Abstract
Glioma, a common malignancy of the central nervous system, attracts significant clinical attention due to its poor prognosis. Glioma stem cells (GSCs), characterized by stem-like properties and substantial heterogeneity, play a crucial role in tumor initiation, progression, and potential recurrence. Moreover, through complex interaction mechanisms, they contribute to the challenges associated with treatment. This review seeks to explore the distinctive characteristics and underlying mechanisms of GSCs, aiming to provide novel theoretical insights and practical strategies for precision therapy in glioma.
Keywords: Glioma stem cells, Glioma, Mechanism, Immunity, Metabolism
Introduction
When central nervous system (CNS) tumors are classified, gliomas account for 90% of all malignant CNS tumors [1]. Glioblastoma (GBM), the most aggressive subtype, constitutes 69% of glioma cases and is characterized by rapid progression and a median survival time of only 14 to 16 months [2]. Consequently, there is a critical need to develop more effective treatment strategies for GBM.
Glioma stem cells (GSCs) demonstrate characteristics such as self-renewal, the ability to differentiate into various lineages, unlimited proliferation, and significant invasiveness [3]. These attributes render GSCs pivotal in driving glioma initiation, progression, resistance to radiotherapy and chemotherapy, and malignant recurrence [4], Consequently, research on GSCs holds substantial theoretical significance and clinical application potential.
This review aims to underscore recent advancements in GSC research, examining their characteristics and underlying mechanisms to establish a theoretical foundation for the development of novel therapeutic strategies. By synthesizing existing research findings, this review further elucidates the critical role of GSCs in glioma development and progression, while also exploring potential future research directions.
Characteristics of GSCs
Definition
Within glioma tissues, GSCs constitute a minor cell population capable of self-renewal and differentiation into multiple lineages [5]. Self-renewal refers to the ability of GSCs to maintain population stability through asymmetric division while generating new GSCs [6], a process essential for the sustained growth and recurrence of tumors. Differentiation along multiple lineages demonstrates that GSCs possess the ability to transform into diverse cell types, thereby contributing to tumor heterogeneity (Fig. 1) and facilitating tumor recurrence and drug resistance [7]. The identification of GSCs has fundamentally altered the conventional understanding of glioma initiation and progression, offering novel insights for precision therapy in glioma.
Fig. 1.
GSC heterogeneity. GSC dynamically maintain population homeostasis through asymmetric division (yielding one stem cell and one differentiated progeny) and symmetric division (generating two stem cells). NPC-like and AC-like states predominantly localize to the tumor core. OPC-like cells are enriched at the infiltrative margin. MES-like subtypes preferentially reside within chronic hypoxic niches. Notably, undifferentiated GSC persist across all tumor regions, demonstrating microenvironmental plasticity. NPC-like: Neural progenitor-like; OPC-like: Oligodendrocyte progenitor-like; AC-like: astrocytic-like; MES-like: Mesenchymal-like (By Figdraw)
Stemness marker molecules
GSCs are typically characterized by the expression of specific stem cell markers. Table 1 enumerates the principal stemness marker molecules and elucidates their mechanisms of action within GSCs. However, the application of CD133 as a classic stem cell marker in the study of GSCs has long been controversial. In GBM, the CD133-positive cell subpopulation exhibits significant stem cell characteristics, including enhanced sphere-forming ability in vitro, drug resistance, and tumorigenicity in vivo [8–10], making it an important marker for studying GSCs. Nevertheless, the limitations of CD133 are equally notable. Studies have shown that its expression is dynamically plastic: some GSCs can downregulate CD133 under hypoxic or metabolic stress conditions [11], while certain CD133-negative cells can regain stem cell characteristics through epigenetic reprogramming [12]. Single-cell sequencing further reveals the presence of CD133-independent stem cell subpopulations in GBM, which maintain their stemness by activating alternative signaling pathways, such as Notch3 [13, 14]. Despite the controversies surrounding CD133 in GSCs research, its functional relevance still renders it an important subject of study.
Table 1.
GSCs stemness marker molecules and their roles
Maker | Description | Primary functions related to glioma | Citations |
---|---|---|---|
CD133/Prominin-1 | Transmembrane glycoprotein | a. Facilitates tumor initiation and stemness maintenance | [153–155] |
b. Serves as a therapeutic target | |||
Oct4/POU5F1 | Transcription factor | a. Involves in the maintenance of glioma stemness | [156–158] |
b. Promotes tumor proliferation and migration | |||
c. Acts as a therapeutic target | |||
Sox2 | Transcription factor | a. Synergistically acts with Oct4 | [5, 159, 160] |
b. Maintains the undifferentiated state of stem cells | |||
c. Influences cancer cell cycle and proliferation efficiency | |||
Nanog | Transcription factor | a. Positively correlates with tumor malignancy | [156, 161] |
b. Participates in metabolic regulation of GSCs, facilitating their survival and proliferation in harsh environments | |||
c. Involved in GSC immune evasion | |||
CD44 | Cell membrane glycoprotein | a. Characterized by high invasiveness and resistance to radiotherapy and chemotherapy | [162, 163] |
b. Hypoxia can induce phenotypic changes in cells | |||
c. Predicts patient prognosis | |||
Nestin | Intermediate filament protein | a. Promotes the formation of tumor spheres | [164, 165] |
Moreover, CD109, a glycosylphosphatidylinositol-anchored glycoprotein, has been proposed as a marker for perivascular GSCs [15]. Studies have demonstrated a clear association between CD109 and the maintenance of GSC stemness as well as disease recurrence [16, 17]. GP130, a co-receptor for cytokines such as interleukin-6 (IL-6) and tumor necrosis factor (TNF), facilitates the activation of signaling pathways, thereby influencing the stemness characteristics of GSCs [18]. The interaction between CD109 and glycoprotein 130 has been identified as a mechanism that promotes the stemness and chemoresistance of GSCs by activating the IL-6/STAT3 signaling pathway. This interaction enhances the tumorigenic potential of GSCs and contributes to their resistance to conventional therapies [19]. Furthermore, the expression of integrin α2 (ITGA2) in GSCs is associated with STAT3 phosphorylation and the activation of epithelial-mesenchymal transition (EMT), underscoring the role of stemness markers in facilitating the invasive behavior of these cells [20]. Additionally, the cellular prion protein (PrPC) and its molecular chaperone Hsp70/90 organizing protein (HOP) are implicated in the regulation of GSC proliferation and self-renewal. The PrPC-HOP complex is crucial for maintaining the stemness of GSCs, and disruption of this complex results in diminished proliferation and impaired self-renewal [21]. In summary, the expression of stem cell markers in GSCs functions not only as an identifier for these cells but also significantly contributes to their proliferation, sustains their undifferentiated state, and augments their invasive potential.
Distribution of GSCs
With advancements in imaging technologies and the identification of molecular markers, the spatial distribution of GSCs within gliomas has become increasingly discernible, allowing for more precise identification of regions enriched with GSCs. Research employing CD133 + and Nestin + markers to locate GSCs has uncovered significant variations in GSC expression across different pathological grades of gliomas, with higher grades exhibiting an increased content of GSCs [22]. Furthermore, GSCs are typically distributed around the microvasculature, emulating the niche architecture of neural stem cells. This niche is modulated by adjacent cells and the cytokines they secrete, which regulate stem cell division and their departure from the niche. The microvasculature is integral to nutrient uptake and the overall functionality of stem cells, with the intricate vascular network ensuring the supply of vital nutrients and oxygen [23]. Additionally, the interaction between endothelial cells and mesenchymal stem cells (MSCs) within the perivascular niche is essential for sustaining stem cell functions and guiding their differentiation pathways. Endothelial cells release various signaling molecules, such as endothelin-1, which can affect MSC fate by promoting differentiation into specific lineages, such as osteogenic and chondrogenic pathways, through the activation of signaling pathways like AKT [24]. This interaction highlights the role of the microvasculature not only in nutrient provision but also in regulating stem cell behavior through paracrine signaling. The distribution of GSCs is influenced not only by their intrinsic properties but also by their close association with the microenvironment. The distribution of GSCs is influenced not only by their intrinsic properties but also by their close association with the tumor microenvironment (TME) in which they reside. The TME serves as the "soil" for GSC survival, encompassing the extracellular matrix, immune cells, vascular networks, and various signaling molecules. Hypoxic conditions significantly impact gene expression regulation, particularly genes involved in the hypoxia response, thereby affecting the growth and invasion of GSCs. Central to this process are hypoxia-inducible factors (HIFs), which regulate the expression of numerous genes that facilitate tumor progression and adaptation to low-oxygen environments. For example, HIF-1α is known to activate a range of downstream target genes that promote angiogenesis, cell survival, and metabolic adaptation, thus contributing to the aggressive nature of GSCs under hypoxic conditions [25, 26]. The hypoxic microenvironment in tumors, such as GBM, not only enhances GSC proliferation but also contributes to their resistance to conventional therapies. This resistance is partially attributed to the upregulation of stemness markers and other survival pathways mediated by HIFs. For instance, the activation of the Wnt/β-catenin signaling pathway under hypoxic conditions has been demonstrated to enhance the invasive and metastatic potential of cancer cells, including GSCs. This pathway is frequently upregulated in response to HIF-1α activation, thereby reinforcing the invasive characteristics of these cells [27, 28]. Furthermore, hypoxia can induce the expression of genes involved in maintaining cancer stem cell properties, such as CD133 and vascular endothelial growth factor (VEGF), which are essential for the self-renewal and differentiation capabilities of GSCs. Consequently, the hypoxic environment not only facilitates the survival of these cells but also augments their ability to form neurospheres and migrate, thereby contributing to tumor progression and recurrence [29, 30]; Conversely, the inflammatory microenvironment enhances GSC migration and adhesion through the release of cytokines and chemokines, thus contributing to tumor heterogeneity and therapeutic resistance. The release of these factors is modulated by various elements, including the presence of other cell types such as oligodendrocyte progenitor cells and macrophages, which establish a supportive niche for GSCs at the tumor periphery [31]. The spatial distribution of GSCs within gliomas is characterized by a complex and dynamic process, shaped by the intrinsic properties of GSCs and the surrounding TME.
Regulatory mechanisms of GSCs
Regulatory mechanisms of GSCs in gliomas encompass insights from genetics, epigenetics, metabolic reprogramming the TME and the immune system.
Genetics and epigenetics
In 2008, the Cancer Genome Atlas (TCGA) database first identified the most prevalent mutated genes in gliomas, such as IDH1/2, TP53, and ATPX. Recent advancements in genomic research have significantly enhanced our comprehension of genetic mutations and structural variations in gliomas [32]. The evolution and diversity of clones during glioma progression are particularly intricate, involving epigenetic regulation that encompasses DNA methylation, histone activity, post-translational modifications (such as methylation and acetylation), and the microRNA modification profile [33]. Research has demonstrated that c-Myc, a crucial regulatory factor, is markedly expressed in GSCs and possesses the capability to activate transcriptional modules associated with stem cell characteristics, thereby inducing apoptosis [34]. GSCs exhibit heightened sensitivity to histone demethylase inhibition compared to their non-stem-like counterparts. This increased sensitivity is attributed to the distinctive chromatin features of GSCs, including the absence of the H3K9me3 mark and mutations in epigenetic regulatory genes (such as KDM4A, EZH2, and DNMT3A). KDM4A, as an H3K9me3-specific demethylase, is often overexpressed or functionally enhanced in GSCs. Its inhibition can directly restore H3K9me3 levels and trigger apoptosis [35, 36]. Mutations in EZH2 (which catalyzes H3K27me3) or DNMT3A (which regulates DNA methylation) may synergistically alter chromatin accessibility, further amplifying the vulnerability of GSCs to epigenetic interventions [37, 38]. In contrast, more differentiated glioma cells exhibit significant resistance to similar drugs due to the retention of more stable epigenetic marks, such as H3K9me3 and DNA methylation patterns [36].
Metabolic reprogramming
Metabolic reprogramming is increasingly acknowledged as a pivotal factor in tumor progression, reflecting a shift in the mechanisms by which cancer cells manage energy production and biosynthesis to support rapid growth and survival. This phenomenon is often characterized by the Warburg effect, wherein cancer cells preferentially utilize glycolysis over oxidative phosphorylation even in the presence of oxygen [39]. The hypoxic microenvironment induced by GBM not only affects the functionality of GSCs but also elevates the reliance on glycolysis [40], Although glycolysis is inefficient in adenosine triphosphate (ATP) synthesis, this metabolic shift serves as a beneficial foundation for the synthesis of novel molecules, such as nucleic acids, pyruvate, and NADPH [41]. Recent research has underscored the dual role of the pentose phosphate pathway (PPP) in cancer cells, including GSCs, under varying oxygenation conditions. Under hypoxic conditions, the PPP is frequently upregulated to satisfy the increased demand for nicotinamide adenine dinucleotide phosphate (NADPH), which is essential for maintaining redox homeostasis and supporting anabolic processes. This is particularly significant in the context of the Warburg effect, where the dependence on glycolysis can result in heightened oxidative stress. The role of the PPP in modulating oxidative stress and inflammation is well-documented, with transaldolase, a pivotal enzyme in the PPP, being implicated in the regulation of these processes and their contribution to carcinogenesis [42]. Metabolic pathways can alternate between glycolysis and the PPP, with glycolysis being activated under hypoxic conditions to facilitate cellular migration and invasion. Conversely, under normoxic conditions, the PPP is activated, promoting cell proliferation [43]. In scenarios of hypoxia combined with glucose deprivation, GSCs enhance the expression of high-affinity glucose transporter (GLUT) proteins to compete effectively with non-stem cells for glucose uptake [44]. Additionally, GSCs augment the synthesis of their metabolites by upregulating the expression of fatty acid synthase and glutamine-metabolizing enzymes, which not only supply energy to GSCs but also modulate the immune microenvironment, thereby further supporting GSC survival and invasion. Compared to the upstream and downstream effects of genetic and epigenetic processes, the influence of the metabolome is more pervasive. Mutations in the IDH1 gene, particularly the R132H variant, have been extensively investigated in the context of GBM and other gliomas. These mutations confer a neomorphic enzymatic activity that disrupts the normal function of isocitrate dehydrogenase 1 (IDH1), resulting in the production of the oncometabolite 2-hydroxyglutarate (2-HG) instead of the typical product, alpha-ketoglutarate (α-KG) [45]. This aberrant enzymatic activity is implicated in the pathogenesis of gliomas by inducing a hypermethylated state of DNA and histones, which can activate oncogenes and inactivate tumor suppressor genes, thereby contributing to tumorigenesis [46]. Furthermore, the accumulation of 2-HG disrupts cellular metabolism and inhibits histone and DNA demethylases, leading to epigenetic modifications that impede cellular differentiation and promote tumor growth [47]. Additionally, mutations in the TERT promoter activate telomere maintenance mechanisms, providing a foundation for the immortalization of GSCs [48]; EGFRvIII reinforces proliferative signals through sustained activation of the PI3K/AKT pathway and synergizes with hypoxia-induced HIF-1α to upregulate glycolytic enzymes (such as PKM2 and LDHA), driving metabolic reprogramming [49, 50]. Loss of ATRX further leads to dysregulation of chromatin remodeling complexes, enhancing the adaptability of GSCs to hypoxic microenvironments, while TP53 mutations impair DNA repair and cell cycle regulation, exacerbating genomic instability [51–54].
Tumor microenvironment
The TME is a complex and dynamic entity composed of various non-cancerous components that play critical roles in either facilitating, supporting, or hindering tumor development [55]. Interactions between stem cells and the TME are well-documented. Hypoxia, a fundamental characteristic of GBM, enhances therapeutic resistance through various mechanisms, notably by inhibiting radicals, thereby reducing the efficacy of radiotherapy [56]. Within the TME of GBM, the interaction between endothelial cells and GSCs is crucial for tumor initiation and progression. This interaction is facilitated by the formation of a perivascular niche, where GSCs are frequently located in close proximity to endothelial cells. Endothelial cells secrete various soluble factors that promote the maintenance and proliferation of GSCs, thereby increasing the aggressiveness and therapy resistance of GBM [57, 58]. Furthermore, the cross-talk between GSCs and endothelial cells extends beyond the maintenance of stemness, involving modulation of the immune microenvironment. This interaction can result in the secretion of extracellular vesicles and exosomes, which carry bioactive molecules that influence the behavior of immune cells, potentially leading to immune evasion and further promoting tumor progression [59, 60]. Pericytes play a crucial role in supporting vascular architecture by characteristically expressing platelet-derived growth factor receptors. They can be derived from GSCs to sustain tumor growth and blood supply. During GBM angiogenesis, the number of pericytes increases in tandem with the disruption of the blood–brain barrier (BBB), serving as a marker for tumor neovascularization [55, 61]. GBM is referred to as a "cold tumor" due to its immunosuppressive microenvironment: hypoxia-induced metabolic suppression of immune cells [62], inhibition of effector T cell function by TGF-β/IL-10 secreted by endothelial cells [63], immune escape induced by GSCs delivering PD-L1 via exosomes [64, 65], immune escape induced by GSCs delivering PD-L1 via exosomes [66]. Transformation strategies require multi-target synergy: combining radiotherapy with STING agonists to activate antigen presentation [67], using CSF1R inhibitors to eliminate tumor-associated macrophages [68], employing IDH inhibitors to correct metabolic abnormalities and enhance immune recognition [69], and combining chimeric antigen receptor T (CAR-T) cells with immune checkpoint blockade to overcome immune tolerance [70, 71], ultimately achieving a therapeutic breakthrough in "cold-to-hot" tumor transformation.
Immune response
Natural killer (NK) cells function as the primary effector cells in GBM immunity, with their mechanisms involving interactions between NK cell receptors and GBM cell ligands, playing a critical role in the initial defense against infections and tumor development. The efficacy of NK cells is predominantly influenced by the complex equilibrium between activating and inhibitory signals transmitted through their surface receptors [72]. These receptors facilitate the ability of NK cells to differentiate between healthy cells and those that are infected or transformed, thereby ensuring appropriate immune responses. Activating receptors, such as NKG2D, identify stress-induced ligands on target cells, leading NK cells to exert cytotoxic effects and produce cytokines. In contrast, inhibitory receptors, including those from the killer cell immunoglobulin-like receptor (KIR) family, recognize self-molecules, such as MHC class I, and inhibit inappropriate NK cell activation. This balance is essential for preventing autoimmunity and ensuring that NK cells are activated only in the presence of legitimate threats [73–75]. Additionally, the activity of NK cells is dynamically modulated by the surrounding microenvironment and the presence of cytokines. Cytokines such as interleukin-15 (IL-15) are pivotal in the priming and activation of NK cells, with proteins like cytokine-inducible SH2-containing protein acting as regulators to maintain homeostasis and prevent overactivation. This regulatory mechanism ensures that NK cells remain effective in their role in immune surveillance [76, 77]. GSCs can evade NK cell-mediated elimination through various mechanisms, including the downregulation of activating ligands, upregulation of ligands that engage inhibitory receptors, recruitment of other immunosuppressive cells [78], and alteration of the chemokine profile within the TME [79]. The remodeling of the chemokine profile within the TME is particularly critical. For instance, the CXCL12-CXCR4 axis mediates the homing of GSCs to perivascular niches and maintains their stemness [80]; CCL2 recruits tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells to construct an immunosuppressive barrier [81–83]; CXCL1/CXCL8 activates neutrophils to release pro-tumor factors such as MMP9 and VEGF [84, 85]; CCL5 regulates the migration of effector T cells but can be hijacked by GSCs to induce the infiltration of regulatory T cells (Tregs) [86, 87]. These chemokines synergistically establish a chemokine gradient, guiding immune cells towards a pro-tumor phenotype and forming a protective microenvironment that hinders NK cell-mediated killing.
T cells are integral to tumor immunity, acting as pivotal effectors in the immune system’s response to cancer. Among the diverse subsets of T cells, CD8 + cytotoxic T lymphocytes (CTLs) are particularly essential due to their capacity to directly eliminate tumor cells. These cells identify tumor antigens presented by major histocompatibility complex (MHC) class I molecules and execute their cytotoxic functions through the release of perforin and granzymes, which induce apoptosis in target cells. Nevertheless, the efficacy of CD8 + T cells can be undermined by various factors within the TME, such as the presence of immunosuppressive cells and molecules that inhibit their function. For example, Tregs can suppress CD8 + T cell activity, thereby promoting tumor immune evasion [88]. In addition to the role of CD8 + T cells, CD4 + T helper cells significantly contribute to antitumor immunity by facilitating the activation and maintenance of CD8 + T cell responses. These cells secrete cytokines that enhance the cytotoxic functions of CD8 + T cells (granzyme B expression increased 2.3-fold) and support the formation of memory T cells, thereby establishing long-term immune surveillance. Nevertheless, the role of CD4 + T cells is multifaceted, as they can also differentiate into Tregs, which promote tumor progression by suppressing effective antitumor immune responses [89]. GSCs can employ various mechanisms to evade T cell-mediated clearance, such as the high expression of PDL1 or CD86 to inhibit CTL activity, downregulation of MHC class I molecules to avoid immune recognition, expansion of immunosuppressive Tregs, and secretion of immunosuppressive factors. Furthermore, GSCs engage in metabolic competition by consuming glucose essential for T cell function and producing immunosuppressive metabolites like adenosine, which depletes tryptophan and induces T cell apoptosis.
Signaling pathways
In examining the complex biological characteristics of GSCs, it is essential to investigate their associated pathways and mechanisms of action. Figure 2 presents some of the common signaling pathways and their respective functions [90–100].
Fig. 2.
Different signaling pathways and their related functions in GSC. The Notch signaling pathway mediates interactions between GSC and endothelial cells, thereby promoting cancer cell invasion/migration. The Wnt pathway regulates angiogenesis, cancer cell invasion/migration, and the maintenance of stemness in GSC. The Sonic hedgehog pathway drives GSC self-renewal, proliferation, and drug resistance, while also inducing the secretion of stem cell factors. The NF-κB pathway sustains GSC self-renewal and proliferation, enhances drug resistance, and increases invasive capacity. The JAK/STAT pathway plays a crucial role in the regulation of GSC stemness maintenance. The PI3K/AKT pathway facilitates cancer cell invasion/migration and supports stem cell proliferation and survival (By Figdraw)
Furthermore, the interplay among various signaling pathways is crucial for the regulation of GSCs. Poly (A)-specific ribonuclease (PARN) has been identified as a pivotal regulator in the activation of the EGFR-STAT3 signaling pathway, thereby facilitating the self-renewal and proliferation of GSCs [101]; The interaction among the ERK, integrin α6, and N-cadherin signaling pathways enhances the invasive capabilities of GSCs [102]; Additionally, the crosstalk between the Wnt/β-catenin and TERT signaling pathways contributes to the maintenance of GSC stem cell characteristics and confers treatment resistance, with CD133 playing a significant role in this interaction [100]; The interaction between the NOTCH and p53 pathways has been demonstrated to influence GSC progression [103]; Circular RNAs, such as circKPNB1 and circZEB1, have been shown to promote the malignant phenotype of GSCs through the TNF-α/NF-κB pathway, establishing a positive feedback loop that enhances GSC proliferation and survival [96, 97]; Lastly, the FMR1/circCHAF1A/miR-211-5p/HOXC8 feedback loop promotes GSC proliferation and tumorigenesis via the MDM2-dependent p53 signaling pathway [104]. Comprehending the intricate interactions among these signaling pathways in GSCs is crucial for identifying novel therapeutic targets and advancing the development of more effective treatments for glioma.
Interventions targeting GSCs
Given the pivotal role of GSCs in the initiation and progression of GBM, as well as their intricate interactions with the TME, therapeutic strategies that focus on the targeted elimination of GSCs by modulating the microenvironment have garnered increasing research interest. However, the BBB, serving as a pivotal physiological barrier that restricts the delivery of drugs to the central nervous system, poses significant challenges to targeted therapy for GSCs due to its unique structure. Studies have demonstrated that more than 98% of small-molecule drugs and virtually all large-molecule therapeutic agents are unable to effectively penetrate the BBB [105]. In recent years, emerging technologies such as nanocarrier systems designed for BBB permeability, focused ultrasound-mediated modulation of barrier permeability, and immune cell-mediated targeted delivery systems have offered novel solutions to overcome this barrier. Consequently, this review emphasizes therapeutic approaches related to immunotherapy and metabolism while exploring nascent BBB penetration technologies. This integrated perspective provides a theoretical framework for developing multimodal treatment strategies.
Targeted BBB
The BBB, serving as the core protective mechanism of the central nervous system, comprises tightly connected endothelial cells, a basement membrane, and a pericyte complex. While effectively blocking pathogens, this complex also poses a significant limitation on the delivery efficiency of drugs into the brain. Targeted therapy against GSCs in GBM faces additional challenges due to the low permeability in hypoxic regions of the tumor core. Currently, innovative strategies to breach the BBB primarily focus on three major directions:
4.1.1. Physical intervention techniques temporarily open barriers in a controlled manner. Focused ultrasound combined with microbubbles (FUS-MB) technology leverages the ultrasonic cavitation effect to enhance the permeability of the BBB. Clinical trials have demonstrated that this approach can increase the intracranial concentration of doxorubicin by 4.7 times [106, 107], and adjustments to ultrasound parameters and microbubble dosages can avoid brain tissue damage [108, 109]. Additionally, magnetically guided iron oxide nanoparticles can traverse the intact BBB through external magnetic field gradients, enabling effective drug delivery within the brain and significantly enhancing drug accumulation in the target region [110].
4.1.2. Biomimetic engineering strategies leverage natural cell penetration mechanisms to overcome barriers. PLGA nanoparticles coated with macrophage membranes exploit the homing properties of immune cells to enhance targeting efficiency by 12-fold [111]. When combined with genetic engineering modifications, these nanoparticles can extend their residence time in vivo and enhance their therapeutic efficacy [112]. Exosomes traverse BBB models through heparin sulfate proteoglycan-mediated transport mechanisms [113], while novel cationic lipid nanoparticles (LNPs), through optimized physicochemical properties, have successfully achieved effective delivery and gene silencing of siRNA in mouse glioblastoma, thereby activating T-cell-dependent antitumor immune responses [114].
4.1.3. Intelligent nanocarriers achieve precise drug release by responding to the tumor microenvironment. pH-sensitive polyethylene glycol-polylactic acid copolymers trigger drug release in acidic microenvironments [115], and by incorporating RGD peptides, they achieve dual targeting of both blood vessels and tumors [116]. Biomimetic liposomes mimic the structure of low-density lipoprotein (LDL) and enhance paclitaxel delivery efficiency through LDL receptor-mediated endocytosis [117].
Despite significant advancements, this field still faces challenges such as balancing delivery efficiency with safety, ensuring stability in large-scale production of carriers, and addressing receptor fluctuations due to tumor heterogeneity [118–120]. In response, cutting-edge research is shifting towards multimodal synergistic strategies, such as combining FUS-MB with engineered exosomes to simultaneously achieve BBB opening and active targeting [121], or utilizing artificial intelligence models to optimize drug selection and design by predicting drug permeability [122]. These interdisciplinary innovations are propelling neuro-oncological treatment towards an era of precise regulation.
Targeted immunotherapy
Immunotherapy, in particular, represents an innovative strategy in cancer treatment, attracting considerable attention due to its potential to induce durable responses in patients with various cancer types. Unlike conventional cancer therapies, which often rely on non-specific interventions such as surgery, radiation, and chemotherapy, immunotherapy leverages the specificity of the immune system to selectively target and eradicate cancer cells. Immunotherapy targeting GSCs primarily involves three principal strategies: tumor vaccines designed to elicit immune responses against antigens present on the surface of GSCs; CAR-T cell therapy, which entails the modification of T cells to specifically target and eradicate GSCs; and immune checkpoint inhibitors that obstruct tumor immune evasion mechanisms, thereby restoring T cell functionality [41]. NK cells have attracted considerable interest due to their potent cytotoxic capabilities and adaptability within the TME. Clinical trials have been conducted using autologous and IL-2-activated NK cells, while allogeneic NK cells or antibodies targeting NK cell inhibitory receptors serve to impede GBM cells from recognizing their own MHC class I molecules [123]. Notably, NK cells engineered with CARs and directed against HER2 have demonstrated the ability to eliminate glioma cells and neurospheres [124]. Additionally, research has shown that NK cells can be educated or primed to enhance their cytotoxic efficacy against glioma cells. For example, NK cell-derived exosomes, when applied to NK cells, have been found to enhance their antitumor activity, resulting in more effective targeting and destruction of tumor cells (removes 86% of tumours in 4 weeks) [125]. Concurrently, CAR-T cell therapy has emerged as a promising strategy for targeting GSCs. A primary approach in utilizing CAR-T cells against GSCs involves the identification and targeting of specific antigens that are overexpressed in GSCs but absent in normal cells. For example, the epidermal growth factor receptor variant III (EGFRvIII) serves as a tumor-specific antigen expressed in a subset of gliomas, including GSCs. CAR-T cells engineered to target EGFRvIII have demonstrated the capacity to recognize and eradicate GSCs, thereby reducing tumor growth and enhancing survival in preclinical models [126, 127]. In addition to EGFRvIII, other antigens such as IL-13 receptor alpha 2 (IL13Rα2) have been identified as viable targets for CAR-T cell therapy against GSCs. Research has shown that CAR-T cells targeting IL13Rα2 can effectively eliminate GSCs both in vitro and in vivo, resulting in significant tumor regression without impacting normal brain tissue [128, 129]. Furthermore, the TME plays a critical role in the efficacy of CAR-T cell therapy. The immunosuppressive characteristics of the TME in GBM can impede the activity of CAR-T cells. To address these challenges, strategies have been developed to enhance the functionality of CAR-T cells, such as incorporating additional features like a TGFβ-trap. This modification not only improves the anti-tumor efficacy of CAR-T cells but also modulates the TME to foster a more pro-inflammatory and anti-tumorigenic milieu [130, 131]. Furthermore, HLA-G, a nonclassical HLA class I molecule, has been identified to form dimers that inhibit NK cell cytotoxicity through interactions with inhibitory receptors. This mechanism has been documented in various cancers, including gliomas, where the TME can promote the formation of HLA-G dimers, thereby shielding tumor cells from NK cell-mediated lysis [132]. Additionally, GSCs have been observed to express HLA-G, contributing to their resistance against NK cell-mediated cytotoxicity [133]. The interaction between killer cell KIRs on NK cells and HLA molecules on target cells is also crucial in modulating the immune response against GSCs. KIR/HLA interactions can inhibit NK cell activation, thus diminishing their capacity to mediate antibody-dependent cellular cytotoxicity (ADCC) against glioma cells. This inhibition exhibits selectivity and may vary contingent upon the specific therapeutic antibodies employed, as demonstrated in studies comparing the effects of rituximab and GA101 (obinutuzumab) [134]. Additionally, the expression of particular HLA alleles and their corresponding supertypes can modulate the immune response to GSCs. Certain HLA supertypes have been correlated with enhanced survival outcomes in patients receiving treatments such as hematopoietic cell transplantation [135], indicating that diversity in HLA presentation may augment immune responsiveness.
Targeted metabolic therapy
Considering the pivotal role of GSCs in metabolic regulation, strategies targeting metabolic pathways have emerged as promising approaches for the selective eradication of GSCs. One mechanism by which GSCs facilitate tumor progression involves the establishment of a positive feedback loop encompassing glycolysis, extracellular acidification, and immunosuppression. The glycolytic shift in GSCs results in lactate production, which is subsequently exported from the cells, leading to extracellular acidification. The acidic milieu within the TME exacerbates its immunosuppressive properties by inhibiting the function of CTLs and NK cells, while simultaneously facilitating the recruitment and polarization of immunosuppressive cell types, such as Tregs and TAMs [136, 137]. The interplay between GSCs and immune cells is pivotal in maintaining this feedback loop. GSCs are capable of secreting factors that modulate immune cell activity, thereby suppressing anti-tumor immune responses. For example, the secretion of transforming growth factor-beta (TGF-β) by GSCs can induce a metabolic shift in immune cells from oxidative phosphorylation to glycolysis, thereby augmenting the immunosuppressive environment [137]. Furthermore, TAMs, which are frequently polarized to the M2 phenotype within the TME, can release cytokines that promote glycolysis in GSCs, thereby perpetuating the feedback loop [138, 139]. Additionally, the acidic microenvironment resulting from increased glycolysis can activate acid-sensing ion channels on immune cells, thereby further facilitating tumor immune evasion. The intricate relationship between metabolic reprogramming and immune modulation underscores the complexity of the TME in GBM and highlights the challenges associated with developing effective therapies [140, 141]. Targeting this positive feedback loop offers a promising therapeutic strategy. By disrupting glycolysis or neutralizing the acidic environment, it may be feasible to restore immune cell function and enhance the efficacy of immunotherapies. For instance, inhibiting key glycolytic enzymes or employing buffering agents to counteract extracellular acidification could potentially disrupt the cycle of immunosuppression and improve patient outcomes [142, 143]. Additionally, regulating glucose metabolism in GSCs presents further opportunities for antitumor effects, as inhibiting GLUT1 can reduce glucose uptake by GSCs, thereby impairing their self-renewal capacity [144]. Hexokinase 2 (HK2), a critical enzyme that catalyzes the initial step of glycolysis, is frequently overexpressed in cancer cells, leading to increased glycolytic activity. This enzyme not only plays a crucial role in glucose metabolism but also facilitates cancer cell survival and proliferation by promoting the Warburg effect. Importantly, elevated HK2 expression was independently associated with poorer prognosis (Cox proportional hazards model, P < 0.006) after adjusting for age [145–147]. Research has demonstrated that targeting HK2 can effectively suppress tumor growth. For example, the application of specific inhibitors such as Benitrobenrazide has been shown to impede cancer cell proliferation by directly targeting HK2, resulting in reduced glycolysis and increased apoptosis in cancer cells [148]. Similarly, xanthohumol, a natural compound, has exhibited anti-tumor effects on GBM by inhibiting glycolysis through the downregulation of HK2 expression. This effect was evidenced by a 50% reduction in tumor volume in murine models [148]. Comparable strategies can also be employed to disrupt fatty acid and glutamine metabolism in GSCs, further inhibiting their growth and survival. Nevertheless, given the heterogeneity of metabolic pathways and the adaptability of GSCs to metabolic stress, such therapies often necessitate combination with other treatment modalities to enhance efficacy in clinical applications. GSCs express high-affinity GLUT3, which confers a survival advantage in hypoglycemic microenvironments, thereby contributing to the maintenance of tumor hierarchy and potentially bearing prognostic significance [149]. The promotion of pyrimidine synthesis not only sustains the nucleotide pool necessary for DNA and RNA synthesis but also fulfills the energy and metabolic demands of GSCs to adapt to diverse tumor microenvironmental conditions [150]. Consequently, targeting pyrimidine synthesis may disrupt the metabolic reliance of GSCs and enhance the therapeutic efficacy against GBM [44, 151, 152].
Conclusions and future perspectives
GSCs have emerged as pivotal drivers of glioma progression, orchestrating tumorigenesis through their self-renewal capabilities, invasive potential, and dynamic interactions with the TME. These cells maintain stemness via dysregulated signaling pathways, including IL-6/STAT3 and EMT, while establishing an immunosuppressive niche through exosomal PD-L1 secretion, Treg recruitment, and metabolic symbiosis. The "cold tumor" phenotype of GBM is reinforced by hypoxia-induced immunosuppression, endothelial-derived TGF-β/IL-10, and lactic acid-driven extracellular acidification, which collectively impair cytotoxic T/NK cell activity and promote immune evasion. GSCs further exploit glycolytic dominance to acidify the TME, activating acid-sensitive ion channels on immune cells and fostering a self-perpetuating cycle of immunosuppression. This cycle is amplified by TGF-β-mediated metabolic reprogramming of immune cells and reciprocal cytokine-enhanced glycolysis in GSCs, creating a feedforward loop that sustains tumor progression. Current therapeutic strategies aim to disrupt these interactions through dual targeting of metabolic and immune pathways. Inhibiting glycolysis (e.g., GLUT1/HK2 suppression), neutralizing acidic microenvironments, or combining IL-6/STAT3 blockade with immune checkpoint inhibitors demonstrates synergistic potential to restore antitumor immunity. Innovative approaches, such as bispecific antibodies co-targeting metabolic enzymes and immune checkpoints, oxidative phosphorylation inhibitors paired with PD-1/CTLA-4 blockade, and CAR-T therapies exploiting GSC glutamine addiction, highlight the promise of combinatorial regimens. Additionally, nanotechnology-driven delivery systems capable of bypassing the BBB and spatially controlled drug release are critical for enhancing therapeutic efficacy. Advances in spatial transcriptomics, CRISPR-based metabolic screens, and engineered immune cells (e.g., TGF-β-resistant CAR-T) will refine therapeutic precision. By integrating AI-driven predictive models to tailor therapies and address resistance, next-generation strategies may transform GBM into a manageable condition. Ultimately, dismantling the GSC-TME axis through metabolic-immune synergy represents a transformative frontier in glioma therapeutics, underscoring the need for translational innovation to bridge mechanistic insights with clinical realities.
Artificial intelligence
The authors declare that they have not use AI-generated work in this manuscript and all work are owned by the authors.
Acknowledgements
We express gratitude to the funding sponsors.
Abbreviations
- GSCs
Glioma stem cells
- CNS
Central nervous system
- GBM
Glioblastoma
- IL-6
Interleukin-6
- TNF
Tumor necrosis factor
- ITGA2
Integrin α2
- EMT
Epithelial-mesenchymal transition
- PrPC
Cellular prion protein
- HOP
Hsp70/90 organizing protein
- MSCs
Mesenchymal stem cells
- TME
Tumor microenvironment
- HIFs
Hypoxia-inducible factors
- VEGF
Vascular endothelial growth factor
- TCGA
The cancer genome atlas
- H3K9me3
H3 lysine 9 trimethylation
- ATP
Adenosine triphosphate
- PPP
Pentose phosphate pathway
- NADPH
Nicotinamide adenine dinucleotide phosphate
- GLUT
Glucose transporter
- 2-HG
2-Hydroxyglutarate
- α-KG
Alpha-ketoglutarate
- BBB
Blood-brain barrier
- CAR-T
Chimeric antigen receptor T
- NK
Natural killer
- KIR
Killer cell immunoglobulin-like receptor
- TAMs
Tumor-associated macrophages
- Tregs
Regulatory T cells
- CTLs
Cytotoxic T lymphocytes
- PARN
Poly (A)-specific ribonuclease
- LNPs
Lipid nanoparticles
- LDL
Low-density lipoprotein
- CAR-T
Chimeric antigen receptor T
- EGFRvIII
Epidermal growth factor receptor variant III
- ADCC
Antibody-dependent cellular cytotoxicity
- HK 2
Hexokinase 2
Author contributions
JYH reviewed the literature and wrote the manuscript; XWY provided relevant literature and reviewed the manuscript; SSH proposed the framework of the article and reviewed the manuscript. All authors read and approved the final manuscript.
Funding
This work was financially supported by Project of Medicine and Health Science and Technology of Zhejiang Province (NO.2024KY015).
Availability of data and materials
Not applicable.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors have declared no competing of interest.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Weller M, Wick W, Aldape K, et al. Glioma. Nat Rev Dis Primers. 2015;1:15017. [DOI] [PubMed] [Google Scholar]
- 2.Forjaz G, Barnholtz-Sloan JS, Kruchko C, et al. An updated histology recode for the analysis of primary malignant and nonmalignant brain and other central nervous system tumors in the surveillance, epidemiology, and end results program. Neurooncol Adv. 2021;3(1):vdaa175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sareddy GR, Viswanadhapalli S, Surapaneni P, et al. Novel KDM1A inhibitors induce differentiation and apoptosis of glioma stem cells via unfolded protein response pathway. Oncogene. 2017;36(17):2423–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Jiang Y, Zhou J, Zhao J, et al. MiR-18a-downregulated RORA inhibits the proliferation and tumorigenesis of glioma using the TNF-α-mediated NF-κB signaling pathway. EBioMedicine. 2020;52: 102651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lopez-Bertoni H, Johnson A, Rui Y, et al. Sox2 induces glioblastoma cell stemness and tumor propagation by repressing TET2 and deregulating 5hmC and 5mC DNA modifications. Signal Transduct Target Ther. 2022;7(1):37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Easwaran S, van Ligten M, Kui M, et al. Enhanced germline stem cell longevity in Drosophila diapause. Nat Commun. 2022;13(1):711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mayer B, Jauch KW, GüNTHERT U, et al. De-novo expression of CD44 and survival in gastric cancer. Lancet. 1993;342(8878):1019–22. [DOI] [PubMed] [Google Scholar]
- 8.Kim JS, Shin DH, Kim JS. Dual-targeting immunoliposomes using angiopep-2 and CD133 antibody for glioblastoma stem cells. J Control Release. 2018;269:245–57. [DOI] [PubMed] [Google Scholar]
- 9.Jamal M, Rath BH, Tsang PS, et al. The brain microenvironment preferentially enhances the radioresistance of CD133(+) glioblastoma stem-like cells. Neoplasia. 2012;14(2):150–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Shin DH, Xuan S, Kim WY, et al. CD133 antibody-conjugated immunoliposomes encapsulating gemcitabine for targeting glioblastoma stem cells. J Mater Chem B. 2014;2(24):3771–81. [DOI] [PubMed] [Google Scholar]
- 11.Matsumoto K, Arao T, Tanaka K, et al. mTOR signal and hypoxia-inducible factor-1 alpha regulate CD133 expression in cancer cells. Cancer Res. 2009;69(18):7160–4. [DOI] [PubMed] [Google Scholar]
- 12.Sun Y, Kong W, Falk A, et al. CD133 (Prominin) negative human neural stem cells are clonogenic and tripotent. PLoS ONE. 2009;4(5): e5498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.He Y, Døssing KBV, Sloth AB, et al. Quantitative evaluation of stem-like markers of human glioblastoma using single-Cell RNA sequencing datasets. Cancers (Basel). 2023;15(5):1557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Galdieri L, Jash A, Malkova O, et al. Defining phenotypic and functional heterogeneity of glioblastoma stem cells by mass cytometry. JCI Insight. 2021;6(4):e128456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Silver DJ, Lathia JD. Revealing the glioma cancer stem cell interactome, one niche at a time. J Pathol. 2018;244(3):260–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bastola S, Pavlyukov MS, Yamashita D, et al. Glioma-initiating cells at tumor edge gain signals from tumor core cells to promote their malignancy. Nat Commun. 2020;11(1):4660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Li C, Cho HJ, Yamashita D, et al. Tumor edge-to-core transition promotes malignancy in primary-to-recurrent glioblastoma progression in a PLAGL1/CD109-mediated mechanism. Neurooncol Adv. 2020;2(1):vdaa163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Shi Y, Zhou W, Cheng L, et al. Tetraspanin CD9 stabilizes gp130 by preventing its ubiquitin-dependent lysosomal degradation to promote STAT3 activation in glioma stem cells. Cell Death Differ. 2017;24(1):167–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Filppu P, Tanjore Ramanathan J, Granberg KJ, et al. CD109-GP130 interaction drives glioblastoma stem cell plasticity and chemoresistance through STAT3 activity. JCI Insight. 2021;6(9):e141486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Zhang J, Li R, Zhang H, et al. ITGA2 as a prognostic factor of glioma promotes GSCs invasion and EMT by activating STAT3 phosphorylation. Carcinogenesis. 2024;45(4):235–46. [DOI] [PubMed] [Google Scholar]
- 21.Iglesia RP, Prado MB, Cruz L, et al. Engagement of cellular prion protein with the co-chaperone Hsp70/90 organizing protein regulates the proliferation of glioblastoma stem-like cells. Stem Cell Res Ther. 2017;8(1):76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Jiang Y, Zhou J, Luo P, et al. Prosaposin promotes the proliferation and tumorigenesis of glioma through toll-like receptor 4 (TLR4)-mediated NF-κB signaling pathway. EBioMedicine. 2018;37:78–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Louissaint A, Rao S, Leventhal C, et al. Coordinated interaction of neurogenesis and angiogenesis in the adult songbird brain. Neuron. 2002;34(6):945–60. [DOI] [PubMed] [Google Scholar]
- 24.Tsai TL, Wang B, Squire MW, et al. Endothelial cells direct human mesenchymal stem cells for osteo- and chondro-lineage differentiation through endothelin-1 and AKT signaling. Stem Cell Res Ther. 2015;6(1):88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Papale M, Buccarelli M, Mollinari C, et al. Hypoxia, inflammation and necrosis as determinants of glioblastoma cancer stem cells progression. Int J Mol Sci. 2020;21(8):2660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Cristofaro I, Limongi C, Piscopo P, et al. M2 receptor activation counteracts the glioblastoma cancer stem cell response to hypoxia condition. Int J Mol Sci. 2020;21(5):1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Huang F, Chen J, Lan R, et al. Hypoxia induced δ-Catenin to enhance mice hepatocellular carcinoma progression via Wnt signaling. Exp Cell Res. 2019;374(1):94–103. [DOI] [PubMed] [Google Scholar]
- 28.Liu HL, Liu D, Ding GR, et al. Hypoxia-inducible factor-1α and Wnt/β-catenin signaling pathways promote the invasion of hypoxic gastric cancer cells. Mol Med Rep. 2015;12(3):3365–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Musah-Eroje A, Watson S. Adaptive changes of glioblastoma cells following exposure to hypoxic (1% Oxygen) tumour microenvironment. Int J Mol Sci. 2019;20(9):2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kahlert UD, Maciaczyk D, Dai F, et al. Resistance to hypoxia-induced, BNIP3-mediated cell death contributes to an increase in a CD133-positive cell population in human glioblastomas in vitro. J Neuropathol Exp Neurol. 2012;71(12):1086–99. [DOI] [PubMed] [Google Scholar]
- 31.Hide T, Komohara Y, Miyasato Y, et al. Oligodendrocyte progenitor cells and macrophages/microglia produce glioma stem cell niches at the tumor border. EBioMedicine. 2018;30:94–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kreth S, Thon N, Kreth FW. Epigenetics in human gliomas. Cancer Lett. 2014;342(2):185–92. [DOI] [PubMed] [Google Scholar]
- 34.Roohinejad Z, Bahramian S, Shamsabadi FT, et al. Upregulation of the c-MYC oncogene and adjacent long noncoding RNAs PVT1 and CCAT1 in esophageal squamous cell carcinoma. BMC Cancer. 2023;23(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Young NL, Dere R. Mechanistic insights into KDM4A driven genomic instability. Biochem Soc Trans. 2021;49(1):93–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mallm JP, Windisch P, Biran A, et al. Glioblastoma initiating cells are sensitive to histone demethylase inhibition due to epigenetic deregulation. Int J Cancer. 2020;146(5):1281–92. [DOI] [PubMed] [Google Scholar]
- 37.Kalkan R. The importance of mutational drivers in GBM. Crit Rev Eukaryot Gene Expr. 2016;26(1):19–26. [DOI] [PubMed] [Google Scholar]
- 38.Harachi M, Masui K, Shimizu E, et al. DNA hypomethylator phenotype reprograms glutamatergic network in receptor tyrosine kinase gene-mutated glioblastoma. Acta Neuropathol Commun. 2024;12(1):40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fendt SM. 100 years of the Warburg effect: a cancer metabolism endeavor. Cell. 2024;187(15):3824–8. [DOI] [PubMed] [Google Scholar]
- 40.Stieber D, Abdul Rahim SA, Niclou SP. Novel ways to target brain tumour metabolism. Expert Opin Ther Targets. 2011;15(10):1227–39. [DOI] [PubMed] [Google Scholar]
- 41.Carruthers RD, Ahmed SU, Ramachandran S, et al. Replication stress drives constitutive activation of the DNA damage response and radioresistance in glioblastoma stem-like cells. Cancer Res. 2018;78(17):5060–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Perl A, Hanczko R, Telarico T, et al. Oxidative stress, inflammation and carcinogenesis are controlled through the pentose phosphate pathway by transaldolase. Trends Mol Med. 2011;17(7):395–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wu A, Wei J, Kong LY, et al. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro Oncol. 2010;12(11):1113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang X, Yang K, Wu Q, et al. Targeting pyrimidine synthesis accentuates molecular therapy response in glioblastoma stem cells. Sci Transl Med. 2019;11(504):eaau4972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Jin G, Reitman ZJ, Spasojevic I, et al. 2-hydroxyglutarate production, but not dominant negative function, is conferred by glioma-derived NADP-dependent isocitrate dehydrogenase mutations. PLoS ONE. 2011;6(2): e16812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Semukunzi H, Roy D, Li H, et al. IDH mutations associated impact on related cancer epidemiology and subsequent effect toward HIF-1α. Biomed Pharmacother. 2017;89:805–11. [DOI] [PubMed] [Google Scholar]
- 47.Kaminska B, Czapski B, Guzik R, et al. Consequences of IDH1/2 mutations in gliomas and an assessment of inhibitors targeting mutated IDH proteins. Molecules. 2019;24(5):968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mancini A, Xavier-Magalhães A, Woods WS, et al. Disruption of the β1L isoform of GABP reverses glioblastoma replicative immortality in a TERT promoter mutation-dependent manner. Cancer Cell. 2018;34(3):513–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zhang Z, Yao L, Yang J, et al. PI3K/Akt and HIF-1 signaling pathway in hypoxia-ischemia (Review). Mol Med Rep. 2018;18(4):3547–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Su X, Yang Y, Guo C, et al. NOX4-derived ROS mediates TGF-β1-induced metabolic reprogramming during epithelial-mesenchymal transition through the PI3K/AKT/HIF-1α pathway in glioblastoma. Oxid Med Cell Longev. 2021;2021:5549047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Teng YC, Sundaresan A, O’Hara R, et al. ATRX promotes heterochromatin formation to protect cells from G-quadruplex DNA-mediated stress. Nat Commun. 2021;12(1):3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ghaleb A, Yallowitz A, Marchenko N. Irradiation induces p53 loss of heterozygosity in breast cancer expressing mutant p53. Commun Biol. 2019;2:436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.BéRUBé NG. ATRX in chromatin assembly and genome architecture during development and disease. Biochem Cell Biol. 2011;89(5):435–44. [DOI] [PubMed] [Google Scholar]
- 54.Aguilera P, López-contreras AJ. ATRX, a guardian of chromatin. Trends Genet. 2023;39(6):505–19. [DOI] [PubMed] [Google Scholar]
- 55.Schiffer D, Annovazzi L, Casalone C, et al. Glioblastoma: microenvironment and niche concept. Cancers (Basel). 2018;11(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Guardia GDA, Correa BR, Araujo PR, et al. Proneural and mesenchymal glioma stem cells display major differences in splicing and lncRNA profiles. NPJ Genom Med. 2020;5:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Sharma A, Shiras A. Cancer stem cell-vascular endothelial cell interactions in glioblastoma. Biochem Biophys Res Commun. 2016;473(3):688–92. [DOI] [PubMed] [Google Scholar]
- 58.Brooks MD, Jackson E, Warrington NM, et al. PDE7B is a novel, prognostically significant mediator of glioblastoma growth whose expression is regulated by endothelial cells. PLoS ONE. 2014;9(9): e107397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Guo X, Sui R, Piao H. Exosomes-mediated crosstalk between glioma and immune cells in the tumor microenvironment. CNS Neurosci Ther. 2023;29(8):2074–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Mondal A, Kumari Singh D, Panda S, et al. Extracellular vesicles as modulators of tumor microenvironment and disease progression in glioma. Front Oncol. 2017;7:144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am J Hematol. 2010;85(8):593–8. [DOI] [PubMed] [Google Scholar]
- 62.Mulligan JK, Rosenzweig SA, Young MR. Tumor secretion of VEGF induces endothelial cells to suppress T cell functions through the production of PGE2. J Immunother. 2010;33(2):126–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bedke T, Pretsch L, Karakhanova S, et al. Endothelial cells augment the suppressive function of CD4+ CD25+ Foxp3+ regulatory T cells: involvement of programmed death-1 and IL-10. J Immunol. 2010;184(10):5562–70. [DOI] [PubMed] [Google Scholar]
- 64.Yang Y, Li CW, Chan LC, et al. Exosomal PD-L1 harbors active defense function to suppress T cell killing of breast cancer cells and promote tumor growth. Cell Res. 2018;28(8):862–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Himes BT, Peterson TE, de Mooij T, et al. The role of extracellular vesicles and PD-L1 in glioblastoma-mediated immunosuppressive monocyte induction. Neuro Oncol. 2020;22(7):967–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lu Y, Chen L, Li L, et al. Exosomes derived from brain metastatic breast cancer cells destroy the blood-brain barrier by carrying lncRNA GS1-600G8.5. Biomed Res Int. 2020;2020:7461727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lauret Marie Joseph E, Kirilovsky A, Lecoester B, et al. Chemoradiation triggers antitumor Th1 and tissue resident memory-polarized immune responses to improve immune checkpoint inhibitors therapy. J Immunother Cancer. 2021;9(7):e002256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Magkouta SF, Vaitsi PC, Pappas AG, et al. CSF1/CSF1R axis blockade limits mesothelioma and enhances efficiency of anti-PDL1 immunotherapy. Cancers (Basel). 2021;13(11):2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wu MJ, Kondo H, Kammula AV, et al. Mutant IDH1 inhibition induces dsDNA sensing to activate tumor immunity. Science. 2024;385(6705):eadl6173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Grosser R, Cherkassky L, Chintala N, et al. Combination immunotherapy with CAR T cells and checkpoint blockade for the treatment of solid tumors. Cancer Cell. 2019;36(5):471–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dosset M, Joseph EL, Rivera Vargas T, et al. Modulation of determinant factors to improve therapeutic combinations with immune checkpoint inhibitors. Cells. 2020;9(7):1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Garnier D, Renoult O, Alves-Guerra MC, et al. Glioblastoma stem-like cells, metabolic strategy to kill a challenging target. Front Oncol. 2019;9:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ganesan S, HöGLUND P. Inhibitory receptor crosslinking quantitatively dampens calcium flux induced by activating receptor triggering in NK cells. Front Immunol. 2018;9:3173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Chen Y, Lu D, Churov A, et al. Research progress on NK cell receptors and their signaling pathways. Mediators Inflamm. 2020;2020:6437057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ravindranath MH, Filippone EJ, Devarajan A, et al. Enhancing natural killer and CD8(+) T cell-mediated anticancer cytotoxicity and proliferation of CD8(+) T cells with HLA-E monospecific monoclonal antibodies. Monoclon Antib Immunodiagn Immunother. 2019;38(2):38–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Delconte RB, Guittard G, Goh W, et al. NK cell priming from endogenous homeostatic signals is modulated by CIS. Front Immunol. 2020;11:75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Koh EK, Lee HR, Son WC, et al. Antitumor effects of NK cells expanded by activation pre-processing of autologous feeder cells before irradiation in colorectal cancer. Oncol Lett. 2023;25(6):232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Kim H, Kim J, Sa JK, et al. Calcipotriol, a synthetic vitamin D analog, promotes antitumor immunity via CD4+T-dependent CTL/NK cell activation. Biomed Pharmacother. 2022;154: 113553. [DOI] [PubMed] [Google Scholar]
- 79.Salazar N, Carlson JC, Huang K, et al. A chimeric antibody against ACKR3/CXCR7 in combination with TMZ activates immune responses and extends survival in mouse GBM models. Mol Ther. 2018;26(5):1354–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Giordano FA, Link B, Glas M, et al. Targeting the post-irradiation tumor microenvironment in glioblastoma via inhibition of CXCL12. Cancers (Basel). 2019;11(3):272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yang Z, Li H, Wang W, et al. CCL2/CCR2 Axis promotes the progression of salivary adenoid cystic carcinoma via recruiting and reprogramming the tumor-associated macrophages. Front Oncol. 2019;9:231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Fei L, Ren X, Yu H, et al. Targeting the CCL2/CCR2 Axis in Cancer Immunotherapy: One stone, three birds? Front Immunol. 2021;12: 771210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gao X, Sui H, Zhao S, et al. Immunotherapy targeting myeloid-derived suppressor cells (MDSCs) in tumor microenvironment. Front Immunol. 2020;11: 585214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Braile M, Cristinziano L, Marcella S, et al. LPS-mediated neutrophil VEGF-A release is modulated by cannabinoid receptor activation. J Leukoc Biol. 2021;109(3):621–31. [DOI] [PubMed] [Google Scholar]
- 85.Yuan M, Zhu H, Xu J, et al. Tumor-derived CXCL1 promotes lung cancer growth via recruitment of tumor-associated neutrophils. J Immunol Res. 2016;2016:6530410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Nie Y, Huang H, Guo M, et al. Breast phyllodes tumors recruit and repolarize tumor-associated macrophages via secreting CCL5 to promote malignant progression, which can be inhibited by CCR5 inhibition therapy. Clin Cancer Res. 2019;25(13):3873–86. [DOI] [PubMed] [Google Scholar]
- 87.Taki M, Abiko K, Baba T, et al. Snail promotes ovarian cancer progression by recruiting myeloid-derived suppressor cells via CXCR2 ligand upregulation. Nat Commun. 2018;9(1):1685. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chen X, Du Y, Lin X, et al. CD4+CD25+ regulatory T cells in tumor immunity. Int Immunopharmacol. 2016;34:244–9. [DOI] [PubMed] [Google Scholar]
- 89.Zamarron BF, Chen W. Dual roles of immune cells and their factors in cancer development and progression. Int J Biol Sci. 2011;7(5):651–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sharifzad F, Ghavami S, Verdi J, et al. Glioblastoma cancer stem cell biology: potential theranostic targets. Drug Resist Updat. 2019;42:35–45. [DOI] [PubMed] [Google Scholar]
- 91.Bhat KPL, Balasubramaniyan V, Vaillant B, et al. Mesenchymal differentiation mediated by NF-κB promotes radiation resistance in glioblastoma. Cancer Cell. 2013;24(3):331–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Rheinbay E, Suvà ML, Gillespie SM, et al. An aberrant transcription factor network essential for Wnt signaling and stem cell maintenance in glioblastoma. Cell Rep. 2013;3(5):1567–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Han B, Wang R, Chen Y, et al. QKI deficiency maintains glioma stem cell stemness by activating the SHH/GLI1 signaling pathway. Cell Oncol (Dordr). 2019;42(6):801–13. [DOI] [PubMed] [Google Scholar]
- 94.Lospinoso Severini L, Quaglio D, Basili I, et al. A smo/gli multitarget hedgehog pathway inhibitor impairs tumor growth. Cancers (Basel). 2019;11(10):1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.McCord AM, Jamal M, Williams ES, et al. CD133+ glioblastoma stem-like cells are radiosensitive with a defective DNA damage response compared with established cell lines. Clin Cancer Res. 2009;15(16):5145–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jiang Y, Zhao J, Liu Y, et al. CircKPNB1 mediates a positive feedback loop and promotes the malignant phenotypes of GSCs via TNF-α/NF-κB signaling. Cell Death Dis. 2022;13(8):697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Zhang G, Jiang Y, Wang Z, et al. FUS/circZEB1/miR-128-3p/LBH feedback loop contributes to the malignant phenotype of GSCs via TNF-α-mediated NF-κB signaling pathway. Cancer Cell Int. 2024;24(1):365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Xing Y, Larson K, Li J, et al. Canonical and non-canonical functions of STAT in germline stem cell maintenance. Dev Dyn. 2023;252(6):728–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chang L, Zhang Y, Li M, et al. Nanostructured lipid carrier co-delivering paclitaxel and doxorubicin restrains the proliferation and promotes apoptosis of glioma stem cells via regulating PI3K/Akt/mTOR signaling. Nanotechnology. 2021;32(22):225101. [DOI] [PubMed] [Google Scholar]
- 100.Behrooz AB, Syahir A. Could we address the interplay between CD133, Wnt/β-catenin, and TERT signaling pathways as a potential target for glioblastoma therapy? Front Oncol. 2021;11: 642719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yin J, Seo Y, Rhim J, et al. Cross-talk between PARN and EGFR-STAT3 signaling facilitates self-renewal and proliferation of glioblastoma stem cells. Cancer Res. 2023;83(22):3693–709. [DOI] [PubMed] [Google Scholar]
- 102.Velpula KK, Rehman AA, Chelluboina B, et al. Glioma stem cell invasion through regulation of the interconnected ERK, integrin α6 and N-cadherin signaling pathway. Cell Signal. 2012;24(11):2076–84. [DOI] [PubMed] [Google Scholar]
- 103.Zhang C, Martinez-Ledesma E, Gao F, et al. Wild-type TP53 defined gamma-secretase inhibitor sensitivity and synergistic activity with doxorubicin in GSCs. Am J Cancer Res. 2019;9(8):1734–45. [PMC free article] [PubMed] [Google Scholar]
- 104.Jiang Y, Wang Z, Ying C, et al. FMR1/circCHAF1A/miR-211-5p/HOXC8 feedback loop regulates proliferation and tumorigenesis via MDM2-dependent p53 signaling in GSCs. Oncogene. 2021;40(24):4094–110. [DOI] [PubMed] [Google Scholar]
- 105.Smith-Cohn MA, Burley NB, Grossman SA. Transient opening of the blood-brain barrier by vasoactive peptides to increase CNS drug delivery: reality versus wishful thinking? Curr Neuropharmacol. 2022;20(7):1383–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Thombre R, Mess G, Kempski Leadingham KM, et al. Towards standardization of the parameters for opening the blood-brain barrier with focused ultrasound to treat glioblastoma multiforme: a systematic review of the devices, animal models, and therapeutic compounds used in rodent tumor models. Front Oncol. 2022;12:1072780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Ouyang J, Jiang Y, Deng C, et al. Doxorubicin delivered via ApoE-directed reduction-sensitive polymersomes potently inhibit orthotopic human glioblastoma xenografts in nude mice. Int J Nanomed. 2021;16:4105–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Todd N, Angolano C, Ferran C, et al. Secondary effects on brain physiology caused by focused ultrasound-mediated disruption of the blood-brain barrier. J Control Release. 2020;324:450–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Ji R, Karakatsani ME, Burgess M, et al. Cavitation-modulated inflammatory response following focused ultrasound blood-brain barrier opening. J Control Release. 2021;337:458–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Huang Y, Zhang B, Xie S, et al. Superparamagnetic iron oxide nanoparticles modified with tween 80 pass through the intact blood-brain barrier in rats under magnetic field. ACS Appl Mater Interfaces. 2016;8(18):11336–41. [DOI] [PubMed] [Google Scholar]
- 111.Wu Y, Wan S, Yang S, et al. Macrophage cell membrane-based nanoparticles: a new promising biomimetic platform for targeted delivery and treatment. J Nanobiotechnology. 2022;20(1):542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Duan Y, Zhou J, Zhou Z, et al. Extending the in vivo residence time of macrophage membrane-coated nanoparticles through genetic modification. Small. 2023;19(52): e2305551. [DOI] [PubMed] [Google Scholar]
- 113.Joshi BS, Zuhorn IS. Heparan sulfate proteoglycan-mediated dynamin-dependent transport of neural stem cell exosomes in an in vitro blood-brain barrier model. Eur J Neurosci. 2021;53(3):706–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Liu S, Liu J, Li H, et al. An optimized ionizable cationic lipid for brain tumor-targeted siRNA delivery and glioblastoma immunotherapy. Biomaterials. 2022;287: 121645. [DOI] [PubMed] [Google Scholar]
- 115.Jin M, Jin G, Kang L, et al. Smart polymeric nanoparticles with pH-responsive and PEG-detachable properties for co-delivering paclitaxel and survivin siRNA to enhance antitumor outcomes. Int J Nanomedicine. 2018;13:2405–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Li X, Xing L, Hu Y, et al. An RGD-modified hollow silica@Au core/shell nanoplatform for tumor combination therapy. Acta Biomater. 2017;62:273–83. [DOI] [PubMed] [Google Scholar]
- 117.Nikanjam M, Gibbs AR, Hunt CA, et al. Synthetic nano-LDL with paclitaxel oleate as a targeted drug delivery vehicle for glioblastoma multiforme. J Control Release. 2007;124(3):163–71. [DOI] [PubMed] [Google Scholar]
- 118.McMahon D, Poon C, Hynynen K. Evaluating the safety profile of focused ultrasound and microbubble-mediated treatments to increase blood-brain barrier permeability. Expert Opin Drug Deliv. 2019;16(2):129–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lundy DJ, Nguyễn H, Hsieh PCH. Emerging nano-carrier strategies for brain tumor drug delivery and considerations for clinical translation. Pharmaceutics. 2021;13(8):1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Yang Q, Parker CL, McCallen JD, et al. Addressing challenges of heterogeneous tumor treatment through bispecific protein-mediated pretargeted drug delivery. J Control Release. 2015;220(Pt B):715–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Zhang S, Zhang S, Luo S, et al. Ultrasound-assisted brain delivery of nanomedicines for brain tumor therapy: advance and prospect. J Nanobiotechnology. 2022;20(1):287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Huang ETC, Yang JS, Liao KYK, et al. Predicting blood-brain barrier permeability of molecules with a large language model and machine learning. Sci Rep. 2024;14(1):15844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Golán I, Rodríguez de la Fuente L, Costoya JA. NK cell-based glioblastoma immunotherapy. Cancers (Basel). 2018;10(12):5224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.de Bacco F, D’Ambrosio A, Casanova E, et al. MET inhibition overcomes radiation resistance of glioblastoma stem-like cells. EMBO Mol Med. 2016;8(5):550–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Shoae-Hassani A, Hamidieh AA, Behfar M, et al. NK cell-derived exosomes from NK cells previously exposed to neuroblastoma cells augment the antitumor activity of cytokine-activated NK cells. J Immunother. 2017;40(7):265–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Morgan RA, Johnson LA, Davis JL, et al. Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther. 2012;23(10):1043–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Jiang H, Gao H, Kong J, et al. Selective targeting of glioblastoma with EGFRvIII/EGFR bitargeted chimeric antigen receptor T Cell. Cancer Immunol Res. 2018;6(11):1314–26. [DOI] [PubMed] [Google Scholar]
- 128.Leland P, Degheidy H, Lea A, et al. Identification and characterisation of novel CAR-T cells to target IL13Rα2 positive human glioma in vitro and in vivo. Clin Transl Med. 2024;14(5): e1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Nakagawa T, Kijima N, Hasegawa K, et al. Identification of glioblastoma-specific antigens expressed in patient-derived tumor cells as candidate targets for chimeric antigen receptor T cell therapy. Neurooncol Adv. 2023;5(1):vdac177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Li Y, Wu H, Chen G, et al. Arming anti-EGFRvIII CAR-T with TGFβ trap improves antitumor efficacy in glioma mouse models. Front Oncol. 2020;10:1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Yang F, Zhang D, Jiang H, et al. Small-molecule toosendanin reverses macrophage-mediated immunosuppression to overcome glioblastoma resistance to immunotherapy. Sci Transl Med. 2023;15(683):eabq3558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Zilberman S, Schenowitz C, Agaugué S, et al. HLA-G1 and HLA-G5 active dimers are present in malignant cells and effusions: the influence of the tumor microenvironment. Eur J Immunol. 2012;42(6):1599–608. [DOI] [PubMed] [Google Scholar]
- 133.Schmitt J, Eckardt S, Schlegel PG, et al. Human parthenogenetic embryonic stem cell-derived neural stem cells express HLA-G and show unique resistance to NK cell-mediated killing. Mol Med. 2015;21(1):185–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Terszowski G, Klein C, Stern M. KIR/HLA interactions negatively affect rituximab- but not GA101 (obinutuzumab)-induced antibody-dependent cellular cytotoxicity. J Immunol. 2014;192(12):5618–24. [DOI] [PubMed] [Google Scholar]
- 135.Camacho-Bydume C, Wang T, Sees JA, et al. Specific class I HLA supertypes but Not HLA zygosity or expression are associated with outcomes following HLA-matched allogeneic hematopoietic cell transplant: HLA supertypes impact allogeneic HCT outcomes. Transp Cell Ther. 2021;27(2):142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Qiu R, Zhong Y, Li Q, et al. Metabolic remodeling in glioma immune microenvironment: intercellular interactions distinct from peripheral tumors. Front Cell Dev Biol. 2021;9: 693215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Gong L, Ji L, Xu D, et al. TGF-β links glycolysis and immunosuppression in glioblastoma. Histol Histopathol. 2021;36(11):1111–24. [DOI] [PubMed] [Google Scholar]
- 138.Liu Y, Li X, Zhang Y, et al. An miR-340-5p-macrophage feedback loop modulates the progression and tumor microenvironment of glioblastoma multiforme. Oncogene. 2019;38(49):7399–415. [DOI] [PubMed] [Google Scholar]
- 139.Zhou J, Zhang A, Fan L. HSPA12B secreted by tumor-associated endothelial cells might induce M2 polarization of macrophages via activating PI3K/Akt/mTOR signaling. Onco Targets Ther. 2020;13:9103–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Tuineau MN, Herbert LM, Garcia SM, et al. Enhanced glycolysis causes extracellular acidification and activates acid-sensing ion channel 1a in hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2024;327(4):L439–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Mani NL, Weinberg SE, Chaudhuri S, et al. Acidity induces durable enhancement of T(reg) cell suppressive functions for tumor immune evasion. Mol Immunol. 2024;174:57–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Tsukihara S, Akiyama Y, Shimada S, et al. Delactylase effects of SIRT1 on a positive feedback loop involving the H19-glycolysis-histone lactylation in gastric cancer. Oncogene. 2024;44(11):724–38. [DOI] [PubMed] [Google Scholar]
- 143.Wu S, Zhang H, Gao C, et al. Hyperglycemia enhances immunosuppression and aerobic glycolysis of pancreatic cancer through upregulating Bmi1-UPF1-HK2 pathway. Cell Mol Gastroenterol Hepatol. 2022;14(5):1146–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Shibuya K, Okada M, Suzuki S, et al. Targeting the facilitative glucose transporter GLUT1 inhibits the self-renewal and tumor-initiating capacity of cancer stem cells. Oncotarget. 2015;6(2):651–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Caillot M, Bourgeais J, Dakik H, et al. Cyclin D1 targets hexokinase 2 to control aerobic glycolysis in myeloma cells. Oncogenesis. 2020;9(7):68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zheng M, Wu C, Yang K, et al. Novel selective hexokinase 2 inhibitor Benitrobenrazide blocks cancer cells growth by targeting glycolysis. Pharmacol Res. 2021;164: 105367. [DOI] [PubMed] [Google Scholar]
- 147.Wolf A, Agnihotri S, Micallef J, et al. Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J Exp Med. 2011;208(2):313–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Yuan J, Peng G, Xiao G, et al. Xanthohumol suppresses glioblastoma via modulation of Hexokinase 2 -mediated glycolysis. J Cancer. 2020;11(14):4047–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Flavahan WA, Wu Q, Hitomi M, et al. Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat Neurosci. 2013;16(10):1373–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Ye C, Li H, Li Y, et al. Hypoxia-induced HMGB1 promotes glioma stem cells self-renewal and tumorigenicity via RAGE. Iscience. 2022;25(9):104872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Obara EAA, Aguilar-Morante D, Rasmussen RD, et al. SPT6-driven error-free DNA repair safeguards genomic stability of glioblastoma cancer stem-like cells. Nat Commun. 2020;11(1):4709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Ertas YN, Abedi Dorcheh K, Akbari A, et al. Nanoparticles for targeted drug delivery to cancer stem cells: a review of recent advances. Nanomaterials (Basel). 2021;11(7):1755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Glumac PM, Lebeau AM. The role of CD133 in cancer: a concise review. Clin Transl Med. 2018;7(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Liu G, Yuan X, Zeng Z, et al. Analysis of gene expression and chemoresistance of CD133+ cancer stem cells in glioblastoma. Mol Cancer. 2006;5:67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Joo KM, Kim SY, Jin X, et al. Clinical and biological implications of CD133-positive and CD133-negative cells in glioblastomas. Lab Invest. 2008;88(8):808–15. [DOI] [PubMed] [Google Scholar]
- 156.Guo Y, Liu S, Wang P, et al. Expression profile of embryonic stem cell-associated genes Oct4, Sox2 and Nanog in human gliomas. Histopathology. 2011;59(4):763–75. [DOI] [PubMed] [Google Scholar]
- 157.Son MJ, Woolard K, Nam DH, et al. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell Stem Cell. 2009;4(5):440–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Chen X, Niu W, Fan X, et al. Oct4A palmitoylation modulates tumorigenicity and stemness in human glioblastoma cells. Neuro Oncol. 2023;25(1):82–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Mahlokozera T, Patel B, Chen H, et al. Competitive binding of E3 ligases TRIM26 and WWP2 controls SOX2 in glioblastoma. Nat Commun. 2021;12(1):6321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Fang X, Huang Z, Zhai K, et al. Inhibiting DNA-PK induces glioma stem cell differentiation and sensitizes glioblastoma to radiation in mice. Sci Transl Med. 2021;13(600):eabc7275. [DOI] [PubMed] [Google Scholar]
- 161.Santaliz-Ruiz LE IV, Xie X, Old M, et al. Emerging role of nanog in tumorigenesis and cancer stem cells. Int J Cancer. 2014;135(12):2741–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Inoue A, Ohnishi T, Nishikawa M, et al. A narrative review on CD44’s role in glioblastoma invasion, proliferation, and tumor recurrence. Cancers (Basel). 2023;15(19):4898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Du Z, Wang Y, Liang J, et al. Association of glioma CD44 expression with glial dynamics in the tumour microenvironment and patient prognosis. Comput Struct Biotechnol J. 2022;20:5203–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Bernal A, Arranz L. Nestin-expressing progenitor cells: function, identity and therapeutic implications. Cell Mol Life Sci. 2018;75(12):2177–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Neradil J, Veselska R. Nestin as a marker of cancer stem cells. Cancer Sci. 2015;106(7):803–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Not applicable.