Estrogen receptor α gene ESR1 amplification may predict endocrine therapy responsiveness in breast cancer patients (2024)

  • Journal List
  • Cancer Sci
  • v.100(6); 2009 Jun
  • PMC11159263

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Estrogen receptor α gene ESR1 amplification may predict endocrine therapy responsiveness in breast cancer patients (1)

Link to Publisher's site

Cancer Sci. 2009 Jun; 100(6): 1012–1017.

Published online 2009 Feb 25. doi:10.1111/j.1349-7006.2009.01145.x

PMCID: PMC11159263

PMID: 19320640

Saori Tomita,1 Zhenhuan Zhang,1 Masahiro Nakano,1 Mutsuko Ibusuki,1 Teru Kawazoe,1 Yutaka Yamamoto,1 and Hirotaka IwaseEstrogen receptor α gene ESR1 amplification may predict endocrine therapy responsiveness in breast cancer patients (2)

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

Estrogen receptor (ER) α plays a crucial role in normal breast development and has also been linked to mammary carcinogenesis and clinical outcome in breast cancer patients. However, the molecular mechanisms controlling the expression of ERα are as yet not fully understood. Gene amplification is one of the important factors regulating protein expression. Recent studies on the amplification of the ESR1 gene, which encodes ERα, have presented conflicting data. Using fluorescence insitu hybridization and real‐time quantitative polymerase chain reaction analysis, we examined the ESR1 status in a series of breast cancer tissues and analyzed its clinical importance. ESR1 gene amplification and gain were found in 22.6 and 11.3% of samples, respectively, as determined by three‐dimensional fluorescence insitu hybridization assay. Moreover, ESR1 amplification and amplification plus gain were significantly negatively correlated with tumor size, number of positive lymph nodes, negative ERα, and positive human epidermal growth factor receptor 2 status. It has also been shown that ESR1 amplification strongly correlates with higher expression levels of ER protein and that patients with ESR1 amplification in their tumors apparently experience longer disease‐free survival than those without. Our data suggest that ESR1 amplification might prove to be helpful in selecting patients who may potentially benefit from endocrine therapy. (Cancer Sci 2009; 100: 1012–1017)

Estrogen receptor (ER) α signaling is known to be necessary for the growth and differentiation of normal breast epithelium, as well as for the initiation and progression of estrogen‐dependent breast cancer. It was reported that the percentage of normal breast epithelial cells staining positive for ERα is generally low,(1) and that expression of the ERα protein fluctuates throughout the menstrual cycle.(2) On the other hand, significantly higher expression has been reported in premalignant breast disease,(3) and the level of ERα expression changes with progression of the disease in individual patients. Clinically, the ERα status of breast cancer patients is widely used both as an indicator for endocrine therapy responsiveness and also for prognosis prediction. However, despite its importance, the molecular mechanisms controlling the expression of ERα are not fully understood.

Nevertheless, it is known that ERα expression is regulated by numerous mechanisms. The silencing of ERα has been identified in breast tumor, including via a mutation within the open reading frame of the ESR1 gene,(4) and via epigenetic changes such as DNA methylation of the promoter‐proximal CpG island in the ESR1 gene.(5, 6) Moreover, it was recently reported that micro‐ribonucleic acid (miRNA)‐206(7, 8) and miRNA‐221/222(9) negatively regulate ERα at the post‐transcriptional level. These abnormalities act alone or in combination to alter the functions or expression levels of ERα.

Gene amplification is one of the important factors regulating protein expression. Many copy number aberrations have been shown to recur across multiple cancer samples. These recurrent copy number aberrations frequently contain oncogenes and tumor‐suppressor genes, and are associated with tumor progression, the clinical course of the cancer, or its responsiveness to therapy.(10) Knowledge of copy number aberration can have an immediate clinical use; for example, an effective diagnosis of the amplification or overexpression of human epidermal growth factor receptor (HER) 2/neu in breast tumors will allow such patients to benefit from treatment with the monoclonal antibody trastuzumab (Herceptin).(11) With respect to ERα, we previously described that loss of heterozygosity of the ER gene does not play an important role in the lack of ER function in breast cancer tissues.(12) On the other hand, some studies have reported amplification of ESR1.(13, 14, 15, 16, 17, 18, 19, 20) For example, Holst and colleagues recently reported that more than 20% of breast cancers harbor genomic amplification of the ESR1 gene. They also found that ESR1 amplification is an indicator for defining a subtype of primary breast cancers that had particularly high ER expression levels and that this subtype might be optimally suited for hormonal therapy.(15) However, contradictory findings on the clinical importance of ESR1 amplification for breast cancer patients have also been reported.(16, 17, 18, 19, 20)

Given the clinical importance of ERα and its gene ESR1 in relation to ERα expression levels, we investigated the status of ESR1 in a series of breast cancer tissues by means of fluorescence insitu hybridization (FISH) and real‐time quantitative polymerase chain reaction (qPCR) analysis, in an attempt to elucidate the relationship between ESR1 status and clinicopathological factors and prognosis of the patients.

Materials and Methods

Patients and breast cancer tissues.  Primary invasive breast carcinoma specimens were obtained by surgical excision from 147 cases of patients at the Department of Breast and Endocrine Surgery, Kumamoto University Hospital (Kumamoto, Japan) between June 2001 and August 2006. FISH staining of ESR1 status was from 133 available cases of paired tissues. These patients were from a consecutive series, and no exclusion criteria were applied. Informed consent was obtained from all patients before surgery. The Ethics Committee of Kumamoto University Graduate School of Medicine (Kumamoto, Japan) approved the study protocol.

The median age of the patients was 62years (range, 30–86years). Postoperative treatment was done in accordance with the recommendations of St Gallen's international consensus meeting for primary breast cancer.(21, 22, 23) On recurrence, patients with ERα‐negative and progesterone receptor‐(PgR) negative tumors were treated with cyclophosphamide+methotrexate+ fluorouracile, fluorouracil+epirubicin+cyclophosphamide, and/or taxanes. Patients with hormone receptor‐positive tumors and non‐visceral metastases were treated with endocrine therapy, such as antiestrogens and aromatase inhibitors. Patients were followed postoperatively every 3months for the first 5years, and every 6months thereafter. The median follow‐up period was 40months (range, 4–87months).

Fluorescence in situ hybridization.  Paraffin‐embedded tissue blocks were sectioned at 4µm thickness and mounted on slides. After deparaffinization and hydration using a commercial SPEC ESR1/CEN6 Dual Color Probe kit (ZytoVision, Bremerhaven, Germany), slides were processed according to the manufacturer's instructions. In brief, the tissue samples on the slides were dewaxed for 10min in an incubator at 70°C, followed by xylene treatment for 10min, twice. After rehydration in a series of graded ethanol solutions followed by rinsing in distilled water, the slides were treated with Pretreatment Solution Citric at 98°C for 15min, and then washed twice with distilled water for 2min.

The samples were treated with pepsin reagent for 15min at 37°C, followed by washing with 2× saline–sodium citrate buffer for 5min and water for 1min. The slides were then dehydrated in a series of graded ethanol solutions and air dried.

Subsequently, the samples were treated with ESR1/CEN‐6 Probe Mix (10µL each) and then a coverslip was added. Finally, the edges of the hybridization area were sealed with rubber cement. For codenaturation of the probe and target DNA, slides were placed at 75°C for 10min and then incubated at 37°C for 14–20h with a Hybridizer instrument (Dako, Glostrup, Denmark).

After hybridization, the slides were washed and dehydrated. Anti‐fading 4′,6‐diamidino‐2‐phenylindole was applied.

Image analysis.  Images were obtained with a fluorescence microscope (DP70; Olympus, Tokyo, Japan) equipped with ×100 UPlan Apo objective lens and image acquisition software (DP Manager; Olympus). For detection of small amplicons, image stacks containing three‐dimensional datasets collected at 1.0–1.5‐µm intervals through the z‐axis were subjected to projections.

In normal cells, the CEN‐6 DNA probe became bound to the centromere area of chromosome 6, and thus one to two red fluorescent signal spots were shown. The ESR1 DNA probe became bound to the ESR1 gene, and thus one to two green fluorescent signals were shown. We classified tissue samples with an ESR1/CEN‐6 ratio greater than 1.0 but less than 2.0 as an ESR1 gain, and those cases with an ESR1/CEN‐6 ratio higher than 2 or with clusters of green signals as indicative of amplification of the ESR1 gene.

Immunohistochemistry.  Immunohistochemical staining was carried out as previously described,(24) on 4‐µm thick tumor sections. Serial sections were prepared from selected blocks and float‐mounted on adhesive‐coated glass slides for ERα, PgR, or HER2 staining. Primary antibodies were monoclonal mouse antihuman ERα antibody (1D5; Dako) at 1:100 dilution, PgR antibody (636; DAKO) at 1:100 dilution, and rabbit antihuman c‐erbB‐2 oncoprotein antibody (Dako) at 1:200 dilution for HER2. The Envision system (EnVision labeled polymer, peroxidase; Dako) was used as the detection system for ERα, PgR, and HER2.

Immunohistochemical scoring.  Immunostained slides were scored after the entire slide had been evaluated by light microscopy. The expression of ERα and PgR was scored by assigning a proportion score and an intensity score according to Allred's procedure.(25) In brief, a proportion score represented the estimated proportion of tumor cells staining positive, as follows: 0, none; 1, <1/100; 2, 1/100–1/10; 3, 1/10–1/3; 4, 1/3–2/3; and 5, >2/3. Any brown nuclear staining in invasive breast epithelium counted toward the proportion score. An intensity score represented the average intensity of the positive cells, as follows: 0, none; 1, weak; 2, intermediate; and 3, strong. The proportion and intensity scores were then added to obtain a total score, which could range from 0 to 8. Tumors with a score of 3 or more were considered to be positive for ERα expression. HER2 immunostaining was evaluated using the same method as used by the HercepTest (Dako). To determine the score of HER2 expression, the membrane staining pattern was estimated and scored on a scale of 0 to 3+. Tumors with scores of 2 or more were considered to be positive for HER2 overexpression.

ESR1 gene amplification detection by real‐time polymerase chain reaction.  Patient and control genomic DNA was extracted using the DNeasy Blood and Tissue Kit (Qiagen, Hilden, Germany) following the manufacturer's protocol. The concentration and purity of the genomic DNA preparations were measured and genomic DNA was stored at 4°C until use.

Probes and primers.  We used the primers and probe for ESR1 as reported by Holst etal.(15) The sequence of the primers and probe were: ESR1 forward primer 5′‐GCCAACGCGCAGGTCTA‐3′ (562–578); ESR1 reverse primer 5′‐GCCGCAGCCTCAGA‐3′ (623–610); and ESR1 TaqMan probe FAM‐CTCCCCTACGGCCCC‐NFQ. The polymerase chain reaction (PCR) product size for ESR1 was 62bp. The final optimized concentration for the ESR1 TaqMan probe and each pair of primers was 250 and 900nM, respectively. RNaseP was chosen as a reference for gene dosage because of its single copy number. Primers and probe for RNaseP were obtained from TaqMan endogenous control kits (4316844; Applied Biosystems).

Real‐time qPCR.  Real‐time qPCR was carried out in a 96‐well optical plate on a 7500 Real‐Time PCR System (Applied Biosystems, Foster City, CA, USA) in a total volume of 20µL, adjusted with sterile water: 4.5µL of DNA (~50ng), 900nM of each ESR1 primer, 10µL of Master Mix (2×), 250nM of ESR1 probe, and 1µL of RNaseP kit (20×). All reactions were carried out in triplicate. Thermal cycling conditions included an initial denaturation at 95°C for 10min, followed by 40 cycles of 15s at 95°C and 60s at 60°C.

Quantitative analysis.  Calculation of the gene copy number was carried out using the comparative Ct method (ΔΔCt) that requires a healthy control sample (diploid) as a calibrator in all amplifications, and the RNaseP gene was coamplified with the ESR1 gene and served as an internal standard.

To use the ΔΔCt method, a validation experiment had to be run to show that the efficiencies of the target and the endogenous control amplifications were approximately equal. Then, we analyzed the samples and calibrated them in triplicate, and in parallel, for ESR1 and RNaseP. ESR1 gene status were defined by the ratio of ESR1 versus RNaseP gene: >2.0 indicated amplification, and between 1.5 and 2.0 indicated a gain.

Statistical analysis.  The χ2‐test for independence was adopted for statistical analysis of associations between ESR1 gene status and clinicopathological factors. A disease‐free survival curve was generated by the Kaplan–Meier method and verified by the log‐rank (Mantel–Cox) test. Differences were considered significant when a P‐value <0.05 was obtained.

Results

ESR1 amplification and gain in relation to clinicopathological factors.  We analyzed the ESR1 gene copy number ratio in 133 primary breast tumors (109 ER‐positive tumors and 24 ER‐negative tumors) by FISH. Of the 133 cases, 30 (22.6%) cases showed amplification for ESR1, and 15 (11.3%) demonstrated ESR1 gene gain.

ESR1 amplification and amplification plus gain were significantly negatively correlated with tumor size (P=0.0025 and P=0.0175, respectively) and the number of positive lymph nodes (P=0.0238 and P=0.0252, respectively), and positively correlated with ERα status (P=0.0035 and P=0.0001, respectively), whereas ESR1 gene amplification plus gain was also significantly negatively correlated with HER2 status (P=0.0256) (Table1).

Table 1

ESR1 amplification and gain in relation to clinicopathological factors

Clinicopathological factorESR1 FISH results
Analyzable (n)Amplification (n (%))P‐valueAmplification +gain (n (%))P
Total13330 (22.6)45 (33.8)
Age (years)
 <50 234 (17.4)0.51465 (21.7)0.1776
 >5011026 (23.6)40 (36.4)
Tumor size (cm)
 <3.0 9528 (29.5)0.0025*38 (40.0)0.0175*
 ≥3.0 382 (5.3)7 (18.4)
No. positive lymph nodes
 0 8223 (28.0)0.0238*33 (40.2)0.0252*
 ≥1 506 (12.0)11 (22.0)
Histological grade
 1 7414 (18.9)0.286023 (31.1)0.4437
 2 2810 (35.7)13 (46.4)
 3 285 (17.9)8 (28.6)
Estrogen receptor
 Positive10930 (27.5)0.0035*45 (41.3)0.0001*
 Negative 240 (0.0)0 (0.0)
Progesterone receptor
 Positive 7918 (22.8)0.939231 (39.2)0.1110
 Negative 5412 (22.2)14 (25.9)
HER2
Negative11929 (24.4)0.144644 (37.0)0.0256*
 Positive 131 (7.7)1 (7.70)

Open in a separate window

*P<0.05 is considered as significant. HER, human epidermal growth factor receptor.

Correlation between ESR1 amplification and ER expression.  To investigate the influence of ESR1 amplification on ER protein levels, we compared ESR1 gene copy number to ER protein expression level by immunohistochemistry using Allred scoring (Table2). Immunohistochemical analysis by Allred was successful in 130/133 (97.7%) breast cancer samples. There was a strong correlation between ESR1 amplification and ERα expression. All tumors with ESR1 amplification showed higher expression levels of ER protein (Allred scores of 7–8); a similar trend was seen with tumors demonstrating ESR1 gain, with 78.5% of these samples showing strong ER expression (Allred scores of 7–8).

Table 2

Comparison of estrogen receptor (ER) amplification and expression

ExpressionESR1 FISH (n)ER immunohistochemistry results (Allred score)
0–2345678%
Normal8627.92.33.54.77.0 9.345.3
Gain14 7.107.107.121.457.1
Amplified30 0000010.090.0

Open in a separate window

FISH, fluorescence insitu hybridization.

Patients with ESR1 amplification in their tumors had better disease‐free survival.  We then examined whether ESR1 amplification in primary breast tumors affected disease‐free survival. In the analysis of disease‐free survival, local recurrences and distant metastases were considered as an event. Among 21 cases of events, there were 16 cases of distant metastases and five of local recurrence.

For this analysis, we stratified the patients into three groups: group 1, tumors with ESR1 amplification; group 2, ER‐positive tumors (Allred scores of 3–8) lacking ESR1 amplification; and group 3, ER‐negative tumors (Allred scores of 0–2). It seems that patients with ESR1 amplification in their tumors experienced longer disease‐free survival than those without ESR1 amplification, even though this difference did not reach statistical significance (Fig.1).

Open in a separate window

Figure 1

Kaplan–Meier analysis of the effect on estrogen receptor (ER) status and ESR1 amplification on disease‐free survival for three groups of cancers: group 1, tumors with ESR1 amplification; group 2, ER‐positive tumors (Allred scores of 3–8) lacking ESR1 amplification; and group 3, ER‐negative tumors (Allred scores of 0–2).

We were unable to analyze the correlation between ESR1 amplification and overall survival analysis because there were no cancer‐related deaths in group 1.

ESR1 amplification by real‐time PCR.  The PCR amplification efficiency for the target gene and the reference gene were determined by constructing a standard curve. The amplification efficiency based on the slopes of the standard curves was 91% for ESR1 and 93% for RNaseP. Because the amplification efficiencies for both genes were approximately equivalent, it was possible to use the comparative Ct method (ΔΔCt) to determine the relative copy number of ESR1. Amplification plots for each triplicate showed near overlap, indicating that the assay was highly reproducible between replicates.

We observed amplification of ESR1 in 2 out of 147 cases (1.4%); in both cases FISH analysis confirmed the amplification. Apart from these two, we found four gain cases, whereas the remainder had a normal ratio of ESR1 to centromere 6 (Fig.2).

Open in a separate window

Figure 2

Frequency distribution of ESR1 amplification ratios in 147 breast cancers analyzed by real‐time quantitative polymerase chain reaction. ESR1 gene status was defined by the ratio of ESR1 versus RNaseP, as follows: >2.0 indicated amplification, between 1.5 and 2.0 indicated gain. The majority of cases had an amplification ratio<1.5, indicating normal copy number of ESR1. Two cases were considered to be amplified by real‐time quantitative polymerase chain reaction.

We investigated the correlation between ESR1 amplification by qPCR and ER status by immunohistochemistry, but no significant results were found (data not shown).

Discussion

In the present study, using FISH, we demonstrated that 30/133 (22.6%) breast cancer samples showed amplification and 15/133 (11.3%) showed gain of ESR1, which is consistent with the findings of Holst etal. that ESR1 gene amplification was frequent in breast cancer.(15) Gene amplification, in general, occurs frequently in breast cancer. Multiple different oncogenes have been described previously as being amplified in breast cancer, including HER2, epidermal growth factor receptor, c‐Myc gene (MYC), cyclin D1 gene (CCND1), and murine double minute 2 gene (MDM2).(26, 27, 28, 29, 30) However, there have been few reports about the frequency of ESR1 gene amplification, even though increased ERα expression levels were found to be associated with breast cancer tumorigenesis and progression. The increased frequency of ESR1 gene amplification reported by Holst etal. has prompted investigations about ESR1 amplification. However, concerning the frequency of ESR1 gene amplification in breast cancer, no consistent conclusions have been drawn.(16, 17, 18, 19, 20) Now, a variety of assays are available for detecting copy number variation (CNV), including Southern blotting, FISH, array‐based comparative genomic hybridization (aCGH) and real‐time qPCR. Southern blotting requires a large amount of genomic DNA and is costly in terms of reagents, work, and time. FISH is considered to be the most precise method for amplification detection,(31) as it is unaffected by the problem of tumor cell heterogeneity (see discussion below). aCGH is a powerful screening tool for detecting DNA copy changes and hence is used to identify cancer‐associated chromosomal aberrations.(32) However, it is more expensive, time‐consuming, and requires special equipment with software. Additionally, aCGH reflects the pattern of changes observed in the dominant clonal populations of a given tumor, which means that changes found in the less prevalent clonal populations are less likely to be identified with this technique.(10) The same applies to real‐time qPCR. However, real‐time qPCR offers several advantages, including speed, sensitivity, reproducibility, and low cost. Furthermore, it is informative due to a large dynamic range of quantification, and the need for very small amounts of starting material. Therefore, we analyzed DNA copy number ratios using real‐time qPCR to confirm the FISH results. We observed, however, a discrepancy between FISH and real‐time qPCR results. We found that only 2/147 (1.4%) samples showed amplification of the ESR1 gene 30/133 (22.6%) using FISH. With regard to ESR1 gene amplification, several studies,(16, 17, 18, 19, 20) have contested the frequency of the phenomenon as reported by Holst etal.(15) The findings from these other studies on ESR1 amplification reported a frequency ranging from only 0 to 10% as detected by a variety of methods, including FISH, aCGH, and real‐time qPCR.(16, 17, 18, 19, 20) The differences between FISH data obtained by various research groups could mainly result from whether manual or automated methodology was applied for scoring hybridization signals. It may also be possible that a proportion of low‐level amplification was missed by automated scoring of FISH signals. Therefore, we carried out manual scoring by taking the image stacks containing three‐dimensional data sets through the z‐axis and analyzed the projection image in order to detect small‐sized amplicons (Fig.3). Analysis using three‐dimensional FISH has proved to be a useful and reliable way to evaluate small nuclear components, such as chromatin arrangement and telomere clustering, as reported previously.(33, 34, 35) Most of the ESR1‐amplified tumors had tiny confluent or small gene clusters of ESR1 (3–10 copies), whereas only 6/30 (20%) cases had large clusters (>10 copies), as observed in the present study (Fig.4). If we had not taken the image stacks into account as described above, the detection rate of ESR1 amplification in our patient population would have been 18/133 (13.5%). On the other hand, it is likely that the inability of array‐CGH and real‐time qPCR to detect CNV was caused by breast tumor heterogeneity. This might have led the normal cells, or the tumor cells without CNV, to dilute the population of mutated tumor cells and thereby lower the average copy number of the population of cells detected. Therefore, further studies using microdissected breast cancer tissue are urgently needed.

Open in a separate window

Figure 3

Image restoration and analyses of three‐dimensional images of the ESR1 locus. (a) A series of images was taken through the z‐axis. (b) Localization of the ESR1 gene (green) and centromere 6 (red) is shown in the two fluorescence insitu hybridization images (z1, z2) and projected one. In a single‐plane image, the ESR1 gene could not be detected, as indicated by an arrowhead in z1 and by an arrow in z2, because of the small size of the amplicon.

Open in a separate window

Figure 4

Examples of fluorescence insitu hybridization findings in breast cancers with (a) normal ESR1 copy number and (b–d) ESR1 amplification. The ESR1 gene probe is labeled in green, the centromere 6 reference probe in red. (a) Two copies of ESR1 per nucleus. (b) Tiny confluent ESR1 gene clusters. (c) Small cluster with three to nine clearly distinguishable ESR1 gene copies. (d) Large clusters of more than 10 gene signals.

We observed that ESR1 amplification was significantly correlated with small tumor size and negative lymph nodes. These data suggest that although ESR1 amplification has a role in the proliferative mechanism of early stage breast cancer, it is not involved in tumor progression. Therefore, it is possible that breast cancer cells with ESR1 amplification have a lower growth potential than those without amplification (which have acquired growth potential via other means), and that such tumor cells with ESR1 amplification will gradually die or be outnumbered by the cells without amplification.

Although the expression status of ERα is a primary determinant in the hormone therapy of breast cancer, it can not reliably predict the responsiveness of the tumors to hormone therapy; not all ERα‐positive breast cancers respond to hormone therapy. This discrepancy between ERα expression status and response to hormonal therapy probably derives from the different estrogen signaling pathway conditions.(36)

Therefore, a new diagnostic marker to screen patients who may potentially benefit from hormone therapy is required. Our results showed that ESR1 amplification was significantly correlated with higher expression levels of ER protein and better disease‐free survival. Considering that most patients with ERα‐positive tumors will have adjuvant antiestrogen treatment, it is likely that ESR1 amplification could be helpful in identifying a subgroup of individuals with breast cancer who would benefit most from the hormone therapy. Further studies in large cohorts of patients are needed to evaluate the predictive value of ESR1 amplification for hormone therapy.

In conclusion, FISH assays appear to be highly sensitive and specific for detecting ESR1 gene amplification, and obtaining images taken through the z‐axis. Our present study has demonstrated ESR1 gene amplification and gain in 22.6 and 11.3% of breast cancer samples, respectively, as determined by FISH assay. ESR1 amplification and amplification plus gain were significant negatively correlated with tumor size, number of positive lymph nodes, and HER2 status, and positively correlated with ERα status. Moreover, it has been shown that ESR1 amplification is strongly correlated with higher expression levels of ER protein and has possibilities as a predictive marker. Our data suggest that ESR1 amplification might prove helpful in selecting patients who may potentially benefit from endocrine therapy.

Acknowledgments

We thank Mrs Noriko Saitoh and Dr Mitsuyoshi Nakao (Department of Regeneration Medicine, Institute of Molecular Embryology and Genetics, Kumamoto University, Japan) for helpful discussions and Ms Yuu Oogaki for her excellent technical support.

References

1. Ricketts D, Turnbull L, Ryall Getal. Estrogen and progesterone receptors in the normal female breast. Cancer Res1991; 51: 1817–22. [PubMed] [Google Scholar]

2. Markopoulos C, Berger U, Wilson P, Gazet JC, Coombes RC. Oestrogen receptor content of normal breast cells and breast carcinomas throughout the menstrual cycle. Br Med J (Clin Res Ed)1988; 296: 1349–51. [PMC free article] [PubMed] [Google Scholar]

3. Allred DC, Mohsin SK, Fuqua SA. Histological and biological evolution of human premalignant breast disease. Endocr Relat Cancer2001; 8: 47–61. [PubMed] [Google Scholar]

4. Herynk MH, Fuqua SA. Estrogen receptor mutations in human disease. Endocr Rev2004; 25: 869–98. [PubMed] [Google Scholar]

5. Iwase H, Omoto Y, Iwata Hetal. DNA methylation analysis at distal and proximal promoter regions of the oestrogen receptor gene in breast cancers. Br J Cancer1999; 80: 1982–6. [PMC free article] [PubMed] [Google Scholar]

6. Parrella P, Poeta ML, Gallo APetal. Nonrandom distribution of aberrant promoter methylation of cancer‐related genes in sporadic breast tumors. Clin Cancer Res2004; 10: 5349–54. [PubMed] [Google Scholar]

7. Adams BD, Furneaux H, White BA. The micro‐ribonucleic acid (miRNA) miR‐206 targets the human estrogen receptor‐alpha (ERα) and represses ERα messenger RNA and protein expression in breast cancer cell lines. Mol Endocrinol2007; 21: 1132–47. [PubMed] [Google Scholar]

8. Kondo N, Toyama T, Sugiura H, Fujii Y, Yamash*ta H.miR‐206 expression is down‐regulated in estrogen receptor alpha‐positive human breast cancer. Cancer Res2008; 68: 5004–8. [PubMed] [Google Scholar]

9. Zhao JJ, Lin J, Yang Hetal. MicroRNA‐221/222 negatively regulates ERalpha and associates with tamoxifen resistance in breast cancer. J Biol Chem2008; 283: 31079–86. [PMC free article] [PubMed] [Google Scholar] Retracted

10. Pinkel D, Albertson DG. Array comparative genomic hybridization and its applications in cancer. Nat Genet2005; 37Suppl (.): S11–17. [PubMed] [Google Scholar]

11. Slamon DJ, Leyland‐Jones B, Shak Setal. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med2001; 344: 783–92. [PubMed] [Google Scholar]

12. Iwase H, Greenman JM, Barnes DM, Bobrow L, Hodgson S, Mathew CG. Loss of heterozygosity of the oestrogen receptor gene in breast cancer. Br J Cancer1995; 71: 448–50. [PMC free article] [PubMed] [Google Scholar]

13. Nembrot M, Quintana B, Mordoh J.Estrogen receptor gene amplification is found in some estrogen receptor‐positive human breast tumors. Biochem Biophys Res Commun1990; 166: 601–7. [PubMed] [Google Scholar]

14. Schuur ER, Weigel RJ. Monoallelic amplification of estrogen receptor‐alpha expression in breast cancer. Cancer Res2000; 60: 2598–601. [PubMed] [Google Scholar]

15. Holst F, Stahl PR, Ruiz Cetal. Estrogen receptor alpha (ESR1) gene amplification is frequent in breast cancer. Nat Genet2007; 39: 655–60. [PubMed] [Google Scholar]

16. Brown LA, Hoog J, Chin SFetal. ESR1 gene amplification in breast cancer: a common phenomenon?Nat Genet2008; 40: 806–7. [PMC free article] [PubMed] [Google Scholar]

17. Horlings HM, Bergamaschi A, Nordgard SHetal. ESR1 gene amplification in breast cancer: a common phenomenon?Nat Genet2008; 40: 807–8. [PubMed] [Google Scholar]

18. Vincent‐Salomon A, Raynal V, Lucchesi C, Gruel N, Delattre O.ESR1 gene amplification in breast cancer: a common phenomenon?Nat Genet2008; 40: 809. [PubMed] [Google Scholar]

19. Reis‐Filho JS, Drury S, Lambros MBetal. ESR1 gene amplification in breast cancer: a common phenomenon?Nat Genet2008; 40: 809–10. [PubMed] [Google Scholar]

20. Adelaide J, Finetti P, Charafe‐Jauffret Eetal. Absence of ESR1 amplification in a series of breast cancers. Int J Cancer2008; 123: 2970–2. [PubMed] [Google Scholar]

21. Goldhirsch A, Glick JH, Gelber RD, Coates AS, Senn HJ. Meeting highlights: International Consensus Panel on the Treatment of Primary Breast Cancer. Seventh International Conference on Adjuvant Therapy of Primary Breast Cancer. J Clin Oncol2001; 19: 3817–27. [PubMed] [Google Scholar]

22. Goldhirsch A, Wood WC, Gelber RD, Coates AS, Thurlimann B, Senn HJ. Meeting highlights: updated international expert consensus on the primary therapy of early breast cancer. J Clin Oncol2003; 21: 3357–65. [PubMed] [Google Scholar]

23. Goldhirsch A, Glick JH, Gelber RD, Coates AS, Thurlimann B, Senn HJ. Meeting highlights: international expert consensus on the primary therapy of early breast cancer 2005. Ann Oncol2005; 16: 1569–83. [PubMed] [Google Scholar]

24. Yamash*ta H, Nishio M, Toyama Tetal. Coexistence of HER2 over‐expression and p53 protein accumulation is a strong prognostic molecular marker in breast cancer. Breast Cancer Res2004; 6: R24–30. [PMC free article] [PubMed] [Google Scholar]

25. Allred DC, Harvey JM, Berardo M, Clark GM. Prognostic and predictive factors in breast cancer by immunohistochemical analysis. Mod Pathol1998; 11: 155–68. [PubMed] [Google Scholar]

26. Slamon DJ, Clark GM, Wong SG, Levin WJ, Ullrich A, McGuire WL. Human breast cancer: correlation of relapse and survival with amplification of the HER‐2/neu oncogene. Science1987; 235: 177–82. [PubMed] [Google Scholar]

27. Ro J, North SM, Gallick GE, Hortobagyi GN, Gutterman JU, Blick M.Amplified and overexpressed epidermal growth factor receptor gene in uncultured primary human breast carcinoma. Cancer Res1988; 48: 161–4. [PubMed] [Google Scholar]

28. Rummukainen JK, Salminen T, Lundin J, Kytola S, Joensuu H, Isola JJ. Amplification of c‐myc by fluorescence insitu hybridization in a population‐based breast cancer tissue array. Mod Pathol2001; 14: 1030–5. [PubMed] [Google Scholar]

29. Hui R, Campbell DH, Lee CSetal. EMS1 amplification can occur independently of CCND1 or INT‐2 amplification at 11q13 and may identify different phenotypes in primary breast cancer. Oncogene1997; 15: 1617–23. [PubMed] [Google Scholar]

30. Courjal F, Cuny M, Rodriguez Cetal. DNA amplifications at 20q13 and MDM2 define distinct subsets of evolved breast and ovarian tumours. Br J Cancer1996; 74: 1984–9. [PMC free article] [PubMed] [Google Scholar]

31. Gray JW, Collins C, Henderson ICetal. Molecular cytogenetics of human breast cancer. Cold Spring Harb Symp Quant Biol1994; 59: 645–52. [PubMed] [Google Scholar]

32. Pollack JR, Sorlie T, Perou CMetal. Microarray analysis reveals a major direct role of DNA copy number alteration in the transcriptional program of human breast tumors. Proc Natl Acad Sci USA2002; 99: 12963–8. [PMC free article] [PubMed] [Google Scholar]

33. Carter KC, Bowman D, Carrington Wetal. A three‐dimensional view of precursor messenger RNA metabolism within the mammalian nucleus. Science1993; 259: 1330–5. [PubMed] [Google Scholar]

34. Cremer M, Von Hase J, Volm Tetal. Non‐random radial higher‐order chromatin arrangements in nuclei of diploid human cells. Chromosome Res2001; 9: 541–67. [PubMed] [Google Scholar]

35. Gotta M, Laroche T, Formenton A, Maillet L, Scherthan H, Gasser SM. The clustering of telomeres and colocalization with Rap1, Sir3, and Sir4 proteins in wild‐type Saccharomyces cerevisiae. J Cell Biol1996; 134: 1349–63. [PMC free article] [PubMed] [Google Scholar]

36. Hayashi SI, Yamaguchi Y.Estrogen signaling pathway and hormonal therapy. Breast Cancer2008; 15: 256–61. [PubMed] [Google Scholar]

Articles from Cancer Science are provided here courtesy of Wiley

Estrogen receptor α gene ESR1 amplification may predict endocrine therapy responsiveness in breast cancer patients (2024)

References

Top Articles
Latest Posts
Article information

Author: Errol Quitzon

Last Updated:

Views: 5965

Rating: 4.9 / 5 (59 voted)

Reviews: 82% of readers found this page helpful

Author information

Name: Errol Quitzon

Birthday: 1993-04-02

Address: 70604 Haley Lane, Port Weldonside, TN 99233-0942

Phone: +9665282866296

Job: Product Retail Agent

Hobby: Computer programming, Horseback riding, Hooping, Dance, Ice skating, Backpacking, Rafting

Introduction: My name is Errol Quitzon, I am a fair, cute, fancy, clean, attractive, sparkling, kind person who loves writing and wants to share my knowledge and understanding with you.