Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 23(1): 775, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596538

RESUMEN

BACKGROUND: Inhibitors of Poly (ADP-Ribose) Polymerases (PARP) provide clinical benefit to patients with breast and ovarian cancers, by compromising the DNA repair activity of cancer cells. Although these agents extend progression-free survival in many patients, responses can be short lived with many patients ultimately progressing. Identification of combination partners that increase dependence of cancer cells to the DNA repair activity of PARPs may represent a strategy to increase the utility of PARP inhibitors. Protein arginine methyltransferase 5 (PRMT5) regulates DNA damage response pathways through splicing and protein modification, and inhibitors of PRMT5 have recently entered clinical trials. METHODS: The effect of PRMT5 inhibition on the levels of DNA damage and repair markers including γH2AX, RAD51, and 53BP1 was determined using high content immunofluorescent imaging. The anti-proliferative activity of the combination of PRMT5 and PARP inhibitors was evaluated using in vitro models of breast and ovarian cancers using both cell lines and ex vivo patient derived xenografts. Finally, the combinations of PRMT5 and PARP inhibitors were evaluated in cell line xenograft models in vivo. RESULTS: Inhibition of PRMT5 by GSK3326595 led to increased levels of markers of DNA damage. The addition of GSK3326595 to the PARP inhibitor, niraparib, resulted in increased growth inhibition of breast and ovarian cancer cell lines and patient derived spheroids. In vivo, the combination improved the partial effects on tumor growth inhibition achieved by either single agent, producing complete tumor stasis and regression. CONCLUSION: These data demonstrate that inhibition of PRMT5 induced signatures of DNA damage in models of breast and ovarian cancer. Furthermore, combination with the PARP inhibitor, Niraparib, resulted in increased anti-tumor activity in vitro and in vivo. Overall, these data suggest inhibition of PRMT5 as a mechanism to broaden and enhance the clinical application of PARP inhibitors.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Humanos , Femenino , Animales , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Daño del ADN , Modelos Animales de Enfermedad , Proteína-Arginina N-Metiltransferasas
2.
Ir J Med Sci ; 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38744793

RESUMEN

PURPOSE: Chronic Obstructive Lung Disease (COPD) remains a major cause of morbidity and mortality across the world. We evaluated survival over 9 years in a cohort of patients with COPD requiring acute inpatient non-invasive ventilation (NIV). We analyzed prognostic indices to evaluate if they were associated with mortality. PATIENTS AND METHODS: We performed a retrospective chart review of all patients who were admitted to St. James's Hospital respiratory ward with COPD and acute hypercapnic respiratory failure who required NIV over a 12-month period and followed their outcomes over 9 years. We investigated the association between survival and potential prognostic variables using univariate analysis and multivariate Cox proportional hazards model. We evaluated the association between survival and the following parameters: age, gender, multiple admissions requiring NIV (> 1 admission in within 12 months of index presentation), home NIV use preadmission, initial arterial blood gas pH, days spent on NIV, serum albumin and serum albumin to serum CRP ratio at admission. RESULTS: Ninety-nine patients with COPD and acute hypercapnic respiratory failure were identified over a 12-month period from January to December 2011. Survival at 1, 2, 5 and 9 years was 65% (n = 64), 42% (n = 42), 25% (n = 25) and 21% (n = 21), respectively. Increasing age (p value < 0.001) and a lower serum albumin (p value < 0.005) were associated with a higher mortality. There was a trend towards improved survival in the group who were treated with home NIV prior to admission compared to no NIV therapy at home but this did not reach statistical significance (Fig. 3, p value = 0.088). CONCLUSION: This study highlights the long-term mortality in patients with COPD admitted with hypercapnic respiratory failure requiring NIV and correlates with prior studies. Increasing age and lower serum albumin were associated with increased mortality. Home NIV may have a protective long-term survival benefit in patients with COPD who have been admitted for acute NIV.

3.
Cancer Immunol Res ; 10(4): 420-436, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35181787

RESUMEN

Protein arginine methyltransferases (PRMT) are a widely expressed class of enzymes responsible for catalyzing arginine methylation on numerous protein substrates. Among them, type I PRMTs are responsible for generating asymmetric dimethylarginine. By controlling multiple basic cellular processes, such as DNA damage responses, transcriptional regulation, and mRNA splicing, type I PRMTs contribute to cancer initiation and progression. A type I PRMT inhibitor, GSK3368715, has been developed and has entered clinical trials for solid and hematologic malignancies. Although type I PRMTs have been reported to play roles in modulating immune cell function, the immunologic role of tumor-intrinsic pathways controlled by type I PRMTs remains uncharacterized. Here, our The Cancer Genome Atlas dataset analysis revealed that expression of type I PRMTs associated with poor clinical response and decreased immune infiltration in patients with melanoma. In cancer cell lines, inhibition of type I PRMTs induced an IFN gene signature, amplified responses to IFN and innate immune signaling, and decreased expression of the immunosuppressive cytokine VEGF. In immunocompetent mouse tumor models, including a model of T-cell exclusion that represents a common mechanism of anti-programmed cell death protein 1 (PD-1) resistance in humans, type I PRMT inhibition increased T-cell infiltration, produced durable responses dependent on CD8+ T cells, and enhanced efficacy of anti-PD-1 therapy. These data indicate that type I PRMT inhibition exhibits immunomodulatory properties and synergizes with immune checkpoint blockade (ICB) to induce durable antitumor responses in a T cell-dependent manner, suggesting that type I PRMT inhibition can potentiate an antitumor immunity in refractory settings.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Proteína-Arginina N-Metiltransferasas , Animales , Arginina , Humanos , Inmunidad , Ratones , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo
4.
J Nanosci Nanotechnol ; 11(9): 8354-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22097583

RESUMEN

In comparison to ITO films prepared by chemical solution deposition on bare substrates, the use of a ZnO buffer layer and Al2O3 barrier layer has been shown to have a significant effect on morphology, measured sheet resistance and therefore resistivity. In the case of quartz substrates, ITO resistivity decreased from 9.6 x 10(-3) ohms cm to 4.3 x 10(-3) ohms cm on incorporation of a ZnO buffer layer and Al2O3 barrier layer, both grown by ALD. A change in surface morphology was observed, due to the presence of the buffer layer, however, the ZnO buffer layer was not found to influence the XRD pattern of the ITO films.

5.
Phys Sportsmed ; 49(1): 45-50, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32326866

RESUMEN

Objectives: Falls and injuries are frequent in professional horseracing. However, professional jockeys spend a large part of their week in horse-related activities outside of racing such as schooling, exercise riding, and yard-related activities. The injury risk related to these activities remains largely unknown internationally. This study aimed to identify the injury prevalence and injury profile of flat and jump jockeys during non-racing activities. Methods: In total 45.6% and 38.5% of all licensed Irish professional flat and jump jockeys completed a cross-sectional self-recall questionnaire examining injuries that occurred outside of racing during 2018. Injury proportion, repeat incidence proportion, and descriptive statistics were calculated. Results: Fifteen percent of professional jockeys sustained an injury outside of a race, half of those injured receiving at least another injury in 2018 and 66.52 injuries per 1,000 falls were noted. Injuries frequently occurred to the upper limb (36.67%), with fractures common (32.00%). Most injuries occurred due to a fall (60.00%) and 77.27% occurred in the gallops. Half of injuries resulted in the jockey missing racing, with 31.00 ± 47.18 (4-180) days lost on average. Twenty-three percent of jockeys attended hospital and 16.67% required surgery due to injury. Interestingly, just under a third did not report their injury to anyone. Conclusion: Injuries to professional jockeys, whilst not as frequent outside of racing, tend to be serious and can affect jockeys financially and impact their availability to ride. Prioritizing injury prevention strategies to maximize availability of jockeys to race is important. Education on the importance of reporting all injuries regardless of where they occur is important to ensure their management and rehabilitation.


Asunto(s)
Accidentes de Trabajo/prevención & control , Accidentes de Trabajo/estadística & datos numéricos , Traumatismos en Atletas/epidemiología , Traumatismos en Atletas/prevención & control , Caballos , Accidentes por Caídas/prevención & control , Accidentes por Caídas/estadística & datos numéricos , Animales , Estudios Transversales , Femenino , Fracturas Óseas/epidemiología , Humanos , Incidencia , Masculino , Prevalencia , Factores de Riesgo , Encuestas y Cuestionarios , Reino Unido/epidemiología
6.
Sci Signal ; 13(619)2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32071169

RESUMEN

High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecological cancer with few effective, targeted therapies. HGSOC tumors exhibit genomic instability with frequent alterations in the protein kinome; however, only a small fraction of the kinome has been therapeutically targeted in HGSOC. Using multiplexed inhibitor beads and mass spectrometry, we mapped the kinome landscape of HGSOC tumors from patients and patient-derived xenograft models. The data revealed a prevalent signature consisting of established HGSOC driver kinases, as well as several kinases previously unexplored in HGSOC. Loss-of-function analysis of these kinases in HGSOC cells indicated MRCKA (also known as CDC42BPA) as a putative therapeutic target. Characterization of the effects of MRCKA knockdown in established HGSOC cell lines demonstrated that MRCKA was integral to signaling that regulated the cell cycle checkpoint, focal adhesion, and actin remodeling, as well as cell migration, proliferation, and survival. Moreover, inhibition of MRCKA using the small-molecule BDP9066 decreased cell proliferation and spheroid formation and induced apoptosis in HGSOC cells, suggesting that MRCKA may be a promising therapeutic target for the treatment of HGSOC.


Asunto(s)
Biomarcadores de Tumor/antagonistas & inhibidores , Cistadenocarcinoma Seroso/tratamiento farmacológico , Proteína Quinasa de Distrofia Miotónica/antagonistas & inhibidores , Neoplasias Ováricas/tratamiento farmacológico , Proteómica/métodos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Espectrometría de Masas/métodos , Terapia Molecular Dirigida/métodos , Proteína Quinasa de Distrofia Miotónica/genética , Proteína Quinasa de Distrofia Miotónica/metabolismo , Clasificación del Tumor , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
7.
Cancer Biol Ther ; 20(7): 1035-1045, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30929564

RESUMEN

Pharmacological inhibition of PARP is a promising approach in treating high grade serous ovarian carcinoma (HGSOC). PARP inhibitors (PARPi) are most active in patients with defects in DNA damage repair (DDR) mechanisms, such as alterations in expression/function of DNA repair and homologous recombination (HR) genes/proteins, including BRCA1 and BRCA2. Benefit of PARPi could be extended towards HR-proficient patients by combining PARPi with agents that functionally abrogate HR. An attractive molecular target for this purpose is heat shock protein 90 (HSP90), which mediates the maturation and stability of several key proteins required for DDR. Here, we tested the hypothesis that targeted inhibition of HSP90 with a small-molecule inhibitor ganetespib would sensitize non-BRCA mutant ovarian carcinoma (OC) cells to PARP inhibition by talazoparib. We used commercially available cell lines, along with several novel HGSOC OC cell lines established in our laboratory. Ganetespib treatment destabilized HSP90 client proteins involved in DNA damage response and cell cycle checkpoint, and disrupted γ-irradiation-induced DNA repair. The effects of the combination of ganetespib and talazoparib on OC cell viability and survival were also analyzed, and among the non-BRCA mutant cell lines analyzed, the combination was synergistic in several cell lines (OVCAR-3, OC-1, OC-16). Together, our data suggest that ganetespib-mediated inhibition of HSP90 effectively disrupts critical DDR pathway proteins and may sensitize OC cells without 'BRCAness' to PARPi. From a clinical perspective, this suggests that HSP90 inhibition has the potential to sensitize some HGSOC patients without HR pathway alterations to PARPi, and potentially other DNA-damage inducing agents.


Asunto(s)
Daño del ADN , Resistencia a Antineoplásicos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Proteínas HSP90 de Choque Térmico/genética , Recombinación Homóloga , Humanos , Inmunohistoquímica , Ratones , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Ftalazinas/farmacología , Radiación Ionizante , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Cell ; 36(1): 100-114.e25, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31257072

RESUMEN

Type I protein arginine methyltransferases (PRMTs) catalyze asymmetric dimethylation of arginines on proteins. Type I PRMTs and their substrates have been implicated in human cancers, suggesting inhibition of type I PRMTs may offer a therapeutic approach for oncology. The current report describes GSK3368715 (EPZ019997), a potent, reversible type I PRMT inhibitor with anti-tumor effects in human cancer models. Inhibition of PRMT5, the predominant type II PRMT, produces synergistic cancer cell growth inhibition when combined with GSK3368715. Interestingly, deletion of the methylthioadenosine phosphorylase gene (MTAP) results in accumulation of the metabolite 2-methylthioadenosine, an endogenous inhibitor of PRMT5, and correlates with sensitivity to GSK3368715 in cell lines. These data provide rationale to explore MTAP status as a biomarker strategy for patient selection.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/deficiencia , Empalme Alternativo , Antineoplásicos/química , Biomarcadores , Línea Celular Tumoral , Sinergismo Farmacológico , Inhibidores Enzimáticos/química , Humanos , Metilación , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Unión Proteica , Proteína-Arginina N-Metiltransferasas/química , Especificidad por Sustrato
10.
Oncogene ; 37(35): 4854-4870, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29773902

RESUMEN

Neural precursor cell expressed, developmentally downregulated 9 (NEDD9) supports oncogenic signaling in a number of solid and hematologic tumors. Little is known about the role of NEDD9 in ovarian carcinoma (OC), but available data suggest elevated mRNA and protein expression in advanced stage high-grade cancers. We used a transgenic MISIIR-TAg mouse OC model combined with genetic ablation of Nedd9 to investigate its action in the development and progression of OC. A Nedd9-/- genotype delayed tumor growth rate, reduced incidence of ascites, and reduced expression and activation of signaling proteins including SRC, STAT3, E-cadherin, and AURKA. Cell lines established from MISIIR-TAg;Nedd9-/- and MISIIR-TAg;Nedd9+/+ mice exhibited altered migration and invasion. Growth of these cells in a syngeneic allograft model indicated that systemic Nedd9 loss in the microenvironment had little impact on tumor allograft growth, but in a Nedd9 wild-type background Nedd9-/- allografts exhibited significantly reduced growth, dissemination, and oncogenic signaling compared to Nedd9+/+ allografts. Gene expression analysis revealed that Nedd9+/+ tumors exhibited more mesenchymal "stem-like" transcriptional program, including increased expression of Aldh1a1 and Aldh1a2. Conversely, loss of Nedd9 resulted in increased expression of differentiation genes, including fallopian tube markers Foxj1, Ovgp1, and Pax8. Collectively, these data suggest that tumor cell-intrinsic Nedd9 expression promotes OC development and progression by broad induction of oncogenic protein signaling and stem/mesenchymal gene expression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Madre Mesenquimatosas/metabolismo , Neoplasias Ováricas/genética , Fosfoproteínas/genética , Transducción de Señal/genética , Animales , Cadherinas/genética , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Humanos , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Oncogenes/genética , Neoplasias Ováricas/patología , Transcripción Genética/genética
11.
Cancer Biol Ther ; 19(7): 554-564, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29737910

RESUMEN

Ovarian cancer ranks fifth in cancer related deaths for women in USA. The high mortality rate associated with ovarian cancer is due to diagnosis at later stages of disease and the high recurrence rate of 60-80%. Recurrent ovarian cancers are more likely to present as multidrug resistance (MDR) leading to unfavorable response from 2nd and 3rd line chemotherapy. Nanoemulsions (NEs) are emerging as an attractive drug delivery system to overcome MDR challenges. NEs can also minimize exposure of therapeutic cargo to normal tissues potentially reducing side effects. In >80% of ovarian cancers, Folate Receptor-α (FR-α) is expressed at 10- to 100-fold higher levels than on non-pathological tissues. Therefore, folate (FA) is being evaluated as an active targeting moiety for FR-α+ ovarian cancer. To improve therapeutic outcome with reduced toxicity, we developed NMI-500, a FA targeted gadolinium (Gd) annotated NE loaded with docetaxel (DTX). NMI-500 has been developed as theranostic agents as Gd will enable physician to acquire real time pharmacodynamics data on NE + DTX accumulation in target lesions. In present study, characterization for key translational metrics of NMI-500 showed size distribution in range of 120 to 150 nm and zeta potential around -45 mV. Active targeting of FA was evaluated against FR-α+ KB cells and results demonstrated significant improvement in cell association which was surface ligand density dependent. We found that NMI-500 was able to inhibit tumor growth in a spontaneous transgenic ovarian cancer model with improved safety profile and this growth inhibition could be longitudinally followed by MRI. These results indicate NMI-500 warrants advancement to clinical trials.


Asunto(s)
Antineoplásicos/administración & dosificación , Docetaxel/administración & dosificación , Portadores de Fármacos/química , Receptor 1 de Folato/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Docetaxel/farmacocinética , Portadores de Fármacos/farmacología , Emulsiones , Endocitosis , Femenino , Ácido Fólico/metabolismo , Gadolinio/química , Gadolinio/farmacología , Humanos , Imagen por Resonancia Magnética/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Imagen Molecular/métodos , Nanopartículas/química , Recurrencia Local de Neoplasia , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Nanomedicina Teranóstica/métodos
12.
Clin Cancer Res ; 23(2): 441-453, 2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27435394

RESUMEN

PURPOSE: To define the safety and effectiveness of T cells redirected against follicle-stimulating hormone receptor (FSHR)-expressing ovarian cancer cells. EXPERIMENTAL DESIGN: FSHR expression was determined by Western blotting, immunohistochemistry, and qPCR in 77 human ovarian cancer specimens from 6 different histologic subtypes and 20 human healthy tissues. The effectiveness of human T cells targeted with full-length FSH in vivo was determined against a panel of patient-derived xenografts. Safety and effectiveness were confirmed in immunocompetent tumor-bearing mice, using constructs targeting murine FSHR and syngeneic T cells. RESULTS: FSHR is expressed in gynecologic malignancies of different histologic types but not in nonovarian healthy tissues. Accordingly, T cells expressing full-length FSHR-redirected chimeric receptors mediate significant therapeutic effects (including tumor rejection) against a panel of patient-derived tumors in vivo In immunocompetent mice growing syngeneic, orthotopic, and aggressive ovarian tumors, fully murine FSHR-targeted T cells also increased survival without any measurable toxicity. Notably, chimeric receptors enhanced the ability of endogenous tumor-reactive T cells to abrogate malignant progression upon adoptive transfer into naïve recipients subsequently challenged with the same tumor. Interestingly, FSHR-targeted T cells persisted as memory lymphocytes without noticeable PD-1-dependent exhaustion during end-stage disease, in the absence of tumor cell immunoediting. However, exosomes in advanced tumor ascites diverted the effector activity of this and other chimeric receptor-transduced T cells away from targeted tumor cells. CONCLUSIONS: T cells redirected against FSHR+ tumor cells with full-length FSH represent a promising therapeutic alternative against a broad range of ovarian malignancies, with negligible toxicity even in the presence of cognate targets in tumor-free ovaries. Clin Cancer Res; 23(2); 441-53. ©2016 AACR.


Asunto(s)
Inmunoterapia , Neoplasias Ováricas/terapia , Receptores de HFE/inmunología , Linfocitos T/inmunología , Animales , Ascitis/inmunología , Ascitis/patología , Exosomas/inmunología , Exosomas/patología , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunohistoquímica , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de HFE/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Rep ; 16(5): 1273-1286, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27452461

RESUMEN

Small-molecule BET bromodomain inhibitors (BETis) are actively being pursued in clinical trials for the treatment of a variety of cancers, but the mechanisms of resistance to BETis remain poorly understood. Using a mass spectrometry approach that globally measures kinase signaling at the proteomic level, we evaluated the response of the kinome to targeted BETi treatment in a panel of BRD4-dependent ovarian carcinoma (OC) cell lines. Despite initial inhibitory effects of BETi, OC cells acquired resistance following sustained treatment with the BETi JQ1. Through application of multiplexed inhibitor beads (MIBs) and mass spectrometry, we demonstrate that BETi resistance is mediated by adaptive kinome reprogramming, where activation of compensatory pro-survival kinase networks overcomes BET protein inhibition. Furthermore, drug combinations blocking these kinases may prevent or delay the development of drug resistance and enhance the efficacy of BETi therapy.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Resistencia a Antineoplásicos/fisiología , Femenino , Humanos , Proteómica/métodos , Transducción de Señal/fisiología
14.
Cancer Res ; 76(9): 2778-90, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197267

RESUMEN

Breast and ovarian cancer patients harboring BRCA1/2 germline mutations have clinically benefitted from therapy with PARP inhibitor (PARPi) or platinum compounds, but acquired resistance limits clinical impact. In this study, we investigated the impact of mutations on BRCA1 isoform expression and therapeutic response. Cancer cell lines and tumors harboring mutations in exon 11 of BRCA1 express a BRCA1-Δ11q splice variant lacking the majority of exon 11. The introduction of frameshift mutations to exon 11 resulted in nonsense-mediated mRNA decay of full-length, but not the BRCA1-Δ11q isoform. CRISPR/Cas9 gene editing as well as overexpression experiments revealed that the BRCA1-Δ11q protein was capable of promoting partial PARPi and cisplatin resistance relative to full-length BRCA1, both in vitro and in vivo Furthermore, spliceosome inhibitors reduced BRCA1-Δ11q levels and sensitized cells carrying exon 11 mutations to PARPi treatment. Taken together, our results provided evidence that cancer cells employ a strategy to remove deleterious germline BRCA1 mutations through alternative mRNA splicing, giving rise to isoforms that retain residual activity and contribute to therapeutic resistance. Cancer Res; 76(9); 2778-90. ©2016 AACR.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Neoplasias Ováricas/genética , Empalme Alternativo/genética , Animales , Proteína BRCA1/metabolismo , Western Blotting , Cisplatino/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Mutación de Línea Germinal , Humanos , Inmunohistoquímica , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Reacción en Cadena de la Polimerasa , Isoformas de Proteínas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cancer Ther ; 14(4): 1035-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25646015

RESUMEN

Ovarian carcinoma is the fifth leading cause of death among women in the United States. Persistent activation of STAT3 is frequently detected in ovarian carcinoma. STAT3 is activated by Janus family kinases (JAK) via cytokine receptors, growth factor receptor, and non-growth factor receptor tyrosine kinases. Activation of STAT3 mediates tumor cell proliferation, survival, motility, invasion, and angiogenesis, and recent work demonstrates that STAT3 activation suppresses antitumor immune responses and supports tumor-promoting inflammation. We hypothesized that therapeutic targeting of the JAK/STAT3 pathway would inhibit tumor growth by direct effects on ovarian carcinoma cells and by inhibition of cells in the tumor microenvironment (TME). To test this, we evaluated the effects of a small-molecule JAK inhibitor, AZD1480, on cell viability, apoptosis, proliferation, migration, and adhesion of ovarian carcinoma cells in vitro. We then evaluated the effects of AZD1480 on in vivo tumor growth and progression, gene expression, tumor-associated matrix metalloproteinase (MMP) activity, and immune cell populations in a transgenic mouse model of ovarian carcinoma. AZD1480 treatment inhibited STAT3 phosphorylation and DNA binding, and migration and adhesion of cultured ovarian carcinoma cells and ovarian tumor growth rate, volume, and ascites production in mice. In addition, drug treatment led to altered gene expression, decreased tumor-associated MMP activity, and fewer suppressor T cells in the peritoneal TME of tumor-bearing mice than control mice. Taken together, our results show pharmacologic inhibition of the JAK2/STAT3 pathway leads to disruption of functions essential for ovarian tumor growth and progression and represents a promising therapeutic strategy.


Asunto(s)
Analgésicos/farmacología , Quinasas Janus/metabolismo , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Analgésicos/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Análisis por Conglomerados , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Integrina alfaVbeta3/genética , Integrina alfaVbeta3/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Pirazoles/administración & dosificación , Pirazoles/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Clin Cancer Res ; 19(18): 5053-67, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23900136

RESUMEN

PURPOSE: Epithelial ovarian cancer (EOC) is usually detected at an advanced stage and is frequently lethal. Although many patients respond to initial surgery and standard chemotherapy consisting of a platinum-based agent and a taxane, most experience recurrence and eventually treatment-resistant disease. Although there have been numerous efforts to apply protein-targeted agents in EOC, these studies have so far documented little efficacy. Our goal was to identify broadly susceptible signaling proteins or pathways in EOC. EXPERIMENTAL DESIGN: As a new approach, we conducted data-mining meta-analyses integrating results from multiple siRNA screens to identify gene targets that showed significant inhibition of cell growth. On the basis of this meta-analysis, we established that many genes with such activity were clients of the protein chaperone HSP90. We therefore assessed ganetespib, a clinically promising second-generation small-molecule HSP90 inhibitor, for activity against EOC, both as a single agent and in combination with cytotoxic and targeted therapeutic agents. RESULTS: Ganetespib significantly reduced cell growth, induced cell-cycle arrest and apoptosis in vitro, inhibited growth of orthotopic xenografts and spontaneous ovarian tumors in transgenic mice in vivo, and inhibited expression and activation of numerous proteins linked to EOC progression. Importantly, paclitaxel significantly potentiated ganetespib activity in cultured cells and tumors. Moreover, combined treatment of cells with ganetespib and siRNAs or small molecules inhibiting genes identified in the meta-analysis in several cases resulted in enhanced activity. CONCLUSION: These results strongly support investigation of ganetespib, a single-targeted agent with effects on numerous proteins and pathways, in augmenting standard EOC therapies.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Neoplasias Ováricas/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Sinergismo Farmacológico , Quimioterapia Combinada , Femenino , Citometría de Flujo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones SCID , Ratones Transgénicos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Paclitaxel/farmacología , Análisis por Matrices de Proteínas , ARN Interferente Pequeño/genética , Triazoles/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Neoplasia ; 14(6): 451-62, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22787427

RESUMEN

Most patients with epithelial ovarian cancer (EOC) experience drug-resistant disease recurrence. Identification of new treatments is a high priority, and preclinical studies in mouse models of EOC may expedite this goal. We previously developed methods for magnetic resonance imaging (MRI) for tumor detection and quantification in a transgenic mouse model of EOC. The goal of this study was to determine whether three-dimensional (3D) fluorescence molecular tomography (FMT) and fluorescent molecular imaging probes could be effectively used for in vivo detection of ovarian tumors and response to therapy. Ovarian tumor-bearing TgMISIIR-TAg mice injected with fluorescent probes were subjected to MRI and FMT. Tumor-specific probe retention was identified in vivo by alignment of the 3D data sets, confirmed by ex vivo fluorescent imaging and correlated with histopathologic findings. Mice were treated with standard chemotherapy, and changes in fluorescent probe binding were detected by MRI and FMT. Ovarian tumors were detected using probes specific for cathepsin proteases, matrix metalloproteinases (MMPs), and integrin α(v)ß(3). Cathepsin and integrin α(v)ß(3) probe activation and retention correlated strongly with tumor volume. MMP probe activation was readily detected in tumors but correlated less strongly with tumor volume. Tumor regression associated with response to therapy was detected and quantified by serial MRI and FMT. These results demonstrate the feasibility and sensitivity of FMT for detection and quantification of tumor-associated biologic targets in ovarian tumors and support the translational utility of molecular imaging to assess functional response to therapy in mouse models of EOC.


Asunto(s)
Carcinoma/diagnóstico , Carcinoma/metabolismo , Integrinas/metabolismo , Imagen Molecular , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/metabolismo , Péptido Hidrolasas/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Carcinoma/patología , Catepsinas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Integrina alfaVbeta3/metabolismo , Integrinas/genética , Imagen por Resonancia Magnética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Ováricas/tratamiento farmacológico , Unión Proteica , Carga Tumoral/efectos de los fármacos
18.
PLoS One ; 6(10): e23666, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21984891

RESUMEN

Niemann-Pick Disease, type C (NPC) is a fatal, neurodegenerative, lysosomal storage disorder. It is a rare disease with broad phenotypic spectrum and variable age of onset. These issues make it difficult to develop a universally accepted clinical outcome measure to assess urgently needed therapies. To this end, clinical investigators have defined emerging, disease severity scales. The average time from initial symptom to diagnosis is approximately 4 years. Further, some patients may not travel to specialized clinical centers even after diagnosis. We were therefore interested in investigating whether appropriately trained, community-based assessment of patient records could assist in defining disease progression using clinical severity scores. In this study we evolved a secure, step wise process to show that pre-existing medical records may be correctly assessed by non-clinical practitioners trained to quantify disease progression. Sixty-four undergraduate students at the University of Notre Dame were expertly trained in clinical disease assessment and recognition of major and minor symptoms of NPC. Seven clinical records, randomly selected from a total of thirty seven used to establish a leading clinical severity scale, were correctly assessed to show expected characteristics of linear disease progression. Student assessment of two new records donated by NPC families to our study also revealed linear progression of disease, but both showed accelerated disease progression, relative to the current severity scale, especially at the later stages. Together, these data suggest that college students may be trained in assessment of patient records, and thus provide insight into the natural history of a disease.


Asunto(s)
Aptitud , Progresión de la Enfermedad , Educación de Pregrado en Medicina , Evaluación Educacional , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Enfermedad de Niemann-Pick Tipo C/patología , Estudiantes , Humanos , Registros Médicos , Estaciones del Año , Índice de Severidad de la Enfermedad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA