Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Environ Res ; 252(Pt 1): 118800, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38555088

ABSTRACT

With global climate changing, hypo-salinity events are increasing in frequency and duration because of continuous rainfall and freshwater inflow, which causes reduced cytosolic osmolarity and cellular stress responses in aquatic animals. Sea cucumbers are considered stenohaline because they lack osmoregulatory organs and are vulnerable to salinity fluctuations. In this study, we performed multiple biochemical assays, de novo transcriptomics, and widely targeted metabolomics to comprehensively explore the osmoregulatory mechanisms and physiological responses of sea cucumber Holothuria moebii to hypo-osmotic stress, which is a representative specie that is frequently exposed to hypo-saline intertidal zones. Our results found that H. moebii contracted their ambulacral feet and oral tentacles, and the coelomic fluid ion concentrations were reduced to be consistent with the environment. The microvilli of intestines and respiratory trees underwent degeneration, and the cytoplasm exhibited swelling and vacuolation. Moreover, the Na+, K+, and Cl- concentrations and Na+/K+-ATPase activity were significantly reduced under hypo-osmotic stress. The decrease in protein kinase A activity and increase in 5'-AMP level indicated a significant inhibition of the cAMP signaling pathway to regulate ion concentrations. And small intracellular organic molecules (amino acids, nucleotides and their derivatives) also play crucial roles in osmoregulation through oxidative deamination of glutamate, nucleotide catabolism, and nucleic acid synthesis. Moreover, lysosomes and peroxisomes removed oxidative damage, whereas antioxidant metabolites, such as N-acetyl amino acids and glutathione, were increased to resist oxidative stress. With prolonged hypo-osmotic stress, glycerophospholipid metabolism was enhanced to maintain membrane stability. Furthermore, acyl-CoA-binding protein activity was significantly inhibited, and only a small amount of acylcarnitine was significantly accumulated, which indicated a disruption in energy metabolism. PPAR signaling pathway and choline content were up-regulated to promote fatty acid metabolism under hypo-osmotic stress. Overall, our results provide new insights into the osmoregulatory mechanisms and physiological responses of sea cucumbers to hypo-osmotic stress.


Subject(s)
Antioxidants , Energy Metabolism , Holothuria , Osmoregulation , Osmotic Pressure , Animals , Holothuria/physiology , Antioxidants/metabolism , Salinity
2.
Fish Shellfish Immunol ; 126: 113-121, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35609761

ABSTRACT

Proprotein Convertase Subtilisin/Kexin Type 9 (PCSK9) in mammals is a multifunctional protein. In this study, PCSK9 of marine fish Epinephelus coioides was characterized. The full-length cDNA of E. coioides PCSK9 was 2458 bp in length containing 185 bp 5' UTR, 263 bp 3' UTR and 2010 bp open reading frame (ORF) encoding 669 amino acids with the predicted molecular weight of 71 kDa and the theoretical PI of 6.6. Similar to other members of PCSK9 family, E. coioides PCSK9 has three conserved domains: Inhibitor_ I9 super family, Peptidases_ S8_ PCSK9_ Proteinase K_ like, and PCSK9_ C-CRD super family. E. coioides PCSK9 mRNA could be detected in all the tissues examined by real-time quantitative PCR, with the highest expression in the brain, followed by skin, trunk kidney, head kidney, intestine, blood, liver, spleen, gill, muscle and heart. E. coioides PCSK9 was distributed in both the cytoplasm and nucleus. The expression of E. coioides PCSK9 was significantly upregulated during Singapore grouper iridovirus (SGIV) infection. Upregulated PCSK9 could significantly affect the activities of nuclear factor kappaB (NF-κB) promoter, SGIV-induced apoptosis, and the expressions of the key SGIV genes (ICP18, LITAT, MCP, and VP19) and the E. coioides proinflammatory factors (IL-6, IL-1ß, IL-8, and TNF-α). The results illustrated that E. coioides PCSK9 might be involved in the pathogen infection by regulating the innate immune response.


Subject(s)
Bass , DNA Virus Infections , Fish Diseases , Iridovirus , Ranavirus , Animals , Cloning, Molecular , Fish Proteins/chemistry , Immunity, Innate/genetics , Iridovirus/physiology , Mammals/genetics , Mammals/metabolism , Proprotein Convertase 9/genetics , Ranavirus/physiology
4.
Oncotarget ; 7(4): 3884-96, 2016 Jan 26.
Article in English | MEDLINE | ID: mdl-26646697

ABSTRACT

Current guidelines for lung cancer treatment with EGFR tyrosine kinase inhibitors (TKI) include only patients with mutated EGFR, although some patients with wildtype EGFR (wt-EGFR) have exhibited positive responses to this therapy as well. Biomarkers predicting the benefit from EGFR-TKIs treatment remain to be determined for patients with wild-type EGFR.Here, we report that wt-EGFR overexpression transformed cells in vitro and induced tumorigenesis in vivo in transgenic mouse models. Wt-EGFR driven lung cancer was hypersensitive to TKI treatment in mouse model. Lung cancer patients with high-expression of wt-EGFR showed longer Overall Survival in comparison to low-expression patients after TKI treatment. Our data therefore suggest that treatment with EGFR inhibitors should be extended to include not only patients with mutated EGFR but also a subset of patients with overexpression of wt-EGFR.


Subject(s)
Bronchi/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , ErbB Receptors/metabolism , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Animals , Biomarkers, Tumor , Blotting, Western , Bronchi/drug effects , Bronchi/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/genetics , Female , Genotype , Humans , Immunoenzyme Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Transgenic , NIH 3T3 Cells , Neoplasm Staging , Prognosis , Survival Rate
5.
Neurocrit Care ; 24(3): 472-80, 2016 06.
Article in English | MEDLINE | ID: mdl-26627226

ABSTRACT

BACKGROUND: Aneurysmal subarachnoid hemorrhage (SAH) is a common condition with relatively poor clinical outcome. Pulmonary complication after SAH is an important contributor to poor outcome. Previous studies have shown that labile zinc and inflammatory mediators participate in many pathophysiological processes. The present study investigated the effects of SAH on the levels of labile zinc and certain proinflammatory factors in rat lung and determined the effect of erythropoietin (EPO) on the pulmonary labile zinc and the inflammatory factor after SAH in rats. METHODS: Experiment 1 aimed to investigate the time course of increase of pulmonary labile zinc, wet/dry weight ratio, and the expression of inflammatory mediators after SAH. In Experiment 2, we chose the maximum time point which lung injury was maximally severity and assessed the effect of EPO on regulation of the pulmonary labile zinc, inflammatory reaction, and wet/dry weight ratio after SAH. RESULTS: SAH caused a gradual increase of pulmonary labile zinc as demonstrated by fluorescence staining with Zinpyr-4. The levels of TNF-α and IL-8 and the lung wet/dry weight ratios were higher in the SAH groups compared to the control group and peaked on 3 days following SAH (p < 0.05). EPO significantly reduced the pulmonary labile zinc, the inflammatory mediators, and the lung wet/dry weight ratio compared with SAH group (p < 0.05). CONCLUSIONS: EPO can protect lung from SAH-induced injury by attenuating pulmonary inflammation and labile zinc accumulation in vivo.


Subject(s)
Erythropoietin/pharmacology , Inflammation Mediators/metabolism , Lung Injury/metabolism , Lung/metabolism , Subarachnoid Hemorrhage/metabolism , Zinc/metabolism , Animals , Disease Models, Animal , Lung/pathology , Lung Injury/prevention & control , Male , Rats , Rats, Sprague-Dawley
6.
Oncotarget ; 6(32): 33019-32, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26361045

ABSTRACT

BACKGROUD: Programmed cell death-ligand 1 (PD-L1) and driver mutations are commonly seen in non-small-cell lung cancer (NSCLC). However, the prevelance of PD-L1 over-expression and its prognostic value in Epstein-Barr virus (EBV) associated pulmonary lymphoepithelioma-like carcinoma (LELC) remains poorly understood. METHODS: A total of 214 NSCLC patients and 113 surgically treated pulmonary LELC patients were included. Paraffin-embedded tumor sections were stained with PD-L1 antibody. Correlations between PD-L1 expression and clinicopathological features as well as survival outcomes were analyzed. RESULTS: The frequency of PD-L1 over-expression in NSCLC was 51.4%. No significant association was observed between common driver mutations and PD-L1 over-expression. Remakably, the positive rate of PD-L1 in pulmonary LELC was 74.3%. High PD-L1 expression was associated with impaired disease-free survival (DFS) compared with low PD-L1 expression (p = 0.008). Multivariate analysis shows that PD-L1 expression level, N stage and M stage were independent prognostic factors for DFS. N stage and M stage but not PD-L1 expression level were significantly associated with overall survival (OS). CONCLUSIONS: PD-L1 over-expression was not related to common driver mutations in NSCLC. Pulmonary LELC have remarkably high incidence of PD-L1 expression. PD-L1 was a negative prognostic factor for DFS in surgically resected pulmonary LELC. These findings may provide a rationale for immunotarget therapy in this virus-associated lung cancer.


Subject(s)
Lung Neoplasms/metabolism , Programmed Cell Death 1 Receptor/biosynthesis , Disease-Free Survival , Female , Humans , Lung Neoplasms/pathology , Male , Middle Aged
7.
Oncotarget ; 6(16): 14209-19, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25895031

ABSTRACT

BACKGROUNDS: Recent clinical trials have shown that immune-checkpoint blockade yields remarkable response in a subset of non-small cell lung cancer (NSCLC) patients. However, few studies directly focus on the association between epidermal growth factor receptor (EGFR) mutational status and programmed cell death-ligand 1 (PD-L1) expression. We examined whether PD-L1 is related to clinicopathologic factors and prognosis in patients with advanced NSCLC treated with EGFR-tyrosine kinase inhibitors (EGFR-TKIs). METHODS: One-hundred and seventy patients with advanced NSCLC were explored. Paraffin-embedded tumour sections were stained with PD-L1 antibody. EGFR mutation was examined by fluorescent quantitative polymerase chain reaction (PCR). The correlations between PD-L1 expression and EGFR status and survival parameters were analyzed. RESULTS: The overall frequency of PD-L1 over-expression was 65.9% (112/170). In lung adenocarcinoma, PD-L1 tended to be associated with mutant EGFR (PD-L1 overexpression in mutant and wild-type EGFR, 64/89 (71.9%) vs. 32/56 (57.1%), respectively; p=0.067). Subgroup analyses showed that high PD-L1 expression was associated with significantly shorter overall survival (OS) in EGFR wild-type patients (p=0.029) but not in EGFR mutant patients (p=0.932) treated with EGFR-TKIs. Even more, for EGFR mutant patients, higher expression of PD-L1 might only signal better outcome with TKIs. CONCLUSIONS: High PD-L1 expression was likely to be associated with the presence of EGFR mutation in advanced lung adenocarcinoma. For EGFR wild-type patients, the PD-L1 over expression can be considered as a poor prognostic indicator of OS.


Subject(s)
B7-H1 Antigen/biosynthesis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , ErbB Receptors/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , ErbB Receptors/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Survival Analysis
8.
Asian Pac J Cancer Prev ; 16(2): 551-7, 2015.
Article in English | MEDLINE | ID: mdl-25684486

ABSTRACT

BACKGROUND: Differentiating morphologic features based on hematoxylin-eosin (HE) staining is the most common method to classify pathological subtypes of non-small-cell lung cancer (NSCLC). However, its accuracy and inter-observer reproducibility in pathological diagnosis of poorly differentiated NSCLC remained to be improved. MATERIALS AND METHODS: We attempted to explore the role of immunohistochemistry (IHC) staining in diagnosing pulmonary squamous cell carcinoma (SQCC) with poorly differentiated features by HE staining or with elevated serum adenocarcinoma-specific tumor markers (AD-TMs). We also compared the difference of epidermal growth factor receptor (EGFR) mutation rate between patients with confirmed SQCC and those with revised pathological subtype. Logistic regression analyses were used to test the association between different factors and diagnostic accuracy. RESULTS: A total of 132 patients who met the eligible criteria and had adequate specimens for IHC confirmation were included. Pathological revised cases in poor differentiated subgroup, biopsy samples and high-level AD-TMs cases were more than those with high/moderate differentiation, surgical specimens and normal-level AD-TMs. Moreover, biopsy sample was a significant factor decreasing diagnostic accuracy of pathological subtype (OR, 4.037; 95% CI 1.446-11.267, p=0.008). Additionally, EGFR mutation rate was higher in patients with pathological diagnostic changes than those with confirmed SQCC (16.7% vs 4.4%, p=0.157). CONCLUSIONS: Diagnosis based on HE staining only might cause pathological misinterpretation in NSCLC patients with poor differentiation or high-level AD-TMs, especially those with biopsy samples. HE staining and IHC should be combined as pathological diagnostic standard. The occurrence of EGFR mutations in pulmonary SQCC might be overestimated.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/metabolism , Carcinoma, Adenosquamous/diagnosis , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Squamous Cell/diagnosis , Cell Differentiation , Lung Neoplasms/diagnosis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cohort Studies , ErbB Receptors/genetics , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Middle Aged , Mutation/genetics , Neoplasm Grading , Neoplasm Staging , Prognosis
9.
Med Oncol ; 32(3): 61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25663066

ABSTRACT

Non-small cell lung cancer (NSCLC) patients harboring KRAS mutation were associated with worse prognosis and lower response to epidermal growth factor receptor (EGFR) target therapy than those with wild-type tumors. However, whether the underlying biological differences are associated with the efficacy of cytotoxic chemotherapy in advanced NSCLC patients remained controversial. We searched electronic databases for eligible literatures. The primary outcomes were objective response rate (ORR), 6-month and 1-year progression-free survival (PFS) rate. The pooled odds ratio (OR) was calculated using random-effect model. Subgroup analyses stratified by literature type, mutation analysis method, therapeutic regimen, patient origin, and EGFR mutation status in KRAS wild-type patients were proposed. Heterogeneity and publication bias were quantitatively evaluated. A total of ten studies involving 1,677 advanced NSCLC patients with known KRAS mutation status who had received first-line chemotherapy were included. KRAS mutants had lower ORR than wild-type patients (25.1 vs. 34.4%) significantly (OR 0.67, 95% CI 0.50-0.88, P = 0.004). Additionally, patients with KRAS mutation had numerically lower 6-month (51.0 vs. 56.8%) and 1-year (10.3 vs. 13.3%) PFS rate than wild-type patients, but there was no significant difference between the two groups (OR 0.75, 95% CI 0.54-1.04, P = 0.08; OR 0.75, 95% CI 0.47-1.21, P = 0.25). Results of the subgroup analyses were almost concordant with the overall ones. This comprehensive analysis revealed that advanced NSCLC patients with KRAS mutations had significantly lower ORR and potentially lower 6-month/1-year PFS rate compared with wild-type patients after first-line chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Disease-Free Survival , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Publication Bias , Treatment Outcome
10.
J Thorac Oncol ; 10(6): 910-23, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25658629

ABSTRACT

INTRODUCTION: Epidermal growth factor receptor (EGFR) mutation status was reported to be associated with programmed death-ligand 1 (PD-L1) expression. However, the molecular mechanism of PD-L1 regulation by EGFR activation and the potential clinical significance of blocking PD-1/PD-L1 in EGFR-mutant non-small-cell lung cancer (NSCLC) treated with EGFR tyrosine kinase inhibitors (TKIs) were largely unknown. METHODS: Western blot, real-time polymerase chain reaction, immunofluorescence, and flow cytometry were employed to explore the association between PD-L1 and EGFR activation. Then, we used EGFR-TKIs and downstream pathways inhibitors to clarify the detailed signaling pathway involved in PD-L1 regulation. Cell apoptosis, viability, and enzyme-linked immunosorbent assay test were used to study the immune suppression by EGFR activation and immune reactivation by EGFR-TKIs and/or PD-1 blocking in tumor cells and human peripheral blood mononuclear cells coculture system. RESULTS: We found that EGFR activation by EGF stimulation, exon-19 deletions, and L858R mutation could induce PD-L1 expression. EGFR activation upregulated PD-L1 through p-ERK1/2/p-c-Jun but not through p-AKT/p-S6 pathway. PD-L1 mediated by EGFR activation could induce the apoptosis of T cells through PD-L1/PD-1 axis in tumor cells and peripheral blood mononuclear cells coculture system. Inhibiting EGFR by EGFR-TKIs could free the inhibition of T cells and enhance the production of interferon-γ. Synergistic tumor cell killing effects were not observed with EGFR-TKIs and anti-PD-1 antibody combination treatment in coculture system. CONCLUSIONS: Our results imply that EGFR-TKIs could not only directly inhibit tumor cell viability but also indirectly enhance antitumor immunity through the downregulation of PD-L1. Anti-PD-1/PD-L1 antibodies could be an optional therapy for EGFR-TKI sensitive patients, especially for EGFR-TKIs resistant NSCLC patients with EGFR mutation. Combination of EGFR-TKIs and anti-PD-1/PD-L1 antibodies treatment in NSCLC is not supported by the current study but warrant more studies to move into clinical practice.


Subject(s)
B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , ErbB Receptors/immunology , Lung Neoplasms/immunology , Protein Kinase Inhibitors/pharmacology , Apoptosis/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Exons , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Sequence Deletion , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Escape/drug effects , Up-Regulation
11.
Tumour Biol ; 36(5): 3389-97, 2015 May.
Article in English | MEDLINE | ID: mdl-25527156

ABSTRACT

Recent studies have shown the prognostic nutritional index (PNI) had prognostic value in some solid tumors. However, no studies have examined its prognostic role in small-cell lung cancer (SCLC) patients. In this retrospective study, 724 consecutive SCLC patients were included between 2006 and 2013. Demographic, clinical, and laboratory data were collected. The PNI was calculated as 10 × serum albumin value (g/dl) + .005 × peripheral lymphocyte count (per mm(3)). Univariate and multivariate analyses were performed to assess the prognostic value of relevant factors. The optimal cut-off value of PNI for OS stratification was determined to be 52.48. A total of 464 and 260 patients were assigned to low and high PNI groups, respectively. Compared with low PNI, high PNI was associated with older age, advanced stage, and elevated lactate dehydrogenase (LDH). Median overall survival (OS) was worse in the low PNI group (low vs high, 15.90 vs 25.27 months; HR, 0.62; p < 0.001). In multivariate analysis, stage, performance status, LDH, and PNI were independent prognostic factors for OS. Subgroup analysis showed PNI was generally a significant prognostic factor in different clinical situations. The assessment of PNI could assist the identification of patients with poor prognosis and be a hierarchical factor in the future SCLC clinical trials.


Subject(s)
Lung Neoplasms/mortality , Small Cell Lung Carcinoma/mortality , Adult , Aged , Aged, 80 and over , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/pathology , Lymphocyte Count , Male , Middle Aged , Prognosis , Retrospective Studies , Serum Albumin/analysis , Small Cell Lung Carcinoma/blood , Small Cell Lung Carcinoma/pathology
12.
Clin Lung Cancer ; 16(2): 144-151.e1, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25454007

ABSTRACT

BACKGROUND: The strong association between smoking history and the presence of epidermal growth factor receptor (EGFR) mutations has been proven in non-small-cell lung cancer (NSCLC), which explains the favorable response to EGFR-tyrosine kinase inhibitor (EGFR-TKI) therapy in nonsmoking NSCLC patients. However, few studies directly focus on the relationship between EGFR-TKI's efficacy and smoking history in NSCLC EGFR-mutant patients. METHODS: Electronic databases were searched for eligible literatures. Data on objective response rates, disease control rates, and progression-free survival (PFS) stratified by smoking status were extracted and synthesized on the basis of a random-effect model. Subgroup and sensitivity analyses were conducted. RESULTS: A total of 9 studies that involved a total of 1029 EGFR-mutant advanced NSCLC patients after EGFR-TKI treatment were included. In overall, nonsmoking was associated with significant prolonged PFS (HR, 0.73, 0.60 to 0.88; P = .001) compared to ever smokers. However, only marginal improvements without statistical significance in objective response rates (odds ratio, 1.11; 95% confidence interval, 0.85 to 1.46; P = .433) and disease control rate (odds ratio, 1.04; 95% confidence interval, 0.82 to 1.33; P = .740) were observed. Subgroup analyses showed that the benefits of PFS in nonsmokers were predominantly presented in pooled results of studies enrolling patients with active EGFR mutations, studies involving previously treated patients, and retrospective studies. Additionally, we failed to observe any significant benefit from nonsmokers in every subgroup for objective response rates and disease control rate. CONCLUSION: For advanced NSCLC patients with EGFR mutations, nonsmoking is associated with longer PFS than ever smoking after EGFR-TKIs treatment. Smoking history should be considered an essential factor in studies regarding EGFR-targeted agents toward EGFR-mutant patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Lung Neoplasms/pathology , Mutation , Protein Kinase Inhibitors/pharmacology , Smoking/adverse effects , Smoking/epidemiology , Treatment Outcome
13.
Sci Rep ; 4: 7268, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25434695

ABSTRACT

The predictive power of age at diagnosis and smoking history for ALK rearrangements and EGFR mutations in non-small-cell lung cancer (NSCLC) remains not fully understood. In this cross-sectional study, 1160 NSCLC patients were prospectively enrolled and genotyped for EML4-ALK rearrangements and EGFR mutations. Multivariate logistic regression analysis was performed to explore the association between clinicopathological features and these two genetic aberrations. Receiver operating characteristic (ROC) curves methodology was applied to evaluate the predictive value. We showed that younger age at diagnosis was the only independent variable associated with EML4-ALK rearrangements (odds ratio (OR) per 5 years' increment, 0.68; p < 0.001), while lower tobacco exposure (OR per 5 pack-years' increment, 0.88; p < 0.001), adenocarcinoma (OR, 6.61; p < 0.001), and moderate to high differentiation (OR, 2.05; p < 0.001) were independently associated with EGFR mutations. Age at diagnosis was a very strong predictor of ALK rearrangements but poorly predicted EGFR mutations, while smoking pack-years may predict the presence of EGFR mutations and ALK rearrangements but with rather limited power. These findings should assist clinicians in assessing the likelihood of EML4-ALK rearrangements and EGFR mutations and understanding their biological implications in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/genetics , Genes, erbB-1/genetics , Lung Neoplasms/epidemiology , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adult , Age Distribution , Aged , Aged, 80 and over , Anaplastic Lymphoma Kinase , China/epidemiology , Comorbidity , Cross-Sectional Studies , Female , Gene Rearrangement/genetics , Genetic Markers/genetics , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Prevalence , Risk Factors , Smoking/epidemiology , Smoking/genetics , Young Adult
14.
Oncotarget ; 5(23): 12189-202, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25361008

ABSTRACT

PD-L1 expression is a feature of Epstein-Barr virus (EBV) associated malignancies such as nasopharyngeal carcinoma (NPC). Here, we found that EBV-induced latent membrane protein 1 (LMP1) and IFN-γ pathways cooperate to regulate programmed cell death protein 1 ligand (PD-L1). Expression of PD-L1 was higher in EBV positive NPC cell lines compared with EBV negative cell lines. PD-L1 expression could be increased by exogenous and endogenous induction of LMP1 induced PD-L1. In agreement, expression of PD-L1 was suppressed by knocking down LMP1 in EBV positive cell lines. We further demonstrated that LMP1 up-regulated PD-L1 through STAT3, AP-1, and NF-κB pathways. Besides, IFN-γ was independent of but synergetic with LMP1 in up-regulating PD-L1 in NPC. Furthermore, we showed that PD-L1 was associated with worse disease-free survival in NPC patients. These results imply that blocking both the LMP1 oncogenic pathway and PD-1/PD-L1 checkpoints may be a promising therapeutic approach for EBV positive NPC patients.


Subject(s)
B7-H1 Antigen/biosynthesis , Epstein-Barr Virus Infections/metabolism , Interferon-gamma/metabolism , Nasopharyngeal Neoplasms/virology , Viral Matrix Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/physiology , Gene Expression Regulation, Viral/physiology , Herpesvirus 4, Human , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/mortality , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transfection , Up-Regulation , Young Adult
15.
Drug Des Devel Ther ; 8: 1083-7, 2014.
Article in English | MEDLINE | ID: mdl-25152614

ABSTRACT

PURPOSE: This retrospective study evaluates the efficacy and safety of S-1 chemotherapy for recurrent and metastatic nasopharyngeal carcinoma patients after failure of platinum-based chemotherapy. PATIENTS AND METHODS: Thirty-nine patients with recurrent and metastatic nasopharyngeal carcinoma who failed previous platinum-based chemotherapy received oral S-1 chemotherapy (twice daily from day 1 to 14) every 3 weeks. The dose of S-1 was determined according to the body surface area (BSA): 40 mg twice a day for BSA <1.25 m(2); 50 mg twice a day for 1.25 m(2) ≤BSA<1.5 m(2); and 60 mg twice a day for BSA ≥1.5 m(2). RESULTS: Treatment was well tolerated. Most adverse events were mild. Grade 3 hematological toxicity occurred in 7.7%. There was one complete response (2.6%) and 12 partial responses (30.7%), giving an overall response rate of 33.3% (95% CI [confidence interval], 21.7-50.8). Median time-to-progression was 5.6 months, and median survival was 13.9 months. One- and 2-year survival rates were 60% and 26%, respectively. CONCLUSION: S-1 monotherapy is considered a safe and effective treatment option for recurrent and metastatic nasopharyngeal carcinoma patients after failure of platinum-based chemotherapy.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Nasopharyngeal Neoplasms/drug therapy , Oxonic Acid/therapeutic use , Tegafur/therapeutic use , Adult , Aged , Antimetabolites, Antineoplastic/adverse effects , Carcinoma , Disease Progression , Drug Combinations , Female , Humans , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Neoplasm Metastasis , Neoplasm Recurrence, Local , Oxonic Acid/adverse effects , Platinum Compounds/therapeutic use , Retrospective Studies , Survival Rate , Tegafur/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL