Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Angew Chem Int Ed Engl ; 62(23): e202302525, 2023 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-36930411

RESUMO

Carbon monoxide (CO) is an endogenous signaling molecule with broad therapeutic effects. Here, a multifunctional X-ray-triggered carbon monoxide (CO) and manganese dioxide (MnO2 ) generation nanoplatform based on metal carbonyl and scintillating nanoparticles (SCNPs) is reported. Attributed to the radioluminescent characteristic of SCNPs, UV-responsive Mn2 (CO)10 is not only indirectly activated to release CO by X-ray but can also be degraded into MnO2 . A high dose of CO can be used as a glycolytic inhibitor for tumor suppression; it will also sensitize tumor cells to radiotherapy. Meanwhile MnO2 , as the photolytic byproduct of Mn2 (CO)10 , has both glutathione (GSH) depletion and Fenton-like Mn2+ delivery properties to produce highly toxic hydroxyl radical (⋅OH) in tumors. Thus, this strategy can realize X-ray-activated CO release, GSH depletion, and ⋅OH generation for cascade cancer radiosensitization. Furthermore, X-ray-activated Mn2+ in vivo demonstrates an MRI contrast effect, making it a potential theranostic nanoplatform.


Assuntos
Nanopartículas , Neoplasias , Humanos , Compostos de Manganês/farmacologia , Compostos de Manganês/uso terapêutico , Óxidos/farmacologia , Monóxido de Carbono/farmacologia , Monóxido de Carbono/uso terapêutico , Raios X , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Linhagem Celular Tumoral , Glutationa/metabolismo , Peróxido de Hidrogênio/uso terapêutico
2.
ACS Nano ; 14(8): 10001-10017, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32658453

RESUMO

Development of an efficient nanoradiosensitization system that enhances the radiation doses in cancer cells to sensitize radiotherapy (RT) while sparing normal tissues is highly desirable. Here, we construct a tumor microenvironment (TME)-responsive disassembled small-on-large molybdenum disulfide/hafnium dioxide (MoS2/HfO2) dextran (M/H-D) nanoradiosensitizer. The M/H-D can degrade and release the HfO2 nanoparticles (NPs) in TME to enhance tumor penetration of the HfO2 NPs upon near-infrared (NIR) exposure, which can solve the bottleneck of insufficient internalization of the HfO2 NPs. Simultaneously, the NIR photothermal therapy increased peroxidase-like catalytic efficiency of the M/H-D nanoradiosensitizer in TME, which selectively catalyzed intratumorally overexpressed H2O2 into highly oxidized hydroxyl radicals (·OH). The heat induced by PTT also relieved the intratumoral hypoxia to sensitize RT. Consequently, this TME-responsive precise nanoradiosensitization achieved improved irradiation effectiveness, potent oxygenation in tumor, and efficient suppression to tumor, which can be real-time monitored by computed tomography and photoacoustic imaging.


Assuntos
Nanopartículas , Neoplasias , Háfnio , Humanos , Peróxido de Hidrogênio , Neoplasias/terapia , Fototerapia , Microambiente Tumoral
3.
Nanoscale ; 11(15): 7157-7165, 2019 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-30919835

RESUMO

The development of a new multifunctional nanomedicine capable of enhancing radiosensitization by photo-induced hyperthermia for the inhibition of cancer growth and metastasis is highly required for efficient treatment of cancer cells. Compared to the first near-infrared (NIR) window, the second NIR window light could provide a maximum penetration depth as well as minimizing autofluorescence due to its low scattering and energy absorption. Here, we report a new versatile theranostic agent based on ternary Cu3BiSe3 nanoparticles (NPs) modified by poly(vinylpyrollidone) (PVP-Cu3BiSe3). Benefiting from their preferable X-ray attenuation ability and strong NIR absorbance in the second NIR biological window, PVP-Cu3BiSe3 NPs can not only deposit more radiation doses to destroy the cancer cells, but also conduct the optical energy into hyperthermia for thermal eradication of tumor tissues and the improvement of the tumor oxygenation to overcome the hypoxia-associated radio-resistance of tumors. According to both in vitro and in vivo results, exposure to an X-ray plus 1064 nm laser completely kills cancer cells and even inhibits tumor metastasis, displaying no warning signs of a relapse. On the other hand, PVP-Cu3BiSe3 NPs can be used as a multi-model imaging agent for X-ray computer tomography (CT) and photoacoustic tomography (PAT) imaging. These demonstrate the potential of PVP-Cu3BiSe3 NPs in multimodal imaging-guided synergetic radiophotothermal therapy of deep-seated tumors and effective inhibition of their metastasis.


Assuntos
Hipertermia Induzida , Raios Infravermelhos , Nanopartículas Metálicas , Neoplasias Experimentais , Técnicas Fotoacústicas , Fototerapia , Radiossensibilizantes , Tomografia Computadorizada por Raios X , Animais , Células HeLa , Humanos , Masculino , Nanopartículas Metálicas/química , Nanopartículas Metálicas/uso terapêutico , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Experimentais/diagnóstico por imagem , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , Radiossensibilizantes/química , Radiossensibilizantes/farmacologia
4.
Adv Mater ; 31(9): e1800662, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30039878

RESUMO

The extensive research of graphene and its derivatives in biomedical applications during the past few years has witnessed its significance in the field of nanomedicine. Starting from simple drug delivery systems, the application of graphene and its derivatives has been extended to a versatile platform of multiple therapeutic modalities, including photothermal therapy, photodynamic therapy, magnetic hyperthermia therapy, and sonodynamic therapy. In addition to monotherapy, graphene-based materials are widely applied in combined therapies for enhanced anticancer activity and reduced side effects. In particular, graphene-based materials are often designed and fabricated as "smart" platforms for stimuli-responsive nanocarriers, whose therapeutic effects can be activated by the tumor microenvironment, such as acidic pH and elevated glutathione (termed as "endogenous stimuli"), or light, magnetic, or ultrasonic stimuli (termed as "exogenous stimuli"). Herein, the recent advances of smart graphene platforms for combined therapy applications are presented, starting with the principle for the design of graphene-based smart platforms in combined therapy applications. Next, recent advances of combined therapies contributed by graphene-based materials, including chemotherapy-based, photothermal-therapy-based, and ultrasound-therapy-based synergistic therapy, are outlined. In addition, current challenges and future prospects regarding this promising field are discussed.


Assuntos
Terapia Combinada/métodos , Portadores de Fármacos/química , Grafite/química , Nanoestruturas/química , Neoplasias/terapia , Tratamento Farmacológico/métodos , Humanos , Hipertermia Induzida/métodos , Campos Magnéticos , Nanomedicina/métodos , Fototerapia/métodos , Terapia por Ultrassom/métodos
5.
Small ; 14(45): e1802290, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30307703

RESUMO

The rising dangers of bacterial infections have created an urgent need for the development of a new generation of antibacterial nanoagents and therapeutics. A new near-infrared 808 nm laser-mediated nitric oxide (NO)-releasing nanovehicle (MoS2 -BNN6) is reported through the simple assembly of α-cyclodextrin-modified MoS2 nanosheets with a heat-sensitive NO donor N,N'-di-sec-butyl-N,N'-dinitroso-1,4-phenylenediamine (BNN6) for the rapid and effective treatment of three typical Gram-negative and Gram-positive bacteria (ampicillin-resistant Escherichia coli, heat-resistant Escherichia faecalis, and pathogen Staphylococcus aureus). This MoS2 -BNN6 nanovehicle has good biocompatibility and can be captured by bacteria to increase opportunities of NO diffusion to the bacterial surface. Once stimulated by 808 nm laser irradiation, the MoS2 -BNN6 nanovehicle not only exhibits photothermal therapy (PTT) efficacy but also can precisely control NO release, generating oxidative/nitrosative stress. The temperature-enhanced catalytic function of MoS2 induced by 808 nm laser irradiation simultaneously accelerates the oxidation of glutathione. This acceleration disrupts the balance of antioxidants, ultimately resulting in significant DNA damage to the bacteria. Within 10 min, the MoS2 -BNN6 with enhanced PTT/NO synergetic antibacterial function achieves >97.2% inactivation of bacteria. The safe synergetic therapy strategy can also effectively repair wounds through the formation of collagen fibers and elimination of inflammation during tissue reconstruction.


Assuntos
Antibacterianos/química , Dissulfetos/química , Molibdênio/química , Óxido Nítrico/química , Antibacterianos/farmacologia , Dano ao DNA/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Luz , Testes de Sensibilidade Microbiana , Doadores de Óxido Nítrico/química , Staphylococcus aureus/efeitos dos fármacos
6.
Adv Healthc Mater ; 7(22): e1800830, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30240165

RESUMO

Although various types of photothermal agents are developed for photothermal cancer therapy, relatively few photothermal agents exhibit high tumor inhibition rate under relatively mild conditions. Herein, a multifunctional Bi2 S3 -Tween 20 nanoplatform loaded with PI3K inhibitor LY294002 is designed as a novel photothermal agent for inhibitor and photothermal synergistic therapy of tumors under mild photothermal therapy conditions. The LY294002 of PI3K inhibitor, after being loaded by Bi2 S3 -Tween 20 nanodots, exhibits greatly increased drug utilization and reduced side effects on normal tissues. In vivo, Bi2 S3 -Tween 20@LY294002 upon near-infrared 808 nm laser irradiation shows potent antitumor activity under relatively mild conditions (power density: 0.6 W cm-2 ). Moreover, the mechanism studies also demonstrate that Bi2 S3 -Tween 20@LY294002 potently kills LoVo cancer cells under low-power near-infrared light irradiation, by downregulating the expression of heat shock protein 70 (HSP70) so as to increase the sensitivity of tumor cell hyperthermia and activating BAX/BAK-regulated mitochondrial apoptosis pathway. The results demonstrate that the newly synthesized multifunctional nanoplatform paves a new avenue for accurate therapy of photothermal-resistant cancer.


Assuntos
Bismuto/química , Cromonas/química , Morfolinas/química , Nanopartículas/química , Polissorbatos/química , Sulfetos/química , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cromonas/farmacologia , Cromonas/uso terapêutico , Regulação para Baixo/efeitos dos fármacos , Proteínas de Choque Térmico HSP70/metabolismo , Hemólise/efeitos dos fármacos , Humanos , Hipertermia Induzida , Raios Infravermelhos , Camundongos , Camundongos Nus , Morfolinas/farmacologia , Morfolinas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/terapia , Inibidores de Fosfoinositídeo-3 Quinase , Fototerapia , Proteína X Associada a bcl-2/metabolismo
7.
Nanoscale ; 10(3): 1517-1531, 2018 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-29303196

RESUMO

Near-infrared (NIR) laser induced phototherapy has been considered as a noninvasive option for cancer therapy. Herein, we report plasmonic PEGylated molybdenum oxide nanoparticles (PEG-MoOx NPs) that were synthesized by using a facile hydrothermal method. The PEG-MoOx NPs exhibit broad absorption at the NIR biological window and remarkable photothermal conversion ability in the first (808 nm) and the second (1064 nm) windows. Moreover, the biocompatible PEG-MoOx NPs exhibit effective cellular uptake and could be eliminated gradually from the liver and spleen in mice. Studies on the therapeutic effects of these NPs under 808 and 1064 nm exposures with mild hyperthermia are conducted. According to the result, exposure to 1064 nm irradiation can not only effectively convert light into heat but also sensitize the formation of reactive oxygen species (ROS), which exert dramatic cancer cell death and suppression in vivo due to the synergic effect of photothermal therapy (PTT) and photodynamic therapy (PDT). In marked contrast, 808 nm irradiation can only execute limited PTT to cancer cells, showing a relatively low inhibition rate in vitro and in vivo. This biodegradable MoOx nanoplatform with synergetic PTT and PDT functionalities upon 1064 nm irradiation provided emerging opportunities for the phototherapy of cancer in nanomedicine.


Assuntos
Hipertermia Induzida , Molibdênio , Nanopartículas , Neoplasias Experimentais/terapia , Fotoquimioterapia , Fototerapia , Animais , Células HeLa , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Óxidos
8.
ACS Appl Mater Interfaces ; 9(16): 14281-14291, 2017 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-28381089

RESUMO

Low water solubility, extensive metabolism, and drug resistance are the existing unavoidable disadvantages of the insoluble drug curcumin in biomedical applications. Herein, we employed d-α-tocopherol polyethylene glycol 1000 succinate (TPGS)-functionalized near-infrared (NIR)-triggered photothermal mesoporous nanocarriers with bamboo charcoal nanoparticles (TPGS-BCNPs) to load and deliver curcumin for improving its bioavailability. This system could considerably increase the accumulation of curcumin in cancer cells for enhanced curcumin bioavailability via simultaneously promoting the cellular internalization of the as-synthesized composite (TPGS-BCNPs@curcumin) by the size effect of NPs and considerably triggering controlled curcumin release from TPGS-BCNPs@curcumin by NIR stimulation and reducing efflux of curcumin by the P-glycoprotein (P-gp) inhibition of TPGS, so as to enhance the therapeutic effect of curcumin and realize a better chemo-photothermal synergetic therapy in vitro and in vivo. Besides cancer therapy, studies indicated that curcumin and some carbon materials could be used as radical scavengers that play an important role in the radioprotection of normal cells. Hence, we also investigated the free-radical-scavenging ability of the TPGS-BCNPs@curcumin composite in vitro to preliminarily evaluate its radioprotection ability for healthy tissues. Therefore, our work provides a multifunctional delivery system for curcumin bioavailability enhancement, chemo-photothermal synergetic therapy of cancer, and radioprotection of healthy tissues.


Assuntos
Nanopartículas , Disponibilidade Biológica , Linhagem Celular Tumoral , Carvão Vegetal , Curcumina , Humanos , Neoplasias
9.
Adv Healthc Mater ; 5(21): 2776-2787, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27717238

RESUMO

The combination of photothermal therapy (PTT) with gene therapy (GT) to improve PTT efficiency and thus eliminate cancer cells under mild hyperthermia is highly needed. Herein, multifunctional WS2 @poly(ethylene imine) (WS2 @PEI) nanoplatform has been designed and constructed for gene-photothermal synergistic therapy of tumors at mild condition. After a surface modification of WS2 with a positively charged PEI, the as-prepared WS2 @PEI nanoplatform can not only act as an efficient survivin-siRNA carrier for GT but also exhibit remarkable near-infrared (NIR) photothermal effects for PTT. On the one hand, the photothermal effects induced by WS2 @PEI upon NIR irradiation can enhance the cellular uptake owing to the increase of the cell membrane permeability, which leads to the remarkable enhancement of silencing efficiency of survivin. On the other hand, the silencing of survivin can increase the apoptosis as well as reduce the heat resistance of cancer cells by downregulating the heat shock protein 70 expressions, which greatly enhance the sensitivity of cancer cells to PTT. As a result, compared to PTT or GT treatment alone, WS2 @PEI mediated synergistic GT/PTT therapy remarkably enhances in vitro cancer cell damage and in vivo tumor elimination.


Assuntos
Iminas/química , Nanopartículas/administração & dosagem , Nanopartículas/química , Neoplasias/terapia , Polietilenos/química , Animais , Apoptose/genética , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/genética , Regulação para Baixo/genética , Terapia Genética/métodos , Proteínas de Choque Térmico HSP70/genética , Células HeLa , Humanos , Hipertermia Induzida/métodos , Proteínas Inibidoras de Apoptose/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fototerapia/métodos , RNA Interferente Pequeno/genética
10.
Adv Healthc Mater ; 5(13): 1627-37, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27276383

RESUMO

Near-infrared-(NIR)-light-triggered photothermal nanocarriers have attracted much attention for the construction of more smart and effective therapeutic platforms in nanomedicine. Here, a multifunctional drug carrier based on a low cost, natural, and biocompatible material, bamboo charcoal nanoparticles (BCNPs), which are prepared by the pyrolysis of bamboo followed by physical grinding and ultrasonication is reported. The as-prepared BCNPs with porous structure possess not only large surface areas for drug loading but also an efficient photothermal effect, making them become both a suitable drug carrier and photothermal agent for cancer therapy. After loading doxorubicin (DOX) into the BCNPs, the resulting DOX-BCNPs enhance drug potency and more importantly can overcome the drug resistance of DOX in a MCF-7 cancer cell model by significantly increasing cellular uptake while remarkably decreasing drug efflux. The in vivo synergistic effect of combining chemotherapy and photothermal therapy in this drug delivery system is also demonstrated. In addition, the BCNPs enhance optoacoustic imaging contrast due to their high NIR absorbance. Collectively, it is demonstrated that the BCNP drug delivery system constitutes a promising and effective nanocarrier for simultaneous bioimaging and chemo-photothermal synergistic therapy of cancer.


Assuntos
Carvão Vegetal , Doxorrubicina , Portadores de Fármacos , Hipertermia Induzida/métodos , Nanopartículas , Neoplasias , Fototerapia/métodos , Animais , Carvão Vegetal/química , Carvão Vegetal/farmacocinética , Carvão Vegetal/farmacologia , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Feminino , Humanos , Células MCF-7 , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/terapia , Poaceae/química , Porosidade , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nanoscale ; 8(22): 11531-42, 2016 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-27203525

RESUMO

Recently, a combination of chemotherapy with photothermal therapy (PTT) has received great attention for the construction of a near infrared (NIR)-controlled drug-delivery system for synergistic treatment of cancer, ultimately resulting in the enhancement of the therapeutic efficacy of anticancer drugs. Here, we developed a novel system for synergistic cancer therapy based on bismuth sulfide (Bi2S3) nanoparticle-decorated graphene functionalized with polyvinylpyrrolidone (PVP) (named PVP-rGO/Bi2S3). The as-prepared PVP-rGO/Bi2S3 nanocomposite has a high storage capacity for anticancer drugs (∼500% for doxorubicin (DOX)) and simultaneously has perfect photothermal conversion efficiency in the NIR region. The results of the in vitro accumulative drug release test manifests that the PVP-rGO/Bi2S3 nanocomposite could be applied as a dual pH- and NIR-responsive nanotherapeutic carrier for the controlled release of DOX from DOX-loaded PVP-rGO/Bi2S3 (PVP-rGO/Bi2S3@DOX). Moreover, the treatment of both cancer cells (including Hela, MCF-7, HepG2 and BEL-7402 cells) and BEL-7402 tumor-bearing mice with the PVP-rGO/Bi2S3@DOX complex followed by NIR laser irradiation produces significantly greater inhibition of cancer cell growth than the treatment with NIR irradiation alone or DOX alone, exhibiting a synergistic antitumor effect. Furthermore, due to the obvious NIR and X-ray absorption ability, the PVP-rGO/Bi2S3 nanocomposite could be employed as a dual-modal contrast agent for both photoacoustic tomography and X-ray computed tomography imaging. In addition to the good biocompatibility, the PVP-rGO/Bi2S3 nanocomposite paves a potential way for the fabrication of theranostic agents for dual-modal imaging-guided chemo-photothermal combined cancer therapy.


Assuntos
Nanocompostos/química , Neoplasias Experimentais/tratamento farmacológico , Fototerapia , Povidona/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Humanos , Raios Infravermelhos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
12.
ACS Nano ; 10(4): 4587-98, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27014806

RESUMO

A key challenge for the use of inorganic nanomedicines in clinical applications is their long-term accumulation in internal organs, which raises the common concern of the risk of adverse effects and inflammatory responses. It is thus necessary to rationally design inorganic nanomaterials with proper accumulation and clearance mechanism in vivo. Herein, we prepared ultrasmall Cu3BiS3 nanodots (NDs) as a single-phased ternary bimetal sulfide for photothermal cancer therapy guided by multispectral optoacoustic tomography (MSOT) and X-ray computed tomography (CT) due to bismuth's excellent X-ray attenuation coefficient. We then monitored and investigated their absorption, distribution, metabolism, and excretion. We also used CT imaging to demonstrate that Cu3BiS3 NDs can be quickly removed through renal clearance, which may be related to their small size, rapid chemical transformation, and degradation in an acidic lysosomal environment as characterized by synchrotron radiation-based X-ray absorption near-edge structure spectroscopy. These results reveal that Cu3BiS3 NDs act as a simple but powerful "theranostic" nanoplatform for MSOT/CT imaging-guided tumor ablation with excellent metabolism and rapid clearance that will improve safety for clinical applications in the future.


Assuntos
Antineoplásicos/farmacocinética , Bismuto/química , Cobre/química , Rim/metabolismo , Nanopartículas/química , Neoplasias/diagnóstico , Neoplasias/terapia , Sulfetos/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Sobrevivência Celular/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos , Tamanho da Partícula , Técnicas Fotoacústicas , Fototerapia/métodos , Reabsorção Renal , Nanomedicina Teranóstica , Distribuição Tecidual , Tomografia Computadorizada por Raios X
13.
Biomaterials ; 76: 11-24, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26517561

RESUMO

Engineering design of plasmonic nanomaterials as on-demand theranostic nanoagents with imaging, drug carrier, and photothermal therapy (PTT) functions have profound impact on treatment of cancer. Here, a facile 'one-pot' template-free hydrothermal route was firstly developed for synthesis of plasmonic oxygen deficiency molybdenum oxide hollow nanospheres functionalized by poly(ethylene glycol) (PEG-MoO(3-x) HNSs). The as-prepared PEG-MoO(3-x) HNSs not only have good biocompatibility but also exhibit obvious localized surface plasmon resonance (LSPR) absorption in the near-infrared (NIR) region. Especially, due to its intrinsic mesoporous properties and effective photothermal conversion efficiency upon 808-nm NIR laser irradiation, the PEG-MoO(3-x) HNSs can be applied as a pH/NIR laser dual-responsive camptothecin (CPT) drug delivery nanoplatform for chemotherapy as well as PTT to cancer cells. A remarkably improved synergistic therapeutic effect to pancreatic (PANC-1) tumor-bearing mice was obtained compared to the result of chemotherapy or PTT alone. Apart from its application for drug delivery, the PEG-MoO(3-x) HNSs can also be employed as an effective contrast nanoagent for photoacoustic (PAT) imaging because of its high NIR absorption, making it promising as a theranostic nanoagent for PAT imaging-guided chemo-photothermal combinational cancer therapy in the nanomedicine field.


Assuntos
Antineoplásicos/uso terapêutico , Hipertermia Induzida , Nanosferas , Neoplasias/terapia , Óxidos/síntese química , Fototerapia , Animais , Terapia Combinada , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Molibdênio , Polietilenoglicóis/química
14.
Nanoscale ; 7(29): 12581-91, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26145146

RESUMO

Non-invasive and real-time imaging of the gastrointestinal (GI) tract is particularly desirable for research and clinical studies of patients with symptoms arising from gastrointestinal diseases. Here, we designed and fabricated silica-coated bismuth sulfide nanorods (Bi2S3@SiO2 NRs) for a non-invasive spatial-temporally imaging of the GI tract. The Bi2S3 NRs were synthesized by a facile solvothermal method and then coated with a SiO2 layer to improve their biocompatibility and stability in the harsh environments of the GI tract, such as the stomach and the small intestine. Due to their strong X-ray- and near infrared-absorption abilities, we demonstrate that, following oral administration in mice, the Bi2S3@SiO2 NRs can be used as a dual-modal contrast agent for the real-time and non-invasive visualization of NRs distribution and the GI tract via both X-ray computed tomography (CT) and photoacoustic tomography (PAT) techniques. Importantly, integration of PAT with CT provides complementary information on anatomical details with high spatial resolution. In addition, we use Caenorhabditis Elegans (C. Elegans) as a simple model organism to investigate the biological response of Bi2S3@SiO2 NRs by oral administration. The results indicate that these NRs can pass through the GI tract of C. Elegans without inducing notable toxicological effects. The above results suggest that Bi2S3@SiO2 NRs pave an alternative way for the fabrication of multi-modal contrast agents which integrate CT and PAT modalities for a direct and non-invasive visualization of the GI tract with low toxicity.


Assuntos
Meios de Contraste/química , Trato Gastrointestinal/patologia , Nanotubos/química , Técnicas Fotoacústicas , Tomografia Computadorizada por Raios X , Administração Oral , Animais , Bismuto/química , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Feminino , Trato Gastrointestinal/diagnóstico por imagem , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia de Fluorescência , Nanotubos/toxicidade , Dióxido de Silício/química , Sulfetos/química
15.
Theranostics ; 5(9): 931-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26155310

RESUMO

The ability to selectively destroy cancer cells while sparing normal tissue is highly desirable during the cancer therapy. Here, magnetic targeted photothermal therapy was demonstrated by the integration of MoS2 (MS) flakes and Fe3O4 (IO) nanoparticles (NPs), where MoS2 converted near-infrared (NIR) light into heat and Fe3O4 NPs served as target moiety directed by external magnetic field to tumor site. The MoS2/Fe3O4 composite (MSIOs) functionalized by biocompatible polyethylene glycol (PEG) were prepared by a simple two-step hydrothermal method. And the as-obtained MSIOs exhibit high stability in bio-fluids and low toxicity in vitro and in vivo. Specifically, the MSIOs can be applied as a dual-modal probe for T2-weighted magnetic resonance (MR) and photoacoustic tomography (PAT) imaging due to their superparamagnetic property and strong NIR absorption. Furthermore, we demonstrate an effective result for magnetically targeted photothermal ablation of cancer. All these results show a great potential for localized photothermal ablation of cancer spatially/timely guided by the magnetic field and indicated the promise of the multifunctional MSIOs for applications in cancer theranostics.


Assuntos
Dissulfetos/farmacocinética , Sistemas de Liberação de Medicamentos/métodos , Compostos Férricos/farmacocinética , Hipertermia Induzida/métodos , Imageamento por Ressonância Magnética/métodos , Molibdênio/farmacocinética , Neoplasias/terapia , Técnicas Fotoacústicas/métodos , Animais , Modelos Animais de Doenças , Dissulfetos/administração & dosagem , Compostos Férricos/administração & dosagem , Células HeLa , Células Hep G2 , Humanos , Raios Infravermelhos , Masculino , Camundongos Endogâmicos BALB C , Molibdênio/administração & dosagem , Nanopartículas/administração & dosagem , Nanomedicina Teranóstica/métodos , Resultado do Tratamento
16.
ACS Nano ; 9(1): 696-707, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25561009

RESUMO

Here, we present a precision cancer nanomedicine based on Bi(2)S(3) nanorods (NRs) designed specifically for multispectral optoacoustic tomography (MSOT)/X-ray computed tomography (CT)-guided photothermal therapy (PTT). The as-prepared Bi(2)S(3) NRs possess ideal photothermal effect and contrast enhancement in MSOT/CT bimodal imaging. These features make them simultaneously act as "satellite" and "precision targeted weapon" for the visual guide to destruction of tumors in vivo, realizing effective tumor destruction and metastasis inhibition after intravenous injection. In addition, toxicity screening confirms that Bi(2)S(3) NRs have well biocompatibility. This triple-modality-nanoparticle approach enables simultaneously precise cancer therapy and therapeutic monitoring.


Assuntos
Bismuto/química , Neoplasias Mamárias Experimentais/diagnóstico , Neoplasias Mamárias Experimentais/terapia , Imagem Multimodal , Nanotubos , Fototerapia , Sulfetos/química , Nanomedicina Teranóstica/métodos , Animais , Linhagem Celular Tumoral , Masculino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Técnicas Fotoacústicas , Tomografia Computadorizada por Raios X
17.
Nanoscale ; 6(17): 10394-403, 2014 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-25047651

RESUMO

We have developed a simple and efficient strategy to fabricate WS2 nanosheets with low toxicity and good water solubility via a liquid exfoliation method by using H2SO4 intercalation and ultrasonication. The as-prepared WS2 nanosheets were employed not only as an NIR absorbing agent for photothermal therapy (PTT) but also as a photosensitizer (PS) carrier for photodynamic therapy (PDT) due to their sheet like structure that offers large surface area to load PS molecules. Moreover, singlet-oxygen generation of the PSs-WS2 complex could be finely controlled by NIR irradiation that could manipulate the PSs release behavior from WS2 nanosheets. The synergistic anti-tumor effect of WS2 nanosheets mediated PDT-PTT was also evaluated carefully and the results clearly showed that the efficacy of combined PDT-PTT treatment of cancer cells is significantly higher than those of PDT-only and PTT-only treatment, indicating enhanced efficiency of the combined therapeutic system. In addition, the WS2 could be used as a computed tomography (CT) contrast agent for bio-imaging since W atoms have strong X-ray attenuation ability, making them a multifunctional theranostic platform for simultaneous imaging-guided diagnosis and therapy.


Assuntos
Hipertermia Induzida/métodos , Nanopartículas Metálicas/administração & dosagem , Azul de Metileno/administração & dosagem , Neoplasias/terapia , Fotoquimioterapia/métodos , Compostos de Tungstênio/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada/métodos , Células HeLa , Humanos , Nanopartículas Metálicas/química , Azul de Metileno/química , Nanocápsulas/administração & dosagem , Nanocápsulas/química , Nanocápsulas/ultraestrutura , Tamanho da Partícula , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/química , Sulfetos/química , Resultado do Tratamento , Compostos de Tungstênio/química
18.
Small ; 10(20): 4160-70, 2014 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-24979184

RESUMO

Light-triggered drug delivery based on near-infrared (NIR)-mediated photothermal nanocarriers has received tremendous attention for the construction of cooperative therapeutic systems in nanomedicine. Herein, a new paradigm of light-responsive drug carrier that doubles as a photothermal agent is reported based on the NIR light-absorber, Rb(x) WO3 (rubidium tungsten bronze, Rb-TB) nanorods. With doxorubicin (DOX) payload, the DOX-loaded Rb-TB composite (Rb-TB-DOX) simultaneously provides a burst-like drug release and intense heating effect upon 808-nm NIR light exposure. MTT assays show the photothermally enhanced antitumor activity of Rb-TB-DOX to the MCF-7 cancer cells. Most remarkably, Rb-TB-DOX combined with NIR irradiation also shows dramatically enhanced chemotherapeutic effect to DOX-resistant MCF-7 cells compared with free DOX, demonstrating the enhanced efficacy of combinational chemo-photothermal therapy for potentially overcoming drug resistance in cancer chemotherapy. Furthermore, in vivo study of combined chemo-photothermal therapy is also conducted and realized on pancreatic (Pance-1) tumor-bearing nude mice. Apart from its promise for cancer therapy, the as-prepared Rb-TB can also be employed as a new dual-modal contrast agent for photoacoustic tomography and (PAT) X-ray computed tomography (CT) imaging because of its high NIR optical absorption capability and strong X-ray attenuation ability, respectively. The results presented in the current study suggest promise of the multifunctional Rb(x)WO3 nanorods for applications in cancer theranostics.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Nanotubos , Técnicas Fotoacústicas , Fototerapia , Rubídio/química , Tomografia Computadorizada por Raios X , Tungstênio/química , Humanos , Raios Infravermelhos , Células MCF-7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA