Purpose: Tumor genotyping is a powerful tool for guiding non–small cell lung cancer (NSCLC) care; however, comprehensive tumor genotyping can be logistically cumbersome. To facilitate genotyping, we developed a next-generation sequencing (NGS) assay using a desktop sequencer to detect actionable mutations and rearrangements in cell-free plasma DNA (cfDNA).

Experimental Design: An NGS panel was developed targeting 11 driver oncogenes found in NSCLC. Targeted NGS was performed using a novel methodology that maximizes on-target reads, and minimizes artifact, and was validated on DNA dilutions derived from cell lines. Plasma NGS was then blindly performed on 48 patients with advanced, progressive NSCLC and a known tumor genotype, and explored in two patients with incomplete tumor genotyping.

Results: NGS could identify mutations present in DNA dilutions at ≥0.4% allelic frequency with 100% sensitivity/specificity. Plasma NGS detected a broad range of driver and resistance mutations, including ALK, ROS1, and RET rearrangements, HER2 insertions, and MET amplification, with 100% specificity. Sensitivity was 77% across 62 known driver and resistance mutations from the 48 cases; in 29 cases with common EGFR and KRAS mutations, sensitivity was similar to droplet digital PCR. In two cases with incomplete tumor genotyping, plasma NGS rapidly identified a novel EGFR exon 19 deletion and a missed case of MET amplification.

Conclusions: Blinded to tumor genotype, this plasma NGS approach detected a broad range of targetable genomic alterations in NSCLC with no false positives including complex mutations like rearrangements and unexpected resistance mutations such as EGFR C797S. Through use of widely available vacutainers and a desktop sequencing platform, this assay has the potential to be implemented broadly for patient care and translational research. Clin Cancer Res; 22(4); 915–22. ©2015 AACR.

See related commentary by Tsui and Berger, p. 790

Translational Relevance

Noninvasive genotyping of cell-free plasma DNA (cfDNA) is a potentially powerful tool for advancing cancer care and translational research, but most established assays are PCR-based and limited to detection of hotspot mutations. Here, we describe the development of a novel rapid targeted next-generation sequencing (NGS) assay for study of cfDNA. Studying 48 cases using a desktop sequencer, this assay was able to detect targetable oncogenic genomic alterations and resistance mechanisms in advanced non–small cell lung cancer without any false positives. The comprehensive coverage afforded by this assay while utilizing a widely available NGS platform has great potential for broad uptake as a tool for noninvasive tumor genotyping.

Genotype-directed targeted therapies are revolutionizing cancer care. Genomic alterations in genes such as EGFR, ALK, KRAS, and BRAF have been validated as powerful predictive biomarkers in the management of non–small cell lung cancer (NSCLC), colorectal cancer, and melanoma; it is now standard to test for these mutations to personalize treatment decisions (1–7). Development of new genotype-directed therapies is widespread in solid tumor oncology, leading to increasing application of next-generation sequencing (NGS) panels that can test tumor biopsies for a wide range of potentially targetable mutations (8, 9). However, routine use of NGS for tumor genotyping presents practical challenges including the availability of adequate biopsy specimens, slow turnaround time, and the need for repeat biopsies after development of drug resistance (9). Given these challenges, it is clear that there is an unmet need for noninvasive assays that can broadly detect actionable genomic alterations.

Many groups, including our own, have investigated noninvasive tumor genotyping of cell-free plasma DNA (cfDNA) as an alternative to tissue genotyping (10–15). Rather than studying circulating cells, these technologies study the free floating DNA contained in the plasma; in advanced cancer patients, a portion of this cfDNA may be derived from the tumor. Plasma genotyping has the potential to be less invasive and faster than tumor genotyping, while also allowing serial assessment of genotype during development of treatment resistance. We recently reported on a highly specific and rapid droplet digital PCR (ddPCR) assay for quantifying the concentration of EGFR and KRAS mutations in cfDNA of advanced NSCLC patients (16, 17). Such PCR-based plasma assays test for mutations at a single site in a gene, but are limited by their inability to detect more complex genomic alterations such as chromosomal rearrangements and their inability to multiplex across several genes. Others have studied NGS of cfDNA using PCR amplicons or tagged DNA baits to enrich for target DNA sequences; however, many such assays are unable to detect rearrangements, whereas other assays rely on massive sequencing and computational processing resulting in unacceptable costs and slow turnaround time (11, 18). Although detection of mutant cfDNA present at low concentration is possible with these approaches despite the more abundant wild-type (germline) DNA, a universal challenge with these highly sensitive genotyping assays is the risk of false positives due to PCR artifact.

In this study, we piloted a novel-targeted NGS approach for the detection of driver mutations and rearrangements in cfDNA from advanced NSCLC patients. Taking cues from traditional hybrid capture approaches that isolate genomic subsets by pull down with probes to genes of interest, our methodology improves on key steps during library generation to reduce sequencing demands and turnaround times. First, to maximize on-target reads to ∼90%, a two-step pull-down process was used that includes both a thermodynamically controlled hybridization step and a kinetically controlled extension step under conditions that neutralize GC bias. Then, to improve signal-to-noise ratio, tags were connected to each captured DNA fragment, so that every read is anchored to its clonal family and to its pull-down probe of origin, facilitating identification of low-frequency mutant alleles and quantification of subtle changes in gene copy numbers (Fig. 1 and Supplementary Methods). We hypothesized that this approach would allow for accurate detection of a broad range of targetable genotypes, including insertions/deletions and rearrangements, in cfDNA from advanced NSCLC patients. Our goal was to leverage a desktop sequencing platform to enable a rapid turnaround time and facilitate widespread clinical adoption.

Figure 1.

Key differences between standard hybrid capture (left) and bias-corrected NGS (right). A, mono-, di-, and trimeric nucleosome cfDNA fragments ranging from 130 to 480 basepairs are isolated. B, in standard hybrid capture, cfDNA fragments are end-repaired and ligated with single primers. In contrast, bias-corrected NGS uses multifunctional adaptors that include sequences for single-primer amplification (red), tags for sample identification (green), and sequence identification tags (blue) that, in conjunction with the fragmentation site (blue dot) identify unique sequence clones. C, in standard hybridization cfDNA fragments are captured with large capture probes (up to 120 bp) that span the genetic region of interest and may result in off-target fragments being isolated (e.g., daisy-chaining off-target DNA). Bias-corrected NGS uses small capture probes (∼40 bp) that are designed to be adjacent to the region of interest. Primer extension of fragments copies genomic and adaptor sequences. Finally, amplification with tailed PCR primers creates sequencing ready clones. D, although both approaches allow sequencing of gene rearrangements, large capture probes designed to target one gene will inefficiently target fragments containing a large amount of fusion partner gene sequence, resulting in poor sensitivity. In bias-corrected NGS, gene junction and partner gene sequence is replicated during primer extension. E, in standard hybrid capture all pulled-down cfDNA (specific and nonspecific) is amplified and sequenced without knowing the exact read or probe which captured the fragment. In bias-corrected NGS, READ_1 identifies the sample ID and the unique sequence identifiers, whereas READ_2 identifies the probe that pulled down each clone, facilitating read analysis, and probe optimization.

Figure 1.

Key differences between standard hybrid capture (left) and bias-corrected NGS (right). A, mono-, di-, and trimeric nucleosome cfDNA fragments ranging from 130 to 480 basepairs are isolated. B, in standard hybrid capture, cfDNA fragments are end-repaired and ligated with single primers. In contrast, bias-corrected NGS uses multifunctional adaptors that include sequences for single-primer amplification (red), tags for sample identification (green), and sequence identification tags (blue) that, in conjunction with the fragmentation site (blue dot) identify unique sequence clones. C, in standard hybridization cfDNA fragments are captured with large capture probes (up to 120 bp) that span the genetic region of interest and may result in off-target fragments being isolated (e.g., daisy-chaining off-target DNA). Bias-corrected NGS uses small capture probes (∼40 bp) that are designed to be adjacent to the region of interest. Primer extension of fragments copies genomic and adaptor sequences. Finally, amplification with tailed PCR primers creates sequencing ready clones. D, although both approaches allow sequencing of gene rearrangements, large capture probes designed to target one gene will inefficiently target fragments containing a large amount of fusion partner gene sequence, resulting in poor sensitivity. In bias-corrected NGS, gene junction and partner gene sequence is replicated during primer extension. E, in standard hybrid capture all pulled-down cfDNA (specific and nonspecific) is amplified and sequenced without knowing the exact read or probe which captured the fragment. In bias-corrected NGS, READ_1 identifies the sample ID and the unique sequence identifiers, whereas READ_2 identifies the probe that pulled down each clone, facilitating read analysis, and probe optimization.

Close modal

Plasma NGS

Targeted NGS of cell-line DNA and plasma cfDNA was performed at Resolution Bioscience as described in Supplementary Methods. Chimeric gene fusions were detected using tiled probes that allow sequencing-based discovery of de novo rearrangements (Supplementary Fig. S1 and Supplementary Table S1).

Plasma ddPCR

For comparison to plasma NGS, plasma ddPCR was performed using an established and validated assay which has been described previously (16). Briefly, this assay emulsifies extracted plasma cfDNA into thousands of droplets which subsequently undergo individual PCR with custom fluorescently labeled probes designed to detect EGFR L858R, EGFR exon 19 deletions, or KRAS G12X (16). Individual droplets are then read in a flow cytometer and the number of positive droplets are quantified (Bio-Rad). Each sample is analyzed in triplicate.

Cell line validation

The targeted NGS panel was validated using genomic DNA from 14 independent, genetically-annotated cell lines harboring four gene fusions, 19 point mutations, and two insertions/deletions (Supplemental Table S2). Cell lines were combined into two separate DNA pools, each containing the genomes of seven cell lines, and systematically blended with normal, wild-type DNA to produce admixtures at 2.5%, 1.0%, 0.4%, and 0.1% dilutions. Prior to NGS, DNA pools were acoustically fragmented to an average size distribution centered around 165 bp and purified by two-sided SPRI to give fragment profiles of 150 to 200 bp that closely approximate cfDNA. Cell lines for the analytical validation experiment were obtained from ATCC (A549, H2228, SK-MEL-2, H1666, SW48), RIKEN (Lc-2/ad), the Broad institute (H1781, SW480, HCT116, H2347, HCC78), and the NCI (H3122). PC-9 and H1975 cells were obtained from the laboratory of Dr. Pasi Jänne. All cell lines were validated to be correct by short tandem repeat (STR) analyses.

Patient population

Patients were identified during their routine lung cancer care at Dana-Farber Cancer Institute. Patients were deemed eligible if they had advanced NSCLC with a known tumor genotype, either untreated or progressive on therapy. Tumor genotyping was performed as part of routine care, either using conventional genotyping assays (PCR, FISH) or a targeted NGS panel when available (5, 19). All patients consented to plasma collection and analysis on an IRB-approved prospective plasma collection protocol or associated correlative science protocols. Following clinical validation, plasma NGS was explored in two patients with high suspicion of a targetable genotype missed on tumor genotyping.

Plasma collection

Plasma was collected prior to initiation of therapy for untreated or progressive advanced NSCLC. Whole blood was collected into 10 mL EDTA containing “purple-top” vacutainer tubes, centrifuged for 10 minutes at 1,200 × g and the plasma supernatant was further cleared by centrifugation for 10 minutes at 3,000 × g. Cleared plasma was stored in cryostat tubes at −80°C until use. cfDNA was isolated using the QIAmp Circulating Nucleic Acid Kit (Qiagen) according to the manufacturer's protocol. DNA was eluted in AVE buffer (100 μL) and stored at −80°C until use.

Blinding of specimens

To ensure data integrity in these experiments, the samples were identified only by sample key. Only the clinical team (GRO, AGS, RSA) that identified patients for study had access to the tissue genotype results. The teams involved in ddPCR and NGS analysis were blinded during data acquisition (plasma isolation, cfDNA extraction, library generation, NGS, and ddPCR analysis) and unblinding was done after NGS and ddPCR results had been reported to the clinical team.

A probe set was developed that covers portions of 11 genes known to be targetable oncogenic drivers in NSCLC. Selected coding regions of eight genes were sequenced (KRAS, EGFR, ALK, HER2, BRAF, NRAS, PIK3CA, MET, and MEK1). In addition, intronic probes were designed to detect genome-level rearrangements that create chimeric gene fusions in ALK, ROS1, and RET. The coding regions of the tumor suppressor TP53 were also included as a control because this gene is commonly mutated in NSCLC. Analysis of the performance of this probe set on plasma DNA showed >80% on-target percentage, which compares favorably with the less than 50% on-target percentage seen in previously published data using standard hybridization selection on plasma DNA (Supplementary Fig. S2 and ref. 18).

The targeted NGS panel was initially validated with dilutions of cell line DNA. Four different dilutions of two DNA pools were sequenced, each derived from seven cell lines harboring previously characterized mutations (Supplemental Table S2). Dilutions of 2.5% to 0.1% resulted in calculated allelic frequencies ranging from 4.5% to 0.01% (Supplementary Fig. S3). Variant calling algorithms (Supplementary Methods) were able to identify mutations that were present at 0.1% or greater with sensitivity and specificity of 88% and 100%. Diagnostic performance improved to 100% sensitivity and specificity for mutations present at an allelic frequency 0.4% or greater (Supplementary Fig. S3 and Table S2). This preliminary analysis of the cell lines allowed the setting of thresholds that ensured high specificity in the subsequent analysis of patient samples.

After validating the NGS platform with DNA dilutions, plasma samples from 48 patients with advanced NSCLC were studied, blinded to the tumor genotyping results (Supplemental Table S3). The median age of these patients was 57, 61% were female and 92% had extra-thoracic metastatic disease. Mean reads per sample was 8.9 million, with a mean coverage per base of 983 unique reads.

The sensitivity of the NGS platform was first studied in 29 of the 48 patients known by tumor genotyping to harbor EGFR and KRAS driver mutations readily assayed with ddPCR (Fig. 2A). Using the tumor genotyping as the gold standard, ddPCR of plasma had a sensitivity of 86%, whereas NGS had a sensitivity of 79%, not significantly different (P = 0.43). Both methods were more sensitive when more cfDNA was available. The allelic fraction of the mutant allele in cfDNA, calculated as the number of mutant reads over wild-type reads, was closely correlated for plasma NGS and plasma ddPCR (Pearson correlation = 0.93, P < 0.001; Fig. 2B).

Figure 2.

Plasma NGS compared with known tumor genotype across a range of genomic equivalents (GE) in the sequencing library. The mutant allele frequency is provided when detected by the plasma genotyping assay (green circle) but not if undetected (red circle). In patients with common EGFR and KRAS mutations (A), plasma NGS has similar sensitivity to plasma ddPCR. Quantification of allelic frequency with plasma NGS and plasma ddPCR are closely correlated (B). In patients with rare genotypes (C), plasma NGS is able to detect a wide range of genomic alterations. In both groups of patients, the rate of detection by plasma NGS increases as the number of GE increases (A, C).

Figure 2.

Plasma NGS compared with known tumor genotype across a range of genomic equivalents (GE) in the sequencing library. The mutant allele frequency is provided when detected by the plasma genotyping assay (green circle) but not if undetected (red circle). In patients with common EGFR and KRAS mutations (A), plasma NGS has similar sensitivity to plasma ddPCR. Quantification of allelic frequency with plasma NGS and plasma ddPCR are closely correlated (B). In patients with rare genotypes (C), plasma NGS is able to detect a wide range of genomic alterations. In both groups of patients, the rate of detection by plasma NGS increases as the number of GE increases (A, C).

Close modal

Detection of rare mutations and rearrangements in cfDNA was next studied in a blinded fashion in 20 of the 48 patients with NSCLC known to harbor a rare mutation or rearrangement on tumor genotyping (Fig. 2C); these included 19 new cases plus one previously studied above, harboring both a KRAS mutation and a PIK3CA mutation. Plasma NGS was able to blindly detect six of eight cases with rearrangements and four cases with rare targetable HER2 and EGFR mutations. Sensitivity in this cohort was 75% (15/20), similar to the prior analysis (Fig. 2C). For each of the six rearrangements detected, the exact breakpoints and fusion partners could be mapped to the genome (Fig. 3A and Supplementary Fig. S4).

Figure 3.

Bias-corrected NGS of cfDNA identifies complex genomic alterations. A, sequencing of the intronic region of RET detects reads extending into KIF5B (inset), predicting a fusion of these two genes. B, in cfDNA from a case with known MET amplification (case 105), MET copy number is significantly increased compared with control probes (P < 0.001), which is not seen in cases without MET amplification. C, two mutations encoding for EGFR C797S are detected in cis with EGFR T790M after resistance to AZD9291. D, in a case with acquired T790M despite no apparent EGFR sensitizing mutation, plasma NGS detects a novel double-deletion in exon 19 of EGFR which would have been missed with many PCR-based genotyping assays.

Figure 3.

Bias-corrected NGS of cfDNA identifies complex genomic alterations. A, sequencing of the intronic region of RET detects reads extending into KIF5B (inset), predicting a fusion of these two genes. B, in cfDNA from a case with known MET amplification (case 105), MET copy number is significantly increased compared with control probes (P < 0.001), which is not seen in cases without MET amplification. C, two mutations encoding for EGFR C797S are detected in cis with EGFR T790M after resistance to AZD9291. D, in a case with acquired T790M despite no apparent EGFR sensitizing mutation, plasma NGS detects a novel double-deletion in exon 19 of EGFR which would have been missed with many PCR-based genotyping assays.

Close modal

Specificity was studied in these 48 patients, each with tumor genotyping positive for an oncogenic driver mutation in EGFR, KRAS, ALK, ROS1, RET, BRAF, or HER2. These oncogenes are established as nonoverlapping on tumor genotyping and are therefore ideal gold standards for assessment of false positives (5, 7). Specificity of the plasma NGS platform was 100% for these seven driver genotypes, with a false positive rate of 0% (95% confidence interval, 0%–10%).

Detection of resistance mutations was explored in 15 of the 48 patients who had plasma collected after development of acquired resistance to a tyrosine kinase inhibitor (Table 1). Of 12 patients with acquired resistance to erlotinib or afatinib, plasma NGS detected T790M in eight. One of the 12 patients had been refractory to erlotinib and afatinib despite harboring an EGFR exon 19 deletion, and tumor NGS had identified high MET amplification. Blinded to the tumor genotype, plasma NGS similarly detected MET amplification, as evidenced by a significant increase in MET copies compared with control (Fig. 3B). No resistance mutations were identified in the remaining three EGFR-mutant cases. Studying two patients who had developed acquired resistance to crizotinib, plasma NGS identified two point mutations in the ALK kinase domain in one case with an ALK rearrangement, and no ROS1 resistance mutations in the other case with a ROS1 rearrangement. One patient was studied who had previously developed T790M-positive resistance to erlotinib, and subsequently developed acquired resistance to the investigational EGFR kinase inhibitor AZD9291 (20); plasma NGS identified two different DNA mutations encoding for EGFR C797S (Fig. 3C), a mutation recently described as a common mediator of acquired resistance to AZD9291 (21). Fourteen of 15 cases had resistance biopsies available for genotyping. Tumor and plasma results were concordant in 12 (86%), whereas in two cases plasma NGS did not detect an acquired T790M mutation detected in tumor. Altogether, sensitivity for the 62 known driver and resistance mutations from the 48 cases was 77% (48/62).

Table 1.

Detection of resistance mechanisms using plasma NGS compared with tissue genotyping of rebiopsy specimens in patients with acquired resistance to kinase inhibitors

Detection of resistance mechanisms using plasma NGS compared with tissue genotyping of rebiopsy specimens in patients with acquired resistance to kinase inhibitors
Detection of resistance mechanisms using plasma NGS compared with tissue genotyping of rebiopsy specimens in patients with acquired resistance to kinase inhibitors

Finally, the plasma NGS assay was explored in two advanced NSCLC patients with high clinical suspicion of possessing a targetable genomic alteration which had been missed on prior tissue genotyping. The initial patient was a 66-year-old female never-smoker who had responded durably to empiric treatment with erlotinib and developed resistance; EGFR genotyping of a rebiopsy using a commercial PCR assay had identified T790M without any sensitizing mutation evident. Plasma ddPCR was first performed and detected 1508 copies/mL of an exon 19 deletion. Plasma NGS then confirmed the presence of a novel double-deletion in exon 19 of EGFR which would have been missed by many commercial PCR assays because these often detect only common exon 19 deletion variants (Fig. 3D). The second patient was a 28-year-old female never-smoker who had progressed on multiple lines of therapy, for whom previous tumor genotyping (including NGS) had revealed no targetable alterations despite four biopsies. Plasma NGS revealed high-level MET amplification which was subsequently confirmed by fluorescent in situ immunohistochemistry (MET:CEP7 ratio >5), and led to the initiation of crizotinib. For each case, turnaround time from blood draw to result reporting in this initial feasibility study was six business days.

In this blinded study, a targeted NGS of cfDNA from advanced NSCLC patients was able to accurately detect a broad range of targetable genomic alterations in NSCLC, including point mutations, insertions/deletions, and rearrangements, with no false positives. This report is the first, to our knowledge, to describe the accurate detection of ALK, ROS1, and RET rearrangements using a single plasma assay without prior knowledge of fusion partners. This represents a dramatic advance over PCR-based plasma genotyping assays which are limited to the detection of hotspot mutations in coding regions (10). This assay was also able to detect both canonical and novel resistance mechanisms, including MET amplification and EGFR C797S (22, 23).

Importantly, this approach uses widely available equipment such as standard EDTA-containing vacutainers and a desktop sequencing platform: any accredited molecular pathology lab with a MiSeq could, with the right technical expertise and bioinformatics support, implement this assay to guide the care of advanced NSCLC patients.

Although others have also studied plasma NGS of lung cancer, this is the first to describe comprehensive and blinded detection of a broad range of alterations with one clinic-ready assay. Detection of hotspot mutations using plasma NGS was described by Couraud and using the IonTorrent platform, achieving a sensitivity of 58% and 87% specificity but without the ability to detect rearrangements or amplifications (11). Newman and others recently demonstrated more comprehensive detection of lung cancer mutations in cfDNA and tumor tissue by hybrid capture using biotinylated oligonucleotide probes on a HiSeq (18); however, this study noted an inefficient capture of fusion rearrangements. Finally, targeted sequencing of cfDNA using PCR amplicons has been successful for detection of single nucleotide variant in multiple types of cancer (15); however, PCR amplicons are not trivial to multiplex and are inherently blind to gene rearrangements. The NGS platform described here overcomes the weaknesses of each of these prior approaches, allowing multiplexed detection of a broad range of alterations with no false positives using an efficient platform. Furthermore, turnaround time from time of blood draw can be as short as 6 days.

Intuitively, we found that the sensitivity of plasma NGS is improved in specimens with a higher quantity of cfDNA, although in some instances targetable genotypes could be detected in specimens with a relatively low number of GE sequenced indicating high DNA shed from the tumor. In this pilot study, sensitivity of plasma NGS was 77%, comparable to most plasma genotyping assays which have reported sensitivity in the range of 70% (24). Sensitivity improves with higher DNA concentration, highlighting how important it will be to understand why the amount of total cfDNA in plasma varies across such a wide range, and whether this variance is due to fluctuations in tumor biology or in methods of extraction. For example, we and others have described previously that plasma genotyping assays are more sensitive in lung cancer patients with extra-thoracic metastases (13, 15, 17, 18). Even with a moderately high sensitivity, the lack of false positives with this assay results in a 100% positive predictive value, meaning that this plasma NGS assay could be used as a screening step before a biopsy sample is taken for genomic analysis: if positive, the results are reliable and can potentially obviate the need for a biopsy, and if negative, a biopsy for further testing may still have value.

Our detection of various resistance mechanisms in patients with acquired resistance to TKIs, including the detection of two different EGFR C797S clones in one case, highlights the potential power of plasma NGS for understanding the heterogeneity of the resistant state. In addition, the C797S alleles identified with plasma NGS were clearly in cis with T790M (Fig. 3), a detail that would be difficult to decipher with a PCR assay and may have treatment implications (22). It is increasingly appreciated that resistant tumors are made up of clones with diverse biologies that may respond differently to therapy (25, 26). We recently showed that three molecular subtypes of acquired resistance to the investigational EGFR inhibitor AZD9291 are apparent by use of serial plasma ddPCR: although all patients started with T790M plus a sensitizing mutation, some lose T790M at resistance whereas some gain C797S (21). However, this analysis required five separate PCR assays: T790M, 19 deletion, L858R, and assays for two C797S variants. Alternatively, one plasma NGS assay can detect these five alterations plus detecting any novel resistance mechanisms that emerge. Our data further suggest that quantification of allelic fraction is similar using plasma NGS and ddPCR, suggesting either could be used to serially monitor plasma genotype concentration.

The findings described in this report are achieved by applying a novel bias-corrected capture technology that builds upon standard sequencing approaches to maximize the efficiency of on-target versus off-target and redundant sequencing reads. By reducing PCR artifacts, this assay can accurately detect mutation present in as few as 0.1% of sequencing reads; in contrast, some tumor NGS platforms are unable to accurately call a mutation unless detected in >10% of sequencing reads. Bias-corrected targeted NGS is an approach that can be applied to any sequencing platform to improve on-target coverage and reduce noise. Here, we apply bias-corrected targeted NGS to desktop sequencing on a MiSeq platform in order to develop a plasma assay with the potential to be rapid and clinic-ready, in contrast to more time intensive assays utilizing HiSeq platforms (18). Yet this NGS approach could also be applied to clinical sequencing panels or discovery efforts to improve sequencing coverage and reduce artifact, particularly when studying clinical specimens from small biopsies with scant DNA.

In conclusion, we have developed and successfully piloted a plasma NGS assay that, using a novel capture and analysis technique, can detect targetable mutations and rearrangements in plasma from advanced NSCLC patients. This is the first plasma NGS assay to demonstrate blinded, multiplexed detection of such a broad range of actionable alterations with no false positive results. By using a widely available desktop sequencing platform and standard vacutainers with the potential for a rapid turnaround time, this assay has the potential for broad uptake and application. Through reducing the barriers between NSCLC patients and genotyping, we hope that plasma NGS will be able to facilitate delivery of targeted therapies and improve outcomes for patients with advanced NSCLC.

A.G. Sacher reports receiving travel reimbursement from AstraZeneca and Roche. C.K. Raymond and L.P. Lim have ownership interest in Resolution Bioscience. P.A. Jänne reports receiving a commercial research grant from AstraZeneca and Astellas; has ownership interest in Gatekeeper Pharmaceuticals; and is a consultant/advisory board member for AstraZeneca, Chugai Pharmaceutical, Merrimack Pharmaceuticals, Pfizer, and Roche. G.R. Oxnard is a consultant/advisory board member for Ariad, AstraZeneca, Clovis, and Sysmex. No potential conflicts of interest were disclosed by the other authors.

Conception and design: C.P. Paweletz, C.K. Raymond, G.R. Oxnard

Development of methodology: A.G. Sacher, C.K. Raymond, A. O'Connell, L.P. Lim, G.R. Oxnard

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.G. Sacher, C.K. Raymond, R.S. Alden, S.L. Mach, Y. Kuang, P.A. Jänne, G.R. Oxnard

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C.P. Paweletz, A.G. Sacher, C.K. Raymond, R.S. Alden, A. O'Connell, Y. Kuang, L. Gandhi, L.P. Lim, G.R. Oxnard

Writing, review, and/or revision of the manuscript: C.P. Paweletz, A.G. Sacher, C.K. Raymond, R.S. Alden, A. O'Connell, S.L. Mach, Y. Kuang, L. Gandhi, P. Kirschmeier, J.M. English, L.P. Lim, P.A. Jänne, G.R. Oxnard

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. O'Connell, J.M. English

Study supervision: P. Kirschmeier, G.R. Oxnard

This work was supported in part by the Department of Defense, Conquer Cancer Foundation of ASCO, Phi Beta Psi Sorority, Stading-Younger Cancer Research Foundation, Expect Miracles Foundation, Harold and Gail Kirstein Lung Cancer Research Fund, and US National Institutes of Health grant R01CA135257 (P.A. Jänne).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Li
T
,
Kung
HJ
,
Mack
PC
,
Gandara
DR
. 
Genotyping and genomic profiling of non-small-cell lung cancer: implications for current and future therapies
.
J Clin Oncol
2013
;
31
:
1039
49
.
2.
Paez
JG
,
Jänne
PA
,
Lee
JC
,
Tracy
S
,
Greulich
H
,
Gabriel
S
, et al
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy
.
Science
2004
;
304
:
1497
500
.
3.
Kwak
EL
,
Bang
YJ
,
Camidge
DR
,
Shaw
AT
,
Solomon
B
,
Maki
RG
, et al
Anaplastic lymphoma kinase inhibition in non-small-cell lung cancer
.
N Engl J Med
2010
;
363
:
1693
703
.
4.
Hauschild
A
,
Grob
JJ
,
Demidov
LV
,
Jouary
T
,
Gutzmer
R
,
Millward
M
, et al
Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial
.
Lancet
2012
;
380
:
358
65
.
5.
Cardarella
S
,
Ortiz
TM
,
Joshi
VA
,
Butaney
M
,
Jackman
DM
,
Kwiatkowski
DJ
, et al
The introduction of systematic genomic testing for patients with non-small-cell lung cancer
.
J Thorac Oncol
2012
;
7
:
1767
74
.
6.
Planchard
D
,
Kim
T
,
Mazières
J
,
Quoix
E
,
Riely
G
,
Barlesi
F
, et al
Dabrafenib in patients with BRAF V600E-mutant advanced non-small cell lung cancer (NSCLC): a multicenter, open-label, phase II trial
(BRF113928).
Ann Oncol
2014
;
25
(
Supplement 5
):
v1
v41
.
7.
Sholl
LM
,
Aisner
DL
,
Varella-Garcia
M
,
Berry
LD
,
Dias-Santagata
D
,
Wistuba
II
, et al
Multi-institutional oncogenic driver mutation analysis in lung adenocarcinoma: the lung cancer mutation consortium experience
.
J Thorac Oncol
2015
.
8.
Wagle
N
,
Berger
MF
,
Davis
MJ
,
Blumenstiel
B
,
Defelice
M
,
Pochanard
P
, et al
High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing
.
Cancer Discov
2012
;
2
:
82
93
.
9.
Hagemann
IS
,
Devarakonda
S
,
Lockwood
CM
,
Spencer
DH
,
Guebert
K
,
Bredemeyer
AJ
, et al
Clinical next-generation sequencing in patients with non-small cell lung cancer
.
Cancer
2015
;
121
:
631
9
.
10.
Luke
JJ
,
Oxnard
GR
,
Paweletz
CP
,
Camidge
DR
,
Heymach
JV
,
Solit
DB
, et al
Realizing the potential of plasma genotyping in an age of genotype-directed therapies
.
J Natl Cancer Inst
2014
;
106(8)
.
11.
Couraud
S
,
Vaca-Paniagua
F
,
Villar
S
,
Oliver
J
,
Schuster
T
,
Blanché
H
, et al
Noninvasive diagnosis of actionable mutations by deep sequencing of circulating free DNA in lung cancer from never-smokers: a proof-of-concept study from BioCAST/IFCT-1002
.
Clin Cancer Res
2014
;
20
:
4613
24
.
12.
Goto
K
,
Ichinose
Y
,
Ohe
Y
,
Yamamoto
N
,
Negoro
S
,
Nishio
K
, et al
Epidermal growth factor receptor mutation status in circulating free DNA in serum: from IPASS, a phase III study of gefitinib or carboplatin/paclitaxel in non-small cell lung cancer
.
J Thorac Oncol
2012
;
7
:
115
21
.
13.
Lee
YJ
,
Yoon
KA
,
Han
JY
,
Kim
HT
,
Yun
T
,
Lee
GK
, et al
Circulating cell-free DNA in plasma of never smokers with advanced lung adenocarcinoma receiving gefitinib or standard chemotherapy as first-line therapy
.
Clin Cancer Res
2011
;
17
:
5179
87
.
14.
Douillard
JY
,
Ostoros
G
,
Cobo
M
,
Ciuleanu
T
,
Cole
R
,
McWalter
G
, et al
Gefitinib treatment in EGFR mutated caucasian NSCLC: circulating-free tumor DNA as a surrogate for determination of EGFR status
.
J Thorac Oncol
2014
;
9
:
1345
53
.
15.
Forshew
T
,
Murtaza
M
,
Parkinson
C
,
Gale
D
,
Tsui
DW
,
Kaper
F
, et al
Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA
.
Sci Transl Med
2012
;
4
:
136ra68
.
16.
Oxnard
GR
,
Paweletz
CP
,
Kuang
Y
,
Mach
SL
,
O'Connell
A
,
Messineo
MM
, et al
Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA
.
Clin Cancer Res
2014
;
20
:
1698
705
.
17.
Sacher
A
,
Oxnard
G
,
Mach
S
,
Messineo
M
,
Jackman
D
,
Jänne
P
, et al
Prediction of lung cancer genotype noninvasively using droplet digital PCR (ddPCR) analysis of cell-free plasma DNA (cfDNA)
. 
American Society of Clinical Oncology Annual Meeting; Chicago, United States
2014
.
18.
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
54
.
19.
Oxnard
G
,
Lydon
C
,
Lindeman
N
,
Shivdasani
P
,
Chin
G
,
Kuo
F
, et al
Implementation of clinical next-generation sequencing (NGS) of non-small cell lung cancer (NSCLC) to identify EGFR amplification as a potentially targetable oncogenic alteration
. 
American Society of Clinical Oncology Annual Meeting; Chicago, USA
2014
.
20.
Jänne
PA
,
Yang
JC
,
Kim
DW
,
Planchard
D
,
Ohe
Y
,
Ramalingam
SS
, et al
AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer
.
N Engl J Med
2015
;
372
:
1689
99
.
21.
Thress
KS
,
Paweletz
CP
,
Felip
E
,
Cho
BC
,
Stetson
D
,
Dougherty
B
, et al
Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M
.
Nat Med
2015
.
22.
Niederst
MJ
,
Hu
H
,
Mulvey
HE
,
Lockerman
EL
,
Garcia
AR
,
Piotrowska
Z
, et al
The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies
.
Clin Cancer Res
2015
;
21
:
3924
33
.
23.
Yu
HA
,
Tian
SK
,
Drilon
AE
,
Borsu
L
,
Riely
GJ
,
Arcila
ME
, et al
Acquired resistance of EGFR-mutant lung cancer to a T790M-Specific EGFR inhibitor: emergence of a third mutation (C797S) in the EGFR tyrosine kinase domain
.
JAMA Oncol
2015
.
24.
Luo
J
,
Shen
L
,
Zheng
D
. 
Diagnostic value of circulating free DNA for the detection of EGFR mutation status in NSCLC: a systematic review and meta-analysis
.
Sci Rep
2014
;
4
:
6269
.
25.
de Bruin
EC
,
McGranahan
N
,
Mitter
R
,
Salm
M
,
Wedge
DC
,
Yates
L
, et al
Spatial and temporal diversity in genomic instability processes defines lung cancer evolution
.
Science
2014
;
346
:
251
6
.
26.
Zhang
J
,
Fujimoto
J
,
Wedge
DC
,
Song
X
,
Seth
S
,
Chow
CW
, et al
Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing
.
Science
2014
;
346
:
256
9
.

Supplementary data