Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
BMC Cancer ; 21(1): 1220, 2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34774004

RESUMEN

BACKGROUND: The predictive role of blood-based tumor mutation burden (bTMB) for selecting advanced nonsmall cell lung cancer (NSCLC) patients who might benefit from immune checkpoint inhibitors (ICIs) is still under debate. Therefore, the purpose of this meta-analysis was to evaluate the efficacy of programmed cell death 1 (PD-1) /programmed cell death ligand 1 (PD-L1) inhibitors versus that of standard-of-care therapy in patients with NSCLC who were bTMB high and bTMB low. METHODS: PubMed, Embase, Cochrane, the Web of Science, and ClinicalTrials.gov were searched systematically from inception to February 2021 for studies of PD-1/PD-L1 inhibitors (durvalumab OR atezolizumab OR avelumab OR pembrolizumab OR Nivolumab) that provided hazard ratios (HRs) for overall survival (OS) or progression-free survival (PFS), or odds ratios (ORs) for objective response rate (ORR) in both bTMB high and bTMB low groups. RESULTS: A total of 2338 patients with advanced or metastatic NSCLC from six randomized controlled trials, which all used chemotherapy (CT) as a control, were included in this study. Compared with CT, PD-1/PD-L1 inhibitor therapy improved OS (HR 0.62, 95% CI 0.52-0.75, P < 0.01), PFS (HR 0.57, 95% CI 0.48-0.67, P < 0.01), and ORR (OR 2.69, 95% CI 1.84-3.93, P < 0.01) in bTMB-high NSCLC patients but not in bTMB-low patients (OS HR 0.86, 95% CI 0.69-1.07, P = 0.17; PFS HR 1.00, 95% CI 0.78-1.27, P = 0.98; ORR OR 0.63, 95% CI 0.49-0.80, P = 0.03). Subgroup analyses showed that these results were consistent across all subgroups (line of therapy, therapy regimen, type of NGS panel, PD-L1 expression, and cutoff value). Meta-regression analysis showed that the proportion of patients with squamous cell histology had no statistical effect on clinical outcomes. Sensitivity analyses illustrated that all results were stable. CONCLUSIONS: The efficacy of PD-1/PD-L1 inhibitor therapy in advanced NSCLC patients may be dependent on bTMB level. Patients with high bTMB tend to obtain significantly better OS, PFS, and ORR from PD-1/PD-L1 inhibitor therapy than from CT. However, because of multiple limitations, including those related to reproducibility, the results are exploratory and should be interpreted with caution.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Tasa de Mutación , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Nivolumab/uso terapéutico , Oportunidad Relativa , Supervivencia sin Progresión , Ensayos Clínicos Controlados Aleatorios como Asunto , Análisis de Regresión , Resultado del Tratamiento
2.
Integr Cancer Ther ; 20: 15347354211058168, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34802261

RESUMEN

BACKGROUND: Celastrus orbiculatus ethyl acetate extract (COE) has shown a strong anti-gastric cancer effect, but the understanding of its mechanism is still lacking. The results of previous studies indicated that COE may be able to inhibit the stemness of gastric cancer stem cells (GCSCs) by regulating PDCD4 and EIF3H expression. AIMS: To explore if COE could inhibit the stemness of GCSCs by regulating PDCD4 and EIF3H expression in vitro and in vivo. PROCEDURE: The GCSCs model was established by stem cell-conditioned culture. Spheroid formation and flow cytometry assays were used to detect the effect of COE on the spheroid formation ability of GCSCs and the percentage of CD44+/CD24+ and ALDH+ cell subpopulations. Western blot analysis was applied to measure the expression of GCSCs biomarkers (Nanog, Oct-4, and SOX-2), PDCD4, and EIF3H in GCSCs treated with COE; and RT-PCR was performed to investigate the effect of COE on PDCD4 mRNA expression in GCSCs. An in vivo tumorigenicity experiment was also conducted to evaluate the effect of COE on tumor-initiating ability of GCSCs in vivo; and the expression of PDCD4 and EIF3H in xenograft tissues was examined by immunohistochemistry (IHC) staining. RESULTS: After culture in stem cell-conditioned medium, SGC7901 cells manifested significantly enhanced spheroid formation ability, upregulated Nanog, Oct-4, and SOX-2 expression and increased percentages of CD44+/CD24+ and ALDH+ cell subpopulations, indicating successful establishment of the GCSCs model. COE treatment significantly inhibited the spheroid formation ability of GCSCs and reduced the percentage of CD44+/CD24+ and ALDH+ cell subpopulations. The western blot analysis showed a significant decrease of Nanog, Oct-4, SOX-2, and EIF3H expression and an increase of PDCD4 expression in GCSCs after COE treatment in a concentration-dependent manner. COE treatment also significantly upregulated the mRNA expression of PDCD4 in GCSCs. In addition, COE displayed a strong inhibitory effect on the tumor-initiating ability of GCSCs in vivo and upregulated PDCD4 and downregulated EIF3H expression in xenograft tissues. CONCLUSION: COE may be able to inhibit GC growth by suppressing the stemness of GCSCs via regulating PDCD4 and EIF3H expression.


Asunto(s)
Celastrus , Neoplasias Gástricas , Proteínas Reguladoras de la Apoptosis , Humanos , Células Madre Neoplásicas , Proteínas de Unión al ARN , Neoplasias Gástricas/tratamiento farmacológico
3.
Aging (Albany NY) ; 11(20): 8860-8878, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31619579

RESUMEN

OBJECTIVE: This study is implemented to probe into the function of lncRNA SBF2-AS1 as a competing endogenous RNA (ceRNA) to sponge microRNA-142-3p (miR-142-3p) in modulating TWF1 expression in the gemcitabine resistance of pancreatic cancer. RESULTS: LncRNA SBF2-AS1 was highly expressed in pancreatic cancer tissues and cells. SBF2-AS1 was found to be associated with gemcitabine resistance in pancreatic cancer. Knock-down of SBF2-AS1 inhibited proliferation, epithelial-mesenchymal transition, while promoting apoptosis of gemcitabine resistant pancreatic cancer cells. SBF2-AS1 inhibited the expression of TWF1 by competitively binding with miR-142-3p in pancreatic cancer. CONCLUSION: Our study demonstrates that knock-down of SBF2-AS1 inhibits the expression of TWF1 by competitively binding with miR-142-3p to induce gemcitabine resistance in pancreatic cancer. METHODS: Expression of SBF2-AS1 was tested in pancreatic cancer tissues and cells. Construction of AsPC-1/GEM and PANC-1/GEM cells with low expression of SBF2-AS1 was performed to determine the biological behaviors of drug-resistant cells. AsPC-1 and PANC-1 cells expressing SBF2-AS1 and/or miR-142-3p were constructed and treated with different concentrations of gemcitabine to detect the sensitivity of the cells to gemcitabine. The binding relationship between SBF2-AS1 and miR-142-3p and between miR-142-3p and TWF1 were determined.


Asunto(s)
Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos , MicroARNs/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Tirosina Quinasas/metabolismo , ARN Largo no Codificante/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Desoxicitidina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , MicroARNs/genética , Proteínas de Microfilamentos/genética , Persona de Mediana Edad , Proteínas Tirosina Quinasas/genética , ARN Largo no Codificante/genética , Regulación hacia Arriba , Gemcitabina
5.
Biomed Res Int ; 2019: 6543230, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30723742

RESUMEN

Vasculogenic mimicry (VM) with the pattern of endothelial independent tubular structure formation lined by aggressive tumor cells mimics regular tumor blood vessels to ensure robust blood supply and correlates with the proliferation, invasion, metastasis, and poor prognosis of malignant tumors, which was demonstrated to be a major obstacle for resistance to antiangiogenesis therapy. Therefore, it is urgent to discover methods to abrogate the VM formation of tumors, which possesses important practical significance for improving tumor therapy. Brucine is a traditional medicinal herb extracted from seeds of Strychnos nux-vomica L. (Loganiaceae) exhibiting antitumor activity in a variety of cancer models. In the present study, the effect of brucine on vasculogenic mimicry and the related mechanism are to be investigated. We demonstrated that, in a triple-negative breast cancer cell line MDA-MB-231, brucine induced a dose-dependent inhibitory effect on cell proliferation along with apoptosis induction at higher concentrations. The further study showed that brucine inhibited cell migration and invasion with a dose-dependent manner. Our results for the first time indicated that brucine could disrupt F-actin cytoskeleton and microtubule structure, thereby impairing hallmarks of aggressive tumors, like migration, invasion, and holding a possibility of suppressing vasculogenic mimicry. Hence, the inhibitory effect of brucine on vasculogenic mimicry was further verified. The results illustrated that brucine significantly suppressed vasculogenic mimicry tube formation with a dose-dependent effect indicated by the change of the number of tubules, intersections, and mean length of tubules. The in-depth molecular mechanism of vasculogenic mimicry suppression induced by brucine was finally suggested. It was demonstrated that brucine inhibited vasculogenic mimicry which might be through the downregulation of erythropoietin-producing hepatocellular carcinoma-A2 and matrix metalloproteinase-2 and metalloproteinase-9.


Asunto(s)
Neovascularización Patológica/tratamiento farmacológico , Estricnina/análogos & derivados , Strychnos nux-vomica/química , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Estricnina/química , Estricnina/farmacología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
6.
Am J Chin Med ; 46(5): 1129-1143, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29976080

RESUMEN

The extract of Celastrus orbiculatus (COE) has been shown to possess anti-Helicobacter pylori (H. pylori) activity and anticancer effects in vitro and in vivo. However, the molecular mechanism by which COE on precancerous lesions of gastric cancer (PLGC) has not been fully elucidated so far. The purpose of this study is to evaluate the effect and mechanism of COE in the rat model of PLGC, after the rat model of PLGC was successfully constructed. The effects of COE in gastric mucosa of rats with PLGC were tested using routine pathology and a transmission electron microscope (TEM) analysis. The protein and mRNA expression levels of epithelial mesenchymal transition (EMT) markers (E-cadherin, N-cadherin and Vimentin) and leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) were detected adopting techniques of immunohistochemistry (IHC), real-time PCR (RT-PCR) and western blot assays. The body weight of PLGC rats was significantly higher in the COE group than that in the untreated group. The process of PLGC was significantly reversed after COE treatment, shown by observing the changes of histopathological morphology and ultrastructure. Gastric mucosal epithelial cells in COE high dose (COE-H) group showed significantly higher expression levels of E-cadherin, and lower expression levels of N-cadherin, Vimentin and Lgr5 than those of the untreated group. COE could suppress the spatial distribution of Lgr5[Formula: see text] cell changes in PLGC rats. These findings suggested that the therapeutic mechanisms of COE in treating PLGC might be related with its effects on reversing the EMT process and inhibiting Lgr5 expression.


Asunto(s)
Celastrus/química , Células Epiteliales/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Mucosa Gástrica/efectos de los fármacos , Extractos Vegetales/administración & dosificación , Receptores Acoplados a Proteínas G/metabolismo , Neoplasias Gástricas/metabolismo , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Mucosa Gástrica/metabolismo , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/fisiopatología
7.
Chin J Integr Med ; 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25382615

RESUMEN

OBJECTIVE: To assess the effect of Celastrus orbiculatus (COE) on growth, invasion and migration of human gastric cancer MGC-803 cells and to explore the possible mechanism. METHODS: The effect of COE on cell viability, apoptosis, adhesion, invasion and migration were studied by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, flow cytometric, cell adhesion and transwell assay, respectively. The activity and expression of matrix metalloproteinase-9 (MMP-9) were determined by gelatin zymography, Western blot and quantitative real-time polymerase chain reaction analysis. Meanwhile, effects of COE on the expression of mitogen-activated protein kinases (MAPKs), serine threonine kinase (Akt), nuclear factor κB (NF-κB) were investigated with Western blot analysis. RESULTS: COE inhibited proliferation and induced apoptosis of MGC-803 cells in a dose-dependent manner. When treated with low-toxic (below 80 µg/mL) doses of COE, cell adhesion, invasion and migration were markedly suppressed. Furthermore, the gelatinolytic activity and expression of MMP-9 were also remarkably suppressed in a dose-dependent manner. In addition, upstream signaling pathways, including the phosphatidylinositol-3 kinase (PI3K)/Akt and NF-κB, were suppressed by COE. Additionally, the PI3K/Akt inhibitor, LY294002, in treating MGC-803 cells potently suppressed cell invasion and migration as well as expression of MMP-9. Similarly, the combined treatment with COE and LY294002 showed a synergistic effect compared with the treatment with COE or LY294002 alone in MGC-803 cells. CONCLUSIONS: COE inhibits invasion and migration of MGC-803 cells by reducing MMP-9 expression. It also inhibit PI3K/Akt and NF-κB signaling pathways, which may offer a novel approach for the treatment of human gastric cancer.

8.
Int Immunopharmacol ; 17(4): 1028-36, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24144813

RESUMEN

Rheumatoid arthritis (RA) joints are in a hypoxic condition. Hypoxia-induced migration and invasion of fibroblast-like synoviocytes (FLSs) are considered to play a critical role in the pathogenesis of RA. Among the key genes upregulated by hypoxia-inducible factor-1α (HIF-1α), CXC chemokine receptor 4 (CXCR4) plays an important role in FLS migration and invasion. Our previous studies have shown that celastrol exerts anti-arthritic effects by inhibiting FLS migration and invasion under normoxic conditions. However, the effect and molecular mechanisms underlying the effect of celastrol on hypoxia-induced FLS migration and invasion are poorly understood. In the present study, we assessed the effect of celastrol on hypoxia-induced FLS migration and invasion. Results showed that celastrol suppressed hypoxia-induced FLS migration and invasion. In addition, we also found that celastrol inhibited hypoxia-induced CXCR4 expression at both the mRNA and the protein levels in RA-FLSs. Meanwhile, it is revealed that celastrol inhibited the transcriptional activity of CXCR4 under hypoxic conditions by suppressing the binding activity of HIF-1α in the CXCR4 promoter, and blocked hypoxia-induced accumulation of nuclear HIF-1α. Furthermore, treatment with HIF-1α inhibitor reduced the hypoxia-induced expression and transcriptional activity of CXCR4. In conclusion, our results indicate that celastrol inhibits hypoxia-induced migration and invasion via suppression of HIF-1α mediated CXCR4 expression in FLSs under hypoxic conditions. These results provide a strong rationale for further testing and validation of the use of celastrol as a new alternative for using in the treatment of RA.


Asunto(s)
Antiinflamatorios/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Hipoxia/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Triterpenos/farmacología , Artritis Reumatoide/metabolismo , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/citología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Triterpenos Pentacíclicos , ARN Interferente Pequeño/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Membrana Sinovial/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...