Abstract
Dynamic susceptibility contrast-enhanced magnetic resonance imaging is routinely used to provide hemodynamic assessment of brain tumors as a diagnostic as well as a prognostic tool. Recently, it was shown that the relative cerebral blood volume (rCBV), obtained from the contrast-enhancing as well as -nonenhancing portion of glioblastoma (GBM), is strongly associated with overall survival. In this study, we aim to characterize the genomic correlates (microRNA, messenger RNA, and protein) of this vascular parameter. This study aims to provide a comprehensive radiogenomic and radioproteomic characterization of the hemodynamic phenotype of GBM using publicly available imaging and genomic data from the Cancer Genome Atlas GBM cohort. Based on this analysis, we identified pathways associated with angiogenesis and tumor proliferation underlying this hemodynamic parameter in GBM.
Keywords: imaging-genomics, perfusion imaging, rCBV, angiogenesis, pathway analysis, data integration
Introduction
Glioblastoma (GBM) is a WHO grade IV tumor with an extremely poor prognosis and a median survival of 12–15 months. Many imaging features are used to characterize GBM, such as structural magnetic resonance imaging (MRI), diffusion MRI, and perfusion MRI.1 Several of these derived radiology- based image features (denoted radiophenotypes) have shown association with the underlying genetics and biology of the tumor.2
Identification of such noninvasive surrogates of clinical outcome and the associated molecular differences might point to a deeper understanding of the underlying disease state.3,4 Several studies5–7 have demonstrated that imaging phenotypes derived from MRI studies might have the potential to serve as noninvasive proxies of cancer-associated clinical outcomes. In this regard, we follow up on recent studies based on dynamic susceptibility contrast-enhanced MRI (DSC-MRI) that identifies a hemodynamic parameter associated with overall survival. Specifically, these studies5,8 pointed to the association of rCBVmax (maximum relative cerebral blood volume of the contrast-enhancing component of GBM) and rCBVNER (rCBV in the nonenhancing region of the tumor) with patient survival.
The Cancer Genome Atlas (TCGA)9 contains comprehensive genomic data (spanning expression, copy number, methylation, microRNA (miRNA), etc.). The Cancer Imaging Archive (TCIA)10 is a publicly available repository of patient-derived images across multiple modalities (single-photon emission computed tomography, computed tomography, and MRI) and multiple tumor types in many of the patients who have corresponding molecular data in TCGA. Using genomic and imaging data from the TCGA-GBM archive, we carried out a bioinformatics analysis to identify molecular correlates of imaging features associated with survival.
Based on the prognostic significance of the hemody-namic parameter (rCBVmax) value, we sought to examine the molecular correlates of rCBVmax via differential expression analysis of messenger RNA (mRNA), miRNA, and protein levels. The intent was to identify molecular entities that are likely regulated across different layers of biological complexity: each modality (mRNA, miRNA, and protein) provides complementary information about the biology of the process of GBM tumorigenesis. Thus, we decided to combine the significantly associated genes (transcriptional correlate), miR-NAs (post-transcriptional correlate), and proteins (signal transduction correlate) to identify if there was a concordant biological process (or processes) that could be inferred when these three–omic modalities were combined. Combining entities found associated with tumor hemodynamic response across different biological layers has the potential to illuminate important regulatory processes.
For this purpose, patients were dichotomized based on median rCBVmax value and then examined for differential expression of mRNA, miRNA, and protein levels. A molecular examination of the two phenotypic groups reveals that genes and miRNAs related to the molecular process of angio-genesis underlie this phenotypic transition between low and high rCBVmax groups.
Materials and Methods
Ethics statement
All patients in this retrospective study had been previously de-identified by TCGA, a publicly available data set that contains no linkage to patient identifiers and is compliant with the Health Insurance Portability and Accountability Act; therefore, a waiver was obtained from the Henry Ford Hospital Institutional Review Board of Henry Ford Hospital, Detroit, MI. This research complied with the principles of the Declaration of Helsinki.
Patient population
Based on the availability of perfusion (DSC) MRI image data from the TCIA and the corresponding genomic expression from TCGA, 50 GBM cases were identified (19 females, 30 males, and 1 unknown). These cases were selected to ensure consistency in glioma history and treatment profile. Specifically, patients with untreated primary GBM were selected. Clinical data for this population were obtained from the cBioPortal for Cancer Genomics.11
Image features
Radiology annotations of the 50 GBM cases and their preoperative images (from TCIA) were obtained from the TCGA Glioma Phenotype Research Group.10 This Health Insurance Portability and Accountability Act-compliant retrospective study was approved by the Henry Ford Hospital Institutional Review Board (IRB #6381). Patients underwent DSC T2*-weighted MR perfusion studies and had gene expression data available from TCGA. Fifteen cases were imaged in a 3 T scanner, while others were imaged in a 1.5 T scanner. rCBVmax was measured from a region of interest of 10 × 10 voxels5 placed on the hottest-appearing tumor region based on qualitative perfusion maps.
Genomic features
Batch-corrected, Level 3 expression data for both mRNA (Affymetrix U133Av2 BI Platform: Affymetrix HT HG U133A) and miRNA (UNC Agilent Human miRNA 8×15K) were downloaded from the UT MD Anderson Genome Data Analysis Center Portal12 version dated June 18, 2013. Protein expression from reverse-phase protein array (RPPA) was downloaded from the Cancer Pro-teome Portal.13 Forty-seven cases had miRNA and mRNA data available, whereas 15 cases had data for all three platforms. Instead of restricting our study to just the 15 common cases for differential expression analysis, we performed analysis of each platform separately and integrated the differential expression results subsequently via pathway analysis.
Defining classes based on median rCBVmax
Based on the prior finding that rCBVmax is associated with survival in a statistical significant manner,5 we sought to examine the molecular correlates of rCBVmax via differential expression analysis. Median rCBVmax across the entire dataset was used to divide the study population into two groups.
Differential expression analysis between cutpoint-induced phenotype classes
Differential expression analysis for mRNA, miRNA, and protein was done to investigate molecular differences between the median rCBVmax-induced phenotype groups. This was done using a two-sided t-test to identify genes, miRNAs, and proteins that are differentially expressed between the two classes. Because of the exploratory nature of this study, no correction was performed for multiple hypothesis testing.
Functional analysis and integrative analysis
For relating differentially expressed miRNAs with differentially expressed mRNAs, the miRNA target filter feature (experimentally validated as well as high confidence sequence matches) is used within Ingenuity Pathway Analysis (IPA) software to find miRNAs that target the differentially expressed genes. Such integrative miRNA:mRNA analysis looks for target relationship (based on sequence affinity, using databases like TargetScan) between miRNAs and the mRNAs derived from differential expression analysis. In addition, the miRNA target filter in IPA looks for concordant changes in expression, ie, anticorrelated expression changes in miRNA:mRNA abundance.
Integrative network analysis of miRNA, mRNA and protein 141 entities via Ingenuity Pathway network analysis
Core analysis and functional analysis were performed on the differentially expressed gene, protein, and miRNA lists. We explored both “Direct and Indirect Interactions” using “Experimentally Observed OR High Confidence Predictions” in the IPA knowledgebase.
Results
Differential expression programs between phenotypic classes reveal the role for cell proliferation and angiogenesis-associated pathways
The differential expression analysis procedure on the mRNA, protein, and miRNA data yields 326 genes, 76 miRNAs, and 8 proteins that are differentially expressed between these two phenotype classes. For completeness, the ontological analysis of these genes, miRNAs, and proteins (using the IPA tool) is presented (Table 1). The key pathways activated in this set are related to cellular development, cellular proliferation, cell death, interleukin signaling, and inflammatory response.14,15
Table 1.
TOP CANONICAL PATHWAYS | ||
---|---|---|
NAME | p-value | RATIO |
LPS/IL-1 mediated inhibition of RXR function | 3.74E-04 | 13/245 (0.053) |
1L-17A signaling in gastric cells | 1.14E-03 | 4/28 (0.143) |
Tight junction signaling | 2.5E-03 | 9/167 (0.054) |
p53 signaling | 2.5E-03 | 7/113 (0.062) |
Pathogenesis of multiple sclerosis | 1.17E-03 | 2/10 (0.2) |
DISEASES AND DISORDERS | ||
NAME | p-value | # MOLECULES |
Cancer | 1.79E-13–7.55E-03 | 311 |
Organismal injury and abnormalities | 1.79E-13–7.55E-03 | 200 |
Reproductive system disease | 1.79E-13–7.554E-03 | 163 |
Gastrointestinal disease | 3.52E-11–6.97E-03 | 159 |
Dermatological diseases and conditions | 3.59E-11–6.97E-03 | 41 |
PHYSIOLOGICAL SYSTEM DEVELOPMENT AND FUNCTION | ||
NAME | p-value | # MOLECULES |
Reproductive system development and function | 1.80E-06–6.97E-03 | 25 |
Tissue morphology | 1.80E-06–7.38E-03 | 80 |
Organismal development | 6.06E-05–7.45E-03 | 109 |
Organismal development | 2.09E-05–7.4E-03 | 65 |
Tumor morphology | 2.21E-05–6.97E-03 | 48 |
MOLECULAR AND CELLULAR FUNCTIONS | ||
NAME | p-value | # MOLECULES |
Cellular development | 1.70E-06–7.23E-03 | 124 |
Cellular growth and proliferation | 1.80E-06–7.38E-03 | 130 |
Cell death and survival | 6.06E-06–7.45E-03 | 130 |
Cell cycle | 2.09E-05–7.45E-03 | 40 |
Cell morphology | 2.21E-04–6.97E-03 | 77 |
Integrated miRNA:mRNA analysis reveals multiple molecules underlying the transition of this perfusion-associated phenotype
We reasoned that combining the differentially expressed miRNAs and mRNAs might reveal a robust mechanism underlying the transition between the two phenotypic classes. Using the IPA tool, we combined the miRNA expression results with the mRNA expression results and found that eight differentially expressed miRNAs (miR-29b-3p, miR495–3p, miR30c/30d, miR-26a-5p, miR296–5p, miR128–3p, miR144–3p, and miR214–3p) target several differentially expressed genes (including PTEN, COL15A1, SPARC, ANPEP, CBFB, STRN, and TMED10) while exhibiting concordant expression correlation (the expression levels of the mRNAs are anticorrelated with the miRNA expression levels).
Several of these miRNAs have been shown to play a key role in GBM. miR-26a regulates PTEN and is observed to be amplified in high-grade gliomas in addition to promoting tumorigenesis.16 miR-29b has been shown to regulate invasion in GBM.17
Examination of the combined set of mRNAs, proteins, and miRNAs via IPA reveals several networks of potential interest. The primary biological processes (based on Fisher’s exact tests for over-representation analysis) are LPS/IL-1- mediated signaling, IL-17A signaling, tight junction signaling, p53 signaling, pathogenesis of multiple sclerosis, etc. (presumably related to angiogenic activity [Table 1]).
Discussion
Perfusion MRI is a promising tool that is now being routinely used to interrogate tumor behavior, such as inflammation and vascularization. It is already being used for treatment monitoring (eg, assessment of changes due to treatment with antiangiogenic therapy). Since it is a much newer tool than conventional MRI, a study of the specific biological processes associated with perfusion imaging is useful to inform the radiologist about what specifically is being assessed using this modality.
Tumor blood volume (rCBVmax) is a measure of total tumor vasculature and angiogenesis. It has been shown to correlate with microvascular density18 as well as genes related to angiogenesis regulation19 and hence is a useful prognostic marker.5,8 Our analysis reveals the role of several angiogenesis-related molecules (at the miRNA, mRNA, and protein levels) associated with this vascular parameter. This functional analysis that spans miRNA, mRNA, and protein entities produces several insights that could perhaps clarify the etiology of GBMs as measured via perfusion imaging in addition to possibly generating new hypotheses that could be validated in a laboratory. Specifically, animal models of GBM with overexpressed gene/miRNA levels might inform on the relationships of these molecules with tumor-associated angiogenic activity.
An examination of the IPA-derived pathways reveals roles for several pathways involved in tumorigenesis, tumor growth, and angiogenesis – specifically, IL-17 signaling is key to tumor survival, inflammation, and angiogenic activity.20,21 Further IL-1 signaling has a role in tumor-mediated angio-genesis.22,23 An examination of the molecular and cellular activities in IPA reveals a role for several tumor hallmarks, such as modulation of tumor morphology, cell proliferation, cell cycle, and apoptosis. Another interesting point is that the context of angiogenesis ties in the observations from our gene set analysis with the pathogenesis of multiple sclerosis, again suggesting a role for angiogenesis modulation.24,25
Angiogenesis is one of the key hallmarks of cancer.26 HER2 signaling and angiogenesis have been shown to be closely linked.27,28 The role of EGFR in GBM is well documented.29 Moreover, its role in modulation of invasion, angiogenesis, and metastasis has been reported; specifically, EGFR signaling contributes to angiogenesis via upregulation of MMP levels and cytokines such as VEGFA and IL8.30 BRCA2 is a tumor suppressor,31 so it is expected that its level goes down in the more extreme rCBV phenotype (suggesting a more aggressive disease). However, its exact relationship with angiogenesis is not clearly characterized, especially in the context of glioma. Several of these proteins have been shown to play a key role in GBM. For example, JNK2 activation correlates strongly with EGFR expression as well as histological grade in GBM.32 JNK2 has been shown to control cell proliferation and apoptosis and thus been implicated in tumorigenesis.33,34 Furthermore, JNK2 has been shown to an upstream regulator of NFAT activity, which is a critical regulator of angiogenesis.35,36
Among the miRNAs listed in Table 2, miR495 has been shown to inhibit proliferation of GBM cells by downregulating CDK6, and thus is heavily downregulated in higher grade gliomas.37 miRNA-26a regulates angiogenesis via modulation of the BMP/SMAD1 signaling.38,39 miR-29b suppresses angiogenesis, migration, and invasion by regulating MMP2 levels; thus, it is expected that it would be down-regulated in the high rCBVmax group, suggesting an activation of these tumorigenic hallmarks. miR-30d has been shown to modulate tumor cell proliferation, angiogenesis, and metastasis.40,41 Their specific role in glioma needs to be investigated more thoroughly.
Table 2.
ENTITY | miRNAa | mRNAb | PROTEIN |
---|---|---|---|
1 | miR26a | PTEN | JNK2 |
2 | miR29b | COL15A1 | HER2 |
3 | miR29b | SPARC | ACC1 |
4 | miR30d | ANPEP | EGFR pY1068, EGFR pY1173 |
5 | miR30d | CBFB | P90RSK |
6 | miR30d | STRN | BRCA2 |
7 | miR30d | TMED10 | Dvl3 |
Notes:
Only the ones that have an experimentally proven relationship of sequence affinity are shown.
Only the ones that have an experimentally proven relationship of sequence affinity are shown.
Conclusions and Future Work
This study reveals the utility of integrating genomic data to dissect the molecular programs underlying the vascular phenotype of the tumor, which is assessed with perfusion imaging. Our hypothesis is that instead of focusing solely on one –omic modality, combining –omic modalities is more likely to illuminate the molecular mechanisms underlying the disease processes. Thus, an integration of mRNA, miRNA, and RPPA data permits the combination of multiple layers of biological complexity: transcriptional, posttranscriptional, and signaling, to infer biological mechanisms underlying this hemodynamic characteristic of GBM.
There are several limitations to address in our study. First, this was a retrospective study performed on a publicly available patient subset, consisting of data acquired on multiple MRI systems with varying protocols. A study that evaluates the robustness of these integrative molecular programs across scanning protocols, scanner resolutions, and a larger sample size is essential to establishing their biological role. A separate dataset with uniformity of treatment regimen is the next step in validating the biological role of these genomic programs. Furthermore, incorporating molecular markers such as IDH mutation status and molecular subtype in a multivariate regression can help to deconvolve the underlying molecular programs underlying the perfusion phenotype in GBM. Also, the top ranking pathways and molecules identified via integrative analysis need to be validated through laboratory studies, possibly via in vivo mouse models of GBM. Finally, an experimental analysis of drugs targeting these signaling molecules and implicated pathways might permit the examination of novel or rational therapeutic combinations for GBM. Methodologically, this work infers differentially expressed entities from median-dichotomized patient groups from rCBVmax. However, examining associated genes/miR-NAs/proteins using the original, continuous-valued rCBVmax values (via measures such as Spearman and Kendall correlation) is another possible avenue for examination. Future work will also involve the examination of computational features derived from perfusion data42–44 and the assessment of molecular alterations (mRNA, miRNA, and protein) underlying those characteristics. This will permit the molecular examination of hemodynamic characteristics other than rCBV, thus illuminating other aspects beyond those routinely obtained in current clinical practice.
Acknowledgments
The authors would like to thank Dr. Laila Poisson for helpful discussions on this manuscript and Tomiko Fisher for assistance with manuscript editing.
Footnotes
ACADEMIC EDITOR: J. T. Efird, Editor in Chief
PEER REVIEW: Four peer reviewers contributed to the peer review report. Reviewers’ reports totaled 759 words, excluding any confidential comments to the academic editor.
FUNDING: The work was supported by CCSG Bioinformatics Shared Resource NCI P30 CA016672, Institutional Research Grant and MDACC Brain Tumor SPORE Career Development Award (5P50CA127001-07), to AR. The authors confirm that the funder had no influence over the study design, content of the article, or selection of this journal.
COMPETING INTERESTS: Authors disclose no potential conflicts of interest.
Paper subject to independent expert blind peer review. All editorial decisions made by independent academic editor. Upon submission manuscript was subject to anti-plagiarism scanning. Prior to publication all authors have given signed confirmation of agreement to article publication and compliance with all applicable ethical and legal requirements, including the accuracy of author and contributor information, disclosure of competing interests and funding sources, compliance with ethical requirements relating to human and animal study participants, and compliance with any copyright requirements of third parties. This journal is a member of the Committee on Publication Ethics (COPE).
Author Contributions
Conception and design of research: AR, RJ. Data preparation, analysis and reporting: GM, GR, RJ, AR. All authors reviewed and approved of the final manuscript.
REFERENCES
- 1.Pope WB, Young JR, Ellingson BM. Advances in MRI assessment of gliomas and response to anti-VEGF therapy. Curr Neurol Neurosci Rep. 2011;11(3):336–44. doi: 10.1007/s11910-011-0179-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Gutman DA, Cooper LA, Hwang SN, et al. MR imaging predictors of molecular profile and survival: multi-institutional study of the TCGA glioblastoma data set. Radiology. 2013;267(2):560–9. doi: 10.1148/radiol.13120118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Diehn M, Nardini C, Wang DS, et al. Identification of noninvasive imaging surrogates for brain tumor gene-expression modules. Proc Natl Acad Sci U S A. 2008;105(13):5213–8. doi: 10.1073/pnas.0801279105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Segal E, Sirlin CB, Ooi C, et al. Decoding global gene expression programs in liver cancer by noninvasive imaging. Nat Biotechnol. 2007;25(6):675–80. doi: 10.1038/nbt1306. [DOI] [PubMed] [Google Scholar]
- 5.Jain R, Poisson L, Narang J, et al. Genomic mapping and survival prediction in glioblastoma: molecular subclassification strengthened by hemodynamic imaging biomarkers. Radiology. 2013;267(1):212–20. doi: 10.1148/radiol.12120846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zinn PO, Mahajan B, Sathyan P, et al. Radiogenomic mapping of edema/cellular invasion MRI-phenotypes in glioblastoma multiforme. PLoS One. 2011;6(10):e25451. doi: 10.1371/journal.pone.0025451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Pope WB, Mirsadraei L, Lai A, et al. Differential gene expression in glioblastoma defined by ADC histogram analysis: relationship to extracellular matrix molecules and survival. AJNR Am J Neuroradiol. 2012;33(6):1059–64. doi: 10.3174/ajnr.A2917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Jain R, Poisson LM, Gutman D, et al. Outcome prediction in patients with glioblastoma by using imaging, clinical, and genomic biomarkers: focus on the nonenhancing component of the tumor. Radiology. 2014;272(2):484–93. doi: 10.1148/radiol.14131691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Haralick RM, Shanmugam K, Dinstein IH. Textural features for image classification. IEEE Trans Syst Man Cybern B Cybern. 1973;3(6):610–21. [Google Scholar]
- 10.Gatrell AC, Bailey TC, Diggle PJ, Rowlingson BS. Spatial point pattern analysis and its application in geographical epidemiology. Trans Inst Br Geogr. 1996;21(1):256–74. [Google Scholar]
- 11.Tesar L, Shimizu A, Smutek D, Kobatake H, Nawano S. Medical image analysis of 3D CT images based on extension of Haralick texture features. Comput Med Imag Grap. 2008;32(6):513–20. doi: 10.1016/j.compmedimag.2008.05.005. [DOI] [PubMed] [Google Scholar]
- 12.Ng F, Ganeshan B, Kozarski R, Miles KA, Goh V. Assessment of primary colorectal cancer heterogeneity by using whole-tumor texture analysis: contrast-enhanced CT texture as a biomarker of 5-year survival. Radiology. 2013;266(1):177–84. doi: 10.1148/radiol.12120254. [DOI] [PubMed] [Google Scholar]
- 13.Li J, Lu Y, Akbani R, et al. The Cancer Proteome Atlas 2014Available at: http://app1.bioinformatics.mdanderson.org/tcpa/_design/basic/index.html.
- 14.Angelo LS, Kurzrock R. Vascular endothelial growth factor and its relationship to inflammatory mediators. Clin Cancer Res. 2007;13(10):2825–30. doi: 10.1158/1078-0432.CCR-06-2416. [DOI] [PubMed] [Google Scholar]
- 15.Imhof BA, Aurrand-Lions M. Angiogenesis and inflammation face off. Nat Med. 2006;12(2):171–2. doi: 10.1038/nm0206-171. [DOI] [PubMed] [Google Scholar]
- 16.Huse JT, Brennan C, Hambardzumyan D, et al. The PTEN-regulating microRNA miR-26a is amplified in high-grade glioma and facilitates gliom-agenesis in vivo. Genes Dev. 2009;23(11):1327–37. doi: 10.1101/gad.1777409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cortez MA, Nicoloso MS, Shimizu M, et al. miR-29b and miR-125a regulate podoplanin and suppress invasion in glioblastoma. Genes Chromosomes Cancer. 2010;49(11):981–90. doi: 10.1002/gcc.20808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jain R, Gutierrez J, Narang J, et al. In vivo correlation of tumor blood volume and permeability with histologic and molecular angiogenic markers in gliomas. AJNR Am J Neuroradiol. 2011;32(2):388–94. doi: 10.3174/ajnr.A2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jain R, Poisson L, Narang J, et al. Correlation of perfusion parameters with genes related to angiogenesis regulation in glioblastoma: a feasibility study. AJNR Am J Neuroradiol. 2012;33(7):1343–8. doi: 10.3174/ajnr.A2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Hayata K, Iwahashi M, Ojima T, et al. Inhibition of IL-17A in tumor micro-environment augments cytotoxicity of tumor-infiltrating lymphocytes in tumor-bearing mice. PLoS One. 2013;8(1):e53131. doi: 10.1371/journal.pone.0053131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Numasaki M, Fukushi J, Ono M, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. 2003;101(7):2620–7. doi: 10.1182/blood-2002-05-1461. [DOI] [PubMed] [Google Scholar]
- 22.Carmi Y, Voronov E, Dotan S, et al. The role of macrophage-derived IL-1 in induction and maintenance of angiogenesis. J Immunol. 2009;183(7):4705–14. doi: 10.4049/jimmunol.0901511. [DOI] [PubMed] [Google Scholar]
- 23.Voronov E, Carmi Y, Apte RN. The role IL-1 in tumor-mediated angiogenesis. Front Physiol. 2014;5:114. doi: 10.3389/fphys.2014.00114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kirk S, Frank JA, Karlik S. Angiogenesis in multiple sclerosis: is it good, bad or an epiphenomenon? J Neurol Sci. 2004;217(2):125–30. doi: 10.1016/j.jns.2003.10.016. [DOI] [PubMed] [Google Scholar]
- 25.Seabrook TJ, Littlewood-Evans A, Brinkmann V, Pöllinger B, Schnell C, Hiestand PC. Angiogenesis is present in experimental autoimmune encepha-lomyelitis and proangiogenic factors are increased in multiple sclerosis lesions. J Neuroinflammation. 2010;7:95. doi: 10.1186/1742-2094-7-95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 27.Alameddine RS, Otrock ZK, Awada A, Shamseddine A. Crosstalk between HER2 signaling and angiogenesis in breast cancer: molecular basis, clinical applications and challenges. Curr Opin Oncol. 2013;25(3):313–24. doi: 10.1097/CCO.0b013e32835ff362. [DOI] [PubMed] [Google Scholar]
- 28.Kumar R, Yarmand-Bagheri R. The role of HER2 in angiogenesis. Semin Oncol. 2001;28(5 suppl 16):27–32. doi: 10.1016/s0093-7754(01)90279-9. [DOI] [PubMed] [Google Scholar]
- 29.Hatanpaa KJ, Burma S, Zhao D, Habib AA. Epidermal growth factor receptor in glioma: signal transduction, neuropathology, imaging, and radioresistance. Neoplasia. 2010;12(9):675–84. doi: 10.1593/neo.10688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Box C, Peak J, Rogers S, Eccles S. EGFR signaling in invasion, angiogenesis and metastasis. In: Haley JD, Gullick WJ, editors. EGFR Signaling Networks in Cancer Therapy Cancer Drug Discovery and Development. New York, NY: Humana Press; 2008. pp. 245–64. [Google Scholar]
- 31.Shahid T, Soroka J, Kong EH, et al. Structure and mechanism of action of the BRCA2 breast cancer tumor suppressor. Nat Struct Mol Biol. 2014;21(11):962–8. doi: 10.1038/nsmb.2899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Yeung YT, McDonald KL, Grewal T, Munoz L. Interleukins in glioblastoma pathophysiology: implications for therapy. Br J Pharmacol. 2013;168(3):591–606. doi: 10.1111/bph.12008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ennis BW, Fultz KE, Smith KA, et al. Inhibition of tumor growth, angiogene-sis, and tumor cell proliferation by a small molecule inhibitor of c-Jun N-terminal kinase. J Pharmacol Exp Ther. 2005;313(1):325–32. doi: 10.1124/jpet.104.078873. [DOI] [PubMed] [Google Scholar]
- 34.Sabapathy K, Wagner EF. JNK2: a negative regulator of cellular proliferation. Cell Cycle. 2004;3(12):1520–3. doi: 10.4161/cc.3.12.1315. [DOI] [PubMed] [Google Scholar]
- 35.Yao K, Cho YY, Bergen HR, III, et al. Nuclear factor of activated T3 is a negative regulator of Ras-JNK1/2-AP-1 induced cell transformation. Cancer Res. 2007;67(18):8725–35. doi: 10.1158/0008-5472.CAN-06-4788. [DOI] [PubMed] [Google Scholar]
- 36.Zaichuk TA, Shroff EH, Emmanuel R, Filleur S, Nelius T, Volpert OV. Nuclear factor of activated T cells balances angiogenesis activation and inhibition. J Exp Med. 2004;199(11):1513–22. doi: 10.1084/jem.20040474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Adlakha YK, Saini N. Brain microRNAs and insights into biological functions and therapeutic potential of brain enriched miRNA−128. Mol Cancer. 2014;13:33. doi: 10.1186/1476-4598-13-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chai ZT, Kong J, Zhu XD, et al. MicroRNA-26a inhibits angiogenesis by downregulating VEGFA through the PIK3C2alpha/Akt/HIF-1alpha pathway in hepatocellular carcinoma. PLoS One. 2013;8(10):e77957. doi: 10.1371/journal.pone.0077957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Icli B, Wara AK, Moslehi J, et al. MicroRNA-26a regulates pathological and physiological angiogenesis by targeting BMP/SMAD1 signaling. Circ Res. 2013;113(11):1231–41. doi: 10.1161/CIRCRESAHA.113.301780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Abba M, Patil N, Allgayer H. MicroRNAs in the regulation of MMPs and metastasis. Cancers. 2014;6(2):625–45. doi: 10.3390/cancers6020625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Su SF, Chang YW, Andreu-Vieyra C, et al. miR-30d, miR-181a and miR-199a-5p cooperatively suppress the endoplasmic reticulum chaperone and sig-naling regulator GRP78 in cancer. Oncogene. 2013;32(39):4694–701. doi: 10.1038/onc.2012.483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zacharaki EI, Morita N, Bhatt P, O’Rourke DM, Melhem ER, Davatzikos C. Survival analysis of patients with high-grade gliomas based on data mining of imaging variables. AJNR Am J Neuroradiol. 2012;33(6):1065–71. doi: 10.3174/ajnr.A2939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ryoo I, Choi SH, Kim JH, et al. Cerebral blood volume calculated by dynamic susceptibility contrast-enhanced perfusion MR imaging: preliminary correlation study with glioblastoma genetic profiles. PLoS One. 2013;8(8):e71704. doi: 10.1371/journal.pone.0071704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Lee J, Jain R, Khalil K, et al. Texture feature ratios from relative CBV maps of perfusion MRI are associated with patient survival in glioblastoma. AJNR Am J Neuroradiol. 2015;37(1):37–43. doi: 10.3174/ajnr.A4534. [DOI] [PMC free article] [PubMed] [Google Scholar]