An ultrasensitive method for quantitating circulating tumor DNA with broad patient coverage

Aaron M Newman, Scott V Bratman, Jacqueline To, Jacob F Wynne, Neville C W Eclov, Leslie A Modlin, Chih Long Liu, Joel W Neal, Heather A Wakelee, Robert E Merritt, Joseph B Shrager, Billy W Loo Jr, Ash A Alizadeh, Maximilian Diehn, Aaron M Newman, Scott V Bratman, Jacqueline To, Jacob F Wynne, Neville C W Eclov, Leslie A Modlin, Chih Long Liu, Joel W Neal, Heather A Wakelee, Robert E Merritt, Joseph B Shrager, Billy W Loo Jr, Ash A Alizadeh, Maximilian Diehn

Abstract

Circulating tumor DNA (ctDNA) is a promising biomarker for noninvasive assessment of cancer burden, but existing ctDNA detection methods have insufficient sensitivity or patient coverage for broad clinical applicability. Here we introduce cancer personalized profiling by deep sequencing (CAPP-Seq), an economical and ultrasensitive method for quantifying ctDNA. We implemented CAPP-Seq for non-small-cell lung cancer (NSCLC) with a design covering multiple classes of somatic alterations that identified mutations in >95% of tumors. We detected ctDNA in 100% of patients with stage II-IV NSCLC and in 50% of patients with stage I, with 96% specificity for mutant allele fractions down to ∼0.02%. Levels of ctDNA were highly correlated with tumor volume and distinguished between residual disease and treatment-related imaging changes, and measurement of ctDNA levels allowed for earlier response assessment than radiographic approaches. Finally, we evaluated biopsy-free tumor screening and genotyping with CAPP-Seq. We envision that CAPP-Seq could be routinely applied clinically to detect and monitor diverse malignancies, thus facilitating personalized cancer therapy.

Figures

Figure 1. Development of CA ncer P…
Figure 1. Development of CAncer Personalized Profiling by deep Sequencing (CAPP-Seq)
(a) Schematic depicting design of CAPP-Seq selectors and their application for assessing circulating tumor DNA. (b) Multi-phase design of the NSCLC selector. Phase 1: Genomic regions harboring known and suspected driver mutations in NSCLC are captured. Phases 2–4: Addition of exons containing recurrent SNVs using WES data from lung adenocarcinomas and squamous cell carcinomas from TCGA (n = 407). Regions were selected iteratively to maximize the number of mutations per tumor while minimizing selector size. Recurrence index = total unique patients with mutations covered per kb of exon. Phases 5,6: Exons of predicted NSCLC drivers, and introns and exons harboring breakpoints in rearrangements involving ALK, ROS1, and RET were added. Bottom: increase of selector length during each design phase. (c) Analysis of the number of SNVs per lung adenocarcinoma covered by the NSCLC selector in the TCGA WES cohort (Training; n = 229) and an independent lung adenocarcinoma WES data set (Validation; n = 183). Results are compared to selectors randomly sampled from the exome (P < 1.0 × 10−6 for the difference between random selectors and the NSCLC selector). (d) Analytical modeling of CAPP-Seq, whole exome and whole genome sequencing for different detection limits of tumor circulating DNA in plasma. Calculations are based on the median number of mutations detected per NSCLC for CAPP-Seq (i.e., 4) and the reported number of mutations in NSCLC exomes and genomes. Additional details are described in Methods. The vertical dotted line represents the median fraction of tumor-derived circulating DNA detected in plasma from patients in this study.
Figure 2. Analytical performance
Figure 2. Analytical performance
(ac) Quality parameters from a representative CAPP-Seq analysis of plasma DNA, including length distribution of sequenced circulating DNA fragments (a), and depth of sequencing coverage across all genomic regions in the selector (b). (c) Variation in sequencing depth across plasma DNA samples from four patients. Orange envelope represents s.e.m. (d) Analysis of background rate for 40 plasma DNA samples collected from 13 patients with NSCLC and five healthy individuals (Supplementary Methods). (e) Analysis of biological background in d focusing on 107 recurrent somatic mutations from a previously reported SNaPshot panel. Mutations found in a given patient’s tumor were excluded. The mean frequency over all subjects was ~0.01%. A single outlier mutation (TP53 R175H) is indicated by an orange diamond. (f) Individual mutations from e ranked by most to least recurrent, according to mean frequency across the 40 plasma DNA samples. The p-value threshold of 0.01 (horizontal line) corresponds to the 99th percentile of global selector background in d. (g) Dilution series analysis of expected versus observed frequencies of mutant alleles using CAPP-Seq. Dilution series were generated by spiking fragmented HCC78 DNA into control circulating DNA. (h) Analysis of the effect of the number of SNVs considered on the estimates of fractional abundance (95% confidence intervals shown in gray). (i) Analysis of the effect of the number of SNVs considered on the mean correlation coefficient between expected and observed cancer fractions (blue dashed line) using data from panel h. 95% confidence intervals are shown for e,f. Statistical variation for g is shown as s.e.m.
Figure 3. Sensitivity and specificity analysis
Figure 3. Sensitivity and specificity analysis
(a) Receiver Operating Characteristic (ROC) analysis of plasma DNA samples from pre-treatment samples and healthy controls, divided into all stages (n = 13 patients) and stages II–IV (n = 9 patients). Area Under the Curve (AUC) values are significant at P < 0.0001. Sn, sensitivity; Sp, specificity. (b) Raw data related to a. TP, true positive; FP, false positive; TN, true negative; FN, false negative. (c) Concordance between tumor volume, measured by CT or PET/CT, and pg mL−1 of ctDNA from pretreatment samples (n = 9), measured by CAPP-Seq. Patients P6 and P9 were excluded due to inability to accurately assess tumor volume and differences related to the capture of fusions, respectively (see Supplementary Methods). Of note, linear regression was performed in non-log space; the log-log axes and dashed diagonal line are for display purposes only.
Figure 4. Noninvasive detection and monitoring of…
Figure 4. Noninvasive detection and monitoring of circulating tumor DNA
(ah) Disease monitoring using CAPP-Seq. (a,b) Disease burden changes in response to treatment in a patient with stage III NSCLC using SNVs and an indel (a), and a patient with stage IV NSCLC using three rearrangement breakpoints (b). (c) Concordance between different reporters (SNVs and a fusion) in a patient with stage IV NSCLC. (d) Detection of a subclonal EGFR T790M resistance mutation in a patient with stage IV NSCLC. The fractional abundance of the dominant clone and T790M-containing clone are shown in the primary tumor (left) and plasma samples (right). (e,f) CAPP-Seq results from post-treatment plasma DNA samples are predictive of clinical outcomes in a patient with stage IIB NSCLC (e) and a patient with stage IIIB NSCLC (f). (g,h) Monitoring of tumor burden following complete tumor resection (g) and Stereotactic Ablative Radiotherapy (SABR) (h) for two patients with stage IB NSCLC. (i) Exploratory analysis of the potential application of CAPP-Seq for biopsy-free tumor genotyping or cancer screening. All plasma DNA samples from patients in Table 1 were examined for the presence of mutant allele outliers without knowledge of the primary tumor mutations (see Supplementary Methods); samples with detectable mutations are shown, along with three samples assumed to be cancer-negative (P1-2, P1-3 and P16-3; Supplementary Methods). The lowest fraction of ctDNA among positive samples was ~0.4% (dashed horizontal line). Error bars in d represent s.e.m. (a,b,eh) Scale bars, 10 cm. Tu, tumor; Ef, pleural effusion; SD, stable disease; PD, progressive disease; PR, partial response; CR, complete response; DOD, dead of disease.

References

    1. Taniguchi K, et al. Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res. 2011;17:7808–7815.
    1. Rosell R, et al. Screening for epidermal growth factor receptor mutations in lung cancer. N Engl J Med. 2009;361:958–967.
    1. Kuang Y, et al. Noninvasive detection of EGFR T790M in gefitinib or erlotinib resistant non-small cell lung cancer. Clin Cancer Res. 2009;15:2630–2636.
    1. Gautschi O, et al. Origin and prognostic value of circulating KRAS mutations in lung cancer patients. Cancer Lett. 2007;254:265–273.
    1. Leary RJ, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med. 2010;2:20ra14.
    1. McBride DJ, et al. Use of cancer-specific genomic rearrangements to quantify disease burden in plasma from patients with solid tumors. Genes, Chromosomes & Cancer. 2010;49:1062–1069.
    1. He J, et al. IgH gene rearrangements as plasma biomarkers in Non- Hodgkin’s lymphoma patients. Oncotarget. 2011;2:178–185.
    1. Forshew T, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4:136ra168.
    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.
    1. Narayan A, et al. Ultrasensitive measurement of hotspot mutations in tumor DNA in blood using error-suppressed multiplexed deep sequencing. Cancer Res. 2012;72:3492–3498.
    1. Dawson SJ, et al. Analysis of Circulating Tumor DNA to Monitor Metastatic Breast Cancer. N Engl J Med. 2013
    1. Murtaza M, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497:108–112.
    1. Crowley E, Di Nicolantonio F, Loupakis F, Bardelli A. Liquid biopsy: monitoring cancer-genetics in the blood. Nature Rev Clinical Oncol. 2013;10:472–484.
    1. Forbes SA, et al. COSMIC (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res. 2010;38:D652–657.
    1. Ding L, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455:1069–1075.
    1. Youn A, Simon R. Identifying cancer driver genes in tumor genome sequencing studies. Bioinformatics. 2011;27:175–181.
    1. Bergethon K, et al. ROS1 rearrangements define a unique molecular class of lung cancers. J Clin Oncol. 2012;30:863–870.
    1. Kwak EL, et al. Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer. N Engl J Med. 2010;363:1693–1703.
    1. Pao W, Hutchinson KE. Chipping away at the lung cancer genome. Nat Med. 2012;18:349–351.
    1. Imielinski M, et al. Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell. 2012;150:1107–1120.
    1. Govindan R, et al. Genomic landscape of non-small cell lung cancer in smokers and never-smokers. Cell. 2012;150:1121–1134.
    1. Koivunen JP, et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clin Cancer Res. 2008;14:4275–4283.
    1. Rikova K, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell. 2007;131:1190–1203.
    1. Fan HC, Blumenfeld YJ, Chitkara U, Hudgins L, Quake SR. Noninvasive diagnosis of fetal aneuploidy by shotgun sequencing DNA from maternal blood. Proc Natl Acad Sci U S A. 2008;105:16266–16271.
    1. Su Z, et al. A platform for rapid detection of multiple oncogenic mutations with relevance to targeted therapy in non-small-cell lung cancer. J Mol Diagn. 2011;13:74–84.
    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.
    1. Iyengar P, Timmerman RD. Stereotactic ablative radiotherapy for non-small cell lung cancer: rationale and outcomes. Journal of the National Comprehensive Cancer Network : JNCCN. 2012;10:1514–1520.
    1. Nesbitt JC, Putnam JB, Jr, Walsh GL, Roth JA, Mountain CF. Survival in early-stage non-small cell lung cancer. Annals Thoracic Surg. 1995;60:466–472.
    1. Aberle DR, et al. Reduced Lung-Cancer Mortality with Low-Dose Computed Tomographic Screening. N Engl J Med. 2011;365:395–409.
    1. Diehl F, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci U S A. 2005;102:16368–16373.
    1. Diehl F, et al. Analysis of mutations in DNA isolated from plasma and stool of colorectal cancer patients. Gastroenterology. 2008;135:489–498.
    1. Chan KC, et al. Cancer genome scanning in plasma: detection of tumor-associated copy number aberrations, single-nucleotide variants, and tumoral heterogeneity by massively parallel sequencing. Clin Chem. 2013;59:211–224.
    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.
    1. Schmitt MW, et al. Detection of ultra-rare mutations by next-generation sequencing. Proc Natl Acad Sci U S A. 2012;109:14508–14513.
    1. Shiroguchi K, Jia TZ, Sims PA, Xie XS. Digital RNA sequencing minimizes sequence-dependent bias and amplification noise with optimized single-molecule barcodes. Proc Natl Acad Sci U S A. 2012;109:1347–1352.
    1. Quail MA, et al. Optimal enzymes for amplifying sequencing libraries. Nat Methods. 2012;9:10–11.
    1. Oyola SO, et al. Optimizing Illumina next-generation sequencing library preparation for extremely AT-biased genomes. BMC Genomics. 2012;13:1.
    1. Fisher S, et al. A scalable, fully automated process for construction of sequence-ready human exome targeted capture libraries. Genome Biol. 2011;12:R1.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25:2078–2079.
    1. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–842.
    1. Koboldt DC, et al. VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing. Genome Res. 2012;22:568–576.
    1. Fisher RA. Statistical methods for research workers. Oliver and Boyd; Edinburgh, London: 1925.

Source: PubMed

3
Abonnieren