Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
PLoS One ; 7(4): e35509, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22536396

RESUMEN

While Magnetic Resonance Thermometry (MRT) has been extensively utilized for non-invasive temperature measurement, there is limited data on the use of high field (≥7T) scanners for this purpose. MR-guided Focused Ultrasound (MRgFUS) is a promising non-invasive method for localized hyperthermia and drug delivery. MRT based on the temperature sensitivity of the proton resonance frequency (PRF) has been implemented in both a tissue phantom and in vivo in a mouse Met-1 tumor model, using partial parallel imaging (PPI) to speed acquisition. An MRgFUS system capable of delivering a controlled 3D acoustic dose during real time MRT with proportional, integral, and derivative (PID) feedback control was developed and validated. Real-time MRT was validated in a tofu phantom with fluoroptic temperature measurements, and acoustic heating simulations were in good agreement with MR temperature maps. In an in vivo Met-1 mouse tumor, the real-time PID feedback control is capable of maintaining the desired temperature with high accuracy. We found that real time MR control of hyperthermia is feasible at high field, and k-space based PPI techniques may be implemented for increasing temporal resolution while maintaining temperature accuracy on the order of 1°C.


Asunto(s)
Hipertermia Inducida , Imagen por Resonancia Magnética , Neoplasias Mamarias Experimentales/terapia , Termómetros , Terapia por Ultrasonido , Animales , Línea Celular Tumoral , Femenino , Ratones , Modelos Biológicos , Trasplante de Neoplasias , Alimentos de Soja , Temperatura , Agua
2.
IEEE Trans Biomed Eng ; 58(7): 2002-12, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21402506

RESUMEN

Gold nanoparticles (GNPs) are nontoxic, can be functionalized with ligands, and preferentially accumulate in tumors. We have developed a 13.56-MHz RF-electromagnetic field (RF-EM) delivery system capable of generating high E-field strengths required for noninvasive, noncontact heating of GNPs. The bulk heating and specific heating rates were measured as a function of NP size and concentration. It was found that heating is both size and concentration dependent, with 5 nm particles producing a 50.6 ± 0.2 °C temperature rise in 30 s for 25 µg/mL gold (125 W input). The specific heating rate was also size and concentration dependent, with 5 nm particles producing a specific heating rate of 356 ± 78 kW/g gold at 16 µg/mL (125 W input). Furthermore, we demonstrate that cancer cells incubated with GNPs are killed when exposed to 13.56 MHz RF-EM fields. Compared to cells that were not incubated with GNPs, three out of four RF-treated groups showed a significant enhancement of cell death with GNPs (p<0.05). GNP-enhanced cell killing appears to require temperatures above 50 °C for the experimental parameters used in this study. Transmission electron micrographs show extensive vacuolization with the combination of GNPs and RF treatment.


Asunto(s)
Oro/química , Hipertermia Inducida/instrumentación , Nanopartículas del Metal/química , Neoplasias/terapia , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Ácido Cítrico , Campos Electromagnéticos , Diseño de Equipo , Calor , Humanos , Hipertermia Inducida/métodos , Microscopía Electrónica de Transmisión , Nanotecnología , Tamaño de la Partícula
3.
Artículo en Inglés | MEDLINE | ID: mdl-21156363

RESUMEN

Mild hyperthermia is increasingly important for the activation of temperature-sensitive drug delivery vehicles. Noninvasive ultrasound thermometry based on a 2-D speckle tracking algorithm was examined in this study. Here, a commercial ultrasound scanner, a customized co-linear array transducer, and a controlling PC system were used to generate mild hyperthermia. Because the co-linear array transducer is capable of both therapy and imaging at widely separated frequencies, RF image frames were acquired during therapeutic insonation and then exported for off-line analysis. For in vivo studies in a mouse model, before temperature estimation, motion correction was applied between a reference RF frame and subsequent RF frames. Both in vitro and in vivo experiments were examined; in the in vitro and in vivo studies, the average temperature error had a standard deviation of 0.7°C and 0.8°C, respectively. The application of motion correction improved the accuracy of temperature estimation, where the error range was 1.9 to 4.5°C without correction compared with 1.1 to 1.0°C following correction. This study demonstrates the feasibility of combining therapy and monitoring using a commercial system. In the future, real-time temperature estimation will be incorporated into this system.


Asunto(s)
Hipertermia Inducida/métodos , Procesamiento de Señales Asistido por Computador , Termografía/métodos , Ultrasonografía/métodos , Algoritmos , Animales , Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Femenino , Procesamiento de Imagen Asistido por Computador/métodos , Ratones , Trasplante de Neoplasias , Fantasmas de Imagen , Sefarosa , Temperatura , Termografía/instrumentación , Transductores , Ultrasonografía/instrumentación
4.
IEEE Trans Biomed Eng ; 57(1): 155-66, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20064754

RESUMEN

A new system is presented for generating controlled tissue heating with a clinical ultrasound scanner, and initial in vitro and in vivo results are presented that demonstrate both transient and sustained heating in the mild-hyperthermia range of 37 ( degrees )C-42 ( degrees )C. The system consists of a Siemens Antares ultrasound scanner, a custom dual-frequency three-row transducer array and an external temperature feedback control system. The transducer has two outer rows that operate at 1.5 MHz for tissue heating and a center row that operates at 5 MHz for B-mode imaging to guide the therapy. We compare the field maps obtained using a hydrophone against calculations of the ultrasound beam based on monochromatic and linear assumptions. Using the finite-difference time-domain (FDTD) method, we compare predicted time-dependent thermal profiles to measured profiles for soy tofu as a tissue-mimicking phantom. In vitro results show differential heating of 6 ( degrees )C for chicken breast and tofu. In vivo tests of the system were performed on three mice bearing Met-1 tumors, which is a model of aggressive, metastatic, and highly vascular breast cancer. In superficially implanted tumors, we demonstrate controlled heating to 42 ( degrees )C. We show that the system is able to maintain the temperature to within 0.1 ( degrees )C of the desired temperature both in vitro and in vivo.


Asunto(s)
Hipertermia Inducida/métodos , Neoplasias Experimentales/diagnóstico por imagen , Terapia por Ultrasonido/métodos , Animales , Pollos , Hipertermia Inducida/instrumentación , Carne , Ratones , Alimentos de Soja , Terapia por Ultrasonido/instrumentación , Ultrasonografía
5.
J Control Release ; 141(2): 128-36, 2010 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-19748536

RESUMEN

To provide a continuous and prolonged delivery of the substrate D-luciferin for bioluminescence imaging in vivo, luciferin was encapsulated into liposomes using either the pH gradient or acetate gradient method. Under optimum loading conditions, 0.17 mg luciferin was loaded per mg of lipid with 90-95% encapsulation efficiency, where active loading was 6 to 18-fold higher than that obtained with passive loading. Liposomal luciferin in a long-circulating formulation had good shelf stability, with 10% release over 3-month storage at 4 degrees C. Pharmacokinetic profiles of free and liposomal luciferin were then evaluated in transgenic mice expressing luciferase. In contrast to rapid in vivo clearance of free luciferin (t(1/2)=3.54 min), luciferin encapsulated into long-circulating liposomes showed a prolonged release over 24h. The first-order release rate constant of luciferin from long-circulating liposomes, as estimated from the best fit of the analytical model to the experimental data, was 0.01 h(-1). Insonation of luciferin-loaded temperature-sensitive liposomes directly injected into one tumor of Met1-luc tumor-bearing mice resulted in immediate emission of light. Systemic injection of luciferin-loaded long-circulating liposomes into Met1-luc tumor-bearing mice, followed by unilateral ultrasound-induced hyperthermia, produced a gradual increase in radiance over time, reaching a peak at 4-7 h post-ultrasound.


Asunto(s)
Benzotiazoles/administración & dosificación , Sistemas de Liberación de Medicamentos , Luminiscencia , Sustancias Luminiscentes/administración & dosificación , Neoplasias Mamarias Experimentales/patología , Animales , Benzotiazoles/química , Benzotiazoles/farmacocinética , Línea Celular Tumoral , Química Farmacéutica , Preparaciones de Acción Retardada , Composición de Medicamentos , Estabilidad de Medicamentos , Femenino , Concentración de Iones de Hidrógeno , Hipertermia Inducida , Inyecciones Intralesiones , Inyecciones Intravenosas , Liposomas , Luciferasas/genética , Luciferasas/metabolismo , Sustancias Luminiscentes/química , Sustancias Luminiscentes/farmacocinética , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Permeabilidad , Solubilidad , Temperatura , Transfección , Ultrasonido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA