Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Res ; 79(9): 2327-2338, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31043430

RESUMO

When pancreatic cancer cannot be removed surgically, patients frequently experience morbidity and death from progression of their primary tumor. Radiation therapy (RT) cannot yet substitute for an operation because radiation causes fatal bleeding and ulceration of the nearby stomach and intestines before achieving tumor control. There are no FDA-approved medications that prevent or reduce radiation-induced gastrointestinal injury. Here, we overcome this fundamental problem of anatomy and biology with the use of the oral EGLN inhibitor FG-4592, which selectively protects the intestinal tract from radiation toxicity without protecting tumors. A total of 70 KPC mice with autochthonous pancreatic tumors received oral FG-4592 or vehicle control ± ablative RT to a cumulative 75 Gy administered in 15 daily fractions to a limited tumor field. Although ablative RT reduced complications from local tumor progression, fatal gastrointestinal bleeding was observed in 56% of mice that received high-dose RT with vehicle control. However, radiation-induced bleeding was completely ameliorated in mice that received high-dose RT with FG-4592 (0% bleeding, P < 0.0001 compared with vehicle). Furthermore, FG-4592 reduced epithelial apoptosis by half (P = 0.002) and increased intestinal microvessel density by 80% compared with vehicle controls. EGLN inhibition did not stimulate cancer growth, as treatment with FG-4592 alone, or overexpression of HIF2 within KPC tumors independently improved survival. Thus, we provide a proof of concept for the selective protection of the intestinal tract by the EGLN inhibition to enable ablative doses of cytotoxic therapy in unresectable pancreatic cancer by reducing untoward morbidity and death from radiation-induced gastrointestinal bleeding. SIGNIFICANCE: Selective protection of the intestinal tract by EGLN inhibition enables potentially definitive doses of radiation therapy. This might allow radiation to be a surgical surrogate for unresectable pancreatic cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2327/F1.large.jpg.


Assuntos
Glicina/análogos & derivados , Prolina Dioxigenases do Fator Induzível por Hipóxia/antagonistas & inibidores , Isoquinolinas/farmacologia , Neoplasias Pancreáticas/mortalidade , Lesões por Radiação/prevenção & controle , Protetores contra Radiação/farmacologia , Radioterapia/mortalidade , Animais , Apoptose , Feminino , Glicina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/radioterapia , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Lesões por Radiação/etiologia , Lesões por Radiação/mortalidade , Radioterapia/efeitos adversos , Fatores de Transcrição/fisiologia , Proteína Supressora de Tumor p53/fisiologia
2.
Nanomedicine (Lond) ; 9(8): 1209-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24063415

RESUMO

AIM: We report a magneto-fluorescent theranostic nanocomplex targeted to neutrophil gelatinase-associated lipocalin (NGAL) for imaging and therapy of pancreatic cancer. MATERIALS & METHODS: Gold nanoshells resonant at 810 nm were encapsulated in silica epilayers doped with iron oxide and the near-infrared (NIR) dye indocyanine green, resulting in theranostic gold nanoshells (TGNS), which were subsequently conjugated with antibodies targeting NGAL in AsPC-1-derived xenografts in nude mice. RESULTS: Anti-NGAL-conjugated TGNS specifically targeted pancreatic cancer cells in vitro and in vivo providing contrast for both NIR fluorescence and T2-weighted MRI with higher tumor contrast than can be obtained using long-circulating, but nontargeted, PEGylated nanoparticles. The nanocomplexes also enabled highly specific cancer cell death via NIR photothermal therapy in vitro. CONCLUSION: TGNS with embedded NIR and magnetic resonance contrasts can be specifically targeted to pancreatic cancer cells with expression of early disease marker NGAL, and enable molecularly targeted imaging and photothermal therapy.


Assuntos
Ouro/uso terapêutico , Nanoconchas/uso terapêutico , Pâncreas/patologia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Proteínas de Fase Aguda/metabolismo , Animais , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/uso terapêutico , Sistemas de Liberação de Medicamentos , Feminino , Corantes Fluorescentes/química , Corantes Fluorescentes/uso terapêutico , Ouro/química , Humanos , Hipertermia Induzida , Lipocalina-2 , Lipocalinas/metabolismo , Imageamento por Ressonância Magnética , Imãs/química , Camundongos Nus , Nanoconchas/química , Proteínas Oncogênicas/metabolismo , Imagem Óptica , Neoplasias Pancreáticas/patologia , Fototerapia
3.
Clin Cancer Res ; 18(18): 4942-53, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22832932

RESUMO

PURPOSE: Development of chemoresistance, poor prognosis, and metastasis often renders the current treatments for colorectal cancer (CRC) ineffective. Whether ursolic acid, a component of numerous medicinal plants, either alone or in combination with capecitabine, can inhibit the growth and metastasis of human CRC was investigated. EXPERIMENTAL DESIGN: The effect of ursolic acid on proliferation of CRC cell lines was examined by mitochondrial dye uptake assay, apoptosis by esterase staining, NF-κB activation by DNA-binding assay, and protein expression by Western blot. The effect of ursolic acid on the growth and chemosensitization was also examined in orthotopically implanted CRC in nude mice. RESULTS: We found that ursolic acid inhibited the proliferation of different colon cancer cell lines. This is correlated with inhibition of constitutive NF-κB activation and downregulation of cell survival (Bcl-xL, Bcl-2, cFLIP, and survivin), proliferative (cyclin D1), and metastatic (MMP-9, VEGF, and ICAM-1) proteins. When examined in an orthotopic nude mouse model, ursolic acid significantly inhibited tumor volume, ascites formation, and distant organ metastasis, and this effect was enhanced with capecitabine. Immunohistochemistry of tumor tissue indicated that ursolic acid downregulated biomarkers of proliferation (Ki-67) and microvessel density (CD31). This effect was accompanied by suppression of NF-κB, STAT3, and ß-catenin. In addition, ursolic acid suppressed EGF receptor (EGFR) and induced p53 and p21 expression. We also observed bioavailability of ursolic acid in the serum and tissue of animals. CONCLUSION: Overall, our results show that ursolic acid can inhibit the growth and metastasis of CRC and further enhance the therapeutic effects of capecitabine through the suppression of multiple biomarkers linked to inflammation, proliferation, invasion, angiogenesis, and metastasis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Triterpenos/uso terapêutico , Animais , Antimetabólitos Antineoplásicos/administração & dosagem , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Apoptose/efeitos dos fármacos , Capecitabina , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Desoxicitidina/toxicidade , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Fluoruracila/farmacologia , Fluoruracila/toxicidade , Humanos , Antígeno Ki-67/metabolismo , Masculino , Camundongos , Camundongos Nus , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Metástase Neoplásica/tratamento farmacológico , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Fator de Transcrição STAT3/metabolismo , Triterpenos/administração & dosagem , Proteínas Supressoras de Tumor/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo , Ácido Ursólico
4.
Int J Cancer ; 131(3): E292-303, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21935918

RESUMO

Agents that can potentiate the efficacy of standard chemotherapy against pancreatic cancer are of great interest. Because of their low cost and safety, patients commonly use a variety of dietary supplements, although evidence of their efficacy is often lacking. One such commonly used food supplement is Zyflamend, a polyherbal preparation with potent anti-inflammatory activities and preclinical efficacy against prostate and oral cancer. Whether Zyflamend has any efficacy against human pancreatic cancer alone or in combination with gemcitibine, a commonly used agent, was examined in cell cultures and in an orthotopic mouse model. In vitro, Zyflamend inhibited the proliferation of pancreatic cancer cell lines regardless of p53 status and also enhanced gemcitabine-induced apoptosis. This finding correlated with inhibition of NF-κB activation by Zyflamend and suppression of cyclin D1, c-myc, COX-2, Bcl-2, IAP, survivin, VEGF, ICAM-1 and CXCR4. In nude mice, oral administration of Zyflamend alone significantly inhibited the growth of orthotopically transplanted human pancreatic tumors, and when combined with gemcitabine, further enhanced the antitumor effects. Immunohistochemical and Western blot analyses of tumor tissue showed that the suppression of pancreatic cancer growth correlated with inhibition of proliferation index marker (Ki-67), COX-2, MMP-9, NF-κB and VEGF. Overall, these results suggest that the concentrated multiherb product Zyflamend alone can inhibit the growth of human pancreatic tumors and, in addition, can sensitize pancreatic cancers to gemcitabine through the suppression of multiple targets linked to tumorigenesis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/farmacologia , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica , Humanos , Antígeno Ki-67/análise , Masculino , Camundongos , Camundongos Nus , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteína Supressora de Tumor p53/análise , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
5.
Int J Nanomedicine ; 6: 259-69, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21423588

RESUMO

PURPOSE: Gold nanoshells (NSs) have already shown great promise as photothermal actuators for cancer therapy. Integrin αvß3 is a marker that is specifically and preferentially overexpressed on multiple tumor types and on angiogenic tumor neovasculature. Active targeting of NSs to integrin αvß3 offers the potential to increase accumulation preferentially in tumors and thereby enhance therapy efficacy. METHODS: Enzyme-linked immunosorbent assay (ELISA) and cell binding assay were used to study the in vitro binding affinities of the targeted nanoconjugate NS-RGDfK. In vivo biodistribution and tumor specificity were analyzed using 64Cu-radiolabeled untargeted and targeted NSs in live nude rats bearing head and neck squamous cell carcinoma (HNSCC) xenografts. The potential thermal therapy applications of NS-RGDfK were evaluated by subablative thermal therapy of tumor xenografts using untargeted and targeted NSs. RESULTS: ELISA and cell binding assay confirmed the binding affinity of NS-RGDfK to integrin αvß3. Positron emission tomography/computed tomography imaging suggested that tumor targeting is improved by conjugation of NSs to cyclo(RGDfK) and peaks at ~20 hours postinjection. In the subablative thermal therapy study, greater biological effectiveness of targeted NSs was implied by the greater degree of tumor necrosis. CONCLUSION: The results presented in this paper set the stage for the advancement of integrin αvß3-targeted NSs as therapeutic nanoconstructs for effective cancer therapy.


Assuntos
Carcinoma de Células Escamosas/diagnóstico por imagem , Carcinoma de Células Escamosas/radioterapia , Integrina alfaVbeta3 , Nanoconjugados/química , Peptídeos Cíclicos/farmacologia , Animais , Carcinoma de Células Escamosas/irrigação sanguínea , Linhagem Celular Tumoral , Radioisótopos de Cobre , Ensaio de Imunoadsorção Enzimática , Ouro/química , Temperatura Alta , Humanos , Hipertermia Induzida/métodos , Integrina alfaVbeta3/antagonistas & inibidores , Integrina alfaVbeta3/metabolismo , Terapia a Laser , Camundongos , Camundongos Nus , Modelos Animais , Nanoconchas/química , Ligação Proteica , Ratos , Ratos Nus , Distribuição Tecidual , Tomografia Computadorizada de Emissão/métodos , Transplante Heterólogo
6.
Nano Lett ; 8(5): 1492-500, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18412402

RESUMO

We report noninvasive modulation of in vivo tumor radiation response using gold nanoshells. Mild-temperature hyperthermia generated by near-infrared illumination of gold nanoshell-laden tumors, noninvasively quantified by magnetic resonance temperature imaging, causes an early increase in tumor perfusion that reduces the hypoxic fraction of tumors. A subsequent radiation dose induces vascular disruption with extensive tumor necrosis. Gold nanoshells sequestered in the perivascular space mediate these two tumor vasculature-focused effects to improve radiation response of tumors. This novel integrated antihypoxic and localized vascular disrupting therapy can potentially be combined with other conventional antitumor therapies.


Assuntos
Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Ouro/uso terapêutico , Hipertermia Induzida/métodos , Nanoestruturas/uso terapêutico , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Animais , Linhagem Celular Tumoral , Humanos , Luz , Masculino , Camundongos , Camundongos Nus , Nanomedicina/métodos , Doses de Radiação , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA