Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Small ; 17(6): e2005728, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33470521

RESUMEN

In order to achieve better antitumor therapeutic efficacy and inhibit tumor metastasis, a multifunctional nanovaccine based on L-arginine (LA)-loaded black mesoporous titania (BMT) is fabricated. In this system, LA is utilized as the exogenous NO supplementation for gas therapy, and BMT is served as acoustic sensitizer for sonodynamic therapy. The ultrasound (US) as the exogenous stimulus can simultaneously trigger BMT and LA to produce singlet oxygen (1 O2 ) and NO gas at tumor sites, respectively. Interestingly, 1 O2 from US-excited BMT can promote the oxidation of LA to produce more NO. The high concentration of 1 O2 and NO in cancer cell can cause intracellular strong oxidative stress level and DNA double-strand breaks to induce cancer cell apoptosis ultimately. The US-triggered BMT@LA "nanovaccine" combining with immune checkpoint inhibitor PD-L1 antibody (αPD-L1) can induce strong antitumor immune response thus effectively killing primary tumors and further inhibiting metastatic tumors. Hence, BMT@LA-based "nanovaccine" combining with αPD-L1 checkpoint blockade treatment can realize synergetic sonodynamic/gas/immunotherapy with enhanced antitumor therapeutic effects.


Asunto(s)
Neoplasias , Arginina , Humanos , Inmunoterapia , Neoplasias/terapia , Titanio
2.
Adv Mater ; 33(4): e2004788, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33289219

RESUMEN

The past decades have witnessed hyperthermia therapy (HTT) as an emerging strategy against malignant tumors. Nanomaterial-based photothermal therapy (PTT) and magnetic hyperthermia (MHT), as highly effective and noninvasive treatment models, offer advantages over other strategies in the treatment of different types of tumors. However, both PTT and MHT cannot completely cure cancer due to recurrence and distal metastasis. In recent years, cancer immunotherapy has attracted widespread attention owing to its capability to activate the body's own natural defense to identify, attack, and eradicate cancer cells. Significant efforts have been devoted to studying the activated immune responses caused by hyperthermia-ablated tumors. In this article, the synergistic mechanism of HTT in immunotherapy, including immunogenic cell death and reversal of the immunosuppressive tumor microenvironment is discussed. The reports of the combination of HTT or HTT-based multimodal therapy with immunotherapy, including immunoadjuvant exploitation, immune checkpoint blockade therapy, and adoptive cellular immunotherapy are summarized. As highlighted, these strategies could achieve synergistically enhanced therapeutic outcomes against both primary tumors and metastatic lesions, prevent cancer recurrence, and prolong the survival period. Finally, current challenges and prospective developments in HTT-synergized immunotherapy are also reviewed.


Asunto(s)
Hipertermia Inducida/métodos , Inmunoterapia/métodos , Animales , Humanos , Neoplasias/terapia
3.
Biomaterials ; 252: 120093, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32422490

RESUMEN

Although synergistic therapy for tumors has displayed significant promise for effective treatment of cancer, developing a simple and effective strategy to build a multi-functional nanoplatform is still a huge challenge. By virtue of the characteristics of tumor microenvironment, such as hypoxia, slight acidity and H2O2 overexpression, Au2Pt-PEG-Ce6 nanoformulation is constructed for collaborative chemodynamic/phototherapy of tumors. Specifically, the Au2Pt nanozymes with multiple functions are synthesized in one step at room temperature. The photosensitizer chlorin e6 (Ce6) is covalently linked to Au2Pt nanozymes for photodynamic therapy (PDT). Interestingly, the Au2Pt nanozymes possess catalase- and peroxidase-like activities simultaneously, which not only can generate O2 for relaxation of tumor hypoxia and enhancement of PDT efficiency but also can produce ∙OH for chemodynamic therapy (CDT). In addition, the high photothermal conversion efficiency (η = 31.5%) of Au2Pt-PEG-Ce6 nanoformulation provides the possibility for photoacoustic (PA) and photothermal (PT) imaging guided photothermal therapy (PTT). Moreover, the presence of high-Z elements (Au and Pt) in Au2Pt-PEG-Ce6 nanoformulation endows it with the ability to act as an X-ray computed tomography (CT) imaging contrast agent. All in all, the Au2Pt-PEG-Ce6 exhibits great potential in multimodal imaging-guided synergistic PTT/PDT/CDT with remarkably tumor specificity and enhanced therapy.


Asunto(s)
Fotoquimioterapia , Porfirinas , Línea Celular Tumoral , Clorofilidas , Peróxido de Hidrógeno , Fármacos Fotosensibilizantes/uso terapéutico , Fototerapia
4.
Small ; 16(14): e1907146, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32162784

RESUMEN

Photoimmunotherapy can not only effectively ablate the primary tumor but also trigger strong antitumor immune responses against metastatic tumors by inducing immunogenic cell death. Herein, Cu2 MoS4 (CMS)/Au heterostructures are constructed by depositing plasmonic Au nanoparticles onto CMS nanosheets, which exhibit enhanced absorption in near-infrared (NIR) region due to the newly formed mid-gap state across the Fermi level based on the hybridization between Au 5d orbitals and S 3p orbitals, thus resulting in more excellent photothermal therapy and photodynamic therapy (PDT) effect than single CMS upon NIR laser irradiation. The CMS and CMS/Au can also serve as catalase to effectively relieve tumor hypoxia, which can enhance the therapeutic effect of O2 -dependent PDT. Notably, the NIR laser-irradiated CMS/Au can elicit strong immune responses via promoting dendritic cells maturation, cytokine secretion, and activating antitumor effector T-cell responses for both primary and metastatic tumors eradication. Moreover, CMS/Au exhibits outstanding photoacoustic and computed tomography imaging performance owing to its excellent photothermal conversion and X-ray attenuation ability. Overall, the work provides an imaging-guided and phototherapy-induced immunotherapy based on constructing CMS/Au heterostructures for effectively tumor ablation and cancer metastasis inhibition.


Asunto(s)
Cobre , Oro , Inmunoterapia , Nanopartículas del Metal , Molibdeno , Neoplasias , Fototerapia , Catalasa/metabolismo , Cobre/química , Oro/química , Humanos , Nanopartículas del Metal/química , Molibdeno/química , Neoplasias/terapia
5.
ACS Appl Mater Interfaces ; 12(10): 11320-11328, 2020 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-32067461

RESUMEN

Nanomaterials with intrinsic peroxidase-like activities are able to catalyze the oxidation of the substrate with the peroxide, which have been widely considered as artificial enzymatic agents in cancer therapy. However, current peroxidase catalytic oxidation treatments generating reactive oxygen species rely highly on hydrogen peroxide and pH, which limit greatly their therapeutic efficiency in the tumor microenvironment. Here, we report a strategy to construct the complex virus-like Fe3O4@Bi2S3 nanocatalysts (F-BS NCs) by connecting typical peroxidase Fe3O4 (MNPs) with a narrow band gap semiconductor Bi2S3 (BS) to enhance the enzymatic activity resorting to the limited intratumoral peroxide and efficient external photothermal stimuli. In this formulation, the integrated F-BS NCs induce cancer-cell death through mild photothermal treatment and sequential photothermal-stimulative catalysis of H2O2 into highly toxic •OH under 808 nm laser, which successfully realize a remarkable synergistic anticancer achievement.


Asunto(s)
Antineoplásicos , Hipertermia Inducida , Nanopartículas de Magnetita/química , Nanomedicina/métodos , Peroxidasa , Animales , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Células HeLa , Humanos , Ratones , Oxidación-Reducción , Peroxidasa/química , Peroxidasa/metabolismo , Peroxidasa/farmacología
6.
ACS Appl Mater Interfaces ; 11(51): 47730-47738, 2019 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-31790193

RESUMEN

To date, the limited light conversion ability and the oxygen-dependent therapeutic process of most photosensitizers make it difficult to achieve satisfactory therapeutic effects in the complex tumor microenvironment, especially the anoxic environment. Herein, the black mesoporous titania (BMT) with large pore size (∼8 nm) is synthesized as a new-style carrier for radical generator drug (AIBI) loading. The BMT as a light transducer can convert near-infrared (NIR) light energy into thermal energy and chemical energy (•OH), contributing to photothermal therapy (PTT) and photodynamic therapy (PDT), respectively. More importantly, AIBI would be thermally decomposed into alkyl radicals (•R) for thermodynamic therapy (TDT). The high concentration of free radicals produced by BMT@AIBI NCs resulted in double-strand breaks (DSBs) of DNA and finally induced cancer cell apoptosis. Since the generation of radicals is unrelated to oxygen, the BMT@AIBI NCs with NIR irradiation presented excellent in vitro and in vivo anticancer results under hypoxic conditions. The reported NIR-induced platform based on BMT@AIBI NCs, which could perform triple energy-conversion processes including light energy to thermal energy, to chemical energy, and to thermal energy then to chemical energy, realizes synergetic photo-thermal-dynamic therapy (PTT, PDT, and TDT) to overcome the problem of tumor hypoxia for enhanced anticancer effects.


Asunto(s)
Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/química , Titanio/química , Animales , Roturas del ADN de Doble Cadena , Humanos , Hipertermia Inducida/métodos , Fototerapia/métodos
7.
Adv Mater ; 31(51): e1905271, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31680346

RESUMEN

The unique tumor microenvironment (TME) facilitates cancer proliferation and metastasis, and it is hard to cure cancer completely via monotherapy. Herein, a multifunctional cascade bioreactor based on hollow mesoporous Cu2 MoS4 (CMS) loaded with glucose oxidase (GOx) is constructed for synergetic cancer therapy by chemo-dynamic therapy (CDT)/starvation therapy/phototherapy/immunotherapy. The CMS harboring multivalent elements (Cu1+/2+ , Mo4+/6+ ) exhibit Fenton-like, glutathione (GSH) peroxidase-like and catalase-like activity. Once internalized into the tumor, CMS could generate ·OH for CDT via Fenton-like reaction and deplete overexpressed GSH in TME to alleviate antioxidant capability of the tumors. Moreover, under hypoxia TME, the catalase-like CMS could react with endogenous H2 O2 to generate O2 for activating the catalyzed oxidation of glucose by GOx for starvation therapy accompanied with the regeneration of H2 O2 . The regenerated H2 O2 can devote to Fenton-like reaction for realizing GOx-catalysis-enhanced CDT. Meanwhile, the CMS under 1064 nm laser irradiation shows remarkable tumor-killing ability by phototherapy due to its excellent photothermal conversion efficiency (η = 63.3%) and cytotoxic superoxide anion (·O2 - ) generation performance. More importantly, the PEGylated CMS@GOx-based synergistic therapy combined with checkpoint blockade therapy could elicit robust immune responses for both effectively ablating primary tumors and inhibiting cancer metastasis.


Asunto(s)
Cobre/química , Portadores de Fármacos/química , Inmunoterapia/métodos , Molibdeno/química , Fototerapia/métodos , Animales , Línea Celular Tumoral , Terapia Combinada , Glucosa Oxidasa/química , Glucosa Oxidasa/uso terapéutico , Ratones
8.
Acta Biomater ; 99: 295-306, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31437636

RESUMEN

Although neodymium vanadate (NdVO4) has been investigated and applied in some fields owing to its intensive ultraviolet (UV) light absorption, weak absorption in visible (Vis) and near infrared (NIR) regions constrains its environmental remediation and biomedical applications. Herein, plasmonic precious metal Au as light trapping agent is deposited onto NdVO4 to form metal/semiconductor hybrid nanostructure for improving the Vis/NIR light absorption. NdVO4/Au heterojunction nanocrystals (NCs) were synthesized by NdVO4 nanorods (NRs) and plasmonic Au nanoparticles (NPs), followed by introducing polyvinylpyrrolidone (PVP) to enhance stability and biocompatibility, which exhibit elevated photocatalytic performance for organic dye degradation, photothermal conversion effect as high as 32.15% and cytotoxic reactive oxygen species (ROS) production ability. NdVO4/Au can be internalized efficiently via endocytosis and cause apparent phototoxicity on HeLa cells. In vivo experiments further show that NdVO4/Au can act as a high-efficiency NIR light-triggered anticancer agent with excellent tumor inhibition effect. In addition, based on outstanding light-to-heat conversion performance and thermal expansion effect under NIR irradiation, NdVO4/Au provides photothermal (PT) and photoacoustic (PA) dual-modal imaging platform for precise cancer diagnosis and treatment. STATEMENTS OF SIGNIFICANCE: It's the first report on integrating precious metal Au and rare earth orthovanadates semiconductor into NdVO4/Au heterojunction NCs. The as-prepared NdVO4/Au heterojunction NCs exhibits improved absorption in Vis/NIR region and increased generation efficiency of photo-induced electron/hole pairs due to the LSPR effect, which results in enhanced photothermal conversion efficiency and the production ability of cytotoxic O2- and OH in comparison with pristine NdVO4. For further clinical application, NdVO4/Au heterojunction NCs could be served as anticancer therapeutic agent for PA/PT dual-modal imaging guided and NIR-triggered photothermal/photodynamic synergistic anticancer treatment.


Asunto(s)
Oro/química , Nanopartículas del Metal/química , Neodimio/química , Neoplasias/terapia , Técnicas Fotoacústicas/métodos , Fotoquimioterapia/métodos , Vanadatos/química , Animales , Antineoplásicos/farmacología , Compuestos Azo/química , Catálisis , Femenino , Células HeLa , Humanos , Rayos Infrarrojos , Rayos Láser , Luz , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión , Nanocompuestos/química , Nanopartículas , Oxígeno/química , Especies Reactivas de Oxígeno/química , Espectroscopía Infrarroja Corta , Nanomedicina Teranóstica/métodos
9.
Nanoscale ; 11(20): 10129-10136, 2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31089645

RESUMEN

The plasmonic cerium vanadate (CeVO4) semiconductor and plasmonic silver (Ag) metal exhibit a localized surface plasmon resonance (LSPR) effect in the visible (Vis)-light region; however, weak absorption in the near-infrared (NIR) region restricts their environmental remediation and biomedical application. Herein, CeVO4/Ag nanohybrids with self-assembled heterostructure and improved Vis/NIR light absorption were synthesized from CeVO4 nanosheets and AgNO3 solution, which could serve as potential solar-driven catalytic agents and near-infrared (NIR) light responsive anticancer agents. Oleic acid-stabilized CeVO4 nanosheets were modified with the HS-PEG1000-OH by the thiol-ene click reaction and presented self-assembly morphology in aqueous solution due to hydrophobic-hydrophobic interactions. Sulfhydryl (-SH) groups provided stable sites for Ag+ ions on the surface of CeVO4, and Ag+ ions could be directly reduced by Ce3+ ions to form CeVO4/Ag heterojunction nanocrystals (NCs). Due to the higher absorption in the Vis/NIR light region than CeVO4 nanosheets, CeVO4/Ag NCs led to the improved solar light responsive photocatalytic degradation of organic dyes. Upon the exposure of these NCs to an 808 nm laser, CeVO4/Ag NCs show high photothermal conversion efficiency, ROS generation ability and photoacoustic (PA) signal for implementing PA imaging-guided photothermal/photodynamic synergistic cancer therapy with better tumor inhibition effect.


Asunto(s)
Cerio/química , Rayos Infrarrojos , Nanocompuestos/química , Plata/química , Vanadatos/química , Animales , Bencenosulfonatos/química , Bencenosulfonatos/metabolismo , Catálisis , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Ratones , Nanocompuestos/uso terapéutico , Nanocompuestos/toxicidad , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/terapia , Ácidos Oléicos/química , Fotoquimioterapia , Fototerapia , Trasplante Heterólogo
10.
Adv Healthc Mater ; 7(20): e1800351, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29938919

RESUMEN

Phototherapy with the properties of specific spatial/temporal selectivity and minimal invasiveness has been acknowledged as one of the most promising cancer therapy types. Among all the photoactive substance for phototherapy, titanium dioxide (TiO2 ) nanomaterials are paid more and more attention due to their outstanding photocatalytic properties, prominent biocompatibility, and excellent chemical stability. However, the wide bandgap (3.0-3.2 eV) of TiO2 limits its absorption only to the ultraviolet (UV) light region. For a long time, UV light-stimulated TiO2 was applied in the phototherapy researches of tumors located in the skin layer, while it is unsatisfactory for most deep-tissue tumors. Due to the maximum penetration into tissue existing in the near-infrared (NIR) region, how to use NIR light to trigger photochemical reaction of TiO2 remains a big challenge. In this review, two strategies to develop and construct NIR-triggered TiO2 -based nanocomposites (NCs) for phototherapy are summarized, and the relevant mechanism and background knowledge of TiO2 -based phototherapy are also given in order to better understand the application value and current situation of TiO2 in phototherapy. Finally, the challenges and research directions of TiO2 in the future clinic phototherapy application are also discussed.


Asunto(s)
Rayos Infrarrojos , Nanocompuestos/uso terapéutico , Fototerapia , Titanio/uso terapéutico , Animales , Humanos , Nanocompuestos/química , Nanocompuestos/ultraestructura
11.
Nanomedicine ; 13(3): 875-883, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27993724

RESUMEN

Anticancer drug doxorubicin hydrochloride (DOX)-loaded photothermal nanocomposite MnFe2O4@mSiO2 with magnetic targeting and T1/T2-weighted dual-mode magnetic resonance imaging of MnFe2O4 core and NIR/pH-coupling sensitive mesoporous silica shell nanocarriers was designed and synthesized successfully. The anticancer drug DOX can be absorbed into mesoporous layer of MnFe2O4@mSiO2 nanocomposite, which shows obvious photothermal/chemo dual-modal synergistic therapies triggered by NIR/pH. Under 808 nm irradiation, MnFe2O4 can transform light into thermo, which can not only ablate tumor cells directly but also promote chemotherapy drugs releasing from mesoporous layer to kill tumor cells. The lower pH can also promote DOX releasing from mesoporous layer to enhance tumor inhibitory effect. It is confirmed that biocompatible DOX-MnFe2O4@mSiO2 nanocomposites can act as a potential multifunctional platform for effective magnetic targeting photothermal/chemo dual-modal synergistic therapies with enhanced anti-tumor efficacy and T1/T2-weighted dual-mode magnetic resonance imaging (MRI) applications in vivo.


Asunto(s)
Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Compuestos Férricos/química , Imagen por Resonancia Magnética/métodos , Compuestos de Manganeso/química , Nanocompuestos/química , Neoplasias/terapia , Dióxido de Silicio/química , Animales , Antineoplásicos/uso terapéutico , Terapia Combinada/métodos , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/uso terapéutico , Doxorrubicina/uso terapéutico , Compuestos Férricos/uso terapéutico , Células HeLa , Humanos , Hipertermia Inducida/métodos , Campos Magnéticos , Compuestos de Manganeso/uso terapéutico , Ratones , Nanocompuestos/uso terapéutico , Nanocompuestos/ultraestructura , Neoplasias/diagnóstico por imagen , Fototerapia/métodos , Dióxido de Silicio/uso terapéutico
12.
J Mater Chem B ; 5(9): 1803-1814, 2017 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-32263921

RESUMEN

Near-infrared (NIR) light induced phototherapy has attracted considerable attention due to its deep therapeutic depth. To improve the therapeutic outcome and address non-selective side effects, the combination of complementary phototherapeutic strategies in a single nanoagent with precise targeting ability may provide an effective approach for cancer therapy. Thus we have developed an 808 nm NIR light triggered nanosystem based on IR806 dye functionalized MnFe2O4 (MFO-IR) for synchronous magnetic targeted and magnetic resonance (MR) imaging guided in vivo photodynamic/photothermal synergistic therapy. In this construction strategy, carboxylic acid functionalized NIR dye IR806 is explored as an 808 nm NIR-excited photosensitizer (PS) for the first time, which can also provide a conjugation site for MnFe2O4 nanoparticles (MFO NPs). Here, monodisperse MFO NPs have multiple capacities as dye carriers, targeting ligands, MRI contrast agents and photothermal agents. MFO-IR nanocomposites (NCs) with negligible toxicity present efficient NIR-mediated photothermal damage and ROS cytotoxicity via the relevant in vitro experimental investigations. With ideal magnetic targeting effects and remarkable NIR light-responsive properties, these MFO-IR NCs exhibit high in vivo tumor localization and could destroy subcutaneous solid tumors completely under an external magnetic field and 808 nm laser irradiation. Consequently, this magnetic nanosystem has great potential for simultaneous diagnosis and precise cancer phototherapy.

13.
Dalton Trans ; 45(33): 13061-9, 2016 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-27529086

RESUMEN

Recently, incorporating multiple components into one nanostructured matrix to construct a multifunctional nanomedical platform has attracted more and more attention for simultaneous anticancer diagnosis and therapy. Herein, a novel anti-cancer nanoplatform has been successfully developed by coating a uniform shell of poly(acrylic acid) (PAA) on the surface of CuS-decorated upconversion nanoparticles (UCNPs). Benefiting from the enhanced 808 nm-excited UCL intensity of the multilayer UCNPs, the unique photothermal properties of CuS and the pH-responsive drug release capacity of the PAA shell, such a nanoplatform design of UCNPs-CuS@PAA (labeled UCP) offers a new route to achieve 808 nm-excited UCL imaging guided chemo/photothermal combination therapy. We have found that the combined chemo/photothermal therapy can significantly improve the therapeutic efficacy compared with chemotherapy or photothermal therapy (PTT) alone. Moreover, the pH/NIR-dependent drug delivery properties, 808 nm-excited UCL imaging, as well as in vitro/in vivo biocompatibility tests were also investigated in detail. These results show promising applications of UCP nanoparticles as a novel theranostic agent for the detection and treatment of tumors.


Asunto(s)
Resinas Acrílicas/administración & dosificación , Cobre/administración & dosificación , Nanopartículas/administración & dosificación , Resinas Acrílicas/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Cobre/química , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Humanos , Luz , Ratones , Microscopía Fluorescente , Nanopartículas/química , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fototerapia , Carga Tumoral/efectos de los fármacos
14.
Nanoscale ; 8(12): 6837-50, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-26956400

RESUMEN

Combining multi-model treatments within one single system has attracted great interest for the purpose of synergistic therapy. In this paper, hollow gold nanospheres (HAuNs) coated with a temperature-sensitive polymer, poly(oligo(ethylene oxide) methacrylate-co-2-(2-methoxyethoxy)ethyl methacrylate) (p(OEGMA-co-MEMA)), co-loaded with DOX and a photosensitizer Chlorin e6 (Ce6) were successfully synthesized. As high as 58% DOX and 6% Ce6 by weight could be loaded onto the HAuNs-p(OEGMA-co-MEMA) nanocomposites. The grafting polymer brushes outside the HAuNs play the role of "gate molecules" for controlled drug release by 650 nm laser radiation owing to the temperature-sensitive property of the polymer and the photothermal effect of HAuNs. The HAuNs-p(OEGMA-co-MEMA)-Ce6-DOX nanocomposites with 650 nm laser radiation show effective inhibition of cancer cells in vitro and enhanced anti-tumor efficacy in vivo. In contrast, control groups without laser radiation show little cytotoxicity. The nanocomposite demonstrates a way of "killing three birds with one stone", that is, chemotherapy, photothermal and photodynamic therapy are triggered simultaneously by the 650 nm laser stimulation. Therefore, the nanocomposites show the great advantages of multi-modal synergistic effects for cancer therapy by a remote-controlled laser stimulus.


Asunto(s)
Quimioterapia/métodos , Nanopartículas del Metal/química , Neoplasias/terapia , Fotoquimioterapia/métodos , Fototerapia/métodos , Animales , Antineoplásicos/química , Materiales Biocompatibles/química , Línea Celular Tumoral , Disulfuros , Sistemas de Liberación de Medicamentos , Oro/química , Células HeLa , Humanos , Rayos Láser , Ratones , Nanocompuestos/química , Nanosferas/química , Polímeros/química , Especies Reactivas de Oxígeno/química , Espectroscopía Infrarroja por Transformada de Fourier , Electricidad Estática , Temperatura , Difracción de Rayos X
15.
ACS Appl Mater Interfaces ; 7(37): 20696-706, 2015 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-26325285

RESUMEN

Near-infrared light is an attractive stimulus due to its noninvasive and deep tissue penetration. Particularly, NIR light is utilized for cancer thermotherapy and on-demand release of drugs by the disruption of the delivery carriers. Here we have prepared a novel NIR-responsive DNA-hybrid-gated nanocarrier based on mesoporous silica-coated Cu1.8S nanoparticles. Cu1.8S nanoparticles, possessing high photothermal conversion efficiency under a 980 nm laser, were chosen as photothermal agents. The mesoporous silica structure could be used for drug storage/delivery and modified with aptamer-modified GC-rich DNA-helix as gatekeepers, drug vectors, and targeting ligand. Simultaneously, the as-produced photothermal effect caused denaturation of DNA double strands, which triggered the drug release of the DNA-helix-loaded hydrophilic drug doxorubicin and mesopore-loaded hydrophobic drug curcumin, resulting in a synergistic therapeutic effect. The Cu1.8S@mSiO2 nanocomposites endocytosed by cancer cells through the aptamer-mediated mode are able to generate rational release of doxorubicin/curcumin under NIR irradiation, strongly enhancing the synergistic growth-inhibitory effect of curcumin against doxorubicin in MCF-7 cells, which is associated with a strong mitochondrial-mediated cell apoptosis progression. The underlying mechanism of apoptosis showed a strong synergistic inhibitory effect both on the expression of Bcl-2, Bcl-xL, Mcl-1, and upregulated caspase 3/9 activity and on the expression level of Bak and Bax. Therefore, Cu1.8S@mSiO2 with efficient synergistic therapeutic efficiency is a potential multifunctional cancer therapy nanoplatform.


Asunto(s)
Aptámeros de Nucleótidos/química , Sistemas de Liberación de Medicamentos , Luz , Nanopartículas/química , Dióxido de Silicio/química , Espectroscopía Infrarroja Corta/métodos , Temperatura , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cobre/química , Curcumina/farmacología , ADN , Doxorrubicina/farmacología , Endocitosis/efectos de los fármacos , Células HEK293 , Humanos , Células MCF-7 , Nanopartículas/ultraestructura , Nanosferas/química , Porosidad , Especies Reactivas de Oxígeno/metabolismo
16.
Biomaterials ; 63: 115-27, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26093792

RESUMEN

To integrate biological imaging and multimodal therapies into one platform for enhanced anti-cancer efficacy, we have designed a novel core/shell structured nano-theranostic by conjugating photosensitive Au25(SR)18 - (SR refers to thiolate) clusters, pH/temperature-responsive polymer P(NIPAm-MAA), and anti-cancer drug (doxorubicin, DOX) onto the surface of mesoporous silica coated core-shell up-conversion nanoparticles (UCNPs). It is found that the photodynamic therapy (PDT) derived from the generated reactive oxygen species and the photothermal therapy (PTT) arising from the photothermal effect can be simultaneously triggered by a single 980 nm near infrared (NIR) light. Furthermore, the thermal effect can also stimulate the pH/temperature sensitive polymer in the cancer sites, thus realizing the targeted and controllable DOX release. The combined PDT, PTT and pH/temperature responsive chemo-therapy can markedly improve the therapeutic efficacy, which has been confirmed by both in intro and in vivo assays. Moreover, the doped rare earths endow the platform with dual-modal up-conversion luminescent (UCL) and computer tomography (CT) imaging properties, thus achieving the target of imaging-guided synergistic therapy under by a single NIR light.


Asunto(s)
Acrilamidas/química , Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sustancias Luminiscentes/química , Nanoconjugados/química , Neoplasias/diagnóstico , Neoplasias/terapia , Ácidos Polimetacrílicos/química , Acrilamidas/uso terapéutico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/uso terapéutico , Doxorrubicina/uso terapéutico , Liberación de Fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Hipertermia Inducida/métodos , Sustancias Luminiscentes/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Nanoconjugados/uso terapéutico , Nanoconjugados/ultraestructura , Imagen Óptica , Fotoquimioterapia/métodos , Ácidos Polimetacrílicos/uso terapéutico , Temperatura , Nanomedicina Teranóstica/métodos , Tomografía Computarizada por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA