Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(41): e2203480119, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36197994

RESUMEN

Fatty acids are an important source of energy and a key component of phospholipids in membranes and organelles. Saturated fatty acids (SFAs) are converted into unsaturated fatty acids (UFAs) by stearoyl Co-A desaturase (SCD), an enzyme active in cancer. Here, we studied how the dynamics between SFAs and UFAs regulated by SCD impacts ovarian cancer cell survival and tumor progression. SCD depletion or inhibition caused lower levels of UFAs vs. SFAs and altered fatty acyl chain plasticity, as demonstrated by lipidomics and stimulated Raman scattering (SRS) microscopy. Further, increased levels of SFAs resulting from SCD knockdown triggered endoplasmic reticulum (ER) stress response with brisk activation of IRE1α/XBP1 and PERK/eIF2α/ATF4 axes. Disorganized ER membrane was visualized by electron microscopy and SRS imaging in ovarian cancer cells in which SCD was knocked down. The induction of long-term mild ER stress or short-time severe ER stress by the increased levels of SFAs and loss of UFAs led to cell death. However, ER stress and apoptosis could be readily rescued by supplementation with UFAs and reequilibration of SFA/UFA levels. The effects of SCD knockdown or inhibition observed in vitro translated into suppression of intraperitoneal tumor growth in ovarian cancer xenograft models. Furthermore, a combined intervention using an SCD inhibitor and an SFA-enriched diet initiated ER stress in tumors growing in vivo and potently blocked their dissemination. In all, our data support SCD as a key regulator of the cancer cell fate under metabolic stress and point to treatment strategies targeting the lipid balance.


Asunto(s)
Supervivencia Celular , Endorribonucleasas , Ácidos Grasos Insaturados , Neoplasias Ováricas , Progresión de la Enfermedad , Ácido Graso Desaturasas , Ácidos Grasos/farmacología , Ácidos Grasos Insaturados/farmacología , Femenino , Humanos , Fosfolípidos , Proteínas Serina-Treonina Quinasas , Estearoil-CoA Desaturasa/metabolismo
2.
Exp Cell Res ; 393(1): 112039, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32376288

RESUMEN

Expression of the homeodomain transcription factor HOXB13 has been demonstrated in several malignancies but its role in tumorigenesis remains elusive. We observed high levels of HOXB13 in poorly differentiated pediatric tumors including a highly aggressive childhood neoplasm - malignant rhabdoid tumor. In a xenograft model of rhabdoid tumor, knockout of HOXB13 diminished tumor growth while partial knockdown of HOXB13 promoted differentiation of tumor cells into bone. These results suggest that HOXB13 enhances rhabdoid malignancy by interfering with mesenchymal stem cell differentiation. Consistent with this hypothesis, overexpression of HOXB13 in mesenchymal progenitor cells inhibited adipogenic, myogenic, and osteogenic differentiation. Mechanistically, we demonstrated that HOXB13 binds to super-enhancer regions regulating genes involved in differentiation and tumorigenesis.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio/metabolismo , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Huesos/metabolismo , Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Genes Homeobox/genética , Humanos , Factores de Transcripción/metabolismo
3.
Gynecol Oncol ; 159(1): 277-284, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32698955

RESUMEN

OBJECTIVE: To investigate the anti-tumor effect of a newly-developed dual inhibitor (APCS-540) of glycogen synthase kinase 3 beta (GSK3B) and histone deacetylases (HDACs) in ovarian cancer cells. METHODS: The effects of APCS-540 on cancer cell proliferation, migration, invasion and cancer stemness were investigated in vitro in human (KURAMOCHI, OVCA420, OVSAHO) and mouse (BR-Luc, ID8, MOSE-HRas-Myc) ovarian cancer cells. Cisplatin-sensitive (A2780) and cisplatin-resistant (A2780cis) cell lines were used to evaluate APCS-540's effect on chemoresistance. The immunocompetent syngeneic mouse model BR-Luc was used to test the effect of APCS-540 on ovarian cancer progression and survival. RESULTS: APCS-540 showed significant anti-tumor effects in vitro in both human and mouse ovarian cancer cells. Importantly, APCS-540 demonstrated marked cytotoxicity against cisplatin-resistant cancer cells and reversed cisplatin-resistance when used in combination with platinum. APCS-540 significantly decreased cancer cell invasion. A significant 66% increase in survival was observed in mice treated with APCS-540 compared to control mice. CONCLUSION: Dual inhibition of GSK3B and HDACs via APCS-540 showed potent anti-tumor activity in vitro and in vivo, suggesting that APCS-540 may provide a novel treatment option for ovarian cancer, including the platinum-resistant disease.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores de Histona Desacetilasas/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ratones , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Inhibidores de Proteínas Quinasas/uso terapéutico
4.
Br J Cancer ; 118(8): 1123-1129, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29555990

RESUMEN

BACKGROUND: Observational studies suggest greater height is associated with increased ovarian cancer risk, but cannot exclude bias and/or confounding as explanations for this. Mendelian randomisation (MR) can provide evidence which may be less prone to bias. METHODS: We pooled data from 39 Ovarian Cancer Association Consortium studies (16,395 cases; 23,003 controls). We applied two-stage predictor-substitution MR, using a weighted genetic risk score combining 609 single-nucleotide polymorphisms. Study-specific odds ratios (OR) and 95% confidence intervals (CI) for the association between genetically predicted height and risk were pooled using random-effects meta-analysis. RESULTS: Greater genetically predicted height was associated with increased ovarian cancer risk overall (pooled-OR (pOR) = 1.06; 95% CI: 1.01-1.11 per 5 cm increase in height), and separately for invasive (pOR = 1.06; 95% CI: 1.01-1.11) and borderline (pOR = 1.15; 95% CI: 1.02-1.29) tumours. CONCLUSIONS: Women with a genetic propensity to being taller have increased risk of ovarian cancer. This suggests genes influencing height are involved in pathways promoting ovarian carcinogenesis.


Asunto(s)
Estatura/fisiología , Carcinoma Epitelial de Ovario/epidemiología , Neoplasias Ováricas/epidemiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estatura/genética , Carcinoma Epitelial de Ovario/genética , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Geografía , Humanos , Análisis de la Aleatorización Mendeliana , Persona de Mediana Edad , Neoplasias Ováricas/genética , Factores de Riesgo , Adulto Joven
5.
Int J Mol Sci ; 19(9)2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134598

RESUMEN

Thymidylate synthase (TYMS) is a crucial enzyme for DNA synthesis. TYMS expression is regulated by its antisense mRNA, ENOSF1. Disrupted regulation may promote uncontrolled DNA synthesis and tumor growth. We sought to replicate our previously reported association between rs495139 in the TYMS-ENOSF1 3' gene region and increased risk of mucinous ovarian carcinoma (MOC) in an independent sample. Genotypes from 24,351 controls to 15,000 women with invasive OC, including 665 MOC, were available. We estimated per-allele odds ratios (OR) and 95% confidence intervals (CI) using unconditional logistic regression, and meta-analysis when combining these data with our previous report. The association between rs495139 and MOC was not significant in the independent sample (OR = 1.09; 95% CI = 0.97⁻1.22; p = 0.15; N = 665 cases). Meta-analysis suggested a weak association (OR = 1.13; 95% CI = 1.03⁻1.24; p = 0.01; N = 1019 cases). No significant association with risk of other OC histologic types was observed (p = 0.05 for tumor heterogeneity). In expression quantitative trait locus (eQTL) analysis, the rs495139 allele was positively associated with ENOSF1 mRNA expression in normal tissues of the gastrointestinal system, particularly esophageal mucosa (r = 0.51, p = 1.7 × 10-28), and nonsignificantly in five MOC tumors. The association results, along with inconclusive tumor eQTL findings, suggest that a true effect of rs495139 might be small.


Asunto(s)
Adenocarcinoma Mucinoso/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Polimorfismo de Nucleótido Simple , Proteínas/genética , ARN sin Sentido/genética , Timidilato Sintasa/genética , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patología , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética , Humanos , Hidroliasas , Modelos Logísticos , Persona de Mediana Edad , Oportunidad Relativa , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas/metabolismo , Sitios de Carácter Cuantitativo , ARN sin Sentido/metabolismo , Riesgo , Transducción de Señal , Timidilato Sintasa/metabolismo
6.
Carcinogenesis ; 36(7): 739-47, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25926422

RESUMEN

ADAM metallopeptidase domain 12 (ADAM12) is a promising biomarker because of its low expression in normal tissues and high expression in a variety of human cancers. However, ADAM12 levels in ovarian cancer have not been well characterized. We previously identified ADAM12 as one of the signature genes associated with poor survival in high-grade serous ovarian carcinoma (HGSOC). Here, we sought to determine if high levels of the ADAM12 protein and/or messenger RNA (mRNA) are associated with clinical variables in HGSOC. We show that high protein levels of ADAM12 in banked preoperative sera are associated with shorter progression-free and overall survival. Tumor levels of ADAM12 mRNA were also associated with shorter progression-free and overall survival as well as with lymphatic and vascular invasion, and residual tumor volume following cytoreductive surgery. The majority of genes co-expressed with ADAM12 in HGSOC were transforming growth factor (TGF)ß signaling targets that function in collagen remodeling and cell-matrix adhesion. In tumor sections, the ADAM12 protein and mRNA were expressed in epithelial cancer cells and surrounding stromal cells. In vitro data showed that ADAM12 mRNA levels can be increased by TGFß signaling and direct contact between epithelial and stromal cells. High tumor levels of ADAM12 mRNA were characteristic of the mesenchymal/desmoplastic molecular subtype of HGSOC, which is known to have the poorest prognosis. Thus, ADAM12 may be a useful biomarker of aggressive ovarian cancer for which standard treatment is not effective.


Asunto(s)
Proteínas ADAM/sangre , Proteínas de la Membrana/sangre , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAM12 , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/cirugía , Pronóstico , Transducción de Señal , Células del Estroma/metabolismo , Células del Estroma/patología , Tasa de Supervivencia , Factor de Crecimiento Transformador beta/metabolismo
7.
Biochem Biophys Res Commun ; 463(4): 551-6, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26047697

RESUMEN

Familial breast and ovarian cancer are often caused by inherited mutations of BRCA1. While current prognoses for such patients are rather poor, inhibition of poly-ADP ribose polymerase 1 (PARP1) induces synthetic lethality in cells that are defective in homologous recombination. BMN 673 is a potent PARP1 inhibitor that is being clinically evaluated for treatment of BRCA-mutant cancers. Using the Brca1-deficient murine epithelial ovarian cancer cell line BR5FVB1-Akt, we investigated whether the antitumor effects of BMN 673 extend beyond its known pro-apoptotic function. Administration of modest amounts of BMN 673 greatly improved the survival of mice bearing subcutaneous or intraperitoneal tumors. We thus hypothesized that BMN 673 may influence the composition and function of immune cells in the tumor microenvironment. Indeed, BMN 673 significantly increases the number of peritoneal CD8(+) T cells and NK cells as well as their production of IFN-γ and TNF-α. These data suggest that the cell stress caused by BMN 673 induces not only cancer cell-intrinsic apoptosis but also cancer cell-extrinsic antitumor immune effects in a syngeneic murine model of ovarian cancer. BMN 673 may therefore serve as a promising adjuvant therapy to immunotherapy to achieve durable responses among patients whose tumors harbor defects in homologous recombination.


Asunto(s)
Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Genes BRCA1 , Neoplasias Ováricas/inmunología , Ftalazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Animales , Femenino , Citometría de Flujo , Ratones , Ratones Noqueados , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Poli(ADP-Ribosa) Polimerasa-1 , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Gynecol Oncol ; 139(3): 394-400, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26348314

RESUMEN

OBJECTIVE: Suboptimal cytoreductive surgery in advanced epithelial ovarian cancer (EOC) is associated with poor survival but it is unknown if poor outcome is due to the intrinsic biology of unresectable tumors or insufficient surgical effort resulting in residual tumor-sustaining clones. Our objective was to identify the potential molecular pathway(s) and cell type(s) that may be responsible for suboptimal surgical resection. METHODS: By comparing gene expression in optimally and suboptimally cytoreduced patients, we identified a gene network associated with suboptimal cytoreduction and explored the biological processes and cell types associated with this gene network. RESULTS: We show that primary tumors from suboptimally cytoreduced patients express molecular signatures that are typically present in a distinct molecular subtype of EOC characterized by increased stromal activation and lymphovascular invasion. Similar molecular pathways are present in EOC metastases, suggesting that primary tumors in suboptimally cytoreduced patients are biologically similar to metastatic tumors. We demonstrate that the suboptimal cytoreduction network genes are enriched in reactive tumor stroma cells rather than malignant tumor cells. CONCLUSION: Our data suggest that the success of cytoreductive surgery is dictated by tumor biology, such as extensive stromal reaction and increased invasiveness, which may hinder surgical resection and ultimately lead to poor survival.


Asunto(s)
Carcinoma/genética , Carcinoma/cirugía , Procedimientos Quirúrgicos de Citorreducción , Neoplasias Ováricas/genética , Neoplasias Ováricas/cirugía , Área Bajo la Curva , Vasos Sanguíneos/patología , Carcinoma/secundario , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Vasos Linfáticos/patología , Invasividad Neoplásica , Neoplasia Residual , Neoplasias Ováricas/patología , Valor Predictivo de las Pruebas , Curva ROC , Células del Estroma
9.
Gynecol Oncol ; 136(3): 542-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25528498

RESUMEN

OBJECTIVE: Ovarian cancer is a hormone-related disease with a strong genetic basis. However, none of its high-penetrance susceptibility genes and GWAS-identified variants to date are known to be involved in hormonal pathways. Given the hypothesized etiologic role of gonadotropins, an assessment of how variability in genes involved in the gonadotropin signaling pathway impacts disease risk is warranted. METHODS: Genetic data from 41 ovarian cancer study sites were pooled and unconditional logistic regression was used to evaluate whether any of the 2185 SNPs from 11 gonadotropin signaling pathway genes was associated with ovarian cancer risk. A burden test using the admixture likelihood (AML) method was also used to evaluate gene-level associations. RESULTS: We did not find any genome-wide significant associations between individual SNPs and ovarian cancer risk. However, there was some suggestion of gene-level associations for four gonadotropin signaling pathway genes: INHBB (p=0.045, mucinous), LHCGR (p=0.046, high-grade serous), GNRH (p=0.041, high-grade serous), and FSHB (p=0.036, overall invasive). There was also suggestive evidence for INHA (p=0.060, overall invasive). CONCLUSIONS: Ovarian cancer studies have limited sample numbers, thus fewer genome-wide susceptibility alleles, with only modest associations, have been identified relative to breast and prostate cancers. We have evaluated the majority of ovarian cancer studies with biological samples, to our knowledge, leaving no opportunity for replication. Using both our understanding of biology and powerful gene-level tests, we have identified four putative ovarian cancer loci near INHBB, LHCGR, GNRH, and FSHB that warrant a second look if larger sample sizes and denser genotype chips become available.


Asunto(s)
Biomarcadores de Tumor/genética , Predisposición Genética a la Enfermedad , Gonadotropinas/metabolismo , Neoplasias Ováricas/genética , Polimorfismo de Nucleótido Simple , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Femenino , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Modelos Logísticos , Neoplasias Ováricas/metabolismo , Factores de Riesgo , Transducción de Señal
10.
Ann Surg Oncol ; 21 Suppl 4: S758-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25124473

RESUMEN

BACKGROUND: Human basal-like breast cancer (BLBC) has a poor prognosis and is often identified by expression of the epidermal growth factor receptor (EGFR). BLBC remains a major clinical challenge because its pathogenesis is not well understood, thus hindering efforts to develop targeted therapies. Recent data implicate the forkhead box C1 (FOXC1) transcription factor as an important prognostic biomarker and functional regulator of BLBC, but its regulatory mechanism and impact on BLBC tumorigenesis remain unclear. METHODS: The association between FOXC1 and EGFR expression in human breast cancer was examined by immunohistochemistry in formalin-fixed tissues and analysis of the TCGA database. The regulation of FOXC1 by EGFR activation was investigated in MDA-MB-468 cells using immunoblotting, qRT-PCR, and luciferase activity assays. This EGFR effect on FOXC1 expression was confirmed using the MDA-MB-468 xenograft model. RESULTS: Both FOXC1 mRNA and protein levels significantly correlated with EGFR expression in human breast tumors. EGFR activation induced FOXC1 transcription through the ERK and Akt pathways in BLBC. EGFR inhibition in vivo reduced FOXC1 expression in xenograft tumors. We also found that FOXC1 knockdown impaired the effects of EGF on BLBC cell proliferation, migration, and invasion. CONCLUSIONS: Our findings uncover a novel EGFR-FOXC1 signaling axis critical for BLBC cell functions, supporting the notion that intervention in the FOXC1 pathway may provide potential modalities for BLBC treatment.


Asunto(s)
Receptores ErbB/genética , Factores de Transcripción Forkhead/genética , ARN Mensajero/análisis , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Receptores ErbB/análisis , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/metabolismo , Gefitinib , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinazolinas/uso terapéutico , ARN Interferente Pequeño/genética , Transfección , Neoplasias de la Mama Triple Negativas/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Regulación hacia Arriba
12.
Gynecol Oncol ; 133(3): 584-90, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24680909

RESUMEN

OBJECTIVE: Women with BRCA-associated ovarian cancer demonstrate excellent responses to Pegylated Liposomal Doxorubicin (PLD). PLD has also been shown to enhance T cell recognition of tumor cells. Here we characterize immunophenotypic changes associated with BRCA1 dysfunction in ovarian cancer cells, and evaluate the T cell contribution to the therapeutic efficacy of PLD in a BRCA1- ovarian cancer model to determine whether enhanced anti-tumor immunity contributes to the improved response to PLD in BRCA1- ovarian cancers. METHODS: The immunophenotype of BRCA1- and wild-type (WT) ovarian cancer cells and their response to PLD were compared in vitro using flow cytometry. T cell recruitment to BRCA1- tumors was evaluated with flow cytometry and immunohistochemistry. The contribution of T cell populations to the therapeutic effect of PLD in a BRCA1- model was evaluated using immunodepleting antibodies with PLD in vivo. RESULTS: The cytotoxic response to PLD was similar in BRCA1- and WT cells in vitro. BRCA1- inactivation resulted in higher expression of Fas and MHC-I at baseline and after PLD exposure. PLD prolonged the survival of BRCA1- tumor bearing mice and increased intratumoral T cell recruitment. CD4+ depletion combined with PLD significantly prolonged overall survival (p=0.0204) in BRCA1- tumor-bearing mice. CONCLUSION: Differences in the immunophenotype of BRCA1- and WT cells are amplified by PLD exposure. The enhanced immunomodulatory effects of PLD in BRCA1- tumors may be exploited therapeutically by eliminating suppressive CD4+ T cells. Our results support further study of combination therapy using PLD and immune agents, particularly in women with BRCA gene mutations.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Doxorrubicina/análogos & derivados , Genes BRCA1 , Inmunomodulación/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Neoplasias Ováricas/genética , Linfocitos T/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/inmunología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/inmunología , Doxorrubicina/farmacología , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Neoplasias Ováricas/inmunología , Polietilenglicoles/farmacología , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Gynecol Oncol ; 132(2): 334-42, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24368280

RESUMEN

OBJECTIVE: To identify molecular prognosticators and therapeutic targets for high-grade serous epithelial ovarian cancers (EOCs) using genetic analyses driven by biologic features of EOC pathogenesis. METHODS: Ovarian tissue samples (n = 172; 122 serous EOCs, 30 other EOCs, 20 normal/benign) collected prospectively from sequential patients undergoing gynecologic surgery were analyzed using RNA expression microarrays. Samples were classified based on expression of genes with potential relevance in ovarian cancer. Gene sets were defined using Rosetta Similarity Search Tool (ROAST) and analysis of variance (ANOVA). Gene copy number variations were identified by array comparative genomic hybridization. RESULTS: No distinct subgroups of EOC could be identified by unsupervised clustering, however, analyses based on genes correlated with periostin (POSTN) and estrogen receptor-alpha (ESR1) yielded distinct subgroups. When 95 high-grade serous EOCs were grouped by genes based on ANOVA comparing ESR1/WT1 and POSTN/TGFBI samples, overall survival (OS) was significantly shorter for 43 patients with tumors expressing genes associated with POSTN/TGFBI compared to 52 patients with tumors expressing genes associated with ESR1/WT1 (median 30 versus 49 months, respectively; P = 0.022). Several targets with therapeutic potential were identified within each subgroup. BRCA germline mutations were more frequent in the ESR1/WT1 subgroup. Proliferation-associated genes and TP53 status (mutated or wild-type) did not correlate with survival. Findings were validated using independent ovarian cancer datasets. CONCLUSIONS: Two distinct molecular subgroups of high-grade serous EOCs based on POSTN/TGFBI and ESR1/WT1 expressions were identified with significantly different OS. Specific differentially expressed genes between these subgroups provide potential prognostic and therapeutic targets.


Asunto(s)
Moléculas de Adhesión Celular/genética , Cistadenocarcinoma Seroso/genética , Proteínas de la Matriz Extracelular/genética , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/genética , Factor de Crecimiento Transformador beta/genética , Carcinoma Epitelial de Ovario , Moléculas de Adhesión Celular/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Análisis por Micromatrices , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Pronóstico , Estudios Prospectivos , Análisis de Supervivencia , Transcriptoma , Factor de Crecimiento Transformador beta/metabolismo
14.
Proc Natl Acad Sci U S A ; 108(2): 745-50, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187397

RESUMEN

Inhibition of the DNA repair enzyme poly(ADP-ribose) polymerase 1 (PARP1) with small molecules has been shown to be an effective treatment for ovarian cancer with BRCA mutations. Here, we report the in vivo administration of siRNA to Parp1 in mouse models of ovarian cancer. A unique member of the lipid-like materials known as lipidoids is shown to deliver siRNA to disseminated murine ovarian carcinoma allograft tumors following intraperitoneal (i.p.) injection. siParp1 inhibits cell growth, primarily by induction of apoptosis, in Brca1-deficient cells both in vitro and in vivo. Additionally, the treatment extends the survival of mice bearing tumors derived from Brca1-deficient ovarian cancer cells but not from Brca1 wild-type cells, confirming the proposed mechanism of synthetic lethality. Because there are 17 members of the Parp family, the inherent complementarity of RNA affords a high level of specificity for therapeutically addressing Parp1 in the context of impaired homologous recombination.


Asunto(s)
Proteína BRCA1/genética , Nanopartículas/química , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Poli(ADP-Ribosa) Polimerasas/genética , ARN Interferente Pequeño/metabolismo , Animales , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Ratones , Nanotecnología/métodos , Trasplante de Neoplasias , Interferencia de ARN , Resultado del Tratamiento
15.
Sci Adv ; 10(16): eadk8805, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38630822

RESUMEN

High-grade serous ovarian carcinoma (HGSOC), the deadliest form of ovarian cancer, is typically diagnosed after it has metastasized and often relapses after standard-of-care platinum-based chemotherapy, likely due to advanced tumor stage, heterogeneity, and immune evasion and tumor-promoting signaling from the tumor microenvironment. To understand how spatial heterogeneity contributes to HGSOC progression and early relapse, we profiled an HGSOC tissue microarray of patient-matched longitudinal samples from 42 patients. We found spatial patterns associated with early relapse, including changes in T cell localization, malformed tertiary lymphoid structure (TLS)-like aggregates, and increased podoplanin-positive cancer-associated fibroblasts (CAFs). Using spatial features to compartmentalize the tissue, we found that plasma cells distribute in two different compartments associated with TLS-like aggregates and CAFs, and these distinct microenvironments may account for the conflicting reports about the role of plasma cells in HGSOC prognosis.


Asunto(s)
Antineoplásicos , Fibroblastos Asociados al Cáncer , Neoplasias Ováricas , Femenino , Humanos , Fibroblastos Asociados al Cáncer/patología , Recurrencia Local de Neoplasia , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Recurrencia , Microambiente Tumoral
16.
NPJ Precis Oncol ; 8(1): 35, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38360876

RESUMEN

Effective targeting of cancer-associated fibroblasts (CAFs) is hindered by the lack of specific biomarkers and a poor understanding of the mechanisms by which different populations of CAFs contribute to cancer progression. While the role of TGFß in CAFs is well-studied, less attention has been focused on a structurally and functionally similar protein, Activin A (encoded by INHBA). Here, we identified INHBA(+) CAFs as key players in tumor promotion and immunosuppression. Spatiotemporal analyses of patient-matched primary, metastatic, and recurrent ovarian carcinomas revealed that aggressive metastatic tumors enriched in INHBA(+) CAFs were also enriched in regulatory T cells (Tregs). In ovarian cancer mouse models, intraperitoneal injection of the Activin A neutralizing antibody attenuated tumor progression and infiltration with pro-tumorigenic subsets of myofibroblasts and macrophages. Downregulation of INHBA in human ovarian CAFs inhibited pro-tumorigenic CAF functions. Co-culture of human ovarian CAFs and T cells revealed the dependence of Treg differentiation on direct contact with INHBA(+) CAFs. Mechanistically, INHBA/recombinant Activin A in CAFs induced the autocrine expression of PD-L1 through SMAD2-dependent signaling, which promoted Treg differentiation. Collectively, our study identified an INHBA(+) subset of immunomodulatory pro-tumoral CAFs as a potential therapeutic target in advanced ovarian cancers which typically show a poor response to immunotherapy.

17.
Nat Commun ; 15(1): 2498, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509063

RESUMEN

T cell-based immunotherapies have exhibited promising outcomes in tumor control; however, their efficacy is limited in immune-excluded tumors. Cancer-associated fibroblasts (CAFs) play a pivotal role in shaping the tumor microenvironment and modulating immune infiltration. Despite the identification of distinct CAF subtypes using single-cell RNA-sequencing (scRNA-seq), their functional impact on hindering T-cell infiltration remains unclear, particularly in soft-tissue sarcomas (STS) characterized by low response rates to T cell-based therapies. In this study, we characterize the STS microenvironment using murine models (in female mice) with distinct immune composition by scRNA-seq, and identify a subset of CAFs we termed glycolytic cancer-associated fibroblasts (glyCAF). GlyCAF rely on GLUT1-dependent expression of CXCL16 to impede cytotoxic T-cell infiltration into the tumor parenchyma. Targeting glycolysis decreases T-cell restrictive glyCAF accumulation at the tumor margin, thereby enhancing T-cell infiltration and augmenting the efficacy of chemotherapy. These findings highlight avenues for combinatorial therapeutic interventions in sarcomas and possibly other solid tumors. Further investigations and clinical trials are needed to validate these potential strategies and translate them into clinical practice.


Asunto(s)
Fibroblastos Asociados al Cáncer , Sarcoma , Neoplasias de los Tejidos Blandos , Femenino , Animales , Ratones , Resistencia a Antineoplásicos , Sarcoma/tratamiento farmacológico , Sarcoma/genética , Linfocitos T Citotóxicos , Microambiente Tumoral , Fibroblastos
18.
Nat Commun ; 15(1): 2528, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38514660

RESUMEN

Intratumoral tertiary lymphoid structures (TLSs) have been associated with improved outcome in various cohorts of patients with cancer, reflecting their contribution to the development of tumor-targeting immunity. Here, we demonstrate that high-grade serous ovarian carcinoma (HGSOC) contains distinct immune aggregates with varying degrees of organization and maturation. Specifically, mature TLSs (mTLS) as forming only in 16% of HGSOCs with relatively elevated tumor mutational burden (TMB) are associated with an increased intratumoral density of CD8+ effector T (TEFF) cells and TIM3+PD1+, hence poorly immune checkpoint inhibitor (ICI)-sensitive, CD8+ T cells. Conversely, CD8+ T cells from immunologically hot tumors like non-small cell lung carcinoma (NSCLC) are enriched in ICI-responsive TCF1+ PD1+ T cells. Spatial B-cell profiling identifies patterns of in situ maturation and differentiation associated with mTLSs. Moreover, B-cell depletion promotes signs of a dysfunctional CD8+ T cell compartment among tumor-infiltrating lymphocytes from freshly isolated HGSOC and NSCLC biopsies. Taken together, our data demonstrate that - at odds with NSCLC - HGSOC is associated with a low density of follicular helper T cells and thus develops a limited number of mTLS that might be insufficient to preserve a ICI-sensitive TCF1+PD1+ CD8+ T cell phenotype. These findings point to key quantitative and qualitative differences between mTLSs in ICI-responsive vs ICI-irresponsive neoplasms that may guide the development of alternative immunotherapies for patients with HGSOC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Neoplasias Ováricas , Estructuras Linfoides Terciarias , Humanos , Femenino , Linfocitos T CD8-positivos , Neoplasias Ováricas/patología , Linfocitos Infiltrantes de Tumor , Fenotipo , Microambiente Tumoral
19.
Pharmacol Ther ; 245: 108413, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37059310

RESUMEN

Reprogramming of cellular metabolism is a hallmark of cancer. Cancer cells undergo metabolic adaptations to maintain tumorigenicity and survive under the attack of immune cells and chemotherapy in the tumor microenvironment. Metabolic alterations in ovarian cancer in part overlap with findings from other solid tumors and in part reflect unique traits. Altered metabolic pathways not only facilitate ovarian cancer cells' survival and proliferation but also endow them to metastasize, acquire resistance to chemotherapy, maintain cancer stem cell phenotype and escape the effects of anti-tumor immune defense. In this review, we comprehensively review the metabolic signatures of ovarian cancer and their impact on cancer initiation, progression, and resistance to treatment. We highlight novel therapeutic strategies targeting metabolic pathways under development.


Asunto(s)
Glucólisis , Neoplasias Ováricas , Humanos , Femenino , Glucólisis/genética , Neoplasias Ováricas/genética , Metabolismo Energético , Redes y Vías Metabólicas , Microambiente Tumoral
20.
bioRxiv ; 2023 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-37503008

RESUMEN

The persistence of ovarian cancer stem-like cells (OvCSCs) after chemotherapy resistance has been implicated in relapse. However, the ability of these relatively quiescent cells to produce the robust tumor regrowth necessary for relapse remains an enigma. Since normal stem cells exist in a niche, and tumor-associated macrophages (TAMs) are the highest abundance immune cell within ovarian tumors, we hypothesized that TAMs may influence OvCSC proliferation. To test this, we optimized OvCSC enrichment by sphere culture and in vitro polarization of monocytes to a TAM-like M2 phenotype. Using cocultures that permitted the exchange of only soluble factors, we found that M2 macrophages increased the proliferation of sphere cells. Longer-term exposure (5-7 days) to soluble TAM factors led to retention of some stem cell features by OvCSCs but loss of others, suggesting that TAMs may support an intermediate stemness phenotype in OvCSCs. Although TAM coculture decreased the percentage of OvCSCs surviving chemotherapy, it increased the overall number. We therefore sought to determine the influence of this interaction on chemotherapy efficacy in vivo and found that inhibiting macrophages improved chemotherapy response. Comparing the gene expression changes in OvCSCs cocultured with TAMs to publicly available patient data identified 34 genes upregulated in OvCSCs by exposure to soluble TAM factors whose expression correlates with outcome. Overall, these data suggest that TAMs may influence OvCSC proliferation and impact therapeutic response.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA