Abstract
Introduction
Methods
Results
Conclusions
Keywords
Introduction
Material and Methods
Targeted NGS Analysis
Statistical Analysis
In Vitro ALK Inhibitor Sensitivity
Results
Patient Characteristics
Characteristics | Patients (%) |
---|---|
N = 76 | |
Sex | |
Male | 36 (47) |
Female | 40 (53) |
Age, years | |
Median | 56 |
Range | 22-82 |
Race | |
White | 58 (76) |
Black/African American | 2 (2) |
Asian | 12 (16) |
Other/unknown | 4 (5) |
Smoking history | |
Current | 3 (4) |
Former | 26 (34) |
Never | 47 (62) |
No. of prior treatments | |
0 | 14 (18) |
1 | 17 (22) |
2 | 18 (24) |
3 | 10 (13) |
≥ 4 | 17 (22) |
Prior ALK TKI treatment | |
ALK TKI naive | 17 (22) |
Prior crizotinib only | 37 (49) |
Prior crizotinib and ceritinib | 9 (12) |
Prior crizotinib and alectinib | 6 (8) |
Prior crizotinib, ceritinib, and alectinib | 6 (8) |
Prior crizotinib, ceritinib, and brigatinib | 1 (1) |
Detection of Disease-Associated Variants in Baseline Plasma Samples

Concordance Between Tissue and Plasma Genotypes
Correlation Between Baseline ctDNA Findings and Response to Ensartinib
Evolution of Response and Resistance During Molecular Surveillance


Comparison of Patient ctDNA With In Vitro ALK Inhibitor Sensitivity Analysis
Discussion
Acknowledgments
Supplementary Data
- Supplementary Tables 1-6 and Supplementary Figures 1-13
- Supplementary Methods
References
- EML4-ALK fusion lung cancer: a rare acquired event.Neoplasia. 2008; 10: 298-302
- Anaplastic lymphoma kinase immunoreactivity correlates with ALK gene rearrangement and transcriptional up-regulation in non–small cell lung carcinomas.Hum Pathol. 2009; 40: 1152-1158
- Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs.JAMA. 2014; 311: 1998-2006
- Crizotinib versus chemotherapy in advanced ALK-positive lung cancer.N Engl J Med. 2013; 368: 2385-2394
- First-line ceritinib versus platinum-based chemotherapy in advanced ALK-rearranged non–small-cell lung cancer (ASCEND-4): a randomised, open-label, phase 3 study.Lancet. 2017; 389: 917-929
- Alectinib versus crizotinib in untreated ALK-positive non–small-cell lung cancer.N Engl J Med. 2017; 377: 829-838
- Lorlatinib in non–small-cell lung cancer with ALK or ROS1 rearrangement: an international, multicentre, open-label, single-arm first-in-man phase 1 trial.Lancet Oncol. 2017; 18: 1590-1599
- Brigatinib in patients with crizotinib-refractory anaplastic lymphoma kinase-positive non-small-cell lung cancer: a randomized, multicenter phase II trial.J Clin Oncol. 2017; 35: 2490-2498
- Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer.Cancer Discov. 2016; 6: 1118-1133
- Cracking the code of resistance across multiple lines of ALK inhibitor therapy in lung cancer.Cancer Discov. 2016; 6: 1084-1086
- Resistance mechanisms to targeted therapies in ROS1(+) and ALK(+) non–small cell lung cancer.Clin Cancer Res. 2018; 24: 3334-3347
- Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: the Lung Cancer Mutation Consortium Experience.J Thorac Oncol. 2015; 10: 768-777
- Analysis of intratumor heterogeneity unravels lung cancer evolution.Mol Cell Oncol. 2015; 2e985549
- Detection of therapeutically targetable driver and resistance mutations in lung cancer patients by next-generation sequencing of cell-free circulating tumor DNA.Clin Cancer Res. 2016; 22: 5772-5782
- Utility of genomic assessment of blood-derived circulating tumor DNA (ctDNA) in patients with advanced lung adenocarcinoma.Clin Cancer Res. 2017; 23: 5101-5111
- A prospective evaluation of circulating tumor cells and cell-free DNA in EGFR-mutant non–small cell lung cancer patients treated with erlotinib on a phase II trial.Clin Cancer Res. 2016; 22: 6010-6020
- Noninvasive genotyping and monitoring of anaplastic lymphoma kinase (ALK) rearranged non–small cell lung cancer by capture-based next-generation sequencing.Oncotarget. 2016; 7: 65208-65217
- Insights into ALK-driven cancers revealed through development of novel ALK tyrosine kinase inhibitors.Cancer Res. 2011; 71: 4920-4931
- Ensartinib (X-396) in ALK-positive non-small cell lung cancer: results from a first-in-human phase I/II, multicenter study.Clin Cancer Res. 2018; 24: 2771-2779
- Bias-corrected targeted next-generation sequencing for rapid, multiplexed detection of actionable alterations in cell-free DNA from advanced lung cancer patients.Clin Cancer Res. 2016; 22: 915-922
- Collection of cell-free DNA for genomic analysis of solid tumors in a clinical laboratory setting.PLoS One. 2017; 12e0176241
- Differential crizotinib response duration among ALK fusion variants in ALK-positive non–small-cell lung cancer.J Clin Oncol. 2016; 34: 3383-3389
- Prevalence and clinicopathological characteristics of ALK fusion subtypes in lung adenocarcinomas from Chinese populations.J Cancer Res Clin Oncol. 2016; 142: 833-843
- Impact of EML4-ALK variant on resistance mechanisms and clinical outcomes in ALK-positive lung cancer.J Clin Oncol. 2018; 36: 1199-1206
- Crizotinib can overcome acquired resistance to CH5424802: is amplification of the MET gene a key factor?.J Thorac Oncol. 2014; 9: e27-e28
- Crizotinib-resistant mutants of EML4-ALK identified through an accelerated mutagenesis screen.Chem Biol Drug Des. 2011; 78: 999-1005
- Identification of a novel ALK G1123S mutation in a patient with ALK-rearranged non–small-cell lung cancer exhibiting resistance to ceritinib.J Thorac Oncol. 2015; 10: e55-e57
- Phase 3 study of ceritinib vs chemotherapy in ALK-rearranged NSCLC patients previously treated with chemotherapy and crizotinib (ASCEND-5): Japanese subset.Jpn J Clin Oncol. 2018; 48: 367-375
- Longitudinal cell-free DNA analysis in patients with small cell lung cancer reveals dynamic insights into treatment efficacy and disease relapse.J Thorac Oncol. 2018; 13: 112-123
- Tracking the evolution of resistance to ALK tyrosine kinase inhibitors through longitudinal analysis of circulating tumor DNA.JCO Precis Oncol. 2018; 2018
- Clinical utility of cell-free DNA for the detection of ALK fusions and genomic mechanisms of ALK inhibitor resistance in non-small cell lung cancer.Clin Cancer Res. 2018; 24: 2758-2770
Article info
Publication history
Footnotes
Disclosure: Dr. Horn has received personal fees from AbbVie, AstraZeneca, Bristol-Myers Squibb, Lilly, Incyte, EMD Serono, Merck, Roche-Genentech, Tessaro, and Xcovery; and reports receiving commercial research support from Boehringer Ingelheim. Dr. Whisenant has received personal fees from Anasys Instruments. Dr. Wakelee is a consultant/advisory board member for AstraZeneca, Genentech/Roche (uncompensated), Merck (uncompensated), Novartis (uncompensated), and Ariad (uncompensated); and has received grants to her institution for conduct of clinical trial work from Genentech/Roche, Novartis, Pfizer, Lilly, Celgene, AstraZeneca/Medimmune, Exelixis, Clovis Oncology, Bristol-Myers Squibb, Gilead, Pharmacyclics, ACEA Biosciences, Merck, and Xcovery. Dr. Reckamp is a consultant/advisory board member for ARIAD Takeda, Exelixis, Guardant, Loxo, and Genentech; and has received grants from Xcovery, AbbVie, Acea, Adaptimmune, Boehringer Ingelheim, Bristol-Myers Squibb, Genentech, Guardant, Janssen, Loxo Oncology, Seattle Genetics, Takeda, and Zeno. Dr. Leal is a consultant/advisory board member for Roche-Genentech, Ariad, Takeda, AstraZeneca, Novartis, AbbVie, and Bristol-Myers Squibb. Dr. Blumenschein has served as a consultant for Abbvie, Adicet, Amgen, ARIAD, Bayer, Bristol-Myers Squibb, Celgene, Clovis, Merck, Novartis, and Xcovery; and has received research funding from Adaptimmune, Bayer, Bristol-Myers Squibb, Celgene, Exelixis, Genetech, GlaxoSmithKline, Hoffman-La Roche, Immatics, Incyte, Macrogenetics, MedImmune, Merck, Novartis, Torque, and Xcovery. Dr. Waqar has received the 1 UM1 CA186704-01 grant; and as institutional PI has received support from F. Hoffmann-La Roche, Ltd., Ariad, Pfizer Pharmaceuticals, Inc., Hengrui Therapeutics, Xcovery, EMD Serono Research and Development Institute, Inc., Checkpoint Therapeutics, Inc., Genentech, Inc., Lilly, Stemcentrx, Inc., Ignyta, Inc., Bristol-Myers Squibb Pharmaceutical, Synemore Biologics Co., Ltd., Novartis Pharmaceuticals Corporation, Merck & Company, Inc., NewLink Genetics Corporation, and Celgene. Dr. Patel is a scientific advisory board member for AstraZeneca, Bristol-Myers Squibb, Illumina, Tempus, and Novartis; and has received research funding from Bristol-Myers Squibb, Eli Lilly, Fate, Incyte, AstraZeneca/MedImmune, Merck, Pfizer, Roche/Genentech, Xcovery, Fate Therapeutics, Genocea, and Iovance. Research funds were distributed through University of California San Diego. Dr. Nieva has received personal fees from Roche and Takeda. Dr. Oxnard is a consultant for Takeda, AstraZeneca, Loxo, Inivata, and GRAIL. Dr. Sanborn is a consultant/advisory board member for ARIAD and Takeda. Drs. Hernandez, Shaffer, Garg, Lim, and Li are employees and shareholders of Resolution Biosciences. Dr. Holzhausen is an employee of Xcovery Holdings, Inc. Drs. Harrow and Liang are employees of and have ownership interests (including patents) in Xcovery Holdings, Inc. Dr. Lovly has served as a consultant for Pfizer, Novartis, Astra Zeneca, Genoptix, Sequenom, Ariad, Takeda, Foundation Medicine, Blueprints Medicine, and Cepheid; and has received research funding from Novartis, AstraZeneca, and Xcovery (the funds were distributed through Vanderbilt University, not given to Dr. Lovly directly). The remaining authors declare no conflict of interest.
Identification
Copyright
User license
Elsevier user license |
Permitted
For non-commercial purposes:
- Read, print & download
- Text & data mine
- Translate the article
Not Permitted
- Reuse portions or extracts from the article in other works
- Redistribute or republish the final article
- Sell or re-use for commercial purposes
Elsevier's open access license policy