Next Article in Journal
Influencing Factors on the Oncuria™ Urinalysis Assay: An Experimental Model
Previous Article in Journal
Transitioning to Combined EBUS EUS-B FNA for Experienced EBUS Bronchoscopist
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Analytical Validation of a Pan-Cancer Panel for Cell-Free Assay for the Detection of EGFR Mutations

Department of Laboratory Medicine and Genetics, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul 06351, Korea
*
Author to whom correspondence should be addressed.
Diagnostics 2021, 11(6), 1022; https://doi.org/10.3390/diagnostics11061022
Submission received: 27 April 2021 / Revised: 28 May 2021 / Accepted: 31 May 2021 / Published: 2 June 2021
(This article belongs to the Section Pathology and Molecular Diagnostics)

Abstract

:
Liquid biopsies have increasingly shown clinical utility. Although next-generation sequencing has been widely used for the detection of somatic mutations from plasma, performance characteristics vary by platform. Therefore, thorough validation is mandatory for clinical use. This study aimed to evaluate the analytical validity of the Oncomine Pan-Cancer Cell-Free Assay. A massively parallel sequencing for the assay was performed using the Ion S5 XL System with Ion 540 kit. The analytical sensitivity and precision were evaluated using pre-characterized reference materials. The specificity was evaluated using plasma from healthy subjects. A comparison with the Cobas EGFR Mutation Test v2 was performed using reference materials and plasma from lung cancer patients. For SNVs and short indels, the analytical sensitivities at variant allele frequencies (VAFs) of 0.1%, 0.5%, and 1% were 50%, 93.4%, and 100% with 20 ng of input, respectively. The overall precision of the true positive variants was 98% at a VAF of 1% with 20 ng input. The assay showed a similar sensitivity to that of the Cobas EGFR Mutation Test v2 at a VAF of 0.5% with 20 ng of input and 100% concordance on clinical samples. The Pan-Cancer Cell-Free Assay can be applied to detect EGFR mutations in advanced lung cancer patients, although follow-up studies will be needed to evaluate the analytical validity for other types of genes and aberrations using clinical samples.

1. Introduction

Cell-free DNA (cfDNA) is a short-fragment nucleotide in the bloodstream. This DNA is produced from cell apoptosis or necrosis but is also actively released from viable cells [1,2,3]. Circulating tumor DNA (ctDNA), as a component of cfDNA of cancer patients, represents the mutation profile of tumor tissues, and the concordance between tissue and plasma ctDNA ranges from ~70% to ~90% for the detection of EGFR variants in nonsmall cell lung cancers [4,5]. Research has been conducted to demonstrate the usefulness of ctDNA in various cancer types for the monitoring of tumor recurrence, the identification of mutations for targeted therapy, and the early detection of tumors [6]. Recently, the guidelines of the American Society of Clinical Oncology for molecular testing of lung cancer patients have been updated. The guideline states that the cfDNA assay can be used to identify EGFR mutations in some clinical situations with limited and/or insufficient tissues [7].
For the detection of ctDNA, a highly sensitive methodology is essential due to the small amount of cfDNA. Furthermore, ctDNA is present in the background of cfDNA derived from hematopoietic cells [8]. Among the various methods available for analysis, real-time PCR, digital droplet PCR, and next-generation sequencing (NGS) are commonly used in clinical laboratories. As a real-time PCR technique, the Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Inc., Branchburg, NJ, USA) is used for detecting several hotspot mutations of the EGFR gene. The test, which uses plasma, as well as formalin-fixed paraffin-embedded tissue specimens, was approved as a companion diagnostic test by the United States Food and Drug Administration [9]. Digital droplet PCR can detect 0.01~0.1% of the mutant burden at hotspot positions [6,10]. An NGS-based analysis, in contrast to a targeted approach, can include up to several hundred genes and detect sequence variations, copy number variants (CNVs), and/or fusion transcripts in a single test. Such an analysis shows a 0.1% limit of detection when using a deeper depth of coverage, special processes such as molecular barcodes (also known as unique molecular identifiers) for the differentiation of true mutations from sequencing artifacts, and computational postprocessing [8,11,12].
This study aimed to evaluate the analytical performance of the Oncomine Pan-Cancer Cell-Free assay (Thermo Fisher Scientific, Waltham, MA, USA) using cell-free total nucleic acid (cfTNA), which is designed to identify single-nucleotide variants (SNVs), small indels, copy number variations (CNVs), and RNA fusions from 52 genes. In addition, we compared the analytical sensitivity of the Oncomine Pan-Cancer Cell-Free assay (Thermo Fisher Scientific, Waltham, MA, USA) to that of the Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Pleasanton, CA, USA).

2. Materials and Methods

2.1. Preparation of Pre-Characterized Reference Materials

For validation of the analytical performance, we used two commercially available control materials. For validation of SNVs and indels, the Multiplex I cfDNA Reference Standard Set (HD778 for a variant allele frequency (VAF) of 1%, HD779 for a VAF of 0.1%, and HD776 for wild type; Horizon Discovery, Waterbeach, UK) was used, which contains 4 EGFR mutations (p.L858R, p.E746_A750del (∆E746-A750), p.T790M, p.A767_V769dup (V769-D770insASV)), 1 KRAS mutation (p.G12D), 2 NRAS mutations (p.Q61K, p.A59T), and 1 PIK3CA mutation (p.E545K). In addition, a library with a VAF of 0.5% was prepared by mixing the mutant control (HD778) and wild-type control (HD776) in equal quantities. For the nucleic acid input, the control material at an initial concentration of 20 ng/μL was further diluted, and different amounts of the nucleic acid input (5, 10, and 20 ng) were used for library preparation. The HD786 structural multiplex cfDNA Reference Standard Set was used for validation of the CNV and fusion. This HD786 reference standard set contains 5% fusion of SLC34A2/ROS1 and CCDC6/RET, 4.5 copies (2.25-fold change) of MET, and 9.5 copies (4.75-fold change) of MYCN. In addition, HD786 was mixed with wild-type control at a ratio of 1:4, yielding a VAF of 1% for the fusion transcript, 2.5 copies (1.25-fold change) of MET, and 3.5 copies (1.75-fold change) of MYCN.

2.2. Preparation of Clinical Samples

We used plasma samples from eight lung cancer patients with EGFR-positive variants detected using the Cobas EGFR Mutation Test v2 (Roche Molecular Systems, Pleasanton, CA, USA). Plasma was separated from the Cell-Free DNA Collection tube (Roche Molecular Systems) by double centrifugation (1600× g for 15 min followed by 16,000× g for 10 min); after completion of the test, the residual volume was archived in a deep freezer (−70 ℃). As the residual volume of plasma of each sample was not sufficient, two EGFR-positive clinical samples were mixed to 4 mL of plasma for DNA extraction. Extraction of cfTNA was performed using the QIAamp® DSP Circulating NA kit (Qiagen, Hilden, Germany). The concentration and size distribution of the nucleic acid were assessed using Qubit 3.0 Fluorometer (Thermo Fisher Scientific, Waltham, MA, USA) and Tapestation (Agilent, Santa Clara, CA, USA), respectively. From the 40 μL of eluted cfTNA, 10 μL (the maximum volume of input recommended by the manufacturer) was used for library preparation if the total amount of cfTNA did not exceed 50 ng. This study was approved by the Institutional Review Board of Samsung Medical Center, Seoul, Korea (2019-01-140).

2.3. Library Preparation, Sequencing, and Variant Calling

The Oncomine Pan-Cancer Cell-Free Assay was used for library preparation, in which DNA fragments were each tagged with a unique molecular tag. Four libraries were loaded on an Ion 540 kit using Ion Chef (Thermo Fisher Scientific, Waltham, MA, USA). One or two Ion 540 chips were sequenced simultaneously using the Ion S5 XL System (Thermo Fisher Scientific). In the initial validation, 16 libraries were sequenced during two consecutive sequencing runs; two 540 chips were analyzed at each sequencing run. In the subsequent six sequencing runs, one control material was included in each run and the corresponding data of the control material were used for validation of reproducibility. Sequence alignment to the hg19 human reference genome and variant calling were performed using Torrent Suite version 5.10.1 (Thermo Fisher Scientific). For variant annotation, Ion Reporter Software 5.10 was used (Thermo Fisher Scientific). Cutoffs for various parameters were used as the default setting. Briefly, molecular coverage, the representation of the number of DNA molecules, of at least 3 with a minimum detection cutoff frequency of 0.065% must be satisfied for variant calling for SNV/indel. For a CNV call, the following criteria must be met: median of the absolute values of all pairwise differences (MAPD) < 0.4, p-value < 10−5, and CNV ratio for a copy number gain >1.15-fold change. For calling a fusion transcript or MET exon 14 skipping, more than 2 molecular counts of the target must have been reported. In addition, at least one of the two control genes (TBP and HMBS) and wild-type controls of MET must have a molecular count above 2. A variant was called when it was present in the “analysis visualization” on Ion Reporter where the TP53 loss of function variant with a molecular frequency less than 1% was further filtered out if the variant had not been defined as a hotspot mutation.

3. Results

3.1. Summary of the Run

An average of 22 million reads was mapped to the reference genome per library, and 96% of the mapped reads were on a target relative to the designed bed file. The mean depth of coverage ranged from 64,000× to 91,000×. The uniformity of each library, which is the percentage of amplicons (bases) covered greater than 20% of the mean amplicon (base) coverage, was 99%.

3.2. Analytical Sensitivity

The analytical sensitivity of SNVs and indels was evaluated with a varying level of VAF and input using the Multiplex I cfDNA Reference Standard (Table 1).
Molecular coverage depended on the amount of input. A molecular coverage above 2000× was obtained with 20 ng of input, whereas molecular coverages of 1100× and 700× were obtained with 10 and 5 ng of input, respectively. With 20 ng of input, the analytical sensitivities at VAFs of 0.1%, 0.5%, and 1% were 50%, 93.4% and 100%, respectively. At a VAF of 0.1%, the EGFR p.E746_A750del variant was detected in a repeated experiment, although other mutations were inconsistently detected below the limit of detection (LOD) level. At a VAF of 0.5%, the EGFR L858R mutation was detected only once in duplicate experiments, while other mutations were detected in both. Although two molecular reads of 1752 molecular depth (0.11%) had the variant, the variant was treated as “NOCALL” with the reason of “&VARCOV<3&VARCOV-TGSM<3”. This means that it did not satisfy the detection threshold of a molecular coverage ≥3.
For evaluation of the analytical sensitivity for fusion, we tested libraries having two levels of VAF or copies at 20 ng. As a result, ROS1 and RET fusion transcripts were detected at VAF levels of both 5% and 1%. For CNV, copy number changes of MET with 4.5 (2.25-fold change) and 2.5 copies (1.25-fold change) were detected, and their observed fold changes were 1.8 and 1.44, respectively (Table 2).

3.3. Analytical Specificity

Analytical specificity was estimated using pooled plasma obtained from healthy subjects aged 30–40 years.
The plasma was tested in seven independent experiments, and 1.0–10 ng of nucleic acid was used. The median molecular depth was 900× (range 583~1600×). As a result, no variant was called except two false positive variants, KIT p.V530I and MAP2K1 p.F129L, from one experiment. The false positive variants were hotspot mutations, and their VAFs were 0.28% and 0.24%, respectively.

3.4. Precision

Precision was estimated from the replicated data of the reference material within a run and/or between runs. For precision at a VAF of 1% with 20 ng of input, two replicates within the run and six replicates with independent batches were performed. The overall precision of the true positive variants was 98% (Table 3).
For individual variants, all except EGFR p.L858R showed a precision of 100%; EGFR p.L858R was detected in 7 of 8 replicates, yielding a precision of 87.5%. Although three molecular reads of 3525 molecular depth (0.09%) had the variant, it was treated as “NOCALL” with the reason of “.&VARCOV-TGSM<3.” This means that it did not satisfy the detection threshold of molecular coverage ≥3 after adjustment using TGSM (TaG Similarity), which is the number of families that supports alleles being falsely generated due to a PCR error or family cloning from sample preparation. Coefficients of variation (CVs) of the observed VAF varied from 16% to 62% according to the variant.
In addition to the verified mutations, the following five variants were called consistently at VAFs of 20~30% across the eight replicated experiments: BRAF p.V600E, CTNNB1 p.S33Y, EGFR p.G719S, MAP2K1 p.Q56P, and PIK3CA p.H1047R. According to the manufacturer’s claim, those variants were confirmed in parent cell lines by whole exome sequencing (https://horizondiscovery.com/en/reference-standards/products/~/link.aspx?_id=D2686851191C40738A817A951F90BA82&_z=z, accessed on 15 January 2021).
In addition to the above variants, FGFR3 amplification was called in 7 of 8 replicates. Each MET amplification, APC p.S1465fs, FBXW7 p.R465C, and HRAS p.G12V were called once.
At a VAF of 0.5% with 20 ng, only the repeatability (intra-run) of true positive variants was estimated from the reference material; the overall repeatability was 94% as the EGFR p.L858R variant was detected only once among the two replicates (Table 1).

3.5. Comparison Between Cobas-EGFR and Pan-Cancer Cell-Free Assay

When using the reference material at VAFs of 0.5 and 1.0% with 20 ng of input, the Pan-Cancer Cell-Free Assay and Cobas-EGFR showed 100% detection (Table 4).
At a VAF of 0.1%, the Pan-Cancer Cell-Free Assay detected two of four variants, whereas Cobas-EGFR detected one of four.
In clinical samples, the amount of input ranged from 0.7 to 53 ng with a molecular depth of 1053~3806×. Across the four libraries from eight clinical samples, all EGFR-mutations detected by Cobas-EGFR were also detected by the Pan-Cancer Cell-Free Assay, showing 100% concordance (Table 5). The lowest VAF was 0.1% for the p.E746_A750del mutation.

4. Discussion

In this paper, we evaluated the analytical validity of the Pan-Cancer Cell-Free Assay on an S5 platform for detecting cancer-associated mutations. As analytical performance depends on the amount of input nucleic acid, as well as VAF, we planned the evaluation protocol with variable VAF, and the amount of input based on reference materials.
It has been suggested that a 0.1% of LOD is required for tumor liquid biopsy, whereas an LOD less than 0.01% is required for early cancer detection [13]. In the present study, the analytical sensitivities were estimated to be 50%, 94%, and 100% at VAFs of 0.1%, 0.5%, and 1% with 20 ng of input, respectively. If limited numbers of mutant molecules are present, the accurate and reliable detection of mutation with a VAF of 0.1% is challenging due to the stochastic sampling issue [14]. This issue is a problem for the Pan-Cancer Cell-Free Assay, as well as others. Previous studies that have included patients with various cancer types showed a mean VAF of stage III/IV greater than 1% [15] and a VAF for second quartiles of 1.3% in metastatic cancers [16], indicating that this assay is suitable for advanced cancer patients.
Detection sensitivity differed by mutation. Though massively parallel sequencing (MPS) is less sensitive for the detection of indels than for SNVs [17,18], the detection of the EGFR p.E746_A750del mutation in this study was better than that of other EGFR mutations and was consistently detected in duplicated experiments at a VAF of 0.1%. The detection of the EGFR p.L858R mutation was less sensitive as it was detected only once in two duplicated experiments at a VAF of 0.5%. In addition, the reproducibility of detection of the EGFR p.L858R mutation at a VAF of 1% was relatively low as one of six additional experiments failed to detect the mutation despite adequate coverage of that position (3525 molecular reads). EGFR exon 19 deletion and L858R are the first and second most common mutations found in Asian lung cancer patients, respectively [19]. This demonstrates the importance of the awareness of issues related to the sensitivity and/or precision for each clinically important mutation.
In the comparison test using the reference material, the Pan-Cancer Cell-Free Assay and Cobas EGFR assay showed comparable results. Both assays showed 100% detection of the EGFR mutations at VAFs of 0.5% and 1% with 20 ng of input. Both assays showed decreased sensitivity at a VAF of 0.1% with the same input. However, the Pan-Cancer Cell-Free assay appeared to be slightly more sensitive than the Cobas EGFR assay, with 50% and 25% detection of the variants, respectively. In a previous study, the Cobas EGFR assay showed 50% detection at a VAF of 0.1%, while the Oncomine Lung cfDNA assay detected all variants [20]. The difference in the detection rate between the previous and current studies might be due to the different amounts of input nucleic acid (66 vs. 20 ng).
The performance of the test can be different even with the same platform or the same assay. Thus, reporting from various studies by independent groups may help to determine the performance of the assay. The previous study using the Oncomine Lung Cell-Free DNA Assay, the same platform with the different assay, demonstrated a sensitivity of 81% with <20 ng of cfDNA at a VAF of 0.1%, whereas our data showed a sensitivity of 50% under the same condition [21]. In the report by the manufacturer, the same assay in the present study showed a sensitivity of 80% at a VAF of 0.1% for SNV [22]. In the present study, most of the analytical performance tests were conducted using reference materials and clinical samples containing only EGFR mutations. Other clinically important mutations including fusion or CNV were not validated by clinical samples. As the performance with clinical samples may not be the same as that with reference materials, further studies would be required for analytical validity of this panel. Although analytical validity has been rarely reported for this assay, clinical validity has been reported in endometrial cancer, brain tumor, and colorectal cancer [23,24,25]. The clinical validity of various cancer types remains to be determined.

5. Conclusions

The Pan-Cancer Cell-Free Assay can be applied to detect EGFR mutations in advanced lung cancer patients, although follow-up studies will be needed to evaluate the analytical validity for other types of genes and aberrations using clinical samples. As sensitivity varies according to variant, the clinical laboratory should evaluate and understand the performance characteristics of the assay prior to its adoption as a clinical test.

Author Contributions

Study organization and data collection, J.-H.J.; data interpretation and writing of the first draft of the manuscript, M.-K.S.; bioinformatics pipeline review, J.-H.P.; study supervision, J.-W.K. All authors discussed the results and commented on the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

This study was approved by the Institutional Review Board of Samsung Medical Center, Seoul, Korea (2019-01-140).

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Stroun, M.; Lyautey, J.; Lederrey, C.; Olson-Sand, A.; Anker, P. About the possible origin and mechanism of circulating DNA apoptosis and active DNA release. Clin. Chim. Acta 2001, 313, 139–142. [Google Scholar] [CrossRef]
  2. Mouliere, F.; Chandrananda, D.; Piskorz, A.M.; Moore, E.K.; Morris, J.; Ahlborn, L.B.; Mair, R.; Goranova, T.; Marass, F.; Heider, K.; et al. Enhanced detection of circulating tumor DNA by fragment size analysis. Sci. Transl. Med. 2018, 10, eaat4921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Schwarzenbach, H.; Hoon, D.S.B.; Pantel, K. Cell-free nucleic acids as biomarkers in cancer patients. Nat. Rev. Cancer 2011, 11, 426–437. [Google Scholar] [CrossRef]
  4. Thress, K.S.; Brant, R.; Carr, T.H.; Dearden, S.; Jenkins, S.; Brown, H.; Hammett, T.; Cantarini, M.; Barrett, J.C. EGFR mutation detection in ctDNA from NSCLC patient plasma: A cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer 2015, 90, 509–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Arriola, E.; Paredes-Lario, A.; García-Gomez, R.; Diz-Tain, P.; Constenla, M.; García-Girón, C.; Márquez, G.; Reck, M.; López-Vivanco, G. Comparison of plasma ctDNA and tissue/cytology-based techniques for the detection of EGFR mutation status in advanced NSCLC: Spanish data subset from ASSESS. Clin. Transl. Oncol. 2018, 20, 1261–1267. [Google Scholar] [CrossRef] [Green Version]
  6. Diaz, L.A.J.; Bardelli, A. Liquid biopsies: Genotyping circulating tumor DNA. J. Clin. Oncol. 2014, 32, 579–586. [Google Scholar] [CrossRef]
  7. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J. Clin. Oncol. 2018, 36, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
  8. Barbany, G.; Arthur, C.; Lieden, A.; Nordenskjold, M.; Rosenquist, R.; Tesi, B.; Wallander, K.; Tham, E. Cell-free tumour DNA testing for early detection of cancer—A potential future tool. J. Intern. Med. 2019, 286, 118–136. [Google Scholar] [CrossRef] [Green Version]
  9. U.S. Food and Drug Administration: Medical Devices: Cobas EGFR Mutation Test v2-P150047. Available online: https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=P150047 (accessed on 6 June 2016).
  10. Li, H.; Jing, C.; Wu, J.; Ni, J.; Sha, H.; Xu, X.; Du, Y.; Lou, R.; Dong, S.; Feng, J. Circulating tumor DNA detection: A potential tool for colorectal cancer management (Review). Oncol. Lett. 2018, 17, 1409–1416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Salk, J.J.; Schmitt, M.W.; Loeb, L.A. Enhancing the accuracy of next-generation sequencing for detecting rare and subclonal mutations. Nat. Rev. Genet. 2018, 19, 269–285. [Google Scholar] [CrossRef] [PubMed]
  12. Kinde, I.; Wu, J.; Papadopoulos, N.; Kinzler, K.W.; Vogelstein, B. Detection and quantification of rare mutations with massively parallel sequencing. Proc. Natl. Acad. Sci. USA 2011, 108, 9530–9535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Aravanis, A.M.; Lee, M.; Klausner, R.D. Next-Generation Sequencing of Circulating Tumor DNA for Early Cancer Detection. Cell 2017, 168, 571–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Heitzer, E.; Haque, I.S.; Roberts, C.E.S.; Speicher, M.R. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat. Rev. Genet. 2019, 20, 71–88. [Google Scholar] [CrossRef]
  15. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Pairawan, S.; Hess, K.R.; Janku, F.; Sanchez, N.S.; Shaw, K.R.M.; Eng, C.; Damodaran, S.; Javle, M.; Kaseb, A.O.; Hong, D.S.; et al. Cell-free Circulating Tumor DNA Variant Allele Frequency Associates with Survival in Metastatic Cancer. Clin. Cancer Res. 2019, 26, 1924–1931. [Google Scholar] [CrossRef] [PubMed]
  17. Lih, C.J.; Harrington, R.D.; Sims, D.J.; Harper, K.N.; Bouk, C.H.; Datta, V.; Yau, J.; Singh, R.R.; Routbort, M.J.; Luthra, R.; et al. Analytical validation of the next-generation sequencing assay for a nationwide signal-finding clinical trial: Molecular analysis for Therapy Choice Clinical Trial. J. Mol. Diagn 2017, 19, 313–327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Gargis, A.S.; Kalman, L.; Berry, M.W.; Bick, D.P.; Dimmock, D.P.; Hambuch, T.; Lu, F.; Lyon, E.; Voelkerding, K.V.; Zehnbauer, B.A.; et al. Assuring the quality of next-generation sequencing in clinical laboratory practice. Nat. Biotechnol. 2012, 30, 1033–1036. [Google Scholar] [CrossRef]
  19. Shi, Y.; Au, J.S.-K.; Thongprasert, S.; Srinivasan, S.; Tsai, C.-M.; Khoa, M.T.; Heeroma, K.; Itoh, Y.; Cornelio, G.; Yang, P.-C. A Prospective, Molecular Epidemiology Study of EGFR Mutations in Asian Patients with Advanced Non–Small-Cell Lung Cancer of Adenocarcinoma Histology (PIONEER). J. Thorac. Oncol. 2014, 9, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kim, Y.; Shin, S.; Lee, K.-A. A Comparative Study for Detection of EGFR Mutations in Plasma Cell-Free DNA in Korean Clinical Diagnostic Laboratories. BioMed Res. Int. 2018, 2018, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. De Luca, G.; Lastraioli, S.; Conte, R.; Mora, M.; Genova, C.; Rossi, G.; Tagliamento, M.; Coco, S.; Bello, M.G.D.; Zupo, S.; et al. Performance of the OncomineTM Lung cfDNA Assay for Liquid Biopsy by NGS of NSCLC Patients in Routine Laboratory Practice. Appl. Sci. 2020, 10, 2895. [Google Scholar] [CrossRef]
  22. Williamson, J.B.; Solano, L.; Yuki, A.; Burkhart, V.D.; Chitwood, J.; Cao, R.; Schageman, J.; Saunders, H.; Cheng, A.; Ha, T.; et al. Analytical validation of the Oncomine Pan-Cancer Cell-Free Assay in a CLIA- and CAP-regulated laboratory for detection of solid tumor-derived variants in blood plasma. J. Clin. Oncol. 2019, 37, e14614. [Google Scholar] [CrossRef]
  23. Moss, E.L.; Gorsia, D.N.; Collins, A.; Sandhu, P.; Foreman, N.; Gore, A.; Wood, J.; Kent, C.; Silcock, L.; Guttery, D.S. Utility of Circulating Tumor DNA for Detection and Monitoring of Endometrial Cancer Recurrence and Progression. Cancers 2020, 12, 2231. [Google Scholar] [CrossRef] [PubMed]
  24. Shah, M.; Takayasu, T.; Moghadamtousi, S.Z.; Arevalo, O.; Chen, M.; Lan, C.; Duose, D.; Hu, P.; Zhu, J.-J.; Roy-Chowdhuri, S.; et al. Evaluation of the Oncomine Pan-Cancer Cell-Free Assay for Analyzing Circulating Tumor DNA in the Cerebrospinal Fluid in Patients with Central Nervous System Malignancies. J. Mol. Diagn. 2021, 23, 171–180. [Google Scholar] [CrossRef] [PubMed]
  25. Chan, R.-H.; Lin, P.-C.; Chen, S.-H.; Lin, S.-C.; Chen, P.-C.; Lin, B.-W.; Shen, M.-R.; Yeh, Y.-M. Clinical Utility of a Cell-Free DNA Assay in Patients With Colorectal Cancer. Front. Oncol. 2021, 11, 589673. [Google Scholar] [CrossRef]
Table 1. Analytical sensitivity using the reference material.
Table 1. Analytical sensitivity using the reference material.
GeneVariantVariant Allele Frequency (%) According to Input (ng)
0.1%0.5%1%
5 ng10 ng20 ng20 ng20 ng
EGFRp.L858R 0.70.940.62
p.E746_A750del 0.260.250.550.591.821.44
p.T790M 0.2 0.440.521.081.31
p.A767_V769dup 0.16 0.080.320.321.070.67
KRASp.G12D 0.530.2 0.370.681.221.25
NRASp.Q61K 0.181.270.861.991.36
p.A59T 0.720.551.352.41
PIK3CAp.E545K 0.160.320.410.971.341.44
Molecular coverage688×1139×2091×2115×2017×
Analytical sensitivity0%
(0/8)
25%
(2/8)
50%
(8/16)
94%
(15/16)
100%
(16/16)
Table 2. Analytical sensitivity for fusion and CNV using the reference material.
Table 2. Analytical sensitivity for fusion and CNV using the reference material.
Variant (Fusion)VAF 1%VAF 5%
ResultMol. Cov. for FusionRead
Cov. for Fusion
ResultMol. Cov. for FusionRead
Cov. for Fusion
SLC34A2–ROS1 Detected311473Detected1274850
CCDC6–RET Detected12510Detected321357
Variant (CNV)2.5 Copies (CN Ratio of 1.25)4.5 Copies (CN Ratio of 2.25)
ResultObserved
CN Ratio
MAPDResultObserved CN RatioMAPD
MET amplificationDetected1.440.233Detected1.80.24
Abbreviations: CN, copy number; CNV, copy number variation; MAPD, median of the absolute values of all pairwise differences; VAF, variant allele frequency.
Table 3. Precision at a VAF of 1% with 20 ng of reference material when testing duplicates within the same batch and six replicates from independent batches.
Table 3. Precision at a VAF of 1% with 20 ng of reference material when testing duplicates within the same batch and six replicates from independent batches.
GeneVariantsExpected VAF (%)Mean and SD of
Observed VAF (%)
CV (%)Detection Rate
EGFRp.L858R1.00.99 ± 0.232387.5% (7/8)
p.E746_A750del 1.38 ± 0.3727100% (8/8)
p.T790M1.00 ± 0.3737100% (8/8)
p.A767_V769dup 0.94 ± 0.2931100% (8/8)
KRASp.G12D1.31.29 ± 0.3225100% (8/8)
NRASp.Q61K1.43 ± 0.2316100% (8/8)
p.A59T1.66 ± 0.4427100% (8/8)
PIK3CAp.E545K0.95 ± 0.5861100% (8/8)
Abbreviations: CV, coefficient of variation; SD, standard deviation; VAF, variant allele frequency.
Table 4. Comparison between Oncomine Pan-Cancer Cell-Free Assay and Cobas-EGFR assay for detection of EGFR mutations using the reference material.
Table 4. Comparison between Oncomine Pan-Cancer Cell-Free Assay and Cobas-EGFR assay for detection of EGFR mutations using the reference material.
Variant0.1%0.5%1%
Pan-CancerCobasPan-CancerCobasPan-CancerCobas
VAF (%)SQIVAF (%)SQIVAF (%)SQI
p.E746_A750del 0.2611.360.5912.921.8214.11
p.A767_V769dup NDND0.322.721.073.1
p.T790M0.2ND0.528.751.089.42
p.L858RNDND0.78.20.948.5
Abbreviations: SQI, semi-quantitative index; VAF, variant allele frequency.
Table 5. Comparison between Pan-Cancer Cell-Free Assay and Cobas-EGFR assay from samples with EGFR mutation.
Table 5. Comparison between Pan-Cancer Cell-Free Assay and Cobas-EGFR assay from samples with EGFR mutation.
Library
No. *
VariantCobas-EGFR AssayPan-Cancer Assay
SQI ValueMolecular DepthMolecular CountObserved VAF (%)
1p.E746_A750del 12.04314430.1
p.E746_A750del 11.18
2p.L858R6.6621102109.95
p.T790M11.61886839.37
3p.L858R7.011540322.08
p.L858R7.87
4p.L858R5.01177050.28
p.L858R5.99
* Each library was prepared by mixing plasma from two clinical samples. Abbreviations: SQI, semi-quantitative index; VAF, variant allele frequency.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

So, M.-K.; Park, J.-H.; Kim, J.-W.; Jang, J.-H. Analytical Validation of a Pan-Cancer Panel for Cell-Free Assay for the Detection of EGFR Mutations. Diagnostics 2021, 11, 1022. https://doi.org/10.3390/diagnostics11061022

AMA Style

So M-K, Park J-H, Kim J-W, Jang J-H. Analytical Validation of a Pan-Cancer Panel for Cell-Free Assay for the Detection of EGFR Mutations. Diagnostics. 2021; 11(6):1022. https://doi.org/10.3390/diagnostics11061022

Chicago/Turabian Style

So, Min-Kyung, Jong-Ho Park, Jong-Won Kim, and Ja-Hyun Jang. 2021. "Analytical Validation of a Pan-Cancer Panel for Cell-Free Assay for the Detection of EGFR Mutations" Diagnostics 11, no. 6: 1022. https://doi.org/10.3390/diagnostics11061022

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop