Virtual Library

Start Your Search

N. Pavlakis

Moderator of

  • +

    MINI 08 - Prognostic/Predictive Biomarkers (ID 106)

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

      MINI08.01 - Quantitative Mass Spectrometry Proteomics Identifies FRalpha and GARFT as Predictive Biomarkers in NSCLC Patients Treated With Pemetrexed (ID 1685)

      16:45 - 18:15  |  Author(s): A.A. Alshehri, E. An, F. Cecchi, T. Hembrough, P.C. Ma, M. Smolkin, S. Wen, M. Monga

      • Abstract
      • Presentation
      • Slides

      Background:
      Lung cancer remains the leading cause of cancer mortality in United States and globally. Pemetrexed combined with platinum chemotherapy is specifically indicated for treatment of non-squamous non-small cell lung cancer (non-sq NSCLC). Pemetrexed is a folate-analog metabolic inhibitor that disrupts folate-dependent processes essential for cell replication. Pemetrexed inhibits thymidylate synthase (TS), dihydrofolate reductase (DHFR), and glycinamide ribonucleotide formyltransferase (GARFT), which are folate-dependent enzymes involved in the de novo biosynthesis of thymidine and purine nucleotides. Folate receptor alpha (FRalpha) is a folate/antifolate transporter protein that is overexpressed by a number of epithelial tumors. The purpose of this study is to identify proteomic biomarkers predictive of response to pemetrexed-based chemotherapy in non-sq NSCLC.

      Methods:
      Patients with advanced non-sq NSCLC who received pemetrexed-based chemotherapy at West Virginia University from 2009 to 2014 were retrospectively identified. Formalin-fixed, paraffin-embedded tumor biopsies were laser microdissected, solubilized, enzymatically digested and subjected to quantitative proteomic analysis. A multiplexed, selected reaction monitoring (SRM) mass spectrometry (MS) assay was used to determine the absolute levels of 46 different candidate proteomic markers, including those in the folate receptor pathway. The Kaplan-Meier method and log-rank test were used in statistical analysis of overall survival (OS) and progression-free survival (PFS).

      Results:
      The 74 patients included in the study had a median follow-up of 26 months, a median OS of 16.6 months (95%CI: 11.6 - 43.4), and a median PFS of 9.61 months (95%CI: 8.43, 12.98). There were 65 patients who received pemetrexed-based regimen as a first line therapy and 9 patients as subsequent salvage treatment. In a comparison between patients who survived >24 months and < 8 months, there were no significant differences between the two groups in terms of sex, age, ECOG performance status, TNM stage at diagnosis, and smoking history. Among the 37 patients with sufficient tumor specimens available for multiplexed proteomic analysis, 30 biomarkers were detected with varying levels of expression. Sixteen additional biomarkers were undetectable. TS protein expression was detected in only 2 patients. Patients whose tumors expressed low levels of GARFT protein (≤900 amol/µg; n=7) had statistically significantly longer median PFS than those whose tumors expressed high levels of GARFT (>900 amol/µg; n=30) (40.6 vs. 11.4 months; p= 0.014). Patients with high FRalpha protein expression (>1510 amol/µg, n=9) had significantly longer median PFS than those with low FRalpha expression (≤1510 amol/µg; n=28) (>50 vs. 11.4 months; p= 0.021). Moreover, the 23 patients with both high GARFT expression (>900 amol/µg) and low FRalpha expression (≤1510 amol/µg) faired considerably worse than the remainder of patients (median PFS 10.1 vs. 40.6 months; p=0.0003).

      Conclusion:
      Multiplexed mass spectrometry-based proteomics offers a feasible and promising approach for tumor biomarker profiling and quantification to predict therapeutic response. Of note, our results show that FRalpha and GARFT protein expression may be predictive of response to pemetrexed-based treatment in patients with non-sq NSCLC. Further investigation is needed to validate the utility of these biomarkers for guiding personalized treatment decisions in clinical practice.

      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.

    • +

      MINI08.02 - Prediction of Response to Pemetrexed in NSCLC by Immunohistochemical Phenotyping Based on Gene Expression Profiles (ID 2793)

      16:45 - 18:15  |  Author(s): S. Visser, J. Hou, K. Bezemer, L. De Vogel, B. Stricker, J. Philipsen, J.G. Aerts

      • Abstract
      • Presentation
      • Slides

      Background:
      A major challenge in the treatment of advanced non-small cell lung cancer (NSCLC) is to identify specific tumor properties that predict response to chemotherapy. Although thymidylate synthase (TS) immunohistochemical (IHC) staining has been extensively studied as a predictive marker for pemetrexed (PEM) sensitivity, its clinical value remains limited. We investigated IHC stainings of different molecular markers linked to the folate metabolic pathway (FMP) identified with gene expression profiling (Hou et al, JTO 2012;7:105-114). We used a population with advanced NSCLC treated with PEM for external validation.

      Methods:
      Resected tumor samples from PEM-naïve NSCLC patients were collected. Gene expression profiling with respect to predicted sensitivity to PEM was based on genes related to FMP. Based on differentially expressed genes, patients were divided into predicted responders (Rs) and non-responders (NRs). Genes showing a strong correlation with these FMP genes and for which IHC stainings were commercially available, were selected for measurement of corresponding protein expressions by IHC stainings. A semiquantitative scoring method was applied, which was used to construct a prediction model for response to PEM. Subsequently, a retrospective cohort of patients with advanced NSCLC was selected, who had received at least two cycles of PEM-based chemotherapy as first-line treatment. IHC staining scores for the same proteins were obtained from tumor tissue. The performance of the prediction model was tested in this population.

      Results:
      From 91 patients resected tumor samples were collected. The majority of patients had early or locally advanced NSCLC (96.3%). Gene expression profiling revealed five markers that showed mRNA levels strongly correlating to FMP genes mRNA levels: TPX2, CPA3, EZH2, MCM2 and TOPO2a. Of 63 patients IHC staining scores of these markers were obtained, which all correlated to their corresponding mRNA levels. The scores were significantly different between predicted NRs and Rs (p<0.05). Testing the IHC markers showed an optimized prediction model with CPA3 (OR=1.71, 95%CI (0.94-3.08)), EZH2 (OR=0.57, 95%CI (0.35-.093)) and TPX2 (OR=0.55, 95%CI (0.29-1.03)) included. With this model 86.5% of the predicted Rs and 72.7% of the predicted NRs were correctly classified. The ROC showed an AUC of 0.883 representing a good discriminatory performance. In the external study population (n=23) the majority of patients had metastatic NSCLC (95.7%). Partial response (PR) was established in 26.1%. Considering patients with PR as responders the prediction model classified 16.7% of the observed Rs and 88.2% of the observed NRs correctly. The ROC showed an AUC of 0.750.

      Conclusion:
      Using external validation this prediction model with IHC staining of FMP correlated markers shows a good specificity, but lacks sensitivity. Again this study shows the limited value of IHC markers as response predictors for PEM in clinical practice. This may be ascribed to the poor relation between IHC and protein activity but the biological significance of FMP genes may also be less important than other factors influencing PEM activity, like pharmacodynamics of PEM e.g. the formation of metabolites. Metabolomics may offer better understanding in cellular processing of PEM and could provide new insights for tailored chemotherapy. Supported by an unrestricted grant from Eli-Lilly, the Netherlands

      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.

    • +

      MINI08.03 - Discussant for MINI08.01, MINI08.02 & MINI08.02b (ID 3546)

      16:45 - 18:15  |  Author(s): D.R. Spigel

      • 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.

    • +

      MINI08.04 - VeriStrat® and Epidermal Growth Factor Receptor Mutation Status in a Phase 1b/2 Study of Cabozantinib +/- Erlotinib in Non-Small Cell Lung Cancer (ID 552)

      16:45 - 18:15  |  Author(s): S.K. Padda, P. Lara Jr., S.N. Gettinger, J.A. Engelman, P.A. Jänne, H. West, L.Y. Zhou, D.S. Subramaniam, J.W. Leach, M.B. Wax, J.W. Neal, D.O. Clary, L.J. Goodman, H.A. Wakelee

      • Abstract
      • Presentation
      • Slides

      Background:
      VeriStrat is a blood-based multivariate proteomic test that predicts response to second line epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) therapy in non-small cell lung cancer (NSCLC). We report a retrospective blinded analysis of VeriStrat classification in plasma samples from a phase 1b/2 trial of cabozantinib (C) +/- erlotinib (E) in metastatic NSCLC patients who had all progressed after benefiting from EGFR TKI therapy. Cabozantinib inhibits the MET/hepatocyte growth factor (HGF) pathway, and VeriStrat may be a surrogate marker for this pathway.

      Methods:
      Patients enrolled into phase 1b (1A:60 mg C+150 mg E, 2A:60 mg C+100 mg E, 3A:100 mg C+100 mg E, 4A:100 mg C+50 mg E, 2B:40 mg C+150 mg E) and phase 2 (Arm A:100 mg C, Arm B:100 mg C+50 mg E). EGFR mutation (EGFRm) status was tested on archival tissue and/or plasma when available. The primary objective was to determine if pre-treatment VeriStrat (VS) classification, good or poor, was prognostic for patients treated with cabozantinib +/- erlotinib. Kaplan-Meier method and log-rank test was used to compare progression-free survival (PFS) of VS-good v. VS-poor patients. Outcomes were stratified by EGFRm status (mutated v. wild type WT/unknown UNK).

      Results:
      Of 79 evaluable patients, 71 were classified as VS-good and 8 as VS-poor. 55.7% had an activating EGFRm (majority exon 19 del/exon 21 L858R) and 12.7% had UNK EGFRm status. There were no significant differences in patient characteristics between VeriStrat-groups. VS-good patients had a statistically improved PFS: VS-good 3.7 mo. (95% CI 3.5-5.4) v. VS-poor 1.9 mo. (95% CI 1.1-3.4), p=0.014. This was still true after excluding 14 patients who had received cabozantinib alone (p=0.005). There was no difference in PFS for VS-good patients when stratified by EGFRm status. There was also no difference in PFS for VS-poor patients with WT/UNK EGFR v. VS-good patients irrespective of EGFRm status. However, VS-poor patients with WT/UNK EGFR had improved PFS compared to VS-poor patients with an EGFRm (3.1 mo. v. 1.6 mo., HR 0.15, 95% CI 0.03-0.68).

      Conclusion:
      VeriStrat is a strong prognostic marker in this study. This study suggests cabozantinib neutralized the worse prognosis of VS-poor patients with WT/UNK EGFR. Given the heterogeneity of treatment dosing, the small number of VS-poor patients, and a high proportion of unknown EGFRm (including T790M) status, this analysis should be considered exploratory.

      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.

    • +

      MINI08.05 - A Survival Comparison Study of Chinese Patients with Primary Lung Adenocarcinoma Harboring ALK Rearrangements Detected in Different Methods with Crizotinib Treatment (ID 3227)

      16:45 - 18:15  |  Author(s): S. Lu, X. Niu, Z. Chen, Z. Zhou, Z. Li, X. Ye, Y. Yu, Y. Xu, M. Liao

      • Abstract
      • Presentation
      • Slides

      Background:
      EML4-ALK is a new driver gene of non-small cell lung cancer (NSCLC) and is associated with response to inhibition with crizotinib. ALK break apart fluorescence in situ hybridization (FISH) assay, Ventana immunohistochemistry (IHC), and reverse transcriptase polymerase chain reaction (RT-PCR) can all be used as the primary assay for detecting ALK fusion events in tumor samples of lung cancer patients with SFDA approval in China. The objective of this study was to analyze the association of ALK rearrangements with clinical outcomes in different ALK testing methods, including FISH, Ventana IHC, and RT-PCR.

      Methods:
      ALK status was assessed by FISH, IHC and RT-PCR in 75 patients with advanced ALK-positive lung adenocarcinoma who had received crizotinib treatment from 2011, May to 2014, Nov in China. Clinicopathologic data and survival outcomes were analyzed. Kaplan-Meier cumulative probability was used to assess different testing methods for survival.

      Results:
      Of all 75 ALK-positive lung adonocarcinoma, there are 23 FISH-positive ALK patients (23/75, 30.7%), 35 IHC-positive ALK patients (35/75, 46.7%) and 17 RT-PCR-positive ALK patients (17/75, 22.7%). 75 patients received crizotinib treatment with IHC-positive and FISH-positive had better progression-free survival (PFS) (P=0.049, Fig A), compared with those with RT-PCR-positive, but not for overall survival (OS) (P=0.074). The median PFS survival for all these 75 patients was 16m, 14m, 8m, respectively, based on the IHC, FISH, and RT-PCR test (Fig A). 23 patients received first-line crizotinib treatment with IHC-positive and FISH-positive had better PFS (P=0.030), compared with those with RT-PCR-positive, but not for OS (P=0.061), either. The median PFS survival for these 23 patients with first-line crizotinib treatment was 12m, 18m, 4.8m, respectively, based on the IHC, FISH, and RT-PCR test. Of all 17 RT-PCR-positive ALK patients, there are 10 E13:A20 fusion type (10/17, 58.8%), 4 E6:A20 fusion type (4/17, 23.5%), 2 E18:A20 fusion type (2/17, 11.8%), and 1 E2:A20 fusion type (1/17, 5.9%). 4 different fusion-type ALK-positive patients detected by RT-PCR received crizotinib treatment with no crizotinib-related PFS significant difference (P=0.312) and no OS difference (P=0.149).Figure 1



      Conclusion:
      ALK-positive patients confirmed by IHC and FISH assay, compared with RT-PCR, maybe have better crizotinib-related PFS. RT-PCR method needs to be further evaluated in clinical practice to identify its role in guiding targeted therapy using crizotinib. And there is no survival difference for different ALK fusion type detected by RT-PCR in our cohort, which need further validation in a large group.

      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.

    • +

      MINI08.06 - Prognostic Significance of FGFR1 Amplification in Patients with Lung Squamous Cell Carcinoma (ID 814)

      16:45 - 18:15  |  Author(s): Q. Zhang, X.-. Zhang, Z. Xie, J. Su, Q. Zhou, X.-. Yang, W.-. Zhong, J.-. Yang, Y.-. Wu

      • Abstract
      • Presentation
      • Slides

      Background:
      The Fibroblast Growth Factor Receptor(FGFR) pathway especially FGFR1 gene copy number gain have attracted continuous attention of researchers for several years. Whereas due to different test methods and distinguishing criteria whether FGFR1 amplification related to patients smoking status or prognosis is still controversial.

      Methods:
      We used fluorescence in situ hybridization (FISH) to detect the gene copy number in paraffin-embedded tissue sections from 200 cases of pulmonary squamous cell carcinoma patients who underwent surgery in Guangdong Lung Caner Institute(GLCL) from 2008 to 2013. All samples had been identified as primary squamous cell carcinoma by postoperative pathology and informed consent. A tumor is defined as FGFR1 amplification positive when FISH results meet one of the following criteria after reviewing at least 100 tumor cells: (1) FGFR1/CEP-8 ratio≥2; (2) mean number of FGFR1 signals≥6; or if (3) ≥10% tumor cell containing more than 15 FGFR1 signals or large clusters. Among them, sample accord with the 3rd standard was defined as focal amplification.

      Results:
      Figure 1 We used fluorescence in situ hybridization (FISH) to detect the gene copy number in paraffin-embedded tissue sections from 200 cases of pulmonary squamous cell carcinoma patients who underwent surgery in Guangdong Lung Caner Institute(GLCL) from 2008 to 2013. All samples had been identified as primary squamous cell carcinoma by postoperative pathology and informed consent. A tumor is defined as FGFR1 amplification positive when FISH results meet one of the following criteria after reviewing at least 100 tumor cells: (1) FGFR1/CEP-8 ratio≥2; (2) mean number of FGFR1 signals≥6; or if (3) ≥10% tumor cell containing more than 15 FGFR1 signals or large clusters. Among them, sample accord with the 3rd standard was defined as focal amplification.



      Conclusion:
      Our results suggested that FGFR1 focal amplification might be an independent risk factor for patients overall survival. Patients with FGFR1 amplification were more likely to disease recurrence. Clinical characteristic including smoking status were not found in association with FGFR1 amplification, suggesting patients with FGFR1 amplification might not be fully enriched through only clinical factors.

      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.

    • +

      MINI08.07 - Discussant for MINI08.04, MINI08.05, MINI08.06 (ID 3327)

      16:45 - 18:15  |  Author(s): P. Hammerman

      • 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.

    • +

      MINI08.08 - VEGF-Mediated Cell Survival in NSCLC: Implications for Epigenetic Targeting of VEGF Receptors as a Therapeutic Approach (ID 2721)

      16:45 - 18:15  |  Author(s): M.P. Barr, K.J. O'Byrne, N. Al Sarraf, S. Cuffe, S. Finn, S.G. Gray

      • Abstract
      • Presentation
      • Slides

      Background:
      We have recently shown that VEGF, at least in part, is an autocrine growth factor for NSCLC cells, mediating its survival effects via VEGFR2 (KDR) in addition to the more novel receptor, Neuropilin-1 (Barr et al., Mol Cancer, 2015). In this study, we evaluated the potential therapeutic utility of histone deacetylase (HDAC) inhibitors in targeting the VEGF-VEGFR signalling axis in non-small cell lung cancer (NSCLC) cells.

      Methods:
      The effect of the HDAC inhibitor, Trichostatin-A (TSA) on modulating the expression of the VEGF receptors, VEGFR1, VEGFR2, NP1 and NP2, in A549 and SKMES-1 cells was examined and validated at the mRNA level and protein levels using RT-PCR and Western blot analysis. Gene expression was further validated by quantitative real-time PCR. To investigate the effect of TSA on the viability of NSCLC cells, these were treated with increasing concentrations of TSA (2.5 ng/ml-250 ng/ml) for 24h. Cell proliferation and apoptosis was measured by BrdU and Annexin V/PI (FACS), respectively. VEGF protein secretion in response to TSA was assessed in conditioned media from lung tumour cells by ELISA. To determine if the effects of TSA on VEGFR receptors were mediated through immediate to early responses, cells were pre-treated with cycloheximide (10 µg/ml) for 2 h followed by treatment with TSA (250 ng/ml) for 24 h. To confirm whether the observed effects of HDAC inhibition by TSA were due to increased histone hyperacetylation at the VEGFR1 and VEGFR2 gene promoters, chromatin immunoprecipitation (ChIP) analysis was carried out following treatment with TSA.

      Results:
      NP1 and NP2 mRNA levels were decreased in both A549 and SKMES-1 lung cancer cells in response to TSA and induced the expression of VEGFR1 and VEGFR2 at higher concentrations. TSA however, had no effect on VEGF mRNA expression. Critically, the effect of TSA was more marked at the protein level, with complete loss of Neuropilin-1 protein. HDAC inhibition resulted in a significant decrease in the viability of A549 and SKMES-1 cells in a dose-dependent fashion. While TSA induced significant apoptosis of both lung tumour cell lines, VEGF was unable to rescue cells from TSA-induced cell death. VEGF secretion was significantly decreased in both cell lines. Treatment with cycloheximide was unable to abrogate the TSA-mediated increase in the VEGF receptors examined, indicating that de novo protein synthesis is not required for these observed effects, but may be due to direct effects at the promoter level. Direct histone acetylation of histones H3 and H4 was observed, indicating an increase in histone hyperacetylation of VEGFR1 and VEGR2 promoters. A significant trend in the modulation of the VEGF receptors similar to that seen in response to TSA was shown when treated with Vorinostat (SAHA).

      Conclusion:
      Epigenetic targeting of the Neuropilin receptors may offer an effective treatment for NSCLC patients in the clinical setting. The possibility of novel targeted agents decreasing the levels, or function, of tumour VEGF receptors, in particular NP1, may lead to more successful treatments and prolonged overall survival in these patients.

      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.

    • +

      MINI08.09 - Anti-Tumor Efficacy of Interleukin-27 in Non Small Cell Lung Cancer (ID 551)

      16:45 - 18:15  |  Author(s): G. Cipollone, I. Airoldi, M.G. Tupone, S. Esposito, M.V. Russo, G. Barbarito, E. Di Carlo

      • Abstract
      • Presentation
      • Slides

      Background:
      Adenocarcinoma (AC) and Squamous Cell Carcinoma (SCC) constitute the commonest lung cancer histotypes, but current therapies still fail to significantly increase their survival rate. An effective immunotherapy to apply alternatively or together with specific treatments may be of great value. Here we asked whether Interleukin (IL)-27, which has revealed powerful antitumor activity in different tumor types and is toxicity-free in humans, is a promising therapeutic choice for NSCLC patients.

      Methods:
      Human lung AC and SCC cell lines were used to assess IL-27’s effect on cancer cell viability, by flow cytometry, and on malignancy-related gene expression, by qRT-PCR. Its effects on tumor growth were assessed in pre-clinical models and examined histopathologically. Expression of IL-27Receptor(R) in clinical samples was assessed by laser capture microdissection followed by qRT-PCR, and by immunohistochemistry.

      Results:
      In vitro, IL-27 was ineffective on cancer cell proliferation or apoptosis, but fostered CXCL3/GROg/MIP2b expression. In vitro and in vivo, IL-27 down-regulated stemness-related genes, namely SONIC HEDGEHOG in AC cells, and OCT4A, SOX2, NOTCH1, KLF4 along with the Epithelial to Mesenchymal Transition (EMT)-related genes NESTIN, SNAI1/Snail, SNAI2/Slug and ZEB1, in SCC cells. In vivo, IL-27 hampered both AC and SCC tumor growth in association with a prominent granulocyte- and macrophage-driven colliquative necrosis, CXCL3 production, and a reduced pluripotency- and EMT-related gene expression. Myeloablation of tumor-bearing hosts mostly abolished IL-27's antitumor effects. In clinical samples, IL-27R was expressed by the majority of AC, 90%, and SCC, 84%. Its expression by the primary tumor was significantly associated with advanced stages of disease (P = 0,02) as assessed by Fisher’s exact test. IL-27R was also expressed by pre-cancerous lesions, microvessels, and by infiltrating immune cells as CD15[+]granulocytes, CD68[+]monocytes/macrophages and CD11c[+]myeloid dendritic cells scattered in the stroma or within the lymph node–like structures, known as tertiary-lymphoid-structures (TLS).

      Conclusion:
      Altogether, our results highlight novel aspects of IL-27’s antitumor potential, specifically in NSCLC, such as the ability to drive myeloid cells towards antitumor activities, and down-regulate stemness genes, particularly in SCC cells, thus suppressing their self-renewal potential. IL-27 may thus be proposed for clinical trials with the prospect of its clinical use in immune-defective or advanced NSCLC patients.

      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.

    • +

      MINI08.10 - Co-Occurrence of Driver Mutations of MAPK and PI3K Pathways in Non Small Cell Lung Cancer: A Report from Lung Cancer Genomics Ireland (LCGI) Study (ID 2627)

      16:45 - 18:15  |  Author(s): S. Rafee, Y. Elamin, S. Toomey, K. Gately, A. Carr, S. Cuffe, S. Nicholson, S.P. Finn, R. Ryan, V. Young, J. Crown, O. Breathnach, P. Morris, E. Kay, A. O'Grady, B. Hennessy, K.J. O'Byrne

      • Abstract
      • Presentation
      • Slides

      Background:
      The mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways are frequently altered in human cancers. Targeting these pathways is an attractive therapeutic strategy in malignant disease. The frequency of single and dual pathway alterations varies substantially across various cancers. Co-occurrence of the MAPK and PI3K pathway aberrations is reported in 5-7% of melanomas, gastric and colorectal cancers, and is associated with a worse clinical outcome. In this report we aim to determine the co-occurrence of the MAPK and PI3K pathway mutations in a large cohort of surgically resected NSCLC tumors.

      Methods:
      We used the platform of Sequenom’s MassArray to perform genotyping for 548 somatic hotspot mutations in 49 genes including genes in the MAPK and PI3K pathways in surgically resected NSCLC tumors. MAPK pathway genes that were screened include: KRAS, HRAS, BRAF, RAF1, MAP3K1, MAP3K2, MAP3K3, MAP3K4, MAP3K5, MAP2K1, MAP2K2, MAP2K3, and PTPN11. PI3K pathway genes that were screened include: PIK3CA, PIK3R1, PIK3R2, PTEN, PDPK1, AKT1, AKT2, and MTOR. Fisher’s exact test was used to determine the statistical significance of association between the MAPK and PI3K pathway mutations. The strength of association was determined in the form of odds ratio.

      Results:
      NSCLC tumors from 356 patients (258 squamous cell, 98 adenocarcinomas) were tested using Sequenom’s MassArray. The frequency of mutations in the MAPK and PI3K pathways was 22.5% (n=80) and 22.8% (n=81) respectively. Among these patients, 38 patients had mutations in both pathways (i.e: 47.5% of patients with a MAPK pathway mutation also had a mutation in the PI3K pathway, and 46.9% of patients with a PI3K pathway mutation also had a mutation in the MAPK pathway, see table 1). Fisher’s exact test revealed that mutations in the MAPK and the PI3K pathways are mutually inclusive (p<0.0001, odds ratio=4.95, 95% CI 2.9-8.5) Table 1: The co-occurrence of MAPK and PI3K pathway mutations in NSCLC

      Pathway/no of patients PI3K WT PI3K MT
      MAPK WT 235 43
      MAPK MT 42 38


      Conclusion:
      38 (10.7%) of 356 NSCLC patients included in the LCGI study had hotspot somatic mutations in both the MAPK and PI3K pathways. Contrary to previous reports, we observed that activating mutations of the MAPK and PI3K pathways are mutually inclusive in NSCLC. These findings may have implications in designing clinical trials of targeted therapies in lung cancer.

      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.

    • +

      MINI08.11 - Genome-Wide Association Analysis Identifies Novel Genetic Determinants of Cancer Metastasis in Korean Lung Adenocarcinoma (ID 2363)

      16:45 - 18:15  |  Author(s): Y. Hwang, H.Y. Son, Y.J. Jung, S.B. Lee, Y.H. Kim, H.J. Lee, I.K. Park, C.H. Kang, J. Kim, Y.T. Kim

      • Abstract
      • Presentation
      • Slides

      Background:
      Non-small cell lung cancer (NSCLC) is characterized by poor prognosis, and few molecular markers were proven to be associated with cancer metastasis. The aim of the study is to identify the associations between single-nucleotide polymorphisms (SNPs) and distant metastasis of adenocarcinoma of the lung in Korean population.

      Methods:
      We conducted a genome-wide association study (GWAS) of in 499 Korean lung adenocarcinomas comparing 4,653,588 SNPs genotypes. Analyses were performed to investigate the association between the germline variations in all genes and cancer metastasis, survival and cancer recurrence.

      Results:
      We undertook a gene-metastasis interaction analysis in a GWAS of lung cancer using a case-control study. (18cases and 481controls) The combined analyses identified two susceptibility loci for metastasis risk in lung adenocarcinoma: SYNE1 [rs117050208, p-value = 1.91 x 10[-14], odds ratio (OR) = 87] and QSOX1 (rs148150589, p-value = 4.46 x 10[-9], OR = 56.5). SYNE1 was known to play crucial roles in the dynamics of genetic progression in colorectal adenocarcinoma and QSOXI was considered a prognostic indicator of metastatic potential in the breast cancer. (Figure1) However, no significant association was found between SNPs and either survival or recurrence. Figure 1



      Conclusion:
      Our data suggest that the SYNE1 rs117050208 and the QSOX1 rs148150589 may serve as potential biomarkers to influence cancer metastasis in the Korean lung adenocarcinoma. The analysis of the rs117050208 and rs148150589 polymorphism can help identify patients at high risk of distant metastasis of lung adenocarcinoma.

      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.

    • +

      MINI08.12 - Proteomic Profiling of Pulmonary Cancer with Squamous Cell Histology (ID 1377)

      16:45 - 18:15  |  Author(s): H. Bohnenberger, D. Yepes, K. Pan, A. Emmert, H. Henric-Petri, F. Bremmer, J. Strecker, A. Lois, L. Fischer, S. Küffer, M. Hinterthaner, H. Wolff, M. Canis, M. Sebastian, B. Danner, P. Ströbel, H. Serve, H. Urlaub, T. Oellerich

      • Abstract
      • Slides

      Background:
      Pathologic differentiation of neoplastic lesions in the lung with squamous cell histology is challenging as appropriate diagnostic immunohistochemical biomarkers are lacking. In particular patients with head and neck cancer and a smoking history can develop both lung metastases and primary lung cancer. Differentiation of primary lung cancer and lung metastases of head and neck cancer is clinically important for therapy and risk stratification. Furthermore, molecular targeted therapies for squamous cell carcinoma of the lung are largely lacking to date. Recent genetic studies uncovered multiple genetic subgroups of squamous cell carcinoma of the lung and moreover potential drug targets. However, the correlation between protein-expression/signaling activation patterns and genetic alterations is strongly influenced by co- and post-transcriptional as well as post-translational regulation. We characterized a broad panel of primary patient-derived formalin-fixed squamous cell carcinomas from lung and head and neck cancer by quantitative mass spectrometry to identify proteomic diagnostic biomarkers, signaling patterns and potential novel drug targets.

      Methods:
      Proteins were extracted from formalin-fixed paraffin-embedded (FFPE) microdissected patient-derived cancer tissues by using the “filter-aided sample preparation (FASP)” method. Purified proteins were subsequently mixed with a cancer-matched isotope labeled quantification standard (Super-SILAC standard) that allows for identification and quantification of thousands of proteins and their phosphorylation sites by high-end mass spectrometry. Using bioinformatics we determined the protein expression and signaling patterns. The biomarkers discovered were validated by immunohistochemistry in additional independent tumor tissues.

      Results:
      In this study we quantitatively characterized the proteomes of 60 primary patient-derived non-small cell lung cancer specimens with squamous cell histology and 25 squamous cell carcinomas from the head and neck region derived from patients that developed lung tumors with similar histology in the course of their disease. Using the Super-SILAC-based mass spectrometric approach we were able to identify and quantify around 2500 proteins per sample. Unsupervised clustering- and principal component analyses revealed that the detected protein expression patterns show a strong correlation with the cellular origin of the analyzed carcinomas. Furthermore, secondary lesions with similar histological morphology in the lung in patients with squamous cell carcinoma of the head and neck region could be classified as primary or metastatic cancer according to their protein expression profiles.

      Conclusion:
      Collectively, this study provides a large set of proteomic biomarkers that can be used to improve lung cancer diagnostics in the future. In particular the differential diagnosis of squamous cell carcinoma/metastases in the lung, that has so far been difficult due to the lack of biomarkers, will be improved by the biomarker panels presented here. Moreover, the expression and activation patterns of kinases discovered in our study is of interest regarding potential novel lung cancer therapies as overexpression or hyperactivation of certain kinases can potentially contribute to the malignant phenotype of lung cancer cells.

      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.

    • +

      MINI08.13 - Driver Mutation Status in Resected Stage I Lung Adenocarcinoma: Correlation with Radiographic CT Findings (ID 3251)

      16:45 - 18:15  |  Author(s): H.Y. Zhou, C.C. Wu, M. Mino-Kenudson, L.F. Tapias, M. Lanuti

      • Abstract
      • Presentation
      • Slides

      Background:
      To indentify the correlation of chest computed tomography appearance and the presence of oncogenic driver mutations in resected stage I lung adenocarcinoma

      Methods:
      Patients with resected stage I lung adenocarcinoma were analyzed from 2008-2012 and categorized into 3 groups: pure ground glass (GGO), mix-solid and ground glass, and solid patterns. All patients underwent driver mutation analysis (26 genes and 89 point mutations) using a multiplex PCR-based assay from paraffin embedded tumors. Disease free survival (DFS) and overall survival (OS) were compared between patients with EGFR, KRAS and the wild-type tumors using Kaplan-Meier methods and Cox regression models.

      Results:
      237 patients who underwent curative resection for stage I lung adenocarcinoma were analyzed with a median follow-up 34 months. Female gender was observed in 68% (160/237) and 21% (50/237) were nonsmokers. Pure GGO was indentified in 9% (n=21), mixed solid in 69% (n=164), and solid in 22% (n=52) of cases. EGFR and KRAS mutation rates were 18.6% (n= 44) and 34.6% (n= 82), respectively. Univariate analysis showed that KRAS-mutated tumors (HR 1.91, 95% CI 1.37-2.67; p<0.01), solid component > 50%, (HR 2.65, 95% CI 1.03-6.8; p=0.04), and smoking status (HR 3.59, 95% CI 1.1-11.8; p=0.03) were associated with worse DFS. In multivariate analysis only KRAS-mutated tumor (HR 1.84, 95% CI 1.31-2.59; p<0.01) was significant for worse DFS. KRAS-mutated tumor was also associated with worse OS in both univariate (HR 1.72, 95% CI 1.14-2.59; p=0.009) and multivariate (HR 1.65, 95% CI 1.09-2.49; p=0.018) analysis. Tumors that harbored >50% solid component on CT chest with a KRAS mutation were associated with worse DFS (HR 2.87, 95% CI 1.4-5.92; p=0.004) and OS (HR 2.51, 95% CI 1.03-6.1; p=0.04) in multivariate analysis compared to wild type tumors that were < 50% solid.

      Conclusion:
      KRAS mutation status and percent solid component on chest CT were predictive of worse outcome in surgically resected stage I lung adenocarcinoma

      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.

    • +

      MINI08.14 - Discussant for MINI08.08, MINI08.09, MINI08.10, MINI08.11, MINI08.12, MINI08.13 (ID 3328)

      16:45 - 18:15  |  Author(s): J.F. Gainor

      • 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.

    • +

      MINI26.01 - Tumour Molecular Profiling and Quantitative Detection of Circulating Biomarkers in Patients with Non-Small Cell Lung Cancer (NSCLC) (ID 317)

      16:45 - 18:15  |  Author(s): E. Karampini, H. Farah, N. Kamal, P. Cane, J. Moorhead, S. Pomplun, J. King, T. Sethi, J. Spicer, F. McCaughan

      • Abstract
      • Presentation
      • Slides

      Background:
      The introduction of targeted therapy has transformed the care of patients with lung cancer by incorporating tumour genotyping into therapeutic decision making. Recent improvements in sequencing technology have allowed for a rapid and broad snapshot of a tumour’s genetic landscape. Circulating cell-free tumour DNA (cfDNA) can be detected in patients with solid organ malignancies and has the potential to be used as a non-invasive biomarker (“liquid biopsy”). By integrating the two approaches, it is possible to detect specific mutational events in diagnostic samples, assess tumour burden, longitudinally monitor the response to therapeutic intervention and detect disease recurrence. As we have shown previously, it may also facilitate the detection of emergent subclonal populations, including variants that confer resistance to specific therapeutic agents.

      Methods:
      30 unselected treatment-naive patients with lung cancer were recruited from clinic. Paired DNA from tumour biopsies and plasma was obtained. Targeted next-generation sequencing (NGS) was performed on the tumour biopsy DNA. Primer sets and probes for identified mutations were optimised and validated on a microdroplet digital PCR (mdPCR) system.

      Results:
      25 of 30 patients in our test cohort had stage IIIB/IV non-small cell lung cancer. 25 of 30 patients (83%) of patients had mutations identified in their diagnostic specimen. 4 out of the 5 patients with no identifiable mutation in their diagnostic specimen presented with early disease and underwent curative surgery. The diagnostic specimens included endobronchial ultrasound (EBUS) samples, percutaneous and pleural biopsies, surgical resection specimens and a brain biopsy. The corresponding mutation was then assayed in cfDNA and was detected in the pre-treatment plasma samples in 90% of patients. Results to date from this cohort will be presented in detail. There has been complete concordance between mutations identified as part of the clinical standard-of-care and our targeted NGS data.

      Conclusion:
      It is feasible to perform a targeted NGS analysis on DNA from standard diagnostic lung cancer specimens and design generic and patient-specific biomarkers for use in a mdPCR assay of cfDNA. We aim to validate this approach and embed it in future clinical trial protocols.

      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.