Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 14(1): 10348, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710798

ABSTRACT

The complete compound of gefitinib is effective in the treatment of lung adenocarcinoma. However, the effect on lung adenocarcinoma (LUAD) during its catabolism has not yet been elucidated. We carried out this study to examine the predictive value of gefitinib metabolism-related long noncoding RNAs (GMLncs) in LUAD patients. To filter GMLncs and create a prognostic model, we employed Pearson correlation, Lasso, univariate Cox, and multivariate Cox analysis. We combined risk scores and clinical features to create nomograms for better application in clinical settings. According to the constructed prognostic model, we performed GO/KEGG and GSEA enrichment analysis, tumor immune microenvironment analysis, immune evasion and immunotherapy analysis, somatic cell mutation analysis, drug sensitivity analysis, IMvigor210 immunotherapy validation, stem cell index analysis and real-time quantitative PCR (RT-qPCR) analysis. We built a predictive model with 9 GMLncs, which showed good predictive performance in validation and training sets. The calibration curve demonstrated excellent agreement between the expected and observed survival rates, for which the predictive performance was better than that of the nomogram without a risk score. The metabolism of gefitinib is related to the cytochrome P450 pathway and lipid metabolism pathway, and may be one of the causes of gefitinib resistance, according to analyses from the Gene Set Enrichment Analysis (GSEA), Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Immunological evasion and immunotherapy analysis revealed that the likelihood of immune evasion increased with risk score. Tumor microenvironment analysis found most immune cells at higher concentrations in the low-risk group. Drug sensitivity analysis found 23 sensitive drugs. Twenty-one of these drugs exhibited heightened sensitivity in the high-risk group. RT-qPCR analysis validated the characteristics of 9 GMlncs. The predictive model and nomogram that we constructed have good application value in evaluating the prognosis of patients and guiding clinical treatment.


Subject(s)
Adenocarcinoma of Lung , Drug Resistance, Neoplasm , Gefitinib , Lung Neoplasms , RNA, Long Noncoding , Tumor Microenvironment , Humans , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Gefitinib/therapeutic use , Gefitinib/pharmacology , RNA, Long Noncoding/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Prognosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Nomograms , Female , Male , Gene Expression Regulation, Neoplastic , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Middle Aged , Aged
2.
Aging (Albany NY) ; 16(3): 2273-2298, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38319706

ABSTRACT

BACKGROUND: Methods for predicting the outcome of lung adenocarcinoma (LUAD) in the clinic are limited. Anoikis is an important route to programmed cell death in LUAD, and the prognostic value of a model constructed with anoikis-related lncRNAs (ARlncRNAs) in LUAD is unclear. METHODS: Transcriptome and basic information for LUAD patients was obtained from the Cancer Genome Atlas. Coexpression and Cox regression analyses were utilized to identify prognostically significant ARlncRNAs and construct a prognostic signature. Furthermore, the signature was combined with clinical characteristics to create a nomogram. Finally, we performed principal component, enrichment, tumor mutation burden (TMB), tumor microenvironment (TME) and drug sensitivity analyses to evaluate the basic research and clinical merit of the signature. RESULTS: The prognostic signature developed with eleven ARlncRNAs can accurately predict that high-risk group patients have a worse prognosis, as proven by the receiver operating characteristic (ROC) curve (AUC: 0.718). Independent prognostic analyses indicated that the risk score is a significant independent prognostic element for LUAD (P<0.001). In the high-risk group, enrichment analysis demonstrated that glucose metabolism and DNA replication were the main enrichment pathways. TMB analysis indicated that the high-risk group had a high TMB (P<0.05). Drug sensitivity analyses can recognize drugs that are sensitive to different risk groups. Finally, 11 ARlncRNAs of this signature were verified by RT-qPCR analysis. CONCLUSIONS: A novel prognostic signature developed with 11 ARlncRNAs can accurately predict the OS of LUAD patients and offer clinical guidance value for immunotherapy and chemotherapy treatment.


Subject(s)
Adenocarcinoma , Lung Neoplasms , RNA, Long Noncoding , Humans , Anoikis/genetics , Prognosis , RNA, Long Noncoding/genetics , Lung , Lung Neoplasms/genetics , Tumor Microenvironment/genetics
3.
Sci Rep ; 13(1): 19151, 2023 11 06.
Article in English | MEDLINE | ID: mdl-37932413

ABSTRACT

Immunogenic cell death (ICD) has been demonstrated to activate T cells to kill tumor cells, which is closely related to tumor development, and long noncoding RNAs (lncRNAs) are also involved. However, it is not known whether ICD-related lncRNAs are associated with the development of lung adenocarcinoma (LUAD). We downloaded ICD-related genes from GeneCards and the transcriptome statistics of LUAD patients from The Cancer Genome Atlas (TCGA) and subsequently developed and verified a predictive model. A successful model was used together with other clinical features to construct a nomogram for predicting patient survival. To further study the mechanism of tumor action and to guide therapy, we performed enrichment analysis, tumor microenvironment analysis, somatic mutation analysis, drug sensitivity analysis and real-time quantitative polymerase chain reaction (RT-qPCR) analysis. Nine ICD-related lncRNAs with significant prognostic relevance were selected for model construction. Survival analysis demonstrated that overall survival was substantially shorter in the high-risk group than in the low-risk group (P < 0.001). This model was predictive of prognosis across all clinical subgroups. Cox regression analysis further supported the independent prediction ability of the model. Ultimately, a nomogram depending on stage and risk score was created and showed a better predictive performance than the nomogram without the risk score. Through enrichment analysis, the enriched pathways in the high-risk group were found to be primarily associated with metabolism and DNA replication. Tumor microenvironment analysis suggested that the immune cell concentration was lower in the high-risk group. Somatic mutation analysis revealed that the high-risk group contained more tumor mutations (P = 0.00018). Tumor immune dysfunction and exclusion scores exhibited greater sensitivity to immunotherapy in the high-risk group (P < 0.001). Drug sensitivity analysis suggested that the predictive model can also be applied to the choice of chemotherapy drugs. RT-qPCR analysis also validated the accuracy of the constructed model based on nine ICD-related lncRNAs. The prognostic model constructed based on the nine ICD-related lncRNAs showed good application value in assessing prognosis and guiding clinical therapy.


Subject(s)
Adenocarcinoma , RNA, Long Noncoding , Humans , Prognosis , Immunogenic Cell Death , Lung , Tumor Microenvironment
4.
Aging (Albany NY) ; 15(19): 10473-10500, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37812189

ABSTRACT

BACKGROUND: Precisely forecasting the prognosis of esophageal squamous cell carcinoma (ESCC) patients is a formidable challenge. Cuproptosis has been implicated in ESCC pathogenesis; however, the prognostic value of cuproptosis-associated long noncoding RNAs (CuRLs) in ESCC is unclear. METHODS: Transcriptomic and clinical data related to ESCC were sourced from The Cancer Genome Atlas (TCGA). Using coexpression and Cox regression analysis to identify prognostically significant CuRLs, a prognostic signature was created. Nomogram models were established by incorporating the risk score and clinical characteristics. Tumor Immune Dysfunction and Rejection (TIDE) scores were derived by conducting an immune landscape analysis and evaluating the tumor mutational burden (TMB). Drug sensitivity analysis was performed to explore the underlying molecular mechanisms and guide clinical dosing. RESULTS: Our risk score based on 5 CuRLs accurately predicted poorer prognosis in high-risk ESCC patients across almost all subgroups. The nomogram that included the risk score provided more precise prognostic predictions. Immune pathways, such as the B-cell receptor signaling pathway, were enriched in the datasets from high-risk patients. High TMB in high-risk patients indicated a relatively poor prognosis. High-risk patients with lower TIDE scores were found to benefit more from immunotherapy. High-risk patients exhibited greater responsiveness to Nilotinib, BI-2536, P22077, Zoledronate, and Fulvestrant, as revealed by drug sensitivity analysis. Real-time PCR validation demonstrated significant differential expression of four CuRLs between ESCC and normal cell lines. CONCLUSIONS: The above risk score and nomogram can accurately predict prognosis in ESCC patients and provide guidance for chemotherapy and immunotherapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , RNA, Long Noncoding , Humans , Prognosis , Esophageal Squamous Cell Carcinoma/genetics , RNA, Long Noncoding/genetics , Esophageal Neoplasms/genetics , Nomograms , Apoptosis
5.
Aging (Albany NY) ; 15(12): 5304-5338, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37379129

ABSTRACT

BACKGROUND: Immunogenic cell death (ICD) is an important part of the antitumor effect, yet the role played by long noncoding RNAs (lncRNAs) remains unclear. We explored the value of ICD-related lncRNAs in tumor prognosis assessment in kidney renal clear cell carcinoma (KIRC) patients to provide a basis for answering the above questions. METHODS: Data on KIRC patients were obtained from The Cancer Genome Atlas (TCGA) database, prognostic markers were identified, and their accuracy was verified. An application-validated nomogram was developed based on this information. Furthermore, we performed enrichment analysis, tumor mutational burden (TMB) analysis, tumor microenvironment (TME) analysis, and drug sensitivity prediction to explore the mechanism of action and clinical application value of the model. RT-qPCR was performed to detect the expression of lncRNAs. RESULTS: The risk assessment model constructed using eight ICD-related lncRNAs provided insight into patient prognoses. Kaplan-Meier (K-M) survival curves showed a more unfavorable outcome in high-risk patients (p<0.001). The model had good predictive value for different clinical subgroups, and the nomogram constructed based on this model worked well (risk score AUC=0.765). Enrichment analysis revealed that mitochondrial function-related pathways were enriched in the low-risk group. The adverse prognosis of the higher-risk cohort might correspond to a higher TMB. The TME analysis revealed a higher resistance to immunotherapy in the increased-risk subgroup. Drug sensitivity analysis can guide the selection and application of antitumor drugs in different risk groups. CONCLUSIONS: This prognostic signature based on eight ICD-associated lncRNAs has significant implications for prognostic assessment and treatment selection in KIRC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Immunogenic Cell Death , Prognosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney , Tumor Microenvironment/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...