Next Article in Journal
Pregnant Women with Multiple Sclerosis: An Overview of Gene Expression and Molecular Interaction Using Bioinformatics Analysis
Previous Article in Journal
LINC00662 Promotes Aggressive Traits by Modulating OCT4 Expression through miR-335-5p in Gallbladder Cancer Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recurrent Glioblastoma—Molecular Underpinnings and Evolving Treatment Paradigms

1
Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
2
Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
3
Harvard Medical School, Harvard University, Boston, MA 02115, USA
4
Department of Neurosurgery, University of Rostock, 18055 Rostock, Germany
5
Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA
6
Division of Biology and Medicine, Brown University, Providence, RI 02912, USA
7
Department of Medicine and Surgery, Unicamillus University, 00131 Rome, Italy
8
Division of Pediatrics, Neuro-Oncology Section, MD Anderson Cancer Center, Houston, TX 77030, USA
9
David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2024, 25(12), 6733; https://doi.org/10.3390/ijms25126733
Submission received: 14 May 2024 / Revised: 13 June 2024 / Accepted: 15 June 2024 / Published: 19 June 2024

Abstract

:
Glioblastoma is the most common and lethal central nervous system malignancy with a median survival after progression of only 6–9 months. Major biochemical mechanisms implicated in glioblastoma recurrence include aberrant molecular pathways, a recurrence-inducing tumor microenvironment, and epigenetic modifications. Contemporary standard-of-care (surgery, radiation, chemotherapy, and tumor treating fields) helps to control the primary tumor but rarely prevents relapse. Cytoreductive treatment such as surgery has shown benefits in recurrent glioblastoma; however, its use remains controversial. Several innovative treatments are emerging for recurrent glioblastoma, including checkpoint inhibitors, chimeric antigen receptor T cell therapy, oncolytic virotherapy, nanoparticle delivery, laser interstitial thermal therapy, and photodynamic therapy. This review seeks to provide readers with an overview of (1) recent discoveries in the molecular basis of recurrence; (2) the role of surgery in treating recurrence; and (3) novel treatment paradigms emerging for recurrent glioblastoma.

1. Introduction

Glioblastoma is the most common and aggressive primary central nervous system (CNS) tumor. The current standard of care for newly diagnosed glioblastoma includes maximal safe resection followed by radiation therapy and temozolomide (TMZ) [1]. Moreover, regimens adding tumor treating fields (TTFs) are increasingly being considered standard of care [2]. Other variations in treatment protocols include differences depending on age and methylation status of O6-methylguanine-DNA methyltransferase (MGMT) [3,4,5]. However, despite well-established protocols for initial treatment and continued efforts to improve treatment options, the median overall survival for newly diagnosed glioblastoma remains poor at 15.6 months after initial treatment, with recurrence typically occurring within 6–9 months of initial diagnosis [6,7].
Maximal safe resection represents the cornerstone of current newly diagnosed glioblastoma treatment, demonstrating both a survival benefit and improved quality of life [8]. As such, new techniques have been developed to maximize resections such as intraoperative MRI or fluorescent guiding agents like 5-aminolevulinic acid (5-ALA), which preferentially accumulates in malignant tissues as protoporphyrin IX [9,10,11,12,13]. However, the complete removal of tumor cells is not possible with contemporary surgery as infiltrating glioblastoma cells persist in the brain, extending beyond the visible tumor [14,15,16]. In fact, postmortem studies have demonstrated that neoplastic cells extend beyond visible enhancement on imaging, making even supramaximal surgery non-curative [17]. Furthermore, as a consequence of initial chemoradiotherapy, residual tumor cells may undergo epigenetic modifications and acquire DNA mismatch repair mutations, which can induce resistance to subsequent therapy [18]. The resistance to chemotherapy and radiation and challenges concerning maximal safe resection continue to be core problems in treating recurrent glioblastoma.
Accordingly, this review first aims to summarize current research in recurrent glioblastoma such as the molecular pathways and genetic mutations that drive cell proliferation (TERT, PTEN, PI3K/Akt, MSH6, and LTBP4) and paracrine signaling of glioma stem cells (GSCs) [19,20]. Secondly, we discuss the advantages and limitations of resection in recurrent glioblastoma and alternatives including laser interstitial thermal therapy (LITT). Finally, we discuss novel treatment paradigms in recurrent glioblastoma including checkpoint inhibitors, chimeric antigen receptor T cell (CAR-T cell) therapy, and OV therapy.

2. Molecular Mechanisms of Recurrence

2.1. Genetic Mutations in Newly Diagnosed Glioblastoma and Recurrent Glioblastoma

To understand the genetic and molecular mechanisms of recurrent glioblastoma, the changes from newly diagnosed glioblastoma to recurrent glioblastoma must be considered. Although the total number of genetic mutations is reported to be relatively stable between newly diagnosed and recurrent glioblastoma [16,21], several dominant mutations at initial diagnosis differ compared with recurrence (Table 1). In recurrent glioblastoma, there is the preservation of clonal mutations such as telomerase reverse transcriptase (TERT) [22] and phosphatase and tensin homolog (PTEN) [23], with a relative loss of epidermal growth factor receptor (EGFR) mutations and amplification [21,24,25] and overexpression of platelet-derived growth factor receptor (PDGF-R). On the other hand, acquired hypermethylation of mismatch repair genes MSH6, LTBP4, and ALKBH5 are almost exclusively observed in recurrent glioblastoma post-initial treatment [21,26].
Gene alterations in TERT have also shown relative clonal stability between newly diagnosed glioblastoma and recurrent glioblastoma [21]. The TERT gene encodes the catalytic domain of telomerase and maintains telomere length and stability. TERT is normally not expressed in somatic cells but can be activated in various types of cancer, leading to uncontrolled proliferation [27]. In both newly diagnosed glioblastoma and recurrent glioblastoma, the TERT gene promoter mutation is commonly conserved, leading to an aggressive, highly proliferative phenotype [53]. Although there is a paucity of information on how TERT mutations could impact tumor recurrence, the presence of TERT promoter mutations is associated with other biomarkers relevant to tumor cell survival/progression such as EGFR amplifications (a dominant mutation in newly diagnosed glioblastoma prior to treatment) and PTEN mutations (dominant in recurrent glioblastoma) [54,55,56].
PTEN is a tumor suppressor gene with a high prevalence of loss in newly diagnosed glioblastoma, sustained at similar levels in recurrent glioblastoma [21,23,34,57,58,59]. PTEN catalyzes the hydrolysis of PIP3, which inhibits the activation of the downstream PI3K/Akt pathway, which in turn regulates cell cycle progression [60,61,62]. Thus, the loss of PTEN characteristically results in the loss of PI3K/Akt-dependent cell cycle inhibition, leading to uncontrolled cell proliferation [63]. These characteristics can also be attributed to the flux through the mammalian target of rapamycin (mTOR), a potent downstream effector of the PI3K/Akt that allows for protein translation, cell growth, proliferation, and survival [64]. An in vivo study by Zhang et al. showed that FK506-binding protein 12 (FKBP12) ligand co-administered with mTOR inhibitors enabled the retention and brain-specific action of mTOR inhibitors, leading to suppressed tumor growth and improved survival without on-target side effects [65]. Upstream of mTOR, the PI3K/Akt pathway is one of the major pathways implicated in the survival, proliferation, invasion, and migration of glioblastoma cancer cells [25]. Studies have identified PI3K-activating mutations in 67–82% of newly diagnosed glioblastoma [34,66] and 91% of recurrent glioblastoma [24,34], indicating a highly conserved pathway of cell proliferation [35]. Multiple pre-clinical studies inhibiting PI3K/Akt substrates have yielded positive treatment results in vitro [67,68]. Furthermore, long non-coding RNAs (lncRNAs) upregulate PI3K/Akt signaling [36,69]. The inhibition of lncRNAs such as LINC01426 and the knockdown of long stress-induced non-coding transcript 5 gene (LSINCT5) inhibit recurrence by attenuating the migratory ability of GSCs via inhibition of PI3K/Akt signaling pathways [70]. PI3K/Akt is not only directly upregulated in GCS lines but also a significant player in the TME. For example, glioblastoma cell lines can repurpose extracellular vesicles to increase proliferation and cell migration via the upregulation of PI3K/Akt [37], also promoting recurrence.
EGFR was the first described transmembrane receptor tyrosine kinase, which results in the activation of downstream pathways regulating proliferation, angiogenesis, migration, survival, and cell differentiation [71,72]. EGFR gene amplifications are present in more than half of glioblastoma [73,74], while EGFRvIII, a truncated receptor resulting from the deletion of exons 2 to 7, is the most common EGFR oncogenic mutation in glioblastoma [75]. EGFRvIII renders the receptor unable to bind any ligand [30], but it is usually co-expressed with the wild-type EGFR. Co-expression results in an autocrine loop that induces glioblastoma cells to produce both the receptor and the ligand [76]. EGFR/EGFRvIII downstream signaling includes constitutive activation of PIP3K/Akt, RAS/MAPK, and Bcl-Xl and has been implicated in glioblastoma cell proliferation, inhibiting apoptosis and enhancing glioblastoma invasiveness [31]. EGFRvIII-specific activation of dedicator of cytokinesis 1 (DOCK1) mediates cell growth and migration [32], whereas EGFRvIII-specific activation of SRC family kinases promotes cell survival in low-energy states [33]. EGFR’s most important ligands in glioblastoma include epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α) [74].
The overexpression of growth factor receptors plays a significant role in glioblastoma recurrence through glioma stem cell renewal and maintenance. In addition to EGFR, PDGF-R also serves as an important gene often overexpressed in newly diagnosed and recurrent glioblastoma [77]. In a study by Kim et al., the researchers observed that the high expression of PDGF-R β in GSCs led to increased expression of SOX-2 and decreased glial fibrillary acid protein (GFAP), demonstrating maintenance of a stem cell-like state in GSCs. In further experiments, Kim et al. used fluorescence-activated cell sorting (FACS) and showed that the inhibition of PDGF-R β resulted in decreased proportions of GSCs in the S phase of the cell cycle [38]. These experiments jointly suggest the important role of PDGF-R β signaling in GSC maintenance. Similarly, recent research by Lane et al. determined that the inhibition of PDGF-R α/β resulted in the outgrowth of neurites in glioblastoma and GSC cell lines [39]. The inhibition of PDGF-R serves as a potential therapeutic target to disrupt a persistent stem-cell-like state, deterring recurrence.
Although there is significant concordance in the genetic makeup between primary and recurrent glioblastomas, several mutational variations have been established between the two. Importantly, recurrent glioblastomas acquire new mutations following initial chemoradiation, including mutations in DNA mismatch repair (MMR) genes MSH6, MLH, and LTBP4 [78]. Indraccolo et al. [78] compared the expression of MMR proteins and matched whole exome sequences of newly diagnosed glioblastoma and subsequent recurrent glioblastomas in 57 patients. In 30% of the recurrent glioblastoma samples, a decrease in MMR protein expression was observed, with concurrent total loss, partial loss, and/or loss of function of MSH2 or MSH6 gene expression. Wang et al. [21] confirmed these results in 65 matched newly diagnosed glioblastoma and recurrent glioblastoma transcriptome and whole exome samples from six international sites from TCGA. All hypermutated samples were observed in recurrent glioblastoma after treatment with TMZ, with 94% acquiring the following mutations absent in matched newly diagnosed glioblastoma samples: MSH2 and MSH6 [79]. Understanding these changes is crucial for targeting recurrent glioblastoma. This acquired resistance to alkylating chemotherapy secondary to MMR MSH6 gene mutation was rescued by downstream peroxisome proliferator-activated receptor (PPAR) inhibition of DNA repair—a targeted therapy for TMZ-resistant recurrent glioblastoma [79]. Also, recurrent glioblastomas often harbor mutations in the latent TGF-binding protein 4 (LTBP4) gene, commonly absent in newly diagnosed glioblastoma-matched samples [21]. The LTBP4 gene drives the expression of proteins upstream of TGF-β that play a role in glioblastoma cell proliferation and migration. In rare cases, the BRAFv600E mutation has been implicated in epithelioid glioblastomas [80]; in patients with recurrence harboring such mutations, BRAF inhibitors have shown promise [81].

2.2. Epigenetic Modifications in Recurrent Glioblastoma

Given the high concordance between genetic mutations in newly diagnosed glioblastoma and in recurrent glioblastoma, increasing emphasis is being placed on transcriptional and epigenetic changes as drivers of recurrence [16] (Figure 1). Epigenetic modifications—like DNA methylation and histone acetylation events—can impact tumor behavior through mechanisms like controlling clonal gene expression [82,83]. For example, exposure to paracrine IFN-γ signaling results in epigenetic changes in GSCs [84] and hybrid glioblastoma-tumor-associated macrophage (TAM) cells with anti-inflammatory markers [85,86]. Histone deacetylase 8 (HDAC8) regulates natural killer (NK) cell cytotoxic activity and contributes to a hypoimmunogenic environment [87], while the deletion of ALKBH5 in hypoxic TME results in the recruitment of anti-inflammatory TAMs [20,88]. ALKBH5 is an N6-methyladenosine demethylase that results in increased oncogene expression responsible for glioblastoma proliferation. Dong et al. [48] demonstrated that the absence of demethylase activity results in the inhibition of CXCL8 expression (a neutrophil chemoattractant), subsequent IL-8 paracrine signaling, and dampening of pro-inflammatory TAMs. Similarly, Zhang et al. [49] demonstrated that ALKBH5 demethylase activity results in patient-derived glioblastoma cell proliferation, which is suppressed when ALKBH5 is silenced. The tumorigenic effects of ALKBH5 also appear to induce a TMZ-resistant state via the demethylation of SOX2 transcripts, resulting in increased SOX2 expression and Wnt5a/b glioblastoma proliferation signaling.
An extensively studied epigenetic mechanism that regulates disease progression in glioblastoma is MGMT, which encodes the DNA repair protein O6-alkylguanine (O6-AG) DNA alkyl transferase (AGT) [89]. The MGMT promoter has been an important target in both newly diagnosed glioblastoma and recurrent glioblastoma as the protein product, AGT, yields resistance to alkylating chemotherapy such as TMZ [51,89]. Hypermethylation of MGMT promoter results in silencing and decreased expression of AGT. Consistently, numerous studies and trials have demonstrated a positive effect of MGMT promoter methylation in response to initial treatment with TMZ in newly diagnosed glioblastomas [3,4,5,50].
Tyrosine kinase inhibitors can cause GSCs to transition into a slow-cycling, persistent state wherein Notch signaling-dependent developmental programs are upregulated [90]. In the persistent GSC state, H3K27 methyltransferase is downregulated, while H3K27 demethylases of KDM6A and KDM6B are upregulated. KDM6A/B are reported to be essential for persistent GSC survival and formation, as KDM6A/B knockouts demonstrate growth reduction in patient-derived persistent glioblastomas. However, upon transition to the persister state, H3K27 acetylation in GSC is associated with a reduction in H3K27me3 and increases in gene expression near enhancer-like elements. These findings indicate potential therapeutic avenues for persistent GCS that exploit epigenetic shifts in methylation and acetylation patterns [90].

2.3. TME Facilitating Glioblastoma Recurrence

Although intrinsic molecular drivers play an important role in tumor behavior, glioblastoma progression is also significantly impacted by the surrounding microenvironment. Although glioblastomas are considered “cold tumors” because of their highly immunosuppressive state, the tumor proper consists of 30–40% non-cancer immune cells [91]. Up to half of these are TAMs—microglia-derived (mgTAM) and monocyte-derived (moTAM) [78,92,93,94]—and the remainder are dendritic cells, T cells, NK cells, and neutrophils [83]. The rate of recurrence and proliferation of glioblastoma has been positively associated with the quantity and phenotype of TAMs, which in turn modulate the activity of T cells, dendritic, and NK cells in the TME [95]. The interaction between GSCs and TAMs in the TME is complex and consists of initial recruitment from the periphery via GSC release of cytokines such as chemokine ligand 2 (CCL2) and colony-stimulating factor 1 (CSF-1) signaling [40,41,42,43]. Once macrophages are within the vicinity of the tumor, they polarize along a pro-inflammatory versus anti-inflammatory axis via a combination of paracrine signals released by GSC and other cells within the TME [96]. IL-10, IL-6, and TGF-β drive polarization of the TME into the anti-inflammatory M2 phenotype, while IFN-γ and STAT1 activity promote a pro-inflammatory M1 phenotype [26,44,95,97,98,99,100].
GSCs and glial cells in the TME release large quantities of TGF-β and IL-10 [44,45,46], enhancing the immunosuppressed environment. Anti-inflammatory TAMs in turn dampen lymphocyte reactivity by inhibiting Th-1 and NK response and infiltration, which enhances Th-2 and Treg activity. This phenomenon is sustained by GSC-inducing genetic changes in TAMs that result in the release of immunosuppressive adenosine [47] and expression of surface molecules that inactivate T cells [83,101]. The cumulative effect is a chronic immunosuppressed state where glioblastoma cells acquire multiple epigenetic modifications and contribute to the hypoimmunogenic TME favorable for survival, proliferation, and resistance to treatment. By understanding and manipulating various components of the TME, glioblastoma treatments can aim to evade immunosuppression to prevent recurrence or treatment resistance.
As neovascularization contributes to maintaining GSCs in the TME, the upregulation of angiogenic elements in glioblastoma has also emerged as an important yet complex player in tumor recurrence and progression. A study in patient-derived glioblastoma cells found that tumor cells supported angiogenic sprouting in hypoxic conditions, forming co-localized networks of sprouting models [102]. One proposed pathway for this occurrence is the upregulation of hypoxia-inducible factor 1 (HIF-1) signaling in the hypoxic TME, releasing proangiogenic growth factors VEGF and basic fibroblast growth factor (bFGF) [103,104]. On the other hand, the upregulation of histamine signaling by GSCs in the TME has also been shown to trigger an increase in the H1-Ca2+-NF-kB axis independent of VEGF activation. In vivo studies with glioblastoma xenograft mice models revealed that pharmacological blockage of H1R using antihistamines impeded the growth of glioblastoma, establishing a potential therapeutic target in the TME for glioblastoma [105]. While it has been well established that neovascularization enables nutrient transport to the TME to promote GSC maintenance, leading to recurrence, the various pathways that directly link angiogenesis to tumor recurrence warrant further investigation.

3. The Role of Surgery in Recurrent Glioblastoma

Glioblastoma cells invade the brain parenchyma diffusely, making complete resection of tumor cells impossible [106,107,108]. Reoperation may be an option for select patients based on recurrence location, preoperative estimate of maximal safe resection of T1 contrast-enhancing disease, preoperative functional status and symptoms, and careful weighing of the risks and benefits [109,110,111,112]. Good candidates for repeat surgery include young patients, Karnofsky Performance Score (KPS) > 70, few comorbidities, and non-eloquent tumor location [113]. However, even in patients who qualify for surgery, the determination of safe and efficacious resection borders often poses an additional challenge because of unspecific FLAIR signals and the limitations of imaging [114]. Although some retrospective studies have reported beneficial outcomes after reoperation, there is no consensus regarding the role of surgery in recurrent glioblastoma [115,116,117]. As such, reoperation in recurrent glioblastoma remains a decision made on a case-by-case basis with multidisciplinary discussions among neurosurgeons, neurooncologists, radiation oncologists, and patients [118].
Multiple retrospective studies suggest a surgical benefit in overall survival (OS) in patients with recurrent glioblastoma. Djamel-Eddine et al. [119] assessed outcomes in 2005–2014 and reported significant survival benefits of repeat surgery; the median OS for patients undergoing one, two, and three surgical procedures were 11, 16, and 18 months respectively. However, while adjuvant treatment after the first surgery was standardized using the temozolomide and radiotherapy protocol [1], there was no standard protocol for subsequent resections, significantly confounding the conclusions regarding the benefit of reoperation on OS in patients with recurrent glioblastoma. In addition, key confounding variables including MGMT methylation status were not accounted for [119]. In another study of 97 patients who underwent reoperation for recurrent glioblastoma, Yong et al. [120] reported an improved median OS of 12.4 months after surgery for recurrent glioblastoma. This effect, though, was confounded by variance in post-operative residual tumor size. Similar to the work by Djamel-Eddine et al. [119], patients in this cohort who underwent repeat surgery were also carefully selected, which introduced selection bias. Nevertheless, a larger retrospective cohort of 503 patients, allowing for multivariate analysis, by Ringel et al. [107] demonstrated the prognostic benefit of surgery in recurrent glioblastoma. Finally, a recent study by Karschnia et al. [121] investigated the prognostic value of surgery at recurrence in 681 patients with recurrent glioblastoma. The study found a survival benefit in patients undergoing resection (11 months), compared with those that did not (7 months), after stratifying for potential clinical confounders [121].
On the other hand, numerous studies have pointed to the lack of benefit of repeat surgery for recurrent glioblastoma. González et al. [112] completed a 15-year retrospective analysis of 350 patients with recurrent glioblastoma. The authors stratified patients eligible for repeat surgery into one of two groups as follows: (1) those who underwent reoperation and adjuvant chemotherapy and (2) those treated only with chemotherapy. Patients who underwent reoperation with adjuvant chemotherapy had a longer median OS of 25 months vs. 17 months compared with the non-surgical arm. Univariate analysis revealed that six cycles of TMZ treatment as well as reoperation impacted OS; however, statistical significance was absent when the most significant covariates were adjusted for [112]. These results were confirmed by Franceschi et al. in a cohort of 232 patients with recurrent glioblastoma [116]. Despite the prolonged median OS in patients who underwent surgery (25.8 vs. 18.6 months), MGMT methylation, age, and progression-free survival (PFS) at 6 months were more strongly correlated with OS than secondary surgery. Nava et al. [111] reported similar findings among 764 patients with recurrent glioblastoma. Their results demonstrated that radiotherapy with temozolomide was the strongest predictor of OS and PFS in recurrent glioblastoma, while reoperation had a minimal effect [111]. Despite conflicting results, most clinicians agree that there is a benefit for reoperation in carefully selected patients [122,123,124,125].
Several factors are important to consider when performing a reoperation on recurrent glioblastoma including the functional status of the patient (i.e., KPS), mass effect, possible preservation of functional tissue (e.g., motor pathways, language pathways), and, crucially, the anticipated extent of resection. Importantly, Suchorska et al. reported that incomplete tumor resection was associated with worse post-recurrence survival compared with no surgery (6.5 months vs. 9.8 months) [126]. Bloch et al. [115] corroborated these findings in a cohort of 107 patients, defining gross-total resection (GTR) as the removal of 95% or greater of tumor volume and subtotal resection (STR) as the removal of less than or equal to 95% of tumor volume. Patients who underwent GTR at recurrence had a significantly greater median survival of 19.0 months compared with those who underwent STR with a median survival of 15.9 months [115]. Of note, defining the extent of resection (EOR) is critical for understanding the relationship between the amount of tumor removed and associated survival benefit. Oppenlander et al. [110] performed a study on 170 patients with recurrent glioblastoma and found a significant survival advantage with as little as 80% EOR, with additional recursive partitioning analysis demonstrating a continued benefit to OS with the highest levels of EOR. Karschnia et al. [121] reported that a residual contrast-enhancing volume threshold ≤1 cm3 compared with >1 cm3 was associated with increased survival. Furthermore, their study also found an exponential increase in hazard ratios for death with higher residual contrast-enhancing tumors. Future studies are required to investigate survival outcomes based on different amounts of tumor resection to discriminate the threshold at which patients begin to experience a survival benefit after reoperation for recurrent glioblastoma.
Patient age is another significant factor when considering reoperation for recurrent glioblastoma. Woernle et al. [117] performed a retrospective analysis of 98 patients who underwent initial surgical resection followed by radiotherapy with concomitant TMZ. The authors reported that repeat surgery resulted in improved OS in the younger group. Neville et al. [127] reported similar findings in a group of 286 patients who had adjuvant carmustine or TMZ. OS at 6, 12, and 24 months was greater for patients who also underwent reoperation for recurrent glioblastoma [127]. However, OS was confounded by younger age, higher KPS, the type of initial management, and reoperation extent. As previously mentioned, a major limitation of these studies is selection bias—patients who undergo repeat surgery are typically younger and otherwise healthier. Furthermore, age influences multiple important prognostic molecular markers such as TERT mutation and response to chemotherapy.
Fluorescence-guided surgery (FGS) using 5-ALA represents a recent promising development in the surgical resection of glioblastoma. FGS resulted in double the rates of GTR in newly diagnosed glioblastoma in a first phase III trial evaluating FGS for brain tumors (65% fluorescence group vs. 36% white light group) [9]. The pro-drug 5-ALA is administered orally to patients just prior to surgery, which leads to the accumulation of a red fluorescent agent, protoporphyrin IX (PpIX). The use of intraoperative microscopes modified for fluorescence imaging, including a violet excitation light and long pass filters to collect the emitted red fluorescence from PpIX, enables neurosurgeons to visualize the PpIX that accumulates in tissues [128,129]. In addition, 5-ALA has been used in recurrent glioblastoma to aid in the visualization of tumor tissue [129,130,131,132,133,134,135]. For example, Hickman et al. analyzed 63 recurrent glioblastoma tumors, of which 84.1% (53/63) showed visible PpIX fluorescence, with most non-fluorescent tumors being isocitrate dehydrogenase (IDH) mutants [136]. Although 5-ALA FGS is considered safe, PpIX fluorescence can be found in reactive-only tissue without the presence of tumor cells in recurrent glioblastoma [136,137]. Accordingly, surgeons should be aware of such false positives, as one study found that 13 out of 313 patients with recurrent glioblastoma demonstrated positive 5-ALA-PpIX fluorescence in reactive-only tissue. Although 5-ALA-PpIX FGS can serve as a powerful adjunct in recurrent glioblastoma cases to help surgeons maximize the extent of resection, like every tool, the surgeon should know the limitations of the technology [137].
The surgical decision to reoperate is heavily guided by factors such as patient age, KPS scores, comorbidities, tumor location, and pre-operative estimation of expected residual contrast enhancement (i.e., ideally < 1 cm3) meanwhile limiting post-operative deficits. Despite the various nuances in determining the efficacy and outcomes of reoperation, most studies showed that younger patients and those with fewer comorbidities have the most favorable reoperation outcomes.

4. Evolving Treatment Paradigms for Recurrent Glioblastoma

Current guidelines for recurrent glioblastoma management recommend enrollment in a clinical trial [138] and follow-up with MRI at 3- to 6-month intervals up to 2 years and then every 6 to 12 months up to the 5-year mark [132]. This section discusses evolving treatment paradigms in recurrent glioblastoma, including immune checkpoint inhibition, cellular therapies, oncolytic viral therapy, novel delivery methods, LITT, photodynamic therapy, and intratumoral microdevices.

4.1. Checkpoint Inhibitors

Several immune checkpoint inhibitors targeting T cell-mediated immunity have shown promise in non-CNS solid tumors. The most relevant T cell checkpoints are the programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) as well as the cytotoxic T-lymphocyte-associated (CTLA) protein axes [139]. These pathways are exploited by glioblastoma tumors to dampen T cell activation, cytotoxicity, and proliferation [140].
Nivolumab and pembrolizumab are two PD-1 receptor blockers that prevent tumor interaction with PD-L1, a protein that inactivates T lymphocytes (Table 2 and Table 3). Cloughesy et al. 2019 and Schalper et al. 2019 demonstrated an OS benefit and increased T-cell receptors among tumor-infiltrating T-lymphocytes, respectively, following nivolumab administration [141,142]. However, in the CheckMate 143 phase 3 trial, published the following year, nivolumab monotherapy compared to bevacizumab in recurrent glioblastoma revealed no improvement in OS [143]. Similarly, the combination of nivolumab and pembrolizumab with/out concurrent bevacizumab yielded no survival benefit [143,144]. Interestingly, in a phase I clinical trial of combination therapy with nivolumab and a regulatable interleukin-12 gene therapy, veledimex, Chiocca et al. [145] demonstrated an increased OS for the combination therapy arm (16.9 months vs. 9.8 months), mediated by increased IFN-γ. Similarly, Lee et al. [146] found that neoadjuvant PD-1 inhibition leads to enhanced IFN-γ signaling and upregulation of CTLA-4, shifting the TME from anti-inflammatory to pro-inflammatory, recruiting cytotoxic T cells. A trial by Nassiri et al. published in Nature Medicine in 2023 combined the oncolytic virus DNX-2401 and intravenous pembrolizumab, demonstrating encouraging results in recurrent glioblastoma therapy. In this trial, 56.2% of the patients had stabilized or improved disease, with a median overall survival of 12.5 months. Three patients in the treatment arm had a durable response, remaining alive at 45, 48, and 60 months [147].
While trials such as CheckMate 143 show a statistically non-significant survival benefit [143], checkpoint inhibitors target a well-described mechanism in recurrent glioblastoma and still present as a promising combination therapy option for recurrent glioblastoma. Clinical trials investigating the concurrent CTLA-4 and anti-PD-1 treatment of recurrent glioblastoma (NCT04606316) are currently ongoing.

4.2. CAR-T Cell Therapy

Chimeric antigen receptors (CARs) are formed through genetic engineering of T cells against specific tumor antigens (Table 4). The unique benefit of CAR-T cell therapy is T cells’ ability to bypass antigen presentation and required co-stimulatory activation, avoiding tumor-specific ignorance [150]. A variety of CAR-T cell therapies have emerged with targets including EGFR/EGFRvIII, IL-13Rα2, and B7-H3 [151]. EGFR/EGFRvIII mutations contribute to increased proliferation, decreased apoptosis, and tumor cell survival in 30–50% of recurrent tumor glioblastoma [76,152]. Despite promising preclinical studies, clinical trials of EGFRvIII-specific CAR-T cell therapy have demonstrated limited efficacy [153,154,155]; however, there have been some favorable reports. A case study by Durgin et al. [156] reported 36-month survival after CAR-T cell EGFRvIII therapy for recurrent glioblastoma. At 29 months, a follow-up MRI brain demonstrated decreased tumor enhancement with persistent peripheral T cell circulation. A similar result was reported by O’Rourke et al. [153], where peripheral expansion of EGFRvIII CAR-T cells resulted in a drop in tumor antigen among five of seven patients who had EGFRvIII CAR-T cell infusions in recurrent glioblastoma enhancing cavities. Additionally, the combination of EGFRvIII CAR-T cells with the immune checkpoint inhibitor pembrolizumab has been demonstrated to be safe, yet not efficacious [157]. An alternative target of CAR-T cell therapy is the IL-13Rα2, expressed almost universally in recurrent glioblastoma. It is a decoy receptor that binds IL-13, inhibiting an immune response [158,159]. IL-13Ra2 is associated with the activation of the PI3K/Akt pathway which mediates tumor progression in recurrent glioblastoma [159,160,161]. Brown et al. recently published phase 1 results using IL-13Rα2 targeting CAR-T cells in 65 patients [162]. An OS of 7.7 months from the time of recurrence was reported; one arm using a refined manufacturing platform delivering IL-13Rα2 targeting CAR-T cells both intraventricularly and intratumorally demonstrated a superior OS of 10.2 months. Finally, interim results from two CAR-T constructs targeting two antigens (EGFR and EGFRvIII by Choi et al. [163] and EGFR and IL-13Rα2 by Bagley et al. [164]) demonstrated rapid tumor regression within days and an acceptable safety profile.

4.3. NK Cell Therapy

NK cell therapies have emerged as a novel treatment serving as an alternative to CART-T therapy [165] (Table 5). NK cells are part of the innate immune system, allowing them to eliminate aberrant cells independent of the T cell activation signal and prevent CAR antigen loss-induced immune escape [166]. Murakami et al. [167] showed that in glioblastoma tumors expressing EGFRvIII growth factors, NK cell lines (CAR-KHYG-1) specific for EGFRvIII could effectively inhibit glioblastoma cell growth via apoptosis. Furthermore, Zhang et al. reported complete lysis of all the ErbB2 cell lines of LN-319, LNT-229, and LN-428 cells by NK-92/5.28.z cells. In in vivo mouse studies, potent antitumor activity and symptom-free survival were demonstrated following stereotactic injection of NK-92/5.28.z. In addition, local therapies of NK cells resulted in robust immune responses curing transplanted syngeneic glioblastoma in four/give immunocompetent mice carrying subcutaneous tumors and five/eight mice carrying intracranial tumors. Impressively, the NK cell injections also led to tumor-specific immunity and memory (IgG titers) after reinjection of glioblastoma cells (ERbB2/GL261) into distal sites, indicating longer-term protection against tumors [168]. With encouraging results in these preclinical studies, NK cell therapy trials are now underway [165].

4.4. Oncolytic Virotherapy

OVs utilize viruses to infect tumor cells resulting in cell lysis and release of tumor antigens, activating an immune response [169] (Table 6 and Table 7). Direct intratumoral delivery of OVs allows for bypassing of the BBB [170]. In a phase 2 clinical trial, Todo et al. tested a triple mutated, third-generation oncolytic HSV type 1 (HSV1, G47∆) in 19 adult patients with recurrent glioblastoma [171]. OV was administered intratumorally in six doses, which resulted in increased tumor-infiltrating CD4+/CD8+ lymphocytes on biopsy, suggesting a targeted immune response against glioblastoma tumor antigens [171]. The patient median OS was 20.2 months after OV therapy and 28.8 months after initial surgical resection, leading to the approval of G47∆ in Japan [171,172].
In a phase I trial, Friedman et al. demonstrated that stereotactic intratumor infusions of a genetically engineered HSV-1, G207 were safe and led to an OS of 12.2 months among 12 children with recurrent high-grade glioma [176]. Evaluation of the TME on biopsy pre- and post-treatment suggested a strong pro-inflammatory T cell response with clusters of T cells sustained between 2 and 9 months post-treatment. Moreover, there was increased T cell infiltration in areas distant from inoculation [176]. More recently, Ling et al. demonstrated that treatment with the oHSV CAN-33110 was associated with changes in tumor T cell counts and clonal diversity, peripheral expansion/contraction of certain T cell clonotypes, and unique tumor transcriptomic signatures of immune activation in a phase 1 trial in recurrent high-grade gliomas [173].
OV combination therapy is of increasing research interest [187]. Nassiri et al. [147] tested a combination therapy of the oncolytic adenovirus DNX-2401 with a checkpoint inhibitor in a cohort of 49 patients with recurrent glioblastoma. A single-dose intratumor delivery of DNX-2401 was followed by intravenous pembrolizumab. The median OS was 12.5 months, greater than the OS after previously reported monotherapies of DNX-2401 (OS: 9.3 months) or PD-1 inhibitors (OS: 9.8 months) [143,181]. Patients showed increased density of immune cells—microglia, macrophages, and CD3+, CD4+, or CD8+ T cells—infiltrating the TME [147]. More importantly, two patients who completed 6 months of pembrolizumab after OV inoculation had an 80% tumor volume reduction and remained alive without progression at the time of publication.

4.5. Novel Delivery Methods

Failure to prevent and treat glioblastoma recurrence may partially be attributed to challenges of drug delivery through the BBB [188]. Novel delivery approaches that couple effective delivery protocols with carriers, including intraventricular infusion of OV and intranasal nanoparticle delivery, show promise in overcoming the delivery challenge set by the BBB [147,171,176,189]. These treatments enable a more focal treatment of glioblastoma, limiting systemic side effects and directly preventing recurrence [190].
Two major disadvantages of intravenous drug administration are the systemic toxicities and the inability of molecules to bypass the BBB. Local intratumor delivery has been the main strategy for overcoming this challenge. However, each treatment infusion requires an invasive neurosurgical procedure, leading to increased complications due to repeat surgeries [191]. Intrathecal or intraventricular delivery is an alternative that could overcome the BBB. This approach has historically been avoided as it was considered high-risk CNS toxicity. In a pre-clinical study, Kang et al. [192] demonstrated that intraventricular delivery of OV G207 in a murine model of CNS disease resulted in increased endothelial-cell mediated toxicity, which could be rescued with a pre-treatment of low-dose OV G207 [191,193].
Studies are also investigating technologies that can increase the CNS concentration of systemically administered drugs. For example, MR-guided ultrasound (MRgFUS) results in the oscillation of pre-formed IV-infused microbubbles, which concentrate the acoustic energy of ultrasound. This causes a transient mechanical separation of endothelial cells, thus “opening” the BBB [194]. Abrahao et al. [195] tested the safety of MRgFUS in a first-in-human feasibility study among four patients with amyotrophic lateral sclerosis. Increased transient BBB permeability, assessed with gadolinium leakage, was achieved in all patients. Multiple pre-clinical glioma models have demonstrated drug delivery safety using MRgFUS [196]. More recently, focal delivery of CAR-T cell therapy, which otherwise results in a potent systemic inflammatory effect, has demonstrated the precision of drug delivery to the local TME when combined with MRgFUS [197].
Nanocarriers and nanotechnology-based drug delivery have also demonstrated positive results in preclinical studies by effectively transporting drugs into the CNS. Because of their small size, nanoparticles can penetrate small capillaries and efficiently pass the BBB through receptor-mediated endocytosis [198,199]. Nanoparticles utilized in glioblastoma treatment have chemotherapeutic agents trapped inside their matrix. The external core membrane undergoes biodegradation within the CNS [200]. Preclinical studies have reported that nanoparticle-delivering agents that inhibit lactate metabolism resulted in the halting of cancer cell growth [201]. The nanoparticles contained lactate oxidase, which, in the hypoxic TME, converted lactate to pyruvic acid and hydrogen peroxide, blocking histone expression and inducing cell cycle arrest. Kumthekar et al. [202] demonstrated the feasibility of using nanoparticles to deliver small interfering RNA (siRNA) in glioblastoma tumors of non-human primates and in a human phase 0 clinical trial. They reported effective localization of gold nanoparticles in tumor cells. Another study investigated intranasally administered polymeric nanoparticles delivering bevacizumab, taking advantage of this technology and a novel administration route [189]. In the preclinical study by Sousa et al. [189], the authors reported that an intranasal administration of nanoparticles led to direct delivery to the CNS, bypassing the BBB through the olfactory and trigeminal nerves [203]. Their results showed that intranasal administration improved CNS bioavailability of bevacizumab, decreasing systemic side effects. However, the therapy did not result in significant tumor shrinkage when comparing nanoparticle delivery to the intravenous administration arms. This, though, is likely explained by the effect of bevacizumab rather than the delivery method [189].

4.6. Laser Interstitial Thermal Therapy, Electrical Fields, Photodynamic Therapy, and Intratumoral Microdevices

LITT presents a potential adjunct to resective surgery [204]. LITT uses an optical fiber that generates heat, leading to tumor cell necrosis or apoptosis [190,204]. Moreover, LITT can sensitize glioma cells to other treatment modalities by altering this primed TME; it can cause immune upregulation and BBB disruption [205,206,207,208]. Mohammadi et al. [204] showed a correlation between tissue damage in recurrent glioblastoma regions with LITT and favorable PFS. Reoperation requires careful patient selection and has high complication rates, to which LITT presents an alternative with comparable outcomes [209,210].
Even less invasive, the Optune TTF device was approved by the FDA in 2011 as a treatment for recurrent glioblastoma [211]. The TTF electrodes are applied to the patient’s scalp and deliver low-intensity and intermediate-frequency alternating electric fields [2,212]. The mechanism is thought to interfere with the polymerization and depolymerization of microtubules in mitotic spindles, leading to disruption in cell replication and cell death [212]. Dono et al. suggested that the aberrant progression into mitosis because of PTEN mutations in recurrent glioblastoma is halted by TTF through the mechanism of mitotic arrest/delay, leading to improved median survival in recurrent glioblastoma patients with PTEN mutations [213]. An established phase 3 clinical trial by Stupp and collaborators showed that TTF devices when worn 18–24 h a day led to similar OS in patients with recurrent glioblastoma undergoing chemotherapy [214]. Shortly after the FDA approval of Optune, Mrugala et al. performed an analysis of the clinical outcomes of TTF device utilization in patients with recurrent glioblastoma across 91 sites in the U.S. Their results revealed that increased compliance with wearing the device (daily compliance ≥ 75%, or ≥18 h daily) led to significantly better median OS at 13.5 months vs. 4.0 months compared with patients who were not compliant [215]. Dono et al. reported that within the TTF monotherapy group, TTF improved the median PPS of recurrent glioblastoma patients with PTEN mutations, yet this improvement did not reach statistical significance (13.9 months versus 10.9 months, p = 0.068) [213]. Despite questions on patient selection and lack of multivariate analyses, the largest benefits of TTF include a minimally invasive approach and an acceptable safety profile [2,211,215,216]. Unfortunately, despite these promising results, uptake in standard-of-care treatment plans has been slow [2,217].
Another potential treatment for recurrent glioblastoma is photodynamic therapy (PDT) [218]. In PDT, a photosensitizer is activated using a specific wavelength of light, which leads to the generation of reactive oxygen species in tissue that has accumulated the photosensitizer and undergone irradiation, which ultimately leads to the death of tumor cells [219]. Agents that have been used clinically for PDT in gliomas include Photofrin, Temoporfin, HpD, and 5-aminolevulinic acid, which leads to overproduction of the photosensitizer protoporphyrin IX [220,221,222,223]. For example, a study in Japan used the photosensitizer talaporfin sodium for PDT in recurrent glioblastoma, where the authors analyzed a group of 70 patients undergoing surgery plus PDT and 38 control patients undergoing surgery only. The PDT group had improved PFS and OS compared to the surgery-only group (5.7 vs. 2.2 months PFS and 16 vs. 12.8 months OS, respectively), and the effect of PDT on OS was maintained after univariate and multivariate analyses. Despite numerous studies with encouraging results, PDT for glioblastoma has not yielded a successful phase III trial. However, there is active research in new photosensitizers for PDT, delivery methods, and ongoing clinical trials using PDT on glioblastoma, including a trial on recurrent glioblastoma using 5-aminlevulinic acid [224].
New technologies can also be used to guide treatment strategies. Recently, in a first-in-human clinical trial, Peruzzi et al. [225] demonstrated the safety and efficacy of a novel biocompatible intratumoral microdevice (IMD) used to deliver nano-doses of chemotherapy to different areas of the TME in real time during tumor resection. The IMDs are inserted into the tumor at the beginning of surgery and are removed at the end of surgery. This approach acknowledges the large heterogeneity in glioblastoma and non-tumor cells within the TME and samples the sensitivity of the tumor to different chemotherapy agents. Besides demonstrating safety and an excellent adverse event profile associated with the IMD, Peruzzi et al. [225] reported that a patient with recurrent glioblastoma had a tumor resistant to all tested chemotherapeutic agents and avoided ineffective adjuvant chemotherapy and unnecessary potential systemic toxicities from the chemotherapy. Sampling chemotherapy sensitivity in an individual patient’s TME in real time demonstrates the potential to personalize adjuvant therapy. Such personalized therapeutic approaches may become critical as large molecular and genetic data have not added practical therapeutic value because of the multiplicity of involved pathways and the epigenetic modifications tumor cells undergo after initial therapy.

5. Conclusions

Glioblastoma is the most aggressive malignancy of the CNS and, despite standard-of-care resection and chemoradiation, most patients succumb to the disease. Extensive research during the past decade has uncovered molecular and genetic bases of glioblastoma recurrence, including driver mutations and major molecular pathways that sustain tumor cell proliferation. There has also been a perspective shift to investigate epigenetic modifications of recurrent tumors and a focus on understanding the interactions between tumor cells and non-tumor cells in the TME. A greater understanding of these mechanisms has guided advancements in treatments. For example, immune checkpoint inhibitors and engineered CAR-T cells utilize lymphocytes’ ability to create a local pro-inflammatory state unfavorable for tumor cell growth and invasion. On the other hand, oncolytic virotherapy introduces a viral pathogen targeted to the tumor cavity to activate a pro-inflammatory response. Novel drug delivery approaches, such as viral capsids or nanoparticles, can also help overcome treatment barriers, like bypassing the BBB and avoiding systemic side effects. Additionally, traditional surgery still plays a critical cytoreductive role in prolonging OS for select patients, although reoperation is often limited by patient characteristics such as age, performance status, tumor location, and involvement of critical brain regions. Alternative or additive treatments include LITT and TTFs. There is still more work required to identify effective treatments and treatment combinations for recurrent glioblastoma, but ongoing research into the mechanisms of recurrence and treatment evasion will help guide future trials.

Author Contributions

Conceptualization, J.D.B. and P.P.; writing—original draft preparation, C.C.; writing—review and editing, C.C., V.S.C., J.V.E.G., S.E.B., L.S., D.D., P.A.V., L.N.T., S.G., L.E., D.M., F.A.G., O.A., T.R.S., G.K.F., P.P. and J.D.B.; project administration, J.D.B. and P.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Institutes of Health (1R01NS116144 and R01CA283905) grants to P.P. and the DFCI/Kiki Leptomeningeal Disease Grant (9620415) to J.D.B. L.S. is supported by a research fellowship from the German Research Foundation (DFG, SP 2144/1-1). Sontag Foundation to P.P.

Conflicts of Interest

P.P. is the founder, sits on the board of directors, and owns equities of Ternalys Therapeutics. J.D.B has an equity position in Treovir Inc. and UpFront Diagnostics. J.D.B is also a co-founder of Centile Bioscience and on the NeuroX1 and QV Bioelectronics scientific advisory boards.

References

  1. Stupp, R.; Mason, W.P.; Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  2. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.M.; 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]
  3. 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]
  4. 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]
  5. 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]
  6. Marenco-Hillembrand, L.; Wijesekera, O.; Suarez-Meade, P.; Mampre, D.; Jackson, C.; Peterson, J.; Trifiletti, D.; Hammack, J.; Ortiz, K.; Lesser, E.; et al. Trends in glioblastoma: Outcomes over time and type of intervention: A systematic evidence based analysis. J. Neuro-Oncol. 2020, 147, 297–307. [Google Scholar] [CrossRef]
  7. van Linde, M.E.; Brahm, C.G.; de Witt Hamer, P.C.; Reijneveld, J.C.; Bruynzeel, A.M.E.; Vandertop, W.P.; van de Ven, P.M.; Wagemakers, M.; van der Weide, H.L.; Enting, R.H.; et al. Treatment outcome of patients with recurrent glioblastoma multiforme: A retrospective multicenter analysis. J. Neuro-Oncol. 2017, 135, 183–192. [Google Scholar] [CrossRef]
  8. Brown, P.D.; Maurer, M.J.; Rummans, T.A.; Pollock, B.E.; Ballman, K.V.; Sloan, J.A.; Boeve, B.F.; Arusell, R.M.; Clark, M.M.; Buckner, J.C. A prospective study of quality of life in adults with newly diagnosed high-grade gliomas: The impact of the extent of resection on quality of life and survival. Neurosurgery 2005, 57, 495–504. [Google Scholar] [CrossRef]
  9. Stummer, W.; Pichlmeier, U.; Meinel, T.; Wiestler, O.D.; Zanella, F.; Reulen, H.-J. 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]
  10. Picart, T.; Pallud, J.; Berthiller, J.; Dumot, C.; Berhouma, M.; Ducray, F.; Armoiry, X.; Margier, J.; Guerre, P.; Varlet, P.; et al. Use of 5-ALA fluorescence-guided surgery versus white-light conventional microsurgery for the resection of newly diagnosed glioblastomas (RESECT study): A French multicenter randomized phase III study. J. Neurosurg. 2024, 140, 987–1000. [Google Scholar] [CrossRef]
  11. Valdes, P.A.; Juvekar, P.; Agar, N.Y.R.; Gioux, S.; Golby, A.J. Quantitative Wide-Field Imaging Techniques for Fluorescence Guided Neurosurgery. Front. Surg. 2019, 6, 31. [Google Scholar] [CrossRef]
  12. Valdés, P.A.; Roberts, D.W.; Lu, F.K.; Golby, A. Optical technologies for intraoperative neurosurgical guidance. Neurosurg. Focus. 2016, 40, E8. [Google Scholar] [CrossRef]
  13. Roberts, D.W.; Valdés, P.A.; Harris, B.T.; Hartov, A.; Fan, X.; Ji, S.; Pogue, B.W.; Leblond, F.; Tosteson, T.D.; Wilson, B.C.; et al. Adjuncts for maximizing resection: 5-aminolevuinic acid. Clin. Neurosurg. 2012, 59, 75–78. [Google Scholar] [CrossRef]
  14. Adamson, C.; Kanu, O.O.; Mehta, A.I.; Di, C.; Lin, N.; Mattox, A.K.; Bigner, D.D. Glioblastoma multiforme: A review of where we have been and where we are going. Expert. Opin. Investig. Drugs 2009, 18, 1061–1083. [Google Scholar] [CrossRef]
  15. Andersen, B.M.; Faust Akl, C.; Wheeler, M.A.; Chiocca, E.A.; Reardon, D.A.; Quintana, F.J. Glial and myeloid heterogeneity in the brain tumour microenvironment. Nat. Rev. Cancer 2021, 21, 786–802. [Google Scholar] [CrossRef]
  16. Birzu, C.; French, P.; Caccese, M.; Cerretti, G.; Idbaih, A.; Zagonel, V.; Lombardi, G. Recurrent glioblastoma: From molecular landscape to new treatment perspectives. Cancers 2021, 13, 47. [Google Scholar] [CrossRef]
  17. Burger, P.C.; Heinz, E.R.; Shibata, T.; Kleihues, P. Topographic anatomy and CT correlations in the untreated glioblastoma multiforme. J. Neurosurg. 1988, 68, 698–704. [Google Scholar] [CrossRef]
  18. Campos, B.; Olsen, L.R.; Urup, T.; Poulsen, H.S. A comprehensive profile of recurrent glioblastoma. Oncogene 2016, 35, 5819–5825. [Google Scholar] [CrossRef]
  19. Montemurro, N.; Pahwa, B.; Tayal, A.; Shukla, A.; De Jesus Encarnacion, M.; Ramirez, I.; Nurmukhametov, R.; Chavda, V.; De Carlo, A. Macrophages in Recurrent Glioblastoma as a Prognostic Factor in the Synergistic System of the Tumor Microenvironment. Neurol. Int. 2023, 15, 595–608. [Google Scholar] [CrossRef]
  20. Yang, J.; Xu, J.; Wang, W.; Zhang, B.; Yu, X.; Shi, S. Epigenetic regulation in the tumor microenvironment: Molecular mechanisms and therapeutic targets. Signal Transduct. Target. Ther. 2023, 8, 210. [Google Scholar] [CrossRef]
  21. Wang, J.; Cazzato, E.; Ladewig, E.; Frattini, V.; Rosenbloom, D.I.S.; Zairis, S.; Abate, F.; Liu, Z.; Elliott, O.; Shin, Y.J.; et al. Clonal evolution of glioblastoma under therapy. Nat. Genet. 2016, 48, 768–776. [Google Scholar] [CrossRef]
  22. 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]
  23. Martinez, R.; Rohde, V.; Schackert, G. Different molecular patterns in glioblastoma multiforme subtypes upon recurrence. J. Neuro-Oncol. 2010, 96, 321–329. [Google Scholar] [CrossRef]
  24. Cloughesy, T.F.; Cavenee, W.K.; Mischel, P.S. Glioblastoma: From molecular pathology to targeted treatment. Annu. Rev. Pathol. Mech. Dis. 2014, 9, 1–25. [Google Scholar] [CrossRef]
  25. 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]
  26. Chen, N.; Peng, C.; Li, D. Epigenetic Underpinnings of Inflammation: A Key to Unlock the Tumor Microenvironment in Glioblastoma. Front. Immunol. 2022, 13, 869307. [Google Scholar] [CrossRef]
  27. Miki, S.; Satomi, K.; Ohno, M.; Matsushita, Y.; Kitahara, M.; Miyakita, Y.; Takahashi, M.; Matsuda, M.; Ishikawa, E.; Matsumura, A.; et al. Highly sensitive detection of TERT promoter mutations in recurrent glioblastomas using digital PCR. Brain Tumor Pathol. 2020, 37, 154–158. [Google Scholar] [CrossRef]
  28. Akyerli, C.B.; Yüksel, Ş.; Can, Ö.; Erson-Omay, E.Z.; Oktay, Y.; Coşgun, E.; Ülgen, E.; Erdemgil, Y.; Sav, A.; von Deimling, A.; et al. Use of telomerase promoter mutations to mark specific molecular subsets with reciprocal clinical behavior in IDH mutant and IDH wild-type diffuse gliomas. J. Neurosurg. 2018, 128, 1102–1114. [Google Scholar] [CrossRef]
  29. He, J.; Zhang, Z.; Ouyang, M.; Yang, F.; Hao, H.; Lamb, K.L.; Yang, J.; Yin, Y.; Shen, W.H. PTEN regulates EG5 to control spindle architecture and chromosome congression during mitosis. Nat. Commun. 2016, 7, 12355. [Google Scholar] [CrossRef]
  30. Gan, H.K.; Cvrljevic, A.N.; Johns, T.G. The epidermal growth factor receptor variant III (EGFRvIII): Where wild things are altered. FEBS J. 2013, 280, 5350–5370. [Google Scholar] [CrossRef]
  31. An, Z.; Aksoy, O.; Zheng, T.; Fan, Q.W.; Weiss, W.A. Epidermal growth factor receptor and EGFRvIII in glioblastoma: Signaling pathways and targeted therapies. Oncogene 2018, 37, 1561–1575. [Google Scholar] [CrossRef]
  32. Feng, H.; Hu, B.; Jarzynka, M.J.; Li, Y.; Keezer, S.; Johns, T.G.; Tang, C.K.; Hamilton, R.L.; Vuori, K.; Nishikawa, R.; et al. Phosphorylation of dedicator of cytokinesis 1 (Dock180) at tyrosine residue Y722 by Src family kinases mediates EGFRvIII-driven glioblastoma tumorigenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 3018–3023. [Google Scholar] [CrossRef]
  33. Cvrljevic, A.N.; Akhavan, D.; Wu, M.; Martinello, P.; Furnari, F.B.; Johnston, A.J.; Guo, D.; Pike, L.; Cavenee, W.K.; Scott, A.M.; et al. Activation of Src induces mitochondrial localisation of de2-7EGFR (EGFRvIII) in glioma cells: Implications for glucose metabolism. J. Cell Sci. 2011, 124, 2938–2950. [Google Scholar] [CrossRef]
  34. Kim, J.; Lee, I.H.; Cho, H.J.; Park, C.K.; Jung, Y.S.; Kim, Y.; Nam, S.H.; Kim, B.S.; Johnson, M.D.; Kong, D.S.; et al. Spatiotemporal Evolution of the Primary Glioblastoma Genome. Cancer Cell 2015, 28, 318–328. [Google Scholar] [CrossRef]
  35. Crespo, S.; Kind, M.; Arcaro, A. The role of the PI3K/AKT/mTOR pathway in brain tumor metastasis. J Cancer Metastasis Treat 2016, 2, 80–89. [Google Scholar] [CrossRef]
  36. Zhou, X.-Y.; Liu, H.; Ding, Z.-B.; Xi, H.-P.; Wang, G.-W. lncRNA SNHG16 promotes glioma tumorigenicity through miR-373/EGFR axis by activating PI3K/AKT pathway. Genomics 2020, 112, 1021–1029. [Google Scholar] [CrossRef]
  37. Pan, J.; Sheng, S.; Ye, L.; Xu, X.; Ma, Y.; Feng, X.; Qiu, L.; Fan, Z.; Wang, Y.; Xia, X.; et al. Extracellular vesicles derived from glioblastoma promote proliferation and migration of neural progenitor cells via PI3K-Akt pathway. Cell Commun. Signal. 2022, 20, 7. [Google Scholar] [CrossRef]
  38. Kim, Y.; Kim, E.; Wu, Q.; Guryanova, O.; Hitomi, M.; Lathia, J.D.; Serwanski, D.; Sloan, A.E.; Weil, R.J.; Lee, J.; et al. Platelet-derived growth factor receptors differentially inform intertumoral and intratumoral heterogeneity. Genes. Dev. 2012, 26, 1247–1262. [Google Scholar] [CrossRef]
  39. 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/p38(MAPK) signalling. Oncogene 2022, 41, 2749–2763. [Google Scholar] [CrossRef]
  40. Pixley, F.J.; Stanley, E.R. CSF-1 regulation of the wandering macrophage: Complexity in action. Trends Cell Biol. 2004, 14, 628–638. [Google Scholar] [CrossRef]
  41. Lamagna, C.; Aurrand-Lions, M.; Imhof, B.A. Dual role of macrophages in tumor growth and angiogenesis. J. Leukoc. Biol. 2006, 80, 705–713. [Google Scholar] [CrossRef]
  42. Yang, L.; Zhang, Y. Tumor-associated macrophages: From basic research to clinical application. J. Hematol. Oncol. 2017, 10, 58. [Google Scholar] [CrossRef]
  43. Zhu, C.; Kros, J.M.; van der Weiden, M.; Zheng, P.; Cheng, C.; Mustafa, D.A.M. Expression site of P2RY12 in residential microglial cells in astrocytomas correlates with M1 and M2 marker expression and tumor grade. Acta Neuropathol. Commun. 2017, 5, 4. [Google Scholar] [CrossRef]
  44. Pombo Antunes, A.R.; Scheyltjens, I.; Lodi, F.; Messiaen, J.; Antoranz, A.; Duerinck, J.; Kancheva, D.; Martens, L.; De Vlaminck, K.; Van Hove, H.; et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat. Neurosci. 2021, 24, 595–610. [Google Scholar] [CrossRef]
  45. Vitkovic, L.; Maeda, S.; Sternberg, E. Anti-inflammatory cytokines: Expression and action in the brain. Neuroimmunomodulation 2001, 9, 295–312. [Google Scholar] [CrossRef]
  46. Gong, D.; Shi, W.; Yi, S.-j.; Chen, H.; Groffen, J.; Heisterkamp, N. TGFβ signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol. 2012, 13, 31. [Google Scholar] [CrossRef]
  47. 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]
  48. Dong, F.; Qin, X.; Wang, B.; Li, Q.; Hu, J.; Cheng, X.; Guo, D.; Cheng, F.; Fang, C.; Tan, Y.; et al. ALKBH5 Facilitates Hypoxia-Induced Paraspeckle Assembly and IL8 Secretion to Generate an Immunosuppressive Tumor Microenvironment. Cancer Res. 2021, 81, 5876–5888. [Google Scholar] [CrossRef]
  49. Zhang, S.; Zhao, B.S.; Zhou, A.; Lin, K.; Zheng, S.; Lu, Z.; Chen, Y.; Sulman, E.P.; Xie, K.; Bögler, O.; et al. m(6)A Demethylase ALKBH5 Maintains Tumorigenicity of Glioblastoma Stem-like Cells by Sustaining FOXM1 Expression and Cell Proliferation Program. Cancer Cell 2017, 31, 591–606.e596. [Google Scholar] [CrossRef]
  50. Storey, K.; Leder, K.; Hawkins-Daarud, A.; Swanson, K.; Ahmed, A.U.; Rockne, R.C.; Foo, J. Glioblastoma Recurrence and the Role of O(6)-Methylguanine-DNA Methyltransferase Promoter Methylation. JCO Clin. Cancer Inform. 2019, 3, 1–12. [Google Scholar] [CrossRef]
  51. Suzuki, T.; Nakada, M.; Yoshida, Y.; Nambu, E.; Furuyama, N.; Kita, D.; Hayashi, Y.; Hayashi, Y.; Hamada, J.-I. The correlation between promoter methylation status and the expression level of O6-methylguanine-DNA methyltransferase in recurrent glioma. Jpn. J. Clin. Oncol. 2011, 41, 190–196. [Google Scholar] [CrossRef]
  52. Christmann, M.; Nagel, G.; Horn, S.; Krahn, U.; Wiewrodt, D.; Sommer, C.; Kaina, B. MGMT activity, promoter methylation and immunohistochemistry of pretreatment and recurrent malignant gliomas: A comparative study on astrocytoma and glioblastoma. Int. J. Cancer 2010, 127, 2106–2118. [Google Scholar] [CrossRef]
  53. Powter, B.; Jeffreys, S.A.; Sareen, H.; Cooper, A.; Brungs, D.; Po, J.; Roberts, T.; Koh, E.-S.; Scott, K.F.; Sajinovic, M.; et al. Human TERT promoter mutations as a prognostic biomarker in glioma. J. Cancer Res. Clin. Oncol. 2021, 147, 1007–1017. [Google Scholar] [CrossRef]
  54. Labussière, M.; Boisselier, B.; Mokhtari, K.; Stefano, A.-L.D.; Rahimian, A.; Rossetto, M.; Ciccarino, P.; Saulnier, O.; Paterra, R.; Marie, Y.; et al. Combined analysis of TERT, EGFR, and IDH status defines distinct prognostic glioblastoma classes. Neurology 2014, 83, 1200–1206. [Google Scholar] [CrossRef]
  55. You, H.; Wu, Y.; Chang, K.; Shi, X.; Chen, X.-D.; Yan, W.; Li, R. Paradoxical prognostic impact of TERT promoter mutations in gliomas depends on different histological and genetic backgrounds. CNS Neurosci. Ther. 2017, 23, 790–797. [Google Scholar] [CrossRef]
  56. Zhang, L.; Liu, Z.; Li, J.; Huang, T.; Wang, Y.; Chang, L.; Zheng, W.; Ma, Y.; Chen, F.; Gong, X.; et al. Genomic analysis of primary and recurrent gliomas reveals clinical outcome related molecular features. Sci. Rep. 2019, 9, 16058. [Google Scholar] [CrossRef]
  57. Liaw, D.; Marsh, D.J.; Li, J.; Dahia, P.L.; Wang, S.I.; Zheng, Z.; Bose, S.; Call, K.M.; Tsou, H.C.; Peacocke, M.; et al. Germline mutations of the PTEN gene in Cowden disease, an inherited breast and thyroid cancer syndrome. Nat. Genet. 1997, 16, 64–67. [Google Scholar] [CrossRef]
  58. Steck, P.A.; Pershouse, M.A.; Jasser, S.A.; Yung, W.K.; Lin, H.; Ligon, A.H.; Langford, L.A.; Baumgard, M.L.; Hattier, T.; Davis, T.; et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nat. Genet. 1997, 15, 356–362. [Google Scholar] [CrossRef]
  59. Garcia-Cao, I.; Song, M.S.; Hobbs, R.M.; Laurent, G.; Giorgi, C.; de Boer, V.C.J.; Anastasiou, D.; Ito, K.; Sasaki, A.T.; Rameh, L.; et al. Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 2012, 149, 49–62. [Google Scholar] [CrossRef]
  60. Maehama, T.; Dixon, J.E. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. J. Biol. Chem. 1998, 273, 13375–13378. [Google Scholar] [CrossRef]
  61. Song, M.S.; Salmena, L.; Pandolfi, P.P. The functions and regulation of the PTEN tumour suppressor. Nat. Reviews. Mol. Cell Biol. 2012, 13, 283–296. [Google Scholar] [CrossRef]
  62. Choi, S.W.; Lee, Y.; Shin, K.; Koo, H.; Kim, D.; Sa, J.K.; Cho, H.J.; Shin, H.-m.; Lee, S.J.; Kim, H.; et al. Mutation-specific non-canonical pathway of PTEN as a distinct therapeutic target for glioblastoma. Cell Death Dis. 2021, 12, 374. [Google Scholar] [CrossRef]
  63. Ohgaki, H.; Kleihues, P. Genetic alterations and signaling pathways in the evolution of gliomas. Cancer Sci. 2009, 100, 2235–2241. [Google Scholar] [CrossRef]
  64. Akhavan, D.; Cloughesy, T.F.; Mischel, P.S. mTOR signaling in glioblastoma: Lessons learned from bench to bedside. Neuro Oncol. 2010, 12, 882–889. [Google Scholar] [CrossRef]
  65. Zhang, Z.; Fan, Q.; Luo, X.; Lou, K.; Weiss, W.A.; Shokat, K.M. Brain-restricted mTOR inhibition with binary pharmacology. Nature 2022, 609, 822–828. [Google Scholar] [CrossRef]
  66. Neilsen, B.K.; Sleightholm, R.; McComb, R.; Ramkissoon, S.H.; Ross, J.S.; Corona, R.J.; Miller, V.A.; Cooke, M.; Aizenberg, M.R. Comprehensive genetic alteration profiling in primary and recurrent glioblastoma. J. Neuro-Oncol. 2019, 142, 111–118. [Google Scholar] [CrossRef]
  67. Koul, D.; Shen, R.; Bergh, S.; Sheng, X.; Shishodia, S.; Lafortune, T.A.; Lu, Y.; de Groot, J.F.; Mills, G.B.; Yung, W.K.A. Inhibition of Akt survival pathway by a small-molecule inhibitor in human glioblastoma. Mol. Cancer Ther. 2006, 5, 637–644. [Google Scholar] [CrossRef]
  68. Gallia, G.L.; Tyler, B.M.; Hann, C.L.; Siu, I.-M.; Giranda, V.L.; Vescovi, A.L.; Brem, H.; Riggins, G.J. Inhibition of Akt inhibits growth of glioblastoma and glioblastoma stem-like cells. Mol. Cancer Ther. 2009, 8, 386–393. [Google Scholar] [CrossRef]
  69. Wang, S.-J.; Wang, H.; Zhao, C.-D.; Li, R. Long noncoding RNA LINC01426 promotes glioma progression through PI3K/AKT signaling pathway and serves as a prognostic biomarker. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 6358–6368. [Google Scholar] [CrossRef]
  70. Liu, B.; Cao, W.; Ma, H. Knockdown of lncRNA LSINCT5 suppresses growth and metastasis of human glioma cells via up-regulating miR-451. Artif. Cells Nanomed. Biotechnol. 2019, 47, 2507–2515. [Google Scholar] [CrossRef]
  71. Jorissen, R.N.; Walker, F.; Pouliot, N.; Garrett, T.P.; Ward, C.W.; Burgess, A.W. Epidermal growth factor receptor: Mechanisms of activation and signalling. Exp. Cell Res. 2003, 284, 31–53. [Google Scholar] [CrossRef]
  72. Herbst, R.S. Review of epidermal growth factor receptor biology. Int. J. Radiat. Oncol. Biol. Phys. 2004, 59, 21–26. [Google Scholar] [CrossRef]
  73. Dhawan, A.; Manem, V.S.K.; Yeaney, G.; Lathia, J.D.; Ahluwalia, M.S. EGFR Pathway Expression Persists in Recurrent Glioblastoma Independent of Amplification Status. Cancers 2023, 15, 670. [Google Scholar] [CrossRef]
  74. Ekstrand, A.J.; James, C.D.; Cavenee, W.K.; Seliger, B.; Pettersson, R.F.; Collins, V.P. Genes for epidermal growth factor receptor, transforming growth factor alpha, and epidermal growth factor and their expression in human gliomas in vivo. Cancer Res. 1991, 51, 2164–2172. [Google Scholar]
  75. van den Bent, M.J.; Gao, Y.; Kerkhof, M.; Kros, J.M.; Gorlia, T.; van Zwieten, K.; Prince, J.; van Duinen, S.; Sillevis Smitt, P.A.; Taphoorn, M.; et al. Changes in the EGFR amplification and EGFRvIII expression between paired primary and recurrent glioblastomas. Neuro Oncol. 2015, 17, 935–941. [Google Scholar] [CrossRef]
  76. Nagane, M.; Coufal, F.; Lin, H.; Bögler, O.; Cavenee, W.K.; Huang, H.J. A common mutant epidermal growth factor receptor confers enhanced tumorigenicity on human glioblastoma cells by increasing proliferation and reducing apoptosis. Cancer Res. 1996, 56, 5079–5086. [Google Scholar]
  77. Cantanhede, I.G.; de Oliveira, J.R.M. PDGF Family Expression in Glioblastoma Multiforme: Data Compilation from Ivy Glioblastoma Atlas Project Database. Sci. Rep. 2017, 7, 15271. [Google Scholar] [CrossRef]
  78. Indraccolo, S.; Lombardi, G.; Fassan, M.; Pasqualini, L.; Giunco, S.; Marcato, R.; Gasparini, A.; Candiotto, C.; Nalio, S.; Fiduccia, P.; et al. Genetic, Epigenetic, and Immunologic Profiling of MMR-Deficient Relapsed Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2019, 25, 1828–1837. [Google Scholar] [CrossRef]
  79. 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. Off. J. Am. Assoc. Cancer Res. 2020, 26, 1690–1699. [Google Scholar] [CrossRef]
  80. 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 BRAF(V600)-Mutant Gliomas: Results From the VE-BASKET Study. J. Clin. Oncol. 2018, 36, 3477–3484. [Google Scholar] [CrossRef]
  81. 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]
  82. Klughammer, J.; Kiesel, B.; Roetzer, T.; Fortelny, N.; Nemc, A.; Nenning, K.H.; Furtner, J.; Sheffield, N.C.; Datlinger, P.; Peter, N.; et al. The DNA methylation landscape of glioblastoma disease progression shows extensive heterogeneity in time and space. Nat. Med. 2018, 24, 1611–1624. [Google Scholar] [CrossRef]
  83. Eisenbarth, D.; Wang, Y.A. Glioblastoma heterogeneity at single cell resolution. Oncogene 2023, 42, 2155–2165. [Google Scholar] [CrossRef]
  84. Gangoso, E.; Southgate, B.; Bradley, L.; Rus, S.; Galvez-Cancino, F.; McGivern, N.; Güç, E.; Kapourani, C.-A.; Byron, A.; Ferguson, K.M.; et al. Glioblastomas acquire myeloid-affiliated transcriptional programs via epigenetic immunoediting to elicit immune evasion. Cell 2021, 184, 2454–2470.e2426. [Google Scholar] [CrossRef]
  85. Cao, M.-F.; Chen, L.; Dang, W.-Q.; Zhang, X.-C.; Zhang, X.; Shi, Y.; Yao, X.-H.; Li, Q.; Zhu, J.; Lin, Y.; et al. Hybrids by tumor-associated macrophages × glioblastoma cells entail nuclear reprogramming and glioblastoma invasion. Cancer Lett. 2019, 442, 445–452. [Google Scholar] [CrossRef]
  86. Fu, W.; Wang, W.; Li, H.; Jiao, Y.; Huo, R.; Yan, Z.; Wang, J.; Wang, S.; Wang, J.; Chen, D.; et al. Single-Cell Atlas Reveals Complexity of the Immunosuppressive Microenvironment of Initial and Recurrent Glioblastoma. Front. Immunol. 2020, 11, 835. [Google Scholar] [CrossRef]
  87. Mormino, A.; Cocozza, G.; Fontemaggi, G.; Valente, S.; Esposito, V.; Santoro, A.; Bernardini, G.; Santoni, A.; Fazi, F.; Mai, A.; et al. Histone-deacetylase 8 drives the immune response and the growth of glioma. Glia 2021, 69, 2682–2698. [Google Scholar] [CrossRef]
  88. Wei, X.; Huo, Y.; Pi, J.; Gao, Y.; Rao, S.; He, M.; Wei, Q.; Song, P.; Chen, Y.; Lu, D.; et al. METTL3 preferentially enhances non-m6A translation of epigenetic factors and promotes tumourigenesis. Nat. Cell Biol. 2022, 24, 1278–1290. [Google Scholar] [CrossRef]
  89. Gerson, S.L. MGMT: Its role in cancer aetiology and cancer therapeutics. Nat. Rev. Cancer 2004, 4, 296–307. [Google Scholar] [CrossRef]
  90. 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.e237. [Google Scholar] [CrossRef]
  91. Murota, Y.; Tabu, K.; Taga, T. Cancer Stem Cell-Associated Immune Microenvironment in Recurrent Glioblastomas. Cells 2022, 11, 2054. [Google Scholar] [CrossRef]
  92. Phillips, J.P.; Eremin, O.; Anderson, J.R. Lymphoreticular cells in human brain tumours and in normal brain. Br. J. Cancer 1982, 45, 61–69. [Google Scholar] [CrossRef]
  93. Darmanis, S.; Sloan, S.A.; Croote, D.; Mignardi, M.; Chernikova, S.; Samghababi, P.; Zhang, Y.; Neff, N.; Kowarsky, M.; Caneda, C.; et al. Single-Cell RNA-Seq Analysis of Infiltrating Neoplastic Cells at the Migrating Front of Human Glioblastoma. Cell Rep. 2017, 21, 1399–1410. [Google Scholar] [CrossRef]
  94. Klemm, F.; Maas, R.R.; Bowman, R.L.; Kornete, M.; Soukup, K.; Nassiri, S.; Brouland, J.-P.; Iacobuzio-Donahue, C.A.; Brennan, C.; Tabar, V.; et al. Interrogation of the Microenvironmental Landscape in Brain Tumors Reveals Disease-Specific Alterations of Immune Cells. Cell 2020, 181, 1643–1660.e1617. [Google Scholar] [CrossRef]
  95. Wang, L.-J.; Lv, P.; Lou, Y.; Ye, J. Gene Expression-Based Predication of RNA Pseudouridine Modification in Tumor Microenvironment and Prognosis of Glioma Patients. Front. Cell Dev. Biol. 2021, 9, 727595. [Google Scholar] [CrossRef]
  96. Pyonteck, S.M.; Akkari, L.; Schuhmacher, A.J.; Bowman, R.L.; Sevenich, L.; Quail, D.F.; Olson, O.C.; Quick, M.L.; Huse, J.T.; Teijeiro, V.; et al. CSF-1R inhibition alters macrophage polarization and blocks glioma progression. Nat. Med. 2013, 19, 1264–1272. [Google Scholar] [CrossRef]
  97. Martinez, F.O.; Sica, A.; Mantovani, A.; Locati, M. Macrophage activation and polarization. Front. Biosci. A J. Virtual Libr. 2008, 13, 453–461. [Google Scholar] [CrossRef]
  98. Lawrence, T.; Natoli, G. Transcriptional regulation of macrophage polarization: Enabling diversity with identity. Nat. Rev. Immunol. 2011, 11, 750–761. [Google Scholar] [CrossRef]
  99. Zanotto-Filho, A.; Gonçalves, R.M.; Klafke, K.; de Souza, P.O.; Dillenburg, F.C.; Carro, L.; Gelain, D.P.; Moreira, J.C.F. Inflammatory landscape of human brain tumors reveals an NFκB dependent cytokine pathway associated with mesenchymal glioblastoma. Cancer Lett. 2017, 390, 176–187. [Google Scholar] [CrossRef]
  100. Dumas, A.A.; Pomella, N.; Rosser, G.; Guglielmi, L.; Vinel, C.; Millner, T.O.; Rees, J.; Aley, N.; Sheer, D.; Wei, J.; et al. Microglia promote glioblastoma via mTOR-mediated immunosuppression of the tumour microenvironment. EMBO J. 2020, 39, e103790. [Google Scholar] [CrossRef]
  101. Wu, L.; Wu, W.; Zhang, J.; Zhao, Z.; Li, L.; Zhu, M.; Wu, M.; Wu, F.; Zhou, F.; Du, Y.; et al. Natural Coevolution of Tumor and Immunoenvironment in Glioblastoma. Cancer Discov. 2022, 12, 2820–2837. [Google Scholar] [CrossRef]
  102. Tatla, A.S.; Justin, A.W.; Watts, C.; Markaki, A.E. A vascularized tumoroid model for human glioblastoma angiogenesis. Sci. Rep. 2021, 11, 19550. [Google Scholar] [CrossRef]
  103. Hambardzumyan, D.; Bergers, G. Glioblastoma: Defining Tumor Niches. Trends Cancer 2015, 1, 252–265. [Google Scholar] [CrossRef]
  104. Gabriely, G.; Wheeler, M.A.; Takenaka, M.C.; Quintana, F.J. Role of AHR and HIF-1alpha in Glioblastoma Metabolism. Trends Endocrinol. Metab. 2017, 28, 428–436. [Google Scholar] [CrossRef]
  105. Chen, J.; Liu, G.; Wang, X.; Hong, H.; Li, T.; Li, L.; Wang, H.; Xie, J.; Li, B.; Li, T.; et al. Glioblastoma stem cell-specific histamine secretion drives pro-angiogenic tumor microenvironment remodeling. Cell Stem Cell 2022, 29, 1531–1546.e7. [Google Scholar] [CrossRef]
  106. Barbagallo, G.M.V.; Jenkinson, M.D.; Brodbelt, A.R. ‘Recurrent’ glioblastoma multiforme, when should we reoperate? Br. J. Neurosurg. 2008, 22, 452–455. [Google Scholar] [CrossRef]
  107. 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-Oncology 2016, 18, 96–104. [Google Scholar] [CrossRef]
  108. Patrick, H.H.; Sherman, J.H.; Elder, J.B.; Olson, J.J. Congress of neurological surgeons systematic review and evidence-based guidelines update on the role of cytoreductive surgery in the management of progressive glioblastoma in adults. J. Neuro-Oncol. 2022, 158, 167–177. [Google Scholar] [CrossRef]
  109. Ammirati, M.; Vick, N.; Liao, Y.L.; Ciric, I.; Mikhael, M. Effect of the extent of surgical resection on survival and quality of life in patients with supratentorial glioblastomas and anaplastic astrocytomas. Neurosurgery 1987, 21, 201–206. [Google Scholar] [CrossRef]
  110. Oppenlander, M.E.; Wolf, A.B.; Snyder, L.A.; Bina, R.; Wilson, J.R.; Coons, S.W.; Ashby, L.S.; Brachman, D.; Nakaji, P.; Porter, R.W.; et al. An extent of resection threshold for recurrent glioblastoma and its risk for neurological morbidity. J. Neurosurg. 2014, 120, 846–853. [Google Scholar] [CrossRef]
  111. Nava, F.; Tramacere, I.; Fittipaldo, A.; Bruzzone, M.G.; Dimeco, F.; Fariselli, L.; Finocchiaro, G.; Pollo, B.; Salmaggi, A.; Silvani, A.; et al. Survival effect of first- and second-line treatments for patients with primary glioblastoma: A cohort study from a prospective registry, 1997–2010. Neuro-Oncology 2014, 16, 719–727. [Google Scholar] [CrossRef]
  112. González, V.; Brell, M.; Fuster, J.; Moratinos, L.; Alegre, D.; López, S.; Ibáñez, J. Analyzing the role of reoperation in recurrent glioblastoma: A 15-year retrospective study in a single institution. World J. Surg. Oncol. 2022, 20, 384. [Google Scholar] [CrossRef]
  113. Dejaegher, J.; Vleeschouwer, S.D. Recurring Glioblastoma: A Case for Reoperation? Exon Publications 2017, 281–296. [Google Scholar] [CrossRef]
  114. Haddad, A.F.; Young, J.S.; Morshed, R.A.; Berger, M.S. FLAIRectomy: Resecting beyond the Contrast Margin for Glioblastoma. Brain Sci. 2022, 12, 544. [Google Scholar] [CrossRef]
  115. Bloch, O.; Han, S.J.; Cha, S.; Sun, M.Z.; Aghi, M.K.; McDermott, M.W.; Berger, M.S.; Parsa, A.T. Impact of extent of resection for recurrent glioblastoma on overall survival: Clinical article. J. Neurosurg. 2012, 117, 1032–1038. [Google Scholar] [CrossRef]
  116. Franceschi, E.; Bartolotti, M.; Tosoni, A.; Bartolini, S.; Sturiale, C.; Fioravanti, A.; Pozzati, E.; Galzio, R.; Talacchi, A.; Volpin, L.; et al. The effect of re-operation on survival in patients with recurrent glioblastoma. Anticancer. Res. 2015, 35, 1743–1748. [Google Scholar]
  117. Woernle, C.M.; Péus, D.; Hofer, S.; Rushing, E.J.; Held, U.; Bozinov, O.; Krayenbühl, N.; Weller, M.; Regli, L. Efficacy of Surgery and Further Treatment of Progressive Glioblastoma. World Neurosurg. 2015, 84, 301–307. [Google Scholar] [CrossRef]
  118. Voisin, M.R.; Zuccato, J.A.; Wang, J.Z.; Zadeh, G. Surgery for Recurrent Glioblastoma Multiforme: A Retrospective Case Control Study. World Neurosurg. 2022, 166, e624–e631. [Google Scholar] [CrossRef]
  119. Djamel-Eddine, Y.-C.; De Witte, O.; Mélot, C.; Lefranc, F. Recurrent glioblastomas: Should we operate a second and even a third time? Interdiscip. Neurosurg. 2019, 18, 100551. [Google Scholar] [CrossRef]
  120. Yong, R.L.; Wu, T.; Mihatov, N.; Shen, M.J.; Brown, M.A.; Zaghloul, K.A.; Park, G.E.; Park, J.K. Residual tumor volume and patient survival following reoperation for recurrent glioblastoma. J. Neurosurg. 2014, 121, 802–809. [Google Scholar] [CrossRef]
  121. Karschnia, P.; Dono, A.; Young, J.S.; Juenger, S.T.; Teske, N.; Häni, L.; Sciortino, T.; Mau, C.Y.; Bruno, F.; Nunez, L.; et al. Prognostic evaluation of re-resection for recurrent glioblastoma using the novel RANO classification for extent of resection: A report of the RANO resect group. Neuro Oncol. 2023, 25, 1672–1685. [Google Scholar] [CrossRef]
  122. Stark, A.M.; Nabavi, A.; Mehdorn, H.M.; Blömer, U. Glioblastoma multiforme-report of 267 cases treated at a single institution. Surg. Neurol. 2005, 63, 162–169. [Google Scholar] [CrossRef]
  123. Helseth, R.; Helseth, E.; Johannesen, T.B.; Langberg, C.W.; Lote, K.; Rønning, P.; Scheie, D.; Vik, A.; Meling, T.R. Overall survival, prognostic factors, and repeated surgery in a consecutive series of 516 patients with glioblastoma multiforme. Acta Neurol. Scand. 2010, 122, 159–167. [Google Scholar] [CrossRef]
  124. Okita, Y.; Narita, Y.; Miyakita, Y.; Ohno, M.; Fukushima, S.; Kayama, T.; Shibui, S. Pathological findings and prognostic factors in recurrent glioblastomas. Brain Tumor Pathol. 2012, 29, 192–200. [Google Scholar] [CrossRef]
  125. Park, C.-K.; Kim, J.H.; Nam, D.-H.; Kim, C.-Y.; Chung, S.-B.; Kim, Y.-H.; Seol, H.J.; Kim, T.M.; Choi, S.H.; Lee, S.-H.; et al. A practical scoring system to determine whether to proceed with surgical resection in recurrent glioblastoma. Neuro-Oncology 2013, 15, 1096–1101. [Google Scholar] [CrossRef]
  126. 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-Oncology 2016, 18, 549–556. [Google Scholar] [CrossRef]
  127. Neville, I.S.; Dos Santos, A.G.; Almeida, C.C.; Abaurre, L.B.; Wayhs, S.Y.; Feher, O.; Teixeira, M.J.; Lepski, G. Reoperation for recurrent glioblastomas: What to expect? Surg. Neurol. Int. 2021, 12, 42. [Google Scholar] [CrossRef]
  128. Li, Y.; Rey-Dios, R.; Roberts, D.W.; Valdés, P.A.; Cohen-Gadol, A.A. Intraoperative fluorescence-guided resection of high-grade gliomas: A comparison of the present techniques and evolution of future strategies. World Neurosurg. 2014, 82, 175–185. [Google Scholar] [CrossRef]
  129. McCracken, D.J.; Schupper, A.J.; Lakomkin, N.; Malcolm, J.; Painton Bray, D.; Hadjipanayis, C.G. Turning on the light for brain tumor surgery: A 5-aminolevulinic acid story. Neuro Oncol. 2022, 24, S52–S61. [Google Scholar] [CrossRef]
  130. Roberts, D.W.; Valdés, P.A.; Harris, B.T.; Fontaine, K.M.; Hartov, A.; Fan, X.; Ji, S.; Lollis, S.S.; Pogue, B.W.; Leblond, F.; et al. Coregistered fluorescence-enhanced tumor resection of malignant glioma: Relationships between δ-aminolevulinic acid-induced protoporphyrin IX fluorescence, magnetic resonance imaging enhancement, and neuropathological parameters. Clinical article. J. Neurosurg. 2011, 114, 595–603. [Google Scholar] [CrossRef]
  131. Roberts, D.W.; Valdés, P.A.; Harris, B.T.; Hartov, A.; Fan, X.; Ji, S.; Leblond, F.; Tosteson, T.D.; Wilson, B.C.; Paulsen, K.D. Glioblastoma multiforme treatment with clinical trials for surgical resection (aminolevulinic acid). Neurosurg. Clin. N. Am. 2012, 23, 371–377. [Google Scholar] [CrossRef]
  132. Valdés, P.A.; Moses, Z.B.; Kim, A.; Belden, C.J.; Wilson, B.C.; Paulsen, K.D.; Roberts, D.W.; Harris, B.T. Gadolinium- and 5-aminolevulinic acid-induced protoporphyrin IX levels in human gliomas: An ex vivo quantitative study to correlate protoporphyrin IX levels and blood-brain barrier breakdown. J. Neuropathol. Exp. Neurol. 2012, 71, 806–813. [Google Scholar] [CrossRef]
  133. Stummer, W.; Stocker, S.; Wagner, S.; Stepp, H.; Fritsch, C.; Goetz, C.; Goetz, A.E.; Kiefmann, R.; Reulen, H.J. Intraoperative detection of malignant gliomas by 5-aminolevulinic acid-induced porphyrin fluorescence. Neurosurgery 1998, 42, 518–525; discussion 525–516. [Google Scholar] [CrossRef]
  134. Valdés, P.A.; Kim, A.; Brantsch, M.; Niu, C.; Moses, Z.B.; Tosteson, T.D.; Wilson, B.C.; Paulsen, K.D.; Roberts, D.W.; Harris, B.T. δ-aminolevulinic acid-induced protoporphyrin IX concentration correlates with histopathologic markers of malignancy in human gliomas: The need for quantitative fluorescence-guided resection to identify regions of increasing malignancy. Neuro Oncol. 2011, 13, 846–856. [Google Scholar] [CrossRef]
  135. Valdés, P.A.; Kim, A.; Leblond, F.; Conde, O.M.; Harris, B.T.; Paulsen, K.D.; Wilson, B.C.; Roberts, D.W. Combined fluorescence and reflectance spectroscopy for in vivo quantification of cancer biomarkers in low- and high-grade glioma surgery. J. Biomed. Opt. 2011, 16, 116007. [Google Scholar] [CrossRef]
  136. Hickmann, A.K.; Nadji-Ohl, M.; Hopf, N.J. Feasibility of fluorescence-guided resection of recurrent gliomas using five-aminolevulinic acid: Retrospective analysis of surgical and neurological outcome in 58 patients. J. Neurooncol 2015, 122, 151–160. [Google Scholar] [CrossRef]
  137. Kamp, M.A.; Felsberg, J.; Sadat, H.; Kuzibaev, J.; Steiger, H.J.; Rapp, M.; Reifenberger, G.; Dibué, M.; Sabel, M. 5-ALA-induced fluorescence behavior of reactive tissue changes following glioblastoma treatment with radiation and chemotherapy. Acta Neurochir. 2015, 157, 207–213; discussion 213–204. [Google Scholar] [CrossRef]
  138. Fisher, J.P.; Adamson, D.C. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021, 9, 324. [Google Scholar] [CrossRef]
  139. Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 3801. [Google Scholar] [CrossRef]
  140. Weenink, B.; French, P.J.; Sillevis Smitt, P.A.E.; Debets, R.; Geurts, M. Immunotherapy in Glioblastoma: Current Shortcomings and Future Perspectives. Cancers 2020, 12, 751. [Google Scholar] [CrossRef]
  141. 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. 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]
  142. Schalper, K.A.; Rodriguez-Ruiz, M.E.; Diez-Valle, R.; López-Janeiro, A.; Porciuncula, A.; Idoate, M.A.; Inogés, S.; Andrea, C.; Cerio, A.; Tejada, S. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat. Med. 2019, 25, 470–476. [Google Scholar] [CrossRef]
  143. Reardon, D.A.; Brandes, A.A.; Omuro, A.; Mulholland, P.; Lim, M.; Wick, A.; Baehring, J.; Ahluwalia, M.S.; Roth, P.; Bähr, O. 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]
  144. Kurz, S.C.; Cabrera, L.P.; Hastie, D.; Huang, R.; Unadkat, P.; Rinne, M.; Nayak, L.; Lee, E.Q.; Reardon, D.A.; Wen, P.Y. PD-1 inhibition has only limited clinical benefit in patients with recurrent high-grade glioma. Neurology 2018, 91, 1355–1359. [Google Scholar] [CrossRef]
  145. Chiocca, E.A.; Gelb, A.B.; Chen, C.C.; Rao, G.; Reardon, D.A.; Wen, P.Y.; Bi, W.L.; Peruzzi, P.; Amidei, C.; Triggs, D. Combined immunotherapy with controlled interleukin-12 gene therapy and immune checkpoint blockade in recurrent glioblastoma: An open-label, multi-institutional phase I trial. Neuro-Oncology 2022, 24, 951–963. [Google Scholar] [CrossRef]
  146. Lee, A.H.; Sun, L.; Mochizuki, A.Y.; Reynoso, J.G.; Orpilla, J.; Chow, F.; Kienzler, J.C.; Everson, R.G.; Nathanson, D.A.; Bensinger, S.J. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma. Nat. Commun. 2021, 12, 6938. [Google Scholar] [CrossRef]
  147. 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]
  148. 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]
  149. Reardon, D.; Kaley, T.; Iwamoto, F.; Baehring, J.; Subramaniam, D.; Rawls, T.; He, Y.; Keler, T.; Yellin, M. ATIM-23. Anti-cd27 agonist antibody varlilumab in combination with nivolumab for recurrent glioblastoma (rgbm): Phase 2 clinical trial results. Neuro-Oncology 2018, 20, vi6. [Google Scholar] [CrossRef]
  150. Bagley, S.J.; Desai, A.S.; Linette, G.P.; June, C.H.; O’Rourke, D.M.C.A.R. T-cell therapy for glioblastoma: Recent clinical advances and future challenges. Neuro-Oncology 2018, 20, 1429–1438. [Google Scholar] [CrossRef]
  151. 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]
  152. Eskilsson, E.; Røsland, G.V.; Solecki, G.; Wang, Q.; Harter, P.N.; Graziani, G.; Verhaak, R.G.W.; Winkler, F.; Bjerkvig, R.; Miletic, H. EGFR heterogeneity and implications for therapeutic intervention in glioblastoma. Neuro-Oncology 2018, 20, 743–752. [Google Scholar] [CrossRef]
  153. O’Rourke, D.M.; Nasrallah, M.P.; Desai, A.; Melenhorst, J.J.; Mansfield, K.; Morrissette JJ, 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]
  154. Yang, J.; Yan, J.; Liu, B. Targeting EGFRvIII for glioblastoma multiforme. Cancer Lett. 2017, 403, 224–230. [Google Scholar] [CrossRef]
  155. 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. = Biomed. Pharmacother. 2019, 113, 108734. [Google Scholar] [CrossRef]
  156. Durgin, J.S.; Henderson, F.J.; Nasrallah, M.P.; Mohan, S.; Wang, S.; Lacey, S.F.; Melenhorst, J.J.; Desai, A.S.; Lee, J.Y.K.; Maus, M.V. Case Report: Prolonged Survival Following EGFRvIII CAR T Cell Treatment for Recurrent Glioblastoma. Front. Oncol. 2021, 11, 669071. [Google Scholar] [CrossRef]
  157. Bagley, S.J.; Binder, Z.A.; Lamrani, L.; Marinari, E.; Desai, A.S.; Nasrallah, M.P.; Maloney, E.; Brem, S.; Lustig, R.A.; Kurtz, G.; et al. Repeated Peripheral Infusions of Anti-EGFRvIII CAR T Cells in Combination with Pembrolizumab Show No Efficacy in Glioblastoma: A Phase 1 Trial. Nat. Cancer 2024, 5, 517–531. [Google Scholar] [CrossRef]
  158. 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, 77769. [Google Scholar] [CrossRef]
  159. Brown, C.E.; Badie, B.; Barish, M.E.; Weng, L.; Ostberg, J.R.; Chang, W.C.; Naranjo, A.; Starr, R.; Wagner, J.; Wright, C. Bioactivity and Safety of IL13Rα2-Redirected Chimeric Antigen Receptor CD8+ T Cells in Patients with Recurrent Glioblastoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2015, 21, 4062–4072. [Google Scholar] [CrossRef]
  160. Tu, M.; Wange, W.; Cai, L.; Zhu, P.; Gao, Z.; Zheng, W. IL-13 receptor α2 stimulates human glioma cell growth and metastasis through the Src/PI3K/Akt/mTOR signaling pathway. Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 2016, 37, 14701–14709. [Google Scholar] [CrossRef]
  161. Thaci, B.; Brown, C.E.; Binello, E.; Werbaneth, K.; Sampath, P.; Sengupta, S. Significance of interleukin-13 receptor alpha 2-targeted glioblastoma therapy. Neuro-Oncology 2014, 16, 1304–1312. [Google Scholar] [CrossRef]
  162. Brown, C.E.; Hibbard, J.C.; Alizadeh, D.; Blanchard, M.S.; Natri, H.M.; Wang, D.; Ostberg, J.R.; Aguilar, B.; Wagner, J.R.; Paul, J.A.; et al. Locoregional delivery of IL-13Rα2-targeting CAR-T cells in recurrent high-grade glioma: A phase 1 trial. Nat. Med. 2024, 30, 1001–1012. [Google Scholar] [CrossRef]
  163. Choi, B.D.; Gerstner, E.R.; Frigault, M.J.; Leick, M.B.; Mount, C.W.; Balaj, L.; Nikiforow, S.; Carter, B.S.; Curry, W.T.; Gallagher, K.; et al. Intraventricular CARv3-TEAM-E T Cells in Recurrent Glioblastoma. N. Engl. J. Med. 2024, 390, 1290–1298. [Google Scholar] [CrossRef]
  164. Bagley, S.J.; Logun, M.; Fraietta, J.A.; Wang, X.; Desai, A.S.; Bagley, L.J.; Nabavizadeh, A.; Jarocha, D.; Martins, R.; Maloney, E.; et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: Phase 1 trial interim results. Nat. Med. 2024, 30, 1320–1329. [Google Scholar] [CrossRef]
  165. Wang, M.; Zhou, Z.; Wang, X.; Zhang, C.; Jiang, X. Natural killer cell awakening: Unleash cancer-immunity cycle against glioblastoma. Cell Death Dis. 2022, 13, 588. [Google Scholar] [CrossRef]
  166. Natural killer cells for cancer immunotherapy: A new CAR is catching up. EBioMedicine 2019, 39, 1–2. [CrossRef]
  167. Murakami, T.; Nakazawa, T.; Natsume, A.; Nishimura, F.; Nakamura, M.; Matsuda, R.; Omoto, K.; Tanaka, Y.; Shida, Y.; Park, Y.S. Novel Human NK Cell Line Carrying CAR Targeting EGFRvIII Induces Antitumor Effects in Glioblastoma Cells. Anticancer. Res. 2018, 38, 5049–5056. [Google Scholar] [CrossRef]
  168. Zhang, C.; Burger, M.C.; Jennewein, L.; Genßler, S.; Schönfeld, K.; Zeiner, P.; Hattingen, E.; Harter, P.N.; Mittelbronn, M.; Tonn, T. ErbB2/HER2-Specific NK Cells for Targeted Therapy of Glioblastoma. J. Natl. Cancer Inst. 2016, 108, djv375. [Google Scholar] [CrossRef]
  169. Peruzzi, P.P.; Chiocca, E.A. Cancer immunotherapy: A vaccine from plant virus proteins. Nat. Nanotechnol. 2016, 11, 214–215. [Google Scholar] [CrossRef]
  170. Bernstock, J.D.; Blitz, S.E.; Hoffman, S.E.; Gerstl, J.V.E.; Chiocca, E.A.; Friedman, G.K. Recent oncolytic virotherapy clinical trials outline a roadmap for the treatment of high-grade glioma. Neuro-Oncol. Adv. 2023, 5, vdad081. [Google Scholar] [CrossRef]
  171. 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]
  172. Ballman, K.V.; Buckner, J.C.; Brown, P.D.; Giannini, C.; Flynn, P.J.; LaPlant, B.R.; Jaeckle, K.A. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro-Oncology 2007, 9, 29–38. [Google Scholar] [CrossRef]
  173. Ling, A.L.; Solomon, I.H.; Landivar, A.M.; Nakashima, H.; Woods, J.K.; Santos, A.; Masud, N.; Fell, G.; Mo, X.; Yilmaz, A.S.; et al. Clinical trial links oncolytic immunoactivation to survival in glioblastoma. Nature 2023, 623, 157–166. [Google Scholar] [CrossRef]
  174. 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]
  175. 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]
  176. Friedman, G.K.; Johnston, J.M.; Bag, A.K.; Bernstock, J.D.; Li, R.; Aban, I.; Kachurak, K.; Nan, L.; Kang, K.-D.; Totsch, S.; et al. Oncolytic HSV-1 G207 Immunovirotherapy for Pediatric High-Grade Gliomas. N. Engl. J. Med. 2021, 384, 1613–1622. [Google Scholar] [CrossRef]
  177. Harrow, S.; Papanastassiou, V.; Harland, J.; Mabbs, R.; Petty, R.; Fraser, M.; Hadley, D.; Patterson, J.; Brown, S.M.; Rampling, R. HSV1716 injection into the brain adjacent to tumour following surgical resection of high-grade glioma: Safety data and long-term survival. Gene Ther. 2004, 11, 1648–1658. [Google Scholar] [CrossRef]
  178. Papanastassiou, V.; Rampling, R.; Fraser, M.; Petty, R.; Hadley, D.; Nicoll, J.; Harland, J.; Mabbs, R.; Brown, M. The potential for efficacy of the modified (ICP 34.5(-)) herpes simplex virus HSV1716 following intratumoural injection into human malignant glioma: A proof of principle study. Gene Ther. 2002, 9, 398–406. [Google Scholar] [CrossRef]
  179. Rampling, R.; Cruickshank, G.; Papanastassiou, V.; Nicoll, J.; Hadley, D.; Brennan, D.; Petty, R.; MacLean, A.; Harland, J.; McKie, E.; et al. Toxicity evaluation of replication-competent herpes simplex virus (ICP 34.5 null mutant 1716) in patients with recurrent malignant glioma. Gene Ther. 2000, 7, 859–866. [Google Scholar] [CrossRef]
  180. 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]
  181. 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. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 1419–1427. [Google Scholar] [CrossRef]
  182. Samson, A.; Scott, K.J.; Taggart, D.; West, E.J.; Wilson, E.; Nuovo, G.J.; Thomson, S.; Corns, R.; Mathew, R.K.; Fuller, M.J.; et al. Intravenous delivery of oncolytic reovirus to brain tumor patients immunologically primes for subsequent checkpoint blockade. Sci. Transl. Med. 2018, 10, eaam7577. [Google Scholar] [CrossRef]
  183. 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]
  184. Forsyth, P.; Roldán, G.; George, D.; Wallace, C.; Palmer, C.A.; Morris, D.; Cairncross, G.; Matthews, M.V.; Markert, J.; Gillespie, Y.; et al. A phase I trial of intratumoral administration of reovirus in patients with histologically confirmed recurrent malignant gliomas. Mol. Ther. 2008, 16, 627–632. [Google Scholar] [CrossRef]
  185. 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]
  186. Desjardins, A.; Gromeier, M.; Herndon, J.E., 2nd; 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]
  187. Karandikar, P.V.; Suh, L.; Gerstl, J.V.E.; Blitz, S.E.; Qu, Q.R.; Won, S.Y.; Gessler, F.A.; Arnaout, O.; Smith, T.R.; Peruzzi, P.P. Positioning SUMO as an immunological facilitator of oncolytic viruses for high-grade glioma. Front. Cell Dev. Biol. 2023, 11, 1271575. [Google Scholar] [CrossRef]
  188. Boockvar, J.A.; Tsiouris, A.J.; Hofstetter, C.P.; Kovanlikaya, I.; Fralin, S.; Kesavabhotla, K.; Seedial, S.M.; Pannullo, S.C.; Schwartz, T.H.; Stieg, P. Safety and maximum tolerated dose of superselective intraarterial cerebral infusion of bevacizumab after osmotic blood-brain barrier disruption for recurrent malignant glioma. Clin. Article. J. Neurosurg. 2011, 114, 624–632. [Google Scholar] [CrossRef]
  189. 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 Off. J. Control. Release Soc. 2019, 309, 37–47. [Google Scholar] [CrossRef]
  190. Solinge, T.S.; Nieland, L.; Chiocca, E.A.; Broekman, M.L.D. Advances in local therapy for glioblastoma—Taking the fight to the tumour. Nat. Rev. Neurol. 2022, 18, 221–236. [Google Scholar] [CrossRef]
  191. Bernstock, J.D.; Blitz, S.; Kang, K.D.; Friedman, G.K. Intraventricular immunovirotherapy; a translational step forward. Oncotarget 2023, 14, 40–43. [Google Scholar] [CrossRef]
  192. Kang, K.D.; Bernstock, J.D.; Totsch, S.K.; Gary, S.E.; Rocco, A.; Nan, L.; Li, R.; Etminan, T.; Han, X.; Beierle, E.A. Safety and Efficacy of Intraventricular Immunovirotherapy with Oncolytic HSV-1 for CNS Cancers. Clin. Cancer Res. 2022, 28, 5419–5430. [Google Scholar] [CrossRef]
  193. Plaksin, M.; Bercovici, T.; Sat Toltsis, G.G.; Grinfeld, J.; Shapira, B.; Zur, Y.; Picciotto, R.; Zadicario, E.; Siddeeq, M.; Wohl, A. Magnetic resonance imaging analysis predicts nanoparticle concentration delivered to the brain parenchyma. Commun. Biol. 2022, 5, 964. [Google Scholar] [CrossRef]
  194. Burgess, A.; Shah, K.; Hough, O.; Hynynen, K. Focused ultrasound-mediated drug delivery through the blood-brain barrier. Expert. Rev. Neurother. 2015, 15, 477–491. [Google Scholar] [CrossRef]
  195. Abrahao, A.; Meng, Y.; Llinas, M.; Huang, Y.; Hamani, C.; Mainprize, T.; Aubert, I.; Heyn, C.; Black, S.E.; Hynynen, K. First-in-human trial of blood-brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound. Nat. Commun. 2019, 10, 4373. [Google Scholar] [CrossRef]
  196. Zhang, Y.; Wang, J.; Ghobadi, S.N.; Zhou, H.; Huang, A.; Gerosa, M.; Hou, Q.; Keunen, O.; Golebiewska, A.; Habte, F.G. Molecular Identity Changes of Tumor-Associated Macrophages and Microglia After Magnetic Resonance Imaging-Guided Focused Ultrasound-Induced Blood-Brain Barrier Opening in a Mouse Glioblastoma Model. Ultrasound Med. Biol. 2023, 49, 1082–1090. [Google Scholar] [CrossRef]
  197. Wu, Y.; Liu, Y.; Huang, Z.; Wang, X.; Jin, Z.; Li, J.; Limsakul, P.; Zhu, L.; Allen, M.; Pan, Y. Control of the activity of CAR-T cells within tumours via focused ultrasound. Nat. Biomed. Eng. 2021, 5, 1336–1347. [Google Scholar] [CrossRef]
  198. Jain, A.; Garg, N.K.; Tyagi, R.K.; Singh, B.; Katare, O.P.; Webster, T.J.; Soni, V. Surface engineered polymeric nanocarriers mediate the delivery of transferrin-methotrexate conjugates for an improved understanding of brain cancer. Acta Biomater. 2015, 24, 140–151. [Google Scholar] [CrossRef]
  199. Abdul Razzak, R.; Florence, G.J.; Gunn-Moore, F.J. Approaches to CNS Drug Delivery with a Focus on Transporter-Mediated Transcytosis. Int. J. Mol. Sci. 2019, 20, 3108. [Google Scholar] [CrossRef]
  200. 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]
  201. Lu, G.; Wang, X.; Li, F.; Wang, S.; Zhao, J.; Wang, J.; Liu, J.; Lyu, C.; Ye, P.; Tan, H. Engineered biomimetic nanoparticles achieve targeted delivery and efficient metabolism-based synergistic therapy against glioblastoma. Nat. Commun. 2022, 13, 4214. [Google Scholar] [CrossRef]
  202. Kumthekar, P.; Ko, C.H.; Paunesku, T.; Dixit, K.; Sonabend, A.M.; Bloch, O.; Tate, M.; Schwartz, M.; Zuckerman, L.; Lezon, R. 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]
  203. Hanson, L.R.; Frey, W. Intranasal delivery bypasses the blood-brain barrier to target therapeutic agents to the central nervous system and treat neurodegenerative disease. BMC Neurosci. 2008, 9 (Suppl. S3), 5. [Google Scholar] [CrossRef]
  204. Mohammadi, A.M.; Hawasli, A.H.; Rodriguez, A.; Schroeder, J.L.; Laxton, A.W.; Elson, P.; Tatter, S.B.; Barnett, G.H.; Leuthardt, E.C. The role of laser interstitial thermal therapy in enhancing progression-free survival of difficult-to-access high-grade gliomas: A multicenter study. Cancer Med. 2014, 3, 971–979. [Google Scholar] [CrossRef]
  205. Watanabe, M.; Tanaka, R.; Hondo, H.; Kuroki, M. Effects of antineoplastic agents and hyperthermia on cytotoxicity toward chronically hypoxic glioma cells. Int. J. Hyperth. Off. J. Eur. Soc. Hyperthermic Oncol. N. Am. Hyperth. Group. 1992, 8, 131–138. [Google Scholar] [CrossRef]
  206. Schildkopf, P.; Ott, O.J.; Frey, B.; Wadepohl, M.; Sauer, R.; Fietkau, R.; Gaipl, U.S. Biological rationales and clinical applications of temperature controlled hyperthermia–implications for multimodal cancer treatments. Curr. Med. Chem. 2010, 17, 3045–3057. [Google Scholar] [CrossRef]
  207. Frey, B.; Weiss, E.M.; Rubner, Y.; Wunderlich, R.; Ott, O.J.; Sauer, R.; Fietkau, R.; Gaipl, U.S. Old and new facts about hyperthermia-induced modulations of the immune system. Int. J. Hyperth. Off. J. Eur. Soc. Hyperthermic Oncol. N. Am. Hyperth. Group. 2012, 28, 528–542. [Google Scholar] [CrossRef]
  208. Man, J.; Shoemake, J.D.; Ma, T.; Rizzo, A.E.; Godley, A.R.; Wu, Q.; Mohammadi, A.M.; Bao, S.; Rich, J.N.; Yu, J.S. Hyperthermia Sensitizes Glioma Stem-like Cells to Radiation by Inhibiting AKT Signaling. Cancer Res. 2015, 75, 1760–1769. [Google Scholar] [CrossRef]
  209. Hoover, J.M.; Nwojo, M.; Puffer, R.; Mandrekar, J.; Meyer, F.B.; Parney, I.F. Surgical outcomes in recurrent glioma: Clinical article. J. Neurosurg. 2013, 118, 1224–1231. [Google Scholar] [CrossRef]
  210. Biase, G.; Garcia, D.P.; Bohnen, A.; Quiñones-Hinojosa, A. Perioperative Management of Patients with Glioblastoma. Neurosurg. Clin. N. Am. 2021, 32, 1–8. [Google Scholar] [CrossRef]
  211. Fabian, D.; Guillermo Prieto Eibl, M.D.P.; Alnahhas, I.; Sebastian, N.; Giglio, P.; Puduvalli, V.; Gonzalez, J.; Palmer, J.D. Treatment of Glioblastoma (GBM) with the Addition of Tumor-Treating Fields (TTF): A Review. Cancers 2019, 11, 174. [Google Scholar] [CrossRef]
  212. Kirson, E.D.; Gurvich, Z.; Schneiderman, R.; Dekel, E.; Itzhaki, A.; Wasserman, Y.; Schatzberger, R.; Palti, Y. Disruption of cancer cell replication by alternating electric fields. Cancer Res. 2004, 64, 3288–3295. [Google Scholar] [CrossRef]
  213. Dono, A.; Mitra, S.; Shah, M.; Takayasu, T.; Zhu, J.J.; Tandon, N.; Patel, C.B.; Esquenazi, Y.; Ballester, L.Y. PTEN mutations predict benefit from tumor treating fields (TTFields) therapy in patients with recurrent glioblastoma. J. Neurooncol 2021, 153, 153–160. [Google Scholar] [CrossRef]
  214. Stupp, R.; Wong, E.T.; Kanner, A.A.; Steinberg, D.; Engelhard, H.; Heidecke, V.; Kirson, E.D.; Taillibert, S.; Liebermann, F.; Dbaly, 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]
  215. Mrugala, M.M.; Engelhard, H.H.; Dinh Tran, D.; Kew, Y.; Cavaliere, R.; Villano, J.L.; Annenelie Bota, D.; Rudnick, J.; Love Sumrall, A.; Zhu, J.J.; et al. Clinical practice experience with NovoTTF-100A system for glioblastoma: The Patient Registry Dataset (PRiDe). Semin. Oncol. 2014, 41 (Suppl. S6), S4–S13. [Google Scholar] [CrossRef]
  216. 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]
  217. 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]
  218. Cramer, S.W.; Chen, C.C. Photodynamic Therapy for the Treatment of Glioblastoma. Front. Surg. 2019, 6, 81. [Google Scholar] [CrossRef]
  219. Kwiatkowski, S.; Knap, B.; Przystupski, D.; Saczko, J.; Kędzierska, E.; Knap-Czop, K.; Kotlińska, J.; Michel, O.; Kotowski, K.; Kulbacka, J. Photodynamic therapy—Mechanisms, photosensitizers and combinations. Biomed. Pharmacother. 2018, 106, 1098–1107. [Google Scholar] [CrossRef]
  220. Vermandel, M.; Dupont, C.; Lecomte, F.; Leroy, H.A.; Tuleasca, C.; Mordon, S.; Hadjipanayis, C.G.; Reyns, N. Standardized intraoperative 5-ALA photodynamic therapy for newly diagnosed glioblastoma patients: A preliminary analysis of the INDYGO clinical trial. J. Neurooncol. 2021, 152, 501–514. [Google Scholar] [CrossRef]
  221. Akimoto, J.; Haraoka, J.; Aizawa, K. Preliminary clinical report on safety and efficacy of photodynamic therapy using talaporfin sodium for malignant gliomas. Photodiagnosis Photodyn. Ther. 2012, 9, 91–99. [Google Scholar] [CrossRef]
  222. Beck, T.J.; Kreth, F.W.; Beyer, W.; Mehrkens, J.H.; Obermeier, A.; Stepp, H.; Stummer, W.; Baumgartner, R. Interstitial photodynamic therapy of nonresectable malignant glioma recurrences using 5-aminolevulinic acid induced protoporphyrin IX. Lasers Surg. Med. 2007, 39, 386–393. [Google Scholar] [CrossRef]
  223. Johansson, A.; Faber, F.; Kniebühler, G.; Stepp, H.; Sroka, R.; Egensperger, R.; Beyer, W.; Kreth, F.W. Protoporphyrin IX fluorescence and photobleaching during interstitial photodynamic therapy of malignant gliomas for early treatment prognosis. Lasers Surg. Med. 2013, 45, 225–234. [Google Scholar] [CrossRef]
  224. 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]
  225. Peruzzi, P.; Dominas, C.; Fell, G.; Bernstock, J.D.; Blitz, S.; Mazzetti, D.; Zdioruk, M.; Dawood, H.Y.; Triggs, D.V.; Ahn, S.W. Intratumoral drug-releasing microdevices allow in situ high-throughput pharmaco phenotyping in patients with gliomas. Sci. Transl. Med. 2023, 15, eadi0069. [Google Scholar] [CrossRef]
Figure 1. Molecular, epigenetic, and extracellular pathways in recurrent glioblastoma.
Figure 1. Molecular, epigenetic, and extracellular pathways in recurrent glioblastoma.
Ijms 25 06733 g001
Table 1. Molecular, epigenetic, and paracrine signaling in recurrent glioblastoma.
Table 1. Molecular, epigenetic, and paracrine signaling in recurrent glioblastoma.
Gene, Epigenetic Modification, or Paracrine SignalingMutational Characteristics in
Recurrent Glioblastoma
References
TERTHighly conserved mutation with the TERT promoter as the most conserved mutation
Correlated with EGFR amplifications, leading to tumor survival and progression
[27]
TERT C228T mutation confers poorer prognosis than C250T[28]
PTEN aberrations lead to microtubule disruptions, resulting in improper cell cycle regulation and mitosis[29]
EGFREGFRvIII results in truncated receptor co-expressed with the wtEGFR, which leads to constitutively activated downstream EGFR signaling
EGFRvIII specific signaling includes DOCK1 and SFK pathways
[30,31,32,33]
PI3K/AktPI3K-activating mutations have been found in 91% of recurrent glioblastomas[34]
Unregulated cell cycle progression[34,35]
lncRNA upregulates PI3K and leads to stem cell migration[21,36]
Glioblastoma-derived EVs upregulate PI3K/Akt proliferation and migratory signaling[37]
PDGF-R α/βHigh expression of PDGF-R β maintains GSC renewal and survival[38]
Knockdown of PDGF-R α/β promotes neurites differentiation in GSC and glioblastoma cell lines[39]
Paracrine signaling via macrophagesCL2 and CSF1 recruit peripheral macrophages [40,41,42,43]
IL-10 and TGF-β signaling polarize TAMs to anti-inflammatory M2[44,45,46]
M2 inhibit Th-1 and NK cell responses[47]
Epigenetic modificationsMSH6, MLH, and LTBP4 lead to resistance to TMZ[21,26]
LTBP4 mutations lead IDH upregulation and increased cell proliferation [21]
ALKBH5 inhibits CXCL8 in hypoxic TME, leading to GSC proliferation[48,49]
Demethylation of MGMT conferred to decreased TMZ sensitivity and shorter survival[50]
MGMT promoter methylation is decreased in recurrent glioblastoma when compared with newly diagnosed glioblastoma[51,52]
TERT = telomerase reverse transcriptase; EGFR = epidermal growth factor receptor; PTEN = phosphatase and tensin homolog; EGFRvIII = mutated EGFR; DOCK1 = dedicator of cytokinesis 1; SFK = SRC family kinases; PI3K = phosphoinositide 3 kinase; lncRNA = long non-coding RNA; EVs = extracellular vesicles; PDGF-R = platelet-derived growth factor receptor; GSC = glioblastoma stem cell; CSF1 = colony-stimulating factor 1; TGF-β = transforming growth factor β; TAM = tumor-associated macrophage; LTBP4 = latent transforming growth factor beta binding protein 4; IDH = isocitrate dehydrogenase; TME = tumor microenvironment; TMZ = temozolomide; MGMT = O(6)-methylguanine-DNA-methyltransferase.
Table 2. Published phase 2 and 3 immune checkpoint inhibitor trials in recurrent glioblastoma.
Table 2. Published phase 2 and 3 immune checkpoint inhibitor trials in recurrent glioblastoma.
Author/Trial Name/IdentifierAgent/ModalityStudy PhaseStudy Outcome
Reardon et al., 2020
[143], CheckMate-143, NCT02017717
Nivolumab vs. bevacizumabPhase 3mPFS = 1.5 months (nivolumab) vs. 3.5 months (bevacizumab);
mOS = 9.8 months (nivolumab) vs. 10.0 months (bevacizumab).
Nayak et al., 2021 [148],
NCT02337491
Pembrolizumab + bevacizumab vs. pembrolizumab monotherapyPhase 2PFS-6 = 26.0% (pembrolizumab + bevacizumab) vs. 6.7% (pembrolizumab monotherapy);
mOS = 8.8 months (pembrolizumab + bevacizumab) vs. 10.3 months (pembrolizumab monotherapy);
ORR = 20% (pembrolizumab + bevacizumab) vs. 0% (pembrolizumab monotherapy).
Cloughesy et al., 2019 [141]Pembrolizumab (neoadjuvant + adjuvant) vs. pembrolizumab (adjuvant only)Phase 2mPFS = 3.3 months (neoadjuvant + adjuvant) vs. 2.4 months (adjuvant only);
mOS = 13.7 months (neoadjuvant + adjuvant) vs. 7.5 months (adjuvant only).
Schalper et al., 2019 [142],
NCT02550249
Neoadjuvant nivolumab (single arm)Phase 2mPFS = 4.1 months;
mOS = 7.3 months;
Higher immune cell infiltration and TCR diversity.
Reardon et al., 2018 [149], NCT02335918Nivolumab + varlilumabPhase 2mOS = 9.7 months.
mPFS = median progression-free survival; mOS = median overall survival; PFS-6 = 6-month progression free survival; ORR = objective response rate; TCR = T cell receptor.
Table 3. Ongoing immune checkpoint inhibitor trials in recurrent glioblastoma.
Table 3. Ongoing immune checkpoint inhibitor trials in recurrent glioblastoma.
Author/Trial Name/IdentifierStatusAgent/ModalityPrimary ObjectiveStudy Phase
NCT04145115Recruiting Ipilimumab + nivolumabTumor response by modified RANO in recurrent glioblastoma with high mutational burden.Phase 2
NCT04323046Active, not recruitingIpilimumab + nivolumabTME changes following neoadjuvant nivolumab and placebo, ipilimumab and placebo, and nivolumab and ipilimumab.
Safety and tolerability of neoadjuvant nivolumab and placebo, ipilimumab and placebo, and nivolumab and ipilimumab.
Phase 1
NCT03890952 Active, not recruitingNivolumab + bevacizumab (BEV) Number of indels as determined using mRNA sequencing.Phase 2
NCT04201873RecruitingPembrolizumab + ATL-DC vaccineInfluence of pembrolizumab on cell cycle-related genetic signatures within the TME.
Influence of ATL-DC vaccination on peripheral T cell response.
Safety/tolerability of pembrolizumab and ATL-DC vaccination.
Phase 1
NCT04013672 Active, not recruiting SurVaxM + Sargramostim + Montanide ISA 51 PFS-6Phase 2
NCT04013672 Active, not recruiting SurVaxM + Sargramostim + Montanide ISA 51PFS-6Phase 2
NCT05465954Recruiting Pembrolizumab + efineptakin alfa OS-9Phase 2
NCT05465954Recruiting Pembrolizumab + efineptakin alfa OS-9Phase 2
NCT04479241 Active, not recruiting Pembrolizumab + lerapolturev ORR; DOR; DRRPhase 2
NCT05053880 Recruiting Pembrolizumab + ACT001 Adverse events; PFS-6Phase 1b/2
NCT05463848 Recruiting Pembrolizumab + Olaparib + Temozolomide TIL density; PFS-6Phase 2
RANO = Response Assessment in Neuro-Oncology; TME = tumor microenvironment; ATL-DC = adjuvant autologous tumor lysate dendritic cell; PFS-6 = 6-month progression-free survival; OS-9 = 9-month overall survival; ORR = objective response rate; DOR = duration of response; DRR = durable radiographic response; TIL = tumor-infiltrating lymphocyte.
Table 4. Ongoing CAR-T cell trials in recurrent glioblastoma.
Table 4. Ongoing CAR-T cell trials in recurrent glioblastoma.
Trial IdentifierStatusAgent/ModalityStudy PhaseTreatment Approach
NCT01454596CompletedEGFRvIII CAR transduced PBLPhase 3Infusion
NCT00730613CompletedAutologous lymphocytesPhase 1Leukapheresis + infusion
NCT01109095CompletedHER-2 CAR CMV-specific CTLsPhase 1Infusion
NCT02208362Active, not recruitingIL13Ralpha2-specific Hinge-optimized 41BB co-stimulatory CAR truncated CD-19 expressing autologous TN/MEM cellsPhase 1Intratumoral catheter
NCT05241392RecruitingB7-H3 targeting CAR-T cellsPhase 1Ommaya device
NCT04077866 Recruiting B7-H3 CAR-T + TMZPhase 1/2ICT/ICV injection (B7-H3 CAR-T) + Oral (TMZ)
NCT05366179Recruiting CAR B7-H3T cellsPhase 1ICV
NCT05474378Recruiting B7-H3CARTPhase 1ICV or dual ICV/ICT
NCT04214392Recruiting Chlorotoxin (EQ)-CD28-CD3zeta-CD19t-expressing CAR T-lymphocytePhase 1 ICT or dual ICT/ICV
NCT03389230 Active, not recruitingHER2(EQ)BBZ/CD19 + T cellsPhase 1Intratumoral/intracavitary
NCT05540873Recruiting IL13alpha2 CAR-T cellsPhase 1Infusion
NCT04003649 Recruiting IL13Ralpha2-specific Hinge-optimized 41BBco-stimulatory CAR/Truncated CD19 expressing autologous TN/MEM+Ipilimumab+nivolumabPhase 1ICV (IL13Ralpha2 CAR T)
IV (ipilimumab+nivoluma)
NCT05627323Recruiting CHM-1101 CAR-T cells Phase 1ICT/ICV dual delivery
NCT05353530 RecruitingEx-vivo expanded autologous IL-8 receptor (CXCR2) modified CD70 CAR (8R70CAR) T cellsPhase 1 Infusion
NCT05577091 Not yet recruiting Autologous Tris-CAR-T cells Phase 1 Infusion
NCT05168423RecruitingCART-EGFR-IL13Rα2Phase 1ICV
NCT05660369RecruitingCARv3-TEAM-E T cellsPhase 1ICV
ICV = intracerebroventricular; ICT = intracranial intratumoral or intracavitary.
Table 5. Ongoing NK cell therapy trials in recurrent glioblastoma.
Table 5. Ongoing NK cell therapy trials in recurrent glioblastoma.
Trial IdentifierStatusAgent/ModalityStudy PhaseTreatment Approach
NCT03383978Recruiting NK-92/5.28z + EzabenlimabPhase 1ICT (NK-92/5.28z), infusion (Ezabenlimab)
NCT04254419Not yet recruiting NK cellsPhase 1Infusion
NCT02100891 Active, not recruiting Donor NK cellPhase 2 Infusion
ICT = intracranial intratumoral or intracavitary.
Table 6. Published OV trials in recurrent glioblastoma.
Table 6. Published OV trials in recurrent glioblastoma.
Trial VirusStudy PhaseTreatment ApproachOutcomes
Todo et al., 2022 [171],
UMIN000015995
G47∆Phase 2 ICTmOS 20.02 months; OS-1yr = 84.2%
Ling et al., 2023 [173], NCT03152318CAN-3110Phase 1ICTmOS = 11.6 months
Markert et al., 2014 [174], NCT00157703G207Phase 1ICTmOS = 7.5 months;
no DLT
Markert et al., 2009 [175]G207Phase 1bICTmOS = 6.6 months;
1 case of transient fever, delirium, hemiparesis
Friedman et al., 2021 [176],
NCT02457845
G207Phase 1ICTmOS = 12.2 months;
radiographic, neuropathologica,l or clinical responses in 11/12 patients
Harrow et al., 2004 [177]HSV1716 Phase 1ICTNo DLT
Papanastassiou et al., 2002 [178]HSV1716Phase 1 ICTNo DLT
Rampling et al., 2000 [179]HSV1716Phase 1ICTNo DLT
Nassiri [147], NCT02798406DXN-2401 + PembrolizumabPhase 1/2ICT (DXN-2401); infusion (Pembrolizumab)No DLT; ORR = 10.4%; OS-1yr = 52.7%
Chiocca et al., 2004 [180]ONYX-015Phase 1ICTNo DLT; mOS = 6.2 months;
mPFS = 46 days
Lang et al., 2018 [181]DNX-2401Phase 1 Single ICT;
ICT followed by resection and injection into resection cavity
mOS (single ICT) = 9.3 months;
mOS (ICT + resection) = 13.0 months
Samson et al., 2018 [182]ReolysinPhase 1bIV infusion of 1010 TCID5Reovirus RNA extensively detected in tumor cells, increased levels of PD-1/PD-L1 in treated patients
Kicielinski et al., 2014 [183]ReolysinPhase 1ICTmOS = 4.6 months; no severe treatment related adverse events
Forsyth et al., 2008 [184]ReolysinPhase 1ICTmOS = 4.8 months
Geletneky et al., 2017 [185]Parvoryx01Phase 1/2(1) ICT followed by resection and injection around resection cavity; (2) Infusions for 5 days prior to resection and injection around resection cavity.mOS = 15.3 months
Desjardins et al., 2018 [186]PVSRIPOPhase 1ICTmOS = 12.5 months
ICT = intracranial intratumoral or intracavitary; mOS=median overall survival; OS-1yr = 1-year overall survival; DLT = dose-limiting toxicity; ORR = objective response rate; mPFS = median progression-free survival; TCID = 50% tissue culture infectious dose.
Table 7. Ongoing OV trials in recurrent glioblastoma.
Table 7. Ongoing OV trials in recurrent glioblastoma.
Trial StatusVirusStudy PhaseTreatment Approach
NCT04482933Not yet recruiting G207Phase 2 ICT
NCT02062827Active, not recruitingM032Phase 1 ICT
NCT03657576RecruitingC134Phase 1 ICT
NCT03896568Recruiting Allogeneic bone marrow-derived human mesenchymal stem cells loaded with DNX-2401Phase 1 Infusion
NCT03911388Recruiting G207Phase 1 ICT (cerebellum)
NCT03043391Active, not recruitingPVSRIPOPhase 1 CED
NCT01582516Completed DNX-2401Phase 1/2CED
NCT01956734Completed DNX-2401Phase 1ICT
NCT00390299Completed MV-CEAPhase 1ICT
NCT02986178Active, not recruiting PVSRIPO:Phase 2CED
NCT02197169 DNX-2401DNX-2401 with IFN-γPhase 1ICT
ICT = intracranial intratumoral or intracavitary; CED = convention-enhanced delivery.
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

Chang, C.; Chavarro, V.S.; Gerstl, J.V.E.; Blitz, S.E.; Spanehl, L.; Dubinski, D.; Valdes, P.A.; Tran, L.N.; Gupta, S.; Esposito, L.; et al. Recurrent Glioblastoma—Molecular Underpinnings and Evolving Treatment Paradigms. Int. J. Mol. Sci. 2024, 25, 6733. https://doi.org/10.3390/ijms25126733

AMA Style

Chang C, Chavarro VS, Gerstl JVE, Blitz SE, Spanehl L, Dubinski D, Valdes PA, Tran LN, Gupta S, Esposito L, et al. Recurrent Glioblastoma—Molecular Underpinnings and Evolving Treatment Paradigms. International Journal of Molecular Sciences. 2024; 25(12):6733. https://doi.org/10.3390/ijms25126733

Chicago/Turabian Style

Chang, Christopher, Velina S. Chavarro, Jakob V. E. Gerstl, Sarah E. Blitz, Lennard Spanehl, Daniel Dubinski, Pablo A. Valdes, Lily N. Tran, Saksham Gupta, Luisa Esposito, and et al. 2024. "Recurrent Glioblastoma—Molecular Underpinnings and Evolving Treatment Paradigms" International Journal of Molecular Sciences 25, no. 12: 6733. https://doi.org/10.3390/ijms25126733

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