Circulating-free DNA Mutation Associated with Response of Targeted Therapy in Human Epidermal Growth Factor Receptor 2-positive Metastatic Breast Cancer

Qing Ye, Fan Qi, Li Bian, Shao-Hua Zhang, Tao Wang, Ze-Fei Jiang, Qing Ye, Fan Qi, Li Bian, Shao-Hua Zhang, Tao Wang, Ze-Fei Jiang

Abstract

Background: The addition of anti-human epidermal growth factor receptor 2 (HER2)-targeted drugs, such as trastuzumab, lapatinib, and trastuzumab emtansine (T-DM1), to chemotherapy significantly improved prognosis of HER2-positive breast cancer patients. However, it was confused that metastatic patients vary in the response of targeted drug. Therefore, methods of accurately predicting drug response were really needed. To overcome the spatial and temporal limitations of biopsies, we aimed to develop a more sensitive and less invasive method of detecting mutations associated with anti-HER2 therapeutic response through circulating-free DNA (cfDNA).

Methods: From March 6, 2014 to December 10, 2014, 24 plasma samples from 20 patients with HER2-positive metastatic breast cancer who received systemic therapy were eligible. We used a panel for detection of hot-spot mutations from 50 oncogenes and tumor suppressor genes, and then used targeted next-generation sequencing (NGS) to identify somatic mutation of these samples in those 50 genes. Samples taken before their first trastuzumab administration and subsequently proven with clinical benefit were grouped into sensitive group. The others were collected after disease progression of the trastuzumab-based therapy and were grouped into the resistant group.

Results: A total of 486 single-nucleotide variants from 46 genes were detected. Of these 46 genes, phosphatidylinositol 4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), proto-oncogene c-Kit (KIT), and tumor protein p53 (TP53) were the most common mutated genes. Seven genes, including epidermal growth factor receptor (EGFR), G protein subunit alpha S (GNAS), HRas proto-oncogene (HRAS), mutL homolog 1 (MLH1), cadherin 1 (CDH1), neuroblastoma RAS viral oncogene homolog (NRAS), and NOTCH1, that only occurred m utations in the resistant group were associated with the resistance of targeted therapy. In addition, we detected a HER2 S855I mutation in two patients who had persistent benefits from anti-HER2 therapy.

Conclusion: Targeted NGS of cfDNA has potential clinical utility to detect biomarkers from HER2-targeted therapies.

Conflict of interest statement

There are no conflicts of interest.

Figures

Figure 1
Figure 1
The constitution ratio of 486 somatic mutations detected from 24 plasma samples. *Genes that owe less than 10 single-nucleotide variants (2%), those genes contain fibroblast growth factor receptor 1 (FGFR1), G protein subunit alpha q (GNAQ), RB transcriptional corepressor 1 (RB1), ABL proto-oncogene 1 (ABL1), B-Raf proto-oncogene (BRAF), fibroblast growth factor receptor 3 (FGFR3), guanine nucleotide-binding protein alpha stimulating (GNAS), HRas proto-oncogene (HRAS), fms-related tyrosine kinase 3 (FLT3), AKT serine/threonine kinase 1 (AKT), ret proto-oncogene (RET), cadherin 1 (CDH1), cyclin-dependent kinase inhibitor 2A (CDKN2A), ERBB2, enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2), isocitrate dehydrogenase 1 (IDH1), isocitrate dehydrogenase 2 (IDH2), MET proto-oncogene (MET), mutL homolog 1 (MLH1), neuroblastoma RAS viral oncogene homolog (NRAS), catenin beta 1 (CTNNB1), janus kinase 2 (JAK2), NOTCH1, protein tyrosine phosphatase nonreceptor type 11 (PTPN11), janus kinase 3 (JAK3), and MPL proto-oncogene (MPL).
Figure 2
Figure 2
Mutational prevalence of 24 human epidermal growth factor receptor 2-positive breast cancer patients’ plasma samples from 46 genes.
Figure 3
Figure 3
The landscape of the identified mutations in seven anti-human epidermal growth factor receptor 2-targeted therapy-resistant genes. The number shown in the box meant the mean allele frequency of the identified mutations in that gene. R: Resistant group. EGFR: Epidermal growth factor receptor; GNAS: Guanine nucleotide-binding protein alpha stimulating; HRAS: HRas proto-oncogene; MLH1: mutL homolog 1; CDH1: Cadherin 1; NRAS: Neuroblastoma RAS viral oncogene homolog.
Figure 4
Figure 4
The gene map of human epidermal growth factor receptor 2 p.S855I mutation identified in circulating-free DNA.
Figure 5
Figure 5
The clinical timeline for two metastatic breast cancer patients with the HER2 p.S855I mutation in cfDNA. (a and b) The clinical treatment history and the dramatic effect of containing anti-HER2 agents’ regimen in the Patient R2 and S2 with HER2 activating mutation, respectively. Values within each circle represent mutation frequency. Positron emission tomography/computed tomography scan showed the clinical complete response in the right breast tumor of patient S8 after eight cycles’ therapy. *TPH: Docetaxel, carboplatin, trastuzumab; RT: Radiotherapy; ANA: Anastrozole; G: Goserelin. †LX: Lapatinib, capecitabine. ‡TXH: Docetaxel, capecitabine, trastuzumab. §The pathological images showed the pathological complete response in the breast tumor of patient S8 after eight cycles’ therapy. HER2: Human epidermal growth factor receptor 2; cfDNA: Circulating-free DNA.

References

    1. Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, et al. Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 2013;31:3997–4013. doi: 10.1200/JCO.2013.50.9984.
    1. Ross JS, Slodkowska EA, Symmans WF, Pusztai L, Ravdin PM, Hortobagyi GN. The HER-2 receptor and breast cancer: Ten years of targeted anti-HER-2 therapy and personalized medicine. Oncologist. 2009;14:320–68. doi: 10.1634/theoncologist.2008-0230.
    1. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 2005;353:1659–72. doi: 10.1056/NEJMoa052306.
    1. Marty M, Cognetti F, Maraninchi D, Snyder R, Mauriac L, Tubiana-Hulin M, et al. Randomized phase II trial of the efficacy and safety of trastuzumab combined with docetaxel in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer administered as first-line treatment: The M77001 study group. J Clin Oncol. 2005;23:4265–74. doi: 10.1200/JCO.2005.04.173.
    1. Gianni L, Eiermann W, Semiglazov V, Manikhas A, Lluch A, Tjulandin S, et al. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): A randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet. 2010;375:377–84. doi: 10.1016/S0140-6736(09)61964-4.
    1. Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, Wallis JW, et al. Whole-genome analysis informs breast cancer response to aromatase inhibition. Nature. 2012;486:353–60. doi: 10.1038/nature11143.
    1. Overman MJ, Modak J, Kopetz S, Murthy R, Yao JC, Hicks ME, et al. Use of research biopsies in clinical trials: Are risks and benefits adequately discussed? J Clin Oncol. 2013;31:17–22. doi: 10.1200/JCO.2012.43.1718.
    1. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366:883–92. doi: 10.1056/NEJMoa1113205.
    1. Garcia-Murillas I, Schiavon G, Weigelt B, Ng C, Hrebien S, Cutts RJ, et al. Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Sci Transl Med. 2015;7:302ra133. doi: 10.1126/scitranslmed.aab0021.
    1. Diaz LA, Jr, Bardelli A. Liquid biopsies: Genotyping circulating tumor DNA. J Clin Oncol. 2014;32:579–86. doi: 10.1200/JCO.2012.45.2011.
    1. Diehl F, Schmidt K, Choti MA, Romans K, Goodman S, Li M, et al. Circulating mutant DNA to assess tumor dynamics. Nat Med. 2008;14:985–90. doi: 10.1038/nm.1789.
    1. Leary RJ, Kinde I, Diehl F, Schmidt K, Clouser C, Duncan C, et al. Development of personalized tumor biomarkers using massively parallel sequencing. Sci Transl Med. 2010;2:20ra14. doi: 10.1126/scitranslmed.3000702.
    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. Shaw JA, Page K, Blighe K, Hava N, Guttery D, Ward B, et al. Genomic analysis of circulating cell-free DNA infers breast cancer dormancy. Genome Res. 2012;22:220–31. doi: 10.1101/gr. 123497.111.
    1. Forshew T, Murtaza M, Parkinson C, Gale D, Tsui DW, Kaper F, et al. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci Transl Med. 2012;4:136ra68. doi: 10.1126/scitranslmed.3003726.
    1. Frenel JS, Carreira S, Goodall J, Roda D, Perez-Lopez R, Tunariu N, et al. Serial next-generation sequencing of circulating cell-free DNA evaluating tumor clone response to molecularly targeted drug administration. Clin Cancer Res. 2015;21:4586–96. doi: 10.1158/1078-0432.CCR-15-0584.
    1. Huang R, Ding P, Yang F. Clinicopathological significance and potential drug target of CDH1 in breast cancer: A meta-analysis and literature review. Drug Des Devel Ther. 2015;9:5277–85. doi: 10.2147/DDDT.S86929.
    1. Thiele S, de Sanctis L, Werner R, Grötzinger J, Aydin C, Jüppner H, et al. Functional characterization of GNAS mutations found in patients with pseudohypoparathyroidism type Ic defines a new subgroup of pseudohypoparathyroidism affecting selectively Gsa-receptor interaction. Hum Mutat. 2011;32:653–60. doi: 10.1002/humu.21489.
    1. Linglart A, Mahon MJ, Kerachian MA, Berlach DM, Hendy GN, Jüppner H, et al. Coding GNAS mutations leading to hormone resistance impair in vitro agonist- and cholera toxin-induced adenosine cyclic 3’,5’-monophosphate formation mediated by human XLalphas. Endocrinology. 2006;147:2253–62. doi: 10.1210/en.2005-1487.
    1. Harkness EF, Barrow E, Newton K, Green K, Clancy T, Lalloo F, et al. Lynch syndrome caused by MLH1 mutations is associated with an increased risk of breast cancer: A cohort study. J Med Genet. 2015;52:553–6. doi: 10.1136/jmedgenet-2015-103216.
    1. Groth C, Fortini ME. Therapeutic approaches to modulating Notch signaling: Current challenges and future prospects. Semin Cell Dev Biol. 2012;23:465–72. doi: 10.1016/j.semcdb.2012.01.016.
    1. Rothé F, Laes JF, Lambrechts D, Smeets D, Vincent D, Maetens M, et al. Plasma circulating tumor DNA as an alternative to metastatic biopsies for mutational analysis in breast cancer. Ann Oncol. 2014;25:1959–65. doi: 10.1093/annonc/mdu288.
    1. Schmid P, Pinder SE, Wheatley D, Macaskill J, Zammit C, Hu J, et al. Phase II randomized preoperative window-of-opportunity study of the pi3k inhibitor pictilisib plus anastrozole compared with anastrozole alone in patients with estrogen receptor-positive breast cancer. J Clin Oncol. 2016;34:1987–94. doi: 10.1200/JCO.2015.63.9179.
    1. Krop IE, Mayer IA, Ganju V, Dickler M, Johnston S, Morales S, et al. Pictilisib for oestrogen receptor-positive, aromatase inhibitor-resistant, advanced or metastatic breast cancer (FERGI): A randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol. 2016;17:811–21. doi: 10.1016/S1470-2045(16)00106-6.
    1. Vuylsteke P, Huizing M, Petrakova K, Roylance R, Laing R, Chan S, et al. Pictilisib PI3Kinase inhibitor (a phosphatidylinositol 3-kinase [PI3K] inhibitor) plus paclitaxel for the treatment of hormone receptor-positive, HER2-negative, locally recurrent, or metastatic breast cancer: Interim analysis of the multicentre, placebo-controlled, phase II randomised PEGGY study. Ann Oncol. 2016;27:2059–66. doi: 10.1093/annonc/mdw320.
    1. Ritter CA, Perez-Torres M, Rinehart C, Guix M, Dugger T, Engelman JA, et al. Human breast cancer cells selected for resistance to trastuzumab in vivo overexpress epidermal growth factor receptor and ErbB ligands and remain dependent on the ErbB receptor network. Clin Cancer Res. 2007;13:4909–19. doi: 10.1158/1078-0432.CCR-07-0701.
    1. Ni J, Zhang L. Evaluation of three small molecular drugs for targeted therapy to treat nonsmall cell lung cancer. Chin Med J. 2016;129:332–40. doi: 10.4103/0366-6999.174484.
    1. Baker AT, Zlobin A, Osipo C. Notch-EGFR/HER2 bidirectional crosstalk in breast cancer. Front Oncol. 2014;4:360. doi: 10.3389/fonc.2014.00360.
    1. Chu BF, Qin YY, Zhang SL, Quan ZW, Zhang MD, Bi JW. Downregulation of notch-regulated ankyrin repeat protein exerts antitumor activities against growth of thyroid cancer. Chin Med J. 2016;129:1544–52. doi: 10.4103/0366-6999.184465.
    1. Osipo C, Patel P, Rizzo P, Clementz AG, Hao L, Golde TE, et al. ErbB-2 inhibition activates Notch-1 and sensitizes breast cancer cells to a gamma-secretase inhibitor. Oncogene. 2008;27:5019–32. doi: 10.1038/onc.
    1. Abravanel DL, Belka GK, Pan TC, Pant DK, Collins MA, Sterner CJ, et al. Notch promotes recurrence of dormant tumor cells following HER2/neu-targeted therapy. J Clin Invest. 2015;125:2484–96. doi: 10.1172/JCI74883.
    1. Han M, Deng HY, Jiang R. Effect of trastuzumab on notch-1 signaling pathway in breast cancer SK-BR3 cells. Chin J Cancer Res. 2012;24:213–9. doi: 10.1007/s11670-012-0213-9.
    1. Pandya K, Meeke K, Clementz AG, Rogowski A, Roberts J, Miele L, et al. Targeting both Notch and ErbB-2 signalling pathways is required for prevention of ErbB-2-positive breast tumour recurrence. Br J Cancer. 2011;105:796–806. doi: 10.1038/bjc.
    1. Bose R, Kavuri SM, Searleman AC, Shen W, Shen D, Koboldt DC, et al. Activating HER2 mutations in HER2 gene amplification negative breast cancer. Cancer Discov. 2013;3:224–37. doi: 10.1158/2159-8290.
    1. Park YH, Shin HT, Jung HH, Choi YL, Ahn T, Park WY, et al. Role of HER2 mutations in refractory metastatic breast cancers: Targeted sequencing results in patients with refractory breast cancer. Oncotarget. 2015;6:32027–38. doi: 10.18632/oncotarget.5184.
    1. Liu S, Wang H, Zhang L, Tang C, Jones L, Ye H, et al. Rapid detection of genetic mutations in individual breast cancer patients by next-generation DNA sequencing. Hum Genomics. 2015;9:2. doi: 10.1186/s40246-015-0024-4.
    1. Schwaederle M, Husain H, Fanta PT, Piccioni DE, Kesari S, Schwab RB, et al. Detection rate of actionable mutations in diverse cancers using a biopsy-free (blood) circulating tumor cell DNA assay. Oncotarget. 2016;7:9707–17. doi: 10.18632/oncotarget.7110.

Source: PubMed

3
Subscribe