Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biomater Adv ; 150: 213425, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37084635

RESUMEN

The efficacy of immune checkpoint therapy is limited by the immunosuppressive tumor microenvironment (TME), and lactate, the most universal component of TME, has been rediscovered that plays important roles in the regulation of metabolic pathways, angiogenesis, and immunosuppression. Here, a therapeutic strategy of acidity modulation combined with programmed death ligand-1 (PD-L1) siRNA (siPD-L1) is proposed to synergistically enhance tumor immunotherapy. The lactate oxidase (LOx) is encapsulated into the hollow Prussian blue (HPB) nanoparticles (NPs) prepared by hydrochloric acid etching followed by the modification with polyethyleneimine (PEI) and polyethylene glycol (PEG) via sulfur bonds (HPB-S-PP@LOx), siPD-L1 is loaded via electrostatic adsorption to obtain HPB-S-PP@LOx/siPD-L1. The obtained co-delivery NPs can accumulate in tumor tissue with stable systemic circulation, and simultaneous release of LOx and siPD-L1 in intracellular high glutathione (GSH) environment after uptake by tumor cells without being destroyed by lysosome. Moreover, LOx can catalyze the decomposition of lactate in the hypoxic tumor tissue with the aid of oxygen release by the HPB-S-PP nano-vector. The results show that the acidic TME regulation via lactate consumption can improve the immunosuppressive TME, including revitalizing the exhausted CD8+ T cells and decreasing the proportion of immunosuppressive Tregs, and synergistically elevating the therapeutic effect of PD1/PD-L1 blockade therapy via siPD-L1. This work provides a novel insight for tumor immunotherapy and explores a promising therapy for triple-negative breast cancer.


Asunto(s)
Antígeno B7-H1 , Neoplasias , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Linfocitos T CD8-positivos/metabolismo , Terapia de Inmunosupresión , Inmunoterapia/métodos , Lactatos , Microambiente Tumoral
2.
Elife ; 122023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-37010266

RESUMEN

Myocardial fibrosis is the characteristic pathology of diabetes-induced cardiomyopathy. Therefore, an in-depth study of cardiac heterogeneity and cell-to-cell interactions can help elucidate the pathogenesis of diabetic myocardial fibrosis and identify treatment targets for the treatment of this disease. In this study, we investigated intercellular communication drivers of myocardial fibrosis in mouse heart with high-fat-diet/streptozotocin-induced diabetes at single-cell resolution. Intercellular and protein-protein interaction networks of fibroblasts and macrophages, endothelial cells, as well as fibroblasts and epicardial cells revealed critical changes in ligand-receptor interactions such as Pdgf(s)-Pdgfra and Efemp1-Egfr, which promote the development of a profibrotic microenvironment during the progression of and confirmed that the specific inhibition of the Pdgfra axis could significantly improve diabetic myocardial fibrosis. We also identified phenotypically distinct Hrchi and Postnhi fibroblast subpopulations associated with pathological extracellular matrix remodeling, of which the Hrchi fibroblasts were found to be the most profibrogenic under diabetic conditions. Finally, we validated the role of the Itgb1 hub gene-mediated intercellular communication drivers of diabetic myocardial fibrosis in Hrchi fibroblasts, and confirmed the results through AAV9-mediated Itgb1 knockdown in the heart of diabetic mice. In summary, cardiac cell mapping provides novel insights into intercellular communication drivers involved in pathological extracellular matrix remodeling during diabetic myocardial fibrosis.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Ratones , Animales , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , Miocardio/patología , Diabetes Mellitus Experimental/complicaciones , Células Endoteliales/patología , Análisis de Expresión Génica de una Sola Célula , Comunicación Celular , Fibrosis , Fibroblastos/patología
3.
Entropy (Basel) ; 25(2)2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36832735

RESUMEN

Linear codes with a few weights have been extensively studied due to their wide applications in secret sharing schemes, strongly regular graphs, association schemes, and authentication codes. In this paper, we choose the defining sets from two distinct weakly regular plateaued balanced functions, based on a generic construction of linear codes. Then we construct a family of linear codes with at most five nonzero weights. Their minimality is also examined and the result shows that our codes are helpful in secret sharing schemes.

4.
Acta Biomater ; 153: 481-493, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36162766

RESUMEN

Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) system adapted from bacteria is a programmable nuclease-based genome editing tool. The long-lasting effect of gene silencing or correction is beneficial in cancer treatment. Considering the need to broaden the practical application of this technology, highly efficient non-viral vectors are urgently required. We prepared a multifunctional non-viral vector that could actively target tumor cells and deliver CRISPR/Cas9 plasmids into nuclei of cancer cells. Protamine sulfate (PS) which contains nuclear localization sequence was utilized to condense plasmid DNA and facilitate nuclei-targeted delivery. Liposome-coated protein/DNA complex avoided the degradation of nuclease in blood circulation. The obtained PS@Lip/pCas9 was further modified with distearoyl phosphoethanolamine-polyethylene glycol-hyaluronic acid (HA) to endow the vector ability to actively target tumor cell. Results suggested that PS@HA-Lip could deliver CRISPR/Cas9 plasmids into nuclei of tumor cells and induce genome editing effect. With the disruption of MTH1 (mutT homolog1) gene, the growth of non-small cell lung cancer was inhibited. Moreover, cell apoptosis in tumor tissue was promoted, and liver metastasis of non-small cell lung cancer (NSCLC) was reduced. Our study has provided a therapeutic strategy targeting MTH1 gene for NSCLC therapy. STATEMENT OF SIGNIFICANCE: CRISPR/Cas9 as a powerful tool for genome editing has drawn much attention. The long-lasting effect possesses unique advantage in cancer treatment. Non-viral vectors have high loading capacity, high safety and low immunogenicity, playing an important role in CRISPR/Cas9 delivery. In our study, a multifunctional non-viral vector for the efficient delivery of CRISPR/Cas9 plasmid was constructed. With the active targeting ligand and nuclei-targeting component, the cargo was efficiently delivered into cell nuclei and exerted genome editing effect. By using this vector, we successfully inhibited the growth and induced the apoptosis of non-small cell lung cancer by disrupting MTH1 expression with good safety. Our work provided an efficient non-vial vector for CRISPR/Cas9 delivery and explored the possibility for cancer treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Sistemas CRISPR-Cas/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Vectores Genéticos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Edición Génica/métodos , ADN
5.
Adv Sci (Weinh) ; 9(16): e2105854, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35355446

RESUMEN

The blood-brain barrier (BBB) severely blocks the intracranial accumulation of most systemic drugs. Inspired by the contribution of the bacterial outer membrane to Escherichia coli K1 (EC-K1) binding to and invasion of BBB endothelial cells in bacterial meningitis, utilization of the BBB invasion ability of the EC-K1 outer membrane for brain-targeted drug delivery and construction of a biomimetic self-assembled nanoparticle with a surface featuring a lipopolysaccharide-free EC-K1 outer membrane are proposed. BBB penetration of biomimetic nanoparticles is demonstrated to occur through the transcellular vesicle transport pathway, which is at least partially dependent on internalization, endosomal escape, and transcytosis mediated by the interactions between outer membrane protein A and gp96 on BBB endothelial cells. This biomimetic nanoengineering strategy endows the loaded drugs with prolonged circulation, intracranial interstitial distribution, and extremely high biocompatibility. Based on the critical roles of gp96 in cancer biology, this strategy reveals enormous potential for delivering therapeutics to treat gp96-overexpressing intracranial malignancies.


Asunto(s)
Biomimética , Nanopartículas , Membrana Externa Bacteriana , Encéfalo , Células Endoteliales/metabolismo , Escherichia coli/metabolismo , Lipopolisacáridos/metabolismo , Nanopartículas/metabolismo
6.
Artif Cells Nanomed Biotechnol ; 49(1): 173-184, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33620265

RESUMEN

Poor cellular uptake and slow intracellular drug release remain the main barriers for the efficient application of micellar delivery system. Taking advantage of the overexpressed CD44 receptor and mild acidic microenvironment of tumour cells, CD44-targeted pH-responsive micelles based on the self-assembly of histidine-hyaluronic acid-dodecylamine (His-HA-DA) were prepared for the delivery of doxorubicin (DOX). These micelles exhibited pH-responsive behaviour with increased particle size, decreased encapsulation efficiency (EE%) of DOX and rapid release of DOX triggered by low pH. Compared with free DOX, DOX/HHD exhibited relatively high cellular uptake mainly via the CD44-mediated endocytosis. The on-demand intracellular release of DOX from DOX/HHD led to improved cytotoxicity. DOX/HHD also showed great penetration efficiency in 3D tumour spheres in vitro. Moreover, these micelles with suitable particle size gained excellent tumour-targeting effects, as well as improved anti-tumour effects and reduced side effects in vivo. In conclusion, these micelles with CD44 targeted and pH-responsive behaviours provide a promising strategy for the efficient delivery of anti-tumour drugs in vivo.


Asunto(s)
Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Liberación de Fármacos , Receptores de Hialuranos/metabolismo , Espacio Intracelular/metabolismo , Línea Celular Tumoral , Endocitosis , Humanos , Concentración de Iones de Hidrógeno , Cinética , Micelas
7.
Theranostics ; 9(20): 5886-5898, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31534526

RESUMEN

Targeting tumor angiogenesis pathway via VEGF siRNA (siVEGF) has shown great potential in treating highly malignant and metastatic non-small cell lung cancer (NSCLC). However, anti-angiogenic monotherapy lacked sufficient antitumor efficacy which suffered from malignant tumor proliferation. Therefore, the combined application of siVEGF and chemotherapeutic agents for simultaneous targeting of tumor proliferation and angiogenesis has been a research hotspot to explore a promising NSCLC therapy regimen. Methods: We designed, for the first time, a rational therapy strategy via intelligently co-delivering siVEGF and chemotherapeutics etoposide (ETO) by multi-functional nanoparticles (NPs) directed against the orthotopic NSCLC. These NPs consisted of cationic liposomes loaded with siVEGF and ETO and then coated with versatile polymer PEGylated histidine-grafted chitosan-lipoic acid (PHCL). We then comprehensively evaluated the anti-angiogenic and anti-proliferation efficiency in the in vitro tumor cell model and in bioluminescent orthotopic lung tumor bearing mice model. Results: The NPs co-delivering siVEGF and ETO exhibited tailor-made surface charge reversal features in mimicking tumor extracellular environment with improved internal tumor penetration capacity and higher cellular internalization. Furthermore, these NPs with flexible particles size triggered by intracellular acidic environment and redox environment showed pinpointed and sharp intracellular cargo release guaranteeing adequate active drug concentration in tumor cells. Enhanced VEGF gene expression silencing efficacy and improved tumor cell anti-proliferation effect were demonstrated in vitro. In addition, the PHCL layer improved the stability of these NPs in neutral environment allowing enhanced orthotopic lung tumor targeting efficiency in vivo. The combined therapy by siVEGF and ETO co-delivered NPs for orthotopic NSCLC simultaneously inhibited tumor proliferation and tumor angiogenesis resulting in more significant suppression of tumor growth and metastasis than monotherapy. Conclusion: Combined application of siVEGF and ETO by the multi-functional NPs with excellent and on-demand properties exhibited the desired antitumor effect on the orthotopic lung tumor. Our work has significant potential in promoting combined anti-angiogenesis therapy and chemotherapy regimen for clinical NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Etopósido/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/química , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Sistemas de Liberación de Medicamentos/métodos , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones Desnudos , Esferoides Celulares/citología , Esferoides Celulares/metabolismo
8.
ACS Appl Mater Interfaces ; 10(43): 36641-36651, 2018 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-30360129

RESUMEN

Drug development involves various evaluation processes to ascertain drug effects and rigorous analysis of biological indicators during in vitro preclinical studies. Two-dimensional (2D) cell cultures are commonly used in numerous in vitro studies, which are poor facsimiles of the in vivo conditions. Recently, three-dimensional (3D) tumor models mimicking the tumor microenvironment and reducing the use of experimental animals have been developed generating great interest to appraise tumor response to treatment strategies in cancer therapy. In this study, silk fibroin (SF) protein and chitosan (CS), two natural biomaterials, were chosen to construct the scaffolds of 3D cell models. Human non-small cell lung cancer A549 cells in the SF/CS scaffolds were found to have a great tendency to gather and form tumor spheres. A549 cell spheres in the 3D scaffolds showed biological and morphological characteristics much closer to the in vivo tumors. Besides, the cells in 3D models displayed better invasion ability and drug resistance than 2D models. Additionally, differences in drug-resistant and immune-related protein levels were found, which indicated that 3D models might resemble the real-life situation. These findings suggested that these 3D tumor models composed of SF/CS are promising to provide a valuable biomaterial platform in the evaluation of anticancer drugs.


Asunto(s)
Quitosano/química , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Fibroínas/química , Neoplasias Pulmonares/tratamiento farmacológico , Microambiente Tumoral , Células A549 , Antineoplásicos/farmacología , Materiales Biocompatibles , Línea Celular Tumoral , Movimiento Celular , Humanos , Microscopía Confocal , Invasividad Neoplásica , Porosidad , Andamios del Tejido
9.
Acta Biomater ; 81: 219-230, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30267887

RESUMEN

To bypass the biological barriers during the drug delivery process, it is desirable to develop smart nanoparticles (NPs) with flexible physical and chemical properties. In this study, a programmed NP delivery system with a pH-triggered detachable PEG layer and a lactobionic acid (Lac)-modified reduction-responsive core was developed to address the "PEG dilemma" and provide an on-demand intracellular release of doxorubicin (DOX). The positively charged DOX-loaded lactobionic acid-chitosan-lipoic acid (DOX/LCL) NPs were prepared and coated with a negatively charged dimethylmaleic acid-PEG-chitosan (PEG-CS-DA) layer to obtain a prolonged circulation time and improve the tumor-targeting effect. After reaching the tumor tissues through a targeted delivery effect, the surface charge of the PEG-CS-DA layer was reversed from negative to positive because of the trigger by the acidic microenvironment (pH 6.8), thus leading to the detachment of the PEG layer. The exposure of positive charges and the active targeting ligand enhanced cellular uptake and facilitated penetration into tumor tissues. Subsequently, the rapid release and diffusion of DOX into the nuclei was triggered by the intracellular high concentration of glutathione, thus leading to cell apoptosis. In conclusion, these programmed pH/reduction-responsive NPs provide a promising strategy for the delivery of antitumor agents in vivo. STATEMENT OF SIGNIFICANCE: In this study, novel programmed pH/reduction-responsive NPs were developed for the delivery of DOX in vivo. These NPs were coated with a negatively charged PEG layer to improve the serum stability and tumor target effect. The PEG layer detached because of the trigger by tumor acidic microenvironment (pH 6.8), thus leading to the exposure of positive charges and the active targeting ligand, which enhanced cellular uptake and facilitated penetration into tumor tissues. Subsequently, the rapid release of DOX was triggered by the intracellular high concentration of glutathione, thereby resulting in enhanced cytotoxicity. These programmed pH/reduction-responsive NPs provide a promising strategy for the delivery of antitumor agents in vivo.


Asunto(s)
Antineoplásicos , Doxorrubicina , Portadores de Fármacos , Nanopartículas , Neoplasias , Microambiente Tumoral , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Liberación de Fármacos , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Nanopartículas/química , Nanopartículas/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología
10.
AAPS J ; 20(2): 34, 2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29476273

RESUMEN

Low accumulation in tumor sites and slow intracellular drug release remain as the obstacles for nanoparticles to achieve effective delivery of chemotherapeutic drugs. In this study, multifunctional micelles were designed to deliver doxorubicin (Dox) to tumor sites to provide more efficient therapy against hepatic carcinoma. The micelles were based on pH-responsive carboxymethyl chitosan (CMCh) modified with a reactive oxygen species (ROS)-responsive segment phenylboronic acid pinacol ester (BAPE) and an active targeted ligand CD147 monoclonal antibody. The Dox-loaded micelles provided rapid and complete drug release in pH 5.3 incubation conditions with 1 mM H2O2. In addition, an in vitro cell uptake study revealed that CD147 modification significantly enhanced cellular internalization due to the high affinity to CD147 receptors, which are overexpressed on tumor cells. An in vivo study revealed that CD147-modified micellar formulations exhibited high accumulation in tumor sites and markedly enhanced antiproliferation effects with fewer side effects than other formulations. In conclusion, this CD147 receptor targeted delivery system with ROS/pH dual sensitivity provides a promising strategy for the treatment of hepatic carcinoma.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Carcinoma Hepatocelular/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hepáticas/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Basigina/inmunología , Línea Celular Tumoral , Preparaciones de Acción Retardada/administración & dosificación , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Micelas , Nanopartículas/química , Nanopartículas/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Pharm ; 15(1): 314-325, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29250957

RESUMEN

In this article, a novel graft polymeric micelle with targeting function ground on aptamer AS1411 was synthesized. The micelle was based on chitosan-ss-polyethylenimine-urocanic acid (CPU) with dual pH/redox sensitivity and targeting effects. This micelle was produced for codelivering Toll-like receptor 4 siRNA (TLR4-siRNA) and doxorubicin (Dox). In vitro investigation revealed the sustained gene and drug release from Dox-siRNA-loaded micelles under physiological conditions, and this codelivery nanosystem exhibited high dual pH/redox sensitivity, rapid intracellular drug release, and improved cytotoxicity against A549 cells in vitro. Furthermore, the micelles loaded with TLR4-siRNA inhibited the migration and invasion of A549. Excellent tumor penetrating efficacy was also noted in the A549 tumor spheroids and solid tumor slices. In vivo, multiple results demonstrated the excellent tumor-targeting ability of AS1411-chitosan-ss-polyethylenimine-urocanic acid (ACPU) micelle in tumor tissues. The micelles exhibited excellent antitumor efficacy and low toxicity in the systemic circulation in lung-tumor-bearing BALB/c mice. These results conclusively demonstrated the great potential of the new graft copolymer micelle with targeting function for the targeted and efficient codelivery of chemotherapeutic drugs and genes in cancer treatment.


Asunto(s)
Micelas , Fosfoproteínas/metabolismo , Polímeros/química , Proteínas de Unión al ARN/metabolismo , Células A549 , Animales , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Liberación de Fármacos , Humanos , Ratones , Fosfoproteínas/química , ARN Interferente Pequeño , Proteínas de Unión al ARN/química , Nucleolina
12.
Mater Sci Eng C Mater Biol Appl ; 82: 234-243, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025653

RESUMEN

In this research, a charge-conversional polymer, poly-l-lysine-lipoic acid (PLL-LA), was prepared by dimethylmaleic anhydride (DA) modification and applied as a carrier with enhanced cell internalization and intracellular pH- and reduction-triggered doxorubicin (Dox) release. The surface charge of dimethylmaleic anhydride-poly-l-lysine-lipoic acid micelles (DA-PLL-LA) was negative at physiological pH and reversed to positive at the extracellular and intracellular pH of cancer cells. At tumor extracellular pH of 6.8, the conjugates underwent a rapid charge-reversible process with almost 80% DA cleavage within 2h, and then endocytosed into the endo/lysosomes more rapidly than at physiological pH of 7.4. The Dox/DA-PLL-LA micelles (Dox-micelles) demonstrated a sustained drug release in vitro under physiological condition, and rapid Dox release was triggered by both extracellular pH and high-concentration reducing glutathione. The Dox-micelles also exhibited enhanced internalization at extracellular pH, rapid intracellular drug release, and improved cytotoxicity against A549 cells in vitro. Excellent tumor-penetrating efficacy was also found in A549 tumor spheroids and solid tumor slices. Moreover, the DA-PLL-LA micelles exhibited excellent tumor-targeting ability in tumor tissues and excellent antitumor efficacy and low systemic toxicity in breast tumor-bearing mice. Therefore, the DA-PLL-LA micelles demonstrated great potential for targeted and efficient drug delivery in cancer treatments.


Asunto(s)
Antibióticos Antineoplásicos/química , Doxorrubicina/química , Portadores de Fármacos/química , Polímeros/química , Células A549 , Animales , Antibióticos Antineoplásicos/uso terapéutico , Antibióticos Antineoplásicos/toxicidad , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/uso terapéutico , Doxorrubicina/toxicidad , Portadores de Fármacos/síntesis química , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Micelas , Microscopía Confocal , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Oxidación-Reducción , Fagocitosis/efectos de los fármacos , Polilisina/química , Polímeros/síntesis química , Ácido Tióctico/química , Trasplante Heterólogo
13.
J Control Release ; 268: 198-211, 2017 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-29061511

RESUMEN

Malignant proliferation and metastasis in non-small cell lung carcinoma (NSCLC) are great challenges for effective clinical treatment through conventional chemotherapy. The combinational therapy strategy of RNA interfering (RNAi) technology and chemotherapeutic agents have been reported to be promising for effective cancer therapy. In this study, based on multifunctional nanoparticles (NPs), the simultaneous delivery of etoposide (ETP) and anti-Enhancer of Zeste Homologue 2 (EZH2) siRNA for the effective treatment of orthotopic lung tumor was achieved. The NPs exhibited pH/redox dual sensitivity verified by particle size changes, morphological changes, and in vitro release of drugs. Confocal microscopy analysis confirmed that the NPs exhibited endosomal escape property and on-demand intracellular drug release behavior, which can protect siRNA from degradation and facilitate the chemotherapeutic effect respectively. In vitro tumor cell motility study demonstrated that EZH2 siRNA loaded in NPs can decrease the migration and invasion capabilities of tumor cells by downregulating the expression of EZH2 mRNA and protein. In particular, an antiproliferation study revealed that the co-delivery of siRNA and ETP in the multifunctional NPs can induce a synergistic therapeutic effect on NSCLC. In vivo targeting evaluation showed that cRGDyC-PEG modification on NPs exhibited a low distribution in normal organs and an obvious accumulation in orthotopic lung tumor. Furthermore, targeted NPs co-delivering siRNA and ETP showed superior inhibition on tumor growth and metastasis and produced minimal systemic toxicity. These findings indicated that multifunctional NPs can be utilized as a co-delivery system, and that the combination of EZH2 siRNA and ETP can effectively treat NSCLC.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteína Potenciadora del Homólogo Zeste 2/genética , Etopósido/administración & dosificación , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Células A549 , Animales , Antineoplásicos Fitogénicos/química , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Terapia Combinada , Liberación de Fármacos , Etopósido/química , Femenino , Humanos , Ratones Desnudos , Nanopartículas/química , Polímeros/administración & dosificación , Polímeros/química , ARN Interferente Pequeño/química
14.
Int J Nanomedicine ; 12: 4241-4256, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28652730

RESUMEN

Physicochemical properties, including particle size, zeta potential, and drug release behavior, affect targeting efficiency, cellular uptake, and antitumor effect of nanocarriers in a formulated drug-delivery system. In this study, a novel stepwise pH-responsive nanodrug delivery system was developed to efficiently deliver and significantly promote the therapeutic effect of doxorubicin (DOX). The system comprised dimethylmaleic acid-chitosan-urocanic acid and elicited stepwise responses to extracellular and intracellular pH. The nanoparticles (NPs), which possessed negative surface charge under physiological conditions and an appropriate nanosize, exhibited advantageous stability during blood circulation and enhanced accumulation in tumor sites via enhanced permeability and retention effect. The tumor cellular uptake of DOX-loaded NPs was significantly promoted by the first-step pH response, wherein surface charge reversion of NPs from negative to positive was triggered by the slightly acidic tumor extracellular environment. After internalization into tumor cells, the second-step pH response in endo/lysosome acidic environment elicited the on-demand intracellular release of DOX from NPs, thereby increasing cytotoxicity against tumor cells. Furthermore, stepwise pH-responsive NPs showed enhanced antiproliferation effect and reduced systemic side effect in vivo. Hence, the stepwise pH-responsive NPs provide a promising strategy for efficient delivery of antitumor agents.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/administración & dosificación , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Quitosano/química , Doxorrubicina/química , Doxorrubicina/farmacocinética , Portadores de Fármacos/química , Evaluación Preclínica de Medicamentos/métodos , Liberación de Fármacos , Femenino , Concentración de Iones de Hidrógeno , Malonatos/química , Ratones Endogámicos BALB C , Nanopartículas/química , Tamaño de la Partícula , Distribución Tisular , Ácido Urocánico/química
15.
Int J Nanomedicine ; 12: 3375-3393, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28490877

RESUMEN

Multidrug resistance (MDR) is a major obstacle for the clinical therapy of malignant human cancers. The discovery of RNA interference provides efficient gene silencing within tumor cells for reversing MDR. In this study, a new "binary polymer" low-density lipoprotein-N-succinyl chitosan-cystamine-urocanic acid (LDL-NSC-SS-UA) with dual pH/redox sensitivity and targeting effect was synthesized for the co-delivery of breast cancer resistance protein small interfering RNA (siRNA) and paclitaxel (PTX). In vivo, the co-delivering micelles can accumulate in tumor tissue via the enhanced permeability and retention effect and the specific recognition and combination of LDL and LDL receptor, which is overexpressed on the surface of tumor cell membranes. The siRNA-PTX-loaded micelles inhibited gene and drug release under physiological conditions while promoting fast release in an acid microenvironment or in the presence of glutathione. The micelles escaped from the lysosome through the proton sponge effect. Additionally, the micelles exhibited superior antitumor activity and downregulated the protein and mRNA expression levels of breast cancer resistance protein in MCF-7/Taxol cells. The biodistribution and antitumor studies proved that the siRNA-PTX-loaded micelles possessed prolonged circulation time with a remarkable tumor-targeting effect and effectively inhibited tumor growth. Therefore, the novel dual pH/redox-sensitive polymers co-delivering siRNA and PTX with excellent biocompatibility and effective reversal of MDR demonstrate a considerable potential in cancer therapy.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Portadores de Fármacos/química , Paclitaxel/administración & dosificación , Polímeros/química , ARN Interferente Pequeño/administración & dosificación , Animales , Línea Celular Tumoral , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Femenino , Humanos , Concentración de Iones de Hidrógeno , Lipoproteínas LDL , Células MCF-7/efectos de los fármacos , Ratones Desnudos , Micelas , Oxidación-Reducción , Paclitaxel/química , Paclitaxel/farmacocinética , Polímeros/administración & dosificación , Interferencia de ARN , ARN Interferente Pequeño/genética , Distribución Tisular
16.
J Biomed Mater Res B Appl Biomater ; 105(5): 1114-1125, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-27008163

RESUMEN

The development of effective and stable carriers of small interfering RNA (siRNA) is important for treating cancer with multidrug resistance (MDR). We developed a new gene and drug co-delivery system and checked its characteristics. Low-density lipoprotein (LDL) was coupled with N-succinyl chitosan (NSC) Lipoic acid (LA) micelles and co-delivered MDR1 siRNA and paclitaxel (PTX-siRNA/LDL-NSC-LA) to enhance antitumor effects by silencing the MDR gene of tumors (Li et al., Adv Mater 2014;26:8217-8224). In our study, we developed a new type of containing paclitaxel-loaded micelles and siRNA-loaded LDL nanoparticle. This "binary polymer" is pH and reduction dual-sensitive core-crosslinked micelles. PTX-siRNA/LDL-NSC-LA had an average particle size of (171.6 ± 6.42) nm, entrapment efficiency of (93.92 ± 1.06) %, and drug-loading amount of (12.35% ± 0.87) %. In vitro, MCF-7 cells, high expressed LDL receptor, were more sensitive to this delivery system than to taxol® and cell activity was inhibited significantly. Fluorescence microscopy showed that PTX-siRNA/LDL-NSC-LA was uptaken very conveniently and played a key role in antitumor activity. PTX-siRNA/LDL-NSC-LA protected the siRNA from degradation by macrophage phagocytosis and evidently down-regulated the level of mdr1 mRNA as well as the expression of P-gp. We tested the target ability of PTX-siRNA/LDL-NSC-LA in vivo in tumor-bearing nude mice. Results showed that this system could directly deliver siRNA and PTX to cancer cells. Thus, new co-delivering siRNA and antitumor drugs should be explored for solving MDR in cancer. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1114-1125, 2017.


Asunto(s)
Quitosano , Técnicas de Transferencia de Gen , Lipoproteínas LDL , Micelas , Proteínas de Neoplasias , Neoplasias Experimentales , Paclitaxel/farmacología , ARN Interferente Pequeño , Ácido Tióctico , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/biosíntesis , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Quitosano/química , Quitosano/farmacología , Femenino , Humanos , Lipoproteínas LDL/química , Lipoproteínas LDL/farmacología , Células MCF-7 , Ratones , Ratones Desnudos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , ARN Interferente Pequeño/biosíntesis , ARN Interferente Pequeño/genética , Ácido Tióctico/química , Ácido Tióctico/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
J Biomed Mater Res B Appl Biomater ; 105(7): 2093-2106, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-27405391

RESUMEN

P-glycoprotein (P-gp) plays an importantrole in multidrug resistance (MDR), proved to be one of the major obstacles in cancer chemotherapy. Cationic polymers could specifically deliver siRNA to tumor cells and thus reverse MDR by the downregulation of P-gp. In this study, a triblock copolymer micelle was prepared based on the polymer of N-succinyl-chitosan-poly-l-lysine-palmitic acid (NSC-PLL-PA) to deliver siRNA-P-gp (siRNA-micelle) or doxorubicin (Dox-micelle). The resulting micelle exhibited an efficient binding ability for siRNA and high encapsulation efficiency for Dox, with an average particle size of ∼170 nm. siRNA-micelle and Dox-micellewere instable at low pH, thereby enhancing tumor accumulation and intracellular release of the encapsulated siRNA and Dox. siRNA-micelle micelles could enhance the knockdown efficacy of siRNA by improving the transfection efficiency, downregulating P-gp expression, and passing the drug efflux transporters, thereby improving the therapeutic effects of Dox-micelle. However, P-gp could transfer from HepG2/ADM to HepG2 cells independent of the expression of mdr1, and the acquired resistance could permit tumor cells to survive and develop intrinsic P-gp-mediated resistance, thereby limiting the desired efficiency of chemotherapeutics. This study demonstrated the effectiveness of siRNA-micelle for tumor-targeted delivery, MDR reversal, and provided an effective strategy for the treatment of cancers that develop MDR. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2093-2106, 2017.


Asunto(s)
Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Micelas , Proteínas de Neoplasias , Neoplasias , ARN Interferente Pequeño , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Células Hep G2 , Humanos , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
18.
ACS Appl Mater Interfaces ; 8(47): 32146-32158, 2016 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-27933846

RESUMEN

The efficient delivery of antitumor agents to tumor sites faces numerous obstacles, such as poor cellular uptake and slow intracellular drug release. In this regard, smart nanoparticles (NPs) that respond to the unique microenvironment of tumor tissues have been widely used for drug delivery. In this study, novel charge-reversal and reduction-responsive histidine-grafted chitosan-lipoic acid NPs (HCSL-NPs) were selected for efficient therapy of breast cancer by enhancing cell internalization and intracellular pH- and reduction-triggered doxorubicin (DOX) release. The surface charge of HCSL-NPs presented as negative at physiological pH and reversed to positive at the extracellular and intracellular pH of the tumor. In vitro release investigation revealed that DOX/HCSL-NPs demonstrated a sustained drug release under the physiological condition, whereas rapid DOX release was triggered by both endolysosome pH and high-concentration reducing glutathione (GSH). These NPs exhibited enhanced internalization at extracellular pH, rapid intracellular drug release, and improved cytotoxicity against 4T1 cells in vitro. Excellent tumor penetrating efficacy was also found in 4T1 tumor spheroids and solid tumor slices. In vivo experiments demonstrated that HCSL-NPs exhibited excellent tumor-targeting ability in tumor tissues as well as excellent antitumor efficacy and low systemic toxicity in breast tumor-bearing BALB/c mice. These results indicated that the novel charge-reversal and reduction-responsive HCSL-NPs have great potential for targeted and efficient delivery of chemotherapeutic drugs in cancer treatments.


Asunto(s)
Nanopartículas , Animales , Doxorrubicina , Portadores de Fármacos , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C
19.
Int J Pharm ; 511(2): 728-40, 2016 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-27484835

RESUMEN

Internal stimuli, such as intracellular lysosomal pH, enzyme, redox and reduction, can be applied to improve biological specificity of chemotherapeutic drugs for cancer therapy. Thus, functionalized copolymers based on their response to specific microenvironment of tumor regions have been designed as smart drug vesicles for enhanced anti-cancer efficiency and reduced side effects. Herein, we reported dually pH/reduction-responsive novel micelles based on self-assembly of carboxymethyl chitosan-cysteamine-N-acetyl histidine (CMCH-SS-NA) and doxorubicin (DOX). The tailor-made dually responsive micelles demonstrated favorable stability in normal physiological environment and triggered rapid drug release in acidic and/or reduction environment. Additionally, the nanocarriers responded to the intracellular environment in an ultra-fast manner within several minutes, which led to the pinpointed release of DOX in tumor cells effectively and ensured higher DOX concentrations within tumor areas with the aid of targeted delivery, thereby leading to enhanced tumor ablation. Thus, this approach with sharp drug release behavior represented a versatile strategy to provide a promising paradigm for cancer therapy.


Asunto(s)
Doxorrubicina/farmacología , Doxorrubicina/farmacocinética , Micelas , Microambiente Tumoral/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Quitosano/análogos & derivados , Quitosano/química , Cisteamina/química , Doxorrubicina/química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Femenino , Histidina/química , Concentración de Iones de Hidrógeno , Hígado/metabolismo , Ratones , Oxidación-Reducción , Ratas , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Sci Rep ; 6: 23859, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27030638

RESUMEN

Co-delivery of chemotherapeutics and siRNA with different mechanisms in a single system is a promising strategy for effective cancer therapy with synergistic effects. In this study, a triblock copolymer micelle was prepared based on the polymer of N-succinyl chitosan-poly-L-lysine-palmitic acid (NSC-PLL-PA) to co-deliver doxorubicin (Dox) and siRNA-P-glycoprotein (P-gp) (Dox-siRNA-micelle). Dox-siRNA-micelle was unstable in pH 5.3 medium than in pH 7.4 medium, which corresponded with the in vitro rapid release of Dox and siRNA in acidic environments. The antitumor efficacy of Dox-siRNA-micelle in vitro significantly increased, especially in HepG2/ADM cells, which was due to the downregulation of P-gp. Moreover, almost all the Dox-siRNA-micelles accumulated in the tumor region beyond 24 h post-injection, and the co-delivery system significantly inhibited tumor growth with synergistic effects in vivo. This study demonstrated the effectiveness of Dox-siRNA-micelles in tumor-targeting and MDR reversal, and provided a promising strategy to develop a co-delivery system with synergistic effects for combined cancer therapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Antibióticos Antineoplásicos/farmacología , Terapia Combinada/métodos , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos/métodos , Neoplasias Hepáticas/terapia , ARN Interferente Pequeño/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Expresión Génica , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , Ratones Desnudos , Micelas , ARN Interferente Pequeño/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...