Next Article in Journal
Thiophene Stability in Photodynamic Therapy: A Mathematical Model Approach
Next Article in Special Issue
Pericytes Are Immunoregulatory Cells in Glioma Genesis and Progression
Previous Article in Journal
Biology and Therapeutic Properties of Mesenchymal Stem Cells in Leukemia
Previous Article in Special Issue
Targeting Innate Immunity in Glioma Therapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glioblastoma Therapy: Past, Present and Future

by
Elena Obrador
1,2,*,
Paz Moreno-Murciano
1,
María Oriol-Caballo
1,2,
Rafael López-Blanch
1,2,
Begoña Pineda
2,
Julia Lara Gutiérrez-Arroyo
3,
Alba Loras
3,
Luis G. Gonzalez-Bonet
4,
Conrado Martinez-Cadenas
3,
José M. Estrela
1,2,5 and
María Ángeles Marqués-Torrejón
3,*
1
Scientia BioTech S.L., 46002 Valencia, Spain
2
Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain
3
Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain
4
Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain
5
Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2024, 25(5), 2529; https://doi.org/10.3390/ijms25052529
Submission received: 23 December 2023 / Revised: 10 February 2024 / Accepted: 16 February 2024 / Published: 21 February 2024
(This article belongs to the Special Issue Glioblastoma: State of the Art and Future Trends)

Abstract

:
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood–brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.

1. Introduction

1.1. The Nature and Prognosis of Glioblastoma

Glioblastoma (GB), categorized as a grade IV astrocytoma, is the most prevalent, aggressive, and lethal primary brain tumor in adults. In 2021, the World Health Organization (WHO) introduced significant changes in the criteria for the diagnosis of gliomas, focusing on the importance of genetic and molecular alterations. According to these new criteria (Figure 1), GB should be diagnosed in adults as an isocitrate dehydrogenase wild-type (IDHwt) diffuse astrocytic glioma if there is microvascular proliferation or necrosis (the conventional criteria), and/or at least one of the following three criteria: concurrent gain of whole chromosome 7 and loss of whole chromosome 10 (+7/−10), telomerase reverse transcriptase (TERT) promoter mutations, and epidermal growth factor receptor (EGFR) amplification [1,2]. The primary driver behind the change in diagnosis criteria is the IDH mutation status, which results in the following modifications: restricting the diagnosis of GB to tumors that do not have IDH mutations (IDHwt); reclassifying tumors previously identified as IDH-mutated GBs as astrocytomas with IDH mutations (grade IV); and establishing the presence of IDH mutations as a requirement for classifying tumors as astrocytomas or oligodendrogliomas Consequently, due to its more favorable prognosis, the previously designated IDH-mutant GB is now categorized within the astrocytomas group, which covers grades II–IV, thus eliminating the term IDH-mutant GB [3]. Moreover, in IDHwt diffuse astrocytomas occurring in younger people, diagnostic consideration should be given to the different types of diffuse pediatric-type gliomas [1]. Gliosarcoma, epithelioid cell GB and giant cell GB are still registered subtypes of GBs, and the term “glioblastoma multiforme” should not be used [4,5]. New clinical trials will need to be designed with these new distinctions in mind [6].
GB typically appears in the cerebral hemispheres, with 95% of these tumors arising in the supratentorial region, especially in the frontal and temporal globes. It infiltrates inside the brain parenchyma and usually does not spread to other parts of the body [7,8,9]. Histologically, GBs are characterized by prominent cellular and nuclear atypia, increased mitotic activity, areas of necrosis, and microvascular proliferation. At least one of these two features must be present for a histologic diagnosis of GB [5]. GB causes death in less than 6 months if untreated [10]. Despite advances in neurosurgery, chemotherapy and radiotherapy, GB remains one of the most treatment-resistant malignancies and its relapse is, in practice, inevitable [7,11,12]. Recurrence often implies a more aggressive form and a median survival of less than 18 months in treated patients [13,14]. Survival beyond 5 years is observed in less than 5.8% of patients [7]. Patients with recurrent GB (rGB) show an approx. 6-month progression-free survival (PFS6) in only 15% of the cases, and overall survival (OS) ranging between 24 and 40 weeks. Survival rate decreases with age [11,15].
As suggested by the moniker “multiforme”, GB has a widespread tumoral heterogeneity and plasticity at the cytopathological, transcriptional, and genomic levels [16,17,18,19,20,21]. Moreover, its highly infiltrative nature and the protection by the blood–brain barrier (BBB) have posed significant treatment challenges [9,22,23]. Glioma stem cells (GSCs) are a small subpopulation of cells within the GB, with genomic instability, self-renewal and tumor-initiating capacity, and the ability to differentiate into different GB subpopulations, being responsible for tumor heterogeneity [24,25,26,27]. Moreover, GSCs are resistant to apoptosis [28,29,30,31], can modulate the components of the tumor microenvironment (TME), are involved in angiogenesis activation and immunosuppression and drive radio/chemoresistance [22,32,33]. The inability of current therapies to eliminate specific GSC subpopulations has been considered a major factor contributing to the inevitable recurrence after treatment [33,34].
Verhaak et al. proposed a four-subtype classification of GB (classical, mesenchymal, proneural, and neural) based on the analysis of mutational changes in 601 genes in the context of The Cancer Genome Atlas (TCGA) [35]. Verhaak’s latest update removed the neural subtype attributing its origin to a peripheral contamination of the tumor samples [20]. The proneural subtype is associated with a more favorable outcome with respect to the mesenchymal, but this difference is relative to the more favorable outcome of IDH-mutant GBs which were consistently classified as proneural GBs [20,36]. Moreover, mesenchymal GSCs are enriched with genes associated with angiogenesis, inflammation, and cell migration/invasion. They tend to develop immunosuppression and exhibit increased radio/chemoresistance, all of which are features linked to a worse prognosis [36,37,38,39]. In any of the cases, the survival difference is minimal because both subtypes can coexist in the same tumor, and dynamic transitions from a proneural to a mesenchymal phenotype can be induced by TNF-α, temozolomide (TMZ), or radiation through an NF-κB-dependent mechanism [20,36,40,41]. Meanwhile, multiplatform analyses of the genetic, epigenetic, and transcriptional profiles have proven useful in refining the classification of gliomas and predicting patient outcomes [42,43,44,45].

1.2. Incidence and Risk Factors

GB is the most common (50.1%) among all malignant brain tumors [15]. The annual incidence is low (≈3.19 per 100,000 people in developed countries) but seems to be increasing in some countries owing to aging populations and improvements in diagnosis, among other factors [46]. The median age of diagnosis is approx. 64 and the incidence increases with age reaching its maximum value (15 per 100,000 people) between 75 and 84 years [11,15,47]. It is extremely rare in a pediatric population (0.15 per 100,000), which usually shows longer survivals. The occurrence of GB is 1.6 times more common in males than females and in Caucasians relative to other ethnicities [15,48].
Beyond rare cases of genetic susceptibility and high-dose radiation exposure, there are no known GB risk factors. An increased risk is seen in some specific genetic diseases, such as hereditary retinoblastoma or Cowden, Turcot, Lynch, Li-Fraumeni and Maffucci syndromes. However, less than 1% of GB patients have a known hereditary disease. Radiation-induced GB can be diagnosed several years after radiation therapy for another tumor or condition in children [49], but no increased risk was observed in adults exposed to IR [50]. Patients diagnosed with previous non-neurological cancers may have an overall elevated incidence of GB compared to the general population [51].

1.3. Criteria to Evaluate Treatment Response and Progression

The MacDonald criteria [52] have traditionally been used to determine treatment response and progression by assessing contrast-enhancing tumor size [by computed tomography (CT) or magnetic resonance imaging (MRI)] along with clinical evaluation and corticosteroid dosage. These criteria categorized the response into four groups: complete response (CR), partial response (PR), stable disease (SD), and progressive disease (PD). Nevertheless, these criteria have several limitations. One is the temporary increase in tumor enhancement (known as pseudo-progression), which occurs in 20–30% of the patients treated with chemo/radiotherapy and challenges differentiation with a real tumor progression. Another limitation is the high radiographic response rates seen with anti-angiogenic agents and other treatments [53]. To address these issues in 2010, the Response Assessment in Neuro-Oncology (RANO) criteria were developed to address these limitations. Although the RANO criteria improved therapy evaluation in high-grade glioma, the assessment of treatment-related side effects can hinder accurate response evaluation. The appearance of new lesions is considered a criterion for disease progression according to both RANO and MacDonald criteria. However, neuro-oncology patients receiving immunotherapies may experience the transient appearance of new enhancing lesions, either locally or in distant sites. In such cases, it is advisable to evaluate imaging findings within 6 months of starting immunotherapy, including the development of new lesions or radiographic progression, as long as there is no significant clinical deterioration [54].
To tackle the challenges in assessing immunotherapy response for neuro-oncology, the immunotherapy RANO (iRANO) criteria were introduced. The iRANO criteria combine the response assessment framework of RANO with guidelines for confirming disease progression, as originally proposed by the Immunotherapy Response Criteria in Solid Tumors to assist in clinical decision-making. The aim is to minimize premature discontinuation of potentially beneficial therapies while ensuring patient safety [54]. However, in most recurrence cases, there is a mixture of tumor cells and tissue affected by radiation injury. Radiologists strive to identify the predominant component of the lesion to determine prognostic factors and categorize the findings according to the RANO criteria, thus providing the most appropriate treatment for the patient. To overcome the aforementioned limitations in the follow-up, incorporating changes measured by advanced MRI and positron emission tomography (PET) imaging, which may precede anatomical changes in tumor volume, shows promise [55,56]. PET may also help to differentiate actual progression from pseudo progression [57]. Additionally, 18F-FMISO-PET can localize regions of hypoxia that are thought to drive radio/chemoresistance in GBs and promote immune suppression [58].

2. Lessons Learned in the Pathophysiology of Glioblastoma

2.1. Glioma Stem Cells and Tumor Microenvironment

The two prevailing hypotheses for the origin of GB are the GSC and the astrocyte de-differentiation theories [25,26,37]. Neural stem cells (NSCs), as unique stem cell type in the brain, have the ability to self-renew and can differentiate into neurons, astrocytes, and oligodendrocytes (Figure 2) [25,59]. NSCs are most active during development, but small populations remain functional in specific stem-cell niches in the adult brain. Compelling evidence suggests that GSCs may arise from NSCs located in the adult subventricular zone (SVZ) [60,61,62,63,64,65], and a recent article provided molecular genetic confirmation of this hypothesis in a preclinical model [60]. GSCs express the mutated genes TERT, PTEN, EGFR, TP53, and PDGF present in NSCs. In addition, there is an evident functional overlap and similarity between both types of stem cells, reflected in numerous shared gene expression patterns such as CD133, Sox10, nestin, vimentin, musashi, GFAP, and Olig1/2 [65,66,67]. Due to the migration ability of GSCs and the unique environment of SVZ (the vascular system of SVZ is richer than that of other brain regions), treatment-resistant GSCs easily migrate to and colonize the SVZ [68]. Consequently, numerous retrospective studies have confirmed that GBs in close contact with the SVZ possess more aggressive patterns of recurrence and worse clinical outcomes [67,69,70]. Therefore, new therapy strategies are being assayed with the aim of targeting SVZ to eradicate NSCs or GSCs [71].
The origin of GB, based on the stem cell theory, explains the versatility and plasticity of heterogeneous GB tumor populations. However, several studies provide evidence suggesting that partially differentiated glial cells, including oligodendrocyte and astrocyte precursors, may play a role in or be responsible for tumorigenesis [60,72,73]. The astrocyte de-differentiation theory is supported by experiments demonstrating the formation of tumors that are histologically similar to GB after activation of oncogenes and/or suppression of tumor suppressor genes in astrocytes [24,72,74,75]. Nevertheless, this manipulation in astrocytes results in their acquisition of stem-cell-like characteristics. Consequently, both hypotheses are not mutually exclusive and explain the presence of cancer stem cells within the tumor [64,76,77]. Moreover, the dedifferentiation of non-GSCs to GSCs further complicates the GSC-targeted therapy [25,78].
GSCs represent a very low percentage of cells within GBs, and are functionally defined and distinguished from their differentiated tumor progeny at central transcriptional, epigenetic, and metabolic regulatory levels [79,80]. Recognized markers of GSCs include CD133 (PROM1) [81], CD44 [82], SOX2 and nestin [76,83], but none of them are specific markers of GSC. Other putative biomarkers are CD15 (FUT4), A2B5 antigen, CD90 (THY1), integrin ITGA6, CD171 (L1CAM), S100A4, ATP-binding cassette transporters and the combination of CD44 and ID1 (reviewed in [84]). GSCs develop genetic variability and possess self-renewal capacity and specific characteristics that support tumor development, heterogeneity, recurrence, immunosuppression and radio and chemotherapeutic resistance [85]. Therefore, the heterogeneity of GB tumor cells can be attributed to the clonal evolution and differentiation/dedifferentiation capacity of GSC [25,66,72,77,86,87]. The GSCs’ ability to adapt to different niches implies that they can dynamically restructure their transcriptional program, inducing the transient expression of genes with specific functions for each cell state [18,36]. Furthermore, microglia and endothelial cells of the perivascular niche produce numerous growth factors that contribute to the support of proliferation, migration, and differentiation of NSCs and GSCs [27,88,89]. In turn, GSCs release transforming growth factor β (TGFβ) that enhances the tumor vasculature and can even transdifferentiate and generate endothelial cells or pericytes to form new tumor vascular niches [90,91]. GSCs exhibit elevated migratory and invasive potential, eliciting infiltration into healthy tissue, thus limiting the effect of total surgical resection and radiotherapy [92]. Residual cells have the ability to regenerate GB in brain regions distant from the initial tumor by acquiring new and different driver mutations that make them resistant to treatments [41]. Consequently, GSCs are more radioresistant than GB cells [93], can be resilient to TMZ-mediated cell death [94], and have mutations that facilitate recurrence after therapy [95]. DNA damage repair mechanisms, such as ATM, ATR, CHK1, and PARP1, are upregulated in GSCs, and CHK1 is preferentially activated following irradiation [96,97].Consequently, GSCs exhibit rapid G2-M cell cycle checkpoint activation and enhanced DNA repair [98]. The preferential activation of DNA damage checkpoint responses [34] and the increased expression of drug efflux pumps and antiapoptotic proteins [99] contribute to GSC recruitment after treatment. Interestingly, the inhibition of DNA repair protein RAD51 homolog 1 has been found to delay G2 cell cycle arrest, thereby sensitizing GSCs to radiation [100].
Ionizing radiation also enhances the motility, invasiveness and aggressiveness of GSCs. The increased motility and invasiveness result from the activation of the HIF(hypoxia-inducible factor)-1α, whereas aggressiveness is attributable to a pro-neural-to-mesenchymal transition associated with the activation of the STAT3 transcriptional factor [101]. STAT3 is overexpressed in GSCs [87] and plays a crucial role in sustaining stem-like characteristics [102]. Moreover, it enhances the expression of pro-tumorigenic genes related to cell cycle progression, extracellular matrix remodeling, as well as the secretion of cytokines and growth factors [103]. Consequently, STAT3 deletion or inhibition in GB cell lines markedly decreases tumorigeneses in vitro and in vivo [103,104] and has a radiosensitizing effect [93]. WP1066, one of the most promising STAT3 inhibitors, will be investigated in a phase II clinical trial for patients with recurrent malignant glioma [105].
GB cells have the ability to manipulate the TME to favor immunosuppression and to develop a niche sustaining tumor growth, invasion, migration, and survival [28,106]. GB cells can evade immune surveillance through the release of various soluble mediators such as TGFβ, IL-10, and PGE-2. In the presence of TGFβ, CD4+ T cells upregulate FoxP3 and differentiate into Treg cells with potent immunosuppressive potential. This cytokine inhibits the expression of cytolytic gene products (perforin, granzyme A, granzyme B, Fas ligand, and IFN-γ) which are co-responsible for CD8+ T-cell-mediated tumor cytotoxicity. Increased secretion of IL-10 is associated with enhanced expression of anti-inflammatory cytokines, such as IL-4, CCL2, and TGFβ. In the presence of IL-10, TAMs downregulate the expression of antigen-presenting molecules, thereby impairing CD4+ T cell activation. In turn, PGE-2 has been shown as a key mediator of immunosuppressive activity through the expansion of myeloid-derived suppressor cells (MDSCs) [107]. In fact, GSCs and GB cells play the role in recruiting and activating MDSCs [108] and M2 macrophages to drive immune suppression [109,110,111]. Simultaneously, GSCs protect themselves from T-cell-mediated killing by secreting extracellular vesicles containing programmed death ligand 1 (PD-L1) [112,113].
Consequently, new therapies that effectively target this important population may help to prevent recurrence and improve patient survival, and for sure, no single therapeutic modality will be effective against such a heterogeneous population of cells.

2.2. Metabolic Features Favoring Growth and Resistance

Metabolic reprogramming plays a crucial role in enabling GB invasive cells to generate the energy required for colonizing the surrounding brain tissue and adapting to hypoxic microenvironments [114,115]. The metabolism of GB is characterized by the upregulation of the PI3K/Akt/mTOR signaling pathway, a high rate of glycolysis, and increased lipid storage [116,117]. Aerobic glycolysis along with glucose consumption and lactate production supports rapid GB growth and correlates with a lower survival rate [118]. Nevertheless, GB cells adapt their metabolism according to glucose availability, which gives them extra resistance to hypoxia or altered redox situations. Selective pressure on GB cells makes them overexpress glucose transporters (GLUT1 and, particularly, GLUT3). GLUT3 has a five-fold higher affinity for glucose compared to GLUT1, thus facilitating glucose uptake in environments with lower glucose concentrations. Additionally, the acquisition of a stem cell state is associated with a significant increase in GLUT3 expression in induced pluripotent cells, and this overexpression correlates with poor glioma patient survival [119,120]. When glucose levels are low, HIF-1α guarantees the upregulation of GLUT3 and hexokinase-2, increasing the glycolytic pathway [121,122].
The activation of sterol regulatory element-binding protein-1, a crucial transcription factor controlling fatty acid and cholesterol synthesis, as well as cholesterol uptake, enables GB to obtain significant quantities of lipids essential for its rapid growth [123]. GSCs exhibit high expression of mediators of lipid metabolism, such as brain-fatty-acid-binding protein (FABP7), which leads to an increase in lipid contents that are specifically metabolized under glucose-deprived conditions [116]. GB cells direct significant amounts of lipids into specialized storage organelles known as lipid droplets, thus avoiding lipotoxicity. This process involves the overexpression of diacylglycerol acyltransferase-1 and sterol-O-acyltransferase-1, which convert surplus fatty acids and cholesterol into triacylglycerol and cholesteryl esters, respectively, increasing the storage as neutral lipids within lipid droplets [123].
Amino acids play a crucial role as important fuels for GB growth. Gene expression profiling has shown an upregulation of the L-Gln importer ASCT2 in GB compared to low-grade gliomas, and L-Gln deprivation has slowed tumor growth in some in vitro studies [124]. The L-Gln-derived glutamate and glucose-derived pyruvate are substrates for the glutamate-pyruvate transaminase 2 (GPT2), which synthetizes α-ketoglutarate. Through GPT2 upregulation, the anaplerotic replenishment of the TCA cycle is possible; otherwise, it is impaired by augmented pyruvate conversion to lactate. In other words, the Warburg effect, manifested as increased lactate release, drives L-Gln addiction in order to maintain the TCA cycle function [124]. Moreover, L-Gln has been shown to promote the mTOR-dependent signaling pathway, a potent driver of GB growth and progression [125,126]. Other amino acids are also utilized to fuel bioenergetic reactions and the synthesis of macromolecules in GBs [114]. L-Asp has been shown to be a limiting metabolite for GB cellular proliferation in hypoxic conditions [127]. L-Arg is involved in GB cell adhesion, and thereby in tumor cell migration and invasion [128]. L-Trp and L-Arg metabolism have also been linked to decreased detection by neighboring immune cells, creating a favorable environment [129].
In gliomas, autocrine glutamatergic signaling has been identified as a promoter of invasion [130]. GB cells release high levels of glutamate, which not only enhances tumor invasiveness but also promotes the turnover of GSCs [131]. In other words, GB cells create a positive feedback system whereby an excess of glutamate promotes their own growth and secondarily causes excitotoxicity-induced cell death in surrounding brain tissue [132]. It is probable that such tissue damage contributes to cerebral edema and the neurotoxicity associated with a growing GB. Consequently, the inhibition of glutamatergic signaling has been proposed as a strategy to mitigate GB-induced neurotoxicity [133].
Moreover, the invasive nature of GB is modulated by cell-to-cell crosstalk within the TME and altered expression of specific genes, such as ANXA2 (encoding the protein annexin A2, a Ca2+-dependent phospholipid-binding protein that helps to organize exocytosis of intracellular proteins to the extracellular domain) [134], GBP2 (encoding the guanylate-binding protein 2, which binds to guanine nucleotides and works in intracellular signaling) [135], FN1 (encoding fibronectin, which binds to integrins and facilitates adhesion, growth, migration, and differentiation) [136], PHIP (encoding the pleckstrin homology domain interacting protein, which regulates growth and survival of GB cells) [137], and SLC2A3 (encoding the glucose transporter 3) [114,138].

2.3. Ion Channels

Different studies have demonstrated the upregulation of Ca2+ selective ion channels in GB, contributing to invasion, proliferation and resistance to apoptosis [139]. By blocking L-type voltage-gated Ca2+ channels, cell invasion is inhibited as filopodia (also known as tumor microtubes, TMs) formation is blocked [140]. Indeed, GB cells possess the ability to extract specific signals from healthy neurons using TMs [141]. Furthermore, inhibition of T-type Ca2+ channels has been shown to induce apoptosis in GB cells [142]. Therefore, blocking Ca2+ could prevent tumorigenesis through several mechanisms, i.e., cell cycle progression, induction of apoptosis and inhibition of cell migration.
K+ ion channels play a crucial role in the proliferation and the resistance to apoptosis in GB. Specifically, certain voltage-gated K+ channels are overexpressed in GBs participating in signaling pathways that promote proliferation and inhibit apoptosis [143]. Some of these effects are due to the role of K+ channels in establishing the resting membrane potential, and therefore affecting the cell cycle. Different studies have shown that inhibition of K+ channels improves survival in GB patients, which emphasizes their role in GB development and progression [144]. Consequently, blocking of ion channels could represent an interesting therapeutic approach against GB progression.

2.4. Epigenetics of Glioblastoma

GB progression is associated with different types of epigenetic alterations, including histone modifications, DNA methylation, chromatin remodeling, and aberrant microRNA (miRNA) [145,146], a group of small non-coding RNA (19–22 nucleotide long) molecules that regulate the post-transcriptional degradation of mRNA [147]. Ciafré et al. performed the first experiment related to miRNAs in GB, investigating the expression of 245 miRNAs using microarrays [148]. The most interesting results came from miR-221 upregulation, and a set of brain-enriched miRNAs (miR-128, miR-181a, miR-181b, and miR-181c) that are down-regulated in GB. At the same time, miRNAs have been shown to be important regulators of gene expression and may also regulate cellular processes, including apoptosis, proliferation, invasion, angiogenesis, and chemoresistance [149,150]. Therefore, microRNAs can be classified according to their role in tumorigenesis (i.e., tumor suppressor or oncogenic). Table 1 summarizes what is known, at present, regarding miRNAs and their role in GB progression and resistance to therapies.
CircRNAs exert their biological effects through four different mechanisms: serving as sponges of RNA binding proteins, modulating parental gene transcription, encoding functional proteins and, most importantly, serving as sponges of miRNAs [189]. As is thoroughly reviewed in [189,190], circRNAs regulate GB proliferation and invasion and are also involved in angiogenesis activation. Good stability, broad distribution and high specificity make circRNAs promising biomarkers for GB prognosis and/or diagnosis, although their clinical implementation still has a long way to go.
Long-noncoding RNAs (lncRNAs) are a class of regulatory noncoding RNAs (>200 nt) that interact with DNA, RNA, and proteins to regulate various biological processes. As reviewed in [191,192,193,194], numerous studies have shown that lncRNA regulates the expression of genes involved in GB tumorigenesis (CHRM3-AS2, DLGAP1-AS1, DGCR10, LINC01057, LINC-PINT, MIR31HG, MIR210HG, NEAT1, NONHSAT079852.2, PVT1, SEMA3B, RBPMS-AS1), progression (ASLNC22381, ASLNC20819, CRNDE, DGCR10, HNF1A-AS1, HOXD-AS2, HRA1B, HOTAIRM1, LINC-PINT, PRADX, NEAT1, OXCT1-AS, TCONS-00004099) and therapeutic resistance (H19, MALAT1, MUF, DANCR, HOTAIR, HOTAIRM1, LINC00511, UCA1, OIP5-AS1, DANCR, FOXO3, HERC2P2) of GB cells. Moreover, lncRNAs exhibit stable secondary structure; thus, some of them (HOTAIR, GAS5, HOXA11-AS, HOTAIRM1, AGAP2-AS1, and AC002456.1) have been proposed as prognostic and diagnostic GB biomarkers [195,196,197,198]. More specifically, SBF2-AS1, MALAT1, CRNDE, TP73-AS1 and LINC00511 have been suggested as biomarkers of TMZ resistance in GB [198]. Lately, some evidence has indicated that lncRNAs also take part in GB cell metabolism. For instance, the lncRNA TP53TG1, under glucose deprivation, may promote cell proliferation and migration by influencing the expression of glucose-metabolism-related genes in glioma cells [199], and the lncRNA LEF1-AS1 facilitates the multiplication of GB cells and impedes apoptosis via the Akt/mTOR pathway [200]. Clinical trials involving the use of lncRNA as biomarkers for GB detection and prognosis are only in the recruitment phase but look promising.
Other epigenetic alterations, such as DNA methylation, histone modifications, and chromatin remodeling, are mechanisms involved in transcriptional activation of critical genes for GB development, lethality and resistance [145,201,202,203]. Thus, several epigenetic agents, including histone methyltransferase inhibitors, DNA methyltransferase inhibitors, histone deacetylase (HDAC) inhibitors, and other agents, are currently being tested for GB treatment in preclinical and clinical trials [146,203]. Protein arginine methyltransferase 5 (PRMT5) is a member of the PRMT family of proteins that plays a key role in the regulation of cellular signaling and gene expression by methylating histones as well as nonhistone proteins [204]. Nuclear expression of PRMT5 negatively correlates with glioma patient survival [205]. Engineered loss of PRMT5 or treatment with CMP5 (a PRMT5 inhibitor) results in apoptosis or loss of self-renewal for differentiated or undifferentiated GB cells, respectively, [206]. CMP5 derails the negative regulation of PTEN by PRMT5, which, in turn, decreases Akt activity in patient-derived GB neurospheres [207].
HDACs have been widely studied in GBM cells due to their relationship with therapeutic resistance, cell proliferation and invasion, angiogenesis and apoptosis [208,209,210,211]. In preclinical studies, HDAC inhibitors (HDACi) have proven to be effective anti-GB agents via multiple mechanisms, such as upregulating the expression of tumor suppressor genes, inhibiting oncogenes, inducing cell cycle arrest, promoting cell apoptosis and differentiation, inhibiting motility/migration, abolishing autophagy and tumor angiogenesis, and upregulating natural killer (NK)-cell-mediated tumor immunity [212,213,214,215]. Additionally, HDACis have demonstrated the capability to reduce cancer stem cell burden in GB tumors by modulating stemness, proliferation, differentiation, cell cycle arrest, apoptosis, autophagy and vasculogenic mimicry of GSCs [209,210,216]. Several HDACis (i.e., valproic acid, voristonat, panobinostat) have been assayed in clinical trials due to their capacity to act as chemo/radio-sensitizers and target GSCs [216]. Voristonat combination regimens with TMZ/radiotherapy and/or bevacizumab (BEV, recombinant humanized monoclonal antibody that blocks VEGFR-A) have proven to be tolerable (NCT01738646), but no statistical improvement in OS and/or PFS was noted [217]. Similar results were obtained with panobinostat in combination with BEV (NCT00859222) [218]. Valproic acid is a potent anticonvulsant that promotes apoptosis and impairs glioma cell proliferation and invasiveness and sensitizes GB cells to several anticancer drugs, such as TMZ, etoposide, gefitinib, nitrosoureas, and radiation therapy [219,220,221]. A meta-analysis [222] and a recent open-label phase II study [223] results seem to confirm that GB patients may experience prolonged survival due to valproic acid administration, providing further justification for a phase III trial of valproic acid/SOC. Levetiracetam (LEV), a relatively new antiepileptic drug, modulates HDAC levels ultimately silencing MGMT, thus increasing TMZ effectiveness in GCSs [224]. Retrospective analyses and an open-label phase II study (NCT02815410) seem to evidence that LEV improves GB patients’ PFS and OS [225,226]. So, it is perhaps time to reconsider the results performed in 2016, where a pooled analysis of a large series of cases treated with valproic acid or levetiracetam failed to find an association with patients’ survival [227]. A double-blind randomized clinical trial (ChiCTR2100049941) focusing on the clinical benefits of LEV + TMZ in the treatment of GB is ongoing in China. Nuclear imaging of HDAC expression in GB can be useful to improve the understanding and role of HDAC enzymes in gliomagenesis and identify patients likely to benefit from HDACi-targeted therapy [215,228].

2.5. The Angiogenetic Capacity of Glioblastoma

Aberrant vascular proliferation, necrosis, and infiltration of surrounding brain tissues are considered “hallmarks” of GB. Neo-vessels form from preexisting blood vessels due to VEGF expression by tumor and stromal cells under hypoxic conditions. The combination of VEGF with FGF (fibroblast growth factor)-2 or PDGF (platelet-derived growth factor) is known to synergistically enhance angiogenesis [229]. Vasculogenic mimicry (VM) is a new mechanism of tumor neovascularization in which highly invasive and genetically dysregulated tumor cells acquire vascular cell function, forming de novo vascular-like structures [230]. The involvement of GSCs in VM has been reported by several studies [231,232]. The disruption of GB vasculature through radiation or anti-angiogenic therapies induces a hypoxic microenvironment that promotes VM as an adaptative strategy to assist GB cells in surviving and progressing even when angiogenesis is blocked [233,234]. In keeping with this idea, the inhibition of vasculogenesis, but not sprouting angiogenesis, prevents the recurrence of GB after irradiation in mice [235].
SCs play a crucial role in VM, mainly due to their high plasticity and potential differentiation into endothelial-like cells [236]. The vascular niche is very important for the maintenance of GSCs as it promotes their survival and proliferation [230,237]. Additionally, communication between endothelial and tumor cells allows tumor vasculature formation and tumor cell dissemination [232,238]. Tumor vasculature has been considered a contributor to treatment resistance and relapse [239]. GSCs seem to be attached to the arterioles but not to the capillaries [240]. Arterioles transport, but do not exchange, gasses and nutrients, and promote a peri-hypoxic area. Integrin ligation causes an activation of the integrin-linked kinase leading to increased HIF-1α, as well as increased VEGF production [241]. HIF-1α acts as a potent activator of angiogenesis by stimulating the production of VEGF-A, PDGF and many other factors that initiate endothelial cell proliferation, invasion, and migration [242]. In GB, HIF-1α is not only influenced by oxygen but also by oncogenic signaling pathways, such as MAPK/ERK, p53, and PI3K/Akt/mTOR [243]. Although many approaches have been tried to inhibit HIF-1α, drugs that only target specific components of the hypoxia signaling pathway have generally failed to produce an enduring clinical response in GB. It is thought that the complete inhibition of HIF-1α is necessary to show potent antitumor activity and to promote the activation of the immune system [243]. Inhibition HIF-2α, which can also block the hypoxia pathway, is an alternative attractive strategy for GB treatment. HIF-2α is specifically overexpressed in GB cells and GSCs, but not in normal tissues [244]. Although the HIF-2α inhibitor PT2385 had limited activity in rGB (phase II, NCT03216499) [245], other HIF-2α inhibitors that are currently under research may help in blocking GB progression. It is also important to mention that recent findings suggest that GB hypoxia regulates gene expression in an HIF-independent way. In that sense, Srivastava et al. demonstrated that FAT1 (a FAT atypical cadherin) modulates the epithelial-mesenchymal transition and stemness gene expression in hypoxic GB [246], and hypoxia induces epigenetic regulation of the transmembrane protein odd Oz, altering DNA methylation status and activating the ODZ1-mediated migration of GB cells [202].
Importantly, Aderetti et al. demonstrated the existence of hypoxic peri-arteriolar GSC niches in GB tumor samples [247]. Apparently, GSCs remain attached to peri-arteriolar niches by the same receptor–ligand interactions as hematopoietic stem cells in the bone marrow. GSCs’ infiltration can be promoted via VEGF secreted by endothelial cells, which may induce the trans-differentiation of GSCs into endothelial cells, promoting angiogenesis and invasiveness [86]. Furthermore, a phenomenon of metabolic zonation has been described depending on the relative distance between the tumor cell and the blood vessel [248]. Proximity to the blood vessels promotes the mammalian target of rapamycin mTOR-derived anabolic metabolism and enhances tumor aggressiveness and therapy resistance [248]. Indeed, GB cells located in the perivascular tier exhibit robust anabolic metabolism and deviate from the Warburg principle by extensively engaging in oxidative phosphorylation. These perivascular cancer cells acquire specific functional characteristics, such as heightened tumorigenicity, enhanced migratory and invasive abilities, and surprisingly, remarkable resistance to chemotherapy and radiation; most of these traits are dependent on the mTOR pathway [248].
The BBB is a major obstacle to drug penetration within the brain parenchyma. Only 20% of small molecules/therapeutics agents cross the BBB and reach tumor cells at an effective concentration. GSC or GB cells protected against therapeutic agents by an intact BBB are the source of tumor recurrence [23]. P-glycoprotein, multidrug resistance proteins, organic anion transporters and breast cancer resistance proteins are especially important efflux pumps within the BBB that limit the accumulation of small-molecule-targeted therapies [249]. To make it more difficult, GSCs overexpress ABC transporters, further hindering drug delivery. ABC transporters promote therapy resistance by promoting the efflux of exogenous compounds, such as TMZ, at the cellular and BBB levels [22]. Infiltrating tumor cells are known to compromise the integrity of the BBB, resulting in a vasculature known as the blood–tumor barrier (BTB), which is highly heterogeneous and characterized by numerous distinct features, i.e., non-uniform permeability and active efflux of molecules [249]. Therefore, delivering therapeutic agents across the BBB and BTB, but avoiding their accumulation in the healthy parenchyma, is essential to making significant progress in GB treatment.

3. Present Therapy and Challenges

3.1. Standard of Care in Newly Diagnosed GB Patients

The Stupp protocol became the standard of care (SOC) for newly diagnosed GB (ndGB) patients since a randomized phase III trial (Table 2) evidenced an improved mOS from 12.1 to 14.6 months and an increase in the 2-year survival rate from 10% to 27% [7]. This SOC includes maximal safe resection, radiotherapy with concurrent (75 mg/m2/day × 6 weeks) and adjuvant TMZ (150–200 mg/m2/day × 5 days for six 28-day cycles). Since then, similar results (mOS 15–18 months) have been observed in other clinical studies [250,251,252]. Despite significant advances in the understanding of the molecular biology and pathophysiology of the GB, SOC has remained unchanged, excepting the possibility of adding or not tumor treating fields (TTFields) [13,253,254].
GB mostly recurs within 2–3 cm from the borders of the initial lesion and with multiple lesions, thus, maximal surgical resection improves survival irrespective of the age of the patient or the molecular status of the tumor [252,268]. Preoperative brain mapping techniques such as navigated transcranial magnetic stimulation (nTMS), magnetoencephalography, functional MRI, and diffusion tract imaging (DTI) are used to facilitate safe resections and minimize surgical complications [269]. Compared to non-nTMS techniques, nTMS has been associated in GB patients with smaller craniotomy size, less residual tumor tissue, shorter hospital stays, and improved survival at 3, 6, and 9 months, with no significant difference in surgery-induced neurological deficits [270].
During surgery, various tools are employed to optimize the extent of resection and minimize residual tumor volume. These include functional monitoring, fluorescence-based visualization of the tumor using 5-aminolevulinic acid (5-ALA), ultrasonography, and intraoperative MRI (ioMRI) [268,269,271]. Additionally, techniques like evoked potentials, electromyography, and brain mapping in awake patients, under local anesthesia, are used to monitor and preserve language and cognition during resections in critical brain areas [272]. The use of the amino acid 5-Ala helps to identify tumor volume and areas of neoplastic infiltration through fluorescent visualization, improves PFS, OS, and reduces postoperative neurological damages [269,273,274,275,276]. 5-Ala has also been effectively used in rGB resection, but the risk of false-positive fluorescence for reactive non-tumor tissue is more remarkable in relapse forms, likely due to an altered BBB [277]. Nevertheless, recently, an off-label fluorophore (sodium fluorescein) has become popular due to numerous benefits compared to 5-Ala, including lower cost, non-toxicity, easy administration and a wide indication for other brain tumors [278]. Microsurgical resection of GB using sodium fluorescein has been associated with an increased GTR rate and OS [279], although it is still considered inferior compared to 5-Ala [280].
Intraoperative ultrasound (ioUS) involves the use of sonography to locate tumor tissue during surgery and to delineate it from healthy brain tissue. As opposed to 5-Ala, which can only identify high-grade gliomas, ioUS is able to identify both low- and high-grade gliomas. In practice, 5-Ala and ioUS are considered complementary techniques [269]. Intraoperative magnetic resonance imaging (IoMRI) improves the accuracy and definition of the tumor and provides near real-time information about the dynamic changes occurring during surgery [281]. Analysis of residual GB volumes and neurological outcomes demonstrates that ioMRI is significantly superior to 5-Ala and white-light surgery at comparable peri- and post-operative morbidities [282]. The combination of ioMRI and 5-Ala facilitated achievement of the highest extent of resection (95%), followed by ioMRI alone (94%), 5-Ala alone (74%), and no imaging (73%), and this was associated with fewer post-chirurgic neurological deficits [283,284]. However, the lack of evidence regarding the cost-effectiveness compared to less advanced techniques raises uncertainty [268,285]. Regardless of the technique used, a postoperative contrast-enhanced MRI should be carried out within 48 h to assess the extent of resection and serve as a baseline for further treatments. Additionally, MRIs are performed every 2–3 cycles of TMZ treatment to monitor the tumor’s response [286].
After surgery, the smallest amount of residual tumor correlates with higher survivals [287,288]. However, radical surgical resection is limited by the highly invasive nature of GB cells [289]. Additionally, postoperative complications are a negative prognostic factor, and in this it is essential to prevent permanent neurologic deficits to safeguard the quality of life of the patients [14,47,289]. Carmustine (BCNU) wafers placed in the tumor resection cavity at the time of surgery provide a modest survival advantage (≈2 months) [290]. Wafer implants have been approved by the FDA and the EMA, but are not included in the SOC mainly due to their limited brain penetration, safety and tolerability, and because the treatment may preclude patients from enrolling into clinical trials [291]. Where surgical resection is not possible, stereotactic biopsy or open biopsy are alternative options for histological diagnosis and further molecular testing, which can determine an optional therapy [292,293]. However, this recommendation is not exempt from criticisms since in GB patients with low-performance status and/or advanced age, biopsies imply very little clinical gain [294].
Compared with surgery alone, postoperative radiotherapy is used to control microscopic unresectable disease, delay neurological deterioration and increase survival [7,295]. Radiotherapy is less efficacious in hypoxic TME due to a lower oxidative stress and because cancer cells develop mechanisms to repair DNA [115].
TMZ is a mono-alkylating agent that induces cytotoxic lesions including N7-methylguanine, N3-methyladenine and O6-methylguanine. N7-methylguanine and N3-methyladenine are repaired by base-excision repair (BER) and contribute minimally to the overall cytotoxicity of TMZ, while O6-methylguanine is repaired by O6-methylguanine-DNA-methyl transferase (MGMT) [296]. Methylation of the MGMT gene promoter (40% of GB patients) causes a reduction in MGMT protein expression and activity that results in persistent O6MeG lesions that trigger replicative stress and cytotoxicity via futile cycles of mismatch repair (MMR) [297]. Therefore, MGMT promoter methylation confers a better prognosis and overall survival (OS) associated with a positive response to alkylating agents in GB patients aged <70 years [7,13,298]. Radiotherapy has been shown to upregulate MGMT, whereas prolonged exposure to alkylating agents may suppress MGMT activity making the cells more susceptible to TMZ [257]. Nevertheless, several trials (Table 2) evidence that extending post-radiation TMZ from 6 to 12 months does not improve PFS6 and is associated with greater toxicity, functional deterioration, and poorer quality of life [257,258].
TMZ is a cornerstone of GB treatment, but its effectiveness is limited by the blood–brain and blood–tumor barriers, and the inherently or acquired GB resistance [19,299,300,301]. Upon TMZ treatment, GB and GSC cells induce DNA repair mechanisms, NF-kB signaling mediated antiapoptotic pathways, the expression of anti-apoptotic Bcl-2 family members, EGFR activity, drug efflux by ATP-binding cassette (ABC) transporters, autophagy-mediated resistance, expression of STAT3 and miRNAs, and overexpression of antioxidant proteins [83,299,301,302,303,304,305]. Nitrosoureas, i.e., lomustine (CCNU), carmustine and procarbazine, were widely used before the availability of TMZ, but their use is now limited to the treatment of rGB. At is shown in Table 2, patients in good physical condition with hypermethylated MTMG promoters (NCT01149109) slightly increase their OS survival by the addition of lomustine to SOC (48.1 vs. 31.4 months) [259,306]. Nevertheless, the benefit of this regimen remains unclear since the sample size was small and few patients were able to complete all six cycles of adjuvant treatment due to the greater hematologic toxicity in the lomustine-TMZ arm [307]. The addition of BEV to the SOC improved PFS but not OS in both AVAglio and RTOG 0825 trials (NCT00943826 and NCT00884741) [260,262].
Up to now, TMZ is still commonly used for GBs with unmethylated MGMT promoters, due to the lack of significant benefits of alternative options (BEV plus irinotecan, dose-dense TMZ, BEV+SOC) [257,260,262,298]. Several preclinical studies demonstrated that O6-benzylguanine (O6BG) or O6-bromothenylguanine inactivate MGMT, but the addition of O6BG to radiation and BCNU treatment (Table 2, NCT00017147) did not provide further benefit and instead increased toxicity [264].

3.2. Tumor-Treating Fields (TTFields)

TTFields is a non-invasive cancer treatment modality that applies low-intensity (0.7–3 V/cm), intermediate-frequency (100–500 kHz), and alternating electric fields over regions of the body where tumors are localized [308,309]. In growing GB cells, TTFields cause chromosome missegregation, disrupt DNA repair, inhibit mitosis and the cell cycle, and induce apoptosis and autophagy [310,311,312,313,314,315,316,317]. TTFields also interfere with the directionality of cancer migration by inducing changes in the organization and dynamics of microtubules and actin and ablate the primary cilia on GB cells that contribute to tumor growth and chemoresistance to TMZ [318]. TTFields also downregulate the expression levels of VEGF, HIF-1α, and matrix metalloproteinases (MMP2 and MMP9), which are necessary for tumor growth, invasion and metastasis [319]. TTFields also increase the membrane permeability of cancer cells and the BBB [320,321], which can help to increase the uptake and bioefficacy of chemotherapeutic drugs. Although this treatment modality reduces the viability of proliferating T cells [322], it also stimulates maturation and phagocytosis by dendritic cells (DCs) [323], increases CD8 T infiltration in TME [315], promotes the production of type I IFNs in GB cells in a cGAS/STING- and AIM2 inflammasome-dependent mechanism [324] and, thereby, facilitates the immune system response. Interestingly, the combination of hyperthermia and TTFields has shown synergistic effects in GB [325].
Over the past decade, TTFields have emerged as a complementary treatment strategy, which is now part of the SOC in GB treatment [289,326,327,328]. The FDA’s approval of rGB was based on the results from the EF-11 trial (NCT00379470) showing that TTFields monotherapy provided similar efficacy compared to the best physician’s choice chemotherapy in patients with rGB, albeit with better quality of life, less toxicity and a lower incidence of serious adverse events [329]. A randomized clinical trial in ndGB patients (NCT00916409) previously treated with chemoradiotherapy showed that patients treated with the TTFields and TMZ had a median free survival (mPFS) of 6.7 months compared to 4.0 months with TMZ alone. The addition of TTFields to the SOC therapy improved median OS (mOS) from 15.6 to 20.5 months without a negative influence on the health-related quality of life [13,330,331]. In ndGB, TTFields are applied within 6 weeks after the end of the radio-chemotherapy, ideally simultaneously with TMZ monotherapy [13,328]. Patients with compliance > 90% showed extended median and 5-year survival rates [256]. The most common adverse effect is skin irritation, occurring in 43% of patients (2% grade 3 or higher) [13,309,327,332], which is generally managed with array relocation and topical treatments including antibiotics and steroids. The frequency of systemic adverse events was 48% in the TTFields-TMZ group and 44% in the TMZ-alone group. Several limitations should be noted in the NCT00916409 trial: (a) only PF patients after the completion of chemoradiation were enrolled, which excluded those who were more likely to have a poor prognosis; (b) randomization in the EF-14 trial occurred over 2 months after diagnosis, which suggests a selection bias of patients who did not have progression after the initial treatment and would therefore likely have a better survival rate; (c) a “sham” device—to better discern a potential placebo-effect of wearing the device—was not used; (d) second-line therapies (chemotherapies, salvage radiation, radiosurgeries or craniotomies) after tumor progression in both groups were not reported while the TTFields plus TMZ group allowed patients to continue TTFields for up to 24 months or after the second GB progression; (e) molecular markers, such as the IDH1/2 status, were not performed [308,333]. Recently, recognized brain cancer experts concluded that TTFields plus TMZ represents a major advance in the field of GB therapy, though other experts maintain their skepticism regarding the use of the TTFields because of the lack of effect in some patients and because the time lengths required to reach (modest) benefits (at least 18 h per day) limit its utility [308,309,334].
Dexamethasone is administered to GB patients to alleviate cerebral edema and provide symptomatic relief. However, the corticoid-induced immunosuppressive effects may also increase infections and decrease survival [335,336]. In fact, a recent meta-analysis revealed that dexamethasone interferes with the therapeutic effects of TTFields [324]. The threshold dose at which dexamethasone can be used with minimal interactions with the TTFields was 4.1 mg per day or lower [337]. Several ongoing clinical trials are studying the optimal timing for TTFields administration (e.g., NCT04471844, NCT04492163, NCT03705351) and the safety and efficacy of the combination of TTFields with other cancer modalities [308,315,316,338,339]. For instance, PriCoTTFields is a phase I/II clinical trial that evaluates the safety and efficacy of TTFields initiated prior and concomitant to combined radiation and TMZ therapy in ndGB patients [340].
TTFields can reduce the DNA double-strand repair by downregulating the activity of the breast cancer type 1 susceptibility (BRCA1) signaling pathway, thereby increasing the sensitivity to the blockade of DNA repair caused by PARP inhibition [341]. Consequently, an ongoing phase II trial (NCT04221503) will try to determine whether niraparib (a PARP inhibitor) can enhance the effect of TTFields in GB patients (NCT04221503). In addition, the combination of TTFields, TMZ and lomustine has shown benefits in ndGB patients [342] and the triple combination of BEV, irinotecan, and TMZ plus TTFields improved the OS of patients with rGB [343]. Mechanisms involved in the acquisition of TTField resistance include activation of voltage-gated Ca2+ channels linked to cell migration [344]; CDK2NA deletion, mTOR (V2006I) mutations [345], and the upregulation of autophagy which can be reversed by combining TTFields with an autophagy inhibitor [312].
In clinical practice, TTFields are mainly used at a frequency of 200 kHz, but preclinical studies show that different GB cell lines respond to other optimal electric frequencies, as is the case of SF188 (400 kHz) or U87 (100 kHz) [346]. This phenomenon highlights the need for further investigation to individualize “TTFields prescription”. Despite the advances associated with the incorporation of TTFields to GB treatment, its clinical use is still quite restricted. EANO guidelines argue that the clinical benefit of TTFields has not been established yet, which contradicts the ASCO-NSO recommendations [253,347]. Certainly, price, regulation, the increase in the efficacy of combined treatments, and likely the development of novel intracranial electrodes, may assist in increasing the utilization and acceptance of TTFields [316].

3.3. Treatment in Special Patient Populations

Elderly patients (>65 years) or patients with a poor functional status have worse prognosis and are less tolerant to toxicities. Surgical resection is not associated with improved survival [348], but according to a recent retrospective single-center study, BCNU wafer implantation during the surgical resection is safe and improves mOS 39.0 months (≥12 implanted wafers) vs. 16.5 months (<12 implanted wafers) in patients in “extreme” neurosurgical conditions (>80 years and patients with preoperative Karnofsky Performance Status score < 50) [349]. Although just a few patients (6/49) reached that number of implants, these results are impressive, since mOS in the “extreme” conditions subgroup was 10.0 months, and there was a significant improvement in the postoperative KPS score compared to the preoperative KPS score.
The combination of TTFields with maintenance TMZ resulted in improved PFS and OS in ≥ 65-year-old patients with ndGB in the phase III EF-14 trial, without affecting patient quality of life [350]. Nevertheless, clinical trials have shown that standard radiotherapy is associated with poor outcomes, especially in patients older than 70 years (Table 1, ISRCTN81470623) [266]. Here, abbreviated courses of radiation therapy must be considered [266,267], although age alone should not represent the sole determining factor for the duration and intensity of the therapy [351]. Hypofractionated radiotherapy schedule (40 Gy delivered in 15) fractions and the addition of concurrent and adjuvant TMZ (NCT00482677) significantly increase survival (9.3 vs. 7.6 months, respectively) without impairing the quality of life [267]. Consequently, partial-brain fractionated radiotherapy with concurrent and adjuvant TMZ is the SOC for elderly patients with good performance status [352,353]. The addition of BEV to radiotherapy had no benefits in elderly patients [354].
A single modality therapy can be considered for patients with poor functional status. RT was more effective than TMZ for unmethylated MGMT-promoter tumors, whereas TMZ was more effective than RT for methylated MGMT-promoter tumors [266,267].

3.4. Options of Treatment in rGB patients

Regardless of the use of multimodality treatments, GB invariably returns after a median interval of less than 10 months, and typically even sooner (≈6 months) in older patients [267]. The genetic and biological changes induced by radiotherapy and/or cytotoxic chemotherapy differentiate rGB from primary tumors. These changes empower GB tumors to navigate the host microenvironment, evade the immune system, and foster intrinsic and acquired resistance to further administration of radiation and/or alkylating agents. Upon recurrence, patients typically exhibit a poor performance status and compromised overall health, with GB tumors often being unresectable, thus requiring substantial use of corticosteroids to manage cerebral edema [355]. This makes rGB prognosis much worse than that of the primary GB.
Actually, although there is no clear SOC salvage therapy for rGB [289], patients who received no salvage treatment had poorer survival than those who received radiation and/or chemotherapy [356]. Therefore, re-resection, re-irradiation and systemic chemotherapy with TMZ rechallenge, nitrosoureas, BEV, and TTFields or clinical trial enrolment to test experimental drugs are considered for all recurrent patients [295,357,358,359,360]. Unfortunately, fewer than 43% of rGB patients were fit enough to be included in clinical trials [361].
Consensus guidelines for selecting candidates for second surgery recommend that patients need to have a good performance status, particularly if more than 6 months have elapsed since the initial surgery [357,362]. According to a retrospective review of the brain tumor database (1997–2016), stereotactic radiosurgery is associated with longer OS and/or PFS in rGB patients with good performance status and small-volume tumor recurrences [363]. In practice, not more than 20–30% of relapsed patients are eligible and only complete resections have any survival benefit (11–17 months) [364,365]. Toxicity to normal brain parenchyma limits re-irradiation in rGB [366]. Radiosurgery or hypofractionated radiotherapy (30–35 Gy in 5–15 fractions) is considered a potentially effective option and is increasingly used for younger patients with good performance status [356,367]. Data from a few prospective studies in rGB suggest that re-irradiation modestly improves PFS compared with systemic treatment alone [356]. OS after re-irradiation (9.7 months) was sufficient to justify this treatment [367,368], but marginal recurrence is significantly more frequent in patients who had prior BEV exposure [369].
Lomustine has become the SOC at relapse in Europe, with thrombocytopenia being the most frequent limiting toxicity [358]. Lomustine is generally preferred to other nitrosoureas given its oral formulation, schedule of administration, and better safety profile. However, lomustine activity is largely restricted to patients with tumors with MGMT promoter methylation and its survival benefit has been found limited: objective response rate was around 10%, mPFS < 2 months, PFS6 was 20%, and OS was 6–9 months [358,370].
One of the most significant features of GB is its hypervascularization, mainly promoted by the hypoxia-facilitated VEGF overexpression in tumor and stromal cells [371]. BEV is an anti-VEGF humanized monoclonal antibody that inhibits tumor-driven angiogenesis and may help in reducing patients’ immune suppression [234,372,373]. rGB with a low apparent diffusion coefficient, large tumor burden, or IDH mutation is more likely to benefit from BEV treatment [374]. BEV gained approval in 2009 for rGB treatment in the US and later in other countries, but BEV is not approved by the EMA as an SOC for rGB [375,376]. BEV has shown promise in extending PFS treating GB, but there is no evidence for its ability to prolong OS [377,378,379,380,381]. The anti-angiogenic effect of BEV decreases contrast uptake during MRI, which can lead to false negatives in recurrences [370]. Despite this, BEV is almost used due to the lack of alternative treatment options, and because it also serves to control brain vasogenic edema [289,375] and to avoid the need for corticoid treatment [382,383]. BEV combined with re-irradiation was found to be safe and tolerable and showed a significant reduction in the incidence of radiation necrosis, patient dependence on corticosteroids and improvement in the Karnofsky score during disease progression-free periods. Survival benefits (10.1 months) have been reported following fractionated stereotactic radiotherapy (35 Gy/10 fractions) and concurrent BEV in a prospective randomized phase II trial [384,385]. A recent retrospective study showed that BEV combined with re-irradiation improved mPFS and mOS to 8 and 13.6 months, respectively [359]. Nevertheless, the validity of these results is constrained by the inclusion of a small number of patients, the heterogeneity of treatment options, and the absence of a control group. Despite these limitations, recent conclusions drawn from a meta-analysis endorse the benefits of this therapeutic option [386]. Lomustine plus BEV for rGB (phase II, NCT01290939) somewhat prolonged PFS but did not confer a survival advantage over treatment with lomustine alone [387]. Although earlier reports suggested that BEV had glucocorticoid-sparing effects, in this trial, the addition of BEV did not reduce the use of glucocorticoids [387].
TTFields did not increase OS (phase III, NCT00379470) but showed efficacy equivalent to chemotherapy commonly used for rGB, with lower toxicity and improved quality of life [329]. According to a recent phase II trial (NCT01894061), the combination of BEV and TTFields is safe and has clinical efficacy in rGB [388].

4. Targeted Therapies

Genetic changes that have been well recognized in GB cells include alterations in the Rb/p16 pathway (> 90%), loss of heterozygosity of 10q (70%), EGFR amplification or mutation (≈50%), TP53 mutations (31%), PDGF receptor gain/amplification (≈25%), mouse double minute homolog 2 (MDM2) gene mutations (10–15%) and the phosphatase and tensin homolog (PTEN) gene mutations (20–34%) [16,389]. Analysis of the large-scale molecular and genomic information present in the Cancer Genome Atlas Program (TCGA) database indicated that p53 pathway (TP53/MDM2/P14arfç), the PI3K/Akt/mTOR pathway, and the RB pathway (CDK4/RB1/P16ink4) are the main signaling pathways involved in GB tumorigenesis, pathophysiology and acquisition of resistance to treatment [16,390,391]. Intrinsically targeting these altered molecules and pathways was seen as a novel avenue in GB treatment. Unfortunately, despite research efforts and clinical trials, except for prolonged PFS afforded by the BEV, no pharmacological intervention has been demonstrated to alter the course of disease [392,393].

4.1. pRB/CDK4/RB1/P16ink4

The retinoblastoma (RB) gene encodes a tumor suppressor protein (pRB) that inhibits the progression of the cell cycle. This gene is inhibited by the cyclin-dependent kinase (CDK) complex, especially CDK4, CCND2, and CDK6. The pRB-controlled pathway is often disrupted due to CDK4/6 amplification or CDKN2A/B loss or mutation [16]. The G1-S phase cell cycle checkpoint is mainly controlled by the kinases pathway, and thus improper formation of the cyclin D-1 complexes with CDK 4/6 leads to the promotion of cell-proliferation-involved carcinogenesis. Homozygous deletion of the CDKN2A-p16INK4α gene in chromosome 9p is one of the three genetic traits of primary GB; in fact, activation of CDK4 is commonly observed in GB cells leading to cell invasion and stemness [394,395,396,397]. Three CDK4/6 inhibitors, including palbociclib, ribociclib and abemaciclib, have been approved by the FDA as monotherapy for treating breast cancers and are under clinical investigation in GB patients (Table 3) [398]. Palbociclib enhanced radiotherapy cytotoxicity in GB xenografts with RB expression [395] but failed to provide a benefit (NCT01227434) [399]. Ribociclib monotherapy exhibited good CNS penetration but showed limited clinical efficacy in patients with rGB, a fact that was attributed to upregulation of the PI3K/mTOR pathway [400]. A recent pharmacokinetic study [398] shows that abemaciclib penetrated into the human brain to a larger extent and was retained longer, thus representing a better treatment option. In GB xenograft models, combined treatment with CDK4/6 inhibitor and oncolytic virus (VSVΔ51) induced severe DNA damage stress, amplified oncolysis, inhibited tumor growth, and prolonged survival [401]. In the phase II trial (NCT02977780), the addition of abemaciclib to the SOC treatment in ndGB patients was well tolerated and prolonged PFS, but there was no evidence of an OS improvement compared to standard radio-chemotherapy [402,403].
CDKN2A encodes two tumor suppressor proteins INK4a (p16INK4a) and ARF (p14ARF), which are crucial regulators of the pRB- and p53-dependent growth control, respectively, [470]. One of the most frequent mutations found in GBs is the homozygous deletion of the p16INK4a/p14ARF/p15INK4b locus [471]. Loss of p14ARF promotes the accumulation of the p53 repressor, reducing p53 levels, and thereby promoting tumorigenesis. In turn, P16INK4a inhibits the association of CDK4 and CDK6 with cyclin D. When p16INK4a is lost, CDK4 and CDK6 associate with cyclin D and participate in the phosphorylation (inhibition) of the RB protein, which in turn facilitates the release of E2F (E2 transcription factor), leading to aberrant cell proliferation. Furthermore, Labuhn et al. found that the co-deletion of ARF and INK4a increased, accordingly with tumor progression, from low- to high-grade gliomas, thus suggesting that deletions of this locus may be fundamental for GB development [471]. A novel piperazine-based benzamide derivative regulates the cell-cycle-related proteins and influences the p16INK4a-CDK4/6-pRb pathway. It significantly inhibited the growth, migration, and invasion of human GB cell lines in vitro. The most interesting aspect of this study is that it could penetrate the BBB with an exceptional brain-to-plasma ratio of 1.07 in vivo, which was accompanied by a superior anti-GB potency on the U87-MG-xenograft model without any apparent host toxicity [472].

4.2. TP53/MDM2/P14arfç

TP53 is a gene that encodes the p53 tumor suppressor protein, commonly referred to as the “Guardian of the Genome” due to its ability to respond to genotoxic stress and to protect the genome by inducing a variety of biological responses including DNA repair, cell cycle arrest, and apoptosis [473,474]. The principal regulators of p53 are the proto-oncogenes MDM2 and ARF (open reading frame). MDM2 is an E3 ubiquitin ligase that hampers p53 by promoting its ubiquitination and subsequent degradation through the proteasome [475]. Conversely, ARF triggers the activation of p53 through direct physical interaction with MDM2, effectively blocking MDM2’s ability to interact with p53. This intricate regulation has fueled intensive research efforts and the development of successful therapeutic approaches to modulate p53 in recent years [476,477,478]. The p53-ARF-MDM2 pathway is dysregulated in 84% of patients with GB and in 94% of the GB cultured cell lines [479]. Furthermore, there is a high occurrence of TP53 and ARF co-inactivation observed in GB [480]. p53 mutations play a particularly significant role in the development of secondary GBs [470], and the insurgence/progression of GB, as well as the related chemoresistance, have often been attributed to MDM2 overexpression [481,482].
Activation of p53 has become a pivotal therapeutic objective within this regulatory trio. In China, starting in 2003, wild-type p53-induced expression within tumor sites has been utilized as a therapeutic approach for several cancer types [480]. Several pharmacological agents have been assayed in order to restore normal p53 functions. One option is to inhibit the MDM2/p53 complex to prevent degradation of the p53, thus restoring the function of wild-type p53 in tumors with mutant p53, and inhibiting gain-of-function mutations in mutant p53 [474,478]. Combining an MDM2 inhibitor with chemotherapy or radiotherapy, higher levels of p53 can be achieved, leading to the activation of apoptosis [482], which is also a viable strategy to overcome resistance to therapy [483,484]. AMG232 (a p53-MDM2 inhibitor) exhibited the most remarkable efficacy against GB stem cells and spheroids [479,485]. The first molecules reported to interrupt the p53–MDM2 interaction were nutlins (1, 2, and 3) [486]. Preclinical studies show that nutlin-3 induces apoptosis and cellular senescence in human GB cancer cells and enhances the efficacy of radiotherapy [487]. TMZ/nutlin3a was synergistic in decreasing growth of wild-type p53 GB cells. The inhibition of cell growth following exposure to TMZ/nutlin3a correlated with (1) activation of the p53 pathway; (2) downregulation of DNA repair proteins; (3) persistence of DNA damage; and (4) decreased invasion. In a xenograft model of intracranial GB, TMZ/nutlin3a treatment resulted in a significant increase in survival compared with single-agent therapy [483]. Nutilin-3 was followed by the development of other MDM2 inhibitors such as idasanutlin (RG7388, NCT03158389), AMG232 (NCT01723020), navtemadlin (NCT03107780) and BI-907828 (NCT05376800), which are undergoing clinical trials for GB and other brain cancers [482].

4.3. PI3K/Akt/mTOR

Constitutive activation and mutation of this [488] signaling pathway play crucial roles in the development of GB and are present in GSCs [297,489]. As illustrated in Figure 3, engagement of an extracellular domain with its ligand activates the intracellular tyrosine kinase that translocates to the plasma membrane resulting in the formation of phosphatidylinositol 3,4,5-triphosphate (PIP3), which further stimulates the serine/threonine kinase phosphoinositide-dependent kinase 1 (PDK1) and protein serine-threonine kinase (Akt, also known as protein kinase B) activities [490]. The tumor suppressor phosphatase and tensin homolog deleted from chromosome 10 (PTEN) negatively regulate the PI3K/Akt pathway by removing the phosphate group from PIP3 to PIP2 [491,492]. PTEN also functions as a lipid phosphatase and in this manner; it is capable of regulating cell polarity, motility and senescence [493] and, in addition, is involved in modulating innate and adaptive immune responses [494]. Akt is a crucial regulator of cell proliferation and survival and is hyperactivated in many cancers. After Akt phosphorylation and induction, mTOR, as a downstream target, is activated. In the PI3K pathway, mTOR acts as a downstream effector and also as an upstream regulator of apoptosis and cell cycle progression. mTOR is a protein kinase localized in two structurally and functionally distinct multiprotein complexes known as mTORC1 and mTORC2 [495,496]. Via the PI3K/Akt pathway, growth factors, low energy status, low oxygen level, and DNA damage converge in mTORC1 activation which, in turn, influences cell growth and proliferation by promoting biosynthetic pathways, limiting catabolic processes, and inhibiting autophagy [497,498]. Differently, mTORC2 activation is triggered by growth factors but does not respond to nutrient availability. Once activated, mTORC2 drives reorganization of the cytoskeletal structure, motility, cell proliferation, and survival and is also involved in the induction of the Warburg effect [495,499]. Recently, it has been shown that acetyl coenzyme A is used by GB cells to induce RICTOR acetylation (a core component of the mTORC2 signaling complex) that results in mTORC2 activation. This mechanism creates an autoactivation loop whereby mTORC2 triggers cell proliferation and growth, bypassing growth factor-activated upstream signaling and rendering GB cells resistant to receptor tyrosine kinase and mTORC2 inhibitors [500].
The PI3K pathway is altered in about 70% of GBs, either by deletion of PTEN or amplification of EGFR and/or VEGFR and/or PDGF receptor (PDGFR) [470,501,502,503]. PTEN is deleted or mutated in 30–60% of GB cases and, consequently, the PI3K signaling pathway is hyperactivated, which in turn accelerates tumor growth, progression, and metastasis [504,505].
EGFR alteration, including overexpression or gene amplification, is present in 50–60% of GBs [9]. EGFR can activate a variety of signal transduction pathways (PI3K/AKT/mTOR, RAS/RAF/MEK/ERK, JAK/STAT, and PKC)[506,507,508]. EGFR gene amplification is detected in 57.4% of primary GBs, whereas the EGFR variant III (EGFRvIII) is a characteristic mutation that emerges later in tumor development [509,510]. EGFRvIII is characterized by a deletion of 267 amino acids in the extracellular domain, leading to a receptor that is unable to bind to a ligand yet is constitutively active [511,512]. Together with its impaired internalization and degradation, the EGFRvIII enhances the tumorigenic potential of GB by activating and sustaining mitogenic, anti-apoptotic and pro-invasive signaling pathways [513]. EGFRvIII is associated with a more aggressive tumor behavior and worse prognosis and contributes to therapy resistance [514,515,516,517]. However, GB samples taken from the first diagnosis and at the time of progression show that in 80–90% of cases, the EGFR amplification status is unchanged, whereas the expression of EGFRvIII often changes [429].
First-generation EGFR inhibitors (gefitinib, erlotinib and lapatinib) were designed to orthosterically block the ATP/substrate-binding pocket of EGFR (Figure 3) [510]. Although these inhibitors showed promising results in inhibiting growth and improving survival in preclinical models, no benefits were observed in clinical trials (Table 3) [435,444,445,518,519,520,521,522,523,524]. Erlotinib as monotherapy or as part of combined treatments has been assayed in multiple trials in rGB and ndGB (Table 3). It is worth mentioning the study carried out by D’Alessandris et al. in which, despite not giving positive results, the administration of BEV and erlotinib was tailored to the molecular profile of the patient’s tumor (VEGF overexpressing tumors or EGFRvIII tumors, respectively) [436]. A phase II trial showed that ndGB patients treated with erlotinib plus SOC had significantly improved OS (19.3 vs. 14.1 months in historical controls) [438], which is surprising because just one year later, another phase II trial reported that combined treatment not just was ineffective but also had an unacceptable toxicity [439]. The effectiveness of erlotinib was also assessed in combination with SOC and with BEV in patients with unmethylated MGMT promoter (Table 3), with a lack of survival benefits [437,439]. Lapatinib, which binds to EGFR better than erlotinib or gefitinib, showed a modest inhibition of EGFR achieved in biopsied post-treatment tumors, highlighting the important limitation of crossing the BBB [525,526].
Second-generation EGFR inhibitors (afatinib and dacomitinib) are designed to bind irreversibly to the tyrosine kinase domain of EGFR and other ERBB family members [527]. Both showed good safety but limited single-reagent activity in clinical trials in rGB patients (Table 3) [404,427,528]. Afatinib (with and without TMZ) showed limited activity in unselected patients, but an increase in the PFS was observed in patients overexpressing EGFR or expressing EGFRvIII [404]. Dacomitinib (PF-00299804) had limited activity in rGB with EGFR amplification, although the molecular characterization of four patients with positive response can be useful to select patients who could benefit from EGFR inhibition [427,528,529]. The third generation of EGFR inhibitors (rociletinib and osimertinib) was designed to target the T790M resistance mutation, which is responsible for about 50% of the acquired resistance to the earlier generation of tyrosine kinase inhibitors [530,531]. Osimertinib (AZD9291) induced GB cell cycle arrest and significantly inhibited colony formation, migration, and invasion to a greater extent than other EGFR tyrosine kinase inhibitors, and prolonged survival of orthotopic GB-bearing mice [532]. Compared with afatinib, AZD9291 demonstrated lower potency in inhibiting EGFRvIII, but it showed excellent BBB penetration making it an attractive candidate for inhibiting EGFR in GB. ZD9291 has been approved for the treatment of lung cancer with good safety and tolerability. ZD9291/BEV combination was marginally effective in most rGB patients with simultaneous EGFR amplification and EGFRvIII mutation, but it is interesting to mention that OS was superior to regorafenib (7.4 months) and a subgroup experienced a long-lasting meaningful benefit [533]. Several anti-EGFR monoclonal antibodies, such as cetuximab, GC1118, ABT-806, ABT-414 and nimotuzumab (Table 3 and Figure 3) failed in clinical trials to increase OS in GB patients [423,424,429,430,451,534,535]. Depatuxizumab mafodotin (depatux-M, ABT-414) is a tumor-specific antibody–drug conjugate comprising ABT-806 and the toxin monomethyl auristatin-F. It was evaluated in a randomized controlled phase II trial (NCT02343406) for EGFR-amplified rGB patients, either as a single agent or in combination with TMZ. The combination therapy demonstrated an improved OS of 19.8% compared to 5.2% in the control group and 10% in the monotherapy group. These findings suggest clinical benefits for the Depatux-M + TMZ combination, particularly in patients relapsing more than 16 weeks after the last TMZ cycle [429]. On the contrary, Depatux-M + TMZ treatment in ndGB patients had negative outcomes in a phase III clinical trial (NCT02573324) [430].
Nimotuzumab is another monoclonal antibody against EGFR that binds more specifically to EGFR overexpressing cells. It showed promising efficacy in a phase II trial for high-grade glioma, but failed to increase survival in a randomized phase III trial in ndGB patients [451]. The heterogeneity of GB tumors, in which EGFR deletion and EGFR amplificated mutations can coexist in different cells, leads to adverse effects arising from collateral inhibition of EGFR in normal tissues, as well as to redundant and alternative compensatory pathways, which represent the most important escape mechanisms that limit the anti-glioma effects of the different EGFR-targeting drugs [536,537,538,539].
Similar to EGFR, the PDGFRs (α and β) are involved in the activation of the PI3K (Figure 3) [540]. PDGFRα is the second most frequently amplified tyrosine kinase receptor in GB behind EGFR, and its expression associates to poor prognosis [541]. Once activated, PDGFR triggers intracellular signaling cascades that regulate cancer cell survival, growth, and progression. Consequently, dysregulation of PDGF signaling stimulates malignant transformation of normal neural stem cells into GB cells and enhances GB cell growth and motility through autocrine signaling [542,543]. No specific PDGFR inhibitor exists; thus, PDGFR is either targeted by specific anti-PDGFR antibodies (IMC-3G3 and MEDI-575) or multi-kinase inhibitors (alone or in combination with other chemotherapeutics). MEDI-575 was well tolerated but showed limited activity in rGB [450]. Pazopanib, sorafenib and sunitinib inhibit both VEGFR and PDGFR but clinical evaluations (Table 3) did not provide any significant benefit in GB [441,452,456,457,458,459,460,522]. Similar results were obtained in other clinical trials with dasatinib [428] and imatinib plus hydroxyurea [544]. ERBB3 (Erb-B2 Receptor Tyrosine Kinase 3), IGF1R, and TGFβR2 seem to be responsible for the resistance to PDGFR inhibitors. Consistent with this notion, a combination of PDGFR inhibitors with inhibitors targeting either ERBB3 or IGF1R more potently suppressed the growth of GB cells than each inhibitor alone. ERBB3 has been suggested as potential prognostic marker and therapeutic target for GB with high PDGFR-α expression [545]. Recently, the PDGFRα/β inhibitor CP-673451 induced differentiation of GSCs and improved the anti-tumor effects of TMZ in vivo using a subcutaneous xenograft mouse model [546].
Targeting angiogenesis has been and continues to be an attractive therapeutic modality in GB. The best-known angiogenesis regulators in GB progression include VEGF, basic fibroblast growth factor (bFGF), PDGF, EGF, TGFβ, MMPs, and angiopoietins [547]. These angiogenic factors are upregulated in GB by a variety of mechanisms including oncogene activation, loss of tumor suppressor gene function, and/or hypoxic microenvironments [243,548]. Loss of the PTEN signaling leads to VEGFR2 expression in GB cells, which may contribute to resistance to anti-angiogenic treatments [549], and is also involved in resistance to TMZ [550]. As was previously mentioned, trials evaluating BEV (Table 1 and Table 3) as monotherapy or in combination with lomustine or TMZ have not improved OS in GB [260,262,387,409,411,412], and lack of discernible benefits was evident in clinical trials where BEV was combined with erlotinib, temsirolimus, sorafenib, or tandutinib in rGB [415,455,464,519]. Despite these negative results, BEV is widely used for the treatment of rGB and, in fact, it is often the control arm in many clinical trials. Regorafenib is an oral multikinase receptor inhibitor that blocks tyrosine kinases active in angiogenesis (VEGFR1–3 and TIE2) and cancer growth (KIT, RET, RAF1, BRAF and BFRAFV600E), growth factors (FGFR and PDGFR), and tumor immunity promoters as the colony-stimulating factor 1 receptor (CSF1R) [370]. Regorafenib has shown superior activity to lomustine in rGB patients with tumors bearing the methylated MGMT promoter. The REGOMA phase II randomized trial (NCT02926222) showed a small mOS benefit of regorafenib (7.4 months) versus lomustine treatment (5.6 months) [454,551]. Age and MGMT methylation appeared to influence OS, thus suggesting that regorafenib treatment could be an alternative in older patients [552]. Regorafenib (phase II/III trial, NCT03970447) has not received the EMA approval, although it is the first-choice treatment for rGB according to Italian Association of Medical Oncology guidelines [553,554]. Most probably, this is due to some questioned results of the clinical trial [555]: (1) low OS in the lomustine group (other clinical trials achieved mOS rates of 8–10 months); (2) the absence of a molecular tumor characterization in 31% of 119 recruited patients; (3) high rate of adverse effects in the regorafenib arm (grade 3–4 adverse events occurred in 56% of patients) in 40% of patients treated with lomustine; and (4) the difference in the two groups of study, i.e., two patients with more favorable prognostics (IDH-mutated GB) were included in the regorafenib group, whereas no IDH mutation was reported in the lomustine group [454]. Multivariate analysis confirmed that MAPK pathway mutations predicted a shorter PFS after regorafenib treatment, while EGFR-altered cases had a better response to regorafenib [556]. Further data from retrospective studies support the efficacy of regorafenib in GB [557], and retrospective analysis on 54 rGB patients treated with regorafenib reports an even longer mOS of 10.2 months [558]. Five trials of regorafenib are ongoing (NCT04810182, NCT04051606) or are being recruited (NCT06095375, NCT03970447, NCT06047379) for the treatment of GB.
Despite the importance of angiogenesis in the growth and acquisition of resistance of GB, none of the anti-angiogenic therapies tested in clinical trials (Table 3), i.e., vandetanib (NCT00821080) [465], cediranib (NCT01062425, NCT00777153, NCT01310855) [421,446,559], dovitinib (NCT01753713) [431], and axitinib (NCT01562197, NCT01290939) [407,560], have managed to improve the benefits obtained of historical controls. Preclinical studies showed that cediranib sensitizes cancer cells to PARP inhibitors by downregulating homology-directed DNA repair [561], but cediranib combined with olaparib (NCT02974621) failed to increase PFS and OS in rGB patients [426]. These results demonstrate that the use of anti-angiogenic agents (either as first- or second-line treatment, and either as a single agent or in combination with chemotherapy), while possibly improving quality of life, does not extend survival in unselected GB patients [230,393,562]. Mechanisms involved in disappointing results of anti-angiogenic chemotherapy in GB include the compensatory switch to alternative angiogenic pathways (i.e., the vasculogenesis mentioned before), GSC transdifferentiation, temporary and dependent treatment dosage and duration until the vasculature returns to normal, the impact of angiogenesis inhibition on tumor distribution of other chemotherapeutic agents [563,564], and upregulation of the hypoxia-inducible protein 2 [244].
Currently, more than 50 PI3K inhibitors have been designed and produced for cancer treatment, but just a few, such as enzastaurin, BKM120, XL147 and XL765, have entered into clinical trials for GB treatment [565,566]. Enzastaurin was well tolerated and had an acceptable hematologic toxicity profile (phase III, NCT00295815), but did not show superior efficacy than lomustine [433]. Currently, buparlisib (BKM120) is the most frequently tested panPI3K inhibitor in clinical trials (Table 3) since it is well tolerated and permeable to the BBB [126]. BKM120 induces G2/M cell cycle arrest and apoptosis in GB cells through microtubule misalignment and mitotic dysfunction in a p53-dependent manner [567], and has shown antiproliferative and proapoptotic properties in various GB cell lines, as well as in xenograft models [568,569,570,571]. However, single-agent BKM120 had minimal response (NCT01339052) in patients with PI3K-activated GB at first or second recurrence. BKM120 had significant brain penetration, and the lack of clinical response was attributed to the incomplete blockade of the PI3K pathway in tumor tissues [417]. Paradoxically, even though BKM120 inhibits Akt phosphorylation, patients harboring PTEN loss and/or PIK3CA mutations were not sensitive to BKM120 treatment [572]. The results of clinical trials (Table 3) in which BKM120 was part of combined treatments with BEV [418], carboplatin, lomustine [419], capmatinib [420] did not show benefits vs. monotherapies.
Akt inhibitors or induction of PTEN expression can reverse the resistance and sensitize cells to chemo- and radiotherapy by impairing DNA repair [573,574]. Akt inhibition induced by perifosine (Figure 3) enhanced the growth inhibition effects of low-dose heavy-ion radiation on GB C6 cells, via proliferation inhibition, apoptosis and oxidative stress [575]. However, perifosine faces certain constraints, such as the restricted ability to penetrate the BBB, which justifies its lack of effectiveness in the clinical trial for rGB (NCT01051557) [453]. The drugs that target Akt, combined with TMZ and fractional radiation, gradually enter the field of vision because chemoradiotherapy should increase the level of phosphorylated Akt [574]. To counteract the impact of Akt, which remains persistently active in GB, an alternative therapeutic strategy involves targeting mTOR, which is hyperactivated in 90% of GBs and is associated with poorer prognosis [505].
The pharmacological inhibition of mTOR inhibits GB growth, induces autophagy and reduces the invasive potential of GSCs [576]. Rapamycin (sirolimus) and its analogs, including everolimus (RAD001), temsirolimus (CCL-779), and ridaforolimus (AP23573), exhibit inhibitory effects on mTORC1 in in vitro and in vivo models and have been extensively clinically studied (Table 3) for GB treatment [442,462,463,492,577,578]. The effectiveness of rapamycin was noticed in terms of antitumor activity during a phase I trial involving patients with rGB [579], but in combination with erlotinib, did not render satisfactory results due to persistent mTORC2 signaling [440,580]. Combining everolimus with conventional SOC leads to increased toxicities and has no survival benefit in ndGB [442]. Surprisingly, these adverse events were not shown when BEV and everolimus were assayed as part of first-line combined modality therapy for GB (NCT00805961). Here, the PFS compared favorably to previous reports with SOC and was similar to results achieved in other trials in which BEV was added to first-line treatment [443]. Third-generation inhibitors of mTOR, (torin1 and 2, vistusertib (AZD2014) and rapalink-1) bivalently target both mTORC1 and mTORC2 [581]. In preclinical models, torin2 effectively inhibited both mTOR pathways in GB, leading to significant suppression of proliferation and migration of GB cells and GSCs [582]. Torin2 displayed a longer half-life, improved water solubility, and better oral bioavailability than torin1. Rapalink-1 targets GSCs and acts synergistically with TTFields to reduce resistance against TMZ [583]. Similarly, vistusertib enhances the radiosensitivity of GSCs in both in vitro and in vivo conditions [584] and, combined with TMZ, shows good safety at the tested dose levels in patients with rGB (phase I, NCT02619864) [585]. The clinical lack of efficacy of mTOR inhibitors can be attributed to the activation of parallel signaling pathways (MAP/ERK) or adaptive and acquired resistance after an initial response [586]. Moreover, mTOR inhibitors act both on tumors and on immune cells; thus, one can hypothesize that the putative anti-tumor efficacy of mTOR inhibitors might be counterbalanced by their suppressive effects on immune cells, thereby building an immunosuppressive environment that facilitates tumor progression [497].
Despite strong preclinical evidence for the therapeutic potential of tyrosine kinase inhibitors targeting the PTEN/PI3K/Akt/mTOR pathway, clinical trials conducted over the past 20 years have not resulted in the desired therapeutic breakthrough for GB [497,503,514,565,587,588]. Brar et al.’s (2022) review focuses on the potential of tyrosine kinase inhibitors in GB therapy and provides a clear insight into the reasons behind the unsuccessful clinical trials in GB, despite the success in treating other cancer types [589].

4.4. RAS/RAF/MEK/ERK

The Ras/RAF/MEK/ERK pathway (or MAPK signaling pathway) is hyperactive in virtually all GBs owing to the overexpression of key regulators like EGFR and PDGFR [546,590]. Activation of the Ras protein occurs through the replacement of GDP by GTP, initiating the activation of MAP kinases. These kinases phosphorylate and activate downstream ERK proteins, which translocate to the nucleus and induce transcriptional pathways, leading to cellular proliferation, survival, and dedifferentiation (Figure 3).
The BRAFV600E missense mutation leads to constitutive activation of the Ras/Raf/MEK/Erk pathway, promoting tumor cell proliferation, survival and inhibition of apoptosis [591,592]. GBs with BRAF mutation differ significantly in location, age of diagnostic, survival rates, and global gene-expression profiles from the rest of GBs [593,594]. Several cases of impressive response to BRAFV600E inhibitors (vemurafenib and dabrafenib) in GB have been reported, including cases of prolonged disease control [595,596,597,598]. The clinical trial (NCT01524978) showed that vemurafenib treatment had a durable antitumor activity in some patients with BRAFV600-mutant gliomas, although efficacy seemed to vary qualitatively by histologic subtype [599].
MEK1/2 are involved in tumor development and apoptosis inhibition, enhancing DNA damage repair capacity and migration/invasion of GB cells. MEK inhibitors (MEK162, trametinib, PD0325901) in GSCs showed antiproliferative and apoptotic cell death and induced neuronal differentiation in sensitive GSCs. Trametinib decreased tumor growth and improved survival in GSC xenografts [600]. Targeting multiple RAS effector pathways with a combination of MEK and mTOR kinase inhibitors is a strategy that has been tested in melanoma and has provided survival benefits in preclinical models of glioma with BRAF mutations or KRAS mutations [601]. At present, concurrent use of trametinib and dabrafenib exhibits notable clinical importance in addressing low-grade gliomas that carry the BRAFV600E mutation [602]. A recent case report presented the case of a young female with BRAFV600E GB who had a prolonged response to targeted therapy with dabrafenib and trametinib [603]. Although the specific BRAFV600E mutation is uncommon in GB patients, dabrafenib in combination with trametinib showed clinically meaningful activity in patients with recurrent or refractory BRAFV600E mutated high/low-grade glioma (phase II, NCT02034110) [432]. The significance of the results is limited because just 31 GB patients were included in the study, but we completely agree with the conclusions of researchers that “genetic testing tumor profiling should be introduced early in the management plan to promptly identify those patients who may be eligible for BRAFV600E-targeted treatment”, which should be extended to all GB studies.

4.5. PARP Inhibitors

Poly(ADP-ribose) polymerase-1 (PARP-1) facilitates the repair of DNA strand breaks. PARP-1 mRNA expression is associated with poor survival in GB patients [604]. Since TMZ induces replicative stress via futile attempts of DNA repair at O6MeG:T mismatches, and PARP inhibitors (PARPis) enhance stress by compromising the stability of stalled replication forks, it seems logical to think that PARPis may restore TMZ sensitivity in GB and GSCs [605,606,607]. In addition, PARP inhibitors synergize with radiation therapy because PARP-1 activity increases 500-fold in the presence of DNA damage [608].
Although several promising PARPis have limited distribution across BBB, some of them reached GB margins in vivo. The brain-to-plasma concentration ratio of veliparib (ABT-888) is substantially higher than other PARPis such as olaparib, rucaparib, or talazoparib. Veliparib restores sensitivity in TMZ-resistant glioma cells and xenografts [609] and extends survival in the MGMT-hypermethylated GB model, but is ineffective in MGMT-unmethylated lines [610]. Unfortunately, veliparib did not increase the efficacy of TMZ in rGB patients (NCT01026493) [467] and had no clinical benefit in ndGB patients with hypermethylated (NCT02152982) [469] or unmethylated-MGMT promoter status (NCT02152982) [468]. The authors support the clinical interest of the last trial, based on the radiosensitizing effects of veliparib [611], and mention “encouraging responses when applied to MGMT-unmethylated cell lines” without citing their sources. Unfortunately, they did not take into consideration the contradictory results previously shown in [610] that are in agreement with the lack of efficacy of the treatment in this trial. More recently, Wu et al.’s (2021) results demonstrated that inhibition of PARylation by PARP inhibitor (talazoparib) reduces MGMT function rendering sensitization to TMZ, providing a rationale for combining PARP inhibitors to sensitize TMZ in MGMT-unmethylated GB [612]. The potential of veliparib in GB can best be demonstrated in patients with PTEN null tumors; therefore, clinical trials with veliparib should evaluate these patients as a separate group [612]. Olaparib combines DNA repair inhibition and impairment of cancer cell respiration as anticancer activities [613]. In vivo, mice treated with TMZ alone or in combination with olaparib showed greater survival than those untreated or with the olaparib monotherapy, as well as a significant decrease in tumor volume; however, there was no significant difference in survival between both groups [614]. Encouragingly, olaparib was found to penetrate brain tumors at radiosensitizing concentrations (NCT01390571). A combination of olaparib and low-dose TMZ was safe and well tolerated (NCT01390571), yielding PFS6 rates [615,616].
PARPi meriting further clinical study include pamiparib and talazoparib (NCT04740190). Compared to olaparib, pamiparib demonstrated improved penetration across the BBB in mice and the strongest capacity to inhibit brain tumor growth in the xenograft model. EGFR-amplified GSCs showed remarkable sensitivity to talazoparib treatment, which significantly suppressed tumor growth in EGFR-amplified subcutaneous models [617]. In the SCLC-derived TMZ-resistant H209 intracranial xenograft model, the combination of pamiparib with TMZ overcomes TMZ resistance and shows significant tumor inhibitory effects and a prolonged life span [618]. These results support the phase I trial (NCT03150862) in which pamiparib is being assayed in combination with SOC in nd/rGB patients [619].
Bisht et al. have recently reviewed several strategies to increase penetration through BBB and reverse the resistance to PARPi [620].

4.6. Other Assayed Strategies

In preclinical studies, statins (well known as hypocholesterolemic drugs) exerted potent anti-GB effects through different mechanisms stemming from the inhibition of the mevalonate cascade, and resulting in the inhibition of proliferation, migration, invasion, and in the induction of apoptosis and autophagy [621,622,623,624,625]. Blockade of the Ras/MEK/ERK and Ras/PI3K/Akt pathways by statins reduces the expression of TGFβ as an angiogenic factor in GB [626]. Synergistic action with other anticancer drugs has also been described [627,628,629]. For instance, atorvastatin augments TMZ’s efficacy in vitro as well as in GB xenografts via prenylation-dependent inhibition of Ras signaling [630]. However, atorvastatin did not improve PFS when was evaluated in combination with radiotherapy and TMZ in GB patients (phase II, NCT02029573) [406].
As tyrosine kinase receptors usually activate similar downstream pathways, in GB upregulation and/or activation of IGF receptors (IGF-1R and IGF-2R), c-Met, and PDGFRβ contribute to resistance to EGFR/EGFRvIII inhibition [505]. Approximately 37% of patients with GB have c-Met overexpression, which plays an important role in promoting invasion and tumor recurrence [631]. Moreover, concomitant c-Met/VEGFR2 overexpression was associated with worse overall survival in GB and is linked to resistance to anti-angiogenic drugs [632]. A phase II study investigated the effect of the monovalent MET inhibitor onartuzumab plus BEV in rGB, but found no evidence of further clinical benefit [413]. Crizotinib in combination with SOC was safe and resulted in a highly promising efficacy for ndGB (phase Ib/ NCT02270034), warranting further investigation [633].
IGF-R1 is overexpressed and is necessary for neoplastic transformation in GB. IGF-1R blockade can inhibit GB growth by different mechanisms, including direct effects on the tumor cells as well as indirect anti-angiogenic effects [634]. High IGF1R expression is associated with chemoresistance to TMZ as well as reduced survival, thus suggesting its possible use as a biomarker [635]. Tyrosine kinase inhibitors, anti-sense oligonucleotides, or monoclonal antibodies (cixutumumab) targeting the IGF-IR have been clinically tested to inhibit IGF signaling in GB, with lack of benefits or inconsistent results [636]. On the contrary, autologous glioma cells treated ex vivo with an antisense oligodeoxynucleotide targeting the IGF-IR, and re-implanted in patients (phase 1b, NCT02507583), seem to increase the OS in ndGB-treated patients [637].
Some natural polyphenols (resveratrol, curcumin, silibinin, quercetin, etc.) have shown antiGB potential and synergize with radio/chemotherapy in vitro. Inhibition of proliferation, migration, cell invasion and angiogenesis are mechanisms proposed to explain the potential effect of polyphenols on reducing GB progression [638,639,640,641,642,643]. Specifically, metformin and resveratrol have been suggested to inhibit GB cell proliferation, invasion and migration by downregulating the PI3K/Akt pathway, activating mTOR, and increasing AMPK phosphorylation [644]. Rutin, epigallocatechin-3-gallate, quercetin and curcumin have shown synergistic effects with TMZ, whereas curcumin enhances the action of etoposide, paclitaxel, cisplatin, camptothecin, and doxorubicin [641,645,646,647]. Nevertheless, most of these proposed mechanisms only have in vitro support. It should be pointed out that, under in vivo conditions, the antitumoral activity of polyphenols is limited due to their short half-life. Even when polyphenols are administered at high doses, their rapid metabolism precludes reaching efficacious anti-tumor concentrations in a growing cancer. In practice, after an oral dose as high as 100 mg/kg, the peak plasma level of resveratrol was 11 ± 4 μM at 10 min, pterostilbene was 25 ± 6 μM at 10 min, EGCG was 5 ± 2 μM at 30 min, curcumin was 27 ± 6 μM at 15 min, quercetine was 8 ± 2 μM at 15 min, and genistein was 17 ± 5 μM at 10 min. Plasma levels of all these PFs were <1 μM at 60 min [648]. Exposure of many different cancer cells (i.e., U87 and LN229 GB cells) to these concentrations for just 1 h does not affect their growth and viability. Our results are supported by Beylerli et al. who recently pointed out that, although potential beneficial effects exerted by polyphenols are promising, their efficacy in vivo is strongly limited by their bioavailability and BBB permeability [646]. Zanotto-Filho et al. showed that resveratrol, a BBB-permeable drug, improved TMZ/curcumin efficacy in brain-implanted tumors in rats [647]. In these experiments, 50 mg curcumin/kg × day and 10 mg resveratrol/kg × day were administered i.p., but it is uncertain at which concentration each polyphenol reached the tumor and for how long. Based on the above discussion, it is easy to deduce that the tumor levels of in vivo administered polyphenols will be very low and acting for a short period of time. Facts that raise many doubts about the real mechanisms involved in the anti-tumor effects. The effect of curcumin has also been assayed using its intratumoral injection [649]. In this case, curcumin (100 mg/kg) inhibited U87 xenografts growth by approx. 50%. However, all the mechanisms proposed for curcumin were studied under in vitro conditions and using unreliable conditions compared to the in vivo setting. Methods to improve polyphenol absorption, pharmacokinetics, and efficacy [650,651,652] are key in order to deliver therapeutic concentrations at specific target sites and to increase the antitumoral bioefficacy of polyphenols in vivo.

5. Immunotherapies

Recent studies show the presence of a variety of immune cell types within the GB TME with a dominance of immunosuppressive cells, i.e., MDSCs, microglia, M2 macrophages, FoxP3+ regulatory T cells (Tregs), and antigen-presenting cells (APCs) (including DCs and bone-marrow-derived macrophages). The presence of M2 macrophages is linked to an increased GB aggressiveness and plays a pivotal role in the acquisition of chemo and radioresistance of GB cells [653,654]. In addition, frequently, CD4+ and CD8+ T cells are functionally deficient, inactivated, or exhausted, often co-expressing immune checkpoint molecules, i.e., programmed cell death receptor 1 (PD-1), lymphocyte activation gene 3 (LAG3) and T cell immunoglobulin mucin 3 (TIM-3) [655]. GB cells secrete immunosuppressive factors such as TGFβ2, PGE-2, IL-1, IL-10 (check Section 2.1) and indoleamine 2,3-dioxygenase (IDO), which work cooperatively to suppress the activity of effector cells and to evading the anti-tumor immune response [516,656]. As described here below, a plethora of novel immunotherapies, i.e., checkpoint inhibitors (ICIs), vaccines, T-cell-based immunotherapies, NK-cell-based therapies, viral therapies, and combined treatments, have been attempted in order to control GB expansion and/or recurrence.

5.1. Checkpoint Inhibitors (ICIs)

Cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death 1 (PD-1), also known as checkpoints, are co-inhibitory receptors expressed on the T cells surface to promote immunotolerance to self-antigens. GBs (and other cancer cells) overexpress programmed cell death ligand 1 (PD-L1) which, upon interaction with PD-1, inhibits T cell proliferation and the T cell receptor (TCR)-dependent IL-2 production and suppresses the CD4+ and CD8+ response [657]. Thus, PD-L1 overexpression triggers T cell exhaustion and leads to the immunosuppressive TME that promotes GB progression and correlates with patients’ worse outcomes [658]. Pro-inflammatory molecules (i.e., IFNs, IL-12 or TGFβ) and several pro-oncogenic transcription factors (HIF-1α, STAT3, EGFR or PTEN loss) have been identified as direct regulators of PD-L1 transcription [391,659].
Monoclonal antibodies targeting CTLA-4 (ipilimumab), PD-1 (pembrolizumab, nivolumab, and cemiplimab) or its associated programmed cell death ligand 1 (PD-L1, atezolimumab, durvalumab and avelumab), known as checkpoint inhibitors (ICIs), have been approved by the FDA for the treatment of various types of cancer [660]. Blockage of the interaction between PD-1 and PD-L1 may re-establish proper immunity against GB. Preclinical studies have confirmed this hypothesis and demonstrated a survival benefit in immunocompetent murine models of GB following dual radiation and anti-PD-1 treatment [661]. The results of the phase II/III trials testing ICIs on GB are presented in Table 4. None of them showed significant increases in mOS. Pembrolizumab was ineffective as monotherapy and combined with BEV for rGB [662]. CheckMate 143 (NCT02017717) compared nivolumab with BEV therapy in rGB patients. Although the follow-up of 12 months did not find significant differences between the two arms (42%), the objective response rate was significant higher with BEV (23.1%) vs. nivolumab (7.8%), disfavoring the experimental drug [663]. The addition of nivolumab to the SOC has been assayed in two clinical trials for ndGB patients with or without hypermethylated MGMT promoters (NCT02617589 and NCT02667587) without evidence of clinical benefits [664,665].
Two clinical studies have demonstrated enhanced T cell responses and a clinical benefit with neoadjuvant ICI treatment versus anti-PD-1 adjuvant administration in patients with rGB [693]. Patients in the neoadjuvant arm received pembrolizumab 14 ± 5 days prior to surgical resection and patients in the adjuvant arm did not. After surgical resection, both groups received pembrolizumab every 3 weeks. Compared to only adjuvant PD-1 blockade, neoadjuvant pembrolizumab confers significant improvement in OS (228.5 days versus 417 days) and PFS (2.4 months versus 3.3 months) [693]. A single-arm phase II clinical trial, utilizing neoadjuvant nivolumab in GB patients, demonstrated similar intratumoral and systemic immune changes (NCT02550249), even though survival advantages could not be demonstrated [683]. Although the pros and cons of starting neoadjuvant therapy are hotly contested in clinical trials [694], preclinical studies indicate the remarkable benefit of starting immunotherapy before surgery [695], which is also in agreement with other clinical assays evaluating the efficacy of nivolumab and neoadjuvant PD-1 blockade in melanoma, breast and lung cancers [696,697]. No improvement in PFS or OS (ISRCTN84434175) was observed with the addition of ipilimumab to SOC in ndGB patients
Co-suppression of CTLA-4 and PD-1 inhibited GB progression in mice models [698,699]. However, the anti-CTLA-4/PD-L1 treatment combination produced grade III-IV toxicities in GB patients, thus drawing into question its safety [700]. TTFields upregulate different immune checkpoints (e.g., PD-L1, CTLA-4, or TIGIT, T cell immunoglobulin and ITIM domain), which supports the possibility that combined treatment may increase the efficacy of immunotherapy [322,323,324]. In that sense, two phase II trials are evaluating possible synergistic effects combining SOC with TTFields plus pembrolizumab (NCT03405792), and nivolumab plus/minus ipilimumab (NCT03430791) in ndGB. The preliminary results of NCT03405792 seem promising because of evidence of an improved OS (24.8 vs. 14.7 months) in ndGB patients that received the complete cocktail treatment vs. controls [684].
A recent meta-analysis concludes that a postoperative combination of radiotherapy, chemotherapy and PD-1 inhibitors may increase antigen cross-presentation activity, promote tumor-lymphocyte infiltration, and increase the expansion of effector T cells. Furthermore, radiotherapy has a synergistic effect on immunotherapy by decreasing the number of tumor cells and upregulating the tumor expression of PD-L1, but evidence of survival benefits is scarce [701]. The limited efficacy of ICIs can be attributed to compensation of blocked PD-1 function by other checkpoint molecules (i.e., LAG-3) [702]; TAMs, which have been observed to remove anti-PD1 antibodies bound to CD8+ T cells [703]; and impaired epigenetics and memory cell formation, resulting in T cell exhaustion [704]. LAG-3 (also known as CD223) is expressed on activated human T (often co-expressed with PD-1), DCs and NK cells [705]. Its binding with the major histocompatibility complex (MHC)-II in APCs promotes apoptosis, decreases proliferation and increases T cell tolerance [705]. The combination of anti-LAG-3 and anti-PD-1 in a variety of tumor models has led to synergistic antitumor efficacy. Harris-Bookman et al. showed that LAG-3 is expressed in human and mouse GB samples, and either knockdown or anti-LAG-3 antibodies improved survival in a preclinical GB model and the efficacy of anti-PD-1 treatment [706]. Currently, a phase I trial (NCT02658981) is underway, testing anti-LAG-3 alone and in combination with anti-PD1 in rGB patients [707]. Ongoing research is exploring the potential of using ICIs in combination with other immunotherapies or targeted therapies to further enhance their effectiveness in treating GB.

5.2. Other Strategies to Avoid Immunosuppression in the GB TME

IDO1, IDO2 and tryptophan 2,3-dioxygenase (TDO2) are “rate-limiting” enzymes in the kynurenine pathway of Trp catabolism, [708,709,710]. The resulting Trp metabolites are involved in immune regulation, energy metabolism and the production of NAD+. Expression of IDO and TDO enzymes in mammals differs by tissue location and by their stimuli-dependent induction [711,712]. They are constitutively expressed in a restricted set of tissues, including placenta, mucosa, and lymphoid organs [713]. IDO1 expression is increased in many human tissues, and it is overexpressed in the majority of cancers (90% of glioma cells) by proinflammatory cytokines, such as IFN-γ and TNF-α, or in response to interaction with tumor-infiltrating T or NK cells [714,715]. IDO1 overexpression in glioma cells impedes an effective immune response through increased apoptosis of CD8+ T cells and by converting naïve T cells into inducible immunosuppressive Tregs [716]. TDO is primarily expressed in the liver and induced by Trp and corticosteroids [717]. Human IDO2 is a relatively inefficient Trp dioxygenase, and little is known about its link to cancer, although different pro-inflammatory stimuli induce its expression in melanoma, pancreatic, gastric, and brain cancers [712]. TDO and IDO2 show higher tissue specificity and much lower enzyme activity than IDO1 [718]. A multitude of cancer cells constitutively express or upregulate IDO1, TDO2, or both, and coerce stromal and tumor-infiltrating immune cells to express IDO1, thus supporting evasion of immunosurveillance [710,719,720]. Increased expression and a high kynurenine/Trp ratio in the peripheral blood have been associated with cancer progression, poor prognosis and lower survival in GB and other cancer patients [721].
Although GB cells typically do not express IDO, it is activated when GB cells are identified by TILs or NK cells and exposed to important anti-cancer cytokines such as IFN-γ and TNF-α. Moreover, IDO1 expression is elevated in GSCs compared to GB cells [722,723]. IDO1 overexpression increases the intratumoral accumulation of immunosuppressive Tregs and decreases OS in experimental mice with brain tumors [708,716,724,725,726]. Compared to healthy controls, GB patients showed decreased serum Trp levels and, surprisingly, reduced Trp metabolite levels. High tumor volume is associated with low systemic metabolite levels, low systemic kynurenine levels and worse OS [727]. Nevertheless, real-time measurements of IDO1 activity in GB patients is still a follow-up strategy = pending development.
Cancer cells require Trp to produce energy, and when its availability is low, they acquire motility in order to search for alternative sources of energy. Trp deprivation inhibits proliferation and induces apoptosis in DCs and T cells through the activation of the serine/threonine-protein kinase GCN-2 (general control non-derepressible 2) [728,729]. GCN-2 activation also inhibits fatty acid production in naïve T cells, which is necessary for their proliferation and activity [730]. However, in vivo, Trp levels show very low fluctuations in the brain [731], and recent studies have questioned the immunosuppression attributed to local Trp starvation [732], thus suggesting that accumulation of kynurenines-associated aryl hydrocarbon receptor (AHR) activation could be the main mechanism involved in the IDO- or TDO2-induced immunosuppression [710,733,734]. In fact, AHR expression is associated with immunosuppression in human tumors and AHR blockade reverses the IDO/TDO-mediated immunosuppression [720]. AHR activation by Trp catabolites promotes TGFβ production in GB [735], generates immune-tolerant Treg cells and DCs, and suppresses anti-tumor immunity [736]. AHR promotes TGFβ production in GB [735], induces immune checkpoints such as PD-L1 [737], and induces immunosuppression via its effects on T cells [738], DCs [739], macrophages [739], B-cells [740], or GB-specific TAMs [741], thereby contributing to the generation of an immunosuppressive milieu [742]. Moreover, Trp metabolites like quinolinic acid, 3-hydoxyanthranilic acid, and 3-hydroxykynurenine have been shown to induce apoptosis in Th1 helper cells, CD8+ effector T cells, and B cells, while sparing immunosuppressive Th2 helper cells [743]. Dysregulation of kynurenine signaling promotes DNA damage, is associated with brain edema formation (which facilitates invasion and motility of cancer cells) and induces NAD+ generation in glioma cells (a mechanism involved in the development of resistance to therapy) [744]. Interestingly, cancer immune suppression was reversed by administration of PEGylated kynureninase that degrades kynurenine into nontoxic and immunologically inert metabolites [745].
Proteolysis targeting chimera (PROTAC) technology has been used in the development of molecules capable of targeting IDO1 [746], and to date, several IDO1 inhibitors (PF-06840003, epacadostat, indoximod, navoximod, BGB-5777, 1-methyl-tryptophan, BMS-986205, GDC-0919) are under clinical trials for GB therapy [724,746,747]. Although the blockade of IDO as monotherapy has not shown any effect on OS in GB patients, IDO inhibition can enhance the response to immunotherapy. In a murine preclinical model of GB, combined treatment with anti-IDO1, anti-CTLA-4, and anti-PD-1 showed synergistic effects, increasing the antitumor effect compared to monotherapies [748]. The combination of an IDO1 inhibitor (BGB-5777 or 1-methyl-tryptophan), PD-1 blockade and irradiation significantly increased OS and resulted in long-term tumor control in 30–40% of mice with advanced GB [749,750]. Furthermore, radiotherapy response can be enhanced by GDC-0919 by reducing radiotherapy-induced immunosuppression [129], whereas IDO inhibitors have been shown to synergize with TMZ and radiation therapy [751]. In mice, PF-06840003 reduced intra-tumoral kynurenic levels and inhibited tumor growth in both monotherapy and, with increased efficacy, in combination with anti-PDL-1 antibodies [752]. Supported by these preclinical data, a phase I multicenter clinical trial (NCT02764151) showed that PF-06840003 was generally well tolerated, had a pharmacodynamic effect and had a durable clinical benefit in a subset of glioma patients [753]. In mice, epacadostat generated a potent immune response by altering the abnormal signaling pathways of cancer cells and reducing kynurenine levels by 90% in both plasma and the tumor tissue [754], and also enhanced the efficacy of TMZ and ICIs improving the OS [755]. Epacadostat combined with anti-PD-1 antibodies (pembrolizumab or nivolumab) improved the response rate and PFS (>6 months) in patients with gastric cancer [756], but in GB patients, non-significant improvements in OS were observed (NCT02327078). Preliminary results of a phase II study (NCT03532295) in rGB with retifanlimab (PD-1 inhibitor) plus or minus epacadostat in combination with BEV and hypofractionated radiotherapy suggests that treatment is well tolerated and had encouraging OS and PFS at the time of data cutoff [685].
Interestingly, advanced age is associated with an increase in brain IDO1 expression, and this is not reversed by treatment with IDO1 inhibitors [757], which can contribute to explaining the decreased survival of older GB patients during treatment with immune checkpoint blockade therapy.

5.3. Vaccines

Vaccines targeting cancer have two types of antigens: tumor-associated antigens present on health tissues and overexpressed in cancer cells, and tumor-specific antigens (TSA, earlier described as “neo-antigens”) present only on cancer cells. Obviously, vaccines based on TSA antigens are more selective and effective, but in highly heterogeneous tumors such as GB, it is difficult to find them. The lack of specificity and high expression of epitopes in GB lead to autoimmunity and side effects such as brain inflammation, which limits the tolerability of vaccine-based strategies [758,759]. Nevertheless, here, we discuss the results obtained with the development of cellular (tumor cell and DC) and non-cellular (peptide) vaccines.

5.3.1. Tumor Cell Vaccines

Early vaccines utilized dead or inactivated tumor cells, with very limited success. To improve the efficacy, gene editing of tumor cells began in the late 1980s, involving the expression of certain immune-stimulating cytokines, granulocyte macrophage colony-stimulating factor (GM-CSF) being one of the most studied [760]. Phase I trials were completed using the latest generation of autologous and allogeneic tumor cell lines secreting GM-CSF (e.g., K-562, lymphoblast cells isolated from the bone marrow of a 53-year-old chronic myelogenous leukemia patient). The effectiveness of vaccination depends on T cell activation and anti-tumor immunity [761]. Additionally, research into the direct injection of formalin-fixed GB as an antigen for treating GB has been studied. A phase I/IIa trial of fractionated radiotherapy, TMZ and autologous formalin-fixed tumor vaccine (AFT) showed a favorable PFS and OS in ndGB patients [675]. IGV-001 consists of autologous GB cells that are incubated with an antisense oligodeoxynucleotide (IMV-001) targeting IGF-1R that is implanted in patients’ abdominal for approximately 48 h, as opposed to other autologous cancer vaccine modalities, which require multiple dosages over weeks. The results are promising because ndGB patients had a PFS of 9.8 months compared to 6.5 months in historical controls receiving SOC [762]. There is a phase IIb trial (NCT04485949) opened for accrual for ndGB patients.

5.3.2. Dendritic Cell Vaccines (DCV)

DC vaccination has gathered considerable attention after some encouraging reports showing acceptable efficacy and safety levels. DCs are powerful APCs capable of inducing acquired and innate immunity responses. DC vaccine preparation involves the isolation of CD-14 positive monocytes from the patient, loading of tumor antigens into the immature DCs, treatment of the DCs with the cytokines (GM-CSF and IL-4) to induce maturity, and finally the preparation of human DC vaccines for re-injection into GB patients [763,764,765].
In a phase I/II prospective non-controlled clinical trial, 37 patients harboring GB or grade 4 astrocytoma received monthly intradermal injections of allogenic DC vaccine, starting at the time of first recurrence after surgery. Compared with patients in the Genomic Data Commons data bank, OS for vaccinated GB patients was 27.6 ± 2.4 months (vs. 16.3 ± 0.7), and it was 59.5 ± 15.9 for vaccinated astrocytoma grade 4 patients (vs. 19.8 ± 2.5). Seven vaccinated patients (two IDH-1-mutated and five wild-type) remained alive at the time of the Lepski et al. report [666].
Results of phase II/III trials on DCV in GB are presented in Table 4. ICT-107 is an autologous DC vaccine targeting six tumor antigens (MAGE-1, HER-2, AIM-2, TRP-2, gp100, IL-13Rα2) specifically overexpressed in glioma stem cells. Patients receiving ICT-107 (NCT01280552, Table 4) reported no significant increases in mOS (17 vs. 15 months) [681]. Yao et al. assayed a DCV loaded with GSC antigens, and results seem to evidence a slight increase in mOS (13.7 vs. 10.7 months) when compared with the placebo control [672]. Northwest Biotherapeutics developed the lysate-loaded dendritic cell vaccine (DCVax®-L) as an adjunct for the treatment of GB [766]. Recently, Liau et al. reported impressive results of nonrandomized phase III clinical trial (NCT00045968) ndGB and rGB patients treated intradermally with DCVax®-L plus SOC vs. contemporaneous matched external control patients treated with SOC [671]. The mOS of the 232 ndGB patients treated with DCVax®-L was 19.3 vs. 16.5 months in controls. Survival at 48 months was 15.5% vs. 9.9%, and at 60 months 13.0% vs. 5.7% in the treated and control arms, respectively. For 64 patients with rGB, mOS was 13.2 months from relapse in the DCVax®-L group vs. 7.8 months in the external control cohort [671]. Despite these promising results, the study design received criticisms [767,768], e.g., the criteria adopted for recruiting GB patients did not consider the latest WHO classifications [253] and there was a lack of evaluation of IDH mutations. Consequently, it is possible that treated patients with longer survivals might have less aggressive GBs [767,768]. In fact, other randomized phase II clinical trials (Table 4, NCT01280552, NCT01213407, NCT03400917) have not shown similar survival benefits [667,669,681]. Compared to the peptide vaccine, the DC vaccine ensures high antigen-presenting efficiency with sufficient exogenous costimulatory signals, whereas DC maturation may be impeded by insufficient costimulatory signals in patients with a weak primary immune status. This can be one explanation for such different results in clinical trials.
Cytomegalovirus is a herpes virus with a high detection rate in many primary or metastatic brain tumors, including GB, whereas it is not detected in surrounding normal brain tissue [769]. Cytomegalovirus has oncogenic features, and its presence in gliomas is linked to the increased production of PGE2, IL-10 and B7-H1. PGE2 activates cell proliferation and angiogenesis, inhibits apoptosis, and activates invasion, and promotes the formation of the TME [770]. IL-10 recruits M2 macrophages that contribute to TME immunosuppression and B7-H1 enhances tumor stem cell migration [771]. Many studies have suggested that anti-cytomegalovirus therapy can restrain glioma progression, and for this purpose, valganciclovir (inhibits viral DNA duplication) [772,773], dendritic cell vaccine and adoptive cytomegalovirus-specific T cell therapy (NCT02661282) [774] have been attempted in GB treatment [770], with promising results. Valganciclovir treatment is well tolerated and, in combination with SOC, significantly increased ndGB patient survival (24.1 months vs. 13.1 months in patients with shorter treatment duration and 13.7 months in the control group) [775], and survival of rGB patients with unmethylated and methylated MGMT promoter genes [776]. Cytomegalovirus-DC vaccines usually target the pp65 antigen. After the radio/chemotherapeutic treatment for ndGB, this vaccine was combined with dose-intensive TMZ and GM-CSF, resulting in a significant increase in survival (Table 4) [670]. A second trial (NCT00639639) compared the effects of tetanus toxoid preconditioning and a DC vaccine with standard TMZ, and resulted in a 36% survival rate at 5 years from diagnosis [777,778]. According to the results of these trials, nearly one-third of the GB patient population receiving cytomegalovirus-specific DC vaccines showed exceptional long-term survival. However, these studies have some limitations: a) consolidative results for the presence of cytomegalovirus within GB are needed; b) the onco-modulatory role of cytomegalovirus for gliomagenesis is not confirmed; and c) it is necessary to confirm positive clinical outcomes in larger and randomized studies [779].
The addition of tumor lysate-pulsed autologous DCs vaccination to tumor resection combined radio-chemotherapy was feasible and safe according to a recent phase II trial (NCT01006044). An increase in tumor-specific immune response was shown in 11/27 evaluated patients, but no correlation between immune response and survival was found [668]. No increase in OS was shown. A new phase II randomized clinical trial (NCT03395587) is now ongoing for ndGB patients [780].
A recent systematic meta-analysis review concludes that adjuvant DC vaccine is feasible, overall toxicity is limited and anti-tumoral cytotoxic responses have been demonstrated. Regimens including DC vaccines led to a significantly longer 1-year and 2-year OS in GB patients after the induction of an immune response (which is not always observed) [781], thus suggesting the need for more time to achieve an anti-GB immune response. The results of these trials robustly support the continued investigation and development of DCV as a treatment for GB.

5.3.3. Peptide Vaccines

Protein/peptide variations derived from mutated genes are distinct to tumor cells and absent in normal cells. As a result, they can serve as TSA to induce immune responses against tumor cells. While only a few mutations are transformed into new epitopes, their presentation by APCs on the human leukocyte antigen (HLA) can lead to T-cell-based immunity. However, many potential tumor antigens do not originate from mutations but arise due to errors or overexpression of normal proteins also present in other tissues. Targeting such antigens may lead to autoimmunity, resulting in unintended effects like brain inflammation [782]. The lack of specificity and the high expression of epitopes in GB pose challenges in the development of peptide vaccine-based strategies. Vaccine adjuvants refer to immunostimulatory components, given in addition to the antigen, to enhance the durability and magnitude of the immune response.
IMA950 is an example of this type of vaccine, containing 11 tumor-associated peptides designed to activate specific T cells. It consists of nine HLA-A2-restricted peptides derived from the surface of GB tumor samples and two HLA-isotype DR-binding peptides that facilitate the CD4+ T cell response. One advantage of using pre-set antigens in the vaccine is that it opens up the possibility of neoadjuvant usage, allowing treatment before tumor samples (through biopsy or surgical resection) become available. To induce the CD8+ T cell response, the vaccine is combined with polyinosinic-polycytidylic acid stabilized with polylysine and carboxymethylcellulose (poly-ICLC), an immunogenic molecule (NCT01920191). In this trial, a multipeptide CD8+ and sustained T helper-1 CD4+ T cell response was observed. For the entire cohort, CD8+ T cell responses to single or multiple peptides were found in 63.2% and 36.8% of patients, respectively. OS was 19 months for GB patients [783]. A randomized phase I/II trial called IMA950-106 (NCT03665545) for rGB is assessing IMA950/Poly-ICLC in combination with pembrolizumab.
Liu et al. characterized the immunogenomic landscape of the aggressive orthotopic GB CT2A model and tested the efficacy of a CT2A-specific neoantigen vaccine combined with an anti-PD-L1 blockade combination [784]. In mice bearing CT2A tumors, the neoantigen vaccine alone did not affect median survival, while immune checkpoint blockade alone only slightly prolonged median survival from 17.5 to 25 days. However, the combination resulted in a 60% long-term survival, thus suggesting that the immune checkpoint blockade facilitates the clonal expansion and activity of neoantigen-specific T cells [784].
In the GAPVAC trial, 16 ndGB patients received two synthesized vaccines: one targeting neoantigens (APVAC2) and the other unmutated peptides (APVAC1), both applied independently by intradermal injections during TMZ therapy. Both vaccines elicited CD8+ and CD4+ T cell responses, inducing 84.7% (APVAC2) and 50% (APVAC1) immunogenicity, and mPFS and OS were encouraging [785]. SurVaxM is a peptide vaccine that is designed to activate the immune system against survivin, which is highly expressed by GB cells. The addition of SurVaxM to the SOC (NCT02455557) appeared to be safe and well tolerated and seemed to have clinical benefits in both methylated and unmethylated ndGB patients [689].
Heat shock protein (HSP) binds TSAs. HSP–peptide complexes can be taken up by antigen-presenting cells and then trigger both innate and adaptive antitumor responses [786]. Therefore, after a simple purification of HSP–peptide complexes from a patient’s tumor, these complexes can be directly administered as a personalized polyvalent antitumor vaccine. HSP peptide complex-96 (HSPPC-96) is the most widely used to treat gliomas [787]. A trial in rGB patients tested this hypothesis and showed an increase in the mOS, a specific peripheral immune response to the peptides bound to HSPPC-96, a focal cellular infiltrate of CD4+ and CD8+ and, in addition, brain biopsies were consistent with specific immune responses at the tumor site [678,679]. A following phase II trial (NCT01814813) failed to demonstrate a survival benefit for patients treated with HSPPC-96 alone or in combination with BEV compared to BEV alone [680]. HSPPC-96 may improve survival for ndGB patients when combined with SOC (NCT02122822) and warrants further study [788].
Up to now, therapeutic vaccines have generally yielded limited efficacy, despite their theoretical basis [360,789]. Combining diverse antigens, including tumor-associated antigens, neoantigens, and pathogen-derived antigens, and optimizing vaccine design or vaccination strategy may help with clinical efficacy improvement [790,791].

5.4. CAR T-Cell Therapy

CAR-T cells are genetically modified T cells (derived from T cells of a patient’s own blood) that express an extracellular TSA recognition domain (CAR, chimeric antigen receptor) and an intracellular activation domain to keep T cells constitutively active. The union between a CAR-T cell and a tumor cell triggers a strong cytotoxic response with the additional release of different proinflammatory cytokines serving as a defense against the immunosuppressive TME [792,793]. Recent studies show that multiple intratumoral (or intraventricular) infusions enhance the persistence of CAR-T cells, their cytotoxic efficacy and the durability of tumor control [794]. Moreover, CAR-T cells can cross the BBB, providing the additional advantage that can be parenterally administered. In a preclinical model of orthotopic GB, a complete antitumor response was achieved when an i.v. dose of anti-GD2 CAR-T cells was administered after focal irradiation. Intravital microscopy images successfully visualized the localization of CAR-T cells and confirmed apoptosis of the tumor cells. According to the authors, radiotherapy allows rapid extravasation and intratumorally expansion of CAR-T cells, leading to a more aggressive and durable immune response [795]. Interestingly, the cytotoxic activity of CAR-T cells is unaffected by TTFields, which provides possible compatibility between both treatments [322].
Six CAR-T cell products have been approved by the FDA for 12 different indications, but none include the GB [796]. Several clinical trials are examining the efficacy of CAR-T cell treatment for GB, being the most tested targets HER2, EGFRvIII, IL-13Rα2, and CD70 [794,797]. The expression of human HER2 plays a crucial role in cell proliferation and motility in cancer cells, and its overexpression in GB is usually associated with an unfavorable prognosis [798]. One potential strategy to optimize the persistence of adoptively transferred T cells relies on the expression of CARs in virus-specific T cells (VSTs) [799]. HER2-CAR–VSTs cells persisted after infusion for up to 12 months in 17 HER2-expressing GB patients. HER2-CAR VST treatment was feasible and safe, and induced transient tumor reduction and/or tumor necrosis effects that resulted in a clinical benefit in 8 of 17 treated patients [800]. Despite this encouraging result, and considering the expression of HER2 in some important organs, the safety of HER2-directed drugs still needs to follow more strict experiments before it is widely used in clinics [801].
IL13Rα2 is rarely expressed in healthy brain tissue and is overexpressed in up to 50–80% of GBs, being associated with poor prognosis [802,803]. Based on preclinical promising results in murine models, a pilot study assayed the efficacy of IL-13Rα2-directed CD8+ cytotoxic T cells (NCT00730613) following tumor resection in three rGB patients [804]. The results of the study demonstrate the feasibility of repetitive intracranial delivery of autologous CART cells via an implanted reservoir/catheter system, showing no serious adverse events. Despite transient regression of the tumor, along with corresponding increases in cytokines and immune cell levels in the cerebrospinal fluid, patients died, most likely due to downregulation of the target antigen IL13Rα2 [804]. One of the patients initially showed a complete remission response after infusion, but had a recurrence 7.5 months later [805].
GD2 is a disialoganglioside overexpressed on tumors of neuroectodermal origin, and especially in GSCs [806]. Animal studies have shown that CAR-T cells against GD2 can effectively eliminate GD2-positive human GB tumors implanted orthotopically in mice without obvious neurotoxicity or off-target effects [807]. GD2-specific fourth-generation chimeric antigen receptor 4SCAR-T cells have been evaluated in a phase I clinical trial assay (NCT03170141) and both single and combined infusions of GD2-specific 4SCAR-T cells were safe and well tolerated in GB patients, with no severe adverse events [805]. More recently, Liu et al. developed a novel combined immunotherapeutic using genetically engineered PBMC-derived induced neural stem cells (iNSCs) expressing HSV-TK and second-generation CAR-NK cells against GB. PBMC-derived iNSCsTK possessed tumor-tropism migration and exhibited considerable anti-tumor activity in the presence of ganciclovir. In addition, combined with GD2NK92, the therapeutic efficacy of iNSCsTK improved the tumor-bearing animal model’s median survival [808].
CARs require higher antigen density for full T cell activation. Hence, the main mechanisms involved in the disease relapse after CAR-T therapy include downregulation of target antigen in response to therapy, inadequate T cell potency, intratumoral heterogeneity, incomplete antigen coverage, upregulation of suppressive ligands (Fas, PD-L1, and other checkpoint molecules), downregulation of anti-inflammatory cytokines (IL-10 and TGF-β), an immunosuppressive tumor microenvironment, and secondary effects of palliative treatment with corticosteroids [809,810,811,812]. CAR T-cell therapy is relatively safe but is not exempt from complications. Neurotoxicity is not an infrequent, and potentially fatal complication. The spectrum of manifestations ranges from delirium and language dysfunction to seizures, coma, and fulminant cerebral edema [813]. Toxicity is related to the location of the tumor and may be reversible with intensive supportive care [814].
The expression of inhibitory immune checkpoints, e.g., PD-1, on CAR-T cells has been associated with a remarkable decrease in their ability to target tumor cells. In this regard, two approaches have been proposed to repress PD-1 expression, i.e., co-administration of anti-PD-1 monoclonal antibodies and CAR-T cells, and PD-1 gene editing in the CAR-T cells [815,816,817]. Work is underway to combine CAR-T cell therapies with immunomodulators designed to maintain the activity of immune cells or to resist specific immunosuppressive mediators. Currently, therapies are being developed using CAR-T cells and multiple antigens (such as B7-H3, CD147, GD2 or MMP2) and a new approach is studying the efficacy of combining EGFR CAR-T with pembrolizumab in rGB [818].

5.5. CAR NK-Cell Therapy

CAR NK-cell therapy offers a different approach to targeting cancer. Firstly, NK cells can recognize tumors composed of heterogeneous cells by employing multiple activating and inhibitory receptors, even when MHC class I (MHC-I) molecules are diminished or absent [819]. Secondly, NK cells play a crucial role in attracting conventional type 1 DC and subsequently CD8+ T cells [820,821]. These functions facilitate the activation of the cancer immunity cycle, which offers the advantage of overcoming the immunosuppressive GB tumor microenvironment [822].
NK cells play a key role in eradicating tumor cells that reduce their surface expression of MHC-I. However, the interaction between MHC-I and killer cell immunoglobulin-like receptors can inhibit NK cell function, leading to reduced destruction of healthy cells [823,824]. Due to these advantageous properties, numerous preclinical and clinical trials have explored the effectiveness of CAR NK-cell-based immunotherapy against GB, utilizing NK cells as either autologous or allogeneic therapy [825]. Allogeneic NK cells offer potential benefits for immune-suppressed patients as they exhibit greater anti-tumor capabilities against GB when compared to exhausted NK cells derived from patients’ peripheral blood mononuclear cells [826]. Cord blood-derived mononuclear cells also hold promise as a valuable source for allogeneic NK cell therapy.
Different approaches utilizing CAR NK-cell therapy include adoptive NK cell therapy (autologous and allogeneic therapy which implies obtaining the patient’s cells and tissues, expanding them ex vivo, and re-infusing them back into the patient), CAR-NK cell therapy (using NK cells engineered to express activating CARs), checkpoint blockade therapy (e.g., combination therapy of NK cells activated by IL-2 and TDK derived from HSP70, and anti-PD1 antibody), and gene editing NK therapy (e.g., the CRISPR/Cas9 genetic-editing system widely utilized to genome-edit T cells to disrupt inhibitory genes such as PD1 and CTLA4) (see [825] for a recent review). CAR-NK cells have advantages in safety profile compared with CAR-T cells, including reduced risk of graft-versus-host disease, cytokine release syndrome, and neurotoxicity [827]. Based on the above considerations and taking into account the inter- and intratumoral heterogeneity of GB, along with its immunosuppressive microenvironment, CAR NK-cell-based immunotherapy appears a suitable approach for addressing GB. In this, it is remarkable the pivotal role of NK cells in inducing anti-tumor responses in CD8+ T cells and other immune cells.

5.6. Multiple Immunotherapeutic Approaches against EGFR

EGFR is also a prevalent molecular target in immunotherapy assays. Rindopepimut (also known as CDX-110) is a small peptide that mimics the mutated region of EGFRvIII and was designed to generate humoral and cytotoxic T cell responses against GB cells expressing this mutation [758]. The combination of rindopepimut with SOC achieved 24-month OS (phase II, NCT00458601), with 50% of patients developing a humoral immune response to the vaccine. Moreover, 82% of the treated patients showed a loss of EGFRvIII expression in tumor recurrence, indicating that the vaccine may be effective in targeting this variant [687]. However, the phase III study (NCT01480479) was interrupted because it did not confirm an increase in mOS [688]. Rindopepimut + BEV was clinically tested (phase II, NCT01498328) to treat EGFRvIII-positive rGB [686]. Rindopepimut-treated patients achieved robust anti-EGFRvIII titers, and the combined treatment significantly enhanced PFS6 (27% vs. 11%) and mOS (12 vs. 8.8 months) [686]. A recent meta-analysis explored the efficacy and safety of various BEV combination regimens in patients with rGB and concluded that BEV + rindopepimut therapy seems to be safer and more effective [828].
In a phase I trial with DC vaccines targeting EGFRvIII, an immune response was demonstrated with in vitro antigen-specific T cell proliferation and in vivo delayed-type hypersensitivity responses, without significant adverse effects. Subsequently, trials have been planned with polyvalent DC vaccines (directed to numerous GB antigen epitopes) that have demonstrated safety despite the higher risk of autoreactive T cell selection [829].
In C57BL/6 mice, 806-28Z CAR-T cells were able to lyse GL261/EGFRvIII cells in a dose-dependent manner, even eradicating xenografted tumors at high doses. Cell dose and granzyme B release were crucial determinants for CAR-T efficacy, even in heterogeneous GB tumors [830]. A phase I study (NCT02209376) administered CAR-T cells to 10 rGB patients via i.v. infusion. One of the patients remained alive for over 18 months without receiving additional therapy. Nonserious toxicities were observed and trafficking of CART-EGFRvIII cells to regions of active GB was shown in seven treated patients, but with a subsequent antigen decrease in five of them accompanied by an increased and robust expression of immunosuppressive markers such as FOXP3 and PD-L, and infiltration by Tregs, which pointed out the induced adaptive resistance to CART-EGFRvIII cells [831]. Mice treated with dose-intensified TMZ, before administration of CAR-T targeting the EGFRvIII antigen, survived for a median of 174.5 days, compared to 69.5 days in mice pretreated with standard doses of TMZ. On this basis, a phase I trial (NCT02664363) in patients with ndGB incorporating dose-intensified TMZ, as a preconditioning regimen prior to CAR immunotherapy, was initiated in 2018 [832].
In addition, strategies assessing co-administration of vaccines and CAR-T engineered with EGFRvIII and PD-1 are being assayed (NCT04003649, NCT04201873, NCT02529072, NCT02287428). Tandem CAR-T cell targeting simultaneously EGFRvIII and IL-13Rɑ2 is now in preclinical investigation, with an activity superior to their monospecific CAR counterparts in heterogeneous GB cell populations [833]. Bispecific T cell engagers (BiTEs) are bispecific antibodies that redirect T cells to target antigen-expressing tumors. BiTE-secreting T cells have been proposed as a valuable therapy in solid tumors, with distinct properties in mono- or multivalent strategies incorporating chimeric antigen receptor CAR-T cells [834]. Combining BiTEs with CAR-T cell therapy has been tested in preclinical models to avoid antigen-negative relapse. Choi et al. [34] used T cells transduced with an EGFRvIII-targeted CAR construct co-expressing BiTEs against EGFR. These BiTEs bound to CD3 on T cells and EGFR simultaneously, allowing the CAR-T cells to target tumor cells expressing EGFRvIII, EGFR, or both. Although EGFRvIII-targeted CAR-T cells were unable to thoroughly treat GB tumors heterogeneous for EGFRvIII expression, CAR-T cells secreting BiTE molecules were able to eliminate murine GB models after their intracranial administration [805].

5.7. Viral Therapies

GB is particularly suitable for viral therapy to overcome TME immunosuppression and growth being surrounded mainly by post-mitotic cells, which allows the use of viruses that require active cell cycles for replication [835]. Viruses used to treat tumors can be divided into two categories: (1) oncolytic viruses (OVs) with the natural capacity to replicate only in cancer cells (reoviruses, and Newcastle disease viruses) or that are vulnerable to genetic manipulation that increases their tumor selectivity (adenoviruses, herpes simplex viruses, vaccina viruses, polioviruses, and measles viruses); and (2) viral vectors with a low replication rate, which are used as vehicles for other therapeutic genes [836]. Any of the viral categories can stimulate an immune response without the expression of an immunomodulatory transgene. This is most notable for OVs, which cause lytic tumor cell death and induce the release of tumor-associated antigens and damage-related molecular patterns that lead to the activation of innate and adaptive immune responses [837]. Immunogenic cell death is characterized by an immune response that indirectly kills the cancer cells using different mechanisms, i.e., apoptosis, necrosis, and autophagy [838]. Moreover, to potentiate anti-cancer immunity, some OVs have been genetically engineered to express different cytokines (GM-CSF, IL12, and IL15), to enhance antigen presentation, or to elicit a more effective immune response against cancer cells [839,840]. The immunostimulatory ability of OVs allows the transformation of “cold tumors” (i.e., those with an immunosuppressive microenvironment) into immune-responsive “hot tumors” [841], and, as a consequence, OVs function not only as direct cancer-killing agents but also as active anticancer vaccines [842].
Genetic modification can have other purposes, such as to enhance the safety of OVs, to improve the tropism of OVs to cancer cells, and to enhance the anti-tumor effect of OVs [843]. For instance, the genetically attenuated and modified ZIKV strain selectively targets GSCs and exhibits good oncolytic activity [844]. Engineered OVs can also introduce specific therapeutic genes including tumor-suppressor genes (i.e., p53, [845]), anti-angiogenic genes [846], and immunostimulatory genes [847], or induce PI3K inhibition [848,849] in GB cells to execute their expression. Administration of the virus must be efficient and safe for successful therapy. The challenge of transporting viruses to the CNS, crossing the BBB, involves potential elimination by the immune system and also limitations on the dose of viruses [850]. The ability to administer a virus systemically could improve efficacy by reaching distant areas where the tumor has spread, although, in the case of GB metastases, these rarely appear [851]. To circumvent the BBB, other promising techniques for drug delivery have been designed such as convection-enhanced delivery (CED) that uses a pressure gradient in a catheter to transfer therapeutic compounds to the interstitial spaces of the CNS [852,853]. OVs can also be administered intra-arterially and increase BBB permeability with focused ultrasounds [854] or using an osmotic agent such as mannitol [855] (see also Figure 4).
Due to the high transfection efficiency and the development of vector engineering techniques, OVs have been widely used and showed promising results in preclinical and clinical studies in GB (see [843,856,857] for a recent review). In addition, OVs have many advantages over conventional immunotherapies, including precise targeting, effective killing rates, and minimal adverse reactions [858]. To date, several clinical trials have been carried out for oncolytic viruses to improve the treatment of GB [836,843,857]. Examples include adenovirus [859,860,861], herpes simplex virus [862,863,864], reovirus [865,866], parvovirus [867], measles virus [868,869,870], poliovirus [853,871], vaccinia virus [872] and Newcastle disease virus [873]. In addition, other ongoing or completed trials have used the following: (1) modified HSV constructs such as G207 (NCT00028158, NCT03911388 and NCT02457845), HSV-1716 (NCT02031965), MVR-C252 (NCT05095441), M032 (NCT02062827), C134 (NCT03657576); and (2) genetically engineered oncolytic adenovirus combined with SOC or immune checkpoint blockade (NCT02197169, NCT01956734, NCT03896568, NCT01582516, and NCT02798406) [874].
Impressive results have been obtained in a phase II trial (UMIN000015995) in which 19 patients with residual or rGB received G47∆ (a triple-mutated, third-generation oncolytic herpes simplex virus type 1) intratumorally for up to six doses. The 1-year OS rate was 84.2%, the mOS 20.2 months after G47∆ initiation and 28.8 months from the initial surgery. Biopsies revealed an increased number of tumor-infiltrating CD4+/CD8+ lymphocytes and a decrease in immunosuppressive Foxp3+ cells. This study led to the approval of G47∆ as the first oncolytic virus in Japan [676]. The immunostimulatory effects of OVs make them excellent immune adjuvants to enhance chemo- and immunotherapy for GB [875]. Intratumoral DNX-2401 (a tumor-selective oncolytic adenovirus) followed by pembrolizumab was safe and had a notable survival benefit in rGB patients (NCT02798406) [673,674]. There is also an additional ongoing phase I study testing DNX-2401 and TMZ combination for rGB patients in Spain (NCT01956734). A live attenuated poliovirus type 1 vaccine (PVSRIPO) has demonstrated promising outcomes in a phase I study (NCT01491893) involving patients with rGB (20% of long-term survivors) [853]. Currently, PVSRIPO is undergoing further investigation in phase II trials for grade IV malignant glioma (NCT02986178) and in combination with pembrolizumab (NCT04479241) for GB [876].
Despite the promising results of some of the trials presented in Table 4 [673,674,676,689,691], others failed to demonstrate any benefit in ndGB [677] or rGB patients [690]. The discrepancy in results can be attributed to a different replication capacity in different hosts, the concomitant or previously administered treatments, or the corticoid coadministration that can limit the immune response. A recent comprehensive analysis conclude that virotherapy has a satisfactory safety profile and can improve the 2- and 3-year survival rates compared with non-virotherapy (2-year survival: 15% vs. 12%; 3-year survival rate: 9% vs. 6%) [877]. Current viral strategies focus on oral treatments and combination therapies [878,879].

5.8. Immunity-Related Adverse Events

ICIs to elicit anti-tumor response can be accompanied by activation of non-specific immune reactions against antigens expressed by normal tissues. The main immune-related adverse events include diarrhea, colitis, hepatitis, skin toxicities (pruritus, mucositis, and maculopapular rash) and endocrinopathies such as hypophysitis and thyroid dysfunction [880]. To increase the beneficial effects of immunotherapy, it is crucial to detect them as soon as possible and, at the same time, develop interventional strategies to control their severity [881].
The side effects of CAR-T treatment for rGB are still acceptable and most patients only suffer from transient discomfort [801]. One of the most important risks associated with CAR T cell therapy is on-target off-tumor toxicity. With the exception of EGFRvIII, all of the GB antigens that are currently being evaluated clinically may be expressed at low levels in normal tissues, which still can result in substantial toxicity [882,883].
The application of immunotherapy to GB implies a substantial concern for immune-associated CNS neurologic complications. Acute neurologic complications associated with oncolytic viruses and other immunotherapy treatments can often be more directly linked to their intratumoral administration than to a secondary immune-response-related adverse event [813]. While serious immune-mediated adverse events are rare overall, sometimes it is difficult to distinguish immune-mediated adverse events from disease progression. iRANO provides guidelines that can be applied to provide consistent metrics in clinical trials as well as daily practice [54].

6. Nanotherapies

Invasive (local administration) and noninvasive tools to deliver drugs are continuously evolving to overcome the BBB [23]. As reviewed in previous works [884,885,886,887], several nanostructures, including polymeric nanoparticles (NPs) (e.g., dendrimers, polymer micelles or nanospheres), inorganic NPs (e.g., silica, iron, gold or graphene NPs), lipid-based NPs (e.g., liposomes, emulsions), nanogels, carbon dots and nano-implants, have been developed as drug delivery systems and potential diagnostic agents for GB over the past decades. These elements can contain active anti-GB agents, such as chemotherapeutic/anti-angiogenic drugs, radio or chemosensitizers, or immune cells along with moieties that specifically target GB cellular receptors/angiogenic blood vessels or facilitate opening of the BBB [888,889].
Nanosystems (under 200 nm) may readily cross the BBB and fenestrated arteries, formed during the angiogenesis process, and accumulate within the tumor. This accumulation may be facilitated due to the weak lymphatic drainage system surrounding the tumor [887] or actively through the addition of targeting moieties to the surface of the NP [890]. The surface charge also plays a significant role since the electrostatic interaction between positively charged NPs and the negative surface charge of the BBB endothelial cells facilitates NP internalization through adsorptive-mediated endocytosis [891]. However, positively charged NPs can induce the generation of reactive oxygen species (ROS), which elevates their toxicity and restricts the in vivo efficacy [892]. Moreover, nanocarriers can also undergo dispersal throughout the brain and cause damage [893]. To overcome these limitations, NP surfaces can include targeting ligands that selectively recognize specific or overexpressed receptors on tumoral cells (folate, transferrin, neurokinin-1 or v3 integrin receptors) [887]. Many of these targeting ligands can also interact with receptors present on the BBB, which enhances the ability of these systems to cross the BBB through receptor-mediated transcytosis. Different targeting ligands (i.e., proteins, peptides and aptamers) have been utilized to promote active targeting of nanocarriers specifically to glioma cells [894,895,896].
Other advantages of NPs include the following: (1) nanoencapsulation increases their half-life activity, for instance in the case of TMZ-loaded chitosan NPs from 1.8 to 13.4 h [897]; (2) they can incorporate additional fluorescent/MRI/radioactive compounds that allow the non-invasive monitoring of its biodistribution [898]; (3) they increase hydrophobic drug solubility while favoring a proper biodistribution and evading the mononuclear phagocyte system catabolism; (4) they can combine different additional therapeutic approaches, such as (although not exclusively) radiotherapy sensitization, immune cells stimulation, or induction of heat/ROS [899].

6.1. In Vivo Imaging, Chemotherapy and Radiotherapy

Iron oxide nanoparticles (IONPs) have demonstrated promising applications as MRI contrast agents to improve the visualization of intracranial neoplasms [900]. The T2* images of Ferumoxtran-10 (first-generation ultra-small superparamagnetic IONPs) also provide complementary information by enhancing the visualization of glioma vascularity, inflammatory components, and differentiation from radiation necrotic areas better than gadolinium-based contrast agents [901]. Other merits of IONPs include their excellent biocompatibility, biodegradability, and low toxicity. Although they have received regulatory approval for clinical use, some IONPs (i.e., abdoscan, sinerem, endorem and clariscan) were recently discontinued by the pharmaceutical industry because of an inconclusive impact on patient management and for marketing reasons [902].
There is recent interest in magnetic particle imaging, a novel real-time and three-dimensional whole-body imaging modality with high tracer sensitivity and temporal–spatial resolution [903]. Iron–platinum particles enhanced the resolution of T2-weighted MRI images and have magnetic properties that help to guide the nanocomposite to the tumor location [904]. In vivo fingerprint Raman Spectroscopy can distinguish between normal brain, tumor tissue and necrosis with accuracies upwards of 90% in patients for grade 2–4 gliomas [905]. Gold NPs can enhance the Raman signal by several orders of magnitude because of their plasmonic effect or “surface-enhanced Raman spectroscopy” phenomenon [906].
A number of anti-cancer drugs have been successfully delivered to the brain using nanocarriers, i.e., TMZ, paclitaxel, docetaxel, cisplatin, doxorubicin (DOX), vincristine and others [887]. Several nanoformulations are being developed to overcome the limitations of TMZ penetration to improve GB treatment [907,908,909]. Sharma et al. developed a polyamidoamine (PAMAM) dendrimer-chitosan conjugate that increased TMZ brain concentration by almost two-fold [910], thus proving that polymer-based nanosystems can increase the stability of TMZ and control its release. Most of these attempts have been carried out by administering NPs directly into the tumor site, which is largely inconvenient in practice. Systemic administration of the dendrimer–rapamycin conjugate (D-Rapa) allows the release of rapamycin into the TME, improving tumor reduction without significant systemic toxicity [911].
PEGylated liposomes increased the plasma concentration of TMZ and were also more concentrated in the brain, suggesting that PEGylation is a good strategy for overcoming the liver and spleen reticuloendothelial system clearance [912]. Unfortunately, the repeated intravenous injections of PEGylated liposomes led to “accelerated blood clearance” due to the induction of anti-PEG antibodies [913], although solid lipid NPs have demonstrated similar properties to liposomes [914].
Polymer-based nanosystems have been used to co-encapsulate TMZ with other chemotherapies including DOX [915], 5-fluoracil [916] and paclitaxel [917]. Behrooz et al. demonstrated that the designed dendrimer-based pharmaceutical system was able to increase the early apoptosis from 28.2% in the case of the administration of the free drugs by up to 73.3% with the encapsulated co-delivery [917]. In orthotopic human GB xenograft models, loco-regional treatment with TMZ-loaded thermogels caused a significant reduction in the growth in tumor recurrences with no systemic toxicity, compared to untreated controls [918]. Transferrin is overexpressed in proliferating cells and cells that have undergone malignant transformation [919]. Indeed, targeting the transferrin receptor has been demonstrated to be a good strategy to increase the crossing of TMZ or other drugs through the BBB and increase their accumulation in brain tissues [920]. Recently, Helal et al. showed that TMZ-loaded albumin NPs had good uptake by GL261 and BL6 GB cells and increased cytotoxicity [921]. Furthermore, when administered i.v., the NP (labeled with a fluorescent molecule) displayed co-localization with the bioluminescent syngeneic BL6 intracranial tumor murine model used, thus highlighting the potential efficacy of this approach to targeting and treating GBs.
Several NP-based strategies have been developed to interfere with VM formation in GBs, such as (Asn-Gly-Arg) peptide (NGR)-modified liposomes containing epirubicin and celecoxib. These NPs are able to cross the BBB, accumulating in the tumor areas of the brain and destroying VM structures in orthotopic xenograft mouse models [922]. Despite a number of treatment options to target GSCs, most of them have failed in recent clinical trials [923]. Recent examples of GSC targeting include CD133-directed gold NPs, which can be utilized as imaging agents for accurate diagnosis of GB [924] or to sensitize GSCs to radiotherapy [925]. Administration of two chemotherapeutics that act through multiple non-overlapping and synergistic mechanisms is expected to improve the efficacy of treatment and prevent cancer cell drug resistance. Dual functionalized liposomes demonstrated ~12 and 3.3 fold increase in DOX and erlotinib accumulation in mouse brains, respectively, compared to free drugs, and showed excellent antitumor efficacy by inducing a ~90% GB regression and significantly increasing the median survival along with minimum toxicity [926].
Inorganic, polymeric, lipidic, and miscellaneous nanoparticles have been developed for nose-to-brain drug delivery against GB [927]. Intranasal administration of BEV-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (size < 200 nm) improved BEV bioavailability in the brain, and a reduction in tumor growth was accompanied by a higher anti-angiogenic effect compared to the free BEV [928]. Intranasal administration of TMZ (NCT04091503) is being studied in the randomized phase I clinical trial.
Silver (AgNPs), gold (AuNPs) and iron oxide NPs have improved the radiotherapy efficacy in vitro and in GB xenografts [929,930,931]. AgNPs showed more powerful radiosensitizing ability than AuNPs leading to a higher rate of apoptotic cell death [930]. More recently, AgNPs exhibited a higher capacity to radiosensitize hypoxic cells than normoxic cells, which represents an important advantage in GB treatment [932]. To this end, several radiosensitizing techniques are currently under investigation. These include the use of PI3K pathway inhibitors [497,933,934], DNA repair inhibitors [935], hyperthermia [936,937], aldehyde dehydrogenase inhibitors [938], or high atomic number metal NPs [939,940].

6.2. Gene Therapy

Aptamers are short three-dimensional structures of single-stranded nucleic acids (RNA or DNA) that bind molecular targets with high affinity. By adopting an unbiased cell-based variant of the original combinatorial systematic evolution of ligands via the exponential (SELEX) enrichment procedure, RNA aptamers that selectively bind GB cells and GSCs have been generated [941,942]. The results prove that they were able to inhibit cell proliferation, migration and stemness, and were able to strongly reduce tumor growth in vivo, thus proving that this approach is a promising innovative diagnostic and therapeutic tool for GB [943,944]. To date, just one clinical trial has assayed aptamers for GB treatment (NCT04121455). The first and only clinical trial to date using aptamer NOX-A12 in GB patients showed promising results and was well tolerated with no dose-limiting toxicities or treatment-related deaths, which is specifically interesting because it has been assayed in GB patients with unmethylated promoters [945]. The addition of the CXCL12 inhibitor NOX-A12 (olaptesed pegol) to SOC and BEV led to an overall OS rate of 83% (n = 5/6) at a median follow-up of 15 months in ndGB patients, according to preliminary data from an expansion arm of the phase 1/2 GLORIA trial [945].
RNA interference (RNAi)-based therapies use small RNA oligonucleotides (21–45 base pairs) of single- or double-stranded RNA molecules to inhibit protein synthesis. A lipopolymeric nanoparticle (LPNP) formulation has shown remarkable affinity for GSCs and can encapsulate multiple siRNAs for targeted anti-GSCs therapy. When directly infused into rat brain tumors, LPNP siRNAs efficiently inhibit tumor growth and offer promising survival benefits. This multiplexed nanomedicine platform holds great potential as a customized anti-GSCs therapy approach [946]. Despite the potential of RNAi-based therapy in cancer treatment, clinical limitations include short circulatory stability, rapid clearance from the body, and inadequate delivery to the brain tumor tissue. To solve these problems, RNAi-aptamers such as RNAi attached to cell-penetrating peptides and RNAi CNS-ligand conjugates have been explored for GB therapy [947,948,949]. Recently, Wang et al. developed an iron oxide NP system for targeted delivery of siRNAs to suppress the TMZ-resistance gene MGMT. The sequential intravenous administration of these NPs and TMZ resulted in increased apoptosis of GSC and GB cells, reduced tumor growth, and significantly prolonged survival as compared to mice treated with TMZ alone, and this without significant tissue toxicities [950].
The Clustered Regularly Interspaced Short Palindromic Repeat associated (Cas) nuclease 9 (CRISPR-Cas9) system is the latest gene editing technology, which stands out as the fastest, highly versatile, and most reliable gene editing tool for discovering genetic alterations, oncogenic targets, and for epigenetic regulation in various cancers, including GB [951]. It uses an engineered single guide RNA (sgRNA) to direct the Cas9 nuclease to specific DNA sections, leading to double-stranded breaks (DSBs). Induced DSBs are repaired through non-homologous end joining to generate insertions or deletions or homology-directed repair of gene modifications at targeted genomic locations [952,953]. CRISPR-Cas 9 gene editing technology offers novel insights into the roles of various genes in regulating proliferation, stemness, angiogenesis, and invasion of GB cell lines [951]. CRISPR-Cas9 screens are utilized to identify new biomarkers, oncogenic drivers, and causes of chemotherapy resistance in cancer. In that sense, the CRISPR-Cas9 methodology was used to identify growth-related subtypes of GB with therapeutical significance [954]. CRISPR-Cas9 has also been used to generate animals carrying genetic mutations to model human diseases. Based on this, the inactivation of multiple tumor suppressor genes such as Nf1, P53, Ptch1, and PTEN in mouse brains has facilitated the development of medulloblastoma and GB disease models [955,956].
The greatest challenge for CRISPR/Cas9 therapy is how to safely and efficiently deliver it to target sites in vivo, which is especially difficult in the CNS. To overcome this problem, CRISPR/Cas9 therapeutic delivery using viral and nonviral-based delivery vehicles is rapidly expanding [957]. Invasive intra-brain injection of CRISPR-Cas9 complexes contained in viral vectors or NPs often leads to serious side effects, i.e., infection, inflammation, swelling, and tissue injury [958]. Thus, noninvasive delivery of encapsulated CRISPR-Cas9 complexes is urgently needed. Rosenblum et al. describe a safe and efficient lipid nanoparticle (LNP) for the delivery of Cas9 mRNA and sgRNAs. A single intracerebral injection of CRISPR-LNPs against PLK1 in aggressive orthotopic GB caused tumor cell apoptosis, inhibited tumor growth by 50%, and improved survival by 30% but showed diverse adverse effects [959].
BBB-penetrating single CRISPR-Cas9 nanocapsules (~30 nm) have been utilized to target GB cells [960]. Mutagenesis and immunogenicity are the disadvantages of using virus-based vectors due to the long-term expression of CRISPR/Cas9 nuclease and sgRNA, so a transient delivery system is preferred to editing the genome for therapeutic purposes. In that sense, nonviral nanoparticle-based vectors have the advantage that they are often nonimmunogenic and permit bulk production [961].
The i.v. administration of Cas9/sgRNA ribonucleoprotein (RNP) complexes for GB treatment is limited by poor in vivo stability of Cas9 protein and sgRNA, low BBB permeability, and non-specific cell targeting [962]. Ruan et al. developed an angiopep-2 decorated, guanidinium and fluorine functionalized polymeric nanoparticle uploaded with Cas9/gRNA. The guanidinium and fluorine domains were both capable of interacting with and stabilizing Cas9/gRNA RNP in blood circulation without impairing its activity. This NP efficiently crossed the BBB, accumulated in brain tumors, suppressed their growth, and significantly improved the median survival time of mice bearing orthotopic GB, while inducing negligible side or off-target effects [960,962]. Liposome-templated hydrogel nanoparticles (LHNPs) with a unique core-shell nanostructure have been developed for the efficient delivery of Cas9 protein and nucleic acids. When combined with minicircle DNA technology, LHNPs show high effectiveness in cell culture. LHNPs can be tailored for targeted gene suppression in malignancies, especially brain tumors [963]. Additionally, LHNPs can be produced using an autocatalytic brain-tumor-targeting mechanism, which allows targeted delivery of CRISPR/Cas9 to brain tumors. Furthermore, LPHNs nanocarriers loaded with CRISPR/Cas9 plasmids targeting the MGMT gene and modified with the cyclic arginine-glycine-aspartic-conjugated (cRGD) peptide (which targets the overexpressed integrin αvβ3 receptors in tumor cells) were successfully synthesized and demonstrated the ability to protect pCas9/MGMT from enzymatic degradation. In vivo, microbubble–LPHN–cRGD complexes safely and locally enhanced the permeability of the BBB under the effect of focused ultrasound radiation. This facilitated the accumulation of NPs in GB tumors, enhanced the therapeutic effect of TMZ and prolonged survival in mice with orthotopic GB tumors [964]. Systemic delivery of CRISPR/Cas9 engulfed into extracellular vesicles showed improved CRISPR-Cas9 loading efficiency, excellent glioma targeting and penetration potentials, and sensitization of GB cells to radiotherapy, in preclinical animal models, without off-target side effects [965].
Recently, gene editing techniques targeting the PD-1/PD-L1 pathway have attracted considerable interest. However, efficient distribution without causing side effects in clinical trials remains a challenge. To address this, a nanoparticle delivery method, using a low-molecular-weight polyethylenimine (PEI) lipid shell and a PLGA core to package the PD-L1 gRNA-CRISPR/Cas9 plasmid, has been developed. This system effectively transfected human U87 glioma cells with PD-L1. The results represent a promising immunotherapy platform for treating GB [966]. In addition, PEI-coated Fe3O4 NPs have been utilized as a vehicle for delivering therapeutic siRNA to GB cells. These NPs effectively reduced cell proliferation and migration [967]. Similar results have been obtained by a dual-sgRNA CRISPR/Cas9 knockout of PD-L1, thus indicating that intracellular PD-L1 is necessary for tumor progression [968]. CRISPR/Cas9 technology has also allowed an increase in the efficacy of CAR immunotherapy in preclinical GB models. Human neutrophils effectively cross the BBB and display effector immunity against cancer cells, but the short lifespan and resistance to genome editing of primary neutrophils have limited their broad application in immunotherapy. Human pluripotent stem cells were engineered with CARs and differentiated into CAR-neutrophils that are loaded with rough silica (SiO2) NPs containing hypoxia-targeting tirapazamine or other drugs, as a dual immunochemotherapy. Systemically administered CAR-neutrophil@R-SiO2-TPZ NPs first attack external normoxic tumor cells by forming immunological synapses and killing tumor cells through phagocytosis. Thereafter, CAR-neutrophils release R-SiO2-TPZ NPs, which are taken up by hypoxic tumor cells. These CAR-neutrophils loaded with drug-containing SiO2 NPs display superior anti-tumor activities against GB, prolong lifespan in mice bearing orthotopic xenografted GBs and reduce off-target drug delivery [969].
Existing approaches to deliver CRISPR-Cas9 to the brain include viral vector delivery (lentivirus and adeno-associated viruses) [970] and nonviral synthetic delivery (gold, lipid, and polymers) [971], which have provided a valuable in vivo proof of principle. Nevertheless, they are not ideal systems for human clinical applications due to their limited BBB penetration and lack of specific brain disease site targeting [960,972]. Moreover, viral vector delivery can generate highly risky immune responses due to off-targeting effects. Current nonviral delivery systems are limited by their loading efficiency, non-disease targeting, and problematic clearance out of the brain [973]. These problems are being solved one by one [960,962,974] and, although most of the aforementioned studies are in preclinical phases, these novel CRISPR-Cas9–brain delivery systems seem a versatile and potent platform for treating GB and other brain diseases.

6.3. Nanosystems Used for Immunotherapeutic Purposes

NP platforms can be designed to efficiently deliver tumor antigens, cytokines, lymphocytes, and therapeutic agents, stimulating the host’s immune system to effectively target tumor cells in a safe manner. CCL21 is a cytokine predominantly expressed in the lymphoid tissue that functions as a chemoattractant for DCs, lymphocytes, and NK cells. Intratumor injection of CCL21-coupled vault NPs enhanced immune response and significantly inhibited GB growth in mice models [975]. T cells can be encapsulated in a poly(ethyleneglycol)-g-chitosan-based degradable hydrogel, which releases T cells to treat GB effectively [911]. Targeted nanoscale immunoconjugates with poly(β-L-malic acid) (a natural biopolymer), covalently attaching a-CTLA-4 or a-PD-1, crossed the BBB and activated a local brain anti-GB immune response. This resulted in an increase in CD8+ T cells, NK cells and macrophages with a decrease in Tregs in the GB tumor area, and in a significantly increased survival compared to animals treated with single free anti-CTLA-4 and anti-PD-1 monoclonal antibodies [976]. Other similar studies also support a greater availability and effectiveness of ICIs when they are incorporated into NPs [977,978]. An immunosuppressive TME in GB was efficiently altered by nanodiamonds containing doxorubicin (Nano-DOX) [979] and biomimetic nanoparticles (CS-I/J@CM NPs) [980], which stimulated the GB cell immunogenicity and activated a potent anti-GB immune response.
Porous biomaterial scaffolds loaded with GM-CSF have proven to be a promising strategy to increase the chemotactic signal and replenish DCs against GB [760]. Based on this idea, Qui et al. designed an injectable nanocomposite/hydrogel system (DOX/RAcNPs@GM gel) consisting of DOX, GM-CSF and imiquimod-loaded antigen-capturing NPs to actively recruit DCs, aiming to elicit a durable anti-tumor response. To confirm this hypothesis, the authors established an orthotopic GB tumor model that was resected on day 14 after tumor engraftment, while the remaining cavity was injected with hydrogel. DOX/RAcNPs synergized with GM-CSF to enhance immune responses, and rats treated with DOX/RAcNPs@gel displayed reduced tumor volume compared with controls or TMZ-treated rats, although all animals died within 32 days (a fact attributed to the insufficient immune infiltrates in the tumor site) [981].
NPs can protect vaccines from degradation and enhance antigen-presenting cell uptake. Various immunostimulants, such as Toll-like receptor (TLR) agonists and stimulator of interferon genes (STING) agonists, have been preclinically or clinically tested for in situ tumor vaccination, but render suboptimal therapeutic efficacy due to poor tumor retention and rapid systemic dissemination [982]. NvIH is a thermo-responsive hydrogel co-encapsulated with ICI antibodies and novel polymeric NPs loaded with three immunostimulatory agonists for Toll-like receptors 7/8/9 (TLR7/8/9) and STING. In situ vaccination with a single dose of NvIH reduced TME immunosuppression, enhanced TME antitumor immune milieu, and elicited systemic antitumor immunity in an orthotopic murine GB model [983].

6.4. Clinical Trials Using NPs for the Treatment of GB

Although numerous in vivo and in vitro studies have been conducted to prove the therapeutic efficacy of nanocarrier-based treatments against GB, few clinical trials using nanotherapies have been completed. NanoTherm® (aminosilane-coated superparamagnetic iron oxide NPs) generates heat upon application of an external alternating magnetic field, inducing thermal ablation of the tumor tissue [178]. Mechanisms involved in hyperthermia-induced chemo/radiosensitisation include the generation of a transient disruption of the BBB, increased blood flow, interference with DNA repair mechanisms, heat-induced damage to ATP-binding cassette (ABC) transporters, changes in tumor cell drug metabolism, and an impaired ability to withstand apoptotic pathways [984]. Accordingly, in a larger phase II study, NanoTherm® was directly injected intratumorally under stereotactic guidance and then heated using an alternating magnetic field. Treatment was combined with fractionated stereotactic radiotherapy. This approach doubled the mOS of rGB patients (up to 13.4 months) and no serious complications were observed [985]. In Europe, these findings led to the approval of NanoTherm® for magnetic hyperthermia therapy application in combination with RT in patients with rGB.
Most clinical trials are based on liposomal drug-delivery systems. Doxorubicin liposomal formulations have been tested following the Stupp regimen (NCT00944801, NCT009448019) without any survival benefit [986,987]. An ongoing phase II trial (NCT01386580) is evaluating the efficacy of glutathione pegylated liposomal doxorubicin (2B3-101) as monotherapy and in combination with trastuzumab in patients with solid tumors and brain metastases or recurrent malignant glioma. SGT-53 is a complex of cationic liposomes encapsulating a normal human wild-type p53 DNA sequence in a plasmid backbone. This complex has been shown to cross the BBB and to deliver the p53 cDNA efficiently and specifically to the tumor cells. SGT-53 significantly chemo-sensitized human GB cell lines (U87 and U251) to TMZ in vitro and in vivo. Furthermore, in an intracranial GB tumor model, two cycles of concurrent treatment with systemically administered SGT-53 and TMZ inhibited tumor growth, increased apoptosis and, most importantly, significantly prolonged median survival [988]. Interestingly, combining anti-PD-1 with SGT-53 was very effective in inhibiting GB growth, inducing tumor cell apoptosis and increasing intratumoral T cell infiltration, resulting in a significant survival benefit in mice bearing intracranial GB [989]. Unfortunately, the phase II trial (NCT02340156) studying the efficacy of SGT-53 and TMZ combined treatment in rGB patients was terminated due to an insufficient number of participants, making it impossible to perform statistical analysis. Ongoing clinical trials include polysiloxane Gd-Chelates-based NP in combination with radiotherapy and TMZ in ndGB patients (NCT04881032), and NP albumin-bound rapamycin (ABI-009) alone or in combination with BEV, TMZ, or other drugs and radiation therapy in rGB and high-stage ndGB patients (NCT03463265); however, the results are not available yet.
siRNA oligonucleotides arranged on the surface of small spherical AuNPs (NU-0129) were designed to target the oncogene BCL2L12. After intravenous administration, NU-0129 uptake into glioma cells correlated with a reduction in tumor-associated Bcl2L12 protein expression, demonstrating the potential of nanoconjugates as a potential brain-penetrant precision medicine [990].

7. Non-Ionizing Energies in GB Therapy

In addition to the use of TTFields, which are part of the SOC for GBs, other types of non-ionizing energies (described here below) are also being developed for therapeutic purposes.

7.1. Laser Interstitial Thermal Therapy

Laser interstitial thermal therapy (LITT) represents a cutting-edge approach for treating brain tumors that are difficult to access through conventional surgery. By inserting a laser catheter into the tumor, LITT may eradicate the tumor by raising its temperature to lethal levels. The catheter implantation process utilizes state-of-the-art computer imaging techniques, ensuring precision and accuracy. Real-time MRI guides the laser through the catheter, enabling neurosurgeons to target the thermal energy solely at the tumor site to minimize damage to surrounding healthy brain tissue. One of the most notable advantages of LITT is its minimally invasive nature and that most patients can return home the day after treatment and quickly resume their normal activities. LITT also offers hope to patients who have not responded to stereotactic radiosurgery or are suffering from radiation necrosis [991,992]. A single-center study [993] and a prospective multicenter registry [994] conclude that LITT can safely reduce intracranial tumor burden in GB patients who have exhausted other adjuvant therapies or are poor candidates for conventional resection techniques. A statistically significant OS advantage was observed in ndGB patients receiving both radiation and chemotherapy within 12 weeks of LITT (16.14 months) versus those who received only one treatment modality or no treatment following LITT (5.36 months) [995]. New neurologic deficits and postprocedural edema (normally resolved with steroid treatment) are the most frequently reported adverse events after LITT [994].

7.2. Focused Ultrasound (LIFU and HIFU)

Focused ultrasound is an early-stage, therapeutic technology that offers possible adjuvant or alternative treatment strategies for GB [996]. This groundbreaking approach involves precisely targeting deep areas of the brain with beams of ultrasonic energy without the need for incisions. This being said, it is important to differentiate between low- and high-intensity (100–10,000 W/cm2) focused ultrasounds (LIFUs and HIFUs). LIFU disrupt the BBB or blood tumor barriers and enhance the uptake of therapeutic agents in the CNS. HIFU can cause thermoablation and mechanical destruction of the tumor. Both can be combined with radiotherapy [997,998].
The advantages of focused ultrasounds over current brain tumor treatments are considerable: (1) eliminating concerns related to surgical wound healing and the risk of infection, making it a safer option for patients; (2) precise targeting; (3) avoiding ionizing radiation exposure; (4) enhancing chemotherapy delivery by temporarily opening the BBB; and (5) their non-invasive nature allows for repeat treatments [854,997].
HIFUs produce frictional heat by causing the vibration of molecules within the tissue. The absorbed energy can quickly elevate the temperature to over 55 °C, which causes protein denaturation, DNA fragmentation and coagulative necrosis when maintained for just a few seconds [999,1000]. This thermoablative process further increases tumor sensitivity to radiation by damaging DNA repair enzymes [997]. To date, clinical data are limited to case reports such as those first reported by Coluccia et al. using MRI-guided HIFUs to achieve tumor ablation without inducing neurological deficits or other adverse effects in a patient with rGB [1001]. Two phase I clinical trials (NCT01473485 and NCT00147056) have evaluated the safety and efficacy of transcranial MRgFUS (magnetic resonance-guided focused ultrasounds) thermoablation for the treatment of either brain metastasis or recurrent glioma, but the results have not been published. MacDonell et al. have recently proposed an interstitial HIFU device that employs an intraparenchymal catheter to induce hyperthermia directly at the tumor tissue, assisted by an MRI-guided robotic system. The advantages of this interstitial device over external MRI-guided HIFU include avoiding attenuation from the skull, improving treatment margins, and enabling concurrent tissue sampling [1002]. Animal studies have demonstrated the feasibility of this technique, but its clinical success has not yet been validated. HIFU technology is approved by the FDA for treatment of several cancers (i.e., prostate, uterine leiomyoma and bone metastasis) and is under investigation for other neoplasms [1003]. The primary limitation of its application in GB is the absence of well-circumscribed lesions [1000].
In contrast, LIFU uses relatively lower energy pulsed waves (around 500 kHz) relying on mechanical perturbation and acoustic cavitation. Cavitation refers to the oscillation and collapse of gas bubbles in response to the compression and refraction of the ultrasonic pressure wave [1003]. LIFU is therefore generally used in conjunction with microbubbles, which can be delivered intravenously and travel to the site targeted by the transducer [1004]. These particles oscillate in the presence of the ultrasound wave, expanding and contracting to produce a stable cavitation effect that disrupts the tight junctions of endothelial cells. Thus, LIFU has been explored as a method to transiently increase the permeability of the BBB to enhance therapeutic delivery, limiting the side effects by ensuring that the impermeable state of the BBB is quickly restored [998]. The precision of LIFU can be enhanced using MRI (MRgLIFU), thus minimizing the effects on healthy tissue [1005]. Furthermore, the opening of the BBB can be confirmed with contrast-enhanced MRI, allowing real-time monitoring of the biological effects of LIFU [1003]. In animal studies, BBB opening is immediate, repeatable, resolves within 6 to 8 h, and does not cause axonal or neuronal injury. The improved delivery of BCNU, TMZ, carboplatin and others, traditionally rendered ineffective by the impermeability of the BBB, has been verified. As a consequence, this treatment not only delayed tumor progression but also enhanced survival in GB animal models [1006,1007,1008]. LIFU has also been used to deliver viruses [1009], cells [1010] and NPs (loaded with imaging agents, therapeutic agents, or both) [1011,1012,1013]. In addition, the microbubbles can be loaded with tumor antigens, allowing a more focused and effective immune response [1014,1015]. It is also noteworthy that preclinical studies suggest that LIFU reduces the TME-induced immunosuppression by increasing infiltration of NK and CD8+ T cells, thus facilitating DCs maturation and diminishing the number of Tregs and MDSCs [1016,1017,1018,1019]. MRgLIFU-mediated BBB disruption has been utilized in the clinical setting to deliver carboplatin, TMZ, doxorubicin, fluorescein or paclitaxel in GB patients [1020,1021,1022]. The treatment was well tolerated, and disruption resulted in a 15–20% increase in contrast enhancement almost instantaneously, resolving after 20–24 h. A single-center trial (NCT02253212) did not show serious adverse events or carboplatin-related neurotoxicity associated with the implantation of a LIFU device with microbubble injection in rGB patients. Patients with documented BBB disruption relative to patients without or with poor BBB disruption had longer PFS (4.11 vs. 2.73 months) and OS (12.94 vs. 8.64 months) [1020]. A small trial (NCT03712293) of six patients with GB treated with multiple cycles of MRgLIFU had an improved penetration of TMZ without immediate or delayed BBB-disruption-related complications. All subjects survived over 1 year, while tumor recurrence was noted in two patients at 11 and 16 months [1021]. These recent results evidence that LIFU-mediated BBB opening increases drug delivery in GB, thus improving tumor control and survival, although larger sample sizes are needed to confirm efficacy and the lack of hemorrhagic complications associated with the procedure [1023].
At present, a number of clinical trials (NCT03551249, NCT04440358, NCT04417088, NCT05370508, NCT06039709) are ongoing or recruiting patients with GB for focused ultrasound treatment (ClinicalTrials.gov, 4 February 2024).

7.3. Photodynamic and Sonodynamic Therapies

Photodynamic therapy (PDT) and sonodynamic therapy (SDT) are emerging modalities for non-invasive cancer treatment, based on the tumor-selective accumulation of non-toxic molecules [photosensitizers (PS) or sonosensitizers (SS)], which are activated by laser light or ultrasound radiation to produce a localized cytotoxic effect via ROS generation [1024,1025,1026,1027]. Apoptotic and necrotic cells elicit the proliferation of effector T cells in the lymph nodes, resulting in further GB eradication. Both techniques can also induce autophagy, endothelial damage, angiogenesis inhibition (associated with ischemia and necrosis), and immune responses [1027,1028,1029,1030].
Protoporphyrin IX (PpIX) and fluorescein are most widely used as PS and as SS due to their safety profile and selective accumulation in tumor cells, and most studies in glioma use 5-aminolevulinic acid (5-ALA) as a precursor of PpIX [1031,1032,1033]. After surgery and adjuvant treatment, the residual tumor is predominantly comprised of resistant GSC clones; thus, several strategies have attempted to enhance 5-ALA uptake by this type of cells in order to increase the efficacy of PDT and SDT [1032,1033]. Moreover, iron chelators, such as deferoxamine and CP94, and ABC transporter inhibitors have been shown to increase PpIX levels in GB cells when utilized as adjuvants [1034,1035]. GSCs exhibit less accumulation of PpIX than non-GSCs, and deferoxamine-induced iron chelation significantly enhances the 5-ALA-mediated PpIX accumulation in GSCs [1034].
Interstitial PDT (iPDT) is a minimally invasive procedure performed in patients whose tumors are present in areas of the brain with readily identifiable neurological function, or in fragile patients who cannot undergo a craniotomy. iPDT is applied via the stereotactic insertion of fiber optic cable(s) into the tumor to deliver photostimulation to the tumor mass after administering a PS to the patient. Intracavitary PDT is applied in the resection cavity at the end of the surgical procedure [1025]. The most frequent PDT-associated adverse events are retinal and cutaneous photosensitivity that can last from several days to a few weeks depending on the PS used and time of exposure. Less frequent adverse events include post-operative hemorrhage, neurological deficits (particularly in the case of large tumors), infection, uncontrolled cerebral edema, and even death [1025]. The use of iPDT against GB over the last 3 decades demonstrates that, overall, the technique is safe and effective. The mPFS was 14.5 months for ndGB and 14 months for rGB patients, which means there is an improvement compared to historical controls [1036]. Different single-center retrospective studies have reported prolonged long-term survivals when using iPDT [1037,1038,1039,1040]. However, the lack of a standard control group, restricted sample sizes, and the lack of information about their characteristics, makes it difficult to draw conclusions. Ongoing clinical trials (NCT04469699, NCT03897491, NCT04391062) are actively investigating the potential of PDT in GB treatment [1027]. The major disadvantage of PDT is the limited penetration of laser light into deep tissues. This could be mitigated using near-infrared radiation (NIR) that can penetrate 3 cm through skin and bone structures [1041]. In fact, NIR photons also diminish both phototoxicity and background autofluorescence, which then leads to improved bio-imaging when compared to traditional fluorescence with visible light [1042].
Recent advances in PDT-GB research are as follows: (1) coupled NIR and photo immunotherapy (NIR-PIT), where a photosensitizer is conjugated to a highly specific monoclonal antibody [1043,1044,1045]; (2) NP-based PDT to augment systemic therapies and avoid skin photosensitization [1046,1047]; (3) NP-based PDT linked to miRNA or other chemotherapeutic agents [1048]; (4) strategies to increase PDT efficacy in hypoxic TME conditions [1049,1050,1051,1052]. Although some studies have demonstrated the enhanced penetration of light by simultaneous application of ultrasounds [1053], combined effects of PDT and SDT did not show any benefit in glioma rat models [1054].
SDT requires the interaction of an SS, ultrasounds, and oxygen. The generation of ROS through the stimulated SS and the ultrasound-activated cavitation effects can together induce apoptosis, necrosis, and autophagy, ultimately causing tumor destruction [1026]. The major advantage of SDT for GB treatment is the ability of ultrasound energy to penetrate into soft tissues more than 10 cm, and the possibility of delivering a tightly focused ultrasound beam for focal treatment [1031]. SDT inhibits tumor growth and increases animal survival in preclinical studies [1028,1055,1056,1057]. A significant step forward was made by Raspagliesi et al., who reported, in a porcine animal model, the first intracranial MRI-guided SDT with fluorescein and 5-ALA using the ExAblate system. The porcine model allows a more precise target definition, and better approaches human conditions [1058]. The efficacy of STD is limited in larger tumors because ultrasound waves cannot penetrate deep enough into the tumor [1059]. Three clinical trials are currently underway to evaluate the safety and feasibility of SDT with 5-ALA in patients with high-grade glioma (NCT04559685), in ndGBs using the ExAblate Model 4000 Type-2 Neuro System (NCT04845919), and the efficacy of SDT using SONALA-001 and Exablate Type-2 devices in subjects with rGB (NCT04988750).

7.4. Microwaves and Pulsed Electric Fields

Microwaves are a form of electromagnetic radiation with wavelengths ranging from about 1 mm to 30 cm, corresponding to frequencies between 1 GHz and 300 GHz, respectively [1060]. Microwaves have been introduced in medicine for cancer diagnosis [1061] and treatment [1062,1063]. However, microwaves have well-known adverse effects on the CNS and can affect neurotransmitter release and, thereby, cause a delay in the signaling process [1064]. Despite these limitations, the research of Rana et al. is an excellent example of how microwave radiation can therapeutically be used to treat GBs [1065]. These authors have shown that a strong electric field (~23 kV/cm) of pulsed high-power microwave (HPM) irradiation causes ROS generation, DNA damage, p53 activation and death in exposed GB U87 cells. Importantly, these authors show that pulse dosage causing damage to GB cells and brain normal cells is different, thus representing a new therapeutic approach that deserves to be tested rapidly in in vivo models. In fact, pulsed microwave-induced thermoacoustic therapy has been recently proposed as a potential alternative modality to precisely and effectively eradicate orthotopic GB. Interestingly, an NP composed of polar amino acids and adenosine-based agonists has been developed having a high microwave absorbance and selective penetration of the BBB. The NP activates the adenosine receptor on the BBB to allow self-passage, and once it is accumulated in the TME, the NP converts absorbed microwaves into ultrasonic shockwaves, which can mechanically destroy tumor cells with minimal damage to adjacent normal brain tissue due to the rapid decay of the ultrasonic shockwave intensity [1066].
Pulsed electrical fields [PEF, high voltage/short-duration (nanoseconds-milliseconds) electrical pulses] have emerged as a non-thermal tissue ablation treatment for malignant neoplasms. This technique, where rod/needle-like electrodes are strategically placed directly in or surrounding the tumor, is associated with the terms electroporation and electro-permeabilization [1067]. Importantly, reversible permeabilization of the cell membrane can also increase the uptake of chemotherapeutics or facilitate transfection approaches [1068]. In the sub-microsecond regime, intracellular effects like nuclear membrane disruption have been observed [1069]. The strength and duration of the nanosecond pulse can create nano-sized pores and electroporation effects [1070,1071,1072]. Depending on these parameters, cell electroporation and permeabilization can be reversible, with membrane recovery typically taking minutes, although intracellular repair may require hours [1073]. Sub-microsecond pulses have also been found to trigger apoptotic cell death in different cell lines and tumor tissues. Apoptosis induction is associated with caspase activation [1074], intracellular Ca2+ release [1075], loss of mitochondrial membrane potential [1076], and DNA damage [1077]. These findings indicate that sub-microsecond PEF protocols, below electroporation thresholds, may offer potential therapeutic benefits for GB treatment and should be explored further [1078].
The first demonstration using irreversible electroporation (IRE) against a dog malignant intracranial glioma was published in 2011 by Garcia et al. [1079]. Canine malignant gliomas share similarities with GB in various clinical, biological, pathologic, molecular, and genetic aspects, making them a valuable model [1080]. In the referenced study, IRE delivery resulted in an approximately 74% reduction in tumor volume. IRE treatment was well tolerated and achieved safe tumor ablation when combined with radiotherapy, anti-edema treatment, and anticonvulsants, with minimal hemorrhage [1079]. More recently, further investigations using the Nanoknife procedure in seven dog glioma models revealed that IRE treatment was successful in six out of seven dogs without inducing or exacerbating edema or causing a significant hemorrhage [1081]. While most adverse effects were minimal or typical of post-operative surgery, one dog experienced severe cerebral edema (due to the tumor location being close to periventricular regions, a common site of occurrence in human glioma). This highlights the importance of considering tumor location and potential effects during the pre-treatment and planning of IRE in the brain. IRE procedures have shown preservation of critical structures and major blood vessels in humans which is an advantage over microwave or radiofrequency ablation methods [1082].
High-frequency irreversible electroporation (H-FIRE, or 2nd generation of IRE) works by delivering short, 1–10 μs pulses in a series of bursts, equivalent to a single monopolar 100 μs pulse used in traditional IRE. However, this approach requires much greater field strength to achieve the same lesion size [1083,1084]. Latouche et al. conducted an experiment using H-FIRE treatment to selectively ablate intracranial meningioma in three dogs. One of the dogs was alive after 6 months without evidence of the presence of a tumor. Another dog was alive but required increased anticonvulsants to control seizure activity, and there were suspicions of residual or recurrent tumors on an MRI 5 months after treatment. Unfortunately, the third died after 76 days due to a recurrent status epilepticus. This study showed no post-operative adverse effects attributed to H-FIRE [1085]. Recently, Campelo et al. reported further evidence that H-FIRE improves survival and immune cell infiltration in rodents with malignant gliomas [1086]. Although the application of these techniques is still in its infancy, available results indicate the potential of H-FIRE’s for brain tumor ablation, thus representing an exciting opportunity for clinical applications.

7.5. Targeted Radionuclide Therapy

Targeted radionuclide therapy (TRT) is based on the use of a molecule labeled with a radionuclide to deliver (through systemic or local administration) a toxic level of radiation to cancer cells. This is achieved by employing a biochemical vector, which is linked to a radionuclide and, in most cases, allows both diagnostic and also therapeutic applications. The energy, range of radiation, and type of emission are critical in targeted radionuclide therapy. Unlike molecular imaging, which involves the use of highly penetrating γ- and positron (β+)-emitting radionuclides, TRT employs β, α, or auger electron emitters with lower penetration capacity but higher ionizing energy [1087]. β particle-emitting radionuclides (e.g., 131I, 90Y, 186/188Re and 177Lu) can irradiate tissue volumes with multicellular dimensions and induce radical formation leading to DNA single-strand breaks. For small tumors, micrometastatic lesions, or residual disease, α-particles (emitted by 213Bi, 225Ac, or 211At) are considered a better option, owing to their short travel distance in tissue (only a few diameters) and high linear energy transfer (LET) (50–230 keV/μm). α-particles induce DNA DSBs that directly trigger cell death, independently of the cell cycle phase, the cell oxygenation level and the MGMT gene promoter methylation status [1088].
Radiolabeled small molecules, radioimmunotherapy (RIT), peptide radionuclide therapy (PRT) and radioNPs are four different modalities of TRT [1089,1090,1091]. RIT uses a monoclonal antibody to achieve targeted vectorization of a radionuclide. Clinical trials for certain antigen targets, like EGFR [1092,1093], tenascin [1094,1095,1096,1097], or DNA histone H1 complex [1098], have shown positive outcomes. Tenascin targeting appears to be one of the most promising RITs for GB. mOS of GB patients treated by fractionated intracavitary radioimmunotherapy with 131I- or 90Y-labeled anti-tenascin monoclonal antibody reached 25.3 months, thus markedly exceeding that of historical controls, being adverse events well controllable [1099]. In the radiopeptide approach, an agonist or antagonist peptide is used to vectorize the radionuclide to a specific receptor overexpressed in cancer cells. In GB clinical trials, radiolabeled somatostatin analogs have been used to target the somatostatin receptor [1100], radiolabeled substance P has been used to target the neurokinin receptor type 1 [1101,1102,1103], and TM-601 (a recombinant version of chlorotoxin) has been used to target the matrix metalloproteinase [1104]. In most of these assays, partial remissions and an improved OS have been observed. TAM and microglia involved in TME immunosuppression are characterized by the upregulation of somatostatin receptor 2; therefore, targeting this receptor has the additional advantage of increasing the immune response against GB [1088]. RadioNP can passively accumulate in the tumor or can have a biologically active peptide or antibody for specific targeting in the same way as the molecular radiopharmaceuticals used in RIT and PRRT.
RadioNPs can be delivered passively or actively using liposomes, metallofullerenes, or lipid nanocapsules. Recently, Georgiou et al. administered 177Lu-AuNPs by CED to treat orthotopic U251-Luc human GB tumors in NRG mice. A high proportion of 177Lu-AuNPs was retained in the U251-Luc tumor for up to 21 days with minimal redistribution to the brain or healthy tissues. The radiation dose in the tumor was 599 Gy, whereas in the surrounding brain, it was 93-fold lower (6.4 Gy), and 2000–3000-fold lower doses were calculated for the contralateral left cerebral hemisphere (0.3 Gy). MRI at 28 days post-treatment showed no visible tumor in mice treated with 177Lu-AuNPs, and 5/8 of them survived up to 150 days, whereas controls had large tumors and required sacrifice within 45 days post-treatment. The results of this study are promising
TRT holds the potential to serve as a potent and supplementary treatment following SOC therapy for primary GBs. It can also be employed as an auxiliary treatment option in cases where the tumor tissue shows resistance to radiation and/or chemotherapy [1088,1105]. Radiopharmaceuticals can be administered systemically or intratumorally/ intracavitarily to circumvent the BBB [1089,1090]. The lack of serious adverse effects and promising results of the previously mentioned phase I/II trials makes TRT an attractive treatment modality that should be considered for phase III trials assays integrated into combined-modality regimens [1106,1107]. As an added advantage, TRT allows interesting theragnostic approaches, thus permitting personalized therapy [1089,1108].

8. Conclusions

No other cancer has witnessed as many therapeutic attempts as GB. Unfortunately, although every treatment option in the Oncotherapy arsenal has been tried, GB almost always recurs. In fact, if you read Dr. Siddhartha Mukherjee’s book The Emperor of All Maladies: A Biography of Cancer, it is easy to think about GB as the emperor of all cancers.
Despite improvements in shorter-term survival rates, the 5-year survival rate after diagnosis remains relatively constant (only 5.8% of all patients). Key advancements, limitations, and perspectives can be summarized as follows:
  • GB location, aggressivity and infiltrative biological behavior represent exceptional challenges. Surgeons face great difficulties in removing all cancerous cells without causing damage to critical brain regions, thus preserving brain function while treating the tumor becomes a delicate balance. Aggressive treatments might easily lead to cognitive impairments and/or neurological deficits.
  • Standard treatment and targeted therapies have only modestly improved patient outcomes, a fact likely due to redundant compensatory mechanisms, insufficient target coverage (in part related to the BBB), and GSC’s ability to remain quiescent or upregulate their molecular defenses until cytotoxic drug concentrations decrease. Mechanisms favor treatment failure and tumor recurrence.
  • Targeting of key contributors to GSC’s self-renewal, survival, and plasticity will contribute to avoiding recurrences. ChemoID test (NCT03632135) uses CSCs isolated from tumor biopsies to select the most effective chemotherapeutic treatment for each patient [1109]. Strategies like this can assist in identifying the best treatment option and in properly designing recruitment for clinical trials.
  • The BBB restricts the entry of many therapeutic agents into the brain, limiting the effectiveness of systemic treatments. Although the BBB and BTB can be disrupted in a variety of ways depending on the stage of the illness, their heterogeneous breakdown makes it quite difficult to achieve uniform drug concentrations inside the tumor. Strategies to improve drug delivery at bioefficacious concentrations include intranasal or intratumoral administrations (with limited acceptance in clinical practice), opening of the BBB with LIFU, and the use of nanocarriers. Especially interesting are multifunctional nanocarriers that allow the delivery of diverse molecules (chemotherapeutic and/or immunotherapeutic and/or imaging agents) and can also be used for theranostic purposes. In addition, receptor-based therapies have the potential to improve clinical outcomes. Nevertheless, most of these technologies are, at this moment, in the development stages.
  • While immunotherapies have revolutionized treatment for other cancers, in GB, patients have not achieved similar positive responses, a fact associated with the unique challenges associated with immunosuppressive TME. GB exhibits an immune-privileged nature characterized by limited lymphocytic infiltration, cytotoxic T cell exhaustion, recruitment of pro-tumorigenic TAMs, downregulation of cancer cells’ MHC I complexes, and abundance of immune-inhibitory molecules such as IL-10 and TGFβ. In our opinion, DC vaccination, oncolytic virotherapy, CAR-cell therapy or combined strategies (with ICIs) seem promising, and outcomes could likely be improved by targeting antigens present in GCS. In any case, immunotherapy should be considered as part of the combined treatment needed to increase patient survival. The integration of immunotherapy with SOC for GB presents opportunities and challenges, since radio/chemotherapy can substantially influence the immune response in two different ways. Radiotherapy can induce immunogenic cell death, thus facilitating the release of tumor antigens that could potentially amplify immunotherapy effectiveness. On the contrary, radio and chemotherapy can also induce lymphopenia and augment the expression of immunosuppressive molecules (PD-L1) within the TME, hence compromising the efficacy of immunotherapeutic treatments. The timing (adjuvant or neoadjuvant) for the best synergistic effect remains a problem to resolve.
  • The implementation of multi-modal combined strategies is essential to increase survival and the quality of life of GB patients. The design of personalized treatments should be based on the genetic profiling and characteristics of this heterogeneous tumor, its specific location, the selection of the most promising targets, including the GSC, and the stimulation of the immune system. Personalized combined treatments offer advantages in terms of efficacy and reduced collateral damage, but the time needed to make decisions and cost represent significant challenges. The use of mathematical models of synergism/antagonism of drugs and pathways will help in predicting drug combinations for this multifactorial disease.
  • We should reconsider what has not been done properly. To enroll patients in a trial, the genetic characteristics of their tumors should be considered, including only those that present the corresponding therapeutic targets. Only in this way will we ensure that results become objective and significant. Administering immunotherapies concurrently with corticosteroids or other immunosuppressive treatments is nonsensical. Such combinations must be considered when analyzing clinical study results to prevent misleading conclusions.
  • TRT, HIFU, PDT and SDT can be alternatives (particularly in patients with poor physical condition), complements to radiotherapy, or become part of the SOC.

Author Contributions

E.O., J.M.E. and M.Á.M.-T. wrote the first draft. M.O.-C., R.L.-B., P.M.-M., B.P., J.L.G.-A. and A.L. prepared the figures and tables with the supervision of the corresponding authors. E.O., J.M.E. and M.Á.M.-T. supervised and edited the final draft. All authors have contributed to gathering and analyzing information. All authors have read and agreed to the published version of the manuscript.

Funding

This study is funded by MCIN/AEI/10.13039/501100011033/ and European Union Next Generation EU/PRTR (grant number CPP2021-8817), and Scientia BioTech S.L.-Spain (grant number SBT-001). The authors gratefully acknowledge the use of the services and facilities of the University of Valencia and the Jaume I University of Castellon. We thank to K. Ferguson for corrections in editing. J.L.G.A. thanks an “Epriex” contract from Plan Nacional de Recuperación, Transformación y Resiliencia. A.L. acknowledges a “Margarita Salas” postdoctoral contract (number 21-076) funded by the European Union-Next generation EU. M.A.M.-T was supported by a ‘Maria Zambrano’ research contract (number MAZ/2021/03 UP2021-021) funded by the European Union-Next generation EU and Ramon y Cajal Research Fellow RYC2022-038481-by MCIN/AEI/ 10.13039/501100011033.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

Authors Elena Obrador, Paz Moreno-Murciano, María Oriol-Caballo, Rafael López-Blanch and José M. Estrela were employed by Scientia BioTech S.L. The author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A Summary. Neuro-Oncology 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  2. Berger, T.R.; Wen, P.Y.; Lang-Orsini, M.; Chukwueke, U.N. World Health Organization 2021 Classification of Central Nervous System Tumors and Implications for Therapy for Adult-Type Gliomas: A Review. JAMA Oncol. 2022, 8, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
  3. Kurokawa, R.; Kurokawa, M.; Baba, A.; Ota, Y.; Pinarbasi, E.; Camelo-Piragua, S.; Capizzano, A.A.; Liao, E.; Srinivasan, A.; Moritani, T. Major Changes in 2021 World Health Organization Classification of Central Nervous System Tumors. RadioGraphics 2022, 42, 1474–1493. [Google Scholar] [CrossRef] [PubMed]
  4. Torp, S.H.; Solheim, O.; Skjulsvik, A.J. The WHO 2021 Classification of Central Nervous System Tumours: A Practical Update on What Neurosurgeons Need to Know—A Minireview. Acta Neurochir 2022, 164, 2453–2464. [Google Scholar] [CrossRef] [PubMed]
  5. Thomas, D.L. 2021 Updates to the World Health Organization Classification of Adult-Type and Pediatric-Type Diffuse Gliomas: A Clinical Practice Review. Chin. Clin. Oncol. 2023, 12, 7. [Google Scholar] [CrossRef] [PubMed]
  6. Singer, L.S.; Feldman, A.Z.; Buerki, R.A.; Horbinski, C.M.; Lukas, R.V.; Stupp, R. The Impact of the Molecular Classification of Glioblastoma on the Interpretation of Therapeutic Clinical Trial Results. Chin. Clin. Oncol. 2021, 10, 38. [Google Scholar] [CrossRef]
  7. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  8. Vehlow, A.; Cordes, N. Invasion as Target for Therapy of Glioblastoma Multiforme. Biochim. Biophys. Acta (BBA) Rev. Cancer 2013, 1836, 236–244. [Google Scholar] [CrossRef]
  9. Davis, M.E. Glioblastoma: Overview of Disease and Treatment. Clin. J. Oncol. Nurs. 2016, 20, S2-8. [Google Scholar] [CrossRef]
  10. Cantrell, J.N.; Waddle, M.R.; Rotman, M.; Peterson, J.L.; Ruiz-Garcia, H.; Heckman, M.G.; Quiñones-Hinojosa, A.; Rosenfeld, S.S.; Brown, P.D.; Trifiletti, D.M. Progress Toward Long-Term Survivors of Glioblastoma. Mayo Clin. Proc. 2019, 94, 1278–1286. [Google Scholar] [CrossRef]
  11. Thakkar, J.P.; Dolecek, T.A.; Horbinski, C.; Ostrom, Q.T.; Lightner, D.D.; Barnholtz-Sloan, J.S.; Villano, J.L. Epidemiologic and Molecular Prognostic Review of Glioblastoma. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1985–1996. [Google Scholar] [CrossRef]
  12. Aldoghachi, A.F.; Aldoghachi, A.F.; Breyne, K.; Ling, K.-H.; Cheah, P.-S. Recent Advances in the Therapeutic Strategies of Glioblastoma Multiforme. Neuroscience 2022, 491, 240–270. [Google Scholar] [CrossRef]
  13. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef]
  14. Revilla-Pacheco, F.; Rodríguez-Salgado, P.; Barrera-Ramírez, M.; Morales-Ruiz, M.P.; Loyo-Varela, M.; Rubalcava-Ortega, J.; Herrada-Pineda, T. Extent of Resection and Survival in Patients with Glioblastoma Multiforme: Systematic Review and Meta-Analysis. Medicine 2021, 100, e26432. [Google Scholar] [CrossRef] [PubMed]
  15. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015–2019. Neuro Oncol. 2022, 24, v1–v95. [Google Scholar] [CrossRef] [PubMed]
  16. Brennan, C.W.; Verhaak, R.G.W.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The Somatic Genomic Landscape of Glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef] [PubMed]
  17. Perrin, S.L.; Samuel, M.S.; Koszyca, B.; Brown, M.P.; Ebert, L.M.; Oksdath, M.; Gomez, G.A. Glioblastoma Heterogeneity and the Tumour Microenvironment: Implications for Preclinical Research and Development of New Treatments. Biochem. Soc. Trans. 2019, 47, 625–638. [Google Scholar] [CrossRef] [PubMed]
  18. Jovčevska, I. Sequencing the next Generation of Glioblastomas. Crit. Rev. Clin. Lab. Sci. 2018, 55, 264–282. [Google Scholar] [CrossRef]
  19. Kim, H.; Zheng, S.; Amini, S.S.; Virk, S.M.; Mikkelsen, T.; Brat, D.J.; Grimsby, J.; Sougnez, C.; Muller, F.; Hu, J.; et al. Whole-Genome and Multisector Exome Sequencing of Primary and Post-Treatment Glioblastoma Reveals Patterns of Tumor Evolution. Genome Res. 2015, 25, 316–327. [Google Scholar] [CrossRef]
  20. Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; deCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y.; et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell 2017, 32, 42–56.e6. [Google Scholar] [CrossRef]
  21. Becker, A.P.; Sells, B.E.; Haque, S.J.; Chakravarti, A. Tumor Heterogeneity in Glioblastomas: From Light Microscopy to Molecular Pathology. Cancers 2021, 13, 761. [Google Scholar] [CrossRef]
  22. Wu, W.; Klockow, J.L.; Zhang, M.; Lafortune, F.; Chang, E.; Jin, L.; Wu, Y.; Daldrup-Link, H.E. Glioblastoma Multiforme (GBM): An Overview of Current Therapies and Mechanisms of Resistance. Pharmacol. Res. 2021, 171, 105780. [Google Scholar] [CrossRef]
  23. Ahmed, M.H.; Canney, M.; Carpentier, A.; Thanou, M.; Idbaih, A. Unveiling the Enigma of the Blood–Brain Barrier in Glioblastoma: Current Advances from Preclinical and Clinical Studies. Curr. Opin. Oncol. 2023, 35, 522–528. [Google Scholar] [CrossRef] [PubMed]
  24. Stiles, C.D.; Rowitch, D.H. Glioma Stem Cells: A Midterm Exam. Neuron 2008, 58, 832–846. [Google Scholar] [CrossRef] [PubMed]
  25. Safa, A.R.; Saadatzadeh, M.R.; Cohen-Gadol, A.A.; Pollok, K.E.; Bijangi-Vishehsaraei, K. Glioblastoma Stem Cells (GSCs) Epigenetic Plasticity and Interconversion between Differentiated Non-GSCs and GSCs. Genes Dis. 2015, 2, 152–163. [Google Scholar] [CrossRef]
  26. Xie, X.P.; Laks, D.R.; Sun, D.; Ganbold, M.; Wang, Z.; Pedraza, A.M.; Bale, T.; Tabar, V.; Brennan, C.; Zhou, X.; et al. Quiescent Human Glioblastoma Cancer Stem Cells Drive Tumor Initiation, Expansion, and Recurrence Following Chemotherapy. Dev. Cell 2022, 57, 32–46.e8. [Google Scholar] [CrossRef]
  27. Hu, Y.; Li, Z.; Zhang, Y.; Wu, Y.; Liu, Z.; Zeng, J.; Hao, Z.; Li, J.; Ren, J.; Yao, M. The Evolution of Tumor Microenvironment in Gliomas and Its Implication for Target Therapy. Int. J. Biol. Sci. 2023, 19, 4311–4326. [Google Scholar] [CrossRef] [PubMed]
  28. Di Nunno, V.; Franceschi, E.; Tosoni, A.; Gatto, L.; Bartolini, S.; Brandes, A.A. Glioblastoma Microenvironment: From an Inviolable Defense to a Therapeutic Chance. Front. Oncol. 2022, 12, 852950. [Google Scholar] [CrossRef] [PubMed]
  29. Schaff, L.R.; Mellinghoff, I.K. Glioblastoma and Other Primary Brain Malignancies in Adults: A Review. JAMA 2023, 329, 574–587. [Google Scholar] [CrossRef] [PubMed]
  30. Furnari, F.B.; Fenton, T.; Bachoo, R.M.; Mukasa, A.; Stommel, J.M.; Stegh, A.; Hahn, W.C.; Ligon, K.L.; Louis, D.N.; Brennan, C.; et al. Malignant Astrocytic Glioma: Genetics, Biology, and Paths to Treatment. Genes Dev. 2007, 21, 2683–2710. [Google Scholar] [CrossRef]
  31. Atkins, R.J.; Ng, W.; Stylli, S.S.; Hovens, C.M.; Kaye, A.H. Repair Mechanisms Help Glioblastoma Resist Treatment. J. Clin. Neurosci. 2015, 22, 14–20. [Google Scholar] [CrossRef]
  32. Yalamarty, S.S.K.; Filipczak, N.; Li, X.; Subhan, M.A.; Parveen, F.; Ataide, J.A.; Rajmalani, B.A.; Torchilin, V.P. Mechanisms of Resistance and Current Treatment Options for Glioblastoma Multiforme (GBM). Cancers 2023, 15, 2116. [Google Scholar] [CrossRef]
  33. Eckerdt, F.; Platanias, L.C. Emerging Role of Glioma Stem Cells in Mechanisms of Therapy Resistance. Cancers 2023, 15, 3458. [Google Scholar] [CrossRef] [PubMed]
  34. Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma Stem Cells Promote Radioresistance by Preferential Activation of the DNA Damage Response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
  35. Verhaak, R.G.W.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; et al. Integrated Genomic Analysis Identifies Clinically Relevant Subtypes of Glioblastoma Characterized by Abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef] [PubMed]
  36. Steponaitis, G.; Tamasauskas, A. Mesenchymal and Proneural Subtypes of Glioblastoma Disclose Branching Based on GSC Associated Signature. Int. J. Mol. Sci. 2021, 22, 4964. [Google Scholar] [CrossRef] [PubMed]
  37. Fayzullin, A.; Sandberg, C.J.; Spreadbury, M.; Saberniak, B.M.; Grieg, Z.; Skaga, E.; Langmoen, I.A.; Vik-Mo, E.O. Phenotypic and Expressional Heterogeneity in the Invasive Glioma Cells. Transl. Oncol. 2019, 12, 122–133. [Google Scholar] [CrossRef]
  38. Seliger, C.; Meyer, A.-L.; Leidgens, V.; Rauer, L.; Moeckel, S.; Jachnik, B.; Proske, J.; Dettmer, K.; Rothhammer-Hampl, T.; Kaulen, L.D.; et al. Metabolic Heterogeneity of Brain Tumor Cells of Proneural and Mesenchymal Origin. Int. J. Mol. Sci. 2022, 23, 11629. [Google Scholar] [CrossRef]
  39. Ah-Pine, F.; Khettab, M.; Bedoui, Y.; Slama, Y.; Daniel, M.; Doray, B.; Gasque, P. On the Origin and Development of Glioblastoma: Multifaceted Role of Perivascular Mesenchymal Stromal Cells. Acta Neuropathol. Commun. 2023, 11, 104. [Google Scholar] [CrossRef]
  40. Teo, W.-Y.; Sekar, K.; Seshachalam, P.; Shen, J.; Chow, W.-Y.; Lau, C.C.; Yang, H.; Park, J.; Kang, S.-G.; Li, X.; et al. Relevance of a TCGA-Derived Glioblastoma Subtype Gene-Classifier among Patient Populations. Sci. Rep. 2019, 9, 7442. [Google Scholar] [CrossRef]
  41. Uribe, D.; Niechi, I.; Rackov, G.; Erices, J.I.; San Martín, R.; Quezada, C. Adapt to Persist: Glioblastoma Microenvironment and Epigenetic Regulation on Cell Plasticity. Biology 2022, 11, 313. [Google Scholar] [CrossRef]
  42. Lee, E.; Yong, R.L.; Paddison, P.; Zhu, J. Comparison of Glioblastoma (GBM) Molecular Classification Methods. Semin. Cancer Biol. 2018, 53, 201–211. [Google Scholar] [CrossRef]
  43. Wang, Z.; Sun, D.; Chen, Y.-J.; Xie, X.; Shi, Y.; Tabar, V.; Brennan, C.W.; Bale, T.A.; Jayewickreme, C.D.; Laks, D.R.; et al. Cell Lineage-Based Stratification for Glioblastoma. Cancer Cell 2020, 38, 366–379.e8. [Google Scholar] [CrossRef]
  44. Yanovich-Arad, G.; Ofek, P.; Yeini, E.; Mardamshina, M.; Danilevsky, A.; Shomron, N.; Grossman, R.; Satchi-Fainaro, R.; Geiger, T. Proteogenomics of Glioblastoma Associates Molecular Patterns with Survival. Cell Rep. 2021, 34, 108787. [Google Scholar] [CrossRef]
  45. Steponaitis, G.; Kucinskas, V.; Golubickaite, I.; Skauminas, K.; Saudargiene, A. Glioblastoma Molecular Classification Tool Based on mRNA Analysis: From Wet-Lab to Subtype. Int. J. Mol. Sci. 2022, 23, 15875. [Google Scholar] [CrossRef]
  46. Grech, N.; Dalli, T.; Mizzi, S.; Meilak, L.; Calleja, N.; Zrinzo, A. Rising Incidence of Glioblastoma Multiforme in a Well-Defined Population. Cureus 2020, 12, e8195. [Google Scholar] [CrossRef] [PubMed]
  47. Tan, A.C.; Ashley, D.M.; López, G.Y.; Malinzak, M.; Friedman, H.S.; Khasraw, M. Management of Glioblastoma: State of the Art and Future Directions. CA A Cancer J. Clin. 2020, 70, 299–312. [Google Scholar] [CrossRef] [PubMed]
  48. Carrano, A.; Juarez, J.J.; Incontri, D.; Ibarra, A.; Cazares, H.G. Sex-Specific Differences in Glioblastoma. Cells 2021, 10, 1783. [Google Scholar] [CrossRef] [PubMed]
  49. Ohgaki, H. Epidemiology of Brain Tumors. In Cancer Epidemiology: Modifiable Factors; Verma, M., Ed.; Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2009; pp. 323–342. ISBN 978-1-60327-492-0. [Google Scholar]
  50. Lopes, J.; Baudin, C.; Leuraud, K.; Klokov, D.; Bernier, M.-O. Ionizing Radiation Exposure during Adulthood and Risk of Developing Central Nervous System Tumors: Systematic Review and Meta-Analysis. Sci. Rep. 2022, 12, 16209. [Google Scholar] [CrossRef] [PubMed]
  51. Dadey, D.Y.A.; Medress, Z.A.; Sharma, M.; Ugiliweneza, B.; Wang, D.; Rodrigues, A.; Parker, J.; Burton, E.; Williams, B.; Han, S.S.; et al. Risk of Developing Glioblastoma Following Non-CNS Primary Cancer: A SEER Analysis between 2000 and 2018. J. Neurooncol. 2023, 164, 655–662. [Google Scholar] [CrossRef] [PubMed]
  52. Ostrom, Q.T.; Patil, N.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. Corrigendum to: CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013-2017. Neuro Oncol. 2020, noaa269. [Google Scholar] [CrossRef] [PubMed]
  53. Wick, W.; Chinot, O.L.; Bendszus, M.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Revil, C.; Kerloeguen, Y.; Cloughesy, T. Evaluation of Pseudoprogression Rates and Tumor Progression Patterns in a Phase III Trial of Bevacizumab plus Radiotherapy/Temozolomide for Newly Diagnosed Glioblastoma. Neuro Oncol. 2016, 18, 1434–1441. [Google Scholar] [CrossRef] [PubMed]
  54. Okada, H.; Weller, M.; Huang, R.; Finocchiaro, G.; Gilbert, M.R.; Wick, W.; Ellingson, B.M.; Hashimoto, N.; Pollack, I.F.; Brandes, A.A.; et al. Immunotherapy Response Assessment in Neuro-Oncology (iRANO): A Report of the RANO Working Group. Lancet Oncol. 2015, 16, e534–e542. [Google Scholar] [CrossRef]
  55. Leao, D.J.; Craig, P.G.; Godoy, L.F.; Leite, C.C.; Policeni, B. Response Assessment in Neuro-Oncology Criteria for Gliomas: Practical Approach Using Conventional and Advanced Techniques. AJNR Am. J. Neuroradiol. 2020, 41, 10–20. [Google Scholar] [CrossRef]
  56. Hooper, G.W.; Ansari, S.; Johnson, J.M.; Ginat, D.T. Advances in the Radiological Evaluation of and Theranostics for Glioblastoma. Cancers 2023, 15, 4162. [Google Scholar] [CrossRef]
  57. Soni, N.; Ora, M.; Jena, A.; Rana, P.; Mangla, R.; Ellika, S.; Almast, J.; Puri, S.; Meyers, S.P. Amino Acid Tracer PET MRI in Glioma Management: What a Neuroradiologist Needs to Know. Am. J. Neuroradiol. 2023. [Google Scholar] [CrossRef] [PubMed]
  58. Reeves, K.M.; Song, P.N.; Angermeier, A.; Manna, D.D.; Li, Y.; Wang, J.; Yang, E.S.; Sorace, A.G.; Larimer, B.M. 18F-FMISO PET Imaging Identifies Hypoxia and Immunosuppressive Tumor Microenvironments and Guides Targeted Evofosfamide Therapy in Tumors Refractory to PD-1 and CTLA-4 Inhibition. Clin. Cancer Res. 2022, 28, 327–337. [Google Scholar] [CrossRef]
  59. Ma, D.K.; Bonaguidi, M.A.; Ming, G.; Song, H. Adult Neural Stem Cells in the Mammalian Central Nervous System. Cell Res. 2009, 19, 672–682. [Google Scholar] [CrossRef]
  60. Lee, J.H.; Lee, J.E.; Kahng, J.Y.; Kim, S.H.; Park, J.S.; Yoon, S.J.; Um, J.-Y.; Kim, W.K.; Lee, J.-K.; Park, J.; et al. Human Glioblastoma Arises from Subventricular Zone Cells with Low-Level Driver Mutations. Nature 2018, 560, 243–247. [Google Scholar] [CrossRef]
  61. Altmann, C.; Keller, S.; Schmidt, M.H.H. The Role of SVZ Stem Cells in Glioblastoma. Cancers 2019, 11, 448. [Google Scholar] [CrossRef] [PubMed]
  62. Kwan, K.; Schneider, J.R.; Patel, N.V.; Boockvar, J.A. Tracing the Origin of Glioblastoma: Subventricular Zone Neural Stem Cells. Neurosurgery 2019, 84, E15–E16. [Google Scholar] [CrossRef]
  63. Hira, V.V.V.; Molenaar, R.J.; Breznik, B.; Lah, T.; Aronica, E.; Van Noorden, C.J.F. Immunohistochemical Detection of Neural Stem Cells and Glioblastoma Stem Cells in the Subventricular Zone of Glioblastoma Patients. J. Histochem. Cytochem. 2021, 69, 349–364. [Google Scholar] [CrossRef]
  64. Beiriger, J.; Habib, A.; Jovanovich, N.; Kodavali, C.V.; Edwards, L.; Amankulor, N.; Zinn, P.O. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front. Oncol. 2022, 12, 790976. [Google Scholar] [CrossRef] [PubMed]
  65. Loras, A.; Gonzalez-Bonet, L.G.; Gutierrez-Arroyo, J.L.; Martinez-Cadenas, C.; Marques-Torrejon, M.A. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life 2023, 13, 905. [Google Scholar] [CrossRef] [PubMed]
  66. Matarredona, E.R.; Pastor, A.M. Neural Stem Cells of the Subventricular Zone as the Origin of Human Glioblastoma Stem Cells. Therapeutic Implications. Front. Oncol. 2019, 9, 779. [Google Scholar] [CrossRef] [PubMed]
  67. Yamaki, T.; Shibahra, I.; Matsuda, K.-I.; Kanemura, Y.; Konta, T.; Kanamori, M.; Yamakawa, M.; Tominaga, T.; Sonoda, Y. Relationships between Recurrence Patterns and Subventricular Zone Involvement or CD133 Expression in Glioblastoma. J. Neurooncol. 2020, 146, 489–499. [Google Scholar] [CrossRef] [PubMed]
  68. Brockman, A.A.; Mobley, B.C.; Ihrie, R.A. Histological Studies of the Ventricular-Subventricular Zone as Neural Stem Cell and Glioma Stem Cell Niche. J. Histochem. Cytochem. 2021, 69, 819–834. [Google Scholar] [CrossRef]
  69. Mistry, A.M.; Hale, A.T.; Chambless, L.B.; Weaver, K.D.; Thompson, R.C.; Ihrie, R.A. Influence of Glioblastoma Contact with the Lateral Ventricle on Survival: A Meta-Analysis. J. Neurooncol. 2017, 131, 125–133. [Google Scholar] [CrossRef]
  70. Huang, R.; Wang, T.; Liao, Z.; Wang, Z.; Ye, M.; Zhou, D.; Xie, H.; Bai, Y.; Qiu, Y.; Liu, Y. A Retrospective Analysis of the Risk Factors Affecting Recurrence Time in Patients with Recurrent Glioblastoma. Ann. Palliat. Med. 2021, 10, 5391–5399. [Google Scholar] [CrossRef]
  71. Li, S.; Dong, L.; Pan, Z.; Yang, G. Targeting the Neural Stem Cells in Subventricular Zone for the Treatment of Glioblastoma: An Update from Preclinical Evidence to Clinical Interventions. Stem Cell Res. Ther. 2023, 14, 125. [Google Scholar] [CrossRef]
  72. Friedmann-Morvinski, D.; Bushong, E.A.; Ke, E.; Soda, Y.; Marumoto, T.; Singer, O.; Ellisman, M.H.; Verma, I.M. Dedifferentiation of Neurons and Astrocytes by Oncogenes Can Induce Gliomas in Mice. Science 2012, 338, 1080–1084. [Google Scholar] [CrossRef]
  73. Shahar, T.; Rozovski, U.; Hess, K.R.; Hossain, A.; Gumin, J.; Gao, F.; Fuller, G.N.; Goodman, L.; Sulman, E.P.; Lang, F.F. Percentage of Mesenchymal Stem Cells in High-Grade Glioma Tumor Samples Correlates with Patient Survival. Neuro Oncol. 2017, 19, 660–668. [Google Scholar] [CrossRef]
  74. Uhrbom, L.; Dai, C.; Celestino, J.C.; Rosenblum, M.K.; Fuller, G.N.; Holland, E.C. Ink4a-Arf Loss Cooperates with KRas Activation in Astrocytes and Neural Progenitors to Generate Glioblastomas of Various Morphologies Depending on Activated Akt1. Cancer Res. 2002, 62, 5551–5558. [Google Scholar]
  75. Li, F.; Liu, X.; Sampson, J.H.; Bigner, D.D.; Li, C.-Y. Rapid Reprogramming of Primary Human Astrocytes into Potent Tumor-Initiating Cells with Defined Genetic Factors. Cancer Res. 2016, 76, 5143–5150. [Google Scholar] [CrossRef]
  76. Bradshaw, A.; Wickremsekera, A.; Tan, S.T.; Peng, L.; Davis, P.F.; Itinteang, T. Cancer Stem Cell Hierarchy in Glioblastoma Multiforme. Front. Surg. 2016, 3, 21. [Google Scholar] [CrossRef]
  77. Lan, X.; Jörg, D.J.; Cavalli, F.M.G.; Richards, L.M.; Nguyen, L.V.; Vanner, R.J.; Guilhamon, P.; Lee, L.; Kushida, M.M.; Pellacani, D.; et al. Fate Mapping of Human Glioblastoma Reveals an Invariant Stem Cell Hierarchy. Nature 2017, 549, 227–232. [Google Scholar] [CrossRef]
  78. Schneider, M.; Ströbele, S.; Nonnenmacher, L.; Siegelin, M.D.; Tepper, M.; Stroh, S.; Hasslacher, S.; Enzenmüller, S.; Strauss, G.; Baumann, B.; et al. A Paired Comparison between Glioblastoma “Stem Cells” and Differentiated Cells. Int. J. Cancer 2016, 138, 1709–1718. [Google Scholar] [CrossRef]
  79. Liau, B.B.; Sievers, C.; Donohue, L.K.; Gillespie, S.M.; Flavahan, W.A.; Miller, T.E.; Venteicher, A.S.; Hebert, C.H.; Carey, C.D.; Rodig, S.J.; et al. Adaptive Chromatin Remodeling Drives Glioblastoma Stem Cell Plasticity and Drug Tolerance. Cell Stem Cell 2017, 20, 233–246.e7. [Google Scholar] [CrossRef]
  80. Gimple, R.C.; Bhargava, S.; Dixit, D.; Rich, J.N. Glioblastoma Stem Cells: Lessons from the Tumor Hierarchy in a Lethal Cancer. Genes Dev. 2019, 33, 591–609. [Google Scholar] [CrossRef]
  81. Glumac, P.M.; LeBeau, A.M. The Role of CD133 in Cancer: A Concise Review. Clin. Transl. Med. 2018, 7, 18. [Google Scholar] [CrossRef]
  82. Si, D.; Yin, F.; Peng, J.; Zhang, G. High Expression of CD44 Predicts a Poor Prognosis in Glioblastomas. Cancer Manag. Res. 2020, 12, 769–775. [Google Scholar] [CrossRef]
  83. Dréan, A.; Rosenberg, S.; Lejeune, F.-X.; Goli, L.; Nadaradjane, A.A.; Guehennec, J.; Schmitt, C.; Verreault, M.; Bielle, F.; Mokhtari, K.; et al. ATP Binding Cassette (ABC) Transporters: Expression and Clinical Value in Glioblastoma. J. Neurooncol. 2018, 138, 479–486. [Google Scholar] [CrossRef]
  84. Vieira de Castro, J.; Gonçalves, C.S.; Hormigo, A.; Costa, B.M. Exploiting the Complexities of Glioblastoma Stem Cells: Insights for Cancer Initiation and Therapeutic Targeting. Int. J. Mol. Sci. 2020, 21, 5278. [Google Scholar] [CrossRef]
  85. Zhang, G.-L.; Wang, C.-F.; Qian, C.; Ji, Y.-X.; Wang, Y.-Z. Role and Mechanism of Neural Stem Cells of the Subventricular Zone in Glioblastoma. World J. Stem Cells 2021, 13, 877–893. [Google Scholar] [CrossRef]
  86. Soda, Y.; Marumoto, T.; Friedmann-Morvinski, D.; Soda, M.; Liu, F.; Michiue, H.; Pastorino, S.; Yang, M.; Hoffman, R.M.; Kesari, S.; et al. Transdifferentiation of Glioblastoma Cells into Vascular Endothelial Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4274–4280. [Google Scholar] [CrossRef]
  87. Garner, J.M.; Ellison, D.W.; Finkelstein, D.; Ganguly, D.; Du, Z.; Sims, M.; Yang, C.H.; Interiano, R.B.; Davidoff, A.M.; Pfeffer, L.M. Molecular Heterogeneity in a Patient-Derived Glioblastoma Xenoline Is Regulated by Different Cancer Stem Cell Populations. PLoS ONE 2015, 10, e0125838. [Google Scholar] [CrossRef]
  88. Matarredona, E.R.; Talaverón, R.; Pastor, A.M. Interactions Between Neural Progenitor Cells and Microglia in the Subventricular Zone: Physiological Implications in the Neurogenic Niche and After Implantation in the Injured Brain. Front. Cell Neurosci. 2018, 12, 268. [Google Scholar] [CrossRef]
  89. David-Bercholz, J.; Kuo, C.T.; Deneen, B. Astrocyte and Oligodendrocyte Responses From the Subventricular Zone After Injury. Front. Cell Neurosci. 2021, 15, 797553. [Google Scholar] [CrossRef]
  90. Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour Vascularization via Endothelial Differentiation of Glioblastoma Stem-like Cells. Nature 2010, 468, 824–828. [Google Scholar] [CrossRef]
  91. Cheng, L.; Huang, Z.; Zhou, W.; Wu, Q.; Donnola, S.; Liu, J.K.; Fang, X.; Sloan, A.E.; Mao, Y.; Lathia, J.D.; et al. Glioblastoma Stem Cells Generate Vascular Pericytes to Support Vessel Function and Tumor Growth. Cell 2013, 153, 139–152. [Google Scholar] [CrossRef]
  92. Molina, J.R.; Hayashi, Y.; Stephens, C.; Georgescu, M.-M. Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation. Neoplasia 2010, 12, 453–463. [Google Scholar] [CrossRef]
  93. Masliantsev, K.; Pinel, B.; Balbous, A.; Guichet, P.-O.; Tachon, G.; Milin, S.; Godet, J.; Duchesne, M.; Berger, A.; Petropoulos, C.; et al. Impact of STAT3 Phosphorylation in Glioblastoma Stem Cells Radiosensitization and Patient Outcome. Oncotarget 2018, 9, 3968–3979. [Google Scholar] [CrossRef]
  94. Beier, D.; Schriefer, B.; Brawanski, K.; Hau, P.; Weis, J.; Schulz, J.B.; Beier, C.P. Efficacy of Clinically Relevant Temozolomide Dosing Schemes in Glioblastoma Cancer Stem Cell Lines. J. Neurooncol. 2012, 109, 45–52. [Google Scholar] [CrossRef]
  95. McCord, M.; Bartom, E.; Burdett, K.; Baran, A.; Eckerdt, F.D.; Balyasnikova, I.V.; McCortney, K.; Sears, T.; Cheng, S.-Y.; Sarkaria, J.N.; et al. Modeling Therapy-Driven Evolution of Glioblastoma with Patient-Derived Xenografts. Cancers 2022, 14, 5494. [Google Scholar] [CrossRef]
  96. Pavlova, G.; Belyashova, A.; Savchenko, E.; Panteleev, D.; Shamadykova, D.; Nikolaeva, A.; Pavlova, S.; Revishchin, A.; Golbin, D.; Potapov, A.; et al. Reparative Properties of Human Glioblastoma Cells after Single Exposure to a Wide Range of X-Ray Doses. Front. Oncol. 2022, 12, 912741. [Google Scholar] [CrossRef]
  97. Vilar, J.B.; Christmann, M.; Tomicic, M.T. Alterations in Molecular Profiles Affecting Glioblastoma Resistance to Radiochemotherapy: Where Does the Good Go? Cancers 2022, 14, 2416. [Google Scholar] [CrossRef] [PubMed]
  98. Ahmed, S.U.; Carruthers, R.; Gilmour, L.; Yildirim, S.; Watts, C.; Chalmers, A.J. Selective Inhibition of Parallel DNA Damage Response Pathways Optimizes Radiosensitization of Glioblastoma Stem-like Cells. Cancer Res. 2015, 75, 4416–4428. [Google Scholar] [CrossRef]
  99. Dean, M.; Fojo, T.; Bates, S. Tumour Stem Cells and Drug Resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
  100. Tachon, G.; Cortes, U.; Guichet, P.-O.; Rivet, P.; Balbous, A.; Masliantsev, K.; Berger, A.; Boissonnade, O.; Wager, M.; Karayan-Tapon, L. Cell Cycle Changes after Glioblastoma Stem Cell Irradiation: The Major Role of RAD51. Int. J. Mol. Sci. 2018, 19, 3018. [Google Scholar] [CrossRef]
  101. Lau, J.; Ilkhanizadeh, S.; Wang, S.; Miroshnikova, Y.A.; Salvatierra, N.A.; Wong, R.A.; Schmidt, C.; Weaver, V.M.; Weiss, W.A.; Persson, A.I. STAT3 Blockade Inhibits Radiation-Induced Malignant Progression in Glioma. Cancer Res. 2015, 75, 4302–4311. [Google Scholar] [CrossRef]
  102. Sherry, M.M.; Reeves, A.; Wu, J.K.; Cochran, B.H. STAT3 Is Required for Proliferation and Maintenance of Multipotency in Glioblastoma Stem Cells. Stem Cells 2009, 27, 2383–2392. [Google Scholar] [CrossRef] [PubMed]
  103. Ganguly, D.; Fan, M.; Yang, C.H.; Zbytek, B.; Finkelstein, D.; Roussel, M.F.; Pfeffer, L.M. The Critical Role That STAT3 Plays in Glioma-Initiating Cells: STAT3 Addiction in Glioma. Oncotarget 2018, 9, 22095–22112. [Google Scholar] [CrossRef] [PubMed]
  104. Wang, Y.; Yang, C.; Sims, M.M.; Sacher, J.R.; Raje, M.; Deokar, H.; Yue, P.; Turkson, J.; Buolamwini, J.K.; Pfeffer, L.M. SS-4 Is a Highly Selective Small Molecule Inhibitor of STAT3 Tyrosine Phosphorylation That Potently Inhibits GBM Tumorigenesis in Vitro and in Vivo. Cancer Lett. 2022, 533, 215614. [Google Scholar] [CrossRef]
  105. Groot, J.d.; Ott, M.; Wei, J.; Kassab, C.; Fang, D.; Najem, H.; O’Brien, B.; Weathers, S.-P.; Matsouka, C.K.; Majd, N.K.; et al. A First-in-Human Phase I Trial of the Oral p-STAT3 Inhibitor WP1066 in Patients with Recurrent Malignant Glioma. CNS Oncol. 2022, 11, CNS87. [Google Scholar] [CrossRef] [PubMed]
  106. Sharma, P.; Aaroe, A.; Liang, J.; Puduvalli, V.K. Tumor Microenvironment in Glioblastoma: Current and Emerging Concepts. Neurooncol. Adv. 2023, 5, vdad009. [Google Scholar] [CrossRef]
  107. Rocha Pinheiro, S.L.; Lemos, F.F.B.; Marques, H.S.; Silva Luz, M.; de Oliveira Silva, L.G.; Faria Souza Mendes dos Santos, C.; da Costa Evangelista, K.; Calmon, M.S.; Sande Loureiro, M.; Freire de Melo, F. Immunotherapy in Glioblastoma Treatment: Current State and Future Prospects. World J. Clin. Oncol. 2023, 14, 138–159. [Google Scholar] [CrossRef] [PubMed]
  108. Otvos, B.; Silver, D.J.; Mulkearns-Hubert, E.E.; Alvarado, A.G.; Turaga, S.M.; Sorensen, M.D.; Rayman, P.; Flavahan, W.A.; Hale, J.S.; Stoltz, K.; et al. Cancer Stem Cell-Secreted Macrophage Migration Inhibitory Factor Stimulates Myeloid Derived Suppressor Cell Function and Facilitates Glioblastoma Immune Evasion. Stem Cells 2016, 34, 2026–2039. [Google Scholar] [CrossRef]
  109. Zhou, W.; Ke, S.Q.; Huang, Z.; Flavahan, W.; Fang, X.; Paul, J.; Wu, L.; Sloan, A.E.; McLendon, R.E.; Li, X.; et al. Periostin Secreted by Glioblastoma Stem Cells Recruits M2 Tumour-Associated Macrophages and Promotes Malignant Growth. Nat. Cell Biol. 2015, 17, 170–182. [Google Scholar] [CrossRef]
  110. Broekman, M.L.; Maas, S.L.N.; Abels, E.R.; Mempel, T.R.; Krichevsky, A.M.; Breakefield, X.O. Multidimensional Communication in the Microenvirons of Glioblastoma. Nat. Rev. Neurol. 2018, 14, 482–495. [Google Scholar] [CrossRef]
  111. Wei, J.; Marisetty, A.; Schrand, B.; Gabrusiewicz, K.; Hashimoto, Y.; Ott, M.; Grami, Z.; Kong, L.-Y.; Ling, X.; Caruso, H.; et al. Osteopontin Mediates Glioblastoma-Associated Macrophage Infiltration and Is a Potential Therapeutic Target. J. Clin. Investig. 2019, 129, 137–149. [Google Scholar] [CrossRef]
  112. Ricklefs, F.L.; Alayo, Q.; Krenzlin, H.; Mahmoud, A.B.; Speranza, M.C.; Nakashima, H.; Hayes, J.L.; Lee, K.; Balaj, L.; Passaro, C.; et al. Immune Evasion Mediated by PD-L1 on Glioblastoma-Derived Extracellular Vesicles. Sci. Adv. 2018, 4, eaar2766. [Google Scholar] [CrossRef]
  113. Himes, B.T.; Peterson, T.E.; de Mooij, T.; Garcia, L.M.C.; Jung, M.-Y.; Uhm, S.; Yan, D.; Tyson, J.; Jin-Lee, H.J.; Parney, D.; et al. The Role of Extracellular Vesicles and PD-L1 in Glioblastoma-Mediated Immunosuppressive Monocyte Induction. Neuro Oncol. 2020, 22, 967–978. [Google Scholar] [CrossRef]
  114. Garcia, J.H.; Jain, S.; Aghi, M.K. Metabolic Drivers of Invasion in Glioblastoma. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
  115. Erices, J.I.; Bizama, C.; Niechi, I.; Uribe, D.; Rosales, A.; Fabres, K.; Navarro-Martínez, G.; Torres, Á.; San Martín, R.; Roa, J.C.; et al. Glioblastoma Microenvironment and Invasiveness: New Insights and Therapeutic Targets. Int. J. Mol. Sci. 2023, 24, 7047. [Google Scholar] [CrossRef]
  116. Hoang-Minh, L.B.; Siebzehnrubl, F.A.; Yang, C.; Suzuki-Hatano, S.; Dajac, K.; Loche, T.; Andrews, N.; Schmoll Massari, M.; Patel, J.; Amin, K.; et al. Infiltrative and Drug-Resistant Slow-Cycling Cells Support Metabolic Heterogeneity in Glioblastoma. EMBO J. 2018, 37, e98772. [Google Scholar] [CrossRef] [PubMed]
  117. Verdugo, E.; Puerto, I.; Medina, M.Á. An Update on the Molecular Biology of Glioblastoma, with Clinical Implications and Progress in Its Treatment. Cancer Commun. 2022, 42, 1083–1111. [Google Scholar] [CrossRef] [PubMed]
  118. Stanke, K.M.; Wilson, C.; Kidambi, S. High Expression of Glycolytic Genes in Clinical Glioblastoma Patients Correlates With Lower Survival. Front. Mol. Biosci. 2021, 8. [Google Scholar] [CrossRef]
  119. Flavahan, W.A.; Wu, Q.; Hitomi, M.; Rahim, N.; Kim, Y.; Sloan, A.E.; Weil, R.J.; Nakano, I.; Sarkaria, J.N.; Stringer, B.W.; et al. Brain Tumor Initiating Cells Adapt to Restricted Nutrition through Preferential Glucose Uptake. Nat. Neurosci. 2013, 16, 1373–1382. [Google Scholar] [CrossRef] [PubMed]
  120. Libby, C.J.; Gc, S.; Benavides, G.A.; Fisher, J.L.; Williford, S.E.; Zhang, S.; Tran, A.N.; Gordon, E.R.; Jones, A.B.; Tuy, K.; et al. A Role for GLUT3 in Glioblastoma Cell Invasion That Is Not Recapitulated by GLUT1. Cell Adh Migr. 2021, 15, 101–115. [Google Scholar] [CrossRef]
  121. Wolf, A.; Agnihotri, S.; Micallef, J.; Mukherjee, J.; Sabha, N.; Cairns, R.; Hawkins, C.; Guha, A. Hexokinase 2 Is a Key Mediator of Aerobic Glycolysis and Promotes Tumor Growth in Human Glioblastoma Multiforme. J. Exp. Med. 2011, 208, 313–326. [Google Scholar] [CrossRef]
  122. Labak, C.M.; Wang, P.Y.; Arora, R.; Guda, M.R.; Asuthkar, S.; Tsung, A.J.; Velpula, K.K. Glucose Transport: Meeting the Metabolic Demands of Cancer, and Applications in Glioblastoma Treatment. Am. J. Cancer Res. 2016, 6, 1599–1608. [Google Scholar] [PubMed]
  123. Kou, Y.; Geng, F.; Guo, D. Lipid Metabolism in Glioblastoma: From De Novo Synthesis to Storage. Biomedicines 2022, 10, 1943. [Google Scholar] [CrossRef] [PubMed]
  124. Obara-Michlewska, M.; Szeliga, M. Targeting Glutamine Addiction in Gliomas. Cancers 2020, 12, 310. [Google Scholar] [CrossRef] [PubMed]
  125. Natarajan, S.K.; Venneti, S. Glutamine Metabolism in Brain Tumors. Cancers 2019, 11, 1628. [Google Scholar] [CrossRef] [PubMed]
  126. Barzegar Behrooz, A.; Talaie, Z.; Jusheghani, F.; Łos, M.J.; Klonisch, T.; Ghavami, S. Wnt and PI3K/Akt/mTOR Survival Pathways as Therapeutic Targets in Glioblastoma. Int. J. Mol. Sci. 2022, 23, 1353. [Google Scholar] [CrossRef]
  127. Garcia-Bermudez, J.; Baudrier, L.; La, K.; Zhu, X.G.; Fidelin, J.; Sviderskiy, V.O.; Papagiannakopoulos, T.; Molina, H.; Snuderl, M.; Lewis, C.A.; et al. Aspartate Is a Limiting Metabolite for Cancer Cell Proliferation under Hypoxia and in Tumours. Nat. Cell Biol. 2018, 20, 775–781. [Google Scholar] [CrossRef]
  128. Karatsai, O.; Stasyk, O.; Redowicz, M.J. Effects of Arginine and Its Deprivation on Human Glioblastoma Physiology and Signaling. Adv. Exp. Med. Biol. 2020, 1202, 243–258. [Google Scholar] [CrossRef]
  129. Kesarwani, P.; Prabhu, A.; Kant, S.; Kumar, P.; Graham, S.F.; Buelow, K.L.; Wilson, G.D.; Miller, C.R.; Chinnaiyan, P. Tryptophan Metabolism Contributes to Radiation-Induced Immune Checkpoint Reactivation in Glioblastoma. Clin. Cancer Res. 2018, 24, 3632–3643. [Google Scholar] [CrossRef]
  130. Lyons, S.A.; Chung, W.J.; Weaver, A.K.; Ogunrinu, T.; Sontheimer, H. Autocrine Glutamate Signaling Promotes Glioma Cell Invasion. Cancer Res. 2007, 67, 9463–9471. [Google Scholar] [CrossRef]
  131. Corsi, L.; Mescola, A.; Alessandrini, A. Glutamate Receptors and Glioblastoma Multiforme: An Old “Route” for New Perspectives. Int. J. Mol. Sci. 2019, 20, 1796. [Google Scholar] [CrossRef]
  132. Savaskan, N.E.; Fan, Z.; Broggini, T.; Buchfelder, M.; Eyüpoglu, I.Y. Neurodegeneration in the Brain Tumor Microenvironment: Glutamate in the Limelight. Curr. Neuropharmacol. 2015, 13, 258–265. [Google Scholar] [CrossRef] [PubMed]
  133. Kumaria, A.; Ashkan, K. Novel Therapeutic Strategies in Glioma Targeting Glutamatergic Neurotransmission. Brain Res. 2023, 1818, 148515. [Google Scholar] [CrossRef] [PubMed]
  134. Maule, F.; Bresolin, S.; Rampazzo, E.; Boso, D.; Puppa, A.D.; Esposito, G.; Porcù, E.; Mitola, S.; Lombardi, G.; Accordi, B.; et al. Annexin 2A Sustains Glioblastoma Cell Dissemination and Proliferation. Oncotarget 2016, 7, 54632–54649. [Google Scholar] [CrossRef] [PubMed]
  135. Yu, S.; Yu, X.; Sun, L.; Zheng, Y.; Chen, L.; Xu, H.; Jin, J.; Lan, Q.; Chen, C.C.; Li, M. GBP2 Enhances Glioblastoma Invasion through Stat3/Fibronectin Pathway. Oncogene 2020, 39, 5042–5055. [Google Scholar] [CrossRef] [PubMed]
  136. Wu, S.; Liu, C.; Wei, X.; Nong, W.-X.; Lin, L.-N.; Li, F.; Xie, X.-X.; Liao, X.-S.; Luo, B.; Zhang, Q.-M.; et al. High Expression of Fibronectin 1 Predicts a Poor Prognosis in Glioblastoma. Curr. Med. Sci. 2022, 42, 1055–1065. [Google Scholar] [CrossRef] [PubMed]
  137. de Semir, D.; Bezrookove, V.; Nosrati, M.; Scanlon, K.R.; Singer, E.; Judkins, J.; Rieken, C.; Wu, C.; Shen, J.; Schmudermayer, C.; et al. PHIP Drives Glioblastoma Motility and Invasion by Regulating the Focal Adhesion Complex. Proc. Natl. Acad. Sci. USA 2020, 117, 9064–9073. [Google Scholar] [CrossRef]
  138. Cosset, É.; Ilmjärv, S.; Dutoit, V.; Elliott, K.; von Schalscha, T.; Camargo, M.F.; Reiss, A.; Moroishi, T.; Seguin, L.; Gomez, G.; et al. Glut3 Addiction Is a Druggable Vulnerability for a Molecularly Defined Subpopulation of Glioblastoma. Cancer Cell 2017, 32, 856–868.e5. [Google Scholar] [CrossRef]
  139. Santoni, G.; Santoni, M.; Nabissi, M. Functional Role of T-Type Calcium Channels in Tumour Growth and Progression: Prospective in Cancer Therapy. Br. J. Pharmacol. 2012, 166, 1244–1246. [Google Scholar] [CrossRef]
  140. Jacquemet, G.; Baghirov, H.; Georgiadou, M.; Sihto, H.; Peuhu, E.; Cettour-Janet, P.; He, T.; Perälä, M.; Kronqvist, P.; Joensuu, H.; et al. L-Type Calcium Channels Regulate Filopodia Stability and Cancer Cell Invasion Downstream of Integrin Signalling. Nat. Commun. 2016, 7, 13297. [Google Scholar] [CrossRef]
  141. Portela, M.; Casas-Tintó, S. New Cellular Dimensions on Glioblastoma Progression. Neurosci. Insights 2020, 15, 2633105520923076. [Google Scholar] [CrossRef]
  142. Kuga, T.; Sadoshima, J.; Tomoike, H.; Kanaide, H.; Akaike, N.; Nakamura, M. Actions of Ca2+ Antagonists on Two Types of Ca2+ Channels in Rat Aorta Smooth Muscle Cells in Primary Culture. Circ. Res. 1990, 67, 469–480. [Google Scholar] [CrossRef]
  143. Pointer, K.B.; Clark, P.A.; Eliceiri, K.W.; Salamat, M.S.; Robertson, G.A.; Kuo, J.S. Administration of Non-Torsadogenic Human Ether-à-Go-Go-Related Gene Inhibitors Is Associated with Better Survival for High hERG-Expressing Glioblastoma Patients. Clin. Cancer Res. 2017, 23, 73–80. [Google Scholar] [CrossRef]
  144. Chen, D.; Song, M.; Mohamad, O.; Yu, S.P. Inhibition of Na+/K+-ATPase Induces Hybrid Cell Death and Enhanced Sensitivity to Chemotherapy in Human Glioblastoma Cells. BMC Cancer 2014, 14, 716. [Google Scholar] [CrossRef]
  145. Dong, Z.; Cui, H. Epigenetic Modulation of Metabolism in Glioblastoma. Semin. Cancer Biol. 2019, 57, 45–51. [Google Scholar] [CrossRef] [PubMed]
  146. Uddin, M.S.; Mamun, A.A.; Alghamdi, B.S.; Tewari, D.; Jeandet, P.; Sarwar, M.S.; Ashraf, G.M. Epigenetics of Glioblastoma Multiforme: From Molecular Mechanisms to Therapeutic Approaches. Semin. Cancer Biol. 2022, 83, 100–120. [Google Scholar] [CrossRef] [PubMed]
  147. Zhao, D. Single Nucleotide Alterations in MicroRNAs and Human Cancer-A Not Fully Explored Field. Non-Coding RNA Res. 2020, 5, 27–31. [Google Scholar] [CrossRef] [PubMed]
  148. Ciafrè, S.A.; Galardi, S.; Mangiola, A.; Ferracin, M.; Liu, C.-G.; Sabatino, G.; Negrini, M.; Maira, G.; Croce, C.M.; Farace, M.G. Extensive Modulation of a Set of microRNAs in Primary Glioblastoma. Biochem. Biophys. Res. Commun. 2005, 334, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
  149. Shea, A.; Harish, V.; Afzal, Z.; Chijioke, J.; Kedir, H.; Dusmatova, S.; Roy, A.; Ramalinga, M.; Harris, B.; Blancato, J.; et al. MicroRNAs in Glioblastoma Multiforme Pathogenesis and Therapeutics. Cancer Med. 2016, 5, 1917–1946. [Google Scholar] [CrossRef] [PubMed]
  150. Chen, M.; Medarova, Z.; Moore, A. Role of microRNAs in Glioblastoma. Oncotarget 2021, 12, 1707–1723. [Google Scholar] [CrossRef] [PubMed]
  151. Wu, D.; Wang, Y.; Fan, L.; Luo, H.; Han, B.; Sun, L.; Wang, X.; Zhang, J.; Cao, L.; Wang, X.; et al. MicroRNA-7 Regulates Glioblastoma Cell Invasion via Targeting Focal Adhesion Kinase Expression. Chin. Med. J. 2011, 124, 2616–2621. [Google Scholar] [PubMed]
  152. Liu, Z.; Jiang, Z.; Huang, J.; Huang, S.; Li, Y.; Yu, S.; Yu, S.; Liu, X. miR-7 Inhibits Glioblastoma Growth by Simultaneously Interfering with the PI3K/ATK and Raf/MEK/ERK Pathways. Int. J. Oncol. 2014, 44, 1571–1580. [Google Scholar] [CrossRef]
  153. Tamim, S.; Vo, D.T.; Uren, P.J.; Qiao, M.; Bindewald, E.; Kasprzak, W.K.; Shapiro, B.A.; Nakaya, H.I.; Burns, S.C.; Araujo, P.R.; et al. Genomic Analyses Reveal Broad Impact of miR-137 on Genes Associated with Malignant Transformation and Neuronal Differentiation in Glioblastoma Cells. PLoS ONE 2014, 9, e85591. [Google Scholar] [CrossRef]
  154. Zhang, X.; Zhang, X.; Hu, S.; Zheng, M.; Zhang, J.; Zhao, J.; Zhang, X.; Yan, B.; Jia, L.; Zhao, J.; et al. Identification of miRNA-7 by Genome-Wide Analysis as a Critical Sensitizer for TRAIL-Induced Apoptosis in Glioblastoma Cells. Nucleic Acids Res. 2017, 45, 5930–5944. [Google Scholar] [CrossRef]
  155. Sun, L.; Yan, W.; Wang, Y.; Sun, G.; Luo, H.; Zhang, J.; Wang, X.; You, Y.; Yang, Z.; Liu, N. MicroRNA-10b Induces Glioma Cell Invasion by Modulating MMP-14 and uPAR Expression via HOXD10. Brain Res. 2011, 1389, 9–18. [Google Scholar] [CrossRef] [PubMed]
  156. Gabriely, G.; Yi, M.; Narayan, R.S.; Niers, J.M.; Wurdinger, T.; Imitola, J.; Ligon, K.L.; Kesari, S.; Esau, C.; Stephens, R.M.; et al. Human Glioma Growth Is Controlled by MicroRNA-10b. Cancer Res. 2011, 71, 3563–3572. [Google Scholar] [CrossRef] [PubMed]
  157. Teplyuk, N.M.; Uhlmann, E.J.; Wong, A.H.-K.; Karmali, P.; Basu, M.; Gabriely, G.; Jain, A.; Wang, Y.; Chiocca, E.A.; Stephens, R.; et al. MicroRNA-10b Inhibition Reduces E2F1-Mediated Transcription and miR-15/16 Activity in Glioblastoma. Oncotarget 2015, 6, 3770–3783. [Google Scholar] [CrossRef] [PubMed]
  158. Li, Y.; Guessous, F.; Zhang, Y.; DiPierro, C.; Kefas, B.; Johnson, E.; Marcinkiewicz, L.; Jiang, J.; Yang, Y.; Schmittgen, T.D.; et al. MicroRNA-34a Inhibits Glioblastoma Growth by Targeting Multiple Oncogenes. Cancer Res. 2009, 69, 7569–7576. [Google Scholar] [CrossRef] [PubMed]
  159. Luan, S.; Sun, L.; Huang, F. MicroRNA-34a: A Novel Tumor Suppressor in P53-Mutant Glioma Cell Line U251. Arch. Med. Res. 2010, 41, 67–74. [Google Scholar] [CrossRef] [PubMed]
  160. Guessous, F.; Zhang, Y.; Kofman, A.; Catania, A.; Li, Y.; Schiff, D.; Purow, B.; Abounader, R. microRNA-34a Is Tumor Suppressive in Brain Tumors and Glioma Stem Cells. Cell Cycle 2010, 9, 1031–1036. [Google Scholar] [CrossRef] [PubMed]
  161. Li, W.-B.; Ma, M.-W.; Dong, L.-J.; Wang, F.; Chen, L.-X.; Li, X.-R. MicroRNA-34a Targets Notch1 and Inhibits Cell Proliferation in Glioblastoma Multiforme. Cancer Biol. Ther. 2011, 12, 477–483. [Google Scholar] [CrossRef]
  162. Vo, D.T.; Qiao, M.; Smith, A.D.; Burns, S.C.; Brenner, A.J.; Penalva, L.O.F. The Oncogenic RNA-Binding Protein Musashi1 Is Regulated by Tumor Suppressor miRNAs. RNA Biol. 2011, 8, 817–828. [Google Scholar] [CrossRef]
  163. Silber, J.; Jacobsen, A.; Ozawa, T.; Harinath, G.; Pedraza, A.; Sander, C.; Holland, E.C.; Huse, J.T. miR-34a Repression in Proneural Malignant Gliomas Upregulates Expression of Its Target PDGFRA and Promotes Tumorigenesis. PLoS ONE 2012, 7, e33844. [Google Scholar] [CrossRef]
  164. Yin, D.; Ogawa, S.; Kawamata, N.; Leiter, A.; Ham, M.; Li, D.; Doan, N.B.; Said, J.W.; Black, K.L.; Phillip Koeffler, H. miR-34a Functions as a Tumor Suppressor Modulating EGFR in Glioblastoma Multiforme. Oncogene 2013, 32, 1155–1163. [Google Scholar] [CrossRef]
  165. Fan, Y.N.; Meley, D.; Pizer, B.; Sée, V. Mir-34a Mimics Are Potential Therapeutic Agents for P53-Mutated and Chemo-Resistant Brain Tumour Cells. PLoS ONE 2014, 9, e108514. [Google Scholar] [CrossRef]
  166. Rathod, S.S.; Rani, S.B.; Khan, M.; Muzumdar, D.; Shiras, A. Tumor Suppressive miRNA-34a Suppresses Cell Proliferation and Tumor Growth of Glioma Stem Cells by Targeting Akt and Wnt Signaling Pathways. FEBS Open Bio 2014, 4, 485–495. [Google Scholar] [CrossRef] [PubMed]
  167. Duan, J.; Zhou, K.; Tang, X.; Duan, J.; Zhao, L. MicroRNA-34a Inhibits Cell Proliferation and Induces Cell Apoptosis of Glioma Cells via Targeting of Bcl-2. Mol. Med. Rep. 2016, 14, 432–438. [Google Scholar] [CrossRef] [PubMed]
  168. Lages, E.; Guttin, A.; El Atifi, M.; Ramus, C.; Ipas, H.; Dupré, I.; Rolland, D.; Salon, C.; Godfraind, C.; deFraipont, F.; et al. MicroRNA and Target Protein Patterns Reveal Physiopathological Features of Glioma Subtypes. PLoS ONE 2011, 6, e20600. [Google Scholar] [CrossRef] [PubMed]
  169. Chan, J.A.; Krichevsky, A.M.; Kosik, K.S. MicroRNA-21 Is an Antiapoptotic Factor in Human Glioblastoma Cells. Cancer Res. 2005, 65, 6029–6033. [Google Scholar] [CrossRef]
  170. Corsten, M.F.; Miranda, R.; Kasmieh, R.; Krichevsky, A.M.; Weissleder, R.; Shah, K. MicroRNA-21 Knockdown Disrupts Glioma Growth In Vivo and Displays Synergistic Cytotoxicity with Neural Precursor Cell–Delivered S-TRAIL in Human Gliomas. Cancer Res. 2007, 67, 8994–9000. [Google Scholar] [CrossRef] [PubMed]
  171. Gabriely, G.; Wurdinger, T.; Kesari, S.; Esau, C.C.; Burchard, J.; Linsley, P.S.; Krichevsky, A.M. MicroRNA 21 Promotes Glioma Invasion by Targeting Matrix Metalloproteinase Regulators. Mol. Cell. Biol. 2008, 28, 5369–5380. [Google Scholar] [CrossRef]
  172. Papagiannakopoulos, T.; Shapiro, A.; Kosik, K.S. MicroRNA-21 Targets a Network of Key Tumor-Suppressive Pathways in Glioblastoma Cells. Cancer Res. 2008, 68, 8164–8172. [Google Scholar] [CrossRef]
  173. Ren, Y.; Zhou, X.; Mei, M.; Yuan, X.-B.; Han, L.; Wang, G.-X.; Jia, Z.-F.; Xu, P.; Pu, P.-Y.; Kang, C.-S. MicroRNA-21 Inhibitor Sensitizes Human Glioblastoma Cells U251 (PTEN-Mutant) and LN229 (PTEN-Wild Type) to Taxol. BMC Cancer 2010, 10, 27. [Google Scholar] [CrossRef] [PubMed]
  174. Skalsky, R.L.; Cullen, B.R. Reduced Expression of Brain-Enriched microRNAs in Glioblastomas Permits Targeted Regulation of a Cell Death Gene. PLoS ONE 2011, 6, e24248. [Google Scholar] [CrossRef] [PubMed]
  175. Godlewski, J.; Nowicki, M.O.; Bronisz, A.; Williams, S.; Otsuki, A.; Nuovo, G.; RayChaudhury, A.; Newton, H.B.; Chiocca, E.A.; Lawler, S. Targeting of the Bmi-1 Oncogene/Stem Cell Renewal Factor by MicroRNA-128 Inhibits Glioma Proliferation and Self-Renewal. Cancer Res. 2008, 68, 9125–9130. [Google Scholar] [CrossRef] [PubMed]
  176. Cui, J.G.; Zhao, Y.; Sethi, P.; Li, Y.Y.; Mahta, A.; Culicchia, F.; Lukiw, W.J. Micro-RNA-128 (miRNA-128) down-Regulation in Glioblastoma Targets ARP5 (ANGPTL6), Bmi-1 and E2F-3a, Key Regulators of Brain Cell Proliferation. J. Neurooncol. 2010, 98, 297–304. [Google Scholar] [CrossRef] [PubMed]
  177. Rao, S.A.; Santosh, V.; Somasundaram, K. Genome-Wide Expression Profiling Identifies Deregulated miRNAs in Malignant Astrocytoma. Mod. Pathol. 2010, 23, 1404–1417. [Google Scholar] [CrossRef] [PubMed]
  178. Lavon, I.; Zrihan, D.; Granit, A.; Einstein, O.; Fainstein, N.; Cohen, M.A.; Cohen, M.A.; Zelikovitch, B.; Shoshan, Y.; Spektor, S.; et al. Gliomas Display a microRNA Expression Profile Reminiscent of Neural Precursor Cells. Neuro-Oncology 2010, 12, 422–433. [Google Scholar] [CrossRef] [PubMed]
  179. Fang, L.; Deng, Z.; Shatseva, T.; Yang, J.; Peng, C.; Du, W.W.; Yee, A.J.; Ang, L.C.; He, C.; Shan, S.W.; et al. MicroRNA miR-93 Promotes Tumor Growth and Angiogenesis by Targeting Integrin-Β8. Oncogene 2011, 30, 806–821. [Google Scholar] [CrossRef] [PubMed]
  180. Hua, D.; Mo, F.; Ding, D.; Li, L.; Han, X.; Zhao, N.; Foltz, G.; Lin, B.; Lan, Q.; Huang, Q. A Catalogue of Glioblastoma and Brain MicroRNAs Identified by Deep Sequencing. OMICS A J. Integr. Biol. 2012, 16, 690–699. [Google Scholar] [CrossRef]
  181. Papagiannakopoulos, T.; Friedmann-Morvinski, D.; Neveu, P.; Dugas, J.C.; Gill, R.M.; Huillard, E.; Liu, C.; Zong, H.; Rowitch, D.H.; Barres, B.A.; et al. Pro-Neural miR-128 Is a Glioma Tumor Suppressor That Targets Mitogenic Kinases. Oncogene 2012, 31, 1884–1895. [Google Scholar] [CrossRef]
  182. Piwecka, M.; Rolle, K.; Belter, A.; Barciszewska, A.M.; Żywicki, M.; Michalak, M.; Nowak, S.; Naskręt-Barciszewska, M.Z.; Barciszewski, J. Comprehensive Analysis of microRNA Expression Profile in Malignant Glioma Tissues. Mol. Oncol. 2015, 9, 1324–1340. [Google Scholar] [CrossRef]
  183. Shan, Z.; Tian, R.; Zhang, M.; Gui, Z.; Wu, J.; Ding, M.; Zhou, X.-F.; He, J. miR128-1 Inhibits the Growth of Glioblastoma Multiforme and Glioma Stem-like Cells via Targeting BMI1 and E2F3. Oncotarget 2016, 7, 78813–78826. [Google Scholar] [CrossRef]
  184. Shi, Z.; Wang, J.; Yan, Z.; You, Y.; Li, C.; Qian, X.; Yin, Y.; Zhao, P.; Wang, Y.; Wang, X.; et al. MiR-128 Inhibits Tumor Growth and Angiogenesis by Targeting p70S6K1. PLoS ONE 2012, 7, e32709. [Google Scholar] [CrossRef] [PubMed]
  185. Chen, R.; Liu, H.; Cheng, Q.; Jiang, B.; Peng, R.; Zou, Q.; Yang, W.; Yang, X.; Wu, X.; Chen, Z. MicroRNA-93 Promotes the Malignant Phenotypes of Human Glioma Cells and Induces Their Chemoresistance to Temozolomide. Biol. Open 2016, 5, 669–677. [Google Scholar] [CrossRef] [PubMed]
  186. Bhaskaran, V.; Nowicki, M.O.; Idriss, M.; Jimenez, M.A.; Lugli, G.; Hayes, J.L.; Mahmoud, A.B.; Zane, R.E.; Passaro, C.; Ligon, K.L.; et al. The Functional Synergism of microRNA Clustering Provides Therapeutically Relevant Epigenetic Interference in Glioblastoma. Nat. Commun. 2019, 10, 442. [Google Scholar] [CrossRef]
  187. Huang, T.; Wan, X.; Alvarez, A.A.; James, C.D.; Song, X.; Yang, Y.; Sastry, N.; Nakano, I.; Sulman, E.P.; Hu, B.; et al. MIR93 (microRNA-93 ) Regulates Tumorigenicity and Therapy Response of Glioblastoma by Targeting Autophagy. Autophagy 2019, 15, 1100–1111. [Google Scholar] [CrossRef]
  188. Huang, T.T.; Sarkaria, S.M.; Cloughesy, T.F.; Mischel, P.S. Targeted Therapy for Malignant Glioma Patients: Lessons Learned and the Road Ahead. Neurotherapeutics 2009, 6, 500–512. [Google Scholar] [CrossRef]
  189. Zhang, L.; Zhang, Y.; Gao, H.; Li, X.; Li, P. Underlying Mechanisms and Clinical Potential of circRNAs in Glioblastoma. Oncol. Res. 2023, 31, 449–462. [Google Scholar] [CrossRef]
  190. Guo, X.; Piao, H. Research Progress of circRNAs in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 791892. [Google Scholar] [CrossRef]
  191. Peng, Z.; Liu, C.; Wu, M. New Insights into Long Noncoding RNAs and Their Roles in Glioma. Mol. Cancer 2018, 17, 61. [Google Scholar] [CrossRef] [PubMed]
  192. Zottel, A.; Šamec, N.; Videtič Paska, A.; Jovčevska, I. Coding of Glioblastoma Progression and Therapy Resistance through Long Noncoding RNAs. Cancers 2020, 12, 1842. [Google Scholar] [CrossRef]
  193. Dai, L.; Liang, W.; Shi, Z.; Li, X.; Zhou, S.; Hu, W.; Yang, Z.; Wang, X. Systematic Characterization and Biological Functions of Non-coding RNAs in Glioblastoma. Cell Prolif. 2022, 56, e13375. [Google Scholar] [CrossRef]
  194. Yadav, G.; Kulshreshtha, R. Metastasis Associated Long Noncoding RNAs in Glioblastoma: Biomarkers and Therapeutic Targets. J. Cell. Physiol. 2022, 237, 401–420. [Google Scholar] [CrossRef]
  195. Shen, J.; Hodges, T.R.; Song, R.; Gong, Y.; Calin, G.A.; Heimberger, A.B.; Zhao, H. Serum HOTAIR and GAS5 Levels as Predictors of Survival in Patients with Glioblastoma. Mol. Carcinog. 2018, 57, 137–141. [Google Scholar] [CrossRef]
  196. Xi, J.; Sun, Q.; Ma, L.; Kang, J. Long Non-Coding RNAs in Glioma Progression. Cancer Lett. 2018, 419, 203–209. [Google Scholar] [CrossRef] [PubMed]
  197. Yu, W.; Ma, Y.; Hou, W.; Wang, F.; Cheng, W.; Qiu, F.; Wu, P.; Zhang, G. Identification of Immune-Related lncRNA Prognostic Signature and Molecular Subtypes for Glioblastoma. Front. Immunol. 2021, 12, 706936. [Google Scholar] [CrossRef] [PubMed]
  198. Eraky, A.M.; Keles, A.; Goodman, S.L.; Baskaya, M.K. Serum Long Non-Coding RNAs as Potential Noninvasive Biomarkers for Glioblastoma Diagnosis, Prognosis, and Chemoresistance. JIN 2022, 21, 111. [Google Scholar] [CrossRef]
  199. Chen, X.; Gao, Y.; Li, D.; Cao, Y.; Hao, B. LncRNA-TP53TG1 Participated in the Stress Response Under Glucose Deprivation in Glioma. J. Cell. Biochem. 2017, 118, 4897–4904. [Google Scholar] [CrossRef] [PubMed]
  200. Wang, J.; Liu, X.; Yan, C.; Liu, J.; Wang, S.; Hong, Y.; Gu, A.; Zhao, P. LEF1-AS1, a Long-Noncoding RNA, Promotes Malignancy in Glioblastoma. OncoTargets Ther. 2017, 10, 4251–4260. [Google Scholar] [CrossRef]
  201. Hegi, M.E.; Liu, L.; Herman, J.G.; Stupp, R.; Wick, W.; Weller, M.; Mehta, M.P.; Gilbert, M.R. Correlation of O 6-Methylguanine Methyltransferase (MGMT) Promoter Methylation With Clinical Outcomes in Glioblastoma and Clinical Strategies to Modulate MGMT Activity. JCO 2008, 26, 4189–4199. [Google Scholar] [CrossRef]
  202. Velásquez, C.; Mansouri, S.; Gutiérrez, O.; Mamatjan, Y.; Mollinedo, P.; Karimi, S.; Singh, O.; Terán, N.; Martino, J.; Zadeh, G.; et al. Hypoxia Can Induce Migration of Glioblastoma Cells Through a Methylation-Dependent Control of ODZ1 Gene Expression. Front. Oncol. 2019, 9, 1036. [Google Scholar] [CrossRef]
  203. Wu, Q.; Berglund, A.E.; Etame, A.B. The Impact of Epigenetic Modifications on Adaptive Resistance Evolution in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 8324. [Google Scholar] [CrossRef]
  204. Scoumanne, A.; Zhang, J.; Chen, X. PRMT5 Is Required for Cell-Cycle Progression and P53 Tumor Suppressor Function. Nucleic Acids Res. 2009, 37, 4965–4976. [Google Scholar] [CrossRef]
  205. Yan, F.; Alinari, L.; Lustberg, M.E.; Martin, L.K.; Cordero-Nieves, H.M.; Banasavadi-Siddegowda, Y.; Virk, S.; Barnholtz-Sloan, J.; Bell, E.H.; Wojton, J.; et al. Genetic Validation of the Protein Arginine Methyltransferase PRMT5 as a Candidate Therapeutic Target in Glioblastoma. Cancer Res. 2014, 74, 1752–1765. [Google Scholar] [CrossRef]
  206. Banasavadi-Siddegowda, Y.K.; Russell, L.; Frair, E.; Karkhanis, V.A.; Relation, T.; Yoo, J.Y.; Zhang, J.; Sif, S.; Imitola, J.; Baiocchi, R.; et al. PRMT5-PTEN Molecular Pathway Regulates Senescence and Self-Renewal of Primary Glioblastoma Neurosphere Cells. Oncogene 2017, 36, 263–274. [Google Scholar] [CrossRef]
  207. Banasavadi-Siddegowda, Y.K.; Welker, A.M.; An, M.; Yang, X.; Zhou, W.; Shi, G.; Imitola, J.; Li, C.; Hsu, S.; Wang, J.; et al. PRMT5 as a Druggable Target for Glioblastoma Therapy. Neuro-Oncology 2018, 20, 753–763. [Google Scholar] [CrossRef] [PubMed]
  208. Yang, W.-B.; Wu, A.-C.; Hsu, T.-I.; Liou, J.-P.; Lo, W.-L.; Chang, K.-Y.; Chen, P.-Y.; Kikkawa, U.; Yang, S.-T.; Kao, T.-J.; et al. Histone Deacetylase 6 Acts Upstream of DNA Damage Response Activation to Support the Survival of Glioblastoma Cells. Cell Death Dis. 2021, 12, 884. [Google Scholar] [CrossRef] [PubMed]
  209. Pastorino, O.; Gentile, M.T.; Mancini, A.; Del Gaudio, N.; Di Costanzo, A.; Bajetto, A.; Franco, P.; Altucci, L.; Florio, T.; Stoppelli, M.P.; et al. Histone Deacetylase Inhibitors Impair Vasculogenic Mimicry from Glioblastoma Cells. Cancers 2019, 11, 747. [Google Scholar] [CrossRef] [PubMed]
  210. Yang, W.; Liu, Y.; Gao, R.; Yu, H.; Sun, T. HDAC6 Inhibition Induces Glioma Stem Cells Differentiation and Enhances Cellular Radiation Sensitivity through the SHH/Gli1 Signaling Pathway. Cancer Lett. 2018, 415, 164–176. [Google Scholar] [CrossRef] [PubMed]
  211. Zhang, I.; Beus, M.; Stochaj, U.; Le, P.U.; Zorc, B.; Rajić, Z.; Petrecca, K.; Maysinger, D. Inhibition of Glioblastoma Cell Proliferation, Invasion, and Mechanism of Action of a Novel Hydroxamic Acid Hybrid Molecule. Cell Death Discov. 2018, 4, 41. [Google Scholar] [CrossRef] [PubMed]
  212. LEE, P.; MURPHY, B.; MILLER, R.; MENON, V.; BANIK, N.L.; GIGLIO, P.; LINDHORST, S.M.; VARMA, A.K.; VANDERGRIFT, W.A.; PATEL, S.J.; et al. Mechanisms and Clinical Significance of Histone Deacetylase Inhibitors: Epigenetic Glioblastoma Therapy. Anticancer. Res. 2015, 35, 615–625. [Google Scholar] [PubMed]
  213. Rahman, M.A.; Engelsen, A.S.T.; Sarowar, S.; Bindesbøll, C.; Birkeland, E.; Goplen, D.; Lotsberg, M.L.; Knappskog, S.; Simonsen, A.; Chekenya, M. Bortezomib Abrogates Temozolomide-Induced Autophagic Flux through an ATG5 Dependent Pathway. Front. Cell Dev. Biol. 2022, 10, 1022191. [Google Scholar] [CrossRef] [PubMed]
  214. Rampazzo, E.; Manfreda, L.; Bresolin, S.; Cani, A.; Mariotto, E.; Bortolozzi, R.; Della Puppa, A.; Viola, G.; Persano, L. Histone Deacetylase Inhibitors Impair Glioblastoma Cell Motility and Proliferation. Cancers 2022, 14, 1897. [Google Scholar] [CrossRef]
  215. Everix, L.; Seane, E.N.; Ebenhan, T.; Goethals, I.; Bolcaen, J. Introducing HDAC-Targeting Radiopharmaceuticals for Glioblastoma Imaging and Therapy. Pharmaceuticals 2023, 16, 227. [Google Scholar] [CrossRef] [PubMed]
  216. Reddy, R.G.; Bhat, U.A.; Chakravarty, S.; Kumar, A. Advances in Histone Deacetylase Inhibitors in Targeting Glioblastoma Stem Cells. Cancer Chemother. Pharmacol. 2020, 86, 165–179. [Google Scholar] [CrossRef]
  217. Ghiaseddin, A.; Reardon, D.; Massey, W.; Mannerino, A.; Lipp, E.S.; Herndon, J.E.; McSherry, F.; Desjardins, A.; Randazzo, D.; Friedman, H.S.; et al. Phase II Study of Bevacizumab and Vorinostat for Patients with Recurrent World Health Organization Grade 4 Malignant Glioma. Oncologist 2018, 23, 157-e21. [Google Scholar] [CrossRef] [PubMed]
  218. Lee, E.Q.; Reardon, D.A.; Schiff, D.; Drappatz, J.; Muzikansky, A.; Grimm, S.A.; Norden, A.D.; Nayak, L.; Beroukhim, R.; Rinne, M.L.; et al. Phase II Study of Panobinostat in Combination with Bevacizumab for Recurrent Glioblastoma and Anaplastic Glioma. Neuro Oncol. 2015, 17, 862–867. [Google Scholar] [CrossRef]
  219. Tsai, H.-C.; Wei, K.-C.; Chen, P.-Y.; Huang, C.-Y.; Chen, K.-T.; Lin, Y.-J.; Cheng, H.-W.; Chen, Y.-R.; Wang, H.-T. Valproic Acid Enhanced Temozolomide-Induced Anticancer Activity in Human Glioma Through the P53-PUMA Apoptosis Pathway. Front. Oncol. 2021, 11, 722754. [Google Scholar] [CrossRef]
  220. Stella, M.; Baiardi, G.; Pasquariello, S.; Sacco, F.; Dellacasagrande, I.; Corsaro, A.; Mattioli, F.; Barbieri, F. Antitumor Potential of Antiepileptic Drugs in Human Glioblastoma: Pharmacological Targets and Clinical Benefits. Biomedicines 2023, 11, 582. [Google Scholar] [CrossRef]
  221. Sullivan, J.K.; Fahey, P.P.; Agho, K.E.; Hurley, S.P.; Feng, Z.; Day, R.O.; Lim, D. Valproic Acid as a Radio-Sensitizer in Glioma: A Systematic Review and Meta-Analysis. Neurooncol. Pract. 2023, 10, 13–23. [Google Scholar] [CrossRef]
  222. Yuan, Y.; Xiang, W.; Qing, M.; Yanhui, L.; Jiewen, L.; Yunhe, M. Survival Analysis for Valproic Acid Use in Adult Glioblastoma Multiforme: A Meta-Analysis of Individual Patient Data and a Systematic Review. Seizure 2014, 23, 830–835. [Google Scholar] [CrossRef]
  223. Krauze, A.V.; Megan, M.; Theresa, C.-Z.; Peter, M.; Shih, J.H.; Tofilon, P.J.; Rowe, L.; Gilbert, M.; Camphausen, K. The Addition of Valproic Acid to Concurrent Radiation Therapy and Temozolomide Improves Patient Outcome: A Correlative Analysis of RTOG 0525, SEER and a Phase II NCI Trial. Cancer Stud. Ther. 2020, 5. [Google Scholar] [CrossRef]
  224. Scicchitano, B.M.; Sorrentino, S.; Proietti, G.; Lama, G.; Dobrowolny, G.; Catizone, A.; Binda, E.; Larocca, L.M.; Sica, G. Levetiracetam Enhances the Temozolomide Effect on Glioblastoma Stem Cell Proliferation and Apoptosis. Cancer Cell Int. 2018, 18. [Google Scholar] [CrossRef]
  225. Hwang, K.; Kim, J.; Kang, S.; Jung, T.; Kim, J.H.; Kim, S.; Kang, S.; Hong, Y.; Kim, T.M.; Kim, Y.J.; et al. Levetiracetam as a Sensitizer of Concurrent Chemoradiotherapy in Newly Diagnosed Glioblastoma: An Open-label Phase 2 Study. Cancer Med. 2021, 11, 371–379. [Google Scholar] [CrossRef]
  226. Pallud, J.; Huberfeld, G.; Dezamis, E.; Peeters, S.; Moiraghi, A.; Gavaret, M.; Guinard, E.; Dhermain, F.; Varlet, P.; Oppenheim, C.; et al. Effect of Levetiracetam Use Duration on Overall Survival of Isocitrate Dehydrogenase Wild-Type Glioblastoma in Adults: An Observational Study. Neurology 2022, 98, e125–e140. [Google Scholar] [CrossRef]
  227. Happold, C.; Gorlia, T.; Chinot, O.; Gilbert, M.R.; Nabors, L.B.; Wick, W.; Pugh, S.L.; Hegi, M.; Cloughesy, T.; Roth, P.; et al. Does Valproic Acid or Levetiracetam Improve Survival in Glioblastoma? A Pooled Analysis of Prospective Clinical Trials in Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2016, 34, 731–739. [Google Scholar] [CrossRef]
  228. Laws, M.T.; Bonomi, R.E.; Kamal, S.; Gelovani, D.J.; Llaniguez, J.; Potukutchi, S.; Lu, X.; Mangner, T.; Gelovani, J.G. Molecular Imaging HDACs Class IIa Expression-Activity and Pharmacologic Inhibition in Intracerebral Glioma Models in Rats Using PET/CT/(MRI) with [18F]TFAHA. Sci. Rep. 2019, 9, 3595. [Google Scholar] [CrossRef]
  229. Cao, R.; Bråkenhielm, E.; Pawliuk, R.; Wariaro, D.; Post, M.J.; Wahlberg, E.; Leboulch, P.; Cao, Y. Angiogenic Synergism, Vascular Stability and Improvement of Hind-Limb Ischemia by a Combination of PDGF-BB and FGF-2. Nat. Med. 2003, 9, 604–613. [Google Scholar] [CrossRef] [PubMed]
  230. Buccarelli, M.; Castellani, G.; Ricci-Vitiani, L. Glioblastoma-Specific Strategies of Vascularization: Implications in Anti-Angiogenic Therapy Resistance. J. Pers. Med. 2022, 12, 1625. [Google Scholar] [CrossRef] [PubMed]
  231. Chiao, M.-T.; Yang, Y.-C.; Cheng, W.-Y.; Shen, C.-C.; Ko, J.-L. CD133+ Glioblastoma Stem-like Cells Induce Vascular Mimicry in Vivo. Curr. Neurovascular Res. 2011, 8, 210–219. [Google Scholar] [CrossRef] [PubMed]
  232. Rosińska, S.; Gavard, J. Tumor Vessels Fuel the Fire in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 6514. [Google Scholar] [CrossRef]
  233. El Hallani, S.; Boisselier, B.; Peglion, F.; Rousseau, A.; Colin, C.; Idbaih, A.; Marie, Y.; Mokhtari, K.; Thomas, J.-L.; Eichmann, A.; et al. A New Alternative Mechanism in Glioblastoma Vascularization: Tubular Vasculogenic Mimicry. Brain 2010, 133, 973–982. [Google Scholar] [CrossRef]
  234. Das, S.; Marsden, P.A. Angiogenesis in Glioblastoma. N. Engl. J. Med. 2013, 369, 1561–1563. [Google Scholar] [CrossRef]
  235. Kioi, M.; Vogel, H.; Schultz, G.; Hoffman, R.M.; Harsh, G.R.; Brown, J.M. Inhibition of Vasculogenesis, but Not Angiogenesis, Prevents the Recurrence of Glioblastoma after Irradiation in Mice. J. Clin. Investig. 2010, 120, 694–705. [Google Scholar] [CrossRef]
  236. Mei, X.; Chen, Y.-S.; Chen, F.-R.; Xi, S.-Y.; Chen, Z.-P. Glioblastoma Stem Cell Differentiation into Endothelial Cells Evidenced through Live-Cell Imaging. Neuro Oncol. 2017, 19, 1109–1118. [Google Scholar] [CrossRef]
  237. Calabrese, C.; Poppleton, H.; Kocak, M.; Hogg, T.L.; Fuller, C.; Hamner, B.; Oh, E.Y.; Gaber, M.W.; Finklestein, D.; Allen, M.; et al. A Perivascular Niche for Brain Tumor Stem Cells. Cancer Cell 2007, 11, 69–82. [Google Scholar] [CrossRef]
  238. Jhaveri, N.; Chen, T.C.; Hofman, F.M. Tumor Vasculature and Glioma Stem Cells: Contributions to Glioma Progression. Cancer Lett. 2016, 380, 545–551. [Google Scholar] [CrossRef] [PubMed]
  239. Charles, N.; Holland, E.C. The Perivascular Niche Microenvironment in Brain Tumor Progression. Cell Cycle 2010, 9, 3012–3021. [Google Scholar] [CrossRef] [PubMed]
  240. Hira, V.V.V.; Aderetti, D.A.; van Noorden, C.J.F. Glioma Stem Cell Niches in Human Glioblastoma Are Periarteriolar. J. Histochem. Cytochem. 2018, 66, 349–358. [Google Scholar] [CrossRef] [PubMed]
  241. Tan, C.; Cruet-Hennequart, S.; Troussard, A.; Fazli, L.; Costello, P.; Sutton, K.; Wheeler, J.; Gleave, M.; Sanghera, J.; Dedhar, S. Regulation of Tumor Angiogenesis by Integrin-Linked Kinase (ILK). Cancer Cell 2004, 5, 79–90. [Google Scholar] [CrossRef]
  242. Zagzag, D.; Lukyanov, Y.; Lan, L.; Ali, M.A.; Esencay, M.; Mendez, O.; Yee, H.; Voura, E.B.; Newcomb, E.W. Hypoxia-Inducible Factor 1 and VEGF Upregulate CXCR4 in Glioblastoma: Implications for Angiogenesis and Glioma Cell Invasion. Lab. Investig. 2006, 86, 1221–1232. [Google Scholar] [CrossRef]
  243. Domènech, M.; Hernández, A.; Plaja, A.; Martínez-Balibrea, E.; Balañà, C. Hypoxia: The Cornerstone of Glioblastoma. Int. J. Mol. Sci. 2021, 22, 12608. [Google Scholar] [CrossRef] [PubMed]
  244. Renfrow, J.J.; Soike, M.H.; West, J.L.; Ramkissoon, S.H.; Metheny-Barlow, L.; Mott, R.T.; Kittel, C.A.; D’Agostino, R.B.; Tatter, S.B.; Laxton, A.W.; et al. Attenuating Hypoxia Driven Malignant Behavior in Glioblastoma with a Novel Hypoxia-Inducible Factor 2 Alpha Inhibitor. Sci. Rep. 2020, 10, 15195. [Google Scholar] [CrossRef] [PubMed]
  245. Strowd, R.; Ellingson, B.; Raymond, C.; Yao, J.; Wen, P.Y.; Ahluwalia, M.; Piotrowski, A.; Desai, A.; Clarke, J.L.; Lieberman, F.S.; et al. Activity of a First-in-Class Oral HIF2-Alpha Inhibitor, PT2385, in Patients with First Recurrence of Glioblastoma. J. Neurooncol. 2023, 165, 101–112. [Google Scholar] [CrossRef] [PubMed]
  246. Srivastava, C.; Irshad, K.; Dikshit, B.; Chattopadhyay, P.; Sarkar, C.; Gupta, D.K.; Sinha, S.; Chosdol, K. FAT1 Modulates EMT and Stemness Genes Expression in Hypoxic Glioblastoma. Int. J. Cancer 2018, 142, 805–812. [Google Scholar] [CrossRef] [PubMed]
  247. Aderetti, D.A.; Hira, V.V.V.; Molenaar, R.J.; van Noorden, C.J.F. The Hypoxic Peri-Arteriolar Glioma Stem Cell Niche, an Integrated Concept of Five Types of Niches in Human Glioblastoma. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 346–354. [Google Scholar] [CrossRef] [PubMed]
  248. Kumar, S.; Sharife, H.; Kreisel, T.; Mogilevsky, M.; Bar-Lev, L.; Grunewald, M.; Aizenshtein, E.; Karni, R.; Paldor, I.; Shlomi, T.; et al. Intra-Tumoral Metabolic Zonation and Resultant Phenotypic Diversification Are Dictated by Blood Vessel Proximity. Cell Metab. 2019, 30, 201–211.e6. [Google Scholar] [CrossRef] [PubMed]
  249. Da Ros, M.; De Gregorio, V.; Iorio, A.L.; Giunti, L.; Guidi, M.; de Martino, M.; Genitori, L.; Sardi, I. Glioblastoma Chemoresistance: The Double Play by Microenvironment and Blood-Brain Barrier. Int. J. Mol. Sci. 2018, 19, 2879. [Google Scholar] [CrossRef] [PubMed]
  250. Faustino, A.C.; Viani, G.A.; Hamamura, A.C. Patterns of Recurrence and Outcomes of Glioblastoma Multiforme Treated with Chemoradiation and Adjuvant Temozolomide. Clinics 2020, 75, e1553. [Google Scholar] [CrossRef]
  251. Lakomy, R.; Kazda, T.; Selingerova, I.; Poprach, A.; Pospisil, P.; Belanova, R.; Fadrus, P.; Vybihal, V.; Smrcka, M.; Jancalek, R.; et al. Real-World Evidence in Glioblastoma: Stupp’s Regimen After a Decade. Front. Oncol. 2020, 10, 840. [Google Scholar] [CrossRef]
  252. AbdelFatah, M.A.R.; Kotb, A.; Said, M.A.; Abouelmaaty, E.M.H. Impact of Extent of Resection of Newly Diagnosed Glioblastomas on Survival: A Meta-Analysis. Egypt. J. Neurosurg. 2022, 37, 3. [Google Scholar] [CrossRef]
  253. Weller, M.; van den Bent, M.; Preusser, M.; Le Rhun, E.; Tonn, J.C.; Minniti, G.; Bendszus, M.; Balana, C.; Chinot, O.; Dirven, L.; et al. EANO Guidelines on the Diagnosis and Treatment of Diffuse Gliomas of Adulthood. Nat. Rev. Clin. Oncol. 2021, 18, 170–186. [Google Scholar] [CrossRef] [PubMed]
  254. Mohile, N.A.; Messersmith, H.; Gatson, N.T.; Hottinger, A.F.; Lassman, A.; Morton, J.; Ney, D.; Nghiemphu, P.L.; Olar, A.; Olson, J.; et al. Therapy for Diffuse Astrocytic and Oligodendroglial Tumors in Adults: ASCO-SNO Guideline. J. Clin. Oncol. 2022, 40, 403–426. [Google Scholar] [CrossRef] [PubMed]
  255. Stupp, R.; Hegi, M.E.; Mason, W.P.; Bent, M.J.; Taphoorn, M.J.B.; Janzer, R.C.; Ludwin, S.K.; Allgeier, A.; Fisher, B.; Belanger, K.; et al. Effects of Radiotherapy with Concomitant and Adjuvant Temozolomide versus Radiotherapy Alone on Survival in Glioblastoma in a Randomised Phase III Study: 5-Year Analysis of the EORTC-NCIC Trial. Lancet Oncol. 2009, 10, 459–466. [Google Scholar] [CrossRef] [PubMed]
  256. Toms, S.A.; Kim, C.Y.; Nicholas, G.; Ram, Z. Increased Compliance with Tumor Treating Fields Therapy Is Prognostic for Improved Survival in the Treatment of Glioblastoma: A Subgroup Analysis of the EF-14 Phase III Trial. J. Neurooncol. 2019, 141, 467–473. [Google Scholar] [CrossRef]
  257. Gilbert, M.R.; Wang, M.; Aldape, K.D.; Stupp, R.; Hegi, M.E.; Jaeckle, K.A.; Armstrong, T.S.; Wefel, J.S.; Won, M.; Blumenthal, D.T.; et al. Dose-Dense Temozolomide for Newly Diagnosed Glioblastoma: A Randomized Phase III Clinical Trial. J. Clin. Oncol. 2013, 31, 4085–4091. [Google Scholar] [CrossRef]
  258. Gately, L.; Mesía, C.; Sepúlveda, J.M.; Del Barco, S.; Pineda, E.; Gironés, R.; Fuster, J.; Hong, W.; Dumas, M.; Gill, S.; et al. A Combined Analysis of Two Prospective Randomised Studies Exploring the Impact of Extended Post-Radiation Temozolomide on Survival Outcomes in Newly Diagnosed Glioblastoma. J. Neurooncol. 2023. [Google Scholar] [CrossRef]
  259. Herrlinger, U.; Tzaridis, T.; Mack, F.; Steinbach, J.P.; Schlegel, U.; Sabel, M.; Hau, P.; Kortmann, R.-D.; Krex, D.; Grauer, O.; et al. Lomustine-Temozolomide Combination Therapy versus Standard Temozolomide Therapy in Patients with Newly Diagnosed Glioblastoma with Methylated MGMT Promoter (CeTeG/NOA-09): A Randomised, Open-Label, Phase 3 Trial. Lancet 2019, 393, 678–688. [Google Scholar] [CrossRef]
  260. Chinot, O.L.; Wick, W.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Carpentier, A.F.; Hoang-Xuan, K.; Kavan, P.; Cernea, D.; et al. Bevacizumab plus Radiotherapy-Temozolomide for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 709–722. [Google Scholar] [CrossRef]
  261. Saran, F.; Chinot, O.L.; Henriksson, R.; Mason, W.; Wick, W.; Cloughesy, T.; Dhar, S.; Pozzi, E.; Garcia, J.; Nishikawa, R. Bevacizumab, Temozolomide, and Radiotherapy for Newly Diagnosed Glioblastoma: Comprehensive Safety Results during and after First-Line Therapy. Neuro Oncol. 2016, 18, 991–1001. [Google Scholar] [CrossRef]
  262. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef]
  263. Wefel, J.S.; Armstrong, T.S.; Pugh, S.L.; Gilbert, M.R.; Wendland, M.M.; Brachman, D.G.; Roof, K.S.; Brown, P.D.; Crocker, I.R.; Robins, H.I.; et al. Neurocognitive, Symptom, and Health-Related Quality of Life Outcomes of a Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma (NRG/RTOG 0825). Neuro Oncol 2021, 23, 1125–1138. [Google Scholar] [CrossRef]
  264. Blumenthal, D.T.; Rankin, C.; Stelzer, K.J.; Spence, A.M.; Sloan, A.E.; Moore, D.F.; Padula, G.D.A.; Schulman, S.B.; Wade, M.L.; Rushing, E.J. A Phase III Study of Radiation Therapy (RT) and O6-Benzylguanine + BCNU versus RT and BCNU Alone and Methylation Status in Newly Diagnosed Glioblastoma and Gliosarcoma: Southwest Oncology Group (SWOG) Study S0001. Int. J. Clin. Oncol. 2015, 20, 650–658. [Google Scholar] [CrossRef] [PubMed]
  265. Wick, W.; Platten, M.; Meisner, C.; Felsberg, J.; Tabatabai, G.; Simon, M.; Nikkhah, G.; Papsdorf, K.; Steinbach, J.P.; Sabel, M.; et al. Temozolomide Chemotherapy Alone versus Radiotherapy Alone for Malignant Astrocytoma in the Elderly: The NOA-08 Randomised, Phase 3 Trial. Lancet Oncol. 2012, 13, 707–715. [Google Scholar] [CrossRef]
  266. Malmström, A.; Grønberg, B.H.; Marosi, C.; Stupp, R.; Frappaz, D.; Schultz, H.; Abacioglu, U.; Tavelin, B.; Lhermitte, B.; Hegi, M.E.; et al. Temozolomide versus Standard 6-Week Radiotherapy versus Hypofractionated Radiotherapy in Patients Older than 60 Years with Glioblastoma: The Nordic Randomised, Phase 3 Trial. Lancet Oncol. 2012, 13, 916–926. [Google Scholar] [CrossRef]
  267. Perry, J.R.; Laperriere, N.; O’Callaghan, C.J.; Brandes, A.A.; Menten, J.; Phillips, C.; Fay, M.; Nishikawa, R.; Cairncross, J.G.; Roa, W.; et al. Short-Course Radiation plus Temozolomide in Elderly Patients with Glioblastoma. N. Engl. J. Med. 2017, 376, 1027–1037. [Google Scholar] [CrossRef] [PubMed]
  268. Bonosi, L.; Marrone, S.; Benigno, U.E.; Buscemi, F.; Musso, S.; Porzio, M.; Silven, M.P.; Torregrossa, F.; Grasso, G. Maximal Safe Resection in Glioblastoma Surgery: A Systematic Review of Advanced Intraoperative Image-Guided Techniques. Brain Sci. 2023, 13, 216. [Google Scholar] [CrossRef]
  269. Gerritsen, J.K.W.; Broekman, M.L.D.; De Vleeschouwer, S.; Schucht, P.; Nahed, B.V.; Berger, M.S.; Vincent, A.J.P.E. Safe Surgery for Glioblastoma: Recent Advances and Modern Challenges. Neurooncol. Pract. 2022, 9, 364–379. [Google Scholar] [CrossRef] [PubMed]
  270. Ille, S.; Sollmann, N.; Hauck, T.; Maurer, S.; Tanigawa, N.; Obermueller, T.; Negwer, C.; Droese, D.; Zimmer, C.; Meyer, B.; et al. Combined Noninvasive Language Mapping by Navigated Transcranial Magnetic Stimulation and Functional MRI and Its Comparison with Direct Cortical Stimulation. J. Neurosurg. 2015, 123, 212–225. [Google Scholar] [CrossRef]
  271. Stummer, W.; Tonn, J.-C.; Mehdorn, H.M.; Nestler, U.; Franz, K.; Goetz, C.; Bink, A.; Pichlmeier, U.; ALA-Glioma Study Group. Counterbalancing Risks and Gains from Extended Resections in Malignant Glioma Surgery: A Supplemental Analysis from the Randomized 5-Aminolevulinic Acid Glioma Resection Study. Clinical Article. J. Neurosurg. 2011, 114, 613–623. [Google Scholar] [CrossRef]
  272. Al-Adli, N.N.; Young, J.S.; Sibih, Y.E.; Berger, M.S. Technical Aspects of Motor and Language Mapping in Glioma Patients. Cancers 2023, 15, 2173. [Google Scholar] [CrossRef] [PubMed]
  273. Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O.D.; Zanella, F.; Reulen, H.-J. ALA-Glioma Study Group Fluorescence-Guided Surgery with 5-Aminolevulinic Acid for Resection of Malignant Glioma: A Randomised Controlled Multicentre Phase III Trial. Lancet Oncol. 2006, 7, 392–401. [Google Scholar] [CrossRef] [PubMed]
  274. Eljamel, S. 5-ALA Fluorescence Image Guided Resection of Glioblastoma Multiforme: A Meta-Analysis of the Literature. Int. J. Mol. Sci. 2015, 16, 10443–10456. [Google Scholar] [CrossRef] [PubMed]
  275. Kiesel, B.; Wadiura, L.I.; Mischkulnig, M.; Makolli, J.; Sperl, V.; Borkovec, M.; Freund, J.; Lang, A.; Millesi, M.; Berghoff, A.S.; et al. Efficacy, Outcome, and Safety of Elderly Patients with Glioblastoma in the 5-ALA Era: Single Center Experience of More Than 10 Years. Cancers 2021, 13, 6119. [Google Scholar] [CrossRef] [PubMed]
  276. Baig Mirza, A.; Christodoulides, I.; Lavrador, J.P.; Giamouriadis, A.; Vastani, A.; Boardman, T.; Ahmed, R.; Norman, I.; Murphy, C.; Devi, S.; et al. 5-Aminolevulinic Acid-Guided Resection Improves the Overall Survival of Patients with Glioblastoma—A Comparative Cohort Study of 343 Patients. Neuro-Oncology Adv. 2021, 3, vdab047. [Google Scholar] [CrossRef] [PubMed]
  277. Chohan, M.O.; Berger, M.S. 5-Aminolevulinic Acid Fluorescence Guided Surgery for Recurrent High-Grade Gliomas. J. Neurooncol. 2019, 141, 517–522. [Google Scholar] [CrossRef]
  278. Smith, E.J.; Gohil, K.; Thompson, C.M.; Naik, A.; Hassaneen, W. Fluorescein-Guided Resection of High Grade Gliomas: A Meta-Analysis. World Neurosurg. 2021, 155, 181–188.e7. [Google Scholar] [CrossRef]
  279. Acerbi, F.; Broggi, M.; Schebesch, K.-M.; Höhne, J.; Cavallo, C.; De Laurentis, C.; Eoli, M.; Anghileri, E.; Servida, M.; Boffano, C.; et al. Fluorescein-Guided Surgery for Resection of High-Grade Gliomas: A Multicentric Prospective Phase II Study (FLUOGLIO). Clin. Cancer Res. 2018, 24, 52–61. [Google Scholar] [CrossRef]
  280. Ahrens, L.C.; Krabbenhøft, M.G.; Hansen, R.W.; Mikic, N.; Pedersen, C.B.; Poulsen, F.R.; Korshoej, A.R. Effect of 5-Aminolevulinic Acid and Sodium Fluorescein on the Extent of Resection in High-Grade Gliomas and Brain Metastasis. Cancers 2022, 14, 617. [Google Scholar] [CrossRef]
  281. Noh, T.; Mustroph, M.; Golby, A.J. Intraoperative Imaging for High-Grade Glioma Surgery. Neurosurg. Clin. N. Am. 2021, 32, 47–54. [Google Scholar] [CrossRef]
  282. Roder, C.; Bisdas, S.; Ebner, F.H.; Honegger, J.; Naegele, T.; Ernemann, U.; Tatagiba, M. Maximizing the Extent of Resection and Survival Benefit of Patients in Glioblastoma Surgery: High-Field iMRI versus Conventional and 5-ALA-Assisted Surgery. Eur. J. Surg. Oncol. (EJSO) 2014, 40, 297–304. [Google Scholar] [CrossRef] [PubMed]
  283. Coburger, J.; Hagel, V.; Wirtz, C.R.; König, R. Surgery for Glioblastoma: Impact of the Combined Use of 5-Aminolevulinic Acid and Intraoperative MRI on Extent of Resection and Survival. PLoS ONE 2015, 10, e0131872. [Google Scholar] [CrossRef] [PubMed]
  284. Nickel, K.; Renovanz, M.; König, J.; Stöckelmaier, L.; Hickmann, A.-K.; Nadji-Ohl, M.; Engelke, J.; Weimann, E.; Freudenstein, D.; Ganslandt, O.; et al. The Patients’ View: Impact of the Extent of Resection, Intraoperative Imaging, and Awake Surgery on Health-Related Quality of Life in High-Grade Glioma Patients—Results of a Multicenter Cross-Sectional Study. Neurosurg. Rev. 2018, 41, 207–219. [Google Scholar] [CrossRef]
  285. Golub, D.; Hyde, J.; Dogra, S.; Nicholson, J.; Kirkwood, K.A.; Gohel, P.; Loftus, S.; Schwartz, T.H. Intraoperative MRI versus 5-ALA in High-Grade Glioma Resection: A Network Meta-Analysis. J. Neurosurg. 2020, 1–15. [Google Scholar] [CrossRef] [PubMed]
  286. Hottinger, A.F.; Stupp, R.; Homicsko, K. Standards of Care and Novel Approaches in the Management of Glioblastoma Multiforme. Chin. J. Cancer 2014, 33, 32–39. [Google Scholar] [CrossRef] [PubMed]
  287. Brown, T.J.; Brennan, M.C.; Li, M.; Church, E.W.; Brandmeir, N.J.; Rakszawski, K.L.; Patel, A.S.; Rizk, E.B.; Suki, D.; Sawaya, R.; et al. Association of the Extent of Resection With Survival in Glioblastoma: A Systematic Review and Meta-Analysis. JAMA Oncol. 2016, 2, 1460–1469. [Google Scholar] [CrossRef]
  288. Shah, A.H.; Heiss, J.D. Neurosurgical Clinical Trials for Glioblastoma: Current and Future Directions. Brain Sci. 2022, 12, 787. [Google Scholar] [CrossRef]
  289. Wen, P.Y.; Weller, M.; Lee, E.Q.; Alexander, B.M.; Barnholtz-Sloan, J.S.; Barthel, F.P.; Batchelor, T.T.; Bindra, R.S.; Chang, S.M.; Chiocca, E.A.; et al. Glioblastoma in Adults: A Society for Neuro-Oncology (SNO) and European Society of Neuro-Oncology (EANO) Consensus Review on Current Management and Future Directions. Neuro-Oncology 2020, 22, 1073–1113. [Google Scholar] [CrossRef]
  290. Westphal, M.; Hilt, D.C.; Bortey, E.; Delavault, P.; Olivares, R.; Warnke, P.C.; Whittle, I.R.; Jääskeläinen, J.; Ram, Z. A Phase 3 Trial of Local Chemotherapy with Biodegradable Carmustine (BCNU) Wafers (Gliadel Wafers) in Patients with Primary Malignant Glioma. Neuro Oncol. 2003, 5, 79–88. [Google Scholar] [CrossRef]
  291. Fisher, J.P.; Adamson, D.C. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021, 9, 324. [Google Scholar] [CrossRef]
  292. Jung, I.-H.; Chang, K.W.; Park, S.H.; Moon, J.H.; Kim, E.H.; Jung, H.H.; Kang, S.-G.; Chang, J.H.; Chang, J.W.; Chang, W.S. Stereotactic Biopsy for Adult Brainstem Lesions: A Surgical Approach and Its Diagnostic Value According to the 2016 World Health Organization Classification. Cancer Med. 2021, 10, 7514–7524. [Google Scholar] [CrossRef]
  293. Katzendobler, S.; Do, A.; Weller, J.; Dorostkar, M.M.; Albert, N.L.; Forbrig, R.; Niyazi, M.; Egensperger, R.; Thon, N.; Tonn, J.C.; et al. Diagnostic Yield and Complication Rate of Stereotactic Biopsies in Precision Medicine of Gliomas. Front. Neurol. 2022, 13, 822362. [Google Scholar] [CrossRef]
  294. Peters, D.R.; Asher, A.L.; Wait, S.D.; Smith, M.D.; Peters, D.R.; Asher, A.L.; Kelly, R.; Boltes, M.; Wait, S.D.; Smith, M.D.; et al. Is There a Role for Stereotactic Biopsy of Unresectable High-Grade Gliomas? A Retrospective Cohort Study of Short-Term Morbidity and Mortality. TNN 2021, 4, 1–7. [Google Scholar] [CrossRef]
  295. Kazmi, F.; Soon, Y.Y.; Leong, Y.H.; Koh, W.Y.; Vellayappan, B. Re-Irradiation for Recurrent Glioblastoma (GBM): A Systematic Review and Meta-Analysis. J. Neurooncol. 2019, 142, 79–90. [Google Scholar] [CrossRef] [PubMed]
  296. Zhang, J.; Stevens, M.F.G.; Bradshaw, T.D. Temozolomide: Mechanisms of Action, Repair and Resistance. Curr. Mol. Pharmacol. 2012, 5, 102–114. [Google Scholar] [CrossRef]
  297. McLendon, R.; Friedman, A.; Bigner, D.; Van Meir, E.G.; Brat, D.J.; Mastrogianakis, G.M.; Olson, J.J.; Mikkelsen, T.; Lehman, N.; Aldape, K.; et al. Comprehensive Genomic Characterization Defines Human Glioblastoma Genes and Core Pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef]
  298. Liu, D.; Yang, T.; Ma, W.; Wang, Y. Clinical Strategies to Manage Adult Glioblastoma Patients without MGMT Hypermethylation. J. Cancer 2022, 13, 354–363. [Google Scholar] [CrossRef] [PubMed]
  299. Singh, N.; Miner, A.; Hennis, L.; Mittal, S. Mechanisms of Temozolomide Resistance in Glioblastoma—A Comprehensive Review. Cancer Drug Resist. 2021, 4, 17–43. [Google Scholar] [CrossRef]
  300. Arora, A.; Somasundaram, K. Glioblastoma vs Temozolomide: Can the Red Queen Race Be Won? Cancer Biol. Ther. 2019, 20, 1083–1090. [Google Scholar] [CrossRef] [PubMed]
  301. Almeida Lima, K.; Osawa, I.Y.A.; Ramalho, M.C.C.; de Souza, I.; Guedes, C.B.; de Souza Filho, C.H.D.; Monteiro, L.K.S.; Latancia, M.T.; Rocha, C.R.R. Temozolomide Resistance in Glioblastoma by NRF2: Protecting the Evil. Biomedicines 2023, 11, 1081. [Google Scholar] [CrossRef]
  302. Lee, S.Y. Temozolomide Resistance in Glioblastoma Multiforme. Genes Dis. 2016, 3, 198–210. [Google Scholar] [CrossRef]
  303. Pang, L.Y.; Saunders, L.; Argyle, D.J. Epidermal Growth Factor Receptor Activity Is Elevated in Glioma Cancer Stem Cells and Is Required to Maintain Chemotherapy and Radiation Resistance. Oncotarget 2017, 8, 72494–72512. [Google Scholar] [CrossRef] [PubMed]
  304. Campos-Sandoval, J.A.; Gómez-García, M.C.; Santos-Jiménez, J.d.L.; Matés, J.M.; Alonso, F.J.; Márquez, J. Antioxidant Responses Related to Temozolomide Resistance in Glioblastoma. Neurochem. Int. 2021, 149, 105136. [Google Scholar] [CrossRef]
  305. Ortiz, R.; Perazzoli, G.; Cabeza, L.; Jiménez-Luna, C.; Luque, R.; Prados, J.; Melguizo, C. Temozolomide: An Updated Overview of Resistance Mechanisms, Nanotechnology Advances and Clinical Applications. Curr. Neuropharmacol. 2021, 19, 513–537. [Google Scholar] [CrossRef] [PubMed]
  306. Weller, J.; Tzaridis, T.; Mack, F.; Steinbach, J.P.; Schlegel, U.; Hau, P.; Krex, D.; Grauer, O.; Goldbrunner, R.; Bähr, O.; et al. Health-Related Quality of Life and Neurocognitive Functioning with Lomustine-Temozolomide versus Temozolomide in Patients with Newly Diagnosed, MGMT-Methylated Glioblastoma (CeTeG/NOA-09): A Randomised, Multicentre, Open-Label, Phase 3 Trial. Lancet Oncol. 2019, 20, 1444–1453. [Google Scholar] [CrossRef]
  307. Stupp, R.; Lukas, R.V.; Hegi, M.E. Improving Survival in Molecularly Selected Glioblastoma. Lancet 2019, 393, 615–617. [Google Scholar] [CrossRef] [PubMed]
  308. Mehta, M.; Wen, P.; Nishikawa, R.; Reardon, D.; Peters, K. Critical Review of the Addition of Tumor Treating Fields (TTFields) to the Existing Standard of Care for Newly Diagnosed Glioblastoma Patients. Crit. Rev. Oncol. /Hematol. 2017, 111, 60–65. [Google Scholar] [CrossRef]
  309. Burri, S.H.; Gondi, V.; Brown, P.D.; Mehta, M.P. The Evolving Role of Tumor Treating Fields in Managing Glioblastoma: Guide for Oncologists. Am. J. Clin. Oncol. 2018, 41, 191–196. [Google Scholar] [CrossRef]
  310. Giladi, M.; Munster, M.; Schneiderman, R.S.; Voloshin, T.; Porat, Y.; Blat, R.; Zielinska-Chomej, K.; Hååg, P.; Bomzon, Z.; Kirson, E.D.; et al. Tumor Treating Fields (TTFields) Delay DNA Damage Repair Following Radiation Treatment of Glioma Cells. Radiat. Oncol. 2017, 12. [Google Scholar] [CrossRef]
  311. Mun, E.J.; Babiker, H.M.; Weinberg, U.; Kirson, E.D.; Von Hoff, D.D. Tumor-Treating Fields: A Fourth Modality in Cancer Treatment. Clin. Cancer Res. 2018, 24, 266–275. [Google Scholar] [CrossRef]
  312. Shteingauz, A.; Porat, Y.; Voloshin, T.; Schneiderman, R.S.; Munster, M.; Zeevi, E.; Kaynan, N.; Gotlib, K.; Giladi, M.; Kirson, E.D.; et al. AMPK-Dependent Autophagy Upregulation Serves as a Survival Mechanism in Response to Tumor Treating Fields (TTFields). Cell Death Dis. 2018, 9, 1074. [Google Scholar] [CrossRef] [PubMed]
  313. Kim, E.H.; Jo, Y.; Sai, S.; Park, M.-J.; Kim, J.-Y.; Kim, J.S.; Lee, Y.-J.; Cho, J.-M.; Kwak, S.-Y.; Baek, J.-H.; et al. Tumor-Treating Fields Induce Autophagy by Blocking the Akt2/miR29b Axis in Glioblastoma Cells. Oncogene 2019, 38, 6630–6646. [Google Scholar] [CrossRef] [PubMed]
  314. Kissling, C.; Di Santo, S. Tumor Treating Fields—Behind and Beyond Inhibiting the Cancer Cell Cycle. CNS Neurol. Disord. Drug Targets 2020, 19, 599–610. [Google Scholar] [CrossRef] [PubMed]
  315. Wu, H.; Yang, L.; Liu, H.; Zhou, D.; Chen, D.; Zheng, X.; Yang, H.; Li, C.; Chang, J.; Wu, A.; et al. Exploring the Efficacy of Tumor Electric Field Therapy against Glioblastoma: An in Vivo and in Vitro Study. CNS Neurosci. Ther. 2021, 27, 1587–1604. [Google Scholar] [CrossRef]
  316. Guo, X.; Yang, X.; Wu, J.; Yang, H.; Li, Y.; Li, J.; Liu, Q.; Wu, C.; Xing, H.; Liu, P.; et al. Tumor-Treating Fields in Glioblastomas: Past, Present, and Future. Cancers 2022, 14, 3669. [Google Scholar] [CrossRef]
  317. Tanzhu, G.; Chen, L.; Xiao, G.; Shi, W.; Peng, H.; Chen, D.; Zhou, R. The Schemes, Mechanisms and Molecular Pathway Changes of Tumor Treating Fields (TTFields) Alone or in Combination with Radiotherapy and Chemotherapy. Cell Death Discov. 2022, 8, 416. [Google Scholar] [CrossRef]
  318. Shi, P.; Tian, J.; Ulm, B.S.; Mallinger, J.C.; Khoshbouei, H.; Deleyrolle, L.P.; Sarkisian, M.R. Tumor Treating Fields Suppression of Ciliogenesis Enhances Temozolomide Toxicity. Front. Oncol. 2022, 12, 837589. [Google Scholar] [CrossRef]
  319. Kim, E.H.; Song, H.S.; Yoo, S.H.; Yoon, M. Tumor Treating Fields Inhibit Glioblastoma Cell Migration, Invasion and Angiogenesis. Oncotarget 2016, 7, 65125. [Google Scholar] [CrossRef]
  320. Chang, E.; Patel, C.B.; Pohling, C.; Young, C.; Song, J.; Flores, T.A.; Zeng, Y.; Joubert, L.-M.; Arami, H.; Natarajan, A.; et al. Tumor Treating Fields Increases Membrane Permeability in Glioblastoma Cells. Cell Death Discov. 2018, 4, 113. [Google Scholar] [CrossRef]
  321. Salvador, E.; Kessler, A.F.; Domröse, D.; Hörmann, J.; Schaeffer, C.; Giniunaite, A.; Burek, M.; Tempel-Brami, C.; Voloshin, T.; Volodin, A.; et al. Tumor Treating Fields (TTFields) Reversibly Permeabilize the Blood-Brain Barrier In Vitro and In Vivo. Biomolecules 2022, 12, 1348. [Google Scholar] [CrossRef]
  322. Diamant, G.; Simchony Goldman, H.; Gasri Plotnitsky, L.; Roitman, M.; Shiloach, T.; Globerson-Levin, A.; Eshhar, Z.; Haim, O.; Pencovich, N.; Grossman, R.; et al. T Cells Retain Pivotal Antitumoral Functions under Tumor-Treating Electric Fields. J. Immunol. 2021, 207, 709–719. [Google Scholar] [CrossRef] [PubMed]
  323. Voloshin, T.; Davidi, S.; Porat, Y.; Shteingauz, A.; Munster, M.; Kaynan, N.; Giladi, M.; Kirson, E.; Weinberg, U.; Plati, Y. IMMU-52. TUMOR TREATING FIELDS (TTFIELDS) INDUCE IMMUNOGENIC CELL DEATH RESULTING IN ENHANCED ANTITUMOR EFFICACY WHEN COMBINED WITH ANTI-PD-1 THERAPY. Neuro Oncol. 2018, 20, vi133. [Google Scholar] [CrossRef]
  324. Chen, D.; Le, S.B.; Hutchinson, T.E.; Calinescu, A.-A.; Sebastian, M.; Jin, D.; Liu, T.; Ghiaseddin, A.; Rahman, M.; Tran, D.D. Tumor Treating Fields Dually Activate STING and AIM2 Inflammasomes to Induce Adjuvant Immunity in Glioblastoma. J. Clin. Investig. 2022, 132, e149258. [Google Scholar] [CrossRef]
  325. Jo, Y.; Han, Y.I.; Lee, E.; Seo, J.; Oh, G.; Sung, H.; Gi, Y.; Kim, H.; Park, S.; Yoon, M. The Combination of Tumor Treating Fields and Hyperthermia Has Synergistic Therapeutic Effects in Glioblastoma Cells by Downregulating STAT3. Am. J. Cancer Res. 2022, 12, 1423–1432. [Google Scholar]
  326. Pandey, M.; Xiu, J.; Mittal, S.; Zeng, J.; Saul, M.; Kesari, S.; Azadi, A.; Newton, H.; Deniz, K.; Ladner, K.; et al. Molecular Alterations Associated with Improved Outcome in Patients with Glioblastoma Treated with Tumor-Treating Fields. Neuro-Oncology Adv. 2022, 4, vdac096. [Google Scholar] [CrossRef]
  327. Bokstein, F.; Blumenthal, D.; Limon, D.; Harosh, C.B.; Ram, Z.; Grossman, R. Concurrent Tumor Treating Fields (TTFields) and Radiation Therapy for Newly Diagnosed Glioblastoma: A Prospective Safety and Feasibility Study. Front. Oncol. 2020, 10, 411. [Google Scholar] [CrossRef]
  328. Krigers, A.; Pinggera, D.; Demetz, M.; Kornberger, L.-M.; Kerschbaumer, J.; Thomé, C.; Freyschlag, C.F. The Routine Application of Tumor-Treating Fields in the Treatment of Glioblastoma WHO° IV. Front. Neurol. 2022, 13. [Google Scholar] [CrossRef]
  329. Stupp, R.; Wong, E.T.; Kanner, A.A.; Steinberg, D.; Engelhard, H.; Heidecke, V.; Kirson, E.D.; Taillibert, S.; Liebermann, F.; Dbalý, V.; et al. NovoTTF-100A versus Physician’s Choice Chemotherapy in Recurrent Glioblastoma: A Randomised Phase III Trial of a Novel Treatment Modality. Eur. J. Cancer 2012, 48, 2192–2202. [Google Scholar] [CrossRef]
  330. Taphoorn, M.J.B.; Dirven, L.; Kanner, A.A.; Lavy-Shahaf, G.; Weinberg, U.; Taillibert, S.; Toms, S.A.; Honnorat, J.; Chen, T.C.; Sroubek, J.; et al. Influence of Treatment With Tumor-Treating Fields on Health-Related Quality of Life of Patients With Newly Diagnosed Glioblastoma: A Secondary Analysis of a Randomized Clinical Trial. JAMA Oncol. 2018, 4, 495–504. [Google Scholar] [CrossRef]
  331. Vymazal, J.; Kazda, T.; Novak, T.; Slanina, P.; Sroubek, J.; Klener, J.; Hrbac, T.; Syrucek, M.; Rulseh, A.M. Eighteen Years’ Experience with Tumor Treating Fields in the Treatment of Newly Diagnosed Glioblastoma. Front. Oncol. 2023, 12, 1014455. [Google Scholar] [CrossRef]
  332. Stupp, R.; Taillibert, S.; Kanner, A.A.; Kesari, S.; Steinberg, D.M.; Toms, S.A.; Taylor, L.P.; Lieberman, F.; Silvani, A.; Fink, K.L.; et al. Maintenance Therapy With Tumor-Treating Fields Plus Temozolomide vs Temozolomide Alone for Glioblastoma: A Randomized Clinical Trial. JAMA 2015, 314, 2535–2543. [Google Scholar] [CrossRef] [PubMed]
  333. Zhu, P.; Zhu, J.-J. Tumor Treating Fields: A Novel and Effective Therapy for Glioblastoma: Mechanism, Efficacy, Safety and Future Perspectives. Chin. Clin. Oncol. 2017, 6, 41. [Google Scholar] [CrossRef] [PubMed]
  334. Rominiyi, O.; Vanderlinden, A.; Clenton, S.J.; Bridgewater, C.; Al-Tamimi, Y.; Collis, S.J. Tumour Treating Fields Therapy for Glioblastoma: Current Advances and Future Directions. Br. J. Cancer 2021, 124, 697–709. [Google Scholar] [CrossRef]
  335. Wong, E.T.; Lok, E.; Gautam, S.; Swanson, K.D. Dexamethasone Exerts Profound Immunologic Interference on Treatment Efficacy for Recurrent Glioblastoma. Br. J. Cancer 2015, 113, 232–241. [Google Scholar] [CrossRef]
  336. Swildens, K.X.; Sillevis Smitt, P.A.E.; van den Bent, M.J.; French, P.J.; Geurts, M. The Effect of Dexamethasone on the Microenvironment and Efficacy of Checkpoint Inhibitors in Glioblastoma: A Systematic Review. Neurooncol. Adv. 2022, 4, vdac087. [Google Scholar] [CrossRef]
  337. Li, S.; Dong, J.; Wang, X.; Meng, X.; Jiang, C.; Cai, J. Dexamethasone and Compliance Affect TTFields Efficacy to Glioblastoma Patients: A Systematic Review and Meta-Analysis. Chin. Neurosurg. J. 2022, 8, 24. [Google Scholar] [CrossRef] [PubMed]
  338. Wang, M.; Zhang, C.; Wang, X.; Yu, H.; Zhang, H.; Xu, J.; Zhao, J.; Jiang, X. Tumor-Treating Fields (TTFields)-Based Cocktail Therapy: A Novel Blueprint for Glioblastoma Treatment. Am. J. Cancer Res. 2021, 11, 1069–1086. [Google Scholar]
  339. Karanam, N.K.; Story, M.D. An Overview of Potential Novel Mechanisms of Action Underlying Tumor Treating Fields-Induced Cancer Cell Death and Their Clinical Implications. Int. J. Radiat. Biol. 2021, 97, 1044–1054. [Google Scholar] [CrossRef]
  340. Glas, M.; Scheffler, B.; Lazaridis, L.; Herrlinger, U.; Pierscianek, D.; Sure, U.; Proescholdt, M.; Hau, P.; Hense, J.; Kleinschnitz, C.; et al. ACTR-49. PriCoTTF: A PHASE I/II TRIAL OF TUMOR TREATING FIELDS PRIOR AND CONCOMITANT TO RADIOTHERAPY IN NEWLY DIAGNOSED GLIOBLASTOMA. Neuro Oncol. 2018, 20, vi22–vi23. [Google Scholar] [CrossRef]
  341. Karanam, N.K.; Srinivasan, K.; Ding, L.; Sishc, B.; Saha, D.; Story, M.D. Tumor-Treating Fields Elicit a Conditional Vulnerability to Ionizing Radiation via the Downregulation of BRCA1 Signaling and Reduced DNA Double-Strand Break Repair Capacity in Non-Small Cell Lung Cancer Cell Lines. Cell Death Dis. 2017, 8, e2711. [Google Scholar] [CrossRef]
  342. Lazaridis, L.; Schäfer, N.; Teuber-Hanselmann, S.; Blau, T.; Schmidt, T.; Oster, C.; Weller, J.; Tzaridis, T.; Pierscianek, D.; Keyvani, K.; et al. Tumour Treating Fields (TTFields) in Combination with Lomustine and Temozolomide in Patients with Newly Diagnosed Glioblastoma. J. Cancer Res. Clin. Oncol. 2020, 146, 787–792. [Google Scholar] [CrossRef]
  343. Lu, G.; Rao, M.; Zhu, P.; Liang, B.; El-Nazer, R.T.; Fonkem, E.; Bhattacharjee, M.B.; Zhu, J.-J. Triple-Drug Therapy With Bevacizumab, Irinotecan, and Temozolomide Plus Tumor Treating Fields for Recurrent Glioblastoma: A Retrospective Study. Front. Neurol. 2019, 10, 42. [Google Scholar] [CrossRef]
  344. Catacuzzeno, L.; Franciolini, F. Role of KCa3.1 Channels in Modulating Ca2+ Oscillations during Glioblastoma Cell Migration and Invasion. Int. J. Mol. Sci. 2018, 19, 2970. [Google Scholar] [CrossRef]
  345. Robins, H.I.; Nguyen, H.N.; Field, A.; Howard, S.; Salamat, S.; Deming, D.A. Molecular Evolution of a Glioblastoma Controlled With Tumor Treating Fields and Concomitant Temozolomide. Front. Oncol. 2018, 8, 451. [Google Scholar] [CrossRef]
  346. Ahmadi-Zeidabadi, M.; Akbarnejad, Z.; Esmaeeli, M.; Masoumi-Ardakani, Y.; Mohammadipoor-Ghasemabad, L.; Eskandary, H. Impact of Extremely Low-Frequency Electromagnetic Field (100 Hz, 100 G) Exposure on Human Glioblastoma U87 Cells during Temozolomide Administration. Electromagn. Biol. Med. 2019, 38, 198–209. [Google Scholar] [CrossRef]
  347. Cruz, N.; Herculano-Carvalho, M.; Roque, D.; Faria, C.C.; Cascão, R.; Ferreira, H.A.; Reis, C.P.; Matela, N. Highlighted Advances in Therapies for Difficult-To-Treat Brain Tumours Such as Glioblastoma. Pharmaceutics 2023, 15, 928. [Google Scholar] [CrossRef] [PubMed]
  348. Baumgarten, P.; Prange, G.; Kamp, M.A.; Monden, D.; Neef, V.; Schwarzer, F.; Dubinski, D.; Dinc, N.; Weber, K.J.; Czabanka, M.; et al. Treatment of Very Elderly Glioblastoma Patients ≥ 75 Years of Age: Whom to Treat. J. Neurooncol. 2023, 165, 509–515. [Google Scholar] [CrossRef]
  349. Roux, A.; Aboubakr, O.; Elia, A.; Moiraghi, A.; Benevello, C.; Fathallah, H.; Parraga, E.; Oppenheim, C.; Chretien, F.; Dezamis, E.; et al. Carmustine Wafer Implantation for Supratentorial Glioblastomas, IDH-Wildtype in “Extreme” Neurosurgical Conditions. Neurosurg. Rev. 2023, 46, 140. [Google Scholar] [CrossRef]
  350. Ram, Z.; Kim, C.-Y.; Hottinger, A.F.; Idbaih, A.; Nicholas, G.; Zhu, J.-J. Efficacy and Safety of Tumor Treating Fields (TTFields) in Elderly Patients with Newly Diagnosed Glioblastoma: Subgroup Analysis of the Phase 3 EF-14 Clinical Trial. Front. Oncol. 2021, 11, 671972. [Google Scholar] [CrossRef]
  351. Wick, A.; Kessler, T.; Elia, A.E.H.; Winkler, F.; Batchelor, T.T.; Platten, M.; Wick, W. Glioblastoma in Elderly Patients: Solid Conclusions Built on Shifting Sand? Neuro Oncol. 2018, 20, 174–183. [Google Scholar] [CrossRef]
  352. Sulman, E.P.; Ismaila, N.; Armstrong, T.S.; Tsien, C.; Batchelor, T.T.; Cloughesy, T.; Galanis, E.; Gilbert, M.; Gondi, V.; Lovely, M.; et al. Radiation Therapy for Glioblastoma: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Guideline. J. Clin. Oncol. 2017, 35, 361–369. [Google Scholar] [CrossRef]
  353. Wee, C.W. Radiotherapy for Newly Diagnosed Glioblastoma in the Elderly: What Is the Standard? Brain Tumor Res. Treat. 2022, 10, 12–21. [Google Scholar] [CrossRef]
  354. Wirsching, H.-G.; Tabatabai, G.; Roelcke, U.; Hottinger, A.F.; Jörger, F.; Schmid, A.; Plasswilm, L.; Schrimpf, D.; Mancao, C.; Capper, D.; et al. Bevacizumab plus Hypofractionated Radiotherapy versus Radiotherapy Alone in Elderly Patients with Glioblastoma: The Randomized, Open-Label, Phase II ARTE Trial. Ann. Oncol. 2018, 29, 1423–1430. [Google Scholar] [CrossRef]
  355. Theeler, B.J.; Gilbert, M.R. Advances in the Treatment of Newly Diagnosed Glioblastoma. BMC Med. 2015, 13, 293. [Google Scholar] [CrossRef]
  356. Shi, W.; Scannell Bryan, M.; Gilbert, M.R.; Mehta, M.P.; Blumenthal, D.T.; Brown, P.D.; Valeinis, E.; Hopkins, K.; Souhami, L.; Andrews, D.W.; et al. Investigating the Effect of Reirradiation or Systemic Therapy in Patients With Glioblastoma After Tumor Progression: A Secondary Analysis of NRG Oncology/Radiation Therapy Oncology Group Trial 0525. Int. J. Radiat. Oncol. Biol. Phys. 2018, 100, 38–44. [Google Scholar] [CrossRef]
  357. Ringel, F.; Pape, H.; Sabel, M.; Krex, D.; Bock, H.C.; Misch, M.; Weyerbrock, A.; Westermaier, T.; Senft, C.; Schucht, P.; et al. Clinical Benefit from Resection of Recurrent Glioblastomas: Results of a Multicenter Study Including 503 Patients with Recurrent Glioblastomas Undergoing Surgical Resection. Neuro Oncol. 2016, 18, 96–104. [Google Scholar] [CrossRef]
  358. Weller, M.; Le Rhun, E. How Did Lomustine Become Standard of Care in Recurrent Glioblastoma? Cancer Treat. Rev. 2020, 87, 102029. [Google Scholar] [CrossRef]
  359. She, L.; Su, L.; Liu, C. Bevacizumab Combined with Re-Irradiation in Recurrent Glioblastoma. Front. Oncol. 2022, 12, 961014. [Google Scholar] [CrossRef]
  360. Neth, B.J.; Webb, M.J.; Parney, I.F.; Sener, U.T. The Current Status, Challenges, and Future Potential of Therapeutic Vaccination in Glioblastoma. Pharmaceutics 2023, 15, 1134. [Google Scholar] [CrossRef]
  361. Skaga, E.; Skretteberg, M.A.; Johannesen, T.B.; Brandal, P.; Vik-Mo, E.O.; Helseth, E.; Langmoen, I.A. Real-World Validity of Randomized Controlled Phase III Trials in Newly Diagnosed Glioblastoma: To Whom Do the Results of the Trials Apply? Neurooncol. Adv. 2021, 3, vdab008. [Google Scholar] [CrossRef]
  362. Brennan, P.M.; Borchert, R.; Coulter, C.; Critchley, G.R.; Hall, B.; Holliman, D.; Phang, I.; Jefferies, S.J.; Keni, S.; Lee, L.; et al. Second Surgery for Progressive Glioblastoma: A Multi-Centre Questionnaire and Cohort-Based Review of Clinical Decision-Making and Patient Outcomes in Current Practice. J. Neurooncol. 2021, 153, 99–107. [Google Scholar] [CrossRef]
  363. Sharma, M.; Schroeder, J.L.; Elson, P.; Meola, A.; Barnett, G.H.; Vogelbaum, M.A.; Suh, J.H.; Chao, S.T.; Mohammadi, A.M.; Stevens, G.H.J.; et al. Outcomes and Prognostic Stratification of Patients with Recurrent Glioblastoma Treated with Salvage Stereotactic Radiosurgery. J. Neurosurg. 2018, 131, 489–499. [Google Scholar] [CrossRef]
  364. Suchorska, B.; Weller, M.; Tabatabai, G.; Senft, C.; Hau, P.; Sabel, M.C.; Herrlinger, U.; Ketter, R.; Schlegel, U.; Marosi, C.; et al. Complete Resection of Contrast-Enhancing Tumor Volume Is Associated with Improved Survival in Recurrent Glioblastoma-Results from the DIRECTOR Trial. Neuro Oncol. 2016, 18, 549–556. [Google Scholar] [CrossRef]
  365. Wann, A.; Tully, P.A.; Barnes, E.H.; Lwin, Z.; Jeffree, R.; Drummond, K.J.; Gan, H.; Khasraw, M. Outcomes after Second Surgery for Recurrent Glioblastoma: A Retrospective Case-Control Study. J. Neurooncol. 2018, 137, 409–415. [Google Scholar] [CrossRef]
  366. Kamiya-Matsuoka, C.; Gilbert, M.R. Treating Recurrent Glioblastoma: An Update. CNS Oncol. 2015, 4, 91–104. [Google Scholar] [CrossRef]
  367. Minniti, G.; Niyazi, M.; Alongi, F.; Navarria, P.; Belka, C. Current Status and Recent Advances in Reirradiation of Glioblastoma. Radiat. Oncol. 2021, 16, 36. [Google Scholar] [CrossRef]
  368. Post, C.C.B.; Kramer, M.C.A.; Smid, E.J.; van der Weide, H.L.; Kleynen, C.E.; Heesters, M.A.A.M.; Verhoeff, J.J.C. Patterns of Re-Irradiation for Recurrent Gliomas and Validation of a Prognostic Score. Radiother. Oncol. 2019, 130, 156–163. [Google Scholar] [CrossRef]
  369. Christ, S.M.; Youssef, G.; Tanguturi, S.K.; Cagney, D.; Shi, D.; McFaline-Figueroa, J.R.; Chukwueke, U.; Lee, E.Q.; Hertler, C.; Andratschke, N.; et al. Re-Irradiation of Recurrent IDH-Wildtype Glioblastoma in the Bevacizumab and Immunotherapy Era: Target Delineation, Outcomes and Patterns of Recurrence. Clin. Transl. Radiat. Oncol. 2023, 44, 100697. [Google Scholar] [CrossRef]
  370. Pineda, E.; Domenech, M.; Hernández, A.; Comas, S.; Balaña, C. Recurrent Glioblastoma: Ongoing Clinical Challenges and Future Prospects. Onco Targets Ther. 2023, 16, 71–86. [Google Scholar] [CrossRef]
  371. Norden, A.D.; Drappatz, J.; Wen, P.Y. Antiangiogenic Therapies for High-Grade Glioma. Nat. Rev. Neurol. 2009, 5, 610–620. [Google Scholar] [CrossRef]
  372. Hegde, P.S.; Wallin, J.J.; Mancao, C. Predictive Markers of Anti-VEGF and Emerging Role of Angiogenesis Inhibitors as Immunotherapeutics. Semin. Cancer Biol. 2018, 52, 117–124. [Google Scholar] [CrossRef] [PubMed]
  373. Tamura, R.; Tanaka, T.; Ohara, K.; Miyake, K.; Morimoto, Y.; Yamamoto, Y.; Kanai, R.; Akasaki, Y.; Murayama, Y.; Tamiya, T.; et al. Persistent Restoration to the Immunosupportive Tumor Microenvironment in Glioblastoma by Bevacizumab. Cancer Sci. 2019, 110, 499–508. [Google Scholar] [CrossRef]
  374. Fu, M.; Zhou, Z.; Huang, X.; Chen, Z.; Zhang, L.; Zhang, J.; Hua, W.; Mao, Y. Use of Bevacizumab in Recurrent Glioblastoma: A Scoping Review and Evidence Map. BMC Cancer 2023, 23, 544. [Google Scholar] [CrossRef]
  375. Cohen, M.H.; Shen, Y.L.; Keegan, P.; Pazdur, R. FDA Drug Approval Summary: Bevacizumab (Avastin) as Treatment of Recurrent Glioblastoma Multiforme. Oncologist 2009, 14, 1131–1138. [Google Scholar] [CrossRef]
  376. Balañá, C.; Etxaniz, O.; Bugés, C.; Martínez, A. Approval Denied by the European Medicines Agency (EMA) for Bevacizumab in the Treatment of High-Grade Glioma Recurrence: A Good Idea or a Grave Error? Clin. Transl. Oncol. 2011, 13, 209–210. [Google Scholar] [CrossRef] [PubMed]
  377. Taal, W.; Oosterkamp, H.M.; Walenkamp, A.M.E.; Dubbink, H.J.; Beerepoot, L.V.; Hanse, M.C.J.; Buter, J.; Honkoop, A.H.; Boerman, D.; Vos, F.Y.F.; et al. Single-Agent Bevacizumab or Lomustine versus a Combination of Bevacizumab plus Lomustine in Patients with Recurrent Glioblastoma (BELOB Trial): A Randomised Controlled Phase 2 Trial. Lancet Oncol. 2014, 15, 943–953. [Google Scholar] [CrossRef]
  378. Franceschi, E.; Brandes, A.A. The Role of Bevacizumab in Recurrent Glioblastoma: New Insights from Randomized Trials. CNS Oncol. 2015, 4, 117–119. [Google Scholar] [CrossRef]
  379. Field, K.M.; Simes, J.; Nowak, A.K.; Cher, L.; Wheeler, H.; Hovey, E.J.; Brown, C.S.B.; Barnes, E.H.; Sawkins, K.; Livingstone, A.; et al. Randomized Phase 2 Study of Carboplatin and Bevacizumab in Recurrent Glioblastoma. Neuro Oncol. 2015, 17, 1504–1513. [Google Scholar] [CrossRef]
  380. Hovey, E.J.; Field, K.M.; Rosenthal, M.A.; Barnes, E.H.; Cher, L.; Nowak, A.K.; Wheeler, H.; Sawkins, K.; Livingstone, A.; Phal, P.; et al. Continuing or Ceasing Bevacizumab beyond Progression in Recurrent Glioblastoma: An Exploratory Randomized Phase II Trial. Neurooncol. Pract. 2017, 4, 171–181. [Google Scholar] [CrossRef]
  381. Brandes, A.A.; Gil-Gil, M.; Saran, F.; Carpentier, A.F.; Nowak, A.K.; Mason, W.; Zagonel, V.; Dubois, F.; Finocchiaro, G.; Fountzilas, G.; et al. A Randomized Phase II Trial (TAMIGA) Evaluating the Efficacy and Safety of Continuous Bevacizumab Through Multiple Lines of Treatment for Recurrent Glioblastoma. Oncologist 2019, 24, 521–528. [Google Scholar] [CrossRef]
  382. Vredenburgh, J.J.; Cloughesy, T.; Samant, M.; Prados, M.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; et al. Corticosteroid Use in Patients with Glioblastoma at First or Second Relapse Treated with Bevacizumab in the BRAIN Study. Oncologist 2010, 15, 1329–1334. [Google Scholar] [CrossRef] [PubMed]
  383. Dirven, L.; van den Bent, M.J.; Bottomley, A.; van der Meer, N.; van der Holt, B.; Vos, M.J.; Walenkamp, A.M.E.; Beerepoot, L.V.; Hanse, M.C.J.; Reijneveld, J.C.; et al. The Impact of Bevacizumab on Health-Related Quality of Life in Patients Treated for Recurrent Glioblastoma: Results of the Randomised Controlled Phase 2 BELOB Trial. Eur. J. Cancer 2015, 51, 1321–1330. [Google Scholar] [CrossRef] [PubMed]
  384. Tsien, C.; Pugh, S.; Dicker, A.P.; Raizer, J.J.; Matuszak, M.M.; Lallana, E.; Huang, J.; Algan, O.; Taylor, N.; Portelance, L.; et al. Randomized Phase II Trial of Re-Irradiation and Concurrent Bevacizumab versus Bevacizumab Alone as Treatment for Recurrent Glioblastoma (NRG Oncology/RTOG 1205): Initial Outcomes and RT Plan Quality Report. Int. J. Radiat. Oncol. Biol. Phys. 2019, 105, S78. [Google Scholar] [CrossRef]
  385. Fleischmann, D.F.; Jenn, J.; Corradini, S.; Ruf, V.; Herms, J.; Forbrig, R.; Unterrainer, M.; Thon, N.; Kreth, F.W.; Belka, C.; et al. Bevacizumab Reduces Toxicity of Reirradiation in Recurrent High-Grade Glioma. Radiother. Oncol. 2019, 138, 99–105. [Google Scholar] [CrossRef] [PubMed]
  386. Marwah, R.; Xing, D.; Squire, T.; Soon, Y.Y.; Gan, H.K.; Ng, S.P. Reirradiation versus Systemic Therapy versus Combination Therapy for Recurrent High-Grade Glioma: A Systematic Review and Meta-Analysis of Survival and Toxicity. J. Neurooncol. 2023, 164, 505–524. [Google Scholar] [CrossRef]
  387. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef]
  388. Fallah, J.; Chaudhary, R.T.; Rogers, L.R.; Wei, W.; Brewer, C.J.; Peereboom, D.M.; Ahluwalia, M.S. Clinical Outcomes of the Combination of Bevacizumab and Ttfields in Patients with Recurrent Glioblastoma: Results of a Phase II Clinical Trial. JCO 2020, 38, 2537. [Google Scholar] [CrossRef]
  389. Lu, Q.R.; Qian, L.; Zhou, X. Convergence of Developmental Origins and Oncogenic Pathways in Malignant Brain Tumors. Wiley Interdiscip. Rev. Dev. Biol. 2019, 8, e342. [Google Scholar] [CrossRef]
  390. Diplas, B.H.; He, X.; Brosnan-Cashman, J.A.; Liu, H.; Chen, L.H.; Wang, Z.; Moure, C.J.; Killela, P.J.; Loriaux, D.B.; Lipp, E.S.; et al. The Genomic Landscape of TERT Promoter Wildtype-IDH Wildtype Glioblastoma. Nat. Commun. 2018, 9, 2087. [Google Scholar] [CrossRef]
  391. Yeo, A.T.; Shah, R.; Aliazis, K.; Pal, R.; Xu, T.; Zhang, P.; Rawal, S.; Rose, C.M.; Varn, F.S.; Appleman, V.A.; et al. Driver Mutations Dictate the Immunologic Landscape and Response to Checkpoint Immunotherapy of Glioblastoma. Cancer Immunol. Res. 2023, 11, 629–645. [Google Scholar] [CrossRef]
  392. Scherm, A.; Ippen, F.M.; Hau, P.; Baurecht, H.; Wick, W.; Gempt, J.; Knüttel, H.; Leitzmann, M.F.; Seliger, C. Targeted Therapies in Patients with Newly Diagnosed Glioblastoma—A Systematic Meta-Analysis of Randomized Clinical Trials. Int. J. Cancer 2023, 152, 2373–2382. [Google Scholar] [CrossRef] [PubMed]
  393. Zhang, A.B.; Mozaffari, K.; Aguirre, B.; Li, V.; Kubba, R.; Desai, N.C.; Wei, D.; Yang, I.; Wadehra, M. Exploring the Past, Present, and Future of Anti-Angiogenic Therapy in Glioblastoma. Cancers 2023, 15, 830. [Google Scholar] [CrossRef] [PubMed]
  394. Schmidt, E.E.; Ichimura, K.; Reifenberger, G.; Collins, V.P. CDKN2 (P16/MTS1) Gene Deletion or CDK4 Amplification Occurs in the Majority of Glioblastomas. Cancer Res. 1994, 54, 6321–6324. [Google Scholar] [PubMed]
  395. Michaud, K.; Solomon, D.A.; Oermann, E.; Kim, J.-S.; Zhong, W.-Z.; Prados, M.D.; Ozawa, T.; James, C.D.; Waldman, T. Pharmacologic Inhibition of Cdk4/6 Arrests the Growth of Glioblastoma Multiforme Intracranial Xenografts. Cancer Res. 2010, 70, 3228–3238. [Google Scholar] [CrossRef]
  396. Olmez, I.; Brenneman, B.; Xiao, A.; Serbulea, V.; Benamar, M.; Zhang, Y.; Manigat, L.; Abbas, T.; Lee, J.; Nakano, I.; et al. Combined CDK4/6 mTOR Inhibition Is Synergistic against Glioblastoma via Multiple Mechanisms. Clin. Cancer Res. 2017, 23, 6958–6968. [Google Scholar] [CrossRef]
  397. Giordano, F.; D’Amico, M.; Montalto, F.I.; Malivindi, R.; Chimento, A.; Conforti, F.L.; Pezzi, V.; Panno, M.L.; Andò, S.; De Amicis, F. Cdk4 Regulates Glioblastoma Cell Invasion and Stemness and Is Target of a Notch Inhibitor Plus Resveratrol Combined Treatment. Int. J. Mol. Sci. 2023, 24, 10094. [Google Scholar] [CrossRef] [PubMed]
  398. Li, J.; Jiang, J.; Wu, J.; Bao, X.; Sanai, N. Physiologically Based Pharmacokinetic Modeling of Central Nervous System Pharmacokinetics of CDK4/6 Inhibitors to Guide Selection of Drug and Dosing Regimen for Brain Cancer Treatment. Clin. Pharmacol. Ther. 2021, 109, 494–506. [Google Scholar] [CrossRef]
  399. Taylor, J.W.; Parikh, M.; Phillips, J.J.; James, C.D.; Molinaro, A.M.; Butowski, N.A.; Clarke, J.L.; Oberheim-Bush, N.A.; Chang, S.M.; Berger, M.S.; et al. Phase-2 Trial of Palbociclib in Adult Patients with Recurrent RB1-Positive Glioblastoma. J. Neurooncol. 2018, 140, 477–483. [Google Scholar] [CrossRef]
  400. Tien, A.-C.; Li, J.; Bao, X.; Derogatis, A.; Kim, S.; Mehta, S.; Sanai, N. A Phase 0 Trial of Ribociclib in Recurrent Glioblastoma Patients Incorporating a Tumor Pharmacodynamic- and Pharmacokinetic-Guided Expansion Cohort. Clin. Cancer Res. 2019, 25, 5777–5786. [Google Scholar] [CrossRef]
  401. Xiao, J.; Liang, J.; Fan, J.; Hou, P.; Li, X.; Zhang, H.; Li, K.; Bu, L.; Li, P.; He, M.; et al. CDK4/6 Inhibition Enhances Oncolytic Virus Efficacy by Potentiating Tumor-Selective Cell Killing and T-Cell Activation in Refractory Glioblastoma. Cancer Res. 2022, 82, 3359–3374. [Google Scholar] [CrossRef]
  402. Lee, E.Q.; Trippa, L.; Fell, G.; Rahman, R.; Arrillaga-Romany, I.; Touat, M.; Drappatz, J.; Welch, M.R.; Galanis, E.; Ahluwalia, M.S.; et al. Preliminary Results of the Abemaciclib Arm in the Individualized Screening Trial of Innovative Glioblastoma Therapy (INSIGhT): A Phase II Platform Trial Using Bayesian Adaptive Randomization. JCO 2021, 39, 2014. [Google Scholar] [CrossRef]
  403. Rahman, R.; Trippa, L.; Lee, E.Q.; Arrillaga-Romany, I.; Fell, G.; Touat, M.; McCluskey, C.; Wiley, J.; Gaffey, S.; Drappatz, J.; et al. Inaugural Results of the Individualized Screening Trial of Innovative Glioblastoma Therapy: A Phase II Platform Trial for Newly Diagnosed Glioblastoma Using Bayesian Adaptive Randomization. J. Clin. Oncol. 2023, 41, 5524–5535. [Google Scholar] [CrossRef]
  404. Reardon, D.A.; Nabors, L.B.; Mason, W.P.; Perry, J.R.; Shapiro, W.; Kavan, P.; Mathieu, D.; Phuphanich, S.; Cseh, A.; Fu, Y.; et al. Phase I/Randomized Phase II Study of Afatinib, an Irreversible ErbB Family Blocker, with or without Protracted Temozolomide in Adults with Recurrent Glioblastoma. Neuro Oncol. 2015, 17, 430–439. [Google Scholar] [CrossRef]
  405. Wick, W.; Dettmer, S.; Berberich, A.; Kessler, T.; Karapanagiotou-Schenkel, I.; Wick, A.; Winkler, F.; Pfaff, E.; Brors, B.; Debus, J.; et al. N2M2 (NOA-20) Phase I/II Trial of Molecularly Matched Targeted Therapies plus Radiotherapy in Patients with Newly Diagnosed Non-MGMT Hypermethylated Glioblastoma. Neuro Oncol. 2019, 21, 95–105. [Google Scholar] [CrossRef]
  406. Altwairgi, A.K.; Alghareeb, W.A.; AlNajjar, F.H.; Alhussain, H.; Alsaeed, E.; Balbaid, A.A.O.; Aldanan, S.; Orz, Y.; Alsharm, A.A. Atorvastatin in Combination with Radiotherapy and Temozolomide for Glioblastoma: A Prospective Phase II Study. Investig. New Drugs 2021, 39, 226–231. [Google Scholar] [CrossRef] [PubMed]
  407. Awada, G.; Ben Salama, L.; De Cremer, J.; Schwarze, J.K.; Fischbuch, L.; Seynaeve, L.; Du Four, S.; Vanbinst, A.-M.; Michotte, A.; Everaert, H.; et al. Axitinib plus Avelumab in the Treatment of Recurrent Glioblastoma: A Stratified, Open-Label, Single-Center Phase 2 Clinical Trial (GliAvAx). J. Immunother. Cancer 2020, 8, e001146. [Google Scholar] [CrossRef] [PubMed]
  408. Duerinck, J.; Du Four, S.; Bouttens, F.; Andre, C.; Verschaeve, V.; Van Fraeyenhove, F.; Chaskis, C.; D’Haene, N.; Le Mercier, M.; Rogiers, A.; et al. Randomized Phase II Trial Comparing Axitinib with the Combination of Axitinib and Lomustine in Patients with Recurrent Glioblastoma. J. Neurooncol. 2018, 136, 115–125. [Google Scholar] [CrossRef] [PubMed]
  409. Herrlinger, U.; Schäfer, N.; Steinbach, J.P.; Weyerbrock, A.; Hau, P.; Goldbrunner, R.; Friedrich, F.; Rohde, V.; Ringel, F.; Schlegel, U.; et al. Bevacizumab Plus Irinotecan Versus Temozolomide in Newly Diagnosed O6-Methylguanine–DNA Methyltransferase Nonmethylated Glioblastoma: The Randomized GLARIUS Trial. JCO 2016, 34, 1611–1619. [Google Scholar] [CrossRef] [PubMed]
  410. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab Alone and in Combination With Irinotecan in Recurrent Glioblastoma. J. Clin. Oncol. 2023, 41, 4945–4952. [Google Scholar] [CrossRef] [PubMed]
  411. Hofland, K.F.; Hansen, S.; Sorensen, M.; Engelholm, S.; Schultz, H.P.; Muhic, A.; Grunnet, K.; Ask, A.; Costa, J.C.; Kristiansen, C.; et al. Neoadjuvant Bevacizumab and Irinotecan versus Bevacizumab and Temozolomide Followed by Concomitant Chemoradiotherapy in Newly Diagnosed Glioblastoma Multiforme: A Randomized Phase II Study. Acta Oncol. 2014, 53, 939–944. [Google Scholar] [CrossRef]
  412. Peters, K.B.; Lou, E.; Desjardins, A.; Reardon, D.A.; Lipp, E.S.; Miller, E.; Herndon, J.E.; McSherry, F.; Friedman, H.S.; Vredenburgh, J.J. Phase II Trial of Upfront Bevacizumab, Irinotecan, and Temozolomide for Unresectable Glioblastoma. Oncologist 2015, 20, 727–728. [Google Scholar] [CrossRef]
  413. Cloughesy, T.; Finocchiaro, G.; Belda-Iniesta, C.; Recht, L.; Brandes, A.A.; Pineda, E.; Mikkelsen, T.; Chinot, O.L.; Balana, C.; Macdonald, D.R.; et al. Randomized, Double-Blind, Placebo-Controlled, Multicenter Phase II Study of Onartuzumab Plus Bevacizumab Versus Placebo Plus Bevacizumab in Patients With Recurrent Glioblastoma: Efficacy, Safety, and Hepatocyte Growth Factor and O6-Methylguanine-DNA Methyltransferase Biomarker Analyses. J. Clin. Oncol. 2017, 35, 343–351. [Google Scholar] [CrossRef] [PubMed]
  414. Tsien, C.I.; Pugh, S.L.; Dicker, A.P.; Raizer, J.J.; Matuszak, M.M.; Lallana, E.C.; Huang, J.; Algan, O.; Deb, N.; Portelance, L.; et al. NRG Oncology/RTOG1205: A Randomized Phase II Trial of Concurrent Bevacizumab and Reirradiation Versus Bevacizumab Alone as Treatment for Recurrent Glioblastoma. J. Clin. Oncol. 2023, 41, 1285–1295. [Google Scholar] [CrossRef]
  415. Odia, Y.; Sul, J.; Shih, J.H.; Kreisl, T.N.; Butman, J.A.; Iwamoto, F.M.; Fine, H.A. A Phase II Trial of Tandutinib (MLN 518) in Combination with Bevacizumab for Patients with Recurrent Glioblastoma. CNS Oncol. 2016, 5, 59–67. [Google Scholar] [CrossRef]
  416. Desjardins, A.; Reardon, D.A.; Coan, A.; Marcello, J.; Herndon, J.E.; Bailey, L.; Peters, K.B.; Friedman, H.S.; Vredenburgh, J.J. Bevacizumab and Daily Temozolomide for Recurrent Glioblastoma. Cancer 2012, 118, 1302–1312. [Google Scholar] [CrossRef]
  417. Wen, P.Y.; Touat, M.; Alexander, B.M.; Mellinghoff, I.K.; Ramkissoon, S.; McCluskey, C.S.; Pelton, K.; Haidar, S.; Basu, S.S.; Gaffey, S.C.; et al. Buparlisib in Patients With Recurrent Glioblastoma Harboring Phosphatidylinositol 3-Kinase Pathway Activation: An Open-Label, Multicenter, Multi-Arm, Phase II Trial. J. Clin. Oncol. 2019, 37, 741–750. [Google Scholar] [CrossRef]
  418. Hainsworth, J.D.; Becker, K.P.; Mekhail, T.; Chowdhary, S.A.; Eakle, J.F.; Wright, D.; Langdon, R.M.; Yost, K.J.; Padula, G.D.A.; West-Osterfield, K.; et al. Phase I/II Study of Bevacizumab with BKM120, an Oral PI3K Inhibitor, in Patients with Refractory Solid Tumors (Phase I) and Relapsed/Refractory Glioblastoma (Phase II). J. Neurooncol. 2019, 144, 303–311. [Google Scholar] [CrossRef]
  419. Rosenthal, M.; Clement, P.M.; Campone, M.; Gil-Gil, M.J.; DeGroot, J.; Chinot, O.; Idbaih, A.; Gan, H.; Raizer, J.; Wen, P.Y.; et al. Buparlisib plus Carboplatin or Lomustine in Patients with Recurrent Glioblastoma: A Phase Ib/II, Open-Label, Multicentre, Randomised Study. ESMO Open 2020, 5, e000672. [Google Scholar] [CrossRef] [PubMed]
  420. van den Bent, M.; Azaro, A.; De Vos, F.; Sepulveda, J.; Yung, W.K.A.; Wen, P.Y.; Lassman, A.B.; Joerger, M.; Tabatabai, G.; Rodon, J.; et al. A Phase Ib/II, Open-Label, Multicenter Study of INC280 (Capmatinib) Alone and in Combination with Buparlisib (BKM120) in Adult Patients with Recurrent Glioblastoma. J. Neurooncol. 2020, 146, 79–89. [Google Scholar] [CrossRef]
  421. Batchelor, T.T.; Won, M.; Chakravarti, A.; Hadjipanayis, C.G.; Shi, W.; Ashby, L.S.; Stieber, V.W.; Robins, H.I.; Gray, H.J.; Voloschin, A.; et al. NRG/RTOG 0837: Randomized, Phase II, Double-Blind, Placebo-Controlled Trial of Chemoradiation with or without Cediranib in Newly Diagnosed Glioblastoma. Neurooncol. Adv 2023, 5, vdad116. [Google Scholar] [CrossRef]
  422. Batchelor, T.T.; Mulholland, P.; Neyns, B.; Nabors, L.B.; Campone, M.; Wick, A.; Mason, W.; Mikkelsen, T.; Phuphanich, S.; Ashby, L.S.; et al. Phase III Randomized Trial Comparing the Efficacy of Cediranib As Monotherapy, and in Combination With Lomustine, Versus Lomustine Alone in Patients With Recurrent Glioblastoma. J. Clin. Oncol. 2013, 31, 3212–3218. [Google Scholar] [CrossRef]
  423. Combs, S.E.; Schulz-Ertner, D.; Hartmann, C.; Welzel, T.; Timke, C.; Herfarth, K.; von Deimling, A.; Edler, L.; Platten, M.; Wick, W.; et al. Phase I/II Study of Cetuximab plus Temozolomide as Radiochemotherapy for Primary Glioblastoma (GERT)—Eudract Number 2005–003911–63; NCT00311857. JCO 2008, 26, 2077. [Google Scholar] [CrossRef]
  424. Hasselbalch, B.; Lassen, U.; Hansen, S.; Holmberg, M.; Sørensen, M.; Kosteljanetz, M.; Broholm, H.; Stockhausen, M.-T.; Poulsen, H.S. Cetuximab, Bevacizumab, and Irinotecan for Patients with Primary Glioblastoma and Progression after Radiation Therapy and Temozolomide: A Phase II Trial. Neuro Oncol. 2010, 12, 508–516. [Google Scholar] [CrossRef]
  425. Lv, S.; Teugels, E.; Sadones, J.; De Brakeleer, S.; Duerinck, J.; Du Four, S.; Michotte, A.; De Grève, J.; Neyns, B. Correlation of EGFR, IDH1 and PTEN Status with the Outcome of Patients with Recurrent Glioblastoma Treated in a Phase II Clinical Trial with the EGFR-Blocking Monoclonal Antibody Cetuximab. Int. J. Oncol. 2012, 41, 1029–1035. [Google Scholar] [CrossRef]
  426. Arrillaga-Romany, I.; Sahebjam, S.; Picconi, D.; Campian, J.; Giglio, P.; Drappatz, J.; Aiken, R.; Villano, J.; Lee, E.; Welch, M.; et al. ACTR-61. A RANDOMIZED PHASE 2 TRIAL OF CEDIRANIB IN COMBINATION WITH OLAPARIB VERSUS BEVACIZUMAB IN PATIENTS WITH RECURRENT GLIOBLASTOMA. Neuro Oncol. 2019, 21, vi27. [Google Scholar] [CrossRef]
  427. Sepúlveda-Sánchez, J.M.; Vaz, M.Á.; Balañá, C.; Gil-Gil, M.; Reynés, G.; Gallego, Ó.; Martínez-García, M.; Vicente, E.; Quindós, M.; Luque, R.; et al. Phase II Trial of Dacomitinib, a Pan-Human EGFR Tyrosine Kinase Inhibitor, in Recurrent Glioblastoma Patients with EGFR Amplification. Neuro Oncol. 2017, 19, 1522–1531. [Google Scholar] [CrossRef]
  428. Lassman, A.B.; Pugh, S.L.; Gilbert, M.R.; Aldape, K.D.; Geinoz, S.; Beumer, J.H.; Christner, S.M.; Komaki, R.; DeAngelis, L.M.; Gaur, R.; et al. Phase 2 Trial of Dasatinib in Target-Selected Patients with Recurrent Glioblastoma (RTOG 0627). Neuro. Oncol. 2015, 17, 992–998. [Google Scholar] [CrossRef] [PubMed]
  429. Van Den Bent, M.; Eoli, M.; Sepulveda, J.M.; Smits, M.; Walenkamp, A.; Frenel, J.-S.; Franceschi, E.; Clement, P.M.; Chinot, O.; De Vos, F.; et al. INTELLANCE 2/EORTC 1410 Randomized Phase II Study of Depatux-M Alone and with Temozolomide vs Temozolomide or Lomustine in Recurrent EGFR Amplified Glioblastoma. Neuro Oncol 2020, 22, 684–693. [Google Scholar] [CrossRef] [PubMed]
  430. Lassman, A.B.; Pugh, S.L.; Wang, T.J.C.; Aldape, K.; Gan, H.K.; Preusser, M.; Vogelbaum, M.A.; Sulman, E.P.; Won, M.; Zhang, P.; et al. Depatuxizumab Mafodotin in EGFR-Amplified Newly Diagnosed Glioblastoma: A Phase III Randomized Clinical Trial. Neuro Oncol. 2022, 25, 339–350. [Google Scholar] [CrossRef]
  431. Sharma, M.; Schilero, C.; Peereboom, D.M.; Hobbs, B.P.; Elson, P.; Stevens, G.H.J.; McCrae, K.; Nixon, A.B.; Ahluwalia, M.S. Phase II Study of Dovitinib in Recurrent Glioblastoma. J. Neurooncol. 2019, 144, 359–368. [Google Scholar] [CrossRef] [PubMed]
  432. Subbiah, V.; Kreitman, R.J.; Wainberg, Z.A.; Gazzah, A.; Lassen, U.; Stein, A.; Wen, P.Y.; Dietrich, S.; de Jonge, M.J.A.; Blay, J.-Y.; et al. Dabrafenib plus Trametinib in BRAFV600E-Mutated Rare Cancers: The Phase 2 ROAR Trial. Nat. Med. 2023, 29, 1103–1112. [Google Scholar] [CrossRef]
  433. Wick, W.; Puduvalli, V.K.; Chamberlain, M.C.; van den Bent, M.J.; Carpentier, A.F.; Cher, L.M.; Mason, W.; Weller, M.; Hong, S.; Musib, L.; et al. Phase III Study of Enzastaurin Compared with Lomustine in the Treatment of Recurrent Intracranial Glioblastoma. J. Clin. Oncol. 2010, 28, 1168–1174. [Google Scholar] [CrossRef]
  434. Yung, W.K.A.; Vredenburgh, J.J.; Cloughesy, T.F.; Nghiemphu, P.; Klencke, B.; Gilbert, M.R.; Reardon, D.A.; Prados, M.D. Safety and Efficacy of Erlotinib in First-Relapse Glioblastoma: A Phase II Open-Label Study. Neuro Oncol. 2010, 12, 1061–1070. [Google Scholar] [CrossRef]
  435. van den Bent, M.J.; Brandes, A.A.; Rampling, R.; Kouwenhoven, M.C.M.; Kros, J.M.; Carpentier, A.F.; Clement, P.M.; Frenay, M.; Campone, M.; Baurain, J.-F.; et al. Randomized Phase II Trial of Erlotinib versus Temozolomide or Carmustine in Recurrent Glioblastoma: EORTC Brain Tumor Group Study 26034. J Clin Oncol 2009, 27, 1268–1274. [Google Scholar] [CrossRef]
  436. D’Alessandris, Q.G.; Montano, N.; Cenci, T.; Martini, M.; Lauretti, L.; Bianchi, F.; Larocca, L.M.; Maira, G.; Fernandez, E.; Pallini, R. Targeted Therapy with Bevacizumab and Erlotinib Tailored to the Molecular Profile of Patients with Recurrent Glioblastoma. Preliminary Experience. Acta Neurochir. 2013, 155, 33–40. [Google Scholar] [CrossRef]
  437. Raizer, J.J.; Giglio, P.; Hu, J.; Groves, M.; Merrell, R.; Conrad, C.; Phuphanich, S.; Puduvalli, V.K.; Loghin, M.; Paleologos, N.; et al. A Phase II Study of Bevacizumab and Erlotinib after Radiation and Temozolomide in MGMT Unmethylated GBM Patients. J. Neurooncol. 2016, 126, 185–192. [Google Scholar] [CrossRef]
  438. Prados, M.D.; Chang, S.M.; Butowski, N.; DeBoer, R.; Parvataneni, R.; Carliner, H.; Kabuubi, P.; Ayers-Ringler, J.; Rabbitt, J.; Page, M.; et al. Phase II Study of Erlotinib plus Temozolomide during and after Radiation Therapy in Patients with Newly Diagnosed Glioblastoma Multiforme or Gliosarcoma. J. Clin. Oncol. 2009, 27, 579–584. [Google Scholar] [CrossRef] [PubMed]
  439. Peereboom, D.M.; Shepard, D.R.; Ahluwalia, M.S.; Brewer, C.J.; Agarwal, N.; Stevens, G.H.J.; Suh, J.H.; Toms, S.A.; Vogelbaum, M.A.; Weil, R.J.; et al. Phase II Trial of Erlotinib with Temozolomide and Radiation in Patients with Newly Diagnosed Glioblastoma Multiforme. J. Neurooncol. 2010, 98, 93–99. [Google Scholar] [CrossRef]
  440. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; Gururangan, S.; Friedman, A.H.; Herndon, J.E.; Marcello, J.; Norfleet, J.A.; McLendon, R.E.; Sampson, J.H.; et al. Phase 2 Trial of Erlotinib plus Sirolimus in Adults with Recurrent Glioblastoma. J. Neurooncol. 2010, 96, 219–230. [Google Scholar] [CrossRef] [PubMed]
  441. Peereboom, D.M.; Ahluwalia, M.S.; Ye, X.; Supko, J.G.; Hilderbrand, S.L.; Phuphanich, S.; Nabors, L.B.; Rosenfeld, M.R.; Mikkelsen, T.; Grossman, S.A.; et al. NABTT 0502: A Phase II and Pharmacokinetic Study of Erlotinib and Sorafenib for Patients with Progressive or Recurrent Glioblastoma Multiforme. Neuro Oncol 2013, 15, 490–496. [Google Scholar] [CrossRef] [PubMed]
  442. Chinnaiyan, P.; Won, M.; Wen, P.Y.; Rojiani, A.M.; Werner-Wasik, M.; Shih, H.A.; Ashby, L.S.; Michael Yu, H.-H.; Stieber, V.W.; Malone, S.C.; et al. A Randomized Phase II Study of Everolimus in Combination with Chemoradiation in Newly Diagnosed Glioblastoma: Results of NRG Oncology RTOG 0913. Neuro Oncol 2018, 20, 666–673. [Google Scholar] [CrossRef]
  443. Hainsworth, J.D.; Shih, K.C.; Shepard, G.C.; Tillinghast, G.W.; Brinker, B.T.; Spigel, D.R. Phase II Study of Concurrent Radiation Therapy, Temozolomide, and Bevacizumab Followed by Bevacizumab/Everolimus as First-Line Treatment for Patients with Glioblastoma. Clin. Adv. Hematol. Oncol. 2012, 10, 240–246. [Google Scholar]
  444. Hegi, M.E.; Diserens, A.-C.; Bady, P.; Kamoshima, Y.; Kouwenhoven, M.C.M.; Delorenzi, M.; Lambiv, W.L.; Hamou, M.-F.; Matter, M.S.; Koch, A.; et al. Pathway Analysis of Glioblastoma Tissue after Preoperative Treatment with the EGFR Tyrosine Kinase Inhibitor Gefitinib--a Phase II Trial. Mol. Cancer Ther. 2011, 10, 1102–1112. [Google Scholar] [CrossRef]
  445. Rich, J.N.; Reardon, D.A.; Peery, T.; Dowell, J.M.; Quinn, J.A.; Penne, K.L.; Wikstrand, C.J.; Van Duyn, L.B.; Dancey, J.E.; McLendon, R.E.; et al. Phase II Trial of Gefitinib in Recurrent Glioblastoma. J. Clin. Oncol. 2004, 22, 133–142. [Google Scholar] [CrossRef]
  446. Brown, N.; McBain, C.; Nash, S.; Hopkins, K.; Sanghera, P.; Saran, F.; Phillips, M.; Dungey, F.; Clifton-Hadley, L.; Wanek, K.; et al. Multi-Center Randomized Phase II Study Comparing Cediranib plus Gefitinib with Cediranib plus Placebo in Subjects with Recurrent/Progressive Glioblastoma. PLoS ONE 2016, 11, e0156369. [Google Scholar] [CrossRef]
  447. Chakravarti, A.; Wang, M.; Robins, H.I.; Lautenschlaeger, T.; Curran, W.J.; Brachman, D.G.; Schultz, C.J.; Choucair, A.; Dolled-Filhart, M.; Christiansen, J.; et al. RTOG 0211: A Phase 1/2 Study of Radiation Therapy with Concurrent Gefitinib for Newly Diagnosed Glioblastoma Patients. Int J Radiat Oncol Biol Phys 2013, 85, 1206–1211. [Google Scholar] [CrossRef]
  448. Sautter, L.; Hofheinz, R.; Tuettenberg, J.; Grimm, M.; Vajkoczy, P.; Groden, C.; Schmieder, K.; Hochhaus, A.; Wenz, F.; Giordano, F.A. Open-Label Phase II Evaluation of Imatinib in Primary Inoperable or Incompletely Resected and Recurrent Glioblastoma. Oncology 2020, 98, 16–22. [Google Scholar] [CrossRef]
  449. Dresemann, G.; Weller, M.; Rosenthal, M.A.; Wedding, U.; Wagner, W.; Engel, E.; Heinrich, B.; Mayer-Steinacker, R.; Karup-Hansen, A.; Fluge, O.; et al. Imatinib in Combination with Hydroxyurea versus Hydroxyurea Alone as Oral Therapy in Patients with Progressive Pretreated Glioblastoma Resistant to Standard Dose Temozolomide. J. Neurooncol. 2010, 96, 393–402. [Google Scholar] [CrossRef] [PubMed]
  450. Phuphanich, S.; Raizer, J.; Chamberlain, M.; Canelos, P.; Narwal, R.; Hong, S.; Miday, R.; Nade, M.; Laubscher, K. Phase II Study of MEDI-575, an Anti-Platelet-Derived Growth Factor-α Antibody, in Patients with Recurrent Glioblastoma. J. Neurooncol. 2017, 131, 185–191. [Google Scholar] [CrossRef] [PubMed]
  451. Westphal, M.; Heese, O.; Steinbach, J.P.; Schnell, O.; Schackert, G.; Mehdorn, M.; Schulz, D.; Simon, M.; Schlegel, U.; Senft, C.; et al. A Randomised, Open Label Phase III Trial with Nimotuzumab, an Anti-Epidermal Growth Factor Receptor Monoclonal Antibody in the Treatment of Newly Diagnosed Adult Glioblastoma. Eur. J. Cancer 2015, 51, 522–532. [Google Scholar] [CrossRef] [PubMed]
  452. Iwamoto, F.M.; Lamborn, K.R.; Robins, H.I.; Mehta, M.P.; Chang, S.M.; Butowski, N.A.; Deangelis, L.M.; Abrey, L.E.; Zhang, W.-T.; Prados, M.D.; et al. Phase II Trial of Pazopanib (GW786034), an Oral Multi-Targeted Angiogenesis Inhibitor, for Adults with Recurrent Glioblastoma (North American Brain Tumor Consortium Study 06-02). Neuro Oncol. 2010, 12, 855–861. [Google Scholar] [CrossRef]
  453. Kaley, T.J.; Panageas, K.S.; Mellinghoff, I.K.; Nolan, C.; Gavrilovic, I.T.; DeAngelis, L.M.; Abrey, L.E.; Holland, E.C.; Lassman, A.B. Phase II Trial of an AKT Inhibitor (Perifosine) for Recurrent Glioblastoma. J. Neurooncol. 2019, 144, 403–407. [Google Scholar] [CrossRef] [PubMed]
  454. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib Compared with Lomustine in Patients with Relapsed Glioblastoma (REGOMA): A Multicentre, Open-Label, Randomised, Controlled, Phase 2 Trial. Lancet Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef] [PubMed]
  455. Galanis, E.; Anderson, S.K.; Lafky, J.M.; Uhm, J.H.; Giannini, C.; Kumar, S.K.; Kimlinger, T.K.; Northfelt, D.W.; Flynn, P.J.; Jaeckle, K.A.; et al. Phase II Study of Bevacizumab in Combination with Sorafenib in Recurrent Glioblastoma (N0776): A North Central Cancer Treatment Group Trial. Clin. Cancer Res. 2013, 19, 4816–4823. [Google Scholar] [CrossRef] [PubMed]
  456. Zustovich, F.; Landi, L.; Lombardi, G.; Porta, C.; Galli, L.; Fontana, A.; Amoroso, D.; Galli, C.; Andreuccetti, M.; Falcone, A.; et al. Sorafenib plus Daily Low-Dose Temozolomide for Relapsed Glioblastoma: A Phase II Study. Anticancer. Res. 2013, 33, 3487–3494. [Google Scholar] [CrossRef]
  457. Hainsworth, J.D.; Ervin, T.; Friedman, E.; Priego, V.; Murphy, P.B.; Clark, B.L.; Lamar, R.E. Concurrent Radiotherapy and Temozolomide Followed by Temozolomide and Sorafenib in the First-Line Treatment of Patients with Glioblastoma Multiforme. Cancer 2010, 116, 3663–3669. [Google Scholar] [CrossRef]
  458. Hutterer, M.; Nowosielski, M.; Haybaeck, J.; Embacher, S.; Stockhammer, F.; Gotwald, T.; Holzner, B.; Capper, D.; Preusser, M.; Marosi, C.; et al. A Single-Arm Phase II Austrian/German Multicenter Trial on Continuous Daily Sunitinib in Primary Glioblastoma at First Recurrence (SURGE 01-07). Neuro Oncol. 2014, 16, 92–102. [Google Scholar] [CrossRef]
  459. Balaña, C.; Gil, M.J.; Perez, P.; Reynes, G.; Gallego, O.; Ribalta, T.; Capellades, J.; Gonzalez, S.; Verger, E. Sunitinib Administered Prior to Radiotherapy in Patients with Non-Resectable Glioblastoma: Results of a Phase II Study. Target. Oncol. 2014, 9, 321–329. [Google Scholar] [CrossRef]
  460. Faye, M.D.; Easaw, J.; De Robles, P.; Agnihotram, R.; Torres-Vasquez, A.; Lamonde, F.; Petrecca, K.; Owen, S.; Panet-Raymond, V.; Shenouda, G.; et al. Phase II Trial of Concurrent Sunitinib, Temozolomide, and Radiotherapy with Adjuvant Temozolomide for Newly Diagnosed MGMT Unmethylated Glioblastoma. Neuro-Oncology Adv. 2023, 5, vdad106. [Google Scholar] [CrossRef]
  461. Chang, S.M.; Wen, P.; Cloughesy, T.; Greenberg, H.; Schiff, D.; Conrad, C.; Fink, K.; Robins, H.I.; De Angelis, L.; Raizer, J.; et al. Phase II Study of CCI-779 in Patients with Recurrent Glioblastoma Multiforme. Investig. New Drugs 2005, 23, 357–361. [Google Scholar] [CrossRef]
  462. Galanis, E.; Buckner, J.C.; Maurer, M.J.; Kreisberg, J.I.; Ballman, K.; Boni, J.; Peralba, J.M.; Jenkins, R.B.; Dakhil, S.R.; Morton, R.F.; et al. Phase II Trial of Temsirolimus (CCI-779) in Recurrent Glioblastoma Multiforme: A North Central Cancer Treatment Group Study. J. Clin. Oncol. 2005, 23, 5294–5304. [Google Scholar] [CrossRef]
  463. Wick, W.; Gorlia, T.; Bady, P.; Platten, M.; van den Bent, M.J.; Taphoorn, M.J.B.; Steuve, J.; Brandes, A.A.; Hamou, M.-F.; Wick, A.; et al. Phase II Study of Radiotherapy and Temsirolimus versus Radiochemotherapy with Temozolomide in Patients with Newly Diagnosed Glioblastoma without MGMT Promoter Hypermethylation (EORTC 26082). Clin. Cancer Res. 2016, 22, 4797–4806. [Google Scholar] [CrossRef] [PubMed]
  464. Lassen, U.; Sorensen, M.; Gaziel, T.B.; Hasselbalch, B.; Poulsen, H.S. Phase II Study of Bevacizumab and Temsirolimus Combination Therapy for Recurrent Glioblastoma Multiforme. Anticancer. Res. 2013, 33, 1657–1660. [Google Scholar] [PubMed]
  465. Kreisl, T.N.; McNeill, K.A.; Sul, J.; Iwamoto, F.M.; Shih, J.; Fine, H.A. A Phase I/II Trial of Vandetanib for Patients with Recurrent Malignant Glioma. Neuro Oncol. 2012, 14, 1519–1526. [Google Scholar] [CrossRef] [PubMed]
  466. Lee, E.Q.; Kaley, T.J.; Duda, D.G.; Schiff, D.; Lassman, A.B.; Wong, E.T.; Mikkelsen, T.; Purow, B.W.; Muzikansky, A.; Ancukiewicz, M.; et al. A Multicenter, Phase II, Randomized, Noncomparative Clinical Trial of Radiation and Temozolomide with or without Vandetanib in Newly Diagnosed Glioblastoma Patients. Clin. Cancer Res. 2015, 21, 3610–3618. [Google Scholar] [CrossRef] [PubMed]
  467. Robins, H.I.; Zhang, P.; Gilbert, M.R.; Chakravarti, A.; de Groot, J.F.; Grimm, S.A.; Wang, F.; Lieberman, F.S.; Krauze, A.; Trotti, A.M.; et al. A Randomized Phase I/II Study of ABT-888 in Combination with Temozolomide in Recurrent Temozolomide Resistant Glioblastoma: An NRG Oncology RTOG Group Study. J. Neurooncol. 2016, 126, 309–316. [Google Scholar] [CrossRef] [PubMed]
  468. Sim, H.-W.; McDonald, K.L.; Lwin, Z.; Barnes, E.H.; Rosenthal, M.; Foote, M.C.; Koh, E.-S.; Back, M.; Wheeler, H.; Sulman, E.P.; et al. A Randomized Phase II Trial of Veliparib, Radiotherapy, and Temozolomide in Patients with Unmethylated MGMT Glioblastoma: The VERTU Study. Neuro Oncol. 2021, 23, 1736–1749. [Google Scholar] [CrossRef] [PubMed]
  469. Sarkaria, J.N.; Ballman, K.V.; Kizilbash, S.H.; Sulman, E.P.; Giannini, C.; Mashru, S.H.; Piccioni, D.E.; Friday, B.E.B.; Dixon, J.G.; Kabat, B.; et al. Randomized Phase II/III Trial of Veliparib or Placebo in Combination with Adjuvant Temozolomide in Newly Diagnosed Glioblastoma (GBM) Patients with MGMT Promoter Hypermethylation (Alliance A071102). JCO 2022, 40, 2001. [Google Scholar] [CrossRef]
  470. Mao, H.; Lebrun, D.G.; Yang, J.; Zhu, V.F.; Li, M. Deregulated Signaling Pathways in Glioblastoma Multiforme: Molecular Mechanisms and Therapeutic Targets. Cancer Investig. 2012, 30, 48–56. [Google Scholar] [CrossRef]
  471. Solomon, D.A.; Kim, J.-S.; Jean, W.; Waldman, T. Conspirators in a Capital Crime: Co-Deletion of p18INK4c and p16INK4a/p14ARF/p15INK4b in Glioblastoma Multiforme. Cancer Res. 2008, 68, 8657–8660. [Google Scholar] [CrossRef]
  472. Lu, Y.; Feng, Y.; Li, Z.; Li, J.; Zhang, H.; Hu, X.; Jiang, W.; Shi, T.; Wang, Z. Novel Piperazine Based Benzamide Derivatives as Potential Anti-Glioblastoma Agents Inhibiting Cell Proliferation and Cell Cycle Progression. Eur. J. Med. Chem. 2022, 227, 113908. [Google Scholar] [CrossRef]
  473. Teodoro, J.G.; Evans, S.K.; Green, M.R. Inhibition of Tumor Angiogenesis by P53: A New Role for the Guardian of the Genome. J. Mol. Med. 2007, 85, 1175–1186. [Google Scholar] [CrossRef] [PubMed]
  474. Nguyen, D.; Liao, W.; Zeng, S.X.; Lu, H. Reviving the Guardian of the Genome: Small Molecule Activators of P53. Pharmacol. Ther. 2017, 178, 92–108. [Google Scholar] [CrossRef] [PubMed]
  475. Nag, S.; Qin, J.; Srivenugopal, K.S.; Wang, M.; Zhang, R. The MDM2-P53 Pathway Revisited. J. Biomed. Res. 2013, 27, 254–271. [Google Scholar] [CrossRef]
  476. Maggi, L.B.; Winkeler, C.L.; Miceli, A.P.; Apicelli, A.J.; Brady, S.N.; Kuchenreuther, M.J.; Weber, J.D. ARF Tumor Suppression in the Nucleolus. Biochim. Biophys. Acta BBA Mol. Basis Dis. 2014, 1842, 831–839. [Google Scholar] [CrossRef] [PubMed]
  477. Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The Multiple Mechanisms That Regulate P53 Activity and Cell Fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef] [PubMed]
  478. Klein, A.M.; De Queiroz, R.M.; Venkatesh, D.; Prives, C. The Roles and Regulation of MDM2 and MDMX: It Is Not Just about P53. Genes Dev. 2021, 35, 575–601. [Google Scholar] [CrossRef] [PubMed]
  479. Sukumar, U.K.; Massoud, T.F.; Paulmurugan, R. P53 Supplementation as a Targeted Cancer Gene Therapy for Glioblastoma. In Glioblastoma Resistance to Chemotherapy: Molecular Mechanisms and Innovative Reversal Strategies; Elsevier: Amsterdam, The Netherlands, 2021; pp. 773–786. ISBN 978-0-12-821567-8. [Google Scholar]
  480. Gao, J.; Aksoy, B.A.; Dogrusoz, U.; Dresdner, G.; Gross, B.; Sumer, S.O.; Sun, Y.; Jacobsen, A.; Sinha, R.; Larsson, E.; et al. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Sci. Signal. 2013, 6. [Google Scholar] [CrossRef] [PubMed]
  481. Costa, B.; Bendinelli, S.; Gabelloni, P.; Da Pozzo, E.; Daniele, S.; Scatena, F.; Vanacore, R.; Campiglia, P.; Bertamino, A.; Gomez-Monterrey, I.; et al. Human Glioblastoma Multiforme: P53 Reactivation by a Novel MDM2 Inhibitor. PLoS ONE 2013, 8, e72281. [Google Scholar] [CrossRef]
  482. Pellot Ortiz, K.I.; Rechberger, J.S.; Nonnenbroich, L.F.; Daniels, D.J.; Sarkaria, J.N. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023, 11, 1879. [Google Scholar] [CrossRef]
  483. Wang, H.; Cai, S.; Bailey, B.J.; Saadatzadeh, M.R.; Ding, J.; Tonsing-Carter, E.; Georgiadis, T.M.; Gunter, T.Z.; Long, E.C.; Minto, R.E.; et al. Combination Therapy in a Xenograft Model of Glioblastoma: Enhancement of the Antitumor Activity of Temozolomide by an MDM2 Antagonist. J. Neurosurg. 2017, 126, 446–459. [Google Scholar] [CrossRef]
  484. Berberich, A.; Kessler, T.; Thomé, C.M.; Pusch, S.; Hielscher, T.; Sahm, F.; Oezen, I.; Schmitt, L.-M.; Ciprut, S.; Hucke, N.; et al. Targeting Resistance against the MDM2 Inhibitor RG7388 in Glioblastoma Cells by the MEK Inhibitor Trametinib. Clin. Cancer Res. 2019, 25, 253–265. [Google Scholar] [CrossRef]
  485. Her, N.-G.; Oh, J.-W.; Oh, Y.J.; Han, S.; Cho, H.J.; Lee, Y.; Ryu, G.H.; Nam, D.-H. Potent Effect of the MDM2 Inhibitor AMG232 on Suppression of Glioblastoma Stem Cells. Cell Death Dis. 2018, 9. [Google Scholar] [CrossRef]
  486. Tovar, C.; Rosinski, J.; Filipovic, Z.; Higgins, B.; Kolinsky, K.; Hilton, H.; Zhao, X.; Vu, B.T.; Qing, W.; Packman, K.; et al. Small-Molecule MDM2 Antagonists Reveal Aberrant P53 Signaling in Cancer: Implications for Therapy. Proc. Natl. Acad. Sci. USA 2006, 103, 1888–1893. [Google Scholar] [CrossRef]
  487. Villalonga-Planells, R.; Coll-Mulet, L.; Martínez-Soler, F.; Castaño, E.; Acebes, J.-J.; Giménez-Bonafé, P.; Gil, J.; Tortosa, A. Activation of P53 by Nutlin-3a Induces Apoptosis and Cellular Senescence in Human Glioblastoma Multiforme. PLoS ONE 2011, 6, e18588. [Google Scholar] [CrossRef]
  488. Rodon, J.; Dienstmann, R.; Serra, V.; Tabernero, J. Development of PI3K Inhibitors: Lessons Learned from Early Clinical Trials. Nat. Rev. Clin. Oncol. 2013, 10, 143–153. [Google Scholar] [CrossRef] [PubMed]
  489. Bleau, A.-M.; Hambardzumyan, D.; Ozawa, T.; Fomchenko, E.I.; Huse, J.T.; Brennan, C.W.; Holland, E.C. PTEN/PI3K/Akt Pathway Regulates the Side Population Phenotype and ABCG2 Activity in Glioma Tumor Stem-like Cells. Cell Stem Cell 2009, 4, 226–235. [Google Scholar] [CrossRef] [PubMed]
  490. Cantley, L.C. The Phosphoinositide 3-Kinase Pathway. Science 2002, 296, 1655–1657. [Google Scholar] [CrossRef] [PubMed]
  491. Mellinghoff, I.K.; Wang, M.Y.; Vivanco, I.; Haas-Kogan, D.A.; Zhu, S.; Dia, E.Q.; Lu, K.V.; Yoshimoto, K.; Huang, J.H.Y.; Chute, D.J.; et al. Molecular Determinants of the Response of Glioblastomas to EGFR Kinase Inhibitors. N. Engl. J. Med. 2005, 353, 2012–2024. [Google Scholar] [CrossRef] [PubMed]
  492. Jhanwar-Uniyal, M.; Dominguez, J.F.; Mohan, A.L.; Tobias, M.E.; Gandhi, C.D. Disentangling the Signaling Pathways of mTOR Complexes, mTORC1 and mTORC2, as a Therapeutic Target in Glioblastoma. Adv. Biol. Regul. 2022, 83, 100854. [Google Scholar] [CrossRef] [PubMed]
  493. Song, M.S.; Salmena, L.; Pandolfi, P.P. The Functions and Regulation of the PTEN Tumour Suppressor. Nat. Rev. Mol. Cell Biol. 2012, 13, 283–296. [Google Scholar] [CrossRef] [PubMed]
  494. Chen, L.; Guo, D. The Functions of Tumor Suppressor PTEN in Innate and Adaptive Immunity. Cell. Mol. Immunol. 2017, 14, 581–589. [Google Scholar] [CrossRef] [PubMed]
  495. Laplante, M.; Sabatini, D.M. mTOR Signaling in Growth Control and Disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef] [PubMed]
  496. Colardo, M.; Segatto, M.; Di Bartolomeo, S. Targeting RTK-PI3K-mTOR Axis in Gliomas: An Update. Int. J. Mol. Sci. 2021, 22, 4899. [Google Scholar] [CrossRef]
  497. Peng, Y.; Wang, Y.; Zhou, C.; Mei, W.; Zeng, C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front. Oncol. 2022, 12. [Google Scholar] [CrossRef] [PubMed]
  498. Xia, Q.; Xu, M.; Zhang, P.; Liu, L.; Meng, X.; Dong, L. Therapeutic Potential of Autophagy in Glioblastoma Treatment With Phosphoinositide 3-Kinase/Protein Kinase B/Mammalian Target of Rapamycin Signaling Pathway Inhibitors. Front. Oncol. 2020, 10, 572904. [Google Scholar] [CrossRef] [PubMed]
  499. Zaytseva, Y.Y.; Valentino, J.D.; Gulhati, P.; Mark Evers, B. mTOR Inhibitors in Cancer Therapy. Cancer Lett. 2012, 319, 1–7. [Google Scholar] [CrossRef]
  500. Masui, K.; Tanaka, K.; Ikegami, S.; Villa, G.R.; Yang, H.; Yong, W.H.; Cloughesy, T.F.; Yamagata, K.; Arai, N.; Cavenee, W.K.; et al. Glucose-Dependent Acetylation of Rictor Promotes Targeted Cancer Therapy Resistance. Proc. Natl. Acad. Sci. USA 2015, 112, 9406–9411. [Google Scholar] [CrossRef] [PubMed]
  501. Bolcaen, J.; Nair, S.; Driver, C.H.S.; Boshomane, T.M.G.; Ebenhan, T.; Vandevoorde, C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals 2021, 14, 626. [Google Scholar] [CrossRef]
  502. Alexandru, O.; Horescu, C.; Sevastre, A.-S.; Cioc, C.E.; Baloi, C.; Oprita, A.; Dricu, A. Receptor Tyrosine Kinase Targeting in Glioblastoma: Performance, Limitations and Future Approaches. Contemp. Oncol. 2020, 24, 55–66. [Google Scholar] [CrossRef]
  503. Liu, H.; Qiu, W.; Sun, T.; Wang, L.; Du, C.; Hu, Y.; Liu, W.; Feng, F.; Chen, Y.; Sun, H. Therapeutic Strategies of Glioblastoma (GBM): The Current Advances in the Molecular Targets and Bioactive Small Molecule Compounds. Acta Pharm. Sin. B 2022, 12, 1781–1804. [Google Scholar] [CrossRef] [PubMed]
  504. Chen, C.; Zhu, S.; Zhang, X.; Zhou, T.; Gu, J.; Xu, Y.; Wan, Q.; Qi, X.; Chai, Y.; Liu, X.; et al. Targeting the Synthetic Vulnerability of PTEN-Deficient Glioblastoma Cells with MCL1 Inhibitors. Mol. Cancer Ther. 2020, 19, 2001–2011. [Google Scholar] [CrossRef] [PubMed]
  505. Khabibov, M.; Garifullin, A.; Boumber, Y.; Khaddour, K.; Fernandez, M.; Khamitov, F.; Khalikova, L.; Kuznetsova, N.; Kit, O.; Kharin, L. Signaling Pathways and Therapeutic Approaches in Glioblastoma Multiforme (Review). Int. J. Oncol. 2022, 60, 69. [Google Scholar] [CrossRef] [PubMed]
  506. Huang, H.S.; Nagane, M.; Klingbeil, C.K.; Lin, H.; Nishikawa, R.; Ji, X.D.; Huang, C.M.; Gill, G.N.; Wiley, H.S.; Cavenee, W.K. The Enhanced Tumorigenic Activity of a Mutant Epidermal Growth Factor Receptor Common in Human Cancers Is Mediated by Threshold Levels of Constitutive Tyrosine Phosphorylation and Unattenuated Signaling. J. Biol. Chem. 1997, 272, 2927–2935. [Google Scholar] [CrossRef] [PubMed]
  507. Maire, C.L.; Ligon, K.L. Molecular Pathologic Diagnosis of Epidermal Growth Factor Receptor. Neuro Oncol. 2014, 16 (Suppl. S8), viii1-6. [Google Scholar] [CrossRef]
  508. Rodriguez, S.M.B.; Kamel, A.; Ciubotaru, G.V.; Onose, G.; Sevastre, A.-S.; Sfredel, V.; Danoiu, S.; Dricu, A.; Tataranu, L.G. An Overview of EGFR Mechanisms and Their Implications in Targeted Therapies for Glioblastoma. Int. J. Mol. Sci. 2023, 24, 11110. [Google Scholar] [CrossRef]
  509. Eskilsson, E.; Rosland, G.V.; Talasila, K.M.; Knappskog, S.; Keunen, O.; Sottoriva, A.; Foerster, S.; Solecki, G.; Taxt, T.; Jirik, R.; et al. EGFRvIII Mutations Can Emerge as Late and Heterogenous Events in Glioblastoma Development and Promote Angiogenesis through Src Activation. Neuro Oncol. 2016, 18, 1644–1655. [Google Scholar] [CrossRef]
  510. An, Z.; Aksoy, O.; Zheng, T.; Fan, Q.-W.; Weiss, W.A. Epidermal Growth Factor Receptor (EGFR) and EGFRvIII in Glioblastoma (GBM): Signaling Pathways and Targeted Therapies. Oncogene 2018, 37, 1561–1575. [Google Scholar] [CrossRef]
  511. Frederick, L.; Wang, X.Y.; Eley, G.; James, C.D. Diversity and Frequency of Epidermal Growth Factor Receptor Mutations in Human Glioblastomas. Cancer Res. 2000, 60, 1383–1387. [Google Scholar]
  512. Guo, G.; Gong, K.; Wohlfeld, B.; Hatanpaa, K.J.; Zhao, D.; Habib, A.A. Ligand-Independent EGFR Signaling. Cancer Res. 2015, 75, 3436–3441. [Google Scholar] [CrossRef]
  513. Gan, H.K.; Kaye, A.H.; Luwor, R.B. The EGFRvIII Variant in Glioblastoma Multiforme. J. Clin. Neurosci. 2009, 16, 748–754. [Google Scholar] [CrossRef]
  514. Lin, B.; Ziebro, J.; Smithberger, E.; Skinner, K.R.; Zhao, E.; Cloughesy, T.F.; Binder, Z.A.; O’Rourke, D.M.; Nathanson, D.A.; Furnari, F.B.; et al. EGFR, the Lazarus Target for Precision Oncology in Glioblastoma. Neuro-Oncology 2022, 24, 2035–2062. [Google Scholar] [CrossRef]
  515. Sampson, J.H.; Heimberger, A.B.; Archer, G.E.; Aldape, K.D.; Friedman, A.H.; Friedman, H.S.; Gilbert, M.R.; Herndon, J.E.; McLendon, R.E.; Mitchell, D.A.; et al. Immunologic Escape after Prolonged Progression-Free Survival with Epidermal Growth Factor Receptor Variant III Peptide Vaccination in Patients with Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2010, 28, 4722–4729. [Google Scholar] [CrossRef]
  516. Chistiakov, D.A.; Chekhonin, I.V.; Chekhonin, V.P. The EGFR Variant III Mutant as a Target for Immunotherapy of Glioblastoma Multiforme. Eur. J. Pharmacol. 2017, 810, 70–82. [Google Scholar] [CrossRef]
  517. Stichel, D.; Ebrahimi, A.; Reuss, D.; Schrimpf, D.; Ono, T.; Shirahata, M.; Reifenberger, G.; Weller, M.; Hänggi, D.; Wick, W.; et al. Distribution of EGFR Amplification, Combined Chromosome 7 Gain and Chromosome 10 Loss, and TERT Promoter Mutation in Brain Tumors and Their Potential for the Reclassification of IDHwt Astrocytoma to Glioblastoma. Acta Neuropathol. 2018, 136, 793–803. [Google Scholar] [CrossRef] [PubMed]
  518. Halatsch, M.-E.; Gehrke, E.E.; Vougioukas, V.I.; Bötefür, I.C.; A-Borhani, F.; Efferth, T.; Gebhart, E.; Domhof, S.; Schmidt, U.; Buchfelder, M. Inverse Correlation of Epidermal Growth Factor Receptor Messenger RNA Induction and Suppression of Anchorage-Independent Growth by OSI-774, an Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor, in Glioblastoma Multiforme Cell Lines. J. Neurosurg. 2004, 100, 523–533. [Google Scholar] [CrossRef] [PubMed]
  519. Sathornsumetee, S.; Desjardins, A.; Vredenburgh, J.J.; McLendon, R.E.; Marcello, J.; Herndon, J.E.; Mathe, A.; Hamilton, M.; Rich, J.N.; Norfleet, J.A.; et al. Phase II Trial of Bevacizumab and Erlotinib in Patients with Recurrent Malignant Glioma. Neuro Oncol. 2010, 12, 1300–1310. [Google Scholar] [CrossRef]
  520. Lal, A.; Glazer, C.A.; Martinson, H.M.; Friedman, H.S.; Archer, G.E.; Sampson, J.H.; Riggins, G.J. Mutant Epidermal Growth Factor Receptor Up-Regulates Molecular Effectors of Tumor Invasion. Cancer Res. 2002, 62, 3335–3339. [Google Scholar] [PubMed]
  521. Raizer, J.J.; Abrey, L.E.; Lassman, A.B.; Chang, S.M.; Lamborn, K.R.; Kuhn, J.G.; Yung, W.K.A.; Gilbert, M.R.; Aldape, K.A.; Wen, P.Y.; et al. A Phase II Trial of Erlotinib in Patients with Recurrent Malignant Gliomas and Nonprogressive Glioblastoma Multiforme Postradiation Therapy. Neuro Oncol. 2010, 12, 95–103. [Google Scholar] [CrossRef] [PubMed]
  522. Reardon, D.A.; Groves, M.D.; Wen, P.Y.; Nabors, L.; Mikkelsen, T.; Rosenfeld, S.; Raizer, J.; Barriuso, J.; McLendon, R.E.; Suttle, A.B.; et al. A Phase I/II Trial of Pazopanib in Combination with Lapatinib in Adult Patients with Relapsed Malignant Glioma. Clin. Cancer Res. 2013, 19, 900–908. [Google Scholar] [CrossRef] [PubMed]
  523. Struve, N.; Riedel, M.; Schulte, A.; Rieckmann, T.; Grob, T.J.; Gal, A.; Rothkamm, K.; Lamszus, K.; Petersen, C.; Dikomey, E.; et al. EGFRvIII Does Not Affect Radiosensitivity with or without Gefitinib Treatment in Glioblastoma Cells. Oncotarget 2015, 6, 33867–33877. [Google Scholar] [CrossRef]
  524. Karami, A.; Hossienpour, M.; Mohammadi Noori, E.; Rahpyma, M.; Najafi, K.; Kiani, A. Synergistic Effect of Gefitinib and Temozolomide on U87MG Glioblastoma Angiogenesis. Nutr. Cancer 2022, 74, 1299–1307. [Google Scholar] [CrossRef]
  525. Vivanco, I.; Robins, H.I.; Rohle, D.; Campos, C.; Grommes, C.; Nghiemphu, P.L.; Kubek, S.; Oldrini, B.; Chheda, M.G.; Yannuzzi, N.; et al. Differential Sensitivity of Glioma- versus Lung Cancer-Specific EGFR Mutations to EGFR Kinase Inhibitors. Cancer Discov. 2012, 2, 458–471. [Google Scholar] [CrossRef]
  526. Roth, P.; Weller, M. Challenges to Targeting Epidermal Growth Factor Receptor in Glioblastoma: Escape Mechanisms and Combinatorial Treatment Strategies. Neuro Oncol. 2014, 16 (Suppl. S8), viii14-19. [Google Scholar] [CrossRef] [PubMed]
  527. Zahonero, C.; Aguilera, P.; Ramírez-Castillejo, C.; Pajares, M.; Bolós, M.V.; Cantero, D.; Perez-Nuñez, A.; Hernández-Laín, A.; Sánchez-Gómez, P.; Sepúlveda, J.M. Preclinical Test of Dacomitinib, an Irreversible EGFR Inhibitor, Confirms Its Effectiveness for Glioblastoma. Mol. Cancer Ther. 2015, 14, 1548–1558. [Google Scholar] [CrossRef] [PubMed]
  528. Sepúlveda, J.M.; Sánchez-Gómez, P.; Vaz Salgado, M.Á.; Gargini, R.; Balañá, C. Dacomitinib: An Investigational Drug for the Treatment of Glioblastoma. Expert Opin. Investig. Drugs 2018, 27, 823–829. [Google Scholar] [CrossRef] [PubMed]
  529. Chi, A.S.; Cahill, D.P.; Reardon, D.A.; Wen, P.Y.; Mikkelsen, T.; Peereboom, D.M.; Wong, E.T.; Gerstner, E.R.; Dietrich, J.; Plotkin, S.R.; et al. Exploring Predictors of Response to Dacomitinib in EGFR-Amplified Recurrent Glioblastoma. JCO Precis. Oncol. 2020, 4, PO.19.00295. [Google Scholar] [CrossRef] [PubMed]
  530. Zhou, W.; Ercan, D.; Chen, L.; Yun, C.-H.; Li, D.; Capelletti, M.; Cortot, A.B.; Chirieac, L.; Iacob, R.E.; Padera, R.; et al. Novel Mutant-Selective EGFR Kinase Inhibitors against EGFR T790M. Nature 2009, 462, 1070–1074. [Google Scholar] [CrossRef] [PubMed]
  531. Walter, A.O.; Sjin, R.T.T.; Haringsma, H.J.; Ohashi, K.; Sun, J.; Lee, K.; Dubrovskiy, A.; Labenski, M.; Zhu, Z.; Wang, Z.; et al. Discovery of a Mutant-Selective Covalent Inhibitor of EGFR That Overcomes T790M-Mediated Resistance in NSCLC. Cancer Discov. 2013, 3, 1404–1415. [Google Scholar] [CrossRef] [PubMed]
  532. Liu, X.; Chen, X.; Shi, L.; Shan, Q.; Cao, Q.; Yue, C.; Li, H.; Li, S.; Wang, J.; Gao, S.; et al. The Third-Generation EGFR Inhibitor AZD9291 Overcomes Primary Resistance by Continuously Blocking ERK Signaling in Glioblastoma. J. Exp. Clin. Cancer Res. 2019, 38, 219. [Google Scholar] [CrossRef]
  533. Cardona, A.F.; Jaramillo-Velásquez, D.; Ruiz-Patiño, A.; Polo, C.; Jiménez, E.; Hakim, F.; Gómez, D.; Ramón, J.F.; Cifuentes, H.; Mejía, J.A.; et al. Efficacy of Osimertinib plus Bevacizumab in Glioblastoma Patients with Simultaneous EGFR Amplification and EGFRvIII Mutation. J. Neurooncol. 2021, 154, 353–364. [Google Scholar] [CrossRef] [PubMed]
  534. Phillips, A.C.; Boghaert, E.R.; Vaidya, K.S.; Mitten, M.J.; Norvell, S.; Falls, H.D.; DeVries, P.J.; Cheng, D.; Meulbroek, J.A.; Buchanan, F.G.; et al. ABT-414, an Antibody-Drug Conjugate Targeting a Tumor-Selective EGFR Epitope. Mol. Cancer Ther. 2016, 15, 661–669. [Google Scholar] [CrossRef] [PubMed]
  535. Choi, S.W.; Jung, H.A.; Cho, H.-J.; Kim, T.M.; Park, C.-K.; Nam, D.-H.; Lee, S.-H. A Multicenter, Phase II Trial of GC1118, a Novel Anti-EGFR Antibody, for Recurrent Glioblastoma Patients with EGFR Amplification. Cancer Med. 2023, 12, 15788–15796. [Google Scholar] [CrossRef] [PubMed]
  536. Lo, H.-W. EGFR-Targeted Therapy in Malignant Glioma: Novel Aspects and Mechanisms of Drug Resistance. CMP 2010, 3, 37–52. [Google Scholar] [CrossRef]
  537. Guo, G.; Narayan, R.N.; Horton, L.; Patel, T.R.; Habib, A.A. The Role of EGFR-Met Interactions in the Pathogenesis of Glioblastoma and Resistance to Treatment. Curr. Cancer Drug Targets 2017, 17, 297–302. [Google Scholar] [CrossRef]
  538. Lee, A.; Arasaratnam, M.; Chan, D.L.H.; Khasraw, M.; Howell, V.M.; Wheeler, H. Anti-Epidermal Growth Factor Receptor Therapy for Glioblastoma in Adults. Cochrane Database Syst. Rev. 2020, 5, CD013238. [Google Scholar] [CrossRef]
  539. Pan, P.C.; Magge, R.S. Mechanisms of EGFR Resistance in Glioblastoma. Int. J. Mol. Sci. 2020, 21, 8471. [Google Scholar] [CrossRef]
  540. Nazarenko, I.; Hede, S.-M.; He, X.; Hedrén, A.; Thompson, J.; Lindström, M.S.; Nistér, M. PDGF and PDGF Receptors in Glioma. Ups. J. Med. Sci. 2012, 117, 99–112. [Google Scholar] [CrossRef]
  541. Higa, N.; Akahane, T.; Yokoyama, S.; Yonezawa, H.; Uchida, H.; Takajo, T.; Otsuji, R.; Hamada, T.; Matsuo, K.; Kirishima, M.; et al. Prognostic Impact of PDGFRA Gain/Amplification and MGMT Promoter Methylation Status in Patients with IDH Wild-Type Glioblastoma. Neurooncol. Adv. 2022, 4, vdac097. [Google Scholar] [CrossRef]
  542. Vassbotn, F.S.; Ostman, A.; Langeland, N.; Holmsen, H.; Westermark, B.; Heldin, C.H.; Nistér, M. Activated Platelet-Derived Growth Factor Autocrine Pathway Drives the Transformed Phenotype of a Human Glioblastoma Cell Line. J. Cell Physiol. 1994, 158, 381–389. [Google Scholar] [CrossRef]
  543. Westermark, B.; Heldin, C.H.; Nistér, M. Platelet-Derived Growth Factor in Human Glioma. Glia 1995, 15, 257–263. [Google Scholar] [CrossRef] [PubMed]
  544. Reardon, D.A.; Dresemann, G.; Taillibert, S.; Campone, M.; van den Bent, M.; Clement, P.; Blomquist, E.; Gordower, L.; Schultz, H.; Raizer, J.; et al. Multicentre Phase II Studies Evaluating Imatinib plus Hydroxyurea in Patients with Progressive Glioblastoma. Br. J. Cancer 2009, 101, 1995–2004. [Google Scholar] [CrossRef] [PubMed]
  545. Song, K.; Yuan, Y.; Lin, Y.; Wang, Y.-X.; Zhou, J.; Gai, Q.-J.; Zhang, L.; Mao, M.; Yao, X.-X.; Qin, Y.; et al. ERBB3, IGF1R, and TGFBR2 Expression Correlate with PDGFR Expression in Glioblastoma and Participate in PDGFR Inhibitor Resistance of Glioblastoma Cells. Am. J. Cancer Res. 2018, 8, 792–809. [Google Scholar] [PubMed]
  546. Lane, R.; Cilibrasi, C.; Chen, J.; Shah, K.; Messuti, E.; Mazarakis, N.K.; Stebbing, J.; Critchley, G.; Song, E.; Simon, T.; et al. PDGF-R Inhibition Induces Glioblastoma Cell Differentiation via DUSP1/p38MAPK Signalling. Oncogene 2022, 41, 2749–2763. [Google Scholar] [CrossRef] [PubMed]
  547. Ahir, B.K.; Engelhard, H.H.; Lakka, S.S. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol. Neurobiol. 2020, 57, 2461–2478. [Google Scholar] [CrossRef] [PubMed]
  548. Shi, T.; Zhu, J.; Zhang, X.; Mao, X. The Role of Hypoxia and Cancer Stem Cells in Development of Glioblastoma. Cancers 2023, 15, 2613. [Google Scholar] [CrossRef]
  549. Di Cristofano, A.; Pandolfi, P.P. The Multiple Roles of PTEN in Tumor Suppression. Cell 2000, 100, 387–390. [Google Scholar] [CrossRef] [PubMed]
  550. Kessler, T.; Sahm, F.; Blaes, J.; Osswald, M.; Rübmann, P.; Milford, D.; Urban, S.; Jestaedt, L.; Heiland, S.; Bendszus, M.; et al. Glioma Cell VEGFR-2 Confers Resistance to Chemotherapeutic and Antiangiogenic Treatments in PTEN-Deficient Glioblastoma. Oncotarget 2015, 6, 31050–31068. [Google Scholar] [CrossRef]
  551. McBain, C.; Lawrie, T.A.; Rogozińska, E.; Kernohan, A.; Robinson, T.; Jefferies, S. Treatment Options for Progression or Recurrence of Glioblastoma: A Network Meta-analysis. Cochrane Database Syst. Rev. 2021, 2021, CD013579. [Google Scholar] [CrossRef]
  552. Fasano, M.; Pirozzi, M.; Famiglietti, V.; Facchini, S.; Caterino, M.; Caroprese, M.; Barillaro, A.; Di Giovanni, I.; Auriemma, A.; Ileana Sara Fattoruso, S.; et al. Clinical Activity of Regorafenib in Elderly Patients with Recurrent Glioblastoma. Mol. Clin. Oncol. 2023, 18, 9. [Google Scholar] [CrossRef]
  553. Leone, A.; Colamaria, A.; Fochi, N.P.; Sacco, M.; Landriscina, M.; Parbonetti, G.; de Notaris, M.; Coppola, G.; De Santis, E.; Giordano, G.; et al. Recurrent Glioblastoma Treatment: State of the Art and Future Perspectives in the Precision Medicine Era. Biomedicines 2022, 10, 1927. [Google Scholar] [CrossRef]
  554. Gaudino, S.; Marziali, G.; Giordano, C.; Gigli, R.; Varcasia, G.; Magnani, F.; Chiesa, S.; Balducci, M.; Costantini, A.M.; Della Pepa, G.M.; et al. Regorafenib in Glioblastoma Recurrence: How to Deal With MR Imaging Treatments Changes. Front. Radiol. 2022, 1, 790456. [Google Scholar] [CrossRef]
  555. Stupp, R. Drug Development for Glioma: Are We Repeating the Same Mistakes? Lancet Oncol. 2019, 20, 10–12. [Google Scholar] [CrossRef]
  556. Chiesa, S.; Mangraviti, A.; Martini, M.; Cenci, T.; Mazzarella, C.; Gaudino, S.; Bracci, S.; Martino, A.; Della Pepa, G.M.; Offi, M.; et al. Clinical and NGS Predictors of Response to Regorafenib in Recurrent Glioblastoma. Sci. Rep. 2022, 12, 16265. [Google Scholar] [CrossRef] [PubMed]
  557. Lombardi, G.; Caccese, M.; Padovan, M.; Cerretti, G.; Pintacuda, G.; Manara, R.; Di Sarra, F.; Zagonel, V. Regorafenib in Recurrent Glioblastoma Patients: A Large and Monocentric Real-Life Study. Cancers 2021, 13, 4731. [Google Scholar] [CrossRef] [PubMed]
  558. Treiber, H.; von der Brelie, C.; Malinova, V.; Mielke, D.; Rohde, V.; Chapuy, C.I. Regorafenib for Recurrent High-Grade Glioma: A Unicentric Retrospective Analysis of Feasibility, Efficacy, and Toxicity. Neurosurg. Rev. 2022, 45, 3201–3208. [Google Scholar] [CrossRef] [PubMed]
  559. Batchelor, T.T.; Duda, D.G.; di Tomaso, E.; Ancukiewicz, M.; Plotkin, S.R.; Gerstner, E.; Eichler, A.F.; Drappatz, J.; Hochberg, F.H.; Benner, T.; et al. Phase II Study of Cediranib, an Oral Pan-Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitor, in Patients with Recurrent Glioblastoma. J. Clin. Oncol. 2010, 28, 2817–2823. [Google Scholar] [CrossRef]
  560. Duerinck, J.; Du Four, S.; Vandervorst, F.; D’Haene, N.; Le Mercier, M.; Michotte, A.; Van Binst, A.M.; Everaert, H.; Salmon, I.; Bouttens, F.; et al. Randomized Phase II Study of Axitinib versus Physicians Best Alternative Choice of Therapy in Patients with Recurrent Glioblastoma. J. Neurooncol. 2016, 128, 147–155. [Google Scholar] [CrossRef] [PubMed]
  561. Kaplan, A.R.; Gueble, S.E.; Liu, Y.; Oeck, S.; Kim, H.; Yun, Z.; Glazer, P.M. Cediranib Suppresses Homology-Directed DNA Repair through down-Regulation of BRCA1/2 and RAD51. Sci. Transl. Med. 2019, 11, eaav4508. [Google Scholar] [CrossRef]
  562. Lombardi, G.; Pambuku, A.; Bellu, L.; Farina, M.; Della Puppa, A.; Denaro, L.; Zagonel, V. Effectiveness of Antiangiogenic Drugs in Glioblastoma Patients: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Crit. Rev. Oncol. Hematol. 2017, 111, 94–102. [Google Scholar] [CrossRef]
  563. Piao, Y.; Liang, J.; Holmes, L.; Henry, V.; Sulman, E.; de Groot, J.F. Acquired Resistance to Anti-VEGF Therapy in Glioblastoma Is Associated with a Mesenchymal Transition. Clin. Cancer Res. 2013, 19, 4392–4403. [Google Scholar] [CrossRef]
  564. Levin, V.A.; Chan, J.; Datta, M.; Yee, J.L.; Jain, R.K. Effect of Angiotensin System Inhibitors on Survival in Newly Diagnosed Glioma Patients and Recurrent Glioblastoma Patients Receiving Chemotherapy and/or Bevacizumab. J. Neurooncol. 2017, 134, 325–330. [Google Scholar] [CrossRef]
  565. Zhao, H.; Wang, J.; Shao, W.; Wu, C.; Chen, Z.; To, S.T.; Li, W. Recent Advances in the Use of PI3K Inhibitors for Glioblastoma Multiforme: Current Preclinical and Clinical Development. Mol. Cancer 2017, 16, 100. [Google Scholar] [CrossRef]
  566. Boylan, J.; Byers, E.; Kelly, D.F. The Glioblastoma Landscape: Hallmarks of Disease, Therapeutic Resistance, and Treatment Opportunities. Med. Res. Arch. 2023, 11. [Google Scholar] [CrossRef]
  567. Koul, D.; Fu, J.; Shen, R.; LaFortune, T.A.; Wang, S.; Tiao, N.; Kim, Y.-W.; Liu, J.-L.; Ramnarian, D.; Yuan, Y.; et al. Antitumor Activity of NVP-BKM120--a Selective Pan Class I PI3 Kinase Inhibitor Showed Differential Forms of Cell Death Based on P53 Status of Glioma Cells. Clin. Cancer Res. 2012, 18, 184–195. [Google Scholar] [CrossRef]
  568. Wen, P.Y.; Lee, E.Q.; Reardon, D.A.; Ligon, K.L.; Alfred Yung, W.K. Current Clinical Development of PI3K Pathway Inhibitors in Glioblastoma. Neuro Oncol. 2012, 14, 819–829. [Google Scholar] [CrossRef]
  569. Jane, E.P.; Premkumar, D.R.; Morales, A.; Foster, K.A.; Pollack, I.F. Inhibition of Phosphatidylinositol 3-Kinase/AKT Signaling by NVP-BKM120 Promotes ABT-737-Induced Toxicity in a Caspase-Dependent Manner through Mitochondrial Dysfunction and DNA Damage Response in Established and Primary Cultured Glioblastoma Cells. J. Pharmacol. Exp. Ther. 2014, 350, 22–35. [Google Scholar] [CrossRef] [PubMed]
  570. Foster, K.A.; Jane, E.P.; Premkumar, D.R.; Morales, A.; Pollack, I.F. NVP-BKM120 Potentiates Apoptosis in Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand-Resistant Glioma Cell Lines via Upregulation of Noxa and Death Receptor 5. Int. J. Oncol. 2015, 47, 506–516. [Google Scholar] [CrossRef] [PubMed]
  571. Netland, I.A.; Førde, H.E.; Sleire, L.; Leiss, L.; Rahman, M.A.; Skeie, B.S.; Miletic, H.; Enger, P.Ø.; Goplen, D. Treatment with the PI3K Inhibitor Buparlisib (NVP-BKM120) Suppresses the Growth of Established Patient-Derived GBM Xenografts and Prolongs Survival in Nude Rats. J. Neurooncol. 2016, 129, 57–66. [Google Scholar] [CrossRef] [PubMed]
  572. Wen, P.Y.; Wen, P.Y.; Yung, W.K.A.; Mellinghoff, I.K.; Ramkissoon, S.; Alexander, B.; Rinne, M.; Colman, H.; Omuro, A.M.; DeAngelis, L.M.; et al. PHASE II TRIAL OF THE PHOSPHATIDYINOSITOL-3 KINASE (PI3K) INHIBITOR BUPARLISIB (BKM120) IN RECURRENT GLIOBLASTOMA CONDUCTED BY THE IVY FOUNDATION EARLY PHASE CLINICAL TRIALS CONSORTIUM. Neuro Oncol. 2014, 16, iii47. [Google Scholar] [CrossRef]
  573. Kao, G.D.; Jiang, Z.; Fernandes, A.M.; Gupta, A.K.; Maity, A. Inhibition of Phosphatidylinositol-3-OH Kinase/Akt Signaling Impairs DNA Repair in Glioblastoma Cells Following Ionizing Radiation. J. Biol. Chem. 2007, 282, 21206–21212. [Google Scholar] [CrossRef]
  574. Yu, Z.; Xie, G.; Zhou, G.; Cheng, Y.; Zhang, G.; Yao, G.; Chen, Y.; Li, Y.; Zhao, G. NVP-BEZ235, a Novel Dual PI3K-mTOR Inhibitor Displays Anti-Glioma Activity and Reduces Chemoresistance to Temozolomide in Human Glioma Cells. Cancer Lett. 2015, 367, 58–68. [Google Scholar] [CrossRef]
  575. Huang, K.; Zhao, W.; Wang, X.; Qiu, Y.; Liu, Z.; Chen, R.; Liu, W.; Liu, B. Akt Inhibition Enhanced the Growth Inhibition Effects of Low-Dose Heavy-Ion Radiation via the PI3K/Akt/P53 Signaling Pathway in C6 Glioblastoma Cells. Front. Oncol. 2021, 11, 649176. [Google Scholar] [CrossRef]
  576. Chandrika, G.; Natesh, K.; Ranade, D.; Chugh, A.; Shastry, P. Suppression of the Invasive Potential of Glioblastoma Cells by mTOR Inhibitors Involves Modulation of NFκB and PKC-α Signaling. Sci. Rep. 2016, 6, 22455. [Google Scholar] [CrossRef]
  577. Markman, B.; Dienstmann, R.; Tabernero, J. Targeting the PI3K/Akt/mTOR Pathway--beyond Rapalogs. Oncotarget 2010, 1, 530–543. [Google Scholar] [CrossRef] [PubMed]
  578. Arcella, A.; Biagioni, F.; Antonietta Oliva, M.; Bucci, D.; Frati, A.; Esposito, V.; Cantore, G.; Giangaspero, F.; Fornai, F. Rapamycin Inhibits the Growth of Glioblastoma. Brain Res. 2013, 1495, 37–51. [Google Scholar] [CrossRef] [PubMed]
  579. Cloughesy, T.F.; Yoshimoto, K.; Nghiemphu, P.; Brown, K.; Dang, J.; Zhu, S.; Hsueh, T.; Chen, Y.; Wang, W.; Youngkin, D.; et al. Antitumor Activity of Rapamycin in a Phase I Trial for Patients with Recurrent PTEN-Deficient Glioblastoma. PLoS Med. 2008, 5, e8. [Google Scholar] [CrossRef] [PubMed]
  580. Gini, B.; Zanca, C.; Guo, D.; Matsutani, T.; Masui, K.; Ikegami, S.; Yang, H.; Nathanson, D.; Villa, G.R.; Shackelford, D.; et al. The mTOR Kinase Inhibitors, CC214-1 and CC214-2, Preferentially Block the Growth of EGFRvIII-Activated Glioblastomas. Clin. Cancer Res. 2013, 19, 5722–5732. [Google Scholar] [CrossRef] [PubMed]
  581. Fan, Q.; Aksoy, O.; Wong, R.A.; Ilkhanizadeh, S.; Novotny, C.J.; Gustafson, W.C.; Truong, A.Y.-Q.; Cayanan, G.; Simonds, E.F.; Haas-Kogan, D.; et al. A Kinase Inhibitor Targeted to mTORC1 Drives Regression in Glioblastoma. Cancer Cell 2017, 31, 424–435. [Google Scholar] [CrossRef]
  582. Jhanwar-Uniyal, M.; Gellerson, O.; Bree, J.; Das, M.; Kleinman, G.; Gandhi, C.D. Defining the Role of mTOR Pathway in the Regulation of Stem Cells of Glioblastoma. Adv. Biol. Regul. 2023, 88, 100946. [Google Scholar] [CrossRef]
  583. Vargas-Toscano, A.; Nickel, A.-C.; Li, G.; Kamp, M.A.; Muhammad, S.; Leprivier, G.; Fritsche, E.; Barker, R.A.; Sabel, M.; Steiger, H.-J.; et al. Rapalink-1 Targets Glioblastoma Stem Cells and Acts Synergistically with Tumor Treating Fields to Reduce Resistance against Temozolomide. Cancers 2020, 12, 3859. [Google Scholar] [CrossRef]
  584. Kahn, J.; Hayman, T.J.; Jamal, M.; Rath, B.H.; Kramp, T.; Camphausen, K.; Tofilon, P.J. The mTORC1/mTORC2 Inhibitor AZD2014 Enhances the Radiosensitivity of Glioblastoma Stem-like Cells. Neuro Oncol. 2014, 16, 29–37. [Google Scholar] [CrossRef]
  585. Lapointe, S.; Mason, W.; MacNeil, M.; Harlos, C.; Tsang, R.; Sederias, J.; Luchman, H.A.; Weiss, S.; Rossiter, J.P.; Tu, D.; et al. A Phase I Study of Vistusertib (Dual mTORC1/2 Inhibitor) in Patients with Previously Treated Glioblastoma Multiforme: A CCTG Study. Investig. New Drugs 2020, 38, 1137–1144. [Google Scholar] [CrossRef]
  586. Albert, L.; Karsy, M.; Murali, R.; Jhanwar-Uniyal, M. Inhibition of mTOR Activates the MAPK Pathway in Glioblastoma Multiforme. Cancer Genom. Proteom. 2009, 6, 255–261. [Google Scholar]
  587. Aldaz, P.; Arozarena, I. Tyrosine Kinase Inhibitors in Adult Glioblastoma: An (Un)Closed Chapter? Cancers 2021, 13, 5799. [Google Scholar] [CrossRef] [PubMed]
  588. Kim, G.; Ko, Y.T. Small Molecule Tyrosine Kinase Inhibitors in Glioblastoma. Arch. Pharm. Res. 2020, 43, 385–394. [Google Scholar] [CrossRef] [PubMed]
  589. Brar, H.K.; Jose, J.; Wu, Z.; Sharma, M. Tyrosine Kinase Inhibitors for Glioblastoma Multiforme: Challenges and Opportunities for Drug Delivery. Pharmaceutics 2022, 15, 59. [Google Scholar] [CrossRef] [PubMed]
  590. Schreck, K.C.; Grossman, S.A.; Pratilas, C.A. BRAF Mutations and the Utility of RAF and MEK Inhibitors in Primary Brain Tumors. Cancers 2019, 11, 1262. [Google Scholar] [CrossRef] [PubMed]
  591. Kowalewski, A.; Durślewicz, J.; Zdrenka, M.; Grzanka, D.; Szylberg, Ł. Clinical Relevance of BRAF V600E Mutation Status in Brain Tumors with a Focus on a Novel Management Algorithm. Target. Oncol. 2020, 15, 531–540. [Google Scholar] [CrossRef]
  592. Andrews, L.J.; Thornton, Z.A.; Saincher, S.S.; Yao, I.Y.; Dawson, S.; McGuinness, L.A.; Jones, H.E.; Jefferies, S.; Short, S.C.; Cheng, H.-Y.; et al. Prevalence of BRAFV600 in Glioma and Use of BRAF Inhibitors in Patients with BRAFV600 Mutation-Positive Glioma: Systematic Review. Neuro Oncol. 2022, 24, 528–540. [Google Scholar] [CrossRef] [PubMed]
  593. McNulty, S.N.; Schwetye, K.E.; Ferguson, C.; Storer, C.E.; Ansstas, G.; Kim, A.H.; Gutmann, D.H.; Rubin, J.B.; Head, R.D.; Dahiya, S. BRAF Mutations May Identify a Clinically Distinct Subset of Glioblastoma. Sci. Rep. 2021, 11, 19999. [Google Scholar] [CrossRef] [PubMed]
  594. Di Nunno, V.; Gatto, L.; Tosoni, A.; Bartolini, S.; Franceschi, E. Implications of BRAF V600E Mutation in Gliomas: Molecular Considerations, Prognostic Value and Treatment Evolution. Front. Oncol. 2022, 12, 1067252. [Google Scholar] [CrossRef] [PubMed]
  595. Beba Abadal, K.; Walsh, M.A.; Yachnis, A.T.; Tran, D.D.; Ghiaseddin, A.P. Eleven Month Progression-Free Survival on Vemurafenib Monotherapy in a Patient With Recurrent and Metastatic BRAF V600E-Mutated Glioblastoma WHO Grade 4. JCO Precis. Oncol. 2017, 1, 1–5. [Google Scholar] [CrossRef] [PubMed]
  596. Ceccon, G.; Werner, J.-M.; Dunkl, V.; Tscherpel, C.; Stoffels, G.; Brunn, A.; Deckert, M.; Fink, G.R.; Galldiks, N. Dabrafenib Treatment in a Patient with an Epithelioid Glioblastoma and BRAF V600E Mutation. Int. J. Mol. Sci. 2018, 19, 1090. [Google Scholar] [CrossRef]
  597. Woo, P.Y.M.; Lam, T.-C.; Pu, J.K.S.; Li, L.-F.; Leung, R.C.Y.; Ho, J.M.K.; Zhung, J.T.F.; Wong, B.; Chan, T.S.K.; Loong, H.H.F.; et al. Regression of BRAF V600E Mutant Adult Glioblastoma after Primary Combined BRAF-MEK Inhibitor Targeted Therapy: A Report of Two Cases. Oncotarget 2019, 10, 3818–3826. [Google Scholar] [CrossRef] [PubMed]
  598. Kushnirsky, M.; Feun, L.G.; Gultekin, S.H.; de la Fuente, M.I. Prolonged Complete Response With Combined Dabrafenib and Trametinib After BRAF Inhibitor Failure in BRAF-Mutant Glioblastoma. JCO Precis. Oncol. 2020, 4, PO.19.00272. [Google Scholar] [CrossRef]
  599. Kaley, T.; Touat, M.; Subbiah, V.; Hollebecque, A.; Rodon, J.; Lockhart, A.C.; Keedy, V.; Bielle, F.; Hofheinz, R.-D.; Joly, F.; et al. BRAF Inhibition in BRAFV600-Mutant Gliomas: Results From the VE-BASKET Study. J. Clin. Oncol. 2018, 36, 3477–3484. [Google Scholar] [CrossRef]
  600. Khan, S.; Martinez-Ledesma, E.; Dong, J.; Mahalingam, R.; Park, S.Y.; Piao, Y.; Koul, D.; Balasubramaniyan, V.; de Groot, J.F.; Yung, W.K.A. Neuronal Differentiation Drives the Antitumor Activity of Mitogen-Activated Protein Kinase Kinase (MEK) Inhibition in Glioblastoma. Neurooncol. Adv. 2023, 5, vdad132. [Google Scholar] [CrossRef]
  601. Yao, T.-W.; Zhang, J.; Prados, M.; Weiss, W.A.; James, C.D.; Nicolaides, T. EGFR Blockade Prevents Glioma Escape from BRAFV600E Targeted Therapy. Oncotarget 2015, 6, 21993–22005. [Google Scholar] [CrossRef]
  602. Bouffet, E.; Hansford, J.R.; Garrè, M.L.; Hara, J.; Plant-Fox, A.; Aerts, I.; Locatelli, F.; van der Lugt, J.; Papusha, L.; Sahm, F.; et al. Dabrafenib plus Trametinib in Pediatric Glioma with BRAF V600 Mutations. N. Engl. J. Med. 2023, 389, 1108–1120. [Google Scholar] [CrossRef]
  603. Venkatesh, A.; Joshi, A.; Allinson, K.; Das, T.; Santarius, T.; Jefferies, S.J.; Harris, F.P.; Jena, R.; Doherty, G.J. Response to BRAF and MEK1/2 Inhibition in a Young Adult with BRAF V600E Mutant Epithelioid Glioblastoma Multiforme: A Case Report and Literature Review. Curr. Probl. Cancer 2021, 45, 100701. [Google Scholar] [CrossRef] [PubMed]
  604. Murnyák, B.; Kouhsari, M.C.; Hershkovitch, R.; Kálmán, B.; Marko-Varga, G.; Klekner, Á.; Hortobágyi, T. PARP1 Expression and Its Correlation with Survival Is Tumour Molecular Subtype Dependent in Glioblastoma. Oncotarget 2017, 8, 46348–46362. [Google Scholar] [CrossRef] [PubMed]
  605. Tentori, L.; Ricci-Vitiani, L.; Muzi, A.; Ciccarone, F.; Pelacchi, F.; Calabrese, R.; Runci, D.; Pallini, R.; Caiafa, P.; Graziani, G. Pharmacological Inhibition of Poly(ADP-Ribose) Polymerase-1 Modulates Resistance of Human Glioblastoma Stem Cells to Temozolomide. BMC Cancer 2014, 14, 151. [Google Scholar] [CrossRef] [PubMed]
  606. Gupta, S.K.; Smith, E.J.; Mladek, A.C.; Tian, S.; Decker, P.A.; Kizilbash, S.H.; Kitange, G.J.; Sarkaria, J.N. PARP Inhibitors for Sensitization of Alkylation Chemotherapy in Glioblastoma: Impact of Blood-Brain Barrier and Molecular Heterogeneity. Front. Oncol. 2018, 8, 670. [Google Scholar] [CrossRef]
  607. Higuchi, F.; Nagashima, H.; Ning, J.; Koerner, M.V.A.; Wakimoto, H.; Cahill, D.P. Restoration of Temozolomide Sensitivity by PARP Inhibitors in Mismatch Repair Deficient Glioblastoma Is Independent of Base Excision Repair. Clin. Cancer Res. 2020, 26, 1690–1699. [Google Scholar] [CrossRef]
  608. Lesueur, P.; Chevalier, F.; El-Habr, E.A.; Junier, M.-P.; Chneiweiss, H.; Castera, L.; Müller, E.; Stefan, D.; Saintigny, Y. Radiosensitization Effect of Talazoparib, a Parp Inhibitor, on Glioblastoma Stem Cells Exposed to Low and High Linear Energy Transfer Radiation. Sci. Rep. 2018, 8, 3664. [Google Scholar] [CrossRef]
  609. Yuan, A.L.; Ricks, C.B.; Bohm, A.K.; Lun, X.; Maxwell, L.; Safdar, S.; Bukhari, S.; Gerber, A.; Sayeed, W.; Bering, E.A.; et al. ABT-888 Restores Sensitivity in Temozolomide Resistant Glioma Cells and Xenografts. PLoS ONE 2018, 13, e0202860. [Google Scholar] [CrossRef]
  610. Gupta, S.K.; Kizilbash, S.H.; Carlson, B.L.; Mladek, A.C.; Boakye-Agyeman, F.; Bakken, K.K.; Pokorny, J.L.; Schroeder, M.A.; Decker, P.A.; Cen, L.; et al. Delineation of MGMT Hypermethylation as a Biomarker for Veliparib-Mediated Temozolomide-Sensitizing Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108, djv369. [Google Scholar] [CrossRef]
  611. Jue, T.R.; Nozue, K.; Lester, A.J.; Joshi, S.; Schroder, L.B.W.; Whittaker, S.P.; Nixdorf, S.; Rapkins, R.W.; Khasraw, M.; McDonald, K.L. Veliparib in Combination with Radiotherapy for the Treatment of MGMT Unmethylated Glioblastoma. J. Transl. Med. 2017, 15, 61. [Google Scholar] [CrossRef]
  612. Lin, F.; de Gooijer, M.C.; Roig, E.M.; Buil, L.C.M.; Christner, S.M.; Beumer, J.H.; Würdinger, T.; Beijnen, J.H.; van Tellingen, O. ABCB1, ABCG2, and PTEN Determine the Response of Glioblastoma to Temozolomide and ABT-888 Therapy. Clin. Cancer Res. 2014, 20, 2703–2713. [Google Scholar] [CrossRef]
  613. Zampieri, L.X.; Sboarina, M.; Cacace, A.; Grasso, D.; Thabault, L.; Hamelin, L.; Vazeille, T.; Dumon, E.; Rossignol, R.; Frédérick, R.; et al. Olaparib Is a Mitochondrial Complex I Inhibitor That Kills Temozolomide-Resistant Human Glioblastoma Cells. Int. J. Mol. Sci. 2021, 22, 11938. [Google Scholar] [CrossRef] [PubMed]
  614. Hwang, K.; Lee, J.-H.; Kim, S.H.; Go, K.-O.; Ji, S.Y.; Han, J.H.; Kim, C.-Y. The Combination PARP Inhibitor Olaparib With Temozolomide in an Experimental Glioblastoma Model. In Vivo 2021, 35, 2015–2023. [Google Scholar] [CrossRef]
  615. Halford, S.E.R.; Cruickshank, G.; Dunn, L.; Erridge, S.; Godfrey, L.; Herbert, C.; Jefferies, S.; Lopez, J.S.; McBain, C.; Pittman, M.; et al. Results of the OPARATIC Trial: A Phase I Dose Escalation Study of Olaparib in Combination with Temozolomide (TMZ) in Patients with Relapsed Glioblastoma (GBM). JCO 2017, 35, 2022. [Google Scholar] [CrossRef]
  616. Hanna, C.; Kurian, K.M.; Williams, K.; Watts, C.; Jackson, A.; Carruthers, R.; Strathdee, K.; Cruickshank, G.; Dunn, L.; Erridge, S.; et al. Pharmacokinetics, Safety, and Tolerability of Olaparib and Temozolomide for Recurrent Glioblastoma: Results of the Phase I OPARATIC Trial. Neuro Oncol. 2020, 22, 1840–1850. [Google Scholar] [CrossRef]
  617. Wu, S.; Gao, F.; Zheng, S.; Zhang, C.; Martinez-Ledesma, E.; Ezhilarasan, R.; Ding, J.; Li, X.; Feng, N.; Multani, A.; et al. EGFR Amplification Induces Increased DNA Damage Response and Renders Selective Sensitivity to Talazoparib (PARP Inhibitor) in Glioblastoma. Clin. Cancer Res. 2020, 26, 1395–1407. [Google Scholar] [CrossRef]
  618. Xiong, Y.; Guo, Y.; Liu, Y.; Wang, H.; Gong, W.; Liu, Y.; Wang, X.; Gao, Y.; Yu, F.; Su, D.; et al. Pamiparib Is a Potent and Selective PARP Inhibitor with Unique Potential for the Treatment of Brain Tumor. Neoplasia 2020, 22, 431–440. [Google Scholar] [CrossRef]
  619. Piotrowski, A.; Puduvalli, V.; Wen, P.; Campian, J.; Colman, H.; Pearlman, M.; Butowski, N.; Battiste, J.; Glass, J.; Cloughesy, T.; et al. ACTR-39. PAMIPARIB IN COMBINATION WITH RADIATION THERAPY (RT) AND/OR TEMOZOLOMIDE (TMZ) IN PATIENTS WITH NEWLY DIAGNOSED OR RECURRENT/REFRACTORY (R/R) GLIOBLASTOMA (GBM); PHASE 1B/2 STUDY UPDATE. Neuro Oncol. 2019, 21, vi21–vi22. [Google Scholar] [CrossRef]
  620. Bisht, P.; Kumar, V.U.; Pandey, R.; Velayutham, R.; Kumar, N. Role of PARP Inhibitors in Glioblastoma and Perceiving Challenges as Well as Strategies for Successful Clinical Development. Front. Pharmacol. 2022, 13, 939570. [Google Scholar] [CrossRef]
  621. Yanae, M.; Tsubaki, M.; Satou, T.; Itoh, T.; Imano, M.; Yamazoe, Y.; Nishida, S. Statin-Induced Apoptosis via the Suppression of ERK1/2 and Akt Activation by Inhibition of the Geranylgeranyl-Pyrophosphate Biosynthesis in Glioblastoma. J. Exp. Clin. Cancer Res. 2011, 30, 74. [Google Scholar] [CrossRef]
  622. Jiang, P.; Mukthavaram, R.; Chao, Y.; Bharati, I.S.; Fogal, V.; Pastorino, S.; Cong, X.; Nomura, N.; Gallagher, M.; Abbasi, T.; et al. Novel Anti-Glioblastoma Agents and Therapeutic Combinations Identified from a Collection of FDA Approved Drugs. J. Transl. Med. 2014, 12, 13. [Google Scholar] [CrossRef]
  623. Samiei, E.; Seyfoori, A.; Toyota, B.; Ghavami, S.; Akbari, M. Investigating Programmed Cell Death and Tumor Invasion in a Three-Dimensional (3D) Microfluidic Model of Glioblastoma. Int. J. Mol. Sci. 2020, 21, 3162. [Google Scholar] [CrossRef]
  624. Afshari, A.R.; Mollazadeh, H.; Henney, N.C.; Jamialahmad, T.; Sahebkar, A. Effects of Statins on Brain Tumors: A Review. Semin. Cancer Biol. 2021, 73, 116–133. [Google Scholar] [CrossRef]
  625. Rendon, L.F.; Tewarie, I.A.; Cote, D.J.; Gabriel, A.; Smith, T.R.; Broekman, M.L.D.; Mekary, R.A. Statins and Gliomas: A Systematic Review of the Preclinical Studies and Meta-Analysis of the Clinical Literature. Drugs 2022, 82, 293–310. [Google Scholar] [CrossRef]
  626. Xiao, A.; Brenneman, B.; Floyd, D.; Comeau, L.; Spurio, K.; Olmez, I.; Lee, J.; Nakano, I.; Godlewski, J.; Bronisz, A.; et al. Statins Affect Human Glioblastoma and Other Cancers through TGF-β Inhibition. Oncotarget 2019, 10, 1716–1728. [Google Scholar] [CrossRef]
  627. Zhu, Z.; Zhang, P.; Li, N.; Kiang, K.M.Y.; Cheng, S.Y.; Wong, V.K.-W.; Leung, G.K.-K. Lovastatin Enhances Cytotoxicity of Temozolomide via Impairing Autophagic Flux in Glioblastoma Cells. Biomed. Res. Int. 2019, 2019, 2710693. [Google Scholar] [CrossRef]
  628. Shojaei, S.; Koleini, N.; Samiei, E.; Aghaei, M.; Cole, L.K.; Alizadeh, J.; Islam, M.I.; Vosoughi, A.-R.; Albokashy, M.; Butterfield, Y.; et al. Simvastatin Increases Temozolomide-Induced Cell Death by Targeting the Fusion of Autophagosomes and Lysosomes. FEBS J. 2020, 287, 1005–1034. [Google Scholar] [CrossRef]
  629. Dastghaib, S.; Shojaei, S.; Mostafavi-Pour, Z.; Sharma, P.; Patterson, J.B.; Samali, A.; Mokarram, P.; Ghavami, S. Simvastatin Induces Unfolded Protein Response and Enhances Temozolomide-Induced Cell Death in Glioblastoma Cells. Cells 2020, 9, 2339. [Google Scholar] [CrossRef]
  630. Peng, P.; Wei, W.; Long, C.; Li, J. Atorvastatin Augments Temozolomide’s Efficacy in Glioblastoma via Prenylation-Dependent Inhibition of Ras Signaling. Biochem. Biophys. Res. Commun. 2017, 489, 293–298. [Google Scholar] [CrossRef]
  631. McCarty, J.H. Glioblastoma Resistance to Anti-VEGF Therapy: Has the Challenge Been MET? Clin. Cancer Res. 2013, 19, 1631–1633. [Google Scholar] [CrossRef] [PubMed]
  632. Carvalho, B.; Lopes, J.M.; Silva, R.; Peixoto, J.; Leitão, D.; Soares, P.; Fernandes, A.C.; Linhares, P.; Vaz, R.; Lima, J. The Role of C-Met and VEGFR2 in Glioblastoma Resistance to Bevacizumab. Sci. Rep. 2021, 11, 6067. [Google Scholar] [CrossRef] [PubMed]
  633. Martinez Garcia, M.; Gil, M.J.; Pineda, E.; Martin Soberón, M.C.; Mesia Barroso, C.; Foro, P.; Capellades, J.; Sarmiento, B.; Bruna, J.; Verger, E.; et al. 401PD—GEINO 1402: A Phase Ib Dose-Escalation Study Followed by an Extension Phase to Evaluate Safety and Efficacy of Crizotinib in Combination with Temozolomide (TMZ) and Radiotherapy (RT) in Patients with Newly Diagnosed Glioblastoma (GB). Ann. Oncol. 2019, 30, v147. [Google Scholar] [CrossRef]
  634. Zamykal, M.; Martens, T.; Matschke, J.; Günther, H.S.; Kathagen, A.; Schulte, A.; Peters, R.; Westphal, M.; Lamszus, K. Inhibition of Intracerebral Glioblastoma Growth by Targeting the Insulin-like Growth Factor 1 Receptor Involves Different Context-Dependent Mechanisms. Neuro Oncol. 2015, 17, 1076–1085. [Google Scholar] [CrossRef]
  635. Maris, C.; D’Haene, N.; Trépant, A.-L.; Le Mercier, M.; Sauvage, S.; Allard, J.; Rorive, S.; Demetter, P.; Decaestecker, C.; Salmon, I. IGF-IR: A New Prognostic Biomarker for Human Glioblastoma. Br. J. Cancer 2015, 113, 729–737. [Google Scholar] [CrossRef]
  636. Tirrò, E.; Massimino, M.; Romano, C.; Martorana, F.; Pennisi, M.S.; Stella, S.; Pavone, G.; Di Gregorio, S.; Puma, A.; Tomarchio, C.; et al. Prognostic and Therapeutic Roles of the Insulin Growth Factor System in Glioblastoma. Front. Oncol. 2021, 10, 612385. [Google Scholar] [CrossRef]
  637. Andrews, D.W.; Garcia, S.; Judy, K.D.; Harshyne, L.A.; Govindarajan, S.; Kenyon, L.; Talekar, K.; Flanders, A.; Atsina, K.-B.; Kim, L.; et al. Abstract CT038: Results of a Phase Ib Trial of an Autologous Cell Vaccine for Newly Diagnosed Glioblastoma. Cancer Res. 2019, 79, CT038. [Google Scholar] [CrossRef]
  638. Dhandapani, K.M.; Mahesh, V.B.; Brann, D.W. Curcumin Suppresses Growth and Chemoresistance of Human Glioblastoma Cells via AP-1 and NFkappaB Transcription Factors. J. Neurochem. 2007, 102, 522–538. [Google Scholar] [CrossRef]
  639. Arabzadeh, A.; Mortezazadeh, T.; Aryafar, T.; Gharepapagh, E.; Majdaeen, M.; Farhood, B. Therapeutic Potentials of Resveratrol in Combination with Radiotherapy and Chemotherapy during Glioblastoma Treatment: A Mechanistic Review. Cancer Cell Int. 2021, 21, 391. [Google Scholar] [CrossRef]
  640. Wong, S.C.; Kamarudin, M.N.A.; Naidu, R. Anticancer Mechanism of Curcumin on Human Glioblastoma. Nutrients 2021, 13, 950. [Google Scholar] [CrossRef] [PubMed]
  641. Zhai, K.; Mazurakova, A.; Koklesova, L.; Kubatka, P.; Büsselberg, D. Flavonoids Synergistically Enhance the Anti-Glioblastoma Effects of Chemotherapeutic Drugs. Biomolecules 2021, 11, 1841. [Google Scholar] [CrossRef] [PubMed]
  642. Zhai, K.; Siddiqui, M.; Abdellatif, B.; Liskova, A.; Kubatka, P.; Büsselberg, D. Natural Compounds in Glioblastoma Therapy: Preclinical Insights, Mechanistic Pathways, and Outlook. Cancers 2021, 13, 2317. [Google Scholar] [CrossRef] [PubMed]
  643. Karkon-Shayan, S.; Aliashrafzadeh, H.; Dianat-Moghadam, H.; Rastegar-Pouyani, N.; Majidi, M.; Zarei, M.; moradi-vastegani, S.; Bahramvand, Y.; Babaniamansour, S.; Jafarzadeh, E. Resveratrol as an Antitumor Agent for Glioblastoma Multiforme: Targeting Resistance and Promoting Apoptotic Cell Deaths. Acta Histochem. 2023, 125, 152058. [Google Scholar] [CrossRef]
  644. Ibrahim, R.S.; Ibrahim, S.S.; El-Naas, A.; Koklesová, L.; Kubatka, P.; Büsselberg, D. Could Metformin and Resveratrol Support Glioblastoma Treatment? A Mechanistic View at the Cellular Level. Cancers 2023, 15, 3368. [Google Scholar] [CrossRef]
  645. Sang, D.-P.; Li, R.-J.; Lan, Q. Quercetin Sensitizes Human Glioblastoma Cells to Temozolomide in Vitro via Inhibition of Hsp27. Acta Pharmacol. Sin. 2014, 35, 832–838. [Google Scholar] [CrossRef] [PubMed]
  646. Beylerli, O.; Beilerli, A.; Shumadalova, A.; Wang, X.; Yang, M.; Sun, H.; Teng, L. Therapeutic Effect of Natural Polyphenols against Glioblastoma. Front. Cell Dev. Biol. 2022, 10, 1036809. [Google Scholar] [CrossRef] [PubMed]
  647. Zanotto-Filho, A.; Braganhol, E.; Klafke, K.; Figueiró, F.; Terra, S.R.; Paludo, F.J.; Morrone, M.; Bristot, I.J.; Battastini, A.M.; Forcelini, C.M.; et al. Autophagy Inhibition Improves the Efficacy of Curcumin/Temozolomide Combination Therapy in Glioblastomas. Cancer Lett. 2015, 358, 220–231. [Google Scholar] [CrossRef] [PubMed]
  648. Estrela, J.M.; Mena, S.; Obrador, E.; Benlloch, M.; Castellano, G.; Salvador, R.; Dellinger, R.W. Polyphenolic Phytochemicals in Cancer Prevention and Therapy: Bioavailability versus Bioefficacy. J. Med. Chem. 2017, 60, 9413–9436. [Google Scholar] [CrossRef] [PubMed]
  649. Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence That Curcumin Suppresses the Growth of Malignant Gliomas in Vitro and in Vivo through Induction of Autophagy: Role of Akt and Extracellular Signal-Regulated Kinase Signaling Pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef]
  650. Piwowarczyk, L.; Mlynarczyk, D.T.; Krajka-Kuźniak, V.; Majchrzak-Celińska, A.; Budzianowska, A.; Tomczak, S.; Budzianowski, J.; Woźniak-Braszak, A.; Pietrzyk, R.; Baranowski, M.; et al. Natural Compounds in Liposomal Nanoformulations of Potential Clinical Application in Glioblastoma. Cancers 2022, 14, 6222. [Google Scholar] [CrossRef]
  651. Liang, Q.; Zhuo, Y.; Wu, X.; Zheng, S.; Zhuang, J.; Wang, K.; Chen, S. Curcumin Combining Temozolomide Formed Localized Nanogel for Inhibition of Postsurgical Chemoresistant Glioblastoma. Nanomedicine 2023, 18, 907–921. [Google Scholar] [CrossRef]
  652. Ali, M.; Benfante, V.; Di Raimondo, D.; Salvaggio, G.; Tuttolomondo, A.; Comelli, A. Recent Developments in Nanoparticle Formulations for Resveratrol Encapsulation as an Anticancer Agent. Pharmaceuticals 2024, 17, 126. [Google Scholar] [CrossRef]
  653. Andersen, R.S.; Anand, A.; Harwood, D.S.L.; Kristensen, B.W. Tumor-Associated Microglia and Macrophages in the Glioblastoma Microenvironment and Their Implications for Therapy. Cancers 2021, 13, 4255. [Google Scholar] [CrossRef]
  654. Wang, G.; Zhong, K.; Wang, Z.; Zhang, Z.; Tang, X.; Tong, A.; Zhou, L. Tumor-Associated Microglia and Macrophages in Glioblastoma: From Basic Insights to Therapeutic Opportunities. Front. Immunol. 2022, 13, 964898. [Google Scholar] [CrossRef]
  655. Fanelli, G.N.; Grassini, D.; Ortenzi, V.; Pasqualetti, F.; Montemurro, N.; Perrini, P.; Naccarato, A.G.; Scatena, C. Decipher the Glioblastoma Microenvironment: The First Milestone for New Groundbreaking Therapeutic Strategies. Genes 2021, 12, 445. [Google Scholar] [CrossRef]
  656. Choi, B.D.; Maus, M.V.; June, C.H.; Sampson, J.H. Immunotherapy for Glioblastoma: Adoptive T-Cell Strategies. Clin. Cancer Res. 2019, 25, 2042–2048. [Google Scholar] [CrossRef]
  657. Heiland, D.H.; Haaker, G.; Delev, D.; Mercas, B.; Masalha, W.; Heynckes, S.; Gäbelein, A.; Pfeifer, D.; Carro, M.S.; Weyerbrock, A.; et al. Comprehensive Analysis of PD-L1 Expression in Glioblastoma Multiforme. Oncotarget 2017, 8, 42214–42225. [Google Scholar] [CrossRef]
  658. Shu, C.; Li, Q. Current Advances in PD-1/PD-L1 Axis-Related Tumour-Infiltrating Immune Cells and Therapeutic Regimens in Glioblastoma. Crit. Rev. Oncol. /Hematol. 2020, 151, 102965. [Google Scholar] [CrossRef]
  659. Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef]
  660. De Felice, F.; Pranno, N.; Marampon, F.; Musio, D.; Salducci, M.; Polimeni, A.; Tombolini, V. Immune Check-Point in Glioblastoma Multiforme. Crit. Rev. Oncol. Hematol. 2019, 138, 60–69. [Google Scholar] [CrossRef]
  661. Zeng, J.; See, A.P.; Phallen, J.; Jackson, C.M.; Belcaid, Z.; Ruzevick, J.; Durham, N.; Meyer, C.; Harris, T.J.; Albesiano, E.; et al. Anti-PD-1 Blockade and Stereotactic Radiation Produce Long-Term Survival in Mice with Intracranial Gliomas. Int. J. Radiat. Oncol. Biol. Phys. 2013, 86, 343–349. [Google Scholar] [CrossRef]
  662. Nayak, L.; Molinaro, A.M.; Peters, K.; Clarke, J.L.; Jordan, J.T.; de Groot, J.; Nghiemphu, L.; Kaley, T.; Colman, H.; McCluskey, C.; et al. Randomized Phase II and Biomarker Study of Pembrolizumab plus Bevacizumab versus Pembrolizumab Alone for Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2021, 27, 1048–1057. [Google Scholar] [CrossRef]
  663. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O.; et al. Effect of Nivolumab vs Bevacizumab in Patients With Recurrent Glioblastoma: The CheckMate 143 Phase 3 Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1003–1010. [Google Scholar] [CrossRef]
  664. Lim, M.; Weller, M.; Idbaih, A.; Steinbach, J.; Finocchiaro, G.; Raval, R.R.; Ansstas, G.; Baehring, J.; Taylor, J.W.; Honnorat, J.; et al. Phase III Trial of Chemoradiotherapy with Temozolomide plus Nivolumab or Placebo for Newly Diagnosed Glioblastoma with Methylated MGMT Promoter. Neuro Oncol. 2022, 24, 1935–1949. [Google Scholar] [CrossRef]
  665. Omuro, A.; Brandes, A.A.; Carpentier, A.F.; Idbaih, A.; Reardon, D.A.; Cloughesy, T.; Sumrall, A.; Baehring, J.; van den Bent, M.; Bähr, O.; et al. Radiotherapy Combined with Nivolumab or Temozolomide for Newly Diagnosed Glioblastoma with Unmethylated MGMT Promoter: An International Randomized Phase III Trial. Neuro Oncol. 2023, 25, 123–134. [Google Scholar] [CrossRef]
  666. Lepski, G.; Bergami-Santos, P.C.; Pinho, M.P.; Chauca-Torres, N.E.; Evangelista, G.C.M.; Teixeira, S.F.; Flatow, E.; de Oliveira, J.V.; Fogolin, C.; Peres, N.; et al. Adjuvant Vaccination with Allogenic Dendritic Cells Significantly Prolongs Overall Survival in High-Grade Gliomas: Results of a Phase II Trial. Cancers 2023, 15, 1239. [Google Scholar] [CrossRef] [PubMed]
  667. Buchroithner, J.; Erhart, F.; Pichler, J.; Widhalm, G.; Preusser, M.; Stockhammer, G.; Nowosielski, M.; Iglseder, S.; Freyschlag, C.F.; Oberndorfer, S.; et al. Audencel Immunotherapy Based on Dendritic Cells Has No Effect on Overall and Progression-Free Survival in Newly Diagnosed Glioblastoma: A Phase II Randomized Trial. Cancers 2018, 10, 372. [Google Scholar] [CrossRef]
  668. Inogés, S.; Tejada, S.; de Cerio, A.L.-D.; Gállego Pérez-Larraya, J.; Espinós, J.; Idoate, M.A.; Domínguez, P.D.; de Eulate, R.G.; Aristu, J.; Bendandi, M.; et al. A Phase II Trial of Autologous Dendritic Cell Vaccination and Radiochemotherapy Following Fluorescence-Guided Surgery in Newly Diagnosed Glioblastoma Patients. J. Transl. Med. 2017, 15, 104. [Google Scholar] [CrossRef]
  669. Bota, D.A.; Taylor, T.H.; Piccioni, D.E.; Duma, C.M.; LaRocca, R.V.; Kesari, S.; Carrillo, J.A.; Abedi, M.; Aiken, R.D.; Hsu, F.P.K.; et al. Phase 2 Study of AV-GBM-1 (a Tumor-Initiating Cell Targeted Dendritic Cell Vaccine) in Newly Diagnosed Glioblastoma Patients: Safety and Efficacy Assessment. J. Exp. Clin. Cancer Res. 2022, 41, 344. [Google Scholar] [CrossRef]
  670. Batich, K.A.; Reap, E.A.; Archer, G.E.; Sanchez-Perez, L.; Nair, S.K.; Schmittling, R.J.; Norberg, P.; Xie, W.; Herndon, J.E.; Healy, P.; et al. Long-Term Survival in Glioblastoma with Cytomegalovirus Pp65-Targeted Vaccination. Clin. Cancer Res. 2017, 23, 1898–1909. [Google Scholar] [CrossRef]
  671. Liau, L.M.; Ashkan, K.; Brem, S.; Campian, J.L.; Trusheim, J.E.; Iwamoto, F.M.; Tran, D.D.; Ansstas, G.; Cobbs, C.S.; Heth, J.A.; et al. Association of Autologous Tumor Lysate-Loaded Dendritic Cell Vaccination With Extension of Survival Among Patients With Newly Diagnosed and Recurrent Glioblastoma: A Phase 3 Prospective Externally Controlled Cohort Trial. JAMA Oncol. 2023, 9, 112. [Google Scholar] [CrossRef]
  672. Yao, Y.; Luo, F.; Tang, C.; Chen, D.; Qin, Z.; Hua, W.; Xu, M.; Zhong, P.; Yu, S.; Chen, D.; et al. Molecular Subgroups and B7-H4 Expression Levels Predict Responses to Dendritic Cell Vaccines in Glioblastoma: An Exploratory Randomized Phase II Clinical Trial. Cancer Immunol. Immunother. 2018, 67, 1777–1788. [Google Scholar] [CrossRef]
  673. Zadeh, G.; Daras, M.; Cloughesy, T.F.; Colman, H.; Kumthekar, P.U.; Chen, C.C.; Aiken, R.; Groves, M.D.; Ong, S.; Ramakrishna, R.; et al. LTBK-04. PHASE 2 MULTICENTER STUDY OF THE ONCOLYTIC ADENOVIRUS DNX-2401 (TASADENOTUREV) IN COMBINATION WITH PEMBROLIZUMAB FOR RECURRENT GLIOBLASTOMA; CAPTIVE STUDY (KEYNOTE-192). Neuro-Oncology 2020, 22, ii237. [Google Scholar] [CrossRef]
  674. Nassiri, F.; Patil, V.; Yefet, L.S.; Singh, O.; Liu, J.; Dang, R.M.A.; Yamaguchi, T.N.; Daras, M.; Cloughesy, T.F.; Colman, H.; et al. Oncolytic DNX-2401 Virotherapy plus Pembrolizumab in Recurrent Glioblastoma: A Phase 1/2 Trial. Nat. Med. 2023, 29, 1370–1378. [Google Scholar] [CrossRef]
  675. Ishikawa, E.; Muragaki, Y.; Yamamoto, T.; Maruyama, T.; Tsuboi, K.; Ikuta, S.; Hashimoto, K.; Uemae, Y.; Ishihara, T.; Matsuda, M.; et al. Phase I/IIa Trial of Fractionated Radiotherapy, Temozolomide, and Autologous Formalin-Fixed Tumor Vaccine for Newly Diagnosed Glioblastoma. J. Neurosurg. 2014, 121, 543–553. [Google Scholar] [CrossRef]
  676. Todo, T.; Ito, H.; Ino, Y.; Ohtsu, H.; Ota, Y.; Shibahara, J.; Tanaka, M. Intratumoral Oncolytic Herpes Virus G47∆ for Residual or Recurrent Glioblastoma: A Phase 2 Trial. Nat. Med. 2022, 28, 1630–1639. [Google Scholar] [CrossRef]
  677. Westphal, M.; Ylä-Herttuala, S.; Martin, J.; Warnke, P.; Menei, P.; Eckland, D.; Kinley, J.; Kay, R.; Ram, Z. Adenovirus-Mediated Gene Therapy with Sitimagene Ceradenovec Followed by Intravenous Ganciclovir for Patients with Operable High-Grade Glioma (ASPECT): A Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2013, 14, 823–833. [Google Scholar] [CrossRef]
  678. Bloch, O.; Crane, C.A.; Fuks, Y.; Kaur, R.; Aghi, M.K.; Berger, M.S.; Butowski, N.A.; Chang, S.M.; Clarke, J.L.; McDermott, M.W.; et al. Heat-Shock Protein Peptide Complex-96 Vaccination for Recurrent Glioblastoma: A Phase II, Single-Arm Trial. Neuro Oncol. 2014, 16, 274–279. [Google Scholar] [CrossRef]
  679. Wang, Y.; Li, C.; Chi, X.; Huang, X.; Gao, H.; Ji, N.; Zhang, Y. Low MxA Expression Predicts Better Immunotherapeutic Outcomes in Glioblastoma Patients Receiving Heat Shock Protein Peptide Complex 96 Vaccination. Front. Oncol. 2022, 12, 865779. [Google Scholar] [CrossRef]
  680. Bloch, O.; Shi, Q.; Anderson, S.K.; Knopp, M.; Raizer, J.; Clarke, J.; Waziri, A.; Colman, H.; Bruce, J.; Olson, J.J.; et al. ATIM-14. ALLIANCE A071101: A PHASE II RANDOMIZED TRIAL COMPARING THE EFFICACY OF HEAT SHOCK PROTEIN PEPTIDE COMPLEX-96 (HSPPC-96) VACCINE GIVEN WITH BEVACIZUMAB VERSUS BEVACIZUMAB ALONE IN THE TREATMENT OF SURGICALLY RESECTABLE RECURRENT GLIOBLASTOMA. Neuro Oncol. 2017, 19, vi29. [Google Scholar] [CrossRef]
  681. Wen, P.Y.; Reardon, D.A.; Armstrong, T.S.; Phuphanich, S.; Aiken, R.D.; Landolfi, J.C.; Curry, W.T.; Zhu, J.-J.; Glantz, M.; Peereboom, D.M.; et al. A Randomized Double-Blind Placebo-Controlled Phase II Trial of Dendritic Cell Vaccine ICT-107 in Newly Diagnosed Patients with Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 5799. [Google Scholar] [CrossRef]
  682. Mulholland, P.J.; Brown, N.F.; McBain, C.; Brazil, L.; Peoples, S.; Jefferies, S.; Harris, F.; Plaha, P.; Vinayan, A.; Brooks, C.; et al. A Randomised Phase II Multicentre Study of Ipilimumab with Temozolomide vs Temozolomide Alone after Surgery and Chemoradiotherapy in Patients with Recently Diagnosed Glioblastoma: Ipi-Glio. JCO 2023, 41, LBA2023. [Google Scholar] [CrossRef]
  683. Schalper, K.A.; Rodriguez-Ruiz, M.E.; Diez-Valle, R.; López-Janeiro, A.; Porciuncula, A.; Idoate, M.A.; Inogés, S.; de Andrea, C.; López-Diaz de Cerio, A.; Tejada, S.; et al. Neoadjuvant Nivolumab Modifies the Tumor Immune Microenvironment in Resectable Glioblastoma. Nat. Med. 2019, 25, 470–476. [Google Scholar] [CrossRef]
  684. Tran, D.D.; Ghiaseddin, A.P.; Chen, D.D.; Le, S.B. Final Analysis of 2-THE-TOP: A Phase 2 Study of TTFields (Optune) plus Pembrolizumab plus Maintenance Temozolomide (TMZ) in Patients with Newly Diagnosed Glioblastoma. JCO 2023, 41, 2024. [Google Scholar] [CrossRef]
  685. Campian, J.L.; Butt, O.H.; Huang, J.; Luo, J.; Tao, Y.; Strowd, R.E.; Hissim, D.C.; Kizilbash, S.H.; Abraham, C.; Ansstas, G.; et al. Preliminary Results of a Phase II Study of Retifanlimab (PD-1 Inhibitor) plus or Minus Epacadostat (IDO1 Inhibitor) in Combination with Bevacizumab and Hypofractionated Radiotherapy for Recurrent Glioblastoma: NCT03532295. JCO 2022, 40, 2058. [Google Scholar] [CrossRef]
  686. Reardon, D.A.; Desjardins, A.; Vredenburgh, J.J.; O’Rourke, D.M.; Tran, D.D.; Fink, K.L.; Nabors, L.B.; Li, G.; Bota, D.A.; Lukas, R.V.; et al. Rindopepimut with Bevacizumab for Patients with Relapsed EGFRvIII-Expressing Glioblastoma (ReACT): Results of a Double-Blind Randomized Phase II Trial. Clin. Cancer Res. 2020, 26, 1586–1594. [Google Scholar] [CrossRef]
  687. Schuster, J.; Lai, R.K.; Recht, L.D.; Reardon, D.A.; Paleologos, N.A.; Groves, M.D.; Mrugala, M.M.; Jensen, R.; Baehring, J.M.; Sloan, A.; et al. A Phase II, Multicenter Trial of Rindopepimut (CDX-110) in Newly Diagnosed Glioblastoma: The ACT III Study. Neuro Oncol. 2015, 17, 854–861. [Google Scholar] [CrossRef]
  688. Weller, M.; Butowski, N.; Tran, D.D.; Recht, L.D.; Lim, M.; Hirte, H.; Ashby, L.; Mechtler, L.; Goldlust, S.A.; Iwamoto, F.; et al. Rindopepimut with Temozolomide for Patients with Newly Diagnosed, EGFRvIII-Expressing Glioblastoma (ACT IV): A Randomised, Double-Blind, International Phase 3 Trial. Lancet Oncol. 2017, 18, 1373–1385. [Google Scholar] [CrossRef] [PubMed]
  689. Ahluwalia, M.S.; Reardon, D.A.; Abad, A.P.; Curry, W.T.; Wong, E.T.; Figel, S.A.; Mechtler, L.L.; Peereboom, D.M.; Hutson, A.D.; Withers, H.G.; et al. Phase IIa Study of SurVaxM Plus Adjuvant Temozolomide for Newly Diagnosed Glioblastoma. J. Clin. Oncol. 2023, 41, 1453–1465. [Google Scholar] [CrossRef] [PubMed]
  690. Cloughesy, T.F.; Petrecca, K.; Walbert, T.; Butowski, N.; Salacz, M.; Perry, J.; Damek, D.; Bota, D.; Bettegowda, C.; Zhu, J.-J.; et al. Effect of Vocimagene Amiretrorepvec in Combination With Flucytosine vs Standard of Care on Survival Following Tumor Resection in Patients With Recurrent High-Grade Glioma: A Randomized Clinical Trial. JAMA Oncol. 2020, 6, 1939–1946. [Google Scholar] [CrossRef] [PubMed]
  691. Brenner, A.J.; Peters, K.B.; Vredenburgh, J.; Bokstein, F.; Blumenthal, D.T.; Yust-Katz, S.; Peretz, I.; Oberman, B.; Freedman, L.S.; Ellingson, B.M.; et al. Safety and Efficacy of VB-111, an Anticancer Gene Therapy, in Patients with Recurrent Glioblastoma: Results of a Phase I/II Study. Neuro Oncol. 2020, 22, 694–704. [Google Scholar] [CrossRef]
  692. Cloughesy, T.F.; Brenner, A.; de Groot, J.F.; Butowski, N.A.; Zach, L.; Campian, J.L.; Ellingson, B.M.; Freedman, L.S.; Cohen, Y.C.; Lowenton-Spier, N.; et al. A Randomized Controlled Phase III Study of VB-111 Combined with Bevacizumab vs Bevacizumab Monotherapy in Patients with Recurrent Glioblastoma (GLOBE). Neuro Oncol. 2020, 22, 705–717. [Google Scholar] [CrossRef]
  693. Cloughesy, T.F.; Mochizuki, A.Y.; Orpilla, J.R.; Hugo, W.; Lee, A.H.; Davidson, T.B.; Wang, A.C.; Ellingson, B.M.; Rytlewski, J.A.; Sanders, C.M.; et al. Neoadjuvant Anti-PD-1 Immunotherapy Promotes a Survival Benefit with Intratumoral and Systemic Immune Responses in Recurrent Glioblastoma. Nat. Med. 2019, 25, 477–486. [Google Scholar] [CrossRef]
  694. Li, F.; Chen, Y.; Wu, J.; Li, C.; Chen, S.; Zhu, Z.; Qin, W.; Liu, M.; Hu, B.; Liu, S.; et al. The Earlier, the Better? A Review of Neoadjuvant Immunotherapy in Resectable Non-Small-Cell Lung Cancer. Chronic Dis. Transl. Med. 2022, 8, 100–111. [Google Scholar] [CrossRef]
  695. Liu, J.; Blake, S.J.; Yong, M.C.R.; Harjunpää, H.; Ngiow, S.F.; Takeda, K.; Young, A.; O’Donnell, J.S.; Allen, S.; Smyth, M.J.; et al. Improved Efficacy of Neoadjuvant Compared to Adjuvant Immunotherapy to Eradicate Metastatic Disease. Cancer Discov. 2016, 6, 1382–1399. [Google Scholar] [CrossRef]
  696. Amaria, R.N.; Reddy, S.M.; Tawbi, H.A.; Davies, M.A.; Ross, M.I.; Glitza, I.C.; Cormier, J.N.; Lewis, C.; Hwu, W.-J.; Hanna, E.; et al. Neoadjuvant Immune Checkpoint Blockade in High-Risk Resectable Melanoma. Nat. Med. 2018, 24, 1649–1654. [Google Scholar] [CrossRef] [PubMed]
  697. Shinada, K.; Murakami, S. Neoadjuvant PD-1 Blockade in Non-Small Cell Lung Cancer: Current Perspectives and Moving Forward. Onco Targets Ther. 2023, 16, 99–108. [Google Scholar] [CrossRef] [PubMed]
  698. Reardon, D.A.; Gokhale, P.C.; Klein, S.R.; Ligon, K.L.; Rodig, S.J.; Ramkissoon, S.H.; Jones, K.L.; Conway, A.S.; Liao, X.; Zhou, J.; et al. Glioblastoma Eradication Following Immune Checkpoint Blockade in an Orthotopic, Immunocompetent Model. Cancer Immunol. Res. 2016, 4, 124–135. [Google Scholar] [CrossRef]
  699. Goswami, S.; Walle, T.; Cornish, A.E.; Basu, S.; Anandhan, S.; Fernandez, I.; Vence, L.; Blando, J.; Zhao, H.; Yadav, S.S.; et al. Immune Profiling of Human Tumors Identifies CD73 as a Combinatorial Target in Glioblastoma. Nat. Med. 2020, 26, 39–46. [Google Scholar] [CrossRef]
  700. Omuro, A.; Vlahovic, G.; Lim, M.; Sahebjam, S.; Baehring, J.; Cloughesy, T.; Voloschin, A.; Ramkissoon, S.H.; Ligon, K.L.; Latek, R.; et al. Nivolumab with or without Ipilimumab in Patients with Recurrent Glioblastoma: Results from Exploratory Phase I Cohorts of CheckMate 143. Neuro-Oncology 2018, 20, 674–686. [Google Scholar] [CrossRef] [PubMed]
  701. Fan, Y.; Wang, T.; Lei, J.; Fei, F.; Liu, J.; Liu, Y. Effects of Postoperative Radiotherapy and Docetaxel and PD-1 Inhibitors on the Survival and Safety of Glioblastoma Patients: A Systematic Review and Meta-Analysis. Ann. Transl. Med. 2022, 10, 1326. [Google Scholar] [CrossRef] [PubMed]
  702. Thommen, D.S.; Schreiner, J.; Müller, P.; Herzig, P.; Roller, A.; Belousov, A.; Umana, P.; Pisa, P.; Klein, C.; Bacac, M.; et al. Progression of Lung Cancer Is Associated with Increased Dysfunction of T Cells Defined by Coexpression of Multiple Inhibitory Receptors. Cancer Immunol. Res. 2015, 3, 1344–1355. [Google Scholar] [CrossRef]
  703. Arlauckas, S.P.; Garris, C.S.; Kohler, R.H.; Kitaoka, M.; Cuccarese, M.F.; Yang, K.S.; Miller, M.A.; Carlson, J.C.; Freeman, G.J.; Anthony, R.M.; et al. In Vivo Imaging Reveals a Tumor-Associated Macrophage-Mediated Resistance Pathway in Anti-PD-1 Therapy. Sci. Transl. Med. 2017, 9, eaal3604. [Google Scholar] [CrossRef]
  704. Pauken, K.E.; Sammons, M.A.; Odorizzi, P.M.; Manne, S.; Godec, J.; Khan, O.; Drake, A.M.; Chen, Z.; Sen, D.R.; Kurachi, M.; et al. Epigenetic Stability of Exhausted T Cells Limits Durability of Reinvigoration by PD-1 Blockade. Science 2016, 354, 1160–1165. [Google Scholar] [CrossRef]
  705. Andrews, L.P.; Marciscano, A.E.; Drake, C.G.; Vignali, D.A.A. LAG3 (CD223) as a Cancer Immunotherapy Target. Immunol. Rev. 2017, 276, 80–96. [Google Scholar] [CrossRef]
  706. Harris-Bookman, S.; Mathios, D.; Martin, A.M.; Xia, Y.; Kim, E.; Xu, H.; Belcaid, Z.; Polanczyk, M.; Barberi, T.; Theodros, D.; et al. Expression of LAG-3 and Efficacy of Combination Treatment with Anti-LAG-3 and Anti-PD-1 Monoclonal Antibodies in Glioblastoma. Int. J. Cancer 2018, 143, 3201–3208. [Google Scholar] [CrossRef]
  707. Mair, M.J.; Kiesel, B.; Feldmann, K.; Widhalm, G.; Dieckmann, K.; Wöhrer, A.; Müllauer, L.; Preusser, M.; Berghoff, A.S. LAG-3 Expression in the Inflammatory Microenvironment of Glioma. J. Neurooncol. 2021, 152, 533–539. [Google Scholar] [CrossRef] [PubMed]
  708. Xu, Y.; Zhang, H.; Sun, Q.; Geng, R.; Yuan, F.; Liu, B.; Chen, Q. Immunomodulatory Effects of Tryptophan Metabolism in the Glioma Tumor Microenvironment. Front. Immunol. 2021, 12, 730289. [Google Scholar] [CrossRef] [PubMed]
  709. Bello, C.; Heinisch, P.P.; Mihalj, M.; Carrel, T.; Luedi, M.M. Indoleamine-2,3-Dioxygenase as a Perioperative Marker of the Immune System. Front. Physiol. 2021, 12, 766511. [Google Scholar] [CrossRef] [PubMed]
  710. Peyraud, F.; Guegan, J.-P.; Bodet, D.; Cousin, S.; Bessede, A.; Italiano, A. Targeting Tryptophan Catabolism in Cancer Immunotherapy Era: Challenges and Perspectives. Front. Immunol. 2022, 13, 807271. [Google Scholar] [CrossRef] [PubMed]
  711. Ball, H.J.; Jusof, F.F.; Bakmiwewa, S.M.; Hunt, N.H.; Yuasa, H.J. Tryptophan-Catabolizing Enzymes—Party of Three. Front. Immunol. 2014, 5, 485. [Google Scholar] [CrossRef]
  712. Dolšak, A.; Gobec, S.; Sova, M. Indoleamine and Tryptophan 2,3-Dioxygenases as Important Future Therapeutic Targets. Pharmacol. Ther. 2021, 221, 107746. [Google Scholar] [CrossRef]
  713. Munn, D.H.; Zhou, M.; Attwood, J.T.; Bondarev, I.; Conway, S.J.; Marshall, B.; Brown, C.; Mellor, A.L. Prevention of Allogeneic Fetal Rejection by Tryptophan Catabolism. Science 1998, 281, 1191–1193. [Google Scholar] [CrossRef] [PubMed]
  714. Zhai, L.; Ladomersky, E.; Lauing, K.L.; Wu, M.; Genet, M.; Gritsina, G.; Győrffy, B.; Brastianos, P.K.; Binder, D.C.; Sosman, J.A.; et al. Infiltrating T Cells Increase IDO1 Expression in Glioblastoma and Contribute to Decreased Patient Survival. Clin. Cancer Res. 2017, 23, 6650–6660. [Google Scholar] [CrossRef] [PubMed]
  715. Jung, M.-Y.; Aibaidula, A.; Brown, D.A.; Himes, B.T.; Cumba Garcia, L.M.; Parney, I.F. Superinduction of Immunosuppressive Glioblastoma Extracellular Vesicles by IFN-γ through PD-L1 and IDO1. Neurooncol. Adv. 2022, 4, vdac017. [Google Scholar] [CrossRef] [PubMed]
  716. Wainwright, D.A.; Balyasnikova, I.V.; Chang, A.L.; Ahmed, A.U.; Moon, K.-S.; Auffinger, B.; Tobias, A.L.; Han, Y.; Lesniak, M.S. IDO Expression in Brain Tumors Increases the Recruitment of Regulatory T Cells and Negatively Impacts Survival. Clin. Cancer Res. 2012, 18, 6110–6121. [Google Scholar] [CrossRef] [PubMed]
  717. Kim, M.; Tomek, P. Tryptophan: A Rheostat of Cancer Immune Escape Mediated by Immunosuppressive Enzymes IDO1 and TDO. Front. Immunol. 2021, 12, 636081. [Google Scholar] [CrossRef]
  718. Song, X.; Si, Q.; Qi, R.; Liu, W.; Li, M.; Guo, M.; Wei, L.; Yao, Z. Indoleamine 2,3-Dioxygenase 1: A Promising Therapeutic Target in Malignant Tumor. Front. Immunol. 2021, 12. [Google Scholar] [CrossRef]
  719. Du, L.; Xing, Z.; Tao, B.; Li, T.; Yang, D.; Li, W.; Zheng, Y.; Kuang, C.; Yang, Q. Both IDO1 and TDO Contribute to the Malignancy of Gliomas via the Kyn–AhR–AQP4 Signaling Pathway. Sig Transduct. Target. Ther. 2020, 5, 1–13. [Google Scholar] [CrossRef]
  720. Campesato, L.F.; Budhu, S.; Tchaicha, J.; Weng, C.-H.; Gigoux, M.; Cohen, I.J.; Redmond, D.; Mangarin, L.; Pourpe, S.; Liu, C.; et al. Blockade of the AHR Restricts a Treg-Macrophage Suppressive Axis Induced by L-Kynurenine. Nat. Commun. 2020, 11, 4011. [Google Scholar] [CrossRef] [PubMed]
  721. Zhai, L.; Genet, M.; Ladomersky, E.; Lauing, K.; Wu, M.; Binder, D.; Kim, L.; Rich, J.; Horbinski, C.; James, C.D.; et al. IMST-39. IDO1 IS PROGNOSTIC FOR GLIOBLASTOMA PATIENT SURVIVAL AND CENTRALLY CORRELATES WITH POTENTLY IMMUNOSUPPRESSIVE MEDIATORS. Neuro-Oncology 2016, 18, vi95. [Google Scholar] [CrossRef]
  722. Ahlstedt, J.; Konradsson, E.; Ceberg, C.; Redebrandt, H.N. Increased Effect of Two-Fraction Radiotherapy in Conjunction with IDO1 Inhibition in Experimental Glioblastoma. PLoS ONE 2020, 15, e0233617. [Google Scholar] [CrossRef]
  723. Ozawa, Y.; Yamamuro, S.; Sano, E.; Tatsuoka, J.; Hanashima, Y.; Yoshimura, S.; Sumi, K.; Hara, H.; Nakayama, T.; Suzuki, Y.; et al. Indoleamine 2,3-Dioxygenase 1 Is Highly Expressed in Glioma Stem Cells. Biochem. Biophys. Res. Commun. 2020, 524, 723–729. [Google Scholar] [CrossRef]
  724. Hosseinalizadeh, H.; Mahmoodpour, M.; Samadani, A.A.; Roudkenar, M.H. The Immunosuppressive Role of Indoleamine 2, 3-Dioxygenase in Glioblastoma: Mechanism of Action and Immunotherapeutic Strategies. Med. Oncol. 2022, 39, 130. [Google Scholar] [CrossRef]
  725. Zhai, L.; Bell, A.; Ladomersky, E.; Lauing, K.L.; Bollu, L.; Nguyen, B.; Genet, M.; Kim, M.; Chen, P.; Mi, X.; et al. Tumor Cell IDO Enhances Immune Suppression and Decreases Survival Independent of Tryptophan Metabolism in Glioblastoma. Clin. Cancer Res. 2021, 27, 6514–6528. [Google Scholar] [CrossRef]
  726. Pilotte, L.; Larrieu, P.; Stroobant, V.; Colau, D.; Dolusic, E.; Frédérick, R.; De Plaen, E.; Uyttenhove, C.; Wouters, J.; Masereel, B.; et al. Reversal of Tumoral Immune Resistance by Inhibition of Tryptophan 2,3-Dioxygenase. Proc. Natl. Acad. Sci. USA 2012, 109, 2497–2502. [Google Scholar] [CrossRef]
  727. Panitz, V.; Končarević, S.; Sadik, A.; Friedel, D.; Bausbacher, T.; Trump, S.; Farztdinov, V.; Schulz, S.; Sievers, P.; Schmidt, S.; et al. Tryptophan Metabolism Is Inversely Regulated in the Tumor and Blood of Patients with Glioblastoma. Theranostics 2021, 11, 9217–9233. [Google Scholar] [CrossRef]
  728. Munn, D.H.; Sharma, M.D.; Baban, B.; Harding, H.P.; Zhang, Y.; Ron, D.; Mellor, A.L. GCN2 Kinase in T Cells Mediates Proliferative Arrest and Anergy Induction in Response to Indoleamine 2,3-Dioxygenase. Immunity 2005, 22, 633–642. [Google Scholar] [CrossRef]
  729. Rashidi, A.; Miska, J.; Lee-Chang, C.; Kanojia, D.; Panek, W.K.; Lopez-Rosas, A.; Zhang, P.; Han, Y.; Xiao, T.; Pituch, K.C.; et al. GCN2 Is Essential for CD8+ T Cell Survival and Function in Murine Models of Malignant Glioma. Cancer Immunol. Immunother. 2020, 69, 81–94. [Google Scholar] [CrossRef]
  730. Eleftheriadis, T.; Pissas, G.; Antoniadi, G.; Liakopoulos, V.; Stefanidis, I. Indoleamine 2,3-Dioxygenase Depletes Tryptophan, Activates General Control Non-Derepressible 2 Kinase and down-Regulates Key Enzymes Involved in Fatty Acid Synthesis in Primary Human CD4+ T Cells. Immunology 2015, 146, 292–300. [Google Scholar] [CrossRef] [PubMed]
  731. Platten, M.; Friedrich, M.; Wainwright, D.A.; Panitz, V.; Opitz, C.A. Tryptophan Metabolism in Brain Tumors—IDO and Beyond. Curr. Opin. Immunol. 2021, 70, 57–66. [Google Scholar] [CrossRef] [PubMed]
  732. Sonner, J.K.; Deumelandt, K.; Ott, M.; Thomé, C.M.; Rauschenbach, K.J.; Schulz, S.; Munteanu, B.; Mohapatra, S.; Adam, I.; Hofer, A.-C.; et al. The Stress Kinase GCN2 Does Not Mediate Suppression of Antitumor T Cell Responses by Tryptophan Catabolism in Experimental Melanomas. Oncoimmunology 2016, 5, e1240858. [Google Scholar] [CrossRef] [PubMed]
  733. Nguyen, N.T.; Kimura, A.; Nakahama, T.; Chinen, I.; Masuda, K.; Nohara, K.; Fujii-Kuriyama, Y.; Kishimoto, T. Aryl Hydrocarbon Receptor Negatively Regulates Dendritic Cell Immunogenicity via a Kynurenine-Dependent Mechanism. Proc. Natl. Acad. Sci. USA 2010, 107, 19961–19966. [Google Scholar] [CrossRef]
  734. Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An Endogenous Tumour-Promoting Ligand of the Human Aryl Hydrocarbon Receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef]
  735. Gramatzki, D.; Pantazis, G.; Schittenhelm, J.; Tabatabai, G.; Köhle, C.; Wick, W.; Schwarz, M.; Weller, M.; Tritschler, I. Aryl Hydrocarbon Receptor Inhibition Downregulates the TGF-Beta/Smad Pathway in Human Glioblastoma Cells. Oncogene 2009, 28, 2593–2605. [Google Scholar] [CrossRef]
  736. Sadik, A.; Somarribas Patterson, L.F.; Öztürk, S.; Mohapatra, S.R.; Panitz, V.; Secker, P.F.; Pfänder, P.; Loth, S.; Salem, H.; Prentzell, M.T.; et al. IL4I1 Is a Metabolic Immune Checkpoint That Activates the AHR and Promotes Tumor Progression. Cell 2020, 182, 1252–1270.e34. [Google Scholar] [CrossRef]
  737. Liu, Y.; Liang, X.; Dong, W.; Fang, Y.; Lv, J.; Zhang, T.; Fiskesund, R.; Xie, J.; Liu, J.; Yin, X.; et al. Tumor-Repopulating Cells Induce PD-1 Expression in CD8+ T Cells by Transferring Kynurenine and AhR Activation. Cancer Cell 2018, 33, 480–494.e7. [Google Scholar] [CrossRef] [PubMed]
  738. Zhang, X.; Liu, X.; Zhou, W.; Du, Q.; Yang, M.; Ding, Y.; Hu, R. Blockade of IDO-Kynurenine-AhR Axis Ameliorated Colitis-Associated Colon Cancer via Inhibiting Immune Tolerance. Cell Mol. Gastroenterol. Hepatol. 2021, 12, 1179–1199. [Google Scholar] [CrossRef]
  739. Goudot, C.; Coillard, A.; Villani, A.-C.; Gueguen, P.; Cros, A.; Sarkizova, S.; Tang-Huau, T.-L.; Bohec, M.; Baulande, S.; Hacohen, N.; et al. Aryl Hydrocarbon Receptor Controls Monocyte Differentiation into Dendritic Cells versus Macrophages. Immunity 2017, 47, 582–596.e6. [Google Scholar] [CrossRef]
  740. Piper, C.J.M.; Rosser, E.C.; Oleinika, K.; Nistala, K.; Krausgruber, T.; Rendeiro, A.F.; Banos, A.; Drozdov, I.; Villa, M.; Thomson, S.; et al. Aryl Hydrocarbon Receptor Contributes to the Transcriptional Program of IL-10-Producing Regulatory B Cells. Cell Rep. 2019, 29, 1878–1892.e7. [Google Scholar] [CrossRef] [PubMed]
  741. Takenaka, M.C.; Gabriely, G.; Rothhammer, V.; Mascanfroni, I.D.; Wheeler, M.A.; Chao, C.-C.; Gutiérrez-Vázquez, C.; Kenison, J.; Tjon, E.C.; Barroso, A.; et al. Control of Tumor-Associated Macrophages and T Cells in Glioblastoma via AHR and CD39. Nat. Neurosci. 2019, 22, 729–740. [Google Scholar] [CrossRef] [PubMed]
  742. Gabriely, G.; Quintana, F.J. Role of AHR in the Control of GBM-Associated Myeloid Cells. Semin. Cancer Biol. 2020, 64, 13–18. [Google Scholar] [CrossRef] [PubMed]
  743. Mohan, A.A.; Tomaszewski, W.H.; Haskell-Mendoza, A.P.; Hotchkiss, K.M.; Singh, K.; Reedy, J.L.; Fecci, P.E.; Sampson, J.H.; Khasraw, M. Targeting Immunometabolism in Glioblastoma. Front. Oncol. 2021, 11, 696402. [Google Scholar] [CrossRef]
  744. Adams, S.; Teo, C.; McDonald, K.L.; Zinger, A.; Bustamante, S.; Lim, C.K.; Sundaram, G.; Braidy, N.; Brew, B.J.; Guillemin, G.J. Involvement of the Kynurenine Pathway in Human Glioma Pathophysiology. PLoS ONE 2014, 9, e112945. [Google Scholar] [CrossRef]
  745. Triplett, T.A.; Garrison, K.C.; Marshall, N.; Donkor, M.; Blazeck, J.; Lamb, C.; Qerqez, A.; Dekker, J.D.; Tanno, Y.; Lu, W.-C.; et al. Reversal of Indoleamine 2,3-Dioxygenase-Mediated Cancer Immune Suppression by Systemic Kynurenine Depletion with a Therapeutic Enzyme. Nat. Biotechnol. 2018, 36, 758–764. [Google Scholar] [CrossRef]
  746. Hu, M.; Zhou, W.; Wang, Y.; Yao, D.; Ye, T.; Yao, Y.; Chen, B.; Liu, G.; Yang, X.; Wang, W.; et al. Discovery of the First Potent Proteolysis Targeting Chimera (PROTAC) Degrader of Indoleamine 2,3-Dioxygenase 1. Acta Pharm. Sin. B 2020, 10, 1943–1953. [Google Scholar] [CrossRef] [PubMed]
  747. Tang, K.; Wu, Y.-H.; Song, Y.; Yu, B. Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitors in Clinical Trials for Cancer Immunotherapy. J. Hematol. Oncol. 2021, 14, 68. [Google Scholar] [CrossRef] [PubMed]
  748. Wainwright, D.A.; Chang, A.L.; Dey, M.; Balyasnikova, I.V.; Kim, C.K.; Tobias, A.; Cheng, Y.; Kim, J.W.; Qiao, J.; Zhang, L.; et al. Durable Therapeutic Efficacy Utilizing Combinatorial Blockade against IDO, CTLA-4, and PD-L1 in Mice with Brain Tumors. Clin. Cancer Res. 2014, 20, 5290–5301. [Google Scholar] [CrossRef]
  749. Ladomersky, E.; Zhai, L.; Lenzen, A.; Lauing, K.L.; Qian, J.; Scholtens, D.M.; Gritsina, G.; Sun, X.; Liu, Y.; Yu, F.; et al. IDO1 Inhibition Synergizes with Radiation and PD-1 Blockade to Durably Increase Survival Against Advanced Glioblastoma. Clin. Cancer Res. 2018, 24, 2559–2573. [Google Scholar] [CrossRef] [PubMed]
  750. Liu, M.; Li, Z.; Yao, W.; Zeng, X.; Wang, L.; Cheng, J.; Ma, B.; Zhang, R.; Min, W.; Wang, H. IDO Inhibitor Synergized with Radiotherapy to Delay Tumor Growth by Reversing T Cell Exhaustion. Mol. Med. Rep. 2020, 21, 445–453. [Google Scholar] [CrossRef] [PubMed]
  751. Li, M.; Bolduc, A.R.; Hoda, M.N.; Gamble, D.N.; Dolisca, S.-B.; Bolduc, A.K.; Hoang, K.; Ashley, C.; McCall, D.; Rojiani, A.M.; et al. The Indoleamine 2,3-Dioxygenase Pathway Controls Complement-Dependent Enhancement of Chemo-Radiation Therapy against Murine Glioblastoma. J. Immunother. Cancer 2014, 2, 21. [Google Scholar] [CrossRef] [PubMed]
  752. Gomes, B.; Driessens, G.; Bartlett, D.; Cai, D.; Cauwenberghs, S.; Crosignani, S.; Dalvie, D.; Denies, S.; Dillon, C.P.; Fantin, V.R.; et al. Characterization of the Selective Indoleamine 2,3-Dioxygenase-1 (IDO1) Catalytic Inhibitor EOS200271/PF-06840003 Supports IDO1 as a Critical Resistance Mechanism to PD-(L)1 Blockade Therapy. Mol. Cancer Ther. 2018, 17, 2530–2542. [Google Scholar] [CrossRef] [PubMed]
  753. Reardon, D.A.; Desjardins, A.; Rixe, O.; Cloughesy, T.; Alekar, S.; Williams, J.H.; Li, R.; Taylor, C.T.; Lassman, A.B. A Phase 1 Study of PF-06840003, an Oral Indoleamine 2,3-Dioxygenase 1 (IDO1) Inhibitor in Patients with Recurrent Malignant Glioma. Investig. New Drugs 2020, 38, 1784–1795. [Google Scholar] [CrossRef] [PubMed]
  754. Prendergast, G.C.; Malachowski, W.J.; Mondal, A.; Scherle, P.; Muller, A.J. Indoleamine 2,3-Dioxygenase and Its Therapeutic Inhibition in Cancer. Int. Rev. Cell Mol. Biol. 2018, 336, 175–203. [Google Scholar] [CrossRef] [PubMed]
  755. Hanihara, M.; Kawataki, T.; Oh-Oka, K.; Mitsuka, K.; Nakao, A.; Kinouchi, H. Synergistic Antitumor Effect with Indoleamine 2,3-Dioxygenase Inhibition and Temozolomide in a Murine Glioma Model. J. Neurosurg. 2016, 124, 1594–1601. [Google Scholar] [CrossRef] [PubMed]
  756. Powderly, J.D.; Klempner, S.J.; Naing, A.; Bendell, J.; Garrido-Laguna, I.; Catenacci, D.V.T.; Taylor, M.H.; Lee, J.J.; Zheng, F.; Zhou, F.; et al. Epacadostat Plus Pembrolizumab and Chemotherapy for Advanced Solid Tumors: Results from the Phase I/II ECHO-207/KEYNOTE-723 Study. Oncologist 2022, 27, 905-e848. [Google Scholar] [CrossRef]
  757. Ladomersky, E.; Zhai, L.; Lauing, K.L.; Bell, A.; Xu, J.; Kocherginsky, M.; Zhang, B.; Wu, J.D.; Podojil, J.R.; Platanias, L.C.; et al. Advanced Age Increases Immunosuppression in the Brain and Decreases Immunotherapeutic Efficacy in Subjects with Glioblastoma. Clin. Cancer Res. 2020, 26, 5232–5245. [Google Scholar] [CrossRef]
  758. Niedbała, M.; Malarz, K.; Sharma, G.; Kramer-Marek, G.; Kaspera, W. Glioblastoma: Pitfalls and Opportunities of Immunotherapeutic Combinations. Onco Targets Ther. 2022, 15, 437–468. [Google Scholar] [CrossRef]
  759. Zhao, T.; Li, C.; Ge, H.; Lin, Y.; Kang, D. Glioblastoma Vaccine Tumor Therapy Research Progress. Chin. Neurosurg. J. 2022, 8, 2. [Google Scholar] [CrossRef]
  760. Parney, I.F.; Chang, L.-J.; Farr-Jones, M.A.; Hao, C.; Smylie, M.; Petruk, K.C. Technical Hurdles in a Pilot Clinical Trial of Combined B7-2 and GM-CSF Immunogene Therapy for Glioblastomas and Melanomas. J. Neurooncol. 2006, 78, 71–80. [Google Scholar] [CrossRef]
  761. Curry, W.T.; Gorrepati, R.; Piesche, M.; Sasada, T.; Agarwalla, P.; Jones, P.S.; Gerstner, E.R.; Golby, A.J.; Batchelor, T.T.; Wen, P.Y.; et al. Vaccination with Irradiated Autologous Tumor Cells Mixed with Irradiated GM-K562 Cells Stimulates Antitumor Immunity and T Lymphocyte Activation in Patients with Recurrent Malignant Glioma. Clin. Cancer Res. 2016, 22, 2885–2896. [Google Scholar] [CrossRef]
  762. Andrews, D.W.; Judy, K.D.; Scott, C.B.; Garcia, S.; Harshyne, L.A.; Kenyon, L.; Talekar, K.; Flanders, A.; Atsina, K.-B.; Kim, L.; et al. Phase Ib Clinical Trial of IGV-001 for Patients with Newly Diagnosed Glioblastoma. Clin. Cancer Res. 2021, 27, 1912–1922. [Google Scholar] [CrossRef] [PubMed]
  763. Bol, K.F.; Schreibelt, G.; Rabold, K.; Wculek, S.K.; Schwarze, J.K.; Dzionek, A.; Teijeira, A.; Kandalaft, L.E.; Romero, P.; Coukos, G.; et al. The Clinical Application of Cancer Immunotherapy Based on Naturally Circulating Dendritic Cells. J. Immunother. Cancer 2019, 7, 109. [Google Scholar] [CrossRef]
  764. Lee-Chang, C.; Lesniak, M.S. Next-Generation Antigen-Presenting Cell Immune Therapeutics for Gliomas. J. Clin. Investig. 2023, 133, e163449. [Google Scholar] [CrossRef] [PubMed]
  765. Santos, P.M.; Butterfield, L.H. Dendritic Cell-Based Cancer Vaccines. J. Immunol. 2018, 200, 443–449. [Google Scholar] [CrossRef]
  766. Polyzoidis, S.; Ashkan, K. DCVax®-L—Developed by Northwest Biotherapeutics. Hum. Vaccin. Immunother. 2014, 10, 3139–3145. [Google Scholar] [CrossRef] [PubMed]
  767. Wick, W.; Bent, M.J. van den First Results on the DCVax Phase III Trial: Raising More Questions than Providing Answers. Neuro-Oncology 2018, 20, 1283. [Google Scholar] [CrossRef]
  768. Pasqualetti, F.; Zanotti, S. Nonrandomised Controlled Trial in Recurrent Glioblastoma Patients: The Promise of Autologous Tumour Lysate-Loaded Dendritic Cell Vaccination. Br. J. Cancer 2023, 1–2. [Google Scholar] [CrossRef] [PubMed]
  769. Peredo-Harvey, I.; Rahbar, A.; Söderberg-Nauclér, C. Presence of the Human Cytomegalovirus in Glioblastomas—A Systematic Review. Cancers 2021, 13, 5051. [Google Scholar] [CrossRef]
  770. Yang, T.; Liu, D.; Fang, S.; Ma, W.; Wang, Y. Cytomegalovirus and Glioblastoma: A Review of the Biological Associations and Therapeutic Strategies. J. Clin. Med. 2022, 11, 5221. [Google Scholar] [CrossRef]
  771. Dziurzynski, K.; Wei, J.; Qiao, W.; Hatiboglu, M.A.; Kong, L.-Y.; Wu, A.; Wang, Y.; Cahill, D.; Levine, N.; Prabhu, S.; et al. Glioma-Associated Cytomegalovirus Mediates Subversion of the Monocyte Lineage to a Tumor Propagating Phenotype. Clin. Cancer Res. 2011, 17, 4642–4649. [Google Scholar] [CrossRef]
  772. Stragliotto, G.; Pantalone, M.R.; Rahbar, A.; Bartek, J.; Söderberg-Naucler, C. Valganciclovir as Add-on to Standard Therapy in Glioblastoma Patients. Clin. Cancer Res. 2020, 26, 4031–4039. [Google Scholar] [CrossRef]
  773. Stragliotto, G.; Pantalone, M.R.; Rahbar, A.; Söderberg-Nauclér, C. Valganciclovir as Add-On to Standard Therapy in Secondary Glioblastoma. Microorganisms 2020, 8, 1471. [Google Scholar] [CrossRef]
  774. Smith, C.; Lineburg, K.E.; Martins, J.P.; Ambalathingal, G.R.; Neller, M.A.; Morrison, B.; Matthews, K.K.; Rehan, S.; Crooks, P.; Panikkar, A.; et al. Autologous CMV-Specific T Cells Are a Safe Adjuvant Immunotherapy for Primary Glioblastoma Multiforme. J. Clin. Investig. 2020, 130, 6041–6053. [Google Scholar] [CrossRef]
  775. Ottenhausen, M.; Bodhinayake, I.; Schaefer, P.M.; Boockvar, J.A. VIGAS and Beyond: The Impact of HCMV-Infection and Its Treatment in Glioblastoma. Neurosurgery 2014, 74, N23. [Google Scholar] [CrossRef] [PubMed]
  776. Pantalone, M.R.; Rahbar, A.; Söderberg-Naucler, C.; Stragliotto, G. Valganciclovir as Add-on to Second-Line Therapy in Patients with Recurrent Glioblastoma. Cancers 2022, 14, 1958. [Google Scholar] [CrossRef]
  777. Mitchell, D.A.; Batich, K.A.; Gunn, M.D.; Huang, M.-N.; Sanchez-Perez, L.; Nair, S.K.; Congdon, K.L.; Reap, E.A.; Archer, G.E.; Desjardins, A.; et al. Tetanus Toxoid and CCL3 Improve Dendritic Cell Vaccines in Mice and Glioblastoma Patients. Nature 2015, 519, 366–369. [Google Scholar] [CrossRef] [PubMed]
  778. Batich, K.A.; Mitchell, D.A.; Healy, P.; Herndon, J.E.; Sampson, J.H. Once, Twice, Three Times a Finding: Reproducibility of Dendritic Cell Vaccine Trials Targeting Cytomegalovirus in Glioblastoma. Clin. Cancer Res. 2020, 26, 5297–5303. [Google Scholar] [CrossRef] [PubMed]
  779. Ahn, J.; Shin, C.; Kim, Y.S.; Park, J.-S.; Jeun, S.-S.; Ahn, S. Cytomegalovirus-Specific Immunotherapy for Glioblastoma Treatments. Brain Tumor Res. Treat. 2022, 10, 135–143. [Google Scholar] [CrossRef]
  780. Rapp, M.; Grauer, O.M.; Kamp, M.; Sevens, N.; Zotz, N.; Sabel, M.; Sorg, R.V. A Randomized Controlled Phase II Trial of Vaccination with Lysate-Loaded, Mature Dendritic Cells Integrated into Standard Radiochemotherapy of Newly Diagnosed Glioblastoma (GlioVax): Study Protocol for a Randomized Controlled Trial. Trials 2018, 19, 293. [Google Scholar] [CrossRef]
  781. Cozzi, S.; Najafi, M.; Gomar, M.; Ciammella, P.; Iotti, C.; Iaccarino, C.; Dominici, M.; Pavesi, G.; Chiavelli, C.; Kazemian, A.; et al. Delayed Effect of Dendritic Cells Vaccination on Survival in Glioblastoma: A Systematic Review and Meta-Analysis. Curr. Oncol. 2022, 29, 881–891. [Google Scholar] [CrossRef]
  782. Bigner, D.D.; Pitts, O.M.; Wikstrand, C.J. Induction of Lethal Experimental Allergic Encephalomyelitis in Nonhuman Primates and Guinea Pigs with Human Glioblastoma Multiforme Tissue. J. Neurosurg. 1981, 55, 32–42. [Google Scholar] [CrossRef]
  783. Migliorini, D.; Dutoit, V.; Allard, M.; Grandjean Hallez, N.; Marinari, E.; Widmer, V.; Philippin, G.; Corlazzoli, F.; Gustave, R.; Kreutzfeldt, M.; et al. Phase I/II Trial Testing Safety and Immunogenicity of the Multipeptide IMA950/Poly-ICLC Vaccine in Newly Diagnosed Adult Malignant Astrocytoma Patients. Neuro Oncol. 2019, 21, 923–933. [Google Scholar] [CrossRef]
  784. Liu, C.J.; Schaettler, M.; Blaha, D.T.; Bowman-Kirigin, J.A.; Kobayashi, D.K.; Livingstone, A.J.; Bender, D.; Miller, C.A.; Kranz, D.M.; Johanns, T.M.; et al. Treatment of an Aggressive Orthotopic Murine Glioblastoma Model with Combination Checkpoint Blockade and a Multivalent Neoantigen Vaccine. Neuro Oncol. 2020, 22, 1276–1288. [Google Scholar] [CrossRef]
  785. Hilf, N.; Kuttruff-Coqui, S.; Frenzel, K.; Bukur, V.; Stevanović, S.; Gouttefangeas, C.; Platten, M.; Tabatabai, G.; Dutoit, V.; van der Burg, S.H.; et al. Actively Personalized Vaccination Trial for Newly Diagnosed Glioblastoma. Nature 2019, 565, 240–245. [Google Scholar] [CrossRef]
  786. Tamura, Y.; Peng, P.; Liu, K.; Daou, M.; Srivastava, P.K. Immunotherapy of Tumors with Autologous Tumor-Derived Heat Shock Protein Preparations. Science 1997, 278, 117–120. [Google Scholar] [CrossRef] [PubMed]
  787. Weller, M.; Roth, P.; Preusser, M.; Wick, W.; Reardon, D.A.; Platten, M.; Sampson, J.H. Vaccine-Based Immunotherapeutic Approaches to Gliomas and Beyond. Nat. Rev. Neurol. 2017, 13, 363–374. [Google Scholar] [CrossRef]
  788. Ji, N.; Zhang, Y.; Liu, Y.; Xie, J.; Wang, Y.; Hao, S.; Gao, Z. Heat Shock Protein Peptide Complex-96 Vaccination for Newly Diagnosed Glioblastoma: A Phase I, Single-Arm Trial. JCI Insight 2018, 3, e99145. [Google Scholar] [CrossRef]
  789. Saxena, M.; van der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic Cancer Vaccines. Nat. Rev. Cancer 2021, 21, 360–378. [Google Scholar] [CrossRef] [PubMed]
  790. Li, L.; Zhou, J.; Dong, X.; Liao, Q.; Zhou, D.; Zhou, Y. Dendritic Cell Vaccines for Glioblastoma Fail to Complete Clinical Translation: Bottlenecks and Potential Countermeasures. Int. Immunopharmacol. 2022, 109, 108929. [Google Scholar] [CrossRef] [PubMed]
  791. Xiong, Z.; Raphael, I.; Olin, M.; Okada, H.; Li, X.; Kohanbash, G. Glioblastoma Vaccines: Past, Present, and Opportunities. eBioMedicine 2024, 100. [Google Scholar] [CrossRef]
  792. Land, C.A.; Musich, P.R.; Haydar, D.; Krenciute, G.; Xie, Q. Chimeric Antigen Receptor T-Cell Therapy in Glioblastoma: Charging the T Cells to Fight. J. Transl. Med. 2020, 18, 428. [Google Scholar] [CrossRef]
  793. Pant, A.; Lim, M. CAR-T Therapy in GBM: Current Challenges and Avenues for Improvement. Cancers 2023, 15, 1249. [Google Scholar] [CrossRef]
  794. Zhang, J.; Siller-Farfán, J.A. Current and Future Perspectives of Chimeric Antigen Receptors against Glioblastoma. Immunother. Adv. 2022, 2, ltac014. [Google Scholar] [CrossRef]
  795. Murty, S.; Haile, S.T.; Beinat, C.; Aalipour, A.; Alam, I.S.; Murty, T.; Shaffer, T.M.; Patel, C.B.; Graves, E.E.; Mackall, C.L.; et al. Intravital Imaging Reveals Synergistic Effect of CAR T-Cells and Radiation Therapy in a Preclinical Immunocompetent Glioblastoma Model. OncoImmunology 2020, 9, e1757360. [Google Scholar] [CrossRef] [PubMed]
  796. Labanieh, L.; Mackall, C.L. CAR Immune Cells: Design Principles, Resistance and the next Generation. Nature 2023, 614, 635–648. [Google Scholar] [CrossRef] [PubMed]
  797. Marei, H.E.; Althani, A.; Afifi, N.; Hasan, A.; Caceci, T.; Pozzoli, G.; Cenciarelli, C. Current Progress in Chimeric Antigen Receptor T Cell Therapy for Glioblastoma Multiforme. Cancer Med. 2021, 10, 5019–5030. [Google Scholar] [CrossRef] [PubMed]
  798. Koka, V.; Potti, A.; Forseen, S.E.; Pervez, H.; Fraiman, G.N.; Koch, M.; Levitt, R. Role of Her-2/Neu Overexpression and Clinical Determinants of Early Mortality in Glioblastoma Multiforme. Am. J. Clin. Oncol. 2003, 26, 332–335. [Google Scholar] [CrossRef]
  799. Pule, M.A.; Savoldo, B.; Myers, G.D.; Rossig, C.; Russell, H.V.; Dotti, G.; Huls, M.H.; Liu, E.; Gee, A.P.; Mei, Z.; et al. Virus-Specific T Cells Engineered to Coexpress Tumor-Specific Receptors: Persistence and Antitumor Activity in Individuals with Neuroblastoma. Nat. Med. 2008, 14, 1264–1270. [Google Scholar] [CrossRef] [PubMed]
  800. Ahmed, N.; Brawley, V.; Hegde, M.; Bielamowicz, K.; Kalra, M.; Landi, D.; Robertson, C.; Gray, T.L.; Diouf, O.; Wakefield, A.; et al. HER2-Specific Chimeric Antigen Receptor–Modified Virus-Specific T Cells for Progressive Glioblastoma: A Phase 1 Dose-Escalation Trial. JAMA Oncol. 2017, 3, 1094–1101. [Google Scholar] [CrossRef] [PubMed]
  801. Liu, X.; Zhao, Z.; Dai, W.; Liao, K.; Sun, Q.; Chen, D.; Pan, X.; Feng, L.; Ding, Y.; Wei, S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers 2023, 15, 4308. [Google Scholar] [CrossRef]
  802. Jarboe, J.S.; Johnson, K.R.; Choi, Y.; Lonser, R.R.; Park, J.K. Expression of Interleukin-13 Receptor Alpha2 in Glioblastoma Multiforme: Implications for Targeted Therapies. Cancer Res. 2007, 67, 7983–7986. [Google Scholar] [CrossRef]
  803. Brown, C.E.; Warden, C.D.; Starr, R.; Deng, X.; Badie, B.; Yuan, Y.-C.; Forman, S.J.; Barish, M.E. Glioma IL13Rα2 Is Associated with Mesenchymal Signature Gene Expression and Poor Patient Prognosis. PLoS ONE 2013, 8. [Google Scholar] [CrossRef]
  804. Brown, C.E.; Badie, B.; Barish, M.E.; Weng, L.; Ostberg, J.R.; Chang, W.-C.; Naranjo, A.; Starr, R.; Wagner, J.; Wright, C.; et al. Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2015, 21, 4062–4072. [Google Scholar] [CrossRef] [PubMed]
  805. Brown, C.E.; Alizadeh, D.; Starr, R.; Weng, L.; Wagner, J.R.; Naranjo, A.; Ostberg, J.R.; Blanchard, M.S.; Kilpatrick, J.; Simpson, J.; et al. Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N. Engl. J. Med. 2016, 375, 2561–2569. [Google Scholar] [CrossRef]
  806. Woo, S.R.; Oh, Y.T.; An, J.Y.; Kang, B.G.; Nam, D.-H.; Joo, K.M. Glioblastoma Specific Antigens, GD2 and CD90, Are Not Involved in Cancer Stemness. Anat. Cell Biol. 2015, 48, 44–53. [Google Scholar] [CrossRef]
  807. Prapa, M.; Chiavelli, C.; Golinelli, G.; Grisendi, G.; Bestagno, M.; Di Tinco, R.; Dall’Ora, M.; Neri, G.; Candini, O.; Spano, C.; et al. GD2 CAR T Cells against Human Glioblastoma. npj Precis. Onc. 2021, 5, 93. [Google Scholar] [CrossRef] [PubMed]
  808. Liu, W.; Zhao, Y.; Liu, Z.; Zhang, G.; Wu, H.; Zheng, X.; Tang, X.; Chen, Z. Therapeutic Effects against High-Grade Glioblastoma Mediated by Engineered Induced Neural Stem Cells Combined with GD2-Specific CAR-NK. Cell Oncol. 2023. [Google Scholar] [CrossRef]
  809. Lee, K.S.; Lee, K.; Yun, S.; Moon, S.; Park, Y.; Han, J.H.; Kim, C.-Y.; Lee, H.S.; Choe, G. Prognostic Relevance of Programmed Cell Death Ligand 1 Expression in Glioblastoma. J. Neurooncol. 2018, 136, 453–461. [Google Scholar] [CrossRef]
  810. Karschnia, P.; Teske, N.; Thon, N.; Subklewe, M.; Tonn, J.-C.; Dietrich, J.; Baumgarten, L. von Chimeric Antigen Receptor T Cells for Glioblastoma: Current Concepts, Challenges, and Future Perspectives. Neurology 2021, 97, 218–230. [Google Scholar] [CrossRef]
  811. Suryadevara, C.M.; Desai, R.; Farber, S.H.; Choi, B.D.; Swartz, A.M.; Shen, S.H.; Gedeon, P.C.; Snyder, D.J.; Herndon, J.E.; Healy, P.; et al. Preventing Lck Activation in CAR T Cells Confers Treg Resistance but Requires 4-1BB Signaling for Them to Persist and Treat Solid Tumors in Nonlymphodepleted Hosts. Clin. Cancer Res. 2019, 25, 358–368. [Google Scholar] [CrossRef] [PubMed]
  812. Verma, A.; Rafiq, S. Chimeric Antigen Receptor (CAR) T Cell Therapy for Glioblastoma. In Cancer Immunotherapies: Solid Tumors and Hematologic Malignancies; Hays, P., Ed.; Cancer Treatment and Research; Springer International Publishing: Cham, Switzerland, 2022; pp. 161–184. ISBN 978-3-030-96376-7. [Google Scholar]
  813. Mantica, M.; Drappatz, J. Immunotherapy Associated Central Nervous System Complications in Primary Brain Tumors. Front. Oncol. 2023, 13, 1124198. [Google Scholar] [CrossRef]
  814. Majzner, R.G.; Ramakrishna, S.; Yeom, K.W.; Patel, S.; Chinnasamy, H.; Schultz, L.M.; Richards, R.M.; Jiang, L.; Barsan, V.; Mancusi, R.; et al. GD2-CAR T Cell Therapy for H3K27M-Mutated Diffuse Midline Gliomas. Nature 2022, 603, 934–941. [Google Scholar] [CrossRef]
  815. Portnow, J.; Wang, D.; Blanchard, M.S.; Tran, V.; Alizadeh, D.; Starr, R.; Dodia, R.; Chiu, V.; Brito, A.; Kilpatrick, J.; et al. Systemic Anti-PD-1 Immunotherapy Results in PD-1 Blockade on T Cells in the Cerebrospinal Fluid. JAMA Oncol. 2020, 6, 1947–1951. [Google Scholar] [CrossRef]
  816. Nakazawa, T.; Natsume, A.; Nishimura, F.; Morimoto, T.; Matsuda, R.; Nakamura, M.; Yamada, S.; Nakagawa, I.; Motoyama, Y.; Park, Y.-S.; et al. Effect of CRISPR/Cas9-Mediated PD-1-Disrupted Primary Human Third-Generation CAR-T Cells Targeting EGFRvIII on In Vitro Human Glioblastoma Cell Growth. Cells 2020, 9, 998. [Google Scholar] [CrossRef]
  817. Shen, L.; Li, H.; Bin, S.; Li, P.; Chen, J.; Gu, H.; Yuan, W. The Efficacy of Third Generation anti-HER2 Chimeric Antigen Receptor T Cells in Combination with PD1 Blockade against Malignant Glioblastoma Cells. Oncol. Rep. 2019, 42, 1549–1557. [Google Scholar] [CrossRef]
  818. Maggs, L.; Cattaneo, G.; Dal, A.E.; Moghaddam, A.S.; Ferrone, S. CAR T Cell-Based Immunotherapy for the Treatment of Glioblastoma. Front. Neurosci. 2021, 15, 662064. [Google Scholar] [CrossRef] [PubMed]
  819. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of Natural Killer Cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef]
  820. Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef]
  821. Zhou, X.; Yu, J.; Cheng, X.; Zhao, B.; Manyam, G.C.; Zhang, L.; Schluns, K.; Li, P.; Wang, J.; Sun, S.-C. The Deubiquitinase Otub1 Controls the Activation of CD8+ T Cells and NK Cells by Regulating IL-15-Mediated Priming. Nat. Immunol. 2019, 20, 879–889. [Google Scholar] [CrossRef] [PubMed]
  822. Grabowski, M.M.; Sankey, E.W.; Ryan, K.J.; Chongsathidkiet, P.; Lorrey, S.J.; Wilkinson, D.S.; Fecci, P.E. Immune Suppression in Gliomas. J. Neurooncol. 2021, 151, 3–12. [Google Scholar] [CrossRef] [PubMed]
  823. Morvan, M.G.; Lanier, L.L. NK Cells and Cancer: You Can Teach Innate Cells New Tricks. Nat. Rev. Cancer 2016, 16, 7–19. [Google Scholar] [CrossRef]
  824. Guillerey, C.; Huntington, N.D.; Smyth, M.J. Targeting Natural Killer Cells in Cancer Immunotherapy. Nat. Immunol. 2016, 17, 1025–1036. [Google Scholar] [CrossRef]
  825. Morimoto, T.; Nakazawa, T.; Maeoka, R.; Nakagawa, I.; Tsujimura, T.; Matsuda, R. Natural Killer Cell-Based Immunotherapy against Glioblastoma. Int. J. Mol. Sci. 2023, 24, 2111. [Google Scholar] [CrossRef]
  826. Lamers-Kok, N.; Panella, D.; Georgoudaki, A.-M.; Liu, H.; Özkazanc, D.; Kučerová, L.; Duru, A.D.; Spanholtz, J.; Raimo, M. Natural Killer Cells in Clinical Development as Non-Engineered, Engineered, and Combination Therapies. J. Hematol. Oncol. 2022, 15, 164. [Google Scholar] [CrossRef]
  827. Balatsoukas, A.; Rossignoli, F.; Shah, K. NK Cells in the Brain: Implications for Brain Tumor Development and Therapy. Trends Mol. Med. 2022, 28, 194–209. [Google Scholar] [CrossRef] [PubMed]
  828. Dongpo, S.; Zhengyao, Z.; Xiaozhuo, L.; Qing, W.; Mingming, F.; Fengqun, M.; Mei, L.; Qian, H.; Tong, C. Efficacy and Safety of Bevacizumab Combined with Other Therapeutic Regimens for Treatment of Recurrent Glioblastoma: A Network Meta-Analysis. World Neurosurg. 2022, 160, e61–e79. [Google Scholar] [CrossRef] [PubMed]
  829. Datsi, A.; Sorg, R.V. Dendritic Cell Vaccination of Glioblastoma: Road to Success or Dead End. Front. Immunol. 2021, 12, 770390. [Google Scholar] [CrossRef] [PubMed]
  830. Chen, M.; Sun, R.; Shi, B.; Wang, Y.; Di, S.; Luo, H.; Sun, Y.; Li, Z.; Zhou, M.; Jiang, H. Antitumor Efficacy of Chimeric Antigen Receptor T Cells against EGFRvIII-Expressing Glioblastoma in C57BL/6 Mice. Biomed. Pharmacother. 2019, 113, 108734. [Google Scholar] [CrossRef] [PubMed]
  831. O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette, J.J.D.; Martinez-Lage, M.; Brem, S.; Maloney, E.; Shen, A.; et al. A Single Dose of Peripherally Infused EGFRvIII-Directed CAR T Cells Mediates Antigen Loss and Induces Adaptive Resistance in Patients with Recurrent Glioblastoma. Sci. Transl. Med. 2017, 9, eaaa0984. [Google Scholar] [CrossRef] [PubMed]
  832. Suryadevara, C.M.; Desai, R.; Abel, M.L.; Riccione, K.A.; Batich, K.A.; Shen, S.H.; Chongsathidkiet, P.; Gedeon, P.C.; Elsamadicy, A.A.; Snyder, D.J.; et al. Temozolomide Lymphodepletion Enhances CAR Abundance and Correlates with Antitumor Efficacy against Established Glioblastoma. Oncoimmunology 2018, 7, e1434464. [Google Scholar] [CrossRef]
  833. Schmidts, A.; Srivastava, A.A.; Ramapriyan, R.; Bailey, S.R.; Bouffard, A.A.; Cahill, D.P.; Carter, B.S.; Curry, W.T.; Dunn, G.P.; Frigault, M.J.; et al. Tandem Chimeric Antigen Receptor (CAR) T Cells Targeting EGFRvIII and IL-13Rα2 Are Effective against Heterogeneous Glioblastoma. Neuro-Oncology Adv. 2023, 5, vdac185. [Google Scholar] [CrossRef] [PubMed]
  834. Yin, Y.; Rodriguez, J.L.; Li, N.; Thokala, R.; Nasrallah, M.P.; Hu, L.; Zhang, L.; Zhang, J.V.; Logun, M.T.; Kainth, D.; et al. Locally Secreted BiTEs Complement CAR T Cells by Enhancing Killing of Antigen Heterogeneous Solid Tumors. Mol. Ther. 2022, 30, 2537–2553. [Google Scholar] [CrossRef] [PubMed]
  835. Wollmann, G.; Ozduman, K.; Van Den Pol, A.N. Oncolytic Virus Therapy for Glioblastoma Multiforme: Concepts and Candidates. Cancer J. 2012, 18, 69–81. [Google Scholar] [CrossRef]
  836. Hamad, A.; Yusubalieva, G.M.; Baklaushev, V.P.; Chumakov, P.M.; Lipatova, A.V. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023, 15, 547. [Google Scholar] [CrossRef]
  837. Haddad, A.F.; Young, J.S.; Aghi, M.K. Using Viral Vectors to Deliver Local Immunotherapy to Glioblastoma. Neurosurg. Focus 2021, 50, E4. [Google Scholar] [CrossRef]
  838. Bartlett, D.L.; Liu, Z.; Sathaiah, M.; Ravindranathan, R.; Guo, Z.; He, Y.; Guo, Z.S. Oncolytic Viruses as Therapeutic Cancer Vaccines. Mol. Cancer 2013, 12, 103. [Google Scholar] [CrossRef]
  839. Kaufman, H.L.; Kohlhapp, F.J.; Zloza, A. Oncolytic Viruses: A New Class of Immunotherapy Drugs. Nat. Rev. Drug Discov. 2015, 14, 642–662. [Google Scholar] [CrossRef] [PubMed]
  840. de Graaf, J.F.; de Vor, L.; Fouchier, R.A.M.; van den Hoogen, B.G. Armed Oncolytic Viruses: A Kick-Start for Anti-Tumor Immunity. Cytokine Growth Factor. Rev. 2018, 41, 28–39. [Google Scholar] [CrossRef] [PubMed]
  841. Martikainen, M.; Essand, M. Virus-Based Immunotherapy of Glioblastoma. Cancers 2019, 11, 186. [Google Scholar] [CrossRef]
  842. Chon, H.J.; Lee, W.S.; Yang, H.; Kong, S.J.; Lee, N.K.; Moon, E.S.; Choi, J.; Han, E.C.; Kim, J.H.; Ahn, J.B.; et al. Tumor Microenvironment Remodeling by Intratumoral Oncolytic Vaccinia Virus Enhances the Efficacy of Immune-Checkpoint Blockade. Clin. Cancer Res. 2019, 25, 1612–1623. [Google Scholar] [CrossRef] [PubMed]
  843. Qi, Z.; Long, X.; Liu, J.; Cheng, P. Glioblastoma Microenvironment and Its Reprogramming by Oncolytic Virotherapy. Front. Cell Neurosci. 2022, 16, 819363. [Google Scholar] [CrossRef] [PubMed]
  844. Wood, H. Neuro-Oncology: A New Role for Zika Virus in Glioblastoma Therapy? Nat. Rev. Neurol. 2017, 13, 640–641. [Google Scholar] [CrossRef]
  845. Fan, X.; Lu, H.; Cui, Y.; Hou, X.; Huang, C.; Liu, G. Overexpression of P53 Delivered Using Recombinant NDV Induces Apoptosis in Glioma Cells by Regulating the Apoptotic Signaling Pathway. Exp. Ther. Med. 2018, 15, 4522–4530. [Google Scholar] [CrossRef]
  846. Hardcastle, J.; Kurozumi, K.; Dmitrieva, N.; Sayers, M.P.; Ahmad, S.; Waterman, P.; Weissleder, R.; Chiocca, E.A.; Kaur, B. Enhanced Antitumor Efficacy of Vasculostatin (Vstat120) Expressing Oncolytic HSV-1. Mol. Ther. 2010, 18, 285–294. [Google Scholar] [CrossRef]
  847. Loskog, A. Immunostimulatory Gene Therapy Using Oncolytic Viruses as Vehicles. Viruses 2015, 7, 5780–5791. [Google Scholar] [CrossRef]
  848. Sánchez-Hernández, L.; Hernández-Soto, J.; Vergara, P.; González, R.O.; Segovia, J. Additive Effects of the Combined Expression of Soluble Forms of GAS1 and PTEN Inhibiting Glioblastoma Growth. Gene Ther. 2018, 25, 439–449. [Google Scholar] [CrossRef]
  849. Xing, F.; Xiao, J.; Wu, J.; Liang, J.; Lu, X.; Guo, L.; Li, P.; Hou, P.; Li, C.; Guo, D. Modulating the Tumor Microenvironment via Oncolytic Virus and PI3K Inhibition Synergistically Restores Immune Checkpoint Therapy Response in PTEN-Deficient Glioblastoma. Signal Transduct. Target. Ther. 2021, 6, 275. [Google Scholar] [CrossRef]
  850. Kaufman, H.L.; Kim, D.W.; DeRaffele, G.; Mitcham, J.; Coffin, R.S.; Kim-Schulze, S. Local and Distant Immunity Induced by Intralesional Vaccination with an Oncolytic Herpes Virus Encoding GM-CSF in Patients with Stage IIIc and IV Melanoma. Ann. Surg. Oncol. 2010, 17, 718–730. [Google Scholar] [CrossRef] [PubMed]
  851. Zadeh, G.; Lang, F.; Daras, M.; Cloughesy, T.; Colman, H.; Ong, S.; Ramakrishna, R.; Vogelbaum, M.; Groves, M.; Nassiri, F.; et al. ATIM-24. INTERIM RESULTS OF A PHASE II MULTICENTER STUDY OF THE CONDITIONALLY REPLICATIVE ONCOLYTIC ADENOVIRUS DNX-2401 WITH PEMBROLIZUMAB (KEYTRUDA) FOR RECURRENT GLIOBLASTOMA; CAPTIVE STUDY (KEYNOTE-192). Neuro-Oncology 2018, 20, vi6. [Google Scholar] [CrossRef]
  852. Mehta, A.M.; Sonabend, A.M.; Bruce, J.N. Convection-Enhanced Delivery. Neurotherapeutics 2017, 14, 358–371. [Google Scholar] [CrossRef] [PubMed]
  853. Desjardins, A.; Gromeier, M.; Herndon, J.E.; Beaubier, N.; Bolognesi, D.P.; Friedman, A.H.; Friedman, H.S.; McSherry, F.; Muscat, A.M.; Nair, S.; et al. Recurrent Glioblastoma Treated with Recombinant Poliovirus. N. Engl. J. Med. 2018, 379, 150–161. [Google Scholar] [CrossRef] [PubMed]
  854. Kong, C.; Chang, W.S. Preclinical Research on Focused Ultrasound-Mediated Blood–Brain Barrier Opening for Neurological Disorders: A Review. Neurol. Int. 2023, 15, 285–300. [Google Scholar] [CrossRef]
  855. D’Amico, R.S.; Khatri, D.; Reichman, N.; Patel, N.V.; Wong, T.; Fralin, S.R.; Li, M.; Ellis, J.A.; Ortiz, R.; Langer, D.J.; et al. Super Selective Intra-Arterial Cerebral Infusion of Modern Chemotherapeutics after Blood-Brain Barrier Disruption: Where Are We Now, and Where We Are Going. J. Neurooncol. 2020, 147, 261–278. [Google Scholar] [CrossRef] [PubMed]
  856. Stavrakaki, E.; Dirven, C.M.F.; Lamfers, M.L.M. Personalizing Oncolytic Virotherapy for Glioblastoma: In Search of Biomarkers for Response. Cancers 2021, 13, 614. [Google Scholar] [CrossRef] [PubMed]
  857. Webb, M.J.; Sener, U.; Vile, R.G. Current Status and Challenges of Oncolytic Virotherapy for the Treatment of Glioblastoma. Pharmaceuticals 2023, 16, 793. [Google Scholar] [CrossRef]
  858. Muthukutty, P.; Yoo, S.Y. Oncolytic Virus Engineering and Utilizations: Cancer Immunotherapy Perspective. Viruses 2023, 15, 1645. [Google Scholar] [CrossRef] [PubMed]
  859. Chiocca, E.A.; Abbed, K.M.; Tatter, S.; Louis, D.N.; Hochberg, F.H.; Barker, F.; Kracher, J.; Grossman, S.A.; Fisher, J.D.; Carson, K.; et al. A Phase I Open-Label, Dose-Escalation, Multi-Institutional Trial of Injection with an E1B-Attenuated Adenovirus, ONYX-015, into the Peritumoral Region of Recurrent Malignant Gliomas, in the Adjuvant Setting. Mol. Ther. 2004, 10, 958–966. [Google Scholar] [CrossRef]
  860. Lang, F.F.; Tran, N.D.; Puduvalli, V.K.; Elder, J.B.; Fink, K.L.; Conrad, C.A.; Yung, W.K.A.; Penas-Prado, M.; Gomez-Manzano, C.; Peterkin, J.; et al. Phase 1b Open-Label Randomized Study of the Oncolytic Adenovirus DNX-2401 Administered with or without Interferon Gamma for Recurrent Glioblastoma. JCO 2017, 35, 2002. [Google Scholar] [CrossRef]
  861. Lang, F.F.; Conrad, C.; Gomez-Manzano, C.; Yung, W.K.A.; Sawaya, R.; Weinberg, J.S.; Prabhu, S.S.; Rao, G.; Fuller, G.N.; Aldape, K.D.; et al. Phase I Study of DNX-2401 (Delta-24-RGD) Oncolytic Adenovirus: Replication and Immunotherapeutic Effects in Recurrent Malignant Glioma. J. Clin. Oncol. 2018, 36, 1419–1427. [Google Scholar] [CrossRef]
  862. Markert, J.M.; Medlock, M.D.; Rabkin, S.D.; Gillespie, G.Y.; Todo, T.; Hunter, W.D.; Palmer, C.A.; Feigenbaum, F.; Tornatore, C.; Tufaro, F.; et al. Conditionally Replicating Herpes Simplex Virus Mutant, G207 for the Treatment of Malignant Glioma: Results of a Phase I Trial. Gene Ther. 2000, 7, 867–874. [Google Scholar] [CrossRef]
  863. Markert, J.M.; Liechty, P.G.; Wang, W.; Gaston, S.; Braz, E.; Karrasch, M.; Nabors, L.B.; Markiewicz, M.; Lakeman, A.D.; Palmer, C.A.; et al. Phase Ib Trial of Mutant Herpes Simplex Virus G207 Inoculated Pre-and Post-Tumor Resection for Recurrent GBM. Mol. Ther. 2009, 17, 199–207. [Google Scholar] [CrossRef]
  864. Markert, J.M.; Razdan, S.N.; Kuo, H.-C.; Cantor, A.; Knoll, A.; Karrasch, M.; Nabors, L.B.; Markiewicz, M.; Agee, B.S.; Coleman, J.M.; et al. A Phase 1 Trial of Oncolytic HSV-1, G207, Given in Combination With Radiation for Recurrent GBM Demonstrates Safety and Radiographic Responses. Mol. Ther. 2014, 22, 1048–1055. [Google Scholar] [CrossRef]
  865. Kicielinski, K.P.; Chiocca, E.A.; Yu, J.S.; Gill, G.M.; Coffey, M.; Markert, J.M. Phase 1 Clinical Trial of Intratumoral Reovirus Infusion for the Treatment of Recurrent Malignant Gliomas in Adults. Mol. Ther. 2014, 22, 1056–1062. [Google Scholar] [CrossRef] [PubMed]
  866. Accomando, W.P.; Rao, A.R.; Hogan, D.J.; Newman, A.M.; Nakao, A.; Alizadeh, A.A.; Diehn, M.; Diago, O.R.; Gammon, D.; Haghighi, A.; et al. Molecular and Immunologic Signatures Are Related to Clinical Benefit from Treatment with Vocimagene Amiretrorepvec (Toca 511) and 5-Fluorocytosine (Toca FC) in Patients with Glioma. Clin. Cancer Res. 2020, 26, 6176–6186. [Google Scholar] [CrossRef]
  867. Geletneky, K.; Hajda, J.; Angelova, A.L.; Leuchs, B.; Capper, D.; Bartsch, A.J.; Neumann, J.-O.; Schöning, T.; Hüsing, J.; Beelte, B.; et al. Oncolytic H-1 Parvovirus Shows Safety and Signs of Immunogenic Activity in a First Phase I/IIa Glioblastoma Trial. Mol. Ther. 2017, 25, 2620–2634. [Google Scholar] [CrossRef]
  868. Allen, C.; Paraskevakou, G.; Iankov, I.; Giannini, C.; Schroeder, M.; Sarkaria, J.; Puri, R.K.; Russell, S.J.; Galanis, E. Interleukin-13 Displaying Retargeted Oncolytic Measles Virus Strains Have Significant Activity Against Gliomas With Improved Specificity. Mol. Ther. 2008, 16, 1556–1564. [Google Scholar] [CrossRef]
  869. Allen, C.; Opyrchal, M.; Aderca, I.; Schroeder, M.A.; Sarkaria, J.N.; Domingo, E.; Federspiel, M.J.; Galanis, E. Oncolytic Measles Virus Strains Have Significant Antitumor Activity against Glioma Stem Cells. Gene Ther. 2013, 20, 444–449. [Google Scholar] [CrossRef]
  870. Kurokawa, C.; Iankov, I.D.; Anderson, S.K.; Aderca, I.; Leontovich, A.A.; Maurer, M.J.; Oberg, A.L.; Schroeder, M.A.; Giannini, C.; Greiner, S.M.; et al. Constitutive Interferon Pathway Activation in Tumors as an Efficacy Determinant Following Oncolytic Virotherapy. J. Natl. Cancer Inst. 2018, 110, 1123–1132. [Google Scholar] [CrossRef]
  871. Gromeier, M.; Lachmann, S.; Rosenfeld, M.R.; Gutin, P.H.; Wimmer, E. Intergeneric Poliovirus Recombinants for the Treatment of Malignant Glioma. Proc. Natl. Acad. Sci. USA 2000, 97, 6803–6808. [Google Scholar] [CrossRef] [PubMed]
  872. Foloppe, J.; Kempf, J.; Futin, N.; Kintz, J.; Cordier, P.; Pichon, C.; Findeli, A.; Vorburger, F.; Quemeneur, E.; Erbs, P. The Enhanced Tumor Specificity of TG6002, an Armed Oncolytic Vaccinia Virus Deleted in Two Genes Involved in Nucleotide Metabolism. Mol. Ther. Oncolytics 2019, 14, 1–14. [Google Scholar] [CrossRef]
  873. Freeman, A.I.; Zakay-Rones, Z.; Gomori, J.M.; Linetsky, E.; Rasooly, L.; Greenbaum, E.; Rozenman-Yair, S.; Panet, A.; Libson, E.; Irving, C.S.; et al. Phase I/II Trial of Intravenous NDV-HUJ Oncolytic Virus in Recurrent Glioblastoma Multiforme. Mol. Ther. 2006, 13, 221–228. [Google Scholar] [CrossRef]
  874. Rong, L.; Li, N.; Zhang, Z. Emerging Therapies for Glioblastoma: Current State and Future Directions. J. Exp. Clin. Cancer Res. 2022, 41, 142. [Google Scholar] [CrossRef]
  875. Lim, M.; Xia, Y.; Bettegowda, C.; Weller, M. Current State of Immunotherapy for Glioblastoma. Nat. Rev. Clin. Oncol. 2018, 15, 422–442. [Google Scholar] [CrossRef]
  876. Sloan, A.E.; Buerki, R.A.; Murphy, C.; Kelly, A.T.; Ambady, P.; Brown, M.; Butowski, N.A.; Cavaliere, R.; Curry, W.T.; Desjardins, A.; et al. LUMINOS-101: Phase 2 Study of PVSRIPO with Pembrolizumab in Recurrent Glioblastoma. JCO 2021, 39, TPS2065. [Google Scholar] [CrossRef]
  877. Chiocca, E.A.; Nassiri, F.; Wang, J.; Peruzzi, P.; Zadeh, G. Viral and Other Therapies for Recurrent Glioblastoma: Is a 24-Month Durable Response Unusual? Neuro Oncol. 2019, 21, 14–25. [Google Scholar] [CrossRef]
  878. Uchida, H.; Marzulli, M.; Nakano, K.; Goins, W.F.; Chan, J.; Hong, C.-S.; Mazzacurati, L.; Yoo, J.Y.; Haseley, A.; Nakashima, H.; et al. Effective Treatment of an Orthotopic Xenograft Model of Human Glioblastoma Using an EGFR-Retargeted Oncolytic Herpes Simplex Virus. Mol. Ther. 2013, 21, 561–569. [Google Scholar] [CrossRef]
  879. Saha, D.; Wakimoto, H.; Peters, C.W.; Antoszczyk, S.J.; Rabkin, S.D.; Martuza, R.L. Combinatorial Effects of VEGFR Kinase Inhibitor Axitinib and Oncolytic Virotherapy in Mouse and Human Glioblastoma Stem-Like Cell Models. Clin. Cancer Res. 2018, 24, 3409–3422. [Google Scholar] [CrossRef]
  880. Spain, L.; Diem, S.; Larkin, J. Management of Toxicities of Immune Checkpoint Inhibitors. Cancer Treat. Rev. 2016, 44, 51–60. [Google Scholar] [CrossRef]
  881. Yang, M.; Oh, I.Y.; Mahanty, A.; Jin, W.-L.; Yoo, J.S. Immunotherapy for Glioblastoma: Current State, Challenges, and Future Perspectives. Cancers 2020, 12, 2334. [Google Scholar] [CrossRef] [PubMed]
  882. Morgan, R.A.; Yang, J.C.; Kitano, M.; Dudley, M.E.; Laurencot, C.M.; Rosenberg, S.A. Case Report of a Serious Adverse Event Following the Administration of T Cells Transduced With a Chimeric Antigen Receptor Recognizing ERBB2. Mol. Ther. 2010, 18, 843–851. [Google Scholar] [CrossRef] [PubMed]
  883. Bedoya, F.; Frigault, M.J.; Maus, M.V. The Flipside of the Power of Engineered T Cells: Observed and Potential Toxicities of Genetically Modified T Cells as Therapy. Mol. Ther. 2017, 25, 314–320. [Google Scholar] [CrossRef]
  884. Nam, L.; Coll, C.; Erthal, L.C.S.; de la Torre, C.; Serrano, D.; Martínez-Máñez, R.; Santos-Martínez, M.J.; Ruiz-Hernández, E. Drug Delivery Nanosystems for the Localized Treatment of Glioblastoma Multiforme. Materials 2018, 11, 779. [Google Scholar] [CrossRef] [PubMed]
  885. Di Filippo, L.D.; Duarte, J.L.; Luiz, M.T.; de Araújo, J.T.C.; Chorilli, M. Drug Delivery Nanosystems in Glioblastoma Multiforme Treatment: Current State of the Art. Curr. Neuropharmacol. 2021, 19, 787–812. [Google Scholar] [CrossRef]
  886. Hsu, J.-F.; Chu, S.-M.; Liao, C.-C.; Wang, C.-J.; Wang, Y.-S.; Lai, M.-Y.; Wang, H.-C.; Huang, H.-R.; Tsai, M.-H. Nanotechnology and Nanocarrier-Based Drug Delivery as the Potential Therapeutic Strategy for Glioblastoma Multiforme: An Update. Cancers 2021, 13, 195. [Google Scholar] [CrossRef]
  887. Marei, H.E. Multimodal Targeting of Glioma with Functionalized Nanoparticles. Cancer Cell Int. 2022, 22, 265. [Google Scholar] [CrossRef] [PubMed]
  888. Wadajkar, A.S.; Dancy, J.G.; Hersh, D.S.; Anastasiadis, P.; Tran, N.L.; Woodworth, G.F.; Winkles, J.A.; Kim, A.J. Tumor-Targeted Nanotherapeutics: Overcoming Treatment Barriers for Glioblastoma. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  889. Alphandéry, E. Nano-Therapies for Glioblastoma Treatment. Cancers 2020, 12, 242. [Google Scholar] [CrossRef] [PubMed]
  890. Rosenblum, D.; Joshi, N.; Tao, W.; Karp, J.M.; Peer, D. Progress and Challenges towards Targeted Delivery of Cancer Therapeutics. Nat. Commun. 2018, 9, 1410. [Google Scholar] [CrossRef] [PubMed]
  891. Qiu, Z.; Yu, Z.; Xu, T.; Wang, L.; Meng, N.; Jin, H.; Xu, B. Novel Nano-Drug Delivery System for Brain Tumor Treatment. Cells 2022, 11, 3761. [Google Scholar] [CrossRef]
  892. Wei, D.; Zhang, N.; Qu, S.; Wang, H.; Li, J. Advances in Nanotechnology for the Treatment of GBM. Front. Neurosci. 2023, 17. [Google Scholar] [CrossRef]
  893. Tang, L.; Feng, Y.; Gao, S.; Mu, Q.; Liu, C. Nanotherapeutics Overcoming the Blood-Brain Barrier for Glioblastoma Treatment. Front. Pharmacol. 2021, 12. [Google Scholar] [CrossRef]
  894. Aravind, A.; Veeranarayanan, S.; Poulose, A.C.; Nair, R.; Nagaoka, Y.; Yoshida, Y.; Maekawa, T.; Kumar, D.S. Aptamer-Functionalized Silica Nanoparticles for Targeted Cancer Therapy. BioNanoSci. 2012, 2, 1–8. [Google Scholar] [CrossRef]
  895. Kuo, Y.-C.; Lee, C.-H. Dual Targeting of Solid Lipid Nanoparticles Grafted with 83-14 MAb and Anti-EGF Receptor for Malignant Brain Tumor Therapy. Life Sci. 2016, 146, 222–231. [Google Scholar] [CrossRef] [PubMed]
  896. Song, P.; Zhao, X.; Xiao, S. Application Prospect of Peptide-Modified Nano Targeting Drug Delivery System Combined with PD-1/PD-L1 Based Immune Checkpoint Blockade in Glioblastoma. Int. J. Pharm. 2020, 589, 119865. [Google Scholar] [CrossRef]
  897. Fang, C.; Wang, K.; Stephen, Z.R.; Mu, Q.; Kievit, F.M.; Chiu, D.T.; Press, O.W.; Zhang, M. Temozolomide Nanoparticles for Targeted Glioblastoma Therapy. ACS Appl. Mater. Interfaces 2015, 7, 6674–6682. [Google Scholar] [CrossRef]
  898. Gonawala, S.; Ali, M.M. Application of Dendrimer-Based Nanoparticles in Glioma Imaging. J. Nanomed. Nanotechnol. 2017, 8, 444. [Google Scholar] [CrossRef] [PubMed]
  899. Ruiz-Molina, D.; Mao, X.; Alfonso-Triguero, P.; Lorenzo, J.; Bruna, J.; Yuste, V.J.; Candiota, A.P.; Novio, F. Advances in Preclinical/Clinical Glioblastoma Treatment: Can Nanoparticles Be of Help? Cancers 2022, 14, 4960. [Google Scholar] [CrossRef] [PubMed]
  900. Neuwelt, E.A.; Várallyay, P.; Bagó, A.G.; Muldoon, L.L.; Nesbit, G.; Nixon, R. Imaging of Iron Oxide Nanoparticles by MR and Light Microscopy in Patients with Malignant Brain Tumours. Neuropathol. Appl. Neurobiol. 2004, 30, 456–471. [Google Scholar] [CrossRef] [PubMed]
  901. Taschner, C.A.; Wetzel, S.G.; Tolnay, M.; Froehlich, J.; Merlo, A.; Radue, E.W. Characteristics of Ultrasmall Superparamagnetic Iron Oxides in Patients with Brain Tumors. AJR Am. J. Roentgenol. 2005, 185, 1477–1486. [Google Scholar] [CrossRef]
  902. Liu, H.; Zhang, J.; Chen, X.; Du, X.-S.; Zhang, J.-L.; Liu, G.; Zhang, W.-G. Application of Iron Oxide Nanoparticles in Glioma Imaging and Therapy: From Bench to Bedside. Nanoscale 2016, 8, 7808–7826. [Google Scholar] [CrossRef]
  903. Gleich, B.; Weizenecker, J. Tomographic Imaging Using the Nonlinear Response of Magnetic Particles. Nature 2005, 435, 1214–1217. [Google Scholar] [CrossRef]
  904. Chan, M.-H.; Chen, W.; Li, C.-H.; Fang, C.-Y.; Chang, Y.-C.; Wei, D.-H.; Liu, R.-S.; Hsiao, M. An Advanced In Situ Magnetic Resonance Imaging and Ultrasonic Theranostics Nanocomposite Platform: Crossing the Blood-Brain Barrier and Improving the Suppression of Glioblastoma Using Iron-Platinum Nanoparticles in Nanobubbles. ACS Appl. Mater. Interfaces 2021, 13, 26759–26769. [Google Scholar] [CrossRef]
  905. Desroches, J.; Jermyn, M.; Pinto, M.; Picot, F.; Tremblay, M.-A.; Obaid, S.; Marple, E.; Urmey, K.; Trudel, D.; Soulez, G.; et al. A New Method Using Raman Spectroscopy for in Vivo Targeted Brain Cancer Tissue Biopsy. Sci. Rep. 2018, 8, 1792. [Google Scholar] [CrossRef]
  906. Schlücker, S. Surface-Enhanced Raman Spectroscopy: Concepts and Chemical Applications. Angew. Chem. Int. Ed. 2014, 53, 4756–4795. [Google Scholar] [CrossRef]
  907. Jatyan, R.; Singh, P.; Sahel, D.K.; Karthik, Y.G.; Mittal, A.; Chitkara, D. Polymeric and Small Molecule-Conjugates of Temozolomide as Improved Therapeutic Agents for Glioblastoma Multiforme. J. Control. Release 2022, 350, 494–513. [Google Scholar] [CrossRef]
  908. Iturrioz-Rodríguez, N.; Sampron, N.; Matheu, A. Current Advances in Temozolomide Encapsulation for the Enhancement of Glioblastoma Treatment. Theranostics 2023, 13, 2734–2756. [Google Scholar] [CrossRef]
  909. Yang, J.; Xu, Y.; Fu, Z.; Chen, J.; Fan, W.; Wu, X. Progress in Research and Development of Temozolomide Brain-Targeted Preparations: A Review. J. Drug Target. 2023, 31, 119–133. [Google Scholar] [CrossRef]
  910. Sharma, A.K.; Gupta, L.; Sahu, H.; Qayum, A.; Singh, S.K.; Nakhate, K.T.; Ajazuddin; Gupta, U. Chitosan Engineered PAMAM Dendrimers as Nanoconstructs for the Enhanced Anti-Cancer Potential and Improved In Vivo Brain Pharmacokinetics of Temozolomide. Pharm. Res. 2018, 35, 9. [Google Scholar] [CrossRef] [PubMed]
  911. Sharma, A.; Liaw, K.; Sharma, R.; Spriggs, T.; Appiani La Rosa, S.; Kannan, S.; Kannan, R.M. Dendrimer-Mediated Targeted Delivery of Rapamycin to Tumor-Associated Macrophages Improves Systemic Treatment of Glioblastoma. Biomacromolecules 2020, 21, 5148–5161. [Google Scholar] [CrossRef] [PubMed]
  912. Hu, J.; Wang, J.; Wang, G.; Yao, Z.; Dang, X. Pharmacokinetics and Antitumor Efficacy of DSPE-PEG2000 Polymeric Liposomes Loaded with Quercetin and Temozolomide: Analysis of Their Effectiveness in Enhancing the Chemosensitization of Drug-Resistant Glioma Cells. Int. J. Mol. Med. 2016, 37, 690–702. [Google Scholar] [CrossRef] [PubMed]
  913. Wang, F.; Ye, X.; Wu, Y.; Wang, H.; Sheng, C.; Peng, D.; Chen, W. Time Interval of Two Injections and First-Dose Dependent of Accelerated Blood Clearance Phenomenon Induced by PEGylated Liposomal Gambogenic Acid: The Contribution of PEG-Specific IgM. J. Pharm. Sci. 2019, 108, 641–651. [Google Scholar] [CrossRef] [PubMed]
  914. Huang, G.; Zhang, N.; Bi, X.; Dou, M. Solid Lipid Nanoparticles of Temozolomide: Potential Reduction of Cardial and Nephric Toxicity. Int. J. Pharm. 2008, 355, 314–320. [Google Scholar] [CrossRef] [PubMed]
  915. Di Martino, A.; Sedlarik, V. Amphiphilic Chitosan-Grafted-Functionalized Polylactic Acid Based Nanoparticles as a Delivery System for Doxorubicin and Temozolomide Co-Therapy. Int. J. Pharm. 2014, 474, 134–145. [Google Scholar] [CrossRef] [PubMed]
  916. Di Martino, A.; Pavelkova, A.; Maciulyte, S.; Budriene, S.; Sedlarik, V. Polysaccharide-Based Nanocomplexes for Co-Encapsulation and Controlled Release of 5-Fluorouracil and Temozolomide. Eur. J. Pharm. Sci. 2016, 92, 276–286. [Google Scholar] [CrossRef] [PubMed]
  917. Behrooz, A.B.; Vazifehmand, R.; Tajudin, A.A.; Masarudin, M.J.; Sekawi, Z.; Masomian, M.; Syahir, A. Tailoring Drug Co-Delivery Nanosystem for Mitigating U-87 Stem Cells Drug Resistance. Drug Deliv. Transl. Res. 2022, 12, 1253–1269. [Google Scholar] [CrossRef]
  918. Gherardini, L.; Vetri Buratti, V.; Maturi, M.; Inzalaco, G.; Locatelli, E.; Sambri, L.; Gargiulo, S.; Barone, V.; Bonente, D.; Bertelli, E.; et al. Loco-Regional Treatment with Temozolomide-Loaded Thermogels Prevents Glioblastoma Recurrences in Orthotopic Human Xenograft Models. Sci. Rep. 2023, 13, 4630. [Google Scholar] [CrossRef] [PubMed]
  919. Ramalho, M.J.; Loureiro, J.A.; Coelho, M.A.N.; Pereira, M.C. Transferrin Receptor-Targeted Nanocarriers: Overcoming Barriers to Treat Glioblastoma. Pharmaceutics 2022, 14, 279. [Google Scholar] [CrossRef] [PubMed]
  920. Kim, S.-S.; Rait, A.; Kim, E.; DeMarco, J.; Pirollo, K.F.; Chang, E.H. Encapsulation of Temozolomide in a Tumor-Targeting Nanocomplex Enhances Anti-Cancer Efficacy and Reduces Toxicity in a Mouse Model of Glioblastoma. Cancer Lett. 2015, 369, 250–258. [Google Scholar] [CrossRef] [PubMed]
  921. Helal, D.O.; Rouatbi, N.; Han, S.; Tzu-Wen Wang, J.; Walters, A.A.; Abdel-Mottaleb, M.M.A.; Kamel, A.O.; Geneidi, A.-S.; Awad, G.A.S.; Al-Jamal, K.T. A Natural Protein Based Platform for the Delivery of Temozolomide Acid to Glioma Cells. Eur. J. Pharm. Biopharm. 2021, 169, 297–308. [Google Scholar] [CrossRef] [PubMed]
  922. Ju, R.-J.; Zeng, F.; Liu, L.; Mu, L.-M.; Xie, H.-J.; Zhao, Y.; Yan, Y.; Wu, J.-S.; Hu, Y.-J.; Lu, W.-L. Destruction of Vasculogenic Mimicry Channels by Targeting Epirubicin plus Celecoxib Liposomes in Treatment of Brain Glioma. Int. J. Nanomed. 2016, 11, 1131–1146. [Google Scholar] [CrossRef]
  923. Tarasov, V.V.; Svistunov, A.A.; Chubarev, V.N.; Zatsepilova, T.A.; Preferanskaya, N.G.; Stepanova, O.I.; Sokolov, A.V.; Dostdar, S.A.; Minyaeva, N.N.; Neganova, M.E.; et al. Feasibility of Targeting Glioblastoma Stem Cells: From Concept to Clinical Trials. Curr. Top. Med. Chem. 2019, 19, 2974–2984. [Google Scholar] [CrossRef]
  924. Cho, J.-H.; Kim, A.-R.; Kim, S.-H.; Lee, S.-J.; Chung, H.; Yoon, M.-Y. Development of a Novel Imaging Agent Using Peptide-Coated Gold Nanoparticles toward Brain Glioma Stem Cell Marker CD133. Acta Biomater. 2017, 47, 182–192. [Google Scholar] [CrossRef]
  925. Kunoh, T.; Shimura, T.; Kasai, T.; Matsumoto, S.; Mahmud, H.; Khayrani, A.C.; Seno, M.; Kunoh, H.; Takada, J. Use of DNA-Generated Gold Nanoparticles to Radiosensitize and Eradicate Radioresistant Glioma Stem Cells. Nanotechnology 2019, 30, 055101. [Google Scholar] [CrossRef]
  926. Lakkadwala, S.; Dos Santos Rodrigues, B.; Sun, C.; Singh, J. Dual Functionalized Liposomes for Efficient Co-Delivery of Anti-Cancer Chemotherapeutics for the Treatment of Glioblastoma. J. Control Release 2019, 307, 247–260. [Google Scholar] [CrossRef] [PubMed]
  927. Kumar, N.; Khurana, B.; Arora, D. Nose-to-Brain Drug Delivery for the Treatment of Glioblastoma Multiforme: Nanotechnological Interventions. Pharm. Dev. Technol. 2023, 28, 1032–1047. [Google Scholar] [CrossRef]
  928. Sousa, F.; Dhaliwal, H.K.; Gattacceca, F.; Sarmento, B.; Amiji, M.M. Enhanced Anti-Angiogenic Effects of Bevacizumab in Glioblastoma Treatment upon Intranasal Administration in Polymeric Nanoparticles. J. Control Release 2019, 309, 37–47. [Google Scholar] [CrossRef] [PubMed]
  929. Bouras, A.; Kaluzova, M.; Hadjipanayis, C.G. Radiosensitivity Enhancement of Radioresistant Glioblastoma by Epidermal Growth Factor Receptor Antibody-Conjugated Iron-Oxide Nanoparticles. J. Neurooncol. 2015, 124, 13–22. [Google Scholar] [CrossRef] [PubMed]
  930. Liu, P.; Jin, H.; Guo, Z.; Ma, J.; Zhao, J.; Li, D.; Wu, H.; Gu, N. Silver Nanoparticles Outperform Gold Nanoparticles in Radiosensitizing U251 Cells in Vitro and in an Intracranial Mouse Model of Glioma. Int. J. Nanomed. 2016, 11, 5003–5014. [Google Scholar] [CrossRef]
  931. Ali, M.Y.; Oliva, C.R.; Noman, A.S.M.; Allen, B.G.; Goswami, P.C.; Zakharia, Y.; Monga, V.; Spitz, D.R.; Buatti, J.M.; Griguer, C.E. Radioresistance in Glioblastoma and the Development of Radiosensitizers. Cancers 2020, 12, 2511. [Google Scholar] [CrossRef] [PubMed]
  932. Liu, Z.; Tan, H.; Zhang, X.; Chen, F.; Zhou, Z.; Hu, X.; Chang, S.; Liu, P.; Zhang, H. Enhancement of Radiotherapy Efficacy by Silver Nanoparticles in Hypoxic Glioma Cells. Artif. Cells Nanomed. Biotechnol. 2018, 46, S922–S930. [Google Scholar] [CrossRef] [PubMed]
  933. Li, X.; Wu, C.; Chen, N.; Gu, H.; Yen, A.; Cao, L.; Wang, E.; Wang, L. PI3K/Akt/mTOR Signaling Pathway and Targeted Therapy for Glioblastoma. Oncotarget 2016, 7, 33440–33450. [Google Scholar] [CrossRef]
  934. Djuzenova, C.S.; Fiedler, V.; Memmel, S.; Katzer, A.; Sisario, D.; Brosch, P.K.; Göhrung, A.; Frister, S.; Zimmermann, H.; Flentje, M.; et al. Differential Effects of the Akt Inhibitor MK-2206 on Migration and Radiation Sensitivity of Glioblastoma Cells. BMC Cancer 2019, 19, 299. [Google Scholar] [CrossRef]
  935. Bindra, R.S.; Chalmers, A.J.; Evans, S.; Dewhirst, M. GBM Radiosensitizers: Dead in the Water…or Just the Beginning? J. Neurooncol. 2017, 134, 513–521. [Google Scholar] [CrossRef]
  936. Schneider, C.S.; Woodworth, G.F.; Vujaskovic, Z.; Mishra, M.V. Radiosensitization of High-Grade Gliomas through Induced Hyperthermia: Review of Clinical Experience and the Potential Role of MR-Guided Focused Ultrasound. Radiother. Oncol. 2020, 142, 43–51. [Google Scholar] [CrossRef]
  937. Grauer, O.; Jaber, M.; Hess, K.; Weckesser, M.; Schwindt, W.; Maring, S.; Wölfer, J.; Stummer, W. Combined Intracavitary Thermotherapy with Iron Oxide Nanoparticles and Radiotherapy as Local Treatment Modality in Recurrent Glioblastoma Patients. J. Neurooncol. 2019, 141, 83–94. [Google Scholar] [CrossRef] [PubMed]
  938. Koh, H.K.; Seo, S.Y.; Kim, J.H.; Kim, H.J.; Chie, E.K.; Kim, S.-K.; Kim, I.H. Disulfiram, a Re-Positioned Aldehyde Dehydrogenase Inhibitor, Enhances Radiosensitivity of Human Glioblastoma Cells In Vitro. Cancer Res. Treat. 2019, 51, 696–705. [Google Scholar] [CrossRef] [PubMed]
  939. Bhattarai, S.R.; Derry, P.J.; Aziz, K.; Singh, P.K.; Khoo, A.M.; Chadha, A.S.; Liopo, A.; Zubarev, E.R.; Krishnan, S. Gold Nanotriangles: Scale up and X-Ray Radiosensitization Effects in Mice. Nanoscale 2017, 9, 5085–5093. [Google Scholar] [CrossRef] [PubMed]
  940. Liu, Y.; Zhang, P.; Li, F.; Jin, X.; Li, J.; Chen, W.; Li, Q. Metal-Based NanoEnhancers for Future Radiotherapy: Radiosensitizing and Synergistic Effects on Tumor Cells. Theranostics 2018, 8, 1824–1849. [Google Scholar] [CrossRef] [PubMed]
  941. Kang, D.; Wang, J.; Zhang, W.; Song, Y.; Li, X.; Zou, Y.; Zhu, M.; Zhu, Z.; Chen, F.; Yang, C.J. Selection of DNA Aptamers against Glioblastoma Cells with High Affinity and Specificity. PLoS ONE 2012, 7, e42731. [Google Scholar] [CrossRef]
  942. Affinito, A.; Quintavalle, C.; Esposito, C.L.; Roscigno, G.; Vilardo, C.; Nuzzo, S.; Ricci-Vitiani, L.; De Luca, G.; Pallini, R.; Kichkailo, A.S.; et al. The Discovery of RNA Aptamers That Selectively Bind Glioblastoma Stem Cells. Mol. Ther. Nucleic Acids 2019, 18, 99–109. [Google Scholar] [CrossRef]
  943. Cesarini, V.; Scopa, C.; Silvestris, D.A.; Scafidi, A.; Petrera, V.; Del Baldo, G.; Gallo, A. Aptamer-Based In Vivo Therapeutic Targeting of Glioblastoma. Molecules 2020, 25, 4267. [Google Scholar] [CrossRef]
  944. Giles, B.; Nakhjavani, M.; Wiesa, A.; Knight, T.; Shigdar, S.; Samarasinghe, R.M. Unravelling the Glioblastoma Tumour Microenvironment: Can Aptamer Targeted Delivery Become Successful in Treating Brain Cancers? Cancers 2023, 15, 4376. [Google Scholar] [CrossRef]
  945. Giordano, F.A.; Layer, J.P.; Leonardelli, S.; Friker, L.L.; Turiello, R.; Corvino, D.; Zeyen, T.; Schaub, C.; Mueller, W.; Sperk, E.; et al. Potential Predictive Biomarker for Response to Radiotherapy and CXCL12 Inhibition in Glioblastoma in the Phase I/II GLORIA Trial. JCO 2023, 41, 2048. [Google Scholar] [CrossRef]
  946. Yu, D.; Khan, O.F.; Suvà, M.L.; Dong, B.; Panek, W.K.; Xiao, T.; Wu, M.; Han, Y.; Ahmed, A.U.; Balyasnikova, I.V.; et al. Multiplexed RNAi Therapy against Brain Tumor-Initiating Cells via Lipopolymeric Nanoparticle Infusion Delays Glioblastoma Progression. Proc. Natl. Acad. Sci. USA 2017, 114, E6147–E6156. [Google Scholar] [CrossRef]
  947. Lozada-Delgado, E.L.; Grafals-Ruiz, N.; Vivas-Mejía, P.E. RNA Interference for Glioblastoma Therapy: Innovation Ladder from the Bench to Clinical Trials. Life Sci. 2017, 188, 26–36. [Google Scholar] [CrossRef] [PubMed]
  948. Singh, P.; Singh, A.; Shah, S.; Vataliya, J.; Mittal, A.; Chitkara, D. RNA Interference Nanotherapeutics for Treatment of Glioblastoma Multiforme. Mol. Pharm. 2020, 17, 4040–4066. [Google Scholar] [CrossRef] [PubMed]
  949. Shetty, K.; Yasaswi, S.; Dutt, S.; Yadav, K.S. Multifunctional Nanocarriers for Delivering siRNA and miRNA in Glioblastoma Therapy: Advances in Nanobiotechnology-Based Cancer Therapy. 3 Biotech. 2022, 12, 301. [Google Scholar] [CrossRef] [PubMed]
  950. Wang, K.; Kievit, F.M.; Chiarelli, P.A.; Stephen, Z.R.; Lin, G.; Silber, J.R.; Ellenbogen, R.G.; Zhang, M. siRNA Nanoparticle Suppresses Drug-Resistant Gene and Prolongs Survival in an Orthotopic Glioblastoma Xenograft Mouse Model. Adv. Funct. Mater. 2021, 31, 2007166. [Google Scholar] [CrossRef]
  951. Al-Sammarraie, N.; Ray, S.K. Applications of CRISPR-Cas9 Technology to Genome Editing in Glioblastoma Multiforme. Cells 2021, 10, 2342. [Google Scholar] [CrossRef]
  952. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A Programmable Dual-RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef] [PubMed]
  953. Mali, P.; Yang, L.; Esvelt, K.M.; Aach, J.; Guell, M.; DiCarlo, J.E.; Norville, J.E.; Church, G.M. RNA-Guided Human Genome Engineering via Cas9. Science 2013, 339, 823–826. [Google Scholar] [CrossRef] [PubMed]
  954. Zhao, N.; Weng, S.; Liu, Z.; Xu, H.; Ren, Y.; Guo, C.; Liu, L.; Zhang, Z.; Ji, Y.; Han, X. CRISPR-Cas9 Identifies Growth-Related Subtypes of Glioblastoma with Therapeutical Significance through Cell Line Knockdown. BMC Cancer 2023, 23, 749. [Google Scholar] [CrossRef]
  955. Zuckermann, M.; Hovestadt, V.; Knobbe-Thomsen, C.B.; Zapatka, M.; Northcott, P.A.; Schramm, K.; Belic, J.; Jones, D.T.W.; Tschida, B.; Moriarity, B.; et al. Somatic CRISPR/Cas9-Mediated Tumour Suppressor Disruption Enables Versatile Brain Tumour Modelling. Nat. Commun. 2015, 6, 7391. [Google Scholar] [CrossRef] [PubMed]
  956. Jin, F.; Jin-Lee, H.J.; Johnson, A.J. Mouse Models of Experimental Glioblastoma. In Gliomas; Debinski, W., Ed.; Exon Publications: Brisbane, Australia, 2021; ISBN 978-0-645-00174-7. [Google Scholar]
  957. Luther, D.C.; Lee, Y.W.; Nagaraj, H.; Scaletti, F.; Rotello, V.M. Delivery Approaches for CRISPR/Cas9 Therapeutics In Vivo: Advances and Challenges. Expert Opin. Drug Deliv. 2018, 15, 905–913. [Google Scholar] [CrossRef] [PubMed]
  958. Rui, Y.; Wilson, D.R.; Green, J.J. Non-Viral Delivery To Enable Genome Editing. Trends Biotechnol. 2019, 37, 281–293. [Google Scholar] [CrossRef] [PubMed]
  959. Rosenblum, D.; Gutkin, A.; Kedmi, R.; Ramishetti, S.; Veiga, N.; Jacobi, A.M.; Schubert, M.S.; Friedmann-Morvinski, D.; Cohen, Z.R.; Behlke, M.A.; et al. CRISPR-Cas9 Genome Editing Using Targeted Lipid Nanoparticles for Cancer Therapy. Sci. Adv. 2020, 6, eabc9450. [Google Scholar] [CrossRef]
  960. Zou, Y.; Sun, X.; Yang, Q.; Zheng, M.; Shimoni, O.; Ruan, W.; Wang, Y.; Zhang, D.; Yin, J.; Huang, X.; et al. Blood-Brain Barrier-Penetrating Single CRISPR-Cas9 Nanocapsules for Effective and Safe Glioblastoma Gene Therapy. Sci. Adv. 2022, 8, eabm8011. [Google Scholar] [CrossRef] [PubMed]
  961. Li, L.; He, Z.-Y.; Wei, X.-W.; Gao, G.-P.; Wei, Y.-Q. Challenges in CRISPR/CAS9 Delivery: Potential Roles of Nonviral Vectors. Hum. Gene Ther. 2015, 26, 452–462. [Google Scholar] [CrossRef]
  962. Ruan, W.; Jiao, M.; Xu, S.; Ismail, M.; Xie, X.; An, Y.; Guo, H.; Qian, R.; Shi, B.; Zheng, M. Brain-Targeted CRISPR/Cas9 Nanomedicine for Effective Glioblastoma Therapy. J. Control. Release 2022, 351, 739–751. [Google Scholar] [CrossRef]
  963. Akram, F.; Haq, I.U.; Sahreen, S.; Nasir, N.; Naseem, W.; Imitaz, M.; Aqeel, A. CRISPR/Cas9: A Revolutionary Genome Editing Tool for Human Cancers Treatment. Technol. Cancer Res. Treat. 2022, 21, 15330338221132078. [Google Scholar] [CrossRef]
  964. Yang, Q.; Zhou, Y.; Chen, J.; Huang, N.; Wang, Z.; Cheng, Y. Gene Therapy for Drug-Resistant Glioblastoma via Lipid-Polymer Hybrid Nanoparticles Combined with Focused Ultrasound. Int. J. Nanomed. 2021, 16, 185–199. [Google Scholar] [CrossRef]
  965. Liu, X.; Cao, Z.; Wang, W.; Zou, C.; Wang, Y.; Pan, L.; Jia, B.; Zhang, K.; Zhang, W.; Li, W.; et al. Engineered Extracellular Vesicle-Delivered CRISPR/Cas9 for Radiotherapy Sensitization of Glioblastoma. ACS Nano 2023, 17, 16432–16447. [Google Scholar] [CrossRef]
  966. Tran, A.; Fierro, J.; Dou, H. CSIG-11. TARGETING PD-L1 IN GLIOBLASTOMA USING NANOPARTICLE-BASED GENE EDITING. Neuro Oncol. 2020, 22, ii29–ii30. [Google Scholar] [CrossRef]
  967. Wang, R.; Degirmenci, V.; Xin, H.; Li, Y.; Wang, L.; Chen, J.; Hu, X.; Zhang, D. PEI-Coated Fe3O4 Nanoparticles Enable Efficient Delivery of Therapeutic siRNA Targeting REST into Glioblastoma Cells. Int. J. Mol. Sci. 2018, 19, 2230. [Google Scholar] [CrossRef]
  968. Fierro, J.; DiPasquale, J.; Perez, J.; Chin, B.; Chokpapone, Y.; Tran, A.M.; Holden, A.; Factoriza, C.; Sivagnanakumar, N.; Aguilar, R.; et al. Dual-sgRNA CRISPR/Cas9 Knockout of PD-L1 in Human U87 Glioblastoma Tumor Cells Inhibits Proliferation, Invasion, and Tumor-Associated Macrophage Polarization. Sci. Rep. 2022, 12, 2417. [Google Scholar] [CrossRef] [PubMed]
  969. Chang, Y.; Cai, X.; Syahirah, R.; Yao, Y.; Xu, Y.; Jin, G.; Bhute, V.J.; Torregrosa-Allen, S.; Elzey, B.D.; Won, Y.-Y.; et al. CAR-Neutrophil Mediated Delivery of Tumor-Microenvironment Responsive Nanodrugs for Glioblastoma Chemo-Immunotherapy. Nat. Commun. 2023, 14, 2266. [Google Scholar] [CrossRef] [PubMed]
  970. Platt, R.J.; Chen, S.; Zhou, Y.; Yim, M.J.; Swiech, L.; Kempton, H.R.; Dahlman, J.E.; Parnas, O.; Eisenhaure, T.M.; Jovanovic, M.; et al. CRISPR-Cas9 Knockin Mice for Genome Editing and Cancer Modeling. Cell 2014, 159, 440–455. [Google Scholar] [CrossRef] [PubMed]
  971. Zheng, Y.; Shen, W.; Zhang, J.; Yang, B.; Liu, Y.-N.; Qi, H.; Yu, X.; Lu, S.-Y.; Chen, Y.; Xu, Y.-Z.; et al. CRISPR Interference-Based Specific and Efficient Gene Inactivation in the Brain. Nat. Neurosci. 2018, 21, 447–454. [Google Scholar] [CrossRef] [PubMed]
  972. Yin, H.; Song, C.-Q.; Dorkin, J.R.; Zhu, L.J.; Li, Y.; Wu, Q.; Park, A.; Yang, J.; Suresh, S.; Bizhanova, A.; et al. Therapeutic Genome Editing by Combined Viral and Non-Viral Delivery of CRISPR System Components in Vivo. Nat. Biotechnol. 2016, 34, 328–333. [Google Scholar] [CrossRef] [PubMed]
  973. Miller, J.B.; Zhang, S.; Kos, P.; Xiong, H.; Zhou, K.; Perelman, S.S.; Zhu, H.; Siegwart, D.J. Non-Viral CRISPR/Cas Gene Editing In Vitro and In Vivo Enabled by Synthetic Nanoparticle Co-Delivery of Cas9 mRNA and sgRNA. Angew. Chem. Int. Ed. Engl. 2017, 56, 1059–1063. [Google Scholar] [CrossRef] [PubMed]
  974. Sinclair, F.; Begum, A.A.; Dai, C.C.; Toth, I.; Moyle, P.M. Recent Advances in the Delivery and Applications of Nonviral CRISPR/Cas9 Gene Editing. Drug Deliv. Transl. Res. 2023, 13, 1500–1519. [Google Scholar] [CrossRef]
  975. Voth, B.L.; Pelargos, P.E.; Barnette, N.E.; Bhatt, N.S.; Chen, C.H.J.; Lagman, C.; Chung, L.K.; Nguyen, T.; Sheppard, J.P.; Romiyo, P.; et al. Intratumor Injection of CCL21-Coupled Vault Nanoparticles Is Associated with Reduction in Tumor Volume in an in Vivo Model of Glioma. J. Neurooncol. 2020, 147, 599–605. [Google Scholar] [CrossRef]
  976. Galstyan, A.; Markman, J.L.; Shatalova, E.S.; Chiechi, A.; Korman, A.J.; Patil, R.; Klymyshyn, D.; Tourtellotte, W.G.; Israel, L.L.; Braubach, O.; et al. Blood–Brain Barrier Permeable Nano Immunoconjugates Induce Local Immune Responses for Glioma Therapy. Nat. Commun. 2019, 10, 3850. [Google Scholar] [CrossRef]
  977. Guo, H.; Wang, R.; Wang, D.; Wang, S.; Zhou, J.; Chai, Z.; Yao, S.; Li, J.; Lu, L.; Liu, Y.; et al. Deliver Anti-PD-L1 into Brain by p-Hydroxybenzoic Acid to Enhance Immunotherapeutic Effect for Glioblastoma. J. Control. Release 2020, 320, 63–72. [Google Scholar] [CrossRef]
  978. Wang, H.; Chao, Y.; Zhao, H.; Zhou, X.; Zhang, F.; Zhang, Z.; Li, Z.; Pan, J.; Wang, J.; Chen, Q.; et al. Smart Nanomedicine to Enable Crossing Blood-Brain Barrier Delivery of Checkpoint Blockade Antibody for Immunotherapy of Glioma. ACS Nano 2022, 16, 664–674. [Google Scholar] [CrossRef] [PubMed]
  979. Li, T.-F.; Li, K.; Wang, C.; Liu, X.; Wen, Y.; Xu, Y.-H.; Zhang, Q.; Zhao, Q.-Y.; Shao, M.; Li, Y.-Z.; et al. Harnessing the Cross-Talk between Tumor Cells and Tumor-Associated Macrophages with a Nano-Drug for Modulation of Glioblastoma Immune Microenvironment. J. Control Release 2017, 268, 128–146. [Google Scholar] [CrossRef] [PubMed]
  980. Wang, T.; Zhang, H.; Qiu, W.; Han, Y.; Liu, H.; Li, Z. Biomimetic Nanoparticles Directly Remodel Immunosuppressive Microenvironment for Boosting Glioblastoma Immunotherapy. Bioact. Mater. 2022, 16, 418–432. [Google Scholar] [CrossRef] [PubMed]
  981. Qiu, Q.; Chen, S.; He, H.; Chen, J.; Ding, X.; Wang, D.; Yang, J.; Guo, P.; Li, Y.; Kim, J.; et al. An Injectable Signal-Amplifying Device Elicits a Specific Immune Response against Malignant Glioblastoma. Acta Pharm. Sin. B 2023. [Google Scholar] [CrossRef] [PubMed]
  982. Melero, I.; Castanon, E.; Alvarez, M.; Champiat, S.; Marabelle, A. Intratumoural Administration and Tumour Tissue Targeting of Cancer Immunotherapies. Nat. Rev. Clin. Oncol. 2021, 18, 558–576. [Google Scholar] [CrossRef] [PubMed]
  983. Cheng, F.; Su, T.; Zhou, S.; Liu, X.; Yang, S.; Lin, S.; Guo, W.; Zhu, G. Single-Dose Injectable Nanovaccine-in-Hydrogel for Robust Immunotherapy of Large Tumors with Abscopal Effect. Sci. Adv. 2023, 9, eade6257. [Google Scholar] [CrossRef] [PubMed]
  984. Mahmoudi, K.; Bouras, A.; Bozec, D.; Ivkov, R.; Hadjipanayis, C. Magnetic Hyperthermia Therapy for the Treatment of Glioblastoma: A Review of the Therapy’s History, Efficacy and Application in Humans. Int. J. Hyperth. 2018, 34, 1316–1328. [Google Scholar] [CrossRef]
  985. Maier-Hauff, K.; Ulrich, F.; Nestler, D.; Niehoff, H.; Wust, P.; Thiesen, B.; Orawa, H.; Budach, V.; Jordan, A. Efficacy and Safety of Intratumoral Thermotherapy Using Magnetic Iron-Oxide Nanoparticles Combined with External Beam Radiotherapy on Patients with Recurrent Glioblastoma Multiforme. J. Neurooncol. 2011, 103, 317–324. [Google Scholar] [CrossRef]
  986. Beier, C.P.; Schmid, C.; Gorlia, T.; Kleinletzenberger, C.; Beier, D.; Grauer, O.; Steinbrecher, A.; Hirschmann, B.; Brawanski, A.; Dietmaier, C.; et al. RNOP-09: Pegylated Liposomal Doxorubicine and Prolonged Temozolomide in Addition to Radiotherapy in Newly Diagnosed Glioblastoma--a Phase II Study. BMC Cancer 2009, 9, 308. [Google Scholar] [CrossRef] [PubMed]
  987. Ananda, S.; Nowak, A.K.; Cher, L.; Dowling, A.; Brown, C.; Simes, J.; Rosenthal, M.A. Cooperative Trials Group for Neuro-Oncology (COGNO) Phase 2 Trial of Temozolomide and Pegylated Liposomal Doxorubicin in the Treatment of Patients with Glioblastoma Multiforme Following Concurrent Radiotherapy and Chemotherapy. J. Clin. Neurosci. 2011, 18, 1444–1448. [Google Scholar] [CrossRef] [PubMed]
  988. Kim, S.-S.; Rait, A.; Kim, E.; Pirollo, K.F.; Chang, E.H. A Tumor-Targeting P53 Nanodelivery System Limits Chemoresistance to Temozolomide Prolonging Survival in a Mouse Model of Glioblastoma Multiforme. Nanomedicine 2015, 11, 301–311. [Google Scholar] [CrossRef] [PubMed]
  989. Kim, S.-S.; Harford, J.B.; Moghe, M.; Slaughter, T.; Doherty, C.; Chang, E.H. A Tumor-Targeting Nanomedicine Carrying the P53 Gene Crosses the Blood-Brain Barrier and Enhances Anti-PD-1 Immunotherapy in Mouse Models of Glioblastoma. Int. J. Cancer 2019, 145, 2535–2546. [Google Scholar] [CrossRef]
  990. Kumthekar, P.; Ko, C.H.; Paunesku, T.; Dixit, K.; Sonabend, A.M.; Bloch, O.; Tate, M.; Schwartz, M.; Zuckerman, L.; Lezon, R.; et al. A First-in-Human Phase 0 Clinical Study of RNA Interference-Based Spherical Nucleic Acids in Patients with Recurrent Glioblastoma. Sci. Transl. Med. 2021, 13, eabb3945. [Google Scholar] [CrossRef]
  991. Melnick, K.; Shin, D.; Dastmalchi, F.; Kabeer, Z.; Rahman, M.; Tran, D.; Ghiaseddin, A. Role of Laser Interstitial Thermal Therapy in the Management of Primary and Metastatic Brain Tumors. Curr. Treat. Options Oncol. 2021, 22, 108. [Google Scholar] [CrossRef]
  992. Lerner, E.C.; Edwards, R.M.; Wilkinson, D.S.; Fecci, P.E. Laser Ablation: Heating up the Anti-Tumor Response in the Intracranial Compartment. Adv. Drug Deliv. Rev. 2022, 185, 114311. [Google Scholar] [CrossRef]
  993. Traylor, J.I.; Patel, R.; Muir, M.; de Almeida Bastos, D.C.; Ravikumar, V.; Kamiya-Matsuoka, C.; Rao, G.; Thomas, J.G.; Kew, Y.; Prabhu, S.S. Laser Interstitial Thermal Therapy for Glioblastoma: A Single-Center Experience. World Neurosurg. 2021, 149, e244–e252. [Google Scholar] [CrossRef]
  994. Alkazemi, M.; Lo, Y.T.; Hussein, H.; Mammi, M.; Saleh, S.; Araujo-Lama, L.; Mommsen, S.; Pisano, A.; Lamba, N.; Bunevicius, A.; et al. Laser Interstitial Thermal Therapy for the Treatment of Primary and Metastatic Brain Tumors: A Systematic Review and Meta-Analysis. World Neurosurg. 2023, 171, e654–e671. [Google Scholar] [CrossRef]
  995. de Groot, J.F.; Kim, A.H.; Prabhu, S.; Rao, G.; Laxton, A.W.; Fecci, P.E.; O’Brien, B.J.; Sloan, A.; Chiang, V.; Tatter, S.B.; et al. Efficacy of Laser Interstitial Thermal Therapy (LITT) for Newly Diagnosed and Recurrent IDH Wild-Type Glioblastoma. Neurooncol. Adv. 2022, 4, vdac040. [Google Scholar] [CrossRef]
  996. Roberts, J.W.; Powlovich, L.; Sheybani, N.; LeBlang, S. Focused Ultrasound for the Treatment of Glioblastoma. J. Neurooncol. 2022, 157, 237–247. [Google Scholar] [CrossRef]
  997. Johansen, P.M.; Hansen, P.Y.; Mohamed, A.A.; Girshfeld, S.J.; Feldmann, M.; Lucke-Wold, B. Focused Ultrasound for Treatment of Peripheral Brain Tumors. Explor. Drug Sci. 2023, 1, 107–125. [Google Scholar] [CrossRef]
  998. Mungur, R.; Zheng, J.; Wang, B.; Chen, X.; Zhan, R.; Tong, Y. Low-Intensity Focused Ultrasound Technique in Glioblastoma Multiforme Treatment. Front. Oncol. 2022, 12. [Google Scholar] [CrossRef]
  999. Elhelf, I.A.S.; Albahar, H.; Shah, U.; Oto, A.; Cressman, E.; Almekkawy, M. High Intensity Focused Ultrasound: The Fundamentals, Clinical Applications and Research Trends. Diagn. Interv. Imaging 2018, 99, 349–359. [Google Scholar] [CrossRef]
  1000. Quadri, S.A.; Waqas, M.; Khan, I.; Khan, M.A.; Suriya, S.S.; Farooqui, M.; Fiani, B. High-Intensity Focused Ultrasound: Past, Present, and Future in Neurosurgery. Neurosurg. Focus 2018, 44, E16. [Google Scholar] [CrossRef] [PubMed]
  1001. Coluccia, D.; Fandino, J.; Schwyzer, L.; O’Gorman, R.; Remonda, L.; Anon, J.; Martin, E.; Werner, B. First Noninvasive Thermal Ablation of a Brain Tumor with MR-Guided Focused Ultrasound. J. Ther. Ultrasound 2014, 2, 17. [Google Scholar] [CrossRef] [PubMed]
  1002. MacDonell, J.; Patel, N.; Rubino, S.; Ghoshal, G.; Fischer, G.; Burdette, E.C.; Hwang, R.; Pilitsis, J.G. Magnetic Resonance-Guided Interstitial High-Intensity Focused Ultrasound for Brain Tumor Ablation. Neurosurg. Focus 2018, 44, E11. [Google Scholar] [CrossRef] [PubMed]
  1003. Hersh, A.M.; Bhimreddy, M.; Weber-Levine, C.; Jiang, K.; Alomari, S.; Theodore, N.; Manbachi, A.; Tyler, B.M. Applications of Focused Ultrasound for the Treatment of Glioblastoma: A New Frontier. Cancers 2022, 14, 4920. [Google Scholar] [CrossRef]
  1004. Arsiwala, T.A.; Sprowls, S.A.; Blethen, K.E.; Adkins, C.E.; Saralkar, P.A.; Fladeland, R.A.; Pentz, W.; Gabriele, A.; Kielkowski, B.; Mehta, R.I.; et al. Ultrasound-Mediated Disruption of the Blood Tumor Barrier for Improved Therapeutic Delivery. Neoplasia 2021, 23, 676–691. [Google Scholar] [CrossRef] [PubMed]
  1005. Hersh, D.S.; Wadajkar, A.S.; Roberts, N.; Perez, J.G.; Connolly, N.P.; Frenkel, V.; Winkles, J.A.; Woodworth, G.F.; Kim, A.J. Evolving Drug Delivery Strategies to Overcome the Blood Brain Barrier. Curr. Pharm. Des. 2016, 22, 1177–1193. [Google Scholar] [CrossRef]
  1006. Liu, H.-L.; Hua, M.-Y.; Chen, P.-Y.; Chu, P.-C.; Pan, C.-H.; Yang, H.-W.; Huang, C.-Y.; Wang, J.-J.; Yen, T.-C.; Wei, K.-C. Blood-Brain Barrier Disruption with Focused Ultrasound Enhances Delivery of Chemotherapeutic Drugs for Glioblastoma Treatment. Radiology 2010, 255, 415–425. [Google Scholar] [CrossRef]
  1007. Wei, H.-J.; Upadhyayula, P.S.; Pouliopoulos, A.N.; Englander, Z.K.; Zhang, X.; Jan, C.-I.; Guo, J.; Mela, A.; Zhang, Z.; Wang, T.J.C.; et al. Focused Ultrasound-Mediated Blood-Brain Barrier Opening Increases Delivery and Efficacy of Etoposide for Glioblastoma Treatment. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 539–550. [Google Scholar] [CrossRef] [PubMed]
  1008. Liu, H.-L.; Huang, C.-Y.; Chen, J.-Y.; Wang, H.-Y.J.; Chen, P.-Y.; Wei, K.-C. Pharmacodynamic and Therapeutic Investigation of Focused Ultrasound-Induced Blood-Brain Barrier Opening for Enhanced Temozolomide Delivery in Glioma Treatment. PLoS ONE 2014, 9, e114311. [Google Scholar] [CrossRef]
  1009. Noroozian, Z.; Xhima, K.; Huang, Y.; Kaspar, B.K.; Kügler, S.; Hynynen, K.; Aubert, I. MRI-Guided Focused Ultrasound for Targeted Delivery of rAAV to the Brain. Methods Mol. Biol. 2019, 1950, 177–197. [Google Scholar] [CrossRef]
  1010. Alkins, R.; Burgess, A.; Ganguly, M.; Francia, G.; Kerbel, R.; Wels, W.S.; Hynynen, K. Focused Ultrasound Delivers Targeted Immune Cells to Metastatic Brain Tumors. Cancer Res. 2013, 73, 1892–1899. [Google Scholar] [CrossRef]
  1011. Nance, E.; Timbie, K.; Miller, G.W.; Song, J.; Louttit, C.; Klibanov, A.L.; Shih, T.-Y.; Swaminathan, G.; Tamargo, R.J.; Woodworth, G.F.; et al. Non-Invasive Delivery of Stealth, Brain-Penetrating Nanoparticles across the Blood-Brain Barrier Using MRI-Guided Focused Ultrasound. J. Control Release 2014, 189, 123–132. [Google Scholar] [CrossRef]
  1012. Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M.; et al. Enhancing Glioblastoma Treatment Using Cisplatin-Gold-Nanoparticle Conjugates and Targeted Delivery with Magnetic Resonance-Guided Focused Ultrasound. Nanomedicine 2018, 14, 1137–1148. [Google Scholar] [CrossRef] [PubMed]
  1013. Janjua, T.I.; Cao, Y.; Ahmed-Cox, A.; Raza, A.; Moniruzzaman, M.; Akhter, D.T.; Fletcher, N.L.; Kavallaris, M.; Thurecht, K.J.; Popat, A. Efficient Delivery of Temozolomide Using Ultrasmall Large-Pore Silica Nanoparticles for Glioblastoma. J. Control Release 2023, 357, 161–174. [Google Scholar] [CrossRef] [PubMed]
  1014. Liu, H.-L.; Hsieh, H.-Y.; Lu, L.-A.; Kang, C.-W.; Wu, M.-F.; Lin, C.-Y. Low-Pressure Pulsed Focused Ultrasound with Microbubbles Promotes an Anticancer Immunological Response. J. Transl. Med. 2012, 10, 221. [Google Scholar] [CrossRef] [PubMed]
  1015. Unga, J.; Hashida, M. Ultrasound Induced Cancer Immunotherapy. Adv. Drug Deliv. Rev. 2014, 72, 144–153. [Google Scholar] [CrossRef] [PubMed]
  1016. Liu, S.; Zhang, Y.; Liu, Y.; Wang, W.; Gao, S.; Yuan, W.; Sun, Z.; Liu, L.; Wang, C. Ultrasound-Targeted Microbubble Destruction Remodels Tumour Microenvironment to Improve Immunotherapeutic Effect. Br. J. Cancer 2023, 128, 715–725. [Google Scholar] [CrossRef] [PubMed]
  1017. Cohen-Inbar, O.; Xu, Z.; Sheehan, J.P. Focused Ultrasound-Aided Immunomodulation in Glioblastoma Multiforme: A Therapeutic Concept. J. Ther. Ultrasound 2016, 4, 2. [Google Scholar] [CrossRef] [PubMed]
  1018. Chen, K.-T.; Chai, W.-Y.; Lin, Y.-J.; Lin, C.-J.; Chen, P.-Y.; Tsai, H.-C.; Huang, C.-Y.; Kuo, J.S.; Liu, H.-L.; Wei, K.-C. Neuronavigation-Guided Focused Ultrasound for Transcranial Blood-Brain Barrier Opening and Immunostimulation in Brain Tumors. Sci. Adv. 2021, 7, eabd0772. [Google Scholar] [CrossRef]
  1019. Bathini, P.; Sun, T.; Schenk, M.; Schilling, S.; McDannold, N.J.; Lemere, C.A. Acute Effects of Focused Ultrasound-Induced Blood-Brain Barrier Opening on Anti-Pyroglu3 Abeta Antibody Delivery and Immune Responses. Biomolecules 2022, 12, 951. [Google Scholar] [CrossRef]
  1020. Idbaih, A.; Canney, M.; Belin, L.; Desseaux, C.; Vignot, A.; Bouchoux, G.; Asquier, N.; Law-Ye, B.; Leclercq, D.; Bissery, A.; et al. Safety and Feasibility of Repeated and Transient Blood-Brain Barrier Disruption by Pulsed Ultrasound in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2019, 25, 3793–3801. [Google Scholar] [CrossRef]
  1021. Park, S.H.; Kim, M.J.; Jung, H.H.; Chang, W.S.; Choi, H.S.; Rachmilevitch, I.; Zadicario, E.; Chang, J.W. One-Year Outcome of Multiple Blood-Brain Barrier Disruptions With Temozolomide for the Treatment of Glioblastoma. Front. Oncol. 2020, 10, 1663. [Google Scholar] [CrossRef] [PubMed]
  1022. Sonabend, A.M.; Gould, A.; Amidei, C.; Ward, R.; Schmidt, K.A.; Zhang, D.Y.; Gomez, C.; Bebawy, J.F.; Liu, B.P.; Bouchoux, G.; et al. Repeated Blood-Brain Barrier Opening with an Implantable Ultrasound Device for Delivery of Albumin-Bound Paclitaxel in Patients with Recurrent Glioblastoma: A Phase 1 Trial. Lancet Oncol. 2023, 24, 509–522. [Google Scholar] [CrossRef] [PubMed]
  1023. Kim, E.; Van Reet, J.; Kim, H.-C.; Kowsari, K.; Yoo, S.-S. High Incidence of Intracerebral Hemorrhaging Associated with the Application of Low-Intensity Focused Ultrasound Following Acute Cerebrovascular Injury by Intracortical Injection. Pharmaceutics 2022, 14, 2120. [Google Scholar] [CrossRef] [PubMed]
  1024. McHale, A.P.; Callan, J.F.; Nomikou, N.; Fowley, C.; Callan, B. Sonodynamic Therapy: Concept, Mechanism and Application to Cancer Treatment. Adv. Exp. Med. Biol. 2016, 880, 429–450. [Google Scholar] [CrossRef] [PubMed]
  1025. Cramer, S.W.; Chen, C.C. Photodynamic Therapy for the Treatment of Glioblastoma. Front. Surg. 2020, 6. [Google Scholar] [CrossRef]
  1026. Zheng, Y.; Ye, J.; Li, Z.; Chen, H.; Gao, Y. Recent Progress in Sono-Photodynamic Cancer Therapy: From Developed New Sensitizers to Nanotechnology-Based Efficacy-Enhancing Strategies. Acta Pharm. Sin. B 2021, 11, 2197–2219. [Google Scholar] [CrossRef] [PubMed]
  1027. Bhanja, D.; Wilding, H.; Baroz, A.; Trifoi, M.; Shenoy, G.; Slagle-Webb, B.; Hayes, D.; Soudagar, Y.; Connor, J.; Mansouri, A. Photodynamic Therapy for Glioblastoma: Illuminating the Path toward Clinical Applicability. Cancers 2023, 15, 3427. [Google Scholar] [CrossRef]
  1028. Song, D.; Yue, W.; Li, Z.; Li, J.; Zhao, J.; Zhang, N. Study of the Mechanism of Sonodynamic Therapy in a Rat Glioma Model. Onco Targets Ther. 2014, 7, 1801–1810. [Google Scholar] [CrossRef]
  1029. Mroz, P.; Hashmi, J.T.; Huang, Y.-Y.; Lange, N.; Hamblin, M.R. Stimulation of Anti-Tumor Immunity by Photodynamic Therapy. Expert Rev. Clin. Immunol. 2011, 7, 75–91. [Google Scholar] [CrossRef]
  1030. Falk-Mahapatra, R.; Gollnick, S.O. Photodynamic Therapy and Immunity: An Update. Photochem. Photobiol. 2020, 96, 550–559. [Google Scholar] [CrossRef]
  1031. Bunevicius, A.; Pikis, S.; Padilla, F.; Prada, F.; Sheehan, J. Sonodynamic Therapy for Gliomas. J. Neurooncol. 2022, 156, 1–10. [Google Scholar] [CrossRef] [PubMed]
  1032. Mahmoudi, K.; Garvey, K.; Bouras, A.; Cramer, G.; Stepp, H.; Jesu Raj, J.; Bozec, D.; Busch, T.; Hadjipanayis, C. 5-Aminolevulinic Acid Photodynamic Therapy for the Treatment of High-Grade Gliomas. J. Neurooncol. 2019, 141, 595–607. [Google Scholar] [CrossRef] [PubMed]
  1033. Hsia, T.; Small, J.L.; Yekula, A.; Batool, S.M.; Escobedo, A.K.; Ekanayake, E.; You, D.G.; Lee, H.; Carter, B.S.; Balaj, L. Systematic Review of Photodynamic Therapy in Gliomas. Cancers 2023, 15, 3918. [Google Scholar] [CrossRef]
  1034. Wang, W.; Tabu, K.; Hagiya, Y.; Sugiyama, Y.; Kokubu, Y.; Murota, Y.; Ogura, S.-I.; Taga, T. Enhancement of 5-Aminolevulinic Acid-Based Fluorescence Detection of Side Population-Defined Glioma Stem Cells by Iron Chelation. Sci. Rep. 2017, 7, 42070. [Google Scholar] [CrossRef]
  1035. Müller, P.; Abdel Gaber, S.A.; Zimmermann, W.; Wittig, R.; Stepp, H. ABCG2 Influence on the Efficiency of Photodynamic Therapy in Glioblastoma Cells. J. Photochem. Photobiol. B 2020, 210, 111963. [Google Scholar] [CrossRef] [PubMed]
  1036. Leroy, H.-A.; Guérin, L.; Lecomte, F.; Baert, G.; Vignion, A.-S.; Mordon, S.; Reyns, N. Is Interstitial Photodynamic Therapy for Brain Tumors Ready for Clinical Practice? A Systematic Review. Photodiagnosis Photodyn. Ther. 2021, 36, 102492. [Google Scholar] [CrossRef] [PubMed]
  1037. Schwartz, C.; Rühm, A.; Tonn, J.-C.; Kreth, S.; Kreth, F.-W. SURG-25INTERSTITIAL PHOTODYNAMIC THERAPY OF DE-NOVO GLIOBLASTOMA MULTIFORME WHO IV. Neuro Oncol. 2015, 17, v219–v220. [Google Scholar] [CrossRef]
  1038. Nitta, M.; Muragaki, Y.; Maruyama, T.; Iseki, H.; Komori, T.; Ikuta, S.; Saito, T.; Yasuda, T.; Hosono, J.; Okamoto, S.; et al. Role of Photodynamic Therapy Using Talaporfin Sodium and a Semiconductor Laser in Patients with Newly Diagnosed Glioblastoma. J. Neurosurg. 2018, 131, 1361–1368. [Google Scholar] [CrossRef]
  1039. Lietke, S.; Schmutzer, M.; Schwartz, C.; Weller, J.; Siller, S.; Aumiller, M.; Heckl, C.; Forbrig, R.; Niyazi, M.; Egensperger, R.; et al. Interstitial Photodynamic Therapy Using 5-ALA for Malignant Glioma Recurrences. Cancers 2021, 13, 1767. [Google Scholar] [CrossRef]
  1040. Foglar, M.; Aumiller, M.; Bochmann, K.; Buchner, A.; El Fahim, M.; Quach, S.; Sroka, R.; Stepp, H.; Thon, N.; Forbrig, R.; et al. Interstitial Photodynamic Therapy of Glioblastomas: A Long-Term Follow-up Analysis of Survival and Volumetric MRI Data. Cancers 2023, 15, 2603. [Google Scholar] [CrossRef]
  1041. Henderson, T.A.; Morries, L.D. Near-Infrared Photonic Energy Penetration: Can Infrared Phototherapy Effectively Reach the Human Brain? Neuropsychiatr. Dis. Treat. 2015, 11, 2191–2208. [Google Scholar] [CrossRef]
  1042. Lee, S.Y.; Lee, R.; Kim, E.; Lee, S.; Park, Y.I. Near-Infrared Light-Triggered Photodynamic Therapy and Apoptosis Using Upconversion Nanoparticles With Dual Photosensitizers. Front. Bioeng. Biotechnol. 2020, 8, 275. [Google Scholar] [CrossRef]
  1043. Burley, T.A.; Mączyńska, J.; Shah, A.; Szopa, W.; Harrington, K.J.; Boult, J.K.R.; Mrozek-Wilczkiewicz, A.; Vinci, M.; Bamber, J.C.; Kaspera, W.; et al. Near-Infrared Photoimmunotherapy Targeting EGFR-Shedding New Light on Glioblastoma Treatment. Int. J. Cancer 2018, 142, 2363–2374. [Google Scholar] [CrossRef]
  1044. Mączyńska, J.; Raes, F.; Da Pieve, C.; Turnock, S.; Boult, J.K.R.; Hoebart, J.; Niedbala, M.; Robinson, S.P.; Harrington, K.J.; Kaspera, W.; et al. Triggering Anti-GBM Immune Response with EGFR-Mediated Photoimmunotherapy. BMC Med. 2022, 20, 16. [Google Scholar] [CrossRef]
  1045. Mohiuddin, T.M.; Zhang, C.; Sheng, W.; Al-Rawe, M.; Zeppernick, F.; Meinhold-Heerlein, I.; Hussain, A.F. Near Infrared Photoimmunotherapy: A Review of Recent Progress and Their Target Molecules for Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 2655. [Google Scholar] [CrossRef] [PubMed]
  1046. Zhang, Y.; He, L.; Wu, J.; Wang, K.; Wang, J.; Dai, W.; Yuan, A.; Wu, J.; Hu, Y. Switchable PDT for Reducing Skin Photosensitization by a NIR Dye Inducing Self-Assembled and Photo-Disassembled Nanoparticles. Biomaterials 2016, 107, 23–32. [Google Scholar] [CrossRef] [PubMed]
  1047. Kang, R.H.; Kim, Y.; Um, H.J.; Kim, J.; Bang, E.-K.; Yeo, S.G.; Kim, D. Glioblastoma Homing Photodynamic Therapy Based on Multifunctionalized Porous Silicon Nanoparticles. ACS Appl. Nano Mater. 2022, 5, 5387–5397. [Google Scholar] [CrossRef]
  1048. Pellosi, D.S.; Paula, L.B.; de Melo, M.T.; Tedesco, A.C. Targeted and Synergic Glioblastoma Treatment: Multifunctional Nanoparticles Delivering Verteporfin as Adjuvant Therapy for Temozolomide Chemotherapy. Mol. Pharm. 2019, 16, 1009–1024. [Google Scholar] [CrossRef] [PubMed]
  1049. Ihata, T.; Nonoguchi, N.; Fujishiro, T.; Omura, N.; Kawabata, S.; Kajimoto, Y.; Wanibuchi, M. The Effect of Hypoxia on Photodynamic Therapy with 5-Aminolevulinic Acid in Malignant Gliomas. Photodiagnosis Photodyn. Ther. 2022, 40, 103056. [Google Scholar] [CrossRef] [PubMed]
  1050. Ma, S.; Wang, F.; Dong, J.; Wang, N.; Tao, S.; Du, J.; Hu, S. Inhibition of Hypoxia-Inducible Factor 1 by Acriflavine Renders Glioblastoma Sensitive for Photodynamic Therapy. J. Photochem. Photobiol. B 2022, 234, 112537. [Google Scholar] [CrossRef] [PubMed]
  1051. Caverzán, M.D.; Oliveda, P.M.; Beaugé, L.; Palacios, R.E.; Chesta, C.A.; Ibarra, L.E. Metronomic Photodynamic Therapy with Conjugated Polymer Nanoparticles in Glioblastoma Tumor Microenvironment. Cells 2023, 12, 1541. [Google Scholar] [CrossRef]
  1052. Omura, N.; Nonoguchi, N.; Fujishiro, T.; Park, Y.; Ikeda, N.; Kajimoto, Y.; Hosomi, R.; Yagi, R.; Hiramatsu, R.; Furuse, M.; et al. Ablation Efficacy of 5-Aminolevulinic Acid-Mediated Photodynamic Therapy on Human Glioma Stem Cells. Photodiagnosis Photodyn. Ther. 2023, 41, 103119. [Google Scholar] [CrossRef] [PubMed]
  1053. Borah, B.M.; Cacaccio, J.; Durrani, F.A.; Bshara, W.; Turowski, S.G.; Spernyak, J.A.; Pandey, R.K. Sonodynamic Therapy in Combination with Photodynamic Therapy Shows Enhanced Long-Term Cure of Brain Tumor. Sci. Rep. 2020, 10, 21791. [Google Scholar] [CrossRef] [PubMed]
  1054. Park, J.; Kong, C.; Shin, J.; Park, J.Y.; Na, Y.C.; Han, S.H.; Chang, J.W.; Song, S.H.; Chang, W.S. Combined Effects of Focused Ultrasound and Photodynamic Treatment for Malignant Brain Tumors Using C6 Glioma Rat Model. Yonsei Med. J. 2023, 64, 233–242. [Google Scholar] [CrossRef]
  1055. Ohmura, T.; Fukushima, T.; Shibaguchi, H.; Yoshizawa, S.; Inoue, T.; Kuroki, M.; Sasaki, K.; Umemura, S.-I. Sonodynamic Therapy with 5-Aminolevulinic Acid and Focused Ultrasound for Deep-Seated Intracranial Glioma in Rat. Anticancer. Res. 2011, 31, 2527–2533. [Google Scholar] [PubMed]
  1056. Suehiro, S.; Ohnishi, T.; Yamashita, D.; Kohno, S.; Inoue, A.; Nishikawa, M.; Ohue, S.; Tanaka, J.; Kunieda, T. Enhancement of Antitumor Activity by Using 5-ALA-Mediated Sonodynamic Therapy to Induce Apoptosis in Malignant Gliomas: Significance of High-Intensity Focused Ultrasound on 5-ALA-SDT in a Mouse Glioma Model. J. Neurosurg. 2018, 129, 1416–1428. [Google Scholar] [CrossRef]
  1057. Wu, S.-K.; Santos, M.A.; Marcus, S.L.; Hynynen, K. MR-Guided Focused Ultrasound Facilitates Sonodynamic Therapy with 5-Aminolevulinic Acid in a Rat Glioma Model. Sci. Rep. 2019, 9, 10465. [Google Scholar] [CrossRef] [PubMed]
  1058. Raspagliesi, L.; D’Ammando, A.; Gionso, M.; Sheybani, N.D.; Lopes, M.-B.; Moore, D.; Allen, S.; Gatesman, J.; Porto, E.; Timbie, K.; et al. Intracranial Sonodynamic Therapy With 5-Aminolevulinic Acid and Sodium Fluorescein: Safety Study in a Porcine Model. Front. Oncol. 2021, 11, 679989. [Google Scholar] [CrossRef]
  1059. Bonosi, L.; Marino, S.; Benigno, U.E.; Musso, S.; Buscemi, F.; Giardina, K.; Gerardi, R.; Brunasso, L.; Costanzo, R.; Iacopino, D.G.; et al. Sonodynamic Therapy and Magnetic Resonance-Guided Focused Ultrasound: New Therapeutic Strategy in Glioblastoma. J. Neurooncol. 2023, 163, 219–238. [Google Scholar] [CrossRef]
  1060. Mumtaz, S.; Rana, J.N.; Choi, E.H.; Han, I. Microwave Radiation and the Brain: Mechanisms, Current Status, and Future Prospects. Int. J. Mol. Sci. 2022, 23, 9288. [Google Scholar] [CrossRef]
  1061. Chandra, R.; Zhou, H.; Balasingham, I.; Narayanan, R.M. On the Opportunities and Challenges in Microwave Medical Sensing and Imaging. IEEE Trans. Biomed. Eng. 2015, 62, 1667–1682. [Google Scholar] [CrossRef]
  1062. Ryan, T.P.; Brace, C.L. Interstitial Microwave Treatment for Cancer: Historical Basis and Current Techniques in Antenna Design and Performance. Int. J. Hyperth. 2017, 33, 3–14. [Google Scholar] [CrossRef]
  1063. Izzo, F.; Granata, V.; Grassi, R.; Fusco, R.; Palaia, R.; Delrio, P.; Carrafiello, G.; Azoulay, D.; Petrillo, A.; Curley, S.A. Radiofrequency Ablation and Microwave Ablation in Liver Tumors: An Update. Oncologist 2019, 24, e990–e1005. [Google Scholar] [CrossRef] [PubMed]
  1064. Hu, C.; Zuo, H.; Li, Y. Effects of Radiofrequency Electromagnetic Radiation on Neurotransmitters in the Brain. Front. Public Health 2021, 9. [Google Scholar] [CrossRef]
  1065. Rana, J.N.; Mumtaz, S.; Choi, E.H.; Han, I. ROS Production in Response to High-Power Microwave Pulses Induces P53 Activation and DNA Damage in Brain Cells: Radiosensitivity and Biological Dosimetry Evaluation. Front. Cell Dev. Biol. 2023, 11. [Google Scholar] [CrossRef] [PubMed]
  1066. Li, W.; Zhang, S.; Xing, D.; Qin, H. Pulsed Microwave-Induced Thermoacoustic Shockwave for Precise Glioblastoma Therapy with the Skin and Skull Intact. Small 2022, 18, e2201342. [Google Scholar] [CrossRef]
  1067. Jenkins, E.P.W.; Finch, A.; Gerigk, M.; Triantis, I.F.; Watts, C.; Malliaras, G.G. Electrotherapies for Glioblastoma. Adv. Sci. 2021, 8, 2100978. [Google Scholar] [CrossRef] [PubMed]
  1068. Lorenzo, M.F.; Arena, C.B.; Davalos, R.V. Maximizing Local Access to Therapeutic Deliveries in Glioblastoma. Part III: Irreversible Electroporation and High-Frequency Irreversible Electroporation for the Eradication of Glioblastoma. In Glioblastoma; De Vleeschouwer, S., Ed.; Codon Publications: Brisbane, Australia, 2017; ISBN 978-0-9944381-2-6. [Google Scholar]
  1069. Schoenbach, K.H.; Beebe, S.J.; Buescher, E.S. Intracellular Effect of Ultrashort Electrical Pulses. Bioelectromagnetics 2001, 22, 440–448. [Google Scholar] [CrossRef] [PubMed]
  1070. Vernier, P.T.; Sun, Y.; Marcu, L.; Craft, C.M.; Gundersen, M.A. Nanoelectropulse-Induced Phosphatidylserine Translocation. Biophys. J. 2004, 86, 4040–4048. [Google Scholar] [CrossRef] [PubMed]
  1071. Pakhomov, A.G.; Shevin, R.; White, J.A.; Kolb, J.F.; Pakhomova, O.N.; Joshi, R.P.; Schoenbach, K.H. Membrane Permeabilization and Cell Damage by Ultrashort Electric Field Shocks. Arch. Biochem. Biophys. 2007, 465, 109–118. [Google Scholar] [CrossRef]
  1072. Pakhomov, A.G.; Bowman, A.M.; Ibey, B.L.; Andre, F.M.; Pakhomova, O.N.; Schoenbach, K.H. Lipid Nanopores Can Form a Stable, Ion Channel-like Conduction Pathway in Cell Membrane. Biochem. Biophys. Res. Commun. 2009, 385, 181–186. [Google Scholar] [CrossRef]
  1073. Kotnik, T.; Rems, L.; Tarek, M.; Miklavčič, D. Membrane Electroporation and Electropermeabilization: Mechanisms and Models. Annu. Rev. Biophys. 2019, 48, 63–91. [Google Scholar] [CrossRef] [PubMed]
  1074. Vernier, P.T.; Li, A.; Marcu, L.; Craft, C.M.; Gundersen, M.A. Ultrashort Pulsed Electric Fields Induce Membrane Phospholipid Translocation and Caspase Activation: Differential Sensitivities of Jurkat T Lymphoblasts and Rat Glioma C6 Cells. IEEE Trans. Dielectr. Electr. Insul. 2003, 10, 795–809. [Google Scholar] [CrossRef]
  1075. White, J.A.; Blackmore, P.F.; Schoenbach, K.H.; Beebe, S.J. Stimulation of Capacitative Calcium Entry in HL-60 Cells by Nanosecond Pulsed Electric Fields *. J. Biol. Chem. 2004, 279, 22964–22972. [Google Scholar] [CrossRef]
  1076. Ren, W.; Beebe, S.J. An Apoptosis Targeted Stimulus with Nanosecond Pulsed Electric Fields (nsPEFs) in E4 Squamous Cell Carcinoma. Apoptosis 2011, 16, 382–393. [Google Scholar] [CrossRef] [PubMed]
  1077. Stacey, M.; Stickley, J.; Fox, P.; Statler, V.; Schoenbach, K.; Beebe, S.J.; Buescher, S. Differential Effects in Cells Exposed to Ultra-Short, High Intensity Electric Fields: Cell Survival, DNA Damage, and Cell Cycle Analysis. Mutat. Res. /Genet. Toxicol. Environ. Mutagen. 2003, 542, 65–75. [Google Scholar] [CrossRef]
  1078. Nuccitelli, R. Application of Pulsed Electric Fields to Cancer Therapy. Bioelectricity 2019, 1, 30–34. [Google Scholar] [CrossRef] [PubMed]
  1079. Garcia, P.A.; Pancotto, T.; Rossmeisl, J.H.; Henao-Guerrero, N.; Gustafson, N.R.; Daniel, G.B.; Robertson, J.L.; Ellis, T.L.; Davalos, R.V. Non-Thermal Irreversible Electroporation (N-TIRE) and Adjuvant Fractionated Radiotherapeutic Multimodal Therapy for Intracranial Malignant Glioma in a Canine Patient. Technol. Cancer Res. Treat. 2011, 10, 73–83. [Google Scholar] [CrossRef]
  1080. Herranz, C.; Fernández, F.; Martín-Ibáñez, R.; Blasco, E.; Crespo, E.; De la Fuente, C.; Añor, S.; Rabanal, R.M.; Canals, J.M.; Pumarola, M. Spontaneously Arising Canine Glioma as a Potential Model for Human Glioma. J. Comp. Pathol. 2016, 154, 169–179. [Google Scholar] [CrossRef]
  1081. Rossmeisl, J.H.; Garcia, P.A.; Pancotto, T.E.; Robertson, J.L.; Henao-Guerrero, N.; Neal, R.E.; Ellis, T.L.; Davalos, R.V. Safety and Feasibility of the NanoKnife System for Irreversible Electroporation Ablative Treatment of Canine Spontaneous Intracranial Gliomas. J. Neurosurg. 2015, 123, 1008–1025. [Google Scholar] [CrossRef]
  1082. Lefevre, M.C.; Dijk, G.; Kaszas, A.; Baca, M.; Moreau, D.; O’Connor, R.P. Integrating Flexible Electronics for Pulsed Electric Field Delivery in a Vascularized 3D Glioblastoma Model. npj Flex. Electron. 2021, 5, 1–9. [Google Scholar] [CrossRef]
  1083. Sano, M.B.; Arena, C.B.; Bittleman, K.R.; DeWitt, M.R.; Cho, H.J.; Szot, C.S.; Saur, D.; Cissell, J.M.; Robertson, J.; Lee, Y.W.; et al. Bursts of Bipolar Microsecond Pulses Inhibit Tumor Growth. Sci. Rep. 2015, 5, 14999. [Google Scholar] [CrossRef] [PubMed]
  1084. Sweeney, D.C.; Reberšek, M.; Dermol, J.; Rems, L.; Miklavčič, D.; Davalos, R.V. Quantification of Cell Membrane Permeability Induced by Monopolar and High-Frequency Bipolar Bursts of Electrical Pulses. Biochim. Biophys. Acta (BBA) Biomembr. 2016, 1858, 2689–2698. [Google Scholar] [CrossRef]
  1085. Latouche, E.L.; Arena, C.B.; Ivey, J.W.; Garcia, P.A.; Pancotto, T.E.; Pavlisko, N.; Verbridge, S.S.; Davalos, R.V.; Rossmeisl, J.H. High-Frequency Irreversible Electroporation for Intracranial Meningioma: A Feasibility Study in a Spontaneous Canine Tumor Model. Technol. Cancer Res. Treat. 2018, 17. [Google Scholar] [CrossRef]
  1086. Campelo, S.N.; Lorenzo, M.F.; Partridge, B.; Alinezhadbalalami, N.; Kani, Y.; Garcia, J.; Saunier, S.; Thomas, S.C.; Hinckley, J.; Verbridge, S.S.; et al. High-Frequency Irreversible Electroporation Improves Survival and Immune Cell Infiltration in Rodents with Malignant Gliomas. Front. Oncol. 2023, 13. [Google Scholar] [CrossRef] [PubMed]
  1087. Lepareur, N.; Ramée, B.; Mougin-Degraef, M.; Bourgeois, M. Clinical Advances and Perspectives in Targeted Radionuclide Therapy. Pharmaceutics 2023, 15, 1733. [Google Scholar] [CrossRef] [PubMed]
  1088. Kunikowska, J.; Morgenstern, A.; Pełka, K.; Bruchertseifer, F.; Królicki, L. Targeted Alpha Therapy for Glioblastoma. Front. Med. 2022, 9, 1085245. [Google Scholar] [CrossRef]
  1089. Cimini, A.; Ricci, M.; Russo, F.; Egidi, M.; Calabria, F.; Bagnato, A.; Schillaci, O.; Chiaravalloti, A. Peptide Receptor Radionuclide Therapy and Primary Brain Tumors: An Overview. Pharmaceuticals 2021, 14, 872. [Google Scholar] [CrossRef]
  1090. Li, Y.; Marcu, L.G.; Hull, A.; Bezak, E. Radioimmunotherapy of Glioblastoma Multiforme—Current Status and Future Prospects. Crit. Rev. Oncol. /Hematol. 2021, 163, 103395. [Google Scholar] [CrossRef]
  1091. Bailly, C.; Vidal, A.; Bonnemaire, C.; Kraeber-Bodéré, F.; Chérel, M.; Pallardy, A.; Rousseau, C.; Garcion, E.; Lacoeuille, F.; Hindré, F.; et al. Potential for Nuclear Medicine Therapy for Glioblastoma Treatment. Front. Pharmacol. 2019, 10. [Google Scholar] [CrossRef]
  1092. Casaco, A.; López, G.; García, I.; Arsenio Rodríguez, J.; Fernández, R.; Figueredo, J.; Torres, L.; Perera Pintado, A.; Batista, J.; Leyva, R.; et al. Phase I Single-Dose Study of Intracavitary-Administered Nimotuzumab Labeled with 188-Re in Adult Recurrent High-Grade Glioma. Cancer Biol. Ther. 2008, 7, 333–339. [Google Scholar] [CrossRef]
  1093. Li, L.; Quang, T.S.; Gracely, E.J.; Kim, J.H.; Emrich, J.G.; Yaeger, T.E.; Jenrette, J.M.; Cohen, S.C.; Black, P.; Brady, L.W. A Phase II Study of Anti–Epidermal Growth Factor Receptor Radioimmunotherapy in the Treatment of Glioblastoma Multiforme: Clinical Article. J. Neurosurg. 2010, 113, 192–198. [Google Scholar] [CrossRef]
  1094. Riva, P.; Franceschi, G.; Frattarelli, M.; Riva, N.; Guiducci, G.; Cremonini, A.M.; Giuliani, G.; Casi, M.; Gentile, R.; Jekunen, A.A.; et al. 131I Radioconjugated Antibodies for the Locoregional Radioimmunotherapy of High-Grade Malignant Glioma--Phase I and II Study. Acta Oncol. 1999, 38, 351–359. [Google Scholar] [CrossRef] [PubMed]
  1095. Reardon, D.A.; Quinn, J.A.; Akabani, G.; Coleman, R.E.; Friedman, A.H.; Friedman, H.S.; Herndon, J.E.; McLendon, R.E.; Pegram, C.N.; Provenzale, J.M.; et al. Novel Human IgG2b/Murine Chimeric Antitenascin Monoclonal Antibody Construct Radiolabeled with 131I and Administered into the Surgically Created Resection Cavity of Patients with Malignant Glioma: Phase I Trial Results. J. Nucl. Med. 2006, 47, 912–918. [Google Scholar]
  1096. Zalutsky, M.R.; Reardon, D.A.; Akabani, G.; Coleman, R.E.; Friedman, A.H.; Friedman, H.S.; McLendon, R.E.; Wong, T.Z.; Bigner, D.D. Clinical Experience with α-Particle–Emitting 211At: Treatment of Recurrent Brain Tumor Patients with 211At-Labeled Chimeric Antitenascin Monoclonal Antibody 81C6. J. Nucl. Med. 2008, 49, 30–38. [Google Scholar] [CrossRef] [PubMed]
  1097. Reardon, D.A.; Zalutsky, M.R.; Akabani, G.; Coleman, R.E.; Friedman, A.H.; Herndon, J.E.; McLendon, R.E.; Pegram, C.N.; Quinn, J.A.; Rich, J.N.; et al. A Pilot Study: 131I-Antitenascin Monoclonal Antibody 81c6 to Deliver a 44-Gy Resection Cavity Boost. Neuro Oncol. 2008, 10, 182–189. [Google Scholar] [CrossRef] [PubMed]
  1098. Hdeib, A.; Sloan, A. Targeted Radioimmunotherapy: The Role of 131I-chTNT-1/B mAb (Cotara®) for Treatment of High-Grade Gliomas. Future Oncol. 2012, 8, 659–669. [Google Scholar] [CrossRef]
  1099. Reulen, H.-J.; Poepperl, G.; Goetz, C.; Gildehaus, F.J.; Schmidt, M.; Tatsch, K.; Pietsch, T.; Kraus, T.; Rachinger, W. Long-Term Outcome of Patients with WHO Grade III and IV Gliomas Treated by Fractionated Intracavitary Radioimmunotherapy. J. Neurosurg. 2015, 123, 760–770. [Google Scholar] [CrossRef] [PubMed]
  1100. Heute, D.; Kostron, H.; von Guggenberg, E.; Ingorokva, S.; Gabriel, M.; Dobrozemsky, G.; Stockhammer, G.; Virgolini, I.J. Response of Recurrent High-Grade Glioma to Treatment with (90)Y-DOTATOC. J. Nucl. Med. 2010, 51, 397–400. [Google Scholar] [CrossRef] [PubMed]
  1101. Krolicki, L.; Bruchertseifer, F.; Kunikowska, J.; Koziara, H.; Królicki, B.; Jakuciński, M.; Pawlak, D.; Apostolidis, C.; Mirzadeh, S.; Rola, R.; et al. Prolonged Survival in Secondary Glioblastoma Following Local Injection of Targeted Alpha Therapy with 213Bi-Substance P Analogue. Eur. J. Nucl. Med. Mol. Imaging 2018, 45, 1636–1644. [Google Scholar] [CrossRef]
  1102. Królicki, L.; Bruchertseifer, F.; Kunikowska, J.; Koziara, H.; Królicki, B.; Jakuciński, M.; Pawlak, D.; Apostolidis, C.; Mirzadeh, S.; Rola, R.; et al. Safety and Efficacy of Targeted Alpha Therapy with 213Bi-DOTA-Substance P in Recurrent Glioblastoma. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 614–622. [Google Scholar] [CrossRef]
  1103. Królicki, L.; Bruchertseifer, F.; Kunikowska, J.; Koziara, H.; Pawlak, D.; Kuliński, R.; Rola, R.; Merlo, A.; Morgenstern, A. Dose Escalation Study of Targeted Alpha Therapy with [225Ac]Ac-DOTA-Substance P in Recurrence Glioblastoma—Safety and Efficacy. Eur. J. Nucl. Med. Mol. Imaging 2021, 48, 3595–3605. [Google Scholar] [CrossRef]
  1104. Mamelak, A.N.; Rosenfeld, S.; Bucholz, R.; Raubitschek, A.; Nabors, L.B.; Fiveash, J.B.; Shen, S.; Khazaeli, M.B.; Colcher, D.; Liu, A.; et al. Phase I Single-Dose Study of Intracavitary-Administered Iodine-131-TM-601 in Adults with Recurrent High-Grade Glioma. J. Clin. Oncol. 2006, 24, 3644–3650. [Google Scholar] [CrossRef]
  1105. Cordier, D.; Forrer, F.; Bruchertseifer, F.; Morgenstern, A.; Apostolidis, C.; Good, S.; Müller-Brand, J.; Mäcke, H.; Reubi, J.C.; Merlo, A. Targeted Alpha-Radionuclide Therapy of Functionally Critically Located Gliomas with 213Bi-DOTA-[Thi8,Met(O2)11]-Substance P: A Pilot Trial. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 1335–1344. [Google Scholar] [CrossRef] [PubMed]
  1106. Gill, M.R.; Falzone, N.; Du, Y.; Vallis, K.A. Targeted Radionuclide Therapy in Combined-Modality Regimens. Lancet Oncol. 2017, 18, e414–e423. [Google Scholar] [CrossRef] [PubMed]
  1107. Bolcaen, J.; Kleynhans, J.; Nair, S.; Verhoeven, J.; Goethals, I.; Sathekge, M.; Vandevoorde, C.; Ebenhan, T. A Perspective on the Radiopharmaceutical Requirements for Imaging and Therapy of Glioblastoma. Theranostics 2021, 11, 7911–7947. [Google Scholar] [CrossRef] [PubMed]
  1108. Dadgar, H.; Jokar, N.; Nemati, R.; Larvie, M.; Assadi, M. PET Tracers in Glioblastoma: Toward Neurotheranostics as an Individualized Medicine Approach. Front. Nucl. Med. 2023, 3. [Google Scholar] [CrossRef]
  1109. Ranjan, T.; Sengupta, S.; Glantz, M.J.; Green, R.M.; Yu, A.; Aregawi, D.; Chaudhary, R.; Chen, R.; Zuccarello, M.; Lu-Emerson, C.; et al. Cancer Stem Cell Assay-Guided Chemotherapy Improves Survival of Patients with Recurrent Glioblastoma in a Randomized Trial. Cell Rep. Med. 2023, 4, 101025. [Google Scholar] [CrossRef] [PubMed]
Figure 1. WHO diagnostic criteria for glioblastoma.
Figure 1. WHO diagnostic criteria for glioblastoma.
Ijms 25 02529 g001
Figure 2. Origin of glioblastoma. During normal embryonic development and in the adult brain, neural stem cells (NSCs) generate glial and neuronal cells. Glioma stem cells (GSCs) arise from NSCs, astrocytes, oligodendrocytes, or glial precursor cells through the activation of oncogenic pathways (inactivation of TP53, NF1 or PTEN). GSCs are described as slow-dividing or quiescent cells, with multilineage differentiation capacity that allows them to differentiate into GB cells and cells with astrocytic, neuronal, and endothelial features and even trans-differentiation abilities. In GB tumors, there exists a dynamic equilibrium between quiescent and proliferative GSCs, and between GSC populations and their lineage-committed counterparts (differentiated non-GSC) that can also dedifferentiate into stem-lineage GSCs. Created with BioRender.com, accessed on 24 January 2024.
Figure 2. Origin of glioblastoma. During normal embryonic development and in the adult brain, neural stem cells (NSCs) generate glial and neuronal cells. Glioma stem cells (GSCs) arise from NSCs, astrocytes, oligodendrocytes, or glial precursor cells through the activation of oncogenic pathways (inactivation of TP53, NF1 or PTEN). GSCs are described as slow-dividing or quiescent cells, with multilineage differentiation capacity that allows them to differentiate into GB cells and cells with astrocytic, neuronal, and endothelial features and even trans-differentiation abilities. In GB tumors, there exists a dynamic equilibrium between quiescent and proliferative GSCs, and between GSC populations and their lineage-committed counterparts (differentiated non-GSC) that can also dedifferentiate into stem-lineage GSCs. Created with BioRender.com, accessed on 24 January 2024.
Ijms 25 02529 g002
Figure 3. PTEN/PI3K/Akt/mTOR and RAS/RAF/MEK/ERK axis, and list of related chemotherapeutics clinically assayed against glioblastoma. Created with BioRender.com.
Figure 3. PTEN/PI3K/Akt/mTOR and RAS/RAF/MEK/ERK axis, and list of related chemotherapeutics clinically assayed against glioblastoma. Created with BioRender.com.
Ijms 25 02529 g003
Figure 4. Viruses modified to infect only GB cells or to specifically induce cancer cell elimination can be delivered either directly into brain tumors or intravenously. Created with BioRender.com. Accessed on 15 January 2024.
Figure 4. Viruses modified to infect only GB cells or to specifically induce cancer cell elimination can be delivered either directly into brain tumors or intravenously. Created with BioRender.com. Accessed on 15 January 2024.
Ijms 25 02529 g004
Table 1. microRNAs: role in GB, functions, and targets.
Table 1. microRNAs: role in GB, functions, and targets.
Tumor-SuppressingOncogenic
NameRegulationTargetsNameRegulationTargets
miRNA-7
[151,152,153,154]
Survival, proliferation, apoptosis, invasion, angiogenesisFAK, EGFR, Akt, c-KIT, TGFβ2, CDK6, Akt2, LRRC4, YBX1, CD24, MTDHmiRNA-10b
[148,155,156,157]
Survival, proliferation, apoptosis, invasion, stemness HOXD10, uPAR, RhoC, PTEN, TFAP2C, BCL2L11, CDKN1A
miRNA-34
[148,158,159,160,161,162,163,164,165,166,167]
Survival, proliferation, apoptosis, migration, invasion, stemness SIRT1, c-Met, Notch1/2, PDGFRA, Msi1, Akt and Wnt miRNA-21
[168,169,170,171,172,173,174]
Survival, proliferation, apoptosis, migration, invasion, chemoresistance HNRPK, TAp63, PDCD4, P53, TGFβ, MMPs, Ras/Raf, SPRY2, ANP32A, PTEN, SMARCA4, ERK, LRRFIP1
miRNA-128
[175,176,177,178,179,180,181,182,183,184,185,186]
Stemness, radioresistance, apoptosis, proliferation, angiogenesisP70S6K1, SUZ12, BMI1, PDGFRA, EGFR, E2F3a, WEE1 and Msi1miRNA-93
[179,187,188]
Survival, proliferation, angiogenesis, stemness Integrin b8
Abbreviations: Akt (Akt serine/threonine kinase), Akt2 (Akt serine/threonine kinase 2), BCL2L11 (BCL2 like 11), BMI1 (BMI1 proto-oncogene, polycomb ring finger), CDKN1A (cyclin dependent kinase inhibitor 1A), CDK6 (cyclin-dependent kinases), CD24 (CD24 molecule), c-KIT (receptor tyrosine kinase), c-Met (MET proto-oncogene, receptor tyrosine kinase), EGFR (Epidermal growth factor receptor), E2F3a (E2F transcription factor 3), FAK (Focal adhesion kinase), HOXD10 (homeobox D10), Integrin b8 (Integrin b8), LRRC4 (leucine rich repeat containing 4), MMPs (matrix metalloproteinases) miR (miRNAs), MTDH (metadherin), Msi1 (musashi RNA binding protein 1), Notch1/2 (notch ½), P70S6K1 (ribosomal protein S6 kinase B1), PDGFRA (platelet derived growth factor receptor alpha), PTEN (phosphatase and tensin homolog), RhoC (ras homolog family member C), SIRT1 (sirtuin 1), SUZ12 (SUZ12 polycomb repressive complex 2 subunit), YBX1 (Y-box binding protein 1), TGFβ2 (transforming growth factor beta 2), uPAR (urokinase-type-plasminogen-activator receptor, WEE1 (WEE1 G2 checkpoint kinase), Wnt (Wingless/Integrated).
Table 2. Phase III clinical trials that support the SOC in ndGB patients.
Table 2. Phase III clinical trials that support the SOC in ndGB patients.
Name/Trial NumberTreatmentNo./Type of PatientsOutcomeRef.
EORTC-NCIC
NCT00006353
RT
RT+TMZ (SOC)
573 ndGB patientsSOC had survival benefits in all groups, including patients ≥ 60 years
mOS: 12.1 m (RT) vs. 14.6 m (RT+TMZ)
OS (2,3,4,5 years): 10.9%, 4.4%, 3%, 1.9% (RT) vs. 27.2%, 16%, 12.1%, 9.8% (SOC)
[7,255]
EF-14
NCT00916409
SOC
SOC+TTFields
695 ndGB patientsThe addition of TTFields to the SOC extended mOS (16 vs. 20.9 m) and mPFS (4.0 vs. 6.7 m) [13,256]
NCT00304031RT+TMZ (SOC)
RT+TMZ (DD)
833 ndGB patients No difference was found in mPFS and mOS (16.6 vs. 14.9 m) between both arms.
GEINO14-01 (Spain) and EX-TEM (Australia) studies confirm the results of this trial
[257,258]
CeTeG/NOA-09 
NCT01149109
SOC+Lomustine
SOC
129 ndGB or gliosarcoma patients with +MGMTCombined treatment might improve mOS (48.1 vs. 31.4 m), but these findings should be interpreted with caution, owing to the small size of the trial[259]
AVAglio
 NCT00943826
SOC+BEV
SOC
921 supratentorial ndGB patientsThe addition of BEV to SOC increased in 4.4 mo. the mPFS but not mOS (16.8 vs. 16.7 m). Maintenance of baseline QoL and performance status were observed with BEV, but with higher rate of AE[260,261]
RTOG 0825
NCT00884741
SOC+BEV (adjuvant)
SOC
637 ndGB patientsThe addition of BEV to SOC increased the PFS in 7.3 m, without differences in mOS. Higher rates of neurocognitive decline, symptom severity, and decline in health related QoL affected patients treated with BEV + SOC.[262,263]
NCT00017147O6BG+BCNU+RT
BCNU+RT
179 ndGB or gliosarcoma patients The addition of O6BG did not provide benefit and caused additional toxicity. Significantly more grade 4/5 AE in the experimental arm.
mOS: 11 m (O6BG + BCNU + RT) vs. 10 m.
[264]
NOA-08-Trial
NCT01502241
TMZ (100 mg/m2)
RT (60 Gy, 2 Gy per fr.).
412 patients with anaplastic astrocytoma or ndGB ≥65 yearsTMZ or RT alone render similar results (mOS: 8.6 vs. 9.6 m).
+MGMT patients had better responses with TMZ, vs. −MGMT patients that better responded to RT.
[265]
ISRCTN81470623TMZ
Hypofractionated RT (34 Gy, 3–4 Gy, fr. over 2 weeks)
Standard RT (60 Gy, 2 Gy fr. over 6 weeks)
291 patients with ndGB > 60 yearsStandard RT was associated with poor outcomes, especially in patients > 70 years. Similar OS (8.4 vs. and 7.4 m) in TMZ vs. hypofractionated RT.
Both strategies should be considered as standard treatment options in the elderly. +MGMT status is a predictive marker for TMZ-derived benefits.
[266]
NCT00482677RT(40 Gy/15 fr.)+ TMZ
RT alone (40 Gy/15 fr.)
562 ndGB patients ≥ 65 yearsPFS (5.3 vs. 3.9 m) and (OS 9.3 vs. 7.6 m) were longer if TMZ was added to RT vs. RT alone. QoL was similar in both groups.[267]
Abbreviations: AE, adverse events; BCNU, carmustine; DD, dose-dense; fr., fraction; +MGMT, methylated MGMT promoter; -MGMT, unmethylated MGMT promoter; m, months; ndGB, new diagnosed glioblastoma; PFS, progression-free survival; O6-BG, O6-benzylguanine; OS, overall survival; QoL, quality of live; SOC, standard of care treatment; TMZ, Temozolomide; TTFields, tumor treating fields; RT, radiation therapy.
Table 3. Selected phase II/II trials for target therapies in GB.
Table 3. Selected phase II/II trials for target therapies in GB.
Trial NumberTreatment AssayedTrial InformationOutcomesRef.
NCT02977780Abemaciclib+SOC
Neratinib+SOC
CC-115+SOC
Phase II in ndGB with -MGMTAddition of abemaciclib or neratinib to the SOC was well-tolerated, but CC-115 associated with ≥grade 3 related toxicities in 58% of patients. An increase in mPFS was observed in the first two treatments and lack of efficacy in CC-115. None of the therapies demonstrated a benefit in OS.[402,403]
NCT00977431Afatinib
TMZ
Afatinib+TMZ
Phase II in rGBAfatinib monotherapy had limited activity, but it increased mPFS in patients overexpressing EGFR or expressing EGFRvIII. Addition of afatinib to TMZ did not improve PFS6 rate (3% vs. 23% vs. 10%) or mPFS. There were no significant differences in OS across the study arms.[404]
NCT03158389Alectinib, Idasanutlin, Palbociclib, Vismodegib, TemsirolimusPhase I/IIa in ndGB with -MGMTThe N2M2 trial is investigating a number of different targeted compounds in combination with RT. No results have been published yet.[405]
NCT02029573Atorvastatin+SOCPhase II in ndGBAtorvastatin did not improve PFS6. High LDL levels seem to be associated with poor outcomes.[406]
NCT03291314Axitinib+avelumabPhase II in rGBEfficacy of the combination is similar to axitinib monotherapy. Addition of avelumab does not show synergistic efficacy. Axitinib confirms its role as a potent corticoid-sparing option to control tumor-related edema.[407]
NCT01562197Axitinib
Axitinib+lomustine
Phase II for rGBAxitinib’s tolerability and clinical outcomes match those of BEV. However, axitinib offers advantages like oral dosing and a short half-life for quick reversibility in urgent interventions. Combined treatment had no benefits. mPFS6 was 26% vs. 17%.[408]
NCT00967330BEV during RT followed by BEV+IRIPhase II in ndGB with -MGMTBEV+ Irinotecan did not alter QoL and resulted in a superior PFS6 rate and mPFS compared with TMZ. [409]
NCT00345163BEV+Irinotecan
BEV
Randomized phase II in rGBBEV alone or in combination with irinotecan, was well tolerated and active in rGB. Estimated PFS6 rates were 42.6% and 50.3%, respectively; the mOS were 9.2 months and 8.7 months, respectively. [410]
NCT00817284BEV+irinotecan+SOC
BEV+SOC
Randomized phase II in ndGBResults did not indicate any benefit from BEV-irinotecan-SOC in first-line therapy as opposed to BEV-SOC in terms of response and PFS.[411]
NCT00979017BEV+irinotecan+TMZ in combination with RTPhase II for unresectable GBCombined treatment was tolerable leading to radiographic response in unresectable and/or subtotally resected GB.[412]
NCT01290939BEV+lomustine
BEV
Lomustine
Phase III in rGBBEV+lomustine improved mOS (12 vs. 8 vs. 8 months) compared to either monotherapy in the prior phase II trial. In this study, despite a somewhat prolonged PFS, there was no mOS advantage over lomustine alone (9.1 vs. 8.1 months).[377,387]
NCT01632228BEV+onartuzumabPhase II in rGBNo survival benefits were evidenced with the combined treatment. mOS was 8.8 months (BEV+onartuzumab) vs. 12.6 months (BEV+placebo).[413]
NCT01730950BEV+RTRandomized phase II in rGBRe-irradiation was shown to be safe and well tolerated. BEV + RT improved PFS6 rate but no difference in OS was found.[414]
NCT00667394BEV+tandutinibPhase II in rGB.The efficacy of combination therapy was comparable to that of BEV monotherapy, but more toxic. [415]
NCT00883298BEV+TMZPhase II in rGBCombined treatment showed efficacy and was well tolerated. The PFS6 and 12-month OS rates were 62.5% and 31.3%, respectively. mOS was 37.1 weeks. The benefits were similar to BEV monotherapy.[416]
NCT01738646BEV+vorinostatPhase II in recurrent grade 4 gliomaCombined treatment was well tolerated, but not improved PFS6 or mOS vs. BEV monotherapy.[217]
NCT01339052BuparlisibPhase II in rGB with PI3K pathway-activatedAlthough buparlisib achieved significant brain penetration, it had minimal efficacy in patients with PI3K-activated rGB.[417]
NCT01349660Buparlisib+BEVPhase II in relapsed/refractory GBThe combination was poorly tolerated, with frequent dose interruptions, and showed no greater efficacy than BEV alone[418]
NCT01934361Buparlisib+carboplatin
Buparlisib+lomustine
Randomized phase Ib/II in rGBNone of the combinations significantly improved the antitumor activity compared with historical data on single-agent carboplatin or lomustine. [419]
NCT01870726Buparlisib+INC280Phase Ib/II in PTEN-deficient GBNo improvement was evidenced in the combined treatment. [420]
NCT01062425Cediranib
Placebo
Randomized phase II in ndGBPFS6 was 46.6% vs. 24.5% (placebo). No difference in OS between the 2 arms. Cediranib had more AEs (≥ 3) than placebo. [421]
NCT00777153Cediranib
Cediranib+lomustine
Lomustine
Phase III in rGBCediranib monotherapy or in combination did not improve PFS or mOS (8.0, 9.4, and 9.8 months, for each group). Cediranib had corticosteroid-sparing effects and clinical activity on deterioration of neurologic function.[422]
NCT00311857Cetuximab+SOCPhase I/II in ndGBEarly data from trimodal therapy indicated feasibility without an increased toxicity profile. OS was 87% at 12 months. +MGMT was not associated with longer OS.[423]
NCT00463073Cetuximab+BEV+
irinotecan
Phase II for rGBPatients with an EGFR amplification lacking EGFRvIII expression had a significantly superior PFS (3.03 vs. 1.63 months) and OS (5.57 vs. 3.97 months). However, the efficacy was not superior to the combination BEV+irinotecan.[424,425]
NCT02974621Cediranib+olaparib
BEV
Randomized phase II for rGB.Treatment with cediranib + olaparib failed to increase PFS and OS in rGB patients.[426]
NCT01520870DacomitinibPhase II in rGB with EGFR amplification or EGFRvIIIDacomitinib had minimal activity but, a subset of patients (4.1%) experienced a durable, clinically meaningful benefit.[427]
NCT00423735DasatinibPhase II in target-selected patients with rGBDasatinib was ineffective in rGB[428]
NCT02343406Depatux-M+TMZ
Depatux-M
Lomustine or TMZ
Randomized phase II in rGB with EGFR amplificationOS: 19.8% vs. 5.2% (control group) vs. 10% (depatux-M). Depatux-M had no impact on QoL, except for more visual disorders.[429]
NCT02573324Depatux-M+SOCPhase III in ndGB with EGFR amplificationCombined treatment enhanced PFS (8.0 vs. 6.3 months), particularly among patients with EGFRvIII-mutant (8.3 vs. 5.9 months) or -MGM tumors. However, no significant OS benefits were evidenced. Most of depatux-M-treated patients had corneal epitheliopathy (61% grade 3–4).[430]
NCT01753713DovitinibPhase II in rGBDovitinib was not efficacious in prolonging the PFS irrespective of prior treatment with anti-angiogenic therapy.[431]
NCT02034110Dabrafenib+trametinibPhase II in BRAFV600E-rare cancersDabrafenib plus trametinib showed clinically meaningful activity (mOS 13.7 months) in high-grade glioma patients with BRAFV600E mutation. Limited significance of result due to the small sample.[432]
NCT00295815Enzastaurin
Lomustine
Phase III in rGBEnzastaurin was well tolerated and had a better hematologic toxicity profile than Lomustine but did not show superior efficacy.[433]
NCT00337883ErlotinibPhase II in rGB assessing if response was related to concomitant use of EIAEDsErlotinib had limited activity. However, in the EIAED subgroup, the PFS6 reached or exceeded historical survival values.[434]
NCT00086879Erlotinib
Control: TMZ or BCNU
Phase II in rGB
Erlotinib had insufficient single-agent activity. PFS6: 11.4% vs. 24.1% in the control arm. Both, PFS and OS were worse in the patients with EGFRvIII in the erlotinib arm.[435]
NCT00671970Erlotinib+BEVPhase II in rGB based on VEGF/EGFRvIII expressionCombined treatment improved mPFS (6.75 vs. 5.5 months) and mOS (17.0 vs. 6.75 months). The significance of the results is limited due to the small size of the cohort.[436]
NCT00720356Erlotinib+BEV (after completion of SOC)Phase II in -MGMT ndGB or gliosarcomaThe combination of erlotinib and BEV was tolerable but did not meet the primary endpoint of increased survival. [437]
NCT00187486Erlotinib+SOCPhase II in ndGB and gliosarcomaErlotinib plus SOC significantly improved OS (19.3 vs. 14.1 months) with respect to historical controls. There was a strong positive correlation between +MGMT and survival.[438]
NCT00274833Erlotinib+SOCPhase II in ndGBThe addition of erlotinib to SOC was not efficacious and had an unacceptable toxicity.[439]
NCT0062243Erlotinib+sirolimusPhase II in rGBErlotinib plus sirolimus was well tolerated but had negligible activity in rGB.[440]
NCT00445588Erlotinib+sorafenibPhase II in progressive and rGBErlotinib and sorafenib have significant pharmacokinetic interactions that may negatively impact the efficacy of the combination regimen.[441]
NCT01062399Everolimus+SOCPhase II in ndGBCombining everolimus with conventional SOC leads to increased toxicities and had no survival benefit[442]
NCT00805961Everolimus+BEV+SOCPhase II in ndGBThe addition of everolimus and BEV to SOC was feasible, deleting the maintenance with TMZ. The PFS compared favorably to previous reports with SOC but was similar to results achieved in other trials in which BEV was added to first-line treatment.[443]
NCT00250887GefitinibPhase II in rGBGefitinib reaches the tumor in high concentrations, efficiently dephosphorylates the target, but does not affect GB growth.[444]
NCT00016991GefitinibPhase II in GB at first relapseTolerable and modest activity. 56.6% of the patients suffered therapy failure within the initial 8-week assessment period.[445]
NCT01310855Gefitinib+ cediranib
Cediranib
Randomized phase II in rGBCombined treatment was tolerated and showed a tendency to improve PFS and response rates. mOS = 7.2 vs. 5.5 months in cediranib arm. Incomplete recruitment led to the study being underpowered.[446]
NCT00014170Gefitinib+RTPhase II in ndGBThe addition of gefitinib to RT was well tolerated, but there were no significant differences in PSF as compared to historical control cohorts treated with RT alone.[447]
NCT00171938Imatinib and/or Hypofractionated RTPhase II for inoperable/incompletely resected GB expressing PDGFRImatinib showed no measurable efficacy.[448]
NCT00154375Imatinib+hydroxyurea
Hydroxyurea
Phase III in TMZ- resistant progressive GBNo clinically meaningful differences were found between the two treatment arms (PFS6 5% vs. 7% in the monotherapy) and the primary study end point was not met.[449]
NCT01268566MEDI-575Phase II in rGBMEDI-575 was well tolerated but showed limited clinical activity[450]
NCT00753246Nimotuzumab+SOCRandomized phase III in ndGNimotuzumab was well tolerated, but its combination with the SOC had no significant survival benefits (22.3 vs. 19.6 months in SOC)[451]
NCT01227434Palbociclib
Palbociclib w/o resection
Phase II in RB1-positive rGBPalbociclib was administered 7 days prior to resection and the analysis of the surgical samples demonstrated tumor biologically effective concentrations, but there was no reduction in RB1 expression or decrease in cell proliferation, thus lacking efficacy.[399]
NCT00459381PazopanibPhase II in rGBPazopanib did not prolong mPFS (12 weeks) but showed in situ biological activity as demonstrated by the radiographic responses.[452]
NCT01051557PerifosinePhase II in rGBPerifosine is tolerable but ineffective as monotherapy for GB.[453]
NCT02926222Regorafenib
Lomustine
Randomized phase II in rGBmOS was significantly improved in the regorafenib group (7.4 months) compared with the lomustine group (5.5 months). Despite the criticisms of the study and the high rate of AEs, the encouraging results led regorafenib to become the SOC in Italy. [454]
NCT00621686Sorafenib+BEVPhase II in rGBNo improvement in the outcome of patients.[455]
NCT00597493Sorafenib+TMZ (LD) Phase II in rGBCombined treatment was feasible and safe, showing some efficacy. Patients: 12% partial response, 43% stable disease, 48% progression. PFS6 was 26% and mOS was 7.4 months. [456]
NCT00544817Sorafenib+SOCPhase II in ndGBThe addition of sorafenib did not improve the efficacy of SOC[457]
NCT00535379SunitinibPhase II in rGBMinimal anti-GB activity and substantial toxicity when sunitinib was given at higher doses. c-KIT expression in vascular endothelial cells was associated with improved PFS.[458]
NCT01100177SunitinibPhase II in non-resectable ndGBNo efficacy as monotherapy.[459]
NCT01100177Sunitinib+SOCPhase II in ndGB with -MGMT The addition of sunitinib modestly increased OS. Sunitinib potentially sensitizes -MGMT GB to adjuvant TMZ.[460]
NCT00022724TemsirolimusPhase II in rGBTemsirolimus was well tolerated, but there was no evidence of efficacy.[461]
NCT00016328TemsirolimusPhase II in rGBTemsirolimus was well tolerated. Radiographic improvement was observed in 36% of patients and was associated with significantly longer median time to progression.[462]
NCT01019434TemsirolimusPhase II in ndGB with -MGMT Temsirolimus showed no clinical benefit compared to TMZ. [463]
NCT00800917Temsirolimus+BEVPhase II in rGBTemsirolimus + BEV treatment was well tolerated but did not improve BEV monotherapy.[464]
NCT00821080VandetanibPhase I/II in recurrent malignant gliomaVandetanib did not have significant activity in unselected patients with recurrent malignant glioma. mOS was 6.3 months in the GB arm. Seizures were an unexpected toxicity of therapy. [465]
NCT00272350Vandetanib+SOCPhase II in ndGB or gliosarcomaThe addition of vandetanib was reasonably well tolerated. However, the regimen did not prolong OS (16.6 months) compared with SOC (15.9 months). [466]
NCT01026493VeliparibRandomized phase I/II in TMZ resistant rGBCombined treatment did not significantly improve PFS6 in the BEV-naïve or BEV-failure patients who were previously treated with TMZ.[467]
ACTRN12615000407594Veliparib+SOCRandomized phase II in ndGB with -MGMT Combined treatment showed no clinical benefit. PFS6: 46% (experimental arm) vs. 31% in SOC. mOS = 12.7 months (experimental arm) vs. 12.8 months (SOC). [468]
NCT02152982Veliparib+SOCPhase II/III in +MGMT ndGBThere was no significant difference in mOS (28.1 vs. 24.8 months in the SOC-treated group).[469]
Abbreviations: AEs, adverse events; BCNU, carmustine; BEV, bevacizumab; EGFR, epidermal growth factor receptor; EIAEDs, CYP3A4 enzyme-inducing antiepileptic drugs; GB, glioblastoma; LD, low dose; +MGMT, methylated MGMT promoter; -MGMT, unmethylated MGMT promoter; mOS, median overall survival; mPFS, median progression-free survival; ndGB, new diagnosed glioblastoma; QoL, quality of life; PFS6 progression-free survival at 6 months; rGB, recurrent glioblastoma; TMZ temozolomide; VEGFR vascular endothelial growth factor receptor; SOC, standard of care treatment; RT, radiotherapy.
Table 4. Selected phase II/III clinical trials on immunotherapy for GB.
Table 4. Selected phase II/III clinical trials on immunotherapy for GB.
Trial NumberTreatmentTrial InformationOutcomeRef.
Allogenic DC vaccinationPhase II for GB or grade 4 astrocytomaOS (27.6 ± 2.4 months) was 75% greater in the vaccinated GB group.[666]
NCT01213407Audencel (tumor lysate-charged autologous DCs)+SOCRandomized phase II in ndGBCombined treatment had no clinical benefits. PFS (28.4% vs. 24.5%) and mOS (≈18.3 months) did not differ significantly between control and vaccine groups.[667]
NCT01006044Autologous DCV (in tumor resection)+SOCPhase II in ndGBTreatment was feasible and safe. Increase in specific immune response (proliferation or cytokine production) was detected in 11/27 evaluated patients. No correlation between immune response and survival was found. mPFS and mOS were 12.7 and 23.4 months, respectively.[668]
NCT03400917AV-GBM-1Phase II in ndGBAV-GBM-1 treatment had numerous AEs affecting the CNS. mPFS was longer than historical benchmarks, but no mOS improvement was noted. mPFS and mOS from ITT enrollment were 10.4 and 16.0 months, respectively. Two-year OS was 27%.[669]
NCT00639639Cytomegalovirus Pp65+SOC+TMZ (DD)Randomized phase II in ndGBPatients showed markedly prolonged mPFS and mOS (25.3 and 41,1 months) compared with historical controls (8.0 and 19.2 months). Four patients remained progression-free 5 years later. [670]
NCT00045968DCVax-L+SOC (ndGB)
DCVax-L (rGB)
Phase III in progressive ndGB or rGBSignificant extension of survival compared with matched external controls.
mOS in ndGB (from randomization): 19.3 vs. 16.5 months
mOS in rGB (from relapse): 13.2 vs. 7.8 months.
[671]
NCT01567202DCV loaded with GSC antigensPhase II for ndGB or rGBExtension in OS to 13.7 months, up from 10.7 months. Low B7-H4 expression identified subgroups of GB patients more responsive to treatment.[672]
NCT02798406DNX-2401+ pembrolizumabPhase II rGBDNX-2401 followed by pembrolizumab was well tolerated with notable survival benefit in selected patients. mOS was 12.5 months; OS at 12 and 18 months was 54.5% and 20.8%, respectively.[673,674]
UMIN000001426 (Japan Clinical trials)Fractionated RT+ TMZ+AFTPhase I/II in ndGBThe treatment was well tolerated and resulted in favorable survival. mPFS, mOS, 2- and 3-year survival rates were: 8.2 months, 22.2 months, 47%, and 38%, respectively.[675]
UMIN000015995
(Japan Clinical trials)
G47∆ Herpes virusPhase II in rGB
G47∆ had a significant survival benefit and good safety profile. mOS was 20.2 months after G47∆ initiation. Biopsies revealed increasing numbers of tumor-infiltrating CD4+/CD8+ lymphocytes and persistent low numbers of Foxp3+ cells.[676]
2004-000464-28
(EU Clinical trials)
Ganciclovir+sitimagene ceradenovec+ SOCRandomized phase III trial in ndGBGanciclovir and sitimagene ceradenovec after resection can increase time to death or to re-intervention, although the treatment did not improve OS (16.32 vs. 14.85 in controls treated with SOC).[677]
NCT00293423HSPPC-96 vaccinePhase II in rGBHSPPC-96 vaccine showed considerable efficacy in terms of OS (10.65 months) and PFS (4.78 months). The improved outcomes were associated with the number of doses and a higher absolute lymphocyte count. [678,679]
NCT01814813HSPPC-96 vaccine+BEVPhase II in rGBThe study failed to demonstrate a survival benefit for patients treated with HSPPC-96 alone or in combination with BEV compared to BEV alone.[680]
NCT01280552ICT-107 DCVPhase II in ndGBPFS increased by 2.2 months, but no significant improvement in OS was shown. The HLA-A2 patient group had higher levels of immune response than the HLA-A1 patient group[681]
ISRCTN84434175Ipilimumab+SOCRandomized phase II in ndGBNo improvement in PFS or OS was observed with the addition of ipilimumab to the SOC.[682]
NCT02017717Nivolumab
BEV
Randomized phase III in rGBmOS (13.4 vs. 14.9 months) was comparable between nivolumab and BEV in rGB patients. The objective response rate was higher with BEV (23.1%) vs. nivolumab (7.8%).[663]
NCT02550249Nivolumab administered pre- and post-surgical intervention.Phase II for ndGB or rGB that required salvage surgical resectionNo obvious survival benefit was substantiated (mPFS and OS were 4.1 and 7.3 months, respectively, in evaluable patients,). Notably, 3 patients treated with nivolumab remained alive after long-term follow-up. [683]
NCT02667587Nivolumab+SOCPhase III for ndGB with +MGMTNivolumab added to SOC did not improve mOS (28.9 months vs. 32.1 months).[664]
NCT02617589Nivolumab+SOCPhase III in ndGB with -MGMTNivolumab added to SOC did not improve mOS (13.4 months vs. 14.9 months).[665]
NCT02337491Pembrolizumab vs. BEVPhase II in rGBPembrolizumab was well tolerated but was ineffective as monotherapy over BEV. [662]
NCT03405792Pembrolizumab+ TTFields+TMZ (maintenance) Phase II in ndGBThe triple combination was well tolerated and demonstrated promising efficacy. mOS was 24.8 vs. 14.7 months in controls. 2-year OS was 52.4% vs. 12.0% in controls. Several patients (40%) with measurable disease achieved a partial to complete response.[684]
NCT03532295Retifanlimab+ BEV+ Hypofractionated RTPhase II in rGBCombined treatment was well-tolerated and had an encouraging OS of 9 months (71.4%) and PFS at the time of data cutoff.[685]
NCT01498328Rindopepimut+ BEVRandomized phase II in rGBPFS6 was 27% and the mOS was 12 months, which means a significant improvement compared with controls (11% and 8.8 months, respectively)[686]
NCT00458601Rindopepimut+ SOCPhase II in ndGB expressing EGFRvIIIAnti-EGFRvIII antibody titers increased ≥4-fold in 85% of patients. PFS at 8.5 months from diagnosis was 66% and 3-year OS was 26%.[687]
NCT01480479Rindopepimut+
SOC
SOC
Randomized phase III in ndGB expressing EGFRvIII The addition of rindopepimut to SOC did not increase mOS, which was ≈20.0 months. [688]
NCT02455557SurVaxM+SOCPhase IIa in ndGBNo serious AEs were attributable to SurVaxM were shown. mPFS and mOS were 11.4 and 25.9 months, measured from first dose of SurVaxM. Specific immune response was evidenced.[689]
NCT02414165Toca 511+ flucytosine
Control: Lomustine, TMZ, or BEV
Randomized phase II/III in astrocytoma or rGBToca 511 + flucytosine did not improve OS (11.1 vs. 12.22 months in control) or other efficacy end points. [690]
NCT02511405VB-111+BEV
BEV
Randomized phase III in rGBPrevious phase II trial (NCT01260506) showed a significant improvement for VB-111 as monotherapy. On the contrary, here, dual administration of VB-111 and BEV failed to improve outcomes, and 67.5% of the patients developed grade ≥3 adverse reactions. The differences were attributed to changes in treatment regimen.[691,692]
Abbreviations: AEs, adverse events; AFT; autologous formalin-fixed tumor vaccine; BEV, bevacizumab; DCV, dendritic cell vaccine; EGFRvIII, epidermal growth factor receptor variant III; GB, glioblastoma; +MGMT, methylated MGMT promoter; -MGMT, unmethylated MGMT promoter; mOS, median overall survival; mPFS, median progression-free survival; ndGB, new diagnosed glioblastoma; QoL, quality of life; PFS6, progression-free survival at 6 months; rGB, recurrent glioblastoma; TMZ temozolomide; TTFields, tumor treating fields; SOC, standard of care treatment; RT, radiotherapy.
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

Obrador, E.; Moreno-Murciano, P.; Oriol-Caballo, M.; López-Blanch, R.; Pineda, B.; Gutiérrez-Arroyo, J.L.; Loras, A.; Gonzalez-Bonet, L.G.; Martinez-Cadenas, C.; Estrela, J.M.; et al. Glioblastoma Therapy: Past, Present and Future. Int. J. Mol. Sci. 2024, 25, 2529. https://doi.org/10.3390/ijms25052529

AMA Style

Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, et al. Glioblastoma Therapy: Past, Present and Future. International Journal of Molecular Sciences. 2024; 25(5):2529. https://doi.org/10.3390/ijms25052529

Chicago/Turabian Style

Obrador, Elena, Paz Moreno-Murciano, María Oriol-Caballo, Rafael López-Blanch, Begoña Pineda, Julia Lara Gutiérrez-Arroyo, Alba Loras, Luis G. Gonzalez-Bonet, Conrado Martinez-Cadenas, José M. Estrela, and et al. 2024. "Glioblastoma Therapy: Past, Present and Future" International Journal of Molecular Sciences 25, no. 5: 2529. https://doi.org/10.3390/ijms25052529

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