A pilot study of ultra-deep targeted sequencing of plasma DNA identifies driver mutations in hepatocellular carcinoma

Ismail Labgaa, Carlos Villacorta-Martin, Delia D'Avola, Amanda J Craig, Johann von Felden, Sebastiao N Martins-Filho, Daniela Sia, Ashley Stueck, Stephen C Ward, M Isabel Fiel, Milind Mahajan, Parissa Tabrizian, Swan N Thung, Celina Ang, Scott L Friedman, Josep M Llovet, Myron Schwartz, Augusto Villanueva, Ismail Labgaa, Carlos Villacorta-Martin, Delia D'Avola, Amanda J Craig, Johann von Felden, Sebastiao N Martins-Filho, Daniela Sia, Ashley Stueck, Stephen C Ward, M Isabel Fiel, Milind Mahajan, Parissa Tabrizian, Swan N Thung, Celina Ang, Scott L Friedman, Josep M Llovet, Myron Schwartz, Augusto Villanueva

Abstract

Cellular components of solid tumors including DNA are released into the bloodstream, but data on circulating-free DNA (cfDNA) in hepatocellular carcinoma (HCC) are still scarce. This study aimed at analyzing mutations in cfDNA and their correlation with tissue mutations in patients with HCC. We included 8 HCC patients treated with surgical resection for whom we collected paired tissue and plasma/serum samples. We analyzed 45 specimens, including multiregional tumor tissue sampling (n = 24), peripheral blood mononuclear cells (PMBC, n = 8), plasma (n = 8) and serum (n = 5). Ultra-deep sequencing (5500× coverage) of all exons was performed in a targeted panel of 58 genes, including frequent HCC driver genes and druggable mutations. Mutations detected in plasma included known HCC oncogenes and tumor suppressors (e.g., TERT promoter, TP53, and NTRK3) as well as a candidate druggable mutation (JAK1). This approach increased the detection rates previously reported for mutations in plasma of HCC patients. A thorough characterization of cis mutations found in plasma confirmed their tumoral origin, which provides definitive evidence of the release of HCC-derived DNA fragments into the bloodstream. This study demonstrates that ultra-deep sequencing of cfDNA is feasible and can confidently detect somatic mutations found in tissue; these data reinforce the role of plasma DNA as a promising minimally invasive tool to interrogate HCC genetics.

Conflict of interest statement

Conflicts of interests: The authors declare no potential conflicts of interest.

Figures

Figure 1
Figure 1
a) Distribution of DNA length fragments in plasma and serum samples. b) Differential coverage for next-generation sequencing in tissue and plasma. Sequencing depth was significant higher in plasma to identify low-frequency mutations.
Figure 2
Figure 2
a) Heat-map of detected mutations with their concordance in tissue and plasma. Each column represents one tumor region and each row is a mutated gene found in tissue. b) Performance of ultra-deep sequencing to detect plasma mutations, on a mutation-basis and on a patient-basis. c) Impact of tumor size, AFP levels and variant allele fraction of the mutation in tissue in detection rate of cfDNA.
Figure 3
Figure 3
TP53 mutations in patient #5: Left panels illustrate read alignments using Integrative Genomics Viewer. Right panels show the percentage of alternative reads found in each compartment [i.e., tissue (top), PBMC (middle), plasma (bottom)]. Two adjacent mutations of TP53, located 3 base pairs apart, with a variant allele fraction of 80% were detected in tissue. The corresponding alternative alleles of these 2 mutations were not detected in PBMC, thus excluding germline events. Ultra-deep sequencing of plasma DNA detected the same 2 mutations in approximately 4% of circulating DNA fragments. Mutations in plasma are found in the exact same reads, identifying their shared origin from their tissue counterparts.
Figure 4
Figure 4
Mutation assay with ddPCR. Displayed are 1D amplitudes for mutant probe (upper lane) and wild-type probe (lower lane) for each mutation assay. Panels from left to right: Plasma and corresponding HCC tissue of same patient, blank (negative control), wild-type DNA (negative control). Blue: single positive droplets for mutant, green: single positive droplets for wild-type, grey: double negative droplets. Detected positive droplets for mutant alleles in plasma are circled in red.

References

    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10:472–484.
    1. Dawson SJ, Tsui DW, Murtaza M, Biggs H, Rueda OM, Chin SF, et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199–1209.
    1. Murtaza M, Dawson SJ, Tsui DW, Gale D, Forshew T, Piskorz AM, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112.
    1. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci Transl Med. 2014;6:224ra224.
    1. Mandel P, Metais P. C R Seances Soc Biol Fil. 1948;142:241–243. [Not Available]
    1. Jiang P, Chan CW, Chan KC, Cheng SH, Wong J, Wong VW, et al. Lengthening and shortening of plasma DNA in hepatocellular carcinoma patients. Proc Natl Acad Sci U S A. 2015
    1. Lin XJ, Chong Y, Guo ZW, Xie C, Yang XJ, Zhang Q, et al. A serum microRNA classifier for early detection of hepatocellular carcinoma: a multicentre, retrospective, longitudinal biomarker identification study with a nested case-control study. Lancet Oncol. 2015;16:804–815.
    1. Russo M, Siravegna G, Blaszkowsky LS, Corti G, Crisafulli G, Ahronian LG, et al. Tumor Heterogeneity and Lesion-Specific Response to Targeted Therapy in Colorectal Cancer. Cancer Discov. 2016;6:147–153.
    1. Tie J, Wang Y, Tomasetti C, Li L, Springer S, Kinde I, 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.
    1. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380–2388.
    1. FDA approves first blood test to detect gene mutation associated with non-small cell lung cancer [news release] Silver Spring MD: U.S. Food and Drug Administration; Jun 1, 2016. Accessed June 1, 2016.
    1. Liao W, Yang H, Xu H, Wang Y, Ge P, Ren J, et al. Noninvasive detection of tumor-associated mutations from circulating cell-free DNA in hepatocellular carcinoma patients by targeted deep sequencing. Oncotarget. 2016;7:40481–40490.
    1. Huang A, Zhao X, Yang XR, Li FQ, Zhou XL, Wu K, et al. Circumventing intratumoral heterogeneity to identify potential therapeutic targets in hepatocellular carcinoma. J Hepatol. 2017
    1. Cai ZX, Chen G, Zeng YY, Dong XQ, Lin MJ, Huang XH, et al. Circulating tumor DNA profiling reveals clonal evolution and real-time disease progression in advanced hepatocellular carcinoma. Int J Cancer. 2017
    1. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. Hepatocellular carcinoma. Nat Rev Dis Primers. 2016;2:16018.
    1. European Association For The Study Of The L, European Organisation For R, Treatment Of C. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2012;56:908–943.
    1. Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, et al. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83:8604–8610.
    1. Kan Z, Zheng H, Liu X, Li S, Barber TD, Gong Z, et al. Whole-genome sequencing identifies recurrent mutations in hepatocellular carcinoma. Genome Res. 2013;23:1422–1433.
    1. De Mattos-Arruda L, Weigelt B, Cortes J, Won HH, Ng CK, Nuciforo P, et al. Capturing intra-tumor genetic heterogeneity by de novo mutation profiling of circulating cell-free tumor DNA: a proof-of-principle. Ann Oncol. 2014;25:1729–1735.
    1. Zhai W, Lim TK, Zhang T, Phang ST, Tiang Z, Guan P, et al. The spatial organization of intra-tumour heterogeneity and evolutionary trajectories of metastases in hepatocellular carcinoma. Nat Commun. 2017;8:4565.
    1. Norton ME, Jacobsson B, Swamy GK, Laurent LC, Ranzini AC, Brar H, et al. Cell-free DNA analysis for noninvasive examination of trisomy. N Engl J Med. 2015;372:1589–1597.
    1. Malapelle U, Mayo de-Las-Casas C, Rocco D, Garzon M, Pisapia P, Jordana-Ariza N, et al. Development of a gene panel for next-generation sequencing of clinically relevant mutations in cell-free DNA from cancer patients. Br J Cancer. 2017;116:802–810.
    1. Zill OA, Greene C, Sebisanovic D, Siew LM, Leng J, Vu M, et al. Cell-Free DNA Next-Generation Sequencing in Pancreatobiliary Carcinomas. Cancer Discov. 2015;5:1040–1048.
    1. Huang A, Zhang X, Zhou SL, Cao Y, Huang XW, Fan J, et al. Detecting Circulating Tumor DNA in Hepatocellular Carcinoma Patients Using Droplet Digital PCR Is Feasible and Reflects Intratumoral Heterogeneity. J Cancer. 2016;7:1907–1914.
    1. Ono A. Circulating Tumor DNA Analysis for Liver Cancers and Its Usefulness as a Liquid Biopsy. Cellular and Molecular Gastroenterology and Hepatology. 2015
    1. Labgaa I, Villanueva A. Liquid biopsy in liver cancer. Discov Med. 2015;19:263–273.
    1. Newman AM, Bratman SV, To J, Wynne JF, Eclov NC, Modlin LA, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat Med. 2014;20:548–554.
    1. Schulze K, Imbeaud S, Letouze E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet. 2015;47:505–511.
    1. Izumchenko E, Chang X, Brait M, Fertig E, Kagohara LT, Bedi A, et al. Targeted sequencing reveals clonal genetic changes in the progression of early lung neoplasms and paired circulating DNA. Nat Commun. 2015;6:8258.
    1. Newman AM, Lovejoy AF, Klass DM, Kurtz DM, Chabon JJ, Scherer F, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat Biotechnol. 2016;34:547–555.
    1. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature. 2017;545:446–451.
    1. Siravegna G, Mussolin B, Buscarino M, Corti G, Cassingena A, Crisafulli G, et al. Clonal evolution and resistance to EGFR blockade in the blood of colorectal cancer patients. Nat Med. 2015;21:795–801.
    1. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364:2507–2516.
    1. Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med. 2017;23:703–713.
    1. Totoki Y, Tatsuno K, Covington KR, Ueda H, Creighton CJ, Kato M, et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat Genet. 2014;46:1267–1273.
    1. The Drug Gene Interaction Database

    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303.
    1. Van der Auwera GA, Carneiro MO, Hartl C, Poplin R, Del Angel G, Levy-Moonshine A, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr Protoc Bioinformatics. 2013;43:11 10 11–33.
    1. Koboldt DC, Zhang Q, Larson DE, Shen D, McLellan MD, Lin L, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576.
    1. Chang MT, Asthana S, Gao SP, Lee BH, Chapman JS, Kandoth C, et al. Identifying recurrent mutations in cancer reveals widespread lineage diversity and mutational specificity. Nat Biotechnol. 2016;34:155–163.

Source: PubMed

3
Abonnere