Open Access

Prognostic prediction of a 12‑methylation gene‑based risk score system on pancreatic adenocarcinoma

  • Authors:
    • Daoqin Dou
    • Shaohua Yang
    • Jiren Zhang
  • View Affiliations

  • Published online on: April 27, 2020     https://doi.org/10.3892/ol.2020.11575
  • Pages: 85-98
  • Copyright: © Dou 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

Pancreatic adenocarcinoma (PAAD) accounts for ~85% of all pancreatic cancer cases and is associated with a less favorable prognosis. Aberrant DNA methylation may influence the progression of PAAD by inducing abnormal gene expression. Methylation data of PAAD samples with prognosis information were obtained from The Cancer Genome Atlas (training set) and European Bioinformatics Institute Array Express databases (validation sets). Using the limma package, the differentially methylated genes in the training dataset were screened. Combined with the Weighted Gene Co‑expression Network Analysis package, the co‑methylated genes in key modules were identified. Then, a cor.test function in R software was applied to explore the functions of key the methylated genes. Correlation analyses of the expression levels and methylation levels of key methylated genes were performed, followed by identification of methylated genes associated with prognosis using Univariate Cox regression analysis. The optimal combination of prognosis related methylated genes was determined using a Cox‑Proportional Hazards (Cox‑PH) model. Subsequently, the risk score prognostic prediction system was constructed by combining the Cox‑PH prognosis coefficients of the selected optimized genes. Based on the constructed risk score system, samples in all datasets were divided into high and low risk samples and the survival status was compared using survival curves. Furthermore, the correlation between independent prognostic factors and the risk score system was determined using the survival package. A total of 50 genes associated with prognosis of PAAD and a 12‑gene optimal combination were obtained, including: CCAAT/enhancer binding protein α, histone cluster 1 H4E, STAM binding protein‑like 1, phospholipase D3, centrosomal protein 55, ssDNA binding protein 4, glutamate AMPA receptor subunit 1, switch‑associated protein 70, adenylate‑cyclase activating polypeptide 1 receptor 1, yippee‑like 3, homeobox C4 and insulin‑like growth factor binding protein 1. Subsequently, a risk score prognostic prediction system of these 12 genes was constructed and validated. In addition, pathological N category, radiotherapy and risk status were identified as independent prognostic factors. Overall, the risk score prognostic prediction system constructed in the present study may be effective for predicting the prognosis of patients with PAAD.

Introduction

Pancreatic cancer (PC) is primarily associated with diabetes, obesity, smoking and genetic conditions, such as germline pathogenic variants and somatic pathogenic variants in DNA damage repair (DDR) genes (1,2). Jaundice, weight reduction, back or abdominal pain, deep-colored urine, pale-colored stools and anorexia are the typical symptoms (3). However, at diagnosis, the cancer has usually metastasized (4,5). The mortality rate of PC is high and there were 411,600 PC-associated deaths globally in 2015 (6). Pancreatic adenocarcinoma (PAAD) is a common type of PC, accounting for ~85% of all PC cases (7). The survival rate of PAAD is very low and the 5-year survival rate was 5% in 2015 (8). A few prognostic indicators are now available for PC, such as C-reactive protein/albumin ratio and neutrophil/lymphocytes ratio (9,10). Therefore, it is of great importance to investigate the prognostic factors of PAAD for improved prediction.

It has been hypothesized that DNA methylation may provide a link between environmental factors contributing to cancer development (11). The stability of the genome and gene expression levels are primarily maintained by a pre-determined pattern of DNA methylation (12). It has been reported that DNA hypermethylation has prognostic value and acts as independent predictor of survival in other cancer types, such as head and neck cancer (13). A previous study reported an association between abnormal methylation of the Reprimo gene and genetic instability and poor survival following surgical resection in patients with PC (14). Hypermethylation of Cyclin D2 is also frequently observed in PC (15). Meanwhile, another study reported a significant difference in the hypermethylation frequency of ALX4, BNC1, HIC1, SEPT9V2,SST, TFPI2 and TAC1 between PAAD samples of stage I, II, III and IV, and these genes are significantly associated with distant metastasis of PAAD (16).

A number of clinical markers of PAAD survival have been recognized, including stage at diagnosis, grading and performance status and the treatment received, such as resection versus no resection and chemotherapy vs. no chemotherapy (1720). It has also been reported that obesity and smoking are associated with a less favorable prognosis of PC (21). Cigarette smoking is associated with the development of ~20% of PC cases and is therefore a consistent risk factor (22). Bioinformatic databases may serve as a valuable tool to further our understanding of the molecular mechanisms underpinning the prognosis of patients with cancer.

The present study aimed to explore the aberrant methylation of genes associated with prognosis of patients with PAAD. The methylation data of genes associated with PAAD were obtained from The Cancer Genome Atlas (TCGA) database and screened for differentially methylated genes (DMGs) associated with prognosis. Subsequently, these data were used to construct a risk score system, which may be effective in predicting the prognosis of patients with PAAD.

Materials and methods

Datasets

The methylation data for the training dataset was obtained from the TGCA database (accessed on 5th June 2018; cancer.gov/tcga), which were based on the Illumina Infinium Human Methylation 450 BeadChip platform. There were a total of 184 samples, 168 of which included prognostic information [mean age, 64.89±11.24 years (range: 40–88); male: female, 94/75; average overall survival time, 17.09±15.22 months; death: survival, 88/80]. The methylation data for the validation training set was obtained from the European Bioinformatics Institute ArrayExpress database (ebi.ac.uk/arrayexpress/), specifically the E-MTAB-5008 and E-MTAB-5571 datasets. The E-MTAB-5008 dataset consisted of 29 PAAD samples with prognostic information and the E-MTAB-5571 dataset consisted of 24 PAAD samples which prognostic information. Both of these datasets were sequenced on the platform of Illumina Infinium Human Methylation 450 BeadChip.

Screening of DMGs

To screen genes associated with PAAD prognosis, samples in the TCGA dataset were divided into less favorable prognosis (defined as a survival time <6 months and death) and more favorable prognosis (defined as survival time >24 months or alive) groups based on a previously described grouping method (23). The methylation loci of genes associated with PAAD prognosis were annotated and combined with the platform annotation information on the Illumina Infinium Human Methylation 450 BeadChip platform and the loci within CpG islands of the genes were selected and used for the following analysis. Using the limma package in R (version 3.34.7) (24), the DMGs between the less favorable prognosis and more favorable prognosis groups were screened according to the following criteria: |log fold change| >0.263 and false discovery rate (FDR) <0.05. Then, the Kernel density curve of the DMGs was generated by calculating the Log 2 (FC).

Identification of co-methylated genes based on Weighted Gene Co-expression Network Analysis (WGCNA)

Co-methylation analysis using the WGCNA package (version 1.63) (25) in R was performed on genes located in CpG islands to identify differentially methylated CpG genes (DMCpGs). The sets of CpG genes with highly related methylation levels under the same biological process or in different tissues were considered as modules. The modules which had a significant association with the methylation levels were identified. The DMCpGs were mapped to the modules and the significant enrichment parameters and fold enrichment were calculated using a hypergeometric test (26). The DMCpGs enriched modules were screened under the following criteria: P<0.05 and a fold enrichment value of >1 was considered to indicate a statistically significant difference. Genes in DMCpGs enriched modules were recognized as key methylation genes and were analyzed using Gene Ontology (GO) enrichment analysis (27) using the Database for Annotation, Visualization and Integrated Discovery (version 6.8) (28).

To meet scale-free network distribution, the weighting parameter ‘power’ in WGCNA algorithm was explored. When the square of the correlation coefficient between log(k) and log[p(k)] reached 0.9, the corresponding value of parameter ‘power’ (power=7) was selected. Under power=7, the mean connectivity of genes was calculated to be 1. Subsequently, the adjacency matrix elements were serialized, and the topological overlap matrix was calculated to evaluate the correlation of gene methylation levels and obtain a system clustering tree. According to the standards of hybrid dynamic shear tree, pruning height (cutHeight) and the minimum number of module genes (minSize) separately were set as 0.95 and 50.

Correlation analysis for the expression levels and methylation levels of key methylated genes

The expression and methylation levels of key methylation genes in matched training PAAD samples were collected and correlation analysis was performed. Pearson correlation coefficients (PCCs) were calculated and the cor.test function (stat.ethz.ch/R-manual/R-devel/library/stats/html/cor.test.html) in R Software was used (29,30). PCCs of single genes were also calculated and the genes with a negative correlation between expression level and methylation level were selected as key methylation genes for subsequent analyses. P<0.05 was considered to indicate a statistically significant difference.

Identification of the methylated genes associated with prognosis

Key methylation genes with a negative correlation between expression level and methylation level were further analyzed for prognosis associated genes. Univariate Cox regression analysis was performed to identify prognosis associated methylation genes using the survival package (version 2.41–1) in R (31). P<0.05 was considered to indicate a statistically significant difference.

Construction of risk score prognostic prediction system

The optimal combination of prognosis related methylation genes was screened using the Cox-Proportional Hazards (Cox-PH) model using the penalized package (version 0.9–50) in R (32). The optimal parameter of ‘lambda’ in the Cox-PH model was calculated through 1,000 cross-validation likelihoods (cvl) (33).

The risk score prognostic prediction system was constructed combining the Cox-PH prognosis coefficients and methylation levels of the selected optimized genes. The resultant formula was: Risk score=∑coef gene × Methylation gene where Coefgene and Methylationgene represented regression coefficient and gene methylation levels, respectively.

The risk scores of samples in the TCGA, E-MTAB-5008 and E-MTAB-5571 datasets were calculated and stratified into high and low risk groups according to the median risk scores. Kaplan-Meier (KM) survival curves (34) of the high and low samples were plotted using the survival package, which were compared with the prognosis of all samples. The area under the receiver operating characteristic (ROC) curve (AUC) was compared, also using the survival package.

Correlation analysis between independent prognostic factors and risk score prognostic prediction system

Using the Cox regression analysis in the survival package (31), independent clinical prognostic factors were screened. Next, the relations between collected factors and the risk score prognostic prediction system were analyzed using KM curves.

Results

Screening of DMGs

Median survival time of samples in the training datasets was 17.09±15.22 months, which is consistent with the time reported in PC (35). There were 13,903 methylation sites containing CpG islands in the training dataset. In TCGA training dataset, based on the predefined method for grouping, each less favorable and more favorable prognosis group had 19 samples, and a total of 1,067 DMGs between the two groups were identified (Fig. 1).

As shown in the Log2 Kernel density curve of the DMGs, 74.98% (800/1,067) of DMGs were significantly hypomethylated and 25.02% (267/1,067) were significantly hypermethylated in the good prognostic group (Fig. 1B). The cluster heatmap of the DMGs suggested that the samples with different prognoses in the TCGA dataset exhibited different methylation levels (Fig. 1C). Furthermore, as the feature factors had different weights in the calculation process for heatmap analysis, there was a slight crossover between good and bad prognosis samples.

Among the CpGs in the 1,067 DMGs, 309, 321, 118, 44, 185 and 90 CpGs were separately located in transcription start site areas, body areas, 5′untranslated regions (UTR), 3′UTR regions, promoter regions and the first exon regions (data not shown). The top 20 DMGs with smaller FDR values were screened and presented in Table I.

Table I.

Top 20 differentially methylated genes with significance.

Table I.

Top 20 differentially methylated genes with significance.

Methylation lociChr.PositionGenesLocationβ-badβ-goodEffectP-valueFDR
cg24902557chr14104786509BTBD6Promoter0.32770.0926−1.8239 5.24×10−7a 4.25×10−6
cg27106909chr1630014398YPEL3Promoter0.31500.1325−1.2489 8.88×10−7a 7.20×10−6
cg25207224chr634319167HMGA1Body0.56450.75470.4188 5.80×10−6a 4.70×10−5
cg27543607chr1688289602CDK103′UTR0.68780.5288−0.3791 6.59×10−6a 5.34×10−5
cg27058889chr155237413BSND1stExon0.54410.75740.4772 6.71×10−6a 5.44×10−5
cg21197594chr9139776802EHMT1Body0.72380.4881−0.5683 9.65×10−6a 7.82×10−5
cg26789779chr537875700GDNFTSS2000.35980.1482−1.2801 9.92×10−6a 8.04×10−5
cg23066982chr626312442 HIST1H4EPromoter0.24460.0759−1.6886 1.12×10−5a 9.08×10−5
cg27575890chr2235237104EIF3DBody0.35920.2527−0.5075 1.16×10−5a 9.44×10−5
cg24864161chr2248870409MOV10L1TSS2000.20220.0689−1.5531 1.21×10−5a 9.84×10−5
cg24460144chr5149550073SLC6A75′UTR0.30930.1822−0.7637 1.43×10−5a 1.16×10−4
cg27576241chr1153301194ADAM15Body0.55300.4002−0.4667 1.50×10−5a 1.21×10−4
cg23930825chr643721343RSPH9Body0.35020.2102−0.7365 1.74×10−5a 1.41×10−4
cg26432961chr641625040FOXP45′UTR0.78970.5080−0.6365 1.79×10−5a 1.45×10−4
cg27626790chr7150305686KCNH2Body0.26270.1002−1.3909 1.80×10−5a 1.45×10−4
cg09730123chr161767949SPSB3Body0.41730.3468−0.2670 1.81×10−5a 1.47×10−4
cg25499067chr823615933NKX2-6Body0.23100.1187−0.9606 1.97×10−5a 1.60×10−4
cg25902939chr1918405350SSBP4Body0.42020.2254−0.8986 2.01×10−5a 1.63×10−4
cg27390009chr1718091226FLIIBody0.50730.3645−0.4768 2.05×10−5a 1.66×10−4
cg24542766chr2238043478RPL3Body0.39240.2856−0.4580 2.10×10−5a 1.70×10−4

a P<0.001. cg indicates methylation ID of a gene; chr., chromosome; Position, position of the methylation sites in chromosomes; Location, location of CpG islands; β-bad, the average methylation level β in bad prognostic groups; β-good, the average methylation level β in good prognostic groups; Effect, log2 fold change; FDR, false discovery rate; TSS, transcription start site; UTR, untranslated region.

Identification of co-methylated genes based on WGCNA

A total of 10 modules were identified (Fig. 2A) and the detailed information of each module is listed in Table II. CpG island genes in 9 modules showed significant a association with methylation levels (P<0.05; correlation coefficients, 0.226–0.729; average correlation coefficient, 0.7742; Table II). The identified DMGs were mapped into each module and their distribution in the modules is shown in Fig. 2B. Two modules were identified as differentially expressed CpG gene enriched modules, black module (comprised of 90 DMGs) and the turquoise module (comprised of 394 DMGs), in which the CpG genes were significantly associated with methylations. The DMGs in these two modules were significantly enriched in 18 GO_Biology Process (BP; such as ‘neuron differentiation’), 7 GO_cellular component (CC; such as ‘integral to plasma membrane’), and 9 GO_molecular function (MF; such as ‘sequence-specific DNA binding’) terms (Table III) and were predominantly associated with transcriptional regulation.

Table II.

A total of 10 modules identified by weighted gene co-expression network analysis.

Table II.

A total of 10 modules identified by weighted gene co-expression network analysis.

ModulesCount of CpGsCorrelation PcorrDMGsEnrichment fold (95% CI) Phyper
Black1590.703 1.01×10−13903.481 (2.622–4.599) 2.20×10−16
Blue4270.587 1.68×10−4630.908 (0.676–1.202) 5.37×10−1
Brown3640.551 2.23×10−3100.169 (0.0799–0.317) 4.83×10−13
Green3520.616 6.05×10−2120.209 (0.107–0.374) 3.49×10−11
Grey5740.226 1.32×10−1480.514 (0.371–0.701) 5.31×10−6
Magenta900.729 9.47×10−16231.572 (0.941–2.529) 7.38×10−2
Pink1380.679 1.44×10−860.267 (0.0961–0.601) 2.88×10−4
Red1910.308 6.69×10−710.0322 (0.000814–0.182) 3.26×10−11
Turquoise13440.687 1.72×10−63941.803 (1.564–2.077) 4.11×10−16
Yellow3590.609 4.27×10−630.0514 (0.0105–0.152) 2.20×10−16

[i] DMGs, differentially methylated genes; CI, confidence interval; Pcorr, P-value for correlation; Phyper, P-value for hypermethylation.

Table III.

Functional terms enriched for the 484 differentially methylated genes involved in black or turquoise modules.

Table III.

Functional terms enriched for the 484 differentially methylated genes involved in black or turquoise modules.

A, Biological process

TermCountP-valueFDR
GO:0030182-neuron differentiation40 1.79×10−10a 3.13×10−7
GO:0006355-regulation of transcription, DNA-dependent95 4.27×10−10a 7.45×10−7
GO:0051252-regulation of RNA metabolic process96 6.29×10−10a 1.10×10−6
GO:0007423-sensory organ development27 1.46×10−9a 2.55×10−6
GO:0007389-pattern specification process28 8.94×10−9a 1.56×10−5
GO:0045165-cell fate commitment20 1.20×10−8a 2.10×10−5
GO:0048568-embryonic organ development22 1.59×10−8a 2.77×10−5
GO:0048598-embryonic morphogenesis28 1.66×10−7a 2.90×10−4
GO:0007267-cell-cell signaling40 9.79×10−7a 1.71×10−3
GO:0006928-cell motion34 1.66×10−6a 2.90×10−3
GO:0045449-regulation of transcription111 2.63×10−6a 4.60×10−3
GO:0003002-regionalization20 3.02×10−6a 5.27×10−3
GO:0007610-behavior33 3.54×10−6a 6.18×10−3
GO:0048666-neuron development27 3.76×10−6a 6.57×10−3
GO:0031328-positive regulation of cellular biosynthetic process42 4.11×10−6a 7.17×10−3
GO:0009891-positive regulation of biosynthetic process42 5.85×10−6a 1.02×10−2
GO:0016477-cell migration23 1.16×10−5a 2.02×10−2
GO:0019226-transmission of nerve impulse26 1.97×10−5a 3.45×10−2

B, Cellular component

TermCountP-ValueFDR

GO:0005887-integral to plasma membrane59 9.12×10−7a 1.21×10−3
GO:0031226~intrinsic to plasma membrane59 1.89×10−6a 2.50×10−3
GO:0044459-plasma membrane part90 2.93×10−6a 3.88×10−3
GO:0034703-cation channel complex15 6.78×10−6a 8.98×10−3
GO:0034705-potassium channel complex12 1.11×10−5a 1.48×10−2
GO:0008076-voltage-gated potassium channel complex12 1.11×10−5a 1.48×10−2
GO:0034702-ion channel complex18 1.99×10−5a 2.64×10−2

C, Molecular function

TermCountP-valueFDR

GO:0043565-sequence-specific DNA binding58 4.65×10−16a 6.55×10−13
GO:0003700-transcription factor activity74 4.42×10−15a 6.52×10−12
GO:0030528-transcription regulator activity85 3.53×10−10a 5.18×10−7
GO:0003677-DNA binding109 2.11×10−8a 3.09×10−5
GO:0022836-gated channel activity25 6.95×10−6a 1.02×10−2
GO:0005261-cation channel activity23 1.01×10−5a 1.48×10−2
GO:0015267-channel activity29 1.41×10−5a 2.07×10−2
GO:0022803-passive transmembrane transporter activity29 1.48×10−5a 2.18×10−2
GO:0005216-ion channel activity27 3.37×10−5a 4.94×10−2

a P<0.001. GO, gene ontology; FDR, false discovery rate.

Correlation analysis of the expression levels and methylation levels of key methylated genes

Overall, the methylation levels and expression levels of the 484 DMGs in the black and turquoise modules were significantly negatively correlated (Cor.=−0.478, P=8.169×10−5; Fig. 2D). A total of 192 DMGs exhibited negative correlation between the expression levels and methylation levels (Table SI).

Construction of the risk score system

A total of 50 genes among the 192 DMGs were found to be associated with prognosis. Following this, a Cox-PH model was utilized to screen the optimal gene combination. When λ=1.1389, the maximum value of cvl was obtained as −458.1914 (Fig. 3A). Using λ=1.1389, a 12-gene optimal combination was acquired: CCAAT/enhancer binding protein α (CEBPα); histone cluster 1 H4E (HIST1H4E); STAM binding protein-like 1, (STAMBPL1) phospholipase D3 (PLD3); centrosomal protein 55 (CEP55); ssDNA binding protein 4 (SSBP4); glutamate AMPA receptor subunit 1 (GRIA1); switch-associated protein 70 (SWAP70); adenylate-cyclase activating polypeptide 1 receptor 1 (ADCYAP1R1); yippee-like 3 (YPEL3); homeobox C4 (HOXC4); and insulin-like growth factor binding protein 1 (IGFBP1) (Fig. 3B; Table IV). Combined with the prognostic coefficients of these 12 optimal genes, the following risk score system was constructed (cg is the methylation ID for corresponding genes.).

Table IV.

Information of the 12 optimal genes.

Table IV.

Information of the 12 optimal genes.

Methylation IDGeneChr.PositionLocationCoefficientHazard ratioP-value
cg22250546CEBPAchr1938483210Promoter−0.47015590.197 2.01×10−2a
cg23066982 HIST1H4Echr626312442Promoter1.4610971.228 4.11×10−2a
cg23264429 STAMBPL1chr10906319835′UTR−0.15437610.369 4.53×10−2a
cg25509871PLD3chr19455633975′UTR0.21249211.082 4.40×10−2a
cg25827255CEP55chr1095246749Promoter−0.70635130.129 2.04×10−2a
cg25902939SSBP4chr1918405350Body0.12680351.139 4.45×10−2a
cg26343183GRIA1chr5152988914Body−2.46425260.276 1.33×10−2a
cg26645401SWAP70chr119643090Body−0.45836470.272 2.02×10−2a
cg27076139 ADCYAP1R1chr731058243TSS15000.06520141.014 1.35×10−2a
cg27106909YPEL3chr1630014398Promoter0.25669941.294 4.32×10−2a
cg27138204HOXC4chr12527323675′UTR0.77077551.679 1.58×10−2a
cg27447599IGFBP1chr745894465TSS2000.1724931.193 2.18×10−2a

a P<0.01. chr, chromosome; coef, correlation coefficient; UTR, untranslated region; TSS, transcription start site.

Risk score=(−0.4701559) × Methylation cg22250546 + (1.461097) × Methylation cg23066982 + (−0.1543761) × Methylation cg23264429 + (0.2124921) × Methylation cg25509871 + (−0.7063513) × Methylation cg25827255+ (0.1268035) × Methylation cg25902939 + (−2.4642526) × Methylation cg26343183+ (−0.4583647) × Methylation cg26645401 +(0.0652014) xMethylation cg27076139+ (0.2566994) × Methylation cg27106909 + (0.7707755) × Methylation cg27138204 + (0.172493) × Methylation cg27447599.

According to the median of the risk scores of the samples in the TCGA dataset, the samples were divided into high and low risk groups. For the TCGA dataset, the comparison between the actual overall survival and risk score system predicting survival of the risk groups was performed and the AUC was 0.976 (Fig. 4A). Moreover, the risk score system was validated in the E-MTAB-5008 (Fig. 4B) and E-MTAB-5571 (Fig. 4C) datasets and the AUCs were 0.919 and 0.924, respectively. The TCGA, E-MTAB-5008 and E-MTAB-5571 datasets had consistent results and all the samples in low risk group had improved survival.

Correlation analysis between independent prognostic factors and risk groups

The clinical information of 168 samples in the TCGA dataset was statistically analyzed, and pathological tumor-node-metastases (TNM) staging system (36), radiotherapy and risk status were identified as independent prognostic factors (Table V). Survival status of high and low risk groups with different pathological N stages (N0 vs. N1) and treatment with radiotherapy (treatment with radiotherapy vs. without radiotherapy) were compared (Fig. 5).

Table V.

The results for screening independent prognostic factors.

Table V.

The results for screening independent prognostic factors.

Univariate coxMultivariate cox


Clinical characteristicsTCGA, n=168HR95% CIP-valueHR95% CIP-value
Age, years, mean ± sd64.89±11.241.0291.008–1.049 5.83×10−3b1.0190.997–1.0413 9.46×10−2
Sex 0.9190.605–1.394 6.89×10−1UnknownUnknownUnknown
  Male94
  Female75
Pathologic M 1.4910.457–4.858 5.05×10−1UnknownUnknownUnknown
  M079
  M14
  Unknown85
Pathologic N 2.1561.276–3.643 3.31×10−3b2.121.139–3.947 1.19×10−2a
  N045
  N1119
  Unknown4
Pathologic T 1.7111.116–2.622 1.35×10−2a1.2210.576–2.585 6.03×10−1
  T18
  T219
  T3136
  T44
  Unknown1
Pathologic stage 1.4881.040 – 2.129 3.12×10−2a0.9960.473–2.096 9.92×10−1
  I20
  II138
  III5
  IV4
  Unknown1
Pathologic grade 1.5381.148–2.061 3.76×10−3b1.3660.993–1.879 5.56×10−2
  130
  288
  347
  42
  Unknown1
History of alcohol usage 1.0990.688–1.757 6.92×10−1UnknownUnknownUnknown
  Yes96
  No56
History of alcohol usage 1.0990.688–1.757 6.92×10−1UnknownUnknownUnknown
  Unknown16
Tobacco history 1.1170.874–1.427 3.75×10−1UnknownUnknownUnknown
  Never61
  Reform56
  Current19
  Unknown32
Chronic pancreatitis history 1.1010.524–2.314 7.99×10−1UnknownUnknownUnknown
  Yes117
  No13
  Unknown38
Diabetes history 0.9030.509–1.602 7.27×10−1UnknownUnknownUnknown
  Yes36
  No99
  Unknown33
Radiotherapy 0.5290.302–0.928 2.39×10−2a0.5020.279–0.906 3.78×10−2a
  Yes38
  No112
  Unknown18
Recurrence 1.5580.951–2.552 7.59×10−2UnknownUnknownUnknown
  Yes40
  No102
  Unknown26
Risk status 2.4531.578–3.815 3.93×10−5c2.1461.289–3.571 3.31×10−3b
  High84
  Low84
Dead UnknownUnknownUnknownUnknownUnknownUnknown
  Death88
  Alive80
Overall survival time, months, mean ± sd)17.09±15.22UnknownUnknownUnknownUnknownUnknownUnknown

a P<0.05

b P<0.01

c P<0.001. TCGA, The Cancer Genome Atlas; HR, Hazard Ratio; CI, confidence interval; sd, standard deviation; T, tumor stage; N, node stage; M, metastasis stage.

Discussion

The mechanisms underlying tumor development and progression of PAAD are complex, and influencing factors include epigenetic regulation of gene expression, epigenetic silencing of genes, oncogenic/tumor suppressor gene mutation, telomere alteration, genomic instability and DNA methylation (3740). The present study aimed to identify potential important key methylated genes in PAAD. A total of 1,067 DMGs were identified in the less favorable prognosis and more favorable prognosis groups. From the 10 modules identified by WGCNA, the black (involving 90 DMGs) and turquoise (involving 394 DMGs) modules, in which the CpG genes were significantly associated with methylations, were selected for further analysis. For the 484 DMGs involved in the two key modules, 18 GO_BP, 7 GO_CC and 9 GO_MF terms were enriched. There were 192 DMGs associated with prognosis (less favorable or more favorable). Correlation analysis indicated that the expression levels and methylation levels of 192 DMGs among the 484 DMGs were negatively correlated. Furthermore, 50 prognosis-associated genes were further screened from the 192 DMGs. After a 12-gene optimal combination (CEBPA, HIST1H4E, STAMBPL1, PLD3, CEP55, SSBP4, GRIA1, SWAP70, ADCYAP1R1, YPEL3, HOXC4 and IGFBP1) was identified, the risk score system was constructed and validated in the TCGA, E-MTAB-5008 and E-MTAB-5571 datasets. In addition, pathological N category, radiotherapy and risk status were found to be independent prognostic factors.

It is demonstrated that upregulation of CCAAT/enhancer binding protein (C/EBP) beta (C/EBPβ), encoded by CEBPB, could restore the anti-cancer functions of Menin in PC (41) The inadequate cytoplasmic localization and abnormal silencing of C/EBP results in its dysfunction, and thus, C/EBP may serve as a novel suppressor in PC cells (42). Downregulated C/EBPα induced by lysine (K)-specific demethylase 6B (KDM6B) promotes the aggressiveness of pancreatic ductal adenocarcinoma (PDAC) cells, indicating that the KDM6B-C/EBPα axis is associated with the progression of PDAC (43,44). Histone H3 modification affects the gene expression and promoter methylation of MUC2, which may be critical for the prognostic prediction of patients with PC (45). The mRNA expression levels of histone H4 is lowered by polyamide-chlorambucil conjugate (1R-Chl) in the MIA PaCa-2 PC cell line and histone H4 genes have elevated histone acetylation in tumor cells (46). Therefore, CEBPA and HIST1H4E may be critical for the survival of PAAD patients.

STAMBPL1 affects the activation of NF-κB through mediating the stability and localization of Tax (47) and NF-κB blockade can inhibit the oncogenicity and metastasis of PC cells (48). Overexpression of CEP55 can promote PC cell aggressiveness via activation of the NF-κB pathway; therefore, CEP55 may be a prognostic factor and therapeutic target for patients with PC (49). Downregulated expression of YPEL1 in PAAD samples is associated with perineural invasion and survival prognosis, thus YPEL1 may serve a role in the malignant transformation of pancreatic tissues (50). Low IGFBP1 plasma levels have a more notable influence in non-smoking patients with PC and predicts an increased risk of PC (51). These data suggest that STAMBPL1, CEP55, YPEL3 and IGFBP1 may be associated with the prognosis of patients with PAAD.

Although PLD3, SSBP4, GRIA1, SWAP70 and ADCYAP1R1 do not have reported associations with PAAD to the best of our knowledge, their influence on other types of human cancer have been reported. Elevated expression and activity of PLD is detected in multiple types of cancer, such as gastric, colorectal, renal, stomach, lung and breast cancers (52), and PLD serves a role in mediating cell proliferation, cell transition, survival signaling and tumor progression (53). SSBP2 is a tumor suppressor gene and the disruption of SSBP2-associated pathways may be involved in the malignant transformation of various tissues (54). GRIA1 is involved in glutamate receptor signaling, which is an epigenetic marker for overall mortality rate of basal-like urothelial carcinomas (55). The oncogene SWAP70 functions in regulating actin rearrangement in basal-like bladder cancer (55) and serves a role in the transformation-associated signaling pathway (56). The promoter hypermethylation level of ADCYAP1 is associated with cervical cancer development and is considered as a promising methylation marker for the early detection of cervical cancer (57). Therefore, PLD3, SSBP4, GRIA1, SWAP70 and ADCYAP1R1 may also be associated with the prognosis of patients with PAAD.

In the present study comprehensive bioinformatics analysis of PAAD samples was performed to identify prognosis-associated genes and to construct a risk score prognostic prediction system. All findings were obtained from relatively small-sized cohorts and thus require further experimental validation. Additionally, the patient cohort samples in the three datasets may exert different clinical features, such as disease stage, historical treatment and demographics, which should be carefully compared in further studies.

In conclusion, 1,067 DMGs were identified and a 12-gene optimal combination consisting of CEBPA, HIST1H4E, STAMBPL1, PLD3, CEP55, SSBP4, GRIA1, SWAP70, ADCYAP1R1, YPEL3, HOXC4 and IGFBP1 was obtained. This 12-gene risk score prognostic prediction system may be valuable for predicting the prognosis of patients with PAAD.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

DD conceived the study design and performed manuscript drafting, SY was responsible for data collection and analysis and JZ performed data interpretation and manuscript writing. All authors read and approved the final manuscript.

Ethical approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Vincent A, Herman J, Schulick R, Hruban RH and Goggins M: Pancreatic cancer. Lancet. 378:607–620. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Palacio S, McMurry HS, Ali R, Donenberg T, Silva-Smith R, Wideroff G, Sussman DA, Rocha Lima CMS and Hosein PJ: DNA damage repair deficiency as a predictive biomarker for FOLFIRINOX efficacy in metastatic pancreatic cancer. J Gastrointest Oncol. 10:1133–1139. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Rabow MW, Petzel MQB and Adkins SH: Symptom management and palliative care in pancreatic cancer. Cancer J. 23:362–373. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Stromnes IM and Greenberg PD: Greenberg, pancreatic cancer: Planning ahead for metastatic spread. Cancer Cell. 29:774–776. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Kanda M, Fujii T, Nagai S, Kodera Y, Kanzaki A, Sahin TT, Hayashi M, Yamada S, Sugimoto H, Nomoto S, et al: Pattern of lymph node metastasis spread in pancreatic cancer. Pancreas. 40:951–955. 2011. View Article : Google Scholar : PubMed/NCBI

6 

McGuire S; World Cancer Report 2014, . Geneva, Switzerland: World Health Organization, International Agency for Research on Cancer. WHO press. 2015.Adv Nutr. 7:418–419. 2016. View Article : Google Scholar

7 

Giulietti M, Righetti A, Principato G and Piva F: LncRNA co-expression network analysis reveals novel biomarkers for pancreatic cancer. Carcinogenesis. 39:1016–1025. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Flattet Y, Yasmaguchi T, Andrejevic-Blant S and Halkic N: Pancreatic adenocarcinoma: The impact of preneoplastic lesion pattern on survival. Biosci Trends. 9:402–406. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Fu YJ, Li KZ, Bai JH and Liang ZQ: C-reactive protein/albumin ratio is a prognostic indicator in Asians with pancreatic cancers: A meta-analysis. Medicine (Baltimore). 98:e182192019. View Article : Google Scholar : PubMed/NCBI

10 

Schlick K, Magnes T, Huemer F, Ratzinger L, Weiss L, Pichler M, Melchardt T, Greil R and Egle A: C-reactive protein and neutrophil/lymphocytes ratio: Prognostic indicator for doubling overall survival prediction in pancreatic cancer patients. J Clin Med. 8:E17912019. View Article : Google Scholar : PubMed/NCBI

11 

Esteller M: Relevance of DNA methylation in the management of cancer. Lancet Oncol. 4:351–358. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Espada J and Esteller M: DNA methylation and the functional organization of the nuclear compartment. Semin Cell Dev Biol. 21:238–246. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Misawa K, Mochizuki D, Imai A, Endo S, Mima M, Misawa Y, Kanazawa T, Carey TE and Mineta H: Prognostic value of aberrant promoter hypermethylation of tumor-related genes in early-stage head and neck cancer. Oncotarget. 7:26087–26098. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Sato N, Fukushima N, Matsubayashi H, Iacobuzio-Donahue CA, Yeo CJ and Goggins M: Aberrant methylation of Reprimo correlates with genetic instability and predicts poor prognosis in pancreatic ductal adenocarcinoma. Cancer. 107:251–257. 2010. View Article : Google Scholar

15 

Matsubayashi H, Sato N, Fukushima N, Yeo CJ, Walter KM, Brune K, Sahin F, Hruban RH and Goggins M: Methylation of cyclin D2 is observed frequently in pancreatic cancer but is also an age-related phenomenon in gastrointestinal tissues. Clin Cancer Res. 9:1446–1452. 2003.PubMed/NCBI

16 

Henriksen SD, Madsen PH, Larsen AC, Johansen MB, Pedersen IS, Krarup H and Thorlacius-Ussing O: Promoter hypermethylation in plasma-derived cell-free DNA as a prognostic marker for pancreatic adenocarcinoma staging. Int J Cancer. 141:2489–2497. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Henriksen SD, Madsen PH, Larsen AC, Johansen MB, Pedersen IS, Krarup H and Thorlacius-Ussing O: Cell-free DNA promoter hypermethylation in plasma as a predictive marker for survival of patients with pancreatic adenocarcinoma. Oncotarget. 8:93942–93956. 2017. View Article : Google Scholar : PubMed/NCBI

18 

Bournet B, Muscari F, Buscail C, Assenat E, Barthet M, Hammel P, Selves J, Guimbaud R, Cordelier P and Buscail L: KRAS G12D mutation subtype is a prognostic factor for advanced pancreatic adenocarcinoma. Clin Transl Gastroenterol. 7:e1572016. View Article : Google Scholar : PubMed/NCBI

19 

Lee B, Lipton L, Cohen J, Tie J, Javed AA, Li L, Goldstein D, Burge M, Cooray P, Nagrial A, et al: Circulating tumor DNA as a potential marker of adjuvant chemotherapy benefit following surgery for localized pancreatic cancer. Ann Oncol. 30:1472–1478. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Loosen SH, Tacke F, Püthe N, Binneboesel M, Wiltberger G, Alizai PH, Kather JN, Paffenholz P, Ritz T, Koch A, et al: High baseline soluble urokinase plasminogen activator receptor (suPAR) serum levels indicate adverse outcome after resection of pancreatic adenocarcinoma. Carcinogenesis. 40:947–955. 2019. View Article : Google Scholar : PubMed/NCBI

21 

Carreras-Torres R, Johansson M, Gaborieau V, Haycock PC, Wade KH, Relton CL, Martin RM, Davey Smith G and Brennan P: The role of obesity, type 2 diabetes, and metabolic factors in pancreatic cancer: A mendelian randomization study. J Natl Cancer Inst. 109:2017. View Article : Google Scholar : PubMed/NCBI

22 

Iodice S, Gandini S, Maisonneuve P and Lowenfels AB: Tobacco and the risk of pancreatic cancer: A review and meta-analysis. Langenbecks Arch Surg. 393:535–545. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Wang P, Wang Y, Hang B, Zou X and Mao JH: A novel gene expression-based prognostic scoring system to predict survival in gastric cancer. Oncotarget. 7:55343–55351. 2016.PubMed/NCBI

24 

R Core Team, . 2012.R: A language and environment for statistical computing. R Foundation for Statistical Computing. (Vienna, Austria). ISBN 3-900051-07-0. http://www.R-project.org/

25 

Wright RM, et al: Samples and traits for WGCNA. Cryobiology. 71:5442015.

26 

Cao J and Zhang S: A bayesian extension of the hypergeometric test for functional enrichment analysis. Biometrics. 70:84–94. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Balakrishnan R, Harris MA, Huntley R, Van Auken K and Cherry JM: A guide to best practices for Gene Ontology (GO) manual annotation. Database (Oxford). 2013:bat0542013. View Article : Google Scholar : PubMed/NCBI

28 

Huang DW, Sherman BT, Tan Q, Kir J, Liu D, Bryant D, Guo Y, Stephens R, Baseler MW, Lane HC and Lempicki RA: DAVID bioinformatics resources: Expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 35:(Web Server Issue). W169–W175. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Stadler L, et al: Optimizing R language execution via aggressive speculation. Symposium on Dynamic Languages. 2016.

30 

Zou KH, Tuncali K and Silverman SG: Correlation and simple linear regression. Radiology. 227:617–622. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Nadarajah S and Bakar AAS: A new R package for actuarial survival models. Computational Statistics. 28:2139–2160. 2013. View Article : Google Scholar

32 

Goeman JJ: L1 penalized estimation in the Cox proportional hazards model. Biom J. 52:70–84. 2010.PubMed/NCBI

33 

Horne JS and Garton EO: Likelihood cross-validation versus least squares cross-validation for choosing the smoothing parameter in kernel home-range analysis. J Wildlife Man. 70:641–648. 2011. View Article : Google Scholar

34 

Nagy Á, Lánczky A, Menyhárt O and Győrffy B: Validation of miRNA prognosis power in hepatocellular carcinoma using expression data of independent datasets. Sci Rep. 8:92272018. View Article : Google Scholar : PubMed/NCBI

35 

Wakeman CJ, Martin IG, Robertson RW, Dobbs BR and Frizelle FA: Pancreatic cancer: Management and survival. ANZ J Surg. 74:941–944. 2015. View Article : Google Scholar

36 

Court CM and Hines OJ: The new American joint committee on cancer TNM staging system for pancreatic cancer-balancing usefulness with prognostication. JAMA Surg. 153:e1836292018. View Article : Google Scholar : PubMed/NCBI

37 

Xia WX, Zhang LH and Liu YW: Weighted gene co-expression network analysis reveal six hub genes involved in and tight junction function in pancreatic adenocarcinoma and their potential use in prognosis. Genet Test Mol Biomarkers. 23:829–836. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Gailhouste L, Liew LC, Hatada I, Nakagama H and Ochiya T: Epigenetic reprogramming using 5-azacytidine promotes an anti-cancer response in pancreatic adenocarcinoma cells. Cell Death Dis. 9:4682018. View Article : Google Scholar : PubMed/NCBI

39 

Ali S, Cohen C, Little JV, Sequeira JH, Mosunjac MB and Siddiqui MT: The utility of SMAD4 as a diagnostic immunohistochemical marker for pancreatic adenocarcinoma, and its expression in other solid tumors. Diagn Cytopathol. 35:644–648. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Sahin IH, Lowery MA, Stadler ZK, Salo-Mullen E, Iacobuzio-Donahue CA, Kelsen DP and O'Reilly EM: Genomic instability in pancreatic adenocarcinoma: A new step towards precision medicine and novel therapeutic approaches. Expert Rev Gastroenteral Hepatol. 10:893–905. 2016.

41 

Cheng P, Chen Y, He TL, Wang C, Guo SW, Hu H, Ni CM, Jin G and Zhang YJ: Menin coordinates C/EBPβ-mediated TGF-β signaling for epithelial-mesenchymal transition and growth inhibition in pancreatic cancer. Mol Ther Nucleic Acids. 18:155–165. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Kumagai T, Akagi T, Desmond JC, Kawamata N, Gery S, Imai Y, Song JH, Gui D, Said J and Koeffler HP: Epigenetic regulation and molecular characterization of C/EBPalpha in pancreatic cancer cells. Int J Cancer. 124:827–833. 2010. View Article : Google Scholar

43 

Yamamoto K, Tateishi K, Kudo Y, Sato T, Yamamoto S, Miyabayashi K, Matsusaka K, Asaoka Y, Ijichi H, Hirata Y, et al: Loss of histone demethylase KDM6B enhances aggressiveness of pancreatic cancer through downregulation of C/EBPα. Carcinogenesis. 35:2404–2414. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Yamamoto K, Tateishi K, Miyabayashi K, Yamamoto S, Yotaro Y, Mohri D, Asaoka Y, Ijichi H, Omata M and Koike K: Reduced Jmjd3 Expression Enhances Aggressiveness of Pancreatic Cancer Through Downregulation of C/EBPα. Gastroenterology. 140:S1442011. View Article : Google Scholar

45 

Yamada N, Hamada T, Goto M, Tsutsumida H, Higashi M, Nomoto M and Yonezawa S: MUC2 expression is regulated by histone H3 modification and DNA methylation in pancreatic cancer. Int J Cancer. 119:1850–1857. 2010. View Article : Google Scholar

46 

Jespersen C, Soragni E, James Chou C, Arora PS, Dervan PB and Gottesfeld JM: Chromatin structure determines accessibility of a hairpin polyamide-chlorambucil conjugate at histone H4 genes in pancreatic cancer cells. Bioorg Med Chem Lett. 22:4068–4071. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Lavorgna A and Harhaj EW: STAMBPL1 is a deubiquitinating enzyme that regulates HTLV-I Tax subcellular localization and NF-kB activation. Retrovirology. 8:1. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Xiong HQ, Abbruzzese JL, Lin E, Wang L, Zheng L and Xie K: NF-kappaB activity blockade impairs the angiogenic potential of human pancreatic cancer cells. Int J Cancer. 108:181–188. 2004. View Article : Google Scholar : PubMed/NCBI

49 

Peng T, Zhou W, Guo F, Wu HS, Wang CY, Wang L and Yang ZY: Centrosomal protein 55 activates NF-κB signalling and promotes pancreatic cancer cells aggressiveness. Sci Rep. 7:59252017. View Article : Google Scholar : PubMed/NCBI

50 

Abiatari I, Kiladze M, Kerkadze V, Friess H and Kleeff J: Expression of YPEL1 in pancreatic cancer cell lines and tissues. Georgian Medical News. 175:60–62. 2009.

51 

Wolpin BM, Michaud DS, Giovannucci EL, Schernhammer ES, Stampfer MJ, Manson JE, Cochrane BB, Rohan TE, Ma J, Pollak MN and Fuchs CS: Circulating insulin-like growth factor binding protein-1 and the risk of pancreatic cancer. Cancer Res. 67:7923–7928. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Diaz-Aragon R, Ramirez-Ricardo J, Cortes-Reynosa P, Simoni-Nieves A, Gomez-Quiroz LE and Perez Salazar E: Role of phospholipase D in migration and invasion induced by linoleic acid in breast cancer cells. Mol Cell Biochem. 457:119–132. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Foster DA and Xu L: Phospholipase D in cell proliferation and cancer. Mol Cancer Res. 1:789–800. 2003.PubMed/NCBI

54 

Wang Y, Klumpp S, Amin HM, Liang H, Li J, Estrov Z, Zweidler-McKay P, Brandt SJ, Agulnick A and Nagarajan L: SSBP2 is an in vivo tumor suppressor and regulator of LDB1 stability. Oncogene. 29:3044–3053. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Tilley SK, Kim WY and Fry RC: Analysis of bladder cancer tumor CpG methylation and gene expression within The Cancer Genome Atlas identifies GRIA1 as a prognostic biomarker for basal-like bladder cancer. Am J Cancer Res. 7:1850–1862. 2017.PubMed/NCBI

56 

Shu CL, Jing-Yang-Lai, Su LC, Chuu CP and Fukui Y: SWAP-70: A new type of oncogene. PLoS One. 8:e592452013. View Article : Google Scholar : PubMed/NCBI

57 

Jung S, Yi L, Jeong D, Kim J, An S, Oh TJ, Kim CH, Kim CJ, Yang Y, Kim KI, et al: The role of ADCYAP1, adenylate cyclase activating polypeptide 1, as a methylation biomarker for the early detection of cervical cancer. Oncol Rep. 25:245–252. 2011.PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Dou D, Yang S and Zhang J: Prognostic prediction of a 12‑methylation gene‑based risk score system on pancreatic adenocarcinoma. Oncol Lett 20: 85-98, 2020
APA
Dou, D., Yang, S., & Zhang, J. (2020). Prognostic prediction of a 12‑methylation gene‑based risk score system on pancreatic adenocarcinoma. Oncology Letters, 20, 85-98. https://doi.org/10.3892/ol.2020.11575
MLA
Dou, D., Yang, S., Zhang, J."Prognostic prediction of a 12‑methylation gene‑based risk score system on pancreatic adenocarcinoma". Oncology Letters 20.1 (2020): 85-98.
Chicago
Dou, D., Yang, S., Zhang, J."Prognostic prediction of a 12‑methylation gene‑based risk score system on pancreatic adenocarcinoma". Oncology Letters 20, no. 1 (2020): 85-98. https://doi.org/10.3892/ol.2020.11575