Genetic landscape and personalized tracking of tumor mutations in Vietnamese women with breast cancer

Van-Anh Nguyen Hoang, Sao Trung Nguyen, Trieu Vu Nguyen, Thanh Huyen Pham, Phuoc Loc Doan, Ngoc Thanh Nguyen Thi, Minh Long Nguyen, Thi Cuc Dinh, Dinh Hoang Pham, Ngoc Mai Nguyen, Duy Sinh Nguyen, Du Quyen Nguyen, Y-Thanh Lu, Thanh Thuy Thi Do, Dinh Kiet Truong, Minh-Duy Phan, Hoai-Nghia Nguyen, Hoa Giang, Lan N Tu, Van-Anh Nguyen Hoang, Sao Trung Nguyen, Trieu Vu Nguyen, Thanh Huyen Pham, Phuoc Loc Doan, Ngoc Thanh Nguyen Thi, Minh Long Nguyen, Thi Cuc Dinh, Dinh Hoang Pham, Ngoc Mai Nguyen, Duy Sinh Nguyen, Du Quyen Nguyen, Y-Thanh Lu, Thanh Thuy Thi Do, Dinh Kiet Truong, Minh-Duy Phan, Hoai-Nghia Nguyen, Hoa Giang, Lan N Tu

Abstract

Breast cancer is the leading cause of cancer death in Vietnamese women, but its mutational landscape and actionable alterations for targeted therapies remain unknown. After treatment, a sensitive biomarker to complement conventional imaging to monitor patients is also lacking. In this prospective multi-center study, 134 early-stage breast cancer patients eligible for curative-intent surgery were recruited. Genomic DNA from tumor tissues and paired white blood cells were sequenced to profile all tumor-derived mutations in 95 cancer-associated genes. Our bioinformatic algorithm was then utilized to identify top mutations for individual patients. Serial plasma samples were collected before surgery and at scheduled visits after surgery. Personalized assay tracking the selected mutations were performed to detect circulating tumor DNA (ctDNA) in the plasma. We found that the mutational landscape of the Vietnamese was largely similar to other Asian cohorts, showing higher TP53 mutation frequency than in Caucasians. Alterations in PIK3CA and PI3K signaling were dominant, particularly in our triple-negative subgroup. Using top-ranked mutations, we detected ctDNA in pre-operative plasma in 24.6-43.5% of the hormone-receptor-positive groups and 76.9-80.8% of the hormone-receptor-negative groups. The detection rate was associated with breast cancer subtypes and clinicopathological features that increased the risk of relapse. Interim analysis after a 15-month follow-up revealed post-operative detection of ctDNA in all three patients that had recurrence, with a lead time of 7-13 months ahead of clinical diagnosis. Our personalized assay is streamlined and affordable with promising clinical utility in residual cancer surveillance. We also generated the first somatic variant dataset for Vietnamese breast cancer women that could lay the foundation for precision cancer medicine in Vietnam.

Keywords: circulating tumor DNA; minimal residual disease; mutational landscape; next-generation sequencing; somatic mutation.

Conflict of interest statement

V‐ANH, PLD, NTNT, MLN, NMN, DQN, Y‐TL, M‐DP, HG and LNT are current employees of Gene Solutions, Vietnam. The remaining authors declare no conflict of interest.

© 2022 Gene Solutions JSC and The Authors. Molecular Oncology published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies.

Figures

Fig. 1
Fig. 1
Schematic of study design and K‐track® assay. (A) 134 patients with primary breast cancer stage I‐III, eligible for curative‐intent surgery were enrolled. Serial plasma samples were collected before surgery and after surgery at scheduled visits. Formalin‐fixed paraffin‐embedded (FFPE) samples of surgically removed tumors were also collected. Patients were followed‐up to record clinical outcomes. (B) Paired FFPE and white blood cells (WBC) DNA of the same individual were sequenced to identify tumor‐specific somatic mutations in 95 cancer‐associated genes. Top 4–5 mutations were selected by our scoring algorithm and tracked in plasma samples using a bespoke multiplex polymerase chain reaction (mPCR) assay and ultra‐deep next‐generation sequencing (NGS).
Fig. 2
Fig. 2
Mutational spectrum of 95 genes in Vietnamese breast cancer women. (A) The number of somatic mutations identified per patient was significantly higher in the HR− HER2+ compared to the HR+ groups. (B) The mutation burden was significantly lower in stage I compared to stage II and III. (C) Pie chart showing the distribution of mutation classes identified in 95 genes. (D) The top 25 significantly mutated genes in the cohort. (E) Frequency of top highly mutated genes in breast cancer was compared between our cohort and published datasets of Caucasian and Asian cohorts. (F) Pairwise analysis identified oncogenic alternations significantly associated with different subtypes. Genes in green color were significantly more altered in a particular group compared to the rest while genes in black color were significantly less altered. HR, Hormone receptors; HER2, Human epidermal growth factor receptor 2. *P < 0.05; Kruskal–Wallis and post hoc Dunn's test for (A) and (B); Fisher's exact test for (F). (A), (B): Boxplots with Tukey whiskers.
Fig. 3
Fig. 3
Oncogenic signaling pathways and actionable alterations in breast cancer subtypes. (A) The top three signaling pathways with frequent oncogenic alterations in our cohort were genome integrity, PI3K signaling and chromatin SWI/SNF remodeling complex. (B) The top three altered signaling pathways were different among breast cancer subtypes. HR+ HER2− had significantly less alterations in genome integrity pathway compared to other groups. Transcription factor pathway was highly mutated specifically in the HR+ HER2− group. (C) Proportion of patients in each subtype that had actionable alterations predictive of treatment response to a drug at different levels of evidence stratified by OncoKB database. HR, Hormone receptors; HER2, Human epidermal growth factor receptor 2. *P < 0.05; Fisher's exact test for (B).
Fig. 4
Fig. 4
Analysis of circulating tumor DNA (ctDNA) in pre‐operative plasma samples. (A) The number of mutations selected to track in each patient was significantly lower in the HR+ HER2− group than in the HR− HER2− group. (B) The percentage of tracked mutations that could be detected in the plasma was significantly lower in the HR+ groups compared to HR‐ groups. (C) Pre‐operative detection rate was associated with breast cancer subtypes and was significantly lower in the HR+ groups compared to HR− groups. (D) The detection rate of PIK3CA H1047R/L in the plasma followed the exact same trend as the overall detection rate even though the mutation was identified at similar variant allele frequency (VAF) in tumors across subtypes. (E) When HR+ groups were stratified by clinicopathological features that increase risk of relapse, the detection rate was significantly lower in the low‐risk than the high‐risk group of HR+ HER2−. (F) Detection was associated with TNM stage as the detection rate in stage I was significantly lower than in stage II and III. Nodal and grade status were not found to affect pre‐operative detection rate. (G) Levels of pre‐operative and post‐operative cell‐free DNA (cfDNA) were not different while VAF of ctDNA significantly reduced after surgery. HR, Hormone receptors; HER2, Human epidermal growth factor receptor 2. *P < 0.05; Kruskal–Wallis and post hoc Dunn's test for (A), (B), (D); Mann–Whitney U test for (G); Chi‐squared test and Fisher's exact test for (C–F). (A), (B), (D), (G): Boxplots with Tukey whiskers.
Fig. 5
Fig. 5
Longitudinal monitoring of circulating tumor DNA (ctDNA) and clinical outcomes. (A) Swimmer plot depicting serial ctDNA results over time and incidence of relapse or metastasis of 32 patients that had been followed up for at least 15 months. This was an interim analysis as the clinical study is ongoing. (B, C) longitudinal plot showing the mean VAF of ctDNA, treatment and clinical status over time of patients ZMB022 and ZMB041. CRT, Chemoradiotherapy; Op, Operation. Molecular relapse detection was 13 months earlier than clinically diagnosed relapse in patient ZMB041.

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. 10.3322/caac.21660
    1. Suh KJ, Kim SH, Kim YJ, Shin H, Kang E, Kim EK, et al. Clinical application of next‐generation sequencing in patients with breast cancer: real‐world data. J Breast Cancer. 2022;25:366–78. 10.4048/jbc.2022.25.e30
    1. Lan NH, Laohasiriwong W, Stewart JF. Survival probability and prognostic factors for breast cancer patients in Vietnam. Glob Health Action. 2013;6:1–9.
    1. Coombes RC, Page K, Salari R, Hastings RK, Armstrong A, Ahmed S, et al. Personalized detection of circulating tumor DNA antedates breast cancer metastatic recurrence. Clin Cancer Res. 2019;25:4255–63. 10.1158/1078-0432.ccr-18-3663
    1. Pedersen AC, Sørensen PD, Jacobsen EH, Madsen JS, Brandslund I. Sensitivity of CA 15‐3, CEA and serum HER2 in the early detection of recurrence of breast cancer. Clin Chem Lab Med. 2013;51:1511–9. 10.1515/cclm-2012-0488
    1. Magbanua MJM, Swigart LB, Wu HT, Hirst GL, Yau C, Wolf DM, et al. Circulating tumor DNA in neoadjuvant‐treated breast cancer reflects response and survival. Ann Oncol. 2021;32:229–39. 10.1016/j.annonc.2020.11.007
    1. Cardoso F, van't Veer LJ, Bogaerts J, Slaets L, Viale G, Delaloge S, et al. 70‐gene signature as an aid to treatment decisions in early‐stage breast cancer. N Engl J Med. 2016;375:717–29. 10.1056/NEJMoa1602253
    1. Sheffield KM, Peachey JR, Method MW, Grimes BR, Brown J, Saverno KR, et al. Recurrence risk in early breast cancer as defined by clinicopathologic features. J Clin Oncol. 2021;39:e18581. 10.1200/JCO.2021.39.15_suppl.e18581
    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.
    1. Li H. Aligning sequence reads, clone sequences and assembly contigs with BWA‐MEM. arXiv. 2013. 10.48550/arXiv.1303.3997
    1. Picard. Broad Institute.
    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.
    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.
    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.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–20.
    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. 10.1093/bioinformatics/btp352
    1. Network CGA. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70.
    1. Pereira B, Chin SF, Rueda OM, Vollan HK, Provenzano E, Bardwell HA, et al. The somatic mutation profiles of 2,433 breast cancers refines their genomic and transcriptomic landscapes. Nat Commun. 2016;7:11479.
    1. Kan Z, Ding Y, Kim J, Jung HH, Chung W, Lal S, et al. Multi‐omics profiling of younger Asian breast cancers reveals distinctive molecular signatures. Nat Commun. 2018;9:1725.
    1. Pan J‐W, Zabidi MMA, Ng P‐S, Meng M‐Y, Hasan SN, Sandey B, et al. The molecular landscape of Asian breast cancers reveals clinically relevant population‐specific differences. Nat Commun. 2020;11:6433. 10.1038/s41467-020-20173-5
    1. O'Meara TA, Tolaney SM. Tumor mutational burden as a predictor of immunotherapy response in breast cancer. Oncotarget. 2021;12:394–400.
    1. Wang Y, Lin L, Li L, Wen J, Chi Y, Hao R, et al. Genetic landscape of breast cancer and mutation tracking with circulating tumor DNA in Chinese women. Aging (Albany NY). 2021;13:11860–76.
    1. Silwal‐Pandit L, Vollan HK, Chin SF, Rueda OM, McKinney S, Osako T, et al. TP53 mutation spectrum in breast cancer is subtype specific and has distinct prognostic relevance. Clin Cancer Res. 2014;20:3569–80. 10.1158/1078-0432.ccr-13-2943
    1. Lang GT, Jiang YZ, Shi JX, Yang F, Li XG, Pei YC, et al. Characterization of the genomic landscape and actionable mutations in Chinese breast cancers by clinical sequencing. Nat Commun. 2020;11:5679.
    1. Nguyen J, Le QH, Duong BH, Sun P, Pham HT, Ta VT, et al. A matched case‐control study of risk factors for breast cancer risk in Vietnam. Int J Breast Cancer. 2016;2016:7164623.
    1. Giuli MV, Giuliani E, Screpanti I, Bellavia D, Checquolo S. Notch signaling activation as a Hallmark for triple‐negative breast cancer subtype. J Oncol. 2019;2019:8707053.
    1. Lipsyc‐Sharf M, Bruin EC, Santos K, McEwen R, Stetson D, Patel A, et al. Circulating tumor DNA and late recurrence in high‐risk hormone receptor‐positive, human epidermal growth factor receptor 2‐negative breast cancer. J Clin Oncol. 2022;40:2408–19. 10.1200/jco.22.00908
    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.
    1. Razavi P, Li BT, Brown DN, Jung B, Hubbell E, Shen R, et al. High‐intensity sequencing reveals the sources of plasma circulating cell‐free DNA variants. Nat Med. 2019;25:1928–37.
    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.
    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.
    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.
    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. 10.1038/s10038-021-00939-5
    1. Garcia‐Murillas I, Chopra N, Comino‐Méndez I, Beaney M, Tovey H, Cutts RJ, et al. Assessment of molecular relapse detection in early‐stage breast cancer. JAMA Oncol. 2019;5:1473–8.
    1. Lin PH, Wang MY, Lo C, Tsai LW, Yen TC, Huang TY, et al. Circulating tumor DNA as a predictive marker of recurrence for patients with stage II‐III breast cancer treated with neoadjuvant therapy. Front Oncol. 2021;11:736769.
    1. Moss J, Zick A, Grinshpun A, Carmon E, Maoz M, Ochana BL, et al. Circulating breast‐derived DNA allows universal detection and monitoring of localized breast cancer. Ann Oncol. 2020;31:395–403. 10.1016/j.annonc.2019.11.014
    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. 10.1158/1078-0432.ccr-21-0410

Source: PubMed

3
Subskrybuj