Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Cancer Med ; 13(10): e6817, 2024 May.
Article in English | MEDLINE | ID: mdl-38112031

ABSTRACT

Circulating tumor DNA (ctDNA) has been widely used as a minimally invasive biomarker in clinical routine. However, a number of factors such as panel design, sample quality, patients' disease stages are known to influence ctDNA detection sensitivity. In this study, we systematically evaluated common factors associated with the variability of ctDNA detection in plasma and investigated ctDNA abundance in bronchoalveolar lavage (BAL). Whole exome profiling was conducted on 61 tumor tissue samples to identify tumor-specific variants, which were then used to design personalized assay MarRyDa® for ctDNA detection. DNA extracted from BAL fluid and plasma were genotyped using MarRyDa® platform. Our analysis showed that histological subtypes and disease stages had significant differences in ctDNA detection rate. Furthermore, we found that DNA purified from BAL supernatants contains the highest levels of ctDNA compared with BAL precipitates and plasma; therefore, utilizing BAL supernatants for tumor detection might provide additional benefits. Finally, we demonstrated that tumor cellularity played significant roles in the design of personalized ctDNA panel which eventually impacts ctDNA detection sensitivity. We suggest setting a flexible criteria for sample quality control and utilization of BAL might benefit more patients in clinics.


Subject(s)
Biomarkers, Tumor , Circulating Tumor DNA , Lung Neoplasms , Humans , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , Lung Neoplasms/genetics , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/blood , Female , Bronchoalveolar Lavage Fluid/chemistry , Male , Precision Medicine/methods , Neoplasm Staging , Early Detection of Cancer/methods , Middle Aged , Aged
2.
Colloids Surf B Biointerfaces ; 218: 112750, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35961116

ABSTRACT

The specific characteristics of the tumor microenvironment (TME) and monotherapy always lead to poor therapy effects for tumors. Hereby, we have developed a smart multifunctional theranostic agent-SSMID (Se@SiO2@MnO2-ICG/DOX) nanocomposites (NCs) that could intelligently respond to the TME for enhanced chemotherapy/photothermal/chemodynamic therapy guided by magnetic resonance imaging (MRI). The SSMID NCs were composed of indocyanine green (ICG) and doxorubicin hydrochloride (DOX) co-loaded porous Se@SiO2 @MnO2. Under the specific conditions of the TME (slightly acidic, H2O2 and GSH overexpression), the MnO2 NPs were specifically decomposed and then SSMID released Mn2+, DOX and Se, which played roles in chemodynamic therapy (CDT), chemotherapy, protecting normal tissues and inhibiting tumor cells by modulating reactive oxygen species (ROS), respectively. MnO2 reacted with glutathione (GSH) and H2O2 to generate O2 and Mn2+, which alleviated tumor hypoxia to improve chemotherapy and depleted GSH to enhance oxidative stress for chemodynamic therapy. More importantly, SSMID NCs could simultaneously exert the photothermal therapy (PTT) effect with near-infrared laser irradiation and promote the release of Mn2+ and DOX to achieve enhanced chemotherapy/chemodynamic therapy. In addition, the released Mn2+ could be used as a T1-weighted MRI contrast agent to monitor tumor location. The SSMID NCs exhibited a pronounced tumor growth inhibitory effect and promising biological safety, which develop a new method to rationally design nano-theranostic agents with enhanced performance for anti-tumor.


Subject(s)
Indocyanine Green , Neoplasms , Cell Line, Tumor , Contrast Media/pharmacology , Doxorubicin/pharmacology , Glutathione/pharmacology , Humans , Hydrogen Peroxide/pharmacology , Indocyanine Green/pharmacology , Manganese Compounds/pharmacology , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Oxides/pharmacology , Photothermal Therapy , Reactive Oxygen Species , Silicon Dioxide/pharmacology , Theranostic Nanomedicine , Tumor Microenvironment
3.
Front Bioeng Biotechnol ; 10: 939158, 2022.
Article in English | MEDLINE | ID: mdl-35814022

ABSTRACT

Based on the safety of prussian blue (PB) in biomedical application, we prepared manganese-based prussian blue (MnPB) nanocatalysts to achieve enhanced photothermal therapy and chemodynamic therapy. And we conducted a series of experiments to explore the therapeutic effects of MnPB nanoparticles (NPs) on non-small cell lung cancer (NSCLC) in vivo and in vitro. For in vitro experiments, the MnPB NPs suppressed growth of A549 cells by reactive oxygen species upregulation and near-infrared irradiation. Moreover, the MnPB NPs could inhibit lung cancer metastasis through downregulating the matrix metalloproteinase (MMP)-2 and MMP-9 expression in A549 cells. And for in vivo experiments, the MnPB NPs inhibited the growth of xenografted tumor effectively and were biologically safe. Meanwhile, Mn2+ as a T1-weighted agent could realize magnetic resonance imaging-guided diagnosis and treatment. To sum up, the results in this study clearly demonstrated that the MnPB NPs had remarkable effects for inhibiting the growth and metastasis of NSCLC and might serve as a promising multifunctional nanoplatform for NSCLC treatment.

4.
Front Chem ; 9: 789934, 2021.
Article in English | MEDLINE | ID: mdl-34820358

ABSTRACT

Non-small cell lung cancer (NSCLC) is considered to be a principal cause of cancer death across the world, and nanomedicine has provided promising alternatives for the treatment of NSCLC in recent years. Photothermal therapy (PTT) and chemodynamic therapy (CDT) have represented novel therapeutic modalities for cancer treatment with excellent performance. The purpose of this research was to evaluate the effects of PPy@Fe3O4 nanoparticles (NPs) on inhibiting growth and metastasis of NSCLC by combination of PTT and CDT. In this study, we synthesized PPy@Fe3O4 NPs through a very facile electrostatic absorption method. And we detected reactive oxygen species production, cell apoptosis, migration and protein expression in different groups of A549 cells and established xenograft models to evaluate the effects of PPy@Fe3O4 NPs for inhibiting the growth of NSCLC. The results showed that the PPy@Fe3O4 NPs had negligible cytotoxicity and could efficiently inhibit the cell growth and metastasis of NSCLC in vitro. In addition, the PPy@Fe3O4 NPs decreased tumor volume and growth in vivo and endowed their excellent MRI capability of observing the location and size of tumor. To sum up, our study displayed that the PPy@Fe3O4 NPs had significant synergistic effects of PTT and CDT, and had good biocompatibility and safety in vivo and in vitro. The PPy@Fe3O4 NPs may be an effective drug platform for the treatment of NSCLC.

6.
Int J Nanomedicine ; 14: 3845-3860, 2019.
Article in English | MEDLINE | ID: mdl-31213805

ABSTRACT

Background: Delay or failure of bone union is a significant clinical challenge all over the world, and it has been reported that bone marrow mesenchymal stem cells (BMSCs) offer a promising approach to accelerate bone fracture healing. Se can modulate the proliferation and differentiation of BMSCs. Se-treatment enhances the osteoblastic differentiation of BMSCs and inhibiting the differentiation and formation of mature osteoclasts. The purpose of this study was to assess the effects of porous Se@SiO2 nanocomposite on bone regeneration and the underlying biological mechanisms. Methods: We oxidized Se2- to develop Se quantum dots, then we used the Se quantum dots to form a solid Se@SiO2 nanocomposite which was then coated with polyvinylpyrrolidone (PVP) and etched in hot water to synthesize porous Se@SiO2 nanocomposite. We used XRD pattern to assess the phase structure of the solid Se@SiO2 nanocomposite. The morphology of porous Se@SiO2 nanocomposite were evaluated by scanning electron microscope (SEM) and the biocompatibility of porous Se@SiO2 nanocomposite were investigated by cell counting kit-8 (CCK-8) assays. Then, a release assay was also performed. We used a Transwell assay to determine cell mobility in response to the porous Se@SiO2 nanocomposite. For in vitro experiments, BMSCs were divided into four groups to detect reactive oxygen species (ROS) generation, cell apoptosis, alkaline phosphatase activity, calcium deposition, gene activation and protein expression. For in vivo experiments, femur fracture model of rats was constructed to assess the osteogenic effects of porous Se@SiO2 nanocomposite. Results: In vitro, intervention with porous Se@SiO2 nanocomposite can promote migration and osteogenic differentiation of BMSCs, and protect BMSCs against H2O2-induced inhibition of osteogenic differentiation. In vivo, we demonstrated that the porous Se@SiO2 nanocomposite accelerated bone fracture healing using a rat femur fracture model. Conclusion: Porous Se@SiO2 nanocomposite promotes migration and osteogenesis differentiation of rat BMSCs and accelerates bone fracture healing, and porous Se@SiO2 nanocomposite may provide clinic benefit for bone tissue engineering.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Femoral Fractures/therapy , Fracture Healing/drug effects , Mesenchymal Stem Cells/cytology , Nanocomposites/chemistry , Osteogenesis/drug effects , Selenium/pharmacology , Silicon Dioxide/pharmacology , Animals , Apoptosis/drug effects , Cells, Cultured , Cytoprotection/drug effects , Disease Models, Animal , Femoral Fractures/diagnostic imaging , Femoral Fractures/pathology , Hydrogen Peroxide/toxicity , Nanocomposites/ultrastructure , Porosity , Rats, Sprague-Dawley , Signal Transduction , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL