Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Front Oncol ; 10: 1786, 2020.
Article in English | MEDLINE | ID: mdl-33014876

ABSTRACT

Cancer has always been an enormous threat to human health and survival. Surgery, radiotherapy, and chemotherapy could improve the survival of cancer patients, but most patients with advanced cancer usually have a poor survival or could not afford the high cost of chemotherapy. The emergence of oncolytic viruses provided a new strategy for us to alleviate or even cure malignant tumors. An oncolytic virus can be described as a genetically engineered or naturally existing virus that can selectively replicate in cancer cells and then kill them without damaging the healthy cells. There have been many kinds of oncolytic viruses, such as herpes simplex virus, adenovirus, and Coxsackievirus. Moreover, they have different clinical applications in cancer treatment. This review focused on the clinical application of oncolytic virus and predicted the prospect by analyzing the advantages and disadvantages of oncolytic virotherapy.

2.
World J Surg Oncol ; 18(1): 146, 2020 Jun 29.
Article in English | MEDLINE | ID: mdl-32600423

ABSTRACT

BACKGROUND: Colon adenocarcinoma (COAD) is a gastrointestinal tumor with a high degree of malignancy. Its deterioration process is closely related to the tumor microenvironment, and transcription factors (TF) play a regulatory role in this process. Currently, there is a lack of exploration between the genes related to the COAD tumor microenvironment and the survival prognosis of patients. Models composed of multiple genes usually predict the survival prognosis of patients more accurately than single genes. We can analyze the multigene models that can predict the prognosis of COAD from the current database. METHODS: The limma package of the R programming language is used for gene differential expression analysis. Kaplan-Meier curve is used to analyze the relationship between the patient risk score model and survival data. The hazard model is used to analyze the relationship between the risk score and the clinical data of COAD patients. The information of immune genes and immune cells is obtained from IMMPORT database and TIMER database. Receiver operating characteristic (ROC) curve is used to judge the stability of the model. RESULTS: We found 7 immune genes, which can built a risk score model to predict the survival prognosis of COAD. According to univariate and multivariate analysis, the risk score can be used as an independent predictor. The content of some immune microenvironment cells will also increase as the risk score increases. CONCLUSIONS: We found 7 immune genes, such as SLC10A2 (solute carrier family 10 member 2), CXCL3 (C-X-C motif chemokine ligand 3), IGHV5-51 (immunoglobulin heavy variable 5-51), INHBA (inhibin subunit beta A), STC1 (stanniocalcin 1), UCN (urocortin), and OXTR (oxytocin receptor), can constitute a model for predicting the prognosis of COAD. They may provide potential therapeutic targets for clinical treatment of COAD.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Colonic Neoplasms/pathology , Computational Biology/methods , Tumor Escape/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Biomarkers, Tumor/immunology , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Databases, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Prognosis , Survival Rate , Tumor Microenvironment
3.
BMC Cancer ; 17(1): 841, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29233126

ABSTRACT

BACKGROUND: The HER2-HER3 heterodimer significantly decreases survival in breast cancer patients. However, the prognostic value of HER2-HER3 overexpression remains unknown in gastric cancer (GC). METHODS: The expression levels of HER2, HER3, Akt, p-Akt, mTOR and p-mTOR were examined in specimens from 120 GC patients by immunohistochemistry and quantitative reverse transcription-PCR. The associations of HER proteins, PI3K/Akt/mTOR pathway-related proteins, clinicopathological features of GC, and overall survival (OS) were assessed. To comprehensively evaluate the prognostic values of pathway-related proteins, meta-analyses were conducted with STATA 11.0. RESULTS: HER2 overexpression was significantly associated with HER3 levels (P = 0.02). HER3 was highly expressed in gastric cancer tissues. High HER2 and HER3 levels were associated with elevated p-Akt and p-mTOR amounts (P < 0.05). Furthermore, HER2-HER3 co-expression was associated with high p-Akt and p-mTOR (P < 0.05) levels. Meanwhile, p-mTOR overexpression was tightly associated with differentiation, depth of invasion, lymph node metastasis, TNM stage and OS (P < 0.05). By meta-analyses, Akt, p-Akt, and mTOR levels were unrelated to clinicopathological characters. HER3 overexpression was associated with depth of invasion (OR = 2.39, 95%CI 1.62-3.54, P < 0.001) and lymph node metastasis (OR = 2.35, 95%CI 1.34-4.11, P = 0.003). Further, p-mTOR overexpression was associated with patient age, tumor location, depth of invasion (OR = 1.63, 95%CI 1.08-2.45, P = 0.02) and TNM stage (OR = 1.73, 95%CI 1.29-2.32, P < 0.001). In addition, HER2-HER3 overexpression corresponded to gradually shortened 5-year OS (P < 0.05), and significant relationships were shown among HER3, p-mTOR overexpression, and 1-, 3-, 5-year OS (P < 0.05). CONCLUSIONS: HER2-HER3 co-expression may potentially enhance mTOR phosphorylation. HER2-HER3 co-expression and p-mTOR are both related to the prognosis of GC patients.


Subject(s)
Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , TOR Serine-Threonine Kinases/metabolism , Aged , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Signal Transduction , Stomach Neoplasms/epidemiology
4.
Biomed Pharmacother ; 96: 450-458, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29031204

ABSTRACT

The purpose of present study was to investigate anti-tumor activity of Ginkgetin (GK) and its mechanism of action in breast cancer. The effects of GK on growth of human breast cancer cell lines MDA-MB-231, BT-474 and MCF-7 were examined by MTT assay. Cells apoptosis in MCF-7 cells were analyzed by TUNEL staining and annexin-V and propidium iodide double staining. The effects of GK on expression of apoptotic associated proteins and mitogen-activated protein kinases (MAPKs) were determined by western blotting. The results showed that GK significantly inhibited proliferation of MDA-MB-231, BT-474 and MCF-7 cells in vitro with time and dose dependent manners and induced apoptosis in MCF-7 cells. GK treatment obviously induced the tumor cells apoptosis and inhibited tumor growth in the MCF-7 xenograft nude mice. GK increased expression of Bax, cleaved-caspase-3, cleaved-caspase-8, cleaved-caspase-9, cleaved-PARP, and decreased the levels of Bcl-2 and survivin in MCF-7 cells. Moreover, GK treatment up-regulated expression of phospho extracellular-related kinase (p-ERK), p-p38 and phospho Jun-amino-terminal kinase (p-JNK) in MCF-7 cells in vitro, and increased numbers of p-p38, p-JNK and p-ERK positive cells in the tumor tissue in vivo. Strikingly, treatment of p38 inhibitor (or JNK inhibitor; ERK inhibitor) significantly prevented GK induced growth inhibition and apoptosis in MCF-7 cells. Collectively, our data exhibit GK exerts well anticancer effects in breast cancer cells, which at least in part, is via activation of the MAPKs. Our results provide a new approach for the treatment of breast cancer.


Subject(s)
Biflavonoids/pharmacology , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , MAP Kinase Signaling System/drug effects , Animals , Biflavonoids/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Female , Humans , MAP Kinase Signaling System/physiology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Random Allocation , Xenograft Model Antitumor Assays/methods
5.
PLoS One ; 12(6): e0176431, 2017.
Article in English | MEDLINE | ID: mdl-28570554

ABSTRACT

OBJECTIVES: Metastasis-associated protein 1 (MTA1) is a transcriptional regulator and significantly associated with prognosis of patients with cancer. However, its role as a potential prognostic marker in digestive tract cancer (DTC) is controversial. In this study, a meta-analysis was conducted to evaluate the MTA1 expression as a predictor of clinicopathology and survival of patients with DTC. METHODS: We searched PubMed, Ovid, Web of Science and Cochrane databases using multiple search strategies for eligible studies. STATA 11.0 software was used to pool the data and analyze the association, odds ratios (ORs) and 95% confidence intervals (CIs) were used to measure the strength of the association. Furthermore, the Newcastle-Ottawa scale was used to evaluate the quality of eligible studies. RESULTS: MTA1 overexpression was strongly associated with depth of invasion (OR = 1.88, 95%CI: 1.05-3.37, P = 0.03), lymph node metastasis (OR = 2.30, 95%CI: 1.76-3.01, P<0.001), vascular invasion (OR = 2.02, 95%CI: 1.40-2.91, P<0.001) and TNM stage (OR = 2.78, 95%CI: 1.63-4.74, P<0.001), and was related to 1- (RR = 1.84, 95%CI: 1.18-2.89, P = 0.008), 3- (RR = 1.74, 95%CI: 1.32-2.30, P<0.001) and 5-year (RR = 1.64, 95%CI: 1.18-2.27, P = 0.003) OS. Further, MTA1 was associated with 1- (RR = 4.16, 95%CI: 1.35-12.81, P = 0.01), 3- (RR = 1.90, 95%CI: 1.02-3.53, P = 0.04) and 5- (RR = 2.17, 95%CI: 1.41-3.32, P<0.001) year DFS. In subgroup analyses based on study quality and tumor type, MTA1 overexpression was obviously related to clinical parameters, such as lymph node metastasis and TNM stage, and was also associated with prognosis of patients with gastrointestinal or esophageal cancer. CONCLUSIONS: MTA1 expression is strongly correlated with metastasis-related variables, and represents a promising prognostic factor in DTC.


Subject(s)
Biomarkers, Tumor/physiology , Gastrointestinal Neoplasms/pathology , Histone Deacetylases/physiology , Repressor Proteins/physiology , Humans , Prognosis , Trans-Activators
6.
PLoS One ; 11(12): e0168085, 2016.
Article in English | MEDLINE | ID: mdl-28005970

ABSTRACT

AIMS: The mammalian target of rapamycin (mTOR) and phosphorylated mTOR (p-mTOR) occurring downstream in the PI3K/Akt/mTOR pathway, are regarded as potential prognostic markers for gastric cancer (GC). However, the prognostic value of mTOR/p-mTOR expression remains controversial. In this study, we determined the expression of mTOR, p-mTOR, p70S6k, and p-p70S6K in GC, and investigated the correlation between their overexpression, clinicopathological parameters, and overall survival (OS). METHODS: The expression of mTOR, p-mTOR, p70S6k, and p-p70S6K was examined in 120 GC patients by immunohistochemistry (IHC). The association of protein expression with clinicopathological features and OS was explored. The p-mTOR expression was detected in normal, adjacent, and GC tissues using Western blot. Eligible studies retrieved from PubMed, Ovid, Web of Science and Cochrane databases, were reviewed in this meta-analysis. RESULTS: IHC showed that the rates of expression of the signal transduction molecules mTOR, p-mTOR, p70S6k and p-p70S6K in GC were 60.8%, 54.2%, 53.3% and 53.3%, respectively. Overexpression of mTOR and p70S6K showed no significant association with clinical variables. Expression of p-mTOR was significantly associated with differentiation (P < 0.01), depth of invasion (P < 0.01), lymph node metastasis (P = 0.04) and TNM stage (P = 0.02). Expression of p-p70S6K was associated with differentiation (P = 0.006), depth of invasion (P < 0.001), and TNM stage (P = 0.02). In survival analysis, differentiation, depth of invasion, lymph node metastasis and TNM stage were not related to OS (all P > 0.05). Furthermore, p-mTOR and p-p70S6K expression, but not mTOR and p70S6K, were tightly associated with OS of GC patients (P = 0.006 and P < 0.001, respectively). In Western blot, p-mTOR was significantly higher in GC tissues than in normal and adjacent tissues. In the present meta-analysis, mTOR overexpression showed no relationship with any clinicopathological variables. However, p-mTOR was correlated with depth of invasion, and TNM stage (all P < 0.05), and its overexpression was associated with a shorter survival time (P < 0.001). CONCLUSION: The results suggest that p-mTOR is a more valuable prognostic factor than mTOR in GC.


Subject(s)
Biomarkers, Tumor/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Phosphorylation , Prognosis , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Survival Rate
7.
PLoS One ; 11(8): e0161219, 2016.
Article in English | MEDLINE | ID: mdl-27536774

ABSTRACT

BACKGROUND AND AIM: Human epidermal growth factor receptor (HER) family plays an important role in gastric cancer (GC), especially HER2. Too much attention has been paid to HER2; however, the functions of HER3 and HER4 overexpression in GC are always ignored. The clinicopathological and prognostic roles of HER3 and HER4 in GC are controversial. In this study, a systematic review and meta-analysis was conducted to evaluate the use of HER3 or HER4 as a predictor of clinicopathology and survival time in GC patients. METHODS: Eligible studies were searched on PubMed, Ovid, Web of Science, and Cochrane databases through multiple search strategies. Data collection and statistical analysis were carried out by the Revman 5.3 software. The Newcastle-Ottawa scale was used to assess the quality of included studies. RESULTS: A total of 448 studies about HER3 overexpression and GC, and 398 studies about HER4 overexpression and GC were searched. Of these, 5 eligible studies about HER3 including 1016 GC patients and 3 eligible studies about HER4 including 793 GC patients met the inclusion criteria. The results showed that HER3 and HER4 overexpression were significantly associated with depth of tumor invasion (OR = 0.44, 95%CI 0.29-0.67, P = 0.0002 and OR = 0.50, 95%CI 0.38-0.86, P = 0.007) and lymph node metastasis (OR = 0.40, 95%CI 0.20-0.77, P = 0.007 and OR = 0.57, 95%CI 0.38-0.86, P = 0.007), and HER3 overexpression reveals a tendency of later tumor node metastases (TNM) stage (OR = 0.50, 95%CI 0.22-1.15, P = 0.10) and predicts a worse survival time (RR = 0.71, 95%CI 0.61-0.84, P<0.00001), while HER4 overexpression had no correlation with TNM stage (OR = 0.60, 95%CI 0.20-1.78) and survival time (RR = 1.09, 95%CI 0.91-1.30). CONCLUSIONS: This meta-analysis indicated that HER3 plays an essential role in the clinicopathology and prognosis of GC. However, HER4 may not be an ideal prognostic factor for GC.


Subject(s)
Receptor, ErbB-3/metabolism , Receptor, ErbB-4/metabolism , Stomach Neoplasms/diagnosis , Biomarkers, Tumor/metabolism , Humans , Prognosis , Stomach Neoplasms/metabolism
8.
BMC Cancer ; 16: 655, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27543107

ABSTRACT

BACKGROUND: Long noncoding RNAs (lncRNAs) have been suggested to be involved in the development and progression of malignancies. However, the investigation of small nucleolar RNA host gene 20 (SNHG20) on cancer progression remains unknown. The present study aims to explore the clinical significance of SNHG20 and its potential molecular mechanism in colorectal cancer (CRC). METHODS: Quantitative real-time PCR (qRT-PCR) was used to measure the SNHG20 expression in a total of 107 CRC tissues and CRC cell lines. Loss of function approach was employed to explore the biological roles of SNHG20 in vitro. Its potential molecular mechanism was further verified by western blotting and qRT-PCR. RESULTS: The results suggested that SNHG20 expression was significantly upregulated in CRC tissues compared to corresponding normal tissues from 107 CRC patients. High expression of SNHG20 was remarkably associated with advanced TNM stage in patients with CRC. Multivariate analyses unraveled that SNHG20 expression was an independent prognostic factor for overall survival in CRC patients. Further functional assays revealed that knockdown of SNHG20 suppressed cell proliferation, invasion and migration, and cell cycle progression in CRC cells. Moreover, SNHG20 regulated cell growth through modulation of a series of cell cycle-associated genes. CONCLUSIONS: Our findings suggest that dysregulation of SNHG20 participates in CRC progression and may serve as a potential therapeutic target in CRC patients.


Subject(s)
Colorectal Neoplasms/genetics , RNA, Long Noncoding/genetics , Up-Regulation , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Disease Progression , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Male , Prognosis , Survival Analysis
9.
Pathol Oncol Res ; 21(3): 727-33, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25573590

ABSTRACT

Studies investigating the association between genetic polymorphism of aldehyde dehydrogenases-2 (ALDH-2) Glu487Lys and colorectal cancer (CRC) risk have reported conflicting results. Given this uncertainty, we carried out a critical analysis of published case-control studies to derive a more precise estimation of this relationship. Published literature from PubMed, EMBASE and China Knowledge Resource Integrated Database were retrieved, and the literature search was updated in June 2014. Eleven studies comprising 6965 subjects were selected (2300 cases and 4665 controls). Overall, our study showed no statistical significance for CRC risk associated with any of the genetic models of ALDH-2 Glu487Lys polymorphism. When studies were stratified for control source, a decreased risk of CRC for participants with Lys/Lys was observed in population based case-control studies [Lys/Lys vs. (Glu/Lys + Glu/Glu): odds ratio (OR) = 0.57, 95% confidence interval (CI) = 0.38-0.87]. Furthermore, we also confirmed the significant correlation between Glu487Lys polymorphism and the influence on the risk of rectal cancer in males [Glu/Glu vs. (Glu/Lys + Lys/Lys): OR = 1.52, 95%CI = 1.10-2.08]. The combined effects of the two gene polymorphisms [ALDH-2 and alcohol dehydrogenase 1B (ADH-1B)] were also studied. Compared with subjects having ALDH-2 Lys+ with ADH-1B His/His, ORs and 95%CIs for those with ALDH-2 Glu/Glu and ADH-1B His/His was 3.42(0.57-20.38). Similar trends were observed for the other two types of comparisons. Our study supports that ALDH-2 Glu487Lys polymorphism is associated with significant reduced risks of CRC in population-based samples, and of rectal cancer in males.


Subject(s)
Aldehyde Dehydrogenase/genetics , Colorectal Neoplasms/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic/genetics , Aldehyde Dehydrogenase, Mitochondrial , Case-Control Studies , China , Female , Humans , Male , Meta-Analysis as Topic , Prognosis , Risk Factors
10.
Tumour Biol ; 35(4): 3431-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24338709

ABSTRACT

Published data on the relationship between T309G polymorphism in the murine double minute 2 (MDM2) gene and susceptibility of digestive tract cancers (DTC) are inconclusive. Thus, the aim of this study is to determine whether MDM2 T309G polymorphism is associated with the risk of diverse DTC, including esophagus, stomach, liver, bile duct, pancreas, and colorectum cancers. Relevant studies were identified up to October 1, 2013. Crude odds ratio (OR) and 95% confidence interval (CI) were used as a measure of the strength of the association. The pooled result based on all studies showed that there was a statistically significant link between MDM2 T309G polymorphism and DTC susceptibility (T vs. G: OR = 0.82, 95%CI = 0.76-0.88). When stratified by race, significant associations were observed for all genetic models among Asians (especially in Chinese population), but not among Caucasians. Subgroup analyses according to tumor location indicated that the genetic variant was associated with esophageal (OR = 0.88, 95%CI = 0.81-0.96 for T vs. G), hepatocellular (OR = 0.69, 95%CI = 0.57-0.84 for T vs. G) and pancreatic cancer risk but not associated with cholangiocarcinoma or colorectum cancer susceptibility. Meanwhile, the G allele was also suggested to be associated with increased gastric cancer risk (OR = 0.68, 95%CI = 0.54-0.87 for TT + TG vs. GG for intestinal type of gastric cancer and OR = 0.18, 95%CI = 0.06-0.50 for TT vs. GG for Helicobacter pylori infection positive stomach cancer). Our study indicates that the MDM2 T309G polymorphism may be an ethnicity-dependent risk factor for DTC, especially for the upper gastrointestinal tract malignancies.


Subject(s)
Digestive System Neoplasms/ethnology , Digestive System Neoplasms/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Digestive System Neoplasms/etiology , Genes, p53 , Genotype , Humans , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...