Skip to main content

REVIEW article

Front. Oncol., 02 March 2022
Sec. Neuro-Oncology and Neurosurgical Oncology
This article is part of the Research Topic Adult Diffuse Glioma Microenvironment: Insight into Cancer Ecology View all 10 articles

Glioblastoma Microenvironment: From an Inviolable Defense to a Therapeutic Chance

Vincenzo Di Nunno&#x;Vincenzo Di Nunno1†Enrico Franceschi*&#x;Enrico Franceschi2*†Alicia TosoniAlicia Tosoni2Lidia GattoLidia Gatto1Stefania BartoliniStefania Bartolini2Alba Ariela Brandes&#x;Alba Ariela Brandes2†
  • 1Department of Oncology, AUSL Bologna, Bologna, Italy
  • 2 Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy

Glioblastoma is an aggressive tumor and is associated with a dismal prognosis. The availability of few active treatments as well as the inexorable recurrence after surgery are important hallmarks of the disease. The biological behavior of glioblastoma tumor cells reveals a very complex pattern of genomic alterations and is partially responsible for the clinical aggressiveness of this tumor. It has been observed that glioblastoma cells can recruit, manipulate and use other cells including neurons, glial cells, immune cells, and endothelial/stromal cells. The final result of this process is a very tangled net of interactions promoting glioblastoma growth and progression. Nonetheless, recent data are suggesting that the microenvironment can also be a niche in which glioblastoma cells can differentiate into glial cells losing their tumoral phenotype. Here we summarize the known interactions between micro-environment and glioblastoma cells highlighting possible therapeutic implications.

Introduction

The 2021 World Health Organization (WHO) Classification defines glioblastoma (GBM) as a diffuse astrocytic glioma without IDH (Isocitrate dehydrogenase) and H3R gene mutations, with enhanced microvascular proliferation, necrosis, and specific alterations like gain of chromosome 7/chromosome 10 loss, EGFR (Epidermal Growth Factor Receptor) amplification, and/or TERT (Telomerase Reverse Transcriptase) mutations (1, 2).

GBM is one of the most fatal primary central nervous system (CNS) malignancies with an estimated 5-years overall survival (OS) of only 6.8% (3, 4). In particular, the prognosis of patients with newly diagnosed GBM ranged from 12 to 18 months (5) while the recurrent disease is associated with a very poor outcome with an estimated OS of 5-10 months (6).

Several efforts have been spent to improve clinical outcomes of patients with GBM, nonetheless, none of the investigated agents have replaced the standard of care represented by maximal safe surgery followed by chemoradiation and adjuvant temozolomide, which has been adopted in 2005 (4). Even in the management of recurrent GBM, few systemic compounds demonstrated clinical efficacy (6).

There are several reasons behind these failures. First, the enrollment of GBM patients within clinical trials is limited only to10% of all GBM patients (7, 8).

However, the major cause explaining the difficult development of effective agents is certainly the complex biology of the disease. Indeed, GBM is associated with specific features which can be summarized in 1) the extremely high heterogeneity presented by tumor cells requiring combined treatment for different subtypes of GBM cancer cells; 2) the lack of biological models able to replicate or at least estimate the interaction between human tumor cells and the surrounding tissue; 3) the complex microenvironment surrounding the tumor (912). Furthermore, the presence of glioblastoma cancer stem cells (CSCs) has been employed to explain the impressive recurrence rate and regenerative proprieties of this tumor (13).

Of note, it has been demonstrated that GBM cells can manipulate the microenvironment surrounding themselves developing a niche sustaining tumor growth and development (14). This process involves immune cells, astrocytes, glial cells, neurons, extra-cellular matrix (ECM), vascular cells, and other cell types (1520). There are several ways by which GBM can communicate with the surrounding tissue. The secretion of soluble factors able to modulate genomic expression and biological behaviors of tumor-associated cells is one of the most obvious and described systems.

Of interest, GBM cells can develop a nuclear and cytoplasmatic ‘‘continuum’’ with neighboring cells. These nanotubes mediate the transfer of protein and inorganic elements (21). Moreover, this also non-secretable molecules such as RNAs, DNA and also mitochondria, and nuclei can be transported from the tumoral to a surrounding cell by these nano-tubules (2224). Notably, this system is also probably responsible for acquired resistance to radiation and systemic temozolomide (TMZ) (23).

GBM cells can also promote the creation of gap junction and cytoplasmatic connections (1520). Finally, the secretion of microvesicles, extracellular vesicles, and exosomes can promote communication with also very distant cells or tissues (1520).

The interactions between tumor cells and immune cells have acquired an increased interest due to the availability of agents able to promote immune-system reactivation. Nonetheless, other interactions have been only partially investigated and could hide novel promising targets for tumor treatment. Here we performed a review summarizing the identified interactions between GBM and its associated microenvironment. We also focused our attention on possible novel treatments targeting these complex interactions.

Immune Cells and Glioblastoma

Immune-contexture assumes a very important role within the GBM microenvironment. It can stimulate the progression and development of tumor cells (25).

In contrast to what was supposed in the past, it has been largely demonstrated that glioblastoma is not an immune ‘‘cold tumor’’ (26, 27). Similar to other tissue, the CNS has its resident immune tissue represented mainly from microglia (28). Furthermore, like other solid tumors, GBM can activate and recall migrating immune cells from systemic tissues and lymphatic vessels (28). Indeed, there is a connection between deep cervical nodes, dural sinus, and lymphatic vessels allowing systemic immune cells to move into the CNS reaching the target site (29, 30). CNS is regulated by several molecular mechanisms able to enhance or suppress the immune response. However, the main difference with other peripheral non-CNS tissues is that the balance between inhibitory and stimulating mechanisms is biased in favor of immune suppression (3135). One of the most important factors mediating an immune-inhibition is the TGFβ2 (transforming growth factor β2) (36, 37). This factor can mediate inhibition of Interleukin 2 mediated T cell survival and reduce the production of critical effector proteins by lymphocytes and other immune cells (36, 37). An increased intracranial pressure, such as that observed during an inflammation response, could be catastrophic and associated with irreparable damage to the neurological tissues. Thus, CNS protects itself preventing prolonged immune response with activation of several mechanisms supporting an immune-depressive status (3135).

Recently, two transcription factors showed to mediate several immune-depressive effects in GBM (38). These factors are represented by SRY-Box Transcription Factor 2 (Sox2) and octamer-binding transcription factor 4 (Oct4) whose activation promotes the suppression of both innate and adaptive immune responses maintaining glioma cell stemness and tumor-propagating potential (38). In particular, the co-expression of Oct4/Sox2 inhibits the expression of CCL5 (C-C Motif Chemokine Ligand 5), CXCL9 (C-X-C Motif Chemokine Ligand 9), CXCL10, and CXCL11 which are essential to induce lymphocyte CD8 effector (Th1 response) attraction (38). Furthermore, they promote the secretion of SPP (signal peptide peptidase), IL8 (Interleukin 8), CXCL5, CCL20, IL6 (interleukin 6) inducing Treg (immune-inhibitory) response and shifting macrophage differentiation toward an immune-regulatory profile more than an immune-active one (38). Several immune checkpoints such as PD-L1 (Programmed Death Receptor Ligand 1), CD70 (Cluster of differentiation 70), A2aR (adenosine A2A receptor), and TDO (Tryptophan 2,3-dioxygenase) are also overexpressed in cells with Sox2/Oct4 overexpression (38). The effects promoted by Sox2/Oct4 are directly mediated by overexpression of the BRD3 (Bromodomain containing protein 3) and BRD4 (Bromodomain containing protein 4) proteins which belong to the Bromodomain and extra terminal motif (BET) proteins family. The BRD3 and BRD4 proteins act modulating the activity of the histone 3 (H3) acetylation mediated by the H3K27Ac enzyme. This mechanism appears of particular interest considering the availability of pan BET inhibitors.

Another recent research investigated RNA expression of GBM-derived sphere-line treated with the BET inhibitor JQ1. The inhibition of BET resulted in a significant modulation of genes responding to Interferon-alpha (enhanced in about 50% of GBM) through a direct transcriptional inhibition more than interference to the JAK (Janus Kinase) -STAT (Signal transducer and activator of transcription) pathway (39).

BET inhibitors have recently been assessed on phase I clinical studies and further trials assessing their clinical efficacy are needed (4047).

In conclusion, differently from other tissues, CNS physiologically employs signals able to reduce an immune innate and adaptive response. This happened to prevent catastrophic effects associated with an uncontrolled inflammation in a dedicated system such as the brain or the spinal cord. In this context, GBM cells adopt several mechanisms to reinforce this inhibition. Glioblastoma stem cells (GSCs) are probably an important component of this complex mechanism as they express important transcription factors such as Sox2 and Oct4 (38). Nonetheless, it is also likely that other pathways converge on the same inhibition suggesting that the inhibition of a singular cascade could not be associated with an effective immune response reactivation.

GBM – Microglia, Myeloid Cells, and Macrophages

Immune resident cells of the CNS are microglia and macrophages including perivascular, meningeal, choroid plexus, and circumventricular macrophages (28). These cells cover over 50% of the GBM tumor load and their composition change during the time and tumor progression (48). In the early phases of GBM development, the microglia is the most represented infiltrating cell subtype while macrophages and myeloid cells composed a large part of GBM volume in advanced phases (49, 50). Differently from macrophages, microglia constitute the resident immune system of the CNS. These cells can move within the CNS but they do not circulate in other tissues. Both microglia and macrophages are ineffective against tumors as their immune response is suppressed by the presence of an immune-depressive cytokines storm (11, 51). Indeed, GBM cells can directly mediate the production of several immune-depressive cytokines. Furthermore, cancer cells can manipulate the secretion and phenotype of surrounding immune cells shifting their phenotype toward an immune-suppressive one and initiating positive feedback leading to an immune-suppressive contexture. Notably, macrophages and myeloid cells are important protagonists of this process.

Myeloid cells constitute a large part of the immune-contexture of GBM. These cells are recruited directly from tumors cells and then can differentiate toward macrophages and monocyte phenotypes (52).

Myeloid cells have been classified into monocytic and granulocytic subtypes. Both these cells inhibit T cell and NK activities. Curiously, some data seem to confirm a negative prognostic role of these cells in males while these same cells can positively activate an immune response in female patients (52).

The main stimulation for macrophages and microglia accrual around the tumor is mediated by the same glioblastoma cells through the production of CCL2 (C-C Motif Chemokine Ligand 2), CCL7 (C-C Motif Chemokine Ligand 7), GDNF (Glial Cell-Derived Neurotrophic Factor), SDF1 (stromal cell-derived factor 1), TNF (tumor necrosis factor), VEGF (vascular endothelial growth factor), ATP (adenosine triphosphate), CSF-1 (Colony-stimulating factor 1), GM-CSF (Granulocyte-Macrophage Colony-Stimulating Factor), and expression of OLIG2 (Oligodendrocyte transcription factor 2) (48, 51, 53). Macrophages and microglia can perpetuate themselves accrual through the production of CCL2 resulting in a positive feedback loop (53). In advanced phases, microglia are localized mainly around tumor tissue while macrophages are localized in perivascular regions (54).

The differentiation of macrophages ranges from two specific phenotypes which are: M1 (immune-response enhancer) or M2 (immune-regulator inhibiting immune response) (55, 56). Although explicative, this classification is not definitive as macrophages can reach an intermediate grade of differentiation activating both genes of the M1 and M2 subtypes (57). In GBM, the resulting phenotype can inhibit immune response through secretion of transforming growth factor β1 (TGFβ1), arginase 1 (ARG1), or interleukin 10 (IL-10) enhancing neo-angiogenesis through VEGF production and extracellular matrix modeling by metalloproteases (MP) (38). On the other hand, these macrophages also produce pro-inflammatory molecules such as IL-β1 (interleukin β1), TNF, IL-5 (interleukin 5), and IL-12 (interleukin 12) which are molecules stimulating the immune response (38).

It has been demonstrated that macrophages promote GBM growth and progression in different ways (11).

First, the macrophage tumor suppression phenotype inhibits the response of other immune cells surrounding the tumor. This immune inhibition is mainly explicated by myeloid cells and differentiated macrophages which miss the activation of natural killer (NK) cells, the production of interferon γ (INF γ) and TGFβ (55). These same cells can induce lymphocytes differentiation toward an immune-regulatory (Th2) profile by TGFβ, reactive oxygen species (ROS), cysteine depletion, L-selectin downregulation, ARG1, and inducible nitric oxide synthase (iNOS2) production (36, 37).

The remodeling of ECM is essential for tumor migration and is mediated by the production of inactive metalloproteases enzymes by GBM cells (48, 58). These pro-Metalloproteases are then activated by enzymes produced by microglia. Moreover, the production of CSF1 by glioblastoma induces the release of the insulin-like growth factor-binding protein 1 (IGFBP1) by microglia which Is essential to promote vascularization and angiogenesis (48, 58, 59).

The administration of macrophages able to restore immune response would be a promising strategy able to partially restore immune-response against the tumor. The development of engineered macrophages (car-M) is at an early stage of assessment but it could be a promising strategy for GBM treatment (60).

Similar to macrophages and microglia also neutrophils and other innate immune cells are attracted by the tumor. In particular, inflammatory factors secreted after a surgical intervention such as IL-8, TNF, and CCL2 can promote neutrophil infiltration in addition to SDF1 and plasminogen activator inhibitor 1 secreted by tumor cells (61, 62). These innate immune cells can facilitate further accrual of macrophages and contribute to microenvironment remodeling.

Lymphocytes and Antigen-Presenting Cells

Antigens exposure can start an adaptive immune response in the CNS like observed in other organs.

The presence of lymphocytes surrounding the tumor has been largely reported in GBM where their concentration correlates positively with survival (63, 64).

When an antigen is recognized by the immune system, a specific lymphocyte clone targeting the same antigen expands itself starting an adaptive immune response. The antigen-presenting cells (APCs) are essential to start the clonal expansion as these cells mediate the presentation of the antigen to the lymphocytes initiating the immune response (29, 30). Antigens captured within the CNS are processed by APC and then presented to lymphocytes probably in deep cervical nodes. Activated lymphocytes can move into the brain directly thanks to the increased permeability resulting from inflammation and neo-vessels or after exposure to antigen by APCs on the meningeal surface (29, 30).

Even if lymphocytes can move around GBM mass their ability to start an immune response against tumor is strongly inhibited by several factors. Once lymphocytes come to the peritumoral tissue they are invested by strong immune-depressive signaling mediated tumor cells and GBM associated microenvironment. The TGFβ1 and TGFβ2 molecules are the main characters for this inhibition (11, 37, 65).

It is important to observe that immune-inhibitory signals are largely provided by the microenvironment more than directly by GBM cells. This is a concrete example of how GBM cells manipulate the microenvironment to sustain their growth and expansion (11). Notably, it has been reported that GBM cells can induce pericytes to produce TGFβ and IL-10 (an inhibitory interleukin) (66).

GBM cells can also release directly inhibitory signals such Fas antigen ligand (FASLG), and other inhibitory molecules. Lymphocytes assume the classical exhaustion phenotype expressing several-inhibitory receptors including the PD-1, T cell membrane protein 3 (TIM3), lymphocyte activation gene 3 protein (LAG3), and T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT) (67).

As already specified, it is interesting to observe that a large part of inhibitory molecules released by GBM cells can be mediated by the Sox2/Oct4 (38).

The complex network of immune-inhibitory signals can partially explain the failure of clinical trials exploring immune-checkpoint inhibitors (ICIs). To date, three different phases III trials failed to show a clinical benefit from the addition of nivolumab in newly diagnosed and recurrent GBM (6870).

In the last years, a novel immune strategy employing engineered T-cells (car-T) has been investigated with favorable preliminary results. In GBM the introduction of reprogrammed T-cells could be ineffective due to the immune shield provided by the tumor microenvironment (70). Strategies aimed to switch the composition of cancer-associated immune cells from an immune-suppressive to an immune-active one are of critical importance. Combinatory strategies employing more immune targets are under investigation (70).

Neurons and Glioblastoma

The brain is composed of over 60% of the white matter which is largely composed of myelinic axons. Neurons, astrocytes, and oligodendrocytes with other glial cells (including microglia) are the most represented cells in the brain and CNS (71).

The study of the interactions between these components and glioblastoma appears of extreme interest as these are strictly related to some of the most important biological and clinical features of GBM cells.

Indeed, connexons between white matter, neurons, and tumors can partially explain the high recurrence rate of the disease as well as the ability to relapse also in a distant site such as the contralateral hemisphere (7174). Of interest, the interaction between neurons and GBCs assumes a particular interest in recent years.

Indeed, the biological niche composed of GBM cells, GSCs, peritumoral oligodendrocytes, astrocytes, and also neuronal axons can both drive differentiation of a GSCs cell toward a tumoral phenotype or, surprisingly, toward a differentiated non-tumoral cell subtype (75). On the other hand, also GBM cells can induce the dedifferentiation of astrocytic cells while astrocytes and oligodendrocytes can support tumor growth in different ways. In particular, astrocytes can support tumor growth and development through the secretion of several cytokines and other soluble factors stimulating directly GBM growth and maintaining an immune-suppressive contexture (76, 77).

Astrocytes

Tumor-associated astrocytes (TAAs) can contribute to GBM growth in different ways. During early phases of tumor development, TAAs respond to initial injury with the secretion of TGFβ, IL-6, and Insulin growth factor 1 (IGF-1) which could contribute to GBM sustainment (76, 77). These same TAAs express the sonic hedgehog (SHH) gene and are concentrated in the perivascular niche of GBM (7880). In early and advanced phases communications between GBM and TAAs occurred especially through the secretion of extracellular vesicles which contribute to stimulating several growth factors by TAAs (81). These include the vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), and epidermal growth factor (EGF) (81).

The contribution of TAAs is of key importance as these cells can stimulate neo-angiogenesis through secretion of VEGF and hypoxia-inducible factor -1 (HIF-1) (82, 83).

Of interest, it has been demonstrated that astrocytes actively participate in chemotherapy resistance by GBM (84). The exact mechanism by which this is possible is still unclear but in vitro studies identified that chemoprotection performed by TAAs is possible only when these cells are connected to cancer cells by gap junctions. Indeed, apoptosis and GBM cells death increased when the development of gap junctions was inhibited by the administration of carbenoxolone (a gap junctions communication inhibitor) (84).

One of the most surprising behaviors of astrocytes is the capacity to be transformed in tumor-initiating cells. In vivo studies demonstrated that several oncogenes including MYC, RAS, and EGFR variant III can dedifferentiate astrocytes in tumoral cells (85, 86). This close relationship suggests a common phenotype shared by GBM and astrocyte supporting the hypothesis that astrocytes are the precursor normal cells by which gliomas originate. It is still unknown the exact mechanism, but it has been observed that glioma cells inhibit the expression of the onco-suppressor p53 in normal astrocytes (87, 88). The malignant transformation of astrocytes required the miR-10b which is silenced in TAAs but is largely expressed and secreted by GBM cells using extracellular vesicles (89).

Although the already cited oncogene (MYC, RAS, and EGFR) can mediate the transformation of astrocyte into malignant glioma cells it is still unclear the exact mechanism by which GBM cells can initiate this process in humans (85, 86). However, the manipulation and conversion of surrounding astrocytes into neoplastic cells can be another issue explaining the well-known recurrence and regeneration proprieties of this tumor

Neurons

Neurons can be manipulated by glioma cells to stimulate tumor progression and invasion. One of the first documented interactions between neurons and GBM is associated with the release of glutamate which could be enhanced in patients with GBM resulting in seizure onset and increased vascular permeability mediated by the N-methyl-D-aspartate receptors (NMDA) (90, 91). Of interest, synapsing signaling of cortical neurons can directly stimulate GBM growth. Neurons and oligodendrocyte precursor cells can secrete the postsynaptic adhesion molecule neuroligin 3 which mediated the activation of several key pathways including the mammalian target of rapamycin (mTOR), phosphoinositide-3-kinase (PI3K), and focal adhesion kinase (FAK) (92). Also, the brain-derived neurotrophic factor chaperone Bip (HSPA5) has been identified as an agent able to stimulate mitosis and cell division (92).

Oligodendrocytes and White Matter

Oligodendrocytes may absolve an inhibitory function against GBM cells. Indeed, these cells can activate the WNT inhibitory factor 1 leading to the inhibition of GBM growth and proliferation (93, 94). In CNS, Oligodendrocytes modulate neural plasticity, metabolic support, and axon activity through myelination. It is well known that GBM can spread through myelinic axons (75). The butterfly shape commonly observed in GBM indicates that tumor cells can invade the contralateral side following the commissure fibers of the corpus callosum (75). Similarly, GBM can spread through the arcuate fasciculus and using the radiation of the corpus callosum (75). It has been supposed that GBM cells adopted myelinated axons as scaffolds and tracks however a recently published study could drastically modify this concept (75). Glioblastoma stem cells are contained in a specific biological niche composed of their microenvironment which influences GSCs fate and differentiation. It is easy to think that GSCs differentiate into cancer cells but it is not always true as in some conditions these cells can differentiate into normal cells without tumorigenicity. It seems that a biological niche composed of white matter can induce GSCs to differentiate toward a pre-oligodendrocyte phenotype losing oncogenic potential (75). This mechanism is similar to that happening during the response to injury (75). Thus, this is an important discovery demonstrating that GSCs can differentiate toward a normal cell phenotype.

The mechanism by which this happens involves the white matter destruction which leads to upregulation of the SOX10 transcription factor. The SOX10 mediates oligodendrocyte differentiation also by cancer cells (75).

Further investigation of this pathway will probably provide new therapeutic insights for the clinical management of GBM.

Cancer Stem Cells

Glioblastoma stem cells explain the extreme clinical aggressiveness of GBM. In GSCs are associated with the high recurrence rate of the tumor even after complete resection (72). These cells are also associated with tumor renewal and resistance to treatment (73). Finally, the ability to spread and relapse in a distant site of the brain such as the contralateral hemisphere could be partially explained by GSCs an interaction between tumors and neurons (7174). As already discussed, the differentiation of GSCs could be shifted toward a tumoral or non-tumoral phenotype according to the biological niche in which these cells are located (75).

Glioblastoma stem cells are localized in a specific niche localized in the perivascular and hypoxic regions of the tumor. These cells are similar to normal neural stem cells (NSCs) which are localized mainly in the subventricular zone of the brain which is a common site of origin for glioblastoma (9597).

It is still unclear if GSCs derived from altered NSCs or mutated glioma cells (98). Glioblastoma Stem Cells are surely associated with GBM progression and recurrence after surgery (99, 100). The surface marker CD133 is one of the most adopted markers to recognize GSCs.

The expression of CD133 is regulated by Sox2 and agents targeting or interfering with this transcription factor can reduce tumor-initiating ability, resistance to chemotherapy, and recurrence (101). Nestin is another upregulated factor reported on GSCs that is directly correlated with poorer survival (98) (102).

Notably, the specific position of GSCs leads to an interaction with Endothelial cells (ECs) and pericytes. It has been supposed that GSCs directly respond to hypoxia stimulating vessels creation and neo-angiogenesis through the production of VEGF (103, 104). Moreover, GSCs can differentiate into ECs and pericytes (103105). The most surprising finding related to GSCs is that these cells can be located at a distance of up to 3 cm from the primary tumor (106). Moreover, their histological identification is almost impossible as these cells are indistinguishable from normal tissue cells (72). Nonetheless, some studies suggest that GSCs derived from the GBM core are different from those isolated by peritumoral tissue presenting different behaviors in terms of proliferative potential and expression of stem-cell markers (72, 107, 108). Even if peritumoral GSCs are less aggressive compared to GSCs from the core of GBM these same cells are also more resistant to temozolomide and radiation therapy (72, 107, 108). As discussed in a previous paragraph, the transcription factors Sox2 and Oct4 are activated in GSCs promoting glioma cell stemness and stimulating several mechanisms leading to innate and adaptive immune response inhibition (38). Curiously, recent data suggest a strong interaction between glioblastoma cancer cells and GSCs (109). This is mainly mediated by the cascade activated by the brain-derived neurotrophic factor (BDNF) secreted by GBM cells and the receptor neurotrophic receptor tyrosine kinase 2 (NTRK2) localized on stem cells. This interaction leads to a paracrine effect resulting in tumor growth and development (109).

Endothelial Cells

Neovascularization and new vessels development are both hallmarks of GBM. The fast growth of the tumor mass required high blood intake thus there are several identified pathways by which GBM cells can interact and manipulate ECs activity (110).

Hypoxia is one of the most important stimulations for tumor growth, vessels development, and acquisition of more aggressive pathological features by tumor cells (111113). In general, hypoxia leads to a metabolic switch by tumor cells which are more likely to promote aerobic glycolysis. Hypoxia induces also an attenuated expression of DNA repair enzymes and impedes the formation of Reactive Oxygen Species (ROS) reducing the cytotoxic effect of radiation therapy (111113).

Notably, ROS can assume a different biological role according to their concentration. Low levels of ROS are associated with stimulation of hormone secretion, synaptic plasticity, and immune response (114, 115). High levels of ROS are instead associated with DNA damage and in particular p53 damage (116). The DNA damage promoted by ROS could partially explain the switch from low to high-grade gliomas (117). The NADPH oxidase (NOX) 4 is activated by PDGF and TGF-β and is a key enzyme associated with ROS production (H2O2) (118). The inhibition of this enzyme could be a promising target in patients with GBM.

One of the most established pathways for new vessels development is the recruitment of EC progenitors by the bone marrow. This is a process occurring during embryogenesis or after an ischemic insult which is also adopted by GBM cells (119, 120). Tumor cells or microenvironments manipulated by GBM cells can secrete the SDF-1 which is the ligand of the CXCR4 receptor expressed by EC progenitors (121, 122). The interaction between SDF-1/CXCR-4 resulted in EC activation and recruitment of novel EC (121, 122).

It has been well established that GBM cells and the surrounding microenvironment can stimulate the production of the VEGF. This factor can drive EC toward the development of novel vessels in a process known as sprouting angiogenesis. Notably, the neoangiogenic promotion carried out by VEGF can be inhibited by the interaction between the Notch receptor and Delta-like canonical Notch ligand 4 (DLL4) (123). Curiously, novel vessels development can originate also through a process known as vasculogenic mimicry (124126). This phenomenon is mediated by the same tumor cells which can differentiate to create a vessels-like structure. In particular, macrophages surrounding tumors can mediate cyclooxygenase-2 (COX-2) activation stimulating vasculogenic mimicry (127). Other supposed mechanisms involved in this pathway are the expression of VE-Cadherin by GBM stem cells resulting from hypoxia and the HIF 1 and HIF 2 secretion. Also, the mTOR expression seems to be involved in this pathway (128).

Vascular co-option is another mechanism by which tumor cells move around pre-existing vessels gaining the access to oxygen and nutrients. Curiously GBM cells can induce secretion of bradykinin by EC cells. Bradykinin plays a chemotaxis effect on GBM cells (129, 130). The interaction between SDF1/CXCR4 (129, 130) and the expression of the EGFRvIII are also involved in this mechanism (129131).

A well-described mechanism associated with neo-angiogenesis is the interaction between VEGF and VEGFR2 and VEGFR1 (132). The interaction between VEGF and VEGFR1 or VEGFR2 induces the phosphorylation and activation of the ERK 1/2 (extracellular signal-regulated kinases 1 and 2) and p38 MAPK (mitogen-activated protein kinase) which together mediated transcription of pro-angiogenic factors (132).

This interaction also results in blood vessels permeability, and proteins lost from blood (110) explaining an increased permeability, edema onset, and increased intracranial pressure. This can explain the clinical benefit experienced by patients treated with the anti-VEGF agent bevacizumab (133136). Even if the administration of bevacizumab is associated with reduced edema and vascularization within tumor masses it failed to show a significant survival benefit among patients with GBM (133136).

The lack of the survival benefit observed could reflect the co-existence of several mechanisms resulting in angiogenesis and vessels development. In this optic, the inhibition of the VEGF mediated by bevacizumab can activate or reinforce other molecular pathways converging on angiogenesis promotion resulting in resistance to the anti-VEGF.

The study of the interactions between immune cells in tumor-associated microenvironment assumes an increased interest (137). Increasing data seem to indicate that hypoxia induces secretion of the VEGF which is one of the main molecules mediating this signaling. In particular Regulatory T lymphocytes (T reg) mediate the production of IL-10, IL-4, and IL-13 inducing differentiation of macrophages into M2 phenotype and stimulating expression of inhibitory B7-H receptor on their membranes (137).

Thus, the inhibition of VEGF could also result in an enhanced immune response against the tumor. This supposed synergic effect has led to the assessment of combination strategies in the clinical setting. Unfortunately, a phase II study assessing the combination between bevacizumab and pembrolizumab (a PD-1 inhibitor) failed to show a significant clinical efficacy on patients with GBM (138).

Targeting the Microenvironment in Glioblastoma

There are very few systemic agents showing clinical efficacy in patients with glioblastoma (139142). As already discussed in previous paragraphs, a great interest in the immune contexture of GBM is explained by the availability of active compounds able to restore immune response against tumors. Nonetheless, immune-checkpoint inhibitors failed to improve the survival of patients with GBM (68). The reason for this failure can be partially explained by the lack of uniformly expressed tumor-specific antigen due to the high heterogeneity of tumor GBM cells as well as the presence of an immune-depressive microenvironment which impairs the ability of immunotherapy to work. Also, GSCs can mediate immune escape (38).

Novel combination strategies are under investigation to overcome these limitations.

For example, it has been demonstrated that other immune checkpoints co-exist with PD-1. These are the indoleamine 2,3-dioxygenase (IDO1), T cell immunoglobulin-mucin-domain containing-3 (TIM-3), and lymphocyte activation gene 3 (LAG3) (143). Novel trials exploring combinations between PD-1 and LAG3 (NCT02658981) or IDO1 (NCT03707457) are under investigation. Similarly, the combination between temozolomide and IDO1 inhibitors is under investigation (NCT02052648).

Previous trials investigating neoantigen vaccination showed that vaccines can induce a significant increase of tumors infiltrating lymphocytes (144). Unfortunately, these infiltrating lymphocytes assume an exhausted phenotype. Thus the co-administration of multi-epitope vaccines and immune-checkpoint inhibitors could be another promising approach (NCT02149225 GAPVAC trial) (144). Strategies assessing co-administration of vaccines and CAR-T engineered with EGFRvIII and PD-1 are also under investigation (NCT04003649, NCT04201873, NCT02529072, NCT02287428) (145147). Also, the administration of co-stimulatory agonists able to enhance T cell function is under investigation. Agonists such as CD27, 4-1BB, OX40 or CD40 are under investigation (NCT04547777, NCT02658981, NCT03688178, NCT04440943) (145147).

Since macrophages are strongly associated with the development of an immune-depressive microenvironment the possibility to develop an engineered cell type of macrophage could be a promising strategy allowing a microenvironment to switch toward an immune-active phenotype (60). This Car-M strategy is still under early assessment however this appears a very promising approach.

As already specified SOX2 and Oct4 are two transcription factors able to modulate a very large amount of genes involved in the immune response. These two factors act through proteins belonging to the BET family. To date, BET inhibitors have been assessed on phase I trials with more of them showing a safety profile (3947). A further investigation of these agents could be important for patients with GBM.

The inhibition of the CSF1 receptor can inhibit the interaction between GBM and cells of the immune innate system including tumor-associated macrophages. Studies on murine models showed that macrophages develop resistance to CSF1 inhibition through the expression of the insulin growth factor 1 (148). The co-administration of CSF1 and IGF1 inhibitors could be an interesting approach as associated to restored immune-active microenvironment resulting from macrophages activation (148).

In the last years, several agents targeting angiogenesis have been tested in GBM without significant results. The only FDA-approved drug is bevacizumab which has been associated with prolonged progression-free survival, a reduction of symptoms related to the tumor but failed to show a significant impact on overall survival (133136). Several efforts are spent to understand the reason for bevacizumab resistance. The very high hypoxia level described inside the tumor can partially explain this failure. Indeed, hypoxia is associated with upregulation of the hypoxia-inducible protein 2 (HIG2) gene with downregulation of the CYLD gene expression. The overexpression of HIG2 resulted in HIF-1β, VEGF expression, and bevacizumab resistance through direct stimulation of hypoxia-inducible factor. Nonetheless, agents targeting HIG2 products failed to improve the clinical outcomes of patients with GBM (149). Novel approaches are aimed to target the angiotensin II receptors (AngII-R) and VEGF (150). However, this approach has been evaluated only in murine models thus assessments on humans are necessary to further assess this approach.

Since GBM cells adopt extracellular vesicles to communicate with surrounding tissue, a system able to reduce vesicles uptake and synthesis can be a promising target. Pre-clinical studies showed that inhibitors of neutral sphingomyelinase (GW4869) can reduce the production of extracellular vesicles while heparin and annexin A1 inhibitors can reduce vesicles intake by target cells (151153).

Direct inhibition of junctions between GBM and microenvironment is also a strategy under investigation. Inhibition of connexin 43 is essential to inhibit the interaction between GBM and astrocytes (154). Another interaction between oligodendrocytes, astrocytes, and GBM cells is the interaction between neuroligin 3/ADAM10 (155). Indeed, GBM cells produce neuroligin 3 which is cleaved from the ADAM10 secreted by neurons and oligodendrocytes (156). The effect is a stimulation of GBM growth and development (11). Thus inhibitors of the ADAM10 appear a promising strategy for GBM which should be further assessed among clinical trials in humans.

Conclusion

GBM remains a fatal disease with limited treatments. The reason for this failure could be partially explained by the development of a complex and effective net of interactions between tumor cells and surrounding tissue cells. The microenvironment resulting from the manipulation carried out by cancer cells can feed and stimulate GBM proliferation hiding and protecting tumor cells from systemic treatments.

Interactions between tumor, endothelial and immune cells are under careful assessment due to the availability of drugs targeting these pathways. Recently, also the associations between glioblastoma and astrocytes, oligodendrocytes, white matter, and neurons come out from the shadow offering novel promising targets with therapeutic implications.

Due to the presence of deep communications between tumor cells and the microenvironment the use of agents targeting more than one altered intracellular cascade at the same time appears a promising approach.

In this optic novel immune-combination including immune checkpoint inhibitors) targeting PD-1/PD-L1, IDO1, TIM3, LAG3), engineered immune cells (CAR-T and CAR M), immune agonists (targeting OX40, CD27,4-1BB, CD40), and BET inhibitors are under investigation. Notably, tumor-associated macrophages are one of the most important cell-associated with the development of an immune-depressive habitat. Thus, a strategy able to reverse this effect such as CAR M assumes a particular interest.

Strategies aimed to inhibit the signaling between GBM cells and the microenvironment are also of key importance as able to inhibit the manipulation carried out by tumor cells on surrounding tissues. Inhibitors of sphingomyelinase, annexin A1 can act on extracellular vesicles secretion and intake while other agents such as ADAM10 inhibitors could directly interfere with the jap junctions connecting GBM to other cells.

Finally, it should be noted that a specific microenvironment composition (such as that described on the white matter) can promote GBM differentiation and convert a tumor cell into an oligodendrocyte well-differentiated element. This surprising finding suggests that there are some elements inside the microenvironment that can provide inhibitory messages to GBM reversing its natural course. Interactions between neurons and glia (especially oligodendrocyte) appear of particular interest and should be further assessed as could hide important targets for novel drugs development. In conclusion, GBM should be considered as a network of interactions in which an action perpetuates against tumor cells result in a response of the associated microenvironment and vice versa. The role of the microenvironment should be always considered during pre-clinical studies and would offer novel targets for patients with GBM.

Author Contributions

VN and LG: writing and draft. EF, AB, AT, and SB: project conception and reviewing. All authors contributed to the article and approved the submitted version.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Glossary

ADAM10: Adam metallopeptidase domain 10

APC: Antigen Presenting Cell

ARG1: arginase 1

A2aR: adenosine A2A receptor

ATP: adenosine triphosphate

BDNF: brain derived neurotrophic factor

BET: Bromodomain and extra terminal motif proteins family

BRD3: Bromodomain containing protein 3

BRD4: Bromodomain containing protein 4

CD70: Cluster of differentiation 70

CD133: prominin-1

CCL2: C-C Motif Chemokine Ligand 2

CCL5: C-C Motif Chemokine Ligand 5

CCL7: C-C Motif Chemokine Ligand 7

COX 2: cyclooxygenase-2

CCL20: C-C Motif Chemokine Ligand 20

CXCL3: C-X-C Motif Chemokine Ligand 3

CXCL5: C-X-C Motif Chemokine Ligand 5

CXCL9: C-X-C Motif Chemokine Ligand 9

CXCL10: C-X-C Motif Chemokine Ligand 10

CXCL11: C-X-C Motif Chemokine Ligand 11

CNS: Central Nervous System

CSF-1: Colony stimulating factor 1

DLL4: Delta-like canonical Notch ligand 4

EC: Endothelial cell

ECM: Extra-cellular matrix

EGF: Epiderma growth factor

EGFR: Epidermal growth factor receptor

ERK 1/2: extracellular signal-regulated kinases 1 and 2

FAK: focal adhesion kinase

FGF: Fibroblast Growth factor

GBM: Glioblastoma

GDNF: Glial Cell Derived Neutrophic Factor

GM-CSF: Granulocyte-Macrophage Colony-Stimulating Factor

GSC: Glioblastoma stem cell

JAK: Jasus Kinase

HGF: Hepatocyte growth factor

HIF-1: hypoxia inducible factor 1

HIF-2: hypoxia inducible factor 2

HIG-2: hypoxia inducible protein 2

HSPA5: brain-derived neurotrophic factor chaperone Bip

IDH: Isocitrate dehydrogenase

IDO1: Indoleamine 2,3-dioxygenase

IGF1: insulin growth factor 1

IGFBP1: insulin-like growth factor-binding protein 1

IL5: Interleukin 5

IL6: Interleukin 6

IL8: Interleukin 8

IL10: Interleukin 10

INF γ: interferon γ

iNOS: inducible nitric oxide synthase

LAG 3: lymphocyte activation gene 3

MAPK: mitogen-activated protein kinase

MGMT: O6 – methylguanine DNA methyltransferase

MP: Metalloprotease

mTOR: mammalian target of rapamycin

NMDA: N-methyl-D-aspartate receptors

NOX 4: NADPH oxidase 4

NSC: Neural stem cell

NTRK2: neurotrophic receptor tyrosine kinase 2

Oct4: Octamer-binding transcription factor 4

OLIG2: Oligodendrocyte transcription factor 2

OS: Overall survival

PD-1: Programmed Death Receptor 1

PD-L1: Programmed Death Receptor Ligand 1

PI3K: phosphoinositide-3-kinase

ROS: reactive oxygen species

SDF1: stromal cell-derived factor 1

SHH: Sonic Hedgehog

SOX2: SRY-Box Transcription Factor 2

SOX10: SRY-Box Transcription Factor 10

SPP: Signal Peptide Peptidase

STAT: Signal transducer and activator of transcription

TAA: Tumor associated astrocytes

TDO: Tryptophan 2,3-dioxygenase

TERT: Telomerase Reverse Transcriptase

TGFβ1: transforming growth factor β1

TIM-3: T cell immunoglobulin-mucin-domain containing3

TMZ: Temozolomide

TNF: tumor necrosis factor

VEGF: vascular endothelial growth factor

WHO: World Health Organization

References

1. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A Summary. Neuro Oncol (2021) 23(8):1231–51. doi: 10.1093/neuonc/noab106

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al. EANO Guidelines on the Diagnosis and Treatment of Diffuse Gliomas of Adulthood. Nat Rev Clin Oncol (2021) 18(3):170–86. doi: 10.1038/s41571-020-00447-z

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of Radiotherapy With Concomitant and Adjuvant Temozolomide Versus Radiotherapy Alone on Survival in Glioblastoma in a Randomised Phase III Study: 5-Year Analysis of the EORTC-NCIC Trial. Lancet Oncol (2009) 10(5):459–66. doi: 10.1016/s1470-2045(09)70025-7

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy Plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N Engl J Med (2005) 352(10):987–96. doi: 10.1056/NEJMoa043330

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Brandes AA, Franceschi E, Paccapelo A, Tallini G, De Biase D, Ghimenton C, et al. Role of MGMT Methylation Status at Time of Diagnosis and Recurrence for Patients With Glioblastoma: Clinical Implications. Oncologist (2017) 22(4):432–7. doi: 10.1634/theoncologist.2016-0254

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Di Nunno V, Franceschi E, Tosoni A, Di Battista M, Gatto L, Lamperini C, et al. Treatment of Recurrent Glioblastoma: State-of-the-Art and Future Perspectives. Expert Rev Anticancer Ther (2020) 20(9):785–95. doi: 10.1080/14737140.2020.1807949

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Di Nunno V, Franceschi E, Tosoni A, Gatto L, Lodi R, Bartolini S, et al. Glioblastoma: Emerging Treatments and Novel Trial Designs. Cancers (Basel) (2021) 13(15). doi: 10.3390/cancers13153750

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Vanderbeek AM, Rahman R, Fell G, Ventz S, Chen T, Redd R, et al. The Clinical Trials Landscape for Glioblastoma: Is it Adequate to Develop New Treatments? Neuro Oncol (2018) 20(8):1034–43. doi: 10.1093/neuonc/noy027

PubMed Abstract | CrossRef Full Text | Google Scholar

9. McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, Mastrogianakis GM. Comprehensive Genomic Characterization Defines Human Glioblastoma Genes and Core Pathways. Nature (2008) 455(7216):1061–8. doi: 10.1038/nature07385

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Brennan CW, Verhaak RG, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The Somatic Genomic Landscape of Glioblastoma. Cell (2013) 155(2):462–77. doi: 10.1016/j.cell.2013.09.034

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Broekman ML, Maas SLN, Abels ER, Mempel TR, Krichevsky AM, Breakefield XO. Multidimensional Communication in the Microenvirons of Glioblastoma. Nat Rev Neurol (2018) 14(8):482–95. doi: 10.1038/s41582-018-0025-8

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Zhao J, Chen AX, Gartrell RD, Silverman AM, Aparicio L, Chu T, et al. Immune and Genomic Correlates of Response to Anti-PD-1 Immunotherapy in Glioblastoma. Nat Med (2019) 25(3):462–9. doi: 10.1038/s41591-019-0349-y

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Bradley CA. Glioblastoma: Stem Cells - Masters of Their Fates. Nat Rev Cancer (2017) 17(10):574–5. doi: 10.1038/nrc.2017.88

CrossRef Full Text | Google Scholar

14. 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). doi: 10.3390/ijms19020436

CrossRef Full Text | Google Scholar

15. Balça-Silva J, Matias D, Dubois LG, Carneiro B, do Carmo A, Girão H, et al. The Expression of Connexins and SOX2 Reflects the Plasticity of Glioma Stem-Like Cells. Transl Oncol (2017) 10(4):555–69. doi: 10.1016/j.tranon.2017.04.005

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Hong X, Sin WC, Harris AL, Naus CC. Gap Junctions Modulate Glioma Invasion by Direct Transfer of Microrna. Oncotarget (2015) 6(17):15566–77. doi: 10.18632/oncotarget.3904

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Joseph JV, Magaut CR, Storevik S, Geraldo LH, Mathivet T, Latif MA, et al. TGF-β Promotes Microtube Formation in Glioblastoma Through Thrombospondin 1. Neuro Oncol (2021). doi: 10.1093/neuonc/noab212

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Thuringer D, Boucher J, Jego G, Pernet N, Cronier L, Hammann A, et al. Transfer of Functional Micrornas Between Glioblastoma and Microvascular Endothelial Cells Through Gap Junctions. Oncotarget (2016) 7(45):73925–34. doi: 10.18632/oncotarget.12136

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Tkach M, Théry C. Communication by Extracellular Vesicles: Where We are and Where We Need to Go. Cell (2016) 164(6):1226–32. doi: 10.1016/j.cell.2016.01.043

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Weil S, Osswald M, Solecki G, Grosch J, Jung E, Lemke D, et al. Tumor Microtubes Convey Resistance to Surgical Lesions and Chemotherapy in Gliomas. Neuro Oncol (2017) 19(10):1316–26. doi: 10.1093/neuonc/nox070

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Osswald M, Jung E, Sahm F, Solecki G, Venkataramani V, Blaes J, et al. Brain Tumour Cells Interconnect to a Functional and Resistant Network. Nature (2015) 528(7580):93–8. doi: 10.1038/nature16071

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Pinto G, Saenz-de-Santa-Maria I, Chastagner P, Perthame E, Delmas C, Toulas C, et al. Patient-Derived Glioblastoma Stem Cells Transfer Mitochondria Through Tunneling Nanotubes in Tumor Organoids. Biochem J (2021) 478(1):21–39. doi: 10.1042/bcj20200710

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Valdebenito S, Audia A, Bhat KPL, Okafo G, Eugenin EA. Tunneling Nanotubes Mediate Adaptation of Glioblastoma Cells to Temozolomide and Ionizing Radiation Treatment. iScience (2020) 23(9):101450. doi: 10.1016/j.isci.2020.101450

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Valdebenito S, Malik S, Luu R, Loudig O, Mitchell M, Okafo G, et al. Tunneling Nanotubes, TNT, Communicate Glioblastoma With Surrounding Non-Tumor Astrocytes to Adapt Them to Hypoxic and Metabolic Tumor Conditions. Sci Rep (2021) 11(1):14556. doi: 10.1038/s41598-021-93775-8

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Ostrom QT, Edelson J, Byun J, Han Y, Kinnersley B, Melin B, et al. Partitioned Glioma Heritability Shows Subtype-Specific Enrichment in Immune Cells. Neuro Oncol (2021) 23(8):1304–14. doi: 10.1093/neuonc/noab072

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Chen Z, Hambardzumyan D. Immune Microenvironment in Glioblastoma Subtypes. Front Immunol (2018) 9:1004. doi: 10.3389/fimmu.2018.01004

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Daubon T, Hemadou A, Romero Garmendia I, Saleh M. Glioblastoma Immune Landscape and the Potential of New Immunotherapies. Front Immunol (2020) 11:585616:585616. doi: 10.3389/fimmu.2020.585616

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Li Q, Barres BA. Microglia and Macrophages in Brain Homeostasis and Disease. Nat Rev Immunol (2018) 18(4):225–42. doi: 10.1038/nri.2017.125

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Aspelund A, Antila S, Proulx ST, Karlsen TV, Karaman S, Detmar M, et al. A Dural Lymphatic Vascular System That Drains Brain Interstitial Fluid and Macromolecules. J Exp Med (2015) 212(7):991–9. doi: 10.1084/jem.20142290

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Rua R, McGavern DB. Advances in Meningeal Immunity. Trends Mol Med (2018) 24(6):542–59. doi: 10.1016/j.molmed.2018.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Basu A, Krady JK, Enterline JR, Levison SW. Transforming Growth Factor Beta1 Prevents IL-1beta-Induced Microglial Activation, Whereas Tnfalpha- and IL-6-Stimulated Activation Are Not Antagonized. Glia (2002) 40(1):109–20. doi: 10.1002/glia.10118

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Irani DN. How Much Control Does the Brain Exert Over the Immune System? Curr Opin Neurol (2002) 15(3):323–6. doi: 10.1097/00019052-200206000-00016

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Lobo-Silva D, Carriche GM, Castro AG, Roque S, Saraiva M. Balancing the Immune Response in the Brain: IL-10 and its Regulation. J Neuroinflamm (2016) 13(1):297. doi: 10.1186/s12974-016-0763-8

CrossRef Full Text | Google Scholar

34. Teeling JL, Perry VH. Systemic Infection and Inflammation in Acute CNS Injury and Chronic Neurodegeneration: Underlying Mechanisms. Neuroscience (2009) 158(3):1062–73. doi: 10.1016/j.neuroscience.2008.07.031

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Vitkovic L, Maeda S, Sternberg E. Anti-Inflammatory Cytokines: Expression and Action in the Brain. Neuroimmunomodulation (2001) 9(6):295–312. doi: 10.1159/000059387

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Masson F, Calzascia T, Di Berardino-Besson W, de Tribolet N, Dietrich PY, Walker PR. Brain Microenvironment Promotes the Final Functional Maturation of Tumor-Specific Effector CD8+ T Cells. J Immunol (2007) 179(2):845–53. doi: 10.4049/jimmunol.179.2.845

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Thomas DA, Massagué J. TGF-Beta Directly Targets Cytotoxic T Cell Functions During Tumor Evasion of Immune Surveillance. Cancer Cell (2005) 8(5):369–80. doi: 10.1016/j.ccr.2005.10.012

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Ma T, Hu C, Lal B, Zhou W, Ma Y, Ying M, et al. Reprogramming Transcription Factors Oct4 and Sox2 Induce a BRD-Dependent Immunosuppressive Transcriptome in GBM-Propagating Cells. Cancer Res (2021) 81(9):2457–69. doi: 10.1158/0008-5472.Can-20-2489

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Gusyatiner O, Bady P, Pham MDT, Lei Y, Park J, Daniel RT, et al. BET Inhibitors Repress Expression of Interferon-Stimulated Genes and Synergize With HDAC Inhibitors in Glioblastoma. Neuro Oncol (2021) 23(10):1680–92. doi: 10.1093/neuonc/noab115

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Ameratunga M, Braña I, Bono P, Postel-Vinay S, Plummer R, Aspegren J, et al. First-in-Human Phase 1 Open Label Study of the BET Inhibitor ODM-207 in Patients With Selected Solid Tumours. Br J Cancer (2020) 123(12):1730–6. doi: 10.1038/s41416-020-01077-z

PubMed Abstract | CrossRef Full Text | Google Scholar

41. Amorim S, Stathis A, Gleeson M, Iyengar S, Magarotto V, Leleu X, et al. Bromodomain Inhibitor OTX015 in Patients With Lymphoma or Multiple Myeloma: A Dose-Escalation, Open-Label, Pharmacokinetic, Phase 1 Study. Lancet Haematol (2016) 3(4):e196–204. doi: 10.1016/s2352-3026(16)00021-1

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Berthon C, Raffoux E, Thomas X, Vey N, Gomez-Roca C, Yee K, et al. Bromodomain Inhibitor OTX015 in Patients With Acute Leukaemia: A Dose-Escalation, Phase 1 Study. Lancet Haematol (2016) 3(4):e186–95. doi: 10.1016/s2352-3026(15)00247-1

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Falchook G, Rosen S, LoRusso P, Watts J, Gupta S, Coombs CC, et al. Development of 2 Bromodomain and Extraterminal Inhibitors With Distinct Pharmacokinetic and Pharmacodynamic Profiles for the Treatment of Advanced Malignancies. Clin Cancer Res (2020) 26(6):1247–57. doi: 10.1158/1078-0432.Ccr-18-4071

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Moreno V, Sepulveda JM, Vieito M, Hernández-Guerrero T, Doger B, Saavedra O, et al. Phase I Study of CC-90010, a Reversible, Oral BET Inhibitor in Patients With Advanced Solid Tumors and Relapsed/Refractory Non-Hodgkin’s Lymphoma. Ann Oncol (2020) 31(6):780–8. doi: 10.1016/j.annonc.2020.03.294

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Piha-Paul SA, Sachdev JC, Barve M, LoRusso P, Szmulewitz R, Patel SP, et al. First-in-Human Study of Mivebresib (ABBV-075), an Oral Pan-Inhibitor of Bromodomain and Extra Terminal Proteins, in Patients With Relapsed/Refractory Solid Tumors. Clin Cancer Res (2019) 25(21):6309–19. doi: 10.1158/1078-0432.Ccr-19-0578

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Postel-Vinay S, Herbschleb K, Massard C, Woodcock V, Soria JC, Walter AO, et al. First-in-Human Phase I Study of the Bromodomain and Extraterminal Motif Inhibitor BAY 1238097: Emerging Pharmacokinetic/Pharmacodynamic Relationship and Early Termination Due to Unexpected Toxicity. Eur J Cancer (2019) 109:103–10. doi: 10.1016/j.ejca.2018.12.020

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Shapiro GI, LoRusso P, Dowlati A, TD K, Jacobson CA, Vaishampayan U, et al. A Phase 1 Study of RO6870810, a Novel Bromodomain and Extra-Terminal Protein Inhibitor, in Patients With NUT Carcinoma, Other Solid Tumours, or Diffuse Large B-Cell Lymphoma. Br J Cancer (2021) 124(4):744–53. doi: 10.1038/s41416-020-01180-1

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Hambardzumyan D, Gutmann DH, Kettenmann H. The Role of Microglia and Macrophages in Glioma Maintenance and Progression. Nat Neurosci (2016) 19(1):20–7. doi: 10.1038/nn.4185

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Bowman RL, Klemm F, Akkari L, Pyonteck SM, Sevenich L, Quail DF, et al. Macrophage Ontogeny Underlies Differences in Tumor-Specific Education in Brain Malignancies. Cell Rep (2016) 17(9):2445–59. doi: 10.1016/j.celrep.2016.10.052

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Müller A, Brandenburg S, Turkowski K, Müller S, Vajkoczy P. Resident Microglia, and Not Peripheral Macrophages, are the Main Source of Brain Tumor Mononuclear Cells. Int J Cancer (2015) 137(2):278–88. doi: 10.1002/ijc.29379

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Li W, Graeber MB. The Molecular Profile of Microglia Under the Influence of Glioma. Neuro Oncol (2012) 14(8):958–78. doi: 10.1093/neuonc/nos116

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Bayik D, Zhou Y, Park C, Hong C, Vail D, Silver DJ, et al. Myeloid-Derived Suppressor Cell Subsets Drive Glioblastoma Growth in a Sex-Specific Manner. Cancer Discov (2020) 10(8):1210–25. doi: 10.1158/2159-8290.Cd-19-1355

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Chang AL, Miska J, Wainwright DA, Dey M, Rivetta CV, Yu D, et al. CCL2 Produced by the Glioma Microenvironment is Essential for the Recruitment of Regulatory T Cells and Myeloid-Derived Suppressor Cells. Cancer Res (2016) 76(19):5671–82. doi: 10.1158/0008-5472.Can-16-0144

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Brandenburg S, Müller A, Turkowski K, Radev YT, Rot S, Schmidt C, et al. Resident Microglia Rather Than Peripheral Macrophages Promote Vascularization in Brain Tumors and are Source of Alternative Pro-Angiogenic Factors. Acta Neuropathol (2016) 131(3):365–78. doi: 10.1007/s00401-015-1529-6

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage Polarization: Tumor-Associated Macrophages as a Paradigm for Polarized M2 Mononuclear Phagocytes. Trends Immunol (2002) 23(11):549–55. doi: 10.1016/s1471-4906(02)02302-5

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Ransohoff RM. A Polarizing Question: Do M1 and M2 Microglia Exist? Nat Neurosci (2016) 19(8):987–91. doi: 10.1038/nn.4338

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, et al. Transcriptome-Based Network Analysis Reveals a Spectrum Model of Human Macrophage Activation. Immunity (2014) 40(2):274–88. doi: 10.1016/j.immuni.2014.01.006

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Du R, Petritsch C, Lu K, Liu P, Haller A, Ganss R, et al. Matrix Metalloproteinase-2 Regulates Vascular Patterning and Growth Affecting Tumor Cell Survival and Invasion in GBM. Neuro Oncol (2008) 10(3):254–64. doi: 10.1215/15228517-2008-001

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Nijaguna MB, Patil V, Urbach S, Shwetha SD, Sravani K, Hegde AS, et al. Glioblastoma-Derived Macrophage Colony-Stimulating Factor (MCSF) Induces Microglial Release of Insulin-Like Growth Factor-Binding Protein 1 (IGFBP1) to Promote Angiogenesis. J Biol Chem (2015) 290(38):23401–15. doi: 10.1074/jbc.M115.664037

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Gatto L, Nunno VD, Franceschi E, Brandes AA. Chimeric Antigen Receptor Macrophage for Glioblastoma Immunotherapy: The Way Forward. Immunotherapy (2021) 13(11):879–83. doi: 10.2217/imt-2021-0054

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Sionov RV, Fridlender ZG, Granot Z. The Multifaceted Roles Neutrophils Play in the Tumor Microenvironment. Cancer Microenviron (2015) 8(3):125–58. doi: 10.1007/s12307-014-0147-5

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Attarha S, Roy A, Westermark B, Tchougounova E. Mast Cells Modulate Proliferation, Migration and Stemness of Glioma Cells Through Downregulation of GSK3β Expression and Inhibition of STAT3 Activation. Cell Signal (2017) 37:81–92. doi: 10.1016/j.cellsig.2017.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Han S, Zhang C, Li Q, Dong J, Liu Y, Huang Y, et al. Tumour-Infiltrating CD4(+) and CD8(+) Lymphocytes as Predictors of Clinical Outcome in Glioma. Br J Cancer (2014) 110(10):2560–8. doi: 10.1038/bjc.2014.162

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Lohr J, Ratliff T, Huppertz A, Ge Y, Dictus C, Ahmadi R, et al. Effector T-Cell Infiltration Positively Impacts Survival of Glioblastoma Patients and is Impaired by Tumor-Derived TGF-β. Clin Cancer Res (2011) 17(13):4296–308. doi: 10.1158/1078-0432.Ccr-10-2557

PubMed Abstract | CrossRef Full Text | Google Scholar

65. D’Alessio A, Proietti G, Sica G, Scicchitano BM. Pathological and Molecular Features of Glioblastoma and its Peritumoral Tissue. Cancers (Basel) (2019) 11(4). doi: 10.3390/cancers11040469

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Valdor R, García-Bernal D, Bueno C, Ródenas M, Moraleda JM, Macian F, et al. Glioblastoma Progression is Assisted by Induction of Immunosuppressive Function of Pericytes Through Interaction With Tumor Cells. Oncotarget (2017) 8(40):68614–26. doi: 10.18632/oncotarget.19804

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Wherry EJ, Kurachi M. Molecular and Cellular Insights Into T Cell Exhaustion. Nat Rev Immunol (2015) 15(8):486–99. doi: 10.1038/nri3862

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Reardon DA, Brandes AA, Omuro A, Mulholland P, Lim M, Wick 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):1003–10. doi: 10.1001/jamaoncol.2020.1024

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Di Nunno V, Franceschi E, Gatto L, Bartolini S, Brandes AA. Predictive Markers of Immune Response in Glioblastoma: Hopes and Facts. Future Oncol (2020) 16(15):1053–63. doi: 10.2217/fon-2020-0047

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Chan HY, Choi J, Jackson C, Lim M. Combination Immunotherapy Strategies for Glioblastoma. J Neurooncol (2021) 151(3):375–91. doi: 10.1007/s11060-020-03481-0

PubMed Abstract | CrossRef Full Text | Google Scholar

71. Cuddapah VA, Robel S, Watkins S, Sontheimer H. A Neurocentric Perspective on Glioma Invasion. Nat Rev Neurosci (2014) 15(7):455–65. doi: 10.1038/nrn3765

PubMed Abstract | CrossRef Full Text | Google Scholar

72. Piccirillo SG, Combi R, Cajola L, Patrizi A, Redaelli S, Bentivegna A, et al. Distinct Pools of Cancer Stem-Like Cells Coexist Within Human Glioblastomas and Display Different Tumorigenicity and Independent Genomic Evolution. Oncogene (2009) 28(15):1807–11. doi: 10.1038/onc.2009.27

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Molina JR, Hayashi Y, Stephens C, Georgescu MM. Invasive Glioblastoma Cells Acquire Stemness and Increased Akt Activation. Neoplasia (2010) 12(6):453–63. doi: 10.1593/neo.10126

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Vehlow A, Cordes N. Invasion as Target for Therapy of Glioblastoma Multiforme. Biochim Biophys Acta (2013) 1836(2):236–44. doi: 10.1016/j.bbcan.2013.07.001

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Brooks LJ, Clements MP, Burden JJ, Kocher D, Richards L, Devesa SC, et al. The White Matter is a Pro-Differentiative Niche for Glioblastoma. Nat Commun (2021) 12(1):2184. doi: 10.1038/s41467-021-22225-w

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Seike T, Fujita K, Yamakawa Y, Kido MA, Takiguchi S, Teramoto N, et al. Interaction Between Lung Cancer Cells and Astrocytes via Specific Inflammatory Cytokines in the Microenvironment of Brain Metastasis. Clin Exp Metastasis (2011) 28(1):13–25. doi: 10.1007/s10585-010-9354-8

PubMed Abstract | CrossRef Full Text | Google Scholar

77. 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):61–9. doi: 10.1007/s13277-015-4242-0

PubMed Abstract | CrossRef Full Text | Google Scholar

78. Clement V, Sanchez P, de Tribolet N, Radovanovic I, Ruiz i Altaba A. HEDGEHOG-GLI1 Signaling Regulates Human Glioma Growth, Cancer Stem Cell Self-Renewal, and Tumorigenicity. Curr Biol (2007) 17(2):165–72. doi: 10.1016/j.cub.2006.11.033

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Brandao M, Simon T, Critchley G, Giamas G. Astrocytes, the Rising Stars of the Glioblastoma Microenvironment. Glia (2019) 67(5):779–90. doi: 10.1002/glia.23520

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Becher OJ, Hambardzumyan D, Fomchenko EI, Momota H, Mainwaring L, Bleau AM, et al. Gli Activity Correlates With Tumor Grade in Platelet-Derived Growth Factor-Induced Gliomas. Cancer Res (2008) 68(7):2241–9. doi: 10.1158/0008-5472.Can-07-6350

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Pink RC, Elmusrati AA, Lambert D, Carter DRF. Royal Society Scientific Meeting: Extracellular Vesicles in the Tumour Microenvironment. Philos Trans R Soc Lond B Biol Sci (2018) 373(1737). doi: 10.1098/rstb.2017.0066

CrossRef Full Text | Google Scholar

82. Vasudev NS, Reynolds AR. Anti-Angiogenic Therapy for Cancer: Current Progress, Unresolved Questions and Future Directions. Angiogenesis (2014) 17(3):471–94. doi: 10.1007/s10456-014-9420-y

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Quail DF, Joyce JA. The Microenvironmental Landscape of Brain Tumors. Cancer Cell (2017) 31(3):326–41. doi: 10.1016/j.ccell.2017.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Chen W, Wang D, Du X, He Y, Chen S, Shao Q, et al. Glioma Cells Escaped From Cytotoxicity of Temozolomide and Vincristine by Communicating With Human Astrocytes. Med Oncol (2015) 32(3):43. doi: 10.1007/s12032-015-0487-0

PubMed Abstract | CrossRef Full Text | Google Scholar

85. Li F, Liu X, Sampson JH, Bigner DD, Li CY. Rapid Reprogramming of Primary Human Astrocytes Into Potent Tumor-Initiating Cells With Defined Genetic Factors. Cancer Res (2016) 76(17):5143–50. doi: 10.1158/0008-5472.Can-16-0171

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Jahani-Asl A, Yin H, Soleimani VD, Haque T, Luchman HA, Chang NC, et al. Control of Glioblastoma Tumorigenesis by Feed-Forward Cytokine Signaling. Nat Neurosci (2016) 19(6):798–806. doi: 10.1038/nn.4295

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Khwaja FW, Svoboda P, Reed M, Pohl J, Pyrzynska B, Van Meir EG. Proteomic Identification of the Wt-P53-Regulated Tumor Cell Secretome. Oncogene (2006) 25(58):7650–61. doi: 10.1038/sj.onc.1209969

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Biasoli D, Sobrinho MF, da Fonseca AC, de Matos DG, Romão L, de Moraes Maciel R, et al. Glioblastoma Cells Inhibit Astrocytic P53-Expression Favoring Cancer Malignancy. Oncogenesis (2014) 3(10):e123. doi: 10.1038/oncsis.2014.36

PubMed Abstract | CrossRef Full Text | Google Scholar

89. El Fatimy R, Subramanian S, Uhlmann EJ, Krichevsky AM. Genome Editing Reveals Glioblastoma Addiction to Microrna-10b. Mol Ther (2017) 25(2):368–78. doi: 10.1016/j.ymthe.2016.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Vazana U, Veksler R, Pell GS, Prager O, Fassler M, Chassidim Y, et al. Glutamate-Mediated Blood-Brain Barrier Opening: Implications for Neuroprotection and Drug Delivery. J Neurosci (2016) 36(29):7727–39. doi: 10.1523/jneurosci.0587-16.2016

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Sattler R, Tyler B, Hoover B, Coddington LT, Recinos V, Hwang L, et al. Increased Expression of Glutamate Transporter GLT-1 in Peritumoral Tissue Associated With Prolonged Survival and Decreases in Tumor Growth in a Rat Model of Experimental Malignant Glioma. J Neurosurg (2013) 119(4):878–86. doi: 10.3171/2013.6.Jns122319

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Venkatesh HS, Johung TB, Caretti V, Noll A, Tang Y, Nagaraja S, et al. Neuronal Activity Promotes Glioma Growth Through Neuroligin-3 Secretion. Cell (2015) 161(4):803–16. doi: 10.1016/j.cell.2015.04.012

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Peferoen L, Kipp M, van der Valk P, van Noort JM, Amor S. Oligodendrocyte-Microglia Cross-Talk in the Central Nervous System. Immunology (2014) 141(3):302–13. doi: 10.1111/imm.12163

PubMed Abstract | CrossRef Full Text | Google Scholar

94. Asslaber M, Schauer S, Gogg-Kamerer M, Bernhart E, Quehenberger F, Haybaeck J. Native Oligodendrocytes in Astrocytomas Might Inhibit Tumor Proliferation by WIF1 Expression. J Neuropathol Exp Neurol (2017) 76(1):16–26. doi: 10.1093/jnen/nlw098

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Calabrese C, Poppleton H, Kocak M, Hogg TL, Fuller C, Hamner B, et al. A Perivascular Niche for Brain Tumor Stem Cells. Cancer Cell (2007) 11(1):69–82. doi: 10.1016/j.ccr.2006.11.020

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Hambardzumyan D, Bergers G. Glioblastoma: Defining Tumor Niches. Trends Cancer (2015) 1(4):252–65. doi: 10.1016/j.trecan.2015.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Li Z, Bao S, Wu Q, Wang H, Eyler C, Sathornsumetee S, et al. Hypoxia-Inducible Factors Regulate Tumorigenic Capacity of Glioma Stem Cells. Cancer Cell (2009) 15(6):501–13. doi: 10.1016/j.ccr.2009.03.018

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, et al. Identification of Human Brain Tumour Initiating Cells. Nature (2004) 432(7015):396–401. doi: 10.1038/nature03128

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Berg TJ, Marques C, Pantazopoulou V, Johansson E, von Stedingk K, Lindgren D, et al. The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2. Cancer Res (2021) 81(8):2101–15. doi: 10.1158/0008-5472.Can-20-1785

PubMed Abstract | CrossRef Full Text | Google Scholar

100. Sharifzad F, Ghavami S, Verdi J, Mardpour S, Mollapour Sisakht M, Azizi Z, et al. Glioblastoma Cancer Stem Cell Biology: Potential Theranostic Targets. Drug Resist Update (2019) 42:35–45. doi: 10.1016/j.drup.2018.03.003

CrossRef Full Text | Google Scholar

101. Ahmed SI, Javed G, Laghari AA, Bareeqa SB, Farrukh S, Zahid S, et al. CD133 Expression in Glioblastoma Multiforme: A Literature Review. Cureus (2018) 10(10):e3439. doi: 10.7759/cureus.3439

PubMed Abstract | CrossRef Full Text | Google Scholar

102. Zhang M, Song T, Yang L, Chen R, Wu L, Yang Z, et al. Nestin and CD133: Valuable Stem Cell-Specific Markers for Determining Clinical Outcome of Glioma Patients. J Exp Clin Cancer Res (2008) 27(1):85. doi: 10.1186/1756-9966-27-85

PubMed Abstract | CrossRef Full Text | Google Scholar

103. D’Alessio A, Proietti G, Lama G, Biamonte F, Lauriola L, Moscato U, et al. Analysis of Angiogenesis Related Factors in Glioblastoma, Peritumoral Tissue and Their Derived Cancer Stem Cells. Oncotarget (2016) 7(48):78541–56. doi: 10.18632/oncotarget.12398

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Lemée JM, Clavreul A, Menei P. Intratumoral Heterogeneity in Glioblastoma: Don’t Forget the Peritumoral Brain Zone. Neuro Oncol (2015) 17(10):1322–32. doi: 10.1093/neuonc/nov119

PubMed Abstract | CrossRef Full Text | Google Scholar

105. Mei X, Chen YS, Chen FR, Xi SY, Chen ZP. Glioblastoma Stem Cell Differentiation Into Endothelial Cells Evidenced Through Live-Cell Imaging. Neuro Oncol (2017) 19(8):1109–18. doi: 10.1093/neuonc/nox016

PubMed Abstract | CrossRef Full Text | Google Scholar

106. Burger PC, Dubois PJ, Schold SC Jr., Smith KR Jr., Odom GL, Crafts DC, et al. Computerized Tomographic and Pathologic Studies of the Untreated, Quiescent, and Recurrent Glioblastoma Multiforme. J Neurosurg (1983) 58(2):159–69. doi: 10.3171/jns.1983.58.2.0159

PubMed Abstract | CrossRef Full Text | Google Scholar

107. Piccirillo SG, Dietz S, Madhu B, Griffiths J, Price SJ, Collins VP, et al. Fluorescence-Guided Surgical Sampling of Glioblastoma Identifies Phenotypically Distinct Tumour-Initiating Cell Populations in the Tumour Mass and Margin. Br J Cancer (2012) 107(3):462–8. doi: 10.1038/bjc.2012.271

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Ruiz-Ontañon P, Orgaz JL, Aldaz B, Elosegui-Artola A, Martino J, Berciano MT, et al. Cellular Plasticity Confers Migratory and Invasive Advantages to a Population of Glioblastoma-Initiating Cells That Infiltrate Peritumoral Tissue. Stem Cells (2013) 31(6):1075–85. doi: 10.1002/stem.1349

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Wang X, Prager BC, Wu Q, Kim LJY, Gimple RC, Shi Y, et al. Reciprocal Signaling Between Glioblastoma Stem Cells and Differentiated Tumor Cells Promotes Malignant Progression. Cell Stem Cell (2018) 22(4):514–28.e5. doi: 10.1016/j.stem.2018.03.011

PubMed Abstract | CrossRef Full Text | Google Scholar

110. Peleli M, Moustakas A, Papapetropoulos A. Endothelial-Tumor Cell Interaction in Brain and CNS Malignancies. Int J Mol Sci (2020) 21(19). doi: 10.3390/ijms21197371

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Beppu T, Kamada K, Yoshida Y, Arai H, Ogasawara K, Ogawa A. Change of Oxygen Pressure in Glioblastoma Tissue Under Various Conditions. J Neurooncol (2002) 58(1):47–52. doi: 10.1023/a:1015832726054

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Colwell N, Larion M, Giles AJ, Seldomridge AN, Sizdahkhani S, Gilbert MR, et al. Hypoxia in the Glioblastoma Microenvironment: Shaping the Phenotype of Cancer Stem-Like Cells. Neuro Oncol (2017) 19(7):887–96. doi: 10.1093/neuonc/now258

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Mihaylova VT, Bindra RS, Yuan J, Campisi D, Narayanan L, Jensen R, et al. Decreased Expression of the DNA Mismatch Repair Gene Mlh1 Under Hypoxic Stress in Mammalian Cells. Mol Cell Biol (2003) 23(9):3265–73. doi: 10.1128/mcb.23.9.3265-3273.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Görlach A, Dimova EY, Petry A, Martínez-Ruiz A, Hernansanz-Agustín P, Rolo AP, et al. Reactive Oxygen Species, Nutrition, Hypoxia and Diseases: Problems Solved? Redox Biol (2015) 6:372–85. doi: 10.1016/j.redox.2015.08.016

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Zuo L, Zhou T, Pannell BK, Ziegler AC, Best TM. Biological and Physiological Role of Reactive Oxygen Species–the Good, the Bad and the Ugly. Acta Physiol (Oxf) (2015) 214(3):329–48. doi: 10.1111/apha.12515

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Lo HW. Targeting Ras-RAF-ERK and its Interactive Pathways as a Novel Therapy for Malignant Gliomas. Curr Cancer Drug Targets (2010) 10(8):840–8. doi: 10.2174/156800910793357970

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Ohgaki H, Kleihues P. Genetic Alterations and Signaling Pathways in the Evolution of Gliomas. Cancer Sci (2009) 100(12):2235–41. doi: 10.1111/j.1349-7006.2009.01308.x

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Li Y, Han N, Yin T, Huang L, Liu S, Liu D, et al. Lentivirus-Mediated Nox4 Shrna Invasion and Angiogenesis and Enhances Radiosensitivity in Human Glioblastoma. Oxid Med Cell Longev (2014) 2014:581732. doi: 10.1155/2014/581732

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Moschetta M, Mishima Y, Sahin I, Manier S, Glavey S, Vacca A, et al. Role of Endothelial Progenitor Cells in Cancer Progression. Biochim Biophys Acta (2014) 1846(1):26–39. doi: 10.1016/j.bbcan.2014.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, et al. Bone Marrow Cells Regenerate Infarcted Myocardium. Nature (2001) 410(6829):701–5. doi: 10.1038/35070587

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Duda DG, Cohen KS, Kozin SV, Perentes JY, Fukumura D, Scadden DT, et al. Evidence for Incorporation of Bone Marrow-Derived Endothelial Cells Into Perfused Blood Vessels in Tumors. Blood (2006) 107(7):2774–6. doi: 10.1182/blood-2005-08-3210

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Wang SC, Yu CF, Hong JH, Tsai CS, Chiang CS. Radiation Therapy-Induced Tumor Invasiveness is Associated With SDF-1-Regulated Macrophage Mobilization and Vasculogenesis. PloS One (2013) 8(8):e69182. doi: 10.1371/journal.pone.0069182

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Hellström M, Phng LK, Hofmann JJ, Wallgard E, Coultas L, Lindblom P, et al. Dll4 Signalling Through Notch1 Regulates Formation of Tip Cells During Angiogenesis. Nature (2007) 445(7129):776–80. doi: 10.1038/nature05571

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Fernández-Cortés M, Delgado-Bellido D, Oliver FJ. Vasculogenic Mimicry: Become an Endothelial Cell “But Not So Much”. Front Oncol (2019) 9:803. doi: 10.3389/fonc.2019.00803

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Hendrix MJ, Seftor EA, Meltzer PS, Gardner LM, Hess AR, Kirschmann DA, et al. Expression and Functional Significance of VE-Cadherin in Aggressive Human Melanoma Cells: Role in Vasculogenic Mimicry. Proc Natl Acad Sci U.S.A. (2001) 98(14):8018–23. doi: 10.1073/pnas.131209798

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Ricci-Vitiani L, Pallini R, Biffoni M, Todaro M, Invernici G, Cenci T, et al. Tumour Vascularization via Endothelial Differentiation of Glioblastoma Stem-Like Cells. Nature (2010) 468(7325):824–8. doi: 10.1038/nature09557

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Rong X, Huang B, Qiu S, Li X, He L, Peng Y. Tumor-Associated Macrophages Induce Vasculogenic Mimicry of Glioblastoma Multiforme Through Cyclooxygenase-2 Activation. Oncotarget (2016) 7(51):83976–86. doi: 10.18632/oncotarget.6930

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Mao XG, Xue XY, Wang L, Zhang X, Yan M, Tu YY, et al. CDH5 is Specifically Activated in Glioblastoma Stemlike Cells and Contributes to Vasculogenic Mimicry Induced by Hypoxia. Neuro Oncol (2013) 15(7):865–79. doi: 10.1093/neuonc/not029

PubMed Abstract | CrossRef Full Text | Google Scholar

129. Krusche B, Ottone C, Clements MP, Johnstone ER, Goetsch K, Lieven H, et al. Ephrinb2 Drives Perivascular Invasion and Proliferation of Glioblastoma Stem-Like Cells. Elife (2016) 5. doi: 10.7554/eLife.14845

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Seifert S, Sontheimer H. Bradykinin Enhances Invasion of Malignant Glioma Into the Brain Parenchyma by Inducing Cells to Undergo Amoeboid Migration. J Physiol (2014) 592(22):5109–27. doi: 10.1113/jphysiol.2014.274498

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Lindberg OR, McKinney A, Engler JR, Koshkakaryan G, Gong H, Robinson AE, et al. GBM Heterogeneity as a Function of Variable Epidermal Growth Factor Receptor Variant III Activity. Oncotarget (2016) 7(48):79101–16. doi: 10.18632/oncotarget.12600

CrossRef Full Text | Google Scholar

132. Szabo E, Schneider H, Seystahl K, Rushing EJ, Herting F, Weidner KM, et al. Autocrine VEGFR1 and VEGFR2 Signaling Promotes Survival in Human Glioblastoma Models in Vitro and in Vivo. Neuro Oncol (2016) 18(9):1242–52. doi: 10.1093/neuonc/now043

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Wick W, Gorlia T, Bendszus M, Taphoorn M, Sahm F, Harting I, et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N Engl J Med (2017) 377(20):1954–63. doi: 10.1056/NEJMoa1707358

PubMed Abstract | CrossRef Full Text | Google Scholar

134. Wefel JS, Armstrong TS, Pugh SL, Gilbert MR, Wendland MM, Brachman DG, et al. Neurocognitive, Symptom, and Health-Related Quality of Life Outcomes of a Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma (NRG/RTOG 0825). Neuro Oncol (2021) 23(7):1125–38. doi: 10.1093/neuonc/noab011

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N Engl J Med (2014) 370(8):699–708. doi: 10.1056/NEJMoa1308573

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, et al. Bevacizumab Plus Radiotherapy-Temozolomide for Newly Diagnosed Glioblastoma. N Engl J Med (2014) 370(8):709–22. doi: 10.1056/NEJMoa1308345

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Tamura R, Tanaka T, Akasaki Y, Murayama Y, Yoshida K, Sasaki H. The Role of Vascular Endothelial Growth Factor in the Hypoxic and Immunosuppressive Tumor Microenvironment: Perspectives for Therapeutic Implications. Med Oncol (2019) 37(1):2. doi: 10.1007/s12032-019-1329-2

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Nayak L, Molinaro AM, Peters K, Clarke JL, Jordan JT, de Groot J, et al. Randomized Phase II and Biomarker Study of Pembrolizumab Plus Bevacizumab Versus Pembrolizumab Alone for Patients With Recurrent Glioblastoma. Clin Cancer Res (2021) 27(4):1048–57. doi: 10.1158/1078-0432.Ccr-20-2500

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Brandes AA, Tosoni A, Franceschi E, Blatt V, Santoro A, Faedi M, et al. Fotemustine as Second-Line Treatment for Recurrent or Progressive Glioblastoma After Concomitant and/or Adjuvant Temozolomide: A Phase II Trial of Gruppo Italiano Cooperativo Di Neuro-Oncologia (GICNO). Cancer Chemother Pharmacol (2009) 64(4):769–75. doi: 10.1007/s00280-009-0926-8

PubMed Abstract | CrossRef Full Text | Google Scholar

140. Franceschi E, Bartolotti M, Tosoni A, Bartolini S, Sturiale C, Fioravanti A, et al. The Effect of Re-Operation on Survival in Patients With Recurrent Glioblastoma. Anticancer Res (2015) 35(3):1743–8.

PubMed Abstract | Google Scholar

141. Tosoni A, Franceschi E, Ermani M, Bertorelle R, Bonaldi L, Blatt V, et al. Temozolomide Three Weeks on and One Week Off as First Line Therapy for Patients With Recurrent or Progressive Low Grade Gliomas. J Neurooncol (2008) 89(2):179–85. doi: 10.1007/s11060-008-9600-y

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Van Den Bent M, Eoli M, Sepulveda JM, Smits M, Walenkamp A, Frenel JS, et al. INTELLANCE 2/EORTC 1410 Randomized Phase II Study of Depatux-M Alone and With Temozolomide vs Temozolomide or Lomustine in Recurrent EGFR Amplified Glioblastoma. Neuro Oncol (2020) 22(5):684–93. doi: 10.1093/neuonc/noz222

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Woroniecka K, Chongsathidkiet P, Rhodin K, Kemeny H, Dechant C, Farber SH, et al. T-Cell Exhaustion Signatures Vary With Tumor Type and are Severe in Glioblastoma. Clin Cancer Res (2018) 24(17):4175–86. doi: 10.1158/1078-0432.Ccr-17-1846

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Singh K, Batich KA, Wen PY, Tan AC, Bagley SJ, Lim M, et al. Designing Clinical Trials for Combination Immunotherapy: A Framework for Glioblastoma. Clin Cancer Res (2021). doi: 10.1158/1078-0432.Ccr-21-2681

CrossRef Full Text | Google Scholar

145. Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim CK, Tobias A, et al. Durable Therapeutic Efficacy Utilizing Combinatorial Blockade Against IDO, CTLA-4, and PD-L1 in Mice With Brain Tumors. Clin Cancer Res (2014) 20(20):5290–301. doi: 10.1158/1078-0432.Ccr-14-0514

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Scott EM, Duffy MR, Freedman JD, Fisher KD, Seymour LW. Solid Tumor Immunotherapy With T Cell Engager-Armed Oncolytic Viruses. Macromol Biosci (2018) 18(1). doi: 10.1002/mabi.201700187

PubMed Abstract | CrossRef Full Text | Google Scholar

147. Gedeon PC, Schaller TH, Chitneni SK, Choi BD, Kuan CT, Suryadevara CM, et al. A Rationally Designed Fully Human Egfrviii : CD3-Targeted Bispecific Antibody Redirects Human T Cells to Treat Patient-Derived Intracerebral Malignant Glioma. Clin Cancer Res (2018) 24(15):3611–31. doi: 10.1158/1078-0432.Ccr-17-0126

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Quail DF, Bowman RL, Akkari L, Quick ML, Schuhmacher AJ, Huse JT, et al. The Tumor Microenvironment Underlies Acquired Resistance to CSF-1R Inhibition in Gliomas. Science (2016) 352(6288):aad3018. doi: 10.1126/science.aad3018

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Lombardi G, Pambuku A, Bellu L, Farina M, Della Puppa A, Denaro L, et al. Effectiveness of Antiangiogenic Drugs in Glioblastoma Patients: A Systematic Review and Meta-Analysis of Randomized Clinical Trials. Crit Rev Oncol Hematol (2017) 111:94–102. doi: 10.1016/j.critrevonc.2017.01.018

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Levin VA, Chan J, Datta M, Yee JL, Jain RK. Effect of Angiotensin System Inhibitors on Survival in Newly Diagnosed Glioma Patients and Recurrent Glioblastoma Patients Receiving Chemotherapy and/or Bevacizumab. J Neurooncol (2017) 134(2):325–30. doi: 10.1007/s11060-017-2528-3

PubMed Abstract | CrossRef Full Text | Google Scholar

151. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. Depletion of Microglia and Inhibition of Exosome Synthesis Halt Tau Propagation. Nat Neurosci (2015) 18(11):1584–93. doi: 10.1038/nn.4132

PubMed Abstract | CrossRef Full Text | Google Scholar

152. Atai NA, Balaj L, van Veen H, Breakefield XO, Jarzyna PA, Van Noorden CJ, et al. Heparin Blocks Transfer of Extracellular Vesicles Between Donor and Recipient Cells. J Neurooncol (2013) 115(3):343–51. doi: 10.1007/s11060-013-1235-y

PubMed Abstract | CrossRef Full Text | Google Scholar

153. Jansen F, Yang X, Hoyer FF, Paul K, Heiermann N, Becher MU, et al. Endothelial Microparticle Uptake in Target Cells is Annexin I/Phosphatidylserine Receptor Dependent and Prevents Apoptosis. Arterioscler Thromb Vasc Biol (2012) 32(8):1925–35. doi: 10.1161/atvbaha.112.253229

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Hitomi M, Deleyrolle LP, Mulkearns-Hubert EE, Jarrar A, Li M, Sinyuk M, et al. Differential Connexin Function Enhances Self-Renewal in Glioblastoma. Cell Rep (2015) 11(7):1031–42. doi: 10.1016/j.celrep.2015.04.021

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Fridman JS, Caulder E, Hansbury M, Liu X, Yang G, Wang Q, et al. Selective Inhibition of ADAM Metalloproteases as a Novel Approach for Modulating Erbb Pathways in Cancer. Clin Cancer Res (2007) 13(6):1892–902. doi: 10.1158/1078-0432.Ccr-06-2116

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Jimenez-Pascual A, Hale JS, Kordowski A, Pugh J, Silver DJ, Bayik D, et al. ADAMDEC1 Maintains a Growth Factor Signaling Loop in Cancer Stem Cells. Cancer Discov (2019) 9(11):1574–89. doi: 10.1158/2159-8290.Cd-18-1308

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: microenvironment, glioblastoma, macrophages, neurons, immune-system

Citation: Di Nunno V, Franceschi E, Tosoni A, Gatto L, Bartolini S and Brandes AA (2022) Glioblastoma Microenvironment: From an Inviolable Defense to a Therapeutic Chance. Front. Oncol. 12:852950. doi: 10.3389/fonc.2022.852950

Received: 11 January 2022; Accepted: 09 February 2022;
Published: 02 March 2022.

Edited by:

Alireza Mansouri, The Pennsylvania State University (PSU), United States

Reviewed by:

Maksim Sinyuk, Abeona Therapeutics, United States
Ryota Tamura, Keio University School of Medicine, Japan

Copyright © 2022 Di Nunno, Franceschi, Tosoni, Gatto, Bartolini and Brandes. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Enrico Franceschi, enricofra@yahoo.it

ORCID: Vincenzo Di Nunno, orcid.org/0000-0003-4441-9834
Enrico Franceschi, orcid.org/0000-0001-9332-4677
Alba Ariela Brandes, orcid.org/0000-0002-2503-9089

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.