Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Asian J Pharm Sci ; 18(1): 100769, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36698441

RESUMO

The siRNA-loaded lipid nanoparticles have attracted much attention due to its significant gene silencing effect and successful marketization. However, the in vivo distribution and release of siRNA still cannot be effectively monitored. In this study, based on the fluorescence resonance energy transfer (FRET) principle, a fluorescence dye Cy5-modified survivin siRNA was conjugated to nanogolds (Au-DR-siRNA), which were then wrapped with lipid nanoparticles (LNPs) for monitoring the release behaviour of siRNA in vivo. The results showed that once Au-DR-siRNA was released from the LNPs and cleaved by the Dicer enzyme to produce free siRNA in cells, the fluorescence of Cy5 would change from quenched state to activated state, showing the location and time of siRNA release. Besides, the LNPs showed a significant antitumor effect by silencing the survivin gene and a CT imaging function superior to iohexol by nanogolds. Therefore, this work provided not only an effective method for monitoring the pharmacokinetic behaviour of LNP-based siRNA, but also a siRNA delivery system for treating and diagnosing tumors.

2.
J Control Release ; 350: 298-307, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36002054

RESUMO

Chimeric receptor T cells (CAR-T) can effectively cure leukemia; however, there are two limitations: a complicated preparation process ex vivo and cytokine release syndrome (CRS). In this study, we constructed a lipid nanoparticle system modified by CD3 antibody on the surface, loading with the plasmid containing the combination gene of interleukin 6 short hairpin RNA (IL-6 shRNA) and CD19-CAR (AntiCD3-LNP/CAR19 + shIL6). The system targeted T cells by the mediation of CD3 antibody and stably transfected T cells to transform them into CAR-T cells with IL-6 knockdown, thus killing CD19-highly expressed leukemia tumor cells and reducing CRS caused by IL-6. In vivo experiments showed that AntiCD3-LNP/CAR19 + shIL6 could stably transfect T cells and produce CAR-T within 90 days to kill the tumor. This significantly prolonged the survival time of leukemia model mice and demonstrated the prepared LNP exhibited the same anti-tumor effect as the traditional CAR-T cells prepared ex vivo. In this study, CAR-T cells were directly produced in vivo after intravenous injection of the lipid nanoparticles, without the need of using the current complex process ex vivo. Additionally, IL-6 expression was silenced, which would be helpful to reduce the CRS and improve the safety of CAR-T therapy. This method improves the convenience of using CAR-T technology and is helpful in further promoting the clinical application of CAR-T.


Assuntos
Leucemia , Receptores de Antígenos Quiméricos , Animais , Antígenos CD19 , Imunoterapia Adotiva/métodos , Interleucina-6/genética , Lipossomos , Camundongos , Nanopartículas , RNA Interferente Pequeno/genética , Receptores de Antígenos Quiméricos/genética , Linfócitos T
3.
J Cancer Res Clin Oncol ; 148(9): 2261-2274, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35445870

RESUMO

PURPOSE: Chimeric antigen receptor (CAR) T cell therapy has demonstrated clinical success in treating haematologic malignancies but has not been effective against solid tumours thus far. Trop2 is a tumour-related antigen broadly overexpressed on a variety of tumours and has been reported as a promising target for pancreatic cancers. Our study aimed to determine whether CAR T cells designed with a fully human Trop2-specific single-chain fragment variable (scFv) can be used in the treatment of Trop2-positive pancreatic tumours. METHODS: We designed Trop2-targeted chimeric antigen receptor engineered T cells with a novel human anti-Trop2 scFv (2F11) and then investigated the cytotoxicity, degranulation, and cytokine secretion profiles of the anti-Trop2 CAR T cells when they were exposed to Trop2 + cancer cells in vitro. We also studied the antitumour efficacy and toxicity of Trop2-specific CAR T cells in vivo using a BxPC-3 pancreatic xenograft model. RESULTS: Trop2-targeted CAR T cells designed with 2F11 effectively killed Trop2-positive pancreatic cancer cells and produced high levels of cytotoxic cytokines in vitro. In addition, Trop2-targeted CAR T cells, which persistently circulate in vivo and efficiently infiltrate into tumour tissues, significantly blocked and even eliminated BxPC-3 pancreatic xenograft tumour growth without obvious deleterious effects observed after intravenous injection into NSG mice. Moreover, disease-free survival was efficiently prolonged. CONCLUSION: These results show that Trop2-targeted CAR T cells equipped with a fully human anti-Trop2 scFv could be a potential treatment strategy for pancreatic cancer and could be useful for clinical evaluation.


Assuntos
Neoplasias Pancreáticas , Receptores de Antígenos Quiméricos , Animais , Antígenos de Neoplasias , Linhagem Celular Tumoral , Humanos , Imunoterapia Adotiva/métodos , Camundongos , Neoplasias Pancreáticas/terapia , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto , Neoplasias Pancreáticas
4.
Adv Mater ; 34(15): e2109969, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35174915

RESUMO

The rapid evolution of cell-based theranostics has attracted extensive attention due to their unique advantages in biomedical applications. However, the inherent functions of cells alone cannot meet the needs of malignant tumor treatment. Thus endowing original cells with new characteristics to generate multifunctional living cells may hold a tremendous promise. Here, the nanoengineering method is used to combine customized liposomes with neutrophils, generating oxygen-carrying sonosensitizer cells with acoustic functions, which are called Acouscyte/O2 , for the visual diagnosis and treatment of cancer. Specifically, oxygen-carried perfluorocarbon and temoporfin are encapsulated into cRGD peptide modified multilayer liposomes (C-ML/HPT/O2 ), which are then loaded into live neutrophils to obtain Acouscyte/O2 . Acouscyte/O2 can not only carry a large amount of oxygen but also exhibits the ability of long circulation, inflammation-triggered recruitment, and decomposition. Importantly, Acouscyte/O2 can be selectively accumulated in tumors, effectively enhancing tumor oxygen levels, and triggering anticancer sonodynamics in response to ultrasound stimulation, leading to complete obliteration of tumors and efficient extension of the survival time of tumor-bearing mice with minimal systemic adverse effects. Meanwhile, the tumors can be monitored in real time by temoporfin-mediated fluorescence imaging and perfluorocarbon (PFC)-microbubble-enhanced ultrasound imaging. Therefore, the nanoengineered neutrophils, i.e., Acouscyte/O2 , are a new type of multifunctional cellular drug, which provides a new platform for the diagnosis and sonodynamic therapy of solid malignant tumors.


Assuntos
Fluorocarbonos , Neoplasias , Terapia por Ultrassom , Animais , Linhagem Celular Tumoral , Lipossomos/uso terapêutico , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/tratamento farmacológico , Neutrófilos , Oxigênio , Espécies Reativas de Oxigênio/uso terapêutico , Terapia por Ultrassom/métodos
5.
J Control Release ; 343: 175-186, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35092721

RESUMO

HMGB1 is an inflammatory factor produced by macrophages after liver injury, which plays a key role in promoting NASH progression and further developing into liver fibrosis and cirrhosis. In this study, a mannose-modified HMGB1-siRNA loaded stable nucleic acid lipid particle delivery system (mLNP-siHMGB1) was constructed to target liver macrophages with mannose receptor mediation, thereby silencing HMGB1 protein expression and treating NASH. We also examined the effect of co-administration with docosahexaenoic acid (DHA), a kind of unsaturated fatty acid, on NASH. The results showed that mLNP-siHMGB1 could target macrophages through mannose receptors, effectively silence HMGB1 gene, reduce the release of HMGB1 protein in the liver, regulate liver macrophages to be an anti-inflammatory M2 phenotype, effectively reduce hepatic lobular inflammation and bullous steatosis in the liver, and restore the liver function of NASH model mice to a normal level. After 8 weeks of combined treatment with mLNP-siHMGB1 and DHA, the liver function of NASH model mice recovered rapidly and the hepatic steatosis returned to normal level. In view of inflammation, a key factor in the progression of NASH, we provided an actively targeted siRNA delivery system in this study, and clarified the important role of the delivery system in phenotypic regulation of liver macrophages in NASH. In addition, we also demonstrated the effectiveness of DHA co-administration in NASH treatment. This study provided a useful idea and scientific basis for the development of therapeutic strategies for NASH in the future.


Assuntos
Proteína HMGB1 , Hepatopatia Gordurosa não Alcoólica , Animais , Modelos Animais de Doenças , Proteína HMGB1/metabolismo , Inflamação/patologia , Lipossomos , Fígado/metabolismo , Cirrose Hepática/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , Hepatopatia Gordurosa não Alcoólica/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico
6.
Pharm Res ; 38(4): 669-680, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33796952

RESUMO

PURPOSE: To address the issue of local drug delivery in tumor treatment, a novel nanoparticle-hydrogel superstructure, namely semi-interpenetrating polymer networks (semi-IPNs) hydrogel composed of poly (ethylene glycol) diacrylate (PEGDA) and hyaluronic acid (HA) and incorporated with paclitaxel (PTX) loaded PLGA nanoparticles (PEGDA-HA/PLGA-PTX), was prepared by in situ UV photopolymerization for the use of local drug delivery. METHODS: Using the gelation time, swelling rate and degradation rate as indicators, the optimal proportion of Irgacure 2959 initiator and the concentration of HA was screened and obtained for preparing hydrogels. Next, paclitaxel (PTX) loaded PLGA nanoparticles (PLGA-PTX NPs) were prepared by the emulsion solvent evaporation method. RESULTS: The mass ratio of the initiator was 1%, and the best concentration of HA was 5 mg/mL in PEGDA-HA hydrogel. In vitro experiments showed that PLGA-PTX NPs had similar cytotoxicity to free PTX, and the cell uptake ratio on NCI-H460 cells was up to 96% by laser confocal microscopy and flow cytometry. The drug release of the PEGDA-HA/PLGA-PTX hydrogel local drug delivery system could last for 13 days. In vivo experiments proved that PEGDAHA/PLGA-PTX hydrogel could effectively inhibit the tumor growth without causing toxic effects in mice. CONCLUSIONS: This study demonstrated that the PEGDA-HA/PLGA-PTX hydrogel is a promising local drug delivery system in future clinical applications for tumor therapy. A photopolymerized semi-interpenetrating polymer networks-based hydrogel incorporated with paclitaxel-loaded nanoparticles was fabricated by in situ UV photopolymerization, providing a promised nanoplatform for local chemotherapy of tumors.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Portadores de Fármacos/química , Hidrogéis/química , Neoplasias/tratamento farmacológico , Paclitaxel/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacocinética , Linhagem Celular Tumoral , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/farmacocinética , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Humanos , Ácido Hialurônico/química , Camundongos , Nanopartículas/química , Neoplasias/patologia , Polietilenoglicóis/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biomed Pharmacother ; 137: 111339, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33550044

RESUMO

Chimeric antigen receptor T cells (CAR-T) immunotherapy has shown promising clinical results in the treatment of leukemia and lymphoma, but the effectiveness is limited for solid tumors. The PD-1/PD-L1 pathway is a key immunosuppressive mechanism for cancer cells to avoid eradication by CAR-T cells. In this study, the shRNA (short hair RNA) gene-silencing technique was used to construct the third-generation of CAR-T cells with PD-1 silencing which targeted CD19 antigen (CD19/△PD-1 CAR-T) and prostate stem cell antigen (PSCA/△PD-1 CAR-T), thereby blocking the PD-1/PD-L1 pathway in treatment of lymphoma and prostate subcutaneous xenograft and enhancing the anti-tumor effect of CAR-T cells. The cell experiments showed that PD-1 silencing in CAR-T cells effectively blocked the PD-1 / PD-L1 pathway. When the ratio of effector to target cell is 8:1, △PD-1 CAR-T cells exhibited higher killing ability and cytokine releasing ability than normal CAR-T cells did. The subcutaneous tumor models were constructed using human chronic myelogenous leukemia cells expressing CD19 (K562-CD19) and human prostate cancer cells expressing PSCA (PC3-PSCA), and treated with CD19/△PD-1 CAR-T and PSCA/△PD-1 CAR-T cells, respectively. The tumor volumes significantly reduced within one week, indicating the good tumor growth inhibitory effect of △PD-1 CAR-T cells. Mice injected with △PD-1 CAR-T cells showed a significantly prolonged survival time compared to those with normal CAR-T cells. This study proved that shRNA-mediated PD-1 silencing technology is an effective strategy for blocking the PD-1/PD-L1 immunosuppression pathway and enhancing the therapeutic effect of CAR-T cells on subcutaneous xenograft. SUMMARY: The effect of CAR-T in treating solid tumors has not been as successful as that in hematological malignancies. The key immunosuppressive mechanism is the expression of PD-1/PD-L1. We used gene silencing technique mediated by shRNA (short hair RNA) to block the PD-1/PD-L1 pathway in lymphoma and prostate tumors, thus enhancing the anti-tumor effect of CAR-T cells on subcutaneous xenograft.


Assuntos
Xenoenxertos/efeitos dos fármacos , Imunoterapia Adotiva/métodos , Leucemia/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Neoplasias da Próstata/tratamento farmacológico , RNA Interferente Pequeno/farmacologia , Animais , Antígenos CD19/metabolismo , Linhagem Celular Tumoral , Inativação Gênica/imunologia , Humanos , Leucemia/imunologia , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Receptor de Morte Celular Programada 1/genética , Neoplasias da Próstata/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nanomedicine (Lond) ; 16(6): 465-480, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33599532

RESUMO

Aim: To construct a long circulatory and sustained releasing H2S system and explore its protective effects on myocardial ischemia and reperfusion (I/R) injury. Materials & methods: Red blood cell (RBC) membrane-coated, diallyl trisulfide (DATS)-carrying mesoporous iron oxide nanoparticles (MIONs) (RBC-DATS-MIONs) were prepared and characterized. Cytotoxicity and cellular uptake were studied in vitro, followed by in vivo assessment of safety, distribution and effect on cardiac function following I/R injury. Results: RBC-DATS-MIONs exhibited excellent biocompatibility, extended circulatory time and controlled-release of H2S in plasma and myocardium. They exhibited superior therapeutic effects on in vitro hypoxia/reoxygenation models and in vivo myocardial I/R models, which involved various mechanisms, including anti-apoptosis, anti-inflammatory and antioxidant activities. Conclusion: This work provides a new potential platform for best utilizing the protective effects of H2S by prolonging its releasing process.


Assuntos
Traumatismo por Reperfusão Miocárdica , Nanopartículas , Membrana Eritrocítica , Humanos , Sulfeto de Hidrogênio , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio
9.
Mol Pharm ; 14(5): 1811-1820, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28388081

RESUMO

The tumor vascular barrier and tumor stroma barrier become the two main obstacles in the in vivo delivery of nanomedicines. In this study, to overcome the two barriers, we used iNGR, a tumor-penetrating peptide, to modify the liposomes to increase their accumulation and penetration in tumor tissues. First, iNGR-modified sterically stabilized liposomes (iNGR-SSL) were prepared, which showed vesicle sizes of about 100 nm and narrow size distribution. The uptake of iNGR-SSL by U87MG cells and HUVECs were significantly more than that of unmodified liposome. The in vivo imaging study demonstrated that iNGR modification remarkably increased the accumulation of the liposome in orthotopic tumor tissues of animal model. The immunofluorescence staining analysis proved that iNGR-SSL could penetrate through tumor blood vessels and into the deep tumor tissues. The cytotoxicity of iNGR-modified doxorubicin-loaded liposomes (iNGR-SSL/DOX) on U87MG and HUVECs cells in vitro was significantly enhanced than that of unmodified doxorubicin-loaded liposomes (SSL/DOX). The iNGR-SSL/DOX also showed comparatively (p < 0.05) stronger cytotoxicity on tumor than SSL/DOX, which should be resulted from the increased tumor accumulation and penetration mediated by iNGR. This study proved that iNGR peptide modification might be an effective method to enhance the tumor penetrating ability of liposomes in tumor tissue and their antitumor effect.


Assuntos
Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/uso terapêutico , Doxorrubicina/química , Doxorrubicina/uso terapêutico , Glioblastoma/tratamento farmacológico , Lipossomos/química , Oligopeptídeos/química , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Células Endoteliais da Veia Umbilical Humana , Humanos , Microscopia Eletrônica de Transmissão
10.
Curr Pharm Des ; 23(20): 3034-3046, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27928957

RESUMO

BACKGROUND: In this study, we developed a drug of IFN-α combined with pPB-SSLs, which specifically target at platelet-derived growth factor receptor-ß (PDGFR-ß). AIM: The aim of this study is to improve the limitations of IFN-α including insufficient drug concentration for the target cells and side-effects causing serious concerns in treatment of hepatic fibrosis. METHODS: We constructed the targeted stable liposomes (SSLs) that not only increase the half-life period of IFN- α, but also can deliver IFN-α to hepatic stellate cells (HSCs). Subsequently, the anti-hepatic-fibrosis effect of pPB-SSL-IFN-α was evaluated both in vitro and in vivo. Immunofluorescent assay showed that the pPB-SSL particles were able to be easily taken up by 3T3 cells. The cellular distribution experiment demonstrated that most of the pPB-SSL-IFN-α would accumulate around the fibroblast, and the cell would be invaded by pPB-SSLIFN- α. RESULTS: The pPB-SSL-IFN-α showed an entrapment efficiency of 39.73 ± 5.21% for IFN-α and the particles reached nanoscale level. It showed more significant alleviated performance for hepatic fibrosis than IFN-α. Both in vitro and in vivo, the pPB-SSL-IFN-α could contribute to reduction or inhibition in the expression of TGF-ß1 and α-SMA even cleavage of caspase-3. Moreover, it was found that the pPB-SSL-IFN-α induced the apoptosis of 3T3 cells by inhibiting the expression of TGF-ß1 as well as α-SMA. Under observation for fibrotic liver of mice treated with pPB-SSL-IFN-α, the semiquantitative score for collagen I, TGF-ß1 and α-SMA were all inferior to the control group and those treated with PEG-IFN-α, SSL-IFN-α or IFN-α. In addition, pPB-SSL-IFN-α has been detected to down-regulate the expression of TNF-α and IL-1ß in comparison with model group (P<0.01). And the phosphorylations of JAK1 and STAT1 were enhanced by pPB-SSL-IFN-αin comparison with model groups (P < 0.01). CONCLUSION: All results of our present research indicated that the pPB-SSL-IFN-α might be an alternative antiliver fibrotic drug and the synthetic method may offer a new access to the anti-hepatic fibrosis research and development.


Assuntos
Sistemas de Liberação de Medicamentos , Interferon-alfa/farmacocinética , Lipossomos/administração & dosagem , Cirrose Hepática Experimental/tratamento farmacológico , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Sinergismo Farmacológico , Interferon-alfa/administração & dosagem , Interferon-alfa/uso terapêutico , Cirrose Hepática Experimental/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...