Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Proc Natl Acad Sci U S A ; 114(48): E10281-E10290, 2017 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-29133392

RESUMEN

Cavitation-facilitated microbubble-mediated focused ultrasound therapy is a promising method of drug delivery across the blood-brain barrier (BBB) for treating many neurological disorders. Unlike ultrasound thermal therapies, during which magnetic resonance thermometry can serve as a reliable treatment control modality, real-time control of modulated BBB disruption with undetectable vascular damage remains a challenge. Here a closed-loop cavitation controlling paradigm that sustains stable cavitation while suppressing inertial cavitation behavior was designed and validated using a dual-transducer system operating at the clinically relevant ultrasound frequency of 274.3 kHz. Tests in the normal brain and in the F98 glioma model in vivo demonstrated that this controller enables reliable and damage-free delivery of a predetermined amount of the chemotherapeutic drug (liposomal doxorubicin) into the brain. The maximum concentration level of delivered doxorubicin exceeded levels previously shown (using uncontrolled sonication) to induce tumor regression and improve survival in rat glioma. These results confirmed the ability of the controller to modulate the drug delivery dosage within a therapeutically effective range, while improving safety control. It can be readily implemented clinically and potentially applied to other cavitation-enhanced ultrasound therapies.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/terapia , Doxorrubicina/análogos & derivados , Sistemas de Liberación de Medicamentos/métodos , Glioma/terapia , Terapia por Ultrasonido/métodos , Acústica/instrumentación , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Carbocianinas/química , Carbocianinas/farmacocinética , Cuerpo Estriado/diagnóstico por imagen , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/instrumentación , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacocinética , Glioma/diagnóstico por imagen , Glioma/metabolismo , Glioma/patología , Hipocampo/diagnóstico por imagen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Proteínas Luminiscentes/química , Proteínas Luminiscentes/farmacocinética , Imagen por Resonancia Magnética , Masculino , Microburbujas , Terapia Molecular Dirigida , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Ratas , Ratas Sprague-Dawley , Transductores , Ondas Ultrasónicas
2.
Ultrasound Med Biol ; 46(8): 1998-2006, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32451192

RESUMEN

As focused ultrasound for blood-brain barrier disruption (FUS-BBBD) has progressed to human application, it has become necessary to consider the potential effects of prior irradiation treatments. Using a murine model, we examined the effects of whole-brain irradiation on FUS-BBBD. We first subjected half of the experimental cohort to daily 3-Gy whole-brain irradiation for 10 consecutive days. Then, microbubble-assisted FUS-BBBD was performed unilaterally while the contralateral sides served as unsonicated controls. FUS-BBBD, as evident by measuring the fluorescence yield of extravasated trypan blue dye, was identified at all sites with minimal or no apparent pathology. The peak fluorescence intensity caused by extravasated dye in the sonicated region was 17.5 ± 12.1% higher after radiation and FUS-BBBD than after FUS-BBBD alone, suggesting that prior radiation of the brain may be a sensitizing factor for FUS-BBBD. Radiation alone-without FUS-BBBD-resulted in mild BBB disruption. Hemorrhagic petechiae were observed in 9 of 12 radiated brains, with 77% of them clearly located outside the sonicated area; no petechiae were found in non-irradiated animals. This radiation protocol did not appear to increase the risk for vascular damage associated with FUS-BBBD.


Asunto(s)
Barrera Hematoencefálica/efectos de la radiación , Irradiación Craneana/efectos adversos , Ultrasonido Enfocado de Alta Intensidad de Ablación , Animales , Encéfalo/patología , Encéfalo/efectos de la radiación , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Ratones , Microburbujas/efectos adversos , Imagen Óptica/métodos
3.
Sci Rep ; 10(1): 8766, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32472017

RESUMEN

We investigated controlled blood-brain barrier (BBB) disruption using a low-frequency clinical transcranial MRI-guided focused ultrasound (TcMRgFUS) device and evaluated enhanced delivery of irinotecan chemotherapy to the brain and a rat glioma model. Animals received three weekly sessions of FUS, FUS and 10 mg/kg irinotecan, or irinotecan alone. In each session, four volumetric sonications targeted 36 locations in one hemisphere. With feedback control based on recordings of acoustic emissions, 98% of the sonication targets (1045/1071) reached a pre-defined level of acoustic emission, while the probability of wideband emission (a signature for inertial cavitation) was than 1%. BBB disruption, evaluated by mapping the R1 relaxation rate after administration of an MRI contrast agent, was significantly higher in the sonicated hemisphere (P < 0.01). Histological evaluation found minimal tissue effects. Irinotecan concentrations in the brain were significantly higher (P < 0.001) with BBB disruption, but SN-38 was only detected in <50% of the samples and only with an excessive irinotecan dose. Irinotecan with BBB disruption did not impede tumor growth or increase survival. Overall these results demonstrate safe and controlled BBB disruption with a low-frequency clinical TcMRgFUS device. While irinotecan delivery to the brain was not neurotoxic, it did not improve outcomes in the F98 glioma model.


Asunto(s)
Antineoplásicos/farmacocinética , Barrera Hematoencefálica , Irinotecán/farmacocinética , Imagen por Resonancia Magnética/métodos , Sonicación/métodos , Inhibidores de Topoisomerasa I/farmacocinética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/análisis , Antineoplásicos/uso terapéutico , Edema Encefálico/etiología , Neoplasias Encefálicas/tratamiento farmacológico , Femenino , Glioma/tratamiento farmacológico , Irinotecán/administración & dosificación , Irinotecán/análisis , Irinotecán/uso terapéutico , Masculino , Microburbujas , Proyectos Piloto , Profármacos/administración & dosificación , Profármacos/análisis , Profármacos/farmacocinética , Profármacos/uso terapéutico , Púrpura/etiología , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Sonicación/efectos adversos , Sonicación/instrumentación , Inhibidores de Topoisomerasa I/administración & dosificación , Inhibidores de Topoisomerasa I/análisis , Inhibidores de Topoisomerasa I/uso terapéutico
4.
Phys Med Biol ; 54(11): 3405-19, 2009 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-19436103

RESUMEN

The purpose of this study was to evaluate the feasibility of using localized harmonic motion (LHM) to monitor and control focused ultrasound surgery (FUS) in VX2 tumors in vivo. FUS exposures were performed on 13 VX2 tumors implanted in nine rabbits. The same transducer induced coagulation and generated a localized oscillatory motion by periodically varying the radiation force. A separate diagnostic ultrasound transducer tracked motion by cross-correlating echo signals at different instances. A threshold in motion amplitude was instituted to cease exposure. Coagulation was confirmed by T2-weighted MR images, thermal dose obtained through MR thermometry and histological examinations. For tumor locations achieving coagulation, the LHM amplitude was 9% (p = 0.04) to 57% (p < 0.0001) lower than that before exposure. Control was successful for 74 (69%) out of 108 cases, with 52 (48%) reaching the threshold and achieving coagulation and 22 (21%) never reaching threshold nor coagulating. For the 34 (31%) unsuccessful exposures, 16 (15%) never reached the threshold but coagulation occurred, and 18 (16%) reached threshold without coagulation confirmed. Noise or radio-frequency signal changes explained motion over- or underestimation in 24 (22%) cases; the remaining 10 (9%) had other causes of error. The control was generally successful, but sudden change or noise in the acquired echo signal caused failure. Coagulation after exposure could be validated by comparing amplitudes before and after exposure.


Asunto(s)
Neoplasias Experimentales/terapia , Terapia por Ultrasonido/métodos , Animales , Masculino , Movimiento (Física) , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/patología , Conejos , Temperatura , Resultado del Tratamiento , Ultrasonografía
5.
Ultrasound Med Biol ; 45(8): 2104-2117, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31101446

RESUMEN

High intensity focused ultrasound (HIFU) mechanical ablation is an emerging technique for non-invasive transcranial surgery. Lesions are created by driving inertial cavitation in tissue, which requires significantly less peak pressure and time-averaged power compared with traditional thermal ablation. The utility of mechanical ablation could be extended to the brain provided the pressure threshold for inertial cavitation can be reduced. In this study, the utility of perfluorobutane (PFB)-based phase-shift nanoemulsions (PSNEs) for lowering the inertial cavitation threshold and enabling focal mechanical ablation in the brain was investigated. We successfully achieved vaporization of PFB-based PSNEs at 1.8 MPa with a 740 kHz focused transducer with a pulsed sonication protocol (duty cycle = 1.5%, 10 min sonication) within intact CD-1 mice brains. Evidence is provided showing that a single bolus injection of PSNEs could be used to initiate and sustain inertial cavitation in cerebrovasculature for at least 10 min. Histologic analysis of brain slices after HIFU exposure revealed ischemic and hemorrhagic lesions with dimensions that were comparable to the focal zone of the transducer. These results suggest that PFB-based PSNEs may be used to significantly reduce the inertial cavitation threshold in the cerebrovasculature and, when combined with transcranial focused ultrasound, enable focal intracranial mechanical ablation.


Asunto(s)
Encéfalo/cirugía , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Animales , Emulsiones , Fluorocarburos , Masculino , Ratones , Modelos Animales , Nanotecnología/métodos , Sonicación , Volatilización
6.
Theranostics ; 9(21): 6284-6299, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534551

RESUMEN

The blood-brain barrier (BBB) restricts delivery of most chemotherapy agents to brain tumors. Here, we investigated a clinical focused ultrasound (FUS) device to disrupt the BBB in rats and enhance carboplatin delivery to the brain using the F98 glioma model. Methods: In each rat, 2-3 volumetric sonications (5 ms bursts at 1.1 Hz for 75s) targeted 18-27 locations in one hemisphere. Sonication was combined with Definity microbubbles (10 µl/kg) and followed by intravenous carboplatin (50 mg/kg). Closed-loop feedback control was performed based on acoustic emissions analysis. Results: Safety and reliability were established in healthy rats after three sessions with carboplatin; BBB disruption was induced in every target without significant damage evident in MRI or histology. In tumor-bearing rats, concentrations of MRI contrast agent (Gadavist) were 1.7 and 3.3 times higher in the tumor center and margin, respectively, than non-sonicated tumors (P<0.001). Tissue-to-plasma ratios of intact carboplatin concentrations were increased by 7.3 and 2.9 times in brain and tumor respectively, at one hour after FUS and 4.2 and 2.4 times at four hours. Tumor volume doubling time in rats receiving FUS and carboplatin increased by 96% and 126% compared to rats that received carboplatin alone and non-sonicated controls, respectively (P<0.05); corresponding increases in median survival were 48% and 66% (P<0.01). Conclusion: Overall, this work demonstrates that actively-controlled BBB disruption with a clinical device can enhance carboplatin delivery without neurotoxicity at level that reduces tumor growth and improves survival in an aggressive and infiltrative rat glioma model.


Asunto(s)
Antineoplásicos/farmacocinética , Barrera Hematoencefálica , Neoplasias Encefálicas/tratamiento farmacológico , Carboplatino/farmacocinética , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Animales , Transporte Biológico , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Neoplasias Encefálicas/diagnóstico por imagen , Medios de Contraste/farmacocinética , Femenino , Glioma/diagnóstico por imagen , Masculino , Microburbujas , Ratas , Sonicación , Ultrasonografía
7.
Ultrasound Med Biol ; 34(5): 834-40, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18207311

RESUMEN

This work investigated the effect of ultrasonic frequency on the threshold for blood-brain barrier (BBB) disruption induced by ultrasound pulses combined with an ultrasound contrast agent. Experiments were performed in rabbits using pulsed sonications at 2.04 MHz with peak pressure amplitudes ranging from 0.3 to 2.3 MPa. BBB disruption was evaluated using contrast-enhanced magnetic resonance imaging. The threshold for BBB disruption was estimated using probit regression. Representative samples with similar amounts of contrast enhancement were examined in light microscopy. Results from these experiments were compared with data from previous studies that used ultrasound frequencies between 0.26 and 1.63 MHz. We found that the BBB disruption threshold (value where the probability for disruption was estimated to be 50%) expressed in terms of the peak negative pressure amplitude increased as a function of the frequency. It appeared to be constant, however, when the exposures were expressed as a function of the mechanical index (peak negative pressure amplitude estimated in situ divided by square root of frequency). Regression of data from all frequencies resulted in an estimated mechanical index threshold of 0.46 (95% confidence intervals: 0.42 to 0.50). Histologic examination of representative samples with similar amounts of blood-brain barrier disruption found that the number of regions containing extravasated red blood cells per unit area was substantially lower on average for lower ultrasound frequencies. This data suggests that the mechanical index is a meaningful metric for ultrasound-induced blood-brain barrier disruption, at least for when other parameters that are not taken into account by the mechanical index are not varied. It also suggests that lower frequency sonication produces less red blood cell extravasation per unit area.


Asunto(s)
Barrera Hematoencefálica , Sonicación , Animales , Medios de Contraste , Sistemas de Liberación de Medicamentos , Análisis de los Mínimos Cuadrados , Imagen por Resonancia Magnética , Masculino , Microburbujas , Conejos
8.
Ultrasound Med Biol ; 34(6): 930-7, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18294757

RESUMEN

Previously, it was shown that low-intensity focused ultrasound pulses applied along with an ultrasound contrast agent results in temporary blood-brain barrier (BBB) disruption. This effect could be used for targeted drug delivery in the central nervous system. This study examined the effects of burst length, pulse repetition frequency (PRF), and ultrasound contrast agent dose on the resulting BBB disruption. One hundred nonoverlapping brain locations were sonicated through a craniotomy in experiments in 26 rabbits (ultrasound frequency: 0.69 MHz, burst: 0.1, 1, 10 ms, PRF: 0.5, 1, 2, 5 Hz, duration: 20 s, peak negative pressure amplitude: 0.1 to 1.5 MPa, Optison dosage 50, 100, 250 microl/kg). For each sonication, BBB disruption was evaluated using contrast-enhanced magnetic resonance imaging. The BBB disruption threshold (the pressure amplitude yielding a 50% probability for BBB disruption) was determined using probit regression for the three burst lengths tested. Tissue effects were examined in light microscopy for representative locations with similar amounts of contrast enhancement from each group. While changing the PRF or the Optison dosage did not result in a significant difference in the magnitude of the BBB disruption (p > 0.05), reducing the burst length resulted in significantly less contrast enhancement (p < 0.01). The BBB disruption thresholds were estimated to be 0.69, 0.47 and 0.36 MPa for 0.1, 1 and 10 ms bursts, respectively. No difference was detected in histology between any experimental groups. This data suggests that over the range of parameters tested, BBB disruption is not affected by PRF or ultrasound contrast agent dose. However, both the BBB disruption magnitude and its threshold depend on the burst length.


Asunto(s)
Albúminas/administración & dosificación , Barrera Hematoencefálica/diagnóstico por imagen , Medios de Contraste/administración & dosificación , Fluorocarburos/administración & dosificación , Acústica , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Encéfalo/patología , Relación Dosis-Respuesta a Droga , Imagen por Resonancia Magnética , Masculino , Conejos , Transductores , Ultrasonografía/métodos
9.
Ultrasound Med Biol ; 44(12): 2609-2624, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30201425

RESUMEN

Thermal ablation of solid tumors via focused ultrasound (FUS) is a non-invasive image-guided alternative to conventional surgical resection. However, the usefulness of the technique is limited in vascularized organs because of convection of heat, resulting in long sonication times and unpredictable thermal lesion formation. Acoustic cavitation has been found to enhance heating but requires use of exogenous nuclei and sufficient acoustic monitoring. In this study, we employed phase-shift nanoemulsions (PSNEs) to promote cavitation and incorporated passive acoustic mapping (PAM) alongside conventional magnetic resonance imaging (MRI) thermometry within the bore of a clinical MRI scanner. Simultaneous PAM and MRI thermometry were performed in an in vivo rabbit tumor model, with and without PSNE to promote cavitation. Vaporization and cavitation of the nanoemulsion could be detected using PAM, which led to accelerated heating, monitored with MRI thermometry. The maximum heating assessed from MRI was well correlated with the integrated acoustic emissions, illustrating cavitation-enhanced heating. Examination of tissue revealed thermal lesions that were larger in the presence of PSNE, in agreement with the thermometry data. Using fixed exposure conditions over 94 sonications in multiple animals revealed an increase in the mean amplitude of acoustic emissions and resulting temperature rise, but with significant variability between sonications, further illustrating the need for real-time monitoring. The results indicate the utility of combined PAM and MRI for monitoring of tumor ablation and provide further evidence for the ability of PSNEs to promote cavitation-enhanced lesioning.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Imagen por Resonancia Magnética/métodos , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/cirugía , Termometría/métodos , Animales , Modelos Animales de Enfermedad , Masculino , Conejos
10.
Ultrasound Med Biol ; 33(4): 584-90, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17337109

RESUMEN

We have developed a method to use low-intensity focused ultrasound pulses combined with an ultrasound contrast agent to produce temporary blood-brain barrier disruption (BBBD). This method could provide a means for the targeted delivery of drugs or imaging agents into the brain. In all our previous work, we used Optison as the ultrasound contrast agent. The purpose of this study was to test the feasibility of using the contrast agent Definity for BBBD. A total of 36 non-overlapping locations were sonicated through a craniotomy in experiments in the brains of nine rabbits (four locations per rabbit; ultrasound [US] frequency: 0.69 MHz; burst: 10 ms; pulse repetition frequency (PRF): 1 Hz; duration: 20 s). The peak negative pressure amplitude ranged from 0.2 to 1.5 MPa. An additional 11 locations were sonicated using Optison at pressure amplitude of 0.5 MPa. Definity and Optison dosages were the same as those used clinically for ultrasound imaging: 10 and 50 microl/kg, respectively. The probability for BBBD (determined using MRI contrast agent enhancement) as a function of pressure amplitude was similar to that found earlier with Optison. For both agents, the probability was estimated to be 50% at 0.4 MPa using probit regression. Histologic examination revealed small, isolated areas of extravasated erythrocytes in some locations. At 0.8 MPa and higher, these areas were sometimes accompanied by tiny (dimensions of 100 microm or less) regions of damaged brain parenchyma. The magnitude of the BBBD was larger with Optison than with Definity at 0.5 MPa (signal enhancement: 13.3% +/- 4.4% vs. 8.4% +/- 4.9%; p = 0.04). In addition, more areas with extravasated erythrocytes were observed with Optison (5.0 +/- 3.5 vs. 1.4 +/- 1.9 areas with extravasation in histology section with largest effect; p = 0.03). We concluded that BBBD is possible using Definity at the dosage of contrast agent and the acoustic parameters tested in this study. The probability for BBBD as a function of pressure amplitude and the type of acute tissue effects were similar to what has been observed using Optison. However, under the experimental conditions used in this study, Optison produced a larger effect for the same acoustic pressure amplitude.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Medios de Contraste/farmacocinética , Fluorocarburos/farmacocinética , Ultrasonografía Doppler de Pulso , Albúminas/farmacocinética , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Estudios de Factibilidad , Imagen por Resonancia Magnética , Masculino , Conejos
11.
Ultrasound Med Biol ; 43(2): 469-475, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27789044

RESUMEN

Numerous researchers are investigating the use of microbubble-enhanced ultrasound to disrupt the blood-brain barrier (BBB) and deliver drugs to the brain. This study investigated the impact of using oxygen as a carrier gas for anesthesia on microbubble activity and BBB disruption. Targets in mice were sonicated in combination with administration of Optison microbubbles (100 µL/kg) under isoflurane anesthesia with either oxygen or medical air. A 690-kHz focused ultrasound transducer applied 10-ms bursts at peak pressure amplitudes of 0.46-0.54 MPa (n = 2) or 0.34-0.36 MPa (n = 5). After sonication of two locations in one hemisphere, the carrier gas for the anesthesia was changed and the sonications were repeated in the contralateral hemisphere. The BBB disruption, measured via contrast-enhanced magnetic resonance imaging, was significantly greater (p < 0.001) with medical air than with oxygen. Harmonic emissions were also greater with air (p < 0.001), while the decay rate of the harmonic emissions was 1.5 times faster with oxygen. A good correlation (R2, 0.46) was observed between the harmonic emissions strength and magnetic resonance imaging signal enhancement. At 0.46-0.54 MPa, both the occurrence and strength of wideband emissions were greater with medical air. However, at lower peak pressure amplitudes of 0.34-0.36 MPa, the strength and probability for wideband emissions were higher with oxygen. Little or no effects were observed in histology at 0.34-0.36 MPa. These findings show that use of oxygen as a carrier gas can result in a substantial diminution of BBB disruption. These results should be taken into account when comparing studies from different researchers and in translating this method to humans.


Asunto(s)
Albúminas/farmacocinética , Barrera Hematoencefálica/metabolismo , Medios de Contraste/farmacocinética , Fluorocarburos/farmacocinética , Microburbujas , Oxígeno/farmacocinética , Ondas Ultrasónicas , Animales , Barrera Hematoencefálica/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Ratones , Modelos Animales
12.
J Control Release ; 250: 77-85, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-27742444

RESUMEN

Drug delivery in brain tumors is challenging because of the presence of blood-brain barrier (BBB) and the blood-tumor barrier (BTB). Focused ultrasound (FUS) combined with microbubbles can enhance the permeability of the BTB in brain tumors, as well as disrupting the BBB in the surrounding tissue. In this study, dynamic contrast-enhanced Magnetic Resonance Imaging (DCE-MRI) was used to characterize FUS-induced permeability changes in a rat glioma model and in the normal brain and to investigate the relationship between these changes and the resulting concentration of the chemotherapy agent doxorubicin (DOX). 9L gliosarcoma cells were implanted in both hemispheres in male rats. At day 10-12 after implantation, FUS-induced BTB disruption using 690kHz ultrasound and Definity microbubbles was performed in one of the tumors and in a normal brain region in each animal. After FUS, DOX was administered at a dose of 5.67mg/kg. The resulting DOX concentration was measured via fluorometry at 1 or 24h after FUS. The transfer coefficient Ktrans describing extravasation of the MRI contrast agent Gd-DTPA was significantly increased in both the sonicated tumors and in the normal brain tissue (P<0.001) between the two DCE-MRI acquisitions obtained before and after FUS, while no significant difference was found in the controls (non-sonicated tumor/normal brain tissue). DOX concentrations were also significantly larger than controls in both the sonicated tumors and in the normal tissue volumes at 1 and 24h after sonication. The DOX concentrations were significantly larger (P<0.01) in the control tumors harvested 1h after FUS than in those harvested at 24h, when the tumor concentrations were not significantly different than in the non-sonicated normal brain. In contrast, there was no significant difference in the DOX concentrations between the tumors harvested at 1 and 24h after FUS or in the concentrations measured in the brain at these time points. The transfer coefficient Ktrans for Gd-DTPA and the drug concentrations showed a good linear correlation (R2=0.56). Overall, these data suggest that FUS and microbubbles can not only increase DOX delivery across the BBB and BTB, but that it is retained in the tissue at significantly enhanced levels for at least 24h. Such enhanced retention may increase the potency of this chemotherapy agent and allow for reduced systemic doses. Furthermore, MRI-based estimates of Gd-DTPA transport across these barriers might be useful to estimate local DOX concentrations in the tumor and in the surrounding normal tissue.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Gliosarcoma/tratamiento farmacológico , Animales , Antineoplásicos/metabolismo , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de la radiación , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Medios de Contraste , Preparaciones de Acción Retardada , Doxorrubicina/metabolismo , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Gadolinio DTPA , Gliosarcoma/irrigación sanguínea , Gliosarcoma/metabolismo , Humanos , Masculino , Microburbujas , Permeabilidad , Ratas , Ratas Sprague-Dawley , Ondas Ultrasónicas
13.
J Neurosurg ; 105(3): 445-54, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16961141

RESUMEN

OBJECT: The goal of this study was to explore the feasibility of using low-frequency magnetic resonance (MR) image-guided focused ultrasound as a noninvasive method for the temporary disruption of the blood-brain barrier (BBB) at targeted locations. METHODS: Rabbits were placed inside a clinical 1.5-tesla MR imaging unit, and sites in their brains were targeted for 20-second burst sonications (frequency 260 kHz). The peak pressure amplitude during the burst varied between 0.1 and 0.9 MPa. Each sonication was performed after an intravenous injection of an ultrasound contrast agent (Optison). The disruption of the BBB was evaluated with the aid of an injection of an MR imaging contrast agent (MAG-NEVIST). Additional tests involving the use of MION-47, a 20-nm magnetic nanoparticle contrast agent, were also performed. The animals were killed at different time points between 3 minutes and 5 weeks postsonication, after which light or electron microscopic evaluation was performed. The threshold for BBB disruption was approximately 0.2 MPa. More than 80% of the brain sites sonicated showed BBB disruption when the pressure amplitude was 0.3 MPa; at 0.4 MPa, this percentage was greater than 90%. Tissue necrosis, ischemia, and apoptosis were not found in tissue in which the pressure amplitude was less than 0.4 MPa; however, in a few areas of brain tissue erythrocytes were identified outside blood vessels following exposures of 0.4 MPa or higher. Survival experiments did not show any long-term adverse events. CONCLUSIONS: These results demonstrate that low-frequency ultrasound bursts can induce local, reversible disruption of the BBB without undesired long-term effects. This technique offers a potential noninvasive method for targeted drug delivery in the brain aided by a relatively simple low-frequency device.


Asunto(s)
Barrera Hematoencefálica/fisiología , Ultrasonido , Albúminas/administración & dosificación , Animales , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/ultraestructura , Medios de Contraste/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Óxido Ferrosoférrico/administración & dosificación , Fluorocarburos/administración & dosificación , Gadolinio DTPA/administración & dosificación , Imagen por Resonancia Magnética , Masculino , Nanopartículas , Nanoestructuras , Conejos
14.
Ultrasound Med Biol ; 32(12): 1923-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17169704

RESUMEN

Histologic effects of focused ultrasound (FUS) exposures combined with an ultrasound contrast agent (Optison) were investigated to examine whether the lesions were dominated by apoptosis or necrosis. The rabbit brains (n = 17) were sonicated (1.5 MHz, peak rarefactional pressure amplitude: 1.4 to 8.8 MPa) after Optison was injected intravenously (IV). MRI and light microscopy were used to examine tissue effects. To detect apoptosis, TUNEL staining based on labeling of DNA strand breaks was used. The average number of apoptotic and necrotic cells in 300 x 220 microm microscopic fields were counted in 18 representative lesions. Lesions in the rabbit brains were created at lowered acoustic power levels when FUS was combined with Optison. In histology, the lesions exhibited red blood cell extravasations and destruction of blood vessels. At 4 h after sonication, the lesions lost many cells, and the remaining cells exhibited both necrotic and apoptotic features. Overall, apoptosis dominated; there were, on average, 32.3 +/- 13.2 apoptotic cells per microscopic field compared with only 5.1 +/- 3.4 necrotic cells per field. In conclusion, FUS combined with Optison could produce lesions that are dominated by apoptosis, presumably induced primarily via ischemia after cavitation-produced damage to the brain vasculature.


Asunto(s)
Albúminas/administración & dosificación , Apoptosis/fisiología , Encéfalo/fisiología , Medios de Contraste/administración & dosificación , Fluorocarburos/administración & dosificación , Sonicación , Animales , Encéfalo/patología , Colorantes , Eosina Amarillenta-(YS) , Colorantes Fluorescentes , Hematoxilina , Etiquetado Corte-Fin in Situ/métodos , Inyecciones Intravenosas , Imagen por Resonancia Magnética/métodos , Masculino , Necrosis , Conejos , Colorantes de Rosanilina , Cloruro de Tolonio , Compuestos de Vanadio
15.
J Ther Ultrasound ; 4: 17, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27213044

RESUMEN

We present here biophysical models to gain deeper insights into how an acoustic stimulus might influence or modulate neuronal activity. There is clear evidence that neural activity is not only associated with electrical and chemical changes but that an electro-mechanical coupling is also involved. Currently, there is no theory that unifies the electrical, chemical, and mechanical aspects of neuronal activity. Here, we discuss biophysical models and hypotheses that can explain some of the mechanical aspects associated with neuronal activity: the soliton model, the neuronal intramembrane cavitation excitation model, and the flexoelectricity hypothesis. We analyze these models and discuss their implications on stimulation and modulation of neuronal activity by ultrasound.

16.
J Neurosurg ; 125(6): 1539-1548, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-26848919

RESUMEN

OBJECTIVE Thermal ablation with transcranial MRI-guided focused ultrasound (FUS) is currently under investigation as a less invasive alternative to radiosurgery and resection. A major limitation of the method is that its use is currently restricted to centrally located brain targets. The combination of FUS and a microbubble-based ultrasound contrast agent greatly reduces the ultrasound exposure level needed to ablate brain tissue and could be an effective means to increase the "treatment envelope" for FUS in the brain. This method, however, ablates tissue through a different mechanism: destruction of the microvasculature. It is not known whether nonthermal FUS ablation in substantial volumes of tissue can safely be performed without unexpected effects. The authors investigated this question by ablating volumes in the brains of normal rats. METHODS Overlapping sonications were performed in rats (n = 15) to ablate a volume in 1 hemisphere per animal. The sonications (10-msec bursts at 1 Hz for 60 seconds; peak negative pressure 0.8 MPa) were combined with the ultrasound contrast agent Optison (100 µl/kg). The rats were followed with MRI for 4-9 weeks after FUS, and the brains were examined with histological methods. RESULTS Two weeks after sonication and later, the lesions appeared as cyst-like areas in T2-weighted MR images that were stable over time. Histological examination demonstrated well-defined lesions consisting of a cyst-like cavity that remained lined by astrocytic tissue. Some white matter structures within the sonicated area were partially intact. CONCLUSIONS The results of this study indicate that nonthermal FUS ablation can be used to safely ablate tissue volumes in the brain without unexpected delayed effects. The findings are encouraging for the use of this ablation method in the brain.


Asunto(s)
Técnicas de Ablación/métodos , Encéfalo/cirugía , Medios de Contraste , Procedimientos Neuroquirúrgicos/métodos , Cirugía Asistida por Computador , Ultrasonografía Intervencional , Animales , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Ultrasonografía Intervencional/métodos
17.
J Neurosurg ; 124(5): 1450-9, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26381252

RESUMEN

OBJECT Transcranial MRI-guided focused ultrasound (TcMRgFUS) is an emerging noninvasive alternative to surgery and radiosurgery that is undergoing testing for tumor ablation and functional neurosurgery. The method is currently limited to central brain targets due to skull heating and other factors. An alternative ablative approach combines very low intensity ultrasound bursts and an intravenously administered microbubble agent to locally destroy the vasculature. The objective of this work was to investigate whether it is feasible to use this approach at deep brain targets near the skull base in nonhuman primates. METHODS In 4 rhesus macaques, targets near the skull base were ablated using a clinical TcMRgFUS system operating at 220 kHz. Low-duty-cycle ultrasound exposures (sonications) were applied for 5 minutes in conjunction with the ultrasound contrast agent Definity, which was administered as a bolus injection or continuous infusion. The acoustic power level was set to be near the inertial cavitation threshold, which was measured using passive monitoring of the acoustic emissions. The resulting tissue effects were investigated with MRI and with histological analysis performed 3 hours to 1 week after sonication. RESULTS Thirteen targets were sonicated in regions next to the optic tract in the 4 animals. Inertial cavitation, indicated by broadband acoustic emissions, occurred at acoustic pressure amplitudes ranging from 340 to 540 kPa. MRI analysis suggested that the lesions had a central region containing red blood cell extravasations that was surrounded by edema. Blood-brain barrier disruption was observed on contrast-enhanced MRI in the lesions and in a surrounding region corresponding to the prefocal area of the FUS system. In histology, lesions consisting of tissue undergoing ischemic necrosis were found in all regions that were sonicated above the inertial cavitation threshold. Tissue damage in prefocal areas was found in several cases, suggesting that in those cases the sonication exceeded the inertial cavitation threshold in the beam path. CONCLUSIONS It is feasible to use a clinical TcMRgFUS system to ablate skull base targets in nonhuman primates at time-averaged acoustic power levels at least 2 orders of magnitude below what is needed for thermal ablation with this device. The results point to the risks associated with the method if the exposure levels are not carefully controlled to avoid inertial cavitation in the acoustic beam path. If methods can be developed to provide this control, this nonthermal approach could greatly expand the use of TcMRgFUS for precisely targeted ablation to locations across the entire brain.


Asunto(s)
Encéfalo/cirugía , Imagen por Resonancia Magnética Intervencional/métodos , Procedimientos Quirúrgicos Ultrasónicos/métodos , Ultrasonografía Intervencional/métodos , Animales , Encéfalo/patología , Estudios de Factibilidad , Macaca mulatta , Imagen por Resonancia Magnética Intervencional/instrumentación , Base del Cráneo/patología , Base del Cráneo/cirugía , Procedimientos Quirúrgicos Ultrasónicos/instrumentación , Ultrasonografía Intervencional/instrumentación
18.
Ultrasound Med Biol ; 42(2): 481-92, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26617243

RESUMEN

The purpose of this study was to investigate the effects on the brain of multiple sessions of blood-brain barrier (BBB) disruption using focused ultrasound (FUS) in combination with micro-bubbles over a range of acoustic exposure levels. Six weekly sessions of FUS, using acoustical pressures between 0.66 and 0.80 MPa, were performed under magnetic resonance guidance. The success and degree of BBB disruption was estimated by signal enhancement of post-contrast T1-weighted imaging of the treated area. Histopathological analysis was performed after the last treatment. The consequences of repeated BBB disruption varied from no indications of vascular damage to signs of micro-hemorrhages, macrophage infiltration, micro-scar formations and cystic cavities. The signal enhancement on the contrast-enhanced T1-weighted imaging had limited value for predicting small-vessel damage. T2-weighted imaging corresponded well with the effects on histopathology and could be used to study treatment effects over time. This study demonstrates that repeated BBB disruption by FUS can be performed with no or limited damage to the brain tissue.


Asunto(s)
Barrera Hematoencefálica/patología , Barrera Hematoencefálica/efectos de la radiación , Ondas de Choque de Alta Energía , Sonicación/métodos , Animales , Barrera Hematoencefálica/fisiología , Dosis de Radiación , Ratas , Ratas Sprague-Dawley
19.
Phys Med Biol ; 61(21): 7664-7687, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27740941

RESUMEN

This study investigated thermal ablation and skull-induced heating with a 230 kHz transcranial MRI-guided focused ultrasound (TcMRgFUS) system in nonhuman primates. We evaluated real-time acoustic feedback and aimed to understand whether cavitation contributed to the heating and the lesion formation. In four macaques, we sonicated thalamic targets at acoustic powers of 34-560 W (896-7590 J). Tissue effects evaluated with MRI and histology were compared to MRI-based temperature and thermal dose measurements, acoustic emissions recorded during the experiments, and acoustic and thermal simulations. Peak temperatures ranged from 46 to 57 °C, and lesions were produced in 5/8 sonicated targets. A linear relationship was observed between the applied acoustic energy and both the focal and brain surface heating. Thermal dose thresholds were 15-50 cumulative equivalent minutes at 43 °C, similar to prior studies at higher frequencies. Histology was also consistent with earlier studies of thermal effects in the brain. The system successfully controlled the power level and maintained a low level of cavitation activity. Increased acoustic emissions observed in 3/4 animals occurred when the focal temperature rise exceeded approximately 16 °C. Thresholds for thermally-significant subharmonic and wideband emissions were 129 and 140 W, respectively, corresponding to estimated pressure amplitudes of 2.1 and 2.2 MPa. Simulated focal heating was consistent with the measurements for sonications without thermally-significant acoustic emissions; otherwise it was consistently lower than the measurements. Overall, these results suggest that the lesions were produced by thermal mechanisms. The detected acoustic emissions, however, and their association with heating suggest that cavitation might have contributed to the focal heating. Compared to earlier work with a 670 kHz TcMRgFUS system, the brain surface heating was substantially reduced and the focal heating was higher with this 230 kHz system, suggesting that a reduced frequency can increase the treatment envelope for TcMRgFUS and potentially reduce the risk of skull heating.


Asunto(s)
Encéfalo/cirugía , Hipertermia Inducida/métodos , Imagen por Resonancia Magnética/métodos , Modelos Teóricos , Ultrasonografía/métodos , Animales , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Femenino , Macaca mulatta , Masculino , Cráneo/diagnóstico por imagen , Cráneo/patología , Cráneo/cirugía , Ultrasonografía/instrumentación
20.
Ultrasound Med Biol ; 31(11): 1527-37, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16286030

RESUMEN

Focused ultrasound offers a method to disrupt the blood-brain barrier (BBB) noninvasively and reversibly at targeted locations. The purpose of this study was to test the safety of this method by searching for ischemia and apoptosis in areas with BBB disruption induced by pulsed ultrasound in the presence of preformed gas bubbles and by looking for delayed effects up to one month after sonication. Pulsed ultrasound exposures (sonications) were performed in the brains of 24 rabbits under monitoring by magnetic resonance imaging (MRI) (ultrasound: frequency = 1.63 MHz, burst length = 100 ms, PRF = 1 Hz, duration = 20 s, pressure amplitude 0.7 to 1.0 MPa). Before sonication, an ultrasound contrast agent (Optison, GE Healthcare, Milwaukee, WI, USA) was injected IV. BBB disruption was confirmed with contrast-enhanced MR images. Whole brain histologic examination was performed using haematoxylin and eosin staining for general histology, vanadium acid fuchsin-toluidine blue staining for ischemic neurons and TUNEL staining for apoptosis. The main effects observed were tiny regions of extravasated red blood cells scattered around the sonicated locations, indicating affected capillaries. Despite these vasculature effects, only a few cells in some of the sonicated areas showed evidence for apoptosis or ischemia. No ischemic or apoptotic regions were detected that would indicate a compromised blood supply was induced by the sonications. No delayed effects were observed either by MRI or histology up to 4 wk after sonication. Ultrasound-induced BBB disruption is possible without inducing substantial vascular damage that would result in ischemic or apoptotic death to neurons. These findings indicate that this method is safe for targeted drug delivery, at least when compared with the currently available invasive methods.


Asunto(s)
Barrera Hematoencefálica/diagnóstico por imagen , Ecoencefalografía/métodos , Imagen por Resonancia Magnética , Albúminas/administración & dosificación , Animales , Apoptosis , Barrera Hematoencefálica/patología , Encéfalo/patología , Isquemia Encefálica/patología , Recuento de Células , Ecoencefalografía/instrumentación , Fluorocarburos/administración & dosificación , Etiquetado Corte-Fin in Situ , Masculino , Microburbujas , Modelos Animales , Neuronas/patología , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA