Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
1.
Genes Dev ; 31(11): 1109-1121, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28698296

RESUMEN

A key feature of high-grade serous ovarian carcinoma (HGSOC) is frequent amplification of the 3q26 locus harboring PRKC-ι (PRKCI). Here, we show that PRKCI is also expressed in early fallopian tube lesions, called serous tubal intraepithelial carcinoma. Transgenic mouse studies establish PRKCI as an ovarian cancer-specific oncogene. Mechanistically, we show that the oncogenic activity of PRKCI relates in part to the up-regulation of TNFα to promote an immune-suppressive tumor microenvironment characterized by an abundance of myeloid-derived suppressor cells and inhibition of cytotoxic T-cell infiltration. Furthermore, system-level and functional analyses identify YAP1 as a downstream effector in tumor progression. In human ovarian cancers, high PRKCI expression also correlates with high expression of TNFα and YAP1 and low infiltration of cytotoxic T cells. The PRKCI-YAP1 regulation of the tumor immunity provides a therapeutic strategy for highly lethal ovarian cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Tolerancia Inmunológica/genética , Isoenzimas/genética , Isoenzimas/inmunología , Neoplasias Ováricas/genética , Proteína Quinasa C/genética , Proteína Quinasa C/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Movimiento Celular/genética , Citocinas/genética , Femenino , Humanos , Isoenzimas/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/fisiopatología , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Señalizadoras YAP
2.
Int J Cancer ; 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38848494

RESUMEN

Extracellular vesicles (EVs) function as natural mediators of intercellular communication, secreted by cells to facilitate cell-cell signaling. Due to their low toxicity, immunogenicity, biodegradability, and potential to encapsulate therapeutic drugs, EVs hold significant therapeutic promise. Nevertheless, their limited targeting ability often diminishes their therapeutic impact. Therefore, enhancing EVs by incorporating targeting units onto their membranes could bolster their targeting capabilities, enabling them to accumulate in specific cells and tissues. In this study, we engineered EVs to fuse ephrin-B2 with the EV membrane protein LAMP2b. This modification aimed to direct the engineered EVs toward the ephrin-B4 receptor expressed on the surface of ovarian cancer cells. The engineered EVs retained their inherent properties, including size, expression of EV membrane proteins, and morphology, upon isolation. In vitro experiments using real-time imaging revealed that EVs engineered with the ephrin-B2 ligand exhibited substantial internalization and uptake by ovarian cancer cells, in stark contrast to native EVs. In vivo, the engineered EVs carrying the ephrin-B2 ligand effectively targeted ovarian cancer cells, surpassing the targeting efficiency of control EVs. This innovative approach establishes a novel targeting system, enhancing the uptake of EVs by ovarian cancer cells. Our findings underscore the potential of using EVs to target cancer cells, thereby enhancing the effectiveness of anti-cancer therapies while minimizing off-target effects and toxicity in normal cells and organs.

3.
Cancer Immunol Immunother ; 73(5): 80, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38554167

RESUMEN

Cancer immunotherapy has seen significant success in the last decade for cancer management by enhancing endogenous cancer immunity. However, immunotherapies developed thus far have seen limited success in the majority of high-grade serous carcinoma (HGSC) ovarian cancer patients. This is largely due to the highly immunosuppressive tumour microenvironment of HGSC and late-stage identification. Thus, novel treatment interventions are needed to overcome this immunosuppression and complement existing immunotherapies. Here, we have identified through analysis of > 600 human HGSC tumours a critical role for Let-7i in modulating the tumoural immune network. Tumoural expression of Let-7i had high positive correlation with anti-cancer immune signatures in HGSC patients. Confirming this role, enforced Let-7i expression in murine HGSC tumours resulted in a significant decrease in tumour burden with a significant increase in tumour T cell numbers in tumours. In concert with the improved tumoural immunity, Let-7i treatment also significantly increased CD86 expression in antigen presenting cells (APCs) in the draining lymph nodes, indicating enhanced APC activity. Collectively, our findings highlight an important role of Let-7i in anti-tumour immunity and its potential use for inducing an anti-tumour effect in HGSC.


Asunto(s)
MicroARNs , Neoplasias Ováricas , Animales , Femenino , Humanos , Ratones , MicroARNs/genética , Neoplasias Ováricas/patología , Linfocitos T/metabolismo , Microambiente Tumoral
4.
Bioconjug Chem ; 30(10): 2675-2683, 2019 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-31560538

RESUMEN

Exosomes have attracted tremendous attention due to their important role in physiology, pathology, and oncology, as well as promising potential in biomedical applications. Although great efforts have been dedicated to investigating their biological properties and applications as natural cancer drug-delivery systems, the systemic biodistribution of exosomes remains underexplored. In addition, exosome-based drug delivery is inevitably hindered by the robust liver clearance, leading to suboptimal tumor retention and therapeutic efficiency. In this study, we report one of the first examples using in vivo positron emission tomography (PET) for noninvasive monitoring of copper-64 (64Cu)-radiolabeled polyethylene glycol (PEG)-modified exosomes, achieving excellent imaging quality and quantitative measurement of blood residence and tumor retention. PEGylation not only endowed exosomes with a superior pharmacokinetic profile and great accumulation in the tumor versus traditionally reported native exosomes but also reduced premature hepatic sequestration and clearance of exosomes, findings that promise enhanced therapeutic delivery efficacy and safety in future studies. More importantly, this study provides important guidelines about surface engineering, radiochemistry, and molecular imaging in obtaining accurate and quantitative biodistribution information on exosomes, which may benefit future exploration in the realm of exosomes.


Asunto(s)
Radioisótopos de Cobre/química , Exosomas/metabolismo , Polietilenglicoles/química , Polietilenglicoles/metabolismo , Tomografía de Emisión de Positrones/métodos , Animales , Línea Celular Tumoral , Ratones , Polietilenglicoles/farmacocinética , Distribución Tisular
5.
Mol Ther Oncolytics ; 31: 100725, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-37781339

RESUMEN

Immunotherapies have emerged as promising strategies for cancer treatment. However, existing immunotherapies have poor activity in high-grade serous ovarian cancer (HGSC) due to the immunosuppressive tumor microenvironment and the associated low tumoral CD8+ T cell (CTL) infiltration. Through multiple lines of evidence, including integrative analyses of human HGSC tumors, we have identified miR-146a as a master regulator of CTL infiltration in HGSC. Tumoral miR-146a expression is positively correlated with anti-cancer immune signatures in human HGSC tumors, and delivery of miR-146a to tumors resulted in significant reduction in tumor growth in both ID8-p53-/- and IG10 murine HGSC models. Increasing miR-146a expression in tumors improved anti-tumor immune responses by decreasing immune suppressive neutrophils and increasing CTL infiltration. Mechanistically, miR-146a targets IL-1 receptor-associated kinase 1 and tumor necrosis factor receptor-associated factor 6 adaptor molecules of the transcription factor nuclear factor κB signaling pathway in ID8-p53-/- cells and decreases production of the downstream neutrophil chemoattractant, C-X-C motif chemokine ligand 1. In addition to HGSC, tumoral miR-146a expression also correlates strongly with CTL infiltration in other cancer types including thyroid, prostate, breast, and adrenocortical cancers. Altogether, our findings highlight the ability of miR-146a to overcome immune suppression and improve CTL infiltration in tumors.

6.
Immunol Cell Biol ; 90(2): 187-96, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21423261

RESUMEN

Oncogene-specific downregulation mediated by RNA interference (RNAi) is a promising avenue for cancer therapy. In addition to specific gene silencing, in vivo RNAi treatment with short interfering RNAs (siRNAs) can initiate immune activation through innate immune receptors including Toll-like receptors, (TLRs) 7 and 8. Two recent studies have shown that activation of innate immunity by addition of tri-phosphate motifs to oncogene-specific siRNAs, or by co-treatment with CpG oligos, can potentiate siRNA antitumor effects. To date, there are no reports on applying such approach against human papillomavirus (HPV)-driven cancers. Here, we characterized the antitumor effects of non-modified siRNAs that can target a specific oncogene and/or recruit the innate immune system against HPV-driven tumors. Following the characterization of silencing efficacy and TLR7 immunostimulatory potential of 15 siRNAs targeting the HPV type 16 E6/E7 oncogenes, we identified a bifunctional siRNA sequence that displayed both potent gene silencing and active immunostimulation effect. In vivo systemic administration of this siRNA resulted in reduced growth of established TC-1 tumors in C57BL/6 mice. Ablation of TLR7 recruitment via 2'O-methyl modification of the oligo backbone reduced these antitumor effects. Further, a highly immunostimulatory, but non-HPV targeting siRNA was also able to exert antitumoral effects although for less prolonged time compared with the bifunctional siRNA. Collectively, our work demonstrates for the first time that siRNA-induced immunostimulation can have antitumoral effects against HPV-driven tumors in vivo, even independent of gene silencing efficacy.


Asunto(s)
Papillomavirus Humano 16/genética , Glicoproteínas de Membrana/genética , ARN Interferente Pequeño/administración & dosificación , Receptor Toll-Like 7/genética , Neoplasias del Cuello Uterino/inmunología , Animales , Línea Celular Tumoral , Femenino , Papillomavirus Humano 16/inmunología , Humanos , Inmunización/métodos , Masculino , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/inmunología , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Represoras/genética , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 7/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/virología
7.
Cancers (Basel) ; 14(17)2022 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-36077735

RESUMEN

Despite having similar histologic features, patients with high-grade serous ovarian carcinoma (HGSC) often experience highly variable outcomes. The underlying determinants for long-term survival (LTS, ≥10 years) versus short-term survival (STS, <3 years) are largely unknown. The present study sought to identify molecular predictors of LTS for women with HGSC. A cohort of 24 frozen HGSC samples was collected (12 LTS and 12 STS) and analyzed at DNA, RNA, and protein levels. OVCAR5 and OVCAR8 cell lines were used for in vitro validation studies. For in vivo studies, we injected OVCAR8 cells into the peritoneal cavity of female athymic nude mice. From RNAseq analysis, 11 genes were found to be differentially expressed between the STS and LTS groups (fold change > 2; false discovery rate < 0.01). In the subsequent validation cohort, transmembrane protein 62 (TMEM62) was found to be related to LTS. CIBERSORT analysis showed that T cells (follicular helper) were found at higher levels in tumors from LTS than STS groups. In vitro data using OVCAR5 and OVCAR8 cells showed decreased proliferation with TMEM62 overexpression and positive correlation with a longevity-regulating pathway (KEGG HSA04213) at the RNA level. In vivo analysis using the OVCAR8-TMEM62-TetON model showed decreased tumor burden in mice with high- vs. low-expressing TMEM62 tumors. Our results demonstrate that restoring TMEM62 may be a novel approach for treatment of HGSC. These findings may have implications for biomarker and intervention strategies to help improve patient outcomes

8.
Mol Cancer Res ; 19(11): 1917-1928, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34348992

RESUMEN

Investigations into the function of nonpromoter DNA methylation have yielded new insights into epigenetic regulation of gene expression. Previous studies have highlighted the importance of distinguishing between DNA methylation in discrete functional regions; however, integrated nonpromoter DNA methylation and gene expression analyses across a wide number of tumor types and corresponding normal tissues have not been performed. Through integrated analysis of gene expression and DNA methylation profiles, we examined 32 tumor types and identified 57 tumor suppressors and oncogenes out of 260 genes exhibiting a correlation of > 0.5 between gene body methylation and gene expression in at least one tumor type. The lymphocyte-specific gene CARD11 exhibits robust association between gene body methylation and expression across 19 of 32 tumor types examined. It is significantly overexpressed in kidney renal cell carcinoma (KIRC) and lung adenocarcinoma (LUAD) tumor tissues in comparison with respective control samples; and is significantly associated with lower overall survival in KIRC. Contrary to its canonical function in lymphocyte NFκB activation, CARD11 activates the mTOR pathway in KIRC and LUAD, resulting in suppressed autophagy. Furthermore, demethylation of a CpG island within the gene body of CARD11 decreases gene expression. Collectively, our study highlights how DNA methylation outside the promoter region can impact tumor progression. IMPLICATIONS: Our study describes a novel regulatory role of gene body DNA methylation-dependent CARD11 expression on mTOR signaling and its impact on tumor progression.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/metabolismo , Metilación de ADN/genética , Linfocitos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , Pronóstico , Transducción de Señal , Transfección
9.
Cell Rep ; 34(6): 108726, 2021 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-33567287

RESUMEN

Tumor and stromal interactions consist of reciprocal signaling through cytokines, growth factors, direct cell-cell interactions, and extracellular vesicles (EVs). Small EVs (≤200 nm) have been considered critical messengers of cellular communication during tumor development. Here, we demonstrate that gain-of-function (GOF) p53 protein can be packaged into small EVs and transferred to fibroblasts. GOF p53 protein is selectively bound by heat shock protein 90 (HSP90), a chaperone protein, and packaged into small EVs. Inhibition of HSP90 activity blocks packaging of GOF, but not wild-type, p53 in small EVs. GOF p53-containing small EVs result in their conversion to cancer-associated fibroblasts. In vivo studies reveal that GOF p53-containing small EVs can enhance tumor growth and promote fibroblast transformation into a cancer-associated phenotype. These findings provide a better understanding of the complex interactions between cancer and stromal cells and may have therapeutic implications.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Vesículas Extracelulares , Mutación con Ganancia de Función , Proteína p53 Supresora de Tumor , Animales , Neoplasias Colorrectales/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/trasplante , Femenino , Células HT29 , Humanos , Ratones , Ratones Noqueados , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
Oncogene ; 40(2): 384-395, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33149280

RESUMEN

Paxillin (PXN), a key component of the focal adhesion complex, has been associated with cancer progression, but the underlying mechanisms are poorly understood. The purpose of this study was to elucidate mechanisms by which PXN affects cancer growth and progression, which we addressed using cancer patient data, cell lines, and orthotopic mouse models. We demonstrated a previously unrecognized mechanism whereby nuclear PXN enhances angiogenesis by transcriptionally regulating SRC expression. SRC, in turn, increases PLAT expression through NF-ĸB activation; PLAT promotes angiogenesis via LRP1 in endothelial cells. PXN silencing in ovarian cancer mouse models reduced angiogenesis, tumor growth, and metastasis. These findings provide a new understanding of the role of PXN in regulating tumor angiogenesis and growth.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Núcleo Celular/metabolismo , Regulación Neoplásica de la Expresión Génica , Neovascularización Patológica/patología , Neoplasias Ováricas/irrigación sanguínea , Paxillin/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Desnudos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Paxillin/antagonistas & inhibidores , Paxillin/genética , Pronóstico , Tasa de Supervivencia , Activador de Tejido Plasminógeno/genética , Activador de Tejido Plasminógeno/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
11.
Cell Rep ; 36(7): 109549, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34407412

RESUMEN

Despite wide use of anti-vascular endothelial growth factor (VEGF) therapy for many solid cancers, most individuals become resistant to this therapy, leading to disease progression. Therefore, new biomarkers and strategies for blocking adaptive resistance of cancer to anti-VEGF therapy are needed. As described here, we demonstrate that cancer-derived small extracellular vesicles package increasing quantities of VEGF and other factors in response to anti-VEGF therapy. The packaging process of VEGF into small extracellular vesicles (EVs) is mediated by the tetraspanin CD63. Furthermore, small EV-VEGF (eVEGF) is not accessible to anti-VEGF antibodies and can trigger intracrine VEGF signaling in endothelial cells. eVEGF promotes angiogenesis and enhances tumor growth despite bevacizumab treatment. These data demonstrate a mechanism where VEGF is partitioned into small EVs and promotes tumor angiogenesis and progression. These findings have clinical implications for biomarkers and therapeutic strategies for ovarian cancer.


Asunto(s)
Vesículas Extracelulares/metabolismo , Tetraspanina 30/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Anciano , Animales , Bevacizumab/farmacología , Bevacizumab/uso terapéutico , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Vesículas Extracelulares/ultraestructura , Femenino , Humanos , Ratones , Ratones Desnudos , Persona de Mediana Edad , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Neoplasias Ováricas/tratamiento farmacológico , Isoformas de Proteínas/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
12.
J Control Release ; 327: 512-532, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32800879

RESUMEN

Nanoparticles (NPs) that permit active targeting promise to play a key role in cancer therapy moving forward. However, in order to successfully advance into clinic, these delivery platforms not only must target individual tumoural cellular components but also require safe, efficient and scalable production. Herein, we review recent and innovative targeted nanoparticle delivery strategies to individual TME components, including cancer-associated blood and lymphatic vessels, pericytes, cancer associated fibroblasts, and cancer stem cells. In contrast to traditional therapies that promote widespread ablation, emerging nano-strategies that specifically modulate different cell populations of the TME, such as targeting pericytes and endothelial cells for vascular normalization, are proving to effectively deliver therapeutics to tumours. Additionally, new smart targeted NPs with transformable characteristics responsive to specific tumour microenvironmental cues demonstrate enhanced spatiotemporal control over cell targeting and therapeutic release. However, translating these therapies to the clinic requires overcoming several significant barriers such as failure to recapitulate the human TME in animal models and issues with NP targeting efficacy, safety and scalable production. We discuss recent efforts to overcome these challenges and innovative means to reduce off-target toxicities. We also highlight important deficiencies in current NP development and offer new perspectives on the design of pre-clinical and clinical trials to accelerate clinical translation of targeted NP platforms.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Células Endoteliales , Humanos , Neoplasias/tratamiento farmacológico , Microambiente Tumoral
13.
Curr Cancer Rep ; 2(1): 54-63, 2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-35814639

RESUMEN

Our purpose was to understand the effects of normoxia or hypoxia on 5-fluorouracil (5-FU) treatment in triple negative breast cancer (TNBC) cells, and characterize the molecular changes in hypoxia inducible factors (HIFs) and cyclooxygenase-2 (COX-2) following treatment. Cell viability and protein levels of HIFs and COX-2 were determined after wild type and HIF silenced MDA-MB-231 cells, and wild type SUM-149 cells, were treated with 5-FU under normoxia or hypoxia. 5-FU reduced cell viability to the same levels irrespective of normoxia or hypoxia. HIF silenced MDA-MB-231 cells showed comparable changes in cell viability, supporting observations that hypoxia and the HIF pathways did not significantly influence cell viability reduction by 5-FU. Our data suggest that HIF-2α accumulation may predispose cancer cells to cell death under hypoxia. SUM-149 cells that have higher COX-2 and HIF-2α following 24 h of hypoxia, were more sensitive to 96 h of hypoxia compared to MDA-MB-231 cells, and were more sensitive to 5-FU than MDA-MB-231 cells. COX-2 levels changed with hypoxia and with 5-FU treatment but patterns were different between the two cell lines. At 96 h, COX-2 increased in both untreated and 5-FU treated cells under hypoxia in MDA-MB-231 cells. In SUM-149 cells, only treatment with 5-FU increased COX-2 at 96 h of hypoxia. Cells that survive hypoxia and 5-FU treatment may exhibit a more aggressive phenotype. Our results support understanding interactions between HIF and COX-2 with chemotherapeutic agents under normoxia and hypoxia, and investigating the use of COX-2 inhibitors in these settings.

14.
Mol Cancer Ther ; 19(8): 1727-1735, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32499298

RESUMEN

XPO1 inhibitors have shown promise in cancer treatment, but mechanisms of resistance to these drugs are not well understood. In this study, we established selective inhibitors of nuclear export (SINE)-resistant ovarian cancer cell lines from in vivo mouse tumors and determined the mechanisms of adaptive XPO1 inhibitor resistance using protein and genomic arrays. Pathway analyses revealed upregulation of the NRG1/ERBB3 pathway in SINE-resistant cells. Depletion of ERBB3 using siRNAs restored the antitumor effect of SINE in vitro and in vivo Furthermore, exogenous NRG1 decreased the antitumor effect of SINE in ovarian cancer cell lines with high ERBB3 expression, but not in those with low expression. These results suggest that NRG1 and ERBB3 expression is a potential biomarker of response to SINE treatment. The antitumor effect of SINE was reduced by exogenous NRG1 in an ERBB3-dependent manner. These findings suggest that NRG1 and ERBB3 are effective biomarkers that should be evaluated in future clinical trials and are relevant therapeutic targets for the treatment of SINE-resistant cancers.


Asunto(s)
Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hidrazinas/farmacología , Carioferinas/antagonistas & inhibidores , Neurregulina-1/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Receptor ErbB-3/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Triazoles/farmacología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Neurregulina-1/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptor ErbB-3/genética , Transcriptoma/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína Exportina 1
15.
Vet Comp Oncol ; 18(2): 206-213, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31441983

RESUMEN

Osteosarcoma is the most common paediatric primary bone malignancy. The major cause of death in osteosarcoma is drug-resistant pulmonary metastasis. Previous studies have shown that thioredoxin reductase 2 is a driver of metastasis in osteosarcoma and can be inhibited by auranofin (AF). Moreover, studies have shown that AF significantly reduces pulmonary metastases in xenotransplant models. Here, we describe a phase I/II study of AF in canine osteosarcoma, a well-recognized spontaneous model of human osteosarcoma. We performed a single-arm multicentre pilot study of AF in combination with standard of care (SOC) (amputation + carboplatin). We recruited 40 dogs to the trial and used a historical SOC-only control group (n = 26). Dogs >15 kg received 9 mg AF q3d PO and dogs <15 kg received 6 mg q3d. Follow-up occurred over at least a 3-year period. Auranofin plus SOC improved overall survival (OS) (P = .036) in all dogs treated. The improved outcome was attributable entirely to improved OS in male dogs (P = .009). At the time of writing, 10 dogs (25%) survive without measurable disease in the treatment group with survival times ranging between 806 and 1525 days. Our study shows that AF improves OS in male dogs when combined with SOC. Our findings have translational relevance for the management of canine and human osteosarcoma. Our data justify a larger multicentre phase 2 trial in dogs and a phase I/II trial in human patients with refractory disease at the time of initial surgery.


Asunto(s)
Antirreumáticos/uso terapéutico , Auranofina/uso terapéutico , Neoplasias Óseas/veterinaria , Carboplatino/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Osteosarcoma/veterinaria , Amputación Quirúrgica/veterinaria , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Antirreumáticos/administración & dosificación , Neoplasias Óseas/terapia , Carboplatino/administración & dosificación , Perros , Quimioterapia Combinada , Femenino , Masculino , Osteosarcoma/terapia , Proyectos Piloto , Factores Sexuales
17.
EBioMedicine ; 43: 127-137, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31056473

RESUMEN

BACKGROUND: Investigations into the function of non-promoter DNA methylation have yielded new insights into the epigenetic regulation of gene expression. However, integrated genome-wide non-promoter DNA methylation and gene expression analyses across a wide number of tumour types and corresponding normal tissues have not been performed. METHODS: To investigate the impact of non-promoter DNA methylation on cancer pathogenesis, we performed a large-scale analysis of gene expression and DNA methylation profiles, finding enrichment in the 3'UTR DNA methylation positively correlated with gene expression. Filtering for genes in which 3'UTR DNA methylation strongly correlated with gene expression yielded a list of genes enriched for functions involving T cell activation. FINDINGS: The important immune checkpoint gene Havcr2 showed a substantial increase in 3'UTR DNA methylation upon T cell activation and subsequent upregulation of gene expression in mice. Furthermore, this increase in Havcr2 gene expression was abrogated by treatment with decitabine. INTERPRETATION: These findings indicate that the 3'UTR is a functionally relevant DNA methylation site. Additionally, we show a potential novel mechanism of HAVCR2 regulation in T cells, providing new insights for modulating immune checkpoint blockade.


Asunto(s)
Regiones no Traducidas 3' , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Genómica , Neoplasias/genética , Linfocitos T/metabolismo , Animales , Biomarcadores de Tumor , Biología Computacional/métodos , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Bases de Datos Genéticas , Epigénesis Genética , Femenino , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genómica/métodos , Receptor 2 Celular del Virus de la Hepatitis A/genética , Humanos , Activación de Linfocitos/inmunología , Ratones , Neoplasias/inmunología , Neoplasias/mortalidad , Pronóstico , Linfocitos T/inmunología
18.
Mol Cancer Ther ; 18(5): 969-979, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30926640

RESUMEN

EP-100 is a synthetic lytic peptide that specifically targets the gonadotropin-releasing hormone receptor on cancer cells. To extend the utility of EP-100, we aimed to identify effective combination therapies with EP-100 for ovarian cancer and explore potential mechanisms of this combination. A series of in vitro (MTT assay, immunoblot analysis, reverse-phase protein array, comet assay, and immunofluorescence staining) and in vivo experiments were carried out to determine the biological effects of EP-100 alone and in combination with standard-of-care drugs. EP-100 decreased the viability of ovarian cancer cells and reduced tumor growth in orthotopic mouse models. Of five standard drugs tested (cisplatin, paclitaxel, doxorubicin, topotecan, and olaparib), we found that the combination of EP-100 and olaparib was synergistic in ovarian cancer cell lines. Further experiments revealed that combined treatment of EP-100 and olaparib significantly increased the number of nuclear foci of phosphorylated histone H2AX. In addition, the extent of DNA damage was significantly increased after treatment with EP-100 and olaparib in comet assay. We performed reverse-phase protein array analyses and identified that the PI3K/AKT pathway was inhibited by EP-100, which we validated with in vitro experiments. In vivo experiment using the HeyA8 mouse model demonstrated that mice treated with EP-100 and olaparib had lower tumor weights (0.06 ± 0.13 g) than those treated with a vehicle (1.19 ± 1.09 g), EP-100 alone (0.62 ± 0.78 g), or olaparib alone (0.50 ± 0.63 g). Our findings indicate that combining EP-100 with olaparib is a promising therapeutic strategy for ovarian cancer.


Asunto(s)
Neoplasias Ováricas/tratamiento farmacológico , Péptidos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Receptores LHRH/genética , Animales , Proteína BRCA1/genética , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Daño del ADN/efectos de los fármacos , Doxorrubicina/farmacología , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Péptidos/síntesis química , Ftalazinas/farmacología , Piperazinas/farmacología , Receptores LHRH/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Mol Cancer Ther ; 18(2): 421-436, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30420565

RESUMEN

Systematic approaches for accurate repurposing of targeted therapies are needed. We developed and aimed to biologically validate our therapy predicting tool (TPT) for the repurposing of targeted therapies for specific tumor types by testing the role of Bromodomain and Extra-Terminal motif inhibitors (BETi) in inhibiting BRD4 function and downregulating Notch3 signaling in ovarian cancer.Utilizing established ovarian cancer preclinical models, we carried out in vitro and in vivo studies with clinically relevant BETis to determine their therapeutic effect and impact on Notch3 signaling.Treatment with BETis or siRNA-mediated BRD4 knockdown resulted in decreased cell viability, reduced cell proliferation, and increased cell apoptosis in vitro. In vivo studies with orthotopic mouse models demonstrated that treatment with BETi decreased tumor growth. In addition, knockdown of BRD4 with doxycycline-inducible shRNA increased survival up to 50% (P < 0.001). Treatment with either BETis or BRD4 siRNA decreased Notch3 expression both in vitro and in vivo BRD4 inhibition also decreased the expression of NOTCH3 targets, including HES1 Chromatin immunoprecipitation revealed that BRD4 was present at the NOTCH3 promoter.Our findings provide biological validation for the TPT by demonstrating that BETis can be an effective therapeutic agent for ovarian cancer by downregulating Notch3 expression.The TPT could rapidly identify candidate drugs for ovarian or other cancers along with novel companion biomarkers.


Asunto(s)
Acetamidas/administración & dosificación , Azepinas/administración & dosificación , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Receptor Notch3/metabolismo , Factores de Transcripción/metabolismo , Acetamidas/farmacología , Animales , Azepinas/farmacología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Mol Cancer Ther ; 18(1): 162-172, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30305341

RESUMEN

For mucinous ovarian cancer (MOC), standard platinum-based therapy is largely ineffective. We sought to identify possible mechanisms of oxaliplatin resistance of MOC and develop strategies to overcome this resistance. A kinome-based siRNA library screen was carried out using human MOC cells to identify novel targets to enhance the efficacy of chemotherapy. In vitro and in vivo validations of antitumor effects were performed using mouse MOC models. Specifically, the role of PRKRA/PACT in oxaliplatin resistance was interrogated. We focused on PRKRA, a known activator of PKR kinase, and its encoded protein PACT because it was one of the five most significantly downregulated genes in the siRNA screen. In orthotopic mouse models of MOC, we observed a significant antitumor effect of PRKRA siRNA plus oxaliplatin. In addition, expression of miR-515-3p was regulated by PACT-Dicer interaction, and miR-515-3p increased the sensitivity of MOC to oxaliplatin. Mechanistically, miR-515-3p regulated chemosensitivity, in part, by targeting AXL. The PRKRA/PACT axis represents an important therapeutic target in MOC to enhance sensitivity to oxaliplatin.


Asunto(s)
Adenocarcinoma Mucinoso/patología , Resistencia a Antineoplásicos , Neoplasias Ováricas/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , ARN Helicasas DEAD-box/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , MicroARNs/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Oxaliplatino , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/farmacología , Proteínas Tirosina Quinasas Receptoras/genética , Ribonucleasa III/metabolismo , Tirosina Quinasa del Receptor Axl
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda