Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mater Today Bio ; 16: 100397, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36081578

RESUMO

In vivo fluorescent imaging by using the new contrast agents emitted at short-wavelength infrared region (NIR II, 1000-1700 â€‹nm) presents an unprecedent advantages in imaging sensitivity and spatial resolution over traditional near-infrared (NIR) light. Recently, Nd-based rare-earth nanocrystals have attracted considerable attention due to the high quantum yield (∼40%) of their emission at NIR II. However, undesirable capture by reticuloendothelial system to bring strong background signal is unsatisfying for tumor discrimination. Here, GSH-sensitive tetrasulfide bond incorporated mesoporous silica shell has entrusted onto Nd-based down-conversion nanocrystals (DCNPs) surface to totally quench the fluorescence of DCNPs. After RGD conjugation on the silica surface, the NIR II contrast agents could actively target to liver tumors. Then tetrasulfide bonds can be broken during the silica framework decomposing in cytoplasm under high GSH concentration to result in NIR II fluorescence explosive recover. Benefiting from this specific response under tumor microenvironment, the NIR II signal in other organs was markedly reduced, while the signal-to-background ratio is prominently enhanced in tumors. Then, solid liver tumors were successfully resected under the guidance of our GSH responsive NIR II fluorescent imaging with no recurrence after 20-day of surgery. Meanwhile, by combining with the ignorable side effects, the Nd-based nanoprobes vastly improved the imaging resolution of tumor margin, opening a paradigm of NIR II fluorescent imaging-guided surgery.

2.
Adv Healthc Mater ; 11(21): e2201986, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36106722

RESUMO

Intracellular oxidative amplification can effectively destroy tumor cells. Additionally, Fe-mediated Fenton reaction often converts cytoplasm H2 O2 to generate extensive hypertoxic hydroxyl radical (• OH), leading to irreversible mitochondrion damage for tumor celleradication, which is widely famous as tumor chemodynamic therapy (CDT). Unfortunately, intracellular overexpressed glutathione (GSH) always efficiently scavenges • OH, resulting in the significantly reduced CDT effect. To overcome this shortcoming and improve the oxidative stress in cytoplasm, Fe3 O4 ultrasmall nanoparticle encapsulated and ICG loaded organo-mesoporous silica nanovehicles (omSN@Fe-ICG) are constructed to perform both photothermal and GSH depletion to enhance the Fenton-like CDT, by realizing intracellular oxidative stress amplification. After this nanoagents are internalized, the tetrasulfide bonds in the dendritic mesoporous framework can be decomposed with GSH to amplify the toxic ROS neration by selectively converting H2 O2 to hydroxyl radicals through the released Fe-based nanogranules. Furthermore, the NIR laser-induced hyperthermia can further improve the Fenton reaction rate that simultaneously destroyed the mitochondria. As a result, the GSH depletion and photothermal assisted CDT can remarkably improve the tumor eradication efficacy.


Assuntos
Hipertermia Induzida , Nanopartículas , Neoplasias , Humanos , Dióxido de Silício , Linhagem Celular Tumoral , Glutationa , Nanopartículas/química , Peróxido de Hidrogênio
3.
ChemMedChem ; 17(19): e202200360, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36000799

RESUMO

The combination of photothermal therapy and chemotherapy are developing as a promising clinical strategy but it urgently needs the high exploration of intelligent multifunctional drug delivery nanovectors. In this paper, we used a versatile method to construct mesoporous polydopamine nanovehicles (MPDA) with the dendritic mesopores loaded with a clinical chemotherapeutic drug, Doxorubicin (MPDA@DOX). The monodisperse nanoagents are spherical with a size of ∼160 nm and pore size of approximately 10 nm. MPDA could efficiently delivery DOX with π-π stacking interaction and acts as the potent photothermal agents. Importantly, MPDA@DOX are preferentially internalized by cancerous cells, then bursting drug release and local hyperthermia generation were observed in conditions representative of the cytoplasm in tumor cells that highly synergistic cell killing effect were found under 808 nm laser irradiation. The fluorescent imaging results of human breast tumor bearing murine model evidenced that MPDA delivery platform have excellent tumor precise targeting effect and in vivo tumor ablation experiment further revealed that MPDA@DOX showed markedly eradicated tumor growth capability under laser exposure. Therefore, this work provided a fascinating strategy based on biocompatible MPDA based drug delivery system for malignant tumors eradication via synergistic therapy.


Assuntos
Nanopartículas , Neoplasias , Animais , Compostos de Diazônio , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Humanos , Indóis , Camundongos , Neoplasias/tratamento farmacológico , Preparações Farmacêuticas , Fototerapia/métodos , Polímeros , Piridinas
4.
Nanomedicine ; 43: 102555, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35390525

RESUMO

The near-infrared fluorescence imaging has been integrated into the operating room to guide tumor resection, potentially reducing the positive margin rates in breast-conserving surgery (BCS). Relative to the widely used first near-infrared fluorescence imaging, imaging in the second near-infrared (NIR-II) region possesses higher contrast and deeper tissue penetration, particularly in the NIR-IIb window, offering many new opportunities for imaging-guided BCS. Here, we fabricated the c(RGDfC) functionalized erbium-based rare-earth nanoparticles (ErNPs@cRGD) with superior optical property in NIR-IIb region. Owing to deeper tissue penetration and efficient tumor targeting, ErNPs@cRGD-based NIR-IIb fluorescence imaging achieved enhanced signal-to-background ratios in tumor visualization, which was able to guide more complete tumor resection, identify multiple microtumors and distinguish malignant lesions from normal tissues in various mice models. Based on these, this NIR-IIb imaging strategy for surgical navigation can significantly reduce positive margin rates and improve prognosis, laying a foundation for the clinical resection of breast cancer.


Assuntos
Neoplasias da Mama , Nanopartículas , Cirurgia Assistida por Computador , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Neoplasias da Mama/cirurgia , Feminino , Fluorescência , Humanos , Camundongos , Nanopartículas/química , Imagem Óptica/métodos , Cirurgia Assistida por Computador/métodos
5.
Adv Sci (Weinh) ; 9(12): e2104728, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35170876

RESUMO

Positive resection margin frequently exists in breast-conserving treatment (BCT) of early-stage breast cancer, and insufficient therapeutic efficacy is common during radiotherapy (RT) in advanced breast cancer patients. Moreover, a multimodal nanotherapy platform is urgently required for precision cancer medicine. Therefore, a biodegradable cyclic RGD pentapeptide/hollow virus-like gadolinium (Gd)-based indocyanine green (R&HV-Gd@ICG) nanoprobe is developed to improve fluorescence image-guided surgery and breast cancer RT efficacy. R&HV-Gd exhibits remarkably improved aqueous stability, tumor retention, and target specificity of ICG, and achieves outstanding magnetic resonance/second near-infrared (NIR-II) window multimodal imaging in vivo. The nanoprobe-based NIR-II fluorescence image guidance facilitates complete tumor resection, improves the overall mouse survival rate, and effectively discriminates between benign and malignant breast tissues in spontaneous breast cancer transgenic mice (area under the curve = 0.978; 95% confidence interval: 0.952, 1.0). Moreover, introducing the nanoprobe to tumors generated more reactive oxygen species under X-ray irradiation, improved RT sensitivity, and reduced mouse tumor progression. Notably, the nanoprobe is biodegradable in vivo and exhibits accelerated bodily clearance, which is expected to reduce the potential long-term inorganic nanoparticle toxicity. Overall, the nanoprobe provides a basis for developing precision breast cancer treatment strategies.


Assuntos
Neoplasias da Mama , Nanopartículas , Cirurgia Assistida por Computador , Animais , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/radioterapia , Neoplasias da Mama/cirurgia , Feminino , Gadolínio , Humanos , Verde de Indocianina , Margens de Excisão , Camundongos , Cirurgia Assistida por Computador/métodos
6.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163548

RESUMO

Owing to their good stability and high photothermal conversion efficiency, the development of carbon-based nanoparticles has been intensively investigated, while the limitation of unsatisfactory cellular internalization impedes their further clinical application. Herein, we report a novel strategy for fabrication of Fe3O4 yolk-shell mesoporous carbon nanocarriers (Fe3O4@hmC) with monodispersity and uniform size, which presented significantly higher cell membrane adsorption and cellular uptake properties in comparison with common solid silica-supported mesoporous carbon nanoparticles with core-shell structure. Moreover, the MRI performance of this novel Fe-based nanoparticle could facilitate precise tumor diagnosis. More importantly, after DOX loading (Fe3O4@hmC-DOX), owing to synergistic effect of chemo-phototherapy, this therapeutic agent exhibited predominant tumor cell ablation capability under 808 nm NIR laser irradiation, both in vitro and in vivo. Our work has laid a solid foundation for therapeutics with hollowed carbon shell for solid tumor diagnosis and therapy in clinical trials.


Assuntos
Neoplasias da Mama/terapia , Carbono/química , Doxorrubicina/administração & dosagem , Nanopartículas Magnéticas de Óxido de Ferro/química , Animais , Neoplasias da Mama/diagnóstico por imagem , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Terapia Combinada , Doxorrubicina/química , Feminino , Camundongos , Nanoestruturas , Tamanho da Partícula , Terapia Fototérmica , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mater Sci Eng C Mater Biol Appl ; 129: 112351, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34579877

RESUMO

Phototherapy has attracted increasing attention in cancer therapy owing to its non-invasive nature, high spatiotemporal selectivity, and negligible side effects. However, a single photosensitizer often exhibits poor photothermal conversion efficiency or insufficient reactive oxygen species (ROS) productivity. Even worse, the ROS can be consumed by tumor overexpressed reductive glutathione, resulting in severely compromised phototherapy. In this paper, we prepared a MnII-coordination driven dual-photosensitizers co-assemblies (IMCP) for imaging-guided self-enhanced PDT/PTT. Specifically, a photothermal agent indocyanine green (ICG), a photodynamic agent chlorin e6 (Ce6), and a transition metal ion (MnII/III) were chosen to synthesize the nanodrug via coordination-driven co-assembly. The as-prepared IMCP exhibited extremely high photosensitizer payload (96 wt%), excellent physiological stability, and outstanding tumor accumulation. Moreover, the existence of MnII not only assists the nanostructure formation but also could competitively coordinate with GSH to minimize the unnecessary ROS consumption, thus improving PDT efficiency. Meanwhile, benefiting from the intrinsic fluorescence, photoacoustic imaging ability of photosensitizers, and the MRI contrast potential of MnII/III, IMCP exhibited superior imaging potential for guiding tumor phototherapy. By changing the excitation wavelength suitably, IMCP could realize the switch between PTT and PDT. In short, the dual-PSs co-assembled nanotheranostic has great potential for multi-modal imaging guided phototherapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Humanos , Verde de Indocianina , Imagem Multimodal , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia
8.
Biomater Sci ; 9(3): 1047, 2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33410825

RESUMO

Correction for 'Tumor acidity-responsive carrier-free nanodrugs based on targeting activation via ICG-templated assembly for NIR-II imaging-guided photothermal-chemotherapy' by Kaihang Xue et al., Biomater. Sci., 2021, DOI: 10.1039/D0BM01864C.

9.
Biomater Sci ; 9(3): 1008-1019, 2021 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-33320145

RESUMO

Carrier-free nanodrugs composed of photosensitizers and chemotherapeutic drugs show great potential in synergistic photothermal-chemotherapy. However, the targeting specificity to tumor cells is still a major obstacle for carrier-free nanodrugs. Meanwhile, almost all exogenous tumor-targeting ligands show no therapeutic effect by themselves. Here, a tumor microenvironment-driven self-targeting supramolecular nanodrug was successfully constructed via an indocyanine green (ICG)-templated small-molecule self-assembly strategy with methotrexate (MTX, folic acid-like antitumor drug) followed by post-insertion of weak acidity-responsive PEG for synergistic photothermal-chemotherapy. Interestingly, the size and morphology could be adjusted by changing the ICG-to-MTX ratio. Notably, the dynamic introduction of PEG not only could temporarily shield self-targeting function in blood to prolong the circulation time, but also could trigger the activation of self-targeting via re-exposing MTX ligands within the tumor microenvironment to enhance cellular uptake. Furthermore, the dePEGylated nanodrug would be disassembled to release MTX on-demand for chemotherapy via both stimuli of stronger lysosomal acidity and an external NIR laser. Moreover, the elimination of tumors could be realized through NIR-II fluorescence/PA imaging-guided synergistic photothermal-chemotherapy. The tumor microenvironment-driven carrier-free nanodrug based on self-targeting activation via ICG-templated assembly might provide a brand-new idea for synergistic photothermal-chemotherapy.


Assuntos
Antineoplásicos , Fotoquimioterapia , Sistemas de Liberação de Medicamentos , Verde de Indocianina , Metotrexato
10.
ACS Appl Mater Interfaces ; 12(29): 32477-32492, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32578429

RESUMO

A carrier-free theranostic nanodrug directly coassembled using a NIR probe and a chemotherapeutic drug is a promising alternative for cancer theranostics. Nevertheless, this nanodrug still faces the limitations of short blood circulation and inefficient tumor accumulation/tumoral cellular uptake in vivo. Meanwhile, most exogenous targeting ligands and poly(ethylene glycol) have no therapeutic effect. Herein, we designed an ultralong-circulating and self-targeting nanodrug by an ordered supramolecular coassembly of indocyanine green (ICG), methotrexate (MTX, chemotherapeutic drug and cancer-cell-specific ligand), and clofarabine (CA). Notably, CA, as a surfactant-like chemotherapeutic drug, was introduced into the initial ICG-MTX coassembly by "Watson-Crick A = T-inspired" hydrogen-bond-driven sequential assembly with MTX. This carrier-free theranostic nanodrug with exceptionally high drug payload (100 wt %) not only showed superior serum stabilities but also displayed ultralong blood circulation (>7 days), enabling efficient accumulation at tumor sites. Moreover, our nanodrugs could be self-recognized by cancer cells and release the drugs on demand through lysosomal acidity and external laser stimulus. Under NIR-II imaging guidance, high-efficiency tumor ablation via synergistic photothermal-chemotherapy could be achieved in one treatment cycle while preventing the tumor recurrence. Our ultralong-circulating and self-recognizing carrier-free theranostic nanodrug based on the "drug-delivering-drug" strategy might have the potential for clinical theranostic application.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Clofarabina/farmacologia , Verde de Indocianina/química , Metotrexato/farmacologia , Neoplasias/tratamento farmacológico , Fotoquimioterapia , Nanomedicina Teranóstica , Animais , Antimetabólitos Antineoplásicos/química , Clofarabina/química , Liberação Controlada de Fármacos , Humanos , Raios Infravermelhos , Substâncias Macromoleculares/química , Metotrexato/química , Camundongos , Neoplasias/diagnóstico por imagem , Imagem Óptica , Tamanho da Partícula , Propriedades de Superfície , Células Tumorais Cultivadas
11.
J Mater Chem B ; 8(17): 3829-3841, 2020 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-32232285

RESUMO

Carrier-free nanodrugs, generated via the straightforward small-molecule self-assembly of anticancer drugs, provide a promising route for cancer chemotherapy. However, their low structural stability, lack of targeting specificity, and poor stimulus responsiveness are still limiting their therapeutic effect. Inspired by Watson-Crick G[triple bond, length as m-dash]C base pairing, the FDA-approved chemo-drug methotrexate (MTX, which can bind with folate receptors) and 5-fluorouracil (5-FU, a DNA/RNA synthetase inhibitor) were adopted for direct assembly into self-recognizing MTX-5-FU nanoparticles via "Watson-Crick-like base pairing"-driven precise supramolecular assembly. Sequentially, our synthesized weak acidity-responsive polyethylene glycol (PEG) was inserted onto the nanoparticle surface to temporarily shield the self-targeting function of MTX and prolong the blood circulation time. Once PEG-MTX-5-FU nanoparticles reached the weakly acidic tumor microenvironment, the PEG corona could be cleaved from their surface and then MTX could be re-exposed to recover its self-recognition ability and significantly elevate tumor cell uptake; furthermore, the de-PEGylated MTX-5-FU nanoparticles could respond to the stronger acidity of lysosome, triggering core disassembly and thus the burst release of both MTX and 5-FU. Further in vitro and in vivo studies consistently confirmed that the nanodrugs exhibited preferable accumulation at the tumor sites with highly synergistic chemotherapeutic effects. The supramolecular recognition-inspired, cascade-triggered self-targeting and controlled release of nanodrugs could be a promising strategy to improve synergistic chemotherapy.


Assuntos
Antineoplásicos/farmacologia , Citosina/química , Fluoruracila/farmacologia , Guanina/química , Metotrexato/farmacologia , Nanopartículas/química , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Fluoruracila/síntese química , Fluoruracila/química , Humanos , Substâncias Macromoleculares/síntese química , Substâncias Macromoleculares/química , Substâncias Macromoleculares/farmacologia , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Metotrexato/síntese química , Metotrexato/química , Camundongos , Estrutura Molecular , Tamanho da Partícula , Propriedades de Superfície , Microambiente Tumoral/efeitos dos fármacos
12.
Mol Pharm ; 16(7): 3133-3144, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31198046

RESUMO

The clinical translation of methotrexate (MTX) is limited because of low aqueous solubility, poor bioavailability, low uptake efficiency, and toxicity concerns. Herein, dual-acting MTX (not only targeting folate receptors but also killing cells via inhibition of intracellular folate metabolism) and hyaluronic acid (HA, targeting CD44 receptors) were selected to be covalently linked by the redox-responsive disulfide bond. The synthesized prodrug (HA-SS-MTX) as a molecular structural motif could self-assemble into simple yet multifunctional nanoparticles (HA-SS-MTX NPs) in aqueous solution. The HA-SS-MTX NPs displayed an average diameter of ∼110 nm with a uniformly spherical shape and maintained stability in different physiological media. Moreover, the HA-SS-MTX NPs could exhibit a sharp redox-dependent response for rapid structure disassembly and sequential MTX release compared to the redox-irresponsive group (HA-ADH-MTX NPs). Furthermore, the results of confocal microscopy and flow cytometry verified that the nanosystems were selectively uptaken by cancer cells via folate and CD44 receptor-mediated internalization through the dual-active targeting mechanism. In addition, HA-SS-MTX NPs could accumulate within tumor sites for a longer period. Notably, in vitro and in vivo antitumor results demonstrated that HA-SS-MTX NPs significantly promoted the death of cancer cells and enhanced the inhibition of tumor growth while reducing the toxicity as compared to MTX and HA-ADH-MTX NPs. Therefore, the smart HA-SS-MTX NPs as the simple and efficient platform have great potential in tumor-targeting drug delivery and therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Antagonistas do Ácido Fólico/metabolismo , Ácido Hialurônico/metabolismo , Metotrexato/metabolismo , Nanopartículas/química , Pró-Fármacos/metabolismo , Neoplasias do Colo do Útero/tratamento farmacológico , Animais , Sobrevivência Celular/efeitos dos fármacos , Dissulfetos/química , Liberação Controlada de Fármacos , Feminino , Antagonistas do Ácido Fólico/química , Antagonistas do Ácido Fólico/farmacologia , Células HeLa , Humanos , Receptores de Hialuronatos/antagonistas & inibidores , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Metotrexato/química , Metotrexato/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oxirredução , Distribuição Tecidual , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
ACS Appl Mater Interfaces ; 11(17): 15262-15275, 2019 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-30964624

RESUMO

Nanoparticles camouflaged by red blood cell (RBC) membranes have attracted considerable attention owing to reservation of structure of membrane and surface proteins, endowing prominent cell-specific function including biocompatibility, prolonged circulation lifetime, and reduced reticular endothelial system (RES) uptake ability. Considering the drawbacks of carrier-free nanomedicine including the serious drug burst release, poor stability, and lack of immune escape function, herein we developed and fabricated a novel RBC membranes biomimetic combinational therapeutic system by enveloping the small molecular drug coassemblies of 10-hydroxycamptothecin (10-HCPT) and indocyanine green (ICG) in the RBC membranes for prolonged circulation, controlled drug release, and synergistic chemo-photothermal therapy (PTT). The self-reorganized RBCs@ICG-HCPT nanoparticles (NPs) exhibited a diameter of ∼150 nm with core-shell structure, high drug payload (∼92 wt %), and reduced RES uptake function. Taking advantage of the stealth functionality of RBC membranes, RBCs@ICG-HCPT NPs remarkably enhanced the accumulation at the tumor sites by passive targeting followed by cellular endocytosis. Upon the stimuli of near-infrared laser followed by acidic stimulation, RBCs@ICG-HCPT NPs showed exceptional instability by heat-mediated membrane disruption and pH change, thereby triggering the rapid disassembly and accelerated drug release. Consequently, compared with individual treatment, RBCs@ICG-HCPT NPs under dual-stimuli accomplished highly efficient apoptosis in cancer cells and remarkable ablation of tumors by chemo-PTT. This biomimetic nanoplatform based on carrier-free, small molecular drug coassemblies integrating imaging capacity as a promising theranostic system provides potential for cancer diagnosis and combinational therapy.


Assuntos
Antineoplásicos Fitogênicos/química , Biomimética , Camptotecina/análogos & derivados , Membrana Celular/química , Raios Infravermelhos , Nanopartículas/química , Neoplasias/terapia , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Camptotecina/química , Camptotecina/farmacologia , Camptotecina/uso terapêutico , Eritrócitos/citologia , Eritrócitos/metabolismo , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Verde de Indocianina/química , Masculino , Camundongos , Camundongos Nus , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Fototerapia , Ratos , Ratos Sprague-Dawley
14.
Mol Pharm ; 16(5): 1982-1998, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30892898

RESUMO

Locating nanomedicines at the active sites plays a pivotal role in the nanoparticle-based cancer therapy field. Herein, a multifunctional nanotherapeutic is designed by using graphene oxide (GO) nanosheets with rich carboxyl groups as the supporter for hyaluronic acid (HA)-methotrexate (MTX) prodrug modification via an adipicdihydrazide cross-linker, achieving synergistic multistage tumor-targeting and combined chemo-photothermal therapy. As a tumor-targeting biomaterial, HA can increase affinity of the nanocarrier toward CD44 receptor for enhanced cellular uptake. MTX, a chemotherapeutic agent, can also serve as a tumor-targeting enhancer toward folate receptor based on its similar structure with folic acid. The prepared nanosystems possess a sheet shape with a dynamic size of approximately 200 nm and pH-responsive drug release. Unexpectedly, the physiological stability of HA-MTX prodrug-decorated GO nanosystems in PBS, serum, and even plasma is more excellent than that of HA-decorated GO nanosystems, while both of them exhibit an enhanced photothermal effect than GO nanosheets. More importantly, because of good blood compatibility as well as reduced undesired interactions with blood components, HA-MTX prodrug-decorated GO nanosystems exhibited remarkably superior accumulation at the tumor sites by passive and active targeting mechanisms, achieving highly effective synergistic chemo-photothermal therapeutic effect upon near-infrared laser irradiation, efficient ablation of tumors, and negligible systemic toxicity. Hence, the HA-MTX prodrug-decorated hybrid nanosystems have a promising potential for synergistic multistage tumor-targeting therapy.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Grafite/química , Nanoconjugados/química , Fotoquimioterapia/métodos , Neoplasias do Colo do Útero/terapia , Adipatos/química , Adipatos/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Sinergismo Farmacológico , Feminino , Grafite/metabolismo , Células HeLa , Humanos , Ácido Hialurônico/química , Ácido Hialurônico/metabolismo , Células MCF-7 , Metotrexato/química , Metotrexato/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células NIH 3T3 , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Distribuição Tecidual , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
ACS Biomater Sci Eng ; 5(9): 4442-4454, 2019 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-33438410

RESUMO

The medical application of nanotechnology is promising for cancer chemotherapy. However, most of the small-molecule drug assemblies still have such disadvantages as serious drug leakage and nonideal synergistic mechanisms, resulting in undesired therapeutic effect. Both nucleoside analogue-based clofarabine (CA) and methotrexate (MTX) were used as the first-line anticancer medication. However, a single-agent chemotherapy still faced many challenges including low bioavailability and toxic side effects to normal tissues due to nonspecific biodistribution of drugs. Herein, we designed and fabricated novel viral-mimicking and carry-free nanodrugs (CA-MTX NPs) via molecular recognition-driven precise self-assembly between CA and MTX. After introduction of mild acid-responsive PEG-lipid on the surface of CA-MTX NPs, the synthetic nanodrugs with a diameter of ∼150 nm exhibited tumor microenvironment-activated characteristics and self-targeting property. The results suggested that our nanodrugs could achieve superior tumor accumulation and synergistically promote the tumor suppression by collaboratively inhibiting dNTP pools. We foresaw that the well-designed smart nanodrugs delivery system would open a new avenue in synergistic cancer therapeutics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...