Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Eur J Pharmacol ; 864: 172719, 2019 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-31586634

RESUMEN

Gambogic acid (GA) is a potential anti-cancer compound that is extracted from the resin of Garciania hanburyi. The present study was designed to evaluate the anti-metastatic effect of GA on melanoma cell lines in vitro and to explore the underlying mechanism. The anti-proliferative activity of GA on melanoma cells was assessed by CCK-8 assay. The Wound-healing, transwell, adhesion, and tube formation assays were performed to examine the inhibition of GA on the cell's migration, invasion, adhesion, and angiogenesis capacities, respectively. Enzymatic activity of MMP-2 and MMP-9 were detected by gelatin zymography assay. Protein expressions regulated by GA treatment were tested by Western blot assay. The present results showed that GA significantly inhibited the proliferation of highly metastatic melanoma A375, B16-F10 cells and human umbilical vein endothelial cells (HUVECs) in time- and doses-dependent manners. Furthermore, GA significantly inhibited the migratory, invasive and adhesive properties of A375 and B16-F10 cells, and tube-forming potential of HUVECs at sub-IC50 concentrations, where no significant cytotoxicity was observed. Mechanistically, GA treatment suppressed the EMT and angiogenesis processes and reduced the enzymatic activity of MMP-2 and MMP-9. Moreover, abnormal PI3K/Akt and ERK signaling pathways in A375 and B16-F10 cells and HUVECs were notably suppressed by GA treatment. Collectively, our results suggest that GA exerts anti-metastasis activity in melanoma cells by suppressing the EMT and angiogenesis through the PI3K/Akt and ERK signaling pathways, and might be used as a phytomedicine against metastatic melanoma.


Asunto(s)
Antineoplásicos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Melanoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/patología , Xantonas/farmacología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Melanoma/irrigación sanguínea , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Cutáneas/irrigación sanguínea , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Xantonas/uso terapéutico , Melanoma Cutáneo Maligno
2.
Mar Drugs ; 17(5)2019 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-31035725

RESUMEN

Melanoma is one of the most malignant and aggressive types of cancer worldwide. Fibroblast growth factor 2 (FGF2) is one of the critical regulators of melanoma angiogenesis and metastasis; thus, it might be an effective anti-cancer strategy to explore FGF2-targeting drug candidates from existing drugs. In this study, we evaluate the effect of the marine drug propylene glycol alginate sodium sulfate (PSS) on FGF2-mediated angiogenesis and invasion. The data shows that FGF2 selectively bound to PSS with high affinity. PSS inhibited FGF2-mediated angiogenesis in a rat aortic ring model and suppressed FGF2-mediated invasion, but not the migration of murine melanoma B16-F10 cells. The further mechanism study indicates that PSS decreased the expression of activated matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9), and also suppressed their activity. In addition, PSS was found to decrease the level of Vimentin in B16-F10 cells, which is known to participate in the epithelial-mesenchymal transition. Notably, PSS did not elicit any changes in cancer cell viability. Based on the results above, we conclude that PSS might be a potential drug to regulate the tumor microenvironment in order to facilitate the recovery of melanoma patients.


Asunto(s)
Alginatos/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Melanoma Experimental/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Alginatos/uso terapéutico , Animales , Aorta/efectos de los fármacos , Organismos Acuáticos/química , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Embrión de Pollo , Membrana Corioalantoides , Evaluación Preclínica de Medicamentos , Transición Epitelial-Mesenquimal , Humanos , Laminaria/química , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Neovascularización Patológica/patología , Neovascularización Fisiológica/efectos de los fármacos , Técnicas de Cultivo de Órganos , Ratas , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Microambiente Tumoral/efectos de los fármacos
3.
Bioelectromagnetics ; 36(6): 464-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26179286

RESUMEN

This study demonstrates that 20-100 GHz range can be used for spatially-accurate focusing of heating inside the skin achieved by varying frequency and exposure beam size, as well as by enforcing air convection. The latter is also used to reduce overheating of skin surface. Heating at different skin depths depending on these parameters is investigated in detail using the hybrid bio-heat equation. In particular, it is shown that decreasing frequency and/or increasing exposure beam size at forced airflow result in elevation of heating of deeper layers of tissue and decrease of skin surface temperature. Changes of water content within 15%, which exceed those due to aging and presence of tumors, only slightly affect heating. Exposure intensity necessary to reach a target temperature significantly increases in different areas of body with elevated blood flow. Dependence on exposure intensity and hyperthermia treatment duration is also investigated and discussed. Results of this study suggest that the lower part of the millimeter-wave range is an attractive alternative for non-invasive thermal treatment of skin cancer with a high spatial resolution.


Asunto(s)
Calor , Hipertermia Inducida/métodos , Microondas/uso terapéutico , Piel/efectos de la radiación , Aire , Circulación Sanguínea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Cinética , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma/terapia , Modelos Biológicos , Piel/irrigación sanguínea , Piel/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia , Agua/metabolismo
4.
Int J Hyperthermia ; 31(6): 615-25, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25982485

RESUMEN

PURPOSE: This study investigates the influence of blood perfusion variability within a tumour and the surrounding healthy tissue during nanoparticle-assisted thermal therapy. It seeks to define ideal therapeutic parameters for a wide range of perfusion rates to attain the desired thermal damage. MATERIAL AND METHODS: Pennes' bioheat model and the Arrhenius model are used to evaluate the thermal damage for a two-dimensional tumour surrounded by healthy tissue. A wide range of tumour perfusion rates were modelled, ranging from moderate to high perfusion in both a homogenously and a heterogeneously perfused tumour. RESULTS: For low perfusion rates, a temporal variation in blood perfusion does not critically influence the thermal damage. For moderately and highly perfused tumours, temporal variation in blood perfusion extends the thermal damage zone by 25-52% compared to a constant perfusion rate. For the tumour size and perfusion conditions under consideration, the ideal therapeutic parameters were found to be irradiation intensity of 1 W/cm(2), and irradiation duration of 105-150 s, for a nanoparticle volume fraction of 0.001%. CONCLUSIONS: It is concluded for low perfusion rates that due to shorter therapeutic duration, nanoparticle-assisted thermal therapy is relatively insensitive to changes in the perfusion rate during the therapy. For moderately and highly perfused tumours, a constant perfusion under-predicts the real thermal damage zone. This study concludes that for moderately and highly perfused tumours the spatial as well as temporal blood perfusion dynamics should be carefully accounted for to get a realistic estimate of thermal damage zone.


Asunto(s)
Hipertermia Inducida , Modelos Biológicos , Nanopartículas/uso terapéutico , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/terapia , Flujo Sanguíneo Regional
5.
Mol Cell Proteomics ; 12(6): 1621-31, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23449826

RESUMEN

Predictive biomarkers are needed to triage patients to the best therapy. We prospectively planned examination of sequential blood, biopsy, and functional imaging with which to confirm the mechanism and to identify potential predictive biomarkers in a phase Ib clinical trial expansion of patients with solid tumors receiving sorafenib/bevacizumab. The maximally tolerated doses of sorafenib at 200 mg twice daily with bevacizumab at 5 mg/kg every other week were given to biopsiable patients. Patients were randomized to receive either sorafenib or bevacizumab monotherapy for the first 28-day cycle with the second drug added with cycle 2. Biopsies, dynamic contrast-enhanced MRI, and fluorodeoxyglucose-proton emission tomography were done pre-therapy and at 2 and 6 weeks (2 weeks into combination therapy). Tumor and serum proteomics, Ras/Raf mutational analysis, and functional imaging results were examined individually and across the dataset to identify potential changes predictive of response to therapy and those that confirm the biochemical drug mechanism(s). Therapy with sorafenib/bevacizumab resulted in clinical benefit in 45% of this mixed solid tumor group. ERK activation and microvessel density were decreased with monotherapy treatment with sorafenib or bevacizumab, respectively; whereas a decreased signal over the group of total AKT, phospho(p)-VEGF receptor2, p-endothelial nitric-oxide synthase, b-RAF, and cleaved poly(ADP-ribose) polymerase was associated with earlier progression of disease. Tumor metabolic activity decreased in those patients with clinical benefits lasting longer than 4 months, and activity increased with progression of disease. Cleavage of caspase 3 and poly(ADP-ribose) polymerase was increased, and Ki67 expression decreased in patients with prolonged clinical benefits, consistent with decreased proliferation and increased apoptosis. The conglomerate analysis, incorporating pharmacodynamic and tumor biochemistry, demonstrated sorafenib/bevacizumab-targeted vascular activity in the tumor. Results suggest potential biomarkers for which changes, as a group, during early therapeutic exposure may predict clinical benefit.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de los Genitales Femeninos/diagnóstico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/uso terapéutico , Neoplasias Cutáneas/diagnóstico , Adulto , Anciano , Bevacizumab , Estudios de Cohortes , Progresión de la Enfermedad , Esquema de Medicación , Quimioterapia Combinada , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Neoplasias de los Genitales Femeninos/irrigación sanguínea , Neoplasias de los Genitales Femeninos/genética , Neoplasias de los Genitales Femeninos/patología , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica , Niacinamida/uso terapéutico , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Valor Predictivo de las Pruebas , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Sorafenib , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
J Mol Med (Berl) ; 91(7): 883-99, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23471434

RESUMEN

Tumor hypoxia is a characteristic of cancer cell growth and invasion, promoting angiogenesis, which facilitates metastasis. Oxygen delivery remains impaired because tumor vessels are anarchic and leaky, contributing to tumor cell dissemination. Counteracting hypoxia by normalizing tumor vessels in order to improve drug and radio therapy efficacy and avoid cancer stem-like cell selection is a highly challenging issue. We show here that inositol trispyrophosphate (ITPP) treatment stably increases oxygen tension and blood flow in melanoma and breast cancer syngeneic models. It suppresses hypoxia-inducible factors (HIFs) and proangiogenic/glycolysis genes and proteins cascade. It selectively activates the tumor suppressor phosphatase and tensin homolog (PTEN) in vitro and in vivo at the endothelial cell (EC) level thus inhibiting PI3K and reducing tumor AKT phosphorylation. These mechanisms normalize tumor vessels by EC reorganization, maturation, pericytes attraction, and lowering progenitor cells recruitment in the tumor. It strongly reduces vascular leakage, tumor growth, drug resistance, and metastasis. ITPP treatment avoids cancer stem-like cell selection, multidrug resistance (MDR) activation and efficiently enhances chemotherapeutic drugs activity. These data show that counteracting tumor hypoxia by stably restoring healthy vasculature is achieved by ITPP treatment, which opens new therapeutic options overcoming hypoxia-related limitations of antiangiogenesis-restricted therapies. By achieving long-term vessels normalization, ITPP should provide the adjuvant treatment required in order to overcome the subtle definition of therapeutic windows for in vivo treatments aimed by the current strategies against angiogenesis-dependent tumors.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Fosfatos de Inositol/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Línea Celular , Línea Celular Tumoral , Células Endoteliales/metabolismo , Femenino , Hipoxia/tratamiento farmacológico , Fosfatos de Inositol/farmacología , Melanoma/irrigación sanguínea , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neovascularización Patológica/tratamiento farmacológico , Oxígeno/metabolismo , Fosfohidrolasa PTEN/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Carga Tumoral/efectos de los fármacos
7.
Iran Biomed J ; 16(2): 59-67, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22801278

RESUMEN

BACKGROUND: A growing body of preclinical data indicates that statins may possess antineoplastic properties; however, some studies have raised the possibility that statins may also have carcinogenic potential. METHODS: An air pouch model was used for angiogenesis. Single or multiple applications of croton oil on the back of Swiss albino mice with or without initiation by dimethylbenz(a)antheracene (DMBA) were used to evaluate the skin tumorgenesis, ultrastructural and histological alterations. RESULTS: Atorvastatin (orally, 10 mg/kg/day) produced a significant (P<0.05) reduction in angiogenesis. Concurrent administration of mevalonate reversed the anti-angiogenic effect of atorvastatin. However, local injection of atorvastatin (200 µg) into the pouches induced a significant (P<0.5) increase in angiogenesis that was not reversed by co-administration of mevalonate. The disturbance of cell polarity, inflammatory response, thickness of epidermal layer, and mitotic index induced by croton oil were inhibited markedly and dose-dependently (P<0.001) by pre-treatment with atorvastatin. In spite of the strong anti-inflammatory and anti-proliferative effects of atorvastatin on epidermal cell proliferation, it was identified that the same doses of atorvastatin in DMBA-initiated and croton oil-promoted skin tumorgenesis in mice increased the incidence of tumors and their conversion into malignant carcinoma. CONCLUSION: The reasons for these discrepancies remain unclear, and could be related to ambivalent effects of atorvastatin on angiogenesis or to specific differences in the experimental conditions. It is suggested that the pro-angiogenic effect of the drug, which could be responsible for promotion of skin tumors, is independent of the 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase inhibition that can be mediated directly by atorvastatin.


Asunto(s)
Epidermis/efectos de los fármacos , Ácidos Heptanoicos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Neovascularización Patológica , Pirroles/farmacología , Neoplasias Cutáneas/patología , 9,10-Dimetil-1,2-benzantraceno , Animales , Atorvastatina , Proliferación Celular/efectos de los fármacos , Aceite de Crotón , Células Epidérmicas , Femenino , Ácidos Heptanoicos/administración & dosificación , Hidroximetilglutaril-CoA Reductasas/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Masculino , Ácido Mevalónico/farmacología , Ratones , Pirroles/administración & dosificación , Ratas , Ratas Wistar , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/inducido químicamente
8.
J Clin Invest ; 122(4): 1427-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22426208

RESUMEN

Localized tissue hypoxia is a consequence of vascular compromise or rapid cellular proliferation and is a potent inducer of compensatory angiogenesis. The oxygen-responsive transcriptional regulator hypoxia-inducible factor 2α (HIF-2α) is highly expressed in vascular ECs and, along with HIF-1α, activates expression of target genes whose products modulate vascular functions and angiogenesis. However, the mechanisms by which HIF-2α regulates EC function and tissue perfusion under physiological and pathological conditions are poorly understood. Using mice in which Hif2a was specifically deleted in ECs, we demonstrate here that HIF-2α expression is required for angiogenic responses during hindlimb ischemia and for the growth of autochthonous skin tumors. EC-specific Hif2a deletion resulted in increased vessel formation in both models; however, these vessels failed to undergo proper arteriogenesis, resulting in poor perfusion. Analysis of cultured HIF-2α-deficient ECs revealed cell-autonomous increases in migration, invasion, and morphogenetic activity, which correlated with HIF-2α-dependent expression of specific angiogenic factors, including delta-like ligand 4 (Dll4), a Notch ligand, and angiopoietin 2. By stimulating Dll4 signaling in cultured ECs or restoring Dll4 expression in ischemic muscle tissue, we rescued most of the HIF-2α-dependent EC phenotypes in vitro and in vivo, emphasizing the critical role of Dll4/Notch signaling as a downstream target of HIF-2α in ECs. These results indicate that HIF-1α and HIF-2α fulfill complementary, but largely nonoverlapping, essential functions in pathophysiological angiogenesis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Circulación Colateral/fisiología , Células Endoteliales/metabolismo , Miembro Posterior/irrigación sanguínea , Isquemia/fisiopatología , Neovascularización Patológica/fisiopatología , Neoplasias Cutáneas/irrigación sanguínea , Proteínas Adaptadoras Transductoras de Señales , Angiopoyetina 2/genética , Angiopoyetina 2/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de Unión al Calcio , Hipoxia de la Célula , Movimiento Celular , Células Cultivadas/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neovascularización Fisiológica/fisiología , Receptores Notch/fisiología , Proteínas Recombinantes de Fusión/fisiología , Recuperación de la Función , Neoplasias Cutáneas/inducido químicamente , Cicatrización de Heridas/fisiología
9.
J Dermatolog Treat ; 22(3): 153-61, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20666667

RESUMEN

A recent study has shown that nanosecond pulsed electric fields (nsPEF) can affect the intracellular structures of melanoma within weeks. nsPEF is a non-drug, non-thermal treatment using ultrashort, intense pulsed electric fields with nanosecond durations. In the current study we followed up melanoma histopathology and metastasis with tissue micro-array 5 months post-nsPEF. After nsPEF treatment, tumor growth, tumor histology, metastasis, peri-tumor vessel and micro-vessel density were examined for the effect of nsPEF treatment on melanoma in vivo. The 17 nsPEF-treated mice were tumor-free for 169 days, significantly longer than those 19 control mice bearing melanoma without nsPEF. Histopathology follow-up showed that melanoma did not recur to the primary injection place after complete elimination. Compared with the control tumor, nsPEF-treated tumors present decreased micro-vessel density in a time-course manner in this survival study. Treatment with nsPEF caused continuous histopathological changes in melanomas, eliminated melanoma without recurrence at the primary site and prolonged animal survival time by inhibiting tumor blood supply and leading to tumor infarction. Thus, nsPEF could be applied in a non-ionizing therapeutic approach, without other agents, to locally treat tumors within a defined boundary.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Femenino , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Melanoma Experimental/secundario , Ratones , Ratones Pelados , Trasplante de Neoplasias , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología
10.
Dis Model Mech ; 3(5-6): 317-32, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20223936

RESUMEN

Innate regulatory networks within organs maintain tissue homeostasis and facilitate rapid responses to damage. We identified a novel pathway regulating vessel stability in tissues that involves matrix metalloproteinase 14 (MMP14) and transforming growth factor beta 1 (TGFbeta(1)). Whereas plasma proteins rapidly extravasate out of vasculature in wild-type mice following acute damage, short-term treatment of mice in vivo with a broad-spectrum metalloproteinase inhibitor, neutralizing antibodies to TGFbeta(1), or an activin-like kinase 5 (ALK5) inhibitor significantly enhanced vessel leakage. By contrast, in a mouse model of age-related dermal fibrosis, where MMP14 activity and TGFbeta bioavailability are chronically elevated, or in mice that ectopically express TGFbeta in the epidermis, cutaneous vessels are resistant to acute leakage. Characteristic responses to tissue damage are reinstated if the fibrotic mice are pretreated with metalloproteinase inhibitors or TGFbeta signaling antagonists. Neoplastic tissues, however, are in a constant state of tissue damage and exhibit altered hemodynamics owing to hyperleaky angiogenic vasculature. In two distinct transgenic mouse tumor models, inhibition of ALK5 further enhanced vascular leakage into the interstitium and facilitated increased delivery of high molecular weight compounds into premalignant tissue and tumors. Taken together, these data define a central pathway involving MMP14 and TGFbeta that mediates vessel stability and vascular response to tissue injury. Antagonists of this pathway could be therapeutically exploited to improve the delivery of therapeutics or molecular contrast agents into tissues where chronic damage or neoplastic disease limits their efficient delivery.


Asunto(s)
Vasos Sanguíneos/enzimología , Vasos Sanguíneos/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Envejecimiento/patología , Animales , Colágenos Fibrilares/metabolismo , Homeostasis , Metaloproteinasa 14 de la Matriz/deficiencia , Ratones , Modelos Biológicos , Planta de la Mostaza , Aceites de Plantas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/patología , Células del Estroma/enzimología , Células del Estroma/patología , Resistencia Vascular
11.
Melanoma Res ; 19(6): 361-71, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19730404

RESUMEN

Nanosecond pulsed electric fields (nsPEFs) can affect the intracellular structures of cells in vitro. This study shows the direct effects of nsPEFs on tumor growth, tumor volume, and histological characteristics of normal skin and B16-F10 melanoma in SKH-1 mice. A melanoma model was set up by injecting B16-F10 into female SKH-1 mice. After a 100-pulse treatment with an nsPEF (40-kV/cm field strength; 300-ns duration; 30-ns rise time; 2-Hz repetition rate), tumor growth and histology were studied using transillumination, light microscopy with hematoxylin and eosin stain and transmission electron microscopy. Melanin and iron within the melanoma tumor were also detected with specific stains. After nsPEF treatment, tumor development was inhibited with decreased volumes post-nsPEF treatment compared with control tumors (P<0.05). The nsPEF-treated tumor volume was reduced significantly compared with the control group (P<0.01). Hematoxylin and eosin stain and transmission electron microscopy showed morphological changes and nuclear shrinkage in the tumor. Fontana-Masson stain indicates that nsPEF can externalize the melanin. Iron stain suggested nsPEF caused slight hemorrhage in the treated tissue. Histology confirmed that repeated applications of nsPEF disrupted the vascular network. nsPEF treatment can significantly disrupt the vasculature, reduce subcutaneous murine melanoma development, and produce tumor cell contraction and nuclear shrinkage while concurrently, but not permanently, damaging peripheral healthy skin tissue in the treated area, which we attribute to the highly localized electric fields surrounding the needle electrodes.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/patología , Melanoma Experimental/terapia , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Piel/química , Animales , Núcleo Celular , Electricidad , Femenino , Hierro/análisis , Melaninas/análisis , Melanoma Experimental/irrigación sanguínea , Ratones , Microscopía Electrónica de Transmisión , Neoplasias Cutáneas/irrigación sanguínea , Células Tumorales Cultivadas
12.
Int J Cancer ; 125(2): 438-45, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19408306

RESUMEN

We have discovered a new, ultrafast therapy for treating skin cancer that is extremely effective with a total electric field exposure time of only 180 microsec. The application of 300 high-voltage (40 kV/cm), ultrashort (300 nsec) electrical pulses to murine melanomas in vivo triggers both necrosis and apoptosis, resulting in complete tumor remission within an average of 47 days in the 17 animals treated. None of these melanomas recurred during a 4-month period after the initial melanoma had disappeared. These pulses generate small, long-lasting, rectifying nanopores in the plasma membrane of exposed cells, resulting in increased membrane permeability to small molecules and ions, as well as an increase in intracellular Ca(2+), DNA fragmentation, disruption of the tumor's blood supply and the initiation of apoptosis. Apoptosis was indicated by a 3-fold increase in Bad labeling and a 72% decrease in Bcl-2 labeling. In addition, microvessel density within the treated tumors fell by 93%. This new therapy utilizing nanosecond pulsed electric fields has the advantages of highly localized targeting of tumor cells and a total exposure time of only 180 microsec. These pulses penetrate into the interior of every tumor cell and initiate DNA fragmentation and apoptosis while at the same time reducing blood flow to the tumor. This new physical tumor therapy is drug free, highly localized, uses low energy, has no significant side effects and results in very little scarring.


Asunto(s)
Terapia por Estimulación Eléctrica , Melanoma Experimental/terapia , Neoplasias Cutáneas/terapia , Animales , Calcio/metabolismo , Femenino , Inmunohistoquímica , Melanoma Experimental/irrigación sanguínea , Ratones , Ratones Desnudos , Técnicas de Placa-Clamp , Recurrencia , Inducción de Remisión , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo
13.
Cancer Lett ; 281(1): 108-16, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19332363

RESUMEN

We examined the anti-angiogenic effects of Ganoderma tsugae methanol extract (GTME) on human epidermoid carcinoma A-431 cells. Our data indicate that GTME inhibits the expression of epidermal growth factor receptor (EGFR) and vascular endothelial growth factor (VEGF) in vitro and in vivo, and also inhibits the capillary tube formation of human umbilical vein endothelial cells (HUVECs). We also show that the suppression of VEGF expression by GTME can be restored by treatment with EGF. These results suggest that GTME inhibits VEGF expression via the suppression of EGFR expression, resulting in the downregulation of VEGF secretion from epidermoid carcinoma A-431 cells. These findings reveal a novel role for G. tsugae in inhibiting EGFR and VEGF expression, which are important for tumor angiogenesis and growth. Thus, GTME may provide a potential therapeutic approach for anti-tumor treatment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/patología , Medicamentos Herbarios Chinos/farmacología , Receptores ErbB/biosíntesis , Ganoderma/química , Proteínas de Neoplasias/biosíntesis , Neovascularización Patológica/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/farmacología , Neoplasias Cutáneas/patología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Línea Celular Tumoral/trasplante , Células Cultivadas/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Endotelio Vascular/citología , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Receptores ErbB/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metanol , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/efectos de los fármacos , Extractos Vegetales/uso terapéutico , Reishi , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Nutr ; 135(12): 2871-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16317135

RESUMEN

Green tea polyphenols (GTPs) show promise as anticarcinogenic agents and may prevent the development of solar UV radiation-induced skin cancer. Here we investigated the mechanisms by which GTPs prevent UVB-induced skin cancer in mice. Two groups of 6- to 7-wk-old female SKH-1 hairless mice were UVB irradiated (180 mJ/cm(2)) 3 times each week for 24 wk. One group consumed water and the other, water containing 2 g/L GTPs. A control group drank water and was not exposed to UVB radiation. UVB-induced tumors and skin biopsies from the control group were analyzed using immunostaining, Western blotting, and gelatinolytic zymography. Oral administration of GTPs reduced UVB-induced tumor incidence (35%), tumor multiplicity (63%), and tumor growth (55%). The GTPs+UVB group had reduced expression of the matrix metalloproteinases (MMP)-2 and MMP-9, which have crucial roles in tumor growth and metastasis, and enhanced expression of tissue inhibitor of MMP in the tumors compared with mice that were treated with UVB alone. The GTPs+UVB group also had reduced expressions of CD31 and vascular endothelial growth factor, which are essential for angiogenesis, and inhibited expression of proliferating cell nuclear antigen in the tumors compared with the UVB group. Additionally, there were more cytotoxic CD8(+) T cells in the tumors of the GTPs+UVB group than in the UVB group and their tumor cells exhibited greater activation of caspase-3, indicating the apoptotic death of the tumor cells. Taken together, these data suggest that in mice, administration of GTPs affects several biomarkers that are involved in UV-carcinogenesis, including inhibition of angiogenic factors and recruitment of cytotoxic T cells in the tumor microenvironment.


Asunto(s)
Flavonoides/administración & dosificación , Neoplasias Inducidas por Radiación/prevención & control , Neovascularización Patológica/prevención & control , Fenoles/administración & dosificación , Neoplasias Cutáneas/diagnóstico por imagen , Neoplasias Cutáneas/prevención & control , , Rayos Ultravioleta , Administración Oral , Animales , Modelos Animales de Enfermedad , Femenino , Flavonoides/farmacología , Guanosina 5'-O-(3-Tiotrifosfato)/uso terapéutico , Ratones , Ratones Pelados , Neoplasias Inducidas por Radiación/irrigación sanguínea , Fenoles/farmacología , Polifenoles , Cintigrafía , Neoplasias Cutáneas/irrigación sanguínea
16.
BMC Cancer ; 4: 53, 2004 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-15324457

RESUMEN

BACKGROUND: The aim of this experimental study was to determine the effect of mild hyperthermia on tumor response and angioneogenesis in an isolated limb perfusion model with a human melanoma xenograft. METHODS: A human melanoma xenograft was implanted into the hindlimbs of 30 athymic nude rats. The animals were randomized into five groups: group I: control, group II: sham group, group III: external hyperthermia with a tissue temperature of 41.5 degrees C for 30 minutes without ILP, group IV: normothermic ILP (tissue temperature 37 degrees C for 30 minutes, group V: hyperthermic ILP (tissue temperature 41.5 degrees C for 30 minutes). Tumor response was evaluated by tumor size determination and immunohistochemical analysis 6 weeks postoperatively. Tissue sections were investigated for expression of CD34 and basic fibroblast growth factor (bFGF). RESULTS: Average tumor volumes of the controls (I) increased from 105 mm3 to 1388 mm3. In the sham operated group (II) tumor volumes were significantly larger than in group I. Tumor volumes in group IV were significantly smaller than in group I and lowest in group V. There were no significant differences in size between group I and group III after six weeks. In group III and IV each, 5 animals showed tumor progression and one had a partial tumor response. In group V only 2 animals showed tumor progression. Immunhistochemical analysis of the tissue sections demonstrated that angioneogenesis was more pronounced in group II than in group I and less pronounced in group IV and V compared with group I. CONCLUSIONS: Our results suggest that even a surgical manipulation such as a skin incision promotes tumor growth, probably by induction of growth factors like bFGF. External hyperthermia of 41.5 degrees C tissue temperature for 30 minutes only has no impact on tumor growth and angioneogenesis in vivo.


Asunto(s)
Hipertermia Inducida , Melanoma/irrigación sanguínea , Melanoma/terapia , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/terapia , Animales , Antígenos CD34/análisis , Biomarcadores de Tumor/análisis , Terapia Combinada , Citoplasma/química , Factor 2 de Crecimiento de Fibroblastos/análisis , Miembro Posterior/irrigación sanguínea , Humanos , Inmunohistoquímica , Masculino , Melanoma/patología , Melanoma/cirugía , Neovascularización Patológica/prevención & control , Perfusión , Distribución Aleatoria , Ratas , Ratas Desnudas , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/cirugía , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Eur J Cancer ; 40(10): 1554-65, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15196540

RESUMEN

This study evaluated the anti-angiogenic activities of erianin in vivo and in vitro. Erianin, a natural product from Dendrobium chrysotoxum, caused moderate growth delay in xenografted human hepatoma Bel7402 and melanoma A375 and induced significant vascular shutdown within 4 h of administering 100 mg/kg of the drug. Erianin also displayed potent anti-angiogenic activities in vitro: it abrogated spontaneous or basic fibroblast growth factor-induced neovascularisation in chick embryo; it inhibited proliferation of human umbilical vein endothelial cells (EC(50) 34.1+/-12.7 nM), disrupted endothelial tube formation, and abolished migration across collagen and adhesion to fibronectin. Erianin also exerted selective inhibition toward endothelial cells, and quiescent endothelium showed more resistance than in proliferative and tumour conditions. In a cytoskeletal study, erianin depolymerised both F-actin and beta-tubulin, more significantly in proliferating endothelial cells than in confluent cells. In conclusion, erianin caused extensive tumour necrosis, growth delay and rapid vascular shutdown in hepatoma and melanoma models; it inhibited angiogenesis in vivo and in vitro and induced endothelial cytoskeletal disorganisation. These findings suggest that erianin has the therapeutic potential to inhibit angiogenesis in vivo and in vitro.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Bibencilos/uso terapéutico , Carcinoma Hepatocelular/irrigación sanguínea , Neoplasias Hepáticas/irrigación sanguínea , Melanoma/irrigación sanguínea , Neoplasias Cutáneas/irrigación sanguínea , Células 3T3 , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Evaluación Preclínica de Medicamentos , Femenino , Neoplasias Hepáticas/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Fenol , Neoplasias Cutáneas/tratamiento farmacológico , Células Tumorales Cultivadas
19.
Br J Cancer ; 78(1): 56-61, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9662251

RESUMEN

The effect of localized hypothermia on microcirculatory and metabolic parameters in s.c. DS sarcomas on the hind foot dorsum of Sprague-Dawley rats was investigated. Tumours were cooled by superfusion of the tumour surface with cooled saline solution to 25 degrees C or 15 degrees C. Control tumours remained at 35 degrees C. These temperatures were maintained for 30 min. In tumour oxygenation measurements, hypothermia at 25 degrees C and 15 degrees C caused progressive decreases in the size of the fraction of pO2 measurements between 0 and 2.5 mmHg together with a reduction in pO2 variability. No significant changes in median or mean pO2 or in the fraction of pO2 measurements between 0 and 5 mmHg, and 0 and 10 mmHg were observed. Using laser Doppler flowmetry, red blood cell flux was found to decrease significantly upon 25 degrees C or 15 degrees C hypothermia treatment to 67% and 37% of starting values respectively, whereas no significant changes were seen in control tumours over the whole observation period. Viscosity was measured in blood and plasma samples over a range of temperatures and was found to increase with decreasing temperature. Assessment of tumour glucose levels showed an increased concentration of glucose following 15 degrees C hypothermia, an observation consistent with a 'slowing down' of glycolysis. No changes in lactate or adenylate phosphate levels were observed. As a way of improving tumour oxygenation, localized hypothermia may therefore be a useful means of radiosensitization.


Asunto(s)
Hipotermia Inducida , Consumo de Oxígeno , Sarcoma Experimental , Neoplasias Cutáneas , Animales , Viscosidad Sanguínea , Flujometría por Láser-Doppler , Masculino , Microcirculación , Ratas , Ratas Sprague-Dawley , Sarcoma Experimental/sangre , Sarcoma Experimental/irrigación sanguínea , Sarcoma Experimental/metabolismo , Sarcoma Experimental/terapia , Neoplasias Cutáneas/sangre , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia
20.
J Neurooncol ; 38(1): 51-7, 1998 May.
Artículo en Inglés | MEDLINE | ID: mdl-9540057

RESUMEN

The effect of the anti-angiogenic agent TNP-470 on tumor growth, vascular area, vascular density and tumor perfusion of two different subcutaneously implanted human glioma xenografts (E98 and E106) in nude mice was evaluated. Vascular parameters were investigated with an image analysis system. For both tumor lines a small but significant tumor growth suppression was observed. However, no differences in vascular parameters between TNP-470 treated tumors and controls could be found after 6 weeks of treatment. It is concluded that although TNP-470 is a promising anti-angiogenic agent in many tumor types, at least 2 glioma lines seem to be partly resistant to its anti-angiogenic effects. Further evaluation of the effects of combination of TNP-470 and cytostatic agents or radiotherapy in human glioma xenografts are required to determine the place of anti-angiogenic therapy in general and treatment with the anti-angiogenic agent TNP-470 more specifically in the treatment of human gliomas.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Glioma/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Sesquiterpenos/uso terapéutico , Animales , División Celular/efectos de los fármacos , Ciclohexanos , Glioma/irrigación sanguínea , Glioma/patología , Humanos , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , O-(Cloroacetilcarbamoil) Fumagilol , Neoplasias Cutáneas/irrigación sanguínea , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA