Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Front Immunol ; 15: 1414716, 2024.
Article in English | MEDLINE | ID: mdl-39315092

ABSTRACT

Ovarian cancer (OC) is the most lethal gynecologic malignancy worldwide. The major clinical challenge includes the asymptomatic state of the disease, making diagnosis possible only at advanced stages. Another OC complication is the high relapse rate and poor prognosis following the standard first-line treatment with platinum-based chemotherapy. At present, numerous clinical trials are being conducted focusing on immunotherapy in OC; nevertheless, there are still no FDA-approved indications. Personalized decision regarding the immunotherapy, including immune checkpoint blockade and immune cell-based immunotherapies, can depend on the effective antigen presentation required for the cytotoxic immune response. The major aim of our study was to uncover tumor-specific transcriptional and epigenetic changes in peripheral blood monocytes in patients with high-grade serous ovarian cancer (HGSOC). Another key point was to elucidate how chemotherapy can reprogram monocytes and how that relates to changes in other immune subpopulations in the blood. To this end, we performed single-cell RNA sequencing of peripheral blood mononuclear cells (PBMCs) from patients with HGSOC who underwent neoadjuvant chemotherapeutic treatment (NACT) and in treatment-naïve patients. Monocyte cluster was significantly affected by tumor-derived factors as well as by chemotherapeutic treatment. Bioinformatical analysis revealed three distinct monocyte subpopulations within PBMCs based on feature gene expression - CD14.Mn.S100A8.9hi, CD14.Mn.MHC2hi and CD16.Mn subsets. The intriguing result was that NACT induced antigen presentation in monocytes by the transcriptional upregulation of MHC class II molecules, but not by epigenetic changes. Increased MHC class II gene expression was a feature observed across all three monocyte subpopulations after chemotherapy. Our data also demonstrated that chemotherapy inhibited interferon-dependent signaling pathways, but activated some TGFb-related genes. Our results can enable personalized decision regarding the necessity to systemically re-educate immune cells to prime ovarian cancer to respond to anti-cancer therapy or to improve personalized prescription of existing immunotherapy in either combination with chemotherapy or a monotherapy regimen.


Subject(s)
Antigen Presentation , Cystadenocarcinoma, Serous , Monocytes , Ovarian Neoplasms , Humans , Female , Monocytes/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/drug therapy , Antigen Presentation/drug effects , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/immunology , Middle Aged , Neoplasm Grading , Gene Expression Regulation, Neoplastic/drug effects , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoadjuvant Therapy/methods , Epigenesis, Genetic
2.
Proc Natl Acad Sci U S A ; 121(34): e2402998121, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39133838

ABSTRACT

Significant racial disparities exist between Black and White patients with uterine serous carcinoma (USC). While the reasons for these disparities are unclear, several studies have demonstrated significantly different rates of driver mutations between racial groups, including TP53. However, limited research has investigated the transcriptional differences of tumors or the composition of the tumor microenvironment (TME) between these groups. Here, we report the single-nuclei RNA-sequencing profiles of primary USC tumors from diverse racial backgrounds. We find that there are significant differences between the tumors of Black and White patients. Tumors from Black patients exhibited higher expression of specific genes associated with aggressiveness, such as PAX8, and axon guidance and synaptic signaling pathways. We also demonstrated that T cell populations are reduced in the tumor tissue compared to matched benign, while anti-inflammatory macrophage populations are retained within the TME. Furthermore, we investigated the connection between PAX8 overexpression and immunosuppression in USC through regulation of several cytokines and chemokines. Notably, we show that PAX8 activity can influence macrophage gene expression and protein secretion. These studies provide a detailed understanding of the USC transcriptome and TME, and identify differences in tumor biology from patients of different racial backgrounds.


Subject(s)
PAX8 Transcription Factor , Signal Transduction , Tumor Microenvironment , Uterine Neoplasms , Humans , Female , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology , PAX8 Transcription Factor/genetics , PAX8 Transcription Factor/metabolism , Signal Transduction/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/immunology , Gene Expression Regulation, Neoplastic , White People/genetics , Single-Cell Analysis , Middle Aged
3.
Front Immunol ; 15: 1394497, 2024.
Article in English | MEDLINE | ID: mdl-38947323

ABSTRACT

Despite advances in surgical and therapeutic approaches, high-grade serous ovarian carcinoma (HGSOC) prognosis remains poor. Surgery is an indispensable component of therapeutic protocols, as removal of all visible tumor lesions (cytoreduction) profoundly improves the overall survival. Enhanced predictive tools for assessing cytoreduction are essential to optimize therapeutic precision. Patients' immune status broadly reflects the tumor cell biological behavior and the patient responses to disease and treatment. Serum cytokine profiling is a sensitive measure of immune adaption and deviation, yet its integration into treatment paradigms is underexplored. This study is part of the IMPACT trial (NCT03378297) and aimed to characterize immune responses before and during primary treatment for HGSOC to identify biomarkers for treatment selection and prognosis. Longitudinal serum samples from 22 patients were collected from diagnosis until response evaluation. Patients underwent primary cytoreductive surgery or neoadjuvant chemotherapy (NACT) based on laparoscopy scoring. Twenty-seven serum cytokines analyzed by Bio-Plex 200, revealed two immune phenotypes at diagnosis: Immune High with marked higher serum cytokine levels than Immune Low. The immune phenotypes reflected the laparoscopy scoring and allocation to surgical treatment. The five Immune High patients undergoing primary cytoreductive surgery exhibited immune mobilization and extended progression-free survival, compared to the Immune Low patients undergoing the same treatment. Both laparoscopy and cytoreductive surgery induced substantial and transient changes in serum cytokines, with upregulation of the inflammatory cytokine IL-6 and downregulation of the multifunctional cytokines IP-10, Eotaxin, IL-4, and IL-7. Over the study period, cytokine levels uniformly decreased in all patients, leading to the elimination of the initial immune phenotypes regardless of treatment choice. This study reveals distinct pre-treatment immune phenotypes in HGSOC patients that might be informative for treatment stratification and prognosis. This potential novel biomarker holds promise as a foundation for improved assessment of treatment responses in patients with HGSOC. ClinicalTrials.gov Identifier: NCT03378297.


Subject(s)
Cystadenocarcinoma, Serous , Cytokines , Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Ovarian Neoplasms/mortality , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/therapy , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/blood , Cystadenocarcinoma, Serous/diagnosis , Cytokines/blood , Middle Aged , Aged , Neoadjuvant Therapy , Phenotype , Cytoreduction Surgical Procedures , Biomarkers, Tumor/blood , Neoplasm Grading , Prognosis , Treatment Outcome , Adult
4.
J Ovarian Res ; 17(1): 140, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970121

ABSTRACT

BACKGROUND: Ovarian serous cystadenocarcinoma, accounting for about 90% of ovarian cancers, is frequently diagnosed at advanced stages, leading to suboptimal treatment outcomes. Given the malignant nature of the disease, effective biomarkers for accurate prediction and personalized treatment remain an urgent clinical need. METHODS: In this study, we analyzed the microbial contents of 453 ovarian serous cystadenocarcinoma and 68 adjacent non-cancerous samples. A univariate Cox regression model was used to identify microorganisms significantly associated with survival and a prognostic risk score model constructed using LASSO Cox regression analysis. Patients were subsequently categorized into high-risk and low-risk groups based on their risk scores. RESULTS: Survival analysis revealed that patients in the low-risk group had a higher overall survival rate. A nomogram was constructed for easy visualization of the prognostic model. Analysis of immune cell infiltration and immune checkpoint gene expression in both groups showed that both parameters were positively correlated with the risk level, indicating an increased immune response in higher risk groups. CONCLUSION: Our findings suggest that microbial profiles in ovarian serous cystadenocarcinoma may serve as viable clinical prognostic indicators. This study provides novel insights into the potential impact of intratumoral microbial communities on disease prognosis and opens avenues for future therapeutic interventions targeting these microorganisms.


Subject(s)
Cystadenocarcinoma, Serous , Immunotherapy , Ovarian Neoplasms , Humans , Female , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Ovarian Neoplasms/microbiology , Ovarian Neoplasms/pathology , Prognosis , Immunotherapy/methods , Middle Aged , Microbiota , Biomarkers, Tumor , Aged , Survival Analysis , Adult
5.
Sci Rep ; 14(1): 16140, 2024 07 12.
Article in English | MEDLINE | ID: mdl-38997411

ABSTRACT

High-grade serous ovarian cancer (HGSOC) is an aggressive disease known to develop resistance to chemotherapy. We investigated the prognostic significance of tumor cell states and potential mechanisms underlying chemotherapy resistance in HGSOC. Transcriptome deconvolution was performed to address cellular heterogeneity. Kaplan-Meier survival curves were plotted to illustrate the outcomes of patients with varying cellular abundances. The association between gene expression and chemotherapy response was tested. After adjusting for surgery status and grading, several cell states exhibited a significant correlation with patient survival. Cell states can organize into carcinoma ecotypes (CE). CE9 and CE10 were proinflammatory, characterized by higher immunoreactivity, and were associated with favorable survival outcomes. Ratios of cell states and ecotypes had better prognostic abilities than a single cell state or ecotype. A total of 1265 differentially expressed genes were identified between samples with high and low levels of C9 or CE10. These genes were partitioned into three co-expressed modules, which were associated with tumor cells and immune cells. Pogz was identified to be linked with immune cell genes and the chemotherapy response of paclitaxel. Collectively, the survival of HGSOC patients is correlated with specific cell states and ecotypes.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/mortality , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/drug therapy , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/immunology , Prognosis , Gene Expression Regulation, Neoplastic , Middle Aged , Neoplasm Grading , Transcriptome , Kaplan-Meier Estimate , Aged , Drug Resistance, Neoplasm/genetics
6.
Cancer Res Commun ; 4(8): 1919-1932, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38984891

ABSTRACT

Chromobox 2 (CBX2), an epigenetic reader and component of polycomb repressor complex 1, is highly expressed in >75% of high-grade serous carcinoma. Increased CBX2 expression is associated with poorer survival, whereas CBX2 knockdown leads to improved chemotherapy sensitivity. In a high-grade serous carcinoma immune-competent murine model, knockdown of CBX2 decreased tumor progression. We sought to explore the impact of modulation of CBX2 on the tumor immune microenvironment (TIME), understanding that the TIME plays a critical role in disease progression and development of therapy resistance. Exploration of existing datasets demonstrated that elevated CBX2 expression significantly correlated with specific immune cell types in the TIME. RNA sequencing and pathway analysis of differentially expressed genes demonstrated immune signature enrichment. Confocal microscopy and co-culture experiments found that modulation of CBX2 leads to increased recruitment and infiltration of macrophages. Flow cytometry of macrophages cultured with CBX2-overexpressing cells showed increased M2-like macrophages and decreased phagocytosis activity. Cbx2 knockdown in the Trp53-null, Brca2-null ID8 syngeneic murine model (ID8 Trp53-/-Brca2-/-) led to decreased tumor progression compared with the control. NanoString immuno-oncology panel analysis suggested that knockdown in Cbx2 shifts immune cell composition, with an increase in macrophages. Multispectral immunohistochemistry (mIHC) further confirmed an increase in macrophage infiltration. Increased CBX2 expression leads to recruitment and polarization of protumor macrophages, and targeting CBX2 may serve to modulate the TIME to enhance the efficacy of immune therapies. SIGNIFICANCE: CBX2 expression correlates with the TIME. CBX2 modulation shifts the macrophage population, potentially leading to an immunosuppressive microenvironment, highlighting CBX2 as a target to improve efficacy of immunotherapy.


Subject(s)
Tumor Microenvironment , Animals , Mice , Humans , Female , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Macrophages/immunology , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
7.
Clin Cancer Res ; 30(16): 3481-3498, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38837893

ABSTRACT

PURPOSE: The purpose of this study was to evaluate RB1 expression and survival across ovarian carcinoma histotypes and how co-occurrence of BRCA1 or BRCA2 (BRCA) alterations and RB1 loss influences survival in tubo-ovarian high-grade serous carcinoma (HGSC). EXPERIMENTAL DESIGN: RB1 protein expression was classified by immunohistochemistry in ovarian carcinomas of 7,436 patients from the Ovarian Tumor Tissue Analysis consortium. We examined RB1 expression and germline BRCA status in a subset of 1,134 HGSC, and related genotype to overall survival (OS), tumor-infiltrating CD8+ lymphocytes, and transcriptomic subtypes. Using CRISPR-Cas9, we deleted RB1 in HGSC cells with and without BRCA1 alterations to model co-loss with treatment response. We performed whole-genome and transcriptome data analyses on 126 patients with primary HGSC to characterize tumors with concurrent BRCA deficiency and RB1 loss. RESULTS: RB1 loss was associated with longer OS in HGSC but with poorer prognosis in endometrioid ovarian carcinoma. Patients with HGSC harboring both RB1 loss and pathogenic germline BRCA variants had superior OS compared with patients with either alteration alone, and their median OS was three times longer than those without pathogenic BRCA variants and retained RB1 expression (9.3 vs. 3.1 years). Enhanced sensitivity to cisplatin and paclitaxel was seen in BRCA1-altered cells with RB1 knockout. Combined RB1 loss and BRCA deficiency correlated with transcriptional markers of enhanced IFN response, cell-cycle deregulation, and reduced epithelial-mesenchymal transition. CD8+ lymphocytes were most prevalent in BRCA-deficient HGSC with co-loss of RB1. CONCLUSIONS: Co-occurrence of RB1 loss and BRCA deficiency was associated with exceptionally long survival in patients with HGSC, potentially due to better treatment response and immune stimulation.


Subject(s)
BRCA1 Protein , BRCA2 Protein , Cystadenocarcinoma, Serous , Ovarian Neoplasms , Retinoblastoma Binding Proteins , Humans , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , BRCA2 Protein/genetics , BRCA2 Protein/deficiency , BRCA1 Protein/genetics , BRCA1 Protein/deficiency , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/immunology , Retinoblastoma Binding Proteins/genetics , Prognosis , Ubiquitin-Protein Ligases/genetics , Neoplasm Grading , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Middle Aged , Germ-Line Mutation , Gene Expression Regulation, Neoplastic , Aged , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism
8.
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
9.
Sci Adv ; 10(20): eadj5428, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38748789

ABSTRACT

High-grade serous ovarian cancer (HGSC) is a challenging disease, especially for patients with immunologically "cold" tumors devoid of tumor-infiltrating lymphocytes (TILs). We found that HGSC exhibits among the highest levels of MYCN expression and transcriptional signature across human cancers, which is strongly linked to diminished features of antitumor immunity. N-MYC repressed basal and induced IFN type I signaling in HGSC cell lines, leading to decreased chemokine expression and T cell chemoattraction. N-MYC inhibited the induction of IFN type I by suppressing tumor cell-intrinsic STING signaling via reduced STING oligomerization, and by blunting RIG-I-like receptor signaling through inhibition of MAVS aggregation and localization in the mitochondria. Single-cell RNA sequencing of human clinical HGSC samples revealed a strong negative association between cancer cell-intrinsic MYCN transcriptional program and type I IFN signaling. Thus, N-MYC inhibits tumor cell-intrinsic innate immune signaling in HGSC, making it a compelling target for immunotherapy of cold tumors.


Subject(s)
Immunity, Innate , N-Myc Proto-Oncogene Protein , Ovarian Neoplasms , Signal Transduction , Female , Humans , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Gene Expression Regulation, Neoplastic , Immunity, Innate/genetics , Interferon Type I/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Neoplasm Grading , Ovarian Neoplasms/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , N-Myc Proto-Oncogene Protein/genetics , N-Myc Proto-Oncogene Protein/metabolism
10.
Am J Pathol ; 194(7): 1272-1284, 2024 07.
Article in English | MEDLINE | ID: mdl-38537936

ABSTRACT

Tumor-infiltrating lymphocytes (TILs) are associated with improved survival in patients with epithelial ovarian cancer. However, TIL evaluation has not been used in routine clinical practice because of reproducibility issues. The current study developed two convolutional neural network models to detect TILs and to determine their spatial location in whole slide images, and established a spatial assessment pipeline to objectively quantify intraepithelial and stromal TILs in patients with high-grade serous ovarian carcinoma. The predictions of the established models showed a significant positive correlation with the number of CD8+ T cells and immune gene expressions. Patients with a higher density of intraepithelial TILs had a significantly prolonged overall survival and progression-free survival in multiple cohorts. On the basis of the density of intraepithelial and stromal TILs, patients were classified into three immunophenotypes: immune inflamed, excluded, and desert. The immune-desert subgroup showed the worst prognosis. Gene expression analysis showed that the immune-desert subgroup had lower immune cytolytic activity and T-cell-inflamed gene-expression profile scores, whereas the immune-excluded subgroup had higher expression of interferon-γ and programmed death 1 receptor signaling pathway. The established evaluation method provided detailed and comprehensive quantification of intraepithelial and stromal TILs throughout hematoxylin and eosin-stained slides. It has potential for clinical application for personalized treatment of patients with ovarian cancer.


Subject(s)
Cystadenocarcinoma, Serous , Deep Learning , Lymphocytes, Tumor-Infiltrating , Ovarian Neoplasms , Humans , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/genetics , Middle Aged , Aged , Prognosis , Stromal Cells/pathology , Stromal Cells/immunology , Stromal Cells/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism
11.
Gynecol Oncol ; 185: 83-94, 2024 06.
Article in English | MEDLINE | ID: mdl-38377762

ABSTRACT

OBJECTIVE: Advanced-stage high-grade serous ovarian cancer (HGSOC) remains a deadly gynecologic malignancy with high rates of disease recurrence and limited, effective therapeutic options for patients. There is a significant need to better stratify HGSOC patients into platinum refractory (PRF) vs. sensitive (PS) cohorts at baseline to improve therapeutic responses and survival outcomes for PRF HGSOC. METHODS: We performed NanoString for GeoMx Digital Spatial Profile (G-DSP) multiplex protein analysis on PRF and PS tissue microarrays (TMAs) to study the bidirectional communication of cancer cells with immune cells in the tumor microenvironment (TME) of HGSOC. We demonstrate robust stratification of PRF and PS tumors at baseline using multiplex spatial proteomic biomarkers with implications for tailoring subsequent therapy. RESULTS: PS patients had elevated apoptotic and anti-tumor immune profiles, while PRF patients had dual AKT1 and WNT signaling with immunosuppressive profiles. We found that dual activity of AKT1 and WNT signaling supported the exclusion of immune cells, specifically tumor infiltrating lymphocytes (TILs), from the TME in PRF tumors, and this was not observed in PS tumors. The exclusion of immune cells from the TME of PRF tumors corresponded to abnormal endothelial cell structure in tumors with dual AKT1 and WNT signaling activity. CONCLUSIONS: We believe our findings provide improved understanding of tumor-immune crosstalk in HGSOC TME highlighting the importance of the relationship between AKT and WNT pathways, immune cell function, and platinum response in HGSOC.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms , Proteomics , Proto-Oncogene Proteins c-akt , Tumor Microenvironment , Humans , Female , Tumor Microenvironment/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Proteomics/methods , Drug Resistance, Neoplasm/immunology , Middle Aged , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Wnt Signaling Pathway/immunology , Aged , Lymphocytes, Tumor-Infiltrating/immunology
12.
Gynecol Oncol ; 184: 74-82, 2024 05.
Article in English | MEDLINE | ID: mdl-38290413

ABSTRACT

OBJECTIVE: Ovarian cancer (OC) is the leading cause of death from gynecologic malignancy in the United States, and biomarkers of patient outcomes are limited. Data using immunohistochemical (IHC) analysis are mixed regarding whether and which tumor infiltrating lymphocytes (TILs) impact survival, and IHC does not adequately quantify rare cell populations, including CD137+ (4-1BB) tumor-reactive TILs. Our study investigates if a higher percentage of CD3+ CD137+ TILs is associated with improved overall survival (OS) in OC. METHODS: Flow cytometry was performed on viably banked OC digests. Chart review and statistical analysis were performed. Forty-seven patients were included, 40 of whom were diagnosed with high-grade serous ovarian carcinoma (HGSOC), papillary serous carcinoma, or undifferentiated histology. RESULTS: A high percentage of CD3+ CD137+ TILs correlated with improved OS (n = 40, r = 0.48, P = 0.0016). Subjects were divided into CD3+ CD137+ TIL high and low groups by the median. Subjects with high CD3+CD137+ TIL frequencies (>9.6%) had longer OS (Wilcoxon rank-sum test; P = 0.0032) and improved OS (logrank test; P = 0.007). Differences in CD3+ or CD3+ CD8+ TILs did not impact survival. CD3+ CD137+ TILs were predictive of OS regardless of germline mutation or debulking status. Analysis of subgroups including late stage HGSOC and late stage HGSOC with primary optimal cytoreduction indicated CD3+ CD137+ TILs correlated with improved OS after adjusting for age and PARP inhibitor use (P = 0.034 and P = 0.016, respectively). CONCLUSIONS: Prevalence of CD3+ CD137+ TILs in digested OC specimens is associated with improved OS, while general TIL markers are not. CD137 has the potential to be a novel biomarker for survival in OC.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Ovarian Neoplasms , Tumor Necrosis Factor Receptor Superfamily, Member 9 , Humans , Female , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Tumor Necrosis Factor Receptor Superfamily, Member 9/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/mortality , Middle Aged , Aged , CD3 Complex/analysis , Adult , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/mortality , Aged, 80 and over
13.
Nature ; 612(7941): 778-786, 2022 12.
Article in English | MEDLINE | ID: mdl-36517593

ABSTRACT

High-grade serous ovarian cancer (HGSOC) is an archetypal cancer of genomic instability1-4 patterned by distinct mutational processes5,6, tumour heterogeneity7-9 and intraperitoneal spread7,8,10. Immunotherapies have had limited efficacy in HGSOC11-13, highlighting an unmet need to assess how mutational processes and the anatomical sites of tumour foci determine the immunological states of the tumour microenvironment. Here we carried out an integrative analysis of whole-genome sequencing, single-cell RNA sequencing, digital histopathology and multiplexed immunofluorescence of 160 tumour sites from 42 treatment-naive patients with HGSOC. Homologous recombination-deficient HRD-Dup (BRCA1 mutant-like) and HRD-Del (BRCA2 mutant-like) tumours harboured inflammatory signalling and ongoing immunoediting, reflected in loss of HLA diversity and tumour infiltration with highly differentiated dysfunctional CD8+ T cells. By contrast, foldback-inversion-bearing tumours exhibited elevated immunosuppressive TGFß signalling and immune exclusion, with predominantly naive/stem-like and memory T cells. Phenotypic state associations were specific to anatomical sites, highlighting compositional, topological and functional differences between adnexal tumours and distal peritoneal foci. Our findings implicate anatomical sites and mutational processes as determinants of evolutionary phenotypic divergence and immune resistance mechanisms in HGSOC. Our study provides a multi-omic cellular phenotype data substrate from which to develop and interpret future personalized immunotherapeutic approaches and early detection research.


Subject(s)
Immune Evasion , Mutation , Ovarian Neoplasms , Female , Humans , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/pathology , Homologous Recombination , Immune Evasion/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Tumor Microenvironment , Transforming Growth Factor beta , Genes, BRCA1 , Genes, BRCA2
14.
Nat Commun ; 13(1): 835, 2022 02 11.
Article in English | MEDLINE | ID: mdl-35149709

ABSTRACT

The majority of high-grade serous ovarian cancers (HGSCs) are deficient in homologous recombination (HR) DNA repair, most commonly due to mutations or hypermethylation of the BRCA1/2 genes. We aimed to discover how BRCA1/2 mutations shape the cellular phenotypes and spatial interactions of the tumor microenvironment. Using a highly multiplex immunofluorescence and image analysis we generate spatial proteomic data for 21 markers in 124,623 single cells from 112 tumor cores originating from 31 tumors with BRCA1/2 mutation (BRCA1/2mut), and from 13 tumors without alterations in HR genes. We identify a phenotypically distinct tumor microenvironment in the BRCA1/2mut tumors with evidence of increased immunosurveillance. Importantly, we report a prognostic role of a proliferative tumor-cell subpopulation, which associates with enhanced spatial tumor-immune interactions by CD8+ and CD4 + T-cells in the BRCA1/2mut tumors. The single-cell spatial landscapes indicate distinct patterns of spatial immunosurveillance with the potential to improve immunotherapeutic strategies and patient stratification in HGSC.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Carcinoma, Ovarian Epithelial/immunology , Cystadenocarcinoma, Serous/immunology , Ovarian Neoplasms/immunology , Tumor Microenvironment/immunology , BRCA1 Protein/genetics , BRCA2 Protein/genetics , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Carcinoma, Ovarian Epithelial/genetics , Cystadenocarcinoma, Serous/genetics , Female , Genes, BRCA1 , Genes, BRCA2 , Genotype , Homologous Recombination , Humans , Mutation , Ovarian Neoplasms/genetics , Prognosis , Proteomics
15.
Sci Rep ; 12(1): 980, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35046435

ABSTRACT

Alternative splicing (AS) events play a crucial role in the tumorigenesis and progression of cancer. Transcriptome data and Percent Spliced In (PSI) values of ovarian cancer patients were downloaded from TCGA database and TCGA SpliceSeq. Totally we identified 1472 AS events that were associated with survival of ovarian serous cystadenocarcinoma (OC) and exon skipping (ES) was the most important type. Univariate and multivariate Cox regression analysis were performed to identify survival-associated AS events and developed the prognostic model based on 11-AS events. The immune cells and different response to cytotoxic T lymphocyte associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) blockers in low-risk and high-risk group of OC patients were analyzed. Ten kinds of immune cells were found up-regulated in low-risk group. Activated B cell, natural killer T cell, natural killer cell and regulatory T cell were associated with survival of OC. The patients in low-risk group had good response to CTLA-4 and PD-1 blockers treatment. Moreover, a regulatory network was established according to the correlation between AS events and splicing factors (SFs). The present study provided valuable insights into the underlying mechanisms of OC. AS events that were correlated with the immune system might be potential therapeutic targets.


Subject(s)
Alternative Splicing , Cystadenocarcinoma, Serous/immunology , Immune Checkpoint Inhibitors/therapeutic use , Ovarian Neoplasms/immunology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/therapy , Female , Humans , Middle Aged , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Proportional Hazards Models , Retrospective Studies
16.
Int J Mol Sci ; 22(20)2021 Oct 18.
Article in English | MEDLINE | ID: mdl-34681879

ABSTRACT

Ovarian cancers include several disease subtypes and patients often present with advanced metastatic disease and a poor prognosis. New biomarkers for early diagnosis and targeted therapy are, therefore, urgently required. This study uses antibodies produced locally in tumor-draining lymph nodes (ASC probes) of individual ovarian cancer patients to screen two separate protein microarray platforms and identify cognate tumor antigens. The resulting antigen profiles were unique for each individual cancer patient and were used to generate a 50-antigen custom microarray. Serum from a separate cohort of ovarian cancer patients encompassing four disease subtypes was screened on the custom array and we identified 28.8% of all ovarian cancers, with a higher sensitivity for mucinous (50.0%) and serous (40.0%) subtypes. Combining local and circulating antibodies with high-density protein microarrays can identify novel, patient-specific tumor-associated antigens that may have diagnostic, prognostic or therapeutic uses in ovarian cancer.


Subject(s)
Adenocarcinoma, Clear Cell/diagnosis , Adenocarcinoma, Mucinous/diagnosis , Antigens, Neoplasm/immunology , Autoantibodies/blood , Biomarkers, Tumor/blood , Cystadenocarcinoma, Serous/diagnosis , Ovarian Neoplasms/diagnosis , Adenocarcinoma, Clear Cell/blood , Adenocarcinoma, Clear Cell/immunology , Adenocarcinoma, Mucinous/blood , Adenocarcinoma, Mucinous/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/blood , Autoantibodies/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Case-Control Studies , Cohort Studies , Cystadenocarcinoma, Serous/blood , Cystadenocarcinoma, Serous/immunology , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Ovarian Neoplasms/blood , Ovarian Neoplasms/immunology , Prognosis , Protein Array Analysis , Young Adult
17.
Medicine (Baltimore) ; 100(40): e27473, 2021 Oct 08.
Article in English | MEDLINE | ID: mdl-34622876

ABSTRACT

BACKGROUND: FOXP4-AS1 expression participates in multiple signal pathways and has been previously reported in colorectal cancer, cervical cancer, and other cancer cells. However, its role on prognosis and immune infiltrates in ovarian serous cystadenocarcinoma (OVs) remains unclear. The purpose of our study was to investigate the expression of FOXP4-AS1 in OVs and its association with immune infiltrates, and determined its prognostic roles in OVs. METHODS: Using The Cancer Genome Atlas (TCGA) database, we retrieved FOXP4-AS1 expression and clinical information for 376 patients with OVs. Wilcoxon rank sum test was used to compare the expression of FOXP4-AS1 in OVs and normal ovarian tissue. Logistic regression was used to analyze the relationship between clinicopathologic features and FOXP4-AS1. Gene Set Enrichment Analysis (GSEA), and single sample Gene Set Enrichment Analysis (ssGSEA) was conducted to investigate the enrich pathways and functions and quantify the extent of immune cells infiltration for FOXP4-AS1. Kaplan-Meier method was used to generate survival curves, and Cox regression was used to analyze the relationship between FOXP4-AS1 and survival rate. RESULTS: High FOXP4-AS1 expression was significantly correlated with tumor FIGO stage (P = .026). Multivariate survival analysis showed that FOXP4-AS1was an independent prognostic marker for overall survival (OS; hazard ratio [HR]: 0.638; 95% confidence interval [CI]:0.467-0.871; P = .001) and disease-specific survival (DSS; HR: 0.649; CI: 0.476-0.885; P = .006). GSEA showed that High FOXP4-AS1 expression may active programmed cell death 1 (PD-1) signaling, the cytotoxic T lymphocyte-associated antigen-4 (CTLA4) pathway, the B cell receptor signaling pathway, apoptosis, fibroblast growth factor receptor (FGFR) signaling, and the Janus-activated kinase signal transducers and activators of transcription (JAK-STAT) signaling pathway. FOXP4-AS1 expression was negatively correlated with markers of immune cells, including aDC, cytotoxic cells and neutrophils. CONCLUSION: High FOXP4-AS1 expression has the potential to be a prognostic molecular marker of favorable survival in OVs.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , RNA, Long Noncoding/biosynthesis , Biomarkers, Tumor , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis
18.
Cancer Res ; 81(22): 5706-5719, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34561272

ABSTRACT

The tumor microenvironment evolves during malignant progression, with major changes in nonmalignant cells, cytokine networks, and the extracellular matrix (ECM). In this study, we aimed to understand how the ECM changes during neoplastic transformation of serous tubal intraepithelial carcinoma lesions (STIC) into high-grade serous ovarian cancers (HGSOC). Analysis of the mechanical properties of human fallopian tubes (FT) and ovaries revealed that normal FT and fimbria had a lower tissue modulus, a measure of stiffness, than normal or diseased ovaries. Proteomic analysis of the matrisome fraction between FT, fimbria, and ovaries showed significant differences in the ECM protein TGF beta induced (TGFBI, also known as ßig-h3). STIC lesions in the fimbria expressed high levels of TGFBI, which was predominantly produced by CD163-positive macrophages proximal to STIC epithelial cells. In vitro stimulation of macrophages with TGFß and IL4 induced secretion of TGFBI, whereas IFNγ/LPS downregulated macrophage TGFBI expression. Immortalized FT secretory epithelial cells carrying clinically relevant TP53 mutations stimulated macrophages to secrete TGFBI and upregulated integrin αvß3, a putative TGFBI receptor. Transcriptomic HGSOC datasets showed a significant correlation between TGFBI expression and alternatively activated macrophage signatures. Fibroblasts in HGSOC metastases expressed TGFBI and stimulated macrophage TGFBI production in vitro. Treatment of orthotopic mouse HGSOC tumors with an anti-TGFBI antibody reduced peritoneal tumor size, increased tumor monocytes, and activated ß3-expressing unconventional T cells. In conclusion, TGFBI may favor an immunosuppressive microenvironment in STICs that persists in advanced HGSOC. Furthermore, TGFBI may be an effector of the tumor-promoting actions of TGFß and a potential therapeutic target. SIGNIFICANCE: Analysis of ECM changes during neoplastic transformation reveals a role for TGFBI secreted by macrophages in immunosuppression in early ovarian cancer.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Extracellular Matrix/pathology , Macrophages/immunology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/pathology , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment , Animals , Apoptosis , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Cystadenocarcinoma, Serous/metabolism , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Female , Humans , Immunosuppressive Agents , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/metabolism , Prognosis , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
J Cancer Res Clin Oncol ; 147(12): 3545-3555, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34476576

ABSTRACT

PURPOSE: An in-depth analysis of the tumor microenvironment of ovarian cancer is needed. The purpose of this study was to elucidate the architecture of the immune microenvironment of high-grade serous ovarian cancers (HGSCs) with or without BRCA1 and BRCA2 mutations. METHODS: A cohort of highly annotated HGSC patients with known germline BRCA1 and BRCA2 status was selected, and pretreatment tumor tissue specimens were analyzed with a multiplexed staining technique aimed at detecting lymphocytes, macrophages, and fibroblasts in the whole tumor area and in specific regions including epithelium, stroma, and perivascular areas. RESULTS: BRCA1- or BRCA2-mutated tumors showed a more immunogenic microenvironment, characterized by a higher abundance of CD8+ and PD-L1+ cells, than did tumors with wild-type BRCA1 and BRCA2. High numbers of PD-L1+ and PD-L1+CD8+ cells were prognostic for event-free survival (hazard ratio [HR]: 0.41, 95% CI 0.21-0.79, p = 0.008 and HR 0.49, 95% CI 0.26-0.91, p = 0.025, respectively), as were high numbers of epithelial PD-L1+ and FAP+PD-L1+ cells (HR 0.52, 95% CI 0.28-0.96, p = 0.037 and HR 0.27, 95% CI 0.08-0.87, p = 0.029) and CD8+ cells (HR 0.51, 95% CI 0.28-0.93, p = 0.027). CONCLUSIONS: This study reveals substantial differences between the immune microenvironment composition of germline BRCA-mutated and BRCA wild-type HGSC.


Subject(s)
Cystadenocarcinoma, Serous/immunology , Ovarian Neoplasms/immunology , Tumor Microenvironment/immunology , Adult , Aged , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Female , Germ-Line Mutation , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL