Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 372
Filter
1.
Sci Rep ; 14(1): 13415, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862557

ABSTRACT

Sperm-associated antigen 5 (SPAG5) regulates cancer cell invasion and is involved in the progression of many cancers. However, the role of SPAG5 in endometrial carcinoma (EC) is still unknown. The purpose of this study was to explore the role of SPAG5 in EC and its potential molecular mechanism. The UALCAN tool and cBioPortal were used to analyze the expression and alterations of SPAG5 in EC, respectively. OncoLnc was used for survival analysis. We analyzed the effects of SPAG5 on immune cell infiltration and the expression levels of immune checkpoints. We also overexpressed and knocked down SPAG5 in EC cells to explore the effect of SPAG5 regulation on migration, invasion, apoptosis, and the cell cycle of EC cells. We found that SPAG5 was overexpressed and the SPAG5 gene was often mutated in EC. High SPAG5 expression was significantly associated with poor overall survival in patients with EC. SPAG5 also affected the level of immune cell infiltration in the TIME and the expression of immune checkpoints lymphocyte activating 3 (LAG3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT) in patients with EC. It may also be involved in the immunotherapy response in these patients. In vitro experiments showed that SPAG5 promotes cancer cell migration and invasion. In conclusion, this study lays the foundation for further understanding the molecular mechanisms of EC involving SPAG5 and contributes to diagnosing and managing this disease.


Subject(s)
Cell Movement , Endometrial Neoplasms , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness , Up-Regulation , Humans , Female , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Cell Movement/genetics , Prognosis , Cell Line, Tumor , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Apoptosis/genetics
2.
Int Immunopharmacol ; 134: 112196, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38759367

ABSTRACT

Cell cycle dysregulation leading to uncontrolled growth is a primary characteristic of malignancy. GSG2, a mitosis-related kinase, affects the normal cell cycle by interfering with the normal dissociation of centromere cohesion, and its overexpression has been shown to play an important role in cancer cells. Here, we investigated the function of GSG2 as a tumor promoter in endometrial carcinoma and its relationship with the immunological microenvironment. We used immunohistochemistry to identify a correlation between the development and prognosis of GSG2 and endometrial cancer. Cell and animal experiments confirmed that GSG2 has a protumorigenic phenotype in endometrial cancer cell lines. Furthermore, using GeneChip analysis and a tumor-immune coculture model, we observed a link between GSG2 expression and the composition of the immune microenvironment. Therefore, we concluded that the activation of the PI3K/AKT pathway by GSG2 may impact DNA repair, disrupt the cell cycle, and regulate the immune response, all of which could increase the ability of EC cells to proliferate malignantly. Consequently, it is anticipated that GSG2 will be a viable therapeutic target in endometrial carcinoma.


Subject(s)
B7-H1 Antigen , Endometrial Neoplasms , Phosphatidylinositol 3-Kinases , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Female , Humans , Mice , Aurora Kinase B , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Cell Line, Tumor , Disease Progression , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase Kinases , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tumor Microenvironment/immunology
3.
Bull Exp Biol Med ; 176(5): 607-611, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38730105

ABSTRACT

The study presents the killer functions of circulating neutrophils: myeloperoxidase activity, the ability to generate ROS, phagocytic activity, receptor status, NETosis, as well as the level of cytokines IL-2, IL-4, IL-6, IL-17A, and IL-18, granulocyte CSF, monocyte chemotactic protein 1, and neutrophil elastase in the serum of patients with uterine myoma and endometrial cancer (FIGO stages I-III). The phagocytic ability of neutrophils in uterine myoma was influenced by serum levels of granulocyte CSF and IL-2 in 54% of the total variance. The degranulation ability of neutrophils in endometrial cancer was determined by circulating IL-18 in 50% of the total variance. In uterine myoma, 66% of the total variance in neutrophil myeloperoxidase activity was explained by a model dependent on blood levels of IL-17A, IL-6, and IL-4. The risk of endometrial cancer increases when elevated levels of monocyte chemotactic protein 1 in circulating neutrophils are associated with reduced ability to capture particles via extracellular traps (96% probability).


Subject(s)
Chemokine CCL2 , Endometrial Neoplasms , Interleukin-17 , Interleukin-6 , Neutrophils , Humans , Female , Neutrophils/metabolism , Neutrophils/immunology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/blood , Endometrial Neoplasms/pathology , Endometrial Neoplasms/metabolism , Interleukin-6/blood , Chemokine CCL2/blood , Interleukin-17/blood , Middle Aged , Interleukin-4/blood , Peroxidase/blood , Peroxidase/metabolism , Interleukin-18/blood , Uterine Neoplasms/blood , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Colony-Stimulating Factor/metabolism , Phagocytosis , Leiomyoma/blood , Leiomyoma/immunology , Leiomyoma/pathology , Leiomyoma/metabolism , Cytokines/blood , Cytokines/metabolism , Leukocyte Elastase/blood , Leukocyte Elastase/metabolism , Adult , Extracellular Traps/metabolism , Extracellular Traps/immunology , Reactive Oxygen Species/metabolism , Aged , Interleukin-2
4.
Medicine (Baltimore) ; 103(19): e38129, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728458

ABSTRACT

BACKGROUND: The prognostic significance of tumor-infiltrating immune cells in endometrial cancer is a subject of ongoing debate. Recent evidence increasingly suggests that these immune cells and cytokines, abundant in endometrial cancer tissues, play a pivotal role in stimulating the body inherent anti-tumor immune responses. METHODS: Leveraging publicly accessible genetic data, we conducted an exhaustive 2-sample Mendelian randomization (MR) study. This study aimed to explore the causal links between 731 immunophenotypes and the risk of endometrial cancer. We thoroughly assessed the robustness, heterogeneity, and potential horizontal pleiotropy of our findings through extensive sensitivity analyses. RESULTS: Our study identified 36 immunophenotypes associated with endometrial cancer risk. Specific immunophenotypes, such as the percentage of Naive-mature B-cells in lymphocytes (OR = 0.917, 95% CI = 0.863-0.974, P = .005), and HLA DR expression on CD14-CD16 + monocytes (OR = 0.952, 95% CI = 0.911-0.996, P = .032), exhibited a negative correlation with endometrial cancer. Conversely, CD127 expression on CD45RA + CD4 + in Treg cells (OR = 1.042, 95% CI = 1.000-1.085, P = .049), and CM CD4+%T in T cell maturation stages (OR = 1.074, 95% CI = 1.012-1.140, P = .018) showed a positive correlation. Reverse MR analysis linked endometrial cancer to 4 immunophenotypes, including a positive correlation with CD127-CD8br %T cell of Treg (OR = 1.172, 95% CI = 1.080-1.270, P = .0001), and negative correlations with 3 others, including CM CD4+%T cell (OR = 0.905, 95% CI = 0.832-0.984, P = .019). CONCLUSION SUBSECTIONS: Our findings underscore a significant causal relationship between immunophenotypes and endometrial cancer in bidirectional MR analyses. Notably, the CM CD4+%T immunophenotype emerged as potentially crucial in endometrial cancer development.


Subject(s)
Endometrial Neoplasms , Mendelian Randomization Analysis , Female , Humans , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology
5.
Ann Oncol ; 35(5): 414-428, 2024 May.
Article in English | MEDLINE | ID: mdl-38431043

ABSTRACT

BACKGROUND: Immunotherapy has transformed the endometrial cancer treatment landscape, particularly for those exhibiting mismatch repair deficiency [MMRd/microsatellite instability-hypermutated (MSI-H)]. A growing body of evidence supports the integration of immunotherapy with chemotherapy as a first-line treatment strategy. Recently, findings from ongoing trials such as RUBY (NCT03981796), NRG-GY018 (NCT03914612), AtTEnd (NCT03603184), and DUO-E (NCT04269200) have been disclosed. MATERIALS AND METHODS: This paper constitutes a review and meta-analysis of phase III trials investigating the role of immunotherapy in the first-line setting for advanced or recurrent endometrial cancer. RESULTS: The pooled data from 2320 patients across these trials substantiate the adoption of chemotherapy alongside immunotherapy, revealing a significant improvement in progression-free survival compared to chemotherapy alone [hazard ratio (HR) 0.70, 95% confidence interval (CI) 0.62-0.79] across all patient groups. Progression-free survival benefits are more pronounced in MMRd/MSI-H tumors (n = 563; HR 0.33, 95% CI 0.23-0.43). This benefit, albeit less robust, persists in the MMR-proficient/microsatellite stable group (n = 1757; HR 0.74, 95% CI 0.60-0.91). Pooled data further indicate that chemotherapy plus immunotherapy enhances overall survival compared to chemotherapy alone in all patients (HR 0.75, 95% CI 0.63-0.89). However, overall survival data maturity remains low. CONCLUSIONS: The incorporation of immunotherapy into the initial treatment for advanced and metastatic endometrial cancer brings about a substantial improvement in oncologic outcomes, especially within the MMRd/MSI-H subset. This specific subgroup is currently a focal point of investigation for evaluating the potential of chemotherapy-free regimens. Ongoing exploratory analyses aim to identify non-responding patients eligible for inclusion in clinical trials.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Endometrial Neoplasms , Immunotherapy , Humans , Female , Endometrial Neoplasms/pathology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/therapy , Endometrial Neoplasms/immunology , Immunotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Clinical Trials, Phase III as Topic , Progression-Free Survival , Randomized Controlled Trials as Topic , Microsatellite Instability , Neoplasm Metastasis
6.
Clin Cancer Res ; 30(9): 1906-1915, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38350001

ABSTRACT

PURPOSE: The response to immune checkpoint inhibitors (ICI) in deficient mismatch repair (dMMR) colorectal cancer and endometrial cancer is variable. Here, we explored the differential response to ICIs according to different mismatch repair alterations. EXPERIMENTAL DESIGN: Colorectal cancer (N = 13,701) and endometrial cancer (N = 3,315) specimens were tested at Caris Life Sciences. Median overall survival (mOS) was estimated using Kaplan-Meier. The prediction of high-, intermediate-, and low-affinity epitopes by tumor mutation burden (TMB) values was conducted using R-squared (R2). RESULTS: Compared with mutL (MLH1 and PMS2) co-loss, the mOS was longer in mutS (MSH2 and MSH6) co-loss in all colorectal cancer (54.6 vs. 36 months; P = 0.0.025) and endometrial cancer (81.5 vs. 48.2 months; P < 0.001) patients. In ICI-treated patients, the mOS was longer in mutS co-loss in colorectal cancer [not reached (NR) vs. 36 months; P = 0.011). In endometrial cancer, the mOS was NR vs. 42.2 months; P = 0.711]. The neoantigen load (NAL) in mutS co-loss compared with mutL co-loss was higher in colorectal cancer (high-affinity epitopes: 25.5 vs. 19; q = 0.017, intermediate: 39 vs. 32; q = 0.004, low: 87.5 vs. 73; q < 0.001) and endometrial cancer (high-affinity epitopes: 15 vs. 11; q = 0.002, intermediate: 27.5 vs. 19; q < 0.001, low: 59 vs. 41; q < 0.001), respectively. R2 ranged from 0.25 in mutS co-loss colorectal cancer to 0.95 in mutL co-loss endometrial cancer. CONCLUSIONS: Patients with mutS co-loss experienced longer mOS in colorectal cancer and endometrial cancer and better response to ICIs in colorectal cancer. Among all explored biomarkers, NAL was higher in mutS co-loss and may be a potential driving factor for the observed better outcomes. TMB did not reliably predict NAL.


Subject(s)
Colorectal Neoplasms , DNA Mismatch Repair , Endometrial Neoplasms , Immune Checkpoint Inhibitors , Mutation , Humans , Female , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Aged , Male , Middle Aged , Endometrial Neoplasms/genetics , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Biomarkers, Tumor/genetics , Adult , Aged, 80 and over , Prognosis , DNA-Binding Proteins/genetics
7.
Nature ; 620(7973): 409-416, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532934

ABSTRACT

Netrin-1 is upregulated in cancers as a protumoural mechanism1. Here we describe netrin-1 upregulation in a majority of human endometrial carcinomas (ECs) and demonstrate that netrin-1 blockade, using an anti-netrin-1 antibody (NP137), is effective in reduction of tumour progression in an EC mouse model. We next examined the efficacy of NP137, as a first-in-class single agent, in a Phase I trial comprising 14 patients with advanced EC. As best response we observed 8 stable disease (8 out of 14, 57.1%) and 1 objective response as RECIST v.1.1 (partial response, 1 out of 14 (7.1%), 51.16% reduction in target lesions at 6 weeks and up to 54.65% reduction during the following 6 months). To evaluate the NP137 mechanism of action, mouse tumour gene profiling was performed, and we observed, in addition to cell death induction, that NP137 inhibited epithelial-to-mesenchymal transition (EMT). By performing bulk RNA sequencing (RNA-seq), spatial transcriptomics and single-cell RNA-seq on paired pre- and on-treatment biopsies from patients with EC from the NP137 trial, we noted a net reduction in tumour EMT. This was associated with changes in immune infiltrate and increased interactions between cancer cells and the tumour microenvironment. Given the importance of EMT in resistance to current standards of care2, we show in the EC mouse model that a combination of NP137 with carboplatin-paclitaxel outperformed carboplatin-paclitaxel alone. Our results identify netrin-1 blockade as a clinical strategy triggering both tumour debulking and EMT inhibition, thus potentially alleviating resistance to standard treatments.


Subject(s)
Endometrial Neoplasms , Epithelial-Mesenchymal Transition , Netrin-1 , Animals , Female , Humans , Mice , Biopsy , Carboplatin/administration & dosage , Carboplatin/pharmacology , Carboplatin/therapeutic use , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Profiling , Netrin-1/antagonists & inhibitors , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA-Seq , Single-Cell Gene Expression Analysis , Tumor Microenvironment/drug effects
8.
Front Immunol ; 14: 1165602, 2023.
Article in English | MEDLINE | ID: mdl-37077909

ABSTRACT

Introduction: The interaction between endometrial cancer (EMC) cells and intratumoral macrophages plays a significant role in the development of the disease. PYD domains-containing protein 3 (NLRP3) inflammasome formation triggers caspase-1/IL-1ß signaling pathways and produces reactive oxygen species (ROS) in macrophages. However, the role of NLRP3-regulated ROS production in macrophage polarization and the subsequent growth and metastasis of EMC remains unknown. Methods: We conducted bioinformatic analysis to compare NLRP3 levels in intratumoral macrophages from EMC and normal endometrium. In vitro experiments involved knocking out NLRP3 in macrophages to shift the polarization from an anti-inflammatory M1-like phenotype to a proinflammatory M2-like phenotype and reduce ROS production. The impact of NLRP3 depletion on the growth, invasion, and metastasis of co-cultured EMC cells was assessed. We also evaluated the effect of NLRP3 depletion in macrophages on the growth and metastasis of implanted EMC cells in mice. Results: Our bioinformatic analysis showed significantly lower NLRP3 levels in intratumoral macrophages from EMC than those from normal endometrium. Knocking out NLRP3 in macrophages shifted their polarization to a proinflammatory M2-like phenotype and significantly reduced ROS production. NLRP3 depletion in M2-polarized macrophages increased the growth, invasion, and metastasis of co-cultured EMC cells. NLRP3 depletion in M1-polarized macrophages reduced phagocytic potential, which resulted in weakened immune defense against EMC. Additionally, NLRP3 depletion in macrophages significantly increased the growth and metastasis of implanted EMC cells in mice, likely due to compromised phagocytosis by macrophages and a reduction in cytotoxic CD8+ T cells. Discussion: Our results suggest that NLRP3 plays a significant role in regulating macrophage polarization, oxidative stress, and immune response against EMC. NLRP3 depletion alters the polarization of intratumoral macrophages, leading to weakened immune defense against EMC cells. The reduction in ROS production by the loss of NLRP3 may have implications for the development of novel treatment strategies for EMC.


Subject(s)
Endometrial Neoplasms , NLR Family, Pyrin Domain-Containing 3 Protein , Tumor-Associated Macrophages , Endometrial Neoplasms/immunology , Neoplasm Transplantation , Heterografts , Humans , Animals , Mice , Oxidative Stress , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phagocytosis , Neoplasm Metastasis
9.
Anticancer Res ; 42(3): 1367-1376, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220229

ABSTRACT

BACKGROUND/AIM: Endometrial carcinoma (EC) is one of the most common gynecological cancers in the Western Hemisphere. Nevertheless, there are not enough appropriate treatment options, especially for advanced stages. The immune checkpoint blockade represents a promising alternative to established cancer therapies by suppressing the immune-inhibitory activity of the immune checkpoint factors programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1). In the present study, we characterized the clinical relevance of the biomarker PD-L1 expression in terms of its prognostic capabilities in EC. PATIENTS AND METHODS: Tumor tissue samples from 87 EC patients were retrospectively analyzed by immunohistochemistry (PD-L1, p16, estrogen receptor, progesterone receptor, HER2/neu, Ki-67, CD3, CD20, CD68). RESULTS: A total of 17.3% of EC patients were PD-L1 positive. PD-L1 status did not represent a suitable prognostic marker in EC, but correlated with T3/T4stage, positive lymph node status, p16 expression, and absence of estrogen and progesterone receptor. PD-L1 positive tissues showed increased infiltration with lymphocytes, monocytes, and macrophages, although not statistically significant in every case. CONCLUSION: In EC, PD-L1 expression has no prognostic significance, but correlates with other oncogenic factors and indicates increased infiltration of the tumor with immune cells. Thus, PD-1/PD-L1 immunecheckpoint blockade seems to be very promising, at least in a subset of EC patients.


Subject(s)
B7-H1 Antigen/analysis , Biomarkers, Tumor/analysis , Endometrial Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Monocytes/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/antagonists & inhibitors , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Female , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunohistochemistry , Immunotherapy , Middle Aged , Neoplasm Staging , Predictive Value of Tests , Retrospective Studies
10.
FEBS Lett ; 596(4): 449-464, 2022 02.
Article in English | MEDLINE | ID: mdl-34855209

ABSTRACT

The Hippo signaling pathway is critical for carcinogenesis. However, the roles of the Hippo signaling pathway in the tumor immune microenvironment have been rarely investigated. This study systematically analyzed the relationship between the Hippo signaling pathway and immune cell infiltration across 32 cancer types. Both bioinformatics analyses and biological experiments revealed that the downstream effector of Hippo signaling YAP1 might inhibit CD8+ T cell infiltration by upregulating the expression of the transcription factor CREB1 in uterine corpus endometrial carcinoma. In addition, esophageal carcinoma (ESCA) patients were classified into three subtypes based on the Hippo-immune gene panel. The subtypes of ESCA had distinct characteristics in immune cell infiltration, immune pathways, and prognosis. Thus, this study also reveals a new classification of the immune subtypes with prognostic characteristics in ESCA.


Subject(s)
Endometrial Neoplasms/genetics , Esophageal Neoplasms/genetics , Hippo Signaling Pathway/genetics , Neoplasms/genetics , Tumor Microenvironment/genetics , YAP-Signaling Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Cell Movement , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/immunology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasms/classification , Neoplasms/immunology , Neoplasms/pathology , Prognosis , Proteomics/methods , Receptors, Antigen, B-Cell/classification , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/classification , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Survival Analysis , Terminology as Topic , YAP-Signaling Proteins/immunology
11.
Exp Biol Med (Maywood) ; 247(3): 221-236, 2022 02.
Article in English | MEDLINE | ID: mdl-34704492

ABSTRACT

Uterine corpus endometrial carcinoma (UCEC) is the third most frequent gynecological malignancies in the female reproductive system. Long non-coding RNAs (lncRNAs) are closely involved in tumor progression. This study aimed to develop an immune subtyping system and a prognostic model based on lncRNAs for UCEC. Paired lncRNAs and non-negative matrix factorization were applied to identify immune subtypes. Enrichment analysis was conducted to assess functional pathways, immune-related genes, and cells. Univariate and multivariate Cox regression analysis were performed to analyze the relation between lncRNAs and overall survival (OS). A prognostic model was constructed and optimized by least absolute shrinkage and selection operator (LASSO) and Akaike information criterion (AIC). Two immune subtypes (C1 and C2) and four paired-prognostic lncRNAs closely associated with overall survival were identified. Some immune features, sensitivity of chemotherapy and immunotherapy, and the relation with immune escape showed variations between two subtypes. A nomogram established based on prognostic model and clinical features was effective in OS prediction. The immune subtyping system based on lncRNAs and the four-paired-lncRNA signature was predictive of UCEC prognosis and can facilitate personalized therapies such as immunotherapy or RNA-based therapy for UCEC patients.


Subject(s)
Biomarkers, Tumor/immunology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Immunotherapy , RNA, Long Noncoding/immunology , Biomarkers, Tumor/genetics , Endometrial Neoplasms/mortality , Endometrial Neoplasms/therapy , Female , Gene Expression Regulation, Neoplastic , Humans , Mutation , Nomograms , Prognosis , RNA, Long Noncoding/genetics , Treatment Outcome , Tumor Escape/genetics , Tumor Escape/immunology
12.
Front Immunol ; 12: 788431, 2021.
Article in English | MEDLINE | ID: mdl-34970268

ABSTRACT

Background: Uterine corpus endometrial carcinoma (UCEC) is a gynecological malignant tumor with low survival rate and poor prognosis. The traditional clinicopathological staging is insufficient to estimate the prognosis of UCEC. It is necessary to select a more effective prognostic signature of UCEC to predict the prognosis and immunotherapy effect of UCEC. Methods: CIBERSORT and weighted correlation network analysis (WGCNA) algorithms were combined to screen modules related to regulatory T (Treg) cells. Subsequently, univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses were used to identify the genes in key modules. The difference in overall survival (OS) between high- and low-risk patients was analyzed by Kaplan-Meier analysis. The Tregs-related risk signature (TRRS) was screened by uni- and multivariate Cox analyses. Afterward, we analyzed the expression difference of TRRS and verified its ability to predict the prognosis of UCEC and the effect of immunotherapy. Results: Red module has the highest correlation with Tregs among all clustered modules. Pathways enrichment indicated that the related processes of UCEC were primarily associated to the immune system. Eight genes (ZSWIM1, NPRL3, GOLGA7, ST6GALNAC4, CDC16, ITPK1, PCSK4, and CORO1B) were selected to construct TRRS. We found that this TRRS is a significantly independent prognostic factor of UCEC. Low-risk patients have higher overall survival than high-risk patients. The immune status of different groups was different, and tumor-related pathways were enriched in patients with higher risk score. Low-risk patients are more likely take higher tumor mutation burden (TMB). Meanwhile, they are more sensitive to chemotherapy than patients with high-risk score, which indicated a superior prognosis. Immune checkpoints such as PD-1, CTLA4, PD-L1, and PD-L2 all had a higher expression level in low-risk group. TRRS expression really has a relevance with the sensitivity of UCEC patients to chemotherapeutic drugs. Conclusion: We developed and validated a TRRS to estimate the prognosis and reflect the immune status of UCEC, which could accurately assess the prognosis of patients with UCEC and supply personalized treatments for them.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma/genetics , Endometrial Neoplasms/genetics , Gene Expression Profiling , Lymphocytes, Tumor-Infiltrating/immunology , T-Lymphocytes, Regulatory/immunology , Transcriptome , Tumor Microenvironment/immunology , Carcinoma/drug therapy , Carcinoma/immunology , Carcinoma/mortality , Case-Control Studies , Clinical Decision-Making , Databases, Genetic , Decision Support Techniques , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/mortality , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genome-Wide Association Study , Humans , Immunotherapy , Nomograms , Predictive Value of Tests , Prognosis , Reproducibility of Results , Risk Assessment , Risk Factors
13.
BMC Cancer ; 21(1): 1203, 2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34763648

ABSTRACT

BACKGROUND: Infiltrating immune and stromal cells are important components of the endometrial cancer (EC) microenvironment, which has a significant effect on the biological behavior of EC, suggesting that unique immune-related genes may be associated with the prognosis of EC. However, the association of immune-related genes with the prognosis of EC has not been elucidated. We attempted to identify immune-related genes with potentially prognostic value in EC using The Cancer Genome Atlas database and the relationship between immune microenvironment and EC. METHODS: We analyzed 578 EC samples from TCGA database and used weighted gene co-expression network analysis to screen out immune-related genes. We constructed a protein-protein interaction network and analyzed it using STRING and Cytoscape. Immune-related genes were analyzed through conjoint Cox regression and random forest algorithm analysis were to identify a multi-gene prediction model and stratify low-risk and high-risk groups of EC patients. Based on these data, we constructed a nomogram prediction model to improve prognosis assessment. Evaluation of Immunological, gene mutations and gene enrichment analysis were applied on these groups to quantify additional differences. RESULTS: Using conjoint Cox regression and random forest algorithm, we found that TRBC2, TRAC, LPXN, and ARHGAP30 were associated with the prognosis of EC and constructed four gene risk models for overall survival and a consistent nomogram. The time-dependent receiver operating characteristic curve analysis revealed that the area under the curve for 1-, 3-, and 5-y overall survival was 0.687, 0.699, and 0.76, respectively. These results were validated using a validation cohort. Immune-related pathways were mostly enriched in the low-risk group, which had higher levels of immune infiltration and immune status. CONCLUSION: Our study provides new insights for novel biomarkers and immunotherapy targets in EC.


Subject(s)
Biomarkers, Tumor/genetics , Endometrial Neoplasms/mortality , Gene Expression Regulation, Neoplastic/immunology , Nomograms , Tumor Microenvironment/genetics , Datasets as Topic , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Middle Aged , Protein Interaction Maps/genetics , Protein Interaction Maps/immunology , ROC Curve , Tumor Microenvironment/immunology
14.
Front Immunol ; 12: 752646, 2021.
Article in English | MEDLINE | ID: mdl-34691070

ABSTRACT

Background: Endometriosis is a common gynecological disease characterized by the presence of endometrial tissue outside the uterus causing chronic inflammation, severe pain, and infertility. However, the innate immunity of gamma-delta (γδ) T lymphocytes in endometriosis has not been characterized. Women with endometriosis present numerous endocrine and immune dysfunctions and elevated risk for endometrial, ovarian, and breast cancers. The tyrosine kinase EphA2 is often overexpressed in cancer including endometrial carcinoma. Methods: We analyzed Vδ1 and Vδ2 γδ T cells in peripheral blood and paired peritoneal fluid samples in endometriosis patients (n = 19) and compared the counts with that of age- and sex-matched healthy donors (n = 33) using flow cytometry. Vδ1 and Vδ2 T cells isolated from healthy donors were used against KLE, RL-95, and Ishikawa endometrial tumor cells in 4 h flow cytometric cytotoxicity assays. The EphA2 blocking studies were performed using antibody, small-molecule inhibitor ALW-II-41-27, and the CRISPR/Cas9. Results: We determined Vδ1 T cells substantially reduced in patients' peripheral blood (p < 0.01) and peritoneal fluid (p < 0.001). No differences were found for circulating Vδ2 T cells compared with peritoneal fluid samples. We observed inherent cytotoxic reactivity of Vδ1 and Vδ2 γδ T lymphocytes against endometrial tumor cells. Importantly, we found reduced specific lysis of EphA2-positive cell lines KLE and RL-95 by Vδ1 T cells in the EphA2 antibody blocking studies and by the EphA2 inhibitor. Furthermore, Vδ1 T-cell-mediated killing was significantly decreased in RL-95 cell EPHA2 knockout. Finally, potent cytolytic activity exerted by Vδ1 T cells was significantly reduced in EPHA2 knockouts in renal A-498 and colon HT-29 carcinoma cell lines. Conclusions: We determined variable levels of Vδ1 and Vδ2 γδ T cells in endometriosis patients. We observed inherent cytotoxic reactivity of γδ T-cell subsets against endometrial cell lines. Specifically, we found that blocking of EphA2 expression resulted in significant inhibition of endometrial tumor killing mediated by Vδ1 γδ T cells. These results suggest that EphA2 is involved in tumor cell lysis and contributes to susceptibility to Vδ1 γδ T cells cytotoxic reactivity.


Subject(s)
Endometrial Neoplasms/immunology , Endometriosis/immunology , Intraepithelial Lymphocytes/immunology , Receptor, EphA2/metabolism , Adult , Cell Line, Tumor , Endometrial Neoplasms/metabolism , Endometriosis/metabolism , Female , Humans , Middle Aged
15.
Int J Mol Sci ; 22(17)2021 Aug 24.
Article in English | MEDLINE | ID: mdl-34502029

ABSTRACT

A tumor cell carrying characteristic genomic alteration(s) exists within its host's microenvironment. The tumor microenvironment (TME) renders holistic support to the tumor via cross-talk between tumor cells and three components of TME, immune components, vascular components, and fibroblast components. The tempero-spatial interaction of tumor cells with its microenvironment is the deterministic factor for tumor growth, progression, resistance to therapy, and its outcome in clinics. TME (1) facilitates proliferation, and the ensuing metastasis-associated phenotypes, (2) perturbs immune surveillance and supports tumor cells in their effort to evade immune recognition, and (3) actively participates in developing drug-induced resistance in cancer cells. Cancer-Associated Fibroblast (CAF) is a unique component of TME. CAF is the host mesenchyme immediately surrounding the tumor cells in solid tumors. It facilitates tumor growth and progression and participates in developing drug resistance in tumor cells by playing a critical role in all the ways mentioned above. The clinical outcome of a disease is thus critically contributed to by the CAF component of TME. Although CAFs have been identified historically, the functional relevance of CAF-tumor cell cross-talk and their influence on angiogenic and immune-components of TME are yet to be characterized in solid tumors, especially in endometrial cancers. Currently, the standard of care for the treatment of endometrial cancers is primarily guided by therapies directed towards the disease's tumor compartment and immune compartments. Unfortunately, in the current state of therapies, a complete response (CR) to the therapy is still limited despite a more commonly achieved partial response (PR) and stable disease (SD) in patients. Acknowledging the limitations of the current sets of therapies based on only the tumor and immune compartments of the disease, we sought to put forward this review based on the importance of the cross-talk between CAF of the tumor microenvironment and tumor cells. The premise of the review is to recognize the critical role of CAF in disease progression. This manuscript presents a systemic review of the role of CAF in endometrial cancers. We critically interrogated the active involvement of CAF in the tumor compartment of endometrial cancers. Here we present the functional characteristics of CAF in the context of endometrial cancers. We review (1) the characteristics of CAF, (2) their evolution from being anti-tumor to pro-tumor, (3) their involvement in regulating growth and several metastasis-associated phenotypes of tumor cells, (4) their participation in perturbing immune defense and evading immune surveillance, and (5) their role in mediating drug resistance via tumor-CAF cross-talk with particular reference to endometrial cancers. We interrogate the functional characteristics of CAF in the light of its dialogue with tumor cells and other components of TME towards developing a CAF-based strategy for precision therapy to supplement tumor-based therapy. The purpose of the review is to present a new vision and initiate a thought process which recognizes the importance of CAF in a tumor, thereby resulting in a novel approach to the design and management of the disease in endometrial cancers.


Subject(s)
Cancer-Associated Fibroblasts , Endometrial Neoplasms/physiopathology , Tumor Microenvironment , Endometrial Neoplasms/immunology , Female , Humans
16.
Immunotherapy ; 13(14): 1179-1192, 2021 10.
Article in English | MEDLINE | ID: mdl-34424031

ABSTRACT

Aims: We investigated immunogenomic signatures and correlated them with survival in ovarian cancer (OV) and endometrial cancer (EC). Materials & method: We used whole transcriptome sequencing data from uterine serous cancer and The Cancer Genome Atlas data of OV and EC (n = 719). Gene expression score was calculated. Population abundance of immune cells were estimated. Results: TGF-ß, myeloid cells, IFN-γ, T cells, B cells and endothelial cells predicted overall survival. Whereas CD47, neutrophils and endothelial cells predicted progression-free survival. In multivariate analyses, TGF-ß, CD47 and monocytic cells predicted survival in high levels of microsatellite instability (MSI-H) EC whereas high IFN-γ trended toward improved survival in the MSI-S EC. High IFN-γ/low TGF-ß and high IFN-γ/low CD47 signatures predicted longer overall survival. Low TGF-ß/low CD47 signature predicted longer overall survival only in the MSI-H EC. Conclusion: Our data support the role of immune markers in predicting survival in OV/EC.


Lay abstract We studied the association of immune markers and immune cells with survival outcome in ovarian and endometrial cancers. We performed gene expression analyses on tumor tissue of 719 patients. We generated signatures for the immune cells and markers and correlated them with survival outcome. We showed that specific immune cells and markers correlated strongly with survival even after we adjusted for other confounding factors. Further, we showed that these immune markers and immune cells have different correlation with survival when stratified in subtypes of endometrial cancer. Our data support the role of these immune markers in predicting survival outcome in ovarian and endometrial cancers.


Subject(s)
Biomarkers, Tumor/immunology , Endometrial Neoplasms/immunology , Ovarian Neoplasms/immunology , Transcriptome/immunology , Tumor Microenvironment/immunology , Adult , Aged , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Retrospective Studies , Exome Sequencing
17.
Gynecol Oncol ; 162(3): 631-637, 2021 09.
Article in English | MEDLINE | ID: mdl-34272092

ABSTRACT

OBJECTIVE: To investigate programmed cell death ligand 1 (PD-L1) expression patterns and define the associations among PD-L1, molecular subtypes, pathological features, and survival in a cohort of 833 patients with endometrial cancer, of whom approximately half had high-risk disease. METHODS: Using direct sequencing of the polymerase epsilon (POLE) exonuclease domain as well as immunohistochemistry for mismatch repair (MMR) proteins (MLH1, PMS2, MSH2, and MSH6) and p53, we stratified endometrial cancers into four molecular subtypes: POLE ultramutated, MMR-deficient, p53-mutant, and non-specific molecular profile (NSMP). PD-L1 was detected via immunohistochemistry and evaluated in tumor cells (TCs) and immune cells (ICs) individually and using the combined positive score (CPS). RESULTS: Positive PD-L1 staining in TCs (≥1%), ICs (≥1%), and in combination (CPS ≥1) was detected in 14.0%, 37.3%, and 45.1% of the samples, respectively. PD-L1 positivity in TCs was more frequent in high-grade than in low-grade tumors, while that in ICs was associated with lymphovascular space invasion, non-endometrioid histology, and deep myometrial invasion. PD-L1 expression in both TCs and ICs was more frequent in POLE ultramutated and MMR-deficient subtypes than in p53-mutant and NSMP subtypes. PD-L1 positivity in TCs, but not in ICs or combined (CPS), was associated with a favorable prognosis in patients with high-risk endometrial cancer. CONCLUSIONS: The distribution and prognostic significance of PD-L1 in TCs versus ICs differ in patients with endometrial cancer, indicating that the separate assessment of PD-L1 in these cells (rather than determining the CPS) may be more relevant to selecting patients eligible for endometrial cancer immunotherapy.


Subject(s)
B7-H1 Antigen/biosynthesis , Endometrial Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/immunology , Cohort Studies , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Humans , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , Retrospective Studies
18.
Gynecol Oncol ; 162(3): 694-701, 2021 09.
Article in English | MEDLINE | ID: mdl-34253388

ABSTRACT

OBJECTIVE: Combined immunohistochemical and molecular classification using the Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) independently predicts prognosis in endometrial carcinoma (EC). As next-generation sequencing (NGS) is entering clinical practice, we evaluated whether more comprehensive immunomolecular profiling (CIMP), including NGS and extended immunohistochemical analysis, could further refine the current ProMisE classification. METHODS: A series of 120 consecutive ECs, classified according to ProMisE, was stained immunohistochemically for CD3, CD8, PD-L1, beta-catenin and L1CAM. An in-house 96 gene NGS panel was performed on a subset of 44 ECs, representing the 4 ProMisE subgroups (DNA polymerase epsilon catalytic subunit exonuclease domain mutated (POLEmut), mismatch repair deficient (MMRd), p53 abnormal (p53 abn) and no specific molecular profile (NSMP) ECs). Cases harboring non-hotspot POLE variants were analyzed with Illumina TruSight Oncology 500 NGS panel (TSO500) as a surrogate for whole-exome sequencing. RESULTS: Eight cases harbored POLE variants, half of which were hotspots. Using TSO500, non-hotspot POLE variants were classified as pathogenic (3) or variant of unknown significance (1). POLEmut and MMRd ECs typically showed higher numbers of CD3+/CD8+ tumor-infiltrating lymphocytes and higher PD-L1 expression in tumor-infiltrating immune cells. p53 abn ECs showed significantly higher L1CAM immunoreactivity and frequently harbored gene amplifications including HER2 (25%), but typically lacked ARID1A or PTEN variants. Beta-catenin-positivity and FGFR2 variants were predominantly found in NSMP ECs. CONCLUSIONS: Our data show that CIMP adds significant value to EC characterization and may help to determine pathogenicity of non-hotspot POLE variants, encountered more frequently than expected in our series. In addition, CIMP may reveal ECs benefitting from immune checkpoint inhibition and allows upfront identification of targetable alterations, such as HER2 amplification in p53 abn ECs.


Subject(s)
Endometrial Neoplasms/genetics , Biomarkers, Tumor/genetics , DNA-Binding Proteins , Endometrial Neoplasms/classification , Endometrial Neoplasms/immunology , Female , High-Throughput Nucleotide Sequencing , Humans , Receptor, Fibroblast Growth Factor, Type 2 , Retrospective Studies , Transcription Factors
19.
Aging (Albany NY) ; 13(12): 16287-16315, 2021 06 16.
Article in English | MEDLINE | ID: mdl-34230220

ABSTRACT

N6-methyladenosine (m6A) RNA methylation is associated with malignant tumor progression and is modulated by various m6A RNA methylation regulator proteins. However, its role in endometrial cancer is unclear. In this work, we analyzed sequence, copy number variation, and clinical data obtained from the TCGA database. Expression was validated using real-time quantitative polymerase chain reaction and immunohistochemistry. Changes in m6A RNA methylation regulators were closely related to the clinicopathological stage and prognosis of endometrial cancer. In particular, ZC3H13, YTHDC1, and METTL14 were identified as potential markers for endometrial cancer diagnosis and prognosis. The TIMER algorithm indicated that immune cell infiltration correlated with changes in ZC3H13, YTHDC1, and METTL14 expression. Meanwhile, ZC3H13 or YTHDC1 knockdown promoted the proliferation and invasion of endometrial cancer cells. Through gene enrichment analysis, we constructed a regulatory network in order to explore the potential molecular mechanism involving ZC3H13, YTHDC1, and METTL14. Virtual screening predicted interactions of potential therapeutic compounds with METTL14 and YTHDC1. These findings advance the understanding of RNA epigenetic modifications in endometrial cancer while identifying m6A regulators associated with immune infiltration, prognosis, and potential treatment strategies.


Subject(s)
Adenosine/analogs & derivatives , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Adenosine/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , DNA Copy Number Variations/genetics , Disease-Free Survival , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Humans , Ligands , Methylation , Middle Aged , Molecular Docking Simulation , Mutation/genetics , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Prognosis , Proportional Hazards Models , RNA, Messenger/genetics , RNA, Messenger/metabolism , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use , Tumor Microenvironment/immunology
20.
Aging (Albany NY) ; 13(12): 16713-16732, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34170849

ABSTRACT

Ferroptosis, a form of programmed cell death induced by excess iron-dependent lipid peroxidation product accumulation, plays a critical role in cancer. However, there are few reports about ferroptosis in endometrial cancer (EC). This article explores the relationship between ferroptosis-related gene (FRG) expression and prognosis in EC patients. One hundred thirty-five FRGs were obtained by mining the literature, retrieving GeneCards and analyzing 552 malignant uterine corpus endometrial carcinoma (UCEC) samples, which were randomly assigned to training and testing groups (1:1 ratio), and 23 normal samples from The Cancer Genome Atlas (TCGA). We established a signature using eight screened FRGs (MDM2, GPX4, PRKAA2, PRNP, SLC11A2, ATP5MC3, PHKG2 and ACO1) related to overall survival using LASSO regression analysis. The samples were divided into low- and high-risk subgroups according to the median risk score. Kaplan-Meier survival curves showed that the low-risk group had better OS. ROC curves showed that this signature performed well in predicting OS (1-, 2-, 3-, and 5-year AUCs of 0.676, 0.775, 0.797, and 0.826, respectively). We systematically analyzed the immune infiltrating profile in UCEC samples from TCGA. Overall, our study identified a novel prognostic signature of 8 FRGs that can potentially predict the prognosis of EC.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Ferroptosis/genetics , Gene Expression Profiling , Lymphocytes, Tumor-Infiltrating/metabolism , Cohort Studies , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Genetic Variation , Humans , Molecular Sequence Annotation , Principal Component Analysis , Prognosis , Protein Interaction Maps/genetics , Reproducibility of Results , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...