scholarly journals A Novel DNA Replication-Related Signature Predicting Recurrence After R0 Resection of Pancreatic Ductal Adenocarcinoma: Prognostic Value and Clinical Implications

Author(s):  
Zengyu Feng ◽  
Kexian Li ◽  
Jianyao Lou ◽  
Mindi Ma ◽  
Yulian Wu ◽  
...  

The aim of any surgical resection for pancreatic ductal adenocarcinoma (PDAC) is to achieve tumor-free margins (R0). R0 margins give rise to better outcomes than do positive margins (R1). Nevertheless, postoperative morbidity after R0 resection remains high and prognostic gene signature predicting recurrence risk of patients in this subgroup is blank. Our study aimed to develop a DNA replication-related gene signature to stratify the R0-treated PDAC patients with various recurrence risks. We conducted Cox regression analysis and the LASSO algorithm on 273 DNA replication-related genes and eventually constructed a 7-gene signature. The predictive capability and clinical feasibility of this risk model were assessed in both training and external validation sets. Pathway enrichment analysis showed that the signature was closely related to cell cycle, DNA replication, and DNA repair. These findings may shed light on the identification of novel biomarkers and therapeutic targets for PDAC.

2021 ◽  
Vol 8 ◽  
Author(s):  
Zengyu Feng ◽  
Peng Chen ◽  
Kexian Li ◽  
Jianyao Lou ◽  
Yulian Wu ◽  
...  

Background: Recurrence after surgery is largely responsible for the extremely poor outcomes for patients with pancreatic ductal adenocarcinoma (PDAC). Ferroptosis is implicated in chemotherapy sensitivity and tumor recurrence, we aimed to find out survival-associated ferroptosis-related genes and use them to build a practical risk model with the purpose to predict PDAC recurrence.Methods: Univariate Cox regression analysis was conducted to obtain prognostic ferroptosis-related genes in The Cancer Genome Atlas (TCGA, N = 140) cohort. Multivariate Cox regression analysis was employed to construct a reliable and credible gene signature. The prognostic performance was verified in a MTAB-6134 (N = 286) validation cohort and a PACA-CA (N = 181) validation cohort. The stability of the signature was tested in TCGA and MTAB-6134 cohorts by ROC analyses. Pathway enrichment analysis was adopted to preliminary illuminate the biological relevance of the gene signature.Results: Univariate and multivariate Cox regression analyses identified a 5-gene signature that contained CAV1, DDIT4, SLC40A1, SRXN1 and TFAP2C. The signature could efficaciously stratify PDAC patients with different recurrence-free survival (RFS), both in the training and validation cohorts. Results of subgroup receiver operating characteristic curve (ROC) analyses confirmed the stability and the independence of this signature. Our signature outperformed clinical indicators and previous reported models in predicting RFS. Moreover, the signature was found to be closely associated with several cancer-related and drug response pathways.Conclusion: This study developed a precise and concise prognostic model with the clinical implication in predicting PDAC recurrence. These findings may facilitate individual management of postoperative recurrence in patients with PDAC.


2021 ◽  
Vol 15 ◽  
pp. 117955492110241
Author(s):  
Hongkai Zhuang ◽  
Zixuan Zhou ◽  
Zuyi Ma ◽  
Shanzhou Huang ◽  
Yuanfeng Gong ◽  
...  

Background: The prognosis of patients with pancreatic ductal adenocarcinoma (PDAC) of pancreatic head remains poor, even after potentially curative R0 resection. The aim of this study was to develop an accurate model to predict patients’ prognosis for PDAC of pancreatic head following pancreaticoduodenectomy. Methods: We retrospectively reviewed 112 patients with PDAC of pancreatic head after pancreaticoduodenectomy in Guangdong Provincial People’s Hospital between 2014 and 2018. Results: Five prognostic factors were identified using univariate Cox regression analysis, including age, histologic grade, American Joint Committee on Cancer (AJCC) Stage 8th, total bilirubin (TBIL), CA19-9. Using all subset analysis and multivariate Cox regression analysis, we developed a nomogram consisted of age, AJCC Stage 8th, perineural invasion, TBIL, and CA19-9, which had higher C-indexes for OS (0.73) and RFS (0.69) compared with AJCC Stage 8th alone (OS: 0.66; RFS: 0.67). The area under the curve (AUC) values of the receiver operating characteristic (ROC) curve for the nomogram for OS and RFS were significantly higher than other single parameter, which are AJCC Stage 8th, age, perineural invasion, TBIL, and CA19-9. Importantly, our nomogram displayed higher C-index for OS than previous reported models, indicating a better predictive value of our model. Conclusions: A simple and practical nomogram for patient prognosis in PDAC of pancreatic head following pancreaticoduodenectomy was established, which shows satisfactory predictive efficacy and deserves further evaluation in the future.


2021 ◽  
Vol 8 ◽  
Author(s):  
Zengyu Feng ◽  
Hao Qian ◽  
Kexian Li ◽  
Jianyao Lou ◽  
Yulian Wu ◽  
...  

Background: Previous prognostic signatures of pancreatic ductal adenocarcinoma (PDAC) are mainly constructed to predict the overall survival (OS), and their predictive accuracy needs to be improved. Gene signatures that efficaciously predict both OS and disease-free survival (DFS) are of great clinical significance but are rarely reported.Methods: Univariate Cox regression analysis was adopted to screen common genes that were significantly associated with both OS and DFS in three independent cohorts. Multivariate Cox regression analysis was subsequently performed on the identified genes to determine an optimal gene signature in the MTAB-6134 training cohort. The Kaplan–Meier (K-M), calibration, and receiver operating characteristic (ROC) curves were employed to assess the predictive accuracy. Biological process and pathway enrichment analyses were conducted to elucidate the biological role of this signature.Results: Multivariate Cox regression analysis determined a 7-gene signature that contained ASPH, DDX10, NR0B2, BLOC1S3, FAM83A, SLAMF6, and PPM1H. The signature had the ability to stratify PDAC patients with different OS and DFS, both in the training and validation cohorts. ROC curves confirmed the moderate predictive accuracy of this signature. Mechanically, the signature was related to multiple cancer-related pathways.Conclusion: A novel OS and DFS prediction model was constructed in PDAC with multi-cohort and cross-platform compatibility. This signature might foster individualized therapy and appropriate management of PDAC patients.


Author(s):  
Zengyu Feng ◽  
Kexian Li ◽  
Jianyao Lou ◽  
Yulian Wu ◽  
Chenghong Peng

BackgroundFor pancreatic ductal adenocarcinoma (PDAC) patients, chemotherapy failure is the major reason for postoperative recurrence and poor outcomes. Establishment of novel biomarkers and models for predicting chemotherapeutic efficacy may provide survival benefits by tailoring treatments.MethodsUnivariate cox regression analysis was employed to identify EMT-related genes with prognostic potential for DFS. These genes were subsequently submitted to LASSO regression analysis and multivariate cox regression analysis to identify an optimal gene signature in TCGA training cohort. The predictive accuracy was assessed by Kaplan–Meier (K-M), receiver operating characteristic (ROC) and calibration curves and was validated in PACA-CA cohort and our local cohort. Pathway enrichment and function annotation analyses were conducted to illuminate the biological implication of this risk signature.ResultsLASSO and multivariate Cox regression analyses selected an 8-gene signature comprised DLX2, FGF9, IL6R, ITGB6, MYC, LGR5, S100A2, and TNFSF12. The signature had the capability to classify PDAC patients with different DFS, both in the training and validation cohorts. It provided improved DFS prediction compared with clinical indicators. This signature was associated with several cancer-related pathways. In addition, the signature could also predict the response to immune-checkpoint inhibitors (ICIs)-based immunotherapy.ConclusionWe established a novel EMT-related gene signature that was capable of predicting therapeutic response to adjuvant chemotherapy and immunotherapy. This signature might facilitate individualized treatment and appropriate management of PDAC patients.


2020 ◽  
Vol 40 (7) ◽  
Author(s):  
Xin Zhao ◽  
Di Cao ◽  
Zhangyong Ren ◽  
Zhe Liu ◽  
Shaocheng Lv ◽  
...  

Abstract Background: Hypermethylation of gene promoters plays an important role in tumorigenesis. The present study aimed to identify and validate promoter methylation-driven genes (PMDGs) for pancreatic ductal adenocarcinoma (PDAC). Methods: Based on GSE49149 and the PDAC cohort of The Cancer Genome Atlas (TCGA), differential analyses of promoter methylation, correlation analysis, and Cox regression analysis were performed to identify PMDGs. The promoter methylation level was assessed by bisulfite sequencing polymerase chain reaction (BSP) in paired tumor and normal tissues of 72 PDAC patients. Kaplan−Meier survival analyses were performed to evaluate the clinical value of PMDGs. Results: In GSE49149, the β-value of the dipeptidyl peptidase like 6 (DPP6) promoter was significantly higher in tumor compared with normal samples (0.50 vs. 0.24, P<0.001). In the PDAC cohort of TCGA, the methylation level of the DPP6 promoter was negatively correlated with mRNA expression (r = −0.54, P<0.001). In a multivariate Cox regression analysis, hypermethylation of the DPP6 promoter was an independent risk factor for PDAC (hazard ratio (HR) = 543.91, P=0.002). The results of BSP revealed that the number of methylated CG sites in the DPP6 promoter was greater in tumor samples than in normal samples (7.43 vs. 2.78, P<0.001). The methylation level of the DPP6 promoter was moderately effective at distinguishing tumor from normal samples (area under ROC curve (AUC) = 0.74, P<0.001). Hypermethylation of the DPP6 promoter was associated with poor overall (HR = 3.61, P<0.001) and disease-free (HR = 2.01, P=0.016) survivals for PDAC patients. Conclusion: These results indicate that DPP6 promoter methylation is a potential prognostic biomarker for PDAC.


2021 ◽  
Vol 11 ◽  
Author(s):  
Zhen Tan ◽  
Yubin Lei ◽  
Bo Zhang ◽  
Si Shi ◽  
Jiang Liu ◽  
...  

BackgroundPancreatic ductal adenocarcinoma (PDAC) is one of the most invasive solid malignancies. Immunotherapy and targeted therapy confirmed an existing certain curative effect in treating PDAC. The aim of this study was to develop an immune-related molecular marker to enhance the ability to predict Stages III and IV PDAC patients.MethodIn this study, weighted gene co-expression network (WGCNA) analysis and a deconvolution algorithm (CIBERSORT) that evaluated the cellular constituent of immune cells were used to evaluate PDAC expression data from the GEO (Gene Expression Omnibus) datasets, and identify modules related to CD4+ T cells. LASSO Cox regression analysis and Kaplan–Meier curve were applied to select and build prognostic multi-gene signature in TCGA Stages III and IV PDAC patients (N = 126). This was followed by independent Stages III and IV validation of the gene signature in the International Cancer Genome Consortium (ICGC, N = 62) and the Fudan University Shanghai Cancer Center (FUSCC, N = 42) cohort. Inherited germline mutations and tumor immunity exploration were applied to elucidate the molecular mechanisms in PDAC. Univariate and Multivariate Cox regression analyses were applied to verify the independent prognostic factors. Finally, a prognostic nomogram was created according to the TCGA-PDAC dataset.ResultsA four-gene signature comprising NAPSB, ZNF831, CXCL9 and PYHIN1 was established to predict overall survival of PDAC. This signature also robustly predicted survival in two independent validation cohorts. The four-gene signature could divide patients into high and low-risk groups with disparity overall survival verified by a Log-rank test. Expression of four genes positively correlated with immunosuppression activity (PD-L1 and PD1). Immune-related genes nomogram and corresponding calibration curves showed significant performance for predicting 3-year survival in TCGA-PDAC dataset.ConclusionWe constructed a novel four-gene signature to predict the prognosis of Stages III and IV PDAC patients by applying WGCNA and CIBERSORT algorithm scoring to transcriptome data different from traditional methods of filtrating for differential genes in cancer and healthy tissues. The findings may provide reference to predict survival and was beneficial to individualized management for advanced PDAC patients.


2021 ◽  
Author(s):  
Boxuan Liu ◽  
Yun Zhao ◽  
Shuanying Yang

Abstract Background: Lung adenocarcinoma is the most occurred pathological type among non-small cell lung cancer. Although huge progress has been made in terms of early diagnosis, precision treatment in recent years, the overall 5-year survival rate of a patient remains low. In our study, we try to construct an autophagy-related lncRNA prognostic signature that may guide clinical practice.Methods: The mRNA and lncRNA expression matrix of lung adenocarcinoma patients were retrieved from TCGA database. Next, we constructed a co-expression network of lncRNAs and autophagy-related genes. Lasso regression and multivariate Cox regression were then applied to establish a prognostic risk model. Subsequently, a risk score was generated to differentiate high and low risk group and a ROC curve and Nomogram to visualize the predictive ability of current signature. Finally, gene ontology and pathway enrichment analysis were executed via GSEA.Results: A total of 1,703 autophagy-related lncRNAs were screened and five autophagy-related lncRNAs (LINC01137, AL691432.2, LINC01116, AL606489.1 and HLA-DQB1-AS1) were finally included in our signature. Judging from univariate(HR=1.075, 95% CI: 1.046–1.104) and multivariate(HR =1.088, 95%CI = 1.057 − 1.120) Cox regression analysis, the risk score is an independent factor for LUAD patients. Further, the AUC value based on the risk score for 1-year, 3-year, 5-year, was 0.735, 0.672 and 0.662 respectively. Finally, the lncRNAs included in our signature were primarily enriched in autophagy process, metabolism, p53 pathway and JAK/STAT pathway. Conclusions: Overall, our study indicated that the prognostic model we generated had certain predictability for LUAD patients’ prognosis.


2021 ◽  
Vol 8 ◽  
Author(s):  
Zengyu Feng ◽  
Kexian Li ◽  
Yulian Wu ◽  
Chenghong Peng

Background: Accumulating evidence shows that the elevated expression of DCBLD2 (discoidin, CUB and LCCL domain-containing protein 2) is associated with unfavorable prognosis of various cancers. However, the correlation of DCBLD2 expression value with the diagnosis and prognosis of pancreatic ductal adenocarcinoma (PDAC) has not yet been elucidated. Methods: Univariate Cox regression analysis was used to screen robust survival-related genes. Expression pattern of selected genes was investigated in PDAC tissues and normal tissues from multiple cohorts. Kaplan–Meier (K–M) survival curves, ROC curves and calibration curves were employed to assess prognostic performance. The relationship between DCBLD2 expression and immune cell infiltrates was conducted by CIBERSORT software. Biological processes and KEGG pathway enrichment analyses were adopted to clarify the potential function of DCBLD2 in PDAC. Results: Univariate analysis, K–M survival curves and calibration curves indicated that DCBLD2 was a robust prognostic factor for PDAC with cross-cohort compatibility. Upregulation of DCBLD2 was observed in dissected PDAC tissues as well as extracellular vesicles from both plasma and serum samples of PDAC patients. Both DCBLD2 expression in tissue and extracellular vesicles had significant diagnostic value. Besides, DCBLD2 expression was correlated with infiltrating level of CD8+ T cells and macrophage M2 cells. Functional enrichment revealed that DCBLD2 might be involved in cell motility, angiogenesis, and cancer-associated pathways. Conclusion: Our study systematically analyzed the potential diagnostic, prognostic and therapeutic value of DCBLD2 in PDAC. All the findings indicated that DCBLD2 might play a considerably oncogenic role in PDAC with diagnostic, prognostic and therapeutic potential. These preliminary results of bioinformatics analyses need to be further validated in more prospective studies.


BMC Cancer ◽  
2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Hongjun Fei ◽  
Songchang Chen ◽  
Chenming Xu

Abstract Background Little data is available on prognostic biomarkers and effective treatment options for Kidney Renal Papillary Cell Carcinoma (KIRP) patients, to find potential prognostic biomarkers and new targets was an urgent mission for KIRP therapy. Methods The differentially expressed autophagy-related genes (DEARGs) were screened out according to the RNA sequencing data in The Cancer Genome Atlas database, then identified survival-related DEARGs to establish a prognostic model for survival predicting of KIRP patients. Then we verified the robustness and validity of the prognostic risk model through clinicopathological data. At last, we evaluate the prognostic value of genes that formed the prognostic risk model individually. Results We analyzed the expression of 232 autophagy-related genes (ARGs) in 289 KIRP and 32 non-tumor tissue cases, and 40 mRNAs were screened out as DEARGs. The functional and pathway enrichment analysis was done and protein–protein interaction network was constructed for all DEARGs. To sift candidate DEARGs associated with KIRP patients’ survival and create an autophagy-related risk prognostic model, univariate and multivariate Cox regression analysis were did separately. Eventually 3 desirable independent prognostic DEARGs (P4HB, NRG1, and BIRC5) were picked out and used for construct the autophagy-related risk model. The accuracy of the prognostic risk model for survival prediction was assessed by Kaplan–Meier plotter, receiver-operator characteristic curve, and clinicopathological correlational analyses. The prognostic value of above 3 genes was verified individually by survival analysis and expression analysis on mRNA and protein level. Conclusions The autophagy-related prognostic model is accurate and applicable, it can predict OS independently for KIRP patients. Three independent prognostic DEARGs can benefit for facilitate personalized target treatment too.


2020 ◽  
Author(s):  
Zengyu Feng ◽  
Minmin Shi ◽  
Kexian Li ◽  
Lingxi Jiang ◽  
Hao Chen ◽  
...  

Abstract Background Cancer stem cells (CSCs) are crucial to malignant behavior and poor prognosis of pancreatic ductal adenocarcinoma (PDAC). In recent years, CSC biology has been widely studied, but practical prognostic signatures based on CSC-related genes have not been established and reported in PDAC. Methods This signature was developed and validated in seven independent PDAC datasets. MTAB-6134 cohort was used as training set, while one Chinese local cohort and five other public cohorts were used for external validation. CSC-related genes with credible prognostic roles were selected to form the signature, and its predictive performance was evaluated by Kaplan-Meier survival, receiver operating characteristic (ROC), and calibration curves. Correlation analysis was employed to clarify the potential biological characteristics. Results A robust signature comprising DCBLD2, GSDMD, PMAIP1, and PLOD2 was developed. It could classify patients into high-risk and low-risk groups, and high risk patients had significantly shorter overall survival (OS) and disease-free survival (DFS) compared with low risk patients. Calibration curves and Cox regression analysis demonstrated the powerful predictive performance. ROC curves showed an improved survival prediction provided by this model. Functional analysis revealed positive association between risk score and CSC marker. These results had cross-dataset compatibility. Conclusions We established a novel four-gene signature based on CSC-related genes which could serve as a powerful prognostic tool in PDAC. Impact This signature can offer potential help for further improving current TNM staging system, and providing data for the development of novel personalized therapeutic strategies in the future.


Sign in / Sign up

Export Citation Format

Share Document