Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Ultrasound Med Biol ; 50(8): 1232-1239, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38760280

RESUMEN

BACKGROUND: Acoustically activated perfluoropropane droplets (PD) formulated from lipid encapsulated microbubble preparations produce a delayed myocardial contrast enhancement that preferentially highlights the infarct zones (IZ). Since activation of PDs may be temperature sensitive, it is unclear what effect body temperature (BT) has on acoustic activation (AA). OBJECTIVE: We sought to determine whether the microvascular retention and degree of myocardial contrast intensity (MCI) would be affected by BT at the time of intravenous injection. METHODS: We administered intravenous (IV) PD in nine rats following 60 min of ischemia followed by reperfusion. Injections in these rats were given at temperatures above and below 36.5°C, with high MI activation in both groups at 3 or 6 min following IV injection (IVI). In six additional rats (three in each group), IV PDs were given only at one temperature (<36.5°C or ≥36.5°C), permitting a total of 12 comparisons of different BT. Differences in background subtracted MCI at 3-6 min post-injection were compared in the infarct zone (IZ) and remote zone (RZ). Post-mortem lung hematoxylin and eosin (H&E) staining was performed to assess the effect potential thermal activation on lung tissue. RESULTS: Selective MCI within the IZ was observed in 8 of 12 rats who received IVI of PDs at <36.5°C, but none of the 12 rats who had IVI at the higher temperature (p < 0.0001). Absolute MCI following droplet activation was significantly higher in both the IZ and RZ when given at the lower BT. H&E indicated significant red blood extravasation in 5/7 rats who had had IV injections at higher BT, and 0/7 rats who had IV PDs at <36.5°C. CONCLUSIONS: Selective IZ enhancement with AA of intravenous PDs is possible, but temperature sensitive. Thermal activation appears to occur when PDs are given at higher temperatures, preventing AA, and increasing unwanted bioeffects.


Asunto(s)
Medios de Contraste , Fluorocarburos , Infarto del Miocardio , Ratas Sprague-Dawley , Animales , Ratas , Infarto del Miocardio/fisiopatología , Masculino , Microburbujas , Temperatura Corporal , Acústica
2.
J Ultrasound Med ; 43(6): 1063-1080, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38440926

RESUMEN

BACKGROUND: Acoustically activatable perfluoropropane droplets (PD) can be formulated from commercially available microbubble preparations. Diagnostic transthoracic ultrasound frequencies have resulted in acoustic activation (AA) predominately within myocardial infarct zones (IZ). OBJECTIVE: We hypothesized that the AA area following acute coronary ischemia/reperfusion (I/R) would selectively enhance the developing scar zone, and target bioeffects specifically to this region. METHODS: We administered intravenous PD in 36 rats and 20 pigs at various stages of myocardial scar formation (30 minutes, 1 day, and 7 days post I/R) to determine what effect infarct age had on the AA within the IZ. This was correlated with histology, myeloperoxidase activity, and tissue nitrite activity. RESULTS: The degree of AA within the IZ in rats was not associated with collagen content, neutrophil infiltration, or infarct age. AA within 24 hours of I/R was associated with increased nitric oxide utilization selectively within the IZ (P < .05 compared with remote zone). The spatial extent of AA in pigs correlated with infarct size only when performed before sacrifice at 7 days (r = .74, P < .01). CONCLUSIONS: Acoustic activation of intravenous PD enhances the developing scar zone following I/R, and results in selective tissue nitric oxide utilization.


Asunto(s)
Fluorocarburos , Infarto del Miocardio , Animales , Fluorocarburos/farmacocinética , Porcinos , Ratas , Infarto del Miocardio/diagnóstico por imagen , Masculino , Medios de Contraste/farmacocinética , Nanopartículas , Ratas Sprague-Dawley , Miocardio/metabolismo , Modelos Animales de Enfermedad , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Microburbujas , Femenino , Ultrasonografía/métodos
3.
Ultrasound Med Biol ; 49(5): 1058-1069, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36797095

RESUMEN

OBJECTIVE: Phase-change contrast agents (PCCAs) are perfluorocarbon nanodroplets (NDs) that have been widely studied for ultrasound imaging in vitro, pre-clinical studies, and most recently incorporated a variant of PCCAs, namely a microbubble-conjugated microdroplet emulsion, into the first clinical studies. Their properties also make them attractive candidates for a variety of diagnostic and therapeutic applications including drug-delivery, diagnosis and treatment of cancerous and inflammatory diseases, as well as tumor-growth tracking. However, control over the thermal and acoustic stability of PCCAs both in vivo and in vitro has remained a challenge for expanding the potential utility of these agents in novel clinical applications. As such, our objective was to determine the stabilizing effects of layer-by-layer assemblies and its effect on both thermal and acoustic stability. METHODS: We utilized layer-by-layer (LBL) assemblies to coat the outer PCCA membrane and characterized layering by measuring zeta potential and particle size. Stability studies were conducted by; 1) incubating the LBL-PCCAs at atmospheric pressure at 37∘C and 45∘C followed by; 2) ultrasound-mediated activation at 7.24 MHz and peak-negative pressures ranging from 0.71 - 5.48 MPa to ascertain nanodroplet activation and resultant microbubble persistence. The thermal and acoustic properties of decafluorobutane gas-condensed nanodroplets (DFB-NDs) layered with 6 and 10 layers of charge-alternating biopolymers, (LBL6NDs and LBL10NDs) respectively, were studied and compared to non-layered DFB-NDs. Half-life determinations were conducted at both 37∘C and 45∘C with acoustic droplet vaporization (ADV) measurements occurring at 23∘C. DISCUSSION: Successful application of up to 10 layers of alternating positive and negatively charged biopolymers onto the surface membrane of DFB-NDs was demonstrated. Two major claims were substantiated in this study; namely, (1) biopolymeric layering of DFB-NDs imparts a thermal stability up to an extent; and, (2) both LBL6NDs and LBL10NDs did not appear to alter particle acoustic vaporization thresholds, suggesting that the thermal stability of the particle may not necessarily be coupled with particle acoustic vaporization thresholds. CONCLUSION: Results demonstrate that the layered PCCAs had higher thermal stability, where the half-lifes of the LBLxNDs are significantly increased after incubation at 37∘C and 45∘C. Furthermore, the acoustic vaporization profiles the DFB-NDs, LBL6NDs, and LBL10NDs show that there is no statistically significant difference between the acoustic vaporization energy required to initiate acoustic droplet vaporization.


Asunto(s)
Fluorocarburos , Neoplasias , Humanos , Medios de Contraste , Nanopartículas Capa por Capa , Acústica , Volatilización , Ultrasonografía/métodos , Microburbujas
4.
Ultrasound Med Biol ; 44(12): 2728-2738, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30228045

RESUMEN

Breast cancer remains a leading cause of death for women throughout the world. Recent advances in medical imaging technologies and tumor targeting agents signify vast potential for progress toward improved management of this global problem. Phase-change contrast agents (PCCAs) are dynamic imaging agents with practical applications in both the research and clinical settings. PCCAs possess characteristics that allow for cellular uptake where they can be converted from liquid-phase PCCAs to gaseous microbubbles via ultrasound energy. Previously, we reported successful internalization of folate-targeted PCCAs in MDA-MB-231 breast cancer cells followed by ultrasound-mediated activation to produce internalized microbubbles. This study examines the binding, internalization and activation of folate-receptor targeted PCCAs in MDA-MB-231 breast cancer cells as a function of gaseous core compositions, incubation time and ultrasound exposure period. In vitro results indicate that internalization and ultrasound-mediated activation of PCCAs were significantly greater using a 50:50 mixture of decafluorobutane:dodecafluoropentane compared with other core compositions: 50:50 octafluoropropane:decafluorobutane (p < 0.0001), decafluorobutane (p < 0.04) and dodecafluoropentane (p < 0.0001). Furthermore, it was found that PCCAs composed of perfluorocarbons with higher boiling points responded with greater activation efficiency when exposed to 12 s of ultrasound exposure as opposed to 4 s of ultrasound exposure. When evaluating different incubation times, it was found that incubating the PCCAs with breast cancer cells for 60 min did not produce significantly greater internalization and activation compared with incubation for 10 min; this was concluded after comparing the number of microbubbles present per cell before ultrasound versus post-ultrasound, and finding a ratio of intracellular microbubbles post-ultrasound/pre-ultrasound, 3.46 versus 3.14, respectively. The data collected in this study helps illustrate further optimization of folate-receptor targeted PCCAs for breast cancer targeting and imaging.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Aumento de la Imagen/métodos , Microburbujas , Ultrasonografía/métodos , Células Cultivadas , Medios de Contraste , Femenino , Humanos
5.
J Vasc Surg ; 68(6S): 105S-113S, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29452833

RESUMEN

BACKGROUND: Molecular imaging of carotid plaque vulnerability to atheroembolic events is likely to lead to improvements in selection of patients for carotid endarterectomy (CEA). The aims of this study were to assess the relative value of endothelial inflammatory markers for this application and to develop molecular ultrasound contrast agents for their imaging. METHODS: Human CEA specimens were obtained prospectively from asymptomatic (30) and symptomatic (30) patients. Plaques were assessed by semiquantitative immunohistochemistry for vascular cell adhesion molecule 1 (VCAM-1), lectin-like oxidized low-density lipoprotein receptor 1, P-selectin, and von Willebrand factor. Established small peptide ligands to each of these targets were then synthesized and covalently conjugated to the surface of lipid-shelled microbubble ultrasound contrast agents, which were then evaluated in a flow chamber for binding kinetics to activated human aortic endothelial cells under variable shear conditions. RESULTS: Expression of VCAM-1 on the endothelium of CEA specimens from symptomatic patients was 2.4-fold greater than that from asymptomatic patients (P < .01). Expression was not significantly different between groups for P-selectin (P = .43), von Willebrand factor (P = .59), or lectin-like oxidized low-density lipoprotein receptor 1 (P = .99). Although most plaques from asymptomatic patients displayed low VCAM-1 expression, approximately one in five expressed high VCAM-1 similar to plaques from symptomatic patients. In vitro flow chamber experiments demonstrated that VCAM-1-targeted microbubbles bind cells that express VCAM-1, even under high-shear conditions that approximate those found in human carotid arteries, whereas binding efficiency was lower for the other agents. CONCLUSIONS: VCAM-1 displays significantly higher expression on high-risk (symptomatic) vs low-risk (asymptomatic) carotid plaques. Ultrasound contrast agents bearing ligands for VCAM-1 can sustain high-shear attachment and may be useful for identifying patients in whom more aggressive treatment is warranted.


Asunto(s)
Arterias Carótidas/diagnóstico por imagen , Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/metabolismo , Imagen Molecular/métodos , Placa Aterosclerótica , Ultrasonografía , Molécula 1 de Adhesión Celular Vascular/análisis , Anciano , Anciano de 80 o más Años , Enfermedades Asintomáticas , Biomarcadores/análisis , Arterias Carótidas/patología , Enfermedades de las Arterias Carótidas/complicaciones , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Medios de Contraste/administración & dosificación , Medios de Contraste/metabolismo , Células Endoteliales/metabolismo , Estudios de Factibilidad , Femenino , Humanos , Inmunohistoquímica , Ataque Isquémico Transitorio/etiología , Ligandos , Masculino , Microburbujas , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Pronóstico , Estudios Prospectivos , Factores de Riesgo , Rotura Espontánea , Accidente Cerebrovascular/etiología
6.
Photoacoustics ; 6: 26-36, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28507898

RESUMEN

We demonstrate a versatile phase-change sub-micron contrast agent providing three modes of contrast enhancement: 1) photoacoustic imaging contrast, 2) ultrasound contrast with optical activation, and 3) ultrasound contrast with acoustic activation. This agent, which we name 'Cy-droplet', has the following novel features. It comprises a highly volatile perfluorocarbon for easy versatile activation, and a near-infrared optically absorbing dye chosen to absorb light at a wavelength with good tissue penetration. It is manufactured via a 'microbubble condensation' method. The phase-transition of Cy-droplets can be optically triggered by pulsed-laser illumination, inducing photoacoustic signal and forming stable gas bubbles that are visible with echo-ultrasound in situ. Alternatively, Cy-droplets can be converted to microbubble contrast agents upon acoustic activation with clinical ultrasound. Potentially all modes offer extravascular contrast enhancement because of the sub-micron initial size. Such versatility of acoustic and optical 'triggerability' can potentially improve multi-modality imaging, molecularly targeted imaging and controlled drug release.

7.
Artículo en Inglés | MEDLINE | ID: mdl-27775902

RESUMEN

Continued advances in the field of ultrasound and ultrasound contrast agents have created new approaches to imaging and medical intervention. Phase-shift perfluorocarbon droplets, which can be vaporized by ultrasound energy to transition from the liquid to the vapor state, are one of the most highly researched alternatives to clinical ultrasound contrast agents (i.e., microbubbles). In this paper, part of a special issue on methods in biomedical ultrasonics, we survey current techniques to prepare ultrasound-activated nanoscale phase-shift perfluorocarbon droplets, including sonication, extrusion, homogenization, microfluidics, and microbubble condensation. We provide example protocols and discuss advantages and limitations of each approach. Finally, we discuss best practice in characterization of this class of contrast agents with respect to size distribution and ultrasound activation.


Asunto(s)
Medios de Contraste/química , Fluorocarburos/química , Microburbujas , Ultrasonografía/métodos , Tamaño de la Partícula
8.
J Control Release ; 243: 69-77, 2016 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-27686582

RESUMEN

Breast cancer is a diverse and complex disease that remains one of the leading causes of death among women. Novel, outside-of-the-box imaging and treatment methods are needed to supplement currently available technologies. In this study, we present evidence for the intracellular delivery and ultrasound-stimulated activation of folate receptor (FR)-targeted phase-change contrast agents (PCCAs) in MDA-MB-231 and MCF-7 breast cancer cells in vitro. PCCAs are lipid-coated, perfluorocarbon-filled particles formulated as nanoscale liquid droplets capable of vaporization into gaseous microbubbles for imaging or therapy. Cells were incubated with 1:1 decafluorobutane (DFB)/octafluoropropane (OFP) PCCAs for 1h, imaged via confocal microscopy, exposed to ultrasound (9MHz, MI=1.0 or 1.5), and imaged again after insonation. FR-targeted PCCAs were observed intracellularly in both cell lines, but uptake was significantly greater (p<0.001) in MDA-MB-231 cells (93.0% internalization at MI=1.0, 79.5% at MI=1.5) than MCF-7 cells (42.4% internalization at MI=1.0, 35.7% at MI=1.5). Folate incorporation increased the frequency of intracellular PCCA detection 45-fold for MDA-MB-231 cells and 7-fold for MCF-7 cells, relative to untargeted PCCAs. Intracellularly activated PCCAs ranged from 500nm to 6µm (IQR=800nm-1.5µm) with a mean diameter of 1.15±0.59 (SD) microns. The work presented herein demonstrates the feasibility of PCCA intracellular delivery and activation using breast cancer cells, illuminating a new platform toward intracellular imaging or therapeutic delivery with ultrasound.


Asunto(s)
Neoplasias de la Mama/metabolismo , Medios de Contraste/administración & dosificación , Sistemas de Liberación de Medicamentos , Receptores de Folato Anclados a GPI/metabolismo , Neoplasias de la Mama/diagnóstico por imagen , Línea Celular Tumoral , Femenino , Fluorocarburos/química , Humanos , Células MCF-7 , Microscopía Confocal , Tamaño de la Partícula , Distribución Tisular , Ultrasonido/métodos
9.
Biomed Opt Express ; 7(7): 2849-60, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27446711

RESUMEN

The use of receptor-targeted lipid microbubbles imaged by ultrasound is an innovative method of detecting and localizing disease. However, since ultrasound requires a medium between the transducer and the object being imaged, it is impractical to apply to an exposed surface in a surgical setting where sterile fields need be maintained and ultrasound gel may cause the bubbles to collapse. Multiphoton microscopy (MPM) is an emerging tool for accurate, label-free imaging of tissues and cells with high resolution and contrast. We have recently determined a novel application of MPM to be used for detecting targeted microbubble adherence to the upregulated plectin-receptor on pancreatic tumor cells. Specifically, the third-harmonic generation response can be used to detect bound microbubbles to various cell types presenting MPM as an alternative and useful imaging method. This is an interesting technique that can potentially be translated as a diagnostic tool for the early detection of cancer and inflammatory disorders.

10.
DNA Repair (Amst) ; 43: 98-106, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27130816

RESUMEN

Non-Homologous End-Joining (NHEJ) is the predominant pathway for the repair of DNA double strand breaks (DSBs) in human cells. The NHEJ pathway is frequently upregulated in several solid cancers as a compensatory mechanism for a separate DSB repair defect or for innate genomic instability, making this pathway a powerful target for synthetic lethality approaches. In addition, NHEJ reduces the efficacy of cancer treatment modalities which rely on the introduction of DSBs, like radiation therapy or genotoxic chemotherapy. Consequently, inhibition of the NHEJ pathway can modulate a radiation- or chemo-refractory disease presentation. The Ku70/80 heterodimer protein plays a pivotal role in the NHEJ process. It possesses a ring-shaped structure with high affinity for DSBs and serves as the first responder and central scaffold around which the rest of the repair complex is assembled. Because of this central position, the Ku70/80 dimer is a logical target for the disruption of the entire NHEJ pathway. Surprisingly, specific inhibitors of the Ku70/80 heterodimer are currently not available. We here describe an in silico, pocket-based drug discovery methodology utilizing the crystal structure of the Ku70/80 heterodimer. We identified a novel putative small molecule binding pocket and selected several potential inhibitors by computational screening. Subsequent biological screening resulted in the first identification of a compound with confirmed Ku-inhibitory activity in the low micro-molar range, capable of disrupting the binding of Ku70/80 to DNA substrates and impairing Ku-dependent activation of another NHEJ factor, the DNA-PKCS kinase. Importantly, this compound synergistically sensitized human cell lines to radiation treatment, indicating a clear potential to diminish DSB repair. The chemical scaffold we here describe can be utilized as a lead-generating platform for the design and development of a novel class of anti-cancer agents.


Asunto(s)
Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , ADN/genética , Autoantígeno Ku/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Cristalografía por Rayos X , ADN/metabolismo , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Rayos gamma , Expresión Génica , Células HeLa , Humanos , Autoantígeno Ku/química , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Simulación del Acoplamiento Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Dominios Proteicos , Multimerización de Proteína , Estructura Secundaria de Proteína , Pirimidinas/síntesis química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Bibliotecas de Moléculas Pequeñas/síntesis química
11.
Ultrasound Med Biol ; 41(5): 1422-31, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25656747

RESUMEN

Phase-change contrast agents in the form of nanoscale droplets can be activated into microbubbles by ultrasound, extending the contrast beyond the vasculature. This article describes simultaneous optical and acoustical measurements for quantifying the ultrasound activation of phase-change contrast agents over a range of concentrations. In experiments, decafluorobutane-based nanodroplets of different dilutions were sonicated with a high-pressure activation pulse and two low-pressure interrogation pulses immediately before and after the activation pulse. The differences between the pre- and post-interrogation signals were calculated to quantify the acoustic power scattered by the microbubbles activated over a range of droplet concentrations. Optical observation occurred simultaneously with the acoustic measurement, and the pre- and post-microscopy images were processed to generate an independent quantitative indicator of the activated microbubble concentration. Both optical and acoustic measurements revealed linear relationships to the droplet concentration at a low concentration range <10(8)/mL when measured at body temperature. Further increases in droplet concentration resulted in saturation of the acoustic interrogation signal. Compared with body temperature, room temperature was found to produce much fewer and larger bubbles after ultrasound droplet activation.


Asunto(s)
Medios de Contraste/química , Fluorocarburos/química , Fluorocarburos/efectos de la radiación , Ondas de Choque de Alta Energía , Nanopartículas/química , Nanopartículas/efectos de la radiación , Medios de Contraste/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Ensayo de Materiales , Tamaño de la Partícula , Transición de Fase , Dosis de Radiación
12.
Phys Med Biol ; 59(2): 379-401, 2014 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-24351961

RESUMEN

Phase-change contrast agents (PCCAs) provide a dynamic platform to approach problems in medical ultrasound (US). Upon US-mediated activation, the liquid core vaporizes and expands to produce a gas bubble ideal for US imaging and therapy. In this study, we demonstrate through high-speed video microscopy and US interrogation that PCCAs composed of highly volatile perfluorocarbons (PFCs) exhibit unique acoustic behavior that can be detected and differentiated from standard microbubble contrast agents. Experimental results show that when activated with short pulses PCCAs will over-expand and undergo unforced radial oscillation while settling to a final bubble diameter. The size-dependent oscillation phenomenon generates a unique acoustic signal that can be passively detected in both time and frequency domain using confocal piston transducers with an 'activate high' (8 MHz, 2 cycles), 'listen low' (1 MHz) scheme. Results show that the magnitude of the acoustic 'signature' increases as PFC boiling point decreases. By using a band-limited spectral processing technique, the droplet signals can be isolated from controls and used to build experimental relationships between concentration and vaporization pressure. The techniques shown here may be useful for physical studies as well as development of droplet-specific imaging techniques.


Asunto(s)
Acústica , Medios de Contraste/química , Fluorocarburos/química , Volatilización
13.
J Acoust Soc Am ; 134(2): 1473-82, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23927187

RESUMEN

Ultrasound contrast agents are known to enhance high intensity focused ultrasound (HIFU) ablation, but these perfluorocarbon microbubbles are limited to the vasculature, have a short half-life in vivo, and may result in unintended heating away from the target site. Herein, a nano-sized (100-300 nm), dual perfluorocarbon (decafluorobutane/dodecafluoropentane) droplet that is stable, is sufficiently small to extravasate, and is convertible to micron-sized bubbles upon acoustic activation was investigated. Microbubbles and nanodroplets were incorporated into tissue-mimicking acrylamide-albumin phantoms. Microbubbles or nanodroplets at 0.1 × 10(6) per cm(3) resulted in mean lesion volumes of 80.4 ± 33.1 mm(3) and 52.8 ± 14.2 mm(3) (mean ± s.e.), respectively, after 20 s of continuous 1 MHz HIFU at a peak negative pressure of 4 MPa, compared to a lesion volume of 1.0 ± 0.8 mm(3) in agent-free control phantoms. Magnetic resonance thermometry mapping during HIFU confirmed undesired surface heating in phantoms containing microbubbles, whereas heating occurred at the acoustic focus of phantoms containing the nanodroplets. Maximal change in temperature at the target site was enhanced by 16.9% and 37.0% by microbubbles and nanodroplets, respectively. This perfluorocarbon nanodroplet has the potential to reduce the time to ablate tumors by one-third during focused ultrasound surgery while also safely enhancing thermal deposition at the target site.


Asunto(s)
Medios de Contraste/química , Fluorocarburos/química , Ultrasonido Enfocado de Alta Intensidad de Ablación/métodos , Calor , Acrilamidas/química , Albúminas/química , Ultrasonido Enfocado de Alta Intensidad de Ablación/instrumentación , Imagen por Resonancia Magnética , Microburbujas , Nanopartículas , Fantasmas de Imagen , Presión , Sonicación , Sonido , Termografía , Factores de Tiempo , Transductores , Volatilización
14.
Phys Med Biol ; 58(13): 4513-34, 2013 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-23760161

RESUMEN

Ultrasonically activated phase-change contrast agents (PCCAs) based on perfluorocarbon droplets have been proposed for a variety of therapeutic and diagnostic clinical applications. When generated at the nanoscale, droplets may be small enough to exit the vascular space and then be induced to vaporize with high spatial and temporal specificity by externally-applied ultrasound. The use of acoustical techniques for optimizing ultrasound parameters for given applications can be a significant challenge for nanoscale PCCAs due to the contributions of larger outlier droplets. Similarly, optical techniques can be a challenge due to the sub-micron size of nanodroplet agents and resolution limits of optical microscopy. In this study, an optical method for determining activation thresholds of nanoscale emulsions based on the in vitro distribution of bubbles resulting from vaporization of PCCAs after single, short (<10 cycles) ultrasound pulses is evaluated. Through ultra-high-speed microscopy it is shown that the bubbles produced early in the pulse from vaporized droplets are strongly affected by subsequent cycles of the vaporization pulse, and these effects increase with pulse length. Results show that decafluorobutane nanoemulsions with peak diameters on the order of 200 nm can be optimally vaporized with short pulses using pressures amenable to clinical diagnostic ultrasound machines.


Asunto(s)
Emulsiones/química , Emulsiones/efectos de la radiación , Gases/química , Microscopía por Video/métodos , Nanopartículas/química , Nanopartículas/efectos de la radiación , Sonicación/métodos , Emulsiones/análisis , Gases/análisis , Gases/efectos de la radiación , Ondas de Choque de Alta Energía , Ensayo de Materiales/métodos , Nanopartículas/ultraestructura , Transición de Fase/efectos de la radiación , Dosis de Radiación
15.
Magn Reson Imaging ; 31(6): 900-10, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23583323

RESUMEN

Dynamic Contrast Enhancement (DCE) MRI has been used to measure the kinetic transport constant, K(trans), which is used to assess tumor angiogenesis and the effects of anti-angiogenic therapies. Standard DCE MRI methods must measure the pharmacokinetics of a contrast agent in the blood stream, known as the Arterial Input Function (AIF), which is then used as a reference for the pharmacokinetics of the agent in tumor tissue. However, the AIF is difficult to measure in pre-clinical tumor models and in patients. Moreover the AIF is dependent on the Fahraeus effect that causes a highly variable hematocrit (Hct) in tumor microvasculature, leading to erroneous estimates of K(trans). To overcome these problems, we have developed the Reference Agent Model (RAM) for DCE MRI analyses, which determines the relative K(trans) of two contrast agents that are simultaneously co-injected and detected in the same tissue during a single DCE-MRI session. The RAM obviates the need to monitor the AIF because one contrast agent effectively serves as an internal reference in the tumor tissue for the other agent, and it also eliminates the systematic errors in the estimated K(trans) caused by assuming an erroneous Hct. Simulations demonstrated that the RAM can accurately and precisely estimate the relative K(trans) (R(Ktrans)) of two agents. To experimentally evaluate the utility of RAM for analyzing DCE MRI results, we optimized a previously reported multiecho (19)F MRI method to detect two perfluorinated contrast agents that were co-injected during a single in vivo study and selectively detected in the same tumor location. The results demonstrated that RAM determined R(Ktrans) with excellent accuracy and precision.


Asunto(s)
Permeabilidad Capilar/fisiología , Compuestos de Flúor/farmacocinética , Interpretación de Imagen Asistida por Computador/métodos , Angiografía por Resonancia Magnética/métodos , Modelos Cardiovasculares , Neoplasias Experimentales/metabolismo , Neovascularización Patológica/metabolismo , Animales , Simulación por Computador , Medios de Contraste/farmacocinética , Femenino , Radioisótopos de Flúor , Aumento de la Imagen/métodos , Angiografía por Resonancia Magnética/normas , Tasa de Depuración Metabólica , Ratones , Ratones SCID , Neoplasias Experimentales/patología , Neovascularización Patológica/patología , Valores de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Distribución Tisular
16.
Ultrasound Med Biol ; 39(5): 893-902, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23453380

RESUMEN

Phase-change contrast agents (PCCAs), which normally consist of nanoscale or microscale droplets of liquid perfluorocarbons in an encapsulating shell, can be triggered to undergo a phase transition to the highly echogenic gaseous state upon the input of sufficient acoustic energy. As a result of the subsequent volumetric expansion, a number of unique applications have emerged that are not possible with traditional ultrasound microbubble contrast agents. Although many studies have explored the therapeutic aspects of the PCCA platform, few have examined the potential of PCCAs for molecular imaging purposes. In this study, we demonstrate a PCCA-based platform for molecular imaging using α(v)ß(3)-targeted nanoscale PCCAs composed of low-boiling-point perfluorocarbons. In vitro, nanoscale PCCAs adhered to target cells, could be activated and imaged with a clinical ultrasound system and produced a six-fold increase in image contrast compared with non-targeted control PCCAs and a greater than fifty-fold increase over baseline. Data suggest that low-boiling-point nanoscale PCCAs could enable future ultrasound-based molecular imaging techniques in both the vascular and extravascular spaces.


Asunto(s)
Medios de Contraste/análisis , Medios de Contraste/síntesis química , Células Endoteliales/diagnóstico por imagen , Fluorocarburos , Imagen Molecular/métodos , Ultrasonografía/métodos , Células Cultivadas , Femenino , Fluorocarburos/síntesis química , Humanos , Proyectos Piloto
17.
Biomaterials ; 33(11): 3262-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22289265

RESUMEN

Recently, an interest has developed in designing biomaterials for medical ultrasonics that can provide the acoustic activity of microbubbles, but with improved stability in vivo and a smaller size distribution for extravascular interrogation. One proposed alternative is the phase-change contrast agent. Phase-change contrast agents (PCCAs) consist of perfluorocarbons (PFCs) that are initially in liquid form, but can then be vaporized with acoustic energy. Crucial parameters for PCCAs include their sensitivity to acoustic energy, their size distribution, and their stability, and this manuscript provides insight into the custom design of PCCAs for balancing these parameters. Specifically, the relationship between size, thermal stability and sensitivity to ultrasound as a function of PFC boiling point and ambient temperature is illustrated. Emulsion stability and sensitivity can be 'tuned' by mixing PFCs in the gaseous state prior to condensation. Novel observations illustrate that stable droplets can be generated from PFCs with extremely low boiling points, such as octafluoropropane (b.p. -36.7 °C), which can be vaporized with acoustic parameters lower than previously observed. Results demonstrate the potential for low boiling point PFCs as a useful new class of compounds for activatable agents, which can be tailored to the desired application.


Asunto(s)
Medios de Contraste/síntesis química , Fluorocarburos/química , Fluorocarburos/efectos de la radiación , Gases/síntesis química , Gases/efectos de la radiación , Nanopartículas/efectos de la radiación , Sonicación/métodos , Medios de Contraste/efectos de la radiación , Ensayo de Materiales , Nanopartículas/química , Transición de Fase/efectos de la radiación , Temperatura de Transición , Ultrasonografía/métodos
18.
Theranostics ; 2(12): 1185-98, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23382775

RESUMEN

Recent efforts using perfluorocarbon (PFC) nanoparticles in conjunction with acoustic droplet vaporization has introduced the possibility of expanding the diagnostic and therapeutic capability of ultrasound contrast agents to beyond the vascular space. Our laboratories have developed phase-change nanoparticles (PCNs) from the highly volatile PFCs decafluorobutane (DFB, bp =-2 °C) and octafluoropropane (OFP, bp =-37 °C ) for acoustic droplet vaporization. Studies with commonly used clinical ultrasound scanners have demonstrated the ability to vaporize PCN emulsions with frequencies and mechanical indices that may significantly decrease tissue bioeffects. In addition, these contrast agents can be formulated to be stable at physiological temperatures and the perfluorocarbons can be mixed to modulate the balance between sensitivity to ultrasound and general stability. We herein discuss our recent efforts to develop finely-tuned diagnostic/molecular imaging agents for tissue interrogation. We discuss studies currently under investigation as well as potential diagnostic and therapeutic paradigms that may emerge as a result of formulating PCNs with low boiling point PFCs.


Asunto(s)
Diagnóstico por Imagen/métodos , Fluorocarburos/química , Nanopartículas/química , Transición de Fase , Ultrasonido/métodos , Animales , Humanos , Volatilización
19.
Langmuir ; 27(17): 10412-20, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21744860

RESUMEN

Recent efforts in the area of acoustic droplet vaporization with the objective of designing extravascular ultrasound contrast agents has led to the development of stabilized, lipid-encapsulated nanodroplets of the highly volatile compound decafluorobutane (DFB). We developed two methods of generating DFB droplets, the first of which involves condensing DFB gas (boiling point from -1.1 to -2 °C) followed by extrusion with a lipid formulation in HEPES buffer. Acoustic droplet vaporization of micrometer-sized lipid-coated droplets at diagnostic ultrasound frequencies and mechanical indices were confirmed optically. In our second formulation methodology, we demonstrate the formulation of submicrometer-sized lipid-coated nanodroplets based upon condensation of preformed microbubbles containing DFB. The droplets are routinely in the 200-300 nm range and yield microbubbles on the order of 1-5 µm once vaporized, consistent with ideal gas law expansion predictions. The simple and effective nature of this methodology allows for the development of a variety of different formulations that can be used for imaging, drug and gene delivery, and therapy. This study is the first to our knowledge to demonstrate both a method of generating ADV agents by microbubble condensation and formulation of primarily submicrometer droplets of decafluorobutane that remain stable at physiological temperatures. Finally, activation of DFB nanodroplets is demonstrated using pressures within the FDA guidelines for diagnostic imaging, which may minimize the potential for bioeffects in humans. This methodology offers a new means of developing extravascular contrast agents for diagnostic and therapeutic applications.


Asunto(s)
Acústica , Medios de Contraste/uso terapéutico , Fluorocarburos/uso terapéutico , Medios de Contraste/química , Fluorocarburos/química , Humanos , Tamaño de la Partícula , Propiedades de Superficie , Volatilización
20.
Ultrasound Med Biol ; 37(9): 1518-30, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21775049

RESUMEN

Currently available microbubbles used for ultrasound imaging and therapeutics are limited to intravascular space due to their size distribution in the micron range. Phase-change contrast agents (PCCAs) have been proposed as a means to overcome this limitation, since droplets formed in the hundred nanometer size range might be able to extravasate through leaky microvasculature, after which they could be activated to form larger highly echogenic microbubbles. Existing PCCAs in the sub-micron size range require substantial acoustic energy to be vaporized, increasing the likelihood of unwanted bioeffects. Thus, there exists a need for PCCAs with reduced acoustic activation energies for use in imaging studies. In this article, it is shown that decafluorobutane, which is normally a gas at room temperature, can be incorporated into metastable liquid sub-micron droplets with appropriate encapsulation methods. The resulting droplets are activatable with substantially less energy than other favored PCCA compounds. Decafluorobutane nanodroplets may present a new means to safely extend ultrasound imaging beyond the vascular space.


Asunto(s)
Medios de Contraste , Fluorocarburos , Microburbujas , Nanopartículas , Ultrasonografía/métodos , Acústica , Medios de Contraste/química , Fluorocarburos/química , Microvasos , Nanopartículas/química , Transductores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...