Serial Tumor Molecular Profiling of Newly Diagnosed HER2-Negative Breast Cancers During Chemotherapy in Combination with Angiogenesis Inhibitors

Joan R E Choo, Yi-Hua Jan, Samuel G W Ow, Andrea Wong, Matilda Xinwei Lee, Natalie Ngoi, Kritika Yadav, Joline S J Lim, Siew Eng Lim, Ching Wan Chan, Mikael Hartman, Siau Wei Tang, Boon Cher Goh, Hon Lyn Tan, Wan Qin Chong, Ang Li En Yvonne, Gloria H J Chan, Shu-Jen Chen, Kien Thiam Tan, Soo Chin Lee, Joan R E Choo, Yi-Hua Jan, Samuel G W Ow, Andrea Wong, Matilda Xinwei Lee, Natalie Ngoi, Kritika Yadav, Joline S J Lim, Siew Eng Lim, Ching Wan Chan, Mikael Hartman, Siau Wei Tang, Boon Cher Goh, Hon Lyn Tan, Wan Qin Chong, Ang Li En Yvonne, Gloria H J Chan, Shu-Jen Chen, Kien Thiam Tan, Soo Chin Lee

Abstract

Background: Breast cancers are heterogeneous with variable clinical courses and treatment responses.

Objective: We sought to evaluate dynamic changes in the molecular landscape of HER2-negative tumors treated with chemotherapy and anti-angiogenic agents.

Patients and methods: Newly diagnosed HER2-negative breast cancer patients received low-dose sunitinib or bevacizumab prior to four 2-weekly cycles of dose-dense doxorubicin and cyclophosphamide. Tumor biopsies were obtained at baseline, after 2 weeks and after 8 weeks of chemotherapy. Next-generation sequencing was performed to assess for single nucleotide variants (SNVs) and copy number alterations (CNAs) of 440 cancer-related genes (ACTOnco®). Observed genomic changes were correlated with the Miller-Payne histological response to treatment.

Results: Thirty-four patients received sunitinib and 18 received bevacizumab. In total, 77% were hormone receptor positive (HER2-/HR+) and 23% were triple negative breast cancers (TNBC). New therapy-induced mutations were infrequent, occurring only in 13%, and appeared early after a single cycle of treatment. Seventy-two percent developed changes in the variant allele frequency (VAF) of pathogenic SNVs; the majority (51%) of these changes occurred early at 2 weeks and were sustained for 8 weeks. Changes in VAF of SNVs were most commonly seen in the PI3K/mTOR/AKT pathway; 13% developed changes in pathogenic mutations, which potentially confer sensitivity to PIK3CA inhibitors. Tumors with poor Miller-Payne response to treatment were less likely to experience changes in VAF of SNVs compared with those with good response (50% [7/14] vs 15% [4/24] had no changes observed at any timepoint, p = 0.029).

Conclusions: Serial molecular profiling identifies early therapy-induced genomic alterations, which may guide future selection of targeted therapies in breast cancer patients who progress after standard chemotherapy.

Clinical trial registration: ClinicalTrials.gov: NCT02790580 (first posted June 6, 2016).

Conflict of interest statement

Soo Chin Lee has received Honoraria and consulting fees from Astra Zeneca, Pfizer, Novartis, Eli Lilly, Roche, ACT Genomics, Eisai; research funding from Taiho, Eisai, Pfizer and ACT Genomics. Samuel GW Ow has received honoraria from AstraZeneca, Pfizer, Lilly, Roche and Novartis. Andrea Wong has received research funding from Otsuka Pharmaceuticals, and has advisory roles with Pfizer, Novartis and Eisai. Natalie Ngoi has received honoraria from AstraZeneca. Joline SJ Lim has consultancy with Pfizer, Novartis; research funding from Synthon. Goh Boon Cher has received honoraria from Novartis, Merck Serono, MSD; has consultancy with Adagene; research support and funding from Bayer, MSD, BMS, Adagene, and Taiho; stock interests in Gilead Sciences and Avantor. Yi-Hua Jan, Shu-Jen Chen and Kien Thiam Tan are under employment by ACT Genomics. Joan RE Choo, Matilda Xinwei Lee, Kritika Yadav, Siew Eng Lim, Ching Wan Chan, Mikael Hartman, Siau Wei Tang, Hon Lyn Tan, Wan Qin Chong, Ang Li En Yvonne, Gloria HJ Chan declared no conflicts of interest.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Consort diagram of sample disposition
Fig. 2
Fig. 2
Tumor SNV and CNA landscape at baseline. Commonly detected SNVs and CNAs by NGS are shown in decreasing order of prevalence. Pathogenic SNVs are labeled as driver mutations while SNVs of unknown significance are labeled as VUS. Copy number gain was defined as copy number between 4–7 while copy number amplification was defined as copy number ≥8. Copy number loss was defined as an observed copy number ≤1 where heterozygous loss (shallow deletion) was defined as observed copy number 1 while homozygous loss (deep deletion) was defined as observed copy number 0s. CNAs copy number alterations; NGS next-generation sequencing; SNV single nucleotide variants; VUS variants of uncertain significance
Fig. 3
Fig. 3
Genomic alterations in signaling molecules that interact with the VEGF angiogenesis pathway at baseline. Percentages of HER2−/HR+ and TNBC tumors (represented on the left and right of each module diagram, respectively) with alterations in signaling molecules that interact with the angiogenesis pathway are shown. Blue highlights the presence of inactivating mutations while red highlights the presence of activating mutations within that gene. The arrows describe whether the interactions are activating or inhibitory upon downstream signaling molecules. Sunitinib and bevacizumab have direct inhibitory effects on certain targets within the signaling cascades which are marked by the symbols. HER2− human epidermal growth factor receptor 2 negative; HR+ hormone receptor positive; TNBC triple-negative breast cancer; VEGF vascular endothelial growth factor
Fig. 4
Fig. 4
Timing of changes in CCF of SNVs. Patients with 3 serial tumor biopsies were analysed for timing of significant changes in VAF of SNVs [measured by the CCF slope (post-treatment CCF/baseline CCF)]. Patients were classified into 4 groups: no changes, early and sustained changes (those who experienced changes at 2 weeks which were persist at 8 weeks), transient changes (those who experienced changes at 2 weeks which subsequently normalized at 8 weeks) and those who experienced late changes alone. The blue bar highlights the patients with no changes, while orange bars highlight those with changes in VAF of SNVs. Of those who experienced changes in VAF of SNVs, majority had early and sustained changes. CCF cancer cell fraction; SNVs single nucleotide variants; VAF variant allele frequency
Fig. 5
Fig. 5
Change in VAF measured by CCF slope (post-treatment CCF/baseline CCF) of pathogenic PIK3CA mutations by treatment cohort. The change in VAF measured by the CCF slope (post-treatment CCF/baseline CCF) of each patient with pathogenic PIK3CA mutations are represented here. A slope of ≤0.8 indicates a decrease in VAF and ≥1.25 indicates an increase in VAF. Of the 20 patients with pathogenic PIK3CA mutations at baseline, 35% had sustained increase in VAF of pathogenic PIK3CA alterations. Significant rise in VAF of pathogenic PI3K mutations only occurred in HER2−/HR+ tumors. CCF cancer cell fraction; VAF variant allele frequency

References

    1. International Agency for Research on Cancer: Breast fact sheet: International Agency for Research on Cancer; 2021. .
    1. Sparano JA, Gray RJ, Makower DF, Pritchard KI, Albain KS, Hayes DF, et al. Adjuvant chemotherapy guided by a 21-gene expression assay in breast cancer. N Engl J Med. 2018;379(2):111–121. doi: 10.1056/NEJMoa1804710.
    1. André F, Ciruelos E, Rubovszky G, Campone M, Loibl S, Rugo HS, et al. Alpelisib for PIK3CA-mutated, hormone receptor-positive advanced breast cancer. N Engl J Med. 2019;380(20):1929–1940. doi: 10.1056/NEJMoa1813904.
    1. Schmid P, Adams S, Rugo HS, Schneeweiss A, Barrios CH, Iwata H, et al. Atezolizumab and Nab-paclitaxel in advanced triple-negative breast cancer. N Engl J Med. 2018;379(22):2108–2121. doi: 10.1056/NEJMoa1809615.
    1. Robson M, Im S-A, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523–533. doi: 10.1056/NEJMoa1706450.
    1. Litton JK, Rugo HS, Ettl J, Hurvitz SA, Gonçalves A, Lee K-H, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379(8):753–763. doi: 10.1056/NEJMoa1802905.
    1. Robinson DR, Wu YM, Vats P, Su F, Lonigro RJ, Cao X, et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet. 2013;45(12):1446–1451. doi: 10.1038/ng.2823.
    1. Chandarlapaty S, Chen D, He W, Sung P, Samoila A, You D, et al. Prevalence of ESR1 mutations in cell-free DNA and outcomes in metastatic breast cancer: a secondary analysis of the BOLERO-2 clinical trial. JAMA Oncol. 2016;2(10):1310–1315. doi: 10.1001/jamaoncol.2016.1279.
    1. Cancer Genome Atlas Network Comprehensive molecular portraits of human breast tumours. Nature. 2012;490(7418):61–70. doi: 10.1038/nature11412.
    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(1):5679. doi: 10.1038/s41467-020-19342-3.
    1. Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, Frederick AM, et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 2012;486(7403):405–409. doi: 10.1038/nature11154.
    1. Arnedos M, Vicier C, Loi S, Lefebvre C, Michiels S, Bonnefoi H, et al. Precision medicine for metastatic breast cancer—limitations and solutions. Nat Rev Clin Oncol. 2015;12(12):693–704. doi: 10.1038/nrclinonc.2015.123.
    1. Wong ALA, Sundar R, Wang T-T, Ng T-C, Zhang B, Tan S-H, et al. Phase Ib/II randomized, open-label study of doxorubicin and cyclophosphamide with or without low-dose, short-course sunitinib in the pre-operative treatment of breast cancer. Oncotarget. 2016;7(39):64089–64099. doi: 10.18632/oncotarget.11596.
    1. Yadav K, Lim J, Choo J, Ow SGW, Wong A, Lee M, et al. Immunohistochemistry study of tumor vascular normalization and anti-angiogenic effects of sunitinib versus bevacizumab prior to dose-dense doxorubicin/cyclophosphamide chemotherapy in HER2-negative breast cancer. Breast Cancer Res Treat. 2022;192(1):131–142. doi: 10.1007/s10549-021-06470-7.
    1. Wolff AC, Hammond MEH, Allison KH, Harvey BE, Mangu PB, Bartlett JMS, et al. Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer: American Society of Clinical Oncology/College of American Pathologists Clinical Practice Guideline Focused Update. J Clin Oncol. 2018;36(20):2105–2122. doi: 10.1200/JCO.2018.77.8738.
    1. Ogston KN, Miller ID, Payne S, Hutcheon AW, Sarkar TK, Smith I, et al. A new histological grading system to assess response of breast cancers to primary chemotherapy: prognostic significance and survival. Breast (Edinburgh, Scotland). 2003;12(5):320–327. doi: 10.1016/S0960-9776(03)00106-1.
    1. Loh JW, Guccione C, Di Clemente F, Riedlinger G, Ganesan S, Khiabanian H. All-FIT: allele-frequency-based imputation of tumor purity from high-depth sequencing data. Bioinformatics. 2019;36(7):2173–2180. doi: 10.1093/bioinformatics/btz865.
    1. Razavi P, Dickler MN, Shah PD, Toy W, Brown DN, Won HH, et al. Alterations in PTEN and ESR1 promote clinical resistance to alpelisib plus aromatase inhibitors. Nature Cancer. 2020;1(4):382–393. doi: 10.1038/s43018-020-0047-1.
    1. Seed G, Yuan W, Mateo J, Carreira S, Bertan C, Lambros M, et al. Gene copy number estimation from targeted next-generation sequencing of prostate cancer biopsies: analytic validation and clinical qualification. Clin Cancer Res. 2017;23(20):6070–6077. doi: 10.1158/1078-0432.CCR-17-0972.
    1. Sun JX, He Y, Sanford E, Montesion M, Frampton GM, Vignot S, et al. A computational approach to distinguish somatic vs. germline origin of genomic alterations from deep sequencing of cancer specimens without a matched normal. PLoS Comput Biol. 2018;14(2):e1005965. doi: 10.1371/journal.pcbi.1005965.
    1. Khiabanian H, Hirshfield KM, Goldfinger M, Bird S, Stein M, Aisner J, et al. Inference of germline mutational status and evaluation of loss of heterozygosity in high-depth, tumor-only sequencing data. JCO Precision Oncol. 2018;2018:1–15.
    1. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. VEGF receptor signalling—in control of vascular function. Nat Rev Mol Cell Biol. 2006;7(5):359–371. doi: 10.1038/nrm1911.
    1. Shah SP, Roth A, Goya R, Oloumi A, Ha G, Zhao Y, et al. The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature. 2012;486(7403):395–399. doi: 10.1038/nature10933.
    1. Balko JM, Giltnane JM, Wang K, Schwarz LJ, Young CD, Cook RS, et al. Molecular profiling of the residual disease of triple-negative breast cancers after neoadjuvant chemotherapy identifies actionable therapeutic targets. Cancer Discov. 2014;4(2):232–245. doi: 10.1158/-13-0286.
    1. Bertheau P, Plassa F, Espié M, Turpin E, de Roquancourt A, Marty M, et al. Effect of mutated TP53 on response of advanced breast cancers to high-dose chemotherapy. Lancet (London, England). 2002;360(9336):852–854. doi: 10.1016/S0140-6736(02)09969-5.
    1. Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N, Sander C. Emerging landscape of oncogenic signatures across human cancers. Nat Genet. 2013;45(10):1127–1133. doi: 10.1038/ng.2762.
    1. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC, et al. The landscape of cancer genes and mutational processes in breast cancer. Nature. 2012;486(7403):400–404. doi: 10.1038/nature11017.
    1. Linderholm BK, Hellborg H, Johansson U, Elmberger G, Skoog L, Lehtiö J, et al. Significantly higher levels of vascular endothelial growth factor (VEGF) and shorter survival times for patients with primary operable triple-negative breast cancer. Ann Oncol. 2009;20(10):1639–1646. doi: 10.1093/annonc/mdp062.
    1. Nahleh Z, Botrus G, Dwivedi A, Jennings M, Nagy S, Tfayli A. Bevacizumab in the neoadjuvant treatment of human epidermal growth factor receptor 2-negative breast cancer: a meta-analysis of randomized controlled trials. Mol Clin Oncol. 2019;10(3):357–365.
    1. Osborne CK, Schiff R. Mechanisms of endocrine resistance in breast cancer. Annu Rev Med. 2011;62(1):233–247. doi: 10.1146/annurev-med-070909-182917.
    1. Drago JZ, Formisano L, Juric D, Niemierko A, Servetto A, Wander SA, et al. FGFR1 amplification mediates endocrine resistance but retains TORC sensitivity in metastatic hormone receptor-positive (HR+) breast cancer. Clin Cancer Res. 2019;25(21):6443–6451. doi: 10.1158/1078-0432.CCR-19-0138.
    1. Formisano L, Stauffer KM, Young CD, Bhola NE, Guerrero-Zotano AL, Jansen VM, et al. Association of FGFR1 with ERα Maintains Ligand-Independent ER Transcription and Mediates Resistance to Estrogen Deprivation in ER(+) Breast Cancer. Clin Cancer Res. 2017;23(20):6138–6150. doi: 10.1158/1078-0432.CCR-17-1232.
    1. Weigel MT, Banerjee S, Arnedos M, Salter J, A'Hern R, Dowsett M, et al. Enhanced expression of the PDGFR/Abl signaling pathway in aromatase inhibitor-resistant breast cancer. Ann Oncol. 2013;24(1):126–133. doi: 10.1093/annonc/mds240.
    1. Mayer IA, Haley BB, Abramson VG, Brufsky A, Rexer B, Stringer-Reasor E, et al. A phase Ib trial of fulvestrant + CDK4/6 inhibitor (CDK4/6i) palbociclib + pan-FGFR tyrosine kinase inhibitor (TKI) erdafitinib in FGFR-amplified/ ER+/ HER2-negative metastatic breast cancer (MBC) Cancer Res. 2021;81(4 Supplement):PD1-03-PD1.
    1. Hui R, Pearson A, Cortes J, Campbell C, Poirot C, Azim HA, et al. Lucitanib for the treatment of HR+/HER2− metastatic breast cancer: results from the multicohort phase II FINESSE study. Clin Cancer Res. 2020;26(2):354–363. doi: 10.1158/1078-0432.CCR-19-1164.
    1. Mavrommati I, Johnson F, Echeverria GV, Natrajan R. Subclonal heterogeneity and evolution in breast cancer. NPJ Breast Cancer. 2021;7(1):155. doi: 10.1038/s41523-021-00363-0.
    1. Bedard PL, Hansen AR, Ratain MJ, Siu LL. Tumour heterogeneity in the clinic. Nature. 2013;501(7467):355–364. doi: 10.1038/nature12627.
    1. Hernandez L, Wilkerson PM, Lambros MB, Campion-Flora A, Rodrigues DN, Gauthier A, et al. Genomic and mutational profiling of ductal carcinomas in situ and matched adjacent invasive breast cancers reveals intra-tumour genetic heterogeneity and clonal selection. J Pathol. 2012;227(1):42–52. doi: 10.1002/path.3990.
    1. Nik-Zainal S, Van Loo P, Wedge DC, Alexandrov LB, Greenman CD, Lau KW, et al. The life history of 21 breast cancers. Cell. 2012;149(5):994–1007. doi: 10.1016/j.cell.2012.04.023.
    1. Yates LR, Gerstung M, Knappskog S, Desmedt C, Gundem G, Van Loo P, et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med. 2015;21(7):751–759. doi: 10.1038/nm.3886.
    1. Zhang L, Feizi N, Chi C, Hu P. Association analysis of somatic copy number alteration burden with breast cancer survival. Front Genet. 2018;9:421. doi: 10.3389/fgene.2018.00421.
    1. Barry WT, Kernagis DN, Dressman HK, Griffis RJ, Hunter JD, Olson JA, et al. Intratumor heterogeneity and precision of microarray-based predictors of breast cancer biology and clinical outcome. J Clin Oncol. 2010;28(13):2198–2206. doi: 10.1200/JCO.2009.26.7245.
    1. Pestrin M, Bessi S, Galardi F, Truglia M, Biggeri A, Biagioni C, et al. Correlation of HER2 status between primary tumors and corresponding circulating tumor cells in advanced breast cancer patients. Breast Cancer Res Treat. 2009;118(3):523–530. doi: 10.1007/s10549-009-0461-7.

Source: PubMed

3
Suscribir