Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.518
Filter
1.
Cancer Immunol Immunother ; 73(6): 112, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38693422

ABSTRACT

OBJECTIVE: The high mortality rate of gastric cancer, traditionally managed through surgery, underscores the urgent need for advanced therapeutic strategies. Despite advancements in treatment modalities, outcomes remain suboptimal, necessitating the identification of novel biomarkers to predict sensitivity to immunotherapy. This study focuses on utilizing single-cell sequencing for gene identification and developing a random forest model to predict immunotherapy sensitivity in gastric cancer patients. METHODS: Differentially expressed genes were identified using single-cell RNA sequencing (scRNA-seq) and gene set enrichment analysis (GESA). A random forest model was constructed based on these genes, and its effectiveness was validated through prognostic analysis. Further, analyses of immune cell infiltration, immune checkpoints, and the random forest model provided deeper insights. RESULTS: High METTL1 expression was found to correlate with improved survival rates in gastric cancer patients (P = 0.042), and the random forest model, based on METTL1 and associated prognostic genes, achieved a significant predictive performance (AUC = 0.863). It showed associations with various immune cell types and negative correlations with CTLA4 and PDCD1 immune checkpoints. Experiments in vitro and in vivo demonstrated that METTL1 enhances gastric cancer cell activity by suppressing T cell proliferation and upregulating CTLA4 and PDCD1. CONCLUSION: The random forest model, based on scRNA-seq, shows high predictive value for survival and immunotherapy sensitivity in gastric cancer patients. This study underscores the potential of METTL1 as a biomarker in enhancing the efficacy of gastric cancer immunotherapy.


Subject(s)
Immunotherapy , Single-Cell Analysis , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/mortality , Humans , Single-Cell Analysis/methods , Immunotherapy/methods , Animals , Mice , Prognosis , Biomarkers, Tumor/genetics , Sequence Analysis, RNA/methods , Female , Male , Gene Expression Regulation, Neoplastic , Xenograft Model Antitumor Assays , Cell Line, Tumor , Random Forest
2.
BMC Cancer ; 24(1): 570, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714987

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most prevalent malignant tumors worldwide and is associated with high morbidity and mortality rates. However, the specific biomarkers used to predict the postoperative prognosis of patients with gastric cancer remain unknown. Recent research has shown that the tumor microenvironment (TME) has an increasingly positive effect on anti-tumor activity. This study aims to build signatures to study the effect of certain genes on gastric cancer. METHODS: Expression profiles of 37 T cell-related genes and their TME characteristics were comprehensively analyzed. A risk signature was constructed and validated based on the screened T cell-related genes, and the roles of hub genes in GC were experimentally validated. RESULTS: A novel T cell-related gene signature was constructed based on CD5, ABCA8, SERPINE2, ESM1, SERPINA5, and NMU. The high-risk group indicated lower overall survival (OS), poorer immune efficacy, and higher drug resistance, with SERPINE2 promoting GC cell proliferation, according to experiments. SERPINE2 and CXCL12 were significantly correlated, indicating poor OS via the Youjiang cohort. CONCLUSIONS: This study identified T cell-related genes in patients with stomach adenocarcinoma (STAD) for prognosis estimation and proposed potential immunotherapeutic targets for STAD.


Subject(s)
Adenocarcinoma , Biomarkers, Tumor , Stomach Neoplasms , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Prognosis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , T-Lymphocytes, Regulatory/immunology , Gene Expression Profiling , Male , Female
3.
Anticancer Res ; 44(5): 2231-2238, 2024 May.
Article in English | MEDLINE | ID: mdl-38677750

ABSTRACT

BACKGROUND/AIM: The prognostic immune and nutritional index (PINI) was developed and reported as a promising prognostic factor. This study aimed to clarify the clinical impact of the PINI in gastric cancer (GC) patients who received curative treatment. PATIENTS AND METHODS: Patients who underwent curative resection for GC at Yokohama City University between 2005 and 2020 were selected based on their medical records. The PINI was calculated by dividing the serum albumin concentration (g/dl) by the serum monocyte concentration. Both measurements were performed prior to surgery. RESULTS: A total of 262 patients were included in this study. Based on the 3- and 5-year overall survival (OS), we set the cutoff value of the PINI at 3.4 in the present study. The 3- and 5-year OS rates were 69.0% and 66.1%, respectively, in the PINI-low group and 90.5% and 82.8% in the PINI-high group. There were significant differences between the two groups (p<0.001). A multivariate analysis of factors associated with OS identified PINI as an independent prognostic factor (hazard ratio=1.996; 95% confidence interval=1.096-3.636, p=0.024). Similar results were observed for RFS. In addition, the PINI status affected the recurrence pattern, postoperative anastomotic leakage, and the introduction of postoperative adjuvant chemotherapy. CONCLUSION: The PINI is a promising nutritional and inflammatory marker for patients with GC and might be a useful marker for the treatment and management of patients with GC.


Subject(s)
Nutrition Assessment , Stomach Neoplasms , Humans , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Stomach Neoplasms/therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/blood , Male , Female , Aged , Prognosis , Middle Aged , Nutritional Status , Adult , Aged, 80 and over , Retrospective Studies , Serum Albumin/analysis , Serum Albumin/metabolism , Gastrectomy
4.
Anticancer Res ; 44(5): 1877-1883, 2024 May.
Article in English | MEDLINE | ID: mdl-38677758

ABSTRACT

BACKGROUND/AIM: Human gastric cancer stem-like cells (CSCs)/cancer-initiating cells can be identified as aldehyde dehydrogenase-high (ALDHhigh) cells. Cancer immunotherapy employing immune checkpoint blockade has been approved for advanced gastric cancer cases. However, the effectiveness of cancer immunotherapy against gastric CSCs/CICs remains unclear. This study aimed to investigate the susceptibility of gastric CSCs/CICs to immunotherapy. MATERIALS AND METHODS: Gastric CSCs/CICs were isolated as ALDHhigh cells using the human gastric cancer cell line, MKN-45. ALDHhigh clone cells and ALDHlow clone cells were isolated using the ALDEFLUOR assay. ALDH1A1 expression was assessed via qRT-PCR. Sphere-forming ability was evaluated to confirm the presence of CSCs/CICs. A model neoantigen, AP2S1, was over-expressed in ALDHhigh clone cells and ALDHlow clone cells, and susceptibility to AP2S1-specific TCR-T cells was assessed using IFNγ ELISPOT assay. RESULTS: Three ALDHhigh clone cells were isolated from MKN-45 cells. ALDHhigh clone cells exhibited a stable phenotype in in vitro culture for more than 2 months. The High-36 clone cells demonstrated the highest sphere-forming ability, whereas the Low-8 cells showed the lowest sphere-forming ability. High-36 cells exhibited lower expression of HLA-A24 compared to Low-8 cells. TCR-T cells specific for AP2S1 showed lower reactivity to High-36 cells compared to Low-8 cells. CONCLUSION: High-36 cells and Low-8 cells represent novel gastric CSCs/CICs and non-CSCs/CICs, respectively. ALDHhigh CSCs/CICs evade T cells due to lower expression of HLA class 1.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Neoplastic Stem Cells , Stomach Neoplasms , T-Lymphocytes, Cytotoxic , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/pathology , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Cell Line, Tumor , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Retinal Dehydrogenase/metabolism , Tumor Escape/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/immunology
5.
Int Immunopharmacol ; 132: 111998, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38593510

ABSTRACT

BACKGROUND: Given the intricate molecular complexities and heterogeneity inherent in T-cell immunotherapy of gastric cancer (GC), elucidative T-cell-related biomarkers were imperative needed for facilitating the prediction of GC patient prognosis and identify potential synergistic therapeutic targets. METHODS: We conducted COX regression analysis in TISIDB, TCGA-STAD, and GEO databases to identify 19 GC T-cell-mediated sensitivity tumor killing (TTK) genes (key GCTTKs). Based on key GCTTKs, we constructed two TTK patterns and analyzed their metabolic pathways, mutation features, clinical data distribution, immune cell infiltration, and prognosis. LASSO regression was performed to develop a T-cell-mediated GC Prognosis (TGCP) model. We validated the TGCP model in GC patients. TAP1 was further selected for investigation of its biological functions and molecular mechanisms. We assessed the potential of TAP1 as a promising therapeutic target for GC using Patient-derived organoids (PDOs)-derived xenografts (PDOXs) models of GC. RESULTS: The TTK patterns display notable disparities. The TGCP model showcases its proficiency in predicting immune response efficacy, effectively distinguishes immunotherapy difference GC patients. Our findings find further confirmation in PDOX models, affirming TAP1 can enhance immunotherapy facilitated by PDL1 inhibitors. Furthermore, Oxaliplatin, by promoting TAP1 expression, augments PDL1 expression, thereby enhancing the efficacy of immunotherapy. CONCLUSIONS: We constructed a TGCP model, which demonstrates satisfactory predictive accuracy. Out of 9 prognostic genes, TAP1 was validated as a synergistic target for Oxaliplatin and PDL1 inhibitors, offering a genetic-level explanation for the synergy observed in GC treatment involving Oxaliplatin in combination with PDL1 inhibitors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 2 , Immunotherapy , Oxaliplatin , Stomach Neoplasms , Stomach Neoplasms/immunology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/therapy , Stomach Neoplasms/genetics , Humans , Animals , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Immunotherapy/methods , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , Mice , T-Lymphocytes/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Drug Synergism , Prognosis
6.
Aging (Albany NY) ; 16(8): 7331-7356, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38656888

ABSTRACT

BACKGROUND: Patients with gastric cancer respond poorly to immunotherapy. There are still unknowns about the biomarkers associated with immunotherapy sensitivity and their underlying molecular mechanisms. METHODS: Gene expression data for gastric cancer were gathered from TCGA and GEO databases. DEGs associated with immunotherapy response came from ICBatlas. KEGG and GO analyses investigated pathways. Hub genes identification employed multiple machine algorithms. Associations between hub genes and signaling pathways, disease genes, immune cell infiltration, drug sensitivity, and prognostic predictions were explored via multi-omics analysis. Hub gene expression was validated through HPA and CCLE. Multiple algorithms pinpointed Cancer-Associated Fibroblasts genes (CAFs), with ten machine-learning methods generating CAFs scores for prognosis. Model gene expression was validated at the single-cell level using the TISCH database. RESULTS: We identified 201 upregulated and 935 downregulated DEGs. Three hub genes, namely CDH6, EGFLAM, and RASGRF2, were unveiled. These genes are implicated in diverse disease-related signaling pathways. Additionally, they exhibited significant correlations with disease-associated gene expression, immune cell infiltration, and drug sensitivity. Exploration of the HPA and CCLE databases exposed substantial expression variations across patients and cell lines for these genes. Subsequently, we identified CAFs-associated genes and established a robust prognostic model. The analysis in the TISCH database showed that the genes in this model were highly expressed in CAFs. CONCLUSIONS: The results unveil an association between CDH6, EGFLAM, and RASGRF2 and the immunotherapeutic response in gastric cancer. These genes hold potential as predictive biomarkers for gastric cancer immunotherapy resistance and prognostic assessment.


Subject(s)
Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Immunotherapy , Machine Learning , Stomach Neoplasms , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/therapy , Humans , Immunotherapy/methods , Drug Resistance, Neoplasm/genetics , Biomarkers, Tumor/genetics , Prognosis , Gene Expression Profiling , Databases, Genetic , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/immunology , Multiomics
7.
Drug Resist Updat ; 74: 101080, 2024 May.
Article in English | MEDLINE | ID: mdl-38579635

ABSTRACT

BACKGROUND: Gastric Cancer (GC) characteristically exhibits heterogeneous responses to treatment, particularly in relation to immuno plus chemo therapy, necessitating a precision medicine approach. This study is centered around delineating the cellular and molecular underpinnings of drug resistance in this context. METHODS: We undertook a comprehensive multi-omics exploration of postoperative tissues from GC patients undergoing the chemo and immuno-treatment regimen. Concurrently, an image deep learning model was developed to predict treatment responsiveness. RESULTS: Our initial findings associate apical membrane cells with resistance to fluorouracil and oxaliplatin, critical constituents of the therapy. Further investigation into this cell population shed light on substantial interactions with resident macrophages, underscoring the role of intercellular communication in shaping treatment resistance. Subsequent ligand-receptor analysis unveiled specific molecular dialogues, most notably TGFB1-HSPB1 and LTF-S100A14, offering insights into potential signaling pathways implicated in resistance. Our SVM model, incorporating these multi-omics and spatial data, demonstrated significant predictive power, with AUC values of 0.93 and 0.84 in the exploration and validation cohorts respectively. Hence, our results underscore the utility of multi-omics and spatial data in modeling treatment response. CONCLUSION: Our integrative approach, amalgamating mIHC assays, feature extraction, and machine learning, successfully unraveled the complex cellular interplay underlying drug resistance. This robust predictive model may serve as a valuable tool for personalizing therapeutic strategies and enhancing treatment outcomes in gastric cancer.


Subject(s)
Drug Resistance, Neoplasm , Fluorouracil , Stomach Neoplasms , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Humans , Drug Resistance, Neoplasm/drug effects , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Oxaliplatin/pharmacology , Oxaliplatin/administration & dosage , Oxaliplatin/therapeutic use , Deep Learning , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Precision Medicine/methods , Male , Female , Middle Aged , Immunotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Signal Transduction/drug effects , Multiomics
8.
Pathol Res Pract ; 257: 155310, 2024 May.
Article in English | MEDLINE | ID: mdl-38663178

ABSTRACT

Gastric carcinoma (GC) is a high heterogeneity and malignant tumor with a poor prognosis. The current implementation of immunotherapy in GC is limited due to the insufficient exploration of immune-related mutations and speculated early mutation events. Therefore, we performed whole-exome sequencing on 40 patients with GC to explore their genetic characteristics, shedding light on the order of genetic events, somatic mutations impacting the immune microenvironment, and potential biomarkers for immunotherapy. Regarding genetic events, TP53 disruptions were identified as frequent and early events in GC progression, often occurring alongside other gene mutations. The mutations occurring in GANS, SMAD4, and POLE were early independent events. Patients harboring CSMD3, FAT4, FLG, KMT2C, LRP1B, MUC5B, MUC16, PLEC, RNF43, SYNE1, TP53, TTN, XIRP2, and ZFHX4 mutations tended to have decreased B cells, T cells, macrophage, neutrophil, and dendritic cells infiltration, except for the ARID1A gene mutations. We also found patients with microsatellite instability-high tumors had higher homologous recombination deficiency (HRD) scores. HRD showed a positive correlation with tumor mutational burden, which might serve as indirect evidence supporting the potential of HRD as a biomarker for GC. These findings highlighted GC's high heterogeneity and complexity and provided valuable insights into the somatic mutations that affect early genetic progression and immune microenvironment.


Subject(s)
Mutation , Stomach Neoplasms , Tumor Microenvironment , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Male , Female , Middle Aged , Biomarkers, Tumor/genetics , Aged , Disease Progression , Exome Sequencing , Adult
9.
Radiother Oncol ; 194: 110213, 2024 May.
Article in English | MEDLINE | ID: mdl-38458258

ABSTRACT

BACKGROUND AND PURPOSE: Poor penetration of transferred T cells represents a critical factor impeding the development of adoptive cell therapy in solid tumors. We demonstrated that iRGD-antiCD3 modification promoted both T cell infiltration and activation in our previous work. Interest in low-dose radiotherapy has recently been renewed due to its immuno-stimulatory effects including T cell recruitment. This study aims to explore the synergistic effects between low-dose radiotherapy and iRGD-antiCD3-modified T cells. MATERIALS AND METHODS: Flow cytometry was performed to assess the expression of iRGD receptors and chemokines. T cell infiltration was evaluated by immunohistofluorescence and in vivo real-time fluorescence imaging and antitumor effects were investigated by in vivo bioluminescence imaging in the gastric cancer peritoneal metastasis mouse model. RESULTS: We found that 2 Gy irradiation upregulated the expression of all three iRGD receptors and T-cell chemokines. The addition of 2 Gy low-dose irradiation boosted the accumulation and penetration of iRGD-antiCD3-modified T cells in peritoneal tumor nodules. Combining 2 Gy low-dose irradiation with iRGD-antiCD3-modified T cells significantly inhibited tumor growth and prolonged survival in the peritoneal metastasis mouse model with a favorable safety profile. CONCLUSION: Altogether, we demonstrated that low-dose radiotherapy could improve the antitumor potency of iRGD-antiCD3-modified T cells by promoting T cell infiltration, providing a rationale for exploring low-dose radiotherapy in combination of other adoptive T cell therapies in solid tumors.


Subject(s)
Stomach Neoplasms , T-Lymphocytes , Animals , Mice , Stomach Neoplasms/radiotherapy , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , T-Lymphocytes/radiation effects , T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Radiotherapy Dosage , Oligopeptides , Peritoneal Neoplasms/radiotherapy , Peritoneal Neoplasms/secondary , Cell Line, Tumor , Female , Combined Modality Therapy
10.
Eur J Gastroenterol Hepatol ; 36(6): 687-694, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38526941

ABSTRACT

The objective of this meta-analysis is to delineate the association between H. pylori CagA serological status and the prevalence of gastric precancerous lesions (GPL). We searched peer-reviewed articles up to October 2023. The extraction of data from the included studies was carried out as well as the quality assessment. Pooled effect sizes were calculated using a random effect model. Thirteen studies met the inclusion criteria, comprising 2728 patients with GPL and 17 612 controls. The aggregate odds ratio (OR) for the association between serum CagA and GPL was 2.74 (95% CI = 2.25-3.32; P  = 0.00; I 2  = 60.4%), irrespective of H. pylori infection status. Within the H. pylori -infected cohort, the OR was 2.25 (95% CI = 1.99-2.56; P  = 0.00; I 2  = 0.0%). Conversely, among the non-infected individuals, the OR was 1.63 (95% CI = 1.04-2.54; P  = 0.038; I 2  = 0.0%). Heterogeneity was explored using subgroup and meta-regression analyses, indicating that the variability between studies likely stemmed from differences in disease classification. Our results demonstrated robustness and negligible publication bias. The meta-analysis underscores a more pronounced association between H. pylori CagA seropositivity and the risk of developing GPL than between seronegativity and the same risk, irrespective of H. pylori infection status at the time. Additionally, the strength of the association was heightened in the presence of an active H. pylori infection. The implications of these findings advocate for the utility of CagA serostatus as a potential biomarker for screening GPL.


Subject(s)
Antigens, Bacterial , Bacterial Proteins , Helicobacter Infections , Helicobacter pylori , Precancerous Conditions , Stomach Neoplasms , Humans , Antigens, Bacterial/immunology , Antigens, Bacterial/blood , Helicobacter pylori/immunology , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Bacterial Proteins/immunology , Stomach Neoplasms/microbiology , Stomach Neoplasms/blood , Stomach Neoplasms/immunology , Precancerous Conditions/microbiology , Precancerous Conditions/blood , Risk Factors , Prevalence
11.
BMB Rep ; 57(4): 188-193, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38449302

ABSTRACT

Gastric cancer (GC), a leading cause of cancer-related mortality, remains a significant challenge despite recent therapeutic advancements. In this study, we explore the potential of targeting cell surface glucose-regulated protein 94 (GRP94) with antibodies as a novel therapeutic approach for GC. Our comprehensive analysis of GRP94 expression across various cancer types, with a specific focus on GC, revealed a substantial overexpression of GRP94, highlighting its potential as a promising target. Through in vitro and in vivo efficacy assessments, as well as toxicological analyses, we found that K101.1, a fully human monoclonal antibody designed to specifically target cell surface GRP94, effectively inhibits GC growth and angiogenesis without causing in vivo toxicity. Furthermore, our findings indicate that K101.1 promotes the internalization and concurrent downregulation of cell surface GRP94 on GC cells. In conclusion, our study suggests that cell surface GRP94 may be a potential therapeutic target in GC, and that antibody-based targeting of cell surface GRP94 may be an effective strategy for inhibiting GRP94-mediated GC growth and angiogenesis. [BMB Reports 2024; 57(4): 188-193].


Subject(s)
Antibodies, Monoclonal , Stomach Neoplasms , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , Humans , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Animals , Mice , Cell Proliferation/drug effects , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/immunology , Neovascularization, Pathologic/metabolism , Mice, Nude
12.
Aging (Albany NY) ; 16(7): 6035-6053, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38536020

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a leading reason for the death of cancer around the world. The immune microenvironment counts a great deal in immunotherapy of advanced tumors, in which T cells exert an indispensable function. METHODS: Single-cell RNA sequencing data were utilized to characterize the expression profile of T cells, followed by T cell-related genes (TCRGs) to construct signature and measure differences in survival time, enrichment pathways, somatic mutation status, immune status, and immunotherapy between groups. RESULTS: The complex tumor microenvironment was analyzed by scRNA-seq data of GC patients. We screened for these T cell signature expression genes and the TCRGs-based signature was successfully constructed and relied on the riskscore grouping. In gene set enrichment analysis, it was shown that pro-tumor and suppressive immune pathways were more abundant in the higher risk group. We also found different infiltration of immune cells in two groups, and that the higher risk samples had a poorer response to immunotherapy. CONCLUSION: Our study established a prognostic model, in which different groups had different prognosis, immune status, and enriched features. These results have provided additional insights into prognostic evaluation and the development of highly potent immunotherapies in GC.


Subject(s)
Adenocarcinoma , Single-Cell Analysis , Stomach Neoplasms , T-Lymphocytes , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Humans , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , T-Lymphocytes/immunology , Prognosis , Male , Female , Gene Expression Regulation, Neoplastic , Immunotherapy , Middle Aged , Transcriptome , Biomarkers, Tumor/genetics , Gene Expression Profiling , Aged
13.
Hematol Oncol Clin North Am ; 38(3): 599-616, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38493074

ABSTRACT

Immune checkpoint inhibitors are rapidly transforming the care of patients with esophagogastric cancer. Particularly, anti-PD-1 therapy has demonstrated promising efficacy in metastatic and resectable disease. In this review, the authors discuss landmark clinical trials, highlight challenges and opportunities in this field, and propose potential directions for future work.


Subject(s)
Esophageal Neoplasms , Immune Checkpoint Inhibitors , Immunotherapy , Stomach Neoplasms , Humans , Esophageal Neoplasms/therapy , Esophageal Neoplasms/immunology , Stomach Neoplasms/therapy , Stomach Neoplasms/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Clinical Trials as Topic
14.
Aging (Albany NY) ; 16(7): 6163-6187, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38552217

ABSTRACT

Kinesin Family Member 3C (KIF3C) assumes a crucial role in various biological processes of specific human cancers. Nevertheless, there exists a paucity of systematic assessments pertaining to the contribution of KIF3C in human malignancies. We conducted an extensive analysis of KIF3C, covering its expression profile, prognostic relevance, molecular function, tumor immunity, and drug sensitivity. Functional enrichment analysis was also carried out. In addition, we conducted in vitro experiments to substantiate the role of KIF3C in gastric cancer (GC). KIF3C expression demonstrated consistent elevation in various tumors compared to their corresponding normal tissues. We further unveiled that heightened KIF3C expression served as a prognostic indicator, and its elevated levels correlated with unfavorable clinical outcomes, encompassing reduced OS, DSS, and PFS in several cancer types. Notably, KIF3C expression exhibited positive associations with the pathological stages of several cancers. Moreover, KIF3C demonstrated varying relationships with the infiltration of various distinct immune cell types in gastric cancer. Functional analysis outcomes indicated that KIF3C played a role in the PI3K-AKT signaling pathway. Drug sensitivity unveiled a positive relationship between KIF3C in gastric cancer and the IC50 values of the majority of identified anti-cancer drugs. Additionally, KIF3C knockdown reduced the proliferation, migration, and invasion capabilities, increased apoptosis, and led to alterations in the cell cycle of gastric cancer cells. Our research has revealed the significant and functional role of KIF3C as a tumorigenic gene in diverse cancer types. These findings indicate that KIF3C may serve as a promising target for the treatment of gastric cancer.


Subject(s)
Biomarkers, Tumor , Kinesins , Stomach Neoplasms , Humans , Kinesins/genetics , Kinesins/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Prognosis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Cell Movement/genetics , Signal Transduction
15.
FEBS Open Bio ; 14(5): 855-866, 2024 May.
Article in English | MEDLINE | ID: mdl-38494433

ABSTRACT

Gastric cancer has a high rate of recurrence, and as such, immunotherapy strategies are being investigated as a potential therapeutic strategy. Although the involvement of immune checkpoints in immunotherapy is well studied, biomechanical cues, such as target cell stiffness, have not yet been subject to the same level of investigation. Changes in the cholesterol content of the cell membrane directly influence tumor cell stiffness. Here, we investigated the effect of cholesterol on NK cell-mediated killing of gastric cancer stem-like cells. We report that surviving tumor cells with stem-like properties elevated cholesterol metabolism to evade NK cell cytotoxicity. Inhibition of cholesterol metabolism enhances NK cell-mediated killing of gastric cancer stem-like cells, highlighting a potential avenue for improving immunotherapy efficacy. This study suggests a possible effect of cancer cell stiffness on immune evasion and offers insights into enhancing immunotherapeutic strategies against tumors.


Subject(s)
Cholesterol , Killer Cells, Natural , Neoplastic Stem Cells , Stomach Neoplasms , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Cholesterol/metabolism , Cell Line, Tumor , Cytotoxicity, Immunologic , Immunotherapy/methods , Tumor Escape/immunology
16.
Aging (Albany NY) ; 16(7): 6098-6117, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38546403

ABSTRACT

PURPOSE: Necroptosis plays an important role in the tumorigenesis, development, metastasis, and drug resistance of malignant tumors. This study explored the new model for assessing stomach adenocarcinoma (STAD) prognosis and immunotherapy by combining long noncoding RNAs associated with necroptosis. METHODS: Patient clinical data and STAD gene expression profiles were curated from The Cancer Genome Atlas (TCGA). Immune-related genes were sourced from a specialized molecular database. Perl software and R software were used for data processing and analysis. Necroptosis-related lncRNAs in STAD were pinpointed via R's correlation algorithms. These lncRNAs, in conjunction with clinical data, informed the construction of a prognostic lncRNA-associated risk score model using univariate and multivariate Cox regression analyses. The model's prognostic capacity was evaluated by Kaplan-Meier survival curves and validated as an independent prognostic variable. Further, a nomogram incorporating this model with clinical parameters was developed, offering refined individual survival predictions. Subsequent analyses of immune infiltration and chemosensitivity within necroptosis-related lncRNA clusters utilized an arsenal of bioinformatic tools, culminating in RT-PCR validation of lncRNA expression. RESULTS: Through rigorous Cox regression, 21 lncRNAs were implicated in the risk score model. Stratification by median risk scores delineated patients into high- and low-risk cohorts, with the latter demonstrating superior prognostic outcomes. The risk model was corroborated as an independent prognostic indicator for STAD. The integrative nomogram displayed high concordance between predicted and observed survival rates, as evidenced by calibration curves. Differential immune infiltration in risk-defined groups was illuminated by the single sample GSEA (ssGSEA), indicating pronounced immune presence in higher-risk patients. Tumor microenvironment (TME) analysis showed that cluster-C3 had the highest score in the analysis of the three TMEs. Through the differential analysis of immune checkpoints, it was found that almost all immune checkpoint-related genes were expressed differently in various tumor clusters. Among them, CD44 expression was the highest. By comparing all drug sensitivities, we screened out 29 drugs with differences in drug sensitivity across different clusters. Risk score gene expression identification results showed that these lncRNAs were abnormally expressed in gastric cancer cell lines. CONCLUSIONS: This investigation provides a robust methodological advance in prognosticating and personalizing immunotherapy for STAD, leveraging quantitatively derived tumor cluster risk scores. It posits the use of necroptosis-related lncRNAs as pivotal molecular beacons for guiding therapeutic strategies and enhancing clinical outcomes in STAD.


Subject(s)
Adenocarcinoma , Necroptosis , RNA, Long Noncoding , Stomach Neoplasms , Tumor Microenvironment , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Adenocarcinoma/drug therapy , Prognosis , Necroptosis/genetics , Gene Expression Regulation, Neoplastic , Male , Female , Drug Resistance, Neoplasm/genetics , Nomograms , Biomarkers, Tumor/genetics , Middle Aged , Kaplan-Meier Estimate
17.
Rev. esp. salud pública ; 98: e202403017, Mar. 2024. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-231922

ABSTRACT

Fundamentos: existen pruebas de la factibilidad de una vacuna para prevenir la infección por helicobacter pylori. Modelizacio-nes en entornos de baja prevalencia informan de una muy probable coste-efectividad a largo plazo. El objetivo de este estudio fue cuantificar su eficiencia en un contexto local.métodos: se simuló la evolución de una cohorte de nacidos a través de un modelo compartimental representativo de varios estados clínicos en relación a la infección por h. Pylori. Se ejecutó dicho modelo bajo las premisas de vacunación en el periodo de lactante y de no intervención. El horizonte temporal fue equivalente a la esperanza de vida y se tuvo en cuenta la perspectiva del sistema de salud.resultados: la vacunación frente a h. Pylori costaría de media 2.168 €/persona más que la no intervención. Con ello se obten-drían 0,32 años de vida ganados ajustados por calidad (avac), lo que implicaría una razón de coste-efectividad incremental (rcei) media de 7.196 €/avac. Para una disposición a pagar de 24.506 €/avac, el 99,96% de las simulaciones resultaron coste-efectivas al alcanzar el horizonte temporal y se cruzó dicho umbral a partir de los treinta años de la vacunación. Las variables que más peso tuvieron para explicar la variabilidad de la rcei fueron, en este orden, la efectividad vacunal, la incidencia de la infección en la primera infancia y el precio de la vacuna. La vacunación dejaría de ser coste-efectiva con un precio mayor de 3.634€/vial o con una cobertura poblacional efectiva menor del 11%.conclusiones: una vacunación frente a la infección por h. Pylori administrada en la infancia sería coste-efectiva a largo plazo en un entorno con las características epidemiológicas y económicas del sur de europa.(AU)


Background: there is sufficient evidence on the feasibility of a vaccine to prevent helicobacter pylori infection. Modeling studies in low prevalence environments report a very probable long-term cost-effectiveness. The objective of this study was to quantify its efficiency in a local context.methods: the evolution of a cohort of newborns was simulated through a compartmental model representing a series of clinical situations regarding h. Pylori infection and related diseases. The model was run under the assumption of both vaccination in the first year of life and no intervention. The time horizon was set as equivalent to the life expectancy and the perspective of the health system was taken into account.results: vaccination against h. Pylori would cost an average of €2,168/person more than no intervention. This would yield an average additional 0.32 quality-adjusted life years gained (qaly), which would entail an incremental cost-effectiveness ratio (icer) of €7,196/qaly. For a willingness to pay of €24,506/qaly, 99.96% of the simulations were cost-effective at eighty-four years old. This threshold was crossed thirty years after vaccination. The variables that carried the most weight in explaining the variability of the icer were, in this order, vaccine effectiveness, the incidence of infection in young children, and the price of the vaccine. Vaccination would cease to be cost-effective with a price greater than €3,634/dose or with effective population coverage less than 11%.(AU)


Subject(s)
Humans , Male , Female , Stomach Neoplasms/economics , Stomach Neoplasms/immunology , Vaccines , Helicobacter , Vaccination
18.
Apoptosis ; 29(5-6): 799-815, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38347337

ABSTRACT

PANoptosis is a form of inflammatory programmed cell death that is regulated by the PANoptosome. This PANoptosis possesses key characteristics of pyroptosis, apoptosis, and necroptosis, yet cannot be fully explained by any of these cell death modes. The unique nature of this cell death mechanism has garnered significant interest. However, the specific role of PANoptosis-associated features in gastric cancer (GC) is still uncertain. Patients were categorized into different PAN subtypes based on the expression of genes related to the PANoptosome. We conducted a systematic analysis to investigate the variations in prognosis and tumor microenvironment (TME) among these subtypes. Furthermore, we developed a risk score, called PANoptosis-related risk score (PANS), which is constructed from genes associated with the PANoptosis. We comprehensively analyzed the correlation between PANS and GC prognosis, TME, immunotherapy efficacy and chemotherapeutic drug sensitivity. Additionally, we performed in vitro experiments to validate the impact of Keratin 7 (KRT7) on GC. We identified two PAN subtypes (PANcluster A and B). PANoptosome genes were highly expressed in PANcluster A. PANcluster A has the characteristics of favorable prognosis, abundant infiltration of anti-tumor lymphocytes, and sensitivity to immunotherapy, thus it was categorized as an immune-inflammatory type. Meanwhile, our constructed PANS can effectively predict the prognosis and immune efficacy of GC. Patients with low PANS have a good prognosis, and have the characteristics of high tumor mutation load (TMB), high microsatellite instability (MSI), low tumor purity and sensitivity to immunotherapy. In addition, PANS can also identify suitable populations for different chemotherapy drugs. Finally, we confirmed that KRT7 is highly expressed in GC. Knocking down the expression of KRT7 significantly weakens the proliferation and migration abilities of GC cells. The models based on PANoptosis signature help to identify the TME features of GC and can effectively predict the prognosis and immune efficacy of GC. Furthermore, the experimental verification results of KRT7 provide theoretical support for anti-tumor treatment.


Subject(s)
Immunotherapy , Stomach Neoplasms , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/diagnosis , Humans , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Keratin-7/genetics , Keratin-7/metabolism , Apoptosis/genetics
19.
Cell Prolif ; 57(5): e13591, 2024 May.
Article in English | MEDLINE | ID: mdl-38319150

ABSTRACT

Highly aggressive gastric cancer (HAGC) is a gastric cancer characterized by bone marrow metastasis and disseminated intravascular coagulation (DIC). Information about the disease is limited. Here we employed single-cell RNA sequencing to investigate peripheral blood mononuclear cells (PBMCs), aiming to unravel the immune response of patients toward HAGC. PBMCs from seven HAGC patients, six normal advanced gastric cancer (NAGC) patients, and five healthy individuals were analysed by single-cell RNA sequencing. The expression of genes of interest was validated by bulk RNA-sequencing and ELISA. We found a massive expansion of neutrophils in PBMCs of HAGC. These neutrophils are activated, but immature. Besides, mononuclear phagocytes exhibited an M2-like signature and T cells were suppressed and reduced in number. Analysis of cell-cell crosstalk revealed that several signalling pathways involved in neutrophil to T-cell suppression including APP-CD74, MIF-(CD74+CXCR2), and MIF-(CD74+CD44) pathways were increased in HAGC. NETosis-associated genes S100A8 and S100A9 as well as VEGF, PDGF, FGF, and NOTCH signalling that contribute to DIC development were upregulated in HAGC too. This study reveals significant changes in the distribution and interactions of the PBMC subsets and provides valuable insight into the immune response in patients with HAGC. S100A8 and S100A9 are highly expressed in HAGC neutrophils, suggesting their potential to be used as novel diagnostic and therapeutic targets for HAGC.


Subject(s)
Leukocytes, Mononuclear , Sequence Analysis, RNA , Single-Cell Analysis , Stomach Neoplasms , Humans , Stomach Neoplasms/immunology , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Stomach Neoplasms/blood , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , Neutrophils/metabolism , Neutrophils/immunology , Male , Female , Middle Aged , Signal Transduction , Aged , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Med Clin (Barc) ; 162(9): 425-427, 2024 May 17.
Article in English, Spanish | MEDLINE | ID: mdl-38418307

ABSTRACT

INTRODUCTION: Gastric cancer is one of the cancers most associated with thromboembolic phenomena. The objective of this article is to study if there is a correlation between thromboembolic phenomena in gastric cancer and tumor expression of PDL-1. METHODS: To this end, the association between thromboembolic events and PDL-1 expression was retrospectively studied in a sample of 46 patients from our hospital. RESULTS: The results obtained revealed a statistically significant difference between the percentage of thromboembolic events between positive and negative PDL-1 with an increase in the latter with a P value of 0.034. CONCLUSION: In conclusion, the expression of PDL-1, and with it, of an inhibitory factor of the cellular immune response, correlates with a decrease in thromboembolic events in patients with gastric cancer, which could indicate the crucial role of the immune response in which thromboembolic events occur.


Subject(s)
B7-H1 Antigen , Stomach Neoplasms , Venous Thrombosis , Humans , Stomach Neoplasms/immunology , Male , Female , Retrospective Studies , Middle Aged , Aged , Venous Thrombosis/etiology , Venous Thrombosis/epidemiology , Incidence , Aged, 80 and over , Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...