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.
Opt Express ; 30(15): 26362-26376, 2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-36236830

RESUMO

The Pancharatnam-Berry (PB) phase can be used to control the phase of circularly polarized electromagnetic waves. However, there are few studies on the modulation of dual-circularly polarized multi-beam using the transmissive coding metasurface. A scheme of spin-controlling multi-beam by transmissive coding metasurface is proposed for dual-circular polarization simultaneously. The transmissive coding metasurface (TCMS) can transmit linearly polarized incidence into multi-beam with orthogonally circular polarization. The phase distribution is designed based the convolution theorem, and the elements of metasurface conforming to the PB phase are arranged according to the phase distribution. In order to compensate the emitting spherical waves into plane waves and realize the transmissive waves with dual-circular polarization, an interesting scheme of elements in different regions with different rotating phase are presented based on the principle of phase compensation. TCMS can transmit linearly polarized waves into two left-hand circularly polarized (LHCP) beams and two right-hand circularly polarized (RHCP) beams. The prototype of TCMS is fabricated and measured, and the experimental results agree well with the simulated data. The transmissive metasurface has potential application in holograms and satellite communication.

2.
Materials (Basel) ; 15(19)2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36234369

RESUMO

Multiple functionalities on a shared aperture are crucial for metasurfaces (MSs) in many applications. In this paper, we propose a coding-feeding metasurface (CFMS) with the multiple functions of high-gain radiation, orbital angular momentum (OAM) generation, and radar cross-section (RCS) reduction based on phase manipulation. The unit cell of the CFMS is composed of a rectangular emission patch and two quasi-Minkowski patches for reflective phase manipulation, which are on a shared aperture. The high-gain radiation and multiple modes of ±1, ±2, and ±3 OAM generation were realized by rationally setting the elements and the phase of their excitation. The CFMS presents a broadband RCS reduction of 8 dB from 3.18 GHz to 7.56 GHz for y-polarization and dual-band RCS reduction for x-polarization based on phase interference. To validate the concept of the CFMS, a prototype was fabricated and measured. The results of the measurement agree well with the simulation. A CFMS with the advantages of light weight and low profile has potential application in detection and wireless communication systems for stealth aircraft.

3.
Int J Nanomedicine ; 16: 7269-7281, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34737564

RESUMO

PURPOSE: Small molecule modified antitumor drug conjugate nanoparticles have the advantages of high drug loading, simple synthesis and preparation, and better biocompatibility. Due to the large demand for exogenous α-linolenic acid (ALA) by tumor cells, we synthesized α-linolenic acid-paclitaxel conjugate (ALA-PTX) and prepared α-linolenic acid-paclitaxel conjugate nanoparticles (ALA-PTX NPs), in order to obtain better tumor cellular uptake and antitumor activity in vitro and in vivo. METHODS: We synthesized and characterized ALA-PTX, and then prepared and characterized ALA-PTX NPs. The cellular uptake, uptake pathways, intracellular behavior, in vitro and in vivo antitumor activity of ALA-PTX NPs were evaluated. RESULTS: The size of ALA-PTX NPs was approximately 110.7±1.7 nm. The drug loading was approximately 90% (w/w) with CrEL-free and organic solvent-free characteristics. The cellular uptake of ALA-PTX NPs was significantly higher than that of PTX injection by MCF-7, MCF-7/ADR and HepG2 cells. In these three cell lines, the cellular uptake of ALA-PTX NPs at 6h was approximately 1.5-2.6 times higher than that of PTX injection. ALA-PTX NPs were ingested through clathrin-mediated endocytosis, then transferred to lysosomes, and could dissolve in cells to play an antitumor activity. The in vitro and in vivo antitumor activity of ALA-PTX NPs was confirmed in MCF-7/ADR and HepG2 cell models and tumor-bearing nude mouse models. CONCLUSION: ALA-PTX NPs developed in our study could provide a new method for the preparation of nano-delivery systems suitable for antitumor therapy that could increase tumor cellular uptake and enhance antitumor activity.


Assuntos
Antineoplásicos Fitogênicos , Antineoplásicos , Nanopartículas , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Endocitose , Camundongos , Paclitaxel , Ácido alfa-Linolênico
4.
Acta Biomater ; 136: 495-507, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34619371

RESUMO

The antitumor immune response involves a cascade of cancer-immunity cycles. Developing a combination therapy aimed at the cancer-immunity cycle is of great importance. In this research, we designed and tested a combined therapeutic-Au nanorod (AuNR)/doxorubicin (DOX) gel (AuNR/DOX gel)-in which the sustained release of DOX was controlled by Pluronic gel. DOX served as an immunogenic tumor cell death (ICD) inducer, triggering the production of damage-associated molecular patterns (DAMPs). Mild photothermal therapy (Mild PTT) produced by 880 nm laser-irradiated AuNRs also generated tumor-associated antigens. Maleimide-modified liposomes (L-Mals), as antigen capturing agents, promoted tumor antigen uptake by DCs. Ultimately, more CD8+ T cells and fewer regulatory T cells (Tregs) infiltrated the tumor, eliciting antitumor responses from the PD-L1 antibody. Our results indicate that this combination strategy promotes a positive shift in the cancer-immunity cycle and holds much promise for combination strategy will lead to development of an antitumor drug delivery system. STATEMENT OF SIGNIFICANCE: Developing a combination therapy for cancer-immunity cycle is of great importance due to antitumor immune response involving a cascade of cancer-immunity cycles. Cancer-immunity cycle usually includes tumor antigen release, antigen presentation, immune activation, trafficking, infiltration, specific recognition of tumor cells by T cells, and finally cancer cell killing. In this research, we designed a combination strategy based on Au nanorod/doxorubicin gel via mild photothermal therapy combined with antigen-capturing liposomes and anti-PD-L1 agent promoting a positive shift in the cancer-immunity cycle. Our results indicate that this combination strategy promotes a positive shift in the cancer-immunity cycle and holds much promise for combination strategy will lead to development of an antitumor drug delivery system.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Doxorrubicina , Melanoma Experimental , Nanotubos , Terapia Fototérmica , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Feminino , Lipossomos , Melanoma Experimental/tratamento farmacológico , Camundongos Endogâmicos C57BL
5.
Drug Deliv ; 28(1): 1603-1615, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34319209

RESUMO

Small molecule-chemotherapeutic drug conjugate nanoparticles (SMCDC NPs) has a great advantage in improving drug loading. However, the factors which influence these conjugates forming stable nanoparticles (NPs) are currently unclear. In our previous studies, we synthesized a series of fatty acid-paclitaxel conjugates and suggested that the changes in the hydrophobic parameters (XlogP), solubility parameters and crystallinity of these fatty acid-paclitaxel conjugates were the key factors for affecting these small molecule-chemotherapeutic drug conjugates (SMCDCs) forming stable NPs in water. Here, we selected clinically widely used chemotherapeutic drug (docetaxel (DTX), doxorubicin (DOX) and irinotecan (Ir)) as model drug, and chose three straight-chain fatty acids (acetic acid (Ac), hexanoic acid (HA) and stearic acid (SA)) and one branched small molecule (N-(tert-butoxycarbonyl) glycine (B-G)) to synthesize 12 SMCDCs. Our results indicated that our prediction criterions obtained from paclitaxel conjugates were also appropriated for these synthesized SMCDCs. We suggested that the present studies expanded the scope of application of the above-mentioned influencing factors, provided research ideas for the rational design of SMCDC forming NPs and a basis for screening NPs with good anticancer activity.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Portadores de Fármacos/química , Ácidos Graxos/química , Nanopartículas/química , Ácido Acético/química , Caproatos/química , Sobrevivência Celular , Química Farmacêutica , Docetaxel/administração & dosagem , Docetaxel/farmacologia , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Irinotecano/administração & dosagem , Irinotecano/farmacologia , Células MCF-7 , Tamanho da Partícula , Solubilidade , Ácidos Esteáricos/química
6.
Drug Deliv ; 28(1): 800-813, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33866918

RESUMO

Cancer immunotherapy is a strategy that is moving to the frontier of cancer treatment in the current decade. In this study, we show evidence that 3-(2-nitrophenyl) propionic acid-paclitaxel nanoparticles (NPPA-PTX NPs), act as immunogenic cell death (ICD) inducers, stimulating an antitumor response which results in synergistic antitumor activity by combining anti-PD-L1 antibody (aPD-L1) in vivo. To investigate the antitumor immunity induced by NPPA-PTX NPs, the expression of both ICD marker calreticulin (CRT) and high mobility group box 1 (HMGB1) were analyzed. In addition, the antitumor activity of NPPA-PTX NPs combined with aPD-L1 in vivo was also investigated. The immune response was also measured through quantitation of the infiltration of T cells and the secretion of pro-inflammatory cytokines. The results demonstrate that NPPA-PTX NPs induce ICD of MDA-MB-231 and 4T1 cells through upregulation of CRT and HMGB1, reactivating the antitumor immunity via recruitment of infiltrating CD3+, CD4+, CD8+ T cells, secreting IFN-γ, TNF-α, and the enhanced antitumor activity by combining with aPD-L1. These data suggest that the combined therapy has a synergistic antitumor activity and has the potential to be developed into a novel therapeutic regimen for cancer patients.


Assuntos
Albuminas/farmacologia , Antineoplásicos/farmacologia , Morte Celular Imunogênica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Nanopartículas/química , Paclitaxel/farmacologia , Albuminas/administração & dosagem , Animais , Antineoplásicos/administração & dosagem , Antígeno B7-H1/imunologia , Calreticulina/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Proteína HMGB1/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Paclitaxel/administração & dosagem , Propionatos/química , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Nanomedicine ; 15: 1809-1821, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32214813

RESUMO

INTRODUCTION: Because tumor-associated inflammation is a hallmark of cancer treatment, in the present study, sorafenib mesoporous silica nanomatrix (MSNM@SFN) co-administrated with flufenamic acid (FFA, a non-steroidal anti-inflammatory drug (NSAID)) was investigated to enhance the anti-tumor activity of MSNM@SFN. METHODS: Metastatic breast tumor 4T1/luc cells and hepatocellular carcinoma HepG2 cells were selected as cell models. The effects of FFA in vitro on cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in 4T1/luc and HepG2 cells were investigated. The in vivo anti-tumor activity of MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) was evaluated in a 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model, respectively. RESULTS: The results indicated that FFA could markedly decrease cell migration, PGE2 secretion, and AKR1C1 and AKR1C3 levels in both 4T1/luc and HepG2 cells. The enhanced anti-tumor activity of MSNM@SFN+FFA compared with that of MSNM@SFN was confirmed in the 4T1/luc metastatic tumor model, HepG2 tumor-bearing nude mice model, and HepG2 orthotopic tumor-bearing nude mice model in vivo, respectively. DISCUSSION: MSNM@SFN co-administrating with FFA (MSNM@SFN+FFA) developed in this study is an alternative strategy for improving the therapeutic efficacy of MSNM@SFN via co-administration with NSAIDs.


Assuntos
Anti-Inflamatórios não Esteroides/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , 20-Hidroxiesteroide Desidrogenases/metabolismo , Membro C3 da Família 1 de alfa-Ceto Redutase/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Dinoprostona/metabolismo , Feminino , Ácido Flufenâmico/administração & dosagem , Células Hep G2 , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Dióxido de Silício/química , Sorafenibe/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Nanomedicine ; 14: 195-204, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30636872

RESUMO

BACKGROUND: 3-(2-Nitrophenyl) propionic acid-paclitaxel (NPPA-PTX) is a paclitaxel (PTX) bioreductive prodrug synthesized by our lab. We hypothesize that NPPA-PTX can self-assemble to form nanoparticles (NPs). MATERIALS AND METHODS: In the present research, the theoretical partition coefficient (XlogP) and Hansen solubility parameters of NPPA-PTX were calculated. NPPA-PTX nanoparticles prepared by NPPA-PTX and DSPE-PEG (NPPA-PTX:DSPE-PEG =1:0.1, w/w) (NPPA-PTX@PEG NPs) were prepared and characterized. The cellular uptake, in vitro antitumor activity, in vivo targeting effect, tumor distribution, in vivo antitumor activity, and safety of NPPA-PTX@PEG NPs were investigated. RESULTS: Our results indicate that NPPA-PTX can self-assemble to form NPPA-PTX@PEG NPs. Both the cellular uptake and safety of NPPA-PTX@PEG NPs were higher than those of Taxol. NPPA-PTX@PEG NPs could target tumor tissues by a passive targeting effect. In tumor tissues, NPPA-PTX@PEG NPs could completely transform into active PTX. The in vivo antitumor activity of NPPA-PTX@PEG NPs was confirmed in MDA-MB-231 tumor-bearing nude mice. CONCLUSION: The bioreductive prodrug NPPA-PTX could self-assemble to form NPs. The safety and antitumor activity of NPPA-PTX@PEG were confirmed in our in vitro and in vivo experiments. The NPPA-PTX@PEG NPs developed in this study could offer a new way of preparing bioreductive prodrug, self-assembled NPs suitable for antitumor therapy.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Nanopartículas/administração & dosagem , Paclitaxel/análogos & derivados , Fenilpropionatos/farmacologia , Pró-Fármacos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Feminino , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos ICR , Camundongos Nus , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Fenilpropionatos/administração & dosagem , Pró-Fármacos/administração & dosagem , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Control Release ; 295: 102-117, 2019 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-30582952

RESUMO

Nucleus-targeting drug delivery systems (NTDDs) deliver chemotherapeutic agents to nuclei in order to improve the efficacy of anti-tumour therapy. Histone H1 (H1) plays a key role in establishing and maintaining higher order chromatin structures and could bind to cell membranes. In the present study, we selected H1 as a target to prepare a novel H1-mediated NTDD. Low molecular weight heparin (LMHP) and doxorubicin (DOX) were combined to form LMHP-DOX. Then, a novel NTDD consisting of LMHP-DOX nanoparticles (LMHP-DOX NPs) was prepared by self-assembly. The characteristics of LMHP-DOX and LMHP-DOX NPs were investigated. Histone H1 high-expressive prostate cancer PC-3M cell line was selected as the cell model. Cellular uptake, and the in vitro and in vivo anti-tumour activity of LMHP-DOX NPs were evaluated on H1 high-expressive human prostate cancer PC-3M cells. Our results indicated that intact LMHP-DOX NPs mediated by H1 could be absorbed by H1 high-expressive PC-3M cells, escape from the lysosomes to the cytoplasm, and localize in the perinuclear region via H1-mediated, whereby DOX could directly enter the cell nucleus and quickly increase the concentration of DOX in the nuclei of H1 high-expressive PC-3M cells to enhance the apoptotic activity of cancer cells. The anti-coagulant activity of LMHP-DOX NPs was almost completely diminished in rat blood compared with that of LMHP, indicating the safety of LMHP-DOX NPs. Compared to traditional NTDD strategies, LMHP-DOX NPs avoid the complicated modification of nucleus-targeting ligands and provide a compelling solution for the substantially enhanced nuclear uptake of chemotherapeutic agents for the development of more intelligent NTDDs.


Assuntos
Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Heparina de Baixo Peso Molecular/administração & dosagem , Histonas/análise , Neoplasias da Próstata/tratamento farmacológico , Animais , Anticoagulantes/administração & dosagem , Anticoagulantes/farmacocinética , Anticoagulantes/farmacologia , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Doxorrubicina/farmacocinética , Doxorrubicina/farmacologia , Sistemas de Liberação de Medicamentos , Liberação Controlada de Fármacos , Heparina de Baixo Peso Molecular/farmacocinética , Heparina de Baixo Peso Molecular/farmacologia , Humanos , Masculino , Nanopartículas/ultraestrutura , Células PC-3 , Neoplasias da Próstata/patologia , Ratos Sprague-Dawley
10.
Int J Nanomedicine ; 13: 1495-1504, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29559778

RESUMO

BACKGROUND: In the present study, the tumor-specific, pH-responsive peptide H7K(R2)2-modified, theranostic liposome-containing paclitaxel (PTX) and superparamagnetic iron oxide nanoparticles (SPIO NPs), PTX/SPIO-SSL-H7K(R2)2, was prepared by using H7K(R2)2 as the targeting ligand, SPIO NPs as the magnetic resonance imaging (MRI) agent, PTX as antitumor drug. METHODS: The PTX/SPIO-SSL-H7K(R2)2 was prepared by a thin film hydration method. The characteristics of PTX/SPIO-SSL-H7K(R2)2 were evaluated. The targeting effect, MRI, and antitumor activity of PTX/SPIO-SSL-H7K(R2)2 were investigated detail in vitro and in vivo in human breast carcinoma MDA-MB-231 cell models. RESULTS: Our results of in vitro flow cytometry, in vivo imaging, and in vivo MR imaging confirmed the pH-responsive characteristic of H7K(R2)2 in MDA-MB-231 cell line in vitro and in vivo. The results of in vivo MRI and in vivo antitumor activity confirmed the theranostic effect of PTX/SPIO-SSL-H7K(R2)2 in MDA-MB-231 tumor-bearing model. CONCLUSION: Considering all our in vitro and in vivo results, we conclude that we developed targeting modified theranostic liposome which could achieve both role of antitumor and MRI.


Assuntos
Compostos Férricos/química , Nanopartículas de Magnetita/química , Neoplasias/tratamento farmacológico , Paclitaxel/uso terapêutico , Peptídeos/química , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Citometria de Fluxo , Humanos , Concentração de Íons de Hidrogênio , Lipossomos , Imageamento por Ressonância Magnética , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias/patologia , Paclitaxel/farmacologia , Distribuição Tecidual/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...