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

Tipo del documento
Intervalo de año de publicación
1.
J Microelectromech Syst ; 29(5): 776-782, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33519169

RESUMEN

We report on isolation, capture, and subsequent elution for analysis of extracellular vesicles derived from human liposarcoma cell conditioned media, using a multi-layer micro-nanofluidic device operated with tangential flow separation. Our device integrates size-based separation followed by immunoaffinity-based capture of extracellular vesicles in the same device. For liposarcomas, this is the first report on isolating, capturing, and then eluting the extracellular vesicles using a micro-nanofluidic device. The results show a significantly higher yield of the eluted extracellular vesicles (~84%) compared to the current methods of ultracentrifugation (~6%) and ExoQuick-based separations (~16%).

2.
Adv Exp Med Biol ; 1226: 73-86, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32030677

RESUMEN

The term "adipose tissue" represents a multicellular and multifunctional organ involved in lipid storage, in hormone and temperature regulation, and in the protection of bones and vital organs from impact-based damage. Emerging evidence now suggests a more malignant role of adipose tissue in promoting cancer onset and progression via the release of secreted factors such as interleukin-6 (IL6) and extracellular vesicles (EVs). These adipose-source factors subsequently affect various aspects of tumorigenesis and/or cancer progression by either directly enhancing the tumor cell oncogenic phenotype or indirectly by the stimulating adjacent normal cells to adopt a more pro-cancer phenotype. Due to the recent growing interest in the role of IL6 and EVs released by adipose tissue in cancer promotion and progression, we are focusing on the protumorigenic impact of fat tissue via IL6 and EV secretion.


Asunto(s)
Tejido Adiposo/metabolismo , Carcinogénesis , Vesículas Extracelulares/metabolismo , Interleucina-6/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral , Humanos
3.
Cancer ; 125(15): 2693-2703, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30980399

RESUMEN

BACKGROUND: Desmoid tumors (DTs) are rare and understudied fibroblastic lesions that are frequently recurrent and locally invasive. DT patients often experience chronic pain, organ dysfunction, decrease in quality of life, and even death. METHODS: Sorafenib has emerged as a promising therapeutic strategy, which has led to the first randomized phase 3 clinical trial devoted to DTs. Concurrently, we conducted a comprehensive analysis of sorafenib efficacy in a large panel of desmoid cell strains to probe for response mechanism. RESULTS: We found distinctive groups of higher- and lower-responder cells. Clustering the lower-responder group, we observed that CTNNB1 mutation was determinant of outcome. Our results revealed that a lower dose of sorafenib was able to inhibit cell viability, migration, and invasion of wild-type and T41A-mutated DTs. Apoptosis induction was observed in those cells after treatment with sorafenib. On the other hand, the lower dose of sorafenib was not able to inhibit cell viability, migration, or invasion or to induce apoptosis in the S45F-mutated DTs. The investigation of autophagy showed the dependency of S45F-mutated DTs on this pathway as a part of cell survival mechanism. Significantly, when autophagy was inhibited genetically or pharmacologically in the S45F mutant cell strains, sensitivity to sorafenib was restored. CONCLUSIONS: Our findings suggest that the response to sorafenib differs when comparing S45F-mutated DTs and T41A-mutated or wild-type DTs. Furthermore, the combination of hydroxychloroquine and sorafenib enhances the antiproliferative and proapoptotic effects in S45F-mutated DT cells, suggesting that profiling ß-catenin status could guide clinical management of desmoid patients who are considering sorafenib treatment.


Asunto(s)
Antineoplásicos/uso terapéutico , Autofagia/efectos de los fármacos , Fibromatosis Agresiva/tratamiento farmacológico , Sorafenib/uso terapéutico , Antineoplásicos/farmacología , Femenino , Humanos , Masculino , Sorafenib/farmacología
4.
Cancer Cell Int ; 18: 89, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29983640

RESUMEN

BACKGROUND: Sarcomas are malignant heterogeneous tumors of mesenchymal derivation. Dedifferentiated liposarcoma (DDLPS) is aggressive with recurrence in 80% and metastasis in 20% of patients. We previously found that miR-133a was significantly underexpressed in liposarcoma tissues. As this miRNA has recently been shown to be a tumor suppressor in many cancers, the objective of this study was to characterize the biological and molecular consequences of miR-133a underexpression in DDLPS. METHODS: Real-time PCR was used to evaluate expression levels of miR-133a in human DDLPS tissue, normal fat tissue, and human DDLPS cell lines. DDLPS cells were stably transduced with miR-133a vector to assess the effects in vitro on proliferation, cell cycle, cell death, migration, and metabolism. A Seahorse Bioanalyzer system was also used to assess metabolism in vivo by measuring glycolysis and oxidative phosphorylation (OXPHOS) in subcutaneous xenograft tumors from immunocompromised mice. RESULTS: miR-133a expression was significantly decreased in human DDLPS tissue and cell lines. Enforced expression of miR-133a decreased cell proliferation, impacted cell cycle progression kinetics, decreased glycolysis, and increased OXPHOS. There was no significant effect on cell death or migration. Using an in vivo xenograft mouse study, we showed that tumors with increased miR-133a expression had no difference in tumor growth compared to control, but did exhibit an increase in OXPHOS metabolic respiration. CONCLUSIONS: Based on our collective findings, we propose that in DDPLS, loss of miR-133a induces a metabolic shift due to a reduction in oxidative metabolism favoring a Warburg effect in DDLPS tumors, but this regulation on metabolism was not sufficient to affect DDPLS.

5.
Cancer ; 121(22): 4088-96, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26349011

RESUMEN

BACKGROUND: Desmoid tumors (DTs) are rare mesenchymal lesions that can recur repeatedly. When it is feasible, DTs are surgically resected; however, this often results in high recurrence rates. Recently, treatment with PF-03084014, a potent γ-secretase inhibitor, has been shown to have antitumor activity in several tumor types by affecting the WNT/ß-catenin pathway. Consequently, Notch pathway inhibition by PF-03084014 might be a promising approach for DT treatment. METHODS: The expression of Notch pathway components was analyzed in DT tissues and cell strains with immunohistochemistry and Western blotting, respectively. A panel of DT cell strains was exposed to PF-03084014 and evaluated for cell proliferation. Antitumor effects were assessed via cell cycle, apoptosis, and migration and invasion analysis. Cells treated with PF-03084014 were characterized with a gene array analysis combined with Ingenuity Pathway Analysis. RESULTS: The results showed that Notch pathway components were expressed at different levels in DTs. Hes1 (Hes Family BHLH Transcription Factor 1) was overexpressed in DT tumors versus dermal scar tissue, and PF-03084014 caused significant decreases in Notch intracellular domain and Hes1 expression in DT cell strains. PF-03084014 decreased DT cell migration and invasion and also caused cell growth inhibition in DT cell strains, most likely through cell cycle arrest. Gene array analysis combined with Ingenuity Pathway Analysis showed that Wnt1-inducible signaling pathway protein 2 possibly regulated Notch and WNT pathways after treatment with PF-03084014 through integrin. CONCLUSION: Our findings suggest that the Notch pathway is an important DT therapeutic target. Furthermore, PF-03084014 has significant antitumor activity against DTs, and it may be an alternative strategy for DT treatment.


Asunto(s)
Fibromatosis Agresiva/tratamiento farmacológico , Receptores Notch/antagonistas & inhibidores , Transducción de Señal/fisiología , Proteínas CCN de Señalización Intercelular/fisiología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Fibromatosis Agresiva/etiología , Fibromatosis Agresiva/patología , Humanos , Invasividad Neoplásica , Receptores Notch/fisiología , Proteínas Represoras/fisiología , Tetrahidronaftalenos/farmacología , Valina/análogos & derivados , Valina/farmacología
6.
J Surg Res ; 178(1): 288-93, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22516345

RESUMEN

Imatinib therapy has undoubtedly contributed to the treatment of metastatic gastrointestinal stromal (GIST) tumors that were previously untreatable. However, disease progression during treatment with tyrosine kinase inhibitors remains an issue in clinical practice not fully explained by KIT and PDGFRA mutation status. We investigated the role of three important signaling molecules (insulin-like growth factor 1 receptor [IGF1R], protein kinase C-θ [PKCθ], and Raf kinase inhibitor protein [RKIP]) that have been implicated in GIST pathogenesis as potential biomarkers for prediction of response to imatinib treatment. We retrospectively reviewed 76 patients with metastatic GIST submitted to imatinib treatment between 2002 and 2007, and analyzed 63 of them. Insulin-like growth factor 1, total PKCθ, phosphorylated PKCθ, and RKIP immunohistochemical expression were correlated with objective response to imatinib treatment and progression-free and overall survival. Median follow-up was 31.2 mo (95% confidence interval, 26.3-36.1 mo). There was a statistically significant association between IGF1R expression and type of response to imatinib treatment (P = 0.05)-that is, higher IGF1R expression was related to lower objective response. However, IGF1R higher expression did not affect progression-free and overall survival. Insulin-like growth factor 1, but not PKCθ and RKIP, emerges as a potential biomarker for prediction of response to imatinib treatment in metastatic GISTs. Validation studies are warranted.


Asunto(s)
Neoplasias Gastrointestinales/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Adulto , Anciano , Antineoplásicos/uso terapéutico , Benzamidas , Femenino , Estudios de Seguimiento , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Tumores del Estroma Gastrointestinal/secundario , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Valor Predictivo de las Pruebas , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas c-kit/genética , Receptor Cross-Talk/efectos de los fármacos , Receptor Cross-Talk/fisiología , Receptor IGF Tipo 1/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Estudios Retrospectivos , Transducción de Señal/fisiología
7.
PLoS One ; 17(10): e0276047, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36240209

RESUMEN

Mutation in the CTNNB1 gene, leading to a deregulation of the WTN/ß-catenin pathway, is a common feature of desmoid tumors (DTs). Many ß-catenin inhibitors have recently been tested in clinical studies; however, BC2059 (also referred as Tegavivint), a selective inhibitor of nuclear ß-catenin that works through binding TBL-1, is the only one being evaluated in a clinical study, specifically for treatment of desmoid tumor patients. Preclinical studies on BC2059 have shown activity in multiple myeloma, acute myeloid leukemia and osteosarcoma. Our preclinical studies provide data on the efficacy of BC2059 in desmoid cell lines, which could help provide insight regarding antitumor activity of this therapy in desmoid tumor patients. In vitro activity of BC2059 was evaluated using desmoid tumor cell lines. Ex vivo activity of BC2059 was assessed using an explant tissue culture model. Pharmacological inhibition of the nuclear ß-catenin activity using BC2059 markedly inhibited cell viability, migration and invasion of mutated DT cells, but with lower effect on wild-type DTs. The decrease in cell viability of mutated DT cells caused by BC2059 was due to apoptosis. Treatment with BC2059 led to a reduction of ß-catenin-associated TBL1 in all mutated DT cells, resulting in a reduction of nuclear ß-catenin. mRNA and protein levels of AXIN2, a ß-catenin target gene, were also found to be downregulated after BC2059 treatment. Taken together, our results demonstrate that nuclear ß-catenin inhibition using BC2059 may be a novel therapeutic strategy for desmoid tumor treatment, especially in patients with CTNNB1 mutation.


Asunto(s)
Neoplasias Óseas , Fibromatosis Agresiva , Fibromatosis Agresiva/patología , Humanos , Mutación , ARN Mensajero/genética , Vía de Señalización Wnt , beta Catenina/metabolismo
8.
J Extracell Vesicles ; 10(4): e12062, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33643547

RESUMEN

We present a resource-efficient approach to fabricate and operate a micro-nanofluidic device that uses cross-flow filtration to isolate and capture liposarcoma derived extracellular vesicles (EVs). The isolated extracellular vesicles were captured using EV-specific protein markers to obtain vesicle enriched media, which was then eluted for further analysis. Therefore, the micro-nanofluidic device integrates the unit operations of size-based separation with CD63 antibody immunoaffinity-based capture of extracellular vesicles in the same device to evaluate EV-cargo content for liposarcoma. The eluted media collected showed ∼76% extracellular vesicle recovery from the liposarcoma cell conditioned media and ∼32% extracellular vesicle recovery from dedifferentiated liposarcoma patient serum when compared against state-of-art extracellular vesicle isolation and subsequent quantification by ultracentrifugation. The results reported here also show a five-fold increase in amount of critical liposarcoma-relevant extracellular vesicle cargo obtained in 30 min presenting a significant advance over existing state-of-art.


Asunto(s)
Vesículas Extracelulares/química , Filtración/métodos , Liposarcoma/química , Nanotecnología/instrumentación , Nanotecnología/métodos , Biomarcadores , Línea Celular Tumoral , Humanos , Neoplasias de Tejido Adiposo/química , Ultracentrifugación/métodos
9.
Oncogene ; 39(34): 5589-5600, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32651460

RESUMEN

Wnt/ß-catenin signaling is one of the key cascades regulating embryogenesis and tissue homeostasis; it has also been intimately associated with carcinogenesis. This pathway is deregulated in several tumors, including colorectal cancer, breast cancer, and desmoid tumors. It has been shown that CTNNB1 exon 3 mutations are associated with an aggressive phenotype in several of these tumor types and may be associated with therapeutic tolerance. Desmoid tumors typically have a stable genome with ß-catenin mutations as a main feature, making these tumors an ideal model to study the changes associated with different types of ß-catenin mutations. Here, we show that the apoptosis mechanism is deregulated in ß-catenin S45F mutants, resulting in decreased induction of apoptosis in these cells. Our findings also demonstrate that RUNX3 plays a pivotal role in the inhibition of apoptosis found in the ß-catenin S45F mutants. Restoration of RUNX3 overcomes this inhibition in the S45F mutants, highlighting it as a potential therapeutic target for malignancies harboring this specific CTNNB1 mutation. While the regulatory effect of RUNX3 in ß-catenin is already known, our results suggest the possibility of a feedback loop involving these two genes, with the CTNNB1 S45F mutation downregulating expression of RUNX3, thus providing additional possible novel therapeutic targets for tumors having deregulated Wnt/ß-catenin signaling induced by this mutation.


Asunto(s)
Neoplasias Abdominales/genética , Poliposis Adenomatosa del Colon/genética , Apoptosis/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Fibromatosis Agresiva/genética , Mutación Missense , Vía de Señalización Wnt/genética , beta Catenina/genética , Neoplasias Abdominales/metabolismo , Neoplasias Abdominales/patología , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Regulación hacia Abajo , Fibromatosis Agresiva/metabolismo , Fibromatosis Agresiva/patología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , beta Catenina/metabolismo
10.
Cancer Res ; 79(19): 4911-4922, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31387924

RESUMEN

Dedifferentiated liposarcoma (DDLPS) is frequently diagnosed late, and patients typically respond poorly to treatments. DDLPS is molecularly characterized by wild-type p53 and amplification of the MDM2 gene, which results in overexpression of MDM2 protein, a key oncogenic process in DDLPS. In this study, we demonstrate that extracellular vesicles derived from patients with DDLPS or from DDLPS cell lines are carriers of MDM2 DNA that can be transferred to preadipocytes, a major and ubiquitous cellular component of the DDLPS tumor microenvironment, leading to impaired p53 activity in preadipocytes and increased proliferation, migration, and production of matrix metalloproteinase 2; treatment with MDM2 inhibitors repressed these effects. Overall, these findings indicate that MDM2 plays a crucial role in DDLPS by enabling cross-talk between tumor cells and the surrounding microenvironment and that targeting vesicular MDM2 could represent a therapeutic option for treating DDLPS. SIGNIFICANCE: Extracellular vesicles derived from dedifferentiated liposarcoma cells induce oncogenic properties in preadipocytes.


Asunto(s)
Adipocitos/metabolismo , Vesículas Extracelulares/metabolismo , Liposarcoma/patología , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Microambiente Tumoral/fisiología , Humanos , Liposarcoma/metabolismo , Células Madre/metabolismo
11.
PLoS One ; 12(11): e0188859, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29186204

RESUMEN

Leiomyosarcoma (LMS) is a malignant soft tissue sarcoma (STS) with a dismal prognosis following metastatic disease. Chemotherapeutic intervention has demonstrated to have modest clinical efficacy with no curative potential in LMS patients. Previously, we demonstrated pan-HDAC inhibition to have a superior effect in various complex karyotypic sarcomas. In this study, our goal is to evaluate the therapeutic efficacy of mocetinostat alone and in combination with gemcitabine in LMS. Human leiomyosarcoma (LMS) cell lines were used for in vitro and in vivo studies. Compounds tested included the class I HDAC inhibitor, mocetinostat, and nucleoside analog, gemcitabine. MTS and clonogenic assays were used to evaluate the effect of mocetinostat on LMS cell growth. Cleaved caspase 3/7 analysis was used to determine the effects of mocetinostat on apoptosis. Compusyn software was used to determine in vitro synergy studies for the combination of mocetinostat plus gemcitabine. A LMS xenograft model in SCID mice was used to test the impact of mocetinostat alone, gemcitabine alone and the combination of mocetinostat plus gemcitabine. Mocetinostat abrogated LMS cell growth and clonogenic potential, and enhanced apoptosis in LMS cell lines. The combination of mocetinostat plus gemcitabine exhibited a synergistic effect in LMS cells in vitro. Similarly, mocetinostat combined with gemcitabine resulted in superior anti-LMS effects in vivo. Mocetinostat reduced the expression of gemcitabine-resistance markers RRM1, RRM2, and increased the expression of gemcitabine-sensitivity marker, hENT1, in LMS cells. LMS are aggressive, metastatic tumors with poor prognosis where effective therapeutic interventions are wanting. Our studies demonstrate the potential utility of mocetinostat combined with gemcitabine for the treatment of LMS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leiomiosarcoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Benzamidas/administración & dosificación , División Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Sinergismo Farmacológico , Humanos , Leiomiosarcoma/patología , Pirimidinas/administración & dosificación , Gemcitabina
12.
Appl Immunohistochem Mol Morphol ; 25(8): 559-565, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-26894649

RESUMEN

Desmoid fibromatosis is a locally aggressive clonal fibroblastic proliferation with high recurrence rates and no metastatic potential. Implicated molecular aberrations occur within the Wnt/ß-catenin pathway (APC and ß-catenin gene mutations). Transforming growth factor-ß (TGF-ß) and connective tissue growth factor (CTGF) are profibrotic growth factors, downstream from nuclear translocation of ß-catenin, that lead to increased fibrogenesis. CTGF (a downstream effector of TGF-ß) is a matricellular protein that modulates the activity of growth factors, adhesion molecules, integrins, and extracellular matrix thus playing a central role in tissue remodeling and fibrosis. Recently there has been growing interest in use of extracellular matrix inhibitors for treatment of various fibrogenic diseases. Desmoid fibromatosis samples (n=15) were evaluated for expression of ß-catenin, TGF-ß, and CTGF using immunohistochemistry on formalin paraffin-embedded material. A control group comprising scar tissue and adjacent normal skin (n=10) were simultaneously immunostained with above mentioned markers. Real-time polymerase chain reaction was performed on frozen specimens of desmoid fibromatosis (n=6) and normal skin (n=2). All 15 desmoid tumors were positive for ß-catenin (surrogate marker of Wnt/ß-catenin pathway dysregulation) which was negative in control normal skin and scar samples. TGF-ß and CTGF were negative in 9 of 10 normal skin controls. TGF-ß and CTGF were positive in all cases of scar tissue. All 15 cases of desmoid tumors were positive for TGF-ß and CTGF. The real-time polymerase chain reaction showed higher expression levels of TGF-ß and CTGF in desmoid fibromatosis compared with normal skin. The high constitutive expression of ß-catenin downstream effectors; TGF-ß, CTGF has the potential for enabling targeted therapy.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibromatosis Agresiva/metabolismo , Mitógenos/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Adulto , Femenino , Fibromatosis Agresiva/patología , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
14.
Cancer Res ; 77(14): 3846-3856, 2017 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-28588009

RESUMEN

Despite the development of combined modality treatments against liposarcoma in recent years, a significant proportion of patients respond only modestly to such approaches, possibly contributing to local or distant recurrence. Early detection of recurrent or metastatic disease could improve patient prognosis by triggering earlier clinical intervention. However, useful biomarkers for such purposes are lacking. Using both patient plasma samples and cell lines, we demonstrate here that miR-25-3p and miR-92a-3p are secreted by liposarcoma cells through extracellular vesicles and may be useful as potential biomarkers of disease. Both miR-25-3p and miR-92a-3p stimulated secretion of proinflammatory cytokine IL6 from tumor-associated macrophages in a TLR7/8-dependent manner, which in turn promoted liposarcoma cell proliferation, invasion, and metastasis via this interaction with the surrounding microenvironment. Our findings provide novel and previously unreported insight into liposarcoma progression, identifying communication between liposarcoma cells and their microenvironment as a process critically involved in liposarcoma progression. This study establishes the possibility that the pattern of circulating miRNAs may identify recurrence prior to radiological detectability while providing insight into disease outcome and as a possible approach to monitor treatment efficacy. Cancer Res; 77(14); 3846-56. ©2017 AACR.


Asunto(s)
Exosomas/metabolismo , Liposarcoma/genética , MicroARNs/metabolismo , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Células HEK293 , Humanos , Liposarcoma/sangre , Liposarcoma/patología , Ratones , Ratones Endogámicos C57BL , Transfección
15.
Clin Cancer Res ; 22(5): 1150-60, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26475335

RESUMEN

PURPOSE: Dedifferentiated liposarcoma (DDLPS) is an aggressive malignancy that can recur locally or disseminate even after multidisciplinary care. Genetically amplified and expressed MDM2, often referred to as a "hallmark" of DDLPS, mostly sustains a wild-type p53 genotype, substantiating the MDM2:p53 axis as a potential therapeutic target for DDLPS. Here, we report on the preclinical effects of SAR405838, a novel and highly selective MDM2 small-molecule inhibitor, in both in vitro and in vivo DDLPS models. EXPERIMENTAL DESIGN: The therapeutic effectiveness of SAR405838 was compared with the known MDM2 antagonists Nutlin-3a and MI-219. The effects of MDM2 inhibition were assessed in both in vitro and in vivo. In vitro and in vivo microarray analyses were performed to assess differentially expressed genes induced by SAR405838, as well as the pathways that these modulated genes enriched. RESULTS: SAR405838 effectively stabilized p53 and activated the p53 pathway, resulting in abrogated cellular proliferation, cell-cycle arrest, and apoptosis. Similar results were observed with Nutlin-3a and MI-219; however, significantly higher concentrations were required. In vitro effectiveness of SAR405838 activity was recapitulated in DDLPS xenograft models where significant decreases in tumorigenicity were observed. Microarray analyses revealed genes enriching the p53 signaling pathway as well as genomic stability and DNA damage following SAR405838 treatment. CONCLUSIONS: SAR405838 is currently in early-phase clinical trials for a number of malignancies, including sarcoma, and our in vitro and in vivo results support its use as a potential therapeutic strategy for the treatment of DDLPS.


Asunto(s)
Indoles/administración & dosificación , Liposarcoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-mdm2/genética , Compuestos de Espiro/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Imidazoles/administración & dosificación , Liposarcoma/genética , Liposarcoma/patología , Ratones , Análisis por Micromatrices , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Piperazinas/administración & dosificación , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 10(7): e0133302, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26200462

RESUMEN

INTRODUCTION: HDAC isoform-specific inhibitors may improve the therapeutic window while limiting toxicities. Developing inhibitors against class I isoforms poses difficulties as they share high homology among their catalytic sites; however, HDAC8 is structurally unique compared to other class I isoforms. HDAC8 inhibitors are novel compounds and have affinity for class I HDAC isoforms demonstrating anti-cancer effects; little is known about their activity in malignant peripheral nerve sheath tumors (MPNST). Recently, we demonstrated anti-MPNST efficacy of HDAC8i in human and murine-derived MPNST pre-clinical models; we now seek to consider the potential therapeutic inhibition of HDAC8 in MPNST. METHODS: Four Human MPNST cell lines, a murine-derived MPNST cell line, and two HDAC8 inhibitors (PCI-34051, PCI-48012; Pharmacyclics, Inc. Sunnyvale, CA) were studied. Proliferation was determined using MTS and clonogenic assays. Effects on cell cycle were determined via PI FACS analysis; effects on apoptosis were determined using Annexin V-PI FACS analysis and cleaved caspase 3 expression. In vivo growth effects of HDAC8i were evaluated using MPNST xenograft models. 2D gel electrophoresis and mass spectrometry were used to identify potential HDAC8 deacetylation substrates. RESULTS: HDAC8i induced cell growth inhibition and marked S-phase cell cycle arrest in human and murine-derived MPNST cells. Relative to control, HDAC8i induced apoptosis in both human and murine-derived MPNST cells. HDAC8i exhibited significant effects on MPNST xenograft growth (p=0.001) and tumor weight (p=0.02). Four potential HDAC8 substrate targets were identified using a proteomic approach: PARK7, HMGB1, PGAM1, PRDX6. CONCLUSIONS: MPNST is an aggressive sarcoma that is notoriously therapy-resistant, hence the urgent need for improved anti-MPNST therapies. HDAC8 inhibition may be useful for MPNST by improving efficacy while limiting toxicities as compared to pan-HDACis.


Asunto(s)
Apoptosis , Histona Desacetilasas/biosíntesis , Proteínas de Neoplasias/biosíntesis , Neurilemoma/enzimología , Proteínas Represoras/biosíntesis , Animales , Línea Celular Tumoral , Femenino , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Humanos , Ratones , Ratones SCID , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neurilemoma/tratamiento farmacológico , Neurilemoma/genética , Neurilemoma/patología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Anticancer Res ; 30(6): 2407-14, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20651400

RESUMEN

BACKGROUND: Prediction of biological behavior is crucial for selection of new therapeutic modalities in GIST. Here, we aimed to assess whether KIT and PDGFRA mutations have survival impact in gastrointestinal stromal tumors (GIST). PATIENTS AND METHODS: Fifty-five Brazilian patients with completely resected GIST were examined for KIT and PDGFRA mutations. The 5-year disease-free survival (DFS) was analyzed. RESULTS: KIT and PDGFRA mutations were identified in 74.5% and 7.3% of patients, respectively. The 5-year DFS rate for all patients was 52.8%. The 5-year DFS rate was lower in patients with tumors having in-frame deletions or concomitant in-frame deletions and insertions affecting codons 557-558 than in patients with tumors having other exon 11 KIT mutations (p=0.023). Conversely, when the patients with concomitant deletion-insertion mutations affecting codons 557-558 were excluded from the analysis, deletions involving codons 557-558 had no influence on 5-year DFS rates. CONCLUSION: Our findings indicate that a specific KIT mutation may be associated with unfavorable behavior in GIST. This finding may have implications on selecting patients for adjuvant therapy.


Asunto(s)
Tumores del Estroma Gastrointestinal/genética , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto , Anciano , Codón , Exones , Femenino , Tumores del Estroma Gastrointestinal/mortalidad , Tumores del Estroma Gastrointestinal/patología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico
18.
São Paulo; s.n; 2015. 113 p. ilus, tab, Quadros.
Tesis en Portugués | LILACS, Inca | ID: biblio-870245

RESUMEN

Os tumores desmóides são proliferações mesenquimais fibroblásticas que, apesar de não originarem metástase, são extremamente invasivos localmente. Esses tumores podem ser esporádicos ou relacionados a síndromes genéticas, como a polipose adenomatosa familiar (FAP). Algumas alterações moleculares já foram relatadas, entretanto, a tumorigênese desses tumores ainda não é totalmente compreendida. Assim, o objetivo desse estudo é analisar e comparar o espectro de mutações somáticas nos tumores desmóides esporádicos e associados a síndromes. Dez tumores desmóides foram analisados por sequenciamento do exoma. O DNA extraído foi utilizado para a construção de uma biblioteca. O DNA fragmentado foi hibridizado com moléculas de RNA correspondentes à região do exoma (50Mb) e posteriormente capturado. As bibliotecas foram amplificadas através de PCR em emulsão e sequenciadas utilizando o SOLiD™ 4 System. Os dados de sequenciamento foram analisados através de programas específicos, e as variantes identificadas somente no tecido tumoral foram selecionadas. A validação de algumas variantes foi realizada por sequenciamento alvo (target sequencing), por ser um método mais sensível através do Ion AmpliSeq e sequenciamento na plataforma Ion PGM.


Desmoid tumors (DTs) are unique mesenchymal fibroblastic proliferationsthat, despite the absolute lack of ability to metastasize, are extremely locallyinvasive. The great majority of DTs occur sporadically, most of these arecaused by somatic mutations in β-catenin (CTNNB1) that lead to aconstitutive activation of WNT pathway. A small number of DTs can occur inthe background of familial adenomatous polyposis (FAP) and are associatedwith inactivating germline mutations in the adenomatous polyposis coli (APC)gene. However, as the genetic profile of desmoid tumors has not beenstudied extensively and remains poorly characterized, the aim of this projectwas to analyze the spectrum of somatic mutations in desmoid tumors. TenDTs were analyzed by massive parallel sequencing (exome). DNA wascaptured by hybridization in solution to cRNA oligonucleotide baits, subjectedto an emulsion PCR, and then sequenced using the SOLiD™ 4 System. Thesequence data were analyzed through specific bioinformatics software andthe variants identified only in tumor were selected for further validation. Forthe sake of higher sensitivity and specificity, target sequencing usingcustomized Ion AmpliSeq™ panels on the Ion PGM™ plataform were used tovalidate the alterations observed using the SOLiD™ 4.


Asunto(s)
Reacción en Cadena de la Polimerasa , Carcinogénesis , Células Madre Mesenquimatosas , Exoma/genética , Fibromatosis Agresiva/genética , Mutación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA