Virtual Library

Start Your Search

Keunchil Park



Author of

  • +

    ES02 - Management of Oncogene Addicted Patients with Stage III NSCLC (ID 5)

    • Event: WCLC 2019
    • Type: Educational Session
    • Track: Treatment of Locoregional Disease - NSCLC
    • Presentations: 1
    • Now Available
    • +

      ES02.02 - Management of EGFR LA Disease (Now Available) (ID 3156)

      10:30 - 12:00  |  Presenting Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Abstract

      Management of EGFR mutation(+) Locally Advanced Non-Small Cell Lung Cancer

      Keunchil Park, MD, PhD

      Division of Hematology-Oncology, Samsung Medical Center,

      Sungkyunkwan University School of Medicine, Seoul, Korea

      The standard of care for the locally advanced non-small cell lung cancer(LA-NSCLC) is concurrent chemoradiotherapy(CCRT). Recently, the addition of consolidation immune checkpoint inhibitor following CCRT demonstrated improved outcome and is now recommended as the new standard of care for this heterogeneous group of LA-NSCLC patients. In advanced/metastatic setting EGFR mutation (+), NSCLC cancer patients define a unique subset with a dramatic response to the EGFR TKIs. Currently, molecular profiling of tumor tissue for EGFR mutations (as part of multiplex testing) is a routine procedure at the time of initial diagnosis of patients with recurrent or metastatic NSCLC. For advanced or metastatic EGFR mutant NSCLC patients EGFR TKI therapy is the treatment of choice and the median overall survival is > 30 months.

      Despite the great success of EGFR TKI in metastatic NSCLC, the role of EGFR TKIs in LA-NSCLC is less well defined and controversial. Early clinical trials in unselected NSCLC patients failed to demonstrate the benefits of EGFR-TKIs as an adjuvant treatment. The first prospective randomized phase III trial to investigate the role of EGFR TKI in earlier stage NSCLC includes the SWOG 0023 study (J Clin Oncol 2008;26:2450-6) which failed to support the role of maintenance gefitinib (versus placebo) following definitive chemoradiation in unresectable stage III NSCLC. The CALGB 30106 phase II study also failed to show the benefits of adding gefitinib to sequential or concurrent chemoradiotherapy in unresectable stage III NSCLC (J Thorac Oncol 2010;5:1382-90). And the randomized prospective placebo-controlled adjuvant gefitinib trial in unselected patients with stage IB–IIIA resected disease (BR.19) was prematurely closed (J Clin Oncol 2013;31:3320-6). Another randomized double-blind trial in adjuvant NSCLC with tarceva (RADIANT) in patients with completely resected state IB to IIIA NSCLC whose tumors expressed EGFR protein by immunohistochemistry or EGFR amplification by fluorescence in situ hybridization did not show prolonged DFS (J Clin Oncol 2015;33:4007-14).

      The prognostic or predictive value of EGFR mutation in early stage NSCLC patients is not well defined. In a retrospective single institutional analysis (J Thorac Oncol. 2012;7: 1815–1822), patients with resected stage I–III lung cancers and EGFR mutation have a lower risk of death compared to patients without EGFR mutation. There was a trend toward improvement in DFS among individuals with resected stages I to III lung adenocarcinomas harboring mutations in EGFR exon 19 or 21 who received adjuvant EGFR TKI therapy (J Thorac Oncol 2011;6:569–575).

      There have been recently reported several prospective trials of adjuvant EGFR TKI in early stage NSCLC patients enriched with EGFR mutation.

      CTONG1104 study (ADJUVANT), a randomized, open-label, phase III trial of adjuvant gefitinib for 24 months versus intravenous vinorelbine plus cisplatin for 4 cycles in patients with completely resected (R0), stage II–IIIA (N1–N2), EGFR-mutant (exon 19 deletion or exon 21 Leu858Arg) NSCLC, demonstrated that adjuvant gefitinib compared to cisplatin-based chemotherapy significantly increases disease-free survival, diminishes toxic effects, and improves HRQoL in patients with completely resected stage II–IIIA EGFR-mutant NSCLC (Lancet Oncol 2018; 19: 139–48).

      In the phase II SELECT trial, adjuvant erlotinib for 2 years in patients with resected stage IA to IIIA EGFR-mutant NSCLC after standard adjuvant chemotherapy with or without radiotherapy (J Clin Oncol 2019;37:97-104) showed an improved 2-year DFS. Patients rechallenged with erlotinib after recurrence experienced durable benefits.

      There are also some neoadjuvant trials of EGFR TKI for locally advanced NSCLC.

      RTOG 1306, a randomized phase II study of individualized combined modality therapy for stage III NSCLC (12-weeks of either erlotinib hydrochloride or crizotinib followed by chemoradiation therapy in stage III NSCLC with EGFR TK mutations or EML4- ALK fusion). The primary objective was to assess whether patients with unresectable LA-NSCLC treated with EGFR or ALK TK targeted agents based on molecular characteristics have a longer progression-free survival than those treated with standard care therapy alone. The study was, however, unfortunately prematurely terminated due to poor accrual (NCT01822496).

      CTONG 1103 is an open-label, randomized trial that compared the efficacy and safety of erlotinib versus gemcitabine plus cisplatin neoadjuvant therapy in patients with exon 19 or 21 EGFR mutations and untreated resectable stage IIIA-N2 NSCLC. The study did not meet the primary end point of ORR with 42 days of neoadjuvant erlotinib, but the secondary end point PFS was significantly improved (J Clin Oncol 37. © 2019; published at jco.org on June 13, 2019: DOI https://doi.org/10.1200/JCO.19.00075)

      There are also several ongoing clinical trials of EGFR TKIs in resected stage IB - IIIA NSCLC with activating EGFR mutations.

      The Adjuvant Lung Cancer Enrichment Marker Identification and Sequencing Trial (ALCHEMIST) is a prospective randomized double-blind placebo-controlled trial to investigate the benefits of the addition of molecularly targeted agents after standard postoperative chemotherapy in patients with resected NSCLC (Clin Cancer Res:2015; 21(24); 5439–44). It is worth noting that the primary objective for the ALCHEMIST adjuvant trials is overall survival (OS).

      ADAURA, a randomized phase III trial (osimertinib vs. placebo in patients with stage IB-IIIA NSCLC, following complete tumor resection with or without adjuvant chemotherapy; NCT02511106) has been designed to assess the efficacy and safety of adjuvant osimertinib versus placebo in patients with resected stage IB-IIIA EGFR mutation-positive (Ex19Del or L858R) NSCLC. The primary efficacy objective is DFS.

      In brief, many neoadjuvant/adjuvant EGFR TKI trials in earlier stage of NSCLC have demonstrated its safety and feasibility with an improved DFS esp. in EGFR mutation(+) LA-NSCLC. However, the real question should be “Can we improve the ‘overall survival’ and thus ultimately the ‘cure rate’ of locally advanced NSCLC with EGFR mutation?” In order to address this issue, there remain several critical questions to be answered, e.g., who is most likely to benefit from adjuvant EGFR TKI, what is the optimal duration of adjuvant TKI, what is the best regimen, etc. Whether the earlier introduction of EGFR targeted therapy in less advanced NSCLC would lead to improved ‘Cure Rate’ remains to be seen in future prospective trials in a larger number of EGFR mutation(+) patients.

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

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

  • +

    MA08 - Pawing the Way to Improve Outcomes in Stage III NSCLC (ID 127)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Treatment of Locoregional Disease - NSCLC
    • Presentations: 1
    • Now Available
    • +

      MA08.03 - Adjuvant Pembrolizumab in N2 Positive NSCLC Treated with Concurrent Chemoradiotherapy Followed by Surgery: Phase II, Prospective Study (Now Available) (ID 1744)

      15:15 - 16:45  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      The standard treatment option for stage IIIA-N2 subgroup is still under discussion with controversies. We hypothesize that immune checkpoint inhibitor consolidation therapy could have an additional role in prolongation of the disease-free survival (DFS) for stage IIIA-N2 NSCLC treated with tri-modalities therapy.

      Method

      This is a phase 2 study evaluating the clinical efficacy of pembrolizumab treatment after CCRT with curative resection in stage IIIA-N2 NSCLC pts (NCT03053856). Pathologically confirmed pts were treated with five cycles of CCRT, weekly paclitaxel (50mg/m2) and cisplatin (25mg/m2) combined with radiotherapy (total of 44Gy over 22 fractions) followed by curative resection. Adjuvant Pembrolizumab (200mg fixed dose) is applied every three weeks up to 2 years or until disease recurrence. The primary objective is disease-free survival of more than 20 months. The first patient was recruited in October 2017, and the data for this abstract was locked at 20th of January, 2019.

      Result

      Total of 40 pts were screened, and 37 pts received treatment. Median age was 64 years (range 39-74), and twenty-three pts were male (62.2%). As a curative surgery, pts received lobectomy (n=34), bi-lobectomy (n=2), or pneumonectomy (n=1). Adenocarcinoma was predominant (n=27, 73.0%). After the neoadjuvant CCRT, down-staging were observed in nine pts (24.3%). The median follow-up duration was 10.6 months (range 3.1-17.2), and pts received a median of 11 cycles (range 1-22) of adjuvant pembrolizumab. DFS is not reached. Fourteen patients discontinued treatment due to disease progression (n=9), adverse events (n=4) and withdraw consent (n=1). There was a case of grade 4 pneumonitis and a case of grade 3 autoimmune hepatitis which lead to discontinuation of the treatment. Otherwise, grade 1-2 hypothyroidism (n=6), pneumonitis (n=5), skin rash (n=3) were observed. Patients with severe immune-related adverse event showed a significantly high percentage of Ki-67 + cells among CD8 T-cells in peripheral blood.

      Conclusion

      This study is the first study to demonstrate the feasibility of adjuvant pembrolizumab monotherapy in stage IIIA-N2 patients. Updated clinical outcome will be presented at the conference.

      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.

  • +

    MA09 - EGFR & MET (ID 128)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Targeted Therapy
    • Presentations: 1
    • Now Available
    • +

      MA09.09 - Long-Term Outcomes to Tepotinib Plus Gefitinib in Patients with <i>EGFR</i>-Mutant NSCLC and MET Dysregulation: 18‑Month Follow-Up (Now Available) (ID 1783)

      15:15 - 16:45  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      In EGFR-mutant NSCLC, MET amplification may cause resistance to EGFR tyrosine kinase inhibitors (TKIs). In a Phase Ib/II study in EGFR TKI-resistant patients with EGFR-mutant MET+ NSCLC, progression-free survival (PFS) and objective response rate (ORR) after ≥6 months of follow-up were improved with tepotinib (a highly selective MET TKI) plus gefitinib, compared with chemotherapy, particularly in patients with MET amplification. Here we present data at ≥18 months of follow-up.

      Method

      Asian patients with advanced, EGFR+, T790M-, MET+ NSCLC with resistance to prior EGFR TKIs were randomized to receive oral tepotinib 500 mg/day+gefitinib 250 mg/day or ≤6 cycles of cisplatin/carboplatin+pemetrexed chemotherapy±pemetrexed maintenance until confirmed progression, unacceptable toxicity, or withdrawal. Primary endpoint was investigator-assessed PFS. Secondary endpoints included ORR, overall survival (OS) and safety. Subgroup analyses were preplanned in MET IHC3+ and MET amplification populations (NCT01982955).

      Result

      Low recruitment halted full enrolment with 55 of 156 planned patients enrolled.

      As of 12-Dec-2018, median (range) duration of treatment with tepotinib+gefitinib was 21.4 (4.6, 110.9) weeks, with 3 patients still receiving treatment; and with pemetrexed was 18.0 (3.0, 60.4) weeks. 15 patients (62.5%) received ≥4 cisplatin/carboplatin cycles.

      Better outcomes were reported with tepotinib+gefitinib vs chemotherapy (Table), particularly in patients with MET IHC3+ (PFS: HR 0.35 [90% CI 0.17–0.74], OS: 0.32 [0.14–0.75]) or MET amplification (PFS: HR 0.13 [90% CI 0.04–0.43], OS: 0.08 [0.01–0.51]).

      Drug-related grade ≥3 adverse events (AEs) occurred in 17 (54.8%) patients receiving tepotinib+gefitinib and 12 (52.2%) patients receiving chemotherapy. Any-cause AEs leading to discontinuation occurred in 3 (9.7%) patients receiving tepotinib+gefitinib and 1 (4.3%) receiving chemotherapy. Dose reductions due to AEs were reported in 5 (16.1%) vs 4 (17.4%) patients.

      Conclusion

      Tepotinib+gefitinib has durable antitumor activity in patients with EGFR-mutant NSCLC with MET IHC3+ or MET amplification, and was generally well tolerated. MET amplification will be further explored as a biomarker for tepotinib.

      Table: Summary of efficacy data

      Population

      Tepotinib + gefitinib

      Chemotherapy

      HR/OR
      (90% CI)

      Overall MET+*

      Patients, n

      31

      24

      mPFS, months (90% CI)

      4.9 (3.9, 6.9)

      4.4 (4.2, 6.8)

      0.67 (0.35, 1.28)

      mOS, months (90% CI)

      17.3 (12.1, 37.3)

      18.7 (15.9, 20.7)

      0.67 (0.33, 1.37)

      ORR, n (%) [90% CI]

      14 (45.2) [29.7, 61.3]

      8 (33.3) [17.8, 52.1]

      1.99 (0.56, 6.87)

      MET IHC3+

      Patients, n

      19

      15

      mPFS, months (90% CI)

      8.3 (4.1, 21.2)

      4.4 (4.1, 6.8)

      0.35 (0.17, 0.74)

      mOS, months (90% CI)

      37.3 (24.2, 37.3)

      17.9 (12.0, 20.7)

      0.32 (0.14, 0.78)

      ORR, n (%) [90% CI]

      13 (68.4) [47.0, 85.3]

      5 (33.3) [14.2, 57.7]

      4.33 (1.03, 18.33)

      MET amplification

      Patients, n

      12

      7

      mPFS, months (90% CI)

      21.2 (8.3, NE)

      4.2 (1.4, 7.0)

      0.13 (0.04, 0.43)

      mOS, months (90% CI)

      37.3 (NE, NE)

      13.1 (3.3, NE)

      0.08 (0.01, 0.51)

      ORR, n (%) [90% CI]

      8 (66.7) [39.1, 87.7]

      3 (42.9) [12.9, 77.5]

      2.67 (0.37, 19.56)

      CEP-7, centromere protein 7; CI, confidence interval; EGFR, epidermal growth factor receptor; GCN, gene copy number; HR, hazard ratio; IHC, immunohistochemistry; IRC, independent review committee; ITT, intention to treat; MET, mesenchymal-epithelial transition factor; NE, not estimable; OR, odds ratio; ORR, objective response rate; OS, overall survival; PFS, progression-free survival

      All efficacy outcomes are investigator-assessed by RECIST v1.1.

      *IHC2+/IHC3+/gene amplification.

      MET amplification is defined as GCN ≥5 and/or MET/CEP-7 ratio ≥2. 17 of 19 patients with MET amplification have MET overexpression (IHC3+).

      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.

  • +

    MA19 - Looking at PROs in Greater Detail - What Patients Actually Want and Expect (ID 147)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Treatment in the Real World - Support, Survivorship, Systems Research
    • Presentations: 1
    • Now Available
    • +

      MA19.06 - Successful Development of Realtime Automatically Updated Data Warehouse in Health Care (ROOT-S) (Now Available) (ID 584)

      11:30 - 13:00  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      Clinical information is often not recorded in an organized way, and converting it to a structured format can be a time-consuming task that may not successfully capture all facets of the information. Clinical Data Warehouse is a real time database that consolidates data from a variety of clinical sources to present a unified view. However, the clinical data extracted from the CDW have not only structured data (SD) but also natural language (NP) generated during clinical practice, and there is a limitation that it is difficult to apply to clinical trials because it is not structured and formatted to find key-point contents. This study aims at developing a systematic and comprehensive cohort through an automatic real-time update system called CDW.

      Method

      The aim of this study was to evaluate clinical data of non-small cell lung cancer, small cell lung cancer, head and neck cancer, thymic cancer, and mesothelioma. In this study, we developed a unique algorithm that is optimized for each disease category using comprehensive natural language processing (NLP) systems and structured information from unstructured free text and structured data capture (SDC). We developed an algorithm using clinical information of patients diagnosed and treated during the past 10 years and designated validation sets of patients diagnosed and treated in 2018 for validation that these algorithms work automatically.

      Result

      We collected clinical data of 23,735 NSCLC patients, 2,077 SCLC patients, 5,032 head and neck cancer patients, 3,948 esophageal cancer patients, 747 thymic cancer patients and 138 mesothelioma patients diagnosed at Samsung Medical Center. We could demonstrate using the validation set that the program accurately extracts the data needed for the cohort of each cancer. The program is updated automatically every 24 hours, the source of each data is indicated separately, and the data that need to be integrated is transformed and systematically organized. The biggest advantage is that the scattered information is systematically integrated and automatically buildup to match the patient's cohort, so you can capture most updated survival or test results or treatment outcomes almost in real time. Data on the development of this program will be presented.

      Conclusion

      This study is the first study that successfully developed and validated real-time updated cohort using CDW. This study suggests a blueprint for constructing a big data -based cohort for clinical research and is expected to be a landmark trial. The detailed analysis of each cancer through the development of the program will be presented.

      wclc.png

      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.

  • +

    MA21 - Non EGFR/MET Targeted Therapies (ID 153)

    • Event: WCLC 2019
    • Type: Mini Oral Session
    • Track: Targeted Therapy
    • Presentations: 1
    • Now Available
    • +

      MA21.10 - Phase II Study of 160mg of Osimertinib in EGFR T790M Positive NSCLC with Brain or Leptomeningeal Metastases Who Progressed on Prior EGFR TKI (Now Available) (ID 1705)

      14:30 - 16:00  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      EGFR tyrosine kinase inhibitor (TKI) has successfully improved clinical outcome in non-small cell lung cancer (NSCLC) with activating EGFR mutation. However, up to 40% of TKI treated patients present with disease progression in the central nerve system (CNS) either as brain metastases (BM) or leptomeningeal metastases (LM). Osimertinib is a 3rd generation EGFR TKI effective in T790M mutant NSCLC and characterized by high blood-brain barrier penetration. In this phase II, multicenter prospective single-arm two cohort study, the clinical efficacy of 160mg of osimertinib in T790M mutant BM or LM patients progressed on prior EGFR TKI was evaluated. (NCT0325712)

      Method

      BM only patients were included in the BM cohort (n=40). Patients with cerebrospinal cytology confirmed LM with or without BM were included in the LM cohort (n=40). 3rd generation TKI, including 80mg of osimertinib, was exposed to 18 patients in BM and 16 patients in LM cohort. T790M need to be identified from either tissue, plasma or cerebrospinal fluid. The primary endpoint was overall response rate (ORR) (H1=30%) for BM cohort and overall survival (OS) (H1=5months) for LM cohort, respectively.

      Result

      Median follow-up duration was 7.9 months for BM and 8.3 months for LM cohort. In BM cohort, median progression-free survival (PFS) was 7.3 months (95% confidential interval [CI] 3.6-13.7), and median OS was not reached (NR). Intracranial ORR and disease control rate (DCR) was 40.0% and 77.5%. Extracranial ORR and DCR was 30.0% and 67.5%. In LM cohort, median PFS was 8.9 months (95%CI 5.6-NR) and median OS was 13.2 months (95%CI 8.0-NR). When response of leptomeningeal lesion is separately evaluated, CR rate was 25.0% (n=10) and non-CR/non-PR rate was 65.0% (n=26). Extracranial ORR and DCR was 22.5% and 85.0%. Intracranial median PFS was not reached in both BM and LM cohort. Grade 3 adverse event (AE) was observed in 7 BM and 11 LM patients. Four patients required dose reduction due to AE. Among the patients who previously received 3rd generation TKI, 33.3% (6 out of 18) in BM cohort and 81.2% (13 out of 16) in LM cohort showed an intracranial DCR to 160mg of osimertinib. Extended survival analyses and exploratory outcomes will be presented at the conference.

      Conclusion

      In this study, 160mg of osimertinib demonstrated promising ORR and survival benefit with tolerable safety profile in EGFR T790M positive NSCLC patients with CNS metastasis who progressed on prior EGFR TKI.

      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.

  • +

    OA14 - Update of Phase 3 Trials and the Role of HPD (ID 148)

    • Event: WCLC 2019
    • Type: Oral Session
    • Track: Immuno-oncology
    • Presentations: 1
    • Now Available
    • +

      OA14.07 - Clinical and Genetic Characterization of Hyperprogression Based on Volumetry in Advanced NSCLC Treated with Immunotherapy (Now Available) (ID 1067)

      11:30 - 13:00  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      Hyperprogressive disease (HPD), characterized by accelerated tumor progression, has been proposed as a new pattern of progression following immune checkpoint inhibitor (ICI) treatment. The aim of this study was to describe the characteristics of HPD and investigate its predictive markers.

      Method

      Clinical and radiological findings of 335 advanced non-small cell lung cancer (NSCLC) patients treated with ICI monotherapy were retrospectively analyzed. Radiological data were quantitatively and longitudinally analyzed for tumor size and volume by comparing baseline and follow-up computerized tomography results. The findings were matched to individual genomic profiles generated by deep sequencing of 380 genes.

      Result

      Among 135 patients with progressive disease (PD), as assessed by RECIST, 48 (14·3% of total and 35·6% among PD) and 44 (13·1% of total and 32·6% among PD) were found to have HPD by volumetric (HPDV) and one-dimensional (HPDR) analysis, respectively. HPDV patients were associated with significantly inferior overall survival (OS) compared with non-HPDV PD patients (median OS (months), 4·7 [95% confidence interval (CI), 3·5–11·9)] vs. 7·9 [95% CI, 6·0–13·5], p=0·004); OS did not differ between HPDR and non-HPDR patients. HPDV status was an independent OS factor. Derived neutrophil-to-lymphocyte ratio (dNLR) greater than 4 and lactate dehydrogenase (LDH) greater than the upper normal limit were significantly associated with HPDV. Moreover, we identified coinciding KRAS and STK11 mutations in the HPDV cohort (3/16), while none were found in the non-HPDV cohort (0/28).

      Conclusion

      Defining HPD treated with ICI based on volumetric measurement is more precise than that based on one-dimensional analysis. Pre-ICI dNLR, LDH, and concurrence of STK11 and KRAS mutations could, thus, be used as potential biomarkers for HPD prediction.

      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.

  • +

    P1.01 - Advanced NSCLC (ID 158)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Advanced NSCLC
    • Presentations: 1
    • Moderators:
    • Coordinates: 9/08/2019, 09:45 - 18:00, Exhibit Hall
    • +

      P1.01-97 - Modified RANO-LM Criteria to Evaluate the Radiological Response of Osimertinib in EGFR T790M Positive NSCLC with Leptomeningeal Metastases (ID 1719)

      09:45 - 18:00  |  Author(s): Keunchil Park

      • Abstract

      Background

      There is no standardized method in tracking the clinical response of leptomeningeal (LM) disease. Based on the recently proposed RANO criteria for LM, we made a further modification which could apply to the clinical trials and real clinical setting. In this study, the feasibility of modified RANO-LM criteria is tested in the exploratory dataset from our prospective clinical trials conducted in EGFR T790M positive non-small cell lung cancer patients with LM (NCT0325712).

      Method

      Based on the previous RANO-LM criteria, up to five leptomeningeal nodules, leptomeningeal enhancement and cranial nerve (CN) enhancement from the baseline was recorded and then changes were scored between -3 to 3 at each post-baseline time point of brain MRI imaging using modified RANO-LM criteria. According to the pre-defined scoring system (figure), all LM lesions were categorized as complete response (CR), partial response (PR), stable disease (SD), or progressive disease (PD). Presence of hydrocephalus or parenchymal lesion is seperately assessed as non-LM lesion

      abstract_figure.png

      Result

      Response of osimertinib in LM was evaluated in 34 patients. Baseline MRI identified LM enhancement in 67.6% (n=23), CN enhancement in 55.9% (n=19), hydrocephalus in 35.3% (n=12) and parenchymal metastases in 79.4% (n=27) but no nodular lesion (n=0). LM specific overall response rate was 63.9% by showing CR (n=10, 29.4%), PR (n=13, 38.3%), SD (n=8, 23.5%), PD (n=1, 2.9%) in evaluable patients. Eight patients showed treatment resistance by showing disease aggravation in LM lesion (n=2), non-LM lesion (n=5) and both (n=1).

      Conclusion

      Modified RANO-LM criteria seems to be effective in evaluating treatment response and disease progression in patients with LM. Despite the complexity of the scoring system, modified RANO-LM criteria is a feasible method to evaluate LM status which could be applicable both to the clinical trials and to daily clinical practice. Further validation in independent patient cohort will be performed.

  • +

    P1.04 - Immuno-oncology (ID 164)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Immuno-oncology
    • Presentations: 2
    • Moderators:
    • Coordinates: 9/08/2019, 09:45 - 18:00, Exhibit Hall
    • +

      P1.04-06 - Tumor Microenvironment Landscape in Lung Adenocarcinoma by Single-Cell Sequencing (ID 1227)

      09:45 - 18:00  |  Author(s): Keunchil Park

      • Abstract

      Background

      Lung cancer is the leading cause of cancer cell death in worldwide. Failure of early detection, high recurrence rate and metastasis all contribute to the low survival rate in this detrimental disease. In particular, frequent brain metastasis confers an imperative challenge in the management of lung cancer. Single-cell RNA sequencing provides specific profiling of cell populations at the single-cell level. Interpretation of single-cell transcriptome data for the discovery of therapeutic targets and prognostic biomarkers is an ongoing challenge in precision cancer medicine. Especially, the influence of immune microenvironment in tumor has been accepted as a key factor for determining the therapeutic outcome. Despite the diversity of immune infiltrates in lung adenocarcinoma, single-cell RNA sequencing has not yet been applied for a large-scale tumor and immune profiling.

      Method

      Samples were obtained from the primary tumor, lymph node or brain metastases, and pleural fluids of 44 patients with lung adenocarcinoma. Following lung tumor resection, normal lung tissues from a distal region as well as normal lymph nodes were collected for comparison. We dissociated the whole tissues into single cell suspension and then performed scRNA-seq using droplet-based 10x Genomics Chromiunm platform.

      Result

      After quality filtering, we cataloged a total of 208,506 cells into 9 distinct cell lineages annotated by expression of known marker genes. We identified epithelial cells including cancer cells, stromal cells (fibroblasts and endothelial cells), immune cells (T, NK, B, myeloid, and MAST cells) as common cell types, and oligodendrocytes uniquely from brain metastases. The most abundant immune cell populations at the lung tumor site were T lymphocytes and myeloid cells. Notably, primary tumor and lymph node metastases showed substantial differences in the immune cell composition. These differences reflected the original tissue microenvironment, grossly altered by tumor progression and invasion. Therefore, our lung adenocarcinoma atlas illustrates the dynamic cellular landscape during cancer progression which may reveal progression associated changes for each cellular component in unprecedented scale and manner.

      Conclusion

      These results provide specific tumor microenvironmental patterns in lung adenocarcinoma. Thus, single cell-level transcriptome profiles provide clues to expand therapeutic windows into the combination therapy with immune and anti-cancer reagents.

    • +

      P1.04-24 - Circulating Suppressive Immune Cells Predict the Efficacy of Anti PD-1 Immunotherapy in Patients with Advanced Non-Small Cell Lung Cancer (ID 466)

      09:45 - 18:00  |  Author(s): Keunchil Park

      • Abstract

      Background

      The major suppressive immune cells in tumor sites are myeloid derived suppressor cell (MDSC), tumor associated macrophage (TAM), and regulatory T (Treg) cell, and the major roles of these suppressive immune cells include hindering T cell activities and supporting tumor progression and survival. In this study, we analyzed the pattern of various circulating suppressive immune cells in patients with non-small cell lung cancer (NSCLC) to see whether those suppressive immune cells hinder T cell activities leading to poor clinical outcomes.

      Method

      Baseline blood samples were collected from stage I to IV NSCLC patients (n=59), and baseline and one week after the therapy paired blood samples from stage IIIB to IV NSCLC patients (n=83) undergoing anti PD-1 immunotherapy either pembrolizumab or nivolumab. The efficacies of peripheral blood suppressive immune cells along with CD39+CD8+ T cells individually or collectively in anti-PD-1 immunotherapy were evaluated using flow cytometry and T cell suppressive assay.

      Result

      G-MDSCs, M-MDSCs, TAMs, Treg cells, and CD39+CD8+ T cells increased according to NSCLC stage, and MDSCs effectively suppressed T cell activities and induced T cell exhaustion ex vivo. Further, the analysis of 83 NSCLC patients treated with anti-PD-1 immunotherapy demonstrated that low G-MDSCs (PPFS = 0.03, Pos = 0.04), M-MDSCs (PPFS = 0.04, Pos = 0.005), TAMs (PPFS = 0.007, Pos = 0.01), and CD39+CD8+ T cells (PPFS = 0.57, Pos = 0.02) were associated with longer progression-free survival (PFS) and overall survival (OS) compared with high groups. When we performed combined analysis of three suppressive immune cells, G-MDSCs, M-MDSCs, and TAMs collectively, patients who had low frequency of all three suppressive immune cells showed more prominent difference of PFS (6.7 months vs 2 months; P = 0.006), OS (8.5 months vs 4.2 months; P = 0.004), and response rate (94.5% vs 50%) compared to patients with high levels of all three suppressive immune cells. We further sorted patients with all suppressive immune cells plus CD39+CD8+ T cells low and high. Again, PFS (6.1 months vs 0.8 months; P = 0.006), OS (11 months vs 2.4 months; P = 0.01), and response rate (85.7% vs 16.7%) of all suppressive immune cells low and CD39+ CD8+ T cells low group were significantly increased.

      Conclusion

      The analysis of 83 advanced NSCLC patients treated with anti-PD-1 immunotherapy demonstrated that G-MDSC, M-MDSC, TAM and CD39+CD8+ T cell frequencies in peripheral blood individually and collectively might be useful as potential predictive biomarkers.

  • +

    P2.01 - Advanced NSCLC (ID 159)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Advanced NSCLC
    • Presentations: 1
    • Moderators:
    • Coordinates: 9/09/2019, 10:15 - 18:15, Exhibit Hall
    • +

      P2.01-07 - Open-Label, Biomarker-Directed Platform Study in NSCLC Patients Who Progressed on an Anti-PD-(L)1-Containing Therapy (HUDSON) (ID 643)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract
      • Slides

      Background

      Immune checkpoint inhibitor (ICI)-containing regimens have significantly improved survival outcomes in first- and second-line non-small cell lung cancer (NSCLC). However, few patients have-durable responses to anti-programmed cell death‑1/programmed cell death-ligand 1 (anti-PD-[L]1)-containing therapy (primary resistance) or other patients progress during anti-PD-(L)1-containing therapy (acquired resistance). HUDSON addresses the urgent need to identify new treatments and understand ICI resistance for patients who progressed after receiving anti-PD-(L)1-containing therapy.

      Method

      HUDSON is a multi-centre, international, multi-arm, platform study (NCT03334617), which will 1) evaluate therapies to reverse ICI resistance and 2) define mechanisms of ICI resistance in patients with NSCLC who have progressed following standard-of-care platinum- and ICI-based therapies. HUDSON consists of biomarker matched and non-matched groups (Figure). Allocation is guided by tumour molecular profile, using a pre-specified algorithm. Pre-existing local next generation sequence (NGS) data enables rapid patient allocation to biomarker-matched groups. Central molecular profiling comprises NGS and immunohistochemistry data. New groups will be added as new translational hypotheses emerge. Translational research will employ serial peripheral blood samples (including ctDNA) and tumour biopsies.

      Figure. Study design and biomarker prevalence

      wclc 2019 abstract figure.jpg

      Result

      Enrolment is ongoing; as of 01 April 2019, patients have been dosed in each of the drug combinations currently open for recruitment. Analyses of tissue and blood samples collected for exploratory research are ongoing, including genomic, transcriptomic and chemistry biomarkers such as tumour mutation burden, human leukocyte antigen status, T-cell receptor repertoire, and peripheral immune activation signatures.

      Conclusion

      Specific differences between patients on individual HUDSON arms that inform anti-PD(L)1 resistance mechanisms, plus learnings from the implementation of this innovative and complex platform study 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.

  • +

    P2.09 - Pathology (ID 174)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Pathology
    • Presentations: 1
    • Moderators:
    • Coordinates: 9/09/2019, 10:15 - 18:15, Exhibit Hall
    • +

      P2.09-07 - Multiple Immunohistochemistry of Non-Small Cell Lung Cancers Reveals Distinct Immune Context (ID 1232)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract

      Background

      Immune checkpoint blockade improves survival in a subset of patients with non-small cell lung cancer (NSCLC). Though increased tumor infiltrating lymphocytes (TILs) correlate with better outcome in many human cancers, biomarkers that predict response to PD-1 pathway inhibitors remain largely unknown. Therefore, a comprehensive evaluation of the composition and distribution of TILs is necessary to demonstrate their roles.

      Method

      To determine the predictive significance of specific immune cells in the tumor microenvironment, we used multiples IHC. Multiplex IHC is a powerful investigative tool which provides objective quantitative data describing the tumor immune context in both immune subset number and location. To evaluate the immune context broadly we used the OPAL staining panel contains CD8, CD4, CD20, CD68, FOXP3, and panCK.

      Result

      Pathologic tumor specimens from 101 patients with recurred or metastatic NSCLC were analyzed. Tumors exhibited a high degree of heterogeneity in the immune infiltrate and there was no significant difference when immune infiltrates were compared by tumor histology. Furthermore, there were no significant immune cell differences in EGFR mutant tumors or tumors that did not harbor mutations in adenocarcinoma. We observed increased Tregs (CD4+/FOXP3+) in PD-L1-positive tumors as compared with PD-L1-negative tumors. Our results showed that the infiltration levels of most T-cell subpopulations within tumor did not significantly associated with survival. However, high infiltration of B cells significantly correlated with overall survival.

      Conclusion

      Increased infiltration of B cells into tumor regions is an independent predictor of better overall survival in NSCLC. These findings suggest that tumor-infiltrating B cells could act as a clinical factor in anti-PD-1 immunotherapy for NSCLC.

  • +

    P2.12 - Small Cell Lung Cancer/NET (ID 180)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Small Cell Lung Cancer/NET
    • Presentations: 1
    • Moderators:
    • Coordinates: 9/09/2019, 10:15 - 18:15, Exhibit Hall
    • +

      P2.12-14 - A Pilot Study of Serial Plasma Metabolomics in Small Cell Lung Cancer Patients (ID 1623)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract

      Background

      Small cell lung cancer (SCLC) is among the deadliest cancers. SCLC is characterized by high proliferation and high turn-over metabolism. There are few results of metabolomics analysis correlated with outcomes of SCLC patients.

      Method

      This study included 27 patients after excluding eight who were treated with immunotherapy or placebo. Median age of the patients was 65 years (range, 43 to 82), and 23 male patients were included. Three (11%) never-smoker patients were included. Twelve patients were limited disease (LD) and 15 were extensive disease (ED) including three patients with brain metastases. All patients were treated with etoposide and platinum (EP, 20 cisplatin and 7 carboplatin), and nine among 12 patients with LD were treated concurrently with radiotherapy since 3rd cycle of EP chemotherapy. We planned to collect blood samples at diagnosis (T1), after two cycles of chemotherapy (T2) and at the first progression (T3). We analyzed 27 T1 samples, 14 T2 samples, and 18 T3 samples. Metabolomics analysis included 183 metabolites (21 amino acids, 19 biogenic amines, 40 acylcarnitines, 14 lysoglycerophosphocholines, 74 glycerophospholipids, and 14 sphingolipids, 1 hexose) using AbsoluteIDQ® p180 kit (Biocrates Life Sciences AG, Innsbruck, Austria).

      Result

      Baseline levels of five acylcarnitines and one glycerophospholipid were associated with progression free survival whereas, the amino acid proline and acylcarnitine (C10:1) were associated to overall survival. In comparison to LD, amino acid, asparagine was elevated while two acylcarnitines (C10:2 and C12:1) were lower in patients with ED. After two cycles of chemotherapy without progression, plasma concentration of 37 lipids (5 LysoPC, 24 glycerophospholipids, 3 sphingolipids and 5 acylcarnitines) were increased. Five mono and poly unsaturated glycerophosphospholipids (PC aa and PC ae) with C42 and C44 fatty acid chains were significantly (adj p < 0.05) lower. Of these, chemotherapy and induction of remission induced change in glycerophospholipid PC ae C42:5 was related to overall survival (p < 0.05). The 7 glycerophospholipids with C42 and C44 fatty acids were elevated when patients relapsed post chemotherapy. Five glycerophospholipid metabolites were significantly lower in response to chemotherapeutic exposure. The metabolites tended to increase towards baseline levels upon relapse.

      Conclusion

      With this preliminary exploratory study, we identified association of metabolites and initial status and outcomes of SCLC. We observed significant modulation in glycerophospholipids upon chemotherapy and relapse which may serve as indicators of therapeutic response and resistance. Further exploration and validation is being underway in larger SCLC population.

  • +

    P2.14 - Targeted Therapy (ID 183)

    • Event: WCLC 2019
    • Type: Poster Viewing in the Exhibit Hall
    • Track: Targeted Therapy
    • Presentations: 4
    • Moderators:
    • Coordinates: 9/09/2019, 10:15 - 18:15, Exhibit Hall
    • +

      P2.14-54 - High Incidence of CNS Metastases in Advanced or Recurrent Non-Small Cell Lung Cancer Patients with RET Fusion (ID 1758)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract

      Background

      Chromosomal rearrangements involving RET, with incidence of 1-2% in non-small-cell lung cancer, define a distinctive molecular subset. Here, we aim to determine the clinicopathological characteristics of patients with advanced NSCLC harboring the RET fusion gene.

      Method

      We identified 59 consecutive cases with RET rearrangements by using break-apart fluorescence in situ hybridization (n=14), next-generation sequencing (n=37), or both (n=8). Clinical data, including baseline characteristics, initial presentation, responses to chemotherapy and/or immunotherapy were retrospectively analyzed.

      Result

      The median age was 56 years, and 53% of patients were male. Approximately half of the patients (51%) were never-smokers. Adenocarcinoma was the predominant histologic subtype (90%), followed by pleomorphic (3%), neuroendocrine (3%), squamous cell (2%), and small cell carcinoma (2%). For the 19 patients with small primary lesions (<3cm), 32% (6/19) had N2 and 37% (7/19) had N3 disease. 17 patients (29%) had an intracranial lesion at the initial presentation or at the time of recurrence, and additional 11 patients (19%) developed brain metastasis during follow-up. Cerebrospinal fluid cytology exam confirmed leptomeningeal seeding in four patients (7%) with concomitant parenchymal metastasis. The median time to development of brain metastases was 19.0 months (range 3.8-51.8).

      Of 30 patients whose fusion partners were identified, kinesin family member 5B (KIF5B) was the most common, followed by coiled-coil domain containing 6 gene (CCDC6), nuclear receptor coactivator 4 (NCOA4), and myosin 5C (MYO5C) and lisH domain and HEAT repeat-containing protein KIAA1468 homolog (KIAA1468). Additionally, a novel fusion partner, phytanoyl-CoA 2-hydroxylase interacting protein like (PHYHIPL), was reported in one patient. Only two patients harbored concomitant EGFR mutation, and no ALK alterations were reported.

      The median overall survival was 35.3 months (95% confidence interval [CI]: 21.7-48.9). In 46 patients who were treated with pemetrexed-based chemotherapy, the overall response rate (ORR) and progression-free survival (PFS) time were 53.0% and 8.5 months (95% CI: 6.3-10.7), respectively. In 13 patients who were treated with immunotherapy, the ORR and PFS were 7.7% and 1.5 months (95% CI: 1.3-1.7), respectively, with no significant difference according to the level of PD-L1 expression.

      Conclusion

      Our study revealed the unique clinical characteristics and outcomes of advanced NSCLC patients harboring RET fusion gene. Considering the high incidence of CNS metastases, relatively poor response to immunotherapy, and a recent development of RET kinase inhibitors (such as cabozantinib and vandetanib), more efforts are warranted for identification of patients with RET fusion as a candidate for targeted therapies.

    • +

      P2.14-57 - Efficacy of 3rd Generation EGFR-TKIs After Failing First or Second Generation EGFR-TKIs in EGFR Mutation-Positive NSCLC (ROOT-EGFR) (ID 1474)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract

      Background

      Over the past decade, treatment of EGFR mutation –positive NSCLC has been revolutionized with the development of next generation EGFR TKIs. Four epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs), erlotinib, gefitinib, afatinib and osimertinib, are currently available in Korea. Recent trials have compared the available and emerging EGFR TKIs head to head. The highly anticipated findings of the phase III FLAURA trial showed a nearly doubling of PFS with frontline osimertinib, a third-generation TKI, versus erlotinib or gefitinib.The optimal sequence of EGFR TKIs is still controversial. And many countries 3rd generation EGFR TKI is not readily available. The purpose of this study is to analyze the efficacy in patients who received first-line gefitinib, erlotinib, or afatinib and followed by 3rd generation EGFR-TKIs.

      Method

      This non-interventional observational study through big data analysis will retrospectively collect de-identified patient data from clinical data warehouse (CDW) using a unique algorithm with Standard Query Language (SQL) called ROOT project.

      Result

      Over 10 years, 2,358 patients were diagnosed recurrent or metastatic non-small cell lung cancer and received 1st or 2nd generation EGFR TKIs at Samsung Medical Center. 72.9% (1720/2358) of EGFR mutation-positive NSCLC patients received 1st or 2nd generation EGFR-TKIs as first line palliative chemotherapy. In total population, 30.8% (727/2358) patients received 3rd generation EGFR TKIs. Among them, patients who received 3rd generation EGFR TKI after 1st or 2nd generation EGF TKIs showed better overall survival (45.0 months (95% CI, 41.8-48.2). Among 1720 patients who received 1st or 2nd generation EGFR-TKIs as first line palliative chemotherapy, 313 patients received 3rd generation EGFR-TKIs as 2nd line chemotherapy and 200 patients received as more than 3rd line chemotherapy. However, there was no difference of overall survival between them (44.0 months vs 47.0months, p-value=0.104).

      Conclusion

      This study was meaningful as a study of what can be the best sequence in EGFR-TKI treatment. Updated and detail clinical and exploratory biomarker outcome will be presented at the meeting.

    • +

      P2.14-59 - Osimertinib in EGFR T790M Advanced NSCLC: Analysis of Uncommon/Complex EGFR Mutations in a Real-World Study (ASTRIS) (ID 1263)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract
      • Slides

      Background

      The challenges of treating uncommon/complex EGFR mutations impact treatment decisions in clinical practice. Osimertinib is a third-generation, irreversible, oral EGFR-TKI that potently and selectively inhibits both EGFR-TKI sensitising (EGFRm) and EGFR T790M mutations and has demonstrated efficacy in NSCLC CNS metastases. ASTRIS is an ongoing, international, real-world study of osimertinib in EGFR T790M positive advanced NSCLC (NCT02474355). We report a subset-analysis of patients with uncommon/complex mutations.

      Method

      Patients with stage IIIB/IV T790M positive NSCLC previously treated with an EGFR-TKI were enrolled and received osimertinib 80 mg once-daily. Progression-free survival (PFS), clinical response and time to treatment discontinuation (TTD) were analysed in both full analysis set (FAS) and patients with uncommon/complex EGFR mutations. Uncommon mutation combinations included: T790M+G719X, T790M+S768I, T790M+ex20ins; complex mutations included T790M+two or more mutation(s).

      Result

      From 18 September 2015 to 15 October 2018 data cut-off, 3015 patients across 16 countries had received ≥1 dose of osimertinib (FAS), 53(2%) of these patients had uncommon/complex EGFR mutations. Baseline demographics between this patient subset and the FAS were similar (Asian: 55%/69%; female: 57%/64%; median age: 59 [30–80] years/62 [range, 27–92]; WHO performance status 2: 9%/11%, respectively). Baseline EGFR mutation status at enrolment of the FAS is shown in Table 1. Clinical outcomes appeared to be lower in the uncommon/complex mutation subset than in the FAS: response rate (measured in a FAS subset with ≥1 documented response assessment) was 50% [95% CI, 35.2, 64.8] in the uncommon/complex mutation group, and 57% [55.2, 58.9] in the FAS; median PFS was 8.1 [5.4, 10.1] and 11.1 [11.0, 12.0] months; median TTD was 9.0 [6.7, 11.5] and 13.5 [12.6, 13.9] months, respectively. Overall survival data are immature.

      astris table 1.png

      Conclusion

      Whilst clinical outcomes appeared to be lower in the uncommon/complex mutation subset than the FAS, they were favourable and support use of osimertinib 80mg in this heterogeneous population.

      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.

    • +

      P2.14-61 - Acquired Resistance to Entrectinib Associated with Activation of RAS Signaling Pathway in ROS1-Rearranged Non-Small Cell Lung Cancer (ID 1219)

      10:15 - 18:15  |  Author(s): Keunchil Park

      • Abstract

      Background

      ROS1 is a receptor tyrosine kinase (RTK) that is not usually expressed at high levels in normal lung tissue. The wild-type function of ROS1 is unknown, and a natural ligand has not been identified. ROS1 gene rearrangements occur in ~1-2% of patients with NSCLC, and have also been identified in colorectal, gastric and ovarian cancers, glioblastoma and cholangiocarcinoma. ROS1 rearrangements with oncogenic potential have been found to constitutively activate ROS1 signaling, and although it remains unclear exactly how the ROS1 fusion proteins are activated, the PI3K/AKT, MAPK/ERK and JAK/STAT3 pathways are known to be involved. Thus, ROS1-rearranged NSCLCs are ‘addicted’ to ROS1 for cell growth and survival. Knocking down or pharmacologically inhibiting ROS1 has been reported to inhibit growth or induce apoptosis in ROS1-rearranged cell lines. Entrectinib is a pan-tyrosine-kinase inhibitor that targets oncogenic rearrangements in NTRK, ROS1 and ALK. The combined results of two clinical trials demonstrated the efficacy of entrectinib in ROS1-rearranged NSCLC. Because the development of drug resistance is inevitable, it would be helpful to determine the mechanisms of entrectinib resistance in a ROS1-rearranged tumor model so that future therapeutic strategies can be developed.

      Method

      To investigate the effects of entrectinib on ROS1-rearranged NSCLC, we used HCC78 cells harboring an SLC34A2-ROS1 fusion. Entrectinib-resistant HCC78 (HCC78ER) cells were newly established in our laboratory through the exposure of HCC78 cells to gradually increasing concentrations of entrectinib (starting at 100 nM and ending with 5 μM) over 6 months.

      Result

      Here, we characterized the molecular basis of resistance in entrectinib-resistant ROS1-rearranged HCC78 cells (HCC78ER cells). These cells were analyzed by next-generation sequencing and genetic profiling, which revealed the acquisition of KRAS G12C and the amplification of KRAS and FGF3. However, there were no secondary mutations in the ROS1 kinase domain. We also found that sustained ERK activation was involved in entrectinib resistance, and that combined treatment with selumetinib resensitized HCC78ER cells to entrectinib in cell viability and colony formation assays.

      Conclusion

      Our data suggest that activation of the RAS signaling pathway can cause entrectinib resistance in ROS1-rearranged NSCLC, and is unlikely to be overcome by sequential single agent ROS1-targeting strategies against such tumors. Instead, co-targeting ROS1 and MEK may be an effective strategy for overcoming entrectinib resistance in ROS1-rearranged NSCLC.

  • +

    PL02 - Presidential Symposium including Top 7 Rated Abstracts (ID 89)

    • Event: WCLC 2019
    • Type: Plenary Session
    • Track:
    • Presentations: 1
    • Now Available
    • +

      PL02.08 - Registrational Results of LIBRETTO-001: A Phase 1/2 Trial of LOXO-292 in Patients with RET Fusion-Positive Lung Cancers (Now Available) (ID 964)

      08:00 - 10:15  |  Author(s): Keunchil Park

      • Abstract
      • Presentation
      • Slides

      Background

      No targeted therapy is currently approved for patients with RET fusion-positive non-small cell lung cancer (NSCLC). LOXO-292 is a highly selective RET inhibitor with activity against diverse RET fusions, activating RET mutations and brain metastases. Based on initial data from LIBRETTO-001, LOXO-292 received FDA Breakthrough Designation for the treatment of RET fusion-positive NSCLC in August 2018.

      Method

      This global phase 1/2 study (87 sites, 16 countries) enrolled patients with advanced RET-altered solid tumors including RET fusion-positive NSCLC (NCT03157128). LOXO-292 was dosed orally in 28-day cycles. The phase 1 portion established the MTD/RP2D (160 mg BID). The phase 2 portion enrolled patients to one of six cohorts based on tumor type, RET alteration, and prior therapies. The primary endpoint was ORR (RECIST 1.1). Secondary endpoints included DoR, CNS ORR, CNS DoR, PFS, OS, safety and PK.

      Result

      As of 17-June 2019, 253 RET fusion-positive NSCLC patients were treated. The primary analysis set (PAS) for LOXO-292 registration, as defined with the US FDA, consists of the first 105 consecutively enrolled RET fusion-positive NSCLC patients who received prior platinum-based chemotherapy; 58 patients (55%) also received prior anti PD-1/PD-L1 agents. The majority of PAS responders have been followed for ≥6 months from first response. Of the remaining 148 patients, 79 had previously been treated with platinum-based chemotherapy, 55 did not receive prior platinum-based chemotherapy and 14 did not have measurable disease at baseline.

      Among PAS patients, the investigator-assessed ORR was 68% (95% CI 58-76%, n=71/105, 2 PRs pending confirmation). Responses did not differ by fusion partner or the type or number of prior therapies, including chemotherapy, anti PD-1/PD-L1 agents and multikinase inhibitors with anti-RET activity. The median DoR was 20.3 months (95% CI 13.8-24.0) with a median follow-up of 8 months; as evidenced by the wide confidence interval, this DoR estimate is not statistically stable due to a low number of events (16 of 69 confirmed responders). The intracranial ORR was 91% (n=10/11: 2 confirmed CRs, 8 confirmed PRs) for patients with measurable brain metastases at baseline.

      The ORR in efficacy evaluable treatment naïve RET fusion-positive NSCLC patients was 85% (95% CI 69-95%, n=29/34, 7 PRs pending confirmation). In the safety data set of all 531 patients, 5 treatment-related AEs occurred in ≥15% of patients: dry mouth, diarrhea, hypertension, increased AST and increased ALT. Most AEs were grade 1-2. Only 9 of 531 (1.7%) patients discontinued LOXO-292 for treatment-related AEs.

      Conclusion

      LOXO-292 had marked antitumor activity in RET fusion-positive NSCLC patients and was well tolerated. These data will form the basis of an FDA NDA submission later this year.

      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.