Subtyping of metastatic breast cancer based on plasma circulating tumor DNA alterations: An observational, multicentre platform study

Zhe-Yu Hu, Yu Tang, Liping Liu, Ning Xie, Can Tian, Binliang Liu, Lixin Zou, Wei Zhou, Yikai Wang, Xuefeng Xia, Quchang Ouyang, Zhe-Yu Hu, Yu Tang, Liping Liu, Ning Xie, Can Tian, Binliang Liu, Lixin Zou, Wei Zhou, Yikai Wang, Xuefeng Xia, Quchang Ouyang

Abstract

Background: After multiple lines of therapies, no guideline or consensus is currently available for the treatment of patients with metastatic breast cancer. This study aims to evaluate the efficacy of a novel re-subtyping and treatment strategy based on ctDNA alterations.

Methods: This observational, multicentre study recruited 223 patients with metastatic breast cancer intending to receive late-line therapy from Dec 1, 2016, to June 31, 2019. This study took place in Hunan Cancer Hospital, the Forth Hospital of Changsha and Zhuzhou Central Hospital in China. ctDNA alterations were assessed by next-generation sequencing (NGS). Patients with druggable ctDNA alterations were treated with corresponding targeted drugs which are clinically available. Other patients received physician-chosen treatment. This study was registered with ClinicalTrials.gov, NCT05079074.

Findings: The progression-free survival (hazard ratio: 0.45, 95% Confidence Interval (CI): 0.33-0.62, P < 0.0001) and disease control rate (89.4% vs. 65.9%, P < 0.0001) were significantly improved in patients who received druggable ctDNA alteration-guided therapy compared with those of patients who received physician-chosen treatment. ctDNA alterations with top rank and high clustering scores were classified into four subtypes based on their functions as follows: 1) extracellular function (ECF), 2) cell proliferation (CP), 3) nuclear function (NF), and 4) cascade signaling pathway (CSP). A significant benefit from ctDNA alteration-guided treatment was observed in patients with NF and CSP ctDNA alterations, with hazard ratios of 0.39 (95% CI: 0.24-0.65, P = 0.0003) and 0.14 (95% CI: 0.04-0.46, P < 0.0001), respectively.

Interpretation: After multiline traditional pathological HR/HER2 subtype-guided therapies, ctDNA testing could identify druggable ctDNA alterations to guide late-line therapy for patients with metastatic breast cancer.

Funding: This work was supported by Key Grants of Research and Development in Hunan Province (2018SK2124, 2018SK2120), Natural Science Foundation of Hunan (2019JJ50360), Hunan Provincial Health Commission Project (B2019085, B2019089 and C2019070), and Changsha Science and Technology Project (kq2004125 and kq2004137).

Keywords: Disease control rate (DCR); Druggable circulating tumor DNA alterations; Genetic alterations of functional pathways; Late-line therapy; Progression-free survival (PFS).

Conflict of interest statement

The authors declare that they have no conflicts of interest.

© 2022 The Authors.

Figures

Figure 1
Figure 1
Kaplan–Meier plot of progression-free survival. Dashes represent censored patients. HR=hazard ratio. Univariate Cox regression analysis was performed to calculate the hazard ratio (HR) with 95% confidence interval (CI) of progression in the ctDNA-guided LLT group versus the traditional group.
Figure 2
Figure 2
Kaplan–Meier plot of progression-free survival. Dashes represent censored patients. HR=hazard ratio. Univariate Cox regression analysis was performed to calculate the hazard ratio (HR) with 95% confidence interval (CI) of progression in the ctDNA-guided LLT group versus the group of the 58 patients with ctDNA alterations but for whom no drugs were available.
Figure 3
Figure 3
ctDNA alterations. A. Heatmaps of baseline ctDNA alteration profiles and the corresponding treatment strategies for patients with B. Circle plot of 442 ctDNA alterations among 420 samples from 223 patients. Genes were clustered by the ‘complete’ method of the hclust function in R. The colors indicate the cutree score, ranging from 1 (blue) to 60 (red). Nearly three-fourths of all ctDNA alterations had a low score (blue, left and bottom regions of the circle).
Figure 3
Figure 3
ctDNA alterations. A. Heatmaps of baseline ctDNA alteration profiles and the corresponding treatment strategies for patients with B. Circle plot of 442 ctDNA alterations among 420 samples from 223 patients. Genes were clustered by the ‘complete’ method of the hclust function in R. The colors indicate the cutree score, ranging from 1 (blue) to 60 (red). Nearly three-fourths of all ctDNA alterations had a low score (blue, left and bottom regions of the circle).
Figure 4
Figure 4
Subgroup analyses of hazard ratios for progression-free survival by ctDNA-based subtype.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70(1):7–30.
    1. Holleczek B, Arndt V, Stegmaier C, Brenner H. Trends in breast cancer survival in Germany from 1976 to 2008–a period analysis by age and stage. Cancer Epidemiol. 2011;35(5):399–406.
    1. Cardoso F, Spence D, Mertz S, et al. Global analysis of advanced/metastatic breast cancer: decade report (2005-2015) Breast. 2018;39:131–138.
    1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7–33.
    1. DeSantis CE, Ma J, Gaudet MM, et al. Breast cancer statistics, 2019. CA Cancer J Clin. 2019;69(6):438–451.
    1. Sundquist M, Brudin L, Tejler G. Improved survival in metastatic breast cancer 1985-2016. Breast. 2017;31:46–50.
    1. Anders CK, Peppercorn J. Treating in the dark: unanswered questions on costs and benefits of late line therapy for metastatic breast cancer. Cancer Invest. 2009;27(1):13–16.
    1. Curtit E, Nerich V, Mansi L, et al. Discordances in estrogen receptor status, progesterone receptor status, and HER2 status between primary breast cancer and metastasis. Oncologist. 2013;18(6):667–674.
    1. Zardavas D, Irrthum A, Swanton C, Piccart M. Clinical management of breast cancer heterogeneity. Nat Rev Clin Oncol. 2015;12(7):381–394.
    1. Hu ZY, Xie N, Tian C, et al. Identifying circulating tumor DNA mutation profiles in metastatic breast cancer patients with multiline resistance. EBioMedicine. 2018;32:111–118.
    1. Metastatic ER+ Breast cancer's genomic landscape. Cancer Discov. 2017;7(2):OF2.
    1. Swallow E, Zhang J, Thomason D, Tan RD, Kageleiry A, Signorovitch J. Real-world patterns of endocrine therapy for metastatic hormone-receptor-positive (HR+)/human epidermal growth factor receptor-2-negative (HER2-) breast cancer patients in the United States: 2002-2012. Curr Med Res Opin. 2014;30(8):1537–1545.
    1. Yang Y, Feng J, Li T, Ge F, Zhao J. CyanOmics: an integrated database of omics for the model cyanobacterium Synechococcus sp. PCC 7002. Database. 2015;2015:1–9.
    1. Hu X, Zhang J, Xu B, et al. Multicenter phase II study of apatinib, a novel VEGFR inhibitor in heavily pretreated patients with metastatic triple-negative breast cancer. Int J Cancer. 2014;135(8):1961–1969.
    1. Gao JJ, Cheng J, Bloomquist E, et al. CDK4/6 inhibitor treatment for patients with hormone receptor-positive, HER2-negative, advanced or metastatic breast cancer: a US Food and Drug Administration pooled analysis. Lancet Oncol. 2020;21(2):250–260.
    1. Robson ME, Tung N, Conte P, et al. OlympiAD final overall survival and tolerability results: Olaparib versus chemotherapy treatment of physician's choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol. 2019;30(4):558–566.
    1. Ma F, Ouyang Q, Li W, et al. Pyrotinib or lapatinib combined with capecitabine in HER2-positive metastatic breast cancer with prior taxanes, anthracyclines, and/or trastuzumab: a randomized, Phase II study. J Clin Oncol. 2019;37(29):2610–2619.
    1. Mounsey LA, Deal AM, Keith KC, et al. Changing natural history of HER2-positive breast cancer metastatic to the brain in the era of new targeted therapies. Clin Breast Cancer. 2018;18(1):29–37.
    1. Baselga J, Im SA, Iwata H, et al. Buparlisib plus fulvestrant versus placebo plus fulvestrant in postmenopausal, hormone receptor-positive, HER2-negative, advanced breast cancer (BELLE-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(7):904–916.
    1. Ni J, Ramkissoon SH, Xie S, et al. Combination inhibition of PI3K and mTORC1 yields durable remissions in mice bearing orthotopic patient-derived xenografts of HER2-positive breast cancer brain metastases. Nat Med. 2016;22(7):723–726.
    1. Turner NC, Kingston B, Kilburn LS, et al. Circulating tumour DNA analysis to direct therapy in advanced breast cancer (plasmaMATCH): a multicentre, multicohort, phase 2a, platform trial. Lancet Oncol. 2020;21(10):1296–1308.
    1. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523–533.
    1. Jiang Z, Li W, Hu X, et al. Tucidinostat plus exemestane for postmenopausal patients with advanced, hormone receptor-positive breast cancer (ACE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2019;20(6):806–815.
    1. Yuan P, Hu X, Sun T, et al. Eribulin mesilate versus vinorelbine in women with locally recurrent or metastatic breast cancer: a randomised clinical trial. Eur J Cancer. 2019;112:57–65.
    1. Im SA, Xu B, Li W, et al. Olaparib monotherapy for Asian patients with a germline BRCA mutation and HER2-negative metastatic breast cancer: OlympiAD randomized trial subgroup analysis. Sci Rep. 2020;10(1):8753.
    1. Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature. 2013;497(7447):108–112.
    1. Esposito A, Criscitiello C, Locatelli M, Milano M, Curigliano G. Liquid biopsies for solid tumors: Understanding tumor heterogeneity and real time monitoring of early resistance to targeted therapies. Pharmacol Ther. 2016;157:120–124.
    1. Swanton C, Govindan R. Clinical implications of genomic discoveries in lung cancer. N Engl J Med. 2016;374(19):1864–1873.
    1. Chabon JJ, Simmons AD, Lovejoy AF, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7:11815.
    1. Diehl F, Li M, Dressman D, et al. Detection and quantification of mutations in the plasma of patients with colorectal tumors. Proc Natl Acad Sci USA. 2005;102(45):16368–16373.

Source: PubMed

3
Abonneren