Next Article in Journal
A Localized Scalable DNA Logic Circuit System Based on the DNA Origami Surface
Previous Article in Journal
Tear Sampling and Biomarker Discovery: A Robust Workflow for Routine Clinical Applications Using UHPLC-MS/MS and Schirmer Strips
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer

by
Malgorzata Szostakowska-Rodzos
1,
Ewa A. Grzybowska
1,*,
Izabella Mysliwy
1,
Renata Zub
2,
Agnieszka Jagiello-Gruszfeld
3,
Maryna Rubach
4,
Aleksandra Konieczna
5 and
Anna Fabisiewicz
1,*
1
Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
2
Cancer Molecular and Genetic Diagnostics Department, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
3
Oncological Clinic, Oncological Mazovian Hospital, al. Solidarności 10, 03-411 Warsaw, Poland
4
Cancer Chemotherapy Day Unit, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
5
Department of Breast Cancer and Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(5), 2038; https://doi.org/10.3390/ijms26052038
Submission received: 28 January 2025 / Revised: 18 February 2025 / Accepted: 21 February 2025 / Published: 26 February 2025
(This article belongs to the Section Molecular Oncology)

Abstract

Monitoring of metastatic breast cancer (mBC) is an important issue in the clinical management of patients. Liquid biopsy has become a non-invasive method for detecting and monitoring cancer in body fluids. The presence of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA) in peripheral blood indicates poor prognosis and may contribute to early detection of progression, but assessment of these levels is still not routine clinical management. The main objective of this study was to estimate the frequency and clinical value of the ESR1 and PIK3CA mutations identified in circulating free DNA (cfDNA.) The second goal was to evaluate whether simultaneous evaluation of CTCs and mutation status in cfDNA increases the prognostic value of liquid biopsy. The results of the analysis of the CTC number and ESR1 and PIK3CA mutations in blood collected from 179 patients with metastatic breast cancer show that ESR1 mutations are more frequent in patients with advanced luminal breast cancer regardless of the type of the treatment. ESR1 mutations appear primarily during progression, as no mutations were found in primary tumor samples. The main conclusion of the study is that combined assessment of CTCs and ESR1 status in liquid biopsy may improve the prognostic value of liquid biopsy.

1. Introduction

Metastatic breast cancer (MBC) is a treatable but still incurable disease. The majority of diagnosed breast cancers are the luminal type expressing estrogen receptors (ERs) and progesterone receptors (PRs) [1]. About 30–50% of patients will eventually relapse due to resistance to the given treatment [2]. This resistance is a consequence of modifications of ERα at the genetic, regulatory, or protein level that allow tumor growth independent of the presence of estrogen. Resistance often develops as a result of acquiring new mutations in the ESR1 gene [3,4]. These mutations are rare (up to 3%) in primary tumors but much more abundant in metastatic lesions, where the rate ranges from 5 to 60% [5]. The most common alterations in ESR1 are point mutations occurring in the ligand binding domain (LBD), in codons 536, 537, 538, and 380 [6,7]. These ESR1-LBD mutations result in constitutively activated ERs, causing decreased sensitivity to endocrine treatments [6,8]. For this reason, tracking mutational changes in ERα during treatment has clinical value and may influence therapeutic decisions during treatment.
Another gene frequently mutated in breast cancer is PIK3CA (coding the catalytic subunit p110α of phosphatidylinositol 3-kinase [PI3K]). The phosphatidylinositol 3-kinase/protein kinase B/mammalian target of the rapamycin (PI3K/AKT/mTOR) pathway is a pivotal intracellular signaling system, and its hyperactivation is a well-known cause of hormonal treatment failure [9]. Mutations in PIK3CA occur mainly in two hotspots at 1047aa and 545aa, accounting for around 70% of all mutations [10,11]. They lead to constitutive activation of PI3K, which has been proposed as a mechanism for endocrine resistance [10].
Due to the heterogeneity of cancer and its dynamic development, recognition of molecular mechanisms responsible for cancer evolution is a challenge. Recently, liquid biopsy has emerged as a noninvasive method for detecting and monitoring cancer in body fluids rather than tumor tissue. Cancer cells release CTCs, ctDNA, RNA (mRNA and micro-RNA), and extracellular vesicles (EV), which can be detected in peripheral blood collected repeatedly during a patient’s treatment [12,13,14]. Unlike tissue biopsy, liquid biopsy can be taken simultaneously with routine blood tests at any stage of the disease. Therefore, a new field of oncology has emerged, focusing on the components of analysis of metastatic tumors circulating in the blood, mainly CTCs and ctDNA. Research in this area will help to better track cancer progression and tailor treatment.
The presence of cfDNA in the blood has proven prognostic significance. Dawson et al. demonstrated that ctDNA is a specific and highly sensitive biomarker in MBC [15], outperforming CTCs in detection frequency and correlation with tumor burden. This was confirmed by other studies [16,17]. Genomic analysis of ctDNA has begun to be incorporated into the clinical management of patients with advanced cancer. The mutational analysis of ESR1 and PIK3CA in cfDNA from MBC patients has been recognized as an important tool for the assessment of the response to treatment and drug efficacy [18,19,20] and has been tested in clinical trials. Mutations in ESR1 and PIK3CA did not show an effect on PFS (progression-free survival) and OS (overall survival) in the MONARCH study [21] but were associated with worse survival in other studies, e.g., the BOLERO-2 trial and the SAFIR02 trial [22,23]. Mutations in other functionally important genes in MBC were tested in the PALOMA-3 trial and PEARL trial, in which TP53 mutations and FGFR1 amplifications were associated with worse outcome regardless of treatment [24,25].
CTCs have been identified as an independent prognostic factor for PFS and OS in the adjuvant, neoadjuvant, and metastatic settings [26,27]. CTC research has also been used as a tool in studying breast cancer heterogeneity [28]. CTC detection and its longitudinal analysis [29,30,31] are still not a clinical standard, and its value as a predictor of disease progression has not yet been established, although many studies suggest that it could be a promising prognostic tool for clinicians [32,33,34]. Until now, several studies have attempted to combine ctDNA information with CTCs [35,36,37], although they mainly focus on cfDNA levels or compare the mutational status of cfDNA and gDNA in CTCs.
The general objective of this study was to evaluate the frequency and clinical value of the ESR1 and PIK3CA mutations identified in cfDNA and to compare these results with primary tumor samples to evaluate if the mutation was originally present in the tumor or if it occurred during metastasis. The other goal was to assess whether the simultaneous evaluation of the status of CTCs and mutational status in cfDNA might strengthen the prognostic value of liquid biopsy. Our work demonstrates that combining the two liquid biopsy approaches results in a better prognosis.

2. Results

2.1. Patients’ Characteristics

In total, 179 patients were enrolled in the study. The clinical characteristics of the patients are summarized in Table 1 and Figure 1. The median follow-up was 53.1 months. The median age of the patients was 63 years at the beginning of the study. Approximately 70% of the patients were characterized with bone metastases. Most of the histological subtypes identified in the primary tumor sample were NST. Most of the patients were treated with radiotherapy and HTH + CHTH + CDK4/6.

2.2. Mutations in cfDNA

In general, mutations in ESR1 or PIK3CA were found in ~63% of the patients. ESR1 mutations were more frequent, as they were found in 53.63% of patients, while PIK3CA mutations in PIK3CA were found in 26.82% of patients (Table 2, Figure 2). Around 15% of the patients were characterized with double mutations in the PIK3CA gene and the ESR1 genes. One patient had double PIK3CA mutations, occurring in p.E545K and p.H1047R hotspots.
For 59 patients, an additional analysis of the mutation status was performed in FFPE (formalin fixed, paraffin embedded) samples as representative samples of a primary tumor. This enabled us to compare the mutational status in primary tumors vs. liquid biopsy. No patient was found to have a mutation in ESR1 in the primary tumor sample, while ~12% were identified with PIK3CA mutations. From this group, ~54% of patients with ESR1 mutations and ~20% with PIK3CA mutations were identified on liquid biopsy (Figure 2). Interestingly, we showed that all ESR1 mutations were identified only in cfDNA samples, while for PIK3CA mutations, the mutation gain during progression was observed only in a few patients. These results highlight that ESR1 mutations develop under the selective pressure of endocrine treatments and might be associated with cancer progression. Therefore, for further validation of the clinical value of cfDNA mutational status, we analyzed data considering only ESR1 mutational status.

2.3. Clinical Value of Liquid Biopsy

  • CTC evaluation
For 96 patients out of a total of 179 patients subjected to ctDNA analysis, an additional CTC evaluation was performed. The characteristics of this group are shown in Table 3. The median age in this group was ~65. CTCs were detected in 36% of the patients, and ≥5 CTCs were found in ~16% of the patients. Patients with ≥5 CTCs detected were characterized with a significantly lower median survival (Figure 2A and Figure 3B). For all patients, an additional evaluation of ESR1 mutational status in cfDNA was performed. To estimate the clinical value of the liquid biopsy, additional analysis was performed for combined liquid biopsy markers as predictors of OS. For patients with ≥5 CTCs and ESR1 mutation in cfDNA material, the median survival was significantly lower than for other patients (11.1 months, compared to 44.3 for patients ≥5 CTCs and N/A for patients with <5 CTCs) (Figure 3C,D). These results highlight that the simultaneous evaluation of liquid biopsy markers might improve the prognostic value of liquid biopsy during treatment.
  • Multivariable COX proportion and hazard regression analysis
To further confirm the clinical value of the simultaneous assessment of the CTCs and the ESR1 mutation, we performed the multivariable Cox proportional hazard regression. The clinicopathological data used for the Cox multivariable model are listed in Table 4 with their reference levels.
Interestingly, the ≥5 CTCs and ESR1 status in cfDNA alone were found to not be significant factors in the univariable and multivariable Cox analysis for the overall survival prognosis. However, when combined, these markers were found to greatly improve the prognostic value of liquid biopsy. The simultaneous presence of ≥5 CTCs and ESR1 mutation in liquid biopsy was found to be a strong predictive factor of OS in univariable (HR = 3.496; 95% CI 1.173–8.484; p-value < 0.05) and multivariable (HR = 3.538; 95% CI 1.126–9.403; p-value < 0.05) analyses (Table 5, Figure 4). Furthermore, these results highlight that combining standard liquid biopsy approaches strongly improves the clinical effectiveness of liquid biopsy as a predictor of OS. Additionally, correlation analysis between ESR1 status and CTC status have shown the absence of correlation (Table S1), supporting the assumption of the independence of these variables.
In a multivariate Cox hazard regression model, we also observed higher HRs for the lobular subtype and combined treatment (HTH + CHTH; HTH + CDK4/6; HTH + CHTH + CDK4/6) (Figure 4). These results might be associated with a low number of patients with the lobular subtype (n < 10), high advancement, and disease aggressiveness for the patients treated with combined therapy.

3. Discussion

Liquid biopsy is one of the most dynamically improving fields in current clinical science. The evaluation of liquid biopsy potential as a prognostic tool is usually done using cfDNA mutational analysis or CTC number counts.
The evaluation of the genetic status of ESR1 and PIK3CA in cfDNA of patients with MBC is recognized as having prognostic and predictive value [38,39,40,41]. Plasma PIK3CA ctDNA specific mutation detected by next generation sequencing is associated with clinical outcomes in advanced breast cancer.
The results of the research carried out in this work confirm previous findings that ESR1 mutations are more frequent in metastatic luminal breast cancer patients irrespective of the type of therapy (endocrine or chemotherapy-based treatments) [38,42] and reveal that pathogenic ESR1 mutations appear mainly during progression, most likely as a result of the selective pressure of endocrine treatments, as no mutations were found in primary tumor samples. PIK3CA mutations are more frequent in primary tumors, and most of them remain present in the advanced stages, but only a few new mutations appear during the metastatic process. These results and the statistical analysis of mutational changes in both genes and their impact on survival (Figure 3) suggest that ESR1 mutations in cfDNA are better suited to be a prognostic tool in MBC. However, our results on the impact of cfDNA mutations were on the borderline of significance (ESR1) or insignificant (PIK3CA) in Kaplan–Meier survival analysis. The clinical relevance of the ESR1 mutations detected in cfDNA evaluated by Kaplan–Meier analysis has been reported several times, with different significance levels [43,44,45,46]. The borderline significance of our analysis may result from the relative heterogeneity of our group of patients in terms of the observation period, so in some patients, resistance-conferring mutations may not be present because there was not enough time for the evolution of the resistance.
However, the results point to the conclusion that this prognostic value of liquid biopsy should be further improved, possibly by combining it with CTC enumeration. This could be achieved by evaluating both values from the same blood sample, which makes it relatively easy to implement in clinical practice.
CTCs are significant prognostic markers in metastatic breast cancer patients. The prognostic value of CTCs has been widely studied, and the presence of ≥5 CTCs was found to be a negative prognostic predictor of OS and PFS [47,48,49]. Longitudinal studies of serially collected samples were reported to improve the prognostic power of CTC enumeration in metastatic breast cancer but require a more organized collection schedule [31,50,51].
In the current study, the presence of CTCs with the ≥5 cutoff was confirmed to be significant. Subsequently, we tested whether the combination of CTC numbers and ESR1 mutational status in blood samples from advanced breast cancer patients can improve the clinical value of a single liquid biopsy. The question of the benefits of combining these two markers has been addressed in several reports, but the authors evaluated the level of cfDNA, its integrity, or a whole profile of genomic alterations and did not evaluate the presence of specific mutation(s) in one gene [35,36,37,52,53].
There are several limitations to our study. First, our findings may have been influenced by heterogeneity of the group, which was not uniformly advanced in the disease. Second, we do not compare mutational status of ESR1 between cfDNA and CTCs, and some reports suggest that the frequency of mutation might be different [54]—however, it could be argued that the cited study had a small sample size and did not achieved significance.
Overall, our findings suggest that the presence of plasma ESR1 mutations in addition to the ≥5 CTC number is unfavorable in the long-term prognosis for these patients and adds additional evidence that early detection of mutation may be clinically helpful for the prediction of treatment efficacy. Additionally, this will help select a specific group of patients who will benefit from a change in treatment. A combined approach would also represent optimal, efficient use of the liquid biopsy sample obtained in one collection.

4. Materials and Methods

4.1. Patients Samples

Blood samples were collected from 179 patients with advanced luminal breast cancer progressing under hormone therapy. Blood collection (9 mL) was carried out once during the treatment follow-up in the Maria Sklodowska-Curie National Institute of Oncology. The selection of patients was carried out by experienced clinicians from the Department of Breast Cancer and Reconstructive Surgery of the Maria Sklodowska-Curie National Research Institute of Oncology. Patients were included for this study between June 2018 and December 2022. The inclusion criteria for the patients were breast cancer with ongoing hormonal treatment, age ≥ 18, and identification of distant metastases. All participants signed an informed consent. Overall, 179 patients were enrolled: 179 patients with cfDNA mutation evaluation and 96 patients with CTCs and cfDNA evaluation.

4.2. Isolation and Preparation of cfDNA

Plasma samples from patients were isolated using the QIAamp Circulating Nucleic Acid Kit (Qiagen; Hilden, Germany) according to the manufacturer’s protocol. The amount of isolated cfDNA was measured using the Quantus system (Promega, Walldorf, Germany) using the QuantiFluor ONE dsDNA system (Promega; Walldorf, Germany). The isolated and measured cfDNA was further used for ddPCR analysis.

4.3. ddPCR Analysis

To test the abundance, ddPCR analysis was performed using the BioRad QX200 Droplet ddPCR system according to the protocol of Schiavon et al. [18]. Probes for ESR1 mutations L536R, Y537S/C, and D538G (Table 6) were purchased from Merck (Rahway, NJ, USA). Reactions were run in multiplexes: L536R with Y537C and Y537S with D538G. For PIK3CA E545K and H1047R mutations, BioRad specific ddPCR Mutations Assays were used (Hercules, CA, USA). The plates were read on a BioRad QX200 droplet reader with BioRad QuantaSoft v1.6.6.0320 software (Hercules, CA, USA).

4.4. Post-Analysis for ddPCR

The estimation of the false positive rate was determined by performing 5 experiments for each assay using WT-only samples, where total amounts of detected mut-positive droplets determined thresholds above which positive droplets in patient samples were to be considered true positive. For each patient, plasma was analyzed in duplicate. Therefore, the PCR results of the patient samples were based on the mean estimated target DNA concentrations (copies/μL) in the merged wells, automatically calculated by the manufacturer’s software. Correction for false positivity was made by subtracting the number of mut-positive droplets detected in the false positive assessment experiments. The mutant allele frequency (MAF) was defined as the number of mut-positive droplets in the total droplet amount (mut-positive and wt-positive). Samples were considered positive if mutation was confirmed in the FFPE sample and mut-positive droplets were found.

4.5. CTC Assessment

After the collection, the blood samples were centrifuged at 2500× g for 15 min. The plasma was collected for the cfDNA analysis, while the buffy coat was transferred to a new 15 mL tube. The CTC assessment with the CytoTrack system was done according to the previously established protocol [34]. The collected cells were stained with the following: Alexa Fluor 488-conjugated pancytokeratin (pan-CK) antibody (1:25) (ThermoFisher Scientific, Waltham, MA, USA), APC conjugated CD45 antibody (1:10) (ThermoFisher Scientific, Waltham, MA, USA), PE conjugated EpCAM antibody (1:10) (ThermoFisher Scientific, Waltham, MA, USA), and 4,6-diamidino-2-phenylindole (DAPI) (Sigma-Aldrich, St Louis, MO, USA) (1:1000). The focus plan for scanning was obtained based on the DAPI stain, at eight places on the disc. Scanning was performed with a 488 nm argon-neon laser, in a spiral pattern with a bandwidth of 10 μm, for 5 min. All signals from the Alexa Fluor 488 emission channel (pan-CK) were recorded and listed in the hotspot table (positive events). The criteria for CTC identification were established as follows: nearly round size with ≥6 µm diameter, visible nucleus, pan-CK signal, and CD45 negative. The clusters were defined as follows: group of ≥3 cells, with at least 3 visible nuclei in the DAPI channel, and with at least 3 cells identified as CTCs.

4.6. FFPE Analysis

FFPE samples obtained from the Department of Cancer Pathomorphology in the Maria Sklodowska-Curie National Research Institute of Oncology were cut 10 µm thin, and up to 8 sections were used for DNA isolation. DNA was isolated using the QIAamp DNA FFPE Tissue Kit (Qiagen, Hilden, Germany) according to the manufacturer’s instructions. The purity of the isolated genetic material was verified on a NanoDrop spectrophotometer (ThermoFisher, Waltham, MA, USA). Only pure DNA with a concentration of at least 50 ng/µL was used for sequencing. Samples were amplified using GoldTaq Polymerase (Applied Biosystems, Waltham, MA, USA) and GeneAmp PCR System 9700 Thermal Cycler (Applied Biosystems, Waltham, MA, USA). PCR products were sequenced using BigDye™ Terminator v3.1 Cycle Sequencing Kit (ThermoFisher, Waltham, MA, USA) and ABI Prism 3130xl Genetic Analyzer (ThermoFisher Waltham, MA, USA). The primers’ sequences are shown in the Table 7.

4.7. Statistical Analysis

Categorized quantitative data at different time points were compared using the Mann–Whitney U test or, if there were more than two categories, the Kruskal–Wallis test. The primary end point was overall survival (OS). OS was defined as the time from blood collection to death from any cause. If an outcome was not reached during the observation time, the variables were censored. Kaplan–Meier plots and log-rank tests were used to illustrate and compare survival between subgroups. Univariable and multivariable hazard ratios (HRs) for selected potential predictors of OS were determined by Cox proportional hazards regression. The fit was measured using the Harrell C index, and the fit of the nested prognostic models was compared using the logarithmic likelihood ratio test (G squared). All data were analyzed using GraphPad Prism 9.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/ijms26052038/s1.

Author Contributions

Analysis of mutations by ddPCR, redaction of abstract, introduction part of discussion: A.F.; CTC results section, statistical analysis, preparation of Table 3, Table 4, Table 5, Table 6 and Table 7, Figure 3 and Figure 4: M.S.-R.; analysis of mutations in FFPE, preparation of Table 1 and Table 2: I.M.; redaction of discussion, Figure 1 and Figure 2, and all manuscript correction: E.A.G.; sequencing and sample collection, R.Z.; selection of patients to the study, collection of clinical data: A.J.-G., M.R. and A.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Maria Skłodowska-Curie Research Institute of Oncology minigrant number SN/MGW19/2024 for A.F., by Narodowe Centrum Nauki, grant number 2016/21/B/NZ2/03473 for E.G., and by 2019/33/N/NZ5/00758 for M.Sz-R.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Ethics Committee of the Maria Sklodowska-Curie National Research Institute of Oncology (31/2020, 18.06.2020 for cfDNA and 84/2020, 17.12.2020 for CTCs).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Material. Further inquiries can be directed to the corresponding authors.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

MBCmetastatic breast cancer
CTCcirculating tumor cell
ctDNAcirculating tumor DNA
cfDNAcirculating free DNA
ERestrogen receptor
PRprogesterone receptor
LBDligand-binding domain
PI3Kphosphatidylinositol 3-kinase
AKTprotein kinase B
mTORmammalian target of the rapamycin
PDXpatient-derived tumor xenograft
EVextracellular vesicle
HRhazard ratio
CIconfidence interval
PFSprogression-free survival
OSoverall survival
MAFmutant allele frequency
HTHhormonal therapy
CHTHchemotherapy
CDK4/6CDK4/6 inhibitors
RTHradiotherapy
ddPCRdigital droplet polymerase chain reaction
FFPEformalin-fixed, paraffin-embedded
WTwild-type
HER2human epidermal growth factor receptor 2

References

  1. Kittaneh, M.; Montero, A.J.; Glück, S. Molecular profiling for breast cancer: A comprehensive review. Biomark. Cancer 2013, 5, 61–70. [Google Scholar] [CrossRef]
  2. Pedersen, R.N.; Esen, B.Ö.; Mellemkjær, L.; Christiansen, P.; Ejlertsen, B.; Lash, T.L.; Nørgaard, M.; Cronin-Fenton, D. The incidence of breast cancer recurrence 10–32 years after primary diagnosis. J. Natl. Cancer Inst. 2022, 114, 391–399. [Google Scholar] [CrossRef]
  3. Aitken, S.J.; Thomas, J.S.; Langdon, S.P.; Harrison, D.J.; Faratian, D. Quantitative analysis of changes in ER, PR and HER2 expression in primary breast cancer and paired nNodal metastases. Ann. Oncol. 2009, 21, 1254–1261. [Google Scholar] [CrossRef]
  4. Simmons, C.; Miller, N.; Geddie, W.; Gianfelice, D.; Oldfield, M.; Dranitsaris, G.; Clemons, M.J. Does confirmatory tumor biopsy alter the management of breast cancer patients with distant metastases? Ann. Oncol. 2009, 20, 1499–1504. [Google Scholar] [CrossRef]
  5. Jeselsohn, R.; Yelensky, R.; Buchwalter, G.; Frampton, G.; Meric-Bernstam, F.; Gonzalez-Angulo, A.M.; Ferrer-Lozano, J.; Perez-Fidalgo, J.A.; Cristofanilli, M.; Goḿez, H.; et al. Emergence of constitutively active estrogen receptor-α mutations in pretreated advanced estrogen receptor-positive breast cancer. Clin. Cancer Res. 2014, 20, 1757–1767. [Google Scholar] [CrossRef]
  6. Robinson, D.R.; Wu, Y.; Vats, P.; Su, F.; Lonigro, R.J.; Cao, X.; Kalyana-Sundaram, S.; Wang, R.; Ning, Y.; Hodges, L.; et al. Activating ESR1 mutations in hormone-resistant metastatic breast cancer. Nat Genet. 2013, 45, 1446–1451. [Google Scholar] [CrossRef]
  7. Toy, W.; Shen, Y.; Won, H.; Green, B.; Sakr, R.A.; Will, M.; Gala, K.; Fanning, S.; King, T.A.; Hudis, C.; et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat. Genet. 2013, 45, 1439–1445. [Google Scholar] [CrossRef] [PubMed]
  8. Toy, W.; Weir, H.; Razavi, P.; Lawson, M.; Goeppert, A.U.; Marie, A.; Smith, A.; Wilson, J.; Morrow, C.; Wong, W.L.; et al. Activating ESR1 mutations differentially affect the efficacy of ER antagonists. Cancer Discov. 2017, 7, 277–287. [Google Scholar] [CrossRef] [PubMed]
  9. Ciruelos Gil, E.M. Targeting the PI3K/AKT/MTOR pathway in estrogen receptor-positive breast cancer. Cancer Treat. Rev. 2014, 40, 862–871. [Google Scholar] [CrossRef]
  10. Fusco, N.; Malapelle, U.; Fassan, M.; Marchiò, C.; Buglioni, S.; Zupo, S.; Criscitiello, C.; Vigneri, P.; Dei Tos, A.P.; Maiorano, E.; et al. PIK3CA mutations as a molecular target for hormone receptor-positive, HER2-negative metastatic breast cancer. Front. Oncol. 2021, 11, 644737. [Google Scholar] [CrossRef] [PubMed]
  11. Samuels, Y.; Wang, Z.; Bardelli, A.; Silliman, N.; Ptak, J.; Szabo, S.; Yan, H.; Gazdar, A.; Powell, S.M.; Riggins, G.J.; et al. High Frequency of mutations of the PIK3CA gene in human cancers. Science 2004, 304, 554. [Google Scholar] [CrossRef]
  12. Guan, X. Cancer metastases: Challenges and opportunities. Acta Pharm. Sin. B 2015, 5, 402–418. [Google Scholar] [CrossRef]
  13. Alix-Panabières, C.; Pantel, K. Liquid biopsy: From discovery to clinical application. Cancer Discov. 2021, 11, 858–873. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, X.; Wang, L.; Lin, H.; Zhu, Y.; Huang, D.; Lai, M.; Xi, X.; Huang, J.; Zhang, W.; Zhong, T. Research progress of CTC, CtDNA, and EVs in cancer liquid biopsy. Front. Oncol. 2024, 14, 1303335. [Google Scholar] [CrossRef]
  15. Dawson, S.-J.; Tsui, D.W.Y.; Murtaza, M.; Biggs, H.; Rueda, O.M.; Chin, S.-F.; Dunning, M.J.; Gale, D.; Forshew, T.; Mahler-Araujo, B.; et al. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 2013, 368, 1199–1209. [Google Scholar] [CrossRef]
  16. Yang, J.; Cheng, L.; Zhang, J.; Chen, L.; Wang, D.; Guo, X.; Ma, X. Predictive value of circulating cell-free DNA in the survival of breast cancer patients a systemic review and meta-analysis. Medicine 2018, 97, e11417. [Google Scholar] [CrossRef] [PubMed]
  17. Lee, J.H.; Jeong, H.; Choi, J.W.; Oh, H.E.; Kim, Y.S. Liquid biopsy prediction of axillary lymph node metastasis, cancer recurrence, and patient survival in breast cancer a meta-analysis. Medicine 2018, 97, e12862. [Google Scholar] [CrossRef]
  18. Schiavon, G.; Hrebien, S.; Garcia-murillas, I.; Cutts, R.J.; Tarazona, N.; Fenwick, K.; Kozarewa, I.; Lopez-knowles, E.; Ribas, R.; Nerurkar, A.; et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci Transl Med. 2016, 7, 313ra182. [Google Scholar] [CrossRef] [PubMed]
  19. Moynahan, M.E.; Chen, D.; He, W.; Sung, P.; Samoila, A.; You, D.; Bhatt, T.; Patel, P.; Ringeisen, F.; Hortobagyi, G.N.; et al. Correlation between PIK3CA mutations in cell-free DNA and everolimus efficacy in HR+, HER2-advanced breast cancer: Results from BOLERO-2. Br. J. Cancer 2017, 116, 726–730. [Google Scholar] [CrossRef]
  20. O’Leary, B.; Hrebien, S.; Morden, J.P.; Beaney, M.; Fribbens, C.; Huang, X.; Liu, Y.; Bartlett, C.H.; Koehler, M.; Cristofanilli, M.; et al. Early circulating tumor DNA dynamics and clonal selection with palbociclib and fulvestrant for breast cancer. Nat. Commun. 2018, 9, 896. [Google Scholar] [CrossRef] [PubMed]
  21. Tolaney, S.M.; Toi, M.; Neven, P.; Sohn, J.; Grischke, E.M.; Llombart-Cussa, A.; Soliman, H.; Wang, H.; Wijayawardana, S.; Jansen, V.M.; et al. Clinical significance of PIK3CA and ESR1 mutations in circulating tumor DNA: Analysis from the MONARCH 2 study of abemaciclib plus fulvestrant. Clin. Cancer Res. 2022, 28, 1500–1506. [Google Scholar] [CrossRef] [PubMed]
  22. Chandarlapaty, S.; Chen, D.; He, W.; Sung, P.; Samoila, A.; You, D.; Bhatt, T.; Patel, P.; Voi, M.; Gnant, M.; 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, 1310–1315. [Google Scholar] [CrossRef] [PubMed]
  23. Mosele, F.; Stefanovska, B.; Lusque, A.; Tran Dien, A.; Garberis, I.; Droin, N.; Le Tourneau, C.; Sablin, M.P.; Lacroix, L.; Enrico, D.; et al. Outcome and molecular landscape of patients with PIK3CA-mutated metastatic breast cancer. Ann. Oncol. 2020, 31, 377–386. [Google Scholar] [CrossRef] [PubMed]
  24. O’leary, B.; Cutts, R.J.; Huang, X.; Hrebien, S.; Liu, Y.; André, F.; Loibl, S.; Loi, S.; Garcia-Murillas, I.; Cristofanilli, M.; et al. Circulating tumor DNA, markers for early progression on fulvestrant with or without palbociclib in ER+ Advanced Breast Cancer. J. Natl. Cancer Inst. 2021, 113, 309–317. [Google Scholar] [CrossRef] [PubMed]
  25. Pascual, J.; Gil-Gil, M.; Proszek, P.; Zielinski, C.; Reay, A.; Ruiz-Borrego, M.; Cutts, R.; Ciruelos Gil, E.M.; Feber, A.; Muñoz-Mateu, M.; et al. Baseline mutations and CtDNA dynamics as prognostic and predictive factors in ER-Positive/HER2-negative metastatic breast cancer patients. Clin. Cancer Res. 2023, 29, 4166–4177. [Google Scholar] [CrossRef] [PubMed]
  26. Janni, W.J.; Rack, B.; Terstappen, L.W.M.M.; Pierga, J.Y.; Taran, F.A.; Fehm, T.; Hall, C.; De Groot, M.R.; Bidard, F.C.; Friedl, T.W.P.; et al. Pooled analysis of the prognostic relevance of circulating tumor cells in primary breast cancer. Clin. Cancer Res. 2016, 22, 2583–2593. [Google Scholar] [CrossRef] [PubMed]
  27. Bidard, F.C.; Michiels, S.; Riethdorf, S.; Mueller, V.; Esserman, L.J.; Lucci, A.; Naume, B.; Horiguchi, J.; Gisbert-Criado, R.; Sleijfer, S.; et al. Circulating tumor cells in breast cancer patients treated by neoadjuvant chemotherapy: A meta-analysis. J. Natl. Cancer Inst. 2018, 110, 560–567. [Google Scholar] [CrossRef] [PubMed]
  28. Rossi, T.; Gallerani, G.; Martinelli, G.; Maltoni, R.; Fabbri, F. Circulating Tumor Cells as a Tool to Untangle the Breast Cancer Heterogeneity Issue. Biomedicines 2021, 9, 1242. [Google Scholar] [CrossRef] [PubMed]
  29. Gerratana, L.; Davis, A.A.; Zhang, Q.; Basile, D.; Rossi, G.; Strickland, K.; Franzoni, A.; Allegri, L.; Mu, Z.; Zhang, Y.; et al. Longitudinal dynamics of circulating tumor cells and circulating tumor DNA for treatment monitoring in metastatic breast cancer. JCO Precis. Oncol. 2021, 5, 943–952. [Google Scholar] [CrossRef]
  30. Magbanua, M.J.M.; Hendrix, L.H.; Hyslop, T.; Barry, W.T.; Winer, E.P.; Hudis, C.; Toppmyer, D.; Carey, L.A.; Partidge, A.H.; Pierga, J.Y.; et al. Serial analysis of circulating tumor cells in metastatic breast cancer receiving first-line chemotherapy. J Natl Cancer Inst. 2021, 113, 443–452. [Google Scholar] [CrossRef]
  31. Szostakowska-Rodzos, M.; Fabisiewicz, A.; Wakula, M.; Tabor, S.; Szafron, L.; Jagiello-Gruszfeld, A.; Grzybowska, E.A. Longitudinal analysis of circulating tumor cell numbers improves tracking metastatic breast cancer progression. Sci. Rep. 2024, 14, 12924. [Google Scholar] [CrossRef] [PubMed]
  32. Zhang, L.; Riethdorf, S.; Wu, G.; Wang, T.; Yang, K.; Peng, G.; Liu, J.; Pantel, K. Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin. Cancer Res. 2012, 18, 5701–5710. [Google Scholar] [CrossRef] [PubMed]
  33. Riethdorf, S.; Muller, V.; Loibl, S.; Nekljudova, V.; Weber, K.; Huober, J.; Fehm, T.; Schrader, I.; Hilfrich, J.; Holms, F.; et al. Prognostic impact of circulating tumor cells for breast cancer patients treated in the neoadjuvant “Geparquattro” trial. Clin Cancer Res. 2016, 23, 5384–5393. [Google Scholar] [CrossRef]
  34. Smit, D.J.; Schneegans, S.; Pantel, K. Clinical applications of circulating tumor cells in patients with solid tumors. Clin. Exp. Metastasis 2024, 41, 403–411. [Google Scholar] [CrossRef]
  35. Kong, S.L.; Liu, X.; Tan, S.J.; Tai, J.A.; Phua, L.Y.; Poh, H.M.; Yeo, T.; Chua, Y.W.; Haw, Y.X.; Ling, W.H.; et al. Complementary sequential circulating tumor cell (CTC) and cell-free tumor DNA (CtDNA) profiling reveals metastatic heterogeneity and genomic changes in lung cancer and breast cancer. Front. Oncol. 2021, 11, 698551. [Google Scholar] [CrossRef] [PubMed]
  36. Rossi, G.; Mu, Z.; Rademaker, A.W.; Austin, L.K.; Strickland, K.S.; Costa, R.L.B.; Nagy, R.J.; Zagonel, V.; Taxter, T.J.; Behdad, A.; et al. Cell-free DNA and circulating tumor cells: Comprehensive liquid biopsy analysis in advanced breast cancer. Clin. Cancer Res. 2018, 24, 560–568. [Google Scholar] [CrossRef] [PubMed]
  37. Madic, J.; Kiialainen, A.; Bidard, F.C.; Birzele, F.; Ramey, G.; Leroy, Q.; Frio, T.R.; Vaucher, I.; Raynal, V.; Bernard, V.; et al. Circulating tumor DNA and circulating tumor cells in metastatic triple negative breast cancer patients. Int. J. Cancer 2015, 136, 2158–2165. [Google Scholar] [CrossRef] [PubMed]
  38. Takeshita, T.; Yamamoto, Y.; Yamamoto-Ibusuki, M.; Tomiguchi, M.; Sueta, A.; Murakami, K.; Omoto, Y.; Iwase, H. Analysis of ESR1 and PIK3CA mutations in plasma cell-free DNA from ER-positive breast cancer patients. Oncotarget 2017, 8, 52142–52155. [Google Scholar] [CrossRef]
  39. Fuqua, S.A.W.; Rechoum, Y.; Gu, G. ESR1 mutations in cell-free DNA of breast cancer: Predictive “Tip of the Iceberg. ” JAMA Oncol. 2016, 2, 1315–1316. [Google Scholar] [CrossRef]
  40. Raei, M.; Heydari, K.; Tabarestani, M.; Razavi, A.; Mirshafiei, F.; Esmaeily, F.; Taheri, M.; Hoseini, A.; Nazari, H.; Shamshirian, D.; et al. Diagnostic Accuracy of ESR1 mutation detection by cell-free DNA in breast cancer: A systematic review and meta-analysis of diagnostic test accuracy. BMC Cancer 2024, 24, 908. [Google Scholar] [CrossRef]
  41. Li, H.; Xu, Y.; Zhao, F.; Song, G.; Rugo, H.S.; Zhang, Y.; Yang, L.; Liu, X.; Shao, B.; Yang, L.; et al. Plasma PIK3CA CtDNA specific mutation detected by Next Generation Sequencing is associated with clinical outcome in advanced breast cancer. Am. J. Cancer Res. 2018, 8, 1873–1886. [Google Scholar] [PubMed]
  42. Li, S.; Shen, D.; Shao, J.; Crowder, R.; Liu, W.; Prat, A.; He, X.; Liu, S.; Hoog, J.; Lu, C.; et al. Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived Xenografts. Cell Rep. 2013, 4, 1116–1130. [Google Scholar] [CrossRef] [PubMed]
  43. Keup, C.; Storbeck, M.; Hauch, S.; Hahn, P.; Sprenger-Haussels, M.; Hoffmann, O.; Kimmig, R.; Kasimir-Bauer, S. Multimodal targeted deep sequencing of circulating tumor cells and matched cell-free DNA provides a more comprehensive tool to identify therapeutic targets in metastatic breast cancer patients. Cancers 2020, 12, 1084. [Google Scholar] [CrossRef] [PubMed]
  44. Takeshita, T.; Yamamoto, Y.; Yamamoto-Ibusuki, M.; Inao, T.; Sueta, A.; Fujiwara, S.; Omoto, Y.; Iwase, H. Clinical significance of monitoring ESR1 mutations in circulating CfDNA in ER positive breast cancer. Oncotarget 2016, 7, 32504. [Google Scholar] [CrossRef] [PubMed]
  45. Zundelevich, A.; Dadiani, M.; Kahana-Edwin, S.; Itay, A.; Sella, T.; Gadot, M.; Cesarkas, K.; Farage-Barhom, S.; Saar, E.G.; Eyal, E.; et al. Erratum. ESR1 mutations are frequent in newly diagnosed metastatic and loco-regional recurrence of endocrine-treated breast cancer and carry worse prognosis. Breast Cancer Res. 2020, 22, 16. [Google Scholar] [CrossRef] [PubMed]
  46. Sim, S.H.; Yang, H.N.; Jeon, S.Y.; Lee, K.S.; Park, I.H. Mutation analysis using cell-free DNA for endocrine therapy in patients with HR+ metastatic breast cancer. Sci. Rep. 2021, 11, 5566. [Google Scholar] [CrossRef]
  47. Peeters, D.J.E.; Van Dam, P.J.; Van Den Eynden, G.G.M.; Rutten, A.; Wuyts, H.; Pouillon, L.; Peeters, M.; Pauwels, P.; Van Laere, S.J.; Van Dam, P.A.; et al. Detection and prognostic significance of circulating tumour cells in patients with metastatic breast cancer according to immunohistochemical subtypes. Br. J. Cancer 2014, 110, 375–383. [Google Scholar] [CrossRef] [PubMed]
  48. Fabisiewicz, A.; Szostakowska-Rodzos, M.; Zaczek, A.J.; Grzybowska, E.A. Circulating tumor cells in early and advanced breast cancer; biology and prognostic value. Int. J. Mol. Sci. 2020, 21, 1671. [Google Scholar] [CrossRef] [PubMed]
  49. Lv, Q.; Gong, L.; Zhang, T.; Ye, J.; Chai, L.; Ni, C.; Mao, Y. Prognostic value of circulating tumor cells in metastatic breast cancer: A systemic review and meta-analysis. Clin. Transl. Oncol. 2016, 18, 322–330. [Google Scholar] [CrossRef] [PubMed]
  50. Magbanua, M.J.M.; Savenkov, O.; Asmus, E.J.; Ballman, K.V.; Scott, J.H.; Park, J.W.; Dickler, M.; Partridge, A.; Carey, L.A.; Winer, E.P.; et al. Clinical significance of circulating tumor cells in hormone receptor–positive metastatic breast cancer patients who received letrozole with or without bevacizumab. Clin. Cancer Res. 2020, 26, 4911–4920. [Google Scholar] [CrossRef] [PubMed]
  51. Fabisiewicz, A.; Szostakowska-Rodzos, M.; Grzybowska, E.A. Improving the prognostic and predictive value of circulating tumor cell enumeration: Is longitudinal monitoring the answer? Int. J. Mol. Sci. 2024, 25, 10612. [Google Scholar] [CrossRef] [PubMed]
  52. Fernandez-Garcia, D.; Hills, A.; Page, K.; Hastings, R.K.; Toghill, B.; Goddard, K.S.; Ion, C.; Ogle, O.; Boydell, A.R.; Gleason, K.; et al. Plasma cell-free DNA (CfDNA) as a predictive and prognostic marker in patients with metastatic breast cancer. Breast Cancer Res. 2019, 21, 149. [Google Scholar] [CrossRef] [PubMed]
  53. Ye, Z.; Wang, C.; Wan, S.; Mu, Z.; Zhang, Z.; Abu-Khalaf, M.M.; Fellin, F.M.; Silver, D.P.; Neupane, M.; Jaslow, R.J.; et al. Association of clinical outcomes in metastatic breast cancer patients with circulating tumour cell and circulating cell-free DNA. Eur. J. Cancer 2019, 106, 133–143. [Google Scholar] [CrossRef]
  54. Smilkou, S.; Ntzifa, A.; Tserpeli, V.; Balgkouranidou, I.; Papatheodoridi, A.; Razis, E.; Linardou, H.; Papadimitriou, C.; Psyrri, A.; Zagouri, F.; et al. Detection Rate for ESR1 Mutations Is Higher in Circulating-Tumor-Cell-Derived Genomic DNA than in Paired Plasma Cell-Free DNA Samples as Revealed by ddPCR. Mol. Oncol. 2025, 1–11. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart with the overall patient data: (A) flowchart of the study design; (B) metastatic status of patients included in the studies, divided into bone metastasis only, bones and other lesions, and locally advanced; (C) CTC status of patients included in the studies.
Figure 1. Flowchart with the overall patient data: (A) flowchart of the study design; (B) metastatic status of patients included in the studies, divided into bone metastasis only, bones and other lesions, and locally advanced; (C) CTC status of patients included in the studies.
Ijms 26 02038 g001
Figure 2. The mutational status in cfDNA of patients included in the study: (A) The graphical representation of mutational hotspots and main mutations detected in this study in PIK3CA and ESR1 genes. (B) The frequency of mutations in PIK3CA and ESR1 in cfDNA; data from 179 patients’ samples. (C) The incidence of mutations in PIK3CA and ESR1 genes in samples from 59 patients for whom the analysis of FFPE and cfDNA mutational status was done; for patients with CTCs detected, the number of CTCs was also included; N/D is for patients without CTCs. red = mutation; green = no mutation.
Figure 2. The mutational status in cfDNA of patients included in the study: (A) The graphical representation of mutational hotspots and main mutations detected in this study in PIK3CA and ESR1 genes. (B) The frequency of mutations in PIK3CA and ESR1 in cfDNA; data from 179 patients’ samples. (C) The incidence of mutations in PIK3CA and ESR1 genes in samples from 59 patients for whom the analysis of FFPE and cfDNA mutational status was done; for patients with CTCs detected, the number of CTCs was also included; N/D is for patients without CTCs. red = mutation; green = no mutation.
Ijms 26 02038 g002
Figure 3. The Kaplan–Meier survival analysis for overall survival (OS) for a group of 96 patients with CTCs and ESR1 mutation. (A) Patients divided according to ESR1 mutational status; mutation detected—red line, wild-type only detected—black line. (B) Patients divided according to the CTC status: ≥5 CTCs—red line, <5 CTCs—black line. (C) Patients divided according to combined CTCs and ESR1 status: ≥5 CTCs and ESR1 mutation—red line, <5 CTCs and wild-type ESR1—black line. (D) Patients divided according to combined CTCs and ESR1 status: ≥5 CTCs—red line, <5 CTCs—black line, ≥5 CTCs and ESR1 mutation—dashed red line, <5 CTCs and wild-type ESR1—dashed black line. The p-values for survival analysis in log-rank test were stated at the bottom right corner of each graph: *—p-value < 0.5; **—p-value < 0.01.
Figure 3. The Kaplan–Meier survival analysis for overall survival (OS) for a group of 96 patients with CTCs and ESR1 mutation. (A) Patients divided according to ESR1 mutational status; mutation detected—red line, wild-type only detected—black line. (B) Patients divided according to the CTC status: ≥5 CTCs—red line, <5 CTCs—black line. (C) Patients divided according to combined CTCs and ESR1 status: ≥5 CTCs and ESR1 mutation—red line, <5 CTCs and wild-type ESR1—black line. (D) Patients divided according to combined CTCs and ESR1 status: ≥5 CTCs—red line, <5 CTCs—black line, ≥5 CTCs and ESR1 mutation—dashed red line, <5 CTCs and wild-type ESR1—dashed black line. The p-values for survival analysis in log-rank test were stated at the bottom right corner of each graph: *—p-value < 0.5; **—p-value < 0.01.
Ijms 26 02038 g003
Figure 4. The graphical representation of the hazard ratios (HRs) and confidence intervals (95% CI) for multivariable Cox proportional hazard regression model. The dashed black line represents HR = 1. HR for the combined CTCs ≥ 5 count and the presence of ESR1 mutation (HR = 3.538; 95%CI 1.126–9.403; p-value 0.0172) is significantly higher than HRs for CTC count and ESR1 mutation alone (HR = 1.814; 95%CI 0.7310–4.124; p-value ≥ 0.05, and HR = 0.5616; 95%CI 0.2686–1.165; p-value ≥ 0.05, respectively).
Figure 4. The graphical representation of the hazard ratios (HRs) and confidence intervals (95% CI) for multivariable Cox proportional hazard regression model. The dashed black line represents HR = 1. HR for the combined CTCs ≥ 5 count and the presence of ESR1 mutation (HR = 3.538; 95%CI 1.126–9.403; p-value 0.0172) is significantly higher than HRs for CTC count and ESR1 mutation alone (HR = 1.814; 95%CI 0.7310–4.124; p-value ≥ 0.05, and HR = 0.5616; 95%CI 0.2686–1.165; p-value ≥ 0.05, respectively).
Ijms 26 02038 g004
Table 1. Clinical characteristics of patients.
Table 1. Clinical characteristics of patients.
VariablesNumber of Patients
Age
<6389
≥6390
HER2 status
HER2+5
HER−161
N/D13
No of meta sites
183
249
≥347
Meta sites
Bones133
Liver67
Lung58
Other81
Hitological subtype
NST133
Lobular16
Other30
Treatment
HTH32
HTH + CHTH36
HTH + CDK4/626
HTH + CHTH + CDK4/685
Radiotherapy
RTH+100
RTH−79
Table 2. Frequency of the mutations identified in the cfDNA material.
Table 2. Frequency of the mutations identified in the cfDNA material.
MutationFrequencyNo of Patients with Mutated Samples
PIK3CA p.E545K12.85%23
PIK3CA p.H1047R14.53%26
ESR1 p.Y537S or D538G53.07%95
ESR1 p.R536R or Y537C2.79%5
Table 3. Characteristics of patients evaluated for CTCs.
Table 3. Characteristics of patients evaluated for CTCs.
VariablesNumber of Patients
Age
<6550
≥6546
HER2 status
HER2+5
HER−86
N/D5
No of meta sites
158
227
≥321
Meta sites
Bones70
Liver32
Lung26
Other36
Hitological subtype
NST75
Other21
Treatment
HTH22
HTH + CHTH15
HTH + CDK4/614
HTH + CHTH + CDK4/645
Radiotherapy
RTH+46
RTH−50
ESR1 status
ESR1 mutation58
ESR1 WT38
Table 4. The clinical variables used for the Cox multivariable model.
Table 4. The clinical variables used for the Cox multivariable model.
Clinical VariableReference Level
TreatmentHTH
Histopathological subtypeNST
Age<65
Table 5. The results of Cox proportional hazard regression analyses.
Table 5. The results of Cox proportional hazard regression analyses.
Univariable Analysis
VariableHR95 CIp-Value
ESR1 mutation0.58320.2853–1.1860.1339
≥5 CTCs1.7750.7423–3.8210.1636
ESR1 mut + ≥5 CTCs3.4961.173–8.4840.0113
Multivariable analysis
VariableHR95 CIp-value
ESR1 mutation0.56160.2686–1.1650.1197
≥5 CTCs1.8140.7310–4.1240.1714
ESR1 mut + ≥5 CTCs3.5381.126–9.4030.0172
Table 6. Sequence of probes and starters used for the assessment of the ESR1 mutation status.
Table 6. Sequence of probes and starters used for the assessment of the ESR1 mutation status.
MutationProbe SequencePrimer Sequence
L536R[6FAM]TGGTGCCCCGCTATGACC[BHQ1]FF 5′AGGCATGGAGCATCTGTACA3′
R5′TTGGTCCGTCTCCTCCA3′
Y537S[6FAM]TGGTGCCCCTCTCTGACCT[BHQ1]FF 5′AGGCATGGAGCATCTGTACA3′
R5′TTGGTCCGTCTCCTCCA3′
D538G[6FAM]CCCTCTATGGCCTGCTGCT[BHQ1]FF 5′AGGCATGGAGCATCTGTACA3′
R5′TTGGTCCGTCTCCTCCA3′
Y537C[6FAM]TGCCCCTCTGTGACCTGCT[BHQ1]FF 5′AGGCATGGAGCATCTGTACA3′
R5′TTGGTCCGTCTCCTCCA3′
WT ESR1[HEX]TGGTGCCCCTCTATGACCTG[BHQ1]FF 5′AGGCATGGAGCATCTGTACA3′
R5′TTGGTCCGTCTCCTCCA3′
Table 7. Sequences of the primers used for Sanger sequencing.
Table 7. Sequences of the primers used for Sanger sequencing.
GeneForward PrimerReverse PrimerProduct Length
ESR1 exon 85′-TCTGTGTCTTCCCACCTACAGT-3′5′-ATGCGATGAAGTAGAGCCCG-3′200 bp
PIK3CA exon 95′-AGCTAGAGACAATGAATTAAGGGA-3′5′-TCCATTTTAGCACTTACCTGTGAC-3′130 bp
PIK3CA exon 205′-AACTGAGCAAGAGGCTTTGGA-3′5′-CAATCGGTCTTTGCCTGCTG-3′200 bp
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Szostakowska-Rodzos, M.; Grzybowska, E.A.; Mysliwy, I.; Zub, R.; Jagiello-Gruszfeld, A.; Rubach, M.; Konieczna, A.; Fabisiewicz, A. The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer. Int. J. Mol. Sci. 2025, 26, 2038. https://doi.org/10.3390/ijms26052038

AMA Style

Szostakowska-Rodzos M, Grzybowska EA, Mysliwy I, Zub R, Jagiello-Gruszfeld A, Rubach M, Konieczna A, Fabisiewicz A. The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer. International Journal of Molecular Sciences. 2025; 26(5):2038. https://doi.org/10.3390/ijms26052038

Chicago/Turabian Style

Szostakowska-Rodzos, Malgorzata, Ewa A. Grzybowska, Izabella Mysliwy, Renata Zub, Agnieszka Jagiello-Gruszfeld, Maryna Rubach, Aleksandra Konieczna, and Anna Fabisiewicz. 2025. "The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer" International Journal of Molecular Sciences 26, no. 5: 2038. https://doi.org/10.3390/ijms26052038

APA Style

Szostakowska-Rodzos, M., Grzybowska, E. A., Mysliwy, I., Zub, R., Jagiello-Gruszfeld, A., Rubach, M., Konieczna, A., & Fabisiewicz, A. (2025). The Combined Assessment of CTC and ESR1 Status in Liquid Biopsy Samples Enhances the Clinical Value of Prediction in Metastatic Breast Cancer. International Journal of Molecular Sciences, 26(5), 2038. https://doi.org/10.3390/ijms26052038

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop