Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
ACS Appl Mater Interfaces ; 16(8): 9968-9979, 2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38358298

RESUMO

Foreseen as foundational in forthcoming oncology interventions are multimodal therapeutic systems. Nevertheless, the tumor microenvironment (TME), marked by heightened glucose levels, hypoxia, and scant concentrations of endogenous hydrogen peroxide could potentially impair their effectiveness. In this research, two-dimensional (2D) Ti3C2 MXene nanosheets are engineered with CeO2 nanozymes and glucose oxidase (GOD), optimizing them for TME, specifically targeting cancer therapy. Following our therapeutic design, CeO2 nanozymes, embodying both peroxidase-like and catalase-like characteristics, enable transformation of H2O2 into hydroxyl radicals for catalytic therapy while also producing oxygen to mitigate hypoxia. Concurrently, GOD metabolizes glucose, thereby augmenting H2O2 levels and disrupting the intracellular energy supply. When subjected to a near-infrared laser, 2D Ti3C2 MXene accomplishes photothermal therapy (PTT) and photodynamic therapy (PDT), additionally amplifying cascade catalytic treatment via thermal enhancement. Empirical evidence demonstrates robust tumor suppression both in vitro and in vivo by the CeO2/Ti3C2-PEG-GOD nanocomposite. Consequently, this integrated approach, which combines PTT/PDT and enzymatic catalysis, could offer a valuable blueprint for the development of advanced oncology therapies.


Assuntos
Hipertermia Induzida , Neoplasias , Nitritos , Elementos de Transição , Humanos , Glucose Oxidase , Peróxido de Hidrogênio , Titânio/farmacologia , Hipertermia , Neoplasias/terapia , Glucose , Hipóxia , Microambiente Tumoral , Linhagem Celular Tumoral
2.
Sci Adv ; 9(44): eadi9980, 2023 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-37910608

RESUMO

Thermoelectric therapy has emerged as a promising treatment strategy for oncology, but it is still limited by the low thermoelectric catalytic efficiency at human body temperature and the inevitable tumor thermotolerance. We present a photothermoelectric therapy (PTET) strategy based on triphenylphosphine-functionalized Cu3VS4 nanoparticles (CVS NPs) with high copper ionic mobility at room temperature. Under near-infrared laser irradiation, CVS NPs not only generate hyperthermia to ablate tumor cells but also catalytically yield superoxide radicals and induce endogenous NADH oxidation through the Seebeck effect. Notably, CVS NPs can accumulate inside mitochondria and deplete NADH, reducing ATP synthesis by competitively inhibiting the function of complex I, thereby down-regulating the expression of heat shock proteins to relieve tumor thermotolerance. Both in vitro and in vivo results show notable tumor suppression efficacy, indicating that the concept of integrating PTET and mitochondrial metabolism modulation is highly feasible and offers a translational promise for realizing precise and efficient cancer treatment.


Assuntos
Nanopartículas , Neoplasias , Humanos , Cobre/química , NAD , Fototerapia/métodos , Neoplasias/terapia , Neoplasias/patologia , Nanopartículas/química , Linhagem Celular Tumoral
3.
J Am Chem Soc ; 145(17): 9488-9507, 2023 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-36998235

RESUMO

Arming activatable mild-photothermal therapy (PTT) with the property of relieving tumor thermotolerance holds great promise for overcoming traditional mild PTT limitations such as thermoresistance, insufficient therapeutic effect, and off-target heating. Herein, a mitochondria-targeting, defect-engineered AFCT nanozyme with enhanced multi-enzymatic activity was elaborately designed as a tumor microenvironment (TME)-activatable phototheranostic agent to achieve remarkable anti-tumor therapy via "electron transport chain (ETC) interference and synergistic adjuvant therapy". Density functional theory calculations revealed that the synergistic effect among multi-enzyme active centers endows the AFCT nanozymes with excellent catalytic activity. In TME, open sources of H2O2 can be achieved by superoxide dismutase-mimicking AFCT nanozymes. In response to the dual stimuli of H2O2 and mild acidity, the peroxidase-mimicking activity of AFCT nanozymes not only catalyzes the accumulation of H2O2 to generate ·OH but also converts the loaded 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) into its oxidized form with strong near-infrared absorption, specifically unlocking its photothermal and photoacoustic imaging properties. Intriguingly, the undesired thermoresistance of tumor cells can be greatly alleviated owing to the reduced expression of heat shock proteins enabled by NADH POD-mimicking AFCT-mediated NADH depletion and consequent restriction of ATP supply. Meanwhile, the accumulated ·OH can facilitate both apoptosis and ferroptosis in tumor cells, resulting in synergistic therapeutic outcomes in combination with TME-activated mild PTT.


Assuntos
Nanopartículas , Neoplasias , Humanos , Terapia Fototérmica , Fototerapia/métodos , Peróxido de Hidrogênio , Transporte de Elétrons , NAD , Nanopartículas/uso terapêutico , Neoplasias/terapia , Linhagem Celular Tumoral , Microambiente Tumoral
4.
ACS Appl Mater Interfaces ; 14(40): 45217-45228, 2022 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-36190449

RESUMO

Nanozymes with the merits of effective enzyme-mimic activities, tunable catalytic properties, pH/temperature tolerance, and high stability have been consumingly researched for nanocatalytic therapy. Herein, the union nanozymes and a natural enzyme nanoplatform (DMSN@CoFe2O4/GOD-PCM) are elaborately designed by simply depositing an ultrasmall cobalt ferrite (CoFe2O4) bimetallic oxide nanozyme and natural glucose oxidase (GOD) that are loaded into the aperture (∼12 nm) of dendritic mesoporous silica (DMSN) for near-infrared-II-enhanced tumor therapy. Upon irradiation, the hyperthermia generated by CoFe2O4 nanozymes unlocks the "gate" of phase-change material (PCM) for releasing GOD, which reshapes the specific tumor microenvironment (TME) through the glucose metabolism pathway. The resulting strengthened acid condition and a considerable amount of H2O2 efficiently initiate the cascade catalysis reactions. Moreover, highly toxic hydroxyl radicals are generated with a Co/Fe dual-cycle system of ultrasmall CoFe2O4 nanozymes. The in situ glutathione consumption and hypoxia relief further amplify oxidative stress. In addition, chemotherapeutic effects due to the cytotoxicity of cobalt ions enhance the therapeutic performance. Collectively, this study provides a proof of concept for TME-reshaped natural and artificial nanozyme cascade catalysis for combined reactive oxygen species-based therapy and chemotherapy.


Assuntos
Hipertermia Induzida , Microambiente Tumoral , Catálise , Linhagem Celular Tumoral , Cobalto , Compostos Férricos , Glucose , Glucose Oxidase , Glutationa , Peróxido de Hidrogênio , Óxidos , Espécies Reativas de Oxigênio/metabolismo , Dióxido de Silício
5.
ACS Nano ; 16(7): 10904-10917, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35797013

RESUMO

Semimetallic nanomaterials as photothermal agents for bioimaging and cancer therapy have attracted tremendous interest. However, the poor photothermal stability, low biocompatibility, and single component limit their therapeutic efficiency in cancer treatment. Here, manganese-doped VSe2 semimetallic nanosheets were prepared and subsequently modified with chitosan (named VSe2/Mn-CS NSs) for combined enzyme catalytic and photothermal therapy. VSe2/Mn-CS NSs show high photothermal property with a photothermal conversion efficiency of 34.61% upon 808 nm near-infrared laser irradiation. In the tumor microenvironment, VSe2/Mn-CS NSs can convert endogenous H2O2 into lethal hydroxyl radicals (•OH) to induce cancer cell apoptosis. The interaction between glutathione (GSH) and Se-Se bonds in VSe2/Mn-CS NSs results in the depletion of GSH level, and the valence states transition of manganese ions is also beneficial for the GSH consumption. This dual depletion of GSH markedly enhances the peroxidase (POD) activity, leading to the high •OH production and the improved therapeutic effect. What is more, the T1-weighted magnetic resonance and photoacoustic imaging endow VSe2/Mn-CS NSs with the ability to guide and track the treatment process. Our study provides a research strategy for the application of semimetallic nanomaterials in cancer diagnosis and treatment.


Assuntos
Hipertermia Induzida , Metaloides , Neoplasias , Humanos , Manganês/uso terapêutico , Peróxido de Hidrogênio , Glutationa , Hipertermia Induzida/métodos , Microambiente Tumoral , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Linhagem Celular Tumoral
6.
ACS Nano ; 16(6): 8939-8953, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35666853

RESUMO

Nanosystem-mediated tumor radiosensitization strategy combining the features of X-ray with infinite penetration depth and high atomic number elements shows considerable application potential in clinical cancer therapy. However, it is difficult to achieve satisfactory anticancer efficacy using clinical radiotherapy for the majority of solid tumors due to the restrictions brought about by the tumor hypoxia, insufficient DNA damage, and rapid DNA repair during and after treatment. Inspired by the complementary advantages of nitric oxide (NO) and X-ray-induced photodynamic therapy, we herein report a two-dimensional nanoplatform by the integration of the NO donor-modified LiYF4:Ce scintillator and graphitic carbon nitride nanosheets for on-demand generation of highly cytotoxic peroxynitrite (ONOO-). By simply adjusting the Ce3+ doping content, the obtained nanoscintillator can realize high radioluminescence, activating photosensitive materials to simultaneously generate NO and superoxide radical for the formation of ONOO- in the tumor. Obtained ONOO- effectively amplifies therapeutic efficacy of radiotherapy by directly inducing mitochondrial and DNA damage, overcoming hypoxia-associated radiation resistance. The level of glutamine synthetase (GS) is downregulated by ONOO-, and the inhibition of GS delays DNA damage repair, further enhancing radiosensitivity. This work establishes a combinatorial strategy of ONOO- to overcome the major limitations of radiotherapy and provides insightful guidance to clinical radiotherapy.


Assuntos
Neoplasias , Ácido Peroxinitroso , Humanos , Óxido Nítrico , Dano ao DNA , Reparo do DNA , Neoplasias/radioterapia
7.
Small ; 18(28): e2200786, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35661402

RESUMO

Mild photothermal therapy (PTT, <45 °C) can prevent tumor metastasis and heat damage to normal tissue, compared with traditional PTT (>50 °C). However, its therapeutic efficacy is limited owing to the hypoxic tumor environment and tumor thermoresistance owing to the overproduction of heat shock proteins (HSPs). Herein, a near-infrared (NIR)-triggered theranostic nanoplatform (GA-PB@MONs@LA) is designed for synergistic mild PTT and enhanced Fenton nanocatalytic therapy against hypoxic tumors. The nanoplatform is fabricated by the confined formation of Prussian blue (PB) nanoparticles in mesoporous organosilica nanoparticles (MONs), followed by the loading of gambogic acid (GA), an HSP90 inhibitor, and coating with thermo-sensitive lauric acid (LA). Upon NIR irradiation, the photothermal effect (44 °C) of PB not only induces apoptosis of tumor cells but also triggers the on-demand release of GA, inhibiting the production of HSP90. Moreover, the delivered heat simultaneously enhances the catalase-like and Fenton activity of PB@MONs@LA in an acidic tumor microenvironment, relieving the tumor hypoxia and promoting the generation of highly toxic •OH. In addition, the nanoplatform enables magnetic resonance/photoacoustic dual-modal imaging. Thus, this study describes a distinctive paradigm for the development of NIR-triggered theranostic nanoplatforms for enhanced cancer therapy.


Assuntos
Antineoplásicos , Hipertermia Induzida , Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Preparações de Ação Retardada , Humanos , Hipertermia Induzida/métodos , Hipóxia/terapia , Neoplasias/terapia , Fototerapia/métodos , Medicina de Precisão , Nanomedicina Teranóstica/métodos , Microambiente Tumoral
8.
ACS Appl Mater Interfaces ; 14(2): 2650-2662, 2022 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-34995459

RESUMO

Smart nanotheranostic systems (SNSs) have attracted extensive attention in antitumor therapy. Nevertheless, constructing SNSs with disease diagnosis ability, improved drug delivery efficiency, inherent imaging performance, and high treatment efficiency remains a scientific challenge. Herein, ultrasmall tin dioxide (SnO2) was assembled with upconversion nanoparticles (UCNPs) to form mesoporous nanocapsules by an in situ hydrothermal deposition method, followed by loading with doxorubicin (DOX) and modification with bovine serum albumin (BSA). pH/near-infrared dual-responsive nanotheranostics was constructed for computed tomography (CT) and magnetic resonance (MR) imaging-induced collaborative cancer treatment. The mesoporous channel of SnO2 was utilized as a reservoir to encapsulate DOX, an antineoplastic drug, for chemotherapy and as a semiconductor photosensitizer for photodynamic therapy (PDT). Furthermore, the DOX-loaded UCNPs@SnO2-BSA nanocapsules combined PDT, Nd3+-doped UCNP-triggered hyperthermia effect, and DOX-triggered chemotherapy simultaneously and demonstrated prominently enhanced treatment efficiency compared to the monotherapy model. Moreover, tin, as one of the trace elements in the human body, has a similar X-ray attenuation coefficient to iodine and therefore can act as a contrast agent for CT imaging to monitor the treatment process. Such an orchestrated synergistic anticancer treatment exhibited apparent inhibition of tumor growth in tumor-bearing mice with negligible side effects. Our study demonstrates nanocapsules with excellent biocompatibility and great potential for cancer treatment.


Assuntos
Antibióticos Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Doxorrubicina/farmacologia , Nanocápsulas/química , Fármacos Fotossensibilizantes/farmacologia , Nanomedicina Teranóstica , Compostos de Estanho/farmacologia , Animais , Antibióticos Antineoplásicos/química , Materiais Biocompatíveis/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Doxorrubicina/química , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Concentração de Íons de Hidrogênio , Raios Infravermelhos , Teste de Materiais , Camundongos , Camundongos Endogâmicos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Tamanho da Partícula , Fármacos Fotossensibilizantes/química , Porosidade , Soroalbumina Bovina/química , Propriedades de Superfície , Compostos de Estanho/química
9.
ACS Nano ; 16(2): 3105-3118, 2022 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-35040328

RESUMO

The clinical application of photothermal therapy (PTT) is severely limited by the tissue penetration depth of excitation light, and enzyme therapy is hampered by its low therapeutic efficiency. As a two-dimensional ultrathin nanosheet with high absorbance in the near-infrared-II (NIR-II) region, the titanium carbide (Ti3C2) nanosheet can be used as a substrate to anchor functional components, like nanozymes and nanodrugs. Here, we decorate Pt artificial nanozymes on the Ti3C2 nanosheets to synthesize Ti-based MXene nanocomposites (Ti3C2Tx-Pt-PEG). In the tumor microenvironment, the Pt nanoparticles exhibit peroxidase-like (POD-like) activity, which can in situ catalyze hydrogen peroxide to generate hydroxyl radicals (•OH) to induce cell apoptosis and necrosis. Meanwhile, the composite shows a desirable photothermal effect upon NIR-II light irradiation with a low power density (0.75 W cm-2). Especially, the POD-like activity is significantly enhanced by the elevated temperature arising from the photothermal effect of Ti3C2Tx. Therefore, satisfactory synergistic PTT/enzyme therapy has been accomplished, accompanied by an applicable photoacoustic imaging capability to monitor and guide the therapeutic process. This work may provide an approach for hyperthermia-amplified nanozyme catalytic therapy, especially based on metal catalysts and MXene nanocomposites.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Catálise , Linhagem Celular Tumoral , Humanos , Hipertermia Induzida/métodos , Nanopartículas/uso terapêutico , Neoplasias/terapia , Titânio/farmacologia , Microambiente Tumoral
10.
Colloids Surf B Biointerfaces ; 197: 111358, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33068823

RESUMO

More effective strategies are needed to improve the treatment of liver cancer. Sono-photodynamic therapy (SPDT) has a more obvious antitumor effect than sonodynamic therapy (SDT) or photodynamic therapy (PDT). We aimed to investigate Glypican-3-targeted, curcumin-loaded microbubbles (GPC3-CUR-MBs)-mediated SPDT in liver cancer cells in vitro and in vivo. GPC3-CUR-MBs were prepared by streptavidin-biotin interactions and the immune ligation method. The characterization and toxicity of GPC3-CUR-MBs and the anti-liver cancer effects of GPC3-CUR-MB-mediated SPDT in vitro and in vivo were studied. We synthetized GPC3-CUR-MBs and found that GPC3-CUR-MBs had no significant toxicity to HepG2 liver cancer cells. In terms of the anti-liver cancer effects in vitro and in vivo, when we used CUR, CUR-MBs or GPC3-CUR-MBs as the sono/photosensitizers, the outcome of SPDT was superior to that of SDT or PDT alone. The outcomes with GPC3-CUR-MBs were better than those with CUR or CUR-MBs in the SDT, PDT or SPDT groups. During the treatment period, the weight of the HepG2 tumor-bearing mice did not decrease significantly, and no significant evidence of lung, heart, liver, spleen and kidney damage was found with H&E staining. Our results indicated that the anti-liver tumor effect of SPDT was better than that of SDT and PDT and that GPC3-CUR-MBs were promising sono/photosensitizers.


Assuntos
Curcumina , Neoplasias Hepáticas , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Curcumina/farmacologia , Glipicanas , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Microbolhas , Fosfolipídeos
11.
J Mater Chem B ; 8(46): 10559-10576, 2020 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-32939520

RESUMO

Photodynamic therapy (PDT) and photothermal therapy (PTT) are well-developed light therapy methods for cancer; however, both have a few areas that need improvement. A sustained PDT effect depends on the sustained generation of reactive oxygen species (ROS); therefore, adjusting the type of photosensitizer or the reaction mechanism to prolong the duration of the oxidation-reduction reaction is a possible solution for the continuation of the PDT effect. Further, if PTT could be combined with other treatments, it would bring about a more satisfactory therapeutic effect. To increase the treatment effect of the above two therapeutic methods, a collaborative treatment model of photo/chemodynamic therapy (PCDT) and PTT is needed and is the focus of this study. On the one hand, PCDT is a therapy that integrates PDT with Fenton-like reactions, and Fenton-like reactions can help PDT to produce more ROS by making better use of H2O2 in the tumor microenvironment. On the other hand, the PTT effect can also promote PCDT effects to some extent because rising temperature can elevate the redox reaction rate. Therefore, a copper oxide semiconductor photosensitizer was selected in this research to realize the abovementioned therapeutic purposes and experimental concepts. A porous silica carrier can facilitate the uniform attachment of the copper oxide photosensitizer to the SiO2 surface to form a relatively uniform nanostructure, and the nanoporous structure can increase the performance of the whole material to a certain extent. Based on these perspectives, SiO2@CuO nanotube (NT), an agent of both Fenton-like photosensitization and photothermal reagent, is synthesized by the hydrothermal co-precipitation template approach to shrink the tumor through the combined effect of PCDT and PTT. In this system, copper ions can participate in the Fenton-like reactions and make better use of H2O2 to generate more ROS. Herein, 808 nm light was chosen for irradiation because of its suitable excitation ability, applicable penetration and low intrinsic damage. The experimental results show that SiO2@CuO NT is a promising agent that combines PCDT and PTT for cancer treatment. This work provides guidance for the synthesis of Fenton-like photosensitizers for the PCDT effect.


Assuntos
Nanotubos de Carbono/química , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/química , Terapia Fototérmica/métodos , Dióxido de Silício/química , Animais , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Terapia Combinada/métodos , Relação Dose-Resposta a Droga , Feminino , Células HeLa , Humanos , Raios Infravermelhos , Camundongos , Fármacos Fotossensibilizantes/administração & dosagem , Porosidade , Dióxido de Silício/administração & dosagem , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/fisiologia
12.
Adv Mater ; 32(42): e2002439, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32914495

RESUMO

Nanocatalytic therapy, using artificial nanoscale enzyme mimics (nanozymes), is an emerging technology for therapeutic treatment of various malignant tumors. However, the relatively deficient catalytic activity of nanozymes in the tumor microenvironment (TME) restrains their biomedical applications. Here, a versatile and bacteria-like PEG/Ce-Bi@DMSN nanozyme is developed by coating uniform Bi2 S3 nanorods (NRs) with dendritic mesoporous silica (Bi2 S3 @DMSN) and then decorating ultrasmall ceria nanozymes into the large mesopores of Bi2 S3 @DMSN. The nanozymes exhibit dual enzyme-mimic catalytic activities (peroxidase-mimic and catalase-mimic) under acidic conditions that can regulate the TME, that is, simultaneously elevate oxidative stress and relieve hypoxia. In addition, the nanozymes can effectively consume the overexpressed glutathione (GSH) through redox reaction. Photothermal therapy (PTT) is introduced to synergistically improve the dual enzyme-mimicking catalytic activities and depletion of the overexpressed GSH in the tumors by photonic hyperthermia. This is achieved by taking advantage of the desirable light absorbance in the second near-infrared (NIR-II) window of the PEG/Ce-Bi@DMSN nanozymes. Subsequently the reactive oxygen species (ROS)-mediated therapeutic efficiency is significantly improved. Therefore, this study provides a proof of concept of hyperthermia-augmented multi-enzymatic activities of nanozymes for tumor ablation.


Assuntos
Materiais Biomiméticos/farmacologia , Glutationa/metabolismo , Hipertermia Induzida , Nanomedicina/métodos , Nanotubos , Neoplasias/terapia , Materiais Biomiméticos/química , Cério/química , Neoplasias/patologia , Polietilenoglicóis/química , Porosidade , Dióxido de Silício/química
13.
Nanoscale ; 12(37): 19293-19307, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32935695

RESUMO

Hypoxia in tumor cells is regarded as the most crucial cause of clinical drug resistance and radio-resistance; thus, relieving hypoxia of tumor cells is the key to enhancing the efficacy of anticancer therapy. As a gas signal molecule of vasodilatation factors, nitric oxide (NO) can relieve the hypoxia status of tumor cells, thereby, enhancing the sensitivity of tumor cells to radiotherapy. However, considering complications of vascular activity, the level of NO required for radiotherapy sensitization cannot be obtained in vivo. In view of this, we design and fabricate a multifunctional bismuth-based nanotheranostic agent, which is functionalized with S-nitrosothiol and termed Bi-SNO NPs. X-rays break down the S-N bond and simultaneously trigger large amount of NO-releasing (over 60 µM). Moreover, the as-prepared Bi-SNO NPs not only possess the capability of absorbing and converting 808 nm NIR photons into heat for photothermal therapy, but also have the ability to increase X-ray absorption and CT imaging sensitivity. In addition, the collaborative radio-, photothermal-, and gas-therapy of Bi-SNO in vivo was further investigated and remarkable synergistic tumor inhibition was realized. Finally, no obvious toxicity of Bi-SNO NPs was observed in the treated mice within 14 days. Therefore, the Bi-SNO developed in this work is an effective nano-agent for cancer theranostics with well-controlled morphology and uniform size (36 nm), which could serve as a versatile CT imaging-guided combined radio-, photothermal- and gas-therapy nanocomposite with negligible side effects.


Assuntos
Nanopartículas , Nanomedicina Teranóstica , Animais , Bismuto , Camundongos , Óxido Nítrico , Fototerapia , Raios X
14.
J Mater Chem B ; 8(26): 5707-5721, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-32510093

RESUMO

Photodynamic therapy (PDT) is a light-based modality for tumor treatment that involves the generation of reactive oxygen species (ROS) by the combination of light, a photosensitizer, and molecular oxygen. Nevertheless, the therapeutic effects of PDT are limited by hypoxic conditions that worsen with oxygen consumption during the PDT process. Photo/chemodynamic therapy (PCDT) based on the Fenton reaction is one strategy to improve ROS generation, provided a highly effective Fenton reagent is developed. In this research, SiO2@Cu7S4 nanotubes (NTs) were synthesized as a PCDT agent. This double-valence metal-sulfide composite material can react with H2O2 at the tumor site. SiO2@Cu7S4 NTs can produce more ROS than the traditional PDT agents, and besides, they can also be used as a photothermal therapy (PTT) agent. SiO2@Cu7S4 NTs will trigger the PTT effect under 808 nm irradiation and generate a large amount of heat to eradicate cancer cells. This heat will also promote the PCDT effect by increasing the reaction rate. Thus, the SiO2@Cu7S4 NT is a suitable material for PCDT and PTT synergistic oncotherapy. The 808 nm laser is selected as the appropriate excitation source, providing adequate penetration and minimal harm to normal cells. The experimental data presented herein demonstrate the promising photosensitive, Fenton-like, and photothermal performance of SiO2@Cu7S4 NTs. Furthermore, the findings could promote the development of PCDT and PTT synergistic therapy. Thus, this research provides a feasible method to design a single, multifunctional material for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Cobre/farmacologia , Nanotubos/química , Fármacos Fotossensibilizantes/farmacologia , Fototerapia , Dióxido de Silício/farmacologia , Compostos de Enxofre/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cobre/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Lasers , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Tamanho da Partícula , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/química , Espécies Reativas de Oxigênio/metabolismo , Dióxido de Silício/química , Compostos de Enxofre/química , Propriedades de Superfície
15.
ACS Nano ; 13(11): 13144-13160, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31609581

RESUMO

Tumor cell metabolism and tumor blood vessel proliferation are distinct from normal cells. The resulting tumor microenvironment presents a characteristic of hypoxia, which greatly limits the generation of oxygen free radicals and affects the therapeutic effect of photodynamic therapy. Here, we developed an oxygen-independent free radical generated nanosystem (CuFeSe2-AIPH@BSA) with dual-peak absorption in both near-infrared (NIR) regions and utilized it for imaging-guided synergistic treatment. The special absorption provides the nanosystem with high photothermal conversion efficiency and favorably matched photoactivity in both I and II NIR biological windows. Upon NIR light irradiation, the generated heat could prompt AIPH release and decompose to produce oxygen-independent free radicals for killing cancer cells effectively. The contrastive research results show that the enhanced therapeutic efficacy of NIR-II over NIR-I is principally due to its deeper tissue penetration and higher maximum permission exposure that benefits from a longer wavelength. Hyperthermia effect and the production of toxic free radicals upon NIR-II laser illumination are extremely effective in triggering apoptosis and death of cancer cells in the tumor hypoxia microenvironment. The high biocompatibility and excellent anticancer efficiency of CuFeSe2-AIPH@BSA allow it to be an ideal oxygen-independent nanosystem for imaging-guided and NIR-II-mediated synergistic therapy via systemic administration.


Assuntos
Antineoplásicos/farmacologia , Materiais Biocompatíveis/farmacologia , Neoplasias da Mama/tratamento farmacológico , Hipertermia Induzida , Hipóxia/terapia , Fotoquimioterapia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Neoplasias da Mama/patologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Radicais Livres/administração & dosagem , Radicais Livres/química , Radicais Livres/farmacologia , Raios Infravermelhos , Camundongos , Imagem Óptica
16.
Dalton Trans ; 48(17): 5817-5830, 2019 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-30977469

RESUMO

In photothermal therapy (PTT), simultaneous achievement of imaging and hyperthermia mediated by a single laser inevitably risks damaging normal tissues before treatment. Herein, a core-shell-structured GdOF:Yb/Er@(GNRs@BSA) nanohybrid was designed and fabricated by conjugating gold nanorods (GNRs) on the surfaces of GdOF:Yb/Er nanoparticles by a facile procedure. By alternating near-infrared (NIR) light appropriately, high photothermal efficiency for PTT and good up-conversion luminescence (UCL) imaging can be achieved in this structure, which can substantially solve the heat-induced risk during the theranostic process. Furthermore, good biocompatibility and phagocytosis can be realized by modifying bovine serum albumin (BSA) on the surface of the GNRs, and the conjugation of folic acid (FA) endows this nanohybrid with targeting function. It is noted that the size of the GNRs prepared by the one-pot method is much smaller than that by the seed-mediated method, which is not only conducive to uniform heat distribution during intratumoral therapy, but also contributes to the nanohybrid metabolic decomposition and fluorescence tracing after treatment. Moreover, this product can also be utilized as a good magnetic resonance imaging (MRI) and computed tomography (CT) contrast agent, which can provide versatile imaging properties in the field of cancer clinical treatment.


Assuntos
Técnicas de Ablação , Raios Infravermelhos , Luminescência , Nanotubos/química , Fototerapia , Materiais Biocompatíveis/química , Érbio/química , Flúor/química , Gadolínio/química , Humanos , Imageamento por Ressonância Magnética , Imagem Óptica , Oxigênio/química , Soroalbumina Bovina/química
17.
ACS Nano ; 12(11): 11000-11012, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30339353

RESUMO

Therapeutic nanosystems which can be triggered by the distinctive tumor microenvironment possess great selectivity and safety to treat cancers via in situ transformation of nontoxic prodrugs into toxic therapeutic agents. Here, we constructed intelligent, magnetic targeting, and tumor microenvironment-responsive nanocatalysts that can acquire oxidation therapy of cancer via specific reaction at tumor site. The magnetic nanoparticle core of iron carbide-glucose oxidase (Fe5C2-GOD) achieved by physical absorption has a high enzyme payload, and the manganese dioxide (MnO2) nanoshell as an intelligent "gatekeeper" shields GOD from premature leaking until reaching tumor tissue. Fe5C2-GOD@MnO2 nanocatalysts maintained inactive in normal cells upon systemic administration. On the contrary, after endocytosis by tumor cells, tumor acidic microenvironment induced decomposition of MnO2 nanoshell into Mn2+ and O2, meanwhile releasing GOD. Mn2+ could serve as a magnetic resonance imaging (MRI) contrast agent for real-time monitoring treatment process. Then the generated O2 and released GOD in nanocatalysts could effectively exhaust glucose in tumor cells, simultaneously generating plenty of H2O2 which may accelerate the subsequent Fenton reaction catalyzed by the Fe5C2 magnetic core in mildly acidic tumor microenvironments. Finally, we demonstrated the tumor site-specific production of highly toxic hydroxyl radicals for enhanced anticancer therapeutic efficacy while minimizing systemic toxicity in mice.


Assuntos
Antineoplásicos/farmacologia , Magnetoterapia , Nanopartículas/química , Microambiente Tumoral/efeitos dos fármacos , Neoplasias do Colo do Útero/tratamento farmacológico , Neoplasias do Colo do Útero/patologia , Animais , Antineoplásicos/química , Compostos Inorgânicos de Carbono/química , Compostos Inorgânicos de Carbono/farmacologia , Catálise , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Glucose Oxidase/química , Glucose Oxidase/metabolismo , Células HeLa , Humanos , Compostos de Ferro/química , Compostos de Ferro/farmacologia , Imageamento por Ressonância Magnética , Compostos de Manganês/química , Compostos de Manganês/farmacologia , Camundongos , Camundongos Endogâmicos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/patologia , Óxidos/química , Óxidos/farmacologia , Tamanho da Partícula , Propriedades de Superfície
18.
Inorg Chem ; 57(16): 9988-9998, 2018 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-30070830

RESUMO

The strategy of diagnosis-to-therapy to realize the integration of imaging and high antitumor efficiency has become the most promising method. Light-induced therapeutic technologies have drawn considerable interest. However, the limited penetration depth of UV/vis excitation and relatively low efficiency are the main obstacles for its further clinic application. For this concern, we presented a facile method to anchor ultrasmall ZnFe2O4 nanoparticles and upconversion luminescence nanoparticles (UCNPs) on graphene oxide (GO) nanosheets (GO/ZnFe2O4/UCNPs, abbreviated as GZUC). To solve the penetration question, here we introduced Tm3+-doped UCNPs to convert the high-penetrated near-infrared (NIR) light into UV/vis photons to activate the photodynamic process. In this system, the dual phototherapy from GO and ZnFe2O4 has been realized upon NIR laser irradiation. Combined with the photodynamic therapy (PDT) based on Fenton reaction that ZnFe2O4 nanoparticles react with excessive H2O2 in tumor microenvironment to produce toxic hydroxyl radicals (·OH), an excellent anticancer efficiency has been achieved. Furthermore, 4-fold imaging including upconversion luminescence (UCL), computed tomography (CT), magnetic resonance imaging (MRI) and photoacoustic tomography (PAT) has been obtained due to its intrinsic properties, thereby successfully realizing diagnosis-monitored therapy. Our demonstration provided a feasible strategy to solve the main problems in current light-triggered theranostic.

19.
Inorg Chem ; 57(9): 4864-4876, 2018 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-29634255

RESUMO

Multifunctional nanotheranostic agent with high performance for tumor site-specific generation of singlet oxygen (1O2) as well as imaging-guidance is crucial to laser-mediated photodynamic therapy. Here, we introduced a versatile strategy to design a smart nanoplatform using phase change material (PCM) to encapsulate photosensitizer (zinc phthalocyanine, ZnPc) in copper sulfide loaded Fe-doped tantalum oxide (Fe-mTa2O5@CuS) nanoparticles. When irradiated by 808 nm laser, the PCM is melted due to the hyperthermia effect from CuS nanoparticles, inducing the release of ZnPc to produce toxic 1O2 triggered by 650 nm light with very low power density (5 mW/cm2). Then, the produced heat and toxic 1O2 can kill tumor cells in vitro and in vivo effectively. Furthermore, the special properties of Fe-mTa2O5 endow the nanoplatform with excellent computed tomography (CT) and T1-weighted magnetic resonance imaging ( T1-MRI) performance for guiding and real-time monitoring of therapeutic effect. This work presents a feasible way to design smart nanoplatform for controllable generation of heat and 1O2, achieving CT/ T1-MRI dual-modal imaging-guided phototherapy.


Assuntos
Cobre/química , Indóis/química , Ferro/química , Imagem Óptica , Compostos Organometálicos/química , Óxidos/química , Fotoquimioterapia , Tantálio/química , Animais , Sobrevivência Celular/efeitos dos fármacos , Cobre/farmacologia , Células HeLa , Humanos , Indóis/farmacologia , Ferro/farmacologia , Imageamento por Ressonância Magnética , Camundongos , Nanopartículas/química , Neoplasias Experimentais/diagnóstico por imagem , Compostos Organometálicos/farmacologia , Óxidos/farmacologia , Tamanho da Partícula , Propriedades de Superfície , Tantálio/farmacologia , Nanomedicina Teranóstica , Tomografia Computadorizada por Raios X
20.
J Mater Chem B ; 6(48): 8148-8162, 2018 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-32254934

RESUMO

Designing multifunctional nanoplatforms for the purpose of simultaneous theranostic modalities is critical to address the challenges of cancer therapy. Also, single modalities of phototherapy, including photothermal therapy (PTT) and photodynamic therapy (PDT), cannot meet the requirements of highly efficient treatment. Here, a core-shell-shell nanostructure consisting of a core of upconversion nanoparticles (UCNPs), a layer of mesoporous silica with anchored ZnO nanodots, and an outer layer of polypyrrole (PPy) was developed. In the proposed construct, the emitted ultraviolet (UV) light from the UCNPs core upon 980 nm near-infrared light irradiation can trigger the ZnO nanodots to activate ambient O2 molecules around cancerous tissues to produce toxic reactive oxygen species (ROS), realizing the PDT function. On the other hand, the coated PPy layer can concurrently give rise to an obvious heat effect upon NIR light illumination, thus achieving synergistic PDT and PTT effects; this results in excellent anti-tumor efficiency in vitro and in vivo. Furthermore, in hand with the upconversion luminescence (UCL) and computed tomography (CT) imaging derived from the UCNPs core, dual-mode imaging directed cancer therapy has been realized.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA