
Pancreatic adenocarcinoma (PAAD) is one of the most common malignant tumors with high mortality rates and a poor prognosis. There is an urgent need to determine the molecular mechanism of PAAD tumorigenesis and identify promising biomarkers for the diagnosis and targeted therapy of the disease.
Three GEO datasets (GSE62165, GSE15471 and GSE62452) were analyzed to obtain differentially expressed genes (DEGs). The PPI networks and hub genes were identified through the STRING database and MCODE plugin in Cytoscape software. GO and KEGG enrichment pathways were analyzed by the DAVID database. The GEPIA database was utilized to estimate the prognostic value of hub genes. Furthermore, the roles of MMP14 and COL12A1 in immune infiltration and tumor-immune interaction and their biological functions in PAAD were explored by TIMER, TISIDB, GeneMANIA, Metascape and GSEA.
A total of 209 common DEGs in the three datasets were obtained. GO function analysis showed that the 209 DEGs were significantly enriched in calcium ion binding, serine-type endopeptidase activity, integrin binding, extracellular matrix structural constituent and collagen binding. KEGG pathway analysis showed that DEGs were mainly enriched in focal adhesion, protein digestion and absorption and ECM-receptor interaction. The 14 genes with the highest degree of connectivity were defined as the hub genes of PAAD development. GEPIA revealed that PAAD patients with upregulated MMP14 and COL12A1 expression had poor prognoses. In addition, TIMER analysis revealed that MMP14 and COL12A1 were closely associated with the infiltration levels of macrophages, neutrophils and dendritic cells in PAAD. TISIDB revealed that MMP14 was strongly positively correlated with CD276, TNFSF4, CD70 and TNFSF9, while COL12A1 was strongly positively correlated with TNFSF4, CD276, ENTPD1 and CD70. GSEA revealed that MMP14 and COL12A1 were significantly enriched in epithelial mesenchymal transition, extracellular matrix receptor interaction, apical junction, and focal adhesion in PAAD development.
Our study revealed that overexpression of MMP14 and COL12A1 is significantly correlated with PAAD patient poor prognosis. MMP14 and COL12A1 participate in regulating tumor immune interactions and might become promising biomarkers for PAAD.
Citation: Yuexian Li, Zhou Su, Biwei Wei, Mengbin Qin, Zhihai Liang. Bioinformatics analysis identified MMP14 and COL12A1 as immune-related biomarkers associated with pancreatic adenocarcinoma prognosis[J]. Mathematical Biosciences and Engineering, 2021, 18(5): 5921-5942. doi: 10.3934/mbe.2021296
[1] | Yan Li, Yuzhang Zhu, Guiping Dai, Dongjuan Wu, Zhenzhen Gao, Lei Zhang, Yaohua Fan . Screening and validating the core biomarkers in patients with pancreatic ductal adenocarcinoma. Mathematical Biosciences and Engineering, 2020, 17(1): 910-927. doi: 10.3934/mbe.2020048 |
[2] | Yi Liu, Long Cheng, Xiangyang Song, Chao Li, Jiantao Zhang, Lei Wang . A TP53-associated immune prognostic signature for the prediction of the overall survival and therapeutic responses in pancreatic cancer. Mathematical Biosciences and Engineering, 2022, 19(1): 191-208. doi: 10.3934/mbe.2022010 |
[3] | Yuan Yang, Lingshan Zhou, Xi Gou, Guozhi Wu, Ya Zheng, Min Liu, Zhaofeng Chen, Yuping Wang, Rui Ji, Qinghong Guo, Yongning Zhou . Comprehensive analysis to identify DNA damage response-related lncRNA pairs as a prognostic and therapeutic biomarker in gastric cancer. Mathematical Biosciences and Engineering, 2022, 19(1): 595-611. doi: 10.3934/mbe.2022026 |
[4] | Fang Niu, Zongwei Liu, Peidong Liu, Hongrui Pan, Jiaxue Bi, Peng Li, Guangze Luo, Yonghui Chen, Xiaoxing Zhang, Xiangchen Dai . Identification of novel genetic biomarkers and treatment targets for arteriosclerosis-related abdominal aortic aneurysm using bioinformatic tools. Mathematical Biosciences and Engineering, 2021, 18(6): 9761-9774. doi: 10.3934/mbe.2021478 |
[5] | Kewei Ni, Gaozhong Sun . The identification of key biomarkers in patients with lung adenocarcinoma based on bioinformatics. Mathematical Biosciences and Engineering, 2019, 16(6): 7671-7687. doi: 10.3934/mbe.2019384 |
[6] | Yong Ding, Jian-Hong Liu . The signature lncRNAs associated with the lung adenocarcinoma patients prognosis. Mathematical Biosciences and Engineering, 2020, 17(2): 1593-1603. doi: 10.3934/mbe.2020083 |
[7] | Tongmeng Jiang, Pan Jin, Guoxiu Huang, Shi-Cheng Li . The function of guanylate binding protein 3 (GBP3) in human cancers by pan-cancer bioinformatics. Mathematical Biosciences and Engineering, 2023, 20(5): 9511-9529. doi: 10.3934/mbe.2023418 |
[8] | Tingting Chen, Wei Hua, Bing Xu, Hui Chen, Minhao Xie, Xinchen Sun, Xiaolin Ge . Robust rank aggregation and cibersort algorithm applied to the identification of key genes in head and neck squamous cell cancer. Mathematical Biosciences and Engineering, 2021, 18(4): 4491-4507. doi: 10.3934/mbe.2021228 |
[9] | Ziyu Wu, Sugui Wang, Qiang Li, Qingsong Zhao, Mingming Shao . Identification of 10 differently expressed lncRNAs as prognostic biomarkers for prostate adenocarcinoma. Mathematical Biosciences and Engineering, 2020, 17(3): 2037-2047. doi: 10.3934/mbe.2020108 |
[10] | Mengyang Han, Xiaoli Wang, Yaqi Li, Jianjun Tan, Chunhua Li, Wang Sheng . Identification of coagulation-associated subtypes of lung adenocarcinoma and establishment of prognostic models. Mathematical Biosciences and Engineering, 2023, 20(6): 10626-10658. doi: 10.3934/mbe.2023470 |
Pancreatic adenocarcinoma (PAAD) is one of the most common malignant tumors with high mortality rates and a poor prognosis. There is an urgent need to determine the molecular mechanism of PAAD tumorigenesis and identify promising biomarkers for the diagnosis and targeted therapy of the disease.
Three GEO datasets (GSE62165, GSE15471 and GSE62452) were analyzed to obtain differentially expressed genes (DEGs). The PPI networks and hub genes were identified through the STRING database and MCODE plugin in Cytoscape software. GO and KEGG enrichment pathways were analyzed by the DAVID database. The GEPIA database was utilized to estimate the prognostic value of hub genes. Furthermore, the roles of MMP14 and COL12A1 in immune infiltration and tumor-immune interaction and their biological functions in PAAD were explored by TIMER, TISIDB, GeneMANIA, Metascape and GSEA.
A total of 209 common DEGs in the three datasets were obtained. GO function analysis showed that the 209 DEGs were significantly enriched in calcium ion binding, serine-type endopeptidase activity, integrin binding, extracellular matrix structural constituent and collagen binding. KEGG pathway analysis showed that DEGs were mainly enriched in focal adhesion, protein digestion and absorption and ECM-receptor interaction. The 14 genes with the highest degree of connectivity were defined as the hub genes of PAAD development. GEPIA revealed that PAAD patients with upregulated MMP14 and COL12A1 expression had poor prognoses. In addition, TIMER analysis revealed that MMP14 and COL12A1 were closely associated with the infiltration levels of macrophages, neutrophils and dendritic cells in PAAD. TISIDB revealed that MMP14 was strongly positively correlated with CD276, TNFSF4, CD70 and TNFSF9, while COL12A1 was strongly positively correlated with TNFSF4, CD276, ENTPD1 and CD70. GSEA revealed that MMP14 and COL12A1 were significantly enriched in epithelial mesenchymal transition, extracellular matrix receptor interaction, apical junction, and focal adhesion in PAAD development.
Our study revealed that overexpression of MMP14 and COL12A1 is significantly correlated with PAAD patient poor prognosis. MMP14 and COL12A1 participate in regulating tumor immune interactions and might become promising biomarkers for PAAD.
Abbreviations: PAAD: pancreatic adenocarcinoma; GEO: Gene Expression Omnibus; DEGs: differentially expressed genes; STRING: Search Tool for the Retrieval of Interacting Genes; MCODE: Molecular Complex Detection; GO: Gene Ontology; KEGG: Kyoto Encyclopedia of Genes and Genomes; DAVID: Database for Annotation, Visualization, and Integrated Discovery; GEPIA: Gene Expression Profiling Interactive Analysis; TIMER: Tumor Immune Estimation Resource; GSEA: Gene Set Enrichment Analysis; ECM: extracellular matrix; PPI: protein-protein interaction; MF, molecular function; BP: biological process; CC: cellular component; TCGA: The Cancer Genome Atlas; GTEx: genotype-tissue expression.
Pancreatic adenocarcinoma (PAAD) is the fourth leading cause of cancer deaths due to its high degree of malignancy, high recurrence rate and poor prognosis [1]. According to published statistics, there were nearly 56,770 new cases in the United States in 2019, and the 5-year survival rate is less than 10% [2]. The mechanisms of PAAD involve complex biological processes, including genomic, epigenetic and metabolic changes. The symptoms of PAAD are atypical, the disease develops rapidly, and there are no sensitive early diagnosis biomarkers or appropriate clinical treatments [3]. Potential biomarkers to assist in the diagnosis and treatment of PAAD have long been a research focus [4]. In recent years, many microarray analysis studies have been conducted to identify key genes involved in the progression of PAAD [5]. However, the key drivers of tumorigenesis remain unclear, which limits the early detection and personalized treatment of PAAD. Thus, it is urgent to reveal the underlying molecular pathogenesis of PAAD and identify effective therapeutic targets.
In this research study, we first used three Gene Expression Synthesis (GEO) datasets (GSE62452, GSE15471 and GSE62165) to obtain differentially expressed genes (DEGs). Next, we constructed a protein-protein interaction (PPI) network using the Search Tool for the Retrieval of Interacting Genes (STRING) database and applied Cytoscape to identify significant modules and hub genes. Subsequently, the Database for Annotation, Visualization, and Integrated Discovery (DAVID) was utilized to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses of the DEGs. We then estimated the prognostic value of the hub genes using the Gene Expression Profiling Interactive Analysis (GEPIA) database. Finally, we identified two genes (MMP14 and COL12A1) that were associated with the prognosis of PAAD patients. We further explored the roles of MMP14 and COL12A1 in tumor-immune interactions and their biological functions in PAAD.
In summary, the results of this study provide new clues to further our understanding of PAAD development and identify new immune-related biomarkers for personalized therapy and prognostic monitoring of PAAD patients.
The GEO is a large public genomic database containing array and sequence-based study data. The following keywords were used to search the GEO database: (pancreas OR pancreatic) AND (cancer OR carcinoma OR tumor OR neoplas* OR malignan* OR adenocarcinoma). The inclusion criteria were as follows: (1) patients diagnosed with PAAD; (2) the samples in microarrays were human tissues from patients with PAAD instead of animal tissues and cell lines; (3) the patients with PAAD were not treated with chemotherapy, radiotherapy, or any other methods; and (4) the microarray profiles included PAAD samples and nontumor tissue samples. The exclusion criteria were as follows: (1) cancer or non-cancer groups with small sample sizes (n < 10) and (2) poor‑quality profiling expression data (0, 0.1 or 1) that accounted for > 30% of the total expression data. Three transcriptome expression profiles, GSE62452, GSE15471 and GSE62165, were obtained from the GEO. The GSE62452 dataset included 69 pancreatic cancer samples and 61 normal pancreatic samples. The GSE15471 dataset consisted of 36 pairs of matched pancreatic tumor and nontumor tissues. The GSE62165 dataset contained 118 PAAD samples and 13 control samples. The three datasets were analyzed using the limma package in R software to identify DEGs. |log2FC| > 1 and adjusted P value < 0.05 were set as the cutoffs.
The STRING [6] database (https://string-db.org/) was utilized to construct a PPI network. A total interaction score > 0.4 was considered statistically significant. Cytoscape [7] is a bioinformatics software used to analyze and visualize the interaction networks between molecules. We used the Molecular Complex Detection (MCODE) plugin to determine the densest and significant module in the PPI network. The criteria were as follows: degree cutoff = 2, node score cutoff = 0.2, max depth = 100 and K-score = 2. We selected the genes with the highest node score and the strongest connectivities as the PAAD hub genes.
DAVID [8] (https://david.ncifcrf.gov/) is an integrated online database that can provide researchers with comprehensive gene functional annotation information to help them understand biological processes. GO analysis annotates genes and analyzes the molecular function (MF), biological process (BP) and cellular component (CC) of these genes [9]. KEGG is another powerful database for the functional interpretation of genomic sequences [10]. We used the DAVID database to analyze the GO and KEGG pathways of the DEGs. All significant GO and KEGG enrichment pathways were visualized with the ggplot package.
GEPIA [11] (http://gepia.cancer-pku.cn/) is a user-friendly database that can be used to analyze and visualize large The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) datasets. GEPIA can perform gene expression comparisons between tumor and control samples, gene correlation analysis and survival analysis. The GEPIA database was utilized to explore the prognostic value of PAAD hub genes. In addition, we downloaded the clinical data of PAAD patients from the TCGA database and analyzed the relationship between hub gene expression and different pathological parameters by the chi-square test. A P value < 0.05 was considered statistically significant.
The Tumor Immune Estimation Resource (TIMER) [12] (https://cistrome.shinyapps.io/timer/) systematically analyzes immune infiltration in cancer samples based on TCGA database data with its own algorithm. We used the TIMER database to explore the correlation between MAD14 and COL12A1 and the infiltration level of 6 types of immune cells; specifically, B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and dendritic cells. Besides, TISIDB [13] (http://cis.hku.hk/TISIDB/), an online database for exploring tumor-immune system interactions, was used to assess the correlation between MMP14 and COL12A1 and the immune system in PAAD. A P value < 0.05 obtained from the Spearman's test was considered statistically significant.
GeneMANIA [14] (http://www.genemania.org), a user-friendly website, can display gene or gene lists that share the same functions as submitted genes and provide a PPI network. The genes interacting with MMP14 and COL12A1, as determined by GeneMANIA, were all input into Metascape [15] for further functional annotations and analyses. Meanwhile, we conducted Gene Set Enrichment Analysis (GSEA) [16] to reveal the functional pathways of MMP14 and COL12A1 in PAAD by using transcription data from the TCGA. A permutation test (1000x) was applied to identify the most significantly involved pathways. NOM p-value < 0.05 and FDR q-value < 0.25 were used as the screening criteria.
After standardization of the microarray results, we identified 4063, 1794 and 296 DEGs from GSE62165, GSE15471 and GSE62452 respectively. Figures 1A–1C show the differential expression of genes in the tumor and normal samples in the three microarrays. A total of 209 common DEGs (153 upregulated and 56 downregulated) in the three datasets were obtained through the online tool Venn diagram (http://bioinformatics.psb.ugent.be/webtools/Venn/) (Figure 1D). The PPI network of DEGs was constructed using the STRING database and then visualized by Cytoscape software, as shown in Figure 1E. The most significant module, with 14 nodes and 83 edges, was obtained based on the PPI network of the DEGs. A total of 14 upregulated genes were identified as hub PAAD genes with a high degree of connectivity, FN1, BGN, THSB2, ASPN, COL11A1, FBN1, COL12A1, COL6A3, SPARC, COL1A1, COL3A1, COL5A2, MMP14 and POSTN (Figure 1F).
The GO analysis results showed that the DEGs were significantly enriched in the biological processes of proteolysis, cell adhesion, skeletal system development, extracellular matrix organization, extracellular matrix disassembly, collagen fibril organization, collagen catabolic process and endodermal cell differentiation (Figure 2A). Changes in cellular components were significantly enriched in extracellular exosomes, extracellular regions, cell surfaces, extracellular space, endoplasmic reticulum lumen, proteinaceous extracellular matrix, collagen trimers and extracellular matrix (Figure 2B). With regard to molecular function, DEGs were mainly enriched in calcium ion binding, serine-type endopeptidase activity, integrin binding, extracellular matrix structural constituent and collagen binding (Figure 2C). KEGG pathway analysis showed that the DEGs were mainly enriched in focal adhesion, protein digestion and absorption and (extracellular matrix) ECM-receptor interaction (Figure 2D).
Using GEPIA data, we noted that among the 14 hub genes, only MMP14 and COL12A1 were associated with the prognosis of PAAD patients. Patients with high MMP14 expression had a worse overall (P = 0.033, HR = 1.6) and disease-free survival rates (P = 0.0035, HR = 1.9) (Figure 3A, B). High expression of COL12A1 indicated a poor overall survival outcome (P = 0.0055, HR = 1.8), but it had no effect on the patient's disease-free survival rate (P = 0.057, HR = 1.5) (Figure 3C, D). In addition, as shown in Table 1, MMP14 expression was correlated with tumor grade and M classification, and COL12A1 expression was associated with tumor grade, stage, and T, N and M classification in PAAD (P value < 0.05). Taken together, these results suggested that the hub genes MMP14 and COL12A1 have clinical relevance in PAAD. Thus, we further explored the roles of MMP14 and COL12A1 in tumor-immune interactions and their biological functions in PAAD.
Parameters | Overall(n = 178) | COL12A1 expression | P | MMP14 expression | P | ||
low (89) | high (89) | high (89) | low (89) | ||||
Primary site (%) | 0.262 | 0.295 | |||||
Body of Pancreas | 14 (7.9) | 9 (10.1) | 5 (5.6) | 9 (10.1) | 5 (5.6) | ||
Head of Pancreas | 139 (78.1) | 65 (73.0) | 74 (83.1) | 69 (77.5) | 70 (78.7) | ||
Tail of Pancreas | 14 (7.9) | 7 (7.9) | 7 (7.9) | 8 (9.0) | 6 (6.7) | ||
Other | 11 (6.2) | 8 (9.0) | 3 (3.4) | 3 (3.4) | 8 (9.0) | ||
Pathological type (%) | 0.094 | 0.094 | |||||
Other Type | 25 (14.1) | 18 (20.2) | 7 (8.0) | 7 (8.0) | 18 (20.2) | ||
Ductal Type | 147 (83.1) | 69 (77.5) | 78 (88.6) | 78 (88.6) | 69 (77.5) | ||
Colloid Carcinoma | 4 (2.3) | 2 (2.2) | 2 (2.3) | 2 (2.3) | 2 (2.2) | ||
Undifferentiated Carcinoma | 1 (0.6) | 0 (0.0) | 1 (1.1) | 1 (1.1) | 0 (0.0) | ||
Grade (%) | 0.044 | 0.018 | |||||
G1 | 31 (17.4) | 22 (24.7) | 9 (10.1) | 8 (9.0) | 23 (25.8) | ||
G2 | 95 (53.4) | 40 (44.9) | 55 (61.8) | 51 (57.3) | 44 (49.4) | ||
G3 | 48 (27.0) | 24 (27.0) | 24 (27.0) | 29 (32.6) | 19 (21.3) | ||
G4 | 2 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | ||
GX | 2 (1.1) | 2 (2.2) | 0 (0.0) | 0 (0.0) | 2 (2.2) | ||
Tumor stage (%) | 0.004 | 0.28 | |||||
Stage Ⅰ | 21 (12.0) | 18 (20.7) | 3 (3.4) | 7 (8.0) | 14 (16.1) | ||
Stage Ⅱ | 147 (84.0) | 66 (75.9) | 81 (92.0) | 76 (86.4) | 71 (81.6) | ||
Stage Ⅲ | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.3) | 1 (1.1) | ||
Stage Ⅳ | 4 (2.3) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
T classification (%) | 0.004 | 0.444 | |||||
T1 | 7 (4.0) | 7 (8.0) | 0 (0.0) | 2 (2.2) | 5 (5.7) | ||
T2 | 24 (13.6) | 17 (19.3) | 7 (7.9) | 10 (11.2) | 14 (15.9) | ||
T3 | 142 (80.2) | 61 (69.3) | 81 (91.0) | 75 (84.3) | 67 (76.1) | ||
T4 | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.2) | 1 (1.1) | ||
TX | 1 (0.6) | 1 (1.1) | 0 (0.0) | 0 (0.0) | 1 (1.1) | ||
N classification (%) | 0.08 | 0.977 | |||||
N0 | 49 (27.7) | 30 (34.1) | 19 (21.3) | 24 (27.0) | 25 (28.4) | ||
N1 | 124 (70.1) | 55 (62.5) | 69 (77.5) | 63 (70.8) | 61 (69.3) | ||
NX | 4 (2.3) | 3 (3.4) | 1 (1.1) | 2 (2.2) | 2 (2.3) | ||
M classification (%) | 0.022 | 0.046 | |||||
M0 | 80 (44.9) | 32 (36.0) | 48 (53.9) | 47 (52.8) | 33 (37.1) | ||
M1 | 4 (2.2) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
MX | 94 (52.8) | 56 (62.9) | 38 (42.7) | (43.8) | (61.8) |
TIMER analysis revealed that MMP14 had significant positive associations with infiltrating levels of CD8+ T cells (r = 0.251, p = 9.24e−4), macrophages (r = 0.417, p = 1.42e−8), neutrophils (r = 0.46, p = 2.5e−10) and dendritic cells (r = 0.463, p = 1.77e−10) in PAAD (Figure 4A). COL12A1 significantly correlated with B cells (r = 0.183, p = 1.67e−2), CD8+ T cells (r = 0.386, p = 1.87e−7), macrophages (r = 0.537, p = 3.58e−14), neutrophils (r = 0.509, p = 1.24e−12) and dendritic cells (r = 0.526, p = 1.55e−13) in PAAD (Figure 4B). Specifically, both MMP14 and COL12A1 were strongly correlated with macrophage, neutrophil and dendritic cell infiltration.
Meanwhile, TISIDB was utilized to comprehensively analyze the relationship between MMP14 and COL12A1 and immune molecules in PAAD. With regard to tumor infiltrating lymphocytes (Figure 5), MMP14 was found to positively correlate with Tcm CD4+ T cells (rho = 0.526, P < 2.26E−16), Act DCs (rho = 0.451, P = 3.6E−10), NK cells (rho = 0.447, P = 5.38E−10) and Tcm CD8+ T cells (rho = 0.432, P = 2.23E−9), while COL12A1 was positively correlated with NK cells (rho = 0.524, P < 2.2E−16), Tcm CD8+ T cells (rho = 0.502, P < 2.2E−16), Treg cells (rho = 0.481, P = 7.19E−12) and Tgd cells (rho = 0.475, P = 2.14E-11). With regard to immunoinhibitors (Figure 6), MMP14 was found to positively correlate with TGFB1 (rho = 0.584, P < 2.26E−16), PDCD1LG2 (rho = 0.366, P = 5.69E−7), HAVCR2 (rho = 0.325, P = 1.01E−5) and TGFBR1 (rho = 0.252, P = 0.000682) while COL12A1 was positively correlated with PDCD1LG2 (rho = 0.583, P < 2.2E−16), TGFBR1 (rho = 0.431, P = 2.46E−9), HAVCR2 (rho = 0.407, P = 2.18E−8) and CD274 (rho = 0.346, P = 2.4E−6). With regard to immunostimulators (Figure 7), MMP14 was strongly positively correlated with CD276 (rho = 0.861, P < 2.2E−16), TNFSF4 (rho = 0.605, P < 2.2E−16), CD70 (rho = 0.573, P < 2.2E−16) and TNFSF9 (rho = 0.426, P = 4.02E−9) while COL12A1 was strongly positively correlated with TNFSF4 (rho = 0.758, P < 2.2E−16), CD276 (rho = 0.594, P < 2.2E−16), ENTPD1 (rho = 0.577, P < 2.2E−16) and CD70 (rho = 0.512, P < 2.2E−16). In conclusion, these results indicate functions in the immune process of PAAD.
Figure 8A, B shows the interaction network and biological functions of MMP14 and COL12A1 and their related genes, respectively. MMP14 was found to be involved in multiple biological processes in PAAD, such as extracellular structure organization, blood vessel development, negative regulation of metallopeptidase activity and amyloid-beta clearance (Figure 8C). COL12A1 was mainly involved in extracellular matrix organization, the pid integrin1 pathway, integrin cell surface interactions, and MET activated PTK2 signaling in PAAD (Figure 8D). Moreover, GSEA was utilized to conduct hallmark and KEGG analyses for MMP14 and COL12A1 expression. The results indicated that the most significant pathways involving MMP14 included epithelial mesenchymal transition, apical junction, ECM receptor interaction, and focal adhesion (Table 2). The most significant pathways involving COL12A1 included epithelial mesenchymal transition, apical junction, focal adhesion, ECM receptor interaction, and regulation of actin cytoskeleton (Table 3).
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.76 | 2.25 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.52 | 2.24 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical surface | 0.52 | 1.90 | ≤ 0.001 | 0.04 |
Hallmark-mitotic spindle | 0.47 | 1.83 | 0.01 | 0.06 |
Hallmark-hypoxia | 0.47 | 1.82 | 0.01 | 0.06 |
Hallmark-TGF-β signaling | 0.56 | 1.82 | 0.01 | 0.05 |
Hallmark-angiogenesis | 0.57 | 1.80 | 0.01 | 0.05 |
Hallmark-WNT-β catenin signaling | 0.47 | 1.69 | 0.02 | 0.09 |
Hallmark-apoptosis | 0.41 | 1.69 | 0.01 | 0.08 |
Hallmark-coagulation | 0.40 | 1.65 | 0.01 | 0.08 |
KEGG-ECM receptor interaction | 0.69 | 2.17 | ≤ 0.001 | ≤ 0.001 |
KEGG-focal adhesion | 0.58 | 2.15 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.46 | 1.98 | ≤ 0.001 | 0.03 |
KEGG-basal cell carcinoma | 0.58 | 1.96 | ≤ 0.001 | 0.03 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.71 | 1.92 | ≤ 0.001 | 0.04 |
KEGG-regulation of actin cytoskeleton | 0.44 | 1.91 | ≤ 0.001 | 0.04 |
KEGG-small cell lung cancer | 0.54 | 1.90 | ≤ 0.001 | 0.03 |
KEGG-pathogenic Escherichia coli infection | 0.55 | 1.83 | 0.01 | 0.06 |
KEGG-bladder cancer | 0.54 | 1.83 | 0.01 | 0.06 |
KEGG-axon guidance | 0.43 | 1.75 | ≤0.001 | 0.10 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.78 | 2.41 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.48 | 2.10 | ≤ 0.001 | 0.01 |
Hallmark-UV response DN | 0.58 | 2.09 | ≤ 0.001 | 0.01 |
Hallmark-TGF-β signaling | 0.59 | 1.97 | ≤ 0.001 | 0.02 |
Hallmark-angiogenesis | 0.61 | 1.93 | 0.01 | 0.02 |
Hallmark-inflammatory response | 0.53 | 1.86 | 0.01 | 0.04 |
Hallmark-coagulation | 0.44 | 1.78 | ≤ 0.001 | 0.05 |
Hallmark-apoptosis | 0.42 | 1.78 | 0.01 | 0.05 |
Hallmark-complement | 0.45 | 1.76 | 0.01 | 0.05 |
Hallmark-notch signaling | 0.52 | 1.70 | 0.02 | 0.07 |
KEGG-focal adhesion | 0.64 | 2.33 | ≤ 0.001 | ≤ 0.001 |
KEGG-ECM receptor interaction | 0.71 | 2.20 | ≤ 0.001 | ≤ 0.001 |
KEGG-regulation of actin cytoskeleton | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-TGF-β signaling | 0.55 | 2.11 | ≤ 0.001 | ≤ 0.001 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.77 | 2.10 | ≤ 0.001 | ≤ 0.001 |
KEGG- small cell lung cancer | 0.56 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- basal cell carcinoma | 0.57 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- melanoma | 0.48 | 1.88 | ≤ 0.001 | 0.04 |
KEGG- adherens junction | 0.53 | 1.87 | 0.01 | 0.04 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |
PAAD is one of the most common digestive system malignancies worldwide. The treatments available for advanced PAAD are limited, and the patient prognosis is poor. The mechanisms leading to malignancy in PAAD remain poorly characterized. Moreover, biomarkers for targeted therapy and prognosis monitoring are still elusive. In many cancers, the level of tumor immune infiltration is closely associated with patient prognosis. Thus, there is an urgent need to identify new immune-associated biomarkers that are associated with prognosis and treatment in PAAD.
In this study, we identified 209 DEGs in PAAD using three microarray datasets from the GEO database. Through GO and KEGG enrichment analyses, we determined the biological functions and pathways of these DEGs in the development of PAAD. GO functional annotations identified many biological pathways associated with the DEGs in PAAD, including cell adhesion, ECM organization, proteolysis, collagen fibril organization, collagen catabolic process and endodermal cell differentiation. KEGG pathway analysis revealed that the DEGs were strongly associated with focal adhesion, ECM-receptor interaction and protein digestion and absorption. These pathways were found to be the most significant pathways in relation to PAAD occurrence and development. Cell adhesion is known to be a key mediator of invasiveness, metastasis, and immune suppression during malignancy since homeostasis in normal tissues is dependent on cell-to-cell adhesion [17]. Mutations and changes in the expression of cell adhesion molecules cadherins, integrins, selectins, and immunoglobulins have been associated with cancer progression in many studies [18]. The dysregulation of the ECM has recently been recognized as a key driver in the development and progression of cancer, as its organization and disassembly control malignant cell behavior and differentiation [19]. Proteolysis is the most fundamental property of malignancy, and its inhibition may be used to attenuate tumor invasion, angiogenesis and migration [20]. Perumal et al found that proteolysis of collagen fibrils is a key process in normal growth, development, repair, and cell differentiation, and in cancerous tumor progression [21]. Collagen is a major component of the ECM, and increasing evidence suggests that it influences tumor cell progression and metastasis through integrins, discoidin domain receptors, tyrosine kinase receptors, and some signaling pathways [22]. Bastidas-Ponce et al described the cellular process of pancreatic morphogenesis and demonstrated that pancreatic formation and development were closely related to endodermal cell differentiation [23]. The functional enrichment results obtained by analyzing the DEGs identified in this study provided novel insights into the mechanisms of PAAD occurrence and development. Based on the PPI network analyzed by Cytoscape, a total of 14 hub genes involved in PAAD development were identified as follows: FN1, BGN, THSB2, ASPN, COL11A1, FBN1, COL12A1, COL6A3, SPARC, COL1A1, COL3A1, COL5A2, MMP14 and POSTN.
To determine whether these hub genes have clinical relevance in PAAD, we used GEPIA to analyze their prognostic value. We found that only MMP14 and COL12A1 were significantly associated with the survival outcomes of PAAD patients among the 14 hub genes. Our results revealed that high expression of MMP14 and COL12A1 tended to predict poor PAAD patient prognosis. In addition, MMP14 expression was correlated with tumor grade and M classification, while the expression of COL12A1 was related to tumor grade, stage, and T, N and M classification. Taken together, these results suggested that the hub genes MMP14 and COL12A1 had clinical relevance in PAAD. Thus, we selected MMP14 and COL12A1 as true hub genes for further analysis. Functional enrichment analysis and GSEA showed that MMP14 and COL12A1 significantly were involved in cancer hallmarks and KEGG pathways, including epithelial mesenchymal transition, ECM receptor interaction, apical junction, and focal adhesion. Proteolytic degradation of ECM and epithelial mesenchymal transition are known to be crucial steps in the development and progression of cancer [19,24]. The results of this study further confirmed that MMP14 and COL12A1 play important roles in the tumorigenesis and progression of PAAD.
MMP14 is a member of the membrane-type matrix metalloprotease (MMP) family with previously known roles in ECM degradation or proteolytic protein processing [25]. Previous studies demonstrated that MMP14 affects cancer cell proliferation and invasion and is usually associated with poor patient prognosis in those with cancers such as bladder cancer, gastric cancer and hepatocellular carcinoma[26,27,28]. A meta-analysis comprehensively evaluated the expression levels of MMP14 and its prognostic value in digestive system carcinomas, such as gastric cancer, esophageal carcinoma, oral cancer and hepatocellular carcinoma [29]. However, there were no data on its prognostic value in PAAD. Previous results indicated that MMP14 mainly exerts its proto-oncogenic functions by regulating processes such as ECM degradation and remodeling, cell invasion, and cancer metastasis [30]. Wei Jiang et al reported that MMP14 expression is required for the induction of epithelial mesenchymal transition and the activation of an invasive program in pancreatic cancer [31]. These results revealed the importance of MMP14 in PAAD and indicated that MMP14 is a potential therapeutic target for PAAD.
COL12A1 encodes the alpha chain of collagen XII, which is a member of the fibril-associated collagen family with interrupted triple helices [32]. Collage XII is an important dominant component of the ECM and interacts with other ECM molecules to provide structural support for cells [33]. As an ECM-related gene, COL12A1 has received increasing attention due to its critical role in human cancer. Some studies have explored the prognostic value of COL12A1 in different cancer types, such as breast cancer, gastric cancer and colorectal cancer [34,35,36]. IDO1 and COL12A1 synergistically participate in the metastasis of gastric cancer via the MAPK pathway[37]. The expression of COL12A1 was found to be increased in ovarian cancer and associated with the drug resistance of cancer cells[38]. In another study, it was revealed that type Ⅳ collagen promotes pancreatic cancer cell proliferation and migration and inhibits apoptosis through an autocrine loop[39]. The expression of Snail, a well-known regulator of epithelial-mesenchymal transition, was found to increase in pancreatic cancer cells in the presence of a collagen-rich milieu, suggesting that the desmoplastic reaction actively contributes to cancer progression[40]. Taken together, these results indicate that COL12A1 might serve as an effective biomarker and therapeutic target for PAAD.
Significant progress has been made in the treatment of malignancies, especially via the use of immunotherapies. In this study, we conducted a correlation analysis between MMP14 and COL12A1 and immune-infiltrating cells in PAAD. Our results demonstrated that the expression levels of both MMP14 and COL12A1 were significantly positively associated with the infiltration levels of macrophages, neutrophils, dendritic cells and NK cells in PAAD (all P < 0.05, all cor > 0.4). Strong evidence has indicated that these tumor-infiltrating cells play significant roles in the development and progression of PAAD. Macrophages, which are the main components of the tumor microenvironment, are known to promote the initiation, progression, angiogenesis, invasion and metastasis of PAAD [41]. A meta-analysis of 1699 patients showed that a high infiltration level of macrophages was significantly associated with poor overall and disease-free survival rates in pancreatic cancer patients [42]. Many studies have established that neutrophils also promote the initiation, development and progression of pancreatic cancer and that high levels of neutrophils predict poor patient prognoses [43]. Dendritic cells are professional antigen-presenting cells and are involved in the induction and regulation of antitumor immune responses. The levels of dendritic cells have been found to be a prognostic factor in pancreatic cancer [44]. Recent clinical evidence has shown that NK cells play a critical role in the antigen-independent immune response against malignant cells and that high NK infiltration predicts poor survival in patients with advanced pancreatic cancer [45]. Patients with a high abundance of these tumor-infiltrating cells had poor survival outcomes, which is consistent with the relationship between high expression of MMP14 and COL12A1 and PAAD patient survival. Previous studies have reported that MMPs promote cancer disease progression, new blood vessel formation, invasion, metastasis, the activity of inflammatory cytokines and chemokines, and avoidance of immune surveillance [46]. MMP14 controls macrophage immune function by modulating inflammatory responses, which triggers the activation of PI3Kδ signal cascades and the remodeling of the Mi-2/NuRD nucleosome complex [47]. In addition, the results of other recent studies suggested that high collagen density can modulate the activity and functions of tumor-infiltrating macrophages and T cells to support cancer cell immune escape, which was associated with poor prognosis in patients with several types of tumor[48,49]. Thus, we hypothesize that the MMP14- and COL12A1-regulated changes in the abundance and functions of tumor immune cells, especially macrophages, neutrophils, dendritic cells and NK cells may lead to invasion, immune system avoidance, and metastasis, which is responsible for the poor prognosis of PAAD patients. However, further experiments and prospective studies are warranted to validate the exact link between these tumor-infiltrating cells and MMP14 and COL12A1 in PAAD.
We also explored the association between MMP14 and COL12A1 and immune markers in PAAD. Among the immunostimulators, there was a high correlation coefficient between the expression of MMP14 and CD276, TNFSF4, CD70, and TNFSF9. There was also a high correlation coefficient between the expression of COL12A1 and TNFSF4, CD276, ENTPD1, and CD70 in PAAD. In terms of immunoinhibitors, MMP14 expression was positively correlated with TGFB1, PDCD1LG2, HAVCR2 and TGFBR1 while COL12A1 was positively correlated with PDCD1LG2, TGFBR1, HAVCR2 and CD274 in PAAD. Macrophages, neutrophils and dendritic cells are an essential component of innate immunity and play a vital role in various inflammatory diseases as well as in tumor progression. Evidence indicates that tumor-infiltrating lymphocytes play a significant role in the promotion or inhibition of tumor growth [50]. Du et al reported that CAR-T cells targeting CD276/B7-H3 effectively controlled the growth of pancreatic ductal adenocarcinoma, ovarian cancer and neuroblastoma in vitro and in mice without obvious toxicity [51]. CD70 is a costimulatory molecule that participates in the survival of regulatory T cells by interacting with its ligand CD27. Many studies have indicated that anti-CD70 therapy is likely beneficial for stimulating antitumor immune responses [52]. TNFSF4 is a costimulatory checkpoint protein that has been proven to enhance the antitumor activity of T cells [53]. TNFSF9 is highly expressed in PAAD tissues and is related to M1 polarization of macrophages [54]. Chen Liang demonstrated that TGFB1 could affect the progression of pancreatic cancer based on the status of SMAD4 [55]. The PD-1 receptor and its ligands PD-L1/CD274 and PD-L2/PDCD1LG2 play key roles in T cell inhibition and exhaustion. Monoclonal antibodies that block the PD-1/PD-L1 pathway have been developed for cancer immunotherapy and enhance T cell function, especially in melanoma, non-small cell lung cancer, renal cell carcinoma and bladder cancer [56]. The interactions between MMP14 and COL12A1 and immune markers have rarely been reported. Our results provide evidence for a strong correlation between MMP14 and COL12A1 and the above immune molecules in PAAD, indicating that MMP14 and COL12A1 may serve as immune-related therapy targets for PAAD.
Our study has some limitations. This study is based on bulk RNA sequencing data used to identify biomarkers for PAAD and gene expression in rare clonal cells may have been missed. Due to the heterogeneity of cancer cells, multiple research methods are needed to reveal the molecular mechanisms of PAAD development. Emerging single-cell RNA sequencing technology, which can study every individual cell in a tumor, has become an important tool for analyzing cell biology, discovering the evolutionary relationships among cells, and revealing the heterogeneity of tumor cells [57]. Single-cell RNA sequencing technology provides a new perspective on the regulatory relationship between gene expression and the cell microenvironment [58]. Moreover, single-cell RNA sequencing technology helps identify biomarkers for predicting the prognosis and therapeutic response of tumors [59]. Recent studies have revealed the value of the application of single-cell RNA sequencing for characterizing cell heterogeneity and identifying specific cell-type biomarkers in pancreatic cancer [60,61]. In future studies, we will integrate and analyze more available data, including single-cell sequencing genomic, transcriptome and proteomic data, to provide new insights into the mechanisms of PAAD development and diagnostic and therapeutic strategies for PAAD patients.
This study identified 14 hub genes and the molecular mechanisms of PAAD development by using bioinformatics analysis. Among the 14 hub genes, we found that overexpression of MMP14 and COL12A1 was significantly correlated with poor PAAD patient prognosis. Furthermore, the expression of MMP14 and COL12A1 is strongly associated with the level of immune cell infiltration and the levels of multiple immunomodulators in PAAD. Our study demonstrates that MMP14 and COL12A1 play an important role in the tumorigenesis and progression of PAAD and might become promising biomarkers and therapeutic targets.
This work was supported by the Nanning Qingxiu District Science and Technology Project (No.2019026), National Natural Science Foundation of China (No. 81960126), and Guangxi Natural Science Foundation (2018GXNSFBA281154).
The authors declare there is no conflict of interest.
[1] |
H. Zhu, T. Li, Y. Du, M. Li, Pancreatic cancer: challenges and opportunities, BMC Med., 16 (2018), 214. doi: 10.1186/s12916-018-1215-3
![]() |
[2] |
R. L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2019, CA Cancer J. Clin., 69 (2019), 7-34. doi: 10.3322/caac.21551
![]() |
[3] |
J. Kleeff, M. Korc, M. Apte, C. La Vecchia, C. D. Johnson, A. V. Biankin, et al., Pancreatic cancer, Nat. Rev. Dis. Primers, 2 (2016), 16022. doi: 10.1038/nrdp.2016.22
![]() |
[4] |
A. Martín-Blázquez, C. Jiménez-Luna, C. Díaz, J. Martínez-Galán, J. Prados, F. Vicente, et al., Discovery of Pancreatic Adenocarcinoma Biomarkers by Untargeted Metabolomics, Cancers, 12 (2020), 1002. doi: 10.3390/cancers12041002
![]() |
[5] |
W. Lu, N. Li, F. Liao, Identification of key genes and pathways in pancreatic cancer gene expression profile by integrative analysis, Genes, 10 (2019), 612. doi: 10.3390/genes10080612
![]() |
[6] |
C. von Mering, M. Huynen, D. Jaeggi, S. Schmidt, P. Bork, B. Snel, STRING: a database of predicted functional associations between proteins, Nucleic Acids Res., 31 (2003), 258-261. doi: 10.1093/nar/gkg034
![]() |
[7] |
P. Shannon, A. Markiel, O. Ozier, N. S. Baliga, J. T. Wang, D. Ramage, et al., Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome Res., 13 (2003), 2498-2504. doi: 10.1101/gr.1239303
![]() |
[8] |
D. W. Huang, B. T. Sherman, Q. Tan, J. Kir, D. Liu, D. Bryant, et al., DAVID Bioinformatics Resources: expanded annotation database and novel algorithms to better extract biology from large gene lists, Nucleic Acids Res., 35 (2007), W169-W175. doi: 10.1093/nar/gkm415
![]() |
[9] |
Gene Ontology Consortium, The Gene Ontology Resource: 20 years and still GOing strong, Nucleic Acids Res., 47 (2019), D330-D338. doi: 10.1093/nar/gky1055
![]() |
[10] |
M. Kanehisa, Y. Sato, M. Kawashima, M. Furumichi, M. Tanabe, KEGG as a reference resource for gene and protein annotation, Nucleic Acids Res., 44 (2016), D457-D462. doi: 10.1093/nar/gkv1070
![]() |
[11] |
Z. Tang, C. Li, B. Kang, G. Gao, C. Li, Z. Zhang, GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses, Nucleic Acids Res., 45 (2017), W98-W102. doi: 10.1093/nar/gkx247
![]() |
[12] |
T. Li, J. Fan, B. Wang, N. Traugh, Q. Chen, J.S. Liu, et al., TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells, Cancer Res., 77 (2017), e108-e110. doi: 10.1158/0008-5472.CAN-17-0307
![]() |
[13] |
B. Ru, C. N. Wong, Y. Tong, J. Y. Zhong, S. S. W. Zhong, W. C. Wu, et al., TISIDB: an integrated repository portal for tumor-immune system interactions, Bioinformatics, 35 (2019), 4200-4202. doi: 10.1093/bioinformatics/btz210
![]() |
[14] |
M. Franz, H. Rodriguez, C. Lopes, K. Zuberi, J. Montojo, G. D. Bader, et al., GeneMANIA update 2018, Nucleic Acids Res., 46 (2018), W60-W64. doi: 10.1093/nar/gky311
![]() |
[15] |
Y. Zhou, B. Zhou, L. Pache, M. Chang, A. H. Khodabakhshi, O. Tanaseichuk, et al., Metascape provides a biologist-oriented resource for the analysis of systems-level datasets, Nat. Commun., 10 (2019), 1523. doi: 10.1038/s41467-019-09234-6
![]() |
[16] |
A. Subramanian, H. Kuehn, J. Gould, P. Tamayo, J.P. Mesirov, GSEA-P: a desktop application for Gene Set Enrichment Analysis, Bioinformatics, 23 (2007), 3251-3253. doi: 10.1093/bioinformatics/btm369
![]() |
[17] |
H. Läubli, L. Borsig, Altered cell adhesion and glycosylation promote cancer immune suppression and metastasis, Front. Immunol., 10 (2019), 2120. doi: 10.3389/fimmu.2019.02120
![]() |
[18] |
M. Janiszewska, M.C. Primi, T. Izard, Cell adhesion in cancer: Beyond the migration of single cells, J. Biol. Chem., 295 (2020), 2495-2505. doi: 10.1074/jbc.REV119.007759
![]() |
[19] |
C. Walker, E. Mojares, A. Del Río Hernández, Role of Extracellular Matrix in Development and Cancer Progression, Int. J. Mol. Sci., 19 (2018), 3028. doi: 10.3390/ijms19103028
![]() |
[20] | M. Wyganowska-Świątkowska, M. Tarnowski, D. Murtagh, E. Skrzypczak-Jankun, J. Jankun, Proteolysis is the most fundamental property of malignancy and its inhibition may be used therapeutically (Review), Int. J. Mol. Med., 43 (2019), 15-25. |
[21] |
S. Perumal, O. Antipova, J. P. Orgel, Collagen fibril architecture, domain organization, and triple-helical conformation govern its proteolysis, Proc. Natl. Acad. Sci., 105 (2008), 2824-2829. doi: 10.1073/pnas.0710588105
![]() |
[22] |
S. Xu, H. Xu, W. Wang, S. Li, H. Li, T. Li, et al., The role of collagen in cancer: from bench to bedside, J. Transl. Med., 17 (2019), 309. doi: 10.1186/s12967-019-2058-1
![]() |
[23] |
A. Bastidas-Ponce, K. Scheibner, H. Lickert, M. Bakhti, Cellular and molecular mechanisms coordinating pancreas development, Development, 144 (2017), 2873-2888. doi: 10.1242/dev.140756
![]() |
[24] |
M. Singh, N. Yelle, C. Venugopal, S. K. Singh, EMT: Mechanisms and therapeutic implications, Pharmacol. Ther., 182 (2018), 80-94. doi: 10.1016/j.pharmthera.2017.08.009
![]() |
[25] |
S. P. Turunen, O. Tatti-Bugaeva, K. Lehti, Membrane-type matrix metalloproteases as diverse effectors of cancer progression, Biochim. Biophys. Acta Mol. Cell Res., 1864 (2017), 1974-1988. doi: 10.1016/j.bbamcr.2017.04.002
![]() |
[26] | J. F. Wang, Y. Q. Gong, Y. H. He, W. W. Ying, X. S. Li, X. F. Zhou, et al., High expression of MMP14 is associated with progression and poor short-term prognosis in muscle-invasive bladder cancer, Eur. Rev. Med. Pharmacol. Sci., 24 (2020), 6605-6615. |
[27] |
A. Kasurinen, S. Gramolelli, J. Hagström, A. Laitinen, A. Kokkola, Y. Miki, et al., High tissue MMP14 expression predicts worse survival in gastric cancer, particularly with a low PROX1, Cancer Med., 8 (2019), 6995-7005. doi: 10.1002/cam4.2576
![]() |
[28] | Y. Jin, Z. Y. Liang, W. X. Zhou, L. Zhou, High MMP14 expression is predictive of poor prognosis in resectable hepatocellular carcinoma, Pathology, 52 (2020), 359-365. |
[29] |
F. Duan, Z. Peng, J. Yin, Z. Yang, J. Shang, Expression of MMP-14 and prognosis in digestive system carcinoma: a meta-analysis and databases validation, J. Cancer, 11 (2020), 1141-1150. doi: 10.7150/jca.36469
![]() |
[30] | O. R. Grafinger, G. Gorshtein, T. Stirling, M. I. Brasher, M. G. Coppolino, β1 integrinmediated signaling regulates MT1-MMP phosphorylation to promote tumor cell invasion, J. Cell Sci., 133 (2020), jcs239152. |
[31] |
W. Jiang, Y. Zhang, K. T. Kane, M. A. Collins, D. M. Simeone, M. P. di Magliano, et al., CD44 regulates pancreatic cancer invasion through MT1-MMP, Mol. Cancer Res., 13 (2015), 9-15. doi: 10.1158/1541-7786.MCR-14-0076
![]() |
[32] |
D. R. Gerecke, P. F. Olson, M. Koch, J. H. Knoll, R. Taylor, D. L. Hudson, et al., Complete primary structure of two splice variants of collagen XⅡ, and assignment of alpha 1(XⅡ) collagen (COL12A1), alpha 1(IX) collagen (COL9A1), and alpha 1(XIX) collagen (COL19A1) to human chromosome 6q12-q13, Genomics, 41 (1997), 236-242. doi: 10.1006/geno.1997.4638
![]() |
[33] |
J. Sapudom, T. Pompe, Biomimetic tumor microenvironments based on collagen matrices, Biomater. Sci., 6 (2018), 2009-2024. doi: 10.1039/C8BM00303C
![]() |
[34] |
Y. H. Xu, J. L. Deng, L. P. Wang, H. B. Zhang, L. Tang, Y. Huang, et al., Identification of Candidate Genes Associated with Breast Cancer Prognosis, DNA Cell Biol., 39 (2020), 1205-1227. doi: 10.1089/dna.2020.5482
![]() |
[35] |
Y. Chen, W. Chen, X. Dai, C. Zhang, Q. Zhang, J. Lu, Identification of the collagen family as prognostic biomarkers and immune-associated targets in gastric cancer, Int. Immunopharmacol., 87 (2020), 106798. doi: 10.1016/j.intimp.2020.106798
![]() |
[36] |
Y. Wu, Y. Xu, Integrated bioinformatics analysis of expression and gene regulation network of COL12A1 in colorectal cancer, Cancer Med., 9 (2020), 4743-4755. doi: 10.1002/cam4.2899
![]() |
[37] |
Z. Xiang, J. Li, S. Song, J. Wang, W. Cai, W. Hu, et al., A positive feedback between IDO1 metabolite and COL12A1 via MAPK pathway to promote gastric cancer metastasis, J. Exp. Clin. Cancer Res., 38 (2019), 314. doi: 10.1186/s13046-019-1318-5
![]() |
[38] |
R. Januchowski, M. Świerczewska, K. Sterzyńska, K. Wojtowicz, M. Nowicki, M. Zabel, Increased Expression of Several Collagen Genes is Associated with Drug Resistance in Ovarian Cancer Cell Lines, J. Cancer, 7 (2016), 1295-1310. doi: 10.7150/jca.15371
![]() |
[39] |
D. Öhlund, O. Franklin, E. Lundberg, C. Lundin, M. Sund, Type Ⅳ collagen stimulates pancreatic cancer cell proliferation, migration, and inhibits apoptosis through an autocrine loop, BMC Cancer, 13 (2013), 154. doi: 10.1186/1471-2407-13-154
![]() |
[40] |
M. A. Shields, S. Dangi-Garimella, S. B. Krantz, D. J. Bentrem, H. G. Munshi, Pancreatic cancer cells respond to type I collagen by inducing snail expression to promote membrane type 1 matrix metalloproteinase-dependent collagen invasion, J. Biol. Chem., 286 (2011), 10495-10504. doi: 10.1074/jbc.M110.195628
![]() |
[41] |
A. Habtezion, M. Edderkaoui, S.J. Pandol, Macrophages and pancreatic ductal adenocarcinoma, Cancer Lett., 381 (2016), 211-216. doi: 10.1016/j.canlet.2015.11.049
![]() |
[42] |
M. Yu, R. Guan, W. Hong, Y. Zhou, Y. Lin, H. Jin, et al., Prognostic value of tumorassociated macrophages in pancreatic cancer: a meta-analysis, Cancer Manag. Res., 11 (2019), 4041-4058. doi: 10.2147/CMAR.S196951
![]() |
[43] |
A. Ocana, C. Nieto-Jiménez, A. Pandiella, A. J. Templeton, Neutrophils in cancer: prognostic role and therapeutic strategies, Mol. Cancer, 16 (2017), 137. doi: 10.1186/s12943-017-0707-7
![]() |
[44] |
A. Deicher, R. Andersson, B. Tingstedt, G. Lindell, M. Bauden, D. Ansari, Targeting dendritic cells in pancreatic ductal adenocarcinoma, Cancer Cell Int., 18 (2018), 85. doi: 10.1186/s12935-018-0585-0
![]() |
[45] |
C. Yang, H. Cheng, Y. Zhang, K. Fan, G. Luo, Z. Fan, et al., Anergic natural killer cells educated by tumor cells are associated with a poor prognosis in patients with advanced pancreatic ductal adenocarcinoma, Cancer Immunol. Immunother., 67 (2018), 1815-1823. doi: 10.1007/s00262-018-2235-8
![]() |
[46] |
S. Quintero-Fabián, R. Arreola, E. Becerril-Villanueva, J.C. Torres-Romero, V. AranaArgáez, J. Lara-Riegos, et al., Role of Matrix Metalloproteinases in Angiogenesis and Cancer, Front. Oncol., 9 (2019), 1370. doi: 10.3389/fonc.2019.01370
![]() |
[47] |
R. Shimizu-Hirota, W. Xiong, B. T. Baxter, S. L. Kunkel, I. Maillard, X.W. Chen, et al., MT1-MMP regulates the PI3Kδ·Mi-2/NuRD-dependent control of macrophage immune function, Genes Dev., 26 (2012), 395-413. doi: 10.1101/gad.178749.111
![]() |
[48] |
A. M. H. Larsen, D. E. Kuczek, A. Kalvisa, M. S. Siersbæk, M. L. Thorseth, A. Z. Johansen, et al., Collagen Density Modulates the Immunosuppressive Functions of Macrophages, J. Immunol., 205 (2020), 1461-1472. doi: 10.4049/jimmunol.1900789
![]() |
[49] |
D. E. Kuczek, A. M. H. Larsen, M. L. Thorseth, M. Carretta, A. Kalvisa, M. S. Siersbæk, et al., Collagen density regulates the activity of tumor-infiltrating T cells, J. Immunother. Cancer, 7 (2019), 68. doi: 10.1186/s40425-019-0556-6
![]() |
[50] |
E. L. Hopewell, C. Cox, S. Pilon-Thomas, L. L. Kelley, Tumor-infiltrating lymphocytes: Streamlining a complex manufacturing process, Cytotherapy, 21 (2019), 307-314. doi: 10.1016/j.jcyt.2018.11.004
![]() |
[51] |
H. Du, K. Hirabayashi, S. Ahn, N. P. Kren, S. A. Montgomery, X. Wang, et al., Antitumor Responses in the Absence of Toxicity in Solid Tumors by Targeting B7-H3 via Chimeric Antigen Receptor T Cells, Cancer Cell, 35 (2019), 221-237. doi: 10.1016/j.ccell.2019.01.002
![]() |
[52] |
J. Jacobs, V. Deschoolmeester, K. Zwaenepoel, C. Rolfo, K. Silence, S. Rottey, et al., CD70: An emerging target in cancer immunotherapy, Pharmacol. Ther., 155 (2015), 1-10. doi: 10.1016/j.pharmthera.2015.07.007
![]() |
[53] |
P. Yin, L. Gui, C. Wang, J. Yan, M. Liu, L. Ji, et al., Targeted delivery of CXCL9 and OX40L by mesenchymal stem cells elicits potent antitumor immunity, Mol. Ther., 28 (2020), 2553-2563. doi: 10.1016/j.ymthe.2020.08.005
![]() |
[54] |
J. Wu, Y. Wang, Z. Jiang, Immune induction identified by TMT proteomics analysis in autoinducer-2 treated macrophages, Expert Rev. Proteomics, 17 (2020), 175-185. doi: 10.1080/14789450.2020.1738223
![]() |
[55] |
C. Liang, J. Xu, Q. Meng, B. Zhang, J. Liu, J. Hua, et al., TGFB1-induced autophagy affects the pattern of pancreatic cancer progression in distinct ways depending on SMAD4 status, Autophagy, 16 (2020), 486-500. doi: 10.1080/15548627.2019.1628540
![]() |
[56] |
K. C. Ohaegbulam, A. Assal, E. Lazar-Molnar, Y. Yao, X. Zang, Human cancer immunotherapy with antibodies to the PD-1 and PD-L1 pathway, Trends Mol. Med., 21 (2015), 24-33. doi: 10.1016/j.molmed.2014.10.009
![]() |
[57] |
S. S. Potter, Single-cell RNA sequencing for the study of development, physiology and disease, Nat. Rev. Nephrol., 14 (2018), 479-492. doi: 10.1038/s41581-018-0021-7
![]() |
[58] |
J. Cheng, J. Zhang, Z. Wu, X. Sun, Inferring microenvironmental regulation of gene expression from single-cell RNA sequencing data using scMLnet with an application to COVID-19, Brief. Bioinform., 22 (2021), 988-1005. doi: 10.1093/bib/bbaa327
![]() |
[59] |
J. Zhang, M. Guan, Q. Wang, J. Zhang, T. Zhou, X. Sun, Single-cell transcriptome-based multilayer network biomarker for predicting prognosis and therapeutic response of gliomas, Brief. Bioinform., 21 (2020), 1080-1097. doi: 10.1093/bib/bbz040
![]() |
[60] |
J. Han, R. A. DePinho, A. Maitra, Single-cell RNA sequencing in pancreatic cancer, Nat. Rev. Gastroenterol. Hepatol., 18 (2021), 451-452. doi: 10.1038/s41575-021-00471-z
![]() |
[61] |
Q. Luo, Q. Fu, X. Zhang, H. Zhang, T. Qin, Application of Single-Cell RNA Sequencing in Pancreatic Cancer and the Endocrine Pancreas, Adv. Exp. Med. Biol., 1255 (2020), 143-152. doi: 10.1007/978-981-15-4494-1_12
![]() |
1. | Chun-Ye Zhang, Shuai Liu, Ming Yang, Clinical diagnosis and management of pancreatic cancer: Markers, molecular mechanisms, and treatment options, 2022, 28, 1007-9327, 6827, 10.3748/wjg.v28.i48.6827 | |
2. | Jinbao Zhang, Meiling Chen, Chuanfa Fang, Peng Luo, A cancer-associated fibroblast gene signature predicts prognosis and therapy response in patients with pancreatic cancer, 2022, 12, 2234-943X, 10.3389/fonc.2022.1052132 | |
3. | Gomathi Venkatasubramanian, Devaki A. Kelkar, Susmita Mandal, Mohit Kumar Jolly, Madhura Kulkarni, Analysis of Yes-Associated Protein-1 (YAP1) Target Gene Signature to Predict Progressive Breast Cancer, 2022, 11, 2077-0383, 1947, 10.3390/jcm11071947 | |
4. | Huan Wu, Hanchu Wang, Zhenyou Jiang, Yue Chen, Identification of Three Core Secretome Genes Associated with Immune Infiltration in High Tumor Mutation Burden Across 14 Major Solid Tumors, 2021, Volume 14, 1178-7074, 6755, 10.2147/IJGM.S333141 | |
5. | Robert J. Rabelo-Fernández, Ricardo A. Noriega Rivera, Yasmarie Santana Rivera, José Tous-Beveraggi, Fatima Valiyeva, Pablo E. Vivas-Mejia, Increased Expression of the RBPMS Splice Variants Inhibits Cell Proliferation in Ovarian Cancer Cells, 2022, 23, 1422-0067, 14742, 10.3390/ijms232314742 | |
6. | Zengwei Tang, Yuan Yang, Qi Zhang, Tingbo Liang, Epigenetic dysregulation-mediated COL12A1 upregulation predicts worse outcome in intrahepatic cholangiocarcinoma patients, 2023, 15, 1868-7083, 10.1186/s13148-022-01413-5 | |
7. | Yao Song, Lei Wang, Kaidong Wang, Yuhua Lu, Pengcheng Zhou, COL12A1 Acts as a Novel Prognosis Biomarker and Activates Cancer-Associated Fibroblasts in Pancreatic Cancer through Bioinformatics and Experimental Validation, 2023, 15, 2072-6694, 1480, 10.3390/cancers15051480 | |
8. | Xuezhou Zhang, Baoan Hong, Hongwei Li, Jiahui Zhao, Mingchuan Li, Dechao Wei, Yongxing Wang, Ning Zhang, Basement membrane-related MMP14 predicts poor prognosis and response to immunotherapy in bladder cancer, 2024, 24, 1471-2407, 10.1186/s12885-024-12489-y | |
9. | Yimin Guo, Shuwen Hua, Baichun Wang, Bing Wang, Chuan-Fan Ding, Yinghua Yan, In situ grown magnetic COF@MOF with a phosphoserine anchor for in-depth N-glycopeptide analysis in serum, 2023, 148, 0003-2654, 5864, 10.1039/D3AN01473H | |
10. | Marina Crespo-Bravo, Annika Hettich, Jeppe Thorlacius-Ussing, Thomas R Cox, Morten A. Karsdal, Nicholas Willumsen, Type XII collagen is elevated in serum from patients with solid tumors: a non-invasive biomarker of activated fibroblasts, 2024, 24, 1591-9528, 10.1007/s10238-024-01431-y | |
11. | Hui Wan, Musu Yuan, Yiwei Fu, Minghua Deng, Continually adapting pre-trained language model to universal annotation of single-cell RNA-seq data, 2024, 25, 1467-5463, 10.1093/bib/bbae047 | |
12. | D. Sinkeviciute, Helene W. Breisnes, A.C. Bay-Jensen, M.A. Karsdal, T. Manon-Jensen, J.H. Mortensen, 2024, 9780443156175, 113, 10.1016/B978-0-443-15617-5.00038-X | |
13. | Weijie Yao, Yanrong Yao, Wen He, Chengsi Zhao, Di Liu, Genwang Wang, Zuozheng Wang, PABPC1 promotes cell proliferation and metastasis in pancreatic adenocarcinoma by regulating COL12A1 expression, 2023, 11, 2050-4527, 10.1002/iid3.919 | |
14. | Zhang Shen, Mian Huang, Jun Lin, Sanshan Wu, Qiong Jin, Inhibitory effect on endometrial cancer: Collagen type XII α1 chain, 2025, 22, 1742-6413, 31, 10.25259/Cytojournal_236_2024 | |
15. | Peng An, Junhong Wang, Rong Fan, Identifying and validating PLAU as a potential prognostic biomarker for PDAC, 2025, 15, 2045-2322, 10.1038/s41598-025-97629-5 | |
16. | Na Li, Nan Zhang, Guanghui Wang, Overexpression of MMP14 is associated with poor prognosis and immune cell infiltration in colon cancer, 2025, 15, 2234-943X, 10.3389/fonc.2025.1564375 | |
17. | Antonella Fanelli, Eugenio Mazzone, Diana Giannuzzi, Laura Marconato, Luca Aresu, Expanding the Spectrum of Canine Diffuse Large B‐Cell Lymphoma Genetic Aberrations Through Whole Genome Sequencing Analysis, 2025, 1476-5810, 10.1111/vco.13059 |
Parameters | Overall(n = 178) | COL12A1 expression | P | MMP14 expression | P | ||
low (89) | high (89) | high (89) | low (89) | ||||
Primary site (%) | 0.262 | 0.295 | |||||
Body of Pancreas | 14 (7.9) | 9 (10.1) | 5 (5.6) | 9 (10.1) | 5 (5.6) | ||
Head of Pancreas | 139 (78.1) | 65 (73.0) | 74 (83.1) | 69 (77.5) | 70 (78.7) | ||
Tail of Pancreas | 14 (7.9) | 7 (7.9) | 7 (7.9) | 8 (9.0) | 6 (6.7) | ||
Other | 11 (6.2) | 8 (9.0) | 3 (3.4) | 3 (3.4) | 8 (9.0) | ||
Pathological type (%) | 0.094 | 0.094 | |||||
Other Type | 25 (14.1) | 18 (20.2) | 7 (8.0) | 7 (8.0) | 18 (20.2) | ||
Ductal Type | 147 (83.1) | 69 (77.5) | 78 (88.6) | 78 (88.6) | 69 (77.5) | ||
Colloid Carcinoma | 4 (2.3) | 2 (2.2) | 2 (2.3) | 2 (2.3) | 2 (2.2) | ||
Undifferentiated Carcinoma | 1 (0.6) | 0 (0.0) | 1 (1.1) | 1 (1.1) | 0 (0.0) | ||
Grade (%) | 0.044 | 0.018 | |||||
G1 | 31 (17.4) | 22 (24.7) | 9 (10.1) | 8 (9.0) | 23 (25.8) | ||
G2 | 95 (53.4) | 40 (44.9) | 55 (61.8) | 51 (57.3) | 44 (49.4) | ||
G3 | 48 (27.0) | 24 (27.0) | 24 (27.0) | 29 (32.6) | 19 (21.3) | ||
G4 | 2 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | ||
GX | 2 (1.1) | 2 (2.2) | 0 (0.0) | 0 (0.0) | 2 (2.2) | ||
Tumor stage (%) | 0.004 | 0.28 | |||||
Stage Ⅰ | 21 (12.0) | 18 (20.7) | 3 (3.4) | 7 (8.0) | 14 (16.1) | ||
Stage Ⅱ | 147 (84.0) | 66 (75.9) | 81 (92.0) | 76 (86.4) | 71 (81.6) | ||
Stage Ⅲ | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.3) | 1 (1.1) | ||
Stage Ⅳ | 4 (2.3) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
T classification (%) | 0.004 | 0.444 | |||||
T1 | 7 (4.0) | 7 (8.0) | 0 (0.0) | 2 (2.2) | 5 (5.7) | ||
T2 | 24 (13.6) | 17 (19.3) | 7 (7.9) | 10 (11.2) | 14 (15.9) | ||
T3 | 142 (80.2) | 61 (69.3) | 81 (91.0) | 75 (84.3) | 67 (76.1) | ||
T4 | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.2) | 1 (1.1) | ||
TX | 1 (0.6) | 1 (1.1) | 0 (0.0) | 0 (0.0) | 1 (1.1) | ||
N classification (%) | 0.08 | 0.977 | |||||
N0 | 49 (27.7) | 30 (34.1) | 19 (21.3) | 24 (27.0) | 25 (28.4) | ||
N1 | 124 (70.1) | 55 (62.5) | 69 (77.5) | 63 (70.8) | 61 (69.3) | ||
NX | 4 (2.3) | 3 (3.4) | 1 (1.1) | 2 (2.2) | 2 (2.3) | ||
M classification (%) | 0.022 | 0.046 | |||||
M0 | 80 (44.9) | 32 (36.0) | 48 (53.9) | 47 (52.8) | 33 (37.1) | ||
M1 | 4 (2.2) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
MX | 94 (52.8) | 56 (62.9) | 38 (42.7) | (43.8) | (61.8) |
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.76 | 2.25 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.52 | 2.24 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical surface | 0.52 | 1.90 | ≤ 0.001 | 0.04 |
Hallmark-mitotic spindle | 0.47 | 1.83 | 0.01 | 0.06 |
Hallmark-hypoxia | 0.47 | 1.82 | 0.01 | 0.06 |
Hallmark-TGF-β signaling | 0.56 | 1.82 | 0.01 | 0.05 |
Hallmark-angiogenesis | 0.57 | 1.80 | 0.01 | 0.05 |
Hallmark-WNT-β catenin signaling | 0.47 | 1.69 | 0.02 | 0.09 |
Hallmark-apoptosis | 0.41 | 1.69 | 0.01 | 0.08 |
Hallmark-coagulation | 0.40 | 1.65 | 0.01 | 0.08 |
KEGG-ECM receptor interaction | 0.69 | 2.17 | ≤ 0.001 | ≤ 0.001 |
KEGG-focal adhesion | 0.58 | 2.15 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.46 | 1.98 | ≤ 0.001 | 0.03 |
KEGG-basal cell carcinoma | 0.58 | 1.96 | ≤ 0.001 | 0.03 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.71 | 1.92 | ≤ 0.001 | 0.04 |
KEGG-regulation of actin cytoskeleton | 0.44 | 1.91 | ≤ 0.001 | 0.04 |
KEGG-small cell lung cancer | 0.54 | 1.90 | ≤ 0.001 | 0.03 |
KEGG-pathogenic Escherichia coli infection | 0.55 | 1.83 | 0.01 | 0.06 |
KEGG-bladder cancer | 0.54 | 1.83 | 0.01 | 0.06 |
KEGG-axon guidance | 0.43 | 1.75 | ≤0.001 | 0.10 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.78 | 2.41 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.48 | 2.10 | ≤ 0.001 | 0.01 |
Hallmark-UV response DN | 0.58 | 2.09 | ≤ 0.001 | 0.01 |
Hallmark-TGF-β signaling | 0.59 | 1.97 | ≤ 0.001 | 0.02 |
Hallmark-angiogenesis | 0.61 | 1.93 | 0.01 | 0.02 |
Hallmark-inflammatory response | 0.53 | 1.86 | 0.01 | 0.04 |
Hallmark-coagulation | 0.44 | 1.78 | ≤ 0.001 | 0.05 |
Hallmark-apoptosis | 0.42 | 1.78 | 0.01 | 0.05 |
Hallmark-complement | 0.45 | 1.76 | 0.01 | 0.05 |
Hallmark-notch signaling | 0.52 | 1.70 | 0.02 | 0.07 |
KEGG-focal adhesion | 0.64 | 2.33 | ≤ 0.001 | ≤ 0.001 |
KEGG-ECM receptor interaction | 0.71 | 2.20 | ≤ 0.001 | ≤ 0.001 |
KEGG-regulation of actin cytoskeleton | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-TGF-β signaling | 0.55 | 2.11 | ≤ 0.001 | ≤ 0.001 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.77 | 2.10 | ≤ 0.001 | ≤ 0.001 |
KEGG- small cell lung cancer | 0.56 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- basal cell carcinoma | 0.57 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- melanoma | 0.48 | 1.88 | ≤ 0.001 | 0.04 |
KEGG- adherens junction | 0.53 | 1.87 | 0.01 | 0.04 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |
Parameters | Overall(n = 178) | COL12A1 expression | P | MMP14 expression | P | ||
low (89) | high (89) | high (89) | low (89) | ||||
Primary site (%) | 0.262 | 0.295 | |||||
Body of Pancreas | 14 (7.9) | 9 (10.1) | 5 (5.6) | 9 (10.1) | 5 (5.6) | ||
Head of Pancreas | 139 (78.1) | 65 (73.0) | 74 (83.1) | 69 (77.5) | 70 (78.7) | ||
Tail of Pancreas | 14 (7.9) | 7 (7.9) | 7 (7.9) | 8 (9.0) | 6 (6.7) | ||
Other | 11 (6.2) | 8 (9.0) | 3 (3.4) | 3 (3.4) | 8 (9.0) | ||
Pathological type (%) | 0.094 | 0.094 | |||||
Other Type | 25 (14.1) | 18 (20.2) | 7 (8.0) | 7 (8.0) | 18 (20.2) | ||
Ductal Type | 147 (83.1) | 69 (77.5) | 78 (88.6) | 78 (88.6) | 69 (77.5) | ||
Colloid Carcinoma | 4 (2.3) | 2 (2.2) | 2 (2.3) | 2 (2.3) | 2 (2.2) | ||
Undifferentiated Carcinoma | 1 (0.6) | 0 (0.0) | 1 (1.1) | 1 (1.1) | 0 (0.0) | ||
Grade (%) | 0.044 | 0.018 | |||||
G1 | 31 (17.4) | 22 (24.7) | 9 (10.1) | 8 (9.0) | 23 (25.8) | ||
G2 | 95 (53.4) | 40 (44.9) | 55 (61.8) | 51 (57.3) | 44 (49.4) | ||
G3 | 48 (27.0) | 24 (27.0) | 24 (27.0) | 29 (32.6) | 19 (21.3) | ||
G4 | 2 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | 1 (1.1) | ||
GX | 2 (1.1) | 2 (2.2) | 0 (0.0) | 0 (0.0) | 2 (2.2) | ||
Tumor stage (%) | 0.004 | 0.28 | |||||
Stage Ⅰ | 21 (12.0) | 18 (20.7) | 3 (3.4) | 7 (8.0) | 14 (16.1) | ||
Stage Ⅱ | 147 (84.0) | 66 (75.9) | 81 (92.0) | 76 (86.4) | 71 (81.6) | ||
Stage Ⅲ | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.3) | 1 (1.1) | ||
Stage Ⅳ | 4 (2.3) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
T classification (%) | 0.004 | 0.444 | |||||
T1 | 7 (4.0) | 7 (8.0) | 0 (0.0) | 2 (2.2) | 5 (5.7) | ||
T2 | 24 (13.6) | 17 (19.3) | 7 (7.9) | 10 (11.2) | 14 (15.9) | ||
T3 | 142 (80.2) | 61 (69.3) | 81 (91.0) | 75 (84.3) | 67 (76.1) | ||
T4 | 3 (1.7) | 2 (2.3) | 1 (1.1) | 2 (2.2) | 1 (1.1) | ||
TX | 1 (0.6) | 1 (1.1) | 0 (0.0) | 0 (0.0) | 1 (1.1) | ||
N classification (%) | 0.08 | 0.977 | |||||
N0 | 49 (27.7) | 30 (34.1) | 19 (21.3) | 24 (27.0) | 25 (28.4) | ||
N1 | 124 (70.1) | 55 (62.5) | 69 (77.5) | 63 (70.8) | 61 (69.3) | ||
NX | 4 (2.3) | 3 (3.4) | 1 (1.1) | 2 (2.2) | 2 (2.3) | ||
M classification (%) | 0.022 | 0.046 | |||||
M0 | 80 (44.9) | 32 (36.0) | 48 (53.9) | 47 (52.8) | 33 (37.1) | ||
M1 | 4 (2.2) | 1 (1.1) | 3 (3.4) | 3 (3.4) | 1 (1.1) | ||
MX | 94 (52.8) | 56 (62.9) | 38 (42.7) | (43.8) | (61.8) |
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.76 | 2.25 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.52 | 2.24 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical surface | 0.52 | 1.90 | ≤ 0.001 | 0.04 |
Hallmark-mitotic spindle | 0.47 | 1.83 | 0.01 | 0.06 |
Hallmark-hypoxia | 0.47 | 1.82 | 0.01 | 0.06 |
Hallmark-TGF-β signaling | 0.56 | 1.82 | 0.01 | 0.05 |
Hallmark-angiogenesis | 0.57 | 1.80 | 0.01 | 0.05 |
Hallmark-WNT-β catenin signaling | 0.47 | 1.69 | 0.02 | 0.09 |
Hallmark-apoptosis | 0.41 | 1.69 | 0.01 | 0.08 |
Hallmark-coagulation | 0.40 | 1.65 | 0.01 | 0.08 |
KEGG-ECM receptor interaction | 0.69 | 2.17 | ≤ 0.001 | ≤ 0.001 |
KEGG-focal adhesion | 0.58 | 2.15 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.46 | 1.98 | ≤ 0.001 | 0.03 |
KEGG-basal cell carcinoma | 0.58 | 1.96 | ≤ 0.001 | 0.03 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.71 | 1.92 | ≤ 0.001 | 0.04 |
KEGG-regulation of actin cytoskeleton | 0.44 | 1.91 | ≤ 0.001 | 0.04 |
KEGG-small cell lung cancer | 0.54 | 1.90 | ≤ 0.001 | 0.03 |
KEGG-pathogenic Escherichia coli infection | 0.55 | 1.83 | 0.01 | 0.06 |
KEGG-bladder cancer | 0.54 | 1.83 | 0.01 | 0.06 |
KEGG-axon guidance | 0.43 | 1.75 | ≤0.001 | 0.10 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |
Name | ES | NES | NOM p-value | FDR q-value |
Hallmark-epithelial mesenchymal transition | 0.78 | 2.41 | ≤ 0.001 | ≤ 0.001 |
Hallmark-apical junction | 0.48 | 2.10 | ≤ 0.001 | 0.01 |
Hallmark-UV response DN | 0.58 | 2.09 | ≤ 0.001 | 0.01 |
Hallmark-TGF-β signaling | 0.59 | 1.97 | ≤ 0.001 | 0.02 |
Hallmark-angiogenesis | 0.61 | 1.93 | 0.01 | 0.02 |
Hallmark-inflammatory response | 0.53 | 1.86 | 0.01 | 0.04 |
Hallmark-coagulation | 0.44 | 1.78 | ≤ 0.001 | 0.05 |
Hallmark-apoptosis | 0.42 | 1.78 | 0.01 | 0.05 |
Hallmark-complement | 0.45 | 1.76 | 0.01 | 0.05 |
Hallmark-notch signaling | 0.52 | 1.70 | 0.02 | 0.07 |
KEGG-focal adhesion | 0.64 | 2.33 | ≤ 0.001 | ≤ 0.001 |
KEGG-ECM receptor interaction | 0.71 | 2.20 | ≤ 0.001 | ≤ 0.001 |
KEGG-regulation of actin cytoskeleton | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-pathway in cancer | 0.50 | 2.19 | ≤ 0.001 | ≤ 0.001 |
KEGG-TGF-β signaling | 0.55 | 2.11 | ≤ 0.001 | ≤ 0.001 |
KEGG-glycosaminoglycan biosynthesis chondroitin sulfate | 0.77 | 2.10 | ≤ 0.001 | ≤ 0.001 |
KEGG- small cell lung cancer | 0.56 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- basal cell carcinoma | 0.57 | 1.99 | ≤ 0.001 | 0.01 |
KEGG- melanoma | 0.48 | 1.88 | ≤ 0.001 | 0.04 |
KEGG- adherens junction | 0.53 | 1.87 | 0.01 | 0.04 |
*Note: ES: enrichment score; NES: normalized enrichment score; NOM: nominal. FDR: false discovery rate. |