Virtual Library

Start Your Search

Myung-Ju Ahn



Author of

  • +

    FP14 - Targeted Therapy - Clinically Focused (ID 252)

    • Event: WCLC 2020
    • Type: Posters (Featured)
    • Track: Targeted Therapy - Clinically Focused
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      FP14.03 - Osimertinib + Savolitinib in pts with EGFRm MET-Amplified/Overexpressed NSCLC: Phase Ib TATTON Parts B and D Final Analysis (ID 3312)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Presentation
      • Slides

      Introduction

      Preliminary data from TATTON (NCT02143466, a multi-arm, multi-drug combination study) suggested that savolitinib, a potent and highly selective MET-tyrosine kinase inhibitor (TKI), plus osimertinib, a third-generation, irreversible, oral epidermal growth factor receptor (EGFR)-TKI, that potently and selectively inhibits T790M and EGFR mutations (EGFRm), may overcome MET-based resistance to EGFR-TKIs in NSCLC. We present the final data from two TATTON expansion cohorts (TATTON Parts B and D); data cutoff 4 March 2020.

      Methods

      Adult patients with locally advanced/metastatic, MET-amplified/overexpressed, EGFRm NSCLC, and disease progression on a prior EGFR-TKI. Most patients had retrospective, central confirmation of MET status by local fluorescent in-situ hybridisation (MET gene copy ≥5 or MET/CEP7 ratio ≥2), next-generation sequencing or immunohistochemistry (+3 in ≥50% of tumour cells). In Part B, patients received osimertinib 80 mg plus savolitinib 600 mg orally once daily; after a protocol amendment, patients ≤55 kg received savolitinib 300 mg. In Part D, patients who had received no prior third-generation EGFR-TKI and were T790M-negative, received osimertinib plus savolitinib 300 mg. The primary endpoint was safety/tolerability; secondary endpoints included objective response rate (ORR), progression-free survival (PFS) and pharmacokinetics.

      Results

      In Parts B and D, 138 and 42 patients respectively, received treatment. Grade ≥3 adverse events (AEs) were reported in 62% and 50% of patients, in Parts B and D respectively; serious AEs were reported in 49% and 38% of patients, respectively. AEs led to discontinuation of savolitinib in 49 (36%) and 15 (36%) patients, and osimertinib in 24 (17%) and 8 (19%) patients, for Parts B and D, respectively. In Part B, seven patients died due to AEs; two cases were possibly treatment-related. In Part D, two patients died due to AEs; neither was considered treatment-related. PFS and ORR results are included in the Table. Pharmacokinetics of savolitinib and osimertinib were consistent with other patient populations in TATTON and previous studies.

      Conclusion

      Osimertinib plus savolitinib was generally well tolerated and the safety profile was in-line with that previously reported. Our results support that osimertinib plus savolitinib may overcome MET-based resistance in patients with NSCLC whose disease has progressed on prior EGFR-TKI. Further exploration of the osimertinib plus savolitinib combination is underway in the SAVANNAH (NCT03778229) and ORCHARD (NCT03944772) studies.

      Table

      Part B: osimertinib 80 mg + savolitinib 600/300* mg

      Part D: osimertinib 80 mg + savolitinib 300 mg

      Endpoint

      Previously treated with a 3G EGFR-TKI

      No prior 3G EGFR-TKI, T790M-negative

      No prior 3G EGFR-TKI, T790M-positive

      No prior 3G EGFR-TKI, T790M-negative

      n=69

      n=51

      n=18

      n=42

      ORR, n (%)

      [95% CI]

      23 (33)

      [22.4, 45.7]

      33 (65)

      [50.1, 77.6]

      12 (67)

      [41.0, 86.7]

      26 (62)

      [45.6, 76.4]

      Median PFS, months
      [95% CI]

      5.5
      [4.1, 7.7]

      9.1
      [5.5,12.8]

      11.1
      [4.1,22.1]

      9.0
      [5.6, 12.7]

      Total PFS events, n (%)

      51 (74)

      36 (71)

      12 (67)

      29 (69)

      * Most patients were enrolled to 600 mg savolitinib, prior to weight-based dosing implementation, but following a protocol amendment, the final 21 patients enrolled in Part B were dosed with savolitinib by body weight as follows: patients who weighed ≤55 kg (n=7) received 300 mg daily and those weighing >55 kg (n=14) received 600 mg daily

      All confirmed responses were partial response.

      3G, third generation; CI, confidence interval; ORR, objective response rate; PFS, progression-free survival

      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.

  • +

    IS02 - Industry Symposium Sponsored by AstraZeneca: Evolving the Role of Immunotherapy in Lung Cancer: ES-SCLC and Unresectable Stage III NSCLC (ID 278)

    • Event: WCLC 2020
    • Type: Industry Symposium
    • Track:
    • Presentations: 1
    • +

      IS02.04 - Defining the Role of Immunotherapy in the Treatment of ES-SCLC (ID 4312)

      10:30 - 11:30  |  Presenting Author(s): Myung-Ju Ahn

      • Abstract

      Abstract not provided

  • +

    MA04 - Health Policy and the Real World (ID 217)

    • Event: WCLC 2020
    • Type: Mini Oral
    • Track: Health Services Research/Health Economics
    • Presentations: 1
    • +

      MA04.06 - Clinical Characteristics and Outcomes in Advanced KRAS Mutant NSCLC – A Multi-Centre Collaboration in Asia (ATORG-005) (ID 3475)

      16:45 - 17:45  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Presentation
      • Slides

      Introduction

      KRAS driver mutations in advanced NSCLC have long been considered to be undruggable. However, promising efficacy data from early phase trials of novel therapies targeting KRAS have renewed focus on KRAS as an oncogenic driver. There is limited data on the prognostic and predictive significance of KRAS mutation subtypes. We present an interim analysis of a real world observational multi-centre study of advanced KRAS mutant NSCLC patients from five countries in Asia, conducted by the Asian Thoracic Oncology Research Group (ATORG).

      Methods

      Patients with advanced KRAS mutant NSCLC treated with at least one line of systemic therapy at tertiary centres in five Asian countries (China, India, Japan, Singapore, South Korea) between Jan 2014 and Dec 2018 were included. Baseline clinical characteristics, molecular profile and treatment outcomes were collected (median follow-up 35.5 months, 95%CI 28.7-50.3).

      Results

      A total of 155 patients were included in this interim analysis, with median age at advanced stage diagnosis 63 years (interquartile range [IQR] 56-70), 93% were ECOG 0-1, 70% were male and 64% were current or ex-smokers. In terms of ethnicity, 39% were Korean, 36% were Chinese, 15% were Japanese, 8% were Indian and 2% were Malay. Baseline histology was adenocarcinoma in 90%, squamous cell carcinoma in 4% and other histologies in 6%. KRAS mutation was detected by NGS in 141 (91%) patients, Sanger sequencing in 12 (8%) patients and RT-PCR in 2 (1%) patients. KRAS G12C (26%) was most common, followed by G12D (23%) and G12V (21%). The incidence of KRAS G12C mutation in patients with a smoking history was 35/99 (35%) compared with 6/56 (11%) in patients without any smoking history. Co-alterations were found with EGFR mutations (14%), ALK fusions (1%), ROS1 fusions (1%) and BRAF mutations (3%). PD-L1 TPS was 0% in 22%, 1-49% in 19%, ≥50% in 14% and unknown/not tested in 45%. Brain metastases were present at advanced stage diagnosis in 25% and lifetime prevalence was 35%. Patients received a median 2 lines of therapy. First-line systemic therapy consisted of chemotherapy alone (66%), targeted therapy (15%) or other therapies (19%). Median time to next treatment (TTNT) on first-line chemotherapy alone was 7.3 months (95%CI 5.0-9.5). Overall, the median TTNT for first-line and second-line therapy was 7.7 (95%CI 6.5–10.0) and 7.0 (95%CI 5.3–10.9) months, respectively. 63% of patients had died, and 37% of patients were still alive or lost to follow-up at the time of data cut-off. Median OS for the overall cohort was 21.6 months (95%CI 15.9-27.6). Median OS was greater in immunotherapy treated (alone or in combination at any line; 45%) versus non-immunotherapy treated (55%) patients (27.6 [95%CI 19.1-37.9] months versus 15.4 [95%CI 10.3-23.7] months, HR 1.8, 95%CI 1.2-2.7, logrank p=0.005).

      Conclusion

      In Asian KRAS mutant NSCLC, duration of first-line therapy and survival outcomes remain poor – emphasising the need for greater therapeutic options for patients with a KRAS driver mutation. Additional sites/countries are planned and recruitment to this study is ongoing.

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

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

  • +

    OA07 - Immuno-biology and Novel Immunotherapeutics from Bench to Bed (ID 228)

    • Event: WCLC 2020
    • Type: Oral
    • Track: Immuno-biology and Novel Immunotherapeutics (Phase I and Translational)
    • Presentations: 1
    • +

      OA07.05 - Discussant (ID 4149)

      10:30 - 11:30  |  Presenting Author(s): Myung-Ju Ahn

      • Abstract
      • Presentation

      Abstract not provided

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

  • +

    P03 - Early Stage/Localized Disease - Clinical Trials in Progress (ID 112)

    • Event: WCLC 2020
    • Type: Posters
    • Track: Early Stage/Localized Disease
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      P03.03 - MERMAID-1: A Phase III Study of Adjuvant Durvalumab plus Chemotherapy in Resected NSCLC Patients with MRD+ Post-Surgery (ID 3322)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Slides

      Introduction

      30% of non-small-cell lung cancer (NSCLC) patients present with surgically resectable disease at diagnosis. To reduce disease recurrence, adjuvant chemotherapy is standard of care (SoC) for patients with completely (R0) resected stage II/III NSCLC. However, 5-year disease-free survival (DFS) rates remain low (~40%), with a 5-year absolute benefit of only 5.4% derived from chemotherapy (Pignon et al. JCO 2008;26:3552-9). Determining who will benefit from adjuvant chemotherapy remains challenging. Identifying minimal residual disease (MRD) through detection of circulating tumour (ct) DNA post-surgery could predict early disease recurrence. Durvalumab is a selective, high‑affinity, human IgG1 monoclonal antibody that blocks PD-L1 binding to PD-1 and CD80. In the phase III PACIFIC trial of patients with unresectable, stage III NSCLC without disease progression following radical platinum-based concurrent chemoradiotherapy, durvalumab improved progression-free survival (PFS) and overall survival (OS), demonstrating its efficacy in situations of residual disease. Further, initial data from POSEIDON showed durvalumab plus chemotherapy prolongs PFS versus chemotherapy in first line treatment of metastatic NSCLC. In the adjuvant setting (i.e. following complete tumour resection), this regimen could provide additional DFS benefit versus chemotherapy alone. MRD assessment could facilitate earlier, more selective adjuvant therapy for MRD+ patients, allowing for treatment intensification for this potentially biologically distinct disease, while minimising overtreatment of MRD– patients. MERMAID‑1 will investigate the efficacy and safety of adjuvant durvalumab + SoC chemotherapy versus placebo + SoC chemotherapy in patients with completely resected stage II/III NSCLC, to assess the benefits of adjuvant therapy in patients with MRD+ status.

      Methods

      MERMAID-1 (NCT04385368) is a phase III, parallel-arm, placebo-controlled, double‑blind, multicentre study, conducted across 16 countries. Patients aged ≥18 years with histologically confirmed EGFR-/ALK- Wild-type, stage II/III NSCLC will enter the first screening period; a personalised MRD panel will be created. Before randomisation, and following assessment of MRD status post-surgery, patients will be further screened for full eligibility (no evidence of disease recurrence, WHO/ECOG performance status 0/1). Patients who have received prior adjuvant therapy or durvalumab, and those with mixed small cell and NSCLC histology or evidence of post-operative disease recurrence are ineligible. MRD status will be determined via ctDNA analysis of plasma samples, collected 3–4 weeks post-surgery, and based on personalised panels (comprised of ≤50 tumour-specific DNA variants) created by whole exome sequencing analysis of the patient’s resected tumour tissue. Approximately 332 patients will be randomised 1:1 (stratified by disease stage, MRD status, and PD-L1 expression) to durvalumab (1500 mg, intravenously) or placebo, plus concurrent SoC chemotherapy, once every three weeks (Q3W) for 12 weeks. Patients will continue with durvalumab monotherapy/placebo Q4W thereafter, until Week 48 or disease recurrence, whichever occurs first. Following a baseline radiological scan prior to randomisation, patients will be assessed Q12W until disease recurrence (per RECIST v1.1). The primary endpoint is DFS in the MRD+ analysis set (investigator-assessed). Secondary endpoints include DFS in the full analysis set (FAS; investigator-assessed); DFS in the MRD+ analysis set and FAS (blinded independent central review); OS in the MRD+ analysis set and FAS; and safety and tolerability, and patient-reported outcomes.

      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.

  • +

    P14 - Immuno-biology and Novel Immunotherapeutics (Phase I and Translational) - Immuno-Biology (ID 153)

    • Event: WCLC 2020
    • Type: Posters
    • Track: Immuno-biology and Novel Immunotherapeutics (Phase I and Translational)
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      P14.25 - Immune Cell Profiling of Hyperprogressive Disease in Patients with Non-Small Cell Lung Cancer Treated with Anti-PD-1/PD-L1 Antibodies (ID 3272)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Slides

      Introduction

      Hyperprogressive disease (HPD) is a distinct pattern of progression characterized by acceleration of tumor growth after treatment with anti-PD-1/PD-L1 antibodies. However, the immunological characteristics have not been fully elucidated in patients with HPD. In the present study, we aimed to uncover the immunological characteristics specific to HPD using peripheral blood obtained before and early after anti-PD-1/PD-L1 treatment in patients with non-small cell lung cancer (NSCLC).

      Methods

      We prospectively recruited patients with metastatic non-small cell lung cancer treated with anti-PD-1/PD-L1 antibodies between April 2015 and April 2018, and collected peripheral blood before treatment and 7-days post-treatment. HPD was defined as ≥ 2-fold increase in both tumor growth kinetics and tumor growth rate between pre-treatment and post-treatment. From peripheral blood mononuclear cells, CD8+ and CD4+ T cells, regulatory T cells, and myeloid derived suppressor cells were analyzed by multi-color flow cytometry.

      Results

      Of 115 patients, 19 (16.5%) developed HPD, 52 experienced durable clinical benefit (DCB; partial response or stable disease ≥6 months), and 44 experienced non-hyperprogressive progression (NHPD). Patients with HPD had significantly lower progression-free survival (P < 0.001) and overall survival (P < 0.001). A total of 72 immune cell parameters were analyzed and the pre-treatment frequency of CD39+ cells among CD8+ T cells was significantly higher in patients with HPD compared to those with NHPD. Other parameters regarding the proliferative response or frequency of total regulatory T cells, effector regulatory T cells (FoxP3hiCD45RA-) or myeloid derived suppressor cells did not significantly differ among DCB, NHPD, and HPD groups.

      Conclusion

      Our findings suggest high pre-treatment frequency of CD39+CD8+ T cells might be a characteristic of HPD. Further investigations in a larger cohort are needed to confirm our results and better delineate the immune landscape of HPD.

      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.

  • +

    P60 - Tumor Biology and Systems Biology - Basic and Translational Science - Immune Bio (ID 198)

    • Event: WCLC 2020
    • Type: Posters
    • Track: Tumor Biology and Systems Biology - Basic and Translational Science
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      P60.09 - High Circulating Regulatory (FoxP3+) T Cells and TGF-β Predict the Response to Anti-PD-1 Immunotherapy in NSCLC Patients (ID 3270)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Slides

      Introduction

      Antitumor immune response induced by immune checkpoint inhibitors including anti-PD-1 or anti-PD-L1 in advanced non-small cell lung cancer (NSCLC) patients has been shown as promising new therapeutic strategies for the last decade. It has been reported that favorable antitumor activities to immune checkpoint inhibitor are strongly correlated with high PD-L1 expression, increased tumor infiltrating lymphocytes, and decreased suppressive immune cells including Treg cells, myeloid-derived suppressor cells (MDSCs), or tumor associated macrophages (TAMs) in various cancer types. The accumulation of Treg cells in tumor bed among various cancers is known to be associated with poor prognosis, as expected from their function inhibiting antitumor immunity and maintaining immune homeostasis. However, Treg cells frequencies in several cancer types showed the opposite results; a high Treg cell infiltration is associated with a favorable prognosis in patients bearing colorectal cancer, or ER- breast cancer.

      Methods

      The patients with NSCLC (stage IIIB to IV) undergoing anti-PD-1 immunotherapy with either pembrolizumab (200 mg every 3 weeks) or nivolumab (2 mg/kg every 2 weeks) were enrolled in a part of a phase II clinical trial (NCT02607631) at Samsung Medical Center (Korea). Baseline and one week after anti-PD-1 therapy peripheral blood samples were collected from March 2017 to February 2018 for discovery cohort (n = 83), and March 2018 to March 2019 for validation cohort (n = 49). Treg cells, PMN-MDSCs, M-MDSCs were analyzed their correlation with clinical outcomes including PFS (progression free survival) and OS (overall survival). mRNA and plasma level of TGF-β were also measured and analyzed the correlation with Treg cells and clinical outcomes.

      Results

      Treg cell frequencies high group showed longer PFS and OS and especially, high Treg cell frequency one week after anti-PD-1 therapy showed more distinct differences compared with Treg cells low group of the patients. Treg cells high frequency group was associated with relatively low PMN-MDSCs frequencies, and combined analysis of Treg cells high and PMN-MDSCs or M-MDSC low group showed better OS compared with Treg cells low and PMN-MDSC or M-MDSC high group. TGF-β mRNA expression correlated with Treg cells and clinical outcomes. Various cytokines in plasma correlated with Treg cells and clinical outcomes.

      Conclusion

      Our results suggest the favorable prognostic value of high circulating CD4+CD25+CD45RA-FoxP3+ T cells (effector Treg cells) frequencies one week after the immunotherapy in NSCLC patients who were treated with anti-PD-1 immunotherapy, either pembrolizumab or nivolumab. Together with Treg cells, the detection of a high TGF-β also identifies a more favorable outcome which may specifically benefit from anti-PD-1 immunotherapy. The understanding of the clinical relevance of the tumor microenvironmental immunologic milieu might provide an important clue when designing novel strategies in cancer immunotherapy, however, blood based Treg cells might also provide alternative sources when tumor biopsy is not available. Therefore, Treg cells frequencies and TGF-β expression level might be used as blood-based biomarkers to predict the response of anti-PD-1 immunotherapy in advanced NSCLC patients.

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

  • +

    P76 - Targeted Therapy - Clinically Focused - EGFR (ID 253)

    • Event: WCLC 2020
    • Type: Posters
    • Track: Targeted Therapy - Clinically Focused
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      P76.15 - Osimertinib Improved Overall Survival in mEGFR NSCLC Patients With Leptomeningeal Metastases Regardless of T790M Mutational Status (ID 1161)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Slides

      Introduction

      Osimertinib, a third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), demonstrated superior penetration of the blood-brain barrier, compared with first- or second-generation EGFR TKIs. However, previous studies have not evaluated whether treatment with osimertinib leads to improved overall survival (OS) for EGFR-mutated NSCLC patients with leptomeningeal metastases (LM) compared with those not treated with osimertinib.

      Methods

      From October 2008 to October 2019, patients with EGFR-mutated NSCLC and cytologically confirmed LM were analyzed for OS according to osimertinib treatment and T790M mutational status. OS was defined as the time from diagnosis of LM to death due to any cause.

      Results

      For the 351 LM patients included in analysis, the median OS (mOS) was 8.1 months (95% confidence interval [CI] 7.2–9.0). T790M mutation was detected in 88 of 197 patients tested, and a total of 110 patients were treated with osimertinib after LM. No significant difference in mOS was demonstrated according to T790M mutational status (10.1 months [95% CI 4.31–15.82] vs. 9.0 [95% CI 6.81–11.21], P = 0.936). However, patients treated with osimertinib showed a superior OS of 17.0 months (95% CI 15.13–18.94) compared with those not treated with osimertinib who showed a mOS 5.5 months (95% CI 4.34–6.63) regardless of T790M mutational status (Hazard ratio 0.36; 95% CI 0.28–0.47, P <0.001). This was a significant survival benefit even compared with those who were never treated with osimertinib but with first-/second-generation EGFR TKIs showing a mOS of 8.7 months (95% CI 7.01–10.39).

      Conclusion

      Osimertinib is a promising treatment option for EGFR-mutated NSCLC with LM regardless of T790M mutational status.

      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.

  • +

    P86 - Targeted Therapy - Clinically Focused - New Target (ID 263)

    • Event: WCLC 2020
    • Type: Posters
    • Track: Targeted Therapy - Clinically Focused
    • Presentations: 1
    • Moderators:
    • Coordinates: 1/28/2021, 00:00 - 00:00, ePoster Hall
    • +

      P86.12 - Cardiac Safety Assessment of Lazertinib in Patients with EGFR Mutation-Positive Advanced Non-Small Cell Lung Cancer (ID 1247)

      00:00 - 00:00  |  Author(s): Myung-Ju Ahn

      • Abstract
      • Slides

      Introduction

      Lazertinib (YH25448, JNJ-73841937) is a highly mutant-selective, irreversible 3rd-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor that targets the activating EGFR mutations (Del19 and L858R), as well as the T790M mutation, while sparing wild type. The recommended phase 2 dose was determined to be 240 mg once daily (QD) in Korean patients based on the results of a first-in-human study in patients with advanced, EGFR-mutated non-small cell lung cancer at doses of 20-320 mg QD. The objective of this evaluation was to assess lazertinib cardiac safety in these patients.

      Methods

      The electrocardiogram (ECG) assessments (absolute and change from baseline QTcF) were performed in an ongoing phase 1/2 lazertinib pharmacokinetics, safety and efficacy study. A total of 224 patients [1st (n=43) and 2nd (n=181) line therapy, 20-320 mg QD dose] with baseline and postdose ECG assessments (in triplicates) along with time-matched plasma concentration data were included in exposure-QTcF analysis using linear regression. The left ventricular ejection fraction (LVEF) was assessed using an echocardiogram or multiple gated acquisition (MUGA) scan at baseline and every 12 weeks.

      Results

      Of 224 evaluable patients, no post-treatment QTcF values >500 ms were reported during the study; 26 (11.6%) patients had a post-baseline QTcF >450 ms including 3 (1.3%) patients with QTcF >480 ms. Of the 221 patients with baseline QTcF confirmed by central assessment, 22 (10.0%) patients had >30 to 60 ms increase and 1 (0.5%) had >60 ms increase in QTcF from baseline. Of the 121 patients at 240 mg, 10 (8.3%) patients had a post-baseline QTcF >450 ms including 1 (0.8%) patient with QTcF >480 ms. There were no clinical symptoms of QTc prolongation observed in any of the patients. At clinically relevant (240 mg QD dosing) plasma steady state Cmax (maximum plasma concentration) of 517.15 (43% coefficient of variation) ng/mL, the upper bound of the two-sided 90% confidence interval for change from baseline QTcF was estimated to 3.9 ms (low concern category). The exposure-QTcF assessment-based prediction suggests that a 2.5-fold and 4.8-fold higher than 517.15 ng/ml plasma concentration would be required to cause the upper bound of two-sided 90% confidence interval for the change from baseline in QTcF of ~10 ms (increasing concern category) and QTcF of ~20 ms (definite concern category), respectively. These results are in line with in vitro (hERG assay), ex-vivo (isolated perfused rabbit heart) and in vivo (instrumented male beagle dogs) preclinical findings. Of 224 patients, no treatment emergent adverse event related with heart failure or clinically meaningful decrease of LVEF was reported.

      Conclusion

      Taken together, preclinical and clinical cardiac safety assessment findings suggest that lazertinib has no clinically relevant effect on QT interval and LVEF. Time-matched plasma concentration and QTcF read-outs as well as LVEF assessments will continue to be collected in all ongoing as well as future lazertinib clinical studies to further confirm that lazertinib has no/minimal cardiac safety risk.

      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.

  • +

    PL04 - A Vision for Clinical Trials in 2020 and Beyond (Japanese, Mandarin, Spanish Translation Available) (ID 145)

    • Event: WCLC 2020
    • Type: Plenary
    • Track: N.A.
    • Presentations: 1
    • +

      PL04.02 - Chair (ID 3918)

      07:00 - 09:00  |  Presenting Author(s): Myung-Ju Ahn

      • Abstract

      Abstract not provided