Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 1444, 2024 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-38365897

RESUMO

Transparent ultrasound transducers (TUTs) can seamlessly integrate optical and ultrasound components, but acoustic impedance mismatch prohibits existing TUTs from being practical substitutes for conventional opaque ultrasound transducers. Here, we propose a transparent adhesive based on a silicon dioxide-epoxy composite to fabricate matching and backing layers with acoustic impedances of 7.5 and 4-6 MRayl, respectively. By employing these layers, we develop an ultrasensitive, broadband TUT with 63% bandwidth at a single resonance frequency and high optical transparency ( > 80%), comparable to conventional opaque ultrasound transducers. Our TUT maximises both acoustic power and transfer efficiency with maximal spectrum flatness while minimising ringdowns. This enables high contrast and high-definition dual-modal ultrasound and photoacoustic imaging in live animals and humans. Both modalities reach an imaging depth of > 15 mm, with depth-to-resolution ratios exceeding 500 and 370, respectively. This development sets a new standard for TUTs, advancing the possibilities of sensor fusion.


Assuntos
Técnicas Fotoacústicas , Humanos , Técnicas Fotoacústicas/métodos , Transdutores , Desenho de Equipamento , Ultrassonografia , Impedância Elétrica
2.
Nanoscale ; 16(2): 742-751, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38086680

RESUMO

The approach of using ferroptosis to treat cancer has garnered attention due to its promising potential. However, the effectiveness of this therapy is often limited by the inherent redox system in cancer cells and the presence of ferritin as an iron ion storage molecule. To address this issue, we have designed a polymeric iron oxide nanocomplex loaded with sulfasalazine as a ferritinophagy-assisted ferroptosis inducing agent. The nanocomplex is based on a pH-responsive drug releasing platform that enables improved ferroptosis anti-cancer therapy. The nanocomplex was synthesized using polymerized phenylboronic acid decorated with iron oxide and further loaded with sulfasalazine by interacting with polymerized phenylboronic acid. Upon entering cancer cells, the nanocomplex releases sulfasalazine at the lysosomal acidic pH, which causes the complex to degrade into the labile iron ion (Fe2+). This process inhibits the production of GSH and reproduces the labile iron ion by degrading ferritin. As a result, an excess iron ion pool is formed, and the facilitated Fenton reaction induces an improved ferroptosis anti-cancer effect. Moreover, our research has demonstrated that the nanocomplex effectively regresses tumors, thereby representing significant inhibition of tumor growth using in vivo models. We believe that this ferritinophagy-assisted ferroptosis strategy using the nanocomplex provides a promising solution for iron-based anti-cancer therapy.


Assuntos
Ferroptose , Sulfassalazina , Sulfassalazina/farmacologia , Ferro , Ferritinas
3.
Biosens Bioelectron ; 216: 114612, 2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-35952434

RESUMO

Theranostic, which integrates the diagnosis and tumor treatment in tandem, is an emerging strategy in cancer treatment. Here, we report a novel and unique theranostic nanoparticle, HBCP NP, based on hexa-BODIPY cyclophosphazene (HBCP). Due to the unique bulky molecular structure of HBCP, this nanoparticle can simultaneously perform near-infrared (NIR) fluorescence imaging and photoacoustic imaging (PAI). Interestingly, since reactive oxygen species (ROS) generation of HBCP NPs is completely inhibited, 'safe' fluorescence imaging is possible without the risk of cell damage even under laser irradiation. Finally, NIR fluorescence imaging and PAI in 4T1 tumor-bearing mice demonstrated selective accumulation of HBCP NPs at tumor sites. In addition, HBCP NPs exhibited excellent photothermal effects under high-power laser irradiation, achieving effective tumor growth inhibition.


Assuntos
Técnicas Biossensoriais , Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Animais , Compostos de Boro , Linhagem Celular Tumoral , Hexosaminidase A , Camundongos , Nanopartículas/química , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Imagem Óptica , Técnicas Fotoacústicas/métodos , Espécies Reativas de Oxigênio , Nanomedicina Teranóstica/métodos
4.
Theranostics ; 12(3): 1247-1266, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154485

RESUMO

Sonodynamic therapy has shown promise as an effective alternative to conventional photodynamic therapy owing to its ability to treat deep-seated tumors. However, the development of stimuli-responsive sonosensitizers with high biocompatibility faces a significant challenge. Methods: In this study, we developed dual stimuli-responsive sonosensitizers with desirable biosafety using extracellular vesicles (EVs), a class of naturally occurring nanoparticles. Indocyanine green (ICG), which functions as both a sonosensitizer and photoacoustic (PA) imaging agent, was loaded into EVs, together with paclitaxel (PTX) and sodium bicarbonate (SBC), to achieve pH-responsive PA imaging-guided chemo-sonodynamic combination therapy. Results: The EVs significantly improved the cellular uptake of ICG, thus triggering enhanced sonodynamic effects in breast cancer cells. SBC-, ICG-, and PTX-loaded EV [SBC-EV(ICG/PTX)] efficiently released the PTX in response to acidic pH in the endo/lysosomes because CO2 bubbles generated from the SBC caused the EV membranes to burst. The drug release was further facilitated by ultrasound (US) treatment, demonstrating dual pH/US-responsive drug release. The ICG- and PTX-loaded EVs exhibited efficient anticancer activity against breast tumor cells owing to the combination of chemo-sonodynamic therapy. High-resolution PA imaging visualized the preferential tumor accumulation of SBC-EV(ICG/PTX) in tumor-bearing mice. Notably, a single intravenous injection of SBC-EV(ICG/PTX) with US irradiation significantly suppressed tumor growth in mice without systemic toxicity. Conclusions: Our findings demonstrate that dual stimuli-responsive SBC-EV(ICG/PTX) are promising sonotheranostic nanoplatforms for safe and efficient chemo-sonodynamic combination cancer therapy and photoacoustic imaging.


Assuntos
Vesículas Extracelulares , Nanopartículas , Neoplasias , Técnicas Fotoacústicas , Animais , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Humanos , Verde de Indocianina , Camundongos , Paclitaxel/farmacologia
5.
J Control Release ; 343: 78-88, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35065158

RESUMO

Tumor-associated macrophages (TAMs), which dampen the therapeutic efficacy of cancer immunotherapy, are the key players in the immunosuppressive tumor microenvironment (TME). Therefore, reprogramming TAMs into tumoricidal M1 macrophages possesses considerable potential as a novel immunotherapy. However, the low bioavailability of polarization agents and limited accumulation of TAMs restrict their anti-tumor efficacy. In this study, we developed a polymer-based hypoxia-responsive nanocomplex to target TAMs in hypoxia for enhanced cancer immunotherapy. We synthesized a hypoxia-cleavable polymer poly(ethylene glycol)-azo-poly(l-lysine) (PEG-azo-PLL) and formulated a nanocomplex by simple mixing PEG-azo-PLL and poly(I:C). By mimicking in vitro hypoxia conditions, PEG-azo-PLL/poly(I:C) complexes could transform the physicochemical properties to enhance the delivery efficiency of poly(I:C) to tumor hypoxia, where M2-like TAMs are accumulated. Furthermore, PEG-azo-PLL/poly(I:C) could successfully reduce the population of M2-like TAMs in hypoxic tumors and promoted infiltration of CD8+ T cells in vivo, resulting in the favorable conversion of immunosuppressive TME. Finally, PEG-azo-PLL/poly(I:C) could elicit a significant in vivo anti-tumor effect in B16F10-bearing mice in addition to a prolonged survival time, demonstrating that the hypoxia-responsive nanocomplex PEG-azo-PLL/poly(I:C) is a promising approach for TAM reprogramming immunotherapy for solid tumors.


Assuntos
Neoplasias , Microambiente Tumoral , Animais , Linfócitos T CD8-Positivos , Hipóxia/terapia , Imunização , Camundongos , Neoplasias/terapia , Polietilenoglicóis/química , Macrófagos Associados a Tumor
6.
Anal Sens ; 2(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37621644

RESUMO

Photoacoustic (PA) imaging relies on the absorption of light by chromophores to generate acoustic waves used to delineate tissue structures and physiology. Here, we demonstrate that Cu(II) efficiently catalyzes the dimerization of diverse near-infrared (NIR) cyanine molecules, including a peptide conjugate. NMR spectroscopy revealed a C-C covalent bond along the heptamethine chains, creating stable molecules under conditions such as a wide range of solvents and pH mediums. Dimerization achieved >90% fluorescence quenching, enhanced photostability, and increased PA signals by a factor of about 4 at equimolar concentrations compared to the monomers. In vivo study with a mouse cancer model revealed that dimerization enhanced tumor retention and PA signal, allowing cancer detection at doses where the monomers are less effective. While the dye dimers highlighted peritumoral blood vessels, the PA signal for dimeric tumor-targeting dye-peptide conjugate, LS301, was diffuse throughout the entire tumor mass. A combination of the ease of synthesis, diversity of molecules that are amenable to Cu(II)-catalyzed dimerization, and the high acoustic wave amplification by these stable dimeric small molecules ushers a new strategy to develop clinically translatable PA molecular amplifiers for the emerging field of molecular photoacoustic imaging.

7.
J Control Release ; 330: 1168-1177, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33212120

RESUMO

Inspired by our previous study, we report a simple yet effective platform for therapeutic antibody delivery. A polymeric phenylboronic acid (pPBA)-antibody nanocomplex was simply formulated by mixing pPBA and antibody, derived by the formation of a pH-responsive phenylboronic ester between the PBA group on pPBA and diol on the inherent glycosylation site of the antibody. We focused on the basic prerequisites for a successful delivery, protection from degradation during the circulation, and release at the target lesion. To evaluate the antibody delivery system, anti-PD-L1, one of the most common antibody therapeutics in immuno-oncology, and mouse colon cancer model with an MC-38 cell line were used. Several in-vitro assays reveal the outstanding protective effect of the nanocomplexes as well as the pH-responsive release of antibodies. Moreover, the anti-PD-L1 nanocomplex exhibited an enhanced circulation as well as a better accumulation in tumor lesions after administration in vivo, which led to a significant antitumor effect in comparison to that of a free antibody. Our nanocomplex platform is a promising antibody delivery system for application in conventional antibody-mediated therapies.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Animais , Ácidos Borônicos/uso terapêutico , Linhagem Celular Tumoral , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico
8.
Adv Healthc Mater ; 10(5): e2000834, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33073497

RESUMO

The tumor microenvironment (TME), which is composed of cancer cells, stromal cells, immune cells, and extracellular matrices, plays an important role in tumor growth and progression. Thus, targeting the TME using a well-designed nano-drug delivery system is emerging as a promising strategy for the treatment of solid tumors. Compared to normal tissues, the TME presents several distinguishable physiological features such as mildly acidic pH, hypoxia, high level of reactive oxygen species, and overexpression of specific enzymes, that are exploited as stimuli to induce specific changes in the nanocarrier structures, and thereby facilitates target-specific delivery of imaging or chemotherapeutic agents for the early diagnosis or effective treatment, respectively. Recently, smart nanocarriers that respond to more than one stimulus in the TME have also been designed to elicit a more desirable spatiotemporally controlled drug release. This review highlights the recent progress in TME-sensitive nanocarriers designed for more efficient tumor therapy and imaging. In particular, the design strategies, challenges, and critical considerations involved in the fabrication of TME-sensitive nanocarriers, along with their in vitro and in vivo evaluations are discussed.


Assuntos
Antineoplásicos , Neoplasias , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Microambiente Tumoral
9.
Adv Healthc Mater ; 9(22): e2001633, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33073526

RESUMO

Nanoparticle (NP)-based drug delivery systems or nanomedicines have broadened the horizon of translational research for decades. Conventional bulk mixing synthesis methods have impeded successful clinical translations of nanomedicines due to the limited ability of the controlled, scalable production with high uniformity. Herein, an on-chip preparation of self-assembled, drug-encapsulated polymeric NPs is presented for their improved uniformity and homogeneity that results in enhanced anti-cancer effect in vitro and in vivo. The NPs are formulated through rapid convective mixing of two aqueous solutions of a hydrophilic polymer and an anti-cancer drug, doxorubicin (DOX), in the swirling microvortex reactor (SMR). Compared to conventional bulk-mixed NPs (BMPs), the microvortex-synthesized NPs (MVPs) exhibit narrower size distributions and better size tunability. It is found that the improved uniformity and homogeneity of the MVPs not only enhance cellular uptake and anti-cancer effect with pH-responsive drug release in vitro, but also result in an improved tumor regression and decreased side effects at off-targeted organs in vivo. The findings demonstrate that uniformly designed NPs with more homogeneous properties can induce a significant enhancement of an anti-cancer effect in vivo. The results show the potential of a high-speed on-chip synthesis as a scalable manufacturing platform for reliable clinical translations of nanomedicines.


Assuntos
Nanopartículas , Neoplasias , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos , Humanos , Concentração de Íons de Hidrogênio , Neoplasias/tratamento farmacológico , Polímeros/uso terapêutico , Resultado do Tratamento
10.
J Control Release ; 327: 627-640, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-32916229

RESUMO

Engineering the membrane of the polymersomes with biologically relevant stimuli-responsive units enables spatial and temporal controlled drug release for effective therapy. Herein, we introduce a new-type of polymersomes featuring reactive oxygen species singlet oxygen (1O2)-labile membrane by employing a versatile stereoregular amphiphilic poly(ethylene glycol)-block-poly(ß-aminoacrylate)-block-poly(ethylene glycol) copolymers, which are synthesized through a facile one pot modular amino-alkynoate click polymerization between secondary amines and activated alkynes. These polymersomes readily co-encapsulate an anticancer drug doxorubicin (DOX) and a near infrared (NIR) photosensitizer IR-780 with hydrophobic characteristics in the membrane, and the resulting polymersomes show efficient uptake by the tumor cells. NIR light irradiation on the tumors, following intraperitoneal injection of the IR-780/DOX co-encapsulated polymersomes, facilitates tumor-specific release of DOX through disassembly of the polymersome nanostructure via 1O2-mediated photocleavage of the membrane. Moreover, IR-780 dye can convert NIR light energy into heat in addition to the generation of 1O2, thus allows to realize both photothermal and photodynamic therapy. Accordingly, the NIR light-mediated on demand chemotherapy, in combination with appreciable phototherapy, of IR-780/DOX co-loaded polymersomes demonstrate an efficient tumor suppression in vivo.


Assuntos
Fotoquimioterapia , Oxigênio Singlete , Linhagem Celular Tumoral , Doxorrubicina , Liberação Controlada de Fármacos , Fototerapia
11.
Biomaterials ; 263: 120402, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32977256

RESUMO

Although graphene oxide (GO) possesses many beneficial functionalities for biomedical usage as itself, modification of GO surface with several polymers or protein is inevitable for in vivo applications; however, such modification limits the degradability of GO due to the steric hindrance. In that context, designing of a surface modified GO carrier that is going to be degraded after its biological function (i.e., drug delivery) is highly desired, especially at complex in vivo level. Herein, we design an unprecedented "catalytic GO nanomedicine" by applying the catalytic DNA, achieving self-degradation of GO in systemic level in the body after the therapy following surface modification. Once the catalytic GO nanomedicines are taken up by mucin1 (MUC1) aptamer-facilitated endocytosis, a photo-switch triggers the release of doxorubicin from the DNA. The single stranded G-quadruplex sequence on the surface of GO forms a quartet structure and becomes DNAzyme by binding with hemin on the GO surface, exhibiting peroxidase effect. Due to the high H2O2 concentration in cancer cells, the catalytic GO nanomedicine generates sufficient amount of strong oxidant, hypochlorous acid (HOCl), inducing GO degradation into small fragments for potential clearance. We demonstrate the potential of our catalytic GO nanomedicine for both therapy and degradation at cellular and complex in vivo environment.


Assuntos
DNA Catalítico , Quadruplex G , Grafite , DNA Catalítico/metabolismo , Hemina , Peróxido de Hidrogênio , Nanomedicina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...