Novel insight on predicting prognosis of gastric cancer based on inflammation

  • Abstract
  • References
  • Citations
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

BackgroundThe tumor microenvironment (TME) and inflammation play vital roles in the development and progression of gastric cancer (GC). However, there are no inflammation-related models that can predict the prognosis and immunotherapy response of GC patients. We aimed to establish a prognostic model based on an inflammation-related gene (IRG) signature that can predict poor clinical outcomes in GC.MethodsWe searched IRGs in The Cancer Genome Atlas (TCGA) database and identified genes differentially expressed in GC. The model was constructed using univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analysis and validated using Gene Expression Omnibus (GEO) database. Receiver operating characteristic (ROC) curve, principal component analysis (PCA), and t-distribution stochastic neighbor embedding (t-SNE) analysis were performed to evaluate model performance. Independent prognostic factor, immune infiltration, cancer stemness, immunotherapy response analysis and gene set enrichment analysis (GSEA) were performed for functional evaluation.ResultsAn inflammation-related risk model was established based on 8 genes (F2, LBP, SERPINE1, ADAMTS12, FABP4, PROC, TNFSF18, and CYSLTR1). Risk score significantly correlated with poor outcomes and independently predicted prognosis. It was also associated with immune infiltration and reflected immunotherapy response.ConclusionsWe established and validated an inflammation-related prognostic model that predicts immune escape and patient prognosis in GC. Our model is expected to improve clinical outcomes by facilitating clinical decision making and the development of individualized treatments.

ReferencesShowing 10 of 28 papers
  • Open Access Icon
  • PDF Download Icon
  • Cite Count Icon 76
  • 10.3389/fimmu.2020.01295
Targeting NK Cell Checkpoint Receptors or Molecules for Cancer Immunotherapy.
  • Jun 23, 2020
  • Frontiers in Immunology
  • Cai Zhang + 1 more

  • Open Access Icon
  • PDF Download Icon
  • Cite Count Icon 122
  • 10.1038/s41598-019-43924-x
FN1, SPARC, and SERPINE1 are highly expressed and significantly related to a poor prognosis of gastric adenocarcinoma revealed by microarray and bioinformatics
  • May 24, 2019
  • Scientific reports
  • Li Li + 7 more

  • Open Access Icon
  • Cite Count Icon 45973
  • 10.1073/pnas.0506580102
Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles
  • Sep 30, 2005
  • Proceedings of the National Academy of Sciences
  • Aravind Subramanian + 10 more

  • Open Access Icon
  • Cite Count Icon 41
  • 10.7717/peerj.7091
Global transcriptomic analysis identifies SERPINE1 as a prognostic biomarker associated with epithelial-to-mesenchymal transition in gastric cancer.
  • Jun 10, 2019
  • PeerJ
  • Bodong Xu + 3 more

  • Open Access Icon
  • Cite Count Icon 637
  • 10.1038/457036b
Inflaming metastasis
  • Dec 31, 2008
  • Nature
  • Alberto Mantovani

  • Open Access Icon
  • PDF Download Icon
  • Cite Count Icon 43
  • 10.1016/j.ctrv.2019.101931
Primary and acquired resistance mechanisms to immune checkpoint inhibition in Hodgkin lymphoma
  • Nov 11, 2019
  • Cancer Treatment Reviews
  • Johanna Veldman + 3 more

  • Cite Count Icon 11
  • 10.1016/j.lfs.2020.118402
Establishment of an immune microenvironment-based prognostic predictive model for gastric cancer
  • Sep 11, 2020
  • Life Sciences
  • Luying Wan + 3 more

  • Open Access Icon
  • Cite Count Icon 75
  • 10.1172/jci123106
Endothelial miR-30c suppresses tumor growth via inhibition of TGF-β-induced Serpine1.
  • Mar 11, 2019
  • Journal of Clinical Investigation
  • James V Mccann + 14 more

  • Open Access Icon
  • Cite Count Icon 2942
  • 10.1016/1074-7613(95)90125-6
Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4
  • Nov 1, 1995
  • Immunity
  • Elizabeth A Tivol + 5 more

  • Open Access Icon
  • Cite Count Icon 437
  • 10.1111/cei.13407
TIGIT as an emerging immune checkpoint.
  • Dec 25, 2019
  • Clinical and Experimental Immunology
  • H Harjunpää + 1 more

CitationsShowing 1 of 1 papers
  • Open Access Icon
  • Research Article
  • Cite Count Icon 23
  • 10.1186/s13046-023-02833-8
Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
  • Oct 3, 2023
  • Journal of Experimental & Clinical Cancer Research : CR
  • Li Xie + 13 more

BackgroundLiver metastasis (LM) is one of the most common distant metastases of gastric cancer (GC). However, the mechanisms underlying the LM of GC (GC-LM) remain poorly understood. This study aimed to identify the tumour-secreted protein associated with GC-LM and to investigate the mechanisms by which this secreted protein remodels the liver microenvironment to promote GC-LM.MethodsData-independent acquisition mass spectrometry (DIA-MS), mRNA expression microarray, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) were performed to identify and validate the GC-secreted proteins associated with GC-LM. A modified intrasplenic injection mouse model of LM was used to evaluate the progression and tumour burden of LM in vivo. Flow cytometry, immunofluorescence (IF), western blots (WB) and IHC were performed to validate the pre-metastatic niche (PMN) formation in the pre-modelling mouse models. mRNA sequencing of PMA-treated THP-1 cells with or without lipopolysaccharide binding protein (LBP) treatment was used to identify the functional target genes of LBP in macrophages. Co-immunoprecipitation (Co-IP), WB, ELISA, IF and Transwell assays were performed to explore the underlying mechanism of LBP in inducing intrahepatic PMN formation.ResultsLBP was identified as a critical secreted protein associated with GC-LM and correlated with a worse prognosis in patients with GC. LBP activated the TLR4/NF-κB pathway to promote TGF-β1 secretion in intrahepatic macrophages, which, in turn, activated hepatic satellite cells (HSCs) to direct intrahepatic fibrotic PMN formation. Additionally, TGF-β1 enhanced the migration and invasion of incoming metastatic GC cells in the liver. Consequently, selective targeting of the TGF-β/Smad signaling pathway with galunisertib demonstrated its efficacy in effectively preventing GC-LM in vivo.ConclusionsThe results of this study provide compelling evidence that serological LBP can serve as a valuable diagnostic biomarker for the early detection of GC-LM. Mechanistically, GC-derived LBP mediates the crosstalk between primary GC cells and the intrahepatic microenvironment by promoting TGF-β1 secretion in intrahepatic macrophages, which induces intrahepatic fibrotic PMN formation to promote GC-LM. Importantly, selectively targeting the TGF-β/Smad signaling pathway with galunisertib represents a promising preventive and therapeutic strategy for GC-LM.

Similar Papers
  • Research Article
  • Cite Count Icon 30
  • 10.3389/fgene.2021.661306
Identification of an EMT-Related Gene Signature for Predicting Overall Survival in Gastric Cancer.
  • Jun 24, 2021
  • Frontiers in genetics
  • Weiyu Dai + 11 more

BackgroundIt has been widely reported that epithelial-mesenchymal transition (EMT) is associated with malignant progression in gastric cancer (GC). Integration of the molecules related to EMT for predicting overall survival (OS) is meaningful for understanding the role of EMT in GC. Here, we aimed to establish an EMT-related gene signature in GC.MethodsTranscriptional profiles and clinical data of GC were downloaded from The Cancer Genome Atlas (TCGA). We constructed EMT-related gene signature for predicting OS by using univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression analyses. Time-dependent receiver operating characteristic (ROC), Kaplan-Meier analysis were performed to assess its predictive value. A nomogram combining the prognostic signature with clinical characteristics for OS prediction was established. And its predictive power was estimated by concordance index (C-index), time-dependent ROC curve, calibration curve and decision curve analysis (DCA). GSE62254 dataset from Gene Expression Omnibus (GEO) was used for external validation. Quantitative real-time PCR (qRT-PCR) was used to detected the mRNA expression of the five EMT-related genes in human normal gastric mucosal and GC cell lines. To further understand the potential mechanisms of the signature, Gene Set Enrichment Analysis (GSEA), pathway enrichment analysis, predictions of transcription factors (TFs)/miRNAs were performed.ResultsA novel EMT-related gene signature (including ITGAV, DAB2, SERPINE1, MATN3, PLOD2) was constructed for OS prediction of GC. With external validation, ROC curves indicated the signature’s good performance. Patients stratified into high- and low-risk groups based on the signature yielded significantly different prognosis. Univariate and multivariate Cox regression suggested that the signature was an independent prognostic variable. Nomogram for prognostication including the signature presented better predictive accuracy and clinical usefulness than the similar model without risk score to some extent with external validation. The qRT-PCR assays suggested that high expression of the five EMT-related genes could be found in human GC cell lines compared with normal gastric mucosal cell line. GSEA and pathway enrichment analysis revealed that focal adhesion and ECM-receptor interaction might be the two important pathways to the signature.ConclusionOur EMT-related gene signature may have practical application as an independent prognostic factor in GC.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 8
  • 10.3389/fonc.2020.522015
Tumor-Infiltrating Lymphocyte-Based Risk Score for Predicting Prognosis in Gastric Cancer.
  • Sep 30, 2020
  • Frontiers in oncology
  • Siyuan Xie + 6 more

Tumor-infiltrating lymphocytes (TILs) in gastric cancer are closely related to clinical prognosis; however, little is known regarding the immune microenvironment in this disease. Thus, RNA-sequencing data from gastric cancer patients were downloaded from the Gene Expression Omnibus (GEO). The proportion of immune cells was determined based on a deconvolution algorithm (CIBERSORT), and gene expression profiles were analyzed in the context of clinical outcomes to construct an immune risk score. Data were analyzed using least absolute shrinkage and selection operator (LASSO) and multivariable Cox regression, to identify prognostic markers of gastric cancer survival. The model included four immune cell types: neutrophils, plasma cells, activated CD4+ memory T cells, and T follicular helper cells. Patients were classified into two subgroups based on risk score, and a significant difference in overall survival (OS) was seen between the subgroups in both the training and testing cohorts, particularly in patients with tumor stages ≥T3. Multivariable analysis revealed that both T-stage and risk score were independent prognostic factors for gastric cancer survival [hazard ratio (HR) 1.505; 95% confidence interval (CI) 1.043–2.173, HR 1.686; 95% CI 1.367–2.080]. Risk scores and clinical factors were then integrated into a nomogram to build a model with both good discriminatory power and accuracy in predicting clinical outcomes. Further analysis using gene set enrichment analysis (GSEA) identified strong associations of immune risk with TGF-β and tumor metastasis-related pathways, which could inform research on the molecular mechanisms of gastric cancer. Collectively, the data presented here suggest that an immune risk model can make an important contribution to predictions prognosis in gastric cancer patients.

  • Research Article
  • Cite Count Icon 3
  • 10.3389/fonc.2024.1485580
Lactylation-related gene signature accurately predicts prognosis and immunotherapy response in gastric cancer.
  • Nov 28, 2024
  • Frontiers in oncology
  • Xuezeng Sun + 6 more

Gastric cancer (GC) is a malignant tumor associated with significant rates of morbidity and mortality. Hence, developing efficient predictive models and directing clinical interventions in GC is crucial. Lactylation of proteins is detected in gastric cancer tumors and is linked to the advancement of gastric cancer. The The Cancer Genome Atlas (TCGA) was utilized to analyze the gene expression levels associated with lactylation. A genetic pattern linked to lactylation was created using Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) regression. The predictive ability of the model was evaluated and confirmed in the Gene Expression Omnibus (GEO) cohort, where patients were divided into two risk groups based on their scores. The study examined the relationship between gene expression and the presence of immune cells in the context of immunotherapy treatment. In vitro cytotoxicity assays, ELISA and PD-1 and PD-L1interaction assays were used to assess the expression of PD-L1 while knocking down SLC16A7. 29 predictive lactylation-related genes with differential expression were discovered. A signature consisting of three genes was developed and confirmed. Patients who had higher risk scores experienced worse clinical results. The group with lower risk showed increased Tumor Immune Dysfunction and Exclusion (TIDE) score and greater responsiveness to immunotherapy. The tumor tissues secrete more lactate acid than normal tissues and express more PD-L1 than normal tissues, that is, lactate acid promotes the immune evasion of tumor cells. In GC, the lactylation-related signature showed strong predictive accuracy. Utilizing both anti-lactylation and anti-PD-L1 may prove to be an effective approach for treating GC in clinical settings. We further proved that one of the lactate metabolism related genes, SCL16A7 could promote the expression of PD-L1 in GC cells. The risk model not only provides a basis for better prognosis in GC patients, but also is a potential prognostic indicator to distinguish the molecular and immune characteristics, and the response from Immune checkpoint inhibitors (ICI) therapy and chemotherapy in GC.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 8
  • 10.3389/pore.2023.1610893
High expression of TMEM200A is associated with a poor prognosis and immune infiltration in gastric cancer
  • Jan 19, 2023
  • Pathology and Oncology Research
  • Hongyang Deng + 5 more

Background: Gastric cancer (GC) is one of the global malignant tumors with high incidence and poor prognosis. Exploring new GC molecular markers is important to improve GC prognosis. Transmembrane protein 200A (TMEM200A) is a member of the family of transmembrane proteins (TMEM). This study is the first to investigate the potential function of TMEM200A and its relationship with immune infiltration in GC.Methods: The differential expression of TMEM200A was determined through the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The receiver operating characteristic (ROC) curve was drawn to assess the diagnostic value of TMEM200A for GC. The relationship between TMEM200A and the clinical characteristics of patients with GC was investigated using the Wilcoxon test and the Kruskal-Wallis test. The effect of TMEM200A on overall survival (OS) was identified using the Kaplan-Meier method, the Log-rank test, the univariate/multivariate Cox regression analysis, and the nomogram prediction model. The co-expressed genes and gene set enrichment analysis (GSEA) were used to explore the potential biological functions of TMEM200A. We used the Tumor Immune Estimation Resource (TIMER) database and the ssGSEA algorithm to estimate the relationship between TMEM200A and immune cell infiltration. Furthermore, we investigated the correlation of TMEM200A with immune checkpoint/immune cell surface markers using the TCGA-STAD data set. Finally, we identified prognosis-related methylation sites in TMEM200A using MethSurv.Results: TMEM200A was highly expressed in GC tissues. TMEM200A had a good diagnostic value for GC. High expression of TMEM200A may shorten the OS of GC patients and may be an independent risk factor for OS in GC patients. TMEM200A participates in the construction of a predictive model with a good predictive effect on the survival rate of GC patients at 1, 3, and 5 years. Co-expressed genes and GSEA indicated that TMEM200A may be an adhesion molecule closely associated with tumor invasion and metastasis. In addition, TMEM200A may be significantly associated with immune cell infiltration and immune checkpoint expression. We also found that TMEM200A contains three methylation sites associated with a poor prognosis.Conclusion: Upregulated TMEM200A may be a promising prognostic marker for GC and is closely associated with the tumor microenvironment (TME).

  • Research Article
  • Cite Count Icon 4
  • 10.21037/atm-22-3980
Identification and verification of a glycolysis-related gene signature for gastric cancer
  • Sep 1, 2022
  • Annals of Translational Medicine
  • Yi Liu + 7 more

BackgroundGlycolysis is a central metabolic pathway for tumor cells. However, the relationship between glycolysis and the prognosis of gastric cancer (GC) patients is not well established. In this study, we sought to construct a glycolysis-related gene signature for GC.MethodsThe messenger ribonucleic acid (mRNA) expression profiles were analyzed using data from The Cancer Genome Atlas (TCGA) database. Glycolysis-related gene sets and pathways were obtained from the Molecular Signatures Database (MSigDB). Subsequently, a prognosis prediction model of the glycolysis-related genes was constructed using Cox and least absolute shrinkage and selection operator (LASSO) regression analyses. An external validation was conducted using data from the Gene Expression Omnibus (GEO) database. Risk scores were also calculated based on the signature. Finally, the correlations between the risk score and overall survival (OS), mutation, immune cell infiltration, immune score, and stromal score were examined in 22 types of infiltrating immune cells.ResultsFifty-five glycolysis-related genes were identified from TCGA database and MSigDB. Using the LASSO and Cox models, 4 novel genes (i.e., VCAN, EFNA3, ADH4, and CLDN9) were identified to construct a gene signature for GC prognosis prediction. The GC patients with low-risk scores had significantly better OS than those with high-risk scores in the training set. Similar results were also found in the independent GEO GSE84437 testing set. Additionally, the degree of cell infiltration in the low-risk group was significantly higher than that in the high-risk group in terms of naive B cells, plasma cells, and T follicular helper cells. In monocytes, M2 macrophages, resting dendritic cells, and resting Mast cells, the degree of infiltration in the high-risk group was significantly higher than that in the low-risk group. The immune score and stromal score of the high-risk group were also significantly higher than those of the low-risk group. Finally, the univariate and multivariate Cox regression analyses showed that 4 glycolysis-related genes were independent prognostic factors for GC.ConclusionsThe established 4 glycolysis-related gene signature may serve as a reliable tool for the prognosis of GC patients and provide a potential glycolysis therapeutic target for GC.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 33
  • 10.3389/fimmu.2022.783495
Clinical Significance and Immunometabolism Landscapes of a Novel Recurrence-Associated Lipid Metabolism Signature In Early-Stage Lung Adenocarcinoma: A Comprehensive Analysis.
  • Feb 10, 2022
  • Frontiers in Immunology
  • Mingchuang Zhu + 11 more

BackgroundThe early-stage lung adenocarcinoma (LUAD) rate has increased with heightened public awareness and lung cancer screening implementation. Lipid metabolism abnormalities are associated with lung cancer initiation and progression. However, the comprehensive features and clinical significance of the immunometabolism landscape and lipid metabolism-related genes (LMRGs) in cancer recurrence for early-stage LUAD remain obscure.MethodsLMRGs were extracted from Gene Set Enrichment Analysis (GSEA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. Samples from The Cancer Genome Atlas (TCGA) were used as training cohort, and samples from four Gene Expression Omnibus (GEO) datasets were used as validation cohorts. The LUAD recurrence-associated LMRG molecular pattern and signature was constructed through unsupervised consensus clustering, time-dependent receiver operating characteristic (ROC), and least absolute shrinkage and selection operator (LASSO) analyses. Kaplan-Meier, ROC, and multivariate Cox regression analyses and prognostic meta-analysis were used to test the suitability and stability of the signature. We used Gene Ontology (GO), KEGG pathway, immune cell infiltration, chemotherapy response analyses, gene set variation analysis (GSVA), and GSEA to explore molecular mechanisms and immune landscapes related to the signature and the potential of the signature to predict immunotherapy or chemotherapy response.ResultsFirst, two LMRG molecular patterns were established, which showed diverse prognoses and immune infiltration statuses. Then, a 12-gene signature was identified, and a risk model was built. The signature remained an independent prognostic parameter in multivariate Cox regression and prognostic meta-analysis. In addition, this signature stratified patients into high- and low-risk groups with significantly different recurrence rates and was well validated in different clinical subgroups and several independent validation cohorts. The results of GO and KEGG analyses and GSEA showed that there were differences in multiple lipid metabolism, immune response, and drug metabolism pathways between the high- and low-risk groups. Further analyses revealed that the signature-based risk model was related to distinct immune cell proportions, immune checkpoint parameters, and immunotherapy and chemotherapy response, consistent with the GO, KEGG, and GSEA results.ConclusionsThis is the first lipid metabolism-based signature for predicting recurrence, and it could provide vital guidance to achieve optimized antitumor for immunotherapy or chemotherapy for early-stage LUAD.

  • Research Article
  • Cite Count Icon 5
  • 10.21037/tcr-23-1755
Gastric cancer prognosis: unveiling autophagy-related signatures and immune infiltrates.
  • Mar 1, 2024
  • Translational Cancer Research
  • Yichen Yin + 4 more

Autophagy played a crucial regulatory role in tumor initiation and progression. Therefore, we aimed to comprehensively analyze autophagy-related genes (ARGs) in gastric cancer, focusing on their expression, prognostic value, and potential functions. The gastric cancer gene chip datasets (GSE79973 and GSE54129) were collected from the Gene Expression Omnibus (GEO) database. Subsequently, the Limma package was employed to identify differentially expressed genes (DEGs) between the normal and disease groups. The selected ARGs were further authenticated using the Human Protein Atlas (HPA) database, The Cancer Genome Atlas (TCGA) database, and GSE19826 database. A total of 15 autophagy-related DEGs, eight of which were upregulated [FKBP1A, IL24, PEA15, HSP90AB1, cathepsin B (CTSB), ITGB1, SPHK1, HIF1A], while seven were downregulated (DAPK2, EIF2AK3, FKBP1B, PTK6, NKX2-3, NFE2L2, PRKCD). Analysis revealed that CTSB was specifically associated with the prognosis of gastric cancer patients. Gene set enrichment analysis (GSEA) showcased a significant enrichment of CTSB-related genes within immune-related pathways. Moreover, correlation analysis demonstrated a clear association between the expression of CTSB and immune infiltration. The upregulation of CTSB in gastric cancer was linked to poor survival and increased immune infiltration. We conjectured that CTSB likely played a critical role in regulating immunity and autophagy in gastric cancer.

  • Research Article
  • Cite Count Icon 11
  • 10.1186/s12876-022-02159-3
Identification of a novel m6A-related lncRNA pair signature for predicting the prognosis of gastric cancer patients
  • Feb 21, 2022
  • BMC Gastroenterology
  • Jun-Mei Wang + 4 more

BackgroundAccumulating studies have demonstrated that lncRNAs play vital roles in the prognosis of gastric cancer (GC); however, the prognostic value of N6-methyladenosine-related lncRNAs has not been fully reported in GC. This study aimed to construct and validate an m6A-related lncRNA pair signature (m6A-LPS) for predicting the prognosis of GC patients.MethodsGC cohort primary data were downloaded from The Cancer Genome Atlas. We analysed the coexpression of m6A regulators and lncRNAs to identify m6A-related lncRNAs. Based on cyclical single pairing along with a 0-or-1 matrix and least absolute shrinkage and selection operator-penalized regression analyses, we constructed a novel prognostic signature of m6A-related lncRNA pairs with no dependence upon specific lncRNA expression levels. All patients were divided into high-risk and low-risk group based on the median risk score. The predictive reliability was evaluated in the testing dataset and whole dataset with receiver operating characteristic (ROC) curve analysis. Gene set enrichment analysis was used to identify potential pathways.ResultsFourteen m6A-related lncRNA pairs consisting of 25 unique lncRNAs were used to construct the m6A-LPS. Kaplan–Meier analysis showed that the high-risk group had poor prognosis. The area under the curve for 5-year overall survival was 0.906, 0.827, and 0.882 in the training dataset, testing dataset, and whole dataset, respectively, meaning that the m6A-LPS was highly accurate in predicting GC patient prognosis. The m6A-LPS served as an independent prognostic factor for GC patients after adjusting for other clinical factors (p < 0.05). The m6A-LPS had more accuracy and a higher ROC value than other prognostic models for GC. Functional analysis revealed that high-risk group samples mainly showed enrichment of extracellular matrix receptor interactions and focal adhesion. Moreover, N-cadherin and vimentin, known biomarkers of epithelial–mesenchymal transition, were highly expressed in high-risk group samples. The immune infiltration analysis showed that resting dendritic cells, monocytes, and resting memory CD4 T cells were significantly positively related to the risk score. Thus, m6A-LPS reflected the infiltration of several types of immune cells.ConclusionsThe signature established by pairing m6A-related lncRNAs regardless of expression levels showed high and independent clinical prediction value in GC patients.

  • Research Article
  • Cite Count Icon 5
  • 10.1016/j.heliyon.2024.e33277
Comprehensive Analysis of angiogenesis associated genes and tumor microenvironment infiltration characterization in cervical cancer
  • Jun 1, 2024
  • Heliyon
  • Shuzhen Li + 2 more

Comprehensive Analysis of angiogenesis associated genes and tumor microenvironment infiltration characterization in cervical cancer

  • Research Article
  • 10.1128/spectrum.02830-24
Prognostic implications of the interaction between intratumoral microbiome and immune response in gastric cancer.
  • Apr 9, 2025
  • Microbiology spectrum
  • Sifan Liu + 6 more

Gastric cancer (GC) prognosis is significantly influenced by intratumoral microbiomes, which modulate host-immune interactions. This study analyzed data from the The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases to identify immune genes associated with GC prognosis and conducted prognostic immune subtypes. GC patients were classified into two distinct prognostic immune phenotypes C1 and C2 based on the non-negative matrix factorization consensus clusters. Phenotype C2 exhibited a better prognosis and distinct immune characteristics, including enhanced presence of Th2 and Th17 cells and improved response to chemotherapy. In contrast, phenotype C1 showed higher expression levels of PDCD1LG2 and TLR9, which were critical immune factors involved in immune regulation. Both phenotypes were linked to immune genes influencing intratumoral microbiomes and GC immunotherapy responses. A prediction risk model was constructed using the LASSO regression analysis and showed great prognostic value for GC patients. The key genes were correlated with immune cells and suppressed the function of the host immune system. The intratumoral microbiomes were strongly associated with the hosts' immune infiltration and significantly interacted with host immune genes to influence GC outcomes. Candidatus Nitrosotenuis plays a significant role in predicting the prognosis of GC patients. This research underscores the pivotal role of intratumoral microbiomes in GC prognosis and supports the development of future personalized therapeutic approaches.IMPORTANCEIncreasing evidence confirms the presence of intratumoral microbiomes. However, the role of the intratumoral microbiomes in the progression of gastric cancer and their relationship with the immune microenvironment remain unclear. Our study classified gastric cancer patients into two immune prognostic subtypes, C1 and C2, using non-negative matrix factorization consensus clusters. The C2 subtype exhibited a better prognosis and more pronounced immune characteristics. Microbiome analyses revealed associations between both subtypes and immune genes that affect intratumoral microbiomes and their responses to immunotherapy. The intratumoral microbiomes were closely linked with host immune infiltration and significantly interacted with immune genes, which influence the prognosis of gastric cancer. Notably, Candidatus Nitrosotenuis showed a significant prognostic value in gastric cancer patients. Our findings highlight the critical role of the intratumoral microbiomes in affecting gastric cancer prognosis and its interaction with the immune microenvironment, supporting future personalized therapeutic approaches.

  • Research Article
  • Cite Count Icon 1
  • 10.4251/wjgo.v16.i3.945
Identification of a novel inflammatory-related gene signature to evaluate the prognosis of gastric cancer patients.
  • Mar 15, 2024
  • World journal of gastrointestinal oncology
  • Jia-Li Hu + 7 more

Gastric cancer (GC) is a highly aggressive malignancy with a heterogeneous nature, which makes prognosis prediction and treatment determination difficult. Inflammation is now recognized as one of the hallmarks of cancer and plays an important role in the aetiology and continued growth of tumours. Inflammation also affects the prognosis of GC patients. Recent reports suggest that a number of inflammatory-related biomarkers are useful for predicting tumour prognosis. However, the importance of inflammatory-related biomarkers in predicting the prognosis of GC patients is still unclear. To investigate inflammatory-related biomarkers in predicting the prognosis of GC patients. In this study, the mRNA expression profiles and corresponding clinical information of GC patients were obtained from the Gene Expression Omnibus (GEO) database (GSE66229). An inflammatory-related gene prognostic signature model was constructed using the least absolute shrinkage and selection operator Cox regression model based on the GEO database. GC patients from the GSE26253 cohort were used for validation. Univariate and multivariate Cox analyses were used to determine the independent prognostic factors, and a prognostic nomogram was established. The calibration curve and the area under the curve based on receiver operating characteristic analysis were utilized to evaluate the predictive value of the nomogram. The decision curve analysis results were plotted to quantify and assess the clinical value of the nomogram. Gene set enrichment analysis was performed to explore the potential regulatory pathways involved. The relationship between tumour immune infiltration status and risk score was analysed via Tumour Immune Estimation Resource and CIBERSORT. Finally, we analysed the association between risk score and patient sensitivity to commonly used chemotherapy and targeted therapy agents. A prognostic model consisting of three inflammatory-related genes (MRPS17, GUF1, and PDK4) was constructed. Independent prognostic analysis revealed that the risk score was a separate prognostic factor in GC patients. According to the risk score, GC patients were stratified into high- and low-risk groups, and patients in the high-risk group had significantly worse prognoses according to age, sex, TNM stage and Lauren type. Consensus clustering identified three subtypes of inflammation that could predict GC prognosis more accurately than traditional grading and staging. Finally, the study revealed that patients in the low-risk group were more sensitive to certain drugs than were those in the high-risk group, indicating a link between inflammation-related genes and drug sensitivity. In conclusion, we established a novel three-gene prognostic signature that may be useful for predicting the prognosis and personalizing treatment decisions of GC patients.

  • Research Article
  • Cite Count Icon 3
  • 10.2174/0109298673278775231101064235
Systematic Analysis of Tumor Stem Cell-related Gene Characteristics to Predict the PD-L1 Immunotherapy and Prognosis of Gastric Cancer.
  • May 1, 2024
  • Current medicinal chemistry
  • Chenchen Wang + 4 more

We aimed to develop a prognostic model with stemness-correlated genes to evaluate prognosis and immunotherapy responsiveness in gastric cancer (GC). Tumor stemness is related to intratumoral heterogeneity, immunosuppression, and anti-tumor resistance. We developed a prognostic model with stemness-correlated genes to evaluate prognosis and immunotherapy responsiveness in GC. We aimed to develop a prognostic model with stemness-correlated genes to evaluate prognosis and immunotherapy responsiveness in GC. We downloaded single-cell RNA sequencing (scRNA-seq) data of GC patients from the Gene-Expression Omnibus (GEO) database and screened GC stemness- related genes using CytoTRACE. We characterized the association of tumor stemness with immune checkpoint blockade (ICB) and immunity. Thereafter, a 9-stemness signature-based prognostic model was developed using weighted gene co-expression network analysis (WGCNA), univariate Cox regression analysis, and the least absolute shrinkage and selection operator (LASSO) regression analysis. The model predictive value was evaluated with a nomogram. Early GC patients had significantly higher levels of stemness. The stemness score showed a negative relationship to tumor immune dysfunction and exclusion (TIDE) score and immune infiltration, especially T cells and B cells. A stemness-based signature based on 9 genes (ERCC6L, IQCC, NKAPD1, BLMH, SLC25A15, MRPL4, VPS35, SUMO3, and CINP) was constructed with good performance in prognosis prediction, and its robustness was validated in GSE26942 cohort. Additionally, nomogram and risk score exhibited the most powerful ability for prognosis prediction. High-risk patients exhibited a tendency to develop immune escape and low response to PD-L1 immunotherapy. We developed a stemness-based gene signature for prognosis prediction with accuracy and reliability. This signature also helps clinical decision-making of immunotherapy for GC patients.

  • Components
  • 10.7717/peerj.12605/fig-5
Figure 5: The correlation between ANXA9 expression and prognosis in GC.
  • Dec 15, 2021

Background Gastric cancer (GC) is the most prevalent malignancy among the digestive system tumors. Increasing evidence has revealed that lower mRNA expression of ANXA9 is associated with a poor prognosis in colorectal cancer. However, the role of ANXA9 in GC remains largely unknown. Material and Methods The Gene Expression Profiling Interactive Analysis (GEPIA) and Human Protein Atlas databases were used to investigate the expression of ANXA9 in GC, which was then validated in the four Gene Expression Omnibus (GEO) datasets. The diagnostic value of ANXA9 for GC patients was demonstrated using a receiver operating characteristic (ROC) curve. The correlation between ANXA9 expression and clinicopathological parameters was analyzed in The Cancer Genome Atlas (TCGA) and UALCAN databases. The Kaplan-Meier (K-M) survival curve was used to elucidate the relationship between ANXA9 expression and the survival time of GC patients. We then performed a gene set enrichment analysis (GSEA) to explore the biological functions of ANXA9. The relationship of ANXA9 expression and cancer immune infiltrates was analyzed using the Tumor Immune Estimation Resource (TIMER). In addition, the potential mechanism of ANXA9 in GC was investigated by analyzing its related genes. Results ANXA9 was significantly up-regulated in GC tissues and showed obvious diagnostic value. The expression of ANXA9 was related to the age, gender, grade, TP53 mutation, and histological subtype of GC patients. We also found that ANXA9 expression was associated with immune-related biological function. ANXA9 expression was also correlated with the infiltration level of CD8+ T cells, neutrophils, and dendritic cells in GC. Additionally, copy number variation (VNV) of ANXA9 occurred in GC patients. Function enrichment analyses revealed that ANXA9 plays a role in the GC progression by interacting with its related genes. Conclusions Our results provide strong evidence of ANXA9 expression as a prognostic indicator related to immune responses in GC.

  • Research Article
  • Cite Count Icon 25
  • 10.21037/jgo-21-325
Differential analysis of RNA methylation regulators in gastric cancer based on TCGA data set and construction of a prognostic model
  • Aug 1, 2021
  • Journal of Gastrointestinal Oncology
  • Jing Li + 6 more

Methylation is one of the common forms of RNA modification, which mainly include N6-methyladenosine (m6A), C5-methylcytidine (m5C), and N1-methyladenosine (m1A). Numerous studies have shown that RNA methylation is associated with tumor development. We aim to construct prognostic models of gastric cancer based on RNA methylation regulators. The transcriptome and clinical data of gastric cancer and normal samples were obtained from the National Cancer Institute Genome Data Commons (NCI-GDC). Use Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression analysis to construct risk models for different types of RNA methylation. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive efficiency of risk characteristics. Cluster heat maps are used to assess the correlation with clinical information. Univariate and multivariate Cox analyses were used to analyze prognostic effects of risk scores. Gene Set Enrichment Analysis (GSEA) analyzes the functional enrichment of RNA methylation genes. And make a separate analysis of the data of Asians. The expression of most of the 30 RNA methylation regulators were significantly different in cancer and paracancerous tissues (P<0.05). Three methylated genes (FTO, ALKBH5, and RBM15) were screened from m6A by LASSO Cox regression analysis. Five methylated genes (FTO, ALKBH5, TRMT61B, RBM15, and YXB1) were selected from the population, and were used to construct two risk ratio models. Survival analysis showed that the survival rate of patients in the low-risk group was significantly higher than that in the high-risk group (P<0.05). All ROC curves indicated that the predictive efficiency of risk characteristics was good [area under the ROC curve (AUC): 0.6-1].Cluster analysis reveals differences in clinical data between the two groups. Univariate and multivariate Cox regression results show that the risk score has independent prognostic value. GSEA showed that pathways such as cell cycle were significantly enriched in the low-risk group, while pathways such as calcium signaling pathway were significantly enriched in the high-risk group. In addition, three methylation models that can predict the prognosis of Asian gastric cancer patients were obtained. The methylation prognosis model constructed in this study can effectively predict the prognosis of gastric cancer patients.

  • Research Article
  • Cite Count Icon 3
  • 10.4251/wjgo.v16.i7.3011
Adipocytes impact on gastric cancer progression: Prognostic insights and molecular features.
  • Jul 15, 2024
  • World journal of gastrointestinal oncology
  • Jia-Rong Shang + 6 more

Adipocytes, especially adipocytes within tumor tissue known as cancer-associated adipocytes, have been increasingly recognized for their pivotal role in the tumor microenvironment of gastric cancer (GC). Their influence on tumor progression and patient prognosis has sparked significant interest in recent research. The main objectives of this study were to investigate adipocyte infiltration, assess its correlation with clinical pathological features, develop a prognostic prediction model based on independent prognostic factors, evaluate the impact of adipocytes on immune cell infiltration and tumor invasiveness in GC, and identify and validate genes associated with high adipocyte expression, exploring their potential diagnostic and prognostic value. To explore the relationship between increased adipocytes within tumor tissue and prognosis in GC patients as well as the associated mechanisms and potential biomarkers, using public databases and clinical data. Using mRNA microarray datasets from the Gene Expression Omnibus database and clinical samples from Jiangsu Provincial Hospital, survival and regression analyses were conducted to determine the relevant prognostic factors in GC. Feature gene selection was performed using least absolute shrinkage and selection operator and support vector machine recursive feature elimination algorithms, followed by differential gene expression analysis, gene ontology, pathway analysis, and Gene Set Enrichment Analysis. Immune cell infiltration was analyzed using the CIBERSORT algorithm. Tumor adipocyte infiltration correlated with poor prognosis in GC, leading to the development of a highly accurate and discriminative prognostic prediction model. Key genes, ADH1B, SFRP1, PLAC9, and FABP4, were identified as associated with high adipocyte expression in GC. The diagnostic and prognostic potential of these identified genes was validated using independent datasets. Downregulation of immune cells was observed in GC with high adipocyte expression. GC with high intratumoral adipocyte expression demonstrated aggressive tumor biology and a poorer prognosis. The genes ADH1B, SFRP1, PLAC9, and FABP4 have been identified as holding diagnostic and prognostic significance in GC. These findings strongly support the use of adipocyte expression as a valuable indicator of tumor invasiveness and anticipated patient outcomes in GC.

More from: Translational Cancer Research
  • Research Article
  • 10.21037/tcr-2025-989
Bioinformatics analysis identifies ULK3 as a novel prognostic and immune-related biomarker in esophageal cancer
  • Oct 1, 2025
  • Translational Cancer Research
  • Kai Cui + 4 more

  • Research Article
  • 10.21037/tcr-2025-957
RNA sequencing and bioinformatics analysis have identified MAST1 as a potential biomarker and therapeutic target for cervical cancer
  • Oct 1, 2025
  • Translational Cancer Research
  • Zhijia Xie + 6 more

  • Research Article
  • 10.21037/tcr-2025-844
Intratumoral disulfidptosis heterogeneity in triple-negative breast cancer, a multiomics integration analysis
  • Oct 1, 2025
  • Translational Cancer Research
  • Zi-Xian Dong + 3 more

  • Research Article
  • 10.21037/tcr-2025-309
Demystifying programmed cell death in lung adenocarcinoma: combined prognostic model construction
  • Sep 27, 2025
  • Translational Cancer Research
  • Yu Li + 5 more

  • Research Article
  • 10.21037/tcr-2024-2651
N6-adenosine methyltransferase METTL4 is a potential prognostic biomarker and associates with cell cycle regulation and immune infiltration in glioma
  • Sep 26, 2025
  • Translational Cancer Research
  • Shuo Zhang + 10 more

  • Research Article
  • 10.21037/tcr-2025-1865
Association between the age-adjusted visceral adiposity index (AVAI) and gynecologic malignancies: a cross-sectional study based on NHANES data
  • Sep 26, 2025
  • Translational Cancer Research
  • Xincheng Zhang + 7 more

  • Research Article
  • 10.21037/tcr-2025-1954
METRN modulates immune escape via CD8+ T cells in breast cancer progression through IL-6 signaling pathway
  • Sep 26, 2025
  • Translational Cancer Research
  • Min Du + 9 more

  • Research Article
  • 10.21037/tcr-2024-2585
Predictive value of random forest prediction model based on postoperative adjuvant transcatheter arterial chemoembolization therapy for survival of hepatocellular carcinoma patients with microvascular invasion: a retrospective cohort study
  • Sep 26, 2025
  • Translational Cancer Research
  • Yanran Dou + 3 more

  • Research Article
  • 10.21037/tcr-2025-1874
Screening of PANoptosis regulator-associated long noncoding RNAs and construction of a survival prognostic model in cutaneous melanoma
  • Sep 26, 2025
  • Translational Cancer Research
  • Jingshu Xiong + 7 more

  • Research Article
  • 10.21037/tcr-2025-1774
Clinical features as prognostic markers in patients with solid tumors treated with programmed cell death protein 1/programmed cell death-ligand 1 inhibitors: a retrospective cohort study
  • Sep 26, 2025
  • Translational Cancer Research
  • Yanli Liu + 7 more

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon