Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Small ; 18(9): e2105465, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34918449

RESUMO

Nanozyme-based chemodynamic therapy (CDT) for fighting bacterial infections faces several major obstacles including low hydrogen peroxide (H2 O2 ) level, over-expressed glutathione (GSH) in infected sites, and inevitable damage to healthy tissue with abundant nonlocalized nanozymes. Herein, a smart ultrasmall Fe3 O4 -decorated polydopamine (PDA/Fe3 O4 ) hybrid nanozyme is demonstrated that continuously converts oxygen into highly toxic hydroxyl radical (•OH) via GSH-depleted cascade redox reactions for CDT-mediated bacterial elimination and intensive wound disinfection. In this system, photonic hyperthermia of PDA/Fe3 O4 nanozymes can not only directly damage bacteria, but also improve the horseradish peroxidase-like activity of Fe3 O4 decorated for CDT. Surprisingly, through photothermal-enhanced cascade catalytic reactions, PDA/Fe3 O4 nanozymes can consume endogenous GSH for disrupting cellular redox homeostasis and simultaneously provide abundant H2 O2 for improving •OH generation, ultimately enhancing the antibacterial performance of CDT. Such PDA/Fe3 O4 can bind with bacterial cells, and reveals excellent antibacterial property against both Staphylococcus aureus and Escherichia coli. Most interestingly, PDA/Fe3 O4 nanozymes can be strongly retained in infected sites by an external magnet for localized long-term in vivo CDT and show minimal toxicity to healthy tissues and organs. This work presents an effective strategy to magnetically retain the therapeutic nanozymes in infected sites for highly efficient CDT with good biosafety.


Assuntos
Radical Hidroxila , Oxigênio , Desinfecção , Glutationa , Peróxido de Hidrogênio , Indóis , Oxirredução , Polímeros
2.
Small ; 18(29): e2202964, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35717674

RESUMO

Nanocatalytic medicine is a burgeoning disease treatment model with high specificity and biosafety in which the nanocatalyst is the core of driving catalytic reaction to generate therapeutic outcomes. However, the robust defense systems in the pathological region would counteract nanocatalyst-initiated therapeutics. Here, a Cu-doped polypyrrole is innovatively developed by a facile oxidative polymerization reaction, which exhibits intriguing multi-catalytic activities, including catalyzing H2 O2 to generate O2 and · OH, and consuming reduced glutathione by a Cu(II)-Cu(I) transition approach. By decorating with sonosensitizers and DSPE-PEG, the obtained CuPPy-TP plus US irradiation can induce severe oxidative damage to tumor cells by amplifying oxidative stress and simultaneously relieving antioxidant capacity in tumors based on the highly effective sonochemical and redox reactions. The notable tumor-specific biodegradability, remarkable cell apoptosis in vitro, and tumor suppression in vivo are demonstrated in this work, which not only present a promising biocompatible antitumor nanocatalyst but also broaden the perspective in oxidative stress-based antitumor therapy.


Assuntos
Polímeros , Pirróis , Catálise , Linhagem Celular Tumoral , Peróxido de Hidrogênio/farmacologia , Polímeros/farmacologia , Microambiente Tumoral
3.
Small ; 17(7): e2005865, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33502106

RESUMO

Nanozyme-based chemodynamic therapy (CDT) has emerged as an effective cancer treatment because of its low side effects and without the requirement of exogenous energy. The therapeutic effect of CDT highlights the pivotal importance of active sites, H2 O2 supplement and the glutathione (GSH) depletion of a nanozyme. The construction of a single kind of catalyst with multiple functions for the enhanced CDT is still a big challenge. In this work, seven types of bimetallic nanoparticles are synthesized using a metal-organic framework (MOF) as a stable host instead of a Fenton or Fenton-like ions supplier. Among them, Cu-Pd@MIL-101 with an alloy loading of 9.5 wt% modified by PEG (9.5% CPMP) is found to exhibit the highest peroxidase (POD) like activity combined with a superoxide dismutase (SOD) mimic activity and the function of GSH depletion. The in vivo results suggest that the stable and ultrafine nanoparticles possess favorable CDT effect for tumor and good biosafety as well as biocompatibility. This work has provided a credible strategy to construct nanozymes with an excellent activity and may pave a new way for the design of enhanced tumor CDT treatment.


Assuntos
Estruturas Metalorgânicas , Nanopartículas , Ligas , Linhagem Celular Tumoral , Peróxido de Hidrogênio
4.
Small ; 15(51): e1904870, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31750615

RESUMO

Tumor hypoxia significantly diminishes the efficacy of reactive oxygen species (ROS)-based therapy, mainly because the generation of ROS is highly oxygen dependent. Recently reported hypoxia-irrelevant radical initiators (AIBIs) exhibit promising potential for cancer therapy under different oxygen tensions. However, overexpressed glutathione (GSH) in cancer cells would potently scavenge the free radicals produced from AIBI before their arrival to the specific site and dramatically limit the therapeutic efficacy. A synergistic antitumor platform (MoS2 @AIBI-PCM nanoflowers) is constructed by incorporating polyethylene-glycol-functionalized molybdenum disulfide (PEG-MoS2 ) nanoflowers with azo initiator and phase-change material (PCM). Under near-infrared laser (NIR) irradiation, the photothermal feature of PEG-MoS2 induces the decomposition of AIBI to produce free radicals. Furthermore, PEG-MoS2 can facilitate GSH oxidation without releasing toxic metal ions, greatly promoting tumor apoptosis and avoiding the introduction of toxic metal ions. This is the first example of the use of intelligent MoS2 -based nanoflowers as a benign GSH scavenger for enhanced cancer treatment.


Assuntos
Dissulfetos/química , Glutationa/química , Molibdênio/química , Neoplasias/terapia , Linhagem Celular Tumoral , Radicais Livres/química , Humanos , Polietilenoglicóis/química , Espécies Reativas de Oxigênio
5.
Adv Healthc Mater ; 13(22): e2400591, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38861753

RESUMO

Calcium overload can lead to tumor cell death. However, because of the powerful calcium channel excretory system within tumor cells, simplistic calcium overloads do not allow for an effective antitumor therapy. Hence, the nanoparticles are created with polyethylene glycol (PEG) donor-modified calcium phosphate (CaP)-coated, manganese-doped hollow mesopores Prussian blue (MMPB) encapsulating glucose oxidase (GOx), called GOx@MMPB@CaP-PEG (GMCP). GMCP with a three-mode enhancement of intratumor reactive oxygen species (ROS) levels is designed to increase the efficiency of the intracellular calcium overload in tumor cells to enhance its anticancer efficacy. The released exogenous Ca2+ and the production of cytotoxic ROS resulting from the perfect circulation of the three-mode ROS outbreak generation that Fenton/Fenton-like reaction and consumption of glutathione from Fe2+/Fe3+and Mn2+/Mn3+ circle, and amelioration of hypoxia from MMPB-guided and GOx-mediated starvation therapy. Photothermal efficacy-induced heat generation owing to MMPB accelerates the above reactions. Furthermore, abundant ROS contribute to damage to mitochondria, and the calcium channels of efflux Ca2+ are inhibited, resulting in a calcium overload. Calcium overload further increases ROS levels and promotes apoptosis of tumor cells to achieve excellent therapy.


Assuntos
Fosfatos de Cálcio , Cálcio , Ferrocianetos , Nanocompostos , Espécies Reativas de Oxigênio , Ferrocianetos/química , Espécies Reativas de Oxigênio/metabolismo , Humanos , Cálcio/metabolismo , Animais , Nanocompostos/química , Camundongos , Fosfatos de Cálcio/química , Linhagem Celular Tumoral , Glucose Oxidase/metabolismo , Glucose Oxidase/química , Camundongos Endogâmicos BALB C , Polietilenoglicóis/química , Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/terapia
6.
Adv Healthc Mater ; 13(17): e2303896, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38551494

RESUMO

Glutathione (GSH) depletion-induced ferroptosis has emerged as a promising treatment for malignant cancer. It works by inactivating glutathione peroxidase 4 (GPX4) and facilitating lipid peroxidation. However, effectively delivering inducers and depleting intracellular GSH remains challenging due to the short half-lives and high hydrophobicity of small-molecule ferroptosis inducers. These inducers often require additional carriers. Herein, diselenide-containing polymers can consume GSH to induce ferroptosis for pancreatic cancer therapy. The diselenide bonds are controllably built into the backbone of the polycarbonate with a targeting peptide CRGD (Cys-Arg-Gly-Asp), which allows for self-assembly into stable nanoparticles (denoted CRNSe) for self-delivery. Significantly, at a concentration of 12 µg mL-1, CRNSe binds to the active site cysteine of GSH resulting in a thorough depletion of GSH. In contrast, the disulfide-containing analog only causes a slight decrease in GSH level. Moreover, the depletion of GSH inactivates GPX4, ultimately inducing ferroptosis due to the accumulation of lipid peroxide in BxPC-3 cells. Both in vitro and in vivo studies have demonstrated that CRNSe exhibits potent tumor suppressive ability with few side effects on normal tissue. This study validates the anti-tumor mechanism of diselenide-containing polymers in addition to apoptosis and also provides a new strategy for inherently inducing ferroptosis in cancer therapy.


Assuntos
Ferroptose , Glutationa , Ferroptose/efeitos dos fármacos , Humanos , Glutationa/metabolismo , Animais , Linhagem Celular Tumoral , Camundongos , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Nanopartículas/química , Antineoplásicos/farmacologia , Antineoplásicos/química , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Polímeros/química , Polímeros/farmacologia , Camundongos Nus , Cimento de Policarboxilato/química , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Camundongos Endogâmicos BALB C
7.
ACS Nano ; 18(26): 17267-17281, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38871478

RESUMO

Intrinsic or acquired resistance to chemical drugs severely limits their therapeutic efficacy in cancer treatment. Various intracellular antioxidant molecules, particularly glutathione (GSH), play a crucial role in maintaining intracellular redox homeostasis by mitigating the overproduced reactive oxygen species (ROS) due to rapid cell proliferation. Notably, these antioxidants also eliminate chemical-drug-induced ROS, eventually diminishing their cytotoxicity and rendering them less effective. In this study, we combined erastin, a GSH biosynthesis inhibitor, with 2'-deoxy-5-fluorouridine 5'-monophosphate sodium salt (FdUMP), an ROS-based drug, to effectively disrupt intracellular redox homeostasis and reverse chemotherapy resistance. Therefore, efficient ferroptosis and apoptosis were simultaneously induced for enhanced antitumor effects. Additionally, we employed small interfering RNA targeting PD-L1 (siPD-L1) as a third agent to block immune-checkpoint recognition by CD8+ T cells. The highly immunogenic cell peroxidates or damage-associated molecular patterns (DAMPs) induced by erastin acted synergistically with downregulated PD-L1 to enhance the antitumor effects. To codeliver these three drugs simultaneously and efficiently, we designed GE11 peptide-modified lipid nanoparticles (LNPs) containing calcium phosphate cores to achieve high encapsulation efficiencies. In vitro studies verified its enhanced cytotoxicity, efficient intracellular ROS induction and GSH/GPX4 downregulation, substantial lipid peroxidation product accumulation, and mitochondrial depolarization. In vivo, this formulation effectively accumulated at tumor sites and achieved significant tumor inhibition in subcutaneous colon cancer (CRC) mouse models with a maximum tumor inhibition rate of 83.89% at a relatively low dose. Overall, a strategy to overcome clinical drug resistance was verified in this study by depleting GSH and activating adaptive immunity.


Assuntos
Antineoplásicos , Apoptose , Antígeno B7-H1 , Regulação para Baixo , Ferroptose , Nanopartículas , Ferroptose/efeitos dos fármacos , Animais , Humanos , Camundongos , Nanopartículas/química , Antígeno B7-H1/metabolismo , Antígeno B7-H1/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Antineoplásicos/farmacologia , Antineoplásicos/química , Regulação para Baixo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Lipídeos/química , Proliferação de Células/efeitos dos fármacos , Feminino , Ensaios de Seleção de Medicamentos Antitumorais , Linhagem Celular Tumoral , Lipossomos
8.
J Pharm Sci ; 113(8): 2258-2267, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38508340

RESUMO

Vitamin K derivatives such as menadione (MD) have been recognized as promising redox-modulating and chemosensitizing agents for anticancer therapy, however, their cellular activities in peptide-targeted nanocarriers have not been elucidated to date. This study provides the guidelines for developing MD-loaded solid lipid nanoparticles (SLN) modified with extracellular matrix (ECM)-derived peptides. Relationships between RGD peptide concentration and changes in DLS characteristics as well as accumulation of SLN in cancer cells were revealed to adjust the peptide-lipid ratio. SLN system maintained adequate nanoparticle concentration and low dispersity after introduction of MD and MD/RGD, whereas formulated MD was protected from immediate conjugation with reduced glutathione (GSH). RGD-modified MD-containing SLN showed enhanced prooxidant, GSH-depleting and cytotoxic activities toward PC-3 prostate cancer cells attributed to improved cellular pharmacokinetics of the targeted formulation. Furthermore, this formulation effectively sensitized PC-3 cells and OVCAR-4 ovarian cancer cells to free doxorubicin and cisplatin so that cell growth was inhibited by MD-drug composition at nontoxic concentrations of the ingredients. These results provide an important background for further improving chemotherapeutic methods based on combination of conventional cytostatics with peptide-targeted SLN formulations of MD.


Assuntos
Antineoplásicos , Doxorrubicina , Lipídeos , Nanopartículas , Oligopeptídeos , Vitamina K 3 , Humanos , Nanopartículas/química , Vitamina K 3/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Doxorrubicina/química , Oligopeptídeos/química , Oligopeptídeos/farmacologia , Lipídeos/química , Masculino , Portadores de Fármacos/química , Feminino , Glutationa/metabolismo , Cisplatino/farmacologia , Cisplatino/administração & dosagem , Células PC-3 , Lipossomos
9.
J Control Release ; 358: 555-565, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37182804

RESUMO

The amplification of reactive oxygen species (ROS) generation and glutathione (GSH) depletion in cancer cells represents a promising strategy to disrupt redox homeostasis for cancer therapy. Quinone methide and its analogs (QM) have recently been recognized as potential GSH scavengers for anticancer applications; however, an effective QM prodrug is yet to be developed. In this study, we prepare a self-immolative polymeric prodrug (SPP), which could be selectively degraded to generate large quantities of QMs in cancer cells during the spontaneous stepwise head-to-tail degradation of SPP. The amphiphilic SPP is self-assembled into nano-sized micelles, allowing for encapsulating 2-methoxy-ß-estradiol (2ME), an anticancer drug that produces a large amount of intracellular ROS. When SPP@2ME, as the cascade-amplified prodrug, is treated on the cancer cells, 2ME is rapidly released at the ROS-rich intracellular environment by degradation of SPP, thus generating more ROS that triggers the degradation of more SPP chains. Such a domino-like cascade-amplified feedback loop significantly amplifies oxidative stress and disrupts the redox homeostasis in cancer cells. This unique strategy provides synergistic anticancer therapeutic efficacy and demonstrates an important perception in innovative and precise nanomedicine.


Assuntos
Antineoplásicos , Nanopartículas , Neoplasias , Pró-Fármacos , Espécies Reativas de Oxigênio/metabolismo , Polímeros/metabolismo , Oxirredução , Glutationa/metabolismo , Linhagem Celular Tumoral
10.
Biomater Adv ; 133: 112616, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35525734

RESUMO

Photodynamic therapy (PDT) is a promising non-invasive and selective cancer treatment. However, its efficacy is curtailed by tumor hypoxia and high levels of glutathione (GSH) in the tumor and addressing both limitations simultaneously remain challenging. Here, an all-in-one nanoplatform was designed using a GSH-responsive nitric oxide (NO) nano-prodrug that synchronously depletes GSH and relieves hypoxia in tumors, enhancing PDT efficacy. The nano-prodrug PEG-PAMAM-PA/SNO was prepared by integrating the GSH-sensitive NO and pheophorbide A (PA) prodrugs N-acetyl-d-penicillamine thiolactone and PAMAM-PA into polyethylene glycol (PEG), and the NPPA/NO and NPPA were then obtained through nanoprecipitation method. This nanoplatform depletes the intracellular antioxidant, GSH, by integrating GSH-responsive NO prodrug and generating NO that relaxes blood vessels, thereby relieving tumor hypoxia and defeating antioxidant defense system in tumor, while PEGylated PAMAM dendrimers have abundant surface functional groups and can greatly prolong their circulation lifetime in the bloodstream. These effects make this GSH-activatable NO nano-prodrug platform an appealing strategy for enhancing PDT's antitumor effects.


Assuntos
Neoplasias , Fotoquimioterapia , Pró-Fármacos , Antioxidantes , Glutationa , Humanos , Hipóxia/tratamento farmacológico , Neoplasias/tratamento farmacológico , Óxido Nítrico , Polietilenoglicóis , Pró-Fármacos/farmacologia
11.
J Colloid Interface Sci ; 612: 246-260, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-34995863

RESUMO

Cancer phototherapy has attracted increasing attention for its effectiveness, relatively low side effect, and noninvasiveness. The combination of photothermal therapy (PTT) and photodynamic therapy (PDT) has been shown to exhibit promising prospects in cancer treatment. However, the tumor hypoxia, high level of intracellular glutathione (GSH), and insufficient photosensitizer uptake significantly limit the PDT efficacy. In this work, we combine oxygen supply, GSH depletion, and tumor targeting in one nanoplatform, folate-decorated mesoporous polydopamine nanoparticles (FA-MPPD) co-loaded with new indocyanine green (IR-820) and perfluorooctane (PFO) (IR-820/PFO@FA-MPPD), to overcome the PDT resistance for enhanced cancer PDT/PTT. IR-820/PFO@FA-MPPD exhibit efficient singlet oxygen generation and photothermal effect under 808 nm laser irradiation, GSH-promoted IR-820 release, and efficient cellular uptake, resulting in high intracellular reactive oxygen species (ROS) level under 808 nm laser irradiation and strong photocytotoxicity in vitro. Following intratumoral injection, IR-820/PFO@FA-MPPD can relieve tumor hypoxia sustainably by PFO-mediated oxygen transport and deplete intracellular GSH by the Michael addition reaction, which boost the PDT effect and lead to the most potent antitumor effect upon 808 nm laser irradiation. The multifunctional IR-820/PFO@FA-MPPD developed in this work offer a relatively simple and effective strategy to potentiate PDT for efficient cancer phototherapy.


Assuntos
Nanopartículas Multifuncionais , Nanopartículas , Neoplasias , Fotoquimioterapia , Linhagem Celular Tumoral , Indóis , Neoplasias/terapia , Fármacos Fotossensibilizantes , Fototerapia , Polímeros
12.
ACS Appl Mater Interfaces ; 13(32): 38127-38137, 2021 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-34347422

RESUMO

Combination therapy has attracted extensive interest in alleviating the shortcomings of monotherapy and enhancing the treatment efficacy. In this work, hollow mesoporous silica nanoparticles (HMSNs) play the role of nanocarriers in the delivery of Cu(II)-doped polydopamine (PDA), termed as HMSNs@PDA-Cu, for synergistic therapy. PDA acts as a traditional photothermal agent to realize photothermal treatment (PTT). Chemodynamic therapy (CDT) is realized by the reaction of Cu(II) with intracellular glutathione (GSH), and subsequently, the generated Cu(I) reacts with H2O2 to produce toxic hydroxyl radical (•OH) through a Fenton-like reaction. The photothermal performance of PDA is improved after its coordination with Cu(II). On the other hand, PDA exhibits superoxide dismutase (SOD)-mimicking activity. PDA converts O2•- to H2O2 and improves the production of H2O2, which promotes the therapeutic effect of CDT. Moreover, the high temperature caused by PTT further enhances the yield of •OH for CDT. This nanotheranostic platform perfectly applied to the tumor depletion of mice, presenting great potential for cancer metastasis therapy in vitro and in vivo.


Assuntos
Cobre/farmacologia , Indóis/farmacologia , Nanopartículas/uso terapêutico , Neoplasias/terapia , Fotoquimioterapia/métodos , Polímeros/farmacologia , Terapia por Ultrassom/métodos , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Camundongos , Hipóxia Tumoral
13.
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
14.
Biomaterials ; 175: 82-92, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29803106

RESUMO

Silica based nanoparticles have emerged as a promising vaccine delivery system for cancer immunotherapy, but their bio-degradability, adjuvanticity and the resultant antitumor activity remain to be largely improved. In this study, we report biodegradable glutathione-depletion dendritic mesoporous organosilica nanoparticles (GDMON) with a tetrasulfide-incorporated framework as a novel co-delivery platform in cancer immunotherapy. Functionalized GDMON are capable of co-delivering an antigen protein (ovalbumin) and a toll-like receptor 9 (TLR9) agonist into antigen presenting cells (APCs) and inducing endosome escape. Moreover, decreasing the intracellular glutathione (GSH) level through the -S-S-/GSH redox chemistry increases the ROS generation level both in vitro and in vivo, facilitating cytotoxic T lymphocyte (CTL) proliferation and reducing tumour growth in an aggressive B16-OVA melanoma tumour model. Our results have shown the potential of GDMON as a novel self-adjuvant and co-delivery nanocarrier for cancer vaccine.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas Anticâncer/administração & dosagem , Glutationa/metabolismo , Melanoma Experimental/terapia , Nanopartículas/química , Dióxido de Silício/química , Animais , Células Apresentadoras de Antígenos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Endossomos/fisiologia , Humanos , Imunoterapia , Melanoma Experimental/imunologia , Melanoma Experimental/patologia , Camundongos , Oligodesoxirribonucleotídeos/administração & dosagem , Ovalbumina/administração & dosagem , Oxirredução , Polietilenoimina/química , Porosidade , Linfócitos T Citotóxicos/patologia , Receptor Toll-Like 9/agonistas
15.
Biomaterials ; 187: 55-65, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30292942

RESUMO

A glutathione (GSH)-sensitive supramolecular nitric oxide (NO) nanogenerator is developed as an all-rounder for enhanced photodynamic therapy (PDT). By integrating GSH-sensitive NO prodrug into the system via LEGO-like host-guest interaction, the nanocarrier could not only deplete intracellular GSH, but also relieve hypoxia at tumor sites through NO mediated blood vessel relaxation. Furthermore, reactive nitrogen species (RNS) with enhanced biocidal activity could be produced by the reaction between NO and reactive oxygen species (ROS), generated from α-cyclodextrin (α-CD) conjugated S-nitrosothiol and light-activated chlorin e6 (Ce6) respectively. Due to multiple combined effects between NO and PDT, the NO acts as the loaded gunpowder inside a 'grenade', 'explosively' amplifying the therapeutic effects that the light responsive 'fuse' Ce6 could exert. The present work may well serve as an inspiration for future creative approaches of photodynamic cancer therapy.


Assuntos
Glutationa/metabolismo , Óxido Nítrico/metabolismo , Fotoquimioterapia/métodos , Espécies Reativas de Nitrogênio/metabolismo , Animais , Hipóxia Celular , Sobrevivência Celular , Clorofilídeos , Ciclodextrinas/química , Portadores de Fármacos , Feminino , Humanos , Luz , Células MCF-7 , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas , Polímeros , Porfirinas/química , Espécies Reativas de Oxigênio/metabolismo , S-Nitrosotióis/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA