Liquid biopsy uncovers distinct patterns of DNA methylation and copy number changes in NSCLC patients with different EGFR-TKI resistant mutations

Hoai-Nghia Nguyen, Ngoc-Phuong Thi Cao, Thien-Chi Van Nguyen, Khang Nguyen Duy Le, Dat Thanh Nguyen, Quynh-Tho Thi Nguyen, Thai-Hoa Thi Nguyen, Chu Van Nguyen, Ha Thu Le, Mai-Lan Thi Nguyen, Trieu Vu Nguyen, Vu Uyen Tran, Bac An Luong, Linh Gia Hoang Le, Quoc Chuong Ho, Hong-Anh Thi Pham, Binh Thanh Vo, Luan Thanh Nguyen, Anh-Thu Huynh Dang, Sinh Duy Nguyen, Duc Minh Do, Thanh-Thuy Thi Do, Anh Vu Hoang, Kiet Truong Dinh, Minh-Duy Phan, Hoa Giang, Le Son Tran, Hoai-Nghia Nguyen, Ngoc-Phuong Thi Cao, Thien-Chi Van Nguyen, Khang Nguyen Duy Le, Dat Thanh Nguyen, Quynh-Tho Thi Nguyen, Thai-Hoa Thi Nguyen, Chu Van Nguyen, Ha Thu Le, Mai-Lan Thi Nguyen, Trieu Vu Nguyen, Vu Uyen Tran, Bac An Luong, Linh Gia Hoang Le, Quoc Chuong Ho, Hong-Anh Thi Pham, Binh Thanh Vo, Luan Thanh Nguyen, Anh-Thu Huynh Dang, Sinh Duy Nguyen, Duc Minh Do, Thanh-Thuy Thi Do, Anh Vu Hoang, Kiet Truong Dinh, Minh-Duy Phan, Hoa Giang, Le Son Tran

Abstract

Targeted therapy with tyrosine kinase inhibitors (TKI) provides survival benefits to a majority of patients with non-small cell lung cancer (NSCLC). However, resistance to TKI almost always develops after treatment. Although genetic and epigenetic alterations have each been shown to drive resistance to TKI in cell line models, clinical evidence for their contribution in the acquisition of resistance remains limited. Here, we employed liquid biopsy for simultaneous analysis of genetic and epigenetic changes in 122 Vietnamese NSCLC patients undergoing TKI therapy and displaying acquired resistance. We detected multiple profiles of resistance mutations in 51 patients (41.8%). Of those, genetic alterations in EGFR, particularly EGFR amplification (n = 6), showed pronounced genome instability and genome-wide hypomethylation. Interestingly, the level of hypomethylation was associated with the duration of response to TKI treatment. We also detected hypermethylation in regulatory regions of Homeobox genes which are known to be involved in tumor differentiation. In contrast, such changes were not observed in cases with MET (n = 4) and HER2 (n = 4) amplification. Thus, our study showed that liquid biopsy could provide important insights into the heterogeneity of TKI resistance mechanisms in NSCLC patients, providing essential information for prediction of resistance and selection of subsequent treatment.

Conflict of interest statement

The authors declare no competing interests.

© 2021. The Author(s).

Figures

Figure 1
Figure 1
The heterogeneity of mutation profiles of 122 Vietnamese NSCLC patients with acquired resistance to first and second generation EGFR TKI. (A) Distribution of TKI resistance associated mutations of 122 Vietnamese NSCLC. (B) Frequencies of EGFR sensitizing mutations (del19, L858R, or rare EGFR mutations) in different resistance mutation profiles. (C) Variant allele frequency (VAF, %) of EGFR sensitizing mutations in different resistance mutation profiles. Data are presented as median VAF, each data point represents one patient. **p < 0.01; ***p < 0.001 (one-way ANOVA, Kruskal–Wallis test).
Figure 2
Figure 2
EGFR dependent (on-target) and independent (off-target) resistance mechanisms showed distinct landscapes of genome-wide methylation and copy number changes. (A, C) Methylation density (MD) (A) and DNA copy number (C) alterations for each 1 Mb bin along the chromosomes (chr1-22). Cases with T790M (n = 5), EGFR amplification (n = 3) and T790M and EGFR amplification co-occurrence (n = 3) were grouped into the on-target group while HER2 amplification (n = 4) and MET amplification (n = 4) cases were grouped into the off-target group. Healthy individuals (n = 20) were used as a reference group to determine the baseline levels of MD and CNA. (B, D) Percent of hypo-methylated bins (B) and CNA bins (D) in cases with different resistance mutation profiles grouped into the on-target group or the off-target group. The component bar graphs showed the comparison between the two groups. Data are shown as mean ± SEM. **p < 0.01, ****p < 0.0001 (Mann–Whitney test). (E) Linear regression and Pearson’s correlation between proportion of hypo-methylated bins and CNA bins in 19 patients from both on-target and off-target groups.
Figure 3
Figure 3
Accumulation of genetic and epigenetic changes in cases with EGFR amplification correlated with the duration of response to TKI treatment. (AC): Linear regression and Pearson’s correlation between proportion of CNA bins (A), hypo-methylated bins (B), EGFR amplification scores (C) and time to treatment resistance (TTTR) in 6 cases with EGFR amplification including 3 cases with co-occurring T790M.
Figure 4
Figure 4
EGFR dependent resistance mechanisms displayed markedly high levels of hyper-methylation in the transcriptional regions of genes involved in regulation of differentiation. (A) Heat-map showed differentially methylated regions (DMRs) among 450 target regions mapped to regulatory regions of cancer related genes across samples within the on-target group (T790M, T790M-EGFR amplification and EGFR amplification) and off-target group (HER2 and MET amplification). Healthy subjects were used as a reference group to determine the basal levels of methylation at target regions. Heatmap visualization was analyzed with ComplexHeatmap package version 2.8.0 (https://www.bioconductor.org/packages/release/bioc/html/ComplexHeatmap.html). Hierarchical clustering was done using Euclidean distance. Red in the scale bar represents a high level of methylation. (B) KEGG and Reactome pathway enrichment analysis using g: Profiler (https://biit.cs.ut.ee/gprofiler) for genes associated with 202 DMRs showing hyper-methylation in the on-target group compared to the off-target group.

References

    1. Bray F, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018;68:394–424. doi: 10.3322/caac.21492.
    1. Sher T, Dy GK, Adjei AA. Small cell lung cancer. Mayo Clin. Proc. 2008;83:355–367. doi: 10.4065/83.3.355.
    1. Mok TS, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 2009;361:947–957. doi: 10.1056/NEJMoa0810699.
    1. Shepherd FA, et al. Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 2005;353:123–132. doi: 10.1056/NEJMoa050753.
    1. Maemondo M, et al. First-line gefitinib in patients aged 75 or older with advanced non-small cell lung cancer harboring epidermal growth factor receptor mutations: NEJ 003 study. J. Thorac. Oncol. 2012;7:1417–1422. doi: 10.1097/JTO.0b013e318260de8b.
    1. Yang JC-H, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): Analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015;16:141–151. doi: 10.1016/S1470-2045(14)71173-8.
    1. Sequist LV, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J. Clin. Oncol. 2013;31:3327–3334. doi: 10.1200/JCO.2012.44.2806.
    1. Passaro A, Jänne PA, Mok T, Peters S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nature Cancer. 2021;2:377–391. doi: 10.1038/s43018-021-00195-8.
    1. Solassol I, Pinguet F, Quantin X. FDA- and EMA-approved tyrosine kinase inhibitors in advanced EGFR-mutated non-small cell lung cancer: safety, tolerability, plasma concentration monitoring, and management. Biomolecules. 2019;9:1–10. doi: 10.3390/biom9110668.
    1. Rosell R, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N. Engl. J. Med. 2009;361:958–967. doi: 10.1056/NEJMoa0904554.
    1. Stewart EL, Tan SZ, Liu G, Tsao MS. Known and putative mechanisms of resistance to EGFR targeted therapies in NSCLC patients with EGFR mutations: A review. Transl Lung Cancer Res. 2015;4:67–81.
    1. Wang J, Wang B, Chu H, Yao Y. Intrinsic resistance to EGFR tyrosine kinase inhibitors in advanced non-small-cell lung cancer with activating EGFR mutations. Onco. Targets. Ther. 2016;9:3711–3726. doi: 10.2147/OTT.S106399.
    1. Santoni-Rugiu E, et al. Intrinsic resistance to EGFR-tyrosine kinase inhibitors in EGFR-mutant non-small cell lung cancer: Differences and similarities with acquired resistance. Cancers. 2019;11:923. doi: 10.3390/cancers11070923.
    1. Kubo T, et al. MET gene amplification or EGFR mutation activate MET in lung cancers untreated with EGFR tyrosine kinase inhibitors. Int. J. Cancer. 2009;124:1778–1784. doi: 10.1002/ijc.24150.
    1. Camidge DR, Pao W, Sequist LV. Acquired resistance to TKIs in solid tumours: Learning from lung cancer. Nat. Rev. Clin. Oncol. 2014;11:473–481. doi: 10.1038/nrclinonc.2014.104.
    1. Jackman D, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J. Clin. Oncol. 2010;28:357–360. doi: 10.1200/JCO.2009.24.7049.
    1. Engelman JA, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039–1043. doi: 10.1126/science.1141478.
    1. Zhang Z, et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012;44:852–860. doi: 10.1038/ng.2330.
    1. Terai H, et al. Activation of the FGF2-FGFR1 autocrine pathway: A novel mechanism of acquired resistance to gefitinib in NSCLC. Mol. Cancer Res. 2013;11:759–767. doi: 10.1158/1541-7786.MCR-12-0652.
    1. Oser MG, Niederst MJ, Sequist LV, Engelman JA. Transformation from non-small-cell lung cancer to small-cell lung cancer: molecular drivers and cells of origin. Lancet Oncol. 2015;16:e165–e172. doi: 10.1016/S1470-2045(14)71180-5.
    1. Glasspool RM, Teodoridis JM, Brown R. Epigenetics as a mechanism driving polygenic clinical drug resistance. Br. J. Cancer. 2006;94:1087–1092. doi: 10.1038/sj.bjc.6603024.
    1. Hou T, et al. Decitabine reverses gefitinib resistance in PC9 lung adenocarcinoma cells by demethylation of RASSF1A and GADD45β promoter. Int. J. Clin. Exp. Pathol. 2019;12:4002–4010.
    1. Maeda M, et al. CpG hypermethylation contributes to decreased expression of PTEN during acquired resistance to gefitinib in human lung cancer cell lines. Lung Cancer. 2015;87:265–271. doi: 10.1016/j.lungcan.2015.01.009.
    1. Yang B, Yang Z-G, Gao B, Shao G-G, Li G-H. 5-Aza-CdR can reverse gefitinib resistance caused by DAPK gene promoter methylation in lung adenocarcinoma cells. Int. J. Clin. Exp. Pathol. 2015;8:12961–12966.
    1. Blair LP, Yan Q. Epigenetic mechanisms in commonly occurring cancers. DNA Cell Biol. 2012;31(Suppl 1):S49–61. doi: 10.1089/dna.2012.1654.
    1. Flavahan WA, Gaskell E, Bernstein BE. Epigenetic plasticity and the hallmarks of cancer. Science. 2017;357:6348. doi: 10.1126/science.aal2380.
    1. Darwiche N. Epigenetic mechanisms and the hallmarks of cancer: an intimate affair. Am. J. Cancer Res. 2020;10:1954–1978.
    1. Brennan K, Flanagan JM. Is there a link between genome-wide hypomethylation in blood and cancer risk? Cancer Prev. Res. 2012;5:1345–1357. doi: 10.1158/1940-6207.CAPR-12-0316.
    1. Zhang W, et al. Global DNA hypomethylation in epithelial ovarian cancer: Passive demethylation and association with genomic instability. Cancers. 2020;12:764. doi: 10.3390/cancers12030764.
    1. Niu X, et al. Genome-wide DNA methylation analysis reveals GABBR2 as a novel epigenetic target for EGFR 19 deletion lung adenocarcinoma with induction erlotinib treatment. Clin. Cancer Res. 2017;23:5003–5014. doi: 10.1158/1078-0432.CCR-16-2688.
    1. Jiang P, et al. Methy-Pipe: an integrated bioinformatics pipeline for whole genome bisulfite sequencing data analysis. PLoS ONE. 2014;9:e100360. doi: 10.1371/journal.pone.0100360.
    1. Chen X, et al. Non-invasive early detection of cancer four years before conventional diagnosis using a blood test. Nat. Commun. 2020;11:1–10.
    1. Zhang C, et al. DNA methylation analysis of the SHOX2 and RASSF1A panel in bronchoalveolar lavage fluid for lung cancer diagnosis. J. Cancer. 2017;8:3585–3591. doi: 10.7150/jca.21368.
    1. Tsou JA, et al. DNA methylation profile of 28 potential marker loci in malignant mesothelioma. Lung Cancer. 2007;58:220–230. doi: 10.1016/j.lungcan.2007.06.015.
    1. Hulbert A, et al. Early detection of lung cancer using DNA promoter hypermethylation in plasma and sputum. Clin. Cancer Res. 2017;23:1998–2005. doi: 10.1158/1078-0432.CCR-16-1371.
    1. Selamat SA, et al. Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res. 2012;22:1197–1211. doi: 10.1101/gr.132662.111.
    1. Nelson HH, et al. Key epigenetic changes associated with lung cancer development: results from dense methylation array profiling. Epigenetics. 2012;7:559–566. doi: 10.4161/epi.20219.
    1. Wen J, Fu J, Zhang W, Guo M. Genetic and epigenetic changes in lung carcinoma and their clinical implications. Mod. Pathol. 2011;24:932–943. doi: 10.1038/modpathol.2011.46.
    1. Jin Y, et al. Mutational profiling of non-small-cell lung cancer patients resistant to first-generation EGFR tyrosine kinase inhibitors using next generation sequencing. Oncotarget. 2016;7:61755–61763. doi: 10.18632/oncotarget.11237.
    1. Liam CK, Wahid MI, Rajadurai P, Cheah YK, Ng TS. Epidermal growth factor receptor mutations in lung adenocarcinoma in Malaysian patients. J. Thorac. Oncol. 2013;8:766–772. doi: 10.1097/JTO.0b013e31828b5228.
    1. Rotow J, Bivona TG. Understanding and targeting resistance mechanisms in NSCLC. Nat. Rev. Cancer. 2017;17:637–658. doi: 10.1038/nrc.2017.84.
    1. Sequist LV, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci. Transl. Med. 2011;3:7526. doi: 10.1126/scitranslmed.3002003.
    1. Kobayashi S, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2005;352:786–792. doi: 10.1056/NEJMoa044238.
    1. Harrison PT, Vyse S, Huang PH. Rare epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer. Semin. Cancer Biol. 2020;61:167–179. doi: 10.1016/j.semcancer.2019.09.015.
    1. You JS, Jones PA. Cancer genetics and epigenetics: Two sides of the same coin? Cancer Cell. 2012;22:9–20. doi: 10.1016/j.ccr.2012.06.008.
    1. Nukaga S, et al. Amplification of EGFR wild-type alleles in non-small cell lung cancer cells confers acquired resistance to mutation-selective EGFR tyrosine kinase inhibitors. Cancer Res. 2017;77:2078–2089. doi: 10.1158/0008-5472.CAN-16-2359.
    1. Buder A, et al. Somatic copy-number alterations in plasma circulating tumor DNA from advanced EGFR-mutated lung adenocarcinoma patients. Biomolecules. 2021;11:618. doi: 10.3390/biom11050618.
    1. Su S-F, et al. Genome-wide epigenetic landscape of lung adenocarcinoma links HOXB9 DNA methylation to intrinsic EGFR-TKI resistance and heterogeneous responses. JCO Precision Oncol. 2021;5:418–431. doi: 10.1200/PO.20.00151.
    1. Wu SG, Shih JY. Management of acquired resistance to EGFR TKI-targeted therapy in advanced non-small cell lung cancer. Mol. Cancer. 2018;17:38. doi: 10.1186/s12943-018-0777-1.
    1. Tran LS, et al. Ultra-deep massively parallel sequencing with unique molecular identifier tagging achieves comparable performance to droplet digital PCR for detection and quantification of circulating tumor DNA from lung cancer patients. PLoS ONE. 2019;14:e0226193. doi: 10.1371/journal.pone.0226193.
    1. Burriesci MS, Lehnert EM, Pringle JR. Fulcrum: Condensing redundant reads from high-throughput sequencing studies. Bioinformatics. 2012;28:1324–1327. doi: 10.1093/bioinformatics/bts123.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514.
    1. Li H, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Newman AM, et al. FACTERA: A practical method for the discovery of genomic rearrangements at breakpoint resolution. Bioinformatics. 2014;30:3390–3393. doi: 10.1093/bioinformatics/btu549.
    1. Shen R, Seshan VE. FACETS: Allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing. Nucleic Acids Res. 2016;44:e131. doi: 10.1093/nar/gkw520.
    1. Scheinin I, et al. DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly. Genome Res. 2014;24:2022–2032. doi: 10.1101/gr.175141.114.
    1. Krueger F, Andrews SR. Bismark: A flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics. 2011;27:1571–1572. doi: 10.1093/bioinformatics/btr167.
    1. Raudvere U, et al. g:Profiler: A web server for functional enrichment analysis and conversions of gene lists (2019 update) Nucleic Acids Res. 2019;47:W191–W198. doi: 10.1093/nar/gkz369.

Source: PubMed

3
Subscribe