Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Invest New Drugs ; 39(5): 1436-1438, 2021 10.
Article in English | MEDLINE | ID: mdl-33913072

ABSTRACT

Gastric cancer (GC) has the third highest rate of cancer incidence and mortality worldwide. First-line immune checkpoint inhibitor (ICI) therapy for advanced GC led to landmark breakthroughs, but which GC patients are most likely to benefit from ICI therapy needs to be investigated in depth and identified via valuable biomarkers. In this letter, we describe superior outcomes in Asian patients than in North American and European patients treated with ICI therapy, and we speculate that positive H. pylori status may be a beneficial prognostic factor for ICI therapy in patients with GC. Many studies have revealed that H. pylori-activated immune responses improve prognosis in patients with GC via increased PD-L1 expression and CD3+ T cells. We propose that H. pylori status should be emphasized in ongoing or forthcoming ICI therapy trials to maximize the benefits of treatment for patients with advanced GC. Further research is required to better understand the mechanisms of inflammation and cancer progression.


Subject(s)
Helicobacter Infections/epidemiology , Immune Checkpoint Inhibitors/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/ethnology , CD3 Complex/metabolism , Hedgehog Proteins/metabolism , Helicobacter pylori , Humans , Racial Groups , Stomach Neoplasms/pathology
2.
Cancer Lett ; 493: 120-127, 2020 11 28.
Article in English | MEDLINE | ID: mdl-32829006

ABSTRACT

LIM kinase 1 (LIMK1) and p21-activated kinase 4 (PAK4) are often over-expressed in breast tumors, which causes aggressive cancer phenotypes and unfavorable clinical outcomes. In addition to the well-defined role in regulating cell division, proliferation and invasion, the two kinases promote activation of the MAPK pathway and cause endocrine resistance through phosphorylating estrogen receptor alpha (ERα). PAK4 specifically phosphorylates LIMK1 and its functional partners, indicating possible value of suppressing both kinases in cancers that over-express PAK4 and/or LIMK1. Here, for the first time, we assessed the impact of combining LIMK1 inhibitor LIMKi 3 and PAK4 inhibitor PF-3758309 in preclinical breast cancer models. LIMK1 and PAK4 pharmacological inhibition synergistically reduced the survival of various cancer cell lines, exhibiting specific efficacy in luminal and HER2-enriched models, and suppressed development and ERα-driven signals in a BT474 xenograft model. In silico analysis demonstrated the cell lines with reliance on LIMK1 were the most prone to be susceptible to PAK4 inhibition. Double LIMK1 and PAK4 targeting therapy can be a successful therapeutic strategy for breast cancer, with a unique efficiency in the subtypes of luminal and HER2-enriched tumors.


Subject(s)
Breast Neoplasms/drug therapy , Lim Kinases/metabolism , Pyrazoles/administration & dosage , Pyrroles/administration & dosage , Thiazoles/administration & dosage , p21-Activated Kinases/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lim Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice , Phosphorylation/drug effects , Pyrazoles/pharmacology , Pyrroles/pharmacology , Thiazoles/pharmacology , Xenograft Model Antitumor Assays , p21-Activated Kinases/antagonists & inhibitors
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
...