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
País de afiliação
Intervalo de ano de publicação
1.
J Control Release ; 260: 92-99, 2017 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-28527736

RESUMO

The aim of this study is to understand the combined and differential biokinetic effects of radiofrequency (RF) electric-field hyperthermia as an adjunctive therapy to [60]fullerene nanoparticle-based drug delivery systems in targeting the micro-vasculature and micro-environments of breast cancer tumors. Intravital microscopy (IVM) is an ideal tool to provide the spatial and temporal resolution needed for quantification in this investigation. The water-soluble and fluorescent [60]fullerene derivative (C60-serPF) was designed to be an amphiphilic nanostructure, which is able to cross several biological membranes and accumulate in tumor tissues by passing through abnormally leaky tumor blood vessels. To elucidate the coupled effects of the highly permeable, but heterogeneous tumor vasculature, with the permeabilizing effects of mild (40-42°C) hyperthermia produced by a local RF field, we controlled variables across tumor and non-tumor mammary gland microvasculature with and without application of RF hyperthermia in each condition. We notice that tumor tissue is characterized by more intense drug extravasation than in contralateral mammary fat pad tissue, which is consistent with enhanced permeability and retention (EPR) effects. The analysis of a permeability parameter (Papp), C60-serPF velocity, and the time of compound influx into the intra- and extra-vascular space suggest that mild RF hyperthermia can improve nanoparticle delivery into tumor tissue.


Assuntos
Adenocarcinoma/metabolismo , Fulerenos/administração & dosagem , Hipertermia Induzida , Neoplasias Mamárias Experimentais/metabolismo , Animais , Transporte Biológico , Linhagem Celular Tumoral , Terapia Combinada , Sistemas de Liberação de Medicamentos , Feminino , Fulerenos/farmacocinética , Camundongos Endogâmicos BALB C , Camundongos Nus , Distribuição Tecidual
2.
Chem Commun (Camb) ; 52(85): 12630-12633, 2016 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-27722511

RESUMO

For potential applications in nano-mediated radiofrequency cancer hyperthermia, the nanomaterial under investigation must increase the heating of any aqueous solution in which it is suspended when exposed to radiofrequency electric fields. This should also be true for a broad range of solution conductivities, especially those that artificially mimic the ionic environment of biological systems. Herein we demonstrate enhanced heating of biologically relevant aqueous solutions using kosmotropes and a hexamalonoserinolamide fullerene.


Assuntos
Ablação por Cateter , Fulerenos/química , Hipertermia Induzida , Nanoestruturas/química , Água/química , Humanos
3.
PLoS One ; 10(8): e0136382, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26308617

RESUMO

Herein, we present a novel imaging platform to study the biological effects of non-invasive radiofrequency (RF) electric field cancer hyperthermia. This system allows for real-time in vivo intravital microscopy (IVM) imaging of radiofrequency-induced biological alterations such as changes in vessel structure and drug perfusion. Our results indicate that the IVM system is able to handle exposure to high-power electric-fields without inducing significant hardware damage or imaging artifacts. Furthermore, short durations of low-power (< 200 W) radiofrequency exposure increased transport and perfusion of fluorescent tracers into the tumors at temperatures below 41°C. Vessel deformations and blood coagulation were seen for tumor temperatures around 44°C. These results highlight the use of our integrated IVM-RF imaging platform as a powerful new tool to visualize the dynamics and interplay between radiofrequency energy and biological tissues, organs, and tumors.


Assuntos
Diagnóstico por Imagem , Hipertermia Induzida , Microscopia Intravital/métodos , Neoplasias Mamárias Animais/patologia , Ondas de Rádio , Algoritmos , Animais , Feminino , Imunofluorescência , Corantes Fluorescentes/farmacocinética , Neoplasias Mamárias Animais/terapia , Camundongos , Distribuição Tecidual
4.
J Natl Cancer Inst ; 106(8)2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25128695

RESUMO

BACKGROUND: Gemcitabine is a potent nucleoside analogue against solid tumors, but development of drug resistance is a substantial problem. Removal of gemcitabine incorporated into DNA by repair mechanisms may contribute to resistance in chemo-refractory solid tumors. Human hepatocellular carcinoma (HCC) is usually very chemoresistant to gemcitabine. METHODS: We treated HCC in vitro and in vivo (orthotopic murine model with human Hep3B or HepG2 xenografts, 7-10 CB17SCID mice per group) with gemcitabine. The role of homologous recombination repair proteins in repairing stalled replication forks was evaluated with hyperthermia exposure and cell-cycle analysis. The Student t-test was used for two-sample comparisons. Multiple group data were analyzed using one-way analysis of variance. All statistical tests were two-sided. RESULTS: We demonstrated that Mre11-mediated homologous recombination repair of gemcitabine-stalled replication forks is crucial to survival of HCC cells. Furthermore, we demonstrated inhibition of Mre11 by an exonuclease inhibitor or concomitant hyperthermia. In orthotopic murine models of chemoresistant HCC, the Hep3B tumor mass with radiofrequency plus gemcitabine treatment (mean ± SD, 180±91mg) was statistically significantly smaller compared with gemcitabine alone (661±419mg, P = .0063). CONCLUSIONS: This study provides mechanistic understanding of homologous recombination inhibiting-strategies, such as noninvasive radiofrequency field-induced hyperthermia, to overcome resistance to gemcitabine in refractory human solid tumors.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Carcinoma Hepatocelular/terapia , Replicação do DNA/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Proteínas de Ligação a DNA/antagonistas & inibidores , Desoxicitidina/análogos & derivados , Hipertermia Induzida/métodos , Neoplasias Hepáticas/terapia , Ondas de Rádio , Reparo de DNA por Recombinação/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Replicação do DNA/genética , DNA de Neoplasias/genética , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Exonucleases/antagonistas & inibidores , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Proteína Homóloga a MRE11 , Camundongos , Camundongos SCID , Neoplasias Experimentais/terapia , Terapia por Radiofrequência , Gencitabina
5.
Nanomedicine ; 10(6): 1121-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24650884

RESUMO

Hepatocellular carcinoma (HCC) is one of the most lethal and chemo-refractory cancers, clearly, alternative treatment strategies are needed. We utilized 10nm gold nanoparticles as a scaffold to synthesize nanoconjugates bearing a targeting antibody (cetuximab, C225) and gemcitabine. Loading efficiency of gemcitabine on the gold nanoconjugates was 30%. Targeted gold nanoconjugates in combination with RF were selectively cytotoxic to EGFR expressing Hep3B and SNU449 cells when compared to isotype particles with/without RF (P<0.05). In animal experiments, targeted gold nanoconjugates halted the growth of subcutaneous Hep3B xenografts in combination with RF exposure (P<0.05). These xenografts also demonstrated increased apoptosis, necrosis and decreased proliferation compared to controls. Normal tissues were unharmed. We have demonstrated that non-invasive RF-induced hyperthermia when combined with targeted delivery of gemcitabine is more effective and safe at dosages ~275-fold lower than the current clinically-delivered systemic dose of gemcitabine. FROM THE CLINICAL EDITOR: In a model of hepatocellular carcinoma, the authors demonstrate that non-invasive RF-induced hyperthermia applied with cetuximab targeted delivery of Au NP-gemcitabine conjugate is more effective and safe at dosages ~ 275-fold lower than the current clinically-used systemic dose of gemcitabine.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Carcinoma Hepatocelular/terapia , Desoxicitidina/análogos & derivados , Ouro/uso terapêutico , Neoplasias Hepáticas/terapia , Nanoconjugados/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/química , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Cetuximab , Desoxicitidina/química , Desoxicitidina/uso terapêutico , Sistemas de Liberação de Medicamentos , Ouro/química , Humanos , Hipertermia Induzida , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos Endogâmicos BALB C , Nanoconjugados/química , Gencitabina
6.
Nanomedicine ; 8(7): 1096-105, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22349096

RESUMO

The use of noninvasive radiofrequency (RF) electric fields as an energy source for thermal activation of nanoparticles within cancer cells could be a valuable addition to the emerging field of nano-mediated cancer therapies. Based on investigations of cell death through hyperthermia, and offering the ability for total-body penetration by RF fields, this technique is thought to complement and possibly outperform existing nano-heat treatments that utilize alternative heat production via optical or magnetic stimuli. However, it remains a challenge to understand fully the complex RF-nanoparticle-intracellular interactions before full system optimization can be engineered. Herein we have shown that liver cancer cells can selectively internalize antibody-conjugated gold nanoparticles (AuNPs) through receptor-mediated endocytosis, with the nanoparticles predominantly accumulating and aggregating within cytoplasmic endolysosomes. After exposure to an external RF field, nonaggregated AuNPs absorbed and dissipated energy as heat, causing thermal damage to the targeted cancer cells. We also observed that RF absorption and heat dissipation is dependent on solubility of AuNPs in the colloid, which is pH dependent. Furthermore, by modulating endolysosomal pH it is possible to prevent intracellular AuNP aggregation and enhance thermal cytotoxicity in hepatocellular cancer cells. FROM THE CLINICAL EDITOR: Gold nanoparticles absorb energy from RF fields and can exert hyperthermic effects leading to cell death. Combining this known effect with antibody-based targeting of the nanoparticles, selective cancer specific hyperthermia induced cell death therapies can be designed, as demonstrated in this article.


Assuntos
Ouro/uso terapêutico , Hipertermia Induzida/métodos , Imunoconjugados/uso terapêutico , Neoplasias Hepáticas/terapia , Nanopartículas/uso terapêutico , Terapia por Radiofrequência , Anticorpos/química , Anticorpos/uso terapêutico , Linhagem Celular Tumoral , Ouro/química , Humanos , Concentração de Íons de Hidrogênio , Imunoconjugados/química , Fígado/metabolismo , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Nanopartículas/química , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA