Circulating tumor DNA analysis depicts subclonal architecture and genomic evolution of small cell lung cancer

Jingying Nong, Yuhua Gong, Yanfang Guan, Xin Yi, Yuting Yi, Lianpeng Chang, Ling Yang, Jialin Lv, Zhirong Guo, Hongyan Jia, Yuxing Chu, Tao Liu, Ming Chen, Lauren Byers, Emily Roarty, Vincent K Lam, Vassiliki A Papadimitrakopoulou, Ignacio Wistuba, John V Heymach, Bonnie Glisson, Zhongxing Liao, J Jack Lee, P Andrew Futreal, Shucai Zhang, Xuefeng Xia, Jianjun Zhang, Jinghui Wang, Jingying Nong, Yuhua Gong, Yanfang Guan, Xin Yi, Yuting Yi, Lianpeng Chang, Ling Yang, Jialin Lv, Zhirong Guo, Hongyan Jia, Yuxing Chu, Tao Liu, Ming Chen, Lauren Byers, Emily Roarty, Vincent K Lam, Vassiliki A Papadimitrakopoulou, Ignacio Wistuba, John V Heymach, Bonnie Glisson, Zhongxing Liao, J Jack Lee, P Andrew Futreal, Shucai Zhang, Xuefeng Xia, Jianjun Zhang, Jinghui Wang

Abstract

Subclonal architecture and genomic evolution of small-cell lung cancer (SCLC) under treatment has not been well studied primarily due to lack of tumor specimens, particularly longitudinal samples acquired during treatment. SCLC is characterized by early hematogenous spread, which makes circulating cell-free tumor DNA (ctDNA) sequencing a promising modality for genomic profiling. Here, we perform targeted deep sequencing of 430 cancer genes on pre-treatment tumor biopsies, as well as on plasma samples collected prior to and during treatment from 22 SCLC patients. Similar subclonal architecture is observed between pre-treatment ctDNA and paired tumor DNA. Mean variant allele frequency of clonal mutations from pre-treatment ctDNA is associated with progression-free survival and overall survival. Pre- and post-treatment ctDNA mutational analysis demonstrate that mutations of DNA repair and NOTCH signaling pathways are enriched in post-treatment samples. These data suggest that ctDNA sequencing is promising to delineate genomic landscape, subclonal architecture, and genomic evolution of SCLC.

Conflict of interest statement

The authors declare the following competing interests: Y.H.G., Y.F.G., X.Y., Y.T.Y., L.P.C., L.Y., Y.X.C., and T.L. are current employees of Geneplus-Beijing. X.Y. and L.Y. hold leadership positions and stocks of Geneplus-Beijing. J.V.H. is a consultant for AstraZeneca, Abbvie, Boehringer Ingelheim, Bristol-Myers Squibb, Medivation, ARIAD, Synta, Oncomed, Novartis, Genentech, and Calithera Biosciences, holds stock in Cardinal Spine LLC and Bio-Tree, and has received funding from AstraZeneca. J.Z. is a consultant for AstraZeneca and receives honoraria from Bristol-Myers Squibb. I.I.W. receives honoraria from Roche/Genentech, Ventana, GlaxoSmithKline, Celgene, Bristol-Myers Squibb, Synta Pharmaceuticals, Boehringer Ingelheim, Medscape, Clovis, AstraZeneca, and Pfizer, and research support from Roche/Genentech, Oncoplex, and HGT. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1
Somatic mutation profiles of 22 SCLC patients from pre-treatment ctDNA sequencing of 430 cancer genes. Twenty-two patients were arranged along the x-axis. Mutation per Mb region, clinical and pathological characters were shown in the upper panel. Genes with somatic mutations were shown in the middle panel. Mutation frequencies of each gene were shown on the left and mutation frequencies of these genes in previous report were shown on the right to each gene. The mutational spectrum was shown at the bottom
Fig. 2
Fig. 2
Somatic mutations detected in paired tumor DNA and ctDNA. Genes with somatic mutations were listed on the x-axis, and samples were shown on the y-axis. Mutations detected only in tumor DNA (tDNA), only in ctDNA or in both were shown in blue, red and orange, respectively
Fig. 3
Fig. 3
Comparison of genomic architecture derived from paired ctDNA versus tumor DNA. CCF of mutations were calculated in ctDNA and tumor DNA. Each dot represents one mutation and the color of each dot indicates the subclone that given mutation was clustered to. Correlation of CCF of all mutations in each pair of samples was shown in the table at lower-right corner. CCF_P: CCF of mutations in ctDNA; CCF_T: CCF of mutations in tumor DNA
Fig. 4
Fig. 4
The association of PFS and OS with ctDNA level measured by the average VAF of clonal mutations. Left, Patients were divided into two groups by ctDNA level. High ctDNA level group (higher than median of 0.18, dot line) was significantly associated with shorter PFS. Three patients were excluded from PFS analysis for three different reasons, including one patient who did not receive any treatment based on the patient's choice, one patient received surgery, and one could not offer out-patient otherapeutic records. Right, Patients were divided into two groups using the median ctDNA level of 0.18. Significantly shorter OS was observed in patients with higher ctDNA level (dot line). Small-cell lung cancer (SCLC) may evolve under treatment. But tumor tissues are often not available to study evolution of SCLC. Here, the authors utilize circulating tumor DNA to investigate the genomic evolution and subclonal architecture of SCLC during therapy

References

    1. Murray N, et al. Importance of timing for thoracic irradiation in the combined modality treatment of limited-stage small-cell lung cancer. The National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 1993;11:336–344. doi: 10.1200/JCO.1993.11.2.336.
    1. Johnson BE, et al. Ten-year survival of patients with small-cell lung cancer treated with combination chemotherapy with or without irradiation. J. Clin. Oncol. 1990;8:396–401. doi: 10.1200/JCO.1990.8.3.396.
    1. Fry WA, Menck HR, Winchester DP. The National Cancer Data Base report on lung cancer. Cancer. 1996;77:1947–1955. doi: 10.1002/(SICI)1097-0142(19960501)77:9<1947::AID-CNCR27>;2-Z.
    1. Lassen U, et al. Long-term survival in small-cell lung cancer: posttreatment characteristics in patients surviving 5 to 18+years–an analysis of 1,714 consecutive patients. J. Clin. Oncol. 1995;13:1215–1220. doi: 10.1200/JCO.1995.13.5.1215.
    1. van Meerbeeck JP, Fennell DA, De Ruysscher DK. Small-cell lung cancer. Lancet. 2011;378:1741–1755. doi: 10.1016/S0140-6736(11)60165-7.
    1. Cancer Genome Atlas Research N.. Comprehensive molecular profiling of lung adenocarcinoma. Nature. 2014;511:543–550. doi: 10.1038/nature13385.
    1. Cancer Genome Atlas N.. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi: 10.1038/nature11252.
    1. Cancer Genome Atlas Research N.. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455:1061–1068. doi: 10.1038/nature07385.
    1. Davies H, et al. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766.
    1. Peifer M, et al. Integrative genome analyses identify key somatic driver mutations of small-cell lung cancer. Nat. Genet. 2012;44:1104–1110. doi: 10.1038/ng.2396.
    1. Rudin CM, et al. Comprehensive genomic analysis identifies SOX2 as a frequently amplified gene in small-cell lung cancer. Nat. Genet. 2012;44:1111–1116. doi: 10.1038/ng.2405.
    1. George J, et al. Comprehensive genomic profiles of small cell lung cancer. Nature. 2015;524:47–53. doi: 10.1038/nature14664.
    1. Bettegowda C, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014;6:224ra224. doi: 10.1126/scitranslmed.3007094.
    1. Tie J, et al. Circulating tumor DNA as an early marker of therapeutic response in patients with metastatic colorectal cancer. Ann. Oncol. 2015;26:1715–1722. doi: 10.1093/annonc/mdv177.
    1. Dawson SJ, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013;368:1199–1209. doi: 10.1056/NEJMoa1213261.
    1. Garcia-Murillas I, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci. Transl. Med. 2015;7:302ra133. doi: 10.1126/scitranslmed.aab0021.
    1. Tie J, et al. Circulating tumor DNA analysis detects minimal residual disease and predicts recurrence in patients with stage II colon cancer. Sci. Transl. Med. 2016;8:346ra392. doi: 10.1126/scitranslmed.aaf6219.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Chae YK, et al. Concordance between genomic alterations assessed by next-generation sequencing in tumor tissue or circulating cell-free DNA. Oncotarget. 2016;7:65364–65373.
    1. Roth A, et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods. 2014;11:396–398. doi: 10.1038/nmeth.2883.
    1. Adzhubei IA, et al. A method and server for predicting damaging missense mutations. Nat. Methods. 2010;7:248–249. doi: 10.1038/nmeth0410-248.
    1. Phallen J, et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci Transl Med. 2017;9(403):pii: eaan2415.. doi: 10.1126/scitranslmed.aan2415.
    1. Razavi P, et al. Performance of a high-intensity 508-gene circulating-tumor DNA (ctDNA) assay in patients with metastatic breast, lung, and prostate cancer. J. Clin. Oncol. 2017;35:LBA11516. doi: 10.1200/JCO.2017.35.15_suppl.LBA11516.
    1. Abbosh C, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature. 2017;545:446–451. doi: 10.1038/nature22364.
    1. Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014;346:256–259. doi: 10.1126/science.1256930.
    1. de Bruin EC, et al. Spatial and temporal diversity in genomic instability processes defines lung cancer evolution. Science. 2014;346:251–256. doi: 10.1126/science.1253462.
    1. Jamal-Hanjani M, et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2109–2121. doi: 10.1056/NEJMoa1616288.
    1. Moraes RVd, Boneti BS, Silva MJDBE, Lima VCCD. Total tumor burden as predictive tool of response and survival of patients with metastatic melanoma treated with nivolumab. J. Clin. Oncol. 2017;35:e21022–e21022. doi: 10.1200/JCO.2017.35.15_suppl.e21022.
    1. Park JH, et al. Tumor burden is predictive of survival in patients with non-small-cell lung cancer and with activating epidermal growth factor receptor mutations who receive gefitinib. Clin. Lung. Cancer. 2013;14:383–389. doi: 10.1016/j.cllc.2012.10.007.
    1. Gerber DE, et al. Baseline tumour measurements predict survival in advanced non-small cell lung cancer. Br. J. Cancer. 2013;109:1476–1481. doi: 10.1038/bjc.2013.472.
    1. Taghipour M, Wray R, Sheikhbahaei S, Wright JL, Subramaniam RM. FDG avidity and tumor burden: survival outcomes for patients with recurrent breast cancer. AJR Am. J. Roentgenol. 2016;206:846–855. doi: 10.2214/AJR.15.15106.
    1. Yeh P, et al. Molecular disease monitoring using circulating tumor DNA in myelodysplastic syndromes. Blood. 2017;129:1685–1690. doi: 10.1182/blood-2016-09-740308.
    1. Olaussen KA, et al. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N. Engl. J. Med. 2006;355:983–991. doi: 10.1056/NEJMoa060570.
    1. Jiang Y, et al. Deep sequencing reveals clonal evolution patterns and mutation events associated with relapse in B-cell lymphomas. Genome Biol. 2014;15:432.
    1. Mar BG, et al. Mutations in epigenetic regulators including SETD2 are gained during relapse in paediatric acute lymphoblastic leukaemia. Nat. Commun. 2014;5:3469. doi: 10.1038/ncomms4469.
    1. Hassan WA, et al. Notch1 controls cell chemoresistance in small cell lung carcinoma cells. Thorac. Cancer. 2016;7:123–128. doi: 10.1111/1759-7714.12297.
    1. Wang VE, et al. Checkpoint inhibitor is active against large cell neuroendocrine carcinoma with high tumor mutation burden. J. Immunother. Cancer. 2017;5:75. doi: 10.1186/s40425-017-0281-y.
    1. Carbone DP, et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2415–2426. doi: 10.1056/NEJMoa1613493.
    1. Voong KR, Feliciano J, Becker D, Levy B. Beyond PD-L1 testing-emerging biomarkers for immunotherapy in non-small cell lung cancer. Ann. Transl. Med. 2017;5:376. doi: 10.21037/atm.2017.06.48.
    1. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348.
    1. Snyder A, et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014;371:2189–2199. doi: 10.1056/NEJMoa1406498.
    1. Reck M, et al. Ipilimumab in combination with paclitaxel and carboplatin as first-line therapy in extensive-disease-small-cell lung cancer: results from a randomized, double-blind, multicenter phase 2 trial. Ann. Oncol. 2013;24:75–83. doi: 10.1093/annonc/mds213.
    1. Reck M, et al. Phase III randomized trial of ipilimumab plus etoposide and platinum versus placebo plus etoposide and platinum in extensive-stage small-cell lung cancer. J Clin Oncol. 2016;34(31):3740–3748. doi: 10.1200/JCO.2016.67.6601.
    1. Antonia SJ, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:883–895. doi: 10.1016/S1470-2045(16)30098-5.
    1. Szustakowski, J. D. Impact of tumor mutation burden on the efficacy of nivolumab or nivolumab+ipilimumab in small cell lung cancer: an exploratory analysis of CheckMate 032. In Proc. World Conference on Lung Cancer 2017 (IASLC, Yokohama, Japan, 2017).
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324.
    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 J, et al. CONTRA: copy number analysis for targeted resequencing. Bioinformatics. 2012;28:1307–1313. doi: 10.1093/bioinformatics/bts146.
    1. Cowell JK, Lo KC. Application of oligonucleotides arrays for coincident comparative genomic hybridization, ploidy status and loss of heterozygosity studies in human cancers. Methods Mol. Biol. 2009;556:47–65. doi: 10.1007/978-1-60327-192-9_5.
    1. Murtaza M, et al. Multifocal clonal evolution characterized using circulating tumour DNA in a case of metastatic breast cancer. Nat. Commun. 2015;6:8760. doi: 10.1038/ncomms9760.

Source: PubMed

3
Prenumerera