Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.112
Filtrar
1.
J Ovarian Res ; 17(1): 208, 2024 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-39434163

RESUMEN

BACKGROUND: Immunotherapy has revolutionized the treatment of ovarian cancer (OC), but different immune microenvironments often constrain the efficacy of immunotherapeutic interventions. Therefore, there is an imperative to delineate novel immune subtypes for development of efficacious immunotherapeutic strategies. METHODS: The immune subtypes of OC were identified by consensus cluster analysis. The differences in clinical features, genetic mutations, mRNA stemness (mRNAsi) and immune microenvironments were analyzed among subtypes. Subsequently, prognostic risk models were constructed based on differentially expressed genes (DEGs) of the immune subtypes using weighted correlation network analysis. RESULTS: OC patients were classified into three immune subtypes with distinct survival rates and clinical features. Different subtypes exhibited varying tumor mutation burdens, homologous recombination deficiencies, and mRNAsi levels. Significant differences were observed among immune subtypes in terms of immune checkpoint expression and immunogenic cell death. Prognostic risk models were validated as independent prognostic factors demonstrated great predictive performance for survival of OC patients. CONCLUSION: In this study, three distinct immune subtypes were identified based on gene sets related to vaccine response, with the C2 subtype exhibiting significantly worse prognosis. While no statistically significant differences in tumor mutation burden (TMB) were observed across the three subtypes, the homologous recombination deficiency (HRD) score and mRNA stemness index (mRNAsi) were notably elevated in the C2 group compared to the others. Immune infiltration analysis indicated that the C2 subtype may have an increased presence of regulatory T (Treg) cells, potentially contributing to a more favorable response to combination therapies involving PARP inhibitors and immunotherapy. These findings offer a precision medicine approach for tailoring immunotherapy in ovarian cancer patients. Moreover, the C3 subtype demonstrated significantly lower expression levels of immune checkpoint genes, a pattern validated by independent datasets, and associated with a better prognosis. Further investigation revealed that the immune-related gene FCRL5 correlates with ovarian cancer prognosis, with in vitro experiments showing that it influences the proliferation and migration of the ovarian cancer cell line SKOV3.


Asunto(s)
Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Pronóstico , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Inmunoterapia/métodos , Mutación , Regulación Neoplásica de la Expresión Génica
2.
J Ovarian Res ; 17(1): 207, 2024 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-39427186

RESUMEN

The MECOM locus is a gene frequently amplified in high-grade serous ovarian carcinoma (HGSOC). Nevertheless, the body of research examining the associations among MECOM transcripts, patient prognosis, and their role in modulating the tumor immune microenvironment (TIME) remains sparse, particularly in large cohorts. This study assessed the expression of MECOM transcripts in 352 HGSOC patients and 88 normal ovarian tissues from the combined GTEx/TCGA database. Using resources such as the UCSC Genome Browser, Ensembl, and NextProt, two transcripts corresponding to classical protein isoforms from MECOM were identified. Cox proportional hazards regression analysis, Kaplan-Meier survival curves, and a comprehensive TIME evaluation algorithm were employed to elucidate the connections between the expression levels of these transcripts and both patient prognosis and TIME status. Chromatin Immunoprecipitation sequencing (ChIP-seq) data for the two protein isoforms, as well as RNA sequencing data post-targeted silencing, were analyzed to identify potential regulatory targets of the different transcription factors. Elevated expression of the MECOM isoform transcripts was correlated with poorer survival in HGSOC patients, potentially through the modulation of cancer-associated fibroblasts (CAFs) and immunosuppressive cell populations. In contrast, higher levels of EVI1 isoform transcripts were linked to enhanced survival, possibly due to the regulation of CD8+ T cells, macrophages, and a reduction in the expression of JUN protein, or its DNA-binding activity on downstream genes. Diverse protein isoforms derived from MECOM were found to differentially affect the survival and tumor development in ovarian cancer patients through specific mechanisms. Investigating the molecular mechanisms underlying disease pathogenesis and identifying potential drug target proteins at the level of splice variant isoforms were deemed crucial.


Asunto(s)
Neoplasias Ováricas , Isoformas de Proteínas , Microambiente Tumoral , Humanos , Femenino , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/mortalidad , Isoformas de Proteínas/genética , Pronóstico , Regulación Neoplásica de la Expresión Génica
3.
Cancer Discov ; 14(10): 1771-1773, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39363745

RESUMEN

Ameliorating the tumor immune microenvironment is a key strategy to improve the therapeutic outcomes of patients with cancer. Sandoval and colleagues demonstrate that iron chelation enhances type I IFN production, promotes NK cell tumor trafficking and activation, and synergizes with chemotherapy drug cisplatin to reduce metastatic ovarian cancer progression in murine models. See related article by Sandoval et al., p. 1901.


Asunto(s)
Células Asesinas Naturales , Neoplasias Ováricas , Microambiente Tumoral , Femenino , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Células Asesinas Naturales/inmunología , Humanos , Animales , Ratones , Microambiente Tumoral/inmunología
4.
Front Immunol ; 15: 1448041, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39376560

RESUMEN

Natural killer (NK) cell therapy represents an attractive immunotherapy approach against recurrent epithelial ovarian cancer (EOC), as EOC is sensitive to NK cell-mediated cytotoxicity. However, NK cell antitumor activity is dampened by suppressive factors in EOC patient ascites. Here, we integrated functional assays, soluble factor analysis, high-dimensional flow cytometry cellular component data and clinical parameters of advanced EOC patients to study the mechanisms of ascites-induced inhibition of NK cells. Using a suppression assay, we found that ascites from EOC patients strongly inhibits peripheral blood-derived NK cells and CD34+ progenitor-derived NK cells, albeit the latter were more resistant. Interestingly, we found that higher ascites-induced NK cell inhibition correlated with reduced progression-free and overall survival in EOC patients. Furthermore, we identified transforming growth factor (TGF)-ß1 to correlate with ascites-induced NK cell dysfunction and reduced patient survival. In functional assays, we showed that proliferation and anti-tumor reactivity of CD34+ progenitor-derived NK cells are significantly affected by TGF-ß1 exposure. Moreover, inhibition of TGF-ß1 signaling with galunisertib partly restored NK cell functionality in some donors. For the cellular components, we showed that the secretome is associated with a different composition of CD45+ cells between ascites of EOC and benign reference samples with higher proportions of macrophages in the EOC patient samples. Furthermore, we revealed that higher TGF-ß1 levels are associated with the presence of M2-like macrophages, B cell populations and T-regulatory cells in EOC patient ascites. These findings reveal that targeting TGF-ß1 signaling could increase NK cell immune responses in high-grade EOC patients.


Asunto(s)
Ascitis , Carcinoma Epitelial de Ovario , Células Asesinas Naturales , Neoplasias Ováricas , Factor de Crecimiento Transformador beta1 , Humanos , Femenino , Factor de Crecimiento Transformador beta1/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Carcinoma Epitelial de Ovario/inmunología , Carcinoma Epitelial de Ovario/mortalidad , Ascitis/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Persona de Mediana Edad , Clasificación del Tumor , Anciano , Pirazoles/uso terapéutico , Pirazoles/farmacología , Quinolinas
5.
J Ovarian Res ; 17(1): 198, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39394143

RESUMEN

Ovarian cancer (OC) remains the primary cause of mortality among gynecological malignancies, and the identification of reliable molecular biomarkers to prognosticate OC outcomes is yet to be achieved. The gene palmitoyl protein thioesterase 2 (PPT2), which has been sparsely studied in OC, was closely associated with metabolism. This study aimed to determine the association between PPT2 expression, prognosis, immune infiltration, and potential molecular mechanisms in OC. We obtained the RNA-seq and clinical data from The Cancer Genome Atlas (TCGA), The Genotype-Tissue Expression (GTEx) and Gene Expression Omnibus (GEO) databases, then Kaplan-Meier analysis, univariate Cox regression, multivariate Cox regression, nomogram, and calibration were conducted to assess and verify the role of PPT2. Gene set enrichment analysis (GSEA) was used to figure out the closely correlated pathways with PPT2. Overexpression experiment was performed to explore the function of PPT2. Our findings showed that PPT2 mRNA expression was apparent down-regulation in OC tissue compared to normal ovarian tissues in TCGA, GTEx datasets, and GEO datasets. This differential expression was also confirmed in our in-house datasets at both the mRNA and protein levels. Decreased PPT2 expression correlated with lower survival rates in TCGA, several GEO datasets, and our in-house datasets. Multivariate analysis revealed that PPT2 was an independent factor in predicting better outcomes for OC patients in TCGA and GEO. A negative correlation was revealed between immune infiltration and PPT2 expression through Single-sample GSEA (ssGSEA). Additionally, PPT2 was negatively correlated with an up-regulated immune score, stromal score, and estimate score, suggesting that patients with low PPT2 expression might benefit more from immunotherapy. Numerous chemical agents showed lower IC50 in patients with high PPT2 expression. In single-cell RNA sequencing (scRNA-seq) analysis of several OC datasets, we found PPT2 was mainly expressed in endothelial cells. Furthermore, we found that PPT2 inhibited OC cell proliferation in vitro. Our results demonstrated that PPT2 was considered a favorable prognostic biomarker for OC and may be vital in predicting response to immunotherapy and chemotherapy. Further research was needed to fully understand the relationship between PPT2 and immunotherapy efficacy in OC patients.


Asunto(s)
Inmunoterapia , Neoplasias Ováricas , Tioléster Hidrolasas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/terapia , Pronóstico , Inmunoterapia/métodos , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica
6.
J Ovarian Res ; 17(1): 197, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39385288

RESUMEN

BACKGROUND: Fibroblast growth factors (FGFs) are cell signaling proteins that perform multiple biological processes in many biological processes (cell development, repair, and metabolism). The dynamics of tumor cells, such as angiogenesis, transformation, and proliferation, have a significant impact on neoplasia and are modulated by FGFs. FGFs' expression and prognostic significance in ovarian cancer (OC), however, remain unclear. METHODS: Through a series of in silico analysis, we investigated the transcriptional, survival data, genetic variation, gene-gene interaction network, ferroptosis-related genes, and DNA methylation of FGFs in OC patients. RESULTS: We discovered that while FGF18 expression levels were higher in OC tissues than in normal OC tissues, FGF2/7/10/17/22 expression levels were lower in the former, and that FGF1/19 expression was related to the tumor stage in OC patients. According to the survival analysis, the clinical prognosis of individuals with OC was associated with the aberrant expression of FGFs. The function of FGFs and their neighboring genes was mainly connected to the cellular response to FGF stimulus. There was a negative correlation between FGF expression and various immune cell infiltration. CONCLUSIONS: This study clarifies the relationship between FGFs and OC, which might provide new insights into the choice of prognostic biomarkers of OC patients.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Neoplasias Ováricas , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/mortalidad , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Pronóstico , Simulación por Computador , Regulación Neoplásica de la Expresión Génica , Metilación de ADN
7.
Front Immunol ; 15: 1465917, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39421747

RESUMEN

Background: This study conducts a bibliometric literature analysis to explore trends in immunotherapy for ovarian cancer from 2019 to 2023. Methods: An extensive online literature search was conducted in the Web of Science Core Collection database to identify English-language articles and reviews related to "trends in immunotherapy", and "ovarian cancer". statistical analysis was performed using VOSviewer to visualize and compare nations, institutions, and journals simultaneously. Results: Our findings highlight contributions by 118 nations, led by the People's Republic of China with 3,167 contributions; Germany followed with 558 and Italy having 547. Of all publications made between 2019-2023, "Frontiers Immunology" had the most publications with 546 total records followed by "cancers ", and "frontiers in oncology" being the most heavily relied upon categories. Annually publication trends increased until 2022 but then declined considerably as a peak of highly-cited papers occurring between 2019 and 2022. Conclusions: Our bibliometric analysis not only maps the evolution of immunotherapy research in ovarian cancer but also provides actionable insights for advancing scientific progress. By identifying emerging trends and key areas, future research can strategically enhance treatment strategies and outcomes for ovarian cancer patients.


Asunto(s)
Bibliometría , Inmunoterapia , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/terapia , Neoplasias Ováricas/inmunología , Inmunoterapia/tendencias , Inmunoterapia/métodos
8.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(10): 909-917, 2024 Oct.
Artículo en Chino | MEDLINE | ID: mdl-39442989

RESUMEN

Objective To screen for the key genes involved in the development of ovarian cancer (OV), analyze the immune cell infiltration and construct a risk model, so as to provide evidence for the early diagnosis, treatment and prognosis evaluation of OV patients. Methods The GSE18520 and GSE6008 datasets were analyzed for differentially expressed genes (DEGs) using the GEO2R data analysis tool, and a Venn diagram was generated. Then, DEGs were subjected to Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, and protein-protein interaction (PPI) networks, as well as mutations, expression and prognosis analysis to identify key genes. Next, a risk model was constructed and immune cell infiltration analysis of key genes was performed. Finally, ovarian cancer tissues were collected as the experimental group, and adjacent normal tissues were collected as the control group. The expression of claudin 4 (CLDN4) mRNA and protein levels were detected using real-time quantitative PCR (RT-qPCR) and Western-blot, and the results were compared between the two groups. Results CLDN4 was identified as a key gene in the development of OV. As its expression increased, the prognosis risk of OV patients worsened, which unfavorably impacted their overall survival (OS). A significant positive correlation was found between CLDN4 and dendritic cell (DC) in the OV microenvironment, and high expression of DCs was significantly associated with better OS in OV patients. The mRNA and protein expression levels of CLDN4 were significantly increased in OV tissues, with statistically significant differences. Conclusion CLDN4 is a key gene in the development of OV, and may serve as a potential biomarker and immunotherapy target for OV.


Asunto(s)
Claudina-4 , Neoplasias Ováricas , Humanos , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Claudina-4/genética , Pronóstico , Regulación Neoplásica de la Expresión Génica , Mapas de Interacción de Proteínas/genética
9.
Nat Commun ; 15(1): 9050, 2024 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-39426954

RESUMEN

Iron is an essential biomineral in the human body. Here, we describe a subset of iron-loaded cancer-associated fibroblasts, termed as FerroCAFs, that utilize iron to induce immunosuppression in prostate cancer and predict an unfavorable clinical outcome. FerroCAFs secrete myeloid cell-associated proteins, including CCL2, CSF1 and CXCL1, to recruit immunosuppressive myeloid cells. We report the presence of FerroCAFs in prostate cancer from both mice and human, as well as in human lung and ovarian cancers, and identify a conserved cell surface marker, the poliovirus receptor. Mechanistically, the accumulated iron in FerroCAFs is caused by Hmox1-mediated iron release from heme degradation. The intracellular iron activates the Kdm6b, an iron-dependent epigenetic enzyme, to induce an accessible chromatin state and transcription of myeloid cell-associated protein genes. Targeting the FerroCAFs by inhibiting the Hmox1/iron/Kdm6b signaling axis incurs anti-tumor immunity and tumor suppression. Collectively, we report an iron-loaded FerroCAF cluster that drives immunosuppression through an iron-dependent epigenetic reprogramming mechanism and reveal promising therapeutic targets to boost anti-tumor immunity.


Asunto(s)
Fibroblastos Asociados al Cáncer , Hemo-Oxigenasa 1 , Hierro , Neoplasias de la Próstata , Humanos , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/genética , Masculino , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/inmunología , Hierro/metabolismo , Animales , Ratones , Hemo-Oxigenasa 1/metabolismo , Hemo-Oxigenasa 1/genética , Femenino , Línea Celular Tumoral , Histona Demetilasas/metabolismo , Histona Demetilasas/genética , Epigénesis Genética , Receptores Virales/metabolismo , Receptores Virales/genética , Hemo/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal , Células Mieloides/metabolismo , Células Mieloides/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Tolerancia Inmunológica , Terapia de Inmunosupresión
10.
J Immunother Cancer ; 12(10)2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39366751

RESUMEN

BACKGROUND: In addition to their established action of synthetic lethality in tumor cells, poly(ADP-ribose) polymerase inhibitors (PARPis) also orchestrate tumor immune microenvironment (TIME) that contributes to suppressing tumor growth. However, it remains not fully understood whether and how PARPis trigger tumor-targeting immune responses. METHODS: To decode the immune responses reshaped by PARPis, we conducted T-cell receptor (TCR) sequencing and immunohistochemical (IHC) analyses of paired clinical specimens before and after niraparib monotherapy obtained from a prospective study, as well as ID8 mouse ovarian tumors. To validate the induction of immunogenic cell death (ICD) by PARPis, we performed immunofluorescence/IHC staining with homologous recombination deficiency tumor cells and patient-derived xenograft tumor tissues, respectively. To substantiate that PARPis elicited tumor cell pyroptosis, we undertook comprehensive assessments of the cellular morphological features, cleavage of gasdermin (GSDM) proteins, and activation of TNF-caspase signaling pathways through genetic downregulation/depletion and selective inhibition. We also evaluated the critical role of pyroptosis in tumor suppression and immune activation following niraparib treatment using a syngeneic mouse model with implanting CRISPR/Cas9 edited Gsdme-/ - ID8 tumor cells into C57BL/6 mice. RESULTS: Our findings revealed that PARPis augmented the proportion of neoantigen-recognized TCR clones and TCR clonal expansion, and induced an inflamed TIME characterized by increased infiltration of both innate and adaptive immune cells. This PARPis-strengthened immune response was associated with the induction of ICD, specifically identified as pyroptosis, which possessed distinctive morphological features and GSDMD/E cleavage. It was validated that the cleavage of GSDMD/E was due to elevated caspase 8 activity downstream of the TNFR1, rather than FAS and TRAIL-R. On PARP inhibition, the NF-κB signaling pathway was activated, leading to increased secretion of TNF-α and subsequent initiation of the TNFR1-caspase 8 cascade. Impeding pyroptosis through the depletion of Gsdme significantly compromised the tumor-suppressing effects of PARP inhibition and undermined the anti-immune response in the syngeneic ID8 mouse model. CONCLUSIONS: PARPis induce a specific type of ICD called pyroptosis via TNF-caspase 8-GSDMD/E axis, resulting in an inflamed TIME and augmentation of tumor-targeting immune responses. These findings deepen our understanding of PARPis activities and point toward a promising avenue for synergizing PARPis with immunotherapeutic interventions. TRIAL REGISTRATION NUMBER: NCT04507841.


Asunto(s)
Neoplasias Ováricas , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Piroptosis , Animales , Femenino , Humanos , Ratones , Línea Celular Tumoral , Gasderminas , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas de Unión a Fosfato/metabolismo , Piperidinas/farmacología , Piperidinas/uso terapéutico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Piroptosis/efectos de los fármacos , Transducción de Señal , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
11.
J Immunother Cancer ; 12(10)2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39401967

RESUMEN

BACKGROUND: Ovarian cancer remains a formidable challenge in oncology, necessitating innovative therapeutic approaches. Claudin-6 (CLDN6), a member of the tight junction molecule CLDN family, exhibits negligible expression in healthy tissues but displays aberrant upregulation in various malignancies, including ovarian cancer. Although several therapeutic modalities targeting CLDN6 are currently under investigation, there is still a need for more potent therapeutic options. While T-cell engagers (TCEs) hold substantial promise as potent immunotherapeutic agents, their current efficacy and safety in terms of target antigen selection and T-cell exhaustion due to only CD3 stimulation without co-stimulation must be improved, particularly against solid tumors. To provide an efficacious treatment option for ovarian cancer, we generated SAIL66, a tri-specific antibody against CLDN6/CD3/CD137. METHODS: Using our proprietary next-generation TCE technology (Dual-Ig), SAIL66 was designed to bind to CLDN6 with one Fab and CD3/CD137 with the other, thereby activating T cells through CD3 activation and CD137 co-stimulation. The preclinical characterization of SAIL66 was performed in a series of in vitro and in vivo studies which included comparisons to a conventional TCE targeting CLDN6 and CD3. RESULTS: Despite the high similarity between CLDN6 and other CLDN family members, SAIL66 demonstrated high specificity for CLDN6, reducing the risk of off-target toxicity. In an in vitro co-culture assay with CLDN6-positive cancer cells, we confirmed that SAIL66 strongly activated the CD137 signal in the Jurkat reporter system, and preferentially induced activation of both CD4+ and CD8+ T cells isolated from human peripheral blood mononuclear cells compared to conventional TCEs. In vivo studies demonstrated that SAIL66 led to a more pronounced increase in intratumor T-cell infiltration and a decrease in exhausted T cells compared with conventional CLDN6 TCE by contribution of CD137 co-stimulation, resulting in better antitumor efficacy in tumor-bearing mouse models. CONCLUSION: Our data demonstrate that SAIL66, designed to engage CLDN6, CD3, and CD137, has the potential to enhance antitumor activity and provide a potent therapeutic option for patients with ovarian and other solid tumors expressing CLDN6. Clinical trials are currently underway to evaluate the safety and efficacy of SAIL66.


Asunto(s)
Claudinas , Humanos , Animales , Ratones , Femenino , Claudinas/metabolismo , Complejo CD3/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Línea Celular Tumoral
12.
J Ovarian Res ; 17(1): 202, 2024 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-39402681

RESUMEN

Ovarian cancer, often referred to as the "silent killer," is notoriously difficult to detect in its early stages, leading to a poor prognosis for many patients. Diagnosis is often delayed until the cancer has advanced, primarily due to its ambiguous and frequently occurring clinical symptoms. Ovarian cancer leads to more deaths than any other cancer of the female reproductive system. The main reasons for the high mortality rates include delayed diagnosis and resistance to treatment. As a result, there is an urgent need for improved diagnostic and treatment options for ovarian cancer. The standard treatments typically involve debulking surgery along with platinum-based chemotherapies. Among patients with advanced-stage cancer who initially respond to current therapies, 50-75% experience a recurrence. Recently, immunotherapy-based approaches to enhance the body's immune response to combat tumor growth have shown promise. Immune checkpoint inhibitors have shown promising results in treating other types of tumors. However, in ovarian cancer, only a few of these inhibitors have been effective because the tumor's environment suppresses the immune system and creates barriers for treatment. This hampers the effectiveness of existing immunotherapies. Nonetheless, advanced immunotherapy techniques and delivery systems based on nanotechnology hold promise for overcoming these challenges.


Asunto(s)
Inmunoterapia , Nanotecnología , Neoplasias Ováricas , Humanos , Femenino , Inmunoterapia/métodos , Neoplasias Ováricas/terapia , Neoplasias Ováricas/inmunología , Nanotecnología/métodos
13.
Oncoimmunology ; 13(1): 2411070, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39364290

RESUMEN

High-grade serious ovarian cancer (HGSOC) is an aggressive malignancy that remains refractory to current immunotherapies. While advanced stage disease has been extensively studied, the cellular and molecular mechanisms that promote early immune escape in HGSOC remain largely unexplored. Here, we report that primary HGSO tumors program neutrophils to inhibit T cell anti-tumor function by activating the endoplasmic reticulum (ER) stress sensor IRE1α. We found that intratumoral neutrophils exhibited overactivation of ER stress response markers compared with their counterparts at non-tumor sites. Selective deletion of IRE1α in neutrophils delayed primary ovarian tumor growth and extended the survival of mice with HGSOC by enabling early T cell-mediated tumor control. Notably, loss of IRE1α in neutrophils sensitized tumor-bearing mice to PD-1 blockade, inducing HGSOC regression and long-term survival in ~ 50% of the treated hosts. Hence, neutrophil-intrinsic IRE1α facilitates early adaptive immune escape in HGSOC and targeting this ER stress sensor might be used to unleash endogenous and immunotherapy-elicited immunity that controls metastatic disease.


Asunto(s)
Estrés del Retículo Endoplásmico , Endorribonucleasas , Neutrófilos , Neoplasias Ováricas , Receptor de Muerte Celular Programada 1 , Proteínas Serina-Treonina Quinasas , Femenino , Animales , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Ratones , Humanos , Estrés del Retículo Endoplásmico/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Resistencia a Antineoplásicos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Cistadenocarcinoma Seroso/patología , Cistadenocarcinoma Seroso/inmunología , Cistadenocarcinoma Seroso/metabolismo , Línea Celular Tumoral , Clasificación del Tumor , Escape del Tumor/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados
14.
Front Immunol ; 15: 1452946, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39355254

RESUMEN

Background: Ovarian cancer (OC) is a global malignancy characterized by metastatic invasiveness and recurrence. Long non-coding RNAs (lncRNAs) and Telomeres are closely connected with several cancers, but their potential as practical prognostic markers in OC is less well-defined. Methods: Relevant mRNA and clinical data for OC were sourced from The Cancer Genome Atlas (TCGA) database. The telomere-related lncRNAs (TRLs) prognostic model was established by univariate/LASSO/multivariate regression analyses. The effectiveness of the TRLs model was evaluated and measured via the nomogram. Additionally, immune infiltration, tumor mutational load (TMB), and drug sensitivity were evaluated. We validated the expression levels of prognostic genes. Subsequently, PTPRD-AS1 knockdown was utilized to perform the CCK8 assay, colony formation assay, transwell assay, and wound healing assay of CAOV3 cells. Results: A six-TRLs prognostic model (PTPRD-AS1, SPAG5-AS1, CHRM3-AS2, AC074286.1, FAM27E3, and AC018647.3) was established, which can effectively predict patient survival rates and was successfully validated using external datasets. According to the nomogram, the model could effectively predict prognosis. Furthermore, we detected the levels of regulatory T cells and M2 macrophages were comparatively higher in the high-risk TRLs group, but the levels of activated CD8 T cells and monocytes were the opposite. Finally, the low-risk group was more sensitive to anti-cancer drugs. The mRNA levels of PTPRD-AS1, SPAG5-AS1, FAM27E3, and AC018647.3 were significantly over-expressed in OC cell lines (SKOV3, A2780, CAOV3) in comparison to normal IOSE-80 cells. AC074286.1 were over-expressed in A2780 and CAOV3 cells and CHRM3-AS2 only in A2780 cells. PTPRD-AS1 knockdown decreased the proliferation, cloning, and migration of CAOV3 cells. Conclusion: Our study identified potential biomarkers for the six-TRLs model related to the prognosis of OC.


Asunto(s)
Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas , ARN Largo no Codificante , Telómero , Humanos , ARN Largo no Codificante/genética , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/mortalidad , Pronóstico , Biomarcadores de Tumor/genética , Telómero/genética , Línea Celular Tumoral , Nomogramas , Persona de Mediana Edad , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo
15.
Semin Immunopathol ; 46(6): 16, 2024 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-39432076

RESUMEN

Ovarian cancer remains a formidable challenge in oncology due to its late-stage diagnosis and limited treatment options. Recent research has revealed the intricate interplay between glycan diversity and the immune microenvironment within ovarian tumors, shedding new light on potential therapeutic strategies. This review seeks to investigate the complex role of glycans in ovarian cancer and their impact on the immune response. Glycans, complex sugar molecules decorating cell surfaces and secreted proteins, have emerged as key regulators of immune surveillance in ovarian cancer. Aberrant glycosylation patterns can promote immune evasion by shielding tumor cells from immune recognition, enabling disease progression. Conversely, certain glycan structures can modulate the immune response, leading to either antitumor immunity or immune tolerance. Understanding the intricate relationship between glycan diversity and immune interactions in ovarian cancer holds promise for the development of innovative therapeutic approaches. Immunotherapies that target glycan-mediated immune evasion, such as glycan-based vaccines or checkpoint inhibitors, are under investigation. Additionally, glycan profiling may serve as a diagnostic tool for patient stratification and treatment selection. This review underscores the emerging importance of glycan diversity in ovarian cancer, emphasizing the potential for unraveling immune interplay and advancing tailored therapeutic prospects for this devastating disease.


Asunto(s)
Neoplasias Ováricas , Polisacáridos , Microambiente Tumoral , Humanos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/terapia , Neoplasias Ováricas/etiología , Femenino , Polisacáridos/metabolismo , Polisacáridos/inmunología , Microambiente Tumoral/inmunología , Glicosilación , Animales , Inmunoterapia/métodos , Escape del Tumor , Susceptibilidad a Enfermedades
16.
Theranostics ; 14(16): 6249-6267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39431011

RESUMEN

Rationale: CD39, a key ectonucleotidase that drives adenosine production, acts as a critical immunosuppressive checkpoint in cancer. Although it has shown promise as a therapeutic target, clinical trials are demonstrating the need for more potent targeting approaches. This need is driving innovation towards the development of novel antibodies and the exploration of strategic combinations with a range of immunotherapies. Methods: An anti-CD39 nanobody was screened and tested for its affinity and binding ability using biolayer interferometry, ELISA and flow cytometry. Blocking ability against soluble and membrane-bound CD39 was measured after CD39 blockade. Internalization was detected using immunofluorescence. The reversal of T-cell function by the anti-CD39 antibody was assessed by CFSE-based T-cell proliferation, CD25 expression and IFN-γ secretion. The in vivo function of tumor growth inhibition was further tested in a mouse model and we also tested the phenotype of immune cells after CD39 antibody administration from tumor tissue, draining lymph nodes and peripheral blood. We inserted the antibody sequence into the chimeric antigen receptor (CAR) construct to induce MSLN CAR-T cells to secret the CD39 antibody, and the efficacy was measured in xenograft models of ovarian cancer. Results: We screened human CD39 antibodies using a VHH library and developed a single-epitope anti-CD39 nanobody, named huCD39 mAb, with high affinity and potent binding and blocking ability. The huCD39 mAb was internalized in a time-dependent manner. The in vitro study revealed that the huCD39 mAb was highly effective in enhancing T-cell proliferation and functionality. In vivo, the huCD39 mAb showed significant anti-tumor efficacy in an immunocompetent mouse model. Flow cytometry analysis demonstrated downregulated CD39 expression in immune cells after antibody administration. We also observed increased CD39 expression in ovarian cancer tissue and in activated CAR T cells. Subsequently, we developed a type of MSLN CAR-T cells secreting huCD39 mAb which showed effective eradication or inhibition in ovarian tumor xenografts. Conclusions: A novel huCD39 mAb with strong blocking ability against human CD39 and potent inhibition of tumor growth has been developed. Furthermore, a modified huCD39 mAb-secreting CAR-T cell has been generated, exhibiting superior efficacy against ovarian cancer. This provides a promising strategy for optimizing immunotherapies in ovarian cancer and potentially other malignancies.


Asunto(s)
Apirasa , Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Anticuerpos de Dominio Único , Linfocitos T , Animales , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Femenino , Apirasa/inmunología , Apirasa/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Anticuerpos de Dominio Único/inmunología , Ratones , Humanos , Línea Celular Tumoral , Linfocitos T/inmunología , Linfocitos T/metabolismo , Inmunoterapia Adoptiva/métodos , Mesotelina , Ensayos Antitumor por Modelo de Xenoinjerto , Antígenos CD/inmunología , Antígenos CD/metabolismo , Proliferación Celular
17.
Sci Rep ; 14(1): 24191, 2024 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-39406772

RESUMEN

Ovarian germ cell tumors (OVGCTs) account for 28% of all diagnosed ovarian cancers, and malignant germ cell tumors specifically account for approximately 13% of diagnosed ovarian cancers in Saudi Arabia. Although most germ cell tumor patients have a high survival rate, patients who experience tumor recurrence have a poor prognosis and present with more aggressive and chemoresistant tumors. The use of immunotherapeutic agents such as PD-L1/PD-1 inhibitors for OVGCTs remains very limited because few studies have described the immunological characteristics of these tumors. This study is the first to investigate PD-L1 expression in ovarian germ cell tumors and explore the role of PD-L1 expression in tumor microenvironment cells and genetic alterations. A total of 34 ovarian germ cell tumors were collected from pathology archives. The collected tumor tissues included ten dysgerminomas, five yolk sac tumors, five immature teratomas, and one mature teratoma, and the remaining samples were mixed germ cell tumors. The tumors were analyzed using immunohistochemical analysis to determine PD-L1 expression, immune cell infiltration and cancer stem cell populations and their correlation with clinical outcome. Furthermore, the genetic alterations in different subtypes of germ cell tumors were correlated with PD-L1 expression and clinical outcome. Datasets for testicular germ cells (TGCTs) were retrieved from The Cancer Genome Atlas (TCGA) and analyzed using cBioPortal (cbioportal.org) and Gene Expression Profiling Interactive Analysis (GEPIA). Compared with yolk sac tumors, dysgerminomas highly express PD-L1 and are associated with high levels of tumor infiltrating lymphocytes (TILs) and stem cell markers. In addition, compared with PD-L1-negative yolk sac tissue, dysgerminomas/seminomas with high PD-L1 expression are associated with more genetic alterations and a better prognosis. Our findings will contribute to the knowledge about the potential benefits of ovarian cancer immunotherapy in specific subsets of germ cell tumor patients and the risk factors for resistance mediated by tumor microenvironment cells.


Asunto(s)
Antígeno B7-H1 , Linfocitos Infiltrantes de Tumor , Neoplasias de Células Germinales y Embrionarias , Neoplasias Ováricas , Microambiente Tumoral , Humanos , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Neoplasias de Células Germinales y Embrionarias/genética , Neoplasias de Células Germinales y Embrionarias/metabolismo , Neoplasias de Células Germinales y Embrionarias/inmunología , Neoplasias de Células Germinales y Embrionarias/patología , Femenino , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Neoplasias Ováricas/metabolismo , Pronóstico , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Microambiente Tumoral/inmunología , Adulto , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Adulto Joven , Adolescente
18.
Front Immunol ; 15: 1458264, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39478854

RESUMEN

Background: The use of gene development data from public database has become a new starting point to explore mitochondrial related gene expression and construct a prognostic prediction model of ovarian cancer. Methods: Data were obtained from the TCGA and ICGC databases, and the intersection with mitochondrial genes was used to obtain the differentially expressed genes. q-PCR, Cox proportional risk regression, minimal absolute contraction and selection operator regression analysis were performed to construct the prognostic risk model, and ROC curve was used to evaluate the model for centralized verification. The association between risk scores and clinical features, tumor mutation load, immune cell infiltration, macrophage activation analysis, immunotherapy, and chemosensitivity was further evaluated. Results: A prognostic risk score model for ovarian cancer patients was constructed based on 12 differentially expressed genes. The score was highly correlated with ovarian cancer macrophage infiltration and was a good predictor of the response to immunotherapy. M1 and M2 macrophages in the ovarian tissue in the OV group were significantly activated, providing a reference for the study of the polarity change of tumor-related macrophages for the prognosis and treatment of ovarian cancer. In terms of drug sensitivity, the high-risk group was more sensitive to vinblastine, Acetalax, VX-11e, and PD-0325901, while the low-risk group was more sensitive to Sabutoclax, SB-505124, cisplatin, and erlotinib. Conclusion: The prognostic risk model of ovarian cancer associated to mitochondrial genes built on the basis of public database better evaluated the prognosis of ovarian cancer patients and guided individual treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Neoplasias Ováricas/mortalidad , Pronóstico , Biomarcadores de Tumor/genética , Mitocondrias/metabolismo , Mitocondrias/genética , Bases de Datos Genéticas , Perfilación de la Expresión Génica , Genes Mitocondriales , Persona de Mediana Edad
19.
Medicine (Baltimore) ; 103(43): e40272, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39470479

RESUMEN

Ovarian cancer (OC) has a limited immunotherapeutic response; hence, this study aimed to investigate the relationship between CXC-chemokine ligand 13 (CXCL13) expression and overall survival (OS) rate, key immune pathways, degree of immune cell infiltration, and progressive disease (PD)-1 checkpoint blockade. A total of 703 differentially expressed genes were obtained from "The Cancer Genome Atlas" (TCGA) database based on the immune and stromal scores of 379 OC patients for getting the targeted gene CXCL13. The association between CXCL13 and OS in OC patients, biological function annotation of CXCL13, and its correlation with immune components were assessed. The results indicated that upregulated CXCL13 expression was positively correlated with better OC patient prognosis. CXCL13 expression was associated with 6 immune-related pathways, 10 immune cells, and PD-1 expression of OC micro-environment. Moreover, high expression of CXCL13 was related to a better tumor response and more extended tumor-stable stage after PD-1 blocking therapy in IMvigor210. The study concluded that CXCL13 could be a prognostic marker and a potential immunotherapy target for OC patients, especially PD-1 checkpoint blockade.


Asunto(s)
Quimiocina CXCL13 , Neoplasias Ováricas , Humanos , Quimiocina CXCL13/metabolismo , Quimiocina CXCL13/genética , Femenino , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/genética , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/metabolismo , Pronóstico , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Microambiente Tumoral/inmunología , Persona de Mediana Edad , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Regulación Neoplásica de la Expresión Génica
20.
Sci Rep ; 14(1): 25239, 2024 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-39448713

RESUMEN

Epithelial ovarian carcinoma (EOC) is the most fatal among female reproductive system tumors. The immune tumor microenvironment and ubiquitin-proteasome pathway are closely related to the proliferation, invasion, and response to chemotherapy in EOC. However, their specific roles in EOC have not been fully elucidated. Therefore, we aimed to recognize potential prognostic markers and novel therapeutic targets for EOC. We constructed the ubiquitin-related signature risk model comprising HSP90AB1, FBXO9, SIGMAR1, STAT1, SH3KBP1, EPB41L2, DNAJB6, VPS18, PPM1G, AKAP12, FRK, and PYGB, specifically for patients with EOC. The high-risk model presented a worse prognosis, primarily associated with the B-cell receptor signaling pathway, ECM receptor interaction, focal adhesion, and actin cytoskeleton regulations. Analysis of the immune landscape revealed a higher abundance of B cells, M2 macrophages, neutrophil CD4 T cells, cancer-associated fibroblasts, macrophage neutrophils, and fibroblasts in the high-risk group. It also exhibited lower tumor mutation burden, mRNAsi, and EREG-mRNAsi and reduced sensitivity to other chemotherapy drugs, except dasatinib. These findings serve as a valuable indicator for personalized treatment strategies and clinical stratification in managing patients with EOC. Additionally, our study will serve as a foundation for future mechanistic research to explore the association between the ubiquitin-proteasome pathway and EOC.


Asunto(s)
Biomarcadores de Tumor , Carcinoma Epitelial de Ovario , Inmunoterapia , Neoplasias Ováricas , Ubiquitina , Humanos , Femenino , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/inmunología , Pronóstico , Biomarcadores de Tumor/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Neoplasias Ováricas/terapia , Inmunoterapia/métodos , Ubiquitina/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Regulación Neoplásica de la Expresión Génica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...