Cancer Res Treat.  2023 Apr;55(2):429-441. 10.4143/crt.2022.891.

Validation and Clinical Application of ONCOaccuPanel for Targeted Next-Generation Sequencing of Solid Tumors

Affiliations
  • 1Department of Pathology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
  • 2Bioinformatics Team, R&D Center, NGeneBio Inc., Seoul, Korea
  • 3Diagnostics Development A Team, R&D Center, NGeneBio Inc., Seoul, Korea

Abstract

Purpose
Targeted next-generation sequencing (NGS) is widely used for simultaneously detecting clinically informative genetic alterations in a single assay. Its application in clinical settings requires the validation of NGS gene panels. In this study, we aimed to validate a targeted hybridization capture-based DNA panel (ONCOaccuPanel) using the Illumina MiSeq sequencing platform. The panel allows the simultaneous detection of single-nucleotide variants (SNVs), insertions, deletions, and copy number changes of 323 genes and fusions of 17 genes in solid tumors.
Materials and Methods
We used 16 formalin-fixed paraffin-embedded (FFPE) tumor samples with previously known genetic mutations and one reference material (HD827) for validation. Moreover, we sequenced an additional 117 FFPE tumor samples to demonstrate the clinical utility of this panel.
Results
Validation revealed a 100% positive percentage agreement and positive predictive value for the detection of SNVs, insertions, deletions, copy number changes, fusion genes, and microsatellite instability–high types. We observed high levels of reproducibility and repeatability (R2 correlation coefficients=0.96-0.98). In the limit of detection assessment, we identified all clinically relevant genes with allele frequencies > 3%. Furthermore, the clinical application of ONCOaccuPanel using 117 FFPE samples demonstrated robust detection of oncogenic alterations. Oncogenic alterations and targetable genetic alterations were detected in 98.2% and 27.4% cases, respectively.
Conclusion
ONCOaccuPanel demonstrated high analytical sensitivity, reproducibility, and repeatability and is feasible for the detection of clinically relevant mutations in clinical settings.

Keyword

High-throughput nucleotide sequencing; DNA mutational analysis; Genetic testing; Validation; Neoplasms

Figure

  • Fig. 1 Bioinformatics pipeline analysis process of ONCOaccuPanel: overall process (A) and SNV and Indel calling process (B). CNV, copy number variation; Indel, insertion and deletion; MAF, Mutation Annotation Format; MMR, mismatch repair; MSI, microsatellite instability; POLE, polymerase ɛ; SNV, single-nucleotide variant; SV, structural variation; VCF, Variant Call Format.

  • Fig. 2 Repeatability test of ONCOaccuPanel. Percentages in the vertical axis and horizontal axis denote the variant of allele frequency (VAF). Indel, insertion and deletion; SNP, single nucleotide polymorphism.

  • Fig. 3 Reproducibility test of ONCOaccuPanel. Percentages in the vertical axis and horizontal axis denote the variant of allele frequency (VAF). Indel, insertion and deletion; SNP, single nucleotide polymorphism.

  • Fig. 4 Estimating the limit of detection of OncoAccupanel; first (A) and second (B) test. Percentages in the vertical axis denote the variant of allele frequency (VAF). Exp, expected VAF; Obs, observed VAF.

  • Fig. 5 Oncogenic mutations and fusions detected in 117 clinical samples.


Cited by  1 articles

BRCA-mutated gastric adenocarcinomas are associated with chromosomal instability and responsiveness to platinum-based chemotherapy
Ji Hyun Oh, Chang Ohk Sung, Hyung-Don Kim, Sung-Min Chun, Jihun Kim
J Pathol Transl Med. 2023;57(6):323-331.    doi: 10.4132/jptm.2023.10.22.


Reference

References

1. Chakravarty D, Solit DB. Clinical cancer genomic profiling. Nat Rev Genet. 2021; 22:483–501.
Article
2. Yatabe Y, Sunami K, Goto K, Nishio K, Aragane N, Ikeda S, et al. Multiplex gene-panel testing for lung cancer patients. Pathol Int. 2020; 70:921–31.
Article
3. Pennell NA, Mutebi A, Zhou ZY, Ricculli ML, Tang W, Wang H, et al. Economic impact of next-generation sequencing versus single-gene testing to detect genomic alterations in metastatic non-small-cell lung cancer using a decision analytic model. JCO Precis Oncol. 2019; 3:1–9.
Article
4. Heist RS, Shim HS, Gingipally S, Mino-Kenudson M, Le L, Gainor JF, et al. MET exon 14 skipping in non-small cell lung cancer. Oncologist. 2016; 21:481–6.
5. Kohno T, Nakaoku T, Tsuta K, Tsuchihara K, Matsumoto S, Yoh K, et al. Beyond ALK-RET, ROS1 and other oncogene fusions in lung cancer. Transl Lung Cancer Res. 2015; 4:156–64.
6. Vaishnavi A, Capelletti M, Le AT, Kako S, Butaney M, Ercan D, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med. 2013; 19:1469–72.
Article
7. Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat Rev Cancer. 2019; 19:495–509.
Article
8. Polyak K. Heterogeneity in breast cancer. J Clin Invest. 2011; 121:3786–8.
Article
9. Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of next-generation sequencing technologies. Nat Rev Genet. 2016; 17:333–51.
Article
10. Jennings LJ, Arcila ME, Corless C, Kamel-Reid S, Lubin IM, Pfeifer J, et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn. 2017; 19:341–65.
11. Aziz N, Zhao Q, Bry L, Driscoll DK, Funke B, Gibson JS, et al. College of American Pathologists’ laboratory standards for next-generation sequencing clinical tests. Arch Pathol Lab Med. 2015; 139:481–93.
Article
12. Rehm HL, Bale SJ, Bayrak-Toydemir P, Berg JS, Brown KK, Deignan JL, et al. ACMG clinical laboratory standards for next-generation sequencing. Genet Med. 2013; 15:733–47.
Article
13. Gargis AS, Kalman L, Berry MW, Bick DP, Dimmock DP, Hambuch T, et al. Assuring the quality of next-generation sequencing in clinical laboratory practice. Nat Biotechnol. 2012; 30:1033–6.
Article
14. Kim J, Park WY, Kim NK, Jang SJ, Chun SM, Sung CO, et al. Good laboratory standards for clinical next-generation sequencing cancer panel tests. J Pathol Transl Med. 2017; 51:191–204.
Article
15. Roy S, Coldren C, Karunamurthy A, Kip NS, Klee EW, Lincoln SE, et al. Standards and guidelines for validating next-generation sequencing bioinformatics pipelines: a joint recommendation of the Association for Molecular Pathology and the College of American Pathologists. J Mol Diagn. 2018; 20:4–27.
Article
16. Kim JE, Chun SM, Hong YS, Kim KP, Kim SY, Kim J, et al. Mutation burden and I index for detection of microsatellite instability in colorectal cancer by targeted next-generation sequencing. J Mol Diagn. 2019; 21:241–50.
Article
17. Umar A, Boland CR, Terdiman JP, Syngal S, de la Chapelle A, Ruschoff J, et al. Revised Bethesda Guidelines for hereditary nonpolyposis colorectal cancer (Lynch syndrome) and microsatellite instability. J Natl Cancer Inst. 2004; 96:261–8.
Article
18. Luchini C, Bibeau F, Ligtenberg MJ, Singh N, Nottegar A, Bosse T, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol. 2019; 30:1232–43.
Article
19. Li MM, Datto M, Duncavage EJ, Kulkarni S, Lindeman NI, Roy S, et al. Standards and guidelines for the interpretation and reporting of sequence variants in cancer: a joint consensus recommendation of the Association for Molecular Pathology, American Society of Clinical Oncology, and College of American Pathologists. J Mol Diagn. 2017; 19:4–23.
Article
20. Nakamura Y, Kawazoe A, Lordick F, Janjigian YY, Shitara K. Biomarker-targeted therapies for advanced-stage gastric and gastro-oesophageal junction cancers: an emerging paradigm. Nat Rev Clin Oncol. 2021; 18:473–87.
Article
21. Denny JC, Collins FS. Precision medicine in 2030-seven ways to transform healthcare. Cell. 2021; 184:1415–9.
Article
22. Meyerson M, Gabriel S, Getz G. Advances in understanding cancer genomes through second-generation sequencing. Nat Rev Genet. 2010; 11:685–96.
Article
23. Cheng DT, Mitchell TN, Zehir A, Shah RH, Benayed R, Syed A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015; 17:251–64.
24. Bolivar AM, Luthra R, Mehrotra M, Chen W, Barkoh BA, Hu P, et al. Targeted next-generation sequencing of endometrial cancer and matched circulating tumor DNA: identification of plasma-based, tumor-associated mutations in early stage patients. Mod Pathol. 2019; 32:405–14.
Article
25. Cancer Genome Atlas Research Network; Albert Einstein College of Medicine; Analytical Biological Services; Barretos Cancer Hospital; Baylor College of Medicine; Beckman Research Institute of City of Hope, et al. Integrated genomic and molecular characterization of cervical cancer. Nature. 2017; 543:378–84.
26. Vang R, Levine DA, Soslow RA, Zaloudek C, Shih Ie M, Kurman RJ. Molecular alterations of TP53 are a defining feature of ovarian high-grade serous carcinoma: a rereview of cases lacking TP53 mutations in the Cancer Genome Atlas Ovarian Study. Int J Gynecol Pathol. 2016; 35:48–55.
Article
27. Ross JS, Ali SM, Wang K, Palmer G, Yelensky R, Lipson D, et al. Comprehensive genomic profiling of epithelial ovarian cancer by next generation sequencing-based diagnostic assay reveals new routes to targeted therapies. Gynecol Oncol. 2013; 130:554–9.
Article
28. Wei S, Hu M, Yang Y, Huang X, Li B, Ding L, et al. Case report: short-term response to first-line crizotinib monotherapy in a metastatic lung adenocarcinoma patient harboring a novel TPR-ROS1 fusion. Front Oncol. 2022; 12:862008.
Article
29. Kao YC, Suurmeijer AJ, Argani P, Dickson BC, Zhang L, Sung YS, et al. Soft tissue tumors characterized by a wide spectrum of kinase fusions share a lipofibromatosis-like neural tumor pattern. Genes Chromosomes Cancer. 2020; 59:575–83.
Article
30. Pascual J, Turner NC. Targeting the PI3-kinase pathway in triple-negative breast cancer. Ann Oncol. 2019; 30:1051–60.
Article
31. Hastings PJ, Lupski JR, Rosenberg SM, Ira G. Mechanisms of change in gene copy number. Nat Rev Genet. 2009; 10:551–64.
Article
32. Pilie PG, Tang C, Mills GB, Yap TA. State-of-the-art strategies for targeting the DNA damage response in cancer. Nat Rev Clin Oncol. 2019; 16:81–104.
Article
33. Li K, Luo H, Huang L, Luo H, Zhu X. Microsatellite instability: a review of what the oncologist should know. Cancer Cell Int. 2020; 20:16.
Article
34. Hampel H, Frankel W, Panescu J, Lockman J, Sotamaa K, Fix D, et al. Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients. Cancer Res. 2006; 66:7810–7.
Article
35. Ferguson SE, Aronson M, Pollett A, Eiriksson LR, Oza AM, Gallinger S, et al. Performance characteristics of screening strategies for Lynch syndrome in unselected women with newly diagnosed endometrial cancer who have undergone universal germline mutation testing. Cancer. 2014; 120:3932–9.
Article
36. Klempner SJ, Fabrizio D, Bane S, Reinhart M, Peoples T, Ali SM, et al. Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: a review of current evidence. Oncologist. 2020; 25:e147–59.
Article
37. Lee SE, Lee MS, Jeon YK, Shim HS, Kang J, Kim J, et al. Interlaboratory comparison study (ring test) of next-generation sequencing-based NTRK fusion detection in South Korea. Cancer Res Treat. 2023; 55:28–40.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr