• Users Online: 155
  • Print this page
  • Email this page


 
 
Table of Contents
REVIEW
Year : 2018  |  Volume : 1  |  Issue : 2  |  Page : 35-42

Molecular mechanisms involved in angiogenesis and potential target of antiangiogenesis in human glioblastomas


Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China

Date of Web Publication30-Apr-2018

Correspondence Address:
Dr. Bao-Hui Liu
Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/glioma.glioma_10_17

Rights and Permissions
  Abstract 

Glioblastoma multiforme (GBM) is the most devastating and common primary malignant brain tumor in adults. Angiogenesis as a hallmark in glioblastoma has attracted more and more attention of the research community. Over the past years, several systematic studies have provided vital results in this field discovering alterations in unanticipated number of genes and other regulatory factors and the associated signaling pathways. Recent discoveries about such genes and signaling pathway associated with angiogenesis in GBM provide a picture of the genomic routes in angiogenesis and antiangiogenesis pathways and may lay the foundation for improved antiangiogenesis therapy and clinical care. In this review, we discuss several such recent progresses in the genes associated with GBM angiogenesis/antiangiogenesis pathways and explore the potential new targets for GBM treatment.

Keywords: Angiogenesis, antiangiogenic therapy, genes, glioblastoma, resistance, signaling pathway


How to cite this article:
Xu Y, Yuan FE, Chen QX, Liu BH. Molecular mechanisms involved in angiogenesis and potential target of antiangiogenesis in human glioblastomas. Glioma 2018;1:35-42

How to cite this URL:
Xu Y, Yuan FE, Chen QX, Liu BH. Molecular mechanisms involved in angiogenesis and potential target of antiangiogenesis in human glioblastomas. Glioma [serial online] 2018 [cited 2023 Oct 2];1:35-42. Available from: http://www.jglioma.com/text.asp?2018/1/2/35/231495


  Introduction Top


Glioblastoma multiforme (GBM), WHO grade IV astrocytoma, is one of the most common primary malignant tumors of the central nervous system in adults.[1],[2] Despite the multimodal therapeutic strategies, including surgery, radiation, and chemotherapy with temozolomide (TMZ), the median overall survival of GBM patients remains as low as a little over 1 year, with the overall 5-year survival rate of <5%.[3],[4]

It is well known that the angiogenesis is a hallmark of cancers.[5] "Angiogenic switch" is a process through which tumor cells develop an angiogenic phenotype, thus initiating angiogenesis.[6] The "angiogenic switch" can promote tumorigenesis and invasion through activating oncogenes and/or downregulating tumor-suppressor genes while upregulating the expression/signaling of angiogenic pathways.[7],[8],[9] Thus, angiogenesis is termed an important influencing factor in tumor growth, and now, increasing number of studies have investigated the potential of antiangiogenic treatment strategy against GBM.[10],[11]

In this review, we discuss several recent progresses in the genes associated with GBM angiogenesis/antiangiogenesis pathways and explore potential new targets in this regard.


  Genes Potentially Related to Glioblastoma Angiogenesis Top


There are a number of genes related to the development and progression of glioblastoma angiogenesis. Here, we focus on several advanced genes.

YKL-40 (chitinase 3 like 1, CHI3L1) is a highly conserved 40-kDa chitin or heparin-binding glycoprotein, which places it into the family of chitinase-like proteins. The gene expression profiling has identified YKL-40 as a cancer-promoter gene in GBM.[12],[13] YKL-40 activates downstream signals focal adhesion kinase (FAK) 397 and extracellular-regulated kinase (ERK) 1/2 through stimulating coordination of Syndecan-1 (Syn-1) and integrin αvβ5 and elevates the expression of vascular endothelial growth factor (VEGF) in glioblastoma cells.[14] Consistently, all these findings established YKL-40 as a proangiogenic factor promoting angiogenesis in GBM through activating coordination of Syn-1, integrin αvβ5, and vascular endothelial cadherin (VE-cad), which stimulates intracellular signaling via FAK861 and ERK 1/2[15] and VEGF receptor 2 (VEGFR2, Flk-1) expression to enhance the endothelial cell (EC)-mediated angiogenesis.[16]

Cluster of differentiation 147 (CD147), a highly glycosylated transmembrane glycoprotein with two Ig-like extracellular domains that belong to the immunoglobulin superfamily, plays a crucial role in GBM angiogenesis.[17] CD147 was recently reported to function via a new mechanism that induces the effective formation of angiogenic niches through the positive feedback with insulin-like growth factor-1 (IGF-1).[18] It is known that VEGF, IGF-1, and hypoxia-inducible factor-1 alpha (HIF-1α) influence angiogenesis of GBM, and CD147 has been reported to induce angiogenesis via HIF-1α-VEGF signaling pathway.[19]

Leucine rich repeat containing 4/Netrin-G ligand-2 (LRRC4/NGL-2), a gene expressed in brain specifically, is a member of the LRRC4/NGL family and is a part of the leucine-rich repeat superfamily.[20] It has been reported that LRRC4/NGL-2 might inhibit cell invasion through regulating the expression of C-X-C chemokine receptor type 4 (CXCR4) and Stromal cell-derived factor 1α (SDF-1α)/CXCR4 axis in vitro.[21] LRRC4 expression might eliminate VEGF-mediated neovascularization, which was demonstrated by Wu et al.[22] where VEGF induced tube-like structure formation in the human umbilical vein endothelial cell was lost in LRRC4β cells. Conceivably, these results portray LRRC4/NGL-2 as a candidate for tumor-suppressor gene, acting against the angiogenesis in GBM. However, further studies are essential to explore detailed mechanisms.

In brief, YKL-40 and CD147 are identified as promoters of tumor through stimulating FAK397 and ERK1/2 pathways and HIF-1α-VEGF signaling pathway, respectively. Conversely, LRRC4/NGL-2 may suppress GBM by eliminating VEGF-mediated angiogenesis.


  Micro-Rnas Potentially Related to Glioblastoma Angiogenesis Top


In recent years, more and more studies have shown microRNAs (miRNAs), as a novel class of noncoding tumor-suppressor or oncogenes, might play key roles in the process of tumorigenesis. In GBM, a group of miRNAs, termed as angiomiRs or anti-angiomiRs, that can contribute toward tumor vascularization was recently identified.[23] Here, we discuss a few of such angiomiRs and anti-angiomiRs.

Hypoxia can augment angiogenesis by influencing miRNA expression in GBM cells. Overexpression of miR-210-3p can increase angiogenesis by inducing HIF, VEGF, and carbonic anhydrase IX (CA9) transcriptional activity. Under hypoxic condition, inhibiting the expression of miR-210-3p impedes HIF-mediated induction of VEGF and CA9 activity, resulting in decreased tumor growth and reduced vascular density, in vivo.[24] Similarly, the expression of miR-21 is usually high in glioblastoma, and HIF-1α is the center for miR-21-induced upregulation of VEGF. This significant correlation of miR-21 with HIF-1α and VEGF implied a link between miR-21 and glioblastoma angiogenesis.[25] miR-296 is another angiomiR, and it has been found that, in GBM, the angiogenic growth factors such as VEGF can elevate miR-296 expression in endothelial cells. Furthermore, downregulation of miR-296 might inhibit human endothelial cells from expressing morphologic characteristics associated with angiogenesis, while upregulation of miR-296 might result in the induction of the same.[26]

miR-15b is an anti-angiomiR which decreases angiogenesis, largely suppressing capillary-like tube formation, via Neuropilin 2 (NRP-2) downregulation.[27] miR-299, located at chromosome 14q32.31, is characterized as a candidate that could suppress GBM. Overexpression or knockdown of miR-299 apparently resulted in suppressing or promoting tumor-induced angiogenesis, respectively, under in vitro assessments. Furthermore, miR-299 overexpression not only suppressed tumor growth but also reduced tumor microvascular density (MVD), inhibiting the tube formation of vascular endothelial cells, in xenograft glioblastoma models.[28]

As we have discussed above, miRNAs play a complex role in the process of GBM angiogenesis. miR-210-3p, miR-21, and miR-296 promote angiogenesis, while miR-15b and miR-299 are potential targets to antiangiogenesis. However, the specific mechanisms of these miRNAs regulating angiogenesis in GBM are still unclear.


  Receptors and Signaling Pathways Potentially Involved in Glioblastoma Multiforme Angiogenesis Top


In GBM, several genetic and molecular alterations modify major signaling pathways that result in brain tumor angiogenesis. Although the involvement of several well-known angiogenic pathways is indubitable, the yet to be understood underlying complex interactions among them, and with few additional unknown players, potentially contribute to the angiogenesis of GBM. Here, we discuss several receptors and signaling pathways potentially involved in GBM angiogenesis.

Three VEGFRs have been reported: VEGFR1, VEGFR2, and VEGFR3, all of which are related to angiogenesis. phosphatidylinositol 3-kinase (PI3K) family members are lipid kinases and play a part in various cellular processes, such as proliferation, migration, and metabolism.[29] The serine/threonine kinase sites (Thr308 and Ser473) on these P13K family members will recruit the downstream AKT to inner membranes and phosphorylate it. It has been found that mTOR pathway regulates many physiological and pathological processes in angiogenesis of tumor cells. In this signaling pathway, mTOR acts not only as an upstream regulator but also as a downstream effector.[30] Furthermore, inhibiting the PI3K pathway not only limits tumor cell growth but also blocks tumor angiogenesis. Interestingly, the PI3K pathway plays a key role in regulating VEGF and VEGFR.[31] In addition, mice treated with FK228 (a histone deacetylase inhibitor) in combination with TMZ significantly decreased the expression of the key members in PI3K/AKT/mTOR signal pathway and significantly enhanced the antitumor efficacy.[32] The phosphorylation of Akt/PKB may inhibit apoptosis and mTOR activation. Endothelial nitric oxide synthase (eNOS) is also involved in angiogenesis through Akt/PKB pathway.[33] Interestingly, overexpression of VEGFR2 is associated with proliferation of tumor cells in GBM. However, the process may be independent of VEGF.[34],[35] The platelet-derived growth factor (PDGF) family has a PDGF/VEGF homology domain which is a common domain to growth factors. Two PDGF receptors (PDGFRs) have been reported, PDGFRα and PDGFRβ.[36],[37] PDGFRβ contributes to tumor expansion by activating genetic alterations associating with DNA karyokinesis and synthesis, through RAS/RAF/ERK/MAPK signaling pathway. Furthermore, signal transduction and transcription activator (STAT) pathway is also involved by both PDGFRα and PDGFRβ, through various phosphorylated tyrosine residues produced by autophosphorylation of PDGFR.[38],[39],[40]

The epidermal growth factor receptor (EGFR) is associated with tumor growth and angiogenesis and is found activated in nearly 50% of primary glioblastomas (GBM).[41] It is structurally related to four human receptors including Her1, Her2, Her3, and Her4, all of which belong to ErbB family. Activation of EGFR can activate a variety of molecules such as SH1, SH2, and STAT, through ligand binging. EGFR is reportedly involved in several signal pathways, including PI3K/AKT/mTOR and RAS/RAF/ERK/MAPK, WNT/β-catenin, and transforming growth factor (TGF)-β signaling.[42],[43] EGFR can modulate both TGF-β pathway, which is involved in invasion and migration of glioblastoma, and Notch1 pathway, which is fundamental to normal development. What's more, downregulation of EGFR may induce tumor cell proliferation, tumor angiogenesis in glioblastoma, and apoptosis in breast cancer.[44],[45],[46] In addition, RAS/MAPK and PI3K/AKT/mTOR signaling pathway regulates cell proliferation, differentiation, tumor angiogenesis, and survival in glioblastoma.[39],[47]

Fibroblast growth factor receptor (FGFR) modulates a series of processes in tumor cells, including FGF-mediated migration and proliferation. FGFR plays a key role in survival and angiogenesis of glioblastoma cell through PI3K/AKT/mTOR signaling pathway. FGF1-FGFR also activates Jnk-p38-MAPK and STAT3-NF-κB pathways, which are crucially associated with tumorigenesis, cell proliferation, and so on.[48] Furthermore, FGF2 has been reported as a prognostic biomarker in glioblastoma [Figure 1] and [Table 1].[49]
Figure 1: Associated receptors and their signaling pathways involved in angiogenesis. RAS/RAF/ERK/MAPK and PI3K/Akt signaling pathways activation can produce a variety of processes involved in angiogenesis such as cell proliferation, migration, and survival

Click here to view
Table 1: Major angiogenic receptors in GBM

Click here to view


The Notch signaling pathway is an intercellular signaling pathway which can affect various cellular processes during embryonic and postnatal development of cells.[50] The Notch1 is naturally expressed in developing vasculature and in extravascular tissues. However, Dll4 and Notch4 expression is specified to vascular endothelial cells.[51] El Hindy et al.[52] demonstrated that the coactions between tumor cells, endothelial cells, and microglia/macrophages may regulate the Dll4-Notch signaling determining typical or atypical vascular patterns in glioblastoma. Furthermore, inhibiting Dll4-Notch signaling pathway can block the formation of functional vessels, but interestingly, it can promote the formation of nonfunctional vessels.[53],[54]

Briefly, VEGFR, EGFR, and FGFR promote angiogenesis via various signaling pathways including Notch signaling pathway, which is a key signaling pathway.


  Potential Mechanisms of Resistance to Antiangiogenic Therapy Top


A formidable challenge to antiangiogenic therapy is the development of resistance to the therapy. Here, we discuss several related potential molecular mechanisms.

Although bevacizumab has been approved for the treatment of GBM patients and most patients are benefited from the treatment initially, these effects are usually limited in confronting tumor regrowth.[55],[56] It has been reported that Dll4-Notch signaling pathway was activated and regulated in the growth of GBM.[57] Interestingly, Dll4 and Jag1 have opposing effects on angiogenesis in GBM.[58] Consistently, it has been reported that when Dll4 is expressed, and Jag1 is not in GBM, then the tumor can develop resistance to bevacizumab mediated antiangiogenesis therapy.[59] The β1 integrins are found to be involved in a series of physiological and pathophysiological processes, including tumorigenesis, proliferation, invasion, and migration of tumor cells, and also have recently been reported to influence the therapeutic resistance in multiple solid cancer models.[60] Recent research demonstrated that blocking β1 integrin in GBM could suppress the associated angiogenesis and tumor growth. Furthermore, the study also showed that suppressing β1 integrin may reverse the resistance to bevacizumab in GBM.[61]

Hypoxia-inducible gene 2 (HIG2) is a marker of hypoxia and is found elevated in several cancer conditions.[62] HIG2 can serve as a diagnostic marker for several cancers and as a potential target for antiangiogenesis therapy.[63] A recent study showed a positive correlation of HIG2 with VEGFA and HIF-1α expression, which ultimately results in resistance against bevacizumab.[64] What's more, HIG2 upregulation may contribute to bevacizumab resistance through other cellular processes such as inhibiting apoptosis, promoting lipid biosynthesis, and activation of WNT signaling pathway.[63],[65],[66]

STAT3 is a receptor that is activated by ligand interaction and can be overexpressed and constitutively activated in tumors.[67],[68] A recent study has found that AZD1480 (a STAT3 inhibitor) combined with cediranib markedly reduced the volume and microvessel density (MVD) of GBM, implying that the STAT3 pathway may mediate resistance to antiangiogenic therapy and the regulation of which might be of use in treating the condition.[69]

Cylindromatosis (CYLD), described as a mutant tumor suppressor gene in familial CYLD,[70] can regulate diverse processes of tumor cell's biology including proliferation, migration, inflammation, and survival.[71] Recently, a reduction in the CYLD expression is noted in gliomas, establishing an inverse relation with the tumor grade and patient prognosis.[72] A recent study found CYLD was downregulated in human GBM tissues under hypoxic condition and acted as an important regulator of hypoxia-induced angiogenesis in GBM.[73] Thus, it may contribute to the antiangiogenesis resistance, and may affect the long-term efficacy of anti-VEGF therapy. On inhibiting VEGF signaling, the tumor and its microenvironment release alternative proangiogenic growth factors to promote VEGF-independent angiogenesis.[74],[75]

Phosphatase and tensin homolog (PTEN), a tumor-suppressor gene, is often inactivated in cancer.[76] Specifically, in GBM, loss of the PTEN leads to VEGFR-2 expression in tumor cells, which may contribute to resistance against antiangiogenic treatments. Furthermore, a recent study showed that overexpression of VEGFR2 in tumor cells could develop early resistance to chemotherapy with TMZ and antiangiogenesis therapy with bevacizumab, in GBM.[77]

The resistance to antiangiogenesis is an emerging severe problem of antiangiogenesis therapy. Expression of Dll4-Notch, HIG2, and CYLD and loss of PTEN are described as potential mechanisms which lead to the resistance.


  New Regulated Factors and Target for Antiangiogenesis Top


As mentioned before, angiogenesis is one of the most obvious hallmarks of glioblastoma, which contrasts glioblastoma from normal tissues.[5],[11] Thus, antiangiogenesis therapy has become a worthy strategy to consider in glioblastoma patients. VEGF has been found to play an important role in the angiogenesis of GBM,[78] and inhibiting the expression of VEGF always seems like to be the most effective therapeutic strategy to inhibit GBM growth.[79] However, this strategy did not meet the expectations in human patients, as shown by studies over the past few years. Here, we discuss several new targets for antiangiogenesis.

Vasculogenic mimicry (VM) [Figure 2] is a newly discovered vascular-like network structure. Maniotis et al.[80] defined and described this structure for malignant melanoma for the first time in 1999. VM is EC-independent, consisting of tumor cells and matrix, and is associated with poor prognosis in GBM patients.[81],[82],[83] More and more emerging studies have confirmed that the involvement of a series of genes and several molecular pathways in VM. Thus, these molecular mechanisms of VM may provide potential targets for antiangiogenic therapy. SU1498 and AZD2171, the inhibitors of VEGFR-2 kinase, have been reported to reduce VM formation in GBM cell lines accompanied by reduced proliferation and tumor growth, both in vivo and in vitro.[84]
Figure 2: Tumor blood supply pattern based on VM. The possible connection between VM channels and the endothelium-dependent vessels: a continuous process of vessels development: (A) VM channels, (B) mosaic vessels, and (C) the endothelium-dependent vessels. (D) Tumor proliferation which leads to hypoxia acts as a trigger to stimulate tumor blood supply cycle. VM: Vasculogenic mimicry

Click here to view


It has been reported that downregulating HIF-1α and mTOR signaling pathway through rapamycin and mTOR siRNA, respectively, may inhibit VM formation.[85] This study not only provided the evidence for a more integrated signaling cascade for VM formation but also demonstrated mTOR as a potential therapeutic target in glioblastoma. Similarly, the TGF inhibitor isoxanthohumol [86] impairs VM formation and the matrix metalloproteinase (MMP) inhibitor chemically modified tetracycline [87] and downregulates VE-cad and MMPs in cancer cells. Francescone et al.[88] showed that targeting VEGFR-2 using Flk-1 shRNA in GBM derived cell lines, limited VM formation, and subsequently inhibited the development of tumors. The results of this study show a key role of VEGFR2 in the formation of VM in GBM and provide a possible therapeutic target, clarifying a molecular mechanism mediating tumor aggressiveness. Chromodomain helicase DNA-binding protein 5 (CHD5) is a novel candidate as a suppressor gene, which is deleted from 1p36 in glioblastomas.[89] A recent study showed that expressing CDH5 in glioblastoma stem cells (GSCs) could benefit GSC-derived neovasculogenesis in GBM, especially under hypoxia.[90] This study also revealed the novel angiogenesis mechanisms contributed by GSCs. These studies on VM is hoped to contribute to the development of antiangiogenesis and antitumor treatments in future.

As mentioned above, hypoxia is one of the most important features of GBM. This condition promotes the expression of HIF-1α, which plays a key role in regulating the biological behaviors of tumor under hypoxic microenvironment. Recent studies demonstrate that vincristine promotes an antiangiogenic effect through the inhibition of HIF-1α in glioblastomas under hypoxia.[91],[92] A recent study has established a positive correlation between the expression of CD31 and HIF-1α in human GBM specimens [93] and characterized N-cadherin and ADAM-10 as biomarkers of aggressiveness in the same specimens.[94] This result may provide a new target for antiangiogenesis therapy. In addition, HIF-1α also induces the overexpression of anterior gradient protein 2 (AGR2), which is important in promoting angiogenesis not only at the level of RNA but also at the level of protein in GBM. Consistently, inhibition of AGR2 may reduce proangiogenesis in GBM cell lines.[95],[96],[97] Thus, this mechanism may provide a potential target to enhance antiangiogenic strategies.

In addition, emerging studies have identified miRNAs as the inhibitors of tumor angiogenesis. A recent study showed that miRNA-7 could effectively suppress angiogenesis by targeting O-GlcNAc transferase (OGT) gene; thus, tube formation and sprouting were suppressed in GBM xenograft in mice.[98] The increased expression of miRNA-16 in GBM cells has a critical effect on repressing endothelial function and angiogenesis by targeting Bmi-1.[99] miR-137 inhibits angiogenesis and proliferation in human glioblastoma by targeting EZH2, and the overexpression of EZH2 can reverse the effect,[100] revealing miRNA-137 as a new target for GBM treatments.

Besides VEGF, VM has been demonstrated to play a critical role in angiogenesis of GBM. Furthermore, AGR2, miRNA-7, miRNA-16, and miRNA-137 may provide a new target for GBM treatments.


  Conclusions and Perspectives Top


Glioblastoma is the most common primary malignant tumor of the central nervous system in adults. In this review, we discussed a series of studies on angiogenesis in glioblastoma, including associated genes, signaling pathways, mechanism of resistance to antiangiogenic therapy, and new strategy to antiangiogenesis. The current antiangiogenic treatments targeting VEGF, such as bevacizumab, have not shown obvious improvement in the overall survival of GBM patients due to the presence of VEGF-independent angiogenic pathway in GBM cells. What's more, the resistance to antiangiogenic therapies has become the new challenge, and the molecular mechanisms of the resistance to antiangiogenic strategy remain to be further explored. VM may be a new potential target for antiangiogenic therapy. The more we understand the mechanisms of glioblastoma angiogenesis, the greater our chances to develop a successful antiangiogenic therapy in GBM patients that effectively improve their quality of life are.

Financial support and sponsorship

Nil.

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol 2016;131:803-20.  Back to cited text no. 1
    
2.
Ostrom QT, Gittleman H, Liao P, Vecchione-Koval T, Wolinsky Y, Kruchko C, et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010-2014. Neuro Oncol 2017;19:v1-88.  Back to cited text no. 2
    
3.
Liu B, Dong H, Lin X, Yang X, Yue X, Yang J, et al. RND3 promotes snail 1 protein degradation and inhibits glioblastoma cell migration and invasion. Oncotarget 2016;7:82411-23.  Back to cited text no. 3
    
4.
Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: Primary brain and central nervous system tumors diagnosed in the United States in 2005-2009. Neuro Oncol 2012;14 Suppl 5:v1-49.  Back to cited text no. 4
    
5.
Hanahan D, Weinberg RA. Hallmarks of cancer: The next generation. Cell 2011;144:646-74.  Back to cited text no. 5
    
6.
Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer 2003;3:401-10.  Back to cited text no. 6
    
7.
Dameron KM, Volpert OV, Tainsky MA, Bouck N. Control of angiogenesis in fibroblasts by p53 regulation of thrombospondin-1. Science 1994;265:1582-4.  Back to cited text no. 7
    
8.
Satchi-Fainaro R, Puder M, Davies JW, Tran HT, Sampson DA, Greene AK, et al. Targeting angiogenesis with a conjugate of HPMA copolymer and TNP-470. Nat Med 2004;10:255-61.  Back to cited text no. 8
    
9.
Takano S, Yamashita T, Ohneda O. Molecular therapeutic targets for glioma angiogenesis. J Oncol 2010;2010:351908.  Back to cited text no. 9
    
10.
Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, et al. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 1999;284:1994-8.  Back to cited text no. 10
    
11.
Wang N, Jain RK, Batchelor TT. New directions in anti-angiogenic therapy for glioblastoma. Neurotherapeutics 2017;14:321-32.  Back to cited text no. 11
    
12.
Nigro JM, Misra A, Zhang L, Smirnov I, Colman H, Griffin C, et al. Integrated array-comparative genomic hybridization and expression array profiles identify clinically relevant molecular subtypes of glioblastoma. Cancer Res 2005;65:1678-86.  Back to cited text no. 12
    
13.
Tanwar MK, Gilbert MR, Holland EC. Gene expression microarray analysis reveals YKL-40 to be a potential serum marker for malignant character in human glioma. Cancer Res 2002;62:4364-8.  Back to cited text no. 13
    
14.
Francescone RA, Scully S, Faibish M, Taylor SL, Oh D, Moral L, et al. Role of YKL-40 in the angiogenesis, radioresistance, and progression of glioblastoma. J Biol Chem 2011;286:15332-43.  Back to cited text no. 14
    
15.
Shao R, Hamel K, Petersen L, Cao QJ, Arenas RB, Bigelow C, et al. YKL-40, a secreted glycoprotein, promotes tumor angiogenesis. Oncogene 2009;28:4456-68.  Back to cited text no. 15
    
16.
Faibish M, Francescone R, Bentley B, Yan W, Shao R. A YKL-40-neutralizing antibody blocks tumor angiogenesis and progression: A potential therapeutic agent in cancers. Mol Cancer Ther 2011;10:742-51.  Back to cited text no. 16
    
17.
Fei F, Li S, Fei Z, Chen Z. The roles of CD147 in the progression of gliomas. Expert Rev Anticancer Ther 2015;15:1351-9.  Back to cited text no. 17
    
18.
Chen Y, Gou X, Ke X, Cui H, Chen Z. Human tumor cells induce angiogenesis through positive feedback between CD147 and insulin-like growth factor-I. PLoS One 2012;7:e40965.  Back to cited text no. 18
    
19.
Wang CH, Yao H, Chen LN, Jia JF, Wang L, Dai JY, et al. CD147 induces angiogenesis through a vascular endothelial growth factor and hypoxia-inducible transcription factor 1α-mediated pathway in rheumatoid arthritis. Arthritis Rheum 2012;64:1818-27.  Back to cited text no. 19
    
20.
Wang JR, Qian J, Dong L, Li XL, Tan C, Li J, et al. Identification of LRRC4, a novel member of leucine-rich repeat (LRR) superfamily, and Its expression analysis in brain tumor. Prog Biochem Biophys 2002;29:233-9.  Back to cited text no. 20
    
21.
Wu M, Chen Q, Li D, Li X, Li X, Huang C, et al. LRRC4 inhibits human glioblastoma cells proliferation, invasion, and proMMP-2 activation by reducing SDF-1 alpha/CXCR4-mediated ERK1/2 and Akt signaling pathways. J Cell Biochem 2008;103:245-55.  Back to cited text no. 21
    
22.
Wu M, Huang C, Li X, Li X, Gan K, Chen Q, et al. LRRC4 inhibits glioblastoma cell proliferation, migration, and angiogenesis by downregulating pleiotropic cytokine expression and responses. J Cell Physiol 2008;214:65-74.  Back to cited text no. 22
    
23.
Wang S, Olson EN. AngiomiRs – Key regulators of angiogenesis. Curr Opin Genet Dev 2009;19:205-11.  Back to cited text no. 23
    
24.
Agrawal R, Pandey P, Jha P, Dwivedi V, Sarkar C, Kulshreshtha R, et al. Hypoxic signature of microRNAs in glioblastoma: Insights from small RNA deep sequencing. BMC Genomics 2014;15:686.  Back to cited text no. 24
    
25.
Hermansen SK, Nielsen BS, Aaberg-Jessen C, Kristensen BW. MiR-21 is linked to glioma angiogenesis: A Co-localization study. J Histochem Cytochem 2016;64:138-48.  Back to cited text no. 25
    
26.
Würdinger T, Tannous BA, Saydam O, Skog J, Grau S, Soutschek J, et al. MiR-296 regulates growth factor receptor overexpression in angiogenic endothelial cells. Cancer Cell 2008;14:382-93.  Back to cited text no. 26
    
27.
Zheng X, Chopp M, Lu Y, Buller B, Jiang F. MiR-15b and miR-152 reduce glioma cell invasion and angiogenesis via NRP-2 and MMP-3. Cancer Lett 2013;329:146-54.  Back to cited text no. 27
    
28.
Cai H, Liu X, Zheng J, Xue Y, Ma J, Li Z, et al. Long non-coding RNA taurine upregulated 1 enhances tumor-induced angiogenesis through inhibiting microRNA-299 in human glioblastoma. Oncogene 2017;36:318-31.  Back to cited text no. 28
    
29.
Engelman JA, Luo J, Cantley LC. The evolution of phosphatidylinositol 3-kinases as regulators of growth and metabolism. Nat Rev Genet 2006;7:606-19.  Back to cited text no. 29
    
30.
Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307:1098-101.  Back to cited text no. 30
    
31.
Liu P, Cheng H, Roberts TM, Zhao JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 2009;8:627-44.  Back to cited text no. 31
    
32.
Wu Y, Dong L, Bao S, Wang M, Yun Y, Zhu R, et al. FK228 augmented temozolomide sensitivity in human glioma cells by blocking PI3K/AKT/mTOR signal pathways. Biomed Pharmacother 2016;84:462-9.  Back to cited text no. 32
    
33.
McCubrey JA, Steelman LS, Chappell WH, Abrams SL, Franklin RA, Montalto G, et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR cascade inhibitors: How mutations can result in therapy resistance and how to overcome resistance. Oncotarget 2012;3:1068-111.  Back to cited text no. 33
    
34.
Logue JS, Morrison DK. Complexity in the signaling network: Insights from the use of targeted inhibitors in cancer therapy. Genes Dev 2012;26:641-50.  Back to cited text no. 34
    
35.
Bai RY, Staedtke V, Riggins GJ. Molecular targeting of glioblastoma: Drug discovery and therapies. Trends Mol Med 2011;17:301-12.  Back to cited text no. 35
    
36.
Puputti M, Tynninen O, Sihto H, Blom T, Mäenpää H, Isola J, et al. Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res 2006;4:927-34.  Back to cited text no. 36
    
37.
Onishi M, Kurozumi K, Ichikawa T, Date I. Mechanisms of tumor development and anti-angiogenic therapy in glioblastoma multiforme. Neurol Med Chir (Tokyo) 2013;53:755-63.  Back to cited text no. 37
    
38.
Westermark B. Platelet-derived growth factor in glioblastoma-driver or biomarker? Ups J Med Sci 2014;119:298-305.  Back to cited text no. 38
    
39.
Vogt PK, Hart JR. PI3K and STAT3: A new alliance. Cancer Discov 2011;1:481-6.  Back to cited text no. 39
    
40.
Mellinghoff IK, Schultz N, Mischel PS, Cloughesy TF. Will kinase inhibitors make it as glioblastoma drugs? Curr Top Microbiol Immunol 2012;355:135-69.  Back to cited text no. 40
    
41.
Nicholas MK, Lukas RV, Jafri NF, Faoro L, Salgia R. Epidermal growth factor receptor-mediated signal transduction in the development and therapy of gliomas. Clin Cancer Res 2006;12:7261-70.  Back to cited text no. 41
    
42.
Paul I, Bhattacharya S, Chatterjee A, Ghosh MK. Current understanding on EGFR and Wnt/β-catenin signaling in glioma and their possible crosstalk. Genes Cancer 2013;4:427-46.  Back to cited text no. 42
    
43.
Ferrari G, Cook BD, Terushkin V, Pintucci G, Mignatti P. Transforming growth factor-beta 1 (TGF-beta1) induces angiogenesis through vascular endothelial growth factor (VEGF)-mediated apoptosis. J Cell Physiol 2009;219:449-58.  Back to cited text no. 43
    
44.
MacDonald BT, He X. Frizzled and LRP5/6 receptors for Wnt/β-catenin signaling. Cold Spring Harb Perspect Biol 2012;4. pii: a007880.  Back to cited text no. 44
    
45.
Xing ZY, Sun LG, Guo WJ. Elevated expression of Notch-1 and EGFR induced apoptosis in glioblastoma multiforme patients. Clin Neurol Neurosurg 2015;131:54-8.  Back to cited text no. 45
    
46.
Guo S, Liu M, Gonzalez-Perez RR. Role of notch and its oncogenic signaling crosstalk in breast cancer. Biochim Biophys Acta 2011;1815:197-213.  Back to cited text no. 46
    
47.
Seshacharyulu P, Ponnusamy MP, Haridas D, Jain M, Ganti AK, Batra SK, et al. Targeting the EGFR signaling pathway in cancer therapy. Expert Opin Ther Targets 2012;16:15-31.  Back to cited text no. 47
    
48.
Chinot OL, de La Motte Rouge T, Moore N, Zeaiter A, Das A, Phillips H, et al. AVAglio: Phase 3 trial of bevacizumab plus temozolomide and radiotherapy in newly diagnosed glioblastoma multiforme. Adv Ther 2011;28:334-40.  Back to cited text no. 48
    
49.
Sooman L, Freyhult E, Jaiswal A, Navani S, Edqvist PH, Pontén F, et al. FGF2 as a potential prognostic biomarker for proneural glioma patients. Acta Oncol 2015;54:385-94.  Back to cited text no. 49
    
50.
Artavanis-Tsakonas S, Rand MD, Lake RJ. Notch signaling: Cell fate control and signal integration in development. Science 1999;284:770-6.  Back to cited text no. 50
    
51.
Iso T, Hamamori Y, Kedes L. Notch signaling in vascular development. Arterioscler Thromb Vasc Biol 2003;23:543-53.  Back to cited text no. 51
    
52.
El Hindy N, Keyvani K, Pagenstecher A, Dammann P, Sandalcioglu IE, Sure U, et al. Implications of dll4-notch signaling activation in primary glioblastoma multiforme. Neuro Oncol 2013;15:1366-78.  Back to cited text no. 52
    
53.
Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y, et al. Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature 2006;444:1083-7.  Back to cited text no. 53
    
54.
Noguera-Troise I, Daly C, Papadopoulos NJ, Coetzee S, Boland P, Gale NW, et al. Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature 2006;444:1032-7.  Back to cited text no. 54
    
55.
Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med 2014;370:699-708.  Back to cited text no. 55
    
56.
Herrlinger U, Schäfer N, Steinbach JP, Weyerbrock A, Hau P, Goldbrunner R, et al. Bevacizumab plus irinotecan versus temozolomide in newly diagnosed O6-methylguanine-DNA methyltransferase nonmethylated glioblastoma: The randomized GLARIUS trial. J Clin Oncol 2016;34:1611-9.  Back to cited text no. 56
    
57.
Zhang JF, Chen Y, Qiu XX, Tang WL, Zhang JD, Huang JH, et al. The vascular delta-like ligand-4 (DLL4)-notch4 signaling correlates with angiogenesis in primary glioblastoma: An immunohistochemical study. Tumour Biol 2016;37:3797-805.  Back to cited text no. 57
    
58.
Qiu XX, Chen L, Wang CH, Lin ZX, Chen BJ, You N, et al. The vascular notch ligands delta-like ligand 4 (DLL4) and jagged1 (JAG1) have opposing correlations with microvascularization but a uniform prognostic effect in primary glioblastoma: A Preliminary study. World Neurosurg 2016;88:447-58.  Back to cited text no. 58
    
59.
Jubb AM, Browning L, Campo L, Turley H, Steers G, Thurston G, et al. Expression of vascular Notch ligands Delta-like 4 and Jagged-1 in glioblastoma. Histopathology 2012;60:740-7.  Back to cited text no. 59
    
60.
Desgrosellier JS, Cheresh DA. Integrins in cancer: Biological implications and therapeutic opportunities. Nat Rev Cancer 2010;10:9-22.  Back to cited text no. 60
    
61.
Carbonell WS, DeLay M, Jahangiri A, Park CC, Aghi MK. B1 integrin targeting potentiates antiangiogenic therapy and inhibits the growth of bevacizumab-resistant glioblastoma. Cancer Res 2013;73:3145-54.  Back to cited text no. 61
    
62.
Kim SH, Wang D, Park YY, Katoh H, Margalit O, Sheffer M, et al. HIG2 promotes colorectal cancer progression via hypoxia-dependent and independent pathways. Cancer Lett 2013;341:159-65.  Back to cited text no. 62
    
63.
Togashi A, Katagiri T, Ashida S, Fujioka T, Maruyama O, Wakumoto Y, et al. Hypoxia-inducible protein 2 (HIG2), a novel diagnostic marker for renal cell carcinoma and potential target for molecular therapy. Cancer Res 2005;65:4817-26.  Back to cited text no. 63
    
64.
Mao XG, Wang C, Liu DY, Zhang X, Wang L, Yan M, et al. Hypoxia upregulates HIG2 expression and contributes to bevacizumab resistance in glioblastoma. Oncotarget 2016;7:47808-20.  Back to cited text no. 64
    
65.
Lewis CA, Brault C, Peck B, Bensaad K, Griffiths B, Mitter R, et al. SREBP maintains lipid biosynthesis and viability of cancer cells under lipid- and oxygen-deprived conditions and defines a gene signature associated with poor survival in glioblastoma multiforme. Oncogene 2015;34:5128-40.  Back to cited text no. 65
    
66.
Mattijssen F, Georgiadi A, Andasarie T, Szalowska E, Zota A, Krones-Herzig A, et al. Hypoxia-inducible lipid droplet-associated (HILPDA) is a novel peroxisome proliferator-activated receptor (PPAR) target involved in hepatic triglyceride secretion. J Biol Chem 2014;289:19279-93.  Back to cited text no. 66
    
67.
Bowman T, Broome MA, Sinibaldi D, Wharton W, Pledger WJ, Sedivy JM, et al. Stat3-mediated Myc expression is required for Src transformation and PDGF-induced mitogenesis. Proc Natl Acad Sci U S A 2001;98:7319-24.  Back to cited text no. 67
    
68.
Abou-Ghazal M, Yang DS, Qiao W, Reina-Ortiz C, Wei J, Kong LY, et al. The incidence, correlation with tumor-infiltrating inflammation, and prognosis of phosphorylated STAT3 expression in human gliomas. Clin Cancer Res 2008;14:8228-35.  Back to cited text no. 68
    
69.
de Groot J, Liang J, Kong LY, Wei J, Piao Y, Fuller G, et al. Modulating antiangiogenic resistance by inhibiting the signal transducer and activator of transcription 3 pathway in glioblastoma. Oncotarget 2012;3:1036-48.  Back to cited text no. 69
    
70.
Bignell GR, Warren W, Seal S, Takahashi M, Rapley E, Barfoot R, et al. Identification of the familial cylindromatosis tumour-suppressor gene. Nat Genet 2000;25:160-5.  Back to cited text no. 70
    
71.
Sun SC. CYLD: A tumor suppressor deubiquitinase regulating NF-kappaB activation and diverse biological processes. Cell Death Differ 2010;17:25-34.  Back to cited text no. 71
    
72.
Song L, Liu L, Wu Z, Li Y, Ying Z, Lin C, et al. TGF-β induces miR-182 to sustain NF-κB activation in glioma subsets. J Clin Invest 2012;122:3563-78.  Back to cited text no. 72
    
73.
Guo J, Shinriki S, Su Y, Nakamura T, Hayashi M, Tsuda Y, et al. Hypoxia suppresses cylindromatosis (CYLD) expression to promote inflammation in glioblastoma: Possible link to acquired resistance to anti-VEGF therapy. Oncotarget 2014;5:6353-64.  Back to cited text no. 73
    
74.
DeLay M, Jahangiri A, Carbonell WS, Hu YL, Tsao S, Tom MW, et al. Microarray analysis verifies two distinct phenotypes of glioblastomas resistant to antiangiogenic therapy. Clin Cancer Res 2012;18:2930-42.  Back to cited text no. 74
    
75.
Lu KV, Bergers G. Mechanisms of evasive resistance to anti-VEGF therapy in glioblastoma. CNS Oncol 2013;2:49-65.  Back to cited text no. 75
    
76.
Di Cristofano A, Pandolfi PP. The multiple roles of PTEN in tumor suppression. Cell 2000;100:387-90.  Back to cited text no. 76
    
77.
Kessler T, Sahm F, Blaes J, Osswald M, Rübmann P, Milford D, et al. Glioma cell VEGFR-2 confers resistance to chemotherapeutic and antiangiogenic treatments in PTEN-deficient glioblastoma. Oncotarget 2015;6:31050-68.  Back to cited text no. 77
    
78.
Plate KH, Breier G, Weich HA, Risau W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 1992;359:845-8.  Back to cited text no. 78
    
79.
Cheng SY, Huang HJ, Nagane M, Ji XD, Wang D, Shih CC, et al. Suppression of glioblastoma angiogenicity and tumorigenicity by inhibition of endogenous expression of vascular endothelial growth factor. Proc Natl Acad Sci U S A 1996;93:8502-7.  Back to cited text no. 79
    
80.
Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe'er J, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: Vasculogenic mimicry. Am J Pathol 1999;155:739-52.  Back to cited text no. 80
    
81.
Mao JM, Liu J, Guo G, Mao XG, Li CX. Glioblastoma vasculogenic mimicry: Signaling pathways progression and potential anti-angiogenesis targets. Biomark Res 2015;3:8.  Back to cited text no. 81
    
82.
El Hallani S, Boisselier B, Peglion F, Rousseau A, Colin C, Idbaih A, et al. A new alternative mechanism in glioblastoma vascularization: Tubular vasculogenic mimicry. Brain 2010;133:973-82.  Back to cited text no. 82
    
83.
Chen Y, Jing Z, Luo C, Zhuang M, Xia J, Chen Z, et al. Vasculogenic mimicry-potential target for glioblastoma therapy: An in vitro and in vivo study. Med Oncol 2012;29:324-31.  Back to cited text no. 83
    
84.
Yao X, Ping Y, Liu Y, Chen K, Yoshimura T, Liu M, et al. Vascular endothelial growth factor receptor 2 (VEGFR-2) plays a key role in vasculogenic mimicry formation, neovascularization and tumor initiation by glioma stem-like cells. PLoS One 2013;8:e57188.  Back to cited text no. 84
    
85.
Huang M, Ke Y, Sun X, Yu L, Yang Z, Zhang Y, et al. Mammalian target of rapamycin signaling is involved in the vasculogenic mimicry of glioma via hypoxia-inducible factor-1α. Oncol Rep 2014;32:1973-80.  Back to cited text no. 85
    
86.
Serwe A, Rudolph K, Anke T, Erkel G. Inhibition of TGF-β signaling, vasculogenic mimicry and proinflammatory gene expression by isoxanthohumol. Invest New Drugs 2012;30:898-915.  Back to cited text no. 86
    
87.
Seftor RE, Seftor EA, Kirschmann DA, Hendrix MJ. Targeting the tumor microenvironment with chemically modified tetracyclines: Inhibition of laminin 5 gamma2 chain promigratory fragments and vasculogenic mimicry. Mol Cancer Ther 2002;1:1173-9.  Back to cited text no. 87
    
88.
Francescone R, Scully S, Bentley B, Yan W, Taylor SL, Oh D, et al. Glioblastoma-derived tumor cells induce vasculogenic mimicry through flk-1 protein activation. J Biol Chem 2012;287:24821-31.  Back to cited text no. 88
    
89.
Fujita T, Igarashi J, Okawa ER, Gotoh T, Manne J, Kolla V, et al. CHD5, a tumor suppressor gene deleted from 1p36.31 in neuroblastomas. J Natl Cancer Inst 2008;100:940-9.  Back to cited text no. 89
    
90.
Mao XG, Xue XY, Wang L, Zhang X, Yan M, Tu YY, et al. CDH5 is specifically activated in glioblastoma stemlike cells and contributes to vasculogenic mimicry induced by hypoxia. Neuro Oncol 2013;15:865-79.  Back to cited text no. 90
    
91.
Escuin D, Kline ER, Giannakakou P. Both microtubule-stabilizing and microtubule-destabilizing drugs inhibit hypoxia-inducible factor-1alpha accumulation and activity by disrupting microtubule function. Cancer Res 2005;65:9021-8.  Back to cited text no. 91
    
92.
Park KJ, Yu MO, Park DH, Park JY, Chung YG, Kang SH, et al. Role of vincristine in the inhibition of angiogenesis in glioblastoma. Neurol Res 2016;38:871-9.  Back to cited text no. 92
    
93.
Musumeci G, Castorina A, Magro G, Cardile V, Castorina S, Ribatti D, et al. Enhanced expression of CD31/platelet endothelial cell adhesion molecule 1 (PECAM1) correlates with hypoxia inducible factor-1 alpha (HIF-1α) in human glioblastoma multiforme. Exp Cell Res 2015;339:407-16.  Back to cited text no. 93
    
94.
Musumeci G, Magro G, Cardile V, Coco M, Marzagalli R, Castrogiovanni P, et al. Characterization of matrix metalloproteinase-2 and -9, ADAM-10 and N-cadherin expression in human glioblastoma multiforme. Cell Tissue Res 2015;362:45-60.  Back to cited text no. 94
    
95.
Hong XY, Wang J, Li Z. AGR2 expression is regulated by HIF-1 and contributes to growth and angiogenesis of glioblastoma. Cell Biochem Biophys 2013;67:1487-95.  Back to cited text no. 95
    
96.
Kang SH, Yu MO, Park KJ, Chi SG, Park DH, Chung YG, et al. Activated STAT3 regulates hypoxia-induced angiogenesis and cell migration in human glioblastoma. Neurosurgery 2010;67:1386-95.  Back to cited text no. 96
    
97.
Kucharzewska P, Christianson HC, Welch JE, Svensson KJ, Fredlund E, Ringnér M, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A 2013;110:7312-7.  Back to cited text no. 97
    
98.
Babae N, Bourajjaj M, Liu Y, Van Beijnum JR, Cerisoli F, Scaria PV, et al. Systemic miRNA-7 delivery inhibits tumor angiogenesis and growth in murine xenograft glioblastoma. Oncotarget 2014;5:6687-700.  Back to cited text no. 98
    
99.
Chen F, Chen L, He H, Huang W, Zhang R, Li P, et al. Up-regulation of microRNA-16 in glioblastoma inhibits the function of endothelial cells and tumor angiogenesis by targeting bmi-1. Anticancer Agents Med Chem 2016;16:609-20.  Back to cited text no. 99
    
100.
Sun J, Zheng G, Gu Z, Guo Z. MiR-137 inhibits proliferation and angiogenesis of human glioblastoma cells by targeting EZH2. J Neurooncol 2015;122:481-9.  Back to cited text no. 100
    


    Figures

  [Figure 1], [Figure 2]
 
 
    Tables

  [Table 1]


This article has been cited by
1 Novel targets to overcome antiangiogenesis therapy resistance in glioblastoma multiforme: Systems biology approach and suggestion of therapy by galunisertib
Morteza Hadizadeh, Akram AminJafari, Sepideh Parvizpour, Sorayya Ghasemi
Cell Biology International. 2022;
[Pubmed] | [DOI]
2 Nutraceuticals Derived from Dietary Spices as Candidate Molecules in Targeting Glioma Signaling Pathways
Vinitha Rani, Pavan S R, Ashwini Prabhu
Revista Brasileira de Farmacognosia. 2022;
[Pubmed] | [DOI]
3 Cytotoxic and apoptotic efficacy of Alkanna tinctoria on glioma cells
Vinitha Rani, Revanasiddappa B. C., Mahendra G. S., Shridhar Deshpande, Jayachandran Venkatesan, Pandurang Appana Dalavi, Ashwini Prabhu
Natural Product Research. 2022; : 1
[Pubmed] | [DOI]
4 Pro- and Antiangiogenic Factors in Gliomas: Implications for Novel Therapeutic Possibilities
Magdalena Groblewska,Barbara Mroczko
International Journal of Molecular Sciences. 2021; 22(11): 6126
[Pubmed] | [DOI]
5 SOX transcription factors and glioma stem cells: Choosing between stemness and differentiation
Milena Stevanovic, Natasa Kovacevic-Grujicic, Marija Mojsin, Milena Milivojevic, Danijela Drakulic
World Journal of Stem Cells. 2021; 13(10): 1417
[Pubmed] | [DOI]
6 Disentangling the therapeutic tactics in GBM: From bench to bedside and beyond
S. Daisy Precilla,Shreyas S. Kuduvalli,Anitha Thirugnanasambandhar Sivasubramania
Cell Biology International. 2020;
[Pubmed] | [DOI]
7 A comprehensive analysis of the angiogenesis-related genes in glioblastoma multiforme vs. brain lower grade glioma
Burcu BITERGE-SUT
Arquivos de Neuro-Psiquiatria. 2020; 78(1): 34
[Pubmed] | [DOI]
8 Role of Matrix Metalloproteinases in Angiogenesis and Cancer
Saray Quintero-Fabián,Rodrigo Arreola,Enrique Becerril-Villanueva,Julio César Torres-Romero,Victor Arana-Argáez,Julio Lara-Riegos,Mario Alberto Ramírez-Camacho,María Elizbeth Alvarez-Sánchez
Frontiers in Oncology. 2019; 9
[Pubmed] | [DOI]
9 Current and Future Trends on Diagnosis and Prognosis of Glioblastoma: From Molecular Biology to Proteomics
Muh-Shi Silantyev,Muh-Shi Falzone,Muh-Shi Libra,Muh-Shi Gurina,Muh-Shi Kardashova,Muh-Shi Nikolouzakis,Muh-Shi Nosyrev,Muh-Shi Sutton,Muh-Shi Panayioti,Muh-Shi Tsatsakis
Cells. 2019; 8(8): 863
[Pubmed] | [DOI]
10 Pericytes in Microvessels: From “Mural” Function to Brain and Retina Regeneration
Nunzia Caporarello,Floriana D’Angeli,Maria Teresa Cambria,Saverio Candido,Cesarina Giallongo,Mario Salmeri,Cinzia Lombardo,Anna Longo,Giovanni Giurdanella,Carmelina Daniela Anfuso,Gabriella Lupo
International Journal of Molecular Sciences. 2019; 20(24): 6351
[Pubmed] | [DOI]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Genes Potentiall...
Micro-Rnas Poten...
Receptors and Si...
Potential Mechan...
New Regulated Fa...
Conclusions and ...
References
Article Figures
Article Tables

 Article Access Statistics
    Viewed6915    
    Printed430    
    Emailed0    
    PDF Downloaded737    
    Comments [Add]    
    Cited by others 10    

Recommend this journal