Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Small ; 19(35): e2301148, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37118853

RESUMO

As an emerging cancer treatment strategy, ferroptosis is greatly restricted by excessive glutathione (GSH) in tumor microenvironment (TME) and low reactive oxygen species (ROS) generation efficiency. Here, this work designs self-assembled copper-alanine nanoparticles (CACG) loaded with glucose oxidase (GOx) and cinnamaldehyde (Cin) for in situ glutathione activated and enzymatic cascade-enhanced ferroptosis and immunotherapy. In response to GSH-rich and acidic TME, CACG allows to effectively co-deliver Cu2+ , Cin, and GOx into tumors. Released Cin consumes GSH through Michael addition, accompanying with the reduction of Cu2+ into Cu+ for further GSH depletion. With the cascade of Cu+ -catalyzed Fenton reactions and enzyme-catalyzed reactions by GOx, CACG could get rid of the restriction of insufficient hydrogen peroxide in TME, leading to a robust and constant generation of ROS. With the high efficiency of GSH depletion and ROS production, ferroptosis is significantly enhanced by CACG in vivo. Moreover, elevated oxidative stress triggers robust immune responses by promoting dendritic cells maturation and T cell infiltration. The in vivo results prove that CACG could efficiently inhibit tumor growth in 4T1 tumor-bearing mouse model without causing obvious systemic toxicity, suggesting the great potential of CACG in enhancing ferroptosis and immunotherapy for effective cancer treatment.


Assuntos
Ferroptose , Nanopartículas , Neoplasias , Animais , Camundongos , Cobre , Espécies Reativas de Oxigênio , Imunoterapia , Glucose Oxidase , Glutationa , Peróxido de Hidrogênio , Microambiente Tumoral , Linhagem Celular Tumoral , Neoplasias/terapia
2.
Adv Sci (Weinh) ; 10(4): e2205480, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36479844

RESUMO

Systematic administration of antibiotics to treat infections often leads to the rapid evolution and spread of multidrug-resistant bacteria. Here, an in situ-formed biotherapeutic gel that controls multidrug-resistant bacterial infections and accelerates wound healing is reported. This biotherapeutic gel is constructed by incorporating stable microbial communities (kombucha) capable of producing antimicrobial substances and organic acids into thermosensitive Pluronic F127 (polyethylene-polypropylene glycol) solutions. Furthermore, it is found that the stable microbial communities-based biotherapeutic gel possesses a broad antimicrobial spectrum and strong antibacterial effects in diverse pathogenic bacteria-derived xenograft infection models, as well as in patient-derived multidrug-resistant bacterial xenograft infection models. The biotherapeutic gel system considerably outperforms the commercial broad-spectrum antibacterial gel (0.1% polyaminopropyl biguanide) in pathogen removal and infected wound healing. Collectively, this biotherapeutic strategy of exploiting stable symbiotic consortiums to repel pathogens provides a paradigm for developing efficient antibacterial biomaterials and overcomes the failure of antibiotics to treat multidrug-resistant bacterial infections.


Assuntos
Anti-Infecciosos , Infecções Bacterianas , Humanos , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bactérias , Poloxaleno/farmacologia , Infecções Bacterianas/tratamento farmacológico
3.
Biomaterials ; 272: 120782, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33819816

RESUMO

Photodynamic therapy (PDT) is widely researched in tumor treatment, but its therapeutic effect is affected by oxygen (O2) concentration of tumor site. Here, we developed a Pd-coordinated π-conjugated extended porphyrin doped porphyrin metal-organic-framework (named as PTP). PTP can achieve near infrared (NIR) O2 concentration ratiometric imaging, solving the problems of short detection wavelengths and influence of self-concentrations. With the NIR excitation wavelength and the ability of higher singlet oxygen (1O2) generation, PTP can induce PDT more effectively. The efficient PDT also mediates cancer immunogenic cell death (ICD), which combines with the immune checkpoint inhibitor αPD-1 to achieve obviously cancer suppression and anti-metastasis effect. This theranostic NIR ratiometric nanoprobe can be used as a pre-evaluation on the outcome of PDT and high-efficient cancer combined treatment system, which will find great potential in tumor diagnosis and treatment.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Neoplasias , Fotoquimioterapia , Porfirinas , Linhagem Celular Tumoral , Humanos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Oxigênio , Fármacos Fotossensibilizantes
4.
ACS Nano ; 14(8): 9848-9860, 2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32658459

RESUMO

Metal carbonyl complexes can readily liberate carbon monoxide (CO) in response to activation stimulus. However, applicability of metal carbonyl complexes is limited because they are unstable under natural ambient conditions of moisture and oxygen. Reported here is the rational design of an iron carbonyl complex delivery nanosystem for the improvement of cancer therapy. We demonstrated that iron pentacarbonyl (Fe(CO)5) can be encapsulated into the cavity of a Au nanocage under an oxygen-free atmosphere and then controllably form iron oxide on the surface of the Au nanocage under aerobic conditions. The formation of iron oxide efficiently avoids the leakage and oxidation of the caged Fe(CO)5. The resulting nanomaterial exhibits excellent safety, biocompatibility, and stability, which can be specifically activated under near-infrared (NIR) irradiation within the tumor environment to generate CO and iron. The released CO causes damage to mitochondria and subsequent initiation of autophagy. More importantly, during autophagy, the nanomaterial that contains iron and iron oxide can accumulate into the autolysosome and result in its destruction. The produced CO and iron show excellent synergistic effects in cancer cells.


Assuntos
Complexos de Coordenação , Compostos de Ferro , Neoplasias , Monóxido de Carbono , Ferro , Neoplasias/tratamento farmacológico
5.
Angew Chem Int Ed Engl ; 59(25): 10087-10094, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31671247

RESUMO

Fluorescent materials exhibiting two-photon induction (TPI) are used for nonlinear optics, bioimaging, and phototherapy. Polymerizations of molecular chromophores to form π-conjugated structures were hindered by the lack of long-range ordering in the structure and strong π-π stacking between the chromophores. Reported here is the rational design of a benzothiadiazole-based covalent organic framework (COF) for promoting TPI and obtaining efficient two-photon induced fluorescence emissions. Characterization and spectroscopic data revealed that the enhancement in TPI performance is attributed to the donor-π-acceptor-π-donor configuration and regular intervals of the chromophores, the large π-conjugation domain, and the long-range order of COF crystals. The crystalline structure of TPI-COF attenuates the π-π stacking interactions between the layers, and overcomes aggregation-caused emission quenching of the chromophores for improving near-infrared two-photon induced fluorescence imaging.


Assuntos
Corantes Fluorescentes/química , Estruturas Metalorgânicas/química , Imagem Óptica/métodos , Animais , Células HeLa , Humanos , Raios Infravermelhos , Camundongos , Camundongos Endogâmicos BALB C , Fótons , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Nat Commun ; 10(1): 3199, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31324770

RESUMO

Most cancer vaccines are unsuccessful in eliciting clinically relevant effects. Without using exogenous antigens and adoptive cells, we show a concept of utilizing biologically reprogrammed cytomembranes of the fused cells (FCs) derived from dendritic cells (DCs) and cancer cells as tumor vaccines. The fusion of immunologically interrelated two types of cells results in strong expression of the whole tumor antigen complexes and the immunological co-stimulatory molecules on cytomembranes (FMs), allowing the nanoparticle-supported FM (NP@FM) to function like antigen presenting cells (APCs) for T cell immunoactivation. Moreover, tumor-antigen bearing NP@FM can be bio-recognized by DCs to induce DC-mediated T cell immunoactivation. The combination of these two immunoactivation pathways offers powerful antitumor immunoresponse. Through mimicking both APCs and cancer cells, this cytomembrane vaccine strategy can develop various vaccines toward multiple tumor types and provide chances for accommodating diverse functions originating from the supporters.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos de Neoplasias/imunologia , Vacinas Anticâncer/imunologia , Membrana Celular/imunologia , Nanopartículas/uso terapêutico , Animais , Fusão Celular , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Feminino , Imunoterapia , Ativação Linfocitária , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T/imunologia , Transcriptoma , Transplante Heterólogo
7.
Adv Mater ; 31(18): e1900499, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30907473

RESUMO

Using the cytomembranes (FMs) of hybrid cells acquired from the fusion of cancer and dendritic cells (DCs), this study offers a biologically derived platform for the combination of immunotherapy and traditional oncotherapy approaches. Due to the immunoactivation implicated in the cellular fusion, FMs can effectively express whole cancer antigens and immunological co-stimulatory molecules for robust immunotherapy. FMs share the tumor's self-targeting character with the parent cancer cells. In bilateral tumor-bearing mouse models, the FM-coated nanophotosensitizer causes durable immunoresponse to inhibit the rebound of primary tumors post-nanophotosensitizer-induced photodynamic therapy (PDT). The FM-induced immunotherapy displays ultrahigh antitumor effects even comparable to that of PDT. On the other hand, PDT toward primary tumors enhances the immunotherapy-caused regression of the irradiation-free distant tumors. Consequently, both the primary and the distant tumors are almost completely eliminated. This tumor-specific immunotherapy-based nanoplatform is potentially expandable to multiple tumor types and readily equipped with diverse functions owing to the flexible nanoparticle options.


Assuntos
Membrana Celular/química , Células Dendríticas/citologia , Imunoterapia , Nanoestruturas/química , Animais , Anticorpos/química , Anticorpos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Receptores de Hialuronatos/imunologia , Estruturas Metalorgânicas/química , Camundongos , Camundongos Nus , Nanoestruturas/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/química , Porfirinas/uso terapêutico , Transplante Heterólogo , Zircônio/química
8.
Biomaterials ; 185: 51-62, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30218836

RESUMO

This study reports a tumor-specific ROS-responsive nanoplatform capable of the combination of nitric oxide (NO)-based gas therapy and sensitized photodynamic therapy (PDT). The nanoplatform is constructed on porous coordination network (PCN), which contains NO donor L-Arg and is concurrently coated with cancer cell membrane (L-Arg@PCN@Mem). Under near infrared light (NIR) irradiation, L-Arg@PCN@Mem produces plenty of reactive oxygen species (ROS) directly for PDT therapy, while a part of ROS take the role of oxidative to converse L-Arg into NO for combined gas therapy. The results indicate that the transformation of ROS to NO can enhance PDT efficacy in hypoxic tumors owing to the ability of NO in freely diffusing into deep hypoxic tumor site. Moreover, homologous targeting function originated from the coating of cancer cells membrane further improves the tumor treatment effect owing to the biotargeting toward homologous tumors. This L-Arg@PCN@Mem nanoplatform provides a new therapy paradigm of overcoming the hypoxia barrier of tumor therapy, and holds great potential for the treatment of tumor and NO-related diseases.


Assuntos
Nanoestruturas/administração & dosagem , Neoplasias/tratamento farmacológico , Doadores de Óxido Nítrico/administração & dosagem , Óxido Nítrico/metabolismo , Fármacos Fotossensibilizantes/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Nanoestruturas/uso terapêutico , Neoplasias/metabolismo , Neoplasias/patologia , Doadores de Óxido Nítrico/uso terapêutico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Porosidade , Hipóxia Tumoral/efeitos dos fármacos
9.
Small ; 14(28): e1801120, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29882235

RESUMO

Hypoxia is reported to participate in tumor progression, promote drug resistance, and immune escape within tumor microenvironment, and thus impair therapeutic effects including the chemotherapy and advanced immunotherapy. Here, a multifunctional biomimetic core-shell nanoplatform is reported for improving synergetic chemotherapy and immunotherapy. Based on the properties including good biodegradability and functionalities, the pH-sensitive zeolitic imidazolate framework 8 embedded with catalase and doxorubicin constructs the core and serves as an oxygen generator and drug reservoir. Murine melanoma cell membrane coating on the core provides tumor targeting ability and elicits an immune response due to abundance of antigens. It is demonstrated that this biomimetic core-shell nanoplatform with oxygen generation can be partial to accumulate in tumor and downregulate the expression of hypoxia-inducible factor 1α, which can further enhance the therapeutic effects of chemotherapy and reduce the expression of programmed death ligand 1 (PD-L1). Combined with immune checkpoints blockade therapy by programmed death 1 (PD-1) antibody, the dual inhibition of the PD-1/PD-L1 axis elicits significant immune response and presents a robust effect in lengthening tumor recurrent time and inhibiting tumor metastasis. Consequently, the multifunctional nanoplatform provides a potential strategy of synergetic chemotherapy and immunotherapy.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Biomimética/métodos , Receptor de Morte Celular Programada 1/metabolismo , Transdução de Sinais , Hipóxia Tumoral/efeitos dos fármacos , Animais , Linfócitos T CD8-Positivos/metabolismo , Catalase/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Citocinas/metabolismo , Doxorrubicina/farmacologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imidazóis/química , Camundongos Endogâmicos C57BL , Nanopartículas/química , Nanopartículas/ultraestrutura , Neoplasias/imunologia , Neoplasias/patologia , Oxigênio/farmacologia , Zeolitas/química
10.
ACS Nano ; 12(5): 4630-4640, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29584395

RESUMO

We report on the benzoporphyrin-based metal-organic framework (TBP-MOF), with 10-connected Zr6 cluster and much improved photophysical properties over the traditional porphyrin-based MOFs. It was found that TBP-MOF exhibited red-shifted absorption bands and strong near-infrared luminescence for bioimaging, whereas the π-extended benzoporphyrin-based linkers of TBP-MOF facilitated 1O2 generation to enhance O2-dependent photodynamic therapy (PDT). It was demonstrated that poly(ethylene glycol)-modified nanoscale TBP-MOF (TBP-nMOF) can be used as an effective PDT agent under hypoxic tumor microenvironment. We also elucidated that the low O2-dependent PDT of TBP-nMOF in combination with αPD-1 checkpoint blockade therapy can not only suppress the growth of primary tumor, but also stimulate an antitumor immune response for inhibiting metastatic tumor growth. We believe this TBP-nMOF has great potential to serve as an efficient photosensitizer for PDT and cancer immunotherapy.


Assuntos
Antineoplásicos/química , Estruturas Metalorgânicas/química , Nanopartículas/química , Metástase Neoplásica/terapia , Fármacos Fotossensibilizantes/química , Porfirinas/química , Zircônio/química , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Antígeno B7-H1/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Xenoenxertos , Humanos , Imunoterapia , Estruturas Metalorgânicas/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Polietilenoglicóis/química , Porfirinas/farmacologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Oxigênio Singlete/metabolismo , Distribuição Tecidual , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA