Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Tipo de estudo
Intervalo de ano de publicação
1.
J Nanobiotechnology ; 21(1): 49, 2023 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-36759822

RESUMO

Aggregation-induced emission luminogens (AIEgens) have emerged as novel phototherapeutic agents with high photostability and excellent performance to induce photodynamic and/or photothermal effects. In this study, a zwitterion-type NIR AIEgens C41H37N2O3S2 (named BITT) with biomimetic modification was utilized for lung cancer therapy. The tumor-associated macrophage (TAM)-specific peptide (CRV) was engineered into the lung cancer cell-derived exosomes. The CRV-engineered exosome membranes (CRV-EM) were obtained to camouflage the BITT nanoparticles (CEB), which targeted both lung cancer cells and TAMs through homotypic targeting and TAM-specific peptide, respectively. The camouflage with CRV-EM ameliorated the surface function of BITT nanoparticles, which facilitated the cellular uptake in both cell lines and induced significant cell death in the presence of laser irradiations in vitro and in vivo. CEB showed improved circulation lifetime and accumulations in the tumor tissues in vivo, which induced efficient photodynamic and photothermal therapy. In addition, CEB induced the tumor microenvironment remodeling as indicated by the increase of CD8 + and CD4 + T cells, as well as a decrease of M2 TAM and Myeloid-derived suppressor cells (MDSCs). Our work developed a novel style of bioinspired AIE aggregates, which could eliminate both lung cancer cells and TAMs, and remodel the tumor environments to achieve an efficient lung cancer therapy. To the best of our knowledge, we are the first to use this style of bioinspired AIE aggregates for photo-mediated immunotherapy in lung cancer therapy.


Assuntos
Neoplasias Pulmonares , Nanopartículas , Humanos , Neoplasias Pulmonares/terapia , Imunoterapia , Peptídeos , Microambiente Tumoral
2.
Asian J Pharm Sci ; 17(5): 666-678, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36382298

RESUMO

The type II prokaryotic CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR/Cas9) adaptive immune system is a cutting-edge genome-editing toolbox. However, its applications are still limited by its inefficient transduction. Herein, we present a novel gene vector, the zwitterionic polymer-inspired material with branched structure (ZEBRA) for efficient CRISPR/Cas9 delivery. Polo-like kinase 1 (PLK1) acts as a master regulator of mitosis and overexpresses in multiple tumor cells. The Cas9 and single guide sgRNA (sgRNA)-encoded plasmid was transduced to knockout Plk1 gene, which was expected to inhibit the expression of PLK1. Our studies demonstrated that ZEBRA enabled to transduce the CRISPR/Cas9 system with large size into the cells efficiently. The transduction with ZEBRA was cell line dependent, which showed ∼10-fold higher in CD44-positive cancer cell lines compared with CD44-negative ones. Furthermore, ZEBRA induced high-level expression of Cas9 proteins by the delivery of CRISPR/Cas9 and efficient gene editing of Plk1 gene, and inhibited the tumor cell growth significantly. This zwitterionic polymer-inspired material is an effective and targeted gene delivery vector and further studies are required to explore its potential in gene delivery applications.

3.
J Colloid Interface Sci ; 625: 532-543, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35749848

RESUMO

The rising risk of lung cancer has become a primary global concern with high mortality and mobility. Presently, clinically used anticancer drugs show limited efficacy and significant side effects. A new generation of anticancer weapons is in great demand for lung cancer therapy. Herein, we have developed a novel style of biomimetic zeolitic imidazolate framework-8 (ZIF-8) based on the merits of cell membranes derived from human bone marrow mesenchymal stem cells (hBMSCs), which can navigate biological bombs herpes simplex virus type I thymidine kinase-encoded plasmids (pHSVtk) and ganciclovir (GCV) to treat lung cancer. The biological bomb-loaded structure can kill transfected lung cancer cells and neighboring lung cancer cells through the "bystander effect," which induces efficient suppression of lung cancer both in vitro and in vivo. The biomimetic nanoparticles show an enhanced circulation lifetime and drug accumulation in the tumor tissues and significantly inhibit the tumors. We have developed a straightforward approach to deliver biological bombs with biomimetic metal-organic frameworks for efficient lung cancer therapy. To the best of our knowledge, this is the first report of such a strategy for lung cancer therapy.


Assuntos
Bombas (Dispositivos Explosivos) , Neoplasias Pulmonares , Estruturas Metalorgânicas , Biomimética , Ganciclovir/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Estruturas Metalorgânicas/farmacologia
4.
J Drug Target ; 30(6): 614-622, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35078385

RESUMO

Lung cancer remains one of the leading causes of death in humans. Gefitinib is an inhibitor of epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) commonly used to suppress tumour growth. However, constantly use of gefitinib results in drug-resistance, reduced efficacy and undesired side effects. To circumvent these drawbacks, targeted and photothermal therapies have emerged as effective strategies. Herein, we are first to adopt a black phosphorus (BP) nanoparticle-based novel delivering strategy by combining gefitinib and cancer cytomembrane to treat non-small cell lung cancer (NSCLC). In these gefitinib-containing nano-carriers, cyanine 5 (Cy5) biotin-labelled BP was incorporated with cancer membrane and then consists of a nanomaterial (BPGM), which enabled to deliver gefitinib to the tumours effectively. The combination of BPGM showed reinforcing effects to suppress NSCLC cells and xenograft tumours without apparent adverse effects both in vitro and in vivo. BPGM facilitated the delivery of gefitinib to tumour tissue and extended its retention time within tumours. These studies thus suggest that BP may serve as novel delivery strategy for lung cancer.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Nanopartículas , Antineoplásicos/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/genética , Gefitinibe/farmacologia , Gefitinibe/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Mutação , Fósforo/farmacologia , Fósforo/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/uso terapêutico
5.
J Nanobiotechnology ; 19(1): 364, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789273

RESUMO

BACKGROUND: Malignant tumor is usually associated with epigenetic dysregulation, such as overexpression of histone deacetylase (HDAC), thus HDAC has emerged as a therapeutic target for cancer. Histone deacetylase inhibitor has been approved for clinical use to treat hematological cancers. However, the low solubility, short circulation lifetime, and high cytotoxicity partially limited their applications in solid tumor. METHODS: The upconversion nanoparticles (UC) modified with mesoporous silica (SUC) was used to load an HDACI, suberoylanilide hydroxamic acid (SAHA), and further camouflaged with M1 macrophage-derived exosome membranes (EMS). EMS was characterized in size and compositions. We also analyzed the epigenetic regulation induced by EMS. Furthermore, we evaluate the biodistribution and in vivo tumor inhibition after the systemic administration of EMS. RESULTS: This novel style spatiotemporal-resolved drug delivery system, EMS showed a high loading efficiency of SAHA. EMS could be taken up by lung cancer cells and lead to efficient epigenetic inhibition. We found that the integrin α4ß1 on M1-EM, was crucial for the homing of EMS to tumor tissues for the first time. In tumor-bearing mice, EMS showed spatiotemporal-resolved properties and facilitated the drug accumulation in the tumors, which induced superior anti-tumor effects. CONCLUSION: This novel style of spatiotemporal-resolved nanoparticles can be used as a theranostic platform for lung cancer therapy.


Assuntos
Epigênese Genética/efeitos dos fármacos , Exossomos , Membranas Artificiais , Sistemas de Liberação de Fármacos por Nanopartículas , Células A549 , Animais , Materiais Biomiméticos , Exossomos/química , Exossomos/metabolismo , Inibidores de Histona Desacetilases , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Nus , Sistemas de Liberação de Fármacos por Nanopartículas/química , Sistemas de Liberação de Fármacos por Nanopartículas/farmacocinética , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , Nanopartículas/química , Proteoma/efeitos dos fármacos , Vorinostat
6.
Pharmacol Res ; 171: 105787, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34314859

RESUMO

We adopted a novel strategy by combining histone deacetylase (HDAC) inhibitors with traditional chemotherapeutics to treat solid tumors. However, chemotherapeutics often have a narrow therapeutic index and need multiple administrations with undesired side effects that lead to the intolerance. To reduce the non-specificity of chemotherapeutics, targeted therapy was introduced to restrict such agents in the tumor with minimum effects on other tissues. We developed bioinspired artificial exosomes (AE), which enabled to deliver chemotherapeutics to the tumors effectively after systemic administration. AE were produced by incorporating membrane proteins from cancer cells into phospholipid liposomes that mimicked the plasma membrane. The synthesized AE were used for the delivery of broad-spectrum chemotherapeutic doxorubicin (DOX) and vorinostat (SAHA), an epigenetic inhibitor. The combination of DOX and SAHA showed synergistic effects on suppressing non-small cell lung cancer cells and xenograft tumors without apparent adverse effects. AE facilitated the delivery of drugs to tumor tissue and extended the retention time of drugs within tumors. Taken together, these studies suggest that the bioengineered artificial exosomes may serve as novel delivery strategy for chemotherapeutics to treat non-small cell lung cancer.


Assuntos
Antineoplásicos/administração & dosagem , Doxorrubicina/administração & dosagem , Portadores de Fármacos/administração & dosagem , Exossomos , Inibidores de Histona Desacetilases/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Vorinostat/administração & dosagem , Animais , Antineoplásicos/química , Linhagem Celular , Doxorrubicina/química , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Epigênese Genética , Humanos , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Carga Tumoral/efeitos dos fármacos , Vorinostat/química
7.
J Colloid Interface Sci ; 603: 319-332, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34186407

RESUMO

HYPOTHESIS: Histone deacetylase inhibitors (HDACIs), such as vorinostat (suberoylanilide hydroxamic acid, SAHA), has become a promising approach for the treatment of metastatic lung cancer. However, HDACIs usually showed a short circulation lifetime, low specificity, and low bioavailability, which limited their therapeutic effect in this field. We supposed that the use of biomimetic nanoparticles enabled to overcome the disadvantages of HDACIs, and improved the inhibition of metastatic lung cancer. EXPERIMENTS: SAHA was encapsulated into a pH-sensitive core constructed with Poly(lactic-co-glycolic acid) (PLAG) and 1,2-dioleoyloxy-3-(trimethylammonium) propane (DOTAP), followed by the camouflage with hybrid membranes derived from red blood cells and metastatic NCI-H1299 lung cancer cells (HRPDS). The physical and chemical properties were characterized with Transmission electron microscope (TEM), Size & Zeta potential analyzer. The cellular uptake was analyzed with Confocal laser scanning microscope (CLSM) and Flow cytometry (FACS). The biological effect analysis was performed with Western blotting (WB), RNA-Sequencing (RNA-Seq), and ChIP-Sequencing (ChIP-Seq). FINDINGS: HRPDS exhibited enhanced circulation lifetime in vivo and homotypic targeting to metastatic cells in the metastatic foci, which induced significant suppression of lung cancer liver metastasis. Our work opens a new avenue for the treatment of metastatic lung cancer by epigenetic inhibition based on this style of biomimetic nanovehicle.


Assuntos
Células Artificiais , Neoplasias Pulmonares , Apoptose , Linhagem Celular Tumoral , Epigênese Genética , Humanos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética
8.
ACS Appl Mater Interfaces ; 12(52): 57732-57745, 2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33326211

RESUMO

Conventional chemotherapy usually induces significant side effects due to its inability to discriminate between cancer and normal cells. Moreover, the efficacy of cancer elimination is still unsatisfied. Here, we fabricated a nanocomposite enabling high-performance dual combination therapy (chemo/photothermal therapy). This style of novel nanocomposites was constructed with doxorubicin (DOX)-loaded mesoporous silica gold (MSG) nanorods, which were further camouflaged with hybrid membranes derived from HeLa cells and red blood cells (HRMSGD). The hybrid membrane-camouflaged structure showed enhanced circulation lifetime and cell line-specific delivery of chemotherapeutics both in vitro and in vivo. The dual combination therapy by HRMSGD showed an unattainable therapeutic effect, compared with a single treatment, and inhibited tumor growth significantly. Furthermore, the nanoplatforms were photoacoustic-responsive, which showed real-time and noninvasive tracking capability. The present study established nanoplatforms with hybrid cell membrane-camouflaged multifunctional gold nanorods, which realized the combination of homotypic targeting, noninvasive tracking, chemotherapy, and photothermal therapy. To the best of our knowledge, this is the first study to use a natural membrane to camouflage mesoporous silica-modified gold nanorods, which opened a new avenue for cancer treatment.


Assuntos
Membrana Celular/química , Portadores de Fármacos/química , Ouro/química , Nanotubos/química , Dióxido de Silício/química , Animais , Fenômenos Químicos , Doxorrubicina/química , Doxorrubicina/farmacologia , Células HeLa , Humanos , Camundongos , Porosidade , Ensaios Antitumorais Modelo de Xenoenxerto
9.
ACS Appl Mater Interfaces ; 12(25): 28034-28046, 2020 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-32469502

RESUMO

Low-density lipoprotein cholesterol (LDL-C) is usually considered as a "bad cholesterol" for it is one of the major risk factors for coronary heart disease. As a scavenger of LDL-C, the low density lipoprotein receptor (LDLR) binds with LDL-C in the liver. However, the protein levels and function of LDLR are regulated by Proprotein convertase subtilisin/kexin type 9 (PCSK9). Loss of PCSK9 induces the increase of LDLR levels and reduction of plasma LDL-C. Here, we developed a novel style of artificial platelets with biomimetic properties, high stability, and long circulation which enabled the efficient delivery of siRNA targeting Pcsk9. The bioinspired nanoparticles induced Pcsk9 mRNA reduction by 66% in vitro. For in vivo studies, the nanoparticles accumulated in the liver to reduce Pcsk9 transcription, which results in ∼28% reduction in plasma LDL-C concentrations with negligible effects on either high density lipoprotein cholesterol (HDL-C) or triglycerides (TGs). These results demonstrated the use of artificial platelets to deliver siRNA and induce effective RNAi therapeutics to specifically lower LDL-C which provides a potential strategy to lower PCSK9 and treat hypercholesterolemia.


Assuntos
LDL-Colesterol/sangue , Nanopartículas/química , Pró-Proteína Convertase 9/metabolismo , Triglicerídeos/sangue , Animais , Plaquetas/química , Plaquetas/metabolismo , Humanos , Fígado/metabolismo , Pró-Proteína Convertase 9/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de LDL/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...