Virtual Library

Start Your Search

L.(. Wang

Moderator of

  • +

    MINI 34 - RNA and miRNA (ID 162)

    • Event: WCLC 2015
    • Type: Mini Oral
    • Track: Biology, Pathology, and Molecular Testing
    • Presentations: 15
    • +

      MINI34.01 - MicroRNA Profiling of a Familial Primary Pulmonary Enteric Adenocarcinoma (ID 397)

      18:30 - 20:00  |  Author(s): I. Garajová, N. Funel, M. Fiorentino, V. Agostini, M. Ferracin, M. Negrini, G.L. Frassineti, C. Rolfo, G. Biasco, E. Giovannetti

      • Abstract
      • Presentation
      • Slides

      Background:
      Primary pulmonary enteric adenocarcinoma (PEAC) is defined as a pulmonary adenocarcinoma with a predominant component (>50%) of intestinal differentiation and tumor cells positive for at least one intestinal marker. Due to its peculiarity and rarity, the optimal clinical management of PEAC patients remains unclear. A total of thirty cases have been described in literature to date, though familial aggregation has never been reported before. This is the first study describing the histological and molecular characterization of the PEAC from a patient with a family member affected by the same tumor.

      Methods:
      We evaluated the molecular characteristics of proband’s PEAC applying a previously validated 47-miRNA signature and applying the predictive method to estimate tissue-of-origin probabilities. Immunohistochemical (IHC) staining (TTF-1, Napsin A, CDX2, cytokeratonins, mucins) and mutational analyses (EGFR, K-RAS, ALK) were performed on formalin-fixed, paraffin-embedded tissue of a patient affected by PEAC.

      Results:
      The familial aggregation of PEAC was associated with similar clinicopathological features (age at diagnosis, smoking-habit, tumor localization, multiple colon polyps), histologic findings (IHC staining negative for TTF-1 and positive for CDX2) and genetic findings (K-RAS(Gly12Asp) mutation, but no EGFR/ALK aberrations). MiRNA profiling revealed main similarities with NSCLC (75.98%), and partial overlap with pancreatic cancer (PDAC, 23.34%), but not with colorectal cancer (less than 0.5%). Notably, this PEAC shares with PDAC key miRNAs associated with tumor aggressiveness (mir-31/-126/-506/-508-3p/-514). Figure 1



      Conclusion:
      In conclusion, we described, for the first time, PEACs in two members of the same family, associated with clinicopathological features. Moreover, miRNA profiling resembled mostly NSCLC, with a partial overlap with PDAC’s pattern that could explain the aggressive behavior compared to most NSCLC and guide future tailored-therapy approaches.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.02 - Prognostic Impact of Bcl-2 Depends on Tumor Histology and Expression of MALAT-1 lncRNA in NSCLC (ID 3221)

      18:30 - 20:00  |  Author(s): L.H. Schmidt, D. Goerlich, C. Rohde, M. Schuler, A. Huge, R. Voss, A. Faldum, C. Mueller-Tidow, W.E. Berdel, R. Wiewrodt

      • Abstract
      • Slides

      Background:
      Apoptosis is a crucial pathway in tumor growth and metastatic development. Apoptotic proteins regulate the underlying molecular cascades and are thought to modulate the tumor response to chemotherapy and radiation. However, the prognostic value of the expression of apoptosis regulators in localized non-small-cell lung cancer (NSCLC) is still unclear.

      Methods:
      We investigated the protein expression of apoptosis regulators Bcl-2, Bcl-xl, Mcl-1, and pp32/PHAPI, and the expression of the lncRNA MALAT-1 in tumor samples from 383 NSCLC patients (median age: 65.6 years; 77.5% male; paraffin embedded tissue microarrays) For statistical analysis correlation tests, Log rank tests and Cox proportional hazard models were applied.

      Results:
      Tumor histology was significantly associated with the expression of Bcl-2, Bcl-xl and Mcl-1 (all p<0.001). Among the tested apoptotic markers only Bcl-2 demonstrated prognostic impact (HR=0.64, p=0.012). For NSCLC patients with non-adenocarcinoma histology, Bcl-2 expression was associated with increased overall survival (p=0.036). Besides tumor histology, prognostic impact of Bcl-2 was also found to depend on MALAT-1 lncRNA expression. Gene expression analysis of A549 adenocarcinoma cells with differential MALAT-1 lncRNA expression demonstrated an influence on the expression of Bcl-2 and its interacting proteins.

      Conclusion:
      Bcl-2 expression was specifically associated with superior prognosis in localized NSCLC. An interaction of Bcl-2 with MALAT-1 lncRNA expression was revealed, which merits further investigation for risk prediction in resectable NSCLC patients.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.03 - Novel microRNA Prognostic Signature in Malignant Pleural Mesothelioma (ID 2988)

      18:30 - 20:00  |  Author(s): F. Grossi, C. Genova, A. Truini, S. Coco, E. Nadal, M.G. Dal Bello, I. Vanni, A. Alama, E. Rijavec, G. Barletta, F. Biello, D. Beer

      • Abstract
      • Presentation
      • Slides

      Background:
      Malignant pleural mesothelioma (MPM) is an aggressive tumor mainly associated with asbestos exposure. MPM patients have a poor outcome (median overall survival (mOS) <1 year), therefore novel therapeutic approaches are needed. MiRNA have been demonstrated to have a role in tumorigenesis and progression in MPM. This study aimed to identify a miRNA signature associated with poor prognosis.

      Methods:
      We identified 26 un-resected MPM patients split as follows: 11 long survivors (LS) OS>3 years and 15 short survivors (SS) OS<1 year. MiRNA expression in 26 FFPE biopsy and 3 normal pleura (NP) was evaluated using Agilent Human miRNA Microarray platform including 2006 miRNA. Expression data were normalized by GeneSpring software (v.12.6). Class-comparison analysis between MPM/NP and SS/LS was performed using a t-test adjusted for multiple comparisons using Benjamini-Hochberg. OS curves were estimated using the Kaplan-Meier method and compared with the log-rank test. In silico validation was performed using miRseq data from TCGA portal based upon 16 patients (LS: 8; SS: 8). Candidate miRNA were assessed by univariate analysis using Kaplan-Meier method and median as cutoff.

      Results:
      Patients’ characteristics: median age 67 years (53-77); 81% males, 19% females; 73% epithelioid histotype, 12% sarcomatoid, 12% biphasic and 1 unspecified MPM. No differences in age, gender and histotype were observed between LS and SS. By class-comparison analysis, 30 miRNAs were significantly up-regulated and 11 down-regulated in MPM vs NP (adjusted p-value <0.05). Fourteen miRNAs were significantly associated with outcome, in the univariate survival analysis and differentially expressed in MPM. A miRNA signature, based on the top 6 prognostic miRNAs (unfavorable, miR-1224; favorable, miR-99a, miR-125b, let-7b, let-7c and let-7i) classified patients into low- or high-risk. High-risk patients showed a significantly shorter median OS (4.1 months, 95% CI 2.2-5.9) as compared with low-risk patients (median not reached, Log-rank p<0.001). In silico validation analysis confirmed that low expression of mir-99a, miR-125b and let-7c was associated with shorter OS. Relevant pathways, such as PI3K/AKT, WNT were associated with these top miRNAs by pathway analysis.

      Conclusion:
      A prognostic miRNA signature was identified by profiling a cohort of un-resected MPM, underlying the clinical potential of miRNA as predictors of survival. An additional validation in a larger independent cohort of MPM is ongoing.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.04 - Serum MicroRNA-139-5p Is a Biomarker of Bone Metastasis in Lung Cancer Patient (ID 884)

      18:30 - 20:00  |  Author(s): S. Xu, Y. Wu, R. Liu, H. Fan, S. Wei, G. Chen, J. Chen

      • Abstract
      • Slides

      Background:
      Lung cancer is the leading cause of cancer mortality worldwide. About 30–40% of patients with lung cancer developed bone metastasis during the course of their disease, with the median survival time of 7 months approximately. Bone lesion is characterized by the imbalance of osteoblastic and osteoclastic activity induced by the tumor cells. Mesenchymal stem cells (MSCs) are the progenitor cells of osteoblast cells, and therefore it is interesting to investigate whether and how MSCs have biological alterations in the tumor microenvironment of lung cancer patients with bone lesion.

      Methods:
      We tested miR-139-5p and Notch1 expression during MSC osteogenic differentiation, and validated the effect of miR-139-5p in MSC osteogenesis by transfection of miR-139-5p mimic and inhibitor. We also collected blood samples of healthy donors and lung cancer with bone metastasis, and tested serum miR-139-5p expression.

      Results:
      We demonstrated that during MSC osteogenic differentiation in vitro, the expression of miR-139-5p increased significantly while Notch1 and its downstream factors decreased. By gain and loss of function studies we showed that miR-139-5p could positively regulate MSC osteogenic differentiation. And luciferase and western blot assays confirmed that miR-139-5p targeted Notch1 expression directly. Moreover, Notch1 knockdown by siRNA could no longer confer miR-139-5p induced MSC osteogenic differentiation. Lung cancer cell A549 and L9981 secreted factors upregulated miR-139-5p expression and meanwhile downregulated Notch1 expression in MSC, as well as impaired ALP activity, the early osteogenic marker of MSCs. More importantly, we observed that serum miR-139-5p was significantly lower in lung cancer patients with lytic bone lesion compared to healthy donors.

      Conclusion:
      We demonstrate for the first time, that miR-139-5p could regulate osteogenic differentiation of MSC by targeting Notch1 mediated signalling pathway. Lung cancer cells could decrease miR-139-5p expression and inhibit MSC osteogenic potential. Importantly, serum miR-139-5p is significantly lower in lung cancer patients with lytic bone lesion compared to healthy donors. Therefore, miR-139-5p might be a biomarker of bone metastasis in lung cancer patients and treatment with miR-139-5p mimic might be an interesting strategy to restore the impaired MSC osteogenic differentiation and to control bone disease in lung cancer patients with bone lesion.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.05 - Discussant for MINI34.01, MINI34.02, MINI34.03, MINI34.04 (ID 3433)

      18:30 - 20:00  |  Author(s): C.J. Rivard

      • Abstract
      • Presentation

      Abstract not provided

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

    • +

      MINI34.06 - MicroRNA 10b, 27a and 27b Are Upregulated in Lung Cancer Cell with Epidermal Growth Factor (EGFR) Mutation Resistant to EGFR TKI (ID 2950)

      18:30 - 20:00  |  Author(s): H. Montes, E.B. Reyes, J. Kevern, P.J. Van Veldhuizen, C.H. Huang

      • Abstract

      Background:
      EGFR Tyrosine Kinase Inhibitor (TKI) is now standard of care in lung cancer patients with EGFR mutation. Invariably, these patients develop resistance and would require treatment with another EGFR TKI or chemotherapy. The gate keeper mutation T790M is responsible for about 50% of resistance cases. There are other mechanisms that could confer resistance in these patients. High expression of microRNA (mir) 10 is associated with worse prognosis in resected lung cancer patients with EGFR mutation. The mir 27a and 27b is associated with increased expression of c-met which is a potential mechanism of resistance to EGFR. We explored the expression of mir 10b and 27a and 27b in lung cancer cell lines with EGFR mutation that were resistant to Erlotinib.

      Methods:
      Lung cancer cell lines with EGFR mutation CRL-2868 and CRL 2871 were treated with 5 nM of erlotinib for 24hours and 72 hours. The erlotinib resistant cells were then harvested and then microRNA profiling was done by real time PCR. HTB177 cell line without EGFR mutation was used as control.

      Results:
      We observed an increased expression of mir 10b, mir27a and mir27b in CRL-2871 compared to control HTB 177 cells. Mir 27b is upregulated in both cell lines. The increase expression of mir 10b and mir 27a were higher in CRL-2871 than the control cells 9 fold and 8 folds respectively. The mir 27b was increased 300 folds in the CRL 2868 compared with control.

      Conclusion:
      We observed upregulation of mir 10b, 27a and 27b in lung cancer cells lines with EGFR mutation that were resistant to Erlotinib treatment. This suggests an epigenetic mechanism of resistance other than the T790M mutation. Further research in patients with EGFR mutation resistance to EGFR TKI should be done to confirm this finding.

    • +

      MINI34.07 - A Novel microRNA Signature Associated with Cisplatin Resistance in NSCLC (ID 2709)

      18:30 - 20:00  |  Author(s): L. Mac Donagh, S.G. Gray, V. Young, R. Ryan, S. Nicholson, N. Leonard, K.J. O'Byrne, S.P. Finn, S. Cuffe, M.P. Barr

      • Abstract
      • Presentation
      • Slides

      Background:
      MicroRNAs (miRNAs) are an abundant class of small non-coding RNAs that range in size from 19 to 25 nucleotides. Alteration in miRNA expression can cause them to act as either tumour suppressor or oncogenes. They have also been shown to regulate a number of processes involved in tumour biology such as metastasis, invasion and angiogenesis. More recently, miRNAs have been linked to chemo- and radio-resistance in many solid tumours, including lung cancer.

      Methods:
      An isogenic model of cisplatin resistance was established by chronically exposing a panel of NSCLC cell lines (MOR, H460, A549, SKMES-1, H1299) to cisplatin for 12 months, generating cisplatin resistant (CisR) sublines from their corresponding age-matched parental (PT) cells. MicroRNA expression profiling was carried out using 7th generation miRCURY LNA™ microRNA arrays consisting of 1,919 miRNAs (Exiqon). MicroRNAs that were significantly increased in CisR sublines were inhibited using antagomirs (Exiqon), while those that were significantly decreased were over-expressed using pre-miRs (Ambion). Functional studies examining clonogenic survival ability, proliferation (BrdU) and apoptosis (Annexin V/PI) were subsequently carried out in the presence or absence of cisplatin. To examine the translational relevance of these microRNAs, their expression was further examined in a cohort of pre-treatment matched normal and tumour lung tissues from NSCLC patients of different histological subtypes. Validation of this miRNA signature is currently being investigated in serum samples from this same cohort of patients and normal controls.

      Results:
      MicroRNA profiling analysis identified ten miRNAs which were significantly altered between parental and corresponding cisplatin resistant lung cancer cell lines. Validation of these miRNAs by real-time PCR (qPCR) identified a specific 5-miR signature that was significantly altered in CisR cells relative to their parental counterparts. Modification of these microRNAs altered the response of resistant cells to the cytotoxic effects of cisplatin and decreased the clonogenic survival of CisR cells when treated with increasing doses of cisplatin (0.1µM-10μM). Significant differential expression was found between normal and tumour tissues across each histological subtype, highlighting the potential use of these microRNAs as markers of response to cisplatin therapy in NSCLC patients. Three miRNAs (miR-A, B, C) belonging to the same family were significantly altered in tumour lung tissue of adenocarcinoma and squamous cell histology compared to matched normal lung tissue. MicroRNA-D expression was significantly altered in squamous cell carcinomas while miR-E was differentially expressed in adenocarcinomas only. Data relating to the expression of this novel signature in the circulation of our NSCLC patient cohort and normal controls will be presented at WCLC 2015.

      Conclusion:
      We have identified and validated a novel miRNA signature associated with cisplatin resistance in a panel of cisplatin resistant cell lines and in patient lung tumours. Genetic manipulation of these specific miRNAs in vitro altered the cisplatin resistant cell response to the cytotoxic effects of cisplatin chemotherapy. The data obtained from this study may provide a basis for the potential development of a companion diagnostic for lung cancer patients who are most likely to benefit, or not, from cisplatin chemotherapy.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.08 - MiR-33B Inhibit EMT Through Wnt/β-Catenin/ZEB1 Pathway in Lung Adenocarcinoma (ID 236)

      18:30 - 20:00  |  Author(s): J.J. Qu, M. Li, T.L. Cheng, J. An, L.M. Cao, H.P. Yang, C.P. Hu

      • Abstract
      • Presentation

      Background:
      Lung adenocarcinoma is one of the main pathological tissue types of lung small cell lung cancer (NSCLC). Tumor metastases are responsible for approximately 90% of lung adenocarcinoma-related death. The epithelial–mesenchymal transition (EMT) is regarded as a critical event during tumor metastasis. From our previous study, we found miR-33b might be involved in the EMT process of lung adenocarcinoma. However, the specific mechanism of miR-33b regulating EMT has not been established.

      Methods:
      Quantitative real time-PCR (qRT-PCR), in situ hybridization and immunohistochemistry were used to investigate expression of miR-33b and ZEB1 in paired lung adenocarcinoma tissues. MiR-33b target gene ZEB1 was investigated using luciferase reporter gene assays. Western blot, qRT-PCR, immunofluorescence were used to compare the expression levels of ZEB1, E-cad, Vim and β-catenin in A549 cells which were transfected miR-33b or miR-NC, anti-miR-33b or anti-miR-NC and in vivo. MTT was used to analysis growth curves of transfected cells with miR-33b or miR-NC, anti-miR-33b or anti-miR-NC, siRNA-NC or siRNA -ZEB1, siRNA-NC or siRNA-β-catenin. The transfected cells were subjected to Transwell migration and invasion assays.

      Results:
      Mean miR-33b levels were significantly lower in lung adenocarcinoma tissues compared with matched non-cancerous tissues (0.024± 0.028 vs 0.063 ± 0.074, P < 0.0001). The level of miR-33b in NSCLC was strongly correlated with lymph node metastasis (P < 0.0001). ZEB1 levels were significantly higher in lung adenocarcinoma tissues compared with matched non-cancerous tissues (0.447± 0.371vs0.084 ± 0.112, P <0.0001). The level of miR-33b in lung adenocarcinoma was negative correlated with ZEB1 (P < 0.0001,r=-0.6077). MiR-33b significantly inhibited EMT in lung adenocarcinoma metastasis in vitro and vivo (P < 0.05).MiR-33b directly targets the ZEB13[']UTR(P < 0.01).β-catenin was down regulation by overexpression of miR-33b (P < 0.01). MiR-33b overexpression and ZEB1 inhibition suppressed lung adenocarcinoma migration and invasion in vitro (P < 0.01).

      Conclusion:
      On the basis of our results, we propose that miR-33b suppress EMT in lung adenocarcinoma through direct targeting of ZEB1 in vitro and in vivo. Moreover, we found that Wnt/β-catenin/ZEB1 pathway regulated by miR-33b might involved in lung adenocarcinoma EMT. These findings provide new insights into the molecular functions of miR-33b as well asthe role of ZEB1 in lung adenocarcinoma metastasis.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

    • +

      MINI34.09 - The Long Non-Coding RNA AK126698 Modulates Wnt/β-Catenin Signaling Through FZD-8 Silencing in Non-Small-Cell Lung Cancer (ID 737)

      18:30 - 20:00  |  Author(s): X. Fu, H. Li

      • Abstract

      Background:
      Recent evidence indicates that long non-coding RNAs (lncRNAs) play a critical role in the regulation of cellular processes, such as differentiation, proliferation and metastasis. These lncRNAs are found to be dysregulated in a variety of cancers. In our previous study, we have demonstrated that lncRNA AK126698 regulated A549 cells cisplatin resistance partly through Wnt/β-catenin signaling. However, the clinical significance of lncRNA AK126698, and its’ molecular mechanisms of controlling cancer cell proliferation and migration are still unclear.

      Methods:
      Expression of lncRNA AK126698 was analyzed in 55 non-small cell lung cancer (NSCLC) tissues and 3 NSCLC cell lines using quantitative polymerase chain reaction (qPCR) assays. Gain and loss of function approaches were used to investigate the biological role of AK126698 in NSCLC cells. The effects of AK126698 on cell proliferation were evaluated by CCK-8 and colony formation assays. Apoptosis was evaluated by flow cytometry. Protein levels of AK126698 targets were determined by western blotting and fluorescent immunohistochemistry.

      Results:
      AK126698 expression was significantly decreased in NSCLC tumor tissues compared with normal lung tissues. Additionally, reduced AK126698 expression was associated with larger tumor size and advanced pathological staging of NSCLC patients. Ectopic expression of AK126698 impaired cell proliferation and migration and induced apoptosis in vitro. However, knockdown of AK126698 expression promoted cell migration and invasionin in vitro. In addition, the expression of FZD8 was inversely correlated with AK126698 in both NSCLC tissues and cell lines and that over-expression of AK126698 could significantly down-regulate the expression of gene downstream of FZD8, including β-catenin, CyclinD1 and c-Myc.

      Conclusion:
      The results of present study indicate that lncRNA AK126698 might serve as a suppressor that regulates the pathogenesis of NSCLC through Wnt/β-catenin signaling pathway. It may provide a new target for therapeutic intervention of NSCLC.

    • +

      MINI34.10 - Discussant for MINI34.06, MINI34.07, MINI34.08, MINI34.09 (ID 3434)

      18:30 - 20:00  |  Author(s): G. Reid

      • Abstract
      • Presentation

      Abstract not provided

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

    • +

      MINI34.11 - Identification and Functional Characterization of Non-Small Cell Lung Cancer-Associated Splice Variants and Splicing Factors (ID 2101)

      18:30 - 20:00  |  Author(s): F. De Miguel, M.J. Pajares, E. Martinez-Terroba, X. Morales, R.D. Sharma, A. Rouzaut, A. Rubio, L. Montuenga, R. Pio

      • Abstract
      • Presentation
      • Slides

      Background:
      Deregulation of alternative splicing has become a hallmark of cancer. In non-small cell lung cancer (NSCLC), the biological importance of splicing is evidenced by the identification of aberrant RNA transcripts associated with somatic mutations in genes encoding splicing factors. We have previously developed ExonPointer, an algorithm optimized to detect differential splicing cassette events from data obtained in microarrays containing probes in exons and junctions. Our present objective was to apply this technology for the identification and characterization of cancer-associated splice variants and splicing factors in NSCLC.

      Methods:
      We applied ExonPointer for the identification of differential splice forms in lung cancer tissues (8 adenocarcinomas, 13 squamous cell carcinomas and 1 large cell carcinoma) and matched normal lung. We validated the events by RT-PCR and used bioinformatics tools, such as DAVID and Ingenuity Pathway Analysis, for cluster enrichment analyses. siRNA knockdown of specific splice isoforms was used for functional analyses. Prognostic studies were performed by immunohistochemistry in 127 primary tissues from patients with NSCLC.

      Results:
      The validation rate for the top 20 differentially expressed splice events identified by ExonPointer was 70%. Gene cluster analyses using the first 250 events showed a significant enrichment of cancer-related clusters such as Cellular Growth and Proliferation, or Cell Death and Survival. Among the validated genes, we identified Extended synaptotagmin-2 (ESYT2). ESYT2 is a membrane protein that mediates fibroblast growth factor receptor-1 (FGFR1) endocytosis and actin dynamics. A significantly different pattern of ESYT2 alternative splicing was found in primary lung tumors as compared to normal lung tissue (p<0.001). In particular, an isoform containing an extra exon was overexpressed in cancer tissues, while the expression of the canonical isoform was decreased. We found a significant correlation between the splicing pattern of ESYT2 and the expression of FGFR1 in a panel of 43 lung cancer cell lines (r=-0.724, p<0.001). Using siRNA downregulation, we analyzed the implication of the ESYT2 isoforms in tumor biology and demonstrated a distinct role of the splice isoforms in actin and tubulin cytoskeleton organization. We also searched for splicing factors responsible for the splicing of ESYT2. We found that the ratio of ESYT2 isoforms correlated with the expression of the splicing factor QKI in lung cancer cell lines (r=-0.793, p<0.001). Moreover, in vitro downregulation of QKI markedly affected ESYT2 splicing. Interestingly, we found a significant enrichment of QKI targets in the list of differentially spliced genes identified by ExonPointer (p<0.001), suggesting that this factor is a critical regulator of splicing in lung cancer. Finally, we observed a significant downregulation of QKI expression in primary NSCLC compared to adjacent normal lung cancer cells (p<0.001), and an association between the nuclear expression of this factor and disease-free survival (HR=0.61; 95%CI=0.35-1.05) or overall survival (HR=0.44; 95%CI=0.21-0.94).

      Conclusion:
      Using a novel analytical tool we have identified new splicing variants with functional relevance in lung cancer. Moreover, changes in splicing events in lung primary tumors were found to be largely regulated by the splicing factor QKI, a potential tumor suppressor gene downregulated in NSCLC and associated with the prognosis of the disease.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.12 - Oncofetal miRNA Expression Inactivates Nuclear Factor I/B, a Critical Regulator of Lung Development and Lung Adenocarcinoma Pathogenesis (ID 3023)

      18:30 - 20:00  |  Author(s): D.D. Becker-Santos, K.L. Thu, L.A. Pikor, C.E. Macaulay, W.W. Lockwood, J.C. English, W.P. Robinson, I. Jurisica, S. Lam, W.L. Lam

      • Abstract
      • Slides

      Background:
      Fetal development shares many biological similarities with tumourigenesis such as high rates of cell proliferation and vasculature restructuring. Particularly in the lung, a number of genes and pathways involved in the development of this organ play important roles in the malignant transformation of adult lung cells. Despite these biological similarities, there is a paucity of information regarding how specific molecular regulators involved in fetal lung development become oncogenes in lung cancer. This is the case for micro RNAs (miRNAs), which have a pivotal role in regulating gene expression during organ development and tumourigenesis. Therefore, a better understanding of the human fetal lung miRNA-transcriptome has the potential to reveal key players in lung cancer development and progression.

      Methods:
      131 pairs of non-small cell lung cancer (NSCLC) tumour and adjacent non-malignant lung tissues and 15 human fetal lung tissue samples were profiled by miRNA-sequencing. Mann–Whitney U tests were performed with Benjamini-Hochberg multiple testing corrections to identify miRNAs abundantly expressed in fetal and tumour tissues but scarce in non-malignant adult lung (i.e. oncofetal miRNAs). To investigate protein-coding genes controlled by the oncofetal miRNAs identified, miRDIP was applied followed by gene reporter luciferase assay experiments. The assessment of associations between patient survival and mRNA expression of selected genes targeted by the oncofetal miRNAs was evaluated in multiple NSCLC cohorts (>1,400 tumour cases). To validate the protein expression of the most prominent gene regulated by the oncofetal miRNAs identified, immunohistochemical (IHC) analysis was performed on a lung adenocarcinoma (LUAD) tissue microarray (TMA).

      Results:
      We describe for the first time a comprehensive, unbiased characterization of miRNA expression in human fetal lung tissue by miRNA sequencing. Through comparison to a large cohort of NSCLC, we identified numerous miRNAs that recapitulate their fetal expression patterns in lung cancer. Strikingly, assessment of the genes potentially regulated by these oncofetal miRNAs led us to identify Nuclear Factor I/B (NFIB), a transcription factor essential for lung development, as being frequently targeted by oncofetal miRNAs. Concordantly, analysis of NFIB expression using RNA-sequencing data for multiple NSCLC cohorts revealed its frequent underexpression in tumours (>60%). Remarkably, low expression of NFIB was significantly associated with poorer survival in LUAD patients but not in squamous cell carcinoma patients, consistent with the functional role of NFIB in distal lung cell differentiation (i.e. cells that are the precursors of LUAD). Furthermore, an NFIB-related gene signature was identified in LUAD tumours and included several well-known lung differentiation markers (TTF-1, ABCA3, GPR116, SFTPB). Finally, the underexpression of NFIB protein was validated on a LUAD TMA, which also revealed that tumours presenting lower levels of this transcription factor are associated with higher grade, biologically more aggressive LUAD (invasive mucinous, micropapillary and solid subtypes).

      Conclusion:
      This work has revealed a prominent mechanism for the downregulation of a crucial gene for lung development, which we found to be associated with aggressive phenotypes of LUAD and consequently, poor patient survival. Elucidating the specific role of NFIB in lung cancer biology will likely lead to the identification of targetable tumour vulnerabilities.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.13 - Long Noncoding RNA BC070487 Represses ZNFX1 during Tobacco-Induced Lung Carcinogenesis (ID 2912)

      18:30 - 20:00  |  Author(s): S. Xi, J. Shan, H. Xu, Z. Xiao, Y. Xiong, S. Oyetunji1, L. Mercedes, M. Zhang, J.A. Hong, D.S. Schrump

      • Abstract
      • Presentation
      • Slides

      Background:
      Limited information exists regarding regulation of gene expression by long noncoding RNAs (lncRNAs) during initiation and progression of tobacco-associated lung cancers. In the present study, an in-vitro model system was used to examine the effects of cigarette smoke on lncRNA expression in human respiratory epithelia and lung cancer cells.

      Methods:
      Micro-array and qRT-PCR techniques were used to assess lncRNA and gene expression profiles in normal human small airway epithelial cells (SAEC) and immortalized human bronchial epithelial cells (HBEC) cultured in the presence or absence of cigarette smoke condensate (CSC). Quantitative chromatin immunoprecipitation (ChIP), methyl-DNA immunoprecipitation (MeDIP), and cross-link immunoprecipitation (CLIP) techniques were used to assess promoter occupancy, DNA methylation, and interaction of lncRNAs with target proteins. Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE) assays were used to identify a regulatory sequence for lncRNA BC070487.

      Results:
      Micro-array analysis demonstrated that under relevant exposure conditions, CSC consistently mediated a 2.5 fold increase in BC070487 expression, and 3.6 fold decrease in its immediate downstream target gene, ZNFX1, in SAEC. qRT-PCR experiments confirmed a 4-7 fold up-regulation of BC070487, with a 4-8 fold down-regulation of ZNFX1 in SAEC and HBEC, as well as Calu-6 and H841 lung cancer cells following CSC exposure. BC070487 expression correlated inversely with expression of ZNFX1 (BC070487: 1.38-10.31 fold up vs ZNFX1: 2.26-26.29 fold down) in primary lung cancers relative to adjacent normal lung parenchyma. Overexpression or depletion of BC070487 inhibited or enhanced expression of ZNFX1, respectively in normal respiratory epithelia and lung cancer cells. CSC as well as BC070487-mediated repression of ZNFX1 coincided with increased occupancy of EZH2, SUZ12, BMI1, and increased levels of H3K27Me3, with decreased H3K4Me3 within the ZNFX1 promoter region. CSC as well as BC070487 overexpression enhanced binding of BC070487 with EZH2, SUZ12 and BMI1 proteins in SAEC and Calu-6 cells. CSC exposure significantly increased DNA methylation in one of three CpG islands spanning the regulatory elements of ZNFX1. In addition, CSC enhanced binding of BC070487 with DNMT3A and DNMT3B with site-specific de novo DNA methylation of ZNFX1 in SAEC and Calu-6 cells. FAIRE assays identified a 800 bp regulatory sequence for BC070487; CSC-mediated activation of BC070487 coincided with increased levels of H3K4Me3, H3K27Ac, H3K36Me3, Sp1, and Ago proteins, with decreased levels of H3K27Me3 and H3K9Me3 within this regulatory region. miR-31, an oncomiR specifically induced by CSC, modulated the transcriptional activity of BC070487 via remodeling the distribution of histone modification marks (Up: H3K4Me3, H3K27Ac, H3K36Me3; Down: H3K27Me3 and H3K9Me3) in the regulatory region of BC070487. Overexpression or depletion of BC070487 increased or inhibited proliferation and invasion of lung cancer cells, and similarly modulated growth of SAEC and HBEC cells. In contrast, ZNFX1 overexpression or depletion decreased or enhanced growth of normal respiratory epithelial cells and lung cancer cells. BC070487 overexpression enhanced growth of lung cancer cells xenografts in athymic nude mice, while forced expression of ZNFX1 arrested growth of these xenografts.

      Conclusion:
      Collectively, these data suggest that CSC-mediated up-regulation of BC070487 suppresses ZNFX1 to promote pulmonary carcinogenesis.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.14 - Biological Role of Prognostic MicroRNAs (MiRNAs) in Squamous Lung Cancer Cell Lines (ID 1171)

      18:30 - 20:00  |  Author(s): M. Filipska, M. Skrzypski, J.J. Bigda, J. Jassem

      • Abstract
      • Presentation
      • Slides

      Background:
      The benefit from postoperative chemotherapy in non-small cell lung cancer (NSCLC) is modest and there are no reliable tools allowing for individual selection of high-risk patients for this management. We earlier demonstrated that overexpression of three miRNAs: miR-662, -192 and -192* may correlate with the risk of distant relapse in squamous cell lung cancer (SCC) patients undergoing pulmonary resection (Skrzypski et al. 2014, Br J Cancer). However, the role of these miRNAs in maintaining aggressive phenotype and/or chemoresistance of SCC is unknown. The aim of this study was to assess the biological role of three abovementioned miRNAs in SCC cells in vitro.

      Methods:
      In the search for appropriate in vitro model we screened by reverse transcription quantitative PCR 11 NSCLC cell lines (H1703, H520, H662, SK-MES-1, A549, H23, H1975, HCC 827, PC9, H460 and Calu-6) for miRNA expression profiles and analyzed their phenotypic features including migration, invasion and clonogenic potential. H520 and H1703 SCC cell lines were transfected with locked nucleic acid miRNA inhibitors. The sensitivity of transfected and wild type cells to cisplatin and etoposide was compared using cytotoxic MTT test. Soft agar colony formation assay was performed to assess the clonogenic potential of transfected vs. non-transfected cells.

      Results:
      Among the analyzed NSCLC cell lines, H520 cells showed natural overexpression of miR-662, -192 and -192*. These cells were resistant to both chemotherapeutics and exhibited an ability to form colonies in soft agar. The inhibition of miR-662 and miR-192 sensitized these cells to etoposide (p=0.004 and 0.016, respectively) but not to cisplatin. Similar results were obtained for H1703 cells (p=0.002 and 0.02, respectively). H520 cells treated with miR-192 and miR-662 inhibitors, compared to negative control, also demonstrated reduced number of colonies in soft agar (p<0.001).

      Conclusion:
      We developed and optimized a cellular model for in vitro studies of biological role of three prognostic miRNAs in SCC. Genes regulated by miR-662 and/or miR-192 may be involved in maintaining the chemoresistance and clonogenic potential of SCC, and these features may be inhibited in a miR-dependent specific manner. Further studies may elucidate predictive value of these genes and a possibility of their targeting with novel therapeutics.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

    • +

      MINI34.15 - Discussant for MINI34.11, MINI34.12, MINI34.13, MINI34.14 (ID 3435)

      18:30 - 20:00  |  Author(s): D. Beer

      • Abstract
      • Presentation
      • Slides

      Abstract not provided

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.



Author of

  • +

    P1.03 - Poster Session/ Treatment of Locoregional Disease – NSCLC (ID 212)

    • Event: WCLC 2015
    • Type: Poster
    • Track: Treatment of Locoregional Disease – NSCLC
    • Presentations: 1
    • +

      P1.03-020 - IMRT Improves Survival in Locally Advanced NSCLC (LA-NSCLC) Receiving Definitive Radiotherapy: A Population Based Time-Trend Analysis (ID 2264)

      09:30 - 17:00  |  Author(s): L.(. Wang

      • Abstract

      Background:
      Currently intensity-modulated radiotherapy (IMRT) is regarded as a promising but unproven therapy for locally advanced non-small cell lung cancer (LA-NSCLC). This study aimed to evaluate the impact of introducing IMRT in LA-NSCLC based on patients receiving definitive radiotherapy (RT) throughout an 11-year span from an academic cancer center.

      Methods:
      Patients treated with definitive RT (≥ 50Gy) between 2000 and 2010 were divided into three eras according to availability of IMRT: 2000 to 2003 (period A, no IMRT, IMRT rate 0%), 2004 to 2006 (period B, introduction of IMRT, IMRT rate 3.5%) and 2007 to 2010 (period C, full access to IMRT, IMRT rate 85.6%). Patients’ characteristics, treatment modality, survival and treatment related toxicities were compared between 3 periods.

      Results:
      A total of 946 patients were analyzed. Less smokers, more stage IIIA diseases and more patients receiving concurrent chemo-radiotherapy (CRT) were observed in period C. The median overall survival (OS), local-regional progression free survival (LRPFS), distant metastasis free survival (DMFS) and progression free survival (PFS) for the whole population, period A, B and C were 19.8 vs. 16.6 vs. 18.2 vs. 23.3 moths, 22.1 vs. 16.2 vs. 18.7 vs. 40.5 months, 20.7 vs. 17.1 vs. 17.0 vs. 33.1 months and 11.4 vs. 10.8 vs. 11.3 vs. 11.9 months, respectively. Accordingly, the 5-y OS, LRPFS, DMFS and PFS were 14.3% vs. 9.8% vs. 12.0% vs. 18.3%, 34.3% vs. 22.9% vs. 28.4% vs. 43.6%, 32.2% vs. 25.2% vs. 23.6% vs. 40.5% and 14.2% vs. 10.7% vs. 11.1% vs. 18.0%, respectively. All survival indexes significantly increased in period C (Figure 1). Multivariate analyses identified IMRT as the independently favorable indicators for all survival indexes. The incidence of radiation induced lung toxicity (RILT) significantly decreased in period C (32.2% vs. 24.9% vs. 12.8%, p < 0.001) whereas that of radiation induced esophagus toxicity (RIET) remained stable (29.4% vs. 39.0% vs. 33.1%, p = 0.064) throughout the overall study period. Figure 1



      Conclusion:
      IMRT was associated with improved tumor control, prolonged survival and decreased RILT, independent of treatment modality and radiation dose.

  • +

    P2.03 - Poster Session/ Treatment of Locoregional Disease – NSCLC (ID 213)

    • Event: WCLC 2015
    • Type: Poster
    • Track: Treatment of Locoregional Disease – NSCLC
    • Presentations: 1
    • +

      P2.03-031 - Subgroup Analysis of East Asian Patients in the Phase III PROCLAIM Trial (ID 1293)

      09:30 - 17:00  |  Author(s): L.(. Wang

      • Abstract

      Background:
      PROCLAIM is a phase III trial comparing overall survival (OS) in patients with stage III, unresectable, nonsquamous non-small cell lung cancer (NSCLC) receiving pemetrexed (Pem) plus cisplatin (Cis) and concurrent thoracic radiation therapy (TRT) for 3 cycles followed by 4 cycles of Pem consolidation (Pem+Cis arm) versus etoposide (Etop) plus Cis and concurrent TRT for 2 cycles followed by up to 2 cycles of consolidation with a platinum-based doublet of choice (Etop+Cis arm). Overall efficacy and safety results for the intent-to-treat (ITT) population (N=598) will be presented in a separate disclosure. Efficacy and safety results from an East Asia (EA) subgroup analysis are presented here.

      Methods:
      A subgroup analysis was performed using the EA randomized population (N=97), which consisted of all patients who were randomized to the study from China (n=61), Taiwan (n=25), and The Republic of Korea (n=11). OS and progression-free survival (PFS) were evaluated by the Kaplan-Meier method and hazard ratios (HRs) were calculated using a Cox regression model. The log-rank test was used to compare treatment arms. Objective response rates (ORRs) were compared using an unadjusted, normal distribution approximation for the difference in rates. ClinicalTrials.gov number NCT00686959.

      Results:
      Baseline characteristics were balanced between treatment arms for EA patients. In the 97 randomized EA patients (n=44 in the Pem+Cis arm; n=53 in the Etop+Cis arm), median PFS was 10.0 months for the Pem+Cis arm and 7.6 months for the Etop+Cis arm (HR: 0.97, 95% confidence interval [CI]: 0.61–1.54, p=0.890). The censoring rate was high for OS (Pem+Cis arm: 43.2%; Etop+Cis arm: 52.8%), and there was no significant difference in OS between the Pem+Cis arm and the Etop+Cis arm (HR: 1.23, 95% CI: 0.70–2.14, p=0.469). The interaction test for region and treatment effect for OS was not significant (p=0.374). The ORRs were 47.7% (95% CI: 32.46–63.31) in the Pem+Cis arm and 34.0% (95% CI: 21.52–48.27) in the Etop+Cis arm. In the 90 treated EA patients (n=44 in the Pem+Cis arm; n=46 in the Etop+Cis arm), the overall incidence of drug-related grade 3/4 treatment-emergent adverse events (TEAEs) was significantly lower in the Pem+Cis arm versus the Etop+Cis arm (61.4% vs. 91.3%; p=0.001). All drug-related grade 3/4 TEAEs occurring in ≥5% of patients had a numerically lower incidence in the Pem+Cis arm than in the Etop+Cis arm except lymphopenia (17 [38.6%] vs. 17 [37.0%]).

      Conclusion:
      For EA patients with nonsquamous NSCLC, Pem+Cis did not improve OS, but did have a good safety profile and numerically improved PFS and ORR compared to Etop+Cis.