Next Article in Journal
A Novel Loss-of-Function SEMA3E Mutation in a Patient with Severe Intellectual Disability and Cognitive Regression
Next Article in Special Issue
Lack of Major Genome-Wide DNA Methylation Changes in Succinate-Treated Human Epithelial Cells
Previous Article in Journal
Locally Secreted Semaphorin 4D Is Engaged in Both Pathogenic Bone Resorption and Retarded Bone Regeneration in a Ligature-Induced Mouse Model of Periodontitis
Previous Article in Special Issue
DNA Hypomethylation Is Associated with the Overexpression of INHBA in Upper Tract Urothelial Carcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

ESR1 Methylation Measured in Cell-Free DNA to Evaluate Endocrine Resistance in Metastatic Breast Cancer Patients

Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Dr. Molewaterplein 40, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(10), 5631; https://doi.org/10.3390/ijms23105631
Submission received: 2 April 2022 / Revised: 13 May 2022 / Accepted: 16 May 2022 / Published: 18 May 2022
(This article belongs to the Special Issue DNA Methylation in Cancer Development and Progression)

Abstract

:
ESR1 methylation was proposed as mechanism for endocrine resistance in metastatic breast cancer patients. To evaluate its potential as a minimally invasive biomarker, we investigated the feasibility of measuring ESR1 methylation in cell-free DNA (cfDNA) and its association with endocrine resistance. First, we provided evidence that demethylation in vitro restores ER expression. Subsequently, we found that ESR1 methylation in cfDNA was not enriched in endocrine-resistant versus endocrine-sensitive patients. Interestingly, we found a correlation between ESR1 methylation and age. Publicly available data confirm an age-related increase in ESR1 methylation in leukocytes, confounding the determination of the ESR1 methylation status of tumors using cfDNA.

1. Introduction

Estrogen is a hormone that exerts a variety of functions in both reproductive tissues and nonreproductive tissues through the activation of ligand-dependent transcription factors, one of which is ERα, encoded by the estrogen receptor 1 gene (ESR1). ERα-mediated transcription is regulated at different levels and involves both direct and indirect mechanisms. ER can directly bind to specific DNA sequences called estrogen response elements (EREs), but ERs can also be recruited onto DNA by tethering via interactions with other transcription factors that regulate transcription in an ERE-independent manner. ERα-mediated transcription is therefore a complex process that involves various coregulatory proteins and transcription factors [1,2].
In addition to its physiological role, ERα is also associated with the development and progression of breast cancer. In patients with estrogen receptor (ER)-positive breast cancer, endocrine therapy, which works by either blocking estrogens from attaching to the ER or by deprivation of estrogen, remains the mainstay of treatment. Although most patients initially respond to endocrine therapy, ultimately all patients will develop endocrine resistance. This poses an important challenge in patients with ER-positive breast cancer, and biomarkers are needed to guide clinical management. Different mechanisms underlying endocrine resistance were identified in metastatic breast cancer (MBC) patients, one of the most recognized mechanisms being the occurrence of activating hotspot mutations in ESR1, ESR1 hotspot mutations are found in around 40% of patients with endocrine-resistant breast cancer [3,4,5,6,7]. They are located within the ligand-binding domain of the gene and result in an estrogen-independent constitutive activation of the ER [8,9]. This enables breast cancer cells to grow independently of the presence of estrogen.
Next to mutations in ESR1, other mechanisms have been proposed to induce estrogen-independent growth of breast cancer cells, highlighting the complex gene regulation of ESR1. For example, inactivating mutations in neurofibromin (NF1), which functions as an ER transcriptional co-repressor [10], were found in endocrine-resistant breast cancer [7]. Over the past few years, the loss of the ER by epigenetic silencing of ESR1 has received increased attention as a potential alternative mechanism of endocrine resistance. Several studies found a correlation between the hypermethylation of cytosine guanine phosphodiesters (CpG dinucleotides) of the ESR1 promoter and decreased ER expression in breast cancer tissue [11,12,13,14,15,16], and few studies also found a correlation of ESR1 promoter methylation with outcomes of endocrine treatment [11,17]. However, whether there is a direct causal association between ESR1 methylation and ER expression has been both proposed [16] and disputed [18].
Following the hypothesis that ESR1 methylation is associated with acquired resistance to endocrine treatment, it is expected to arise during treatment. To test this hypothesis, studying ESR1 methylation repetitively during treatment is required. A non-invasive tool to study tumor biology in blood is cell-free DNA (cfDNA), which is fragmented DNA present in the circulation likely composed of all dying cells in the body. As such, cfDNA mostly originates from leukocytes [19]. However, in patients with cancer, a varying fraction of the DNA that circulates in blood is tumor-derived (ctDNA). In most patients with metastatic breast cancer, this tumor-specific fraction does not exceed 10% [20]. However, this fraction can be effectively used to repeatedly study the genetic composition of the tumor for purposes such as the monitoring of treatment response [21,22].
The current study aims to evaluate whether determining ESR1 methylation in cfDNA may provide a minimally invasive biomarker for endocrine resistance. For this purpose, we first established that ER expression was indeed directly associated with DNA methylation in vitro. Subsequently, we evaluated whether the detection of ESR1 methylation in cfDNA was feasible and associated with endocrine resistance.

2. Results

2.1. ER Expression Is Regulated by Methylation

We first set out to provide direct evidence that demethylation by 5-aza-2′-deoxycytidine (DAC) restores ER expression in the ER-negative MDA-M-436 cell line. MDA-MB-436 cells were treated with DAC for 5 days. After 5 days, we observed a decrease in ESR1 methylation, compared to cells without any treatment or treated only with the DAC solvent dimethyl sulfoxide (DMSO, Figure 1 top). Concurringly, the mRNA expression of ESR1 was restored in these DAC-treated MDA-MB-436 cells compared to untreated and mock-treated cells (Figure 1 bottom). These results show that ER expression is regulated by DNA-methylation, although the exact methylation site responsible for silencing of ESR1 remains to be elucidated.

2.2. ESR1 Methylation Prevalence in cfDNA and Its Prognostic Value

The methylation of ESR1 was successfully determined in all 31 patients from cohort 1 and 18/20 healthy blood donors (HBDs; EDTA tubes), as well as 17/18 patients from cohort 2 and 7/10 HBDs (CellSave tubes). Figure 2 summarizes the cohorts and samples included in this study. ESR1 methylation levels of HBDs were not significantly different between Ethylenediaminetetraacetic acid (EDTA, n = 7) and CellSave tubes (n = 18) from different donors (Mann–Whitney U p = 0.809). The median age of patients was higher than the median age of healthy donors in cohort 1 (Mann–Whitney U p = 0.009) but not cohort 2 (Mann–Whitney U p = 0.391, Table S1). In patient samples, ESR1 methylation levels exceeded the cut-off in 12/31 (39%) endocrine treatment-naïve patients from cohort 1 and in 7/17 (41%) patients from cohort 2 post endocrine treatment, which was not significantly different (Fisher’s exact p = 1, Figure 3A). Importantly, the presence of ESR1 methylation in plasma at baseline was not predictive for progression-free survival on first-line treatment with an aromatase inhibitor (AI) in cohort 1 (logrank p = 0.660). From the 19 patients in which we detected ESR1 methylation, 3 also carried an ESR1 mutation (16%), whereas in the 28 patients without ESR1 methylation, 8 showed an ESR1 mutation (29%, Fisher’s exact p = 0.319). The proportion of patients who were positive for ESR1 methylation significantly increased with age (Figure 3B, chi-square test for trend p < 0.001).

2.3. ESR1 Methylation and Age

Following up on these results in our relatively small cohort, we investigated whether methylation levels in white blood cells were age-related in public data [23]. Interestingly, we found that the methylation of the CpG dinucleotides covered by our assay was moderately, but significantly, associated with age (Figure 3C, Spearman rho = 0.171, p < 0.001). This association with age was seen for all CpG dinucleotides in the region of promoter A (Figure 3D) and promoter B (Figure 3E), although the latter association was only weak. CpG dinucleotides in the region of promoter C were highly methylated in white blood cells in all cases, irrespective of age (Figure 3F). The locations of ESR1 promoter regions and cg probes are summarized in Table S2.

3. Discussion

The primary aim of this study was to evaluate whether ESR1 methylation was indeed involved in regulation of ER expression and, as such, may provide a minimally invasive biomarker for the detection of endocrine resistance.
Treatment with a demethylating agent increased the expression of ESR1 in vitro. Although this experiment does not pinpoint which methylation site is responsible for ESR1 silencing, these findings are in line with other studies suggesting that the methylation of ESR1 negatively regulates ER expression in vivo [11,12,13,14,15,16]. Notwithstanding the fact that ESR1 methylation appears to be at least partly involved in the regulation of its expression, we did not find an association between the presence of ESR1 methylation in cfDNA and progression in endocrine treatment, nor was baseline ESR1 methylation predictive for outcomes of first-line treatment with AIs. Despite its limitations in sample size, our study was sufficiently powered to detect a similar increase in ESR1 methylation from endocrine-sensitive to endocrine-resistant patients as was previously observed for ESR1 mutation incidence. Additionally, the anticipated mutual exclusivity between methylation and mutation was not observed. Instead, we found that the proportion of ESR1-methylation-positive patients increased with age. Although this observation was only modest in our patient samples, we found a more pronounced positive correlation with age in WBCs using public data. Irrespective of whether ESR1 methylation indeed suppresses ER expression, this observation limits the use of ESR1 methylation in cfDNA as a biomarker of endocrine resistance. None of the earlier studies that investigated ESR1 methylation in serum or plasma reported an association with age, nor did they describe whether HBDs were age-matched [11,14,24,25]. In our analyses, the reference samples were not fully age-matched as the blood bank from which the reference samples were obtained set the maximum age for blood donation at 80 years. This might have resulted in a slight overestimation of ESR1 methylation incidence in part of our samples. Based on a mutation analysis, 53% of the patients had detectable ctDNA with a median variant allele frequency of 5.6% anda range from 0.04 to 45%. These were not correlated with the ESR1 methylation level (Spearman’s rho = −0.066, p = 0.719). Together with the fact that we observed a significant correlation between age and ESR1 promoter methylation in WBCs, it is likely that part of the ESR1 methylation signal in cfDNA was not tumor-specific. Although we set a cut-off using data from HBDs, and all HBDs were aged >50, our HBDs were not fully age-matched. This observation underlines the importance of using age-matched HBDs when performing epi-genetic analysis of cfDNA, which is often a complicating factor.
The fact that ESR1 contains multiple promoter regions further complicates formal comparison of findings between studies. Most previous studies focused on promoter A, which is closest to exon 1 and located within a CpG island (i.e., region enriched for CpG dinucleotides). Alternatively, methylation of CpG dinucleotides located in promoter C, and not within promoter A itself, was suggested to be associated with the altered transcription of the ERα gene in AI-resistant cell lines [12]. However, we found that promoter C was highly methylated in WBCs, complicating the analysis of this region in cfDNA. Further underlining the importance of genomic location, another study suggested that the methylation of ER enhancers and not ER promoters were responsible for reduced ESR1 binding and the decreased expression of ER regulators [18]. Of those ER enhancers, one enhancer site was located within ESR1 itself. However, this CpG dinucleotide site is also a common SNP, limiting its utility as a routine biomarker when analyzed as a single-marker.
In conclusion, while it was proposed that epigenetic silencing of the ER receptor might play a role in endocrine resistance in breast cancer, our results in cfDNA did not find support for this. We note that the evaluation of the tumor’s ESR1 methylation status in cfDNA may be confounded by the presence of a background age-related methylation signal of ESR1 in WBCs.

4. Methods

4.1. Clinical Samples

We included plasma samples from patients with ER+/HER2- MBC, selected from three studies. Cohort 1 comprised patients starting treatment with first-line aromatase-inhibitors (CareMore-AI study, trialregister.nl, number NL4884), whereas cohort 2 included patients progressing on any line of endocrine therapy (TAX-ESR1 study on breast cancer, trialregister.nl, number NL7280 and the CareMore-Trastuzumab study on breast cancer, trialregister.nl, number NL4977). The study was performed in accordance with the Declaration of Helsinki. All patients gave written informed consent prior to study procedures. The median age of patients in both cohorts was 66 years (cohort 1 range: 47–88 years; cohort 2 range: 55–70 years). In cohort 1, samples were collected in EDTA tubes and centrifuged within 24 h after collection. Using the same tube type and processing, samples from 20 female healthy donors with a median age of 58 years (range: 55–74 years) were used as a reference. In cohort 2, samples were collected in CellSave tubes and isolated within 96 h after collection. Using the same tube type and processing, samples from 10 female healthy donors with a median age of 61 years (range: 55–70 years) were used as a reference. EDTA (n = 20) and CellSave plasma (n = 10) processed similarly from age- and gender-matched healthy blood donors (HBD) were used as references. cfDNA was isolated from plasma using the Maxwell® (MX) RSC ccfDNA Plasma Kit (Promega, Madison, WI, USA) in cohort 1 and the QIAamp Circulating Nucleic Acids kit (Qiagen, Venlo, The Netherlands) in cohort 2.

4.2. ESR1 Mutations

For all samples, ESR1 mutations were detected using the Oncomine™ Breast Cancer panel containing 10 breast cancer specific genes and including molecular barcoding to establish a limit of detection (LOD) as low as 0.1% (Life Technologies, Carlsbad, CA, USA). For all samples, 10 ng DNA input was used for library preparation. Samples with a sequencing coverage <500 molecules were excluded from analysis. Additionally, variants were annotated as true variants when they were identified in at least 3 unique molecules.

4.3. Culturing Cell Lines and DAC-Treatment

MDA-MB-436 and HCC1187 breast cancer cells (ATCC, HTB-130 & CRL-2322; authenticated by Short Tandem Repeat profiling) were cultured in RPMI1640 glutamax medium (Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% heat-inactivated fetal bovine serum, 100 µg/mL penicillin, and 80 µg/mL streptomycin. To analyze restoration of ER mRNA expression following demethylation, MDA-MB-436 cells were treated with 0.1 µM, 0.2 µM, and 0.5 µM 5-aza-2′ deoxycytidine (DAC) (Sigma, St. Louis, MO, USA) and diluted in DMSO daily for 5 days, along with untreated and mock-treated cells. The HCC1187 cell line was excluded from DAC treatment because ESR1 is unmethylated in this cell line [26] (GSE57342). Genomic DNA or RNA was extracted from cell line pellets using the Nucleospin Tissue kit (Macherey-Nagel, Düren, Germany) or Trizol (Tebu-Bio, Heerhugowaard, The Netherlands), respectively, according to the manufacturer’s instructions.

4.4. ESR1-Targeted Quantitative Methylation-Specific PCR (qMSP)

Based on the RefSeq-curated sequence, ESR1 has 3 promoter regions, referred to as promoter A, B, and C, according to literature (Figure 4) [12]. Of these, only promoter A is located within a CpG island. The methylation status of ESR1 promotor region A of DAC-treated and control cell lines and patient cfDNA was determined by qMSP analysis after sodium bisulfite conversion (BSC). BSC was performed with the EZ DNA Methylation kit (Zymo Research, Orange, CA, USA) using 10 ng cfDNA for patient samples and with 500 ng genomic DNA for cell line samples following the manufacturer’s instructions. We adapted previously published ESR1 primers [17,25,27] to amplify the methylated promotor region of ESR1 in a duplex reaction with the β-actin (ACTB) reference gene. Details of the primers and probes used are given in Table S3. The sensitivity and efficiency of our duplex assay were determined with the MDA-MB-436 cell-line DNA, which is methylated at promoter A of ESR1, in serial dilution using unmethylated HCC1187 DNA after whole genome amplification. Our duplex assay detects methylated ESR1 down to 0.1% in a background of unmethylated DNA (Figure S1) and down to 0.1 ng of total input of methylated DNA (Figure S2). qMSP reactions were carried out in a final reaction volume of 25 µL, containing 300 nM of each primer, 125 nM of each probe, 12.5 µL 2x EpiTect MethyLight Master Mix (w/o ROX, Qiagen, Venlo, The Netherlands), and 1 µL 50x diluted ROX Dye Solution. qPCR reactions were performed on the Mx3000P and Mx3005P QPCR Systems (Stratagene, La Jolla, CA, USA). Samples with an ACTB-Ct value above 32 were excluded for data analysis. Methylation values of ESR1 were normalized to ACTB using the comparative Ct method (2−ΔCT). Genomic DNA from methylated, MDA-MB-436 cells was used as positive control, whereas whole-genome-amplified HCC1187 DNA served as unmethylated control DNA.

4.5. Reverse-Transcriptase Quantitative PCR (RT-PCR)

Two μg of the isolated RNA was converted to cDNA using the Thermo Scientific RevertAid H Minus First Strand cDNA Synthesis Kit (Fermentas, Thermo Scientific, Waltham, MA, USA), followed by an RNase H step (Ambion, Life Technologies, Carlsbad, CA, USA) to digest any remaining RNA. Quantitative real-time PCR targeting exon 4 of ESR1 [28] was performed in triplicate with the Mx3000P QPCR machine (Agilent Technologies, Santa Clara, CA, USA) using ABgene Absolute Universal or Absolute SYBR Green with ROX PCR reaction mixtures (Thermo Scientific, Waltham, MA, USA) according the manufacturer’s instructions.

4.6. Statistical Analysis

cfDNA from healthy donors (EDTA n = 10, CellSave n = 20) was used to determine a cut-off for ESR1 methylation positivity, which was defined as the upper limit of the 95% confidence interval. Cut-offs were separately determined for EDTA and CellSave tubes. Statistical tests used are specified throughout the Results section. p values < 0.05 were considered statistically significant. Statistical analysis was performed using SPSS Statistics, version 25.0 (ICM Co., Armonk, NY, USA).

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23105631/s1.

Author Contributions

Conceptualization, M.K.B., T.D., J.W.M.M. and S.M.W.; formal analysis, M.K.B., T.D. and S.M.W.; writing—original draft preparation, M.K.B., T.D. and S.M.W.; writing—review and editing, S.S., J.W.M.M. and S.M.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Dutch Cancer Society (project no. NKB-EMCR-2016-108154).

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the Institutional Review Board (or Ethics Committee) of the Erasmus Medical Center (CareMore-AI study, MEC14-588, date of approval: 10 February 2015; CareMore-Trastuzumab study, MEC14-589, date of approval: 18 February 2015; TAX-ESR1 study, MEC17-497, date of approval: 23 November 2017).

Informed Consent Statement

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

Data Availability Statement

Sequences of the primers and probes that were used are provided in the Supplementary Data. Methylation data from white blood cells were publicly available and downloaded from NCBI’s Gene Expression Omnibus, using the following accession number: GSE40279.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Klinge, C.M. Estrogen receptor interaction with estrogen response elements. Nucleic Acids Res. 2001, 29, 2905–2919. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Yaşar, P.; Ayaz, G.; User, S.D.; Güpür, G.; Muyan, M. Molecular mechanism of estrogen-estrogen receptor signaling. Reprod. Med. Biol. 2016, 16, 4–20. [Google Scholar] [CrossRef] [PubMed]
  3. Razavi, P.; Li, B.T.; Brown, D.N.; Jung, B.; Hubbell, E.; Shen, R.; Abida, W.; Juluru, K.; De Bruijn, I.; Hou, C.; et al. High-intensity sequencing reveals the sources of plasma circulating cell-free DNA variants. Nat. Med. 2019, 25, 1928–1937. [Google Scholar] [CrossRef] [PubMed]
  4. Schiavon, G.; Hrebien, S.; Garcia-Murillas, I.; Cutts Rosalind, J.; Pearson, A.; Tarazona, N.; Fenwick, K.; Kozarewa, I.; Lopez-Knowles, E.; Ribas, R.; et al. Analysis of ESR1 mutation in circulating tumor DNA demonstrates evolution during therapy for metastatic breast cancer. Sci. Transl. Med. 2015, 7, ra182–ra313. [Google Scholar] [CrossRef] [Green Version]
  5. Fribbens, C.; O’Leary, B.; Kilburn, L.; Hrebien, S.; Garcia-Murillas, I.; Beaney, M.; Cristofanilli, M.; Andre, F.; Loi, S.; Loibl, S.; et al. Plasma ESR1 Mutations and the Treatment of Estrogen Receptor–Positive Advanced Breast Cancer. J. Clin. Oncol. 2016, 34, 2961–2968. [Google Scholar] [CrossRef]
  6. Lopez-Knowles, E.; Pearson, A.; Schuster, G.; Gellert, P.; Ribas, R.; Yeo, B.; Cutts, R.; Buus, R.; Garcia-Murillas, I.; Haynes, B.; et al. Molecular characterisation of aromatase inhibitor-resistant advanced breast cancer: The phenotypic effect of ESR1 mutations. Br. J. Cancer 2019, 120, 247–255. [Google Scholar] [CrossRef]
  7. Razavi, P.; Chang, M.T.; Xu, G.; Bandlamudi, C.; Ross, D.S.; Vasan, N.; Cai, Y.; Bielski, C.M.; Donoghue, M.T.A.; Jonsson, P.; et al. The Genomic Landscape of Endocrine-Resistant Advanced Breast Cancers. Cancer Cell 2018, 34, 427–438.e6. [Google Scholar] [CrossRef] [Green Version]
  8. Robinson, D.R.; Wu, Y.-M.; 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] [Green Version]
  9. Toy, W.; Shen, Y.; Won, H.; Green, B.; Sakr, R.A.; Will, M.; Li, Z.; Gala, K.; Fanning, S.; King, T.A.; et al. ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat. Genet. 2013, 45, 1439–1445. [Google Scholar] [CrossRef] [Green Version]
  10. Zheng, Z.-Y.; Anurag, M.; Lei, J.T.; Cao, J.; Singh, P.; Peng, J.; Kennedy, H.; Nguyen, N.-C.; Chen, Y.; Lavere, P.; et al. Neurofibromin Is an Estrogen Receptor-α Transcriptional Co-repressor in Breast Cancer. Cancer Cell 2020, 37, 387–402.e7. [Google Scholar] [CrossRef]
  11. Mastoraki, S.; Strati, A.; Tzanikou, E.; Chimonidou, M.; Politaki, E.; Voutsina, A.; Psyrri, A.; Georgoulias, V.; Lianidou, E. ESR1 Methylation: A Liquid Biopsy–Based Epigenetic Assay for the Follow-up of Patients with Metastatic Breast Cancer Receiving Endocrine Treatment. Clin. Cancer Res. 2018, 24, 1500. [Google Scholar] [CrossRef] [Green Version]
  12. Tsuboi, K.; Nagatomo, T.; Gohno, T.; Higuchi, T.; Sasaki, S.; Fujiki, N.; Kurosumi, M.; Takei, H.; Yamaguchi, Y.; Niwa, T.; et al. Single CpG site methylation controls estrogen receptor gene transcription and correlates with hormone therapy resistance. J. Steroid Biochem. Mol. Biol. 2017, 171, 209–217. [Google Scholar] [CrossRef] [PubMed]
  13. Ramos, E.A.S.; Camargo, A.A.; Braun, K.; Slowik, R.; Cavalli, I.J.; Ribeiro, E.M.S.F.; Pedrosa, F.d.O.; de Souza, E.M.; Costa, F.F.; Klassen, G. Simultaneous CXCL12 and ESR1 CpG island hypermethylation correlates with poor prognosis in sporadic breast cancer. BMC Cancer 2010, 10, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Martínez-Galán, J.; Torres, B.; del Moral, R.; Muñoz-Gámez, J.A.; Martín-Oliva, D.; Villalobos, M.; Núñez, M.I.; Luna, J.d.D.; Oliver, F.J.; Almodóvar, J.M.R.d. Quantitative detection of methylated ESR1 and 14-3-3-σ gene promoters in serum as candidate biomarkers for diagnosis of breast cancer and evaluation of treatment efficacy. Cancer Biol. Ther. 2008, 7, 958–965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Mirza, S.; Sharma, G.; Prasad, C.P.; Parshad, R.; Srivastava, A.; Gupta, S.D.; Ralhan, R. Promoter hypermethylation of TMS1, BRCA1, ERα and PRB in serum and tumor DNA of invasive ductal breast carcinoma patients. Life Sci. 2007, 81, 280–287. [Google Scholar] [CrossRef] [PubMed]
  16. Shenker, N.S.; Flower, K.J.; Wilhelm-Benartzi, C.S.; Dai, W.; Bell, E.; Gore, E.; El Bahrawy, M.; Weaver, G.; Brown, R.; Flanagan, J.M. Transcriptional implications of intragenic DNA methylation in the oestrogen receptor alpha gene in breast cancer cells and tissues. BMC Cancer 2015, 15, 337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Widschwendter, M.; Siegmund, K.D.; Müller, H.M.; Fiegl, H.; Marth, C.; Müller-Holzner, E.; Jones, P.A.; Laird, P.W. Association of Breast Cancer DNA Methylation Profiles with Hormone Receptor Status and Response to Tamoxifen. Cancer Res. 2004, 64, 3807. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Stone, A.; Zotenko, E.; Locke, W.J.; Korbie, D.; Millar, E.K.A.; Pidsley, R.; Stirzaker, C.; Graham, P.; Trau, M.; Musgrove, E.A.; et al. DNA methylation of oestrogen-regulated enhancers defines endocrine sensitivity in breast cancer. Nat. Commun. 2015, 6, 7758. [Google Scholar] [CrossRef] [Green Version]
  19. Snyder, M.W.; Kircher, M.; Hill, A.J.; Daza, R.M.; Shendure, J. Cell-free DNA Comprises an In Vivo Nucleosome Footprint that Informs Its Tissues-Of-Origin. Cell 2016, 164, 57–68. [Google Scholar] [CrossRef] [Green Version]
  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] [Green Version]
  21. Heitzer, E.; Ulz, P.; Geigl, J.B. Circulating Tumor DNA as a Liquid Biopsy for Cancer. Clin. Chem. 2015, 61, 112–123. [Google Scholar] [CrossRef] [PubMed]
  22. Merker, J.D.; Oxnard, G.R.; Compton, C.; Diehn, M.; Hurley, P.; Lazar, A.J.; Lindeman, N.; Lockwood, C.M.; Rai, A.J.; Schilsky, R.L.; et al. Circulating Tumor DNA Analysis in Patients With Cancer: American Society of Clinical Oncology and College of American Pathologists Joint Review. J. Clin. Oncol. 2018, 36, 1631–1641. [Google Scholar] [CrossRef] [PubMed]
  23. Hannum, G.; Guinney, J.; Zhao, L.; Zhang, L.; Hughes, G.; Sadda, S.; Klotzle, B.; Bibikova, M.; Fan, J.B.; Gao, Y.; et al. Genome-wide methylation profiles reveal quantitative views of human aging rates. Mol. Cell 2013, 49, 359–367. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Zurita, M.; Lara, P.C.; del Moral, R.; Torres, B.; Linares-Fernández, J.L.; Arrabal, S.R.; Martínez-Galán, J.; Oliver, F.J.; Ruiz de Almodóvar, J.M. Hypermethylated 14-3-3-σ and ESR1 gene promoters in serum as candidate biomarkers for the diagnosis and treatment efficacy of breast cancer metastasis. BMC Cancer 2010, 10, 217. [Google Scholar] [CrossRef] [Green Version]
  25. Müller, H.M.; Widschwendter, A.; Fiegl, H.; Ivarsson, L.; Goebel, G.; Perkmann, E.; Marth, C.; Widschwendter, M. DNA Methylation in Serum of Breast Cancer Patients. Cancer Res. 2003, 63, 7641. [Google Scholar]
  26. Li, H.; Chiappinelli, K.B.; Guzzetta, A.A.; Easwaran, H.; Yen, R.-W.C.; Vatapalli, R.; Topper, M.J.; Luo, J.; Connolly, R.M.; Azad, N.S.; et al. Immune regulation by low doses of the DNA methyltransferase inhibitor 5-azacitidine in common human epithelial cancers. Oncotarget 2014, 5, 587–598. [Google Scholar] [CrossRef]
  27. Kirn, V.; Strake, L.; Thangarajah, F.; Richters, L.; Eischeid, H.; Koitzsch, U.; Odenthal, M.; Fries, J. ESR1-promoter-methylation status in primary breast cancer and its corresponding metastases. Clin. Exp. Metastasis 2018, 35, 707–712. [Google Scholar] [CrossRef]
  28. Sieuwerts, A.M.; Meijer-van Gelder, M.E.; Timmermans, M.; Trapman, A.M.A.C.; Garcia, R.R.; Arnold, M.; Goedheer, A.J.W.; Portengen, H.; Klijn, J.G.M.; Foekens, J.A. How ADAM-9 and ADAM-11 Differentially From Estrogen Receptor Predict Response to Tamoxifen Treatment in Patients with Recurrent Breast Cancer: A Retrospective Study. Clin. Cancer Res. 2005, 11, 7311–7321. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Restoration of ESR1/ER after DAC treatment. (Top): Methylation decrease in ESR1 promotor region A is observed in MDA-MB-436 after DAC treatment by qMSP using the 2−ΔCt method, compared to the ACTB housekeeping gene. (Bottom): Increased ER expression, measured by targeting ESR1 exon 4 mRNA after cDNA synthesis, is observed in MDA-MB-436 after DAC treatment by qPCR using the 2−ΔCt method compared to the average of the PBGD and HPRT1 housekeeping genes. Error bars represent the standard deviation. * ESR1 expression levels were multiplied 250× for scale unification.
Figure 1. Restoration of ESR1/ER after DAC treatment. (Top): Methylation decrease in ESR1 promotor region A is observed in MDA-MB-436 after DAC treatment by qMSP using the 2−ΔCt method, compared to the ACTB housekeeping gene. (Bottom): Increased ER expression, measured by targeting ESR1 exon 4 mRNA after cDNA synthesis, is observed in MDA-MB-436 after DAC treatment by qPCR using the 2−ΔCt method compared to the average of the PBGD and HPRT1 housekeeping genes. Error bars represent the standard deviation. * ESR1 expression levels were multiplied 250× for scale unification.
Ijms 23 05631 g001aIjms 23 05631 g001b
Figure 2. Flow chart of patient samples and healthy donor samples that were included in this study.
Figure 2. Flow chart of patient samples and healthy donor samples that were included in this study.
Ijms 23 05631 g002
Figure 3. (A,B). ESR1 methylation in cfDNA from patients from cohorts A and B. (A). Prevalence of ESR1 methylation in cfDNA is not increased in endocrine-resistant patients; Fisher’s exact test (B). Prevalence of ESR1 methylation in cfDNA increased with age; chi-square test for trend (CF). ESR1 methylation in cfDNA from healthy subjects, and association with age. Data were publicly available and downloaded from Hannum et al. Mol Cell 2013. (C). Beta value of cg15980539, targeted by our ESR1 methylation assay (D). Median beta value of CpG dinucleotides located at promoter A (E). Median beta value of CpG dinucleotides located at promoter B (F). Median beta value of CpG dinucleotides located at promoter C.
Figure 3. (A,B). ESR1 methylation in cfDNA from patients from cohorts A and B. (A). Prevalence of ESR1 methylation in cfDNA is not increased in endocrine-resistant patients; Fisher’s exact test (B). Prevalence of ESR1 methylation in cfDNA increased with age; chi-square test for trend (CF). ESR1 methylation in cfDNA from healthy subjects, and association with age. Data were publicly available and downloaded from Hannum et al. Mol Cell 2013. (C). Beta value of cg15980539, targeted by our ESR1 methylation assay (D). Median beta value of CpG dinucleotides located at promoter A (E). Median beta value of CpG dinucleotides located at promoter B (F). Median beta value of CpG dinucleotides located at promoter C.
Ijms 23 05631 g003
Figure 4. ESR1 exon 1 and positions of ESR1 promoter regions, based on the RefSeq curated ESR1 sequence. The genomic coordinates are relative to the ESR1 startcodon located at chr6:151,807,913 (hg38 reference genome). The red line indicates the location of the qMSP that was used to evaluate the ESR1 methylation status in cohorts 1 and 2.
Figure 4. ESR1 exon 1 and positions of ESR1 promoter regions, based on the RefSeq curated ESR1 sequence. The genomic coordinates are relative to the ESR1 startcodon located at chr6:151,807,913 (hg38 reference genome). The red line indicates the location of the qMSP that was used to evaluate the ESR1 methylation status in cohorts 1 and 2.
Ijms 23 05631 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bos, M.K.; Deger, T.; Sleijfer, S.; Martens, J.W.M.; Wilting, S.M. ESR1 Methylation Measured in Cell-Free DNA to Evaluate Endocrine Resistance in Metastatic Breast Cancer Patients. Int. J. Mol. Sci. 2022, 23, 5631. https://doi.org/10.3390/ijms23105631

AMA Style

Bos MK, Deger T, Sleijfer S, Martens JWM, Wilting SM. ESR1 Methylation Measured in Cell-Free DNA to Evaluate Endocrine Resistance in Metastatic Breast Cancer Patients. International Journal of Molecular Sciences. 2022; 23(10):5631. https://doi.org/10.3390/ijms23105631

Chicago/Turabian Style

Bos, Manouk K., Teoman Deger, Stefan Sleijfer, John W. M. Martens, and Saskia M. Wilting. 2022. "ESR1 Methylation Measured in Cell-Free DNA to Evaluate Endocrine Resistance in Metastatic Breast Cancer Patients" International Journal of Molecular Sciences 23, no. 10: 5631. https://doi.org/10.3390/ijms23105631

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