Gene expression profile for predicting survival of patients with meningioma

  • Authors:
    • Feng Chen
    • Chun-Xiang Xiang
    • Yi Zhou
    • Xiang-Sheng Ao
    • Da-Quan Zhou
    • Peng Peng
    • Hai-Quan Zhang
    • Han-Dong Liu
    • Xing Huang
  • View Affiliations

  • Published online on: November 25, 2014     https://doi.org/10.3892/ijo.2014.2779
  • Pages: 791-797
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Current staging methods are inadequate for predicting the overall survival of meningioma. DNA microarray technologies improve the understanding of tumour progression. We analysed genome wide expression profiles of 119 meningioma samples from two previous published DNA microarray studies. The Cox proportional hazards regression models were applied to identify overall survival related gene signature. A total of 449 genes (109 upregulated and 340 downregulated) were identified as differentially expressed in meningioma. Among these differentially expressed genes, 37 genes were identified to be related to meningioma overall survival. Our 37-gene signature is closely associated with overall survival among patients with meningioma. This gene expression profile could provide an optimization of the clinical management and development of new therapeutic strategies for meningioma.

Introduction

Meningioma usually appears in the senior age of life and accounts to ~22% of intracranial tumours in males and 38% in females (1). According to the WHO grading criteria, meningioma is divided into three grades: benign meningioma (grade I), atypical meningioma (grade II) and anaplastic meningioma (grade III). Anaplastic meningioma shows poorer prognosis than other common types of meningioma with a median overall survival time 2 years. Due to the difficulty of managing meningioma recurrence (2) and the low predictive powers of clinic pathological factors, biomarkers to identify high-risk patients with a poor prognosis are strongly needed. From a previous study conducted in a total 124 samples from 105 patients using the high-resolution FISH-technology (iFISH), it is reported that the deletion of chromosome 1p may be an independent marker of meningioma recurrence and progression (3).

DNA microarray technology has enabled the simultaneous measurements of expression levels of thousands of genes in a group sample of tumour tissue, which could be used to provide prognostic information, or discriminate between various histologic subtypes. For the last decade, many research groups have used this technology to define prognostic gene signatures in many different tumour types, such as ovarian (4) breast (5), gastric (6), lung cancer (7) and neuroblastomas (8). The survival related gene signature might capture tumour progression status and could serve as a prerequisite to a more patient-tailored therapy.

Nevertheless, to the best of our knowledge, no study in which gene expression profiling was used to predict meningioma overall survival has been published yet. In the present study, we analysed genome-wide expression profiles of 119 meningioma samples using DNA microarray technologies. We applied Cox proportional hazards regression models and identified 37 prognostic genes related to meningioma overall survival, which could provide an optimization of the clinical management and development of new therapeutic strategies for meningioma.

Materials and methods

Data collection

Gene expression profiling datasets from two previously published studies analysed in the present study were referred as dataset 1 and dataset 2. Dataset 1 consists of 51 samples, while dataset 2 consists of 68 samples.

For dataset 1 (9), there are 47 meningioma samples and 4 normal meninges. As previously discussed, the selection criteria of these samples include availability of enough tumour RNA, high quality of the extracted RNA and the representativeness of its cytogenetic profiles as defined by interphase fluorescence in situ hybridization (iFISH) within the whole series of tumours. Total RNA was extracted using RNeasy Mini kit (Quiagen, Valencia, CA, USA). The RNA integrity was assessed using the Agilent 2100 Bioanalyzer (Agilent Technologies, Inc., Palo Alto, CA, USA). The labelling process was performed according to the protocols from Affymetrix. Labelled RNA was hybridized to Human Genome U133A microarray, after quality checking on GeneChips Test 3 Arrays. Washing and scanning were performed using Fluidics Station 400 and GeneChip Scanner (Affymetrix).

For dataset 2 (10), tumour biopsies from 43 female and 25 male subjects with sporadic meningioma were selected from the UCLA Neuro-oncology Program Tissue Bank through institutional review board approved protocols. RNA was extracted from 20–50 mg tumour pieces using Qiagen™ (Qiagen) RNeasy Mini kits per manufacturer’s protocols. The extracted total RNA was assessed for integrity using the 2100 Bioanalyzer by Agilent Technologies. Total RNA (1 μg) was used for single-round biotinylated probe synthesis using the Affymetrix Array Station device made by Caliper Life Sciences (Hopkinton, MA, USA) by the manufacturer’s protocols. Labelled and sheared cRNA was manually applied to Affymetrix Human Genome U133 Plus 2.0 Arrays (Affymetrix). All microarrays were scanned using the Affymetrix GeneChip scanner 3000.

Data processing

We retrieved the raw fluorescence intensity data within CEL files from NCBI GEO database with accession number GSE43290 (dataset 1) and GSE16581 (dataset 2). The raw datasets were preprocessed with gcrma algorithm, as implemented with R packages from Bioconductor (http://www.bioconductor.org). The gcrma algorithm (11) adjusts for the background intensities in Affymtrix array data by including optical noise and non-specific binding (NSB). It then converts background adjusted probe intensities to expression measures using the same normalization and summarization methods implanted by the robust multiarray average (RMA) (12) algorithm. Because two different microarray platforms were used in these datasets, the probe sets had to be matched to identical genes. Based on the latest official symbol annotation provided by the manufacturer, we developed a Perl script to match probe sets among datasets 1 and 2. This process produced a total of 13,879 genes on both the two Affymetrix microarray systems: Human Genome U133A microarray and Human Genome U133 Plus 2.0 Arrays.

Microarray and statistical analysis

We then used the statistical software programs R, the R-package, limma (13), for the analysis of gene expression microarray data from dataset 1. This algorithm uses linear models, as well as Empirical Bayesian methods, for analysing designed experiments and assessment of differential expression. The Cox proportional hazards regression analysis was conducted to identify survival related genes. Survival analysis was performed using the R-package ‘survival’ (14). Overall survival (OS) was analysed using the Kaplan-Meier product-limit method and the significance of the variables was measured by the log-rank test. Hierarchical clustering based on Euclidean distance and Ward’s clustering method were used to show the expression patterns of survival-related genes in dataset 2. We further analysed the Gene Ontology and canonical pathways with the use of DAVID (The Database for Annotation, Visualization and Integrated Discovery) tools (15).

Results

Clinical characteristics

The clinical characteristics of samples in dataset 1 and 2 are summarized in Table I. We used dataset 1 for the differential expression analysis to pre-select the survival gene candidates. Within this dataset, a total of 51 patients (16 males and 35 females with a median age 65 years; range, 23–84 years) were analysed. All tumours were diagnosed and classified according to WHO criteria: 33 patients (64.71%) were diagnosed as grade I, 12 patients (23.53%) as grade II, and 2 patients (3.92%) as grade III. On the other hand, we used dataset 2 for the identification of survival related gene expression profile. Among the 68 samples (25 males and 43 females with a median age 64 years; range, 32–89 years), 43 patients (63.24%) were diagnosed as grade I, 19 patients (27.94%) as grade II and 6 patients (8.82%) as grade III. The median survival time is 1,726 days with range of 19–3,387 days.

Table I

Clinical characteristics of the patients with meningioma tumours.

Table I

Clinical characteristics of the patients with meningioma tumours.

DemographicsDataset 1, n=51Dataset 2, n=68
Survival time, days
 MedianNA1,726
 RangeNA19–3,387
Age, years
 Median6564
 Range23–8432–89
Gender
 Male1625
 Female3543
Grade
 I3343
 II1219
 III26
 Normal4NA
Identification of a 37-gene signature for meningioma prognosis

Our hypothesis is that the genes involved in the progression of meningioma may also contribute to the prediction of prognosis. Therefore, we firstly conducted differential expression analysis in dataset 1 using Affymetrix Human Genome U133A microarray. A total of 449 genes (109 upregulated and 340 downregulated) were identified as significantly differentially expressed in meningioma. As shown in Fig. 1, the threshold is set as a log-odds value of >4.6 (99% probability that the gene is differentially expressed) and a fold-change >2-fold. The 449 genes were used as a discovery set for the identification of survival related profiles in meningioma.

To investigate the prognostic potential of these genes, we prepared dataset 2. To deal with cross-platform microarray data appropriately, we mapped the probe set IDs from two microarray platforms to official symbols. A univariate Cox proportional hazard model showed that expression levels of 51 probes (representing 37 non-redundant genes) were correlated with overall survival time (P<0.05) (Table II). As shown in Fig. 2, the 37-gene signature was the dominant characteristics that permitted the stratification of individuals into cancerous and normal groups. To determine whether the gene expression profiles could accurately predict overall survival, hierarchical clustering was used to classify all of the samples from dataset 2 into two groups as high- and low-risk groups (Fig. 3A). Kaplan-Meier analysis demonstrated that the high- and low-risk groups were significantly different in their overall survival (P<0.01) (Fig. 3B).

Table II

Thirty-seven genes composing the survival related profile.

Table II

Thirty-seven genes composing the survival related profile.

SymbolChromosome locationDescription
ARHGDIA17q25.3Rho GDP dissociation inhibitor (GDI) α
ATP6V1G26p21.3ATPase, H+ transporting, lysosomal 13 kDa, V1 subunit G2
BCAS120q13.2Breast carcinoma amplified sequence 1
CD2003q12-q13CD200 molecule
CNOT319q13.4CCR4-NOT transcription complex, subunit 3
CSF317q11.2-q12Colony stimulating factor 3 (granulocyte)
CSGALNACT210q11.21Chondroitin sulfate N-acetylgalactosaminyltransferase 2
CTBP14p16C-terminal binding protein 1
CXCL34q21Chemokine (C-X-C motif) ligand 3
EIF2S3Xp22.2-p22.1Eukaryotic translation initiation factor 2, subunit 3 γ, 52 kDa
EIF5A17p13-p12Eukaryotic translation initiation factor 5A
ELAVL119p13.2ELAV (embryonic lethal, abnormal vision, Drosophila)-like 1 (Hu antigen R)
ERG21q22.3v-ets erythroblastosis virus E26 oncogene homolog (avian)
ESR16q25.1Estrogen receptor 1
FKBP819p12FK506 binding protein 8, 38 kDa
FUS16p11.2Fused in sarcoma
HBD11p15.5Hemoglobin, δ
HLA-F6p21.3Major histocompatibility complex, class I, F
HMGN121q22.2High mobility group nucleosome binding domain 1
HNRNPR1p36.12Heterogeneous nuclear ribonucleoprotein R
IL84q13-q21Interleukin 8
L1CAMXq28L1 cell adhesion molecule
LOC1001295186q25.3Uncharacterized LOC100129518
MAFG17q25.3v-maf musculoaponeurotic fibrosarcoma oncogene homolog G (avian)
MBP18q23Myelin basic protein
NUPL113q12.13Nucleoporin like 1
PAPOLA14q32.31Poly(A) polymerase α
PRRC2B9q34.13Proline-rich coiled-coil 2B
SCAF421q22.1SR-related CTD-associated factor 4
SENP317p13SUMO1/sentrin/SMT3 specific peptidase 3
SET9q34SET nuclear oncogene
SETP4Xq21.1SET pseudogene 4
SOD26q25.3Superoxide dismutase 2, mitochondrial
SOX1022q13.1SRY (sex determining region Y)-box 10
SUMO217q25.1SMT3 suppressor of mif two 3 homolog 2 (S. cerevisiae)
USP464q12Ubiquitin specific peptidase 46
WBP413q14.11WW domain binding protein 4
Characterization of survival related profile

We conducted gene set enrichment analysis to understand the biological characteristics of the 37-gene signature. The gene signature includes three oncogenes, FUS (FUS RNA binding protein), ERG (v-ets avian erythroblastosis virus E26 oncogene homolog) and SET (SET nuclear proto-oncogene); two cell differentiation markers, CD200 (CD200 molecule) and L1CAM (L1 cell adhesion molecule); five transcription factors, ESR1 (estrogen receptor 1), MAFG (v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog G), CNOT3 (CCR4-NOT transcription complex, subunit 3), SOX10 (sex determining region Y-box 10) and ERG.

To characterize the gene list based on GO classification on ‘biological process’, ‘molecular function’ and ‘cellular component’, we examined in which categories the gene signature was significantly enriched. The 37 genes were significantly (P-value <0.05) enriched in 16 GO categories. In the biological process class, the genes were notably enriched in behaviour (GO:0007610), regulation of cell proliferation (GO:0042127), pigment cell differentiation (GO:0050931), regulation of cell adhesion (GO:0030155), negative regulation of cellular component organization (GO:0051129), neutrophil chemotaxis (GO:0030593) and mRNA metabolic process (GO:0016071). In cellular component class, the genes were significantly enriched in axon part (GO:0033267), internal side of plasma membrane (GO:0009898), integral to organelle membrane (GO:0031301), intrinsic to organelle membrane (GO:0031300), cytosol (GO:0005829), axon (GO:0030424), plasma membrane part (GO:0044459) and terminal button (GO:0043195). In the molecular function class, the genes were only enriched in translation factor activity, nucleic acid binding (GO:0008135) (Fig. 4).

However, we could not identify any pathway that the 37 genes were significantly enriched in.

Discussion

In the present study, we identified a 37-gene prognostic signature for meningioma patients across two types of microarray expression profiling datasets by using Cox proportional hazard models. This gene signature was independently predictive of survival and outperforming current pathological staging criteria.

Our survival gene signature consists of genes that showed prognostic potential in other cancer types. For instance, the gene ARHGDIA [Rho GDP dissociation inhibitor (GDI) alpha] is associated with mesothelioma (16,17) prognosis and could serve as an independent prognostic factor in hepatocellular carcinoma (18). Another example is the gene CD200. CD200 is expressed in most PCM (plasma cell myeloma) cases and its expression level remains stable even during the treatment process, which could serve as a useful marker for the prognosis of PCM (19,20). It is also reported that CD200 could be a new prognostic factor in acute myeloid leukaemia (21). ESR1, a ligand-activated transcription factor, involves in pathological processes of breast (2225), prostate (26) and non-small cell lung cancer (27).

Surprisingly, from the differential expression analysis, a small number of genes that upregulated or downregulated were identified in meningioma comparing to normal meninges. This suggests that meningioma and normal meninges may remain largely homogeneous at global gene expression level. However, the identified differentially expressed genes appear to be clinically and even biologically important, which could ultimately influence the prognostic outcome.

To the best of our knowledge, the present study is the first to identify a prognostic signature for meningioma prognosis based on genome wide expression profiling technologies. However, because most of the current public accessible meningioma data lack sufficient follow-up information, we could not evaluate the prognostic predictive power in other independent datasets. For future work, we will continuously work on optimizing the prognosis model by integrating more meningioma datasets.

In conclusion, these results suggest that the 37-gene signature could serve as a useful tool to predict the meningioma progression and survival. To apply in the clinical practice, a prospective multi-centre study is still needed to improve the predictive ability and reliability of the gene expression profile. Nevertheless, the survival related gene expression profile could provide an optimization of the clinical management and development of new therapeutic strategies for meningioma.

References

1 

Claus EB, Bondy ML, Schildkraut JM, Wiemels JL, Wrensch M and Black PM: Epidemiology of intracranial meningioma. Neurosurgery. 57:1088–1095. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Pasquier D, Bijmolt S, Veninga T, et al: Atypical and malignant meningioma: outcome and prognostic factors in 119 irradiated patients. A multicenter, retrospective study of the Rare Cancer Network. Int J Radiat Oncol Biol Phys. 71:1388–1393. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Linsler S, Kraemer D, Driess C, et al: Molecular biological determinations of meningioma progression and recurrence. PloS One. 9:e949872014. View Article : Google Scholar : PubMed/NCBI

4 

Konstantinopoulos PA, Spentzos D and Cannistra SA: Gene-expression profiling in epithelial ovarian cancer. Nat Clin Pract Oncol. 5:577–587. 2008. View Article : Google Scholar : PubMed/NCBI

5 

van ‘t Veer LJ, Dai H, van de Vijver MJ, et al: Gene expression profiling predicts clinical outcome of breast cancer. Nature. 415:530–536. 2002. View Article : Google Scholar

6 

Motoori M, Takemasa I, Yano M, et al: Prediction of recurrence in advanced gastric cancer patients after curative resection by gene expression profiling. Int J Cancer. 114:963–968. 2005. View Article : Google Scholar : PubMed/NCBI

7 

Chen HY, Yu SL, Chen CH, et al: A five-gene signature and clinical outcome in non-small-cell lung cancer. N Engl J Med. 356:11–20. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Schramm A, Schulte JH, Klein-Hitpass L, et al: Prediction of clinical outcome and biological characterization of neuroblastoma by expression profiling. Oncogene. 24:7902–7912. 2005. View Article : Google Scholar : PubMed/NCBI

9 

Tabernero MD, Maillo A, Gil-Bellosta CJ, et al: Gene expression profiles of meningiomas are associated with tumor cytogenetics and patient outcome. Brain Pathol. 19:409–420. 2009. View Article : Google Scholar

10 

Lee Y, Liu J, Patel S, et al: Genomic landscape of meningiomas. Brain Pathol. 20:751–762. 2010. View Article : Google Scholar

11 

Wu Z, Irizarry RA, Gentleman R, Martinez-Murillo F and Spencer F: A model-based background adjustment for oligonucleotide expression arrays. J Am Stat Assoc. 99:909–917. 2004. View Article : Google Scholar

12 

Irizarry RA, Hobbs B, Collin F, et al: Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics. 4:249–264. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Smyth GK: Limma: linear models for microarray data. Bioinformatics and Computational Biology Solutions Using R and Bioconductor. Springer; New York: pp. 397–420. 2005, View Article : Google Scholar

14 

Therneau TM and Grambsch PM: Modeling Survival Data: Extending the Cox Model. Springer; New York: 2000

15 

Huang da W, Sherman BT and Lempicki RA: Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 4:44–57. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Gordon GJ, Bueno R and Sugarbaker DJ: Genes associated with prognosis after surgery for malignant pleural mesothelioma promote tumor cell survival in vitro. BMC Cancer. 11:1692011. View Article : Google Scholar : PubMed/NCBI

17 

Gordon GJ, Dong L, Yeap BY, et al: Four-gene expression ratio test for survival in patients undergoing surgery for mesothelioma. J Natl Cancer Inst. 101:678–686. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Li W, Wang H, Jin X and Zhao L: Loss of RhoGDI is a novel independent prognostic factor in hepatocellular carcinoma. Int J Clin Exp Pathol. 6:2535–2541. 2013.PubMed/NCBI

19 

Douds JJ, Long DJ, Kim AS and Li S: Diagnostic and prognostic significance of CD200 expression and its stability in plasma cell myeloma. J Clin Pathol. 67:792–796. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Alapat D, Coviello-Malle J, Owens R, et al: Diagnostic usefulness and prognostic impact of CD200 expression in lymphoid malignancies and plasma cell myeloma. Am J Clin Pathol. 137:93–100. 2012. View Article : Google Scholar

21 

Tonks A, Hills R, White P, et al: CD200 as a prognostic factor in acute myeloid leukaemia. Leukemia. 21:566–568. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Caizzi L, Ferrero G, Cutrupi S, et al: Genome-wide activity of unliganded estrogen receptor-alpha in breast cancer cells. Proc Natl Acad Sci USA. 111:4892–4897. 2014. View Article : Google Scholar

23 

Gao QG, Chan HY, Man CW and Wong MS: Differential ERalpha-mediated rapid estrogenic actions of ginsenoside Rg1 and estren in human breast cancer MCF-7 cells. J Steroid Biochem Mol Biol. 141:104–112. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Martinez-Galan J, Torres-Torres B, Nunez MI, et al: ESR1 gene promoter region methylation in free circulating DNA and its correlation with estrogen receptor protein expression in tumor tissue in breast cancer patients. BMC Cancer. 14:592014. View Article : Google Scholar : PubMed/NCBI

25 

Toy W, Shen Y, Won H, et al: ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet. 45:1439–1445. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Gu Z, Wang G and Chen W: Estrogen receptor alpha gene polymorphisms and risk of prostate cancer: a meta-analysis involving 18 studies. Tumour Biol. 35:5921–5930. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Atmaca A, Al-Batran SE, Wirtz RM, et al: The validation of estrogen receptor 1 mRNA expression as a predictor of outcome in patients with metastatic non-small cell lung cancer. Int J Cancer. 134:2314–2321. 2014. View Article : Google Scholar

Related Articles

Journal Cover

February-2015
Volume 46 Issue 2

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen F, Xiang C, Zhou Y, Ao X, Zhou D, Peng P, Zhang H, Liu H and Huang X: Gene expression profile for predicting survival of patients with meningioma. Int J Oncol 46: 791-797, 2015
APA
Chen, F., Xiang, C., Zhou, Y., Ao, X., Zhou, D., Peng, P. ... Huang, X. (2015). Gene expression profile for predicting survival of patients with meningioma. International Journal of Oncology, 46, 791-797. https://doi.org/10.3892/ijo.2014.2779
MLA
Chen, F., Xiang, C., Zhou, Y., Ao, X., Zhou, D., Peng, P., Zhang, H., Liu, H., Huang, X."Gene expression profile for predicting survival of patients with meningioma". International Journal of Oncology 46.2 (2015): 791-797.
Chicago
Chen, F., Xiang, C., Zhou, Y., Ao, X., Zhou, D., Peng, P., Zhang, H., Liu, H., Huang, X."Gene expression profile for predicting survival of patients with meningioma". International Journal of Oncology 46, no. 2 (2015): 791-797. https://doi.org/10.3892/ijo.2014.2779