Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Endometrial Endometrioid Carcinoma Metastases Show Decreased ER-Alpha and PR-A Expression Compared to Matched Primary Tumors

  • Carla Bartosch ,

    carlabartosch@med.up.pt

    Affiliations Department of Pathology, Portuguese Oncology Institute-Porto, Porto, Portugal, Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute- Porto, Porto, Portugal, Medical Faculty, University of Porto, Porto, Portugal

  • Sara Monteiro-Reis,

    Affiliations Department of Pathology, Portuguese Oncology Institute-Porto, Porto, Portugal, Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute- Porto, Porto, Portugal

  • Renata Vieira,

    Affiliation Department of Pathology, Portuguese Oncology Institute-Porto, Porto, Portugal

  • Armindo Pereira,

    Affiliation Department of Pathology, Centro Hospitalar São Joao, Porto, Portugal

  • Marta Rodrigues,

    Affiliation Department of Pathology, Centro Hospitalar São Joao, Porto, Portugal

  • Carmen Jerónimo ,

    ‡ These authors are joint senior authors on this work.

    Affiliations Cancer Biology and Epigenetics Group, Research Center of Portuguese Oncology Institute- Porto, Porto, Portugal, Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar-ICBAS, University of Porto, Porto, Portugal

  • José M. Lopes

    ‡ These authors are joint senior authors on this work.

    Affiliations Medical Faculty, University of Porto, Porto, Portugal, Department of Pathology, Centro Hospitalar São Joao, Porto, Portugal, IPATIMUP, Institute of Molecular Pathology and Immunology, Porto, Portugal

Abstract

Patients with endometrial endometrioid carcinoma (EEC) that present with advanced primary disease and develop recurrences have a poor outcome. The phenotype of EEC metastases and recurrences is poorly studied. We evaluated the morphological features and ER-alpha/PRA/p53 immunohistochemical expression of a sample of 45 EEC metastases compared to matched primary tumors. Additionally, we studied methylation levels of ER-alpha/PRA gene promoters. The distribution of histological FIGO grade was significantly different in metastases, which disclosed higher grade than primary tumors (p = 0.005). Mitotic index was significantly lower in metastases compared to matched primary tumors (p<0.001). ER-alpha (p = 0.002) and PRA (p<0.001) median H-scores were significantly lower in metastases than in matched primary EECs, but there was no significant difference concerning p53 expression (p = 0.056). ER-alpha/PRA expression differences did not correlate with differences in metastases morphology. ER-alpha/PRA gene promoter levels were globally low (range: 0% to 11.9%). One case showed higher ER-alpha gene promoter methylation in metastasis compared to matched EEC primary tumor. Regarding PRA, there was a significant higher frequency of its promotor methylation in metastases compared to primary tumors (51.6% vs. 22.7%, p = 0.022). In conclusion, EEC metastatic disease displays phenotypic changes along with ER-alpha and PRA decreased expression compared to primary tumors. ER-alpha and PRA gene promoter methylation seems to play a limited role in the etiology of these alterations. PR expression assessment for hormonal treatment decision of patients with advanced tumors, may be more adequate in metastases than in EEC primary tumors.

Introduction

In general, patients with endometrial endometrioid carcinoma (EEC) have a good prognosis. However, approximately 25% of patients present with advanced primary disease outside the uterus at the time of diagnosis, and 15–20% of patients have recurrences later in the course of disease [1, 2]. Most patients with advanced and recurrent EEC will die of disease, and death rates are reported to be increasing [3, 4]. Characterization of EEC metastases and recurrences is poorly studied.

Steroid hormones, namely estrogens and progestins, play a major role in EEC. Estrogens have been associated with endometrial cancer development and growth, while progestins have an opposite effect, promoting differentiation and growth inhibition [5]. The effects of both hormones in EEC are mediated by their nuclear receptors. Loss of estrogen receptor (ER) and progesterone receptor (PR) expression, as well as p53 expression, have been associated with aggressive phenotypes and poor survival in EEC primary tumors [2, 6, 7]. Gene promotor hypermethylation has been suggested as one of the mechanisms responsible for ER and PR expression loss [812]. Furthermore, the expression levels of each of the ER subtypes (alpha and beta) and the different PR isoforms (A and B), seem to regulate proliferation and differentiation of endometrial cells, and were postulated to be altered in endometrial cancer [1316].

Patients with advanced or recurrent EEC might be candidates for hormone therapy, being the response to hormone treatment dependent on PR expression [5]. However, assessment of hormone receptor expression is usually not routinely performed before treatment initiation, and when requested is usually performed in the primary tumor. Although studies on ER and PR expression in EEC metastases or recurrences are limited, it has been suggested that hormone receptor expression diminishes in metastatic disease [1719].

In this study we aimed to evaluate the morphological phenotype and ER/PR/p53 immunohistochemical expression of a sample of EEC metastases compared to matched primary tumors. Additionally, we studied the methylation levels of ER and PR gene promoters both in EEC primary tumors and corresponding metastases.

Methods

Patients and histological evaluation

We identified EEC patients diagnosed with metastasis at two Portuguese tertiary centers [Centro Hospitalar S. João (CHSJ) and Portuguese Institute of Oncology—Porto (IPOP)], between 2000 and 2013, by searching the respective Pathology Department databases.

Cases with histological confirmation of metastasis, either at diagnosis or during follow-up, were selected. Cases with ambiguous histology and synchronous ovarian endometrioid carcinoma, only local vaginal recurrence, cytological confirmation of metastasis or unavailable formalin fixed paraffin embedded (FFPE) tissue, were excluded. Altogether, 45 pairs of EEC primary tumors and metastases were included in the study, after approval by the Ethics Committee of both participating institutions [CHSJ (CES44/2010)/IPOP (CES494-010)]. Because this was a retrospective study, based on archive paraffin-embedded tissue and many patients were deceased when the study was started, no informed consent was procured and data was anonymized and de-identified prior to analysis.

The patients’ clinical files and pathological material were retrospectively reviewed. Data collected included patients’ age at diagnosis, tumor size, histological subtype, lymphovascular invasion, International Federation of Gynecology and Obstetrics (FIGO) stage, treatment (radiotherapy, hormone therapy and chemotherapy), site of metastases, progression free and overall survival.

All histological slides were reviewed by a gynecopathologist (CB). Histological subtype, histological FIGO grade (according to the WHO criteria) and mitotic index [number of mitosis per 10 high power fields (HPF)] were evaluated both in primary tumors and metastases.

Immunohistochemistry

ER, PR and p53 protein expression was assessed by immunohistochemistry. Selected FFPE blocks were sectioned at 3 μm thickness. Sections were deparaffinized in xylene and rehydrated through a graded ethanol series. Antigen retrieval was achieved by incubating the sections in preheated EDTA buffer (pH 8) for ER-alpha and in citrate buffer (pH6) for PRA and p53 in a water bath (98°C) for 20 minutes. Endogenous peroxidase was blocked and the slides were incubated for 1h using the following antibodies: ER alpha (clone 6F11, mouse monoclonal antibody, 1:100, Novocastra Leica Biosystems), PRA (clone 16, mouse monoclonal antibody, 1:150, Novocastra Leica Biosystems) and p53 (clone DO7, mouse monoclonal antibody, 1:200, Dako). Finally, sections were incubated with Poly-HRP-GAM/R/R IgG (ImmunoLogic) and counterstained with hematoxylin. Appropriate positive and negative controls were also included in each assay.

ER-alpha, PRA and p53 expression was evaluated by optical microscopy using an image analysis program (GenASIs Go-Path). Fifteen random fields of the tumor in each slide (corresponding to a mean of 7138 tumors cells per case) were photographed. In each photo tumor cells were selected and evaluated using H-score. H-score was defined by the intensity grade of staining (0–3) multiplied by the percentage of positive cells, resulting in a range of possible scores of 0–300.

DNA extraction

Representative sections of EEC primary tumors and matched metastases larger than 5mm (n = 31), were selected for DNA extraction. Enriched tumor cell areas were marked on a HE-stained slide and used as a template for macroscopic dissection. The corresponding areas of ten consecutive 7μm thick sections placed on glass slides were scratched into 1.5mL tubes using a sterile, single-use scalpel. Tissue samples were deparaffinized and then submitted to overnight digestion, in a dried bath incubator at 55°C, using 1mL of digestion buffer (composed by Tris-HCl 1M, EDTA 0,1M, Tween 20 and sterile bidistilled water), and proteinase K (20mg/ml, 50μL) (Sigma-Aldrich, Germany). Genomic DNA was extracted by standard phenol–chlorophorm method, precipitated with ethanol and eluted in 20 μl of water [20].

Bisulfite modification and quantitative methylation-specific PCR

DNA samples from were subjected to sodium bisulfite modification, using the EZ DNA Methylation-Gold Kit (Zymo Research, Orange, CA, USA) according to manufacturer’s instructions.

Methylation analysis of estrogen (ESR1) and progesterone (PRA) receptor gene promoters was performed by quantitative methylation-specific PCR (qMSP). Bisulfite-modified genomic DNA was tested using four sets of primers that recognize the methylated sequence located at the promoter regions of ERS1, and PRA genes. Primers used were those previously published [10, 21]: PRA forward: 5’-ACGGGTTATTTTTTTTTCG-3’, reverse: 5’-TAAAATATACGCCCTCCACG-3’; ESR1 forward 5’-GGCGTTCGTTTTGGGATTG-3’, reverse 5’-GCCGACACGCGAACTCTAA-3’.

The qMSP was performed in a 10μl reaction volume including 10 μM forward and reverse primers, 50 ng bisulfite treated DNA from tissue samples, and 5μL Kapa Sybr Fast qPCR master mix (Kapa Byosystems). Each sample was analyzed in triplicate in 384-well plates using the LightCycler 480 System (Roche). The geometric mean of the two closest values for each sample was used for data analysis. A standard curve was generated from 1:5 serial dilutions of bisulfite-converted commercially available methylated DNA (CpGenome Universal Methylated DNA; Millipore Billerica). The universal methylated DNA sample was also used as a positive control for the qMSP reaction. Additionally, all plates contained two water blanks as negative controls. All samples were also tested using primers for non–CpG-containing regions of an internal reference gene, β-actin (ACTB) to normalize for input DNA (primers: forward 5’- TGGTGATGGAGGAGGTTTAGTAAGT- 3’, reverse 5’- AACCAATAAAACCTACTCCTCCCTTAA- 3’). The level of methylated DNA (percent of methylated reference, PMR) was calculated using the following formula: [(target gene/ACTB)sample / (target gene/ACTB)positive control] x 100. Additionally PMR values were dichotomized to methylated or unmethylated using a cut-off of 4%, as used in a number of prior publications [22, 23].

Statistical analysis

Data was tabulated and analyzed using STATA (STATACorp, Texas, USA). We performed a pairwise analysis to compare differences between primary tumors and metastases. Marginal homogeneity test was used to compare the distribution of histological FIGO grade across primary tumors and matched metastases. Wilcoxon sign rank test was used to compare median differences in mitotic index, ER-alpha, PRA and p53 expression, and continuous methylation levels between matched primary tumors and metastases. McNemar test was used to compare dichotomized methylation levels between primary tumors and metastases. We used Mann-Whitney and Kruskal-Wallis tests to evaluate the associations of immunohistochemical results with clinical-pathological parameters. Spearman’s rank correlation coefficient was used to evaluate the correlation between ER, PR and mitotic index changes. A p value equal or inferior to 0.05 was considered significant.

Results

Clinical and pathological features

The clinicopathological features of our cohort of patients are depicted in Table 1. The mean age of patients was 63.8 years (range: 39–87). All 45 patients included in the study underwent total hysterectomy and the majority (84.5%) was staged as FIGO III/IV. Most (66.7%) underwent adjuvant radiotherapy and 31.1% had adjuvant chemotherapy.

thumbnail
Table 1. Clinicopathological features of EEC patients diagnosed with metastases.

https://doi.org/10.1371/journal.pone.0134969.t001

Cases were classified as endometrioid carcinomas with no specification (n = 31, 68.9%) and as endometrioid carcinomas with squamous differentiation (n = 14, 31.1%). Focal mucinous differentiation was identified in 5 (11.1%), and 4 (8.9%) had areas of microcystic elongated and fragmented (MELF) pattern. Median tumor size was 6.5 cm (range: 1.4, 11). Thirty (66.7%) invaded more than half of the myometrium, 12 (26.7%) had endocervical stromal invasion and 37 (82.2%) had lymphovascular invasion.

Metastases were diagnosed synchronous to the endometrial carcinoma (n = 28, 62.2%) or later in the course of disease (n = 17, 37.8%). They were located in pelvic/para-aortic lymph (60%) nodes, peritoneum (26.7%), and other extra-pelvic distant sites (13.3%). Nine (20.0%) patients also had local vaginal recurrence. Treatment of metastatic and local recurrent disease included radiotherapy (n = 7, 15.6%), chemotherapy (n = 10, 22.2%) and hormone therapy (n = 12, 26.7%).

At the end of the study 20 (44.4%) patients had died of disease, 1 (2.22%) died of other causes, 23 (51.1%) were alive, and 1 (2.22%) was lost to follow-up. The median follow-up time for survivors was 33.9 months, ranging from 7 months to 11 years. The median disease free survival was 50.9 months (95% CI 29.0–69.2). The overall median survival was 56.7 months (95% CI 40.1–133.5) with a 5-year survival rate of 49.3%.

Histological comparison of primary tumors and metastases

The morphological features of ECC primary tumors and metastases were similar in most of the cases (n = 31, 68.9%), but there were cases with discordant FIGO grade (n = 14, 31.1%) (Figs 1 and 2). The distribution of histological FIGO grade was significantly different when comparing metastases to matched primary tumors (p = 0.005). As described in Table 2 and Fig 3, metastases disclosed more frequently higher histological FIGO grade than primary tumors. On the other hand, mitotic rate was significantly lower in metastases when compared to matched primary tumors, with a median difference of -8 mitoses/10 HPF (p<0.001). This difference remains significant even when limiting the analysis to metastases at diagnosis (-8 mitoses/10 HPF, p<0.001).

thumbnail
Fig 1. Histological features.

Morphological similar EEC primary tumor (A) and matched lymph node metastasis (B). Scale bar = 200μm.

https://doi.org/10.1371/journal.pone.0134969.g001

thumbnail
Fig 2. Histological features.

Morphological discordant EEC primary tumor (A) and matched peritoneal recurrence (B), showing higher histological FIGO grade in the recurrence. Scale bar = 100μm.

https://doi.org/10.1371/journal.pone.0134969.g002

thumbnail
Fig 3. Immunohistochemical expression.

Box-plots showing ER-alpha, PRA and p53 immunohistochemical expression levels in EEC primary tumors compared to metastases.

https://doi.org/10.1371/journal.pone.0134969.g003

thumbnail
Table 2. Comparison of morphological features and ER-alpha, PRA and p53 immunohistochemical expression between EEC primary tumors and metastases.

https://doi.org/10.1371/journal.pone.0134969.t002

Immunohistochemical ER-alpha, PRA and p53 expression

ER expression was intermediate to strong and diffuse in most EEC primary tumors, with a median staining extent of 77.1% positive cells and a median H-score of 175.1 (range: 0.3, 259.2). PR expression was also strong and diffuse in most EEC primary tumors, with a median staining extent of 71.5% positive cells, and a median H-score of 191.2 (range: 0, 273.8). There was a tendency for lower ER-alpha/PRA H-scores in EECs with higher histological FIGO grade. This was statistically significant for PRA (median PRA H-score by grade: I- 207.4, II-199.9, III-109.6; p = 0.022), but not significant for ER-alpha expression (median ER-alpha H-score by grade: I- 186.6, II-175.9, III-1355; p = 0.190).

The expression of p53 was within the levels considered as physiologic in the majority of EEC primary tumors, with scattered nuclei staining (median 19.9% positive cells, range: 0, 88.4%). The median p53 H-score was 38.4, ranging from 0 to 244.8. There was a tendency for higher p53 H-scores in EEC with higher histological FIGO grade, but not statistically significant (median p53 H-score by grade: I- 35.4, II-40.4, III-43.5; p = 0.343).

The differences of expression between metastasis and primary tumor of each case are plotted in Fig 4. ER-alpha expression was significantly lower in metastases than in matched EEC primary tumors, with a median difference of -21.9 (p = 0.002). The same was observed when comparing PRA expression with a median difference of -32.6 (p<0.001). In contrast, for p53 there was no significant difference in expression between metastases and matched primary tumors (median difference = 2.5, p = 0.056). Additionally, the decreased expression in ER-alpha and PRA were correlated with each other (rs = 0.58, p<0.001).

thumbnail
Fig 4. Immunohistochemical features.

Scatter plots showing H-score differences in ER-alpha/PRA/p53 expression in EEC metastases matched to primary tumors and corresponding immunohistochemical representative examples. Plot horizontal bars corresponds to zero differences between primary tumors and metastases. It is evident that most cases showed a negative change of imunoexpression H-score, i.e. a decrease, of ER-alpha and PRA in metastases. Scale bar = 100μm.

https://doi.org/10.1371/journal.pone.0134969.g004

There was no relationship between the differences in ER-alpha and PRA expression and the differences found in tumor grade (ER-alpha: p = 0.783, PRA: 0.564) nor with differences found in mitotic index (ER-alpha: p = 0.906, PRA: p = 0.776). The PRA expression difference between metastases and primary tumors is significantly higher for FIGO stage I/II tumors compared to FIGO stage III/IV, but there was no association of ER-alpha and p53 expression with FIGO stage (ER-alpha: 0.079, PRA: 0.031, p53: 0.824). The decrease in ER-alpha and PRA expression also did not associate with the remaining clinicopathological parameters, namely metastasis site (ER-alpha: p = 0.397, PRA: p = 0.800), lymphovascular invasion (ER-alpha: p = 0.190, PRA: p = 0.764), radiotherapy (ER-alpha: p = 0.174, PRA: p = 0.211), chemotherapy (ER-alpha: p = 0.485, PRA: p = 0.883).

ER-alpha and PRA gene promoter methylation levels

Promotor methylation levels, expressed as PMR, of the genes studied (ESR1, PRA) were in general mild, varying from 0% to 11.9%. Using PMR as a continuous variable, there was a tendency for higher promoter methylation levels in metastases compared to primary tumors for ESR1 (median PMR: primary tumors = 0.83% vs. metastases = 1.03%, p = 0.349), PRA (median PMR: primary tumors = 3.27% vs. metastases = 4.01%, p = 0.125), but without any statistical significance (Fig 5).

thumbnail
Fig 5. Gene promoter methylation levels.

ER-alpha and PRA gene promoters methylation levels in EEC primary tumors compared to metastases.

https://doi.org/10.1371/journal.pone.0134969.g005

Analyzing dichotomized PMR, one (3%) of the metastases displayed ESR1 gene promotor methylation, whereas no methylation of ESR1 gene promotor was found in EEC primary tumors. Regarding PRA, there was a significant higher frequency of its promotor methylation in metastases (n = 16, 51.6%) compared to EEC primary tumors (n = 7, 22.7%) (p = 0.022).

There was no significant correlation between differences in immunohistochemistry expression and differences in methylation levels neither for estrogen nor progesterone receptors. However, the group of cases that showed a change in PRA promotor methylation status in metastases compared to the matched EEC primary tumors, displayed a larger difference in immunohistochemical PRA expression (median H-score difference = -41, range: -201, -4.6) compared to cases with no change in methylation status (median H-score difference = -13.8, range: -132.7, -75.1), but without statistical significance (p = 0.095). The metastasis with ESR1 promoter hypermethylation did not express ER-alpha.

Discussion

In our study we compared the morphology, ER-alpha, PRA and p53 immunoexpression of metastases and matched primary tumors in a series of 45 EECs. We showed that metastases had significantly higher FIGO grade, lower mitotic index and decreased ER-alpha and PRA expression compared to matched EEC primary tumors. No significant difference was found regarding p53 expression. Additionally, we found a higher frequency of hormone receptors gene promotor hypermethylation in metastases compared to matched primary tumors, particularly in PRA gene.

Very few studies have addressed the histological characterization of endometrial metastases/recurrences [1719]. Soslow et al described a series of endometrial carcinomas in which discrepancies between recurrences and primary were found [18]. Particularly, they found a tendency for endometrial carcinomas to become morphological ambiguous or with higher grade in recurrences. Santacana et al. also commented on a higher histological grade in EEC vaginal recurrences of eight cases [17]. Our quantitative evaluation supports these findings showing a significant higher FIGO grade in 45 EEC metastases compared to matched primary tumors. This is in line with the hypothesis that metastases represent selected EEC primary tumor clones that acquire features associated with invasion and metastatic capability and/or tumor progression, as postulated by Darwinian tumor evolution models [24]. Noteworthy, histological FIGO grade may not always adequately indicate the metastatic potential of a heterogeneous tumor. Since FIGO grade relies on the proportional evaluation of aggressiveness-related features, namely solid pattern and cytological atypia, it is possible that low-grade FIGO tumors may harbor focal aggressive tumor clones.

Interestingly, in our study, metastases had a significant lower mitotic index than primary EEC. This finding might be due, in part, to the effect of cytotoxic agents used in EC adjuvant treatment, as reported in other cancer models [25, 26]. However, most (62.2%) of our cases comprised metastases detected at diagnosis, that displayed a significant decrease in mitotic index. In this setting it is possible that EEC metastatic tumor cells may have shifted their cellular mechanisms towards invasion/migration, as opposed to cell division, a concept often referred to as go-or-grow [27]. The proliferative activity of metastases remains controversial in cancer. While studies have reported a decrease in proliferation markers, e.g. breast and esophageal carcinoma lymph node metastases [28, 29], others suggested that metastases are more proliferative than matched primaries, e.g. omental ovarian cancer metastases [30]. In endometrial cancer, to the best of our knowledge, only one study evaluated Ki-67 in local vaginal recurrences and matched primaries, showing a significant higher Ki-67 expression in recurrences [17]. Importantly, a low mitotic index does not always imply a low proliferative activity, and mitotic index does not always correlates with Ki-67 expression [31].

ER and PR expression observed in our EEC primary tumors was similar to what has been reported in the literature, showing lower expression with higher tumor grade [32]. There was a significant decrease both in ER-alpha and PRA expression in metastases compared to primary tumors. Interestingly, there was no significant association between the decrease in hormone receptors and morphological changes. Soslow et al. also reported a frequent decrease of PR expression in matched primary and recurrent endometrial carcinomas; and in addition noted that there was no discordant morphology in the three EEC cases described as having PR expression discordance [18]. Though not statistically significant, Santacana et al. reported decreased expression of PR in recurrences compared to primary tumors [17]. Similarly, using unmatched endometrial carcinoma primary tumors and metastases Tangen et al. also reported a highest proportion of PR loss in the metastases [19]. The antibodies used in these three studies detected both PR isoforms. Fujimoto et al., using southern blot, described 3 cases of metastatic endometrial carcinoma in which PRA mRNA was suppressed compared to the primary tumor [33]. The same authors, in a later study showed that the ratio of ER-beta/ER-alpha immunohistochemical expression was increased in endometrial cancer metastases compared to primary tumors [34]. Interestingly, in breast carcinomas the discordance of hormone receptor status between metastases and primary tumors is fairly well documented [3537]. Hormone receptor conversion by immunohistochemistry in distant breast cancer metastases occurs in a significant proportion patients, and has been shown to be more frequent for PR. Additionally, following the same trend as in breast cancer, hormone receptor expression changes in endometrial carcinoma may also have important implications for therapeutic strategies. EEC patients benefit from hormonal treatment, in particular those for which chemotherapy is not an option due to older age and comorbidities [5]. Nevertheless, the response rate to hormonal treatment reported in the literature is highly variable (11–56%), and usually poor [5]. This variation might be explained by the decrease of hormonal receptors in metastases as observed in the present study. Thus, assessment of hormone receptor status in recurrent/metastatic disease might prove more useful to tailor the treatment of patients with advanced EEC.

Regarding p53 expression we did not observe any significant differences between metastases and matched EEC primary tumors. While Soslow et al. also reported that p53 immunoexpression is usually concordant when evaluating matched pairs, with few exceptions, Santacana et al. reported that p53 expression was higher in patients with post-radiation recurrences [17, 18]. Thus, further studies are needed to clarify the role of p53 in EEC recurrences/metastases.

ER and PR changes during endometrial cancer progression most probably result from genetic, epigenetic or posttranslational alterations. In our study we searched for gene promoter DNA methylation as this is a well-recognized mechanism of gene silencing [38]. Globally, we found low ER and PR methylation levels in EEC primary tumors. However, when comparing primary tumors with metastases, using dichotomized PMR, statistically significant higher methylation levels were found for PRA in matched EEC metastases. This result is in agreement with the PR isoform A decrease protein expression observed in immunohistochemistry. There are only a few studies on ER and PR gene methylation in endometrial carcinomas, mainly using cell lines and rarely primary tumor samples. Reduced ER expression has not been consistently reported to be associated with ER genes promoter methylation [8, 9, 39]. Regarding PR, the functional role of its isoforms in the endometrium is not fully understood; nevertheless, some studies in knockout mice reported that PRA expression is necessary to mediate the anti-proliferative responses to progesterone and to inhibit estrogen-induced proliferation [13]. Additionally, PRA has been also shown to function as a transcriptional inhibitor of other steroid hormone receptors, such as ER and PR-B [40]. Hence, it is possible that a decrease in PRA isoform expression and its gene promoter methylation might play a role in tumor progression. PRA methylation was associated with progestin-resistance in breast carcinoma [41]. In vitro studies have suggested that in endometrial carcinoma it is the PRB isoform that plays a major role in carcinogenesis, by regulating anti-proliferative activity and invasiveness [42, 43]. PRB isoform, compared to PRA, seems to be underexpressed in high-grade endometrial carcinoma, but loss of both PR isoforms has been associated with poor prognosis [14, 15, 44, 45]. Noteworthy, other mechanisms were also shown to alter PR function in endometrial carcinoma, such as complex of nucleic acid sequence aberrations (e.g. PROGINS) [46], PR promoter polymorphisms [46, 47], and exon region methylation [12, 48]. Moreover, there is recent evidence that PRA also plays a major role in endometrial carcinogenesis, namely by decreasing PR transcription and respective expression in the stroma of progesterone refractory endometrial carcinomas [49].

Some limitations of our study include possible selection bias since we only analyzed EEC metastases that have been biopsied or excised. Furthermore, the evaluation of ER/PR immunohistochemical expression might have been affected by intratumoral heterogeneity; however, most EEC usually showed homogeneous staining. Also, we should mention that the antibodies used were specific only for ER alpha and PRA isoform. Even though studying PRB and ER-beta expression would allow a broader knowledge of hormonal status in EEC metastatic progression, our results clearly showed that ER-alpha and PRA are deregulated in that process.

The apparent discrepancies regarding methylation results obtained by us and those reported by others might be due to the different sensitivities of the method/primers used and to the stringent criteria in the analysis.

Conclusion

EEC metastatic progression seems to be related to loss of ER-alpha and PRA expression, but does not appear to depend on the increase of p53 expression. Conventional morphological markers of EEC aggressiveness such as histological FIGO grade are relevant in EEC metastases, but do not associate with ER-alpha and PRA loss in metastases. The decrease in PRA expression highlights that, for patients candidates to hormone therapy, receptor status assessment might be more informative in metastases than in EEC primary tumors. Gene promotors’ aberrant methylation seems to play a limited role in the decrease of PRA expression in metastatic EEC.

Author Contributions

Conceived and designed the experiments: CB CJ JML. Performed the experiments: CB SMR RV AP. Analyzed the data: CB MR. Wrote the paper: CB CJ JML.

References

  1. 1. Chi DS, Barakat RR, Palayekar MJ, Levine DA, Sonoda Y, Alektiar K, et al. The incidence of pelvic lymph node metastasis by FIGO staging for patients with adequately surgically staged endometrial adenocarcinoma of endometrioid histology. International journal of gynecological cancer: official journal of the International Gynecological Cancer Society. 2008;18(2):269–73. pmid:18334008.
  2. 2. Salvesen HB, Haldorsen IS, Trovik J. Markers for individualised therapy in endometrial carcinoma. The Lancet Oncology. 2012;13(8):e353–61. pmid:22846840.
  3. 3. Creasman WT, Odicino F, Maisonneuve P, Quinn MA, Beller U, Benedet JL, et al. Carcinoma of the corpus uteri. FIGO 26th Annual Report on the Results of Treatment in Gynecological Cancer. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2006;95 Suppl 1:S105–43. pmid:17161155.
  4. 4. Howlader N, Noone AM, Krapcho M, Neyman N, Aminou R, Waldron W, et al. SEER Cancer Statistics Review, 1975–2008, National Cancer Institute. Bethesda, MD. Available: http://seer.cancer.gov/csr/1975_2008/.
  5. 5. Decruze SB, Green JA. Hormone therapy in advanced and recurrent endometrial cancer: a systematic review. International journal of gynecological cancer: official journal of the International Gynecological Cancer Society. 2007;17(5):964–78. pmid:17442022.
  6. 6. Trovik J, Wik E, Werner HM, Krakstad C, Helland H, Vandenput I, et al. Hormone receptor loss in endometrial carcinoma curettage predicts lymph node metastasis and poor outcome in prospective multicentre trial. Eur J Cancer. 2013;49(16):3431–41. pmid:23932335.
  7. 7. Huvila J, Talve L, Carpen O, Edqvist PH, Ponten F, Grenman S, et al. Progesterone receptor negativity is an independent risk factor for relapse in patients with early stage endometrioid endometrial adenocarcinoma. Gynecol Oncol. 2013;130(3):463–9. pmid:23777659.
  8. 8. Hori M, Iwasaki M, Shimazaki J, Inagawa S, Itabashi M. Assessment of hypermethylated DNA in two promoter regions of the estrogen receptor alpha gene in human endometrial diseases. Gynecol Oncol. 2000;76(1):89–96. pmid:10620447.
  9. 9. Navari JR, Roland PY, Keh P, Salvesen HB, Akslen LA, Iversen OE, et al. Loss of estrogen receptor (ER) expression in endometrial tumors is not associated with de novo methylation of the 5' end of the ER gene. Clin Cancer Res. 2000;6(10):4026–32. pmid:11051252.
  10. 10. Sasaki M, Dharia A, Oh BR, Tanaka Y, Fujimoto S, Dahiya R. Progesterone receptor B gene inactivation and CpG hypermethylation in human uterine endometrial cancer. Cancer Res. 2001;61(1):97–102. pmid:11196205.
  11. 11. Xiong Y, Dowdy SC, Gonzalez Bosquet J, Zhao Y, Eberhardt NL, Podratz KC, et al. Epigenetic-mediated upregulation of progesterone receptor B gene in endometrial cancer cell lines. Gynecol Oncol. 2005;99(1):135–41. pmid:16024066.
  12. 12. Ren Y, Liu X, Ma D, Feng Y, Zhong N. Down-regulation of the progesterone receptor by the methylation of progesterone receptor gene in endometrial cancer cells. Cancer genetics and cytogenetics. 2007;175(2):107–16. pmid:17556066.
  13. 13. Mulac-Jericevic B, Mullinax RA, DeMayo FJ, Lydon JP, Conneely OM. Subgroup of reproductive functions of progesterone mediated by progesterone receptor-B isoform. Science. 2000;289(5485):1751–4. pmid:10976068.
  14. 14. Miyamoto T, Watanabe J, Hata H, Jobo T, Kawaguchi M, Hattori M, et al. Significance of progesterone receptor-A and -B expressions in endometrial adenocarcinoma. J Steroid Biochem Mol Biol. 2004;92(3):111–8. pmid:15555905.
  15. 15. Jongen V, Briet J, de Jong R, ten Hoor K, Boezen M, van der Zee A, et al. Expression of estrogen receptor-alpha and -beta and progesterone receptor-A and -B in a large cohort of patients with endometrioid endometrial cancer. Gynecol Oncol. 2009;112(3):537–42. pmid:19108875.
  16. 16. Zannoni GF, Monterossi G, De Stefano I, Gargini A, Salerno MG, Farulla I, et al. The expression ratios of estrogen receptor alpha (ERalpha) to estrogen receptor beta1 (ERbeta1) and ERalpha to ERbeta2 identify poor clinical outcome in endometrioid endometrial cancer. Hum Pathol. 2013;44(6):1047–54. pmid:23266443.
  17. 17. Santacana M, Yeramian A, Velasco A, Bergada L, Gatius S, Garcia V, et al. Immunohistochemical features of post-radiation vaginal recurrences of endometrioid carcinomas of the endometrium: role for proteins involved in resistance to apoptosis and hypoxia. Histopathology. 2012;60(3):460–71. pmid:22276609.
  18. 18. Soslow RA, Wethington SL, Cesari M, Chiappetta D, Olvera N, Shia J, et al. Clinicopathologic analysis of matched primary and recurrent endometrial carcinoma. The American journal of surgical pathology. 2012;36(12):1771–81. pmid:23154767.
  19. 19. Tangen IL, Werner HM, Berg A, Halle MK, Kusonmano K, Trovik J, et al. Loss of progesterone receptor links to high proliferation and increases from primary to metastatic endometrial cancer lesions. Eur J Cancer. 2014;50(17):3003–10. pmid:25281525.
  20. 20. Jeronimo C, Monteiro P, Henrique R, Dinis-Ribeiro M, Costa I, Costa VL, et al. Quantitative hypermethylation of a small panel of genes augments the diagnostic accuracy in fine-needle aspirate washings of breast lesions. Breast cancer research and treatment. 2008;109(1):27–34. pmid:17549626
  21. 21. Ehrlich M, Woods CB, Yu MC, Dubeau L, Yang F, Campan M, et al. Quantitative analysis of associations between DNA hypermethylation, hypomethylation, and DNMT RNA levels in ovarian tumors. Oncogene. 2006;25(18):2636–45. pmid:16532039; PubMed Central PMCID: PMC1449872.
  22. 22. Eads CA, Lord RV, Wickramasinghe K, Long TI, Kurumboor SK, Bernstein L, et al. Epigenetic patterns in the progression of esophageal adenocarcinoma. Cancer Res. 2001;61(8):3410–8. pmid:11309301.
  23. 23. Ogino S, Kawasaki T, Brahmandam M, Cantor M, Kirkner GJ, Spiegelman D, et al. Precision and performance characteristics of bisulfite conversion and real-time PCR (MethyLight) for quantitative DNA methylation analysis. The Journal of molecular diagnostics: JMD. 2006;8(2):209–17. pmid:16645207; PubMed Central PMCID: PMC1867588.
  24. 24. Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Annual review of pathology. 2013;8:277–302. pmid:23092187.
  25. 25. Burcombe R, Wilson GD, Dowsett M, Khan I, Richman PI, Daley F, et al. Evaluation of Ki-67 proliferation and apoptotic index before, during and after neoadjuvant chemotherapy for primary breast cancer. Breast cancer research: BCR. 2006;8(3):R31. pmid:16790076; PubMed Central PMCID: PMC1557736.
  26. 26. Penault-Llorca F, Abrial C, Raoelfils I, Chollet P, Cayre A, Mouret-Reynier MA, et al. Changes and predictive and prognostic value of the mitotic index, Ki-67, cyclin D1, and cyclo-oxygenase-2 in 710 operable breast cancer patients treated with neoadjuvant chemotherapy. The oncologist. 2008;13(12):1235–45. pmid:19091781.
  27. 27. Gallaher J, Anderson AR. Evolution of intratumoral phenotypic heterogeneity: the role of trait inheritance. Interface focus. 2013;3(4):20130016. pmid:24511380; PubMed Central PMCID: PMC3915835.
  28. 28. Calascibetta A, Cabibi D, Rausa L, Aragona F, Barresi E, Martorana A, et al. Difference in Ki67 and thymidylate synthase expression in primary tumour compared with metastatic nodes in breast cancer patients. Nucleosides, nucleotides & nucleic acids. 2006;25(9–11):1193–6. pmid:17065089.
  29. 29. Yano K, Okamura T, Yoshida Y, Ichiyoshi Y, Yasumoto K. Proliferative activity of esophageal carcinomas and their lymph node metastases: comparison using argyrophilic nucleolar organizer region staining. Journal of surgical oncology. 1997;65(4):274–9. pmid:9274793.
  30. 30. Brodsky AS, Fischer A, Miller DH, Vang S, MacLaughlan S, Wu HT, et al. Expression profiling of primary and metastatic ovarian tumors reveals differences indicative of aggressive disease. PLoS One. 2014;9(4):e94476. pmid:24732363; PubMed Central PMCID: PMC3986100.
  31. 31. Jalava P, Kuopio T, Juntti-Patinen L, Kotkansalo T, Kronqvist P, Collan Y. Ki67 immunohistochemistry: a valuable marker in prognostication but with a risk of misclassification: proliferation subgroups formed based on Ki67 immunoreactivity and standardized mitotic index. Histopathology. 2006;48(6):674–82. pmid:16681683.
  32. 32. Reid-Nicholson M, Iyengar P, Hummer AJ, Linkov I, Asher M, Soslow RA. Immunophenotypic diversity of endometrial adenocarcinomas: implications for differential diagnosis. Mod Pathol. 2006;19(8):1091–100. pmid:16648864.
  33. 33. Fujimoto J, Ichigo S, Hirose R, Sakaguchi H, Tamaya T. Clinical implication of expression of progesterone receptor form A and B mRNAs in secondary spreading of gynecologic cancers. J Steroid Biochem Mol Biol. 1997;62(5–6):449–54. pmid:9449248.
  34. 34. Fujimoto J, Sakaguchi H, Aoki I, Toyoki H, Tamaya T. Clinical implications of the expression of estrogen receptor-alpha and -beta in primary and metastatic lesions of uterine endometrial cancers. Oncology. 2002;62(3):269–77. pmid:12065875.
  35. 35. Dieci MV, Barbieri E, Piacentini F, Ficarra G, Bettelli S, Dominici M, et al. Discordance in receptor status between primary and recurrent breast cancer has a prognostic impact: a single-institution analysis. Annals of oncology: official journal of the European Society for Medical Oncology / ESMO. 2013;24(1):101–8. pmid:23002281.
  36. 36. Amir E, Clemons M, Purdie CA, Miller N, Quinlan P, Geddie W, et al. Tissue confirmation of disease recurrence in breast cancer patients: pooled analysis of multi-centre, multi-disciplinary prospective studies. Cancer treatment reviews. 2012;38(6):708–14. pmid:22178456.
  37. 37. Hoefnagel LD, van de Vijver MJ, van Slooten HJ, Wesseling P, Wesseling J, Westenend PJ, et al. Receptor conversion in distant breast cancer metastases. Breast cancer research: BCR. 2010;12(5):R75. pmid:20863372; PubMed Central PMCID: PMC3096964.
  38. 38. Esteller M. Epigenetics in cancer. N Engl J Med. 2008;358(11):1148–59. pmid:18337604.
  39. 39. Sasaki M, Kotcherguina L, Dharia A, Fujimoto S, Dahiya R. Cytosine-phosphoguanine methylation of estrogen receptors in endometrial cancer. Cancer Res. 2001;61(8):3262–6. pmid:11309276.
  40. 40. Wen DX, Xu YF, Mais DE, Goldman ME, McDonnell DP. The A and B isoforms of the human progesterone receptor operate through distinct signaling pathways within target cells. Mol Cell Biol. 1994;14(12):8356–64. pmid:7969170; PubMed Central PMCID: PMC359374.
  41. 41. Wargon V, Fernandez SV, Goin M, Giulianelli S, Russo J, Lanari C. Hypermethylation of the progesterone receptor A in constitutive antiprogestin-resistant mouse mammary carcinomas. Breast Cancer Res Treat. 2011;126(2):319–32. pmid:20440553.
  42. 42. Smid-Koopman E, Blok LJ, Kuhne LC, Burger CW, Helmerhorst TJ, Brinkmann AO, et al. Distinct functional differences of human progesterone receptors A and B on gene expression and growth regulation in two endometrial carcinoma cell lines. J Soc Gynecol Investig. 2003;10(1):49–57. pmid:12517594.
  43. 43. Dai D, Wolf DM, Litman ES, White MJ, Leslie KK. Progesterone inhibits human endometrial cancer cell growth and invasiveness: down-regulation of cellular adhesion molecules through progesterone B receptors. Cancer Res. 2002;62(3):881–6. pmid:11830547.
  44. 44. Sakaguchi H, Fujimoto J, Hong BL, Nakagawa Y, Tamaya T. Drastic decrease of progesterone receptor form B but not A mRNA reflects poor patient prognosis in endometrial cancers. Gynecol Oncol. 2004;93(2):394–9. pmid:15099952.
  45. 45. Kumar NS, Richer J, Owen G, Litman E, Horwitz KB, Leslie KK. Selective down-regulation of progesterone receptor isoform B in poorly differentiated human endometrial cancer cells: implications for unopposed estrogen action. Cancer Res. 1998;58(9):1860–5. pmid:9581825.
  46. 46. Pijnenborg JM, Romano A, Dam-de Veen GC, Dunselman GA, Fischer DC, Groothuis PG, et al. Aberrations in the progesterone receptor gene and the risk of recurrent endometrial carcinoma. J Pathol. 2005;205(5):597–605. pmid:15726651.
  47. 47. De Vivo I, Huggins GS, Hankinson SE, Lescault PJ, Boezen M, Colditz GA, et al. A functional polymorphism in the promoter of the progesterone receptor gene associated with endometrial cancer risk. Proc Natl Acad Sci U S A. 2002;99(19):12263–8. pmid:12218173; PubMed Central PMCID: PMC129433.
  48. 48. Ghabreau L, Roux JP, Niveleau A, Fontaniere B, Mahe C, Mokni M, et al. Correlation between the DNA global methylation status and progesterone receptor expression in normal endometrium, endometrioid adenocarcinoma and precursors. Virchows Archiv: an international journal of pathology. 2004;445(2):129–34. pmid:15221375.
  49. 49. Janzen DM, Rosales MA, Paik DY, Lee DS, Smith DA, Witte ON, et al. Progesterone receptor signaling in the microenvironment of endometrial cancer influences its response to hormonal therapy. Cancer Res. 2013;73(15):4697–710. pmid:23744837; PubMed Central PMCID: PMC3752407.