Open Access

Construction of a 26‑feature gene support vector machine classifier for smoking and non‑smoking lung adenocarcinoma sample classification

  • Authors:
    • Lei Yang
    • Lu Sun
    • Wei Wang
    • Hao Xu
    • Yi Li
    • Jia‑Ying Zhao
    • Da‑Zhong Liu
    • Fei Wang
    • Lin‑You Zhang
  • View Affiliations

  • Published online on: December 7, 2017     https://doi.org/10.3892/mmr.2017.8220
  • Pages: 3005-3013
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The present study aimed to identify the feature genes associated with smoking in lung adenocarcinoma (LAC) samples and explore the underlying mechanism. Three gene expression datasets of LAC samples were downloaded from the Gene Expression Omnibus database through pre‑set criteria and the expression data were processed using meta‑analysis. Differentially expressed genes (DEGs) between LAC samples of smokers and non‑smokers were identified using limma package in R. The classification accuracy of selected DEGs were visualized using hierarchical clustering analysis in R language. A protein‑protein interaction (PPI) network was constructed using gene interaction data from the Human Protein Reference Database for the DEGs. Betweenness centrality was calculated for each node in the network and genes with the greatest BC values were utilized for the construction of the support vector machine (SVM) classifier. The dataset GSE43458 was used as the training dataset for the construction and the other datasets (GSE12667 and GSE10072) were used as the validation datasets. The classification accuracy of the classifier was tested using sensitivity, specificity, positive predictive value, negative predictive value and area under curve parameters with the pROC package in R language. The feature genes in the SVM classifier were subjected to pathway enrichment analysis using Fisher's exact test. A total of 347 genes were identified to be differentially expressed between samples of smokers and non‑smokers. The PPI network of DEGs were comprised of 202 nodes and 300 edges. An SVM classifier comprised of 26 feature genes was constructed to distinguish between different LAC samples, with prediction accuracies for the GSE43458, GSE12667 and GSE10072 datasets of 100, 100 and 94.83%, respectively. Furthermore, the 26 feature genes that were significantly enriched in 9 overrepresented biological pathways, including extracellular matrix‑receptor interaction, proteoglycans in cancer, cell adhesion molecules, p53 signaling pathway, microRNAs in cancer and apoptosis, were identified to be smoking‑related genes in LAC. In conclusion, an SVM classifier with a high prediction accuracy for smoking and non‑smoking samples was obtained. The genes in the classifier may likely be the potential feature genes associated with the development of patients with LAC who smoke.

Introduction

Lung cancer is the most common cause of cancer-associated fatality in men and the second most common in women (1). The 5-year survival rate following diagnosis of lung cancer is 15.6%, making it one of the worst prognostic malignant tumors (2). The survival rate is lower compared with breast, colon and prostate cancer (2). Cigarette smoking is responsible for ~90% of lung cancer incidences and leads to decreased survival rates (3).

The major histological types of lung cancer include adenocarcinoma, squamous cell carcinoma, large cell carcinoma and small cell carcinoma. The incidence of lung adenocarcinoma (LAC) increased gradually and this lung cancer has been the most frequently occurring histological type in most parts of the world in recent years (4). Adenocarcinoma account for ~40% of all lung cancer cases (5). Smoking is a major cause of lung adenocarcinoma (6). However, the causes of the increase in adenocarcinomas are not clear.

Sequencing data from large-scale databases, such as The Cancer Genome Atlas, have aided in identification of novel factors and potentially targetable alterations in lung adenocarcinomas (7). A number of smoking-associated genes have been revealed in LAC, including the cyclin D1 A870 G gene, and polymorphisms of this gene have been indicated to modulate smoking-induced lung cancer risk (8). Estrogen receptor α promotes smoking carcinogen-induced lung carcinogenesis via cytochrome P450 1B1 (9). The interactions between smoking, polymorphisms of human 8-oxoguanine DNA glycosylase and p53 are associated with the development of lung cancer (10). Interactions between smoking, fragile histidine triad gene alterations (11) and excision repair cross-complementation group 1 polymorphisms (12) have also been reported in lung cancer. However, the recognized genetic changes in patients with LAC who are smokers remain to be elucidated and further studies are necessary to determine the underlying molecular mechanism of smoking-induced LAC.

A recent study has aimed to identify smoking-associated genes via the differential analysis of RNA sequencing data (13). The study analyzed two datasets with only two samples and identified 1,603 differentially expressed genes (DEGs). The authors also identified that the possible alternative splicing of gene FCGBP may have an impact on lung cancer. However, the small sample size could lead to low reliability of the results.

In the present study, three gene expression datasets of smokers and non-smokers with LAC (>50 samples/group) were obtained and DEGs were identified using meta-analysis. A protein-protein interaction (PPI) network of the DEGs was constructed with the betweenness centrality (BC) analysis for the selection of feature genes. Using the feature genes, a support vector machine (SVM) classifier, which is able to distinguish between samples from smokers and non-smokers with a high classification accuracy, was constructed. The feature genes in the SVM classifier were considered as the smoking-related genes in LAC and enrichment analysis was conducted to identify significant pathways.

Materials and methods

Gene expression data

To collect gene expression data from patients with LAC who smoke or do not smoke, the Gene Expression Omnibus (GEO; www.ncbi.nlm.nih.gov/geo/) database was used and the key words ‘lung adenocarcinoma’, ‘Homo sapiens’ and ‘smoke’ were searched. The following inclusion criteria were used to extract the corresponding datasets: i) They were gene expression data; ii) they were from LAC samples; iii) information concerning smoking was described; and iv) ≥50 samples were included in each dataset. A total of three datasets were collected from the GEO database, including GSE43458 (14), GSE10072 (15) and GSE12667 (16) (Table I).

Table I.

Data of the three collected gene expression datasets.

Table I.

Data of the three collected gene expression datasets.

Accession numberPlatformTotal samples (n)Non-smokers (n)Smokers (n)
GSE43458 HuGene-1_0-st-v11104040
GSE10072HG-U133A1071642
GSE12667HG-U133_Plus_2  75  843

Raw data in these three datasets were analyzed with the affy package in R 3.2.1 (http://bioconductor.org/packages/release/bioc/html/affy.html) (17). Probes were subsequently mapped into genes. Probes corresponding to one gene were averaged as the final expression value of the gene. Normalization was performed with package limma (18) of R to conduct the analysis of the datasets.

Screening of DEGs

Meta-analysis was used to enforce the analytical reliability for gene expression data by combining data from different datasets. DEGs associated with smoking in the three gene expression datasets were screened via meta-analysis using the MetaDE.ES package of R (19). The method tested the heterogeneity of gene expression value from three datasets with three statistic parameters: Tau2, Q-value and Qpval. Subsequently, differential expression of genes between smoking and non-smoking samples was assessed by determining the P-value and false discovery rate (FDR). To determine the DEGs associated with smoking, tau2=0, Qpval >0.05 and FDR <0.05 were set as the cut-off points. Bidirectional clustering analysis using the pheatmap package in R language (https://cran.r-project.org/web/packages/pheatmap/index.html), which was based on the euclidean distance calculations for gene expression values, was also conducted to examine whether the selected DEGs were able to distinguish different samples, as described previously (20).

Construction of PPI network

To investigate the interactions of DEGs, the DEGs were mapped to the PPI database using the Human Protein Reference Database (21). The interactions of DEGs obtained were constructed into a PPI network with the proteins that were connected with at least three DEGs. The network was visualized with Cytoscape (22).

Calculation of BC

Feature genes that function as hub nodes in the PPI network were screened using a BC algorithm (23). BC represented the degree of node in the network and was calculated as follows:

CB(v)=∑t≠v≠u∈Vσst(v)σst

Where σst is the total number of shortest paths from node s to node t; σst(ν) is the number of shortest paths from s to t going through v; BC scores were between 0 and 1, and a higher BC score indicated a higher degree of the node.

Training and validation of SVM classifier

SVM classifier comprises of feature genes that distinguishes between different samples (24,25). To construct the SVM classifier, one of the downloaded datasets, GSE43458 (containing 40 non-smokers and 40 smokers) was selected as the training dataset basing on the top 10, 20, 30, 40 and 50 feature genes ranked by BC scores. The feature genes in the SVM classifier that could exactly distinguish between different samples in GSE42458 were subjected to two-way clustering analysis using pheatmap package in R 3.1.4 (https://cran.r-project.org/web/packages/pheatmap/index.html). Sample similarity matrices were also obtained by computing the Pearson's correlation coefficients of these genes using Cor package in R 3.1.4 (https://stat.ethz.ch/R-manual/R-devel/library/stats/html/cor.html) and top 50 genes were selected for further analysis. The clustering and similarity matrices were visualized using heatmaps in pheatmap package in R 3.1.4 (https://cran.r-project.org/web/packages/pheatmap/index.html).

The SVM classifier was validated with two independent datasets, GSE10072 and GSE12667. Sensitivity (Se), specificity (Sp), positive predictive value (PPV), negative predictive value (NPV) and area under curve (AUC) were calculated using the pROC package in R language (https://cran.r-project.org/web/packages/pROC/index.html) to examine the classification accuracy of the SVM classifier as described previously (26,27).

Pathway enrichment analysis

Feature gene-related Kyoto Encyclopedia of Genes and Genomes pathways (http://www.genome.jp/kegg/) were revealed using Fisher's exact test as follows:

p=1–∑i=0x–1(Mi)(N–MK–i)(NK)

Where N represented the total number of genes; M represented the number of genes in the pathway; and K indicated the number of feature genes.

Results

DEGs

A total of 12,476 genes were in the three gene expression datasets, and according to the set criteria, 347 DEGs between smoking and non-smoking LAC samples were identified. The top 10 DEGs ranked by FDR are listed in Table II. As indicated in Fig. 1, the 347 DEGs distinguished the samples of smokers from the non-smokers.

Table II.

Top 10 candidate feature genes by FDR.

Table II.

Top 10 candidate feature genes by FDR.

IDP-valueFDR tau2QpvalQvalExpression
ABCB11 1.52×10−05 5.59×10−040 9.27×10−01 8.28×10−03Up
ABCB6 2.27×10−03 2.23×10−020 9.63×10−01 2.18×10−03Up
ABCC2 2.11×10−03 2.11×10−020 9.51×10−01 3.83×10−03Up
ABCG5 4.01×10−06 2.10×10−040 9.00×10−01 1.58×10−02Up
ACD 4.81×10−06 2.38×10−040 9.40×10−01 5.71×10−03Up
ADAMTS5 1.88×10−04 3.79×10−030 9.10×10−01 1.26×10−02Up
AGT 2.89×10−03 2.64×10−020 8.47×10−01 3.71×10−02Up
AIM1L 1.00×10−20 7.47×10−190 8.26×10−01 4.84×10−02Up
AKAP6 1.15×10−04 2.69×10−030 9.82×10−01 5.08×10−04Down
ALPL 5.01×10−03 3.88×10−020 9.06×10−01 1.40×10−02Down

[i] FDR, false discovery rate; UP, upregulation in smokers; DOWN, downregulation in smokers.

PPI network

A PPI network containing 202 nodes (genes) and 300 edges (connection between nodes) was obtained (Fig. 2). The proteins that were connected with ≥3 DEGs were also included in the PPI network. Degree distribution of genes in the network is indicated in Fig. 3. Similar to biological networks, the PPI network was scale-free, with the majority of genes (80 genes) exhibiting small degrees (Log transformed degree <1) and few genes (only 5) exhibiting larger degrees (Log transformed degree between 3 and 4). The genes with high degrees were hub genes, indicating their roles in the development of smoking-associated LAC.

Feature genes

BC was calculated for each node in the PPI network. The top 10 genes by BC value were considered as the feature genes, including high mobility group box 1 (HMGB1); dynein light chain LC8-type 1; tubulin α 4a; 14-3-3 protein γ; tyrosine 3-monooxygenase; spectrin β, non-erythrocytic 1; ubiquilin 4; DNA methyltransferase 1 (DNMT1); enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and glucocorticoid modulatory element binding protein 1 (Table III).

Table III.

Top 10 genes ranked using BC.

Table III.

Top 10 genes ranked using BC.

GeneBCExpressionDegreeP-valueFDRQpvalQval
HMGB1 1.98×10−01Down11 1.63×10−03 1.76×10−02 8.95×10−01 1.74×10−02
DYNLL1 1.77×10−01Up15 1.00×10−20 7.47×10−19 8.92×10−01 1.83×10−02
TUBA4A 1.37×10−01Up10 2.08×10−05 7.32×10−04 8.50×10−01 3.56×10−02
YWHAG 1.20×10−0111 9.86×10−01 9.95×10−01 5.38×10−062.07×10
YWHAQ 1.07×10−01Up10 2.40×10−04 4.55×10−03 8.46×10−01 3.77×10−02
SPTBN1 1.04×10−01Down  7 9.39×10−04 1.23×10−02 8.53×10−01 3.43×10−02
UBQLN4 1.00×10−01  7 9.31×10−01 9.67×10−01 5.95×10−02 3.55×100
DNMT1 9.98×10−02Up  7 1.20×10−04 2.77×10−03 8.61×10−01 3.05×10−02
EZH2 8.51×10−02Up  8 2.65×10−03 2.48×10−02 8.44×10−01 3.89×10−02
GMEB1 8.45×10−02Down  8 2.31×10−04 4.40×10−03 9.32×10−01 7.20×10−03

[i] BC, betweenness centrality; UP, upregulation in smokers; DOWN, downregulation in smokers; -, no significant difference in expression; FDR, false discovery rate.

SVM classifier

Feature genes with the greatest BC values were used to construct the SVM classifier basing on dataset GSE43458. There were 8, 11, 14, 16, 18, 20, 22 and 26 feature genes in the top 10, 15, 20, 25, 30, 35, 40 and 50 genes, respectively. The training process is indicated in Fig. 4. The accuracy of the classifier reached 100% when the 26 feature genes in the top 50 were included. Therefore, the classifier comprised by these 26 feature genes were chosen as the final SVM classifier. These feature genes included Cbl proto-oncogene B (CBLB), DNMT1, EZH2, HMGB1, integrin α-5 (ITGA5), MDK, protein kinase C ι (PRKCI) and sprouty receptor tyrosine kinase signaling antagonist 2 (SPRY2).

Hierarchical clustering was performed for samples from the training dataset using the 26 feature genes (Fig. 5). The classifier separated samples of smokers from samples of non-smokers in dataset GSE43458 (Fig. 6A).

The SVM classifier was validated using dataset GSE12667 and GSE10072. The classification accuracy in GSE12667 was 100% (Fig. 6B). In GSE10072, the classifier identified 42 smokers (42/42, 100%) and 13 non-smokers (13/16, 81.25%), and total accuracy was 94.83% (55/58) (Fig. 6C; Table IV). The classifier demonstrated high accuracy of 100, 100 and 94.83% in GSE43458, GSE12667 and GSE10072, respectively. Se, Sp, PPV, NPV and AUC results (Table IV) and receiver operating characteristic curves were generated (Fig. 7).

Table IV.

Prediction results of the support vector machine classifier in the three datasets.

Table IV.

Prediction results of the support vector machine classifier in the three datasets.

DatasetSamples (n)Accuracy (%)SeSpPPVNPVAUC
GSE434588010011111
GSE126675110011111
GSE100725894.8310.8130.93310.994

[i] Se, sensitivity; Sp, specificity; PPV, positive predictive value; NPV, negative predictive value; AUC, area under curve.

Overrepresented biological pathways

The 26 feature genes were indicated to be significantly enriched in nine biological pathways (Table V): Extracellular matrix (ECM)-receptor interaction, proteoglycans in cancer, cell adhesion molecules, pathogenic Escherichia coli infection, p53 signaling pathway, microRNAs in cancer, bacterial invasion of epithelial cells, apoptosis and hematopoietic cell lineage.

Table V.

A total of 9 biological pathways significantly overrepresented by the 26 feature genes.

Table V.

A total of 9 biological pathways significantly overrepresented by the 26 feature genes.

IDTermP-valueGenes
hsa04512Extracellular matrix-receptor interaction 5.68×10−03SDC4, ITGA5, COL3A1
hsa05205Proteoglycans in cancer 9.82×10−03SDC4, CBLB, ITGA5, SDC2
hsa04514Cell adhesion molecules 2.17×10−02SDC4, CD4, SDC2
hsa05130Pathogenic Escherichia coli infection 2.19×10−02YWHAQ, TUBA4A
hsa04115p53 signaling pathway 3.21×10−02APAF1, BID
hsa05206MicroRNAs in cancer 3.29×10−02EZH2, SPRY2, ITGA5, DNMT1
hsa05100Bacterial invasion of epithelial cells 3.91×10−02CBLB, ITGA5
hsa04210Apoptosis 4.86×10−02APAF1, BID
hsa04640Hematopoietic cell lineage 4.96×10−02CD4, ITGA5

Discussion

In the present study, three gene expression datasets were obtained and a total of 347 DEGs were identified in samples from smokers with LAC compared with non-smokers with LAC using meta-analysis. A PPI network including 202 nodes and 300 edges was constructed, from which 26 feature genes were identified. The SVM classifier of these 26 genes separated smokers from non-smokers with an accuracy >94% in all the three datasets. Pathway enrichment analysis demonstrated that these feature genes were primarily associated with cancer development- and metastasis-associated pathways, including ECM-receptor interaction, proteoglycans in cancer, cell adhesion molecules, p53 signaling pathway, microRNAs in cancer and apoptosis.

Due to the generalization ability, SVM has been widely used for analysis, including data classification and function approximation (2830). SVM classifier has been demonstrated to distinguish whether one cancer sample type possessed distinctive signatures of gene expressions compared with other sample types (31). In the present study, an SVM classifier with 26 feature genes successfully distinguished LAC samples of smokers and non-smokers using bioinformatics analysis. Yousef et al (32) previously conducted a similar study for the identification of biomarkers, by integrating interaction networks and an SVM classifier, and subsequently obtained >90% accuracy in classification of selected microarray datasets. Furthermore, a previous study also demonstrated that the discriminant analysis based on an SVM classifier achieved satisfactory results in the classification of lung cancer samples (33).

Specific genes within the 26 feature genes have been implicated in lung cancer or LAC. CBLB is a regulator of T-cell response (34). It has been reported that the single nucleotide polymorphisms of CBLB may predict the definitive radiotherapy outcomes for non-small cell lung cancer (NSCLC) (34). CBLB is associated with icotinib-induced apoptosis and G1 phase arrest of epidermal growth factor receptor mutation-positive NSCLC (35).

DNMT1 is responsible for maintaining methylation patterns following DNA replication and has an important role in the development of various types of cancer (36). DNA methylation alterations are recognized as key epigenetic changes in cancer, influencing the chromosomal instability through global hypomethylation and aberrant gene expression via the alterations in methylation levels (37). The tobacco-specific carcinogen nicotine-derived nitrosamine ketone induces the accumulation of DNMT1 in patients with lung cancer (38). Furthermore, DNMT1 inhibits the expression of, the tumor suppressor Wnt7a in NSCLC (39).

EZH2 is a member of the polycomb-group family, which is associated with maintaining the transcriptional repressive state of genes over successive cell generations (40). Yoon et al (41) previously suggested a correlation between the genotype variants in EZH2 and reduced lung cancer risk. Additionally, Zhang et al (42) determined that miR-138 inhibited tumor growth through the repression of EZH2 in NSCLC. Notably, a recent study indicated that EZH2 silencing with RNA interference induced G2/M arrest in human lung cancer cells in vitro (43), and Wang et al (44) recently demonstrated that EZH2 overexpression was associated with a poor prognosis for patients with LAC. In the present study, it was indicated that EZH2 was upregulated in the samples of smokers and thus the present findings suggest that EZH2 upregulation may result from smoking.

HMGB1 has a role in tumor cell migration (45). Shen et al (46) indicated that the expression of HMGB1 correlates with the progression of NSCLC. ITGA5 is considered as a prognostic indicator in NSCLC (47).

MDK promotes cell growth, migration and angiogenesis, in particular during tumorigenesis (48). A previous study indicated that MDK protein overexpression is correlated with the malignant status and prognosis of NSCLC (49). Furthermore, MDK has been targeted as a therapeutic biomarker for lung cancer (50).

PRKCI is required for lung tumorigenesis as genetic loss of PRKCI inhibits Kras-initiated hyperplasia and subsequent lung tumor formation in vivo (51). SPRY2 inhibits cell migration and proliferation in NSCLC (52). In addition, a previous study has indicated that downregulation of SPRY2 in NSCLC contributes to tumor malignancy (53).

Smoking can cause LAC and the incidence of this disease increased in recent years (4). However, the reason for this increase and the mechanism underlying smoking-associated development of LAC remain to be elucidated. The present study identified genes implicated in smoking-associated LAC, including CBLB, DNMT1, EZH2, HMGB1, ITGA5, MDK, PRKCI and SPRY2. Most of these genes have been reported in association with malignancy and certain were associated with lung cancer. The identification of these characteristic genes may aid in elucidating the mechanism underlying smoking associated-lung adenocarcinoma. Although further experiments such as validation the gene and protein expression level in the smoking and non-smoking LAC samples were not performed limited by the LAC samples available, these results may provide information to other researchers in the field.

In conclusion, a number of key genes have been revealed in smokers with LAC and some of these have been implicated in lung cancer. However, the associations between the 26 feature genes, smoking and LAC remain to be fully elucidated with further studies.

Acknowledgements

The present study was supported by The Natural Science Foundation of Heilongjiang Province (The Science Foundation for Returned Overseas Students in Heilongjiang Province; grant nos. LC2016036 and LC2015040) and Research Foundation of Harbin Medical University Affiliated Second Hospital (grant nos. KYBS2015-10 and KYBS2015-01).

References

1 

Centers for Disease Control and Prevention (CDC), . State-specific trends in lung cancer incidence and smoking-United States, 1999–2008. MMWR Morb Mortal Wkly Rep. 60:1243–1247. 2011.PubMed/NCBI

2 

Nanavaty P, Alvarez MS and Alberts WM: Lung cancer screening: Advantages, controversies, and applications. Cancer Control. 21:9–14. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Bryant A and Cerfolio RJ: Differences in epidemiology, histology, and survival between cigarette smokers and never-smokers who develop non-small cell lung cancer. Chest. 132:185–192. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Nakamura H and Saji H: Worldwide trend of increasing primary adenocarcinoma of the lung. Surg Today. 44:1004–1012. 2014. View Article : Google Scholar : PubMed/NCBI

5 

Kong J, Xu F, He M, Chen K and Qian B: The incidence of lung cancer by histological type: A population-based study in Tianjin, China during 1981–2005. Respirology. 19:1222–1228. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Cancer Genome Atlas Research Network, . Comprehensive molecular profiling of lung adenocarcinoma. Nature. 511:543–550. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Devarakonda S, Morgensztern D and Govindan R: Genomic alterations in lung adenocarcinoma. Lancet Oncol. 16:e342–e351. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Gautschi O, Hugli B, Ziegler A, Bigosch C, Bowers NL, Ratschiller D, Jermann M, Stahel RA, Heighway J and Betticher DC: Cyclin D1 (CCND1) A870G gene polymorphism modulates smoking-induced lung cancer risk and response to platinum-based chemotherapy in non-small cell lung cancer (NSCLC) patients. Lung Cancer. 51:303–311. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Li MY, Liu Y, Liu LZ, Kong AW, Zhao Z, Wu B, Long X, Wu J, Ng CS, Wan IY, et al: Estrogen receptor alpha promotes smoking-carcinogen-induced lung carcinogenesis via cytochrome P450 1B1. J Mol Med. 93:1221–1233. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Cheng Z, Wang W, Song YN, Kang Y and Xia J: hOGG1, p53 genes, and smoking interactions are associated with the development of lung cancer. Asian Pac J Cancer Prev. 13:1803–1808. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Zhang J, Chen D, Shen QM, Tian DL, Jiang YH, Yin HN and Li HW: Association between cigarette smoking and FHIT gene alterations in Chinese lung cancer. Lung Cancer. 29:2352000. View Article : Google Scholar

12 

Zhou W, Liu G, Park S, Wang Z, Wain JC, Lynch TJ, Su L and Christiani DC: Gene-smoking interaction associations for the ERCC1 polymorphisms in the risk of lung cancer. Cancer Epidemiol Biomarkers Prev. 14:491–496. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Zhou C, Chen H, Han L, Xue F, Wang A and Liang YJ: Screening of genes related to lung cancer caused by smoking with RNA-Seq. Eur Rev Med Pharmacol Sci. 18:117–125. 2014.PubMed/NCBI

14 

Kabbout M, Garcia MM, Fujimoto J, Liu DD, Woods D, Chow CW, Mendoza G, Momin AA, James BP, Solis L, et al: ETS2 mediated tumor suppressive function and MET oncogene inhibition in human non-small cell lung cancer. Clin Cancer Res. 19:3383–3395. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Landi MT, Dracheva T, Rotunno M, Figueroa JD, Liu H, Dasgupta A, Mann FE, Fukuoka J, Hames M, Bergen AW, et al: Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One. 3:e16512008. View Article : Google Scholar : PubMed/NCBI

16 

Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, et al: Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 455:1069–1075. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Gautier L, Cope L, Bolstad BM and Irizarry RA: Affy-analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 20:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Smyth GK: Limma: Linear models for microarray dataBioinformatics and Computational Biology Solutions Using R and Bioconductor. Gentleman R, Carey V, Huber W, Irizarry R and Dudoit S: Springer; New York: pp. 397–420. 2004

19 

Wang X, Kang DD, Shen K, Song C, Lu S, Chang LC, Liao SG, Huo Z, Tang S, Ding Y, et al: An R package suite for microarray meta-analysis in quality control, differentially expressed gene analysis and pathway enrichment detection. Bioinformatics. 28:2534–2536. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Wang L, Cao C, Ma Q, Zeng Q, Wang H, Cheng Z, Zhu G, Qi J, Ma H, Nian H and Wang Y: RNA-seq analyses of multiple meristems of soybean: Novel and alternative transcripts, evolutionary and functional implications. BMC Plant Biol. 14:1692014. View Article : Google Scholar : PubMed/NCBI

21 

Keshava Prasad TS, Goel R, Kandasamy K, Keerthikumar S, Kumar S, Mathivanan S, Telikicherla D, Raju R, Shafreen B, Venugopal A, et al: Human protein reference database-2009 update. Nucleic Acids Res. 37(Database issue): D767–D772. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Smoot ME, Ono K, Ruscheinski J, Wang PL and Ideker T: Cytoscape 2.8: New features for data integration and network visualization. Bioinformatics. 27:431–432. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Barthélemy M: Betweenness centrality in large complex networks. Eur Phy J Conden Matter Com Sys. 38:163–168. 2004.

24 

Zhang HH, Ahn J, Lin X and Park C: Gene selection using support vector machines with non-convex penalty. Bioinformatics. 22:88–95. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Brown MP, Grundy WN, Lin D, Cristianini N, Sugnet CW, Furey TS, Ares M Jr and Haussler D: Knowledge-based analysis of microarray gene expression data using support vector machines. Proc Natl Acad Sci USA. 97:pp. 262–267. 2000; View Article : Google Scholar : PubMed/NCBI

26 

Stojanović M, Andjelković Apostolović M, Stojanović D, Milosević Z, Ignjatović A, Lakusić VM and Golubović M: Understanding sensitivity, specificity and predictive values. Vojnosanit Pregl. 71:1062–1065. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Parikh R, Mathai A, Parikh S, Chandra Sekhar G and Thomas R: Understanding and using sensitivity, specificity and predictive values. Indian J Ophthalmol. 56:45–50. 2008. View Article : Google Scholar : PubMed/NCBI

28 

Orru G, Pettersson-Yeo W, Marquand AF, Sartori G and Mechelli A: Using support vector machine to identify imaging biomarkers of neurological and psychiatric disease: A critical review. Neurosci Biobehav Rev. 36:1140–1152. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Fan XJ, Wan XB, Huang Y, Cai HM, Fu XH, Yang ZL, Chen DK, Song SX, Wu PH, Liu Q, et al: Epithelial-mesenchymal transition biomarkers and support vector machine guided model in preoperatively predicting regional lymph node metastasis for rectal cancer. Br J Cancer. 106:1735–1741. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Han M, Dai J, Zhang Y, Lin Q, Jiang M, Xu X, Liu Q and Jia J: Support vector machines coupled with proteomics approaches for detecting biomarkers predicting chemotherapy resistance in small cell lung cancer. Oncol Rep. 28:2233–2238. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Guyon I, Weston J and Barnhill S: Gene selection for cancer classification using support vector machines. Machine Learning. 46:389–422. 2002. View Article : Google Scholar

32 

Yousef M, Ketany M, Manevitz L, Showe LC and Showe MK: Classification and biomarker identification using gene network modules and support vector machines. BMC Bioinformatics. 10:3372009. View Article : Google Scholar : PubMed/NCBI

33 

Guan P, Huang D, He M and Zhou B: Lung cancer gene expression database analysis incorporating prior knowledge with support vector machine-based classification method. J Exp Clin Cancer Res. 28:1032009. View Article : Google Scholar : PubMed/NCBI

34 

Li P, Wang X, Liu Z, Liu H, Xu T, Wang H, Gomez DR, Nguyen QN, Wang LE, Teng Y, et al: Single nucleotide polymorphisms in CBLB, a regulator of T-cell response, predict radiation pneumonitis and outcomes after definitive radiotherapy for non-small-cell lung cancer. Clin Lung Cancer. 17:253–262. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Mu X, Zhang Y, Qu X, Hou K, Kang J, Hu X and Liu Y: Ubiquitin ligase Cbl-b is involved in icotinib (BPI-2009H)-induced apoptosis and G1 phase arrest of EGFR mutation-positive non-small-cell lung cancer. Biomed Res Int. 2013:7263752013. View Article : Google Scholar : PubMed/NCBI

36 

Rountree MR, Bachman KE, Herman JG and Baylin SB: DNA methylation, chromatin inheritance, and cancer. Oncogene. 20:3156–3165. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Kerr KM, Galler JS, Hagen JA, Laird PW and Laird-Offringa IA: The role of DNA methylation in the development and progression of lung adenocarcinoma. Dis Markers. 23:5–30. 2007. View Article : Google Scholar : PubMed/NCBI

38 

Lin RK, Hsieh YS, Lin P, Hsu HS, Chen CY, Tang YA, Lee CF and Wang YC: The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients. J Clin Invest. 120:521–532. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Tennis MA, Vanscoyk MM, Wilson LA, Kelley N and Winn RA: Methylation of Wnt7a is modulated by DNMT1 and cigarette smoke condensate in non-small cell lung cancer. PLoS One. 7:e329212012. View Article : Google Scholar : PubMed/NCBI

40 

McCabe MT and Creasy CL: EZH2 as a potential target in cancer therapy. Epigenomics. 6:341–351. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Yoon KA, Gil HJ, Han J, Park J and Lee JS: Genetic polymorphisms in the polycomb group gene EZH2 and the risk of lung cancer. J Thorac Oncol. 5:10–16. 2010. View Article : Google Scholar : PubMed/NCBI

42 

Zhang H, Zhang H, Zhao M, Lv Z, Zhang X, Qin X, Wang H, Wang S, Su J, Lv X, et al: miR-138 inhibits tumor growth through repression of EZH2 in non-small cell lung cancer. Cell Physiol Biochem. 31:56–65. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Xia H, Zhang W, Li Y, Guo N and Yu C: EZH2 silencing with RNA interference induces G2/M arrest in human lung cancer cells in vitro. Biomed Res Int. 2014:3487282014. View Article : Google Scholar : PubMed/NCBI

44 

Wang X, Zhao H, Lv L, Bao L, Wang X and Han S: Prognostic significance of EZH2 expression in non-small cell lung cancer: A meta-analysis. Sci Rep. 6:192392016. View Article : Google Scholar : PubMed/NCBI

45 

Zhang C, Ge S, Hu C, Yang N and Zhang J: miRNA-218, a new regulator of HMGB1, suppresses cell migration and invasion in non-small cell lung cancer. Acta Biochim Biophys Sin (Shanghai). 45:1055–1061. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Shen X, Hong L, Sun H, Shi M and Song Y: The expression of high-mobility group protein box 1 correlates with the progression of non-small cell lung cancer. Oncol Rep. 22:535–539. 2009.PubMed/NCBI

47 

Zheng W, Jiang C and Li R: Integrin and gene network analysis reveals that ITGA5 and ITGB1 are prognostic in non-small-cell lung cancer. Onco Targets Ther. 9:2317–2327. 2016. View Article : Google Scholar : PubMed/NCBI

48 

Jono H and Ando Y: Midkine: A novel prognostic biomarker for cancer. Cancers. 2:624–641. 2010. View Article : Google Scholar : PubMed/NCBI

49 

Yuan K, Chen Z, Li W, Gao CE, Li G, Guo G, Yang Y, Ai Y, Wu L and Zhang M: MDK protein overexpression correlates with the malignant status and prognosis of non-small cell lung cancer. Arch Med Res. 46:635–641. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Hao H, Maeda Y, Fukazawa T, Yamatsuji T, Takaoka M, Bao XH, Matsuoka J, Okui T, Shimo T, Takigawa N, et al: Inhibition of the growth factor MDK/midkine by a novel small molecule compound to treat non-small cell lung cancer. PLoS One. 8:e710932013. View Article : Google Scholar : PubMed/NCBI

51 

Regala RP, Davis RK, Kunz A, Khoor A, Leitges M and Fields AP: Atypical protein kinase C{iota} is required for bronchioalveolar stem cell expansion and lung tumorigenesis. Cancer Res. 69:7603–7611. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Sutterlüty H, Mayer CE, Attems J, Setinek U, Mikula M, Mikulits W, Micksche M and Berger W: Inhibition of cell migration and proliferation in non-small cell lung cancer (NSCLC) by Sprouty 2 (Spry2) via K-Ras dependent and independent pathways. Cancer Res. 66 Suppl 8:S349–S350. 2006.

53 

Sutterlüty H, Mayer CE, Setinek U, Attems J, Ovtcharov S, Mikula M, Mikulits W, Micksche M and Berger W: Down-regulation of Sprouty2 in non-small cell lung cancer contributes to tumor malignancy via extracellular signal-regulated kinase pathway-dependent and -independent mechanisms. Mol Cancer Res. 5:509–520. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2018
Volume 17 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang L, Sun L, Wang W, Xu H, Li Y, Zhao JY, Liu DZ, Wang F and Zhang LY: Construction of a 26‑feature gene support vector machine classifier for smoking and non‑smoking lung adenocarcinoma sample classification. Mol Med Rep 17: 3005-3013, 2018
APA
Yang, L., Sun, L., Wang, W., Xu, H., Li, Y., Zhao, J. ... Zhang, L. (2018). Construction of a 26‑feature gene support vector machine classifier for smoking and non‑smoking lung adenocarcinoma sample classification. Molecular Medicine Reports, 17, 3005-3013. https://doi.org/10.3892/mmr.2017.8220
MLA
Yang, L., Sun, L., Wang, W., Xu, H., Li, Y., Zhao, J., Liu, D., Wang, F., Zhang, L."Construction of a 26‑feature gene support vector machine classifier for smoking and non‑smoking lung adenocarcinoma sample classification". Molecular Medicine Reports 17.2 (2018): 3005-3013.
Chicago
Yang, L., Sun, L., Wang, W., Xu, H., Li, Y., Zhao, J., Liu, D., Wang, F., Zhang, L."Construction of a 26‑feature gene support vector machine classifier for smoking and non‑smoking lung adenocarcinoma sample classification". Molecular Medicine Reports 17, no. 2 (2018): 3005-3013. https://doi.org/10.3892/mmr.2017.8220