Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Phys Med Biol ; 69(14)2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38914104

RESUMEN

Objective.Pulsed focused ultrasound (FUS) can deliver therapeutics to the brain by using intravenous microbubbles (MBs) to open the blood-brain barrier (BBB). MB emissions indicate treatment outcomes, like BBB opening (harmonics) and damage (broadband). Typically, a pulse repetition frequency (PRF) of 1 Hz is used, but the effect of PRF on MBs is not fully understood. We investigated the effect of PRF on MB activity and tracer delivery.Approach.The effect of PRF (0.125, 0.25, 0.5, 1, and 2 Hz) on MB activity was monitored through harmonic and wideband emissions during FUS sonications of the rat brain at 274.3 kHz. BBB opening was quantified through fluorescence imaging to estimate the concentration of Trypan Blue (TB) dye following a 75-pulse FUS exposure for PRFs of 1 and 0.25 Hz.Main results.At a fixed acoustic pressure, the percentage change in maximum harmonic amplitude compared to the control (PRF = 1 Hz) decreased with increasing PRF, with a median change of 73.8% at 0.125 Hz and -38.3% at 2 Hz. There was no difference in the pressure threshold for broadband emissions between PRFs of 0.25 and 1 Hz. PRF = 0.25 Hz, led to a 68.2% increase in the mean concentration of TB measured after FUS, with a 53.9% increase in the mean harmonic sum, compared with PRF = 1 Hz. Harmonic emissions-based control at PRF = 0.25 Hz yielded similar TB delivery, with less damage at histology, compared with 1 Hz.Significance.For a fixed number of FUS pulses, reducing the PRF was shown to increase the magnitude of harmonic emissions and TB delivery, but not the threshold for broadband emissions. While further research is necessary to understand the mechanisms involved, these results may be useful to improve clinical safety margins and sensitivity to detecting small harmonic signals from cavitating MBs.


Asunto(s)
Barrera Hematoencefálica , Sistemas de Liberación de Medicamentos , Microburbujas , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de la radiación , Animales , Ratas , Ondas Ultrasónicas , Ratas Sprague-Dawley , Masculino , Sonicación/métodos
2.
J Control Release ; 372: 194-208, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38897294

RESUMEN

PURPOSE: We report our experience disrupting the blood-brain barrier (BBB) to improve drug delivery in glioblastoma patients receiving temozolomide chemotherapy. The goals of this retrospective analysis were to compare MRI-based measures of BBB disruption and vascular damage to the exposure levels, acoustic emissions data, and acoustic simulations. We also simulated the cavitation detectors. METHODS: Monthly BBB disruption (BBBD) was performed using a 220 kHz hemispherical phased array focused ultrasound system (Exablate Neuro, InSightec) and Definity microbubbles (Lantheus) over 38 sessions in nine patients. Exposure levels were actively controlled via the cavitation dose obtained by monitoring subharmonic acoustic emissions. The acoustic field and sensitivity profile of the cavitation detection system were simulated. Exposure levels and cavitation metrics were compared to the level of BBBD evident in contrast-enhanced MRI and to hypointense regions in T2*-weighted MRI. RESULTS: Our treatment strategy evolved from using a relatively high cavitation dose goal to a lower goal and longer sonication duration and ultimately resulted in BBBD across the treatment volume with minimal petechiae. Subsonication-level feedback control of the exposure using acoustic emissions also improved consistency. Simulations of the acoustic field suggest that reflections and standing waves appear when the focus is placed near the skull, but their effects can be mitigated with aberration correction. Simulating the cavitation detectors suggest variations in the sensitivity profile across the treatment volume and between patients. A correlation was observed with the cavitation dose, BBBD and petechial hemorrhage in 8/9 patients, but substantial variability was evident. Analysis of the cavitation spectra found that most bursts did not contain wideband emissions, a signature of inertial cavitation, but biggest contribution to the cavitation dose - the metric used to control the procedure - came from bursts with wideband emissions. CONCLUSION: Using a low subharmonic cavitation dose with a longer duration resulted in BBBD with minimal petechiae. The correlation between cavitation dose and outcomes demonstrates the benefits of feedback control based on acoustic emissions, although more work is needed to reduce variability. Acoustic simulations could improve focusing near the skull and inform our analysis of acoustic emissions. Monitoring additional frequency bands and improving the sensitivity of the cavitation detection could provide signatures of microbubble activity associated with BBB disruption that were undetected here and could improve our ability to achieve BBB disruption without vascular damage.

3.
IEEE Trans Biomed Eng ; PP2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38814760

RESUMEN

OBJECTIVE: Holographic methods can be used with phased array transducers to shape an ultrasound field. We tested a simple method to create holograms with a hemispherical 1024-element phased array transducer and explored how it could benefit ultrasound-mediated blood-brain barrier (BBB) disruption. METHODS: With this method, individual acoustic simulations for each element of the transducer were simultaneously loaded into computer memory. Each element's phase was systematically modulated until the combined field matched a desired pattern. The method was evaluated with a 220 kHz transducer being tested clinically to enhance drug delivery via BBB disruption. The holograms were evaluated in a tissue-mimicking phantom and in vivo in experiments disrupting the BBB in rats and in a macaque. We also explored whether this approach could mitigate secondary reflections from the skull using simulations of transcranial focusing in clinical treatments of transcranial sonication for BBB disruption. RESULTS: This approach can enlarge the focal volume in a patient-specific manner and could reduce the number of sonication targets needed to disrupt large volumes, improve the homogeneity of the disruption, and improve our ability to detect microbubble activity in tissues with low vascular density. Simulations suggest that the method could also mitigate secondary reflections during transcranial sonication.

4.
Sci Rep ; 14(1): 4831, 2024 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-38413663

RESUMEN

Focused Ultrasound (FUS) has been shown to sensitize tumors outside the brain to Radiotherapy (RT) through increased ceramide-mediated apoptosis. This study investigated the effects of FUS + RT in healthy rodent brains and F98 gliomas. Tumors, or striata in healthy rats, were targeted with microbubble-mediated, pulsed FUS (220 kHz, 102-444 kPa), followed by RT (4, 8, 15 Gy). FUS + RT (8, 15 Gy) resulted in ablative lesions, not observed with FUS or RT only, in healthy tissue. Lesions were visible using Magnetic Resonance Imaging (MRI) within 72 h and persisted until 21 days post-treatment, indicating potential applications in ablative neurosurgery. In F98 tumors, at 8 and 15 Gy, where RT only had significant effects, FUS + RT offered limited improvements. At 4 Gy, where RT had limited effects compared with untreated controls, FUS + RT reduced tumor volumes observed on MRI by 45-57%. However, survival benefits were minimal (controls: 27 days, RT: 27 days, FUS + RT: 28 days). Histological analyses of tumors 72 h after FUS + RT (4 Gy) showed 93% and 396% increases in apoptosis, and 320% and 336% increases in vessel-associated ceramide, compared to FUS and RT only. Preliminary evidence shows that FUS + RT may improve treatment of glioma, but additional studies are required to optimize effect size.


Asunto(s)
Neoplasias Encefálicas , Glioma , Ratas , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/radioterapia , Microburbujas , Línea Celular Tumoral , Glioma/diagnóstico por imagen , Glioma/radioterapia , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Ceramidas/farmacología , Barrera Hematoencefálica
5.
Adv Mater ; 35(52): e2308150, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37949438

RESUMEN

Microbubbles (MB) are widely used for ultrasound (US) imaging and drug delivery. MB are typically spherically shaped, due to surface tension. When heated above their glass transition temperature, polymer-based MB can be mechanically stretched to obtain an anisotropic shape, endowing them with unique features for US-mediated blood-brain barrier (BBB) permeation. It is here shown that nonspherical MB can be surface-modified with BBB-specific targeting ligands, thereby promoting binding to and sonopermeation of blood vessels in the brain. Actively targeted rod-shaped MB are generated via 1D stretching of spherical poly(butyl cyanoacrylate) MB and via subsequently functionalizing their shell with antitransferrin receptor (TfR) antibodies. Using US and optical imaging, it is demonstrated that nonspherical anti-TfR-MB bind more efficiently to BBB endothelium than spherical anti-TfR-MB, both in vitro and in vivo. BBB-associated anisotropic MB produce stronger cavitation signals and markedly enhance BBB permeation and delivery of a model drug as compared to spherical BBB-targeted MB. These findings exemplify the potential of antibody-modified nonspherical MB for targeted and triggered drug delivery to the brain.


Asunto(s)
Barrera Hematoencefálica , Microburbujas , Receptores de Transferrina , Sonicación , Barrera Hematoencefálica/metabolismo , Receptores de Transferrina/metabolismo , Ligandos , Sistemas de Liberación de Medicamentos , Anticuerpos , Animales , Ratones , Femenino , Ratones Endogámicos BALB C , Línea Celular , Células Endoteliales/metabolismo
6.
Ultrasound Med Biol ; 47(7): 1747-1760, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33879388

RESUMEN

Transient opening of the blood-spinal cord barrier has the potential to improve drug delivery options to the spinal cord. We previously developed short-burst phase-keying exposures to reduce focal depth of field and mitigate standing waves in the spinal canal. However, optimal short-burst phase-keying parameters for drug delivery have not been identified. Here, the effects of pressure, treatment duration, pulse length, burst repetition frequency and burst length on resulting tissue effects were investigated. Increased in situ pressures (0.23-0.33 MPa) led to increased post-treatment T1-weighted contrast enhancement in magnetic resonance imaging (p = 0.015). Increased treatment duration (120 vs. 300 s) led to increased enhancement, but without statistical significance (p = 0.056). Increased burst repetition frequency (20 vs. 40 kHz) yielded a non-significant increase in enhancement (p = 0.064) but corresponded with increased damage observed on histology. No difference was observed in enhancement between pulse lengths of 2 and 10 ms (p = 0.912), corresponding with a sharp drop in the recorded second harmonic signal during the first 2 ms of the pulse. Increasing the burst length from two to five cycles (514 kHz) led to increased enhancement (p = 0.014). Results indicate that increasing the burst length may be the most effective method to enhance drug delivery. Additionally, shorter pulse lengths may allow more interleaved targets, and therefore a larger treatment volume, within one sonication.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Médula Espinal/anatomía & histología , Médula Espinal/diagnóstico por imagen , Animales , Barrera Hematoencefálica , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Ultrasonografía
7.
IEEE Trans Biomed Eng ; 67(5): 1387-1396, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31442968

RESUMEN

OBJECTIVE: We previously developed short burst, phase keying (SBPK) focused ultrasound (FUS) to mitigate standing waves in the human vertebral canal. Here, we show microbubble emissions from these pulses can be detected through the human vertebral arch and that these pulses are effective for blood-spinal cord barrier (BSCB) opening. METHODS: At f0 = 514 kHz, circulating microbubbles were sonicated through ex vivo human vertebrae (60 kPa-1 MPa) using a dual-aperture approach and SBPK exposures engineered to incorporate pulse inversion (PI). Signals from a 250 kHz receiver were analyzed using PI, short-time Fourier analysis and the maximum projection over the pulse train. In rats (n = 14), SBPK FUS+microbubbles was applied to 3 locations/spinal cord at fixed pressures (∼0.20-0.47 MPa). MRI and histology were used to assess opening and tissue damage. RESULTS: In human vertebrae between 0.2-0.4 MPa, PI amplified the microbubble/baseline ratio at f0/2 and 2f0 by 202 ± 40% (132-291%). This was maximal at 0.4 MPa, coinciding with the onset of broadband emissions. In vivo, opening was achieved at 40/42 locations, with mean MRI enhancement of 46 ± 32%(16%-178%). Using PI, f0/2 was detected at 14/40 opening locations. At the highest pressures (f0/2 present) histology showed widespread bleeding throughout the focal region. At the lowest pressures, opening was achieved without bleeding. CONCLUSION: This study confirmed that PI can increase sensitivity to transvertebral detection of microbubble signals. Preliminary in vivo investigations show that SBPK FUS can increase BSCB permeability without tissue damage. SIGNIFICANCE: SBPK is a clinically relevant pulse scheme and, in combination with PI, provides a means of mediating and monitoring BSCB opening noninvasively.


Asunto(s)
Barrera Hematoencefálica , Microburbujas , Animales , Imagen por Resonancia Magnética , Ratas , Médula Espinal/diagnóstico por imagen , Columna Vertebral , Ultrasonografía
8.
Theranostics ; 10(17): 7758-7774, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32685018

RESUMEN

Blood-spinal cord barrier opening, using focused ultrasound and microbubbles, has the potential to improve drug delivery for the treatment of spinal cord pathologies. Delivering and detecting ultrasound through the spine is a challenge for clinical translation. We have previously developed short burst, phase keying exposures, which can be used in a dual-aperture configuration to address clinical scale targeting challenges. Here we demonstrate the use of these pulses for blood-spinal cord barrier opening, in vivo in pigs. Methods: The spinal cords of Yorkshire pigs (n=8) were targeted through the vertebral laminae, in the lower thoracic to upper lumbar region using focused ultrasound (486 kHz) and microbubbles. Four animals were treated with a combination of pulsed sinusoidal exposures (1.0-4.0 MPa, non-derated) and pulsed short burst, phase keying exposures (1.0-2.0 MPa, non-derated). Four animals were treated using ramped short burst, phase keying exposures (1.8-2.1 MPa, non-derated). A 250 kHz narrowband receiver was used to detect acoustic emissions from microbubbles. Blood-spinal cord barrier opening was assessed by the extravasation of Evans blue dye. Histological analysis of the spinal cords was used to assess tissue damage and excised vertebral samples were used in benchtop experiments. Results: Ramped short burst, phase keying exposures successfully modified the blood-spinal cord barrier at 16/24 targeted locations, as assessed by the extravasation of Evans blue dye. At 4 of these locations, opening was confirmed with minimal adverse effects observed through histology. Transmission measurements through excised vertebrae indicated a mean transmission of (47.0 ± 7.0 %) to the target. Conclusions: This study presents the first evidence of focused ultrasound-induced blood-spinal cord barrier opening in a large animal model, through the intact spine. This represents an important step towards clinical translation.


Asunto(s)
Barrera Hematoencefálica/efectos de la radiación , Sistemas de Liberación de Medicamentos/métodos , Enfermedades de la Médula Espinal/tratamiento farmacológico , Médula Espinal/efectos de la radiación , Ondas Ultrasónicas , Animales , Barrera Hematoencefálica/metabolismo , Humanos , Masculino , Microburbujas , Modelos Animales , Médula Espinal/irrigación sanguínea , Médula Espinal/metabolismo , Sus scrofa
9.
Artículo en Inglés | MEDLINE | ID: mdl-30273151

RESUMEN

Focused ultrasound has been shown to increase the permeability of the blood-brain barrier and its feasibility for opening the blood-spinal cord barrier has also been demonstrated in small animal models, with great potential to impact the treatment of spinal cord (SC) disorders. For clinical translation, challenges to transvertebral focusing of ultrasound energy on the human spinal canal, such as a focal depth of field and standing-wave formation, must be addressed. A dual-aperture approach using multifrequency and phase-shift keying (PSK) strategies for achieving a controlled focus in human thoracic vertebrae was investigated through numerical simulations and benchtop experiments in ex vivo human vertebrae. An ~85% reduction in the focal depth of field was achieved compared to a single-aperture approach at 564 kHz. Short-burst (two-cycle) excitations in combination with PSK were found to suppress the formation of standing waves in ex vivo human thoracic vertebrae when focusing through the vertebral laminae. The results make an important contribution toward the development of a clinical-scale approach for targeting ultrasound therapy to the SC.


Asunto(s)
Canal Medular/diagnóstico por imagen , Vértebras Torácicas/diagnóstico por imagen , Terapia por Ultrasonido/métodos , Ultrasonografía/métodos , Simulación por Computador , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA