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










Base de dados
Intervalo de ano de publicação
1.
Nat Med ; 30(3): 740-748, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38302715

RESUMO

Although antiprogrammed death 1 antibody plus chemotherapy has recently been approved for first-line esophageal squamous cell carcinoma (ESCC), antiprogrammed death-ligand 1 antibody may offer another combination option in this setting. In this multicenter, randomized, double-blinded phase 3 trial a total of 540 adults (aged 18-75 years) with unresectable, locally advanced, recurrent or metastatic ESCC and who had not received systemic treatment were enrolled. All patients were randomized at 2:1 to receive either sugemalimab (an anti-PD-L1 antibody; 1,200 mg) or placebo every 3 weeks for up to 24 months, plus chemotherapy (cisplatin 80 mg m-2 on day 1 plus 5-fluorouracil 800 mg m-2 day-1 on days 1-4) every 3 weeks for up to six cycles. At the prespecified interim analysis this study had met dual primary endpoints. With a median follow-up of 15.2 months, the prolongation of progression-free survival was statistically significant with sugemalimab plus chemotherapy compared with placebo plus chemotherapy (median 6.2 versus 5.4 months, hazard ratio 0.67 (95% confidence interval 0.54-0.82), P = 0.0002) as assessed by blinded independent central review. Overall survival was also superior with sugemalimab chemotherapy (median 15.3 versus 11.5 months, hazard ratio 0.70 (95% confidence interval 0.55-0.90, P = 0.0076). A significantly higher objective response rate (60.1 versus 45.2%) as assessed by blinded independent central review was observed with sugemalimab chemotherapy. The incidence of grade 3 or above treatment-related adverse events (51.3 versus 48.4%) was comparable between the two groups. Sugemalimab plus chemotherapy significantly prolonged progression-free survival and overall survival in treatment-naïve patients with advanced ESCC, with no unexpected safety signal. The ClinicalTrials.gov identifier is NCT04187352 .


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Adulto , Humanos , Anticorpos Monoclonais Humanizados/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Cisplatino/efeitos adversos , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/induzido quimicamente , Pessoa de Meia-Idade , Idoso
5.
J Gene Med ; 22(9): e3213, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32386256

RESUMO

BACKGROUND: The present study explored the role and mechanism of microRNA-874-3p (miR-874-3p) in the migration of the osteosarcoma cell line, U-2 OS. METHODS: The expression profile of osteosarcoma (OS) microRNA (GSE65071) datasets was downloaded from the Gene Expression Omnibus (GEO) database (https://www.ncbi.nlm.nih.gov/geo) to identify differentially expressed miRNAs in OS and its biological functions. A quantitative reverse transcription-polymerase chain reaction was performed to detect the expression of miR-874-3p and its target gene regulator of G protein 4 (RGS4) in human osteosarcoma cells U-2 OS and normal osteoblast hFOB1.19. Plasmid overexpression miR-874-3p and pcDNA-RGS4 were transfected into U-2 OS using Lipofectamine 2000 (Thermo Fisher, Waltham, MA, USA). Cell migration was measured using Transwell migration assays. Bioinformatic analysis and luciferase reporter assay were conducted to search for the target gene of miR-874-3p. RESULTS: In total, 167 differentially expressed miRNAs were detected after the analysis of GSE65071; of which 78 were up-regulated genes and 89 were down-regulated. miR-874-3p was down-regulated and selected for further analysis. The expression level of miR-874-3p in U-2 OS cells was significantly decreased compared to the hFOB1.19 cell line (p < 0.05). Overexpression of miR-874-3p significantly inhibited the proliferation and migration of U-2 OS cells and overexpression of RGS4 reversed the inhibitory effect of miR-874-3p on U-2 OS cells. Through luciferase report analyses and bioinformatic analysis, RGS4 may be the candidate target gene of miR-874-3p. CONCLUSIONS: In conclusion, overexpression of miR-874-3p suppressed OS cell proliferation and migration. Thus, miR-874-3p might present a therapeutic agent for the treatment of OS.


Assuntos
Proliferação de Células/genética , MicroRNAs/genética , Osteossarcoma/genética , Proteínas RGS/genética , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Osteossarcoma/patologia
6.
J Environ Pathol Toxicol Oncol ; 38(3): 217-227, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31679309

RESUMO

BACKGROUND: Natural active components have been reported to serve as adjuvant medications in the clinical practice of cancer therapeutics. However, the antineoplastic roles of atractylenolide III (ATL) are rarely reported. In the present study, we assessed the functions of ATL combined with docetaxel in gastric cancer cells. METHODS: Cell viability and cytotoxic activity were evaluated using CCK-8 and LDH-based cytotoxicity assays, respectively. Protein expression levels were measured by western blotting analysis. Annexin V-FITC/PI staining was used to evaluate cell apoptosis using flow cytometry. RESULTS: AGS and SGC-7901 cell viability was significantly inhibited in ATL combined with docetaxel group compared with docetaxel treatment alone. The levels of LDH, apoptosis rate, and the ratio of BAX to Bcl-2 were significantly elevated in combination treatment group compared to docetaxel treatment alone. Intriguingly, docetaxel combined with ATL resulted in a significant decrease in FGFR1, FGFR2, and FGFR4 protein expression compared with docetaxel treatment alone. Knockout of FGFR1, -2, and -4 exhibited a similar role of medications to inhibit growth and induce apoptosis in AGS and SGC-7901 cells. CONCLUSIONS: ATL and docetaxel treatment performed the synergistic effects on the inhibition of growth and induction of apoptosis in gastric cancer cells, and the underlying mechanism was mediated, at least partially, through the inhibition of FGFR1, -2, and -4.


Assuntos
Antineoplásicos/farmacologia , Docetaxel/farmacologia , Lactonas/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Sesquiterpenos/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Humanos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Neoplasias Gástricas/tratamento farmacológico
7.
Transl Lung Cancer Res ; 8(6): 989-999, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32010577

RESUMO

BACKGROUND: Bevacizumab is a monoclonal antibody (mAb) against vascular endothelial growth factor (VEGF) and used for treatments of various cancers. Due to the high costs of bevacizumab treatments, a biosimilar provides an affordable alternative therapy for cancer patients. METHODS: In this randomized, double-blind, multicenter, phase 3 study, locally advanced, metastatic or recurrent non-squamous non-small cell lung cancer (NSCLC) patients with wild-type epidermal growth factor receptor were enrolled and randomized (1:1) into IBI305 or bevacizumab groups. Patients received 6 cycles of paclitaxel/carboplatin plus IBI305 or bevacizumab 15 mg/kg intravenously followed by IBI305 or bevacizumab 7.5 mg/kg maintenance until disease progression, unacceptable toxicity or death. The primary endpoint was confirmed objective response rate (ORR) by an independent radiological review committee (IRRC) and secondary endpoints included disease control rate (DCR), progression-free survival (PFS), duration of response (DOR), overall survival (OS) and safety. RESULTS: A total of 450 NSCLC patients were enrolled (224 in IBI305 group and 226 in bevacizumab group). ORRs were 44.3% for IBI305 and 46.4% for bevacizumab, and the ORR ratio was 0.95 (90% CI: 0.803 to 1.135), within the predefined equivalence margin of 0.75 to 1.33. No significant difference in PFS (7.64 vs. 7.77 m, P=0.9987) was observed between the 2 groups. Serious adverse events (AEs) occurred in 33.5% (75/224) of patients in the IBI305 group and 37.6% (85/226) in the bevacizumab group. AEs ≥ grade 3 were similar in the IBI305 and bevacizumab groups [84.4% (189/224) vs. 89.8% (203/226), P=0.085]. CONCLUSIONS: IBI305 is similar to bevacizumab in terms of efficacy and safety. TRIAL REGISTRATION: Clinicaltrials.org Identifier: NCT02954172. Registered on 3 November 2016. Https://clinicaltrials.gov/.

8.
Drug Dev Ind Pharm ; 41(8): 1343-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25156484

RESUMO

Paclitaxel (PTX) is a natural broad-spectrum anticancer drug with poor aqueous solubility. PTX nanocrystals were formulated to improve the water solubility, and PTX nanosuspensions were prepared using anti-solvent precipitation, and then organic solvent and surfactants were removed by filtering through a vacuum system. The physical characterization of PTX nanocrystals were measured by transmission electron microscope, X-ray diffraction and differential scanning calorimetry. In addition, saturation solubility, in vitro release, stability and pharmacokinetic characteristics were examined. The average particle size of PTX nanocrystals was ∼200 nm, and they had a stable potential and a uniform distribution. Paclitaxel nanocrystals can effectively improve drug solubility and in vitro release. PTX pharmacokinetic and tissue distribution studies were compared after intravenous administration of nanocrystals versus a commercial injection formulation. PTX nanocrystals were rapidly distributed with a longer elimination phase. Moreover, tissue distribution indicated that PTX nanocrystals are mainly absorbed by the liver and spleen and may offer reduced renal and cardiovascular toxicity which may reduce side effects.


Assuntos
Nanopartículas/química , Nanopartículas/metabolismo , Paclitaxel/síntese química , Paclitaxel/farmacocinética , Animais , Antineoplásicos Fitogênicos/síntese química , Antineoplásicos Fitogênicos/farmacocinética , Feminino , Camundongos , Tamanho da Partícula , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia , Difração de Raios X
9.
J Nanosci Nanotechnol ; 12(8): 6163-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22962722

RESUMO

Ginsenosides, the major chemical composition of Chinese white ginseng (Panax ginseng C. A. Meyer), can inhibit tumor, enhance body immune function, prevent neurodegeneration. In this paper, for the first time we reported that the amount of ginsenosides in the equivalent extraction of the nanoscale Chinese white ginseng particles (NWGP) was 2.5 times more than that of microscale Chinese white ginseng particles (WGP). And the extractions from NWGP (1000 microg/ml) reached a high tumor inhibition of 64% exposed to human lung carcinoma cells (A549) and 74% exposed to human cervical cancer cells (Hela) after 72 h. Our work shows that the nanoscale Chinese WGP greatly improves the bioavailability of ginsenosides.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Ginsenosídeos/isolamento & purificação , Panax/química , Extratos Vegetais/farmacologia , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , História do Século XIX , Humanos , Microscopia Eletrônica de Varredura , Nanotecnologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...