Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Pharm Sci ; 110(2): 876-887, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33166581

RESUMEN

Prostate cancer is the most common malignant tumor with bone metastasis, and there is still no ideal treatment for bone metastasis of prostate cancer. In this study, a pH and GSH dual sensitive calcium phosphate-polymer hybrid nanoparticle (DTX@Cap/HP) was prepared to co-deliver zoledronate (ZOL) and docetaxel (DTX) to treat bone metastasis of prostate cancer. DTX@Cap/HP exhibited high bone binding affinity and released more DTX and ZOL in acidic and high GSH concentration environment. A large amount of DTX@Cap/HP was uptaken by PC-3 cell in acidic medium than that in neutral medium. DTX@Cap/HP obviously reduced PC-3 cell proliferation and bone lesion in in-vitro 3D model of bone metastases of prostate cancer. Besides, DTX@Cap/HP also exhibited stronger anti bone metastases of prostate cancer activity in vivo as compared with the same dose of DTX + ZOL, which resulted from the co-delivery of DTX and ZOL to bone metastases of prostate cancer by DTX@Cap/HP and the synergistic effects of DTX and ZOL. DTX@Cap/HP has great potential in the treatment of bone metastases of prostate cancer.


Asunto(s)
Antineoplásicos , Nanopartículas , Neoplasias de la Próstata , Antineoplásicos/uso terapéutico , Fosfatos de Calcio , Línea Celular Tumoral , Docetaxel , Humanos , Masculino , Polímeros , Neoplasias de la Próstata/tratamiento farmacológico , Ácido Zoledrónico/uso terapéutico
3.
Nanomedicine (Lond) ; 15(9): 833-849, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32163008

RESUMEN

Aim: To prepare pH-sensitive nanoparticle composed of alendronate (ALN) and poly(amidoamine) (PAMAM) to treat bone metastases of lung cancer. Methods: The solvent evaporation method was used to prepare docetaxel (DTX)-loaded ALN-PAMAM nanoparticles (DTX@ALN-PAMAM). Results: The in vitro results showed DTX@ALN-PAMAM significantly enhanced the anticancer activity of DTX and inhibited the formation of osteoclasts. DTX@ALN-PAMAM concentrated at bone metastasis site in mice, which resulted in the suppression of bone resorption, pain response and growth of bone metastases. Eventually, the therapeutic effect of DTX on bone metastases of lung cancer was obviously improved. Conclusion: ALN modified PAMAM nanoparticle could be an effective platform for the treatment of bone metastases of lung cancer.


Asunto(s)
Antineoplásicos , Neoplasias Óseas , Neoplasias Pulmonares , Nanopartículas , Alendronato , Animales , Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Docetaxel , Sistemas de Liberación de Medicamentos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Poliaminas
4.
Nanomedicine ; 21: 102054, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31310809

RESUMEN

Bone is one of the prone metastatic sites of lung cancer. Osteoclast plays an important role in bone resorption and the growth of bone metastases of lung cancer. In order to treat bone metastases of lung cancer, we reported a docetaxel (DTX)-loaded nanoparticle, DTX@AHP, which could target dually at osteoclasts and bone metastatic tumor cells. The in vitro drug release from DTX@AHP exhibited pH and redox responsive characteristics. DTX@AHP displayed high binding affinity with bone matrix. In addition, DTX@AHP significantly inhibited the differentiation of RAW264.7 into osteoclast and effectively inhibited the proliferation of osteoclasts and tumor cells in in-vitro 3D bone metastases model of lung cancer. DTX@AHP could accumulate in bone metastases sites in vivo. Consequently, DTX@AHP not only markedly inhibited the growth of bone metastases of lung cancer but also reduced osteolysis in tumor-bearing mice. DTX@AHP exhibited great potential in the treatment of bone metastases of lung cancer.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Docetaxel/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Nanopartículas/administración & dosificación , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Proliferación Celular/efectos de los fármacos , Docetaxel/química , Sistemas de Liberación de Medicamentos , Liberación de Fármacos , Xenoinjertos , Humanos , Neoplasias Pulmonares/patología , Ratones , Nanopartículas/química , Metástasis de la Neoplasia , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteólisis/inducido químicamente , Células RAW 264.7
5.
Nanomedicine (Lond) ; 14(8): 1011-1032, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30925116

RESUMEN

AIM: The objective of this study was to deliver a ring-closed form of 10-hydroxycamptothecin (HCPT) to the mitochondria and nucleus to treat colorectal cancer. MATERIALS & METHODS: HCPT-loaded nanoparticle HCPT@PLGA-PEG2k-triphenylphosphonium/PLGA-hyd-PEG4k-folic acid (PT/PHF) and HCPT@PT/PLGA-SS-PEG4k-folic acid (PSF) were prepared by using emulsion-solvent evaporation method. RESULTS: In vitro experimental results indicated HCPT@PT/PHF and HCPT@PT/PSF maintained a large amount of HCPT in active form, and delivered more HCPT to the nucleus and mitochondria of the tumor cell, which resulted in the enhancement of cytotoxicity of HCPT. In vivo experimental results indicated that HCPT@PT/PHF and HCPT@PT/PSF delivered more ring-closed form of HCPT to tumor tissue, which led to strong antitumor activity. CONCLUSION: HCPT@PT/PHF and HCPT@PT/PSF could enhance therapeutic efficacy of HCPT to colorectal cancer.


Asunto(s)
Camptotecina/análogos & derivados , Núcleo Celular/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Portadores de Fármacos/química , Mitocondrias/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Transporte Biológico , Camptotecina/farmacología , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Liberación de Fármacos , Emulsiones/química , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Ratones , Ratones Desnudos , Modelos Animales , Nanopartículas/química , Tamaño de la Partícula , Poliésteres/química , Polietilenglicoles/química , Propiedades de Superficie , Distribución Tisular
6.
Nanomedicine ; 16: 236-249, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30639669

RESUMEN

Efficient delivery of antioxidant drugs into mitochondria of ischemic cardiomyocytes where reactive oxygen species largely induced is a major challenge for precise treatment of myocardial ischemia-reperfusion injury. Herein, we report a smart dual-shell polymeric nanoparticle, MCTD-NPs, which utilizes multistage continuous targeted strategy to deliver reactive oxygen species scavenger specifically to mitochondria of ischemic cardiomyocytes upon systemic administration. In vitro experiments indicated that the intracellular uptake of MCTD-NPs was specifically enhanced in hypoxia reoxygenation injured H9c2 cells. MCTD-NPs selectively delivered resveratrol to mitochondria of hypoxia reoxygenation injured H9c2 cells. In addition, MCTD-NPs increased the viability of H/R injured H9c2 cell through eliminating mitochondrial ROS, decreasing mPTP opening and blocking mitochondria-dependent apoptotic pathway. In vivo experiments revealed that MCTD-NPs increased the distribution of resveratrol in the ischemic myocardium and subsequently reduced infarct size in MI/RI rats. These results demonstrated a novel platform for specific delivery of antioxidant to mitochondria to treat MI/RI.


Asunto(s)
Antioxidantes/uso terapéutico , Mitocondrias/metabolismo , Animales , Antioxidantes/administración & dosificación , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular , Humanos , Etiquetado Corte-Fin in Situ , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo
7.
J Nanobiotechnology ; 17(1): 18, 2019 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683110

RESUMEN

BACKGROUND: Cyclosporin A (CsA) is a promising therapeutic drug for myocardial ischemia reperfusion injury (MI/RI) because of its definite inhibition to the opening of mitochondrial permeability transition pore (mPTP). However, the application of cyclosporin A to treat MI/RI is limited due to its immunosuppressive effect to other normal organ and tissues. SS31 represents a novel mitochondria-targeted peptide which can guide drug to accumulate into mitochondria. In this paper, mitochondria-targeted nanoparticles (CsA@PLGA-PEG-SS31) were prepared to precisely deliver cyclosporin A into mitochondria of ischemic cardiomyocytes to treat MI/RI. RESULTS: CsA@PLGA-PEG-SS31 was prepared by nanoprecipitation. CsA@PLGA-PEG-SS31 showed small particle size (~ 50 nm) and positive charge due to the modification of SS31 on the surface of nanoparticles. CsA@PLGA-PEG-SS31 was stable for more than 30 days and displayed a biphasic drug release pattern. The in vitro results showed that the intracellular uptake of CsA@PLGA-PEG-SS31 was significantly enhanced in hypoxia reoxygenation (H/R) injured H9c2 cells. CsA@PLGA-PEG-SS31 delivered CsA into mitochondria of H/R injured H9c2 cells and subsequently increased the viability of H/R injured H9c2 cell through inhibiting the opening of mPTP and production of reactive oxygen species. In vivo results showed that CsA@PLGA-PEG-SS31 accumulated in ischemic myocardium of MI/RI rat heart. Apoptosis of cardiomyocyte was alleviated in MI/RI rats treated with CsA@PLGA-PEG-SS31, which resulted in the myocardial salvage and improvement of cardiac function. Besides, CsA@PLGA-PEG-SS31 protected myocardium from damage by reducing the recruitment of inflammatory cells and maintaining the integrity of mitochondrial function in MI/RI rats. CONCLUSION: CsA@PLGA-PEG-SS31 exhibited significant cardioprotective effects against MI/RI in rats hearts through protecting mitochondrial integrity, decreasing apoptosis of cardiomyocytes and myocardial infract area. Thus, CsA@PLGA-PEG-SS31 offered a promising therapeutic method for patients with acute myocardial infarction.


Asunto(s)
Ciclosporina/administración & dosificación , Ciclosporina/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Mitocondrias/metabolismo , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Oligopéptidos/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Ciclosporina/farmacocinética , Ciclosporina/farmacología , Modelos Animales de Enfermedad , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Poro de Transición de la Permeabilidad Mitocondrial , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Tamaño de la Partícula , Ratas
8.
Mol Pharm ; 15(3): 1296-1308, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29432025

RESUMEN

The experiment aims to increase antitumor activity while decreasing the systemic toxicity of doxorubicin (DOX). Charge reversible and mitochondria/nucleus dual target lipid hybrid nanoparticles (LNPs) was prepared. The in vitro experimental results indicated that LNPs released more amount of DOX in acidic environment and delivered more amount of DOX to the mitochondria and nucleus of tumor cells than did free DOX, which resulted in the reduction of mitochondrial membrane potential and the enhancement of cytotoxicity of LNPs on tumor cells. Furthermore, the in vivo experimental results indicated that LNPs delivered more DOX to tumor tissue and significantly prolonged the retention time of DOX in tumor tissue as compared with free DOX, which consequently resulted in the high antitumor activity and low systemic toxicity of LNPs on tumor-bearing nude mice. The above results indicated that charge reversible mitochondria/nucleus dual targeted lipid hybrid nanoparticles greatly enhanced therapeutic efficacy of DOX for treating lung cancer.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Núcleo Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Portadores de Fármacos/química , Mitocondrias/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Línea Celular Tumoral , Femenino , Humanos , Concentración de Iones de Hidrógeno , Lípidos/química , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Desnudos , Nanopartículas/química , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Artif Cells Nanomed Biotechnol ; 46(sup1): 380-391, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29336169

RESUMEN

In order to inhibit the growth of lung cancer bone metastasis and reduce the bone resorption at bone metastasis sites, a bone metastasis target micelle DOX@DBMs-ALN was prepared. The size and the zeta potential of DOX@DBNs-ALN were about 60 nm and -15 mV, respectively. DOX@DBMs-ALN exhibited high binding affinity with hydroxyapatite and released DOX in redox-responsive manner. DOX@DBMs-ALN was effectively up taken by A549 cells and delivered DOX to the nucleus of A549 cells, which resulted in strong cytotoxicity on A549 cells. The in vivo experimental results indicated that DOX@DBMs-ALN specifically delivered DOX to bone metastasis site and obviously prolonged the retention time of DOX in bone metastasis site. Moreover, DOX@DBMs-ALN not only significantly inhibited the growth of bone metastasis tumour but also obviously reduced the bone resorption at bone metastasis sites without causing marked systemic toxicity. Thus, DOX@DBMs-ALN has great potential in the treatment of lung cancer bone metastasis.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Resorción Ósea/tratamiento farmacológico , Doxorrubicina/química , Doxorrubicina/farmacología , Neoplasias Pulmonares/patología , Micelas , Células A549 , Animales , Neoplasias Óseas/patología , Proliferación Celular/efectos de los fármacos , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Femenino , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Oxidación-Reducción/efectos de los fármacos , Fosfatidiletanolaminas/química , Distribución Tisular
10.
Nanomedicine ; 14(3): 991-1003, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29339188

RESUMEN

In order to enhance the penetration of small interference RNA against the polo-like kinase I (siPLK1) across BBB to treat glioblastoma (GBM), transferrin (Tf) modified magnetic nanoparticle (Tf-PEG-PLL/MNP@siPLK1) was prepared. The in vitro experiments indicated that Tf-PEG-PLL/MNP@siPLK1 enhanced the cellular uptake of siPLK1, which resulted in an increase of gene silencing effect and cytotoxicity of Tf-PEG-PLL/MNP@siPLK1 on U87 cells. Besides, Tf-PEG-PLL/MNP@siPLK1 significantly inhibited the growth of U87 glioblastoma spheroids and markedly increased the BBB penetration efficiency of siPLK1 with the application of external magnetic field in in-vitro BBB model. The in vivo experiments indicated that siPLK1 selectively accumulated in the brain tissue, and markedly reduced tumor volume and prolonged the survival time of GBM-bearing mice after Tf-PEG-PLL/MNP@siPLK1 was injected to GBM-bearing mice via tail vein. The above data indicated that magnet and transferrin co-modified nanoparticle enhanced siPLK1 penetration across BBB and increased its anti GBM activity in vivo.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Silenciador del Gen , Glioblastoma/terapia , Nanopartículas de Magnetita/administración & dosificación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Transferrina/química , Animales , Barrera Hematoencefálica/patología , Ciclo Celular , Proteínas de Ciclo Celular/administración & dosificación , Proteínas de Ciclo Celular/genética , Supervivencia Celular , Endocitosis , Glioblastoma/genética , Glioblastoma/patología , Nanopartículas de Magnetita/química , Ratones , Tamaño de la Partícula , Proteínas Serina-Treonina Quinasas/administración & dosificación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/genética , Quinasa Tipo Polo 1
11.
Oncotarget ; 8(26): 42772-42788, 2017 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-28514759

RESUMEN

Bcl-2 gene is an important target to treat lung cancer. The small interference RNA (siRNA) of Bcl-2 gene (siBcl-2) can specifically silence Bcl-2 gene. However, naked siBcl-2 is difficult to accumulate in the tumor tissue to exert its activity. In this paper, a calcium phosphate lipid hybrid nanoparticle that possessed charge reversible property was prepared to enhance the activity of siBcl-2 in vivo. The average diameter and zeta potential of siBcl-2 loaded calcium phosphate lipid hybrid nanoparticles (LNPS@siBcl-2) were 80 nm and -13 mV at pH7.4 whereas the diameter and zeta potential changed to 1506 nm and +9 mV at pH5.0. LNPS@siBcl-2 could efficiently deliver siBcl-2 to the cytoplasm and significantly decreased the expression of Bcl-2 in A549 cells. Moreover, the in vivo experimental results showed that most of the Cy5-siBcl-2 accumulated in tumor tissue after LNPS@Cy5-siBcl-2 was administered to tumor-bearing mice by tail vein injection. Meanwhile, the expression of Bcl-2 was decreased but the expression of the BAX and Caspase-3 was increased in tumor tissue. LNPS@siBcl-2 significantly inhibited the growth of tumor in tumor-bearing mice without any obvious systemic toxicity. Thus, the charge reversible calcium phosphate lipid hybrid nanoparticle was an excellent siBcl-2 delivery carrier to improve the activity of siBcl-2 in vivo. LNPS@siBcl-2 has potential in the treatment of lung cancer.


Asunto(s)
Fosfatos de Calcio/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Animales , Fosfatos de Calcio/química , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Interferente Pequeño/química
12.
Nanoscale ; 9(19): 6264-6277, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28470315

RESUMEN

Bone is an especially prone metastatic site for breast cancer, and to block the vicious cycle between bone resorption and tumor growth is an important strategy for the treatment of breast cancer bone metastasis. In this paper, pH- and redox-sensitive as well as breast cancer bone metastasis-targeting nanoparticles (DOX@ALN-(HA-PASP)CL) were prepared, and also their anti-tumor activity and anti-bone resorption effect were investigated in detail. The in vitro experimental results indicated that DOX released from DOX@ALN-(HA-PASP)CL exhibited a GSH-, DTT- and pH-dependent manner. Moreover, in an in vitro 3D breast cancer bone metastasis model, DOX@ALN-(HA-PASP)CL decreased bone resorption through inhibiting the proliferation of human breast cancer cells (MDA-MB-231 cells) and reducing the activity of osteoclasts. The in vivo experimental results indicated that a large amount of DOX was delivered to a breast cancer bone metastasis site after tumor-bearing mice were treated with DOX@ALN-(HA-PASP)CL; meanwhile, DOX@ALN-(HA-PASP)CL significantly decreased the tumor volume and bone resorption in tumor-bearing mice without causing obvious systemic toxicity. In conclusion, the in vitro and in vivo experimental results indicate that DOX@ALN-(HA-PASP)CL has great potential in the treatment of breast cancer bone metastasis.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Resorción Ósea/tratamiento farmacológico , Neoplasias de la Mama/patología , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Nanopartículas , Animales , Neoplasias Óseas/secundario , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Femenino , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Desnudos , Oxidación-Reducción , Células RAW 264.7 , Ratas
13.
Mol Pharm ; 14(3): 746-756, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28146635

RESUMEN

Doxorubicin (DOX) is a broad-spectrum chemotherapy drug to treat tumors. However, severe side effects and development of DOX resistance hinder its clinical application. In order to overcome DOX resistance, DOX/TPP-DOX@Pasp-hyd-PEG-FA micelles were prepared by using newly synthesized comb-like amphiphilic material Pasp-hyd-PEG-FA. Drug released in vitro from micelles showed a pH-dependent manner. DOX/TPP-DOX@Pasp-hyd-PEG-FA induced more apoptosis in KB cell and MCF-7/ADR cell than DOX@Pasp-hyd-PEG-FA. Confocal laser scanning microscopy experiment indicated that DOX/TPP-DOX@Pasp-hyd-PEG-FA delivered TPP-DOX and DOX to the nucleus and mitochondria of the tumor cell simultaneously. Thus, DOX/TPP-DOX@Pasp-hyd-PEG-FA could significantly damage the mitochondrial membrane potential. DOX/TPP-DOX@Pasp-hyd-PEG-FA markedly shrinked the tumor volume in tumor-bearing nude mice grafted with MCF-7/ADR cell as compared with the same dose of free DOX. DOX was mainly accumulated in tumor tissue after DOX/TPP-DOX@Pasp-hyd-PEG-FA was injected to tumor-bearing nude mice by tail vein. After free DOX was injected to tumor-bearing nude mice by tail vein, DOX widely distributed through the whole body. Therefore, mitochondria and nucleus dual delivery system has potential in overcoming DOX resistance.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Femenino , Ácido Fólico/química , Humanos , Concentración de Iones de Hidrógeno , Células KB , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Desnudos , Micelas , Polietilenglicoles/química
14.
Sci Rep ; 6: 35267, 2016 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-27731405

RESUMEN

With the extensive application of doxorubicin (DOX), DOX resistance has become one of the main obstacles to the effective treatment of breast cancer. In this paper, DOX and resveratrol (RES) were co-encapsulated in a modified PLGA nanoparticle (NPS) to overcome the DOX resistance. CLSM results indicated that DOX and RES were simultaneously delivered into the nucleus of DOX-resistant human breast cancer cells by DOX/RES-loaded NPS. Consequently, DOX/RES-loaded NPS showed significant cytotoxicity on MDA-MB-231/ADR cells and MCF-7/ADR cells. Furthermore, DOX/RES-loaded NPS could overcome DOX resistance by inhibiting the expression of drug resistance-related protein such as P-gp, MRP-1 and BCRP, and induce apoptosis through down-regulating the expression of NF-κB and BCL-2. In tumor-bearing mice, DOX/RES-loaded NPS mainly delivered DOX and RES to tumor tissue. Compared with free DOX, DOX/RES-loaded NPS significantly inhibited the DOX-resistant tumor growth in tumor-bearing mice without causing significant systemic toxicity. In a word, DOX/RES-loaded NPS could overcome the DOX resistance and had the potential in the treatment of DOX-resistant breast cancer.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Portadores de Fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Nanopartículas , Estilbenos/administración & dosificación , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Resveratrol
15.
Sci Rep ; 5: 16125, 2015 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-26530454

RESUMEN

In order to overcome drug resistant and enhance antitumor activity of DOX, a new pH-sensitive micelle (DOX/DQA-DOX@DSPE-hyd-PEG-AA) was prepared to simultaneously deliver DOX to nucleus and mitochondria. Drug released from DOX/DQA-DOX@DSPE-hyd-PEG-AA showed a pH-dependent manner. DOX/DQA-DOX@DSPE-hyd-PEG-AA induced the depolarization of mitochondria and apoptosis in MDA-MB-231/ADR cells and A549 cells, which resulted in the high cytotoxicity of DOX/DQA-DOX@DSPE-hyd-PEG-AA against MDA-MB-231/ADR cells and A549 cells. Confocal microscopy confirmed that DOX/DQA-DOX@DSPE-hyd-PEG-AA simultaneously delivered DQA-DOX and DOX to the mitochondria and nucleus of tumor cell. After DOX/DQA-DOX@DSPE-hyd-PEG-AA was injected to the tumor-bearing nude mice by the tail vein, DOX was mainly found in tumor tissue. But DOX was widely distributed in the whole body after the administration of free DOX. Compared with free DOX, the same dose of DOX/DQA-DOX@DSPE-hyd-PEG-AA significantly inhibited the growth of DOX-resistant tumor in tumor-bearing mice without obvious systemic toxicity. Therefore, dual subcellular compartment delivery of DOX greatly enhanced the antitumor activity of DOX on DOX-resistant tumor. DOX/DQA-DOX@DSPE-hyd-PEG-AA has the potential in target therapy for DOX-resistant tumor.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Doxorrubicina/farmacología , Portadores de Fármacos/química , Animales , Antibióticos Antineoplásicos/metabolismo , Antibióticos Antineoplásicos/uso terapéutico , Antibióticos Antineoplásicos/toxicidad , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/metabolismo , Doxorrubicina/uso terapéutico , Femenino , Humanos , Concentración de Iones de Hidrógeno , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Micelas , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Distribución Tisular , Trasplante Heterólogo
16.
Mol Pharm ; 11(5): 1378-90, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24720787

RESUMEN

Doxorubicin (DOX) is a broad-spectrum antitumor drug used in the clinic. However, it can cause serious heart toxicity. To increase the therapeutic index of DOX and to attenuate its toxicity toward normal tissues, we conjugated DOX with either α-linolenic acid (LNA) or palmitic acid (PA) by a hydrazone or an amide bond to produce DOX-hyd-LNA, DOX-ami-LNA, DOX-hyd-PA, and DOX-ami-PA. The cytotoxicity of DOX-hyd-LNA on HepG2, MCF-7, and MDA-231 cells was higher compared to that of DOX, DOX-ami-LNA, DOX-hyd-PA, and DOX-ami-PA. The cytotoxicity of DOX-hyd-LNA on HUVECs was lower than that of DOX. DOX-hyd-LNA released significantly more DOX in pH 5.0 medium than it did in pH 7.4 medium. DOX-hyd-LNA induced more apoptosis in MCF-7 and HepG2 cells than DOX or DOX-ami-LNA. Significantly more DOX was released from DOX-hyd-LNA in both MCF-7 and HepG2 cells compared with DOX-ami-LNA. Compared to free DOX, a biodistribution study showed that DOX-hyd-LNA greatly increased the content of DOX in tumor tissue and decreased the content of DOX in heart tissue after it was intravenously administered. DOX-hyd-LNA improved the survival rate, prolonged the life span, and slowed the growth of the tumor in tumor-bearing nude mice. These results indicate that DOX-hyd-LNA improved the therapeutic index of DOX. Therefore, DOX-hyd-LNA is a potential compound for use as a cancer-targeting therapy.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/uso terapéutico , Doxorrubicina/química , Doxorrubicina/uso terapéutico , Ácido alfa-Linolénico/química , Animales , Antineoplásicos/administración & dosificación , Línea Celular , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Femenino , Células Hep G2 , Humanos , Hidrazonas/química , Concentración de Iones de Hidrógeno , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Ratones Desnudos , Ácido Palmítico/química , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Anticancer Drugs ; 25(7): 751-66, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590167

RESUMEN

The experiment aimed to increase the drug-delivery efficiency of poly-lactic-co-glycolic acid (PLGA) nanoparticles. Lipid-polymer hybrid nanoparticles (LPNs-1) were prepared using PLGA as a hydrophobic core and FA-PEG-hyd-DSPE as an amphiphilic shell. Uniform and spherical nanoparticles with an average size of 185 nm were obtained using the emulsification solvent evaporation method. The results indicated that LPNs-1 showed higher drug loading compared with naked PLGA nanoparticles (NNPs). Drug release from LPNs-1 was faster in an acidic environment than in a neutral environment. LPNs-1 showed higher cytotoxicity on KB cells, A549 cells, MDA-MB-231 cells, and MDA-MB-231/ADR cells compared with free doxorubicin (DOX) and NNPs. The results also showed that, compared with free DOX and NNPs, LPNs-1 delivered more DOX to the nuclear of KB cells and MDA-MB-231/ADR cells. LPNs-1 induced apoptosis in KB cells and MDA-MB-231/ADR cells in a dose-dependent manner. The above data indicated that DOX-loaded LPNs-1 could kill not only normal tumor cells but also drug-resistant tumor cells. These results indicated that modification of PLGA nanoparticles with FA-PEG-hyd-DSPE could considerably increase the drug-delivery efficiency and LPNs-1 had potential in the delivery of chemotherapeutic agents in the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Ácido Fólico/análogos & derivados , Nanopartículas/química , Fosfatidilcolinas/química , Polietilenglicoles/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Doxorrubicina/administración & dosificación , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Ácido Láctico/química , Ácido Láctico/metabolismo , Nanopartículas/metabolismo , Fosfatidilcolinas/metabolismo , Polietilenglicoles/metabolismo , Ácido Poliglicólico/química , Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico
18.
Mini Rev Med Chem ; 13(10): 1462-74, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23815579

RESUMEN

Melatonin, an endogenous ligand for melatonin receptor, plays an important role in modulating various physiological activities through acting on different subtypes MT1, MT2 or the binding site MT3. The distinct roles of the receptor subtypes provide great potential for receptor-specific pharmacological agents. Melatonin has no subtypeselectivity, so it is very important to develop different subtype-selective ligand for receptor subtype research and drug development. In order to provide guidance for developing high selective ligand, this paper focused on the MT2-selective ligands, which developed well in the past years. The MT2-selective ligands, mainly focusing on binding data on MT1 and MT2 receptor, are reviewed in detail according to their structural classes, and the relative pharmacophore, receptor binding models and the relationship between the structure of ligand and the affinity along with selectivity for receptor subtype were discussed, which may facilitate the exploration of more potent and effective MT2-selective ligands.


Asunto(s)
Melatonina/metabolismo , Melatonina/farmacología , Receptor de Melatonina MT2/antagonistas & inhibidores , Receptor de Melatonina MT2/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Melatonina/química , Estructura Molecular , Relación Estructura-Actividad
19.
J Pharm Sci ; 102(2): 530-40, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169439

RESUMEN

Folate-aminocaproic acid-doxorubicin (FA-AMA-DOX) was synthesized and characterized by H NMR spectroscopy and mass spectrometry. Cytotoxicity and cellular uptake experiments were performed in KB and HepG2 cells, which express folic acid receptor, and the cell line A549, which does not express folic acid receptor. Cytotoxicity was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and cellular uptake was monitored using fluorescence microscopy. The amount of DOX released from FA-AMA-DOX was much greater at pH 5.0 than that at pH 6.5 or 7.4. The cytotoxicity of FA-AMA-DOX toward KB and HepG2 cells was greater than that of DOX or AMA-DOX at the same concentrations, and cytotoxicity could be attenuated by FA in a dose-dependent manner. On the contrary, the cytotoxicity of FA-AMA-DOX and AMA-DOX toward A549 cells was lower than that of DOX at the same concentration, and cytotoxicity could not be reduced by FA. Compared with FA-AMA, FA-AMA-DOX increased the intracellular accumulation of DOX in KB cells. These results suggested that FA-AMA-DOX have suitable attributes for the active targeting of folate-receptor-positive tumor cells and for releasing the chemotherapeutic agent, DOX, in situ; it therefore has potential as a novel cancer therapeutic.


Asunto(s)
Antineoplásicos/síntesis química , Doxorrubicina/síntesis química , Portadores de Fármacos/síntesis química , Ácido Fólico/síntesis química , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Portadores de Fármacos/metabolismo , Portadores de Fármacos/farmacología , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Células Hep G2 , Humanos , Concentración de Iones de Hidrógeno
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...