Virtual Library

  • +

    Poster Display Session (ID 63)

    • Event: ELCC 2017
    • Type: Poster Display Session
    • Track:
    • Presentations: 133
    • Moderators:
    • Coordinates: 5/07/2017, 12:30 - 13:00, Hall 1
    • +

      4P - PD-L1 in NSCLC cytology (ID 446)

      12:30 - 12:30  |  Author(s): M. Kovacevic, I. Kern, S. Gabrič

      • Abstract

      Background:
      PD-L1 is a predictive biomarker for NSCLC, which is determined by immunohistochemistry. Significant number of NSCLCs are diagnosed from cytology samples. No study of PD-L1 expression in NSCLC cytology samples was published to date. The aim of this study was to evaluate possiblity of immunocytochemical determination of PD-L1 status in primary and metastatic NSCLC.

      Methods:
      We examined 50 consecutive cytology samples from 50 patients (19 TBNAs of mediastinal lymph nodes, 9 FNABs of peripheral lymph nodes, 15 TBNAs of lung, 4 pleural effusions, 2 FNABs of subcutaneous mass and one US – guided FNAB of liver). Methanol – fixed cytospins were prepared for immunocytochemistry using PD-L1 mouse monoclonal antibody (clone 22C3, Dako, USA) on an automated staining platform (Benchmark, Ventana/Roche, USA). Samples containing 100 or more tumor cells were considered representative. PD-L1 expression was evaluated on tumor cells with membranous staining. PD-L1 positivity was defined by cutoff value of 1%. 18 patients had concurrent histology samples used for PD-L1 immunohistochemistry (FFPE sections, same PD-L1 antibody clone and platform).

      Results:
      We found 37 (74%) adenocarcinomas, 7 (14%) squamous cell carcinomas, while 6 (12%) remained NSCLC-NOS. 74% of all NSCLC samples showed positive immunocytochemical reaction with PD-L1. 27 (73%) of all adenocarcinomas, 4 (57%) of all squamous cell carcinomas and 6 (100%) of all NSCLC-NOS showed positive immunocytochemical reaction with PD-L1. In patients with both cytology and histology samples, concordance in PD-L1 expression was 78%. All types of cytology samples examined in the study showed to be representative for evaluation. Most problems occurred in evaluation of pleural effusion due to nonspecific cytoplasmic staining for PD-L1 in histiocytes.

      Conclusions:
      Cytology samples are adequate for evaluation of PD-L1 expression in primary and metastatic NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Clinic Golnik

      Funding:
      University Clinic Golnik

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      5P - Repeated biopsy for mutational analysis on EGFR-mutated non-small cell lung cancer (EGFR-NSCLC): Feasibility and safety (ID 493)

      12:30 - 12:30  |  Author(s): G. Anguera, M. Riudavets, A. Torrego, V. Pajares, A. Gimenez, L. López-Vilaró, E. Martínez, J.C. Trujillo-Reyes, E. Lerma, M. Majem

      • Abstract

      Background:
      The majority of non-small cell lung cancer (NSCLC) patients (pts) with epidermal growth factor receptor (EGFR) mutation develop resistance to EGFR tyrosine kinase inhibitors, in 50-60% of the cases, mediated by the EGFR T790M-mutation. Repeated biopsy (Bx) is necessary after progression on initial therapy to check T790M status. The aim of this study is to evaluate the feasibility and safety of repeated Bx in clinical practice.

      Methods:
      Retrospective analysis of 110 repeated biopsies (Bxs) performed in 74 pts with advanced NSCLC during the last 4 years, 30 of them with EGFR-NSCLC underwent to 42 Bxs. The technical success rates for the repeated Bx and the adequacy rates of specimens were evaluated. Bx-related complications were recorded. Clinical details were collected.

      Results:
      42 repeated Bxs were performed in 30 pts with EGFR-NSCLC (6 men (20%), 24 women (80%)). Mean age 62 (36-82). Mean number of repeated Bxs per patient: 1 (1-3). At diagnosis (dx), exon 19 mutation was detected in 20 pts (66,7%) and exon 21 mutation in 10 (33,3%). The main reasons for repeated Bx were: mutational analysis at dx 5 (16,7%), insufficient material at dx 4 (13,3%) and detection of T790M at the moment of progression 21 (70%). The technical success rate for Bx was 35/42 (83.3%), and postprocedural complications occurred in 2/42 cases: 1 pneumothorax and 1 low bleeding. Bx specimens came mostly from primary tumor (25/42, 59,5%, followed by: lymph nodes (2/42, 4,8%), pleural fluid (3/42, 7,15%), liquid Bx (3/42, 7,15%), metastasis sites (9/42, 21,4%): bone 2, liver 2, spinal fluid 2, pleural 2, adrenal gland 1. The most used technique was bronchoscopy (19/42, 45,2%), followed by: percutaneous biopsy (11/42, 26,2%), thoracocentesis (4/42, 9,5%). Other techniques: liquid Bx 3 cases (7,15%), surgery 3 (7,15%) and lumbar puncture 2 (4,8%). Results from repeated Bx were used to select the next line of treatment in 28/42 procedures (66,7%), and 16 of them (57,1%) allowed to include the patient in a clinical trial.

      Conclusions:
      Our data demonstrate that repeated Bx in EGFR-NSCLC is safe and clinically feasible. Findings from repeated biopsy were used to direct subsequent treatment in 66.7% of pts and 16 of them (57,1%) allowed to include the patient in a clinical trial.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hospital de la Santa Creu i Sant Pau

      Funding:
      Hospital de la Santa Creu i Sant Pau

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      6P - Metformin sensitizes lung cancer cells to treatment by the tyrosine kinase inhibitor erlotinib (ID 315)

      12:30 - 12:30  |  Author(s): X. Wang, Y. Xiang, B. Gao

      • Abstract

      Background:
      Lung cancer is one of the deadliest malignant tumors with limited treatment options. Although targeted therapy, using tyrosine-kinase inhibitors, such as erlotinib, has shown therapeutic benefit, only 15% patients with mutated epidermal growth factor receptor (EGFR) in lung cancer cells are sensitive. Therefore, additional therapeutic strategy should be developed. Metformin (Met) has been used to treat type 2 diabetes (T2D) for nearly 60 years. Met reduces circulating glucose and insulin levels by inhibiting gluconeogenesis in the liver. Recently Met class drugs have been shown their anticancer properties, including in combination therapy, in which Met inhibits the growth of tumor initiating cells in breast cancer cell lines and prevents the relapse of tumors in vivo when combining with chemotherapy.

      Methods:
      The effects of Met to sensitizing wild type EGFR lung cancer cells to erlotinib were examined in vitro by using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT), flow cytometry analysis, Western blot analysis, chemotaxis assay, as well as in vivo experiments.

      Results:
      We found that MET sensitized lung cancer cells bearing wild-type EGFR to erlotinib treatment by enriching cancer cells expressing higher levels of EGFR with persistent phosphorylation. As a consequence, combination of MET and erlotinib more efficiently inhibited the growth of lung cancer cells both in vitro and in mice with xenografted tumors.

      Conclusions:
      These results suggest a novel approach to treating lung cancer cases which are originally resistant to erlotinib.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Ruijin Hospital

      Funding:
      This project was supported in part by Shanghai Municipal Health and Family Planning Commission Research Grants (Youth) 20154Y0089.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      7P - Population-level effect of targeted therapy in patients with advanced pulmonary adenocarcinoma: A Swiss prospective cohort study (ID 453)

      12:30 - 12:30  |  Author(s): C. Schwegler, D. Kaufmann, O. Gautschi, S. Aebi, J. Diebold, D. Pfeiffer

      • Abstract

      Background:
      Large cancer centres in the USA demonstrated that molecular diagnosis and targeted therapy improved overall survival (OS) of patients with advanced pulmonary adenocarcinoma (Kris, JAMA 2014). We validated this finding in a rural area of Switzerland, served by private practices, community hospitals, and a tertiary referral centre.

      Methods:
      We conducted a prospective cohort study with the Cancer Registry of Central Switzerland, established in 2010, covering 4 cantons and 517'000 inhabitants. We enrolled all residents newly diagnosed with stage IV pulmonary adenocarcinoma from 2010 until 2014. From the registry, the central pathology and the residents’ offices, we obtained the date of diagnosis and death, gender, smoking, tumour histology and stage, molecular testing (on demand), and therapy. We used the Kaplan-Meier method and a log-rank test to compare OS, and Cox proportional hazards model for association with age, gender and smoking. The cutoff date was February 2016. No adjustment was made for multiple testing. The study was approved by the IRB.

      Results:
      348 patients were included in the study. Median age at diagnosis was 66 years (range, 30-94), 190 (55%) patients were men, 197 (57%) were smokers or former smokers. Molecular testing (PCR, IHC and FISH) was performed in 279 (80%), 132 (38%) had oncogenic driver mutations (ODM): KRAS (17%), EGFR (12%), ALK (6%), HER2 (2%), BRAF (1%), RET (0,5%) or MET (0,5%). 56 patients with an ODM, mostly EGFR (34) and ALK (12), received genotype-matched targeted therapy, at least 25 (45%) of whom in a clinical trial or named patient program. Median OS was 18 months for patients with ODM and targeted therapy, 8 months for patients with ODM and conventional therapy, and 10 months for patients with no ODM and conventional therapy. For patients with ODM and targeted therapy, OS was significantly better than for patients with ODM and conventional therapy (HR 0.64, p = 0.04).

      Conclusions:
      Rigorous testing combined with optimal access to targeted therapy in clinical trials, improved the prognosis of patients with advanced pulmonary adenocarcinoma and EGFR mutation or ALK rearrangement. For patients with tumours lacking actionable mutations, further advances are expected with immunotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      J. Diebold, Institute of Pathology, Cantonal Hospital Lucerne, Lucerne, Switzerland

      Funding:
      Cancer League of Central Switzerland, Lucerne, Switzerland

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      8P - Expression of genes associated with anti-viral response in EGFR mutant non-small cell lung cancer (NSCLC) (ID 346)

      12:30 - 12:30  |  Author(s): N. Karachaliou, A. Giménez-Capitán, A. Drozdowskyj, E. Aldeguer, M. González Cao, A.F. Cardona, G. López-Vivanco, J.M. Sánchez, M.D.L.L. Gil, R. Rosell

      • Abstract

      Background:
      Up-regulation of endogenous retroviruses induces interferon response ("viral mimicry") and potentiates response to immune checkpoint therapy. Pattern-recognition receptors (PRRs), including retinoic acid-inducible gene-I (RIG-I), could up-regulate interferon pathway activating signal transducers and activators of transcription (STAT1), which ultimately induces CD274 (PD-L1) mRNA expression. We assume that alterations in the interferon pathway could be present in EGFR mutant NSCLC. Activation of STAT3 stimulates DNMT1 repressing PRRs. We recently identified YAP1 as a compensatory mechanism of resistance to gefitinib and osimertinib in EGFR mutant cells. Activation of YAP1 up-regulates CXCL5, a chemokine attracting myeloid-derived suppressor cell.

      Methods:
      Total RNA from 53 EGFR mutant NSCLC patients (pts), was reversed transcribed and analyzed by qRT-PCR. RIG-1, STAT1, CD274, DNMT1 and CXCL5 mRNA were examined with specific primers/probes in triplicates. Progression-free survival (PFS) and overall survival (OS) were estimated.

      Results:
      Fifty-three EGFR mutant NSCLC pts treated with erlotinib were analyzed, 65% were female, 65% never-smoked, 32% had brain metastases, 37% had bone metastases and 68% had exon 19 deletion. A close correlation was found between the RIG-1 and STAT1 mRNA levels (r = 0.42, p = 0.02). An anti-correlation trend was noted between DNMT1 and STAT1. Median PFS was 22 mo, 12.9 mo and 8.6 mo for pts with high, intermediate and low PD-L1 mRNA, respectively (P = 0.04). Median PFS was numerically longer for pts with low levels of DNMT1, RIG1 STAT1 and CXCL5, although the differences were not statistically significant. A similar trend was observed for OS.

      Conclusions:
      CD274 (PD-L1) mRNA could be a prognostic marker in EGFR mutant NSCLC pts. Down-modulation of CD274 indicates alterations in PRRs or downstream interferon signaling factors. The pathway dysregulation is multifactorial, and the role of STAT3 and YAP1 hyperactivation merits further research. DNMT1 overexpression ablates STAT1. Since the cyclin-dependent kinase 4 (CDK4) interacts with DNMT1, therapies with CDK4 inhibitors can directly neutralize the main defects in the interferon "viral mimicry" pathway.

      Clinical trial identification:


      Legal entity responsible for the study:
      Institut d\'Investigació en Ciències Germans Trias i Pujol, Badalona, Spain Institut Català d\'Oncologia, Hospital Germans Trias i Pujol, Badalona, Spain

      Funding:
      Institut d\'Investigació en Ciències Germans Trias i Pujol, Badalona, Spain Institut Català d\'Oncologia, Hospital Germans Trias i Pujol, Badalona, Spain

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      9P - Knockdown of MFN2 gene expression inhibits lung adenocarcinoma cell proliferation (ID 254)

      12:30 - 12:30  |  Author(s): Y. Lou, Y. Zhang, J. Xu, P. Gu, W. Zhang, X. Zhang, H. Zhong, L. Jiang, B. Han

      • Abstract

      Background:
      Mitofusin-2(MFN2) was initially identified as a hyperplasia suppressor in hyper-proliferative vascular smooth muscle cells of hypertensive rat arteries, which has also been implicated in various cancers. There exists a controversy in whether it is an oncogene or exerting anti-proliferative effect on tumor cells. Our previous cell cycle analysis and MTT assay showed that cell proliferation was inhibited in MFN2 deficient A549 human lung adenocarcinoma cells. MFN2-knockdown induced gene expression changes in A549 cells was analyzed by microarray assay and then functional pathway enrichment analysis revealed that six pathways were enriched in deregulated genes including Cell cycle, DNA replication, ECM-receptor interaction, Focal adhesion, MAPK signaling pathway and Chemokine signaling pathway, as we previously reported.

      Methods:
      MFN2 expression at protein level was examined in 30 pair lung adenocarcinoma/adjacent normal lung samples with immunohistochemistry staining. Then MFN2 knockdown was performed in human lung adenocarcinoma cells A549 with lentiviral-mediated shRNA strategy. The expression changes of downstream factors were determined in A549 cells by western blot. Furthermore, tumor models in nude mice were generated and tumor formation and progression in these mice were analysed.

      Results:
      As compared to adjacent normal lung tissues, MFN2 expression was significantly higher in lung adenocarcinoma tissues with positive MFN2 signals in 90% (27/30) lung adenocarcinoma tissues and only in 26.7% (8/30) adjacent normal tissues. The downregulation of RAP1A and upregulation of RALB and ITGA2 identified in MFN2-knockdown cells by microarray analysis were confirmed by western blot. In vivo, tumor models in nude mice were generated. Tumor formation and progression in nude mice suggested that MFN2 knockdown reduced tumorigenesis of A549 cells.

      Conclusions:
      The current study confirmed the anti-proliferative effect of MFN2 deficiency and its risk in lung adenocarcinoma.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      This work was supported by the National Natural Science foundation of China (81572249 and 81201839).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      10P - In vivo evaluation of combertestatin A-4 phosphate for lung cancer by bioluminescence imaging and multispectral optoacoustic tomography (ID 293)

      12:30 - 12:30  |  Author(s): L. Liu, S. Kumari, J. Campbell, R.P. Mason, L. Zhang

      • Abstract

      Background:
      Vascular disrupting agents (VDA) promise significant therapeutic efficacy against solid tumors by selectively destroying tumor vasculature. Combretestatin A-4 phosphate (CA4P) is a well-characterized VDA that inhibits microtubule polymerization by binding to the colchicine-binding site of beta-tubulin [1,2]. This disruption causes a decrease in blood flow, leading to ischemia and cell death. Photoacoustic tomography (PAT) is a non-invasive technique which allows for high-resolution (∼100μm) spectral imaging. The iThera Multispectral Optoacoustic Tomography inVision 256-TF (MSOT) system allows for high throughput, in vivo imaging of small animals. The MSOT has access to a range of frequencies (680-980nm) with 1nm resolution and so is able to differentiate between numerous different contrasts agents simultaneously.

      Methods:
      Female and male nude mice (each group n = 6) were implanted with 2x10[6] H1299 lung cancer cells subcutaneously. Ten days after implantation, the mice image were acquired in transaxial sections through the tumor region using nine wavelengths ranging from 700-880 nm by MSOT device and bioluminescence images (BLI) were taken by IVIS Spectrum at different time points. Images were reconstructed and processed using manufacturer’s software. Percentage of hemoglobin saturation (%SO~2~) was calculated as %SO~2~ = [HbO~2~/(HbO~2~+Hb)]*100.

      Results:
      VDA caused significant perfusion reduction and hypoxiation at 1 and 3 hours post VDA and mostly recovered at 24 hours. The %SO~2~ of tumor peripheral vessels was decreased from 42.5% at baseline to 33% and 28% at 1 and 3 hours respectively. The %SO2 of the tumor core reduced from 43% at baseline to 27% and 12% at 1 and 3 hours respectively. There was no change in %SO2 of normal tissue (spine). Similarly, BLI signal was reduced by > 90% within 3 hours after administration of CA4P indicating vascular disruption which was confirmed by histology.

      Conclusions:
      We demonstrated the feasibility of MSOT to detect changes in oxyhemoglobin and deoxyhemoglobin based on their unique absorption spectra in order to further elucidate the mechanism of action. CA4P caused significant reduction of perfusion and oxygenation within the tumor. The results indicate strong rationale for further development, particularly in combination with additional therapies.

      Clinical trial identification:


      Legal entity responsible for the study:
      Li Liu

      Funding:
      CPRIT RP160617

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      11P - Prognostic significance of different immunohistochemical markers in small cell lung cancer (ID 407)

      12:30 - 12:30  |  Author(s): E. Gkika, S. Beitinger, S. Adebahr, T. Schimek-Jasch, S. Wiesemann, C. Waller, A. Grosu, U. Nestle, G. Kayser

      • Abstract

      Background:
      To evaluate the role of different immunohistochemical markers concerning the overall and progression free survival in patients with small cell lung cancer (SCLC).

      Methods:
      A total of 104 consecutive patients with histologically proven SCLC (limited and extensive disease) treated with chemotherapy and radiotherapy at the University Hospital of Freiburg, between 2005 and 2012 and available pathologic slices, were included in the analysis. The expression of different immunohistochemical markers from the tissue biopsy such as CD56, CD44, Chromogranin, Synaptophysin, TTF-1, GLUT-1, Hif-1 alpha, PD-1 und PD-L1 and MIB-1/KI-67 as well as the LDH und NSE from the initial blood sample were correlated with the overall survival (OS) and progression free survival (PFS) using the Kaplan Meier Method and Cox proportional hazards ratio. Furthermore, for CD44, Hif-1 alpha and GLUT-1 H-Scores were generated for further analysis.

      Results:
      Of the included 104 patients, 48 had limited disease (LD) and 53 extensive disease (ED) at diagnosis. The median overall survival calculated from diagnosis was 21 (95%CI 19-23) months for patients with LD and 13 (95%CI 11-15) months for patients with ED. Patients with higher LDH (HR: 1.003 95% CI 1.001-1.005, p < 0.001) and NSE (HR:1.004, 95%CI: 1.002-1.007, p < 0.001) at diagnosis had a worse OS. These factors correlated also with a worse PFS (HR: 1.002 95% CI 1.000-1.003 p = 0.011 and HR: 1.004, 95% 1.001-1.006, p = 0.002, respectively). Concerning the immunohistchemical markers only the expression of Synaptophysin correlated with a worse OS (HR 1.890, CI 95% 1.067-3.346, p = 0.029) but not with the PFS (HR: 1.761, 95% CI 0.963-3.222, p = 0.066). All other markers did not correlate with OS or PFS.

      Conclusions:
      A positive immunhistochemical staining of synaptophysisn in patients with SCLC correlated with worse prognosis.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Freiburg

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      12P - Thyroid transcription factor-1 as a prognosticator in patients with metastatic lung adenocarcinoma without EGFR sensitizing mutations (ID 335)

      12:30 - 12:30  |  Author(s): J.Y. Park, S.H. Jang, J. Kim, S. Park, Y.I. Hwang, K. Jung, J.W. Seo

      • Abstract

      Background:
      Thyroid transcription factor-1 (TTF-1) expression is known not only as a diagnostic marker but also a good prognosticator among patients with lung adenocarcinoma. However, good prognosis of TTF-1 expression might be due to epidermal growth factor receptor (EGFR) sensitizing mutations because of the positive correlation between TTF-1 expression and EGFR mutations. The purpose of this study was to explore prognostic impact of TTF-1 expression according to EGFR sensitizing mutation status in lung adenocarcinoma.

      Methods:
      This is a retrospective analysis of patients with 1) stage IV lung adenocarcinoma having available TTF-1 immunohistochemistry result, 2) ECOG performance status 0-2, and 3) receiving systemic anti-cancer treatment. The data were extracted from the registry of Hallym University Sacred Heart Hospital in Korea, between March 2006 and March 2016.

      Results:
      Of the 697 patients with lung adenocarcinoma, 144 patients were included for analysis. The mean age was 65.2 ± 11.6 years (female; 42.4%). EGFR sensitizing mutations were detected in 72/144 (50.0%) patients. TTF-1 was positive in 116/144 (80.6%) patients, and it had a significant correlation with EGFR sensitizing mutations (p < 0.001). Patients with TTF-1 positive lung adenocarcinoma had longer overall survival (OS) than TTF-1 negative (21.4 vs. 6.5 months, p < 0.001). In Cox regression analysis, TTF-1 positivity (hazard ratio [HR] 0.50; 95% CI: 0.31-0.83; p = 0.007), Stage IV, M1a (HR 0.62; 95% CI, 0.40–0.97; p = 0.034), good performance status (ECOG=0; HR 0.52; 95% CI, 0.33–0.82; p = 0.005) and EGFR sensitizing mutations (HR 0.62; 95% CI, 0.39–0.97; p = 0.038) were independently associated with prolonged OS. In the subgroup of patients with wild type EGFR adenocarcinoma, TTF-1 positivity was also good prognosticator for OS (HR 0.58; 95% CI: 0.33-0.99; p = 0.046), and for progression-free survival (PFS) of the first-line cytotoxic chemotherapy (HR 0.43; 95% CI, 0.24–0.78; p = 0.006). (Table).rnTable: 12PCox proportional hazard model of overall survival for 72 patients with lung adenocarcinoma harboring wild type EGFRrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      CovariateUnivariate analysisMultivariate analysis
      P valueHR (95% CI)P valueHR (95% CI)
      Age < 70 vs. ≥ 700.9860.995 (0.607-1.633)0.5631.175 (0.680-2.030)
      Female vs. male0.0060.425 (0.231-0.780)0.4700.563 (0.119-2.676)
      Never smoker vs. ever smoker<0.0010.443 (0.248-0.790)0.9800.981 (0.222-4.330)
      Stage IV, M1a vs. M1b0.0620.602 (0.353-1.027)0.0460.569 (0.327-0.991)
      ECOG 0 vs. 1-20.0160.529 (0.316-0.887)0.0760.587 (0.325-1.057)
      TTF-1 positive vs. negative0.0600.607 (0.360-1.022)0.0460.575 (0.334-0.991)
      Pemetrexed, 1[st] line vs. non-pemetrexed containing regimen0.8620.956 (0.576-1.587)0.4800.829 (0.492-1.395)
      rnHR=Hazard ratio; CI=confidence intervals, ECOG = Eastern Cooperative Oncology Group; EGFR = epidermal growth factor receptor; TTF-1 = thyroid transcription factor 1.rn

      Conclusions:
      TTF-1 expression was a good prognosticator for OS and PFS in patients with stage IV lung adenocarcinoma without EGFR sensitizing mutations.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hallym University Sacred Heart Hospital Institutional Ethics Committee

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      13P - Characterizing the DNA damage response in putative stem cells of resected normal lung and matched NSCLC patient samples (ID 462)

      12:30 - 12:30  |  Author(s): T.M. Marti, C.C. Tièche, R. Peng, S.R.R. Hall, L. Froment, P. Dorn, S. Berezowska, R.A. Schmid

      • Abstract

      Background:
      More than 80% of lung tumors are non-small cell lung cancers (NSCLC). Tumor initiation and propagation are mediated by tumor initiating cells (TICs). In NSCLC and glioblastoma, TICs are characterized by high glycine decarboxylase (GLDC) expression. The DNA damage response (DDR) network evolved to maintain genome integrity and its dysregulation is associated with TICs and therapy resistance. Our aim was to establish a protocol to characterize the DDR status in NSCLC TICs.

      Methods:
      We evaluated a small cohort of eleven consenting patients who underwent elective surgery for lung cancer. Dissection of the tumor and non-adjacent normal lung tissue was performed by a pathologist. Tissue was dissociated to a single cell suspension, which was stained and analyzed by multicolor flow cytometry (FACS).

      Results:
      Based on EpCAM and GLDC expression, we identified four distinct subpopulations within the lineage negative compartment (live cells/CD31-/CD45-). In normal lung samples, a rare subpopulation was characterized by increased expression of the epithelial marker EpCAM and GLDC. Relative GLDC expression was increased in matched tumor samples. In normal samples, GLDC expression was associated with proliferation as indicated by Ki-67 staining, which was increased in matched tumor samples. Basal activation of the DDR as indicated by H2AX phosphorylation was increased in tumor versus normal samples. Upon DNA damage induction in both normal and cancer samples, H2AX was phosphorylated in the majority of the EpCAM+/GLDC+/- cells, in a smaller fraction of the EpCAM-/GLDC+ cells and absent in EpCAM-/GLDC- cells. Interestingly, the relative increase in basal versus DNA damage-induced H2AX phosphorylation was significantly higher in normal compared to matched cancer subpopulations. Further, variation in H2AX phosphorylation levels was higher in cancer samples. A larger cohort will be required to correlate DDR activation with the mutational status of the NSCLC samples.

      Conclusions:
      We established a FACS-based protocol allowing us to analyze DDR activation in normal versus tumor tissue and bulk versus cancer stem cells. This protocol can easily be adapted for the analysis of other types of patient tissue samples.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital Bern

      Funding:
      This work was supported by the Bernese Cancer League and the Swiss Cancer Research (KFS-3530-08-2014) to TMM.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      14P - Histone H2AX phosphorylation increases apoptosis of lung cancer cells by miR-3196/PUMA pathway (ID 213)

      12:30 - 12:30  |  Author(s): C. Lu, C. Xu, L. Zhang, L. Duan

      • Abstract

      Background:
      Increasing amount of studies indicate that histone H2AX acts as a novel tumor suppressor, but the detailed mechanism of apoptosis in tumor cells regulated by H2AX remains elusive.

      Methods:
      Microarray assay was performed to analyze differentially expressed apoptosis-related miRNAs regulated by H2AX in lung cancer cells. The flow cytometry (FCM) assay was performed for detection of apoptosis. Quantitative RT-PCR (qRT-PCR) was used to detect miR-3196 expression. Chromatin immunoprecipitation (ChIP) together with qRT-PCR was used to evaluate the binding of gamma-H2AX and RNA polymerase II to the miR-3196 promoter, and H3K27 trimethylation level in the promoter region of miR-3196. H2AX, BIRC7, PUMA and β-actin protein levels were determined by western blotting. The miR-3196 binding to PUMA mRNA at the 3′-UTR and miR-3196 promoter activity were evaluated by luciferase assay. Lipofectamine 2000 was used for transfection of vectors and miR-3196 mimics or miR-3196 inhibitor.

      Results:
      The data from microarray assay demonstrated that histone H2AX was involved in regulation of miRNAs expression during lung cancer cell apoptosis. H2AX knockdown affected the expression of 16 miRNAs, resulting in the downregulation of 1 and the upregulation of 15 miRNAs. Among the upregulated miRNAs, miR-3196 was identified as a target of H2AX and shown to inhibit apoptosis in lung cancer cells by targeting p53 upregulated modulator of apoptosis (PUMA). Phosphorylated H2AX (gamma-H2AX) was shown to bind to the promoter of miR-3196 and regulate its expression. In addition, H2AX phosphorylation increased H3K27 trimethylation in the promoter region of miR-3196 and inhibited the binding of RNA polymerase II to the promoter of miR-3196, leading to the inhibition of miR-3196 transcription. Taken together, these results indicated that miR-3196 is inhibited by H2AX phosphorylation and attenuates lung cancer cell apoptosis by downregulating PUMA.

      Conclusions:
      Histone H2AX phosphorylation increases apoptosis of lung cancer cells via the miR-3196/PUMA pathway.

      Clinical trial identification:
      This study does not contain clinical trial.

      Legal entity responsible for the study:
      Air Force General Hospital (PLA)

      Funding:
      National Natural Science Foundation of China

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      15P - PLGF regulates crosstalk between non-small cell lung cancer cells and tumor-associated macrophages in cancer vascularization and growth (ID 253)

      12:30 - 12:30  |  Author(s): W. Zhang, Y. Lou, B. Yan, Y. Zhao, S. Cui, L. Jiang, B. Han

      • Abstract

      Background:
      The growth and invasion of non-small cell lung cancer (NSCLC) require assistance of tumor-associated vascularization, the underlying molecular mechanisms of which remain eluted. Recently, we reported that placental growth factor (PLGF) was expressed by NSCLC cells, and promoted the metastasis of NSCLC cells. Here, we showed that NSCLC cells produced and secreted PLGF to signal to tumor-associated macrophages (TAM) through the surface expression of the receptor for PLGF, Flt-1, on macrophages.

      Methods:
      Ten week-old male NOD/SCID mice were used for transplantation of 10[7] AAV-transduced/labeled A549 cells by tail vein injection. Bones from 12-week-old male C57BL/6 mice were flushed with macrophage culture media. Isolated bone-marrow-derived macrophages (10[5]) were co-cultured either with equal number of A549 cells (10[5]) with/without of 10µmol/l SB431542, or with/without recombinant PLGF (100ng), or with/without of 10 µg/l sFlt-1. Two days after co-culture, the changes in A549 cell number were determined with an MTT assay, and the macrophage subtypes were determined by flow cytometry.

      Results:
      In a transwell co-culture system, PLGF secreted by NSCLC cells triggered macrophage polarization to a TAM subtype that promote growth of NSCLC cells. Moreover, polarized TAM seemed to secrete transforming growth factor β 1 (TGFβ1) to enhance the growth of endothelial cells in a HUVEC assay.

      Conclusions:
      Thus, our studies suggest that the cross-talk between TAM and NSCLC cells via PLGF/Flt-1 and TGFβ receptor signaling may promote the growth and vascularization of NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      This work was supported by the National Natural Science foundation of China (81402378).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      16P - Carvacrol induces growth inhibition and circumvents chemoresistance via inhibition of STAT3/Skp2/p27 pathway in non-small lung cancer cells (ID 269)

      12:30 - 12:30  |  Author(s): K. Kim, C. Lee

      • Abstract

      Background:
      S-phase kinase-associated protein 2 (Skp2) which constitues SCF complex and plays a role to recognize the substrates has been known to act as a proto-oncogene. In this study, we examined the effect of carvacrol, an active compound of oregano, on Skp2 inactivation and the underlying mechanisms in non-small lung cancer cells.

      Methods:
      Reagents and antibodies. Carvacrol was obtained from Sigma-Aldrich. A549 and H460 cells were purchased from the ATCC. For Western blot analysis, specific antibodies against Skp2, phospho-STAT3, STAT3, p21, p27, and GAPDH, as well as secondary antibodies were obtained from Santa Cruz Biotechnology.rnCell viability measurement. Cells were treated with carvacrol for 24 h and stained wtih 0.4% Trypan blue solution. Dye-excluding viable cells were counted under the microscope.rnClonogenic assay. Cells were seeded into 24 well plates and treated with carvacrol and then cultured for the next 7 to 10 days to form colonies. Colonies of > 50 cells were stained with crystal violet.rnEctopic expression of Skp2. To ectopically express Skp2, the recombinant plasmid, pcDNA3-Skp2-myc, was constructed and transfected into cells using Lipofectamine 2000. To establish stable cell lines, the transfected cells were cultured in the presence of 400 μg/ml of G418.rnsiRNA transfection. To reduce Skp2 expression, cells were transfected with siRNA targeting Skp2 or control siRNA.

      Results:
      Carvacrol treatment of A549 as well as H460 cells caused to reduce Skp2 protein level in dose-dependent manner. RT-PCR assay was also found that Skp2 mRNA level was reduced by carvacrol, suggesting the transcriptional down-regulation of Skp2 expression by carvacrol. We next found that the cytotoxic effect of carvacrol was attenuated in Skp2 overexpression in A459 cells and further observed its synergistic anti-proliferative effect in cells transfected with Skp2 specific siRNA. In addition, carvacrol was found to result in apoptotic cell death.

      Conclusions:
      Taken together, these data indicate that carvacrol targets Skp2 to inhibit cell proliferation and to cause apoptotic cell death in non-small lung cancer cells.

      Clinical trial identification:


      Legal entity responsible for the study:
      Yeungnam University College of Medicine, Republic of Korea

      Funding:
      Yeungnam University College of Medicine, Republic of Korea

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      19P - Domestic cooking fuel as a risk factor for lung cancer in women: A case control study (ID 227)

      12:30 - 12:30  |  Author(s): S.S. Katpattil

      • Abstract

      Background:
      Tobacco smoking is the most common risk factor for lung cancer. But a significant proportion of lung cancer occurs in non-smokers. Indoor pollution due to domestic fuels has been recently implicated as a causative agent in lung cancer especially in women. We conducted a case control study to find out the role of domestic cooking fuel as a risk factor for lung cancer in Indian women.

      Methods:
      In a case control study 67 women with proven lung cancer were recruited. Forty-six females having a non-malignant respiratory disease constituted the control group. The patients and controls were asked about the exposure in various cooking fuels using a questionnaire.

      Results:
      There were 50(74.6%) non-smokers and 17(25.4%) smokers among the female cancer cases (p = 0.016). Adenocarcinoma was the commonest histological type of malignancy (n = 26, 38.8%) in the whole group and was the predominant form in the non-smoking females. Tobacco smoking was the most important risk factor for lung cancer with OR of 4.87 (95% CI 1.34-17.76). Among non-smokers out of all the cooking fuels the risk of development of lung cancer was highest for biomass fuel exposure with an odds ratio of 5.33 (95% CI 1.7-16.7). Use of mixed fuels was associated with a lesser risk (OR = 3.04, 95% CI 1.1-8.38).

      Conclusions:
      This study indicated that domestic cooking fuel exposure is an important risk factor in the causation of lung cancer among women in addition of exposure to tobacco smoke.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      Al Iqbal Hospital

      Funding:
      Al Iqbal Hospital

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      21P - The efficacy of combination therapy with varenicline and bupropion for smoking cessation (ID 329)

      12:30 - 12:30  |  Author(s): D.A. Johns

      • Abstract

      Background:
      Extended use of combined pharmacotherapies to treat tobacco dependence may increase smoking abstinence. The objective of this study was to evaluate smoking abstinence by using bupropion alone, varenicline alone and combination of both.

      Methods:
      We conducted a randomized, prospective, double-blind, trial with 300 smokers enrolled. Group 1 bupropion alone (150 mg twice daily), group 2 varenicline alone (1 gm twice daily), group 3 varenicline + bupropion (1 gm twice daily+ 150 mg twice daily). The drugs were given for 12 weeks. Main outcome measures: The primary outcome was carbon monoxide-confirmed continuous abstinence rates (CAR) and the assessment of nicotine withdrawal symptoms. Nicotine dependence was assessed with the 6-item Fagerstrom Test for Nicotine Dependence scores ≥ 6 indicate high dependence. During the treatment phase, the Minnesota Nicotine Withdrawal Scale was used to assess withdrawal symptoms.

      Results:
      Subjects in the group 1,2 and 3 (n = 100 each) were comparable in age, smoking history, number of daily smoked cigarettes, and nicotine dependence. After the treatment there was reduced cigarette cravings (17.5 ± 1.1vs 15 ±3 vs 13.1 ± 1) and total nicotine withdrawal symptoms (17.8 ± 4.3 vs 15.4 ± 5.1 vs13.2 ± 2.1) during the treatment period. As compared to B and VR monotherapy, VR+B produced significant increases in CAR at 6 months (OR, 1.63; CI, 1.10-2.20, OR, 1.52; CI, 1.00-2.30 and OR, 1.72; CI, 1.06-2.67, respectively); results were similar, but somewhat stronger, when 7PP was used at 10 weeks (OR, 1.69; CI, 1.05-2.53, OR, 1.57; CI, 1.03-2.41 and OR, 1.82; CI, 1.09-2.83, respectively)All medications were well tolerated, but participants in the VR groups experienced more fatigue, digestive symptoms (e.g., nausea, diarrhea), and sleep-related concerns (e.g., abnormal dreams, insomnia). The frequency of serious adverse events did not differ between groups.

      Conclusions:
      Dual medication was found to enhance success in the early phases of quitting.

      Clinical trial identification:
      REF/2017/03/013652

      Legal entity responsible for the study:
      Abhilash Clinic

      Funding:
      N/A

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      23P - Pictorial warning on cigarette packets and its role in tobacco cessation: Questionnaire survey among cigarette smokers in Chennai (ID 320)

      12:30 - 12:30  |  Author(s): C.J.K. Francis

      • Abstract

      Background:
      Warning labels on cigarette packages are meant to communicate such smoking-associated risks. The study is designed to find out the effectiveness of pictorial warnings present on cigarette packets in India for tobacco cessation among cigarette smokers.

      Methods:
      A questionnaire was distributed to 800 current smokers attending an outpatient department of a college. Statistical analysis was done to find association between socioeconomic status and effectiveness of pictures to quit cigarette smoking.

      Results:
      48% smokers perceive text warning is an efficient method to create awareness. 56% emphasized the importance of pictorial warning and greater area to be covered. 43% felt that warning on cigarette packets helped them to quit smoking.

      Conclusions:
      Though pictorial warning is an effective method to improve the awareness among smokers on the ill effects of smoking, the size, area covered and the position of the picture on cigarette packets needs to be reviewed to improve the quit rate.

      Clinical trial identification:
      na

      Legal entity responsible for the study:
      N/A

      Funding:
      C.J.K. Francis

      Disclosure:
      Nestle India Limited

    • +

      25P - Tobacco consumption pattern among auto rickshaw drivers in Chennai city, Tamil Nadu, India (ID 310)

      12:30 - 12:30  |  Author(s): D.L. Francis

      • Abstract

      Background:
      Tobacco use is a major preventable cause of premature death and diseases, currently leading to five million deaths worldwide which are expected to raise over eight million deaths worldwide by 2030. India is the second largest consumer of tobacco in the world. This study is contemplated with an aim to assess the prevalence of tobacco consumption and the associated factors involved in its consumption, as this group of the population is under constant pressure and account for the workforce of the country. So through this study we could be able to know the awareness of ill effect, reasons & amount of consumption.

      Methods:
      Across sectional descriptive study was conducted among Auto Rickshaw Drivers in Chennai City. Auto drivers who were working for more than two years and present on the day of examination and who were willing to participate in the study were included. Cluster random sampling technique was used. 400 samples were selected from 40 auto stands of various parts of Chennai City. Data was collected using a Survey Proforma which comprised of a Questionnaire which can assess the frequency of consumption, age of initiation, the amount of consumption, mental stress, economic factors, any past history of disease and most importantly the awareness towards oral cancer. Age, tobacco consumption pattern, reasons of consumption, amount of consumption, harmful effects of tobacco are the variables.

      Results:
      Prevalence among auto rickshaw drivers for consumption of tobacco products was very high (87%). Auto rickshaw drivers were mostly used tobacco in the form of Gutkha (72%) and bidi (40%) in comparison to other products. It also shows that they use cheap tobacco products. Most of the auto rickshaw drivers start using tobacco products in age less than 18 years (80%) and associated factors for tobacco use are due to friends and their influence (78%). Awareness level among auto rickshaw driver was high (70%) but still uses tobacco products because of its addiction (66%). In the opinion of auto rickshaw drivers increase in tax may reduce it consumption and the majority of drivers (70%) think that tobacco must be banned.

      Conclusions:
      Prevalence of tobacco use among auto rickshaw drivers was very high. They are in definite need of tobacco cessation activities.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      N/A

      Funding:
      N/A

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      26P - Smoking habits and awareness about anti-smoking acts among general public in Gurgaon, Haryana, India (ID 319)

      12:30 - 12:30  |  Author(s): C.J.K. Francis

      • Abstract

      Background:
      India is the world’s third largest tobacco-growing country. The Indian scenario as far as tobacco consumption is concerned is far worse because of the prevalence of the tobacco chewing habit which covers a wide spectrum of socioeconomic and ethnic groups and is spread over urbanized area as well as remote village. Despite the facts, that the harmful effects of tobacco chewing and smoking are widely known, many young people start smoking during adolescence, largely because they believe that smoking will boost their social acceptability and image. This study was contemplated with an aim to assess tobacco/smoking habits and awareness about anti-smoking act among general public in Gurgaon, Haryana, India.

      Methods:
      A structured questionnaire consisting of 14 questions related to tobacco/smoking habits and awareness about anti-smoking act were asked to general public and their response was recorded. Random sampling method was used and data was collected from a cross-sectional survey. Anti-tobacco counselling was given on the spot and followed.

      Results:
      The study population consisted of total 430 individuals, male 364 (84.65%) and females 66 (15.34%). Then the questionnaires were asked and statistically analyzed. Around 286 (78.57%) from 364 males were indulged in some form of tobacco usage (smoker =32.86%, tobacco chewer = 16.78%, both =11.18%, alcohol +tobacco user =21.67%). In the present study, most common cause of tobacco use was pleasure 40.5%, inducing factor were friends 53.1% followed by parents and siblings.

      Conclusions:
      36.20% patients used tobacco as second hand exposure in job places. 54.8% were aware about the anti-smoking act in public places, so only 8.6% people from all males enrolled, were smoking in public places.

      Clinical trial identification:
      na

      Legal entity responsible for the study:
      N/A

      Funding:
      C.J.K. Francis

      Disclosure:
      Nestle India Limited

    • +

      27P - Factors causing treatment delays and its impact on treatment outcome in patients of lung cancer: An analysis (ID 214)

      12:30 - 12:30  |  Author(s): D. Kumar, S. Singh

      • Abstract

      Background:
      Delays in diagnosis and treatment are undesirable in cancer management. In this analysis, we tried to identify factors causing delay in diagnosis and subsequently its effect on outcome in patients of lung cancer presenting to the oncology department.

      Methods:
      55 patients with proven histopathological diagnosis of lung cancer were interviewed. Results were analysed statistically.

      Results:
      M:F=3:1.Mean age 49 years (32-65 years).NSCLC:SCLC=4:1, majorly adenocarcinoma. The duration of symptoms ranged from 1 month to 2 years. The number of illiterate patients presented mainly in advanced stages as compared to their educated counterparts and the difference was statistically significant (P<0.001). The number of patients presenting directly to the department was 8, those diagnosed outside and referred to us was 29 while those diagnosed and received some form of oncologic treatment outside and referred thereafter was 18. The difference in the primary delay between patients presenting directly to the institute versus those diagnosed outside was significant (P=0.01). The mean time to starting definitive treatment after presentation to the outpatient was 3 days (range 0-15 days) and was very significantly (P< 0.001) less than the secondary delays caused to the other two subsets of patients. The most important patient reasons for the delay in diagnosis were - financial constraints (20/55, 28.3%), ignorance about disease (9/55, 16.36%); dependency to escort (5/55, 9.0%), far off native place (7/55,12.7%), faith in local practitioner (3/55, 5.4%), family occasions (2/55, 3.6%), fear of death (2/55, 3.6%). 4/55 (7.2%) were initially diagnosed as tuberculosis and hence the delay, and 3/55 (5.4%) had no reason cited. Stages at diagnosis were stage II (4/55, 7.2%), stage III (41/55, 74.5%), stage IV (10/55,18.1%). Advanced stage disease patients had statistically significant poorer outcomes compared with those with earlier stage disease.

      Conclusions:
      Factors causing delayed presentation are both patient and system related. Awareness regarding the early signs and symptoms of lung cancer should be encouraged through various media channels. At the same time, the system needs to intensify its efficiency to avoid secondary delays that adversely affect the treatment outcome.

      Clinical trial identification:
      not applicable

      Legal entity responsible for the study:
      N/A

      Funding:
      None

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      28P - Impact of a pharmaceutical consultation: Review of a pilot study (ID 382)

      12:30 - 12:30  |  Author(s): E. Clou, J. Gaudas, K. Ahmed, D. Avenin, A. Esteso, G. Galula, J. Fillon, J.P. Lotz, I. Debrix

      • Abstract

      Background:
      The development of oral anticancer drug has resulted in several challenges. Prescriptions require a specific monitoring of the patient during his treatment. The objective of this work, resulted from collaboration among health professionals, was to assess the feasibility of setting up a pharmaceutical consultation in department of medical oncology. In addition, we aimed to evaluate the impact of this consultation in reducing prescription errors, drug-drug interactions finding, the early detection of adverse events (AEs) and in the patients’ adherence to both their treatments and the present approach.

      Methods:
      Before the initiation of oral chemotherapy, the oncologist proposed to the patient at participating in a pharmaceutical consultation. Following the patients’ agreement, the pharmacist collected several datas (medical history, community pharmacy medical prescriptions, treating physician) and agreed to an appointment with him. This process allowed to obtain the Best Possible Medication History and establish with the patient the treatment plan. During consultation, the resident assessed the patients’ compliance (with Morisky Scale questionnaire) and informed about the preventive and corrective measures to be implemented in case of AEs. All this datas were related in a specific notebook. A telephone follow-up was proposed to the patient. At last, a report containing all the pharmaco-therapeutic analysis was sent to the oncologist.

      Results:
      The pharmaceutical consultations allowed follow-up of 17 patients. Of the 19 pharmaceutical interventions conducted during the consultations, 94% involved drug-drug interactions. All patients participated to the telephone follow-up. 49 telephones calls allowed the detection of 25 AEs, half of which were referred to other health professionals. For 2 patients, anticancer drug had to be stopped for a bad tolerance finding telephone follow-up. Compliance remained constant over time for all patients. An average score of 9.4/10 was noted by patients for global satisfaction with the activity.

      Conclusions:
      Those results have showed that pharmaceutical consultation could enhance quality management of patients treated by oral anticancer drugs. It highlighted the major interest of this new health professional’s collaboration.

      Clinical trial identification:


      Legal entity responsible for the study:
      Isabelle Debrix

      Funding:
      Hospital Tenon (Paris 20ème)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      29P - Baseline characteristics of participants in pl-208: A multi-center trial of the prolung test™ (Transthoracic Bioconductance Measurement) as an adjunct to CT chest scans for the risk stratification of patients with pulmonary lesions suspicious for lung cancer (ID 334)

      12:30 - 12:30  |  Author(s): R.C. Yung, D. Ost, M. Simoff, C. Reddy, A. Goyal, I. Barjakarevic, M. Garff, K. Larson, J. O'Driscoll, S. Makani

      • Abstract

      Background:
      Lung cancer screening with low dose CTs shows numerous indeterminate pulmonary lesions (IPLs). Followup evaluation entail additional radiation (full dose CT/PET) or invasive biopsies. Alternative, novel adjunctive risk-stratification pathways are needed. Volume-averaged thoracic bioconductance, is a non-invasive technology measured transcutaneously by ProLung’s Electro-Pulmonary Nodule Scan (EPNS). A single center study had demonstrated potential to risk stratify such lesions.[1] with sensitivity/specificity of 90%/92% respectively and ROC >0.90. We present interim patient data of PL-208, a pivotal multicenter EPNS validation study.

      Methods:
      This is IRB approved and prospectively enrolled IPNs being evaluated. It is a two-phase study consisting of 350 subjects, 200 in the algorithm stabilization phase and 150 subjects in a subsequent algorithm validation phase. EPNS is performed within 60 days of the CT and pre tissue biopsies. In this prospective study, all study subjects & investigators are blinded to the EPNS results.

      Results:
      We present data on the initial 133 subjects in the table comparing data between the earlier feasibility study FML204[1] and PL208. 46(35%) subjects were asymptomatic; the rest had symptoms leading to imaging. Diagnoses include: 84 (63%) lung cancers, 4 (3%) non-primary lung cancers, 20 (15%) benign by biopsy, and 25 (19%) benign by CT follow-up.

      Conclusions:
      PL-208 cohort has a similar gender and age distribution to the PL204 feasibility study using EPNS as a risk-stratifying tool in evaluating suspicious IPLs. PL-208 includes asymptomatic nodules detected during LDCT screening, and hence more subjects with small T1a IPLs (17% Vs 7%), but overall a majority of subjects have IPNs < 3 cm (59%/56%). Of LCs diagnosed, 38%/41% are in stages I/II. [1]Yung et al. Transcutaneous Computed Bioconductance Measurement. J Thorac Oncol 2012; 7:681–9.\r\nTable: 29PComparison FML-204 and PL-208 patient characteristics\r\n

      \r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n\r\n
      SUBJECTSFML-204PL-208
      Age Mean(range)64 (34-80)65 (22-85)
      Male (%)22 (54%)68 (51%)
      Symptomatic41 (100%)87 (65%)
      COPD diagnosis19 (46%)27 (20%)
      NODULE FEATURES\r\n\r\n
      Solitary/multiple17(41%)/24 (59%)45 (34%)/88 (66%)
      0-10 mm (T1a)2 (7%)23 (17%)
      11-20 mm (T1b)13 (32%)35 (26%)
      21-30 mm (T1c)7 (17%)21 (16%)
      31-50 mm (T2a)9 (22%)25 (19%)
      > 50 mm (T2b/c)8 (20%)22 (17%)
      Solid37 (90%)111 (83%)
      Part solid/GGO3 (7%)/1 (2%)13 (10%)/9 (7%)
      DIAGNOSIS\r\n\r\n
      Benign by biopsy/CT-fu9 (22%)/3 (7%)20 (15%)/25 (19%)
      Small cell lung cancer2 (5%)10 (8%)
      Adenocarcinoma20 (49%)51 (38%)
      Squamous cell carcinoma6 (15%)17 (13%)
      Carcinoid1 (2%)6 (5%)
      Non-lung malignancy0 (0%)4 (3%)
      STAGE\r\n\r\n
      I11 (38%)20 (24%)
      II1 (3%)12 (14%)
      III6 (21%)23 (27%)
      IV11 (38%)20 (24%)
      Other0 (0%)9 (11%)
      \r\n

      Clinical trial identification:
      NCT01566682

      Legal entity responsible for the study:
      ProLungDx

      Funding:
      ProLungDx

      Disclosure:
      R.C. Yung: Conducted study with ProLung (corporate) sponsorship of study. Lead author also on ProLung Advisory Board, have received consultation fees and travel support (to meetings). Have been offered stock options. D. Ost: Conducted study with ProLung (corporate) sponsorship of study. Have received consultation fees on Advisory Board. M. Simoff, C. Reddy, A. Goyal, I. Barjakarevic: Conducted study with ProLung (corporate) sponsorship of study. M. Garff: Employee of ProLungDx. K. Larson: Employee of ProLung. J. O\'Driscoll: Chief Medical Officer and member of the Board of Directors for ProLungDx. S. Makani: Funded corporate sponsored research from ProLungDx.

    • +

      31P - Targeted therapy for patients with ALK positive NSCLC: Results from the transalpine cohort (ID 332)

      12:30 - 12:30  |  Author(s): C. Britschgi, M. Rechsteiner, R. Delaloye, M. Früh, G. Metro, O. Gautschi, S.I. Rothschild, R.A. Stahel, P.J. Wild, A. Curioni-Fontecedro

      • Abstract

      Background:
      Anaplastic lymphoma kinase (ALK) rearrangements occur in 3-5% of lung adenocarcinoma. ALK tyrosine kinase inhibitors (ALK-TKIs) are used for the treatment of these patients as standard of care, in clinical trials or in expanded access programs. The aim of this study was to analyze the clinical characteristics of response and progression to different ALK-TKIs and to study mechanisms of resistance using next generation sequencing (NGS).

      Methods:
      This collaborative study was performed in five institutions in Switzerland and Italy. Clinical data from patients diagnosed with ALK positive NSCLC (i.e. break in 2p23 in FISH) in the palliative setting were collected and analyzed. If available, biopsies both before starting ALK-TKI and at progression were subjected to NGS using the Ion AmpliSeq Comprehensive Cancer Panel.

      Results:
      We collected clinical data of 139 patients. 117 have so far undergone first-line treatment with chemotherapy (ChT) (n = 87, 74%), ALK-TKIs (n = 29, 25%) or other (n = 1, 1%). Second-line therapy was given in 92 patients; 16 received ChT (17%), 68 ALK-TKIs (74%), 8 immuno- (I-O) and other therapies (9%). 55 patients underwent third-line therapy comprising ChT (n = 11, 20%), ALK-TKIs (n = 40, 75%), or I-O and others (n = 3, 5%). Fourth-line treatment was given to 24 patients; ChT (n = 8, 33%), ALK-TKIs (n = 15, 63%) and other treatments (n = 1, 4%). 11 patients have received five or more lines of therapy. Crizotinib was given to 102 patients (81% in first- or second-line) resulting in a median PFS of 11.7 months (95% confidence interval [CI] 8.2-13.1). Ceritinib was given to 37 patients (76% in third- or later line) with a median PFS of 6.5 months (95% CI 3.7-9.4). 26 patients received alectinib (85% in third- or later line) with a median PFS of 12.4 months (95% CI 8.4-17.0). So far, NGS has been performed in 5 patients before and after ALK-TKI therapy. Two secondary ALK mutations were detected (ALK p.I1171S and p.C1156Y) conferring responsiveness to further ALK-TKIs.

      Conclusions:
      This study confirms the activity of ALK-TKIs in ALK positive NSCLC and allowed the documentation of important clinical data in the real-life setting. Moreover, an in-depth molecular analysis of resistance mechanisms was performed, which will now be expanded to more patients from the cohort.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital Zurich

      Funding:
      University Hospital Zurich, Novartis

      Disclosure:
      S.I. Rothschild: Compensation (to the institution) for advisory boards from Novartis, Pfizer and Roche. Compensation (to the institution) for invited talks from Pfizer and Roche. All other authors have declared no conflicts of interest.

    • +

      32P - Effects of BTK inhibitor on gefitinib-resistance non-small cell lung cancer (ID 162)

      12:30 - 12:30  |  Author(s): T. Hong, Y. Fang, Y. Chen

      • Abstract

      Background:
      Lung cancer is the most frequent cause of cancer death in the world. EGFR-mutant lung cancer is a subtype of non–small cell lung cancer (NSCLC) that exhibits sensitivity to EGFR tyrosine kinase inhibitors (TKIs) such as gefitinib (Iressa); however, acquired resistance was developed after a median of 9–14 months. In our previous study, we used two primary TKI-resistant lung cancer cell lines (CL25 and CL100) and a genome-wide human kinase and phosphatase RNAi screening and found that silencing Bruton tyrosine kinase (BTK) significantly inhibited the NSCLC viability. BTK is a non-receptor tyrosine kinase of the Tec kinase family and plays an important role in B-cell receptor signaling. Therefore, we wanted to combine primary TKI (Iressa) with BTK inhibitors (Ibrutinib and CC-292) as a cancer treatment for EGFR TKI-resistant NSCLC patients.

      Methods:
      First, we verified that BTK knockdown decreased cell viability in EGFR TKI-resistant CL25 cells and enhanced the sensitivity to Iressa treatment. Second, we test the 50% inhibitory concentration (IC50) of BTK inhibitors (Ibrutinib and CC-292) in NSCLC cells by WST-1 assay. Next, we analyzed the effects of BTK inhibitors on the downstream signaling of BTK by western blot. Furthermore, we investigated the mechanism of growth inhibition triggered by BTK inhibitors in CL25 cells. The cell cycle distribution and cell death were examined. Finally, we tried to understand whether BTK inhibitors could enhance the gefitinib -induced cell death in EGFR TKI-resistant CL25 cells by combined treatment with gefitinib and BTK inhibitors.

      Results:
      By a genome-wide human kinase and phosphatase RNAi screening and found that BTK may contribute to the primary resistance. Knockdown of BTK expression decreased the NSCLC cell viability in a dose dependent manner and increased the gefitinib-induced cell death in both EGFR TKI-resistant CL25 and CL100 cells. BTK knockdown increased the levels of apoptotic and autophagic markers and cyclinD1 by western blot analysis. Moreover, BTK inhibitors affected BTK, NFkB, PI3K/AKT and EGFR signaling in EGFR TKI-resistant CL25 cells. Besides, treatment with Ibrutinib or CC-292 at the concentration of 4 fold IC50 induced G1 arrest in CL25 cells. Finally, combination of BTK inhibitors and gefitinib could not enhance the gefitinib-induced cell death.

      Conclusions:
      According to above results, we found that BTK may be a candidate contributing to the primary EGFR-TKI resistance in NSCLC cells. Knockdown of BTK induces the autophagic cell death and the G1 arrest in primary EGFR TKI-resistant CL25 cells and enhances the gefitinib-induced cell death while BTK inhibitors induce G1 arrest but cannot enhance the gefitinib-induced cell death.

      Clinical trial identification:


      Legal entity responsible for the study:
      National Cheng Kung University

      Funding:
      Ministry of Science and Technology, Taiwan.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      33P - The screening and characterization of aptamer against gefitinib-resistant cells (ID 161)

      12:30 - 12:30  |  Author(s): Y. Chen, J. Wu, T. Hong

      • Abstract

      Background:
      The EGFR tyrosine kinase inhibitor (TKI) gefitinib serves as first-line drug for patients with advanced non-small cell lung cancer (NSCLC) harboring EGFR mutation. The challenge of this target therapy is the acquired resistance due to T790M mutation after gefitinib treatment. Aptamers are single-strand DNA molecules that form 3D structures and specifically bind to target components. In this study, we wanted to isolate aptamers with recognition ability for gefitinib-resistant NSCLC cells and look for another mechanism that contributes to the acquired resistance that is not caused by T790M mutation.

      Methods:
      We used PC9 (gefitinib-sensitive cells) and PC9-IR (gefitinib-resistant cells with no T790M mutation) cells to select specific aptamers that bound to membrane proteins which were overexpressed in PC9-IR cells. We used suction-type microfluidic control module to perform cell-systematic evolution of ligands by exponential enrichment (Cell-SELEX) and obtained some aptamers that had more affinity to bind to PC9-IR. We examined the specificity of the aptamers and observed whether aptamers could bind to cell membranes of PC9-IR. In addition, we tested the cell cytotoxicity of the aptamers.

      Results:
      We obtained the aptamer, AP16-23F, which had greater affinity to bind to PC9-IR. In order to test whether AP16-23F could recognize the cells with gefitinib resistance, we used AP16-23F to isolate cells from PC9 by fluorescence-activated cell sorting (FACS). The results showed that the cells selected by AP16-23F were more resistant to gefitinib.

      Conclusions:
      We have isolated an aptamer with specificity for binding and capturing gefitinib-resistant NSCLC cells. This aptamer may be useful for drug resistance detection and may have the potential to deliver anti-cancer drug to gefitinib-resistant cells in the future.

      Clinical trial identification:


      Legal entity responsible for the study:
      National Cheng Kung University

      Funding:
      Ministry of Science and Technology, Taiwan

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      34P - Discovery of 1,3,5-triazine-monastrol based novel EGFR-tyrosine kinase inhibitor against human lung carcinoma (ID 371)

      12:30 - 12:30  |  Author(s): U.P. Singh, J.K. Shrivastava, A. Verma, H.R. Bhat

      • Abstract

      Background:
      In India, the number of new lung cancer cases has been increased with an annual rise of 15-20%. This poses a significant burden on already struggling healthcare services of the country. Thus, more efforts have been directed towards the easy access of the medicines and development of novel cost effective drug for the under-privileged. Consequently, introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) has led to dramatic clinical improvement in non-small cell lung cancer (NSCLC). However, resistance to these inhibitors either via intrinsic and acquired resistance, put a selective pressure on the development of novel inhibitors. Therefore, the present study was aimed to develop novel 1,3,5-triazine derivatives as EGFR-TKIs for lung carcinoma.

      Methods:
      The 1,3,5-triazine-monastrol compounds were synthesized via multi-component reaction. The compounds were tested for determination of anticancer activity three human NSCLC cell lines A549, H157 and H52. The compounds were also tested for effect on cell growth inhibition and apoptosis through cell cycle arrest assay. The docking analysis was also carried out with EGFR-TK domain (PDB ID: 1M17) to elucidate vital structural residues necessary for bioactivity.

      Results:
      The cytotoxicity studies results suggest that, synthesized derivatives exhibit considerable inhibition with average IC~50~ for compound 7a were found to be 3.21, 6.32 and 11.05 µmol/l. It has been found that, 7h showed augmention in the number of cells in G0–G1 phase, with a subsequent decrease in the cells belonging to S and G2–M phase. It also causes modulation of tumor cell apoptosis in a concentration-dependent way. In docking study, it has been found that, compound 7h, 7l, 7m, 7e were found to be the most efficient analogues to attenuate EGFR-TKs via creating MET769, ASP831, LYS721 and CYS773 amino acids comparable to erlotinib.

      Conclusions:
      In conclusion, 1,3,5-triazines-monstrol conjugates has shown promising antitumor activity via inhibition EGFR-TK in lung carcinoma and my find serve as a lead for further research.

      Clinical trial identification:


      Legal entity responsible for the study:
      SHUATS

      Funding:
      SHUATS

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      35P - CDK7 inhibition as a promising therapeutic strategy in lung squamous cell carcinoma with SOX2 amplification (ID 188)

      12:30 - 12:30  |  Author(s): J.C. Lee, J.Y. Hur, J.K. Rho, H.R. Kim, C. Choi

      • Abstract

      Background:
      Molecular targeted therapy has much improved the survival of lung adenocarcinoma while few advances have been made in the treatment of lung squamous cell carcinoma (SCC). SOX2 amplification is one of the most common genetic alterations in SCC. We investigated the effects of THZ1, a potent inhibitor of cyclin-dependent kinase 7 (CDK7) which plays a key role in gene transcription, on SCC cell lines with SOX2 amplification.

      Methods:
      SOX2-amplified SCC cell lines (H520, H1703, HCC95) and SCC cell lines without SOX2 amplification (H226, SK-MES1, H1975) were used. Along with the general drug efficacy of THZ1 and the effect of SOX2 knockout by CRISPR on cell growth, the differential gene expression following THZ1 treatment was evaluated by microarray analysis.

      Results:
      Genetic inhibition of SOX2 by CRISPR resulted in the suppression of cell growth in SOX2-amplified SCC cell lines. The treatment of THZ1 led to the suppression of cell growth and apoptotic cell death only in SOX2-amplified SCC cell lines while the modest growth-inhibitory effect of cisplatin was not different according to the status of SOX2 amplification. The phosphorylation of carboxyl-terminal domain (CTD) of RNAPII and the expression of SOX2 and survivin were decreased by THZ1 in Western blotting. Accordingly, the expression of transcription-associated genes including SOX2 was down-regulated by THZ1 in SOX2-amplified SCC cell lines.

      Conclusions:
      THZ1 could effectively control the proliferation and survival of SOX2-amplified SCC cells with the decrease of global transcriptional activity. It suggests that CDK7 inhibition leading to suppression of transcription may be considered as one of promising therapeutic options in lung SCC with SOX2 amplification.

      Clinical trial identification:


      Legal entity responsible for the study:
      Jae Cheol Lee

      Funding:
      Asan Medical Center

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      36P - Circulating tumor DNA (ctDNA) as predictive biomarker in NSCLC patients treated with nivolumab (ID 401)

      12:30 - 12:30  |  Author(s): F. Passiglia, A. Perez, A. Listì, M. Castiglia, E. Musso, C. Ancona, S. Rizzo, M. Alù, L. Blasi, A. Russo

      • Abstract

      Background:
      Nivolumab is a programmed death-1 (PD-1) inhibitor recently approved for the treatment of NSCLC patients who failed prior chemotherapy. Searching for predictive biomarkers of immunotherapy efficacy is an area of intensive investigation for translational research. Monitoring circulating tumor DNA (ctDNA) during nivolumab treatment could help clinicians to predict the immunotherapy efficacy and ultimately improve the management of patients.

      Methods:
      From August 2015 to January 2017, 25 NSCLC patients receiving intravenous nivolumab 3 mg/kg every two weeks were included within a translational study. All the patients underwent CT-scan every 6 cycles and responses were evaluated by Response Evaluation Criteria in Solid Tumors (RECIST). Peripheral blood samples were obtained from the patients at baseline and after 4 cycles of therapy and the quantification of ctDNA (ng/µL plasma) was performed by qubit dsDNA HS assay and confirmed by qPCR evaluating a 115 bp fragment of ALU repeat. The Mann Whitney test was used for intergroup comparisons of two independent samples. A p-value <0.05 was used as a threshold for statistical significance. Survival analysis was performed using Kaplan–Meier method, providing median and p-value.

      Results:
      Among the 20 patients included, 13 experienced progression disease (PD) at the first CT-scan, 3 partial response (PR), 4 stable disease (SD); 7 patients are still alive at the time of analysis. In 20 patients evaluable for ctDNA analysis at baseline, median ctDNA was significantly higher in patients with time to progression (TTP) < 3 months as compared to TTP > 3 months (p: 0.03). Similarly, in 17 patients evaluable for ctDNA analysis after 4 cycles, median ctDNA was significantly higher in patients with time to progression (TTP) < 3 months as compared to TTP > 3 months (p: 0.042). A not significant trend toward a TTP benefit was observed in patients with baseline ctDNA < 0.49 ng/µL as compared to ctDNA > 0.49 ng/µL (5 vs 3 months; p: 0.06); however, ctDNA after 4 cycles < 0.56 ng/µL was significantly associated with an improved TTP as compared to ctDNA > 0.56 ng/µL (p: 0.05). No significant differences in overall survival (OS) have been observed between these subgroups of patients.

      Conclusions:
      The preliminary results of this study showed that high ctDNA plasma levels are associated with early PD in NSCLC treated with nivolumab, suggesting a potential role of ctDNA as early predictor of response to immunotherapy. These preliminary data need to be explored and confirmed by prospective studies including larger cohorts of patients.

      Clinical trial identification:
      N.A

      Legal entity responsible for the study:
      Palermo University Hospital

      Funding:
      Palermo University Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      37P - Can we perform subtyping of non-small cell lung cancer patients by use of lung tissue metabolic fingerprinting? (ID 477)

      12:30 - 12:30  |  Author(s): M. Ciborowski, J. Kisluk, K. Pietrowska, P. Samczuk, E. Parfieniuk, T. Kowalczyk, M. Kozlowski, A. Kretowski, J. Niklinski

      • Abstract

      Background:
      In case of NSCLC patients targeted agents (TAs) and newer chemotherapeutics yielded differences in outcomes according to histologic subgroups. About 62% of adenocarcinoma (ADC) patients have driver mutations in targeted oncogenes and TAs can be successfully administrated to these patients. Similar mutations are rarely observed in squamous cell carcinoma (SCC) and TAs are currently unavailable for this group of patients. A correct histologic classification is important for treatment planning and novel therapeutic options are awaited for SCC patients. Metabolomics has potential for revealing metabolic pathways altered by disease and for biomarkers discovery. The purpose of this study was to evaluate whether LC-MS metabolomics of lung tumor tissue can be used to classify NSCLC patients according to histological subtype: ADC, SCC or large cell carcinoma (LCC).

      Methods:
      The study was performed in the group of 25 NSCLC patients (62±8 years old, BMI=25±2, 32% females) who underwent curative tumor surgery. The patients were classified as ADC (n = 8), LCC (n = 4), and SCC (n = 13). Metabolites were extracted from tumor and adjacent control lung tissue and analyzed by LC-QTOF-MS. Differences between tumor and control tissue were evaluated by paired t-test or Wilcoxon rank test. One-way ANOVA was used to select metabolites discriminating NSCLC subtypes. Multivariate analysis was used for samples classification.

      Results:
      Control and tumor tissue samples were well separated on OPLS-DA model (R2=0.96, Q2=0.86). The main metabolites responsible for separation were lysophospholipids (+51-384%, p-value 0.02-0.0004) and acylcarnitines (+59-477%, 0.05-0.0007). PLS-DA model showed good separation of NSCLC subtypes (R2=0.99, Q2=0.7). Among metabolites responsible for this separation sphingosine (p = 0.03), four acylcarnitines (p-value 0.03-0.001), Lyso PEs 18:1 (p = 0.04) and 16:0 (p = 0.03) as well as Lyso PC 16:1 (p = 0.01) can be mentioned.

      Conclusions:
      NSCLC subtypes can be discriminated based on tumor tissue metabolic fingerprinting. Obtained results are preliminary and require further validation in a larger group of patients.

      Clinical trial identification:
      This study is a research project which was approved by the Ethics Committee of the Medical University of Bialystok (No. R-I-003/262/2004).

      Legal entity responsible for the study:
      Medical University of Bialystok

      Funding:
      National Science Centre, Poland (2014/13/B/NZ5/01256).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      38P - Flow cytometry immunophenotyping of pleural fluid cytology in patients with diffuse large B-cell lymphoma (DLBCL) and malignant pleural effusion (ID 377)

      12:30 - 12:30  |  Author(s): F. Lumachi, P. Ubiali, R. Tozzoli, S.C. Sulfaro, S.M. Basso

      • Abstract

      Background:
      Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of NHL and comprises up to 35% of cases. In patients with DLBCL the clinical presentation varies widely. Malignant pleural effusion (MPE) represents a common occurrence in cancer patients with either thoracic and extra-thoracic malignancies, often requiring invasive diagnostic procedures, including video-assisted thoracoscopic surgery (VATS)-guided biopsy. Approximately 20% of patients with DLBCL may develop MPE. The aim of this study was to demonstrate the usefulness of flow cytometry phenotyping (FCP) of pleural cytology (PC) in patients with MPE due to DLBCL.

      Methods:
      A retrospective chart review of 40 (15 (37.5%) males and 25 (62.5%) females, median age 69 tears, range 46-85 years) patients with histologically confirmed MPE was performed at a tertiary referral Medical Center. All patients underwent PC and FCP before VATS-guided biopsy. For the FCP, the FACSCanto II system and the FACSDiva software (BD Biosciences, Franklin Lakes, USA) were used. Compensation beads were obtained with single stains of each antibody to determine the compensation settings, and the settings were applied in FlowJo software (Tree Star, Ashland, USA) after data collection. Sample cells were stained with a panel of antibodies, including CD5, CD10, CD19, CD23, Kappa, Lambda, and CD45 (Dako, Glostrup, Denmark). In the presence of a T-cell population with aberrant phenotypes or a B-cell population with monotypic light chains, the diagnosis of DLBCL-related pleural effusion was made.

      Results:
      Final histology showed 5 (12.5%) patients with DLBCL and 35 (87.5%) with MPE from other malignancies (mainly breast cancer and lung cancer). The age (68.0±10.1 vs. 67.5±14.2 years; p = 0.94) and the male to female ratio (p = 0.64) did not differ between groups. The results are reported in the Table. The sensitivity and negative predictive value of FCP reached 100%, the specificity was the same as obtained with the PC alone, but the accuracy was superior (97.5% vs. 85%; p = 0.003).rnTable: 38PResults of flow cytometry phenotyping and pleural cytologyrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      ResultsFlow cytometry immunophenotypingPleural fluid cytology
      Sensitivity100%0%
      Specificity97.1% (95% CI: 91.6-100)97.1% (95% CI: 91.6-100)
      Negative predictive value100%97.2%
      Clinical accuracy97.5%85.0%
      Prevalence12.5%12.5%
      False positive rate2.9%2.9%
      False negative rate0%14.3%
      Likelihood ratio positive350
      Likelihood ratio negative01.3
      rn

      Conclusions:
      FCP of PC has great value in confirming (or excluding) the relationship between pleural effusion and malignancy, including lymphoma, exhibiting a significantly higher accuracy that that of PC alone. FCP should be performed in all patients with a history of DLBCL who exhibit suspicious MPE.

      Clinical trial identification:


      Legal entity responsible for the study:
      Università degli Studi di Padova

      Funding:
      Università degli Studi di Padova

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      39P - Efficacy of Tumor Treating Fields (TTFields) and anti-PD-1 in non-small cell lung cancer (NSCLC) preclinical models (ID 457)

      12:30 - 12:30  |  Author(s): U. Weinberg, T. Voloshin, O.T. Yitzaki, N. Kaynan, M. Giladi, A. Shteingauz, M. Munster, S. Cahal, E.D. Kirson, Y. Palti

      • Abstract

      Background:
      Tumor Treating Fields (TTFields) are an effective anti-neoplastic treatment modality delivered via non-invasive application of low intensity, intermediate frequency, alternating electric fields. TTFields is approved for the treatment of both newly diagnosed and recurrent glioblastoma. TTFields interrupt mitosis in cancer cells by disrupting microtubules and septin filaments, which play key roles in mitosis. The mitotic effects of TTFields include abnormal chromosome segregation that trigger different forms of cell death. We evaluated TTFields’ effect on immunogenic cell death and its efficacy when combined with an immune checkpoint inhibitor (αPD1) in NSCLC.

      Methods:
      Murine Lewis lung carcinoma (LLC) cells were treated with TTFields using the inovitro[TM] system. Levels of cell surface calreticulin (CRT) and intracellular ATP levels were evaluated using flow cytometry. High mobility group box 1 (HMGB1) secretion was measured using an ELISA assay. Mice inoculated with LLC cells were treated with isotype control, TTFields, αPD-1, or TTFields + αPD-1. Tumor volume monitoring and intra-tumor immune cell profiling were performed.

      Results:
      TTFields induced elevated cell surface expression of CRT, decreased ATP levels, and promoted HMGB1 secretion. In vivo, the combined treatment of TTFields + α-PD-1 led to a significant decrease in lung tumor volume compared to all three other groups (P < 0.001). Significant increase in CD45+ tumor infiltrating cells was observed in the TTFields + αPD-1-treated mice. Infiltrating cells demonstrated a significant upregulation of surface PD-L1 expression. Both F4/80+CD11b+ cells and CD11c+ cells exhibited higher tumor infiltration and elevated PD-L1 expression, as compared to the control group. These findings indicate enhanced inflammatory antitumor environment conferred by the combination of TTFields + αPD-1.

      Conclusions:
      Our results demonstrate that TTFields treatment potentiates immunogenic cell death in NSCLC cancer cells. Combining TTFields with specific immunotherapies such as anti-PD-1 may enhance antitumor immunity and result in increased tumor control. A phase III clinical study on TTFields in combination with either PD-1 inhibitors or docetaxel in NSCLC is underway.

      Clinical trial identification:
      Not applicable. Preclinical research.

      Legal entity responsible for the study:
      Novocure Ltd

      Funding:
      Novocure Ltd

      Disclosure:
      U. Weinberg: Full time employee of Novocure. T. Voloshin, O.T. Yitzaki, N. Kaynan, M. Giladi, A. Shteingauz, M. Munster, S. Cahal, E.D. Kirson: Full time employee of Novocure Ltd. Y. Palti: Shareholder at Novocure Ltd, NY.

    • +

      40P - Antitumoral effect and reduced systemic toxicity in mice after intra-tumoral injection of an in vivo solidifying calcium sulfate formulation with docetaxel (ID 201)

      12:30 - 12:30  |  Author(s): M. Sandelin, S. Grudén, V. Rasanen, P. Micke, M. Hedeland, N. Axén, M. Jeansson

      • Abstract

      Background:
      Docetaxel is a cytostatic agent approved for treatment of non-small cell lung cancer as well as other cancers. Its effectiveness in clinical practice is associated with a variety of acute and long term side effects. To reduce the systemic side effects, a local slow-release depot formulation based on calcium sulfate with docetaxel for intra-tumoral administration was developed.

      Methods:
      Two formulations with a twofold difference in docetaxel drug load (corresponding to 12.5 and 25.0 mg/kg body weight) were manufactured with the calcium sulfate based NanoZolid™ technology. The formulations were injected intra-tumorally as a paste which solidified within the tumor. The effects of the two local depot formulations were tested in female mice (n = 60) inoculated with subcutaneous Lewis lung carcinoma cells. The slow-release depots were compared to systemic intraperitoneal injections of docetaxel (25.0 mg/kg) and a calcium sulfate placebo formulation. Tumor volumes were measured and systemic side effects were evaluated using body weight and cell counts from whole blood as well as plasma concentrations. In addition, a histological evaluation was performed.

      Results:
      Both docetaxel formulations showed a significantly higher antitumor efficacy compared to placebo, which was comparable to that of systemic administration of docetaxel. Moreover, the intra-tumoral depot formulations with docetaxel showed reduced systemic toxicity compared to systemic treatment, including less weight loss and no decrease in blood cell counts. The extracellular deposits of calcium sulfate, observed in all groups with intra-tumoral injections, did not lead to significant inflammatory changes.

      Conclusions:
      It was shown that intra-tumoral slow-release depots of calcium sulfate with docetaxel were well tolerated and released docetaxel at amounts sufficient for satisfactory local antitumor effect and for a low level plasma detection. The use of such depots can be an alternative to intravenous injection as an efficient antitumoral treatment with reduced systemic toxicity.

      Clinical trial identification:


      Legal entity responsible for the study:
      Uppsala University, Department of Immunology, Genetics and Pathology, Marie Jeansson

      Funding:
      This study was supported by an IGP Young Investigator grant (MJ), the Swedish Research Council (521-2012-865, MJ), Åke Wiberg Foundation (738866289, MJ), Magnus Bergvalls Foundation (24942-1-2013, 2014-00055, MJ), and Ture Stenholms Foundation for Surgical Research (MS)

      Disclosure:
      S. Grudén: Consultant for LIDDS AB. N. Axén: Consultant for LIDDS AB and have equity interests in LIDDS AB. All other authors have declared no conflicts of interest.

    • +

      41P - Development of novel 1,2,4-triazole-thiol derivatives as dual PI3K/mTOR inhibitor against the non-small cell lung cancer (NSCLC) (ID 435)

      12:30 - 12:30  |  Author(s): A. Verma, V. Kumar, U.P. Singh

      • Abstract

      Background:
      PI3K catalyzes the ATP-dependent phosphorylation of the 3'-hydroxyl group of phosphatidylinositols and plays an important role in cell growth and survival. However, most of the inhibitors in this signaling pathway have inhibitory effect against both PI3K and mammalian target of rapamycin (mTOR) kinases. It is well known that the mTOR, regulate cell proliferation, apoptosis, angiogenesis, and metabolism through both AKT-dependent and AKT-independent mechanisms. Agents targeting PI3Ks with additional mTOR kinase inhibitory have more advantageous than alone inhibition. Our efforts to identify potent and efficacious PI3K/mTOR dual inhibitors resulted in the discovery of a series of substituted 1,2,4-triazole-thiol derivatives.

      Methods:
      The entire set of the designed compounds were assessed on the Lipinski rule of five for drug-likeness prediction using Molinspiration and synthesized via novel synthetic procedure. The anticancer activity was evaluated against panel of three human NSCLC cell lines A549, H157 and H52 together with effect on cell cycle. The compounds were also evaluated for inhibitory activity against PI3K and mTOR via kinase inhibitory assay together with molecular docking experiments (PI3K; 3s2a.pdb) to rationalize the mechanism of action.

      Results:
      The compounds were developed in excellent yield and found to be significantly activity against the panel of the cell lines. The most active compound 6b showed significant inhibitory activity with IC50 of 4.56, 7.23 and 9.21 µmol/l against A549, H157 and H52, respectively, with highest activity against A549 (IC50 = 1.13 µmol/l). The compound 6b causes a significant arrest of G2/M phase and induces tumor cell apoptosis in a concentration dependent manner. In a PI3K/mTOR inhibition assay, 6b showed IC50 of 1.43±0.21 µM and 2.32±0.12 against PI3Ks and mTOR, respectively. Moreover, the docking results showed that 6b efficiently inhibit PI3Ks via channeling in ATP pocket with Ki of 436.57 nM. It showed to interact with Met804, ILE831, ILE879.

      Conclusions:
      In conclusion, a novel series of 1,2,4-triazole-thiol derivatives has been developed as potent inhibitor of PI3K/mTOR against lung cancer with favorable drug likeness profile.

      Clinical trial identification:


      Legal entity responsible for the study:
      SHUATS

      Funding:
      SHUATS

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      42P - Dual inhibitory effects of novel naringenin analogue in tobacco-carcinogen induced lung cancer via inhibition of PI3K/Akt/mTOR pathway (ID 337)

      12:30 - 12:30  |  Author(s): V. Kumar, A. Verma, P.C. Bhatt

      • Abstract

      Background:
      Phosphoinositide 3- kinase (PI3K)-AKT-mammalian target of rapamycin (mTOR) pathways is considered as the signalling pathway which activates the diverse cellular function viz., survival, cell expansion, vesicular transport and proliferation and found frequently dysregulated pathway in lung cancer. Consequently, flavonoids based inhibitors play a key kinase role in the pathway including mTOR, PI3K and AKT, have been extensively scrutinized in targeting the oncology in recent years. The common pathway to PI3K-Akt-mTOR used to target during the lung cancer therapy. Therefore, the current study was aimed to peruse the naringenin as dual PI3K/mTOR for lung cancer.

      Methods:
      In the current experimental study mice were randomly divided into 7 groups with 12 mice in each groups The oxidative stress was evaluated in term of antioxidant parameters such as myeloperoxidase (MPO) and superoxide dismutase (SOD), respectively. The proinflammatory cytokines viz., interleukin (IL)-1β, tumor necrosis factor-α (TNF-α) and interleukin (IL)-6 were measured via using the standard enzyme-linked immunosorbent assay kits. The concentration of PI3K, P-PI3K, mTOR, P-mTOR Akt and P-Akt were determined via using the Western blot techniques. We also performed the histopathological study to identify the changes during the disease.

      Results:
      Naringenin significantly suppressed the oxidative stress via improving the status of endogenous antioxidant parameters such as SOD and MPO in a dose dependent manner. disease control group mice confirmed the change in protein levels of PI3K/Akt/mTOR pathway in lung as compared to normal control, which were significantly down-regulated by the naringenin in a dose dependent manner. In the histological study, we observed that the naringenin substantially reduced the benzopyrene induced neutrophils in lung tissue.

      Conclusions:
      It can be concluded that naringenin has shown promising anticancer effect via attenuation of PI3K/Akt/mTOR against lung cancer and signifies the potential therapeutic relevance for further development.

      Clinical trial identification:


      Legal entity responsible for the study:
      SHUATS

      Funding:
      SHUATS

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      43P - A phase I-II clinical trial to evaluate the safety, pharmacokinetics and efficacy of high dose intravenous ascorbic acid synergy with mEHT in Chinese patients with stage III-IV non-small cell lung cancer (ID 225)

      12:30 - 12:30  |  Author(s): J. Ou, X. Zhu, Y. Lu, C. Zhao, H. Zhang, X. Gui, X. Wang, X. Zhang, T. Zhang, C.L.K. Pang

      • Abstract

      Background:
      Ascorbic acid (AA) infusion and modulated electrohyperthermia (mEHT) have been widely used by integrative cancer practitioners for many years. However, there are no safety and pharmacokinetics data in Chinese cancer patients.

      Methods:
      Blood ascorbic acid in the fasting state was obtained from 35 NSCLC patients; selecting from them 15 patients with stage III-IV entered the phase I -II study. They were randomized allocated into 3 groups, and received doses 1.0, 1.2, 1.5g/kgAA infusions. Pharmacokinetic profiles were obtained when they received solely IVAA at concentrations of 1g/kg, 1.2g/kg, and 1.5g/kg, and when IVAA in combination with mEHT. The process was applied 3 times a week (every other day, weekend days off) for 4 weeks. Participants in the first group received intravenous AA (IVAA) when mEHT was finished; in the second group IVAA was administered simultaneously with mEHT; and in the third group IVAA was applied first, and followed with mEHT. All patients started the trial doses of 1 g/kg for 8 treatments. When there was no DLT observed, the test dose for patients increased to 1.2 g/kg continuously for 8 treatments, and then the test dose for patients was escalated to 1.5 g/kg continuously for 8 treatments. DLT was defined as any reversible grade ≥3 adverse events, whether haematological or non-haematological.

      Results:
      Fasting plasma AA levels were significantly correlated with stage of the disease. Peak concentrations of AA were significantly higher in the simultaneous treatments than in other combinations with mEHT or in solely IVAA-managed groups. The average scores for the functioning scales continuously increased and the symptoms gradually decreased over the full cycle of the study. 3/7 patients with squamous cell lung cancer were CR, 4/7 were PR. 1/8 patient with adenocarcinoma cell lung cancer was PR, 5/8 were SD, and 2/8 died.

      Conclusions:
      IVAA synergies with simultaneous mEHT is safe and the concomitant application significantly increases the plasma AA level for NSCLC patients. QOL is improved when they receive the above treatments. Patients diagnosed with squamous cell lung cancer were sensitive to the above two treatments.

      Clinical trial identification:
      NCT02655913

      Legal entity responsible for the study:
      Junwen Ou

      Funding:
      Clifford Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      47P - Multi detector CT evaluation of suspicious malignant lung masses with its combined wash-in & wash-out features and their histopathological correlation (ID 237)

      12:30 - 12:30  |  Author(s): S. Khanduri, S. Bhagat

      • Abstract

      Background:
      Lung cancer is the leading cause of cancer death in the world. Detection of malignancy at an early stage and with precision is the utmost objective of radiological evaluation. The final diagnosis of lung cancer is histopathological evaluation of the mass by biopsy or fine needle aspiration cytology (FNAC) only. This study was carried out with an aim of evaluating the suspected malignant masses by Multidetector CT (MDCT) along with their contrast wash-in & wash-out characterisctics with their histopathological correlation.

      Methods:
      After obtaining ethical approval, 50 cases of suspected lung cancer, by clinical and radiographic evaluation, were enrolled in the study. These patients underwent CT thorax (non contrast, contrast & delayed scans) on 384 slice Siemens Somatom Force®. After undergoing the radiological evaluation, biopsy of the mass was performed either using CT guidance or bronchoscopy guidance. Radiological and histopathological findings were correlated.

      Results:
      Sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV) and diagnostic accuracy of MDCT against histopathology were 97.3%, 76.9%, 92.3%, 90.9%, and 92.0%, respectively. Sensitivity and specificity calculated through receiver operating characteristic curve for wash-in (Net early enhancement) for >17.00 HU were found to be 97.3% and 76.9% while that of wash out characterization of lung mass - Relative wash-out at < 18.83% were 97.3% and 93.3%, and absolute washout at < 65.48% were 97.3% and 93.3% in predicting malignancy.

      Conclusions:
      MDCT serves as an excellent tool for early diagnosis of lung cancer and it is an important tool for cases where biopsy or FNAC is not possible. Faster and newer CT techniques such as contrast wash-in and wash out have greater diagnostic accuracy than the conventional diagnostic techniques. So a combination of conventional and newer techniques can further increase the diagnostic accuracy of MDCT in diagnosing lung cancers and obviate the need for invasive methods for biopsy/FNAC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Era\'s Lucknow Medical College & Hospital

      Funding:
      Era\'s Lucknow Medical College & Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      48P - Evaluation of neoplasms incidentally detected by preoperative FDG-PET/CT for primary lung cancer (ID 362)

      12:30 - 12:30  |  Author(s): Y. Noda, H. Matsudaira, S. Arakawa, K. Yoshida, T. Morikawa

      • Abstract

      Background:
      Fluorodeoxyglucose Positron emission tomography/computed tomography (FDG-PET/CT) is an effective modality generally used in preoperative evaluation for primary lung cancer. In some cases, FDG-PET/CT found incidental neoplastic lesions and also detected unexpected pre-malignancy or second malignancy. Our study aimed to examine incidental findings detected by preoperative PET/CT and its impact on management for lung cancer.

      Methods:
      From April 2010 until August 2015, 149 patients underwent surgery for primary lung cancer at Katsushika Medical Center and 111 patients of them acquired preoperative PET/CT were evaluated retrospectively. Incidental findings were followed up by some imaging test, clinician feedback, pathological examination and various endoscopy.

      Results:
      The objects included 32 women and 79 men. The mean age was 70.6±8.46 years old. Excluding known diseases and thoracic cage lesions, abnormal FDG uptakes on PET/CT observed at 40 patients (36%). 8 cases (7.2%) of them required other modalities for further evaluation and the sites of abnormal uptake were prostate, larynx, thyroid gland, 3 cases of colon, bone and lymph node each.3 cases (2.7%) of colon had an indication for treatment. 2 cases were colon polyp and the other case was colon cancer. 2 cases underwent endoscopic mucosal resection. The other case underwent surgery for the colon cancer before treatment for the lung cancer because of neoplastic obstruction. This case was pointed out liver metastasis 6 months later from surgery of the colon cancer. We diagnosed liver lesion as colon cancer metastasis and we are doing chemotherapy to this case.

      Conclusions:
      A malignant or pre-malignant lesion was found in 0.9% and 1.8%, respectively. Colon was major lesion of abnormal uptake of FDG in our study, and 1 case required colon resection before lung cancer. The case had a significant change in management for treatment. We conclude preoperative FDG PET/CT might be useful modality in identifying second primary cancers or pre-malignancy lesions in patients with primary lung cancer. Further investigations are needed to evaluate incidental findings detected by preoperative FDG PET/CT.

      Clinical trial identification:


      Legal entity responsible for the study:
      Jikei University School of Medicine

      Funding:
      Jikei University School of Medicine

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      49P - Computed tomographic features of pulmonary sarcomatoid carcinoma (ID 499)

      12:30 - 12:30  |  Author(s): M. Attia, K. Abdelmoula, H. Neji, M. Affes, S. Hantous-Zannad, A. Mekkaoui, I. Baccouche, K. Ben Miled-M'rad

      • Abstract

      Background:
      Pulmonary sarcomatoid carcinoma is a rare tumor. It is subdivided into 5 subtypes and representing approximately 0.4% of non-small cell lung cancer. The aim of our work is to illustrate the computed tomography (CT) features of this tumor.

      Methods:
      This was a retrospective case series of 18 consecutive patients with a pulmonary sarcomatoid carcinoma, collected between January 2011 and December 2016. All patients had undergone a CT scan for the diagnosis, staging and therapeutic response evaluation. CT- guided percutaneous biopsy and a histological analysis were performed in all cases for the pathological confirmation.

      Results:
      The mean age of patients was 56.8 years (range, 46-73 years). The CT appearance of the tumor was similar for all our patients: Large mass, peripheral, heterogeneous density, with massive necrotic tissue component that strongly heightened after contrast injection; tumor had a locoregional extension with parietal and bone invasion (n = 5) and a distance extension (n = 4) to the adrenal glands. Pathological examination found a pleomorphic carcinoma (n = 3), a giant cell carcinoma (n = 1) and a carcinosarcoma (n = 2). Sarcomatoid carcinoma diagnosis was increased for the remaining patients without precision of histological subtype.

      Conclusions:
      Pulmonary sarcomatoid carcinoma seems to have CT scan characteristics that distinguish it from other non-small cell lung cancers, specifically: diagnosis size, peripheral necrosis and increased vascularization of its fleshy portion with a local aggressiveness and a high metastatic potential.

      Clinical trial identification:


      Legal entity responsible for the study:
      Abderrahmen Mami Hospital

      Funding:
      Abderrahmen Mami Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      50P - Usefulness of F-18 FDG PET/CT to identify metastatic mediastinal lymph node in NSCLC patients with bilateral benign mediastinal lymph node hyperplasia (ID 272)

      12:30 - 12:30  |  Author(s): Y.S. Kim, S.H. Yoon, B.S. Son, D.H. Kim, K. Kim, S. Kim

      • Abstract

      Background:
      F-18 FDG PET/CT is superior to CT imaging in identifying metastatic mediastinal lymph node (MLN) involvement in NSCLC. However, the accuracy of F-18 FDG PET in nodal staging is substantially reduced by false increases in F-18 FDG uptake in the inflammatory nodes. The efficacy of this technique is controversial because of reactively increased F-18 FDG uptake in the mediastinum, especially in regions endemic for the granulomatous disease. Here, we determined the qualitative and quantitative parameters of F-18 FDG-PET/CT for evaluation of mediastinal nodes to distinguish between malignant and benign lesions.

      Methods:
      27 patients with pathologically documented NSCLC who showed bilateral F-18 FDG-avid MLNs on pre-treatment staging PET/CT image were included. 104 MLNs were pathologically analysed through EBUS-TBNA or MLN dissection. The MLN with prominent F-18 FDG uptake compared with other F-18 FDG-avid LNs via visual assessment was qualitatively considered as metastasis. The maximum standardized uptake value (SUVmax) of LN and SUVmax of LN to the liver and blood pool were calculated for quantifying F-18 FDG uptake. SUVmax/contra was obtained to identify metastatic MLNs.

      Results:
      The qualitative evaluation showed sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) for separating metastatic LN from benign reactive hyperplasia of 66.7%, 87.0%, 74.3%, and 88.2%, respectively. SUVmax of LN and normalized SUVmax of LN to the liver and blood pool did not significantly differ; however, SUVmax/contra was significantly higher for metastatic LNs than benign lesions. Receiver-operating-characteristic derived SUVmax/contra cut-off was 1.18 (AUC, 0.734). The quantitative evaluation showed sensitivity, specificity, PPV, and NPV of 66.7%, 75.3%, 77.4%, and 86.4%, respectively.

      Conclusions:
      Qualitative interpretation of F-18 FDG PET/CT was superior to quantitative parameters in discriminating metastatic LNs from benign reactive hyperplasia in patients with bilateral F-18 FDG-avid mediastinal LNs. We suggest SUVmax/contra may aid in the interpretation of mediastinal nodal staging in patients with NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      None

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      52P - Combined chemotherapy with etoposide, irinotecan plus cisplatin compared with topotecan monotherapy as the second-line treatment in sensitive relapsed small cell lung cancer: A retrospective analysis (ID 308)

      12:30 - 12:30  |  Author(s): H. Wang

      • Abstract

      Background:
      Topotecan is currently the only drug approved by US Food and Drug Administration (FDA) for treatment of relapsed small-cell lung cancer (SCLC) patients. We did this study to attempt to investigate whether combined chemotherapy with etoposide, irinotecan plus cisplatin (PEI) was superior to topotecan alone as second-line treatment for sensitive relapsed SCLC patients.

      Methods:
      Between January 2009 and November 2015, a total of 82 relapsed SCLC patients were admitted to the affiliated Cancer Hospital of Zhengzhou University in this retrospective analysis. Among them, 44 patients were treated with combined PEI chemotherapy, while 38 patients were treated with topotecan monotherapy. Disease control rates (DCR), progression-free survival (PFS), overall survival (OS) and adverse reaction were recorded.

      Results:
      A total of 82 patients were analyzed. The patients treated with combined chemotherapy had longer median overall survival (OS) (16.3 months vs 10.1 months) than those treated with topotecan (P = 0.001), and median progression-free survival (PFS) time was slightly but significantly longer (6.2 months vs 4.1 months), than the topotecan group. Compared with the topotecan group, more common grade 3 or 4 adverse events were recorded for the combination chemotherapy group, including leucopenia, anaemia, thrombocytopenia. Grade 3-4 non-hematological toxicity was not common. When compared with younger patients, combined chemotherapy-treated patients (≥ 68 years of age) experienced higher rates of leucopenia, anemia, thrombocytopenia and fatigue. Serious adverse events were reported in two patients in the topotecan group and seven patients in the combination group. Two treatment-related deaths (one each of pulmonary infection and pneumonitis) occurred in the combination group, one treatment-related death (pneumonitis) occurred in the topotecan group.

      Conclusions:
      Combined chemotherapy with etoposide, irinotecan plus cisplatin (PEI) is not inferior to topotecan monotherapy as the second-line treatment.

      Clinical trial identification:
      none

      Legal entity responsible for the study:
      None

      Funding:
      The National Natural Science Foundation of China (No.81272600)

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      53P - Systematic literature reviews of second and third-line treatments used for small-cell lung cancer (SCLC) (ID 246)

      12:30 - 12:30  |  Author(s): R. Goulding, M. Lorenzi, Y. Yuan, S. Bobiak, N. Hertel, B. Korytowsky, J.R. Penrod, J. Jansen

      • Abstract

      Background:
      While SCLC is initially sensitive to first-line chemotherapy and radiotherapy, most patients relapse, and outcomes with second-line treatments remain bleak. The purpose of this study was to gather evidence and assess appropriate methodologies for evidence synthesis of the following clinically relevant outcomes: efficacy, safety, and health-related quality of life (HRQoL) in adult patients treated with existing second and third-line SCLC treatments, including immunotherapy, which was recently recommended in the NCCN SCLC treatment guidelines.

      Methods:
      A systematic literature search was conducted using MEDLINE, EMBASE, and the Cochrane Register of Controlled Clinical Trials, and carried out by 2 independent reviewers. Manual searches of clinicaltrials.gov, the WHO International Clinical Trials Registry Platform, and 5 relevant conferences (ASCO, AACR, ESMO, WCLC, and ELCC) were performed to identify unpublished randomized clinical trials (RCTs) potentially eligible for inclusion. RCTs that reported outcomes of interest, including overall survival, progression-free survival, objective response rate, and/or HRQoL, for the treatments and population of interest, were identified for this review. Corresponding trial, patient baseline characteristics, and outcome data were extracted for all included studies.

      Results:
      17,550 citations were identified, including 970 conference abstracts and 25 clinical trial registry entries. Nine SCLC RCTs were identified studying treatments of interest, including cisplatin, carboplatin, etoposide, amrubicin, irinotecan, gemcitabine, and topotecan; however, there were a lack of RCTs for innovative treatments including nivolumab.

      Conclusions:
      A lack of RCT evidence for all relevant treatments in the target population may result in disconnected treatment networks, such that trial comparisons involving all treatments of interest may not be feasible using the standard network meta-analysis methodology. Alternative methods that incorporate single-arm evidence may be required to estimate the relative clinical efficacy, safety, and HRQoL outcomes of second- and third-line treatments of relevant SCLC comparators.

      Clinical trial identification:


      Legal entity responsible for the study:
      Bristol-Myers Squibb and Precision Health Economics

      Funding:
      Bristol-Myers Squibb

      Disclosure:
      R. Goulding: Employee of Precision Health Economics, funding from Bristol-Myers Squibb for the work on the abstract. Y. Yuan, S. Bobiak, N. Hertel, B. Korytowsky, J.R. Penrod: Employee of Bristol-Myers Squibb with stock/ownership options. J. Jansen: Employee of Precision Health Economics and reports that Precision Health Economics received funding for this work. All other authors have declared no conflicts of interest.

    • +

      54P - Up front chemotherapy dosing and relative dose intensity in extensive stage small cell lung cancer (ID 344)

      12:30 - 12:30  |  Author(s): A.W. Mølby, B.E. Laursen, U. Falkmer, T. McCulloch, N.A. Jensen, L.Ø. Poulsen

      • Abstract

      Background:
      Extensive-stage small cell lung cancer (ES-SCLC) is an extremely aggressive malignant disease where patients often present with a poor performance status (PS), high age and several comorbidities. Reduced dose up front is a possibility for these patients. We investigated the impact of reduced dose up front and the impact of Relative dose intensity (RDI), age and comorbidity on overall survival (OS).

      Methods:
      Data of a real-time registration study of a consecutive cohort, of patients treated with carboplatin and etoposide for ES-SCLC during 2012 and 2013 in the North Denmark Region, were analyzed. Patients only treated with one cycle of chemotherapy were excluded. Comorbidity was assessed according to the Charlson Comorbidity Index (CCI). Area Under the Curve (AUC), for carboplatin dose, was used for analysis of RDI. All dose reductions were made simultaneously in both carboplatin and etoposide. Multivariate statistical survival analysis was made using Cox Regression analysis calculating a Hazard Ratio (HR) for OS.

      Results:
      In all, data from 75 patients were analyzed; mean age was 68 years; 44/75 (59%) were in PS 0-1 and 37/75 (49%) had 1 or more comorbidities. Reduced dose at baseline was applied to 25/75 (33%) of the patients. These patients had a HR 1.82, p-value = 0.070. RDI of 85% or below, was applied to 42/75 (56%) and showed a HR of 0.66, p-value = 0.228. Compared to PS 0-1, PS 2 showed a HR of 2.35 (p = 0.014) and PS 3-4 a HR of 8.40 (p < 0.000). Age, CCI, polypharmacy and LDH ≥ 300 U/L showed no statistical significant impact on OS. Hematological toxicity was the main reason for dose reductions and delays during chemotherapy, which occurred 19/75 (25%) and 47/75 (63%), respectively. Five patients died due to febrile neutropenia/sepsis. Fourteen long-term survivors were seen with an OS of more than 20 months.

      Conclusions:
      These results suggest that full dose chemotherapy should be applied up front, though poor PS is a major limiting factor, whereas high age should not be. RDI showed no impact on OS, but larger studies are needed. Data concerning fourteen long-term survivors will be presented at the meeting.

      Clinical trial identification:


      Legal entity responsible for the study:
      Aalborg University Hospital, Department of Oncology

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      55P - Clinical significance of EBUS-TBNA in patients with small cell lung cancer (ID 433)

      12:30 - 12:30  |  Author(s): Y.M. Lee

      • Abstract

      Background:
      Small cell lung cancer (SCLC) is associated with rapid tumor growth and early dissemination. A mass in or adjacent to the hilum/mediastinum is particularly characteristic of SCLC. Transbronchial needle aspiration (TBNA) is minimally invasive and safe method for the evaluation of lung masses. The diagnostic sensitivity of TBNA is particularly high for SCLC (64-93%). The addition of endobronchial ultrasound (EBUS) guidance to TBNA (EBUS-TBNA) has greatly enhanced the diagnostic yield and safety of the procedure. The aim of this retrospective study was to assess the clinical characteristics of SCLC diagnosed by EBUS-TBNA and its clinical significance.

      Methods:
      From January 2011 to December 2015, SCLC diagnosed by EBUS-TBNA were enrolled. For each patient, we documented patients characterisitics, size and locations of L/Ns, bronchoscopic findings and clinical courses. All patients underwent routine clinical assessment and a CT scan prior to EBUS-TBNA.

      Results:
      A total of 100 patients (64[64%] with limited-stage and 36[36%] with extensive-stage disease) were included in this study. Sex and Age distribution was M: F = 92.8 with a mean age 66.3±16.2 years. The most common chief complains were cough (n = 51), dyspnea (n = 18), chest pain (n = 15), blood tinged sputum (n = 10) and hoarseness (n = 6). The majority of patients was smokers (n = 95, 95%) and heavy smoker (>40pack.yr) was 74 (74%). 26 patients (26%) had hilar/mediastinal masses without a primary parenchymal lesion. Tumor marker CEA mean value was 16.4 in LD and 27.6 in ED. In bronchoscopy findings, 79 (79%) cases show only bronchial obstruction by external compression. The median size of target lesion examined by EBUS-TBNA was 33mm (range 11.5-74mm). Of the 100 EBUS-TBNA samples, 100(100%) samples yielded a diagnosis of SCLC. The most common target lymph node was at station 4R, followed by 7 and 11R. The median number and duration of aspirates per lesion was 2 (range 1-5) and 13mins (10-33). The most common distatant metastasis organ was bone (n = 30), follow by liver (n = 15), adrenal gland (n = 7) and brain (n = 6). The most common comorbidity was COPD (n = 34) followed by Heart disease (n = 15) and DM (n = 13) and DILD (n = 7). 30 patients of SCLC were not received chemotherapy. In chemotherapy group, overall response rate was 78.5% (LD: 88%, ED: 55%). No procedure-related complications were noted.

      Conclusions:
      EBUS-TBNA is a safe and highly accurate procedure for the diagnosis of SCLC. It is recommended a first diagnostic test if a patient is clinically suspected with SCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Busan Paik Hospital

      Funding:
      Busan Paik Hospital

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      56P - Pro-GRP in small cell lung cancer (ID 288)

      12:30 - 12:30  |  Author(s): S. Cavalieri, F. Nichetti, D. Morelli, F. de Braud, A. Martinetti, E. Sottotetti, K. Dotti, M. Prisciandaro, F. Corti, M. Platania

      • Abstract

      Background:
      NSE was historically the recommended tumor marker for SCLC. It stains up to 80% of SCLCs in tissue examinations but is also elevated in the sera of 20–30% of patients with NSCLC. NSE has low sensitivity, particularly in patients with limited disease (LD). Recently, pro-gastrin-releasing peptide (pro-GRP) became available as a sensitive, specific and reliable tumor marker for patients with SCLC.

      Methods:
      We retrospectively analyzed pro-GRP in pts with SCLC and NSCLC treated from 2015 to 2016 at our Center. Serum pro-GRP level was measured with electrochemiluminescence method at our laboratory; it was considered elevated if higher than 77.8 pg/ml. Statistical tests used to study differences between pro-GRP medians were Mann-Whitney U test for SCLC vs NSCLC and Fisher’s exact test for DD vs LD. To calculate sensitivity (Sn), specificity (Sp), positive and negative predictive values (PPV and NPV respectively) we considered: SCLC with elevated pro-GRP as true positive (TP), NSCLC with normal pro-GRP as true negative (TN), SCLC with normal pro-GRP as false negative (FN), NSCLC with elevated pro-GRP as false positive (FP).

      Results:
      A total of 46 pts were studied (33 men, 13 women), whose median age was 67 years (range 20-78). 20 pts had SCLC (16 DD, 4 LD) and 26 metastatic or unresectable advanced NSCLC. Median pro-GRP level was 1398 pg/ml (range 43-50000) in SCLC and 26 pg/ml (range 6-119.2) in NSCLC (difference 1372 pg/ml; SCLC/NSCLC ratio 53.76; p < 0.01). TPs were 18, FNs 2, TNs 25, FP 1. Sn was 90%, Sp 96.15%, PPV 94.73% and NPV 92.59%. All the TPs had diffuse disease, the 2 FNs had limited disease, the only FP had a poorly differentiated adenocarcinoma. Median pro-GRP was 182.25 pg/ml (range 43-642) in LD pts and 2522 pg/ml (range 88-50000) in those with DD (absolute difference 2339.75 pg/ml; ratio DD/LD 13.83; p = 0.026). Pro-GRP was elevated in all DD pts and in half of LD pts (2/4): Both of them had a high burden of loco-regional disease.

      Conclusions:
      Our data confirm that pro-GRP is sensitive for SCLC diagnosis and that it might help to discriminate DD from LD. Since high marker levels are related to a high disease burden, pro-GRP may have a negative prognostic significance. Follow up is required to define its role in clinical practice in monitoring response to treatment.

      Clinical trial identification:
      Not applicable

      Legal entity responsible for the study:
      Fondazione IRCCS Istituto Nazionale dei Tumori

      Funding:
      Fondazione IRCCS Istituto Nazionale dei Tumori

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      57P - small cell lung cancer: Retrospective review of an institution (ID 508)

      12:30 - 12:30  |  Author(s): A. Silva, I. Guerreiro, C. Castro, M. Brandao, A. Rodrigues, C. Oliveira, I. Pousa, J. Oliveira, I. Azevedo, M. Soares

      • Abstract

      Background:
      Small cell lung cancer (SCLC) account for 10 to 15% of lung cancers. Despite the high rate of response to chemotherapy and radiotherapy, it is characterized by a rapid growth and development of widespread metastases.

      Methods:
      Retrospective review of patients with SCLC with diagnosis between January 2013 and December 2016 in our institution - Portuguese Oncology Center, Porto. Descriptive analysis and survival evaluation by Kaplan-Meier method. Evaluation of differences between survival curves by log rank test.

      Results:
      From the 144 patients included, 115 (79.9%) were male and the mean age was 65 years (range 42-87). The majority of patients had an ECOG PS 0-1 at diagnosis (n = 91, 64.1%). History of smoking was present in 115 patients (79.8%). Ki-67 was analyzed in 38 patients (36.4%) and was ≥50% in 30 patients. 108 patients (75%) had metastatic disease at diagnosis, more frequently bone (n = 61, 42.4%) and liver (n = 50, 34.7%) metastasis. Initial treatment had palliative intent in 118 patients (81.9%), and 41 (28.5%) underwent only symptomatic treatment. The most commonly used chemotherapy regimen was the doublet of platinum and etoposide (n = 98, 95.1%). The overall response rate (ORR) was 64%. Median overall survival (OS) was 5.5 months (95% confidence interval (CI) 2.9-7.9). Within patients submitted to radical treatment (n = 26, 18.1%), there was relapse in 15 (57.7%), with a median disease free survival of 10.5 months (95% CI: 9.4-11.7). Within patients submitted to palliative treatment there was progression in 43 (not evaluated in 28), with a median progression free survival of 6.3 months (95% CI: 5.9-6.7). The median OS of the patients without metastatic disease at diagnosis was significantly higher than those with metastatic disease (median 15.9 months, 95% CI: 12.3-19.4 versus 3.1 months (95% CI: 2.1 -4.1, p < 0.001).

      Conclusions:
      The results obtained in our sample are in agreement with the existing literature. The discovery of predictive and predictive biomarkers as well as new forms of treatment is urgently needed.

      Clinical trial identification:


      Legal entity responsible for the study:
      Instituto Portugues de Oncologia do Porto (Portuguese Oncology Institute of Porto, Portugal)

      Funding:
      Instituto Portugues de Oncologia do Porto (Portuguese Oncology Institute of Porto, Portugal)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      58TiP - IMpower133: Phase I/III trial of first-line atezolizumab with carboplatin and etoposide in ES-SCLC (ID 421)

      12:30 - 12:30  |  Author(s): M. Reck, A.S. Mansfield, S.V. Liu, T.S.K. Mok, X. Tang, S. Lam, F. Kabbinavar, A. Lopez-Chavez, A. Sandler, L. Horn

      • Abstract

      Background:
      Platinum-based chemotherapy (chemo) with etoposide is the current first-line standard of care for the majority of patients (pts) with ES-SCLC. However, survival outcomes remain poor (median OS, < 1 year) despite initial response rates ranging from 50%-70%. Atezolizumab (atezo) is an anti–PD-L1 mAb that prevents the interaction of PD-L1 with its receptors, PD-1 and B7.1, and restores antitumor T-cell activity. Tolerable safety with promising durability of response has been shown with atezo in pts with ES-SCLC: confirmed ORR was 6% (n = 1/17 [partial response]; DOR of 7 mo) by RECIST v1.1 and 24% by immune-related response criteria (irRC; n = 4/17, with 2 pts on atezo for ≥ 12 mo). Preliminary data also indicate the potential synergy between atezo and platinum-based chemo in NSCLC whereby durable responses may translate into improved survival compared with atezo alone. IMpower133 (NCT02763579), a global, Phase I/III, randomized, multicenter, double-blinded, placebo-controlled trial, will evaluate the efficacy and safety of 1L atezo + carboplatin + etoposide compared with placebo + carboplatin + etoposide in treatment-naive pts with ES-SCLC.

      Trial design:
      Eligibility criteria include ES-SCLC, measurable disease (RECIST v1.1), ECOG PS 0-1 and no prior systemic anticancer treatment. Exclusion criteria include untreated CNS metastases and history of autoimmune disease. Submission of tumor tissue is a study requirement but pts will be enrolled regardless of biomarker status. Stratification factors are sex, ECOG performance status and presence of treated brain metastases. Eligible pts will be randomized 1:1 to receive four 21-day cycles of atezo (1200 mg IV) or placebo in combination with carboplatin (AUC 5 mg/mL/min IV, d 1) and etoposide (100 mg/m[2] IV, d 1-3), followed by maintenance therapy with atezo or placebo until PD per RECIST v1.1. Pts can continue with treatment until persistent radiographic PD, symptomatic deterioration or unacceptable toxicity. Co-primary endpoints are investigator-assessed PFS per RECIST v1.1 and OS. Secondary efficacy endpoints include investigator-assessed ORR and DOR. Safety and tolerability will also be assessed. Approximately 400 pts will be enrolled.

      Clinical trial identification:
      NCT02763579

      Legal entity responsible for the study:
      F. Hoffmann-La Roche Ltd/Genentech Inc.

      Funding:
      F. Hoffmann-La Roche Ltd/Genentech Inc.

      Disclosure:
      M. Reck: Honoraria for lectures and consultancy with Boehringer-Ingelheim, Hoffmann-La Roche, Lilly, MSD, BMS, AstraZeneca, Celgene, Merck, Pfizer. A.S. Mansfield: Advisory Board: Genentech, BMS, and Trovagene. S.V. Liu: Advisory board/consultant for: Genentech, Pfizer, Ariad, Celgene, Boehringer Ingelheim, Lilly. T.S.K. Mok: Grant/Speaker/AdBoards: AZ, BI, Pfizer, Novartis, SFJ, Roche, MSD, Clovis, BMS, Eisai, Taiho, Lilly, Merck, ACEA, Vertex, BMS, geneDecode, OncoGenex, Celgene, Ignyta, Taiho; Stock: Sanomics; Board: IASLC, CLCRF, CSCO, HKCTS. X. Tang: Roche employee. S. Lam: Roche/Genentech: employee, stock. F. Kabbinavar, A. Lopez-Chavez, A. Sandler: Employee, stock: Roche/Genentech. L. Horn: Advisory board: Genentech; Consulting: Abbvie, Merck, Lilly, Xcovery and EMD Serono.

    • +

      62P - Risk factors associated with early vs late recurrence in stage I lung adenocarcinoma (ID 436)

      12:30 - 12:30  |  Author(s): J. Qian, J. Xu, S. Wang, W. Yang, F. Qian, B. Zhang, R. Wang, X. Zhang, H. Wang, B. Han

      • Abstract

      Background:
      Patients with stage I lung adenocarcinoma (ADC) develop recurrences after complete surgical resection, both early and over the long term. This study aimed to identify and compare risk factors for early and late recurrence after surgery in stage I lung ADC patients.

      Methods:
      We analyzed recurrences among 5904 patients with stage I lung ADC who underwent curative operations at Shanghai Chest Hospital between January 1, 2008 and December 31, 2014. Recurrences were classified as within 2 years (early), and more than 2 years (late) after surgery. The clinicopathologic findings were compared between patients with early and late recurrences. Both univariate and multivariate analyses were performed, incorporating factors of gender, age at diagnosis, operation type, tumor location and size, pathologic subtype, cell differentiation, lymphovascular invasion, visceral pleural invasion, and smoking history.

      Results:
      Recurrence occurred in totally 628 (10.6%) patients, with early and late recurrence in 300 and 328 patients, respectively. Early and late recurrences shared common risk factors of advanced age at diagnosis (P = 0.013 and P = 0.006, respectively), poorly cell differentiation (both P = 0.000), larger tumor size (both P = 0.000), and visceral pleural invasion (both P = 0.000). Early recurrence was additionally associated with male (HR, 1.327; 95%CI, 1.042-1.690; P = 0.022), sublobar resection (HR, 2.353; 95%CI, 1.634-3.387; P = 0.000), tumor located in the right middle lobe (HR, 1.711; 95%CI, 1.634-3.387; P = 0.005) and solid, micropapillary or variant subtypes (HR, 5.437; 95%CI, 1.258-23.509; P = 0.023).

      Conclusions:
      These findings suggest the existence of different risk factors accounting for early vs late recurrences among patients with stage I lung ADC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      Shanghai Chest Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      63P - Diabetes predisposes patients to atrial fibrillation after robotic-assisted video-thoracoscopic pulmonary lobectomy (ID 514)

      12:30 - 12:30  |  Author(s): S. Reynolds, J. Glover, M. Echavarria, E. Ng, F. Velez-Cubian, C. Moodie, J. Garrett, J. Fontaine, E. Toloza

      • Abstract

      Background:
      Pre-existing diabetes has been described as a risk factor for the development of atrial fibrillation (AFib) following non-cardiac surgery. This study aimed to determine if diabetes and associated comorbidities increase the risk of AFib following robotic-assisted pulmonary lobectomy and improve risk stratification of diabetics.

      Methods:
      Excluding patients with preoperative history of AFib, 353 consecutive patients who underwent robotic-assisted video thoracoscopic (RAVTS) lobectomy by one surgeon from October 2010 to August 2016 were retrospectively analysed. Patients were studied with respect to the presence of diabetes, coronary artery disease, heart failure, kidney failure, peripheral vascular disease, and other known associated comorbidities. Chi-Square (X[2]), Fisher’s exact test, and Student’s t-test were used to compare variables, with significance at p ≤ 0.05.

      Results:
      In this study, 64 patients with diabetes were identified, 11 (17.2%) of whom developed AFib following RAVTS lobectomy. Patients with diabetes were at higher risk of developing of AFib following surgery (OR 2.52, 95% CI 1.15 to 5.50, p = 0.02). The average age of diabetics who developed AFib was 72.7 years and 68.4 years for those who did not (p = 0.07). Known comorbidities in patients with diabetes did not confer additional risk, including hypertension (p = 1.00), hyperlipidaemia (p = 1.00), cardiomyopathy (p = 0.17), coronary artery disease (p = 0.27), and obesity (p = 0.67). There was a trend toward increased risk in diabetics with kidney disease, although it failed to reach significance (p = 0.07). Being a former smoker was the only independent risk factor identified, as 90.9% of diabetics with AFib were former smokers (OR 10.38, 95% CI 1.24 to 86.95, p = 0.03). Pack-years did not increase risk for AFib, with 47.2 pack-years on average being reported in those who developed AFib, while patients without AFib averaged 49.6 pack-years (p = 0.87). Furthermore, there was no significant difference in pre-operative percent forced expiratory volume in 1 second of diabetics who did and did not develop AFib (87.0% vs 80.4%, p = 0.45).

      Conclusions:
      Patients with diabetes are at higher risk for developing AFib after RAVTS lobectomy. Known comorbidities of diabetes, including obesity, hyperlipidaemia, and kidney disease, did not confer an increased risk for the development of AFib after surgery; however, being a former smoker puts diabetics at 10-times greater risk than current or never smokers with diabetes.

      Clinical trial identification:
      Not applicable.

      Legal entity responsible for the study:
      Eric M. Toloza, M.D., Ph.D.

      Funding:
      Moffitt Cancer Center and University of South Florida Health Morsani College of Medicine

      Disclosure:
      J. Fontaine, E. Toloza: Honoraria as robotic thoracic surgery observation site and proctor for Intuitive Surgical Corp. All other authors have declared no conflicts of interest.

    • +

      64P - Smoking history as a risk factor for atrial fibrillation following robotic-assisted video-thoracoscopic pulmonary lobectomy (ID 515)

      12:30 - 12:30  |  Author(s): J. Glover, S. Reynolds, M. Echavarria, E. Ng, F. Velez-Cubian, C. Moodie, J. Garrett, J. Fontaine, E. Toloza

      • Abstract

      Background:
      Smoking history has been correlated to the development of atrial fibrillation (AFib) after noncardiac thoracic surgery, increasing hospital length of stay, post-operative mortality, and costs. This study sought to determine the effects of smoking history and pulmonary function on the development of AFib following robotic-assisted pulmonary lobectomy to allow for more targeted dispositioning of post-lobectomy patients.

      Methods:
      We retrospectively analysed 353 consecutive patients without history of AFib who underwent robotic-assisted video thoracoscopic (RAVTS) lobectomy by one surgeon from October 2010 to August 2016. Patients were analysed with respect to smoking status, pack-years, months of smoking cessation, and pulmonary function. Chi-Square (X[2]), Fisher’s exact test, and Student’s t-test were used to compare variables, with significance at p ≤ 0.05.

      Results:
      In our study, 17 of 144 men (11.8%) and 16 of 209 women (7.7%) experienced new-onset AFib following RAVTS lobectomy (p = 0.19). The average age of people who developed AFib was 72.8 years (yrs) and 66.4 yrs for those who did not (p < 0.001). Former smokers represented 72.7% of new AFib cases, current smokers 21.2%, and never smokers 6.1% (p = 0.009). Former smokers were at higher risk than both never (OR 5.30, 95% CI 1.22 to 23.09, p = 0.03) and current smokers (OR 2.62, 95% CI 1.09 to 6.31, p = 0.03). Former smokers who developed AFib also were older (74.6 vs. 69.1 yrs, p = 0.004) and more often diabetic (OR 3.27, 95% CI 1.31 to 8.17, p = 0.01). There was no difference in AFib rates for light (≤15 pack-years) and heavy (>15 pack-years) smokers (p = 0.21). Never smokers fared better than light (p = 0.02) but not heavy (p = 0.13) smokers. There was no difference in pack-years for former and current smokers who developed AFib (p = 0.11). For all groups, the development of AFib was independent of pre-operative pulmonary function as measured by percent of predicted forced expiratory volume in 1 second and percent of predicted diffusion capacity of the lung for carbon monoxide (p = 0.09 and 0.63, respectively). The development of AFib was also unaffected by the presence of COPD (p = 0.80).

      Conclusions:
      Former and light smokers are at higher risk than both current and never smokers for developing AFib after RAVTS lobectomy, independent of pack-years and pre-operative pulmonary function. Duration of smoking cessation prior to lobectomy does not change the likelihood of developing AFib.

      Clinical trial identification:
      Not applicable.

      Legal entity responsible for the study:
      Eric M. Toloza, M.D., Ph.D.

      Funding:
      Moffitt Cancer Center and University of South Florida Health Morsani College of Medicine

      Disclosure:
      J. Fontaine, E. Toloza: Have received honoraria as robotic thoracic surgery observation site and proctor for Intuitive Surgical Corp. All other authors have declared no conflicts of interest.

    • +

      65P - Lymph node metastases in clinically node negative peripheral non-small cell lung cancer (ID 207)

      12:30 - 12:30  |  Author(s): T. Mehmood

      • Abstract

      Background:
      Small lung cancers are increasingly detected with the advent of low dose spiral computed tomography and lung cancer screening programme, and these lesions are frequently subsolid and low risky pN+ disease. Integrated positron emission tomography/computed tomography (PET/CT) is widely used in lymph node staging with higher accuracy and sublobar resection may be appropriate in cN0 NSCLC. This retrospective study was designed to identify the risk factors and pattern of lymph node metastases in NSCLC.

      Methods:
      107 consecutive cN0 patients with 1 cm to 2 cm peripheral NSCLC who underwent PET-CT scans followed by curative-intent resections in our hospital were enrolled in this study. Clinical and pathological data were analyzed by multivariate analysis retrospectively, including tumor size, tumor SUVmax, ratio SUVmax tumor/SUVmax liver. Lymph nodes of metastases were analyzed in pN+ patients.

      Results:
      8.5% (9/107) PET-CT diagnosed N0 NSCLC cases had pathological lymph node metastases, including 8 N1 and 3 N2 involvement (1 skipping N2 metastases). Univariable and multivariable analysis of clinicopathological factors (including tumor size, ratio SUVmax tumor/liver) found no independent risk factor for lymph node metastases. All N(+) cases were adenocarcinoma while 66.7% (6/9) of the N(+) cases were single station metastases. The lymph node metastasis rate of solid NSCLC and GGO were 10.7% and 0% (P=0.224), respectively.

      Conclusions:
      The lymph node rate of 1 cm to 2 cm NSCLC is relatively high and intrapulmonary lymph nodes were higher risk than mediastinal lymph nodes. However, no predictors of lymph node metastases were detected in this population. Sublobar resection can be an alternative procedure for GGO lesions as no lymph node metastases were found, but for solid NSCLC, thorough lymph node sampling should be performed to rule out lymph node metastases before making the decision of segmentectomy.

      Clinical trial identification:


      Legal entity responsible for the study:
      SKMH, Lahore

      Funding:
      SKMH, Lahore

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      66P - Effect of age on risk for atrial fibrillation following robotic-assisted video-thoracoscopic pulmonary lobectomy (ID 517)

      12:30 - 12:30  |  Author(s): J. Glover, S. Reynolds, E. Ng, M. Echavarria, F. Velez-Cubian, C. Moodie, J. Garrett, J. Fontaine, E. Toloza

      • Abstract

      Background:
      Aging is a known risk factor for several post-operative comorbidities, including atrial fibrillation (AFib), leading to increased length of stay and mortality. This study was designed to investigate the effect of age and related comorbidities on new-onset atrial fibrillation after robotic-assisted pulmonary lobectomy to better identify patients at greatest risk.

      Methods:
      We conducted a retrospective analysis of 353 consecutive patients without history of preoperative AFib who underwent robotic-assisted video-thoracoscopic (RAVTS) lobectomy by one surgeon from October 2010 to August 2016. Patients were analysed with respect to age and associated comorbidities, such as hypertension, hyperlipidaemia, and diabetes. Chi-Square (X[2]), Fisher’s exact test, and Student’s t-test were used to compare variables, with significance at p ≤ 0.05.

      Results:
      The average age of participants who developed post-operative AFib (n = 33) was significantly higher, 72.8 years (yrs) vs. 66.4 yrs (p < 0.001). There was a decreased risk in patients under 70 yrs, with only 11 (5.3%) developing AFib (OR 0.31, CI 0.14-0.66, p = 0.002). By contrast, 22 (15.3%) of the elderly (age ≥70 yrs) experienced new-onset A-fib (OR 2.54, 95% CI 1.19 to 5.41, p = 0.02). Those under 60 yrs were at least risk (OR 0.10, 95% CI 0.01 to 0.75, p = 0.02), and those ≥80 yrs were at greatest risk (OR 2.74, 95% CI 1.03 to 7.28, p = 0.04). While the elderly in our cohort had a higher rate of many well-described comorbidities, including hypertension, hyperlipidaemia, coronary artery disease, and cardiomyopathy, none of these conferred an increased risk of post-operative AFib. Among those over 70 yrs, only BMI, particularly an obese BMI (≥30 kg/m[2]), led to a higher risk (p = 0.02 and p = 0.003, respectively). Among the elderly, obese patients developed AFib at 2.3 times the rate of those with normal or overweight BMI’s (OR 4.21, 95% CI 1.66 to 10.68, p = 0.002). Conversely, only being a former smoker increased the risk of AFib in those under 70 yrs (OR 4.79, 95% CI 1.23 to 18.70, p = 0.02). The risk in former smokers was independent of pack-years and duration of cessation prior to surgery. Furthermore, the risk of AFib was not significantly affected by intra-operative complications in either group.

      Conclusions:
      Patients aged 70 years or older are at increased risk of AFib after RAVTS lobectomy, particularly if they are obese. Those under 70 years are at increased risk if they are former smokers.

      Clinical trial identification:
      Not applicable.

      Legal entity responsible for the study:
      Eric M. Toloza, M.D., Ph.D.

      Funding:
      Moffitt Cancer Center and University of South Florida Health Morsani College of Medicine

      Disclosure:
      J. Fontaine, E. Toloza: Have received honoraria as robotic thoracic surgery observation site and proctor for Intuitive Surgical Corp. All other authors have declared no conflicts of interest.

    • +

      67P - CT-guided fine-needle aspiration biopsy of pulmonary lesions under 15 mm of diameter: Results on 68 consecutive patients (ID 303)

      12:30 - 12:30  |  Author(s): D. Tosi, A. Palleschi, L. Rosso, P. Mendogni, I. Righi, R. Carrinola, F. Damarco, M. Cattaneo, L. Santambrogio

      • Abstract

      Background:
      Most reports on lung fine needle aspiration cytology (FNAC) demonstrate that the diagnostic accuracy tends to decrease with the size of the lesions, and that the frequency of complications increases as the size of lesions decreases. The aim of this prospective study was to describe the accuracy and incidence of complications related to FNAC of solitary pulmonary nodules (SPNs) of 15 mm or less in diameter. Moreover, we evaluated how this procedure during the initial evaluation of patients with SPN can reduce the number of unnecessary surgery.

      Methods:
      From January 2012 to December 2014, 225 patients with an SPN between 7-15 mm in diameter were referred to our Institution. Patients with risk factors such as ASA 3, FEV1 <70% of predicted, cardiac comorbidity or previous chest surgery were enrolled. A total of 68 patients were candidate to CT guided FNAC.

      Results:
      The prevalence of malignant pathology in the whole population was 61%. Forty-nine out of 68 smears (72%) were considered adequate for diagnosis. Sensitivity was 83%, specificity was 100%. Positive and negative predictive values were 83% and 100%. Out of the total number, 16 patients (23%) avoided surgery. A post-biopsy pneumothorax was detected in 27 cases (39%). The pneumothorax rate was significantly affected by the number of passages (p = 0.01).

      Conclusions:
      In our experience the diagnostic accuracy value was high. Subjecting patients with undiagnosed pulmonary lesions to FNAC during the initial evaluation significantly reduces the number of unnecessary surgical procedures.

      Clinical trial identification:
      None

      Legal entity responsible for the study:
      Fondazione Ca\' Granda Policlinico - Milan

      Funding:
      Fondazione Ca\' Granda Policlinico - Milan

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      68P - Circulating tumor cells spillage after lung nodule biopsy (ID 497)

      12:30 - 12:30  |  Author(s): M. Sarfaty, N. Abdel-Rahman, A. Moore, J. Pfannkuche, C. Chudak, M.R. Kramer, N. Peled

      • Abstract

      Background:
      Lung cancer has high recurrence rate even when diagnosed at an early stage. Biopsy is currently the procedure of choice for the investigation of pulmonary lesions, yet it is unclear whether the biopsy itself releases tumor cells into the circulation and attributes to the late distal recurrence. Studies of various malignancies show the ability to identify circulating tumor cells (CTC) even in early stage cancer. The number of CTC correlates with disease outcome. The aim of this study was to quantify the spillage of tumor cells after nodule biopsy in early lung cancer.

      Methods:
      Patients with pulmonary nodules undergoing computed tomography biopsy enrolled into this study. CellCollector[TM] (GILUPI, Germany) was used for CTC detection before and after the procedure. This filtration device isolates CTC in-vivo through a standard cannula inserted to the cubbital vein for thirty minutes via epithelial cell adhesion molecule antibodies. Patients served as their own controls. Trans thoracic biopsy was done by a 19G needle with a standard procedure of one pass. The study was approved by the institutions' ethics committee.

      Results:
      13 patients were enrolled to between 02/2016 and 08/2016. Five patients were excluded due to partial procedure. Eight patients were eligible for analysis. Three had benign diseases (Granuloma, Interstitial pneumonitis. and five had lung cancer (Adenocarcinoma -4; Squamous-1); 3 had an elevation of the CTC count (0->2, 0->3, 2->8) and 2 had a depletion (8->2, 5->2). All 5 patients were male, with a median age of 65 years (range 54-72), four of them were past smokers and one never smoker. No adverse events were reported.

      Conclusions:
      With the limitation of low number of participants, this preliminary data shows that trans thoracic biopsy may increase CTC in some patients with lung cancer. The study is ongoing and the enlarged cohort will be presented.

      Clinical trial identification:


      Legal entity responsible for the study:
      Rabin Medical Center Ethics Comittee

      Funding:
      GILUPI

      Disclosure:
      J. Pfannkuche: Managing director at GILUPI, which provided the kits for this study. C. Chudak: Marketing & Sales Manager at GILUPI, which provided the kits for this study. All other authors have declared no conflicts of interest.

    • +

      69P - CT-Guided radiofrequency ablation of early stage NSCLC (ID 168)

      12:30 - 12:30  |  Author(s): X. Zhang

      • Abstract

      Background:
      To prospectively evaluate the safety and efficacy of radiofrequency (RF) ablation of early stage NSCLC with diameter of less than 30 mm.

      Methods:
      Forty patients (24 men and 16 women; age range, 35–78 years; mean age, 58 years) with early stage NSCLC (40 lesions) which were conformed by biopsy were enrolled in during 2014-2016. RF ablation was performed in tumors by CT guidance, each with a diameter of less than 30 mm (mean deviation, 14 mm). Written informed consent was obtained in this prospective study that was approved by the local ethics committee. Follow-up CT scans were obtained within 48 hours after treatment and at 2 and 6months thereafter to evaluate treatment outcome and complications. Lung spirometry measurements were obtained before and 2 months after RF ablation.

      Results:
      Forty successful RF ablation treatments were performed. The complications occured in the periprocedural period were 9 cases of minor pneumothorax and 4 cases of sputum cruentum. There were 35 lesions of reduced size, 13 lesions of stabilized lesion size, and 2 lesions of increased size on the follow-up Contrast-enhanced CT images at 6-month. There was no modification of respiratory function was found between the spirometry measurements before the treatment and 2 months after (P < 0.05).

      Conclusions:
      RF ablation is a safe and effective method to treat early stage NSCLC with diameter of less than 30 mm. Larger studies are necessary to fully evaluate its potential combination with other treatment techniques.

      Clinical trial identification:


      Legal entity responsible for the study:
      Xiaobo Zhang

      Funding:
      The Radiology Department of Chinese PLA General Hospital

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      70P - A retrospective analysis to explore the value of gemcitabine combined with cisplatin as adjuvant chemotherapy of NSCLC (ID 375)

      12:30 - 12:30  |  Author(s): D. Ma, J. Wang, X. Hao, Y. Wang, X. Hu, P. Xing, J. Li

      • Abstract

      Background:
      To evaluate the value of gemcitabine combined with cisplatin (GP) as adjuvant chemotherapy in radical resection of non-small cell lung cancer (NSCLC).

      Methods:
      This study reviewed the 100 patients’ charts with radical resection of NSCLC treated with cisplatin/gemcitabine as adjuvant chemotherapy between June 2007 and December 2010 in CAMS (Chinese Academy of Medical Sciences).

      Results:
      Among the 100 cases, eighty-two (82%) was male and the median age was 59 years (range, 36-73 years). Forty-two (42%) patients were adenocarcinoma, while fifty-five (55%) were squamous cell carcinoma. Most patients had pathologic IIB (29%) and IIIA (44%) stage disease, whereas others were IA (2%), IB (14%), IIA (6%), and IIIB (5%). Surgical methods included sleeve resection (12%), pneumonectomy (14%), and lobectomy (73%).The completion rate of 4 cycles of chemotherapy was 85%, of which 76 case (76%) completed the planned full-dose chemotherapy. And the main reason for the reduction in gemcitabine doses in thirteen patients were grade 3/4 myelosuppression, mainly neutropenia and thrombocytopenia. The median dose and dose intensity were 8377.1 mg/m[2] and 708 mg/(m[2]·week) for gemcitabine, and 293.38 mg/m[2] and 25.24 mg/(m[2]·week) for cisplatin respectively. With good compliance to treatment, toxicities observed were tolerable and managable. During a median follow-up duration of 73.1 months, the median disease-free survival(DFS) was 33.8 months (95%CI: 15.938-51.676). Patients with the squamous cell carcinoma (HR 0.404, 95% CI 0.241-0.676, P = 0.001) and pathologic stage I (HR 4.379, 95% CI 1.721-11.142, P = 0.002) were associated with better DFS from univariate and multivariate analyses. The survival rate at 1 year, 2 years and 5 years was 94%, 77% and 55%, while the survival rate without recurrence was 64%, 53% and 39% respectively.

      Conclusions:
      As one of the adjuvant chemotherapy regimens, gemcitabine combined with cisplatin is well tolerated. Patients with squamous cell carcinomas or stage I can benefit from it better.

      Clinical trial identification:


      Legal entity responsible for the study:
      None

      Funding:
      None

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      71TiP - A randomized, phase 3 trial with anti-PD-1 monoclonal antibody pembrolizumab (MK-3475) versus placebo for patients with early stage NSCLC after resection and completion of standard adjuvant therapy (EORTC/ETOP 1416-PEARLS) (ID 399)

      12:30 - 12:30  |  Author(s): L. Paz-Ares, B. Hasan, U. Dafni, J. Menis, E. De Maio, K. Oselin, I. Albert, M. Faehling, P. Van Schil, M.E.R. O'Brien

      • Abstract

      Background:
      In the last 5 years no major advances have been made in the treatment of early stage NSCLC. Checkpoint inhibitors have shown promising clinical efficacy in advanced, refractory non-small cell lung cancer (NSCLC), but they have not yet been explored in the adjuvant setting. PEARLS (NCT02504372) is international, triple-blinded, placebo-controlled, randomized phase III trial to compare pembrolizumab versus placebo after complete resection of stage IB (T ≥ 4 cm), II and IIIA NSCLC, followed by standard adjuvant chemotherapy, if appropriate as per local guidelines, in patients who have signed the informed consent.

      Trial design:
      Eligible patients are those with completely resected stage IB (T ≥ 4 cm), II and IIIA NSCLC that have or have not received adjuvant platinum-based chemotherapy and whose PD-L1 status is known: negative (TPS=0%) versus weak positive (TPS = 1-49%) versus strong positive (TPS≥50%). Co-primary endpoints are disease-free survival in the PD-L1 strong positive subgroup and in the overall population. An HR = 0.78 is targeted for the whole population with 640 disease free survival events from a sample size of 1380 randomized patients. Secondary endpoints include disease-free survival in the PD-L1 positive subgroup, overall survival in each subpopulation and in the overall population, lung cancer specific survival, and safety and tolerability. The exploratory endpoints will assess pharmacokinetics, immunogenicity, quality of life and potential biomarkers of treatment response. Recruitment started in January 2016 and is currently ongoing. As of November 17, 2016, among the 102 randomized patients so far, 44 (43.1%), 32 (31.4%) and 26 (25.5%) patients have negative, weak positive and strong positive PD-L1 status respectively.

      Clinical trial identification:
      (EudraCT number 2015-000575-27) (NCT02504372)

      Legal entity responsible for the study:
      MSD EORTC, ETOP

      Funding:
      MSD

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      75P - Predictive value of CT texture analysis in lung cancer patients undergoing nivolumab (ID 231)

      12:30 - 12:30  |  Author(s): V. Nardone, P. Tini, E. Aldi, P. Pastina, L. Sebaste, G. Battaglia, S.F. Carbone, V. Ricci, L. Pirtoli, P. Correale

      • Abstract

      Background:
      Nivolumab is a human mAb to PD-1 with significant antitumor activity in a number of solid malignancies including NSCLC, kidney cancer and melanoma. At the present, no predictive factor has been identified for this drug, thus its administration is mostly empirical, at price of frequent adverse events and high costs. In this context, we evaluated whether baseline CT texture analysis (TA) could be used to identify advanced NSCLC patients who will benefit by Nivolumab treatment, by taking in consideration that the therapeutic effects PD-1 blockade depends by its ability of reactivating a pre-existing immuneresponse, whose activity is strictly related to the presence of necrosis, hypoxia, and inflammation in the tumor sites, which can be evaluated by imaging assessments.

      Methods:
      A retrospective analysis was performed on a sample of seventeen NSCLC patients who received salvage therapy with Nivolumab 3 mg/kg every 15 days between October 2015 and January 2017 with a median follow up of twelve months. The gross primary tumor volume before treatment (baseline) was contoured on pre and post contrast CT sequences. TA parameters were extrapolated by using LifeX Software ©, tested for reliability and then correlated with patients’ outcome, in particular with early failure defined as a confirmed disease progression or death within 6 months (7 patients), by means of univariate and multivariate analysis.

      Results:
      We found a significant correlation among TA parameters and patients’ outcome at univariate analysis. In fact, early vs longer responders showed differences in term of volume in voxel (p:0.049), entropy (p:0.046), compacity (p:0.033), GLCM-contrast (p: 0.018), GLCM-dissimilarity (p:0.017), LRHGE (p:0.019), coarseness (p:0.036), contrast (0.020), ZP (p:0.025), pre contrast: GLCM-contrast (p:0.031), GLCM-dissimilarity (p:0.035), contrast (p:0.038), SZE (p:0.029), ZP (p:0.040). Multivariate analysis (Logistic regression) confirmed a significant correlation between early failure and post contrast GLCM dissimilarity (p:0.011, OR: 3.30, AUC: 0.800; 95% CI 0.578-1.00).

      Conclusions:
      Our results, if confirmed on a larger series, suggest that TA may predict early failure, and therefore may help the selection of patients who may benefit by Nivolumab treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Azienda Ospedaliera Universitaria Senese

      Funding:
      Azienda Ospedaliera Universitaria Senese

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      76P - Interpretation and prognostic value of PET-CT after induction chemotherapy with or without radiation in IIIA-N2 non-small cell lung cancer patients who receive curative surgery (ID 200)

      12:30 - 12:30  |  Author(s): J.H. Lee

      • Abstract

      Background:
      We evaluated the correlation of clinical staging on positron emission tomography-computed tomography (PET-CT) and pathologic staging and the prognostic value of PET-CT after induction chemotherapy in patients with locally advanced non-small cell lung cancer (NSCLC).

      Methods:
      We analyzed 42 cases of clinical stage IIIA-N2 NSCLC who received 2 to 4 cycles of pre-operative chemotherapy with or without radiation followed by curative resection. The maximum standard uptake value (SUVmax) of the suspected lesion on PET-CT was recorded. PET-CT findings after induction chemotherapy were compared with those of initial PET-CT and pathology after surgery.

      Results:
      The accuracy of PET-CT in restaging of the primary tumor after induction chemotherapy was 50.0%. 18 (42.8%) of 42 patients were underestimated ycT stage, and 3 (7.1%) of 42 patients was overestimated ycT stage by PET-CT scan. The accuracy of PET-CT in restaging of the nodal disease was 71.4%. 6 (14.3%) of 42 patients were underestimated ycN stage, and 6 (14.3%) of 42 patients were overestimated ycN stage as compared with pathologic staging. The 2-year overall survival (OS) and relapse-free survival (RFS) rate were 68.5% and 40.9%, respectively. Complete responders (ycT0N0M0) on PET-CT after induction chemotherapy had a significantly longer RFS time than did incomplete responders (28.3 months versus 9.1 months, P = 0.021).

      Conclusions:
      Complete response on PET-CT after induction chemotherapy with or without radiation was a good prognostic indicator for RFS in stage IIIA-N2 NSCLC patients who received surgery. However, response evaluation on PET-CT after induction chemotherapy should be interpreted with caution due to its unacceptable accuracy.

      Clinical trial identification:
      None declared.

      Legal entity responsible for the study:
      Jong Hoon Lee

      Funding:
      None

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      77P - Bronchoscopic photodynamic therapy for treatment of microscopic residual disease post resection in non-small cell lung cancer (ID 257)

      12:30 - 12:30  |  Author(s): A. Biswas, A. Khokar, S. Fernandez-Bussy, M.A. Jantz, H.J. Mehta

      • Abstract

      Background:
      The goal of lung cancer surgery is a complete tumor resection (R0 resection) with tumor-free resection margins. However, 4-5% of lung cancer resections have evidence of microscopic residual disease which is associated with worse prognosis as compared to complete resection. Definitive management for such patients is re-operation and resection of residual tumor. However, a significant percent of patients may not be able to tolerate another surgery and for those patient’s definitive management is not well studied. We treated these patients with stage I cancer and mucosal residual disease with bronchoscopic PDT (photoynamic therapy) and report our experience here.

      Methods:
      We conducted a retrospective analysis of patients who underwent definitive surgery for early stage lung cancer. All patients with R1 resection, stage I disease with mucosal residual tumor and or carcinoma in situ along the stump site were treated with bronchoscopic photodynamic therapy and were studied. Patient characteristics, histology, type and site of surgery, pattern of recurrence, recurrence status, and survival data were evaluated. Adverse events were recorded.

      Results:
      A total of 11 patients (Table) with mucosal R1 resection were treated with PDT along the stump site between 2007 and 2013. The breakdown according to the pattern of residual disease was as follows: CIS in 3 and MRD in 8 patients. One patient (9%) had local recurrence 1 year after treatment and was treated with radiation along the stump site. Four patients (36%) had no evidence of recurrence to date after a median follow up of 4 years and other 6 patients had evidence of regional (16%) or distant (45%) and were treated appropriately for the same. Local control rate was 91%. The median overall survival and progression free survival in our cohort of patients were 45 and 26 months respectively. One patient developed pneumonia and one patient has evidence of photosensitivity reaction.rnTable: 77Prn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn rnrn
      Patient #AgeSexOperationHistologyType of residual diseaseSite of treatmentPattern of recurrencePFS monthsOverall survival monthsAlive
      153MLobectomyAdeno caMRDLLLregional4966Y
      280FLobectomySCCaCISLULn/an/a45N
      380FSegmentectomyAdeno caMRDRMLDistant1011N
      457MLobectomySCCaCISRLLn/an/a70Y
      566MLobectomySCCaMRDLLLDistant2832N
      640MLobectomyAdeno caMRDRULregional3659N
      766FLobectomyAdeno caMRDRLLn/an/a86Y
      867FLobectomySCCaMRDRMLDistant2634N
      976MSleeve lobectomySCCaMRDLLLn/an/a39N
      1072FSegmentectomySCCaCISRULLocal2473Y
      1174FLobectomySCCaMRDLULDistant2125N
      rn

      Conclusions:
      Conclusion: Bronchoscopic photodynamic therapy (PDT) is a nonthermal ablative technique that can be used to treat central airway disease in adults. In conclusion, based on our findings, PDT is a safe and effective alternative therapy for patients with mucosal residual disease post R1 resection. Although this was a retrospective evaluation with a small sample, our data suggest that selected NSCLC patients after R1 resection may benefit from bronchoscopic PDT.

      Clinical trial identification:
      This is not a clinical trial.

      Legal entity responsible for the study:
      University of Florida

      Funding:
      None

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      78P - Comparison of chemoradiotherapy treatment strategies in stage III non-small cell lung cancer among elderly patients from multiple data sources (ID 365)

      12:30 - 12:30  |  Author(s): H. Pang, T. Stinchcombe, P. Cheng, A.W. Lee, V.H. Lee, E.E. Vokes, X. Wang

      • Abstract

      Background:
      Comparative effectiveness research can benefit from combining data from multiple sources. This analysis integrally evaluated the survival benefits of combined modality therapies (CMTs) in locally advanced non-small cell lung cancer (NSCLC) based on individual patient data from US and Chinese populations.

      Methods:
      Two patient populations were included. SEER-Medicare cohort consists of 65 years or higher who were diagnosed with stage III (IIIA/IIIB) NSCLC from 2006 to 2010 and received combined modality treatment. Queen Mary Hospital cohort from Hong Kong for aged 65 or higher who were diagnosed with stage III (IIIA/IIIB) NSCLC from 2007 to 2016. The four CMTs of interest were concurrent (CMT-ONLY), sequential chemotherapy followed radiation (CMT-SEQ), induction followed by concurrent (CMT-IND) and concurrent followed by consolidation (CMT-CON) chemoradiation. The primary endpoint for was overall survival. Patients hospitalized for neutropenia were ascertained through inpatient claim with the claim occurring within 130 days after the first chemotherapy. Stepwise multivariable regression models and propensity score adjusted models were used to control confounding variables. We also compared the findings with our previous study based on clinical trials data.

      Results:
      2682 locally advanced NSCLC patients were included. For CMT-ONLY, CMT-SEQ, CMT-IND, and CMT-CON respectively, their corresponding median overall survivals were 13.0 (95% CI 12.0-14.0), 13.0 (95% CI 12.0-16.0), 17.0 (95% CI 15.0-20.0), and 15.4 (95% CI 14.0-17.0) months. CMT-IND and CMT-CON had survival benefit over CMT-ONLY and CMT-SEQ. Patients receiving CMT-SEQ were relatively healthier patients in this study based on the Charlson comorbidity weights. 7.9%, 2.8%, 8.6%, and 9.5% were hospitalized for neutropenia for CMT-ONLY, CMT-SEQ, CMT-IND, and CMT-CON, respectively. CMT-SEQ had a lower hospitalization for neutropenia rate than the other CMTs.

      Conclusions:
      The findings on efficacy and toxicity are quite consistent with previously reported studies based on clinical trials or observational databases.

      Clinical trial identification:


      Legal entity responsible for the study:
      Duke University

      Funding:
      NIH, HMRF

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      79P - Long term survival of stage IIIB non-small cell lung cancer (NSCLC) patients treated with concurrent chemoradiation (ID 208)

      12:30 - 12:30  |  Author(s): T. Mehmood

      • Abstract

      Background:
      The optimal treatment strategy for Stage IIIB NSCLC patients with a T4N0-1 tumor is a matter of debate. In prospective combined modality series including surgery, the median overall survival (OS) is approximately 24 months. We hypothesized that results comparable to regimens including surgery can be achieved with concurrent chemoradiation in this patient group.

      Methods:
      In our retrospectively collected database of NSCLC patients, all patients with T4 (mediastinal invasion) N0-1 NSCLC receiving concurrent chemoradiation were included. One patient had a recurrence after previous pneumonectomy. All patients were given 3 cycles of chemotherapy (cisplatin and etoposide). Radiotherapy (RT) was started at the 2nd course of chemotherapy. OS was calculated from date of diagnosis (Kaplan-Meier method). Toxicity was scored according to CTCAEv3.0.

      Results:
      42 patients (8 females, 34 males) with a median age of 62.5 ± 9 years (44-80 years) were included from January 2005 until December 2009. Stage distribution: 86% T4N0 (n = 36), 14% T4N1 (n = 6). The maximal tumor dose was 66 Gy using conventional fractionation. The median prescribed mean lung dose was 15 ± 4.4 Gy (5.03 -19.9 Gy). Acute toxicity: 1 patient experienced grade 3 dyspnea during RT. Grade 3 dysphagia occurred in 5 patients (12%) during RT requiring tube feeding in 3 of these patients (7%). Dysphagia persisted later than 1 month after RT in 1 patient (2%). Grade 3 dysphagia only occurred in patients treated concurrently. Grade 3 cough occurred in 1 patient during RT, no patient experienced grade 3 cough 1 month after RT. 2 patients died within 3 months after start of RT, one due to myocardial infarction, one of unknown causes. Severe late toxicity was not present: no grade 3 complications more than 3 months after the end of radiotherapy. With a median follow-up of 42 months, the median OS for the whole group is 34 months (95% CI 24-43 months). 2-year OS survival is 55%.

      Conclusions:
      Concurrent accelerated chemoradiation using an individualized dose prescription is a valid treatment strategy for stage IIIb, T4N0-1 NSCLC patients yielding very promising OS results with low toxicity.

      Clinical trial identification:


      Legal entity responsible for the study:
      SKMH, Lahore

      Funding:
      SKMH, Lahore

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      80TiP - A feasibility trial evaluating the addition of nivolumab to standard first-line chemo-radiotherapy in locally advanced stage IIIA/B NSCLC: The ETOP 6-14 NICOLAS trial (ID 455)

      12:30 - 12:30  |  Author(s): S. Peters, R..A. Stahel, M. Kassapian, M. Guckenberger, E. Felip, A..J. De Langen, R.M. Huber, H. Roschitzki-Voser, A. Piguet, D. De Ruysscher

      • Abstract

      Background:
      Sequential or concomitant chemo-radiotherapy is the treatment of choice for stage III NSCLC. One attempt to improve the long-term survival is an immunotherapeutic strategy. The monoclonal antibody nivolumab targets and inhibits PD-1, an immune receptor on activated T-cells and responsible for abrogating anti-cancer immune response. The role of immune-checkpoint inhibitors is currently being evaluated in this indication as monotherapy or in combination with chemotherapy as well as after completion of chemo-radiotherapy but it has not yet been assessed in combination with radiotherapy. While anecdotal data of concurrent therapy suggests an acceptable toxicity profile, a formal prospective assessment is required before embarking on trials comparing concurrent versus sequential checkpoint inhibitors and radiotherapy.

      Trial design:
      NICOLAS is a phase II feasibility trial evaluating the administration of nivolumab concomitantly with standard first-line chemo-radiotherapy for locally advanced stage IIIA/B NSCLC. Additional inclusion criteria include adequate organ function and performance status 0-1. Chemotherapy consists of three cycles of cisplatin plus vinorelbine, etoposide or pemetrexed. Radiotherapy to the chest will be delivered either sequentially to or concurrently with chemotherapy. The initial nivolumab dose is 240 mg Q2W for eight cycles in the sequential, and 360 mg Q3W for four cycles in the concurrent chemo-radiotherapy schedule, followed by 480 mg Q4W for maximum one year for both regimens. A total of 43 patients will be recruited into the trial. The primary endpoint is grade > =3 pneumonitis observed up to 6 months after radiotherapy. The goal is to reject a 6-month pneumonitis-free rate of < =67%, tested at a rate of 85%, with 85% power and 5% one-sided alpha. An interim analysis will be performed after 21 patients have completed a 3-month follow-up on nivolumab treatment. Safety is closely monitored by the ETOP IDMC at their regular meetings every three months. NICOLAS is sponsored by ETOP with financial support of Bristol-Meyers Squibb. Accrual is ongoing and as of January 2017, 18 patients have been enrolled.

      Clinical trial identification:
      EudraCT: 2014-005097-11

      Legal entity responsible for the study:
      European Thoracic Oncology Platform (ETOP)

      Funding:
      Bristol-Meyers Squibb

      Disclosure:
      R.A. Stahel: Honoraria as consultant at advisory boards from Abbvie, Astra Zeneca, BMS, Boehringer Ingelheim, Eli Lilly, MSD, Pfizer and Roche and as speaker from Astellas, Astra Zeneca, Lilly, MSD, Novartis and Roche. R.M. Huber: Honoraria as consultant at advisory boards from BMS. All other authors have declared no conflicts of interest.

    • +

      97P - Brigatinib in crizotinib-refractory ALK+ NSCLC: Updates from the pivotal randomized phase 2 Trial (ALTA) (ID 247)

      12:30 - 12:30  |  Author(s): M.J. Hochmair, M. Tiseo, K.L. Reckamp, H.L. West, H.J. Groen, C.J. Langer, W. Reichmann, D. Kerstein, D. Kim, D.R. Camidge

      • Abstract

      Background:
      In a phase 1/2 trial (NCT01449461), the investigational next-generation anaplastic lymphoma kinase (ALK) inhibitor brigatinib (BRG) showed promising activity in crizotinib-treated ALK-positive non–small cell lung cancer (ALK+ NSCLC) patients (pts); because tumor responses and adverse events (AEs) varied with starting dose, two BRG regimens were evaluated in ALTA (NCT02094573).

      Methods:
      Pts with crizotinib-refractory advanced ALK+ NSCLC were stratified by presence of brain metastases and best response to prior crizotinib and randomized 1:1 to receive BRG at 90 mg qd in arm A or 180 mg qd with a 7-day lead-in at 90 mg in arm B. Investigator-assessed confirmed objective response rate (ORR) per RECIST v1.1 was the primary endpoint.

      Results:
      222 pts were enrolled (112 in arm A, 110 in arm B); median age was 54 years, 57% were female, 74% had received chemotherapy, and 69% had brain metastases. As of 29 February 2016, 57%/69% of pts in arms A/B were receiving BRG, with 7.8/8.3-month median follow-up. Investigator-assessed efficacy by arm and subgroup is shown below. Per independent review committee, as of 16 May 2016, confirmed ORR was 48%/53% and median PFS was 9.2/15.6 months in arms A/B. Treatment-emergent AEs with ≥25% overall frequency (A/B, n = 109/n=110 treated) were nausea 33%/40%, diarrhea 19%/38%, headache 28%/27%, and cough 18%/34%; grade ≥3 events with ≥3% frequency were hypertension 6%/6%, increased blood creatine phosphokinase 3%/9%, pneumonia 3%/5%, and increased lipase 4%/3%. A subset of pulmonary AEs with early onset (median onset: Day 2) occurred in 14/219 (6%) treated pts (3%, grade ≥3); no such events occurred after escalation to 180 mg in arm B, and 7/14 pts were successfully retreated.

      Conclusions:
      BRG yielded substantial efficacy, with an acceptable safety profile, in both arms. 180 mg with 90 mg lead-in showed an improvement in efficacy endpoints, particularly PFS, with no increase in early pulmonary AEs, compared with 90 mg. Investigator-Assessed Efficacy by Subgroup.rnTable: 97PDrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn rnrn
      rnArm A n = 112Arm B n = 110Total N = 222
      Confirmed ORR, n/N (%)rnrnrn
       All pts[a]50/112 (45)59/110 (54)109/222 (49)
       Racernrnrn
        Asian18/39 (46)18/30 (60)36/69 (52)
        Non-Asian32/73 (44)41/80 (51)73/153 (48)
       Prior chemotherapyrnrnrn
        Yes35/83 (42)44/81 (54)79/164 (48)
        No15/29 (52)15/29 (52)30/58 (52)
       Best response to prior crizotinibrnrnrn
        CR or PR36/71 (51)47/73 (64)83/144 (58)
        Other14/41 (34)12/37 (32)26/78 (33)
       Baseline brain metastasesrnrnrn
        Yes31/80 (39)43/74 (58)74/154 (48)
        No19/32 (59)16/36 (44)35/68 (51)
      Median PFS, monthsrnrnrn
       All pts9.212.911.1
       Racernrnrn
        Asian8.811.111.1
        Non-Asian9.212.911.8
       Prior chemotherapyrnrnrn
        Yes8.812.911.8
        No9.28.19.2
       Best response to prior crizotinibrnrnrn
        CR or PR11.115.615.6
        Other7.412.99.2
       Baseline brain metastasesrnrnrn
        Yes9.211.811.1
        No7.415.615.6
      rnCR = complete response, ORR = objective response rate, PFS = progression-free survival, PR = partial responsernaPrimary endpointrn

      Clinical trial identification:
      NCT02094573

      Legal entity responsible for the study:
      ARIAD Pharmaceuticals, Inc.

      Funding:
      ARIAD Pharmaceuticals, Inc.

      Disclosure:
      M. Tiseo: Consulting or advisory role (AstraZeneca, BMS, Boehringer Ingelheim, Eli Lilly, Novartis, Otsuka, Pierre Fabre), research funding (ARIAD). K.L. Reckamp: Consulting or advisory role (ARIAD), research funding (ARIAD). H.L. West: Consulting or advisory role (ARIAD, Astellas, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Merck, Novartis, Pfizer, Roche/Genentech, Trovagene), speakers’ bureau (ARIAD, Eli Lilly, Roche/Genentech). H.J. Groen: Consulting or advisory role (Eli Lilly, MSD, Novartis, Pfizer, Roche). C.J. Langer: Honoraria (BMS, Lilly/ImClone, Roche/Genentech), consulting or advisory role (Abbott, ARIAD, AstraZeneca, Bayer/Onyx, BMS, Cancer Support Community, Celgene, Clarient, Clovis Oncology, Lilly/ImClone, Merck, Millennium, Roche/Genentech), research funding (Advantagene, ARIAD, Celgene, Clovis Oncology, GSK, Inovio, Merck, Roche/Genentech). W. Reichmann, D. Kerstein: Employment, stock and other ownership interests (ARIAD). D.R. Camidge: Honoraria (ARIAD), research funding (ARIAD). All other authors have declared no conflicts of interest.

    • +

      98P - Retrospective indirect comparison of alectinib phase II data vs ceritinib real-world data in ALK+ NSCLC after progression on crizotinib (ID 379)

      12:30 - 12:30  |  Author(s): J. Davies, M. Martinec, R. Martina, P. Delmar, M. Coudert, W. Bordogna, S. Golding, G. Crane

      • Abstract

      Background:
      Approvals of second-line ALK inhibitors (ALKi) ceritinib and alectinib are based on single-arm trials that lack comparative efficacy data to support health technology assessments. We assessed if real-world data (RWD) could provide this by acting as an external control for single-arm studies by comparing data generated from alectinib trials and ceritinib patient data.

      Methods:
      We retrospectively analysed patients (pts) with ALK+ advanced NSCLC receiving an ALKi after crizotinib failure. The alectinib arm (ALC; n = 183) included pts from the phase II NP28673/NP28761 studies. To generate the ceritinib control arm (CER; n = 67) eligibility criteria similar to the alectinib trials was applied to the Flatiron Health’s electronic health record database. A propensity score based on prognostic factors was generated and applied by inverse probability treatment weighting. A multivariate Cox model was used to evaluate the association of ALC compared with CER on overall survival (OS) adjusting for age, sex, prior treatment, race and stage at diagnosis. Summary data from the CER trial ASCEND-2 were re-digitised to compare with CER RWD.

      Results:
      Prior to re-weighting, the arms were heavily imbalanced. Key differences between the arms included age, prior treatments and baseline CNS metastases (median: 53 vs 61 yrs, 36% vs 13% [≥3 lines], 61% vs 35%, ALC vs CER). After weighting, balance was achieved across all key prognostic factors with a standardised mean difference <10% for all factors. A multivariate Cox model showed ALC was associated with lower risk of death (HR 0.65; 95% CI 0.48–0.88). Adjusted median OS was 24.2 months for ALC (95% CI 17.5–NR) vs 15.6 months for CER (95% CI 15.5–18.6). Median OS in the RWD CER group was similar to that reported in the CER ASCEND-2 trial (14.9 months).

      Conclusions:
      The results show that ALC is associated with prolonged OS vs CER, which was consistent through numerous sensitivity analyses. CER RWD median OS was similar to that observed in ASCEND 2, validating this analysis. For single-arm studies, RWD can serve as an external control for producing comparative data.

      Clinical trial identification:
      NP28673 NP28761

      Legal entity responsible for the study:
      F. Hoffmann-La Roche

      Funding:
      F. Hoffmann-La Roche

      Disclosure:
      J. Davies, G. Crane: Employee at Roche Products Ltd. P. Delmar, M. Coudert: Employee at F. Hoffmann-La Roche, Ltd. M. Martinec, W. Bordogna, S. Golding: Employee: F. Hoffmann-La Roche; Stock ownership: F. Hoffmann-La Roche. All other authors have declared no conflicts of interest.

    • +

      99P - Efficacy of gefitinib in advanced squamous cell lung cancer harboring epidermal growth factor receptor mutations: A pooled analysis of individual patient data (ID 300)

      12:30 - 12:30  |  Author(s): D. Tao, N. Zhang, Y. Wang

      • Abstract

      Background:
      Gefitinib has demonstrated clinical benefits for patients suffering from lung adenocarcinoma with activating mutation of epidermal growth factor receptor (EGFR). However, the efficacy of gefitinib for advanced squamous cell lung cancer (SqCLC) harboring EGFR mutation is still unclear.

      Methods:
      We conducted a pooled analysis of individual patient data based on the published reports that included patients with advanced SqCLC harboring EGFR mutation who were treated with gefitinib. The patients selected for the analysis were advanced SqCLC patients harboring EGFR mutations who were treated with gefitinib and described in reports containing the data of the histology, EGFR mutation status and type, response to gefitinib, and outcome of these patients.

      Results:
      Tweenty-six patients were selected from ten reports for the pooled analysis. One patient obtained a complete response (CR) and five patients obtained a partial response (PR); the response rate (RR) was 23.1%. The median progression-free survival (mPFS) and overall survival (mOS) was 3.1 months and 10.8 months, respectively. The RR and PFS were significantly inferior in the SqCLC patients harboring EGFR mutations to non-SqCLC patients harboring EGFR mutations selected from the same published reports (RR: 23.1% vs. 62.2%, P < 0.001; PFS: 3.1 vs. 9.2 months, P < 0.001, respectively). While no significant difference in OS was observed between the two groups (OS: 10.8 vs. 17.7 months, P = 0.082).

      Conclusions:
      The efficacy of gefitinib was inferior in advanced SqCLC harboring EGFR mutation as compared to non-SqCLC harboring EGFR mutation. The underlining mechanisms of resistance to EGFR-TKI in SqCLC with EGFR mutation should be deeply explored in the further studies.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chongqing Cancer Hospital & Cancer Institute

      Funding:
      Chongqing Cancer Hospital & Cancer Institute

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      100P - Afatinib vs gefitinib for treatment-naïve patients with EGFRm+ NSCLC (LUX-Lung 7): Analysis of time to treatment failure and impact of afatinib dose adjustment (ID 356)

      12:30 - 12:30  |  Author(s): J.C. Yang, L. Paz-Ares, E. Tan, K. O'Byrne, L. Zhang, M. Boyer, T. Mok, V. Hirsh, J. Fan, K. Park

      • Abstract

      Background:
      PFS was significantly improved in LUX-Lung 7 with afatinib (A) vs gefitinib (G). Time to treatment failure (TTF) was a co-primary endpoint to reflect clinical practice of continuing TKI tx beyond radiologic progression in the absence of clinical deterioration. An analysis of TTF and a post-hoc analysis of the impact of dose adjustment of A on PFS and AEs are reported here.

      Methods:
      Patients (pts) were randomized to A 40mg/d or G 250mg/d until progressive disease (PD) or beyond if deemed beneficial. The dose of A could be reduced by 10mg decrements to a minimum of 20mg in the event of selected drug-related (DR) AEs. TTF was analyzed using a stratified log-rank test and Kaplan-Meier methods. PFS was compared between pts who had a dose reduction within 6 mos and those who received ≥40mg for 6 mos. Incidence/severity of common AEs before/after dose reduction was assessed.

      Results:
      319 pts were randomized (160 A, 159 G). At data cut-off (21 Aug 2015), 87.5% A and 93.7% G pts had discontinued tx, mostly due to radiologic PD (69.4 vs 74.8%) or toxicity (11.3 vs 10.7%). 35.0% A and 29.6% G pts with clinical benefit continued tx beyond radiologic PD. Pts remained on tx significantly longer with A vs G (median TTF 13.7 vs 11.5 mos; HR 0.73 [95% CI 0.58–0.92]; p = 0.007; pts on tx at 2 yrs: 25.0 vs 13.2%). TTF subgroup analyses favored A. Risk of tx failure was reduced with A vs G regardless of EGFRm type or race. Median tx duration beyond PD with A and G was 2.7 and 2.0 mos, respectively. 63 pts (39%) treated with A had a dose reduction to 30mg; 21 (13%) had further reduction to 20mg. There was no significant difference in PFS in pts who received <40 mg or ≥ 40 mg (median 12.8 vs 11.0 mos; HR 1.3 [95% CI 0.9–2.0]; p = 0.14). Dose reduction of A reduced the incidence/severity of DR AEs: grade ≥3 diarrhea, rash/acne and stomatitis were reduced from 25.4%, 20.6% and 7.9%, to 9.5%, 3.2% and 3.2%, respectively.

      Conclusions:
      TTF was significantly improved with first-line A vs G in EGFRm+ NSCLC, which testifies to the tolerability of A, and suggests that it may confer additional clinical benefit in pts who continue tx beyond PD. Dose adjustment of A reduced the frequency/intensity of DR AEs without compromising efficacy.

      Clinical trial identification:
      LUX-Lung 7: EudraCT No: 2011-001814-33

      Legal entity responsible for the study:
      Boehringer Ingelheim

      Funding:
      Boehringer Ingelheim

      Disclosure:
      J.C-H. Yang: Ad board and honoraria: BI, Lilly, Bayer, Roche/Genentech/Chugai, Astellas, MSD, Merck Serono, Pfizer, Novartis, Clovis, Celgene, Merrimack, Yuhan Pharmaceuticals, BMS, Ono pharmaceutical Daiichi, Sankyo, and AZ. L. Paz-Ares: Honoraria from Pfizer, Bristol-Myers Squibb, MSD, Novartis, Roche, Eli Lilly, Boehringer lngelheim, Clovis Oncology, AstraZeneca, and Amgem. K. O\'Byrne: Ad board, speaker bureau, travel to international conferences and honoraria: AZ, BMS, Roche-Genentech, MSD, Pfizer, BI. Ad board and speaker bureau: Novartis. 3 Patents: 1 on novel drugs, 2 on biomarkers, IP held by Queensland University of Technology. M. Boyer: Ad board: BMS, Merck Sharpe and Dohme, Pfizer Board of Directors: IASLC Research: Pfizer, Genentech, BI, AZ, Novartis, Merck Sharpe and Dohme, Clovis Honoraria: Merck Sharpe and Dohme, BI, BMS, AZ. T. Mok: Receipt of grants/research supports: AstraZeneca, BI, Pfizer, Novartis, SFJ, Roche, MSD, Clovis Oncology, BMS; Receipt of honoraria or consultation fees: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, Merck Serono, MSD, Janssen, Clovis Oncology, BioMarin, GSK, Novartis, SFJ Pharmaceutical, ACEA Biosciences, Inc., Vertex Pharmaceuticals, BMS, AVEO & Biodesix, Prime Oncology, Amgen; Participation in a company sponsored speaker’s bureau: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, MSD, Amgen, Janssen, Clovis Oncology, GSK, Novartis, BMS, PrIME Oncology; Stock shareholder: Sanomics Limited. V. Hirsh: Honoraria for participating on advisory boards for Boehringer Ingelheim, AstraZeneca, Roche, Merck, Eli Lilly, Pfizer, Amgen, and Bristol-Myers Squibb. K. Park: Participated on advisory boards for Astellas, AstraZeneca, Boehringer Ingelheim, Clovis Oncology, Eli Lilly, Hanmi, MSD, Novartis, and Roche. All other authors have declared no conflicts of interest.

    • +

      101P - Association between time to progression and subsequent survival in previously treated BRAF V600E-mutant metastatic non-small cell lung cancer (ID 416)

      12:30 - 12:30  |  Author(s): M. Sasane, J. Li, J. Zhang, J. Zhao, M.L. Ricculli, S. Redhu, J. Signorovitch

      • Abstract

      Background:
      Time to progression (TTP) has been associated with prolonged post-progression survival (PPS) in ALK positive non-small cell lung cancer (NSCLC). However, such associations have not yet been assessed among previously treated metastatic NSCLC patients with BRAF V600 mutation who received BRAF-specific targeted therapies. This study evaluated the TTP-PPS association among previously treated BRAF V600E-mutant metastatic NSCLC patients receiving the BRAF inhibitor dabrafenib as a single agent or in combination with the MEK inhibitor trametinib.

      Methods:
      Patients who experienced disease progression in an open-label, non-randomized, Phase II study (NCT01336634) of 2nd-line or later treatment with dabrafenib monotherapy or combination therapy with trametinib were included. Progression was defined per RECIST v1.1. Time from progression to death (PPS) was studied in a Kaplan-Meier analysis with patients stratified by shorter (< 6 months) versus longer TTP (≥ 6 months). The association between TTP and PPS was also quantified in Cox proportional hazards models.

      Results:
      A total of 84 patients who progressed on dabrafenib monotherapy or combination therapy were included in this study, with 77% having ECOG performance score > 0 at screening. Prior to progression, 57 received dabrafenib monotherapy and 27 received combination therapy with trametinib. Among all patients included, 60 (71%) died during post-progression follow-up. Patients with TTP ≥ 6 months experienced significantly longer PPS compared to those with TTP < 6 months (median PPS: 9.5 vs. 2.7 months, log-rank p-value < 0.001). In the proportional hazards model for the full cohort, each 3 months of longer TTP was associated with a 32% lower hazard of death following progression (hazard ratio [95% confidence interval]: 0.68 [0.57, 0.83]). A similar positive association between TTP and PPS was observed in each treatment group (monotherapy: 0.70 [0.57, 0.88]; combination therapy: 0.57 [0.34, 0.97]).

      Conclusions:
      A longer duration of TTP after treatment with dabrafenib monotherapy or combination therapy was significantly associated with a longer duration of PPS.

      Clinical trial identification:
      NCT01336634

      Legal entity responsible for the study:
      Novartis

      Funding:
      Financial support for the study were provided by Novartis.

      Disclosure:
      M. Sasane: Employee of Novartis and own Novartis stock or stock options. J. Li: Employed by Analysis Group, Inc., payment from Novartis for participation in this research. J. Zhang: Employee of Novartis and own Novartis stock or stock options. J. Zhao: Employed by Analysis Group, Inc., payment from Novartis for participation in this research. M.L. Ricculli: Employed by Analysis Group, Inc., payment from Novartis for participation in this research. S. Redhu: Employee of Novartis and own Novartis stock or stock options. J. Signorovitch: Employed by Analysis Group, Inc., payment from Novartis for participation in this research.

    • +

      102P - LUX-Lung 8 phase III trial: Analysis of long-term response to second-line afatinib in patients with advanced squamous cell carcinoma (SCC) of the lung (ID 355)

      12:30 - 12:30  |  Author(s): J.C. Yang, G. Goss, E. Felip, S. Lu, A. Ardizzoni, S.M. Gadgeel, V. Georgoulias, N. Dupuis, E. Ehrnrooth, J. Soria

      • Abstract

      Background:
      In LUX-Lung 8, afatinib (A; 40mg/day) significantly improved OS (median 7.9 vs 6.8 months, p = 0.008) and PFS (2.6 vs 1.9 months, p = 0.010) versus erlotinib (E; 150mg/day) in pts with pretreated SCC of the lung (n = 795). 12-month (36 vs 28%; p = 0.016) and 18-month survival (22 vs 14%; p = 0.013) were significantly higher with A than E, indicating that some pts derive prolonged benefit from A. This is a post-hoc analysis of baseline characteristics and efficacy/safety of A in long-term responders (LTRs; treatment for ≥12 months). Archived tumor samples and serum were analyzed to identify potential biomarkers.

      Methods:
      Tumor samples were retrospectively analyzed using next-generation sequencing (NGS); EGFR expression was determined by IHC. Pre-treatment serum samples were analyzed with VeriStrat[®] and classified as VeriStrat-Good or VeriStrat-Poor.

      Results:
      21/398 pts treated with A were LTRs. Six pts were still on treatment at the time of data cut-off. The median duration of treatment was 17.6 months (range: 12.3–27.6). Baseline characteristics were similar to the overall dataset (median age: 64 y [range: 54–81]; male: 76%; Asian: 29%; ECOG 0/1: 33%/67%; best response to chemotherapy CR or PR/SD: 48%/52%; current and ex-smokers: 90%). Median PFS was 16.6 months (range: 2.8–25.8); median OS was 21.1 months (12.9–31.6). The most common treatment-related AEs (all grade/grade 3) were: diarrhea (81%/5%); rash/acne (71%/5%); stomatitis (29%/5%). AEs generally occurred soon after treatment onset. Six pts had dose reductions due to related AEs. NGS data in ten LTRs will be presented. Genomic aberrations in the ErbB gene family were identified in 50% of these pts (overall dataset: 26.5%). Of 17 pts assessed by VeriStrat, 88% were VeriStrat-Good (overall dataset: 62%). IHC data were available for only one LTR (EGFR-).

      Conclusions:
      Baseline characteristics of LTRs to A were similar to the overall dataset. A conferred median OS of almost 2 years in this subgroup. A was well tolerated with predictable, transient AEs. Though biomarker data look promising, the cohort was too small to identify any clear NGS/VeriStrat predictive signals; further studies are required.

      Clinical trial identification:
      LUX-Lung 8: EudraCT No: 2011-002380-24

      Legal entity responsible for the study:
      Boehringer Ingelheim

      Funding:
      Boehringer Ingelheim

      Disclosure:
      J.C-H. Yang: Ad board and honoraria: BI, Lilly, Bayer, Roche/Genentech/Chugai, Astellas, MSD, Merck Serono, Pfizer, Novartis, Clovis, Celgene, Merrimack, Yuhan Pharmaceuticals, BMS, Ono pharmaceutical Daiichi, Sankyo, and AZ. G. Goss: Participated on advisory boards for AstraZeneca, Boehringer Ingelheim, Pfizer, Eli Lilly, Bristol-Myers Squibb, and Celgene. E. Felip: Participated on advisory boards for Eli Lilly, Pfizer, Roche, MSD, and Boehringer Ingelheim. Felip has receieved lecture fees from AstraZeneca, Bristol-Myers Squibb, and Novartis. A. Ardizzoni: Received honoraria and participated on advisory boards for Bristol-Myers Squibb, MSD, Eli-Lilly, and Boehringer Ingelheim. Ardizzoni has received honoraria from Pfizer and Bayer. S.M. Gadgeel: Participated on advisory boards for Boehringer Ingelheim, Pfizer, Genentech, ARIAD, AstraZeneca, Bristol-Myers Squibb, and Roche. N. Dupuis: Employee of and owns stock in Biodesix. E. Ehrnrooth: Employee of Boehringer Ingelheim. J-C. Soria: Astrazeneca, Astex, GSK, Gammamabs, Lilly, MSD, Merus, Pfizer, Pharmamar, Pierre Fabre, Roche, Sanofi, Servier, Symphogen, Takeda. All other authors have declared no conflicts of interest.

    • +

      103P - Re-biopsy for advanced non-small cell lung cancer after EGFR tyrosine kinase inhibitor therapy: CT characteristics of patients with T790M mutation and the use of various re-biopsy procedures (ID 385)

      12:30 - 12:30  |  Author(s): H.J. Koo, M.Y. Kim, C. Choi, J.C. Lee, S. Park

      • Abstract

      Background:
      Re-biopsy for mutation analysis of non-small cell lung cancer (NSCLC) after EGFR-tyrosine kinase inhibitor treatment is important to determine further chemotherapy regimen. There have been no studies about the radiologic characteristics of NSCLC with T790 mutation and the use of the various re-biopsy procedures.

      Methods:
      Between January and December 2016, 78 patients underwent re-biopsy for mutation analysis of NSCLC, and among them, 76 were assessed with adequate specimen. Patients’ treatment course, serial CT scans and pathologic reports were retrospectively reviewed. Re-biopsy methods are varied: EBUS or BFS-guided (n = 27), CT-guided (n = 18), fluoroscopy-guided (n = 5) biopsies, US-guided supraclavicular lymph node (n = 6) or other sites (n = 6) biopsies and pleural fluid analysis (n = 14). CT images obtained at the time of initial biopsy and re-biopsy were compared between patients with and without T790M mutation. Re-biopsy associated complications were assessed.

      Results:
      Among 76 patients, 40 (52.6%) presented T790M mutation on re-biopsy. Progression free survivals between patients with and without T790M mutation were not statistically different (322 and 389 days, respectively). On initial CT, pleural retraction (odds ratio (OR), 4.1; p = 0.03) and the presence of pleural metastasis (OR, 3.4; p = 0.03) were significant factors that related to the positive T790M mutation by multivariate logistic analysis. Pleural retraction (OR, 26.8, p = 0.03) and pleural metastasis (OR, 11.4; p = 0.004) are also shown as significant factors that related to the positive T790M mutation on CT obtained at the time of re-biopsy. Three patients developed pneumothorax, and two were managed by chest tube insertion. One patient who was negative T790M mutation on pleural fluid analysis finally diagnosed as positive T790M mutation by following CT-guided biopsy.

      Conclusions:
      Pleural retraction and pleural metastasis were significantly associated factors to positive T790M mutation in NSCLC patients who underwent re-biopsy. Negative T790M on pleural fluid analysis could not give a guarantee for true negative, and further core biopsy might be recommended.

      Clinical trial identification:


      Legal entity responsible for the study:
      None

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      104P - Efficacy of ceritinib in a "real-world" population of crizotinib-refractory ALK-positive NSCLCs: Results of the Italian compassionate use program (ID 422)

      12:30 - 12:30  |  Author(s): G. Metro, A. Passaro, G. Lo Russo, V. Gregorc, R. Giusti, E. Capelletto, O. Martelli, F.L. Cecere, L. Bonanno, R. Chiari

      • Abstract

      Background:
      Ceritinib has been approved by EMA for the treatment of patients (pts) with anaplastic lymphoma kinase (ALK)-positive advanced non-small cell lung cancer (NSCLC) who progress on crizotinib. Nevertheless, this drug is not reimbursed in Italy by the National Health Care System as of January 2017. The aim of this study was to assess the efficacy of ceritinib administered within a compassionate use (CU) program made available to crizotinib-refractory patients.

      Methods:
      This collaborative study involved multiple institutions in Italy. Clinical data from crizotinib-refractory pts with ALK-positive NSCLC who were requested ceritinib compassionate use (CU) were collected and analyzed.

      Results:
      Twenty-four centres took part to the study, for a total of 70 pts who were requested ceritinib CU. Of these, 63 pts received at least one dose of ceritinib 750 mg/d from July 2014 to January 2017. Pts characteristics were as follows: median age 56 years (22-86), 34/63 (54%) female, 43/63 (68%) never smokers, 12/63 (19%) ECOG PS ≥ 2, 13/63 (21%) pretreated with ≥ 2 lines of chemotherapy, 49/63 (78%) metastatic to the brain. Median time on prior crizotinib was 370 days (51-1644). The most common any grade treatment-related adverse events (TRAEs) were nausea (60%, 6% grade 3 or 4), vomiting (49%, 5% grade 3 or 4), diarrhea (51% 2% grade 3 or 4), ALT elevation (48%, 17% grade 3 or 4), AST elevation (49%, 17% grade 3 or 4), and fatigue (59%, 8% grade 3 or 4). Dose reduction due to TRAEs occurred in 31/63 pts (49%). Out of 31 pts, 15 pts (49%) reduced the dose to 600 mg/d, 10 pts (32%) to 450 mg/d, and 6 pts (19%) to 300 mg/d. Unusual TRAEs consisted of an increase in serum creatinine in 3 pts. Of the 55 patients who were evaluable for response, 21 pts (38%) responded to treatment. Overall, at a median follow-up of 6.2 months (0.5-26), median progression-free survival (PFS) was 7.6 months and 6-month PFS was 59%.

      Conclusions:
      The ceritinib CU program in Italy confirms the efficacy of the drug in a "real-world" setting, with a safety profile that is similar to that observed in clinical trials. A high rate of dose adjustments due to TRAEs was observed, which, however, did not appear to influence the activity of the drug.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      N/A

      Funding:
      Associazione Italiana per la Ricerca contro il Cancro

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      105P - Prognostic factors in crizotinib (CRZ)-resistant anaplastic lymphoma kinase (ALK+) non-small cell lung cancer (NSCLC) patients (Pts) (ID 469)

      12:30 - 12:30  |  Author(s): K.L. Reckamp, J. Lee, J. Huang, I. Proskorovsky, W. Reichmann, M. Krotneva, D. Kerstein, H. Huang

      • Abstract

      Background:
      The aim of this analysis was to assess prognostic factors in patients with CRZ-resistant ALK+ NSCLC associated with progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) based on the Phase 2 ALTA Trial (NCT02094573) of brigatinib.

      Methods:
      Analyses used data from ALTA Arm B (180 mg qd with a 7-day lead-in at 90 mg, N = 110). Potential prognostic factors were evaluated using univariate and multivariate Cox proportional hazards models for PFS and OS, and logistic regression for ORR. Potential prognostic factors included age, sex, race, ECOG performance status, prior radiotherapy (RT), prior RT to brain, prior chemotherapy, prior platinum-based chemotherapy, smoking status, number of prior regimens, best response to prior CRIZ, number of metastatic sites, and active brain lesions. ALTA trial was not powered to detect differences in outcomes by these factors, thus a threshold of p < 0.5 was used to select variables into all models. Factors with hazard ratios >1.30 or < 0.77 were also included in the final models for PFS and OS.

      Results:
      As of February 29, 2016, median follow-up was 8.3 months, and independent review committee (IRC) assessed median PFS was 15.6 months [95% CI: 11.0, NR] (31 events). Median OS was not reached (17 deaths). IRC-assessed ORR was 52.7% [95% CI: 43.0%, 62.3%]. Prognostic factors are shown in the table.rnTable: 105PImportant prognostic factors in CRIZ-resistant ALK+ NSCLC treated with brigatinib 180 mg qd with a 7-day lead-in at 90 mg (N = 110)rn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      Prognostic FactorPFS (HR [95% CI])*OS (HR [95% CI])*ORR (OR [95% CI])**
      Age (per 1-year increase)0.98 [0.95, 1.01]
      Sex Male vs female1.79 [0.84, 3.79]
      Race Asian vs non-Asian1.29 [0.55, 3.04]0.15 [0.02, 1.25]1.87 [0.73, 5.00]
      ECOG performance status 1 vs 0 2 vs 00.72 [0.33, 1.58] 3.24 [0.99, 10.60]2.56 [0.72, 9.06] 4.02 [0.73, 22.14]0.64 [0.26, 1.53] 0.19 [0.02, 1.07]
      Prior chemotherapy Yes vs no0.42 [0.19, 0.95]0.34 [0.11, 1.04]1.90 [0.71, 5.24]
      Best response to prior CRIZ CR/PR vs any other status or unknown0.56 [0.25, 1.27]0.74 [0.24, 2.33]1.88 [0.77, 4.70]
      Prior radiotherapy to brain Yes vs no2.21 [1.02, 4.78]0.51 [0.17, 1.57]
      Active brain lesions Yes vs no0.45 [0.15, 1.39]
      Smoking status Never vs current/former/unknown0.49 [0.14, 1.67]2.45 [1.04, 5.97]
      Number of metastatic sites 3 vs 1-2 4+ vs 1-21.05 [0.20, 5.58] 2.66 [0.59, 12.03]0.34 [0.10, 1.08] 0.81 [0.27, 2.36]
      rnAbbreviations: PFS = progression-free survival; OS = overall survival; ORR = objective response rate; HR = hazard ratio; OR = odds ratio; CI = confidence interval; CR = complete response; PR = partial response; ECOG = Eastern Cooperative Oncology Group;rn*HR < 1 indicates lower risk of progression or death vs reference group;rn**OR > 1 indicates higher odds of response vs reference grouprn

      Conclusions:
      In this analysis of CRZ-resistant ALK+ NSCLC patients, a history of prior chemotherapy was associated with longer PFS, while prior radiotherapy was associated with shorter PFS, both likely due to unmeasured patient characteristics. Never smokers had more than double the odds of response, versus current or former smokers. ECOG status 2 was nominally associated with shorter PFS and OS and lower ORR, and presence of active brain lesions was associated with longer OS.

      Clinical trial identification:
      The trial protocol number is NCT02094573.

      Legal entity responsible for the study:
      Evidera Inc.

      Funding:
      ARIAD Pharmaceuticals

      Disclosure:
      K.L. Reckamp: Consulting or advisory role (ARIAD), research funding (ARIAD). J. Lee, M. Krotneva: Employee of Evidera Inc., which provides consulting services to pharmaceutical organizations. Evidera Inc. received funding from ARIAD pharmaceuticals. J. Huang: Employment, consulting or advisory role (ARIAD). W. Reichmann, D. Kerstein, H. Huang: Employment, stock shareholder (ARIAD). All other authors have declared no conflicts of interest.

    • +

      106P - Brain metastases (BM) development in molecular selected non-small cell lung cancer (NSCLC) patients included in clinical trials (ID 305)

      12:30 - 12:30  |  Author(s): S. Cedres, N. Pardo, A. Navarro-Mendivil, A. Martinez, A. Martinez de Castro, J. Remon, F. Amair, J. Zeron, M. Vilaro, E. Felip

      • Abstract

      Background:
      The molecular profiling of patients (p) with advanced NSCLC identifies several oncogenic drivers that can be targeted with selective inhibitors. We aimed to assess the characteristics and brain development of p with molecular alterations at our center treated with targeted agents.

      Methods:
      EGFR, KRAS, HER2 mutated p and ALK, ROS1 and RET rearrangements positive p enrolled onto clinical trials between 2009 and 2015 at our center were included in this analysis. A cohort of wild type (WT) adenocarcinoma p was selected as comparator. Overall survival (OS) was estimated by the Kaplan Meier method.

      Results:
      200 p were collected (76 WT, 45 EGFR, 51 ALK, 21 KRAS, 3 ROS1, 2 HER2 and 2 RET). Median age 57 years (26-82), 52% men, 60% performance status (PS) 1, 59% smokers, 98% stage IV and 92% adenocarcinoma. First treatment was selective inhibitor in 73% of EGFR and 58% of ALK p. Median follow up was 23 months (m) (95% CI 1.6-104.6). The OS (immature with 58% of deaths) was 33m for all p and 57m EGFR, 40m ALK, 31m KRAS and 19m WT. We found differences in OS for molecular selected population vs WT (55m vs 19 m p < 0.001), women (55m vs 23m, p = 0.002), PS 0/1 vs PS2 (21 vs 7m, p < 0.001) and non-smokers (51 vs 23 m smokers, p = 0.002). Brain metastases were detected in 86 p (36 ALK, 25 WT, 14 EGFR, 8 KRAS, 2 ROS and 1 RET) and 87% received local therapy. BM were more frequent in women, non-smokers and ALK p (p < 0.001). BM developed at a median of 6m from diagnosis of NSCLC (6m molecular selected and 5m WT, p = 0.44) and median OS after development of BM was 14m (28m EGFR, 26m ALK and 8m WT, p < 0.001). No differences in OS were detected in p with or without BM (p > 0.05). Independently of target agent, we did not found significant differences in OS p with BM treated with local therapy vs systemic treatment (p > 0.005). P who initiated the EGFR and ALK inhibitors after diagnosis of BM had greater benefit than those p who began treatment before diagnosis of BM (86m vs 57m for EGFR and 55m vs 35m for ALK respectively, p > 0.05 in both).

      Conclusions:
      Molecular selected p treated with targeted agents have prolonged survival. Brain metastases is a frequent site of disease progression, but the prognosis of these p is impressive independently of local therapies.

      Clinical trial identification:
      not aplicable

      Legal entity responsible for the study:
      N/A

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      107P - The real world experience of first generation Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor (EGFR-TKI) in patients with advanced lung cancer: Explorative analysis of big data from national health insurance service of Korea (ID 437)

      12:30 - 12:30  |  Author(s): B.S. Kwon, J.C. Lee, S. Kim, M.Y. Kim, C. Choi

      • Abstract

      Background:
      Although the effectiveness of EGFR-TKI in patients with advanced lung cancer has been proven in previous trials, the real-world efficacy of EGFR-TKI has not been investigated. The object of this study was to evaluate the outcomes of first-generation EGFR-TKI in female patients with advanced lung cancer using big data analysis.

      Methods:
      We retrospectively reviewed the Korean health insurance service data of advanced lung cancer patients from January 2004 to December 2013. Patients older than 20 years in palliative setting were grouped into three categories: Group A received 1st-generation EGFR-TKI ≥6 months, group B given EGFR-TKI < 6 months, and group C was treated with cytotoxic chemotherapy alone. For each group, we determined progression-free survival (PFS) and overall survival (OS).

      Results:
      Of 11,045 patients enrolled in the study, 6,170 (55.8%) received EGFR-TKI at least 1 month, and 4,875 (44.2%) never treated with EGFR-TKI. 2,572 were in the group A and 3,598 were in the group B. In the group C, platinum based doublet agent or monotherapy such as docetaxel, gemcitabine and pemetrexed was administered. The median OS of patients treated with EGFR-TKI was significantly longer than that of EGFR-TKI naïve patients (19.1 months [95% confidence interval (CI) 18.5-19.7] vs 9.5 months [95% CI 9.1-9.8]; p < 0.0001). In subgroup analysis, the median OS was 30.3 months [95% CI 29.5-31.2] in the group A, compared with 12.3 months [95% CI 11.9-12.7] in the group B and 9.5 months [95% CI 9.1-9.8] in the group C (p < 0.001). Patients age < 65 years, treated with EGFR-TKI ≥ 6months, and received docetaxel, pemetrexed chemotherapy were independent predictors of longer OS (Hazard ratio (HR) 0.78 [95% CI 0.74-0.81]; p = 0.002, HR 0.41 [95% CI 0.38-0.43]; p < 0.001, HR 0.81 [95% CI 0.77-0.85]; p < 0.001, HR 0.77 [95% CI 0.74-0.81]; p < 0.001, respectively). Median PFS was 15.8 months [95% CI 15.1-16.3] in group A and 3.7 months [95% CI 3.5-3.8] in group B (p < 0.001).

      Conclusions:
      The current study demonstrated that EGFR-TKI conferred a significant PFS and OS benefit in female patient with advanced lung cancer in real-world.

      Clinical trial identification:


      Legal entity responsible for the study:
      Asan Medical Center

      Funding:
      None

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      108P - NSCLC patients harbouring a rare or complex EGFR mutation are more often smokers and might not benefit from first line TKI therapy (ID 479)

      12:30 - 12:30  |  Author(s): D. Kauffmann-Guerrero, K. Kahnert, S. Reu, A. Jung, Z. Synyaeva, P. Mertsch, R.M. Huber

      • Abstract

      Background:
      TKI therapy is definitely standard in first line treatment of patients with advanced NSCLC harbouring a classic EGFR mutation (Exon 19 deletion or Exon 21 L858R). However, a certain percentage of patients have rare or complex EGFR mutation with unknown relevance regarding prognosis and response to TKIs. For those patients, therapy decisions remain challenging.

      Methods:
      343 patients with NSCLC, who underwent EGFR mutation testing, were analysed in this study with regard to epidemiological characteristics as age, sex and smoking status as well to EGFR mutation status classified into wild type, classic, rare, synonymous, T790M and complex mutations. Further rare and complex mutations of 12 patients who received TKI therapy were analysed regarding to response to TKI treatment.

      Results:
      282 (82%) of all patients were EGRF wild type, whereas 61 (18%) harboured an EGFR mutation. Most of them appeared to be classic mutations 32 (9%), followed by rare (16 (5%)) and complex (7 (2%)) mutations. Synonymous and T790M resistance mutations were found in 3 and 4 patients respectively. EGFR mutations were significantly more frequent in women than in men. It was noticeable that patients with rare or complex mutations were significantly more often smokers compared to classic EGFR mutations. Further the rare and complex mutations showed to be less responsive to TKI therapy.

      Conclusions:
      Smoking seems to enhance the probability of rare or complex EGFR mutations. Besides, those mutations might not benefit from first line TKI therapy, so that the presence of EGFR mutation alone should not necessarily lead to induction of a TKI therapy and in case of TKI treatment to close surveillance. In fact, type of mutation, reports of response and patient characteristic as performance status should lead the decision.

      Clinical trial identification:


      Legal entity responsible for the study:
      Department of Internal Medicine V, University Munich

      Funding:
      Department of Internal Medicine V, University of Munich

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      109P - Sequential administration of EGFR-TKI and pemetrexed achieved a long period of response in advanced NSCLC patients with EGFR-mutant tumors (ID 322)

      12:30 - 12:30  |  Author(s): Y. Chen, Q. Zhu, L. Dai, Z. Ding

      • Abstract

      Background:
      The study aimed to explore the efficacy of sequential administration of EGFR TKI and pemetrexed doublet chemotherapy (P), two effective therapies, in advanced NSCLC patients with EGFR mutant tumors.

      Methods:
      This was a retrospective study where patients prescribed with EGFR-TKI (gefitinib or icotinib) from Jan 2013 to Jan 2016 were screened. Patients must have metastastic diseases harboring TKI-sensitizing EGFR mutation. They must be older than 18 years, and have evaluable target lesions. Whether TKI or P was used in the first line, it must be switched to the other in the second line. PFS in both first line (PFS1) and second line (PFS2) was collected.

      Results:
      We screened totally 550 patients (gefitinib n = 455, icotinib n = 95) to enroll a cohort of 37 patients. However, 1 patient was found to subject adjuvant TKI therapy and therefore was excluded. They were all adenocarcinoma except 1 adenosquamous carcinoma. Gender, good PS, mutation type were equally distributed (male n = 19 vs female n = 17, PS:0 n = 21 vs PS:1 n = 15, exon 19 del n = 14 vs L858R n = 14 vs other type n = 8). The median age was 50.5 years. For the whole cohort, the total PFS (PFS1+PFS2) was 18.1 m (95%CI: 15.2-21.1 m). For the patients receiving first-line TKI, PFS1 and PFS2 were 10.3 m and 6.6 m. And for those receiving first-line P, PFS1 and PFS2 were 3.4 m and 11.5 m.

      Conclusions:
      Our results argued the sequential usage of TKI and P achieve a long period of response in advanced NSCLC patients with EGFR-mutant tumors, comparable to that of synchronous administration (15.4 m, JMIT study).

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      West China Hospital, Sichuan University

      Funding:
      West China Hospital, Sichuan University

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      110P - Afatinib for patients with advanced NSCLC pretreated with chemotherapy and an EGFR tyrosine kinase inhibitor: Retrospective analysis of the Swiss Afatinib Named Patient Program (ID 505)

      12:30 - 12:30  |  Author(s): M. Ehmann, L. Wannesson, M. Pless, M. Früh, O. Gautschi, A. Curioni-Fontecedro, D. Betticher, M. Mark, A. Ochsenbein, S.I. Rothschild

      • Abstract

      Background:
      Epidermal growth factor receptor (EGFR) mutations are found in 12-15% of lung adenocarcinoma patients in European countries. Afatinib is a second-generation EGFR tyrosine kinase inhibitor (TKI) and is approved for stage IV NSCLC patients with common EGFR mutations. Based on the LUX-Lung 1 trial a named patient program for afatinib was initiated in Switzerland. We thus aimed to evaluate afatinib activity in patients which where previous treated with chemotherapy and first-generation TKIs.

      Methods:
      This multicentre retrospective analysis was performed in 11 institutions in Switzerland. We reviewed clinical records of patients included in the afatinib NPP.

      Results:
      Between 03/2011 and 04/2014 a total of 69 patients were included in the NPP. Baseline characteristics were obtained from all these patients. Follow-up data were accessible from 41 patients. Median age of the population was 63 years (range, 46-79). 68% of patients were female and 28% were never smoker. Adenocarcinoma was the predominant histological subtype (93%). 56 patients (81%) had a proven EGFR mutation. Of those, 29 patients (52%) had a deletion 19, 16 patients (29%) had a L858R mutation in exon 21. A T790M mutation was detected in 10 patients (18%). 50 patients (73%) received treatment with erlotinib and 14 patients (20%) with gefitinib before inclusion in the NPP. 31 patients were evaluable for response assessment by RECIST 1.1. One patient (3.2%) achieved a complete remission, 4 patients (12.9%) showed a partial remission and 3 patients (9.7%) disease stabilization. Mean duration of afatinib therapy was 200 days (95% CI 146-255). Mean overall survival (OS) from diagnosis of metastatic NSCLC was 17.2 months (95%CI 11.9-22.6). In multivariate analysis, EGFR mutation was associated with response to afatinib.

      Conclusions:
      This study confirms the activity of afatinib in pretreated lung adenocarcinoma. The benefit is larger in patients with EGFR mutation positive tumors and mainly in those with classical mutations (deletion 19 or point mutation L858R in exon 21).

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Unrestricted educational grant by Boehringer-Ingelheim

      Disclosure:
      S.I. Rothschild: Advisory Board Boehringer-Ingelheim (honorary paid to the institution). All other authors have declared no conflicts of interest.

    • +

      111P - Pharmacist led proactive follow-up algorithm for advanced EGFR positive NSCLC patients on afatinib (ID 513)

      12:30 - 12:30  |  Author(s): P.K. Cheema, A. Thawer, J. Leake, S.Y. Cheng, S. Khanna, J.C. Victor

      • Abstract

      Background:
      Afatinib is a standard first-line therapy for advanced EGFR positive NSCLC. We implemented a pharmacy led proactive follow-up algorithm for pts prescribed afatinib to identify and manage early adverse events (AEs) (Table). Management of AEs was standardized.

      Methods:
      This was a retrospective chart review of all advanced EGFR positive NSCLC pts at the Sunnybrook Odette Cancer Centre from April 1, 2015 to July 31, 2016 that received afatinib following institution of our algorithm. This study evaluated the impact of our algorithm and characteristics of real world AEs.

      Results:
      We included 33 pts. Median age was 64 and 55%, 79%, 59%, 27% and 88% were female, PS < 2, Asian, smokers, and treated as 1st line, respectively. Median follow up was 249 days (d). Median time on afatinib was 157 d (IQR: 27 to 304). Prophylactic use of topical hydrocortisone/clindamycin was 55% and 46% for an oral tetracycline. All pts had 1 drug related AE, 18% were grade 3/4. Most common AEs were diarrhea 88%, rash 82%, stomatitis 58%, paronychia 46%, nausea 39% and fatigue 39%. Median time to 1st drug related AE was 17 d (IQR: 7 to 126), with early median time to onset of diarrhea 8 d, stomatitis 13 d, rash 15 d, fatigue 18 d and nausea 25 d and late onset of paronychia 75 d, transaminitis 114 d and anorexia 133 d. Median dose of afatinib was 40 mg/daily, 34% of pts had > or = 1 dose reduction and 9% discontinued afatinib due to AEs. Proactive calls identified 37% of all drug related AEs, 33% of grade 3/4 AEs, 58% of first drug related AEs and identified 2 patients that were noncompliant. Only 3% of AEs were identified by an ER visit/urgent clinic visit.rnTable: 111PPhamacist led follow-up algorithm for pts prescribed afatinibrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn rnrn
      Time on drugDay 1Day 5Day 10Day 14Day 17
      InterventionVisit with pharmacyProactive pharmacy callProactive pharmacy callRoutine clinic visit with medical oncologyProactive pharmacy call
      Variables assessedPatient education of side effects and consent obtained for proactive callsAdherence Rash Diarrhea StomatitisAdherence Rash Diarrhea Stomatitis Paronychia Nausea FatigueClinical assessment and laboratory monitoring of CBC, creatinine and liver function testsAdherence Rash Diarrhea Stomatitis Paronychia Nausea Fatigue Anorexia
      rn

      Conclusions:
      Our algorithm resulted in early identification and management of AEs with a low rate of urgent assessments and discontinuation due to toxicity while maintaining the ideal dose of afatinib. This algorithm provides a tool for centres prescribing afatinib.

      Clinical trial identification:


      Legal entity responsible for the study:
      Dr. Parneet Cheema, PI and Sunnybrook Health Sciences Centre

      Funding:
      Boehringer Ingelheim

      Disclosure:
      P.K. Cheema: Advisory board and research grants: Boehringer Ingelheim. A. Thawer: Advisory board and research grant: Boehringer Ingelheim. S.Y. Cheng, S. Khanna: Advisory board: Boehringer Ingelheim. All other authors have declared no conflicts of interest.

    • +

      112P_PR - Immune response and adverse events to influenza vaccine in cancer patients undergoing PD-1 blockade (ID 502)

      12:30 - 12:30  |  Author(s): S.I. Rothschild, C. Balmelli, L. Kaufmann, M. Stanczak, M. Syedbasha, D. Vogt, O. Gautschi, A. Egli, A. Zippelius, H. Laeubli

      • Abstract

      Background:
      Immune checkpoint inhibitors have been introduced into standard clinical practice. Since cancer patients are at risk to develop complications when infected with seasonal influenza viruses – in particular patients with lung cancer that often have pre-existing lung disorders – it is recommended to vaccinate oncological patients. Concerns have been raised about the safety of influenza vaccination in patients undergoing checkpoint blockade. It is feared that immune stimulation via PD-1/PD-L1 blockade might induce an overshooting immune response.

      Methods:
      Patients undergoing checkpoint blockade were vaccinated with a trivalent influenza vaccination between October and November 2015. For an age-matched control cohort, the partners of the patients were vaccinated and included in our analysis as healthy controls. Safety and frequency of immune-related adverse events were evaluated. Antibody titers against antigens and strains that were included in the trivalent influenza vaccination were measured by hemagglutination inhibition assay in patients undergoing PD-1 blockade and age-matched controls. Cytokine/chemokine profile and changes in peripheral immune cells in a cohort of cancer patients undergoing immunotherapy with PD-1/PD-L1 blockade were also studies.

      Results:
      We included 23 patients at two institutions in Switzerland (University Hospital Basel and Cantonal Hospital Lucerne). Most patients have been treated with PD-1 blocking antibodies for at least 6 weeks at the time of vaccination. The frequency of irAEs was at 52.2% and 6 of 23 patients (26.1%) hat severe grade 3 or 4 irAEs. This frequency is significantly higher than in our general experience at our center (all grades 25.49%, grade 3 or 4 9.8%). There was no major difference over time in the generation of antibody titers in all three viral lines tested. Peripheral leukocyte counts and also cytokine/inflammatory chemokine levels did not change significantly shortly after vaccination.

      Conclusions:
      While the vaccination against seasonal influenza viruses seems to produce good serological protection and no short term toxicity of the vaccination could be observed, the increased rate of immunological toxicity is concerning and should be studied in a larger patient population.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      University Hospital Basel

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      113P - Clinical outcomes and quality of life (QoL) in adults with advanced refractory non-small cell lung cancer (NSCLC) patients receiving nivolumab (Nivo) as 2+ line treatment: Interim analysis of expanded access program (ID 391)

      12:30 - 12:30  |  Author(s): K.K. Laktionov, L.V. Bolotina, V.V. Breder, A.S. Danilova, F.V. Moiseenko, T.P. Nikitina, R.V. Orlova, E.A. Filippova, S.A. Protsenko, T.I. Ionova

      • Abstract

      Background:
      Therapy with immune checkpoints changed the paradigm of treatment in many solid tumors including NSCLC. Recently several drugs of this group were approved in second and first line. Still the data on their use in NSCLC is quite limited. The aim of this study was to evaluate benefits/risks of Nivo treatment within the expanded access program in refractory NSCLC patients both from physician’s and patient’s perspective. We report interim analysis on response rates, safety and QoL.

      Methods:
      Adult pts with advanced refractory NSCLC received Nivo 3 mg/kg q2w in accordance with expanded access program. Tumor response was assessed using RECIST v. 1.1. Adverse events (AEs) were evaluated by NCI CTCAE v3.0. QoL was assessed by RAND SF-36, symptom severity – by ESAS-R. Group comparisons were made using Mann-Whitney test.

      Results:
      From July of 2015 to December of 2016 with the median follow-up time of 11 weeks 141 pts were enrolled in 7 centers in RF. Clinical characteristics: 64.5% – males; median age – 60.5 (29 − 79); ECOG PS 0-1/2-3 – 78.3%/21.3%; former or current smokers – 71%; non squamous NSCLC – 65.2%; ≥2 lines of previous systemic treatment – 54%. At baseline pts had poor QoL comparing with healthy controls – 0.283 vs 0.505 (p < 0.001); 68.8% had moderate-to-severe symptoms. The most dramatic QoL worsening was observed for physical functioning and role functioning, p < 0.001. During 4 weeks of treatment Integral QoL index increased by 50% in 53% of pts. Efficacy was evaluated in 51 pts (median first evaluation – 9 weeks), PR – 6/51, SD – 30/51, PD – 15/51. Early deaths from cancer occurred in 7 pts; early deaths not related to cancer – 2 pts. 15 pts discontinued Nivo prematurely (<2 mos) because of rapid clinical worsening. 66 pts were not evaluated for response on cut-off. AEs were registered in 35 pts (median of Nivo treatment – 7 weeks); among them 8 with grade 3-4 AEs.

      Conclusions:
      QoL is dramatically compromised in advanced refractory NSCLC pts. Early data from this study supports that Nivo is effective and well tolerated by this patient population.

      Clinical trial identification:


      Legal entity responsible for the study:
      Multinational Center for QoL Research

      Funding:
      The study was supported by the grant of BMS

      Disclosure:
      K.K. Laktionov, L.V. Bolotina, V.V. Breder, A.S. Danilova, F.V. Moiseenko, T.P. Nikitina, R.V. Orlova, E.A. Filippova, S.A. Protsenko, T.I. Ionova: Study supported by the grant of BMS

    • +

      114P - Phase II study of nivolumab in patients with advanced non-small cell lung cancer (NSCLC) in Korea (ID 441)

      12:30 - 12:30  |  Author(s): E.K. Cho, J.H. Kang, J. Han, J.S. Lee, D. Kim, S. Kim, Y.J. Min, K.H. Lee, J. Kim, K. Park

      • Abstract

      Background:
      Nivolumab (BMS-936558/ONO-4538), a fully human IgG4, PD-1 immune-checkpoint inhibitor antibody, has shown durable clinical activity in several tumor types. Recently, two phase III studies (CheckMate-017 and -057) demonstrated that nivolumab improved overall survival (OS) than docetaxel in second-line of squamous (SQ) and non-squamous (NSQ) Non-Small Cell Lung Cancer (NSCLC), respectively. Here, we report the results of a phase II study to evaluate the efficacy and safety of nivolumab in Korean patients (pts) with previously treated advanced SQ and NSQ NSCLC.

      Methods:
      This study requires pts aged ≥ 20 years with ECOG Performance Status (PS) of 0 or 1, stage IIIB/IV or recurrent NSCLC and at least one prior chemotherapy including platinum containing regimen. Pts received nivolumab 3 mg/kg IV Q2W until progression or unacceptable toxicity. The primary endpoint in this study was the objective response rate (ORR) (RECIST v1.1).

      Results:
      Nivolumab was administered to 100 NSCLC pts (SQ: 44, NSQ: 56), male/female: 78 (SQ: 44, NSQ: 34)/22 (SQ: 0, NSQ: 22); PS 0/1: 14 (SQ: 6, NSQ: 8)/86 (SQ: 38, NSQ: 48); aged 29 to 80 [median: 66.5] years (SQ: 40 to 80 [median: 69.5], NSQ: 29 to 77 [median: 63.5]); Stage IIIB/IV/recurrence: 6 (SQ: 5, NSQ: 1)/91 (SQ: 37, NSQ: 54)/3 (SQ: 2, NSQ: 1)). In SQ and NSQ NSCLC, ORR was 15.9% (7/44) and 23.2% (13/56), respectively. Median progression-free survival was 2.6 mo and 5.3 mo, respectively. Complete Response was observed in 2.3% (1/44) and 1.8% (1/56), respectively. Median OS was 12.3 mo and 16.3 mo, respectively. Median follow-up was 8.9 mo and 12.3 mo, respectively. Most common adverse drug reaction (ADR) was decreased appetite 15.9% (7/44), followed by pyrexia 9.1% (4/44) in SQ NSCLC, and decreased appetite 12.5% (7/56), followed by pruritus 10.7% (6/56), fatigue 8.9% (5/56), pyrexia 5.4% (3/56) and nausea 5.4% (3/56) in NSQ NSCLC. Grade 3-4 ADR was observed in 6.8% (3/44) and 10.7% (6/56) of SQ and NSQ NSCLC, respectively. No interstitial lung disease and no grade 5 ADRs were observed in this study.

      Conclusions:
      Nivolumab was considered to be effective and used safely in Korean pts with SQ and NSQ NSCLC as well as in non-Korean pts with SQ and NSQ NSCLC.

      Clinical trial identification:
      NCT02175017

      Legal entity responsible for the study:
      Ministry of Food and Drug Safety in Korea

      Funding:
      Ono Pharmaceutical

      Disclosure:
      J.H. Kang: Honoraria; Bristol-Myers Squibb, Boehringer Ingelheim Consulting or Advisory Role; Bristol-Myers Squibb, Amgen Research Funding; AstraZeneca, Lilly. K. Park: Consulting or Advisory Role; Astellas Pharma, AstraZeneca, Boehringer Ingelheim, Clovis Oncology, Lilly, Hanmi, Kyowa Hakko Kirin, Novartis, Ono Pharmaceutical, Roche, Speakers\' Bureau; Boehringer Ingelheim Research Funding: AstraZeneca. All other authors have declared no conflicts of interest.

    • +

      115P - Immune related adverse events (irAE) and disease response with nivolumab in pre-treated advanced non-small cell lung cancer (NSCLC) (ID 428)

      12:30 - 12:30  |  Author(s): E. Connolly, G. Mallesara, I. Nordman

      • Abstract

      Background:
      IrAE with nivolumab are recognised. Incidence in practice and correlation with disease response in NSCLC has not been well characterised.

      Methods:
      A retrospective, descriptive analysis was carried out on 40 patients who received nivolumab on a compassionate access program from July 2015 to November 2016. Disease response was assessed by RECIST criteria.

      Results:
      The median age of patients was 63 years (range 28-80). 24 patients were female (60%). 17 received nivolumab as second line therapy, 12 as third line, 9 as fourth line, and 2 as fifth line. Best response included partial response (PR) in 15% (n = 6), stable disease (SD) in 40% (n = 16), progressive disease (PD) in 38% (n = 15) and unknown in 8% (n = 3). Treatment is ongoing in 30% (n = 12) (mean 6 months, range 2-18 months). 70% (n = 28) had irAE of any grade. Development of rash (n = 11; 28%) and pruritis (n = 10; 25%) were the most frequent irAE experienced. Grade 3 pruritus, pneumonitis, hepatitis, rash, arthralgia, and fatigue were seen. 2 patients developed grade 3 biopsy proven immune related colitis. Treatment requiring endocrinopathies occurred; hypothyroid (13%; n = 5), hyperthyroid (5%; n = 2) and hypoadrenal (5%; n = 2). Therapy was ceased due to grade 4 rash (n = 1), grade 3 hepatitis (n = 1), progressive neurological symptoms (n = 1) and psychiatric symptoms requiring hospital admission with no prior history of psychiatric illness (n = 1). The psychiatric and neurological symptoms resolved with treatment cessation and both patients have ongoing disease control on observation. A death occurred from suspected immune related hepatitis. Importantly, 5 of 6 patients who had PR developed grade 3 and above irAE. Of 13 patients who received treatment for greater than 3 months with SD, 11 developed irAE (85%). Of 10 patients on treatment for greater than 6 months with disease control (PR or SD); 7 developed grade 3 irAE or treatment requiring endocrinopathies. Of 10 non responders (PD within 3 months) 40% (n = 4) developed irAE; all grade 1. Clinically there appears to be a trend between response and development of irAE.

      Conclusions:
      The irAE are in line with published data however are of higher incidence and severity in this case series. There may be a trend of response and development of irAE.

      Clinical trial identification:
      not applicable

      Legal entity responsible for the study:
      Calvary Mater Newcastle

      Funding:
      Study conducted by Calvary Mater Newcastle. Nivolumab received on compassionate access programme from Bristol-Myers Squibb (BMS) however BMS have not been involved in data collection or interpretation.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      116P - A randomized study to evaluate safety of DCVAC/LUCA added to chemotherapy with carboplatin and pemetrexed vs. chemotherapy alone in patients with stage IV non-small cell lung cancer (ID 239)

      12:30 - 12:30  |  Author(s): H. Zhong, R. Zhong, B. Yan

      • Abstract

      Background:
      DCVAC/LuCa is an active autologous cellular immunotherapy consisting of autologous dendritic cells (DCs) loaded with NSCLC antigens (whole tumor cell antigens of tumor cell lines, like Her2/neu, MAGE-A3 and Survivin, et al.). DCVAC has the ability to induce an immune response, including cytotoxic CD8+ T cells, against tumor-associated antigens expressed by patients’ cancer cells. However, advanced-stage NSCLC with a heavy tumor burden establishes a harsh landscape for immunotherapy due to immune tolerance towards tumor antigens. Combination of DC treatment and chemotherapy is anticipated to achieve stronger immune responses than either of the treatments alone. AEs were collected in this study to evaluate the safety of DCVAC/LuCa added to chemotherapy with pemetrexed and carboplatin vs. chemotherapy alone in patients with stage IV non-small cell lung cancer.

      Methods:
      This is a randomized, open-label study. A total of 20 newly diagnosed stage IV, non-squamous, wild-type EGFR, ALK-negative or unknown NSCLC patients treated between January 2016- December 2016 in Shanghai Chest Hospital were enrolled.10 patients were randomized to group A: DCVAC/LuCa + chemotherapy (4-6 cycles of pemetrexed/carboplatin followed by pemetrexed as maintenance therapy); 10 patients were randomized to group B: 4-6 cycles of pemetrexed/carboplatin followed by pemetrexed as maintenance therapy.Patients in the group A started with DCVAC/LuCa (5 × 106 DCs/aliquots) treatment on Day 15 (+/- 3 days) of chemotherapy Cycle 3.The initial 5 doses of DCVAC/LCa were administered at a 3-week interval. The DCVAC/LuCa was then injected every 6 weeks up to the maximum number of 15 doses (75 × 106 DCs/15 aliquots). AEs were collected and analyzed.

      Results:
      The common adverse events in both group were chemotherapy related leukopenia, hemoglobin decrease etc. All AEs were grade 1 or 2 according to CTCAE V4.03, and there were no grade 4 toxicities or treatment-related deaths. One patient in group A got non-infectious fever and returned to normal without treatment.

      Conclusions:
      In patients with stage IV NSCLC, DCVAC/LuCa therapy was well tolerated with the favorable safety profile.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      Sotio Medical Research (Beijing) Co., Ltd

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      117P - Symptomatic pneumonitis in the irradiated lung after nivolumab: Three case studies (ID 295)

      12:30 - 12:30  |  Author(s): O. Roengvoraphoj, C. Eze, M. Li, F. Manapov

      • Abstract

      Background:
      Nivolumab is a feasible therapy option in patients with advanced NSCLC who progress after first-line conventional treatment. There is limited information about an overlapping toxicity of nivolumab when applied after primary multimodality treatment. Here, we describe symptomatic pneumonitis in the irradiated lung in patients undergoing second- or third-line nivolumab therapy.

      Methods:
      Case 1: A 66-year-old female patient who presented with squamous NSCLC stage ypT2a pN2 cM0 underwent adjuvant thoracic irradiation. Nivolumab was started 6 months’ post-radiotherapy when recurrent disease was detected on restaging CT. Twelve days after the first nivolumab treatment, the patient developed grade 2 dyspnea and cough.rnCase 2: A 76-year-old male patient with non-squamous NSCLC stage cT1a cN2 cM1b (single metastatic brain tumor) recieved intracranial stereotactic radiosurgery followed by thoracic RT to a total dose of 66Gy. Second-line nivolumab was started 6 months later and after the fourth cycle (70 days after the first nivolumab treatment), the patient developed grade 2 dyspnea and cough.rnCase 3: A 56-year-old female patient with metastatic NSCLC was treated with Cisplatin/Pemetrexed followed by irradiation to the brain and thorax. Due to systemic progression, second-line chemotherapy with docetaxel/nintedanib. Six months later, the patient was started on nivolumab. After 6 cycles (77 days after the first cycle of nivolumab), the patient developed grade 2 coughing and dyspnea.

      Results:
      In all patients comprehensive radiological and functional diagnostic as well as bronchoscopy were performed after onset of respiratory symptoms. Imaging analysis was strongly consistent with parenchyma changes in the irradiated lung volume receiving 15 to 20Gy. Nivolumab treatment was interrupted and patients were treated with systemic corticoids for one to two months with rash alleviation of symptoms.

      Conclusions:
      Three cases of symptomatic pneumonitis in patients with advanced NSCLC treated with nivolumab were described. Interruption of immunotherapy and systemic corticosteroid therapy for several weeks was necessary. Future prospective investigation of the described phenomenon is urgently indicated.

      Clinical trial identification:


      Legal entity responsible for the study:
      Deparment of Radiation Oncology LMU Munich

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      118P - Early onset of endocrine alterations during PD-1 blockade in advanced NSCLC patients (ID 381)

      12:30 - 12:30  |  Author(s): C.E. Onesti, A. Botticelli, M. Occhipinti, F.R. Di Pietro, I. Zizzari, C. Napoletano, M. Nuti, S. Lauro, F. Mazzuca, P. Marchetti

      • Abstract

      Background:
      Immune checkpoint inhibitors have been widely studied in recent years for solid tumors treatment, including lung cancer. Despite advantages observed in term of survival, new toxicities, such as endocrinopathies, have been observed. The most common are thyroid dysfunctions, hypophysitis and adrenal insufficiency. Diabetes mellitus and low level of testosterone are observed with anti-CTLA4 antibodies, but no data for anti-PD-1 and anti-PD-L1 are reported so far.

      Methods:
      Patients with histological diagnosis of squamous NSCLC, eligible for treatment with nivolumab, with ECOG PS 0-2 and adequate organs function were enrolled in our observational prospective study. All patients underwent blood sampling before starting treatment, after one and two months. All samples were analyzed for adeno-pituitary hormones (LH, FSH, ACTH, PRL, TSH) and for hormones secreted by target glands (FT3, FT4, 17-β-estradiol, testosterone, cortisol).

      Results:
      We enrolled 11 patients (6 M, 5 F; median age 65 y, range 44-82 y) affected by squamous NSCLC, receiving nivolumab in second line treatment. Three patients had hypothyroidism treated with levothyroxine in past medical history. The majority of patients (72.7%) showed endocrine alterations during treatment, three of which were symptomatic. Three males showed a reduction of testosterone level, in one case associated with decreased libido. A woman had a raised testosterone with hirsutism. Two patients experienced an increase in LH and FSH level and one patient a reduction. Moreover, we observed two cases of hyperthyroidism, one of which symptomatic in a patient with hypothyroidism in past medical history, two cases of increased ACTH level, one case of PRL reduction, one of 17-β-estradiol raise, one of cortisol reduction and one of cortisol raise. In all the cases endocrine alterations onset early during the treatment.

      Conclusions:
      We observe high rate of endocrine alterations in patients receiving nivolumab, in most cases asymptomatic. Many alterations observed are not usually evaluated and symptoms are often underestimated. More and wider studies could help to manage symptoms for a better quality of life and to investigate the mechanisms underlying endocrine disorders.

      Clinical trial identification:
      Not Applicable

      Legal entity responsible for the study:
      \"Sapienza\" University of Rome, Department of Medical Oncology

      Funding:
      \"Sapienza\" University of Rome, Department of Medical Oncology

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      119P - A phase 2 randomized open-label study of ramucirumab (RAM) plus first-line platinum-based chemotherapy in patients (pts) with recurrent or advanced non-small cell lung cancer (NSCLC): Final results from squamous pts (ID 174)

      12:30 - 12:30  |  Author(s): S. Thomas, R.C. Doebele, D. Spigel, M. Tehfe, M. Reck, S. Verma, A. Zimmermann, E. Alexandris, P. Lee, P. Bonomi

      • Abstract

      Background:
      Vascular endothelial growth factor receptors (VEGFR) and their ligands are key regulators of angiogenesis and implicated in pathogenesis of NSCLC. RAM, a human IgG1 monoclonal antibody, specifically binds to VEGFR-2 thereby preventing receptor activation and angiogenesis. In a phase 2 trial (NCT01160744) addition of RAM to platinum-pemetrexed chemotherapy demonstrated clinical activity and acceptable safety in pts with advanced non-squamous NSCLC; here we report data from squamous (SQ) NSCLC pts treated with RAM in combination with first-line gemcitabine plus platinum chemotherapy.

      Methods:
      Eligible pts had Stage IV SQ NSCLC, ECOG PS ≤ 2, and no prior VEGF/VEGFR therapy nor chemotherapy for stage IV disease. Pts were randomized 1:1 into either Arm C: gemcitabine 1000 mg/m[2] + carboplatin AUC=5/cisplatin 75 mg/m[2] (GEM + Cb/Cis) or Arm D: ramucirumab 10 mg/kg + gemcitabine + carboplatin/cisplatin (RAM + GEM + Cb/Cis). Pts received first-line therapy from 4 to 6 cycles (21-day cycle); patients in Arm D, in absence of progressive disease, entered a maintenance phase with RAM alone. Primary objective was progression-free survival (PFS). Secondary endpoints were overall survival (OS), objective response rate (ORR), disease control rate (DCR), change in tumor size (CTS), duration of response and safety.

      Results:
      140 pts were randomized (GEM + Cb/Cis: 69; RAM + GEM + Cb/Cis: 71). The median PFS was 5.4 m GEM + Cb/Cis and 5.6 m for RAM + GEM + Cb/Cis; HR 0.88 (90% CI, 0.64, 1.22; p = 0.52). Median OS was 11.3 m for GEM + Cb/Cis and 10.4 m for RAM + GEM + Cb/Cis; HR 0.93 (90% CI, 0.68, 1.27; p = 0.68). ORR was significantly improved in RAM + GEM + Cb/Cis (46.5%) compared to GEM + Cb/Cis (24.6%) (p = 0.007). No statistically significant difference was observed in DCR and CTS between the two arms. Grade ≥ 3 adverse events occurring in > 10% of pts on RAM administered arm were: anemia, neutropenia and thrombocytopenia.

      Conclusions:
      The primary endpoint of PFS was not met. The addition of RAM to GEM + Cb/Cis significantly improved ORR in pts with SQ NSCLC with no new unexpected safety findings.

      Clinical trial identification:
      NCT01160744 JVBL

      Legal entity responsible for the study:
      Eli Lilly and Company, Indianapolis, IN

      Funding:
      Eli Lilly and Company, Indianapolis, IN

      Disclosure:
      R.C. Doebele: Grants from Ignyta, Threshold Pharmaceuticals, Strategia; patent from NTRK1 FISH; royalties paid to Abbott Molecular; Licensing fees for biologic materials Ariad, Loxo, Chugai, Blueprint Medicines; other from Ariad, Trovagene, Guardant Health, AstraZeneca. D. Spigel: Novartis Speakers Bureau (uncompensated). M. Tehfe: Advisory board committee and Honoraria for speaker: Lilly, Celegene, BMS, Merck. M. Reck: Personal fees from Boehringer-Ingelheim, Hoffmann-La Roche, Lilly, MSD, BMS, AstraZeneca, Celgene, Merck, Pfizer, outside the submitted work. A. Zimmermann, E. Alexandris, P. Lee: Employee and stockholder of Eli Lilly and Company. P. Bonomi: Personal fees from Eli Lilly, Roche Genentech, Pfizer, Celgene, Bristol Myers Squibb, Astra Zeneca, Merck, outside the submitted work. All authors have declared no conflicts of interest.

    • +

      120P - Subgroup analysis of adenocarcinoma patients refractory to first-line chemotherapy from REVEL: A randomized phase III study of docetaxel with ramucirumab or placebo for second-line treatment of stage IV non–small cell lung cancer (NSCLC) (ID 423)

      12:30 - 12:30  |  Author(s): M. Reck, L. Paz-Ares, D. Moro-Sibilot, F.A. Shepherd, F. Cappuzzo, K.B. Winfree, E. Alexandris, A. Sashegyi, R. Varea, M. Pérol

      • Abstract

      Background:
      In the phase III REVEL trial, ramucirumab plus docetaxel significantly improved median overall survival (OS), median progression-free survival (PFS), and objective response rate (ORR) in patients with advanced NSCLC who progressed after first-line platinum therapy, independent of histology. The REVEL trial also showed that ramucirumab plus docetaxel therapy improved median OS, median PFS, and ORR in adenocarcinoma patients who were refractory to first-line platinum therapy and in patients categorized as rapid progressors. Here, we report safety and quality of life (QoL) outcomes in refractory adenocarcinoma patients who participated in the REVEL trial.

      Methods:
      Patients were refractory if they had a best response of progressive disease to first-line treatment. Patients were randomized 1:1 to receive docetaxel 75 mg/m[2] in combination with either ramucirumab 10 mg/kg or placebo every 21 days until disease progression, unacceptable toxicity, or death. Treatment-emergent adverse events (TEAEs) were assessed according to the NCI-CTCAE, version 4.0. Quality of life was measured by the Lung Cancer Symptom Scale (LCSS).

      Results:
      Of the 1253 patients randomized (ramucirumab + docetaxel: 628; docetaxel + placebo: 625), 17% (ramucirumab + docetaxel: 9%; docetaxel + placebo: 8%) were adenocarcinoma patients refractory to first-line therapy. The safety overview and LCSS scores are presented in the table.rnTable: 120PSafety and QoL Outcomes of Refractory Adenocarcinoma Patients Treated in REVELrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      TEAEs, n (%)Ramucirumab + Docetaxel N = 111Placebo + Docetaxel N = 101
      Any TEAE108 (97)101 (100)
      Grade ≥3 TEAEs82 (74)73 (72)
      Serious TEAEs47 (42)48 (48)
      TEAEs leading to discontinuation6 (5)4 (4)
      TEAEs leading to death4 (4)11 (11)
      LCSS scores, time to deteriorationHazard Ratio (95% CI)
      Loss of appetite1.45 (0.91, 2.32)
      Fatigue0.90 (0.58, 1.41)
      Cough1.29 (0.77, 2.14)
      Dyspnea1.06 (0.64, 1.76)
      Hemoptysis1.55 (0.59, 4.07)
      Pain1.14 (0.71, 1.84)
      Symptom distress1.12 (0.69, 1.81)
      Activity level1.01 (0.64, 1.59)
      Global QoL0.98 (0.63, 1.52)
      Total LCSS0.81 (0.47, 1.41)
      ASBI0.83 (0.46, 1.50)
      rnNote: The primary LCSS analysis was time to deterioration, defined as the time from randomization to the first 15 mm increase. ASBI, average symptom burden index; CI, confidence interval; LCSS, Lung Cancer Symptom Scale; QoL, quality of life; TEAE, treatment-emergent adverse event.rn

      Conclusions:
      Our analysis did not identify any new safety concerns or increased detriment in QoL for this subgroup of patients. Safety outcomes for refractory adenocarcinoma patients were consistent with the outcomes for refractory patients with all histologies and the intent-to-treat population.

      Clinical trial identification:
      NCT01168973

      Legal entity responsible for the study:
      Eli Lilly and Company

      Funding:
      Eli Lilly and Company

      Disclosure:
      M. Reck: Personal fees from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly and Company, Hoffmann-La Roche, Merck Sharp & Dohme, Novartis, and Pfizer. L. Paz-Ares: Personal fees from AMGEM, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, CLOVIS, Eli Lilly and Company, Hoffmann-La Roche, Merck Sharp & Dohme, Novartis, and Pfizer. D. Moro-Sibilot: Personal fees from AMGEN, Ariad, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly and Company, Hoffmann-La Roche, Merck Sharp & Dohme, Novartis, and Pfizer. F.A. Shepherd, M. Pérol: Personal fees from Eli Lilly and Company. F. Cappuzzo: Personal fees from AstraZeneca, Hoffmann-La Roche, and Pfizer. K.B. Winfree: Employee of Eli Lilly and Company and reports personal fees from Eli Lilly and Company. E. Alexandris: Employee of Eli Lilly and Company. A. Sashegyi, R. Varea: Employee and shareholder of Eli Lilly and Company.

    • +

      121P - Safety and efficacy of bevacizumab in "fragile" patients with advanced non-small cell lung cancer (ID 492)

      12:30 - 12:30  |  Author(s): C. Maragkos, D. Grapsa, G. Koumakis, A. Polyzos, K. Syrigos

      • Abstract

      Background:
      Bevacizumab is the first antiangiogenic agent approved for treatment of advanced non-squamous NSCLC. Elderly or other “fragile” patients, such as those with poor performance status (PS), comprise the majority of the NSCLC population, but are typically underrepresented in clinical trials. The primary aim of this study was to investigate the efficacy and safety of first-line bevacizumab in “real-world” patients with advanced NSCLC.

      Methods:
      The medical records of all patients with histologically or cytologically confirmed advanced-stage non-squamous NSCLC, treated with first-line bevacizumab (plus chemotherapy) at the Oncology Clinic of “Sotiria” Athens University Hospital between January 2008 and December 2012 were retrospectively reviewed. Selected patients were stratified according to age, i.e. elderly (> 70 years) vs non-elderly (≤70 years), ECOG PS (0-1 vs 2-3) and other demographic, clinicopathological and treatment data. Main outcome measures were overall survival (OS) and treatment-related toxicity.

      Results:
      A total of 145 cases (mean age/SD=61.7 ± 10.6 years) were included. 23,4% of patients were elderly, while 15.8% had poor PS (2-3). Maintenance bevacizumab (with or without pemetrexed) was administered in 24.1% of all cases, including only patients with good PS, and mainly non-elderly. The presence of comorbidities was significantly associated with poor PS and age> 70 years. No statistically significant difference with regard to OS, or type and frequency of side effects was observed between elderly and non-elderly patients. A similar toxicity profile was also observed between patients with good versus poor PS. Absence of maintenance bevacizumab and poor PS were both significantly associated with reduced OS, both in univariate [HR (95% CI): 0.45 (0.22-0.9); p = 0.023 and HR (95% CI): 2.16 (1.29-3.64); p = 0.004, respectively] and in multivariate analysis [[HR (95% CI): 0.47 (0.22-0.99); p = 0.048 and HR (95% CI): 1.76 (1.04-3.00); p = 0.036, respectively].

      Conclusions:
      Our findings suggest that first-line bevacizumab may show similar efficacy and toxicity in elderly versus non-elderly patients with advanced NSCLC. Further prospective data are needed to confirm these observations.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Athens

      Funding:
      University of Athens

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      122P - Response to Metabolically Supported Chemotherapy (MSCT) with weekly carboplatin/paclitaxel in advanced stage/metastatic (Stage IV) non-small cell lung cancer: A survival, efficacy and tolerability analysis (ID 184)

      12:30 - 12:30  |  Author(s): M.S. Iyikesici, A. Slocum, A.K. Slocum, E. Turkmen, F.B. Berkarda

      • Abstract

      Background:
      Metabolically supported chemotherapy (MSCT), is defined as the application of standard chemotherapy protocols concomitant to the administration of pharmacological doses of regular insulin and the development of hypoglycemia, and following fasting starting the previous day. This study aims to evaluate the efficacy and the tolerability of MSCT combining carboplatin and paclitaxel in the treatment of advanced/metastatic non-small cell lung cancer (NSCLC).

      Methods:
      This study is a retrospective single center analysis of all patients diagnosed with metastatic (stage IV) NSCLC that received MSCT combining carboplatin and paclitaxel at our clinic between March 2010 and June 2015.

      Results:
      44 patients were included in our study. While 39 (88.6%) of the patients were male, their median age was 65 years (range 35-87). Adenocarcinoma was diagnosed in 34 (77.3%) of the patients. 18 (40.9%) of the patients had brain metastases and ECOG PS was ≥2 in the case of 36 (81.8%) of the patients. When the response evaluation for this study was carried out at the end of 6 cycles of treatment, 42 out of 44 patients included in this analysis remained alive. Statistical analysis revealed a mean overall survival (OS) of 43.4 months, mean progression-free survival (PFS) of 41.8 months and 1-year survival rate of 86.1%.

      Conclusions:
      This study demonstrates that a metabolically supported form of applying weekly paclitaxel at a dose of 75 mg/m[2] combined with weekly carboplatin at a dose of AUC 2 may bring about remarkable improvements in the survival rates of patients with advanced and metastatic NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Kemerburgaz University Bahcelievler Medical Park Hospital and ChemoThermia Oncology Center

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      123P - A retrospective study in Chinese patients: Is there a role of nanoparticle albumin bound paclitaxel in advanced NSCLC? (ID 393)

      12:30 - 12:30  |  Author(s): Y. Zhu, P. Xing, L. Shan, S. Chen, X. Hao, J. Li

      • Abstract

      Background:
      We aimed to report the safety and short-term efficacy of nanoparticle albumin bound paclitaxel (Nab-PTX) in advanced non-small cell lung cancer (NSCLC) in Chiina as a second line and later treatment, and explored its efffcacy affected by previous paclitaxel treatment wether or not.

      Methods:
      Advanced NSCLC patients who failed in prior treatment and received weekly Nab-PTX regimen on days 1, 8 (a dose of 130 mg/m2/week) treatment were included. The primary efficacy endpoint was progression-free survival (PFS). Toxicity was evaluated with NCI-CTCAE 3.0.

      Results:
      A total of 98 patients at the Cancer Institute & Hospital of the Chinese Academy of Medical Sciences (Beijing, China) between June 2010 and July 2015 were enrolled. The median PFS and overall survival (OS) were 4.34 months (95% CI 3.508 to 5.165 months) and 11.73 months (95% CI 9.211 to 14.247 months), respectively. PFS was no significant difference between patients with previous paclitaxel treatment and without previous paclitaxel treatment (median, 4.11 versus 4.53 months, respectively, p = 0.195). OS was also no significant difference between the two arms (median, 9.69 versus 14.62 months, respectively, p = 0.190). The objective responses rate (ORR) and disease control rate (DCR) of all patients were 22.4% and 74.5%, respectively. The ORR and DCR were 23.0% and 70.5%, respectively in one arm with previous paclitaxel treatment, while in another arm without previous paclitaxel treatment, the results were 21.6% and 81.1%. No significant difference in ORR (p = 0.533) and DCR (p = 0.244) between the two arms. Grade 3 or higher adverse events (AEs) of all patients was neutropenia (25.5%), leukopenia (12.4%), peripheral neuropathy (5.1%), myalgia/arthralgia (5.1%), anaemia (3.1%), and fatigue (1.0%), respectively.

      Conclusions:
      The Nab-PTX was effective and well tolerated as second line or later treatment in advanced NSCLC patients. Even used paclitaxel treatment previously did not affect the efficacy and PFS of Nab-PTX.

      Clinical trial identification:
      Preview and Finish

      Legal entity responsible for the study:
      Junling Li

      Funding:
      Junling Li

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      124P - Driverless lung carcinoma: Impact of expanded RNA and protein-based testing on detection of actionable biomarkers (ID 396)

      12:30 - 12:30  |  Author(s): Z. Gatalica, J. Xiu, W. Chen, T. Maney, A. Voss, N. Plowman

      • Abstract

      Background:
      In recent years, massively parallel-gene sequencing (NGS) has improved the detection of targetable mutations in lung cancer (NSCLC). We have identified NSCLC cases that, despite extensive DNA sequencing and targeted in-situ hybridizations (ISH) for ROS1, ALK and cMET alterations, have no druggable target or conventional lung cancer pathogenic mutation identified ("driverless" cancer). Expanded platform testing (RNA and protein-based) was performed on these driverless cases in order to assess the ability of these technologies to identify actionable drug targets.

      Methods:
      A review of 522 NSCLC cases (Caris Life Sciences, Phoenix, AZ) tested with NGS (592 gene sequencing panel, Agilent SureSelectXT; Illumina NextSeq) and ISH yielded 21 patients (F:M=12:9) without characteristic genetic alterations. Expanded testing included an RNA-based fusion panel (52 genes, Archer FusionPlex) and protein-expression (IHC) testing for EGFR, TS and PD-L1.

      Results:
      Expanded platform profiling identified targetable NTRK gene fusions (NTRK3:ETV6 and NTRK1:TPM3) in 2 cases and c-MET exon 14 skipping in 1 case. IHC identified PD-L1 expression in 7 (3 low and 4 high TPS), EGFR over-expression (H-score>200) in 7, and TS under expression in 13 cases. Initial NGS panel identified 2 low allele frequency pathogenic mutations (PIK3CA and GNAS), and 3 gene amplifications (MDM2, CDK4 and CDKN2A), as potential non-characteristic drivers in 4 cases. Total mutational load (TML) range was 1-10/Mb (mean 5.2/Mb).

      Conclusions:
      With the routine use of NGS a small proportion of cases (4.2%) remain without standard biomarker-guided therapy recommendation. They are characterized by a lower TML (5.2/Mb) than reported for NSCLC (e.g. TCGA mean: 8.9/Mb). A significant benefit from expanded multiplatform testing (RNA- and protein-based) included detection of biomarkers for immune check point inhibitors (33% eligible) and targeted therapies (23% eligible: NTRK and c-Met inhibitors). Over-expression of EGFR and under expression of TS (57% and 72%. respectively) could provide additional information for therapy guidance in specific cancer types.

      Clinical trial identification:
      Not applicable

      Legal entity responsible for the study:
      Caris Life Sciences

      Funding:
      Caris Life Sciences

      Disclosure:
      Z. Gatalica, A. Voss: Employment: Caris Life Sciences. J. Xiu, W. Chen, T. Maney: Employment: Caris LIfe Sciences. All other authors have declared no conflicts of interest.

    • +

      125P - Clinical research platform into molecular testing, treatment and outcome of non-small cell lung carcinoma Patients (CRISP): A prospective German registry in stage IV NSCLC, AIO-TRK-0315 (ID 388)

      12:30 - 12:30  |  Author(s): F. Griesinger, W.E.E. Eberhardt, N. Marschner, M. Jänicke, A. Fleitz, J. Sahlmann, A. Karatas, A. Hipper, M. Sebastian, M. Thomas

      • Abstract

      Background:
      Treatment in NSCL is quickly evolving and new agents make it to the routine practice at a rapid pace. Whether outcome and PRO data generated in clinical trials with often narrow inclusion and exclusion criteria will hold up in the routine practice is of high interest, especially due to the increasing costs of new drugs. Therefore, registry data are of ever increasing importance to patients, physicians and reimbursement institutions.

      Methods:
      Therefore, we have started a prospective, clinical registry for patients with metastatic NSCLC. The purpose of CRISP is to set up a national clinical research platform to document representative data on molecular testing, sequences of systemic therapies and other treatment modalities, course of disease in patients with advanced or metastatic NSCLC in Germany not amenable to curative treatment. A particular focus is on molecular biomarker testing of patients before the start of 1[st] -line treatment. The data shall be used to assess the current state of care and to develop recommendations concerning topics that could be improved. PRO assessment will provide large-scale data on quality of life and anxiety/depression for real-life patients in routine practice. In addition, two questionnaires (concerning individual quality of life and patient-caregiver communication) will be validated in German patients with metastatic NSCLC. Furthermore, CRISP will set up a decentral tissue annotation for future collaborative, investigational scientific biomarker testing. This study will be carried out in up to 150 representative cancer centers in all therapeutic sectors in Germany. More than 8000 patients will be recruited and followed up to a maximum of 3 years, respectively until death.

      Results:
      The first patients have been included in Dec. 2015. As of Jan. 2[nd] 2017, 71 centers are recruiting patients and 529 patients have been recruited. Preliminary data will be presented at the meeting for molecular testing, demographic data as well as treatment stratification in the 1[st] line setting.

      Conclusions:
      The registry CRISP will be the first to present representative real life data, covering all treatment settings of patients with NSCLC in Germany.

      Clinical trial identification:
      NCT02622581

      Legal entity responsible for the study:
      AIO-Studien-gGmbH

      Funding:
      CRISP is supported by Grants from AstraZeneca GmbH, Boehringer Ingelheim Pharma GmbH & Co. KG, Bristol-Myers Squibb GmbH & Co. KGaA, Celgene GmbH, Lilly Deutschland GmbH, MSD Sharp & Dohme GmbH, Novartis Pharma GmbH, and Pfizer Pharma GmbH.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      126P - The clinical impact of multiplex ctDNA gene analysis in lung cancer (ID 476)

      12:30 - 12:30  |  Author(s): S. Geva, T. Twito, A. Dvir, L. Soussan-Gutman, M. Ilouze, L.C. Roisman, E. Dudnik, A. Zer, R.B. Lanman, N. Peled

      • Abstract

      Background:
      Next-generation sequencing (NGS) of cell-free circulating tumor DNA (ctDNA) enables a non-invasive option for comprehensive genomic analysis of lung cancer patients. In this study, we evaluated the clinical utility of ctDNA sequencing on treatment strategy.

      Methods:
      In this retrospective study, data was collected from files of 109 NSCLC patients at the Thoracic Cancer Unit at Rabin Medical Center, Israel, between 2014-2017. Plasma samples from advanced non-small cell lung cancer (NSCLC) patients were analyzed by a commercial test (Guardant 360[TM]), using massively parallel paired-end synthesis to sequence a targeted gene panel. This test allows the detection of somatic alterations such as point mutations, indels, fusions and copy number amplifications.

      Results:
      109 consecutive NSCLC patients were included in this study. Median age at diagnosis was 63 years, 81% had adenocarcinoma. 39% (43/109) performed ctDNA analysis before 1[st] line therapy (Group A) and 61% (66/109) on progression (Group B), among them 42% (28/66) after progression on EGFR TKI (Group B1). ctDNA analysis yielded lung cancer related actionable mutations in 40% (44/109) of the patients; 32% (14/43) in group A and 45% (30/66) in group B; 71% (20/28) in group B1. Treatment decision was taken toward targeted therapy subsequent to NGS analysis in 27% of patients. 68 individual actionable genomic alterations were found (table).rnTable: 126PGenetic alterations frequencies among groups A, B and B1rn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      Group A: Upfront NGS (n = 43, 19 individual mutations)Group B: NGS on progression (n = 66, 49 individual mutations)Group B1: NGS on progression on EGFR TKIs (n = 28, 34 individual mutations)
      EGFR Sensitizing52% (10/19)EGFR Sensitizing45% (22/49)EGFR Sensitizing59% (20/34)
      MET16% (3/19)MET25% (12/49)EGFR T790M23% (8/34)
      ERBB210.5% (2/19)EGFR T790M16% (8/49)MET12% (4/34)
      BRAF V600E10.5% (2/19)RET6% (3/49)ERBB23% (1/34)
      RET10.5% (2/19)ERBB26% (3/49)ALK3% (1/34)
      rnrnALK2% (1/49)rnrn
      rnrnResponse assessment (RECIST) for 20 patients with evaluable response to targeted therapy showed complete response in 5% (1/20), partial response in 35% (7/20), stable disease in 25% (5/20) and progressive disease in 35% (7/20). Response rate was 20% (1/5) for group A, 47% (7/15) for group B, among them 67% (6/9) for group B1. Total objective response rate (ORR) was 40%.

      Conclusions:
      Comprehensive ctDNA testing revealed possible treatment options for 40% of patients analyzed. The highest impact was seen in the progressors on EGFR therapy. These positive results emphasize the utility of liquid biopsy analysis to guide clinicians to select the right therapy for the right patient.

      Clinical trial identification:


      Legal entity responsible for the study:
      Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel.

      Funding:
      Institute of Oncology, Davidoff Cancer Center, Rabin Medical Center, Petach Tikva, Israel.

      Disclosure:
      S. Geva: Travel grant from Teva Pharmaceuticals. Honorarium from Guardant Health, Inc. T. Twito, A. Dvir, L. Soussan-Gutman: Employee of Oncotest (subsidiary of Teva pharmaceuticals), the distributor of Guardant360 in Israel. E. Dudnik: Consultant of BI. Honorary lectures for BI, Roche, AstraZeneca and MSD. R.B. Lanman: Employee with stock ownership in Guardant Health, Inc. N. Peled: Consultant for Pfizer, BI, Roche, AZ, MSD, BMS, Lilly, Novartis, and NovellusDx. All other authors have declared no conflicts of interest.

    • +

      127P - Prognostic significance of advanced lung cancer inflammation index (ALI) In untreated and treated stage IV non-small cell lung cancer (NSCLC): An Australian cancer centre experience (ID 172)

      12:30 - 12:30  |  Author(s): H. Mandaliya, M. Jones, C. Oldmeadow, I. Nordman

      • Abstract

      Background:
      The role of advanced lung cancer inflammation index (ALI) at diagnosis is increasingly explored as an independent prognostic factor of survival in stage IV NSCLC. Post-treatment changes in ALI and the potential impact on survival are not clear in this group of patients. We aimed to evaluate prognostic role of ALI in stage IV NSCLC at diagnosis as well as post treatment.

      Methods:
      A retrospective descriptive study was conducted for patients with stage IV NSCLC actively treated at Calvary Mater Newcastle, Australia from 2010 to 2015. Advanced lung cancer inflammation index (ALI = BMI x Albumin/NLR; BMI=Body mass index, NLR=Neutrophil-to-lymphocyte ratio, Albumin=Serum albumin g/dl) values calculated at diagnosis and post first cycle chemotherapy/targeted treatment. Demographic variables summarised and estimates of Kaplan-Meier (KM) survival distribution for overall survival (OS) generated. Extended Cox regression used to derive OS hazard ratios of predictive variables. Project approved by Research Ethics Committee.

      Results:
      A total of 279 patients with Stage IV NSCLC were treated during the study time. Baseline ALI was available for 276 patients and post first treatment cycle for 189 patients. The Cox PH model suggested ALI was a prognostic factor for OS, hazard ratio = 0.982 (95% CI: 0.973-0.991). Post first-cycle treatment, individuals with mean baseline ALI, had a hazard ratio = 0.391 (95% CI: 0.211-0.629) that was also increasing multiplicatively as a function of baseline ALI. At baseline, KM estimate suggested median survival was 6.23 months (95% CI: 4.83-9.27) for patients with ALI < 18 compared to 14.70 months (95% CI: 11.63-18.20) for those with ALI > 18. Post first-cycle treatment, not adjusted for baseline, patients with ALI < 18 median survival was 5.23 months (95% CI: 3.27-9.07) compared to 12.67 months (95% CI: 10.47-15.13) for ALI > 18.

      Conclusions:
      ALI has a strong association with survival from baseline and post first-cycle treatment adjusted for baseline ALI. High pretreatment and post treatment ALI predicts for a longer survival while low pretreatment and post treatment ALI predicts for a shorter survival.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hunter New England Human Research Ethics Committee

      Funding:
      Department of Medical Oncology, Calvary Mater Newcastle, Australia Hunter Medical Research Institute, Australia

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      128P - Serum DKK-1 as a clinical and prognostic factor in non-small cell lung cancer patients with bone metastases (ID 192)

      12:30 - 12:30  |  Author(s): R. Qiao, T. Chu, B. Han, R. Zhong, Q. Chang, J. Teng, J. Pei

      • Abstract

      Background:
      The Wnt/β-catenin pathway plays a crucial role in tumor pathogenesis, specifically in cell proliferation, angiogenesis, motility and invasiveness. The activity of Wnt family ligands is antagonized by several secreted factors including Dickkopf (DKK). A member of the Dickkopf family, DKK-1, is a 35 kDa secreted protein that is a potent inhibitor of Wnt/β-catenin signaling. The study was designed to evaluate the association between serum Dickkopf-1(DKK-1) and non- small cell lung cancer(NSCLC) bone metastases.

      Methods:
      Serum DKK-1and CEA levels were quantified in 318 NSCLC patients, 140 with osseous metastases and 178 with extraosseous metastases. We used receiver operating characteristics (ROC) to evaluate the predictive qualities of these parameters for bone metastases.

      Results:
      Serum DKK-1 levels were significantly higher in patients with osseous metastases compared with patients with extraosseous metastases (P < 0.001). ROC curves showed that the optimum cutoff was 311.8 ng/ml (area under curve [AUC] 0.791, 95% confidence interval [CI] 0.739–0.843, sensitivity 77.1% and specificity 71.4%). ROC analysis also showed that testing both DKK-1 and CEA increased the detection accuracy for NSCLC bone metastases compared with CEA alone (AUC 0.797, 95% CI 0.746–0.848, sensitivity 82.9% and specificity 68.9%; DKK-1 plus CEA vs. DKK-1 alone P = 0.370; DKK-1 plus CEA vs. CEA alone P = 0.0001). Thus, serum DKK-1 correlated with the number of bone lesions (P = 0.042). In osseous metastases group, Kaplan–Meier analysis showed that patients with high serum DKK-1 levels had poorer overall survival than patients with low serum DKK-1 levels (P = 0.025), and multivariable analyses showed serum DKK-1 to be an independent prognostic factor for overall survival (P = 0.029).

      Conclusions:
      Our data shows that serum DKK-1 levels are increased in NSCLC patients with bone metastases. More importantly this is the first report to show that high serum DKK-1 levels are associated with the extent of bone metastases and poor survival in NSCLC patients with bone metastases.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      The Science and Technology Development Fund of Shanghai Chest Hospital (Grant No. 2014YZDC10101)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      129P - Procathepsin D as a novel diagnostic marker for malignant pleural effusion of lung cancer (ID 341)

      12:30 - 12:30  |  Author(s): C.Y. Lee, J.Y. Hong, M. Lee, S.H. Jang

      • Abstract

      Background:
      Approximately 20% of pleural effusions are due to malignancy, and 50% of these are due to primary lung cancer. However, the etiology of the effusions is often obscure and various diagnostic procedures may be required in order to find their cause. Cathepsin D (CD) as a lysosomal aspartic protease is one of the most important intracellular enzymes considered to be substantially involved in tumor invasion. Procathespin D (pCD) is overexpressed and secreted by cells of various tumor types. The aim of this study is to evaluate the serum and pleural fluid levels of CD and pCD as a differential diagnostic tool in patients with pleural effusion caused by malignant or benign process.

      Methods:
      Patients and pleural fluid collection: The present study included 85 patients (40 patients with lung cancer, 30 patients with tuberculosis, 10 patients with parapneumonic effusion, 5 patients with transudate effusion) who underwent thoracentesis and pleural biopsy. Clinical and pathology data, including tumor type were acquired for all patients. Analysis of CD and pCD: Amounts of CD in the pleural fluid and serum were determined with a cathepsin D ELISA kit and pCD with a sandwich ELISA method.

      Results:
      Concentration of pleural CD was not significantly different in patients with malignant pleural effusion compared with non malignant effusion (mean 13.34 vs. 16.72 ng/ml; p > 0.05), and also the concentration of serum CD was not significantly different (mean 41.34 vs. 36.52 ng/ml; p > 0.05). Plasma pCD of malignant pleural effusion and non malignant effusion were 42.03 ng/ml and 38.59 ng/ml, respectively (p > 0.05) but pleural pCD of malignant pleural effusion and non malignant effusion were 84.24 ng/ml and 33.82 ng/ml, respectively with significance (p < 0.05).

      Conclusions:
      In this small cohort, the pleural pCD of lung cancer patients showed elevated levels compared with that of non malignant effusion. The pleural fluid levels of pCD could be a diagnostic marker of malignant effusion with lung cancer and further prospective studies might shed more light on the value of CD and pCD in the diagnostics of pleural effusion.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chang Youl Lee

      Funding:
      Hallym University Medical Center

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      130P - EGFR mutations in lung adenocarcinoma and brain metastases: A Croatian single institution experience (ID 512)

      12:30 - 12:30  |  Author(s): K.B. Sreter, S. Kukulj, S. Smojver-Jezek, S. Seiwerth, M. Jakopovic, M. Samarzija

      • Abstract

      Background:
      The brain, bones and lungs are common sites of metastasis in non-small cell lung cancer. We aimed to investigate the metastatic pattern of epidermal growth factor receptor (EGFR) mutations by analyzing the incidence of different metastatic sites in EGFR positive (+) lung adenocarcinoma patients with brain metastases (BM).

      Methods:
      Data from medical records at the Clinic for Respiratory Diseases "Jordanovac" were collected for this retrospective cohort study. Caucasian Croatian patients with primary lung adenocarcinoma (PLA) and EGFR+ mutation status (2014-2015) were included.

      Results:
      Of 116 EGFR+ patients, 24 (21.0%) were diagnosed with BM. The majority of EGFR+ patients (n = 17, 70.8%) were less than 65 years old at BM diagnosis. There were fewer males (n = 4, 16.7%) than females. Only four patients (all female, 16.7%) were active smokers at diagnosis of PLA. Median age at diagnosis of BM was 62 years (range: 43-78 years). Most patients (n = 20, 83.3%) had good performance status (PS, ECOG 0-1) and normal to increased body mass index (n = 17, 70.8%). Weight loss at presentation was reported by 10 patients (41.6%). The majority (n = 16, 66.7%) initially presented to the emergency department. In 8 patients (33.3%), symptoms related to BM appeared prior to or at the same time as the PLA. Oral tyrosine kinase inhibitor (TKI) treatment was received in second line (n = 8, 33.3%) after progression of disease following first line chemotherapy. Most patients with multiple BM (n = 18, 75.0%) received whole brain palliative radiotherapy; three could not due to poor PS. The main extracranial sites of metastases were bone (n = 16, 66.7%), liver (n = 7, 29.2%) and pleura (n = 19, 79.2%). The most common EGFR mutations were single exon 19 deletion (n = 13, 54.2%) and exon 21 L858R (n = 5, 20.8%). One patient (4.2%) had a double mutation (exon 19 and 21) and another (4.2%) had a rare single exon 18 mutation. Exon 20 T790M mutation occurred in 16.7% of patients (n = 4). The median overall survival (mOS) was 7.7 months versus 20.1 months from time of diagnosis of BM versus from PLA, respectively.

      Conclusions:
      Early diagnosis of BM and extracranial metastases in EGFR mutant PLA patients is key to improving clinical outcomes, quality of life, and overall survival.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital Centre Zagreb, Clinic for Respiratory Diseases \"Jordanovac\"

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      131P - T790M co-exists with other secondary resistance mechanisms in EGFR mutation positive NSCLC and are associated with inferior outcomes (ID 159)

      12:30 - 12:30  |  Author(s): R. Teng, A. Takano, W. Tan, M. Ang, C. Toh, Q. Ng, W. Lim, E. Tan, T. Lim, D.S. Tan

      • Abstract

      Background:
      Compared to conventional chemotherapy, NSCLC adenocarcinoma patients with EGFR-activating mutations (M+) have superior response to EGFR TKIs. However, all patients develop acquired resistance (AR) to TKIs, most commonly through T790M mutation. Due to intratumour heterogeneity, additional non-T790M mechanisms of AR could coexist with the T790M mutation. Little is known about the prevalence and clinical outcomes of patients with co-existing mechanisms of AR.

      Methods:
      145 patients with EGFR M+ NSCLC underwent a repeat biopsy after developing AR to TKI therapy. All specimens underwent histological review, while 116 (80%) underwent molecular profiling with multiple parallel high-sensitivity assays (including a 29-gene NGS panel) to detect genetic aberrations linked to TKI resistance. Kaplan Meier survival curves were plotted and compared with the log-rank test.

      Results:
      53.4% TKI-resistant patients (n = 57/106) were T790M-positive, while 61% patients had a non-T790M mechanism of AR including CMET polysomy or amplification, PTEN Loss, PIK3CA substitution and small cell change. 10 patients had lost their original EGFR-activating mutation. Up to 42% (n = 34/80) had a mutation in an NSCLC driver gene (including BRAF, DDR2, NRAS) detected by a 29-gene panel. 51% patients treated with 1[st]-generation gefitinib or erlotinib (n = 101) were T790M-positive, compared to only 20% of patients treated with 2[nd]-generation afatinib (n = 15). There was also a difference (p = 0.071) in T790M prevalence between patients treated with TKI post-progression (37/67, 55%) and those treated with chemotherapy (15/41, 36.5%). Notably, almost half the T790M-positive subjects harboured co-existing non-T790M mechanisms of AR (28/57, 49.1%). These patients had inferior OS of 42 months (95% CI 36.9-47.1) compared to patients with solely T790M (58 months, 95% CI 36.5-132.8).

      Conclusions:
      While T790M is widely-accepted as the predominant mechanism of AR, the spectrum of mutations can vary greatly depending on the type of prior therapy. Moreover, other mechanisms of AR could coexist with T790M and portend inferior clinical outcomes, suggesting that targeting other mechanisms of AR beyond T790M may be clinically-relevant.

      Clinical trial identification:


      Legal entity responsible for the study:
      Ministry of Health Singapore

      Funding:
      National Cancer Centre Singapore

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      132P - Real world treatment patterns and outcomes for mNSCLC patients receiving second and third-line therapy in Germany (ID 349)

      12:30 - 12:30  |  Author(s): N. Yu, I. Reyes-Rivera, J. Hipp, M. Zou, N. Pillai, S. Hammerschmidt

      • Abstract

      Background:
      Metastatic non-small cell lung cancer (mNSCLC) is the leading cause of cancer death worldwide. Recent approvals of targeted therapies, especially immunotherapies, may change the treatment landscape, but little is known about their real-world impact. This study (MO39087) assessed real world treatment patterns and outcomes of patients (pts) receiving second- (2L) and/or third-line (3L) therapy for mNSCLC in Germany.

      Methods:
      A retrospective, non-interventional, observational study was performed with secondary data from the German Oncology Network of outpatient treatment centres. Participating office-based oncologists across Germany provided de-identified longitudinal patient-level data via electronic case report forms for mNSCLC pts. Pts were followed from start of 2L and 3L (index dates) until death/loss to follow up. Pt characteristics and treatment patterns were described. Overall survival (OS) and progression-free survival (PFS) from index dates were calculated using the Kaplan-Meier method.

      Results:
      Of pts diagnosed between Jan 2007–Jul 2016, 340 and 123 pts started 2L and 3L therapy, respectively. Of these, 56% in 2L and 59% in 3L were treated in 2014 or later. The top 3 regimens in 2L were monotherapies of docetaxel (n = 55, 16%), vinorelbine (n = 41, 12%) and pemetrexed (n = 40, 12%). In 3L, the top 3 regimens were monotherapies of docetaxel (n = 19, 15%), vinorelbine (n = 17, 14%) and gemcitabine (n = 11, 9%). Since 2014, an increase in pts on targeted therapies was seen in 2L (25%, 47/192, n = 15 for immunotherapy) vs before 2014 (14%, 20/148). The same trend was also seen in 3L (since 2014, 25%, 18/72, n = 9 for immunotherapy; before 2014, 16%, 8/51). Median OS and PFS was 8 mths (95% CI 6.6–8.9) and 5 mths (95% CI 4.2–5.3) for 2L, and 9 mths (95% CI 6.4–10.3) and 4 mths (95% CI 3.0–5.9) for 3L.

      Conclusions:
      Despite a trend of increased uptake of 2/3L targeted therapies since 2014, prescription levels were still low and single agent chemotherapy still dominates. Given the small number of pts and short observation time, especially for immunotherapy, further studies will be needed when a broader uptake of immunotherapy is expected, to fully assess the impact on treatment patterns and survival.

      Clinical trial identification:
      M039087

      Legal entity responsible for the study:
      Roche Pharmaceutical

      Funding:
      Roche Pharmaceutical

      Disclosure:
      N. Yu: Roche employee. I. Reyes-Rivera: Employee: F. Hoffmann-La Roche Ltd., Stock ownership: F. Hoffmann-La Roche Ltd. J. Hipp: Employee: Roche Pharma AG, and Stock ownership: F. Hoffmann La-Roche. M. Zou: Corporate-sponsored research by Roche. S. Hammerschmidt: Advisory board or board of directors: Astra Zeneca, Boehringer-Ingelheim, BMS, Novartis, Pfizer, Roche. All other authors have declared no conflicts of interest.

    • +

      133P - The influence of body composition on TTFields intensity in the lungs (ID 466)

      12:30 - 12:30  |  Author(s): U. Weinberg, N. Urman, H.S. Hershkovich, Z. Bomzon, E.D. Kirson, Y. Palti

      • Abstract

      Background:
      Tumor Treating Fields (TTFields) are low intensity, alternating electric fields in the intermediate frequency range that disrupt mitosis. The therapy is FDA approved for the treatment of glioblastoma, and a pivotal study testing the efficacy of TTFields in non-small cell lung cancer is planned. TTFields are delivered through two pairs of transducer arrays (TL) placed on the patient's skin. Since the efficacy of TTFields increases with intensity, it is important to identify factors that influence field intensity in the lungs. Therefore, it is important to understand how body shape and composition influence the field intensity. Here we present a computer-simulation-based study investigating the effect of body size, shape and composition on the field distribution in the lungs.

      Methods:
      The study was performed using the Sim4Life software package and realistic computational phantoms of a female (ELLA), male (DUKE) and obese male (FATS). Various array layouts were placed on the models, and the distribution of TTFields within their lungs were calculated and compared.

      Results:
      For all models, uniform field distributions within the lungs were obtained when the arrays were axially-aligned with the parenchyma as much as anatomically possible. The layouts that generated the highest average field intensities were those in which one pair of arrays delivered an electric field from the anterolateral to the posterior-contralateral aspect of the patient, with the second pair inducing the field from the antero-contralateral to the posterolateral aspect of the patient. In all models, these layouts led to average field intensities in the lungs of above the therapeutic threshold (>1 V/cm). The highest field intensities developed in DUKE's lungs and the lowest field intensities developed in FATS's lungs. Analysis suggests that field attenuation was caused primarily by layers of fat. Hence, the lower field intensities in the lungs of ELLA and FATS can be largely attributed to the thick layers of fat present in FATS and the fatty tissue in ELLA's breasts.

      Conclusions:
      This study provides insights into how TTFields distribution in the lungs is influenced by body composition. These insights will help to optimize TL placement and design in the future.

      Clinical trial identification:


      Legal entity responsible for the study:
      Novocure

      Funding:
      Novocure

      Disclosure:
      U. Weinberg, N. Urman, H.S. Hershkovich, E.D. Kirson: Employee of Novocure. Z. Bomzon: Paid employee of Novocure Ltd. Y. Palti: The author is a shareholder in Novocure.

    • +

      134P - Applicable CT-guided percutaneous radiofrequency ablation (RFA) in the treatment of unresectable Lung Ground Glass Opacity (GGO) (ID 348)

      12:30 - 12:30  |  Author(s): Y.T. Wei

      • Abstract

      Background:
      Radiofrquency ablation (RFA) is a locoregional therapeutic for variable unresectable malignancies and currently used for lung cancer (either primary or metastatic). It is minimally invasive and accurate for percutaneous RFA under CT guidance. This study was to investigate the safety and efficacy of RFA in the treatment of unresectable lung Ground Glass Opacity (GGO).

      Methods:
      From June 2015 to June 2016 a total of 68 patients (29 males and 39 females with a median age of 46 y, range, 29-78 y) with 83 lesions (GGO) have been enrolled in this study. As approved by institutional review board, all patients were informed and signed the consent forms beforehand. All the preselected lesions were proven by needle biopsy under CT guidance percutaneously prior to the procedure. In total 60 malignancies were ultimately included and CT-guided percutaneous RFA was serially performed with pre-set power of 40-80w and ablation time of 12-25 min. Imaging follow-up served as evaluation method.

      Results:
      All of the procedures were performed to completion. There were 18 patients with pneumothorax postoperatively and of those 6 needed a closed thoracic drainage. 8 patients showed slight bleeding during the procedures. Follow-up CT scanning at 30-, 90-, and 180-day postoperatively have shown that 56 of 60 lesions turned consolidated and with volume diminished. 4 of 60 were without significant dimensional change at 30-day follow-up, 3 of the 4 with accentuation were detected on 90-day CT follow-up and needed a reoperation of RFA.

      Conclusions:
      CT-guided percutaneous RFA is safe and effective. It offers good control and is well-tolerated as a viable option for unresectable GGO.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chinese PLA General Hospital

      Funding:
      Chinese PLA General Hospital

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      135P - Coxsackievirus type B3 is a potent oncolytic virus against KRAS-mutant non-small cell lung cancer (ID 242)

      12:30 - 12:30  |  Author(s): H. Deng, T.D. Sliva, Y. Xue, Y. Mohamud, J. Zhang, W. Lockwood, H. Luo

      • Abstract

      Background:
      Lung cancer is one of the most leading causes of cancer-related death worldwide. Over 85% of lung cancers are non-small cell lung cancer (NSCLC), for which the 5-year survival rate is extremely low (∼15.9%). Most NSCLCs are caused by the accumulation of genomic alterations, among which epidermal growth factor receptor (EGFR) mutation and KRAS mutation are two of the most predominant types. Although patients with EGFR-mutant NSCLCs have manifested a good response to EGFR inhibitors, there is a paucity of effective treatments for the KRAS-mutant NSCLCs and new strategies are urgently needed. Coxsackievirus type B3 (CV-B3) is a non-enveloped, human-pathogenic enterovirus that causes mild flu-like symptoms in adults. Due to its highly lytic nature, CV-B3 has yielded an increased efficacy of viral-mediated oncolysis as compared to other viruses, which makes it as a good candidate for cancer treatment.

      Methods:
      Seven NSCLC cell lines (A549, H2030, H23, H1975, PC-9, H3255 and HCC4006) and three normal lung epithelial cells (HPL1D, HAE and BEAS2B) were selected for this study. Cells were infected with CV-B3 (MOI 0.01) for 48hrs. Cytopathic effects caused by virus infection were observed by light microscope, followed by crystal violet staining. MTS assay were conducted to examine the resistance of normal lung epithelial cells upon CVB3 infection. The supernatants were collected to determine the virus titres by plaque assay. Coxsackievirus and adenovirus receptor (CAR) expression was examined via western blot.

      Results:
      Our studies found that CV-B3 treatment led to a significant reduction of cell survival in KRAS-mutant NSCLCs but not EGFR-mutant NSCLCs nor normal lung epithelial cells. MTS assay results demonstrated CV-B3 infection didn’t lead to a significant enhancement of cell death in normal lung epithelial cells. Furthermore, we showed that virus titres within the supernatants of KRAS-mutant NSCLCs are significantly higher than both EGFR-mutant NSCLCs and normal lung epithelial cells. Finally, we demonstrated that CAR expression levels were significantly increased in KRAS-mutant NSCLCs.

      Conclusions:
      Our study found that CV-B3 is an effective and safe oncolytic virus against KRAS-mutant NSCLCs.

      Clinical trial identification:


      Legal entity responsible for the study:
      Center for Heart and Lung Innovation, St. Paul’s Hospital, Department of Pathology and Laboratory Medicine, University of British Columbia

      Funding:
      British Columbia Lung Association

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      136P - Long-term safety and efficacy of darbepoetin alfa in subjects with stage IV NSCLC receiving multi-cycle chemotherapy (ID 357)

      12:30 - 12:30  |  Author(s): P. Gascón, R. Nagarkar, M. Šmakal, K. Syrigos, C.H. Barrios, J. Cárdenas Sánchez, L. Zhang, D. Henry, D. Tomita, C. De Oliveira Brandao

      • Abstract

      Background:
      Darbepoetin alfa (DA) is an erythropoiesis-stimulating agent (ESA) that has been shown to increase hemoglobin levels and reduce the rate of transfusions in patients with chemotherapy-induced anemia (CIA). Most studies have not shown an association between ESA use and poor outcomes, but some clinical trials have reported increased mortality and/or tumor progression. This trial was therefore designed to address the safety of DA for CIA in patients with non-small cell lung cancer (NSCLC).

      Methods:
      Study 20070782 is a randomized, double-blind, noninferiority trial to compare DA with placebo, and is enrolling patients with NSCLC with CIA. Eligible patients are ≥ 18 years old with Eastern Cooperative Oncology Group (ECOG) status ≤ 1, stage IV NSCLC, no prior adjuvant/neoadjuvant NSCLC therapy, ≥ 2 cycles first-line chemotherapy planned (≥ 6 weeks total), and screening hemoglobin ≤ 11 g/dL. Approximately 3,000 patients from up to 500 global sites will be randomized 2:1 to DA (500 mcg) or placebo every 3 weeks (Q3W) until disease progression or end of chemotherapy. At hemoglobin > 12 g/dL, study drug is withheld until hemoglobin ≤ 12 g/dL. Transfusions are allowed when necessary. Endpoints include overall survival (OS; primary) and progression-free survival (PFS; secondary), and will be analyzed when ∼2,700 deaths have occurred. Additional safety endpoints include tumor response and rate of thromboembolic events. Superiority of DA to placebo in transfusion rates will be tested if noninferiority is achieved for OS and PFS.

      Results:
      As of January 1, 2017, a total of 2,447 patients have enrolled. The independent data monitoring committee has conducted 10 reviews of unblinded data (which included a planned formal interim analysis at 60% of planned total number of 2,700 deaths to test for harm), and has recommended continuation of the trial without changes.

      Conclusions:
      Study 20070782 is the largest clinical trial in NSCLC to date, and will provide comprehensive data on the safety and efficacy of DA in patients with CIA.

      Clinical trial identification:
      NCT00858364

      Legal entity responsible for the study:
      Amgen Inc.

      Funding:
      Amgen Inc.

      Disclosure:
      C.H. Barrios: Pfizer, Novartis, Amgen, AstraZeneca, Boehringer Ingelheim, Roche, Lilly, Sanofi, GSK, Taiho, Mylan, Merrimack, Merck, Abbvie, Astellas, Biomarin, BMS, Daiichi Sankyo, Abraxis, AB Science, Asana, Medivation, Daiichi Sankyo, Exelixis, ImClone, LEO. L. Zhang: Investigator/consultant: AstraZeneca, Pfizer, Lilly, BMS, Roche, Boehringer Ingelheim. D. Tomita, C. De Oliveira Brandao: Employee and shareholder of Amgen Inc. All other authors have declared no conflicts of interest.

    • +

      137P - Cost-effectiveness of afatinib versus erlotinib for the treatment of squamous non-small cell lung cancer in France after a first-line platinum based therapy (ID 176)

      12:30 - 12:30  |  Author(s): M. Pignata, K. Le Lay, L. Luciani, C. McConnachie, J. Gordon, C. Chouaid, S. Roze

      • Abstract

      Background:
      Non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancers and has an extremely poor prognosis. Among the different histologies, squamous NSCLC represents 20 to 30% of them. Afatinib is an irreversible ErbB family blocker approved in Europe for squamous NSCLC after a first-line platinum based therapy. The objective of the present study was to evaluate the cost-effectiveness of afatinib versus erlotinib in this setting in France.

      Methods:
      The study population was taken from the LUX-Lung 8 trial which compared afatinib with erlotinib in patients with squamous NSCLC. The analysis was performed from the perspective of healthcare funders and affected patients in France. A state transition model was developed to evaluate cost-effectiveness based on progression-free survival and overall survival in the trial. Life expectancy, quality-adjusted life expectancy and direct costs were evaluated over a 10-year time horizon. Future costs and clinical benefits were discounted at 4% annually. Deterministic and probabilistic sensitivity analyses were performed.

      Results:
      Model projections indicated that patients-treated with afatinib benefitted from longer life expectancy than those treated with erlotinib (0.94 years versus 0.78 years respectively) translating to an increase of 0.094 quality-adjusted life years (QALYs). The total cost of treatment over a 10-year time horizon was higher for afatinib than erlotinib, EUR 12,364 versus EUR 9,510, leading to an incremental cost-effectiveness ratio of EUR 30,277 per QALY gained for afatinib versus erlotinib. Sensitivity analyses showed that the base case findings were stable under variation in a range of model inputs.

      Conclusions:
      Based on data from the LUX-Lung 8 trial, afatinib was projected to improve clinical outcomes versus erlotinib, with an 89% probability of being cost-effective assuming a willingness to pay of EUR 50,000 per QALY gained, following a first-line platinum based therapy for patients with squamous NSCLC in France.

      Clinical trial identification:


      Legal entity responsible for the study:
      HEVA HEOR

      Funding:
      Boehringer Ingelheim

      Disclosure:
      M. Pignata, C. McConnachie, S. Roze: The study was supported by funding from Boehringer Ingelheim. K. Le Lay: Employee of Boehringer Ingelheim. L. Luciani: Employee of Boehringer Ingelheim. J. Gordon: Employee of Boehringer Ingelheim. C. Chouaid: In the past 5 years, received fees for attending scientific meetings, speaking, organizing research or consulting from AstraZeneca, Boehringer Ingelheim, and Hoffman la Roche.

    • +

      138TiP - An open-label phase 3b/4 safety trial of flat-dose nivolumab plus ipilimumab in patients with advanced non-small cell lung cancer (NSCLC) (ID 164)

      12:30 - 12:30  |  Author(s): L. Paz-Ares, B. Lash, I. Albert, G. Gagnon, C. Chakmakjian, N. Ready, W. Hu, L. Krug, J. Fairchild, R.N. Pillai

      • Abstract

      Background:
      The combination of nivolumab and ipilimumab, immune checkpoint inhibitors with distinct but complementary mechanisms of action, is approved as first-line therapy for metastatic melanoma and has shown encouraging clinical activity in other tumors, including NSCLC. In CheckMate 012, a multi-cohort phase 1 trial in chemotherapy-naïve patients with NSCLC, nivolumab (3 mg/kg) plus ipilimumab (1 mg/kg) yielded objective response rates of up to 47%; discontinuation rates due to treatment-related adverse events were similar to those with nivolumab monotherapy. Data indicate comparable pharmacokinetic, safety, and efficacy profiles for 240 mg flat-dose nivolumab and 3 mg/kg nivolumab. This open-label phase 3b/4 study (ClinicalTrials.gov identifier: NCT02869789) will characterize the safety of flat-dose nivolumab plus ipilimumab in patients with advanced NSCLC. This study will also evaluate this combination in special patient populations who are typically excluded from NSCLC trials.

      Trial design:
      Adult patients with stage IV/recurrent NSCLC and no prior systemic anticancer therapy (cohort A; n = 400), or with stage IIIb/IV NSCLC and recurrence or progression during or after one prior platinum doublet chemotherapy regimen (cohort B; n = 400) will be enrolled. Patients are required to have assessment of programmed death-1 ligand 1 expression, Eastern Cooperative Oncology Group performance status (ECOG PS) 0–1, and no untreated brain metastases, carcinomatous meningitis, autoimmune disease, or active malignancy requiring concurrent intervention. A third cohort (A1; n = ∼200) with no prior systemic therapy will have ECOG PS 2 or one or more of the following: asymptomatic untreated brain metastases, renal or hepatic dysfunction, and/or HIV. All patients will receive flat-dose nivolumab (240 mg every 2 weeks) plus weight-based ipilimumab (1 mg/kg every 6 weeks). Endpoints are shown in the table.rnTable: 138 TiPStudy endpointsrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn rnrn
      PrimarySecondary
      Number and percentage of patients with high-grade treatment-related select and immune-mediated adverse eventsProgression-free survival
      Objective response rate
      Duration of response
      Patient-reported outcomes based on the Functional Assessment of Cancer Therapy-Lung (FACT-L)
      rn

      Clinical trial identification:
      NCT02869789

      Legal entity responsible for the study:
      Bristol-Myers Squibb

      Funding:
      Bristol-Myers Squibb

      Disclosure:
      L. Paz-Ares: Medical advisor for: Lilly, Roche, MSD, BMS, Celgene, Pfizer, Boehringer Ingelheim, Bayer, Clovis, and Astra Zeneca. C. Chakmakjian: Speaker\'s Bureau for: BMS. N. Ready: Honoraria from: BMS, Merck; Consultant for: BMS, Merck, Novartis, Abbvie. W. Hu, L. Krug, J. Fairchild: Bristol-Myers Squibb employee. All other authors have declared no conflicts of interest.

    • +

      139TiP - A non-interventional biomarker study in patients (pts) with non-small cell lung cancer (NSCLC) of adenocarcinoma histology who are treated with nintedanib according to the approved label (LUME-BioNIS) (ID 260)

      12:30 - 12:30  |  Author(s): M. Reck, A.J. Staal-Van den Brekel, A. Mellemgaard, N. Morsli, A. Ellingboe, R. Kaiser, K. Pietzko, T. Kitzing, J. Braunger, K. Kerr

      • Abstract

      Background:
      Nintedanib+docetaxel significantly improved overall survival (OS) of pts with advanced adenocarcinoma NSCLC. There are currently no validated tumour- or serum-derived biomarkers to predict the efficacy of antiangiogenic therapy. The objective of this study is to investigate whether tumour-based gene/protein expression patterns or genomic markers, alone or combined with clinical covariates, could predict treatment effect in pts with adenocarcinoma NSCLC receiving nintedanib (Vargatef[®]).

      Trial design:
      In this non-interventional study at 86 mainly European sites, new biomarker data and clinical characteristics will be collected from ∼300 pts who receive nintedanib as part of routine treatment. Pts must be eligible for nintedanib+docetaxel, i.e. have advanced adenocarcinoma NSCLC after first-line chemotherapy, and will receive nintedanib 200 mg twice daily (Days 2–21 of 21-day cycle) and docetaxel (75 mg/m[2]; Day 1). The primary outcome is OS in relation to exploratory biomarker assessment, including gene expression profile, tumour genomic alterations and protein analysis. Tumour tissue samples obtained prior to first-line therapy are required along with informed consent. Mutation analysis of nintedanib target genes (VEGFR1–3, FGFR1–3, PDGFR α/β) and driver genes (EGFR, KRAS, ALK, BRAF, PIK3CA) will be conducted, as well as evaluation of tumour protein expression (e.g. PD-L1, CD133) and proliferation (Ki-67) markers by immunohistochemistry. One ∼2 mL blood sample (or a buccal swab) will be collected at baseline or after nintedanib initiation alongside routine blood sampling to analyse the potential influence of genetic variants in angiogenesis-related genes (e.g. single nucleotide polymorphisms in VEGFR1). Pts will be followed up every 6 months. The primary outcome will be analysed after 250 deaths. Gene expression patterns and tumour genomics in relation to efficacy will be analysed using univariate and multivariate regression models. Adverse events will be assessed. All analyses will be exploratory and considered hypothesis-generating. The study is ongoing (NCT02671422).

      Clinical trial identification:
      NCT02671422

      Legal entity responsible for the study:
      Boehringer Ingelheim Pharma GmbH & Co. KG

      Funding:
      Boehringer Ingelheim Pharma GmbH & Co. KG

      Disclosure:
      M. Reck: Author reports personal fees from Boehringer-Ingelheim, Hoffmann-La Roche, Lilly, MSD, BMS, AstraZeneca, Celgene, Merck and Pfizer. K. Kerr: Personal fees from Boehringer Ingelheim, during the conduct of the study. All other authors have declared no conflicts of interest.

    • +

      140TiP - CheckMate 384: A phase 3b/4 dose-frequency optimization trial of nivolumab in advanced or metastatic non-small cell lung cancer (ID 261)

      12:30 - 12:30  |  Author(s): N. Reinmuth, R. Harris, P. Mitchell, E.B. Garon, J. Zhu, I. Chang, G. Selvaggi, E. Pichon

      • Abstract

      Background:
      Nivolumab, an anti-programmed death-1 antibody, is approved for the treatment of various cancers. Based on efficacy and safety findings across multiple tumor types, the approved dose of nivolumab was, until recently, 3 mg/kg every 2 weeks (Q2W). In September 2016, the approved nivolumab dose in non-small cell lung cancer (NSCLC), melanoma, and renal cell carcinoma was modified in the United States to a flat dose of 240 mg Q2W. Reducing the frequency of nivolumab administration may increase convenience and compliance while maintaining efficacy and safety in patients receiving long-term nivolumab therapy. CheckMate 384, a randomized, open-label phase 3b/4 trial (NCT02713867) was designed to evaluate less frequent nivolumab dosing (480 mg every 4 weeks [Q4W] vs 240 mg Q2W) in patients with advanced/metastatic NSCLC following up to 12 months of prior treatment with nivolumab 3 mg/kg or 240 mg Q2W.

      Trial design:
      Eligible patients are adults with advanced/metastatic squamous or non-squamous NSCLC and Eastern Cooperative Oncology Group performance status 0–2 who received prior intravenous nivolumab 3 mg/kg or 240 mg Q2W for up to 12 months and achieved a complete or partial response or stable disease confirmed on 2 consecutive assessments. Patients with untreated, symptomatic central nervous system metastases are not eligible. Patients are randomized 1:1 to receive intravenous nivolumab in 1 of 2 flat-dose schedules: 240 mg Q2W or 480 mg Q4W. Randomization is stratified by histology and response to pre-study nivolumab treatment (complete/partial response vs stable disease). Endpoints are shown in the table. The primary objective is to compare 6-month and 1-year progression-free survival (PFS) rates between patients who received nivolumab 480 mg Q4W and those who received 240 mg Q2W. Planned enrollment is 620 patients.rnTable: 140TiPStudy endpointsrn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn rnrn
      PrimarySecondary
      PFS rates at 6 months and 1 year after randomization (co-primary)PFS rate at 1 year after randomization by tumor histology and by response to pre-study nivolumab before randomization
      PFS rate at 2 years after randomization
      Overall survival rate
      Safety and tolerability, as assessed by incidence and severity of adverse events
      rn

      Clinical trial identification:
      NCT02713867

      Legal entity responsible for the study:
      Bristol-Myers Squibb

      Funding:
      Bristol-Myers Squibb

      Disclosure:
      N. Reinmuth: Personal fees (speakers and consulting honoraria): Bristol-Myers Squibb, Hoffmann-La Roche, Lilly, Novartis, Boehringer-Ingelheim, AstraZeneca, Amgen, Pfizer, MSD. R. Harris: Consultant (personal fees): Bristol-Myers Squibb. P. Mitchell: Advisory board member: AstraZeneca, Roche, Boehringer-Ingelheim, BMS, MSD, Celgene; Honoraria: Roche; Travel grants: Roche, BMS. E.B. Garon: Research Funding (received by my institution): AstraZeneca, BMS, Merck, Genentech, Eli Lilly, Pfizer, Novartis, Boehringer Ingelheim, Mirati. J. Zhu, I-F. Chang: BMS Employment and BMS Stock Ownership. S. Selvaggi: Bristol-Myers Squibb employee and shareholder and has received travel, accommodation, and expense assistance from Bristol-Myers Squibb. All other authors have declared no conflicts of interest.

    • +

      141TiP - ASTRIS: A multicenter, real world treatment study of osimertinib in EGFR T790M positive non-small cell lung cancer (NSCLC) (ID 278)

      12:30 - 12:30  |  Author(s): P.K. Cheema, Y. Chen, F. de Marinis, H.C. Freitas, S. Kim, A. Milner, M. Provencio, J. Rigas, Y. Wu

      • Abstract

      Background:
      EGFR tyrosine kinase inhibitors (TKIs) are the first-line treatment for patients with EGFR mutation positive advanced NSCLC. Resistance develops due to a secondary EGFR T790M mutation in approximately 60% of cases. Osimertinib is an oral, irreversible, central-nervous system (CNS) active, EGFR-TKI selective for both EGFR-TKI sensitising and T790M resistance mutations. In a Phase III trial versus platinum-based doublet chemotherapy in patients with T790M positive NSCLC (AURA3), osimertinib provided a significantly longer progression-free survival (PFS; median 10.1 mths vs. 4.4 mths; hazard ratio 0.30; 95% CI 0.23, 0.41; p < 0.001) and higher response rates (71% vs 31%; odds ratio 5.39; p < 0.001) (Mok et al, New Engl J Med 2016). Currently, there is limited evidence of the efficacy of osimertinib outside of clinical trials.

      Trial design:
      ASTRIS is a Phase III open-label, single-arm, multi-national, real world treatment study assessing the efficacy and safety of osimertinib in patients with advanced T790M mutation positive NSCLC, who have previously received an EGFR-TKI. Approximately 3500 patients will be enrolled across Asia, Europe and North and South America on a rolling basis. Country level enrolment is to stop within 6 months of market licence approval or national reimbursement. Key inclusion criteria are adults with locally advanced or metastatic NSCLC having a confirmed T790M mutation who received a prior EGFR-TKI and have World Health Organization performance status 0 − 2. T790M status must be confirmed by an appropriately validated test. Patients with asymptomatic CNS metastases, not requiring an increasing corticosteroid dose within 2 weeks prior to osimertinib administration, are allowed. Osimertinib 80 mg will be administered orally once daily for as long as the patient continues to receive clinical benefit, as judged by the investigator. The primary efficacy outcome is overall survival: secondary outcomes include investigator assessed response rate, PFS, and time to treatment discontinuation. Baseline demographics and disease characteristics, T790M mutation testing results and safety will also be reported.

      Clinical trial identification:
      NCT02474355 (27 May 2015)

      Legal entity responsible for the study:
      AstraZeneca

      Funding:
      AstraZeneca

      Disclosure:
      P.K. Cheema: Advisory board: Astrazeneca, Bristol Myers Squibb, Novartis, Boehringer Ingelheim, Pfizer, Merck Research sponsors: Astrazeneca, Hoffmann La Roche, Boehringer Ingelheim. Y-M. Chen: Advisory board of AstraZeneca, Boehringer Ingelheim, Roche and Merck Sharp & Dohme. H.C. Freitas: Member of ASTRIS steering committee, a research program supported by Astra Zeneca. A. Milner: Employee of AstraZeneca, who sponsored the study. M. Provencio: Honoraria for consultancy work from MSD, Bristol-Myers Squibb, Roche and Astrazeneca. J. Rigas: Consultant, through Kelly Service, for AstraZeneca in Global Medical Affairs on osimertinib (TAGRISSO). Y-L. Wu: Speaker fees from AstraZeneca, Roche, Eli Lilly. All other authors have declared no conflicts of interest.

    • +

      142TiP - A prospective multi-center study to investigate the EGFR-TKI resistance profile, treatment algorithm and clinical outcome in Chinese patients with advanced EGFRm+ NSCLC who have received prior first generation EGFR TKI (PRECENT study, CCTC-1601, NCT02988141) (ID 280)

      12:30 - 12:30  |  Author(s): W. Liang, L. Wu, L. Wang, Y. Huang, R. Xu, L. Zhang, J. He

      • Abstract

      Background:
      The resistance mechanism is impactful to make treatment strategy after prior first generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) failure and the profile has well established in western population; while without solid data among Asian population. In addition, methods used in determining previously reported resistance profiles are flawed in failing to detect concomitant or heterogeneous mechanisms. High throughput next-generation sequencing (NGS), which allows parallel multiplex genotyping with tissue or plasma sample, can help in addressing these issues. Furthermore, little was known about the treatment algorithms and clinical outcomes of different mechanisms in China. Thus, the study aims to investigate the resistance mechanism, treatment strategy and clinical outcome for those patients with prior EGFR-TKI in China.

      Trial design:
      We are going to perform a prospective, multi-center study to obtain: a) the biomarker profile of EGFR-TKI acquired resistance detected based on paired tissue and blood respectively; b) concordance of T790M detection by NGS in blood sample with that in tissue sample as reference; c) the clinical outcomes (ORR, DCR, PFS of subsequent treatment and OS) of patients with different resistance mechanisms by NGS in tissue samples and blood samples. All the paired re-biopsy tissue and blood samples will be collected and subjected to NGS panel testing by central lab. Treatment strategy will be summarized and described. The assessment will be performed every 6 weeks until objective disease progression as defined by RECIST1.1. Survival follow-up will be conducted every 6 weeks until death, lost to follow-up, withdrawal of consent or the DCO (data cut off). A sample size of 100 patients will provide 80% power to detect at least 1 case of resistance mechanism with a proportion of 1.5% and a precision of 9% for an assumed concordance 70% of T790M mutation between tissue and blood samples. First subject will be enrolled in Feb 2017.

      Clinical trial identification:
      NCT02988141

      Legal entity responsible for the study:
      Guangdong Association of Thoracic Disease (China)

      Funding:
      AstraZeneca

      Disclosure:
      L. Zhang: Member on an advisory board and receives lecture fees from AstraZeneca. All other authors have declared no conflicts of interest.

    • +

      143TiP - IMpower110: Phase III trial of 1L atezolizumab in PD-L1–selected chemotherapy-naive NSCLC (ID 364)

      12:30 - 12:30  |  Author(s): J. Jassem, F. de Marinis, D.R. Spigel, S. Lam, S. Mocci, A. Sandler, A. Lopez-Chavez, Y. Deng, G. Giaccone, R.S. Herbst

      • Abstract

      Background:
      Despite poor survival and toxicities, chemotherapy is the standard of care and remains the main first-line option for patients (pts) with advanced NSCLC non-squamous (non-sq) and squamous (sq) histology without genetic driver alterations. Immunotherapies targeting PD-L1/PD-1 are available for 2L+ NSCLC but remain to be fully studied in the 1L setting. Atezolizumab (atezo), an anti–PD-L1 mAb, prevents PD-L1 interacting with PD-1 and B7.1, restoring tumor-specific T-cell immunity. Significant and clinically relevant survival benefit has been shown with atezo in previously treated NSCLC, regardless of PD-L1 expression on tumor cells (TC) and/or tumor-infiltrating immune cells (IC). IMpower110 (NCT02409342), a global Phase III randomized, multicenter, open-label trial, will evaluate efficacy and safety of 1L atezo vs cisplatin (cis)/carboplatin (carbo) + pemetrexed (pem) or gemcitabine (gem) in PD-L1–selected chemotherapy-naive pts with advanced non-sq or sq NSCLC, respectively.

      Trial design:
      Inclusion criteria include stage IV non-sq or sq NSCLC, measurable disease (RECIST v1.1), ECOG PS 0-1, no prior chemotherapy for advanced NSCLC and centrally assessed PD-L1 expression of ≥ 1% on TC or IC (TC1/2/3 or IC1/2/3 with VENTANA SP142 IHC assay; expected prevalence, ≈ 65%). Exclusion criteria include active or untreated CNS metastases, prior immune checkpoint blockade therapy or autoimmune disease. Pts will be randomized 1:1 to receive atezo or cis/carbo + pem (non-sq)/gem (sq) (4 or 6 21-day cycles are allowed). Pts receiving atezo may continue until loss of clinical benefit, while pts in the comparator arm can receive pem maintenance (non-sq) or best supportive care (sq) until disease progression. Stratification factors are sex, ECOG PS, histology (non-sq vs sq) and centrally assessed PD-L1 expression by IHC. Co-primary endpoints are PFS and OS. Key secondary endpoints are ORR, DOR, IRF-assessed PFS (RECIST v1.1) and TTD in pt-reported lung cancer symptoms. Safety and PK will also be evaluated. Tumor biopsies at progression will be assessed for immunologic biomarkers associated with responses to atezo and to differentiate unusual responses from radiographic progression. Approximately 570 pts will be enrolled.

      Clinical trial identification:
      NCT02409342

      Legal entity responsible for the study:
      F. Hoffmann-La Roche Ltd/Genentech Inc., a member of the Roche Group

      Funding:
      F. Hoffmann-La Roche Ltd/Genentech Inc., a member of the Roche Group

      Disclosure:
      J. Jassem: Speaker: AstraZeneca, Roche, Pfizer; Advisory roles: AstraZeneca, Boehringer, BMS, Celgene, G1 Therapeutics, Merck, Pfizer, Pierre Fabre, Roche; Travel support: Roche, Boehringer. F. de Marinis: Consultation fees received from Roche/BMS/Boehringer/Novartis/Pfizer/MSD/Astrazeneca. D.R. Spigel: Consulting/Advisory Role: Genentech (uncompensated); Travel, accommodation, expenses: Genentech. S. Lam, S. Mocci, A. Sandler, A. Lopez-Chavez, Y. Deng: Employee, stock: Roche/Genentech. G. Giaccone: Consulting or Advisory Role: Clovis, Boehringer-Ingelheim; Celgene; Research grants: Karyopharm, Astra-Zeneca; Eli-Lilly. R.S. Herbst: Consultant and research support from Genentech

    • +

      144TiP - CheckMate 227: A randomized, open-label phase 3 trial of nivolumab, nivolumab plus ipilimumab, or nivolumab plus chemotherapy versus chemotherapy in chemotherapy-naïve patients with advanced non-small cell lung cancer (NSCLC) (ID 394)

      12:30 - 12:30  |  Author(s): L. Paz-Ares, J. Brahmer, M.D. Hellmann, M. Reck, K. O'Byrne, H. Borghaei, W.J. Geese, H. Lu, F.E. Nathan, S. Ramalingam

      • Abstract

      Background:
      Platinum-based chemotherapy is standard-of-care first-line therapy for most patients with advanced NSCLC, but the clinical benefit is modest. Although first-line nivolumab, an immune checkpoint inhibitor antibody, did not improve progression-free survival or overall survival (OS) versus chemotherapy in patients with advanced NSCLC and ≥5% programmed death-1 ligand 1 (PD-L1) expression, OS compared favorably with historical controls of first-line platinum-based chemotherapy. Combining nivolumab with chemotherapy in this setting may increase the durability of tumor responses and broaden the population of patients to derive benefit. In a multi-cohort phase 1 study (CheckMate 012) in chemotherapy-naïve patients with advanced NSCLC, nivolumab plus chemotherapy had promising clinical activity, regardless of tumor PD-L1 expression, and a manageable safety profile. CheckMate 227 is a 2-part, randomized, open-label phase 3 trial (NCT02477826), evaluating first-line nivolumab, nivolumab plus ipilimumab, or nivolumab plus chemotherapy versus chemotherapy in patients with advanced NSCLC.

      Trial design:
      Part 1 of CheckMate 227, which has completed accrual, enrolled adult patients with stage IV/recurrent NSCLC, no prior systemic anticancer therapy, and assessment of PD-L1 expression at screening. Patients with ≥1% PD-L1 expression were randomized 1:1:1 to nivolumab, nivolumab plus ipilimumab, or chemotherapy arms; those with <1% PD-L1 expression were randomized 1:1:1 to nivolumab plus ipilimumab, nivolumab plus chemotherapy, or chemotherapy arms. In part 2, ∼480 previously untreated patients with advanced NSCLC, regardless of PD-L1 expression level, will be randomized 1:1 to receive histology-based platinum doublet chemotherapy alone or in combination with nivolumab. Part 2 of CheckMate 227, which is the focus of this presentation, allows for the evaluation of first-line nivolumab plus chemotherapy in a broad group of patients with advanced NSCLC across the PD-L1–expressing continuum.

      Clinical trial identification:
      NCT02477826

      Legal entity responsible for the study:
      Bristol-Myers Squibb

      Funding:
      Bristol-Myers Squibb

      Disclosure:
      L. Paz-Ares: Served as a medical advisor for the following companies: Lilly, Roche, MSD, BMS, Celgene, Pfizer, Boehringer-Ingelheim, Bayer, Clovis, and AstraZeneca. J. Brahmer: Received research grants and served as an uncompensated advisory board member for Bristol-Myers Squibb. M.D. Hellmann: Received grants from Genentech and Bristol-Myers Squibb. Received personal fees from Genentech, Bristol-Myers Squibb, Merck, AstraZeneca, Novartis, and Janssen. M. Reck: Received consultant fees and served on speaker\'s bureau for the following companies: Roche, Lilly, Bristol-Myers Squibb, MSD, AstraZeneca, Pfizer, Boehringer-Ingelheim, and Celgene. K. O\'Byrne: Received honoraria, speaker bureau and/or travel and registration support for national and international meetings from BMS, Boehringer-Ingelheim, Astrazeneca, Lilly Oncology, Novartis, MSD, Roche-Genentech and Pfizer. H. Borghaei: The institution has a clinical trial agreement w/BMS. Consultant/advisory board member for: BMS, Lilly, Genentech, Celgene, EMD-Serono, Merck, Pfizer, Trovagene, Millenium, & Boehringer-Ingelheim. Received grants from: Millenium, Merck, & Celgene. W.J. Geese, H. Lu: BMS Employee and stock holder. F.E. Nathan: BMS employee. S. Ramalingam: Served on ad hoc scientific advisory board meetings the following companies: Astra Zeneca, BMS, Boehringer Ingelheim, Celgene, Ariad, Amgen, Lilly, Merck, Genentech. Also received honoraria from BMS.

    • +

      145TiP - TTFields combined with PD-1 inhibitors or docetaxel for 2nd line treatment of non-small cell lung cancer (NSCLC): Phase 3 LUNAR study (ID 408)

      12:30 - 12:30  |  Author(s): U. Weinberg, O. Farber, M. Giladi, Z. Bomzon, E.D. Kirson

      • Abstract

      Background:
      Tumor Treating Fields (TTFields) are a non-invasive, anti-mitotic treatment modality. TTFields disrupt the formation of the mitotic spindle, and dislocation of intracellular constituents. TTFields significantly extend the survival of newly diagnosed glioblastoma patients when combined with temozolomide. Efficacy of TTFields in NSCLC has been shown preclinically and in a phase I/II pilot study with pemetrexed, where overall survival (OS) improved by > 5 months vs historical controls.

      Trial design:
      We hypothesize that adding TTFields to 2nd line therapies in advanced NSCLC will increase OS. Patients (N = 512) with squamous or non-squamous NSCLC are enrolled in this Phase 3 study LUNAR [NCT02973789]. Patients are stratified by 2[nd] line therapy (PD-1 inhibitor or docetaxel), histology (squamous vs. non-squamous) and geographical region. Key inclusion criteria are 1st disease progression (RECIST 1.1), ECOG 0-1, no prior surgery or radiation therapy, no electronic medical devices in the upper torso, and absence of brain metastasis. Docetaxel or PD-1 inhibitors (either nivolumab or pembrolizumab) are given at standard doses. TTFields are applied to the upper torso for at least 18 hours/day, allowing patients to maintain daily activities. TTFields are continued until progression in the thorax and/or liver according to the immune-related response criteria (irRC). Follow up is performed once q6 weeks, including CT scans of the chest and abdomen. On progression in the upper torso, patients are followed monthly for survival. The primary endpoint is superiority in OS between patients treated with TTFields in combination with either docetaxel or PD-1 inhibitors, compared to docetaxel or PD-1 inhibitors alone. A co-primary endpoint compares the OS in patients treated with TTFields and docetaxel to those treated with PD-1 inhibitors alone in a non-inferiority analysis. Secondary endpoints include progression-free survival, radiological response rate based on the irRC, quality of life based on the EORTC QLQ C30 questionnaire and severity & frequency of adverse events. The sample size is powered to detect a HR of 0.75 in TTFields-treated patients versus control group.

      Clinical trial identification:
      NCT02973789

      Legal entity responsible for the study:
      Novocure Ltd

      Funding:
      Novocure Ltd

      Disclosure:
      U. Weinberg, O. Farber, M. Giladi, Z. Bomzon: Full time employee of Novocure. E.D. Kirson: Full time employee of Novocure Ltd

    • +

      146TiP - EXalt3: A phase III study of ensartinib (X-396) in anaplastic lymphoma kinase (ALK)-positive non-small cell lung cancer (NSCLC) (ID 473)

      12:30 - 12:30  |  Author(s): L. Horn, Y. Wu, M. Reck, C. Liang, F. Tan, K. Harrow, V. Oertel, G. Dukart, T.S.K. Mok

      • Abstract

      Background:
      Ensartinib is a novel, potent anaplastic lymphoma kinase (ALK) small molecule tyrosine kinase inhibitor (TKI) with additional activity against MET, ABL, Axl, EPHA2, LTK, ROS1 and SLK. Ensartinib has demonstrated activity in ALK treatment naïve and previously treated patients and has a generally well tolerated safety profile.

      Trial design:
      eXalt3 (NCT02767804) is a global, randomized, open-label phase III study comparing the efficacy and safety of ensartinib to crizotinib in ALK- positive TKI naïve non-small cell lung cancer (NSCLC) patients. It is being conducted in > 100 sites in North America, South America, Europe, and the Asia/Pacific region. Enrollment began in 2016. The primary efficacy endpoint is progression free survival (PFS) assessed by independent radiology review. Secondary efficacy endpoints include overall survival, response rates (overall and central nervous system [CNS]), PFS by investigator assessment, time to response, duration of response, and time to CNS progression. Approximately 270 patients with ALK+ NSCLC who have received no prior ALK TKI and up to one prior chemotherapy regimen will be randomized 1:1 to ensartinib 225 mg QD, or crizotinib 250 mg BID, with stratification based on prior chemotherapy, ECOG performance status (PS), CNS metastases and geographic region. Eligibility includes patients ≥ 18 years of age, stage IIIB or IV ALK+ NSCLC. Patients are required to have measurable disease per RECIST 1.1, adequate organ function, and an ECOG PS of ≤ 2. Adequate tumor tissue (archival or fresh biopsy) must be available for central testing. The study has >80% power to detect a superior effect of ensartinib over crizotinib in PFS at a 2-sided alpha level of 0.05.

      Clinical trial identification:
      NCT02767804

      Legal entity responsible for the study:
      Xcovery Holding Company

      Funding:
      Xcovery Holding Company

      Disclosure:
      L. Horn: Consulting for Xcovery Holding Company, BMS, BI, Abbvie, Genentech, Merck. Y-L. Wu: Speaker fees from AstraZeneca, Roche, Eli Lilly. M. Reck: Honoraria for lectures and consultancy: Hoffmann-La Roche, Lilly, BMS, MSD, AstraZeneca, Merck, Celgene, Boehringer-Ingelheim, Pfizer, Novartis. C. Liang, K. Harrow, V. Oertel, G. Dukart: Full-Time Employee- Xcovery Holding Company. F. Tan: Manager- Xcovery Holding Company Chief Medical Officer- Betta Pharmaceuticals. T.S.K. Mok: Grant/Research Support from AstraZeneca, BI, Pfizer, Novartis, SFJ, Roche, MSD, Clovis Oncology, BMS, Eisai, Taiho; Speaker’s fees with: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, MSD, Novartis, BMS, Taiho; Major Stock Shareholder in: Sanomics Ltd.; Advisory Board for: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, Clovis Oncology, Merck Serono, MSD, Novartis, SFJ Pharmaceutical, ACEA Biosciences, Inc., Vertex Pharmaceuticals, BMS, geneDecode Co., Ltd., OncoGenex Technologies Inc., Celgene, Ignyta, Inc.; Board of Directors: IASLC, Chinese Lung Cancer Research Foundation Ltd., Chinese Society of Clinical Oncology (CSCO), Hong Kong Cancer Therapy Society (HKCTS).

    • +

      148P - Metastatic site location may influence the diagnostic accuracy of plasma EGFR-mutation testing in NSCLC: A pooled analysis (ID 406)

      12:30 - 12:30  |  Author(s): F. Passiglia, A. Listì, N. Barraco, A. Galvano, D. Fanale, L. Incorvaia, V. Bazan, C. Rolfo, A. Russo

      • Abstract

      Background:
      Recent studies evaluated the potential role of EGFR mutation testing by using circulating tumor DNA (ctDNA) isolated from plasma of NSCLC patients, overall showing a lower sensitivity compared with the standard tissue genotyping. However, it’s less clear if the presence of extrathoracic (M1b) disease may enhance the ability to identify EGFR mutations in plasma. This pooled analysis aims to evaluate the association between metastatic site location and sensitivity of ctDNA analysis.

      Methods:
      Data from all published studies, that evaluated the sensitivity of plasma-based EGFR-mutation testing, stratified by metastatic site location (extrathoracic (M1b) vs intrathoracic (M1a)) were collected by searching in PubMed, Cochrane Library, American Society of Clinical Oncology, and World Conference of Lung Cancer, meeting proceedings. Pooled Odds ratio (OR) and 95% confidence intervals (95% CIs) were calculated for the ctDNA analysis sensitivity, according to the metastatic site location.

      Results:
      A total of seven studies, with 1233 patients, were eligible. Pooled analysis showed that the sensitivity of EGFR-mutation testing by ctDNA was significantly higher in patients with extrathoracic disease (M1b) compared to patients with intrathoracic (M1a) disease (OR: 4.29; 95% CIs: 2.20 – 8.38).

      Conclusions:
      These data suggest that the location of metastatic sites significantly influences the diagnostic accuracy of ctDNA analysis. Particularly the ability to identify EGFR activating mutations in plasma of NSCLC patients is significantly higher in the presence of M1b vs M1a disease. These observations could influence the clinical management of EGFR-mutated patients.

      Clinical trial identification:
      N.A

      Legal entity responsible for the study:
      Palermo University Hospital

      Funding:
      Palermo University Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      149P - PD-L1 expression patterns in the metastatic tumors to the lung: A comparative study with the primary non-small cell lung cancer (ID 251)

      12:30 - 12:30  |  Author(s): Z. Gatalica, J. Senarathne, S. Vranic

      • Abstract

      Background:
      Immune check point inhibitors (anti-PD-1/PD-L1) therapy has revolutionized cancer treatment of several, advanced and chemotherapy resistant malignancies. PD-L1 expression on tumor (TC) and/or inflammatory cells (IC) has been associated with a more favorable therapy response. We compared PD-L1 distribution in a large cohort of advanced tumors metastatic to the lungs and compared it with the primary lung non-small cell carcinomas (NSCLC).

      Methods:
      The study groups included 176 metastatic cancers and 81 NSCLC. Expression of PD-L1 was assessed using immunohistochemistry (SP142, Ventana). PD-L1 positivity was defined as 2+ intensity at ≥ 5% in TC or IC cells. All cases were further stratified into 4 categories based on the expression presence or absence of PD-L1 expression on tumor or IC cells. PD-L1 expression was correlated with total mutational load (TML) measured in tumors using NGS.

      Results:
      Overall TC PD-L1 positivity was significantly higher in NSCLC compared with metastatic tumors (28% vs. 14%, p = 0.009) although some metastatic cancers (e.g. triple-negative breast and head/neck carcinomas, melanoma) exhibited higher TC PD-L1 expression. In contrast, overall IC PD-L1 expression was predominantly observed in metastatic tumors (28% vs. 0%, p < 0.001). The IC PD-L1 expression ranged from 0% for metastatic renal cell carcinomas to 36-38% in the metastatic breast and colon carcinomas and melanoma. Consequently, the stratification based on PD-L1 distribution (TC vs. IC), resulted in significantly different patterns between the primary and metastatic tumors (p < 0.001, Table). Mean TML (±SD) for NSCLC (10±5.6) differed significantly from metastatic carcinomas from other sites (6.6±2.7) (p = 0.013).

      Conclusions:
      Our study indicate that a substantial proportion of metastatic tumors to the lung exhibit PD-L1 expression on either tumor or inflammatory (immune) cells and are potentially amenable for the treatment with immune check point inhibitors.rnTable: 149Prn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      HistotypesTME categories (PD-L1 expression)Total
      TC+/IC+TC-/IC-TC+/IC-TC-/IC+
      NSCLC05823081
      Metastatic tumors81111641176
      Total81693941257
      rn

      Clinical trial identification:


      Legal entity responsible for the study:
      None

      Funding:
      Caris Life Sciences

      Disclosure:
      Z. Gatalica: Employee of Caris Life Sciences. J. Senarathne: Jude Senarathne is an employee of Caris Life Sciences. S. Vranic: Received honoraria from Caris Life Sciences.

    • +

      150P - Immunohistochemical characteristics of brain metastases and corresponding primary lung cancer (ID 360)

      12:30 - 12:30  |  Author(s): D. Marinova, Y. Slavova, S. Nachev, D. Dimitrova, E. Mekov, M. Mihailov, D. Kostadinov, V. Youroukova

      • Abstract

      Background:
      About 40% of patients with lung cancer (LC) develop brain metastases (BM). The prognosis is poor. The study of BM is important for better understanding of the biology of LC.

      Methods:
      Surgically resected BMs and corresponding primary LCs from 30 patients (men n = 25, 83%; age 55±9) were studied: adenocarcinoma (AC)-21, squamous cell carcinoma (SCC)-5, small cell lung carcinoma (SCLC)-4. The histological subtype and immunohistochemical expression of TTF-1, p63, cytokeratin 7, synaptophysin, Ki-67 (proliferative activity) and CD31 (number intratumoral microvessels-NIM) were evaluated.

      Results:
      The histology of LC compared with BM is different in half of the AC and without difference in SCC and SCLC. ACs are mainly with acinar (53% cases) and solid (37%) components. Metastatic ACs are more often with papillary (47%) component. In 47% of AC BM has different histological structure than LC. The acinar AC are predominantly papillar in 70% of BM showing that the papillary component metastasize most frequently. TTF-1 is expressed in greater number of lung AC (n = 20, 95%), but with lower mean levels of expression, while the corresponding BM express the marker less frequently (n = 16, 76%), but when it is presented it has higher mean values of expression (45.44 vs73.88, p = 0.011). P63 is expressed in high percentage in all SCC (n = 5,100%); there is no difference in expression between LC and BM (80.6 vs 81.6, p = 0.68). Cytokeratin 7 is expressed in all AC equally regardless of the site - primary or metastatic. Ki-67 proliferative index (PI) is higher in SCLC than in lung AD (p = 0.008), in SCLC BM than in AD BM (p < 0.001), in SCLC BM than in SCC BM (p = 0.008). It was found that the Ki-67 PI BM is higher than that of AC LC (30 vs. 17, p = 0.003), in SCC (35 vs.27, p = 0.048) but without difference in SCLC (p = 0.141). CD31 establish vascular invasion in LC – NIM is higher in AC than in SCLC (55vs.31, p = 0.003), in SCC than in SCLC (61vs.31, p = 0.009), no difference between AC and SCC (66 vs.61, p = 0.467). There were no significant differences between LC and BM.

      Conclusions:
      There are differences between primary LC and corresponding BM – in histology structure, in immunohistochemical expression, and in proliferative activity.

      Clinical trial identification:


      Legal entity responsible for the study:
      Medical University, Sofia.

      Funding:
      Supported by Grant 360/2015-Contract Nr.76/2015 funded by Medical University, Sofia.

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      151P - Predictors of survival in patients with bone metastasis of lung cancer (ID 354)

      12:30 - 12:30  |  Author(s): S. Jagadeesan

      • Abstract

      Background:
      In non-small cell lung cancer patients (NSCLC), median survival from the time patients develop bone metastasis is classically described being inferior to 6 months. We investigated the subcategory of patients having skeletal-related-event revealing NSCLC. The purpose of this study was to assess the impact of bone involvement on overall survival and to determine biological and tumoral prognosis factors on OS.

      Methods:
      We assessed the survival rates after bone metastasis and prognostic factors in 28 patients with bone metastases from lung cancer. We first assessed the survival rates and explored various prognostic factors of 28 patients with bone metastasis from lung cancer. We then preliminarily ascertained in a small group of patients whether treatment with an EGFR inhibitor had the potential to influence survival.

      Results:
      The cumulative survival rates after bone metastasis from lung cancer were 59.9% at 6 months, 31.6% at 1 year, and 11.3% at 2 years. The mean survival was 9.7 months (median, 7.2 months; range, 0.1–74.5 months). A favorable prognosis was more likely in women and patients with adenocarcinoma, solitary bone metastasis, no metastases to the appendicular bone, no pathologic fractures, performance status 1 or less, use of systemic chemotherapy, and use of an epithelial growth factor receptor inhibitor.

      Conclusions:
      Bone metastases are a common problem in advanced lung cancer. While the benefits of bone-targeted therapies have been demonstrated, their use is limited in non-trial populations. If better predictive markers of individual risk were available this might increase the appropriate use of bone-targeted agents.

      Clinical trial identification:


      Legal entity responsible for the study:
      S. Jagadeesan

      Funding:
      N/A

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      152P - Relationship between overall survival and preoperative parameters in patients with colorectal cancer and synchronous liver and lung metastases (ID 451)

      12:30 - 12:30  |  Author(s): F. Lumachi, S.M. Basso, U. Zuccon, F. Maffeis, A. Del Conte, P. Ubiali

      • Abstract

      Background:
      The overall survival (OS) of patients with synchronous colorectal cancer (CRC) and pulmonary metastases (PMs) is significantly shorter, but the optimal surgical strategy, including simultaneous colorectal and liver resection vs. delayed liver metastasectomy, has not been defined. The aim of this retrospective study was to evaluate the relationship between OS and preoperative parameters, including age, number and median size of metastases, in patients with PMs from CRC and synchronous or metachronous liver metastases (LMs).

      Methods:
      The medical charts of a group of 20 patients (9 man, 11 women, median age 60 years, range 31-75) who underwent curative surgery for CRC and developed both PMs and synchronous or metachronous LMs were reviewed. Eleven patients aged <65 years, whilst 9 patients aged 65 years or older. Written informed consent was obtained from all the participants. The Pearson correlation coefficient (R) and the linear regression equation calculation was obtained. Because the data were not normally distributed, the Mann-Whitney U-test was used to evaluate statistical significance of correlations.

      Results:
      The median OS was 12.0 months (range 1-58 months) and the overall disease-free interval (DFI) was 2.7±1.9 months. The OS showed a difference in favor of the older (≥65 years) patients (43.3±21.3 vs. 20.1±23.3, p = 0.036). As expected, a strong direct linear relationship between OS and DFI (R = 0.8469, p < 0.000001) was found. There was no significant correlation between OS and both median size (R=−0.3976, p = 0.8256, regression line equation: mm = 21.820349609352-0.20184496105481OS) and number of metastases (R=−0.2975, p = 0.2025, regression line equation: Number=3.294978403666-0.039208413596717OS). Surprisingly, a direct strong relationship between OS and the age of the patients (R = 0.6098, p = 0.0043, regression line equation: age=52.147836178944 + 0.46213761830125OS) was found.

      Conclusions:
      Our results suggest that patients with both PMs and LMs from CRC aged ≥65 years should undergo metastasectomy (in lack of contraindication to surgery) regardless of other parameters, including number and size of metastases, because OS directly correlates with age.

      Clinical trial identification:


      Legal entity responsible for the study:
      Università degli Studi di Padova

      Funding:
      Università degli Studi di Padova

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      153P - Predictive markers of survival in patients with pulmonary metastases and malignant pleural effusion (ID 404)

      12:30 - 12:30  |  Author(s): F. Lumachi, S.M. Basso, R. Tozzoli, A. Del Conte, U. Zuccon, P. Ubiali

      • Abstract

      Background:
      Lung metastases (LMs) can be identified in up to 30-50% of all cancer patients, and represent the result of metastatic spread to the lungs from the several cancers. The presence of LMs seriously affects overall survival (OS), and the onset of pleural effusion further reduces the life expectancy of the patients. The aim of this retrospective study was to evaluate the usefulness of carcinoembryonic antigen (CEA), lactate dehydrogenase (LDH), and C-reactive protein (CRP) measurement in the pleural fluid of patients with LMs and malignant pleural effusion (MPE).

      Methods:
      The medical records of 22 patients (median age 68 years, range 46-86) with LMs (mainly from breast, urinary tract, and colorectal cancers) and MPE were analyzed. There were 13 (59.1%) males and 9 (40.9%) females. All patients underwent video-assisted thoracoscopic (VAT)-assisted thoracentesis and subsequent pleural fluid examination, including CEA, LDH, and CRP, which were measured using a chemiluminescent (CLIA) immunoassay (sandwich CLIA with native CEA coated to magnetic microbeads), a spectrophotometric assay (L-lactate as substrate), and an immunonephelometric assay (polystyrene particles coated with anti-human CRP monoclonal antibodies), respectively.

      Results:
      The OS was 6.7±5.2 months (range 1-23 months), and the levels of pleural markers were 10.4±21.6 ng/mL (CEA), 418.4±342.9 U/L (LDH), and 6.2±9.1 mg/L (CRP). No relationship was found between OS and the age of the patients (R = 0.14, p = 0.542), LDH (R=-0.31, p = 0.169) or CRP (R=-0.33, p = 0.136). There was a significant direct correlation between OS and CEA (R = 0.66, p = 0.0007). The relative regression line equations are reported in the table.rnTable: 153Prn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      ParameterRegression line equation
      Age of the patientsOS = 63.323797651973 + 0.23725477330884 age
      CEAOS = 2.0616207417742-4.4634344843623 CEA
      LDHOS = 470.3738751303-20.900941750959 LDH
      CRPOS = 12.210673177537-0.84687826996861
      rn

      Conclusions:
      In patients with MPE and LMs, only CEA pleural levels significantly related to OS, and can be considered a useful predictive factor. Further studies will eventually confirm our results.

      Clinical trial identification:


      Legal entity responsible for the study:
      Università degli Studi di Padova

      Funding:
      Università degli Studi di Padova

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      154P - Analysis of factors affecting survival in patients with simultaneous liver and pulmonary metastases from colorectal cancer (ID 450)

      12:30 - 12:30  |  Author(s): F. Lumachi, S.M. Basso, U. Zuccon, F. Maffeis, A. Del Conte, P. Ubiali

      • Abstract

      Background:
      In patients with colorectal cancer (CRC) the presence of simultaneous liver (LMs) and pulmonary metastases (PMs) is uncommon, but may significantly affect overall survival (OS). The aim of this study was to identify the prognostic factors (PFs) influencing OS in such a group of patients.

      Methods:
      We retrospectively reviewed the medical records of 36 patients (mean age 62.2±10.5 years) with CRC who underwent curative surgical resection and developed PMs during follow up requiring video-assisted thoracoscopic (VATS) metastasectomy. There were 21 (58.3%) males and 15 (41.7%) females. Twenty (55.6%) patients had already LMs at the time of VATS. Survival curves were estimated by Kaplan-Meyer method and compared with log rank testing. Cox proportional hazard model (HR) calculation (stepwise logistic regression) was used in the multivariate analysis, to identify the independent variables. A p-value of < 0.01 (two-tailed) was considered statistically significant.

      Results:
      The OS of the entire population ranged widely (30.6±25.1 months), according to patient age (p = 0.004), presence of involved mesenteric (p = 0.0003) or thoracic (p = 0.0001) lymph nodes, number of the involved nodes (p = 0.006), and simultaneous LMs and PMs (p = 0.0002). There was no relationship between gender (p = 0.67) or the mean size of metastases (p = 0.53) and OS. A tendency towards a correlation between age and the presence of lymph node involvement (p = 0.050) was also observed. The regression analysis showed that the number of LMs (HR = 1.37, 95% CI: 1.14-1.67, p = 0.0011), and the presence of node involvement, both thoracic (HR = 11.49, 95% CI: 1.49-88.58, p = 0.0191) and mesenteric (HR = 5.79, 95% CI: 1.27-26.30, p = 0.0230), were independent parameters affecting OS.

      Conclusions:
      In patients with CRC with both LMs and PMs only lymph node involvement and their number should be considered as negative PFs. In conclusion, the results of liver metastasectomy are independent of age, gender, and size of metastases, and thus this therapeutic approach should be suggested in all node-negative patients suitable for surgery, regardless of other PFs.

      Clinical trial identification:


      Legal entity responsible for the study:
      Università degli Studi di Padova

      Funding:
      Università degli Studi di Padova

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      155P - Combination of hepatic surgery and minimally invasive thoracic surgery using a subxyphoid approach through an abdominal incision (ID 323)

      12:30 - 12:30  |  Author(s): M. Lim, D.H. Kim

      • Abstract

      Background:
      If an intrathoracic mass is found in a patient scheduled for open hepatic surgery, combined surgery is not recommended because of the high risk. If combined surgery is essential, the thoracic approach must be minimally invasive. We here introduce a subxyphoid approach through an abdominal incision to minimize the invasiveness of combined thoracic and hepatic surgery.

      Methods:
      From April 2012 to December 2016, 17 patients requiring combined hepatic and thoracic surgery were treated via the subxyphoid approach through an abdominal incision. We retrospectively analyzed the clinical data and evaluated the feasibility of the procedure.

      Results:
      The hepatic lesions included 7 cases (41.2%) of colon cancer metastases and 10 cases (58.8%) of primary hepatocellular carcinoma. The types of liver resection performed included two wedge resections (11.8%), two multiple wedge resections (11.8%), eight segmentectomies (47.1%), three lobectomies (17.5%), and two other procedures (11.8%). Unilateral, mediastinal, and bilateral subxyphoid approaches toward thoracic surgery were employed in 11 (64.7%), 2 (11.8%), and 4 (23.5%) cases, respectively. The thoracic surgeries included six cases (35.3%) of wedge resection, five cases (29.4%) of multiple wedge resection, two (11.8%) segmentectomies, and four (23.5%) mediastinal mass excisions. The mean number of lung masses excised was 1.7±1.0 (range 1–5). No lung-related morbidities were noted; one patient died as a result of the abdominal procedure. The mean thoracic and total operation times were 79.7±63.0 min (range, 20–205 min) and 411.0±109.0 min (range, 255–635 min), respectively. Mean ICU and mean hospital stay did not differ between the patients described above and others who underwent open hepatic surgery during the same period.

      Conclusions:
      The subxyphoid approach through an abdominal incision allows aggressive treatment of intrathoracic masses in patients scheduled for open hepatic surgery. This approach did not have a significant impact on the time of operation and the period of the recovery. The approach is especially suitable for patients with bilateral lung lesions.

      Clinical trial identification:


      Legal entity responsible for the study:
      Department of Cardiovascular and Thoracic Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital

      Funding:
      Department of Cardiovascular and Thoracic Surgery, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      156P - Clinical outcome of cyberknife radiosurgery in brain metastases of non-small cell lung cancer: A single institutional experience (ID 444)

      12:30 - 12:30  |  Author(s): S. Lee, Y. Kim, Y.K. Won, S.J. Lee, J.H. Hong, Y.N. Kang, J.H. Kang, S. Hong, S.J. Kim, Y.K. Kim

      • Abstract

      Background:
      Stereotactic radiosurgery is a less invasive therapeutic modality for brain metastasis of non-small cell lung cancer (NSCLC). We retrospectively reviewed a single institutional experience using Cyberknife radiosurgery (CKRS).

      Methods:
      The patients diagnosed with brain metastasis of NSCLC who were treated with CK-RS from 2006 to 2016 in Seoul St. Mary’s Hospital were analyzed. Total 304 targets from 150 patients were included. The histology of NSCLC patients was as follows: adenocarcinoma (85%), squamous cell carcinoma (9%), and others (7%). Median 22 Gy (range: 17-30Gy, 1-8 fx) was given. Median 2 targets (range: 1-7 targets) were treated per patient. Whole brain radiotherapy (WBRT) was given to 44.7% of the patients. Total 68 patients (45.5%) were mutation positive and targeted therapy was given to 95 patients (63.3%).

      Results:
      Median follow-up time was 11.4 mo (range: 0-94.6 mo) from the last day of CKRS. Response to CKRS was observed in 86.1% of the targets. Median time to response was 2.9 mo (range: 0.2-58.8 mo). At the time of analysis, there were 14 recurrences (4.6%) and median time to recurrence was 13.3 mo (range: 4.1-62.6 mo). Intracranial failure defined as appearance of new metastasis other than sites previously treated with CKRS was observed in 81 patients (54.7%) at median 8.9 mo (range: 7-10.8 mo). The 1-year intracranial failure rates of the patients who received targeted therapy were 64.8% versus 55.3% of those who did not, respectively (P = 0.38). The 1-year intracranial failure rates of the patients who received CKRS alone were 64% versus 58.3% of those who received both CKRS and WBRT, respectively (P = 0.58). Radiation-associated change on MRI was observed in 98 targets (32.3%) at median 7.0 mo (range: 0.4-68.8 mo). Eleven patients (7.3%) required steroids for symptom alleviation at median 21 days (range: 7-42 days). Five patients (3.0%) underwent surgery after median 7.0 mo (range: 2.8-74.2 mo). The patients survived median 12.6 mo (range: 0.3-94.8 mo) after CKRS.

      Conclusions:
      According to our institutional experience, CKRS achieved adequate local control with tolerable toxicity in patients with brain metastasis of NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      The institutional review board reviewed and approved of the study.

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      157P - Mediastinal masses: A study of our cases by transthoracic needle biopsy (TTNB) versus open surgery as a diagnostic procedure (ID 187)

      12:30 - 12:30  |  Author(s): F. Gradica, L. Lisha, D. Argjiri, A. Cani, A. Leka, F. Kokici, S. Gradica, Y. Vata, F. Gradica, V. Rexha

      • Abstract

      Background:
      Mediastinal tumors are an uncommon abnormalities found in clinical practice. Transthoracic needle biopsy (TTNB) is done with imaging guidance and most frequently by a radiologist but and by a thoracic surgeon, for the aim is to diagnose a defined mass. It is integral in the diagnosis and treatment of many thoracic diseases, and is an important alternative to more invasive surgical procedures. Open biopsy is done by chamberlein procedures under general anesthesia.

      Methods:
      We evaluate the different malignant mediastinal mass (MMs) in various age groups and the sensitivity and early mortality rate of open biopsy 2004 to 2013 and transthoracic needle biopsy (TTNB) and core needle biopsy (CNB) 2013-2016. This was a prospective study of 80 patients who were consulted for MMs and underwent open biopsy from 2004 to 2013 and 20 patients underwent by thoracic surgeon TTNB and CNB under guidance of ultrasound or computed tomography (CT) scan from 2013 to 2016. Cytology and histological examinations were evaluated in all patients.

      Results:
      Among 80 cases, 63 were male and 17 were female were diagnosed by open biopsy from 2004 to 2013. Among 20 cases, 13 males and 7 females were diagnosed by TTNB. Mean age of presentation was 57 years old (ranging from 50-75 years old). Metastatic carcinoma and nonHodgkin's lymphoma are the common AMMs. Adequate tissue material by open biopsy was obtained in 80 cases and 17 of 20 cases (85%) by TTNB. Of these 17 patients, 15 (88,2%) cases were diagnosed correctly by TTNB, whereas 3 (11,8%) cases were not diagnosed definitely by TTNB. The sensitivity of TTNB for MMs was 85%, and no mortality whereas open biopsy were correctly diagnosed in 77 of 80 cases (96.25%) patients and operative mortality rate 2 patients (2.5%) and operative major compliactions in 3 (3.7%) patents had massive operative bleeding. Three cases (3.7%) had died after several mounths after open biopsy with out histological diagnosis. There is no significant difference of TTNB and open biopsy in carcinoma patients (P > 0.05). Operative mortality rate was higher for open biopsy in carcinomatous patients (3.7%) than for TTNB (0%).

      Conclusions:
      CT scan-guided TTNB and CNB in combination with FNAC are safe, minimally invasive, and cost-effective procedure, with low morbitity rate and major complications, which can provide a precise diagnosis in the MMs, and may obviate the need for invasive surgical approach. Invasive surgical approach should be performed whenever the diagnosis by TTNB or CNB is suspected of carcinoma but not established.

      Clinical trial identification:
      -

      Legal entity responsible for the study:
      N/A

      Funding:
      N/A

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      158TiP - Open label, multi-center, prospective study to investigate the efficacy and safety of osimertinib in brain metastases from patients with EGFR T790M positive NSCLC who have received prior therapy with an EGFR-TKI (APOLLO Study, NCT02972333) (ID 250)

      12:30 - 12:30  |  Author(s): L. Wang, L. Xing, L. Cao, L. Wang, X. Wang, J. Feng, Y. Shu, Y. Shi, Y. Song, J. Yu

      • Abstract

      Background:
      The development of EGFR-TKI have led to significant advances in patients with tumors harboring EGFR mutations (EGFRm). However, increasing incidence of central nervous system (CNS) metastasis (∼40%), including leptomeningeal metastasis (LM) and brain metastasis (BM), has been reported, particular in EGFR mutant NSCLC receiving EGFR TKI treatment. Due to limited blood brain barrier (BBB) penetration of current EGFR inhibitors (i.e. 1st generation EGFR TKIs such as gefitinib, erlotinib or icotinib and 2nd generation TKIs e.g. afatinib), these drugs can only exhibit limited efficacy on CNS metastasis. Furthermore, the acquiring resistance may commonly occur due to the development of EGFR T790M mutation. Osimertinib is a novel oral, potent and irreversible inhibitor of both EGFRm sensitizing and T790M resistance mutants. In a combined analysis from AURA (NCT01802632) and AURA2 (NCT02094261) study, the ORR of pts with CNS metastases was 56%, whilst 64% in pts without CNS metastases, demonstrating the potential benefits of osimertinib in pts with BM.

      Trial design:
      This is an open label, multi-center, prospective study to investigate the efficacy and safety of osimertinib in pts with BM. Pts with confirmed EGFR T790M positive NSCLC who received prior therapy with an EGFR-TKI and concurrent with brain metastasis will be enrolled. All eligible patients will have access to osimertinib 80mg once-daily as long as they show clinical benefit as judged by the investigator and in the absence of discontinuation criteria. All consenting pts will be required to provide CSF and blood samples pre-treatment, 6 weeks after treatment and at PD. All pts receiving osimertinib will be followed for clinical outcomes (tumor response, survival, etc) and patient reported outcomes at baseline and every 12 weeks (± 7days investigator per RECIST 1.1 until objective disease progression, intolerant toxicity, loss of follow up). A sample size of 100 patients will provide 80% power to evaluate the treatment profile of Osimertinib and its impact on the molecular evolution. The First subject in is on 11[th] January, 2017.

      Clinical trial identification:
      APOLLO Study (protocol number: NCT02972333) was released on November 23, 2016.

      Legal entity responsible for the study:
      Jinming Yu, Shandong Cancer Hospital

      Funding:
      AstraZeneca

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      159TiP - TTFields and radiosurgery for 1-10 brain metastases from NSCLC: The Phase 3 METIS study (ID 409)

      12:30 - 12:30  |  Author(s): M.P. Mehta, V. Gondi, P.D. Brown

      • Abstract

      Background:
      Tumor Treating Fields (TTFields) are non-invasive, loco- regional, anti-mitotic treatment modality, based on low intensity alternating electric fields. Efficacy of TTFields in non-small cell lung cancer (NSCLC) has been demonstrated in multiple in vitro and in vivo models, and in a phase I/II clinical study. TTFields treatment to the brain was shown to be safe and to extend overall survival in newly-diagnosed glioblastoma patients.

      Trial design:
      The study objectives are to test the efficacy, safety and neurocognitive outcomes of TTFields in this patient population. Patients (N = 270) with 1-10 brain metastases (BM) from NSCLC are randomized in a ratio of 1:1 to receive stereotactic radio surgery (SRS) followed by either TTFields or supportive care alone. Patients are followed-up every two months until 2[nd] cerebral progression. Patients in the control arm may cross over to receive TTFields at the time of 2[st] cerebral progression. Key inclusion criteria: Karnofsky performance status (KPS) of 70 or above, 1 inoperable or 2-10 brain lesions amenable to SRS, optimal standard therapy for the extracranial disease, no brain-directed therapy, no signs of significantly increased intracranial pressure, and no electronic implantable devices in the brain. Endpoints: Time to 1[st] cerebral progression based on the RANO-BM Criteria or neurological death (primary); time to neurocognitive failure based on the following tests: HVLT, COWAT and TMT; overall survival; radiological response rate; quality of life; adverse events severity and frequency (secondary). Treatment: Continuous TTFields at 150 kHz for at least 18 hours per day are applied to the brain within 7 days of SRS. The treatment system is a portable medical device allowing normal daily activities. The device delivers TTFields to the brain using 4 Transducer Arrays, which may be covered by a wig or a hat for cosmetic reasons. Patients receive the best standard of care for their systemic disease. Statistical Considerations: This is a prospective, randomized, multicenter study for 270 patients. The sample size was calculated using a log-rank test (based on Lakatos 1988 and 2002) and has 80% power at a two sided alpha of 0.05 to detect a hazard ratio of 0.57.

      Clinical trial identification:
      NCT02831959

      Legal entity responsible for the study:
      Novocure

      Funding:
      Novocure

      Disclosure:
      M.P. Mehta: Grants/research support: Novocure; Cellectar (both to institution, not to self) Advisor/board member: Board of Directors of Pharmacyclics Consultant: Cavion, Novocure, Varian, Agenus, Insys, Remedy, IBA Stock shareholder: Pharmacyclics. V. Gondi: Educational honoraria: Novocure Consultant: INSYX Therapeutics. All other authors have declared no conflicts of interest.

    • +

      160P - BAP1 in advanced sporadic malignant pleural mesothelioma (ID 304)

      12:30 - 12:30  |  Author(s): A. El Bastawisy, R. Shehab, A. Bahnassy, N. Sabri

      • Abstract

      Background:
      BAP1 mutation has been previously described in familial malignant pleural mesothelioma (MPM), however a little is known about its role in sporadic MPM.

      Methods:
      This is a prospective study including all eligible cases of MPM presenting to NCI, Cairo University, Egypt during the years 2013-2016. BAP1 analysis was done by PCR. Patients received first line chemotherapy (Pemetrexed/platinum). Primary objective was to evaluate the rate of BAP1 mutation in MPM, secondary objectives: To evaluate the correlation of BAP1 mutation and objective response rate (ORR), progression free survival (PFS) and overall survival (OS) in advanced MPM.

      Results:
      Among 120 cases, 38.5% showed positive BAP1 mutation.56.5% in the mutated group showed overall clinical benefit (CR+PR+SD) versus 46.2% in the wild group. (P = 0.34). One year PFS in the mutated group was 50.5% versus 38.5% in the wild group. (P = 0.31). One-year OS in the mutated group was 54.5% versus 47.6% in the wild group. (P = 0.67).

      Conclusions:
      BAP1 mutation is present in high frequency in a Caucasian population however it was not associated with clinical outcome after first line platinum based chemotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      National Cancer Institute and Faculty of Pharmacy, Cairo University, Egypt

      Funding:
      National Cancer Institute and Faculty of Pharmacy, Cairo University, Egypt

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      161P - DKK1 stabilization as a new malignant pleural mesothelioma therapeutic avenue (ID 490)

      12:30 - 12:30  |  Author(s): A. Guazzelli, E. Bakker, F. Sotgia, M. Listanti, M. Krstic-Demonacos, L. Mutti

      • Abstract

      Background:
      Malignant Pleural Mesothelioma (MPM) is a cancer with a hypoxic microenvironment. Hypoxic niches favor “stemness” in cancer cell formation and therefore represent a novel therapeutic target. DKK1 (dickkopf) abrogates WNT signaling via lipoprotein-related protein 5/6 (LRP5/6) and secreted frizzled related proteins (sFRPs)-mediated interference. Doxycycline (DXC) has the potential to be a repositioned drug with potent inhibition of mitochondrial biogenesis, targeting cancer stem-like cell (CSC) generation. The aim of this study has been to assess the effect of DXC on DKK1 expression and MPM-CSC generation.

      Methods:
      We have conducted a microarray analysis in MPM cell lines in normoxic and hypoxic (1% O~2~) conditions and on the basis of this experiment we have performed Western blot analysis of DKK1 and assessed MPM-CSCs generation (meso-spheres) in the same conditions before and following treatment with Doxycycline.

      Results:
      Microarray analysis showed a significant increase in DKK1 following treatment with DXC (7.56 fold). However, hypoxic conditions showed progressive DKK1 degradation compared to normoxic conditions (as assessed by Western blotting), thus confirming the role of WNT signaling in hypoxia. Treatment with DXC promoted DKK1 stabilization in both normoxia and hypoxia and significantly prevented CSC generation via hampering DKK1 degradation in hypoxic conditions.

      Conclusions:
      We have identified that both hypoxia and doxycycline exert differential cell-specific effects on DKK1 expression in cell monolayers, thus identifying DKK1 as a potential target for therapeutic development. DKK stabilization by DXC in hypoxic conditions paves the avenue of new therapeutic approaches aimed at WNT signaling regulation.

      Clinical trial identification:
      N/A

      Legal entity responsible for the study:
      Luciano Mutti\'s research group at the University of Salford

      Funding:
      G.I.Me. – Gruppo Italiano Mesotelioma and University of Salford

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      162P - Targeting the RON/MET/TAM signalling network in mesothelioma (ID 447)

      12:30 - 12:30  |  Author(s): A. Baird, M. Jarzabek, L. Shiels, S. Raeppel, S. Finn, S. Cuffe, H.I. Pass, I. Schmitt-Opitz, A.T. Byrne, S.G. Gray

      • Abstract

      Background:
      Malignant pleural mesothelioma (MPM) is an aggressive inflammatory cancer, with limited treatment options and poor survival rates. Simultaneously targeting the RON/MET/TAM family of receptor tyrosine kinases (RTK) may provide an effective novel therapeutic strategy for this disease.

      Methods:
      Expression of RON/MET/TAM and associated ligands were assessed in a cohort of patient samples and MPM cell lines. In vitro and in vivo experiments were undertaken to determine the efficacy of single and multi RTK targeting agents (LCRF0004, RXDX-106, BMS-777607).

      Results:
      mRNA expression of the RON/MET/TAM family was detected in a large panel of normal pleural and MPM cell lines. In a cohort of patient samples, mRNA levels of flRON, sfRON, c-MET, Axl and Tyro3 were increased in MPM tumour samples compared with benign pleural (p < 0.05). There was no difference detected in MERTK expression. In addition, MSP was also elevated in tumour tissue (p < 0.05), whereas GAS6 was not. Furthermore, no MET Exon 14 skipping mutations were detected. All RTK targeting agents displayed in vitro efficacy in terms of reduced proliferation and increased apoptosis. In an in vivo SQ xenograft model the multi-target TKI (BMS-777607) demonstrated superior anti-tumour activity compared with a single targeting agent (LCRF0004).

      Conclusions:
      Data suggests that a multiple TKI, targeting the RON/MET/TAM signalling network, is superior to selective RTKi as an interventional strategy in MPM.

      Clinical trial identification:
      N/A

      Legal entity responsible for the study:
      St. James\'s Hospital

      Funding:
      HRB

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      164P - Heart radiation dose as a risk factor for dyspnea worsening after multimodality treatment for non-small cell lung cancer and pleural mesothelioma: An exploratory analysis (ID 500)

      12:30 - 12:30  |  Author(s): A. Botticella, C. Billiet, G. Defraene, C. Draulans, K. Nackaerts, C. Deroose, J. Coolen, P. Nafteux, D. De Ruysscher

      • Abstract

      Background:
      The purpose of our study is to quantify the influence of heart dose on the early and late onset of dyspnea in a cohort of radiotherapy (RT)-treated non-small cancer patients (NSCLC) and malignant pleural mesothelioma (MPM) patients after multimodality treatment, according to the type of surgery.

      Methods:
      In 121 patients with multimodality-treated NSCLC and MPM the maximal dyspnea score (CTCAE 4.0) before RT and at an early (≤6 months) and a late (7-12 months) time point were obtained. Included patients needed to be clinically and radiologically progression-free 9 months after the end of RT. The difference (Δ) between the maximal dyspnea at ≤ 6 months and the pre-RT dyspnea or the maximal dyspnea at 7-12 months and the pre-RT dyspnea was calculated.

      Results:
      Our results show that 44% (50/113) of patients developed an early worsening of at least 1 point in their dyspnea score (Δdyspnea ≥1) after the end of RT. As independent predictors of an early worsening, we identified the MHD (for Δdyspnea ≥1: OR = 1.032, p = 0.04) and the dyspnea score before RT (for Δdyspnea ≥1: OR = 0.40, p = 0.0001; for Δdyspnea ≥2: OR = 0.35, p = 0.05). At a later timepoint, only the dyspnea score before RT (OR: 0.40, p = 0.001) was identified as predictor of for Δdyspnea ≥1.

      Conclusions:
      Our results, albeit exploratory, suggest that heart dose may play a role in the early worsening of the dyspnea in a heterogeneous cohort of multimodality treated RT patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      KUL UHasselt-ZOL-Jessa

      Funding:
      This study is part of the Limburg Clinical Research Program (LCRP) UHasselt-ZOL-Jessa, supported by the foundation Limburg Sterk Merk, province of Limburg, Flemish government, Hasselt University, Ziekenhuis Oost-Limburg and Jessa Hospital. This work was partly funded by the European Union’s Seventh Framework Programme for research, technological development and demonstration under grant agreement no 601826 (REQUITE). CB is supported by the Cancer foundation Limburg and a grant from ‘Kom op tegen kanker’. AB is supported by a grant from the Stichting tegen kanker/Fondation contre le cancer (CA/2014/354) and by Kom op Tegen Kanker (Stand up to Cancer, The Flemish Cancer Society).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      165P - Malignant pleural mesothelioma: A systematic review of first-line chemotherapy and analysis of Hong Kong cohort (ID 281)

      12:30 - 12:30  |  Author(s): H. Pang, J. Ji, K. Yan, C.C. Leung, D. Lam

      • Abstract

      Background:
      Malignant pleural mesothelioma (MPM) is a rare but aggressive cancer. Although the standard first-line chemotherapy for MPM has been established, many different treatments are still under investigation. Additionally, epidemiologic analysis of MPM patients in Hong Kong has not been conducted in recent years.

      Methods:
      To review the progress in first-line chemotherapy treatment for MPM patients between 2010 and 2016, and to study the epidemiology and prognostic factors of Hong Kong MPM patients from 2002 to 2015. A thorough literature search was performed on PubMed and Cochrane Library (CENTRAL), restricted to English papers released between January 2010 and May 2016. Hong Kong MPM patients were identified from Pneumoconiosis Medical Board (PMB) by reviewing medical records. Kaplan-Meier and log-rank tests were used to compare survival between categories. Prognostic factors were identified by Cox regression analysis.

      Results:
      22 studies were included in the review. Pemetrexed/cisplatin is the recommended first-line regime now. Studies suggested that carboplatin plus pemetrexed can be an alternative regimen to reduce toxicity. The addition of bevacizumab to pemetrexed/cisplatin showed promising results and may become a new paradigm. Gemcitabine plus cisplatin, especially administrated in low dose prolonged infusion, showed comparable efficacy with lower cost. From the Hong Kong PMB clinic, 102 MPM patients were identified, with male predominance, mean age of 69.1 years at diagnosis, mean latency of 49 years, median overall survival of 11.7 months and mainly epithelioid histological subtype. Other possible prognostic factors like symptoms of chest pain and dyspnea were explored.

      Conclusions:
      Cisplatin plus pemetrexed, has been supported in routine practices settings. Carboplatin can substitute cisplatin to reduce toxicity. Bevacizumab plus cisplatin-pemetrexed triplet with appropriate management of treatment-related toxicity is the most promising first-line treatment. Hong Kong MPM patients share similarities in terms of prognostic factors with those in the literature.

      Clinical trial identification:


      Legal entity responsible for the study:
      HKU

      Funding:
      Pneumoconiosis Compensation Fund Board

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      166P - Thymoma and thymic carcinoma: A real-life retrospective analysis (ID 472)

      12:30 - 12:30  |  Author(s): I. Attili, S. Frega, A. Pavan, G. Pasello, V. Polo, G. Zago, F. Rea, F. Calabrese, P. Conte, L. Bonanno

      • Abstract

      Background:
      Thymic epithelial tumors (TETs) include thymoma and thymic carcinoma, a group of rare and heterogeneous thoracic cancers. The management is primarily surgical, but pre- and post-operatory chemotherapy and radiotherapy are to be evaluated. Recurrences after surgery occur in 10-15% of resected tumors.

      Methods:
      We retrospectively collected TETs consecutively treated at our center between 2005 to 2016 and analyzed clinical and pathological features, response to treatment and patterns of relapse. Patients with uncomplete records or with less than 12-month follow-up were excluded from survival analysis.

      Results:
      Study population included 57 TETs. Median age was 55 years, male: female ratio was 1:1, all were ECOG PS 0-1 at diagnosis. Histologic subtype A to B1 thymoma was 21%, B2-B3 47.4%, thymic carcinoma 21%, others 11%. Paraneoplastic syndromes were present at diagnosis in 1/3 of the cases (85% myasthenia gravis), mainly less differentiated histologies (85% B2 thymoma or higher). C-kit immunohistochemistry has been evaluated only in 30 cases, 1/3 resulting positive, 80% associated to thymic carcinoma. 50 cases were included in survival analysis: median follow up was 52 months (m) (95%CI: 30-74m), median overall survival (OS) has not been reached (121m- NA) and was not affected by recurrences (p: 0.34). Treatments and relapse patterns are summarized in table. 66% of patients developed disease relapse (51% pleura) regardless of histology and radical surgery, median time to first relapse was 30 m (95% CI: 6-54m), median OSr (OS from first relapse) was not reached (47m-NA) and was significantly worse for thymic carcinomas (p: <0.0001); OSr was not affected by treatment type, number or site of relapse; the only variable able to affect time to relapse was histology: ῃ[2] 0.27.rnTable: 166PTreatment and relapse patterns in TETs [s: surgery; rt: radiotherapy; c: combined; r: relapse]rn

      rnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrnrn
      rnLocal treatment (s: rt: c)Systemic treatmentSystemic+ Local (s: rt: c)Best supportive care
      1[st] diagnosis (N:57)27 (12: 0: 15)327 (8: 4: 15)0
      1[st] r (N:33)14 (5: 2: 7)106 (1: 2: 3)3
      2[nd] r (N:17)9 (6: 1: 2)53 (1:1: 1)0
      3[rd] r (N:9)1 (1: 0: 0)51 (0: 0: 1)2
      4[th] r(N:4)1 (0: 0: 1)11 (0: 0: 1)1
      TOT5224386
      rn

      Conclusions:
      Real-life management of TETs include multidisciplinary evaluation, which is essential also at relapse to integrate local and systemic treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Istituto Oncologico Veneto

      Funding:
      Istituto Oncologico Veneto

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      167P - Role of surgery in the treatment of thymic carcinoma based on a 5-year experience (ID 325)

      12:30 - 12:30  |  Author(s): F.F. Caushi, I. Skenduli, J. Shkurti, D. Xhemalaj, A. Hatibi, R. Kortoci, B. Gega, H. Hafizi

      • Abstract

      Background:
      Invasive thymomas and thymic carcinomas are relatively rare neoplasms, which together represent about 0.2-1.5% of all malignancies. Thymic carcinomas account for only 0.06% of all thymic neoplasms and have extremely poor prognosis. In about 50% of the patients, thymic carcinomas are detected by chance with plain-film chest radiography. Nowadays the surgery is going to earn more and more space in their multimodality treatment because of new techniques and new technology.

      Methods:
      We reviewed all cases with histologically confirmed thymic carcinoma treated between 2011 and 2016. This study was performed to investigate the tumor characteristics, treatment approach, and prognosis of patients with thymic carcinoma.

      Results:
      There were 47% males and 53% females in this study. Mean age was 45,4 years. Thymic carcinoma was diagnosed in 13% of patients with mediastinal tumors, as opposed to 60% for thymoma. None of the thymic carcinoma patients had concomitant myasthenia gravis. All of the patients received surgical intervention and the diagnosis was made by pathologic study. The pathologic subtypes of thymic carcinoma included 40% squamous cell carcinomas, 35% undifferentiated carcinomas, 20% carcinoid tumors, and the rest other types of tumors. Surgery performed was en-bloc resection of the tumor in 22% of cases accompanied by partial resection of superior vena cava, total resection of the left innominate vein and partial pericardectomy, and debulking surgery in 18%. In all other cases surgery was undertaken for diagnostic purposes. No perioperative deaths occurred. All cases were treated postoperatively with adjuvant radio and/or chemotherapy. The mean of hospitalized days after surgery was 12±2.

      Conclusions:
      Owing to the paucity of cases, optimal management of thymic carcinoma has yet to be defined, but one thing is for sure, that it requires multimodality treatment. Our experience supports the pivotal role of surgery in managing thymic carcinoma. En-bloc resection of the tumor is an essential treatment for favorable outcomes of locally advanced thymic tumors, while debulking surgery may be considered for patients in advanced stages because it minimizes the tumour size and area for irradiation postoperatively.

      Clinical trial identification:


      Legal entity responsible for the study:
      Fatmir Caushi

      Funding:
      Fatmir Caushi

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      168P - Clinical and prognostic characteristics of primary pulmonary non-Hodgkin’s lymphoma: A retrospective analysis of 38 cases in a Chinese population (ID 194)

      12:30 - 12:30  |  Author(s): R. Qiao, B. Han, R. Zhong, Y. Zhao, T. Chu, L. Jiang, H. Zhong

      • Abstract

      Background:
      Primary pulmonary non-Hodgkin’s lymphoma(NHL) is very rare, and although the prognosis is favorable, clinical features, beneficial diagnostic procedures, prognostic factors and optimal management have not been clearly defined.

      Methods:
      In this study, thirty-eight cases of primary pulmonary NHL treated in Shanghai Chest Hospital during a 10-year period were retrospectively reviewed, and clinicopathological features and prognosis were analyzed.

      Results:
      There were twenty-eight patients with mucosa-associated lymphoid tissue (MALT) lymphoma, three with diffuse large B-cell lymphoma, one with peripheral T-cell lymphoma, one with mantle cell lymphoma and five with unclassified B-cell lymphoma. The cohort consisted of 21 male and 17 female patients with a median age of 57.5 years. At presentation, 36.8% of patients were asymptomatic, and unilateral tumors occurred more frequently than bilateral and predominantly in the right lung. Thirty-three patients underwent surgical resection single or combination chemotherapy, and five patients received combination chemotherapy alone. Overall survival(OS) was significantly longer in patients with MALT lymphoma than that of non-MALT lymphoma (129.9 vs. 71.5 months, P = 0.019 by log-rank test). Patients who had received surgical resection had a better OS (126 vs. 65.4 months, P = 0.036 by log-rank test). Additionally, multivariate analysis showed that elevated serum lactate dehydrogenase (LDH) level was independently associated with a poor OS (P = 0.048).

      Conclusions:
      Primary pulmonary NHL has atypical clinical manifestations and non-specific imaging changes. Surgical resection is vital in clarifying the diagnosis and obtaining a favorable prognosis. Serum LDH level was an independent prognostic factor.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      Shanghai Chest Hospital

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      169P - Pulmonary carcinoid tumors: Experience from an oncology center (ID 454)

      12:30 - 12:30  |  Author(s): L. Bei, A. Silva, A.L. Cunha, C. Franco, J. Oliveira, I. Azevedo, I. Pousa, A. Rodrigues, M. Soares, D. Pereira

      • Abstract

      Background:
      Pulmonary carcinoid (PC) tumors are a rare group of pulmonary neoplasms, characterized by neuroendocrine differentiation and indolent clinical behavior. Typical carcinoid (TC) account for the majority of cases, are low-grade and rarely metastasizes. Atypical carcinoid (AC) have worse prognosis, with greater tendency to metastasize and recur locally.

      Methods:
      Retrospective analysis of histologically confirmed PCs diagnosed between 2008 and 2016. Descriptive analysis of the main demographic, clinical and prognostic characteristics was performed. Overall survival (OS) was evaluated using the Kaplan-Meier.

      Results:
      We identified 55 patients (PTS). TC 58.2% (n = 32), AC 36.4% (n = 20), not other specified 3.6% (n = 2) and one case of combined TC and AC (bilateral). Female 56.4% (n = 31), male 43.6% (n = 24). Median age 62 years (range 18-83). Current smoker (SMK) 10.9% (n = 6), ex-SMK 10.9% (n = 6) and non-SMK 58.2% (n = 32). Secondary malignancies in 27.3% (n = 15) of PTS: thyroid (n = 4), breast (n = 3), prostate (n = 3), colorectal (n = 2), endometrium (n = 1), cervix (n = 1) and non-small lung cancer (n = 1). Staging (AJCC 7th Ed.): I (65.5%, n = 36), II (7.3%, n = 4), III (7.3%, n = 4), IV (16.4%, n = 9). Surgery was performed in 38 (86.3%) PTS with localized disease: lobectomy 78,9% (n = 30) pneumonectomy 13.2% (n = 5) and segmentectomy 7.9% (n = 3) and the majority (>90%) had systematic nodal dissection. One PT had adjuvant chemotherapy (CT) - stage IIIA. Recurrence was documented in 5 PTS submitted to complete resection. For advanced or recurrent disease, first line systemic treatment (Tx) was proposed in 9 PTS: somatostatin analogues (SA) (n = 3), CT (n = 3) and SA + CT (n = 3). The CT regimens were platinum + etoposide, streptozocin (STZ) + 5-FU and STZ + doxorubicin. Two PTS were treated after disease progression with further CT, one of them received 177Lutetium DOTA octreotide. 4 PTS underwent radiotherapy in palliative setting. Radioembolization was used to treat liver metastasis in 3 PTS. 5-year OS was 76,2%.

      Conclusions:
      PC’s diagnosis and Tx require an experienced multidisciplinary team. Our results corroborate the literature data regarding epidemiology, high percentage of localized disease amenable for surgery and OS. Despite the emergence of new therapies, advanced disease remains with limited Tx options.

      Clinical trial identification:


      Legal entity responsible for the study:
      Instituto Português de Oncologia do Porto

      Funding:
      Instituto Português de Oncologia do Porto

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      170TiP - LUME-Meso: Randomised phase II/III study of nintedanib (N) + pemetrexed/cisplatin (PEM/CIS) followed by maintenance N or placebo (P) in chemo-naïve patients with malignant pleural mesothelioma (MPM) (ID 274)

      12:30 - 12:30  |  Author(s): S. Popat, R. Gaafar, A. Nowak, A. Tsao, J. Van Meerbeeck, N. Vogelzang, T. Nakano, D. Velema, N. Morsli, G. Scagliotti

      • Abstract

      Background:
      PEM/CIS is the standard first-line treatment for MPM, with median overall survival (OS) of ∼1 year. N is a triple angiokinase inhibitor of vascular endothelial growth factor (VEGF) receptors 1–3, platelet-derived growth factor (PDGF) receptors α/β and fibroblast growth factor receptors 1–3, as well as Src and Abl kinases. VEGF and PDGF overexpression are associated with poor prognosis in MPM, and N has demonstrated efficacy in preclinical MPM models. We performed a randomised Phase II trial of N or P + PEM/CIS in MPM followed by maintenance N or P; progression-free survival (PFS) was the primary endpoint. An internal Data Monitoring Committee recommended the study be expanded to include a confirmatory Phase III part. With regulatory authority guidance, the Phase II data were unblinded, demonstrating a PFS benefit with N (hazard ratio 0.56, 95% confidence interval 0.34–0.91; p = 0.017); these data assisted in planning the Phase III part including sample size estimation, and N was granted U.S. Food & Drug Administration orphan drug designation for the treatment of MPM in December 2016. The Phase III part (NCT01907100) is recruiting.

      Trial design:
      For Phase III, 450 chemo-naïve patients worldwide (>100 sites in 27 countries) aged ≥18 years with unresectable MPM of epithelioid histology and Eastern Cooperative Oncology Group performance score 0–1 will be randomised 1:1 to receive up to 6 21-day cycles of PEM (500 mg/m[2])/CIS (75 mg/m[2]) on Day 1 plus N or P (200 mg twice daily, Days 2–21), followed by N or P monotherapy until disease progression or undue toxicity. The primary endpoint is PFS; the key secondary endpoint is OS. An adaptive design will be used at the time of the primary PFS analysis to reassess the number of OS events for sufficient OS power. Other secondary endpoints are objective response and disease control (using modified Response Evaluation Criteria in Solid Tumors). The frequency/severity of adverse events and health-related quality of life will also be assessed. An exploratory analysis of predictive/prognostic biomarkers is planned.

      Clinical trial identification:
      NCT01907100

      Legal entity responsible for the study:
      Boehringer Ingelheim Pharma GmbH & Co. KG

      Funding:
      Boehringer Ingelheim Pharma GmbH & Co. KG

      Disclosure:
      S. Popat: Acknowledges NHS funding to the NIHR Biomedical Research Centre at The Royal Marsden and the ICR, and is consultant to and has received honoraria from Boehringer Ingelheim and Eli Lilly. A. Nowak: Acknowledges funding from the National Health and Medical Research Council of Australia to the National Centre for Asbestos Related Diseases. A. Tsao: Received honoraria from Eli Lilly, Roche, Novartis, AstraZeneca, Ariad, Boehringer Ingelheim, Genentech, BMS, Seattle Genetics, and has received research funding from Eli Lilly, AstraZeneca, Millennium, BMS, Seattle Genetics, and Polaris. J. Van Meerbeeck: Received institutional funding through research grants from the Belgian Foundation against Cancer and Flemish Kom op tegen Kanker Fund. N. Vogelzang: Received an honorarium from Boehringer lngelheim for services on the steering committee of this study. D. Velema: Employee of Boehringer Ingelheim. N. Morsli: Employee of Boehringer Ingelheim. G. Scagliotti: Consultant for Eli Lilly and has received honoraria from Eli Lilly, Roche, Pfizer, Novartis, AstraZeneca, and Clovis Oncology. All other authors have declared no conflicts of interest.

  • +

    Welcome reception (ID 33)

    • Event: ELCC 2017
    • Type: Welcome reception
    • Track:
    • Presentations: 1
    • Moderators:
    • Coordinates: 5/05/2017, 13:00 - 13:30, Hall 1
    • +

      Welcome Reception (ID 567)

      13:00 - 13:20  |  Author(s): F.R. Hirsch

      • Abstract

      Abstract not provided

  • +

    Poster Display session (Friday) (ID 65)

    • Event: ELCC 2018
    • Type: Poster Display session
    • Track:
    • Presentations: 186
    • Now Available
    • Moderators:
    • Coordinates: 4/13/2018, 12:30 - 13:00, Hall 1
    • +

      4P - Assessment of PD-L1 expression by immunohistochemistry in histological and cytological non-small cell lung carcinoma (NSCLC) in the era of immunotherapy: A national Irish study (Now Available) (ID 630)

      12:30 - 12:30  |  Presenting Author(s): A. Fabre  |  Author(s): K. Breen, J. McCormack

      • Abstract
      • Slides

      Background:
      Evasion of immune system is a hallmark of cancer, which enables cancer cells to escape the attack from immune cells, by expression of immune inhibitory signalling proteins such as programmed death-ligand-1 (PD-L1); PD-L1 binds to programmed death-1 (PD-1) expressed on immune cells (T, B, dendritic and NK T-cells) to suppress anti-cancer immunity. Anti PD-L1antibodies (such as pembrolizumab) are now being used for the treatment of some cancers.

      Methods:
      We assessed the expression of PD-L1 on NSCLC by immunohistochemistry using the Dako IHC22C3 pharmDx® kit on the Dako Autostainer link48®. Slides were read and scored using the Dako guidelines based on a Tumour Proportion Score (TPS) (% of tumour cells expressing PD-L1, ≤1% (negative), 1–49% (low), ≥50% (positive)). Immune cells were not scored.

      Results:
      The 16 months-patient cohort was made up of 870 patients, from SVUH (43.9%) and Irish national hospitals (61.2%), 49.5% females and 51.5% males. Overall staining patterns were as follows: 31.5% positive (high TPS >50%), 23.1% low (low TPS 1–49%), 42.8% negative (TPS <1%). Specimens included cytology (22.9%, of which 57% were EBUS samples), biopsy (77.1%, 39% were lung/bronchial biopsies) and surgical (15.6%) specimens. Adenocarcinomas represented 59.3% of all NSCLC and 33.5% had a high TPS (≥50%) score. Squamous cell carcinomas (33.7%) were positive (≥50%) in 29.7%. Histology and cytology samples had similar distribution of high, low and negative TPS cases.

      Conclusions:
      This is the first comprehensive collection of PD-L1 testing data in NSCLC in Ireland. Our results broadly with the KEYNOTE-010 study validating our scoring. Some of these patients are now receiving pembrolizumab treatment. Heterogenous tumour populations and strong staining of inflammatory cells can make assessment difficult.

      Clinical trial identification:


      Legal entity responsible for the study:
      St. Vincent's University Hospital Dublin

      Funding:
      MSD

      Disclosure:
      A. Fabre, K. Breen, J. McCormack: Financial funding received from MSD.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      5P - Heterogeneity of PD-L1 expression in primary tumors and paired lymph node metastases of non-small cell lung cancer (Now Available) (ID 501)

      12:30 - 12:30  |  Presenting Author(s): Y. Saito  |  Author(s): S. Horiuchi, H. Morooka, T. Ibi, N. Takahashi, T. Ikeya, E. Hoshi, S. Shimizu

      • Abstract
      • Slides

      Background:
      Programmed cell death ligand 1 (PD-L1) expression by immunohistochemistry (IHC) is an important predictive factor for patients with metastatic non-small cell lung cancer (NSCLC). Although PD-L1 IHC 22C3 pharmDx (Dako)® is the first FDA-approved companion diagnostic test for PD-L1 testing in NSCLC, it remains controversial which specimen is preferred from primary tumor or metastatic tissue.

      Methods:
      We analyzed formalin-fixed paraffin-embedded tissues of 24 paired primary tumor and metastatic lymph node (LN) from patients with pathological N1 or N2 NSCLC. All of them underwent surgical lung resection and LN dissection (or sampling) at Saitama Cardiovascular and Respiratory Center, and none of them received any chemotherapy or radiotherapy before surgery. According to PD-L1 IHC 22C3 pharmDx interpretation manual, all specimens were stained by using EnVision FLEX visualization system on Autostainer Link 48 in laboratory of LSI Medience Corporation in Japan. Next, we scored and divided each sample into three levels based on a Tumor Proportion Score (TPS); 1) Group 1 (TPS: <1%), 2) Group 2 (TPS: 1–49%) and 3) Group 3 (TPS: >=50%).

      Results:
      All patients were Asian (Japanese) with average age 67.3 years old (49–83). There were 8 females (33.3%), 8 never smoker (33.3%), 19 adenocarcinomas (79.2%) including 7 EGFR mutation and 1 ALK positive tumor. Number of Group 1, 2 and 3 were 5 (20.9%), 15 (62.5%), 4 (16.7%) in primary tumor, while that were 11 (45.9%), 9 (37.5%), 4 (16.7%) in metastatic LN, respectively. No primary tumor in Group 3 paired metastatic LN showing high PD-L1 expression.

      Conclusions:
      We found apparent discrepancy of TPS between primary tumor and metastatic LN of NSCLC, and there are concerns that will cause serious problem when we decide chemotherapeutic agents. It is future subject to explore which site should be biopsied for PD-L1 IHC.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Saitama Cardiovascular and Respiratory Center Research Grant, Eiken Chemical Co., Ltd

      Disclosure:
      Y. Saito: This work was supported by the Saitama Cardiovascular and Respiratory Center Research Grant (Grant No. TE17, for Yuichi Saito) and Eiken Chemical Co., Ltd. These funding bodies had no rule in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      6P - Mechanisms of regulating PD-L1 expression in non-small cell lung cancer during EMT process (ID 226)

      12:30 - 12:30  |  Presenting Author(s): F. Li  |  Author(s): L. Liang

      • Abstract

      Background:
      Non-small cell lung cancer (NSCLC) is one of the most severe malignant tumors in the world. Recent studies have reported the important role of PD-L1 in immune escape of tumor microenvironment.

      Methods:
      In this study, we explored the molecular mechanisms in regulating the PD-L1 expression during the epithelial-to-mesenchymal transition (EMT) process treated with transforming growth factor β-1 (TGFβ-1) and -2 (FGF-2) in NSCLC. The phenotypic alteration of EMT was evaluated by wound healing assay and western blot. And the protein expression was evaluated by Western Blot and Flow Cytometry Analysis. Furthermore, the expression of PD-L1 was increased significantly treated with Gefitinib in tumor xenograft model.

      Results:
      The results showed that EMT promoted the expression of PD-L1 remarkably in A549 cell line while have no obvious influence on H1650 and H1975 cell lines. Furthermore, AKT pathway inhibitor Ly294002, Erk pathway inhibitor PD98059 and TAK1 pathway inhibitor 5Z-7 inhibited the expression of PD-L1 in A549 and H1650, but not H1975 in EMT process. Besides, our study indicated that AKT, Erk and TAK1 pathways regulated the expression of PD-L1 by mediating transportation of the transcription factor Stat3 and the subunit p65 of NF-κB from cytoplasm to nucleus.

      Conclusions:
      Take together, our study supports the role of ERK, AKT and TAK1 in mediating the expression of PD-L1 during EMT process and represents a promising strategy for treatment NSCLC in clinic.

      Clinical trial identification:
      none

      Legal entity responsible for the study:
      Peking University Third Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      7P - The regulation of FOXP3 expression by GLI-1 in non-small cell lung cancer cells and its influence on lung cancer cell stemness (ID 174)

      12:30 - 12:30  |  Presenting Author(s): H. Qi  |  Author(s): H. Jia, G.G. Chen

      • Abstract

      Background:
      Later numerous publications have demonstrated that lymphocytic FOXP3 is significantly associated with immune suppression. However, there are conflict data concerning its function among malignant cells. In colorectal cancer cells the inhibition of FOXP3 expression can result in increased cancer stemness. However whether this is the case in lung CSC or whether it is related to GLI-1 remains unclear.

      Methods:
      In this study, a series of in vivo and in vitro experiments will be conducted to clarify the regulation of FOXP3 expression in non-small-cell lung cancer cells, and its influence upon lung cancer cell stemness.

      Results:
      The cells with FOXP3 over expression and negative controls were applied for mRNA expression microarray experiments, and the results showed that when the expression of FOXP3 was up regulated, the expression of GLI-1 was oppositely regulated. Three NSCLC cell lines were treated with NNK for different periods. The results show that with the treat time progressing, the expression of FOXP3 was up regulated with the expression of GLI-1 down regulated. When the FOXP3 expression was up regulated, the expression of GLI-1 was suppressed, while the expression of cancer stem cell markers includes SOX2, Nanog and CD133 were also up regulated, which meant the cancer stemness increased. When the GLI-1 expression was up regulated, the expression of FOXP3 was increased, and the expression of cancer stem cell markers include SOX2 and Nanog were also up regulated, which meant the cancer stemness increased. For tumors with FOXP3 over expressed, the expression of GLI-1 was up regulated and the cancer stemness increased. GLI-1 and FOXP3 might bind with each other inside cells. It was easier for H460-FOXP3 cells to form tumor sphere, and the diameter is much larger.

      Conclusions:
      GLI-1 might influence lung cancer stem cells by regulating the expression of FOXP3.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chinese University of Hong Kong

      Funding:
      General Research Fund from Chinese University of Hong Kong

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      8P - Noncanonical Wnt11, a tumor suppressive gene by antagonizing canonical Wnt signaling, represents a putative molecularly therapeutic target in lung cancer (ID 502)

      12:30 - 12:30  |  Presenting Author(s): X. Wang  |  Author(s): L. Li, T.S.K. Mok, Q. Tao

      • Abstract

      Background:
      Most studies focused on the role of canonical Wnt signaling pathway, an increasing field of research, however, is concerning about the noncanonical Wnt pathways. The role of Wnt11, a noncanonical Wnt family member, has not been established in lung cancer. Epigenetic inactivation of tumor suppressive genes through promoter CpG methylation is a fundamental regulatory process during tumorigenesis.

      Methods:
      The expression levels of Wnt11 were assessed in human normal tissues and lung cancer cell lines panel by semi-quantitative reverse transcription-PCR(RT-PCR). The promoter CpG methylation of Wnt11 were tested in bisulfite treated DNA by methylation-specific PCR (MSP). Western blots assay, colony formation assay, cell proliferation assay, wound-healing assay, dual-luciferase reporter assay and apoptosis assay were used to characterize the changes induced by overexpression of Wnt11.

      Results:
      In our study, analysis of Wnt11 expression revealed it was broadly expressed in human normal adult and fetal tissues, while it's frequently downregulated or silenced in multiple lung cancer cell lines. By performing methylation-specific PCR (MSP), promoter CpG methylation of Wnt11 were frequently detected in multiple lung cancer cell lines. Functional assays show that ectopic expression of Wnt11 could suppress tumor cell growth, possibly through inducing apoptosis. Moreover, Wnt11 represses canonical Wnt/β-catenin signaling and AKT signaling pathway. Wnt11 overexpression also reversed EMT and downregulated stem cell markers.

      Conclusions:
      Together our data suggest that in lung cancer, Wnt11 is lost by methylation and represents a tumor suppressor by antagonizing canonical Wnt/β-catenin signaling and AKT signaling pathway. Restoration of Wnt11 expression through demethylation could be an important therapeutic approach in the treatment of lung cancer.

      Legal entity responsible for the study:
      The Chinese University of Hong Kong

      Funding:
      RGC (TBRS #T12-401/13R), China Natural Science Foundation (NSFC #81572327), Johns Hopkins Singapore, and VC special research fund from The Chinese University of Hong Kong

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      9P - Mesenchymal stem cells’ microvesicles from primary and metastatic NSCLC niches differentially modulate lung cancer cells (Now Available) (ID 160)

      12:30 - 12:30  |  Presenting Author(s): O. Attar-Schneider  |  Author(s): L. Drucker, M. Gottfried

      • Abstract
      • Slides

      Background:
      Novel therapeutic approaches are urgently needed in lung cancer, particularly ones that address the malignant cells in their stroma microenvironment critical to drug resistance and disease relapse. The stroma constituents, mesenchymal stem cells (MSCs), interact with cancer cells by various methods including transfer of microvesicles (MVs) that vehicle protein, mRNA and microRNAs thereby altering recipient cells’ phenotype. Here, we examined the effect of MSCs’ MVs from primary (lung) and metastatic (bone marrow (BM)) niches on NSCLC cells with emphasis on translation initiation's (TI) role in the process.

      Methods:
      Lung and BM MSCs’ MVs were isolated and applied to NSCLC cell lines (H1299, H460). MVs uptake was confirmed and NSCLC cells were assayed for: viability (WST-1); cell count/death (trypan); proliferation (PCNA); migration (scratch); autophagy; TI status (factors, regulators, targets); MAPKs activation (immunoblotting).

      Results:
      We observed increased viability, proliferation and migration of Lung-MSCs’ MVs treated NSCLC cells whereas, BM-MSCs’ MVs treated cells showed reductions (50–90% and −40%, respectively p < 0.05). Correspondingly, Lung MSCs’ MVs elevated TI status whereas BM-MSCs’ MVs diminished it (60–120% and −30–70%, respectively p < 0.05). The opposite trend was also evident in MAPK activation and autophagy induction.

      Conclusions:
      Our observations not only depict a role for MSCs’ MVs in NSCLC phenotype but also display distinct differences between the primary and metastatic niches and may emphasize specific factors that shape disease progression. Further studies into the mechanism underlying the MVs-NSCLC cells’ contact and cargo transfer may promote the design of a therapeutic approach that will sabotage the dialogue between NSCLC cells and MSCs.

      Clinical trial identification:


      Legal entity responsible for the study:
      Meir Medical Center

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      10P - The micro-environmental cross talk between mast cells and lung cancer cells through cell-to-cell contact (Now Available) (ID 170)

      12:30 - 12:30  |  Presenting Author(s): R. Shemesh  |  Author(s): Y. Gorzalczany, N. Peled, R. Sagi-Eisenberg

      • Abstract
      • Slides

      Background:
      Mast cells (MCs) are key effectors in allergic reactions, but are also involved in tissue remodeling, wound healing and protection against pathogens. MCs infiltrate tumors and their number within the tumor microenvironment in certain cancer types, such as lung cancer, has been correlated with poor prognosis. The nature of crosstalk between lung cancer and MCs remains poorly resolved. In this study, we investigated the activation patterns within the MCs following cell-to-cell contact with lung cancer cells.

      Methods:
      Human MCs (HMC-1 and LAD-2) were exposed to Human lung cancer cell (H1299) derived membranes to recapitulate cell contact mediated activation. Lysates of MCs were tested for protein expression and posttranslational modifications (i.e. phosphorylation) by targeted western blotting. We unraveled the intracellular signaling molecules that are necessary for this signaling pathway by a pharmacological approach using several inhibitors. Each treatment was repeated at least 3 times.

      Results:
      H1299 membrane exposure activated the ERK1/2 MAP kinases in HMC-1 and in LAD-2 cells. AKT signaling was also activated in LAD-2 cells as a result of this contact. Furthermore, we discovered that inhibition of protein kinase C (PKC) augments the activation of ERK1/2 in LAD-2 cells, while it inhibits ERK1/2 activation in HMC-1 cells. Activation of AKT is inhibited by PI3K and PKC inhibitors.

      Conclusions:
      Our results suggest that H1299 membranes activate ERK1/2 in MCs. In addition, we discovered that there is an important difference between ERK1/2 MAP kinase signal transduction in HMC-1 and LAD-2 cells, whereby PKC is an inhibitor of the H1299 stimulated activation of ERK1/2 in LAD-2 cells, while it mediates ERK 1/2 activation in HMC-1 cells. Furthermore, we can conclude that H1299 membranes activate AKT and that the activation is mediated by PI3K and PKC. Rachel Shemesh and Yaara Gorzalczany contributed equally to this work. Correspondence Authors: Nir Peled, Ronit Sagi-Eisenberg.

      Clinical trial identification:


      Legal entity responsible for the study:
      Prof. Nir Peled, Prof. Ronit Sagi-Eisenberg

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      11P - Glycolysis enzyme screenings identify non-glycolytic function of aldolase A that interacts and alters F-actin dynamics to promote lung cancer metastasis (Now Available) (ID 168)

      12:30 - 12:30  |  Presenting Author(s): Y. Chang  |  Author(s): M. Hsiao

      • Abstract
      • Slides

      Background:
      In recent years, cancer metastasis remains a serious issue for drug development and target therapy. Although the metabolic reprogramming including glycolysis appears to promote cancer metastasis, the molecular mechanism by which this occurs remains unclear.

      Methods:
      From high throughput screening of glycolytic enzymes, we investigated aldolase A (ALDOA) as the most significant enzyme promoting cell metastasis in vitro and in vivo. Furthermore, we recruited the enzyme inhibitors and enzyme-dead constructs to compromised function of aldolase A.

      Results:
      We established a His-tagged ALDOA construct model and found ALDOA directed protein-protein interaction (PPI) with g-actin and the status correlated with malignant cancer cells, whereas in normal cell it did not. Meta-analysis of intergraded RNA and protein levels suggested ALDOA could be a prognostic marker in several cancer types. Moreover, compared with non-tumor tissues increased levels of ALDOA and g-actin are commonly detected in malignancies and strongly predict a worse prognosis in cancer patients. Therefore, we designed a specific peptide to block the interaction between ALDOA and g-actin to decrease the metastatic ability of cancer cells in vitro and prolong survival rate in vivo.

      Conclusions:
      These findings suggest a new therapeutic strategy for targeting the cancer-associated PPI between ALDOA and g-actin to combat metastatic cancers.

      Clinical trial identification:


      Legal entity responsible for the study:
      Genomics Research Center, Academia Sinica, Taiwan

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      12P - Cigarette smoke promotes proliferation of non-small cell lung cancer (NSCLC) via enhancing glucose uptake and glycolysis (Now Available) (ID 454)

      12:30 - 12:30  |  Presenting Author(s): I. Hsieh  |  Author(s): F. Cheng, C. Tu, C. Chen, T. Hsia, B. Wang, Y. Yeh, W. Huang

      • Abstract
      • Slides

      Background:
      Cigarette smoking has been demonstrated as an important risk factor for lung cancer and causes poor prognosis. The critical roles of aerobic glycolysis and Warburg effect in supplying building blocks and energy for proliferation of lung cancer cells have also been well documented. However, it still remains unclear whether and how cigarette smoke alters the glucose uptake and glucose metabolism to promote the proliferation of NSCLC.

      Methods:
      Glycolysis and mitochondrial respiration rate were quantified by Seahorse XF analyzer. The gene expressions were assessed by quantitative RT-PCR and immunoblot. The cell lines were manipulated to inhibit or knockdown glucose transporters by glucose transporter competitive inhibitors and si-RNA. Fluorescence–labeled glucose analogue was employed to measure the glucose uptake ability. Cell viability and proliferation rate were analyzed by MTT assay and IncuCyte live cell analysis system respectively. Biochemical assays were used to evaluate the levels of glycolytic products.

      Results:
      NSCLC A549 and NCI-H292 cell lines were treated with cigarette smoke extract (CSE) or its carcinogenic ingredient Benzo[α]pyrene (B[α]P) as the cigarette smoke exposure models. Cigarette smoke promoted glucose uptake and glucose metabolism in NSCLC by increasing glucose transporter 3 (GLUT3) and sodium glucose transporter 1 (SGLT1) expression. The enhancing glycolysis activity was demonstrated in [13]C labelling LC/MS detection and further confirmed in biochemical assays of endogenous lactate and NADH production. Inhibiting glucose transporters were related to promoting survival in cigarette smoke treated cancer cells.

      Conclusions:
      The results suggest that targeting glucose transporters or glucose metabolism related enzymes in combination with the standard treatment are potential therapeutic strategies in treating smoker lung cancer patients.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      China Medical University, Taiwan

      Funding:
      Ministry of Science and Technology, Taiwan

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      13P - The identification of binding targets for pro-metastatic protein NIFK in lung cancer (ID 325)

      12:30 - 12:30  |  Presenting Author(s): T.C. Lin

      • Abstract

      Background:
      We previously identified nucleolar protein interacting with the FHA domain of pKi-67 (NIFK) as a poor prognostic marker in lung cancer. Furthermore, NIFK overexpression promotes cancer metastasis via decreasing casein kinase 1α (CK1α) expression, a suppressor of pro-metastatic TCF4/β-catenin signaling. We further observe the axis is induced by the downregulation of a novel transcription factor RUNX1. However, the interplay between NIFK, CK1α and RUNX1 remains largely unknown. In this study, we aim to unravel the interactive mechanism of the signaling axis NIFK/RUNX1/CK1α, and to characterize its potential clinical value in cancer patient cohort.

      Methods:
      NIFK is pointed out to possess RNA binding function via the RNA Recognition Motif (RRM). RNA immunoprecipitation assay was performed using specific NIFK antibody in lung cancer PC13 and A549 cells after NIFK overexpression. RNA targets were analyzed by RT-PCR and Next Generation Sequencing (NGS). In addition, relative NIFK, RUNX1 and CK1α expression and its association with patient survival outcome were investigated in lung cancer cohorts.

      Results:
      We observe the increase in amount of RNA binding targets after NIFK overexpression respectively in PC13 and A549 cells as comparing with the control. Moreover, NIFK appears to bind with RUNX1 RNA but not CK1α suggesting it is the initial step of NIFK-mediated TCF4/β-catenin signaling regulation. NIFK also leads to RUNX1 RNA instability of which mechanism remains to be explored. RUNX1 is observed to associate with good prognosis in lung cancer datasets (jacob-00182-CANDF and GSE13213). In addition, relative expression of the axis is further revealed in lung cancer cohort.

      Conclusions:
      We first show the interactive mechanism of pro-metastatic molecule NIFK in regulating TCF4/β-catenin signaling, which might contribute to therapeutic target design for lung cancer patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chang Gung Memorial Hospital

      Funding:
      Ministry of Science and Technology, Taiwan

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      14P - MMP-2 1306 C/T promoter polymorphism and smoking: A possible role as risk factors in lung cancer (Now Available) (ID 384)

      12:30 - 12:30  |  Presenting Author(s): A. Daniele  |  Author(s): D. Rosa, V. Lapadula, I. Abbate, D. Galetta, E.S. Montagna, A. Catino

      • Abstract
      • Slides

      Background:
      Lung cancer is the leading cause of cancer death in both gender accounting for 15% of deaths among young people (0–49 years), 30% among adults (50–69 years) and 27% among those over seventy. Matrix metalloproteases (MMPs) are a large family of proteins consisting of at least 26 human MMPs. They are zinc-dependent endopeptidases that cleave components of the extracellular matrix (ECM) and basement membrane and has been reported that play an important role in several steps of cancer development. Many studies have shown that a nucleotide polymorphism (–1306C3T) in the MMP2 promoter in addition to smoking and age are major risk factors for the onset of lung cancer. The aim of this study was to examine the association of MMP-2 polymorphism and smoke as a risk for cancer in 71 Italians lung cancer patients.

      Methods:
      Seventy-one patients (56 men and 15 women) aged between 44 and 84 years with adeno or squamous carcinoma in inoperable stage IIIA/IIIB/IV were enrolled in this study; information about clinical and histopathological data and smoking habits were collected through the clinical charts; genotyping was performed using direct DNA sequencing.

      Results:
      44/71 patients (62%) had the CC genotype, 15/71 (21%) patients had CT genotype and 12/71 (16%) had TT genotype. About smoking 47/71 (66%) patients were smokers, 5/71 (7%) patients had never smoked and 19/71 (26%) of them were ex-smokers and had stopped for at least 3 years. In subgroup of smokers patients, the overall average number of years of smoke was 44, the average number of smoked cigarettes was 25 and the number of pack- year 48; the smoker group was divided into heavy smokers smoking from 50 to 60 cigarettes/day, a group of heavy smokers smoking from 11 to 49 cigarettes/day and light smokers smoking from 2 to 10 cigarettes/day. A higher allelic frequency of CC genotype and stage IV disease in the heavy and mean smoking group were observed compared to light smokers (p < 0.001 and p < 0.0001 respectively) while the TT genotype was much more frequent in stage III and in subgroup of light smokers (p < 0.05).

      Conclusions:
      These preliminary results suggest a significant association between the mmp2-1306c/t polymorphism and smoke that could represent a significant risk factors of developing lung cancer.

      Clinical trial identification:


      Legal entity responsible for the study:
      IRCCS Istituto Tumori Giovanni Paolo II

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      15P - Comprehensive profiling of lincRNAs in lung adenocarcinoma of never smokers reveals their important roles in cancer development and prognosis (ID 362)

      12:30 - 12:30  |  Presenting Author(s): Y. Li  |  Author(s): X. Zhang, Z. Sun

      • Abstract

      Background:
      Long intergenic non-coding RNA (lincRNA) is a family of gene transcripts whose functions are largely unknown in disease process. Although cigarette smoking is the main cause of lung cancer, lung cancer in never smokers is a separate entity and its underlying cause is little known. Growing evidence suggests lincRNAs play a significant role in cancer development and progression; however, such data is lacking for lung cancer in never-smokers.

      Methods:
      This study conducted comprehensive profiling of lincRNAs from RNA-seq data in 27 pairs of lung adenocarcinoma and its normal tissues of never smokers. Both known and novel lincRNAs segregated tumors from normal tissues clearly. About a third of lincRNAs were differentially expressed between tumors and normal samples and most of them were coordinated with their putative protein gene targets. More interestingly, lincRNAs defined two clusters of tumors that were associated with tumor aggressiveness and patient survival.

      Results:
      We identified a subset of lincRNAs that were differentially expressed but also associated with patient survival. Almost perfect concordance was observed for differentially expressed lincRNAs and lincRNAs associated with survival from the discovery set were also predictive for survival in the validation set of 85 transcriptomes.

      Conclusions:
      With further validation, these lincRNAs could serve as potential diagnostic and prognostic markers.

      Clinical trial identification:


      Legal entity responsible for the study:
      Mayo Clinic

      Funding:
      The National Natural Science Foundation of China (No.81472835)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      16P - Long intergenic non-coding RNA 00152 promotes lung adenocarcinoma proliferation via interacting with EZH2 and repressing IL24 expression (ID 496)

      12:30 - 12:30  |  Presenting Author(s): C. Qinnan  |  Author(s): W. Zhaoxia

      • Abstract

      Background:
      Numerous studies have shown that long non-coding RNAs (lncRNAs) behave as a novel class of transcript during multiple cancer processes, such as cell proliferation, apoptosis, migration, and invasion. LINC00152 is located on chromosome 2p11.2, and has a transcript length of 828 nucleotides. The biological role of LINC00152 in LAD (lung adenocarcinoma) remains unknown.

      Methods:
      Quantitative reverse transcription PCR (qRT-PCR) was used to detect LINC00152 expression in 60 human LAD tissues and paired normal tissues. In vitro and in vivo studies showed the biological function of LINC00152 in tumour progression. RNA transcriptome sequencing technology was performed to identify the downstream suppressor IL24 (interleukin 24) which was further examined by qRT-PCR, western bolt and rescue experiments. RNA immunoprecipitation (RIP), RNA pulldown, and Chromatin immunoprecipitation (ChIP) assays were carried out to reveal the interaction between LINC00152, EZH2 and IL24.

      Results:
      LINC00152 expression was upregulated in 60 human LAD tissues and paired normal tissues. High levels of LINC00152 expression were correlated with advanced TNM stage, larger tumor size, and lymph node metastasis, as well as shorter survival time. Silencing of LINC00152 suppressed cell growth and induced cell apoptosis. LINC00152 knockdown altered the expression of many downstream genes, including IL24. LINC00152 could interact with EZH2 and inhibit IL24 transcription. Moreover, the ectopic expression of IL24 repressed cell proliferation and partly reversed LINC00152 overexpression-induced promotion of cell growth in LAD.

      Conclusions:
      Our study reveals an oncogenic role for LINC00152 in LAD tumorigenesis, suggesting that it could be used as a therapeutic target in LAD treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      The Second Affiliated Hospital of Nanjing Medical University

      Funding:
      National Natural Science Foundation of China (No. 81472198), the Key Clinical Medicine Technology Foundation of Jiangsu Province (No. BL2014096), the Medical Innovation Team Foundation of the Jiangsu Provincial Enhancement Health Project (No.21), “333 high class Talented Man Project” (No. BRA2016509)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      17P - Long non-coding RNA SNHG20 promotes non-small cell lung cancer cell progression by silencing of P21 expression (ID 336)

      12:30 - 12:30  |  Presenting Author(s): C. Zhenyao  |  Author(s): C. Xin, W. Zhaoxia

      • Abstract

      Background:
      Mounting evidence demonstrates that long non-coding RNAs (lncRNAs) are novel transcripts governing multiple biological processes, and that their dysregulation is involved in the development and progression of multiple types of cancer. Small Nucleolar RNA Host Gene 20 (SNHG20) is a 2183 bp lncRNA. The clinical relevance of SNHG20 and its molecular mechanisms involved in non-small cell lung cancer (NSCLC) have not been well documented.

      Methods:
      Expression of SNHG20 was analyzed in 42 paired NSCLC tissues and five NSCLC cell lines by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). Relationship between SNHG20 levels and NSCLC patients clinicopathologic feature was analyzed. Kaplan–Meier survival analysis was used to evaluate the prognosis of patients with high or low SNHG20 expression. Univariate and multivariate survival analysis were performed to further confirm the prognostic role of SNHG20. SNHG20 siRNAs and overexpression vector were transfected into NSCLC cells to knockdown or upregulate SNHG20 expression. In vitro and in vivo studies showed the biological role of SNHG20 in lung cancer cell proliferation and migration. RIP and ChIP assays were carried out to uncover the mechanism of SNHG20's regulation of underlying targets.

      Results:
      We found that SNHG20 expression was markedly upregulated in NSCLC tissues, and its upregulation was associated with larger tumor size (P = 0.012), lymph node invasion (P = 0.005) and TNM stage (P = 0.008). Kaplan–Meier survival analysis indicated that patients with low SNHG20 expression level had better progression-free survival (P = 0.003) and overall survival (P = 0.001) than those with high SNHG20 expression. Further univariate and multivariable Cox regression analysis suggested that increased SNHG20 was an independent prognostic indicator for this disease. Knockdown of SNHG20 repressed NSCLC cell proliferation, migration and induced cell apoptosis. Mechanistic investigations revealed that SNHG20 could interact with EZH2, thereby repressing P21 expression.

      Conclusions:
      Our findings indicate that SNHG20 is significantly upregulated in NSCLC tissues and it may represent a new candidate for use in NSCLC diagnosis, prognosis and therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      The Second Affiliated Hospital of Nanjing Medical University

      Funding:
      National Natural Science Foundation of China (No. 81472198,81672307); the Key Clinical Medicine Technology Foundation of Jiangsu Province (No. BL2014096); the Medical Innovation Team Foundation of the Jiangsu Provincial Enhancement Health Project (No.21); “333 high class Talented Man Project” (No. BRA2016509)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      18P - The pseudogene DUXAP10 promotes an aggressive phenotype through binding with LSD1 and repressing LATS2 and RRAD in non-small cell lung cancer (ID 497)

      12:30 - 12:30  |  Presenting Author(s): W. Chenchen  |  Author(s): W. Zhaoxia

      • Abstract

      Background:
      Pseudogenes have been considered as non-functional transcriptional relics of human genomic for long time. However, recent studies revealed that they play a plethora of roles in diverse physiological and pathological processes, especially in cancer, and many pseudogenes are transcribed into long noncoding RNAs and emerging as a novel class of lncRNAs. However, the biological roles and underlying mechanism of pseudogenes in the pathogenesis of non-small cell lung cancer are still incompletely elucidated.

      Methods:
      Real-time polymerase chain reaction (PCR) was used to examine DUXAP10 expression in NSCLC cell lines/tissues compared with normal epithelial cells/adjacent non-tumorous tissues. Cox proportional hazards regression models were performed to further confirm the prognostic role of DUXAP10. Cell proliferation assays were performed to detect the biological effects of DUXAP10 in gastric cancer cells. Real-time PCR, western-blot and immunohistochemistry were used to evaluate the mRNA and protein expression of LATS2 and RRAD.

      Results:
      This study identifies a putative oncogenic pseudogene DUXAP10 in NSCLC, which is located in 14q11.2 and 2398 nt in length. Firstly, we found that DUXAP10 was significantly up-regulated in 93 human NSCLC tissues and cell lines, and increased DUXAP10 was associated with patients’ poorer prognosis and short survival time. Furthermore, the loss and gain of functional studies including growth curves, migration, invasion assays and in vivo studies verify the oncogenic roles of DUXAP10 in NSCLC. Finally, the mechanistic experiments indicate that DUXAP10 could interact with Histone demethylase Lysine specific demethylase1 (LSD1) and repress tumor suppressors Large tumor suppressor 2 (LATS2) and Ras-related associated with diabetes (RRAD) transcription in NSCLC cells.

      Conclusions:
      These findings demonstrate DUXAP10 exerts the oncogenic roles through binding with LSD1 and epigenetic silencing LATS2 and RRAD expression. Our investigation reveals the novel roles of pseudogene in NSCLC, which may serve as new target for NSCLC diagnosis and therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      The Second Affiliated Hospital of Nanjing Medical University

      Funding:
      National Natural Science Foundation of China (No. 81472198), the Key Clinical Medicine Technology Foundation of Jiangsu Province (No BL2014096), the Medical Innovation Team Foundation of the Jiangsu Provincial Enhancement Health Project (No.21), “333 high class Talented Man Project” (No. BRA2016509)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      19P - Single sample predictor of non-small cell lung cancer histology based on gene expression analysis of archival tissue (Now Available) (ID 444)

      12:30 - 12:30  |  Presenting Author(s): A. Karlsson  |  Author(s): H. Cirenajwis, K. Ericson-Lindqvist, C. Reuterswärd, M. Jönsson, A. Patthey, A.F. Behndig, M. Johansson, M. Planck, J. Staaf

      • Abstract
      • Slides

      Background:
      In non-small cell lung cancer (NSCLC), histological classification dictates choice of patient therapy. In this study, we aimed to establish a gene expression based single-sample predictor (SSP) for histological classification of NSCLC tumors using archival tissue that may be used in parallel with e.g. gene fusion detection as a multicomponent single assay.

      Methods:
      A NanoString probe set was designed to target 12 genes routinely used as IHC markers for histological subtyping as well as fusion genes known to be frequently active in NSCLC. Gene expression data was derived from NanoString analysis of 78 formalin-fixed paraffin-embedded (FFPE) NSCLCs with known histological subtypes (development cohort). A SSP was trained using AIMS (1) in the development cohort for prediction of adenocarcinoma (AC), squamous cell carcinoma (SqCC), or neuroendocrine tumors (NE). The AIMS model was applied to 11 FFPE tumors classified as large cell carcinomas (LCC) according to the WHO2004 classification (2), and 199 fresh frozen NSCLC tumors analyzed by RNA sequencing (GSE81089)(3). Finally, the SSP will be applied to 11 NSCLC-not otherwise specified (NOS) cases (4) subjected to in-depth pathological re-evaluation.

      Results:
      The SSP was successfully applied to NanoString data from 11 LCCs re-classified as AC, SqCC and LCC according to the revised WHO2015 guidelines (2). Of reclassified LCC tumors, 100% of AC cases and 75% (3/4) of SqCC tumors were correctly identified. In GSE81089, the SSP was erratically successful depending on histology of the tumor classified, with 97.4% concordance for AC, 97.1% for SqCC, but mismatch for 3 out of 5 NE tumors. In summary, the SSP could successfully classify tumors of AC and SqCC histology in both validation cohorts but could less successfully classify non-AC and non-SqCC tumors respectively.

      Conclusions:
      Gene expression based SSP can accurately classify AC and SqCC histology. Expanded transcriptional profiling may be required to capture all aspects of lung cancer biology for precise and possibly refined histological subtyping of individual cases. Gene expression-based analysis could serve as a promising complement to existing techniques, providing a useful multicomponent assay for lung cancer diagnostics.

      Clinical trial identification:


      Legal entity responsible for the study:
      Lund University

      Funding:
      The Swedish Cancer Society, the Mrs Berta Kamprad Foundation, the Gunnar Nilsson Cancer Foundation, the Crafoord Foundation, BioCARE a Strategic Research Program at Lund University, the Gustav V:s Jubilee Foundation, Skane University Hospital Foundation, and governmental funding (ALF)

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      20P - ArgSS1 expression in squamous cell carcinoma of the lung (Now Available) (ID 294)

      12:30 - 12:30  |  Presenting Author(s): E. Cathcart-Rake  |  Author(s): A. Roden, A. Mansfield

      • Abstract
      • Slides

      Background:
      Diminished or absent expression of the enzyme argininosuccinate synthetase 1 (ArgSS1 or ASS1) portends chemotherapy resistance and poor prognosis in a number of tumor types, suggesting arginine depletion is a viable therapeutic strategy in ArgSS1 deficient tumors. ArgSS1 expression and its implications on prognosis have not been studied in squamous cell carcinoma of the lung.

      Methods:
      Resected pathology specimens were collected from patients with squamous cell carcinoma of the lung and analyzed for ArgSS1 expression by IHC. ArgSS1 expression was deemed intact if there was >50% expression in tumor cells and deficient if there was ≤50%. Chart reviews were conducted to abstract patient demographic and oncologic information. Pearson's correlations were utilized to evaluate potential associations between ArgSS1 status and oncologic variables. The Kaplan-Meier estimator was used to evaluate a potential association between ArgSS1 expression and survival which was compared with a log-rank test.

      Results:
      Sixty-seven specimens of squamous cell carcinoma of the lung were evaluated. Patients were diagnosed at a mean age of 66.9 years (SD 9.1). The majority of patients were male (64.2%). 97% were current or former smokers, and 89.6% had early stage disease. Most patients (64.2%) had moderately differentiated tumors. Nineteen (28.3%) tumors were ArgSS1 deficient. We did not find any significant associations between ArgSS1 expression and gender, tobacco use, stage at diagnosis, or tumor differentiation. There was no significant difference in overall survival between those who had intact ArgSS1 expression (median 42 months, interquartile range 28-not reached) and those who did not (median not reached, interquartile range 14-not reached; HR = 0.19; p = 0.66).

      Conclusions:
      Despite the associations of ArgSS1 deficiency and poor survival in other malignancies, we did not observe an association between ArgSS1 and overall survival in patients with squamous cell carcinoma of the lung. Our findings might be limited by our sample size. Even if ArgSS1 is not a prognostic marker in squamous cell carcinoma of the lung, it may still have predictive significance given the development of arginine deaminases in clinical trials. Preclinical work is ongoing with cell lines to study this further.

      Clinical trial identification:


      Legal entity responsible for the study:
      Mayo Clinic

      Funding:
      Has not received any funding

      Disclosure:
      A. Mansfield: Payments to his institution from AbbVie, Genentech, BMS and Trovagene for consulting, and payment to his institution from Novartis for research. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      21P - Influence of delayed and prolonged fixation on the evaluation of immunohistochemical staining on pulmonary resection specimen (Now Available) (ID 539)

      12:30 - 12:30  |  Presenting Author(s): M. Van Seijen  |  Author(s): L. Brcic, A. Navarro, I. Sansano, M. Béndek, B. Witte, I. Brcic, R. Kammler, R.A. Stahel, E. Thunnissen

      • Abstract
      • Slides

      Background:
      Pre-analytical factors as fixation time has influence on morphology of diagnostic and predictive immunohistochemical staining and are increasingly used in the evaluation of lung cancer. Our aim was to investigate if variations in fixation time influence the outcome of immunohistochemical staining in lung cancer.

      Methods:
      From lung cancer resections specimen with tumor size >4 cm, 10 fragments were sampled: 2 received standard fixation, 5 delayed, and 3 prolonged fixation. After paraffin embedding, tissue microarrays (TMA) were made. Immunohistochemistry (IHC) of 20 antibodies were applied and scored for quality and intensity of staining. TMA slides were scored by core/case: quality of cores and IHC intensity in cores of sufficient quality.

      Results:
      Tissue with delay in start of fixation showed deterioration of tissue quality leading to reduction in the evaluation of staining for CK-7, Keratin MNF116, Cam5.2, CK5/6, TTF-1, CMET, Napsin-A, D2-40, PD-L1 antibodies. Prolonged fixation had no influence on the performance of immunohistochemical stains.

      Conclusions:
      Delay of fixation negatively effects the evaluation of IHC staining. Therefore, tissue should be fixated soon after surgery.

      Clinical trial identification:


      Legal entity responsible for the study:
      European Thoracic Oncology Platform (ETOP)

      Funding:
      VU University Medical Center

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      22P - Clonal and subclonal occurrence of oncogenic mutations in lung adenocarcinoma (ID 642)

      12:30 - 12:30  |  Presenting Author(s): V. Tischler  |  Author(s): M.A. Ihle, A. Stenzinger, W. Weichert, W. Jochum, R. Büttner, J. Wolf, M. Peifer, R. Thomas

      • Abstract

      Background:
      Increasing evidence emerges that oncogenic mutations in cancer occur at both clonal and subclonal levels. Targeted drug treatment programs based on clinical mutation testing do not consider the clonal or subclonal frequencies of oncogenic mutations but the pure presence. Our aim was to study the clonal and subclonal frequencies of (oncogenic) mutations in lung adenocarcinoma (LUAD).

      Methods:
      Fresh frozen and formalin-fixed paraffin embedded tumor and normal tissues of 50 LUAD patients were investigated by WES. Computational analysis for accurate estimation of purity, ploidy and absolute copy numbers followed by assessment of the cancer cell fractions (CCF, ratio of the observed and the expected allelic fraction of a mutation) for each detected mutation in the individual adenocarcinoma was performed. Statistical distribution of CCFs was performed to assign mutations to distinct clonal and subclonal clusters. Clonal and subclonal mutational status was verified by digital droplet PCR of selected cases. Tumor mutational burden was calculated as exonic non-synonymous mutations per case. Mutational signatures were determined.

      Results:
      In the study cohort, 14 (28%) tumor samples were composed of clonal mutations and 36 (72%) of clonal and subclonal mutations. Of the 36 cases with subclonal clusters, 22 (61%) showed 1 subclonal cluster, 11 (31%) revealed 2 subclonal clusters and 3 (8%) 3 subclonal clusters. Among the subclonal mutations, we found known oncogenic mutations: KRAS c.34G > T (p.G12C), KRAS c.35G > T (G12V), KRAS c.182_183delinsTC (p.Q61L). The average number of tumor exonic non-synonymous mutations per case was 155 (median 103, ranging from 27 to 1593). The average exonic non-synonymous mutation rate was 5 per million base pairs. We found mutational signatures that are correlated with tobacco smoking, age, AID/APOBEC.

      Conclusions:
      We detected clonal and subclonal mutations in the majority of LUAD. We found oncogenic mutations in KRAS and EGFR. Our findings indicate the importance of the determination of clonal and subclonal status of oncogenic mutations and could also be meaningful for the estimation of tumor mutational burden as potential clinical marker.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Swiss Cancer League

      Disclosure:
      R. Thomas: Ownership interests (including patents) for AstraZeneca, Bayer, Novartis, and Roche; consultant/advisory board member for AstraZeneca, Bayer, Boehringer-Ingelheim, Clovis, Daiichi-Sankyo, Johnson & Johnson, Lilly, Merck, MSD, New Oncology, Puma, Roche, and Sanofi-Aventis. All other authors have declared no conflicts of interest.

    • +

      23P - Genomic profiling of gene aberrations in 323 Chinese NSCLC patients (Now Available) (ID 203)

      12:30 - 12:30  |  Presenting Author(s): Y. Liu  |  Author(s): C. Xu, W. Wang, Z. Song, Y. Zhu, Y. Guan, R. Chen, G. Chen, T. Lv, Y. Song

      • Abstract
      • Slides

      Background:
      Therapeutic approaches to non-small cell lung cancer (NSCLC) have shifted toward an emphasis on molecularly targeted therapy in genotypic subsets of patients such as EGFR, ALK, ROS1. Patients with driver mutations receiving matched target drugs could have significantly longer progression free and overall survival. In this study, we aimed to analyse the genomic alterations of NSCLC.

      Methods:
      Formalin-fixed paraffin-embedded tumor samples of 323 Chinese NSCLC patients including 193 males (59.75%) and 130 females (40.25%) with a median age of 58 were collected for next-generation sequencing (NGS)-based 59-genes panel assay. Genomic alterations including single base substitution, short and long insertions/deletions, copy number variations, and gene fusions in selected genes were assessed.

      Results:
      Different histological subtypes of adenocarcinoma (278/323, 86.07%), squamous carcinoma (32/323, 9.91%), mixed carcinoma (7/323, 2.17%) and large cell carcinoma (6/323, 1.86%) were included in the Chinese NSCLC cohort. The top ranked genomic alterations were TP53 (182/323, 56.35%), EGFR (133/323, 41.18%), MSH2(51/323, 15.79%), TSC2 (46/323, 14.24%), MSH6 (30/323, 9.29%), ALK fusions (28/323, 8.67%), MET (22/323, 6.81%), KRAS (22/323, 6.81%), BRAF (20/323, 6.19%), PIK3CA (18/323, 5.57%), HER2 (16/323, 4.95%), ROS1 fusions (9/323, 2.79%), and RET fusions (8/323, 2.48%), which makes up 90.40% of the 323 patients with at least one driver mutation. In addition to common driver mutations, rare mutation types such as HIP1-ALK, CEP72-ROS1, RAD18-ROS1 and FGFR3-TACC3 were also detected by deep sequencing assay.

      Conclusions:
      With the help of NGS, our study revealed the landscape of driver gene mutations in 323 Chinese NSCLC patients, and we also found the most target locations that might be treated by targeted therapies. Further studies may emerge whether concurrent mutations, mutation burden and the number of actionable mutation are associated with survival outcome in NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Yueping Liu

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      24P - Proviral integration site for Moloney murine leukemia virus-1 (PIM-1) inhibition with AZD1208 to prevent resistance to osimertinib in EGFR mutant NSCLC (Now Available) (ID 399)

      12:30 - 12:30  |  Presenting Author(s): J. Bracht  |  Author(s): N. Karachaliou, J. Berenguer, I. Attili, M. Ito, C. Codony-Servat, J. Codony-Servat, M. Filipska, N. Gil, R. Rosell

      • Abstract
      • Slides

      Background:
      Although treatment with EGFR tyrosine kinase inhibitors (TKIs) is initially effective in a subgroup of lung cancer patients, therapy resistance ultimately occurs. Resistance mechanisms consist of overexpression and co-expression of receptor tyrosine kinases (RTKs) and overexpression of STAT3. PIM1 was shown to be an important regulator of both RTKs and STAT3 expression, and inhibition of PIM1 with the pan-PIM inhibitor, AZD1208, might prevent STAT3 and RTK up-regulation after TKI treatment. This research focuses on combining TKI treatment with AZD1208 to prevent therapy resistance in different EGFR mutant NSCLC cell lines.

      Methods:
      Using TaqMan quantitative-PCR we studied basal mRNA expression levels of PIM1 and PIM3 in 5 EGFR-mutant NSCLC cell lines. Cells were then exposed to single osimertinib or AZD1208 treatment, or the combination, to determine the combination index (CI) and changes in cell viability. Moreover, changes in protein expression of different RTKs, STAT3 and downstream effectors of STAT3 were studied in 2 cell lines using western blotting.

      Results:
      The PC9 and H1975 cell lines were shown to have high PIM1 and STAT3 expression compared to the other EGFR mutant NSCLC cell lines. Combined osimertinib and AZD1208 treatment showed moderate synergism in all cell lines, with CIs ranging from 0.75 to 0.86 in triplicate experiments. Western blot experiments indicate that osimertinib treatment leads to upregulation of pCDCP1, pYAP1, pPaxillin and pSTAT3 in the PC9 and H1975 cell lines, suggesting initiation of resistance to single osimertinib treatment. In contrast, single AZD1208 treatment does not induce, or even lower expression of these proteins compared to baseline levels. When combining both treatments, the osimertinib-induced pSTAT3 up-regulation can be prevented with AZD1208. The effect of AZD1208 on RTK expression will be further explored.

      Conclusions:
      Single TKI treatment in EGFR mutant NSCLC induces expression of RTKs and STAT3, ultimately leading to therapy resistance. Inhibition of PIM1 with AZD1208 can abolish the osimertinib-induced phosphorylation of STAT3, and thereby prevents activation of resistance pathways.

      Clinical trial identification:


      Legal entity responsible for the study:
      IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain

      Funding:
      Fundació Obra Social “La Caixa”

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      25P - A non-pathway-specific approach in EGFR and KRAS mutant or squamous cell histology non-small cell lung cancer (NSCLC) (ID 551)

      12:30 - 12:30  |  Presenting Author(s): M. Ito  |  Author(s): C. Codony-Servat, J. Codony-Servat, I. Attili, J. Berenguer, J. Bracht, N. Gil, M. Okada, N. Karachaliou, R. Rosell

      • Abstract

      Background:
      p21-activated kinase 1 (PAK1) stimulates growth and metastasis in several types of tumors, including NSCLC. Protein kinase C iota (PKCi) is an enzyme highly expressed in NSCLC that regulates PAK1 signaling. We have previously shown that cancer pathway-specific intervention, like EGFR inhibitors in EGFR mutant NSCLC, results in parallel compensatory activation of other pathways, including the receptor tyrosine kinases AXL and MET, the transmembrane protein CUB domain-containing protein-1 (CDCP1) or the transcriptional regulators STAT3 and YAP1. We have now explored whether a non-pathway-specific approach can be efficient in three subsets of NSCLC.

      Methods:
      Three lung cancer cell lines were used: HCC827 and H23 lung adenocarcinoma cells that carry EGFR and KRAS mutations respectively, and H520 PAK1 amplified squamous NSCLC cells. Cell viability assays and western blotting were applied to evaluate the effect of IPA-3 (PAK1 inhibitor) plus auranofin (PKCi inhibitor). We used a Chou-Talalay modified method for drug combination studies that offers quantitative definition for additive effect (combination index [CI] = 1), synergism (CI < 1), and antagonism (CI > 1).

      Results:
      We found a differential PAK1 expression or activation profile in the three models, with H520 cells being the ones with the highest PAK1 expression and activation. IPA-3 plus auranofin was highly synergistic in HCC827, H23 and H520 cells with CIs of less than 0.4. In the EGFR mutant HCC827 cell line, IPA-3 plus afatinib or osimertinib was additive (CI = 0.9), or slightly synergistic (CI = 0.8). In the same cell line, the combination of IPA-3 plus auranofin abrogated EGFR and downstream signaling (ERK, AKT, STAT3, YAP1) and inhibited the expression and activation of AXL, MET and CDCP1. IPA-3 plus auranofin was, similarly, highly synergistic in H23 (CI = 0.3) and H520 (CI = 0.3) cells.

      Conclusions:
      For the first time, we are reporting that a non-pathway specific combination can be effective in EGFR and KRAS mutant as well as squamous NSCLC. The combination could control the counter-regulatory pathways that are made apparent and ultimately cause resistance when a pathway specific-intervention is applied.

      Clinical trial identification:


      Legal entity responsible for the study:
      IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain

      Funding:
      International Association for the Study of Lung Cancer (IASLC)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      26P - Incidence of T790M mutations after progression on first Line EGFR TKIS in advanced NSCLC: Real time data from a tertiary cancer institute in North India (ID 422)

      12:30 - 12:30  |  Presenting Author(s): U. Batra  |  Author(s): P. Jain, P. Babu, M. Sharma

      • Abstract

      Background:
      The incidence of EGFR mutations vary according to geographic region of the world. The EGFR rates in the western world are 15% whereas the incidence of EGFR mutation in the EASTERN population is 45–55%. In India, the incidence of EGFR mutation is approx. 33%. Similarly, there is a paucity of data regarding the incidence of T790 M mutation following EGFR TKIs from the Indian subcontinent. This study was therefore undertaken to analyse the incidence of T790M mutation in the Indian population following therapy with EGFFR TKIs.

      Methods:
      EGFR mutant NSCLC stage IV patients who received Tyrosine kinase inhibitors and progressed on TKI were analysed. T790M status was determined using the droplet digital PCR.

      Results:
      41 patients were selected using above selection criteria. T790M mutation was detected in 22 of 41 patients (54%). 21 of 22 patients had retained the primary EGFR mutation also. Out of these 22 patients, del19 was present in 14 patients and 8 patients had L858R mutation. Most common site of progression was pleural and plural effusion. Median duration of treatment on TKI before progression is 289.7 days, highest duration being 1290 days and lowest 45 days.

      Conclusions:
      Exact incidence of T790M mutations after progression on TKI s in Asian population is not exactly known and requires large data, as incidence may be different than reported in western population.

      Clinical trial identification:
      none

      Legal entity responsible for the study:
      All authors

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      27P - ALK immunohistochemistry scores do not predict sensitivity to crizotinib in fluorescence in situ hybridization-positive non-small cell lung cancer patients (Now Available) (ID 512)

      12:30 - 12:30  |  Presenting Author(s): G. Metro  |  Author(s): R. Chiari, D. Giannarelli, A. Sidoni, G. Bellezza

      • Abstract
      • Slides

      Background:
      Anaplastic lymphoma kinase (ALK) immunohistochemistry (IHC) is an established screening method for the detection of ALK gene rearrangements by fluorescence in situ hybridization (FISH) in patients with advanced non-small cell lung cancer. However, little is known about the correlation between ALK IHC scores and sensitivity to treatment with an ALK-tyrosine kinase (-TKI) inhibitor.

      Methods:
      ALK IHC score was evaluated through a 4-tiered system (0, 1+, 2+, 3+) based on the D5F3 clone (Cell Signaling). Any number of cells stained was considered as ALK positive, and the strongest staining present was the final score. H-score was assessed using the following formula: [1 × (% cells 1+) + 2 × (% cells 2+) + 3 × (% cells 3+)] resulting into a score from 0 to 300. All ALK IHC score positive tumors underwent confirmatory FISH (Vysis ALK Break Apart Probe kit).

      Results:
      From June 2011 to May 2017, 50 double ALK IHC/FISH-positive patients were identified at our Institution. The majority of patients had an ALK IHC score ≥2+ [5/50 (10%) score 1+; 23/50 (46%) score 2+; 22/50 (44%) score 3+], with 47/50 (94%) tumors having any ALK IHC positivity in ≥50% of cells. H-score was more commonly ≥100 [2/50 (4%) 1-<100; 14/50 (28%) ≥100-<200; 34/50 (68%) ≥200–300]. Overall, 31 patients were treated with the ALK-TKI crizotinib for advanced disease, whose main charachteristics were as follows: median age 50 years (28–80), male/female (42%/58%), never/ever smokers (61%/39%), performance status 0/1 (74%/26%), chemotherapy-pretreated (90%). The results showed no significant correlation between the intensity of ALK IHC score or H-score with regard to response to crizotinib. Similarly, no significant association was noted between the intensity of ALK IHC score as well as the H-score and progression-free survival.

      Conclusions:
      In double ALK IHC/FISH-positive patients ALK IHC scores are typically skewed towards higher values, and ALK protein is expressed in a mostly diffuse way. Such preferentially higher distribution of ALK IHC scores as well as diffuse ALK staining might imply difficulty in setting a discriminatory treshold of activity with regard to sensitivity to crizotinib. This, in turn, could justify the absence of predictivity of ALK IHC scores.

      Clinical trial identification:


      Legal entity responsible for the study:
      Giulio Metro

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      28P - The study of ALK rearrangement in advanced primary non-small cell lung cancer and associated metastatic lesions (Now Available) (ID 201)

      12:30 - 12:30  |  Presenting Author(s): C. Xu  |  Author(s): W. Wang, W. Zhuang, Y. Tian, J. Zhang, M. Fang, Y. Chen, G. Chen, T. Lv, Y. Song

      • Abstract
      • Slides

      Background:
      ALK rearrangement in non-small cell lung cancer (NSCLC) patients has recently been identified as a driver gene and benefited from crizotinib treatment. However, no data are available for ALK rearrangement NSCLC about the relationship between primary and metastatic patients. The aim of this study was to examine the positive rate of ALK rearrangement in primary and metastatic NSCLC, and to investigate their relationships.

      Methods:
      From January 2013 to May 2015, 384 cases of primary NSCLC, 246 cases of matched metastatic tumors, and 47 cases of normal lung specimens, as the control group, were collected in our multicenter. The positive rate of ALK rearrangement among the NSCLC population was established, and thus the consistency of ALK rearrangement in advanced primary NSCLC and associated metastases and the relationship between ALK rearrangement and clinical data was analyzed.

      Results:
      The positive rate of ALK rearrangement on primary tumor was 11.46% (44/384). For those 246 paired cases, the positive rate on primary tumor was 10.98% (27/246), with that of metastases 7.32% (18/246). Among the 246 cases, there were two cases whose metastases were positive but primary tumors were negative and 11 case whose primary tumors were positive but metastases were negative. Positive rate of ALK rearrangement was higher in the primary lesions than in metastases. It was of statistical significance between the two groups (χ[2] = 112.208, P < 0.001). The positive rate of primary tumors could be predicted by metastases (κ = 0.683, P < 0.001). The sensitivity was 59.26% (16/27) and the specificity was 99.09% (217/219).

      Conclusions:
      The metastases of NSCLC can predict ALK rearrangement of the primary lesions. It can be used as an alternative means for metastases to detect ALK rearrangement which are not readily available.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chunwei Xu

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      29P - Preliminary assessment of a targeted break-point NGS assay for ALK gene fusion detection in lung adenocarcinoma samples from Chilean patients (ID 241)

      12:30 - 12:30  |  Presenting Author(s): M. Freire  |  Author(s): R. Lizana, L. Ramos, G. Sepúlveda, A. Blanco, S. Chernilo, O. Arén, C. Fernández, R. Armisén

      • Abstract

      Background:
      Lung cancer is the leading cause of cancer death in women and men. The most common mutations in lung adenocarcinoma are in EGFR and KRAS, and along with ten other genes (e.g. ALK and MET) they show a prevalence of 5% or less. As many of these mutations are clinically actionable, it is essential to know the mutation state for all of these genes. Therefore, it is necessary to evaluate multiple types of mutations (SNP, fusions, and CNV) of numerous genes in a small amount of FFPE tissue. We performed a preliminary orthogonal validation of the next generation sequencing based Oncomine Focus Assay (OFA, Thermo Fisher) for the determination of ALK gene fusion in lung adenocarcinoma samples from Chilean patients under standard clinical settings.

      Methods:
      This work analyzed 722 lung adenocarcinoma samples from patients recruited in the NIRVANA study by an immunohistochemistry-based ALK test (Ventana) and OFA (including an ALK gene fusion breakpoint assay). Twenty-eight ALK-positive cases by Ventana ALK, OFA or both and 22 negative samples for both tests, were analyzed using a validated (in EU and China) qPCR EML4-ALK fusion gene detection kit (AmoyDx).

      Results:
      Using the EML4-ALK qPCR fusion kit as benchmark, both Ventana ALK and OFA have a sensitivity of 75% [CI95%:51–91]. However, OFA presents a 96% [CI95%: 80–>99] specificity vs. 88% [CI95%:69–97] for Ventana ALK. Therefore, the positive predictive value is 94% [CI95%:68–99] for OFA and 83% [CI95%:63–94] for Ventana ALK. The most commons ALK gene fusions were exon 20 and 6 of EML4.

      Conclusions:
      When these techniques were compared using “real world” lung adenocarcinoma samples, OFA presented an advantage in the ability to predict positive values and was more specific than Ventana ALK test. These results indicate the need for further research to validate the use of OFA panel for the determination of these gene rearrangements.

      Clinical trial identification:
      NIRVANA study NCT03220230

      Legal entity responsible for the study:
      Pfizer Chile

      Funding:
      Pfizer

      Disclosure:
      M. Freire, R. Lizana, L. Ramos: Currently Pfizer employee and directly involved in this research initiative. G. Sepúlveda, A. Blanco: Currently Pfizer employee and directly involved in this research initiative, through the data analysis. S. Chernilo, O. Arén: Principal Investigator in one of the centers that recruit subject to this study; involved in the results discussion and review; Pfizer funding to research center for the subjects recruitment. C. Fernández: Collaborates with one of Principal Investigator and also gives Anatomopathological support to this study; Involved in the results discussion and review; Pfizer funding to anatomopathological team for the samples analyzed. R. Armisén: Currently Pfizer employee, head of research at the center where this research was done; involved in the results discussion and review.

    • +

      30P - Feasibility of anti-ROS1 SP384 for detection of ROS1 protein (Now Available) (ID 433)

      12:30 - 12:30  |  Presenting Author(s): I. Menzl  |  Author(s): B. Richardson, C. Le, D. Smith, A.E. Hanlon Newell, C. Bell, G. Pate, R.S.P. Huang

      • Abstract
      • Slides

      Background:
      Determination of ROS1 positivity in non-small cell lung cancer (NSCLC) is essential for appropriate identification of patients who may respond to ROS tyrosine kinase inhibitor therapy. ROS1 positivity is often clinically detected by fluorescence in situ hybridization (FISH). A ROS1 antibody that can detect ROS1 protein with high sensitivity, specificity, and consistency would allow for immunohistochemistry (IHC) screening for ROS1 positivity. Here, we present data on the feasibility of using VENTANA ROS1 (SP384) Rabbit Monoclonal Primary Antibody (anti-ROS1 SP384) for the detection of ROS1 protein.

      Methods:
      Anti-ROS1 SP384 has been optimized with VENTANA OptiView DAB IHC Detection Kit and on BenchMark IHC/ISH instruments. Anti-ROS1 SP384 was evaluated for sensitivity, specificity, and consistency of staining for the ROS1 protein. Sensitivity and specificity were assessed through immunoreactivity using tissue microarrays (TMAs) containing NSCLC (n = 40), cholangiocarcinoma (n = 64), melanoma (n = 90), 20 non-neoplastic tissue types, and additional 15 neoplastic tissues. The following parameters were assessed: percent target cell staining, overall intensity of target cell staining on a scale of 0–3 in 0.25 increments, and reactivity which is deemed as sample with >0% ROS1 staining. Consistency of anti-ROS1 SP384 staining was assessed on BenchMark GX, XT and ULTRA platforms and for 3 different antibody lots.

      Results:
      Anti-ROS1 SP384 exhibited high sensitivity and specificity in when staining TMAs containing a range of neoplastic and non-neoplastic tissues and when comparing the results to prevalence of ROS1 expression cited in the literature. The antibody showed expected expression of ROS1 in NSCLC, cholangiocarcinoma, melanoma, normal kidney tubules, and type II pneumocytes. The remaining cases included on the TMA were largely non-reactive. Staining performance of anti-ROS1 SP384 was consistent across 3 platforms and 3 antibody lots.

      Conclusions:
      Herein, we demonstrate that it is feasible to use anti-ROS1 SP384 to detect ROS1 protein due to the high sensitivity, specificity, and consistency of the antibody. Further studies are ongoing in previously characterized patient samples to assess antibody performance, clinical utility, and staining interpretation guidance.

      Clinical trial identification:


      Legal entity responsible for the study:
      Ventana Medical Systems, Inc.

      Funding:
      Ventana Medical Systems, Inc.

      Disclosure:
      I. Menzl, B. Richardson, C. Le, D. Smith, A.E. Hanlon Newell, G. Pate, R.S.P. Huang: Employee of Roche/Ventana Medical Systems and owns stock in Roche. C. Bell: Employee of Roche/Ventana Medical Systems.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      31P - Analysis of ROS1 rearrangement non-small cell lung cancer cell blocks from pleural effusion (Now Available) (ID 202)

      12:30 - 12:30  |  Presenting Author(s): W. Wang  |  Author(s): C. Xu, W. Zhuang, Y. Tian, J. Xu, M. Fang, Y. Chen, G. Chen, T. Lv, Y. Song

      • Abstract
      • Slides

      Background:
      ROS1 rearrangement in non-small cell lung cancer (NSCLC) patients has recently been identified as a driver gene event and patients benefi from crizotinib treatment. The aim of this study was to investigate the clinical value of ROS1 rearrangement non-small cell lung cancer (NSCLC) cell blocks from pleural effusion.

      Methods:
      Two hundred and fifteen cases of ROS1 rearrangement non-small cell lung cancer (NSCLC) blocks cell from pleural effusion, and 404 cases of tissues were analysed by the reverse transcription polymerase chain reaction (RT-PCR) method. The consistency of ROS1 rearrangement was examined in 74 cases of patients with tissues and cell blocks.

      Results:
      ROS1 rearrangement was found in 7 of 215 cell blocks (positive detection rate of 3.26%). ROS1 rearrangement was detected in 8 of 404 tissue blocks (positive detection rate of 1.98%). There were 71 cases of the 74 (95.95%) cases that had the same consistency as tissue block. ROS1 rearrangement was detected in 2 of 74 (2.70%) cell blocks, and 5 of 74 (6.76%) tissue blocks.

      Conclusions:
      The rate of ROS1 rearrangement in cell blocks of NSCLC is higher than in matched tissue blocks. The patients with malignant pleural effusion are likely to tend to ROS1 rearrangement.

      Clinical trial identification:


      Legal entity responsible for the study:
      Wenxian Wang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      32P - Serum circulating cell free DNA as potential diagnostic and prognostic biomarker in non-small cell lung cancer (Now Available) (ID 262)

      12:30 - 12:30  |  Presenting Author(s): A. Alhanafy  |  Author(s): S. El Shafei, M. Habib, R. Abdellatif, M. Haggag

      • Abstract
      • Slides

      Background:
      Cell-free DNA (ccf-DNA) can be found in healthy persons, persons with malignant and nonmalignant diseases as Chronic Obstructive Pulmonary Disease. In cancer, ccf-DNA results from tumor necrosis, lysis of circulating cancer cells or of micro-metastases and active release. Recently the first liquid biopsy test for analysis of driver gene mutation from cfDNA becomes a reality in clinical practice for patients with NSCLC with use of peripheral blood sample for EGFR mutation detection as predictive marker of response to EGFR-targeted therapy. The aim of this study was to study the concentrations and integrity index of ccf-DNA as a diagnostic and prognostic marker.

      Methods:
      This study was carried out in Menoufia University hospital in the period from October 2015 to September 2017. It was conducted on 140 individuals classified into: Group I: included 60 patients with NSCLC, Group II: included 40 patients with COPD. Group III: included 40 healthy controls. For NSCLC patients Staging was done according to the American Joint Committee on Cancer: the 7th edition. Real-time ALU-qPCR used to assess the concentration and integrity index of serum ccf-DNA.

      Results:
      The mean age was (56.8 ± 9.8, 52.9 ± 11.1 & 53.7 ± 9.0) for the three studied groups respectively with non significant difference between them. NSCLC patients demonstrated significantly higher values of each of ALU 215, ALU 247, DNA integrity than both COPD patients and controls, with non significant difference between the last two groups. The ROC curve analysis demonstrated that the total accuracy of ALU 247 was 92.1% at a cutoff point 325, 0.98 AUC, 96.7% sensitivity, 88.7% specificity, 86.6% PPV& 97.3% NPV, while ALU 115 recorded (0.93, 90%,78.7%, 76.1, 91.3 & 83.6) for AUC, sensitivity, specificity, PPV, NPV and accuracy respectively at a cutoff point 565. It was noticed that DNA integrity has AUC 0.65, sensitivity 75%, specificity 42.5%, 49.5% PPV, 69.4% NPP and total accuracy 56.4% at a cutoff point 0.48. Among NSCLC patients, ALU 115 & ALU 247 increased significantly in advanced tumor and more elevation was noticed in metastatic patients. Overall survival of NSCLC patients in relation to ALU 247, ALU 115 & DNA integrity documented a significant relationship between low DNA integrity and better survival of those patients.

      Conclusions:
      Serum DNA concentrations may be valuable in early diagnosis and potential prognostic marker in NSCLC patients

      Clinical trial identification:


      Legal entity responsible for the study:
      Menoufia University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      33P - Expression of biomarkers IDH1, CEA, TPA and CYFRA21-1 in peripheral blood and tissue of non-small cell lung carcinoma patients detected by real-time PCR (Now Available) (ID 467)

      12:30 - 12:30  |  Presenting Author(s): A. Mishra  |  Author(s): N. Singh, D.K. Sahu, S. Kumar

      • Abstract
      • Slides

      Background:
      Lung cancer is the leading cause of cancer-related death worldwide. In India, lung cancer constitutes 6.9 per cent of all new cancer cases and 9.3 per cent of all cancer related deaths in both sexes. The propensity for tumor biomarkers to be detected in serum at an early disease stage has become an area of interest for clinicians. This study aimed to evaluate the expression of four known serum biomarkers, namely, Isocitrate Dehydrogenase-1(IDH-1), carcinoembryonic antigen(CEA), Tissue Polypeptide Antigen (TPA), cytokeratin19(CYFRA-21-1) in tumor tissue and peripheral blood of patients with non-small cell lung cancer, detected by relative quantification real-time PCR and to analyze the association with benign lung tissue samples.

      Methods:
      Fifty patients affected by non small cell lung carcinoma of Stage IIIB, in which histopathologically identified 21 patients were adenocarcinoma and 29 patients were squamous lung carcinoma and fifty individuals which affected by benign lung disease were studied in this study. cDNA were made from each sample after taking peripheral blood samples and tissue sample during surgery, extracting total RNA. simultaneously detecting four genes in each sample was examined by the real-time PCR technique.

      Results:
      The IDH1 was up-regulated in tumor patients compared with corresponding benign lung individuals in tissue samples 36 out of 50 and in blood sample was up-regulated in 40/50, its sensitivity was determined at 72% in tumor tissue sample and 80% in blood sample, CEA was up-regulated 34 out of 50 patients in tissue and in blood sample up-regulated in 41/50, its sensitivity was determined at 68% in tumor tissue sample and 82% in blood sample, TPA was up-regulated 43 out of 50 patients in tissue and in blood sample up-regulated in 45/50, its sensitivity was determined at 86% in tumor tissue sample and 90% in blood sample, CYFRA21-1 was up-regulated 37 out of 50 patients in tissue and in blood sample was up-regulated in 43/50, its sensitivity was determined at 74% in tumor tissue sample and 86% in blood sample.

      Conclusions:
      Hence, we further validate the known biomarkers both in tissue and peripheral blood and propose their diagnostic and prognostic utility.

      Clinical trial identification:


      Legal entity responsible for the study:
      Institutional Ethical Committee

      Funding:
      King George's Medical University, Lucknow, India

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      34P - Recommendations for EGFR mutation testing with a fast and fully automated sample-to-result system: How to optimize DNA input instead of using lung cancer tissue? (Now Available) (ID 563)

      12:30 - 12:30  |  Presenting Author(s): C. Chapusot

      • Abstract
      • Slides

      Background:
      EGFR mutation detection on non-small cell lung carcinoma (NSCLC) is mandatory for optimal care of metastatic patients. Rearrangement/amplification of ALK or ROS, PDL1 expression level, BRAF, HER2 mutational status is also required. These different tests are time and tissue consuming. Therefore, molecular diagnostic laboratories need to elaborate strategies in order to optimize the realization of these different tests from limited biopsy specimens. The Idylla™ EGFR Mutation Test provided by Biocartis® is a fully automated sample–to–result test based on real-time PCR. The test allows the detection of 51 EGFR mutations directly from slices of FFPE tissue samples in less than three hours with an overall agreement of 96% with pyrosequencing. Nevertheless, further molecular tests need DNA and further tissue processing. In this project, the feasibility of EGFR testing from extracted DNA of NSCLC samples using Idylla™ EGFR was evaluated.

      Methods:
      EGFR mutational status was evaluated with the Idylla™ EGFR cartridge in two independent series of 20 retrospective and 83 prospective cases using DNA samples extracted from NSCLC instead of tissue sections.

      Results:
      Tumoral cellularity, DNA quantity and quality evaluated with a Tapestation Agilent® were taken into account in addition to the WT DNA CQ value calculated by the Idylla™ Explore software to optimize the DNA input. EGFR mutational status of the 20 retrospective cases previously analyzed with pyrosequencing was assessed in order to check the agreement of the fully automated method. A prospective series of 83 samples was then analyzed. All the results were interpreted and validated with the use of the Idylla™ test specific software, and qPCR curves were visualized by the Idylla™ Explore software.

      Conclusions:
      These results suggest some recommendations to use already extracted DNA with a fully automated sample–to–result system taking into account preanalytics and analytics features to safely characterize EGFR mutational status on lung cancer samples.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital of Dijon

      Funding:
      Biocartis company

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      37P - Epidemiology and management of bronchopulmonary carcinoma in eastern Morocco of 738 cases (Now Available) (ID 217)

      12:30 - 12:30  |  Presenting Author(s): I. Alloubi  |  Author(s): K.Y. Belmokhtar, M. Bellaoui

      • Abstract
      • Slides

      Background:
      In Morocco, bronchopulmonary cancer (CBP) is a major healthcare problem whose main etiological factor is tobacco. However, in the Eastern Kingdom region no study has been done on the epidemiological characteristics, pathological, clinical, molecular and therapeutic aspects of CBP.

      Methods:
      This is a descriptive retrospective study aimed at determine the epidemiological, pathological, clinical and therapy of CBP of patients at the Regional Oncology Center (ROC) Hassan II Oujda.

      Results:
      The study population consisted of 738 patients, 671 men and 67 women. The mean age was 59.1 ± 11.9 years (median = 58 years). Smokers are mostly men and represent 87% of patients. Average smoking consumption was 37.58 ± 20.85-pack years with an average duration of 30.92 ± 11.18 years. The symptoms that prompted the consultation were: cough in 49.59% of cases, chest pain in 45.39% of cases, hemoptysis in 32.11% of cases, dyspnea in 30.89% of cases. Adenocarcinoma was the predominant histological type (47%) followed by epidermoid carcinomas (27%) and small cell carcinomas (13%). Involvement of the right lung was frequent (55%), 36% of the lesions involved the lobe upper right. Almost all patients (97%) were diagnosed in advanced stages (III and IV). In this series, 65% of patients were treated with chemotherapy, 31% with radiotherapy and surgical treatment was only done in 6% of cases.

      Conclusions:
      This study reports, for the first time, the epidemiological data, pathology, clinical characteristics and therapy of CBP in the Eastern Region of Morocco for a period of 10 years (2005–2014). It was found that the diagnosis is often made at a late stage of the disease, hence the need for early detection and prevention based on the fight against tobacco.

      Clinical trial identification:


      Legal entity responsible for the study:
      Genetic Department, Faculty of Medicine Oujda Morocco

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      38P - Survival of lung cancer: Bangladesh perspective (Now Available) (ID 239)

      12:30 - 12:30  |  Presenting Author(s): R. Parveen  |  Author(s): P.S. Akhtar, N. Khatun, M.J. Islam

      • Abstract
      • Slides

      Background:
      Lung cancer, the most common and the leading cause of cancer death in the world, is increasingly being recognized in Bangladesh. Comprising one third of all male cancers, it is mostly presented at later stages. This study aimed at finding out the survival of lung cancer patients and also the important prognostic factors behind survival who were registered and treated in National Institute of cancer Research & Hospital.

      Methods:
      This study was done from Jan 2012 to June 2015 at Dept. of Medical Oncology, NICRH. The lung cancer patients attending NICRH in two years (from Jan 2012 to Dec 2013) were selected. After thorough evaluation, symptomatic and supportive management as well as chemotherapy and/or radiotherapy were given on priority basis. The patients were followed up every 4–8 weekly up to survival or maximum 42 months whichever was earlier. The patients were assessed during each visit by clinical status, investigations and treatment response. All events of death were documented properly.

      Results:
      Out of 2264 lung patients who were registered and treated during the period of Jan'12 –Dec'13, 1067 were in contact and eligible for the study. The male female ratio was 4.2:1 (862 male and 205 female). Female patients were younger (mean age of male 58.24 years and female 52.18yrs), 80% of all cases belonged to poor and lower middle-class families with 54% illiterate and 26% had primary education. Their performance status was mostly ECOG grade 2 & 3 (>80%). Male patients were smokers (90%) and females were mostly betel nut and/or tobacco chewers (66%) and also smokers (27%). All cases were at stage III and stage IV diseases with 86% non-small cell carcinoma and 14% small cell carcinoma. About 50% patients survived six months. One year survival of all cases was 27%; female 32% and male 25%. Mean survival of male and female patients was respectively 7.82 and 16.63 months. Survival of female patients was significant (P value-0.001) and survival of younger patients (<40 years) were also significant, 10.06 months (P value-0.001).

      Survival in months according to gender and age
      GenderMean Survival Time (months)95% confidence intervalP-value
      Male7.8207.300–8.3410.001 (HS)
      Female16.63814.153–19.124
      Overall8.7808.237–9.322
      Age group
      <=4010.0637.723–12.4030.001 (HS)
      41–509.6308.369–10.891
      51–609.4818.527–10.435
      61–707.6426.736–8.547
      >706.1714.944–7.399
      Overall8.7808.237–9.322


      Conclusions:
      Lung cancer patients mostly presented at advanced stage (inoperable), with poor performance status. After supportive, symptomatic and anticancer treatment (chemotherapy and radiotherapy), one year survival of lung cancer was 27%. Female and young age <40 years patients’ survival was significantly higher than male and aged patients (>40yrs).

      Clinical trial identification:


      Legal entity responsible for the study:
      Prof. Parveen Shahida Akhtar

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      39P - Lung cancer in Morocco: Results of a retrospective study (ID 516)

      12:30 - 12:30  |  Presenting Author(s): A. Haimer  |  Author(s): F. Habib, A. Soulaymani, A. Mokhtari, H. Hami

      • Abstract

      Background:
      Lung cancer is a major cause of morbidity and mortality worldwide, with an estimated 1.8 million new cases of lung cancer (13% of the total) and 1.6 million cancer deaths in 2012 (19.4% of the total) (GLOBOCAN 2012). The aim of this study is to determine the epidemiological characteristics of lung cancer in Morocco.

      Methods:
      This is a descriptive retrospective study of lung cancer cases diagnosed and treated at Al Azhar Oncology Center in Rabat between 2005 and 2015.

      Results:
      During the period of study, there were 615 cases diagnosed with lung cancer; 85.9% in men and 14.1% in women, giving a male-female ratio of 6.1 and representing 7.7% of all new cases of cancer reported during this period. The average age of the patients at diagnosis was 59.6 ± 11.4 years (range 4–94 years). Lung cancer is related to age with only 2.6% of cases diagnosed in persons younger than 40 years, 86.4% in those aged 40–74 years and 11% in those aged 75 years and over. Among all detected cases, 2.4% were diagnosed with metastatic disease and 21% died from lung cancer during the study period, accounting for 25.9% of all cancer deaths.

      Conclusions:
      Despite the limitations of the available data, it is clear that there are several barriers to access to cancer control in developing countries. This includes prevention, early detection, diagnosis and treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Ibn Tofail University, Kenitra, Morocco

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      40P - Comparison of clinical characteristics and survival of lung cancer patients in a Canadian province and in the United States according to insurance status (Now Available) (ID 609)

      12:30 - 12:30  |  Presenting Author(s): A. Matutino  |  Author(s): E. Kornaga, M. Dean, A.A.L. Pereira, G. Bebb

      • Abstract
      • Slides

      Background:
      Cancer outcomes in the United States (US) was shown to be influenced by insurance status for the most types of cancer. In Canada, health care access is similar for the whole population. The Affordable Care Act (ACA) was implemented to expand access to health care coverage in the US. This study aimed to compare clinical characteristics and survival of lung cancer pts in a Canadian province and in the US according to insurance status over the years.

      Methods:
      Data was obtained from the US SEER database and the Alberta, Canada Glans-Look lung cancer database. Pts were included if they were 18-64yo and diagnosed between 2007–2012. To account for the introduction of the ACA in the US, data was also analyzed by years 2007–2009 vs 2010–2012. Overall survival (OS) was evaluated over 26 months and the 25th, 50th and 75th percentiles of survival time, if reached, were estimated. Pearson's χ[2] was used to assess significance of associations with diagnosis year, and unadjusted associations were compared using the log-rank test. Hazard ratio's (HR) were estimated using the Cox proportional hazards model.

      Results:
      A total of 65,791 pts from the SEER database and 1,034 pts from the Canada Glans-Look database were included. The comparison of survival outcomes of the Canadian population vs the US population based on insurance status (insured, Medicaid, uninsured) showed that the Canadian survival was similar to the US Medicaid group (11.7 vs 10 m at percentile 50 of survival), while US insured group had a better outcome of 16 months (p < 0001). When comparing 2007–2009 vs 2010–2012, overall survival remained similar for the Canadian population (10.9 vs 13 m, p = 0.8876), while it improved in the post-ACA years for the US population (13 vs 14 m p = 0.0002). In both Canadian and US population, female sex, earlier stages and adenocarcinoma pts presented with better survival outcomes (p < 0.0001).

      Conclusions:
      Among lung cancer pts, the Canadian outcomes resembled the US Medicaid group for survival outcomes. In the US population, Medicaid and uninsured pts experienced worse survival. Survival rates in all insurance groups improved after the implementation of the ACA in the US, remaining similar in the Canadian population.

      Clinical trial identification:


      Legal entity responsible for the study:
      Alberta Health Services

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      41P - Gender based variations in presentation and management of lung cancer at a south Asian tertiary referral centre (ID 447)

      12:30 - 12:30  |  Presenting Author(s): V. Vashistha  |  Author(s): C. Choudhari, A. Garg, A. Gupta, G. Parthasarathy, C. Mohan, K. Madan, V. Hadda, G.C. Khilnani, A. Mohan

      • Abstract

      Background:
      The clinical presentation and management strategies of lung cancer for lesser income women in South Asia has rarely been evaluated. We sought to compare the baseline characteristics, duration of symptoms, histology, staging, and rates of treatment between women and men at our public tertiary referral center in northern India.

      Methods:
      A retrospective review of patients diagnosed with lung cancer between January 1, 2008 and December 31, 2016 was completed. Baseline variables, previous treatment for tuberculosis, length of symptoms, tumor characteristics, and forms of treatment were collected and compared between women and men.

      Results:
      1370 total patients were selected including 230 females (16.8%). Compared to men, women were younger (mean {SD} age, 54.6 {10.9} vs. 58.4 {10.8}; p < 0.001), less likely current or previous smokers (34.1% vs. 86.1%; p < 0.001), had a lower rate of secondary education (46.3% vs. 69.4%, p < 0.001), and increased rate of previous tuberculosis treatment (37.3% vs. 29.6%, p = 0.03). No differences in duration (p = 0.47) of symptoms at time of diagnosis, rates (p = 0.92) of non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC), or rates (p = 0.20) of ultimately receiving some form of cancer treatment between women and men were observed. Women had higher rates of stage IV disease (75.0% vs. 62.2%, p < 0.001) for NSCLC but shared no difference (p = 0.48) in rates of extensive stage for SCLC compared to men.

      Conclusions:
      Women with lung cancer in our region are younger, more likely non-smokers, less well educated, more likely to have received recent anti-tuberculosis therapy before being correctly diagnosed, and more likely to present with metastatic disease. Clinicians should maintain a high index of suspicion even in younger women presenting with new-onset respiratory symptoms which may mimic tuberculosis.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      42P - Is there a delay in diagnosis of lung cancer in women? (Now Available) (ID 442)

      12:30 - 12:30  |  Presenting Author(s): Y. Tolwin  |  Author(s): B. Silverman, N. Peled

      • Abstract
      • Slides

      Background:
      Lung cancer remains a major cause of death in the world, and while it was considered in the past to be primarily a male disease, the number of female patients has risen in recent years, such that rates among women are similar to those among men. Nevertheless, it has been found previously (in cardiovascular disease) that when there is a sex specific stereotype to a disease, it may remain entrenched in medical diagnostic processes, so as to cause belated diagnosis among the other sex. Here we aim to characterize the effects of sex on lung cancer diagnosis.

      Methods:
      We performed a retrospective analysis using two cohorts, 458,132 patients from the USA using the SEER (Statistics, Epidemiology and End Results) database, and 30330 patients from Israel. Patient cohorts were analyzed for sex-based differences by tumor type and stage at diagnosis, and results were stratified by race and analyzed with data regarding possible confounders such as smoking and socio-economic factors.

      Results:
      Male patients were more likely than female patients to be diagnosed at stage 3–4, consistent across lung cancer types, cancer registries, smoking, and racial and socioeconomic backgrounds. The exception to this was the arab population in the Israeli cohort, where there was no significant difference between men and women in the percentage diagnosed in later stages. The difference between the percentage of men vs. women diagnosed in stages 3–4 correlated negatively with increased female ever smokers and with squamous and small cell carcinoma, and were not correlated with the rate of cancer in women, or the difference between male and female cancer rates. Race was shown to have a significant effect on the percentage of women diagnosed in later stages.

      Conclusions:
      Results do not show a general belated diagnosis of lung cancer in women. In fact, the opposite appears to be the case. Results appear to point to the fact that smoking women are more likely to be diagnosed at later stages, which is consistent with current literature. Israeli arab women may suffer from belated lung cancer diagnosis, despite very low levels of smoking, perhaps owing to social and cultural causes.

      Clinical trial identification:


      Legal entity responsible for the study:
      Institute of Oncology, Soroka Medical Center & Ben-Gurion University

      Funding:
      Has not received any funding

      Disclosure:
      N. Peled: Advisor & honorarium from AZ, BI, BMS, Lilly, MSD, Novartis, Pfizer, Roche, Takeda, FMI. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      43P - A network-based signature to predict the survival of non-smoking lung adenocarcinoma (ID 388)

      12:30 - 12:30  |  Presenting Author(s): Q. Mao  |  Author(s): F. Jiang, L. Xu

      • Abstract

      Background:
      A substantial increase in the number of non-smoking lung adenocarcinoma (LAC) patients draws extensively attention in the past decades. Effective biomarkers are needed to identify high-risk patients to guide the therapy. Here, we provided a network-based signature to predict the survival of non-smoking LAC.

      Methods:
      Gene expression profiles were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Significant gene co-expression networks and hub genes were identified by Weighted Gene Co-expression Network Analysis (WGCNA). Potential mechanisms and pathways of co-expression networks were analyzed by Gene Ontology (GO). The predictive signature was constructed by penalized Cox regression analysis and tested in two independent datasets.

      Results:
      Two distinct co-expression modules were significantly correlated with non-smoking status across four GEO datasets. GO revealed that nuclear division and cell cycle pathways were main mechanisms of the blue module and that genes in the turquoise module were involved in lymphocyte activation and cell adhesion pathways. Seventeen genes were selected from hub genes at an optimal lambda value and built the prognostic signature. The prognostic signature distinguished the survival of non-smoking LAC (training: hazard ratio (HR) = 3.696, 95%confident interval (CI): 2.025–6.748, p < 0.001; testing: HR = 2.9, 95%CI:1.322–6.789, p = 0.006; HR = 2.78, 95%CI:1.658–6.654, p = 0.022) and had moderate predictive abilities in the training and validation datasets.

      Conclusions:
      The prognostic signature is a promising predictor of non-smoking LAC patients, which might benefit to clinical practice and precision therapeutic management.

      Clinical trial identification:


      Legal entity responsible for the study:
      Lin Xu

      Funding:
      National Natural Science Foundation of China (Nos. 81472702, 81501977 and 81672294)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      44P - A large population-based study on large cell neuroendocrine lung cancer: A SEER database analysis (ID 293)

      12:30 - 12:30  |  Presenting Author(s): X. Fu  |  Author(s): H. Zhang, Y. Yang, M. Liu, J. Lu, X. Liang, K. Nan, Y. Yao, T. Tian

      • Abstract

      Background:
      Large cell neuroendocrine lung cancer (LCNELC) is a subtype of lung cancer with neuroendocrine morphology and differentiation on immunohistochemistry, a high mitotic rate and non-small cell cytological features. However large population-based study on the clinicalpathological characteristics of LCNELC is lacking. Nomogram provides a visualized equation that the behavior of a predictor is represented in scales. In this study, we aim to explore the potential associations between clinicopathological factors and prognosis in SEER-18 database and to establish a nomogram model to predict the prognosis of LCNELC.

      Methods:
      We used the SEER-18 database to study the prognosis of LCNELC patients from 2000 to 2014 in the United States. All statistical analyses were performed by R software. We used packages “SEERaBomb”, “survival”, “rms”, and “rcorrp.cens” to obtain data and to build and evaluate the nomogram.

      Results:
      A total number of 1231 patients were enrolled. Sex, marital status, age at diagnosis, tumor size, AJCC TNM stage, and SEER histologic stage affect the prognosis of LCNELC patients. We included these factors to develop the nomogram prediction model. The Harrel's C-index showed that the nomogram model has a better prediction than traditional AJCC TNM staging system. We evaluated different surgeries for patients at early and advanced TNM stages as well as different SEER histologic stages, and suggested that early TNM stage or localized and regional SEER histologic stages patients benefit from surgical resection, especially lobectomy or bilobectomy.

      Conclusions:
      Age, sex, marital status, tumor size, TNM stage, SEER histologic stage, and both radiation and surgery treatment are independent prognostic variants for LCNELC. And early TNM stage or localized and regional SEER stage of LCNELC patients benefit from surgical resection, especially lobectomy or bilobectomy.

      Clinical trial identification:


      Legal entity responsible for the study:
      The First Affiliated Hospital of Xi'an Jiaotong University

      Funding:
      Natural Science Foundation of Shaanxi Province (No.2017JM8019). International cooperation project in science and technology of Shaanxi province (No. 2016KW-017). Wu Jieping Medical Foundation (No. 50603020).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      48P - Clinical characteristics of Korean lung cancer patients with programmed death-ligand 1 expression (ID 301)

      12:30 - 12:30  |  Presenting Author(s): H. Park

      • Abstract

      Background:
      Programmed death-ligand 1 (PD-L1) is a transmembrane protein binds to the programmed death-1 (PD-1) receptor and anti-PD-1 therapy enables the immune response against tumors. The aim of this study was to assess the clinical and pathologic characteristics of PD-L1 positive lung cancer patients in Korea.

      Methods:
      We retrospectively reviewed the clinical data of pathologically proven lung cancer patients, and collected 267 cases of formalin-fixed, paraffin-embedded tissue sample from single institution. PD-L1 expression was detected by immunohistochemical assay using Monoclonal Mouse Anti-PD-L1, Clone 22C3. PD-L1 protein expression is determined by using Tumor Proportion Score (TPS), the percentage of viable tumor cells showing partial or complete membrane staining. The specimen should be considered PD-L1 positive as TPS ≥ 50% of tumor cells. We categorized according to the percentage of TPS; more than 1% or 50%.

      Results:
      A total of 267 patients were enrolled and major histologic types were adenocarcinoma (ADC)(69.3%). The majority was smoker (67.4%) and clinical stage IV (60.7%). 31 (11.6%) cases of EGFR mutation and 17 (6.4%) cases of ALK FISH positive were included. The patients who showed TPS ≥ 1% and 50% were 116 (42%) and 58 (21%), respectively. More than 1% of TPS group was older than below (64.83 ± 9.38 vs. 61.73 ± 10.78, p = 0.014). More than 1% of TPS group was consisted of ADC (67.8%) and squamous cell carcinoma(SqC) (29.6%) histology. And more than 50% of TPS group was composed of ADC (72.4%), SqC (22.4%). More than 1% of TPS group was significantly older than less (64.83 ± 9.38 years vs. 61.73 ± 10.78 years, p = 0.014). The rate of poorly differentiated pathology was significantly higher in TPS ≥ 1% group (42(40.8%) vs. 32(25.8%)) and TPS ≥ 50% group (25(53.2%) vs. 49(27.2%)). There was no difference in smoking, EGFR mutation, ALK rearrangement status or biopsy site.

      Conclusions:
      In Korean lung cancer patients, PD-L1 positive group defined as TPS ≥ 1% was older than negative group. And major histology was poorly differentiated non-small cell lung cancer in both TPS ≥ 1% and 50% groups.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chonnam National University Hospital, Hwasun

      Funding:
      Has not received any funding

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      49P - Diagnosis of small pulmonary lesions by transbronchial lung biopsy with radial endobronchial ultrasound and virtual bronchoscopic navigation versus CT-guided transthoracic needle biopsy: A systematic review and meta-analysis (ID 307)

      12:30 - 12:30  |  Presenting Author(s): J.H. Chang  |  Author(s): Y.J. Ryu, J.H. Lee

      • Abstract

      Background:
      Advances in bronchoscopy and computed tomography (CT)-guided lung biopsy have improved the approach to small pulmonary lesions (PLs), leading to an increase in preoperative histological diagnosis. The purpose of this meta-analysis was to evaluate the efficacy and safety issues between transbronchial lung biopsy with radial endobrochial ultrasound and virtual bronchoscopic navigation (TBLB-rEBUS&VBN) as a bronchoscopic (BR) approach and CT-guided transthoracic needle biopsy (CT-TNB) as a percutaneous (PC) approach for the tissue diagnosis of small PLs.

      Methods:
      A systematic search for relevant studies was performed in May 2016 using five electronic databases: MEDLINE, EMBASE, Cochrane Library Central Register of Controlled Trials, Web of Science, and Scopus; the selected papers were assessed using meta-analysis. The papers were limited to those published since 2000 that studied small PLs ≤ 3 cm in diameter. To select studies of TBLB-rEBUS&VBN, rEBUS with a GS was used for peripheral pulmonary lesion (PPL) diagnosis and virtual bronchoscopy was used as a navigational method. The three methods of conventional CT-guided transthoracic needle biopsy, CT fluoroscopy-guided transthoracic biopsy, and C-arm cone-beam CT-guided transthoracic biopsy were used for CT-TNB.

      Results:
      From 7345 records, 9 papers on the bronchoscopic approach and 15 papers on the percutaneous approach were selected. The pooled diagnostic yield was 75% (95% confidence interval [CI], 69–80) in the BR approach and 93% (95% CI, 90–96) in the PC approach. Especially for tissue biopsy of PLs <2 cm, we recommend prioritizing CT-TNB, which had a 26% better diagnostic yield than TBLB-rEBUS&VBN. However, for PLs >2 cm but ≤3 cm, the yield in the BR approach improved to 81% (95% CI, 75–85). Complications of pneumothorax and hemorrhage were rare in the BR approach and quite common in the PC approach.

      Conclusions:
      CT-TNB was superior to TBLB-rEBUS & VBN for the evaluation of small PLs. However, for lesions greater than 2 cm, the BR approach was quite valuable when considering its diagnostic yield of over 80% with its low risk of procedure-related complications.

      Clinical trial identification:


      Legal entity responsible for the study:
      Jung Hyun Chang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      50P - The expression and significance of miR-146a and miR-146b in BALB/c mice pulmonary precancerous lesions induced by fine particulate matter (ID 235)

      12:30 - 12:30  |  Presenting Author(s): T. Hou  |  Author(s): G. Wang

      • Abstract

      Background:
      Lung cancer is one of the commonest malignant tumors threatening the health of human being. The morbidity and mortality of lung cancer increased rapidly, early detection and early treatment is the key to increase the survival rate of lung cancer patients. Studies have confirmed that air pollution is closely related to the occurrence of lung cancer. Air pollution (mainly PM2.5) has been classified as a carcinogen by the International Agency for Research on Cancer. MicroRNAs (miRNAs) are endogenous, noncoding RNAs consisting of 21–23 nucleotides which are stable existence in the blood. It is one kind of potential diagnostic markers in serum. In order to verify whether microRNA-146 can be used as early warning indicators of precancerous lesion of lung cancer.

      Methods:
      30 BALB/c mice were treated with noninvasive tracheal instillation of fine particulate matter suspension at different doses for 90 days (two times one week), histopathological changes, pro-infammatory factors levels, pulmonary functions and the relative expression of microRNA-146 were detected.

      Results:
      Histopathological changes showed atypical hyperplasia of alveolar epithelial cells, alveolar macrophages with engulfed particles and lymphocyte aggregation in bronchiole and alveolar. Pro-inflammatory factors IL-6, IFN-γ and TNF-α were increased significantly, however, PIF and PEF were decreased significantly. Lung resistance Increased and MVV reduced from the general tendency, the relative expressions of miR-146a and miR-146b were up-regulated remarkably in treatment groups compared to the control group.

      Conclusions:
      MiR-146a and miR-146b may be novel warning biomarkers for early detection of pulmonary precancerous lesions.

      Clinical trial identification:
      NO

      Legal entity responsible for the study:
      Peking University First Hopital

      Funding:
      Beijing Municipal Natural Science Foundation (Key Program: No.7161013)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      53P - Lung cancer: Beyond EGFR and ALK dichotomy (Now Available) (ID 359)

      12:30 - 12:30  |  Presenting Author(s): C.T. Satheesh  |  Author(s): A.D. Bhatt, M. Singhal, R. Aarthi, K. Shubham, G. Madhura, V. Maka, R. Naik, S. Iyer, G. sHAFI

      • Abstract
      • Slides

      Background:
      Lung cancer, one of the most frequent cancers worldwide has long relied on molecular testing of major biomarkers such as EGFR/ALK. Achieving superior clinical outcomes needs a comprehensive emphasis beyond contemporary EGFR and ALK. Several technologies arose in par with molecular testing for EGFR/ALK, most of them failing to comprehend beyond due to a universal skepticism among clinicians though recommended in NCCN guidelines.

      Methods:
      A total 137 lung cancer cases from NGS tested data (PositiveSelect) comprising 91 males and 46 females were investigated. EGFR, ALK positivity were used for data dichotomization to understand therapeutic utility of rare alterations beyond EGFR/ALK.

      Results:
      Upon dichotomization, 28% were identified with EGFR + ALK variants favoring direct EGFR/ALK targeted therapeutics. The remaining 72% harbored no EGFR/ALK variants descending into the category of chemotherapy in current clinical practice. Similarly, 23% harbored EGFR variants carrying the beneficial effects of EGFR TKIs and remaining 77% displayed no EGFR variants sloping towards chemotherapy. The study conquered the incompetence of targeted drug utilization on conventional diagnosis of EGFR/ALK in (EGFR) and (EGFR + ALK) negative cohorts. Based on our analysis of EGFR negative cohort, we identified clinically actionable variants in KRAS (7%), BRAF (2%), ERBB2 (1%), MET (2%) and RET (3%) expressing potential for targeted therapy excepting EGFR TKIs. Correspondingly, KRAS (6%), ERBB2 (1%), MET (1%) and RET (3%) variants were identified in EGFR + ALK negative cohort enabling the utility of targeted therapeutics apart from EGFR/ALK. Only 35% of the two negative limbs were categorized into chemotherapy which would have been entire cohort otherwise.

      Conclusions:
      The study accentuates the potential of comprehensive genomics in ascertaining the hallmarks of lung cancer beyond EGFR/ALK dichotomy also liaising between theory and utilization of broad spectrum genomic testing among medical professionals to circumvent chemotherapy. Thus, chemotherapy dependence in EGFR/ALK negative cohort could be effectively curtailed by clinicians evidencing better clinical outcomes.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      Positive Bioscience

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      54P - Epidermal growth factor receptor expression (EGFR) in serum as a marker of treatment response and survival in advanced squamous cell lung cancer (Now Available) (ID 353)

      12:30 - 12:30  |  Presenting Author(s): A. Mohan  |  Author(s): A. Ansari, M. Masroor, A. Saxena, K. Luthra, R.M. Pandey, D. Jain, R. Kumar, G.C. Khilnani, R. Guleria

      • Abstract
      • Slides

      Background:
      Reliable biomarkers are needed to prognostigate patients with advanced lung cancer following chemotherapy. This study evaluated the association of serum EGFR expression with disease severity and treatment response in advanced Squamous cell lung cancer.

      Methods:
      Newly diagnosed subjects with advanced Squamous cell lung cancer (stage IIIB and IV) were enrolled. Relevant demographic data were recorded, including performance status (assessed by Karnofsky performance status [KPS] and European Co-operative Oncology Group [ECOG] scoring system). Epidermal growth growth factor receptor (EGFR) expression was estimated in serum using reverse transcriptase polymerase chain reaction (RT-PCR) at baseline and following four cycles of Carboplatin – Paclitaxel chemotherapy. Response was assessed using the RECIST 1.1 criteria. Objective response rate (ORR) was defined as Complete remission (CR) or partial response (PR) and Disease control rate (DCR) was defined as CR/PR/Stable disease (SD). Kaplan meier curve was used to compare the overall survival (OS) between subjects based on median EGFR expression level as the cut-off.

      Results:
      A total of 82 subjects were enrolled. These included 79 (96.3%) males with mean (SD) age of 61.9 (9.8) years and 48.8% having metastatic disease. Majority were current / former smokers (97.6%); 59.7% had KPS ranging between 40 -70 and 48.1% had ECOG of 0/II. The baseline mean (± SD) serum EGFR expression was 17.8 ± 9.3 fold increase over control values, with median (min., max.) of 18.2 (4.4, 42.8). Following chemotherapy, ORR and DCR were 59.7% and 77.3% respectively. Significant reduction in EGFR expression was observed with median (min, max.) absolute and percentage reduction of 6.2 fold (−3.1, 33.6), and 54.2 % (−55.6, 78.4) respectively; p < 0.001. No significant association was observed between change in EGFR expression and age, gender, disease stage, performance status, ORR or DCR. Subjects with baseline EGFR expression of greater than 16.0 fold had significantly worse OS than those with <16.0 fold increase.

      Conclusions:
      EGFR is over-expressed in advanced squamous cell lung cancer and is significantly down-regulated following chemotherapy; additionally, baseline expression is a useful marker of overall survival following chemotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Department of Biotechnology, India

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      56P - Plasma circulating ctDNA: Potential biomarker in non-small cell lung carcinoma and clinical significance (ID 355)

      12:30 - 12:30  |  Presenting Author(s): V. Goel  |  Author(s): S. Tiwari, V. Talwar, N. Pattnaik, P. Dash, S. Raina, M.C. John

      • Abstract

      Background:
      Circulating cell free tumour DNA (ctDNA) from liquid biopsy is a potential source of tumour genetic material especially in case of non-availability of tissue biopsy for EGFR testing. Detectable levels of oncogenic driver mutations in peripheral blood have been shown to be associated with poorer prognosis and good predictor of EGFR TKI efficacy.

      Methods:
      Liquid biopsy was performed on 95 NSCLC patients with matched tumour tissue for genomic analysis. An EGFR mutation of one major molecular subtype in NSCLC was performed on massive parallel sequencing. Single gene EGFR mutation analysis was performed on the ctDNA by using ultra deep sequencing on the HiSeq platform. Then custom designed bioinformatics algorithms were used to detect somatic mutations at allele frequencies as low as 0.01%.

      Results:
      Overall concordance of mutation status between 95 pairs of tissue and plasma ctDNA samples for EGFR mutation status was about 93%. 30.5% (29/95) of the study subset was EGFR mutated on tissue typing and 27.36% (26/95) in ctDNA. Positive predictive value was 100% and negative predictive value was 95.6% – with diagnostic accuracy of 97%. A false negative rate of 4% was observed in this study. 12 out of 95 (12.63%) samples which had rare Exon19 deletions and complex indels could be confidently detected by NGS methods only. An objective efficacy response rate for Gefitinib was estimated at 70%, with a disease control rate of 94%. Median period of follow-up was 13.9 months. Median PFS was 16.8 months (95% CI 11.168–26.198).

      Conclusions:
      12% of newly diagnosed NSCLC patients could get the additional benefit of targeted therapy, by using the NGS which detected oncogenic driver mutations. Liquid Biopsy offer significant diagnostic, prognostic, and predictive information.

      Clinical trial identification:


      Legal entity responsible for the study:
      Rajiv Gandhi Cancer Hospital and Research Center

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      57P - Targeted sequencing of plasma-derived cfDNA in patients with metastatic NSCLC (Now Available) (ID 557)

      12:30 - 12:30  |  Presenting Author(s): R. Pasquale  |  Author(s): F. Bergantino, F. Fenizia, L. Forgione, C. Roma, A. De Luca, G. Rocco, A. Morabito, N. Normanno

      • Abstract
      • Slides

      Background:
      Circulating cell free DNA (cfDNA) is an alternative to tumor tissue for molecular profiling in non-small cell lung cancer (NSCLC). Next generation sequencing (NGS) with targeted panels can concurrently evaluate multiple actionable mutations.

      Methods:
      NGS analysis was performed on tissue samples with the Oncomine Solid Tumour DNA kit, while plasma samples were analyzed with the Oncomine Lung cfDNA Assay. Droplet Digital PCR (ddPCR) was performed with the QX200 System to solve discordant cases.

      Results:
      We performed NGS analysis of tumor samples and matched cfDNA obtained from 102 patients with metastatic NSCLC before systemic treatment. NGS detected EGFR mutations in 21/25 plasma samples from EGFR-mutant patients (sensitivity 84%), and in 3/77 samples from EGFR wild type (wt) patients (specificity 96.1%), with a concordance rate of 93.1%. Analysis with ddPCR confirmed in plasma samples the absence of EGFR variants in false-negative cases according to NGS; in the 3 cases with EGFR mutant-plasma and wt-tissue, ddPCR confirmed the presence of EGFR mutations at low allelic frequency in both plasma and tissue samples, therefore confirming the specificity of NGS analysis. We also evaluated in the cohort of 77 EGFR wt tumors the concordance between tumor and plasma for the mutations in genes other than the EGFR covered by both NGS panels. The mean concordance was low (57.3%). In particular, a high discordance among KRAS mutations was observed. Out of 11 cases with KRAS mutations in tissue, only 3 showed also KRAS mutations in plasma. Analysis with ddPCR identified KRAS mutations in the cfDNA from 4/5 available samples that were negative by NGS, suggesting a low sensitivity of the panel for RAS mutations. In addition, NGS analysis revealed KRAS mutations in 3 cases that were negative on tissue. These variants were validated by ddPCR, confirming the specificity of NGS analysis and suggesting tumor heterogeneity for these variants.

      Conclusions:
      Our study showed that plasma-NGS is a suitable method for EGFR genotyping in NSCLC. Relatively low sensitivity and tumor heterogeneity might limit the ability of NGS to identify driver alterations other than EGFR variants.

      Clinical trial identification:


      Legal entity responsible for the study:
      Istituto Nazionale Tumori “Fondazione G. Pascale”- IRCCS, Naples, Italy

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      58P - Co-treatment of trametinib (MEK inhibitor) with TPX-0005 (Src/FAK/JAK2 inhibitor) synergistic in KRAS mutant NSCLC cell lines and CDCP1 acts as a biomarker in KRAS mutant patients (p) (Now Available) (ID 407)

      12:30 - 12:30  |  Presenting Author(s): C. Codony-Servat  |  Author(s): M. Llanos Gil Moreno, J. Bracht, I. Attili, C. Lazzari, V. Gregorc, M. Ito, S. Viteri, N. Karachaliou, R. Rosell

      • Abstract
      • Slides

      Background:
      KRAS mutant lung adenocarcinoma has a dismal prognosis. In the current study, we identified combination targets for trametinib, a MEK inhibitor, which acts downstream of KRAS to suppress signaling through MAPK cascade. However, we have previously shown that selumetinib (MEK inhibitor) in KRAS mutant NSCLC cells caused a rebound of ERK, AKT and STAT3, as well as YAP phosphorylation (Y357), NOTCH3 and activation of RTKs, AXL and MET. We hypothesize that, in KRAS mutant NSCLC, suppression of MAPK could lead to activation of Src-YAP1-AXL-MET. Since CUB domain-containing protein 1 (CDCP1) activates Src, we consider that CDCP1 could be a biomarker of Src activation.

      Methods:
      Cell viability assay, colony formation assay and western blotting were performed to assess the treatment of trametinib plus TPX-0005 in a panel of 8 NSCLC cell lines: A549, Calu6, H23, H460, H2009, H2030, H441, H727. Synergy between trametinib and TPX-0005 was assessed via the Chou-Talalay method to estimate the combination index (CI). CI values <0.7 were considered synergistic, with decreasing CI values indicating greater synergy. We examined tumor samples of 32 KRAS mutant NSCLC p for CDCP1 mRNA levels.

      Results:
      The combination of trametinib with TPX-0005 was synergistic or additive in all cell lines tested. H23 and Calu6 were the most synergistic, followed by H441 and H2030. In the majority of cell lines examined, the colony formation assay resulted in an almost complete abrogation of tumor cell colonies, particularly in the H23 and A549. Western blotting indicated that the combination of trametinib with TPX-0005 abolished the phosphorylation of STAT3 (Y705), paxillin (Y118), a readout of FAK, and Src (Y416). The median PFS of 32 KRAS mutant NSCLC p was 2.5 months and the overall survival was 13.4 months. According to CDCP1 levels, the median PFS was 3.5 months for those with low CDCP1 and 1.4 months for those with high CDCP1 (P = 0.012). The median survival was 16.3 months for p with low CDCP1, and only 3.2 months for those with high CDCP1 (P = 0.023).

      Conclusions:
      The combination of trametinib plus TPX-0005 shows significant in-vitro activity in the majority of KRAS mutant NSCLC cell lines and the mRNA levels of CDCP1 could be a biomarker in KRAS mutant NSCLC p, indicating the activation of Src-YAP signaling. Clinical trials with the combination of MEK inhibitors with TPX-0005 are warranted.

      Clinical trial identification:


      Legal entity responsible for the study:
      IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain

      Funding:
      Fundació Obra Social “La Caixa”

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      59P - Circulating microRNAs as novel predictive markers of afatinib efficacy in squamous cell lung cancer (SCC): An exploratory sub-analysis of the LUX-Lung 8 trial (Now Available) (ID 593)

      12:30 - 12:30  |  Presenting Author(s): Y. Gaston Math_  |  Author(s): P. Fogel, S. Martin-Lannerée, C. Marcaillou, E. Lallet, N. Krämer, N. Gibson, F. Solca, E. Ehrnrooth, J. Cadranel

      • Abstract
      • Slides

      Background:
      Screening of circulating microRNAs is a promising avenue for the discovery of novel biomarkers in cancer therapy. In the LUX-Lung 8 trial, the ErbB family inhibitor Afatinib (A) provided a significant progression free survival (PFS) and overall survival (OS) benefit compared to the EGFR TKI Erlotinib (E) in 2nd line treatment of advanced SCC. To detect new markers associated with clinical outcomes, we performed a post hoc extensive screening of miRNAs in serum samples from a subset of LUX-Lung 8 trial patient population.

      Methods:
      1787 miRNA expression levels were measured by miRNA-seq using the Illumina HiSeq4000 platform in baseline (BL) (n = 133) and cycle 2 (C2) (n = 109) serum samples from 133 LUX-Lung 8 patients randomly selected with stratification on smoking, treatment, and interval from last dose of chemotherapy. All variables were tested for their BL prognostic and predictive value on OS in a Cox model adjusted for prognostic factors. For miRNAs with FDR adjusted p-value <0.05, a data-driven cut-off was determined and Kaplan Meier estimates were performed comparing low- and high-expressers. Analysis of C2 values was conducted.

      Results:
      29 miRNAs were found to be prognostic of OS and 8 to be predictive of treatment effect on OS with FDR p-value p < 0.05. MiR-3150b-3p was the most significant predictive variable (FDR p < 0.001). In the A arm, median OS was 2.7 m in miR-3150b-3p high- vs 8.5 m in low-expressers (HR = 3.4, p = 0.0001) and 5.7 m in high-expressers treated with E (HR = 2.7, p = 0.01). Furthermore, in the A arm, but not in the E arm, high miR-3150b-3p expression at C2 in patients with low BL values was associated with a shorter OS compared to patients remaining low (7.4 m vs 10.4 m, HR = 2; p = 0.08).

      Conclusions:
      Using a rigorous methodology, we found new highly prognostic markers and several novel markers with high predictive value of a differential treatment benefit between A and E. This suggests the two drugs have different mechanism of action. Because this was an exploratory analysis and the effect size could be over-estimated, those results should be replicated in a larger study.

      Clinical trial identification:
      NCT01523587

      Legal entity responsible for the study:
      IntegraGen

      Funding:
      IntegraGen

      Disclosure:
      Y. Gaston Mathé, S. Martin-Lannerée, C. Marcaillou, E. Lallet: Employee of the study sponsor. P. Fogel: Subcontractor of the study sponsor. N. Krämer: Subcontractor of the clinical trial sponsor and owner of the rights on afatinib, Boehringer-Ingelheim. N. Gibson, F. Solca, E. Ehrnrooth: Employee of the clinical trial sponsor and owner of the rights on afatinib, Boehringer-Ingelheim. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      60P - Circulating miR-31 as a predictive marker of EGFR TKI treatment efficacy in squamous cell lung cancer (SCC): A sub-analysis of the LUX-Lung 8 trial (Now Available) (ID 205)

      12:30 - 12:30  |  Presenting Author(s): Y. Gaston Math_  |  Author(s): P. Fogel, S. Martin-Lannerée, C. Marcaillou, E. Lallet, N. Krämer, N. Gibson, F. Solca, E. Ehrnrooth, J. Cadranel

      • Abstract
      • Slides

      Background:
      MicroRNA miR-31-3p and 5p have been reported to predict anti-EGFR cetuximab efficacy in metastatic colorectal cancer but have not been studied in other indications. In the LUX-Lung 8 trial, ErbB family blocker Afatinib (A) provided a significant PFS and OS benefit compared to EGFR TKI Erlotinib (E) in 2nd line treatment of advanced SCC. We retrospectively analyzed the association of miR-31-3p and miR-31-5p serum levels with treatment benefit in a sub-population of this study.

      Methods:
      MiRNA expression levels from baseline (BL) (n = 133) and cycle 2 (C2) (n = 109) serum samples from 133 LUX-Lung 8 trial patients selected randomly with stratification on smoking status, treatment and interval from last dose of chemotherapy were measured by miRNA-Seq using Illumina HiSeq4000 platform. MiR-31-3p, miR-31-5p or combined miR-31 were tested for their prognostic and predictive value of treatment effect on OS using Cox models adjusted for prognostic factors and Kaplan Meier estimates after determination of a data-driven cut-off defining low or high expressers.

      Results:
      Mir-31-3p was detected in 19/133 BL samples and was predictive of E efficacy (p < 0.01). MiR-31-5p was prognostic of OS in the E (p = 0.03) but not in the A arm. Patients with measurable miR-31-3p or high miR-31-5p had a better OS when treated with A compared to E (miR-31-3p: 15 m [6.3;70] vs 4 m [2.7;7.8], HR = 0.3, p < 0.05; miR-31-5p: 7.3 m [4.7;16.5] vs 3.3 m [2.4;6.5], HR = 0.5, p = 0.07). No difference between treatment arms was seen in patients with no measurable miR-31-3p or low miR-31-5p expression. In the E arm only, high miR-31 expression at C2 in patients with low BL miR-31 was associated with a shorter OS compared to patients remaining low (4.2 m [2.8;9.2] vs 10.3 m [7.8;13.0], HR = 0.5, p = 0.05).

      Conclusions:
      MiR-31-3p/5p serum levels were predictive of E treatment efficacy. Patients with high BL miR-31 levels had lower benefit of E and higher benefit of A, suggesting different mechanisms of action for the drugs. Increase of miR-31 levels during treatment with E was associated with poor outcomes. Measurement of circulating mir-31 has potential to help optimizing treatment choice between E and A for SCC patients. Those results should be replicated in a larger study.

      Clinical trial identification:
      NCT01523587

      Legal entity responsible for the study:
      IntegraGen

      Funding:
      IntegraGen

      Disclosure:
      Y. Gaston Mathé: Employee of IntegraGen, the sponsor of the study and owner of IP rights on the disclosed results. P. Fogel: Fees from the study sponsor for the performance of the statistical analysis. S. Martin-Lannerée, C. Marcaillou, E. Lallet: Employee of the study sponsor. N. Krämer: Subcontracter for the clinical trial sponsor and afatinib product owner, Boehringer-Ingelheim. N. Gibson: Employee of the clinical trial sponsor and afatinib product rights owner Boehringer-Ingelheim. F. Solca, E. Ehrnrooth: Employee of the clinical trial sponsor and owner of the rights on afatinib, Boehringer-Ingelheim. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      61P - mir-125b plays a tumor suppressor role in inflammation-related non-small cell lung cancer via repressing IGF-1 signal pathway (ID 509)

      12:30 - 12:30  |  Presenting Author(s): Y. Zhang  |  Author(s): B. Han, S. Hu, Y. Lou, T. Chu, J. Qian, Q. Chang

      • Abstract

      Background:
      Epidemiologic data have indicated that chronic inflammation was highly associated with the pathogenesis of lung cancer. However, the molecular relations between inflammation and lung cancer have not been well understood. MicroRNAs could connect the inflammatory response with tumorigenesis through regulating their cancer-related targets. The aim of the present study was to identify the core miRNA in inflammation-related lung cancer and its potential mechanisms.

      Methods:
      RT-PCR was used to detect the expression of miRNAs and mRNAs. CKK8 and flow cytometry assays was performed for the function experiments. Microarray analysis and IPA analysis were used to predict the potential signal pathway.

      Results:
      Mir-125b was the most dramatically up-regulated miRNAs after treated with IFN-r, whereas after stimulated with IL-10, mir-125b was the most strikingly down-regulated ones. Restoration of mir-125b expression could completely overrode the impact of IL-10 on both cell proliferation and apoptosis in NSCLC cell lines. And the level of mir-125b was significantly lower in 30 NSCLC tumor tissues compared with normal controls (P < 0.0001). Microarray analysis found 69 up-regulated genes and 105 down-regulated genes after down-regulate mir-125b. And IPA analysis indicated that IGF-1 signaling pathway was dramatically activated. The results were validated by RT-PCR.

      Conclusions:
      MiR-125b might play a tumor suppressor role via inhibiting IGF-1 signaling in inflammation-related lung cancer.

      Clinical trial identification:


      Legal entity responsible for the study:
      Yanwei Zhang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      62P - MAP kinase signaling pathway in pulmonary adenocarcinomas: A study from India (ID 553)

      12:30 - 12:30  |  Presenting Author(s): V. Singh  |  Author(s): P.S. Malik, D. Jain

      • Abstract

      Background:
      Tyrosine kinase inhibitors (TKIs) are effective in EGFR and ALK driven non-small cell lung cancer (NSCLC). However, drug insensitivity, tumor recurrence, and resistance caused by second mutations in the EGFR or aberrant bypass signaling are key challenges. To overcome this, the focus has been shifted to genomic analysis of various EGFRdownstream signaling pathways. One of the vital EGFR downstream signaling pathways that perhaps provide roadmaps for targeted therapy is MAP kinase pathway. We aim to study key genes of MAP kinase pathway in patients with pulmonary adenocarcinomas (PADC).

      Methods:
      All PADC patients since 2015 were included after obtaining clinical details. Histomorphology was ascertained prior to mutational analysis. Real-Time PCR (EGFRexon 18–21 and KRASexon 2) and Sanger Sequencing (BRAFexon 11 and 15 and MEK1exon 2) technologies were used. The clinical and pathologic correlation was done.

      Results:
      Of 118 PADC patients (M: F-2.3:1), 26.2% (31/118) patients harboured EGFR mutations and were predominantly females. The most common EGFR mutation was exon 19 deletion. EGFR mutation positive cases showed mostly acinar histology. KRAS mutations (G12A) were observed in 21.1% (25/118) patients and were predominantly older males with mucinous histology. Four (3.3%) patients harboured coexisting EGFR and KRAS mutations. BRAF mutations (L597V, I953V) were seen in 2.5% (3/118) patients, however, there was no coexistence of BRAF mutations with other oncogenic drivers such as KRAS, EGFR mutations or ALK fusion. Additionally, BRAFpolymorphism was seen in 15.0% cases. None of the cases showed MEK1 mutation.

      Conclusions:
      This study examined the genetic alterations of genes involved in MAP kinase pathway in PADC of Indian patients. The frequency of KRAS mutation is higher than reported previously in the literature. Secondly, mutations in EGFR and KRAS showed coexistence. The high rate of KRAS mutations and coexistence of genomic alterations in Indian patients should be validated in a large number of cases for better patient management.

      Clinical trial identification:


      Legal entity responsible for the study:
      AIIMS, New Delhi, India

      Funding:
      ICMR, Lady Tata Trust, and AIIMS

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      63P - Prevalence and spectrum of germline mutations among patients with familial lung cancer (ID 228)

      12:30 - 12:30  |  Presenting Author(s): M. Kanwal

      • Abstract

      Background:
      Although lung cancer is generally thought to be environmentally provoked but rare familial forms of lung cancer has been described previously, suggesting there may be genetic susceptibility factors. However, due to the apparently sporadic nature of lung cancer, little attention has been paid to the role of genetic predisposition in lung cancer. To address this, we used FLC samples obtained from the Chinese population in highly air-polluted regions to screen for novel germline mutations in lung cancer.

      Methods:
      Through a whole genome sequencing (WGS) analysis of the nine subjects (four lung cancer patients and five normal family members of FLC), we obtained a whole genome dataset of DNA alterations in FLC samples. A total of 1218 genes were identified with mutations of multiple types. Subsequently, the top 12 highly mutated genes were selected for validation by PCR and DNA sequencing in an expanded sample set including FLC, sporadic lung cancer, and healthy population.

      Results:
      Mutations of the five genes (ARHGEF5, ANKRD20A2, ZNF595, ZNF812, MYO18B) may be potential germline mutations of lung cancer. We also analyzed specific mutations within the 12 genes and found that some specific mutations within the MUC12, FOXD4L3 and FOXD4L5genes showed higher frequencies in the samples of FLC and/or lung cancer tissue, compared with the healthy population. Moreover, some genes with copy number variation may be potentially associated with a predisposition to lung cancer.

      Conclusions:
      Our study uncovered the mutation spectrum in FLC of the Chinese population. Insights from this study will help direct further efforts to enhance our understanding of genetic predisposition in lung cancer. The investigation of novel and known gene mutations detected by the present study may contribute to evaluate functional impacts of these mutations not only in FLC but in sporadic lung cancer as well.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      National Science Foundation of China (81272617), the 973 Program (2011CB510104), and the Yunnan Province Science and Technology Department (Y103951111)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      64P - Cisplatin-resistant phenotype: Characterization, adaptation to EGFR signaling and sensitivity to EGFR inhibitors in NSCLC cells (Now Available) (ID 419)

      12:30 - 12:30  |  Presenting Author(s): V. Pamidiboina  |  Author(s): F. Tian, R. Kiefl, M. Schaule, R.M. Huber

      • Abstract
      • Slides

      Background:
      The emergence of platinum resistance is a significant obstacle to clinical management of lung cancer. We aimed to analyze the EGFR signaling and efficacy of EGFR inhibitors in acquired cisplatin resistance. Better understanding forms a basis for the development of novel combination therapies that could enhance patient survival.

      Methods:
      An isogenic clinical model was used to induce resistance in a panel of cell lines (H838, HCC827, H1975 and H1650 NSCLC cells) and H1339, an SCLC cell line. Cells were exposed to cisplatin (1 μg/ml/3 hrs/week) followed by recovery periods over of 4 weeks, and cisplatin–resistant phenotype (CRP) derived from original, age–matched naïve cells. They were then characterized by survival, proliferation, colony formation, and apoptosis. EGFR family receptors, phosphorylation and downstream signalling was assessed by EGFR phosphorylation and the PathScan Signaling array. The effects of EGFR TKIs (erlotinib, gefitinib, afatinib, and rociletinib) on CRP cells was evaluated at clinical concentrations.

      Results:
      CRP cells demonstrated increased survival, proliferation and resistance to apoptosis against the cisplatin challenge. CRP cells displayed altered expression of EGFR receptor family and their phosphorylation and critical nodes of signaling. But their appearance varied from cell line to cell line in comparison to their respective controls. The EGFR TKIs (except erlotinib on H838 cells) showed similar effects on CRP and their naïve cells.

      Conclusions:
      Our results identified CRP of NSCLC cells, which exhibited enhanced total EGFR and Met protein expression and their phosphorylation. This altered the expression of critical oncoproteins. The information can be used to design combination therapies with other TKIs to improve patient life.

      Clinical trial identification:


      Legal entity responsible for the study:
      Respiratory Medicine and Thoracic Oncology (LMU Klinikum der Universität München)

      Funding:
      German Center for Lung Research (DZL), Germany

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      65P - Fatty acid synthase (FASN) inhibition effect on EGFR TKIs sensitive and resistant cells (Now Available) (ID 574)

      12:30 - 12:30  |  Presenting Author(s): S. Ruíz Martínez  |  Author(s): J. Bosch-Barrera, M. Planas, L. Feliu, R. Porta-Balanya, T. Puig

      • Abstract
      • Slides

      Background:
      EGFR tyrosine kinases inhibitors (TKIs) are effective therapies for NSCLC patients whose tumors harbor an EGFR activating mutation, however, this treatment is not curative because of both primary and secondary resistance to EGFR TKIs. Several resistance mechanisms to EGFR-TKIs have been described, among which, there is the T790M mutation. A recently described mechanism of multidrug resistance is the FASN overexpression. FASN is a multifunctional enzyme essential for the endogenous synthesis of long-chain fatty acids.

      Methods:
      We have worked with the lung adenocarcinoma cell line PC9 carrying the EGFR TKI sensitizing exon 19 deletion (ΔE746-A750) and 3 gefitinib-resistant PC9 derived cell lines (2 T790M positive and 1 T790M negative). Cell viability was determined by the MTT method for Gefitinib, osimertinib and two FASN inhibitors (EGCG and G28). Apoptosis was assessed by PARP cleavage. Molecular effects were analyzed by studying changes in the expression and/or activation of EGFR signaling pathways by western blot. The effects of the double combination of EGFR TKIs and FASN inhibitors in all cell lines were studied and synergy between drugs was determined using CompuSyn software.

      Results:
      All cell lines have significantly different IC50 values for Gefitinib except for the two T790M positive ones. Osimertinib has a concentration-dependent cytostatic effect on sensitive cells. All the studied cell lines are sensitive to FASN inhibition. The cytotoxicity caused by FASN inhibition is independent of the T790M mutation that leads to gefitinib resistance. Apoptosis is induced in all cell lines at all treatments. While gefitinib and osimertinib activate the Signal Transducer and Activator of Transcription 3 (STAT3) in all cell lines, EGCG and G28 reduce the STAT3 activation found in all gefitinib-resistant cell lines. Combination experiments using FASN inhibitor G28 and EGFR TKIs show mostly additive effect and synergism at some concentrations.

      Conclusions:
      In summary, we show cytotoxicity effect of EGCG and G28 in NSCLC cells sensitive and resistant to EGFR TKIs probably due to inhibition of FASN/STAT3 signalling. FASN inhibition should overcome EGFR TKI resistance and may serve as a novel target therapy to improve EGFR-based cancer therapy in lung cancer.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Girona

      Funding:
      AstraZeneca

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      66P - PALB2 mRNA expression as a predictive marker in advanced non-small cell lung cancer (NSCLC) patients (p) treated with docetaxel-cisplatin (Now Available) (ID 405)

      12:30 - 12:30  |  Presenting Author(s): N. Karachaliou  |  Author(s): J. Berenguer, I. Chaib, J.L. Ramírez Serrano, M.T. Moran Bueno, B. Massuti Sureda, S. Calabuig Fariñas, C. Camps, M. Provencio Pulla, R. Rosell

      • Abstract
      • Slides

      Background:
      PALB2, the partner and localizer of BRCA2, binds directly to BRCA1 and is essential for homologous recombination repair and, henceforth, could influence the effect of docetaxel-cisplatin therapy. Previous studies have shown that PALB2 impedes the degradation of nuclear factor erythroid 2-related factor 2 (NRF2) through the binding and sequestration of its inhibitor Kelch-like ECH-associated protein (KEAP1). Over-expression of NRF2 could activate NOTCH signaling and lead to enrichment of cancer stem cells. In the current study, we examine the mRNA levels of PALB2 in advanced NSCLC p treated with docetaxel-cisplatin.

      Methods:
      We assessed PALB2 mRNA levels as potential biomarkers in tumor tissue from 177 cisplatin plus docetaxel-treated NSCLC p from the NCT00617656/GECP-BREC trial. The relationship of the PALB2 mRNA levels with the PFS, OS and response were examined.

      Results:
      Median age 62; 79.1% male; 51.4% adenocarcinoma. Results of PALB2 mRNA expression were as follows: PFS was 5.6 months (m) for p with high/intermediate (H-I) PALB2 and 4.1 m for p with low (L) PALB2 (p = 0.0018). OS was 13.2 m for p with H-I PALB2 compared to 9.9 for p with L PALB2 (p = 0.0377). In the univariate analysis, H-I PALB2 was a marker of longer PFS (HR = 0.56, 95% CI, 0.38, 0.80; p = 0.002) and OS (HR = 0.64, 95% CI, 0.41, 0.98; p = 0.0394). In the multivariate analysis, only H-I PALB2 was associated with longer PFS (here HR = 0.56 and p = 0.0022) and OS (HR = 0.61 and p = 0.0343). Among 143 p with data for response and PALB2 expression, 49.5% of p with H-I PALB2 were responders, compared to only 28% with L PALB2 (p = 0.0131).

      Conclusions:
      Higher PALB2 mRNA levels were associated with higher response, longer PFS and OS in NSCLC p treated with docetaxel-cisplatin. However, PALB2 could also be a readout of NRF2 activity and NOTCH signaling, indicative of an increase in cancer stem cells, providing hints for combinatory therapy with gamma secretase inhibitors to prevent the increase of stem-like cells and further improve outcome to docetaxel-cisplatin therapy. Experiments in NSCLC cell lines are ongoing.

      Clinical trial identification:


      Legal entity responsible for the study:
      IGTP, Germans Trias i Pujol Research Institute, Badalona, Barcelona, Spain

      Funding:
      Fundació Obra Social “La Caixa”

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      67P - The influence of circulating tumor DNA analysis on response to immunotherapy in non-small cell lung cancer (NSCLC) (ID 603)

      12:30 - 12:30  |  Presenting Author(s): R. Grinberg  |  Author(s): L.C. Roisman, S. Geva, L. Soussan-Gutman, A. Dvir, R. Yair, T. Twito, R. Larman, N. Peled

      • Abstract

      Background:
      International guidelines have advocated molecular profiling as part of the standard diagnostic evaluation for advanced NSCLC, with the goal of identifying driver mutations for which effective therapies or clinical trials are available. In advanced NSCLC, immunotherapy demonstrated good response rates with some responses being remarkably durable. Understanding the molecular determinants of response to immunotherapies is a key challenge in oncology. Notably, tumor mutational burden detected by tissue next generation sequencing (NGS) was found to be correlated with response to immune checkpoint inhibitors.

      Methods:
      In this retrospective study, data were collected on NSCLC patients treated in multiple medical centers in Israel between 2014 and 2017. We used NGS on cell-free circulating tumor DNA (ctDNA) to evaluate whether mutational burden influences the response to immunotherapy in these patients.

      Results:
      Overall, 336 NSCLC patients underwent NGS on ctDNA. Of these 336 patients, 192 (57%) were females and 144 (43%) were males. The average age (range) was 64 (23–103) years. Clinical treatment information is currently available for 117 patients, of whom 50 (43%) received immune check-point inhibitors. Rates of stable disease, partial and complete responses (RECIST criteria), as well as progression-free survival and overall survival will be reported. In addition, to unravel the genomic determinants of response to immunotherapy we will use the blood-derived ctDNA to understand if hypermutated ctDNA is a predictive biomarker of response to immunotherapy.

      Conclusions:
      ctDNA collection was feasible in 336 patients. Prediction model to associate the ctDNA signature with response to immunotherapy will be presented.

      Clinical trial identification:


      Legal entity responsible for the study:
      Soroka Medical Hospital

      Funding:
      Has not received any funding

      Disclosure:
      L.C. Roisman: Lectures fees from Roche, Astrazenca, MSD, Pfizer. S. Geva: Travel grant from Teva Pharmaceuticals, honorarium from Guardant Health. L. Soussan-Gutman, A. Dvir, R. Yair, T. Twito: Works at Oncotest-Teva, which is a distributor of Guardant Health in Israel. R. Larman: Employee by Guardant Health inc. N. Peled: Advisor & honorarium from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, FoundationMedicine, Gaurdant360, MSD, Novartis, NovellusDx, Pfizer, Roche, Takeda. All other authors have declared no conflicts of interest.

    • +

      68P - In vivo study of a novel chemoablative, thermoresponsive hydrogel for intratumoural administration in a murine A549 xenograft model (ID 280)

      12:30 - 12:30  |  Presenting Author(s): S.M. Rossi  |  Author(s): B.K. Ryan, H.M. Kelly

      • Abstract

      Background:
      Direct intratumoural (IT) injection of chemotherapeutics into solid tumours has potential to achieve high local concentrations, reducing systemic toxicity and off target side effects. However, this approach is limited by rapid drug clearance from the tumour, resulting in inaccurate and unpredictable dosing. Thermoresponsive hydrogels undergo a characteristic phase change in response to temperature. This allows for minimally invasive administration of a liquid via needle/catheter into the tumour, with localisation and retention following gelation at physiological temperatures. Drug loading of hydrogels facilitates localised, controlled delivery of chemotherapeutics to the tumour, with reduced off site toxicity.

      Methods:
      Preclinical assessment of blank and drug loaded formulations was conducted in a murine A549 xenograft model (approved by the RCSI ethics committee). 1 × 10[6] A549-luciferase cells were subcutaneously injected into the right flank of female Athymic Nude-Foxn1[nu] mice (n = 6 per group). At a tumour volume of approx. 250 mm[3], 100 μL of hydrogel (blank or drug loaded) or saline was injected IT using a 22G needle. Tumour growth was assessed using callipers and an IVIS® Spectrum in vivo imaging system over 14 days. On day 14, mice were sacrificed; tumours, liver and kidney excised, fixed and embedded in paraffin for histological and immunohistochemistry studies.

      Results:
      Two hours after IT injection, IVIS® imaging indicated localisation and retention of the hydrogel at injection site. At day 7 and 14 following blank or drug loaded hydrogel treatment, a significant reduction in tumour growth was observed compared to saline (p = 0.001). Retention of the hydrogel at the tumour site for up to 14 days was observed in ex vivo tumour tissue. No change in welfare scoring was observed during the study, with 100% survival at day 14, indicating treatment did not result in acute off site toxicity.

      Conclusions:
      Preliminary preclinical studies have shown the blank and drug loaded hydrogels significantly reduce tumour growth. Further studies are required to fully elucidate the underlying mechanism of action. IT administration of this formulation may represent a potential new adjuvant approach in lung cancer treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Royal College of Surgeons in Ireland

      Funding:
      Enterprise Ireland

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      69P - The diabetes drug canagliflozin sensitizes non-small cell lung cancer (NSCLC) to radiotherapy and chemotherapy (ID 494)

      12:30 - 12:30  |  Presenting Author(s): T. Tsakiridis  |  Author(s): L. Villani, L. Broadfield, K. Marcinko, E. Tsakiridis, P. Ellis, P. Muti, G. Steinberg

      • Abstract

      Background:
      Radiotherapy (RT) and chemotherapy (ChT) are key treatments for NSCLC, but this disease is highly resistant to cytotoxic therapy. Metabolism determines tumor cell survival and response to therapy. Oncogenic pathways like EGFR- > K-Ras- > mTOR stimulate glycolysis, protein and lipid synthesis (lipogenesis). This is balanced by the action of metabolic stress sensors like AMP-activated kinase (AMPK), which is activated by stressors, including inhibition of mitochondrial oxidative phosphorylation (OxPhos) cascade. AMPK mediates energy conservation through a global regulation on metabolism that includes suppression of lipogenesis, protein synthesis and mTOR. Canagliflozin is an approved and effective diabetes drug that blocks the Na-glucose co-transporter 2 (SGLT2), which controls glycemia by blocking glucose re-uptake in the kidney. However, we found that canagliflozin also inhibits mitochondria OxPhos Complex I and activates AMPK, leading to blockade of lipogenesis and a significant anti-tumor activity in NSCLC.

      Methods:
      Adenocarcinoma A549 and H1299 NSCLC cells were subjected to canagliflozin, RT and cisplatin ChT treatments and were analyzed with proliferation, clonogenic survival and immunoblotting experiments.

      Results:
      We report that canagliflozin enhances the response of NSCLC cells to RT and ChT. Canagliflozin, but not other clinically used SGLT2 inhibitors, like Empagliflozin or Dapagliflozin, suppresses proliferation and clonogenic survival of A549 and H1299 cells and enhances their response to RT and cisplatin ChT. Importantly, canagliflozin mediates these effects at clinically achievable low micromolar doses (5–30 mM). These effects are associated with potent inhibition of OxPhos complex I, phosphorylation and activation of AMPK, inhibitory phosphorylation of Acetyl-CoA carboxylase (ACC) and suppression of the mTOR pathway.

      Conclusions:
      This work suggests that canagliflozin is a promising metabolism-targeting therapy that may be able to improve clinical RT/ChT responses in NSCLC. Being, a well-tolerated, approved and widely used medication, canagliflozin may be able to enter early phase NSCLC trials in the near future.

      Clinical trial identification:


      Legal entity responsible for the study:
      McMaster University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      70P - Immunological efficacy of anti-tumor dendritic cell vaccine application in patients with non-small cell lung cancer (ID 410)

      12:30 - 12:30  |  Presenting Author(s): O. Gorbach  |  Author(s): N. Khranovska, O. Skachkova, M. Inomistova, V. Sovenko, A. Ganul, V. Orel

      • Abstract

      Background:
      Immunotherapy using dendritic cells (DC) represents a novel and promising therapeutic method for patients with non-small cell lung cancer (NSCLC). The aim was to examine the molecular and cellular mechanisms of anticancer immune responses during immunotherapy as well identify the immune parameters that may predict DC vaccine therapy efficiency.

      Methods:
      One hundred patients with IIB-IIIA stage NSCLC were enrolled into the study. Patients were randomly allocated into two groups: 1st – patients received DC-vaccine after surgery, 2nd – received surgery only. Original construction of DC vaccine has been used: autologous DCs with mechanically heterogenized microparticles of tumor cells. Patients received four intravenous injections with one-month interval and immunological monitoring was performed before each injection.

      Results:
      The most notable changes in cell-mediated immune response was observed after the 4[th] injection of DC vaccine. Namely, the balance of Th1- and Th2-mediated immune response was changed, in particular the number of CD3[+]IFN-γ[+] lymphocytes prevailed over CD3[+]IL-4[+] lymphocytes. Moreover, the CD4[+]CD45RO[+] memory cell numbers increased in peripheral blood and the TGF-β mRNA expression level was significantly decreased in circulating leucocytes (p < 0.05) compared with that in patients without DC-vaccine treatment. According to Cox proportional hazard model, changes in the number of CD4[+]CD45RO[+] memory cells (p < 0.05), TGF-β mRNA expression level (p < 0.02) and ratio of CD3[+]IFN-γ[+] lymphocytes to CD3[+]IL-4[+] lymphocytes (p < 0.02) allow us reliably to determine the development of relapses in patients with NSCLC. Using ROC analysis, we established the optimal criteria for these immunological markers, which can separate groups by low and high risk of NSCLC recurrence: <0.74 for ratio of CD3[+]IFN-γ[+] lymphocytes to CD3[+]IL-4[+] lymphocytes (p < 0.001, AUC = 0.92, Se = 100%, Sp = 75%); ≤0.62 × 10[3] cells/mL for CD4[+]CD45RO[+] memory cells (p < 0.001, AUC = 0.91, Se = 100%, Sp = 82%); >0.034 a.u. for TGF-β mRNA expression (p < 0.05, AUC = 0.73, Se = 67%, Sp = 100%).

      Conclusions:
      Immunological markers with prognostic significance regarding the duration of the event-free period have been established in patients with NSCLC after DC vaccine treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      National Cancer Institute of the MPH Ukraine

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      73P - PET and neck US for the detection of cervical lymphadenopathy in patients with lung cancer and mediastinal lymphadenopathy (ID 420)

      12:30 - 12:30  |  Presenting Author(s): M. Ahmed

      • Abstract

      Background:
      Cervical lymph nodes are frequently involved in patients with lung cancer and indicate inoperability. Some guidelines recommend neck ultrasound (NUS) in patients with bulky mediastinal lymphadenopathy. Positron emission tomography (PET) is indicated for patients with potentially curable disease. We aimed to assess the diagnostic yield of NUS and the diagnostic accuracy of PET for cervical lymphadenopathy among patients with suspected lung cancer and mediastinal lymphadenopathy.

      Methods:
      Records of all patients with lung cancer who underwent a NUS over a consecutive 5-year period were reviewed. Only patients with mediastinal lymphadenopathy on CT were included. The diagnostic accuracy of PET was assessed with NUS-guided fine needle aspiration cytology used as the reference test.

      Results:
      During the study period 123 patients met the inclusion criteria. Malignant cervical lymphadenopathy was confirmed in 39.8% (95% CI 31.1–49.1%). PET-CT had a specificity of 81.1%, sensitivity of 87.5%, negative predictive value of 96.8% and positive predictive value of 50% for the detection of cervical lymphadenopathy, and it contributed no additional staging information in the neck area. Overall, PET led to changes in management in only 2.2% of cases.

      Conclusions:
      A significant proportion of patients with lung cancer and mediastinal lymphadenopathy have cervical lymphadenopathy detected by NUS. In this group of patients PET offers minimal additional value in staging and management.

      Clinical trial identification:


      Legal entity responsible for the study:
      Galway University Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      74P - Spatial concordance of tumor proliferation and accelerated repopulation from pathologic images to 18F-FLT PET images: A basic study guided for PET-based radiotherapy dose painting (ID 158)

      12:30 - 12:30  |  Presenting Author(s): C.M. Li  |  Author(s): X. Meng

      • Abstract

      Background:
      PET imaging with [18]F-fluorothymidine ([18]F-FLT) can potentially be used to identify tumor subvolumes for selecting dose escalation in radiation therapy. The aim of this study was to monitor tumor cell proliferation and repopulation during fractionated radiotherapy and investigate the spatial concordance of tumor cell proliferation and repopulation with [18]F-FLT tracer uptake.

      Methods:
      Mice bearing A549 xenograft tumors were assigned to 5 different irradiated groups (3f/6d, 6f/12d, 9f/18d, 12f/24d and 18f/36d) with 2 Gy/fractions and non-irradiated group. Serial [18]F-FLT micro PET scans were performed at different time points, the maximum of standard uptake value (SUVmax) were measured to detect the feasible time of tumor repopulation during irradiation. Ex vivo images of the spatial pattern of intratumor [18]F-FLT uptake and Ki-67 labeling index (LI) were obtained from thin tumor tissue sections. A layer-by-layer comparison between SUVmax and Ki-67 LI results, including the thresholds at which maximum overlap occurred between FLT-segmented areas and areas of active cell proliferation, were conducted to evaluate the spatial imaging pathology correlation.

      Results:
      The SUVmax were observed decreases in the 3f/6d group (P = 0.000), compared to these for non-irradiated tumors. However, it was significantly increased in the 6f/12d later, and then gradually reduced with treatment time prolonged again after 6f/12d group. Proliferation changes on pathology imaging at 6f/12d were also confirmed. Significant correlations were found between the SUVmax and Ki-67 LI of all ROIs in each in vitro tumor of cell proliferation group (Ps < 0.001). Similar results were also found in each tumor of accelerated repopulation group (Ps < 0.001). Furthermore, both the mean ORRs were more than 50% in all layer of the tumor cell proliferation and accelerated groups. Regions of high-intensity [18]F-FLT uptake in the autoradiographs exhibited prominent staining for Ki-67.

      Conclusions:
      [18]F-FLT PET may be a promising imaging surrogate of tumor proliferative response to fractionated radiotherapy and might help make adaptive radiation oncology treatment plan.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shandong Cancer Hospital

      Funding:
      National Natural Science Foundation of China (81472810) and the Science and Technology Development Plans of Shandong Province (2014GSF118138)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      76P - Robustness of radiomic features in [18F]-FDG PET/CT and [18F]-FDG PET/MR (ID 304)

      12:30 - 12:30  |  Presenting Author(s): D. Vuong  |  Author(s): M. Bogowicz, M. Huellner, P. Veit-Haibach, N. Andratschke, J. Unkelbach, M. Guckenberger, S. Tanadini-Lang

      • Abstract

      Background:
      Radiomics is a promising tool for identification of new prognostic biomarkers. However, image reconstruction settings may affect the absolute values of radiomic features, which reduces their value as reliable biomarkers. PET/MR is becoming more available and often replaces PET/CT. The aim of this study was to quantify to what extend [18F]-FDG PET/CT radiomics models can be transferred to [18F]-FDG PET/MR.

      Methods:
      Nine patients with non-small cell lung cancer underwent first an [18F]-FDG PET/MR scan followed by an [18F]-FDG PET/CT scan (SIGNA PET/MR and Discovery PET/CT 690, GE Healthcare) with a delay time of 38 min +/−5 min. Patients had one single FDG injection for both scans. The primary tumors were segmented independently on the PET scans from PET/CT and PET/MR with two semi-automated methods (gradient-based and threshold-based). Resampling was performed to the lowest resolution. In total, 1358 radiomic features were calculated, i.e. shape (18), intensity (17), texture (137), wavelets (1186). The intra-class correlation coefficient was used to compare the radiomic features in both image modalities. An ICC >0.9 was considered stable among both types of PET scans.

      Results:
      The median relative volume difference of the tumors segmented on PET/CT and PET/MR was 4.8% (range 0.4–39.9%) for the gradient-based and 18.0% (range 0.7–71.2%) for the threshold-based method. A larger number of radiomic features was stable using the gradient-based method compared to the threshold-based method, which concurs with the improved reproducibility of tumor volume using gradient-based method. More than 70.6% of shape and intensity features yielded an ICC >0.9 among both segmentation methods. However, only 51.5% of texture and 27.2% of wavelet features reached this criterion (for gradient-based and even less in threshold-based method). In the wavelet features analysis, more features were robust in smoothed images (low-pass filtering) in comparison to images with emphasized heterogeneity (high-pass filtering).

      Conclusions:
      Shape and intensity radiomic features were robust comparing the two types of [18F]-FDG PET scans (PET/CT and PET/MR). In contrast, texture and wavelet features showed reduced stability, which needs to be considered for their use in prognostic modelling.

      Clinical trial identification:


      Legal entity responsible for the study:
      Dr. Stephanie Tanadini-Lang

      Funding:
      Has not received any funding

      Disclosure:
      M. Guckenberger: Committee Member EORTC, ESTRO Board of Directors, Head of Working Group DEGRO. All other authors have declared no conflicts of interest.

    • +

      77P - Is there an incremental benefit with 68 Ga DOTA PET/CT in staging of broncho-pulmonary carcinoid tumors? (ID 615)

      12:30 - 12:30  |  Presenting Author(s): G. Karimundackal

      • Abstract

      Background:
      The staging of well differentiated bronchopulmonary neuroendocrine tumours (carcinoids) is complicated by the unpredictable incidence of nodal and distal metastases. PET CECT which has become the standard for staging of lung cancers has proven ineffective in the staging of carcinoid tumours. 68 Ga DOTA NOC PET/CT which depends on radio-tracer uptake in somatostatin receptors appears to be an attractive modality for the staging of these neuroendocrine tumours.

      Methods:
      We performed a retrospective analysis of patients who underwent 68 Ga DOTA NOC PET/CT followed by surgical resection from October 2014 to November 2017. Data was retrieved regarding demographics, standardized uptake value (SUV), surgery performed and final histopathology report including degree of differentiation, nodal positivity and Mib index. The study included only patients who underwent resection since the focus of the study on corelation with histopathological features. An attempt was made to corelate the SUV with diagnosis of typical vs atypical carcinoid, nodal metastases and Mib index.

      Results:
      During the study period 38 patients underwent surgical resection following DOTA PET. All details of imaging including SUV were available for 35 patients, while complete histopathological details were available for 37 patients. DOTA PET was not able to differentiate between typical and atypical carcinoids (32 vs 3) based on SUV. The mean SUV of typical carcinoids was 41.4(SD- 41.2) whereas that of atypical carcinoids was 34.4(SD-39.4) and the difference was not statistically significant (p = 0.7). 6/37 cases had nodal metastases of which DOTA PET could corrrectly identify only 1. There were 2 false positive reports and 5 false negative reports, (sensitivity 16.6% and specificity 93.5%). The mean SUV of the false positive nodes (n = 2 cases) was 7 which calls into question the specificity of the tracer. No corelation between SUV and Mib index could be demonstrated, p = 0.37.

      Conclusions:
      In this study 68 Ga DOTA NOC PET CECT could not help in predicting the histopathological nature of carcinoid tumours, atypical vs typical, presence of nodal metastases or Mib index. This study is limited by the small numbers enrolled, a larger series may help us incoming to a more definitive conclusion.

      Clinical trial identification:


      Legal entity responsible for the study:
      Tata Memorial Hospital

      Funding:
      Has not received any funding

      Disclosure:
      The author has declared no conflicts of interest.

    • +

      79P - Small cell lung cancer in the young: Characteristics, timeliness and outcome data (Now Available) (ID 334)

      12:30 - 12:30  |  Presenting Author(s): S. Jiang  |  Author(s): Y. Han, Y. Huang, X. Hao, J. Li, X. Hu, Z. Xiao, H. Wang, Y. Wang, Y. Shi

      • Abstract
      • Slides

      Background:
      Patients with small cell lung cancer (SCLC) younger than 40 are limited in number and are understudied. Our research aimed to assess the characteristics, timeliness of cancer development, and outcomes of this patient population.

      Methods:
      Records of Patients under the age of 40 with SCLC at the Chinese Academy of Medical Sciences between January 2006 and December 2015 were reviewed and evaluated.

      Results:
      One hundred and three patients (67.0% limited-stage, 33.0% extensive-stage) were included, along with 54 (52.4%) never-smokers. The median duration of the diagnostic interval and the median survival time (MST) were 51.0 days and 24.0 months, respectively. Survival was significantly better for limited-stage patients than for those in the extensive-stage group (MST, 28.0 vs. 14.0 months, p < 0.0001). Patients with shorter diagnostic times (≤1 month) tended to have a better prognosis than those with a longer delay to diagnosis (MST, 25.0 vs. 23.0 months, p = 0.061), which was more pronounced in the limited stage group (MST, 48.0 vs. 26.0 months, p = 0.043). A timely diagnosis also exerted a positive impact on progression-free survival (PFS) in both the limited-staged patients (36.0 vs. 13.0 months, p = 0.061) and extensive-staged patients (6.0 vs. 3.0 months, p = 0.047). Further analysis identified a correlation between the antibiotic treatment before SCLC diagnosis and the diagnostic interval (Spearman Rho = 0.294, p = 0.003). Multivariate analysis suggested that empirical antibiotherapy history, disease stage, and tumor location independently correlated with survival.

      Conclusions:
      Our study identified distinct characteristics of patients with SCLC under 40. More timely care could improve patient outcomes especially among those at a limited-stage.

      Clinical trial identification:


      Legal entity responsible for the study:
      National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      80P - Understanding the patient perspective of small cell lung cancer (SCLC) (Now Available) (ID 265)

      12:30 - 12:30  |  Presenting Author(s): A. Rydén  |  Author(s): S. Ollis, E. Love, A.L. Shields, H. Jiang, P. Dennis

      • Abstract
      • Slides

      Background:
      When developing a patient-reported outcomes assessment strategy, empirical evidence based on literature review and therapeutic area experts should be complemented with input directly from patients to reliably reflect the patients’ experiences. Therefore, a set of research activities were initiated to identify, describe, and substantiate concepts that reflect the important and relevant symptoms, treatment-related side effects, and impacts of extensive stage (ES)-SCLC.

      Methods:
      Twenty-five articles were reviewed, four clinical experts participated in advice meetings and 17 US patients diagnosed with ES-SCLC were interviewed. Trained interviewers used a semi-structured interview guide to elicit information from patients. Interviews were transcribed, coded, and analysed using qualitative data analytic methods.

      Results:
      Patients’ mean age was 65.2 years (SD = 9.8) and patients self-reported as white (n = 11, 64.7%), black or African American (n = 5, 29.4%), and American Indian or Alaska Native (n = 1, 5.9%). Thirty-one signs/symptoms of ES-SCLC were elicited with fatigue being mentioned by most patients (n = 10). Also, the most frequently reported SCLC symptoms were shortness of breath (n = 6, 35.3%), pain (n = 5, 29.4%), and weight loss (n = 4, 23.5%). Patients also reported numerous ways that their lives were impacted by ES-SCLC (n = 68 concepts), and treatment-related side-effects they experienced (n = 41 concepts). These findings, along with evidence from literature review and clinical experts, showed that shortness of breath, pain, weight loss, cough, fatigue, nausea, paraneoplastic syndromes, haemoptysis, and loss of appetite were reported as signs and symptoms of SCLC in all three research activities.

      Conclusions:
      This study provides an overview of symptoms, treatment-related side effects, and impacts associated with ES-SCLC from the patient perspective. The results suggest that patients with SCLC have similar key symptoms to patients with non-small cell lung cancer, therefore similar measurement strategies can be applied. Using this evidence, recommendations can be made for ES-SCLC clinical trial outcomes that resonate with primary stakeholders including patients, healthcare providers, regulatory agencies, and payers.

      Clinical trial identification:
      Not applicable.

      Legal entity responsible for the study:
      AstraZeneca PLC

      Funding:
      AstraZeneca

      Disclosure:
      A. Rydén, H. Jiang, P. Dennis: AstraZeneca: full-time employment and stock ownership. S. Ollis, E. Love, A.L. Shields: Employee of Adelphi Values, which receives payment from pharmaceutical companies to conduct outcomes-based research.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      81P - The influence of comorbidity on health utility score (HUS) and health-related quality of life (HRQoL) in small cell lung cancer (SCLC) compared to non-small cell lung cancer (NSCLC) (ID 531)

      12:30 - 12:30  |  Presenting Author(s): A. Vedadi  |  Author(s): S. Shakik, M..C. Brown, W. Xu, F.A. Shepherd, N. Leighl, P.A. Bradbury, G. Liu, G. O'Kane

      • Abstract

      Background:
      A significant number of patients with SCLC suffer from comorbidities. In particular, heart failure and chronic obstructive pulmonary disease are often present. Comorbidity alone has been validated as an independent negative survival prognostic factor in SCLC. Despite this, comorbidities are rarely systematically assessed in relation to quality of life. Here, we examine the effect of comorbidity on HUS and quality of life in SCLC in relation to NSCLC.

      Methods:
      Histologically confirmed SCLC or NSCLC, were recruited from the Princess Margaret Cancer Centre. Demographics, treatment toxicity, and Patient Reported Charlson Comorbidity Index (PRO-CCI) were collected. For statistical analysis, univariable linear regression was applied to evaluate each variable and its association with HUS. A multivariable model with a backwards selection algorithm of univariable factors associated with HUS was employed.

      Results:
      Comorbidiity differentially drives HUS in SCLC compared to NSCLC (interaction p < 0.0001) and is an independent negative predictor of HRQoL (Table). Specifically, heart failure and COPD were two main modulators in SCLC quality of life (p < 0.05). Other prognostic factors, notably treatment-related symptom severity and performance status at diagnosis, did not differentially modulate HUS (Table). SCLC patients had a significantly higher number of comorbidities and lower HUS relative to NSCLC; nonetheless, comorbidity had the greatest magnitude of inverse relationship with HUS.

      Interaction TermsInteraction p-valueSCLCNSCLC
      CCI<0.0001−0.09 (−0.01, −0.06)−0.02 (−0.05, 0.01)
      Average severity of symptoms0.70−0.04 (−0.06, −0.02)−0.04 (−0.05, −0.02)
      Performance status at diagnosis0.53−0.06 (−0.11, −0.02)−0.08 (−0.12, −0.05)


      Conclusions:
      Comorbidity differentially modulates HUS in SCLC compared to NSCLC and is an important criterion to evaluate prior to treatment. We suspect that extensive smoking histories in the SCLC population may be driving these differential effects on HUS (Table). Interactions between factors differentially modulating HUS in SCLC and NSCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Princess Margaret Hospital (University Health Network)

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      82P - Chemotherapy versus combined chemoradiotherapy: Survival patterns among patients with extensive disease small cell lung cancer (ID 283)

      12:30 - 12:30  |  Presenting Author(s): I. Uthman  |  Author(s): M. Hassan

      • Abstract

      Background:
      Extensive Disease Small Cell Lung Cancer (ED-SCLC) represents 30% of lung cancer. While surgery is not an option, chemotherapy is traditionally currently recommended as the standard treatment. As oncologists seek superior modality for better survival, we conducted this study to evaluate the efficacy of combined chemotherapy and thoracic radiotherapy in improving the survival of patients with ED-SCLC, and to identify the characteristics of best candidates.

      Methods:
      Using the SEER database, we extracted the data of 5443 patients with ED-SCLC from 2010 to 2013. There were 2665 (49%) patients who underwent combined chemotherapy and thoracic radiotherapy, while 2778 (51%) of them received chemotherapy only. Patients’ demographics and TNM classification were assessed.

      Results:
      Patients with ED-SCLC had better outcomes when they received combined chemoradiation, compared with those who received chemotherapy alone, where overall 1-year relative survival rates were 40.2% and 24.1%, respectively (p value <0.001). All patients’ characteristics favor combination over chemotherapy except those aged below 40 years and those over 80 years. Subgroup analysis revealed better survival rates among female patients, patients aged between 40 and 59 years, and those with metastasis to the other lung or free regional lymph nodes. On the other hand, no advantage was allocated to those of specific race or primary site.

      Survival of Patients
      VariablesChemoradiationChemotherapyp value
      Sex:
       Male37.1%21.5%<0.001**
       Female43.9%27.0%<0.001**
      Age:
       20–3928.6%80.1%0.914
       40–5943.3%25.0%<0.001**
       60–7940.0%24.3%<0.001**
       ≥8021.8%19.6%0.6
      Race:
       White39.9%23.7%<0.001**
       Black42.5%27.7%0.002**
       Others40.9%25.4%0.002**
      Primary Site:
       Upper Lobe40.3%24.3%<0.001**
       Middle Lobe47.5%22.6%0.001**
       Lower Lobe38.4%23.9%<0.001**
       Overlapping Lesion44.5%25.4%0.01*
      Lymph Nodes Involvement:
       No Nodes Involved53.6%,35.5%<0.001**
       Single Ipsilateral Node46.8%31.6%<0.001**
       Multiple Ipsilateral Nodes37.5%22.3%<0.001**
      Distant Metastasis:
       Lung37.5%,21.0%<0.001**
       Liver30.2%19.2%<0.001**
       Brain31.5%22.4%<0.001**
       Bone31.6%21.2%<0.001**


      Conclusions:
      Combined chemoradiation in patients with ED-SCLC shows higher 1-year relative survival rates when compared with chemotherapy alone. Further studies should be conducted to weigh the efficacy versus safety to consider using chemoradiation as an alternative modality for ED-SCLC.

      Legal entity responsible for the study:
      Inas Uthman

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      83P - Radiotherapy improves the survival of patients with extensive disease small cell lung cancer: A propensity matched analysis of surveillance, epidemiology and end results database (Now Available) (ID 281)

      12:30 - 12:30  |  Presenting Author(s): R. Zhang  |  Author(s): P. Li, Q. Li, Y. Qiao, T. Xu, Q. Song, Z. Fu

      • Abstract
      • Slides

      Background:
      The survival advantage of radiotherapy (RT) for patients with extensive disease small cell lung cancer (ED-SCLC) has not been adequately evaluated.

      Methods:
      We analyzed stage IV SCLC patients enrolled from a large data from the Surveillance, Epidemiology, and End Results (SEER) registry through January 2010 and January 2012. Propensity score (PS) analysis with 1:1 matching and the nearest neighbor matching method were performed to ensure well-balanced characteristics of all comparison groups. Kaplan-Meier and Cox proportional hazardous model were used to evaluate the overall survival (OS), cancer-specific survival (CSS), and corresponding 95% confidence interval (95%CI).

      Results:
      Overall, for all metastatic ED-SCLC, receiving radiotherapy was associated with both improved OS and CSS. Generally speaking, radiotherapy for thoracic lesion and any metastatic sites could significantly improve the OS and CSS, except for brain metastasis. Before PS matching, radiotherapy significantly improved the survival of ED-SCLC patients with metastasis to brain (OS, HR = 0.71, with 95% CI = 0.61–0.83), however, the OS improvement became insignificant after PS matching (OS, HR = 0.91, with 95% CI = 0.74–1.12). For those M1a-SCLC patients without pleural effusion, radiotherapy, most likely to the primary site, also significantly improved the survival (P < 0.001). Furthermore, for those ED-SCLC patients with ≥ 2 metastatic sites, i.e., poly-metastatic ED-SCLC patients, radiation significantly improved the median OS from 6.0 months to 8.0 months (P = 0.015) and the median CSS from 7.0 months to 8.0 months (P = 0.020).

      Conclusions:
      The results suport radiotherapy in addition to chemotherapy might improve the survival of patients with metastatic ED-SCLC in a PS-matched patient cohort from the large SEER database. Although it should be adequately studied in phase III trials, it is prudent to select patients for radiotherapy in metastatic ED-SCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      Dr. Zhenming Fu, Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China

      Funding:
      National Science Foundation of China (NSFC)

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      84P - Patterns of care for patients with small cell lung cancer: A survey of German radiation oncology institutions on recommendations for prophylactic cranial irradiation (Now Available) (ID 471)

      12:30 - 12:30  |  Presenting Author(s): C. Eze  |  Author(s): O. Roengvoraphoj, M. Dantes, R. Abdo, N. Schmidt-Hegemann, C. Belka, F. Manapov

      • Abstract
      • Slides

      Background:
      Prophylactic cranial irradiation in SCLC was first proposed in 1973 and is recommended for patients demonstrating good response to initial therapy. The general pattern of care amongst radiation oncologists in Germany has not been previously evaluated. We conceptualized and conducted this survey to assess patterns of care.

      Methods:
      We surveyed radiation oncology institutions in Germany via an online questionnaire sent by e-mail to member institutions of the German Society for Radiation Oncology (DEGRO e.V.). The questions were based on respondent and treatment characteristics with emphasis on prophylactic cranial irradiation in limited- and extensive-stage SCLC.

      Results:
      We received a total of 95 responses (29% response rate). Of which 64 were completed and returned and hence eligible for evaluation. Sixty-one percent of respondents were between the ages of 50–59, 88% with over 15 years of experience in the management of lung malignancies. Sixty-seven percent of the institutions stage their patients initially with 18F-FDG-PET/CT. Of the 64 responses, 97% recommended delivery of PCI in therapy responders with LS-SCLC compared to 67% with ES-SCLC. Interestingly, a quarter of respondents offered hippocampal-avoidance PCI with only 38% following their patients with serial brain imaging following PCI. Neuropsychological testing was generally not routinely performed.

      Conclusions:
      German radiation oncology institutions showed interesting variations in certain aspects yet in general congruence in SCLC management and PCI delivery in accordance with the updated national and international guidelines. Future randomized clinical trials will further reduce discrepancies regarding delivery of PCI especially in ES-SCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital, LMU Munich

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      85P - Prophylactic cranial irradiation in patients with extensive-stage small cell lung cancer (ES-SCLC): A tertiary cancer centre experience (Now Available) (ID 381)

      12:30 - 12:30  |  Presenting Author(s): A. Srivastava  |  Author(s): K. Shelly, F. Lin

      • Abstract
      • Slides

      Background:
      Prophylactic cranial irradiation (PCI) remains the standard of care in patients who respond to first-line chemotherapy for ES-SCLC, although several recent studies have questioned its benefit with regard to survival.

      Methods:
      To see whether PCI was associated with improved survival in ES-SCLC patients, we conducted a retrospective study at a regional cancer centre in New Zealand. All ES-SCLC patients who had at least one cycle of chemotherapy from 2001 through April 2017 were included. The available medical records were reviewed to extract clinical variables: age, sex, comorbidities, metastatic sites, chemotherapy [regimen, and number of chemotherapy lines (NCL)], radiotherapy [palliative, consolidation chest radiotherapy (CCRT), or PCI], and overall survival (OS). Univariate analysis stratified by PCI status (received vs. not received) was performed. Stepwise Cox's regression was used to identify and adjust for confounders that affect survival by PCI status. The subgroups were examined by Kaplan-Meier method.

      Results:
      327 ES-SCLC patients who received ≥1 cycle of chemotherapy were identified; of the 245 patients with available records, median OS was 7.7 months (95% CI: 5.7–8.6 months). In 49 patients (20%) who received PCI, the median OS was 14.3 months (95% CI: 11.5–17.4), compared to 6.3 months (95% CI: 5.2–7.3) in patients who did not receive PCI (unadjusted hazard ratio (HR) for death: 0.34, 95% CI: 0.24–0.48). After adjusting for three covariates (NCL, whether ≥4 cycles of first-line chemotherapy was given, and whether CCRT was delivered), however, difference in survival was no longer significant (adjusted HR: 0.66, 95% CI: 0.42–1.04). No difference was observed in OS among subgroups. 85 patients (35%) who had <4 cycles of first-line chemotherapy (and did not receive more chemotherapy) had a dismal prognosis (median OS: 2.9 months, 95% CI: 1.8–3.4).

      Conclusions:
      Though ES-SCLC patients treated with PCI appear to have a better survival, but this positive association is not independent of treatment characteristics. Without further data from randomised trials, PCI should be used judiciously considering its toxicities and detriments to quality of life.

      Clinical trial identification:


      Legal entity responsible for the study:
      Waikato District Health Board

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      86P - Prophylactic cranial irradiation in SCLC: A survey of German radiation oncology institutions on recommendations for brain imaging (Now Available) (ID 573)

      12:30 - 12:30  |  Presenting Author(s): C. Eze  |  Author(s): O. Roengvoraphoj, M. Dantes, R. Abdo, N. Schmidt-Hegemann, C. Belka, F. Manapov

      • Abstract
      • Slides

      Background:
      Prophylactic cranial irradiation in SCLC is recommended in therapy responders to primary treatment. The landmark PCI trials largely took place in the pre-MRI era. Recently, the role of brain imaging in PCI has come under increased scrutiny. We conducted this survey to gauge the patterns of brain imaging in PCI amongst radiation oncologists in Germany.

      Methods:
      We surveyed radiation oncology institutions in Germany via an online questionnaire sent by e-mail to member institutions in the online directory of the German Society for Radiation Oncology (DEGRO e.V.). The questions covered aspects of brain imaging in limited- and extensive-stage SCLC.

      Results:
      We received a total of 39 responses. Of which 34 were completed and eligible for further evaluation. 97% and 92% of respondents recommended brain imaging prior to initial treatment in LS- and ES-SCLC, respectively with contrast-enhanced MRI being the preferred modality in 86% of cases. Ninety-one percent of respondents recommended brain imaging not older than 4 weeks prior to PCI delivery with 71% recommending commencing PCI <4 weeks after primary treatment. Of all responses, 38% followed their patients regularly with serial brain imaging following PCI with the absolute majority preferring image acquisition only in symptomatic patients. On detection of brain metastases (BMs), 59% recommended stereotactic radiosurgery (SRS) or repeat whole brain radiotherapy depending on the number of BMs.

      Conclusions:
      Patterns of brain imaging in Germany are in general consistent with the guidelines with contrast-enhanced MRI being the preferred modality for brain staging prior to primary treatment and especially after multimodality treatment and before PCI delivery.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital, LMU Munich

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      87P - Assessment of the prognostic utility of the enumeration of circulating tumour cells at the end of treatment and disease relapse in small cell lung cancer patients (Now Available) (ID 612)

      12:30 - 12:30  |  Presenting Author(s): A.J. Vickers  |  Author(s): A. Xenofontos, L. Carter, R. Bernabe, M. Carter, L. Franklin, K. Morris, J. Pierce, C. Dive, F. Blackhall

      • Abstract
      • Slides

      Background:
      Small cell lung cancer (SCLC) is an aggressive cancer characterised by early invasion and metastasis, the rapid emergence of treatment resistance and poor prognosis. Previous research has demonstrated that the number of circulating tumour cells (CTCs) at baseline is prognostic in SCLC, associated with poorer overall survival (OS). Our aim was to explore the prognostic utility of the quantification of CTCs at the end of treatment (EoT) and the development of disease relapse, in addition to baseline.

      Methods:
      This was a single-centre study assessing CTC numbers in 105 patients with SCLC recruited to a lung cancer biomarker programme between 2009 and 2017. CTC enumeration was performed with CellSearch™ at baseline and at least one further timepoint including EoT, 1[st] or 2[nd] disease relapse. Clinical data was extracted from the electronic patient records database. Data was analysed for trends between CTC number and progression free (PFS) and OS.

      Results:
      Baseline measurements were stratified into groups of ≥50 and <50 CTCs/7.5 ml. Patients in the ≥50 CTCs/7.5 ml group had significantly lower mean PFS (9.6 vs 5.8 months, p < 0.05) and OS (14.4 vs 9.3 months, p < 0.05). EOT CTCs were significantly lower than at baseline (mean = 73 vs 632 CTCs/7.5 ml, p < 0.05) and lower than 1[st] (mean = 202 CTCs/7.5 ml) and 2[nd] (mean = 602 CTCs/7.5 ml) relapse. EOT CTCs were split into groups of 0, 1–20 and >20 CTCs/7.5 ml. Patients in the >20 CTCs/7.5 ml group had the shortest PFS (mean = 4.6 months) and significantly shorter OS (mean = 5.6 months, p < 0.05) compared to those with less CTCs. The same CTC groups were used for the 1[st] relapse, with decreasing mean PFS (3.9 vs 6.8 months) and OS (10.1 vs 17.1 months) as patients moved from the most to the least favourable groups.

      Conclusions:
      This study indicates that EoT CTC number may be predictive of both PFS and OS. We also noted a negative correlation between relapse CTC numbers and survival times, but further research is required to determine the statistical significance of this relationship. With further work, CTCs may have a role in evaluating response to treatment and monitoring for relapse.

      Clinical trial identification:


      Legal entity responsible for the study:
      The Christie NHS Foundation Trust

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      88P - Feasibility of outpatient dinutuximab (D) and irinotecan (I) for second-line treatment of relapsed or refractory small cell lung cancer (RR SCLC): Part 1 of an open-label, randomized, phase 2/3 study (Now Available) (ID 589)

      12:30 - 12:30  |  Presenting Author(s): M.J. Edelman  |  Author(s): O. Juan, A. Navarro, G. Golden, A. Saunders

      • Abstract
      • Slides

      Background:
      D, a chimeric monoclonal antibody that binds cell-surface GD2 expressed on SCLC, can cause significant pain. Combined tolerability with I in RR SCLC patients (pts) is unknown.

      Methods:
      Part 1, an intrasubject dose-escalation lead in to the main study, investigated outpatient use of D + I. Eligibility: RR SCLC following first-line platinum-based therapy with a life expectancy of ≥12 weeks, PS 0-1. Pts received intravenous (IV) D and I (fixed dose 350 mg/m[2]) on Day 1 q 21-days. D was dosed 10 mg/m[2], and increased 2 mg/m[2]/cycle, if pain was < Grade 2, no opioid medications were required, and prior dose was otherwise tolerated. Doses could be decreased based on toxicity observed. Pretreatment included IV hydration, antihistamines, and antipyretics. Pts were monitored for 4 hours after D. Pts remain on treatment until intolerance, progression, or death.

      Results:
      12 pts were treated (8 male) in US and Spain. Mean (range) age was 68 (47–79) years. A median (range) of 3 (2–4) cycles were completed per pt. Median (range) D dose achieved was 14 (10–16) mg/m[2]. 121 adverse events (AEs) were reported in the Table. There were no grade (gr) 5 AEs. Pain AEs, attributable to D, were mainly gr 1 (80%), with most resolving within hours of infusion; one subject experienced gr 3 AEs, and received the same D dose in the subsequent cycle. Overall, pain did not result in D dose reductions, discontinuations or hospitalizations. Most other AEs were GI in nature and likely attributable to I.

      AEGrade, N subjects (%)
      1234
      Pain
      Back pain5 (41.7%)1 (8.3%)1 (8.3%)0
      Pain in extremity3 (25.0%)000
      Abdominal pain1 (8.3%)1 (8.3%)00
      Arthralgia1 (8.3%)01 (8.3%)0
      Headache2 (16.7%)000
      Non-pain
      Diarrhea10 (83.3%)2 (16.7%)00
      Nausea5 (41.7%)3 (25.0%)00
      Cough3 (25.0%)1 (8.3%)00
      Vomiting3 (25.0%)1 (8.3%)00
      Anaemia2 (16.7%)01 (8.3%)0
      Constipation3 (25.0%)000
      Decreased appetite3 (25.0%)1 (8.3%)00
      Asthenia2 (16.7%)1 (8.3%)00
      Dehydration2 (16.7%)000
      Fatigue1 (8.3%)1 (8.3%)00
      Hypomagnesaemia1 (8.3%)1 (8.3%)00
      Infusion related reaction1 (8.3%)000


      Conclusions:
      D up to 16 mg/m[2] + I have been tolerated with no unanticipated AEs. Part 2 of the study has begun and will randomize ∼460 subjects to: I vs. D + I vs. topotecan.

      Clinical trial identification:
      EudraCT 2017-000758-20 ClinTrials NCT03098030

      Legal entity responsible for the study:
      United Therapeutics Corporation

      Funding:
      United Therapeutics Corporation

      Disclosure:
      M.J. Edelman: Primary investigator for this corporate-sponsored research. O. Juan, A. Navarro: Principal investigator for this corporate-sponsored research. G. Golden, A. Saunders: Employed by United Therapeutics Corporation who is the sponsor of this study.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      89P - The role of palliative care in extensive stage lung cancer (Now Available) (ID 215)

      12:30 - 12:30  |  Presenting Author(s): Y. Ahmed

      • Abstract
      • Slides

      Background:
      Small cell lung cancer (SCLC) represents approximately 10–15% of all lung cancers and is characterized by an aggressive disease course. Despite being a relatively chemotherapy- and radiation- sensitive disease, recurrence and eventual chemoresistance is frequent. prognosis in these patients remains poor, with a median survival of 10–12 months from diagnosis. These patients may experience significant physical and psychological distress throughout the course of their disease, resulting in impaired quality of life (QOL). Early implementation of palliative care can lead to improved symptom control, reduced distress, limit hospitalizations and aid in the decision making with regards to the delivery of care, ultimately providing improved satisfaction with end of life care.

      Methods:
      A retrospective, single-center audit of all patients diagnosed with small cell lung cancer over a six-year period (Jan 2010–Dec 2016). Patients were identified using the Hospitals’ Inpatient Enquiry (HIPE) database. Clinical data were retrieved from hospital pathology and imaging reports as well as patient records. Statistical analysis was performed using a Kaplan-Meier analysis and Pearson correlation coefficient.

      Results:
      115 patients were diagnosed with SCLC. 91/115 patients (79%) were diagnosed with extensive stage disease (40 female patients (44%) and 51 male (56%)). Median age at diagnosis was 66 years. Median ECOG performance status was 1. 63/91 of these patients (69%) received a palliative care review within one month of diagnosis. 51 patients (56%) were alive at six months, with 29 (32%) alive at one year. Patients in the group receiving early palliative care intervention demonstrated improved overall survival, and this correlation was statistically significant (p = 0.01). The median number of hospitalizations in this cohort was also lower (median = 1) than those with delayed palliative care input (median = 2), however, this result was not statistically significant.

      Conclusions:
      Early palliative care input improves quality of life and translates into a survival benefit. Increased awareness of the benefits of palliative care in extensive stage small cell lung cancer at diagnosis is paramount in ensuring optimal patient outcomes and patient satisfaction with their oncological care.

      Clinical trial identification:
      not clinical trial

      Legal entity responsible for the study:
      HSE

      Funding:
      Has not received any funding

      Disclosure:
      The author has declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      90P - Study of intestinal flora differences between small cell lung cancer (SCLC) patients and healthy people (ID 390)

      12:30 - 12:30  |  Presenting Author(s): L. Zhang  |  Author(s): J. Chen

      • Abstract

      Background:
      SCLC accounts for approximately 14% of lung cancer. There are a large number of symbiotic microorganisms in the intestinal tract of the human body. Under normal circumstances, intestinal flora, the body and the external environment maintain a relative balance to maintain the health of the body. If the balance is broken, the body will show an alteration of intestinal flora, which may cause disease.

      Methods:
      (1) Clinical fecal specimens collection: Fresh fecal samples were collected before the first cycle of chemotherapy and labeled as 1–14. 14 healthy people were matched. (2) Intestinal bacteria DNA extraction. (3) The changes of intestinal flora were analyzed by PCR-DGGE. Cutting of the bands with significant differences in sequencing to determine the classification of bacteria. (4) Results analysis: Quantity One software and SPSS19.0 software were used to analyze the results.

      Results:
      (1) Intestinal flora analysis in SCLC group and healthy group: The diversity index and abundance of the healthy group was higher than those of the SCLC group, the difference in diversity index was statistically significant (P < 0.05), but the difference in abundance was not significant (P > 0.05). (2) Sequencing results analysis: Brightness of bands 1, 3, 7 and 8 were higher in the SCLC group than in the healthy group; brightness of bands 2, 4, 5, 6 and 9 were higher in the SCLC group than in the healthy group. The results show that the sequence similarity of bands 1 and 3 with Eubacterium xylanophilumisis were 98% and 97%, respectively; the sequence similarity of bands 2 and 6 with Prevotella copri 91% and 97%, respectively; the sequence similarity of band 4 with Blotia sp.was 93%; the sequence similarity of band 5 with Dialister succinatiphilus was 92%; the sequence similarity of band 7 with Eubacterium eligens was 100%; the sequence similarity of band 8 with Clostridium sp. was 99%; the sequence similarity of band 9 with Pseudobutyrivibriorum inisis 100%.

      Conclusions:
      (1) Intestinal flora differ between SCLC patients and healthy people, the difference in diversity index is significant, but the difference in abundance is not significant. (2) The increase of Eubacterium xylanophilum, Eubacterium eligens and Clostridium sp.are positively correlated with the occurrence of SCLC. The decrease of Prevotellacopri and Pseudobutyrivibriorum inis are positively correlated with the occurrence of SCLC.

      Clinical trial identification:


      Legal entity responsible for the study:
      2nd Hospital Affiliated to Dalian Medical University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      92P - Expression of TNFRII in serum is correlated with the significant risk of subcentimeter lung adenocarcinoma (ID 312)

      12:30 - 12:30  |  Presenting Author(s): Y. Zhang  |  Author(s): B. Han, B. Sun, K. Yu, T. Chu, J. Qian, Q. Chang

      • Abstract

      Background:
      With the rapid advances of LDCT screening for lung cancer, the opportunity to detect subcentimeter NSCLC is gradually increasing. But, even subcentimeter NSCLCs are not always in the early stage. Thus, it is quite important for us to judge the possibility of malignancy for these patients, even the tumor size is less than 10 mm. Chronic inflammation is well established as a hallmark in lung carcinogenesis. The aim of the present study is to evaluate the correlation between inflammation biomarkers and the risk for subcentimeter lung adenocarcinoma.

      Methods:
      Inflammatory biomarkers were measured in 71 subcentimeter lung adenocarcinoma patients and 71 age-, sex- and smoking-matched healthy controls by using the Luminex bead-based assay.

      Results:
      The expression level of TNFRII is significantly down-regulated in subcentimeter lung adenocarcinoma patients compared with the healthy controls (P < 0.001). And the results were validated by oncomine data mining analysis. Elevated levels of TNFRII were associated with an 89% reduced risk for subcentimeter lung adenocarcinoma. (OR = 0.11, 95% CI: 0.04–0.30, P = 2.4 × 10[−5]). BLC was associated with a 2.70-fold (95% CI: 1.31–5.58, P = 7.0 × 10[−3]) increased risk of subcentimeter lung adenocarcinoma for the comparison of patients in the higher-level group with the lower-level group. To yield more information, the BLC/TNFRII ratio was created to examine their prediction for the risk of subcentimeter lung adenocarcinoma, and there was a 35-fold increased risk for patients in the higher-level group relative to patients in the lower-level group. Further ROC curve analysis revealed that TNFRII was a significant diagnostic biomarker for subcentimeter lung adenocarcinoma, with the area under the curve of 0.73 (95% CI: 0.65–0.82, P = 2.0 × 10[−6]). The sensitivity, specificity and accuracy were 0.75, 0.72 and 0.73, respectively.

      Conclusions:
      Our findings demonstrated that TNFRII was associated with the significant risk of subcentimeter lung adenocarcinoma and could be a promising biomarker for accessorily diagnosing subcentimeter lung adenocarcinoma.

      Clinical trial identification:


      Legal entity responsible for the study:
      Yanwei Zhang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      93P - DNA damage repair protein expression and EGFR gene status in NSCLC (ID 504)

      12:30 - 12:30  |  Presenting Author(s): J. Wang  |  Author(s): W. Luo

      • Abstract

      Background:
      Epidermal growth factor receptor (EGFR) is considered to be one of the key driver genes in non-small cell lung cancer (NSCLC). This study explored the potential association between EGFR and DNA damage repair gene expression (Ku70 and Rad51), and their relationship with prognosis in patients with NSCLC.

      Methods:
      We reviewed the clinical information of 79 patients. The expression of Ku70 and Rad51 were determined via immunohistochemistry in surgically resected of lung adenocarcinoma and densitometry analysis using image-pro plus 6.0 software. The clinical prognostic value of protein expression was investigated with univariate and multivariate survival analysis.

      Results:
      Compared with EGFR mutant patients, mean integral optical density (IOD) of Rad51 and Ku70 had no statistically significant difference in the wild type. Separating patients into low and high expression group according to the median value of IOD, Spearman's test showed no significant correlation between Ku70 and Rad51 expression level and EGFR gene status, while tumor tissue with T1-T2 staging had a lower Ku70 protein expression than T3-T4 (p < 0.05) according to the IOD. No statistical significant difference was found between in mean IOD of Ku70 and Rad51 between recurrence and non-recurrence. Kaplan-Meier analysis revealed that patients with Ku70 low expression tended to have poorer DFS than high expression group (20.6 vs. 22.7 months, p > 0.05). PFS of both Ku70 and Rad51 low expression group had an improving tendency comparing to high expression group (Ku70: 9.0 vs. 6.0 months, and Rad51: 10.0 vs. 7.0 months, both p > 0.05). Multivariate analysis showed, (negative vs. positive) was an independent impact factor of DFS; gender (female vs. male), EGFR status (mutant vs. wild type) and Ku70 expression level (low vs. high) were independent impact factors of PFS.

      Logistic regression between different proteins and possible impact factor
      ProteinVariablesSEExp(B)p
      Ku70EGFR0.4880.6840.688
      T-stage0.8330.0440.044
      Rad51EGFR0.4780.9250.925
      T-stage0.7320.1640.164


      Conclusions:
      We provide clinical evidence that Ku70 protein expression level is lower in higher T-stage tumor tissue than the lower. DNA damage repair protein expression level has not been detected to have significant correlation with EGFR gene status and disease recurrence, while tumor high expression level of Ku70 protein might have worse response to systemic therapy. However, further study is required.

      Clinical trial identification:
      This is an retrospective study and approve by the clinical trial committee of the hospital.

      Legal entity responsible for the study:
      Thoracic Oncology Center, West China Hospital

      Funding:
      State Key Laboratory of Biotherapy and Cancer Center of Sichuan province in China

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      94P - Lung cancer recurrence in patients with pre-operative circulating tumor DNA and elevated tumor markers (Now Available) (ID 449)

      12:30 - 12:30  |  Presenting Author(s): S. Isaksson  |  Author(s): A.M. George, M. Jönsson, H. Cirenajwis, H. Brunnström, P. Jönsson, J. Staaf, L.H. Saal, M. Planck

      • Abstract
      • Slides

      Background:
      Recurrence after surgically treated non-small cell lung cancer (NSCLC) is frequent. A subset of NSCLCs harbors mutations that are routinely analyzed in tumor tissue but can also be detected in cell-free circulating tumor DNA (ctDNA). We performed mutation analysis of tumors to subsequently quantify corresponding mutated ctDNA in pre-operative plasma. Furthermore, we analyzed five tumor markers in pre-operative serum to study the potential of these blood-based methods to predict lung cancer recurrence.

      Methods:
      Plasma and serum were collected pre-operatively from 167 patients surgically treated for primary lung adenocarcinoma at the Lund University Hospital 2005–2014. Tumor specimens were analyzed with Next-Generation Sequencing (NGS). 76/167 tumors harbored at least one mutation in either of the EGFR, BRAF or KRAS genes. Cell-free DNA from corresponding plasma (0.6 to 1.4 mL) was analyzed for mutations in these three genes using the IBSAFE method, an innovation upon standard droplet digital PCR. The tumor markers carcinoembryonic antigen, neuron-specific enolase, cancer antigen 125, human epididymis protein 4 and carbohydrate antigen 19–9 were analyzed in serum with electrochemiluminiscence immunoassay.

      Results:
      So far ctDNA and tumor markers have been analyzed in 41 cases. Twenty-nine patients had ≥1 elevated tumor markers and 10 had detectable ctDNA. Information about recurrence was missing in two patients. Sixteen patients were diagnosed with recurrence. Of these, nine had ≥1 elevated tumor markers, three had detectable ctDNA and two had both ctDNA and ≥1 tumor markers. Of 16 patients without recurrence, 10 had at ≥1 elevated tumor marker, one had detectable ctDNA, and one patient had positive ctDNA in combination with one elevated tumor marker. Four patients had stage IV disease at time of diagnosis, two of them with ctDNA and all four with ≥1 tumor markers.

      Conclusions:
      In summary, ctDNA and/or tumor markers might be useful to identify NSCLC patients with increased risk of recurrence but it remains to be investigated in larger studies and with greater plasma volumes how these biomarkers may fit into lung cancer management.

      Clinical trial identification:


      Legal entity responsible for the study:
      Lund University

      Funding:
      The Swedish Cancer Society, Skane University Hospital Foundation, the Mrs Berta Kamprad Foundation, the Gustav V:s Jubilee Foundation, the Gunnar Nilsson Cancer Foundation, BioCARE a Strategic Research Program at Lund University, and governmental funding (ALF)

      Disclosure:
      A.M. George, L.H. Saal: Named inventor of related intellectual property and shareholder of SAGA Diagnostics. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      95P - Intubated versus non-intubated general anesthesia for minimally invasive video-assisted thoracic surgery (VATS) in octogenarians (Now Available) (ID 268)

      12:30 - 12:30  |  Presenting Author(s): N. Ilic  |  Author(s): D. Ilic, J. Juricic, D. Krnic, N. Frleta Ilic, I. Simundza, D. Orsulic

      • Abstract
      • Slides

      Background:
      In recent years, non-intubated video-assisted thoracic surgey has gained popularity worldwide, especially in elderly lung patients. The main goal of this surgical practice is to achieve an overall improvement of patient management and outcome thanks to the avoidance of side-effects related to general anesthesia (GA) and single-lung ventilation. In this study we tried to compare a non-intubated general anesthetic technique with an intubated general anesthetic technique for VATS.

      Methods:
      Sixty patients aged 80 and more scheduled for VATS lung surgery, were allocated randomly into two groups with 30 patients each. The first group received standard general anesthesia with double lumen tube. The second group underwent a non-intubated anesthetic technique. Heart rate, mean arterial pressure, end-tidal CO~2~ and the visual analog pain score (VAS) measurements were recorded during the surgery and 24 hours after the surgery. Both groups received ultrasound guided paravertebral block before surgery with single injection of 20 ml of 0.25% levobupivacaine. VATS lung biopsy, sublobar or lobar resection were equally distributed in both groups of patients.

      Results:
      Time for anesthetic procedure was shorter in the non-intubated group. VATS lobecotomy was performed in the usual manner in all patients without any intraoperative complications. VAS scores in the first 24 hours werecomparable. We found significantlly shorter recovery times, reduced oxygen requirement, and shorter hospital stays in the non-intubated group. There were no significant differences in intraoperative blood loss, the operation time or postoperative complications between the non-intubated group and the intubated group of patients.

      Conclusions:
      In this study, our experience has shown that non-intubated VATS is a safe and feasible surgery for elderly lung cancer patients with certain advantages for the patients undergoing VATS. Our results indicate that we can achieve day surgery for selected patients. Further clinical studies should be carried out in order to improve surgical outcomes in elderly LC patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Ilic Nenad

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      96P - The role of bronchial stump reinforcement by flap in prevention of broncho-pleural fistula after major lung resections (Now Available) (ID 624)

      12:30 - 12:30  |  Presenting Author(s): F. Caushi  |  Author(s): I. Skenduli, A. Mezini, A. Hatibi, H. Hafizi, S. Bala, E. Hysa, F. Kokiqi

      • Abstract
      • Slides

      Background:
      Lung resection remain the treatment of choice especially for bronchogenic carcinoma and as well as for intractable end stage localized lung disease such as tuberculosis, bronchiectasis, lung abscess and complicated hydatid cyst. The development of bronchopleural fistula (BPF) remain the most devastating complications that may arise after lung resection especially after pneumonectomy. Bronchial stump reinforcement has been shown to significantly reduce incidence of BPF. There have been described various coverage techniques as: intercostal muscle flap, pleural flap, pericardial fat pad, diaphragm and azygous vein. The aim of this study is to compare the efficiency of bronchial stump reinforcement with flap versus non-reinforcement after major lung resection.

      Methods:
      This is a retrospective study where was analised the data for 300 patients who underwent major lung resections. 38 patients underwent pulmonectomy and the rest lobectomy, bilobectomy and sleeve lobectomy. The follow up period varied from 6 months up to 10 years. The patients were divided into 2 groups according to method of bronchial stump reinforcement or not. Two groups were matched as regard to: patient age, sex distribution, lung pathology and risk factors.

      Results:
      78.1% of patients had the diagnosis of NSCLC where 10% of them were subjected to neo-adjuvant chemotherapy. Only in 50% of patients that underwent pulmonectomy, the bronchial stump was reinforced by diverse nature of flap. Meanwhile, according to patients that underwent the other types of lung resections, the bronchial stump was reinforced by flap only in 7% of cases. There was 3% mortality in both groups for a period of postoperative follow up 3 months. Complications were observed in both groups, but BPF happened in 20 patients where only in three of them was performed the bronchial stump reinforcement.

      Conclusions:
      Intercostals muscle flap, pericardial fat pad and pleural flap are valuable and effective methods in prevention of broncho-pleural fistula following lung resection especially in pulmonectomies and broncho-sleeve lung resections. Surgical techniques using flap reinforcement seems to play a major role in the prevention of BPF especially after neo-adjuvant chemotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Fatmir Caushi

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      97P - Multistep prediction of cardio-pulmonary morbidity after lung cancer surgery (ID 558)

      12:30 - 12:50  |  Presenting Author(s): N. Lukavetskyy  |  Author(s): R. Litviniak, I. Hipp, T. Fetsych

      • Abstract

      Background:
      The aim of the study is to establish “multistep” criteria to clarify the high-risk patients with resectable lung cancer. The purpose of the present study was to identify preoperative risk factors in lung cancer patients.

      Methods:
      Retrospective review of the clinical records of all patients operated on thoracic department in 2010–2011. None of the patient received preoperative chemo and/or-radiotherapy. Patients older than 70 years, benign lung lesions, lung metastases, or patients after non-curative lung resection were excluded. A total of 168 patients met criteria for the study. All factors were divided into 3 groups: initial (36 factors incl. TNM, histology, presence of comorbidities etc), functional (29 factors – lung function parameters, echocardiography), and surgery-related (14 factors incl. type of surgery and lymphadenectomies etc.). We analyzed postoperative mortality, pulmonary complications, arrhythmia and hypertension. All of the variables that were found to be significant in the univariate analyses were entered into the multivariate analyses using a forward step-wise logistic regression model in all group separately.

      Results:
      Group 1. Our model identifies no risk factors for postoperative mortality and hypertension. Diabetes mellitus is associated with higher rates of arrhythmia. Predictors that are associated with pulmonary complication include: the history of second primary cancer, neuroendocrine tumours, rare type of lung tumours. Group 2. There are no risk factors for arrhythmia and hypertension. Left ventricular ejection fraction <45 is associated with postoperative mortality, forced expiratory flow at 25% – pulmonary complication. Group 3. The following criteria related with postoperative mortality – number of unobstructed (by tumours) lung segments, pulmonary complications – ventilation time, arrhythmia – number of resected lymph nodes, hypertension – surgery with resection adjacement structures.

      Conclusions:
      There are risk factors associated with higher rates of specific postoperative mortality and morbidity but need to be validated by prospective study.

      Clinical trial identification:


      Legal entity responsible for the study:
      Lviv Medical University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      98P - Trends in and impact of hospital-acquired adverse events in patients with lung cancer undergoing lung resection (ID 341)

      12:50 - 12:50  |  Presenting Author(s): A. Gupta  |  Author(s): K.C. Mara, K. Kernstine, S. Khanna, D.E. Gerber

      • Abstract

      Background:
      Hospital-acquired adverse events are a measure of quality of care delivered and may adversely impact patient outcomes. It is unknown if there been a change in the rates and impact of these adverse events with their increasing recognition as quality measures. We analyzed hospital-acquired adverse events in patients with lung cancer undergoing lung resection using the National Hospital Discharge Survey (NHDS) database from 2001 to 2010.

      Methods:
      NHDS collects clinical information on patients dismissed from non-Federal short-stay United States hospitals. Demographics, diagnoses, procedures, and dismissal information were abstracted using ICD-9 diagnosis (162.X) and procedure codes (32.9, 32.3X, 32.4X, and 32.5X). The Agency for Healthcare Research and Quality Patient Safety Indicators (PSIs) were utilized to identify hospital-acquired adverse events. Weighted analyses were performed using SAS version 9.4.

      Results:
      An estimated 585,408 patients with lung cancer underwent lung resection during the study period and were included in the analysis; 58.3% were >65 years, 49.1% women, 66% white. Of these, 153,609 (26.2%) suffered from ≥1-PSI event during their hospitalization. The proportion of patients suffering from ≥1-PSI event consistently increased from 22.0% in 2001–2002, to 34.8% in 2009–2010, p value <0.01. Compared to those with no PSI, patients with ≥1-PSI experienced higher in-hospital mortality (2.2%, vs 15.7%, adjusted odds ratio, 7.8, 95% CI 5.1–11.8), and prolonged length-of-stay (7.8 days, vs 12.5 days, adjusted mean difference, 4.6 days, 95% CI 3.7–5.5), p value for both <0.01.

      Conclusions:
      There was a substantial increase in the frequency of potentially avoidable adverse events in this national database of lung cancer patients undergoing resection, associated with worse outcomes. Although this may in part be due to increasing recognition, interventions are required to prevent these potentially avoidable events.

      Clinical trial identification:
      Not applicable

      Legal entity responsible for the study:
      Arjun Gupta

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      100P - Prognostic factors in surgically resected N2 small cell lung cancer: Significance of the subcarinal node (ID 316)

      12:50 - 12:50  |  Presenting Author(s): R. Qiao  |  Author(s): J. Xu, Y. Zhang, W. Yang, B. Zhang, S. Wang, R. Zhong, B. Han

      • Abstract

      Background:
      Surgical resection is being increasingly used for stage IIIA-N2 small cell lung cancer (SCLC). The aim of this study was to determine the prognostic factors in patients with pathologic N2 (pN2) stage IIIA SCLC.

      Methods:
      A retrospective analysis of 163 consecutive patients with pN2 stage IIIA SCLC who underwent pulmonary resections and systemic lymphadenectomies was conducted. Potential clinicopathological factors that could influence OS were statistically analyzed. The prognostic significance was examined by Cox regression analysis.

      Results:
      The median overall survival (OS) was 10.6 months. Multiple-station lymph node metastasis indicated a poorer prognosis than single-station involvement (p = 0.003). With respect to the station of lymph node metastasis, the OS was only related to the involvement of the subcarinal node, regardless of tumor location (p < 0.05). Multivariate analysis showed two statistically significant risk factors for survival, including multiple-station lymph node and subcarinal node metastasis (hazard ratio [HR] = 1.76, 95% confidence interval [CI]:1.11–2.78, p = 0.015; HR = 1.61, 95% CI: 1.03–2.50, p = 0.036, respectively).

      Conclusions:
      We found multiple-station N2 metastases and subcarinal node involvement were independent prognostic factors for worse survival in pN2 stage IIIA SCLC patients, which may be helpful to identify a valid subpopulation of N2 patients who can benefit from surgical intervention and guide postoperative adjuvant therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai Chest Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      101P - Overtreatment in early lung cancer: Survival is independent of pathological confirmation (Now Available) (ID 302)

      12:50 - 12:50  |  Presenting Author(s): F. Zehentmayr  |  Author(s): M. Sprenger, G. Fastner, F. Sedlmayer

      • Abstract
      • Slides

      Background:
      Lung cancer causes 27% of all cancer deaths. About 10% of the patients are diagnosed in UICC stages I-IIa. Although pathological proof is compulsory before cancer treatment, some patients are unable or reluctant to undergo invasive diagnostic procedures. Nonetheless, these individuals are frequently treated similarly to patients with histologically confirmed disease, which might bear the risk of treating indolent or even benign tumors. A SEER-analysis [1] found a significantly better OS in patients with clinically diagnosed tumors without pathological confirmation, which made the authors conclude that patients with such lesions are probably overtreated. We analysed our own data as a non-selected single centre cohort to compare survival data of patients with and without pathological proof.

      Methods:
      163 patients with NSCLC UICC I-IIa irradiated between 2002 and 2016 were eligible. Of these, 123 patients had pathological proof of disease. In these cases, irradiation treatment was administered in the same way as if these patients had a confirmed malignant disease. In accordance with international guidelines, the decision was based on the patient's age and smoking history, new or growing nodule on CT and positive PET-scan.

      Results:
      Both overall (OS) and cancer specific survival (CSS) did not differ between patients with pathological confirmation compared with those without: median OS 40,1 (range: 0,3–147,5) months versus 39,4 (range: 0,5–162) months (p = 0,958), median CSS 51,7 (3,7–129,5) months versus 113,4 (0,5–162) months (p = 0,585). Patients with pathological confirmation had higher UICC stage (p-value = 0,002) and received more chemotherapy (p-value < 0,001).

      Conclusions:
      Our study revealed no difference in OS and CSS for irradiated stage I-IIa lung cancer patients with and without histological confirmation, thus indicating a comparable benefit from treatment. A main reason probably lies in the strict adherence to a modern diagnostic work-up including PET-CT scan for all patients.[1] Shaikh T (2016) Absence of Pathological Proof of Cancer Associated with Improved Outcomes in Early-Stage Lung Cancer. J Thorac Oncol 11 (7):1112–1120.

      Clinical trial identification:


      Legal entity responsible for the study:
      Paracelsus Medical University, University Hospital Salzubrg, Department of Radiation Oncology

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      102P - Stereotactic lung radiotherapy for early-stage NSCLC: An institution&#x0027;s experience (Now Available) (ID 606)

      12:50 - 12:50  |  Presenting Author(s): N. Ferreira  |  Author(s): C. Travancinha, T. Antunes, J. Fonseca, M. Labareda, F. Santos

      • Abstract
      • Slides

      Background:
      Stereotactic body radiation therapy (SBRT) is the primary alternative to surgery for patients with clinical stage I or II non-small cell lung cancer (NSCLC) who have significant comorbidity that precludes safe resection and for those who refuse surgery.

      Methods:
      Patients with clinical stage I or II NSCLC treated with SBRT in our institution, between January 1[st] 2015 and March 31[st] 2017, were retrospectively analyzed. Control and survival rates were calculated using the Kaplan-Meier method. Acute and late toxicities were graded according to the CTCAE v4.0.

      Results:
      78 patients were identified, 58 men and 20 women, with a median age of 73 years (range 50–90), and a ECOG performance status of 0 or 1 in 69% of these patients. Of the biopsy proven patients (63%), 53% were adenocarcinomas and 37% were squamous cell carcinomas. 52 patients were staged as IA (T1N0), 23 as stage IB (T2aN0) and 3 as stage IIB (T3N0), all of which were clinically staged with PET-CT. The majority of the tumors (72%) were peripherally located, with a median size of 23 mm. 48Gy in 4 fractions was the most prescribed scheme (76%), followed by 50Gy in 5 fractions (14%). 70 patients were eligible for assessment of recurrence. With a median follow-up of 17.5 months (range 3.5–50.6), 17 (24%) had disease recurrence, of which 11 (65%) had metastatic disease and 10 (59%) failed locally, with isolated distant or local relapse occurring in 5 (29%) cases each. The 1- and 2-year local control was 94% and 81%, respectively. The 2-year regional control, distant metastasis free survival and overall survival (OS) was 87%, 76% and 65%, respectively. Median OS was 37.8 months. The performance status was correlated with survival (p < 0.0001), whereas the tumor histology (adenocarcinoma) predicted for distant relapse (p = 0.03). No grade 3 toxicity was reported. Late grade 2 toxicity occurred in 29% of patients (15% pneumonitis, 9% fatigue, 3% rib fracture, 6% chest wall pain, 2% esophagitis).

      Conclusions:
      In our population of early stage lung cancer patients treated with SBRT, high rates of local control were achieved, with overall survival outcomes and toxicity profile comparable to other published data, confirming the established role of SBRT in stage I/II non-small cell lung cancer patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Instituto Português de Oncologia de Lisboa

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      103P - Stereotactic body radiotherapy followed by radiofrequency ablation for inoperable stage Ib non-small cell lung cancer (ID 169)

      12:50 - 12:50  |  Presenting Author(s): H. Ding  |  Author(s): S. Chen, T. Chen, M. Su, C. Zhu, Q. Zheng

      • Abstract

      Background:
      Hypoxia in the center of large tumor directly leads to radiation resistance. Concurrently, heat loss results in incomplete ablation oflarge tumors abutting airways or blood vessels. The combining stereotactic body radiation therapy (SBRT) and radiofrequency ablation (RFA) for non-small cell lung cancer (NSCLC) treatment could overcome respective shortcoming.

      Methods:
      In this study, we report our experience with combined SBRT and RFA in 27 medically inoperable patients with biopsy-proven T2aN0M0 NSCLC. Patients were treated with SBRT to a dose of 50 Gy followed by CT-guided RFA within 7 days.

      Results:
      There were no treatment-related deaths. At a mean follow-up period of 29.6 months (range, 3 to 56 months), 15 patients (55.6%) died, with 1-year, 2-year, and 3-year cumulative survival rates of 77.8%, 63.0% and 48.1%, respectively. Threeof the deaths were cancer related. Four (14.8%) and five (18.5%) patients had local and distant recurrence, respectively.

      Conclusions:
      We concluded that SBRT followed by RFA appears effective for centrally located NSCLC. Survival rates appear to be better than with SBRT or RFA alone. Interactions between the two modalities warrants deeper investigation.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      104P - Adjuvant chemotherapy may improve prognosis in surgically resected stage I NSCLC with lymphovascular invasion (Now Available) (ID 481)

      12:50 - 12:50  |  Presenting Author(s): S. Wang  |  Author(s): J. Xu, F. Qian, W. Yang, B. Zhang, J. Qian, R. Qiao, B. Han

      • Abstract
      • Slides

      Background:
      The 8[th] edition of the TNM classification for non-small cell lung cancer (NSCLC) has recently been approved. Lymphovascular invasion (LVI) has been reported to be a strong risk factor for stage I patients. Meanwhile, the efficacy of adjuvant chemotherapy for surgically resected pathologic stage I NSCLC is controversial. This study aimed at exploring the association between adjuvant chemotherapy and survival in stage I NSCLC patients with LVI.

      Methods:
      A total of 2600 patients with stage I NSCLC treated in the Shanghai Chest Hospital (2008–2012) were included in the analysis, of which 221 were pathologically diagnosed with LVI. We divided these patients into an ACT (adjuvant-chemotherapy) group and a surgery alone group. By using the Kaplan–Meier method and Cox proportional hazard regression model, we explored whether lymphovascular invasion was a poor prognostic factor and the application of adjuvant chemotherapy could improve the prognosis.

      Results:
      For all stage I NSCLC patients, it was observed that patients with LVI had an unfavorable Lung-cancer specific survival (LCSS) (hazard ratio [HR]: 1.604; 95% confidence interval [CI]: 1.124–2.289; P = 0.009) and recurrence-free survival (RFS) (HR: 1.943; 95% CI: 1.491–2.532; P < 0.001). The presence of LVI was suspected to be correlated with larger tumor size, and adenocarcinoma. Analysis of 221 patients with LVI indicated an increased LCSS (HR: 0.31; 95% CI: 0.161–0.595; P < 0.001) and RFS (HR: 0.53; 95% CI: 0.530–0.286; P = 0.044) with adjuvant chemotherapy treatment. We saw significant differences in LCSS and RFS in patients treated with adjuvant chemotherapy with both stage IA and stage IB disease.

      Conclusions:
      For all stage I NSCLC patients, LVI was correlated with poorer prognosis, which was improved by adjuvant chemotherapy. Our preliminary study suggests that adjuvant chemotherapy might be an appropriate option for stage I NSCLC patients with LVI.

      Clinical trial identification:


      Legal entity responsible for the study:
      Wang Shuyuan

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      105P - Feasibility study of adjuvant chemotherapy with cisplatin and pemetrexed short hydration regimen for completely resected non-small cell lung cancer (ID 358)

      12:50 - 12:50  |  Presenting Author(s): R. Dokuni  |  Author(s): M. Tachihara, S. Tokunaga, K. Okuno, M. Yamamoto, K. Kobayashi, Y. Tanaka, Y. Funada, Y. Maniwa, Y. Nishimura

      • Abstract

      Background:
      Cisplatin-based adjuvant chemotherapy improved overall survival and relapse free survival (RFS) of patient with completely resected stage II and III non-small cell lung cancer (NSCLC). However, the adverse effects of cisplatin-based regimen have not satisfied both patients and clinicians yet, and most of patients have to be hospitalized during chemotherapy. Recently, pemetrexed regimen as adjuvant chemotherapy was reported to result less toxicity than vinorelbine. Additionally, safety of short hydration method for cisplatin administration has been reported. The aim of our study was to investigate the feasibility of pemetrexed plus cisplatin with short hydration as adjuvant chemotherapy in patients with completely resected NSCLC.

      Methods:
      21 completely resected non-squamous NSCLC patients with pathological stage IIA, IIB, and IIIA were enrolled. Adjuvant chemotherapy was started between four and eight weeks after surgery, and consisted of 4 cycles of pemetrexed (500 mg/m[2]) plus cisplatin (75 mg/m[2]) every 3 weeks with short hydration. The first cycle of each patient was administered with hospitalization, and the others were in outpatient clinic in principle. The primary endpoint was the completion rate of treatment, and secondary endpoints included toxicity, two-year RFS rate, and outpatient treatment rate (rate of outpatient chemotherapy at second cycle).

      Results:
      Patients were median age 66 years (range 57 to 75) and gender 57% male/43% female. All cases were adenocarcinoma. Pathological stages were IIA 52%, IIB 19%, and IIIA 29%. 81% of patients received 4 cycles of therapy as scheduled. Only 1 patient needed decrease of chemotherapy dose and the reason was prolonged anorexia. No grade 3 or 4 hematologic toxicity was observed. Except for one grade-3 pulmonary thromboembolism and two grade-3 anorexia the adverse events were relatively mild. Increased serum creatinine was observed only one patient (grade 1). Outpatient treatment rate was 90%. Two-year RFS rate was 59%.

      Conclusions:
      Adjuvant pemetrexed and cisplatin chemotherapy with short hydration for resected non-squamous NSCLC is safety and able to be performed in outpatient basis.

      Clinical trial identification:


      Legal entity responsible for the study:
      Kobe University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      106P - The value of adjuvant chemotherapy in patients with resected stage IB solid predominant and solid non-predominant lung adenocarcinoma (Now Available) (ID 472)

      12:50 - 12:50  |  Presenting Author(s): S. Cao  |  Author(s): J. Xu, H. Zhong

      • Abstract
      • Slides

      Background:
      The adjuvant chemotherapy(ACT) of stage IB lung adenocarcinoma remain controversial, and solid ingredients were supposed to be connected to the poor survival. We are intended to explore the benefits adjuvant chemotherapy made on patients in IB with solid ingredients.

      Methods:
      a number of 334 completely resected patients with lung adenocarcinoma in stage IB from 2006 to 2015 were reviewed. All the pathological slides were evaluated with solid ingredients composed.

      Results:
      Our data showed that although disease-free survival (DFS)(p = 0.661) and overall survival (OS)(p = 0.130) were not significantly different in solid growth pattern with or without ACT, patients with solid predominant patterns tend to have longer DFS [hazard ratio (HR) 0.403, p = 0.021)]and OS (HR 0.286, p = 0.009) with ACT. In patients with solid non-predominant patterns, receiving ACT had little influence in DFS(p = 0.231) and OS (p = 0.611).

      Conclusions:
      the solid predominant pattern in postoperative patients of stage IB could benefit from adjuvant, and solid non-predominant pattern couldn't.

      Clinical trial identification:


      Legal entity responsible for the study:
      Shanghai chest hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      107P - Meta-analysis on defining optimal follow up of patients with early stage NSCLC after definitive therapy (ID 260)

      12:50 - 12:50  |  Presenting Author(s): D. Wankhede

      • Abstract

      Background:
      This is a meta-analysis to evaluate the impact of various follow-up intensities and strategies on the outcome of patients after definitive therapy for early stage NSCLC.

      Methods:
      All the original RCTs comparing the follow-up protocols of different definitive therapies on early stage NSCLC were considered up to November 2017. Specifically, the Jadad score was used to evaluated and appraise the quality of the selected randomized trials.

      Results:
      In the five RCTs, a total of 1,826 patients with early stage NSCLC undergoing various definitive therapies were randomized into less intensive or minimal follow-up group (n = 984) and intensive follow-up (n = 842). The female: male ratio was 1: 1.45 and 1.26:1 in the intensive and less intensive follow-up groups, respectively. A critical evaluation of all the articles that were included in the present meta-analysis revealed that most of the patients that took part in the study had a follow-up of at least 3 years. Nonetheless, three studies reported a drop out rate that varied between zero and 7% for the less intensive follow-up category and from zero to 38.2% for the minimal or less intensive follow-up group.

      Conclusions:
      The meta-analysis revealed that in selected patients with stage IIIA-N2 NSCLC, surgical resection does not improve the overall survival rate as is the case with radiotherapy. None of the follow up protocols was associated with increase in recurrence rate or overall survival rate. The ideal follow up should be individualized.

      Clinical trial identification:


      Legal entity responsible for the study:
      Durgesh Wankhede

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      108TiP - A multi-center, prospective, randomized controlled clinical trial: Comparison between wedge resection and segmentectomy in the surgical treatment of ground glass opacity-dominant stage IA non-small cell lung cancer (Now Available) (ID 507)

      12:50 - 12:50  |  Presenting Author(s): L. Liu  |  Author(s): H. Liao

      • Abstract
      • Slides

      Background:
      Sublobectomy is an optimal choice for selected early-stage non-small cell lung cancer (NSCLC) patients. However, the criteria for the choice of two different types of sublobectomy methods (wedge resection and segmentectomy) is not clear yet. It was reported that tumor size and the percentage of the ground glass opacity (GGO) were the key parameters for the choice of the surgeries. The study was designed to verify the presumption that for GGO-dominant early stage NSCLC patients (cT1a-T1bN0M0, and GGO component ≥75%), wedge resection is equal to segmentetomy in long term survival.

      Trial design:
      The trial is a multi-center, prospective randomized, open-labelled, active-control, non-inferiority study. In the study, the cT1a-T1bN0M0 NSCLC patients with GGO component ≥75% who intended to have surgeries were enrolled. Patients were randomized into wedge resection + hilum and mediastinal lymph nodes dissection or sampling group (experimental group) and segmentectomy + hilum and mediastinal lymph nodes dissection or sampling group (control group). The primary endpoint was 5-year disease-free survival (DFS) rate. The secondary endpoints consist of 3-year DFS rate, 5/10-year overall survival rate (OSR), rates of morbidity and 30-day mortality rates, and lung function change. All the study data were descriptively analyzed. Comparison for the differences in DFS was performed by the Log-rank test. The CMHX[2] tests were completed to compare the differences in 3-year DFS and 5/10-year OSR after taking the center effects into considerations. The differences in effectiveness were compared accordingly. In the case that statistical significance was confirmed between the groups, the non-inferiority comparisons would be completed afterwards. The impact factors of effectiveness were measured by the or the Cox modeling, or the Logistic regression analysis. The safety was measured by (serious) adverse events with a Chi-square test. To summarize, the purposes of the study are to seek for a reliable criteria of selection of different type of sublobectomy for early stage NSCLC.

      Clinical trial identification:
      NCT02718365

      Legal entity responsible for the study:
      Lunxu Liu

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      113P - Exosomal miRNAs in peripheral blood as novel diagnostic biomarkers of radioresistant lung adenocarcinoma (ID 391)

      12:50 - 12:50  |  Presenting Author(s): C. Zhu  |  Author(s): L. Wang, Q. Zhang, H. Xu, J. Tong, Y. Wan, Q. Zheng

      • Abstract

      Background:
      Radioresistance occurs in a high proportion of patients with lung adenocarcinoma (LA), resulting in poor response to radiation and dim prognosis. Early identification of radioresistance would accordingly guide adjustment of treatment regimens. Cell-derived exosomes containing proteins and nucleic acids can contribute to intercellular communication. Emerging evidence demonstrates exosomes are intimately involved in therapeutic resistance, and thus exosomes potentially can be used to predict radioresistance.

      Methods:
      Five patients with radioresistant LA and five healthy volunteers as negative controls were enrolled. Exosomes in 1 ml plasma were isolated with lipid nanoprobe approach, and miRNA from each sample was isolated with Qiagen miRNeasy kit. miRNA expression profiling was analyzed using microarrays, and quantitative real-time PCR was further used to validate miRNAs showing differential expression among two groups. Subsequently, selected miRNAs were validated in additional 27 patients with LA to evaluate their diagnostic potential.

      Results:
      Microarray analysis revealed there were 7 miRNAs expressed at a remarkably higher level in patients with radioresistant LA in comparison with that of healthy volunteers, and qRT-PCR further validated 3 miRNAs. Of note, exosomal miR-96-3p miR-10b-5p and miR-21 highly expressed in patients with radioresistant LA. The corresponding diagnostic specificity in predicting radioresistance was 0.916, 0.902 and 0.871, respectively. Moreover, exosomal miR-96-3p intensely indicates poor overall survival (with hazard ratio [95% confidence interval]: 2.38 (1.41–3.86), p < 0.001). However, it does not show significant correlation with tumor size and differentiation degree (p > 0.05).

      Conclusions:
      Our preliminary results indicate three exosomal miRNAs potentially can be used as non-invasive diagnosis biomarker of radioresistant LA.

      Clinical trial identification:


      Legal entity responsible for the study:
      International Ethical Guidelines for Biomedical Research Involving Human Subjects

      Funding:
      Youngth Programm of Natural Science Foundation of Jiangsu Province (BK20170134)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      114P - Evaluation of pulmonary function parameters after moderate hypofractionated image-guided thoracic irradiation in locally advanced node-positive non-small cell lung cancer patients with very limited lung function (Now Available) (ID 633)

      12:50 - 12:50  |  Presenting Author(s): F. Manapov  |  Author(s): O. Roengvoraphoj, J. Taugner, M. Dantes, C. Wijaya, C. Belka, C. Eze

      • Abstract
      • Slides

      Background:
      To evaluate the changes in pulmonary function parameters (PFT) after moderate hypofractionated image-guided thoracic irradiation (Hypo-IGRT) in locally advanced node-positive non-small cell lung cancer patients with very limited lung function.

      Methods:
      PFT was measured in 8 patients with NSCLC UICC stage IIIA, IIIB and IIIC (UICC 8[th] Edition) and very limited PFT (FEV1 ≤ 1 L and/or DLCO ≤ 40% and/or long-term oxygen therapy) prior to as well as 3 and 6 months after Hypo-IGRT. Vital capacity (VC), forced expiratory volume in 1s (FEV1), and single-breath diffusing capacity of the lung for CO (DLCO-SB) as PFT parameters were analyzed. Hypo-IGRT was delivered to a total dose of 45 Gy (ICRU) in 15 fractions under daily image-guidance.

      Results:
      Eight patients (5 men/3 women) were treated with Hypo-IGRT. The median follow-up was 20 months. COPD GOLD III and IV was diagnosed in 2 (25%) and 4(50%) patients, respectively. Five (63%) patients were on long-term oxygen treatment. The median initial VC, FEV1 and DLCO-SB was 1.69L/64.8% predicted (range: 1.36–2.66/33–80%), 1L/39.4% predicted (range: 0.78–1.26/28–60%) and 33.3% (range: 13.3–54), respectively. The median value for VC, FEV1 and DLCO-SB 3 months after Hypo-IGRT was 2.05L/56.35% predicted (range: 1.34–2.33/47–81.5%), 1.08L/47.5% predicted (range: 0.74–1.60/30.8–59.59%) and 38.55% (range: 24–68), respectively. At 6 months post-treatment, the mean value for VC, FEV1 and DLCO-SB was 1.64L/66% predicted (range: 1.41–2.79/35.5–75.5%), 1.0L/47% predicted (range: 0.65–1.28/24.5–54.10%) and 31% (range: 27–43%), respectively. The estimated median overall survival (OS) was not reached for the entire cohort.

      Conclusions:
      No significant decrease in all tested parameters was found until 6 months after Hypo-IGRT. In this small study, Hypo-IGRT was safely delivered in locally advanced node-positive NSCLC patients with very limited lung function who were not suitable candidates for conventional treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital, LMU Munich

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      115P - Heart and pulmonary artery radiation doses in non-small cell lung cancer (Now Available) (ID 309)

      12:50 - 12:50  |  Presenting Author(s): K. Thippu Jayaprakash  |  Author(s): J. Rubasingham, C.J. Ingle, D. Muthukumar

      • Abstract
      • Slides

      Background:
      Heart and pulmonary artery (PA) radiation doses following radical (chemo) radiotherapy to non-small cell lung cancer (NSCLC) correlate with overall survival (OS). We aimed to quantify radiation doses delivered to heart and PA during radical (chemo) radiotherapy in NSCLC, and to analyse dose volume histogram (DVH) based parameters that were associated with poor OS.

      Methods:
      NSCLC patients who underwent radical (chemo) radiotherapy using 3-D conformal radiotherapy between 2015 and 2017 were identified from our institutional radiotherapy electronic database. Heart and PA were contoured according to the Radiotherapy Oncology Group Trial 1106 contouring atlas. Maximum, minimum and mean radiation doses delivered to heart and PA were quantified. DVH based parameters, volume of heart received 5 Gy (V5), and volume of PA received 40, 45, 50, 55 and 60 Gy (V40, V45, V50, V55 and V60 respectively) were obtained from radiotherapy plans and compared with values that showed correlation with poor OS in NSCLC.

      Results:
      Out of a total of nineteen patients, 63% had stage III tumours. Twelve patients were treated with 60 Gy/30 (over 6 weeks) and seven were treated with 55 Gy/20 (over 4 weeks) radiation dose-fractionation schedules. For heart, average radiation doses of mean, minimum and maximum radiation doses were 7.49, 0.33 and 44.18 Gy and corresponding PA radiation doses were 27.14, 5.37 and 52.67 Gy. Evidence suggests mean radiation dose of heart >7.8 Gy and heart V5 of >72.3% correlate with poor OS. In our study, we observed mean heart dose and V5 were less than these values in 74% and 89% patients respectively. PA DVH based parameters, V40 ≥ 80%, V45 > 70%, V50 ≥ 45%, V55 ≥ 32% and V60 > 37 correlates with poor OS. In our cohort of patients, V40, V45, V50, V55 and V60 were less than these values in 95%, 89%, 79%, 74% and 89% patients.

      Conclusions:
      In this retrospective study, we found heart and PA DVH based radiation dose parameters in NSCLC radical (chemo) radiotherapy were less than those of published values that were associated with poor OS. It is possible to achieve this in 3-D conformal radiotherapy planning for most patients. Including these DVH based parameters during radiotherapy planning would ensure radiation doses to these critical organs are optimised.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      116P - Comparison of conformity and homogeneity index values of VMAT and non-VMAT techniques used in lung cancer radiotherapy (Now Available) (ID 328)

      12:50 - 12:50  |  Presenting Author(s): A. Ozen  |  Author(s): M. Akcay, K. Duruer, O. Bozdogan, I.S. Surenkok, D. Etiz

      • Abstract
      • Slides

      Background:
      The purpose of this study is to compare Conformity (CI) and Homogeneity (HI) Index values of different IMRT techniques (VMAT vs non-VMAT) in lung cancer patients who treated in our clinic.

      Methods:
      We evaluated 37 locally advanced lung cancer patients who previously treated as curatively with IMRT (Volumetric and non-Volumetric (Forward or Inverse) planning techniques) in Eskisehir Osmangazi University Hospital between 2015 July and 2017 September. For each patient, CI and HI values were calculated retrospectively using different formulas according to literature.

      Results:
      Median prescribed dose was 64Gy (range: 57.5–66). When we compared CI values, there was a statistically different result in favor of VMAT only which is calculated according with Paddick's formula. When we compared HI values, there were no statistically different results between IMRT techniques (Table).Table:Calculated CI and HI values of VMAT and non-VMAT techniques

      VMAT(n = 21)Non-VMAT(n = 16)p
      Dose (Gy)Min57.5057.50NS
      Max64.0066.00
      Mean61.5261.37
      Median64.0061.00
      CI (RTOG)PIV/TVMin1.07061.2558NS[a]
      Max1.86173.9451
      Mean1.47871.9063
      Median1.41931.6721
      CI (Knöös)TV~PIV~/TVMin0.94210.9016NS[b]
      Max1.00001.0000
      Mean0.99020.9833
      Median0.99840.9881
      CI (Paddick)TV~PIV~[2]/(TV × PIV)Min0.53720.25350.040[a]
      Max0.84390.7895
      Mean0.67840.5656
      Median0.70460.5830
      HI (Shaw et al.)D~max~/D~mean~Min1.07531.0931NS[b]
      Max1.37991.2472
      Mean1.15551.1620
      Median1.13501.1589
      HI (Salt-Lomax)D~%98~/D~presc~Min0.98460.9814NS[b]
      Max1.04461.0424
      Mean1.02201.0157
      Median1.02631.0161
      HI (Paddick)D~%2~/D~%98~Min1.03091.0484[b]
      Max1.13151.1092
      Mean1.06321.0769
      Median1.05631.0737
      HI (ICRU 83)(D~%2~–D~%98~)/D~%50~Min0.03040.0472NS[b]
      Max0.12170.1015
      Mean0.06080.0731
      Median0.05470.0707
      PIV = Prescribed isodose volume TV = Target volume TV~PIV~ = Intersection of Prescribed isodose volume and Target volumeaMann-Whitney TestbStudent T-Test

      Conclusions:
      The VMAT technique is superior to the non-VMAT techniques in achieving the conformity index which is obtained by the Paddick's approach that a more complex and comprehensive formula is used to calculate the conformity index.

      Clinical trial identification:


      Legal entity responsible for the study:
      Eskisehir Osmangazi University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      117P - Feasibility of moderate hypofractionated image-guided thoracic irradiation for locally advanced node-positive non-small cell lung cancer patients with very limited lung function (Now Available) (ID 626)

      12:50 - 12:50  |  Presenting Author(s): F. Manapov  |  Author(s): O. Roengvoraphoj, J. Taugner, M. Dantes, C. Wijaya, C. Belka, C. Eze

      • Abstract
      • Slides

      Background:
      To determine the feasibility of moderate hypofractionated image-guided thoracic irradiation (Hypo-IGRT) in locally advanced node-positive non-small cell lung cancer patients with very limited pulmonary function.

      Methods:
      Eight patients with NSCLC stage IIIA-C and highly diminished pulmonary function (FEV1 ≤ 1.0 L and/or DLCO-SB ≤ 40% and/or long-term oxygen therapy) were treated with Hypo-IGRT. Planning was based on 18F-FDG-PET/CT and 4-D computed tomography (4D-CT). GTV included primary tumor and involved lymph nodes (short-axis ≥1 cm and/or PET-positive). CTV was not generated. ITV was defined through the overlap of GTVs on 10 phases of 4D-CT. Isotropic margin of 5 mm was added to ITV to generate the PTV. Hypo-IGRT was delivered to a total dose of 45 Gy (ICRU) in 15 fractions under daily image-guidance.

      Results:
      Eight patients completed Hypo-IGRT. The median follow-up was 20 months. The median age was 64 years. Two (25%), 4 (50%) and 2 (25%) patients presented with NSCLC stage IIIA, IIIB and IIIC, respectively. There were 7 (88%) patients with ECOG 2 and 1 (12%) patient with ECOG 3. Three patients received chemotherapy prior to Hypo-IGRT. Median initial DLCO-SB was 33.3% (range: 13.3–54) and the median initial FEV1 was 1L/39% predicted (range: 0.78–1.26L/28–60%). The median PTV was 226.9 cm³ (range: 100.17–379.80). The median overall survival (OS), progression-free survival (PFS) and distant metastasis-free survival (DMFS) for the entire cohort were not reached. Mean OS was 42 months (95% CI: 31.7–52.6). Mean PFS was 38 months (95% CI: 17.8–45.7). The 1- and 2-year OS rates were 100% and 87.5%. The 6- and 12- months PFS rates were 100% and 62.5%. Three patients developed local failure. Median mean lung dose was 9.4 Gy (range: 5.3–11.6). V15 and V20 for both lungs were 22% (range: 10–25) and 15% (range: 6–19). Median mean esophageal dose was 12.76 Gy (range: 2.1–26.7). There was no case of radiation pneumonitis. Four patients developed grade 2 acute radiation esophagitis.

      Conclusions:
      This analysis shows that Hypo-IGRT can be considered in locally advanced node-positive NSCLC patients with very limited pulmonary function who are inadequate for conventional therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital, LMU Munich

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      118P - A comparative study of sequential chemoradiation vs concurrent chemoradiation vs concurrent chemoradiation followed by consolidation chemotherapy in unresectable NSCLC (ID 200)

      12:50 - 12:50  |  Presenting Author(s): A. Datta  |  Author(s): K.B. Choudhury, A. Majumdar

      • Abstract

      Background:
      To compare the Response Rates, acute and late toxicities and Time to Progression (TTP) in the 3 arms.

      Methods:
      Patients with unresectable NSCLC were prospectively analysed from August 2011 – September 2013. They were randomized to 3 arms of Sequential Chemoradiation (ARM A), Concurrent Chemoradiation followed by consolidation chemotherapy (ARM B) and Concurrent Chemoradiation (ARM C). Arm A received 2 cycles induction while Arm B received 2 cycles consolidation chemotherapy. Arm C was the Control arm. Drugs used were Paclitaxel (175 mg/m[2])/Carboplatin (AUC 5). Their dose in concurrent setting were 45 mg/m[2] and AUC 2 respectively. The dose of radiation used was 60 Gy in 30 fractions. Response Rate was assessed using RECIST v1.1 criteria while acute and late toxicities were assessed by RTOG/EORTC (skin, upper GI, Dysphagia) and CTCAE v4.0 (dyspnoea, haematological and peripheral neuropathy) criteria. For TTP Kaplan Meier survival analysis with Log rank was used for inter-treatment comparisons.

      Results:
      72 patients (male = 68, female = 4) were randomised in the 3 arms in 1:1:1 allocation of whom 64 patients (all males) were available for analysis (Arm A = 20, Arm B = 21, Arm C = 23). All the 3 arms were comparable in their baseline parameters. Chi Square test analysis showed non-significant statistical difference in response rates between the 3 arms. TTP was superior with median PFS of 7 months (95% C.I. 5.2mths, 8.8 months) in Arm C (Concurrent Chemoradiation arm). Grade 2 & 3 acute skin toxicities in Arms B (33% & 9.5%) & C (21.7% & 4.3%) were more (p < 0.001). Grade 2 & 3 upper GI toxicity in Arms B (57.1% & 28.6%) & C (87.0% & 13%) were higher (p < 0.001). Grade 2 & 3 Haematological toxicity was more (p < 0.001) in Arms B (42.9% & 57.1%) & C (52.2% & 47.8%). Grade 2 & 3 Dysphagia was more (p < 0.001) in Arms B (57.1% & 28.6%) & C (87% & 13%). Peripheral neuropathy was higher in Arms B & C (p < 0.001) with Arm C having the maximum Grade 2 neuropathy (78.3%). There was no difference among the late skin toxicity among the 3 arms (p = 0.869).

      Conclusions:
      There was no difference in the response rates across the 3 arms. The Concurrent Chemoradiation arm had the maximum Time to progression with maximum acute toxicities.

      Clinical trial identification:


      Legal entity responsible for the study:
      IPGMER and SSKM Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      119P - Cisplatin/etoposide or paclitaxel/carboplatin with concurrent radiation therapy in stage IIIB non-small cell lung cancer: A one-year phase II trial at a low resource setting (Now Available) (ID 489)

      12:50 - 12:50  |  Presenting Author(s): M. Shuayb  |  Author(s): M.S.J.R. Shahi, M.M. Hossen

      • Abstract
      • Slides

      Background:
      Concurrent chemoradiation therapy (CCRT) has been established as the standard of care for patients with stage IIIB non-small cell lung cancer (NSCLC). The comparative benefits of two extensively used regimens: cisplatin/etoposide (PE) and paclitaxel/carboplatin (PC), with thoracic radiation therapy (TRT) remain largely undefined. To explore the distinct advantage of multiagent CCRT with taxane (PC) over CCRT with traditional platinum doublet therapy (PE), an interventional study was designed and conducted at the Department of Radiotherapy of Dhaka Medical College Hospital.

      Methods:
      Patients were randomly assigned to receive the following treatments: PE arm, cisplatin (50 mg/m[2]) on days 1,8 and 29 and etoposide (50 mg/m[2]) on days 1–5 and 29–33 plus 45 Gy of TRT; PC arm, weekly paclitaxel (45 mg/m[2]) and carboplatin (AUC = 2) plus 45 Gy of TRT. RECIST criteria, Kaplan-Meier method and RTOG morbidity criteria were employed to determine treatment response, survival and acute & late toxicity respectively. Student's t test, Chi squared test, Fisher's exact test and Log rank test were used to compare across the arms in different aspects.

      Results:
      A total of 60 patients (30 in each arm) were randomized. The objective response and disease control rate were 51.85% in PE and 60.71% in PC (P = 0.61) and 85.19% in PE and 78.57% in PC (P = 1) respectively. Treatment with PE had significant survival advantage in terms of 1-year overall survival (60.6% vs. 42.9%; P = 0.04) and 1 year progression free survival (44.6% vs. 31.0%; P = 0.04). Grade 3/4 neutropaenia occurred in 20% of PE and 16.67% of PC patients (P = 1). Acute oesophagitis (grade 1/2, 53.33% vs. 70%; grade 3/4, 13.33% vs. 30%, P < 0.01) and upper GI symptoms (grade 1/2: 63.33 vs. 100%; P < 0.01) were significantly more pronounced in PC group. PC arm developed significantly greater rate of pneumonitis (grade 1/2, 16.67% vs. 36.67%; grade 3/4, 0% vs. 10%; P = 0.02). PC arm had 4 treatment related deaths.

      Conclusions:
      There was substantial incongruity between PE and PC based CCRT regimen in the treatment of stage IIIB NSCLC. With meticulous patient selection and follow up, CCRT with PE can be an acceptable protocol in this setting.

      Clinical trial identification:
      German Clinical Trials Register. DRKS00012599. Date 20.07.2017

      Legal entity responsible for the study:
      Ethical Review Committee of Dhaka Medical College Hospital, BD on 22.10.2014 (Ethical Clearance Certificate # ERC-DMC/ECC/2014/168). Registered by Bangladesh College of Physicians and Surgeons (BCPS) on 11.11.2014 (Registration # PSN-0037)

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      120P - Optimal chemotherapy regimen for concurrent chemo-radiotherapy of locally advanced unresectable stage III non-small cell lung cancer (NCSLC) (Now Available) (ID 597)

      12:50 - 12:50  |  Presenting Author(s): N. Jokhadze  |  Author(s): I.Z. Kiladze, L. Katselashvili, M. Kacharava, N. Jankarashvili, I. Zumbadze, N. Kalandarishvili, T. Melkadze, E. Gogua

      • Abstract
      • Slides

      Background:
      Combination platinum-based chemo-radiotherapy is considered as a standard treatment of locally advanced unresectable NSCLC. The exact sequence, optimal and effective chemotherapy regimens to be used in combination with radiation therapy are all still subject of debates. Cisplatin/Etoposide (EC) and Paclitaxel/Carboplatin (PC) both demonstrated efficacy in the treatment of locally advanced unresectable NSCLC and are considered as most widely used regimens in this setting. However, there are very limited number of randomized trials comparing EC vs PC face to face.

      Methods:
      In our open labeled prospective study a total 52 patients were randomized (1:1) to receive 60–66 Gy intensity-modulated radiotherapy with either Etoposide 50 mg/m[2] d1-5, 29–33 and Cisplatin 50 mg/m[2] d 1, 8, 29 and 36 (ARM1) or Paclitaxel 45 mg/m[2] weekly and Carboplatin AUC = 2 mg/mL/min weekly (ARM2). Study inclusion criteria were: unresectable stage IIIA/IIIB/IIIC, NSCLC proved by pathology; treatment naïve, age 18–70; ECOG ≤ 2; no serious comorbidities; no contraindications to chemo-radiotherapy, chest CT in recent 2 weeks. The primary endpoint of the study was progression-free survival and 1year survival. Secondary endpoints were toxicity profiles of two regimens.

      Results:
      With median follow-up time 48 months 1-year survival was significantly higher in the EP arm than that of PC arm. Estimated difference was 27% (p value = 0.05). PFS was also higher in EP arm compared to PC arm (46% vs 5%, p value = 0.05). No significant difference in incidence of Grade >2 radiation pneumonitis and Grade >2 radiation esophagitis was observed among two arms. Incidence of Grade >3 hematologic toxicities was also similar in two groups.

      Conclusions:
      CP used concurrently with thoracic radiation showed better 1year survival and PFS than weekly PC. In terms of toxicity profiles these two regimens did not show statistically significant difference in our study. To determine which regimen is associated with better long term survival or PFS larger randomized studies with longer follow-up periods are needed.

      Clinical trial identification:
      GYOG0005

      Legal entity responsible for the study:
      Georgian Young Oncologists Group

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      121P - Primary surgery followed by chemotherapy versus definitive concurrent chemoradiotherapy in locally advanced non-small cell lung cancer (Now Available) (ID 542)

      12:50 - 12:50  |  Presenting Author(s): I.Z. Kiladze  |  Author(s): V. Kuchava, N. Joxadze, L. Katselashvili, T. Melkadze

      • Abstract
      • Slides

      Background:
      Approximately 30–35% of patients present with locally advanced NSCLC(LAD-NSCLC) The majority of them can be treated with a combination of chemo. and radiation; however, a subset of stage III pat. are considered surgical candidates may require a modification of this plan. The purpose of this study is to compare effectiveness of primary surgery and chemotherapy versus chemoradiation in patients with LAD-NSCLC. To evaluate the survival parameters and treatment complications.

      Methods:
      NSCLC pat. with clinical stage IIIA and selected IIIB from 2014 to 2016 were identified retrospectively in two Institutions. Medical records reviewed. All patients were grouped in 2 treatment arms. Surgical arm: surg.+adj.chemotherapy and ChRT arm: Definitive Chemoradiotherapy.

      Results:
      The medical records of 75 patients (39 Surgical and 35 ChRT arm) were reviewed. More than 1/2 in both arms were with scc (51% and 61%). The type of surgery was 15 lobec- and 24 pneumonectomies with complete mediastinal l/node dissection. In CHRT arm G3-4 hematology toxicity occurred in 19pat. and 6 patients had acute non-hematology toxicity(G3). No acute G 4 radiation toxicity developed. In surg. arm only 1 severe bleeding developed and reoperation was performed. 2 patients had wound healing problems. 5 treatment related deaths occurred: 3 in the surgical arm (2 pulmonary embolus and 1 cardiac complication) and 2 patients in chemoradiation arm(2 PE). The 1-year survival rate was 69% vs 61% in surg. and ChRT arms respectively, which was not statistically significant (p = 0.4604). No difference in the 1-year survival was observed in Stage IIIA 68% vs 58% (p = 0.5627) and in Stage IIIB 72% vs 62% (p = 0.5540) between arms. No difference on median survival time (17.5 vs 16.8 m).

      Conclusions:
      Treatment effectiveness, complications and 1-year survival rate were equal between the surgical and ChRT arms. These results seem to indicate primary surgery as the treatment of choice for stage IIIA and selected patients with stage IIIB NSCLC, whenever a complete resection is thought to be technically feasible and the patient's conditions compatible with the extent of the planned surgery. Further results of 3- and 5-year survival rates are awaited.

      Clinical trial identification:
      GYO LAD-NSCLC 002

      Legal entity responsible for the study:
      Georgian Group of Young Oncology

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      122P - Pathological response rates after induction radio-chemotherapy in stage III NSCLC (ID 229)

      12:50 - 12:50  |  Presenting Author(s): F. Casas  |  Author(s): D. Sanchez, D. Martínez, M. Boada, N. Vilariño, R. Martin, P. Paredes, I. Volmer, K.S. Cortés, M. Parera

      • Abstract

      Background:
      This retrospective analysis was designed to assess pathological tumor response to radical (60 Gy) radio-chemotherapy (RTx/CTx) and surgery in stage III NSCLC (TNM 8th edition).

      Methods:
      We included patients treated from a phase II trial and a few patients with salvage surgery after multidisciplinary decision by Lung Committee (05/2012–10/2017). Induction CTX (CDDP/Carbo doublets) was with concurrent RTx (59–62 Gy). Pathologic N2 were by mediastinoscopy (Md) or endobronchial ultrasound (EBUS). CT scans were performed within 3–4 weeks of treatment. Pathologic examination of residual viable tumor was take in account pT and pN, downstaging and pCR.

      Results:
      Median age: 59 years (range 41–77). Gender: 65.5% was male. Stage: 58.6% IIIA, 37.9% IIIB, 1 3.4% IV (brain metastases treated with radiosurgery). TNM: 27.6% T2b, 24.1% T3, 20.7% T4, 10.3% T2b, 10.3% T1c and 6.9% T1b, 6.9%; N0 86.2% N2,6.9% N3; 3.4% M1b (Brain). Histological: Adenocar 62.1%, 34.5% squamous, 3.4% NSCLC. N2-N3: 62.1% by EBUS, 3.4% by Md, 20.7% needed both. Nodal station distribution: 37.9% 1- N1 and 1-N2; 31% 1-N2, 13.8% 2-N2 and 1-N1, 6,9% 1-N1, 1-N2 and 1-N3, 3,4% 2-N2 and 6,9% N0. CTx: CDDP + Vp16 - 58.75% and CDDP + VNR 27.6%, Toxicites CTx/RTx: Anemia G3 (13.8%), G4 (3.4%); leucopenia, G3 (20.7%), G4 (17.2%); neutropenia, G3 (24.1%), G4 (10.3%). Esophagitis G2 (31%); 27,6% were hospitalised by toxicities. Radiological response: 1 (3.4%) progression, 12 (41.4%) stable, 16 (55.2%) partial response. Surgery: 86.2%; 58.6% lobectomy (LB), 3.4% biLB, 3.4% pneumectomy, 3.4% LB + bronchoplasty, 10.3% LB + ribs resection, 6.9% LB + ribs+ hemivertebral resection. Downstaging: 69%. pCR: 20.7%; Histological tumor viability (Tv): 24.1% pCR, 20.7% a 5% Tv, 17.2% between 10–15% Tv, 13.7% between 20 and 30% Tv, 6.8% between 40 and 50% Tv, 3.4% with 90% Tv; Histological lymph node Nv: 72.4% pCR, 6.8% between 5 and 10% Nv, 6.8% between 40 and 100% Nv.

      Conclusions:
      RTx/CTx and surgery in stage III NSCLC presents high rates of downstaging with a high percentage of toxicities that required an experienced multidisciplinary team able to anticipate and treat these conditions. We consider the different pathological responses must be correlated with an adequate and long follow-up that led us to analyze possible recurrence patterns with an impact on the management, survival and quality of life of these patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Clinic

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      124P - Cardiac events in stage III non-small cell lung cancer (NSCLC): An attention shift to cardio-oncology collaboration (Now Available) (ID 587)

      12:50 - 12:50  |  Presenting Author(s): J. Degens  |  Author(s): D. De Ruysscher, B. Kietselaer, A. Schols, A. Dingemans

      • Abstract
      • Slides

      Background:
      Recently it was shown that 15% of stage III NSCLC patients, treated with thoracic radiotherapy in dose-escalated trials, suffer from late cardiac events.[1] However, the prevalence of pre-existent cardiac co-morbidity in daily clinical practice in these patients and the development of cardiac events during follow-up is still unclear. As these patients are treated with curative intent and have a five survival rate 25%, there is need for a study to investigate the development of cardiac events and the relation with pre-existent cardiac co-morbidity.

      Methods:
      In this retrospective cohort study a thorough patient file search was carried out in 153 patients diagnosed with stage III NSCLC, treated with (chemo-)radiotherapy between 2006 and 2011 in our center. Primary endpoint was the incidence of pre-existing cardiac comorbidity and relation with the development of serious cardiac events, defined as CTCAE 4.0 grade >2, within five years after a curative treatment with (chemo)radiotherapy. Cardiovascular risk prediction was calculated for each patient according to WHO/ISH, which indicates the 10-year risk of a serious cardiovascular event.

      Results:
      Pre-existing cardiac comorbidity was seen in 46 patients (31.1%) with most frequently myocardial infarct/ coronary artery disease (9.8%) and arrhythmia (7.8%). WHO/ISH cardiovascular risk prediction was > 10% in 60.1% of the patients. Serious cardiac events appeared in 26% of the patients in the second year after treatment (20.3%). Most frequent cardiac events were arrhythmia (9.2%), myocardial infarction (6.5%), congestive heart failure (4.6%) and pericardial effusion (4.6%).Table:Serious cardiac events within subgroups of the study population

      Total N = 153Cardiac history N = 46No cardiac history N = 102WHO/ISH Cardiac event risk > 10% N = 78
      Cardiac events CTCAE-score > 2 (N/%)38 (26%)14 (30%)24 (23%)18 (23%)
      Missing (N)7---
      Median time to event 1–2 years (N/%)17 (11.6%)6 (13%)11 (10.8%)16 (20.5%)


      Conclusions:
      In daily clinical practice 1/3th of patients with stage III NSCLC, treated with (chemo-)radiotherapy, have pre-existing cardiac comorbidity. In addition, 26% develop a serious cardiac event during follow-up, even in patients without cardiac history. Therefor it is important to identify patients at risk in order to prevent these cardiac events.

      Clinical trial identification:


      Legal entity responsible for the study:
      Maastricht University Medical Center

      Funding:
      Has not received any funding

      Disclosure:
      D. De Ruysscher: Consulting or advisory role to disclose: Bristol-Myers Squibb I have research funding to disclose: Brsitol-Myers-Squibb (BMS). A-M. Dingemans: Reports other from Roche/Genentech, other from MSD Oncology, other from AstraZeneca, other from Pfizer, other from Lilly, other from Boehringer Ingelheim, other from Bristol-Myers Squibb, other from Clovis Oncology, outside the submitted work. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      125P - Brain metastases in patients with stage N2 non-small cell lung cancer (Now Available) (ID 347)

      12:50 - 12:50  |  Presenting Author(s): C. Lowrie  |  Author(s): E. McCully, S. Ansel, P. McLoone, N. Mohammed

      • Abstract
      • Slides

      Background:
      At diagnosis, 60% of NSCLC patients (pts) have potentially curable locally advanced disease with N0, N1 or N2 lymph node status. Guidelines recommend surgical resection of locally advanced NSCLC followed by adjuvant chemotherapy (SIGN 370). The selection of pts for curative treatment requires a PET scan and pathological nodal staging. Accurate clinical staging of lung cancer ensures best treatment. NICE guidelines recommend consideration of brain imaging in pts selected for treatment with curative intent especially if N2 disease is diagnosed. In clinical practice, many relapse within 1 year despite combination treatment. Brain metastases (mets) are common in NSCLC but there are now more treatment options available. We investigated the frequency of brain mets in pts with post-operative pN2 (pathological N2) disease.

      Methods:
      A retrospective audit of West of Scotland Cancer Network was performed of pts with pN2 NSCLC between September 2011 and November 2016. Data collection included demographics, staging investigations, and clinical outcomes using electronic patient records.

      Results:
      We identified 169 NSCLC pts (86 females, 83 males) with pN2 disease. Clinical staging showed 73 pts (43%) cN0, 42 (25%) cN1 and 53 (31%) cN2. 32 pts (19%) had pre-treatment brain imaging. This was higher among pts with cN2 disease (34%). Imaging modality was primarily CT, with MRI used for 9 pts (28%). Follow-up imaging included brain in another 12 (8%). Distant relapse occurred within 3 months of treatment for 11 pts of whom 2 had symptomatic brain mets. Overall 84 (50%) pts experienced distant relapse of which 23% (19/84) had brain mets. Median overall survival of all pts was 25.3 (95%CI 18.5–29.8) months. There was no statistically significant difference in the survival of pts with brain mets compared to other distant mets; median survival 20.3 (8.1–25.3) months vs 17.8 (14.5–21.8) months respectively, log-rank test p = 0.56.

      Conclusions:
      This audit shows that only 34% of cN2 pts underwent pre-operative brain imaging. 1% relapsed within 3 months of surgery with brain mets and overall 23% were diagnosed with brain mets. The median overall survival for this group is comparable with the published series and there was no survival difference between pts with brain mets and other distant metastatic disease.

      Clinical trial identification:


      Legal entity responsible for the study:
      NHS Greater Glasgow and Clyde

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      126TiP - SAKK 16/14: Anti-PD-L1 antibody durvalumab (MEDI4736) in addition to neoadjuvant chemotherapy in patients with stage IIIA(N2) non-small cell lung cancer (NSCLC): A multicenter single-arm phase II trial (Now Available) (ID 638)

      12:50 - 12:50  |  Presenting Author(s): S.I. Rothschild  |  Author(s): A. Zippelius, S. Savic Prince, M. Gonzalez, W. Weder, A. Xyrafas, C. Rusterholz, M. Pless

      • Abstract
      • Slides

      Background:
      Improving the outcome of locally advanced non-small cell lung cancer (NSCLC) is one of the major challenges in thoracic oncology. SAKK substantially contributed to establish a standard of care for patients with stage III NSCLC: The trial SAKK 16/96 established neoadjuvant chemotherapy with three cycles of cisplatin and docetaxel. The randomized trial SAKK 16/00 showed no benefit by adding neoadjuvant radiotherapy as third treatment modality. Our results consistently showed a 5-year overall survival (OS) of 37%. Recently, the PACIFIC trial showed significantly improved progression-free survival for durvalumab as consolidation therapy after definitive chemoradiotherapy in unresectable stage III NSCLC.

      Trial design:
      This is a single-arm phase II clinical trial designed to evaluate the addition of perioperative immunotherapy with durvalumab to the previously established standard of care for stage IIIA(N2) patients. Eligible patients with WHO performance status 0–1 and age of 18–75 years must have pathologically proven NSCLC stage IIIA(N2) (T1-3 N2 M0) according to the 7th edition of the TNM classification, irrespective of histological subtype, genomic aberrations or PD-L1 expression status. Tumor tissue has to be available for the mandatory translational research. Patients whose tumor is deemed resectable at diagnosis receive three cycles of chemotherapy with cisplatin 100 mg/m[2] and docetaxel 85 mg/m[2] every three weeks followed by two cycles of durvalumab 750 mg every two weeks. Following surgery, patients will be treated with durvalumab 750 mg every two weeks for 12 months. The primary endpoint of the trial is event-free survival at 12 months. Secondary endpoints include OS, objective response, nodal down-staging, complete resection, pattern of recurrence and toxicity. Additionally, a large translation research program accompanies the trial investigating potential predictive biomarkers of anti-PD-L1 therapy. Based on the data of first 25 operated patients and given that the results showed that their 30-day post-operative mortality is less than 10%, according to the decision rule described in the protocol of the trial there is no reason for further detailed safety analysis (evaluated by an IDMC) and thus shall continue as per protocol.

      Clinical trial identification:
      NCT 02572843

      Legal entity responsible for the study:
      Swiss Group for Clinical Cancer Research

      Funding:
      Swiss Group for Clinical Cancer Research; AstraZeneca; Rising Tide Foundation, Gateway for Cancer Research

      Disclosure:
      S.I. Rothschild: SAKK 16/14 study is supported by AstraZeneca. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      127TiP - Phase I study of neo-adjuvant Stereotactic Body Radiotherapy (SBRT) in operable patients with borderline resectable locally advanced NSCLC (LA-NSCLC) (LINNEARRE I STUDY: NCT02433574) (ID 493)

      12:50 - 12:50  |  Presenting Author(s): T. Tsakiridis  |  Author(s): N. Isfahanian, N. Nguyen, J. Wright, W. Hanna, A. Swaminath, Y. Shargal, J. Agzaraian, M. Wierzbicki, T. Chow

      • Abstract

      Background:
      Patients with NSCLC who undergo incomplete resection (R1) have significantly worse survival compared to those with complete resection (R0). The efficacy of Stereotactic Body Radiotherapy (SBRT) in treating small lung tumors is established, but its use as neo-adjuvant therapy in locally-advanced (LA)-NSCLC is not investigated. We hypothesized that pre-operative SBRT can be done safely and could improve rates of R0 resection in LA-NSCLC. This concept could introduce a rapid, convenient and less toxic neoadjuvant therapy in LA-NSCLC to help improve rates of complete resection. LINNEARRE I is single institution phase I study that investigates the safety and feasibility of delivering, timely, neo-adjuvant hypo-fractionated RT of escalating Biological Equivalent Doses (BED) approaching those of SBRT.

      Trial design:
      A total of twenty patients with biopsy proven T3-T4, N0-1, M0, NSCLC will be enrolled. Patients must be deemed as medically operable, by a thoracic surgeon, but at risk of
      Clinical trial identification:
      NCT02433574

      Legal entity responsible for the study:
      Juravinski Cancer Center

      Funding:
      Juravinski Cancer Center Foundation

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      145P - Impact of next-generation sequencing on survival in lung cancer (Now Available) (ID 595)

      12:50 - 12:50  |  Presenting Author(s): S. Geva  |  Author(s): A. Belilovski Rozenblum, L.C. Roisman, M. Ilouze, E. Dudnik, A. Zer, O. Rotem, N. Peled

      • Abstract
      • Slides

      Background:
      Next-generation sequencing (NGS) enables a comprehensive genomic analysis of lung cancer patients. It has uncovered many novel genetic abnormalities and identified actionable genomic alterations in lung tumors that previously tested “negative” by conventional non-NGS tests. In this study, we evaluated the clinical impact of NGS on overall survival of advanced non-small cell lung cancer (NSCLC) patients.

      Methods:
      In this retrospective study, 234 consecutive stage IIIb/IV NSCLC patients who performed hybrid capturing NGS were enrolled in Israel, between 2011–2017. Hybrid capture-based NGS was performed by Foundation Medicine and Guardant 360[TM] if tissue was not available.

      Results:
      234 consecutive NSCLC patients were included in this study. 62% (145/234) performed tissue NGS and 38% (89/234) performed liquid NGS. Median age at diagnosis was 63 years. 84% had adenocarcinoma. 37% were never-smokers. The patients were divided into 4 groups according to the identification of NCCN-approved actionable genomic alterations on NGS analysis: Group A did not have an actionable target; Group B discovered an actionable target but did not receive targeted treatment, due to high performance status or preference to use immunotherapy for PD-L1 positive patients; Group C received targeted therapy subsequent to NGS analysis, among them 75 received treatment according to NCCN guidelines, 9 off-protocol and 7 received immunotherapy due to high tumor mutation burden found on NGS analysis; Group D received targeted therapy due to standard molecular tests, as PCR, IHC and FISH performed prior to NGS. Median Overall survival (OS) is summarized in the Table (p = 0.0278).Table: (Abstract 145P)Overall Survival

      Total Cohort (n = 234)Group A 46% (108/234)Group B 5.5% (13/234)Group C 39% (91/234)Group D 9.5% (22/234)
      Median OS20.9 months (95% CI: 19.0–28.7)17.8 months (95% CI: 13.1–24.2)10.8 months (95% CI: 4.1–NA)25.7 months (95% CI: 19.9–41.7)37.7 months (95% CI: 26.7–NA)


      Conclusions:
      This study evaluates the clinical impact of comprehensive NGS testing by demonstrating the survival advantage of patients with actionable targets discovered through NGS. Comprehensive tissue and liquid-based NGS have revealed targeted treatment options for a significant number of patients. Overall Survival of patients treated with tailored therapy subsequent to NGS analysis was positively impacted in comparison to patients without an actionable target, only exceeded by patients who received targeted treatment subsequent to upfront standard molecular tests.

      Clinical trial identification:


      Legal entity responsible for the study:
      Soroka Cancer Center, Ben-Gurion University, Beer Sheva, Israel.

      Funding:
      Has not received any funding

      Disclosure:
      S. Geva: Travel grant from Teva Pharmaceuticals, Honorarium from Guardant Health. L.C. Roisman: Lectures fees: Roche, MSD, Pfizer, Astrazenca. M. Ilouze: Honorarium from Pfizer, MSD, Roche and Takeda. A. Zer: Personal fees from Roche, grants and personal fees from BMS, personal fees from AstraZeneca, personal fees from BI, outside the submitted work. N. Peled: Advisor & Honorarium from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, FoundationMedicine, Gaurdant360, MSD, Novartis, NovellusDx, pfizer, Roche, Takeda. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      146P - The clinical impact of comprehensive cfDNA genomic testing in lung cancer (Now Available) (ID 566)

      12:50 - 12:50  |  Presenting Author(s): S. Geva  |  Author(s): A. Belilovski Rozenblum, R. Grinberg, A. Dvir, L. Soussan-Gutman, L.C. Roisman, E. Dudnik, A. Zer, O. Rotem, N. Peled

      • Abstract
      • Slides

      Background:
      Next-generation sequencing (NGS) of cell-free circulating tumor DNA (cfDNA) enables a non-invasive option for comprehensive genomic analysis of lung cancer patients. Currently there is insufficient data in regard to the impact of cfDNA analysis on clinical decision making. In this study, we evaluated the clinical utility of cfDNA sequencing on treatment strategy and progression-free survival in non-small cell lung cancer (NSCLC) patients.

      Methods:
      In this retrospective study, data was collected from files of 116 NSCLC patients monitored between the years 2014–2017 in Israel. Plasma samples from stage IIIb/IV NSCLC patients were analyzed by a commercial test (Guardant 360), using hybrid capture, single molecule barcoding and massively parallel paired-end synthesis to sequence a targeted gene panel. This test allows the detection of somatic alterations such as point mutations, indels, fusions and copy number amplifications.

      Results:
      116 consecutive NSCLC patients were included in this study. Median age at diagnosis was 63 years, male:female ratio was 1:1.7. 40% (47/116) were never-smokers, 83% (96/116) had adenocarcinoma. 41.4% (48/116) were tested before 1[st] line therapy (Group A), 34.5% (40/116) upon progression on chemotherapy or immunotherapy (Group B1) and 24.1% (28/116) upon progression on EGFR TKIs (Group B2). The most common genes were EGFR sensitizing mutations (25.9%, 30/116), MET amplifications and/or exon 14 skipping mutations or resistance point mutation (9.5%, 11/116) and EGFR T790M mutations (6.9%, 8/116). Clinical outcome of cfDNA analysis and targeted therapy for the entire cohort and for each group are summarized in the Table.Table: (Abstract 146P)Clinical Outcome of cfDNA Analysis and Targeted Therapy

      Total (n = 116)Group A (n = 48)Group B1 (n = 40)Group B2 (n = 28)
      Drug-associated actionable Mutations (On/Off Lable)65% (75/116)65% (31/48)52.5% (21/40)82% (23/28)
      Lung Cancer Related Actionable Mutations (NCCN guidelines)41% (48/116)31% (15/48)32.5% (13/40)71% (20/28)
      Tretmanet change (Impact on Decision)26% (30/116)23% (11/48)25% (10/40)32% (9/28)
      Response Evaluable93% (28/30)82% (9/11)100% (10/10)100% (9/9)
      Response not Evaluable7% (2/30)18% (2/11) Early cessation of treatment d/t toxicity0% (0/10)0% (0/9)
      Response Assessment (RECIST): CR4% (1/28)0% (0/9)0% (0/10)11% (1/9)
      Response Assessment (RECIST): PR39% (11/28)44% (4/9)30% (3/10)44% (4/9)
      Response Assessment (RECIST): SD32% (9/28)56% (5/9)20% (2/10)22% (2/9)
      Response Assessment (RECIST): PD25% (7/28)0% (0/9)50% (5/10)22% (2/9)
      Objective Response Rate43% (12/28)44% (4/9)30% (3/10)55.5% (5/9)
      Disease Control Rate75% (21/28)100% (9/9)50% (5/10)78% (7/9)
      Median Duration of Treatment5 months (6/28 ongoing)9 months (4/9 ongoing)3.5 months (0/10 ongoing)4 months (2/9 ongoing)
      Durable Disease Control Rate (over 4 months)43% (13/30)64% (7/11)20% (2/10)44% (4/9)
      Median PFS3.6 months7.3 months2.5 months3.3 months


      Conclusions:
      This study extends the evidence for clinical utility of comprehensive NGS testing by demonstrating durability of response to plasma-detected genomic alterations. cfDNA NGS changes treatment decisions in a significant number of patients in this retrospective study. It also has the potential to reduce undergenotyping of advanced NSCLC patients, while reducing costs and complications of biopsies, and facilitating more precise use of targeted therapy as well as immunotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Soroka Cancer Center, Ben-Gurion University, Beer Sheva, Israel.

      Funding:
      Has not received any funding

      Disclosure:
      S. Geva: Travel grant from Teva Pharmaceuticals, Honorarium from Guardant Health. A. Dvir, L. Soussan-Gutman: Employee of Oncotest (subsidiary of Teva pharmaceuticals), the distributor of Guardant360 in Israel. L.C. Roisman: Lectures fees: Roche, MSD, Pfizer, Astrazenca. A. Zer: Personal fees from Roche, grants and personal fees from BMS, personal fees from AstraZeneca, personal fees from BI, outside the submitted work. N. Peled: Advisor & Honorarium from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Eli Lilly, FoundationMedicine, Gaurdant360, MSD, Novartis, NovellusDx, pfizer, Roche, Takeda. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      147P - Clinical testing of ctDNA from NSCLC patients using A 17-gene liquid biopsy mutation panel (Now Available) (ID 590)

      12:50 - 12:50  |  Presenting Author(s): D.A. Eberhard  |  Author(s): J. Bennett, D. Davison, C. Hammond, A. Petty, J. Pluenneke, A. Dei Rossi, G. Alexander, D. Paragas, M. Lopatin

      • Abstract
      • Slides

      Background:
      The Oncotype SEQ® Liquid Select™ assay is a next-generation sequencing gene panel assay performed as a laboratory-developed test in a central CLIA-certified laboratory by Genomic Health, Inc. Oncotype SEQ® detects clinically relevant genomic alterations in 17 genes using ctDNA isolated from blood plasma. Here we summarize genomic findings from patients (pts) tested after commercial launch of Oncotype SEQ® in June 2016.

      Methods:
      126 NSCLC pts from 17 community cancer centers in the US were tested with Oncotype SEQ® in routine clinical care. Proprietary technologies and bioinformatics tools were used to identify actionable genomic variants, which were reported to pts’ medical records.

      Results:
      Tested NSCLC pts were stages IV (124), III (1) and II (1). Diagnoses were adenocarcinoma (94), squamous cell carcinoma (19), other (11), or unknown primary (2). Mean age (range) was 69 y (44–90 y), 64% were ≥65 y, 16% were >80 y. 56% were female. Results were reported for 112 (89%) pts. 135 gene variants were detected in 72/112 (64%) pts across 12 genes. 66% of identified variants were SNV, 5% indels, 27% CNV, and 2% fusions. 39 pts had 1 variant, 19 had 2, and 14 had ≥3. In nonsquamous NSCLC, 91/117 (78%) reported variants were SNV/indel/fusions and 26/117 (22%) were CNV; in squamous tumors, these were 7/16 (44%) and 9/16 (56%), respectively. Clinically actionable variants specified in FDA drug labels or in NCCN guidelines for NSCLC were reported for 42/112 (38%) pts in EGFR (13 variants), ALK (3), KRAS (21), MET (7), BRAF (2), and ERBB2 (1). These were observed at allele fractions as low as 0.12% for KRAS G12D and ranging from 0.26% to 22% for EGFR E746_A750del (n = 4). Other reported variants were potentially actionable, either specified in FDA labels of drugs approved for other tumor types, associated with active therapeutic clinical trials in NSCLC, or represented possible germline variants warranting genetic counseling.

      Conclusions:
      Oncotype SEQ® sensitively identifies gene variants that are important to inform optimal management decisions for pts with advanced-stage NSCLC. A retrospective review of medical records to examine concurrent tissue testing, treatments received, and clinical outcomes is ongoing and will be presented.

      Clinical trial identification:
      N/A

      Legal entity responsible for the study:
      Genomic Health, Inc.

      Funding:
      Genomic Health, Inc.

      Disclosure:
      D.A. Eberhard, J. Bennett, D. Davison, C. Hammond, A. Petty, J. Pluenneke, A. Dei Rossi, G. Alexander, D. Paragas, M. Lopatin: Employment and stock ownership: Genomic Health.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      148P - Method comparison for detection of epidermal growth factor receptor (EGFR) T790M mutation in matched biopsied tumour sections and plasma samples (Now Available) (ID 172)

      12:50 - 12:50  |  Presenting Author(s): J. Longshore  |  Author(s): S. Patel, B. Collins, M. Cantarini, S. Jenkins

      • Abstract
      • Slides

      Background:
      The correct identification of EGFR mutations is crucial to ensure that patients are assigned the most appropriate targeted treatment. Few studies have assessed EGFR mutation test performance in detecting the T790M resistance mutation in patients with advanced non-small cell lung cancer (NSCLC), using both biopsied tumour and plasma samples.

      Methods:
      We assessed the performance of the cobas® EGFR Mutation Test (v1 US-IVD, tissue; v2 CE-IVD, plasma), and therascreen® EGFR RGQ PCR Kit (CE-IVD v1, tissue) and EGFR Plasma RGQ PCR Kit (CE-IVD, plasma), using next-generation sequencing (NGS; Illumina MiSeq™) as the reference method. DNA was extracted from formalin fixed paraffin embedded tumour and plasma samples. Matched biopsy tumour tissue and plasma samples were taken from patients with advanced NSCLC and confirmed EGFR-tyrosine kinase inhibitor sensitising mutation, following disease progression on the most recent line of therapy, during screening for the AURA Phase II extension study.

      Results:
      288 tissue samples and 135 matched plasma samples were tested with the cobas® and therascreen tests, and compared with an NGS reference method (Table). When analysing tumour tissue, the two tests were similarly concordant for the detection of T790M, exon 19 deletions, and L858R mutation when using NGS as the reference. In our analysis, however, a higher invalid rate was observed with the therascreen test. When analysing plasma ctDNA, we observed a higher positive percent agreement with the NGS reference using the cobas® plasma test, indicating higher sensitivity for detection of EGFR mutations in plasma ctDNA, with similar specificity.Table: (Abstract 148P)

      Tumour tissue samples*NGS tissue test (reference)
      T790MEx19delL858R
      cobas tissue test n = 285PPA (95% CI)91 (86, 95)98 (95, 100)93 (86, 98)
      NPA (95% CI)98 (91, 100)91 (84, 96)100 (97, 100)
      therascreen tissue test n = 245PPA (95% CI)93 (88, 96)96 (92, 99)91 (82, 96)
      NPA (95% CI)97 (89, 100)89 (81, 95)99 (96, 100)
      Plasma samples*NGS plasma test (reference)
      T790MEx19delL858R
      cobas plasma test n = 135PPA (95% CI)92 (84, 97)97 (91, 100)86 (70, 95)
      NPA (95% CI)92 (80, 98)88 (77, 95)98 (93, 100)
      therascreen plasma test n = 133PPA (95% CI)65 (54, 75)77 (66, 86)76 (58, 89)
      NPA (95% CI)98 (89, 100)97 (88, 100)100 (96, 100)
      *Invalid results were excluded for the calculation of PPA and NPA. CIs were calculated using Clopper-Pearson exact method for binomial proportions. CI, confidence interval; Ex19del, exon 19 deletions; NGS, next-generation sequencing; NPA, negative percentage agreement (using NGS as the reference); PPA, positive percentage agreement (using NGS as the reference).

      Conclusions:
      When a viable test result is obtained using tumour tissue, the cobas® and therascreen tests appear to perform similarly for detection of EGFR mutations. When analysing plasma ctDNA, the cobas® test shows greater sensitivity compared to the therascreen test.

      Clinical trial identification:
      ClinicalTrials.gov NCT01802632

      Legal entity responsible for the study:
      AstraZeneca

      Funding:
      AstraZeneca

      Disclosure:
      S. Patel: Former employee of Qiagen and current employee of, and shareholder in, AstraZeneca. B. Collins: Consultancy fees from AstraZeneca. M. Cantarini: Shareholder in, former full-time employee of, and current part-time contractor for, AstraZeneca. S. Jenkins: Employee of, and shareholder in, AstraZeneca. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      149P - Real-world outcomes with first-line afatinib in EGFR mutant NSCLC adenocarcinoma: A single centre experience exploring effects of dose-reduction (Now Available) (ID 274)

      12:50 - 12:50  |  Presenting Author(s): N. Tokaca  |  Author(s): M. O'Brien, J. Bhosle, N. Yousaf, R. Kumar, S. Popat, D. Walder

      • Abstract
      • Slides

      Background:
      Tyrosine kinase inhibitors of epidermal growth factor receptor (EGFR-TKI) are indicated for first-line treatment of EGFR+ advanced/metastatic NSCLC, having demonstrated superior progression free survival (PFS) and tolerability over chemotherapy in this setting. Afatinib is the only EGFR-TKI to have also shown superior overall survival (OS) over chemotherapy in patients with EGFR 19del mutations, albeit with higher toxicity: 53% and 28% dose reductions (DR) were reported in LUX-LUNG 3 (LL3) and LUX-LUNG 6 (LL6) trials, respectively. Effects of toxicities on treatment delivery and efficacy in the real-world UK population are unknown.

      Methods:
      Retrospective review of outcomes in patients with EGFR+ treatment naïve advanced/metastatic NSCLC treated with afatinib at a single UK centre. Primary endpoint: PFS. Key secondary endpoints: rate of DR; relative dose intensity; toxicities; objective response rate (ORR); PFS according to DR vs no DR; OS overall, by DR vs. no DR, and by mutation type. Survival analyses were performed using Kaplan-Meier methods and compared using the log-rank test.Table:Patient demographics and baseline characteristics (n = 44)

      Patient demographics and baseline characteristic (n = 44)No. (%)
      Age (median, range)63.5 (31–85)
      Gender
       M19 (43.2)
       F25 (56.8)
      Ethnicity
       Caucasian29 (65.9)
       Asian11 (25)
       Other4 (9.1)
      Stage at diagnosis
      2 (4.5)
       IIIA3 (6.8)
       IIIB1 (2.3)
       IV38 (86.4)
      ECOG performance status at start of afatinib
       09 (20.5)
       129 (65.9)
       26 (13.6)
      Comorbidities
       None18 (40.9)
       Mild23 (52.3)
       Moderate3 (6.8)
      EGFR mutation
       Exon 19 del29 (65.9)
       L858R11 (25.0)
       G719X2 (4.5)
       S768I1 (2.3)
       Exon 20 ins1 (2.3)
      Starting dose of afatinib
       40 mg40 (90.9)
       30 mg4 (9.1)


      Results:
      44 patients received first-line afatinib (30–40 mg) between September 2012 and July 2017. Patient characteristics are shown in the Table. 70% patients had at least one DR, 29% during the first cycle. Relative dose intensity was 77.1%. The most common toxicity was diarrhoea (32%), followed by skin rash (22%) and paronychia (18%). Out of 42 evaluable patients, 74% achieved partial response (56% and 67% in LL3 and LL6, respectively). Disease control rate was 93% (LL3: 90%, LL6: 93%). After median follow-up of 26 months, 27/42 patients had disease progression or death on afatinib, 10 patients remained on afatinib and 5 switched to other EGFR-TKI due to intolerable toxicities. mPFS was 12.3mo (LL3: 11.1mo, LL6: 11mo). mPFS in patients with a DR was 22.7mo vs. 12.3mo if no DR (HR 0.69, p = 0.38). Median OS was 31.4mo (LL3: 28.2mo, LL6: 23.1mo). There was no significant difference in mOS for patients with DR vs. no DR (31.4 vs 24.4mo, HR 1.51, p = 0.46). There was a trend towards greater OS for patients with EGFR del19, but not statistically significant (p = 0.23). EGFR T790M testing was available for 19 patients after progression on afatinib, with 6 positive for T790M, all of whom went on to a third-generation EGFR-TKI.

      Conclusions:
      Dose reductions on afatinib are required in a majority of real-world patients, with no significant detrimental impact on efficacy and long-term survival outcomes which, in our cohort, were consistent with trial data.

      Clinical trial identification:


      Legal entity responsible for the study:
      Royal Marsden Hospital

      Funding:
      Has not received any funding

      Disclosure:
      M. O'Brien: Advisory work for BI. J. Bhosle: Honoraria from Boehringer Ingelheim. S. Popat: Consulting/advisory for Boehringer Ingelheim. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      150P - Correlation between erlotinib-induced rash and efficacy in first-line therapy of patients with advanced non-small cell lung cancer (NSCLC) expressing epidermal growth factor receptor (EGFR)-mutation: A prospective, multi-center, open-label, single-arm, phase II study (Now Available) (ID 408)

      12:50 - 12:50  |  Presenting Author(s): M. Gottfried  |  Author(s): S. Keren Rosenberg, J. Dudnik, M. Wollner, J. Bar, A. Onn, O. Frenkel, N. Maimon

      • Abstract
      • Slides

      Background:
      Skin rash is the most common adverse event following erlotinib treatment, reported in about 75% of patients. Several retrospective analyses have suggested that erlotinib-induced skin rash may be associated with better therapeutic outcomes. This phase II TIME (Tarceva In Mutated EGFR patients) study assessed the relationship between erlotinib-induced rash and clinical efficacy in EGFR-mutated advanced NSCLC patients receiving erlotinib first line.

      Methods:
      Patients ≥18 years of age, with EGFR mutated stage IV or inoperable stage IIIB NSCLC, previously untreated with any systemic anti-neoplastic therapy for their advanced stage disease, were enrolled to receive oral erlotinib at an initial daily dose of 150 mg until disease progression or unacceptable toxicity. The primary endpoint was progression-free survival (PFS) according to rash grade.

      Results:
      Sixty patients (41 women, 19 men; median age, 70.7 years) were enrolled in 12 medical centers across Israel. Median PFS from the time of enrollment was 10.4 months (95% CI, 7.5–15.3). The incidence of grade 0, 1, 2, and 3 rash was 6.8%, 22.0%, 39.0% and 32.2%, respectively. Kaplan Meier survival analysis showed that patients with grade 2–3 rash had a statistically significant longer median PFS of 12.0 months (95% CI, 9.3–17.1) compared to patients with grade 0–1 rash who had a median PFS of 5.0 months (95% CI, 2.0–13.5), hazard ratio = 0.38, (95% CI, 0.20–0.71; p = 0.002). Similar results were observed in differentiation between exon 19 and exon 21 mutations.

      Conclusions:
      Albeit a relatively small sample of patients, the results of this prospective study strongly indicate that skin rash during treatment with erlotinib represents a significant predictive factor of efficacy in patients treated for advanced stage NSCLC. These results support previously published retrospective data. Patients might be reassured that rash severity indicates response to treatment. Moreover, an absence of rash as a response to treatment might be a negative prognostic factor in this group of patients.

      Clinical trial identification:
      ClinicalTrials.gov ID: NCT01174563

      Legal entity responsible for the study:
      Roche Pharmaceuticals (Israel) LTD

      Funding:
      Roche Pharmaceuticals (Israel) LTD

      Disclosure:
      M. Gottfried: Advisory Board and corporate sponsored research: Pfizer, BI, MSD, BMS, Roche, AZ, Abbvie. S. Keren Rosenberg: Advisory Boards: Roche, Pfizer, MSD, AZ, BI, Takeda Corporate-sponsored research: Roche. J. Dudnik: Advisory Boards: BI, Astra-Zeneca Corporate-sponsored studies: Roche, Astra-Zeneca, MSD, BMS, BI. M. Wollner: Corporate-sponsored research and consultations fees: Roche, MSD, BMS, AZ, BI, Pfizer Consultation fee: Takeda. J. Bar: Corporate-sponsored research: BI, AZ, Pfizer, Merck, Abbvie, Roche, AZ, MSD, BMS, BI Consulting fees: Roche, Pfizer, Takeda, Abbvie, VBL, BI, AZ, MSD, BMS. A. Onn: Advisory Board: Roche, BI, AZ, MSD. O. Frenkel: Employee of Roche Pharmaceuticals (Israel) Ltd. N. Maimon: Corporate-sponsored research: Roche, MSD, AZ, BI, Pfizer, Abbvie, BMS.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      151P - Maintenance therapy using tyrosine kinase inhibitor (erlotinib) or pemetrexed in metastatic/locally advanced in EGFR mutation-negative lung cancer: Comparison of results (Now Available) (ID 522)

      12:50 - 12:50  |  Presenting Author(s): K. Gupta  |  Author(s): A. Joshi, V. Noronha, S.K. Parthiban, V. Patil, A. Janu, R. Kaushal, K. Prabhash

      • Abstract
      • Slides

      Background:
      Maintenance therapy of locally advanced or metatstatic non-small cell lung cancer (NSCLC) other than predominantly squamous cell histology in patients whose disease has not progressed following 4 to 6 cycles of platinum-based doublet therapy has been standard of care. Pemetrexed or Tyrosine Kinase Inhibitors like Erlotinib have both been used as either continuous maintenance or switch maintenance therapy.

      Methods:
      All patients of NSCLC other than the Squamous Cell carcinoma who have completed either 4 or 6 cycles of platinum and pemetrexed and have either CR/PR/SD on response assessment scan post induction treatment and willing to participate in the study were randomized to receive either pemetrexed or TKI- Erlotinib. Patients were followed up till death. PS and OS were calculated for each arm and indirectly compared.

      Results:
      Two hundred patients were randomized to receive either pemetrexed or erlotinib fron November 2014 to March 2017. Median age of cohort was 55(28–79). Of the 200 patients, 132 patients were male and 68 were female. PS was 0–1 in 195 patients. Smokers constituted 63% (126/200). Majority of patients had stable disease (62.5%), post completion of induction chemotherapy (125/200). Median number of cycles was 4 (4–6). Median PFS in Pemetrexed arm was 4.46 month (95%CI; 3.98 to 4.95), while in Erlotinib arm the median PFS was 4.5 month (95% CI; 3.98 to 4.95) (Hazard ratio = 0.98; 95% CI, 0.714 to 1.369, p-value 0.945). Median OS from starting induction therapy was 16.6 months (15.2–17.9 months) in Pemetrexed arm versus 18.3 months (95% CI 13.75–22.91 months) in Erlotinib arm (p-0.321). [HR = 1.222 (95% CI 0.821–1.818)].

      Conclusions:
      On indirect comparison, maintenance treatment with Pemetrexed and Erlotinib has similar PFS and OS.

      Clinical trial identification:


      Legal entity responsible for the study:
      Tata Memorial Hospital Centre, Mumbai, India

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      152P - Impaired liver function was associated with PFS of EGFR TKI treatment (Now Available) (ID 247)

      12:50 - 12:50  |  Presenting Author(s): Q. Zhu  |  Author(s): Y. Chen, Y. Hong, Z. Ding

      • Abstract
      • Slides

      Background:
      EGFR TKI treatment revolutionized the standard of care for advanced NSCLC harboring EGFR mutation, but the surrogate markers for the efficacy need to be refined. Elevated transaminase was a dose-limiting factor for TKI treatment, while its prognostic significance was not explored before.

      Methods:
      This was a retrospective study where pathologically confirmed NSCLC patients prescribed with first-line EGFR TKI (gefitinib, erlotinib, or icotinib) were enrolled. But those with concomitant other cancer, or target lesion resected were excluded. In addition, patients who took drugs significantly affecting liver function were excluded. The clinical data were retrieved through a pre-established database. The highest transaminase level was recorded during the treatment course. The PFS was defined as the interval between the initiation of the treatment and the date when first sign of tumor progression was significant.

      Results:
      From Oct 2013 to Oct 2016, 208 patients were enrolled. Most of them were non-smokers (70.7%, vs smokers), had adenocarcinoma (93.3%, vs non-adeno), and took gefitinib (48.6%, vs 36.5% icotinib, vs 13.9% erlotinib). The median age was 59.5 year (range: 31–85). Female (55.8%, vs male) and exon 19 Del (54.8%, vs 38.7% L858R, vs 6.5% others) were distributed dominantly. Gefitinib was associated with the most frequent occurrence of elevated transaminase (42.5%, icotinib 26.3%, and erlotinib 32.2%), esp. ALT (40.6%, 19.7%, and 25.8% respectively). Interestingly, an inverse relationship was found between PFS and the elevated ALT. Those with elevated ALT tended to have shorter PFS (9.5 m) than those with normal ALT (12.6 m, p = 0.011, HR = 0.68). However, this correlation was not for AST (10.0 and 11.5 m respectively, p = 0.237, HR = 0.75). In Cox multivariate regression model, elevated ALT indicated shorter PFS, independent of gender, age (<60), mutation (exon19 Del or other), and smoking status.

      Conclusions:
      Elevated ALT level but not AST level was inversely related to the PFS of TKI treatment.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      153P - EGFR T790M co-exists with sensitive mutations in the same cell group in lung adenocarcinoma patients (Now Available) (ID 157)

      12:50 - 12:50  |  Presenting Author(s): X. Shi  |  Author(s): S. Wu, H. Wang, Z. Liang, X. Zeng

      • Abstract
      • Slides

      Background:
      EGFR TKI targeted therapy has improved lung adenocarcinoma patients’ prognosis tremendously, but almost all of patients inevitably develop acquired resistance to these agents, and EGFR T790M mutation is the major contributors. Previous work has shown that after TKI therapy, lung adenocarcinoma patients kept the sensitive mutation and acquired resistance mutation simultaneously by sequencing methods or in vitro cell line experiments. Whether the two different type mutations are in the same cell group or in two different cell groups is unknown.

      Methods:
      RNA in situ hybridization methods was employed to examined EGFR T790M and L858R mutation in lung adenocarcinoma cancer tissues which was obtained before and after TKI therapy. EGFR expression was examined by immunohistochemistry. EGFR mutation were detected by ARMS PCR methods.

      Results:
      Twenty-four patients were enrolled in this study which were divided into 3 groups. Group 1: 4 patients who had concurrent primary T790M and sensitive EGFR mutation. Group 2: 14 patients who acquired T790M mutation after receiving TKI therapy. Among them, 6 patients had biopsy tissues before and after TKI therapy. 8 patients only own tissues after TKI therapy. Group 3: 6 patients who had sensitive EGFR mutation and received TKI therapy, but re-biopsy tissues didn't have EGFR T790M. We found that the results of RNA ISH and ARMS PCR methods was identical in the majority of the examined tissues. Only one repeated biopsy tissue didn't identify EGFR T790M after TKI therapy by PCR in group 3, while the RNA ISH method detected T790M in this tissue which contain only 100 tumor cells. In the serial cut slides, we observed that T790M and L858R mutations were in the same cell group, not only in the primary resistance cases, but also in the acquired resistance cases. For the two cases which had tissues available after receiving third generation TKI therapy, we observed that T790M disappeared in the repeated biopsy specimen, leaving the sensitive mutation which existed from the beginning.

      Conclusions:
      EGFR sensitive mutation and T790M co-exist in the same cell groups. EGFR sensitive mutation is a trunk and drive mutation, while T790M is a passenger mutation during the treatment process by TKI therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Peking Union Medical College Hospital

      Funding:
      Peking Union Medical College Youth Fund, the Fundamental Research Funds for the Central Universities (Project NO. 3332016002). Pathology Research Centre of the China Academy of Medical Sciences (Project No. 2016ZX310176-2)

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      154P - CNS response to osimertinib in patients with EGFR mutated lung adenocarcinoma: Real world data (ID 524)

      12:50 - 12:50  |  Presenting Author(s): R. Devjak  |  Author(s): N. Turnsek Hitij, K. Mohorcic, M. Rajer, T. Cufer

      • Abstract

      Background:
      Driver mutation of epidermal frowth factor (EGFR) is presented in non-small cell lung cancer (NSCLC) at about 10%. Osimertinib (Tagrisso®) is a 3rd generation tyrosine kynase inhibitor (TKI) for EGFR mutated advanced lung adenocarcinoma with or without T790M resistant mutation. Particularly osimertinib showed a good penetration through blood-brain barrier and efficacy in central nervous system (CNS) metastases. In this, single-center, retrospective study we analyzed course of the CNS disease of metastatic NSCLC patients treated with osimertinib in any line of therapy.

      Methods:
      Patients with EGFRm advanced NSCLC who received osimertinib (80 mg daily) after progression on prior EGFR TKI (n = 23) or upfront (n = 2) were analyzed. All patients performed CT or MRI before osimertinib. We collected data on: presence of CNS metastases, line of the treatment on osimertinib, previous treatment of CNS metastases, efficacy and date of progression. Responses were evaluated radiologically every 8–16 weeks according to RECIST criteria. All patients had at least one evaluation.

      Results:
      In the present study 25 patients were included which have been treated from October 2015 untill November 2017. Among analyzed patients 10/25 (40%) had CNS (brain only) metastases before initiation with osimertinib, 4/10 received prior CNS treatment with radiotheraphy or surgery. Maximum osimertinib treatment duration was 46 /56 weeks for patients with/without CNS metastases, respectively. Median duration of treatment at the time of analysis was 27 weeks with 11/25 of patients still ongoing treatment. CNS objective response rate was 70% (CR 0%, PR 50%, SD 20%). Patients with progressive disaese (30%) had progression of all tumor leasons including CNS. There was no isolated CNS progression during osimertinib treament observed among all analyzed patients (n = 25).

      Conclusions:
      In our real world analysis osimertinib showed comparable CNS efficacy to clinical trial results with no CNS only progressive disease.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Clinic Golnik

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      155P - Antitumoral efficacy of dual blockade of EGFR signaling by osimertinib in combination with selumetinib or cetuximab in activated EGFR human NSCLC tumor models (ID 536)

      12:50 - 12:50  |  Presenting Author(s): F. Morgillo  |  Author(s): C.M. Della Corte, R. Alfieri, P. Petronini, S. La Monica, F. Papaccio, F. Ciardiello

      • Abstract

      Background:
      Osimertinib is the gold standard for activated-EGFR-NSCLC patient treatment. The aim of this work was to test the efficacy of a complete EGFR-inhibition by the addition of the monoclonal antibody, cetuximab, or the MEK1/2-inhibitor, selumetinib, to osimertinibin EGFR-mutated-NSCLC in vivo models.

      Methods:
      We evaluated combinations of osimertinib plus selumetinib/cetuximab in HCC827(E746-A759del/T790M-), H1975(L858R/T790M+) and PC9-T790M (E746-A759del /T790M+) xenografts, in second line after the developing of resistance to osimertinib and in first line, and we explored mechanisms of resistance to these treatments.

      Results:
      The addition of selumetinib or cetuximab to osimertinib in second line reverted the sensibility to osimertinib in the majority of mice, with a response rate (RR) of 50–80%, and a median PFS (mPFS) of first plus second line of therapy of 28 weeks. The early use of combinations in first line increased the RR to 90%, with amPFSnot reached in all combination arms in the three xenografts models, with a statistically significant superiority (p < 0,005) as compared to osimertinib, that achieved in first line a mPFS of 17–18 weeks. Moreover, on ex vivo primary cell cultures obtained from osimertinib plus selumetinib-resistant tumors, we found hedgehog pathway activation and we demonstrated that triple combination with a Smo inhibitor plus osimertinib.

      Conclusions:
      We demonstrated that a dual vertical EGFR blockade with osimertinib plus selumetinib/cetuximab is a novel effective therapeutic option in EGFR-mutated-NSCLC and that Hedgehog pathway activation and its interplay with MAPK is involved in resistance to these combination treatments.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Campania “L. Vanvitelli”

      Funding:
      AstraZeneca partially supported

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      156P - ASTRIS, a real-world study with osimertinib in patients with non-small cell lung cancer (NSCLC) EGFR T790M mutated: Characteristics and diagnostic methods used for patients included in Spain (ID 547)

      12:50 - 12:50  |  Presenting Author(s): D. Vicente Baz  |  Author(s): A. Paredes Lario, M.T. Moran Bueno, B. Massuti Sureda, N. Reguard, R. Álvarez, A. Insa Molla, O. Juan, G. Marquez, M. Provencio Pulla

      • Abstract

      Background:
      We present demographic and diagnostic data for the first planned interim analysis of ASTRIS study, currently ongoing.

      Methods:
      ASTRIS is a single-arm, open-label, phase IIIb clinical trial to evaluate the efficacy and safety of osimertinib monotherapy in real practice. Eligible patients had stage IIIB-IV NSCLC with a T790M mutation determined by a locally validated test (not restricted by sample type), had received at least a previous EGFR-TKI, ECOG 0-2, with no history of interstitial lung disease or QTc prolongation. Asymptomatic and stable CNS metastases were allowed. Patients received osimertinib 80 mg once daily.

      Results:
      We included 132 patients in 18 centers, 130 began treatment. At data cut-off (3 Nov 2016), 72% continued in the study, median follow-up 5.2 (<1–12) months. Median age 66 (32–89) years, 69% women, 98% caucasian, 85% ECOG 0/1, 15% ECOG 2, 84% stage IV, 40% cerebral / leptomeningeal metastasis, 42% previous chemotherapy, 34% previous radiotherapy. EGFR-TKIs: gefinitib (43%), erlotinib (57%), afatinib (17%) and dacomitinib (2%). Only patients with a T790M positive test result were treated in the study: 73 (56%) were recruited to the study after a positive tissue test, 47 (36%) after a positive plasma test, 4 (3%) cytology and 6 (5%) after testing another specimen type. The origin of the biopsy tissue was primary tumor (60%), metastasis (40%). The local laboratory was used in 62% of the patients. Testing methods: Roche cobas (50%), Qiagen therascreen (17%), PCR-Invader (20%), TaqMan (9%), ARMS-PCR (1%), Illumina MiSeq / HiSeq (1%), AMOY (1%) and others (2%). Other EGFR mutations were EXON 19 deletions (58%), L858R (27%), S768I (5%) and G719X (3%).

      Conclusions:
      The patient profile included in the study expands the patient population studied in the published studies, including excluded patients (PS-2, treated with non-marketed TKIs) or characteristics typical of the Spanish population, with a majority of Caucasians. The data from the ASTRIS study will give external validity to the results obtained in studies published with osimertinib.

      Clinical trial identification:
      NCT02474355

      Legal entity responsible for the study:
      AstraZeneca

      Funding:
      AstraZeneca

      Disclosure:
      D. Vicente Baz: Consultant or Advisory Role: Astra Zeneca, BMS, Pfizer, Roche Research Funding: Pfizer, Boehringer. G. Marquez: AstraZeneca employee. All other authors have declared no conflicts of interest.

    • +

      157P - Second-line afatinib for patients with locally advanced or metastatic NSCLC harbouring common EGFR mutations: A phase IV study (Now Available) (ID 198)

      12:50 - 12:50  |  Presenting Author(s): S. Thongprasert  |  Author(s): S. Geater, D. Clement, A. Abdelaziz, J. Reyer-Igama, D. Jovanovic, S. Suresh, A. Cseh, R. Gaafar

      • Abstract
      • Slides

      Background:
      The oral, irreversible ErbB family blocker, afatinib, is approved as first-line treatment for patients with EGFR mutation-positive (EGFRm+) NSCLC, demonstrating superior progression-free survival (PFS) and tolerability versus platinum-based chemotherapy. However, chemotherapy is still commonly used as first-line therapy in this patient population. In the LUX-Lung 2 study, second-line afatinib demonstrated clinical activity and an acceptable safety profile in patients with advanced NSCLC harbouring common EGFR (Del19/L858R) mutations following chemotherapy; however, the starting dose was 50 mg/day for most patients. Here, we report efficacy and safety of second-line afatinib at the approved dose of 40 mg/day in this patient population.

      Methods:
      In this open-label, single-arm Phase IV study, tyrosine kinase inhibitor-naïve patients with EGFRm+ (Del19/L858R) NSCLC who progressed after failure of first-line chemotherapy received afatinib (starting dose 40 mg/day) until disease progression or other reasons necessitating withdrawal. The primary endpoint was investigator-assessed objective response (OR); secondary endpoints were PFS and disease control. Safety was also assessed.

      Results:
      The study was conducted across 24 sites in 7 countries. Sixty patients were enrolled into the study and treated with afatinib for a median duration of 11.5 months. The mean age of patients was 55.9 years; 55% were female. An OR was achieved by 50% of treated patients; median duration of response was 13.8 months. 50 patients achieved disease control; median duration was 11.9 months. 39 patients experienced an event contributing to PFS, with median PFS of 10.9 months. The most common treatment-related adverse events were diarrhoea (72%), rash/acne (58%), and paronychia (27%); no deaths were related to afatinib treatment.

      Conclusions:
      At the approved starting dose of 40 mg/day, second-line afatinib demonstrated efficacy and a tolerable safety profile in patients with EGFRm+ (Del19/L858R) NSCLC. The outcomes are consistent with previous studies of afatinib. These findings support the use of second-line 40 mg/day afatinib in chemotherapy pre-treated patients.

      Clinical trial identification:
      NCT02208843

      Legal entity responsible for the study:
      Boehringer Ingelheim

      Funding:
      Boehringer Ingelheim

      Disclosure:
      S. Geater: Honoraria from AstraZeneca and Boehringer Ingelhiem, Research funding from Samsung, AstraZeneca, Boehringer Ingelhiem and Roche. D. Jovanovic: Membership on an advisory board or board of directors: Member of local and regional (Adriatic) BI AdB. R. Gaafar: Membership of advisory board: Boehringer Ingelheim, EliLilly, Astra Zeneca, MSD, Corporate sponsored research between company and NCI, Cairo University and investigator Roche International, BI, MsD. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      158P - Afatinib in patients with EGFR mutation-positive (EGFRm+) NSCLC harbouring uncommon mutations: Overview of clinical data (Now Available) (ID 567)

      12:50 - 12:50  |  Presenting Author(s): A. Märten  |  Author(s): N. Yamamoto, C. Yu, S.I. Ou, C. Zhou, Y. Wu

      • Abstract
      • Slides

      Background:
      Approximately 10–12% of patients with EGFRm+ NSCLC have tumours harbouring uncommon EGFR mutations; however, there is a paucity of clinical data on the sensitivity of these tumours to EGFR TKIs. Preclinical data indicate that the irreversible ErbB family blocker, afatinib, has similar activity against certain uncommon mutations (e.g. L858M/S768I) to that against the common L858R mutation.[1,2]

      Methods:
      Here, we review the key clinical data available for first-line afatinib in EGFRm+ NSCLC harbouring uncommon EGFR mutations.

      Results:
      Post-hoc analysis of the randomised LUX-Lung 2/3/6 trials[3] demonstrates that afatinib is clinically active against certain uncommon EGFR mutations. Among 75 afatinib-treated patients with tumours harbouring uncommon mutations in these trials the objective response rate against G719X (n = 18), L861Q (n = 16) and S768I (n = 8) single/compound mutations was 78%, 56% and 100%. Response rate was lower in patients with exon 20 insertions (n = 23; 9%) or de novo T790M (n = 14; 14%). In 38 patients with uncommon mutations/duplications in exons 18–21 PFS was 10.7 months (95% CI 5.6–14.7) and OS was 19.4 months (95% CI 16.4–26.9). The clinical activity of afatinib against uncommon mutations is substantiated by observations outside of the randomised controlled trial setting. In a Phase IIIb single-arm open-label study of afatinib in its registered indication, 67 of 479 TKI-naïve patients had NSCLC with uncommon mutations[4] (exon 20 insertions; L861Q; G719S/A/C; T790M; S768I: 5.2; 4.6; 2.3; 1.0; 1.9%, respectively). Median PFS in these patients was 9.1 months (95% CI 5.6–13.6).[4] In an analysis of 165 EGFRm+ NSCLC patients treated with first-line afatinib in real-world practice in South Korea median PFS in those with uncommon mutations excluding T790M (n = 10) was not reached vs 4.7 months in those with T790M (n = 4; p = 0.01).[5]

      Conclusions:
      Afatinib has shown preclinical and clinical activity in patients with NSCLC harbouring uncommon EGFR mutations, including G719X, L861Q and S768I. These data could help inform clinical decisions in this patient population. 1. Saxon. J Thorac Oncol 2017;12:884 2. Banno. Cancer Sci 2016;107:1134 3. Yang. Lancet Oncol 2015;16:830 4. Wu. WCLC 2017 P3.01-036 5. Kim. WCLC 2017 P3.01-023

      Clinical trial identification:
      N/A

      Legal entity responsible for the study:
      Boehringer Ingelheim

      Funding:
      Boehringer Ingelheim

      Disclosure:
      A. Märten: Employment: Boehringer Ingelheim. N. Yamamoto: Membership on an advisory board or board of directors: BI, AZ, Chugai Corporate-sponsored research: BI. C-J. Yu: Membership on an advisory board or board of directors: Roche, ONO, Chugai, Novartis. Y-L. Wu: Honoraria: AstraZeneca, Roche, Eli Lilly, Pfizer, Sanofi. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      159P - Clinical activity of afatinib in a cohort of patients with lung adenocarcinoma harbouring uncommon EGFR mutations: A Spanish retrospective multicentre study (ID 269)

      12:50 - 12:50  |  Presenting Author(s): M.T. Moran Bueno  |  Author(s): C. Aguado, M. Dómine, A. Gomez Rueda, A. Calles, S.M. Cedres, N. Viñolas, D. Isla, R. Palmero, M. Sereno Moyano

      • Abstract

      Background:
      Uncommon EGFR mutations (u-EGFRm) in exons 18–21 account for 12–15% of overall EGFR mutations in non-small cell lung cancer (NSCLC). Afatinib has shown activity against some u-EGFRm, especially G719X, L861Q, and S768I; although other u-EGFRm (Ins20, de novo T790M) have a lower likelihood of response. U-EGFRm are a heterogeneous group of molecular alterations and have also been reported as co-mutations with other EGFR mutations (complex mutations). We examined the molecular and clinical characteristics and efficacy of afatinib in a cohort of patients with advanced NSCLC patients carrying u-EGFRm in Spanish clinical practice.

      Methods:
      Medical records of 67 NSCLC patients with u-EGFRm treated with afatinib between 2012 and 2017 at 23 Spanish institutions were reviewed. U-EGFRm were analysed as complex mutations (Group A), Ins20 (Group B), or single mutations (Group C). Efficacy data were evaluated in terms of tumour response and overall survival (OS).

      Results:
      Of the 67 patients, 96% were Caucasian, all were adenocarcinoma, 46% were never smokers and 37% were former smokers, 79% were Stage IV at diagnosis, 79% had ECOG PS 0–1. Group A complex u-EGFRm consisted of double mutations of G719X + E709F, G719X + S768I, G719X + L861Q, L858R + T790M, L858R + S768I, Del19 + S768I, Del19 + L747S, or R776C + L861Q. No differences in clinical characteristics were found between Group A (n = 20), Group B (n = 23), and Group C (n = 24). Afatinib was administered as 1st line therapy in 80% of patients. Median time on therapy was 4.2 months (range 2.0–12.9). Eighteen percent of patients started afatinib at a reduced dose and 24% of patients required a dose reduction. Response to afatinib was significantly higher in Group A and C (70% and 54%, respectively), compared with Group B (13%; pairwise comparison p < 0.001 and 0.013, respectively, Table). Median OS (mOS) for the entire cohort was 19.9 months (9.7–30.1). Hazard ratio for OS were 0.27 (95% CI 0.10-0-71; p = 0.009) and 0.40 (95% CI 0.17–0.95; p = 0.037) for Group A and C compared to Group B, respectively.

      u-EGFRmGroup A (complex)n = 20Group B (Ins20)n = 23Group C (other)n = 24
      Response to afatinib, n (%)
      Complete response1 (5.0)0 (0)1 (4.2)
      Partial response13 (65.0)3 (13.0)12 (50.0)
      Stable disease3 (15.0)8 (34.8)5 (20.8)
      Progressive disease0 (0)10 (43.5)3 (12.5)
      Not evaluable3 (15.0)2 (8.7)3 (12.5)
      mOS (95% CI), months28.8 (20.7–36.8)10.7 (6.2–15.3)19.9 (10.8–29.0)
      p = 0.014


      Conclusions:
      In clinical practice, afatinib was active in u-EGFRm NSCLC, particularly in complex and single mutations. Further strategies are needed for patients with ins20, a subgroup u-EGFRm with a lower clinical benefit from afatinib.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hospital Germans Trias i Pujol, Badalona, Barcelona, Spain

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      160P - Real world study of lung squamous cell carcinoma patients with EGFR mutation treated with EGFR-TKI (Now Available) (ID 527)

      12:50 - 12:50  |  Presenting Author(s): L. Miao  |  Author(s): G. Lin, Y. Wu, J. Chen, X. Ai, M. Yuan, R. Chen

      • Abstract
      • Slides

      Background:
      EGFR tyrosine kinase inhibitors (TKIs) have greatly improved the outcomes of EGFR mutation-positive adenocarcinomas of the lung; however, the role of genetic testing and efficacy of EGFR-TKI in lung squamous cell carcinoma (SCC) remains controversial.

      Methods:
      A total of 265 patients with advanced SCC who underwent genetic testing at our institute from 2016 to 2017 were included for analysis. Mutation profiling was performed with targeted capture based next-generation sequencing, which enables simultaneously assess single-nucleotide variants, insertions/deletions, rearrangements, and somatic copy-number alterations across 59 genes. Response assessment was done using Response Evaluation Criteria In Solid Tumors (RECIST) 1.1. Kaplan–Meier method was used for calculating time to progression (TTP).

      Results:
      EGFR mutation was detected in 28 out of 265 patients with SCC (10.6%). Of the patients, 12 was with EGFR exon 21 L858R mutation, 10 was exon 19 E746_A750del mutation and 2 was exon 21 L861Q mutation. In addition, 8 was exon 20 mutations with 2 T790M mutation, 2 S768I mutation and 1 P772_H773insR, S768_D770dup, V774M, N771dup mutation resprectively. For the 2 patients with de novo T790M mutation, one has concurrent L858R mutation and the other EGFR N771dup. Furthermore, 10 out of the 27 patients received TKIs during their treatment. Five patients were treated with chemotherapy as first line, and five were EGFR-TKI as the first line. The median TTP on EGFR TKI was 10.5 months. There was no significant difference of TTP between the two groups treated with EGFR-TKI as first line or not (16 months vs 7 months, p = 0.19).

      Conclusions:
      EGFR mutations are present in over 10% SCC patients. These patients predict a better outcome if given TKI, but it may be inferior to the outcomes of EGFR-positive adenocarcinomas treated with TKI.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      161P - The survival of advanced non-small cell lung cancer with epidermal growth factor receptor in Thailand: A single instituition review (Now Available) (ID 183)

      12:50 - 12:50  |  Presenting Author(s): A. Supavavej  |  Author(s): M. Tandhansakul

      • Abstract
      • Slides

      Background:
      EGFR TKIs can improve progression free survival in treatment naive EGFR mutant NSCLC compared with platinum based chemotherapy, but no statistical significant difference in overall survival. The median survival time of EGFR mutant NSCLC treated at least 2 systemic chemotherapy regimens and one EGFR TKIs was 34.8 months in previous study.

      Methods:
      We retrospectively reviewed the medical record of advanced NSCLC cases treated with EGFR TKIs in Chulabhorn Hospital during 2009–2012. The primary objective was to determine the median survival time of advanced NSCLC patients who received EGFR-TKIs in Chulabhorn Hospital. The secondary objective was to identify the difference of survival between EGFR mutation status groups. The overall survival was analyzed by Kaplan Meier curve. The overall survival in EGFR mutation status was compared by Log rank test.

      Results:
      There were 73 patients with median follow up time of 17.09 months. EGFR TKIs were given as first, second, and third line treatment in 18 (24.7%), 29 (39.7%), and 22 (30.1%) patients, respectively. EGFR mutation status was positive, negative and unknown in 14 (19.17%), 15 (20.8%), and 43 (59.7%) patients. Exon 19 deletion mutation was presented in 8 (57.1%) patients while there was 5 (35.7%) patients with exon 21 L858R substitution. There was one patient with exon 19 G729E substituition. The median overall survival time was 30.01 months (95%CI: 18.71, 49.48 months) in all patients, 49.49 months (95%CI: 15.93, NR) in EGFR mutant (N= 14), 20.96 months (95%CI: 10.64, NR) in EGFR wild type (N= 15), and 27.74 months (95%CI: 18.71, NR) in unknown EGFR mutation status (N = 43). No statistical significant difference of overall survival was noted between EGFR mutation status (p values = 0.51).

      EGFR mutationNmedian OS(months)95%CI
      positive1449.4915.93,NR
      negative1520.9610.64,NR
      unknown1727.7418.01,NR
      total7230.0218.71,49.49


      Conclusions:
      The median survival time for Thai patients treated with EGFR TKIs was comparable with historical data irrespective to EGFR mutation status. In EGFR mutant cases, the median survival time was nearly 4 years. EGFR TKIs in any lines of treatment can prolong the survival of NSCLC patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chulabhorn Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      162P - Responses to EGFR TKIs and ALK TKIs in advanced NSCLC patients harboring concomitant EGFR and ALK alterations (Now Available) (ID 503)

      12:50 - 12:50  |  Presenting Author(s): S. Wang  |  Author(s): T. Chu, B. Zhang, B. Han, B. Yan, R. Qiao

      • Abstract
      • Slides

      Background:
      Previous studies indicated that Epidermal growth factor receptor (EGFR) mutation and anaplastic lymphoma kinase (ALK) rearrangement are mutually exclusive in non-small cell lung cancer (NSCLC). However, cases diagnosed with concomitant EGFR and ALK alterations has been occasionally reported. This study aimed to assess the prevalence of this small subset patients and optimize clinical management.

      Methods:
      We retrospectively collected clinical outcomes of 29 cases who had concomitant EGFR and ALK alterations from 5816 lung cancer patients tested EGFR mutation and ALK rearrangement between 2011–2017 in the Shanghai Chest Hospital. Meanwhile, we identified 103 cases harboring double positive mutations from a literature search. Of these 132 cases, 81 patients received EGFR tyrosine kinase inhibitor (EGFR-TKI) or ALK-TKI treatment.

      Results:
      The frequency of EGFR/ALK co-alterations was 0.5% (29/5816; 95%CI:0.3%-0.7%) in NSCLC in our center. For all 132 cases, there is a prevalence of women (67 female, 46 male, 19 not reported), Asian (87Asian, 44 Caucasian, 1 not reported) and never smoker patients (77 never smokers, 21 smokers, 34 not reported). We divided the patients into three groups according to EGFR or ALK TKIs treatment: A: single EGFR TKI group (36 cases), B: single ALK TKI group (14 cases) and C: both TKIs (31 cases). All patients were assessed for TKIs responses. The disease control rate (DCR) of EGFR-TKI was 81.5%, whereas the DCR of ALK-TKI was 89.1%. The median PFS of three groups were 12.4, 15.9 and 24.1months, respectively (P = 0.02). The PFS of group A and C had statistically sigificant difference (P = 0.006). But the PFS of group A and B, B and C did not have statistical significance (P = 0.338, P = 0.335).

      Conclusions:
      EGFR mutations and ALK rearrangement do coexist in NSCLC. In cases with double positive mutations, our preliminary study suggests that PFS of those who received double TKIs is longer than those who received only one TKI. Combination of both TKIs might be an appropriate choice. The result of the study indicates that ALK TKI might be preferentially administered when TKI is initiated as first-line treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Wang shuyuan

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      163P - The influence of 2<sup>nd</sup> and 3<sup>rd</sup> generation TKI in EGFR mt+ and ALK+ patients on OS and PFS: Results of the NOWEL network (Now Available) (ID 402)

      12:50 - 12:50  |  Presenting Author(s): J. Roeper  |  Author(s): M. Netchaeva, A. Lueers, M. Falk, M. Tiemann, C. Wesseler, G. Wiest, S. Sackmann, D. Ukena, F. Griesinger

      • Abstract
      • Slides

      Background:
      EGFR TKI treatment is standard of care in patients with metastasized NSCLC carrying an activating EGFR mutation. Targeted therapies achieve a higher ORR, OS, PFS and a better quality of life than chemotherapy in mt+ patients. With the advent of 2[nd] and 3[rd] generation TKÍs effective in 1[st] generation TKI resistant tumors, we wanted to study the impact of these drugs on the outcome of patients in a real life setting in 3 lung cancer centers.

      Methods:
      1477 patients from three cancer centers diagnosed with non-squamous cell NSCLC stage IV (UICC 7) were examined. Methods for the mutation testing was performed according to the German Oncopedia guidelines using either Sanger Sequencing or COBAS® or Next Generation Sequencing (hybrid capture NGS, New Oncology Cologne).

      Results:
      945/1477 (64%) consecutive patients with non-squamous cell NSCLC from three cancer centers were studied for the presence of tumor mutations, especially for EGFR and ALK mutations. The EGFR mutation rate was 16% (149/912), and the ALK-translocation rate 4% (26/700). Median OS in EGFR mt+ patients was 23 months (n = 154) compared to 11 months (n = 763) in patients with EGFR WT (p < 0.001). Median OS in EGFR mt+ patients depending on the center was 23 (n = 102) vs. 28 (n = 38) vs. 16 (n = 14) months respectively (center 1 vs. center 2 vs. center 3). Median OS in ALK mt+ patients was 24 months (n = 19) in center 1, 11 months (n = 5) in center 2 and in center 3 median OS was not reached (p < 0.025). The use of 3[rd] generation TKI Osimertinib lead to a significantly higher OS (n = 21, median OS 55 months) than the use of only 1[st] and 2[nd] generation TKI (n = 118, median OS 22 months, p < 0.001). The hazard ratio HR for patients treated without Osimertinib was 2.77 [95% CI 1.454–5.305] (p < 0.002). Similarly, use of 2[nd] and 3[rd] generation ALKi impacted significantly on median OS: Crizotinib alone (n = 7), 17 months, Crizotinib followed by Ceritinib and/or Brigatinib/Alectinib (n = 13) median OS was not reached and 3 months for other therapies (n = 6) (p < 0.000).

      Conclusions:
      Small differences in OS were observed, depending on the treatment centers, but the use of multiple EGFR and ALK-I impacted highly significantly on the outcome of patients with EGFR and ALK-alterations in a real life setting.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Oldenburg

      Funding:
      Has not received any funding

      Disclosure:
      M. Netchaeva: Advisory Boards: Roche, AstraZeneca, BMS Travel: Boehringer-Ingelheim, Celegene, Pharma Mar, Amgen Consultant: Boehringer-Ingelheim. M. Falk: Advisory boards: Boehringer Ingelheim, Pfizer, Roche. M. Tiemann: Advisory boards: Novartis, Boehringer, Roche, Astra Zeneca Scientific Support: Novartis F. Griesinger: Advisory Boards: Ariad, Astra-Zeneca, Boehringer-Ingelheim, Bristol-Myer-Squibb, Celgene, Clovis, Lilly, Merck-Sharp-Dome, Novartis, Pfizer, Roche Travel Support: Ariad, Astra-Zeneca, Boehringer-Ingelheim, Bristol-Myer-Squibb, Celgene, Lilly, Merck-Sharp-Dome, Novartis, Pfizer, Roche Scientific Support: Astra-Zeneca, Boehringer-Ingelheim, Bristol-Myer-Squibb, Celgene, Lilly, Merck-Sharp-Dome, Novartis, Pfizer, Roche Shares: none. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      164P - Feasibility of liquid biopsy using plasma and platelets for detection of ALK rearrangements in non-small cell lung cancer (Now Available) (ID 297)

      12:50 - 12:50  |  Presenting Author(s): C. Park  |  Author(s): H. Park, H. Cho, Y. Choi, I. Oh, Y. Kim

      • Abstract
      • Slides

      Background:
      The aim of this study is to investigate the feasibility of liquid biopsy using plasma and platelets for detection of ALK rearrangement.

      Methods:
      FISH was performed in 664 patients between January 2015 and May 2017. We retrospectively analyzed the FFPE tissue and blood sample to detect ALK rearrangement using multiplex RT-PCR from 30 advanced NSCLC patients who had available tissue specimen. Total RNA were extracted from FFPE cell blocks, plasma and platelets, respectively. EML4-ALK fusion RNA was detected using PANAqPCR[TM] EML4-ALK fusion gene detection kit.

      Results:
      28 patients were FISH positive and 2 were negative. In a validation data compared with FISH, RT-PCR using FFPE tissue demonstrated 57.1% sensitivity and 69.2% accuracy. Liquid biopsy (plasma or platelets-positive) had higher sensitivity (96.4%) and accuracy (93.3%). Among the specimen of liquid biopsy, platelets showed slightly higher sensitivity and accuracy than plasma (82.1 and 83.3% vs 78.6 and 76.7%). Compared with FFPE tissue using RT-PCR, liquid biopsy showed 100% sensitivity, 20.0% specificity and 69.2% accuracy. Median proportion of positive cells in FISH was higher in subgroups of liquid biopsy with positive result (Plasma, 30.0 vs 15.0%, p = 0.062; Platelets 30.0 vs 20.0%, p = 0.104). In 18 patients with crizotinib treatment, platelets-positive subgroup showed a tendency of longer duration of treatment (7.2 vs 1.5 months, p = 0.071) and higher response rate (57.1 vs 0.0%, p = 0.092). However, platelets-positive subgroup showed significantly higher disease control rate than platelets-negative subgroup when they were treated with crizotinib (85.7 vs 25.0%, p = 0.044).

      Conclusions:
      Plasma and platelets are valuable sources for liquid biopsy using RT-PCR technique in detection of ALK rearrangement, and could play a supplementary role in diagnosis of ALK-positive NSCLC. Furthermore, platelets may be useful for predicting the treatment outcome of crizotinib.

      Clinical trial identification:


      Legal entity responsible for the study:
      Legal entity responsible for the study

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      165P - Analysis of our experience of ROS-1 patients in advanced NSCLC (Now Available) (ID 518)

      12:50 - 12:50  |  Presenting Author(s): N. Pande  |  Author(s): A. Joshi, V. Noronha, V. Patil, A. Mahajan, K. Prabhash

      • Abstract
      • Slides

      Background:
      ROS1 is a receptor tyrosine kinase receptor and it belongs to insulin receptor family.[1] It acts as a driver oncogene in 1 to 2% of NSCLC patients. A homology exists between ALK and ROS1 Kinase domains. Crizotinib binds with high affinity to both ALK and ROS1, which is consistent with this homology. Crizotinib is approved for patients with the ROS1 translocation including those who have received chemotherapy and those who are treatment naïve. We report our experience in a tertiary cancer care centre in ROS-1 positive patients.

      Methods:
      Patients who were ROS-1 positive by break-apart Fluorescence In situ Hybridization (FISH) and who had advanced NSCLC being planned treatment with palliative intent were included in our study. We had a total of 22 patients whose details were obtained from the prospective lung cancer audit database that is maintained in the department of medical oncology. Details of patients demographic data (age, gender, comorbidities, smoking status and performance status), tumour characteristics (histology, stage, number and sites of metastases) treatment (crizotinib dose, sequence of treatment, dose interruptions, treatment used before and after crizotinib,) efficacy and side effects were retrieved. Response evaluation done by RECIST 1.1 criteria.

      Results:
      A total of 22 patients satisfied the predefined selection criteria. There were 13 males and 9 females in this group and the median age of the population was 53 years. 10 patients received Platinum doublet as first line and 1 received Erlotinib as first line in view of poor PS. Six patients could be started on Crizotinib as first line. A total of 16 patients took Crizotinib either in first or second line. The patients who were on Crizotinib had good tolerance with none experiencing any grade 3/4 toxicity. The median follow up was 244 days in non-crizotinib arma and 322 days in Crizotinib arm. The estimated PFS was 79 days in non-crizotinib arm and 573 days in those who received Crizotinib. The estimated OS was 79 days vs not reached in Crizotinib arm.

      Conclusions:
      In conclusion, crizotinib is effective treatment with acceptable side effect profile in patients with ROS1 rearranged advanced NSCLC with significant improvement in PFS and OS.

      Clinical trial identification:


      Legal entity responsible for the study:
      Nikhil Pande

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      166P - Evidence-based conclusions and inclinations of pemetrexed, taxane and bevacizumab in advanced lung cancer (Now Available) (ID 331)

      12:50 - 12:50  |  Presenting Author(s): S. Shaheen  |  Author(s): L. Ji, J.W. Morgan, S. Otoukesh, H. Mirshahidi

      • Abstract
      • Slides

      Background:
      Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death. In advanced lung cancer patients, lacking targetable mutations, platinum-based chemotherapy is a mainstay treatment. Pemetrexed and taxane are used in combination with platinum, however, there are no large randomized trials comparing first-line of these agents. Furthermore, addition of bevacizumab is sometimes used in the treatment of these patients. The objectives of this study were to compare survival in patients treated with pemetrexed vs taxane in addition to platinum, with and without bevacizumab.

      Methods:
      We extracted data for metastatic lung adenocarcinomas diagnosed 2005–2015 from the California Cancer Registry (CCR) assessing lung cancer-specific Cox proportional mortality hazards ratios (HR) for patients that had received 1. platinum + taxane vs platinum + pemetrexed and 2. bevacizumab (Y/N).

      Results:
      Of 6,034 patients enrolled in the study, 3,593 (58.5%) received platinum and taxane, 2,788 (41.5%) received platinum and pemetrexed, 1,712 (28.4%), received bevacizumab. Median survival among patients treated with pemetrexed was significantly longer than those that received taxane (12.4 months vs 10.1 months; p < 0.001). Likewise, median survival time among patients treated with bevacizumab (Y/N) was 14.8 months vs 9.8 months; p < 0.001. Significantly improved survival was evident for younger patients <65 years (HR = 0.94; 95%CI = 0.88–0.99), females (HR = 0.78; 95%CI = 0.74–0.83), Asians (HR = 0.74; 95%CI = 0.68–0.80) and Hispanics (HR = 0.86; 0.78–0.95), the highest SES quintiles (HR = 0.87; 95%CI = 0.78–0.96), and for patients treated with platinum and pemetrexed (HR = 0.82; 95%CI = 0.77–0.87) and bevacizumab (HR = 0.78; 95%CI = 0.73–0.83). Our finding revealed increases in use of pemetrexed with reductions in taxanes, while bevacizumab use peaked in 2008, followed by decline.

      Conclusions:
      Our study showed survival was significantly improved in patients who received pemetrexed in addition to platinum as compared to taxanes. Although bevacizumab has lost favour, our findings revealed additional survival benefit when combined with either chemotherapy regimen.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      167P - Efficacy of pemetrexed-based chemotherapy in advanced lung adenocarcinoma patients with ROS-1 rearrangement (Now Available) (ID 413)

      12:50 - 12:50  |  Presenting Author(s): B. Zhang  |  Author(s): T. Chu, J. Xu, Y. Zhang, B. Yan, Y. Dong, J. Qian, R. Qiao, S. Wang, B. Han

      • Abstract
      • Slides

      Background:
      When chemotherapy is commenced as first-line treatment in advanced lung adenocarcinoma patients with ROS-1 rearrangement, it is unclear that which agent should be preferentially administered. The aim of this study is to compare the therapeutic efficacy of pemetrexed-containing (Pem-C) and non-pemetrexed-containing (Non-Pem-C) chemotherapy in these patients.

      Methods:
      We retrospectively identified patients who were demonstrated to be ROS-1 positive by multiplex reverse-transcriptase polymerase chain reaction (RT-PCR) between October 2014 and December 2016. Those who received platinum-based dual agent chemotherapy as palliative treatment were included for further analysis.

      Results:
      A total of 4596 consecutive individuals were screened and 55 eligible individuals were enrolled into this study. In first-line treatment, patients who received Pem-C treatment (n = 39) derived a higher objective response rate (ORR, 40.0% vs. 7.1%, P = 0.02) and progression-free survival (PFS1, 7.0 months vs. 3.9 months, P < 0.01) compared with those who received Non-Pem-C treatment (n = 16). However, in later-line treatment, progression-free survival (PFS2) was not statistically superior in the Pem-C group (3.1 months, 95% CI: 0.6–5.6 months) compared with the Non-Pem-C group (1.9 months, 95% CI: 0.1–3.1 months, P = 0.12).

      Conclusions:
      Pem-C treatment resulted in better clinical outcomes compared with other agents in patients with ROS-1 rearrangement when initiated as first-line treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Bo Zhang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      168P - Pemetrexed dosing regimens in patients with advanced NSCLC (Now Available) (ID 617)

      12:50 - 12:50  |  Presenting Author(s): S. Visser  |  Author(s): S. Koolen, P. de Bruijn, B. Stricker, R. Mathijssen, J. Aerts

      • Abstract
      • Slides

      Background:
      There is a lack of rationale to use body surface area (BSA)-based dosing if BSA is not a predictor of systemic clearance and thus total systemic exposure (AUC). Using population pharmacokinetic (Pop-PK) modelling, we evaluated the influence of BSA and renal function on pemetrexed (PMX) clearance and compared different dosing strategies.

      Methods:
      In a prospective observational multi-center study, patients with stage IIIB/IV non-small-cell lung cancer (NSCLC) receiving PMX(/platinum) first- or second-line were enrolled. PMX (500 mg/m[2]) was administered as a 10-min intravenous infusion every 21 days. Prior to and weekly after each PMX administration, plasma sampling was performed (cycle-PK) and glomerular filtration rate (eGFR) was estimated. In a subgroup, sampling was also performed at 10,30 minutes and 1,2,4 and 8 hours after start of PMX infusion (24h PK). With the ultimate Pop-PK model, we simulated different dosing strategies and compared differences in median AUC (interquartile range [IQR]) and interindividual variability (coefficient of variation [CV]) of AUC to standard BSA-based dosing.

      Results:
      For 106 of the 165 patients, concentrations of PMX were quantified (24h PK, n = 15; cycle PK, n = 106). A two-compartment model (population estimate (%standard error of the estimate) in terms of PMX clearance (CL; 4.58L/h (3.1%)), central distribution volume (V~1~; 15.9L (3.3%)), peripheral distribution volume (V~2~; 21.6L (5.0%)), intercompartimental clearance (Q; 0.05L/h (4.7%)) and between-patient variability of CL (16.7%), fitted PK data appropriately. Covariate eGFR significantly reduced between-patient variability in CL from 20.2% to 16.7% (p < 0.005), while BSA did not. Compared to BSA-based dosing (CV 22.5%, AUC 206 (IQR 178–240)), simulation of eGFR-based dosing (CV 18.5%, AUC 206 (IQR 183–232)) and fixed dose of 900 mg with 25% dose reduction if eGFR < 60 (CV 19.1%, AUC 197 (174–224)) both showed less interindividual variability of AUC.

      Conclusions:
      Although we currently dose PMX based on BSA, our data show better rationale for eGFR-based dosing as it offers additional control of systemic exposure to PMX, indicated by the decreased variability. Using fixed dose of 900 mg with 25% dose reduction if eGFR < 60 may be an acceptable alternative in clinical practice.

      Clinical trial identification:


      Legal entity responsible for the study:
      Erasmus MC

      Funding:
      ZonMw

      Disclosure:
      J. Aerts: Member of scientific committee ELCC 2018 consultant/advisory role with Eli Lilly and Company All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      169P - Nephrotoxicity of pemetrexed maintenance in patients with advanced NSCLC: Two cohort studies (Now Available) (ID 534)

      12:50 - 12:50  |  Presenting Author(s): S. Visser  |  Author(s): J. Huisbrink, N. Van 'T Veer, J. Van Toor, T. Van Boxem, N. Van Walree, B. Stricker, J. Aerts

      • Abstract
      • Slides

      Background:
      Optimal survival benefit from different lines of anticancer treatment in advanced non-small-cell lung cancer (NSCLC) requires conservation of renal function. We evaluated the development of renal impairment and its clinical significance during pemetrexed maintenance.

      Methods:
      In a prospective multi-center cohort study, patients with advanced (stage IIIB/IV) NSCLC treated with first-line (platinum-combined) or second-line pemetrexed were enrolled. After platinum-based induction patients were eligible for pemetrexed maintenance if no disease progression occurred. We evaluated the incidence of acute and chronic kidney disease (AKD/CKD), treatment discontinuation frequency due to renal impairment and associations of clinical variables with AKD during maintenance. Both AKD and CKD were defined in accordance with the Kidney Disease: Improving Global Outcomes (KDIGO) clinical practice guidelines. We validated findings in an independent cohort of advanced NSCLC patients treated with pemetrexed maintenance.

      Results:
      In total 213 patients received pemetrexed. In the primary cohort 165 patients started induction of whom 47 (28%) continued maintenance. During maintenance 16 patients (34%) developed AKD, leading to CKD in 11 (69%) and treatment discontinuation in 9 (56%) in the primary cohort. Lower eGFR (unit 5 mL/min/1.73 m[2]) before start of induction (OR 1.32, 95%CI: (1.06–1.64) and relative decline (per 10%) in eGFR during induction (OR 2.11, 95%CI: 1.22–3.65) were univariably associated with AKD during maintenance. In the independent cohort (n = 48), these similar associations could be demonstrated. In this cohort, 24 patients (50%) developed AKD, leading to CKD in 13 (54%) and treatment discontinuation in 7 (29%).

      Conclusions:
      Patients are at risk for renal impairment during pemetrexed maintenance, which may jeopardize further lines of anticancer treatment. Patients whose renal function is impaired before – or declines during – induction treatment are more vulnerable. Renal protective strategies for patients at increased risk might be beneficial, such as continuation of hydration during pemetrexed maintenance or dosing based on renal function.

      Clinical trial identification:


      Legal entity responsible for the study:
      Erasmus MC

      Funding:
      ZonMw

      Disclosure:
      J. Aerts: Member of Scientific Committee ELCC; Consultant/advisory role with Eli Lilly and Company. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      170P - A phase II trial of nab-paclitaxel and gemcitabine in patients with non-small cell lung cancer previously treated with platinum based chemotherapy (ID 346)

      12:50 - 12:50  |  Presenting Author(s): Y. Hatakeyama  |  Author(s): M. Tachihara, T. Kiriu, A. Hata, T. Nagano, M. Yamamoto, K. Kobayashi, H. Ohnishi, N. Katakami, Y. Nishimura

      • Abstract

      Background:
      Nab-Paclitaxel plus gemcitabine significantly improved overall survival, progression-free survival, and response rate in patients with metastatic pancreatic adenocarcinoma. Combination treatment increases intra-tumoral gemcitabine levels attributable to a decrease in the primary gemcitabine metabolizing enzyme, cytidine deaminase. Based on these data, we planned to assess the efficacy and safety of combination with nab-PTX + gemcitabine in patients with non-small-cell lung cancer (NSCLC) previously treated with platinum based chemotherapy.

      Methods:
      Patients with advanced NSCLC with progressive disease to platinum-based chemotherapy, ECOG performance status (PS) 0 or 1, and adequate kidney, liver and bone marrow function were eligible. Treatment consisted of nab-paclitaxel (100 mg/m[2]) + gemcitabine (1000 mg/m[2]) on days 1 and 8 of each 3-week cycle until progression disease or unacceptable toxicity. The primary endpoint was progression-free survival (PFS).

      Results:
      Of the 28 patients enrolled, 28 were evaluable for response and toxicity. The median age was 68 years (range 47–79), 23 male and 5 female. Histology subtypes were: adenocarcinoma 19 patients, squamous cell carcinoma 9 patients. Seventeen patients had ECOG PS 1 and 11 patients had PS 0. Twenty-four patients were 2[nd] line and 4 patients were 3[rd] line. The median number of cycles administered was 4(range 1–10). The overall response rate was 17.9%. The disease control rate was 67.9%. The median progression-free survival was 3.3 months (95% confidence interval [CI] = 1.6–4.7). Four patients (14.3%) had grade 3 anemia, 3 patients (10.7%) had grade 3 thrombocytopenia, 5 patients (17.9%) had grade 4 neutropenia. However, no patients developed febrile neutropenia. Remarkable non-hematologic toxicity was interstitial pneumonia with grade 3 in 4 patients (14.3%), neuropathy with grade 1 in 2 patients (7.1%) and infections with grade 3 in 2 patients (7.1%).

      Conclusions:
      The efficacy of nab-Paclitaxel in combination with gemcitabine in advanced second or third-line NSCLC patients was limited and the high onset of interstitial pneumonia was unacceptable.

      Clinical trial identification:


      Legal entity responsible for the study:
      Division of Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      171P - A phase I/II study of weekly nab-paclitaxel plus cisplatin in chemotherapy-naïve patients with advanced non-small cell lung cancer (ID 300)

      12:50 - 12:50  |  Presenting Author(s): Y. Kono  |  Author(s): Y. Hattori, K. Nishino, R. Uozumi, S. Itoh, T. Inoue, T. Kumagai, S. Morita, F. Imamura, M. Satouchi

      • Abstract

      Background:
      In the treatment of non-small-cell lung cancer (NSCLC), solvent-based paclitaxel (sb-paclitaxel) plus cisplatin can be expected to have high response rate, but strong neurotoxicity is observed. Currently, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) plus carboplatin is considered as one of the standard regimen, and a higher response rate and lower neurotoxicity are expected as compared with sb-paclitaxel plus carboplatin. Therefore, we planned this study to evaluate the efficacy and safety of nab-paclitaxel plus cisplatin in chemotherapy-naïve patients with advanced NSCLC.

      Methods:
      Chemotherapy-naïve patients with advanced NSCLC were eligible. In phase 1 dose escalation cohort (3 + 3 design), patients received nab-paclitaxel (80 or 100 mg/m[2] iv on days 1, 8 and 15) plus cisplatin (60 or 75 mg/m[2] iv on day 1) every 4 weeks. The maximum tolerated dose was not reached. Nab-paclitaxel (100 mg/m[2] iv on days 1, 8 and 15) plus cisplatin (75 mg/m[2] iv on day1) every 4 weeks was selected for phase 2 cohort. The primary endpoint was objective response rate (ORR).

      Results:
      23 patients (phase 1, n = 6; phase 2, n = 17) were enrolled from October 2013 to September 2017, and 22 patients were eligible. The median age was 67.5 years (range 37–75 years), 90.9% were males, 100% had smoked, 45.5% had adenocarcinoma and 81.8% had Stage IV. The ORR was 59.1% (95% confidence interval (CI); 38.7–76.7%) and the disease control rate was 86.4% (95% CI; 66.7–95.3%). The median progression free survival was 5.1 months (95% CI; 4.0–6.7 months) and the median overall survival was 24.2 months (95% CI; 8.4- not estimable (NE) months). The common grade ≥ 3 adverse events was neutropenia (31.8%), leukopenia (27.3%), lung infection (18.2%) and hyponatremia (18.2%). There was one grade 2 interstitial pneumonia and no treatment related death.

      Conclusions:
      Nab-paclitaxel plus cisplatin was well tolerated and associate with encouraging response outcomes in chemotherapy-naïve patients with advanced NSCLC. Further investigation is warranted.

      Clinical trial identification:
      UMIN000011776 (17 Sep 2013)

      Legal entity responsible for the study:
      N/A

      Funding:
      Taiho Pharmaceutical

      Disclosure:
      Y. Hattori: Honoraria: Astra Zeneca, Boehringer Ingelheim, Chugai Pharmaceutical, Eli Lilly, MSD, Novartis, Ono Pharmaceutical, Taiho Pharmaceutical Research funding: Bristol-Myers Squibb, MSD. S. Morita: Honoraria: Taiho Pharmaceutical. F. Imamura: Honoraria: Taiho Pharmaceutical Research funding: Taiho Pharmaceutical. M. Satouchi: Honoraria: Astra Zeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Chugai Pharmaceutical, Eli Lilly, MSD, Novartis, Ono Pharmaceutical, Pfizer, Taiho Pharmaceutical Research funding: Astellas, Astra Zeneca, Bristol-Myers Squibb, Chugai Pharmaceutical, Eli Lilly, MSD, Ono Pharmaceutical, Novartis, Pfizer. All other authors have declared no conflicts of interest.

    • +

      172P - Safety and efficacy phase 2 study of nab-paclitaxel maintenance treatment after nab-paclitaxel plus carboplatin in stage IIIB/IV non-small cell lung cancer (Now Available) (ID 313)

      12:50 - 12:50  |  Presenting Author(s): A. Nakao  |  Author(s): J. Uchino, N. Nagata, K. Takayama, K. Watanabe, M. Fujita

      • Abstract
      • Slides

      Background:
      Maintenance chemotherapy is being approved as a new treatment paradigm to improve the outcome of advanced NSCLC. Pemetrexed has become recognized as the most promising drug for maintenance therapy, and demonstrated benefits both in progression-free survival (PFS) and overall survival (OS) in the PARAMOUNT study. Since carboplatin plus nab-paclitaxel is also a less toxic regimen, nab-paclitaxel could be a candidate for better maintenance chemotherapy. Therefore, we conducted a phase II study to evaluate the nab-paclitaxel maintenance treatment after nab-paclitaxel plus carboplatin, in terms of safety and efficacy for advanced NSCLC.

      Methods:
      This trial was an open-label, single-arm, multi-center, phase II study. Patients with advanced NSCLC, with no previous systemic chemotherapy (TKIs were allowed), with measurable lesion, and an ECOG PS 0 or 1 participated. Patients received nab-paclitaxel 100 mg/m[2] on days 1, 8, and 15, every 4 weeks in combination with carboplatin at AUC 6 on day 1 of each 4-week cycle for induction. Patients, with no detected disease progression in induction chemotherapy, received nab-paclitaxel monotherapy maintenance until disease progression. The primary endpoint was the PFS. Secondary endpoints were the objective response rate (ORR), OS, safety including peripheral neuropathy, and maintenance therapy transition rate.

      Results:
      A total of 39 patients were enrolled to receive induction therapy, and 15 patients were treated with maintenance nab-paclitaxel; the transition rate was 38.5%. The median PFS, measured from the transition of maintenance therapy, was 6.5 (90%CI: 1.4–11.4) months among 15 patients. PFS was superior to that of the PARAMOUNT study. However, the lower limit of the 90% confidence interval, 1.4 months, was lower than the threshold 3.0 months. Therefore, it was not statistically significant. The most common grade ≥ 3 toxicities observed were hematologic; neutropenia (55.0%), anemia (15.0%), and febrile neutropenia (5.0%), with no increase in the maintenance therapy.

      Conclusions:
      The rate of transition to maintenance therapy was lower than expected. Although nab-paclitaxel may become a candidate regimen for maintenance therapy, this study did not demonstrate statistically significant results because of the small number of enrolments.

      Clinical trial identification:


      Legal entity responsible for the study:
      Department of Respiratory Medicine, Faculty of Medicine, Fukuoka University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      173P - Apatinib plus S-1 showes encouraging response in advanced non-small-cell lung canceras as laterline chemotherapy (Now Available) (ID 320)

      12:50 - 12:50  |  Presenting Author(s): Z. wu  |  Author(s): J. Wu

      • Abstract
      • Slides

      Background:
      There is no standard treatment strategy for patients with advanced non-small cell lung cancer (NSCLC) who experienced progression with three or more lines of chemotherapy. Apatinib, a new tyrosine kinase inhibitor targeting vascular endothelial growth factor receptor 2 (VEGFR-2), has been shown confirming antitumor activity and manageable toxicities in breast and gastric cancers. Clinical trials of apatinib on non-small cell lung cancer show that progression-free survival is improved, but the objective response rate is still low. However, it remains to be explored whether the combined treatment of apatinib plus S1 could be further effective on NSCLC.

      Methods:
      Effects of apatinib, S-1 and apatinib plus s-1 were assessed on two NSCLC cell lines (adenocarcinoma A549 and squamous carcinoma NCI-H520) and xenograft model by injecting NCI-H520 cells. Furthermore, we retrospectively assessed the efficacy and safety of apatinib plus S1 in 12 patients with advanced NSCLC after the failure of second or third-line chemotherapy.

      Results:
      Apatinib plus S-1 strengthened the effect of S-1 and apatinib alone on the NSCLC cell lines, and NCI-H520 was more susceptible. Co-administration delayed the tumour growth than mono-therapy in the xenograft model. There were 12 patients available for efficacy and safety evaluation. 4/12 (33%) patients experienced dose reduction during treatment. Followed up to Dec 20, 2017, the median during time of treatment was 3.5 months. According to RECIST criteria, the disease control rate was 83%, 10/12 (partial response 50%, 6/12 and stable disease 33%, 4/10). Patients with lung squamous cell carcinoma could benefit more than those with lung adenocarcinoma (partial response 100%, 3/3 vs 33%, 3/9). The most frequent treatment-related adverse events were secondary hypertension (41.6%, 5/12), oral mucositis (50%, 6/12), hand-foot syndrome (33%, 4/12) and fatigue (33%, 4/12). Main grade 3 or 4 toxicities were hypertension (16.6%, 2/12), oral mucositis (8.3%, 1/12) and fatigue (8.3%, 1/12).

      Conclusions:
      Apatinib plus S1 exhibits superior activity and acceptable toxicity for the heavily pretreated patients with advanced non-small cell lung cancer. Patients with lung squamous cell carcinoma could benefit more from this treament.

      Clinical trial identification:


      Legal entity responsible for the study:
      Chinese Peoples Liberation Army General Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      174P - Aprepitant, palonosetron and dexamethasone proved effective to prevent chemotherapy-related nausea and vomiting in lung cancer (ID 378)

      12:50 - 12:50  |  Presenting Author(s): Y. Zhang  |  Author(s): N. Yang, F. Zeng, F. Wu

      • Abstract

      Background:
      This study aimed to determine the antiemetic efficacy and safety of aprepitant, palonosetron and dexamethasone in patients with locally advanced or metastatic LC receiving one day full dose cisplatin-based combination chemotherapy (75 mg/m[2]).

      Methods:
      In this open-label, single-arm, single-centre study, 122 patients were enrolled as treatment group, and the antiemetic therapy consisted of palonosetron 0.75 mg on day 1, aprepitant 125 mg on day 1 and 80 mg on days 2–3 and dexamethasone 15 mg on days 1–3. With following, 125 patients were chosen as control group using Propensity Score Matching. The percentage of patients, who received rescue antiemetic medication, with metoclopramide included, was set as the primary endpoint. The secondary endpoints were complete response (CR; no vomiting/retching or rescue medication) in the overall period (0–120 h), and the rate of nausea and vomiting in all the patients who has already been suffered with nausea and vomiting without using aprepitant before. The incidence and severity of nausea were assessed on the basis of the Common Terminology Criteria for Adverse Events v4.03 and a subjective rating scale completed by patients.

      Results:
      There was no difference between two groups with personal characteristics. The percentage of patients who received rescue antiemetic medication (metoclopramide) in treatment group (n = 18) and control group (n = 39) were 14.8% and 37.1% (p < 0.05) respectively. The cumulative occurrence rate of vomiting for all chemotherapy cycles in treatment group and control group were 90.63% and 78.93% (p < 0.05). There was no difference between two groups with the cumulative occurrence rate of nausea for all chemotherapy cycles 69.79% and 73.03% (p > 0.05) respectively. After using aprepitant, the rate of nausea and vomiting were decreased to 37.5% (N = 6, p < 0.05) and 25% (N = 4, p < 0.05) in all the patients who has already been suffered with nausea and vomiting (100%, N = 16) without using aprepitant before. There was no drug related intolerance side effects.

      Conclusions:
      The combination antiemetic therapy was effective and well-tolerated in patients with locally advanced or metastatic LC receiving 1-day full dose cisplatin-based combination chemotherapy.

      Clinical trial identification:
      NCT02161991

      Legal entity responsible for the study:
      Yongchang Zhang

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      175P - A prospective, randomized, double-blind study of pre-chemotherapy hydration with or without mannitol to prevent renal toxicity in non-small cell lung cancer (NSCLC) patients receiving cisplatin-gemcitabine chemotherapy (Now Available) (ID 321)

      12:50 - 12:50  |  Presenting Author(s): N. Thavornpattanpong

      • Abstract
      • Slides

      Background:
      Cisplatin-induced renal toxicity is the most serious adverse event in cisplatin-gemcitabine treated NSCLC patients. Currently there is no gold standard pre-medication regimen that can prevent cisplatin-induced nephrotoxicity.

      Methods:
      We randomized 55 patients who received cisplatin 80 mg/m[2] every 3–4 weeks into two hydration arms. The patients of each arm received one liter of 0.9% sodium chloride solution pre-hydration and also received a half liter of 0.9% sodium chloride solution post hydration. Both arm receives cisplatin (80 mg/m[2] IV: d1) plus gemcitabine (1000 mg/m[2] IV: d1, 8). In the 0.9% sodium chloride solution added with mannitol arm, 40 g of mannitol was mixed with the cisplatin. Chemotherapy was given at 21-day intervals for 6 cycles or until the patient developed intolerable toxicity or progressive disease, whichever occurred first. Clinical parameters, renal function, clinical response and adverse events were recorded. Renal toxicity was defined as more than 20% reduction in creatinine clearance compared with baseline level.

      Results:
      29 patients were randomized to the 0.9% sodium chloride solution arm and 26 patients were randomized to 0.9% sodium chloride solution added with 40 g of mannitol arm. There were no significant differences between randomized groups in age, gender, comorbid disease and baseline creatinine clearance. For each group, decreases in creatinine clearance of more than 20% were 82% in arm 0.9% sodium chloride solution alone and 54% in arm 0.9% sodium chloride solution added with 40 g of mannitol (p-value = 0.021). The decline rate of creatinine clearance in arm 0.9% sodium chloride was greater than in the other arm (p-value = 0.004). There were no differences in the safety profiles.

      Conclusions:
      Hydration with 0.9% sodium chloride solution alone appears to be associated with greater cisplatin nephrotoxicity than 0.9% sodium chloride solution with added mannitol.

      Clinical trial identification:


      Legal entity responsible for the study:
      Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University Thailand

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      176P - PrediCare: A new diagnostic tool predicting imminent disease progression in advanced NSCLC patients by machine-learning integration of three serum biomarkers (ID 495)

      12:50 - 12:50  |  Presenting Author(s): Y. Kogan  |  Author(s): M. Kleiman, S. Shannon, M. Elishmereni, E. Taub, L. Aptekar, R. Brenner, R. Berger, H. Nechushtan, Z. Agur

      • Abstract

      Background:
      In advanced non-small cell lung cancer (NSCLC), most patients deteriorate rapidly and die within 1 year of diagnosis. Forecasting disease progression just prior to its clinical manifestation would allow an earlier switch to the next treatment line, thus preventing major deterioration in the patient's stature and potentially improving response to therapy. However, present serum tumor biomarkers, e.g. carcinoembryonic antigen (CEA), lack the power to signal progression. We developed PrediCare, an innovative diagnostic for continuous monitoring and alerting to forthcoming progression in late-stage NSCLC.

      Methods:
      PrediCare was constructed by machine-learning modeling, and designed to process patient data throughout treatment. Data of late-stage NSCLC patients under first-line standard-of-care therapies, collected in a retrospective observational trial (NCT02577627), served for algorithm training and testing. The algorithm's predictive ability was evaluated using diverse features of 1–3 longitudinally measured serum tumor markers (CEA, CA125, CA15.3), as pre-selected by receiver-operating-characteristic analysis. Performance was evaluated by cross-validation.

      Results:
      A total of 167 NSCLC patients were assessed, the median follow-up time being 101 days. The CEA/CA125/CA15.3 combination showed statistically significant prediction ability, while the use of only 1–2 markers had lower performance. Combining the 3 markers, PrediCare accurately predicted 87/165 of the progression events (52.6% sensitivity), with 15/165 false positives (91.1% specificity). Positive predictive value, negative predictive value, accuracy and Cohen's kappa were 68%, 85%, 81% and 0.47, respectively.

      Conclusions:
      PrediCare is a new individualized medicine software tool, predicting imminent disease progression in advanced NSCLC. This improves treatment planning, and potentially increases survival. Our technology uses standard tumor biomarkers, but integrates three of them in a unique way that offers superiority over their current interpretation in the clinic. Testing of PrediCare under a larger biomarker panel is underway.

      Clinical trial identification:


      Legal entity responsible for the study:
      Optimata Ltd.

      Funding:
      Optimata Ltd.

      Disclosure:
      Y. Kogan, M. Kleiman, S. Shannon, M. Elishmereni, E. Taub, Z. Agur: Employee of Optimata Ltd., which sponsored the study. R. Brenner, R. Berger: Member of the advisory board of Optimata Ltd., which sponsored the study. All other authors have declared no conflicts of interest.

    • +

      177P - Clinicopathologic characteristics in large cell neuroendocrine carcinomas of the lung (LCNCL): The experience of one center (ID 596)

      12:50 - 12:50  |  Presenting Author(s): M. Vaslamatzis  |  Author(s): A. Laskarakis, T.K. Tegos, M. Pavlakis, T. Argyrakos, N. Alevizopoulos, C. Zoublios, E. Vasili, E. Gioti, K. Athanasiadi

      • Abstract

      Background:
      LCNCL is a newly recognized clinicopathologic entity, characterized by the cell morphology and the immunohistochemical evidence of neuroendocrine markers. The optimal treatment for LCNCL is not yet established. The aim of our study was to describe the clinicopathologic findings, the outcome and treatment toxicity in LCNCL patients (pts).

      Methods:
      Twenty-five pts (4%), smokers (>40 pack/year), with LCNCL, among 585 with small cell lung cancer (SCLC), were admitted & treated consecutively in our Unit between 1/1996–12/2017. Patients’ characteristics are shown in the Table. The presenting symptoms, at diagnosis were cough ± heamoptysis ± fever, dyspnea, Superior Vena Cava Syndrome (SVC), bone pain ± subcutaneous nodules & chest pain in 8 (32%), 5 (20%), 4 (16%), 3 (12%) pts, while 5 (20%) absolutely asymptomatic, were diagnosed during routine check-up. Paraneoplastic tetraplegy & myopathy had 1 (4%) and 1 (4%) pts. Eight (32%) pts underwent curative surgery (5 lobectomy, 3 pneumonectomy), while the diagnosis in other pts was established by biopsy of enlarged cervical lymph nodes in 2 (8%), liver biopsy in 1 (4%), adrenal biopsy in 1 (4%) and confirmed by bronchoscopy in all cases. 17 stage IV pts had, at presentation, metastases in: liver, bones, adrenals, CNS, lung and breast in 7 (41%), 8 (47%) 5 (29%), 4 (24%) & 1 (6%) cases.

      Results:
      All pts received Cisplatin-Etoposide ± mediastinal ± cranial RT. Response Rate was documented in 20 (80%), median PFS was 8 (2-93+) months (mo) and OS 12 (4-93+) mo. In median follow up of 1 (1+−93+) mo, 22 (88%) died. Still alive are 3 (12%) patients. One (4%) patient, pT2N2 IIIA underwent left upper lobectomy & then received adjuvant chemotherapy + mediastinal RT + prophylactic cranial irradiation (PCI). He is still alive after 93+ mo. The other 2 with stages IIIA and IV with limited bone disease are in very good partial response for 33+ and 36+ mo. The mPFS and mOS for stages ≤ IIIA and IIIB + IV are 9 vs 3 and 16 vs 6 mo. Grade III & IV febrile leukopenia, thrombocytopenia & anemia had 4, 2 and 2 patients respectively.

      No of pts25
      Median (m) age66 (33–77)ys
      Men/Female19/6
      ECOG
      04 (16%)
      114 (56%)
      23 (12%)
      34 (16%)
      40 (0%)
      Histology
      pure LCNCL21 (84%)
      Mixed LCNCL-SCLC4 (16%)
      cStage
      Ib1 (4%)
      IIa1 (4%)
      IIb1 (4%)
      IIIa4 (16%)
      IIIb1 (4%)
      IV17 (68%)
      Ki-67 (%)60 (40–90%)
      NSE (+)25 (100%)
      TTF-1 (+)24 (96%)
      Chromogranine/Synaptophysin (+)24 (96%)
      CK7 (+)25 (100%)
      CD56 (+)25 (100%)


      Conclusions:
      (1) LCNCL is an unusual type of lung carcinoma with strong correlation with smoking. (2) Surgery seems to be beneficial for early stage disease, but on it's own it doesn't appear to be sufficient & adjuvant chemotherapy consisting of Cisplatin/Etoposite±RT is considered mandatory.

      Clinical trial identification:


      Legal entity responsible for the study:
      Vaslamatzis Michael Head of the Oncology Department Evangelismos General Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      178P - Smo inhibitor LDE225 reverses epithelial-mesenchymal transition (EMT) in non-small cell lung cancer (NSCLC) cells (ID 478)

      12:50 - 12:50  |  Presenting Author(s): Y. Yu  |  Author(s): Y. Cao, B. Pan, S. Jin

      • Abstract

      Background:
      LDE225 is a potent and selective oral inhibitor of Smo, a key component of the hedgehog (Hh) signaling pathway. It has been reported that LDE225 can inhibit the invasion in many solid tumors, and thus attenuate tumor metastasis, but its mechanism of action has been unclear in non-small cell lung cancers(NSCLCs). The aim of the present study was to evaluate whether LDE225 can reverse the epithelial-mesenchymal transition(EMT) induced by co-cultured with macrophage(THP-1) cell lines in NSCLC cells, and its possible mechanisms.

      Methods:
      The NSCLC cells (H460 and H1299) were co-cultured with THP-1 cells for 72h to obsreve EMT formation, and then were treated with 5μm LDE225 for 24h. Wound healing assays and transwell assays were used to detect the migratory and invasive activity of lung cancer cells. The EMT-related factors (E-cadherin and vimentin) were detected by western bloting analysis. SHH and GLI1, as the Hh pathway-related factors, were also detected by western bloting analysis. ELISA assays used to detect the concentration of TGF-β1 in the conditioned medium (CM) obtained from THP-1 cells before and after co-culture with NSCLC cells.

      Results:
      Our data showed that the expressions of E-cadherin and vimentin could significantly modulate after NSCLC cells co-culture with THP-1 cells, which further enhanced the capacity of the NSCLC cells to migrate and invade the extracellular matrix. LDE225 was able to compromise these effects. Additionally, LDE225 remarkably inhibited the activation of Hh signaling pathway in NSCLC cells after co-cultured with THP-1 cells. Further analysis showed that the concentration of TGF-β1 in the CM from co-culture of NSCLC cells & THP-1 cells was significantly lower in the presence of LDE225. It can suppresse the EMT and mimick the effect of LDE225 on NSCLC cells when administration of anti-TGF-β1 neutralizing antibody during co-culture.

      Conclusions:
      Our study confirmd that LDE225 can reverse lung cancer cells EMT and migration via down-regulating the concentration of TGF-β1 in co-culture with THP-1 cells.

      Clinical trial identification:


      Legal entity responsible for the study:
      3rd Affiliated Hospital of Harbin Medical University

      Funding:
      National Natural Scientific Foundation of China (No: 81572824, 81673007 and 81773133), postdoctoral scientific research developmental fund of Heilongjiang Province, Health Department of Heilongjiang Provincial of China (No 2011-124) and Harbin Medical University Cancer Hospital major project Foundation (No: JJZ-2010-01).

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      179P - The cytoplasmic LKB1 promotes the growth of human lung adenocarcinoma by enhancing autophagy (ID 342)

      12:50 - 12:50  |  Presenting Author(s): M.J. Liu  |  Author(s): L. Jiang, X. Fu, X. Liang

      • Abstract

      Background:
      Liver Kinase B1 (LKB1), as a tumor suppression gene, is associated with various kinds of cancers, including lung cancer. However, it is unclear whether LKB1 can be also located in the nucleus of NSCLC cells and if yes, what its function is during the development and progression of NSCLC.

      Methods:
      Full length LKB1 (LKB1~L~) and LKB1 without nuclear localization signal (LKB1~S~) were introduced into lung adenocarcinoma cell A549 respectively. MTT assays, colony formation assays, flow cytometry analysis, transmission electron microscopy and western blot were used to evaluate the effect of LKB1~L~ and LKB1~S~ in vitro. The subcutaneous tumor model in nude mice was used to evaluate the effect of LKB1~L~ and LKB1~S~ in vivo. The expression profile of LKB1 was examined in 190 lung adenocarcinoma tissues by immunohistochemistry and was analyzed combined with clinical parameters.

      Results:
      In this study, we found that cytoplasmic LKB1 promoted the growth of lung adenocarcinoma by enhancing autophagy and it was independent of the AMPK/mTOR signaling. In 190 lung adenocarcinoma samples, we found that LKB1 was expressed in both the nucleus and cytoplasm of lung adenocarcinoma cells and the levels of nuclear LKB1 were inversely associated the levels of cytoplasmic LKB1 in lung adenocarcinoma tissues. Furthermore, the different subcellular localizations of LKB1 were correlated with opposite clinical parameters in this population. While the cytoplasmic LKB1 expression was associated with poor differentiation and advanced TNM stage, the nuclear LKB1 expression was related to well differentiation and early TNM stage. Moreover, the cytoplasmic LKB1 was associated with poor OS in this population and was an independent risk factor for prognosis.

      Conclusions:
      In summary, our data demonstrated that the cytoplasmic LKB1 without nuclear localization signal promoted A549 cell survival in vitro and lung tumor growth in vivo. Furthermore, the cytoplasmic LKB1 was associated with poor prognosis in lung adenocarcinoma cancer. Therefore, our findings may provide new insights into the pathogenesis of lung adenocarcinoma and novel biomarkers for prognosis of lung adenocarcinoma.

      Clinical trial identification:


      Legal entity responsible for the study:
      The First Affiliated Hospital of Xi'an Jiaotong University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      180P - Significant progress in palliative treatment of NSCLC over the last decades: Correlation of treatment milestones with survival in unselected patients (Now Available) (ID 645)

      12:50 - 12:50  |  Presenting Author(s): S.I. Rothschild  |  Author(s): R. Nachbur, N. Herzog, J. Passweg, M. Pless

      • Abstract
      • Slides

      Background:
      Therapeutic options for patients (pts) with metastatic non-small cell lung cancer (mNSCLC) have considerably changed during the last decades. Introduction of 3rd-generation chemotherapeutic agents (1997), molecularly targeted drugs (2009) and immune checkpoint inhibitors (2015) are milestones in the treatment of NSCLC. We analyzed the outcomes of all consecutive patients with mNSCLC in a single institution to determine whether these milestones improved the outcome of unselected patients in a real-world setting.

      Methods:
      576 consecutive patients with palliative treatment for NSCLC at the University Hospital Basel between 1990 and 2016 were analyzed. Probabilities of survival were calculated using the Kaplan-Meier estimator. Groups were compared using the log-rank test. Multivariate analysis was performed to determine factors associated with improved overall survival (OS).

      Results:
      Mean age at diagnosis was 62.9 years, 68% were male, 81% were smokers and 55% had adenocarcinoma. Four cohorts of pts were created according to the described milestones in NSCLC therapy and approval of the drugs. There was a significant statistical difference in median OS for the four-time periods: 1990–1996 8.1 months, 1997–2008 10.7 months, 2009–2014 9.0 months, and 2015–2016 12.4 months (p = 0.027). The four cohorts did not differ significantly in gender, stage, performance and smoking status. In the multivariate analysis, the time period was an independent prognostic factor for OS (p < 0.001). A further independent prognostic factors for OS was sequential therapy with at least two therapies (p = 0.002). Lack of response to 1st-line therapy was an independent negative prognostic factor (p < 0.0001).

      Conclusions:
      Our analysis shows that results obtained in prospective clinical trials are applicable to an unselected real-world population of NSCLC pts. In the period from 2009 to 2014 there was no overall progress, perhaps because introduction of targeted agents benefitted only a minority of selected pts harboring a specific molecular aberration. On the whole, a clinically meaningful and encouraging improvement of survival in m NSCLC was observed in the last decades.

      Clinical trial identification:


      Legal entity responsible for the study:
      University Hospital Basel

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      181P - Real-world comparative effectiveness of treatment sequences in metastatic non-small cell lung cancer with squamous histology (Now Available) (ID 458)

      12:50 - 12:50  |  Presenting Author(s): H. Yang  |  Author(s): B.P. Levy, J.E. Signorovitch, O. Patterson-Lomba, C.Q. Xiang, M. Parisi

      • Abstract
      • Slides

      Background:
      Commonly used first-line (1L) chemotherapies for metastatic non-small cell lung cancer (mNSCLC) include gemcitabine + platinum-based agent (Gem/Platinum), nab-paclitaxel + carboplatin (nab-P/C), and sb-paclitaxel + carboplatin (sb-P/C). Head-to-head trials and prospective data comparing these treatments and subsequent lines of treatment including immunotherapy (IO) are limited.

      Methods:
      This retrospective cohort study compared the efficacy of 1L Gem/Platinum, nab-P/C, and sb-P/C among mNSCLC patients with squamous histology, and among patients who subsequently received 2L IO therapy. Medical records of adult patients who initiated these 1L treatments between 06/2014–10/2015 were reviewed by 132 participating physicians. Overall survival (OS) and progression free survival (PFS) was evaluated using Kaplan-Meier curves, and compared between cohorts using Cox proportional hazards model adjusting for baseline characteristics.

      Results:
      Medical records of 458 mNSCLC patients with squamous histology receiving Gem/Platinum (n = 139), nab-P/C (n = 159), or sb-P/C (n = 160) as 1L therapy were reviewed. Demographics, ECOG and number of comorbidities were balanced among 1L cohorts. 1L nab-P/C was associated with a longer median OS (23.9 months) than Gem/Platinum (16.9 months; adjusted HR vs nab-P/C = 1.55, p < 0.05) and sb-P/C (18.3 months; adjusted HR vs nab-P/C = 1.42, p = 0.10). There were no statistically significant differences in PFS (median PFS of 8.8, 8.0 and 7.6 months for Gem/Platinum, nab-P/C, and Sb-P/C, respectively). 37%, 46% and 40% of patients with 1L Gem/Platinum, nab-P/C and Sb-P/C received a 2L IO. For the subgroup of patients in the three chemotherapy groups who received both 1L chemotherapy and 2L IO therapy, there were no statistically significant differences in OS (median OS of 27.3, 25.0 and 23.0 months, respectively).

      Conclusions:
      In a nationwide sample of mNSCLC patients with squamous histology, 1L nab-P/C was associated with a longer median OS compared with 1L Gem/Platinum, and a statistically non-significant difference compared with 1L sb-P/C. Median OS was similar across 1L agents among the subgroup of patients who sequenced to a 2L IO.

      Clinical trial identification:


      Legal entity responsible for the study:
      Celgene Corporation

      Funding:
      Celgene Corporation

      Disclosure:
      H. Yang, J.E. Signorovitch, O. Patterson-Lomba, C.Q. Xiang,: Employee of Analysis Group, Inc., which has received consultancy fees from Celgene. B.P. Levy: Consultancy fees from Celgene. M. Parisi: Celgene employee.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      182P - Real world anti-PD-L1 treatment (tx) outcomes in a multinational European non-small cell lung cancer (NSCLC) cohort with focus on toxicity (tox) and brain metastases (BM): Preliminary data from an EORTC young investigators lung cancer group collaborative analysis (Now Available) (ID 432)

      12:50 - 12:50  |  Presenting Author(s): R. Tassi  |  Author(s): F. Taylor, S. Canova, L. Low, O. Abdel-Rahman, B. Hasan, E. De Maio, A. Levy, B. Besse, L. Hendriks

      • Abstract
      • Slides

      Background:
      Anti-PD-(L)1 (IO) tx has been approved for treating incurable NSCLC patients (pts) in 1st line and beyond. Outcome of pts with BM is not well known as these pts were often excluded from clinical trials. Similarly, there are limited data in pts that discontinue tx due to tox (TOX+). We evaluated outcome of pts with BM at baseline IO (BM+) and IO tox pts in a real world European cohort.

      Methods:
      Retrospective multicenter data collection (1 NL, 1 UK, 2 IT) including all consecutive IO treated NSCLC pts. Primary outcomes: progression free and overall survival (PFS/OS) from start IO to progression (PD) or death, respectively of BM+ pts and TOX+ pts (≥gr 2 IO related tox). Secondary: BM development during IO.

      Results:
      Between 05-′15 and 10-′17 150 pts received IO: 26% pembro, 74% nivo. 5.3%, 60.7% and 34% received IO as 1st, 2nd or ≥2 line, respectively. 60.7% male, median age 65.7 years, WHO PS 0-1/2-3/unknown: 89.3/9.3%/1.3%, respectively, 87% current/former smoker, 65% non-squamous histology, 4% EGFR/ALK driver mutation (pretreated with TKI). 25 pts (16.7%) BM+ (1 leptomeningeal (LM), 81% of whom pretreated with radiation), baseline characteristics did not significantly differ between BM+ and BM−. 12.6% of all pts developed/had PD of BM during IO. Median FU was 7.8 months (mo). Median PFS was 11.7 vs 4.6 mo (p < 0.001) for BM+ vs BM-, median PFS of the LM pt was 0.3 mo. Median OS was not reached for BM+ pts and was 9.4 mo for BM- pts (p < 0.001). 46 pts (30.6%) had grade ≥ 2 IO related tox, 13 pts (8.7%) discontinued IO without having PD (76.9% grade ≥3). Median time on IO for TOX+ discontinuing pts was 4.8 mo. 38.5% of the TOX+ pts had PD after discontinuation. Median PFS and OS of TOX+ vs TOX- pts was 16.3 vs 10.2 and 17.7 vs 15.0 mo respectively with a trend towards longer survival for TOX+ pts (both p ns).

      Conclusions:
      With limited FU, (irradiated) BM+ is not a negative prognostic marker in IO treated pts, as PFS/OS are significantly longer compared to BM-. Development/PD of BM during IO is low. Discontinuation because of tox while responding does not negatively impact on survival in most pts.

      Clinical trial identification:


      Legal entity responsible for the study:
      Authors

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      183P - Progression of central nervous system metastases in advanced non-small cell lung cancer patients effectively treated with first-generation EGFR-TKI (Now Available) (ID 395)

      12:50 - 12:50  |  Presenting Author(s): M. Yu  |  Author(s): Q. Zhao, Y. Li, S. Zhang, X. Zhou, J. Wang, Y. Wang, Y. Lu, Y. Liu

      • Abstract
      • Slides

      Background:
      Central nervous system (CNS) progression is frequently detected in patients with favorable initial responses to first-generation EGFR-TKIs, but data are incomplete with respect to clinical features and prognostic factors of CNS failure in this population.

      Methods:
      We retrospectively evaluated 420 advanced non-small cell lung cancer (NSCLC) patients treated with first-generation EGFR-TKI for over 3 months. We analyzed CNS progression of these patients, defining as newly developed CNS metastases or progression of preexisting brain lesions after EGFR-TKI treatment.

      Results:
      Of the 420 patients, 99 (23.6%) with CNS progression after EGFR-TKI initiation were identified. The median time to CNS progression was 12 months (95% CI 10.5–13.5). Patients with L858R mutation were more likely to experience CNS failure than those with exon 19 deletion (P = 0.008). For patients with previous brain metastases, the median time to CNS progression of patients with EGFR-TKI and local CNS therapy was significantly longer than those treated with EGFR-TKI alone (14.0 months vs. 11.2 months; P = 0.016). The median survival time after CNS progression was 11.2 months (95% CI 8.8–13.5). L858R mutation, multiple brain metastases progression and CNS with other site failures were negative prognostic factor, while localized CNS therapy was the only significant favorable prognostic factor for overall survival (OS) after CNS progression.

      Conclusions:
      For advanced NSCLC patients treated successfully with first-generation EGFR-TKI, careful monitoring for CNS progression should be considered, especially for those with L858R mutation. Localized CNS therapy should be considered for CNS progression.

      Clinical trial identification:
      This study was approved by the Institutional Review Board of West China Hospital, Sichuan University and informed consent was obtained from each patient for the use of tissue in molecular analysis

      Legal entity responsible for the study:
      West China Hospital, Sichuan UniverWest China Hospital, Sichuan University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      184P - Clinico-radiological pattern of response to nivolumab in stage IV NSCL: A real life experience over two years (Now Available) (ID 627)

      12:50 - 12:50  |  Presenting Author(s): M. Perna  |  Author(s): V. Scotti, C. Muntoni, C. Moroni, F. Giannelli, D. Cozzi, F. Mazzoni, V. Bonti, D. Lavacchi, L. Livi

      • Abstract
      • Slides

      Background:
      Immune checkpoint inhibitors are novel and effective drugs in the treatment of stage IV non-small cell lung cancer (NSCLC). These drugs are associated with a specific immune-related patterns of response, including cases of pseudoprogression (Seymour L Lancet Oncol 2017) and hyperprogression (as defined as > 50% tumor burden compared with basal CT; Kato S Clin Cancer Res 2017).

      Methods:
      Our purpose was to evaluate the pattern of response in a cohort of patients with stage IIIB-IV NSCLC treated with nivolumab in two oncologic units. Radiologic evaluation was correlated with clinical benefit from the treatment in all the series.

      Results:
      We overall selected 86 patients, treated between August 2015 and August 2017. 40 patients were excluded because of absence of a target lesion, previous radiotherapy to a target lesion, follow up inferior to three months. 46 patients (12 males, 34 females) were considered for radiological review, made by a senior and a junior radiologists specialized in thoracic imaging. Median age was 68 years (range 49–84). Stage of disease was IIIB in 12 patients, IV in 34 cases. Histology was adenocarcinoma in 23 patients (50%), squamous cell carcinoma in 23 (50%). 31 patients underwent nivolumab as second line treatment, 15 patients in third line or more. Median number of cycles was 8 (range 1–48). After a radiological review according to iRecist criteria, pseudoprogressions were observed in 4 patients (9%); discrepancy between clinical benefit and radiological response occurred usually at first or second evaluation. Median time to pseudoprogression was 6,2 months (5,3–13). One patient experienced hyperprogression, with rapidly decreasing performance status.

      CharateristicsPatients (total number: 46)
      Sex
       Male 33 (71,7%)
       Female 13 (28,3%)
      Pack years
       0 5 (10,9%)
       ≤30 10 (21,7%)
       >30 26 (56,5%)
       ND 5 (10,9%)
      Stage
       IIIb 12 (26%)
       IV 34 (74%)
      Histology
       Adenocarcinoma 23 (50%)
       Squamous cell carcinoma 23 (50%)
      Number of cycles
       ≤10 23 (50%)
       10–20 10 (21,8%)
       20–30 8 (17,4%)
       >30 6 (10,8%)
      Radiological results
       Pseudoprogression 4 (9%)
       Hyperprogression 1 (2,2%)


      Conclusions:
      Pseudoprogression in immunotherapy treatment is a rare but possible event in NSCLC treatment. Immunotherapy treatment should be carefully interrupted in patients with clinical benefit. A small percentage of patients do not benefit from immunotherapy and have a rapid progression of disease during the first courses of immunotherapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Prof. Lorenzo Livi

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      185P - Retrospective study of tumour responses and toxicities associated with anti-PD1 antibody therapy in elderly cancer patients(>75yo) in comparison to patients ≤75yo with metastatic non-small cell lung cancer (Now Available) (ID 324)

      12:50 - 12:50  |  Presenting Author(s): L.C. CHONG  |  Author(s): N. Corsini, J. Hogan-Doran, M. Brown

      • Abstract
      • Slides

      Background:
      Antibodies directed against PD1 (anti PD1) are approved in Australia for the second line treatment of metastatic non-small cell lung cancer (met NSCLC). There is limited data on the efficacy and tolerability of anti-PD1 in elderly cancer patients. The purpose of this study was to retrospectively evaluate the responses and toxicities in elderly (>75yo) compared with younger patients (≤ 75yo) with met NSCLC treated with anti-PD1.

      Methods:
      56 patients (36 patients ≤75yo; 20 patients >75yo) with metastatic non-small cell lung carcinoma treated with anti-PD1 at Royal Adelaide Hospital, Australia, between January 2010 to January 2017 were retrospectively identified. Patient demographics, metastatic non-small cell lung cancer characteristics, response rates, toxicity profiles and survival data were collected. Statistical comparisons were made between the older (>75yo) and younger (≤75yo) patients.

      Results:
      Adenocarcinoma was the most common histological type of non-small cell lung cancer in both groups. Patients that achieved stable disease in ≤75yo (20/36, 55.6%) were slightly lower compared with >75yo (15/20, 75%). Patients that progressed on anti-PD1 were higher in ≤75yo (12/36, 33.3%) compared with >75yo (5/20, 25%). Median overall survival (OS) and progression-free survival (PFS) was 8.0 months (95% CI, 0.00 to 16.6 months) and 5.0 months (95% CI, 1.47 to 8.53 months), respectively, in the ≤75yo group and 20.0 months (95% CI, 9.2 to 30.8 months) and 6.0 months (95% CI, 4.55 to 7.45 months), respectively, in the >75yo. The log-ranked test indicates that the survival distributions do not differ for OS or PFS (OS, p = .88 and PFS, p > .05). The rates of adverse events were similar between both groups with the most common adverse event being rash and fatigue (grade 1–2). Anti-PD1 treatment discontinuation occurred in 26/36 (72.2%) of ≤75yo and 17/20 (85%) of >75yo. Main reason for treatment discontinuation was progression of disease, 23/36 (63.9%) of ≤75yo and 15/20 (75%) of >75yo.

      Conclusions:
      Anti-PD1 antibodies are safe and effective in elderly patients with metastatic non-small cell lung cancer. Response rate and toxicity profiles are similar between ≤75yo and >75yo.

      Clinical trial identification:


      Legal entity responsible for the study:
      Authors

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      186P - Thyroid toxicity as an immune-related adverse event in patients with non-small cell lung cancer during treatment with immune checkpoint inhibitors (Now Available) (ID 364)

      12:50 - 12:50  |  Presenting Author(s): K. Syrigos  |  Author(s): A. Charpidou, A. Asimakopoulou, I. Vathiotis, I. Gkiozos, S. Tsagouli, P. Boura, D. Grapsa, E. Kotteas

      • Abstract
      • Slides

      Background:
      Programmed cell death protein 1(PD-1)/programmed death-ligand 1(PD-L1) inhibitors have improved outcomes for cancer patients, in exchange for novel immune-related adverse events (irAEs), including thyroid toxicity that is documented in up to 10% of cases.

      Methods:
      The medical records of 42 patients with stage IV non-small cell lung cancer (NSCLC), treated with anti-PD-1/PD-L1 monoclonal antibodies, were retrospectively evaluated. Thyroid function tests (TFTs) were assessed both at baseline and at regular 6-week intervals. Demographics and clinicopathological features of patients and frequency, grade, clinical course and management of thyroid toxicity were recorded and analyzed.

      Results:
      Thyroid toxicity was observed in 12 cases (28.6%). 6 patients developed hyperthyroidism, which was only transient in 4 of them (66.7%), before advancing to hypothyroidism. De novo hypothyroidism ensued in 2 patients. The remaining 4 patients were hypothyroid, receiving hormone replacement therapy at baseline and needed dose adjustment due to thyroid toxicity. Median time on treatment until development of thyroid toxicity was 47 days (range from 15 to 251 days). Grade I events were recorded in 6 patients (50%), grade II in 5 (41.7%) and only one patient (8.3%) experienced grade III thyroid toxicity. Oral levothyroxine was prescribed for symptomatic hypothyroidism. B-blockers in combination with low doses of oral methylprednisolone (a maximum of 16 mg per day) were administered in symptomatic hyperthyroid patients. TFTS became normal in 11 patients; 4 individuals needed long-lasting hormone replacement. None of our patients had to permanently discontinue anti-cancer immunotherapy.

      Conclusions:
      A relatively high rate of immune-mediated thyroid toxicity was observed in our cohort. Low doses of oral methylprednisolone were effective in controlling clinical hyperthyroidism, while grade III/IV thyroid toxicity was rare. Further studies are warranted to explore the possibility of an increased susceptibility to immune-mediated thyroid-related adverse events in the Greek population, and the factors underlying this phenomenon.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Athens

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      187P - Nivolumab-related immune-related adverse events in advanced NSCLC predict therapeutic objective response (Now Available) (ID 461)

      12:50 - 12:50  |  Presenting Author(s): J. Rogado  |  Author(s): V. Pacheco Barcia, B. Vera, P. Toquero, R. Mondejar, A.I. Ballesteros Garcia, O. Donnay, N. Romero Laorden, R. Colomer Bosch, J.M. Sánchez-Torres

      • Abstract
      • Slides

      Background:
      It has been described that the efficacy of Nivolumab, an anti-PD-1 inmune checkpoint inhibitor antibody, in melanoma is associated with the development of immune-related adverse events (irAEs). This relationship is not defined in other malignant diseases. Our study explores whether Nivolumab-induced irAEs in Non-Small-Cell Lung Cancer (NSCLC) are associated with treatment efficacy.

      Methods:
      We carried out a review of the medical records of 40 cases of NSCLC treated with Nivolumab between January 2015 and May 2017 at the Hospital Universitario de La Princesa of Madrid. The efficacy of Nivolumab was evaluated using objective response (OR) criteria of iRECIST, progression-free survival (PFS) and overall survival (OS). Odds Ratio tests were performed to determine the association between irAEs and OR and log-rank and cox proportional hazards models for PFS and OS.

      Results:
      25% of the patients treated with Nivolumab developed irAEs (10/40). The most frequent toxicity was hypothyroidism, with 6 cases, five of them in grade 1, and one grade 3. Other toxicities were hepatitis, colitis, nephritis and hyperthiroidism immune-related (one patient each, all grade 1–2). Nivolumab response was as follows: 1 patient had complete response and 13 patients had partial response (objective response -OR-, 14/40: 35%); 17 patients had stable disease, and 9 patients had progressive disease at the first evaluation. Six of the 10 patients developing irAEs had an OR, compared to 8/30 patients without IRAEs (60% vs. 26%, OR 4.1, IC95% 0.83–20.3, p = 0.056). Patients who developed irAEs had similar PFS (6 vs 5.5 months, HR 1.59, CI 95% 0.7–3.7, p = 0.2) and OS (28 vs 21 months, HR 1.8, CI 95% 0.8–4.5, p = 0.1).

      Conclusions:
      We have found an association between the development of Nivolumab related irAEs and the OR using iRECIST criteria, in patients with NSCLC, but not PFS or OS. Patients who develop irAEs presents four times more probability to have an OR to Nivolumab.

      Clinical trial identification:


      Legal entity responsible for the study:
      Instituto de investigación sanitaria Princesa

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      188P - Treatment switching–adjusted overall survival (OS) in KEYNOTE-024: First-line pembrolizumab versus chemotherapy in patients with advanced non-small cell lung cancer (NSCLC) (ID 622)

      12:50 - 12:50  |  Presenting Author(s): M. Reck  |  Author(s): D. Rodríguez-Abreu, A.G. Robinson, R. Hui, T. Csőszi, K. Vandormael, W. Malbecq, M..C. Pietanza, J. Brahmer

      • Abstract

      Background:
      In KEYNOTE-024, pembrolizumab was superior to chemotherapy for advanced untreated NSCLC with PD-L1 tumor proportion score (TPS) ≥50% and no sensitizing EGFR mutations/ALK translocations. We present OS adjusted for the potential bias introduced by switching treatment from chemotherapy to pembrolizumab after a median of 25.2 months of follow-up.

      Methods:
      Patients were randomized to pembrolizumab 200 mg every 3 weeks or investigator-choice platinum-doublet chemotherapy with optional pemetrexed maintenance for patients with nonsquamous histology. Patients in the chemotherapy arm could switch to pembrolizumab (prior to the second interim analysis that revealed superiority, only patients with progressive disease confirmed by blinded, independent central radiology review were eligible). Three statistical methods were applied to adjust for treatment switching: the simplified 2-stage method, the rank preserving structural failure time (RPSFT) method, and the inverse probability of censoring weighting (IPCW) method.

      Results:
      305 patients were randomized; all but 1 patient in the chemotherapy arm received protocol-specified treatment. 82 (54%) patients had switched from chemotherapy to pembrolizumab per protocol. OS results per the intent-to-treat (ITT) analysis and the 3 adjustment methods are listed in the Table. The 3 methods adjusting for treatment switching in the chemotherapy arm provided larger treatment estimates (lower HRs) than the ITT estimate. As there was some evidence towards a deviation from the common treatment effect assumed in the RPSFT method, and the IPCW method is more prone to bias in the presence of relatively small sample sizes, the simplified 2-stage method was the favored method in this study. Results obtained with the RPSFT and IPCW methods were consistent with those from the 2-stage analyses.Table: (Abstract: 188P)Treatment switching–adjusted OS in KEYNOTE-024 after median follow-up of 25.2 months

      Median OS, months (95% CI)Pembrolizumab(n = 154)Chemotherapy(n = 151)HR (95% CI)
      ITT analysis30.0 (18.3–not reached)14.2 (9.8–19.0)0.63 (0.47–0.86) 2-sided log-rank P = 0.003
      2-stage methodN/A8.7 (7.3–11.5)0.49 (0.34–0.69)
      RPSFT methodN/A11.8 (8.7–not reached)0.52 (0.33–0.75)
      IPCW methodN/A11.8 (8.7–21.3)0.52 (0.33–0.80)


      Conclusions:
      In this study, pembrolizumab demonstrated an OS advantage compared to chemotherapy as first-line therapy, which becomes more pronounced when utilizing treatment switching analyses that adjust for bias introduced when switching from chemotherapy to pembrolizumab. Pembrolizumab remains a standard-of-care first-line therapy for patients with advanced NSCLC expressing PD-L1 TPS ≥50%.

      Clinical trial identification:
      NCT02142738

      Legal entity responsible for the study:
      Merck & Co., Inc., Kenilworth, NJ, USA

      Funding:
      This study and medical writing assistance were funded by Merck & Co., Inc., Kenilworth, NJ, USA.

      Disclosure:
      M. Reck: Advisory board member for Roche, Lilly, AstraZeneca, BMS, MSD, Merck, Boehringer-Ingelheim, Celgene, Pfizer, and Novartis; Speakers’ bureaus for Roche, Lilly, AstraZeneca, BMS, MSD, Merck, Boehringer-Ingelheim, Celgene, Pfizer, and Novartis; Research funding from Boehringer-Ingelheim. A.G. Robinson: Honoraria from Merck & Co., Inc., Kenilworth, NJ; his institution has received research funding from AstraZeneca, Merck & Co., Inc. (Kenilworth, NJ USA), Bristol-Myers Squibb, and Roche Canada. R. Hui: Honoraria from MSD, BMS, AstraZeneca, Boehringer-Ingelheim, and Pfizer; Consulting or Advisory role for MSD, AstraZeneca, Pfizer, and Novartis. K. Vandormael: Employee of MSD Europe. W. Malbecq: Employee of and owns stock in MSD Europe. M.C. Pietanza: Employee of Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc., Kenilworth, NJ USA. J. Brahmer: Advisory board member for BMS (uncompensated), Celgene, Eli Lilly, Merck; Research funding from BMS, MedImmune/AstraZeneca, Merck. All other authors have declared no conflicts of interest.

    • +

      189P - Are pretreatment neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR) and advanced lung cancer inflamation index (ALI) levels useful in predicting the outcomes of patients with advanced non-small cell lung cancer? (Now Available) (ID 429)

      12:50 - 12:50  |  Presenting Author(s): B. Komurcuoglu  |  Author(s): A. Evkan, G. Karakurt

      • Abstract
      • Slides

      Background:
      Systemic inflammation is both involved in the pathogenesis of cancer and can be activated by oncogenic changes in cancer cells. Inflammation parameters in cancer patients can increase due to different reasons such as cachexia, anorexia, pain. Systemic inflammatory markers in cancer are easily calculated using routine laboratory data, which can be used to identify high-risk patients, and predict the progression of the disease.

      Methods:
      Between January 2013 and December 2015, 115 cases with advanced lung cancer diagnosed at our hospital were retrospectively reviewed. The cases were staged according to TNM 7. NLR (neutrophil lymphocyte ratio), PLR (platelet lymphocyte ratio) and BMI (body mass index) and ALI (advanced lung cancer inflammation index) values of patients were calculated. ALI was calculated as (BMI x Alb/NLR). For ALI 18, NLR 5, PLR 250 cut off levels were accepted. These values were compared with progression-free survival and overall survival of patients.

      Results:
      Our study of 115 patients, 15 (13.04%) were female and 100 (86.96%) were male. The mean age of the cases was 61.51 + 9.69 (min 41, max 83). 38 patients were stage 3B and 77 patients were stage 4. Patients histopathologic patterns were 43 adenocarcinomas, 37 squamous cell, 27 small cell and 8 non-small cell lung cancer. 19 of the patients were palliatively treated according to performance status and additional chronic illnesses. 73 cases were chemotherapy (CT), 7 cases were CRT, 13 cases were consecutive CT-RT, 1 case was operated on after induction, 2 cases only thoracic RT was applied. There were NLR <5.00 77 (67%) and >5.00 38 (33%) patients for overall survival (OS). The average time for OS under NLR 5.00 was 9.67 months, 5.95 per month above 5.00. A statistically significant difference was observed in the NLR value for the time period up to the OS (p:0.026; p < 0.05). OS was observed in 44 (38%) of the patients under levels of 18.00 ALI, and 71 (62%) of patients over 18.00 ALI. The average time for OS under ALI 18.00 was 4.24 months versus 10.13 months for the others. A statistically significant difference was observed between the ALI value and the OS (p:0.000; p < 0.05). There was no significant difference between PFS and OS for PLR value (p > 0.05).

      Conclusions:
      We found that the NLR and ALI values at diagnosis were related to survival in patients with advanced lung cancer, and that the cut-off values indicated may be useful in predicting the survival and prognosis of the patient.

      Clinical trial identification:


      Legal entity responsible for the study:
      Izmir Dr. Suat Seren Education Hospital Etic Comitty

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      190P - Are neutrophil-to-lymphocyte ratio (NLR) and neutrophil count percentage (NCP) predictors of nivolumab outcome and toxicity in NSCLC? (Now Available) (ID 463)

      12:50 - 12:50  |  Presenting Author(s): J. Rogado  |  Author(s): V. Pacheco Barcia, P. Toquero, B. Vera, R. Mondejar, N. Romero Laorden, A.I. Ballesteros Garcia, O. Donnay, R. Colomer Bosch, J.M. Sánchez-Torres

      • Abstract
      • Slides

      Background:
      There is limited data of the effect of inflammatory markers on Nivolumab efficacy. We assessed the association between NLR and NCP in NSCLC and the efficacy of Nivolumab. In order to establish whether the effect was either predictive or prognostic, we studied NLR and NCP in two independent cohorts, one treated with Nivolumab and one treated with chemotherapy. Finally, we also analyzed the relationship of NLR and NCP with immune-related adverse events (irAEs).

      Methods:
      Data from 40 NSCLC patients treated with Nivolumab between January 2015 and May 2017 were retrospectively collected. Population was dichotomized according to NLR of 5 and NCP of 80%. The association between NLR or NCP and progression free survival (PFS) and overall survival (OS) was analyzed by univariate and multivariate models. A cohort of 54 chemotherapy-treated NSCLC patients was also analyzed. The association between development of irAEs and NLR or NCP was estudiad by chi square test and Odds Ratio.

      Results:
      Multivariate analysis demonstrated that patients treated with Nivolumab and baseline NLR < 5 have a favourable PFS (6 vs 2 months, HR 8.3, p < 0.001) and OS (26 vs 10.5 months, HR 4.40, p < 0.000001) than cases with NLR ≥ 5. Patients treated with nivolumab and NCP < 80% have also a favourable PFS (6 vs 1.5 months, HR 0.10, p < 0.001) and OS (21.5 vs 9.5 months, HR 0.21, p < 0.05) than cases with NCP ≥ 80%. Patients who received chemotherapy and NLR < 5 have significative better PFS and OS too (15.5 vs 6.5 months, HR 5.9, p < 0.00001) and OS (24 vs 17 months, HR 6.7, p < 0.0001), but this was not observed with NCP < 80%, since no significant diferences showed in the multivariate analysis (PFS 4 vs 2.5 months, p = 0.8; OS 14 vs 9.5 months, p = 0.54). In our analysis, NLR and NCP were not associated with the development of irAEs.

      Conclusions:
      Neutrophil count percentage is a predictive biomarker of Nivolumab clinical course in NSCLC. NCP < 80% was associated with improved PFS and OS. In contrast, neutrophil-to-lymphocyte ratio had a prognostic but not a predictive value. Our results may relevant in the future when patients with NSCLC initiate nivolumab therapy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Instituto de investigación sanitaria Princesa

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      192P - KRAS as predictive biomarker of response to checkpoint inhibitors in NSCLC (Now Available) (ID 538)

      12:50 - 12:50  |  Presenting Author(s): A. Addeo  |  Author(s): J. Torralvo, P. Dietrich

      • Abstract
      • Slides

      Background:
      AntiPD1/antiPDL1-based immunotherapy has changed dramatically the prognosis of non small cell lung cancer (NSCLC)pwith a substantial improvement of overall survival (OS) and even presenting long lasting responses in a subset of patients (pts). Several factors have been associated with the likelihood of better survival, which include the smoking exposure and the presence of KRAS-mutation according to data from randomized clinical trials that compared chemotherapy to these immunotherapeutic (IO) agents.

      Methods:
      We have collected all the cases of stage IV NSCLC pts treated with IO agents within our institution form January 2016 to december 2017 with particular interest to pts harbouring KRAS –mutation. We have assessed their clinical outcomes: disease control rate (DCR) Partial response (PR), progression free survival (PFS) and overall survival (OS).

      Results:
      45 patients with advanced NSCLC were treated with anti PD1 or anti PDL1 (Nivolumab 71%, Pembrolizumab 22% and Atezolizumab 7%, respectively) from January 2016 to December 2018 within our Institution. Twenty (44%) pts were identified as Adenocarcinoma with KRAS-mutation. The subgroup included 39% of female, median age of 61 years, and all patients had PS0-1. The immunotherapy (IO) was administered as 1st, 2nd and ≥ 3rd therapy in 7%, 82% et 11% of the 45 pts, respectively: DCR was 74% with 49% PR and in 40% of pts the response lasted ≥12 months. Within the KRAS mutation group (20 pts) the DCR was 83.5% and the PR was 67%. Within this cohort the PFS was 9.5 months and OS was 42 months. At the time of analysis 7/45 pts were still receiving the immunotherapy treatment.

      Conclusions:
      Our retrospective analysis has showed that KRAS-mutation pts represent a subgroup of pts that seem to substantially benefit from IO agents in terms of both response rate, PFS and OS. This is in line with the literature and despite the limitations of a retrospective analysis, it confirms the possible strong correlation among KRAS mutation and response to IO. Further prospective studies are warranted to better define the role of KRAS mutation in this context.

      Clinical trial identification:


      Legal entity responsible for the study:
      HUG

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      193TiP - Phase 3 study of epacadostat plus pembrolizumab with or without platinum-based chemotherapy vs pembrolizumab plus chemotherapy for first-line metastatic non-small cell lung cancer (mNSCLC): ECHO-306/KEYNOTE-715 (ID 415)

      12:50 - 12:50  |  Presenting Author(s): M. Garassino  |  Author(s): S. Rubin, Y. Zhao, Y. Luo, A. Samkari, R. Hui

      • Abstract

      Background:
      Combination therapies targeting broad immunosuppression in the tumor microenvironment (TME) may prolong survival. Upregulation of the indoleamine 2,3 dioxygenase-1 (IDO1) enzyme within TME contributes to an immunosuppressive state responsible for tumor escape from immune surveillance. Epacadostat (E), a potent and selective inhibitor of IDO1, is under investigation in several tumor types. Pembrolizumab (P) is FDA-approved for NSCLC. In a phase 1/2 study, E + P showed minimal additive toxicity vs monotherapy and encouraging efficacy in NSCLC. The ECHO-306/KEYNOTE-715 global study compares the efficacy and safety of first-line E + P±chemotherapy vs P + chemotherapy in patients with mNSCLC.

      Trial design:
      Eligible patients: age ≥18, stage IV NSCLC (no EGFR-sensitizing mutation and ROS1/ALK translocations), ECOG PS ≤1, no prior systemic therapy for mNSCLC, and no prior IDO1 inhibitors or immune checkpoint therapies. Patients (∼1062) will be randomized 1:1:1 (E 100 mg oral BID + P 200 mg IV Q3W±platinum-based chemotherapy [nonsquamous: pemetrexed 500 mg/m[2] + cisplatin 75 mg/m[2] or carboplatin AUC 5 Q3W for 4 cycles followed by pemetrexed 500 mg/m[2] Q3W; squamous: paclitaxel 175–200 mg/m[2] + carboplatin AUC 5–6 Q3W for 4 cycles]; or E-matched placebo + P + chemotherapy) and stratified by tumor histology (squamous vs nonsquamous), PD-L1 status (tumor proportion score <50% vs ≥50%), ECOG PS (0 vs 1), and region (East Asia vs non-East Asia). Patients will receive treatment for ≤35 cycles of P or until disease progression (PD), intolerable toxicity, or investigator/patient decision to withdraw. Treatment may continue after initial radiographic PD in eligible patients. Patients may discontinue treatment after confirmed complete response. Primary endpoints: OS and PFS. Secondary endpoints: ORR per RECIST v1.1 (blinded central review; weeks 6, 12, 18, Q9W up to week 54 and Q12W thereafter), safety and tolerability, and E pharmacokinetics. Adverse events (AEs) will be monitored throughout the study and for 30 days (90 days for serious AEs) after end of treatment and graded per CTCAE v4.0.

      Clinical trial identification:
      NCT03322566

      Legal entity responsible for the study:
      Incyte Corporation, Wilmington, DE, USA and Merck & Co., Inc., Kenilworth, NJ, USA

      Funding:
      Incyte Corporation, Wilmington, DE, USA and Merck & Co., Inc., Kenilworth, NJ, USA

      Disclosure:
      M. Garassino: Received research funding from AstraZeneca (Inst), Bristol-Myers Squibb (Inst), Lilly (Inst), Merck Sharp & Dohme (Inst), and Roche (Inst), and received travel/accommodations/expenses from AstraZeneca, Bristol-Myers Squibb, and Roche. S. Rubin, Y. Zhao: Employee and stockholder of Incyte. Y. Luo, A. Samkari: Employee and stockholder of Merck. R. Hui: Advisory board member for Merck Sharp and Dohme, AstraZeneca, Novartis, Roche, and Bristol-Myers Squibb. Received speaker honorarium from Merck Sharp and Dohme.

    • +

      194TiP - eXalt3: Phase 3 randomized study comparing ensartinib to crizotinib in anaplastic lymphoma kinase (ALK) positive non-small cell lung cancer (NSCLC) patients (ID 644)

      12:50 - 12:50  |  Presenting Author(s): L. Horn  |  Author(s): Y. Wu, M. Reck, C. Liang, F. Tan, K. Harrow, V. Oertel, G. Dukart, T.S.K. Mok

      • Abstract

      Background:
      Ensartinib (X-396) is a novel, potent ALK small molecule tyrosine kinase inhibitor (TKI) with additional activity against MET, ABL, Axl, EPHA2, LTK, ROS1 and SLK. Ensartinib is well-tolerated and has shown promising activity in NSCLC patients in a phase 1/2 study in patients that were both ALK TKI naïve and patients that received prior crizotinib, as well as those with CNS metastases. The safety profile of ensartinib appears to be different than other ALK TKIs.

      Trial design:
      In this global, phase 3, open-label, randomized study, approximately 270 patients with ALK+ NSCLC who have received no prior ALK TKI and up to one prior chemotherapy regimen will be randomized with stratification by prior chemotherapy (0/1), performance status (0 − 1/2), brain metastases at screening (absence/presence), and geographic region (Asia/other), to receive oral ensartinib (225 mg, once daily) or crizotinib (250 mg, twice daily) until disease progression or intolerable toxicity.Eligibility also includes patients ≥ 18 years of age, stage IIIB or IV ALK+ NSCLC. Patients are required to have measurable disease per RECIST 1.1, adequate organ function, and an ECOG PS of ≤ 2. Adequate tumor tissue (archival or fresh biopsy) must be available for central testing. The primary endpoint is progression-free survival assessed by independent radiology review based on RECIST v. 1.1 criteria. Secondary efficacy endpoints include overall survival, response rates (overall and central nervous system [CNS]), PFS by investigator assessment, time to response, duration of response, and time to CNS progression. The study has > 80% power to detect a superior effect of ensartinib over crizotinib in PFS at a 2-sided alpha level of 0.05.Phase 3 recruitment began in June, 2016 and currently has 98 active sites in 20 countries. The duration of recruitment will be approximately 24 months.

      Clinical trial identification:
      NCT02767804

      Legal entity responsible for the study:
      Xcovery Holding Corporation

      Funding:
      Betta Pharmaceuticals

      Disclosure:
      L. Horn: Consulting for Xcovery Holding Company, BMS, BI, Abbvie, Genentech, Merck. Y-L. Wu: Speaker fees from AstraZeneca, Roche, Eli Lily. M. Reck: Honoraria for lectures and consultancy: Hoffman-La Roche, Lily, BMS, MSD, AstraZeneca, Merck, Celgene, Boehringer-Ingelheim, Pfizer, Novartis. C. Liang, K. Harrow, V. Oertel, G. Dukart: Full-Time Employee: Xcovery Holding Corporation, F. Tan: Manager: Xcovery Holding Company Chief Medical Officer: Betta Pharmaceuticals. T.S.K. Mok: Grant/Research Support from AstraZeneca, BI, Pfizer, Novartis, SFJ, Roche, MSD, Clovis Oncology, BMS, Eisai, Taiho; Speaker's fees with: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, MSD, Novartis, BMS, Taiho; Major Stock Shareholder in: Sanomics Ltd.; Advisory Board for: AstraZeneca, Roche/Genentech, Pfizer, Eli Lilly, BI, Clovis Oncology, Merck Serono, MSD, Novartis, SFJ Pharmaceutical, ACEA Biosciences, Inc., Vertex Pharmaceuticals, BMS, geneDecode Co., Ltd., OncoGenex Technologies Inc., Celgene, Ignyta, Inc.; Board of Directors: IASLC, Chinese Lung Cancer Research Foundation Ltd., Chinese Society of Clinical Oncology (CSCO), Hong Kong Cancer Therapy Society (HKCTS).

    • +

      195TiP - A phase 3, randomized, double-blind study of epacadostat plus pembrolizumab vs pembrolizumab as first-line therapy for metastatic non-small cell lung cancer (mNSCLC) expressing high PD-L1 levels (ECHO-305/KEYNOTE-654) (ID 414)

      12:50 - 12:50  |  Presenting Author(s): L. Paz-Ares  |  Author(s): S. Rubin, Y. Zhao, L. Xu, A. Samkari, M. Awad

      • Abstract

      Background:
      Epacadostat (E) is a potent, highly selective oral inhibitor of indoleamine 2,3 dioxygenase-1 (IDO1), a tryptophan-catabolizing enzyme that shifts the tumor microenvironment to an immunosuppressive state responsible for tumor escape from immune surveillance. Pembrolizumab (P), a PD-1 inhibitor, is the standard of care for treatment-naive patients with mNSCLC, high PD-L1 expression (tumor proportion score [TPS] ≥50%), and no EGFR or ALK genomic aberrations. Preliminary data from the phase 1/2 ECHO-202/KEYNOTE-037 study suggest that E + P is generally well tolerated and active in NSCLC regardless of PD-L1 expression. The ECHO-305/KEYNOTE-654 global study compares the efficacy and safety of first-line E + P vs P in patients with PD-L1 high mNSCLC.

      Trial design:
      Eligible patients are ≥18 years old with stage IV NSCLC for whom EGFR-, ALK-, or ROS1-directed therapy is not indicated, TPS ≥50%, ECOG PS 0 or 1, and no prior systemic therapy for mNSCLC. Patients who received prior IDO1 inhibitors or immune checkpoint therapies are excluded. Approximately 588 patients will be randomized 1:1 to E 100 mg oral BID + P 200 mg IV Q3W or P + E-matched placebo. Patients will be stratified by tumor histology (squamous vs nonsquamous), ECOG PS (0 vs 1), and geographical location (East Asia vs non-East Asia). Treatment will continue for up to 35 cycles of P or until disease progression, intolerable toxicity, or investigator or patient decision to withdraw. Treatment may continue beyond initial radiographic progression in eligible patients. Patients may discontinue treatment following confirmed complete response. The primary endpoints are PFS and OS. Secondary endpoints include ORR, duration of response, safety, tolerability, and E pharmacokinetics. Response will be assessed by RECIST v1.1 (blinded independent central review) every 9 weeks up to week 54 and every 12 weeks thereafter. Adverse events (AEs) will be monitored throughout the study and for 30 days (90 days for serious AEs) after end of treatment and graded per CTCAE v4.0.

      Clinical trial identification:
      NCT03322540

      Legal entity responsible for the study:
      Incyte Corporation, Wilmington, DE, USA and Merck & Co., Inc., Kenilworth, NJ, USA

      Funding:
      Incyte Corporation, Wilmington, DE, USA and Merck & Co., Inc., Kenilworth, NJ, USA

      Disclosure:
      L. Paz-Ares: Honoraria from AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Lilly, MSD, Pfizer, Roche, Merck and Novartis. S. Rubin, Y. Zhao: Employee and stockholder of Incyte. L. Xu, A. Samkari: Employee and stockholder of Merck. M. Awad: Consulting or advisory role for Abbvie, ARIAD, AstraZeneca/MedImmune, Boehringer Ingelheim, Bristol-Myers Squibb, Clovis Oncology, Foundation Medicine, Genentech, Merck, Nektar, and Pfizer.

    • +

      196TiP - CheckMate 384: A phase 3b/4 dose-frequency optimization trial of nivolumab in advanced or metastatic NSCLC (Now Available) (ID 248)

      12:50 - 12:50  |  Presenting Author(s): E. Garon  |  Author(s): N. Reinmuth, R. Harris, P. Mitchell, J. Zhu, G. Selvaggi, S. Agrawal, E. Pichon

      • Abstract
      • Slides

      Background:
      Nivolumab, an anti-programmed death-1 antibody, is approved for previously treated metastatic non-small cell lung cancer (NSCLC) and various other cancers. Based on efficacy and safety results, the approved dosing frequency is every 2 weeks (Q2W) until progression or discontinuation due to toxicity. CheckMate 153 evaluated nivolumab treatment (tx) duration and demonstrated improved outcomes with continuous tx; 1-year progression-free survival (PFS) rates were higher in patients who continued vs stopped tx after 1 year. As an alternative to stopping tx, reducing dosing frequency could optimize convenience during long-term nivolumab therapy while maintaining efficacy and safety; recent exposure-response modeling predicted the benefit–risk profile of nivolumab 480 mg every 4 weeks (Q4W) to be similar to 3 mg/kg Q2W. CheckMate 384, a randomized, open-label phase 3b/4 trial was designed to evaluate less frequent nivolumab dosing (480 mg Q4W vs 240 mg Q2W) in patients with advanced/metastatic NSCLC and prior nivolumab tx.

      Trial design:
      Eligible patients are adults with advanced/metastatic squamous or non-squamous NSCLC and Eastern Cooperative Oncology Group performance status 0–2 who received prior nivolumab 3 mg/kg Q2W or 240 mg Q4W for up to 12 months with a complete or partial response or stable disease confirmed by 2 consecutive assessments. Patients with untreated, symptomatic central nervous system metastases are not eligible. Patients are randomized 1:1 to receive intravenous nivolumab 240 mg Q2W or 480 mg Q4W. Randomization is stratified by histology and response to pre-study nivolumab tx (complete/partial response vs stable disease). Endpoints are shown in the Table. The primary objective is to compare 6-month and 1-year PFS rates between patients who received nivolumab 480 mg Q4W and those who received 240 mg Q2W. Planned enrollment is 620 patients.Table:Study endpoints

      PrimarySecondary
      PFS rates at 6 months and 1 year after randomization (co-primary)PFS rate at 1 year after randomization by tumor histology and by response to pre-study nivolumab before randomization
      PFS rate at 2 years after randomization
      Overall survival rate
      Safety and tolerability, as assessed by incidence and severity of adverse events
      Clinical trial identification: NCT02713867; First posted: March 21, 2016

      Clinical trial identification:
      NCT02713867; First posted: March 21, 2016

      Legal entity responsible for the study:
      Bristol-Myers Squibb

      Funding:
      Bristol-Myers Squibb

      Disclosure:
      E. Garon: Research funding from AstraZeneca, Bristol-Myers Squibb, Eli Lilly, Genentech, Merck, Novartis, and Pfizer. N. Reinmuth: Speaker and consulting honoraria and fees from Amgen, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Hoffmann-La Roche, Lilly, MSD, Novartis, and Pfizer. R. Harris: Consultant for Bristol-Myers Squibb. P. Mitchell: Advisory boards for AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, MSD, Merck Serono, and Roche; Received research funding from Merck Serono. Received honoraria and travel grants from Roche. J. Zhu, G. Selvaggi, S. Agrawal: Employee and shareholder of Bristol-Myers Squibb. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      197TiP - A phase 1/2 trial of selinexor combined with docetaxel for previously treated, advanced KRAS mutant non-small cell lung cancer (ID 244)

      12:50 - 12:50  |  Presenting Author(s): A. Gupta  |  Author(s): H. West, D..R. Camidge, T.E. Stinchcombe, L. Horn, J. Saltarski, J. Bolluyt, M.A. White, D.E. Gerber

      • Abstract

      Background:
      Selinexor is an oral, reversible inhibitor of exportin-1 (XPO1), the major nuclear export protein responsible for nucleocytoplasmic transport of multiple tumor suppressor proteins and cell cycle regulators. Upregulation of XPO1 results in tumorigenesis and inactivation of apoptosis. Pharmacologic targeting of this process, termed selective inhibition of nuclear export (SINE), has demonstrated anti-tumor efficacy in preclinical and clinical trials. The most extensively studied SINE to date, selinexor (KPT-330; Karyopharm Therapeutics, Inc., Newton, MA), has demonstrated single-agent anticancer activity and synergistic effects in combination regimens against multiple cancer types. SINE may have particular relevance in KRAS-driven tumors, for which this treatment strategy demonstrates significant synthetic lethality. Thus, we designed a phase 1/2 study to assess the safety and tolerability of selinexor in combination with docetaxel in patients with previously treated, advanced KRAS mutant non-small cell lung cancer (NSCLC).

      Trial design:
      This is a multicenter, single-arm, open-label, non-blinded trial. Eligible patients are aged ≥18 years with advanced NSCLC and by central testing documenting presence of KRAS mutation (codons 12, 13, or 61). We will enroll 9–18 patients in the dose escalation cohort and 35 patients in the expansion cohort at five institutions with UT Southwestern as the primary coordinating center for this study. Dose escalation will be performed in a traditional 3 + 3 design. Patients will receive selinexor orally once weekly, with a lead in period of 1 week before chemotherapy initiation, in combination with standard doses of docetaxel given once every 3 weeks. Treatment will be administered in 21-day cycles. Dose limiting toxicities will be assessed based on the first cycle (7-day lead-in + 21-day cycle = 28 day) toxicity. The primary objective is to determine the safety and the maximum tolerated dose and recommended Phase 2 dose of selinexor administered with the standard dose of docetaxel. The sample size estimate is based on the exact one-sample binomial test for proportions. Enrollment will open in January 2018.

      Clinical trial identification:
      NCT03095612

      Legal entity responsible for the study:
      University of Texas Southwestern Medical Center, Dallas, TX, USA

      Funding:
      Karyopharm Therapeutics, Inc., Newton, MA

      Disclosure:
      L. Horn: Has consulting for Abbvie, Astra Zeneca, Lilly, BMS, Merck, Roche-Genentech, Xcovery. D.E. Gerber: Research funding from Karyopharm. All other authors have declared no conflicts of interest.

    • +

      198TiP - Afatinib plus EGF pathway targeting immunization (EGF-PTI) as first line therapy for EGFR mutant NSCLC: The EPICAL study (Now Available) (ID 332)

      12:50 - 12:50  |  Presenting Author(s): N. Karachaliou  |  Author(s): A. Cardona Zorrilla, D. Rodríguez-Abreu, M. Cobo Dols, N. Reguart Aransay, S. Viteri, J. Codony-Servat, M.A. Molina-Vila, E. D'Hondt, R. Rosell

      • Abstract
      • Slides

      Background:
      EGF-PTI elicits an antibody response against EGF and suppresses EGFR signaling. It is a vaccine with autologous humanized recombinant EGF conjugated with recombinant P64K protein of Neisseria meningitides. We previously showed signal transducer and activator of transcription 3 (STAT3) activation as a mechanism of innate resistance to EGFR tyrosine kinase inhibitors (TKIs) in EGFR mutant (EGFR+) NSCLC1,2. Anti-EGF antibodies block STAT3 activation in EGFR+ cells alone and in combination with EGFR TKIs. They also delay the emergence of resistance to afatinib in vitro3. Sera from patients immunized with EGF-PTI block the EGF-induced EGFR and ERK phosphorylation in serum starved cells. Based on the above data we designed the first phase Ib study to evaluate the safety and efficacy of afatinib plus EGF-PTI as 1st line therapy for metastatic EGFR+ NSCLC patients (NTC….).

      Trial design:
      This is a multicenter open-label exploratory phase Ib clinical study in Spain and Colombia. Thirty metastatic EGFR+ NSCLC patients will receive treatment with afatinib 40 mg daily plus EGF PTI. EGF PTI will be administered (4 injections, 4.8 mL) on day 1, 14, 28, 43 and 92 (immunization phase) and every 3 months (2 injections, 2.4 mL) (maintenance phase). Treatment will continue until disease progression, intolerable adverse events, consent withdrawal or noncompliance with the study protocol. The primary objective of the study is to evaluate the safety and tolerability of the combination. Secondary objectives include overall response rate, progression free survival, overall survival, time to treatment failure, duration of response and disease control rate. A pre-specified secondary objective of the study is to unravel the molecular mechanisms underlying the potential efficacy of afatinib plus EGF-PTI. To this end, EGFR, ERK, AKT and STAT3 phosphorylation levels will be longitudinally evaluated in EGF-stimulated serum starved cells treated with sera from the patients. Longitudinal blood analyses of EGFR mutations and the expression of a panel of possible predictive biomarkers in the baseline tissue will be also performed.

      Clinical trial identification:
      EudraCT number: 2017-003237-28

      Legal entity responsible for the study:
      Pangaea Oncology.

      Funding:
      Bioven

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      199TiP - TTFields concurrent with standard of care for stage 4 non-small cell lung cancer (NSCLC) following platinum failure: Phase 3 LUNAR study (Now Available) (ID 523)

      12:50 - 12:50  |  Presenting Author(s): U. Weinberg  |  Author(s): O. Farber, M. Giladi, Z. Bomzon, E. Kirson

      • Abstract
      • Slides

      Background:
      Tumor Treating Fields (TTFields) is a non-invasive, anti-mitotic treatment that disrupt the formation of the mitotic spindle and dislocation of intracellular constituents. TTFields plus temozolomide significantly extend survival in newly diagnosed glioblastoma. Efficacy of TTFields in NSCLC has been shown preclinically and their safety in a phase I/II pilot study with pemetrexed. We hypothesize that adding TTFields to immune checkpoint inhibitor or docetaxel following platinum doublet failure will increase OS.

      Trial design:
      Patients (N = 534), with squamous or non-squamous NSCLC, are stratified by their selected standard therapy (immune checkpoint inhibitors or docetaxel), histology (squamous vs. non-squamous) and geographical region. Key inclusion criteria are disease progression while on or after platinum-based therapy, ECOG 0-2, no electronic medical devices in the upper torso, and absence of brain metastasis. Docetaxel or immune checkpoint inhibitors are given at standard doses. TTFields are applied to the upper torso for at >18 hours/day, allowing patients to maintain daily activities. TTFields are continued until progression in the thorax and/or liver. Follow up is performed once q6 weeks, including CT scans of the chest and abdomen. On progression in the thorax and/or liver, patients have 3 post-progression follow up visits and are then followed monthly for survival. The primary endpoint is superiority in OS between patients treated with TTFields in combination with the standard of care treatments versus (vs) standard of care treatments alone. Key secondary endpoints compare the OS in patients treated with TTFields and docetaxel vs docetaxel alone, and patients treated with TTFields and immune checkpoint inhibitors vs those treated with immune checkpoint inhibitors alone. An exploratory analysis will test non-inferiority of TTFields with docetaxel compared to checkpoint inhibitors alone. Secondary endpoints include progression-free survival, radiological response rate, quality of life based on the EORTC QLQ C30 questionnaire and severity & frequency of adverse events. The sample size is powered to detect a HR of 0.75 in TTFields-treated patients versus control group.

      Clinical trial identification:
      NCT02973789

      Legal entity responsible for the study:
      Novocure

      Funding:
      Novocure

      Disclosure:
      U. Weinberg, O. Farber, M. Giladi, Z. Bomzon, E. Kirson: Novocure employee

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      201P - Isolation of circulating tumor cells and potential heterogeneity analysis in advanced non-small cell lung cancer (ID 148)

      12:50 - 12:50  |  Presenting Author(s): H. Zhang  |  Author(s): Q. Zhang, J. Nong, J. Wang, Z. Yan, L. Yi, X. Cheng, Z. Liu, S. Zhang

      • Abstract

      Background:
      Liquid biopsy as a noninvasive testing method is attractive not only for evaluation of prognosis but also for detection of the molecular drivers of tumors and specific DNA mutations that predict a response or resistance to targeted agents in the era of personalized medicine. A tumor biopsy depicts only a single site, which might be inadequate for characterization of the tumor because of intratumoral and intermetastatic heterogeneity. Circulating tumor DNA offers a “real-time” tool for serial monitoring of tumor genomes by providing accessible genetic biomarkers for cancer diagnosis, prognosis, and response to therapy; thus, circulating tumor DNA has progressively become routinely used in clinical oncology, Another type of liquid biopsy focuses on analysis of, while circulating tumor cells (CTCs) which is potentially more practical for obtaining genome-wide information and facilitating heterogeneity analysis.

      Methods:
      The CellSearch System is the only validated method for CTC detection that has been approved by the Food and Drug Administration. Based on this principle, we enrolled 60 patients with newly diagnosed and locally advanced or metastatic non-small cell lung cancer, CTCs were isolated by micromanipulation followed by whole-genome amplification, then analyzed by quantitative PCR for epidermal growth factor receptor (EGFR) mutation.

      Results:
      In 63% of these patients ≥1 CTC was isolated (median, 2 CTCs; range, 0–16 cells per 4.5 ml of blood). We then investigated whether the captured CTCs were suitable for molecular characterization. In five of six patients, the amplified CTC genomic DNA was further analyzed that proved with copy number variation. Three of these patients (1,3, and 5 CTCs, respectively) in whom the mutational status of the primary tumor was known showed specific EGFR mutations possibly identical to those of the primary tumor, while only one showed exactly the same mutation and the others showed clearly different mutations from the primary tumor.

      Conclusions:
      These preliminary results suggest that the use of CTCs may be feasible for heterogeneity analysis of tumor mutations.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hongtao Zhang, Shucai Zhang

      Funding:
      Capital features – Multicentre clinical study of Ectipitidine Hydrochloride in the Treatment of Advanced Lung Cancer with EGFR21 Exon Sensitive Mutation(Z161100000516107) Beijing Municipal Science and Technology Commission (Z151100004015104)

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      202P - The clone evolutionary landscape and genomic characteristics of osteosarcoma and lung metastasis (Now Available) (ID 204)

      12:50 - 12:50  |  Presenting Author(s): J. Lai  |  Author(s): C. Xu, W. Wang, Z. Song, Y. Chen, Y. Zhu, S. Cai, G. Chen, T. Lv, Y. Song

      • Abstract
      • Slides

      Background:
      Tumor metastasis is still the main cause of cancer related mortality, so it is important to identify the key molecules in each step of tumor metastasis and develop new strategies for prevention and control of tumor metastasis. Osteosarcoma (OS) is a primary malignant bone tumor that has a high potential to metastasize to lungs. Few consistent clinically actionable mutations have been reported and no further improvements have been made in the last decades regarding survival. Therefore, biomarkers prognosticating for overall survival or the development of lung metastases in patients with OS may improve personalized care. The study of our aim is to investigate harbor distinct genetic alterations beyond those observed in primary tumors.

      Methods:
      Next-generation sequencing (NGS)-based 381 genes panel assay were performed on ten patients with primary OS and matched lung metastatic tumors. A set of high confident SNV, small insertion and indel and CNV in each sample were identified.

      Results:
      There were diversified metastatic progression during lung metastasis of OS including parallel evolution (6/10) and linear evolution (4/10), and metastasis-to-metastasis spread was also found in two patients with multiple metastasis. Multiple novel significantly mutated genes were identified, including CREBBP, LRP1B, MAP3K1 and LRP1B in lung metastases, SETD2, GNAS, and H3F3A in primary tumors, and CDKN2A in both. Copy number analysis indicated recurrent CNAs, including NFKBIA gain, MCL1 gain, and MYC gain in lung metastases, AMER1 loss, CTNNB1 gain in primary tumors, and CDKN2B gene family loss in both. Furthermore, phylogenetic analyses revealed that paired primary tumors and metastases underwent parallel evolution with few ubiquitous clonal mutations, suggesting that OS metastases are likely to be derived from primary tumors at a very early stage of their evolution.

      Conclusions:
      The evolution of primary and metastatic tumors were very complex. Our findings strongly support a parallel evolution model of primary and metastatic tumors. Moreover, the much higher mutation load and significantly mutated genes that are specifically associated with lung metastases may provide immune checkpoint inhibitor and target therapeutic insight for OS.

      Clinical trial identification:


      Legal entity responsible for the study:
      Jinhuo Lai

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      203P - Combined pulmonary metastasectomy and chemotherapy in patients with colorectal lung metastases and concurrent extrapulmonary disease (ID 282)

      12:50 - 12:50  |  Presenting Author(s): M. Faisal  |  Author(s): I. Uthman, A. Abo Bakr

      • Abstract

      Background:
      Lung metastases from colorectal cancer (CRC) with the presence of concurrent extra-pulmonary disease is considered a very poor prognosis. Chemotherapy was the only advised treatment. Nowadays with the evolution in surgical oncology techniques, imaging and medical oncology treatment, a better management is present for stage IV colorectal cancer with a prolonged overall survival. We aim to evaluate the benefit of adding surgical resection to chemotherapy in patients with colorectal lung metastasis and extra-pulmonary diseases.

      Methods:
      We extracted the data of 1423 colorectal cancer patients with pulmonary and extra-pulmonary metastases from 2010 to 2013, using SEER database. Ninety-nine (7%) patients had combined chemotherapy and pulmonary metastasectomy, while 1324 (93%) of them received chemotherapy only.

      Results:
      Patients who received combination of chemotherapy and pulmonary metastasectomy had better 1-year survival rates (83.5%), compared to those who received chemotherapy only (68.3%). We observed a statistically significant difference between the two modalities in white patients, patients aged between 40 to 59 years, tumors arising in sigmoid colon or rectum, patients with elevated CEA levels, and those with metastasis to the lungs only or both lung and liver.Table: (Abstract: 203P)

      VariablesCombined TherapyChemotherapyp value (difference in modalitiesp value (difference in combined therapy)
      Sex
       Male79.4%66.6%0.007**0.399
       Female87.1%70.6%<0.001**
      Age
       20–3966.7%77.8%0.2190.201
       40–5989.7%74.2%<0.001**
       60–7973.0%66.2%0.159
       ≥80100%43.2%0.221
      Race
       White86.7%69.3%<0.001**0.687
       Black74.3%65.4%0.061
       Others77.9%66.1%0.036**
      Primary Site
       Cecum76.1%69.0%0.7500.003*
       Ascending Colon70.0%58.0%0.401
       Hepatic0%55.1%0.361
       Flexure Transverse Colon100%55.5%0.605
       Splenic Flexure60.3%59.2%0.937
       Descending Colon75.3%64.3%0.068
       Sigmoid Colon85.5%68.5%0.002**
       Rectum100%74%<0.001**
      Distant Metastasis
       Lung Only94.5%78.9%0.002**0.007*
       Lung and Liver77.4%64.7%0.002**
       Lung and Brain100%25.3%0.198
       Lung and Bone50.8%65.9%0.783
      CEA Level
       Normal95.7%81.4%0.3780.446
       Elevated79.9%66.2%<0.001**


      Conclusions:
      Combining pulmonary metastasectomy with chemotherapy in CRC patients with pulmonary and extrapulmonary metastases shows higher 1-year relative survival rates when compared to using chemotherapy alone.

      Clinical trial identification:


      Legal entity responsible for the study:
      Inas Uthman

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      204P - Clinicopathological features, treatment patterns and outcomes of pulmonary metastasectomy (Now Available) (ID 546)

      12:50 - 12:50  |  Presenting Author(s): P. Shivanna  |  Author(s): S. Deo, N.K. Shukla, S. Bhoriwal, R. Thakur, S. Bakshi, R. Palaniappan, S. Kumar

      • Abstract
      • Slides

      Background:
      Lung is the most common metastatic site, accounting up to 80% of metastases in solid tumors. Although the presence of metastases in patients with malignant tumors is a sign of advanced systemic disease and is considered as non-curative, recent trials have shown survival benefit with pulmonary metastasectomy in selected group of patients. The reason behind some tumors metastasizing exclusively to the lung is unknown. Surgical resection of lung metastasis may significantly impact the long-term outcome in this subset of carefully selected patients. In this study we analyze the results of pulmonary metastasectomy.

      Methods:
      A prospectively maintained database in the Department of Surgical Oncology, BRA IRCH, AIIMS, New Delhi, India from 1995 to 2017 was accessed and the data of all patients undergoing pulmonary metastasectomy for various primary tumors were analyzed. Clinicopathologic, epidemiological factors, surgical details, post metastasectomy survival and prognostic factors were analyzed.

      Results:
      A total of 139 patients underwent pulmonary metastasectomy from 1995 to 2017. A total of 195 surgical procedures were carried out for pulmonary metastases, which included surgeries for recurrent metastatic diseases. Patients were predominantly male (M: F = 106:33) and belonged to various age groups ranging from 11years to 68 years (mean = 30 years). Lower limb was the most common site of primary and bone osteosarcoma was the predominant histology of primary tumour (46%), followed by soft tissue sarcomas. Majority of patients underwent open metastasectomy of whom 97% underwent R0 resection. Largest tumor size was 7cms. There were no perioperative mortality. Nineteen patients developed pulmonary recurrence requiring repeat metastasectomy.

      Conclusions:
      Pulmonary metastasis is a form of systemic failure in many solid tumors. Vast majority end up in palliation. However, in a small subset of patients, pulmonary metastasis are suitable for surgical resection. Pulmonary metastasectomy in combination with or without systemic therapy plays a role in prolonging survival in this subset of patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      All India Institute of Medical Sciences

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      205P - RNA-seq analysis of lung adenocarcinomas reveals different circular RNA expression profiles between non-metastatic and metastatic patients (Now Available) (ID 488)

      12:50 - 12:50  |  Presenting Author(s): D. Wu  |  Author(s): X. Qi, B. Qin

      • Abstract
      • Slides

      Background:
      The identification of cancer-associated circular RNAs (circRNAs) and investigation of their molecular and biological functions are important to understand the molecular tumorigenesis and progression in cancer devolpment. However, their role in non-small cell lung cancer (NSCLC) is unknown. We aimed to examine the expression profile of circRNAs in NSCLC and to evaluate its biological role and clinical significance in tumor progression.

      Methods:
      Six patients with lung adenocarcinomas were included and divided into non-metastatic group (n = 3) and metastatic group (n = 3). The gene expression changes were examined. GO and KEGG Pathway enrichment analysis were performed for the host genes of the differentially expressed circRNAs. CircRNA-miRNA interaction were predicted by popular target prediction software, and network was constructed by Cytoscape software. We also selected 5 genes for further verification by real-time quantitative Polymerase Chain Reaction.

      Results:
      The circRNAs chip detected more than 30,000 circRNAs expressed in peripheral blood samples from the six lung adenocarcinomas patients. And about 200 circRNAs were ≥2-fold differentially expressed between non-metastatic group and metastatic group, of which 174 circRNAs were up-regulated and 3 were down-regulated in the metastatic group compared with non-metastatic group. In addition, we conformed and verified the transcription level of 5 abnormally up-regulated circRNAs by RT-qPCR. Moreover, bioinformatics analysis demonstrated the biological processes of Fc gamma R-mediated phagocytosis, vascular endothelial growth factor signaling pathway and endocrine resistance. We also selected the top 10 significant Gene Ontology terms and selected pathways for a brief summary.

      Conclusions:
      We performed a global analysis of expression profile of circRNAs in metastatic lung adenocarcinoma. Using bioinformatics analysis, our study has reveled and identified the related circRNAs involved in tumorigenesis and metastasis process. Our results may stimulate a deeper understanding of the disease, and lead to the development of potential therapies and the identification of novel biomarkers.

      Clinical trial identification:


      Legal entity responsible for the study:
      Wu Dan, Department of Oncology, The Fifth Hospital of Xiamen, China

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      206P - Isolated splenic metastasis from non-small cell lung cancer: Study of an extremely rare clinical entity with review of all cases reported in the literature (ID 515)

      12:50 - 12:50  |  Presenting Author(s): N. Mitsimponas  |  Author(s): M. Mitsogianni, A. Giagounidis

      • Abstract

      Background:
      Rarely, non-small cell lung cancer metastases have been reported in soft tissue, kidney, peritoneum, spleen, pancreas, intestine, bone marrow, eye, ovary, thyroid, heart, breast, nasal cavity and tonsils. As an isolated event, splenic metastasis from non-small cell lung cancer is exceedingly rare. Until now, only 29 cases have been reported in the medical literature.

      Methods:
      We have reviewed the medical literature concerning this extremely rare clinical entity and we have found 29 cases. We have studied these cases concerning different parameters like age, gender, histology and side of primary lung lesion, time to splenic metastasis, symptoms of metastasis and treatment of primary tumour and treatment of splenic metastasis.

      Results:
      In these cases, was recorded a strong male predominance (58%), ranging in age from 49 to 82 (mean. 62,3 years). In most of the reports the primary tumor was located in the right lung. The most common histopathological subtype of lung cancer with isolated splenic metastasis was adenocarcinoma (44% of cases), following from the squamous cell cancer (17% of cases) and large cell lung cancer (17% of cases). Splenic metastasis was synchronous in 12 cases and metachronous in other 17 cases. In the 17 cases of metachronous isolated splenic metastasis the median interval between the diagnosis of primary tumor and isolated splenic metastasis was 22,2 months (range 2 to 96 months). The majority of cases were asymptomatic (62% of cases) and the diagnosis was serendipitously made at follow up exams. Some patients presented with splenic rupture (12% of cases), abdominal pain (21% of cases) and fever (3% of cases). The majority of patients underwent a surgical resection of the primary tumor (65% of cases). In the most cases, the patients underwent a splenectomy (83% of cases). The vast majority of patients died 1 to 49 months after splenectomy.

      Conclusions:
      The appearance of this rare clinical entity ranges in age, symptoms and histological type of tumour. The management of this entity in the same way with an oligometastatic NSCLC (splenectomy), whenever feasible, seems to be the most appropriate treatment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Nikolaos Mitsimponas

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      207P - A comparison of outcomes of stereotactic body radiotherapy versus metastasectomy in patients with pulmonary metastases (ID 541)

      12:50 - 12:50  |  Presenting Author(s): Y.H. Lee  |  Author(s): K.M. Kang, H. Choi, I.B. Ha, H. Jeong, J.H. Song, B.K. Jeong

      • Abstract

      Background:
      Since the concept of oligometastases was proposed, several investigators have suggested that aggressive local treatment of oligometastatic disease may be helpful in improving patient survival. However, optimal local therapy is still not clear it is difficult to compare the treatment outcomes of SBRT vs. metastasectomy in prospective randomized trials. Hence, the aim of this study was to compare the efficacy of SBRT and metastasectomy for the treatment of pulmonary metastases. Additionally, we sought to identify patients who are most likely to benefit from local treatment.

      Methods:
      Total 51 patients (median age, 65 years) were included. Twenty-one patients received SBRT using the Cyberknife system (total radiation doses 48–60 Gy delivered in 3–5 equal fractions) and 30 underwent metastasectomy, most (93.3%) with wedge resection. The median follow-up duration was 13.7 months. Local control rate, progression free survival (PFS), and overall survival (OS) were assessed.

      Results:
      The median tumor size was larger in the SBRT group (2.5 cm) than in the metastasectomy group (1.25 cm; P = 0.015). Other synchronous metastases were prevalent in the SBRT group (57.1% vs. 20%; P = 0.006). The local control rate was similar in both groups (P = 0.163). PFS was longer in patients who underwent metastasectomy than in those who received SBRT (P = 0.02), with 1- and 2-year PFS rates of 51.1% and 46% in the former vs. 23.8% and 11.9% in the latter, respectively. The corresponding OS rates were 95% and 81.8% vs. 79.5% and 68.2%, respectively (P = 0.534). On multivariate analysis, synchronous metastasis was associated with poor PFS, and tumor size was the most significant factor influencing OS. There were no differences in PFS and OS between the metastasectomy and SBRT groups after dividing patients according to the presence or absence of synchronous metastases.

      Conclusions:
      SBRT and metastasectomy for pulmonary metastases produce similar local control and OS outcomes. SBRT is effective and has a broader indication than metastasectomy; however, patients with synchronous metastases are less likely to benefit from local treatment, whether via SBRT or surgery.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      208P - Metastatic non-small cell lung carcinoma: About 420 cases (ID 639)

      12:50 - 12:50  |  Presenting Author(s): H. Loubna

      • Abstract

      Background:
      Non-small cell lung carcinoma NSCLC is the leading cause of cancer death in the world. Its incidence is closely related to smoking. The prognosis of metastatic NSCLC remains poor despite major therapeutic advances.

      Methods:
      This is a descriptive retrospective study of patients treated for metastatic non-small cell lung carcinoma in the medical oncology department of HASSAN II University Hospital of Fez, over a period of 10 years, from January 2007 to July 2017.

      Results:
      420 cases have been identified. The average age of our patients was 58.63 years old [31–86]. The male sex was dominant with a percentage of 86%. Smoking was found in 76.1% of patients while occupational exposure to a risk factor was reported in 4% of cases. The disease is declared by cough in 45% of cases. Adenocarcinoma represents 47.1% of all histological subtypes. Most patients (62%) had a performance status that exceeded 1. The most frequent metastatic site was lung (45% of cases) followed by bone (28% of cases). 66% of patients received platinum based first line chemotherapy associated with Bevacizumab in non-squamous cell carcinomas. 30% of cases who had an EGFR mutation research had a mutation and received Erotinib or Afatinib. None of the patients was tested for ALK rearrangement or PDL 1 overexpression. 19% of cases received second-line systemic treatment with Docetaxel in 79.7% of cases, Pemetrexed in 17.5% and Erlotinib in 2.7% of cases. The overall response rate after first treatment line was 42%: 4% partial response, 38% stability and 56% progression. Whereas 75% of patients with tyrosine kinase inhibitors responded. After a median follow-up of 48 months, the median SSP and SG were 5.3 months and 11.2 months, respectively.

      Conclusions:
      The results of our series confirm the poor prognosis of metastatic NSCLC and emphasizes the need of implementing early diagnosis strategies and smoking quit in our country

      Clinical trial identification:
      Background : Non-small cell lung carcinoma NSCLC is the leading cause of cancer death in the world. Its incidence is closely related to smoking. The prognosis of metastatic NSCLC remains poor despite major therapeutic advances. Method : This is a descriptive retrospective study of patients treated for metastatic non-small cell lung carcinoma in the medical oncology department of HASSAN II University Hospital of Fez , over a period of 10 years, from January 2007 to July 2017. Results : 420 cases have been identified. The average age of our patients was 58.63 years old [31-86]. The male sex was dominant with a percentage of 86%. Smoking was found in 76.1% of patients while occupational exposure to a risk factor was reported in 4% of cases. The disease is declared by cough in 45% of cases. Adenocarcinoma represents 47.1% of all histological subtypes. Most patients (62%) had a performance status that exceeded 1. The most frequent metastatic site was lung (45% of cases) followed by bone (28% of cases). 66% of patients received platinum based first line chemotherapy associated with Bevacizumab in non squamous cell carcinomas. 30 % of cases who had an EGFR mutation research had a mutation and received Erotinib or Afatinib. None of the patients was tested for ALK rearrangement or PDL 1 overexpression. 19% of cases received second-line systemic treatment with Docetaxel in 79.7% of cases, Pemetrexed in 17.5% and Erlotinib in 2.7% of cases. The overall response rate with chemotherapy was … Whereas 75% of patients with tyrosine kinase inhibitors responded. After a median follow-up of 48 months, the median SSP and SG were 5.3 months and 11.2 months, respectively. Conclusion : The results of our series confirm the poor prognosis of metastatic NSCLC and emphasizes the need of implementing early diagnosis strategies and smoking quit in our country

      Legal entity responsible for the study:
      Department of Medical Oncology, HASSAN II University Hospital-Fez

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      209P - The management of brain disease (BD) in ALK-rearranged non-small cell lung cancer (ALK NSCLC): Can systemic treatment delay the initiation of local treatments? Experience of the Ramón y Cajal University Hospital (RyCUH, Madrid) (Now Available) (ID 625)

      12:50 - 12:50  |  Presenting Author(s): R. Martin Huertas  |  Author(s): I. Ciscar Garcia, C. Saavedra Serrano, J.J. Serrano Domingo, E. Corral de la Fuente, J.J. Soto Castillo, A. Gomez Rueda, M.E. Olmedo Garcia, P. Garrido Lopez

      • Abstract
      • Slides

      Background:
      The arrival of oral tyrosine kinase inhibitors (TKI) to the treatment of ALK NSCLC is becoming a paradigm shift in the treatment of BD (brain metastases and meningeal carcinomatosis), given the intracranial activity thereof, with the possibility of delaying the start of local treatments, as radiotherapy (RT), radiosurgery or neurosurgery, and the comorbidity that they entail.

      Methods:
      Between October 2013 and October 2017 were registered patients diagnosed with ALK NSCLC at the RyCUH. Patients with BD were considered eligible, well at diagnostic of the primary tumor, well at progression of it. The systemic treatment carried out is collected as well as the local treatment at diagnosis of the BD if this is done. We studied systemic (PFSs) and brain progression free survival (PFSb) as well as overall survival (OS) according to the type of treatment and moment received. Other variables were included as neurological symptomatology and corticotherapy.

      Results:
      Of the 33 patients diagnosed of ALK NSCLC, 42.42% had BD at some point of their evolution (57.14% at diagnosis and 42.86% at progression). As a first-line systemic treatment, 57.14% received chemotherapy and 42.85% received a TKI, although 71.43% received a TKI as a second-line treatment. 57.14% of the patients received whole radiation therapy and 28.57% radiosurgery at some point during their evolution. PFSb is higher in those patients without brain metastases at diagnosis, who initiate systemic treatment, and who perform local treatment at brain progression (14 months), compared to patients who change systemic treatment for brain progression (12 months). The median OS is higher in patients who do not initially receive local treatment (25.5 months) versus those who do (21 months).

      Conclusions:
      There are multiple strategies in the management of ALK NSCLC BD. Systemic treatment with TKIs allows to delay local treatment with RT or surgery and comorbidity. However, the use of RT or surgery for exclusive brain progression with controlled systemic disease allows the maintenance of TKI and prolong the PFSs.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hospital Universitario Ramón y Cajal (Madrid)

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      210P - Brain oligometastases in NSCLC: Therapeutic effect and prognostic factors of WBRT combined with intracarotid infusion of elemene (ID 236)

      12:50 - 12:50  |  Presenting Author(s): Y. XU  |  Author(s): X. Lv, Y. Liu, B. Dong

      • Abstract

      Background:
      To retrospectively analyze the curative effect of WBRT combined with intracarotid infusion of elemene following intracranial local treatment in NSCLC patients with brain oligometastases and investigate the related factors affecting the prognosis of patients.

      Methods:
      Clinical data of patients admitted in the Affiliated Hospital of Dalian Medical University between Jan. 2000 and Dec. 2015 were collected. Inclusion criteria: (1) pathologically diagnosed as NSCLC; (2) brain metastasis confirmed by enhanced MR, with 1–3 lesions; (3) no metastases found in other parts of the body; (4) primary pulmonary lesions were effectively treated; (5) surgery or SRS were performed for intracranial lesions. Patents in the observation group were treated with WBRT alone with a dose of 40Gy/20f; and those in the study group were treated with the same WBRT scheme as the observation group, and 1000 mg elemene was infused daily for 15–20 days.

      Results:
      Forty-one patients had complete follow-up data. There were 22 males and 19 females, with the median age of 58 years; age >60 years in 17 cases and ≤60 in 24 cases; KPS score of 60 in 6 cases and KPS >60 in 35 cases. Followed the local treatment, 17 patients were treated with WBRT alone and 24 with WBRT combined with elemene. The median OS of all patients was 16.1 months and the median iPFS was 12 months. The 1- and 2- year survival rates were 68.2% and 24.4%, respectively. The median OS of the WBRT group and WBRT combined with elemene group were 14.8 and 18 months (χ2 = 2.328, P = 0.127) and the median iPFS was 10.3 and 13.8 months (χ2 = 4.010, P = 0.045), respectively. KPS > 60, single brain metastases, extracranial lesion control and the use of targeted therapy were the predictive factors for a good prognosis and multivariate analysis indicated that single brain metastases, extracranial focus control and targeted therapy were the independent prognostic factors for a favorable prognosis.

      Conclusions:
      WBRT combined with intracarotid infusion of elemene can prolong the median iPFS of NSCLC patients with brain oligometastases. Single brain metastasis, extracranial lesion control and targeted therapy are the independent prognostic factors for a favorable prognosis.

      Clinical trial identification:


      Legal entity responsible for the study:
      The First Affiliated Hospital of Dalian Medical University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      211P - First analysis of a real-world study to explore the character and treatment outcome of epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) on non-small cell lung cancer (NSCLC) with brain metastases (BM) (ID 528)

      12:50 - 12:50  |  Presenting Author(s): H. Zhang  |  Author(s): L. Liu, C. Zhang, N. Zhang, H. Su, Y. Song, J. Min

      • Abstract

      Background:
      Lung Cancer is the most common cancer in China. Brain metastasis(BM) is the main reason for treatment failure of advanced lung cancer and treatment strategy is unclear in real world practice. Recently, some data showed that EGFR-TKI was an efficient option for NSCLC BM patients. Further evidence on EGFR-TKI plus standard therapy in clinical practice is highly expected.

      Methods:
      Based on our clinical database, we collected the clinical data of patients diagnosed with NSCLC and BM during 2014 and 2017. The therapeutic effectiveness was compared between the chemoradiation and chemoradiation + EGFR-TKI. Log-rank test was used in the comparison of the two groups. Cox's proportional hazard models were used in the factor analyses.

      Results:
      Total 153 patients were analyzed in the study, 61 patients treated by chemoradiation and 92 patients by chemoradiation + EGFR-TKI. Baseline characters of age, location and T stage of primary disease, and metastatic numbers were balanced in two groups. Gefitinib is the most frequently used TKI in clinical practice, composing about 48%(44/92). In chemoradiation + EGFR-TKI, 72 patients were performed with EGFR gene test and of them, 56 patients (78%) were EGFR positive. In chemoradiation, 29 patients were performed with EGFR gene test and 2 patients (6.9%) were EGFR positive. EGFR positive rate was 57% (58/101) in the total patients. The OS trend obviously showed that patients got more advantage from chemoradiation + EGFR-TKI compared with chemoradiation. Median OS was 27.0 months in the chemoradiation + EGFR-TKI and 16.6 months in the chemoradiation. Subgroup 24 months survival rate analysis of Gefitinib vs other TKI is 0.732 and 0.427, p = 0.0124. This preliminary analysis indicated that adding TKI to chemoradiation treatment could improve OS (hazard ratio [HR] 1.887) on NSCLC BM patients.

      Conclusions:
      The first analysis presents the survival benefit of EGFR-TKI plus standard treatment on NSCLC BM patients. Further data from this real-world study will be generated as we continue recruiting more patients in near future.

      Clinical trial identification:


      Legal entity responsible for the study:
      Oncology Department, Tangdu Hospital affiliated with the PLA Airforce Medical University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      212P - Clinical experience and predictive factors of response to whole brain radiotherapy in metastatic lung cancer: A single institute experience (ID 462)

      12:50 - 12:50  |  Presenting Author(s): V.V. Pareek  |  Author(s): R. Bhalavat, M. Chandra

      • Abstract

      Background:
      Metastases in lung cancer especially to the brain is associated with poor outcomes and warrants palliative care in form of whole brain radiation therapy or best supportive care. Prognostic factors for overall survival are seen in many studies. However, there are few studies evaluating the role and predictive factors of whole brain radiation therapy (WBRT). The aim of this study was to identify the clinical outcomes and assess the predictive factors associated with treatment outcomes with WBRT in lung metastases.

      Methods:
      From 2009 to 2016, from medical records of Jupiter Hospital, 95 patients with histopathologically proven lung cancer and on imaging proven for solitary brain metastases, were evaluated retrospectively for demographic, treatment and clinical outcome parameters. Univariate and multivariate analysis were assessed for various impact of parameters in treatment outcomes age, Charlson comorbidity score, gender, marital status, use of chemotherapy, other metastatic sites, weight loss, BMI and edema and extent of edema/tumor ratio (E/T). T2 weighted and diffusion weighted images were assessed for the size comparison of E/T ratio.

      Results:
      Poor outcomes were related to small cell lung cancer (p = 0.006), E/T ratio > 1.5 (p < 0.001), median tumor diameter <2 cm was associated with better prognosis. Low Charlson comorbidity score, male gender, presence of extra-cranial metastases, weight loss >10% and higher BMI were associated with poorer outcomes. On subset analysis as per the risk factors, median survival was 5.6 months’ vs 9.2 months in poor risk factors vs the better prognostic factors. Also, raised LDH was found to be closesly associated as a prognostic factor for inferior outcomes. Patients with SCLC exhibited an improved RR compared with patients with NSCLC.

      Conclusions:
      This study evaluates predictive factors for response of BMs to WBRT in lung cancer. In cases where outcomes are poor, they can be considered for best supportive care and these require quality of life assessments to predict outcome comparison. The patients with better prognostic factors can also be looked in to for higher dose delivery with boost to solitary metastases for better outcomes and needs to be assessed.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      217P - Impact of VEGF and CD74 expression on response and survival of malignant pleural mesothelioma patients treated with gemcitabine and cisplatin: A phase II retrospective study (Now Available) (ID 565)

      12:50 - 12:50  |  Presenting Author(s): D.A. Soliman  |  Author(s): M.A.A. Ellithy, A.S. Saad, M.S. Jassim, H. Abou Gabal, W. Osman

      • Abstract
      • Slides

      Background:
      Malignant pleural mesothelioma is a fatal and aggressive disease; most patients present with advanced stages. Mesothelioma have one of the highest circulating serum VEGF levels of any solid tumor which is considered as a poor prognostic factor in this disease. Macrophage migration inhibitory factor (MIF) and its receptor CD74 found to be associated with angiogenic activity. Role of MIF has not been shown yet in MPM. This study targets the correlation between expression of VEGF and CD74 (on stored retrospective 44 paraffin embedded mesothelioma specimens) and tumor response to chemotherapy Gemcitabine and platinum combination. The study also figures out the prognostic impact of VEGF and CD74 on the progression free survival (PFS) and overall survival (OS) of the studied patients.

      Methods:
      Immunohistochemical staining for VEGF scored from 0 to 3 represent the percentage of cytoplasmic positively stained cells in the tumor. The CD74 expression was recorded in the tumor and the stroma semiquantitatively using the histoscore method with the final score resulting from the percentage of tumor cells staining positively multiplied by the staining intensity grade. Both CD74 and VEGF immunohistochemical markers were furtherly categorized in the statistical analyses as none (0)/low (1) vs medium (2)/high expression (3).

      Results:
      Combined med/high expression of CD74-TS and VEGF are significantly associated with poor response to gemcitabine and cisplatin chemotherapy (P = 0.03). VEGF expression level did not correlate with PFS (P = 0.09) while high CD74 (T) (P = 0.001) and (S) (P = 0.006) expression significantly impair PFS. Med/high VEGF expression is significantly associated with shorter OS (P = 0.025) whereas, med/high expression of CD74-TS yield a highly significant shorter OS. (P = 0.001).

      Conclusions:
      High expression of VEGF and CD74 T&S are inversely correlated with OS and response to chemotherapy with gemcitabine and cisplatin in Egyptian mesothelioma patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Clinical Oncology Department Ethical Committe

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      219P - Outcomes of malignant pleural mesothelioma (MPM) patients (p) treated after first line chemotherapy (CT) (Now Available) (ID 510)

      12:50 - 12:50  |  Presenting Author(s): S. Cedres Perez  |  Author(s): F. Amair, A. Martinez-Marti, A. Navarro, N. Pardo, J. Remon-Masip, J.M. Miquel, G. Villacampa, R. Dienstmann, E. Felip

      • Abstract
      • Slides

      Background:
      The increasing incidence and poor outcome associated with MPM requires identification of novel treatment options. Initial reports have demonstrated beneficial effects of pemetrexed, gemcitabine and vinorelbine in previously treated MPM, however there is no clear agreement on the role of different agents after first line. The aim of this study is to evaluate the outcomes of p with MPM treated in second and third lines at our institution.

      Methods:
      91 MPM p treated with CT or experimental agents in clinical trials beyond first line between September 2002 and October 2017 were retrospectively reviewed (49 p received a third line regimen). Survival was calculated using the Kaplan–Meier method and log-rank test was used for statistical comparison. The associations of type of regimen with outcomes were assessed with Cox proportional-hazard models.

      Results:
      Patient's characteristics: median age 65 years (29–84 years), males: 73%, performance status 1: 66%, asbestos exposure: 81%, stage III at diagnosis: 48%, epithelial subtype: 77%. All p were treated with chemotherapy in first line, 77% received cisplatin plus pemetrexed, 19% carboplatin plus pemetrexed and 4% others. Median PFS in first line was 5.1 months (m; CI95% 4.6–5.2). Median overall survival after first line was 11.6 m (9.8–15.9). Regimen offered as second- or third-line: platinum doublet in 24%, vinorelbine in 32%, gemcitabine in 10%, immunotherapy in 22%, and targeted agents in 12%. Median PFS in second or third line with platinum doublet was 4.9 m (4.3–5.9), vinorelbine 2.7 m (2.1–3.8), gemcitabine 3 m (1.4 – NA), immunotherapy 3 m (2.2–4.5) and targeted agents 3 m (0.8 – NA) (p > 0.05). In the third line setting, median survival after initiation of vinorelbine was 8.5 m (5.5 – NA) versus 3 m (3 – NA) for gemcitabine-treated p (p = 0.001).

      Conclusions:
      In our single institution series of previously treated MPM p, second or third line treatment produces modest benefit, with no clear differences in outcome for CT or clinical trial alternatives. Further research is necessary to treat p who failed to first line CT, including choice of therapy sequence in the second and third lines.

      Clinical trial identification:
      NA

      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      220P - Effects of pifithrin-μ on the growth of acidity-tolerant malignant mesothelioma cells and epithelial-mesenchymal transition (ID 264)

      12:50 - 12:50  |  Presenting Author(s): S. Lee  |  Author(s): Y. Lee

      • Abstract

      Background:
      Heat shock protein 70, which protects cells from various forms of cellular stress, has gained significant attention as a potential therapeutic target in various malignancies.

      Methods:
      Here, pifithrin-μ, an effective dual inhibitor of HSP70 and p53, was employed to examine its anticancer activities and to analyze its possible effect for epithelial-mesenchymal transition (EMT) in malignant mesothelioma cells.

      Results:
      MSTO-211HAcT cells, pre-adapted in medium containing lactic acid, showed more resistance to anticancer drugs, cisplatin and gemcitabine, when compared with their parental acid-sensitive MSTO-211H cells. Pifithrin-μ treatment induced cell death in a p53-independent manner and developed EMT-like phenomenon, which was characterized by an elongated cell morphology, a decrease in the levels of epithelial cell markers, including E-cadherin, claudin 1 and b-catenin, and an increase in the level of mesenchymal markers, including vimentin and Snail, and increased migratory and invasive properties in MSTO-211HAcT cells. Moreover, p53 knockdown significantly enhanced the pifithrin-μ-mediated changes of critical EMT markers, migration and invasion, and anoikis resistance.

      Conclusions:
      Collectively, pifithrin-μ may contribute to malignant progression by promoting the EMT, at least in part, through the p53 inhibition, despite its preferential growth-inhibiting and apoptosis-promoting effects on MSTO-211HAcT cells under acidic extracellular environment.

      Clinical trial identification:


      Legal entity responsible for the study:
      Soonchunhyang University

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      221P - Treatment experience of pleural malignant mesothelioma in a tertiary referral center in Mexico (ID 533)

      12:50 - 12:50  |  Presenting Author(s): A.M. Gutierrez  |  Author(s): G. Nuñez-Cerrillo, Y.A. Remolina-Bonilla, A. Armengol-Alonso, R. Trejo-Rosales

      • Abstract

      Background:
      Malignant pleural mesothelioma (MPM) is considered a relatively rare tumor and has an extremely poor prognosis. There is a lack of data in Mexico where asbestos industry still exists. Therefore, the creation of databases is necessary to determine the dimension of this problem in our country.

      Methods:
      A retrospective study was performed including patients with MPM diagnosis attended between 2012 to 2016 at Hospital de Oncología, Centro Médico Nacional Siglo XXI in Mexico City. The objective of our study was to describe clinical characteristics and oncologic outcomes. A survival analysis was conducted taking into account the different treatment arms and clinical stages.

      Results:
      A total of 99 patients diagnosed with MPM and complete clinical records were included. 67% were men and 33% women. Median age at diagnosis was 64 years, 40.4% had a history of asbestos exposure, 66% had performance status 0–1. Pleural effusion was present in 97%. Most of the patients had advanced disease (65% stage IV and 29% stage III). 14.1% underwent pleurodesis. In 3% radical pleurectomy was performed. 15% received only palliative care with median overall survival of 3 months. First line regimens were pemetrexed with platin compound in 36.5%, while no pemetrexed-contained regimens were administered in 63.5% (gemcitabine/platinum in 49%). Overall response was 15.5% and stable disease 50%. Median overall survival and progression-free survival were 10 (0–50) months and 4 (0–8) months respectively, with no difference between treatment groups (p = 0.38). 29% received second-line therapy, most frequently (48%) oral vinorelbine-based regimens with clinical benefit of 46% and median PFS 3 (0–16) months.

      Conclusions:
      To our best knowledge, this is the largest report about the treatment of advanced MPM in Mexico. Limited by its retrospective design, this study confirms better outcomes in patients treated with chemotherapy and also highlights oral vinorelbine activity in second-line setting. Prognosis of advanced MPM is poor and prospective trials in Mexico are needed.

      Clinical trial identification:


      Legal entity responsible for the study:
      Hospital de Oncología, Centro Médico Nacional Siglo XXI, Departamento de Oncología Médica

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      222P - Women: The other face of malignant pleural mesothelioma (Now Available) (ID 483)

      12:50 - 12:50  |  Presenting Author(s): M.E. Brandão  |  Author(s): M. Rocha, J.C. Silva, L. Nascimento, T. Gomes, B. Conde, A. Fernandes

      • Abstract
      • Slides

      Background:
      Malignant pleural mesothelioma (MPM) is a rare asbestos-related malignancy known to affect men in high-risk occupations. Recent publication data have suggested an increase in MPM cases among women but with a potential survival advantage of female gender. We sought to investigate gender differences in epidemiology, clinical characteristics and outcome among our MPM patients.

      Methods:
      We retrospectively reviewed the medical records of all MPM patients diagnosed in our center between 2000 and 2017. Data regarding demographics, asbestos exposure, symptoms, histology, tumor staging, CT findings, treatment and outcome were obtained for all patients.

      Results:
      We identified 29 cases of MPM, of which 38% occurred in women. The median age at diagnosis was similar for both genders (79 years in women vs 77 years in men). Domestic environment was the most important source of asbestos exposure for women (36%) whereas the vast majority of men cases (67%) were related to occupational exposure. In a significant proportion of women (54%) no source of asbestos exposure could be identified. The most common presenting symptom in the overall cohort was dyspnea, but women were more likely to present with cough and chest pain (82% vs 33% for cough; 73% vs 28% for chest pain). Pleural plaques were found in 36% of women and 72% of men. The median time to diagnosis was 2 months for both genders. Stage IV (TNM) disease was diagnosed in 64% of women and 83% of men. Women had a worse performance status at diagnosis (ECOG > 3 in 27% vs 5% in men). The main therapeutic strategies were best supportive care in women (64%) and chemotherapy in men (56%).There was a trend towards distant progression of the disease in women (54%) and local progression with invasion of lung parenchyma in men (44%). One-year survival rate was inferior in women (27% vs 39%).

      Conclusions:
      In our cohort, women didn't fit the traditional mesothelioma patient profile. Gender differences were found in modalities of asbestos exposure, presenting symptoms, radiological appearance, disease progression and outcome. Women demonstrated a worse 1-year survival rate, which may be attributed to the worse performance status at diagnosis, lower proportion of women treated with chemotherapy and higher rate of distant metastasis.

      Clinical trial identification:


      Legal entity responsible for the study:
      Centro Hospitalar de Trás-os-Montes e Alto Douro

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      223P - Sarcomatoid lung carcinoma: An uncommon and deadly entity (Now Available) (ID 628)

      12:50 - 12:50  |  Presenting Author(s): A. Sharma  |  Author(s): E. Contreras, K. Sandoval, L. Nicholson, J. Waalen, M. Bhangoo

      • Abstract
      • Slides

      Background:
      Pulmonary sarcomatoid carcinoma is a rare type of non-small cell lung cancer with poor prognosis. It is more common in males, has a strong association with smoking, and makes up mere 1.3% of all lung cancers. Our study examined the epidemiology, natural history, and prognostic factors of sarcomatoid lung carcinoma using a population-based registry.

      Methods:
      The Surveillance, Epidemiology, and End Results (SEER) Program database was utilized to isolate cases by tumor site and histology codes. The association between clinical and demographic characteristics and long-term survival was assessed.

      Results:
      A total of 582 histology confirmed cases were identified between 1973 and 2014. The median age of the patients was 68 years (range 29–96). Of the patients with a known tumor stage (N = 477), 74.8% had regional or distant stage, and 95% of patients with known histologic grade, had poorly or undifferentiated histologic features. 60.8% received surgical intervention. The median overall survival was 12 months (95% CI 9–14 months). In a multivariate analysis, age, sex, and SEER tumor stage were found to be a significant prognostic factors for disease specific survival. The prognosis of sarcomatoid carcinoma of the lung remains dismal, even in patients with resectable disease. Patients who underwent cancer directed surgery (64%) had median survival of 19 months with 1, 5 and 10-year survival rates of 63.7%, 33.6% and 28.8% respectively. Those patients who did not undergo cancer directed surgery (36%) had a poorer prognosis with a median survival of 4 months and 1, 5 and 10-year survival rates of 19.8%, 6.7% and 6.7% respectively.

      Conclusions:
      Sarcomatoid lung carcinoma often presents as a high grade, advanced malignacy with aggressive behavior and a dire prognosis. Patients are younger and yet have shorter survial times than those with more common lung cancers. Emphasis on new strategies for early detection and identification of additional risk factors, and noval therapies will be important to improve the outcome for patients with this malignancy.

      Clinical trial identification:


      Legal entity responsible for the study:
      Atul Narayan Sharma

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      224P - The role of surgery in the treatment of lung neuroendocrine tumors (NETs) based on a 3-year experience of a thoracic department (Now Available) (ID 484)

      12:50 - 12:50  |  Presenting Author(s): B. von Amann  |  Author(s): C. Rodrigues, A. Ribeiro, C. Bárbara, F. Félix

      • Abstract
      • Slides

      Background:
      NETs are a heterogeneous family of neoplasms comprising typical and atypical carcinoid (TC and AC), large-cell neuroendocrine carcinoma (LCNEC) and small cell lung carcinoma (SCLC). Surgical resection is the primary therapy for carcinoid tumors of the lung but plays a minor role in treatment of SCLC and LCNEC because tumors are often locally advanced or have metastasized at the time of presentation. Patients with limited cancer may demonstrate better disease control upon surgical treatment.

      Methods:
      Retrospective analysis of histologically confirmed lung NETs submitted to surgical treatment between 2015 and 2017 in a thoracic surgery unit of a tertiary hospital. Descriptive analysis of the main demographic, clinical, imagiological features and surgical treatment was performed.

      Results:
      Of the 1799 patients admitted on the unit, included 72 patients. 24 TC (33.4%), 32 AC (44.4%), 14 LCNEC (19.4%) and 2 SCLC (2.8%). 58.3% (n = 42) were female. The median age was 65 years (range 24–83) without difference by gender. Current smoker (SMK) 19.4% (n = 14), ex-SMK 26.4% (n = 19) and non-SMK 54.2% (n = 39). Median lesion dimension in CT scan was 25.7 mm, located in 2 lobes in 9 patients. PET was performed in 90% patients with median 6.3 SUV. Airway obstruction in 18% of patients. Lobectomy performed in 72.2% (52 patients, 6 of them involving 2 lobes) and segmentectomy performed in 27.8% (20 patients, 5 of them involving 2 lobes). The surgical approach was thoracotomy in 58.3% (n = 42) and minimally invasive (VATS) in 41.7% (n = 30). The mean length of stay was 10 days. The median pathological lesion size was 33.2 × 21.5 × 6 mm; Ki-67 antigen expression was evaluated in 43% patients (n = 31). Staging: I (65.3%, n = 47), II (20.9%, n = 15), III (6.9%, n = 5), IV (6.9%, n = 5). 92% TC were stage I; 15.6% AC were stage III and IV; 28.6% LCNEC were stage III and IV.

      Conclusions:
      This study reinforces the importance of the differential diagnosis of lung NETs in order to predict their behavior. Highlights the surgical treatment in TC in which the percentage of advance disease is low. On the other hand, AC and more aggressive tumors, such as LCNEC and SCLC should be treated in a multimodal concept reinforcing the importance of an experienced multidisciplinary team.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      225P - Surgical experience of advanced bronchopulmonary carcinoids: An analysis from a tertiary care hospital of North India (ID 372)

      12:50 - 12:50  |  Presenting Author(s): N. Kumar  |  Author(s): S. Deo, N.K. Shukla, S. Boriwal, V. Kumar, P. Ramanathan, D. Jain, K. Madan, S. Kumar

      • Abstract

      Background:
      Bronchopulmonary carcinoid is an uncommon, distinct malignant potential lung cancer. Surgery is the mainstay of treatment. The aim of this study was to analyze the epidemiology, complex surgical approaches and their outcomes.

      Methods:
      This was a retrospective study done in the Department of Surgical Oncology of a tertiary teaching institute of North India. All the case records were analyzed for clinical case history, preoperative treatment, surgical procedures, postoperative events, adjuvant treatment, histopathology and outcome.

      Results:
      A total of 30 patients who were operated on from 2012 to 2017 for bronchopulmonary carcinoids were reviewed. The mean age was 36 years with male predominance. 50% had a history of a cough and hemoptysis. Four patients were smokers. Six patients had been treated for tuberculosis and one patient received neoadjuvant chemotherapy due to histopathological misdiagnosis. One-third of patients had underlying pneumonia. Bronchoscopy-guided intraluminal tumor debulking was done in 5 cases to treat obstructive pneumonia before taking up for surgery. Ten pneumonectomies (including carinoplasty with latissimus dorsi muscle cover in three patients) were performed in patients with tumor in the main bronchus with extrabronchial spread. Other operative procedures included 13 bilobectomies, 7 lobectomies, bronchoplastic procedures in 4 patients (1 in bilobectomy and 3 in lobectomy). Postoperative mortality was nil; 6 patients had lobar collapse, high ICD output (conservatively treated) and surgical site infection postoperatively. In histopathology, 21 had typical and 9 had atypical carcinoids and all were margin negative except one. Two patients had lymph node metastasis and adjuvant chemotherapy was given to both. With the median follow-up of 22 months, all the patients are disease-free and doing well.

      Conclusions:
      Diagnostic dilemma and late presentation are major concerns in India. Aggressive surgical procedures including sleeve resection with bronchoplasty may be needed to achieve margin-negative resection because of good prognosis and long-term survival. In some cases bronchoscopic debulking can be done preoperatively to optimize the patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      All India Institute of Medical Science

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      226P - Accuracy of mutation analysis in small tissues from transbronchial lung biopsy with radial probe endobronchial ultrasound (ID 437)

      12:50 - 12:50  |  Presenting Author(s): M.K. Lee  |  Author(s): J.S. Eom

      • Abstract

      Background:
      There is little evidence to support the reliability of molecular analysis using transbronchial lung biopsy combined with radial probe endobronchial ultrasound and a guide sheath (EBUS-GS) specimens. In order to verify the accuracy of epidermal growth factor receptor (EGFR) mutation and anaplastic lymphoma kinase (ALK) gene rearrangement results using EBUS-GS specimens, we conducted a retrospective study using a prospectively collected database.

      Methods:
      From December 2015 to May 2017, consecutive patients who received EBUS-GS for the diagnosis of peripheral lung nodule or mass were registered. For the comparisons of EGFR mutation and ALK gene rearrangement analysis using EBUS-GS specimen, molecular analysis using surgical specimen was considered as gold standard. As a result, 70 patients who received both EBUS-GS and subsequent surgical treatment were included.

      Results:
      Using surgical specimens, EGFR mutation and ALK gene rearrangement were detected in 37% and 4%, respectively. In comparisions with reference value, the accuracy of EGFR mutation and ALK gene rearrangement correlations analysis of using EBUS-GS specimens were revealed as 97% and 100%, respectively.

      Conclusions:
      The present study demonstrated that the molecular studies, such as EGFR mutation and ALK rearrangement analysis, with the EBUS-GS specimen were reliable comparing with those using surgical specimen.

      Clinical trial identification:


      Legal entity responsible for the study:
      Pusan National University Hospital

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      227P - Stereotactic body radiotherapy with helical tomotherapy for lung tumors: A single-center experience (ID 404)

      12:50 - 12:50  |  Presenting Author(s): V. Figlia  |  Author(s): F. Cuccia, V. Valenti, A. Tripoli, G. Terranova, A. Lo Casto, G. Failla, T. Cucchiara, G. Mortellaro, G. Ferrera

      • Abstract

      Background:
      Several studies report favorable outcomes for stereotactic body radiotherapy (SBRT) in the treatment of early stage NSCLC or lung metastases. Here we introduce our experience of Helical TomoTherapy (HT) SBRT for primary and secondary lung tumors.

      Methods:
      Between March 2014 and June 2017, 57 lung lesions in 56 patients (pts), median age 72 years (range 26–91) underwent ablative HT-SBRT: 34 pts (60.7%) for primary tumors and 22 pts (39.3%) for lung metastases. Staging was performed by contrast-enhanced chest CT and PET-CT scans. Immobilization was obtained with breast board and abdominal pressure mould mask. The planning target volume (PTV) included a margin of 10 mm in cranio-caudal direction and 5 mm in all other directions around the internal target volume (ITV), defined basing on the volumetric sum of the clinical target volumes of a free breathing planning CT and a pretreatment MVCT scan acquired in treatment position. According to tumor site, different schedules of RT were used: 60–70 Gy in 8–10 alternate fractions for peripheral lesions (n = 47) and 50 Gy and 60 Gy in 10 daily fractions for central (n = 10). Median BED10 was 96 Gy (range 75–119). Treatment-related toxicity was evaluated using CTCAE v4.0 scale. For the first year of follow-up, physical evaluation and chest CT were conducted every 2–3 months.

      Results:
      Median GTV and PTV sizes were respectively 6.14 and 22.86 cc (range, 0.87–75.1 and 65.15–131.03). Median duration of RT was 15 days (range 10–20) and all pts were successfully treated with mild acute adverse events. Concurrent EGF-R-targeting TKIs were administered in 4 cases. With a median follow-up of 17 months (range 6–39) no ≥ G3 radiation pneumonitis or ≥ G2 esophagitis occurred; only 1 patient showed G2 non-cardiac chest pain, 2 pts G2 radiation pneumonitis. At the time of the analysis, 4 local failures were registered, resulting in Local Control (LC) rates of 97.1% for primary tumors and 84.8% for metastatic lesions; 49 pts are alive and 7 dead: 1y- and 2y- Overall survival rates were 96.6% and 87.8% for primary lesions and 94.1% and 79% for metastases, respectively.

      Conclusions:
      HT-SBRT in primary or metastatic lung tumors demonstrates low risk of normal tissue complications and high rates of LC.

      Clinical trial identification:


      Legal entity responsible for the study:
      ARNAS Civico Hospital, Palermo, Italy

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      228P - Augmentation of NAD+ by NQO1 activation attenuates cisplatin-mediated hearing impairment (ID 548)

      12:50 - 12:50  |  Presenting Author(s): S. Yang  |  Author(s): K.K. Kwon, H. So

      • Abstract

      Background:
      Cisplatin [cis-diaminedichloroplatinum-II] is an extensively used chemotherapeutic agent, and one of its most adverse effects is ototoxicity. A number of studies have demonstrated that these effects are related to oxidative stress and DNA damage. However, the precise mechanism underlying cisplatin-associated ototoxicity is still unclear. The cofactor nicotinamide adenine dinucleotide (NAD[+]) has emerged as a key regulator of cellular energy metabolism and homeostasis. Although a link between NAD[+]-dependent molecular events and cellular metabolism is evident, it remains unclear whether modulation of NAD[+] levels has an impact on cisplatin-induced hearing impairment.

      Methods:
      To investigate whether augmentation of NAD[+] by NQO1 activation using b-Lapachone (b-Lap) attenuates cisplatin-mediated hearing impairment, male C57BL/6 mice and NQO1 knockout mice on a C57BL/6 background were used. For analysis of the auditory threshold, auditory brainstem response (ABR) was recorded. For biochemical analysis, we measured the enzymatic activity of SIRT1, PARP1, ROS production, NAD+/NADH ratio, mRNA levels of miR-34a and pro-inflammatory cytokines. Immunohistochemistry and western blot analysis were also performed.

      Results:
      We have demonstrated for the first time that both the protein expression level and the activity of SIRT1 were suppressed by the reduction of intracellular NAD[+] levels in cisplatin-treated cochlear tissue. We also found that the decrease in SIRT1 protein expression and its activity after cisplatin exposure were mediated by the increase in transcriptional activity of p53 for miR-34a expression and PARP-1 activation causing NAD[+]-depletion, respectively. However, the increase in cellular NAD[+] levels by NQO1 activation using b-Lap prevented mice from cisplatin-induced cochlear damage and hearing impairment through the modulation of PARP-1, SIRT1, p53, and NF-kB.

      Conclusions:
      Considering that b-Lap itself did not attenuate the tumoricidal effect of cisplatin, these results suggest that the direct modulation of the cellular NAD[+] level by pharmacological agents could be a promising therapeutic strategy for enhancing the efficacy of cisplatin chemotherapy without its adverse effects.

      Clinical trial identification:


      Legal entity responsible for the study:
      N/A

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

    • +

      229P - Clinical and laboratory markers of prognosis in lung cancer patients with hypercalcemia (Now Available) (ID 329)

      12:50 - 12:50  |  Presenting Author(s): K. Syrigos  |  Author(s): A. Ntineri, P. Bakakos, D. Grapsa, E. Dalakou, E. Kotteas, A. Charpidou

      • Abstract
      • Slides

      Background:
      Hypercalcemia is a severe complication in lung cancer patients, imposing prompt recognition, investigation and treatment. The primary aim of our study was to further investigate the clinical and laboratory prognostic features in a cohort of lung cancer patients with hypercalcemia.

      Methods:
      The medical records of a total of 100 sequential, non-selected, patients with histologically or cytologically-confirmed diagnosis of small or non-small cell lung cancer (SCLC/NSCLC) and hypercalcemia, diagnosed and treated at the Oncology Unit of Sotiria Athens General Hospital between December 2015 to April 2016 were retrospectively reviewed. Demographic, clinical and laboratory patients’ characteristics were correlated with outcome data.

      Results:
      Mean age (±SD) of our patient population at the diagnosis of hypercalcemia was 64.3 (±8,8) years; male to female ratio was 86/14. The albumin-corrected calcium was 11.0 ± 0.6 mg/dl (range 10.5–13.9). The majority of patients had non-small cell lung cancer (89%), mainly of squamous type (48.3%) and adenocarcinoma (43.8%), and stage IV disease (57%). Development of bone metastases (BMs) was observed in 17.7% of patients, while 5% of patients had received bisphosphonates. Hypercalcemia-leukocytosis syndrome was associated with advanced stage (p = 0.001) and the presence of bone metastases (p < 0.05). Median time to progression was 3.5 months (interquartile range: 1.2–7.2) and median overall survival (mOS) was 13.8 months (95% CI: 7.3–20.2). In univariate Cox proportional hazard models, patients with lower serum albumin levels (p < 0.001), concomitant presence of leukocytosis (p < 0.05), advanced disease stage (p < 0.01) and negative history of resected disease (p < 0.001) had higher risk of death. In multivariate Cox regression analysis, serum albumin (HR 0.37, 95% CI 0.16–0.86; p < 0.05), metastatic disease (HR 1.92, 95% CI 1.01–3.68; p < 0.05) and the history of surgical resection (HR 0.25, 95%CI 0.10–0.61; p < 0.01) were independent predictors of survival.

      Conclusions:
      Our study results, derived from a combined cohort of SCLC and NSCLC patients with hypercalcemia, suggest that serum albumin levels may provide independent prognostic information in this setting.

      Clinical trial identification:


      Legal entity responsible for the study:
      University of Athens

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      230P - Experience with the implant of vascular access devices by medical oncologists in a non-surgical setting (Now Available) (ID 465)

      12:50 - 12:50  |  Presenting Author(s): A. Revuelta  |  Author(s): D. Rodríguez Rubí, L. Fáez, M.D.P. Solís, S. Fernández Arrojo, C. Iglesias Gómez, D. Gómez, J. del Río Fernández, A. Castillo Trujillo, E. Esteban González

      • Abstract
      • Slides

      Background:
      Totally implantable central venous catheters are widely used in the management of patients (pts) with malignant diseases to facilitate drug delivery for the new therapeutic protocols. These are based on continuous administration and higher doses of chemotherapeutic agents with relative phlebitis problems and supportive treatment. Staff of our department, specially trained in the routine implantation of central venous accesses were in charge of the procedure. The technique was carried out under local anesthesia in a special suite of day hospital, under strict aseptic measures without fluoroscopic control.

      Methods:
      From Sep 94 to January 2017, 1665 devices (port-a-cath systems [PS]) were implanted in 1627 pts, with a median age of 50.5 yr (range 14–81), and median K.I. 70% (50–100), female 982/male 683. Venous access: right interior jugular 983, left subclavian 316, right subclavian 333, left interior jugular 33. A thorax X-ray was performed after each procedure and in 216 pts prophylactic antibiotics were given.

      Results:
      The venous access remained implanted a median of 438 days (1- +2210). Complications occurred in 266 placements (16%): infections 116 (7%); deep venous thrombosis 66 (4%) obstruction 10 (0.6%); malpositioned 16 (2%); fractures/migration 28 (1.7%); pneumothorax 6 (0.32%); local skin necrosis 7 (0.6%). 520 devices were removed, 347 (66%) after completing planned therapy, and 173 (34%) due to complications [Infections (92), migration (22), malposition (12), venous thrombosis (26), obstruction (11) and skin necrosis (10)]. Cost-effectiveness of venous catheters in a non-surgical setting compared with devices implanted by interventional vascular radiologists in operating room turned out to be 1000 euro cheaper for each device.

      Conclusions:
      Our experience suggests that implant of vascular access devices by medical oncologist in a non-surgical setting has similar or even less complications and is more cost effective with regard to radiology suite and operating room placement procedures.

      Clinical trial identification:


      Legal entity responsible for the study:
      SESPA

      Funding:
      Has not received any funding

      Disclosure:
      All authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      231P_PR - Impact of the continuity of nursing care delivered by a pivot nurse in oncology on improving satisfaction and quality of life of patients with advanced lung cancer (Now Available) (ID 270)

      12:50 - 12:50  |  Presenting Author(s): E. Kassouf  |  Author(s): M. Tehfe, M. Floresci, N. Blais

      • Abstract
      • Slides

      Background:
      Health care organizations around the globe have been implementing different strategies aimed at improving their care systems to obtain better patient- physician interaction and resolve underlying issues leading to patient dissatisfaction. In an effort to improve continuity of care inside a network of interdisciplinary teams, the Ministry of Health and Social Services of Québec has implemented the recruitment of pivot nurses in oncology services.

      Methods:
      This study was conducted at the Notre Dame university hospital in Montreal. Patients were selected from the outpatient admissions’ list, three months after the start of their treatment, and divided into two cohorts: the continuity of care (CC) cohort, where patients were followed by a PNO, and the usual care (UC) cohort, who received standard care from the oncology clinic staff. Participants in both groups completed the Princess Margaret Hospital Patient Satisfaction with Doctor Questionnaire (PMH/PSQ-MD), the FACT-L Scale for quality of life assessment and questions evaluating the understanding of their health status and disease. Another ten questions were addressed specifically to the CC in regards to nursing care and the health care system in Quebec.

      Results:
      In total, 65 patients were recruited, 82% of whom were assigned to the CC cohort. Analysis of the PMH PSQ-MD questionnaire demonstrated superior outcomes in regards to information exchange, empathy, and quality of life for the CC cohort (p < .001). The FACT-L questionnaire also favored the CC cohort in terms of physical well being, social and family well being, emotional well being, and functional well being. These differences were highly significant and translated into better satisfaction when comparing the total score of the CC cohort to the UC cohort (p < .0001). Other variables examined revealed an adequate fulfilment of the PNO role as regarded by the participants, except for matters of an intimate nature.

      Conclusions:
      The PNO appears to have a substantial role in the care of patients with advanced lung cancer. Continuity of care seems to improve patients’ quality of life and satisfaction by reducing the symptom strain experienced in the ambulatory patients.

      Clinical trial identification:


      Legal entity responsible for the study:
      Centre Hospitalier de l'Université de Montréal

      Funding:
      Has not received any funding

      Disclosure:
      E. Kassouf: Personal fees from Lilly, Sanofi Canada, Amgen, Celgene, and Bristol-Myers Squibb outside the submitted work. M. Tehfe: Personal fees from Lilly, Amgen, and Celgene outside the submitted work. M. Floresci: Personal fees from Lilly, Merck, and Boehringer Ingelheim outside the submitted work. N. Blais: Personal fees from Merck, Pfizer, AstraZeneca, and Bristol-Myers Squibb outside the submitted work.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.

    • +

      232TiP - Lanreotide autogel/depot in lung neuroendocrine tumours: The randomized, double-blind, placebo-controlled, international phase 3 SPINET Study (Now Available) (ID 600)

      12:50 - 12:50  |  Presenting Author(s): X.M. Truong Thanh  |  Author(s): D. Reidy-Lagunes, M. Kulke, E. Wolin, S. Singh, D. Ferone, D. Hoersch, A. Houchard, M. Caplin, E. Baudin

      • Abstract
      • Slides

      Background:
      Treatment options for advanced lung neuroendocrine tumours (NETs) are limited. The phase 3 CLARINET study demonstrated antitumour efficacy of the somatostatin analogue (SSA) lanreotide autogel/depot (LAN) 120 mg/28 days vs. placebo for metastatic gastroenteropancreatic grade 1/2 (Ki-67 < 10%) NETs, but prospective data on SSAs in advanced lung NETs are lacking. This study evaluates antitumour efficacy and safety of LAN 120 mg in patients with advanced lung NETs.

      Trial design:
      SPINET is a large double-blind, placebo-controlled phase 3 study. Main inclusion criteria are adults with well-differentiated typical/atypical, metastatic and/or unresectable lung NETs, at least one measurable lesion on CT/MRI imaging (RECIST v1.1), positive somatostatin receptor imaging, ECOG PS0–1. Patients have a maximum of two previous courses of cytotoxic chemotherapy, molecular-targeted therapy or interferon. A total of 216 patients from 80 sites across the USA, Canada and Europe will be randomized 2:1 to either LAN 120 mg/28 days or placebo, both with best supportive care, until progressive disease (PD)/death or unacceptable toxicity. Patients experiencing PD on placebo may opt to receive LAN 120 mg in an open-label extension. All patients experiencing PD will be followed to document survival, quality-of-life (QoL) and subsequent treatment. Recruitment began in July 2016. The primary endpoint is progression-free survival (PFS, time from randomization to PD/death; central review, RECIST v1.1). Main secondary endpoints include, objective response rate, overall survival, time to treatment failure, change in chromogranin A, QoL, and safety. SPINET is the first prospective randomized trial designed to assess LAN 120 mg in typical/atypical carcinoid lung NETs.

      Clinical trial identification:
      NCT02683941; EudraCT: 2015-004992-62

      Legal entity responsible for the study:
      Ipsen

      Funding:
      Ipsen

      Disclosure:
      X-M. Truong Thanh, A. Houchard: Employee of Ipsen. D. Reidy-Lagunes: Honoraria from Novartis; Consultancy and speakers’ bureau fees from Ipsen, Novartis, and Pfizer; Research funding from Novartis. M. Kulke: Consultancy fees from Ipsen, and Novartis. E. Wolin: Consultancy and advisory fees from Ipsen, and Advanced Accelerator applications. D. Ferone: Renumeration for services from Novartis, Ipsen, and Pfizer. D. Hoersch: Fees from Lexicon; Grants, personal fees and other from Novartis, Ipsen, and Pfizer. M. Caplin: Honoraria, consultancy and speakers’ bureau fees from Ipsen, Advanced Accelerator Applications, and Novartis. E. Baudin: Remuneration for services (advisory board or board of directors; corporate-sponsored research; consulting fee; research investigator; speaker) from Ipsen, Novartis, Pfizer, and Advanced Accelerator Applications. All other authors have declared no conflicts of interest.

      IASLC Members: To view this content or have the option to purchase this event, click here to login.
      Conference Attendees & Access Code holders: Click here to enter your Access Code. Already entered your Access Code? Please login.