Skip to main content

Advertisement

Log in

Circulating tumor cells, circulating tumor DNA, and disease characteristics in young women with metastatic breast cancer

  • Preclinical study
  • Published:
Breast Cancer Research and Treatment Aims and scope Submit manuscript

Abstract

Background

Clinical and genomic data from patients with early-stage breast cancer suggest more aggressive disease in premenopausal women. However, the association between age, disease course, and molecular profile from liquid biopsy in metastatic breast cancer (MBC) is not well characterized.

Methods

Patients were classified as premenopausal (< 45 years), perimenopausal (45–55 years), or postmenopausal (> 55 years). Cohort 1 consisted of patients with MBC who consented for prospective serial evaluation of circulating tumor cells (CTCs) using CellSearch™. Cohort 2 included patients who, as part of routine care, had circulating tumor DNA (ctDNA) sequenced by the Guardant360™ assay. Clinicopathologic data were collected from retrospective review to compare disease features between premenopausal and postmenopausal women.

Results

Premenopausal women represented 26% of 138 patients in Cohort 1 and 21% of 253 patients in Cohort 2. In Cohort 1, younger patients had a shorter time to metastases and a higher prevalence of lung and brain metastases. Overall, there were similar rates of ≥ 5 CTCs/7.5 mL, HER2 + CTC expression, and CTC clusters between pre- and postmenopausal women. However, for those with triple negative breast cancer, premenopausal women had a higher proportion of ≥ 5 CTCs/7.5 mL. In Cohort 2, premenopausal women had a higher incidence of FGFR1 (OR 2.75, p = 0.022) and CCND2 (OR 6.91, p = 0.024) alterations. There was no difference in the ctDNA mutant allele frequency or the number of detected alterations between these age groups.

Conclusions

Our data reveal that premenopausal women diagnosed with MBC have unique clinical, pathologic, and molecular features when compared to their postmenopausal counterparts. Our results highlight FGFR1 inhibitors as potential therapeutics of particular interest among premenopausal women.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

Data availability

The datasets generated and/or analyzed during the current study are not publicly available due ongoing data collection but are available from the corresponding author on reasonable request.

References

  1. Fidler MM et al (2017) Cancer incidence and mortality among young adults aged 20–39 years worldwide in 2012: a population-based study. Lancet Oncol 18(12):1579–1589

    Article  Google Scholar 

  2. Siegel RL, Miller KD, Jemal A (2020) Cancer statistics, 2020. CA Cancer J Clin 70(1):7–30

    Article  Google Scholar 

  3. Liao S et al (2015) The molecular landscape of premenopausal breast cancer. Breast Cancer Res 17:104

    Article  Google Scholar 

  4. Fu J et al (2018) How young is too young in breast cancer?-Young breast cancer is not a unique biological subtype. Clin Breast Cancer 18(1):e25–e39

    Article  Google Scholar 

  5. Keegan TH et al (2012) Occurrence of breast cancer subtypes in adolescent and young adult women. Breast Cancer Res 14(2):R55

    Article  CAS  Google Scholar 

  6. O’Brien KM et al (2015) Risk factors for young-onset invasive and in situ breast cancer. Cancer Causes Control 26(12):1771–1778

    Article  Google Scholar 

  7. Im S-A et al (2019) Overall survival with ribociclib plus endocrine therapy in breast cancer. N Engl J Med 381(4):307–316

    Article  CAS  Google Scholar 

  8. Cristofanilli M et al (2004) Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med 351(8):781–791

    Article  CAS  Google Scholar 

  9. Hrebien S et al (2019) Early ctDNA dynamics as a surrogate for progression-free survival in advanced breast cancer in the BEECH trial. Ann Oncol 30(6):945–952

    Article  CAS  Google Scholar 

  10. Bidard F-C et al (2020) Efficacy of circulating tumor cell count-driven vs clinician-driven first-line therapy choice in hormone receptor-positive, ERBB2-negative metastatic breast cancer: the STIC CTC randomized clinical trial. JAMA Oncol. https://doi.org/10.1001/jamaoncol.2020.5660

    Article  Google Scholar 

  11. Hayashi N et al (2012) Prognostic value of HER2-positive circulating tumor cells in patients with metastatic breast cancer. Int J Clin Oncol 17(2):96–104

    Article  Google Scholar 

  12. Amintas S et al (2020) Circulating tumor cell clusters: united we stand divided we fall. Int J Mol Sci 21(7):2653

    Article  CAS  Google Scholar 

  13. Shah AN et al (2019) Abstract P3-01-19: HER2-positive circulating tumor cells (CTCs) in advanced breast cancer (BC): a feature independent of BC subtype. Cancer Res 79(4 Supplement):P3-01–19

    Google Scholar 

  14. Higgins MJ et al (2012) Detection of tumor PIK3CA status in metastatic breast cancer using peripheral blood. Clin Cancer Res 18(12):3462–3469

    Article  CAS  Google Scholar 

  15. Anders CK et al (2008) Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol 26(20):3324–3330

    Article  Google Scholar 

  16. Kolečková M et al (2017) Age-associated prognostic and predictive biomarkers in patients with breast cancer. Oncol Lett 13(6):4201–4207

    Article  Google Scholar 

  17. Gnerlich JL et al (2009) Elevated breast cancer mortality in women younger than age 40 years compared with older women is attributed to poorer survival in early-stage disease. J Am Coll Surg 208(3):341–347

    Article  Google Scholar 

  18. Azim HA Jr et al (2015) Genomic aberrations in young and elderly breast cancer patients. BMC Med 13:266–266

    Article  Google Scholar 

  19. Cristofanilli M et al (2019) The clinical use of circulating tumor cells (CTCs) enumeration for staging of metastatic breast cancer (MBC): international expert consensus paper. Crit Rev Oncol Hematol 134:39–45

    Article  Google Scholar 

  20. Gao J et al (2013) Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6(269):pl1

    Article  Google Scholar 

  21. Tolaney SM et al (2019) Randomized phase II study of eribulin mesylate (E) with or without pembrolizumab (P) for hormone receptor-positive (HR +) metastatic breast cancer (MBC). J Clin Oncol 37(15_suppl):1004

    Article  Google Scholar 

  22. Razavi P et al (2018) The genomic landscape of endocrine-resistant advanced breast cancers. Cancer Cell 34(3):427-438.e6

    Article  CAS  Google Scholar 

  23. Turner N et al (2020) Abstract GS3-06: results from the plasmaMATCH trial: a multiple parallel cohort, multi-centre clinical trial of circulating tumour DNA testing to direct targeted therapies in patients with advanced breast cancer (CRUK/15/010). Cancer Res 80(4 Supplement):GS3-06

    Google Scholar 

  24. Shah AN et al (2020) Hormone receptor-positive/human epidermal growth receptor 2-negative metastatic breast cancer in young women: emerging data in the era of molecularly targeted agents. Oncologist 25(6):e900–e908

    Article  Google Scholar 

  25. Bardia A et al (2019) Abstract PD2-08: Ribociclib with endocrine therapy for premenopausal patients with hormone receptor-positive, HER2-negative advanced breast cancer: biomarker analyses from the phase III randomized MONALEESA-7 trial. Cancer Res 79(4 Supplement):PD2-08

    Google Scholar 

  26. Leary B et al (2018) The genetic landscape and clonal evolution of breast cancer resistance to Palbociclib plus Fulvestrant in the PALOMA-3 trial. Cancer Discov 8(11):1390

    Article  Google Scholar 

  27. Dawood S et al (2008) Circulating tumor cells in metastatic breast cancer. Cancer 113(9):2422–2430

    Article  Google Scholar 

  28. Rack B et al (2014) Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J Natl Cancer Inst 106(5):dju066

    Article  Google Scholar 

  29. Turner N et al (2010) FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer. Cancer Res 70(5):2085–2094

    Article  CAS  Google Scholar 

  30. André F et al (2014) Comparative genomic hybridisation array and DNA sequencing to direct treatment of metastatic breast cancer: a multicentre, prospective trial (SAFIR01/UNICANCER). Lancet Oncol 15(3):267–274

    Article  Google Scholar 

  31. Perez-Garcia J et al (2018) Targeting FGFR pathway in breast cancer. Breast 37:126–133

    Article  CAS  Google Scholar 

  32. Formisano L et al (2019) Aberrant FGFR signaling mediates resistance to CDK4/6 inhibitors in ER+ breast cancer. Nat Commun 10(1):1373

    Article  Google Scholar 

  33. Yamamoto-Ibusuki M, Arnedos M, André F (2015) Targeted therapies for ER+/HER2− metastatic breast cancer. BMC Med 13(1):137

    Article  Google Scholar 

  34. Wang S, Ding Z (2017) Fibroblast growth factor receptors in breast cancer. Tumor Biol 39(5):1010428317698370

    Google Scholar 

  35. Turner NC et al (2020) Abstract OT2-07-01: a phase 2 study of futibatinib (TAS-120) in metastatic breast cancers harboring fibroblast growth factor receptor (FGFR) amplifications (FOENIX-MBC2). Cancer Res 80(4 Supplement):OT2-07–01

    Google Scholar 

  36. Boyle P (2012) Triple-negative breast cancer: epidemiological considerations and recommendations. Ann Oncol 23(Suppl 6):vi7–vi12

    Article  Google Scholar 

Download references

Funding

Lynn Sage Research Foundation.

Author information

Authors and Affiliations

Authors

Contributions

ANS, KC, LG, and MC involved in concept/design of work; ANS, KC, LG, AAD, QZ, and SJ collected the data; ANS, KC, and LG performed data analysis/interpretation; ANS and KC wrote the manuscript; and all involved in critical revision of the article and final approval of the version to be published.

Corresponding author

Correspondence to Ami N. Shah.

Ethics declarations

Conflict of interest

Ami N. Shah: Honorarium—Taiho, Daiichi-Sankyo, Pfizer; Advisory Board—d AbbVie; Lorenzo Gerratana: Advisory board—Lilly; Amir Behdad: Fees for Non-CME Services Received Directly from Commercial Interest or their Agents (e.g., speakers' bureaus); Author; Pfizer, Bayer, Foundation Medicine; Massimo Cristofanilli: Advisory Board—G1 Therapeutics, Cytodyn, Sermonix, LIlly, Foundation Medicine. Fees for Non-CME Services Received Directly from Commercial Interest or their Agents—Pfizer. Contracted Research—G1 Therapeutics. Others: none.

Ethical approval

This protocol was approved by the institutional review board at the Robert H. Lurie Comprehensive Cancer Center of Northwestern University (STU00203283 and STU00214133).

Consent to participate

Patients provided written consent to participate in part 1.

Informed consent

Requirement for informed consent was waived for part 2 for this retrospective review of de-identified patient data.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shah, A.N., Carroll, K.J., Gerratana, L. et al. Circulating tumor cells, circulating tumor DNA, and disease characteristics in young women with metastatic breast cancer. Breast Cancer Res Treat 187, 397–405 (2021). https://doi.org/10.1007/s10549-021-06236-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10549-021-06236-1

Keywords

Navigation