Skip to main content
Neuro-Oncology Advances logoLink to Neuro-Oncology Advances
. 2025 Sep 9;7(Suppl 4):iv19–iv31. doi: 10.1093/noajnl/vdae204

The glioma microenvironment and its impact on antitumor immunity

Landon J Hansen 1, Christopher M Jackson 2,
PMCID: PMC12418595  PMID: 40933033

Abstract

Gliomas are a heterogeneous group of intrinsic brain tumors that are among the most difficult cancers to treat. Diffuse invasion into normal brain tissue prevents complete surgical resection; therefore, adjuvant therapy is necessary to curtail tumor progression and recurrence. High-grade, isocitrate dehydrogenase wild-type gliomas, also known as glioblastomas, are particularly resistant to treatment. Despite aggressive therapy with maximal safe resection, radiation, and chemotherapy, the median survival remains less than 2 years and has changed little in the past 2 decades. A major focus of therapeutic development for cancer treatment is immunotherapy, which aims to enhance the immune system’s ability to destroy tumor cells wherever they reside. While cancer immunotherapy has dramatically improved outcomes for patients with advanced melanoma, lung cancer, and many other malignancies, immunotherapies have not yet demonstrated the ability to reliably improve survival for glioblastoma patients. One of the fundamental challenges to developing effective immunotherapy for glioblastoma is the heterogenous and complex tumor microenvironment (TME), where there are multiple anatomic, molecular, and functional barriers to generating and sustaining antitumor immunity. Recent insights into the contributions of specific components of the glioma tumor microenvironment are leading the way from a trial-and-error approach to rationally targeted combination therapies. In this focused review, we discuss specific characteristics of the TME that impede immunotherapy for glioma and approaches in various stages of development aimed at overcoming these barriers.

Keywords: Immunotherapy, tumor microenvironment, anti-tumor immunity, tumor metabolism, immune checkpoints


Gliomas are the most common type of primary brain tumor and are characterized by their infiltrative nature and poor prognosis. These tumors impact tens of thousands of individuals worldwide each year and pose a substantial challenge to treatment due to their resistance to conventional therapies.1 Gliomas are classified based on genetic and histologic characteristics, with grade 4 lesions known as glioblastoma (GBM) being the most common and aggressive form. The median survival time for glioma patients varies from over 10 years for grade 2 oligodendrogliomas to only 15 months for patients diagnosed with GBM, even with comprehensive treatment involving maximum safe surgical resection, radiation, and chemotherapy.2 Gliomas are particularly difficult to treat due to multiple factors including the invasive dispersion of tumor cells, the delicate nature of the surrounding host organ, the unique immune environment of the CNS, the restricted access of the blood-brain barrier, and the inherent cellular resistance to therapy.

Given the diffusely infiltrative nature of this disease into the indispensable and poorly healing surrounding brain tissue, the precise and adaptive nature of immunotherapy is particularly appealing. Immunotherapy has revolutionized care for many cancer types and is now a mainstay of cancer therapy. The concept of arming the patient’s immune system with the tools necessary to defend itself against invasive and recalcitrant tumor cells has reshaped the way we understand and approach cancer.3 Despite recent progress, however, the promise of immunotherapy-based approaches for treating brain tumors remains to be fully realized. One of the chief reasons is that these tumors are profoundly immunosuppressive and employ nonredundant, parallel mechanisms to escape immune surveillance, recognition, and elimination.4–9

A tumor microenvironment conducive to potent and sustained antitumor immune responses is critical for effective immunotherapy. The glioma TME is a complex interaction of tumor cells, astrocytes, immune cells, fibroblasts, endothelial cells, growth factors, cytokines, and metabolites (Figure 1). This intricate ecosystem of signaling molecules and cellular cross-talk determines the likelihood of generating and sustaining antitumor immune responses. The general term “immune cells” in this instance includes a diverse array of tumor-infiltrating cell types including microglia, macrophages, myeloid-derived suppressor cells (MDSCs), neutrophils, dendritic cells, Tregs, CD4+ T cells, CD8+ T cells, natural killer (NK) cells, and B cells.10–16 In this focused review, we will discuss the most potent immunosuppressive mechanisms at work in the TME and explore strategies to shift the TME to an environment conducive to antitumor immune activity.

Figure 1.

Figure 1.

Schematic summarizing selected therapeutic strategies targeting the TME. Central to the schematic is the tumor cell which instigates the process of tumor formation and the creation of the tumor microenvironment. Cells that are recruited to the microenvironment include microglia, macrophages, T cells, NK cells, dendritic cells, reactive astrocytes, fibroblasts, and others. These cells secrete factors and components that influence each other and modify the extracellular matrix. Tumor cells also interact with neurons and form structural connections like synapses that facilitate tumor growth. Metabolic derangements result in autocrine and paracrine signaling in the microenvironment through secreted molecules and metabolites. Strategies to target glioma growth may focus on any of these aspects of the microenvironment, only a small sample of which are illustrated here. Created with BioRender.

Structural Components of the TME

Tumor Cells

The foundation of the brain TME is the tumor cells themselves. Glioma, like all cancers, is initiated by the underlying genetic alterations that enable deregulated cell growth and proliferation, transforming normal cells into tumor cells. This process of transformation allows tumor cells to ignore cues from their microenvironment that normally regulate cell growth, such as cellular density, nutrient and oxygen availability, growth factors (or the lack thereof), and extracellular matrix (ECM) support. The mutations and copy number alterations that arise in glioma have been particularly well-characterized, with distinct patterns of mutations specific to each of the glioma subtypes.

One of the defining genetic alterations of gliomas is the isocitrate dehydrogenase (IDH1) mutation. As of 2021, the World Health Organization classifies gliomas based on whether or not they have IDH mutations, due to the clear prognostic and diagnostic utility.17 IDH mutant tumors include astrocytoma, which also have mutations in ATRX and TP53, and oligodendroglioma, which has TERT promoter mutations and loss of chromosome arms 1p and 19q.17–20 IDH mutant tumors that also have CDKN2A/CDKN2B deletion or histologic characteristics of microvascular proliferation and necrosis behave more aggressively and are given a grade 4 designation. IDH wild-type (WT) tumors, which are the most aggressive, include GBM, and have mutations in the TERT promoter, loss of chromosomal arm 10q (PTEN gene locus), loss of the chromosome 9p21 housing tumor suppressor genes CDKN2A/CDKN2B and MTAP, amplification of chromosome 7, and frequent mutations in EGFR, TP53, PIK3CA, PTEN, AKT, and STAT. Most of these mutations function to deregulate the cell cycle controls, allowing unchecked tumor cell proliferation. Some genetic alterations also play a role in protecting tumor cells from the immune response and suppressing immune function, a requirement for tumor growth. Additional attention will be given to some of these immunosuppressive alterations in later sections.

While in many cancer types, a higher tumor mutational burden correlates with increased antigen recognition and improved immune response, the same association has not been found with GBM.21,22 In fact, some studies have shown the inverse, with high mutational burden associated with worse survival,23 although this is confounded by the fact that higher mutation number is also associated with older age and IDH WT status. One study, however, looked at patients that underwent immunotherapy with either immune checkpoint blockade or recombinant polio virotherapy and found that those with a very low tumor mutational burden had improved survival.24

The standard of care treatment for high-grade glioma is surgical resection followed by radiation and temozolomide chemotherapy.2 Temozolomide is a DNA alkylating agent that demonstrates single-agent antitumor activity against GBM. The mechanism of temozolomide is directly counted by the action of DNA-repair enzyme O6-methylguanine-DNA methyltransferase (MGMT), and levels of tumor MGMT expression and promoter methylation have been shown to correlate with patient survival.25 One of the main side effects of temozolomide is lymphopenia,26 with decreased circulating CD8+ T cells and NK cells, but increased Tregs.27,28 Interesting, despite T-cell depletion by temozolomide, patients coadministered temozolomide and an EGFRvIII vaccine was able to develop potent cellular and humoral immune responses that were enhanced by higher temozolomide doses.28 Of note, dendritic cells are relatively unaffected by temozolomide and thus may be able to exert more influence in the TMZ-depleted microenvironment.29

Immune Infiltrates

Macrophages are the most abundant cell type in gliomas, comprising a staggering 30%–50% of the cells in a tumor mass.16 It has been shown that a combination of tissue-resident microglia and bone marrow–derived macrophages both contribute to the tumor-associated macrophage (TAM) population. Microglia are embryologically yolk sac–derived and reside in normal brain tissue, where they account for 10%–20% of the non-neuronal population in the brain.30 They play an important role in brain development, homeostatic maintenance, angiogenesis, synaptic formation, and protection from pathogens.31,32 Extra-parenchymal, bone marrow–derived macrophages typically reside in the perivascular spaces and the meninges.33 Both microglia and macrophages infiltrate gliomas, and the density of infiltration correlates with glioma progression and grade, with more highly infiltrated lesions being more aggressive.14,15,34 Glioma cells secrete high levels of chemoattractants including CCL2, CCL7, MCP-3, CSF-1, G-CSF, Lox, and Osteopontin.35–38 These attractants stimulate robust infiltration of macrophages and microglia into the tumor where they are manipulated into a pro-tumoral role. In addition to attractants, tumor cells secrete an abundance of anti-inflammatory cytokines and metabolites, such as IL-6, IL-10, TGF-β, adenosine, glucocorticoids, and prostaglandins.39–44 The combination of chemoattractants and immunosuppressive signals results in a resident macrophage population that is unable to carry out their expected functions. Ideally, macrophages, along with other antigen-presenting cells (APCs), would function by engulfing tumor cell debris and presenting processed peptides on major histocompatibility complexes (MHC) I and II for T-cell surveillance. Macrophage activation, mediated by IFN-γ involves secretion of TNF-α, chemokines, cytokines, and nitric oxide to augment the pro-inflammatory environment.45 Macrophage activation to an inflammatory state (M1-like phenotype) is normally balanced in the body by subsequent transition to an anti-inflammatory (M2-like) state to promote healing.46 While the M1 vs M2 polarization model is an oversimplification of macrophage activation states in gliomas, markers of these phenotypes are useful in identifying cell states that are pro or anti-inflammatory. In the classically activated M1 state, macrophages produce inflammatory cytokines and are identified by cell surface markers CD80, CD86, and MHC II. In the alternatively activated M2 state, macrophages secrete factors to dampen the immune system and promote tissue healing and remodeling, including ECM deposition and angiogenesis. This M2 polarization is the predominant macrophage phenotype observed in gliomas and can be detected by expression of markers CD163, CD206, and arginase-1 (ARG-1).47,48 Alternative (M2) activation is triggered by the abovementioned anti-inflammatory factors (IL-4, IL-10, IL-13, CSF-1, TGF-β) and maintained autocrine signaling by macrophage secretion of IL-10 and expression of the IL-10 receptor.49–51 As expected, expression of M2 markers correlates with higher glioma grade.47 This is because these M2 macrophages in the TME do not recognize tumor cells as intruders, but rather, as healing tissue for which they carry out supportive roles of promoting proliferation, connectivity, and angiogenesis.

Dendritic cells (DCs) constitute about 5% of the cell population in IDH WT glioma and 10% of the population in IDH mutant glioma.52 Dendritic cells are antigen-processing and -presenting cells and interact with both the innate and adaptive immune system including T cells, B cells, mast cells, and NK cells. They are not found in the quiescent brain parenchyma but reside in vascular-rich compartments such as the meninges and choroid plexus. In gliomas, the infiltrative dendritic cell population consists of different subtypes, such as conventional DC type 1 (cDC1), conventional DC type 2 (cDC2), premature DCs (pre-DC), migratory DCs (CD2), and plasmacytoid DCs, though they display limited function compared to their peripheral blood counterparts.52,53 Suppressive components of the TME, such as macrophage-derived IL-10, limit full maturation and antigen-presenting capability.52,54 Recent work using single-cell RNA-seq analysis has identified additional DC subsets in blood and GBM tissue,53,55 though further investigation is needed to understand the role each of these subsets plays in the microenvironment.

MDSCs are a group of myeloid progenitor and precursor cells consisting of macrophages, dendritic cells, and granulocytes in different stages of differentiation. Under the chronic inflammatory conditions of the TME, immature myeloid cells do not fully differentiate into a mature myeloid cell type, causing accumulation of an immunosuppressive population of cells known as MDSCs.56 There are different subtypes of MDSCs based on which differentiation markers they express, namely granulocytic (G-MDSC), mononuclear (M-MDSC), and early-stage (E-MDSC), while all lacking mature markers of myeloid cells such as MHCII-HLA DR.57 The MDSC cell population is particularly immunosuppressive, with ability to block T cell, NK cell, macrophage, and dendritic cell function. They utilize reactive oxygen species and reactive nitrogen species, generated through NADPH oxidase or ARG-1 to induce oxidative stress that blocks T-cell or NK cell function. MDSCs also secrete soluble factors such as IL-10, TGF-β, and IFN-γ that are able to polarize macrophages to M2 phenotype, inhibit DC cell stimulation of T cells, and influence T reg differentiation.56

Tumor-infiltrating lymphocytes (TILs) are an essential component of the host immune response and the body’s most effective weapon against tumor growth. The number of infiltrating T cells in glioma tissue is associated with improved survival independent of age, MGMT status, or treatment.11 There have been observations that the number of CD8+ TILs in glioma is inversely correlated with tumor grade58 and with EGFR amplification and homozygous PTEN loss.59 The overall density of lymphocytes in glioma tissue is low, comprising only 1% of all cell types.60 In addition, the glioma microenvironment is particularly adept at inducing T-cell dysfunction through immunosuppressive cytokines, metabolites, immune checkpoints, antigen suppression, and regulatory T cells.4,5 Together, with low TIL density, these microenvironmental factors render T-cell responses ineffective.

Virtually every cell in the body displays internally processed peptide fragments on their cell surface in MHC I receptors for T-cell inspection. APCs that engulf pathogens, debris, or apoptotic cells present peptide fragments on MHC II receptors. When T cells interact with MHC I and II receptors that contain irregular sequences (ie, tumor mutations), this can trigger a cascade of T-cell activation and proliferation, releasing an army of CD4+ T helper cells and CD8+ cytotoxic T cells to scan the body for the aberrant sequence. When a T helper cell locates mutant peptides on a tumor cell, they secrete pro-inflammatory cytokines to recruit cytotoxic T cells and other immune cells to the target. Cytotoxic T cells are then able to kill tumor cells through secretion of perforin and granzymes to induce apoptosis. T regulatory cells (Tregs) serve to temper the inflammatory response once the threat has been neutralized. While this seems like a straightforward program to eliminate any cell that is going awry, tumor cells employ multiple evasive maneuvers to avoid detection. For example, tumor cells downregulate MHC I expression, so that antigens are not presented on the cell surface for T-cell detection. Even if functional APCs are able to present tumor peptides on MHC II for T-cell activation, the T cells are unable to locate the mutant tumor cells that lack MHC I. Loss of MHC I expression is correlated with worse clinical outcomes, and resistance to immune checkpoint blockade and adoptive immunotherapy.61

Tumors also utilize innate immune regulatory programs to quell antitumor immune responses. For example, T cells express surface inhibitory proteins, called checkpoints, such as programmed-death 1 (PD-1), T-cell immunoglobulin and mucin domain-containing protein-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and cytotoxic T lymphocyte Antigen 4 (CTLA-4). One of the best-known, and successfully modulated, mechanisms of immune evasion is upregulation of checkpoint ligands on tumor cells, such as the PD-1 ligand, PDL-1, which can bind to PD-1 on surveilling T cells and prevent activation even when a tumor antigen is detected.62 Chronic antigen exposure in the setting of anti-inflammatory signals, such as occurs in the glioma microenvironment, results in hyporesponsive T-cell states that share many similarities with the exhaustion observed in chronic viral infections. This hypofunction is marked by even further upregulation of multiple checkpoint receptors and a state of severe T-cell dysfunction.5 In addition to these active immunosuppressive processes, at baseline the TME is an uninviting place for T cells. Nutrient deficiency, hypoxia, high lactate levels, acidic pH, and high levels of released ions and metabolites such as potassium (K+) and adenosine all limit function of infiltrating T cells. A major line of tumor defense is inactivation of the APCs as described above, limiting antigen presentation and co-stimulation of T cells and secreting anti-inflammatory signals.

Stromal Cells

Gliomas are diffuse tumors that spread throughout the brain parenchyma. As such, the normal parenchymal cells become involved in the tumor mass and part of the TME. Like the immune cells we have discussed, stromal cells are not merely trapped in the TME but play an active role in tumor growth. Astrocytes are the most common glial cell in the brain and are best-known for their role in structural support, blood-brain barrier maintenance, synapse structural and functional maintenance, nutrient dispersion, and other homeostatic roles.63,64 Reactive astrocytes have been found in close association with glioma cells and have been found to promote tumor proliferation and invasion through secretion of stromal cell-derived factor 1 (SDF-1) and metalloproteinases.65 Tumor-associated astrocytes are predominately found in the tumor niche, along with fibroblasts, pericytes, and endothelial cells.66 This is also the primary location of the glioma stem-like cell (GSC) population.67 Similar to immune cells, reactive astrocytes are initially recruited to aid in brain healing, but quickly become hijacked to help with tumor progression. GBM cells produce RANKL, which acts on the astrocyte RANK receptor, to activate the NF-κB pathway. These activated astrocytes in turn secrete high levels of TGF-β, TNF-α, IGF-1, and GDF-15 which support glioma cell proliferation.68,69 Astrocytes have also been shown to facilitate glioma cell resistance to hypoxia through CCL20 secretion, and resistance to chemotherapy/apoptosis through GAP junctions and helping glioma cells clear calcium ions.70

Recent work demonstrated that neurons directly participate and support glioma development through aberrant synapses and secretion of growth stimuli. Gliomas integrate into neuronal circuitry where they form synapses with neurons and receive depolarizing currents which promote glioma growth and proliferation through voltage-sensitive signaling pathways.71 Glioma cells secrete the synopatogenic molecule thrombospondin-1 (TSP-1), promoting functional connectivity with neighboring neurons. Neurons also secrete synaptic molecule neuroligin-3 at neuronal-glioma synapses, which promotes glioma cell proliferation through the PI3K-MTOR pathway and results in feed-forward neuroligin-3 expression by glioma cells.72,73 Of note, these areas of high functional connectivity between neurons and glioma cells are inversely correlated with areas of immune activity.74 Animal models show that TSP-1 knockout not only abrogates neuronal connectivity but also restores pro-inflammatory responses, prolonging survival in immunocompetent but not immunodeficient models.74 This suggests that glioma cells utilize functional connectivity with neurons not only for neuron-derived growth signals but also to manipulate the microenvironment to evade immune cells by mimicking neuronal mechanisms.

Fibroblasts are additional stromal cells that play a role in tumor cell invasion and growth. These cells play roles in processing tumor-secreted metabolites and waste products to optimize tumor growth and immune evasion.75 Glioma-associated fibroblasts have also been shown to play an important role in matrix metalloprotease production for ECM restructuring.76,77 Recent work has also shown that cancer-associated fibroblasts produce extra domain A fibronectin which binds to macrophage TLR4 receptor to stimulate M2 polarization. Spatial transcriptomics also demonstrates close association between fibroblasts and GSCs in the perivascular niche. GSCs stimulate fibroblasts with PDGF and TGF-β and fibroblasts in turn secrete HGF and osteopontin.75

Endothelial cells play a critical role in tumor vascularization as well as carry an immunomodulatory role through cross-talk with immune cells.78 The proliferation, migration, and morphogenesis of endothelial cells is essential to angiogenesis, and angiogenesis is essential to glioma growth and progression.78,79 In fact, one of the defining histologic features of high-grade glioma is vascular proliferation. Vascular endothelial growth factor (VEGF) is a secreted signaling molecule that promotes angiogenesis. VEGF is secreted by tumor cells and TAMs to promote neovascularization. The main signals driving VEGF production are either cytokine cross-talk as described above, or hypoxia and HIF-1α signaling. Endothelial cells also play a role in promoting immunosuppression in the TME by upregulating PD-L1 expression on their cell surface in response to VEGF signaling and hypoxia.78

Extracellular Matrix

The ECM is a network of macromolecules such as chondroitin sulfate proteoglycans, heparan sulfate proteoglycans, hyaluronic acid, laminins, tenascins, and glycoprotein-fibronectin that provide the scaffold for the extracellular tissue microenvironment.80 The normal brain ECM differs from other tissues in that it lacks collagen (except in basal lamina of blood vessels) and carries out important roles in neuronal migration, differentiation, and maturation.81 The ECM of gliomas relative to normal brain has upregulated hyaluronic acid, tenascins, and fibronectin, with higher levels of these molecules being correlated with higher tumor grade and aggressiveness. Upregulation of these ECM components has been shown to increase the mobility and invasiveness of tumor cells. The ECM in gliomas is a dynamic structure that is actively remodeled through MMPs to facilitate tumor cell invasion, with new ECM deposition being a constant process.81

Demographic Factors

Age

There is a strong association between age at the time of glioma diagnosis and outcomes.82 One factor is the genetic composition of the tumor, as IDH mutant gliomas occur predominately in younger adults while IDH WT gliomas occur in a bimodal distribution in children and elderly adults.17 The incidence of IDH WT GBM progressively increases throughout adulthood and peaks in the eighth decade of life.82 Within the same glioma subtype classification, advanced age is associated with poorer outcomes in general and has been shown to have decreased survival benefit with immunotherapy treatment.83 General population studies show that circulating CD8+ T-cell numbers progressively decrease in peripheral circulation with age, while regulatory T-cell numbers increase.84 Advanced age is also associated with immunosuppressive gene expression profile in the brain including increased PD-L1 and IDO1 expression.84 While outcomes are certainly multifactorial, it is clear that the immune system is less capable of an antitumor response in the elderly population.

Gender

Men have higher incidence of glioma and worse survival outcomes compared to women.82,85,86 Investigations into molecular differences between gliomas in males and females have revealed differences in mutations and copy number alterations, with males having amplification of AKT1 on chromosome 17 and females having amplification of MYC on chromosome 8. Males also have enrichment of PIK3R1 and NF1 mutations while females are more likely to have PIK3CA mutations.86 Overall, females have a higher aneuploidy fraction and tumor mutational burden. Women also are more likely to have methylation of the MGMT promoter. Analysis of tumor gene expression patterns shows differences in transcriptional signatures with increased neuronal transcriptional signatures in males with IDH WT tumors compared to females.87 Aside from molecular differences in tumor cells, there are also broader TME differences that have been reported. Males have increased monocytic MDSCs in the TME.88 There are also intrinsic T-cell differences between sexes, with males having decreased T-cell infiltration into gliomas and increased rates of T-cell exhaustion associated with hormonal and sex gene differences.89

Metabolism and the TME

The brain TME is an area of high energy demand and insufficient vascular supply. One of the first noted characteristics of cancer cells is the increased rate of glycolysis, known as the Warburg Effect, discovered by Otto Warburg in 1927.90 The brain normally utilizes alternate energy sources like lactate and ketone bodies during periods of high energy demand. Tumor cells prioritize metabolic pathways that facilitate high rates of cell turnover and provide the energy and building components necessary for cell proliferation. Oncogenic pathways altered in cancer that regulate cell proliferation naturally also regulate cellular metabolism, increasing rates of glycolysis and secretion of lactic acid in the microenvironment.91 Glioma cells have been shown to upregulate the rate-limiting glycolytic enzyme hexokinase2.92,93 Lactic acid accumulation leads to an acidic microenvironment with a pH around 6.5 in glioma, compared to 7.2 in normal brain.94 Mass spectrometry imaging confirms elevated lactate levels in glioma tissue compared to surrounding tissue,95 and higher lactate levels in IDH WT vs IDH mutant gliomas.96 Serum lactate levels are also elevated in glioma patients, particularly with higher grade lesions.97 Lactic acid promotes angiogenesis and cancer cell migration and inhibits immune cell function.98 High lactate levels are associated with decreased CD8 T-cell infiltration in gliomas.99 When TAMs are exposed to lactate, HIF-1α is stabilized and VEGF production increases.100 In addition to glycolysis, gliomas also have upregulated activity through the pentose phosphate pathway, glutamine metabolism, and fatty acid oxidation. These all represent energy sources and pathways essential to generating biosynthesis components for cellular division that are necessary in GBM and provide the tumor with metabolic plasticity to deal with a dynamic nutrient microenvironment.101

Control of immune cell metabolism in the TME is not fully understood. Identifying targetable pathways to drive specific metabolic programs has the potential to provide the metabolic support necessary for immune cells to perform antitumor functions under duress. We recently identified a novel mechanism by which tumors inhibit immune cell function is through the metabolically active cytokine Meteorin-like (METRNL).102 T cells have limited resources in the TME and are susceptible to metabolic insufficiency.103 T cells that undergo repeated stimulation in this context enter a hypofunctional state characterized by upregulation of checkpoint molecules as well as secretion of METRNL. METRNL acts via autocrine and paracrine signaling to induce T-cell mitochondrial dysfunction and shift metabolic activity to glycolysis, temporarily conserving energy but decreasing T-cell function.102 Ablating METRNL improves T-cell function and supports tumor control. Future studies will determine if METRNL blockade or ablation alone is adequate metabolic support for endogenous and/or genetically modified antitumor T cells to remain active in the TME long-term.

Mutations in IDH1/2 alter the enzymatic active site and disrupt the normal metabolic function of converting isocitrate to α-ketoglutarate, and instead confer the neomorphic function of converting α-ketoglutarate to 2-hydroxyglutarate (2-HG). 2-HG cannot be utilized by the cell causing it to accumulate within the cell and within the TME. IDH mutation is believed to be one of the earliest mutations that occurs in low-grade glioma formation and has been shown to heavily impact tumor cell epigenetics and gene expression patterns. Investigations of the impact of 2-HG on the microenvironment have shown that 2-HG impairs T-cell chemotaxis, accumulation, and function through interference with STAT signaling pathways and decreased NFAT and NF-κB-mediated transcription.104–106 2-HG has also been shown to directly impair dendritic cell maturation, downregulating MHC II and IL-6 and upregulating immunosuppressive mediators CD206 and PD-L1, resulting in decreased T-cell activation and IFN-γ production.52

In 1975, it was first demonstrated that extracellular adenosine suppresses the activity of cytotoxic T cells.107 Subsequently, the effects of adenosine on the immune system have been further elucidated but translation to therapeutic strategies has lagged. There are 4 adenosine G protein-coupled receptors, A1, A2A, A2B, and A3, which are expressed to varying degrees on the surface of lymphocytes, macrophages, and other cells in the TME. Activation of T-cell receptors results in upregulation of A2A expression on T cells, and activation of toll-like receptors on macrophages upregulates expression of both A2A and A2B receptors.108 Adenosine stimulation of A2A on T cells results in intracellular cAMP production and downstream impairment of T-cell activation and proliferation, and causes upregulation of co-inhibitory molecules PD-1, TIM-3, LAG-3, and CTLA-4 as well as differentiation of CD4+ T cells into Tregs.40,109–111 Stimulation of A2A and A2B receptors on microglia and macrophages results in M2 polarization and increased expression of IL-10, VEGFA, and TGF-β.42,112 Gliomas manipulate this signaling mechanism in the TME through upregulation of adenosine levels. At baseline, adenosine levels are increased in the TME due to high cellular turnover. Following cell death, ATP is rapidly released into the extracellular space, shifting concentrations from the nanomolar to micromolar range.113 ATP is a pro-inflammatory molecule in the extracellular space; however, in the TME it is quickly hydrolyzed to adenosine, which is potently immunosuppressive. Tumor cells, and other cells in the TME, such as fibroblasts, express high levels of ectonucleotidases CD39 and CD73. CD39 is responsible for hydrolyzing extracellular ATP to ADP and AMP, and CD73 subsequently converts AMP to adenosine.114,115 Adenosine can then be catabolized by membrane-bound adenosine deaminase into inosine, or transported into cells.108

One of the most frequent genetic alterations in GBM is loss of the 9p21 locus including tumor suppressor genes CDKN2A, CDKN2B, and MTAP. MTAP deletion has been shown to have prognostic significance, correlating with shortened disease-free survival in GBM patients.116

Loss of 9p21 has been associated with an immunosuppressive microenvironment across several cancer types and resistance to immune checkpoint therapy.9 Further studies demonstrated that this immunosuppressive effect of 9p21 loss was specific to MTAP deletion and not CDKN2A or CDKN2B deletion.117 When MTAP enzyme function is lost, its substrate methylthioadenosine (MTA), a purine metabolite, is left to accumulate to high levels within and around the tumor cells, in a manner similar to 2-HG in IDH mutant gliomas. MTA inhibits several methyltransferase enzymes intracellularly, resulting in epigenetic changes to tumor cells,116,118,119 and has also been shown to have an impact on its microenvironment, likely by acting on purinergic receptors as well as affecting methyltransferase activity within other cells of the TME.6,117,120 Exposure of macrophages to MTA has been shown to downregulate TNF-α production and to shift macrophages to an M2-polarized phenotype with elevated expression of VEGFA and IL-10,116,121 and MTA has been shown to directly suppress the proliferation and function of T lymphocytes.122 As 9p21 loss is one of the cancer-initiating events in the process of tumorigenesis, it is likely that the resultant metabolic alteration of the TME is one of the first immune escape mechanisms employed by tumor cells and further cements the importance of purinergic signaling in immune modulation.

Therapeutic Targets in the TME

While the number and variety of immune evasion mechanisms in the TME are daunting, each offers a potential opportunity for therapeutic intervention (Figure 1). The most studied immunotherapeutic approach to date has been immune checkpoint blockade using antibodies directed toward surface inhibitory checkpoint proteins, such as anti-PD-1 or anti-CTLA-4. While this approach has been revolutionary in treating other cancer types, it has failed to show the same efficacy against GBM.123–125 Although there is some evidence that neoadjuvant administration has benefit,126,127 further work must be done to identify patient subsets that are most likely to benefit from checkpoint inhibitor treatment and trial novel combination therapies.128 A summary of phase III immunotherapy trials for glioma treatment is listed in Table 1.

Table 1.

Phase III Glioma Immunotherapy Trials (as of November 16, 2024 in clinicaltrials.gov)

Clinical Trial ID Study Name Start Date Status Agent Primary Outcome
NCT00045968 Study of a Drug [DCVax®-L] to Treat Newly Diagnosed GBM Brain Cancer (GBM) December 2006 Completed Autologous DC exposed to tumor antigen Increased OS in Newly Diagnosed GBMa
NCT03149003 A Study of DSP-7888 Dosing Emulsion in Combination with Bevacizumab in Patients With Recurrent or Progressive Glioblastoma Following Initial Therapy December 2017 Completed Ombipepimut (Synthetic epitopes of WT1) + bevacizumab No difference in OSa
NCT01480479 Phase III Study of Rindopepimut/GM-CSF in Patients With Newly Diagnosed Glioblastoma (ACT IV) November 2011 Completed Rindopepimut EGFRVIII peptide Vaccine No difference in OSa
NCT00088400 Comparison of TransMID (Transferrin-mediated delivery of diphtheria toxin) vs Standard Treatment of Cancerous Brain Tumors July 2004 Completed Tf-CRM107 Transferrin-Diphtheria toxin conjugate No difference in OSa
NCT01759810 Proteome-based Personalized Immunotherapy of Glioblastoma December 2012 Active Antigen-loaded DCs, co-cultured T cells, hematopoietic stem cells All-cause mortality
NCT04396860 Testing the Use of the Immunotherapy Drugs Ipilimumab and Nivolumab Plus Radiation Therapy Compared to the Usual Treatment (Temozolomide and Radiation Therapy) for Newly Diagnosed MGMT Unmethylated Glioblastoma September 2020 Active, Not Recruiting Ipilimumab Nivolumab OS
NCT04277221 Autologous Dendritic Cell/Tumor antigen for Adjuvant Immunotherapy in Standard Treatment of Recurrent Glioblastoma Multiforme (GBM) September 2019 Recruiting Antigen-loaded autologous DCs OS
NCT03548571 Dendritic Cell Immunotherapy Against Cancer Stem Cells in Glioblastoma Patients Receiving Standard Therapy April 2018 Recruiting DCs transfected w/ survivin, hTERT, and GBM stem cell mRNA PFS
NCT05235737 The Assessment of Immune Response in Newly Diagnosed Glioblastoma Patients Treated with Pembrolizumab (PIRG) March 2022 Recruiting Pembrolizumab OS
PFS
NCT06556563 EF-41/KEYNOTE D58: Phase 3 Study of Optune Concomitant With Temozolomide Plus Pembrolizumab in Newly Diagnosed Glioblastoma November 2024 Recruiting Optune + Pembrolizumab OS
NCT05685004 Study of Neoantigen-Specific Adoptive T cell Therapy for Newly Diagnosed MGMT Negative Glioblastoma Multiforme (GBM) September 2023 Recruiting Tumor antigen-stimulated autologous T cells OS
NCT05100641 AV-GBM-1 vs Control as Adjunctive Therapy Following Surgery and RT/TMZ in Newly Diagnosed GBM January 2024 Recruiting Antigen-loaded autologous DCs OS

Abbreviations: OS, overall survival; PFS, progression-free survival.

aFinal study result.

Another focus of investigation has been targeting the immunosuppressive effects of infiltrative macrophages.129 This includes treatment with IL-12 and inhibitors of CSF-1R to inhibit TAM function,130,131 inhibition of SDF-1 to inhibit chemotaxis,132 antibiotic treatment to inhibit microglial function,133 or promotion of M1 polarization through inhibition of STAT3 signaling.134

Altering the cellular composition of the TME through exogenous introduction of autologous immune cells through adoptive cell transfer has been another major effort. In these approaches immune cells (NK cells, dendritic cells, and T cells) are harvested and then modified through cytokine and antigen stimulation or genetic engineering and then reintroduced to the patient. A recent phase III trial using lysate-loaded autologous dendritic cell vaccine infusion showed significant survival improvement in both newly diagnosed and recurrent GBM (Table 1).135 Another highly investigated approach is the chimeric antigen receptor (CAR) T cell, which involves engineering the patients T cells to express a receptor that targets a tumor-specific antigen.136 This allows the T cell to bypass MHC-dependent antigen recognition. Recent work at 2 institutions demonstrates rapid radiographic responses in recurrent GBM after a single infusion of CAR T cells. In one study, the CAR was engineered to both target the epidermal growth factor receptor (EGFR) variant III tumor-specific antigen, as well as locally secrete t-cell-engaging antibody molecules against the WT EGFR protein.137 In the other study, the CAR was bivalent targeting an epitope of EGFR and IL13Rα2.138 While the results were mostly transient, these early results demonstrate the incredible potential of this line of therapy.

The metabolic influence of the TME on T cells is another recent focus in cancer treatment. Inhibition of adenosine production through ectonucleotidases (anti-CD39, anti-CD73) or direct inhibition of adenosine receptors A2A and A2B are being explored in clinical trials for various cancer types.139 Drugs that inhibit glycolysis/hexokinase 2,140,141 block glutamine synthesis and metabolism,142 or inhibit glucose and/or amino acid uptake143 are all approaches that are being investigated to normalize the metabolism of the TME in a way that can impair tumor cell growth and improve immune function. Similarly, targeting the effect of lactic acid buildup through inhibition of lactate dehydrogenase is gaining traction,139,144,145 as well as targeting hypoxia signaling pathways.146–148 While direct angiogenesis inhibition with anti-VEGF-A inhibitors has so far not shown to significantly improve survival in GBM patients, multi-modal antiangiogenic therapies and combination with other therapies are being investigated.149–151 Inhibitors of mutant IDH (Vorasidenib), which block the production of 2-HG, normalizing TCA cycle flow, have recently been shown to significantly improve progression-free survival in IDH mutant gliomas.152 With the discovery that functional connectivity between neurons and glioma cells promotes tumor growth, efforts to interfere with tumor-neuron synapses revealed that Gabapentin, a common antiepileptic and pain modulation drug that targets synaptogenic thrombospondin signaling, reduces glioma-neuron synapse formation and significantly reduces GBM growth.153

Targeting the ECM has been recognized as a high-yield area of therapeutic investigation. Modifying fibronectins, tenascins, fibulin-3, and hyaluronic acid deposition with blocking antibodies has been shown to decrease tumor growth, invasion, and neovascularization in preclinical models.154–157 Additionally, enzymatic breakdown of ECM components using hyaluronidase, matrix metalloproteinases, or chondroitinase has been shown to improve distribution of immunotherapeutic agents through the thick tumor ECM. Preclinical models utilizing oncolytic adenovirus armed with soluble hyaluronidase or oncolytic herpes simplex virus armed with matrix metalloproteinases or chondroitinase ABC each demonstrated improved viral distribution through the tumor and greater antitumor efficacy.158–160

Conclusions

The intricate and hostile glioma microenvironment presents a formidable challenge to the body’s natural immune functions and to therapeutic interventions. The TME is an active and dynamic participant in tumor progression and expands the challenge of treating glioma beyond merely targeting the tumor cells themselves. The interplay between glioma cells, immune infiltrates, stromal cells, and the ECM creates a complex ecosystem that fosters tumor growth, immune evasion, and therapeutic resistance.

Despite significant advances in our understanding of glioma biology, the immunosuppressive landscape of the TME remains a major barrier to effective treatment. Glioma cells exploit various mechanisms to suppress immune function, including the recruitment and reprogramming of TAMs, the expansion of MDSCs, and the inhibition of T-cell activity through immune checkpoints and metabolic reprogramming. Metabolic adaptations of glioma cells further render the microenvironment inhospitable to immune cell function. Additionally, the interactions between glioma cells and the surrounding stromal components, such as astrocytes and endothelial cells, promote a pro-tumoral environment that supports tumor cell survival and expansion.

To overcome these challenges, any therapeutic approach aimed at eradicating glioma must include strategies to modulate the TME, overcome immune cell dysfunction, and target the metabolic pathways that contribute to immunosuppression. While the complexity of the glioma microenvironment poses significant hurdles, it also offers numerous therapeutic targets. Continued research into the cellular and molecular mechanisms that drive immunosuppression and tumor progression in gliomas is crucial for the development of novel, more effective treatments. By unraveling the complexities of the glioma microenvironment, we can lay the foundation for innovative therapeutic strategies that may one day offer hope to patients with this devastating disease.

Contributor Information

Landon J Hansen, Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

Christopher M Jackson, Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.

Supplement sponsorship

This article appears as part of the supplement “Immunotherapy for Brain Tumors,” sponsored by the Wilkins Family Chair in Neurosurgical Brain Tumor Research.

Conflict of interest statement

C.M.J. is a consultant and shareholder of Egret Therapeutics. C.M.J. holds Amgen stock. C.M.J. has received research support from Biohaven, InCephalo, and Grifols. Johns Hopkins University has filed a provisional patent on METRNL blockade for cancer treatment on which he is an inventor. Johns Hopkins University has filed a provisional patent on PD-1 agonists for brain ischemia on which he is an inventor. Stanford University has filed a provisional on using PD-1 agonists for myocardial infarction on which he is an inventor.

References

  • 1. Ostrom QT, Price M, Neff C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2016-2020. Neuro Oncol. 2023;25(12 Suppl 2):iv1–iv99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Stupp R, Mason WP, van den Bent MJ, et al. ; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. [DOI] [PubMed] [Google Scholar]
  • 3. Beatty GL, Gladney WL.. Immune escape mechanisms as a guide for cancer immunotherapy. Clin Cancer Res. 2015;21(4):687–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Chongsathidkiet P, Jackson C, Koyama S, et al. Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nat Med. 2018;24(9):1459–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Woroniecka K, Chongsathidkiet P, Rhodin K, et al. T-cell exhaustion signatures vary with tumor type and are severe in glioblastoma. Clin Cancer Res. 2018;24(17):4175–4186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Hansen LJ, Yang R, Roso K, et al. MTAP loss correlates with an immunosuppressive profile in GBM and its substrate MTA stimulates alternative macrophage polarization. Sci Rep. 2022;12(1):4183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Nduom EK, Weller M, Heimberger AB.. Immunosuppressive mechanisms in glioblastoma. Neuro Oncol. 2015;17(Suppl 7):vii9–vii14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Coy S, Wang S, Stopka SA, et al. Single-cell spatial analysis reveals the topology of immunomodulatory purinergic signaling in glioblastoma. Nat Commun. 2022;13(1):4814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Han G, Yang G, Hao D, et al. 9p21 loss confers a cold tumor immune microenvironment and primary resistance to immune checkpoint therapy. Nat Commun. 2021;12(1):5606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. El Andaloussi A, Lesniak MS.. An increase in CD4+CD25+FOXP3+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro Oncol. 2006;8(3):234–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Kmiecik J, Poli A, Brons NH, et al. Elevated CD3+ and CD8+ tumor-infiltrating immune cells correlate with prolonged survival in glioblastoma patients despite integrated immunosuppressive mechanisms in the tumor microenvironment and at the systemic level. J Neuroimmunol. 2013;264(1–2):71–83. [DOI] [PubMed] [Google Scholar]
  • 12. Parney IF, Waldron JS, Parsa AT.. Flow cytometry and in vitro analysis of human glioma-associated macrophages. Laboratory investigation . J Neurosurg. 2009;110(3):572–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Hussain SF, Yang D, Suki D, Grimm E, Heimberger AB.. Innate immune functions of microglia isolated from human glioma patients. J Transl Med. 2006;4:15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Pinton L, Masetto E, Vettore M, et al. The immune-suppressive microenvironment of human gliomas depends on the accumulation of bone marrow-derived macrophages in the center of the lesion. J ImmunoTher Cancer. 2019;7(1):58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Li W, Graeber MB.. The molecular profile of microglia under the influence of glioma. Neuro Oncol. 2012;14(8):958–978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Charles NA, Holland EC, Gilbertson R, Glass R, Kettenmann H.. The brain tumor microenvironment . Glia. 2012;60(3):502–514. [DOI] [PubMed] [Google Scholar]
  • 17. Louis DN, Perry A, Wesseling P, et al. The 2021 WHO classification of tumors of the central nervous system: a summary. Neuro Oncol. 2021;23(8):1231–1251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Killela PJ, Pirozzi CJ, Healy P, et al. Mutations in IDH1, IDH2, and in the TERT promoter define clinically distinct subgroups of adult malignant gliomas. Oncotarget. 2014;5(6):1515–1525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Killela PJ, Reitman ZJ, Jiao Y, et al. TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal. Proc Natl Acad Sci U S A. 2013;110(15):6021–6026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Brennan CW, Verhaak RG, McKenna A, et al. ; TCGA Research Network. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Merchant M, Ranjan A, Pang Y, et al. Tumor mutational burden and immunotherapy in gliomas. Trends Cancer. 2021;7(12):1054–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Hodges TR, Ott M, Xiu J, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro Oncol. 2017;19(8):1047–1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Wang L, Ge J, Lan Y, et al. Tumor mutational burden is associated with poor outcomes in diffuse glioma. BMC Cancer. 2020;20(1):213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Gromeier M, Brown MC, Zhang G, et al. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy. Nat Commun. 2021;12(1):352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Hegi ME, Diserens AC, Godard S, et al. Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin Cancer Res. 2004;10(6):1871–1874. [DOI] [PubMed] [Google Scholar]
  • 26. Brock CS, Newlands ES, Wedge SR, et al. Phase I trial of temozolomide using an extended continuous oral schedule. Cancer Res. 1998;58(19):4363–4367. [PubMed] [Google Scholar]
  • 27. Fadul CE, Fisher JL, Gui J, et al. Immune modulation effects of concomitant temozolomide and radiation therapy on peripheral blood mononuclear cells in patients with glioblastoma multiforme. Neuro Oncol. 2011;13(4):393–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Sampson JH, Aldape KD, Archer GE, et al. Greater chemotherapy-induced lymphopenia enhances tumor-specific immune responses that eliminate EGFRvIII-expressing tumor cells in patients with glioblastoma. Neuro Oncol. 2011;13(3):324–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Karachi A, Dastmalchi F, Mitchell DA, Rahman M.. Temozolomide for immunomodulation in the treatment of glioblastoma. Neuro Oncol. 2018;20(12):1566–1572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Ginhoux F, Prinz M.. Origin of microglia: current concepts and past controversies. Cold Spring Harb Perspect Biol. 2015;7(8):a020537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Harry GJ. Microglia during development and aging. Pharmacol Ther. 2013;139(3):313–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Fetler L, Amigorena SN.. Brain under surveillance: the microglia patrol. Science. 2005;309(5733):392–393. [DOI] [PubMed] [Google Scholar]
  • 33. Faraco G, Park L, Anrather J, Iadecola C.. Brain perivascular macrophages: characterization and functional roles in health and disease. J Mol Med (Berl). 2017;95(11):1143–1152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Liu H, Sun Y, Zhang Q, et al. Pro-inflammatory and proliferative microglia drive progression of glioblastoma. Cell Rep. 2021;36(11):109718. [DOI] [PubMed] [Google Scholar]
  • 35. Chen P, Zhao D, Li J, et al. Symbiotic macrophage-glioma cell interactions reveal synthetic lethality in PTEN-null glioma. Cancer Cell. 2019;35(6):868–884.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Wei J, Marisetty A, Schrand B, et al. Osteopontin mediates glioblastoma-associated macrophage infiltration and is a potential therapeutic target. J Clin Invest. 2019;129(1):137–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Platten M, Kretz A, Naumann U, et al. Monocyte chemoattractant protein-1 increases microglial infiltration and aggressiveness of gliomas. Ann Neurol. 2003;54(3):388–392. [DOI] [PubMed] [Google Scholar]
  • 38. Coniglio SJ, Eugenin E, Dobrenis K, et al. Microglial stimulation of glioblastoma invasion involves epidermal growth factor receptor (EGFR) and colony-stimulating factor 1 receptor (CSF-1R) signaling. Mol Med. 2012;18(1):519–527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Wei J, Barr J, Kong LY, et al. Glioma-associated cancer-initiating cells induce immunosuppression. Clin Cancer Res. 2010;16(2):461–473. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 40. Ohta A. A metabolic immune checkpoint: adenosine in tumor microenvironment. Front Immunol. 2016;7:109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Zou JP, Morford LA, Chougnet C, et al. Human glioma-induced immunosuppression involves soluble factor(s) that alters monocyte cytokine profile and surface markers. J Immunol. 1999;162(8):4882–4892. [PubMed] [Google Scholar]
  • 42. Csóka B, Selmeczy Z, Koscsó B, et al. Adenosine promotes alternative macrophage activation via A2A and A2B receptors. FASEB J. 2012;26(1):376–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Haskó G, Pacher P.. Regulation of macrophage function by adenosine. Arterioscler Thromb Vasc Biol. 2012;32(4):865–869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Ghiringhelli F, Bruchard M, Chalmin F, Rébé C.. Production of adenosine by ectonucleotidases: a key factor in tumor immunoescape. J Biomed Biotechnol. 2012;2012:473712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Roesch S, Rapp C, Dettling S, Herold-Mende C.. When immune cells turn bad-tumor-associated microglia/macrophages in glioma. Int J Mol Sci . 2018;19(2):436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Mantovani A, Sica A, Sozzani S, et al. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004;25(12):677–686. [DOI] [PubMed] [Google Scholar]
  • 47. Prosniak M, Harshyne LA, Andrews DW, et al. Glioma grade is associated with the accumulation and activity of cells bearing M2 monocyte markers. Clin Cancer Res. 2013;19(14):3776–3786. [DOI] [PubMed] [Google Scholar]
  • 48. Karimi E, Yu MW, Maritan SM, et al. Single-cell spatial immune landscapes of primary and metastatic brain tumours. Nature. 2023;614(7948):555–563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Sawada M, Suzumura A, Hosoya H, Marunouchi T, Nagatsu T.. Interleukin-10 inhibits both production of cytokines and expression of cytokine receptors in microglia. J Neurochem. 1999;72(4):1466–1471. [DOI] [PubMed] [Google Scholar]
  • 50. Tang Y, Le W.. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2016;53(2):1181–1194. [DOI] [PubMed] [Google Scholar]
  • 51. Zhang H, Luo YB, Wu W, et al. The molecular feature of macrophages in tumor immune microenvironment of glioma patients. Comput Struct Biotechnol J. 2021;19:4603–4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Friedrich M, Hahn M, Michel J, et al. Dysfunctional dendritic cells limit antigen-specific T-cell response in glioma. Neuro Oncol. 2023;25(2):263–276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Pombo Antunes AR, Scheyltjens I, Lodi F, et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat Neurosci. 2021;24(4):595–610. [DOI] [PubMed] [Google Scholar]
  • 54. Ruffell B, Chang-Strachan D, Chan V, et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 2014;26(5):623–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Villani AC, Satija R, Reynolds G, et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017;356(6335):eaah4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Mi Y, Guo N, Luan J, et al. The emerging role of myeloid-derived suppressor cells in the glioma immune suppressive microenvironment. Front Immunol. 2020;11:737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Gieryng A, Kaminska B.. Myeloid-derived suppressor cells in gliomas. Contemp Oncol (Pozn). 2016;20(5):345–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Han S, Zhang C, Li Q, et al. Tumour-infiltrating CD4(+) and CD8(+) lymphocytes as predictors of clinical outcome in glioma. Br J Cancer. 2014;110(10):2560–2568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Rutledge WC, Kong J, Gao J, et al. Tumor-infiltrating lymphocytes in glioblastoma are associated with specific genomic alterations and related to transcriptional class. Clin Cancer Res. 2013;19(18):4951–4960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. González-Tablas Pimenta M, Otero A, Arandia Guzman DA, et al. Tumor cell and immune cell profiles in primary human glioblastoma: impact on patient outcome. Brain Pathol. 2021;31(2):365–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Dhatchinamoorthy K, Colbert JD, Rock KL.. Cancer immune evasion through loss of MHC Class I antigen presentation. Front Immunol. 2021;12:636568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A. 2002;99(19):12293–12297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Kim JH, Park JA, Lee SW, et al. Blood-neural barrier: intercellular communication at glio-vascular interface. J Biochem Mol Biol. 2006;39(4):339–345. [DOI] [PubMed] [Google Scholar]
  • 64. Chung WS, Allen NJ, Eroglu C.. Astrocytes control synapse formation, function, and elimination. Cold Spring Harb Perspect Biol. 2015;7(9):a020370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Barbero S, Bajetto A, Bonavia R, et al. Expression of the chemokine receptor CXCR4 and its ligand stromal cell-derived factor 1 in human brain tumors and their involvement in glial proliferation in vitro. Ann N Y Acad Sci. 2002;973:60–69. [DOI] [PubMed] [Google Scholar]
  • 66. Zhao X, Chen R, Liu M, et al. Remodeling the blood-brain barrier microenvironment by natural products for brain tumor therapy. Acta Pharm Sin B. 2017;7(5):541–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Chen J, Mao S, Li H, et al. The pathological structure of the perivascular niche in different microvascular patterns of glioblastoma. PLoS One. 2017;12(8):e0182183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Roth P, Junker M, Tritschler I, et al. GDF-15 contributes to proliferation and immune escape of malignant gliomas. Clin Cancer Res. 2010;16(15):3851–3859. [DOI] [PubMed] [Google Scholar]
  • 69. Placone AL, Quiñones-Hinojosa A, Searson PC.. The role of astrocytes in the progression of brain cancer: complicating the picture of the tumor microenvironment. Tumour Biol. 2016;37(1):61–69. [DOI] [PubMed] [Google Scholar]
  • 70. Chen W, Wang D, Du X, et al. Glioma cells escaped from cytotoxicity of temozolomide and vincristine by communicating with human astrocytes. Med Oncol. 2015;32(3):43. [DOI] [PubMed] [Google Scholar]
  • 71. Venkatesh HS, Morishita W, Geraghty AC, et al. Electrical and synaptic integration of glioma into neural circuits. Nature. 2019;573(7775):539–545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Venkatesh HS, Tam LT, Woo PJ, et al. Targeting neuronal activity-regulated neuroligin-3 dependency in high-grade glioma. Nature. 2017;549(7673):533–537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Venkatesh HS, Johung TB, Caretti V, et al. Neuronal activity promotes glioma growth through neuroligin-3 secretion. Cell. 2015;161(4):803–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Nejo T, Krishna S, Jimenez C, et al. Glioma-neuronal circuit remodeling induces regional immunosuppression. bioRxiv. 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Jain S, Rick JW, Joshi RS, et al. Single-cell RNA sequencing and spatial transcriptomics reveal cancer-associated fibroblasts in glioblastoma with protumoral effects. J Clin Invest. 2023;133(5):147087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Sameshima T, Nabeshima K, Toole BP, et al. Glioma cell extracellular matrix metalloproteinase inducer (EMMPRIN) (CD147) stimulates production of membrane-type matrix metalloproteinases and activated gelatinase A in co-cultures with brain-derived fibroblasts. Cancer Lett. 2000;157(2):177–184. [DOI] [PubMed] [Google Scholar]
  • 77. Valera PS, Plou J, García I, et al. SERS analysis of cancer cell-secreted purines reveals a unique paracrine crosstalk in MTAP-deficient tumors. Proc Natl Acad Sci U S A. 2023;120(52):e2311674120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Fang J, Lu Y, Zheng J, et al. Exploring the crosstalk between endothelial cells, immune cells, and immune checkpoints in the tumor microenvironment: new insights and therapeutic implications. Cell Death Dis. 2023;14(9):586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Yang C, Passos Gibson V, Hardy P.. The role of MiR-181 family members in endothelial cell dysfunction and tumor angiogenesis. Cells. 2022;11(10):1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Lau LW, Cua R, Keough MB, Haylock-Jacobs S, Yong VW.. Pathophysiology of the brain extracellular matrix: a new target for remyelination. Nat Rev Neurosci. 2013;14(10):722–729. [DOI] [PubMed] [Google Scholar]
  • 81. Mohiuddin E, Wakimoto H.. Extracellular matrix in glioblastoma: opportunities for emerging therapeutic approaches. Am J Cancer Res. 2021;11(8):3742–3754. [PMC free article] [PubMed] [Google Scholar]
  • 82. Wang GM, Cioffi G, Patil N, et al. Importance of the intersection of age and sex to understand variation in incidence and survival for primary malignant gliomas. Neuro Oncol. 2022;24(2):302–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Ladomersky E, Zhai L, Lauing KL, et al. Advanced age increases immunosuppression in the brain and decreases immunotherapeutic efficacy in subjects with glioblastoma. Clin Cancer Res. 2020;26(19):5232–5245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Ladomersky E, Scholtens DM, Kocherginsky M, et al. The coincidence between increasing age, immunosuppression, and the incidence of patients with glioblastoma. Front Pharmacol. 2019;10:200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Miller KD, Ostrom QT, Kruchko C, et al. Brain and other central nervous system tumor statistics, 2021. CA Cancer J Clin. 2021;71(5):381–406. [DOI] [PubMed] [Google Scholar]
  • 86. Jang B, Yoon D, Lee JY, et al. Integrative multi-omics characterization reveals sex differences in glioblastoma. Biol Sex Differ. 2024;15(1):23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Huang Y, Shan Y, Zhang W, et al. Sex differences in the molecular profile of adult diffuse glioma are shaped by IDH status and tumor microenvironment. Neuro Oncol. 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Bayik D, Zhou Y, Park C, et al. Myeloid-derived suppressor cell subsets drive glioblastoma growth in a sex-specific manner. Cancer Discov. 2020;10(8):1210–1225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Lee J, Nicosia M, Hong ES, et al. Sex-biased T-cell exhaustion drives differential immune responses in glioblastoma. Cancer Discov 2023;13(9):2090–2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Warburg O, Wind F, Negelein E.. The metabolism of tumors in the body. J Gen Physiol. 1927;8(6):519–530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Cohen IJ, Blasberg R.. Impact of the tumor microenvironment on tumor-infiltrating lymphocytes: focus on breast cancer. Breast Cancer (Auckl). 2017;11:1178223417731565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Stanke KM, Wilson C, Kidambi S.. High expression of glycolytic genes in clinical glioblastoma patients correlates with lower survival. Front Mol Biosci. 2021;8:752404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Guo D, Tong Y, Jiang X, et al. Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IκBα. Cell Metab. 2022;34(9):1312–1324.e6. [DOI] [PubMed] [Google Scholar]
  • 94. Walsh JJ, Parent M, Akif A, et al. Imaging hallmarks of the tumor microenvironment in glioblastoma progression. Front Oncol. 2021;11:692650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Kampa JM, Kellner U, Marsching C, et al. Glioblastoma multiforme: metabolic differences to peritumoral tissue and IDH-mutated gliomas revealed by mass spectrometry imaging. Neuropathol. 2020;40(6):546–558. [DOI] [PubMed] [Google Scholar]
  • 96. Wenger KJ, Steinbach JP, Bähr O, Pilatus U, Hattingen E.. Lower lactate levels and lower intracellular pH in patients with. AJNR Am J Neuroradiol. 2020;41(8):1414–1422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Branco M, Linhares P, Carvalho B, et al. Serum lactate levels are associated with glioma malignancy grade. Clin Neurol Neurosurg. 2019;186:105546. [DOI] [PubMed] [Google Scholar]
  • 98. Walenta S, Mueller-Klieser WF.. Lactate: mirror and motor of tumor malignancy. Semin Radiat Oncol. 2004;14(3):267–274. [DOI] [PubMed] [Google Scholar]
  • 99. Wang Z, Dai Z, Zhang H, et al. Tumor-secreted lactate contributes to an immunosuppressive microenvironment and affects CD8 T-cell infiltration in glioblastoma. Front Immunol. 2023;14:894853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Husain Z, Huang Y, Seth P, Sukhatme VP.. Tumor-derived lactate modifies antitumor immune response: effect on myeloid-derived suppressor cells and NK cells. J Immunol. 2013;191(3):1486–1495. [DOI] [PubMed] [Google Scholar]
  • 101. Kant S, Kesarwani P, Prabhu A, et al. Enhanced fatty acid oxidation provides glioblastoma cells metabolic plasticity to accommodate to its dynamic nutrient microenvironment. Cell Death Dis. 2020;11(4):253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Jackson CM, Pant A, Dinalankara W, et al. The cytokine meteorin-like inhibits anti-tumor CD8. Immunity. 2024;57(8):1864–1877.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Reina-Campos M, Scharping NE, Goldrath AW.. CD8. Nat Rev Immunol. 2021;21(11):718–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Kohanbash G, Carrera DA, Shrivastav S, et al. Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. J Clin Invest. 2017;127(4):1425–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Amankulor NM, Kim Y, Arora S, et al. Mutant IDH1 regulates the tumor-associated immune system in gliomas. Genes Dev. 2017;31(8):774–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Bunse L, Pusch S, Bunse T, et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat Med. 2018;24(8):1192–1203. [DOI] [PubMed] [Google Scholar]
  • 107. Wolberg G, Zimmerman TP, Hiemstra K, Winston M, Chu LC.. Adenosine inhibition of lymphocyte-mediated cytolysis: possible role of cyclic adenosine monophosphate. Science. 1975;187(4180):957–959. [DOI] [PubMed] [Google Scholar]
  • 108. Allard B, Allard D, Buisseret L, Stagg J.. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol. 2020;17(10):611–629. [DOI] [PubMed] [Google Scholar]
  • 109. Leone RD, Sun IM, Oh MH, et al. Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother. 2018;67(8):1271–1284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Ohta A, Kini R, Subramanian M, Madasu M, Sitkovsky M.. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front Immunol. 2012;3:190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Ohta A, Gorelik E, Prasad SJ, et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci U S A. 2006;103(35):13132–13137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Koscsó B, Csóka B, Selmeczy Z, et al. Adenosine augments IL-10 production by microglial cells through an A2B adenosine receptor-mediated process. J Immunol. 2012;188(1):445–453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E.. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer. 2018;18(10):601–618. [DOI] [PubMed] [Google Scholar]
  • 114. Kaczmarek E, Koziak K, Sévigny J, et al. Identification and characterization of CD39/vascular ATP diphosphohydrolase. J Biol Chem. 1996;271(51):33116–33122. [DOI] [PubMed] [Google Scholar]
  • 115. Yegutkin GG, Henttinen T, Samburski SS, Spychala J, Jalkanen S.. The evidence for two opposite, ATP-generating and ATP-consuming, extracellular pathways on endothelial and lymphoid cells. Biochem J. 2002;367(Pt 1):121–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Hansen LJ, Sun R, Yang R, et al. MTAP loss promotes stemness in glioblastoma and confers unique susceptibility to purine starvation. Cancer Res. 2019;79(13):3383–3394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Gjuka D, Adib E, Garrison K, et al. Enzyme-mediated depletion of methylthioadenosine restores T cell function in MTAP-deficient tumors and reverses immunotherapy resistance. Cancer Cell. 2023;41(10):1774–1787.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Kryukov GV, Wilson FH, Ruth JR, et al. MTAP deletion confers enhanced dependency on the PRMT5 arginine methyltransferase in cancer cells. Science. 2016;351(6278):1214–1218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Mavrakis KJ, McDonald ER, Schlabach MR, et al. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science. 2016;351(6278):1208–1213. [DOI] [PubMed] [Google Scholar]
  • 120. Limm K, Wallner S, Milenkovic VM, Wetzel CH, Bosserhoff AK.. The metabolite 5’-methylthioadenosine signals through the adenosine receptor A2B in melanoma. Eur J Cancer. 2014;50(15):2714–2724. [DOI] [PubMed] [Google Scholar]
  • 121. Keyel PA, Romero M, Wu W, et al. Methylthioadenosine reprograms macrophage activation through adenosine receptor stimulation. PLoS One. 2014;9(8):e104210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Henrich FC, Singer K, Poller K, et al. Suppressive effects of tumor cell-derived 5’-deoxy-5’-methylthioadenosine on human T cells. Oncoimmunol. 2016;5(8):e1184802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Lim M, Weller M, Idbaih A, et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro Oncol. 2022;24(11):1935–1949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Omuro A, Reardon DA, Sampson JH, et al. Nivolumab plus radiotherapy with or without temozolomide in newly diagnosed glioblastoma: results from exploratory phase I cohorts of CheckMate 143. Neurooncol. Adv. 2022;4(1):vdac025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Reardon DA, Brandes AA, Omuro A, et al. Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: the CheckMate 143 phase 3 randomized clinical trial. JAMA Oncol. 2020;6(7):1003–1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Schalper KA, Rodriguez-Ruiz ME, Diez-Valle R, et al. Neoadjuvant nivolumab modifies the tumor immune microenvironment in resectable glioblastoma. Nat Med. 2019;25(3):470–476. [DOI] [PubMed] [Google Scholar]
  • 127. Cloughesy TF, Mochizuki AY, Orpilla JR, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25(3):477–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Arrieta VA, Dmello C, McGrail DJ, et al. Immune checkpoint blockade in glioblastoma: from tumor heterogeneity to personalized treatment. J Clin Invest. 2023;133(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Lin C, Wang N, Xu C.. Glioma-associated microglia/macrophages (GAMs) in glioblastoma: immune function in the tumor microenvironment and implications for immunotherapy. Front Immunol. 2023;14:1123853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Chiocca EA, Gelb AB, Chen CC, et al. Combined immunotherapy with controlled interleukin-12 gene therapy and immune checkpoint blockade in recurrent glioblastoma: an open-label, multi-institutional phase I trial. Neuro Oncol. 2022;24(6):951–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Butowski N, Colman H, De Groot JF, et al. Orally administered colony-stimulating factor 1 receptor inhibitor PLX3397 in recurrent glioblastoma: an ivy foundation early phase clinical trials consortium phase II study. Neuro Oncol. 2016;18(4):557–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Lee EQ, Duda DG, Muzikansky A, et al. Phase I and biomarker study of plerixafor and bevacizumab in recurrent high-grade glioma. Clin Cancer Res. 2018;24(19):4643–4649. [DOI] [PubMed] [Google Scholar]
  • 133. Cohen AL, Anker CJ, Johnson B, et al. Repeat radiation with bevacizumab and minocycline in bevacizumab-refractory high-grade gliomas: a prospective phase 1 trial. J Neurooncol. 2020;148(3):577–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Groot J, Ott M, Wei J, et al. A first-in-human phase I trial of the oral p-STAT3 inhibitor WP1066 in patients with recurrent malignant glioma. CNS Oncol. 2022;11(2):CNS87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Liau LM, Ashkan K, Brem S, et al. Association of autologous tumor lysate-loaded dendritic cell vaccination with extension of survival among patients with newly diagnosed and recurrent glioblastoma: a phase 3 prospective externally controlled cohort trial. JAMA Oncol. 2023;9(1):112–121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Luksik AS, Yazigi E, Shah P, Jackson CM.. CAR T cell therapy in glioblastoma: overcoming challenges related to antigen expression. Cancers (Basel). 2023;15(5):1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Choi BD, Gerstner ER, Frigault MJ, et al. Intraventricular CARv3-TEAM-E T cells in recurrent glioblastoma. N Engl J Med. 2024;390(14):1290–1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Bagley SJ, Logun M, Fraietta JA, et al. Intrathecal bivalent CAR T cells targeting EGFR and IL13Rα2 in recurrent glioblastoma: phase 1 trial interim results. Nat Med. 2024;30(5):1320–1329. [DOI] [PubMed] [Google Scholar]
  • 139. Verma NK, Wong BHS, Poh ZS, et al. Obstacles for T-lymphocytes in the tumour microenvironment: therapeutic challenges, advances and opportunities beyond immune checkpoint. EBioMed. 2022;83:104216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Chiasserini D, Davidescu M, Orvietani PL, et al. 3-Bromopyruvate treatment induces alterations of metabolic and stress-related pathways in glioblastoma cells. J Proteomics. 2017;152:329–338. [DOI] [PubMed] [Google Scholar]
  • 141. Guo J, Xue Q, Liu K, et al. Dimethylaminomicheliolide (DMAMCL) suppresses the proliferation of glioblastoma cells via targeting pyruvate kinase 2 (PKM2) and rewiring aerobic glycolysis. Front Oncol. 2019;9:993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Zhao J, Ma X, Gao P, et al. Advancing glioblastoma treatment by targeting metabolism. Neoplasia. 2024;51:100985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Pająk B, Siwiak-Niedbalska E, Jaśkiewicz A, Sołtyka M, Domoradzki T.. WP1234-A novel anticancer agent with bifunctional activity in a glioblastoma model. Biomedicines. 2022;10(11):2799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Khan F, Lin Y, Ali H, et al. Lactate dehydrogenase A regulates tumor-macrophage symbiosis to promote glioblastoma progression. Nat Commun. 2024;15(1):1987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Guyon J, Fernandez-Moncada I, Larrieu CM, et al. Lactate dehydrogenases promote glioblastoma growth and invasion via a metabolic symbiosis. EMBO Mol Med. 2022;14(12):e15343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kao TW, Bai GH, Wang TL, et al. Novel cancer treatment paradigm targeting hypoxia-induced factor in conjunction with current therapies to overcome resistance. J Exp Clin Cancer Res. 2023;42(1):171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Domènech M, Hernández A, Plaja A, Martínez-Balibrea E, Balañà CH.. The cornerstone of glioblastoma. Int J Mol Sci . Nov 222021;22(22). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Wang G, Wang JJ, Fu XL, Guang R, To ST.. Advances in the targeting of HIF-1α and future therapeutic strategies for glioblastoma multiforme (review). Oncol Rep. 2017;37(2):657–670. [DOI] [PubMed] [Google Scholar]
  • 149. Zhang AB, Mozaffari K, Aguirre B, et al. Exploring the past, present, and future of anti-angiogenic therapy in glioblastoma. Cancers (Basel). Jan 292023;15(3):830. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Ameratunga M, Pavlakis N, Wheeler H, et al. Anti-angiogenic therapy for high-grade glioma. Cochrane Database Syst Rev. 2018;11(11):CD008218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Shamshiripour P, Hajiahmadi F, Lotfi S, et al. Next-generation anti-angiogenic therapies as a future prospect for glioma immunotherapy; from bench to bedside. Front Immunol. 2022;13:859633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Mellinghoff IK, van den Bent MJ, Blumenthal DT, et al. ; INDIGO Trial Investigators. Vorasidenib in IDH1- or IDH2-mutant low-grade glioma. N Engl J Med. 2023;389(7):589–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Krishna S, Choudhury A, Keough MB, et al. Glioblastoma remodelling of human neural circuits decreases survival. Nature. 2023;617(7961):599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Sabari J, Lax D, Connors D, et al. Fibronectin matrix assembly suppresses dispersal of glioblastoma cells. PLoS One. 2011;6(9):e24810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Xia S, Lal B, Tung B, et al. Tumor microenvironment tenascin-C promotes glioblastoma invasion and negatively regulates tumor proliferation. Neuro Oncol. 2016;18(4):507–517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Nandhu MS, Behera P, Bhaskaran V, et al. Development of a function-blocking antibody against Fibulin-3 as a targeted reagent for glioblastoma. Clin Cancer Res. 2018;24(4):821–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Wolf KJ, Shukla P, Springer K, et al. A mode of cell adhesion and migration facilitated by CD44-dependent microtentacles. Proc Natl Acad Sci U S A. 2020;117(21):11432–11443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Martinez-Quintanilla J, He D, Wakimoto H, Alemany R, Shah K.. Encapsulated stem cells loaded with hyaluronidase-expressing oncolytic virus for brain tumor therapy. Mol Ther. 2015;23(1):108–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Sette P, Amankulor N, Li A, et al. GBM-targeted oHSV armed with matrix metalloproteinase 9 enhances anti-tumor activity and animal survival. Mol Ther Oncolytics. 2019;15:214–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Jaime-Ramirez AC, Dmitrieva N, Yoo JY, et al. Humanized chondroitinase ABC sensitizes glioblastoma cells to temozolomide. J Gene Med. 2017;19(3). [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Neuro-Oncology Advances are provided here courtesy of Oxford University Press

RESOURCES