Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 396
Filter
1.
Sci Adv ; 10(38): eado6491, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292790

ABSTRACT

Neoantigens arising from somatic mutations are tumor specific and induce antitumor host T cell responses. However, their sequences are individual specific and need to be identified for each patient for therapeutic applications. Here, we present a proteogenomic approach for neoantigen identification, named Neoantigen Selection using a Surrogate Immunopeptidome (NESSIE). This approach uses an autologous wild-type immunopeptidome as a surrogate for the tumor immunopeptidome and allows human leukocyte antigen (HLA)-agnostic identification of both HLA class I (HLA-I) and HLA class II (HLA-II) neoantigens. We demonstrate the direct identification of highly immunogenic HLA-I and HLA-II neoantigens using NESSIE in patients with colorectal cancer and endometrial cancer. Fresh or frozen tumor samples are not required for analysis, making it applicable to many patients in clinical settings. We also demonstrate tumor prevention by vaccination with selected neoantigens in a preclinical mouse model. This approach may benefit personalized T cell-mediated immunotherapies.


Subject(s)
Antigens, Neoplasm , Histocompatibility Antigens Class II , Histocompatibility Antigens Class I , Humans , Antigens, Neoplasm/immunology , Animals , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/genetics , Mice , Histocompatibility Antigens Class II/immunology , Female , Cancer Vaccines/immunology , Colorectal Neoplasms/immunology , Peptides/immunology , Endometrial Neoplasms/immunology , Immunotherapy/methods
2.
J Clin Invest ; 134(17)2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39225094

ABSTRACT

Obesity is a known driver of endometrial cancer. In this issue of the JCI, Gómez-Banoy and colleagues investigated a cohort of patients with advanced endometrial cancer treated with immune checkpoint inhibitors targeting the interaction between programmed cell death receptor-1 (PD-1) and its ligand (PD-L1). Notably, a BMI in the overweight or obese range was paradoxically associated with improved progression-free and overall survival. A second paradox emerged from CT analyses of visceral adipose tissue, viewed as an unhealthy fat depot in most other contexts, the quantity of which was also associated with improved treatment outcomes. Though visceral adiposity may have value as a biomarker to inform personalized treatment strategies, of even greater impact would be if a therapeutic strategy emerges from the future identification of adipose-derived mediators of this putative anticancer immune-priming effect.


Subject(s)
Endometrial Neoplasms , Intra-Abdominal Fat , Humans , Female , Endometrial Neoplasms/immunology , Endometrial Neoplasms/therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/drug therapy , Intra-Abdominal Fat/immunology , Immunotherapy , Immune Checkpoint Inhibitors/therapeutic use , Obesity/immunology , Obesity/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology
3.
Nat Commun ; 15(1): 7695, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227583

ABSTRACT

Neoadjuvant immune checkpoint blockade (ICB) has shown unprecedented activity in mismatch repair deficient (MMRd) colorectal cancers, but its effectiveness in MMRd endometrial cancer (EC) remains unknown. In this investigator-driven, phase I, feasibility study (NCT04262089), 10 women with MMRd EC of any grade, planned for primary surgery, received two cycles of neoadjuvant pembrolizumab (200 mg IV) every three weeks. A pathologic response (primary objective) was observed in 5/10 patients, with 2 patients showing a major pathologic response. No patient achieved a complete pathologic response. A partial radiologic response (secondary objective) was observed in 3/10 patients, 5/10 patients had stable disease and 2/10 patients were non-evaluable on magnetic resonance imaging. All patients completed treatment without severe toxicity (exploratory objective). At median duration of follow-up of 22.5 months, two non-responders experienced disease recurrence. In-depth analysis of the loco-regional and systemic immune response (predefined exploratory objective) showed that monoclonal T cell expansion significantly correlated with treatment response. Tumour-draining lymph nodes displayed clonal overlap with intra-tumoural T cell expansion. All pre-specified endpoints, efficacy in terms of pathologic response as primary endpoint, radiologic response as secondary outcome and safety and tolerability as exploratory endpoint, were reached. Neoadjuvant ICB with pembrolizumab proved safe and induced pathologic, radiologic, and immunologic responses in MMRd EC, warranting further exploration of extended neoadjuvant treatment.


Subject(s)
Antibodies, Monoclonal, Humanized , DNA Mismatch Repair , Endometrial Neoplasms , Immune Checkpoint Inhibitors , Neoadjuvant Therapy , Humans , Female , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/diagnostic imaging , Immune Checkpoint Inhibitors/therapeutic use , Middle Aged , Antibodies, Monoclonal, Humanized/therapeutic use , Aged , Adult , Treatment Outcome
4.
Cancer Rep (Hoboken) ; 7(9): e70009, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39324703

ABSTRACT

BACKGROUND: Endometrial cancer (EC) stands as the predominant gynecological malignancy impacting the female reproductive system on a global scale. N6-methyladenosine, cuproptosis- and ferroptosis-related biomarker is beneficial to the prognostic of tumor patients. Nevertheless, the correlation between m6A-modified lncRNAs and ferroptosis, copper-induced apoptosis in the initiation and progression of EC remains unexplored in existing literature. AIMS: In this study, based on bioinformatics approach, we identified lncRNAs co-expressing with cuproptosis-, ferroptosis-, m6A- related lncRNAs from expression data of EC. By constructing the prognosis model in EC, we screened hub lncRNA signatures affecting prognosis of EC patients. Furthermore, the guiding value of m6A-modified ferroptosis-related lncRNA (mfrlncRNA) features was assessed in terms of prognosis, immune microenvironment, and drug sensitivity. METHOD: Our research harnessed gene expression data coupled with clinical insights derived from The Cancer Genome Atlas (TCGA) collection. To forge prognostic models, we adopted five machine learning approaches, assessing their efficacy through C-index and time-independent ROC analysis. We pinpointed prognostic indicators using the LASSO Cox regression approach. Moreover, we delved into the biological and immunological implications of the discovered lncRNA prognostic signatures. RESULTS: The survival rate for the low-risk group was markedly higher than that for the high-risk group, as evidenced by a significant log-rank test (p < 0.001). The LASSO Cox regression model yielded concordance indices of 0.76 for the training set and 0.77 for the validation set, indicating reliable prognostic accuracy. Enrichment analysis of gene functions linked the identified signature predominantly to endopeptidase inhibitor activity, highlighting the signature's potential implications. Additionally, immune function and drug density emphasized the importance of early diagnosis in EC. CONCLUSION: Five hub lncRNAs in EC were identified through constructing the prognosis model. Those genes might be potential biomarkers to provide valuable reference for targeted therapy and prognostic assessment of EC.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Ferroptosis , RNA, Long Noncoding , Humans , Female , RNA, Long Noncoding/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/mortality , Endometrial Neoplasms/immunology , Endometrial Neoplasms/drug therapy , Ferroptosis/genetics , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Adenosine/analogs & derivatives , Adenosine/metabolism , Immunotherapy/methods , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Computational Biology , Middle Aged
5.
BMC Cancer ; 24(1): 1100, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39232704

ABSTRACT

BACKGROUND: Regulatory T (Treg) cells reportedly play crucial roles in tumor angiogenesis as well as antitumor immunity. In order to explore their therapeutic potential, we investigated the precise prognostic impact of Treg markers in endometrial carcinoma. METHODS: We performed multiplexed immunofluorescence and quantitative image analyses of CD25, FOXP3, CTLA4, and CD45RA in tumor specimens from 176 consecutive patients treated at our institution for primary endometrial carcinomas. Bioinformatics analyses were further conducted to corroborate the findings. RESULTS: High CD25+, FOXP3+, and CD25+FOXP3+CD45RA- stromal cell counts correlated with better overall survival (OS) (p = 0.00019, 0.028 and 0.0012) and MSI-high (p = 0.015, 0.016 and 0.047). High CD45RA+ stromal cell count was associated with superficial myometrial invasion (p = 0.0038). Bioinformatics survival analysis by Kaplan-Meier plotter showed that high CD25, FOXP3, CTLA4, and CD45RA mRNA expressions correlated with better OS (p = 0.046, 0.00042, 0.000044, and 0.0022). Univariate and multivariate analyses with various clinicopathologic prognostic factors indicated that high CD25+ or CD25+FOXP3+CD45RA- stromal cell count was significant and independent for favorable OS (p = 0.0053 and 0.0015). We subsequently analyzed the correlations between the multiplexed immunofluorescence results and treatment-free interval (TFI) after primary chemotherapy in recurrent cases, finding no significant associations. Further analysis revealed that high ratio of CD25+ : CD8+ cell count or CD25+FOXP3+CD45RA- : CD8+ cell count correlated with longer TFI (p = 0.021 and 0.021). CONCLUSION: The current observations suggest that the balance between CD25+ or CD25+FOXP3+CD45RA- cells and CD8+ cells, corresponding to promoting or inhibiting effect on tumor angiogenesis, affect tumor chemosensitivity leading to prognostic significance. CD25+FOXP3+CD45RA- effector Treg tumor infiltration may serve as a useful prognostic biomarker and a potential target for immunotherapeutic manipulation of tumor chemosensitivity by novel management for advanced/recurrent endometrial carcinomas.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Forkhead Transcription Factors , Interleukin-2 Receptor alpha Subunit , Leukocyte Common Antigens , T-Lymphocytes, Regulatory , Humans , Female , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/mortality , Endometrial Neoplasms/genetics , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Prognosis , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Biomarkers, Tumor/metabolism , Leukocyte Common Antigens/metabolism , Middle Aged , Interleukin-2 Receptor alpha Subunit/metabolism , Aged , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Adult , Kaplan-Meier Estimate , CTLA-4 Antigen/metabolism , Aged, 80 and over
6.
Lab Invest ; 104(9): 102126, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39174007

ABSTRACT

This study used artificial intelligence (AI)-based analysis to investigate the immune microenvironment in endometrial cancer (EC). We aimed to evaluate the potential of AI-based immune metrics as prognostic biomarkers. In total, 296 cases with EC were classified into 4 molecular subtypes: polymerase epsilon ultramutated (POLEmut), mismatch repair deficiency (MMRd), p53 abnormal (p53abn), and no specific molecular profile (NSMP). AI-based methods were used to evaluate the following immune metrics: total tumor-infiltrating lymphocytes (TIL), intratumoral TIL, stromal TIL, and tumor cells using Lunit SCOPE IO, as well as CD4+, CD8+, and FOXP3+ T cells using immunohistochemistry (IHC) by QuPath. These 7 immune metrics were used to perform unsupervised clustering. PD-L1 22C3 IHC expression was also evaluated. Clustering analysis demonstrated 3 distinct immune microenvironment groups: immune active, immune desert, and tumor dominant. The immune-active group was highly prevalent in POLEmut, and it was also seen in other molecular subtypes. Although the immune-desert group was more frequent in NSMP and p53mut, it was also detected in MMRd and POLEmut. POLEmut showed the highest levels of CD4+ and CD8+ T cells, total TIL, intratumoral TIL, and stromal TIL with the lowest levels of FOXP3+/CD8+ ratio. In contrast, p53abn in the immune-active group showed higher FOXP3+/CD4+ and FOXP3+/CD8+ ratios. The immune-active group was associated with favorable overall survival and recurrence-free survival. In the NSMP subtype, a significant association was observed between immune active and better recurrence-free survival. The PD-L1 22C3 combined positive score (CPS) showed significant differences among the 3 groups, with the immune-active group having the highest median CPS and frequency of CPS ≥ 1%. The immune microenvironment of EC was variable within molecular subtypes. Within the same immune microenvironment group, significant differences in immune metrics and T cell composition were observed according to molecular subtype. AI-based immune microenvironment groups served as prognostic markers in ECs, with the immune-active group associated with favorable outcomes.


Subject(s)
Endometrial Neoplasms , Lymphocytes, Tumor-Infiltrating , Tumor Microenvironment , Humans , Female , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Tumor Microenvironment/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Prognosis , Middle Aged , Aged , Adult , Biomarkers, Tumor/metabolism
7.
Am J Reprod Immunol ; 92(2): e13916, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166450

ABSTRACT

BACKGROUND: Uterine endometrial cancer (UEC) is a common gynecological estrogen-dependent carcinoma, usually accompanied by intermenstrual bleeding. Active heme metabolism frequently plays an increasingly important role in many diseases, especially in cancers. Tumor-associated macrophages (TAMs) are the major population in the immune microenvironment of UEC. However, the roles of heme metabolisms in the crosstalk between UEC cells (UECCs) and macrophages are unclear. MATERIALS AND METHODS: In our study, by using TCGA database analysis, integration analysis of the protein-protein interaction (PPI) network and sample RNA transcriptome sequencing were done. The expression level of both heme-associated molecules and iron metabolism-related molecules were measured by quantitative real-time polymerase chain reaction. Heme level detection was done through dehydrohorseradish peroxidase assay. In addition to immunohistochemistry, phagocytosis assay of macrophages, immunofluorescence staining, intracellular ferrous iron staining, as well as enzyme-linked immune sorbent assay were performed. RESULTS: In the study, we verified that heme accumulation in UECCs is apparently higher than in endometrial epithelium cells. Low expression of succinate dehydrogenase B under the regulation of estrogen contributes to over-production of succinate and heme accumulation in UECC. More importantly, excessive heme in UECCs impaired macrophage phagocytosis by regulation of CD36. Mechanistically, this process is dependent on toll-like receptor (TLR4)/type I interferons alpha (IFN Iα) regulatory axis in macrophage. CONCLUSION: Collectively, these findings elucidate that active heme metabolism of UECCs directly decreases phagocytosis by controlling the secretion of TLR4-mediated IFN Iα and the expression of CD36, and further contributing to the immune escape of UEC.


Subject(s)
CD36 Antigens , Endometrial Neoplasms , Heme , Interferon Type I , Phagocytosis , Signal Transduction , Toll-Like Receptor 4 , Female , Humans , Toll-Like Receptor 4/metabolism , Heme/metabolism , Endometrial Neoplasms/immunology , Endometrial Neoplasms/metabolism , Interferon Type I/metabolism , CD36 Antigens/metabolism , Macrophages/immunology , Macrophages/metabolism , Tumor Microenvironment/immunology
8.
Sci Rep ; 14(1): 19111, 2024 08 17.
Article in English | MEDLINE | ID: mdl-39154063

ABSTRACT

Endometrial cancer (EC) is associated with significant risk factors such as polycystic ovarian syndrome (PCOS) and sedentary behavior. In our study, we aim to employ machine learning algorithms to investigate the potential molecular processes that underlie their interaction and explore their respective roles in the diagnosis and immunotherapy of EC. The GEO database provides access to microarray data, which was utilized in this study to identify gene expression modules associated with PCOS and sedentary behavior, using weighted gene expression network analysis (WGCNA). Cluego software was then employed to investigate the energy enrichment of shared pathways in both PCOS and sedentary individuals, and differential gene analysis was used to confirm another two databases. The miRNAs-mRNAs controlled network was constructed to verify the pathway. The immune-related factors of the shared pathway in EC were then analyzed. Finally, to validate our findings, we conducted cell experiments using EC cell lines (AN3CA, KLE, Ishikawa, RL95-2, and HEC-1A). We found that increased intracellular aromatic compound anabolism is a common feature of both PCOS and sedentary individuals. We then developed a disease pathway model that was based on the common genetic characteristics of PCOS and sedentary behavior. We utilized pathway typing in EC samples and found a significant survival difference between the two subgroups, with the upregulated expression type exhibiting an immune-hot phenotype. Finally, the experimental results confirmed the expression of the hub gene (NAA15) in EC. The findings of our study suggest that genes related to the intracellular aromatic compound metabolic pathway can be used for immunotherapy of EC.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Polycystic Ovary Syndrome , Sedentary Behavior , Humans , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/immunology , Polycystic Ovary Syndrome/metabolism , Female , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Biomarkers, Tumor/genetics , Gene Regulatory Networks , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , MicroRNAs/genetics , MicroRNAs/metabolism , Gene Expression Profiling , Machine Learning
9.
J Ovarian Res ; 17(1): 162, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39123216

ABSTRACT

BACKGROUND: Dachsous cadherin related 1 (DCHS1) is one of calcium-dependent adhesion membrane proteins and is mainly involved in the development of mammalian tissues. There is a lack of more detailed research on the biological function of DCHS1 in pan-cancer. MATERIALS AND METHODS: We evaluated the expression, the prognostic value, the diagnostic value and genomic alterations of DCHS1 by using the databases, including TCGA, UALCAN, HPA, GEPIA2.0 and GSCA. We employed the databases of UCSC, TIMER2.0, TISIDB, GSCA to analyze the association between DCHS1 expression and the immune microenvironment, stemness, TMB, MSI and anticancer drug sensitivity. BioGRID, STRING and GEPIA2.0 were used to perform protein interaction and functional enrichment analysis. Real-time quantitative PCR, CCK8, Transwell assay and Western blot were performed to determine the function of DCHS1 in UCEC. RESULTS: DCHS1 is differentially expressed in many cancers and its expression is significantly associated with tumor prognosis and diagnosis. DCHS1 expression was significantly correlated with the infiltration of cancer-associated fibroblasts (CAFs), Endothelial cell (ECs), and Hematopoietic stem cell in most cancers. In addition, DCHS1 was significantly associated with sensitivity to many antitumor drugs. Functional enrichment analysis revealed that DCHS1-related proteins were involved in Focal adhesion, Endometrial cancer and Wnt signaling pathway. GSEA results showed that DCHS1 was related to epithelial-mesenchymal transition (EMT) in many cancers. In vitro experiments in UCEC showed that DCHS1 regulated cell proliferation, migration and EMT. CONCLUSIONS: Our findings indicated that DCHS1 might be a novel prognostic and diagnostic biomarker and immunotherapy target, and plays an important role in the proliferation, migration and EMT in UCEC.


Subject(s)
Biomarkers, Tumor , Endometrial Neoplasms , Epithelial-Mesenchymal Transition , Female , Humans , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cadherin Related Proteins , Cell Line, Tumor , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Gene Expression Regulation, Neoplastic , Prognosis , Tumor Microenvironment/immunology
10.
Mol Biomed ; 5(1): 32, 2024 08 14.
Article in English | MEDLINE | ID: mdl-39138733

ABSTRACT

Endometrial cancer (UCEC) is one of three major malignant tumors in women. The HOX gene regulates tumor development. However, the potential roles of HOX in the expression mechanism of multiple cell types and in the development and progression of tumor microenvironment (TME) cell infiltration in UCEC remain unknown. In this study, we utilized both the The Cancer Genome Atlas (TCGA) database and International Cancer Genome Consortium (ICGC) database to analyze transcriptome data of 529 patients with UCEC based on 39 HOX genes, combing clinical information, we discovered HOX gene were a pivotal factor in the development and progression of UCEC and in the formation of TME diversity and complexity. Here, a new scoring system was developed to quantify individual HOX patterns in UCEC. Our study found that patients in the low HOX score group had abundant anti-tumor immune cell infiltration, good tumor differentiation, and better prognoses. In contrast, a high HOX score was associated with blockade of immune checkpoints, which enhances the response to immunotherapy. The Real-Time quantitative PCR (RT-qPCR) and Immunohistochemistry (IHC) exhibited a higher expression of the HOX gene in the tumor patients. We revealed that the significant upregulation of the HOX gene in the epithelial cells can activate signaling pathway associated with tumour invasion and metastasis through single-cell RNA sequencing (scRNA-seq), such as nucleotide metabolic proce and so on. Finally, a risk prognostic model established by the positive relationship between HOX scores and cancer-associated fibroblasts (CAFs) can predict the prognosis of individual patients by scRNA-seq and transcriptome data sets. In sum, HOX gene may serve as a potential biomarker for the diagnosis and prediction of UCEC and to develop more effective therapeutic strategies.


Subject(s)
Endometrial Neoplasms , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Humans , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Female , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Prognosis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcriptome , Genes, Homeobox/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Databases, Genetic , Gene Expression Profiling , Middle Aged
11.
BMC Womens Health ; 24(1): 429, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068426

ABSTRACT

BACKGROUND: Given the significant role of immune-related genes in uterine corpus endometrial carcinoma (UCEC) and the long-term outcomes of patients, our objective was to develop a prognostic risk prediction model using immune-related genes to improve the accuracy of UCEC prognosis prediction. METHODS: The Limma, ESTIMATE, and CIBERSORT methods were used for cluster analysis, immune score calculation, and estimation of immune cell proportions. Univariate and multivariate analyses were utilized to develop a prognostic risk model for UCEC. Risk model scores and nomograms were used to evaluate the models. String constructs a protein-protein interaction (PPI) network of genes. The qRT-PCR, immunofluorescence, and immunohistochemistry (IHC) all confirmed the genes. RESULTS: Cluster analysis divided the immune-related genes into four subtypes. 33 immune-related genes were used to independently predict the prognosis of UCEC and construct the prognosis model and risk score. The analysis of the survival nomogram indicated that the model has excellent predictive ability and strong reliability for predicting the survival of patients with UCEC. The protein-protein interaction network analysis of key genes indicates that four genes play a pivotal role in interactions: GZMK, IL7, GIMAP, and UBD. The quantitative real-time polymerase chain reaction (qRT-PCR), immunofluorescence, and immunohistochemistry (IHC) all confirmed the expression of the aforementioned genes and their correlation with immune cell levels. This further revealed that GZMK, IL7, GIMAP, and UBD could potentially serve as biomarkers associated with immune levels in endometrial cancer. CONCLUSION: The study identified genes related to immune response in UCEC, including GZMK, IL7, GIMAP, and UBD, which may serve as new biomarkers and therapeutic targets for evaluating immune levels in the future.


Subject(s)
Endometrial Neoplasms , Nomograms , Female , Humans , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Prognosis , Risk Assessment/methods , Protein Interaction Maps/genetics , Middle Aged , Biomarkers, Tumor/genetics , Cluster Analysis
13.
Cancer ; 130(18): 3082-3086, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-38962843

ABSTRACT

In the past year, notable advances were achieved toward improving oncological outcomes in patients with advanced and recurrent endometrial cancer because of reporting of high-level results of several phase 3 clinical trial combining an immune checkpoint inhibitor with chemotherapy in the first-line setting. For the first time, patients with recurrent or advanced endometrial cancer have options for treatment that are superior to traditional chemotherapy alone. What remains to be determined is population specificity of these recommendations. All four major studies were in agreement that patients with endometrial cancer with deficient mismatch repair markedly benefited from addition of an immune checkpoint inhibitor for progression-free survival; some showed preliminary results demonstrating a potential overall survival. Molecular characterization details are needed to determine if and which patients with tumors that are mismatch proficient should receive this new combination approach. PLAIN LANGUAGE SUMMARY: Combining an immune checkpoint inhibitor with chemotherapy in the first-line setting treatment of patients with advanced endometrial cancer improved progression-free survival, especially in patients with mismatch repair deficiency. Improving patient selection with potential biomarkers of sensitivity and biomarkers of resistance is key in developing the next clinical trials and will assist in directing therapy to the correct patients and minimize toxicity.


Subject(s)
Endometrial Neoplasms , Immune Checkpoint Inhibitors , Immunotherapy , Humans , Female , Endometrial Neoplasms/therapy , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , DNA Mismatch Repair , Progression-Free Survival , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
14.
Aging (Albany NY) ; 16(14): 11248-11274, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39079132

ABSTRACT

Endometrial cancer (EC) is a fatal gynecologic tumor. Bioinformatic tools are increasingly developed to screen out molecular targets related to EC. Our study aimed to identify stemness-related prognostic biomarkers for new therapeutic strategies in EC. In this study, we explored the prognostic value of cancer stem cells (CSCs), characterized by self-renewal and unlimited proliferation, and its correlation with immune infiltrates in EC. Transcriptome and somatic mutation profiles of EC were downloaded from TCGA database. Based on their stemness signature and DEGs, EC patients were divided into two subtypes via consensus clustering, and patients in Stemness Subtype I presented significantly better OS and DFS than Stemness Subtype II. Subtype I also displayed better clinicopathological features, and genomic variations demonstrated different somatic mutation from subtype II. Additionally, two stemness subtypes had distinct tumor immune microenvironment patterns. In the end, three machine learning algorithms were applied to construct a 7-gene stemness subtype risk model, which were further validated in an external independent EC cohort in our hospital. This novel stemness-based classification could provide a promising prognostic predictor for EC and may guide physicians in selecting potential responders for preferential use of immunotherapy. This novel stemness-dependent classification method has high value in predicting the prognosis, and also provides a reference for clinicians in selecting sensitive immunotherapy methods for EC patients.


Subject(s)
Endometrial Neoplasms , Machine Learning , Mutation , Neoplastic Stem Cells , Tumor Microenvironment , Humans , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/mortality , Female , Prognosis , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Transcriptome , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Middle Aged , Gene Expression Profiling
15.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38955419

ABSTRACT

BACKGROUND: Patients with mismatch repair-deficient (MMRd) endometrial cancer (EC) can derive great benefit from immune checkpoint inhibitors (ICI). However not all responses and predictors of primary resistance are lacking. METHODS: We compared the immune tumor microenvironment of MMRd EC ICI-responders (Rs) and ICI non-responders (NRs), using spatial multiplexed immune profiling and unsupervised hierarchical clustering analysis. RESULTS: Overall, NRs exhibited drastically lower CD8+, absent terminally differentiated T cells, lack of mature tertiary lymphoid structures and dendritic cells, as well as loss of human leukocyte antigen class I. However, no single marker could predict R versus NR with confidence. Clustering analysis identified a combination of four immune features that demonstrated that accurately predicted ICI response, with a discriminative power of 92%. Finally, 80% of NRs lacked programmed death-ligand 1, however, 60% exhibited another actionable immune checkpoint (T-cell immunoglobulin and mucin containing protein-3, indoleamine 2,3-dioxygenase 1, or lymphocyte activation gene 3). CONCLUSIONS: These findings underscore the potential of immune tumor microenvironment features for identifying patients with MMRd EC and primary resistance to ICI who should be oriented towards trials testing novel immunotherapeutic combinations.


Subject(s)
DNA Mismatch Repair , Endometrial Neoplasms , Immune Checkpoint Inhibitors , Humans , Female , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/immunology , Endometrial Neoplasms/genetics , Tumor Microenvironment , Middle Aged , Aged
16.
J Clin Invest ; 134(17)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38900575

ABSTRACT

BACKGROUNDObesity is the foremost risk factor in the development of endometrial cancer (EC). However, the impact of obesity on the response to immune checkpoint inhibitors (ICI) in EC remains poorly understood. This retrospective study investigates the association among BMI, body fat distribution, and clinical and molecular characteristics of EC patients treated with ICI.METHODSWe analyzed progression-free survival (PFS) and overall survival (OS) in EC patients treated with ICI, categorized by BMI, fat-mass distribution, and molecular subtypes. Incidence of immune-related adverse events (irAEs) after ICI was also assessed based on BMI status.RESULTS524 EC patients were included in the study. Overweight and obese patients exhibited a significantly prolonged PFS and OS compared with normal BMI patients after treatment with ICI. Multivariable Cox's regression analysis confirmed the independent association of overweight and obesity with improved PFS and OS. Elevated visceral adipose tissue (VAT) was identified as a strong independent predictor for improved PFS to ICI. Associations between obesity and OS/PFS were particularly significant in the copy number-high/TP53abnormal (CN-H/TP53abn) EC molecular subtype. Finally, obese patients demonstrated a higher irAE rate compared with normal BMI individuals.CONCLUSIONObesity is associated with improved outcomes to ICI in EC patients and a higher rate of irAEs. This association is more pronounced in the CN-H/TP53abn EC molecular subtype.FUNDINGNIH/NCI Cancer Center; MSK Gerstner Physician Scholars Program; National Center for Advancing Translational Sciences (NCATS); Cycle for Survival; Breast Cancer Research Foundation.


Subject(s)
Adiposity , Body Mass Index , Endometrial Neoplasms , Immune Checkpoint Inhibitors , Obesity , Humans , Female , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Endometrial Neoplasms/immunology , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/genetics , Middle Aged , Aged , Obesity/immunology , Retrospective Studies , Progression-Free Survival , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Mod Pathol ; 37(8): 100532, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38848896

ABSTRACT

Endometrial serous carcinoma (ESC) is an uncommon, aggressive type of endometrial cancer. While immune checkpoint blockade has emerged as a promising treatment option for endometrial carcinomas, research on the expression of immune checkpoints that could serve as prospective immunotherapy targets in ESC is limited. We examined the prevalence and prognostic value of lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin and ITIM domain (TIGIT), V-domain immunoglobulin (Ig) suppressor of T-cell activation (VISTA), and indoleamine 2,3-dioxygenase 1 (IOD1) in 94 cases of ESC and correlated their expression with CD8+ and FOXP3+ tumor-infiltrating lymphocytes (TILs). We observed a positive correlation among LAG-3, TIGIT, and VISTA expressed on immune cells, and among these markers and CD8+ and FOXP3+ TIL densities. In Kaplan-Meier survival analysis, tumors with high levels of LAG-3 and TIGIT expression had better progression-free survival (PFS) and overall survival (OS) than those with lower levels of expression (LAG-3: PFS, P = .03, OS, P = .04; TIGIT: PFS, P = .01, OS, P = .009). In multivariate analysis, only high TIGIT expression was of independent prognostic value for better OS. VISTA expression in immune or tumor cells, and IDO1 expression in tumor cells, did not show a significant association with survival. Our data indicate that LAG-3, TIGIT, and VISTA immune checkpoints have roles in the microenvironment of ESC, and their expression patterns highlight the complex interactions among the different components of this system. High levels of these markers, together with high CD8+ TIL, suggest the potential immunogenicity of a subset of these tumors. Further studies are needed to elucidate the roles of various immune components in the ESC microenvironment and their association with intrinsic tumor properties.


Subject(s)
Antigens, CD , B7 Antigens , Biomarkers, Tumor , Endometrial Neoplasms , Indoleamine-Pyrrole 2,3,-Dioxygenase , Lymphocyte Activation Gene 3 Protein , Lymphocytes, Tumor-Infiltrating , Receptors, Immunologic , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Antigens, CD/metabolism , B7 Antigens/metabolism , Biomarkers, Tumor/analysis , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/immunology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Endometrial Neoplasms/mortality , Endometrial Neoplasms/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Prognosis , Receptors, Immunologic/metabolism , Tumor Microenvironment/immunology
18.
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
19.
Gynecol Oncol ; 187: 1-11, 2024 08.
Article in English | MEDLINE | ID: mdl-38696842

ABSTRACT

OBJECTIVE: FAT3 and LRP1B are two tumor suppressor genes with high mutation frequency in multiple cancer types, we sought to investigate the prognostic and immunological significance of these two genes in EC. METHODS: Based on a cohort of 502 EC samples, we conducted a comprehensive analysis of its multidimensional data types including genomic, transcriptomic, and clinical information, the potential impact of FAT3 and LRP1B co-mutation on antitumor immune response and prognosis were systematically discussed. RESULTS: We observed that FAT3 and LRP1B co-mutation was not only defined a dataset with prominently increased TMB, decreased tumor aneuploidy, and specially enriched in MSI-H subtype, but also manifested increased expression of immune-related markers, especially exclusive upregulation of PD-L1 levels and higher PD-L1+/CD8A+ proportion. Further analysis focused on lymphocyte infiltration and pathway enrichment explored the immune cell composition of the microenvironment and underlying molecular mechanisms affecting tumor development. Furthermore, EC patients with FAT3 and LRP1B co-mutation possessed significantly prolonged PFS and OS, and the co-mutation status was proved to be an independent prognostic factor. And a nomogram with high predictive performance was constructed by incorporating co-mutation with clinical features. More strikingly, the prognosis of MSI-H patients in EC with co-mutation was significantly improved, and their survival reached a level consistent with the POLE subtype. CONCLUSIONS: In endometrial cancer, co-mutation of FAT3 and LRP1B not only leads to activation of the immune state, but also represents a subgroup with an improved prognosis, particularly in the MSI-H subtype.


Subject(s)
Endometrial Neoplasms , Mutation , Receptors, LDL , Female , Humans , Middle Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/immunology , Endometrial Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , Receptors, LDL/genetics , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
20.
Gynecol Oncol ; 187: 85-91, 2024 08.
Article in English | MEDLINE | ID: mdl-38735144

ABSTRACT

BACKGROUND: The combination of immune checkpoint inhibitors (ICIs) and platinum-based chemotherapy has emerged as a highly promising primary option for advanced or recurrent endometrial cancer (EC). The study aimed to evaluate treatment efficacy of ICIs with cytotoxic chemotherapy in EC. METHODS: We conducted a comprehensive review of randomized controlled trials up to November 11, 2023, focusing on immunotherapy combined with chemotherapy versus chemotherapy alone for EC. The primary endpoint was the pooled hazard ratio (HR), which was further analyzed across subgroups based on mismatch repair (MMR) status, race, histology, and programmed death-ligand 1 (PD-L1) status. The protocol was registered in PROSPERO (CRD42023475669). FINDINGS: Four trials with 2335 patients were analyzed. ICIs with chemotherapy significantly prolonged progression-free survival (PFS) (HR, 0.70; 95% CI, 0.62-0.79) and overall survival (OS) (HR, 0.75; 95% CI, 0.63-0.89) compared to chemotherapy alone. Stratification by MMR status showed substantial benefits for dMMR (PFS; HR, 0.33; 95% CI, 0.26-0.43; OS; HR, 0.37; 95% CI, 0.22-0.91) over pMMR cohorts in both PFS and OS. In the subgroup analysis, there was significant PFS advantage in Caucasian (HR, 0.63; 95% CI, 0.54-0.72) over non-Caucasian, in endometrioid histology (HR, 0.66; 95% CI, 0.56-0.78) over non-endometrioid, and in PD-L1 positive (HR, 0.39; 95% CI, 0.19-0.81) over PD-L1 negative population. INTERPRETATION: ICIs combined with platinum-based chemotherapy significantly prolonged PFS and OS in patients with advanced or recurrent EC. Patients with dMMR status, Caucasians, endometrioid histology, and positive PD-L1 status showed significant PFS benefits, emphasizing the need for personalized treatment approaches to improve outcomes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Endometrial Neoplasms , Immune Checkpoint Inhibitors , Neoplasm Recurrence, Local , Humans , Female , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/pathology , Endometrial Neoplasms/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/immunology , Randomized Controlled Trials as Topic , Progression-Free Survival
SELECTION OF CITATIONS
SEARCH DETAIL