Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ACS Pharmacol Transl Sci ; 7(6): 1864-1873, 2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38898951

RESUMEN

Design strategies that lead to a more focused in vivo delivery of functionalized nanoparticles (NPs) and their cargo can potentially maximize their therapeutic efficiency while reducing systemic effects, broadening their clinical applications. Here, we report the development of a noncovalent labeling approach where immunoglobulin G (IgG)-decorated NPs can be directed to a cancer cell using a simple, linear bispecific protein adaptor, termed MFE23-ZZ. MFE23-ZZ was created by fusing a single-chain fragment variable domain, termed MFE23, recognizing carcinoembryonic antigen (CEA) expressed on tumor cells, to a small protein ZZ module, which binds to the Fc fragment of IgG. As a proof of concept, monoclonal antibodies (mAbs) were generated against a NP coat protein, namely, gas vesicle protein A (GvpA) of Halobacterium salinarum gas vesicles (GVs). The surface of each GV was therapeutically derivatized with the photoreactive agent chlorin e6 (Ce6GVs) and anti-GvpA mAbs were subsequently bound to GvpA on the surface of each Ce6GV. The bispecific ligand MFE23-ZZ was then bound to mAb-decorated Ce6GVs via their Fc domain, resulting in a noncovalent tripartite complex, namely, MFE23.ZZ-2B10-Ce6GV. This complex enhanced the intracellular uptake of Ce6GVs into human CEA-expressing murine MC38 colon carcinoma cells (MC38.CEA) relative to the CEA-negative parental cell line MC38 in vitro, making them more sensitive to light-induced cell killing. These results suggest that the surface of NP can be rapidly and noncovalently functionalized to target tumor-associated antigen-expressing tumor cells using simple bispecific linkers and any IgG-labeled cargo. This noncovalent approach is readily applicable to other types of functionalized NPs.

2.
Theranostics ; 14(5): 1794-1814, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38505609

RESUMEN

Rationale: The acoustic stimulation of microbubbles within microvessels can elicit a spectrum of therapeutically relevant bioeffects from permeabilization to perfusion shutdown. These bioeffects ultimately arise from complex interactions between microbubbles and microvascular walls, though such interactions are poorly understood particularly at high pressure, due to a paucity of direct in vivo observations. The continued development of focused ultrasound methods hinges in large part on establishing links between microbubble-microvessel interactions, cavitation signals, and bioeffects. Methods: Here, a system was developed to enable simultaneous high-speed intravital imaging and cavitation monitoring of microbubbles in vivo in a chorioallantoic membrane model. Exposures were conducted using the clinical agent DefinityTM under conditions previously associated with microvascular damage (1 MHz, 0.5-3.5 MPa, 5 ms pulse length). Results: Ultrasound-activated microbubbles could be observed and were found to induce localized wall deformations that were more pronounced in smaller microvessels and increased with pressure. A central finding was that microbubbles could extravasate from microvessels (from 34% of vessels at 1 MPa to 79% at 3 MPa) during insonation (94% within 0.5 ms) and that this occurred more frequently and in progressively larger microvessels (up to 180 µm) as pressure was increased. Following microbubble extravasation, transient or sustained red blood cell leakage ensued at the extravasation site in 96% of cases for pressures ≥1 MPa. Conclusions: The results here represent the first high-speed in vivo investigation of high-pressure focused ultrasound-induced microbubble-microvessel interactions. This data provides direct evidence that the process of activated microbubble extravasation can occur in vivo and that it is linked to producing microvessel wall perforations of sufficient size to permit red blood cell leakage. The association of red blood cell leakage with microbubble extravasation provides mechanistic insight into the process of microvessel rupture, which has been widely observed in histology.


Asunto(s)
Membrana Corioalantoides , Microburbujas , Animales , Microscopía , Ultrasonografía/métodos , Microscopía Intravital
3.
Ultrasonics ; 138: 107223, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38553135

RESUMEN

Thrombotic occlusions of large blood vessels are increasingly treated with catheter based mechanical approaches, one of the most prominent being to employ aspiration to extract clots through a hollow catheter lumen. A central technical challenge for aspiration catheters is to achieve sufficient suction force to overcome the resistance of clot material entering into the distal tip. In this study, we examine the feasibility of inducing cavitation within hollow cylindrical transducers with a view to ultimately using them to degrade the mechanical integrity of thrombus within the tip of an aspiration catheter. Hollow cylindrical radially polarized PZT transducers with 3.3/2.5 mm outer/inner diameters were assessed. Finite element simulations and hydrophone experiments were used to investigate the pressure field distribution as a function of element length and resonant mode (thickness, length). Operating in thickness mode (∼5 MHz) was found to be associated with the highest internal pressures, estimated to exceed 23 MPa. Cavitation was demonstrated to be achievable within the transducer under degassed water (10 %) conditions using hydrophone detection and high-frequency ultrasound imaging (40 MHz). Cavitation clouds occupied a substantial portion of the transducer lumen, in a manner that was dependent on the pulsing scheme employed (10 and 100 µs pulse lengths; 1.1, 11, and 110 ms pulse intervals). Collectively the results support the feasibility of achieving cavitation within a transducer compatible with mounting in the tip of an aspiration format catheter.


Asunto(s)
Ultrasonido Enfocado de Alta Intensidad de Ablación , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Transductores , Succión , Catéteres , Agua
4.
Adv Sci (Weinh) ; 11(4): e2304453, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38032129

RESUMEN

Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.


Asunto(s)
Microburbujas , Neoplasias , Ratones , Animales , Cobre , Ultrasonografía , Lípidos/química
5.
ACS Nano ; 18(1): 410-427, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38147452

RESUMEN

Focused ultrasound-stimulated microbubbles can induce blood flow shutdown and ischemic necrosis at higher pressures in an approach termed antivascular ultrasound. Combined with conventional therapies of chemotherapy, immunotherapy, and radiation therapy, this approach has demonstrated tumor growth inhibition and profound synergistic antitumor effects. However, the lower cavitation threshold of microbubbles can potentially yield off-target damage that the polydispersity of clinical agent may further exacerbate. Here we investigate the use of a monodisperse nanodroplet formulation for achieving antivascular effects in tumors. We first develop stable low boiling point monodisperse lipid nanodroplets and examine them as an alternative agent to mediate antivascular ultrasound. With synchronous intravital imaging and ultrasound monitoring of focused ultrasound-stimulated nanodroplets in tumor microvasculature, we show that nanodroplets can trigger blood flow shutdown and do so with a sharper pressure threshold and with fewer additional events than conventionally used microbubbles. We further leverage the smaller size and prolonged pharmacokinetic profile of nanodroplets to allow for potential passive accumulation in tumor tissue prior to antivascular ultrasound, which may be a means by which to promote selective tumor targeting. We find that vascular shutdown is accompanied by inertial cavitation and complex-order sub- and ultraharmonic acoustic signatures, presenting an opportunity for effective feedback control of antivascular ultrasound.


Asunto(s)
Neoplasias , Humanos , Ultrasonografía , Acústica , Microvasos/diagnóstico por imagen , Microscopía Intravital , Microburbujas
6.
Ultrasonics ; 132: 107006, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37116399

RESUMEN

Ultrafast ultrasound imaging enables the visualization of rapidly changing blood flow dynamics in the chambers of the heart. Singular value decomposition (SVD) filters outperform conventional high pass clutter rejection filters for ultrafast blood flow imaging of small and shallow fields of view (e.g., functional imaging of brain activity). However, implementing SVD filters can be challenging in cardiac imaging due to the complex spatially and temporally varying tissue characteristics. To address this challenge, we describe a method that involves excluding the proximal portion of the image (near the chest wall) and divides the reduced field of view into overlapped segments, within which tissue signals are expected to be spatially and temporally coherent. SVD filtering with automatic selection of cut-off singular vector orders to remove tissue and noise signals is implemented for each segment. Auto-thresholding is based on the coherence of spatial singular vectors, delineating tissue, blood, and noise subspaces within a spatial similarity matrix calculated for each segment. Filtered blood flow signals from the segments are reconstructed and then combined and Doppler processing is used to form a set of blood flow images. Preliminary experimental results suggest that the spatially segmented approach improves the separation of the tissue and blood subsets in the spatial similarity matrix so that automatic thresholding is significantly improved, and tissue clutter can then be rejected more effectively in cardiac ultrafast imaging, compared to using the full field of view. In the case studied, spatially segmented SVD improved the rate of correct automatic selection of thresholds from 78% to 98.7% for the investigated cases and improved the post-filter power of blood signals by an average of more than 10 dB during a cardiac cycle.


Asunto(s)
Procesamiento de Señales Asistido por Computador , Ultrasonografía Doppler , Velocidad del Flujo Sanguíneo/fisiología , Ultrasonografía Doppler/métodos , Ultrasonografía/métodos , Corazón/diagnóstico por imagen , Fantasmas de Imagen , Procesamiento de Imagen Asistido por Computador/métodos
7.
Theranostics ; 13(1): 250-266, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36593952

RESUMEN

Rationale: Focused ultrasound-stimulated microbubbles have been shown to be capable of inducing blood flow shutdown and necrosis in a range of tissue types in an approach termed antivascular ultrasound or nonthermal ablation. In oncology, this approach has demonstrated tumor growth inhibition, and profound synergistic antitumor effects when combined with traditional platforms of chemo-, radiation- and immune-therapies. However, the exposure schemes employed have been broad and underlying mechanisms remain unclear with fundamental questions about exposures, vessel types and sizes involved, and the nature of bubble behaviors and their acoustic emissions resulting in vascular damage - impeding the establishment of standard protocols. Methods: Here, ultrasound transmitters and receivers are integrated into a murine dorsal window chamber tumor model for intravital microscopy studies capable of real-time visual and acoustic monitoring during antivascular ultrasound. Vessel type (normal and tumor-affected), caliber, and viability are assessed under higher pressure conditions (1, 2, and 3 MPa), and cavitation signatures are linked to the biological effects. Results: Vascular events occurred preferentially in tumor-affected vessels with greater incidence in smaller vessels and with more severity as a function of increasing pressure. Vascular blood flow shutdown was found to be due to a combination of focal disruption events and network-related flow changes. Acoustic emissions displayed elevated broadband noise and distinct sub- and ultra-harmonics and their associated third-order peaks with increasing pressure. Conclusions: The observed vascular events taken collectively with identified cavitation signatures provide an improved mechanistic understanding of antivascular ultrasound at the microscale, with implications for establishing a specific treatment protocol and control platform.


Asunto(s)
Neoplasias , Animales , Ratones , Humanos , Ultrasonografía , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Acústica , Microvasos/diagnóstico por imagen , Microscopía Intravital , Microburbujas
8.
Ultrason Sonochem ; 93: 106291, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36640460

RESUMEN

Therapeutic focused ultrasound in combination with encapsulated microbubbles is being widely investigated for its ability to elicit bioeffects in the microvasculature, such as transient permeabilization for drug delivery or at higher pressures to achieve 'antivascular' effects. While it is well established that the behaviors of microbubbles are altered when they are situated within sufficiently small vessels, there is a paucity of data examining how the bubble population dynamics and emissions change as a function of channel (vessel) diameter over a size range relevant to therapeutic ultrasound, particularly at pressures relevant to antivascular ultrasound. Here we use acoustic emissions detection and high-speed microscopy (10 kframes/s) to examine the behavior of a polydisperse clinically employed agent (Definity®) in wall-less channels as their diameters are scaled from 1200 to 15 µm. Pressures are varied from 0.1 to 3 MPa using either a 5 ms pulse or a sequence of 0.1 ms pulses spaced at 1 ms, both of which have been previously employed in an in vivo context. With increasing pressure, the 1200 µm channel - on the order of small arteries and veins - exhibited inertial cavitation, 1/2 subharmonics and 3/2 ultraharmonics, consistent with numerous previous reports. The 200 and 100 µm channels - in the size range of larger microvessels less affected by therapeutic focused ultrasound - exhibited a distinctly different behavior, having muted development of 1/2 subharmonics and 3/2 ultraharmonics and reduced persistence. These were associated with radiation forces displacing bubbles to the distal wall and inducing clusters that then rapidly dissipated along with emissions. As the diameter transitioned to 50 and then 15 µm - a size regime that is most relevant to therapeutic focused ultrasound - there was a higher threshold for the onset of inertial cavitation as well as subharmonics and ultraharmonics, which importantly had more complex orders that are not normally reported. Clusters also occurred in these channels (e.g. at 3 MPa, the mean lateral and axial sizes were 23 and 72 µm in the 15 µm channel; 50 and 90 µm in the 50 µm channel), however in this case they occupied the entire lumens and displaced the wall boundaries. Damage to the 15 µm channel was observed for both pulse types, but at a lower pressure for the long pulse. Experiments conducted with a 'nanobubble' (<0.45 µm) subpopulation of Definity followed broadly similar features to 'native' Definity, albeit at a higher pressure threshold for inertial cavitation. These results provide new insights into the behavior of microbubbles in small vessels at higher pressures and have implications for therapeutic focused ultrasound cavitation monitoring and control.


Asunto(s)
Sonicación , Terapia por Ultrasonido , Sonicación/métodos , Microburbujas , Acústica , Terapia por Ultrasonido/métodos
9.
Drug Deliv ; 29(1): 1176-1183, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35393905

RESUMEN

BACKGROUND: Delivery of viral vectors as gene therapies to treat neurodegenerative diseases has been hampered by the inability to penetrate the blood brain barrier (BBB) and invasive or non-targeted delivery options prone to inducing immune responses. MR guided focused ultrasound (MR-g-FUS) and microbubbles have demonstrated safe, temporary, targeted BBB permeabilization clinically. METHODS: We developed clinically scalable, microbubble drug conjugates (MDCs) for the viral gene therapy, AAV.SIRT3-myc [adeno-associated virus expressing myc-tagged SIRT3], which has previously been shown to have disease modifying effects in animal models of Parkinson's disease (PD). The lipid shells of the perfluorocarbon gas MDCs were covalently conjugated to antibodies with binding specificity to AAVs. Following systemic (iv) delivery of AAV.SIRT3-myc MDCs, MR-g-FUS was used to deliver SIRT3-myc to brain regions affected in PD. SIRT3-myc expression was determined post mortem, using immunohistochemistry. RESULTS: An in vitro, SH-SY5Y cell culture model was used to show that the localized destruction of MDCs using ultrasound exposures within biological safety limits dissociated AAV2-GFP (green fluorescent protein) from the MDCs in the targeted area while maintaining their transduction capacity. In rats, MR-g-FUS resulted in BBB permeabilization in the striatum and substantia nigra (SNc). SIRT3-myc was expressed in the striatum, but not the SNc. CONCLUSION: These studies demonstrate that MDCs combined with MR-g-FUS are an effective method for delivery of viral vector gene therapies, such as AAV.SIRT3, to brain regions affected in PD. This technology may prove useful as a disease-modifying strategy in PD and other neurodegenerative disorders.


Asunto(s)
Sistemas de Liberación de Medicamentos , Enfermedad de Parkinson , Sirtuina 3 , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Dependovirus/genética , Proteínas Fluorescentes Verdes/genética , Imagen por Resonancia Magnética/métodos , Microburbujas , Enfermedad de Parkinson/terapia , Ratas , Sirtuina 3/metabolismo , Sirtuina 3/farmacología
10.
Artículo en Inglés | MEDLINE | ID: mdl-33513102

RESUMEN

There has been growing interest in nanobubbles (NBs) for vascular and extravascular ultrasound contrast imaging and therapeutic applications. Studies to date have generally utilized low frequencies (<12 MHz), high concentrations (>109 mL-1), and uncalibrated B-mode or contrast-mode on commercial systems without reporting investigations on NB signatures upon which the imaging protocols should be based. We recently demonstrated that low concentrations (106 mL-1) of porphyrin-lipid-encapsulated NBs scatter nonlinearly at low (2.5, 8 MHz) and high (12.5, 25, 30 MHz) frequencies in a pressure threshold-dependent manner that is advantageous for amplitude modulation (AM) imaging. Here, we implement pressure-calibrated AM at high frequency on a commercial preclinical array system to enhance sensitivity to nonlinear scattering of three phospholipid-based NB formulations. With this approach, improvements in contrast to tissue ratio relative to B-mode between 12.4 and 22.8 dB are demonstrated in a tissue-mimicking phantom, and between 6.7 and 14.8 dB in vivo.


Asunto(s)
Medios de Contraste , Diagnóstico por Imagen , Fantasmas de Imagen , Ultrasonografía
11.
Ultrasonics ; 110: 106245, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32932144

RESUMEN

Emerging contrast imaging studies have highlighted the potential of nanobubbles for both intravascular and extravascular applications. Reports to date on nanobubbles have generally utilized low frequencies (<12 MHz), high concentrations (>109 mL-1), and B-mode or contrast-mode on preclinical and clinical systems. However, none of these studies directly examined nanobubble acoustic signatures systematically to implement nonlinear imaging schemes in a methodical manner based on nanobubble behaviour. Here, nanobubble nonlinear behaviour is investigated at high frequencies (12.5, 25, 30 MHz) and low concentration (106 mL-1) in a channel phantom, with different pulse types in single- and multi-pulse sequences to examine behaviour under conditions relevant to high frequency imaging. Porphyrin nanobubbles are demonstrated to initiate nonlinear scattering at high frequencies in a pressure-threshold dependent manner, as previously observed at low frequencies. This threshold behaviour was then utilized to demonstrate enhanced nanobubble imaging with pulse inversion, amplitude modulation, and a combination of the two, progressing towards the improved sensitivity and expanded utility of these ultrasound contrast agents.

12.
Theranostics ; 10(25): 11690-11706, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052241

RESUMEN

Background: There has been growing interest in nanobubbles for their potential to extend bubble-mediated ultrasound approaches beyond that of their larger microbubble counterparts. In particular, the smaller scale of nanobubbles may enable them to access the tumor extravascular compartment for imaging and therapy in closer proximity to cancer cells. Compelling preliminary demonstrations of the imaging and therapeutic abilities of nanobubbles have thus emerged, with emphasis on their ability to extravasate. However, studies to date rely on indirect histologic evidence that cannot confirm whether the structures remain intact beyond the vasculature - leaving their extravascular potential largely untapped. Methods: Nanobubble acoustic scattering was assessed using a recently reported ultra-stable formulation at low concentration (106 mL-1) and frequency (1 MHz), over a range of pressures (100-1500 kPa) in a channel phantom. The pressure-dependent response was utilized as a basis for in vivo experiments where ultrasound transmitters and receivers were integrated into a window chamber for simultaneous intravital multiphoton microscopy and acoustic monitoring in tumor-affected microcirculation. Microscopy and acoustic data were utilized to assess passive and active delivery of nanobubbles and determine whether they remained intact beyond the vasculature. Results: Nanobubbles exhibit pressure-dependent nonlinear acoustic scattering. Nanobubbles are also found to have prolonged acoustic vascular pharmacokinetics, and passively extravasate intact into tumors. Ultrasound stimulation of nanobubbles is shown to actively enhance the delivery of both intact nanobubbles and shell material, increasing their spatial bioavailability deeper into the extravascular space. A range of acute vascular effects were also observed. Conclusion: This study presents the first direct evidence that nanobubbles passively and actively extravasate intact in tumor tissue, and is the first to directly capture acute vascular events from ultrasound-stimulation of nanobubbles. The insights gained here demonstrate an important step towards unlocking the potential of nanobubbles and extending ultrasound-based applications.


Asunto(s)
Medios de Contraste/administración & dosificación , Microburbujas , Microscopía Acústica/métodos , Nanopartículas/administración & dosificación , Neoplasias/diagnóstico por imagen , Animales , Línea Celular Tumoral , Humanos , Ratones , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neoplasias/irrigación sanguínea , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Sci Adv ; 6(36)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32917589

RESUMEN

Thermosensitive liposomes represent an important paradigm in oncology, where hyperthermia-mediated release coupled with thermal bioeffects enhance the effectiveness of chemotherapy. Their widespread clinical adoption hinges upon performing controlled targeted hyperthermia, and a leading candidate to achieve this is temperature-based magnetic resonance imaging (MRI)-guided focused ultrasound (MRgFUS). However, the current approach to hyperthermia involves exposures lasting tens of minutes to hours, which is not possible to achieve in many circumstances because of blood vessel cooling and respiratory motion. Here, we investigate a novel approach to overcome these limitations: to use fractionated ultrashort (~30 s) thermal exposures (~41° to 45°C) to release doxorubicin from a thermosensitive liposome. This is first demonstrated in a dorsal chamber tumor model using two-photon microscopy. Thermal exposures were then conducted with a rabbit tumor model using a custom MRgFUS system incorporating temperature feedback control. Drug release was confirmed, and longitudinal experiments demonstrated profoundly enhanced tumor growth inhibition and survival.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neoplasias , Animales , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Liposomas , Imagen por Resonancia Magnética , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Conejos
14.
Ultrasound Med Biol ; 46(10): 2861-2870, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32732167

RESUMEN

Submicron phase-change droplets are an emerging class of ultrasound contrast agent. Compared with microbubbles, their relatively small size and increased stability offer the potential to passively extravasate and accumulate in solid tumors through the enhanced permeability and retention effect. Under exposure to sufficiently powerful ultrasound, these droplets can convert into in situ gas microbubbles and thus be used as an extravascular-specific contrast agent. However, in vivo imaging methods to detect extravasated droplets have yet to be established. Here, we develop an ultrasound imaging pulse sequence within diagnostic safety limits to selectively detect droplet extravasation in tumors. Tumor-bearing mice were injected with submicron perfluorobutane droplets and interrogated with our imaging-vaporization-imaging sequence. By use of a pulse subtraction method, median droplet extravasation signal relative to the total signal within the tumor was estimated to be Etumor=37±5% compared with the kidney Ekidney=-2±8% (p < 0.001). This work contributes toward the advancement of volatile phase-shift droplets as a next-generation ultrasound agent for imaging and therapy.


Asunto(s)
Medios de Contraste , Fluorocarburos , Microburbujas , Neoplasias/diagnóstico por imagen , Volatilización , Animales , Ratones , Ultrasonografía/métodos
15.
Ultrasound Med Biol ; 46(8): 2104-2112, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32473846

RESUMEN

Although intravascular ultrasound (IVUS) is an important tool in guiding complex coronary interventions, the resolution of existing commercial IVUS devices is considerably poorer than that of optical coherence tomography. Dual-frequency IVUS (DF IVUS), incorporating a second, higher frequency transducer, has been proposed as a possible method of overcoming this limitation. Although preliminary studies have shown that DF IVUS can produce complementary images, including large-scale morphology and high detail of superficial features, it has not yet been determined that this approach would be feasible in a more clinically relevant environment. The purpose of this study was to demonstrate the first in vivo use of a 30/80 MHz DF IVUS catheter in visualizing coronary vessels in a porcine model. In addition, two commercially available stents were studied in vitro and in vivo. Clear subjective improvement of visualization of superficial structures is demonstrated, and sufficient dynamic range is achieved to image through both the catheter sheath and blood in vivo.


Asunto(s)
Prótesis Vascular , Vasos Coronarios/diagnóstico por imagen , Stents , Ultrasonografía Intervencional/métodos , Animales , Implantación de Prótesis Vascular/métodos , Femenino , Porcinos
16.
Nano Lett ; 20(6): 4512-4519, 2020 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-32374617

RESUMEN

Ultrasound-activated nanobubbles are being widely investigated as contrast agents and therapeutic vehicles. Nanobubbles hold potential for accessing the tumor extravascular compartment, though this relies on clinically debated passive accumulation for which evidence to date is indirect. We recently reported ultrasound-triggered conversion of high payload porphyrin-encapsulated microbubbles to nanobubbles, with actively enhanced permeability for local delivery. This platform holds implications for optical/ultrasound-based imaging and therapeutics. While promising, it remains to be established how nanobubbles are generated and whether they extravasate intact. Here, insights into the conversion process are reported, complemented by novel simultaneous intravital and acoustic monitoring in tumor-affected functional circulation. The first direct acoustic evidence of extravascular intact nanobubbles are presented. These insights collectively advance this delivery platform and multimodal micro- and nanobubbles, extending their utility for imaging and therapeutics within and beyond the vasculature.


Asunto(s)
Medios de Contraste , Microburbujas , Neoplasias , Ultrasonografía , Acústica , Humanos , Nanotecnología
17.
Phys Med Biol ; 64(13): 135003, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31082815

RESUMEN

Ultrasound stimulated microbubbles have been shown to be capable of breaking up blood clots through micro-scale interactions occurring near the clot surface. However, only a small fraction of bubbles circulating in the bloodstream will be in close proximity to such boundaries, where they must be to elicit therapeutic effects. Here, the accumulation and subsequent behavior of microbubbles displaced from an overlying flow channel to a boundary under radiation forces were examined. Experimental data were acquired using a novel high speed microscopy configuration and simulations were conducted to provide insight into the accumulation process. There was broad agreement between experiments and simulations, both indicating that the size distribution and number of bubbles arriving at the boundary depended on channel flow rate, applied pressure, and bubble concentration. For example, higher flow rates and lower pressures favored the accumulation of larger bubbles relative to the native agent distribution. Moreover, bubble dynamics were dependent on the surface type, exhibiting rapid translation along agarose gel surfaces whereas on fibrin surfaces, they accumulated in localized regions inducing repetitive strain cycles. The results indicate that the process of bringing bubbles from within a vessel to a boundary is complex and should be an important consideration in the development of therapeutic applications such as sonothrombolysis.


Asunto(s)
Acústica , Medios de Contraste , Microburbujas , Fantasmas de Imagen , Ultrasonografía/métodos , Coagulación Sanguínea , Fibrina/química , Humanos , Propiedades de Superficie
18.
Ultrasound Med Biol ; 45(2): 500-512, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30447880

RESUMEN

Checkpoint inhibitor (CI) immunotherapy is playing an increasingly prominent role in the treatment of cancer but is effective and durable in only a subset of patients. There are concerted efforts to improve CI therapy through the use of multiple CIs or use of CIs in combination with other anti-cancer agents. Here we investigate the use of "anti-vascular" ultrasound-stimulated microbubble (USMB) treatments in combination with anti-PD-1 CI therapy. The colorectal cancer cell line CT26 was used to conduct longitudinal growth studies along with acute experiments to assess ultrasound-induced anti-tumor immune responses using flow cytometry and enzyme-linked immunospot (ELISPOT) analysis. Longitudinal experiments indicated that USMB + anti-PD-1 treatments significantly enhanced tumor growth inhibition and animal survival relative to monotherapies. Flow cytometry and ELISPOT data did not clearly support a T cell-dependent mechanism for the enhancement. Therefore, the results indicate the ability of anti-vascular USMBs to increase the anti-tumor effects of CI therapy; the specific mechanisms of enhancement remain to be established.


Asunto(s)
Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Inmunoterapia/métodos , Microburbujas , Ultrasonido/métodos , Animales , Línea Celular Tumoral , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C
19.
J Acoust Soc Am ; 144(5): 2859, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30522286

RESUMEN

It is established that radiation forces can be used to transport ultrasound contrast agents, particularly for molecular imaging applications. However, the ability to model and control this process in the context of therapeutic ultrasound is limited by a paucity of data on the translational dynamics of encapsulated microbubbles under the influence of longer pulses. In this work, the translation of individual microbubbles, isolated with optical tweezers, was experimentally investigated over a range of diameters (1.8-8.8 µm, n = 187) and pressures (25, 50, 100, 150, and 200 kPa) with millisecond pulses. Data were compared with theoretical predictions of the translational dynamics, assessing the role of shell and history force effects. A pronounced feature of the displacement curves was an effective threshold size, below which there was only minimal translation. At higher pressures (≥150 kPa) a noticeable structure emerged where multiple local maxima occurred as a function of bubble size. The ability to accurately capture these salient features depended on the encapsulation model employed. In low Reynolds number conditions (i.e., low pressures, or high pressures, off-resonance) the inclusion of history force more accurately fit the data. After pulse cessation, bubbles exhibited substantial displacements consistent with the influence of history effects.

20.
Phys Med Biol ; 63(21): 215001, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30272572

RESUMEN

Nanobubbles hold potential for expanding utility of ultrasound contrast-based applications to extravascular targets, but their acoustic response and the effects of the surrounding environment remain relatively unexplored. Here we investigate the dynamics of porphyrin-encapsulated nanobubbles (diameter <0.4 µm; 106 ml-1) at clinically relevant frequencies (2.5 MHz and 8 MHz) as a function of pressure (0.1-1.0 MPa) in vessel- and tissue-mimicking phantoms to gain an understanding of nanobubble behaviour in intra- and extravascular compartments. The results provide the first direct observation that nanobubbles can initiate nonlinear scattering, and that they do so in a pressure-dependent manner. It is further demonstrated that while nanobubbles in confining media require higher pressures for nonlinearities and demonstrate reduced scattering, they can exhibit sustained and non-destructive cavitation. Bubble models are then used to gain mechanistic insights into experimentally observed nanobubble dynamics and confirm sensitivity to nonlinear shell rheology, particularly to radially-dependent surface tension and the characteristic time constant for shear-thinning.


Asunto(s)
Vesículas Extracelulares/química , Microvasos/química , Nanoestructuras/química , Fantasmas de Imagen , Porfirinas/química , Medios de Contraste , Humanos , Dispersión de Radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA