Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Cancer Res ; 83(11): 1883-1904, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37074042

RESUMEN

The EGFR and TGFß signaling pathways are important mediators of tumorigenesis, and cross-talk between them contributes to cancer progression and drug resistance. Therapies capable of simultaneously targeting EGFR and TGFß could help improve patient outcomes across various cancer types. Here, we developed BCA101, an anti-EGFR IgG1 mAb linked to an extracellular domain of human TGFßRII. The TGFß "trap" fused to the light chain in BCA101 did not sterically interfere with its ability to bind EGFR, inhibit cell proliferation, or mediate antibody-dependent cellular cytotoxicity. Functional neutralization of TGFß by BCA101 was demonstrated by several in vitro assays. BCA101 increased production of proinflammatory cytokines and key markers associated with T-cell and natural killer-cell activation, while suppressing VEGF secretion. In addition, BCA101 inhibited differentiation of naïve CD4+ T cells to inducible regulatory T cells (iTreg) more strongly than the anti-EGFR antibody cetuximab. BCA101 localized to tumor tissues in xenograft mouse models with comparable kinetics to cetuximab, both having better tumor tissue retention over TGFß "trap." TGFß in tumors was neutralized by approximately 90% in animals dosed with 10 mg/kg of BCA101 compared with 54% in animals dosed with equimolar TGFßRII-Fc. In patient-derived xenograft mouse models of head and neck squamous cell carcinoma, BCA101 showed durable response after dose cessation. The combination of BCA101 and anti-PD1 antibody improved tumor inhibition in both B16-hEGFR-expressing syngeneic mouse models and in humanized HuNOG-EXL mice bearing human PC-3 xenografts. Together, these results support the clinical development of BCA101 as a monotherapy and in combination with immune checkpoint therapy. SIGNIFICANCE: The bifunctional mAb fusion design of BCA101 targets it to the tumor microenvironment where it inhibits EGFR and neutralizes TGFß to induce immune activation and to suppress tumor growth.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias , Animales , Humanos , Ratones , Anticuerpos Monoclonales Humanizados/uso terapéutico , Carcinoma de Células Escamosas/terapia , Línea Celular Tumoral , Cetuximab/farmacología , Cetuximab/uso terapéutico , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/terapia , Factor de Crecimiento Transformador beta , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias/terapia
2.
Expert Opin Biol Ther ; 14(8): 1145-59, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24762115

RESUMEN

INTRODUCTION: Cancer cells acclimatize to the harsh tumor microenvironment by altering cellular metabolism in favor of aerobic glycolysis. This process provides a source of energy and also generates essential components for macromolecular biosynthesis, which enables cellular survival. As the dependence of cancer cells on glycolysis affects tumorigenesis, it has become an attractive target for therapeutic intervention. Several preclinical studies have shown the effectiveness of using biological targets from the glycolytic pathway for anticancer therapy. AREAS COVERED: This review provides an insight into the glycolytic pathway, highlighting potential targets for glycolytic inhibition. We then discuss recent advancement in delivery strategies that have the potential to circumvent some of the problems posed by current glycolytic inhibitors, enabling resurrection of abandoned therapeutic agents. EXPERT OPINION: Targeting the glycolysis pathway is a tactical approach for cancer therapy. However, the current nonspecific therapeutic strategies have several drawbacks such as poor bioavailability, unfavorable pharmacokinetic profile and associated nonspecific toxicity, thereby limiting preclinical investigation. In recent years, nanoparticle systems have received recognition for the delivery of therapeutic agents directly to the tumor tissue. Thus, it is envisaged that this strategy can be expanded for the delivery of current glycolytic inhibitors specifically to tumor tissues providing improved anticancer activity.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Glucólisis , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Aerobiosis , Animales , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Respiración de la Célula/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Sistemas de Liberación de Medicamentos/tendencias , Glucólisis/efectos de los fármacos , Glucólisis/fisiología , Humanos , Redes y Vías Metabólicas/efectos de los fármacos , Terapia Molecular Dirigida/tendencias
3.
Cancer Lett ; 334(1): 86-94, 2013 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-23111102

RESUMEN

Pancreatic cancer is fourth leading cause of cancer-related deaths in the United States of America. In spite of recent advances in the current therapeutic modalities such as surgery, radiation and chemotherapy patients, the average five year survival rate remains still less than 5%. Recently, compounds from natural sources receive ample of attention as anti-cancer agents. Many epidemiological studies published over the past few decades provide a strong correlation between consumption of vegetables, fruits or plant derived products and reduced incidence of cancer. The present review focuses on the potential antitumor effects of various natural products.


Asunto(s)
Anticarcinógenos/farmacología , Neoplasias Pancreáticas/prevención & control , Fitoquímicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Capsaicina/química , Capsaicina/farmacología , Curcumina/química , Curcumina/farmacología , Humanos , Isotiocianatos/química , Isotiocianatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Resveratrol , Estilbenos/química , Estilbenos/farmacología ,
4.
PLoS One ; 6(5): e20151, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647434

RESUMEN

We evaluated the mechanism of capsaicin-mediated ROS generation in pancreatic cancer cells. The generation of ROS was about 4-6 fold more as compared to control and as early as 1 h after capsaicin treatment in BxPC-3 and AsPC-1 cells but not in normal HPDE-6 cells. The generation of ROS was inhibited by catalase and EUK-134. To delineate the mechanism of ROS generation, enzymatic activities of mitochondrial complex-I and complex-III were determined in the pure mitochondria. Our results shows that capsaicin inhibits about 2.5-9% and 5-20% of complex-I activity and 8-75% of complex-III activity in BxPC-3 and AsPC-1 cells respectively, which was attenuable by SOD, catalase and EUK-134. On the other hand, capsaicin treatment failed to inhibit complex-I or complex-III activities in normal HPDE-6 cells. The ATP levels were drastically suppressed by capsaicin treatment in both BxPC-3 and AsPC-1 cells and attenuated by catalase or EUK-134. Oxidation of mitochondria-specific cardiolipin was substantially higher in capsaicin treated cells. BxPC-3 derived ρ(0) cells, which lack mitochondrial DNA, were completely resistant to capsaicin mediated ROS generation and apoptosis. Our results reveal that the release of cytochrome c and cleavage of both caspase-9 and caspase-3 due to disruption of mitochondrial membrane potential were significantly blocked by catalase and EUK-134 in BxPC-3 cells. Our results further demonstrate that capsaicin treatment not only inhibit the enzymatic activity and expression of SOD, catalase and glutathione peroxidase but also reduce glutathione level. Over-expression of catalase by transient transfection protected the cells from capsaicin-mediated ROS generation and apoptosis. Furthermore, tumors from mice orally fed with 2.5 mg/kg capsaicin show decreased SOD activity and an increase in GSSG/GSH levels as compared to controls. Taken together, our results suggest the involvement of mitochondrial complex-I and III in capsaicin-mediated ROS generation and decrease in antioxidant levels resulting in severe mitochondrial damage leading to apoptosis in pancreatic cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Capsaicina/farmacología , Complejo III de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Estrés Oxidativo/efectos de los fármacos , Neoplasias Pancreáticas/patología , Adenosina Trifosfato/metabolismo , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Capsaicina/antagonistas & inhibidores , Catalasa/metabolismo , Línea Celular Tumoral , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo III de Transporte de Electrones/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Disulfuro de Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Homeostasis/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/metabolismo , Oxidación-Reducción/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
5.
PLoS One ; 6(10): e25799, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22016776

RESUMEN

Our previous studies have shown that benzyl isothiocyanate (BITC) suppresses pancreatic tumor growth by inhibiting STAT-3; however, the exact mechanism of tumor growth suppression was not clear. Here we evaluated the effects and mechanism of BITC on pancreatic tumor angiogenesis. Our results reveal that BITC significantly inhibits neovasularization on rat aorta and Chicken-Chorioallantoic membrane. Furthermore, BITC blocks the migration and invasion of BxPC-3 and PanC-1 pancreatic cancer cells in a dose dependant manner. Moreover, secretion of VEGF and MMP-2 in normoxic and hypoxic BxPC-3 and PanC-1 cells was significantly suppressed by BITC. Both VEGF and MMP-2 play a critical role in angiogenesis and metastasis. Our results reveal that BITC significantly suppresses the phosphorylation of VEGFR-2 (Tyr-1175), and expression of HIF-α. Rho-GTPases, which are regulated by VEGF play a crucial role in pancreatic cancer progression. BITC treatment reduced the expression of RhoC whereas up-regulated the expression of tumor suppressor RhoB. STAT-3 over-expression or IL-6 treatment significantly induced HIF-1α and VEGF expression; however, BITC substantially suppressed STAT-3 as well as STAT-3-induced HIF-1α and VEGF expression. Finally, in vivo tumor growth and matrigel-plug assay show reduced tumor growth and substantial reduction of hemoglobin content in the matrigel plugs and tumors of mice treated orally with 12 µmol BITC, indicating reduced tumor angiogenesis. Immunoblotting of BITC treated tumors show reduced expression of STAT-3 phosphorylation (Tyr-705), HIF-α, VEGFR-2, VEGF, MMP-2, CD31 and RhoC. Taken together, our results suggest that BITC suppresses pancreatic tumor growth by inhibiting tumor angiogenesis through STAT-3-dependant pathway.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Isotiocianatos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Pancreáticas/irrigación sanguínea , Factor de Transcripción STAT3/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Técnicas In Vitro , Isotiocianatos/uso terapéutico , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Ratas , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
6.
Clin Cancer Res ; 17(7): 1784-95, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21350002

RESUMEN

PURPOSE: Our previous studies have shown that benzyl isothiocyanate (BITC) suppress pancreatic cancer growth by inducing apoptosis but the molecular mechanism was unclear. In this study we hypothesized the involvement of PI3K/AKT/FOXO pathway in BITC-induced apoptosis. EXPERIMENTAL DESIGN: Mice were implanted BxPC-3 tumor xenografts and orally gavaged with 12 µmol BITC. Plasma and tumor BITC concentration was estimated by liquid chromatography/tandem mass spectrometry. BxPC-3 and PanC-1 cells were used to elucidate PI3K/AKT/FOXO pathway. Electrophoretic mobility shift assay (EMSA), DNA binding activity, immunofluorescence, and gene transfection were used to delineate the mechanism. RESULTS: BITC-treated mice showed 43% less tumor growth as compared with control mice and correlated well with the therapeutic concentrations of 6.5 µmol/L BITC achieved in plasma and 7.5 µmol/g BITC in tumor tissue. Western blot analyses and immunohistochemistry revealed that tumors from BITC-treated mice showed reduced phosphorylation of PI3K, AKT, PDK1, mTOR, FOXO1, and FOXO3a and increased apoptosis. Complementing our in vivo results, we made similar observations in a dose- and time-dependent manner in BITC-treated BxPC-3 and Panc-1 cells. Binding of FOXO1 with 14-3-3 proteins was also reduced drastically by BITC treatment indicating nuclear retention of FOXO1 and this observation was further confirmed with EMSA, immunofluorescence, DNA binding, and upregulation of FOXO-responsive proteins Bim, p27, and p21 in BxPC-3 cells. Overexpression of AKT by transient transfection significantly blocked the modulation of FOXO proteins and protected the cells from BITC-mediated apoptosis and growth suppression. CONCLUSIONS: Our results provide convincing evidence on the involvement of PI3K/AKT/FOXO pathway in BITC-mediated pancreatic tumor growth suppression.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción Forkhead/metabolismo , Isotiocianatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Acetilación , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Proteína de Unión a CREB/metabolismo , Caspasa 3/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Humanos , Quinasa I-kappa B/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Sirtuinas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Trasplante Heterólogo , Carga Tumoral/efectos de los fármacos , Factores de Transcripción p300-CBP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA