Next Article in Journal
Identification of Key Active Constituents in Eucommia ulmoides Oliv. Leaves Against Parkinson’s Disease and the Alleviative Effects via 4E-BP1 Up-Regulation
Next Article in Special Issue
TPP-Based Nanovesicles Kill MDR Neuroblastoma Cells and Induce Moderate ROS Increase, While Exerting Low Toxicity Towards Primary Cell Cultures: An In Vitro Study
Previous Article in Journal
Seco-Duocarmycin SA in Aggressive Glioblastoma Cell Lines
Previous Article in Special Issue
Antitumor Activities of a Humanized Cancer-Specific Anti-HER2 Monoclonal Antibody, humH2Mab-250 in Human Breast Cancer Xenografts
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Understanding Neovascularization in Glioblastoma: Insights from the Current Literature

by
Mariagiovanna Ballato
1,
Emanuela Germanà
1,
Gabriele Ricciardi
1,2,
Walter Giuseppe Giordano
1,
Pietro Tralongo
1,
Mariachiara Buccarelli
3,
Giorgia Castellani
3,
Lucia Ricci-Vitiani
3,
Quintino Giorgio D’Alessandris
4,
Giuseppe Giuffrè
5,
Cristina Pizzimenti
6,
Vincenzo Fiorentino
5,
Valeria Zuccalà
5,
Antonio Ieni
5,
Maria Caffo
7,
Guido Fadda
5 and
Maurizio Martini
5,*
1
Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy
2
Istituto Clinico Polispecialistico C.O.T. Cure Ortopediche Traumatologiche s.pa., 98124 Messina, Italy
3
Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
4
Department of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
5
Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
6
Pathology Unit, Papardo Hospital, 98158 Messina, Italy
7
Biomedical and Dental Sciences and Morphofunctional Imaging, Unit of Neurosurgery, University of Messina, 98122 Messina, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(6), 2763; https://doi.org/10.3390/ijms26062763
Submission received: 6 February 2025 / Revised: 15 March 2025 / Accepted: 17 March 2025 / Published: 19 March 2025
(This article belongs to the Special Issue New Molecular Mechanisms and Advanced Therapies for Solid Tumors)

Abstract

Glioblastomas (GBMs), among the most aggressive and resilient brain tumors, characteristically exhibit high angiogenic potential, leading to the formation of a dense yet aberrant vasculature, both morphologically and functionally. With these premises, numerous expectations were initially placed on anti-angiogenic therapies, soon dashed by their limited efficacy in concretely improving patient outcomes. Neovascularization in GBM soon emerged as a complex, dynamic, and heterogeneous process, hard to manage with the classical standard of care. Growing evidence has revealed the existence of numerous non-canonical strategies of angiogenesis, variously exploited by GBM to meet its ever-increasing metabolic demand and differently involved in tumor progression, recurrence, and escape from treatments. In this review, we provide an accurate description of each neovascularization mode encountered in GBM tumors to date, highlighting the molecular players and signaling cascades primarily involved. We also detail the key architectural and functional aspects characteristic of the GBM vascular compartment because of an intricate crosstalk between the different angiogenic networks. Additionally, we explore the repertoire of emerging therapies against GBM that are currently under study, concluding with a question: faced with such a challenging scenario, could combined therapies, tailored to the patient’s genetic signatures, represent an effective game changer?

1. Introduction

Glial tumors, also known as gliomas, are malignant neoplasms that grow in the brain or, less frequently, in the spinal cord, whose cellular origin remains a matter of debate. This kind of tumor presumably arises from improperly reactivated glial cells or from aberrant neural stem/glial progenitor cells, but the different hypotheses are not necessarily mutually exclusive; distinct subtypes might have distinct tumor-initiating cells [1,2,3,4,5,6]. Gliomas can occur at any age, even in children, representing the most prevalent and aggressive primary tumors of the central nervous system (CNS) [7]. The disease is more frequently diagnosed in Caucasians, ranging from 45 to 65 years old [8,9,10,11,12,13]. Epidemiological evidence has also revealed higher incidence and shorter lifespan in males than in females, suggesting the possible existence of sex-related aspects in tumorigenesis, survival rate, and response to therapies [8,12,14,15,16,17]. However, regardless of gender, advanced age appears to be a negative prognostic factor, with poorer outcomes [18,19].
Gliomas are mostly sporadic tumors, although a small fraction of cases (about 5%) show familial features with the occurrence of the disease in two or more members of a family, generally with early onset [20,21]. In this context, recent genome-wide association studies (GWASs) through the identification of 25 putative risk loci (like TERT 5p15.33, MDM4 1q32.1, TP53 17p13.1, EGFR 7p11.2, and others) seem to support the scenario of a possible polygenic susceptibility to gliomas. The underlying molecular mechanisms, however, are still poorly understood [21,22,23]. In addition, approximately 1–2% of glial tumors are strictly associated with hereditary diseases, such as neurofibromatosis type 1, Lynch, Li–Fraumeni, Turcot, and Cowden syndromes [17,24]. Furthermore, several potential predisposing factors, including exposure to environmental agents, lifestyle choices, and unhealthy behaviors that may be related to a higher incidence of gliomas, are currently being studied. Individuals with a history of allergies or asthma, curiously, seem less inclined to develop glial tumors. However, to date, exposure to ionizing radiation is the only proven and well-documented environmental risk factor [17,21].
Given the level of heterogeneity, an integrated and layered diagnosis, although challenging, is required to ensure the most appropriate patient management. Specifically, gliomas form a wide collection of low- to high-grade tumors classified in compliance with the 2021 fifth edition World Health Organization (WHO) classification of tumors of the CNS [25,26,27]. Based on histopathological aspects, immunohistochemical features, and molecular/cytogenetic profiles, indeed, adult-type diffuse gliomas are currently categorized into three tumor types, as follows: (1) isocitrate dehydrogenase (IDH)-mutant and 1p/19q-codeleted oligodendrogliomas, (2) IDH-mutant astrocytomas, and (3) IDH-wildtype glioblastomas [28]. Furthermore, tumor grading, from grade 1 (the least aggressive) to grade 4 (the most aggressive) in terms of malignancy, is established not only by evaluating different histological criteria (mitosis, nuclear atypia, microvascular proliferation, and necrosis) but also by assessing molecular characteristics [29,30]. Moreover, under the updated classification, the presence of CDKN2A/B homozygous deletion is sufficient to consider an astrocytoma IDH-mutant, WHO grade 4, even without microvascular proliferation and/or necrosis. In the same way, an IDH-wildtype astrocytoma falls under the group of glioblastomas in the presence of at least one of the following characteristics: necrosis and/or microvascular proliferation and/or TERT promoter mutation and/or EGFR gene amplification and/or concomitant gain of chromosome 7 and loss of chromosome 10 [28].
Glioblastoma multiforme (GBM), with a median overall survival (OS) of around 15 months, represents, among gliomas, the most malignant and poorest prognosis form [31,32,33]. Hallmarks such as rapid growth, a highly infiltrative nature, a pronounced tendency to recur, and refractoriness to therapeutic options make GBM essentially incurable [34,35]. A “one-size-fits-all” approach (Stupp protocol) is currently used for glioblastoma patients. Specifically, the standard of care involves an aggressive, multimodal, tripartite strategy: safe surgical debulking, radiotherapy, and adjuvant chemotherapy with the alkylating drug temozolomide (TMZ) [35,36,37]. However, despite ongoing efforts to counteract the inexorable progression of the disease, treatment plan effectiveness is insufficient, leading to only slight improvements in survival rates [38].
One of the key features of GBMs is their extensive vascularization [39]. This signature trait has led to hopes for the success of anti-angiogenic therapies that have emerged in the last decades as a promising treatment option for several types of solid tumors [40]. By starving GBM, indeed, given the great vascular density, it was expected that this could stop or slow down its progression, leading to better prognoses [41]. Furthermore, by normalizing the tumor vascular network, anti-angiogenic therapies would have improved the tissue distribution and effectiveness of chemotherapeutic agents [42]. With these premises, GBM angiogenesis has become an important research focus, and in 2009, the FDA approved bevacizumab monotherapy—a recombinant humanized antibody against VEGF—for recurrent glioblastoma treatment [43,44,45]. However, as suggested by various clinical studies, while angiogenesis inhibitors (usually combined with conventional therapies) tend to improve the OS and/or PFS when used for the treatment of several tumors such as metastatic colon–rectal cancer, advanced non-small-cell lung cancer, or metastatic renal cell carcinoma, the introduction of anti-angiogenic therapies for the management of GBM patients, although initially promising, turned out to be only a “meteor” [46,47,48,49,50,51,52,53,54]. Apart from tumor recurrence or acquired resistance to angiogenic inhibitors, some evidence paradoxically reports a worsening in GBM aggressiveness after bevacizumab therapy [55]. The marginal clinical benefits may be attributed to the tumor resorting to alternative modes of neovascularization, aside from canonical angiogenesis, eventually exacerbated or activated as a compensation strategy by anti-angiogenic drugs. GBM harbors an exceptional level of intra- and inter-tumor heterogeneity as well, which poses further challenges in treatment. With the advances of groundbreaking research, three distinct molecular subtypes have been identified, namely classical (CL), proneural (PN), and mesenchymal (MES), characterized by different genetic backgrounds, prognoses, and therapeutic sensitivity [56]. Presumably, distinct subtypes exploit alternative neovascularization strategies, pro-angiogenic factors, and escape routes in a temporally and spatially variable manner. GBM with a mesenchymal signature, known for its worst outcomes, appears to strongly activate the trans-differentiation mechanism, for example, compared to the other subtypes, as suggested by the higher angiogenic potential of MES-GBM cancer stem cells (GSCs) relative to those of the PN subtype [57,58,59]. Moreover, unlike the other phenotypes, the mesenchymal one shows the greatest upregulation of pro-angiogenic factors like VEGF, VEGFR1/2, and PECAM1 (a signaling molecule expressed specifically by the endothelium). Additionally, the MES vasculature exhibits a higher Ki-67 index than the PN and CL subtypes, indicating enhanced levels of endothelial proliferation [60]. Therefore, understanding the inter-patient heterogeneity associated with different modes of neovascularization, as well as the multitude of underlying molecular dynamics and actors involved, is crucial for contributing to the development of novel personalized therapies.
In the present article, we review the multiple modes of neovascularization observed in the GBM ecosystem so far, summarizing the key factors and principle pathways involved. We also emphasize the structural features of tumor vasculature, as the result of the complex interplay between different neovascularization mechanisms. Moreover, we provide an updated version of the emerging therapeutic scenario in the fight against GBM angiogenesis, highlighting the urgent need for a different and innovative therapeutic approach, based on the specific molecular profile of the patient, with the possibility of combining therapies to hit multiple pathways, also those involved in angiogenesis.

2. GBM Vascularization: Morphological and Functional Aspects

Angiogenesis, the generation of new blood vessels from pre-existing vascular networks, holds a pivotal role in both physiological and pathological settings, including cancer [61]. In order to grow, infiltrate the surrounding tissue, and eventually metastasize, tumors need an increased blood supply to meet the rising demand for nutrients, oxygen, and waste removal [62]. In this context, compared with other solid tumors, GBM has a marked angiogenic potential [63]. Interestingly, however, tumor-associated blood vessels are characterized by various morphological and functional abnormalities, deserving of a deeper evaluation (Figure 1) [64]. Neovascularization in GBM is often described as accelerated yet dysfunctional [65,66]. In depth, newly sprouted blood vessels appear dilated, fenestrated, and highly permeable; have a tortuous course; and give rise to a poorly branched and disorganized tree-like structure. Sometimes, immature or dead-end blood vessels are also formed [67,68]. In addition, GBM vasculature does not present the classical hierarchical organization into arterioles, venules, and capillaries [69]. Moreover, “glomeruloid microvascular proliferations”—vascular clusters made of hyperplastic endothelial cells, smooth muscle cells, and pericytes—are also present, commonly near necrotic foci [68].
At a single-cell level, GBM-associated endothelial cells (GECs) reveal larger sizes, larger nuclei, multiple nucleoli, and a flattened, veil-like appearance, unlike the cobblestone-shaped ones typically found in normal brain tissue [70,71]. GECs are also more resistant to apoptosis and migrate faster, exploiting chemo-kinesis instead of chemotaxis [70]. This blood vessel dysmorphia ultimately causes several functional defects, leading to increased regional hypoxia, a non-homogenous blood supply, enhanced permeability, and possible edema formation along with increased interstitial fluid pressure which overall hinders effective drug delivery [69,72,73,74]. Actually, a broad spectrum of different neovascularization patterns participates in the creation of such a particular vascular bed [75].

3. Different Strategies of Neovascularization in GBM

Sprouting angiogenesis represents the canonical process by which solid tumors build new blood vessels. However, advances in research have identified several alternative mechanisms implicated at various levels in tumor growth, progression, and drug resistance [76]. Currently, to what extent each mechanism plays a role in human brain cancer remains a controversial issue, as well as their involvement in the development of resistance to anti-angiogenetic drugs, as supported by clinical evidence [77,78]. Understanding the relative contribution in the context of GBM would lead to a deeper comprehension of the biology of angiogenesis. Recognizing how the various mechanisms interact (if they work together or sometimes antagonize each other or are occasionally activated as a compensatory mechanism) and what the underlying regulatory pathways are is a necessary step in designing more effective therapeutic approaches [76,79].
To date, (1) sprouting angiogenesis, (2) vasculogenesis, (3) vessel co-option, (4) vascular intussusception, (5) vessel mimicry, and (6) the trans-differentiation of cancer stem-like cells are all strategies that GBM can exploit to ensure a proper “fuel supply” [80]. Vessel co-option and vessel mimicry, in particular, have recently emerged as important players in several tumor types, as they are involved in both metastasis and resistance mechanisms to vascular-directed therapies [78,81,82].

3.1. Sprouting Angiogenesis

Angiogenesis, more specifically termed sprouting angiogenesis (SA), is a dynamic but strictly monitored process in time and space, which ensures the formation of vascular branches from pre-existing blood vessels when required, maintaining regular homeostasis [83,84]. Insufficient angiogenesis can lead to chronic wounds, ischemia, or hair loss, while excessive angiogenesis is often associated with eye disease, rheumatoid arthritis, or cancer [85].
SA represents the principal mechanism by which neovascularization occurs after birth, even if in the healthy adult it becomes largely quiescent [85,86]. The process turns on/off based on a delicate balance between pro- and anti-angiogenic factors [85]. Numerous mechanical (increasing blood flow, shear stress), chemical (hypoxic conditions, increased levels of nitric oxide), and molecular (growth factors, chemokines CXC-1, -2, -3, -5, -6, -7, and -8, hypoxia-inducible factor, integrins, and others) signals can trigger angiogenesis [87]. On the other hand, there exist main endogenous angiogenesis inhibitors (endostatin, angiostatin, chemokines CXC-4, -9, -10, -11, -12, and -14, thrombospondin, pigment epithelium-derived factor, interferon-α, -β, and -γ), often peptides or protein fragments present in the bloodstream or confined to the extracellular matrix [88,89].
Angiogenesis is a complex and multi-step process structured as follows: (1) angiogenic input; (2) degradation of extracellular matrix and dissolution of capillary basement membrane and activation of quiescent endothelial cells; (3) proliferation and migration of endothelial tip cells (cells located at the tips of the growing vessel that migrate following the VEGF concentration gradient); (4) tubulogenesis (the formation of an endothelial tube-like structure); and (5) the formation of a mature basement membrane with pericytes [83,90,91]. In GBM, with the “angiogenic switch” (Figure 2), the transition from dormant, avascular hyperplasia to the characteristic highly vascularized tumor with exuberant angiogenesis, the balance is disrupted and tips in favor of the pro-angiogenic factors [75,92]. Blood vessel sprouting, caused by an increase in angiogenesis activators, is promoted by hypoxic microenvironments, genetic alterations, and crosstalk between different cell types [63,83].
The key players and the principle molecular signaling pathways orchestrating abnormal angiogenesis in GBM are specifically described below.

3.1.1. Hypoxia and Hypoxia-Inducible Factors

The hypoxic microenvironment—low tumor oxygenation—is the main driver of GBM angiogenesis. In the early stages, the tumor mass, being smaller than 1 mm, takes the necessary oxygen and nutrients from the pre-existing tissue vasculature [93]. However, beyond this size, in line with Folkman’s theory, the resulting hypoxic tumor ecosystem instigates the formation of new blood vessels through a series of genetic changes, necessary to feed the rapidly growing malignancy [93,94].
Hypoxia-inducible factor-1 (HIF-1) is the key mediator of cellular adaptation to hypoxic stress [95]. HIF-1 is a ubiquitously expressed transcription factor whose biological activity revolves around the formation of a heterodimer comprising the oxygen-labile α-subunit and the stable nuclear β-subunit [96]. Under normoxic cellular conditions, cells by default continuously produce but also rapidly degrade HIF1-α. In particular, with good oxygen levels, the prolyl hydroxylase domain (PHD) 1-3 hydroxylates HIF-α at the two proline residues 402 and 564 within the oxygen-dependent degradation domain (ODDD). The hydroxylated HIF1-α is then recognized and polyubiquitinated by Von Hippel–Lindau (VHL) E3 ubiquitin ligase and thus marked for proteasomal degradation. In contrast, under hypoxic conditions, HIF1-α is stabilized and accumulates in the cellular nucleus, where it forms a heterodimer with the β-subunit. Through binding to hypoxia response elements (HREs), HIF-1 ultimately modulates the expression of numerous target genes [95,96]. In GBM, due to the robust proliferation index and the aberrant angiogenesis, a highly hypoxic environment typically occurs [97]. In this context, HIF promotes the transcription of several pro-angiogenic factors, including VEGF, PDGF, angiopoietins, and their cognate receptors, as well as MMPs [95]. High levels of HIF-1 positively correlate with GBM development, metastasis, systemic dissemination, angiogenesis, and resistance to treatments [98,99]. Sometimes, in GBM, HIF can be activated even in normoxic conditions in the case of EGFR amplifications/mutations or in the case of PTEN/TP53 loss [99].

3.1.2. Pro-Angiogenic Factors

VEGF. Vascular endothelial growth factors (VEGFs) portray a family of homodimeric glycoproteins (molecular weight of about 40 kDa) with a cystine-knot motif that includes seven different members: VEGF-A, VEGF-B, VEGF-C, VEGF-D, the placental growth factor (PIGF), viral VEGF-E, and snake venom VEGF-F [100,101,102]. These factors trigger signal transduction cascades, binding to a three-member family of tyrosine kinase receptors with different specificity: vascular endothelial growth factor receptor-1 (VEGFR-1), VEGFR-2, and VEGFR-3 [103]. VEGF-R1 binds to VEGF-A and -B and PIGF, and VEGF-R2 is specific to VEGF-A, -C, and -D, while VEGF-R3 selectively binds to VEGF-C and -D. Furthermore, VEGF receptors perform their biological functions by interacting with different co-receptors, such as neuropilin-1 and -2 and heparan sulfate proteoglycans [104].
Researchers’ attention has mainly focused on the multifunctional cytokine VEGF-A, the most representative regulator of both normal and pathological angiogenesis [105,106,107].
Within a tumor context, various cell types secrete VEGF-A, including cancer cells but also endothelial cells, macrophages, and fibroblasts [107]. Although VEGF-A binds to both VEGFR-1 and VEGFR-2, VEGF-A/VEGFR-2 is the dominant signaling pathway [108]. Several factors produced in response to hypoxic tumor microenvironments (HIFs, PDGF, EGF) or independently of hypoxia can induce VEGF and VEGFR upregulation in GBM [54,108,109]. In particular, high levels of VEGF-A mRNA were detected, especially near necrotic areas in GBM tumors [110]. Furthermore, the VEGF-A expression rate increases from low-grade to high-grade gliomas and negatively correlates with GBM patients’ survival [111,112]. Moreover, GBM exhibits the highest content of VEGF protein, compared with other brain tumors, such as ependymomas, meningiomas, and medulloblastomas [113]. Triggering signaling pathways (such as PI3K/AKT/mTOR, RAS/RAF/MEK/ERK, SRC/FAK, and p38 MAPK), VEGF-A/VEGFR-2 promotes vascular permeability as well as EC proliferation and migration. However, besides stimulating tumor angiogenesis, VEGF-A is also implicated in the growth, survival, and infiltration of tumor cells, particularly in aggressive tumors. Although VEGF mainly acts through paracrine mechanisms by binding to receptors expressed on the tumor vasculature, GBM cancer cells are also able to express VEGFRs, stimulating malignancy progression through autocrine signaling [54,92,113,114]. Furthermore, it has been observed that VEGF-A promotes tumorigenesis and angiogenesis in human GBM cancer stem-like cells, though it is still unclear whether it also stimulates their proliferation and through what mechanisms [115]. The alternative splicing of VEGF-A pre-mRNA allows for the production of various protein isoforms, characterized by different molecular weights, biochemical properties, and biological functions. VEGF-121, the lighter variant with no heparin-binding domain, is the main isoform in circulating blood, believed to be predominantly involved in vascular permeability and less involved in tumor angiogenesis [116,117]. The heavier isoforms, VEGF-A189 and VEGF-A206, are closely anchored to the extracellular matrix by their two heparin-binding domains, serving as a valuable source of VEGF [116,118,119]. Instead, the intermediate-weight VEGF-A165 isoform, which has a single heparin-binding domain, presents in-between ECM-binding properties, showing excellent bioavailability [116,118]. As recently suggested by D’Alessandris et al., GBMs can synthetize all VEGF isoforms. Therefore, since bevacizumab binds to all VEGF splicing products, the sensitivity of each GBM tumor to anti-angiogenic treatment may depend on the relative amount of the various isoforms produced [120]. Martini and colleagues, using a brain xenograft model of human GBM cells, observed that VEGF-A121 plasma levels correlate with tumor size and that VEGF-A121 concentration in peripheral blood markedly decreases after bevacizumab administration. Additionally, if higher levels of circulating VEGF-121 may result in less bevacizumab being available to target the other, more clinically relevant VEGF isoforms, the research group demonstrated that patients with lower baseline VEGF-121 concentrations and lower VEGF-121 reduction after bevacizumab treatment had better prognoses. Therefore, the circulating VEGF-A121 levels could be a potential predictive biomarker for response to bevacizumab in GBM patients [116].
Apart from VEGF-R2, VEGF-R1 under physiological conditions can prevent excessive blood vessels sprouting by acting as a negative regulator, but when upregulated in tumors, it seems to contribute to aberrant angiogenesis [121]. VEGFR-3, instead, normally mediates lymphoid proliferation, but evidence suggests its involvement in tumor metastases. However, more investigations are required in this thematic area [121].
FGF. The human genome encodes 22 polypeptides (molecular weights range from 17 to 34 kDa) that form the broad family of fibroblast growth factors (FGFs), with FGF1 (acid FGF) and FGF2 (basic FGF) being the first members to be identified in the 1970s [122,123,124,125]. FGFs mediate the behaviors (proliferation, differentiation, migration, survival) of various cell types, tissue maintenance, and wound healing through fibroblast growth factor receptor (FGFR) activation [124,126,127]. However, improper FGF/FGFR signaling is also implicated in cancer progression and tumor-induced angiogenesis [128]. Previous studies have widely highlighted FGF upregulation in various tumor contexts [129,130,131,132,133,134,135]. In glioblastoma cases, specifically, increased levels of both FGF2 mRNA and proteins have been reported through Northern blotting, immunohistochemistry, and in situ hybridization.
Moreover, the expression rate of FGF2 was positively correlated with tumor aggressiveness and vascular density [136,137]. Compelling evidence, in fact, shows that FGF2 plays a crucial pro-angiogenic role in GBM [138]. Basically, FGF2, in synergy with VEGF, stimulates endothelial cell replication and migration as well as the proteolysis of the extracellular matrix, necessary for the formation of new blood vessels [139,140,141,142]. Interestingly, FGF inhibitors have shown anti-angiogenic effects in murine models, also leading to an increase in tumor cell apoptosis [143]. Indeed, it has been observed that FGF2 can promote glioma cell survival by upregulating BCL2 and seems to support, together with EGF, the self-renewal of GBM stem-like cells [144,145]. In addition, an in vitro study by Toyoda et al. demonstrated how FGF2, secreted by GBM cells, can improve blood–tumor barrier functionality, contributing to the typical GBM refractoriness to therapies [146]. Furthermore, aberrant FGF2/FGFR signals are also involved in acquired resistance to anti-VEGF drugs in GBM patients, but the related mechanisms remain to be clarified [147].
FGFRs comprise a group of five transmembrane tyrosine kinase receptors, from FGFR1 to FGFR5 (FGFR5 is a truncated receptor, lacking the cytoplasmic tyrosine kinase domain) [148]. Cognate ligand/receptor interactions trigger various intracellular molecular pathways, including RAS/MAPK, PLCγ, PI3K-AKT, and STATs [149,150,151,152]. However, approximately 8% of gliomas harbor FGFR genomic aberrations in the form of activating mutations, amplifications, and structural rearrangements, which contribute to tumor progression. FGFR-TACC fusion and FGFR1/FGFR3 amplifications constitute the most common variants [153,154,155].
TGF-β. The three well-characterized transforming growth factor-β (TGF-β) isoforms (TGF-β 1, -β 2, and -β 3) are 25 kDa cytokines with pleiotropic effects, secreted as inactivate precursors almost ubiquitously in the human body [156]. TGF-βs, by activating three specific membrane receptors (TβRI, TβRII, TβRIII), trigger both Smad-dependent and Smad-independent molecular pathways (PI3K/AKT, RHO, PAR6, ERK, JUNK, p38, NF-κB, and TRAF4/6), leading to different, context-dependent, cellular responses [156,157,158]. Intriguingly, TGF-β shows a dualistic behavior in tumorigenesis, acting as a cancer repressor in healthy tissues/premalignant stages but also turning into a strong promoter of tumor progression and malignancy in advanced disease [159]. Specifically, dysregulated TGF-β signals drive more aggressive tumor phenotypes supporting angiogenesis, infiltration, metastasis, immunosurveillance evasion, and epithelial-to-mesenchymal transition programs [159,160,161,162,163,164,165]. Even in GBM, the so-called “TGF-β paradox” has been widely observed [166,167]. Increased TGF-β has been found in the most aggressive gliomas and represents a poor prognostic marker, both in terms of the PFS and OS [167]. TGF-β, secreted by cancer cells or other cell types in the surrounding microenvironment, also encourages GBM neovascularization, mainly by augmenting the levels of the pro-angiogenic factor insulin-like growth factor-binding protein 7 (IGFBP7) in the tumor endothelial fraction [168]. Furthermore, impaired TGF-β signaling indirectly stimulates endothelial cell proliferation and ECM remodeling in GBMs, increasing VEGF production and MMP functionality, respectively [169,170]. Additionally, cell proliferation through TGF-β/Smad has been highlighted in human glioma cell lines. Indeed, apart from TGF-β, phospho-Smad overexpression has been demonstrated by in vivo studies [171].
PDGF. Initially isolated from human platelet α-granules but also released by other cell types, platelet-derived growth factor (PDGF) acts as a potent stimulator of fibroblast, glial cell, and smooth muscle cell proliferation, apart from being involved in migration and chemotaxis [172,173,174,175,176]. The PDGF family includes four protein chains (PDGF-A, PDGF-B, PDGF-C, PDGF-D) that can variously combine to generate disulfide-bonded homodimers (PDGF-AA, PDGF-BB, PDGF-CC, PDGF-DD) and heterodimers (PDGF-AB), mediating all the biological effects ascribable to PDGF, through PDGFR activation [177,178]. Platelet-derived growth factor receptors (PDGFR-α and PDGFR-β) are single-pass membrane tyrosine kinase glycoproteins with a molecular weight of approximately 180 kDa [179].
After proper stimulation by the ligand, PDGFRs trigger multiple downstream signaling cascades, including PI3K/AKT, JAK/STAT, the Notch pathway, and MAPK/ERK [180,181]. Aberrant PDGF signaling networks show tumorigenic potential in a variety of tumor forms, contributing to progression, metastasis, angiogenesis, the EMT, and anti-VEGF escape [182,183,184,185]. Both ligands and their receptors were found to be upregulated in gliomas. About 20% of GBMs exhibit genetic alterations in PDGF/PDGFR signaling, commonly PDGFs activating mutations/chromosomal rearrangements or PDGFR amplifications/overexpression [182,186,187]. Mounting evidence demonstrates that PDGF-B is the main factor involved in promoting GBM angiogenesis by stimulation through its receptor PDGFR-β, pericyte recruitment, and increasing VEGF levels in the developing tumor vasculature [188]. Furthermore, in GBM tissues, in situ hybridization analyses have revealed the enhanced expression of both PDGF-B and PDGFR-β mRNA in the proliferating endothelial component, particularly in glomeruloid structures and small capillaries, suggesting the existence of autocrine angiogenesis-stimulating loops [189]. GCSCs express PDGFR-β more frequently than PDGFR-α. In GBM cell lines, the genetic or pharmacological inhibition of PDGFR-β has been proven to cause a lowering of GSCS self-renewal potential and a decrease in GBM progression [186]. Instead, PDGFR-α targeting in vitro promotes apoptosis in GCSCs resistant to anti-Notch/anti-EGFR therapy [190]. It has also been suggested that PDGF-α/β inhibition with CP-673451 both attenuates proliferation/invasion rates and stimulates differentiation in GBM/GCSCs cell lines via DUSP1/p38 MAPK [191].
Angiopoietins. Originally discovered in 1996, angiopoietins constitute a small class of four endothelial growth factors, angiopoietin-1 (Ang1), Ang2, Ang3 (in mouse), and Ang4 (in humans), which structurally share an amino-terminal “superclustering” region, followed by a coiled-coil motif involved in multimerization and a carboxy-terminal fibrinogen-like domain necessary for receptor binding [192,193,194]. All angiopoietins act through Tie-2 (also known as TEK), a tyrosine kinase receptor specifically expressed on endothelial cells and their precursors [195,196,197]. The Tie family also includes Tie-1, a poorly defined orphan receptor that presumably affects Ang1-Ang2/Tie-2 signaling, modulating both physiological and pathological neovascularization [198,199].
Ang1 is a natural TEK agonist, essentially involved in vascular maturation and quiescence, whereas Ang2 functions as a context-specific agonist or antagonist of TEK, promoting angiogenesis or blood vessel destabilization/cell death in the presence or absence of VEGF-A, respectively [193,200,201]. Interestingly, the angiopoietin/Tie2 ligand/receptor system is frequently perturbed in cancer patients. It has been remarked that an altered expression profile of Angs can intensify the aggressiveness of ovarian cancer, lung carcinoma, and human gliomas, while their contribution to tumor angiogenesis is not well defined [202,203,204,205]. Various studies conducted on this research topic have revealed conflicting data, reporting, in some cases, pro-angiogenic roles and, in other cases, anti-vascular effects of upregulated Ang1 or Ang2 [206]. In GBM, the Ang/Tie-2 axis presumably contributes to aberrant angiogenesis [206]. Furthermore, Northern blot and in situ hybridization analyses showed the differential expression of Ang ligands in human GBM tissue samples: the upregulation of Ang1 was found in tumor cells, while Ang2 mRNA was detected only in the endothelial cells within the tumor mass but exclusively in correspondence with small capillaries. In addition, immunohistochemistry indicated enhanced levels of Tie-2 receptor in tumor vasculature, compared to in healthy brain tissue, in a rat glioma model [207]. Moreover, apart from endothelial cells, GBM tumor cells are also capable of expressing the Tie-2 receptor [208]. The exact biological significance of this imbalanced pathway in GBM has yet to be elucidated. However, it has been observed that a double pharmacological blockade of VEGF and Ang2 in two murine GBM models (Gl261 and U87) allowed the normalization of tumor vasculature and interfered with malignancy progression, leading to better prognoses [209]. Other experts have highlighted that the protein levels of Ang4 are also enhanced in human GBM tissues. Being implicated in tumor angiogenesis and development, it could be a potential future therapeutic target [208].

3.1.3. Proteinases

MMPs. The family of matrix metalloproteinases (MMPs) includes a large collection of endopeptidases with different substrate specificity, which specifically require zinc and calcium to perform their enzymatic activity [210]. Described for the first time in the tadpole, MMPs have rapidly emerged as the group of proteases mainly involved in ECM remodeling [211]. Collectively, by cleaving internal peptide bonds, MMPs are able to break down the majority of protein components constituting connective tissues such as collagen, elastin, fibronectin, gelatin, and casein [211]. ECM degradation is a crucial step both during embryonic development and in postnatal life. However, apart from mediating several physiological processes, dysregulated MMP activities are commonly found in pathological states, including tumor neovascularization, invasion, and spread [212,213].
Basically, MMPs augment angiogenesis through multiple mechanisms spanning from increasing the bioavailability of ECM-bound pro-angiogenic mediators to facilitating endothelial cell migration and pericyte detachment from growing vessels [214].
Comparing different cancer types, RNA-seq analyses have recently outlined a highly heterogeneous MMP expression, showing overall enhanced levels of MMP1, MMP9, MMP10, MMP11, and MMP13 but the downregulation of MMP2, MMP7, MMP23B, MMP27, and MMP28 [213]. The potential role of MMPs in glioma pathogenesis has also been investigated. In this respect, accumulating data have highlighted an increased expression of MMP1, MMP-9, MMP-11, and MMP-19 in GBM tissues, both at the mRNA and protein levels. Upregulated MMP-1 and MMP-9 negatively correlate with the patient’s prognosis, while MMP-11 overexpression seems to be associated with a more aggressive GBM phenotype [215,216]. Furthermore, it has been observed that an unbalanced MMP9 activity elevates the rate of cell proliferation and the clonogenic potential in U87 GBM cell lines [217]. Moreover, a randomized phase III study has shown that newly diagnosed GBM patients with low MMP9 plasma levels respond better to bevacizumab, with an improved OS, reinforcing the hypothesis of its concrete involvement in GBM angiogenesis [218]. The therapeutic value of conventional chemotherapy combined with a broad-spectrum MMP inhibitor in the treatment of GBM has recently been studied in some clinical trials, with encouraging data [219].

3.2. Vasculogenesis

During embryo development, the primitive vascular system originates from the ex novo assembly of endothelial precursor cells derived from the mesoderm, during a process known as vasculogenesis [220,221]. For a long time, it has been considered a mechanism exclusive to the intrauterine period [222]. Even in adult life, in reality, different scenarios require vasculogenesis activation like wound healing, stroke, ischemia, or tumor development [223]. Bone marrow, hematopoietic stem cells, myeloid cells, and tissue-resident cells (adipose, cardiac, dental, or neural tissue) can all be putative sources of adult endothelial progenitor cells (EPCs). Subsequent to GBM’s recruitment of EPCs, they predominantly occupy the hypoxic niches and start to differentiate into mature endothelial cells, initiating the formation of new blood vessels [222,224,225]. In GBM patients, as with other tumor types, the circulating levels of EPCs were found to be elevated compared to in healthy individuals. Furthermore, it has been shown that an increase in glioma malignancy grade is positively correlated with a higher fraction of VEGF-R2+ cells [226]. Moreover, EPC recruitment, which appears more prominent in GSC-enriched GBM tumors, is also further intensified after radiotherapy treatment. In a radiotherapy-treated GBM xenograft model, it was observed that the tumor was able to recover its damaged endothelial fraction through the induction of vasculogenesis. Both HIF-1 (activated by hypoxia and reoxygenation after radiotherapy) and stromal cell-derived factor-1 (SDF-1), along with its receptor CXCR4, play a crucial role in recruiting bone marrow-derived cells (BMDCs) to the tumor [227,228,229,230]. Through the pharmacological targeting of HIF-1 or the inhibition of CXCR4 with neutralizing antibodies, the influx of BMDCs was interrupted, contrasting tumor recovery. These results reiterate the need to target GBM on multiple fronts, also considering the existence of vasculogenesis as a compensatory mechanism of neovascularization exploited by the tumor [231].

3.3. Vessel Co-Option

That cancerous cells can co-opt a pre-existing vascular system has been demonstrated, for the first time, in a rat C6 glioma model. Tumors affecting the brain, liver, and lungs, organs characterized by a high vascularization level, show the phenomenon more commonly. Specifically, during blood vessel co-option, tumor cells, given their infiltrating ability, penetrate the healthy tissue and migrate along the pre-existing capillaries, eventually wrapping them and guiding them into the tumor mass so that host vasculature-dependent proliferation can start [82,232]. Recent studies suggest that the vessel co-option (VC) mechanism can be intrinsic to the tumor or eventually exacerbated by anti-angiogenetic therapies/chemoradiotherapy, allowing GBM to recur inexorably [233,234].
There is currently no detailed information on the vessel co-option modus operandi due to the challenges related to the longitudinal observation of VC in vivo and the lack of adequate in vitro models [235]. Ang-2, IL-8, CDC42, CXCR4/SDF1α, bradykinin, ephrinB2 (only when upregulated), and Olig2/Wnt7a have emerged as some of the key players in driving GBM vessel co-option [234].

3.4. Vascular Intussusception

Vascular intussusception (VI) takes place when a pre-existing blood vessel splits into two functional blood vessels. Vascular remodeling occurs through blood vessel invagination followed by the formation of intraluminal intussusceptive pillars that ultimately create a double lumen [77,79]. The existence of intussusceptive microvascular growth as one of the potential backup mechanisms to ensure tumor perfusion has emerged in GBM but also in melanoma, colon cancer, breast carcinoma, and B-cell non-Hodgkin’s lymphoma [236]. An angiogenic switch from sprouting to intussusception associated with malignancy recovery has been reported after ionizing radiation or anti-VEGF treatment (PTK787/ZK222854) in tumor xenografts in nude mice [237]. Compared to canonical neovascularization, VI requires less energy and time consumption [236]. In addition, newly formed blood vessels are less permeable and appear more refractory to anti-angiogenic inhibitors. Every aspect is particularly useful for GBM progression [236,238]. The mediators leading this process have not yet been identified [77].

3.5. Vasculogenic Mimicry

Under certain conditions, GBM aggressive tumor cells, marked by high plasticity, organize spatially to form vessel-like channels that are then connected with the pre-existing vascular system in order to provide the tumor mass with enough nutrients and oxygen [239,240]. First discovered in melanoma, two different vascular mimicry (VM) modalities have been identified in tumors so far: the vasculogenic mimicry of the patterned matrix mode and the vasculogenic mimicry of the tubular mode [239,241]. In the first type, the channels have a completely distinct morphology compared to endothelial blood vessels, while in the second case, vasculogenic-like structures closely resemble capillaries [241]. Both strategies have been observed in GBM, and the incidence of VM is often associated with resistance to anti-angiogenic drugs and worse prognoses [240,241,242,243,244]. Indeed, the research group of Angara et al. has previously outlined increased VM in GBM following anti-angiogenic treatments (Vatalanib), supporting the hypothesis that it is an alternative neovascularization mechanism, mainly induced by hypoxic conditions but aggravated by angiogenesis inhibitors [239,245]. The so-called tumor-derived endothelial cells (TDECs)—mature GBM tumor cells trans-differentiated into cells with an endothelial phenotype—play a key role in promoting VM [246].

3.6. Trans-Differentiation of Cancer Stem-Like Cells

Recent research efforts have highlighted how the aggressive behavior of GBM is largely attributable to GBM stem-like cells (GSCs), a small fraction of cancerous cells, with high plasticity and self-renewal capacity, involved in tumor initiation, maintenance, and progression [247,248]. Cancer stem-like cells (CSCs) were first identified at the end of the 20th century. Bonnet and Dick, indeed, were pioneers in isolating CSCs from acute myelogenous leukemia [249]. Since then, CSCs have become a hotspot on which many research groups have focused. Their discovery has certainly provided new insights into understanding tumor biology and eventually finding novel molecular biomarkers.
GSCs, typically located near hypoxic perivascular niches, show a slow-growing status and a marked resistance to conventional treatments (both chemo- and radiotherapy) [250]. Endowed with stemness properties, GSCs exhibit a high differentiation potential, causing extensive tumor heterogeneity, a hallmark of GBM [250]. It is now widely accepted that GSCs can also trans-differentiate into an endothelial lineage, directly contributing to new blood vessel formation [251]. In support of this, Ricci-Vitiani et al. found that, on average, 60% of GBM endothelial cells showed the same chromosomal aberrations as those reported in GBM tumor cells, suggesting a cancerous origin [252]. Furthermore, they observed trans-differentiation both in vitro, with the isolation of CD133+/CD31− GBM cells capable of obtaining an endothelial phenotype, and in vivo using a xenograft model [253].
Moreover, the work of De Pascalis et al. has shown that in GBM recurring after radiotherapy, the cerebral endothelium undergoes radiation-induced senescence, and the fraction of tumor-derived cells with an endothelial phenotype increases by a factor of 2 to 2.7 compared to tumors from primary surgery. Therefore, GSCs likely play a compensatory role by supporting the neovascularization of the brain endothelium undergoing radiation-induced senescence [254]. This GSC-associated neovascularization mechanism seems closely connected to hypoxia and occurs independently of the VEGF/VEGFR signaling pathway. Therefore, bevacizumab, or other drugs that target the same pathway, have not proven to be very effective at blocking the trans-differentiation of GSCs into GSC-derived endothelial cells (GdECs) [253]. However, the efficacy of 349 different compounds in inducing cell death in both GSC and GdEC cell lines was comprehensively evaluated, with elesclomol (STA-4783) emerging as the most effective. Elesclomol compromises the survival of both GSCs and GdECs, inducing a significant increase in mitochondrial reactive oxygen species, leading to non-apoptotic, copper-dependent cell death. Furthermore, as confirmed both in vitro and in vivo, the combination of elesclomol and TMZ enhances cytotoxicity compared to TMZ alone [255].

4. Treating GBM: Between Pitfalls and Emerging Therapies

The current lack of successful therapies represents one of the major limitations in GBM management. Several aspects contribute to making treatment options insufficient. Firstly, removing the entire tumor is tricky given its critical location and highly infiltrative nature. Even when the mass appears completely excised, GBM invariably comes back. Furthermore, the presence of the blood–brain barrier (BBB) significantly obstructs effective CNS drug delivery [256]. Most GBM patients are also intrinsically refractory to chemo-/radiotherapy or develop resistance at a later time, a phenomenon primarily driven by CSCs [257,258]. In addition, both the great intra-tumor heterogeneity and the marked immunosuppressive microenvironment are closely related to the failure of current treatments [259,260]. Moreover, aberrant angiogenesis and the coexistence of multiple modes of neovascularization complicate the effectiveness of anti-angiogenic therapies [261].
The therapeutic landscape in the GBM setting is, however, currently evolving, and immense efforts have been directed towards overcoming the Stupp protocol, the gold-standard treatment to date, through the development of more promising therapeutic approaches [262].
Bevacizumab (BEV), or Avastin, is a recombinant monoclonal IgG1 antibody, humanized, specifically directed against VEGF-A, typically upregulated in GBM [63,263,264,265]. Based on data from two phase II studies, BRAIN and NCI 06-C-0064E, it was FDA-approved in 2009 as a single agent for the treatment of recurrent glioblastoma, but its concrete ability to improve the prognosis remains to be clarified [266,267]. In particular, in the BRAIN trial, 167 patients with GBM in their first or second relapse were randomly assigned to receive BEV, alone or in combination with Irinotecan (a topoisomerase I inhibitor). The 6-month PFS rates were 42.6% and 50.3%, respectively, and the median OS rates were 9.2 months and 8.7 months, respectively. Moreover, the group receiving BEV plus Irinotecan experienced an increased incidence of grade 3 or higher treatment-related adverse events compared to the BEV-alone group [268]. In the 06-C-0064E study, a single-center experience conducted by the National Cancer Institute (NCI), instead, a cohort of 48 patients with recurrent GBM was treated with BEV monotherapy until progression, resulting in a 6-month PFS of 29% and a median OS of 31 weeks [269].
Given the single-agent treatment failure, the spotlight has shifted to the possible application of polytherapy. The combination of two or more anti-cancer drugs, indeed, has recently emerged as a novel promising angle to explore, aiming to better prognoses and increase therapeutic potential while reducing side effects [270].
In this respect, different clinical trials have examined the efficacy and toxicity of BEV together with other chemotherapeutic drugs or radiotherapy, highlighting improvements in the OS and/or PFS [271,272,273,274]. For example, the potential of BEV or Lomustine as single agents versus a combination of BEV plus Lomustine was evaluated in a group of 153 patients with recurrent GBM, in a randomized controlled phase II study called the BELOB trial. The primary end point was a 9-month OS. The main secondary end points were the median PFS, median OS, objective response rate, and association of the outcome with MGMT (O6-methylguanine-DNA methyltransferase) promoter methylation status and IDH status. Overall, the 6-month PFS and the median OS were 16% and 8 months in the BEV group, 13% and 8 months in the Lomustine group, and 42% and 12 months in the BEV plus Lomustine group, respectively. The safety analysis showed that, after a reduction in the Lomustine dose, the combination treatment was generally well tolerated, suggesting promising prospects for the use of BEV integrated with Lomustine [273]. Later, following the findings of the BELOB study, the EORTC (European Organization for Research and Treatment of Cancer) 26101 phase III trial enrolled a total of 437 patients with recurrent GBM, of whom 149 underwent Lomustine treatment alone, and 238 received Lomustine plus BEV for a period of 37 months. Despite a longer PFS in the group treated with the combination therapy (4.2 months vs. 1.5 months), there was no significant difference in the median OS between the two cohorts (9.1 months in the combination group and 7.6 months in the monotherapy group) [266,275]. More precisely, in the RTOG 0825 trial, 637 newly diagnosed GBM patients were randomly assigned to receive either BEV or a placebo, in addition to TMZ and radiotherapy. While the study suggested an improvement in the PFS (10.7 months in the BEV group compared to 7.3 months in the placebo group), it showed no evidence of a survival benefit. The median OS was, indeed, 15.7 months in the BEV group and 16.1 months in the placebo group, respectively [263]. Based on the same rationale, a second placebo-controlled study, called the AVAglio trial, enrolled a total of 921 newly diagnosed GMB patients who were randomly assigned to the BEV group (458 patients) or to the placebo group (463 patients). The addition of BEV to TMZ + radiotherapy prolonged the PFS (8.4 months in the BEV group, 4.3 months in the placebo group) and extended the baseline performance status, along with decreasing the glucocorticoid demand. Conversely, the duration of OS observed did not significantly differ between the two cohorts (16.8 months in the BEV group compared to 16.7 months in the placebo group) [276]. However, AVAglio patients were then retrospectively evaluated for molecular subtype, and the group with proneural GBM actually showed an increase in OS after treatment with Bevacizumab added to first-line therapy [277].
The potential role of alternative angiogenic inhibitors, including monoclonal antibodies (Carotuximab anti-endoglin), tyrosine kinase inhibitors involved in angiogenesis (Regorafenib: anti-VEGFR1,2,1, FGFR1, PDGFR-β, Sorafenib anti-VEGFR2/3, PDGFR-β, Raf kinase, and FLT3; Sunitinib: anti-VEGFR2, PDGFR-α/β, and KIT; Cediranib: a pan-VEGFR inhibitor; and others), and recombinant fusion proteins (Aflibercept, a decoy receptor anti-VEGF), either as monotherapy or in combination with other anti-cancer drugs, is being investigated in numerous ongoing phase I, II, or III clinical studies, widely described in the current literature with promising preliminary results [42,278,279,280,281,282,283,284,285,286,287,288].
The introduction of immunotherapy in clinical practice has changed the management of previously difficult-to-treat solid tumors. However, immunotherapy in GBM has not led to significant improvements in clinical outcomes. GBM is a cold tumor, capable of implementing various immune escape strategies [260,289]. Nivolumab (anti-PD-1), Pembrolizumab (anti-PDL-1), and Ipilimumab (anti-CTLA4) are among the main immune checkpoint inhibitors tested as single agents in several clinical trials against recurrent or newly diagnosed GBM but with marginal clinical benefits [260]. However, the therapeutic efficacy of immune checkpoint inhibitors combined with anti-angiogenic drugs or with vaccine-based/oncolytic virus-based/CAR-T-based approaches is currently under critical evaluation [260,290].
The details of the most relevant clinical trials involving anti-angiogenic drugs, used either alone or as part of a polytherapy regimen, in patients with recurrent or newly diagnosed GBM, are provided in Table 1.
Questions regarding the optimal dosing regimen of BEV and other emerging drugs for GBM management remain unresolved. While maximizing the likelihood of a response, it is equally important to minimize therapy-related toxicity. However, the persistently high incidence of drug-related side effects continues to be a significant limitation and requires further investigation. From this perspective, for example, a phase II, comparative, randomized, single-center clinical trial was specifically performed to compare the efficacy and safety of low-dose BEV (5 mg/kg) plus Lomustine versus standard-dose BEV (10 mg/kg) alone in 69 adults with recurrent GBM. The results showed a trend towards a longer median PFS in the low-dose BEV + Lomustine arm in patients with their first relapse [291].
Furthermore, the presence of the blood–brain barrier, due to its highly protective nature, complicates proper drug delivery to the brain, possibly resulting in suboptimal concentrations of therapeutic agents at the tumor site and necessitating the development of strategies to enhance drug penetration and reduce off-target effects [292].
Ultimately, one of the key challenges for the future will be the identification of effective tumor-specific biomarkers and their integration with the assessment of each GBM’s genetic signature, to predict which patient subgroups could benefit most from novel anti-angiogenic therapies, either alone or in combination with other drugs. In this context, new research avenues are currently exploring the potential of various angiogenic regulators as biomarkers (such as lower concentrations of VEGF-A121 as a predictive circulating biomarker for response to BEV or downregulated HIF1-α as a predictive tissue biomarker for response to TMZ), in addition to identifying potential GSC biomarkers (such as CD133, CD44, SOX2, CD15, CD70, ALDH3A1, and nestin) for the development of targeted and personalized therapies [116,293,294].

5. Conclusions

GBM is hallmarked by a singular vascular bed, in terms of morphology, architecture, and functionality, which reflects a complex underlying biology. Tumor aspects like multiple pro-angiogenic mediators and pathways, alternative strategies of neovascularization, high intra- and inter-tumor heterogeneity, and strong resilience to current treatments emphasize the imperative need to design novel standards of care in the fight against GBM, specifically tailored to each patient’s molecular signature, which may impact various aspects of multifaceted neovascularization. Could combination therapies, corroborated by a more detailed comprehension of GBM’s intricate landscape, represent valid options to improve patients’ clinical outcomes and their healthy lifespan in the near future?

Author Contributions

M.B. (Mariagiovanna Ballato) and M.M. are the principal authors and main contributors to writing this manuscript. E.G., W.G.G., G.R., M.B. (Mariachiara Buccarelli), V.F. and G.C. reviewed the literature. L.R.-V., Q.G.D., C.P., G.G., V.Z. and G.F. have read and revised this manuscript. P.T., A.I. and M.C. revised the English language. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Acknowledgments

We thank Rosa Scarfì for her technical support.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ah-Pine, F.; Khettab, M.; Bedoui, Y.; Slama, Y.; Daniel, M.; Doray, B.; Gasque, P. On the Origin and Development of Glioblastoma: Multifaceted Role of Perivascular Mesenchymal Stromal Cells. Acta Neuropathol. Commun. 2023, 11, 104. [Google Scholar] [CrossRef]
  2. Loras, A.; Gonzalez-Bonet, L.G.; Gutierrez-Arroyo, J.L.; Martinez-Cadenas, C.; Marques-Torrejon, M.A. Neural Stem Cells as Potential Glioblastoma Cells of Origin. Life 2023, 13, 905. [Google Scholar] [CrossRef] [PubMed]
  3. Youssef, G.; Miller, J.J. Lower Grade Gliomas. Curr. Neurol. Neurosci. Rep. 2020, 20, 21. [Google Scholar] [CrossRef] [PubMed]
  4. Zong, H.; Parada, L.F.; Baker, S.J. Cell of Origin for Malignant Gliomas and Its Implication in Therapeutic Development. Cold Spring Harb. Perspect. Biol. 2015, 7, a020610. [Google Scholar] [CrossRef]
  5. Weller, M.; Wen, P.Y.; Chang, S.M.; Dirven, L.; Lim, M.; Monje, M.; Reifenberger, G. Glioma. Nat. Rev. Dis. Primers 2024, 10, 33. [Google Scholar] [CrossRef]
  6. Garcia, M.R.; Feng, Y.; Vasudevaraja, V.; Galbraith, K.; Serrano, J.; Thomas, C.; Radmanesh, A.; Hidalgo, E.T.; Harter, D.H.; Allen, J.C.; et al. Clinical, Pathological, and Molecular Characteristics of Diffuse Spinal Cord Gliomas. J. Neuropathol. Exp. Neurol. 2022, 81, 865–872. [Google Scholar] [CrossRef]
  7. Aggarwal, P.; Luo, W.; Pehlivan, K.C.; Hoang, H.; Rajappa, P.; Cripe, T.P.; Cassady, K.A.; Lee, D.A.; Cairo, M.S. Pediatric versus Adult High Grade Glioma: Immunotherapeutic and Genomic Considerations. Front. Immunol. 2022, 13, 1038096. [Google Scholar] [CrossRef]
  8. Wang, G.-M.; Cioffi, G.; Patil, N.; Waite, K.A.; Lanese, R.; Ostrom, Q.T.; Kruchko, C.; Berens, M.E.; Connor, J.R.; Lathia, J.D.; et al. Importance of the Intersection of Age and Sex to Understand Variation in Incidence and Survival for Primary Malignant Gliomas. Neuro Oncol. 2022, 24, 302–310. [Google Scholar] [CrossRef]
  9. Mo, Z.; Xin, J.; Chai, R.; Woo, P.Y.; Chan, D.T.; Wang, J. Epidemiological Characteristics and Genetic Alterations in Adult Diffuse Glioma in East Asian Populations. Cancer Biol. Med. 2022, 19, 1440. [Google Scholar] [CrossRef]
  10. Persaud-Sharma, D.; Burns, J.; Trangle, J.; Castro, G.; Barengo, N.; Moulik, S.; Manuel Lozano, J. Demographic Variation in the Frequency of Gliomas in Florida. Medicina 2019, 55, 5. [Google Scholar] [CrossRef]
  11. Ostrom, Q.T.; Egan, K.M.; Nabors, L.B.; Gerke, T.; Thompson, R.C.; Olson, J.J.; LaRocca, R.; Chowdhary, S.; Eckel-Passow, J.E.; Armstrong, G.; et al. Glioma risk associated with extent of estimated European genetic ancestry in African-Americans and Hispanics. Int. J. Cancer 2019, 146, 739. [Google Scholar] [CrossRef] [PubMed]
  12. Frosina, G.; Casella, C.; Puppo, A.; Marani, E.; Campanella, D.; Boni, L.; Fontana, V. Epidemiology of Malignant Brain Tumors in Genova, Italy. 1993–2017. Sci. Rep. 2024, 14, 27300. [Google Scholar] [CrossRef]
  13. Mesti, T.; Ocvirk, J. Malignant Gliomas: Old and New Systemic Treatment Approaches. Radiol. Oncol. 2016, 50, 129. [Google Scholar] [CrossRef] [PubMed]
  14. Daswani, B.; Khan, Y. Insights into the Role of Estrogens and Androgens in Glial Tumorigenesis. J. Carcinog. 2021, 20, 10. [Google Scholar] [CrossRef]
  15. Rhun, E.L.; Weller, M. Sex-Specific Aspects of Epidemiology, Molecular Genetics and Outcome: Primary Brain Tumours. ESMO Open 2020, 5, e001034. [Google Scholar] [CrossRef]
  16. Stabellini, N.; Krebs, H.; Patil, N.; Waite, K.; Barnholtz-Sloan, J.S. Sex Differences in Time to Treat and Outcomes for Gliomas. Front. Oncol. 2021, 11, 630597. [Google Scholar] [CrossRef]
  17. Davis, M.E. Epidemiology and Overview of Gliomas. Semin. Oncol. Nurs. 2018, 34, 420–429. [Google Scholar] [CrossRef]
  18. Ostrom, Q.T.; Kinnersley, B.; Armstrong, G.; Rice, T.; Chen, Y.; Wiencke, J.K.; McCoy, L.S.; Hansen, H.M.; Amos, C.I.; Bernstein, J.L.; et al. Age-Specific Genome-Wide Association Study in Glioblastoma Identifies Increased Proportion of ‘Lower Grade Glioma’-like Features Associated with Younger Age. Int. J. Cancer 2018, 143, 2359–2366. [Google Scholar] [CrossRef]
  19. Krigers, A.; Demetz, M.; Thomé, C.; Freyschlag, C.F. Age Is Associated with Unfavorable Neuropathological and Radiological Features and Poor Outcome in Patients with WHO Grade 2 and 3 Gliomas. Sci. Rep. 2021, 11, 17380. [Google Scholar] [CrossRef]
  20. Choi, D.-J.; Armstrong, G.; Lozzi, B.; Vijayaraghavan, P.; Plon, S.E.; Wong, T.C.; Boerwinkle, E.; Muzny, D.M.; Chen, H.-C.; Gibbs, R.A.; et al. The Genomic Landscape of Familial Glioma. Sci. Adv. 2023, 9, eade2675. [Google Scholar] [CrossRef]
  21. Ostrom, Q.T.; Adel Fahmideh, M.; Cote, D.J.; Muskens, I.S.; Schraw, J.M.; Scheurer, M.E.; Bondy, M.L. Risk Factors for Childhood and Adult Primary Brain Tumors. Neuro-Oncology 2019, 21, 1357–1375. [Google Scholar] [CrossRef] [PubMed]
  22. Kinnersley, B.; Houlston, R.S.; Bondy, M.L. Genome-Wide Association Studies in Glioma. Cancer Epidemiol. Biomark. Prev. 2018, 27, 418–428. [Google Scholar] [CrossRef] [PubMed]
  23. Kinnersley, B.; Labussière, M.; Holroyd, A.; Di Stefano, A.-L.; Broderick, P.; Vijayakrishnan, J.; Mokhtari, K.; Delattre, J.-Y.; Gousias, K.; Schramm, J.; et al. Genome-Wide Association Study Identifies Multiple Susceptibility Loci for Glioma. Nat. Commun. 2015, 6, 8559. [Google Scholar] [CrossRef]
  24. Segura, P.P.; Quintela, N.V.; García, M.M.; Berrón, S.d.B.; Sarrió, R.G.; Gómez, J.G.; Castaño, A.G.; Martín, L.M.N.; Rubio, O.G.; Losada, E.P. SEOM-GEINO Clinical Guidelines for High-Grade Gliomas of Adulthood (2022). Clin. Transl. Oncol. 2023, 25, 2634. [Google Scholar] [CrossRef]
  25. Giannopoulou, A.-I.; Kanakoglou, D.S.; Piperi, C. Transcription Factors with Targeting Potential in Gliomas. Int. J. Mol. Sci. 2022, 23, 3720. [Google Scholar] [CrossRef]
  26. Toader, C.; Eva, L.; Costea, D.; Corlatescu, A.D.; Covache-Busuioc, R.-A.; Bratu, B.-G.; Glavan, L.A.; Costin, H.P.; Popa, A.A.; Ciurea, A.V. Low-Grade Gliomas: Histological Subtypes, Molecular Mechanisms, and Treatment Strategies. Brain Sci. 2023, 13, 1700. [Google Scholar] [CrossRef]
  27. Byun, Y.H.; Park, C.-K. Classification and Diagnosis of Adult Glioma: A Scoping Review. Brain NeuroRehabilitation 2022, 15, e23. [Google Scholar] [CrossRef]
  28. Nafe, R.; Porto, L.; Samp, P.-F.; You, S.-J.; Hattingen, E. Adult-Type and Pediatric-Type Diffuse Gliomas: What the Neuroradiologist Should Know. Clin. Neuroradiol. 2023, 33, 611–624. [Google Scholar] [CrossRef]
  29. Komori, T. Grading of Adult Diffuse Gliomas According to the 2021 WHO Classification of Tumors of the Central Nervous System. Lab. Investig. 2022, 102, 126–133. [Google Scholar] [CrossRef]
  30. Park, Y.W.; Vollmuth, P.; Foltyn-Dumitru, M.; Sahm, F.; Ahn, S.S.; Chang, J.H.; Kim, S.H. The 2021 WHO Classification for Gliomas and Implications on Imaging Diagnosis: Part 1—Key Points of the Fifth Edition and Summary of Imaging Findings on Adult-Type Diffuse Gliomas. J. Magn. Reson. Imaging 2023, 58, 677–689. [Google Scholar] [CrossRef]
  31. Fekete, B.; Werlenius, K.; Tisell, M.; Pivodic, A.; Smits, A.; Jakola, A.S.; Rydenhag, B. What Predicts Survival in Glioblastoma? A Population-Based Study of Changes in Clinical Management and Outcome. Front. Surg. 2023, 10, 1249366. [Google Scholar] [CrossRef] [PubMed]
  32. Luo, C.; Song, K.; Wu, S.; Hameed, N.U.F.; Kudulaiti, N.; Xu, H.; Qin, Z.-Y.; Wu, J.-S. The Prognosis of Glioblastoma: A Large, Multifactorial Study. Br. J. Neurosurg. 2021, 35, 555–561. [Google Scholar] [CrossRef] [PubMed]
  33. Marenco-Hillembrand, L.; Wijesekera, O.; Suarez-Meade, P.; Mampre, D.; Jackson, C.; Peterson, J.; Trifiletti, D.; Hammack, J.; Ortiz, K.; Lesser, E.; et al. Trends in Glioblastoma: Outcomes over Time and Type of Intervention: A Systematic Evidence Based Analysis. J. Neurooncol. 2020, 147, 297–307. [Google Scholar] [CrossRef] [PubMed]
  34. Hatoum, A.; Mohammed, R.; Zakieh, O. The Unique Invasiveness of Glioblastoma and Possible Drug Targets on Extracellular Matrix. Cancer Manag. Res. 2019, 11, 1843–1855. [Google Scholar] [CrossRef]
  35. Goenka, A.; Tiek, D.; Song, X.; Huang, T.; Hu, B.; Cheng, S.-Y. The Many Facets of Therapy Resistance and Tumor Recurrence in Glioblastoma. Cells 2021, 10, 484. [Google Scholar] [CrossRef]
  36. Bonosi, L.; Marrone, S.; Benigno, U.E.; Buscemi, F.; Musso, S.; Porzio, M.; Silven, M.P.; Torregrossa, F.; Grasso, G. Maximal Safe Resection in Glioblastoma Surgery: A Systematic Review of Advanced Intraoperative Image-Guided Techniques. Brain Sci. 2023, 13, 216. [Google Scholar] [CrossRef]
  37. Tomar, M.S.; Kumar, A.; Srivastava, C.; Shrivastava, A. Elucidating the Mechanisms of Temozolomide Resistance in Gliomas and the Strategies to Overcome the Resistance. Biochim. Biophys. Acta Rev. Cancer 2021, 1876, 188616. [Google Scholar] [CrossRef]
  38. Brown, N.F.; Ottaviani, D.; Tazare, J.; Gregson, J.; Kitchen, N.; Brandner, S.; Fersht, N.; Mulholland, P. Survival Outcomes and Prognostic Factors in Glioblastoma. Cancers 2022, 14, 3161. [Google Scholar] [CrossRef]
  39. Mosteiro, A.; Pedrosa, L.; Ferrés, A.; Diao, D.; Sierra, À.; González, J.J. The Vascular Microenvironment in Glioblastoma: A Comprehensive Review. Biomedicines 2022, 10, 1285. [Google Scholar] [CrossRef]
  40. Tu, J.; Liang, H.; Li, C.; Huang, Y.; Wang, Z.; Chen, X.; Yuan, X. The Application and Research Progress of Anti-Angiogenesis Therapy in Tumor Immunotherapy. Front. Immunol. 2023, 14, 1198972. [Google Scholar] [CrossRef]
  41. Mukherjee, A.; Madamsetty, V.S.; Paul, M.K.; Mukherjee, S. Recent Advancements of Nanomedicine towards Antiangiogenic Therapy in Cancer. Int. J. Mol. Sci. 2020, 21, 455. [Google Scholar] [CrossRef] [PubMed]
  42. Schulte, J.D.; Aghi, M.K.; Taylor, J.W. Anti-Angiogenic Therapies in the Management of Glioblastoma. Chin. Clin. Oncol. 2021, 10, 37. [Google Scholar] [CrossRef] [PubMed]
  43. Cohen, M.H.; Shen, Y.L.; Keegan, P.; Pazdur, R. FDA Drug Approval Summary: Bevacizumab (Avastin) as Treatment of Recurrent Glioblastoma Multiforme. Oncologist 2009, 14, 1131–1138. [Google Scholar] [CrossRef] [PubMed]
  44. Fisher, J.P.; Adamson, D.C. Current FDA-Approved Therapies for High-Grade Malignant Gliomas. Biomedicines 2021, 9, 324. [Google Scholar] [CrossRef]
  45. Sener, U.; Islam, M.; Webb, M.; Kizilbash, S.H. Antiangiogenic Exclusion Rules in Glioma Trials: Historical Perspectives and Guidance for Future Trial Design. Neuro-Oncol. Adv. 2024, 6, vdae039. [Google Scholar] [CrossRef]
  46. Qu, C.-Y.; Zheng, Y.; Zhou, M.; Zhang, Y.; Shen, F.; Cao, J.; Xu, L.-M. Value of Bevacizumab in Treatment of Colorectal Cancer: A Meta-Analysis. World J. Gastroenterol. 2015, 21, 5072–5080. [Google Scholar] [CrossRef]
  47. Cao, D.; Zheng, Y.; Xu, H.; Ge, W.; Xu, X. Bevacizumab Improves Survival in Metastatic Colorectal Cancer Patients with Primary Tumor Resection: A Meta-Analysis. Sci. Rep. 2019, 9, 20326. [Google Scholar] [CrossRef]
  48. Cutsem, E.V.; Rivera, F.; Berry, S.; Kretzschmar, A.; Michael, M.; DiBartolomeo, M.; Mazier, M.-A.; Canon, J.-L.; Georgoulias, V.; Peeters, M.; et al. Safety and Efficacy of First-Line Bevacizumab with FOLFOX, XELOX, FOLFIRI and Fluoropyrimidines in Metastatic Colorectal Cancer: The BEAT Study. Ann. Oncol. 2009, 20, 1842–1847. [Google Scholar] [CrossRef]
  49. Soria, J.-C.; Mauguen, A.; Reck, M.; Sandler, A.B.; Saijo, N.; Johnson, D.H.; Burcoveanu, D.; Fukuoka, M.; Besse, B.; Pignon, J.-P.; et al. Systematic Review and Meta-Analysis of Randomised, Phase II/III Trials Adding Bevacizumab to Platinum-Based Chemotherapy as First-Line Treatment in Patients with Advanced Non-Small-Cell Lung Cancer. Ann. Oncol. 2013, 24, 20–30. [Google Scholar] [CrossRef]
  50. Liu, Y.; Li, H.-M.; Wang, R. Effectiveness and Safety of Adding Bevacizumab to Platinum-Based Chemotherapy as First-Line Treatment for Advanced Non-Small-Cell Lung Cancer: A Meta-Analysis. Front. Med. 2021, 8, 616380. [Google Scholar] [CrossRef]
  51. Ascha, M.S.; Wang, J.F.; Kumthekar, P.; Sloan, A.E.; Kruchko, C.; Barnholtz-Sloan, J.S. Bevacizumab for the Treatment of Non-Small Cell Lung Cancer Patients with Synchronous Brain Metastases. Sci. Rep. 2019, 9, 17792. [Google Scholar] [CrossRef] [PubMed]
  52. Thompson Coon, J.S.; Liu, Z.; Hoyle, M.; Rogers, G.; Green, C.; Moxham, T.; Welch, K.; Stein, K. Sunitinib and Bevacizumab for First-Line Treatment of Metastatic Renal Cell Carcinoma: A Systematic Review and Indirect Comparison of Clinical Effectiveness. Br. J. Cancer 2009, 101, 238–243. [Google Scholar] [CrossRef] [PubMed]
  53. Feldman, D.R.; Ged, Y.; Lee, C.-H.; Knezevic, A.; Molina, A.M.; Chen, Y.-B.; Chaim, J.; Coskey, D.T.; Murray, S.; Tickoo, S.K.; et al. Everolimus plus Bevacizumab Is Effective First-Line Treatment for Patients with Advanced Papillary Variant Renal Cell Carcinoma: Final Results from a Phase II Trial. Cancer 2020, 126, 5247–5255. [Google Scholar] [CrossRef]
  54. Zhang, A.B.; Mozaffari, K.; Aguirre, B.; Li, V.; Kubba, R.; Desai, N.C.; Wei, D.; Yang, I.; Wadehra, M. Exploring the Past, Present, and Future of Anti-Angiogenic Therapy in Glioblastoma. Cancers 2023, 15, 830. [Google Scholar] [CrossRef]
  55. Funakoshi, Y.; Takigawa, K.; Hata, N.; Kuga, D.; Hatae, R.; Sangatsuda, Y.; Fujioka, Y.; Otsuji, R.; Sako, A.; Yoshitake, T.; et al. Changes in the Relapse Pattern and Prognosis of Glioblastoma After Approval of First-Line Bevacizumab: A Single-Center Retrospective Study. World Neurosurg. 2022, 159, e479–e487. [Google Scholar] [CrossRef]
  56. Khan, A.B.; Lee, S.; Harmanci, A.S.; Patel, R.; Latha, K.; Yang, Y.; Marisetty, A.; Lee, H.-K.; Heimberger, A.B.; Fuller, G.N.; et al. CXCR4 expression is associated with proneural-to-mesenchymal transition in glioblastoma. Int. J. Cancer 2023, 152, 713–724. [Google Scholar] [CrossRef]
  57. Azam, Z.; To, S.T.; Tannous, B.A. Mesenchymal Transformation: The Rosetta Stone of Glioblastoma Pathogenesis and Therapy Resistance. Adv. Sci. 2020, 7, 2002015. [Google Scholar] [CrossRef]
  58. Luo, J.; Wang, Z.; Zhang, X.; Yu, H.; Chen, H.; Song, K.; Zhang, Y.; Schwartz, L.M.; Chen, H.; Liu, Y.; et al. Vascular Immune Evasion of Mesenchymal Glioblastoma Is Mediated by Interaction and Regulation of VE-Cadherin on PD-L1. Cancers 2023, 15, 4257. [Google Scholar] [CrossRef]
  59. Xu, C.; Hou, P.; Li, X.; Xiao, M.; Zhang, Z.; Li, Z.; Xu, J.; Liu, G.; Tan, Y.; Fang, C. Comprehensive Understanding of Glioblastoma Molecular Phenotypes: Classification, Characteristics, and Transition. Cancer Biol. Med. 2024, 21, 363–381. [Google Scholar] [CrossRef]
  60. Phillips, H.S.; Kharbanda, S.; Chen, R.; Forrest, W.F.; Soriano, R.H.; Wu, T.D.; Misra, A.; Nigro, J.M.; Colman, H.; Soroceanu, L.; et al. Molecular Subclasses of High-Grade Glioma Predict Prognosis, Delineate a Pattern of Disease Progression, and Resemble Stages in Neurogenesis. Cancer Cell 2006, 9, 157–173. [Google Scholar] [CrossRef]
  61. Huang, Y.-J.; Nan, G.-X. Oxidative Stress-Induced Angiogenesis. J. Clin. Neurosci. 2019, 63, 13–16. [Google Scholar] [CrossRef]
  62. Jiang, X.; Wang, J.; Deng, X.; Xiong, F.; Zhang, S.; Gong, Z.; Li, X.; Cao, K.; Deng, H.; He, Y.; et al. The Role of Microenvironment in Tumor Angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 204. [Google Scholar] [CrossRef]
  63. Ahir, B.K.; Engelhard, H.H.; Lakka, S.S. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol. Neurobiol. 2020, 57, 2461–2478. [Google Scholar] [CrossRef] [PubMed]
  64. Guyon, J.; Chapouly, C.; Andrique, L.; Bikfalvi, A.; Daubon, T. The Normal and Brain Tumor Vasculature: Morphological and Functional Characteristics and Therapeutic Targeting. Front. Physiol. 2021, 12, 622615. [Google Scholar] [CrossRef] [PubMed]
  65. Gerstner, E.R.; Emblem, K.E.; Yen, Y.-F.; Dietrich, J.; Jordan, J.T.; Catana, C.; Wenchin, K.L.; Hooker, J.M.; Duda, D.G.; Rosen, B.R.; et al. Vascular Dysfunction Promotes Regional Hypoxia after Bevacizumab Therapy in Recurrent Glioblastoma Patients. Neurooncol. Adv. 2020, 2, vdaa157. [Google Scholar] [CrossRef]
  66. Brisson, L.; Henrique Geraldo, L.; Bikfalvi, A.; Mathivet, T. The Strange Microenvironment of Glioblastoma. Rev. Neurol. 2023, 179, 490–501. [Google Scholar] [CrossRef]
  67. Charalambous, C.; Chen, T.C.; Hofman, F.M. Characteristics of Tumor-Associated Endothelial Cells Derived from Glioblastoma Multiforme. Neurosurg. Focus. 2006, 20, E22. [Google Scholar] [CrossRef]
  68. Vartanian, A.; Singh, S.K.; Agnihotri, S.; Jalali, S.; Burrell, K.; Aldape, K.D.; Zadeh, G. GBM’s Multifaceted Landscape: Highlighting Regional and Microenvironmental Heterogeneity. Neuro Oncol. 2014, 16, 1167–1175. [Google Scholar] [CrossRef]
  69. Sounni, N.E.; Paye, A.; Host, L.; Noël, A. MT-MMPS as Regulators of Vessel Stability Associated with Angiogenesis. Front. Pharmacol. 2011, 1, 111. [Google Scholar] [CrossRef]
  70. Testa, E.; Palazzo, C.; Mastrantonio, R.; Viscomi, M.T. Dynamic Interactions between Tumor Cells and Brain Microvascular Endothelial Cells in Glioblastoma. Cancers 2022, 14, 3128. [Google Scholar] [CrossRef]
  71. Neubauer, K.; Zieger, B. Endothelial Cells and Coagulation. Cell Tissue Res. 2022, 387, 391–398. [Google Scholar] [CrossRef] [PubMed]
  72. Mathivet, T.; Bouleti, C.; Van Woensel, M.; Stanchi, F.; Verschuere, T.; Phng, L.; Dejaegher, J.; Balcer, M.; Matsumoto, K.; Georgieva, P.B.; et al. Dynamic Stroma Reorganization Drives Blood Vessel Dysmorphia during Glioma Growth. EMBO Mol. Med. 2017, 9, 1629–1645. [Google Scholar] [CrossRef] [PubMed]
  73. Barlow, K.D.; Sanders, A.M.; Soker, S.; Ergun, S.; Metheny-Barlow, L.J. Pericytes on the Tumor Vasculature: Jekyll or Hyde? Cancer Microenviron. 2013, 6, 1–17. [Google Scholar] [CrossRef] [PubMed]
  74. Tate, M.C.; Aghi, M.K. Biology of Angiogenesis and Invasion in Glioma. Neurotherapeutics 2009, 6, 447–457. [Google Scholar] [CrossRef]
  75. Sanati, M.; Afshari, A.R.; Amini, J.; Mollazadeh, H.; Jamialahmadi, T.; Sahebkar, A. Targeting Angiogenesis in Gliomas: Potential Role of Phytochemicals. J. Funct. Foods 2022, 96, 105192. [Google Scholar] [CrossRef]
  76. Dudley, A.C.; Griffioen, A.W. Pathological Angiogenesis: Mechanisms and Therapeutic Strategies. Angiogenesis 2023, 26, 313–347. [Google Scholar] [CrossRef]
  77. Plate, K.H.; Scholz, A.; Dumont, D.J. Tumor Angiogenesis and Anti-Angiogenic Therapy in Malignant Gliomas Revisited. Acta Neuropathol. 2012, 124, 763–775. [Google Scholar] [CrossRef]
  78. Pezzella, F.; Ribatti, D. Vascular Co-option and Vasculogenic Mimicry Mediate Resistance to Antiangiogenic Strategies. Cancer Rep. 2020, 5, e1318. [Google Scholar] [CrossRef]
  79. Belotti, D.; Pinessi, D.; Taraboletti, G. Alternative Vascularization Mechanisms in Tumor Resistance to Therapy. Cancers 2021, 13, 1912. [Google Scholar] [CrossRef]
  80. Holst, C.B.; Pedersen, H.; Obara, E.A.A.; Vitting-Seerup, K.; Jensen, K.E.; Skjøth-Rasmussen, J.; Lund, E.L.; Poulsen, H.S.; Johansen, J.S.; Hamerlik, P. Perspective: Targeting VEGF-A and YKL-40 in Glioblastoma—Matter Matters. Cell Cycle 2021, 20, 702–715. [Google Scholar] [CrossRef]
  81. Cuypers, A.; Truong, A.-C.K.; Becker, L.M.; Saavedra-García, P.; Carmeliet, P. Tumor Vessel Co-Option: The Past & the Future. Front. Oncol. 2022, 12, 965277. [Google Scholar]
  82. Ribatti, D.; Annese, T.; Tamma, R. Vascular Co-Option in Resistance to Anti-Angiogenic Therapy. Front. Oncol. 2023, 13, 1323350. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, R.; Yao, Y.; Gao, H.; Hu, X. Mechanisms of Angiogenesis in Tumour. Front. Oncol. 2024, 14, 1359069. [Google Scholar] [CrossRef]
  84. Brat, D.J.; Van Meir, E.G. Glomeruloid Microvascular Proliferation Orchestrated by VPF/VEGF. Am. J. Pathol. 2001, 158, 789–796. [Google Scholar] [CrossRef]
  85. Ionescu, C.; Oprea, B.; Ciobanu, G.; Georgescu, M.; Bică, R.; Mateescu, G.-O.; Huseynova, F.; Barragan-Montero, V. The Angiogenic Balance and Its Implications in Cancer and Cardiovascular Diseases: An Overview. Medicina 2022, 58, 903. [Google Scholar] [CrossRef]
  86. Wong, A.L.; Haroon, Z.A.; Werner, S.; Dewhirst, M.W.; Greenberg, C.S.; Peters, K.G. Tie2 Expression and Phosphorylation in Angiogenic and Quiescent Adult Tissues. Circ. Res. 1997, 81, 567–574. [Google Scholar] [CrossRef]
  87. Al Sabti, H. Therapeutic Angiogenesis in Cardiovascular Disease. J. Cardiothorac. Surg. 2007, 2, 49. [Google Scholar] [CrossRef]
  88. Sund, M.; Nyberg, P.; Eikesdal, H.P. Endogenous Matrix-Derived Inhibitors of Angiogenesis. Pharmaceuticals 2010, 3, 3021–3039. [Google Scholar] [CrossRef]
  89. Saman, H.; Raza, S.S.; Uddin, S.; Rasul, K. Inducing Angiogenesis, a Key Step in Cancer Vascularization, and Treatment Approaches. Cancers 2020, 12, 1172. [Google Scholar] [CrossRef]
  90. Logsdon, E.A.; Finley, S.D.; Popel, A.S.; Gabhann, F.M. A Systems Biology View of Blood Vessel Growth and Remodelling. J. Cell. Mol. Med. 2014, 18, 1491–1508. [Google Scholar] [CrossRef]
  91. Senger, D.R.; Davis, G.E. Angiogenesis. Cold Spring Harb. Perspect. Biol. 2011, 3, a005090. [Google Scholar] [CrossRef] [PubMed]
  92. Bulnes, S.; Bengoetxea, H.; Ortuzar, N.; Argandoña, E.G.; Garcia-Blanco, Á.; Rico-Barrio, I.; Lafuente, J.V. Angiogenic Signalling Pathways Altered in Gliomas: Selection Mechanisms for More Aggressive Neoplastic Subpopulations with Invasive Phenotype. J. Signal Transduct. 2012, 2012, 597915. [Google Scholar] [CrossRef] [PubMed]
  93. Gupta, M.K.; Qin, R.-Y. Mechanism and Its Regulation of Tumor-Induced Angiogenesis. World J. Gastroenterol. 2003, 9, 1144–1155. [Google Scholar] [CrossRef]
  94. Sebestyén, A.; Kopper, L.; Dankó, T.; Tímár, J. Hypoxia Signaling in Cancer: From Basics to Clinical Practice. Pathol. Oncol. Res. 2021, 27, 1609802. [Google Scholar] [CrossRef]
  95. Yang, Y.; Sun, M.; Wang, L.; Jiao, B. HIFs, Angiogenesis, and Cancer. J. Cell Biochem. 2013, 114, 967–974. [Google Scholar] [CrossRef]
  96. Ziello, J.E.; Jovin, I.S.; Huang, Y. Hypoxia-Inducible Factor (HIF)-1 Regulatory Pathway and Its Potential for Therapeutic Intervention in Malignancy and Ischemia. Yale J. Biol. Med. 2007, 80, 51–60. [Google Scholar]
  97. Kaur, B.; Khwaja, F.W.; Severson, E.A.; Matheny, S.L.; Brat, D.J.; Van Meir, E.G. Hypoxia and the Hypoxia-Inducible-Factor Pathway in Glioma Growth and Angiogenesis. Neuro-Oncology 2005, 7, 134–153. [Google Scholar] [CrossRef]
  98. Begagić, E.; Bečulić, H.; Džidić-Krivić, A.; Kadić Vukas, S.; Hadžić, S.; Mekić-Abazović, A.; Šegalo, S.; Papić, E.; Muchai Echengi, E.; Pugonja, R.; et al. Understanding the Significance of Hypoxia-Inducible Factors (HIFs) in Glioblastoma: A Systematic Review. Cancers 2024, 16, 2089. [Google Scholar] [CrossRef]
  99. Chédeville, A.L.; Madureira, P.A. The Role of Hypoxia in Glioblastoma Radiotherapy Resistance. Cancers 2021, 13, 542. [Google Scholar] [CrossRef]
  100. Calvo, P.M.; Hernández, R.G.; de la Cruz, R.R.; Pastor, A.M. Role of Vascular Endothelial Growth Factor as a Critical Neurotrophic Factor for the Survival and Physiology of Motoneurons. Neural Regen. Res. 2022, 18, 1691–1696. [Google Scholar] [CrossRef]
  101. Dakowicz, D.; Zajkowska, M.; Mroczko, B. Relationship between VEGF Family Members, Their Receptors and Cell Death in the Neoplastic Transformation of Colorectal Cancer. Int. J. Mol. Sci. 2022, 23, 3375. [Google Scholar] [CrossRef] [PubMed]
  102. Iyer, S.; Acharya, K.R. Tying the Knot: The Cystine Signature and Molecular-Recognition Processes of the Vascular Endothelial Growth Factor Family of Angiogenic Cytokines. FEBS J. 2011, 278, 4304–4322. [Google Scholar] [CrossRef]
  103. Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis. Genes Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef] [PubMed]
  104. Stuttfeld, E.; Ballmer-Hofer, K. Structure and Function of VEGF Receptors. IUBMB Life 2009, 61, 915–922. [Google Scholar] [CrossRef] [PubMed]
  105. Lisi, L.; Pia Ciotti, G.M.; Chiavari, M.; Ruffini, F.; Lacal, P.M.; Graziani, G.; Navarra, P. Vascular Endothelial Growth Factor Receptor 1 in Glioblastoma-associated Microglia/Macrophages. Oncol. Rep. 2020, 43, 2083–2092. [Google Scholar] [CrossRef]
  106. Azimi-Nezhad, M.; Stathopoulou, M.G.; Bonnefond, A.; Rancier, M.; Saleh, A.; Lamont, J.; Fitzgerald, P.; Ndiaye, N.C.; Visvikis-Siest, S. Associations of Vascular Endothelial Growth Factor (VEGF) with Adhesion and Inflammation Molecules in a Healthy Population. Cytokine 2013, 61, 602–607. [Google Scholar] [CrossRef]
  107. Goel, H.L.; Mercurio, A.M. VEGF Targets the Tumour Cell. Nat. Rev. Cancer 2013, 13, 871–882. [Google Scholar] [CrossRef]
  108. Apte, R.S.; Chen, D.S.; Ferrara, N. VEGF in Signaling and Disease: Beyond Discovery and Development. Cell 2019, 176, 1248–1264. [Google Scholar] [CrossRef]
  109. Maity, A.; Pore, N.; Lee, J.; Solomon, D.; O’Rourke, D.M. Epidermal Growth Factor Receptor Transcriptionally Up-Regulates Vascular Endothelial Growth Factor Expression in Human Glioblastoma Cells via a Pathway Involving Phosphatidylinositol 3′-Kinase and Distinct from That Induced by Hypoxia. Cancer Res. 2000, 60, 5879–5886. [Google Scholar]
  110. Plate, K.H.; Breier, G.; Weich, H.A.; Risau, W. Vascular Endothelial Growth Factor Is a Potential Tumour Angiogenesis Factor in Human Gliomas in Vivo. Nature 1992, 359, 845–848. [Google Scholar] [CrossRef]
  111. Sonoda, Y.; Kanamori, M.; Deen, D.F.; Cheng, S.-Y.; Berger, M.S.; Pieper, R.O. Overexpression of Vascular Endothelial Growth Factor Isoforms Drives Oxygenation and Growth but Not Progression to Glioblastoma Multiforme in a Human Model of Gliomagenesis. Cancer Res. 2003, 63, 1962–1968. [Google Scholar] [PubMed]
  112. Flynn, J.R.; Wang, L.; Gillespie, D.L.; Stoddard, G.J.; Reid, J.K.; Owens, J.; Ellsworth, G.B.; Salzman, K.L.; Kinney, A.Y.; Jensen, R.L. Hypoxia-Regulated Protein Expression, Patient Characteristics, and Preoperative Imaging as Predictors of Survival in Adults with Glioblastoma Multiforme. Cancer 2008, 113, 1032–1042. [Google Scholar] [CrossRef] [PubMed]
  113. Huang, H.; Held-Feindt, J.; Buhl, R.; Mehdorn, H.M.; Mentlein, R. Expression of VEGF and Its Receptors in Different Brain Tumors. Neurol. Res. 2005, 27, 371–377. [Google Scholar] [CrossRef] [PubMed]
  114. Ghalehbandi, S.; Yuzugulen, J.; Pranjol, M.Z.I.; Pourgholami, M.H. The Role of VEGF in Cancer-Induced Angiogenesis and Research Progress of Drugs Targeting VEGF. Eur. J. Pharmacol. 2023, 949, 175586. [Google Scholar] [CrossRef]
  115. Xu, C.; Wu, X.; Zhu, J. VEGF Promotes Proliferation of Human Glioblastoma Multiforme Stem-like Cells through VEGF Receptor 2. Sci. World J. 2013, 2013, 417413. [Google Scholar] [CrossRef]
  116. Martini, M.; de Pascalis, I.; D’Alessandris, Q.G.; Fiorentino, V.; Pierconti, F.; Marei, H.E.-S.; Ricci-Vitiani, L.; Pallini, R.; Larocca, L.M. VEGF-121 Plasma Level as Biomarker for Response to Anti-Angiogenetic Therapy in Recurrent Glioblastoma. BMC Cancer 2018, 18, 553. [Google Scholar] [CrossRef]
  117. Wallensten, J.; Mobarrez, F.; Åsberg, M.; Borg, K.; Beser, A.; Wilczek, A.; Nager, A. Isoforms of Soluble Vascular Endothelial Growth Factor in Stress-Related Mental Disorders: A Cross-Sectional Study. Sci. Rep. 2021, 11, 16693. [Google Scholar] [CrossRef]
  118. Yamakuchi, M.; Okawa, M.; Takenouchi, K.; Bibek, A.; Yamada, S.; Inoue, K.; Higurashi, K.; Tabaru, A.; Tanoue, K.; Oyama, Y.; et al. VEGF-A165 Is the Predominant VEGF-A Isoform in Platelets, While VEGF-A121 Is Abundant in Serum and Plasma from Healthy Individuals. PLoS ONE 2023, 18, e0284131. [Google Scholar] [CrossRef]
  119. Vempati, P.; Popel, A.S.; Mac Gabhann, F. Extracellular Regulation of VEGF: Isoforms, Proteolysis, and Vascular Patterning. Cytokine Growth Factor Rev. 2014, 25, 1–19. [Google Scholar] [CrossRef]
  120. D’Alessandris, Q.G.; Martini, M.; Cenci, T.; Capo, G.; Ricci-Vitiani, L.; Larocca, L.M.; Pallini, R. VEGF Isoforms as Outcome Biomarker for Anti-Angiogenic Therapy in Recurrent Glioblastoma. Neurology 2015, 84, 1906–1908. [Google Scholar] [CrossRef]
  121. Lohela, M.; Bry, M.; Tammela, T.; Alitalo, K. VEGFs and Receptors Involved in Angiogenesis versus Lymphangiogenesis. Curr. Opin. Cell Biol. 2009, 21, 154–165. [Google Scholar] [CrossRef] [PubMed]
  122. Ornitz, D.M.; Itoh, N. Fibroblast Growth Factors. Genome Biol. 2001, 2, reviews3005.1–reviews3005.12. [Google Scholar] [CrossRef] [PubMed]
  123. Beenken, A.; Mohammadi, M. The FGF Family: Biology, Pathophysiology and Therapy. Nat. Rev. Drug Discov. 2009, 8, 235–253. [Google Scholar] [CrossRef] [PubMed]
  124. Farooq, M.; Khan, A.W.; Kim, M.S.; Choi, S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021, 10, 3242. [Google Scholar] [CrossRef]
  125. Zhuang, L.; Falquet, L.; Trueb, B. Genome-Wide Comparison of FGFRL1 with Structurally Related Surface Receptors. Exp. Ther. Med. 2010, 1, 161–168. [Google Scholar] [CrossRef]
  126. Kostas, M.; Lampart, A.; Bober, J.; Wiedlocha, A.; Tomala, J.; Krowarsch, D.; Otlewski, J.; Zakrzewska, M. Translocation of Exogenous FGF1 and FGF2 Protects the Cell against Apoptosis Independently of Receptor Activation. J. Mol. Biol. 2018, 430, 4087–4101. [Google Scholar] [CrossRef]
  127. Giacomini, A.; Grillo, E.; Rezzola, S.; Ribatti, D.; Rusnati, M.; Ronca, R.; Presta, M. The FGF/FGFR System in the Physiopathology of the Prostate Gland. Physiol. Rev. 2021, 101, 569–610. [Google Scholar] [CrossRef]
  128. Liu, G.; Chen, T.; Ding, Z.; Wang, Y.; Wei, Y.; Wei, X. Inhibition of FGF-FGFR and VEGF-VEGFR Signalling in Cancer Treatment. Cell Prolif. 2021, 54, e13009. [Google Scholar] [CrossRef]
  129. Sahores, A.; Figueroa, V.; May, M.; Liguori, M.; Rubstein, A.; Fuentes, C.; Jacobsen, B.M.; Elía, A.; Rojas, P.; Sequeira, G.R.; et al. Increased High Molecular Weight FGF2 in Endocrine-Resistant Breast Cancer. Horm. Cancer 2018, 9, 338–348. [Google Scholar] [CrossRef]
  130. Donnem, T.; Al-Shibli, K.; Al-Saad, S.; Busund, L.-T.; Bremnes, R.M. Prognostic Impact of Fibroblast Growth Factor 2 in Non-Small Cell Lung Cancer: Coexpression with VEGFR-3 and PDGF-B Predicts Poor Survival. J. Thorac. Oncol. 2009, 4, 578–585. [Google Scholar] [CrossRef]
  131. Gnanapragasam, V.J.; Robinson, M.C.; Marsh, C.; Robson, C.N.; Hamdy, F.C.; Leung, H.Y. FGF8 Isoform b Expression in Human Prostate Cancer. Br. J. Cancer 2003, 88, 1432–1438. [Google Scholar] [CrossRef] [PubMed]
  132. Gazzaniga, P.; Gandini, O.; Gradilone, A.; Silvestri, I.; Giuliani, L.; Magnanti, M.; Gallucci, M.; Saccani, G.; Frati, L.; Agliano, A.M. Detection of Basic Fibroblast Growth Factor mRNA in Urinary Bladder Cancer: Correlation with Local Relapses. Int. J. Oncol. 1999, 14, 1123–1127. [Google Scholar] [CrossRef] [PubMed]
  133. Tai, A.L.S.; Sham, J.S.T.; Xie, D.; Fang, Y.; Wu, Y.-L.; Hu, L.; Deng, W.; Tsao, G.S.W.; Qiao, G.-B.; Cheung, A.L.M.; et al. Co-Overexpression of Fibroblast Growth Factor 3 and Epidermal Growth Factor Receptor Is Correlated with the Development of Nonsmall Cell Lung Carcinoma. Cancer 2006, 106, 146–155. [Google Scholar] [CrossRef]
  134. Suzuki, K.; Tokue, A.; Kamiakito, T.; Kuriki, K.; Saito, K.; Tanaka, A. Predominant Expression of Fibroblast Growth Factor (FGF) 8, FGF4, and FGF Receptor 1 in Nonseminomatous and Highly Proliferative Components of Testicular Germ Cell Tumors. Virchows Arch. 2001, 439, 616–621. [Google Scholar] [CrossRef]
  135. Sonvilla, G.; Allerstorfer, S.; Stättner, S.; Karner, J.; Klimpfinger, M.; Fischer, H.; Grasl-Kraupp, B.; Holzmann, K.; Berger, W.; Wrba, F.; et al. FGF18 in Colorectal Tumour Cells: Autocrine and Paracrine Effects. Carcinogenesis 2008, 29, 15–24. [Google Scholar] [CrossRef]
  136. Takahashi, J.A.; Fukumoto, M.; Igarashi, K.; Oda, Y.; Kikuchi, H.; Hatanaka, M. Correlation of Basic Fibroblast Growth Factor Expression Levels with the Degree of Malignancy and Vascularity in Human Gliomas. J. Neurosurg. 1992, 76, 792–798. [Google Scholar] [CrossRef]
  137. Takahashi, J.A.; Mori, H.; Fukumoto, M.; Igarashi, K.; Jaye, M.; Oda, Y.; Kikuchi, H.; Hatanaka, M. Gene Expression of Fibroblast Growth Factors in Human Gliomas and Meningiomas: Demonstration of Cellular Source of Basic Fibroblast Growth Factor mRNA and Peptide in Tumor Tissues. Proc. Natl. Acad. Sci. USA 1990, 87, 5710–5714. [Google Scholar] [CrossRef]
  138. Kaya, B.; Çiçek, O.; Erdi, F.; Findik, S.; Karatas, Y.; Esen, H.; Keskin, F.; Kalkan, E. Intratumoral Hemorrhage-Related Differences in the Expression of Vascular Endothelial Growth Factor, Basic Fibroblast Growth Factor and Thioredoxin Reductase 1 in Human Glioblastoma. Mol. Clin. Oncol. 2016, 5, 343–346. [Google Scholar] [CrossRef]
  139. Numasaki, M.; Ito, K. VEGF-A, HGF and bFGF Are Involved in IL-17A-Mediated Migration and Capillary-like Vessel Formation of Vascular Endothelial Cells. Iran. J. Immunol. 2021, 18, 103–110. [Google Scholar]
  140. Yoshida, A.; Anand-Apte, B.; Zetter, B.R. Differential Endothelial Migration and Proliferation to Basic Fibroblast Growth Factor and Vascular Endothelial Growth Factor. Growth Factors 1996, 13, 57–64. [Google Scholar] [CrossRef]
  141. Pepper, M.S.; Ferrara, N.; Orci, L.; Montesano, R. Potent Synergism between Vascular Endothelial Growth Factor and Basic Fibroblast Growth Factor in the Induction of Angiogenesis in Vitro. Biochem. Biophys. Res. Commun. 1992, 189, 824–831. [Google Scholar] [CrossRef] [PubMed]
  142. Qiao, D.; Meyer, K.; Mundhenke, C.; Drew, S.A.; Friedl, A. Heparan Sulfate Proteoglycans as Regulators of Fibroblast Growth Factor-2 Signaling in Brain Endothelial Cells. Specific Role for Glypican-1 in Glioma Angiogenesis. J. Biol. Chem. 2003, 278, 16045–16053. [Google Scholar] [CrossRef] [PubMed]
  143. Cuevas, P.; Carceller, F.; Angulo, J.; González-Corrochano, R.; Cuevas-Bourdier, A.; Giménez-Gallego, G. Antiglioma Effects of a New, Low Molecular Mass, Inhibitor of Fibroblast Growth Factor. Neurosci. Lett. 2011, 491, 1–7. [Google Scholar] [CrossRef] [PubMed]
  144. Powers, C.J.; McLeskey, S.W.; Wellstein, A. Fibroblast Growth Factors, Their Receptors and Signaling. Endocr. Relat. Cancer 2000, 7, 165–197. [Google Scholar] [CrossRef]
  145. Li, G.; Chen, Z.; Hu, Y.-D.; Wei, H.; Li, D.; Ji, H.; Wang, D.-L. Autocrine Factors Sustain Glioblastoma Stem Cell Self-Renewal. Oncol. Rep. 2009, 21, 419–424. [Google Scholar]
  146. Toyoda, K.; Tanaka, K.; Nakagawa, S.; Thuy, D.H.D.; Ujifuku, K.; Kamada, K.; Hayashi, K.; Matsuo, T.; Nagata, I.; Niwa, M. Initial Contact of Glioblastoma Cells with Existing Normal Brain Endothelial Cells Strengthen the Barrier Function via Fibroblast Growth Factor 2 Secretion: A New in Vitro Blood-Brain Barrier Model. Cell Mol. Neurobiol. 2013, 33, 489–501. [Google Scholar] [CrossRef]
  147. Zahra, F.T.; Sajib, M.S.; Mikelis, C.M. Role of bFGF in Acquired Resistance upon Anti-VEGF Therapy in Cancer. Cancers 2021, 13, 1422. [Google Scholar] [CrossRef]
  148. Sleeman, M.; Fraser, J.; McDonald, M.; Yuan, S.; White, D.; Grandison, P.; Kumble, K.; Watson, J.D.; Murison, J.G. Identification of a New Fibroblast Growth Factor Receptor, FGFR5. Gene 2001, 271, 171–182. [Google Scholar] [CrossRef]
  149. Harding, M.J.; Nechiporuk, A.V. Fgfr-Ras-MAPK Signaling Is Required for Apical Constriction via Apical Positioning of Rho-Associated Kinase during Mechanosensory Organ Formation. Development 2012, 139, 3130–3135. [Google Scholar] [CrossRef]
  150. Mohammadi, M.; Honegger, A.M.; Rotin, D.; Fischer, R.; Bellot, F.; Li, W.; Dionne, C.A.; Jaye, M.; Rubinstein, M.; Schlessinger, J. A Tyrosine-Phosphorylated Carboxy-Terminal Peptide of the Fibroblast Growth Factor Receptor (Flg) Is a Binding Site for the SH2 Domain of Phospholipase C-Gamma 1. Mol. Cell Biol. 1991, 11, 5068–5078. [Google Scholar]
  151. Reis-Filho, J.S.; Simpson, P.T.; Turner, N.C.; Lambros, M.B.; Jones, C.; Mackay, A.; Grigoriadis, A.; Sarrio, D.; Savage, K.; Dexter, T.; et al. FGFR1 Emerges as a Potential Therapeutic Target for Lobular Breast Carcinomas. Clin. Cancer Res. 2006, 12, 6652–6662. [Google Scholar] [CrossRef] [PubMed]
  152. Hart, K.C.; Robertson, S.C.; Kanemitsu, M.Y.; Meyer, A.N.; Tynan, J.A.; Donoghue, D.J. Transformation and Stat Activation by Derivatives of FGFR1, FGFR3, and FGFR4. Oncogene 2000, 19, 3309–3320. [Google Scholar] [CrossRef] [PubMed]
  153. Helsten, T.; Elkin, S.; Arthur, E.; Tomson, B.N.; Carter, J.; Kurzrock, R. The FGFR Landscape in Cancer: Analysis of 4,853 Tumors by Next-Generation Sequencing. Clin. Cancer Res. 2016, 22, 259–267. [Google Scholar] [CrossRef] [PubMed]
  154. Dono, A.; El Achi, H.; Bundrant, B.E.; Goli, P.S.; Zhu, P.; Ozkizilkaya, H.I.; Esquenazi, Y.; Ballester, L.Y. Infiltrating Gliomas with FGFR Alterations: Histologic Features, Genetic Alterations, and Potential Clinical Implications. Cancer Biomark. 2023, 36, 117–131. [Google Scholar] [CrossRef]
  155. Jimenez-Pascual, A.; Siebzehnrubl, F.A. Fibroblast Growth Factor Receptor Functions in Glioblastoma. Cells 2019, 8, 715. [Google Scholar] [CrossRef]
  156. Danielpour, D. Advances and Challenges in Targeting TGF-β Isoforms for Therapeutic Intervention of Cancer: A Mechanism-Based Perspective. Pharmaceuticals 2024, 17, 533. [Google Scholar] [CrossRef]
  157. Vander Ark, A.; Cao, J.; Li, X. TGF-β Receptors: In and beyond TGF-β Signaling. Cell. Signal. 2018, 52, 112–120. [Google Scholar] [CrossRef]
  158. Tie, Y.; Tang, F.; Peng, D.; Zhang, Y.; Shi, H. TGF-Beta Signal Transduction: Biology, Function and Therapy for Diseases. Mol. Biomed. 2022, 3, 45. [Google Scholar] [CrossRef]
  159. Oft, M.; Heider, K.-H.; Beug, H. TGFβ Signaling Is Necessary for Carcinoma Cell Invasiveness and Metastasis. Curr. Biol. 1998, 8, 1243–1252. [Google Scholar] [CrossRef]
  160. Davies, M.; Prime, S.S.; Eveson, J.W.; Price, N.; Ganapathy, A.; D’Mello, A.; Paterson, I.C. Transforming Growth Factor-β Enhances Invasion and Metastasis in Ras-Transfected Human Malignant Epidermal Keratinocytes. Int. J. Exp. Pathol. 2012, 93, 148–156. [Google Scholar] [CrossRef]
  161. Zhang, J.; Deng, Y.-T.; Liu, J.; Gan, L.; Jiang, Y. Role of Transforming Growth Factor-Β1 Pathway in Angiogenesis Induced by Chronic Stress in Colorectal Cancer. Cancer Biol. Ther. 2024, 25, 2366451. [Google Scholar] [CrossRef] [PubMed]
  162. Yang, L.; Liu, Z.; Wen, T. Multiplex Fluorescent Immunohistochemistry Quantitatively Analyses Microvascular Density (MVD) and the Roles of TGF-β Signalling in Orchestrating Angiogenesis in Colorectal Cancer. Transl. Cancer Res. 2019, 8, 429. [Google Scholar] [CrossRef] [PubMed]
  163. Hou, A.J.; Shih, R.M.; Uy, B.R.; Shafer, A.; Chang, Z.L.; Comin-Anduix, B.; Guemes, M.; Galic, Z.; Phyu, S.; Okada, H.; et al. IL-13Rα2/TGF-β Bispecific CAR-T Cells Counter TGF-β-Mediated Immune Suppression and Potentiate Anti-Tumor Responses in Glioblastoma. Neuro-Oncology 2024, 26, 1850–1866. [Google Scholar] [CrossRef] [PubMed]
  164. Ghahremanifard, P.; Chanda, A.; Bonni, S.; Bose, P. TGF-β Mediated Immune Evasion in Cancer—Spotlight on Cancer-Associated Fibroblasts. Cancers 2020, 12, 3650. [Google Scholar] [CrossRef]
  165. Zhang, J.; Thorikay, M.; van der Zon, G.; van Dinther, M.; Ten Dijke, P. Studying TGF-β Signaling and TGF-β-Induced Epithelial-to-Mesenchymal Transition in Breast Cancer and Normal Cells. J. Vis. Exp. 2020, 27, e61830. [Google Scholar]
  166. Zhang, Q.; Yu, N.; Lee, C. Mysteries of TGF-β Paradox in Benign and Malignant Cells. Front. Oncol. 2014, 4, 94. [Google Scholar] [CrossRef]
  167. Bruna, A.; Darken, R.S.; Rojo, F.; Ocaña, A.; Peñuelas, S.; Arias, A.; Paris, R.; Tortosa, A.; Mora, J.; Baselga, J.; et al. High TGFbeta-Smad Activity Confers Poor Prognosis in Glioma Patients and Promotes Cell Proliferation Depending on the Methylation of the PDGF-B Gene. Cancer Cell 2007, 11, 147–160. [Google Scholar] [CrossRef]
  168. Pen, A.; Moreno, M.J.; Durocher, Y.; Deb-Rinker, P.; Stanimirovic, D.B. Glioblastoma-Secreted Factors Induce IGFBP7 and Angiogenesis by Modulating Smad-2-Dependent TGF-β Signaling. Oncogene 2008, 27, 6834–6844. [Google Scholar] [CrossRef]
  169. Hovis, G.; Chandra, N.; Kejriwal, N.; Hsieh, K.J.-Y.; Chu, A.; Yang, I.; Wadehra, M. Understanding the Role of Endothelial Cells in Glioblastoma: Mechanisms and Novel Treatments. Int. J. Mol. Sci. 2024, 25, 6118. [Google Scholar] [CrossRef]
  170. Krishnan, S.; Szabo, E.; Burghardt, I.; Frei, K.; Tabatabai, G.; Weller, M. Modulation of Cerebral Endothelial Cell Function by TGF-β in Glioblastoma: VEGF-Dependent Angiogenesis versus Endothelial Mesenchymal Transition. Oncotarget 2015, 6, 22480–22495. [Google Scholar] [CrossRef]
  171. Piek, E.; Westermark, U.; Kastemar, M.; Heldin, C.H.; van Zoelen, E.J.; Nistér, M.; Ten Dijke, P. Expression of Transforming-Growth-Factor (TGF)-Beta Receptors and Smad Proteins in Glioblastoma Cell Lines with Distinct Responses to TGF-Beta1. Int. J. Cancer 1999, 80, 756–763. [Google Scholar] [CrossRef]
  172. Heldin, C.-H.; Westermark, B. Mechanism of Action and In Vivo Role of Platelet-Derived Growth Factor. Physiol. Rev. 1999, 79, 1283–1316. [Google Scholar] [CrossRef] [PubMed]
  173. Michalevicz, R.; Katz, F.; Stroobant, P.; Janossy, G.; Tindle, R.W.; Hoffbrand, A.V. Platelet-Derived Growth Factor Stimulates Growth of Highly Enriched Multipotent Haemopoietic Progenitors. Br. J. Haematol. 1986, 63, 591–598. [Google Scholar] [CrossRef] [PubMed]
  174. Deuel, T.F.; Senior, R.M.; Huang, J.S.; Griffin, G.L. Chemotaxis of Monocytes and Neutrophils to Platelet-Derived Growth Factor. J. Clin. Investig. 1982, 69, 1046–1049. [Google Scholar] [CrossRef]
  175. Grotendorst, G.R.; Chang, T.; Seppä, H.E.; Kleinman, H.K.; Martin, G.R. Platelet-Derived Growth Factor Is a Chemoattractant for Vascular Smooth Muscle Cells. J. Cell Physiol. 1982, 113, 261–266. [Google Scholar] [CrossRef]
  176. De Donatis, A.; Comito, G.; Buricchi, F.; Vinci, M.C.; Parenti, A.; Caselli, A.; Camici, G.; Manao, G.; Ramponi, G.; Cirri, P. Proliferation versus Migration in Platelet-Derived Growth Factor Signaling: The Key Role of Endocytosis. J. Biol. Chem. 2008, 283, 19948–19956. [Google Scholar] [CrossRef]
  177. Bartoschek, M.; Pietras, K. PDGF Family Function and Prognostic Value in Tumor Biology. Biochem. Biophys. Res. Commun. 2018, 503, 984–990. [Google Scholar] [CrossRef]
  178. Westermark, B.; Heldin, C.H.; Nistér, M. Platelet-Derived Growth Factor in Human Glioma. Glia 1995, 15, 257–263. [Google Scholar] [CrossRef]
  179. Mahadevan, D.; Yu, J.-C.; Saldanha, J.W.; Thanki, N.; McPhie, P.; Uren, A.; LaRochelle, W.J.; Heidaran, M.A. Structural Role of Extracellular Domain 1 of α-Platelet-Derived Growth Factor (PDGF) Receptor for PDGF-AA and PDGF-BB Binding (*). J. Biol. Chem. 1995, 270, 27595–27600. [Google Scholar] [CrossRef]
  180. Zhang, H.; Bajraszewski, N.; Wu, E.; Wang, H.; Moseman, A.P.; Dabora, S.L.; Griffin, J.D.; Kwiatkowski, D.J. PDGFRs Are Critical for PI3K/Akt Activation and Negatively Regulated by mTOR. J. Clin. Investig. 2007, 117, 730–738. [Google Scholar] [CrossRef]
  181. Zou, X.; Tang, X.-Y.; Qu, Z.-Y.; Sun, Z.-W.; Ji, C.-F.; Li, Y.-J.; Guo, S.-D. Targeting the PDGF/PDGFR Signaling Pathway for Cancer Therapy: A Review. Int. J. Biol. Macromol. 2022, 202, 539–557. [Google Scholar] [CrossRef] [PubMed]
  182. Pandey, P.; Khan, F.; Upadhyay, T.K.; Seungjoon, M.; Park, M.N.; Kim, B. New Insights about the PDGF/PDGFR Signaling Pathway as a Promising Target to Develop Cancer Therapeutic Strategies. Biomed. Pharmacother. 2023, 161, 114491. [Google Scholar] [CrossRef] [PubMed]
  183. Chen, J.; Yuan, W.; Wu, L.; Tang, Q.; Xia, Q.; Ji, J.; Liu, Z.; Ma, Z.; Zhou, Z.; Cheng, Y.; et al. PDGF-D Promotes Cell Growth, Aggressiveness, Angiogenesis and EMT Transformation of Colorectal Cancer by Activation of Notch1/Twist1 Pathway. Oncotarget 2016, 8, 9961–9973. [Google Scholar] [CrossRef]
  184. Heldin, C.-H.; Lennartsson, J. Structural and Functional Properties of Platelet-Derived Growth Factor and Stem Cell Factor Receptors. Cold Spring Harb. Perspect. Biol. 2013, 5, a009100. [Google Scholar] [CrossRef]
  185. Gacche, R.N.; Assaraf, Y.G. Redundant Angiogenic Signaling and Tumor Drug Resistance. Drug Resist. Updates 2018, 36, 47–76. [Google Scholar] [CrossRef]
  186. Kim, Y.; Kim, E.; Wu, Q.; Guryanova, O.; Hitomi, M.; Lathia, J.D.; Serwanski, D.; Sloan, A.E.; Weil, R.J.; Lee, J.; et al. Platelet-Derived Growth Factor Receptors Differentially Inform Intertumoral and Intratumoral Heterogeneity. Genes Dev. 2012, 26, 1247–1262. [Google Scholar] [CrossRef]
  187. Heldin, C.-H.; Lennartsson, J.; Westermark, B. Involvement of Platelet-Derived Growth Factor Ligands and Receptors in Tumorigenesis. J. Intern. Med. 2018, 283, 16–44. [Google Scholar] [CrossRef]
  188. Guo, P.; Hu, B.; Gu, W.; Xu, L.; Wang, D.; Huang, H.-J.S.; Cavenee, W.K.; Cheng, S.-Y. Platelet-Derived Growth Factor-B Enhances Glioma Angiogenesis by Stimulating Vascular Endothelial Growth Factor Expression in Tumor Endothelia and by Promoting Pericyte Recruitment. Am. J. Pathol. 2003, 162, 1083–1093. [Google Scholar] [CrossRef]
  189. Hermansson, M.; Nistér, M.; Betsholtz, C.; Heldin, C.H.; Westermark, B.; Funa, K. Endothelial Cell Hyperplasia in Human Glioblastoma: Coexpression of mRNA for Platelet-Derived Growth Factor (PDGF) B Chain and PDGF Receptor Suggests Autocrine Growth Stimulation. Proc. Natl. Acad. Sci. USA 1988, 85, 7748–7752. [Google Scholar] [CrossRef]
  190. Cenciarelli, C.; Marei, H.E.; Zonfrillo, M.; Pierimarchi, P.; Paldino, E.; Casalbore, P.; Felsani, A.; Vescovi, A.L.; Maira, G.; Mangiola, A. PDGF Receptor Alpha Inhibition Induces Apoptosis in Glioblastoma Cancer Stem Cells Refractory to Anti-Notch and Anti-EGFR Treatment. Mol. Cancer 2014, 13, 247. [Google Scholar] [CrossRef]
  191. Lane, R.; Cilibrasi, C.; Chen, J.; Shah, K.; Messuti, E.; Mazarakis, N.K.; Stebbing, J.; Critchley, G.; Song, E.; Simon, T.; et al. PDGF-R Inhibition Induces Glioblastoma Cell Differentiation via DUSP1/p38MAPK Signalling. Oncogene 2022, 41, 2749–2763. [Google Scholar] [CrossRef] [PubMed]
  192. Davis, S.; Aldrich, T.H.; Jones, P.F.; Acheson, A.; Compton, D.L.; Jain, V.; Ryan, T.E.; Bruno, J.; Radziejewski, C.; Maisonpierre, P.C.; et al. Isolation of Angiopoietin-1, a Ligand for the TIE2 Receptor, by Secretion-Trap Expression Cloning. Cell 1996, 87, 1161–1169. [Google Scholar] [CrossRef] [PubMed]
  193. Valenzuela, D.M.; Griffiths, J.A.; Rojas, J.; Aldrich, T.H.; Jones, P.F.; Zhou, H.; McClain, J.; Copeland, N.G.; Gilbert, D.J.; Jenkins, N.A.; et al. Angiopoietins 3 and 4: Diverging Gene Counterparts in Mice and Humans. Proc. Natl. Acad. Sci. USA 1999, 96, 1904–1909. [Google Scholar] [CrossRef] [PubMed]
  194. Barton, W.A.; Tzvetkova, D.; Nikolov, D.B. Structure of the Angiopoietin-2 Receptor Binding Domain and Identification of Surfaces Involved in Tie2 Recognition. Structure 2005, 13, 825–832. [Google Scholar] [CrossRef]
  195. Kontos, C.D.; Stauffer, T.P.; Yang, W.-P.; York, J.D.; Huang, L.; Blanar, M.A.; Meyer, T.; Peters, K.G. Tyrosine 1101 of Tie2 Is the Major Site of Association of P85 and Is Required for Activation of Phosphatidylinositol 3-Kinase and Akt. Mol. Cell. Biol. 1998, 18, 4131–4140. [Google Scholar] [CrossRef]
  196. Reusch, P.; Barleon, B.; Weindel, K.; Martiny-Baron, G.; Gödde, A.; Siemeister, G.; Marmé, D. Identification of a Soluble Form of the Angiopoietin Receptor TIE-2 Released from Endothelial Cells and Present in Human Blood. Angiogenesis 2001, 4, 123–131. [Google Scholar] [CrossRef]
  197. Schnürch, H.; Risau, W. Expression of Tie-2, a Member of a Novel Family of Receptor Tyrosine Kinases, in the Endothelial Cell Lineage. Development 1993, 119, 957–968. [Google Scholar] [CrossRef]
  198. La Porta, S.; Roth, L.; Singhal, M.; Mogler, C.; Spegg, C.; Schieb, B.; Qu, X.; Adams, R.H.; Baldwin, H.S.; Savant, S.; et al. Endothelial Tie1–Mediated Angiogenesis and Vascular Abnormalization Promote Tumor Progression and Metastasis. J. Clin. Investig. 2018, 128, 834–845. [Google Scholar] [CrossRef]
  199. Savant, S.; Porta, S.L.; Budnik, A.; Busch, K.; Hu, J.; Tisch, N.; Korn, C.; Valls, A.F.; Benest, A.V.; Terhardt, D.; et al. The Orphan Receptor Tie1 Controls Angiogenesis and Vascular Remodeling by Differentially Regulating Tie2 in Tip and Stalk Cells. Cell Rep. 2015, 12, 1761–1773. [Google Scholar] [CrossRef]
  200. Jeansson, M.; Gawlik, A.; Anderson, G.; Li, C.; Kerjaschki, D.; Henkelman, M.; Quaggin, S.E. Angiopoietin-1 Is Essential in Mouse Vasculature during Development and in Response to Injury. J. Clin. Investig. 2011, 121, 2278–2289. [Google Scholar] [CrossRef]
  201. Mueller, S.B.; Kontos, C.D. Tie1: An Orphan Receptor Provides Context for Angiopoietin-2/Tie2 Signaling. J. Clin. Investig. 2016, 126, 3188–3191. [Google Scholar] [CrossRef] [PubMed]
  202. Brunckhorst, M.K.; Xu, Y.; Lu, R.; Yu, Q. Angiopoietins Promote Ovarian Cancer Progression by Establishing a Procancer Microenvironment. Am. J. Pathol. 2014, 184, 2285–2296. [Google Scholar] [CrossRef] [PubMed]
  203. Martoglio, A.-M.; Tom, B.D.M.; Starkey, M.; Corps, A.N.; Charnock-Jones, D.S.; Smith, S.K. Changes in Tumorigenesis- and Angiogenesis-Related Gene Transcript Abundance Profiles in Ovarian Cancer Detected by Tailored High Density cDNA Arrays. Mol. Med. 2000, 6, 750–765. [Google Scholar] [CrossRef] [PubMed]
  204. Hu, B.; Jarzynka, M.J.; Guo, P.; Imanishi, Y.; Schlaepfer, D.D.; Cheng, S.-Y. Angiopoietin 2 Induces Glioma Cell Invasion by Stimulating Matrix Metalloprotease 2 Expression through the Avβ1 Integrin and Focal Adhesion Kinase Signaling Pathway. Cancer Res. 2006, 66, 775–783. [Google Scholar] [CrossRef]
  205. Dong, Z.; Chen, J.; Yang, X.; Zheng, W.; Wang, L.; Fang, M.; Wu, M.; Yao, M.; Yao, D. Ang-2 Promotes Lung Cancer Metastasis by Increasing Epithelial-Mesenchymal Transition. Oncotarget 2018, 9, 12705–12717. [Google Scholar] [CrossRef]
  206. Machein, M.R.; Knedla, A.; Knoth, R.; Wagner, S.; Neuschl, E.; Plate, K.H. Angiopoietin-1 Promotes Tumor Angiogenesis in a Rat Glioma Model. Am. J. Pathol. 2004, 165, 1557–1570. [Google Scholar] [CrossRef]
  207. Stratmann, A.; Risau, W.; Plate, K.H. Cell Type-Specific Expression of Angiopoietin-1 and Angiopoietin-2 Suggests a Role in Glioblastoma Angiogenesis. Am. J. Pathol. 1998, 153, 1459–1466. [Google Scholar] [CrossRef]
  208. Brunckhorst, M.K.; Wang, H.; Lu, R.; Yu, Q. Angiopoietin-4 Promotes Glioblastoma Progression by Enhancing Tumor Cell Viability and Angiogenesis. Cancer Res. 2010, 70, 7283–7293. [Google Scholar] [CrossRef]
  209. Peterson, T.E.; Kirkpatrick, N.D.; Huang, Y.; Farrar, C.T.; Marijt, K.A.; Kloepper, J.; Datta, M.; Amoozgar, Z.; Seano, G.; Jung, K.; et al. Dual Inhibition of Ang-2 and VEGF Receptors Normalizes Tumor Vasculature and Prolongs Survival in Glioblastoma by Altering Macrophages. Proc. Natl. Acad. Sci. USA 2016, 113, 4470–4475. [Google Scholar] [CrossRef]
  210. Choi, Y.C.; Dalakas, M.C. Expression of Matrix Metalloproteinases in the Muscle of Patients with Inflammatory Myopathies. Neurology 2000, 54, 65–71. [Google Scholar] [CrossRef]
  211. Jaoude, J.; Koh, Y. Matrix Metalloproteinases in Exercise and Obesity. Vasc. Health Risk Manag. 2016, 12, 287–295. [Google Scholar] [PubMed]
  212. Cui, N.; Hu, M.; Khalil, R.A. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog. Mol. Biol. Transl. Sci. 2017, 147, 1–73. [Google Scholar] [PubMed]
  213. Gobin, E.; Bagwell, K.; Wagner, J.; Mysona, D.; Sandirasegarane, S.; Smith, N.; Bai, S.; Sharma, A.; Schleifer, R.; She, J.-X. A Pan-Cancer Perspective of Matrix Metalloproteases (MMP) Gene Expression Profile and Their Diagnostic/Prognostic Potential. BMC Cancer 2019, 19, 581. [Google Scholar] [CrossRef]
  214. Rundhaug, J.E. Matrix Metalloproteinases and Angiogenesis. J. Cell Mol. Med. 2005, 9, 267–285. [Google Scholar] [CrossRef]
  215. Stojic, J.; Hagemann, C.; Haas, S.; Herbold, C.; Kühnel, S.; Gerngras, S.; Roggendorf, W.; Roosen, K.; Vince, G.H. Expression of Matrix Metalloproteinases MMP-1, MMP-11 and MMP-19 Is Correlated with the WHO-Grading of Human Malignant Gliomas. Neurosci. Res. 2008, 60, 40–49. [Google Scholar] [CrossRef]
  216. Wu, Z.; Yang, Y.; Chen, M.; Zha, Y. Matrix Metalloproteinase 9 Expression and Glioblastoma Survival Prediction Using Machine Learning on Digital Pathological Images. Sci. Rep. 2024, 14, 15065. [Google Scholar] [CrossRef]
  217. Xue, Q.; Cao, L.; Chen, X.-Y.; Zhao, J.; Gao, L.; Li, S.-Z.; Fei, Z. High Expression of MMP9 in Glioma Affects Cell Proliferation and Is Associated with Patient Survival Rates. Oncol. Lett. 2017, 13, 1325–1330. [Google Scholar] [CrossRef]
  218. Jiguet-Jiglaire, C.; Boissonneau, S.; Denicolai, E.; Hein, V.; Lasseur, R.; Garcia, J.; Romain, S.; Appay, R.; Graillon, T.; Mason, W.; et al. Plasmatic MMP9 Released from Tumor-Infiltrating Neutrophils Is Predictive for Bevacizumab Efficacy in Glioblastoma Patients: An AVAglio Ancillary Study. Acta Neuropathol. Commun. 2022, 10, 1. [Google Scholar] [CrossRef]
  219. Gabelloni, P.; Da Pozzo, E.; Bendinelli, S.; Costa, B.; Nuti, E.; Casalini, F.; Orlandini, E.; Da Settimo, F.; Rossello, A.; Martini, C. Inhibition of Metalloproteinases Derived from Tumours: New Insights in the Treatment of Human Glioblastoma. Neuroscience 2010, 168, 514–522. [Google Scholar] [CrossRef]
  220. Goldie, L.C.; Nix, M.K.; Hirschi, K.K. Embryonic Vasculogenesis and Hematopoietic Specification. Organogenesis 2008, 4, 257–263. [Google Scholar] [CrossRef]
  221. Drake, C.J. Embryonic and Adult Vasculogenesis. Birth Defects Res. C Embryo Today 2003, 69, 73–82. [Google Scholar] [CrossRef] [PubMed]
  222. Kässmeyer, S.; Plendl, J.; Custodis, P.; Bahramsoltani, M. New Insights in Vascular Development: Vasculogenesis and Endothelial Progenitor Cells. Anat. Histol. Embryol. 2009, 38, 1–11. [Google Scholar] [CrossRef]
  223. Tepper, O.M.; Capla, J.M.; Galiano, R.D.; Ceradini, D.J.; Callaghan, M.J.; Kleinman, M.E.; Gurtner, G.C. Adult Vasculogenesis Occurs through in Situ Recruitment, Proliferation, and Tubulization of Circulating Bone Marrow-Derived Cells. Blood 2005, 105, 1068–1077. [Google Scholar] [CrossRef] [PubMed]
  224. Chopra, H.; Hung, M.K.; Kwong, D.L.; Zhang, C.F.; Pow, E.H.N. Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int. 2018, 2018, 9847015. [Google Scholar] [CrossRef]
  225. Blatchley, M.R.; Hall, F.; Wang, S.; Pruitt, H.C.; Gerecht, S. Hypoxia and Matrix Viscoelasticity Sequentially Regulate Endothelial Progenitor Cluster-Based Vasculogenesis. Sci. Adv. 2019, 5, eaau7518. [Google Scholar] [CrossRef]
  226. Huizer, K.; Sacchetti, A.; Dik, W.A.; Mustafa, D.A.; Kros, J.M. Circulating Proangiogenic Cells and Proteins in Patients with Glioma and Acute Myocardial Infarction: Differences in Neovascularization between Neoplasia and Tissue Regeneration. J. Oncol. 2019, 2019, 3560830. [Google Scholar] [CrossRef]
  227. Du, R.; Lu, K.V.; Petritsch, C.; Liu, P.; Ganss, R.; Passegué, E.; Song, H.; Vandenberg, S.; Johnson, R.S.; Werb, Z.; et al. HIF1alpha Induces the Recruitment of Bone Marrow-Derived Vascular Modulatory Cells to Regulate Tumor Angiogenesis and Invasion. Cancer Cell 2008, 13, 206–220. [Google Scholar] [CrossRef]
  228. Moeller, B.J.; Cao, Y.; Li, C.Y.; Dewhirst, M.W. Radiation Activates HIF-1 to Regulate Vascular Radiosensitivity in Tumors: Role of Reoxygenation, Free Radicals, and Stress Granules. Cancer Cell 2004, 5, 429–441. [Google Scholar] [CrossRef]
  229. Jin, D.K.; Shido, K.; Kopp, H.-G.; Petit, I.; Shmelkov, S.V.; Young, L.M.; Hooper, A.T.; Amano, H.; Avecilla, S.T.; Heissig, B.; et al. Cytokine-Mediated Deployment of SDF-1 Induces Revascularization through Recruitment of CXCR4+ Hemangiocytes. Nat. Med. 2006, 12, 557–567. [Google Scholar] [CrossRef]
  230. Folkins, C.; Shaked, Y.; Man, S.; Tang, T.; Lee, C.R.; Zhu, Z.; Hoffman, R.M.; Kerbel, R.S. Glioma Tumor Stem-like Cells Promote Tumor Angiogenesis and Vasculogenesis via Vascular Endothelial Growth Factor and Stromal-Derived Factor 1. Cancer Res. 2009, 69, 7243–7251. [Google Scholar] [CrossRef]
  231. Kioi, M.; Vogel, H.; Schultz, G.; Hoffman, R.M.; Harsh, G.R.; Brown, J.M. Inhibition of Vasculogenesis, but Not Angiogenesis, Prevents the Recurrence of Glioblastoma after Irradiation in Mice. J. Clin. Investig. 2010, 120, 694–705. [Google Scholar] [CrossRef] [PubMed]
  232. Ribatti, D.; Pezzella, F. Vascular Co-Option and Other Alternative Modalities of Growth of Tumor Vasculature in Glioblastoma. Front. Oncol. 2022, 12, 874554. [Google Scholar] [CrossRef] [PubMed]
  233. Pichol-Thievend, C.; Anezo, O.; Pettiwala, A.M.; Bourmeau, G.; Montagne, R.; Lyne, A.-M.; Guichet, P.-O.; Deshors, P.; Ballestín, A.; Blanchard, B.; et al. VC-Resist Glioblastoma Cell State: Vessel Co-Option as a Key Driver of Chemoradiation Resistance. Nat. Commun. 2024, 15, 3602. [Google Scholar] [CrossRef] [PubMed]
  234. Seano, G.; Jain, R.K. Vessel Co-Option in Glioblastoma: Emerging Insights and Opportunities. Angiogenesis 2020, 23, 9–16. [Google Scholar] [CrossRef]
  235. Bae, J.; Kim, M.-H.; Han, S.; Park, S. Development of Tumor-Vasculature Interaction on Chip Mimicking Vessel Co-Option of Glioblastoma. BioChip J. 2023, 17, 77–84. [Google Scholar] [CrossRef]
  236. Nico, B.; Crivellato, E.; Guidolin, D.; Annese, T.; Longo, V.; Finato, N.; Vacca, A.; Ribatti, D. Intussusceptive Microvascular Growth in Human Glioma. Clin. Exp. Med. 2010, 10, 93–98. [Google Scholar] [CrossRef]
  237. Hlushchuk, R.; Riesterer, O.; Baum, O.; Wood, J.; Gruber, G.; Pruschy, M.; Djonov, V. Tumor Recovery by Angiogenic Switch from Sprouting to Intussusceptive Angiogenesis after Treatment with PTK787/ZK222584 or Ionizing Radiation. Am. J. Pathol. 2008, 173, 1173–1185. [Google Scholar] [CrossRef]
  238. Hlushchuk, R.; Makanya, A.N.; Djonov, V. Escape Mechanisms after Antiangiogenic Treatment, or Why Are the Tumors Growing Again? Int. J. Dev. Biol. 2011, 55, 563–567. [Google Scholar] [CrossRef]
  239. Kirschmann, D.A.; Seftor, E.A.; Hardy, K.M.; Seftor, R.E.B.; Hendrix, M.J.C. Molecular Pathways: Vasculogenic Mimicry in Tumor Cells: Diagnostic and Therapeutic Implications. Clin. Cancer Res. 2012, 18, 2726–2732. [Google Scholar] [CrossRef]
  240. Angara, K.; Borin, T.F.; Arbab, A.S. Vascular Mimicry: A Novel Neovascularization Mechanism Driving Anti-Angiogenic Therapy (AAT) Resistance in Glioblastoma. Transl. Oncol. 2017, 10, 650–660. [Google Scholar] [CrossRef]
  241. El Hallani, S.; Boisselier, B.; Peglion, F.; Rousseau, A.; Colin, C.; Idbaih, A.; Marie, Y.; Mokhtari, K.; Thomas, J.-L.; Eichmann, A.; et al. A New Alternative Mechanism in Glioblastoma Vascularization: Tubular Vasculogenic Mimicry. Brain 2010, 133, 973–982. [Google Scholar] [CrossRef] [PubMed]
  242. Yue, W.-Y.; Chen, Z.-P. Does Vasculogenic Mimicry Exist in Astrocytoma? J. Histochem. Cytochem. 2005, 53, 997–1002. [Google Scholar] [CrossRef] [PubMed]
  243. Wang, S.; Ke, Y.; Lu, G.; Song, Z.; Yu, L.; Xiao, S.; Sun, X.; Jiang, X.; Yang, Z.; Hu, C. Vasculogenic Mimicry Is a Prognostic Factor for Postoperative Survival in Patients with Glioblastoma. J. Neurooncol. 2013, 112, 339–345. [Google Scholar] [CrossRef] [PubMed]
  244. Mei, X.; Chen, Y.-S.; Zhang, Q.-P.; Chen, F.-R.; Xi, S.-Y.; Long, Y.-K.; Zhang, J.; Cai, H.-P.; Ke, C.; Wang, J.; et al. Association between Glioblastoma Cell-Derived Vessels and Poor Prognosis of the Patients. Cancer Commun. 2020, 40, 211–221. [Google Scholar] [CrossRef]
  245. Angara, K.; Rashid, M.H.; Shankar, A.; Ara, R.; Iskander, A.; Borin, T.F.; Jain, M.; Achyut, B.R.; Arbab, A.S. Vascular Mimicry in Glioblastoma Following Anti-Angiogenic and Anti-20-HETE Therapies. Histol. Histopathol. 2017, 32, 917–928. [Google Scholar]
  246. Soda, Y.; Marumoto, T.; Friedmann-Morvinski, D.; Soda, M.; Liu, F.; Michiue, H.; Pastorino, S.; Yang, M.; Hoffman, R.M.; Kesari, S.; et al. Transdifferentiation of Glioblastoma Cells into Vascular Endothelial Cells. Proc. Natl. Acad. Sci. USA 2011, 108, 4274–4280. [Google Scholar] [CrossRef]
  247. Niechi, I.; Erices, J.I.; Carrillo-Beltrán, D.; Uribe-Ojeda, A.; Torres, Á.; Rocha, J.D.; Uribe, D.; Toro, M.A.; Villalobos-Nova, K.; Gaete-Ramírez, B.; et al. Cancer Stem Cell and Aggressiveness Traits Are Promoted by Stable Endothelin-Converting Enzyme-1c in Glioblastoma Cells. Cells 2023, 12, 506. [Google Scholar] [CrossRef]
  248. Lathia, J.D.; Mack, S.C.; Mulkearns-Hubert, E.E.; Valentim, C.L.L.; Rich, J.N. Cancer Stem Cells in Glioblastoma. Genes Dev. 2015, 29, 1203–1217. [Google Scholar] [CrossRef]
  249. Wang, Z.; Zhang, H.; Xu, S.; Liu, Z.; Cheng, Q. The Adaptive Transition of Glioblastoma Stem Cells and Its Implications on Treatments. Signal Transduct. Target. Ther. 2021, 6, 124. [Google Scholar] [CrossRef]
  250. Tang, X.; Zuo, C.; Fang, P.; Liu, G.; Qiu, Y.; Huang, Y.; Tang, R. Targeting Glioblastoma Stem Cells: A Review on Biomarkers, Signal Pathways and Targeted Therapy. Front. Oncol. 2021, 11, 701291. [Google Scholar] [CrossRef]
  251. Scully, S.; Francescone, R.; Faibish, M.; Bentley, B.; Taylor, S.L.; Oh, D.; Schapiro, R.; Moral, L.; Yan, W.; Shao, R. Transdifferentiation of Glioblastoma Stem-Like Cells into Mural Cells Drives Vasculogenic Mimicry in Glioblastomas. J. Neurosci. 2012, 32, 12950–12960. [Google Scholar] [CrossRef] [PubMed]
  252. Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour Vascularization via Endothelial Differentiation of Glioblastoma Stem-like Cells. Nature 2010, 468, 824–828. [Google Scholar] [CrossRef] [PubMed]
  253. Huang, Z.; Wu, T.; Liu, A.Y.; Ouyang, G. Differentiation and Transdifferentiation Potentials of Cancer Stem Cells. Oncotarget 2015, 6, 39550–39563. [Google Scholar] [CrossRef] [PubMed]
  254. De Pascalis, I.; Morgante, L.; Pacioni, S.; D’Alessandris, Q.G.; Giannetti, S.; Martini, M.; Ricci-Vitiani, L.; Malinverno, M.; Dejana, E.; Larocca, L.M.; et al. Endothelial Trans-Differentiation in Glioblastoma Recurring after Radiotherapy. Mod. Pathol. 2018, 31, 1361–1366. [Google Scholar] [CrossRef]
  255. Buccarelli, M.; D’Alessandris, Q.G.; Matarrese, P.; Mollinari, C.; Signore, M.; Cappannini, A.; Martini, M.; D’Aliberti, P.; De Luca, G.; Pedini, F.; et al. Elesclomol-Induced Increase of Mitochondrial Reactive Oxygen Species Impairs Glioblastoma Stem-like Cell Survival and Tumor Growth. J. Exp. Clin. Cancer Res. 2021, 40, 228. [Google Scholar] [CrossRef]
  256. Cai, Q.; Fan, H.; Li, X.; Giannotta, M.; Bachoo, R.; Qin, Z. Optical Modulation of the Blood-Brain Barrier for Glioblastoma Treatment. Bio Protoc. 2024, 14, e4920. [Google Scholar] [CrossRef]
  257. Gerritsen, J.K.W.; Broekman, M.L.D.; De Vleeschouwer, S.; Schucht, P.; Nahed, B.V.; Berger, M.S.; Vincent, A.J.P.E. Safe Surgery for Glioblastoma: Recent Advances and Modern Challenges. Neurooncol. Pract. 2022, 9, 364–379. [Google Scholar] [CrossRef]
  258. Singh, N.; Miner, A.; Hennis, L.; Mittal, S. Mechanisms of Temozolomide Resistance in Glioblastoma—A Comprehensive Review. Cancer Drug Resist. 2021, 4, 17–43. [Google Scholar] [CrossRef]
  259. Fan, J.; Liu, J.; Zhang, B.; Wang, X.; Wang, X.; Liang, J.; Li, Y.; Zhang, Y.; Zhang, C.; Yu, S.; et al. GPR65 Contributes to Constructing Immunosuppressive Microenvironment in Glioma. Neurosurg. Rev. 2024, 47, 417. [Google Scholar] [CrossRef]
  260. Rodriguez, S.M.B.; Tataranu, L.G.; Kamel, A.; Turliuc, S.; Rizea, R.E.; Dricu, A. Glioblastoma and Immune Checkpoint Inhibitors: A Glance at Available Treatment Options and Future Directions. Int. J. Mol. Sci. 2024, 25, 10765. [Google Scholar] [CrossRef]
  261. Rosińska, S.; Gavard, J. Tumor Vessels Fuel the Fire in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 6514. [Google Scholar] [CrossRef] [PubMed]
  262. McMahon, D.J.; Gleeson, J.P.; O’Reilly, S.; Bambury, R.M. Management of Newly Diagnosed Glioblastoma Multiforme: Current State of the Art and Emerging Therapeutic Approaches. Med. Oncol. 2022, 39, 129. [Google Scholar] [CrossRef] [PubMed]
  263. Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A Randomized Trial of Bevacizumab for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef] [PubMed]
  264. Moen, M.D. Bevacizumab: In Previously Treated Glioblastoma. Drugs 2010, 70, 181–189. [Google Scholar] [CrossRef]
  265. Liu, Z.-L.; Chen, H.-H.; Zheng, L.-L.; Sun, L.-P.; Shi, L. Angiogenic Signaling Pathways and Anti-Angiogenic Therapy for Cancer. Signal Transduct. Target. Ther. 2023, 8, 1–39. [Google Scholar] [CrossRef]
  266. Batchelor, T.T.; Reardon, D.A.; de Groot, J.F.; Wick, W.; Weller, M. Antiangiogenic Therapy for Glioblastoma: Current Status and Future Prospects. Clin. Cancer Res. 2014, 20, 5612–5619. [Google Scholar] [CrossRef]
  267. Ghiaseddin, A.; Peters, K.B. Use of Bevacizumab in Recurrent Glioblastoma. CNS Oncol. 2015, 4, 157–169. [Google Scholar] [CrossRef]
  268. Friedman, H.S.; Prados, M.D.; Wen, P.Y.; Mikkelsen, T.; Schiff, D.; Abrey, L.E.; Yung, W.K.A.; Paleologos, N.; Nicholas, M.K.; Jensen, R.; et al. Bevacizumab Alone and in Combination with Irinotecan in Recurrent Glioblastoma. J. Clin. Oncol. 2009, 27, 4733–4740. [Google Scholar] [CrossRef]
  269. Kreisl, T.N.; Kim, L.; Moore, K.; Duic, P.; Royce, C.; Stroud, I.; Garren, N.; Mackey, M.; Butman, J.A.; Camphausen, K.; et al. Phase II Trial of Single-Agent Bevacizumab Followed by Bevacizumab Plus Irinotecan at Tumor Progression in Recurrent Glioblastoma. J. Clin. Oncol. 2009, 27, 740–745. [Google Scholar] [CrossRef]
  270. Bayat Mokhtari, R.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination Therapy in Combating Cancer. Oncotarget 2017, 8, 38022–38043. [Google Scholar] [CrossRef]
  271. Ren, X.; Ai, D.; Li, T.; Xia, L.; Sun, L. Effectiveness of Lomustine Combined With Bevacizumab in Glioblastoma: A Meta-Analysis. Front. Neurol. 2021, 11, 603947. [Google Scholar] [CrossRef] [PubMed]
  272. Tsien, C.I.; Pugh, S.L.; Dicker, A.P.; Raizer, J.J.; Matuszak, M.M.; Lallana, E.C.; Huang, J.; Algan, O.; Deb, N.; Portelance, L.; et al. NRG Oncology/RTOG1205: A Randomized Phase II Trial of Concurrent Bevacizumab and Reirradiation Versus Bevacizumab Alone as Treatment for Recurrent Glioblastoma. J. Clin. Oncol. 2023, 41, 1285–1295. [Google Scholar] [CrossRef] [PubMed]
  273. Taal, W.; Oosterkamp, H.M.; Walenkamp, A.M.E.; Dubbink, H.J.; Beerepoot, L.V.; Hanse, M.C.J.; Buter, J.; Honkoop, A.H.; Boerman, D.; de Vos, F.Y.F.; et al. Single-Agent Bevacizumab or Lomustine versus a Combination of Bevacizumab plus Lomustine in Patients with Recurrent Glioblastoma (BELOB Trial): A Randomised Controlled Phase 2 Trial. Lancet Oncol. 2014, 15, 943–953. [Google Scholar] [CrossRef]
  274. She, L.; Su, L.; Liu, C. Bevacizumab Combined with Re-Irradiation in Recurrent Glioblastoma. Front. Oncol. 2022, 12, 961014. [Google Scholar] [CrossRef]
  275. Wick, W.; Gorlia, T.; Bendszus, M.; Taphoorn, M.; Sahm, F.; Harting, I.; Brandes, A.A.; Taal, W.; Domont, J.; Idbaih, A.; et al. Lomustine and Bevacizumab in Progressive Glioblastoma. N. Engl. J. Med. 2017, 377, 1954–1963. [Google Scholar] [CrossRef]
  276. Chinot, O.L.; Wick, W.; Mason, W.; Henriksson, R.; Saran, F.; Nishikawa, R.; Carpentier, A.F.; Hoang-Xuan, K.; Kavan, P.; Cernea, D.; et al. Bevacizumab plus Radiotherapy-Temozolomide for Newly Diagnosed Glioblastoma. N. Engl. J. Med. 2014, 370, 709–722. [Google Scholar] [CrossRef]
  277. Sandmann, T.; Bourgon, R.; Garcia, J.; Li, C.; Cloughesy, T.; Chinot, O.L.; Wick, W.; Nishikawa, R.; Mason, W.; Henriksson, R.; et al. Patients With Proneural Glioblastoma May Derive Overall Survival Benefit From the Addition of Bevacizumab to First-Line Radiotherapy and Temozolomide: Retrospective Analysis of the AVAglio Trial. J. Clin. Oncol. 2015, 33, 2735–2744. [Google Scholar] [CrossRef]
  278. Ahluwalia, M.S.; Rogers, L.R.; Chaudhary, R.; Newton, H.; Ozair, A.; Khosla, A.A.; Nixon, A.B.; Adams, B.J.; Seon, B.K.; Peereboom, D.M.; et al. Endoglin Inhibitor TRC105 with or without Bevacizumab for Bevacizumab-Refractory Glioblastoma (ENDOT): A Multicenter Phase II Trial. Commun. Med. 2023, 3, 120. [Google Scholar] [CrossRef]
  279. Lombardi, G.; Caccese, M.; Padovan, M.; Cerretti, G.; Pintacuda, G.; Manara, R.; Di Sarra, F.; Zagonel, V. Regorafenib in Recurrent Glioblastoma Patients: A Large and Monocentric Real-Life Study. Cancers 2021, 13, 4731. [Google Scholar] [CrossRef]
  280. Caccese, M.; Desideri, I.; Villani, V.; Simonelli, M.; Buglione, M.; Chiesa, S.; Franceschi, E.; Gaviani, P.; Stasi, I.; Caserta, C.; et al. REGOMA-OSS: A Large, Italian, Multicenter, Prospective, Observational Study Evaluating the Efficacy and Safety of Regorafenib in Patients with Recurrent Glioblastoma. ESMO Open 2024, 9, 102943. [Google Scholar] [CrossRef]
  281. Hottinger, A.F.; Ben Aissa, A.; Espeli, V.; Squiban, D.; Dunkel, N.; Vargas, M.I.; Hundsberger, T.; Mach, N.; Schaller, K.; Weber, D.C.; et al. Phase I Study of Sorafenib Combined with Radiation Therapy and Temozolomide as First-Line Treatment of High-Grade Glioma. Br. J. Cancer 2014, 110, 2655–2661. [Google Scholar] [CrossRef] [PubMed]
  282. Grisanti, S.; Ferrari, V.D.; Buglione, M.; Agazzi, G.M.; Liserre, R.; Poliani, L.; Buttolo, L.; Gipponi, S.; Pedersini, R.; Consoli, F.; et al. Second Line Treatment of Recurrent Glioblastoma with Sunitinib: Results of a Phase II Study and Systematic Review of Literature. J. Neurosurg. Sci. 2019, 63, 458–467. [Google Scholar] [CrossRef] [PubMed]
  283. Reardon, D.A.; Vredenburgh, J.J.; Coan, A.; Desjardins, A.; Peters, K.B.; Gururangan, S.; Sathornsumetee, S.; Rich, J.N.; Herndon, J.E.; Friedman, H.S. Phase I Study of Sunitinib and Irinotecan for Patients with Recurrent Malignant Glioma. J. Neurooncol. 2011, 105, 621–627. [Google Scholar] [CrossRef]
  284. de Groot, J.F.; Lamborn, K.R.; Chang, S.M.; Gilbert, M.R.; Cloughesy, T.F.; Aldape, K.; Yao, J.; Jackson, E.F.; Lieberman, F.; Robins, H.I.; et al. Phase II Study of Aflibercept in Recurrent Malignant Glioma: A North American Brain Tumor Consortium Study. J. Clin. Oncol. 2011, 29, 2689–2695. [Google Scholar] [CrossRef]
  285. Nayak, L.; de Groot, J.; Wefel, J.S.; Cloughesy, T.F.; Lieberman, F.; Chang, S.M.; Omuro, A.; Drappatz, J.; Batchelor, T.T.; DeAngelis, L.M.; et al. Phase I Trial of Aflibercept (VEGF Trap) with Radiation Therapy and Concomitant and Adjuvant Temozolomide in Patients with High-Grade Gliomas. J. Neurooncol. 2017, 132, 181–188. [Google Scholar] [CrossRef]
  286. Lombardi, G.; De Salvo, G.L.; Brandes, A.A.; Eoli, M.; Rudà, R.; Faedi, M.; Lolli, I.; Pace, A.; Daniele, B.; Pasqualetti, F.; et al. Regorafenib Compared with Lomustine in Patients with Relapsed Glioblastoma (REGOMA): A Multicentre, Open-Label, Randomised, Controlled, Phase 2 Trial. Lancet Oncol. 2019, 20, 110–119. [Google Scholar] [CrossRef]
  287. Janssen, J.B.E.; Brahm, C.G.; Driessen, C.M.L.; Nuver, J.; Labots, M.; Kouwenhoven, M.C.M.; Sanchez Aliaga, E.; Enting, R.H.; de Groot, J.C.; Walenkamp, A.M.E.; et al. The STELLAR Trial: A Phase II/III Randomized Trial of High-Dose, Intermittent Sunitinib in Patients with Recurrent Glioblastoma. Brain Commun. 2024, 6, fcae241. [Google Scholar] [CrossRef]
  288. Batchelor, T.T.; Mulholland, P.; Neyns, B.; Nabors, L.B.; Campone, M.; Wick, A.; Mason, W.; Mikkelsen, T.; Phuphanich, S.; Ashby, L.S.; et al. Phase III Randomized Trial Comparing the Efficacy of Cediranib as Monotherapy, and in Combination with Lomustine, versus Lomustine Alone in Patients with Recurrent Glioblastoma. J. Clin. Oncol. 2013, 31, 3212–3218. [Google Scholar] [CrossRef]
  289. Xiong, W.; Li, C.; Kong, G.; Wan, B.; Wang, S.; Fan, J. Glioblastoma: Two Immune Subtypes under the Surface of the Cold Tumor. Aging 2022, 14, 4357–4375. [Google Scholar] [CrossRef]
  290. Mougel, A.; Terme, M.; Tanchot, C. Therapeutic Cancer Vaccine and Combinations With Antiangiogenic Therapies and Immune Checkpoint Blockade. Front. Immunol. 2019, 10, 467. [Google Scholar] [CrossRef]
  291. Weathers, S.-P.; Han, X.; Liu, D.D.; Conrad, C.A.; Gilbert, M.R.; Loghin, M.E.; O’Brien, B.J.; Penas-Prado, M.; Puduvalli, V.K.; Tremont-Lukats, I.; et al. A Randomized Phase II Trial of Standard Dose Bevacizumab versus Low Dose Bevacizumab plus Lomustine (CCNU) in Adults with Recurrent Glioblastoma. J. Neurooncol. 2016, 129, 487–494. [Google Scholar] [CrossRef] [PubMed]
  292. Rodà, F.; Caraffi, R.; Picciolini, S.; Tosi, G.; Vandelli, M.A.; Ruozi, B.; Bedoni, M.; Ottonelli, I.; Duskey, J.T. Recent Advances on Surface-Modified GBM Targeted Nanoparticles: Targeting Strategies and Surface Characterization. Int. J. Mol. Sci. 2023, 24, 2496. [Google Scholar] [CrossRef] [PubMed]
  293. Lo Dico, A.; Martelli, C.; Diceglie, C.; Lucignani, G.; Ottobrini, L. Hypoxia-Inducible Factor-1α Activity as a Switch for Glioblastoma Responsiveness to Temozolomide. Front. Oncol. 2018, 8, 249. [Google Scholar] [CrossRef] [PubMed]
  294. Hassn Mesrati, M.; Behrooz, A.B.; Abuhamad, A.Y.; Syahir, A. Understanding Glioblastoma Biomarkers: Knocking a Mountain with a Hammer. Cells 2020, 9, 1236. [Google Scholar] [CrossRef]
Figure 1. Some important morphological aspects of angiogenesis in GBM (E&E staining). (A) The image shows glomeruloid vessels (vessels with multiple lumina) and endothelial multilayering because of endothelial hyperplasia (200× magnification). (B) Atypical, branched vessel network (100× magnification). (C) An island of viable tumor cells encircling the blood vessels in a radial pattern (200× magnification). (D) Ectatic hyalinized vessels and intervening spindle-shaped stromal cells (200× magnification).
Figure 1. Some important morphological aspects of angiogenesis in GBM (E&E staining). (A) The image shows glomeruloid vessels (vessels with multiple lumina) and endothelial multilayering because of endothelial hyperplasia (200× magnification). (B) Atypical, branched vessel network (100× magnification). (C) An island of viable tumor cells encircling the blood vessels in a radial pattern (200× magnification). (D) Ectatic hyalinized vessels and intervening spindle-shaped stromal cells (200× magnification).
Ijms 26 02763 g001
Figure 2. Schematic representation of the “angiogenic switch” in GBM. During tumor progression, the needle of the scale shifts in favor of pro-angiogenic factors, released by GBM cancer cells and TME, under hypoxic conditions, resulting in aberrant growth of GBM vasculature. Abbreviations: GBM, glioblastoma; TME, tumor microenvironment; VEGF, vascular endothelial growth factor; FGF, fibroblast growth factor; TGF-beta, transforming growth factor-beta; PDGF, platelet-derived growth factor; ANG, angiopoietin; MMP, matrix metalloproteinase; PDGFR, platelet-derived growth factor receptor; VEGFR, vascular endothelial growth factor receptor. This figure was created with Servier Medical Art (https://smart.servier.com).
Figure 2. Schematic representation of the “angiogenic switch” in GBM. During tumor progression, the needle of the scale shifts in favor of pro-angiogenic factors, released by GBM cancer cells and TME, under hypoxic conditions, resulting in aberrant growth of GBM vasculature. Abbreviations: GBM, glioblastoma; TME, tumor microenvironment; VEGF, vascular endothelial growth factor; FGF, fibroblast growth factor; TGF-beta, transforming growth factor-beta; PDGF, platelet-derived growth factor; ANG, angiopoietin; MMP, matrix metalloproteinase; PDGFR, platelet-derived growth factor receptor; VEGFR, vascular endothelial growth factor receptor. This figure was created with Servier Medical Art (https://smart.servier.com).
Ijms 26 02763 g002
Table 1. List of the major clinical trials targeting angiogenesis in GBM, including information such as trial name, phase, number of patients, dose, progression-free survival (PFS), overall survival (OS), and treatment-related side effects.
Table 1. List of the major clinical trials targeting angiogenesis in GBM, including information such as trial name, phase, number of patients, dose, progression-free survival (PFS), overall survival (OS), and treatment-related side effects.
Trial Name/
Author
TreatmentPhaseNo. of
Patients
Dose6-Months PFSMedian
OS
Adverse Effects
BRAIN
[268]
BEV alone/
BEV+
Irinotecan
II167 patients with recurrent GBM10 mg/kg every 2 weeks of BEV alone
or
10 mg/kg of BEV + 340 mg/m2 or 125 mg/m2 of Irinotecan (if taking or not taking enzyme-inducing antiepileptic drugs, respectively) every 2 weeks
42.6%
BEV alone
50.3%
BEV+
Irinotecan
9.2 months
BEV alone
8.7 months
BEV+
Irinotecan
Hypertension,
fatigue, headache,
convulsion,
diarrhea,
nausea, and
neutropenia
NCI
06-C-0064E
[269]
BEVII48 patients with recurrent GBM10 mg/kg every 2 weeks29%31 weeksThromboembolic events,
hypertension,
hypophosphatemia, and thrombocytopenia
BELOB
[273]
BEV/
Lomustine alone or BEV + Lomustine
II153 patients with recurrent GBM110 mg/m2 every 6 weeks of Lomustine alone or
10 mg/kg every 2 weeks of BEV alone
or
90/110 mg/m2 every 6 weeks of Lomustine combined with 10 mg/kg every 2 weeks of BEV
13%
Lomustine alone
16% BEV alone
42% BEV + Lomustine
8 months
Lomustine
alone
8 months
BEV alone
12 months BEV+
Lomustine
Hypertension,
fatigue, and
infections
EORTC
26101
[275]
BEV + Lomustine or Lomustine aloneIII437
patients with progressive GBM
10 mg/kg every 2 weeks of BEV + 90 mg/m2 every 6 weeks of Lomustine
or
110 mg/m2 every 6 weeks of Lomustine alone
Not
available
9.1 months BEV + Lomustine
8.6 months Lomustine alone
Pulmonary embolism,
arterial hypertension,
and
hematologic toxic
effects
RTOG
0825
[263]
BEV/placebo
+ TMZ +
radiotherapy
III637 patients with newly diagnosed GBM10 mg/kg every 2 weeks of BEV + TMZ + radiotherapyNot
available
15.7 monthsLymphopenia, neutropenia, fatigue, and
thrombocytopenia
AVAglio
[276]
BEV/placebo + TMZ +
radiotherapy
III921 patients with newly diagnosed GBM10 mg/kg every 2 weeks of BEV + TMZ + radiotherapyNot
available
16.8 months
BEV
16.7 months
placebo
Hypertension, proteinuria, thromboembolia, and wound
healing complications
ENDOT
[278]
Carotuximab
(TRC105) alone or TRC105 + BEV
following radiation, TMZ, and BEV therapy
II22 patients with GBM that had progressed after chemoradiation10 mg/kg weekly
TRC105 alone/
10 mg/kg split into two doses with 3 mg/kg administered on cycle 1 day 8 and 7 mg/kg administered on cycle 1 day 11
with TRC105 + BEV
13.3%
(calculated for the 15
evaluable patients treated with BEV +
TRC105)
5.7 months
(calculated for the 15
evaluable patients treated with BEV + TRC105)
Headache, epistaxis,
fatigue, TIA,
lower leg edema,
pulmonary embolism,
and
sinusitis
REGOMA
[286]
Regorafenib
or Lomustine
II119 patients with recurrent GBM160 mg once daily for the first 3 weeks of each 4-week cycle of Regorafenib or 110 mg/m2 every 6 weeks of LomustineNot
available
7.4 months
in the Regorafenib group/
5.6 months in the
Lomustine group
Hand–foot skin reaction,
increased lipase,
increased blood bilirubin, decreased platelet/
lymphocyte count,
and
neutropenia
Hottinger, A.F
[281]
Sorafenib + TMZ + radiotherapyI17 patients with newly diagnosed GBM400 mg 2 times daily86.7%17.8 monthsThrombocytopenia,
neutropenia, alopecia,
nausea, vomiting,
hypophosphatemia,
and
fatigue
STELLAR
[287]
Sunitinib
or Lomustine (in the control arm)
I32 patients with recurrent GBM in part I
37 patients with recurrent GBM in part II
Part I
300 mg Q1W of Sunitinib
or
110 mg/m2
of Lomustine once every six weeks
Part II
700 mg Q2W of Sunitinib
or
110 mg/m2
of Lomustine once every six weeks
8% in part I with Sunitinib vs.
29% with
Lomustine
14% in part II with Sunitinib vs. 15% with
Lomustine
6.5 months in part I with Sunitinib
vs. 4.7 months with
Lomustine
4.7 months in part II with Sunitinib vs. 7 months with
Lomustine
Thrombocytopenia,
fatigue,
leukopenia,
diarrhea,
nausea,
and
vomiting
REGAL
[288]
Cediranib alone/in combination with Lomustine vs. Lomustine plus placeboIII325 patients with
recurrent GBM
Cediranib alone (30 mg), Cediranib (20 mg) + Lomustine (110 mg/m2),
or
Lomustine
(110 mg/m2) + placebo
16%
Cediranib alone
35%
Cediranib + Lomustine 25%
Lomustine alone
8.0 months Cediranib alone,
9.4 months Cediranib +
Lomustine
9.8 months
Lomustine alone
Diarrhea,
thrombocytopenia,
neutropenia,
and
hypertension
De Groot, J.F
[284]
AfliberceptII42 patients with GBM and 16 patients with anaplastic glioma4 mg/kg on day 1 of every 2-week cycle.7.7% for
GBM
39 weeksFatigue,
thromboembolia,
wound healing,
and
CNS ischemia
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ballato, M.; Germanà, E.; Ricciardi, G.; Giordano, W.G.; Tralongo, P.; Buccarelli, M.; Castellani, G.; Ricci-Vitiani, L.; D’Alessandris, Q.G.; Giuffrè, G.; et al. Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. Int. J. Mol. Sci. 2025, 26, 2763. https://doi.org/10.3390/ijms26062763

AMA Style

Ballato M, Germanà E, Ricciardi G, Giordano WG, Tralongo P, Buccarelli M, Castellani G, Ricci-Vitiani L, D’Alessandris QG, Giuffrè G, et al. Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. International Journal of Molecular Sciences. 2025; 26(6):2763. https://doi.org/10.3390/ijms26062763

Chicago/Turabian Style

Ballato, Mariagiovanna, Emanuela Germanà, Gabriele Ricciardi, Walter Giuseppe Giordano, Pietro Tralongo, Mariachiara Buccarelli, Giorgia Castellani, Lucia Ricci-Vitiani, Quintino Giorgio D’Alessandris, Giuseppe Giuffrè, and et al. 2025. "Understanding Neovascularization in Glioblastoma: Insights from the Current Literature" International Journal of Molecular Sciences 26, no. 6: 2763. https://doi.org/10.3390/ijms26062763

APA Style

Ballato, M., Germanà, E., Ricciardi, G., Giordano, W. G., Tralongo, P., Buccarelli, M., Castellani, G., Ricci-Vitiani, L., D’Alessandris, Q. G., Giuffrè, G., Pizzimenti, C., Fiorentino, V., Zuccalà, V., Ieni, A., Caffo, M., Fadda, G., & Martini, M. (2025). Understanding Neovascularization in Glioblastoma: Insights from the Current Literature. International Journal of Molecular Sciences, 26(6), 2763. https://doi.org/10.3390/ijms26062763

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop