Evidence-based guidelines recommend surveillance after treatment of localized breast cancer including history, physical examination, and annual mammography (1,2). Laboratory tests including circulating tumor markers and imaging studies beyond mammography are not recommended in asymptomatic patients, although these recommendations are based on clinical trials done in an era when diagnostic, imaging and therapeutic options were limited (3). Recently, the U.S. Food and Drug Administration (FDA) granted regulatory approval for apalutamide in the treatment of nonmetastatic castration-resistant prostate cancer based on the endpoint of metastasis-free survival (4), and use of an integral biomarker (serum prostate-specific antigen [PSA]) to select men at high risk for developing metastasis and lacking distant disease based on standard diagnostic imaging (5). This paradigm provides a tenable model for evaluating a similar strategy in nonmetastatic breast cancer.
Application of such a strategy requires the use of biomarkers that exhibit and a high degree of analytic and clinical validity, which would be required to ultimately establish clinical utility—a biomarker-directed treatment change that results in clinical benefit (6). Similar to PSA in prostate cancer, circulating MUC-1 antigen assays (eg, CA15-3, CA 27.29) have been evaluated for surveillance in breast cancer, although the trials were underpowered, and survival was not improved for those who underwent surveillance (7–10). Other blood-based biomarkers offer potential for greater sensitivity and specificity in identifying individuals at high risk for recurrence, including circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA), microRNA, noncoding RNAs, and tumor-educated platelets (11). The only FDA-cleared, analytically validated assay is one that allows detection and enumeration of CTCs in metastatic breast, colorectal, and prostate cancer (CellSearch System, Menarini Silicon Biosystems, Bologna, Italy) (12,13). The assay involves an automated system for immunomagnetic capturing of cells that express epithelium-specific cell adhesion molecule (EpCAM) on the cell surface. CTCs are detectable in 65% of patients with metastatic breast cancer, and a CTC count of greater than 5 cells per 7.5 mL of blood is associated with statistically significant inferior progression-free survival and overall survival (12), providing evidence for clinical validity. Other studies have shown that CTCs are detectable using the same assay in about 20–25% of patients with localized nonmetastatic breast cancer. CTCs also provide independent prognostic information (whether obtained before or after surgery, including after neoadjuvant or adjuvant chemotherapy) providing additional evidence for clinical validity (14–16).
In this issue of JNCI, Trapp et al. report the association between CTCs detected 2 years after completion of adjuvant chemotherapy in 1087 patients with stage II–III breast cancer enrolled in the phase III SUCCESS A trial (17) in which CTCs were found to be prognostic at diagnosis (15). CTCs were detected in 18.2% of patients (median= 1 cell, range = 1–99 cells per 7.5 mL blood) at 2 years, and was associated with a 3.9-fold increased risk of death and a 2.3-fold higher recurrence risk in multivariable models that included clinicopathologic features and CTC status at baseline; sensitivity analysis showed this effect only in HER2-negative disease. Another report from this same study found that among 206 subjects enrolled in the SUCCESS study with follow-up information and known CTC status at 5 years, 7.8% were CTC-positive at 5 years (median = 1 cell, range = 1–53 cells per 7.5 mL blood), and was associated with a six-fold increase in recurrence (18). Finally, in a separate study including 353 patients with hormone receptor-positive, HER2-negative, stage II–III breast cancer treated with adjuvant chemotherapy and endocrine therapy, CTCs were detectable after a median follow-up of 5.1 years in 5.1% (median = 1 cell, range = 1–15 cells per 7.5 mL blood), and was associated with a 13.1-fold increase in recurrence risk in multivariable analysis adjusted for other prognostic covariates; the median time between the positive CTC assay and recurrence was 2.8 years (19). Because imaging was not performed in any of these trials at the time of the positive CTC assay, it is currently unknown what proportion of CTC-positive subjects would be found to have clinical evidence of asymptomatic metastatic disease.
Despite convincing evidence of the analytic and clinical validity of the CTC assay, clinical utility has not been established. Only one study that was specifically designed to determine clinical utility failed to show improved survival in metastatic breast cancer when systemic chemotherapy was changed in patients with persistently high CTC count (>5 CTCs per 7.5 mL blood) after 3 weeks of chemotherapy (13). No studies have been specifically designed to evaluate whether CTCs or other novel biomarkers may be used for surveillance in order to select individuals with nonmetastatic breast cancer after local therapy at high risk of clinical recurrence (20). Moreover, methodology now exists to characterize CTCs rather than simply enumerate them, as well as to identify and quantify somatic tumor mutations in blood or urine (11). This could yield insights into the mechanism underlying disease recurrence such as tumor dormancy (21) and selection of treatment approaches. These strategies may be particularly promising for preventing early recurrence within 5 years of diagnosis typically associated with hormone receptor-negative breast cancer (22), or later recurrence up to 15 years or longer after diagnosis in hormone receptor-positive breast cancer (23). The availability of more effective therapies for metastatic breast cancer, such as immune checkpoint blockade for triple-negative disease (24), and CDK 4/6 inhibitors (25) or novel oral selective estrogen receptor downregulators (26) for hormone receptor-positive disease, offers potential for early intervention that could ultimately delay or even prevent metastasis.
The time has never been better for a reappraisal of surveillance in early breast cancer in a prospective clinical trial, with one potential design schematically depicted in Figure 1. Key elements of the trial include: 1) selection of patients with nonmetastatic breast cancer at high clinical risk of recurrence based on classical clinicopathologic features (27); 2) further enrichment of the high-risk population based on an integral biomarker such as the CTC assay, with imaging to exclude distant metastasis in CTC-positive patients; and 3) testing interventions that may delay or prevent distant metastasis in the CTC-positive cohort in whom imaging has excluded distant metastasis. Such a trial should include a cross-platform comparison of the integral CTC assay used for treatment selection with other CTC assays, ctDNA assays that are both prognostic and potentially predictive response to specific therapies (eg, ESR1 mutations and response to SERDs) (28–30), and serial assays to screen high-risk populations at multiple timepoints after diagnosis and before recurrence, or as an intermediate pharmacodynamic biomarker of drug response. We have never been better positioned to launch such a trial—until we do, the clinical utility of CTCs and other liquid biopsy biomarkers remains unproven in this setting.
Notes
Affiliations of authors: Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY (JAS); University of Utah, Huntsman Cancer Institute, Salt Lake City, UT (NLH).
The authors report no disclosures.
References
- 1. Khatcheressian JL, Hurley P, Bantug E, et al. Breast cancer follow-up and management after primary treatment: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2013;317:961–965. [DOI] [PubMed] [Google Scholar]
- 2. Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/American Society of Clinical Oncology Breast Cancer Survivorship Care Guideline. J Clin Oncol. 2016;346:611–635. [DOI] [PubMed] [Google Scholar]
- 3. Henry NL, Henry LN, Hayes DF, et al. Promoting quality and evidence-based care in early-stage breast cancer follow-up. J Natl Cancer Inst. 2014;1064:dju034.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Beaver JA, Kluetz PG, Pazdur R.. Metastasis-free survival—a new end point in prostate cancer trials. N Engl J Med. 2018;37826:2458–2460. [DOI] [PubMed] [Google Scholar]
- 5. Smith MR, Saad F, Chowdhury S, et al. Apalutamide treatment and metastasis-free survival in prostate cancer. N Engl J Med. 2018;37815:1408–1418. [DOI] [PubMed] [Google Scholar]
- 6. Hayes DF, Bast RC, Desch CE, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 1996;8820:1456–1466. [DOI] [PubMed] [Google Scholar]
- 7. Molina R, Zanon G, Filella X, et al. Use of serial carcinoembryonic antigen and CA 15.3 assays in detecting relapses in breast cancer patients. Breast Cancer Res Treat. 1995;361:41–48. [DOI] [PubMed] [Google Scholar]
- 8. Kokko R, Holli K, Hakama M.. Ca 15-3 in the follow-up of localised breast cancer: a prospective study. Eur J Cancer. 2002;389:1189–1193. [DOI] [PubMed] [Google Scholar]
- 9. Chan DW, Beveridge RA, Muss H, et al. Use of Truquant BR radioimmunoassay for early detection of breast cancer recurrence in patients with stage II and stage III disease. J Clin Oncol. 1997;156:2322–2328. [DOI] [PubMed] [Google Scholar]
- 10. Mariani L, Miceli R, Michilin S, Gion M.. Serial determination of CEA and CA 15.3 in breast cancer follow-up: an assessment of their diagnostic accuracy for the detection of tumour recurrences. Biomarkers. 2009;142:130–136. [DOI] [PubMed] [Google Scholar]
- 11. Babayan A, Pantel K.. Advances in liquid biopsy approaches for early detection and monitoring of cancer. Genome Med. 2018;101:21.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Cristofanilli M, Budd GT, Ellis MJ, et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N Engl J Med. 2004;3518:781–791. [DOI] [PubMed] [Google Scholar]
- 13. Smerage JB, Barlow WE, Hortobagyi GN, et al. Circulating tumor cells and response to chemotherapy in metastatic breast cancer: SWOG S0500. J Clin Oncol. 2014;3231:3483–3489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lucci A, Hall CS, Lodhi AK, et al. Circulating tumour cells in non-metastatic breast cancer: a prospective study. Lancet Oncol. 2012;137:688–695. [DOI] [PubMed] [Google Scholar]
- 15. Rack B, Schindlbeck C, Jückstock J, et al. Circulating tumor cells predict survival in early average-to-high risk breast cancer patients. J Natl Cancer Inst. 2014;1065:dju066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Janni WJ, Rack B, Terstappen LW, et al. Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer. Clin Cancer Res. 2016;2210:2583–2593. [DOI] [PubMed] [Google Scholar]
- 17. Trapp E, Janni W, Schindlbeck C, et al. Presence of circulating tumor cells in high-risk early breast cancer during follow-up and prognosis. J Natl Cancer Inst. 2019;1114:380–387. [DOI] [PubMed] [Google Scholar]
- 18. Janni W, Rack BK, Fasching PA, et al. Persistence of circulating tumor cells in high risk early breast cancer patients five years after adjuvant chemotherapy and late recurrence: results from the adjuvant SUCCESS A trial. J Clin Oncol. 2016;36(suppl; abstr 515). [Google Scholar]
- 19. Sparano J, O'Neill A, Alpaugh K, et al. Association of circulating tumor cells with late recurrence of estrogen receptor-positive breast cancer: a secondary analysis of a randomized clinical trial [published online ahead of print Jul 26, 2018]. JAMA Oncol. 2018. doi: 0.1001/jamaoncol.2018.2574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Bidard FC, Fehm T, Ignatiadis M, et al. Clinical application of circulating tumor cells in breast cancer: overview of the current interventional trials. Cancer Metastasis Rev. 2013;32(1–2):179–188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Hurst RE, Bastian A, Bailey-Downs L, Ihnat MA.. Targeting dormant micrometastases: rationale, evidence to date and clinical implications. Ther Adv Med Oncol. 2016;82:126–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Sparano JA, Zhao F, Martino S, et al. Long-term follow-up of the E1199 phase III trial evaluating the role of taxane and schedule in operable breast cancer. J Clin Oncol. 2015;3321:2353–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Pan H, Gray R, Braybrooke J, et al. 20-year risks of breast-cancer recurrence after stopping endocrine therapy at 5 years. N Engl J Med. 2017;37719:1836–1846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in patients with advanced triple-negative breast cancer: phase Ib KEYNOTE-012 study. J Clin Oncol. 2016;3421:2460–2467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Deng Y, Ma G, Li W, Wang T, Zhao Y, Wu Q.. CDK4/6 inhibitors in combination with hormone therapy for HR(+)/HER2(-) advanced breast cancer: a systematic review and meta-analysis of randomized controlled trials [published online ahead of print May 4, 2018]. Clin Breast Cancer. 2018; pii: S1526-8209(17)30798-X. doi: 10.1016/j.clbc.2018.04.017. [DOI] [PubMed] [Google Scholar]
- 26. Bihani T, Patel HK, Arlt H, et al. Elacestrant (RAD1901), a selective estrogen receptor degrader (SERD), has antitumor activity in multiple ER(+) breast cancer patient-derived xenograft models. Clin Cancer Res. 2017;2316:4793–4804. [DOI] [PubMed] [Google Scholar]
- 27. Dowsett M, Sestak I, Regan MM, et al. Integration of clinical variables for the prediction of late distant recurrence in patients with estrogen receptor-positive breast cancer treated with 5 years of endocrine therapy: CTS5. J Clin Oncol. 2018;3619:1941–1948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Schiavon G, Hrebien S, Garcia-Murillas I, et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2015;7313:313ra182.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Chandarlapaty S, Chen D, He W, 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;210:1310–1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Toy W, Weir H, Razavi P, et al. Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 2017;73:277–287. [DOI] [PMC free article] [PubMed] [Google Scholar]