Enhancer of zeste 2 (EZH2) governs gene reprogramming during cardiac hypertrophy through epigenetic remodeling, a process regulated by numerous non-coding RNAs (ncRNAs). However, the dynamic interaction between EZH2 and ncRNAs upon hypertrophic stimulation remains elusive. Here we performed an unbiased profiling for EZH2-associated ncRNAs in mouse hearts treated with Angiotensin II (AngII) at different time points (0, 4, and 24 h). The interactions between EZH2 and long ncRNAs (lncRNAs), Chaer, Mirt1, Hotair, and H19, were validated by PCR. RIP-seq analysis identified a total of 126 ncRNAs to be significantly associated with EZH2. These ncRNAs covers all five categories including intergenic, antisense, intron-related, promoter-related and both antisense and promoter-related. According to their changing patterns after AngII treatment, these ncRNAs were clustered into four groups, constantly enhanced, transiently enhanced, constantly suppressed and transiently suppressed. Structural prediction showed that EZH2 bound to hairpin motifs in ncRNAs including snoRNAs. Interaction strength prediction and RNA pull-down assay confirmed the direct interaction between EZH2 and Snora33. Interestingly, two antisense lncRNAs of Malat1, Gm20417, and Gm37376, displayed different binding patterns from their host gene after AngII treatment, suggesting a crucial role of this genomic locus in modulating EZH2 behavior. Our findings reveal the profile of EZH2-associated ncRNAs upon hypertrophic stimulation, and imply a dynamic regulation of EZH2 function in cardiac hypertrophy.
Osteosarcoma is the most common malignant bone tumor that seriously threatens the lives of teenagers and children. Zinc finger (ZNF) protein genes encode the largest transcription factor family in the human genome. Aberrant expressions of ZNF protein genes widely occur in osteosarcoma, and these genes are therefore attractive biomarker candidates for prognosis prediction. In this study, we conducted a comprehensive analysis of ZNF protein genes in osteosarcoma and identified prognosis-related ZNF protein genes. Then, we constructed a prognostic signature based on seven prognosis-related ZNF protein genes and stratified patients into high- and low-risk groups. The seven genes included MKRN3, ZNF71, ZNF438, ZNF597, ATMIN, ZNF692, and ZNF525. After validation of the prognostic signature in internal and external cohorts, we constructed a nomogram including clinical features such as sex and age and the relative risk score based on the risk signature. Functional enrichment analysis of the risk-related differentially expressed genes revealed that the prognostic signature was closely associated with immune-related biological processes and signaling pathways. Moreover, we found significant differences between the high- and low-risk groups for the scores of diverse immune cell subpopulations, including CD8+ T cells, neutrophils, Th1 cells, and TILs. Regarding immune function, APC co-inhibition, HLA, inflammation promotion, para-inflammation, T-cell co-inhibition, and the type I IFN response were significantly different between the high- and low-risk groups. Of the seven ZNF protein genes, lower expressions of ATMIN, MKRN3, ZNF71, ZNF438, and ZNF597 were correlated with a high risk, while higher expressions of ZNF525 and ZNF692 were associated with a high risk. The Kaplan-Meier survival analysis suggested that lower expressions of ATMIN, ZNF438, and ZNF597 and the higher expression of ZNF692 were associated with worse overall survival in osteosarcoma. In conclusion, our ZNF protein gene-based signature was a novel and clinically useful prognostic biomarker for osteosarcoma patients.
Background: Although growth advantage of certain clones would ultimately translate into a clinically visible disease progression, radiological imaging does not reflect clonal evolution at molecular level. Circulating tumor DNA (ctDNA), validated as a tool for mutation detection in lung cancer, could reflect dynamic molecular changes. We evaluated the utility of ctDNA as a predictive and a prognostic marker in disease monitoring of advanced non-small cell lung cancer (NSCLC) patients. Methods: This is a multicenter prospective cohort study. We performed capture-based ultra-deep sequencing on longitudinal plasma samples utilizing a panel consisting of 168 NSCLC-related genes on 949 advanced NSCLC patients with driver mutations to monitor treatment responses and disease progression. The correlations between ctDNA and progression-free survival (PFS)/overall survival (OS) were performed on 248 patients undergoing various treatments with the minimum of 2 ctDNA tests. Results: The results of this study revealed that higher ctDNA abundance (P=0.012) and mutation count (P=8.5×10−4) at baseline are associated with shorter OS. We also found that patients with ctDNA clearance, not just driver mutation clearance, at any point during the course of treatment were associated with longer PFS (P=2.2×10−16, HR 0.28) and OS (P=4.5×10−6, HR 0.19) regardless of type of treatment and evaluation schedule. Conclusions: This prospective real-world study shows that ctDNA clearance during treatment may serve as predictive and prognostic marker across a wide spectrum of treatment regimens.
Blue lasers are becoming more widely used in the diagnosis and treatment of bladder cancer; however, their photobiomodulation effects on bladder cancer cells remains unclear. The purpose of the current study was to explore the photobiomodulation effect of blue laser irradiation on bladder cancer progression and the associated mechanisms. The human uroepithelial cell line SV-HUC-1 and human bladder cancer cell lines T24 and EJ were exposed to blue laser irradiation (450 nm) at various energy densities, and cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and the levels of the proteins associated with the MAPK pathway proteins were determined. A significant decrease in cell viability was observed in a density-dependent manner after blue laser irradiation at > 4 J/cm 2 in both bladder cancer cell lines. However, the blue laser did not reduce cell viability in SV-HUC-1 cells until the energy density exceeded 16 J/cm 2 . Meanwhile, Ki67 levels, reflecting cell proliferation and senescence, were also significantly decreased after blue laser irradiation at 4 J/cm 2 and 8 J/cm 2 in the absence of cell cycle arrest. Moreover, blue laser irradiation at 4 J/cm 2 and 8 J/cm 2 caused a reduction in cell migration and invasion and also reduced the expression levels of MMP-2, MMP-9, Snail, N-cadherin, phospho-MEK and phospho-ERK, and elevated the expression levels of E-cadherin. Meanwhile ERK activator(tBHQ) significantly reversed the irradiation-induced suppression of proliferation, migration and invasion in T24 and EJ cell lines. The present study showed that blue laser irradiation inhibited bladder cancer proliferation in a density-dependent manner and inhibited bladder cancer progression by suppressing migration, invasion, and the EMT process in T24 and EJ cell lines. This inhibition was possibly mediated via suppression of the MAPK/MEK/ERK pathway. Thus, the use of a low-energy blue laser in the diagnosis and treatment of bladder cancer is possibly safe and may have an anti-tumor effect.
The homeobox (HOX) family genes have been linked to multiple types of tumors, while their effect on malignant behaviors of clear cell renal cell carcinoma (ccRCC) and clinical significance remains largely unknown. Here, we comprehensively analyzed the expression profiles and prognostic value of HOX genes in ccRCC using datasets from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. We developed a prognostic signature comprising eight HOX genes ( HOXB1 , HOXA7 , HOXB5 , HOXD8 , HOXD9 , HOXB9 , HOXA9 , and HOXA11 ) for overall survival prediction in ccRCC and it allowed patients to be subdivided into high- and low-risk groups. Kaplan-Meier survival analysis in all the internal and external cohorts revealed significant difference in clinical outcome of patients in different risk groups, indicating the satisfactory predictive power of the signature. Additionally, we constructed a prognostic nomogram by integrating signature-derived risk score and clinical factors such as gender, age, T and M status, which might be helpful for clinical decision-making and designing tailored management schedules. Immunological analysis revealed that the regulatory T cells (Tregs) infiltrated differently between the two subgroups in both TCGA and ICGC cohorts. ssGSEA method showed that the enrichment scores for mast cells were significantly lower in high-risk group compared with the low-risk group, which was consistent in both TCGA and ICGC cohorts. As for the related immune function, the enrichment scores of APC co-inhibition, para-inflammation, and type II IFN response were consistently lower in high-risk group in both cohorts. Of the eight HOX genes, the mRNA and protein levels of HOXD8 were downregulated in ccRCC than that in normal tissues, and decreased expression of HOXD8 was associated with increased tumor grade and stage, and lymph node metastasis. Survival analysis revealed that lower expression of HOXD8 predicted worse overall survival in ccRCC. In conclusion, our HOX gene-based signature was a favorable indicator to predict the prognosis of ccRCC cases and associated with immune cell infiltration. HOXD8 might be a tumor suppressor gene in ccRCC and a potential predictor of tumor progression.
Contrast-induced nephropathy (CIN) is a common adverse event in the diagnosis and treatment of coronary intervention. The current study investigated the predictive effect of preoperative fibrinogen (FIB) combined with antithrombin III (AT-III) on CIN following percutaneous coronary intervention (PCI). A total of 394 patients who underwent PCI between October 2018 and May 2019 were selected for the present study. Pre-procedural FIB levels and AT-III activity were measured before PCI. CIN was defined as a 0.5 mg/dl or 25% increase in serum creatinine levels 48 to 72 h after exposure to a radiocontrast agent. Patients were classified into CIN and non-CIN groups. CIN occurred in 48 (12.2%) patients. The serum FIB levels were significantly higher in patients who developed CIN compared with those who did not develop CIN. In addition, AT-III levels ≤89.5% were associated with higher rates of CIN. Logistical regression analysis showed that high FIB, and low AT-III and albumin levels were high-risk factors associated with CIN. For FIB, the area under the receiver operating characteristic curve (AUC) for predicting CIN was 0.653. The optimal cut-off value was 3.48 g/l with a sensitivity of 45.8% and a specificity of 79.7% [95% confidence interval (CI): 0.603-0.701; P=0.0002)]. For AT-III, the AUC was 0.711, and the optimal cut-off value was 89.5%, with a sensitivity of 81.3% and specificity of 58.2% (95% CI: 0.659-0.758; P<0.0001). When combining FIB and AT-III, the AUC was 0.747. The optimal cut-off value was 0.090424, with a diagnostic sensitivity of 93.8% and specificity of 46.6% (95% CI: 0.697-0.792; P<0.0001). The results showed that FIB combined with AT-III resulted in improved predictive accuracy of CIN (FIB vs. AT-III, AUC=0.653 vs. 0.711, P=0.292; FIB vs. FIB + AT-III, AUC=0.653 vs. 0.747, P=0.012; AT-III vs. FIB + AT-III, AUC=0.711 vs. 0.747, P=0.138). Pre-procedural levels of FIB, AT-III and albumin were independently associated with an increased risk of CIN. Furthermore, the results suggested that the combination of FIB and AT-III was a better predictor of CIN after PCI.
Abstract Background: Entinostat is a novel oral benzamide HDACi characterized by its selectivity for class 1 HDACs as well as a unique pharmacokinetic (PK) and pharmacodynamic (PD) profile. Previous results have demonstrated the ability of entinostat to synergize in vitro and in vivo with the small molecule EGFR inhibitors, gefitinib and erlotinib as well as EGFR directed antibody, cetuximab to inhibit growth of EGFRi resistant NSCLC cells. The mechanism for the combined efficacy may involve a) targeting of EGFR gene and protein expression b) down-regulation of EGFR-mediated signal transduction pathway c) alteration of the tumor phenotype to re-sensitize cancer cells to EGFRi. Methods: The growth inhibitory effect of the combination of erlotinib and entinostat in H1975 and H1933 cells was determined using a 5-day 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. In vivo efficacy of entinostat was tested in athymic nude mice bearing NSCLC xenografts, A549 and H460. Combined treatment of entinostat with cetuximab was studied in H460 xenografts Results: We have extended our previous work examining the effect of entinostat on EGFRi resistance through testing of entinostat combined with erlotinib in cell line models of acquired resistance and evaluating the combination of entinostat with cetuximab in vivo in a xenograft model. Our results demonstrate that the combination of entinostat and erlotinib has enhanced activity in H1975 NSCLC cells which harbor the EGFRi T790M mutation. In addition the combination is more effective in H1933 NSCLC cells which have a met amplification. To further characterize the potential mechanism of action by which entinostat sensitizes NSCLC cells to EGFRi we have also tested entinostat in vivo in A549 and H460 xenograft models. Entinostat exhibits a dose response inhibition of tumor growth in both models and analysis of molecular markers is ongoing. In our previous experiments examining impact of entinostat on EGFRi resistance we reported on the combined efficacy of entinostat and cetuximab in the A549 NSCLC xenograft model. We have followed up on those initial findings through testing in a second xenograft model (H460) and will report those results. Conclusions: Entinostat enhances the effect of EGFR inhibitors in models of acquired EGFR resistance. The results from these experiments support the rationale of combining HDACi and EGFRi (small molecules as well as antibodies) in clinical studies in NSCLC. Ongoing clinical trials testing these hypothesis include ENCORE-401, a randomized, placebo controlled phase 2 study of erlotinib +/− entinostat in NSCLC patients and a pilot study, ENCORE-403 which was initiated to test the effect of entinostat on resistance to the EGFRi erlotinib in NSCLC. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 5439.
// Di Zheng 1, * , Min Hu 2, * , Yu Bai 2 , Xuehua Zhu 2 , Xuesong Lu 3 , Chunyan Wu 4 , Jiying Wang 1 , Li Liu 1 , Zheng Wang 3 , Jian Ni 1 , Zhenfan Yang 2 and Jianfang Xu 1 1 Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, China 2 IMED Asia, AstraZeneca, Shanghai, China 3 Research and Development Information, AstraZeneca, Shanghai, China 4 Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School, Shanghai, China * These authors contributed equally to this work Correspondence to: Jianfang Xu, email: xujianfang63@aliyun.com Zhenfan Yang, email: Pamela.Yang@astrazeneca.com Keywords: EGFR, NSCLC, osimertinib, drug resistance, G796D Received: March 08, 2017 Accepted: May 04, 2017 Published: May 16, 2017 ABSTRACT Osimertinib is an effective third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) approved in multiple countries and regions for patients with EGFR T790M mutation-positive non-small cell lung cancer (NSCLC). Despite impressive initial tumor responses, development of drug resistance ultimately limits the benefit of this compound. Mechanisms of resistance to osimertinib are just beginning to emerge, such as EGFR C797S and L718Q mutations, BRAF V600E and PIK3CA E545K mutations, as well as ERBB2 and MET amplification. However, a comprehensive view is still missing. In this study, we presented the first case of Chinese NSCLC patient who developed resistance to osimertinib, and discovered de novo EGFR G796D mutation as a potential mechanism. Our findings provided insights into mechanisms of resistance to osimertinib and highlighted tumor heterogeneity and clonal evolution during the development of drug resistance.
Literatures regarding the prevalence and clinical significance of compound EGFR mutations are limited. Until now, none of retrospective or prospective research has focused on in cis compound EGFR mutations except case reports. In this study, we screened a cohort of 3,000 treatment-naïve Chinese advanced NSCLC patients using capture-based ultra-deep targeted sequencing to evaluate the prevalence of EGFR in cis compound mutations and the efficacy of EGFR-TKI in this population. Of the 3,000 patients screened, 1,266 (42.2%) had EGFR mutation; among them, 15 patients (1.2%) harboring in cis compound EGFR mutations, with 10 patients carrying EGFR L858R in combination with a rare mutation and five patients carrying two rare EGFR mutations. No patient with EGFR 19del was observed. Interestingly, no in trans configuration was identified in this cohort. All of the patients harboring in cis compound EGFR mutations were non-smokers, histologically diagnosed with adenocarcinoma and received first-generation EGFR-TKI. Furthermore, our data also revealed that patients with in cis compound EGFR mutations exhibit comparable PFS to first generation EGFR-TKI comparing to patients with single activating EGFR mutation. This observation was further supported by in silico molecular modeling analyses which demonstrated in cis compound mutations do not alter the ATP-binding pocket of EGFR, thus having no effect on the interaction between gefitinib and EGFR.