Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Electrophoresis ; 44(15-16): 1234-1246, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37431197

RESUMO

Dielectrophoresis (DEP) is a successful method to recover nanoparticles from different types of fluid. The DEP force acting on these particles is created by an electrode microarray that produces a nonuniform electric field. To apply DEP to a highly conducting biological fluid, a protective hydrogel coating over the metal electrodes is required to create a barrier between the electrode and the fluid. This protects the electrodes, reduces the electrolysis of water, and allows the electric field to penetrate into the fluid sample. We observed that the protective hydrogel layer can separate from the electrode and form a closed domed structure and that collection of 100 nm polystyrene beads increased when this occurred. To better understand this collection increase, we used COMSOL Multiphysics software to model the electric field in the presence of the dome filled with different materials ranging from low-conducting gas to high conducting phosphate-buffered saline fluids. The results suggest that as the electrical conductivity of the material inside the dome is reduced, the whole dome acts as an insulator which increases electric field intensity at the electrode edge. This increased intensity widens the high-intensity electric field factor zone resulting in increased collection. This informs how dome formation results in increased particle collection and provides insight into how the electric field can be intensified to the increase collection of particles. These results have important applications for increasing the recovery of biologically-derived nanoparticles from undiluted physiological fluids that have high conductance, including the collection of cancer-derived extracellular vesicles from plasma for liquid biopsy applications.


Assuntos
Eletricidade , Software , Eletroforese/métodos , Condutividade Elétrica , Eletrodos
2.
Pharm Res ; 34(10): 2025-2035, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28791550

RESUMO

PURPOSE: A major challenge facing nanoparticle-based delivery of chemotherapy agents is the natural and unavoidable accumulation of these particles in healthy tissue resulting in local toxicity and dose-limiting side effects. To address this issue, we have designed and characterized a new prodrug nanoparticle with controllable toxicity allowing a locally-delivered light trigger to convert the payload of the particle from a low to a high toxicity state. METHODS: The nanoparticles are created entirely from light-activatable prodrug molecules using a nanoprecipitation process. The prodrug is a conjugate of doxorubicin and photocleavable biotin (DOX-PCB). RESULTS: These DOX-PCB nanoparticles are 30 times less toxic to cells than doxorubicin, but can be activated to release pure therapeutic doxorubicin when exposed to 365 nm light. These nanoparticles have an average diameter of around 100 nm and achieve the maximum possible prodrug loading capacity since no support structure or coating is required to prevent loss of prodrug from the nanoparticle. CONCLUSIONS: These light activatable nanoparticles demonstrate tunable toxicity and can be used to facilitate future therapy development whereby light delivered specifically to the tumor tissue would locally convert the nanoparticles to doxorubicin while leaving nanoparticles accumulated in healthy tissue in the less toxic prodrug form.


Assuntos
Antineoplásicos/farmacologia , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Nanopartículas/química , Pró-Fármacos/química , Células A549 , Antineoplásicos/química , Antineoplásicos/toxicidade , Biotina/química , Linhagem Celular Tumoral , Doxorrubicina/química , Doxorrubicina/toxicidade , Liberação Controlada de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Luz , Tamanho da Partícula , Polietilenoglicóis/química , Pró-Fármacos/farmacologia , Propriedades de Superfície
3.
J Biol Phys ; 43(4): 493-510, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29124623

RESUMO

Gas-filled microbubbles attached to cell surfaces can interact with focused ultrasound to create microstreaming of nearby fluid. We directly observed the ultrasound/microbubble interaction and documented that under certain conditions fluorescent particles that were attached to the surface of live cells could be removed. Fluorescently labeled liposomes that were larger than 500 nm in diameter were attached to the surface of endothelial cells using cRGD targeting to αvß3 integrin. Microbubbles were attached to the surface of the cells through electrostatic interactions. Images taken before and after the ultrasound exposure were compared to document the effects on the liposomes. When exposed to ultrasound with peak negative pressure of 0.8 MPa, single microbubbles and groups of isolated microbubbles were observed to remove targeted liposomes from the cell surface. Liposomes were removed from a region on the cell surface that averaged 33.1 µm in diameter. The maximum distance between a single microbubble and a detached liposome was 34.5 µm. Single microbubbles were shown to be able to remove liposomes from over half the surface of a cell. The distance over which liposomes were removed was significantly dependent on the resting diameter of the microbubble. Clusters of adjoining microbubbles were not seen to remove liposomes. These observations demonstrate that the fluid shear forces generated by the ultrasound/microbubble interaction can remove liposomes from the surfaces of cells over distances that are greater than the diameter of the microbubble.


Assuntos
Adesão Celular , Lipossomos/isolamento & purificação , Lipossomos/metabolismo , Microbolhas , Ondas Ultrassônicas , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Ligantes , Eletricidade Estática , Propriedades de Superfície
4.
Small ; 11(38): 5088-96, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26274918

RESUMO

The effect of complex biological fluids on the surface and structure of nanoparticles is a rapidly expanding field of study. One of the challenges holding back this research is the difficulty of recovering therapeutic nanoparticles from biological samples due to their small size, low density, and stealth surface coatings. Here, the first demonstration of the recovery and analysis of drug delivery nanoparticles from undiluted human plasma samples through the use of a new electrokinetic platform technology is presented. The particles are recovered from plasma through a dielectrophoresis separation force that is created by innate differences in the dielectric properties between the unaltered nanoparticles and the surrounding plasma. It is shown that this can be applied to a wide range of drug delivery nanoparticles of different morphologies and materials, including low-density nanoliposomes. These recovered particles can then be analyzed using different methods including scanning electron microscopy to monitor surface and structural changes that result from plasma exposure. This new recovery technique can be broadly applied to the recovery of nanoparticles from high conductance fluids in a wide range of applications.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/química , Plasma/química , Eletrodos , Eletroforese , Humanos , Processamento de Imagem Assistida por Computador , Microfluídica , Nanopartículas/ultraestrutura , Dióxido de Silício/química , Espectrofotometria Ultravioleta
5.
Opt Lett ; 40(12): 2834-7, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-26076274

RESUMO

A new optical contrast agent has been developed by exposing dye-loaded microbubbles to a rapidly-cooled thermal treatment to homogenize the dye distribution across the surface. Ultrasound causes these microbubbles to oscillate in size which changes the self-quenching efficiency of the dye molecules creating a "blinking" signal. We demonstrate for the first time that these microbubbles can reproducibly generate second, third, and even fourth harmonic fluorescence intensity modulations, in addition to the fundamental frequency of the driving ultrasound. Detecting these harmonic signals could produce a higher signal-to-noise ratio for fluorescence imaging in medical applications by allowing fundamental frequency interference and artifacts to be filtered out.


Assuntos
Meios de Contraste , Corantes Fluorescentes , Microbolhas , Ondas Ultrassônicas , Temperatura Alta
6.
Small ; 10(16): 3316-24, 2014 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-24839198

RESUMO

The nanoscale surface features of lipid-coated microbubbles can dramatically affect how the lipids interact with one another as the microbubble diameter expands and contracts under the influence of ultrasound. During microbubble manufacturing, the different lipid shell species naturally partition forming concentrated lipid islands. In this study the dynamics of how these nanoscale islands accommodate the expansion of the microbubbles are monitored by measuring the fluorescence intensity changes that occur as self-quenching lipophilic dye molecules embedded in the lipid layer change their distance from one another. It was found that when the dye molecules were concentrated in islands, less than 5% of the microbubbles displayed measurable fluorescence intensity modulation indicating the islands were not able to expand sufficiently for the dye molecules to separate from one another. When the microbubbles were heated and cooled rapidly through the lipid transition temperature the islands were melted creating an even distribution of dye about the surface. This resulted in over 50% of the microbubbles displaying the fluorescence-modulated signal indicating that the dye molecules could now separate sufficiently to change their self-quenching efficiency. The separation of the surface lipids in these different formations has significant implications for microbubble development as ultrasound and optical contrast agents.


Assuntos
Fluorescência , Nanoestruturas , Ultrassom , Lipídeos/química
7.
J Acoust Soc Am ; 136(6): 3422, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25480086

RESUMO

The collapse dynamics of lipid monolayer-coated microbubbles in the clinically-relevant size range under 6 µm in diameter have not been studied directly due to their small size obscuring the collapse visualization. This study investigates the influence of inter-microbubble distance on the shape of lipid debris clouds created by the collapse of the microbubble destroying the microbubble lipid monolayer. The shape was highly influenced by the fluid motion that occurred as the microbubbles collapsed. It was observed that at inter-microbubble distances smaller than 37 µm the microbubbles began to interact with one another resulting in distorted and ellipsoid-shaped debris clouds. At inter-microbubble distances less than 10 µm, significantly elongated debris clouds were observed that extended out from the original microbubble location in a single direction. These distortions show a significant distance-dependent interaction between microbubbles. It was observed that microbubbles in physical contact with one another behaved in the same manner as separate microbubbles less than 10 µm apart creating significantly elongated debris clouds. It can be hypothesized that small inter-microbubble distances influence the microbubble to collapse asymmetrically resulting in the creation of fluid jets that contribute to the formation of debris fields that are elongated in a single direction.


Assuntos
Estimulação Acústica , Meios de Contraste , Hidrodinâmica , Microbolhas , Ultrassonografia , Lipídeos , Software , Gravação em Vídeo
8.
Adv Sci (Weinh) ; : e2309220, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39023197

RESUMO

Gut microbiome composition is tied to diseases ranging from arthritis to cancer to depression. However, mechanisms of action are poorly understood, limiting development of relevant therapeutics. Organ-on-chip platforms, which model minimal functional units of tissues and can tightly control communication between them, are ideal platforms to study these relationships. Many gut microbiome models are published to date but devices are typically fabricated using oxygen permeable polydimethylsiloxane, requiring interventions to support anaerobic bacteria. To address this challenge, a platform is developed where the chips are fabricated entirely from gas-impermeable polycarbonate without tapes or gaskets. These chips replicate polarized villus-like structures of the native tissue. Further, they enable co-cultures of commensal anaerobic bacteria Blautia coccoides on the surface of gut epithelia for two days within a standard incubator. Another complication of commonly used materials in organ-on-chip devices is high ad-/absorption, limiting applications in high-resolution microscopy and biomolecule interaction studies. For future communication studies between gut microbiota and distal tumors, an additional polycarbonate chip design is developed to support hydrogel-embedded tissue culture. These chips enable high-resolution microscopy with all relevant processing done on-chip. Designed for facile linking, this platform will make a variety of mechanistic studies possible.

9.
Soft Matter ; 9(8): 2384-2388, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23526919

RESUMO

Fluorescent microbubbles have been fabricated with the capacity to have their emission modulated by ultrasound. These contrast agent particles could potentially be used in the future to extract fluorescence modulation from a strong light background to increase imaging depth and resolution in scattering media. Fluorescence intensity modulation was demonstrated at the ultrasound driving frequency.

10.
Front Immunol ; 14: 1162905, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37081897

RESUMO

Augmenting adaptive immunity is a critical goal for developing next-generation cancer therapies. T and B cells infiltrating the tumor dramatically influence cancer progression through complex interactions with the local microenvironment. Cancer cells evade and limit these immune responses by hijacking normal immunologic pathways. Current experimental models using conventional primary cells, cell lines, or animals have limitations for studying cancer-immune interactions directly relevant to human biology and clinical translation. Therefore, engineering methods to emulate such interplay at local and systemic levels are crucial to expedite the development of better therapies and diagnostic tools. In this review, we discuss the challenges, recent advances, and future directions toward engineering the tumor-immune microenvironment (TME), including key elements of adaptive immunity. We first offer an overview of the recent research that has advanced our understanding of the role of the adaptive immune system in the tumor microenvironment. Next, we discuss recent developments in 3D in-vitro models and engineering approaches that have been used to study the interaction of cancer and stromal cells with B and T lymphocytes. We summarize recent advancement in 3D bioengineering and discuss the need for 3D tumor models that better incorporate elements of the complex interplay of adaptive immunity and the tumor microenvironment. Finally, we provide a perspective on current challenges and future directions for modeling cancer-immune interactions aimed at identifying new biological targets for diagnostics and therapeutics.


Assuntos
Neoplasias , Animais , Humanos , Neoplasias/patologia , Microambiente Tumoral
11.
Pharmaceutics ; 14(11)2022 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-36365214

RESUMO

Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble-nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid-polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin-avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble-nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.

12.
Adv Healthc Mater ; 10(1): e2001125, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32996270

RESUMO

Stem cell fate is closely intertwined with microenvironmental and endogenous cues within the body. Recapitulating this dynamic environment ex vivo can be achieved through engineered biomaterials which can respond to exogenous stimulation (including light, electrical stimulation, ultrasound, and magnetic fields) to deliver temporal and spatial cues to stem cells. These stimuli-responsive biomaterials can be integrated into scaffolds to investigate stem cell response in vitro and in vivo, and offer many pathways of cellular manipulation: biochemical cues, scaffold property changes, drug release, mechanical stress, and electrical signaling. The aim of this review is to assess and discuss the current state of exogenous stimuli-responsive biomaterials, and their application in multipotent stem cell control. Future perspectives in utilizing these biomaterials for personalized tissue engineering and directing organoid models are also discussed.


Assuntos
Células-Tronco , Engenharia Tecidual , Materiais Biocompatíveis , Diferenciação Celular , Alicerces Teciduais
13.
Lab Chip ; 21(7): 1318-1332, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33877235

RESUMO

Tumor-secreted exosomes and other extracellular vesicles (EVs) in circulation contain valuable biomarkers for early cancer detection and screening. We have previously demonstrated collection of cancer-derived nanoparticles (NPs) directly from whole blood and plasma with a chip-based technique that uses a microelectrode array to generate dielectrophoretic (DEP) forces. This technique enables direct recovery of NPs from whole blood and plasma. The biomarker payloads associated with collected particles can be detected and quantified with immunostaining. Accurately separating the fluorescence intensity of stained biomarkers from background (BG) levels becomes a challenge when analyzing the blood from early-stage cancer patients in which biomarker concentrations are low. To address this challenge, we developed two complementary techniques to standardize the quantification of fluorescently immunolabeled biomarkers collected and concentrated at predictable locations within microfluidic chips. The first technique was an automated algorithm for the quantitative analysis of fluorescence intensity at collection regions within the chip compared to levels at adjacent regions. The algorithm used predictable locations of particle collection within the chip geometry to differentiate regions of collection and BG. We successfully automated the identification and removal of optical artifacts from quantitative calculations. We demonstrated that the automated system performs nearly the same as a human user following a standard protocol for manual artifact removal with Pearson's r-values of 0.999 and 0.998 for two different biomarkers (n = 36 patients). We defined a usable dynamic range of fluorescence intensities corresponding to 1 to 2000 arbitrary units (a.u.). Fluorescence intensities within the dynamic range increased linearly with respect to exposure time and particle concentration. The second technique was the implementation of an internal standard to adjust levels of biomarker fluorescence based on the relative collection efficiency of the chip. Use of the internal standard reduced variability in measured biomarker levels due to differences in chip-to-chip collection efficiency, especially at low biomarker concentrations. The internal standard did not affect linear trends between fluorescence intensity and exposure time. Adjustments using the internal standard improved linear trends between fluorescence intensity and particle concentration. The optical quantification techniques described in this paper can be easily adapted for other lab-on-a-chip platforms that have predefined regions of biomarker or particle collection and that rely on fluorescence detection.


Assuntos
Exossomos , Vesículas Extracelulares , Humanos , Dispositivos Lab-On-A-Chip , Microfluídica , Plasma
14.
Adv Mater ; 32(7): e1905914, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31922627

RESUMO

Hydrogels are formed using various triggers, including light irradiation, pH adjustment, heating, cooling, or chemical addition. Here, a new method for forming hydrogels is introduced: ultrasound-triggered enzymatic gelation. Specifically, ultrasound is used as a stimulus to liberate liposomal calcium ions, which then trigger the enzymatic activity of transglutaminase. The activated enzyme catalyzes the formation of fibrinogen hydrogels through covalent intermolecular crosslinking. The catalysis and gelation processes are monitored in real time and both the enzyme kinetics and final hydrogel properties are controlled by varying the initial ultrasound exposure time. This technology is extended to microbubble-liposome conjugates, which exhibit a stronger response to the applied acoustic field and are also used for ultrasound-triggered enzymatic hydrogelation. To the best of the knowledge, these results are the first instance in which ultrasound is used as a trigger for either enzyme catalysis or enzymatic hydrogelation. This approach is highly versatile and can be readily applied to different ion-dependent enzymes or gelation systems. Moreover, this work paves the way for the use of ultrasound as a remote trigger for in vivo hydrogelation.


Assuntos
Enzimas/química , Hidrogéis/química , Ondas Ultrassônicas , Cloreto de Cálcio/química , Catálise , Reagentes de Ligações Cruzadas/química , Fibrinogênio/química , Cinética , Lipossomos/química , Microbolhas , Fosfatidiletanolaminas/química , Fosforilcolina/química , Polietilenoglicóis/química
15.
Acad Radiol ; 13(3): 363-7, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16488849

RESUMO

RATIONALE AND OBJECTIVES: This experiment was directed to explore the effects of ultrasound microbubbles on gene structure in vitro and green fluorescent protein (GFP) plasmid transfer into skeletal muscles in vivo. By establishing a rat ischemic hind limb model, the effects of ultrasound-mediated microbubble destruction on vascular endothelial growth factor (VEGF) gene transfection to skeletal muscles were also studied in vivo. MATERIALS AND METHODS: Ultrasound irradiation was applied on the mixture of microbubbles and GFP plasmid in vitro. Gel electrophoresis was used to detect the effects of ultrasound and microbubbles on GFP plasmid. For in vivo experiments, ultrasound irradiation was applied on the hind limb after directly injecting microbubbles into the hind limb of Wistar rats. Directly after treatment, the skeletal muscles were harvested to observe the microstructure. We also studied the transfer rate of GFP plasmid DNA into the skeletal muscles of rats by applying ultrasound and microbubble technique. Furthermore, a naked VEGF plasmid was applied to study the feasibility of angiogenesis by using rats ischemia models. RESULTS: Gel electrophoresis of plasmid DNA showed that there was no difference between the groups. By studying the hematoxylin and eosin stained pictures of the skeletal muscles, we found that ultrasound irradiation of skeletal muscle after injection of microbubbles could cause the exudation of the red blood cells, whereas it had no effects on the microstructure of muscle fibers. In vivo experiments showed that an ultrasound microbubble could enhance the transfer of plasmid DNA to the skeletal muscles. CONCLUSIONS: The ultrasound-mediated microbubble technique provides an effective noninvasive method for gene therapy.


Assuntos
Técnicas de Transferência de Genes , Microbolhas , Músculo Esquelético , Ultrassom , Animais , Proteínas de Fluorescência Verde/genética , Músculo Esquelético/citologia , Plasmídeos , Ratos , Ratos Wistar , Transfecção , Fator A de Crescimento do Endotélio Vascular/genética
16.
Nat Commun ; 6: 8264, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26372413

RESUMO

A major challenge in neuroscience is to reliably activate individual neurons, particularly those in deeper brain regions. Current optogenetic approaches require invasive surgical procedures to deliver light of specific wavelengths to target cells to activate or silence them. Here, we demonstrate the use of low-pressure ultrasound as a non-invasive trigger to activate specific ultrasonically sensitized neurons in the nematode, Caenorhabditis elegans. We first show that wild-type animals are insensitive to low-pressure ultrasound and require gas-filled microbubbles to transduce the ultrasound wave. We find that neuron-specific misexpression of TRP-4, the pore-forming subunit of a mechanotransduction channel, sensitizes neurons to ultrasound stimulus, resulting in behavioural outputs. Furthermore, we use this approach to manipulate the function of sensory neurons and interneurons and identify a role for PVD sensory neurons in modifying locomotory behaviours. We suggest that this method can be broadly applied to manipulate cellular functions in vivo.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Interneurônios/fisiologia , Locomoção/fisiologia , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPC/genética , Ondas Ultrassônicas , Animais , Caenorhabditis elegans , Interneurônios/metabolismo , Microbolhas , Neurônios/metabolismo , Neurônios/fisiologia , Células Receptoras Sensoriais/metabolismo
17.
Ultrasonics ; 54(8): 2090-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25059435

RESUMO

Lipid monolayer coated microbubbles are currently being developed to identify vascular regions that express certain surface proteins as part of the new technique of ultrasound molecular imaging. The microbubbles are functionalized with targeting ligands which bind to the desired cells holding the microbubbles in place as the remaining unbound microbubbles are eliminated from circulation. Subsequent scanning with ultrasound can detect the highly reflectant microbubbles that are left behind. The ultrasound scanning and detection process results in the destruction of the microbubble, creating lipid fragments from the monolayer. Here we demonstrate that microbubbles targeted to 4T1 murine breast cancer cells and human umbilical cord endothelial cells leave behind adhered fragments of the lipid monolayer after exposure to ultrasound with peak negative pressures of 0.18 and 0.8MPa. Most of the observed fragments were large enough to be resistant to receptor mediated endocytosis. The fragments were not observed to incorporate into the lipid membrane of the cell over a period of 96min. They were not observed to break into smaller pieces or significantly change shape but they were observed to undergo translation and rotation across the cell surface as the cells migrated over the substrate. These large fragments will apparently remain on the surface of the targeted cells for significant periods of time and need to be considered for their potential effects on blood flow through the microcapillaries and potential for immune system recognition.


Assuntos
Membrana Celular , Lipídeos/química , Microbolhas , Imagem Molecular/métodos , Ultrassom , Veias Umbilicais/citologia , Antígenos de Neoplasias/química , Moléculas de Adesão Celular/química , Técnicas de Cultura de Células , Endotélio Vascular/citologia , Molécula de Adesão da Célula Epitelial , Desenho de Equipamento , Humanos , Lecitinas/química , Proteínas de Membrana/química , Microscopia de Fluorescência , Peptídeos Cíclicos/química , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenoglicóis/química
18.
Drug Des Devel Ther ; 7: 375-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667309

RESUMO

The inherently toxic nature of chemotherapy drugs is essential for them to kill cancer cells but is also the source of the detrimental side effects experienced by patients. One strategy to reduce these side effects is to limit the healthy tissue exposure by encapsulating the drugs in a vehicle that demonstrates a very low leak rate in circulation while simultaneously having the potential for rapid release once inside the tumor. Designing a vehicle with these two opposing properties is the major challenge in the field of drug delivery. A triggering event is required to change the vehicle from its stable circulating state to its unstable release state. A unique mechanical actuation type trigger is possible by harnessing the size changes that occur when microbubbles interact with ultrasound. These mechanical actuations can burst liposomes and cell membranes alike allowing for rapid drug release and facilitating delivery into nearby cells. The tight focusing ability of the ultrasound to just a few cubic millimeters allows for precise control over the tissue location where the microbubbles destabilize the vehicles. This allows the ultrasound to highlight the tumor tissue and cause rapid drug release from any carrier present. Different vehicle designs have been demonstrated from carrying drug on just the surface of the microbubble itself to encapsulating the microbubble along with the drug within a liposome. In the future, nanoparticles may extend the circulation half-life of these ultrasound triggerable drug-delivery vehicles by acting as nucleation sites of ultrasound-induced mechanical actuation. In addition to the drug delivery capability, the microbubble size changes can also be used to create imaging contrast agents that could allow the internal chemical environment of a tumor to be studied to help improve the diagnosis and detection of cancer. The ability to attain truly tumor-specific release from circulating drug-delivery vehicles is an exciting future prospect to reduce chemotherapy side effects while increasing drug effectiveness.


Assuntos
Antineoplásicos/administração & dosagem , Microbolhas , Neoplasias/terapia , Veículos Farmacêuticos , Terapia por Ultrassom/métodos , Diagnóstico por Imagem , Humanos
19.
J Control Release ; 155(3): 358-66, 2011 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-21745505

RESUMO

The use of focused ultrasound can be an effective method to locally highlight tumor tissue and specifically trigger the activation of echogenic drug delivery vehicles in an effort to reduce systemic chemotherapy side effects. Here we demonstrate a unique ultrasound triggered vehicle design and fabrication method where the payload and a perfluorocarbon gas microbubble are both encapsulated within the internal aqueous space of a liposome. This nested lipid shell geometry both stabilized the microbubble and ensured it was spatially close enough to interact with the liposome membrane at all times. The internal microbubble was shown to fragment the outer liposome membrane upon exposure to ultrasound at intensities of 1-1.5MPa. The focused ultrasound allowed the release of the internal payload to localized regions within tissue phantoms. The vehicles showed high payload loading efficiency of 16%, stability in blood of several hours, and low level macrophage recognition in vitro. High speed fluorescent videos present the first optical images of such vehicles interacting with ultrasound. This ability to open the outer membrane in small regions of deep tissue could provide a second level of spatial and temporal control beyond biochemical targeting, making these particles promising for in vivo animal studies.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Ondas de Choque de Alta Energia , Microbolhas , Veículos Farmacêuticos/química , Estabilidade de Medicamentos , Fluorocarbonos/química , Lipídeos/química , Lipossomos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA