Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
ACS Nano ; 18(5): 4189-4204, 2024 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-38193384

RESUMO

cGAS-STING signaling plays a critical role in radiotherapy (RT)-mediated immunomodulation. However, RT alone is insufficient to sustain STING activation in tumors under a safe X-ray dose. Here, we propose a radiosensitization cooperated with cGAS stimulation strategy by engineering a core-shell structured nanosized radiosensitizer-based cGAS-STING agonist, which is constituted with the hafnium oxide (HfO2) core and the manganese oxide (MnO2) shell. HfO2-mediated radiosensitization enhances immunogenic cell death to afford tumor associated antigens and adequate cytosolic dsDNA, while the GSH-degradable MnO2 sustainably releases Mn2+ in tumors to improve the recognition sensitization of cGAS. The synchronization of sustained Mn2+ supply with cumulative cytosolic dsDNA damage synergistically augments the cGAS-STING activation in irradiated tumors, thereby enhancing RT-triggered local and system effects when combined with an immune checkpoint inhibitor. Therefore, the synchronous radiosensitization with sustained STING activation is demonstrated as a potent immunostimulation strategy to optimize cancer radio-immuotherapy.


Assuntos
Háfnio , Compostos de Manganês , Neoplasias , Humanos , Compostos de Manganês/farmacologia , Óxidos/farmacologia , Óxidos/uso terapêutico , Imunoterapia , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Nucleotidiltransferases
2.
J Colloid Interface Sci ; 660: 869-884, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38277843

RESUMO

Infiltration and activation of intratumoral T lymphocytes are critical for immune checkpoint blockade (ICB) therapy. Unfortunately, the low tumor immunogenicity and immunosuppressive tumor microenvironment (TME) induced by tumor metabolic reprogramming cooperatively hinder the ICB efficacy. Herein, we engineered a lactate-depleting MOF-based catalytic nanoplatform (LOX@ZIF-8@MPN), encapsulating lactate oxidase (LOX) within zeolitic imidazolate framework-8 (ZIF-8) coupled with a coating of metal polyphenol network (MPN) to reinforce T cell response based on a "two birds with one stone" strategy. LOX could catalyze the degradation of the immunosuppressive lactate to promote vascular normalization, facilitating T cell infiltration. On the other hand, hydrogen peroxide (H2O2) produced during lactate depletion can be transformed into anti-tumor hydroxyl radical (•OH) by the autocatalytic MPN-based Fenton nanosystem to trigger immunogenic cell death (ICD), which largely improved the tumor immunogenicity. The combination of ICD and vascular normalization presents a better synergistic immunopotentiation with anti-PD1, inducing robust anti-tumor immunity in primary tumors and recurrent malignancies. Collectively, our results demonstrate that the concurrent depletion of lactate to reverse the immunosuppressive TME and utilization of the by-product from lactate degradation via cascade catalysis promotes T cell response and thus improves the effectiveness of ICB therapy.


Assuntos
Estruturas Metalorgânicas , Neoplasias , Humanos , Ácido Láctico/farmacologia , Estruturas Metalorgânicas/farmacologia , Peróxido de Hidrogênio/farmacologia , Linfócitos T , Imunoterapia , Linhagem Celular Tumoral , Microambiente Tumoral
3.
ACS Nano ; 17(14): 13195-13210, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37256771

RESUMO

Radiotherapy (RT) is one of the important clinical treatments for local control of triple-negative breast cancer (TNBC), but radioresistance still exists. Ferroptosis has been recognized as a natural barrier for cancer progression and represents a significant role of RT-mediated anticancer effects, while the simultaneous activation of ferroptosis defensive system during RT limits the synergistic effect between RT and ferroptosis. Herein, we engineered a tumor microenvironment (TME) degradable nanohybrid with a dual radiosensitization manner to combine ferroptosis induction and high-Z effect based on metal-organic frameworks for ferroptosis-augmented RT of TNBC. The encapsulated l-buthionine-sulfoximine (BSO) could inhibit glutathione (GSH) biosynthesis for glutathione peroxidase 4 (GPX4) inactivation to break down the ferroptosis defensive system, and the delivered ferrous ions could act as a powerful ferroptosis executor via triggering the Fenton reaction; the combination of them induces potent ferroptosis, which could synergize with the surface decorated Gold (Au) NPs-mediated radiosensitization to improve RT efficacy. In vivo antitumor results revealed that the nanohybrid could significantly improve the therapeutic efficacy and antimetastasis efficiency based on the combinational mechanism between ferroptosis and RT. This work thus demonstrated that combining RT with efficient ferroptosis induction through nanotechnology was a feasible and promising strategy for TNBC treatment.


Assuntos
Ferroptose , Neoplasias de Mama Triplo Negativas , Humanos , Anestésicos Locais , Butionina Sulfoximina , Fibrinolíticos , Glutationa , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Acta Biomater ; 141: 364-373, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35063709

RESUMO

Tumor vasculature-targeting therapy either using angiogenesis inhibitors or vascular disrupting agents offers an important new avenue for cancer therapy. In this work, a tumor-specific catalytic nanomedicine for enhanced tumor ablation accompanied with tumor vasculature disruption and angiogenesis inhibition was developed through a cascade reaction with enzyme glucose oxidase (GOD) modified on Fe-based metal organic framework (Fe-MOF) coupled with anti-VEGFR2.The GOD enzyme could catalyze the intratumoral glucose decomposition to trigger tumor starvation and yet provide abundant hydrogen peroxide as the substrate for Fenton-like reaction catalyzed by Fe-MOF to produce sufficient highly toxic hydroxyl radicals for enhanced chemodynamic therapy and instantly attacked tumor vascular endothelial cells to destroy the existing vasculature, while the anti-VEGFR2 antibody guided the nanohybrids to target blood vessels and block the VEGF-VEGFR2 connection to prevent angiogenesis. Both in vitro and in vivo results demonstrated the smart nanohybrids could cause the tumor cell apoptosis and vasculature disruption, and exhibited enhanced tumor regression in A549 xenograft tumor-bearing mice model. This study suggested that synergistic targeting tumor growth and its vasculature network would be more promising for curing solid tumors. STATEMENT OF SIGNIFICANCE: Cooperative destruction of tumor cells and tumor vasculature offers a potential avenue for cancer therapy. Under this premise, a tumor-specific catalytic nanomedicine for enhanced tumor ablation accompanied with tumor vasculature disruption and new angiogenesis inhibition was developed through a cascade reaction with glucose oxidase modified on the surface of iron-based metal organic framework coupled with VEGFR2 antibody. The resulting data demonstrated that a therapeutic regimen targeting tumor growth as well as its vasculature with both existing vasculature disruption and neovasculature inhibition would be more potential for complete eradication of tumors.


Assuntos
Estruturas Metalorgânicas , Neoplasias , Animais , Catálise , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Glucose Oxidase/química , Humanos , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
5.
ACS Appl Mater Interfaces ; 13(45): 53504-53518, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34704726

RESUMO

With the recent success of immune checkpoint blockade (ICB) in cancer immunotherapy, there has been renewed interest in evaluating the combination of ICB inhibitors with radiotherapy (RT) in clinical trials in view of the localized RT-initiated vaccination effect, which can be augmented further by systemic immune-stimulating agents. Unfortunately, traditional RT/ICB accompanies severe toxicity from high-dose ionizing irradiation and low response rate from RT-aggravated immunosuppression, among which M2-type tumor-associated macrophages (TAMs) play an important role. Herein, CpG-decorated gold (Au) nanoparticles (CpG@Au NPs) were fabricated to improve the RT/ICB efficacy by immune modulation under low-dose X-ray exposure, where Au NPs served as radioenhancers to minimize the radiotoxicity, and yet acted as nanocarriers to deliver CpG, a toll-like receptor 9 agonist, to re-educate immunosuppressive M2 TAMs to immunostimulatory M1 counterparts, thus arousing innate immunity and meanwhile priming T cell activation. When combined with an anti-programmed death 1 antibody, irradiated CpG@Au led to consistent abscopal responses that efficiently suppressed distant tumors in a bilateral GL261 tumor-bearing model. This work thus demonstrates that CpG@Au-mediated macrophage reeducation could efficiently modulate the tumor-immune microenvironment for synergistic RT/ICB.


Assuntos
Glioma/terapia , Ouro/farmacologia , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia , Nanocompostos/química , Oligodesoxirribonucleotídeos/farmacologia , Macrófagos Associados a Tumor/efeitos dos fármacos , Animais , Ouro/química , Inibidores de Checkpoint Imunológico/química , Camundongos , Oligodesoxirribonucleotídeos/química , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
6.
Mol Med Rep ; 19(5): 4475-4483, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30896885

RESUMO

microRNAs (miRNAs) are frequently aberrantly expressed in colorectal cancer (CRC) and are considered to serve a critical role in the onset and development of CRC by binding to its target transcription factor. The aim of the present study was to examine the role of miRNA (miR)­183­5p in the proliferation, invasion and migration of CRC cells, in addition to its underlying mechanism. Reverse transcription­quantitative polymerase chain reaction analysis was used to detect the expression level of miR­183­5p. MTT and Transwell assays were performed to examine proliferation and invasion in SW620 cells. Western blot analysis was performed to determine the protein expression of reticulocalbin­2 (RCN2), matrix metalloproteinase­2, ß­catenin, cyclin D1 and c­Myc. miR­183­5p expression was significantly upregulated in the CRC tissues compared with adjacent normal tissues. In addition, the inhibition of miR­183­5p suppressed proliferation, invasion and migration in SW620 cells. miR­183­5p downregulation or overexpression regulated the CRC cell cycle, invasion and migration by modulating RCN2 expression. Furthermore, the Wnt/ß­catenin pathway was observed to be involved in the inhibitory effect of miR­183­5p downregulation in CRC cell proliferation, invasion and migration. These results provided evidence that the downregulation of miR­183­5p inhibits CRC proliferation and invasion by regulating the RCN2/Wnt/ß­catenin pathway. miR­183­5p and RCN2 may serve an important role in the molecular etiology of CRC and have potential applications in CRC treatment.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Neoplasias Colorretais/patologia , MicroRNAs/metabolismo , Via de Sinalização Wnt , Regiões 3' não Traduzidas , Adulto , Antagomirs/metabolismo , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Movimento Celular , Neoplasias Colorretais/metabolismo , Regulação para Baixo , Feminino , Humanos , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Pessoa de Meia-Idade , Interferência de RNA , RNA Interferente Pequeno/metabolismo
7.
Adv Healthc Mater ; 7(10): e1800022, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29508557

RESUMO

Owing to the progressive development of metal-organic frameworks (MOFs) synthetic processes and their unique characters associated with the excellent performance-selectable composition, tunable pore scale, large surface area, and good thermal stability, MOFs have captured the interest and the imagination of an increasing number of scientists working in different fields. In the area of biomedical applications, MOFs are especially involved in sensing, molecular imaging, and drug delivery, with strong contributions to the whole nanomedicine area. Recently, these materials have been scaled down to nanometer sizes with the advancement of chemical synthesis gradually reaching an adjustable level. This review mainly discusses and summarizes the general synthesis, properties, and biomedical applications of nanoscaled MOFs and their composites in biosensing, imaging, and cancer therapy within the latest three years. The remaining challenges and future opportunities in this field, in terms of processing techniques, maximizing composite properties, and prospects for clinical applications, are also indicated.


Assuntos
Técnicas Biossensoriais/métodos , Sistemas de Liberação de Medicamentos/métodos , Estruturas Metalorgânicas , Imagem Molecular/métodos , Nanoestruturas , Neoplasias , Animais , Humanos , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/uso terapêutico , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA