Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Adv Healthc Mater ; 12(30): e2301787, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37660271

RESUMO

The demand for engineered scaffolds capable of delivering multiple cues to cells continues to grow as the interplay between cell fate with microenvironmental and external cues is revealed. Emphasis has been given to develop stimuli-responsive scaffolds. These scaffolds are designed to sense an external stimulus triggering a specific response (e.g., change in the microenvironment, release therapeutics, etc.) and then initiate/modulate a desired biofunction. Here, magnetic-responsive carboxylated multi-walled carbon nanotubes (cMWCNTs) are integrated into 3D collagen/polylactic acid (PLA) scaffold via a reproducible filtration-based method. The integrity and biomechanical performance of the collagen/PLA scaffolds are preserved after cMWCNT integration. In vitro safety assessment of cMWCNT/collagen/PLA scaffolds shows neither cytotoxicity effects nor macrophage pro-inflammatory response, supporting further in vitro studies. The cMWCNT/collagen/PLA scaffolds enhance chondrocytes metabolic activity while maintaining high cell viability and extracellular matrix (i.e., type II collagen and aggrecan) production. Comprehensive in vitro study applying static and pulsed magnetic field on seeded scaffolds shows no specific cell response in dependence with the applied field. This result is independent of the presence or absence of cMWCNT into the collagen/PLA scaffolds. Taken together, these findings provide additional evidence of the benefits to exploit the CNTs outstanding properties in the design of stimuli-responsive scaffolds.


Assuntos
Nanotubos de Carbono , Engenharia Tecidual , Engenharia Tecidual/métodos , Alicerces Teciduais , Colágeno , Poliésteres , Fenômenos Magnéticos
2.
J Control Release ; 357: 67-83, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36921725

RESUMO

K-RAS is a highly relevant oncogene that is mutated in approximately 90% of pancreatic cancers and 20-25% of lung adenocarcinomas. The aim of this work was to develop a new anti-KRAS siRNA therapeutic strategy through the engineering of functionalized lipid nanoparticles (LNPs). To do this, first, a potent pan anti-KRAS siRNA sequence was chosen from the literature and different chemical modifications of siRNA were tested for their transfection efficacy (KRAS knockdown) and anti-proliferative effects on various cancer cell lines. Second, a selected siRNA candidate was loaded into tLyp-1 targeted and non-targeted lipid nanoparticles (LNPs). The biodistribution and antitumoral efficacy of selected siRNA-loaded LNP-prototypes were evaluated in vivo using a pancreatic cancer murine model (subcutaneous xenograft CFPAC-1 tumors). Our results show that tLyp-1-tagged targeted LNPs have an enhanced accumulation in the tumor compared to non-targeted LNPs. Moreover, a significant reduction in the pancreatic tumor growth was observed when the anti-KRAS siRNA treatment was combined with a classical chemotherapeutic agent, gemcitabine. In conclusion, our work demonstrates the benefits of using a targeting approach to improve tumor accumulation of siRNA-LNPs and its positive impact on tumor reduction.


Assuntos
Nanopartículas , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/uso terapêutico , Distribuição Tecidual , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética
3.
Biomacromolecules ; 24(10): 4444-4453, 2023 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-36753733

RESUMO

Polymeric micelles are among the most extensively used drug delivery systems. Key properties of micelles, such as size, size distribution, drug loading, and drug release kinetics, are crucial for proper therapeutic performance. Whether polymers from more controlled polymerization methods produce micelles with more favorable properties remains elusive. To address this question, we synthesized methoxy poly(ethylene glycol)-b-(N-(2-benzoyloxypropyl)methacrylamide) (mPEG-b-p(HPMAm-Bz)) block copolymers of three different comparable molecular weights (∼9, 13, and 20 kDa), via both conventional free radical (FR) and reversible addition-fragmentation chain transfer (RAFT) polymerization. The polymers were subsequently employed to prepare empty and paclitaxel-loaded micelles. While FR polymers had relatively high dispersities (D ∼ 1.5-1.7) compared to their RAFT counterparts (D ∼ 1.1-1.3), they formed micelles with similar pharmaceutical properties (e.g., size, size distribution, critical micelle concentration, cytotoxicity, and drug loading and retention). Our findings suggest that pharmaceutical properties of mPEG-b-p(HPMAm-Bz) micelles do not depend on the synthesis route of their constituent polymers.


Assuntos
Elétrons , Micelas , Polimerização , Polietilenoglicóis , Polímeros , Portadores de Fármacos
4.
Int J Pharm X ; 4: 100124, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35898812

RESUMO

Poly (alkyl cyanoacrylate) (PACA) polymeric nanoparticles (NPs) are promising drug carriers in drug delivery. However, the selection of commercially available alkyl cyanoacrylate (ACA) monomers is limited, because most monomers were designed for use in medical and industrial glues and later repurposed for drug encapsulation. This study therefore aimed to seek out novel ACA materials for use in NP systems using a toxicity led screening approach. A multistep strategy, including cytotoxicity screening of alcohols as degradation products of PACA (44 alcohols), NPs (14 polymers), and a final in vivo study (2 polymers) gave poly (2-ethylhexyl cyanoacrylate) PEHCA as a promising novel PACA candidate. For the first time, this work presents cytotoxicity data on several novel ACAs, PEHCA in vivo toxicity data, and miniemulsion polymerisation-based encapsulation of the cabazitaxel and NR688 in novel PACA candidates. Furthermore, several of the ACA candidates were compatible with a wider selection of lipophilic active pharmaceutical ingredients (APIs) versus commercially available controls. Combined, this work demonstrates the potential benefits of expanding the array of available ACA materials in drug delivery. Novel ACAs have the potential to encapsulate a wider range of APIs in miniemulsion polymerisation processes and may also broaden PACA applicability in other fields.

5.
Drug Deliv Transl Res ; 12(9): 2207-2224, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35799027

RESUMO

State-of-the-art in vitro test systems for nanomaterial toxicity assessment are based on dyes and several staining steps which can be affected by nanomaterial interference. Digital holographic microscopy (DHM), an interferometry-based variant of quantitative phase imaging (QPI), facilitates reliable proliferation quantification of native cell populations and the extraction of morphological features in a fast and label- and interference-free manner by biophysical parameters. DHM therefore has been identified as versatile tool for cytotoxicity testing in biomedical nanotechnology. In a comparative study performed at two collaborating laboratories, we investigated the interlaboratory variability and performance of DHM in nanomaterial toxicity testing, utilizing complementary standard operating procedures (SOPs). Two identical custom-built off-axis DHM systems, developed for usage in biomedical laboratories, equipped with stage-top incubation chambers were applied at different locations in Europe. Temporal dry mass development, 12-h dry mass increments and morphology changes of A549 human lung epithelial cell populations upon incubation with two variants of poly(alkyl cyanoacrylate) (PACA) nanoparticles were observed in comparison to digitonin and cell culture medium controls. Digitonin as cytotoxicity control, as well as empty and cabazitaxel-loaded PACA nanocarriers, similarly impacted 12-h dry mass development and increments as well as morphology of A549 cells at both participating laboratories. The obtained DHM data reflected the cytotoxic potential of the tested nanomaterials and are in agreement with corresponding literature on biophysical and chemical assays. Our results confirm DHM as label-free cytotoxicity assay for polymeric nanocarriers as well as the repeatability and reproducibility of the technology. In summary, the evaluated DHM assay could be efficiently implemented at different locations and facilitates interlaboratory in vitro toxicity testing of nanoparticles with prospects for application in regulatory science.


Assuntos
Holografia , Microscopia , Digitonina , Holografia/métodos , Humanos , Técnicas In Vitro , Microscopia/métodos , Reprodutibilidade dos Testes
6.
Drug Deliv Transl Res ; 12(9): 2039-2041, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35882743
7.
Drug Deliv Transl Res ; 12(9): 2114-2131, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35426570

RESUMO

Biodistribution of nanoencapsulated bioactive compounds is primarily determined by the size, shape, chemical composition and surface properties of the encapsulating nanoparticle, and, thus, less dependent on the physicochemical properties of the active pharmaceutical ingredient encapsulated. In the current work, we aimed to investigate the impact of formulation type on biodistribution profile for two clinically relevant nanoformulations. We performed a comparative study of biodistribution in healthy rats at several dose levels and durations up to 14-day post-injection. The studied nanoformulations were nanostructured lipid carriers incorporating the fluorescent dye IR780-oleyl, and polymeric nanoparticles containing the anticancer agent cabazitaxel. The biodistribution was approximated by quantification of the cargo in blood and relevant organs. Several clear and systematic differences in biodistribution were observed, with the most pronounced being a much higher (more than 50-fold) measured concentration ratio between cabazitaxel in all organs vs. blood, as compared to IR780-oleyl. Normalized dose linearity largely showed opposite trends between the two compounds after injection. Cabazitaxel showed a higher brain accumulation than IR780-oleyl with increasing dose injected. Interestingly, cabazitaxel showed a notable and prolonged accumulation in lung tissue compared to other organs. The latter observations could warrant further studies towards a possible therapeutic indication within lung and conceivably brain cancer for nanoformulations of this highly antineoplastic compound, for which off-target toxicity is currently dose-limiting in the clinic.


Assuntos
Antineoplásicos , Nanopartículas , Nanoestruturas , Animais , Portadores de Fármacos/química , Lipídeos/química , Nanopartículas/química , Polímeros , Ratos , Distribuição Tecidual
8.
Drug Deliv Transl Res ; 12(9): 2089-2100, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35318565

RESUMO

The development of drug nanocarriers based on polymeric, lipid and ceramic biomaterials has been paving the way to precision medicine, where the delivery of poorly soluble active compounds and personalized doses are made possible. However, the nano-size character of these carriers has been demonstrated to have the potential to elicit pathways of the host response different from those of the same biomaterials when engineered as larger size implants and of the drugs when administered without a carrier. Therefore, a specific regulatory framework needs to be made available that can offer robust scientific insights and provide safety data by reliable tests of these novel nano-devices. In this context, the present work presents a multistep protocol for the in vitro assessment of the hemocompatibility of nanocarriers of different physicochemical properties. Poly (ethyl butyl cyanoacrylate) nanoparticles and lipid-based (LipImage™ 815) nanoparticles of comparable hydrodynamic diameter were tested through a battery of assays using human peripheral blood samples and recapitulating the main pathways of the host response upon systemic administration; i.e., protein interactions, fibrinogen-platelet binding, cytotoxicity, and inflammatory response. The data showed the sensitivity and reproducibility of the methods adopted that were also demonstrated to determine individual variability as well as to discriminate between activation of pathways of inflammation and unintended release of inflammatory signaling caused by loss of cell integrity. Therefore, this multistep testing is proposed as a reliable protocol for nanoparticle development and emerging regulatory frameworks.


Assuntos
Nanopartículas , Materiais Biocompatíveis , Portadores de Fármacos/química , Reação a Corpo Estranho , Humanos , Lipídeos/química , Teste de Materiais , Nanopartículas/química , Preparações Farmacêuticas , Reprodutibilidade dos Testes
9.
Drug Deliv Transl Res ; 12(9): 2075-2088, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35182369

RESUMO

The detection of biomedical organic nanocarriers in cells and tissues is still an experimental challenge. Here we developed an imaging strategy for the label-free detection of poly (ethylbutyl cyanoacrylate) (PEBCA) particles. Experiments were carried out with phagocytic NR8383 macrophages exposed to non-toxic and non-activating concentrations of fluorescent (PEBCA NR668 and PEBCA NR668/IR), non-fluorescent (PEBCA), and cabazitaxel-loaded PEBCA particles (PEBCA CBZ). Exposure to PEBCA NR668 revealed an inhomogeneous particle uptake similar to what was obtained with the free modified Nile Red dye (NR668). In order to successfully identify the PEBCA-loaded cells under label-free conditions, we developed an imaging strategy based on enhanced darkfield microscopy (DFM), followed by confocal Raman microscopy (CRM) and time-of-flight secondary ion mass spectrometry (ToF-SIMS). Nitrile groups of the PEBCA matrix and PEBCA ions were used as suitable analytes for CRM and ToF-SIMS, respectively. Masses found with ToF-SIMS were further confirmed by Orbitrap-SIMS. The combined approach allowed to image small (< 1 µm) PEBCA-containing phagolysosomes, which were identified as PEBCA-containing compartments in NR8383 cells by electron microscopy. The combination of DFM, CRM, and ToF-SIMS is a promising strategy for the label-free detection of PEBCA particles.


Assuntos
Cianoacrilatos , Espectrometria de Massa de Íon Secundário , Macrófagos , Microscopia Confocal , Espectrometria de Massa de Íon Secundário/métodos
10.
J Control Release ; 326: 164-171, 2020 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-32681950

RESUMO

The situation of the COVID-19 pandemic reminds us that we permanently need high-value flexible solutions to urgent clinical needs including simplified diagnostic technologies suitable for use in the field and for delivering targeted therapeutics. From our perspective nanotechnology is revealed as a vital resource for this, as a generic platform of technical solutions to tackle complex medical challenges. It is towards this perspective and focusing on nanomedicine that we take issue with Prof Park's recent editorial published in the Journal of Controlled Release. Prof. Park argued that in the last 15 years nanomedicine failed to deliver the promised innovative clinical solutions to the patients (Park, K. The beginning of the end of the nanomedicine hype. Journal of Controlled Release, 2019; 305, 221-222 [1]. We, the ETPN (European Technology Platform on Nanomedicine) [2], respectfully disagree. In fact, the more than 50 formulations currently in the market, and the recent approval of 3 key nanomedicine products (e. g. Onpattro, Hensify and Vyxeos), have demonstrated that the nanomedicine field is concretely able to design products that overcome critical barriers in conventional medicine in a unique manner, but also to deliver within the cells new drug-free therapeutic effects by using pure physical modes of action, and therefore make a difference in patients lives. Furthermore, the >400 nanomedicine formulations currently in clinical trials are expecting to bring novel clinical solutions (e.g. platforms for nucleic acid delivery), alone or in combination with other key enabling technologies to the market, including biotechnologies, microfluidics, advanced materials, biomaterials, smart systems, photonics, robotics, textiles, Big Data and ICT (information & communication technologies) more generally. However, we agree with Prof. Park that " it is time to examine the sources of difficulty in clinical translation of nanomedicine and move forward ". But for reaching this goal, the investments to support clinical translation of promising nanomedicine formulations should increase, not decrease. As recently encouraged by EMA in its roadmap to 2025, we should create more unity through a common knowledge hub linking academia, industry, healthcare providers and hopefully policy makers to reduce the current fragmentation of the standardization and regulatory body landscape. We should also promote a strategy of cross-technology innovation, support nanomedicine development as a high value and low-cost solution to answer unmet medical needs and help the most promising innovative projects of the field to get better and faster to the clinic. This global vision is the one that the ETPN chose to encourage for the last fifteen years. All actions should be taken with a clear clinical view in mind, " without any fanfare", to focus "on what matters in real life", which is the patient and his/her quality of life. This ETPN overview of achievements in nanomedicine serves to reinforce our drive towards further expanding and growing the maturity of nanomedicine for global healthcare, accelerating the pace of transformation of its great potential into tangible medical breakthroughs.


Assuntos
Sistemas de Liberação de Medicamentos , Nanomedicina , Animais , COVID-19 , Ensaios Clínicos como Assunto , Infecções por Coronavirus/terapia , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos/métodos , Humanos , Nanomedicina/métodos , Nanotecnologia/métodos , Neoplasias/terapia , Pandemias , Pneumonia Viral/terapia
11.
Drug Deliv Transl Res ; 10(3): 726-729, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32141035

RESUMO

This commentary article conveys the views of the board of the Nanomedicine and Nanoscale Delivery Focus Group of the Controlled Release Society regarding the decision of the United States National Cancer Institute (NCI) in halting funding for the Centers of Cancer Nanotechnology Excellence (CCNEs), and the subsequent editorial articles that broadened this discussion. Graphical abstract.


Assuntos
Nanomedicina/economia , National Cancer Institute (U.S.)/organização & administração , Neoplasias/tratamento farmacológico , Grupos Focais , Humanos , Estados Unidos
15.
Expert Opin Drug Deliv ; 15(12): 1249-1261, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30415585

RESUMO

INTRODUCTION: Ultrasound in combination with microbubbles can make cells and tissues more accessible for drugs, thereby achieving improved therapeutic outcomes. In this review, we introduce the term 'sonopermeation', covering mechanisms such as pore formation (traditional sonoporation), as well as the opening of intercellular junctions, stimulated endocytosis/transcytosis, improved blood vessel perfusion and changes in the (tumor) microenvironment. Sonopermeation has gained a lot of interest in recent years, especially for delivering drugs through the otherwise impermeable blood-brain barrier, but also to tumors. AREAS COVERED: In this review, we summarize various in vitro assays and in vivo setups that have been employed to unravel the fundamental mechanisms involved in ultrasound-enhanced drug delivery, as well as clinical trials that are ongoing in patients with brain, pancreatic, liver and breast cancer. We summarize the basic principles of sonopermeation, describe recent findings obtained in (pre-) clinical trials, and discuss future directions. EXPERT OPINION: We suggest that an improved mechanistic understanding, and microbubbles and ultrasound equipment specialized for drug delivery (and not for imaging) are key aspects to create more effective treatment regimens by sonopermeation. Real-time feedback and tools to predict therapeutic outcome and which tumors/patients will benefit from sonopermeation-based interventions will be important to promote clinical translation.


Assuntos
Sistemas de Liberação de Medicamentos , Microbolhas , Neoplasias/tratamento farmacológico , Animais , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Endocitose , Humanos , Microambiente Tumoral , Ultrassonografia
16.
Biointerphases ; 13(1): 011004, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382205

RESUMO

The primary goal of our investigation was the development of a versatile immobilization matrix based on archaeal tetraether lipids that meets the most important prerequisites to render an implant surface bioactive by binding specific functional groups or functional polymers with the necessary flexibility and an optimal spatial arrangement to be bioavailable. From this point of view, it appears obvious that numerous efforts made recently to avoid initial bacterial adhesion on catheter surfaces as an important prerequisite of material associated infection episodes have shown only a limited efficiency since the bioactive entities could not be presented in an optimal conformation and a stable density. A significant improvement of this situation can be achieved by highly specific biomimetic modifications of the catheter surfaces. The term "biomimetic" originates from the fact that specific archaeal tetraether lipids were introduced to form a membrane analog monomolecular spacer system, which (1) can be immobilized on nearly all solid surfaces and (2) chemically modified to present a tailor-made functionality in contact with aqueous media either to avoid or inhibit surface fouling or to equip any implant surface with the necessary chemical functionality to enable cell adhesion and tissue integration. Ultrathin films based on tetraether lipids isolated from archaea Thermoplasma acidophilum were used as a special biomimetic immobilization matrix on the surface of commercial medical silicon elastomers. A complete performance control of the membrane analog coatings was realized in addition to biofunctionality tests, including the proof of cytotoxicity and hemocompatibility according to DIN EN ISO 10993. In order to make sure that the developed immobilization matrix including the grafted functional groups are biocompatible under in vivo-conditions, specific animal tests were carried out to examine the in vivo-performance. It can be concluded that the tetraether lipid based coating systems on silicone have shown no signs of cytotoxicity and a good hemocompatibility. Moreover, no mutagenic effects, no irritation effects, and no sensitization effects could be demonstrated. After an implantation period of 28 days, no irregularities were found.


Assuntos
Materiais Biomiméticos/síntese química , Materiais Revestidos Biocompatíveis/síntese química , Lipídeos/isolamento & purificação , Membranas/metabolismo , Thermoplasma/química , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/toxicidade , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/toxicidade , Hemólise , Humanos , Metabolismo dos Lipídeos , Membranas/química , Camundongos , Próteses e Implantes/efeitos adversos , Coelhos , Silício , Propriedades de Superfície
17.
Int J Mol Sci ; 18(11)2017 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-29156588

RESUMO

Although nanotoxicology has become a large research field, assessment of cytotoxicity is often reduced to analysis of one cell line only. Cytotoxicity of nanoparticles is complex and should, preferentially, be evaluated in several cell lines with different methods and on multiple nanoparticle batches. Here we report the toxicity of poly(alkyl cyanoacrylate) nanoparticles in 12 different cell lines after synthesizing and analyzing 19 different nanoparticle batches and report that large variations were obtained when using different cell lines or various toxicity assays. Surprisingly, we found that nanoparticles with intermediate degradation rates were less toxic than particles that were degraded faster or more slowly in a cell-free system. The toxicity did not vary significantly with either the three different combinations of polyethylene glycol surfactants or with particle size (range 100-200 nm). No acute pro- or anti-inflammatory activity on cells in whole blood was observed.


Assuntos
Cianoacrilatos/toxicidade , Nanopartículas/toxicidade , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica , Cianoacrilatos/química , Feminino , Células Hep G2 , Humanos , Masculino , Nanopartículas/química , Tamanho da Partícula , Polietilenoglicóis , Tensoativos
18.
Mol Pharm ; 14(8): 2560-2569, 2017 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-28170271

RESUMO

Protein adsorption on nanoparticles (NPs) used in nanomedicine leads to opsonization and activation of the complement system in blood, which substantially reduces the blood circulation time of NPs. The most commonly used method to avoid protein adsorption is to coat the NPs with polyethylene glycol, so-called PEGylation. Although PEGylation is of utmost importance for designing the in vivo behavior of the NP, there is still a considerable lack of methods for characterization and fundamental understanding related to the PEGylation of NPs. In this work we have studied four different poly(butyl cyanoacrylate) (PBCA) NPs, PEGylated with different types of PEG-based nonionic surfactants-Jeffamine M-2070, Brij L23, Kolliphor HS 15, Pluronic F68-or combinations thereof. We evaluated the PEGylation, both quantitatively by nuclear magnetic resonance (NMR), thermogravimetric analysis (TGA), and time-of-flight secondary ion mass spectrometry (ToF-SIMS) and qualitatively by studying ζ-potential, protein adsorption, diffusion, cellular interactions, and blood circulation half-life. We found that NMR and ToF-SIMS are complementary methods, while TGA is less suitable to quantitate PEG on polymeric NPs. It was found that longer PEG increases both blood circulation time and diffusion of NPs in collagen gels.


Assuntos
Nanopartículas/química , Polímeros/química , Embucrilato/química , Espectroscopia de Ressonância Magnética , Metacrilatos/química , Nanomedicina/métodos , Tensoativos/química , Termogravimetria
20.
Contrast Media Mol Imaging ; 10(5): 356-66, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25930237

RESUMO

Microbubbles (MBs) are routinely used as contrast agents for ultrasound imaging. The use of ultrasound in combination with MBs has also attracted attention as a method to enhance drug delivery. We have developed a technology platform incorporating multiple functionalities, including imaging and therapy in a single system consisting of MBs stabilized by polyethylene glycol (PEG)-coated polymeric nanoparticles (NPs). The NPs, containing lipophilic drugs and/or contrast agents, are composed of the widely used poly(butyl cyanoacrylate) (PBCA) polymer and prepared in a single step. MBs stabilized by these NPs are subsequently prepared by self-assembly of NPs at the MB air-liquid interface. Here we show that these MBs can act as contrast agents for conventional ultrasound imaging. Successful encapsulation of iron oxide NPs inside the PBCA NPs is demonstrated, potentially enabling the NP-MBs to be used as magnetic resonance imaging (MRI) and/or molecular ultrasound imaging contrast agents. By precise tuning of the applied ultrasound pulse, the MBs burst and the NPs constituting the shell are released. This could result in increased local deposit of NPs into target tissue, providing improved therapy and imaging contrast compared with freely distributed NPs.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Embucrilato/química , Microbolhas , Imagem Multimodal/métodos , Nanopartículas/química , Polímeros/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA