Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors

Viktor A Adalsteinsson, Gavin Ha, Samuel S Freeman, Atish D Choudhury, Daniel G Stover, Heather A Parsons, Gregory Gydush, Sarah C Reed, Denisse Rotem, Justin Rhoades, Denis Loginov, Dimitri Livitz, Daniel Rosebrock, Ignaty Leshchiner, Jaegil Kim, Chip Stewart, Mara Rosenberg, Joshua M Francis, Cheng-Zhong Zhang, Ofir Cohen, Coyin Oh, Huiming Ding, Paz Polak, Max Lloyd, Sairah Mahmud, Karla Helvie, Margaret S Merrill, Rebecca A Santiago, Edward P O'Connor, Seong H Jeong, Rachel Leeson, Rachel M Barry, Joseph F Kramkowski, Zhenwei Zhang, Laura Polacek, Jens G Lohr, Molly Schleicher, Emily Lipscomb, Andrea Saltzman, Nelly M Oliver, Lori Marini, Adrienne G Waks, Lauren C Harshman, Sara M Tolaney, Eliezer M Van Allen, Eric P Winer, Nancy U Lin, Mari Nakabayashi, Mary-Ellen Taplin, Cory M Johannessen, Levi A Garraway, Todd R Golub, Jesse S Boehm, Nikhil Wagle, Gad Getz, J Christopher Love, Matthew Meyerson, Viktor A Adalsteinsson, Gavin Ha, Samuel S Freeman, Atish D Choudhury, Daniel G Stover, Heather A Parsons, Gregory Gydush, Sarah C Reed, Denisse Rotem, Justin Rhoades, Denis Loginov, Dimitri Livitz, Daniel Rosebrock, Ignaty Leshchiner, Jaegil Kim, Chip Stewart, Mara Rosenberg, Joshua M Francis, Cheng-Zhong Zhang, Ofir Cohen, Coyin Oh, Huiming Ding, Paz Polak, Max Lloyd, Sairah Mahmud, Karla Helvie, Margaret S Merrill, Rebecca A Santiago, Edward P O'Connor, Seong H Jeong, Rachel Leeson, Rachel M Barry, Joseph F Kramkowski, Zhenwei Zhang, Laura Polacek, Jens G Lohr, Molly Schleicher, Emily Lipscomb, Andrea Saltzman, Nelly M Oliver, Lori Marini, Adrienne G Waks, Lauren C Harshman, Sara M Tolaney, Eliezer M Van Allen, Eric P Winer, Nancy U Lin, Mari Nakabayashi, Mary-Ellen Taplin, Cory M Johannessen, Levi A Garraway, Todd R Golub, Jesse S Boehm, Nikhil Wagle, Gad Getz, J Christopher Love, Matthew Meyerson

Abstract

Whole-exome sequencing of cell-free DNA (cfDNA) could enable comprehensive profiling of tumors from blood but the genome-wide concordance between cfDNA and tumor biopsies is uncertain. Here we report ichorCNA, software that quantifies tumor content in cfDNA from 0.1× coverage whole-genome sequencing data without prior knowledge of tumor mutations. We apply ichorCNA to 1439 blood samples from 520 patients with metastatic prostate or breast cancers. In the earliest tested sample for each patient, 34% of patients have ≥10% tumor-derived cfDNA, sufficient for standard coverage whole-exome sequencing. Using whole-exome sequencing, we validate the concordance of clonal somatic mutations (88%), copy number alterations (80%), mutational signatures, and neoantigens between cfDNA and matched tumor biopsies from 41 patients with ≥10% cfDNA tumor content. In summary, we provide methods to identify patients eligible for comprehensive cfDNA profiling, revealing its applicability to many patients, and demonstrate high concordance of cfDNA and metastatic tumor whole-exome sequencing.

Conflict of interest statement

T.R.G., L.A.G., N.W. are consultants and equity holders in Foundation Medicine, Inc. M.M. was previously a consultant and equity holder in Foundation Medicine, Inc. C.-Z.Z. is a consultant and equity holder in Pillar Biosciences. The authors have filed a patent application on methods described in this manuscript. The remaining authors declare no competing financial interests.

Figures

Fig. 1
Fig. 1
Copy number and tumor fractions from ULP-WGS. a cfDNA workflow. b Genome-wide copy number from 0.1× ULP-WGS of cfDNA from a healthy donor. c Genome-wide copy number from 25× WGS and 0.1× WGS of cell-free DNA from a metastatic breast cancer patient (MBC_315), and 1× WGS and WES of matched tumors from this patient. SCNA for tumor WES and cfDNA 25× coverage WGS were predicted using TITAN (“Methods”). d Comparison of copy ratios between ULP-WGS of cfDNA with deep (>10×) WGS of the same cfDNA sample, WGS (1×) of matched tumors from 22 metastatic breast cancer (MBC) patients, and WES (average mean target coverage 173×) of matched tumors from 41 MBC and prostate cancer (CRPC) patients. Log2 copy ratios were computed as normalized read coverage for each 1 Mb (WGS/ULP-WGS) and the mean of overlapping 50 kb bins (WES) after adjustment for tumor fraction/purity. The correlation of copy ratios between tumor and cfDNA was computed using Spearman rank correlation (coefficient ρ). F-measure (F1) is the harmonic mean of the CNA positive predictive value (precision) and sensitivity (recall) performance. Recall is defined as the proportion of SCNA gain/loss in tumor biopsy also observed in ULP-WGS of cfDNA (“Methods”). e Comparison of tumor fractions estimated from ULP-WGS and WES of cfDNA. Samples (n = 35) with similar tumor ploidy (difference < 0.75 and ploidy ≥1.5) estimated in both ULP-WGS and tumor WES are shown. The correlation between the two data types was calculated using Pearson correlation (coefficient r). Red line denotes y = x. WES tumor fractions were estimated using ABSOLUTE (shown) and TITAN (Supplementary Fig. 15, Supplementary Data 6)
Fig. 2
Fig. 2
Comparison of whole-exome sequencing of cfDNA to whole-exome sequencing of matched tumor biopsies. a Fraction of clonal (≥0.9 cancer cell fraction, CCF) and subclonal (<0.9 CCF) SSNVs detected by MuTect in WES of tumor biopsies and confirmed (i.e., supported by ≥3 variant reads) in WES of cfDNA. Sites with <3 reads that had power <0.9 for mutation calling were not included when computing the fraction of SNVs confirmed (“Methods”). b Fraction of clonal and subclonal SSNVs detected in WES of cfDNA and confirmed in WES of tumor biopsies. For 18 patients with WES of cfDNA at a second time point t2, SSNVs not detected in the matched tumor biopsy but confirmed at t2 are indicated with black. c Analysis of clonal dynamics in an ER+ breast cancer patient diagnosed with metastatic disease 1.5 years (yrs) prior to biopsy and cfDNA collection (t1, Day 0). Clustering analysis of CCF for SSNVs between matched tumor biopsy and cfDNA (t1) is shown in the left panel. The right panel shows the CCF of four mutation clusters, one containing ESR1 L536P (Subclonal Cluster 1, orange) and the other containing ESR1 D538G (Subclonal Cluster 2, light blue), at t1 and t2 (51 days apart) from a patient with ER+ metastatic breast cancer being treated with a SERD. The lymph node biopsy was taken at the same time as cfDNA t1. Mutations were clustered by the CCFs for each pair of samples using Phylogic (“Methods”). Error bars represent the 95% credible interval of the joint posterior density of the clusters. Mutations, excluding indels, having ≥90% estimated power based on coverage in both samples are shown; clusters with fewer than three mutations are excluded. The number of mutations in each cluster is indicated in the legend in parentheses
Fig. 3
Fig. 3
Genomic alterations of known significance and applicability to large cohorts. a, b The alteration status of significantly mutated genes predicted by MutSig2CV, (a), focal SCNAs (b), and known cancer-associated genes are shown for cfDNA and tumor biopsies from 27 metastatic breast cancer (MBC) patients. Mutated genes with MutSig2CV q-value < 0.1 are statistically significant. Mutations that were exclusively detected in one sample may be present at low CCF in the other matched sample but were excluded from the frequency calculation. SCNA frequencies were computed for oncogenes (MYC to ERBB2) and tumor suppressors (BRCA1 to ATM) using only amplification and deletion status, respectively. Mutations were predicted using MuTect and SCNAs were predicted using ReCapSeg and ABSOLUTE. Red dot indicates distinct mutations in tumor and cfDNA. c Mutational signatures in whole-exome sequencing of cfDNA and tumor biopsies were predicted using a Bayesian non-negative matrix factorization (NMF) approach (“Methods”). Samples with predicted biallelic inactivation of BRCA1/2 are indicated in red and blue. Black line denotes y = x; blue line denotes model fit using linear least squares regression. d Neoantigen burden, defined as the number of predicted neoantigen SSNVs, was calculated using NetMHCpan (“Methods”). Black line denotes y = x; blue line denotes model fit using linear least squares regression. e Applicability to many patients with metastatic cancer. Tumor fractions estimated from ULP-WGS of cfDNA from 903 blood samples from 391 patients with metastatic breast cancer and 536 blood samples from 129 patients with metastatic prostate cancers. The earliest blood drawn for each patient is shown. Samples with coverage <0.05× were excluded

References

    1. Diehl F, et al. Circulating mutant DNA to assess tumor dynamics. Nat. Med. 2008;14:985–990. doi: 10.1038/nm.1789.
    1. Dawson S-J, 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. Newman AM, et al. An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage. Nat. Med. 2014;20:548–554. doi: 10.1038/nm.3519.
    1. Bettegowda C, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 2014;6:224ra24. doi: 10.1126/scitranslmed.3007094.
    1. Russo M, et al. Tumor heterogeneity and lesion-specific response to targeted therapy in colorectal cancer. Cancer Discov. 2015;6:147–153. doi: 10.1158/-15-1283.
    1. Lanman RB, et al. Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA. PLoS ONE. 2015;10:e0140712. doi: 10.1371/journal.pone.0140712.
    1. Newman AM, et al. Integrated digital error suppression for improved detection of circulating tumor DNA. Nat. Biotechnol. 2016;34:547–555. doi: 10.1038/nbt.3520.
    1. Murtaza M, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112. doi: 10.1038/nature12065.
    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.
    1. Butler TM, et al. Exome sequencing of cell-free DNA from metastatic cancer patients identifies clinically actionable mutations distinct from primary disease. PLoS ONE. 2015;10:e0136407. doi: 10.1371/journal.pone.0136407.
    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. Beroukhim R, et al. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463:899–905. doi: 10.1038/nature08822.
    1. Heitzer E, et al. Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 2013;5:30. doi: 10.1186/gm434.
    1. Leary RJ, et al. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 2012;4:162ra154. doi: 10.1126/scitranslmed.3004742.
    1. Chan KCA, et al. Noninvasive detection of cancer-associated genome-wide hypomethylation and copy number aberrations by plasma DNA bisulfite sequencing. Proc. Natl Acad. Sci. USA. 2013;110:18761–18768. doi: 10.1073/pnas.1313995110.
    1. Carter SL, et al. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 2012;30:413–421. doi: 10.1038/nbt.2203.
    1. Ha G, et al. TITAN: inference of copy number architectures in clonal cell populations from tumor whole-genome sequence data. Genome Res. 2014;24:1881–1893. doi: 10.1101/gr.180281.114.
    1. Snyder MW, Martin K, Hill AJ, Daza RM, Shendure J. Cell-free DNA comprises an in vivo nucleosome footprint that informs its tissues-of-origin. Cell. 2016;164:57–68. doi: 10.1016/j.cell.2015.11.050.
    1. Szpechcinski A, et al. Cell-free DNA levels in plasma of patients with non-small-cell lung cancer and inflammatory lung disease. Br. J. Cancer. 2015;113:476–483. doi: 10.1038/bjc.2015.225.
    1. Robinson D, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–1228. doi: 10.1016/j.cell.2015.05.001.
    1. Grasso CS, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487:239–243. doi: 10.1038/nature11125.
    1. Van Allen EM, et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nat. Med. 2014;20:682–688. doi: 10.1038/nm.3559.
    1. Lawrence MS, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. doi: 10.1038/nature12213.
    1. Lawrence MS, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501. doi: 10.1038/nature12912.
    1. Ciriello G, et al. Comprehensive molecular portraits of invasive lobular breast cancer. Cell. 2015;163:506–519. doi: 10.1016/j.cell.2015.09.033.
    1. Cohen, O. et al. Whole Exome and Transcriptome Sequencing of Resistant ER+ Metastatic Breast Cancer. In Proc. 2016 San Antonio Breast Cancer Symposium Abstr. S1-01 (2016).
    1. Alexandrov LB, Nik-Zainal S, Siu HC, Leung SY, Stratton MR. A mutational signature in gastric cancer suggests therapeutic strategies. Nat. Commun. 2015;6:8683. doi: 10.1038/ncomms9683.
    1. Kasar S, et al. Whole-genome sequencing reveals activation-induced cytidine deaminase signatures during indolent chronic lymphocytic leukaemia evolution. Nat. Commun. 2015;6:8866. doi: 10.1038/ncomms9866.
    1. Kim J, et al. Somatic ERCC2 mutations are associated with a distinct genomic signature in urothelial tumors. Nat. Genet. 2016;48:600–606. doi: 10.1038/ng.3557.
    1. Alexandrov LB, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421. doi: 10.1038/nature12477.
    1. Mateo J, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N. Engl. J. Med. 2015;373:1697–1708. doi: 10.1056/NEJMoa1506859.
    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. Hoof I, et al. NetMHCpan, a method for MHC class i binding prediction beyond humans. Immunogenetics. 2009;61:1–13. doi: 10.1007/s00251-008-0341-z.
    1. Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 2015;33:1152–1158. doi: 10.1038/nbt.3344.
    1. Nielsen M, et al. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS ONE. 2007;2:e796. doi: 10.1371/journal.pone.0000796.
    1. Curtis C, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486:346–352.
    1. Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412.
    1. Gundem G, et al. The evolutionary history of lethal metastatic prostate cancer. Nature. 2015;520:353–357. doi: 10.1038/nature14347.
    1. Brastianos PK, et al. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 2015;5:1164–1177. doi: 10.1158/-15-0369.
    1. Sun K, et al. Plasma DNA tissue mapping by genome-wide methylation sequencing for noninvasive prenatal, cancer, and transplantation assessments. Proc. Natl Acad. Sci. USA. 2015;112:E5503–E5512. doi: 10.1073/pnas.1508736112.
    1. Lohr JG, et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat. Biotechnol. 2014;32:479–484. doi: 10.1038/nbt.2892.
    1. Oh WK, et al. Development of an integrated prostate cancer research information system. Clin. Genitourin. Cancer. 2006;5:61–66. doi: 10.3816/CGC.2006.n.019.
    1. Ha G, et al. Integrative analysis of genome-wide loss of heterozygosity and monoallelic expression at nucleotide resolution reveals disrupted pathways in triple-negative breast cancer. Genome Res. 2012;22:1995–2007. doi: 10.1101/gr.137570.112.
    1. Van Loo P, et al. Allele-specific copy number analysis of tumors. Proc. Natl Acad. Sci. USA. 2010;107:1–6. doi: 10.1073/iti0110107.
    1. Yau C, et al. A statistical approach for detecting genomic aberrations in heterogeneous tumor samples from single nucleotide polymorphism genotyping data. Genome Biol. 2010;11:R92.
    1. Chapman MA, et al. Initial genome sequencing and analysis of multiple myeloma. Nature. 2011;471:467–472. doi: 10.1038/nature09837.
    1. DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 2011;43:491–498. doi: 10.1038/ng.806.
    1. Berger MF, et al. The genomic complexity of primary human prostate cancer. Nature. 2012;470:214–220. doi: 10.1038/nature09744.
    1. Cibulskis K, et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics. 2011;27:2601–2602. doi: 10.1093/bioinformatics/btr446.
    1. Costello M, et al. Discovery and characterization of artifactual mutations in deep coverage targeted capture sequencing data due to oxidative dna damage during sample preparation. Nucleic Acids Res. 2013;41:e67. doi: 10.1093/nar/gks1443.
    1. Ramos AH, et al. Oncotator: cancer variant annotation tool. Hum. Mutat. 2015;36:E2423–E2429. doi: 10.1002/humu.22771.
    1. Saunders CT, et al. Strelka: accurate somatic small-variant calling from sequenced tumor- normal sample pairs. Bioinformatics. 2012;28:1811–1817. doi: 10.1093/bioinformatics/bts271.
    1. Olshen AB, Venkatraman ES, Lucito R, Wigler M. Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics. 2004;5:557–572. doi: 10.1093/biostatistics/kxh008.
    1. Venkatraman ES, Olshen AB. A faster circular binary segmentation algorithm for the analysis of array CGH data. Bioinformatics. 2007;23:657–663. doi: 10.1093/bioinformatics/btl646.
    1. McKenna A, et al. The genome analysis toolkit: a mapreduce framework for analyzing next- generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110.
    1. Landau DA, et al. Evolution and impact of subclonal mutations in chronic lymphocytic leukemia. Cell. 2013;152:714–726. doi: 10.1016/j.cell.2013.01.019.
    1. Harrow J, et al. GENCODE: the reference human genome annotation for the ENCODE project. Genome Res. 2012;22:1760–1774. doi: 10.1101/gr.135350.111.
    1. Stachler MD, et al. Paired exome analysis of Barrett’s esophagus and adenocarcinoma. Nat. Genet. 2015;47:1047–1055. doi: 10.1038/ng.3343.
    1. Kasar S, et al. Whole-genome sequencing reveals activation-induced cytidine deaminase signatures during indolent chronic lymphocytic leukaemia evolution. Nat. Commun. 2015;6:8866. doi: 10.1038/ncomms9866.
    1. Tan VYF, Fevotte C. Automatic relevance determination in nonnegative matrix factorization with the beta-divergence. IEEE Trans. Pattern Anal. Mach. Intell. 2013;35:1592–1605. doi: 10.1109/TPAMI.2012.240.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Campbell PJ, Stratton MR. Deciphering signatures of mutational processes operative in human cancer. Cell Rep. 2013;3:246–259. doi: 10.1016/j.celrep.2012.12.008.
    1. Shukla SA, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat. Biotechnol. 2015;33:1152–1158. doi: 10.1038/nbt.3344.
    1. Hoof I, et al. NetMHCpan, a method for MHC class I binding prediction beyond humans. Immunogenetics. 2009;61:1–13. doi: 10.1007/s00251-008-0341-z.

Source: PubMed

3
S'abonner