Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Drug Deliv ; 28(1): 1923-1931, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34550040

RESUMO

Transdermal drug delivery for local or systemic therapy provides a potential anticancer modality with a high patient compliance. However, the drug delivery efficiency across the skin is highly challenging due to the physiological barriers, which limit the desired therapeutic effects. In this study, we prepared liposome-in-hydrogels containing a tumor targeting photosensitizer IR780 (IR780/lipo/gels) for tumor photothermal therapy (PTT). The formulation effectively delivered IR780 to subcutaneous tumor and deep metastatic sites, while the hydrogels were applied on the skin overlying the tumor or on an area of distant normal skin. The photothermal antitumor activity of topically administered IR780/lipo/gels was evaluated following laser irradiation. We observed significant inhibition of the rate of the tumor growth without any toxicity associated with the topical administration of hydrogels. Collectively, the topical administration of IR780/lipo/gels represents a new noninvasive and safe strategy for targeted tumor PTT.


Assuntos
Hidrogéis/química , Indóis/farmacologia , Lipossomos/química , Fármacos Fotossensibilizantes/farmacologia , Terapia Fototérmica/métodos , Administração Cutânea , Animais , Peso Corporal , Química Farmacêutica , Portadores de Fármacos/química , Indóis/administração & dosagem , Indóis/efeitos adversos , Indóis/farmacocinética , Terapia com Luz de Baixa Intensidade/efeitos adversos , Terapia com Luz de Baixa Intensidade/métodos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/efeitos adversos , Fármacos Fotossensibilizantes/farmacocinética , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Pharm ; 589: 119763, 2020 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-32898629

RESUMO

Phototherapy exerts its anticancer effects by converting laser radiation energy into hyperthermia or reactive singlet oxygen (1O2). In this study, we developed chitosan nanoparticles (CS NPs) encapsulating both photothermal (IR780) and photodynamic (5-Aminolevulinic acid (5-ALA)) reagents for photothermally enhanced photodynamic therapy by noninvasive oral administration. The 5-ALA&IR780@CS NPs were stable in acidic conditions similar to the gastric environment, which greatly improved drug oral absorption and local accumulation in subcutaneous mouse colon tumors (CT-26 cells) following oral gavage. Mechanistic studies revealed that the co-delivery system can lead to photothermally enhanced photodynamic effects against cancer cells by increasing oxidative stress, including the elevation of ROS, superoxide and 1O2 production. Additionally, significant therapeutic efficacy for cancer treatment were observed in vivo after oral administration of 5-ALA&IR780@CS NPs, without causing any overt adverse effects. Our work highlights the great potential of photothermally enhanced photodynamic therapy by CS NPs for colon cancer management via oral route.


Assuntos
Quitosana , Neoplasias do Colo , Hipertermia Induzida , Nanopartículas , Fotoquimioterapia , Animais , Neoplasias do Colo/tratamento farmacológico , Indóis , Camundongos
3.
J Nanobiotechnology ; 18(1): 124, 2020 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-32887622

RESUMO

BACKGROUND: Chemotherapy is a standard cancer treatment which uses anti-cancer drugs to destroy or slow the growth of cancer cells. However, chemotherapy has limited therapeutic effects in bladder cancer. One of the reasons of this resistance to chemotherapy is that higher levels of glutathione in invasive bladder cancer cells. We have fabricated nanoparticles that respond to high concentrations of glutathione and near-infrared laser irradiation in order to increase the drug accumulation at the tumor sites and combine chemotherapy with photothermal therapy to overcome the challenges of bladder cancer treatment. METHODS: The DOX&IR780@PEG-PCL-SS NPs were prepared by co-precipitation method. We investigated the tumor targeting capability of NPs in vitro and in vivo. The orthotopic bladder cancer model in C57BL/6 mice was established for in vivo study and the photothermal effects and therapeutic efficacy of NPs were evaluated. RESULTS: The DOX&IR780@PEG-PCL-SS NPs were synthesized using internal cross-linking strategy to increase the stability of nanoparticles. Nanoparticles can be ingested by tumor cells in a short time. The DOX&IR780@PEG-PCL-SS NPs have dual sensitivity to high levels of glutathione in bladder cancer cells and near-infrared laser irradiation. Glutathione triggers chemical structural changes of nanoparticles and preliminarily releases drugs, Near-infrared laser irradiation can promote the complete release of the drugs from the nanoparticles and induce a photothermal effect, leading to destroying the tumor cells. Given the excellent tumor-targeting ability and negligible toxicity to normal tissue, DOX&IR780@PEG-PCL-SS NPs can greatly increase the concentration of the anti-cancer drugs in tumor cells. The mice treated with DOX&IR780@PEG-PCL-SS NPs have a significant reduction in tumor volume. The DOX&IR780@PEG-PCL-SS NPs can be tracked by in vivo imaging system and have good tumor targeting ability, to facilitate our assessment during the experiment. CONCLUSION: A nanoparticle delivery system with dual sensitivity to glutathione and near-infrared laser irradiation was developed for delivering IR780 and DOX. Chemo-photothermal synergistic therapy of both primary bladder cancer and their metastases was achieved using this advanced delivery system.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Musculares/tratamento farmacológico , Nanopartículas/química , Nanopartículas/uso terapêutico , Polímeros/química , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Terapia Combinada , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Tratamento Farmacológico/métodos , Humanos , Raios Infravermelhos , Terapia a Laser , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Musculares/patologia , Neoplasias Musculares/radioterapia , Músculos/efeitos dos fármacos , Fototerapia/métodos , Polietilenoglicóis , Sensibilidade e Especificidade , Succinimidas , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/radioterapia
4.
Front Chem ; 8: 601649, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33520933

RESUMO

Malignant cancer is a devastating disease often associated with a poor clinical prognosis. For decades, modern drug discoveries have attempted to identify potential modulators that can impede tumor growth. Cancer stem cells however are more resistant to therapeutic intervention, which often leads to treatment failure and subsequent disease recurrence. Here in this study, we have developed a specific multi-target drug delivery nanoparticle system against breast cancer stem cells (BCSCs). Therapeutic agents curcumin and salinomycin have complementary functions of limiting therapeutic resistance and eliciting cellular death, respectively. By conjugation of CD44 cell-surface glycoprotein with poly(lactic-co-glycolic acid) (PLGA) nanoparticles that are loaded with curcumin and salinomycin, we investigated the cellular uptake of BCSCs, drug release, and therapeutic efficacy against BCSCs. We determined CD44-targeting co-delivery nanoparticles are highly efficacious against BCSCs by inducing G1 cell cycle arrest and limiting epithelial-mesenchymal transition. This curcumin and salinomycin co-delivery system can be an efficient treatment approach to target malignant cancer without the repercussion of disease recurrence.

5.
Cell Physiol Biochem ; 51(4): 1566-1583, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30497066

RESUMO

BACKGROUND/AIMS: Glioblastoma multiforme (GBM) is the most devastating and widespread primary central nervous system tumour in adults, with poor survival rate and high mortality rates. Existing treatments do not provide substantial benefits to patients; therefore, novel treatment strategies are required. Peiminine, a natural bioactive compound extracted from the traditional Chinese medicine Fritillaria thunbergii, has many pharmacological effects, especially anticancer activities. However, its anticancer effects on GBM and the underlying mechanism have not been demonstrated. This study was conducted to investigate the potential antitumour effects of peiminine in human GBM cells and to explore the related molecular signalling mechanisms in vitro and in vivo Methods: Cell viability and proliferation were detected with MTT and colony formation assays. Morphological changes associated with autophagy were assessed by transmission electron microscopy (TEM). The cell cycle rate was measured by flow cytometry. To detect changes in related genes and signalling pathways in vitro and in vivo, RNA-seq, Western blotting and immunohistochemical analyses were employed. RESULTS: Peiminine significantly inhibited the proliferation and colony formation of GBM cells and resulted in changes in many tumour-related genes and transcriptional products. The potential anti-GBM role of peiminine might involve cell cycle arrest and autophagic flux blocking via changes in expression of the cyclin D1/CDK network, p62 and LC3. Changes in Changes in flow cytometry results and TEM findings were also observed. Molecular alterations included downregulation of the expression of not only phospho-Akt and phospho-GSK3ß but also phospho-AMPK and phospho-ULK1. Furthermore, overexpression of AKT and inhibition of AKT reversed and augmented peiminine-induced cell cycle arrest in GBM cells, respectively. The cellular activation of AMPK reversed the changes in the levels of protein markers of autophagic flux. These results demonstrated that peiminine mediates cell cycle arrest by suppressing AktGSk3ß signalling and blocks autophagic flux by depressing AMPK-ULK1 signalling in GBM cells. Finally, peiminine inhibited the growth of U251 gliomas in vivo. CONCLUSION: Peiminine inhibits glioblastoma in vitro and in vivo via arresting the cell cycle and blocking autophagic flux, suggesting new avenues for GBM therapy.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Cevanas/uso terapêutico , Glioblastoma/tratamento farmacológico , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Antineoplásicos Fitogênicos/farmacologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cevanas/farmacologia , Feminino , Fritillaria/química , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos
6.
Nanomedicine ; 14(7): 2283-2294, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29981460

RESUMO

Tumor angiogenesis is a key step in the process of tumor development, and antitumor angiogenesis has a profound influence on tumor growth. Herein we report a dual-function drug delivery system comprising a Near-infrared (NIR) dye and an anti-angiogenic drug within liposomes (Lip-IR780-Sunitinib) for enhanced antitumor therapy. The hydrophobic NIR dye IR780 was loaded into the liposome phospholipid bilayer, and the bilayer would be disrupted by laser irradiation so that anti-angiogenic drug sunitinib release would be activated remotely at the tumor site. The released hydrophilic sunitinib could potentially target multiple VEGF receptors on the tumor endothelial cell surface to inhibit angiogenesis. Meanwhile, IR780-loaded liposomes kill the cancer cells by photothermal therapy. Lip-IR780-Sunitinib exhibited enhanced anti-tumor and anti-angiogenic effects in vitro and in vivo. This system facilitates easy and controlled release of cargos to achieve anti-tumor angiogenesis and photothermal therapy.


Assuntos
Neoplasias da Mama/terapia , Hipertermia Induzida , Indóis/química , Lipossomos/administração & dosagem , Neovascularização Patológica/terapia , Fototerapia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Animais , Apoptose , Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Terapia Combinada , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Indóis/administração & dosagem , Lasers , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/patologia , Espectroscopia de Luz Próxima ao Infravermelho , Sunitinibe/química , Sunitinibe/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Adv Healthc Mater ; 7(4)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29227047

RESUMO

Polymeric materials that respond to a variety of endogenous and external stimuli are actively developed to overcome the main barriers to successful systemic delivery of therapeutic nucleic acids. Here, an overview of viable stimuli that are proved to improve systemic delivery of nucleic acids is provided. The main focus is placed on nucleic acid delivery systems (NADS) based on polymers that respond to pathological or physiological changes in pH, redox state, enzyme levels, hypoxia, and reactive oxygen species levels. Additional discussion is focused on NADS suitable for applications that use external stimuli, such as light, ultrasound, and local hyperthermia.


Assuntos
Técnicas de Transferência de Genes , Ácidos Nucleicos/química , Polímeros/química , Fosfatos de Cálcio/química , Humanos , Concentração de Íons de Hidrogênio , Hipertermia Induzida , Ácidos Nucleicos/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Nanomedicine (Lond) ; 12(9): 1043-1055, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28440689

RESUMO

AIM: The aim of the present study was to use hemoglobin (Hb) nanoparticles (NPs) to improve oral bioavailability of a near-infrared dye IR780 for in vivo antitumor application in photothermal therapy. METHODS: One-step acid-denaturing method was used to encapsulate IR780 into self-assembled Hb NPs (IR780@Hb NPs). Pharmacokinetics, biodistribution and antitumor effect were studied in vivo. RESULTS: The Hb NPs showed high stability in enzymatic and acidic conditions similar to the gastric environment, and enhanced absorption of IR780 into the blood. In vivo imaging revealed that IR780 could accumulate at the tumor sites and effectively caused photothermal effect, which resulted in tumor ablation after oral administration in tumor-bearing mice. CONCLUSION: Hb NPs represent a promising delivery system for improving oral absorption of photosensitizer dyes, which could open new treatment modalities in cancer.


Assuntos
Corantes/administração & dosagem , Portadores de Fármacos/química , Hemoglobinas/química , Indóis/administração & dosagem , Nanopartículas/química , Neoplasias/terapia , Fármacos Fotossensibilizantes/administração & dosagem , Administração Oral , Animais , Células CACO-2 , Linhagem Celular Tumoral , Corantes/uso terapêutico , Humanos , Hipertermia Induzida/métodos , Indóis/uso terapêutico , Masculino , Camundongos Endogâmicos BALB C , Modelos Moleculares , Neoplasias/patologia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia/métodos , Distribuição Tecidual
9.
Sci Rep ; 6: 27421, 2016 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-27263444

RESUMO

Combination of photothermal and photodynamic therapy (PTT/PDT) offer unique advantages over PDT alone. However, to achieve synergetic PDT/PTT effect, one generally needs two lasers with different wavelengths. Near-infrared dye IR-780 could be used as photosensitizer both for PTT and PDT, but its lipophilicity limits its practical use and in vivo efficiency. Herein, a simple multifunctional IR780-loaded nanoplatform based on transferrin was developed for targeted imaging and phototherapy of cancer compatible with a single-NIR-laser irradiation. The self-assembled transferrin-IR780 nanoparticles (Tf-IR780 NPs) exhibited narrow size distribution, good photo-stability, and encouraging photothermal performance with enhanced generation of ROS under laser irradiation. Following intravenous injection, Tf-IR780 NPs had a high tumor-to-background ratio in CT26 tumor-bearing mice. Treatment with Tf-IR780 NPs resulted in significant tumor suppression. Overall, the Tf-IR780 NPs show notable targeting and theranostic potential in cancer therapy.


Assuntos
Hipertermia Induzida/métodos , Nanopartículas , Neoplasias/terapia , Fotoquimioterapia , Fototerapia/métodos , Transferrina/química , Animais , Indóis/farmacologia , Indóis/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/tratamento farmacológico , Espectrofotometria Ultravioleta
10.
Mol Pharm ; 13(3): 829-38, 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26713719

RESUMO

PEG-IR780-C13 micelles have been demonstrated to be a novel photothermal agent with tumor-targeting property. This study was designed to explore the feasibility of applying PEG-IR780-C13 micelles and near-infrared (NIR) irradiation for thermal ablation of renal tumor by using an in situ tumor model. In addition, the potential thermal injury to normal renal tissue was evaluated. PEG-IR780-C13 micelles were intended to accumulate in renal tumor after systemic delivery. In vitro results revealed that PEG-IR780-C13 micelles were uptaken by RENCA cells mainly through caveola-mediated endocytosis and mainly distributed in late endosomes and lysosomes. Upon NIR irradiation, PEG-IR780-C13 micelles generated heat effectively both in vitro and in vivo, exhibiting a promising photothermal therapeutic property. The photothermal effect of PEG-IR780-C13 micelles could effectively destroy RENCA cells in vitro and adequately inhibit growth of in situ renal tumor in vivo. Meanwhile, PEG-IR780-C13 micelles mediated photothermal therapy (PTT) resulting in only limited injury to normal renal tissue surrounding tumor sites. Our data indicated that PEG-IR780-C13 micelles mediating PTT could generate tumor-specific heat for destruction of renal tumor in a minimally invasive way, providing a novel strategy for thermal ablation of renal tumor.


Assuntos
Hipertermia Induzida , Indóis/farmacologia , Raios Infravermelhos , Neoplasias Renais/terapia , Fototerapia , Polietilenoglicóis/química , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Terapia Combinada , Citometria de Fluxo , Técnicas Imunoenzimáticas , Neoplasias Renais/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Micelas , Microscopia Confocal , Espectroscopia de Luz Próxima ao Infravermelho , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA