the infiltrating glioma cells to the neighboring tissues. Gliomas usually lead to an immunosuppressive environment and prevent normal immune cell reactions against cancer cells. Hence, immunotherapy is the best strategy to overcome the challenges and activate the immune system against tumor formation. Gliomas are known for their molecular and cellular heterogeneity. Immunotherapy can target specific antigens expressed on tumor cells, including tumor-associated antigens and neoantigens, addressing the diversity of cancer cells within the tumor [56,57].
Immunotherapy coupled with nanocarrier-based drug delivery presents a revolutionary approach to treating glioblastoma. Overcoming the BBB, nanocarriers enabled targeted delivery of immunotherapeutic agents, and enhanced drug bioavailability while minimizing systemic side effects are the advantages of nanocarrier-based immunotherapy in gliomas. The personalized nature of nanocarrier systems, tailored to individual tumor characteristics, promises a more effective and precise treatment. By encapsulating immunomodulatory agents, these carriers boost the immune response within the tumor microenvironment. Additionally, the ability to administer combination therapies and mitigate systemic toxicity underscores the potential of this innovative strategy in overcoming the challenges posed by glioblastoma, offering hope for improved patient outcomes. Checkpoint inhibitors, cytokines, and antigenic peptides are major immunomodulatory agents that can be delivered using nanocarriers to modulate the immune system’s response against glioblastoma [58,59].
Kuang et al. studied the effect of macrophage-directed immunotherapy with chemotherapy in orthotopic glioma. Doxorubicin and an immune checkpoint inhibitor (1-methyltryptophan, 1MT) were loaded on mesoporous silica nanoparticles modified with iRGD. The nanocarrier showed the ability to penetrate the BBB and accumulate drug molecules. The nanocarrier leads to the activation of cytotoxic CD8+ T lymphocytes and ions of CD4+ T cells in both GL261 cells cocultured with splenocytes in vitro and GL261-luc orthotopic tumors in vivo. The expression of antitumor cytokines was found to be upregulated, while protumor proteins were downregulated in the tumor tissues [60].
The Applications of Nanoparticles for medulloblastoma and glioma therapy are summarized in Table 1.

3.5. Nanoformulations Used for the Treatment of Neuroblastoma

Neuroblastoma (NB) is a complex pediatric tumor that originates from the neural crest and is the most common extracranial solid tumor in children, accounting for 15% of pediatric tumor-related deaths and 8–10% of all childhood malignancies. Most of the malignant cells are found in the adrenal medulla, but they can manifest as localized or metastatic tumors in the paraspinal ganglia, thorax, pelvis, and neck [67]. There are now several alternative therapeutic options for localized NB at different stages. Since most children are inoperable at the time of diagnosis due to metastases, even though complete resection of the primary NB is expected to greatly improve overall survival, the primary treatments in most cases still involve radiotherapy, chemotherapy, immunotherapy, differentiation-inducing therapy, and autologous hematopoietic stem cell transplantation [68]. Nano-based methods for treating pediatric neuroblastoma are an emerging area of interest and possible treatment plans in the upcoming years.
Treatment for neuroblastoma, a cancer of the nerve tissue that affects young children, is still challenging. When it comes to targeted therapy, imaging, and drug distribution for neuroblastoma, nano-based methods have distinct benefits. Solid lipid NPs, polymer micelles, nanoliposomes, nanocapsules, nanospheres, and nanomedicines are the primary forms of nanoparticles (NPs) [40]. Graphene oxide nanoribbons were developed by Mari et al. [69] to investigate their effects on human neuroblastoma cells. In one of the cell lines, they discovered that these nanoribbons stimulated autophagy and increased the synthesis of reactive oxygen species (ROS) within the first 48 h of exposure. Both cell lines observed a brief increase in ROS generation and autophagy at low doses; however, neither cell growth inhibition nor cell death was brought about by these effects [69]. Li et al. [70] studied zinc oxide nanoparticles (ZnO NPs) that were produced using Clausena lansium Peel aqueous extracts and zinc nitrate. These developed ZnO NPs were found to affect the regulation of autophagy and apoptotic proteins in SH-SY5Y neuroblastoma cells, leading to DNA damage, ROS generation, decreased cell stability, and viability. The utilization of N-acetyl-L-cysteine (NAC) was shown to mitigate ROS effects and prevent apoptosis, suggesting that ZnO NPs have the potential to induce cell death in neuroblastoma cells through the production of intracellular ROS. Kalashnikova et al. [71] developed and tested nanoceria and dextran-nanoceria formulations loaded with curcumin for treating childhood neuroblastoma. The formulations effectively killed neuroblastoma cells, particularly in MYCN-amplified cases, without damaging the healthy cells. This nanoparticle-induced oxidative stress stabilized HIF-1α and triggered the caspase-dependent apoptosis. These results offer a promising alternative to traditional drug therapies for aggressive cancers. Mohammadniaei et al. [72] developed a promising cell differentiation therapy using silver-coated bismuth selenide nanoparticles. The developed nanoparticles can be functionalized with a unique RNA structure to inhibit micro-RNA-17 and release retinoic acid, facilitating the transformation of cancer cells into neurons. This innovative research reports on the hydrophobicity challenges and offers a new method for drug delivery and real-time monitoring of the differentiation process, potentially advancing diagnostic and therapeutic agents. Zhang et al. [73] have demonstrated that Nab-paclitaxel exerted significant cytotoxicity against various pediatric solid tumor cell lines in vitro, with dose-dependent effects studied. In vivo studies on rhabdomyosarcoma and neuroblastoma xenograft models showed antitumor activity and increased survival in the metastatic model. Nab-paclitaxel induced tumor cell-cycle arrest and apoptosis, and its higher tumor/plasma drug ratio favored its efficacy compared to paclitaxel, even in paclitaxel-resistant relapsed tumors [73].
The applications of nanoparticles for neuroblastoma therapy are summarized in Table 2.

3.6. Nanoformulations Used for the Treatment of Retinoblastoma

Retinoblastoma is a disease in which malignant (cancer) cells form in the tissues of the retina. Retinoblastoma can expand to other parts of the body, such as the brain and spine [88]. Pediatric retinoblastoma (RB) is an uncommon and occasionally inherited malignancy. Because of alterations in the tumor-suppressor genes and the lack of a targeted, efficient, and cost-effective therapy, retinoblastoma is an uncommon form of cancer that is difficult to diagnose and treat. As such, there is a critical need for innovative treatments to address these issues [89]. External beam radiation, episcleral plaque radiation, cryotherapy, enucleation, and photocoagulation were conventional therapies for children with RB [90]. Ocular malignancies present unique problems, and improved penetration of the retinal pigment epithelium by monotherapies is required [91].
Several nano-applications have been investigated recently to overcome these obstacles. The application of nanotechnologies in the detection and management of cancers and eye conditions has grown rapidly in recent years [92,93,94]. Among the most useful nanotechnology-based ocular delivery methods include nanoliposomes, polymeric nanoparticles (PNPs), nanocapsules, nanocages, nano-micelles, nano-dendrimers, and nanohydrogels, which offer several benefits over standard diagnostics and treatments [95,96,97]. Moradi et al. [98] assess the combined effects of gold nanoparticles (Au-NPs) and ultrasonic hyperthermia on Y79 cells. Cells were exposed to ultrasonic irradiation with or without 60 nm Au-NPs, and their viability was measured 48 h later. Results showed that hyperthermia alone reduced cell viability after 4 min, while in the presence of Au-NPs, this effect was observed after 4.5 min. Higher Au-NP concentrations increased cytotoxicity. This research concludes that the use of Au-NPs enhances the sensitivity of cells to hyperthermia induced by ultrasound [98]. A multifunctional nanoparticle system has been developed for the diagnosis and treatment of retinoblastoma. The nanoparticles, consisting of magnetic hollow mesoporous gold nanocages (AuNCs) loaded with muramyl dipeptide (MDP) and perfluoropentane (PFP), enable advanced imaging (photoacoustic, ultrasound, and magnetic resonance) for diagnosis and enhance low-intensity focused ultrasound (LIFU) therapy. These nanoparticles, when combined with LIFU, effectively target and treat RB tumors, leading to tumor cell death, while MDP activates dendritic cells (DCs) for improved immune response. The multifunctional nanoparticles offer potential for multimodal imaging-guided LIFU therapy and show promise for RB treatment with high safety [99]. Silver nanoparticles (AgNPs) are increasingly used in medical and commercial products due to their potent antibacterial properties. Rajanahalli et al. [100] have investigated the impact of AgNPs on mouse embryonic stem cells (mESCs). They revealed that AgNPs with different surface coatings altered cell morphology, induced cell cycle arrest at G1 and S phases, and reduced pluripotency marker Oct4A while promoting the expression of stress-related isoforms. The findings suggested that AgNPs’ toxicity is linked to excessive reactive oxygen species (ROS) production, with polysaccharide coating mitigating this effect [100]. Qu et al. [98,101] developed EpCAM-conjugated mesoporous silica nanoparticles (EpCMSN) to effectively deliver carboplatin (CRB) for the treatment of retinoblastoma (RB), a rare eye tumor. EpCMSN demonstrated enhanced cellular uptake and superior anticancer effects compared to free CRB, with a significantly lower IC50 value of 1.38 μg/mL. The specific receptor-mediated internalization of EpCMSN, targeting EpCAM receptors, suggests a promising approach for targeted treatment of RB and other ocular malignancies [101]. Photothermal therapy, with its minimal invasiveness and high specificity, addresses issues associated with traditional drug treatment for tumors. However, its limited tissue penetration hinders clinical application. Using a nano-platform comprising liposomes and indocyanine green (ICG) introduced a novel strategy for treating retinoblastoma by enhancing ICG stability and enabling imaging-guided photothermal therapy, making use of the eye’s transparency to infrared light. In this study, ICG-loaded liposome nanoparticles (ILP) were developed, offering targeted tumor treatment and improved imaging capabilities, holding promise for image-guided tumor phototherapy. Figure 4 depicts the fluorescence and photoacoustic imaging capabilities of ILP [102]. Cerium-doped titania nanoparticles (Ce-doped TiO2) were studied by Kartha et al. [103] using a cost-effective sol-gel method, and their enhanced photodynamic anticancer effects were evaluated on Y79 retinoblastoma cells. The study investigated the structural and optical properties of pure and Ce-doped TiO2, revealing cerium’s presence through X-ray diffraction and Raman spectra. Additionally, microscopy analysis showed that both TiO2 variants exhibited spherical shapes. The findings indicated that cerium doping in TiO2 enhances its photodynamic anticancer activity [103]. The applications of nanoparticles for retinoblastoma therapy are discussed in Table 3.

4. Clinical Trials

Pediatric brain tumors are some of the most devastating childhood diseases, with high mortality rates and significant long-term morbidity for survivors. Conventional treatment options like surgery, radiation, and chemotherapy often have severe side effects and limited efficacy. As mentioned in Table 4 (retrieved from https://clinicaltrials.gov/, accessed on 15 February 2024), emerging nanotherapeutic strategies offer a promising avenue for improving the treatment of pediatric brain tumors by overcoming these limitations. Nanoparticles, with their unique size and properties, can be designed to target tumor cells more effectively, deliver drugs with greater precision, and reduce systemic toxicity. Here is a brief overview of some ongoing clinical studies exploring nano-therapeutic strategies against pediatric brain tumors.

5. Conclusions

Pediatric brain tumors are considered the most frequent type of pediatric cancer, and they pose a tremendous therapeutic challenge owing to their tendency to infiltrate and disseminate to surrounding tissues, restricting the use of surgery as a feasible mono-therapeutic strategy. Furthermore, the difficulty in delivering medications to the brain tumor site in effective therapeutic concentrations while evading the blood–brain barrier (BBB) represents another challenge for cancer conquering. Consequently, nanomedicines have emerged as a promising therapeutic approach to circumvent the hurdles encountered with conventional therapy, along with improving the bioavailability of drug payloads. Nanotechnology-based delivery systems can effectively cross the BBB, and when decorated with receptors that are overexpressed both by BBB-building cells and cancer cells, they can discriminate cancer cells from surrounding healthy ones, thus directing the therapeutic agents towards malignant cells. However, various challenges must be carefully considered, including biocompatibility issues and clearance modulation. Nonetheless, various strategies have been implemented in recent years to overcome these drawbacks, and, along with the growing body of knowledge in the molecular genetics of brain tumors, the scientific community is unquestionably close to a major breakthrough in the development of efficient, safe, and low-cost nanosystems capable of imaging and treating brain cancers without inflicting remarkable damage to healthy tissue.

Author Contributions

Conceptualization, A.V.V.V.R.K., H.V.G. and S.M.D.R.; validation, A.P.J., M.K. and R.A.M.O.; formal analysis, G.K.K., A.S.A.L., A.M. and S.M.D.R.; resources, P.T.K., H.V.G. and A.M.; data curation, A.P.J., M.K. and R.A.M.O.; writing—original draft preparation, A.V.V.V.R.K., G.K.K., P.T.K. and A.P.J.; writing—review and editing, P.T.K., M.K., R.A.M.O., A.S.A.L., and A.M.; visualization, A.V.V.V.R.K., G.K.K. and A.S.A.L.; supervision, H.V.G. and S.M.D.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Mueller, S.; Chang, S. Pediatric brain tumors: Current treatment strategies and future therapeutic approaches. Neurotherapeutics 2009, 6, 570–586. [Google Scholar] [CrossRef] [PubMed]
  2. Moin, A.; Rizvi, S.M.D.; Hussain, T.; Gowda, D.V.; Subaiea, G.M.; Elsayed, M.M.A.; Ansari, M.; Alanazi, A.S.; Yadav, H. Current Status of Brain Tumor in the Kingdom of Saudi Arabia and Application of Nanobiotechnology for Its Treatment: A Comprehensive Review. Life 2021, 11, 421. [Google Scholar] [CrossRef] [PubMed]
  3. Abedalthagafi, M.; Mobark, N.; Al-Rashed, M.; AlHarbi, M. Epigenomics and immunotherapeutic advances in pediatric brain tumors. NPJ Precis. Oncol. 2021, 5, 34. [Google Scholar] [CrossRef] [PubMed]
  4. Udaka, Y.T.; Packer, R.J. Pediatric Brain Tumors. Neurol. Clin. 2018, 36, 533–556. [Google Scholar] [CrossRef] [PubMed]
  5. Yang, S.; Wallach, M.; Krishna, A.; Kurmasheva, R.; Sridhar, S. Recent Developments in Nanomedicine for Pediatric Cancer. J. Clin. Med. 2021, 10, 1437. [Google Scholar] [CrossRef] [PubMed]
  6. Pui, C.H.; Gajjar, A.J.; Kane, J.R.; Qaddoumi, I.A.; Pappo, A.S. Challenging issues in pediatric oncology. Nat. Rev. Clin. Oncol. 2011, 8, 540–549. [Google Scholar] [CrossRef] [PubMed]
  7. Surendiran, A.; Sandhiya, S.; Pradhan, S.C.; Adithan, C. Novel applications of nanotechnology in medicine. Indian J. Med. Res. 2009, 130, 689–701. [Google Scholar] [PubMed]
  8. Emerich, D.F.; Thanos, C.G. Nanotechnology and medicine. Expert Opin. Biol. Ther. 2003, 3, 655–663. [Google Scholar] [CrossRef]
  9. Ando, H.; Kobayashi, S.; Abu Lila, A.S.; Eldin, N.E.; Kato, C.; Shimizu, T.; Ukawa, M.; Kawazoe, K.; Ishida, T. Advanced therapeutic approach for the treatment of malignant pleural mesothelioma via the intrapleural administration of liposomal pemetrexed. J. Control. Release 2015, 220, 29–36. [Google Scholar] [CrossRef]
  10. Abu Lila, A.S.; Ishida, T.; Kiwada, H. Recent advances in tumor vasculature targeting using liposomal drug delivery systems. Expert Opin. Drug Deliv. 2009, 6, 1297–1309. [Google Scholar] [CrossRef]
  11. Knudson, A.G., Jr. Genetics and the etiology of childhood cancer. Pediatr. Res. 1976, 10, 513–517. [Google Scholar] [CrossRef] [PubMed]
  12. Castaneda, M.; den Hollander, P.; Kuburich, N.A.; Rosen, J.M.; Mani, S.A. Mechanisms of cancer metastasis. Semin. Cancer Biol. 2022, 87, 17–31. [Google Scholar] [CrossRef] [PubMed]
  13. Liu, K.-W.; Pajtler, K.W.; Worst, B.C.; Pfister, S.M.; Wechsler-Reya, R.J. Molecular mechanisms and therapeutic targets in pediatric brain tumors. Sci. Signal. 2017, 10, eaaf7593. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, S.; Chung, D.H. Pediatric solid malignancies: Neuroblastoma and Wilms’ tumor. Surg. Clin. N. Am. 2006, 86, 469–487. [Google Scholar] [CrossRef] [PubMed]
  15. Dhall, G. Medulloblastoma. J. Child. Neurol. 2009, 24, 1418–1430. [Google Scholar] [CrossRef] [PubMed]
  16. Millard, N.E.; De Braganca, K.C. Medulloblastoma. J. Child. Neurol. 2016, 31, 1341–1353. [Google Scholar] [CrossRef] [PubMed]
  17. Packer, R.J.; Cogen, P.; Vezina, G.; Rorke, L.B. Medulloblastoma: Clinical and biologic aspects. Neuro-Oncology 1999, 1, 232–250. [Google Scholar] [CrossRef] [PubMed]
  18. Kijima, N.; Kanemura, Y. Molecular Classification of Medulloblastoma. Neurol. Med. Chir. 2016, 56, 687–697. [Google Scholar] [CrossRef]
  19. Rausch, T.; Jones, D.T.; Zapatka, M.; Stütz, A.M.; Zichner, T.; Weischenfeldt, J.; Jäger, N.; Remke, M.; Shih, D.; Northcott, P.A.; et al. Genome sequencing of pediatric medulloblastoma links catastrophic DNA rearrangements with TP53 mutations. Cell 2012, 148, 59–71. [Google Scholar] [CrossRef]
  20. Lee, M.J.; Hatton, B.A.; Villavicencio, E.H.; Khanna, P.C.; Friedman, S.D.; Ditzler, S.; Pullar, B.; Robison, K.; White, K.F.; Tunkey, C.; et al. Hedgehog pathway inhibitor saridegib (IPI-926) increases lifespan in a mouse medulloblastoma model. Proc. Natl. Acad. Sci. USA 2012, 109, 7859–7864. [Google Scholar] [CrossRef]
  21. Kim, J.; Tang, J.Y.; Gong, R.; Lee, J.J.; Clemons, K.V.; Chong, C.R.; Chang, K.S.; Fereshteh, M.; Gardner, D.; Reya, T.; et al. Itraconazole, a commonly used antifungal that inhibits Hedgehog pathway activity and cancer growth. Cancer Cell 2010, 17, 388–399. [Google Scholar] [CrossRef]
  22. Roussel, M.F.; Stripay, J.L. Modeling pediatric medulloblastoma. Brain Pathol. 2020, 30, 703–712. [Google Scholar] [CrossRef] [PubMed]
  23. Bolin, S.; Borgenvik, A.; Persson, C.U.; Sundström, A.; Qi, J.; Bradner, J.E.; Weiss, W.A.; Cho, Y.-J.; Weishaupt, H.; Swartling, F.J. Combined BET bromodomain and CDK2 inhibition in MYC-driven medulloblastoma. Oncogene 2018, 37, 2850–2862. [Google Scholar] [CrossRef]
  24. Borgenvik, A.; Čančer, M.; Hutter, S.; Swartling, F.J. Targeting MYCN in Molecularly Defined Malignant Brain Tumors. Front. Oncol. 2020, 10, 626751. [Google Scholar] [CrossRef]
  25. Hussain, T.; Paranthaman, S.; Rizvi, S.M.D.; Moin, A.; Gowda, D.V.; Subaiea, G.M.; Ansari, M.; Alanazi, A.S. Fabrication and Characterization of Paclitaxel and Resveratrol Loaded Soluplus Polymeric Nanoparticles for Improved BBB Penetration for Glioma Management. Polymers 2021, 13, 3210. [Google Scholar] [CrossRef] [PubMed]
  26. MacDonald, T.J.; Aguilera, D.; Kramm, C.M. Treatment of high-grade glioma in children and adolescents. Neuro-Oncology 2011, 13, 1049–1058. [Google Scholar] [CrossRef]
  27. Minturn, J.E.; Fisher, M.J. Gliomas in children. Curr. Treat. Options Neurol. 2013, 15, 316–327. [Google Scholar] [CrossRef] [PubMed]
  28. Maris, J.M. Recent advances in neuroblastoma. N. Engl. J. Med. 2010, 362, 2202–2211. [Google Scholar] [CrossRef] [PubMed]
  29. Mei, H.; Wang, Y.; Lin, Z.; Tong, Q. The mTOR signaling pathway in pediatric neuroblastoma. Pediatr. Hematol. Oncol. 2013, 30, 605–615. [Google Scholar] [CrossRef]
  30. Louis, C.U.; Shohet, J.M. Neuroblastoma: Molecular pathogenesis and therapy. Annu. Rev. Med. 2015, 66, 49–63. [Google Scholar] [CrossRef]
  31. Mora, J. Dinutuximab for the treatment of pediatric patients with high-risk neuroblastoma. Expert Rev. Clin. Pharmacol. 2016, 9, 647–653. [Google Scholar] [CrossRef] [PubMed]
  32. Vitanza, N.A.; Partap, S. Pediatric Ependymoma. J. Child. Neurol. 2016, 31, 1354–1366. [Google Scholar] [CrossRef] [PubMed]
  33. Kilday, J.P.; Rahman, R.; Dyer, S.; Ridley, L.; Lowe, J.; Coyle, B.; Grundy, R. Pediatric ependymoma: Biological perspectives. Mol. Cancer Res. 2009, 7, 765–786. [Google Scholar] [CrossRef] [PubMed]
  34. Merchant, T.E.; Fouladi, M. Ependymoma: New therapeutic approaches including radiation and chemotherapy. J. Neurooncol. 2005, 75, 287–299. [Google Scholar] [CrossRef] [PubMed]
  35. Thorp, N.; Gandola, L. Management of Ependymoma in Children, Adolescents and Young Adults. Clin. Oncol. R Coll. Radiol. 2019, 31, 162–170. [Google Scholar] [CrossRef] [PubMed]
  36. Arabzade, A.; Zhao, Y.; Varadharajan, S.; Chen, H.C.; Jessa, S.; Rivas, B.; Stuckert, A.J.; Solis, M.; Kardian, A.; Tlais, D.; et al. ZFTA-RELA Dictates Oncogenic Transcriptional Programs to Drive Aggressive Supratentorial Ependymoma. Cancer Discov. 2021, 11, 2200–2215. [Google Scholar] [CrossRef] [PubMed]
  37. Rudà, R.; Bosa, C.; Magistrello, M.; Franchino, F.; Pellerino, A.; Fiano, V.; Trevisan, M.; Cassoni, P.; Soffietti, R. Temozolomide as salvage treatment for recurrent intracranial ependymomas of the adult: A retrospective study. Neuro-Oncology 2016, 18, 261–268. [Google Scholar] [CrossRef]
  38. Antonelli, R.; Jiménez, C.; Riley, M.; Servidei, T.; Riccardi, R.; Soriano, A.; Roma, J.; Martínez-Saez, E.; Martini, M.; Ruggiero, A.; et al. CN133, a Novel Brain-Penetrating Histone Deacetylase Inhibitor, Hampers Tumor Growth in Patient-Derived Pediatric Posterior Fossa Ependymoma Models. Cancers 2020, 12, 1922. [Google Scholar] [CrossRef]
  39. Nehra, M.; Uthappa, U.T.; Kumar, V.; Kumar, R.; Dixit, C.; Dilbaghi, N.; Mishra, Y.K.; Kumar, S.; Kaushik, A. Nanobiotechnology-assisted therapies to manage brain cancer in personalized manner. J. Control. Release 2021, 338, 224–243. [Google Scholar] [CrossRef]
  40. Aleassa, E.M.; Xing, M.; Keijzer, R. Nanomedicine as an innovative therapeutic strategy for pediatric cancer. Pediatr. Surg. Int. 2015, 31, 611–616. [Google Scholar] [CrossRef]
  41. Rodríguez-Nogales, C.; González-Fernández, Y.; Aldaz, A.; Couvreur, P.; Blanco-Prieto, M.J. Nanomedicines for Pediatric Cancers. ACS Nano 2018, 12, 7482–7496. [Google Scholar] [CrossRef]
  42. Pandit, R.; Chen, L.; Götz, J. The blood-brain barrier: Physiology and strategies for drug delivery. Adv. Drug Deliv. Rev. 2020, 165-166, 1–14. [Google Scholar] [CrossRef] [PubMed]
  43. Tang, W.; Fan, W.; Lau, J.; Deng, L.; Shen, Z.; Chen, X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem. Soc. Rev. 2019, 48, 2967–3014. [Google Scholar] [CrossRef] [PubMed]
  44. Khan, N.H.; Mir, M.; Ngowi, E.E.; Zafar, U.; Khakwani, M.M.A.K.; Khattak, S.; Zhai, Y.K.; Jiang, E.S.; Zheng, M.; Duan, S.F.; et al. Nanomedicine: A Promising Way to Manage Alzheimer’s Disease. Front. Bioeng. Biotechnol. 2021, 9, 630055. [Google Scholar] [CrossRef]
  45. Choi, J.; Rui, Y.; Kim, J.; Gorelick, N.; Wilson, D.R.; Kozielski, K.; Mangraviti, A.; Sankey, E.; Brem, H.; Tyler, B.; et al. Nonviral polymeric nanoparticles for gene therapy in pediatric CNS malignancies. Nanomedicine 2020, 23, 102115. [Google Scholar] [CrossRef] [PubMed]
  46. Kim, J.; Dey, A.; Malhotra, A.; Liu, J.; Ahn, S.I.; Sei, Y.J.; Kenney, A.M.; MacDonald, T.J.; Kim, Y. Engineered biomimetic nanoparticle for dual targeting of the cancer stem-like cell population in sonic hedgehog medulloblastoma. Proc. Natl. Acad. Sci. USA 2020, 117, 24205–24212. [Google Scholar] [CrossRef] [PubMed]
  47. Meng, W.; Kallinteri, P.; Walker, D.A.; Parker, T.L.; Garnett, M.C. Evaluation of poly (glycerol-adipate) nanoparticle uptake in an in vitro 3-D brain tumor co-culture model. Exp. Biol. Med. 2007, 232, 1100–1108. [Google Scholar] [CrossRef] [PubMed]
  48. Kumar, V.; Wang, Q.; Sethi, B.; Lin, F.; Coulter, D.W.; Dong, Y.; Mahato, R.I. Polymeric nanomedicine for overcoming resistance mechanisms in hedgehog and Myc-amplified medulloblastoma. Biomaterials 2021, 278, 121138. [Google Scholar] [CrossRef]
  49. Gao, J.; Wang, Z.; Liu, H.; Wang, L.; Huang, G. Liposome encapsulated of temozolomide for the treatment of glioma tumor: Preparation, characterization and evaluation. Drug Discov. Ther. 2015, 9, 205–212. [Google Scholar] [CrossRef]
  50. Sun, T.; Wu, H.; Li, Y.; Huang, Y.; Yao, L.; Chen, X.; Han, X.; Zhou, Y.; Du, Z. Targeting transferrin receptor delivery of temozolomide for a potential glioma stem cell-mediated therapy. Oncotarget 2017, 8, 74451–74465. [Google Scholar] [CrossRef]
  51. Gu, G.; Gao, X.; Hu, Q.; Kang, T.; Liu, Z.; Jiang, M.; Miao, D.; Song, Q.; Yao, L.; Tu, Y.; et al. The influence of the penetrating peptide iRGD on the effect of paclitaxel-loaded MT1-AF7p-conjugated nanoparticles on glioma cells. Biomaterials 2013, 34, 5138–5148. [Google Scholar] [CrossRef]
  52. Bhunia, S.; Vangala, V.; Bhattacharya, D.; Ravuri, H.G.; Kuncha, M.; Chakravarty, S.; Sistla, R.; Chaudhuri, A. Large Amino Acid Transporter 1 Selective Liposomes of l-DOPA Functionalized Amphiphile for Combating Glioblastoma. Mol. Pharm. 2017, 14, 3834–3847. [Google Scholar] [CrossRef] [PubMed]
  53. Chai, Z.; Hu, X.; Wei, X.; Zhan, C.; Lu, L.; Jiang, K.; Su, B.; Ruan, H.; Ran, D.; Fang, R.H.; et al. A facile approach to functionalizing cell membrane-coated nanoparticles with neurotoxin-derived peptide for brain-targeted drug delivery. J. Control. Release 2017, 264, 102–111. [Google Scholar] [CrossRef] [PubMed]
  54. Luo, Z.; Yan, Z.; Jin, K.; Pang, Q.; Jiang, T.; Lu, H.; Liu, X.; Pang, Z.; Yu, L.; Jiang, X. Precise glioblastoma targeting by AS1411 aptamer-functionalized poly (l-γ-glutamylglutamine)-paclitaxel nanoconjugates. J. Colloid Interface Sci. 2017, 490, 783–796. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, Y.; Zhai, M.; Chen, Z.; Han, X.; Yu, F.; Li, Z.; Xie, X.; Han, C.; Yu, L.; Yang, Y.; et al. Dual-modified liposome codelivery of doxorubicin and vincristine improve targeting and therapeutic efficacy of glioma. Drug Deliv. 2017, 24, 1045–1055. [Google Scholar] [CrossRef] [PubMed]
  56. Segura-Collar, B.; Hiller-Vallina, S.; de Dios, O.; Caamaño-Moreno, M.; Mondejar-Ruescas, L.; Sepulveda-Sanchez, J.M.; Gargini, R. Advanced Immunotherapies for Glioblastoma: Tumor Neoantigen Vaccines in Combination with Immunomodulators. Acta Neuropathol. Commun. 2023, 11, 79. [Google Scholar] [CrossRef] [PubMed]
  57. Bovenberg, M.S.S.; Degeling, M.H.; Tannous, B.A. Cell-Based Immunotherapy Against Gliomas: From Bench to Bedside. Mol. Ther. 2013, 21, 1297–1305. [Google Scholar] [CrossRef] [PubMed]
  58. Tang, L.; Zhang, M.; Liu, C. Advances in Nanotechnology-Based Immunotherapy for Glioblastoma. Front. Immunol. 2022, 13, 882257. [Google Scholar] [CrossRef] [PubMed]
  59. Šamec, N.; Zottel, A.; Paska, A.V.; Jovčevska, I. Nanomedicine and Immunotherapy: A Step Further towards Precision Medicine for Glioblastoma. Molecules 2020, 25, 490. [Google Scholar] [CrossRef]
  60. Kuang, J.; Song, W.; Yin, J.; Zeng, X.; Han, S.; Zhao, Y.P.; Tao, J.; Liu, C.J.; He, X.H.; Zhang, X.Z. IRGD Modified Chemo-Immunotherapeutic Nanoparticles for Enhanced Immunotherapy against Glioblastoma. Adv. Funct. Mater. 2018, 28, 1800025. [Google Scholar] [CrossRef]
  61. Ahmad, S.; Khan, I.; Pandit, J.; Emad, N.A.; Bano, S.; Dar, K.I.; Rizvi, M.M.A.; Ansari, M.D.; Aqil, M.; Sultana, Y. Brain Targeted Delivery of Carmustine Using Chitosan Coated Nanoparticles via Nasal Route for Glioblastoma Treatment. Int. J. Biol. Macromol. 2022, 221, 435–445. [Google Scholar] [CrossRef]
  62. Jafari, S.; Tavakoli, M.B.; Zarrabi, A. Lomustine Loaded Superparamagnetic Iron Oxide Nanoparticles Conjugated with Folic Acid for Treatment of Glioblastoma Multiforma (GBM). Iran. J. Pharm. Res. 2020, 19, 134. [Google Scholar] [CrossRef]
  63. Zhao, Y.Z.; Lin, Q.; Wong, H.L.; Shen, X.T.; Yang, W.; Xu, H.L.; Mao, K.L.; Tian, F.R.; Yang, J.J.; Xu, J.; et al. Glioma-Targeted Therapy Using Cilengitide Nanoparticles Combined with UTMD Enhanced Delivery. J. Control. Release 2016, 224, 112–125. [Google Scholar] [CrossRef]
  64. Kamali, M.; Webster, T.J.; Amani, A.; Hadjighassem, M.R.; Malekpour, M.R.; Tirgar, F.; Khosravani, M.; Adabi, M. Effect of Folate-Targeted Erlotinib Loaded Human Serum Albumin Nanoparticles on Tumor Size and Survival Rate in a Rat Model of Glioblastoma. Life Sci. 2023, 313, 121248. [Google Scholar] [CrossRef] [PubMed]
  65. Lakkadwala, S.; Singh, J. Co-Delivery of Doxorubicin and Erlotinib through Liposomal Nanoparticles for Glioblastoma Tumor Regression Using an in Vitro Brain Tumor Model. Colloids Surf. B Biointerfaces 2019, 173, 27–35. [Google Scholar] [CrossRef] [PubMed]
  66. Hassanzadeganroudsari, M.; Soltani, M.; Heydarinasab, A.; Apostolopoulos, V.; Akbarzadehkhiyavi, A.; Nurgali, K. Targeted Nano-Drug Delivery System for Glioblastoma Therapy: In Vitro and in Vivo Study. J. Drug Deliv. Sci. Technol. 2020, 60, 102039. [Google Scholar] [CrossRef]
  67. Mobasheri, T.; Rayzan, E.; Shabani, M.; Hosseini, M.; Mahmoodi Chalbatani, G.; Rezaei, N. Neuroblastoma-targeted nanoparticles and novel nanotechnology-based treatment methods. J. Cell Physiol. 2021, 236, 1751–1775. [Google Scholar] [CrossRef] [PubMed]
  68. Perkins, S.M.; Shinohara, E.T.; DeWees, T.; Frangoul, H. Outcome for children with metastatic solid tumors over the last four decades. PLoS ONE 2014, 9, e100396. [Google Scholar] [CrossRef]
  69. Mari, E.; Mardente, S.; Morgante, E.; Tafani, M.; Lococo, E.; Fico, F.; Valentini, F.; Zicari, A. Graphene Oxide Nanoribbons Induce Autophagic Vacuoles in Neuroblastoma Cell Lines. Int. J. Mol. Sci. 2016, 17, 1995. [Google Scholar] [CrossRef] [PubMed]
  70. Li, F.; Song, L.; Yang, X.; Huang, Z.; Mou, X.; Syed, A.; Bahkali, A.H.; Zheng, L. Anticancer and genotoxicity effect of (Clausena lansium (Lour.) Skeels) Peel ZnONPs on neuroblastoma (SH-SY5Y) cells through the modulation of autophagy mechanism. J. Photochem. Photobiol. B 2020, 203, 111748. [Google Scholar] [CrossRef]
  71. Kalashnikova, I.; Mazar, J.; Neal, C.J.; Rosado, A.L.; Das, S.; Westmoreland, T.J.; Seal, S. Nanoparticle delivery of curcumin induces cellular hypoxia and ROS-mediated apoptosis via modulation of Bcl-2/Bax in human neuroblastoma. Nanoscale 2017, 9, 10375–10387. [Google Scholar] [CrossRef]
  72. Mohammadniaei, M.; Yoon, J.; Choi, H.K.; Placide, V.; Bharate, B.G.; Lee, T.; Choi, J.-W. Multifunctional Nanobiohybrid Material Composed of Ag@Bi2Se3/RNA Three-Way Junction/miRNA/Retinoic Acid for Neuroblastoma Differentiation. ACS Appl. Mater. Interfaces 2019, 11, 8779–8788. [Google Scholar] [CrossRef]
  73. Zhang, L.; Marrano, P.; Kumar, S.; Leadley, M.; Elias, E.; Thorner, P.; Baruchel, S. Nab-paclitaxel is an active drug in preclinical model of pediatric solid tumors. Clin. Cancer Res. 2013, 19, 5972–5983. [Google Scholar] [CrossRef] [PubMed]
  74. Yesil-Celiktas, O.; Pala, C.; Cetin-Uyanikgil, E.O.; Sevimli-Gur, C. Synthesis of silica-PAMAM dendrimer nanoparticles as promising carriers in Neuro blastoma cells. Anal. Biochem. 2017, 519, 1–7. [Google Scholar] [CrossRef] [PubMed]
  75. Vignaroli, G.; Calandro, P.; Zamperini, C.; Coniglio, F.; Iovenitti, G.; Tavanti, M.; Colecchia, D.; Dreassi, E.; Valoti, M.; Schenone, S.; et al. Improvement of pyrazolo[3,4-d]pyrimidines pharmacokinetic properties: Nanosystem approaches for drug delivery. Sci. Rep. 2016, 6, 21509. [Google Scholar] [CrossRef] [PubMed]
  76. Pieper, S.; Onafuye, H.; Mulac, D.; Cinatl, J., Jr.; Wass, M.N.; Michaelis, M.; Langer, K. Incorporation of doxorubicin in different polymer nanoparticles and their anticancer activity. Beilstein J. Nanotechnol. 2019, 10, 2062–2072. [Google Scholar] [CrossRef]
  77. Bacanlı, M.; Eşi, M.Ö.; Erdoğan, H.; Sarper, M.; Erdem, O.; Özkan, Y. Evaluation of cytotoxic and genotoxic effects of paclitaxel-loaded PLGA nanoparticles in neuroblastoma cells. Food Chem. Toxicol. 2021, 154, 112323. [Google Scholar] [CrossRef] [PubMed]
  78. Viale, M.; Vecchio, G.; Monticone, M.; Bertone, V.; Giglio, V.; Maric, I.; Cilli, M.; Bocchini, V.; Profumo, A.; Ponzoni, M.; et al. Fibrin Gels Entrapment of a Poly-Cyclodextrin Nanocarrier as a Doxorubicin Delivery System in an Orthotopic Model of Neuroblastoma: Evaluation of In Vitro Activity and In Vivo Toxicity. Pharm. Res. 2019, 36, 115. [Google Scholar] [CrossRef]
  79. Wang, G.; Hu, W.; Chen, H.; Shou, X.; Ye, T.; Xu, Y. Cocktail Strategy Based on NK Cell-Derived Exosomes and Their Biomimetic Nanoparticles for Dual Tumor Therapy. Cancers 2019, 11, 560. [Google Scholar] [CrossRef]
  80. Piazzini, V.; Vasarri, M.; Degl’Innocenti, D.; Guastini, A.; Barletta, E.; Salvatici, M.C.; Bergonzi, M.C. Comparison of Chitosan Nanoparticles and Soluplus Micelles to Optimize the Bioactivity of Posidonia oceanica Extract on Human Neuroblastoma Cell Migration. Pharmaceutics 2019, 11, 655. [Google Scholar] [CrossRef]
  81. Zhao, J.; Yao, L.; Nie, S.; Xu, Y. Low-viscosity sodium alginate combined with TiO2 nanoparticles for improving neuroblastoma treatment. Int. J. Biol. Macromol. 2021, 167, 921–933. [Google Scholar] [CrossRef] [PubMed]
  82. Burgos-Panadero, R.; El Moukhtari, S.H.; Noguera, I.; Rodríguez-Nogales, C.; Martín-Vañó, S.; Vicente-Munuera, P.; Cañete, A.; Navarro, S.; Blanco-Prieto, M.J.; Noguera, R. Unraveling the extracellular matrix-tumor cell interactions to aid better targeted therapies for neuroblastoma. Int. J. Pharm. 2021, 608, 121058. [Google Scholar] [CrossRef]
  83. Patel, R.B.; Ye, M.; Carlson, P.M.; Jaquish, A.; Zangl, L.; Ma, B.; Wang, Y.; Arthur, I.; Xie, R.; Brown, R.J.; et al. Development of an In Situ Cancer Vaccine via Combinational Radiation and Bacterial-Membrane-Coated Nanoparticles. Adv. Mater. 2019, 31, e1902626. [Google Scholar] [CrossRef] [PubMed]
  84. Subramanian, C.; White, P.T.; Kuai, R.; Kalidindi, A.; Castle, V.P.; Moon, J.J.; Timmermann, B.N.; Schwendeman, A.; Cohen, M.S. Synthetic high-density lipoprotein nanoconjugate targets neuroblastoma stem cells, blocking migration and self-renewal. Surgery 2018, 164, 165–172. [Google Scholar] [CrossRef] [PubMed]
  85. Kulkarni, S.; Pandey, A.; Nikam, A.N.; Nannuri, S.H.; George, S.D.; Fayaz, S.M.A.; Vincent, A.P.; Mutalik, S. ZIF-8 nano confined protein-titanocene complex core-shell MOFs for efficient therapy of Neuroblastoma: Optimization, molecular dynamics and toxicity studies. Int. J. Biol. Macromol. 2021, 178, 444–463. [Google Scholar] [CrossRef] [PubMed]
  86. Lin, Y.S.; Chen, Y.; Tsai, Y.H.; Tseng, S.H.; Lin, K.S. In Vivo Imaging of Neuroblastomas Using GD2-Targeting Graphene Quantum Dots. J. Pediatr. Surg. 2021, 56, 1227–1232. [Google Scholar] [CrossRef] [PubMed]
  87. Di Paolo, D.; Pastorino, F.; Brignole, C.; Corrias, M.V.; Emionite, L.; Cilli, M.; Tamma, R.; Priddy, L.; Amaro, A.; Ferrari, D.; et al. Combined Replenishment of MiR-34a and Let-7b by Targeted Nanoparticles Inhibits Tumor Growth in Neuroblastoma Preclinical Models. Small 2020, 16, 1906426. [Google Scholar] [CrossRef]
  88. Dimaras, H.; Corson, T.W. Retinoblastoma, the visible CNS tumor: A review. J. Neurosci. Res. 2019, 97, 29–44. [Google Scholar] [CrossRef]
  89. Fabian, I.D.; Onadim, Z.; Karaa, E.; Duncan, C.; Chowdhury, T.; Scheimberg, I.; Ohnuma, S.I.; Reddy, M.A.; Sagoo, M.S. The management of retinoblastoma. Oncogene 2018, 37, 1551–1560. [Google Scholar] [CrossRef]
  90. Shields, C.L.; Shields, J.A.; De Potter, P.; Minelli, S.; Hernandez, C.; Brady, L.W.; Cater, J.R. Plaque radiotherapy in the management of retinoblastoma. Use as a primary and secondary treatment. Ophthalmology 1993, 100, 216–224. [Google Scholar] [CrossRef]
  91. Bhavsar, D.; Subramanian, K.; Sethuraman, S.; Krishnan, U.M. Management of retinoblastoma: Opportunities and challenges. Drug Deliv. 2016, 23, 2488–2496. [Google Scholar] [CrossRef] [PubMed]
  92. Wei, D.; Zhang, N.; Qu, S.; Wang, H.; Li, J. Advances in nanotechnology for the treatment of GBM. Front. Neurosci. 2023, 17, 1180943. [Google Scholar] [CrossRef] [PubMed]
  93. Barzegar Behrooz, A.; Talaie, Z.; Syahir, A. Nanotechnology-Based Combinatorial Anti-Glioblastoma Therapies: Moving from Terminal to Treatable. Pharmaceutics 2022, 14, 1697. [Google Scholar] [CrossRef] [PubMed]
  94. Gusmão, L.A.; Matsuo, F.S.; Barbosa, H.F.G.; Tedesco, A.C. Advances in nano-based materials for glioblastoma multiforme diagnosis: A mini-review. Front. Nanotechnol. 2022, 4, 836802. [Google Scholar] [CrossRef]
  95. Barani, M.; Mukhtar, M.; Rahdar, A.; Sargazi, G.; Thysiadou, A.; Kyzas, G.Z. Progress in the Application of Nanoparticles and Graphene as Drug Carriers and on the Diagnosis of Brain Infections. Molecules 2021, 26, 186. [Google Scholar] [CrossRef] [PubMed]
  96. Kenchegowda, M.; Rahamathulla, M.; Hani, U.; Begum, M.Y.; Guruswamy, S.; Osmani, R.A.M.; Gowrav, M.P.; Alshehri, S.; Ghoneim, M.M.; Alshlowi, A.; et al. Smart Nanocarriers as an Emerging Platform for Cancer Therapy: A Review. Molecules 2021, 27, 146. [Google Scholar] [CrossRef] [PubMed]
  97. Bilal, M.; Barani, M.; Sabir, F.; Rahdar, A.; Kyzas, G.Z. Nanomaterials for the treatment and diagnosis of Alzheimer’s disease: An overview. NanoImpact 2020, 20, 100251. [Google Scholar] [CrossRef]
  98. Moradi, S.; Mokhtari-Dizaji, M.; Ghassemi, F.; Sheibani, S.; Amoli, F.A. The effect of ultrasound hyperthermia with gold nanoparticles on retinoblastoma Y79 cells. Gold. Bull. 2020, 53, 111–120. [Google Scholar] [CrossRef]
  99. Wang, M.; Yang, Q.; Li, M.; Zou, H.; Wang, Z.; Ran, H.; Zheng, Y.; Jian, J.; Zhou, Y.; Luo, Y.; et al. Multifunctional Nanoparticles for Multimodal Imaging-Guided Low-Intensity Focused Ultrasound/Immunosynergistic Retinoblastoma Therapy. ACS Appl. Mater. Interfaces 2020, 12, 5642–5657. [Google Scholar] [CrossRef]
  100. Rajanahalli, P.; Stucke, C.J.; Hong, Y. The effects of silver nanoparticles on mouse embryonic stem cell self-renewal and proliferation. Toxicol. Rep. 2015, 2, 758–764. [Google Scholar] [CrossRef]
  101. Qu, W.; Meng, B.; Yu, Y.; Wang, S. EpCAM antibody-conjugated mesoporous silica nanoparticles to enhance the anticancer efficacy of carboplatin in retinoblastoma. Mater. Sci. Eng. C Mater. Biol. Appl. 2017, 76, 646–651. [Google Scholar] [CrossRef] [PubMed]
  102. Liu, Y.; Han, Y.; Chen, S.; Liu, J.; Wang, D.; Huang, Y. Liposome-based multifunctional nanoplatform as effective therapeutics for the treatment of retinoblastoma. Acta Pharm. Sin. B 2022, 12, 2731–2739. [Google Scholar] [CrossRef]
  103. Kartha, B.; Thanikachalam, K.; Vijayakumar, N.; Alharbi, N.S.; Kadaikunnan, S.; Khaled, J.M.; Gopinath, K.; Govindarajan, M. Synthesis and characterization of Ce-doped TiO2 nanoparticles and their enhanced anticancer activity in Y79 retinoblastoma cancer cells. Green Process. Synth. 2022, 11, 143–149. [Google Scholar] [CrossRef]
  104. Mitra, M.; Kandalam, M.; Rangasamy, J.; Shankar, B.; Maheswari, U.K.; Swaminathan, S.; Krishnakumar, S. Novel epithelial cell adhesion molecule antibody conjugated polyethyleneimine-capped gold nanoparticles for enhanced and targeted small interfering RNA delivery to retinoblastoma cells. Mol. Vis. 2013, 19, 1029–1038. [Google Scholar]
  105. Godse, R.; Rathod, M.; De, A.; Shinde, U. Intravitreal galactose conjugated polymeric nanoparticles of etoposide for retinoblastoma. J. Drug Deliv. Sci. Technol. 2021, 61, 102259. [Google Scholar] [CrossRef]
  106. Sims, L.B.; Tyo, K.M.; Stocke, S.; Mahmoud, M.Y.; Ramasubramanian, A.; Steinbach-Rankins, J.M. Surface-Modified Melphalan Nanoparticles for Intravitreal Chemotherapy of Retinoblastoma. Invest. Ophthalmol. Vis. Sci. 2019, 60, 1696–1705. [Google Scholar] [CrossRef] [PubMed]
  107. Silva-Abreu, M.; Calpena, A.C.; Espina, M.; Silva, A.M.; Gimeno, A.; Egea, M.A.; García, M.L. Optimization, Biopharmaceutical Profile and Therapeutic Efficacy of Pioglitazone-loaded PLGA-PEG Nanospheres as a Novel Strategy for Ocular Inflammatory Disorders. Pharm. Res. 2018, 35, 11. [Google Scholar] [CrossRef]
  108. Delrish, E.; Ghassemi, F.; Jabbarvand, M.; Lashay, A.; Atyabi, F.; Soleimani, M.; Dinarvand, R. Biodistribution of Cy5-labeled Thiolated and Methylated Chitosan-Carboxymethyl Dextran Nanoparticles in an Animal Model of Retinoblastoma. J. Ophthalmic Vis. Res. 2022, 17, 58–68. [Google Scholar] [CrossRef]
  109. Delrish, E.; Jabbarvand, M.; Ghassemi, F.; Amoli, F.A.; Atyabi, F.; Lashay, A.; Soleimani, M.; Aghajanpour, L.; Dinarvand, R. Efficacy of topotecan nanoparticles for intravitreal chemotherapy of retinoblastoma. Exp. Eye Res. 2021, 204, 108423. [Google Scholar] [CrossRef]
  110. Passos Gibson, V.; Derbali, R.M.; Phan, H.T.; Tahiri, H.; Allen, C.; Hardy, P.; Chain, J.L. Survivin silencing improved the cytotoxicity of carboplatin and melphalan in Y79 and primary retinoblastoma cells. Int. J. Pharm. 2020, 589, 119824. [Google Scholar] [CrossRef]
  111. Tabatabaei, S.N.; Derbali, R.M.; Yang, C.; Superstein, R.; Hamel, P.; Chain, J.L.; Hardy, P. Co-delivery of miR-181a and melphalan by lipid nanoparticles for treatment of seeded retinoblastoma. J. Control. Release 2019, 298, 177–185. [Google Scholar] [CrossRef] [PubMed]
  112. Narayana, R.V.L.; Jana, P.; Tomar, N.; Prabhu, V.; Nair, R.M.; Manukonda, R.; Kaliki, S.; Coupland, S.E.; Alexander, J.; Kalirai, H.; et al. Carboplatin- and Etoposide-Loaded Lactoferrin Protein Nanoparticles for Targeting Cancer Stem Cells in Retinoblastoma In Vitro. Invest. Ophthalmol. Vis. Sci. 2021, 62, 13. [Google Scholar] [CrossRef]
  113. Ahmed, F.; Ali, M.J.; Kondapi, A.K. Carboplatin loaded protein nanoparticles exhibit improve anti-proliferative activity in retinoblastoma cells. Int. J. Biol. Macromol. 2014, 70, 572–582. [Google Scholar] [CrossRef] [PubMed]
  114. Shome, D.; Kalita, D.; Jain, V.; Sarin, R.; Maru, G.B.; Bellare, J.R. Carboplatin loaded polymethylmethacrylate nano-particles in an adjunctive role in retinoblastoma: An animal trial. Indian J. Ophthalmol. 2014, 62, 585–589. [Google Scholar] [CrossRef] [PubMed]
  115. Alsaab, H.; Alzhrani, R.M.; Kesharwani, P.; Sau, S.; Boddu, S.H.; Iyer, A.K. Folate Decorated Nanomicelles Loaded with a Potent Curcumin Analogue for Targeting Retinoblastoma. Pharmaceutics 2017, 9, 15. [Google Scholar] [CrossRef] [PubMed]
  116. Mitra, M.; Misra, R.; Harilal, A.; Sahoo, S.K.; Krishnakumar, S. Enhanced in Vitro Antiproliferative Effects of EpCAM Antibody-Functionalized Paclitaxel-Loaded PLGA Nanoparticles in Retinoblastoma Cells. Mol. Vis. 2011, 17, 2724. [Google Scholar] [PubMed]
  117. N’Diaye, M.; Vergnaud-Gauduchon, J.; Nicolas, V.; Faure, V.; Denis, S.; Abreu, S.; Chaminade, P.; Rosilio, V. Hybrid Lipid Polymer Nanoparticles for Combined Chemo- And Photodynamic Therapy. Mol. Pharm. 2019, 16, 4045–4058. [Google Scholar] [CrossRef]
  118. Gallud, A.; Warther, D.; Maynadier, M.; Sefta, M.; Poyer, F.; Thomas, C.D.; Rouxel, C.; Mongin, O.; Blanchard-Desce, M.; Morère, A.; et al. Identification of MRC2 and CD209 Receptors as Targets for Photodynamic Therapy of Retinoblastoma Using Mesoporous Silica Nanoparticles. RSC Adv. 2015, 5, 75167–75172. [Google Scholar] [CrossRef]
Figure 1. Schematic representation of nano-therapeutics for pediatric brain tumors.
Figure 1. Schematic representation of nano-therapeutics for pediatric brain tumors.
Brainsci 14 00185 g001
Figure 2. The role of nanoparticles in overcoming the BBB for efficient delivery of therapeutic moieties to treat AD. (A) Image of the human brain. (B) Components of the BBB. (C) Functionalized nanoparticles (NPs) for imaging and targeted drug delivery to the AD brain. (D) Different pathways of transport (a–e) across BBB are utilized by functionalized NPs. (a) Transport of NPs through cellular transport proteins; (b) transport of NPs through tight junctions; (c) transport of NPs via receptor-mediated transcytosis; (d) transport of NPs via transcellular pathway following diffusion, specifically adopted by gold NPs; and (e) transport of cationic NPs and liposomes via adsorption-mediated transcytosis. (E) Effect of functionalized NPs in treating AD via the degradation of tau aggregates and efflux of Aβ fibrils after getting solubilized by the NPs. AD: Alzheimer’s disease; NPs: nanoparticles; BBB: blood–brain barrier. Adapted with permission from [44].
Figure 2. The role of nanoparticles in overcoming the BBB for efficient delivery of therapeutic moieties to treat AD. (A) Image of the human brain. (B) Components of the BBB. (C) Functionalized nanoparticles (NPs) for imaging and targeted drug delivery to the AD brain. (D) Different pathways of transport (a–e) across BBB are utilized by functionalized NPs. (a) Transport of NPs through cellular transport proteins; (b) transport of NPs through tight junctions; (c) transport of NPs via receptor-mediated transcytosis; (d) transport of NPs via transcellular pathway following diffusion, specifically adopted by gold NPs; and (e) transport of cationic NPs and liposomes via adsorption-mediated transcytosis. (E) Effect of functionalized NPs in treating AD via the degradation of tau aggregates and efflux of Aβ fibrils after getting solubilized by the NPs. AD: Alzheimer’s disease; NPs: nanoparticles; BBB: blood–brain barrier. Adapted with permission from [44].
Brainsci 14 00185 g002
Figure 3. Observation of coumarin-6-labeled NP, NP co-administered with iRGD, MT1-NP, and MT1-NP co-administered with iRGD distribution in the brains of nude mice with intracranial C6 glioma, 3 h after intravenous administration. Analysis conducted on frozen sections using a confocal microscope revealed blood vessels marked with anti-CD31 (red), nuclei stained with DAPI (blue), and NPs depicted in green. Scale bars indicate 50 mm. Adapted with permission from reference [51].
Figure 3. Observation of coumarin-6-labeled NP, NP co-administered with iRGD, MT1-NP, and MT1-NP co-administered with iRGD distribution in the brains of nude mice with intracranial C6 glioma, 3 h after intravenous administration. Analysis conducted on frozen sections using a confocal microscope revealed blood vessels marked with anti-CD31 (red), nuclei stained with DAPI (blue), and NPs depicted in green. Scale bars indicate 50 mm. Adapted with permission from reference [51].
Brainsci 14 00185 g003
Figure 4. Assessing the fluorescence and photoacoustic imaging capabilities of ILP. (A) In vivo, observe the fluorescence and photoacoustic images of ICG and ILP at various time intervals. (B,C) Analyze the fluorescent intensity (B) and photoacoustic intensity (C) of tumor tissue quantitatively. (D) Examine ex vivo fluorescence images of ICG and ILP at 6 h. (E) Quantify the fluorescent intensity of different tissues at the 6 h mark. Herein p < 0.05 is flagged with one star (*) and p < 0.01 is flagged with two stars (**). Adapted with permission from reference [102].
Figure 4. Assessing the fluorescence and photoacoustic imaging capabilities of ILP. (A) In vivo, observe the fluorescence and photoacoustic images of ICG and ILP at various time intervals. (B,C) Analyze the fluorescent intensity (B) and photoacoustic intensity (C) of tumor tissue quantitatively. (D) Examine ex vivo fluorescence images of ICG and ILP at 6 h. (E) Quantify the fluorescent intensity of different tissues at the 6 h mark. Herein p < 0.05 is flagged with one star (*) and p < 0.01 is flagged with two stars (**). Adapted with permission from reference [102].
Brainsci 14 00185 g004
Table 1. Applications of Nanoparticles for medulloblastoma and glioma therapy.
Table 1. Applications of Nanoparticles for medulloblastoma and glioma therapy.
Carriers #Targeting Ligand #Targeting Receptor/Area #Therapeutic Molecule #Cell Line and/or Animal Model Used #Outcome/Key Findings #Ref.
Poly (beta-amino ester) (PBAE) nanoparticles--Herpes simplex virus type I thymidine kinaseAT/RT implanted miceGreater median overall survival in mice implanted with AT/RT[45]
High-density lipoprotein-mimetic nanoparticlesapolipoprotein A1
anti-CD15
SR-B1 CD15 antigenLDE225SHH MB cellsSR-B1-mediated intracellular cholesterol depletion in SHH MB cells.[46]
mPEG-b-PCC-g-DC copolymer-based NPsApoE-targeting peptide COG-133ApoE receptorMDB5
SF2523
SHH-MB tumor-bearing NSG miceReduction in tumors in orthotopic SHH-MB tumor-bearing NSG mice[48]
Liposomes--TemozolomideMicePreferential accumulation in the brain[49]
PAMAM-PEG- nanoparticlesTransferrinTransferrin-1 receptorsTemozolomidenude mouse intracranial xenograft models.Anticancer activity against O6-methylguanine-DNA- methyltransferase gene promoter methylation.[50]
PEG-PLA nanoparticles MT1-AF7p peptideMembrane type-1 matrix metalloproteinase paclitaxel C6 glioma-bearing nude mice Enhanced survival time in intracranial C6 glioma-bearing nude mice [51]
liposome LAT 1LAT-1 receptorAmphi-DOPAglioma-bearing C57BL/6J miceOverall survivability increased by 60% in glioma-bearing C57BL/6J mice[52]
RBC coated nanoparticle CDX peptidenAChRsDoxorubicinglioma bearing nude miceHigh brain targeting, superior therapeutic activity with less toxicity[53]
poly (L-γ-glutamyl-glutamine)- nanoconjugates aptamer AS1411NucleolinPaclitaxel U87 MG cells and intracranial glioblastoma-bearing nude miceHigher anti-glioma effect with enhanced median survival time[54]
Liposome T7 DA7RTransferrin receptors, VEGFR 2 receptorsDoxorubicin and vincristineHUVEC cells, C6 Cells, glioma-bearing mice High anti-glioma effect in in vivo studies[55]
Chitosan-coated PLGA nanoparticles-BrainCarmustineU87 MG cell line
Albino Wistar rats
Enhanced cytotoxicity in cell lines and AUC in brain[61]
PG-SPIONsFolic acidFolate receptorsLomustineU87 MG cell lineEnhanced cellular uptake[62]
CGT nanoparticles CGTintegrins avβ3 and avβ5 -Rat glioblastoma modelUTMD with CGT therapy improved the CGT delivery, prolonged tumor retention, apoptosis, and median survival period[63]
Human serum albumin nanoparticlesFolic acidFolate receptorsErlotinibU87MG and C6 cells
rat glioblastoma model
Improved apoptosis and tumor reduction compared to pure drug[64]
Liposomes--Doxorubicin and erlotinibU87 MG cell linesImproved apoptosis[65]
poly (butyl cyanoacrylate) (PBCA) NPsmAb-Carboplatin.Rat glioblastoma modelLonger survival time[66]
# ApoE—Apolipoprotein E, AT—Atypical Teratoid, CGT—Cilengitide, DOPA—Dihydroxyphenylalanine, HUVEC—Human Umbilical Vein Endothelial Cells, LAT 1—Large Amino Acid Transporter1, mAb—Mono clonal antibody, nAChRs—Nicotinic Acetylcholine Receptors, NSG—NOD Scid Gamma, PAMAM—Polyamidoamine, PBAE—Poly (Beta-Amino Ester), PEG-PLA—Poly (Ethylene Glycol)—Poly (Lactic Acid), PG-SPIONS—polyglycerol coated superparamagnetic iron oxide nanoparticles, RT—Rhabdoid Tumors, SHH-MB—Sonic Hedgehog Medulloblastoma, SR-B1—Scavenger Receptor Class B Type 1, UTMD—ultrasound-targeted microbubble destruction, VEGFR—Vascular Endothelial Growth Factor Receptor.
Table 2. Applications of nanoparticles for neuroblastoma therapy.
Table 2. Applications of nanoparticles for neuroblastoma therapy.
Carriers/Moieties #Therapeutic Molecule #In Vitro/In Vivo Models #Outcome/Key Findings #Ref.
Silica-PAMAM dendrimer HybridAnthocyaninsNeuro-2A brain neuroblastoma from mouse and Vero (African green monkey kidney) normal cell linesThe Hybrid nanoparticles (134.8 nm) with +19.78 mV zeta potential showed effective cytotoxicity against Neuroblastoma (Neuro 2A) cells, with 87.9% inhibition due to anthocyanin release. This system appears to be primarily therapeutic in its current application. The placebo nanoparticles were non-toxic to the cells.[74]
LiposomesPyrazolo[3,4-d]pyrimidinesSprague Dawley ratsLiposomal encapsulation effectively overcame the poor water solubility of pyrazolo[3,4-d]pyrimidines; this study focuses on the therapeutic approach and makes them more suitable for clinical drug development.[75]
PLA- and PLGA-based nanoparticlesDoxorubicinNeuroblastoma cell line UKF-NB-3These kinds of nanoparticles can enhance in vivo drug activity through the EPR effect and overcome transporter-mediated drug resistance. The primary focus of the study is on delivering doxorubicin as a therapeutic agent against neuroblastoma cells.[76]
PLGA (Poly(lactic-co-glycolic acid) nanoparticlesPaclitaxel (taxol)Human neuroblastoma cells (SH-SY5Y)Paclitaxel-loaded PLGA nanoparticles exhibited cytotoxicity with cell viability below 50% at concentrations > 10 nM and induced genotoxic effects, suggesting their potential as a biocompatible carrier for neuroblastoma treatment and the developed system was used for a therapeutic approach.[77]
Cyclodextrin-Fibrin gels (FBGs)Doxorubicin (Dox)Mouse orthotopic NB model (SHSY5YLuc+ cells implanted into the left adrenal glandIncrease in the therapeutic index of Dox when locally administered via FBGs loaded with oCD-NH2/Dox for neuroblastoma treatment. Overall, the research presented in the article focuses on improving the therapeutic efficacy of Dox delivery for neuroblastoma treatment, with possibilities for exploring theranostic applications in the future.[78]
Biomimetic Core-Shell NNsTherapeutic miRNAHuman neuroblastoma CHLA-255 cells and CHLA-255-luc tumor-bearing nonobese diabetic/severe combined immunodeficient (NOD/SCID) miceNN/NKEXO cocktail for targeted neuroblastoma therapy, efficient miRNA delivery, dual tumor growth inhibition, and potential clinical application. Prepared systems are primarily focused on a therapeutic approach.[79]
Soluplus or Chitosan NanoparticlesPosidonia oceanica (POE)SH-SY5Y human neuroblastoma cell lineNPs improved the aqueous solubility and stability of POE and enhanced its inhibitory effect on cancer cell migration, likely due to efficient encapsulation. Overall, the research presented in the article focuses on improving the therapeutic application of the nano-formulations.[80]
Alginate-TiO2 TMZ NanoparticlesTemozolomide (TMZ)Human neuroblastoma cells SH-SY5YThe developed system seems to focus primarily on a therapeutic approach, as these nanoparticles exhibit higher cytotoxicity against neuroblastoma cells, potentially impacting neuroblastoma treatment.[81]
Solid lipid (Precirol®) and αv integrins (ligand)Etoposide and CilengitideHR-NB cell lines and MYCN-amplified cell linesCombination therapy with cilengitide enhanced efficacy against high-risk neuroblastoma cells. This research offers theragnostic potential by targeting ECM-tumor cell interactions, inhibiting VN-integrin binding, modulating ECM stiffness, and employing nanoencapsulated chemotherapeutic agents to enhance the therapeutic index and overall effectiveness in high-risk neuroblastoma treatment.[82]
Bacterial Membrane-coated Nanoparticle (BNP)PC7A/CpG polyplex core with bacterial membraneB78 melanoma tumors engrafted in syngeneic miceBNP enhances immune recognition of tumor neoantigens post-radiation, improving dendritic cell uptake and cross-presentation, resulting in robust antitumor T-cell responses in mice with melanoma or neuroblastoma. The developed system seems to focus primarily on a therapeutic approach; therefore, the current application leans toward immunotherapy,[83]
Synthetic High-Density Lipoprotein (HDL) Nanoparticles4,19,27-tri acetyl withanolide AHuman NB cell lines SH-EP, SH-SY5Y, IMR32 and SK-N-As and tumor-bearing miceTreatment reduced sphere formation, invasion, migration, and cancer stem cell markers in neuroblastoma cells. However, the targeting of SR-B1 and its potential for influencing CSC functions also suggest the potential for theranostic applications, where the nanoparticles could be used for both diagnosis and treatment.[84]
Core-Shell MOF of ZincTitanocene Dichloride (TC) loaded Lactoferrin (Lf)Neuroblastoma- IMR-32 cells and Wistar ratsZIF-8 framework loaded with Lf-TC and 5-Fluorouracil exhibited potential for Neuroblastoma therapy, confirmed through in vitro cell studies and in vivo safety assessments in Wistar rats. The Lf-TC and 5FU-loaded ZIF-8 framework serves as nanoplatforms for tumor phototherapy, with the potential for transformation into a theranostic platform through additional imaging moiety modifications.[85]
Graphene Quantum Dots (GQDs)Anti-GD2 AntibodyNIH3T3 mouse fibroblast cell line and BE(2)-M17 human neuroblastoma cell line and nude miceThis study focuses on the theranostics potential of anti-GD2/GQDs and demonstrates the potential use of Anti-GD2/GQDs for targeting and imaging of neuroblastomas in vivo.[86]
Nanocarriers coated-cationic liposomes functionalized with antibodies against GD2 receptormiR-34a and let-7bNB tumor cells, orthotopic xenografts, pseudometastatic models, athymic micePromising therapeutic efficacy of miR-34a and let-7b combined replacement, Support for clinical application as adjuvant therapy for high-risk NB patients.[87]
# NKEXOs: Natural killer cell-derived exosomes, NNs: core–shell nanoparticles, TiO2: Titanium dioxide, NB: Neuroblastoma, ECM: Extracellular matrix, CSC: Cancer Stem Cell, ZIF-8: Zeolitic Imidazolate Framework.
Table 3. Applications of nanoparticles for retinoblastoma therapy.
Table 3. Applications of nanoparticles for retinoblastoma therapy.
Carriers/Moieties #Therapeutic Molecule #In Vitro/In Vivo Models #Outcome/Key Findings #Ref.
AuNP-PEI-EpCAM Antibody (EpAb)EpCAM-specific siRNAY79 retinoblastoma cellsNovel nanocarrier successfully delivered EpCAM-specific siRNA to retinoblastoma (RB) cells, leading to significant gene knockdown. The nanoparticles were well-tolerated by cells, and their conjugation with the EpCAM antibody enhanced internalization and therapeutic efficacy for RB. Gold nanoparticles also hold the potential for imaging in diagnosis.[104]
Galactose-Chitosan Anchored Etoposide PLGA NPs (GC-ENP)Etoposide (ETP)Y-79 retinoblastoma cellsGC-ENP, with high entrapment efficiency and galactose targeting, demonstrates increased uptake in retinoblastoma cells (Y-79) and enhanced cytotoxicity, making it a promising drug delivery system for retinoblastoma treatment and enhancing the therapeutic application of the developed system.[105]
PLGA NanoparticlesMelphalanY79 cellsSurface modification improves efficacy in retinoblastoma cells, particularly with MPG-NPs. Prepared systems are primarily focused on a therapeutic approach, and this system enhances cell association, but some NPs remain on the cell surface rather than internalizing.[106]
Nanospheres (NSs) of PGZPioglitazone (PGZ)Y-79 cell line and male pigsThe Polymeric nanoparticles effectively encapsulated PGZ, showing optimal characteristics with sustained drug release, good ocular tolerance, and significant in vivo anti-inflammatory potential, offering a promising approach for ocular inflammation treatment and suggesting a purely therapeutic approach.[107]
CMD-TCs-NPs Y79 retinoblastoma cells and Wistar albino ratsCMD-TCs-NPs show smaller size, positive zeta potential, and higher affinity for retinoblastoma tumors in rat eyes when administered intravitreally, while CMD-TMC-NPs remained in the vitreous and did not reach the retina. These findings suggest CMD-TCs-NPs’ potential for more effective drug delivery in retinoblastoma treatment.[108]
Thiolated Chitosan Nanoparticles (TPH-TCs-NPs)Topotecan (TPH)Human retinoblastoma cells (Y79), xenograft-rat-model of retinoblastomaTPH-TCs-NPs enhanced drug loading, improved control over drug release, and increased treatment efficacy for retinoblastoma. Thiolated chitosan demonstrates improved interaction with cell membranes, leading to higher cellular uptake of the drug. Therefore, this study primarily focuses on the therapeutic approach.[109]
siRNA-loaded switchable LNPSurvivin siRNAY79 retinoblastoma cells and primary human RB cells.Sequential siRNA survivin followed by chemotherapy sensitizes cancer cells to carboplatin and melphalan, showing promise in treating retinoblastoma (RB) without affecting healthy cells. The study suggests careful drug screening to find synergy with survivin for future in vivo testing.[110]
Lipid Nanoparticles (LNP)Melphalan and miR-181aY79 retinoblastoma cells and Sprague Dawley ratsCo-delivery of melphalan and miR-181a using 171 nm switchable LNP with high encapsulation efficiencies enhanced therapeutic efficiency, reducing the expression of anti-proliferative and anti-apoptotic genes while increasing pro-apoptotic gene expression. [111]
Lactoferrin nanoparticles (Lf-Nps)Carboplatin (CPT) and Etoposide (ETP)Retinoblastoma (Rb) Y79 cellsThe Nanoformulations of Lf-CPT and Lf-ETP enhance drug uptake, intracellular retention, and cytotoxicity, particularly in Rb Y79 CSCs, offering the potential for improved targeted therapy and therapeutic efficacy and better clinical outcomes by overcoming chemoresistance in cancer stem cells (CSCs).[112]
Apo-nano-carbo and Lacto-nano-carbo nanoparticlesCarboplatinHuman retinoblastoma cell line Y79These nanoparticles demonstrated pH-dependent drug release and receptor-mediated endocytosis for targeted delivery, resulting in greater intracellular uptake and anti-proliferative activity (IC50 = 4.31 μg ml−1 and 4.16 μg ml−1, respectively) compared to soluble carboplatin (IC50 = 13.498 μg ml−1).[113]
Polymethylmethacrylate nanoparticlesCarboplatinSprague Dawley ratsIntra-vitreal carboplatin concentrations were significantly higher with novel carboplatin-loaded polymethylmethacrylate nanoparticles (NPC) compared to the commercially available carboplatin (CAC), indicating enhanced trans-scleral permeability for potential use in treating advanced retinoblastoma. Therefore, this study primarily focuses on the therapeutic approach.[114]
Folic Acid-Conjugated Polymeric MicellesCurcumin-Difluorinated (CDF)Retinoblastoma cell lines (Y-79 and WERI-RB1)The Folic acid-conjugated micelles loaded with CDF increased CDF solubility and showed significant anticancer activity on retinoblastoma cell lines (Y-79 and WERI-RB). This formulation holds promise as an alternative approach to retinoblastoma therapies; therefore, the study focuses on the therapeutic potential of the developed system.[115]
EpCAM antibody-functionalized PLGA NPs.PaclitaxelY79 retinoblastoma cellsEpCAM antibody-functionalized biodegradable NPs show potential for tumor-selective drug delivery and overcoming drug resistance in retinoblastoma treatment. Therefore, this study primarily focuses on the therapeutic approach.[116]
Hybrid Lipid Polymer NanoparticlesBeta-lapachone (β-Lap)Retinoblastoma cellsThis study focuses on a combined chemo- and photodynamic therapy (PDT) approach, aiming to synergistically treat retinoblastoma with both β-Lap and m-THPC encapsulated in LNPs.[117]
Mesoporous silica nanoparticlesanti-MRC2 and/or anti-CD209Human retinoblastoma cancer cells (Y-79 and WERI-Rb1)Identifies elevated expression of two receptors, MRC2 and CD209, in retinoblastoma, leading to the creation of mesoporous silica nanoparticles (MSN) equipped with anti-MRC2 and/or anti-CD209 antibodies for targeted PDT and imaging.[118]
# AuNP-PEI-EpCAM Antibody: Gold nanoparticles-polyethyleneimine-Epithelial cell adhesion molecule monoclonal antibody conjugated, CMD-TCs-NPs thiolated and methylated chitosan-carboxymethyl dextran nanoparticles, siLNP: siRNA-loaded switchable lipid nanoparticles, Apo-nano-carbo: carboplatin loaded apotranferrin and Lacto-nano-carbo: lactoferrin loaded nanoparticles, PLGA NPs: Poly(lactic-co-glycolic acid) nanoparticles, PDT: Photodynamic therapy.
Table 4. Clinical trials using nano-therapeutics in pediatric brain tumors.
Table 4. Clinical trials using nano-therapeutics in pediatric brain tumors.
Carriers/Nanoparticles #Condition #Therapeutic Agent #Phase #Status #NCT Code
LiposomesGlioblastomaC225-ILs-doxPhase 1CompletedNCT03603379
Gold NanoparticleRecurrent GlioblastomaNU-0129 IVEarly Phase 1CompletedNCT03020017
Ultra-small iron oxide particleBrain NeoplasmsCombidex as MRI contrast agentPhase 2TerminatedNCT00659334
Small iron particlesChildhood Brain NeoplasmDSC-MRI with ferumoxytolEarly Phase 1CompletedNCT00978562
Nanoparticle Formulation MTX110Diffuse Intrinsic Pontine GliomaPanobinostatPhase 1
Phase 2
CompletedNCT03566199
LiposomesBrain tumorDoxorubicinPhase 1CompletedNCT00019630
LiposomesBrain and Central Nervous System TumorsCytarabinePhase 1Unknown statusNCT00003073
MTX110 and gadoliniumDiffuse Intrinsic Pontine GliomaInfusatePhase 1CompletedNCT04264143
LiposomesNeuroblastomaDoxorubicinPhase 1TerminatedNCT02536183
LiposomeNeuroblastomaDoxorubicinPhase 1WithdrawnNCT02557854
LiposomeNeuroblastomaIrinotecan Sucrosofate Phase 1RecruitingNCT02013336
# C225-ILs-dox: Doxorubicin-loaded Anti-EGFR-immunoliposomes.
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

Ravi Kiran, A.V.V.V.; Kumari, G.K.; Krishnamurthy, P.T.; Johnson, A.P.; Kenchegowda, M.; Osmani, R.A.M.; Abu Lila, A.S.; Moin, A.; Gangadharappa, H.V.; Rizvi, S.M.D. An Update on Emergent Nano-Therapeutic Strategies against Pediatric Brain Tumors. Brain Sci. 2024, 14, 185. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci14020185

AMA Style

Ravi Kiran AVVV, Kumari GK, Krishnamurthy PT, Johnson AP, Kenchegowda M, Osmani RAM, Abu Lila AS, Moin A, Gangadharappa HV, Rizvi SMD. An Update on Emergent Nano-Therapeutic Strategies against Pediatric Brain Tumors. Brain Sciences. 2024; 14(2):185. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci14020185

Chicago/Turabian Style

Ravi Kiran, Ammu V. V. V., G. Kusuma Kumari, Praveen T. Krishnamurthy, Asha P. Johnson, Madhuchandra Kenchegowda, Riyaz Ali M. Osmani, Amr Selim Abu Lila, Afrasim Moin, H. V. Gangadharappa, and Syed Mohd Danish Rizvi. 2024. "An Update on Emergent Nano-Therapeutic Strategies against Pediatric Brain Tumors" Brain Sciences 14, no. 2: 185. https://0-doi-org.brum.beds.ac.uk/10.3390/brainsci14020185

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