Identification of seven‐gene marker to predict the survival of patients with lung adenocarcinoma using integrated multi‐omics data analysis

1 INTRODUCTION

Lung cancer is a major cause of cancer-associated death worldwide,1 and risk factors cover family history, age, smoking, and air pollution.2 Two histological types of lung cancer are small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC).3, 4 Lung adenocarcinoma (LUAD) is the main type of NSCLC,5 and its incidence has been increasing steadily in the past few decades. Tumor, lymph node, and metastasis (TNM) phase is currently the most widely used system to assess the clinical outcome of cases with LUAD.6 However, the prognosis of LUAD patients with the same pathologic stage varies greatly.7-10 Currently, an effective system for predicting the outcome of LUAD is needed to help clinicians evaluate treatment outcomes and promote personalized therapy.

A number of studies were conducted for finding biomarkers predictive of long-term LUAD survival. Biological markers are mainly categorized into several classes, firstly, single molecule as a separated prognosis-related index, covering squamous cell antigen (SCC), CA125, or other new markers; secondly, many genetic markers as prognostic genes identified using gene expression profiles with high-throughput screening. There are several systematic biological methods for identifying gene biomarkers associated with LUAD prognosis and constructing genetic features. For instance, Wang et al established a 4-gene signature based on gene expression profiling using least absolute shrinkage and selection operator (LASSO) selection operator Cox regression and weighted gene correlation network analysis11; Liu et al. used a GLYCOLYSIS-related genes to identify a 4-gene signature applying Cox proportional hazard regression12; Chen et al. built a 11-gene signature by meta-analysis of gene expressions13; Li et al. developed a 3-gene signature using network biology method analysis.14 Noticeably, these above gene signatures have been independently verified in external datasets, but have not been clinically applied. Thus, it is crucial to analyze the biological function more comprehensively to identify genes associated with LUAD prognosis.

To effectively build a reliable LUAD prognostic process-associated gene signature, this study developed a scientific pipeline for identifying LUAD-associated gene markers. Single nucleotide mutations, gene expression profiles, and copy number variation data for LUAD patients were acquired from GEO and TCGA databases. A 7-gene signature was established by integrating transcriptomics and genomics data to screen prognostic markers. The ability of the signature to predict LUAD survival was validated in external validation and internal test sets. The results validated that the 7-gene signal participated in vital biology courses and pathways in LUAD. Similarly, GSEA analysis also showed consistent outcomes. The current data indicated that the 7-gene signature could efficiently estimate cancer prognostic of LUAD patients, providing a better understanding of the molecular mechanism of LUAD prognosis.

2 MATERIALS AND METHODS 2.1 Data collection and processing

A total of 516 samples containing SNP 6.0 chip copy number variation data, 576 FPKM samples containing RNA-Seq data, 738 samples containing clinical follow-up information were obtained from the UCSC Cancer Browser (https://xenabrowser.net/datapages/). 543 samples of mutation annotation information (MAF) were downloaded from GDC client. On May 25, 2019, the GSE3121015 dataset containing a total of 246 samples with standardized expression characteristics and clinic-related data was downloaded from GEO. In the GSE31210, 226 samples with clinical follow-up information were acquired. From the TCGA RNA-Seq data, follow-up information of 513 LUAD samples was filtered and randomly divided into 2 groups, one group as a training set (N = 256) and another as a test set (N = 257). All these samples were surgical cases collected before the first treatment. Samples with follow-up information in the GSE31210 dataset served as external validation sets. Copy number variation (CNV) dataset, GSE36363 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE36363), mutation dataset, and LUSC-KR (https://dcc.icgc.org/projects/LUSC-KR) were used as multi-omics validation datasets. For sample information of each group, see Table 1. The flow chart of this research is shown in Figure 1.

TABLE 1. Clinical information statistics of three sets of datasets Characteristic TCGA training datasets (n = 256) TCGA test datasets (n = 257) GSE31210 (n = 226) Age(years) <=50 29 19 27 >50 227 236 199 Survival Status Living 161 167 191 Dead 95 90 35 Gender Female 135 141 121 Male 121 116 105 Pathologic_T T 1 82 89 T 2 141 134 T 3 24 22 T 4 7 11 Pathologic_N N 0 166 170 N 1 53 41 N 2 31 38 N 3 1 1 Pathologic_M M 0 174 169 M 1/M X 80 86 Tumor Stage Stage Ⅰ 140 140 168 Stage Ⅱ 66 54 58 Stage Ⅲ 38 42 0 Stage Ⅳ 8 17 0 image

Work flow chart

2.2 Univariate Cox proportional hazards regression

To identify genes closely related to OS from the train dataset, we performed univariate Cox proportional hazard regression study as previously described in Jin-Cheng et al.16 p < 0.05 was the threshold.

2.3 Data analysis on copy number variation

GISTIC detects both focal and broad (probably overlapping) reappearing events. We used GISTIC 2.017 software to determine genes with significant amplification or deletion. The parameter thresholds for fragments with amplification or deletion lengths were ˃ 0.1 and p < 0.05.

2.4 Analysis of gene mutation

Significantly mutated genes in the MAF file of TCGA mutation data were screened by Mutsig 2.0 software. The threshold was set to p < 0.05.

2.5 Development of a prognostic immune gene signature Genes significantly related to patient OS, amplification, deletions, and mutations were selected, and those showing prognostic significance were obtained applying randomized survival forest algorithm.18 According to Jin et al,19 random survival forest in R package was used for filtering genes, iterations number of Monte Carlo was 100, and the number of previous progressions was 5. Characteristic genes were defined as genes exhibiting relative significance higher than 0.4. Further Cox regression study based on multiple variables was performed, and the following risk scoring mode was built: urn:x-wiley:08878013:media:jcla24190:jcla24190-math-0001In which N denotes quantity of prognosis-related genes, urn:x-wiley:08878013:media:jcla24190:jcla24190-math-0002 refers to the expression level of prognosis-related genes, and urn:x-wiley:08878013:media:jcla24190:jcla24190-math-0003 represents the assessed gene regression coefficient in the multivariate Cox regression study. 2.6 Analysis of functional enrichment

Pathway enrichment analysis of Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) was performed in the R package clusterprofiler,20 identifies over-represented GO terms in biology-related courses, cellular element and molecular function, and KEGG pathway. Here, in the study, FDR <0.05 represented statistics-related significance.

GSEA21 was conducted using the JAVA program (http://software.broadinstitute.org/gsea/downloads.jsp) with the MSigDB22 on C2 canonical pathway gene set collection containing 1320 gene sets. After conducting 1000 permutations, gene sets of a p < 0.05 showing noticeable up-regulation were filtered.

2.7 Statistical analysis

For comparing survival risk between the two risk groups, the Kaplan-Meier (KM) curve was drawn using the score of median risk as a cutoff. For testing whether genetic markers were independent prognostic factor, a multivariate Cox regression study was performed. Statistical significance was determined when p < 0.05. AUC study was carried out in the R package pROC. The heat map was drawn using R package Pheatmap. Default parameter was used in all analyses in R software version 3.4.3, unless otherwise specified.

3 RESULTS 3.1 Identification of gene sets in correlation with survival of LUAD cases

For samples of the TCGA train set, the study performed univariate regression to examine the relation between gene expression and patients’ overall survival (OS). Among the 2071 prognostic genes identified, 1161 genes were of HR<1, while 910 genes were of HR>1. The top 20 genes were listed in Table 2.

TABLE 2. The top20 most relevant genes for OS gene HR coefficient Z-score p value ENSG00000107859 1.48694912 0.39672645 5.72186084 1.05E−08 ENSG00000148704 1.434454599 0.360784707 5.57425514 2.49E−08 ENSG00000107984 1.533873881 0.427796484 4.962716871 6.95E−07 ENSG00000138829 1.422130151 0.352155854 4.647037727 3.37E−06 ENSG00000156687 1.46213808 0.379899803 4.635544845 3.56E−06 ENSG00000140478 1.425955115 0.354841845 4.561167459 5.09E−06 ENSG00000178462 1.400199512 0.336614735 4.294593782 1.75E−05 ENSG00000185888 1.376019257 0.319194734 4.263138276 2.02E−05 ENSG00000163975 1.531203385 0.426053952 4.23019025 2.33E−05 ENSG00000159217 1.353621993 0.302783958 4.06409894 4.82E−05 ENSG00000165891 1.456027917 0.375712123 4.011611337 6.03E−05 ENSG00000133466 1.506641141 0.409882763 4.007282786 6.14E−05 ENSG00000266265 1.354880774 0.303713461 3.976929914 6.98E−05 ENSG00000106031 1.355842036 0.30442269 3.962817439 7.41E−05 ENSG00000161714 1.454645204 0.374762025 3.892559471 9.92E−05 ENSG00000251258 1.325733251 0.281965703 3.885360438 0.000102178 ENSG00000121691 0.659952564 −0.415587319 −3.877219552 0.000105657 ENSG00000179241 1.462860113 0.380393501 3.8721129 0.000107896 ENSG00000145192 1.33684358 0.290311298 3.865936536 0.000110664 ENSG00000197213 1.315374686 0.274121558 3.816051598 0.000135604 3.2 Genomic variation identification with gene set

Genes showing significant deletion or amplification were identified by GISTIC 2.0 using copy number variation data from TCGA. There were 20 significantly amplified fragments in the genome of 230 genes (Figure 2A); noticeably, these fragments involved many important genes such as significant amplification of KRAS in the 12p12.1 segment (q value = 2.42E-12), significant amplification of EGFR in the 7p11.2 segment (q value = 2.16E-09), and significant expansion of ERBB2 in the 17q12 segment (q value = 2.01E-05). Moreover, there were 21 significant deletion fragments (Figure 2B) involving 1878 genes, and among these genes, CD1 showed significant absence in the 1p13.2 segment (q value = 2.63E-05), APC had significant deletion in the5q13 segment (q value = 0.017957), and CDKN2B also showed a great deletion in the 9p21.3 segment (q value = 1.08E-81). Mutsig2 was used to screen significantly mutated genes using TCGA mutational annotation data; here, a total of 438 genes with significant mutation frequencies were detected. Figure 2C lists the top 50 genes in the sample showing the most significant framework insertions or deletions, missense mutations, synonymous mutations, nonsense mutations, framework shifts, other nonsynonymous, or cleavage sites. Among the 50 genes, KRAS, RB1, SMAD4, TP53, EGFR, and BRAF were closely involved in the LUAD development.

image

mRNAs located in the focal CNA peaks are LUAD-related. Scores and false-discovery rates (q values) from GISTIC 2.0 for alterations (x-axis) plotted against genome positions (y-axis); dotted lines indicate the centromeres. A: The amplifications (red) of genes. B: The deletions (blue) of genes. The green line represents 0.25 q value cutoff point that determines significance. C: The most significant mutation in the top 50 genes, the upper histogram shows the total number of nonsynonymous and synonymous mutations in each of these 50 genes, the histogram on the right shows the number of samples in which 50 genes have mutated in all samples. Different colors in the heat map indicate the type of mutation, and the gray color means no mutation

3.3 Functional analysis on genomic variant genes

For investigating the functions of genes with genomic variations, 2261 deleted or amplified genes and significant mutant genes screened based on copy number variation were integrated. GO biological process and KEGG functional up-regulation study was conducted on the 2261 genes. KEGG enrichment analysis revealed that natural killer cell-mediated cytotoxicity, t-cell receptor signaling pathway, MAPK signaling pathway, chemokine signaling pathway, Foxo signaling pathway, other KEGG biological pathways, and non-small-cell lung cancer were related to the cancer development (Figure 3A). In the category of biological process, the pathways were mainly enriched in metabolic process, cell communication, cell differentiation, developmental process, and other GO terms (Figure 3B). Interestingly, these above terms were closely correlated with cancer progress. Our data indicated that the genes of these genomic variants were linked to tumor development.

image

Functional enrichment analysis on 2261 genomic variant genes. A: Enriched KEGG biological pathways. B: Enriched GO terms in the “biological process” category. Different colors indicate different saliency, and different sizes reflect the number of genes

3.4 A 7-gene signature for LUAD survival was built Genomic variant genes and prognosis-related genes were integrated, and a sum of 218 genes in the intersection of the three groups was selected as a candidate gene. Based on the relation of the quantity of classification trees and error rate, random forest was used in feature selection (Figure 4A). Here, genes with a significance greater than 0.4 were recruited to build a gene signature, and here, 7 genes were acquired (Table 3). The 7 genes were ranked for their out-of-bag value (Figure 4B). The 7-gene signature was built with Cox regression analysis based on multiple variables as follow: urn:x-wiley:08878013:media:jcla24190:jcla24190-math-0004 image

Establishment of a 7-gene signature for LUAD survival. A: The relationship between the number of classification trees and the error rate. B: The order of importance of 7 genes out-of-bag. C: KM survival curve distribution of the 7-gene signature in the TCGA training set. D: The ROC curve and AUC of the 7-gene signature classification. E: The risk score, survival status and survival time, and the expressions of the 7 genes of the TCGA training set

TABLE 3. 7-genes was significantly associated with the overall survival in the training-set patients Ensembl Gene ID Symbol HR Z-score p value Importance Relative Importance ENSG00000168273 SMIM4 0.78 −2.208803 2.72E−02 0.0119 1 ENSG00000104140 RHOV 1.30 2.641546 8.25E−03 0.009 0.755 ENSG00000083123 BCKDHB 0.78 −2.290767 2.20E−02 0.0087 0.7351 ENSG00000164796 CSMD3 1.33 3.658676 2.54E−04 0.0085 0.7152 ENSG00000214013 GANC 0.73 −2.486935 1.29E−02 0.0077 0.649 ENSG00000138829 FBN2 1.42 4.647038 3.37E−06 0.0056 0.4702 ENSG00000254585 MAGEL2 1.20 2.275242 2.29E−02 0.0051 0.4305

The sample risk score was obtained, and the samples were divided according to the medium the risk score (cutoff = −0.04131651). Patients’ prognosis in the two risk groups was significantly different (Figure 4C). Our result showed that the 3-year AUC of the model was 0.71 in the training set (Figure 4D). Analysis on expression correlation of the 7 genes and risk score showed that high expression and high-risk correlation of RHOV, CSMD3, FBN2, and MAGEL2 were risk factors, whereas low-risk correlation of SMIM4, BCKDHB, GANC, and high expression was protective factors (Figure 4E).

3.5 The robustness of the 7-gene signature was verified

To verify the robustness of the 7-gene signature, the sample risk score in the test set was calculated. According to the threshold of the training set, we divided the samples into two groups, and noticeable prognostic diversifications between the 2 groups were observed (Figure 5A). Analysis of ROC showed a 5-year AUC of 0.68 (Figure 5B). The risk score and the expression of the 7 genes were consistent in the training set (Figure 5C). Thus, the model was validated as an effective prognostic classifier in the TCGA dataset.

image

Robustness of the 7-gene signal model was verified in test dataset. A: KM curve in the test set sample. B: The ROC curve and AUC of the 7-gene signature classified in the test dataset. C: The relation between risk scores and the expressions of the 7 genes in the test set samples

GEO platform was an external dataset for verifying the classification of the gene prediction system in different platforms. The signature was applied to determine sample risk score. The cutoff of the training set was the threshold for grouping samples into low- and high-risk groups. We found that patients’ prognosis was evidently better in the low-risk group than the high-risk group (Figure 6A). Moreover, ROC analysis showed a 3-year AUC of 0.66, which was similar to the training set (Figure 6B), and the relationship between the risk score and the expressions of the 7 genes was also consistent in the training set (Figure 6C). To conclude, our 7-gene signature model showed a prognostic significance in both external and internal datasets.

image Robustness of the 7-gene signal model was verified in GSE31210 dataset. A: KM survival curve distribution of the 7-gene signature in GSE31210. B: ROC curves and AUC of the 7-gene signature classification. C: The relationship of expressions of the 7 genes, survival time, risk score, and survival status in GSE31210 3.6 The 7-gene signature model was clinically independent

To identify whether the 7-gene signature system was independent in clinical applications, the 95% confident interval (CI) of HR, relevant hazard ratio (HR), and p value were determined by performing multivariate and univariate Cox regression in the TCGA test set, TCGA training set, and GSE31210 data with clinical information. We then analyzed the clinical information of TCGA, GSE65858 patients’ data covering tumor stage N stage, M stage, T stage, gender, age, and group information using our 7-gene signature (Table 4). Analysis from univariate Cox regression showed that in the TCGA training set, high-risk group, pathologic T3, pathologic N1, pathologic T2, pathologic N2/N3, tumor stage II, tumor stage III, tumor stage IV displayed noticeable associations with survival. However, from multivariate Cox regression analysis, only high-risk group (p = 3.40E-05, CI = 1.52–3.27, HR = 2.24, 95%) was clinically independent. In the TCGA test set, from the results of univariate Cox regression analysis, we observed that pathologic N1, pathologic T4, pathologic T3, pathologic N2/N3, tumor stage II, tumor stage IV, tumor stage III, and high-risk group were evidently related to survival. Multivariate Cox regression demonstrated that pathologic T3, pathologic N1, and high-risk group (p = 0.002, 95% CI = 1.25–2.79, HR = 1.86) were clinically independent. In GSE65858, from the data of univariate Cox regression analysis, it could be found that tumor stage II and high-risk group were noticeably correlated with survival. Corresponding multivariate Cox regression study demonstrated that high-risk group (p = 0.008, CI = 1.17–2.91, HR = 1.85, 95%) and tumor stage II were clinically independent. Hence, the 7-gene prediction system was a prognostic marker independent of other clinical factors.

TABLE 4. Univariate and multivariate Cox regression analysis identifies clinical factors and clinical independence associated with prognosis Variables Univariate analysis Multivariable analysis HR 95%CI of HR p value HR 95%CI of HR p value TCGA training datasets 7-gene risk score Low-risk group 1 (reference) 1 (reference) High-risk group 2.72 2.01–3.68 9.530E−11 2.24 1.52–3.27 3.40E−05 Age 1.01 0.99–1.03 0.606 1.01 0.99–1.03 0.277 Female 1 (reference) 1 (reference) Male 0.84 0.56–1.26 0.40 0.85 0.54–1.31 0.468 Pathologic T 1 1 (reference) 1 (reference) Pathologic T 2 1.73 1.02–2.93 0.042 1.36 0.77–2.37 0.286 Pathologic T 3 2.99 1.39–6.38 4.86E−03 1.56 0.59–4.11 0.370 Pathologic T 4 1.78 0.52–6.08 3.56E−01 0.83 0.18–3.77 0.808

留言 (0)

沒有登入
gif