Tumor genomic profiling and personalized tracking of circulating tumor DNA in Vietnamese colorectal cancer patients

Huu Thinh Nguyen, Trieu Vu Nguyen, Van-Anh Nguyen Hoang, Duc Huy Tran, Ngoc An Le Trinh, Minh Triet Le, Tuan-Anh Nguyen Tran, Thanh Huyen Pham, Thi Cuc Dinh, Tien Sy Nguyen, Ky Cuong Nguyen The, Hoa Mai, Minh Tuan Chu, Dinh Hoang Pham, Xuan Chi Nguyen, Thien My Ngo Ha, Duy Sinh Nguyen, Du Quyen Nguyen, Y-Thanh Lu, Thanh Thuy Do Thi, Dinh Kiet Truong, Quynh Tho Nguyen, Hoai-Nghia Nguyen, Hoa Giang, Lan N Tu, Huu Thinh Nguyen, Trieu Vu Nguyen, Van-Anh Nguyen Hoang, Duc Huy Tran, Ngoc An Le Trinh, Minh Triet Le, Tuan-Anh Nguyen Tran, Thanh Huyen Pham, Thi Cuc Dinh, Tien Sy Nguyen, Ky Cuong Nguyen The, Hoa Mai, Minh Tuan Chu, Dinh Hoang Pham, Xuan Chi Nguyen, Thien My Ngo Ha, Duy Sinh Nguyen, Du Quyen Nguyen, Y-Thanh Lu, Thanh Thuy Do Thi, Dinh Kiet Truong, Quynh Tho Nguyen, Hoai-Nghia Nguyen, Hoa Giang, Lan N Tu

Abstract

Background: Colorectal cancer (CRC) is the fifth most common cancer with rising prevalence in Vietnam. However, there is no data about the mutational landscape and actionable alterations in the Vietnamese patients. During post-operative surveillance, clinical tools are limited to stratify risk of recurrence and detect residual disease.

Method: In this prospective multi-center study, 103 CRC patients eligible for curative-intent surgery were recruited. Genomic DNA from tumor tissue and paired white blood cells were sequenced to profile all tumor-derived somatic mutations in 95 cancer-associated genes. Our bioinformatic algorithm identified top mutations unique for individual patient, which were then used to monitor the presence of circulating tumor DNA (ctDNA) in serial plasma samples.

Results: The top mutated genes in our cohort were APC, TP53 and KRAS. 41.7% of the patients harbored KRAS and NRAS mutations predictive of resistance to Cetuximab and Panitumumab respectively; 41.7% had mutations targeted by either approved or experimental drugs. Using a personalized subset of top ranked mutations, we detected ctDNA in 90.5% of the pre-operative plasma samples, whereas carcinoembryonic antigen (CEA) was elevated in only 41.3% of them. Interim analysis after 16-month follow-up revealed post-operative detection of ctDNA in two patients that had recurrence, with the lead time of 4-10.5 months ahead of clinical diagnosis. CEA failed to predict recurrence in both cases.

Conclusion: Our assay showed promising dual clinical utilities in residual cancer surveillance and actionable mutation profiling for targeted therapies in CRC patients. This could lay foundation to empower precision cancer medicine in Vietnam and other developing countries.

Keywords: circulating tumor (ctDNA); minimal residual disease (MRD); mutational landscape; next-generation sequencing (NGS); somatic mutation.

Conflict of interest statement

V-AH, T-AT, TH, DQN, Y-TL, H-NN, HG and LT are current employees of Gene Solutions, Vietnam. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Nguyen, Nguyen, Nguyen Hoang, Tran, Le Trinh, Le, Nguyen Tran, Pham, Dinh, Nguyen, Nguyen The, Mai, Chu, Pham, Nguyen, Ngo Ha, Nguyen, Nguyen, Lu, Do Thi, Truong, Nguyen, Nguyen, Giang and Tu.

Figures

Figure 1
Figure 1
Schematic of study design and K-Track® assay. (A) 103 patients with primary colorectal cancer stage I-IV, eligible for curative-intent surgery were enrolled. Serial plasma samples were collected before surgery and at scheduled visits after surgery. FFPE samples of surgically removed tumors were also collected. Clinical outcomes were recorded at each visit. (B) Genomic DNA of paired FFPE and WBC were sequenced to profile all tumor-specific somatic alterations in 95 cancer-associated genes. Top 5 mutations were selected by our K-Track® scoring algorithm and then used to monitor ctDNA presence in plasma samples by a bespoke multiplex PCR assay and ultra-deep sequencing at an average of 100,000X.
Figure 2
Figure 2
Mutational spectrum of 95 genes in the Vietnamese colorectal cancer patients. (A) The average number of tumor-derived mutations was 7 mutations per patient and not different by stage. (B) The mutation burden was not different by the tumor site. (C) Pie chart showing the distribution of mutation classes identified in 95 genes. (D) The top 25 significantly mutated genes in our cohort. (E) Mutation frequency of top mutated genes in our cohort was compared with published datasets of Caucasian and Asian cohorts. (F) Mutually exclusive and co-occurring mutated genes in our dataset. *P < 0.05; Kruskal-Wallis and post hoc Dunn’s test for (A, B).
Figure 3
Figure 3
Oncogenic signaling pathways and actionable alterations in the Vietnamese colorectal cancer patients. (A) The top three signaling pathways with frequent oncogenic alterations in our cohort were Wnt/β-catenin signaling, genome integrity, and MAPK signaling. (B) Proportions of patients harboring mutations in KRAS and NRAS predictive of resistance to Cetuximab and Panitumumab respectively. Frequency of the specific resistance mutations was also illustrated. (C) Proportions of patients carrying mutations that are candidate biomarkers for response to drugs with compelling clinical evidence (level 3) or laboratory evidence (level 4) as classified by the OncoKB database.
Figure 4
Figure 4
Detection of ctDNA in plasma samples. (A) The average number of mutations selected to track was 5 mutations per patient regardless of cancer stage. (B) Detection rate of ctDNA in pre-operative plasma samples was 90.5%. (C) Pre-operative ctDNA detection rate was associated with TNM stage, as the rate in stage I was significantly lower than in stage II and III. Nodal involvement, histological grade and CEA level status did not affect the detection rate. (D) Pre-operative CEA level was found elevated (≥5 ng/mL) in only 41.3% patients. (E) Total levels of cfDNA were not different between pre-operative and post-operative plasma samples while ctDNA and CEA levels significantly reduced after surgery. *P < 0.05; Kruskal-Wallis and post hoc Dunn’s test for (A); Chi-squared test and Fisher’s exact test for (C); Mann-Whitney U test for (E).
Figure 5
Figure 5
Longitudinal monitoring of ctDNA and clinical outcomes of patients. (A) Swimmer plot depicting ctDNA results over time and incidence of relapse in 19 patients that had been followed up for at least 16 months. This was an interim analysis as the study is on-going. (B, C) Longitudinal plot showing the mean VAF of ctDNA, CEA level, treatment and clinical status over time of patients ZMC002 and ZMC006. Molecular relapse detection was 4 months earlier than clinically diagnosed relapse in patient ZMC006. CEA level was still normal at the time point when ctDNA was found positive. Op, operation, CRT, chemoradiotherapy.

References

    1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. . Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin (2021) 71:209–49. doi: 10.3322/caac.21660
    1. Le DD, Vo TV, Sarakarn P. Overall survival rate of Vietnamese patients with colorectal cancer: A hospital-based cohort study in the central region of Vietnam. Asian Pac J Cancer Prev (2021) 22:3569–75. doi: 10.31557/APJCP.2021.22.11.3569
    1. Parikh AR, Van Seventer EE, Siravegna G, Hartwig AV, Jaimovich A, He Y, et al. . Minimal residual disease detection using a plasma-only circulating tumor DNA assay in patients with colorectal cancer. Clin Cancer Res (2021) 27:5586–94. doi: 10.1158/1078-0432.CCR-21-0410
    1. Fang L, Yang Z, Zhang M, Meng M, Feng J, Chen C. Clinical characteristics and survival analysis of colorectal cancer in China: a retrospective cohort study with 13,328 patients from southern China. Gastroenterol Rep (Oxf) (2021) 9:571–82. doi: 10.1093/gastro/goab048
    1. Duffy MJ. Carcinoembryonic antigen as a marker for colorectal cancer: is it clinically useful? Clin Chem (2001) 47:624–30. doi: 10.1093/clinchem/47.4.624
    1. Sørensen CG, Karlsson WK, Pommergaard HC, Burcharth J, Rosenberg J. The diagnostic accuracy of carcinoembryonic antigen to detect colorectal cancer recurrence - a systematic review. Int J Surg (2016) 25:134–44. doi: 10.1016/j.ijsu.2015.11.065
    1. Reinert T, Henriksen TV, Christensen E, Sharma S, Salari R, Sethi H, et al. . Analysis of plasma cell-free DNA by ultradeep sequencing in patients with stages I to III colorectal cancer. JAMA Oncol (2019) 5:1124–31. doi: 10.1001/jamaoncol.2019.0528
    1. Tie J, Cohen JD, Wang Y, Christie M, Simons K, Lee M, et al. . Circulating tumor DNA analyses as markers of recurrence risk and benefit of adjuvant therapy for stage III colon cancer. JAMA Oncol (2019) 5:1710–7. doi: 10.1001/jamaoncol.2019.3616
    1. DePristo MA, Banks E, Poplin R, Garimella KV, Maguire JR, Hartl C, et al. . A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet (2011) 43:491–8. doi: 10.1038/ng.806
    1. Andrews S. FastQC: A quality control tool for high throughput sequence data (2010). Available at: .
    1. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA-MEM. arXiv: Genomics (2013). Available at: .
    1. Picard. Broad Institute. Available at: .
    1. McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. . The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res (2010) 20:1297–303. doi: 10.1101/gr.107524.110
    1. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res (2018) 28:1747–56. doi: 10.1101/gr.239244.118
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for illumina sequence data. Bioinformatics (2014) 30:2114–20. doi: 10.1093/bioinformatics/btu170
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. . The sequence Alignment/Map format and SAMtools. Bioinformatics (2009) 25:2078–9. doi: 10.1093/bioinformatics/btp352
    1. Cancer Genome Atlas Network . Comprehensive molecular characterization of human colon and rectal cancer. Nature (2012) 487:330–7. doi: 10.1038/nature11252
    1. Ellrott K, Bailey MH, Saksena G, Covington KR, Kandoth C, Stewart C, et al. . Scalable open science approach for mutation calling of tumor exomes using multiple genomic pipelines. Cell Syst (2018) 6:271–281.e7. doi: 10.1016/j.cels.2018.03.002
    1. Giannakis M, Mu XJ, Shukla SA, Qian ZR, Cohen O, Nishihara R, et al. . Genomic correlates of immune-cell infiltrates in colorectal carcinoma. Cell Rep (2016) 15:857–65. doi: 10.1016/j.celrep.2016.03.075
    1. Huang W, Li H, Shi X, Lin M, Liao C, Zhang S, et al. . Characterization of genomic alterations in Chinese colorectal cancer patients. Jpn J Clin Oncol (2021) 51:120–9. doi: 10.1093/jjco/hyaa182
    1. Lee CS, Song IH, Lee A, Kang J, Lee YS, Lee IK, et al. . Enhancing the landscape of colorectal cancer using targeted deep sequencing. Sci Rep (2021) 11:8154. doi: 10.1038/s41598-021-87486-3
    1. Chakravarty D, Gao J, Phillips S, Kundra R, Zhang H, Wang J, et al. . OncoKB: a precision oncology knowledge base. JCO Precis Oncol (2017) 1:1–16. doi: 10.1200/PO.17.00011
    1. Zhao Q, Wang F, Chen YX, Chen S, Yao YC, Zeng ZL, et al. . Comprehensive profiling of 1015 patients' exomes reveals genomic-clinical associations in colorectal cancer. Nat Commun (2022) 13:2342. doi: 10.1038/s41467-022-30062-8
    1. Su MW, Chang CK, Lin CW, Chu HW, Tsai TN, Su WC, et al. . Genomic and metabolomic landscape of right-sided and left-sided colorectal cancer: Potential preventive biomarkers. Cells (2022) 11(3):527. doi: 10.3390/cells11030527
    1. Zhuang Y, Wang H, Jiang D, Li Y, Feng L, Tian C, et al. . Multi gene mutation signatures in colorectal cancer patients: predict for the diagnosis, pathological classification, staging and prognosis. BMC Cancer (2021) 21:380. doi: 10.1186/s12885-021-08108-9
    1. Chen M, Zhang Q, Chen H, Wang Z, Xu T, QI C, et al. . FAT4 mutation as a potential predictive biomarker for immunotherapy combined with anti-angiogenic therapy in MSS metastatic colorectal cancer. J Clin Oncol (2022) 40:e15504–4. doi: 10.1200/JCO.2022.40.16_suppl.e15504
    1. Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, et al. . Oncogenic signaling pathways in the cancer genome atlas. Cell (2018) 173:321–337.e10. doi: 10.1016/j.cell.2018.03.035
    1. Abbosh C, Birkbak NJ, Wilson GA, Jamal-Hanjani M, Constantin T, Salari R, et al. . Phylogenetic ctDNA analysis depicts early-stage lung cancer evolution. Nature (2017) 545:446–51. doi: 10.1038/nature22364
    1. Henriksen TV, Reinert T, Rasmussen MH, Demuth C, Løve US, Madsen AH, et al. . Comparing single-target and multitarget approaches for postoperative circulating tumour DNA detection in stage II-III colorectal cancer patients. Mol Oncol (2022) 16(20):3654–65. doi: 10.1002/1878-0261.13294
    1. 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. doi: 10.1038/nm.3519
    1. Diefenbach RJ, Lee JH, Stewart A, Menzies AM, Carlino MS, Saw RPM, et al. . Anchored multiplex PCR custom melanoma next generation sequencing panel for analysis of circulating tumor DNA. Front Oncol (2022) 12:820510. doi: 10.3389/fonc.2022.820510
    1. Poh J, Ngeow KC, Pek M, Tan KH, Lim JS, Chen H, et al. . Analytical and clinical validation of an amplicon-based next generation sequencing assay for ultrasensitive detection of circulating tumor DNA. PloS One (2022) 17:e0267389. doi: 10.1371/journal.pone.0267389
    1. Watanabe K, Nakamura Y, Low SK. Clinical implementation and current advancement of blood liquid biopsy in cancer. J Hum Genet (2021) 66:909–26. doi: 10.1038/s10038-021-00939-5
    1. Tarazona N, Gimeno-Valiente F, Gambardella V, Zuñiga S, Rentero-Garrido P, Huerta M, et al. . Targeted next-generation sequencing of circulating-tumor DNA for tracking minimal residual disease in localized colon cancer. Ann Oncol (2019) 30:1804–12. doi: 10.1093/annonc/mdz390
    1. Schøler LV, Reinert T, Ørntoft MW, Kassentoft CG, Árnadóttir SS, Vang S, et al. . Clinical implications of monitoring circulating tumor DNA in patients with colorectal cancer. Clin Cancer Res (2017) 23:5437–45. doi: 10.1158/1078-0432.CCR-17-0510
    1. Shirasu H, Taniguchi H, Watanabe J, Kotaka M, Yamazaki K, Hirata K, et al. . O-11 monitoring molecular residual disease by circulating tumor DNA in resectable colorectal cancer: Molecular subgroup analyses of a prospective observational study GALAXY in CIRCULATE-Japan. Ann Oncol (2021) 32:S222–3. doi: 10.1016/j.annonc.2021.05.015
    1. Henriksen TV, Tarazona N, Frydendahl A, Reinert T, Gimeno-Valiente F, Carbonell-Asins JA, et al. . Circulating tumor DNA in stage III colorectal cancer, beyond minimal residual disease detection, toward assessment of adjuvant therapy efficacy and clinical behavior of recurrences. Clin Cancer Res (2022) 28:507–17. doi: 10.1158/1078-0432.CCR-21-2404
    1. Vidal J, Casadevall D, Bellosillo B, Pericay C, Garcia-Carbonero R, Losa F, et al. . Clinical impact of presurgery circulating tumor DNA after total neoadjuvant treatment in locally advanced rectal cancer: A biomarker study from the GEMCAD 1402 trial. Clin Cancer Res (2021) 27:2890–8. doi: 10.1158/1078-0432.CCR-20-4769

Source: PubMed

3
구독하다