Skip to main content
Erschienen in: BMC Cancer 1/2018

Open Access 01.12.2018 | Research article

Expression of estrogen receptor beta correlates with adverse prognosis in resected pancreatic adenocarcinoma

verfasst von: Hendrik Seeliger, Ioannis Pozios, Gerald Assmann, Yue Zhao, Mario H. Müller, Thomas Knösel, Martin E. Kreis, Christiane J. Bruns

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

The relevance of estrogen receptor (ER) expression in pancreatic ductal adenocarcinoma (PDAC) is largely unknown. Clinical trials targeting ER with selective estrogen receptor modulators in pancreatic cancer did not show any benefit. Here, we analyze the impact of recently characterized ER isoform beta on survival in a cohort of patients with resected PDAC.

Methods

Eighty-four patients having undergone pancreatic resection for PDAC at a single institution were identified. Tissue microarrays were constructed of archival tumor specimens. The expression of ER beta was determined by immunohistochemistry and quantified by a system established for estrogen receptor expression in breast cancer. ER beta expression was then correlated with clinicopathological parameters, and univariate and multivariate survival analyses were performed.

Results

Nuclear expression of ER beta was found in 31% of tumors. No significant correlation was found between ER beta expression and TNM status, tumor grade, age or sex. Univariate analysis revealed nodal metastasis and the expression of ER beta as factors correlating with a shorter overall survival and disease free survival. When comparing ER beta expression in patients surviving more than 24 months with those who died from the tumor within 12 or 24 months, respectively, a significantly lower ER beta expression was found in the long term survivors. In multivariate analysis, ER beta expression was demonstrated to be an independent predictor of shorter overall survival.

Conclusions

In resected PDAC, expression of ER beta seems to correlate with poor prognosis. These data may help to identify patients who may benefit from additional systemic therapy including selective estrogen receptor modulators.
Abkürzungen
CI
Confidence interval
CRT
Chemoradiotherapy
CTX
Chemotherapy
DFS
Disease free survival
DP
Distal pancreatectomy
EGF
Epidermal growth factor
EGFR
Epidermal growth factor receptor
ER
Estrogen receptor
MAP kinase
Mitogen activated protein kinase
OS
Overall survival
PD
Partial pancreatoduodenectomy
PDAC
Pancreatic ductal adenocarcinoma
PPPD
pylorus preserving partial pancreatoduodenectomy
TMA
Tissue microarray
TP
Total pancreatectomy

Background

Pancreatic ductal adenocarcinoma (PDAC) is among the leading causes of cancer-related mortality in western countries [1]. In the last decade, overall survival improved only marginally. To date, standard therapeutic regimens consist of surgery, cytotoxic chemotherapy, irradiation, or a combination [2, 3] and result in an overall 5 year survival of less than 10 %. More recently, newer agents targeted against molecular determinants of cancer cells or tumor vessels, or both, have been tested in clinical trials to expand the therapeutic armamentarium [4, 5]. When characterizing molecular targets for potential prognostic and therapeutic use, differences in the sex distribution have led to the investigation of the role of estrogen receptors (ER) in the development and progression of pancreatic cancer and other malignancies [611]. Most early trials with the selective estrogen receptor modulator tamoxifen in PDAC yielded only a moderate survival benefit while showing an acceptable safety profile [1214]. This concept of inhibition of ER mediated effects however did not take into account recently reported existence of differential signaling of ER isoforms ERα and ERβ [1517].
Human ERβ was cloned in 1996 for the first time and subsequently was shown to have a ligand binding specificity and a signaling response to estrogen agonists that is distinct from ERα [1518]. While ERα was demonstrated to promote tumor growth and angiogenesis in breast cancer and many other solid tumor types, the role of ERβ is defined much less clearly.
In ERα negative breast cancer specimens, ERβ was shown to correlate with a higher proliferation index [9]. Furthermore, in breast cancer patients, ERβ was characterized as a response marker of the selective estrogen receptor modulator tamoxifen in unselected cohorts and in patients negative for ERα [19, 20]. In non small cell lung cancer patients results are controversial. While in one study, high ERβ expression served as a negative prognostic marker and correlated with a worse outcome [21], a metaanalysis failed to find a consistent correlation of ERβ expression with survival [22]. Beside ERβ-specific effects, ERβ activation seems to interfere with EGF receptor signaling via an activation of the MAP kinase [10, 23].
While pancreatic cancer cell lines were reported to express ERβ [24], there is no consistent information available on the expression of ERβ in human pancreatic ductal adenocarcinoma specimens and its correlation with histopathological parameters and prognostic consequences [8]. The present study was designed to analyze the influence of ERβ expression on overall and disease free survival in PDAC. Here, in a cohort of 94 patients having undergone a pancreatic resection, we correlate ERβ expression in a tissue microarray derived from intraoperative tumor specimens with clinicopathological and survival parameters.

Methods

Patients

We identified 111 consecutive patients from a prospective database of patients operated for ductal pancreatic adenocarcinoma at a single institution. Of these, clinicopathological information and prospectively collected follow up data as well as archived tumor material were available for evaluation in 84 patients. Patients with distant metastases were excluded from the study as well as patients who died within 30 days after resection. Specifically, information on age, sex, date of the primary surgery, perioperative irradiation or chemotherapy, TNM tumor status and grading, date last seen, date of death, cause of death, and date of the first identification of tumor progression were extracted from the original patient charts and a regional tumor registry database. The study was approved by the Ethics Committee of the Hospital of the University of Munich. Due to the retrospective nature of the study, explicit consent was not required.

Tissue microarray construction

Paraffin embedded archive tissue material of tumor and surrounding normal pancreatic tissue was used to generate tissue microarrays (TMAs) after confirming the histological diagnosis of PDAC by a pathologist blinded for the clinical data.
TMAs were prepared essentially as published before [25]. In brief, the area of interest to be sampled was identified and marked on an areal slide corresponding to each paraffin block. Three tissue core biopsies, each 0.6 mm in diameter, were punched out of the donor paraffin block and then arrayed in each of the respective recipient TMA blocks using a manual arrayer (Beecher Instruments, Sun Prairie, WI). Edge confusion was ensured by incubating the TMAs at 37 °C for 30 min. Sections of 2 μm thickness were cut onto adhesive glass slides (Super Frost Plus, Menzel, Braunschweig, Germany).

Immunohistochemistry

Immunohistochemistry for ERβ was performed using standard technique. Briefly, after deparaffinization and rehydration, slides were blocked with bovine serum albumin. The primary antibody (Rabbit polyclonal to estrogen receptor beta, Abcam, Cambridge, UK), was added in a dilution of 1:200 and incubated overnight at 4 °C. After blocking endogenous peroxidase with 7.5% hydrogen peroxide, a horseradish-peroxidase conjugated polyclonal goat anti-rabbit secondary antibody (Dako, Hamburg, Germany) was added and incubated for 30 min at room temperature. Slides were counterstained with hematoxylin.

Histopathological evaluation

ERβ expression was quantified analogous to the scoring system proposed by Remmele and Stegner used for ERα in breast cancer [26]. Briefly, staining intensity was scored from 0 (no reaction) to 3 (strong reaction), and the percentage of stained nuclei was scored from 0 (no positive nuclei) to 4 (more than 80% positive nuclei). The scores of staining intensity and stained nuclei were multiplied, yielding a total core of 0 to 12. Positive expression of ERβ was defined as a score of 3 or more. Scoring was performed by two independent pathologists blinded for the clinical data.

Statistical analysis

Statistical analyses were performed by utilizing IBM SPSS statistics 23 software package (IBM, Armonk, NY). Chi-square tests were applied to test correlation between categorical variables. Survival curves were calculated according to Kaplan-Meier, with differences in survival between strata of low and high ERβ expression and clinicopathological parameters detected by the log-rank test. Multivariate analysis was performed using the Cox proportional hazards model including variables with a p value of less than 0.15 in univariate analyses. A p value of less than 0.05 was considered statistically significant.

Results

Demographic data

The study cohort consisted of 84 patients, 41 men and 43 women with a median age of 65.6 years at the time of the operation (range 32–82 years). Demographic and clinicopathological characteristics of the patients are summarized in Table 1. At the time of the analysis, 63 patients (75.0%) had died from the tumor, and three more patients had tumor progression.
Table 1
Clinicopathological parameters of 84 patients with resected pancreatic ductal adenocarcinoma
Variable
 
n
%
Sex
female
43
51.2
male
41
48.8
Age
≤60 years
24
28.6
> 60 years
60
71.4
Type of operation
PD
35
41.7
PPPD
34
40.5
Distal pancreatectomy
10
11.9
TP
5
6.0
T status
T1
1
1.2
T2
10
11.9
T3
68
81.0
T4
5
6.0
N status
N0
35
41.7
N1
49
58.3
Residual tumora
R0
39
48.1
R1
42
51.9
Histological grading
G1
2
2.4
G2
28
33.3
G3
54
64.3
Perioperative therapy
Chemotherapy
5
6.0
Chemoradiation
49
58.3
none
30
35.7
PD partial pancreatoduodenectomy (Kausch-Whipple procedure), PPPD pylorus preserving partial pancreatoduodenectomy (Traverso-Longmire procedure), DP distal pancreatectomy, TP total pancreatectomy
amissing information on resection status in three patients

Expression of ERβ

A nuclear expression of the ERβ was detected in 26 PDAC tumor specimens (31.0%). Representative slides are shown in Fig. 1. No correlation was seen between ER expression and other clinicopathological parameters, such as sex, age, T and N stage, and histological grading. Furthermore, additional therapy (chemotherapy or chemoradiation) and ER expression did not correlate (Table 2). Interestingly, in adjacent normal pancreatic tissue, ER beta expression was detected in 41 patients (48.8%). A downregulation of ER beta expression in tumor tissue, compared to normal tissue, as defined by a lower staining score, was seen in 42 cases (50.0%).
Table 2
Correlation of estrogen receptor beta (ERβ) expression with clinicopathological parameters
Variable
n
ERβ expression
P value
Total
84
26 (31.0%)
 
Sex
female
43
16 (37.2%)
0.243
male
41
10 (24.4%)
Age
≤60 years
24
7 (29.2%)
1.000
> 60 years
60
19 (31.7%)
T status
1–2
11
3 (27.3%)
1.000
3–4
73
23 (31.5%)
N status
0
35
11 (31.4%)
1.000
1
49
15 (30.6%)
Tumor grade
1–2
30
7 (23.3%)
0.328
3
54
19 (35.2%)
Residual tumora
R0
39
11 (28.2%)
0.487
 
R1
42
15 (35.7%)
 
Perioperative therapy
Surgery alone
30
13 (43.3%)
0.086
Chemoradiation/Chemotherapy
54
13 (24.1%)
amissing information on resection status in three patients

Univariate survival analysis

Mean overall survival of all patients after resection of the primary tumor was 27.0 months (95% confidence interval 22.3–31.6 months), and mean disease free survival was 21.2 months (95% confidence interval 17.2–25.1 months). Patients with ERβ expressing tumors survived 16.6 months compared to 30.9 months in patients without ERβ expression (p = 0.009, Fig. 2a). Disease free survival was 13.5 months in patients with ERβ expression compared to 23.5 months in patients with no ERβ expression (p = 0.037, Fig. 2b). Overall survival in nodal positive patients was 21.5 months versus 33.1 months in nodal negative patients (p = 0.021, Fig. 2a). Disease free survival was 18.5 months in nodal positive patients which was significantly shorter compared to 25.1 months in patients with negative nodal status (p = 0.066, Fig. 2b). Details are given in Table 3 and Table 4. In long term survivors (overall survival 24 months and more, n = 25) ERβ expression was detected in 25%, while patients who survived less than 12 months (n = 25) showed ERβ expression in 44% (Fig. 3, p = 0.0027).
Table 3
Univariate analysis of prognostic factors for overall survival in resected pancreatic ductal adenocarcinoma
Variable
 
Mean OS [months]
95% CI
Median OS [months]
p value
Total
 
27.0
22.3
31.6
21.4
 
Sex
female
23.1
17.7
28.5
16.3
0.122
male
30.2
22.5
37.9
34.2
 
Age
≤60 years
29.4
21.6
37.1
21.4
0.398
> 60 years
25.4
20.1
30.8
20.2
 
T status
T1–2
26.2
15.2
37.2
17.0
0.969
T3–4
26.8
21.8
31.9
22.2
 
N status
N0
33.1
26.1
40.2
34.2
0.021
N1
21.5
16.1
26.8
16.3
 
Tumor grading
G1–2
28.1
21.3
34.9
36.8
0.228
G3
25.3
19.6
31.0
16.7
 
Residual tumor
R0
31.4
24.0
38.8
34.2
0.052
R1
19.8
15.7
23.9
16.7
 
ERβ expression
negative
30.9
25.2
36.7
34.2
0.009
positive
16.6
12.7
20.4
12.2
 
Perioperative
CRT/CTX
25.5
18.5
 
32.5
17.4
0.800
therapy
none
27.2
21.5
 
32.9
21.4
 
CI confidence interval, OS overall survival, CRT chemoradiotherapy, CTX chemotherapy
Table 4
Univariate analysis of prognostic factors for disease free survival in resected pancreatic ductal adenocarcinoma
Variable
 
Mean DFS [months]
95% CI
Median DFS [months]
p value
Total
 
21.2
17.2
25.1
15.0
 
Sex
female
20.4
15.2
25.6
12.2
0.485
male
22.0
16.1
27.9
16.3
 
Age
≤60 years
22.4
15.0
29.8
16.1
0.704
> 60 years
20.5
15.8
25.2
15.0
 
T status
T1–2
25.6
13.5
37.7
16.3
0.701
T3–4
20.7
16.4
25.0
15.0
 
N status
N0
25.1
18.9
31.2
20.0
0.066
N1
18.5
13.6
23.5
13.0
 
Tumor grading
G1–2
24.4
17.3
31.6
20.1
0.293
G3
19.5
14.8
24.1
14.8
 
Residual tumor
R0
25.9
19.4
32.5
21.5
0.018
R1
15.1
11.7
18.5
13.3
 
ERβ expression
negative
23.5
18.7
28.4
16.3
0.037
positive
13.5
9.6
17.4
8.7
 
Perioperative
CRT/CTX
21.4
16.5
26.3
15.0
0.800
therapy
none
20.4
13.5
27.2
16.7
 
CI confidence interval, DFS disease free survival, CRT chemoradiotherapy, CTX chemotherapy

Multivariate survival analysis

To validate ERβ expression as an independent prognostic indicator in PDAC on overall survival, multivariate regression analysis was performed. Expression of ERβ was demonstrated to be an independent prognostic indicator of overall survival (hazard ratio 1.938, p = 0.047). Of the remaining variables tested only positive nodal status showed a trend towards adverse survival however did not become statistically significant (hazard ratio 1.831, p = 0.069). Male sex and residual tumor status also failed to show statistical significance in the multivariate survival analysis. Details are shown in Table 5.
Table 5
Multivariate analysis of prognostic factors for overall survival in pancreatic ductal adenocarcinoma
Parameter
Hazard ratio
95% CI
p value
ERβ expression
1.938
1.010–3.720
0.047
N1
1.831
0.954–3.517
0.069
Residual tumor
1.704
0.894–3.247
0.105
Male sex
0.628
0.355–1.311
0.251
CI confidence interval

Discussion

In the present study, ERβ was expressed on PDAC in 31% of all patients. Expression of ERβ did not correlate with any of the clinicopathological parameters examined, however ERβ expression was strongly associated with an adverse overall survival and disease free survival in univariate analyses. Multivariate analysis showed that ERβ expression on tumor cells was an independent prognostic factors of overall survival.
To our knowledge, this study is the largest series on expression of ER on pancreatic neoplasms. The fact that ERα is not detectable with immunohistochemical methods on PDAC tissue is concordant with several other smaller studies [27, 28]. However, there are two studies which showed ERα expression on mRNA level on PDAC [29, 30]. Whether this finding reflects ERα protein levels being expressed in very small amounts not detectable with immunohistochemical methods, or a missing translation of ERα mRNA in PDAC is unknown. Interestingly, mucinous cystic tumors seem to express ERα more frequently than PDAC, possibly reflecting the “ovarian-type stroma” defining mucinous cystic tumors [31, 32]. Data on ERα expression in this entity is still pending.
ERβ was however expressed in nontumorous tissue, and to a lesser extent in the corresponding PDAC specimens. Compared to normal pancreatic tissue, an overall loss of ERβ expression in PDAC was detected in the majority of the investigated cases, suggesting ERβ loss as a molecular event in the line of tumor progression. Yet, the presence of ERβ expression in the tumor correlates with an adverse prognosis. This phenomenon may be explained by a crosstalk of ERβ signal transduction and other pathways that are activated during of tumor progression, leading to a more aggressive tumor phenotype in those subjects with an unchanged ERβ signaling pathway. In fact, non-ligand dependent ER signaling is well characterized. The ligand dependent pathway of ER signaling is initiated by steroid ligand binding to the ER. In the non-ligand dependent pathway, activated kinase growth factor receptors phosphorylate ER, leading to its activation [3336]. Loss of ERβ expression during tumor progression was described in several other tumor entities [6, 37, 38]. Seemingly, interplay between ERβ expression and tissue specific distribution of growth factors may be important for subsequent tumor progression.
In colon cancer cell lines, ERβ was shown to be the predominant ER, whereas ERβ mRNA expression was a lot lower and similar to normal tissue [6]. Similar results were obtained when examining ERβ expression in human tumor samples [37, 39].
No significant correlation was found between ERβ expression and clinicopathological features of patients and PDAC specimens. A trend was seen towards a lower ERβ expression in male patients, but statistical significance was not reached. Circulating estrogen levels may have some effect on ERβ expression. In fact, an upregulation of ERβ by estrogen has been described previously [40]. However, since the majority of the female patients in our cohort is postmenopausal, as reflected by age distribution, this effect must be regarded as questionable. Similarly, a trend was detected towards a higher expression of ERβ in less differentiated tumors, underlining a role of ERβ in tumor progression towards a more aggressive phenotype. This finding is supported by recent data in breast cancer, where ERβ expression was found to correlate with tumor grading and higher expression of the proliferation marker Ki-67 in women with ERα negative breast cancer [9]. Similarly, in esophageal cancer, a correlation of ERβ expression with poor differentiation status and tumor stage was found in squamous cell carcinomas and in adenocarcinomas [41].
In the analyzed cohort, ERβ expression was found to strongly correlate with a reduction of overall survival and disease free survival in patients with resected pancreatic adenocarcinoma in univariate and multivariate analyses. Multivariate analysis of overall survival, revealed that the expression of ERβ is an independent negative prognostic factor. Patients with lymph node metastases had a shorter survival, although this did not reach statistical significance. Interestingly, resection status (R0 versus R1) also failed to be a statistically significant prognostic factor of overall survival and disease free survival in the multivariate analysis. This finding may be partially explained by underestimation of the number of R1 resections [42], as examination of circumferential resection margin was not incorporated in routine pathological protocol when specimens were analyzed originally.
At present, clinical data on the impact of ER expression on solid tumors on survival except in breast cancer is not sufficient to establish a clear prognostic role of the different ER subtypes. In esophageal squamous cell carcinoma and gastric adenocarcinoma, expression of ERα in the absence of ERβ was described to correlate with an adverse prognosis [38, 43]. In colorectal cancer, loss of ERβ expression correlates with advanced cancer stages and poor survival [44]. In hepatocellular cancer, both ERα and ERβ are expressed [45]. Presence of a variant ERα in hepatocellular carcinoma correlates with shorter survival, compared to wild type ER [46].
However, some clinical and experimental data support the hypothesis that ERβ expression may lead to a more aggressive tumor phenotype. In ERα negative breast cancer, ERβ expression correlates with an increased Ki-67 expression, suggesting a higher proliferation rate within the tumor cells. In the same cohort, ERβ expression positively correlated with advanced tumor grade [9]. Similar findings were described in sarcoma patients [11]. In vitro proliferation of non small cell lung cancer cells was reduced by siRNA mediated elimination of ERβ signaling [10]. In contrast, ERβ deficiency results in an enhanced tumorigenesis in the small bowel, but not in the colon of Apc (min/+) mice, suggesting a tumor suppressor effect of ERβ [47].
The exact role of ER signaling in solid tumors remains to be defined. Our data strongly suggest a tumor promoting role of ERβ signaling in PDAC, which is in line with previously published data on non small cell lung cancer [21, 48]. However, there are studies showing an effect of ERβ on tumor suppression, especially in colon cancer [4951]. These conflicting results may be explained by differences in the tissue distribution of the ER subtypes and their splicing variants. Moreover, ER signaling is embedded in a complex signaling network controlling tumor cell growth and proliferation to the effect that context specific signaling interactions lead to different effects in different tissue types [52]. In the present study, the phosphorylation status of ERβ was not examined. One can speculate that differences of tissue specific ERβ phosphorylation lead to differential ER mediated actions that are mediated by ligand independent ER signaling. Specifically, an extensive crosstalk between epidermal growth factor receptor (EGFR) and ER mediated pathways is well documented in several tumor types [40]. Since a strong EGFR expression is present in PDAC, ER phosphorylation by EGFR mediated growth signals and the resulting proliferative stimulus may be an important contributor to the adverse prognostic effects observed. In fact, EGFR inhibition combined with ER signaling disruption resulted in a marked inhibition of tumor xenograft growth [10].
A limitation of the present study is the different perioperative treatment of the patients included. In the cohort presented, 58% of the patients received perioperative chemoradiation. Although a clear survival benefit of perioperative chemoradiation in patients with resectable PDAC has not been shown [53], there is a possible impact of irradiation and/or chemotherapy on ER expression [54]. However, whether this is true for the ERβ subtype and is unknown, and its clinical significance remains unclear. To clarify this issue, additional studies may correlate the expression of ERβ on pretherapeutic tumor biopsies with the expression on surgical specimens after neoadjuvant therapy.

Conclusions

Here, the expression of ERβ was analyzed on surgical specimens of patients with PDAC and correlated with overall and disease free survival. ERβ was expressed on 31% of PDAC surgical specimens. A correlation between ERβ expression and an adverse prognosis in resected PDAC seems to exist. These data may be useful in defining a role of ERβ expression as a prognostic indicator and as a potential molecular target in patients with advanced PDAC.

Acknowledgements

The authors thank Andrea Sendelhofert for excellent technical assistance in constructing and staining the tissue microarrays.

Funding

HS performed the study design, data collection, analysis and interpretation and wrote the manuscript while on a scientific rotation position funded by the Deutsche Forschungsgemeinschaft (DFG, KFO128). Further, the authors acknowledge support from the DFG and the Open Access Publication Fund of Charité – Universitätsmedizin Berlin.

Availability of data and materials

The datasets used and analysed during the study are available from the corresponding author on reasonable request.
The study was approved by the Ethics Committee of the Hospital of the University of Munich. Due to the retrospective nature of the study, explicit consent was not required.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.CrossRef Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67(1):7–30.CrossRef
2.
Zurück zum Zitat Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73–85.CrossRef Kamisawa T, Wood LD, Itoi T, Takaori K. Pancreatic cancer. Lancet. 2016;388(10039):73–85.CrossRef
3.
Zurück zum Zitat Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. New Engl J Med. 2014;371(11):1039–49.CrossRef Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. New Engl J Med. 2014;371(11):1039–49.CrossRef
4.
Zurück zum Zitat Chiaravalli M, Reni M, O'Reilly EM. Pancreatic ductal adenocarcinoma: state-of-the-art 2017 and new therapeutic strategies. Cancer Treat Rev. 2017;60:32–43.CrossRef Chiaravalli M, Reni M, O'Reilly EM. Pancreatic ductal adenocarcinoma: state-of-the-art 2017 and new therapeutic strategies. Cancer Treat Rev. 2017;60:32–43.CrossRef
5.
Zurück zum Zitat Ottaiano A, Capozzi M, De Divitiis C, De Stefano A, Botti G, Avallone A, Tafuto S. Gemcitabine mono-therapy versus gemcitabine plus targeted therapy in advanced pancreatic cancer: a meta-analysis of randomized phase III trials. Acta Oncol. 2017;56(3):377–83.CrossRef Ottaiano A, Capozzi M, De Divitiis C, De Stefano A, Botti G, Avallone A, Tafuto S. Gemcitabine mono-therapy versus gemcitabine plus targeted therapy in advanced pancreatic cancer: a meta-analysis of randomized phase III trials. Acta Oncol. 2017;56(3):377–83.CrossRef
6.
Zurück zum Zitat Campbell-Thompson M, Lynch IJ, Bhardwaj B. Expression of estrogen receptor (ER) subtypes and ERbeta isoforms in colon cancer. Cancer Res. 2001;61(2):632–40.PubMed Campbell-Thompson M, Lynch IJ, Bhardwaj B. Expression of estrogen receptor (ER) subtypes and ERbeta isoforms in colon cancer. Cancer Res. 2001;61(2):632–40.PubMed
7.
Zurück zum Zitat Martineti V, Picariello L, Tognarini I, Carbonell Sala S, Gozzini A, Azzari C, Mavilia C, Tanini A, Falchetti A, Fiorelli G, et al. ERbeta is a potent inhibitor of cell proliferation in the HCT8 human colon cancer cell line through regulation of cell cycle components. Endocr Relat Cancer. 2005;12(2):455–69.CrossRef Martineti V, Picariello L, Tognarini I, Carbonell Sala S, Gozzini A, Azzari C, Mavilia C, Tanini A, Falchetti A, Fiorelli G, et al. ERbeta is a potent inhibitor of cell proliferation in the HCT8 human colon cancer cell line through regulation of cell cycle components. Endocr Relat Cancer. 2005;12(2):455–69.CrossRef
8.
Zurück zum Zitat Satake M, Sawai H, Go VL, Satake K, Reber HA, Hines OJ, Eibl G. Estrogen receptors in pancreatic tumors. Pancreas. 2006;33(2):119–27.CrossRef Satake M, Sawai H, Go VL, Satake K, Reber HA, Hines OJ, Eibl G. Estrogen receptors in pancreatic tumors. Pancreas. 2006;33(2):119–27.CrossRef
9.
Zurück zum Zitat Skliris GP, Leygue E, Curtis-Snell L, Watson PH, Murphy LC. Expression of oestrogen receptor-beta in oestrogen receptor-alpha negative human breast tumours. Br J Cancer. 2006;95(5):616–26.CrossRef Skliris GP, Leygue E, Curtis-Snell L, Watson PH, Murphy LC. Expression of oestrogen receptor-beta in oestrogen receptor-alpha negative human breast tumours. Br J Cancer. 2006;95(5):616–26.CrossRef
10.
Zurück zum Zitat Marquez-Garban DC, Chen HW, Fishbein MC, Goodglick L, Pietras RJ. Estrogen receptor signaling pathways in human non-small cell lung cancer. Steroids. 2007;72(2):135–43.CrossRef Marquez-Garban DC, Chen HW, Fishbein MC, Goodglick L, Pietras RJ. Estrogen receptor signaling pathways in human non-small cell lung cancer. Steroids. 2007;72(2):135–43.CrossRef
11.
Zurück zum Zitat Greenberg JA, Somme S, Russnes HE, Durbin AD, Malkin D. The estrogen receptor pathway in rhabdomyosarcoma: a role for estrogen receptor-beta in proliferation and response to the antiestrogen 4’OH-tamoxifen. Cancer Res. 2008;68(9):3476–85.CrossRef Greenberg JA, Somme S, Russnes HE, Durbin AD, Malkin D. The estrogen receptor pathway in rhabdomyosarcoma: a role for estrogen receptor-beta in proliferation and response to the antiestrogen 4’OH-tamoxifen. Cancer Res. 2008;68(9):3476–85.CrossRef
12.
Zurück zum Zitat Taylor OM, Benson EA, McMahon MJ. Clinical trial of tamoxifen in patients with irresectable pancreatic adenocarcinoma. The Yorkshire Gastrointestinal Tumour Group. Br J Surg. 1993;80(3):384–6.CrossRef Taylor OM, Benson EA, McMahon MJ. Clinical trial of tamoxifen in patients with irresectable pancreatic adenocarcinoma. The Yorkshire Gastrointestinal Tumour Group. Br J Surg. 1993;80(3):384–6.CrossRef
13.
Zurück zum Zitat Tomao S, Romiti A, Massidda B, Ionta MT, Farris A, Zullo A, Brescia A, Santuari L, Frati L. A phase II study of gemcitabine and tamoxifen in advanced pancreatic cancer. Anticancer Res. 2002;22(4):2361–4.PubMed Tomao S, Romiti A, Massidda B, Ionta MT, Farris A, Zullo A, Brescia A, Santuari L, Frati L. A phase II study of gemcitabine and tamoxifen in advanced pancreatic cancer. Anticancer Res. 2002;22(4):2361–4.PubMed
14.
Zurück zum Zitat Wong A, Chan A. Survival benefit of tamoxifen therapy in adenocarcinoma of pancreas. A case-control study. Cancer. 1993;71(7):2200–3.CrossRef Wong A, Chan A. Survival benefit of tamoxifen therapy in adenocarcinoma of pancreas. A case-control study. Cancer. 1993;71(7):2200–3.CrossRef
15.
Zurück zum Zitat Mosselman S, Polman J, Dijkema R. ER beta: identification and characterization of a novel human estrogen receptor. FEBS Lett. 1996;392(1):49–53.CrossRef Mosselman S, Polman J, Dijkema R. ER beta: identification and characterization of a novel human estrogen receptor. FEBS Lett. 1996;392(1):49–53.CrossRef
16.
Zurück zum Zitat Kuiper GG, Gustafsson JA. The novel estrogen receptor-beta subtype: potential role in the cell- and promoter-specific actions of estrogens and anti-estrogens. FEBS Lett. 1997;410(1):87–90.CrossRef Kuiper GG, Gustafsson JA. The novel estrogen receptor-beta subtype: potential role in the cell- and promoter-specific actions of estrogens and anti-estrogens. FEBS Lett. 1997;410(1):87–90.CrossRef
17.
Zurück zum Zitat Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science. 1997;277(5331):1508–10.CrossRef Paech K, Webb P, Kuiper GG, Nilsson S, Gustafsson J, Kushner PJ, Scanlan TS. Differential ligand activation of estrogen receptors ERalpha and ERbeta at AP1 sites. Science. 1997;277(5331):1508–10.CrossRef
18.
Zurück zum Zitat Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997;138(3):863–70.CrossRef Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, Gustafsson JA. Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology. 1997;138(3):863–70.CrossRef
19.
Zurück zum Zitat Gruvberger-Saal SK, Bendahl PO, Saal LH, Laakso M, Hegardt C, Eden P, Peterson C, Malmstrom P, Isola J, Borg A, et al. Estrogen receptor beta expression is associated with tamoxifen response in ERalpha-negative breast carcinoma. Clin Cancer Res. 2007;13(7):1987–94.CrossRef Gruvberger-Saal SK, Bendahl PO, Saal LH, Laakso M, Hegardt C, Eden P, Peterson C, Malmstrom P, Isola J, Borg A, et al. Estrogen receptor beta expression is associated with tamoxifen response in ERalpha-negative breast carcinoma. Clin Cancer Res. 2007;13(7):1987–94.CrossRef
20.
Zurück zum Zitat Hopp TA, Weiss HL, Parra IS, Cui Y, Osborne CK, Fuqua SA. Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer. Clin Cancer Res. 2004;10(22):7490–9.CrossRef Hopp TA, Weiss HL, Parra IS, Cui Y, Osborne CK, Fuqua SA. Low levels of estrogen receptor beta protein predict resistance to tamoxifen therapy in breast cancer. Clin Cancer Res. 2004;10(22):7490–9.CrossRef
21.
Zurück zum Zitat Skjefstad K, Grindstad T, Khanehkenari MR, Richardsen E, Donnem T, Kilvaer T, Andersen S, Bremnes RM, Busund LT, Al-Saad S. Prognostic relevance of estrogen receptor alpha, beta and aromatase expression in non-small cell lung cancer. Steroids. 2016;113:5–13.CrossRef Skjefstad K, Grindstad T, Khanehkenari MR, Richardsen E, Donnem T, Kilvaer T, Andersen S, Bremnes RM, Busund LT, Al-Saad S. Prognostic relevance of estrogen receptor alpha, beta and aromatase expression in non-small cell lung cancer. Steroids. 2016;113:5–13.CrossRef
22.
Zurück zum Zitat Ma L, Zhan P, Liu Y, Zhou Z, Zhu Q, Miu Y, Wang X, Jin J, Li Q, Lv T, et al. Prognostic value of the expression of estrogen receptor beta in patients with non-small cell lung cancer: a meta-analysis. Transl Lung Cancer Res. 2016;5(2):202–7.CrossRef Ma L, Zhan P, Liu Y, Zhou Z, Zhu Q, Miu Y, Wang X, Jin J, Li Q, Lv T, et al. Prognostic value of the expression of estrogen receptor beta in patients with non-small cell lung cancer: a meta-analysis. Transl Lung Cancer Res. 2016;5(2):202–7.CrossRef
23.
Zurück zum Zitat Song RX, Zhang Z, Chen Y, Bao Y, Santen RJ. Estrogen signaling via a linear pathway involving insulin-like growth factor I receptor, matrix metalloproteinases, and epidermal growth factor receptor to activate mitogen-activated protein kinase in MCF-7 breast cancer cells. Endocrinology. 2007;148(8):4091–101.CrossRef Song RX, Zhang Z, Chen Y, Bao Y, Santen RJ. Estrogen signaling via a linear pathway involving insulin-like growth factor I receptor, matrix metalloproteinases, and epidermal growth factor receptor to activate mitogen-activated protein kinase in MCF-7 breast cancer cells. Endocrinology. 2007;148(8):4091–101.CrossRef
24.
Zurück zum Zitat Konduri S, Schwarz RE. Estrogen receptor beta/alpha ratio predicts response of pancreatic cancer cells to estrogens and phytoestrogens. J Surg Res. 2007;140(1):55–66.CrossRef Konduri S, Schwarz RE. Estrogen receptor beta/alpha ratio predicts response of pancreatic cancer cells to estrogens and phytoestrogens. J Surg Res. 2007;140(1):55–66.CrossRef
25.
Zurück zum Zitat Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998;4(7):844–7.CrossRef Kononen J, Bubendorf L, Kallioniemi A, Barlund M, Schraml P, Leighton S, Torhorst J, Mihatsch MJ, Sauter G, Kallioniemi OP. Tissue microarrays for high-throughput molecular profiling of tumor specimens. Nat Med. 1998;4(7):844–7.CrossRef
26.
Zurück zum Zitat Remmele W, Stegner HE. Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe. 1987;8(3):138–40.PubMed Remmele W, Stegner HE. Recommendation for uniform definition of an immunoreactive score (IRS) for immunohistochemical estrogen receptor detection (ER-ICA) in breast cancer tissue. Pathologe. 1987;8(3):138–40.PubMed
27.
Zurück zum Zitat Ollayos CW, Riordan GP, Rushin JM. Estrogen receptor detection in paraffin sections of adenocarcinoma of the colon, pancreas, and lung. Arch Pathol Lab Med. 1994;118(6):630–2.PubMed Ollayos CW, Riordan GP, Rushin JM. Estrogen receptor detection in paraffin sections of adenocarcinoma of the colon, pancreas, and lung. Arch Pathol Lab Med. 1994;118(6):630–2.PubMed
28.
Zurück zum Zitat Yeh TS, Jan YY, Chiu CT, Ho YB, Chen TC, Lee KF, Chan KM, Hsu JC, Hwang TL, Chen MF. Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut. 2002;51(5):712–6.CrossRef Yeh TS, Jan YY, Chiu CT, Ho YB, Chen TC, Lee KF, Chan KM, Hsu JC, Hwang TL, Chen MF. Characterisation of oestrogen receptor, progesterone receptor, trefoil factor 1, and epidermal growth factor and its receptor in pancreatic cystic neoplasms and pancreatic ductal adenocarcinoma. Gut. 2002;51(5):712–6.CrossRef
29.
Zurück zum Zitat Iwao K, Miyoshi Y, Ooka M, Ishikawa O, Ohigashi H, Kasugai T, Egawa C, Noguchi S. Quantitative analysis of estrogen receptor-alpha and -beta messenger RNA expression in human pancreatic cancers by real-time polymerase chain reaction. Cancer Lett. 2001;170(1):91–7.CrossRef Iwao K, Miyoshi Y, Ooka M, Ishikawa O, Ohigashi H, Kasugai T, Egawa C, Noguchi S. Quantitative analysis of estrogen receptor-alpha and -beta messenger RNA expression in human pancreatic cancers by real-time polymerase chain reaction. Cancer Lett. 2001;170(1):91–7.CrossRef
30.
Zurück zum Zitat Singh S, Baker PR, Poulsom R, Wright NA, Sheppard MC, Langman MJ, Neoptolemos JP. Expression of oestrogen receptor and oestrogen-inducible genes in pancreatic cancer. Br J Surg. 1997;84(8):1085–9.CrossRef Singh S, Baker PR, Poulsom R, Wright NA, Sheppard MC, Langman MJ, Neoptolemos JP. Expression of oestrogen receptor and oestrogen-inducible genes in pancreatic cancer. Br J Surg. 1997;84(8):1085–9.CrossRef
31.
Zurück zum Zitat Izumo A, Yamaguchi K, Eguchi T, Nishiyama K, Yamamoto H, Yonemasu H, Yao T, Tanaka M, Tsuneyoshi M. Mucinous cystic tumor of the pancreas: immunohistochemical assessment of “ovarian-type stroma”. Oncol Rep. 2003;10(3):515–25.PubMed Izumo A, Yamaguchi K, Eguchi T, Nishiyama K, Yamamoto H, Yonemasu H, Yao T, Tanaka M, Tsuneyoshi M. Mucinous cystic tumor of the pancreas: immunohistochemical assessment of “ovarian-type stroma”. Oncol Rep. 2003;10(3):515–25.PubMed
32.
Zurück zum Zitat Thompson LD, Becker RC, Przygodzki RM, Adair CF, Heffess CS. Mucinous cystic neoplasm (mucinous cystadenocarcinoma of low-grade malignant potential) of the pancreas: a clinicopathologic study of 130 cases. Am J Surg Pathol. 1999;23(1):1–16.CrossRef Thompson LD, Becker RC, Przygodzki RM, Adair CF, Heffess CS. Mucinous cystic neoplasm (mucinous cystadenocarcinoma of low-grade malignant potential) of the pancreas: a clinicopathologic study of 130 cases. Am J Surg Pathol. 1999;23(1):1–16.CrossRef
33.
Zurück zum Zitat Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29(2):217–33.CrossRef Arpino G, Wiechmann L, Osborne CK, Schiff R. Crosstalk between the estrogen receptor and the HER tyrosine kinase receptor family: molecular mechanism and clinical implications for endocrine therapy resistance. Endocr Rev. 2008;29(2):217–33.CrossRef
34.
Zurück zum Zitat Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001;276(13):9817–24.CrossRef Campbell RA, Bhat-Nakshatri P, Patel NM, Constantinidou D, Ali S, Nakshatri H. Phosphatidylinositol 3-kinase/AKT-mediated activation of estrogen receptor alpha: a new model for anti-estrogen resistance. J Biol Chem. 2001;276(13):9817–24.CrossRef
35.
Zurück zum Zitat Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Strom A, Treuter E, Warner M, et al. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 2007;87(3):905–31.CrossRef Heldring N, Pike A, Andersson S, Matthews J, Cheng G, Hartman J, Tujague M, Strom A, Treuter E, Warner M, et al. Estrogen receptors: how do they signal and what are their targets. Physiol Rev. 2007;87(3):905–31.CrossRef
36.
Zurück zum Zitat Shupnik MA. Crosstalk between steroid receptors and the c-Src-receptor tyrosine kinase pathways: implications for cell proliferation. Oncogene. 2004;23(48):7979–89.CrossRef Shupnik MA. Crosstalk between steroid receptors and the c-Src-receptor tyrosine kinase pathways: implications for cell proliferation. Oncogene. 2004;23(48):7979–89.CrossRef
37.
Zurück zum Zitat Konstantinopoulos PA, Kominea A, Vandoros G, Sykiotis GP, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G, Papavassiliou AG. Oestrogen receptor beta (ERbeta) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer. 2003;39(9):1251–8.CrossRef Konstantinopoulos PA, Kominea A, Vandoros G, Sykiotis GP, Andricopoulos P, Varakis I, Sotiropoulou-Bonikou G, Papavassiliou AG. Oestrogen receptor beta (ERbeta) is abundantly expressed in normal colonic mucosa, but declines in colon adenocarcinoma paralleling the tumour’s dedifferentiation. Eur J Cancer. 2003;39(9):1251–8.CrossRef
38.
Zurück zum Zitat Nozoe T, Oyama T, Takenoyama M, Hanagiri T, Sugio K, Yasumoto K. Significance of immunohistochemical expression of estrogen receptors alpha and beta in squamous cell carcinoma of the esophagus. Clin Cancer Res. 2007;13(14):4046–50.CrossRef Nozoe T, Oyama T, Takenoyama M, Hanagiri T, Sugio K, Yasumoto K. Significance of immunohistochemical expression of estrogen receptors alpha and beta in squamous cell carcinoma of the esophagus. Clin Cancer Res. 2007;13(14):4046–50.CrossRef
39.
Zurück zum Zitat Slattery ML, Sweeney C, Murtaugh M, Ma KN, Wolff RK, Potter JD, Caan BJ, Samowitz W. Associations between ERalpha, ERbeta, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomark Prev. 2005;14(12):2936–42.CrossRef Slattery ML, Sweeney C, Murtaugh M, Ma KN, Wolff RK, Potter JD, Caan BJ, Samowitz W. Associations between ERalpha, ERbeta, and AR genotypes and colon and rectal cancer. Cancer Epidemiol Biomark Prev. 2005;14(12):2936–42.CrossRef
40.
Zurück zum Zitat Caiazza F, Galluzzo P, Lorenzetti S, Marino M. 17Beta-estradiol induces ERbeta up-regulation via p38/MAPK activation in colon cancer cells. Biochem Biophys Res Commun. 2007;359(1):102–7.CrossRef Caiazza F, Galluzzo P, Lorenzetti S, Marino M. 17Beta-estradiol induces ERbeta up-regulation via p38/MAPK activation in colon cancer cells. Biochem Biophys Res Commun. 2007;359(1):102–7.CrossRef
41.
Zurück zum Zitat Kalayarasan R, Ananthakrishnan N, Kate V, Basu D. Estrogen and progesterone receptors in esophageal carcinoma. Dis Esophagus. 2008;21(4):298–303.CrossRef Kalayarasan R, Ananthakrishnan N, Kate V, Basu D. Estrogen and progesterone receptors in esophageal carcinoma. Dis Esophagus. 2008;21(4):298–303.CrossRef
42.
Zurück zum Zitat Verbeke CS, Menon KV. Variability in reporting resection margin status in pancreatic cancer. Ann Surg. 2008;247(4):716–7.CrossRef Verbeke CS, Menon KV. Variability in reporting resection margin status in pancreatic cancer. Ann Surg. 2008;247(4):716–7.CrossRef
43.
Zurück zum Zitat Xu CY, Guo JL, Jiang ZN, Xie SD, Shen JG, Shen JY, Wang LB. Prognostic role of estrogen receptor alpha and estrogen receptor beta in gastric cancer. Ann Surg Oncol. 2010;17(9):2503–9.CrossRef Xu CY, Guo JL, Jiang ZN, Xie SD, Shen JG, Shen JY, Wang LB. Prognostic role of estrogen receptor alpha and estrogen receptor beta in gastric cancer. Ann Surg Oncol. 2010;17(9):2503–9.CrossRef
44.
Zurück zum Zitat Rudolph A, Toth C, Hoffmeister M, Roth W, Herpel E, Jansen L, Marx A, Brenner H, Chang-Claude J. Expression of oestrogen receptor beta and prognosis of colorectal cancer. Br J Cancer. 2012;107(5):831–9.CrossRef Rudolph A, Toth C, Hoffmeister M, Roth W, Herpel E, Jansen L, Marx A, Brenner H, Chang-Claude J. Expression of oestrogen receptor beta and prognosis of colorectal cancer. Br J Cancer. 2012;107(5):831–9.CrossRef
45.
Zurück zum Zitat Iavarone M, Lampertico P, Seletti C, Francesca Donato M, Ronchi G, del Ninno E, Colombo M. The clinical and pathogenetic significance of estrogen receptor-beta expression in chronic liver diseases and liver carcinoma. Cancer. 2003;98(3):529–34.CrossRef Iavarone M, Lampertico P, Seletti C, Francesca Donato M, Ronchi G, del Ninno E, Colombo M. The clinical and pathogenetic significance of estrogen receptor-beta expression in chronic liver diseases and liver carcinoma. Cancer. 2003;98(3):529–34.CrossRef
46.
Zurück zum Zitat Villa E, Colantoni A, Camma C, Grottola A, Buttafoco P, Gelmini R, Ferretti I, Manenti F. Estrogen receptor classification for hepatocellular carcinoma: comparison with clinical staging systems. J Clin Oncol. 2003;21(3):441–6.CrossRef Villa E, Colantoni A, Camma C, Grottola A, Buttafoco P, Gelmini R, Ferretti I, Manenti F. Estrogen receptor classification for hepatocellular carcinoma: comparison with clinical staging systems. J Clin Oncol. 2003;21(3):441–6.CrossRef
47.
Zurück zum Zitat Giroux V, Lemay F, Bernatchez G, Robitaille Y, Carrier JC. Estrogen receptor beta deficiency enhances small intestinal tumorigenesis in ApcMin/+ mice. Int J Cancer. 2008;123(2):303–11.CrossRef Giroux V, Lemay F, Bernatchez G, Robitaille Y, Carrier JC. Estrogen receptor beta deficiency enhances small intestinal tumorigenesis in ApcMin/+ mice. Int J Cancer. 2008;123(2):303–11.CrossRef
48.
Zurück zum Zitat Fan S, Liao Y, Liu C, Huang Q, Liang H, Ai B, Fu S, Zhou S. Estrogen promotes tumor metastasis via estrogen receptor beta-mediated regulation of matrix-metalloproteinase-2 in non-small cell lung cancer. Oncotarget. 2017;8(34):56443–59.CrossRef Fan S, Liao Y, Liu C, Huang Q, Liang H, Ai B, Fu S, Zhou S. Estrogen promotes tumor metastasis via estrogen receptor beta-mediated regulation of matrix-metalloproteinase-2 in non-small cell lung cancer. Oncotarget. 2017;8(34):56443–59.CrossRef
49.
Zurück zum Zitat Niv Y. Estrogen receptor beta expression and colorectal cancer: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2015;27(12):1438–42.CrossRef Niv Y. Estrogen receptor beta expression and colorectal cancer: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2015;27(12):1438–42.CrossRef
50.
Zurück zum Zitat Stevanato Filho PR, Aguiar Junior S, Begnami MD, Ferreira FO, Nakagawa WT, Spencer R, Bezerra TS, Boggiss PE, Lopes A. Estrogen receptor beta as a prognostic marker of tumor progression in colorectal Cancer with familial adenomatous polyposis and sporadic polyps. Pathol Oncol Res. 2017;24(3):533–40.CrossRef Stevanato Filho PR, Aguiar Junior S, Begnami MD, Ferreira FO, Nakagawa WT, Spencer R, Bezerra TS, Boggiss PE, Lopes A. Estrogen receptor beta as a prognostic marker of tumor progression in colorectal Cancer with familial adenomatous polyposis and sporadic polyps. Pathol Oncol Res. 2017;24(3):533–40.CrossRef
51.
Zurück zum Zitat Topi G, Ehrnstrom R, Jirstrom K, Palmquist I, Lydrup ML, Sjolander A. Association of the oestrogen receptor beta with hormone status and prognosis in a cohort of female patients with colorectal cancer. Eur J Cancer. 2017;83:279–89.CrossRef Topi G, Ehrnstrom R, Jirstrom K, Palmquist I, Lydrup ML, Sjolander A. Association of the oestrogen receptor beta with hormone status and prognosis in a cohort of female patients with colorectal cancer. Eur J Cancer. 2017;83:279–89.CrossRef
52.
Zurück zum Zitat Miller MM, McMullen PD, Andersen ME, Clewell RA. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol. 2017;47(7):564–80.CrossRef Miller MM, McMullen PD, Andersen ME, Clewell RA. Multiple receptors shape the estrogen response pathway and are critical considerations for the future of in vitro-based risk assessment efforts. Crit Rev Toxicol. 2017;47(7):564–80.CrossRef
53.
Zurück zum Zitat Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011;150(3):476–3.CrossRef Assifi MM, Lu X, Eibl G, Reber HA, Li G, Hines OJ. Neoadjuvant therapy in pancreatic adenocarcinoma: a meta-analysis of phase II trials. Surgery. 2011;150(3):476–3.CrossRef
54.
Zurück zum Zitat Torlakovic E, Lilleby W, Berner A, Torlakovic G, Chibbar R, Furre T, Fossa SD. Differential expression of steroid receptors in prostate tissues before and after radiation therapy for prostatic adenocarcinoma. Int J Cancer. 2005;117(3):381–6.CrossRef Torlakovic E, Lilleby W, Berner A, Torlakovic G, Chibbar R, Furre T, Fossa SD. Differential expression of steroid receptors in prostate tissues before and after radiation therapy for prostatic adenocarcinoma. Int J Cancer. 2005;117(3):381–6.CrossRef
Metadaten
Titel
Expression of estrogen receptor beta correlates with adverse prognosis in resected pancreatic adenocarcinoma
verfasst von
Hendrik Seeliger
Ioannis Pozios
Gerald Assmann
Yue Zhao
Mario H. Müller
Thomas Knösel
Martin E. Kreis
Christiane J. Bruns
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4973-6

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.