11 research outputs found

    Molecular landscape of osimertinib resistance in patients and patient-derived preclinical models

    Get PDF
    Introduction: Osimertinib is a third-generation EGFR tyrosine kinase inhibitor (TKI) that is approved for the use of EGFR-mutant non-small cell lung cancer (NSCLC) patients. In this study, we investigated the acquired resistance mechanisms in NSCLC patients and patient-derived preclinical models. Methods: Formalin-fixed paraffin-embedded tumor samples and plasma samples from 55 NSCLC patients who were treated with osimertinib were collected at baseline and at progressive disease (PD). Next-generation sequencing was performed in tumor and plasma samples using a 600-gene hybrid capture panel designed by AstraZeneca. Osimertinib-resistant cell lines and patient-derived xenografts and cells were generated and whole exome sequencing and RNA sequencing were performed. In vitro experiments were performed to functionally study the acquired mutations identified. Results: A total of 55 patients and a total of 149 samples (57 tumor samples and 92 plasma samples) were analyzed, and among them 36 patients had matched pre- and post-treatment samples. EGFR C797S (14%) mutation was the most frequent EGFR-dependent mechanism identified in all available progression samples, followed by EGFR G824D (6%), V726M (3%), and V843I (3%). Matched pre- and post-treatment sample analysis revealed in-depth acquired mechanisms of resistance. EGFR C797S was still most frequent (11%) among EGFR-dependent mechanism, while among EGFR-independent mechanisms, PIK3CA, ALK, BRAF, EP300, KRAS, and RAF1 mutations were detected. Among Osimertinib-resistant cell lines and patient-derived models, we noted acquired mutations which were potentially targetable such as NRAS p.Q61K, in which resistance could be overcome with combination of osimertinib and trametinib. A patient-derived xenograft established from osimertinib-resistant patient revealed KRAS p.G12D mutation which could be overcome with combination of osimertinib, trametinib, and buparlisib. Conclusion: In this study, we explored the genetic profiles of osimertinib-resistant NSCLC patient samples using targeted deep sequencing. In vitro and in vivo models harboring osimertinib resistance revealed potential novel treatment strategies after osimertinib failure.ope

    Repotrectinib Exhibits Potent Antitumor Activity in Treatment-Naรฏve and Solvent-Front-Mutant ROS1-Rearranged Non-Small Cell Lung Cancer

    Get PDF
    Purpose: Although first-line crizotinib treatment leads to clinical benefit in ROS1+ lung cancer, high prevalence of crizotinib-resistant ROS1-G2032R (ROS1G2032R) mutation and progression in the central nervous system (CNS) represents a therapeutic challenge. Here, we investigated the antitumor activity of repotrectinib, a novel next-generation ROS1/TRK/ALK-tyrosine kinase inhibitor (TKI) in ROS1+ patient-derived preclinical models. Experimental design: Antitumor activity of repotrectinib was evaluated in ROS1+ patient-derived preclinical models including treatment-naรฏve and ROS1G2032R models and was further demonstrated in patients enrolled in an on-going phase I/II clinical trial (NCT03093116). Intracranial antitumor activity of repotrectinib was evaluated in a brain-metastasis mouse model. Results: Repotrectinib potently inhibited in vitro and in vivo tumor growth and ROS1 downstream signal in treatment-naรฏve YU1078 compared with clinically available crizotinib, ceritinib, and entrectinib. Despite comparable tumor regression between repotrectinib and lorlatinib in YU1078-derived xenograft model, repotrectinib markedly delayed the onset of tumor recurrence following drug withdrawal. Moreover, repotrectinib induced profound antitumor activity in the CNS with efficient blood-brain barrier penetrating properties. Notably, repotrectinib showed selective and potent in vitro and in vivo activity against ROS1G2032R. These findings were supported by systemic and intracranial activity of repotrectinib observed in patients enrolled in the on-going clinical trial. Conclusions: Repotrectinib is a novel next-generation ROS1-TKI with improved potency and selectivity against treatment-naรฏve and ROS1G2032R with efficient CNS penetration. Our findings suggest that repotrectinib can be effective both as first-line and after progression to prior ROS1-TKI.ope

    Modeling Clinical Responses to Targeted Therapies by Patient-Derived Organoids of Advanced Lung Adenocarcinoma

    Get PDF
    Purpose: Patient-derived organoids (PDO) of lung cancer has been recently introduced, reflecting the genomic landscape of lung cancer. However, clinical relevance of advanced lung adenocarcinoma organoids remains unknown. Here, we examined the ability of PDOs to predict clinical responses to targeted therapies in individual patients and to identify effective anticancer therapies for novel molecular targets. Experimental design: Eighty-four organoids were established from patients with advanced lung adenocarcinoma. Formalin-fixed, paraffin-embedded tumor specimens from corresponding patients were analyzed by whole-exome sequencing (n = 12). Organoids were analyzed by whole-exome sequencing (n = 61) and RNA sequencing (n = 55). Responses to mono or combination targeted therapies were examined in organoids and organoid-derived xenografts. Results: PDOs largely retained somatic alterations including driver mutations of matching patient tumors. PDOs were able to recapitulate progression-free survival and objective responses of patients with non-small cell lung cancer receiving clinically approved tyrosine kinase inhibitors. PDOs recapitulated activity of therapeutic strategies under clinical investigation. YUO-071 harboring an EGFR exon 19 deletion and a BRAF G464A mutation and the matching patient responded to dabrafenib/trametinib combination therapy. YUO-004 and YUO-050 harboring an EGFR L747P mutation was sensitive to afatinib, consistent with the response in the matching patient of YUO-050. Furthermore, we utilized organoids to identify effective therapies for novel molecular targets by demonstrating the efficacy of poziotinib against ERBB2 exon 20 insertions and pralsetinib against RET fusions. Conclusions: We demonstrated translational relevance of PDOs in advanced lung adenocarcinoma. PDOs are an important diagnostic tool, which can assist clinical decision making and accelerate development of therapeutic strategies.ope

    A phase II study of poziotinib in patients with recurrent and/or metastatic head and neck squamous cell carcinoma

    Get PDF
    Background: In phase I studies, poziotinib has shown meaningful efficacy against various types of cancers. This phase 2 study aimed to investigate the efficacy and safety of poziotinib in recurrent and/or metastatic head and neck squamous cell carcinoma (R/M-HNSCC). Methods: Overall, 49 patients were enrolled (median age, 62 years; age range, 21-78 years). Patients received a median of two prior treatments including chemotherapy and others and received 12 mg poziotinib orally once daily as part of a 28-day cycle. The primary endpoint was objective response rate (ORR), and the secondary endpoints were progression-free survival (PFS) and overall survival (OS). Targeted capture sequencing was performed using available tissues to identify translational biomarkers related to clinical response. Results: ORR was 22.4%, median PFS was 4.0 months (95% confidence interval [CI], 1.8-6.2 months), and median OS was 7.6 months (95% CI, 4.4-10.8 months). The most common treatment-related adverse events were acneiform rash (85%) and mucositis (77%). A grade 3 or higher adverse event was acneiform rash (3%). Targeted capture sequencing was performed in 30 tissue samples. TP53 and PIK3CA were the most frequently mutated genes (43%), followed by CCND1 (33%) and EGFR (30%). Mutations in ERBB2, ERBB3, and ERBB4, which are HER family genes, were observed in 17%, 13%, and 10% samples, respectively. There was no difference in the frequency of somatic mutations in the HER family genes between the clinically benefitted and non-benefitted groups. Conclusion: Compared to other pan-HER inhibitors, poziotinib showed clinically meaningful efficacy in heavily treated R/M-HNSCC. Clinical trial registration number: NCT02216916.ope

    The Liability Threshold Model for Predicting the Risk of Cardiovascular Disease in Patients with Type 2 Diabetes: A Multi-Cohort Study of Korean Adults

    No full text
    Personalized risk prediction for diabetic cardiovascular disease (DCVD) is at the core of precision medicine in type 2 diabetes (T2D). We first identified three marker sets consisting of 15, 47, and 231 tagging single nucleotide polymorphisms (tSNPs) associated with DCVD using a linear mixed model in 2378 T2D patients obtained from four population-based Korean cohorts. Using the genetic variants with even modest effects on phenotypic variance, we observed improved risk stratification accuracy beyond traditional risk factors (AUC, 0.63 to 0.97). With a cutoff point of 0.21, the discrete genetic liability threshold model consisting of 231 SNPs (GLT231) correctly classified 87.7% of 2378 T2D patients as high or low risk of DCVD. For the same set of SNP markers, the GLT and polygenic risk score (PRS) models showed similar predictive performance, and we observed consistency between the GLT and PRS models in that the model based on a larger number of SNP markers showed much-improved predictability. In silico gene expression analysis, additional information was provided on the functional role of the genes identified in this study. In particular, HDAC4, CDKN2B, CELSR2, and MRAS appear to be major hubs in the functional gene network for DCVD. The proposed risk prediction approach based on the liability threshold model may help identify T2D patients at high CVD risk in East Asian populations with further external validations.ope

    Design and Optimization of Pilot-Scale Bunsen Process in Sulfur-Iodine (SI) Cycle for Hydrogen Production

    No full text
    Sulfur-Iodine cycle (SI cycle)์€ ์š”์˜ค๋“œ์™€ ํ™ฉ์„ ์ฒจ๊ฐ€ํ•˜์—ฌ ์ตœ์ข…์ ์œผ๋กœ ๋ฌผ์„ ์—ดํ™”ํ•™์ ์œผ๋กœ ๋ถ„ํ•ดํ•˜์—ฌ ์‚ฐ์†Œ์™€ ์ˆ˜์†Œ๋ฅผ ์ƒ์‚ฐํ•˜๋Š” ๊ณต์ •์œผ๋กœ ํ™ฉ์‚ฐ๋ถ„ํ•ด, ์š”์˜ค๋“œํ™” ์ˆ˜์†Œ ๋ถ„ํ•ด, ๋ถ„์  ๋ฐ˜์‘ ๋“ฑ ์„ธ๊ฐ€์ง€ ๋ฐ˜์‘๋“ค๋กœ ์ด๋ฃจ์–ด์ ธ ์žˆ๋‹ค. ๋ถ„์   ๋ฐ˜์‘์€ ๋‘๊ฐ€์ง€ ๊ณต์ • ์ค‘๊ฐ„์— ์กด์žฌํ•˜๋ฏ€๋กœ ๋‘ ๋ฐ˜์‘์— ํ•„์š”ํ•œ ํ™”ํ•™๋ฌผ์„ ์กฐ๋‹ฌํ•˜๋Š” ์—ญํ• ๋กœ ์ด์— ๋Œ€ํ•œ ์ƒ๋ถ„๋ฆฌ ๋ฐ ๋ฐ˜์‘๊ธฐ์— ๋Œ€ํ•œ ๋ถ„์„์ด ์ค‘์š”ํ•˜๋‹ค. ๋ณธ ์—ฐ๊ตฌ์—์„œ๋Š” 50 L/hr ์ˆ˜์†Œ๋ฅผ ์ƒ์‚ฐํ•˜๋Š” pilot scale์˜ Sulfur-Iodine Cycle ์ค‘ ๋ถ„์   ๊ณต์ •์— ๋Œ€ํ•œ ๋ชจ์‚ฌ, ๋ฏผ๊ฐ๋„ ๋ถ„์„, ๋ฏผ๊ฐ๋„ ๋ถ„์„์„ ํ† ๋Œ€๋กœํ•œ ๊ฐ๊ฐ ์ƒ๋ถ„๋ฆฌ๊ธฐ์™€ ๋ถ„์   ๋ฐ˜์‘๊ธฐ์— ๋Œ€ํ•œ ์ตœ์  ์กฐ๊ฑด์„ ์ œ์‹œํ•˜์˜€๋‹ค. ์—ด์—ญํ•™ ๋ฌผ์„ฑ์น˜์˜ ๊ณ„์‚ฐ์„ ์œ„ํ•ด Electrolyte Non-Random Two Liquid (ELECNRTL) model ์‚ฌ์šฉํ•˜์˜€๋‹ค. ๋ชจ๋ธ์— ๋Œ€ํ•œ ์‹ ๋ขฐ๋„ ํ™•๋ณด๋ฅผ ์œ„ํ•ด์„œ ์‹ค์ œpilot scale์˜ ๊ณต์ • ๋ฐ์ดํ„ฐ์™€ ๊ฒ€์ฆ์„ ์ˆ˜ํ–‰ํ•˜์˜€๋‹ค. ๋ฐ˜์‘๊ธฐ์˜ ์ข…๋ฅ˜๋ฅผ ์„ ์ •ํ•˜๊ธฐ ์œ„ํ•ด Continuous Stirred Tank Reactor (CSTR)๊ณผ Plug Flow Reactor (PFR) ๋™์ผํ•œ ์˜จ๋„ ๋ฐ ๋ถ€ํ”ผ ๋ณ€ํ™”์—์„œ SO2 ์ „ํ™˜์œจ์„ ๋น„๊ตํ•˜์˜€๋‹ค. ์ƒ๋ถ„๋ฆฌ๊ธฐ ์„ ์ •์„ ์œ„ํ•ด 3 ์ƒ ๋ถ„๋ฆฌ ์‹œ์Šคํ…œ(๊ธฐ์ฒด-์•ก์ฒด-์•ก์ฒด)๊ณผ ์•ก์ฒด-๊ธฐ์ฒด ๋ถ„๋ฆฌ ํ›„ ์•ก์ฒด-์•ก์ฒด ๊ตฌ์กฐ์—์„œ H2SO4 ์ƒ๊ณผ HIX ์ƒ์—์„œ์˜ ๋ถˆ์ˆœ๋ฌผ๋“ค์„ ๋น„๊ตํ•˜์˜€๋‹ค. PFR์—์„œ ์˜จ๋„, ์ง€๋ฆ„, ๊ธธ์ด๋ฅผ ๊ฒฐ์ • ๋ณ€์ˆ˜๋กœ SO2 ์ „ํ™˜์œจ์„ ์ตœ๋Œ€ํ™” ํ•˜๊ธฐ ์œ„ํ•œ ์ตœ์ ํ™”๋ฅผ ์ˆ˜ํ–‰ํ•˜์˜€๋Š”๋ฐ, ์˜จ๋„ 121 oC์™€PFR์˜ ์ง€๋ฆ„์ด 0.20 m ๋ฐ ๊ธธ์ด 7.6 m ์ผ ๋•Œ SO2 ์ „ํ™˜์œจ์ด 98% ์ตœ์  ๊ฒฐ๊ณผ์ž„์„ ํ™•์ธํ•˜์˜€๋‹ค. ๊ธฐ์กด pilot scale๊ณผ ๋™์ผํ•œ์šด์ „ ์กฐ๊ฑด ํ•˜์— PFR์˜ ์ง€๋ฆ„ 3/8 inch, ๊ธธ์ด 3.0 m, 120 oC ์ผ ๋•Œ ์ธ์ž… ๋ชฐ๋Ÿ‰์ธ I2 ๋ฐ H2O๋ฅผ ๊ฒฐ์ • ๋ณ€์ˆ˜๋กœ SO2 ์ „ํ™˜์œจ์—๋Œ€ํ•œ ์ตœ์ ํ™”๋ฅผ ์ˆ˜ํ–‰ํ•˜์˜€์„ ๋•Œ, SO2 ์ „ํ™˜์œจ์ด 10% ์ผ๋•Œ H2O ๋ฐ I2 ์˜ ์ธ์ž… ๋ชฐ๋Ÿ‰์€ ๊ฐ๊ฐ 17%์™€ 22%๋กœ ๊ฐ์†Œํ•˜์˜€๋‹ค. ์•ž์„ ์กฐ์—… ์กฐ๊ฑด ์ตœ์ ํ™” ์กฐ๊ฑด (121 oC, ์ง€๋ฆ„ 0.20m, ๊ธธ์ด: 7.6m) ๊ฒฝ์šฐ์—๋Š” SO2 ์ „ํ™˜์œจ์ด 98% ์ผ ๋•Œ H2O๊ฐ€ 1% ๊ทธ๋ฆฌ๊ณ  I2๊ฐ€7% ๊ฐ์†Œํ•˜์˜€๋‹ค. ์ƒ๋ถ„๋ฆฌ๊ธฐ์—์„œ HIX ์ƒ๋‚ด H2SO4 ์ตœ์†Œํ™”ํ•˜๋Š” ๋ชฉ์ ํ•จ์ˆ˜์—์„œ ๊ทธ์— ์ƒ์‘ํ•˜๋Š” ์˜จ๋„, I2์™€ H2O๋ฅผ ๊ฒฐ์ • ๋ณ€์ˆ˜๋กœ์„ค์ •ํ•˜์˜€์„ ๋•Œ, H2O ๋ชฐ๋Ÿ‰์ด ๊ธฐ์กด๊ณต์ •๋ณด๋‹ค 17% ๊ฐ์†Œํ•˜๊ณ  I2 ๋ชฐ๋Ÿ‰์ด 24% ๊ฐ์†Œํ•˜์˜€์„ ๋•Œ ์ตœ์†Œ ๋ถˆ์ˆœ๋ฌผ์ด ์ƒ์„ฑํ•˜์˜€๋‹ค.11Nscopuskciothe

    Preclinical Study of a Biparatopic METxMET Antibody-Drug Conjugate, REGN5093-M114, Overcomes MET-driven Acquired Resistance to EGFR TKIs in EGFR-mutant NSCLC

    No full text
    Purpose: MET amplification is a frequent mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI) in patients with EGFR-mutated non-small cell lung cancer (NSCLC), and combined treatment with EGFR TKIs and MET TKIs has been explored as a strategy to overcome resistance. However, durable response is invariably limited by the emergence of acquired resistance. Here, we investigated the preclinical activity of REGN5093-M114, a novel antibody-drug conjugate targeting MET in MET-driven patient-derived models. Experimental Design: Patient-derived organoids, patient-derived cells, or ATCC cell lines were used to investigate the in vitro/in vivo activity of REGN5093-M114. Results: REGN5093-M114 exhibited significant antitumor efficacy compared with METTKI or unconjugated METxMET biparatopic antibody (REGN5093). Regardless of MET gene copy number, MET-overexpressed TKI-naive EGFR-mutant NSCLC cells responded to REGN5093-M114 treatment. Cell surface MET expression had the most predictive power in determining the efficacy of REGN5093-M114. REGN5093-M114 potently reduced tumor growth of EGFR-mutant NSCLC with PTEN loss or MET Y1230C mutation after progression on prior osimertinib and savolitinib treatment. Conclusions: Altogether, REGN5093-M114 is a promising candidate to overcome the challenges facing functional MET pathway blockade.,restrictio

    Clinical decision support algorithm based on machine learning to assess the clinical response to anti-programmed death-1 therapy in patients with non-small-cell lung cancer

    No full text
    Objective: Anti-programmed death (PD)-1 therapy confers sustainable clinical benefits for patients with non-small-cell lung cancer (NSCLC), but only some patients respond to the treatment. Various clinical characteristics, including the PD-ligand 1 (PD-L1) level, are related to the anti-PD-1 response; however, none of these can independently serve as predictive biomarkers. Herein, we established a machine learning (ML)-based clinical decision support algorithm to predict the anti-PD-1 response by comprehensively combining the clinical information. Materials and methods: We collected clinical data, including patient characteristics, mutations and laboratory findings, from the electronic medical records of 142 patients with NSCLC treated with anti-PD-1 therapy; these were analysed for the clinical outcome as the discovery set. Nineteen clinically meaningful features were used in supervised ML algorithms, including LightGBM, XGBoost, multilayer neural network, ridge regression and linear discriminant analysis, to predict anti-PD-1 responses. Based on each ML algorithm's prediction performance, the optimal ML was selected and validated in an independent validation set of PD-1 inhibitor-treated patients. Results: Several factors, including PD-L1 expression, tumour burden and neutrophil-to-lymphocyte ratio, could independently predict the anti-PD-1 response in the discovery set. ML platforms based on the LightGBM algorithm using 19 clinical features showed more significant prediction performance (area under the curve [AUC] 0.788) than on individual clinical features and traditional multivariate logistic regression (AUC 0.759). Conclusion: Collectively, our LightGBM algorithm offers a clinical decision support model to predict the anti-PD-1 response in patients with NSCLC.restrictio
    corecore