scholarly journals Construction and Validation of a Ferroptosis-Related Prognostic Model for Gastric Cancer

2021 ◽  
Vol 2021 ◽  
pp. 1-14
Author(s):  
Xiaotao Jiang ◽  
Qiaofeng Yan ◽  
Linling Xie ◽  
Shijie Xu ◽  
Kailin Jiang ◽  
...  

Background. Gastric cancer (GC), an extremely aggressive tumor with a very different prognosis, is the third leading cause of cancer-related mortality. We aimed to construct a ferroptosis-related prognostic model that can be distinguished prognostically. Methods. The gene expression and the clinical data of GC patients were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus database (GEO). The ferroptosis-related genes were obtained from the FerrDb. Using the “limma” R package and univariate Cox analysis, ferroptosis-related genes with differential expression and prognostic value were identified in the TCGA cohort. Last absolute shrinkage and selection operator (LASSO) Cox regression was applied to shrink ferroptosis-related predictors and construct a prognostic model. Functional enrichment, ESTIMATE algorithm, and single-sample gene set enrichment analysis (ssGSEA) were applied for exploring the potential mechanism. GC patients from the GEO cohort were used for validation. Results. An 8-gene prognostic model was constructed and stratified GC patients from TCGA and meta-GEO cohort into high-risk groups or low-risk groups. GC patients in high-risk groups have significantly poorer OS compared with those in low-risk groups. The risk score was identified as an independent predictor for OS. Functional analysis revealed that the risk score was mainly associated with the biological function of extracellular matrix (ECM) organization and tumor immunity. Conclusion. In conclusion, the ferroptosis-related model can be utilized for the clinical prognostic prediction in GC.

2021 ◽  
Vol 11 ◽  
Author(s):  
Kebing Huang ◽  
Xiaoyu Yue ◽  
Yinfei Zheng ◽  
Zhengwei Zhang ◽  
Meng Cheng ◽  
...  

Glioma is well known as the most aggressive and prevalent primary malignant tumor in the central nervous system. Molecular subtypes and prognosis biomarkers remain a promising research area of gliomas. Notably, the aberrant expression of mesenchymal (MES) subtype related long non-coding RNAs (lncRNAs) is significantly associated with the prognosis of glioma patients. In this study, MES-related genes were obtained from The Cancer Genome Atlas (TCGA) and the Ivy Glioblastoma Atlas Project (Ivy GAP) data sets of glioma, and MES-related lncRNAs were acquired by performing co-expression analysis of these genes. Next, Cox regression analysis was used to establish a prognostic model, that integrated ten MES-related lncRNAs. Glioma patients in TCGA were divided into high-risk and low-risk groups based on the median risk score; compared with the low-risk groups, patients in the high-risk group had shorter survival times. Additionally, we measured the specificity and sensitivity of our model with the ROC curve. Univariate and multivariate Cox analyses showed that the prognostic model was an independent prognostic factor for glioma. To verify the predictive power of these candidate lncRNAs, the corresponding RNA-seq data were downloaded from the Chinese Glioma Genome Atlas (CGGA), and similar results were obtained. Next, we performed the immune cell infiltration profile of patients between two risk groups, and gene set enrichment analysis (GSEA) was performed to detect functional annotation. Finally, the protective factors DGCR10 and HAR1B, and risk factor SNHG18 were selected for functional verification. Knockdown of DGCR10 and HAR1B promoted, whereas knockdown of SNHG18 inhibited the migration and invasion of gliomas. Collectively, we successfully constructed a prognostic model based on a ten MES-related lncRNAs signature, which provides a novel target for predicting the prognosis for glioma patients.


2020 ◽  
Author(s):  
Ming Liu ◽  
Jiayi Xie ◽  
Xiaobei Luo ◽  
Yaxin Luo ◽  
Side Liu ◽  
...  

Abstract Background: Gastric cancer (GC) is one of the most prevalent malignant cancers around the world. Given that abnormal RNA binding proteins (RBPs) are involved in the tumorigenesis, we aimed to explore the potential value of RBPs-associated genes in gastric cancer.Methods: RNA-seq and clinical data were retrieved from The Cancer Genome Atlas (TCGA) database and differentially expressed RBPs genes were screened. GO and KEGG pathway enrichment analyses were implemented to elucidate the roles of RBPs in GC. The protein-protein interaction (PPI) networks of RBPs were carried out, and the hub genes were determined by MCODE built in Cytoscape. The TCGA-STAD dataset was randomly divided into training and testing groups. A prognostic signature including five RBPs was developed within the training cohort after Cox regression and Lasso regression analyses. We used Kaplan–Meier (KM) and receiver operating characteristic (ROC) curves to evaluate the capacity of the model in both groups. Then, a nomogram based on hub RBPs expression was established. Gene Set Enrichment Analysis was performed between the high-risk and low-risk group.Results: A total of 166 up-regulated RBPs and 130 down-regulated RBPs were identified. Via Cox regression and Lasso regression analysis within the training group, five hub RBPs (RNASE1, SETD7, BOLL, PPARGC1B, MSI2) were screened and the prognostic model was constructed. The risk score was calculated and gastric cancer patients were divided into high-risk and low-risk groups. In multivariate analysis, risk score was still an independent prognostic indicator (HR = 1.80, 95% CI = 1.45-2.22, P < 0.01). Patients with low risk had favorable survival rate in both training and testing group compared to those at high risk (P < 0.001). The areas under the ROC curves (AUC) of the prognostic model are 0.718 in the training cohort and 0.651 in the testing cohort. The hub RBPs-based nomogram model exhibited excellent ability to predict the OS of GC. GSEA illustrated that several cancer-related signaling pathways were enriched in patients with a high-risk score.Conclusions: This study discovered a five RBPs signature which might provide a potential prognostic value to GC patients.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1093-1093
Author(s):  
Fong Chun Chan ◽  
Anja Mottok ◽  
Alina S Gerrie ◽  
Maryse M Power ◽  
Kerry J Savage ◽  
...  

Abstract Introduction: Disease progression remains a significant clinical burden in classical Hodgkin lymphoma (CHL) with 25-30% of patients relapsing after first-line treatment. The current standard of care for relapsed CHL is high dose chemotherapy followed by autologous stem cell transplantation (ASCT). This secondary therapy only cures approximately 50% of patients, with virtually no reliable biomarkers to identify the patients in which this salvage treatment regimen fails. The specific aim of this study was to establish the extent of changes in tumor microenvironment (TME) composition between matched primary and relapse samples, and to build and validate a prognostic model for post-ASCT outcomes using relapse samples. Materials & Methods:NanoString digital gene expression profiling was used to ascertain the gene expression of 784 genes of interest from 245 biopsies sampled from 174 CHL patients. This cohort included 90 patients with single biopsies performed at first diagnosis (primary), 13 patients with single biopsies taken at relapse, and 71 patients with paired biopsies taken at first diagnosis and at relapse. All patients received ABVD as first-line treatment, and 151 patients went on to receive ASCT. The 784 genes of interest were selected based on previously reported associations with outcome in CHL and/or components of the TME. Spearman statistics were used for pairwise correlations and log-rank tests were used to assess survival differences. Bootstrap aggregation with concordance statistics (C-stat) was used to calculate the post-ASCT prognostic properties and a two-sample t-test was used to compare primary and relapse samples. Penalized elastic-net multivariate cox regression was used for model construction. An independent, similarly treated, cohort of 31 relapse biopsies was used for model validation. Results: Comparative gene expression analysis revealed that 17 of the 71 patients (24%) exhibited poor correlations between their paired primary and relapse samples (r2 < 0.75) - indicative of significant differences in their TME compositions. Amongst these differences was a striking inverse correlation between macrophage and B-cell gene expression pattern changes (r2 = -0.809). We validated these findings by using CD20 and CD163 immunohistochemistry confirming this inverse correlation of relative changes in macrophage and B cell content in the TME (r2= -0.645). Patients who exhibited poor gene expression correlations had inferior post-ASCT failure-free survival (FFS) (3-year: 38.5%) compared to patients with high gene expression correlations (3-year: 77%; P = 0.005). A comparative C-stat analysis of prognostic properties between primary and relapse samples demonstrated that relapse samples contain superior prognostic features for prediction of post-ASCT outcomes (relapse C-stat 0.785 ± 0.073 vs. primary C-stat 0.594 ± 0.079, P < 0.001). To this end, we developed a gene expression prognostic model using penalized Cox regression that was based on gene expression measurements in relapse samples (RHL30). RHL30 was able to risk stratify patients according to post-ASCT outcomes in the training cohort (5-year post-ASCT FFS: 23.8% in high-risk vs. 77.5% in low-risk; 5-year post-ASCT overall survival [OS]: 30.9% in high-risk vs. 85.4% in low-risk). This was validated in an independent cohort of 31 patients with relapsed CHL with a 5-year post-ASCT FFS of 37.5% in the high-risk vs. 70.1% in the low-risk groups (P = 0.017) and 5-year post-ASCT OS of 37.5% in the high-risk vs. 71.6% in the low-risk groups (P = 0.006). Conclusions: The TME gene expression profile differs significantly between matched primary and relapse CHL samples in a subset of patients with relapsed CHL. Gene expression measurements derived from relapse samples contain superior predictive properties for response to ASCT. To this end, we have developed a novel clinically applicable prognostic model (RHL30), derived from relapse samples, that identifies patients who have a high likelihood to benefit from ASCT (low-risk), and conversely a subgroup of patients who may benefit from additional or alternative therapeutic approaches (high-risk). Disclosures Connors: Seattle Genetics: Research Funding; Bristol Myers Squib: Research Funding; Millennium Takeda: Research Funding; NanoString Technologies: Research Funding; F Hoffmann-La Roche: Research Funding. Scott:NanoString Technologies: Patents & Royalties: named inventor on a patent for molecular subtyping of DLBCL that has been licensed to NanoString Technologies.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Qian Yan ◽  
Wenjiang Zheng ◽  
Boqing Wang ◽  
Baoqian Ye ◽  
Huiyan Luo ◽  
...  

Abstract Background Hepatocellular carcinoma (HCC) is a disease with a high incidence and a poor prognosis. Growing amounts of evidence have shown that the immune system plays a critical role in the biological processes of HCC such as progression, recurrence, and metastasis, and some have discussed using it as a weapon against a variety of cancers. However, the impact of immune-related genes (IRGs) on the prognosis of HCC remains unclear. Methods Based on The Cancer Gene Atlas (TCGA) and Immunology Database and Analysis Portal (ImmPort) datasets, we integrated the ribonucleic acid (RNA) sequencing profiles of 424 HCC patients with IRGs to calculate immune-related differentially expressed genes (DEGs). Survival analysis was used to establish a prognostic model of survival- and immune-related DEGs. Based on genomic and clinicopathological data, we constructed a nomogram to predict the prognosis of HCC patients. Gene set enrichment analysis further clarified the signalling pathways of the high-risk and low-risk groups constructed based on the IRGs in HCC. Next, we evaluated the correlation between the risk score and the infiltration of immune cells, and finally, we validated the prognostic performance of this model in the GSE14520 dataset. Results A total of 100 immune-related DEGs were significantly associated with the clinical outcomes of patients with HCC. We performed univariate and multivariate least absolute shrinkage and selection operator (Lasso) regression analyses on these genes to construct a prognostic model of seven IRGs (Fatty Acid Binding Protein 6 (FABP6), Microtubule-Associated Protein Tau (MAPT), Baculoviral IAP Repeat Containing 5 (BIRC5), Plexin-A1 (PLXNA1), Secreted Phosphoprotein 1 (SPP1), Stanniocalcin 2 (STC2) and Chondroitin Sulfate Proteoglycan 5 (CSPG5)), which showed better prognostic performance than the tumour/node/metastasis (TNM) staging system. Moreover, we constructed a regulatory network related to transcription factors (TFs) that further unravelled the regulatory mechanisms of these genes. According to the median value of the risk score, the entire TCGA cohort was divided into high-risk and low-risk groups, and the low-risk group had a better overall survival (OS) rate. To predict the OS rate of HCC, we established a gene- and clinical factor-related nomogram. The receiver operating characteristic (ROC) curve, concordance index (C-index) and calibration curve showed that this model had moderate accuracy. The correlation analysis between the risk score and the infiltration of six common types of immune cells showed that the model could reflect the state of the immune microenvironment in HCC tumours. Conclusion Our IRG prognostic model was shown to have value in the monitoring, treatment, and prognostic assessment of HCC patients and could be used as a survival prediction tool in the near future.


2021 ◽  
Vol 12 ◽  
Author(s):  
Dongjie Chen ◽  
Hui Huang ◽  
Longjun Zang ◽  
Wenzhe Gao ◽  
Hongwei Zhu ◽  
...  

We aim to construct a hypoxia- and immune-associated risk score model to predict the prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). By unsupervised consensus clustering algorithms, we generate two different hypoxia clusters. Then, we screened out 682 hypoxia-associated and 528 immune-associated PDAC differentially expressed genes (DEGs) of PDAC using Pearson correlation analysis based on the Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression project (GTEx) dataset. Seven hypoxia and immune-associated signature genes (S100A16, PPP3CA, SEMA3C, PLAU, IL18, GDF11, and NR0B1) were identified to construct a risk score model using the Univariate Cox regression and the Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression, which stratified patients into high- and low-risk groups and were further validated in the GEO and ICGC cohort. Patients in the low-risk group showed superior overall survival (OS) to their high-risk counterparts (p &lt; 0.05). Moreover, it was suggested by multivariate Cox regression that our constructed hypoxia-associated and immune-associated prognosis signature might be used as the independent factor for prognosis prediction (p &lt; 0.001). By CIBERSORT and ESTIMATE algorithms, we discovered that patients in high-risk groups had lower immune score, stromal score, and immune checkpoint expression such as PD-L1, and different immunocyte infiltration states compared with those low-risk patients. The mutation spectrum also differs between high- and low-risk groups. To sum up, our hypoxia- and immune-associated prognostic signature can be used as an approach to stratify the risk of PDAC.


2020 ◽  
Author(s):  
Jianfeng Zheng ◽  
Jinyi Tong ◽  
Benben Cao ◽  
Xia Zhang ◽  
Zheng Niu

Abstract Background: Cervical cancer (CC) is a common gynecological malignancy for which prognostic and therapeutic biomarkers are urgently needed. The signature based on immune‐related lncRNAs(IRLs) of CC has never been reported. This study aimed to establish an IRL signature for patients with CC.Methods: The RNA-seq dataset was obtained from the TCGA, GEO, and GTEx database. The immune scores(IS)based on single-sample gene set enrichment analysis (ssGSEA) were calculated to identify the IRLs, which were then analyzed using univariate Cox regression analysis to identify significant prognostic IRLs. A risk score model was established to divide patients into low-risk and high-risk groups based on the median risk score of these IRLs. This was then validated by splitting TCGA dataset(n=304) into a training-set(n=152) and a valid-set(n=152). The fraction of 22 immune cell subpopulations was evaluated in each sample to identify the differences between low-risk and high-risk groups. Additionally, a ceRNA network associated with the IRLs was constructed.Results: A cohort of 326 CC and 21 normal tissue samples with corresponding clinical information was included in this study. Twenty-eight IRLs were collected according to the Pearson’s correlation analysis between immune score and lncRNA expression (P < 0.01). Four IRLs (BZRAP1-AS1, EMX2OS, ZNF667-AS1, and CTC-429P9.1) with the most significant prognostic values (P < 0.05) were identified which demonstrated an ability to stratify patients into low-risk and high-risk groups by developing a risk score model. It was observed that patients in the low‐risk group showed longer overall survival (OS) than those in the high‐risk group in the training-set, valid-set, and total-set. The area under the curve (AUC) of the receiver operating characteristic curve (ROC curve) for the four IRLs signature in predicting the one-, two-, and three-year survival rates were larger than 0.65. In addition, the low-risk and high-risk groups displayed different immune statuses in GSEA. These IRLs were also significantly correlated with immune cell infiltration. Conclusions: Our results showed that the IRL signature had a prognostic value for CC. Meanwhile, the specific mechanisms of the four-IRLs in the development of CC were ascertained preliminarily.


2020 ◽  
Author(s):  
Bin Wu ◽  
Yi Yao ◽  
Yi Dong ◽  
Si Qi Yang ◽  
Deng Jing Zhou ◽  
...  

Abstract Background:We aimed to investigate an immune-related long non-coding RNA (lncRNA) signature that may be exploited as a potential immunotherapy target in colon cancer. Materials and methods: Colon cancer samples from The Cancer Genome Atlas (TCGA) containing available clinical information and complete genomic mRNA expression data were used in our study. We then constructed immune-related lncRNA co-expression networks to identify the most promising immune-related lncRNAs. According to the risk score developed from screened immune-related lncRNAs, the high-risk and low-risk groups were separated on the basis of the median risk score, which served as the cutoff value. An overall survival analysis was then performed to confirm that the risk score developed from screened immune-related lncRNAs could predict colon cancer prognosis. The prediction reliability was further evaluated in the independent prognostic analysis and receiver operating characteristic curve (ROC). A principal component analysis (PCA) and gene set enrichment analysis (GSEA) were performed for functional annotation. Results: Information for a total of 514 patients was included in our study. After multiplex analysis, 12 immune-related lncRNAs were confirmed as a signature to evaluate the risk scores for each patient with cancer. Patients in the low-risk group exhibited a longer overall survival (OS) than those in the high-risk group. Additionally, the risk scores were an independent factor, and the Area Under Curve (AUC) of ROC for accuracy prediction was 0.726. Moreover, the low-risk and high-risk groups displayed different immune statuses based on principal components and gene set enrichment analysis.Conclusions: Our study suggested that the signature consisting of 12 immune-related lncRNAs can provide an accessible approach to measuring the prognosis of colon cancer and may serve as a valuable antitumor immunotherapy.


2021 ◽  
Author(s):  
Junliang Li ◽  
Lingfang Zhang ◽  
Tiankang Guo

Abstract Background. Peritoneal metastatic gastric cancer (PMGC) is very common, and usually, the prognosis is poor. There is currently an absence of accurate methods for the early diagnosis and prediction of peritoneal metastasis (PM). This highlights the need to develop strategies to identify the risk of PMGC. Methods. We performed a comprehensive discovery of biomarkers to predict PM by analyzing profiling datasets from GSE62254. The prognostic PM-related genes were obtained using the univariate Cox regression analysis, followed by a least absolute shrinkage and selection operator regression (LASSO) to establish a risk score model. The gene set enrichment analysis (GSEA) was used to determine the pathway enrichment in both the high- and low-risk groups. The 1-, 3-, and 5-year overall survival (OS) rates and area under the receiver operating characteristic curve (ROC) were used to compare the predictive accuracy-based risk stratification. In addition, an unsupervised clustering algorithm was applied to divide patients into subgroups according to the PM-related genes. Results. We identified 10 genes (MMP12, TAC1, TSPYL5, PPP1R14A, TMSB15B, NPY1R, PCDH9, EPM2AIP1, TIG7, and DYNC1I1) for PMGC diagnosis. The OS rates between the high- and low-risk groups at 1-, 3-, and 5-years were significantly different in the training and validation sets. The AUCs at 1-, 3-, and 5-years in the training set were 0.71, 0.74, and 0.73, respectively. In the validation set, the AUCs at 1-, 3-, and 5-years were 0.68, 0.66, and 0.69, respectively. The 10 gene signatures were correlated with immune cell infiltration in both the high- and low-risk groups. In addition, based on the GSEA, several significant pathways were enriched in the high-risk PMGC group, such as the Wnt and transforming growth factor beta (TGF-β) signaling pathway and leukocyte transendothelial migration pathway. Furthermore, unsupervised cluster analysis showed that the model could distinguish the level of risk among patients with PMGC. Conclusions. Overall, 10 gene signatures were identified for PMGC risk prediction. These may be valuable in making clinical decisions to improve treatment outcomes in patients with PMGC.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Pu Wu ◽  
Jinyuan Shi ◽  
Wei Sun ◽  
Hao Zhang

Abstract Background Pyroptosis is a form of programmed cell death triggered by inflammasomes. However, the roles of pyroptosis-related genes in thyroid cancer (THCA) remain still unclear. Objective This study aimed to construct a pyroptosis-related signature that could effectively predict THCA prognosis and survival. Methods A LASSO Cox regression analysis was performed to build a prognostic model based on the expression profile of each pyroptosis-related gene. The predictive value of the prognostic model was validated in the internal cohort. Results A pyroptosis-related signature consisting of four genes was constructed to predict THCA prognosis and all patients were classified into high- and low-risk groups. Patients with a high-risk score had a poorer overall survival (OS) than those in the low-risk group. The area under the curve (AUC) of the receiver operator characteristic (ROC) curves assessed and verified the predictive performance of this signature. Multivariate analysis showed the risk score was an independent prognostic factor. Tumor immune cell infiltration and immune status were significantly higher in low-risk groups, which indicated a better response to immune checkpoint inhibitors (ICIs). Of the four pyroptosis-related genes in the prognostic signature, qRT-PCR detected three of them with significantly differential expression in THCA tissues. Conclusion In summary, our pyroptosis-related risk signature may have an effective predictive and prognostic capability in THCA. Our results provide a potential foundation for future studies of the relationship between pyroptosis and the immunotherapy response.


2021 ◽  
Vol 11 ◽  
Author(s):  
Xiaoxia Tong ◽  
Xiaofei Qu ◽  
Mengyun Wang

BackgroundCutaneous melanoma (CM) is one of the most aggressive cancers with highly metastatic ability. To make things worse, there are limited effective therapies to treat advanced CM. Our study aimed to investigate new biomarkers for CM prognosis and establish a novel risk score system in CM.MethodsGene expression data of CM from Gene Expression Omnibus (GEO) datasets were downloaded and analyzed to identify differentially expressed genes (DEGs). The overlapped DEGs were then verified for prognosis analysis by univariate and multivariate COX regression in The Cancer Genome Atlas (TCGA) datasets. Based on the gene signature of multiple survival associated DEGs, a risk score model was established, and its prognostic and predictive role was estimated through Kaplan-Meier (K-M) analysis and log-rank test. Furthermore, the correlations between prognosis related genes expression and immune infiltrates were analyzed via Tumor Immune Estimation Resource (TIMER) site.ResultsA total of 103 DEGs were obtained based on GEO cohorts, and four genes were verified in TCGA datasets. Subsequently, four genes (ADAMDEC1, GNLY, HSPA13, and TRIM29) model was developed by univariate and multivariate Cox regression analyses. The K-M plots showed that the high-risk group was associated with shortened survival than that in the low-risk group (P &lt; 0.0001). Multivariate analysis suggested that the model was an independent prognostic factor (high-risk vs. low-risk, HR= 2.06, P &lt; 0.001). Meanwhile, the high-risk group was prone to have larger breslow depth (P&lt; 0.001) and ulceration (P&lt; 0.001).ConclusionsThe four-gene risk score model functions well in predicting the prognosis and treatment response in CM and will be useful for guiding therapeutic strategies for CM patients. Additional clinical trials are needed to verify our findings.


Sign in / Sign up

Export Citation Format

Share Document