Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
J Cancer ; 13(9): 2768-2774, 2022.
Article in English | MEDLINE | ID: mdl-35812183

ABSTRACT

Background: The present study was designed to examine the prognostic value of a systemic inflammation marker-BAN score, which was established based on body-mass-index (BMI), albumin (ALB) and neutrophil-lymphocyte ratio (NLR) in resectable esophageal squamous cell carcinoma (ESCC) patients. Methods: A total of 420 newly diagnosed ESCC patients in our hospital between January 2008 and December 2013 were included. Their baseline characteristics were retrospectively reviewed and collected. BAN score was calculated as (BMI × ALB/ NLR). The optimal cutoff value for BAN score was defined as 28.0 in terms of survival. Patients were then allocated to high BAN (≥ 28.0) and low BAN (< 28.0) score groups. Results: Pretreatment BAN score was significantly associated with tumor length, white blood cell count, BMI, ALB and NLR levels. However, no significant difference was observed in patients' age, gender, tumor location, degree of differentiation, depth of invasion, lymph node involvement, tumor-node-metastasis (TNM) stage or other variables between groups. Moreover, those with high pretreatment BAN scores (≥ 28.0) tended to have favorable disease free survival (DFS) [hazard ratio (HR), 0.650; 95% confidence interval (CI), 0.481-0.877; P = 0.005] and overall survival (OS) (HR, 0.608; 95% CI, 0.445-0.829; P = 0.002) by univariate analysis. Furthermore, multivariate Cox regression analysis suggested that high BAN score (≥ 28.0) could serve as an independent predictor for both DFS (HR, 0.726; 95% CI, 0.532-0.993; P = 0.045) and OS (HR, 0.670; 95% CI, 0.485-0.927; P = 0.016). Conclusions: Pretreatment BAN score could independently predict long-term survival for resectable ESCC patients.

2.
J Cancer ; 10(25): 6244-6251, 2019.
Article in English | MEDLINE | ID: mdl-31772657

ABSTRACT

Background: To investigate the prognostic significance of the cumulative score based on preoperative fibrinogen and pre-albumin (FP score) in patients with gastric cancer after radical gastrectomy. Methods: Baseline characteristics, preoperative fibrinogen and pre-albumin levels were retrospectively reviewed in patients who underwent radical gastrectomy. The optimal cut-off values for fibrinogen and pre-albumin were defined as 4.0 g/L and 230.0 mg/L, respectively. Patients with elevated fibrinogen (≥ 4.0 g/L) and decreased pre-albumin (< 230.0 mg/L) levels were allocated an FP score of 2, those with only one of these two abnormalities were assigned a score of 1, and those with neither of the two abnormalities were allocated a score of 0. The prognostic value was examined by univariate and multivariate regression analyses. Results: The preoperative FP score was significantly correlated with age, tumor size, fibrinogen level, pre-albumin level and white blood cell count. No significant differences based on sex, tumor location, degree of differentiation, depth of invasion, lymph node status, tumor-node-metastasis (TNM) stage or adjuvant chemotherapy were identified between the groups. In addition, univariate survival analysis revealed that a high preoperative FP score was significantly associated with unfavorable disease-free survival (DFS) [hazard ratio (HR), 1.482; 95% confidence interval (CI), 1.222-1.796; P < 0.001] and overall survival (OS) (HR, 1.623; 95% CI, 1.315-2.002; P < 0.001). Moreover, after adjusting for other factors, a high preoperative FP score remained an independent predictor for impaired DFS (HR, 1.434; 95% CI, 1.177-1.747; P < 0.001) and OS (HR, 1.413; 95% CI, 1.136-1.758; P = 0.002) in multivariate Cox regression analysis. Conclusions: The preoperative FP score significantly predicts long-term survival for gastric cancer patients who have undergone radical gastrectomy.

3.
Pathol Res Pract ; 214(11): 1758-1764, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30249505

ABSTRACT

The rs36084323 A > G polymorphism in programmed cell death-1(PD-1) gene has been reported to be associated with cancer risk. However, the results of previous studies were inconsistent. Therefore, we performed a meta-analysis to identify the potential association, by searching the PubMed, EMBASE, Cochrane Library, and the Chinese CNKI, WANFANG and CBM databases. Data were extracted and odds ratios (ORs) with 95% confidence intervals (CIs) were estimated to assess the strength of the association. A total of 10 relevant studies involving 4445 cancer cases and 5126 controls were recruited. Overall, the results indicated that the PD-1 rs36084323 A > G polymorphism was not statistically associated with cancer risk. However, stratified analysis revealed that there was a statistically reduced cancer risk in Asians(G vs. A, OR = 0.89, 95%CI:0.81-0.97, P = 0.008, I2 = 48.8%; GG vs. AA, OR = 0.79, 95% CI:0.66-0.94, P = 0.008, I2 = 48.7%; GG/AG vs. AA, OR = 0.87, 95%CI:0.76-0.98, P = 0.017, I2 = 34.9%; GG vs. AG/AA, OR = 0.85, 95%CI:0.75-0.97, P = 0.027, I2 = 40%) and in the patients with EOC(AG vs. AA, OR = 0.69, 95%CI:0.54-0.90, P = 0.005, I2 = 0%; GG/AG vs. AA, OR = 0.67, 95%CI:0.52-0.85, P = 0.001, I2 = 0). Meta-regression showed that ethnicity (P = 0.029) but not cancer types (P = 0.792), source of controls (P = 0.207) or ample size (P = 0.585) were the sources of heterogeneity. This meta-analysis demonstrates the PD-1 rs36084323 A > G polymorphism is associated with decreased cancer risk in Asian, and suggests it could potentially serve as a biomarker to screen high-risk individuals. Large-scale and well-designed case-control studies are needed to enrich the evidence of this result.


Subject(s)
Genetic Predisposition to Disease/genetics , Neoplasms/genetics , Programmed Cell Death 1 Receptor/genetics , Asian People/genetics , Humans , Polymorphism, Single Nucleotide
4.
World J Gastroenterol ; 23(6): 986-998, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28246472

ABSTRACT

AIM: To clarify the mechanisms involved in the critical endoplasmic reticulum (ER) stress initiating unfolded protein response pathway modified by melatonin. METHODS: Hepatoma cells, HepG2, were cultured in vitro. Flow cytometry and TUNEL assay were used to measure HepG2 cell apoptosis. Western blotting and quantitative reverse transcription-polymerase chain reaction methods were used to determine the protein and messenger RNA levels of ER stress and apoptosis related genes' expression, respectively. Tissue microarray construction from patients was verified by immunohistochemical analysis. RESULTS: In the present study, we first identified that melatonin selectively blocked activating transcription factor 6 (ATF-6) and then inhibited cyclooxygenase-2 (COX-2) expression, leading to enhanced liver cancer cell apoptosis under ER stress condition. Dramatically increased CCAAT-enhancer-binding protein homologous protein level, suppressed COX-2 and decreased Bcl-2/Bax ratio by melatonin or ATF-6 siRNA contributed the enhanced HepG2 cell apoptosis under tunicamycin (an ER stress inducer) stimulation. In clinical hepatocellular carcinoma patients, the close relationship between ATF-6 and COX-2 was further confirmed. CONCLUSION: These findings indicate that melatonin as a novel selective ATF-6 inhibitor can sensitize human hepatoma cells to ER stress inducing apoptosis.


Subject(s)
Activating Transcription Factor 6/antagonists & inhibitors , Antioxidants/pharmacology , Apoptosis/drug effects , CCAAT-Enhancer-Binding Proteins/metabolism , Cyclooxygenase 2/metabolism , Endoplasmic Reticulum Stress/drug effects , Melatonin/pharmacology , Unfolded Protein Response/drug effects , Activating Transcription Factor 6/genetics , Down-Regulation , Hep G2 Cells , Humans , In Situ Nick-End Labeling , Liver Neoplasms , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tunicamycin/pharmacology , bcl-2-Associated X Protein/metabolism
5.
Int J Radiat Biol ; 89(12): 1079-86, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23875954

ABSTRACT

PURPOSE: To investigate the radiosensitizing effect and mechanism of action by the natural product Paeonol on lung adenocarcinoma both in vitro and in vivo. MATERIALS AND METHODS: Two lung adenocarcinoma cell lines (human lung adenocarcinoma cell line A549 and mouse Lewis lung carcinoma (LLC) cell line) were chosen for this research. In order to select the experimental concentrations of Paeonol, cytotoxicity was determined using a MTT (3-[4,5-dimethylthiazol-2-yl]-2,5- diphenyltetrazolium bromide) assay. A clonogenic assay was performed to measure the radiosensitizing effects. Apoptosis was determined by the Tunel (terminal deoxynucleotidyl transferase-mediated dUTP nick and labeling) assay and flow cytometry. Protein expression was analyzed by Western blotting. To test the radiosensitizing effect in vivo, a transplanted tumor model was established. RESULTS: The MTT assay showed that Paeonol inhibited proliferation of cells. Paeonol concentration ranged from an IC5 (5% inhibiting concentration) to an IC20 and was used at non-toxic concentrations for subsequent experiments. The clonogenic assay showed that Paeonol enhanced the radiosensitivity of cells. Data from the Tunel assay and flow cytometry verified that Paeonol enhanced radiation-induced apoptosis. Paeonol inhibited the activation of the PI3K/AKT (Phosphatidylinositol 3-kinase/ Protein Kinase B) pathway and down-regulated the expression of COX-2 (Cyclooxygenase-2) and Survivin. Paeonol (1718 mg/kg) combined with 10 Gy irradiation inhibited the growth of a transplanted tumor model in vivo, resulting in the longest tumor growth time, tumor growth delay and the highest inhibition ratio when compared with the radiotherapy alone group. CONCLUSIONS: It is reported for the first time that Paeonol has a radiosensitizing effect on lung adenocarcinoma both in vitro and in vivo. This effect could be related to the augmentation of radiation-induced apoptosis and the inhibition of the PI3K/Akt signalling pathway and its downstream proteins: COX-2 and Survivin.


Subject(s)
Acetophenones/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Apoptosis , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Lewis Lung , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cyclooxygenase 2/metabolism , Drug Screening Assays, Antitumor , Humans , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Survivin , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...