Virtual Library

Start Your Search

David Planchard

Moderator of

  • +

    ES 08 - Molecular Diagnostics and Targeted Therapy (ID 517)

    • Event: WCLC 2017
    • Type: Educational Session
    • Track: Chemotherapy/Targeted Therapy
    • Presentations: 5
    • +

      ES 08.01 - Overview of Diagnostics and Pathology (ID 7614)

      11:00 - 12:30  |  Presenting Author(s): Tetsuya Mitsudomi

      • Abstract
      • Presentation
      • Slides

      Abstract:
      Recent advances in molecular biology have revealed that lung cancer is not a single disease and that there are subsets of non-small cell lung cancer (NSCLC) with specific genetic alterations that are critical to the growth and survival of cancer cells. Alterations of the EGFR, ALK and ROS1 gene, which are present in a mutually exclusionary fashion, are representative driver oncogene mutations. Targeted drugs against each driver oncogene usually result in dramatic tumor shrinkage and prolongation of progression free survival (PFS) compared with conventional platinum doublet chemotherapy. However, there is only a weak association between WHO pathologic classification 2015 and type of driver oncogenes. Therefore, it is of utmost importance to identify who are likely to benefit from targeted drugs by performing molecular tests for each lung cancer patient who is a candidate for drug therapy. A list of driver oncogenes is further expanding; BRAF, RET, MET, HER2, NTRK1 are being recognized as new drivers that can be exploited in the clinic. It is getting more practical to screen these molecular alterations by use of next generation sequencing technology, rather than to detect each gene alterations one by one using different platforms. We have also known that not all the tumors with mutations of the same gene behave similarly. For example, while deletional mutation in exon 19 and L858R in exon 21 are two representative mutations that sensitize cancer cells to EGFR-tyrosine kinase inhibitors (TKI), G719X in exon 18 has an intermediate sensitivity and insertional mutation in exon 20 or de novo T790M are known to be resistant. It has been shown that there is a heterogeneity in efficacy of EGF-TKIs depending on the class of mutation. For example, afatinib is active among other EGFR-TKIs for exon 18 mutations. Furthermore, a certain molecular context is known to be associated with primary resistance even within lung cancers with the same EGFR mutations. For example, it is reported that mutations in the PI3K/AKT/mTOR pathway (AKT1, PIK3CA, STK11, PTEN) or TP53 mutations are more frequent in non-responders and are associated with shorter PFS. This context dependence may present in other driver oncogenes, too. Acquired resistance is almost inevitable in the treatment of lung cancer with targeted drug. Mechanisms of this resistance has been extensively studied and now we know there are at least 3 types of mechanisms; i.e., 1) target modification by the secondary mutation that alters the affinity between the drug and the target relative to the affinity between ATP and the target (e.g., T790M in EGFR, L1196M in ALK), 2) accessory pathway activation that bypass the inhibitory effect of the drug(e.g., Met amplification in EGFR), and histologic transformation, such as small cell lung cancer transformation and epithelial-mesenchymal transition. We are now able to use the newer generation of TKIs to treat some of the resistance due to the secondary mutation of the target gene. Osimertinib has recently been shown to prolong PFS of patients who acquired resistance to EGFR-TKI through T790M mutation compared with platinum-pemetrexed in the AURA 3 trial. Therefore, detection of this mutation which accounts for about 50~60 % of the acquired resistance against EGFR-TKI is important. However, re-biopsy is sometimes more challenging compared with that in the first-line setting, and therefore detection of T790M in cell-free DNA in plasma has been rapidly developed and is now approved in regulatory authorities in several countries. There is another issue which should be taken into consideration when treating patients with acquired resistance. When there are multiple metastatic lesions, resistance mechanisms may vary from one tumor to another. Hence, it can happen that while one tumor shrinks but others increase in size. It may be reasonable and thus beneficial for patients when treatment is planned according to most prevalent mechanism of resistance in the plasma as a sum of total resistant mechanism. In this talk, I would like to overview recent advances of molecular diagnosis in targeted therapy of lung cancer and also like to discuss future perspectives in this field.

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

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

    • +

      ES 08.02 - Update of the Management of EGFR-Positive NSCLC (ID 7615)

      11:00 - 12:30  |  Presenting Author(s): James Chih-Hsin Yang

      • Abstract
      • Presentation
      • Slides

      Abstract not provided

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

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

    • +

      ES 08.03 - Update of the Management of ALK-Positive NSCLC (ID 7616)

      11:00 - 12:30  |  Presenting Author(s): D. Ross Camidge

      • Abstract
      • Presentation
      • Slides

      Abstract:
      ALK positive NSCLC represents 2-7% of advanced NSCLC. Three ALK TKIs have shown positive first line trials against either platinum-doublet chemotherapy (ceritinib and crizotinib) or against crizotinib (alectinib). At least three other first line trials against crizotinib are ongoing (brigatinib, ensartinib and lorlatinib). Activity of different ALK TKIs post crizotinib are characterized by comparable response rates but differing toxicity profiles, durations of benefit and extent of CNS activity. With changes in the first line standard, data post non-crizotinib ALK TKIs continues to emerge with attendant caution re the applicability of both biological and clinical data currently available for clinical decision making. Advances in our understanding of CNS trial endpoints has also helped facilitate cross trial comparisons of CNS activity of these different agents. Chemotherapeutic, radiotherapeutic and immunotherapeutic options other than ALK TKIs have all generated different clinical datasets - defining both some reasonable clinical options and some clearly in need of additional research.

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

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

    • +

      ES 08.04 - Emerging Target Therapy in NSCLC (ID 7617)

      11:00 - 12:30  |  Presenting Author(s): Enriqueta Felip

      • Abstract
      • Presentation
      • Slides

      Abstract:
      As a result of recent advances, systematic genomic testing for patients with non-small cell lung cancer (NSCLC) is the new standard of care in clinical decision-making, due to the identification of driver molecular alterations that have triggered the development of new molecules targeting these specific alterations in cancer cells. Several studies have enabled to conclude that both EGFR-mutant and ALK-positive NSCLC constitute two defined subgroups of oncogene-driven tumors with potentially effective targeted therapy. Furthermore, approximately 15-20% of NSCLC diagnosed in Europe and North America bear EGFR mutations or ALK rearrangements, enhancing the significance of the development of drugs capable of interfering with their intracellular effects. Based on these results, the identification of other activating mutations has been pursued in hopes of improving survival in NSCLC by specifically treating these genomic alterations. These potential therapeutic targets include ROS1, BRAF, RET, HER2, MET exon 14 skipping mutations and NTRK, among others. Here, we seek to review the characteristics of emerging targets that enable interaction with molecules that specifically target these receptors in lung adenocarcinomas, as well as the results of preliminary studies that assess the efficacy of these new strategies applied to NSCLC. ROS1 ROS1 rearrangement characterizes a small subset (1%–2%) of NSCLC and is associated with slight/never smoking patients and adenocarcinoma histology. Crizotinib was shown to harbor relevant activity in ROS1-rearranged NSCLC. A number of agents including ceritinib, lorlatinib and entrectinib are now been developed in order to overcome the resistance to crizotinib. At present, ROS1-rearranged patients represents a clearly defined NSCLC molecular subgroup with highly active therapeutic options. BRAF BRAF mutations occur in 2%–4% of patients with NSCLC, with the most common resulting in a glutamate substitution for valine at codon 600 (V600E). Non‐V600E BRAF mutations make up the remaining BRAF mutations and may be either activating (i.e., G469A/V, K601E, L597R) or inactivating (i.e., D594G, G466V). Efforts targeting BRAF‐mutant NSCLC to date have almost exclusively focused on patients with V600E‐mutant disease. Direct inhibition of mutant BRAF and/or the downstream mitogen-activated protein kinase kinase has led to good outcomes survival in patients with BRAF-mutant metastatic NSCLC. Dabrafenib plus trametinib achieved 63·2% response rate (RR) in BRAF(V600E)-mutant NSCLC. RET RET fusions are detected in 1-2% of lung adenocarcinomas and a number of genes, such as KIF5B, CCDC6, NCO4 and TRIMM33, can act as fusion partners. In a global registry of RET positive NSCLC patients, 41 received a RET inhibitor achieving a median progression-free survival of 2.9 months and a median overall survival of 6.8 months. Response rate was 34% for those patients receiving cabozantinib and 27% for those receiving vandetanib. Overall RET inhibitors strategies seem active in a subgroup of patients with RET-rearranged NSCLC. However, RR is lower to that observed in EGFR-mutated/ALK-positive patients. HER2 HER2 mutations are identified in about 2-4% of NSCLC and are critical for lung carcinogenesis. A number of series shows the chemosensitivity of HER2-driven NSCLC, and the potential interest of HER2-targeted agents. In a recent study, NSCLC patients with HER2 mutations were treated with T-DM1 and achieved a 44% RR. MET Approximately 2-3% of NSCLCs harbor activating mutations of the MET proto-oncogene that cause exon 14 skipping (METex14) and accumulation of c-Met lacking a juxtamembrane domain. Recently, the clinical activity of anti-Met-targeted therapy was demonstrated in patients harboring MET exon 14 skipping lung cancer. MET seems a relevant target in NSCLC and a number of clinical trials with MET inhibitors in this population are now ongoing. NTRK TRK rearrangements represent the molecular driver of a subset of solid tumors, including 1-2% of NSCLCs. Preliminary data indicate that molecularly selected NSCLC patients harboring NTRK fusions derive an unprecedented clinical benefit from Trk-directed targeted therapies. There are two different targeted agents, entrectinib and larotrectinib, that are in phase II testing for any patients who have solid tumors with NTRK rearrangement, including NSCLC patients. Both drugs have achieved dramatic responses, regardless of histology in earlier phase I studies. In a study presented at ASCO 2017, larotrectinib has demonstrated consistent and durable antitumor activity in TRK fusion cancers, across a wide range of ages and tumor types. REFERENCES Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS-1 rearranged non-small-cell lung cancer: N Engl J Med. 2014; 371:1963-1971. Planchard D, Besse B, Groen HJ, et al. Dabrafenib plus trametininb in patients with previously treated BRAF (V600E)-mutant metastatic non-small cell lung cancer: an opne-label, multicentre phase 2 trial. Lancet Oncol. 2016;17:984-993. Gautschi O, Milia J, Filleron T, et al. Targeting RET in Patients With RET-Rearranged Lung Cancers: Results From the Global, Multicenter RET Registry. J Clin Oncol. 2017;35:1403-1410. Mazières J, Barlesi F, Filleron T, et al. Lung cancer patients with HER2 mutations treated with chemotherapy and HER2-targeted drugs: results from the European EUHER2 cohort. Ann Oncol. 2016;27:281-286. Lu X, Peled N, Greer J, et al. MET exon 14 mutation encodes an actionable therapeutic target in lung adenocarcinoma. Cancer Res. 2017 May 18. [Epub ahead of print]. Drilon A, Nagasubramanian R, Blake JF, et al. A Next-Generation TRK Kinase Inhibitor Overcomes Acquired Resistance to Prior TRK Kinase Inhibition in Patients with TRK Fusion-Positive Solid Tumors. Cancer Discov. 2017 Jun 3. [Epub ahead of print]. Hyman DM, Laetsch TW, Kummar S, et al. The efficacy of larotrectinib (LOXO-101), a selective tropomyosin receptor kinase (TRK) inhibitor, in adult and pediatric TRK fusion cancers. J Clin Oncol. 2017;35 (suppl; abstr LBA2501). Riely GL. What, When, and How of Biomarker Testing in Non-Small Cell Lung Cancer. J Natl Compr Canc Netw. 2017;15:686-688. Jordan EJ, Kim HR, Arcila ME, et al. Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies. Cancer Discov. 2017;7:596-609.

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

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

    • +

      ES 08.05 - Oncogenomics for Clinicians (ID 7618)

      11:00 - 12:30  |  Presenting Author(s): Matthew Meyerson

      • Abstract
      • Presentation
      • Slides

      Abstract not provided

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

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



Author of

  • +

    MA 05 - Immuno-Oncology: Novel Biomarker Candidates (ID 658)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Immunology and Immunotherapy
    • Presentations: 1
    • +

      MA 05.03 - The Early Monitoring of Derived Neutrophil-To Lymphocyte Ratio (dNLR) Could Be a Surrogate Marker of Benefit of Immunotherapy in NSCLC  (ID 10147)

      15:45 - 17:30  |  Author(s): David Planchard

      • Abstract
      • Presentation
      • Slides

      Background:
      Baseline high derived NLR (dNLR>3, neutrophils/(leucocytes-neutrophils) ratio) has recently correlated with no benefit to immune checkpoint inhibitors (ICI) in advanced NSCLC, but the dynamic monitoring of dNLR has not been assessed in this population.

      Method:
      dNLR at baseline, at 2[nd] cycle and at progressive disease were retrospectively collected in advanced NSCLC patients treated with ICI from November 2012 to April 2017, in a multicentric cohort (N= 292) from 4 European centers. The primary endpoint was overall survival (OS), and secondary endpoints were progression free survival (PFS), response rate (RR) and disease control rate (DCR).

      Result:
      Out of 292 patients (67%) were males, 264 (92%) smokers and 239 (83%) with PS ≤1, with median age 64 years; 153 (52%) had adenocarcinoma and 114 (30%) squamous; 44 (15%) were KRASmut, 11 (4%) EGFRmut and 3 (1%) ALK positive. PDL1 was ≥ 1% by immunohistochemistry in 67 (76%), negative in 21 (24%) and unknown in 204 patients. The median of prior lines was 1 (0-10). The median follow-up was 12 months (m) [11-14]. The median PFS and OS were 4m [3-5] and 11m [9-15]. Baseline dNLR was>3 in 106 patients (36%) and at 2[nd] cycle in 90 patients (32%). dNLR>3 at baseline and at 2[nd] cycle were associated with poor PFS (p<0.0001 and p=0.0008, respectively), poor OS (both p<0.0001) and progressive disease (p=0.002 and p=0.005, respectively). At 2[nd] cycle of ICI, the dNLR status (> high or ≤ 3 low) changed in 63 patients: in 38 (14%) dNLR decreased; in 25 (9%) dNLR increased. According to the dNLR monitoring (combining dNLR at baseline et at 2[nd] cycle), the median OS was 17m (95%CI 13-NA) when dNLR remained low (n=153), 10m (95%CI 7-NA) when dNLR changed (n=64) and 4m (95%CI 3-7) when dNLR remained high (dNLR>3, n=64, p<0.0001).The dNLR monitoring was also associated with PFS (p=0.002), RR and DCR (p=0.003 and p=0.013, respectively).

      Conclusion:
      Monitoring dNLR at baseline and at 2[nd] cycle could be a routinely tool to early assess benefit to ICI in NSCLC patients on treatment. The dNLR monitoring showed a strong correlation with OS and PFS. Modification of dNLR between baseline and 2[nd] cycle impacts outcomes in NSCLC patients treated with ICI.

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

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

  • +

    MA 10 - Immunotherapy I (ID 664)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Immunology and Immunotherapy
    • Presentations: 1
    • +

      MA 10.11 - Hyperprogressive Disease (HPD) Is Frequent in Non-Small Cell Lung Cancer (NSCLC) Patients (Pts) Treated with Anti PD1/PD-L1 Agents (IO) (ID 10222)

      11:00 - 12:30  |  Author(s): David Planchard

      • Abstract
      • Presentation
      • Slides

      Background:
      Using Tumor Growth Rate (TGR), HPD was identified in 9% of 131 advanced cancer pts, treated with IO in a single institution (Champiat et al. 2017). In this study, we explored HPD in a large, multicenter cohort of advanced NSCLC pts treated with IO.

      Method:
      We performed a retrospective analysis of consecutive NSCLC pts treated with IO, in 8 institutions, between November 2012 and April 2017. Eligibility criteria required, for each patient: 2 CT scans performed before starting IO and one during IO, an interval between two CT scans ≥2 weeks or 3 months (m) and presence of target lesions. CT scans were centrally assessed according to RECIST 1.1 criteria. We calculated TGR before IO (TGR pre-IO) and during IO (TGR IO); patients were defined HPD if they had progression disease (PD) at first evaluation during IO and a ≥ 2-fold increase in the TGR IO compared to TGR pre-IO. Median overall survival (mOS) was estimated using Kaplan-Meier method for the total population and HPD pts.

      Result:
      Among 419 eligible pts, 86 were excluded for inadequate intervals between CT scans. Among 333 evaluable pts, 63% were male, 46% ≥65 years, 43% smokers; 12% had PS ≥ 2, 65% adenocarcinoma, 45% ≥3 metastatic sites, 22% KRAS mutation, 4% EGFR mutation, 1% ALK rearrangement; 21% had PD-L1 positive status, 10% negative, 69% unknown, >90% received single agent PD-1 inhibitor in ≥ 2 line. Response rate (RR) to IO was 18%, median follow up was 12 m [10-14]. 33% of pts had TGR IO ≥1 (not regressing tumors), 25% had TGR IO ≥ 2-fold TGR pre-IO and 54 pts (16%) had HPD. 15 pts (4%) had confirmed pseudoprogression, 3 were initially qualified as HPD. Compared to not-HPD, HPD pts had more frequently ≥ 3 metastatic sites at baseline (59% vs 43% p=0,02) and more new lung lesions during IO (34% vs 17% p=0,007). PD-L1 negative status was more common among HPD pts but the association was borderline significant (53% vs 28% p=0,05). Age, clinical, molecular characteristics, RR to treatment before IO, baseline tumor burden, liver or brain new lesions during IO were not different according to HPD status. mOS was 13 m [10-17] in the total population, 5 m [3-8] in HPD pts.

      Conclusion:
      HPD occurred in 16% of advanced NSCLC pts treated with IO and was associated with plurimetastatic disease and appearance of new lung lesions. Further work is needed to characterize HPD prognostic value.

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

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

  • +

    MA 11 - Emerging Diagnostic/Biomarkers in NSCLC (ID 668)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Advanced NSCLC
    • Presentations: 1
    • +

      MA 11.01 - Liquid Biopsies for Monitoring BRAF Mutation (V600E) in Advanced BRAF (V600E) Non-Small Cell Lung Cancer (NSCLC) (ID 10232)

      11:00 - 12:30  |  Author(s): David Planchard

      • Abstract
      • Presentation
      • Slides

      Background:
      Circulating tumor DNA (ctDNA) has been shown beneficial in monitoring EGFR mutations in blood, especially for the detection of resistance mutations, like T790M in NSCLC patients. However, the role of BRAF (V600E) ctDNA for monitoring the patient’s response has not been studied yet. The aim of this study was to determine the clinical relevance of BRAF (V600E) ctDNA for monitoring the response to BRAF inhibitors in a prospective cohort of advanced NSCLC BRAF (V600E) patients.

      Method:
      We prospectively enrolled advanced NSCLC patients with BRAF (V600E) treated with BRAF +/- MEK inhibitors in our institution. A blood sample was collected at different time points, including at baseline, during treatment and at progressive disease. ctDNA BRAF analysis was performed using the Inivata InVision platform (enhanced tagged-amplicon next-generation sequencing (eTAM-Seq).

      Result:
      Between June 2016 and June 2017, 14 patients have been included. Eight patients (57%) were females, 9 (64%) non-smokers, with a median age of 63 years (35-70). All the patients had adenocarcinoma and BRAF (V600E) mutation in tissue analysis. Thirteen patients (93%) had stage IV at diagnosis, 7 patients (50%) with bone, 6 (43%) pleural and 4 (29%) lung metastasis. The median of lines of treatment received was 2 (1-4). Thirteen patients (93%) received BRAF + MEK inhibitor and 1 patient (14%) BRAF inhibitor, with an objective response rate of 64% (1 complete, 8 partial response) and disease control rate of 86%. BRAF mutation detection was tested under treatment in 12 patients (86%). Longitudinal analysis was performed from the serial sampling in 6 patients to date: 4 patients (67%) were ctDNA positive for BRAF (V600E) at time of progression, with a range of allelic frequency of 0.11-6.16%. BRAF mutation was not detectable in patients with objective response (2/6, 33%) at time of sample collection(s). Additional BRAF (V600E) NSCLC patient samples are being analyzed.

      Conclusion:
      Liquid biopsy for monitoring BRAF (V600E) using ctDNA appears to be feasible and useful in advanced NSCLC patients. Updated longitudinal results for the complete patient cohort will be presented at the meeting.

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

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

  • +

    MA 19 - Mesothelioma: Bench to Bedside (ID 680)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Mesothelioma
    • Presentations: 1
    • +

      MA 19.03 - Nintedanib + Pemetrexed/Cisplatin in Malignant Pleural Mesothelioma (MPM): Phase II Biomarker Data from the LUME‑Meso Study (ID 8111)

      11:00 - 12:30  |  Author(s): David Planchard

      • Abstract
      • Presentation
      • Slides

      Background:
      Nintedanib is a triple angiokinase inhibitor. LUME-Meso (NCT01907100) is a randomised, Phase II/III study of ≤6 cycles of nintedanib+pemetrexed/cisplatin versus placebo+pemetrexed/cisplatin, followed by nintedanib or placebo maintenance, in chemo-naïve patients with MPM. In Phase II results, nintedanib+pemetrexed/cisplatin improved progression-free survival (PFS) versus control (hazard ratio [HR]=0.54; p=0.010), with a trend for prolonged overall survival (OS; HR=0.77; p=0.319). Benefit was most pronounced in patients with epithelioid tumours. Since no pharmacodynamic/predictive biomarkers are validated for anti-angiogenic therapies, exploratory analyses were conducted to investigate potential associations of plasma-derived angiogenic factors and genomic markers with treatment outcome in the LUME-Meso Phase II epithelioid population.

      Method:
      Blood samples were collected at baseline and, for patients receiving maintenance, at monotherapy Cycle 3 (C3mono) and end of monotherapy (EoTmono). Analyses focused on 58 angiogenic factors (Human AngiogenesisMAP[®] panel, Myriad RBM) and single-nucleotide polymorphisms (SNPs) in genes implicated in mesothelioma and/or associated with response to anti-angiogenic therapies in other tumour types (VEGFR1, VEGFR3 and mesothelin). Associations of biomarkers with treatment effect were evaluated by Cox regression and tested for interaction with false discovery rate (FDR) adjustment. Adjusted mean changes in angiogenic factor levels were compared between arms by ANCOVA. Analyses were exploratory, limited by small sample size, and considered hypothesis generating.

      Result:
      Of 77 patients with epithelioid tumours, angiogenic factor and genomic data were available for 71 and 67 patients, respectively. PFS/OS benefit of nintedanib appeared potentially more pronounced in patients with baseline plasma endoglin level below the median. There were possible weak associations between major homozygous genotypes for two VEGFR3 SNPs (rs307821 G/G and rs307826 A/A), and OS benefit and between VEGFR1 SNP rs9582036 A/A genotype and PFS benefit. Biomarker treatment associations were limited by small subgroup size, especially for low-frequency SNPs, and interaction tests were not significant after FDR adjustment. Regarding pharmacodynamic effects, adjusted mean change in interleukin-8 levels with nintedanib was greater from baseline to C3mono and lower from C3mono until EoTmono, compared with placebo. Nintedanib showed lower adjusted mean changes versus placebo for VEGFR2 from baseline to C3mono, and for VEGFR2 and VEGFR3 from baseline to EoTmono.

      Conclusion:
      These analyses represent the first biomarker results for nintedanib-treated MPM. While there seemed to be signals for greater PFS and OS improvement in patients with low plasma endoglin and major homozygous VEGFR1/3 genotypes, no biomarkers showed clear significant association with treatment benefit. These findings warrant further evaluation in the Phase III study.

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

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

  • +

    P1.03 - Chemotherapy/Targeted Therapy (ID 689)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Chemotherapy/Targeted Therapy
    • Presentations: 1
    • +

      P1.03-013 - Monitoring of ALK Fusions and Mutations in Advanced ALK Positive Non-Small Cell Lung Cancer (NSCLC) Patients (ID 10208)

      09:30 - 16:00  |  Author(s): David Planchard

      • Abstract

      Background:
      Co-isolated exosomal RNA and cfDNA from plasma can be used for detection of genomic alteration such as EML4-ALK fusion RNA and ALK resistance mutations in NSCLC patients. The clinical utility of this liquid biopsy for response monitoring is under investigation. The aim of this study was to evaluate liquid biopsy as tool for monitoring response to treatment in a prospective cohort of ALK-positive NSCLC patients.

      Method:
      Consecutive ALK positive NSCLC patients treated with systemic therapies in our institution were enrolled. After informed consent, blood samples were prospectively collected for longitudinal analysis during treatment and at progression. Exosomal RNA and cfDNA co-isolated from plasma was used for detection of EML4-ALK fusion RNAs by the qPCR-based ExoDx Lung(ALK)™-test as well as for analysis of ALK-resistance mutations by ExoDx NGS sequencing.

      Result:
      From Aug 2016 to date, 23 patients were enrolled in the study, 14 (61%) were females, 15 (65%) non-smokers, median age of 50 years (23-76). All patients had adenocarcinoma and were tissue positive for ALK by immunohistochemistry 14 (61%) and/or FISH 16 (70%). Nineteen patients (83%) had stage IV disease at diagnosis, with brain involvement in 7 patients (37%), bone in 11 (48%) and liver in 2 (11%). The median number of ALK inhibitors received was 2 (0-4). Twenty-one patients (91%) received ALK inhibitors (5 crizotinib, 3 ceritinib, 13 next-generation inhibitors) and 2 chemotherapy, with an objective response rate of 48%. Five out of 8 patients (63%) that were treatment naïve (baseline) or progressive disease (PD) at the time of collection, were positive for EML4-ALK by liquid biopsy, 1 of 4 samples (25%) at baseline, and 4 of 4 samples (100%) at PD, were positive by liquid biopsy. EML4-ALK variant 1 was detected in two (40%) and variant 3 in three patients (60%). All 26 samples collected during objective response or stable disease (100%) were negative for EML4-ALK by liquid biopsy. The ALK resistance mutation panel was performed on 2 samples from patients with PD, and both were detected positive for ALK resistance mutations, L1196M (variant 1) and G1202R (variant 3), respectively.

      Conclusion:
      The monitoring of ALK fusions on exosomal RNA by liquid biopsy is applicable in the clinic and closely correlated to disease control. ALK mutations detection using liquid biopsy can be an accurate tool for assessing the resistance to ALK inhibitors. Updated results from up to 30 patients will be available for the final presentation.

  • +

    P1.04 - Clinical Design, Statistics and Clinical Trials (ID 690)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Clinical Design, Statistics and Clinical Trials
    • Presentations: 1
    • +

      P1.04-001 - Osimertinib with Ramucirumab or Necitumumab in Advanced T790M-positive EGFR-Mutant NSCLC: Preliminary Ph1 Study Results (ID 7940)

      09:30 - 16:00  |  Author(s): David Planchard

      • Abstract
      • Slides

      Background:
      Combination studies of a first- or second-generation EGFR tyrosine kinase inhibitor (TKI) and either a VEGF or EGFR-targeting monoclonal antibody have recently shown promising clinical results in EGFR-mutant non-small cell lung cancer (NSCLC) patients. The preliminary safety results from the phase 1 study JVDL (NCT02789345), combining third-generation EGFR TKI osimertinib (Osi) with human IgG1 monoclonal antibodies ramucirumab (Ram) or necitumumab (Neci), are reported.

      Method:
      Eligible pts naïve to third-generation EGFR TKI therapy with advanced EGFR T790M-positive NSCLC who progressed after initial EGFR TKI therapy were enrolled. In the dose-finding portion, following a dose de-escalation 3+3 design, patients received daily oral Osi (80 mg) and either 10 mg/kg intravenous (IV) Ram on day 1 (D1) every two weeks (Q2W), or 800 mg (IV) Neci on D1 and D8 Q3W. Primary objective of the study is to assess the safety and tolerability of Ram or Neci combined with Osi, and secondary objectives include preliminary evaluation of efficacy.

      Result:
      As of data cutoff on 09-May-2017, 7 pts were treated in the completed dose-finding portion: 3 pts with Ram+Osi (Arm A) and 4 pts (1 non-evaluable and replaced) with Neci+Osi (Arm B). No DLTs were observed in either arm, and the initial dose level became the recommended dose for expansion cohort. After the DLT observation period was complete, the only Grade ≥3 (Gr≥3) treatment-related adverse event (TRAE) was dermatitis acneiform (Arm B), with one unrelated Gr≥3 treatment-emergent AE (TEAE) of increased lipase (Arm B) and one serious AE of Gr2 diverticulitis (unrelated to study treatment) (Arm A). Expansion cohort A of Ram+Osi is fully enrolled with 22 pts. Safety data were available for 18 out of 22 cohort A patients. Gr≥3 TEAEs were reported in 4 patients, including dyspnea (unrelated [n=1]), decreased appetite (unrelated [n=1]), and hypertension (related [n=2]). Three patients reported serious adverse events (none related to study treatment): Gr3 dyspnea and Gr2 pyrexia, Gr2 dyspnea, and Gr2 urinary tract infection. No death was reported in patients in the dose-finding portion, and one death unrelated to study treatment was reported in the expansion cohort.

      Conclusion:
      No DLTs were observed and no unexpected safety signals were seen to date. The recommended dose for expansion cohort was the initial dose level of 10 mg/kg ramucirumab IV Q2W with oral 80 mg osimertinib. Additional safety and efficacy observation for the combination of Ram+Osi is ongoing, and will be presented at the meeting.

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

  • +

    P1.08 - Locally Advanced NSCLC (ID 694)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Locally Advanced NSCLC
    • Presentations: 1
    • +

      P1.08-009 - Neutrophilia as Prognostic Biomarker in Locally Advanced Stage III Lung Cancer (ID 8920)

      09:30 - 16:00  |  Author(s): David Planchard

      • Abstract

      Background:
      To study the prognostic value of leucocyte disorders in two retrospective cohorts of stage III Non-Small Cell Lung Cancer (NSCLC) patients, and to compare their accuracy with established prognostic markers.

      Method:
      Clinical records of consecutive previously untreated NSCLC patients in our Institution between June 2001 and September 2016 for stage III NSCLC were collected. The prognostic value of pretreatment leucocyte disorders was examined, with focus on patterns of relapse and survival. Leukocytosis and neutrophilia were defined as a leukocyte count or a neutrophil count exceeding 10 and 7 G/L, respectively.

      Result:
      We identified 238 patients (145 patients prospectively registered through MSN study (NCT02105168) with 136 additional patients), displaying baseline leukocytosis or neutrophilia in 39% and 40% respectively. Most were diagnosed with adenocarcinoma (48%), and stage IIIB NSCLC (58%). 3-year actuarial overall survival (OS) and progression-free survival (PFS) were 35% and 27% respectively. Local relapses were reported in 100 patients (42%), and distant metastases in 132 patients (55%). In multivariate analysis, leukocytosis, neutrophilia, and induction chemotherapy regimen based on carboplatin/paclitaxel were associated with worse OS and PFS (p<0.05). Neutrophilia independently decreased Locoregional Control (LRC) (HR=2.5, p<0.001) and Distant Metastasis Control (DMC) (HR=2.1, p<0.001). Neutrophilia was significantly associated with worse brain metastasis control (p=0.004), mostly in adenocarcinoma patients (p<0.001). Figure 1



      Conclusion:
      In stage III NSCLC patients, treated with concurrent cisplatin-based chemoradiation, baseline leukocytosis and neutrophilia predict OS, PFS, LRC, and DMC. In addition with previously available markers, this independent cost-effective biomarker could help to stratify stage III NSCLC population with more accuracy.

  • +

    P2.01 - Advanced NSCLC (ID 618)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Advanced NSCLC
    • Presentations: 1
    • +

      P2.01-038 - Determinants of Frailty and Treatment Toxicity in Non-Small Cell Lung Cancer Patient (ID 9566)

      09:00 - 16:00  |  Author(s): David Planchard

      • Abstract
      • Slides

      Background:
      Platinum-based chemotherapy remains a first line treatment for advanced non-small-cell lung cancers (NSCLC). Despite better individualization of treatment, some patients will seek frequent medical attention because of cancer-related complications or treatment toxicity. This can negatively impact patient’s quality of life and health care resources. This study aimed to identify biological and clinical factors predictive of frailty and treatment toxicity among NSCLC patients eligible for first-line platinum-based chemotherapy.

      Method:
      Using our institutional medical charts, we retrospectively extracted data on stage III and IV NSCLC patients diagnosed between December 2011 and November 2015 who had received a first-line platinum based chemotherapy. The primary outcome is defined as any unplanned emergency visit and/or unplanned hospitalization for cancer or treatment related complications. Using multivariate logistic regression model with step by step method, we defined baseline biological and clinical determinants associated with the primary outcome.

      Result:

      Table 1. First Multivariate Analysis
      Variable OR 95% CI
      Age ≥ 62 Years-old 1.61 0.70 - 3.68
      Adenocarcinoma - Squamous Cell Carcinoma - NSCLC other 1 2.43 0.50 0.61- 9.61 1.45 – 1.74
      Performance scale ≥ 1 1.35 0.57 – 3.18
      Number of metastasis ≥ 2 1.36 0.58 – 3.18
      Pleural metastasis 2.04 0.53 – 7.86
      Weight loss ≥10% or ≥3 kg 1.00 0.41 – 2.43
      ≥ 3 prescription drugs per day 0.98 0.42 – 2.28
      Current smoker - Former Smoker - Never smoker 1 0.56 1.10 0.24 – 1.30 0.24 – 5.11
      Neutrophils count ≥ 7500/ mm[3] 1.57 0.70 – 3.54
      Lymphocytes count ≤ 1000/ mm[3] 1.04 0.34 – 3.22
      Albumin ≤ 35 g/L 2.70 0.93 – 7.69
      LDH ≥ 247 U/L 0.93 0.37 – 2.30
      We included 227 patients. Mean age was 60 years old, 65% were male, 46% current smokers, 10% PS 2-3 and 74% had adenocarcinoma histology. 20,7% patients had locally advanced disease (Stage III) treated by chemoradiation and 78,4% had metastatic disease treated by exclusive chemotherapy. Median overall survival (OS) was 15 months and PFS 6 months. Overall, 55 % (122/227) met the primary outcome. There were 14 variables (Table 1) included in the first multivariate analysis before computer based step by step approach. In the final model (not shown), albumin level <35 g/L (OR 2.24 95% IC 1.14- 4.38, p= 0.02) was an independent predictor of the primary outcome. There was also a trend for squamous cell carcinoma subtype (OR 2.27 95% IC 0.872- 5.914, p= 0.09).

      Conclusion:
      Low albumin level is a determinant of frailty in patients eligible for platinum-based chemotherapy. Early intervention in these subgroups could benefit patient’s quality of life and health care expenses. (Medicoeconomic analysis will be presented).

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

  • +

    P2.07 - Immunology and Immunotherapy (ID 708)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Immunology and Immunotherapy
    • Presentations: 2
    • +

      P2.07-005 - Impact of Baseline Leptomeningeal and Brain Metastases on Immunotherapy Outcomes in Advanced Non-Small Cell Lung Cancer (NSCLC) Patients (ID 7958)

      09:30 - 16:00  |  Author(s): David Planchard

      • Abstract

      Background:
      Central nervous system (CNS) involvement is frequent in NSCLC patients and associated with poor prognosis. However, its impact on immune checkpoints inhibitors’ (ICI) outcomes remains unknown.

      Method:
      We retrospectively collected the clinical and imaging data of a cohort of 271 patients treated with ICI in our institute from Nov. 2012 to April 2017. We analyzed overall survival (OS), progression-free survival (PFS), overall response rate (ORR) and disease control rate (DCR), and CNS outcomes using brain CT scan and/or MRI. Both body and CNS outcomes were assessed prospectively by investigators.

      Result:
      With a median follow up of 17 months (95% IC 15-21), 259 patients were evaluated, 48 (19%) had CNS involvement before immunotherapy; 225 were (87%) smokers, 78% had PS ≤1, with median age of 63.1; 166 (64%) had adenocarcinoma; 67 (26%) were KRASmut, 14 (5%) EGFRmut and 3 (1%) ALK positive. PDL1 was ≥1% by immunohistochemistry in 68 (28%), negative in 28 (11%) and unknown in 163 patients. Median number of prior lines was 1 (0-11). The global ORR was 20%. The median OS was 8 months (95% IC 6-11). No difference was observed in OS between CNS+ vs. CNS- population (p= 0.09). The global ORR was 18% vs. 20%, in CNS+ and CNS- patients, respectively (p=1). To date, CNS–relative data are available for 36 patients: n= 32 brain metastasis, n=7 meningeal carcinomatosis, including 4 cytological positivity, n=2 leptomeningeal and n=1 medullar metastasis. Thirty-one patients (86%) had brain target lesions and 15 were evaluable for CNS outcome (CNS progressive disease (PD) before starting ICI and/or no brain radiation therapy (RT) in the previous 6 months. Median interval between consecutive CNS assessments was 2 months. Twenty-two had CNS PD before immunotherapy: 41% (9/22) received radiation therapy (RT) the month before immunotherapy (4 whole brain RT, 5 stereotactic). No differences were observed according to prior RT, with a median OS of 10 months (95%IC 2-NR) vs. 8 months. (95%IC 5-NR) for prior vs. no prior RT (p=0.79). The median OS for the 7 patients with meningeal carcinomatosis was 2 months (0 to 20). The CNS ORR was 27% (4/15, 3 partial, 1 complete response) and CNS DCR was 60% (9/15). One CNS pseudo progression (7%) and one dissociated brain response (7%) were observed.

      Conclusion:
      CNS involvement did not seem to be associated with a negative impact on immunotherapy outcomes in advanced NSCLC patients. Final analysis of the entire cohort will be presented.

    • +

      P2.07-060 - Response Assessment and Subgroups Analysis According to the Lung Immune Prognostic Index (LIPI) for Immunotherapy in Advanced NSCLC Patients (ID 10179)

      09:30 - 16:00  |  Author(s): David Planchard

      • Abstract

      Background:
      LIPI is a score that combine dNLR (neutrophils/(leucocytes-neutrophils) and lactate dehydrogenase (LDH) and correlate with prognosis of NSCLC patients treated with immune checkpoint inhibitors (ICI). We report the predictive role of LIPI on response and in various subgroups of patients.

      Method:
      Baseline dNLR and LDH were retrospectively collected in 431 patients treated with ICI from Nov. 2012 to Jan. 2017, from 8 European centers. LIPI delineates 3 groups: good (dNLR<3+LDH3 or LDH>ULN), poor (dNLR>3+LDH>ULN). Response rate (RR) and disease control rate (DCR) were assessed according to the investigator’s criteria. The subgroup analysis was performed according to the age, histology, performance status (PS) and PD-L1 status by immunohistochemistry (positivity if ≥ 1% on tumor cells).

      Result:
      With a median follow-up of 12.8 months (m.) [95%CI 11.9-14], 431 patients were included. Baseline characteristics are summarized in table 1. The median overall survival (OS) and progression-free survival (PFS) were 10.5m. [95%CI 9.5-13] and 3.9m. [3-4.4], respectively. The median OS was 4.8m. vs. 10 m. vs. 16.5m., and median PFS was 2m. vs. 3.1m. vs. 5m. for the poor, intermediate and good LIPI groups, respectively (both p<0.0001). LIPI was correlated with response rate (p<0.0001). In multivariate analysis, the intermediate and poor group were associated with progressive disease, with an OR of 2.20 [CI95% 1.26-3.84] p=0.005) and an OR of 3.04 [CI95% 1.46-6.36] p=0.003), respectively. LIPI was correlated with OS, regardless the age (<70 years (p<0.0001) vs. older (p=0.0006) and the histology non-squamous (p<0.0001) vs. squamous (p=0.02). In PS 0-1 and in smoker population, LIPI correlated with OS (both p<0.0001), but not in PS ≥2 (12%) and non-smokers (8%). LIPI was correlated with OS for positivity (p=0.01) and unknown PD-L1 (p=0.0001), but not negativity.

      LIPI 0 Good (N=162, 37%) LIPI 1 Intermediate (N=206, 48%) LIPI 2 Poor (N= 63, 15%) All population cohort N = 431 (%)
      Sex
      Male 102 (63) 131 (64) 42 (67) 275 (64)
      Age at diagnosis
      Median (years, range) 62 (36;86) 63 (29;86) 62 (39;84) 62 (29;86)
      Smoking status
      Non-smoker 13 (8) 18 (9) 5 (8) 36 (8)
      Former 80 (49) 115 (56) 46 (73) 241 (56)
      Current 67 (42) 69 (33) 11 (17) 147 (34)
      Unknown 2 4 1 7
      Histology
      Non-squamous 111 (69) 132 (64) 41 (65) 284 (66)
      Squamous 51 (31) 74 (36) 22 (35) 147 (34)
      Molecular alteration
      EGFR mutation 3 (2) 13 (6) 3 (5) 19 (4)
      ALK rearrangement 2 (1) 2 (1) 1 (2) 5 (1)
      KRAS mutation 34 (21) 31 (15) 8 (13) 73 (17)
      PDL1 status
      Negative 16 (36) 14 (25) 1 (5) 31 (25)
      Positive 28 (64) 43 (75) 20 (95) 91 (75)
      Unknown 118 149 42 337
      Performance Status
      0 51 (32) 45 (22) 10 (16) 106 (25)
      1 96 (60) 132 (64) 42 (67) 270 (63)
      ≥ 2 12 (8) 28 (14) 11 (17) 51 (12)
      Stage at diagnosis
      IIIb 18 (11) 33 (16) 14 (22) 65 (15)
      IV 101 (62) 135 (66) 38 (60) 274 (64)
      Metastases sites
      Median (Range) 2 (0;6) 2 (0;7) 2 (1;7) 2 (0-7)
      Bone 43 (27) 58 (28) 20 (32) 121 (28)
      Liver 28 (17) 39 (19) 16 (25) 83 (19)
      Brain 22 (14) 19 (9) 9 (14) 50 (12)
      Immunotherapy
      PD1 inhibitor 133 (82) 167 (81) 48 (76) 348 (81)
      PDL1 inhibitor 19 (12) 34 (17) 12 (19) 65 (15)
      PDL1 inhibitor- CTLA4 inhibitor 10 (6) 5 (2) 3 (5) 18 (4)
      Immunotherapy line
      Median (Range) 2 (1;11) 2 (1;12) 2 (1;8) 2 (1-12)
      Response rate
      Complete response (CR) 6 (4) 3 (1) 0 (0) 8 (2)
      Partial response (PR) 42 (26) 53 (26) 18 (28) 113 (26)
      Stable disease (SD) 66 (41) 59 (29) 8 (13) 133 (31)
      Progression 40 (25) 81 (39) 33 (52) 154 (36)
      NA 8 10 4 25
      Dissociated response 14 (9) 15 (7) 2 (3) 31 (7)


      Conclusion:
      Baseline LIPI predicts response to ICI, and was correlated with OS regardless of age and histology.