Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Mar 3.
Published in final edited form as: J Pathol. 2012 Jul 18;228(1):20–30. doi: 10.1002/path.4056

Use of mutation profiles to refine the classification of endometrial carcinomas

Melissa K McConechy 1,#, Jiarui Ding 2,4,#, Maggie CU Cheang 3, Kimberly Wiegand 1, Janine Senz 1, Alicia Tone 1, Winnie Yang 1, Leah Prentice 1, Kane Tse 6, Thomas Zeng 6, Helen McDonald 6, Amy P Schmidt 9, David G Mutch 10, Jessica N McAlpine 8, Martin Hirst 6,7, Sohrab P Shah 2,4, Cheng-Han Lee 5, Paul J Goodfellow 9, C Blake Gilks 1,5,11, David G Huntsman 1,2,11
PMCID: PMC3939694  NIHMSID: NIHMS536965  PMID: 22653804

Abstract

The classification of endometrial carcinomas is based on pathological assessment of tumour cell type; the different cell types (endometrioid, serous, carcinosarcoma, mixed, and clear cell) are associated with distinct molecular alterations. This current classification system for high-grade subtypes, in particular the distinction between high-grade endometrioid (EEC-3) and serous carcinomas (ESC), is limited in its reproducibility and prognostic abilities. Therefore, a search for specific molecular classifiers to improve endometrial carcinoma subclassification is warranted. We performed target enrichment sequencing on 393 endometrial carcinomas from two large cohorts, sequencing exons from the following 9 genes; ARID1A, PPP2R1A, PTEN, PIK3CA, KRAS, CTNNB1, TP53, BRAF and PPP2R5C. Based on this gene panel each endometrial carcinoma subtype shows a distinct mutation profile. EEC-3s have significantly different frequencies of PTEN and TP53 mutations when compared to low-grade endometrioid carcinomas. ESCs and EEC-3s are distinct subtypes with significantly different frequencies of mutations in PTEN, ARID1A, PPP2R1A, TP53, and CTNNB1. From the mutation profiles we were able to identify subtype outliers, i.e. cases diagnosed morphologically as one subtype but with a mutation profile suggestive of a different subtype. Careful review of these diagnostically challenging cases suggested that the original morphological classification was incorrect in most instances. The molecular profile of carcinosarcomas suggests two distinct mutation profiles for these tumours; endometrioid-type (PTEN, PIK3CA, ARID1A, KRAS mutations), and serous-type (TP53 and PPP2R1A mutations). While this nine gene panel does not allow for a purely molecularly based classification of endometrial carcinoma, it may prove useful as an adjunct to morphological classification and serve as an aid in the classification of problematic cases. If used in practice, it may lead to improved diagnostic reproducibility and may also serve to stratify patients for targeted therapeutics.

Keywords: Endometrial carcinoma, uterine, mutation profiles, endometrioid, serous, carcinosarcoma, classification

Introduction

The incidence of endometrial carcinoma is rising in the western world, and it is currently the most common type of gynaecological carcinoma [1]. This increase has been linked to increased obesity, increased life expectancy and tamoxifen use in women [2]. The classical pathogenic dualistic model proposed by Bokhman in 1983, placed endometrial carcinomas into one of two groups; estrogen-dependent endometrioid carcinomas, and estrogen-independent non-endometrioid carcinomas [3]. The classification of endometrial carcinomas used in clinical practice is based on histopathological assessment to determine cell type and grade [4-5], and is used in guiding therapy [6-7]. Endometrioid endometrial carcinomas (EECs) represent 70-80% of cases, are generally low grade (grade 1 or 2) with favourable prognosis, and most are cured by hysterectomy alone [8-9]. However, less common high-grade (grade 3) endometrioid carcinomas (EEC-3) have a significantly worse prognosis [5, 10]. The remaining 20-30% of non-endometrioid subtypes consist mostly of serous, and less commonly carcinosarcoma (previously known as MMMT or mixed malignant mullerian tumours), mixed histology, and clear cell carcinomas. These non-endometrioid tumours are not generally graded in the WHO grading system [6], are considered high-grade, as they are associated with poor outcomes [11]. Recent reports have shown the current pathological classification and grading system of high-grade endometrial carcinomas is limited in both reproducibility and prognostic ability [10, 12-14].

Molecular alterations in the PI3K/AKT, MAPK, and WNT signalling pathways have been implicated in the pathogenesis of specific endometrial carcinoma subtypes [15-18]. Thus there is a rationale for using mutational profiles in the classification of these tumours. EECs are molecularly recognized by frequent mutations in PTEN, PIK3CA, KRAS, CTNNB1, FGFR2, and microsatellite instability (MSI) [8, 19-22]. Recent studies have identified mutations in ARID1A [24], PIK3R1 [23], and PIK3R2 [24] in EECs. Endometrial serous carcinomas (ESCs), and carcinosarcomas characteristically do not harbour a high frequency of these mutations, however, TP53 [8, 19-20], and PPP2R1A [25-26] mutations are known to be common in ESC. TP53 mutations are also detected in carcinosarcomas [27] and EEC-3s [10, 28].

Next-generation sequencing technologies has allowed sequencing of multiple genes and samples simultaneously [24], making large mutational studies achievable. As no single gene is a sensitive or specific marker for endometrial carcinoma subtypes, it is likely that the analysis of gene panels will be needed to guide subclassification. The aim of this study was to determine the mutation profiles of a large series of endometrial carcinomas, based on oncogenes and tumour suppressor genes known to be important in carcinogenesis, in an attempt to improve the classification of endometrial carcinomas.

Materials and Methods

Patient Samples

We obtained 152 endometrial tumours, and 90 corresponding buffy coat specimens originating from the BC Cancer Agency and Vancouver General Hospital via the OvCaRe Tissue Biobank repository, Vancouver, BC, Canada. Patients were informed for written consent, and research ethics approved as previously described [25]. An additional 260 endometrial tumour DNA samples were obtained from Washington University, St. Louis, Missouri. The endometrial subtype, grade and microsatellite instability data was previously determined in these cases. All samples from both centers’ have undergone review by gynaecological pathologists.

Exon Sequencing

Genomic DNA (500ng) was used for indexed Illumina library construction [29], then underwent targeted enrichment using biotinylated RNA capture probes generated from cDNA clones or PCR amplicons [30] representing exons of ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, BRAF, TP53, and PPP2R5C and sequenced using Illumina (GAIIx).

Bioinformatics Analysis

Short reads were aligned to the human genome (hg18) using the BWA aligner v0.5.9 [31]. A Random Forest classifier trained on validated SNVs was used to remove false-positive calls [32]. SNVs in the Catalogue of Somatic Mutations in Cancer (COSMIC) [33] were considered to be true positives, so a 99% cutoff threshold was selected (Figure S1). Mean coverage was plotted for cases with and without mutations (Figure S2). Details found in Supplementary materials and methods.

DNA validations

Select predicted SNVs were validated using Sanger sequencing as previously described [25]. See Supplementary materials and methods.

Identifying outlier cases

Outliers were identified by observing mutation profiles that did not fit the original diagnosed histological subtype; defined as ESC with PTEN and/or ARID1A mutations, and low-grade EECs with only TP53 and/or PPP2R1A mutations. With the goal of comparing mutational outliers with immuno-profiles, formalin-fixed embedded paraffin blocks were only available for 147/156 Vancouver cases, for the construction of a Tissue Microarray (TMA). For details see Supplementary materials and methods. These cases were used for the characterization of mutational outliers, by correlating with morphology and immunohistochemistry (IHC), and retrospectively reviewed by two independent pathologists, using the full hysterectomy case, without knowledge of mutation or IHC data.

Statistical Analysis

Fisher exact tests and multivariable logistic regression analysis were used to test the significance of associations between mutations within subtypes. All tests were two-tailed and p-value < 0.05 were considered significant. Fisher exact tests were not adjusted for multiple comparisons. The multivariable logistic regression model used step-wise selection based on the likelihood ratio test, with all genes included. The Hosmer-Lemeshow test was used to assess the goodness-of-fit of the estimated logistic regression models.

Results

To determine the mutation frequencies in various subtypes of endometrial carcinomas, we used exon capture sequencing of ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, BRAF, TP53, and PPP2R5C. This resulted in the detection of somatic nonsynonymous missense, truncating, indels (insertions/deletions), and splice site mutations in 90.1% (353/392) of cases. The characteristics of the endometrial carcinomas, with histology subtypes and grade, are summarized in Table 1. We have stratified these carcinomas into low-grade (grade 1 and 2) EECs, EEC-3, ESC, carcinosarcoma, mixed, and undifferentiated, based on routine histopathological assessment, to determine the differences in mutational profiles. All mutational data are summarized in Table S1. The mutation frequencies of ARID1A, PTEN, PIK3CA, PPP2R1A, TP53, and CTNNB1 are significantly different across four subtypes of endometrial carcinomas (Table 2).

Table 1.

Summary of all endometrial carcinoma subtypes.

All Subtypes
Endometrioid 306
Grade 1 169
Grade 2 107
Grade 3 30
Serous 37
Mixed* 4
Undifferentiated 3
Carcinosarcoma 42
Total 392
*

Includes one cases as mixed serous and endometrioid carcinoma, one case mixed G2 and G3 endometrioid and clear cell carcinoma, and two cases as mixed serous and clear cell carcinoma.

Table 2.

The frequency of mutations within all endometrial subtypes.

Low-Grade Endometrioid (G1 and 2) (n=276) High-Grade Endometrioid (G3) (n=30) Serous (n=37) Carcinosarcoma (n=42) p-value across all subtypes (chi-squared test)
PTEN 185 (67.0%) 27 (90.0%) 1 (2.7%) 14 (33.3%) 4.63E-17
PIK3CA 105 (38.0%) 17 (56.7%) 10 (27.0%) 12 (28.6%) 0.0480
ARID1A 129 (46.7%) 18 (60.0%) 4 (10.8%) 10 (23.8%) 5.77E-06
KRAS 46 (16.6%) 8 (26.7%) 3 (8.1%) 7 (16.7%) 0.2434
CTNNB1 66 (23.8%) 6 (20.0%) 1 (2.7%) 2 (4.8%) 1.19E-03
PPP2R1A 19 (6.9%) 3 (10.0%) 16 (43.2%) 9 (21.4%) 1.50E-09
TP53 28 (10.1%) 9 (30.0%) 25 (67.6%) 27 (64.3%) 2.79E-23
BRAF 8 (2.9%) 2 (6.7%) 2 (5.4%) 1 (2.4%) 0.6186
PPP2R5C 1 (0.4%) 2 (6.7%) 0 (0%) 0 (0%) 0.002

Bold indicates significant p-values <0.05

High-grade and low-grade endometrioid carcinomas have similar mutation profiles but differ in frequencies of TP53 mutations

Low-grade EECs have high to moderate frequencies of mutations in PTEN, ARID1A, PIK3CA, and CTNNB1 (Table 2), with a higher frequency of mutations of PTEN, ARID1A, PIK3CA, KRAS, PPP2R1A and TP53 seen in EEC-3s (Table 2). The comparison of mutations in low-grade EEC and EEC-3 showed that PTEN (p=0.0111) and TP53 (p=0.0046) mutation frequencies are significantly different (Table 3). Multivariable logistic regression also revealed that PTEN (p=0.007) and TP53 (p<0.001) mutations significantly distinguish EEC-3 from low-grade EEC (Table 4).

Table 3.

Univariate Fisher exact test (p-values) to show significant differences between mutation profiles of each endometrial carcinoma subtypes.

Low-Grade Endometrioid vs High-Grade Endometrioid Low-Grade Endometrioid vs Serous High-Grade Endometrioid vs Serous High-Grade Endometrioid vs Carcinosarcoma Serous vs Carcinosarcoma
PTEN 0.0111 6.58E-15 2.57E-14 1.09E-06 4.30E-04
PIK3CA 0.0522 0.2091 0.0235 0.0276 1.0000
ARID1A 0.1826 1.38E-05 2.42E-05 0.0030 0.1522
KRAS 0.2000 0.2328 0.0525 0.3814 0.3215
CTNNB1 0.8211 1.23E-03 0.0394 0.0602 1.0000
PPP2R1A 0.4630 4.96E-08 2.95E-03 0.3365 0.0526
TP53 4.62E-03 8.56E-14 3.17E-03 0.0080 0.8151
BRAF 0.2555 0.3352 1.0000 0.5669 0.5972
PPP2R5C 0.0263 1.0000 0.1967 0.1702 NA

Bold indicates significant p-values <0.05

Table 4.

Multivariable logistic regression analysis of gene mutations between endometrial carcinoma subtypes. Reported values are only the most significant genes selected by the step-wise selection method based on the Likelihood ratio test.

Gene (Marker) Low-grade Endometrioid (n=276) vs. High-grade Endometrioid (n=30) Low-grade Endometrioid (n=276) vs. =Serous (n=37) High-grade Endometrioid (n=30) vs. Serous (n=37) High-grade Endometrioid (n=30) vs. Carcinosarcoma (n=42) Serous (n=37) vs. Carcinosarcoma (n=42)

p-value Odds ratio* to high-grade endometrioid p-value Odds ratio* to serous p-value Odds ratio* to serous p-value Odds ratio* to carinosarcoma p-value Odds ratio* to carinosarcoma

PTEN 0.007 5.61 (1.6-19.7) 1.89E-04 0.02 (0.002-0.14) 3.75E-05 0.05 (0.01-0.22) 6.24E-03 19.41 (2.3-162.6)
PIK3CA
ARID1A 0.080 0.3 (0.08-1.2)
KRAS
CTNNB1
PPP2R1A 2.7E-04 13.28 (3.3-53.4) 0.0736 5.12 (0.86-30.7) 0.0446 0.32 (0.1-0.97)
TP53 7.04E-04 4.95 (2.0-12.5) 7.64E-05 7.64 (2.8-20.9)
BRAF 1.40E-02 18.9 (1.8-196.7)
PPP2R5C
*

Odds ratio (95% CI)

Endometrial serous carcinomas show a distinct mutation profile

Of 37 ESCs, high frequencies of mutations were found in TP53, PPP2R1A, and PIK3CA (Table 2). TP53 and/or PPP2R1A mutations were found in 28/37 (75.7%) of ESCs, accounting for the majority of aberrations in this subtype (Figure 1). The comparison of EEC-3 to ESC revealed significantly different mutation frequencies for ARID1A, PTEN, PIK3CA, CTNNB1, PPP2R1A, and TP53 (p <0.05) (Table 3). Low frequencies to zero mutation events were noted for some genes common in both ESCs and EEC-3. In an attempt to keep all the multivariate analyses consistent across the subtype comparisons, we included the same list of genes in the logistic regression model building between EEC-3 and ESC. As a result, there was no one reliable multivariable logistic regression model built, based on the mutation markers, to distinguish between these two subtypes (Table 4). As expected, the mutational profiles of low-grade EEC and ESC are significantly different (Table 3). Multivariable logistic regression shows, PTEN (p <0.001) with a trend of ARID1A (p=0.08) mutations associated with low-grade EEC, whereas PPP2R1A and TP53 (p <0.001) are associated with ESC (Table 4).

Figure 1. Mutation profiles of endometrial subtypes.

Figure 1

A. Low-grade endometrioid carcinoma, including grade 1 and 2 tumours (n=276); B. High-grade endometrioid carcinoma, grade 3 tumours (n=30); C. Serous carcinoma (n=37); D. Carcinosarcoma (n=42), (+) indicates carcinosarcomas with heterologous differentiation elements; E. Undifferentiated and mixed histology subtypes, (a) undifferentiated carcinomas, (b) mixed low-grade EEC with serous carcinoma, (c) mixed endometrioid and clear cell carcinoma, (d) mixed serous and clear cell carcinoma. Rows indicate genes, columns represent tumour cases. Coloured bars indicate mutations’ including; missense, truncating, indels and splice site mutations. Grey bars indicate no mutations were detected. (*) indicates serous carcinoma outliers with ARID1A mutations; (#) indicates low-grade EECs and EEC-3s mutation outliers with serous-type mutations (TP53 or PPP2R1A).

Cases with discordant morphological diagnosis and mutational profiles

As discussed, ESCs were found to have a high frequency of mutations in TP53 and PPP2R1A (Figure 1). From the mutation profiles we identified three histology-defined ESC cases with ARID1A and PTEN mutations and lacked TP53 mutations, a profile more indicative of EECs (Figure 1). Other studies have not found ARID1A or PTEN mutations in ESCs, however there have been limited studies testing for ARID1A mutations in endometrial carcinomas [34-36]. On independent histopathological review of these three cases, all were mixed tumours consisting predominantly of ESC, but with minor components of low-grade EEC in two cases, and EEC-3 with clear cell carcinoma in one case (Table 5). For the two mixed ESC and low-grade EEC cases, we confirmed the section of tumour sample used for DNA extraction and subsequent sequencing exclusively contained the ESC component (Figure 2); however it harboured mutations with an endometrioid profile. Immunostaining is recommended for use in diagnostically problematic cases [37], although not universally used. These three cases showed a non-serous IHC profile; p53 normal expression and p16 negative expression, while one expressed ER and PR (Table 5).

Table 5.

Outlier cases with pathological review, IHC and mutation profile.

ID 841 1120 220 895 511 1034 611
Original Subtype Serous carcinoma Serous carcinoma Serous carcinoma Low-grade endometrioid carcinoma Low-grade endometrioid carcinoma Low-grade endometrioid carcinoma Low-grade endometrioid carcinoma
Review mixed serous (80%) and low-grade endometrioid carcinoma, with adjacent endometrium showing focal complex atypical hyperplasia mixed serous (60%) and low-grade endometrioid carcinoma, with adjacent endometrium showing complex atypical hyperplasia Grade 3 endometrioid with clear cell changes Grade 2 endometrioid (extensively myometrial-invasive and LVI) Grade 3 endometrioid Mixed low-grade (G2) endometrioid and serous carcinoma Serous carcinoma
p53-IHCa 1 1 1 2 2 2 2
ER-IHCb 0 1 0 1 1 1 1
P16-IHCb 0 0 0 0 0 1 1
PR-IHCb 0 1 0 1 1 1 NA
PTEN-IHCc 1 1 0 0 0 0 0
ARID1A p.Q420*, p.R1335* p.Q2176fs p.Q548fs, p.G1847fs
PTEN p.L265fs splice site acceptor
PIK3CA p.G106V, p.V344M p.Q546K, p.H1047Y Y1021C
KRAS p.G13D p.G12A
PPP2R1A p.R182W p.P179L
TP53 p.R282W p.H193L p.R248Q p.S241F
CTNNB1
BRAF p.A526V, p.P403fs
PPP2R5C
a

Scoring; 0= loss of expression, 1= normal expression, 2= over-expression

b

Scoring; 0= no expression, 1= over-expression

c

Scoring; 1=normal expression, 0= loss of expression

Figure 2. A case originally diagnosed as serous carcinoma, but with an ARID1A mutation and no TP53 mutation, is a mixed low-grade endometrioid and serous carcinoma (case #1120).

Figure 2

A. A mix of a grade 1 endometrioid (left half ) and high-grade serous (right half) carcinoma, 40X magnification; B. High power (100X) image of histologically distinct low-grade endometrioid carcinoma; C. High power (100X) images of serous carcinoma component, of which the sampling of tumour was used for mutation sequencing; D. Atypical complex hyperplasia in the background endometrium 40X magnification.

We also identified four outlier low-grade EECs that contained TP53 mutations and lacked PTEN mutations, which were also diagnostically challenging cases. Upon review, two cases showed morphological features of serous, and one case was re-classified from low-grade EEC to EEC-3. One outlier remained classified as low-grade EEC, however it was noted that this case showed extensive myometrial invasion and widespread lymphovascular invasion. By IHC, abnormal p53 expression was confirmed in all cases. All were, however, ER-positive with PTEN loss of expression, features found primarily in EECs. In two of these cases, p16 was strongly expressed (Table 5). In summary, these seven outlier cases showed features intermediate between ESC and EEC in morphological, IHC and genetic analysis (Table 5, Table S2).

We also performed unsupervised hierarchical clustering analysis on the 147 cases with IHC and mutational status (Figure S3, Table S2). This shows most low-grade EEC and EEC-3 subtypes cluster together, while the remaining EEC-3, serous and mixed cases are scattered. The mutational outliers with the diagnosed subtype are indicated, as well as the new classification.

Carcinosarcomas show either an endometrioid or serous mutation profile

Endometrial carcinosarcomas are relatively rare, and their classification as an endometrial carcinoma subtype or as a distinct entity is under debate [38]. In our analysis, of carcinosarcomas we found mutations in TP53, PTEN, PIK3CA, ARID1A, and PPP2R1A (Table 2). Two subgroups of carcinosarcomas were identified; one group characterized by mutations in PTEN and ARID1A (endometrioid-type), and a second group with TP53 and PPP2R1A mutations more similar to ESC (Figure 1). Heterologous differentiation of the sarcomatous component was observed in a subset of tumors from both groups. Histopathological reviews of cases were not available; therefore it was not possible to correlate morphological features and mutational profiles of endometrioid-like or serous-like in the epithelial components of these tumours.

Mutations involving signalling pathways in endometrial carcinomas

By mutational analysis of multiple genes, it is possible to identify different mutations involving a single signalling pathway that may be functionally equivalent, and to examine the relationship between mutations involving different genes/pathways. Mutations in the PI3K and MAPK signalling pathways are known to be important in EECs, therefore we further examined the prevalence of mutations in PTEN, PIK3CA, KRAS, ARID1A and CTNNB1. We found 211/276 (76.5%) low-grade EECs have PTEN and/or PIK3CA mutations (Figure 1). Co-existent PTEN and PIK3CA mutations were identified in 79/276 (28.6%) low-grade EECs, and 16/30 (53.3%) EEC-3s (p=0.0112). AR1D1A mutations have recently been identified in low-grade EECs; however the relationship of these mutations with other pathways such as PI3K and WNT has not been examined [34]. Of the low-grade EECs with ARID1A mutations, 112/129 (86.8%) have mutations within PTEN and/or PIK3CA (p=0.0002). EEC-3s with ARID1A mutations (n=18) all have PTEN mutations, and 13/18 (72.2%) also have PIK3CA mutations. Thus there is a significant association between ARID1A and PTEN/PIK3CA mutations.

Microsatellite instability

MSI is a feature of the endometrioid subtype, therefore we determined the MSI status of 241/276 low-grade EECs and 13/30 EEC-3s. We found 97/241 (40.2%) of the low-grade EECs are MSI positive, compared to 8/13 (61.5%) of EEC-3 (Table S1).

Discussion

Endometrial carcinoma is a heterogeneous disease, comprised of multiple subtypes with differing risk factors, precursor lesions, and outcomes. Lack of reproducibility in histopathological diagnosis of endometrial carcinoma subtypes has hindered progress. For example, while some studies have found that EEC-3 and ESC have different outcomes [39], other studies have not [10]. This difference may reflect inclusion of different cases, based on subtly different diagnostic criteria, within these cohorts. Robust and reproducible diagnostic categories are an important first step in moving towards subtype-specific treatment, as is happening for ovarian carcinoma [40-41]. However in the case of endometrial carcinoma, it is likely that molecular markers will be needed to improve the suboptimal performance of conventional histopathological assessment [42]. With the advent of next-generation sequencing technologies, the molecular profiles of many tumour cell types are being extensively characterized. The knowledge of these mutation profiles can potentially be used diagnostically for subclassification, and to identify relevant targets for the development/deployment of targeted therapeutics. In this study, we performed exon capture sequencing of nine genes in two large cohorts of endometrial carcinomas, revealing differing mutational landscapes for endometrial carcinoma subtypes.

As demonstrated in previous studies, we identified high frequencies of mutations within PTEN, PIK3CA, ARID1A, KRAS and CTNNB1, and lack of TP53 mutations in low-grade EECs. EEC-3s demonstrate a similar pattern of mutations, but with a significantly increased frequency of TP53 mutations. High frequencies of PTEN mutations in EECs confirm this is an early driver event in tumour progression. Our results show that the frequency of MSI cases is similar in low-grade EEC and EEC-3, which supports the view that the majority of EEC-3s have progressed from low-grade EEC [10].

Recent studies identified a high frequency of concurrent PTEN and PIK3CA mutations in endometrial carcinomas [15, 24], but not in any other tumour type investigated to date [24]. In this study, we also observed this phenomenon in low-grade EECs and EEC-3s, but not in ESC or carcinosarcoma. We have determined that in low-grade EECs and EEC-3s, ARID1A mutations are significantly associated with concurrent mutations in PTEN and PIK3CA, a novel finding suggesting a cooperative role of these pathways in EEC tumourigenesis.

ESCs have frequent TP53 and PPP2R1A mutations, and lack mutations in PTEN, ARID1A and CTNNB1, a mutational profile distinct from that of EECs. While it was not possible to classify tumours solely based on this nine gene mutation panel, we were able to use the mutation profile as a diagnostic adjunct for morphological subclassification in individual cases. This is an attractive prospect given the significant problems in distinguishing EEC-3 and ESC highlighted in recent studies [5, 13-14, 28, 37, 43]. We observed mutational outliers where the original diagnosis did not fit the mutation profile, specifically ESC cases with ARID1A mutations, and low-grade EECs with only TP53 mutations. In most of these outlier cases, retrospective review by two independent pathologists resulted in reclassification, agreeing with the subtype-specific mutation patterns rather than the original diagnosis.

It has previously been proposed that ESC may arise through two different tumourigenic pathways, i.e. from progression through hyperplasia and low-grade EEC, or arising via high-grade endometrial intraepithelial carcinoma, in an estrogen-independent pathway [44]. In this study, we observed two tumours initially diagnosed as ESC that showed an endometrioid mutation profile. On retrospective review the diagnosis for both was changed to mixed serous and endometrioid. This observation is not novel but does give further support to ESCs arising in some cases by an alternative molecular pathway, rather than the classical Type 2 pathway (Figure 3, Figure S4) [9]. This further suggests that the classification of endometrial carcinomas cannot be encompassed by a simple dualistic model. In particular, the high-grade subtypes show considerable heterogeneity not reflected adequately in a Type 1 versus Type 2 model. Future studies will be required to address the following issues: 1. How reproducible is molecularly supported subtype diagnoses? 2. If diagnoses can be made reproducibly, do subtypes show significant differences in stage at diagnosis, pattern of spread, prognosis or response to treatment? Only after those questions are addressed can subtype-specific management move forward, and mutation-based treatment decisions can be made for challenging diagnoses.

Figure 3. Mutational analysis may be an effective tool to classify morphologically problematic cases into biologically relevant treatment groups.

Figure 3

Intermediate high-grade cell types tend to be diagnostically challenging cases, often with multiple morphological features of endometrioid and/or serous carcinomas. The addition of mutation profiles can lead to reproducible diagnosis and the future of mutation-based treatment decisions for targeted therapeutics. Blue and red colours indicate distinct mutation profiles for low-grade EEC and serous carcinomas. Yellow indicates the cases were the mutational profiles will aid in separating out the appropriate histological subtype and dictate appropriate treatment options for patients.

We also investigated the molecular profiles of carcinosarcomas. These tumours are generally rare with poor prognosis [45], and are composed of a mixture of carcinomatous and sarcomatous elements [46]. While previous studies have not identified a high number of mutations in this subtype [47], we have shown a moderate frequency of mutations in the majority of genes sequenced. This discrepancy may be due to limited exon sequencing in previous studies; in the current study all exons of these genes were interrogated. Two patterns of mutations were observed; an endometrioid-type mutation profile (ARID1A, PTEN, PIK3CA, KRAS) or a serous mutation profile (TP53, PPP2R1A). This suggests a dualistic molecular evolution of carcinosarcomas with an endometrioid-like or serous-like mutation pattern (Figure 3). Further validation studies will be necessary to determine if these molecular profiles are associated with different morphological features in the carcinomatous or sarcomatous components, or are associated with outcome differences.

We acknowledge that there are limitations of this study; we were unable to perform full histopathological reviews of many cases, including all carcinosarcomas. There were also limited numbers of cases of EEC-3 and ESC in this study, therefore independent validation studies, linked with outcome [48], will be needed in these tumour types. There is also uncertainty about the sensitivity of the exon capture method, and false negatives are likely to be present in this data set. The TCGA endometrial sequencing effort will prove to be useful in validating the observations of this study.

In conclusion, we have identified distinct molecular profiles that may aid in endometrial carcinoma classification leading to more reproducible diagnoses. Although endometrial carcinoma subtypes diagnoses and grade are currently used in guiding patient management, mutational analysis is emerging as a realistic option in clinical practice. In the future, we predict that the mutational classification of endometrial carcinomas will become an important tool in diagnosis, guiding mutation-based targeted treatment decisions. Mutation profiles are already being applied in other cancers for selecting targeted therapeutics, for example BRAF inhibitors in malignant melanoma [49] and BRAF and EGFR targeting in colorectal cancers [50-51]. Determination of the role of mutational analysis in assessment of endometrial carcinomas will require additional study, with careful comparison of molecular versus conventional subclassification.

Supplementary Material

Supp Figure 1
Supp Figure 2
Supp Figure 3
Supp Figure 4
Supp Table 1
Supp Table 2
Supp Table Legends
Supplementary Materials and Methods

Acknowledgments

This work is supported by grants from the British Columbia (BC) Cancer Foundation and the Vancouver General Hospital (VGH)–University of British Columbia Hospital Foundation (to the OvCaRe ovarian cancer research team in Vancouver) and the Canadian Institutes of Health Research (CIHR). Work supported at Washington University in part by RO1CA71754 and P50CA134254 to PJG. We thank all the women who donated the samples used in this study. In addition, we would like to thank Jason Madore, Christine Chow and Sally Cheng and the Sequence Production and LIMS groups at Canada's Michael Smith Genome Sciences Centre for technical assistance; and the fellows of the University of British Columbia Gynaecologic Oncology Program for obtaining consent from patients for data in the OvCaRe tumour bank.

List of Abbreviations

EEC

Endometrial Endometrioid Carcinoma

EEC-3

High-grade (grade 3) Endometrial Endometrioid Carcinoma

ESC

Endometrial Serous Carcinoma

TMA

Tissue Microarray

MSI

Microsatellite Instability

TCGA

The Cancer Genome Atlas

Footnotes

Authors have no conflict of interests to report.

Statement of author contributions

MKM, SPS, MH, BG and DGH conceived and designed the study. MKM, JD, JS, WY, MH, KT, TZ, and HM carried out experiments. BG and CHL performed pathological reviews. MM, JD, MCUC, KW, JS, WY, AT, LP, APS, DGM, JNM, SPS, PJG, BG, CHL and DGH collected data and performed data analysis and assisted in interpretation of data. MCUC, MKM, JD performed statistical analysis. MKM, JD, BG, CHL and DGH wrote the manuscript and created the figures. DGH and PJG provided endometrial samples for sequencing. All authors reviewed and approved the manuscript.

References

  • 1.Evans T, Sany O, Pearmain P, et al. Differential trends in the rising incidence of endometrial cancer by type: data from a UK population-based registry from 1994 to 2006. Br J Cancer. 2011;104:1505–1510. doi: 10.1038/bjc.2011.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Bray F, Dos Santos Silva I, Moller H, et al. Endometrial cancer incidence trends in Europe: underlying determinants and prospects for prevention. Cancer Epidemiol Biomarkers Prev. 2005;14:1132–1142. doi: 10.1158/1055-9965.EPI-04-0871. [DOI] [PubMed] [Google Scholar]
  • 3.Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. 1983;15:10–17. doi: 10.1016/0090-8258(83)90111-7. [DOI] [PubMed] [Google Scholar]
  • 4.Amant F, Moerman P, Neven P, et al. Endometrial cancer. The Lancet. 2005;366:491–505. doi: 10.1016/S0140-6736(05)67063-8. [DOI] [PubMed] [Google Scholar]
  • 5.Clarke BA, Gilks CB. Endometrial carcinoma: controversies in histopathological assessment of grade and tumour cell type. Journal of clinical pathology. 2010;63:410–415. doi: 10.1136/jcp.2009.071225. [DOI] [PubMed] [Google Scholar]
  • 6.Bocker W. The WHO classification of breast tumours and tumours of the female genital organs: Pathology and genetics. Pathology of the Kidneys and Urinary Passages - Molecular Vascular Pathology and Angiogenesis. 2002;86:116–119. [PubMed] [Google Scholar]
  • 7.Prat J. Prognostic parameters of endometrial carcinoma. Human pathology. 2004;35:649–662. doi: 10.1016/j.humpath.2004.02.007. [DOI] [PubMed] [Google Scholar]
  • 8.Samarnthai N, Hall K, Yeh IT. Molecular profiling of endometrial malignancies. Obstetrics and gynecology international. 2010;2010:162363. doi: 10.1155/2010/162363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Catasus L, Gallardo A, Prat J. Molecular genetics of endometrial carcinoma. 2009;15:554–563. [Google Scholar]
  • 10.Alkushi A, Kobel M, Kalloger SE, et al. High-grade endometrial carcinoma: serous and grade 3 endometrioid carcinomas have different immunophenotypes and outcomes. International journal of gynecological pathology : official journal of the International Society of Gynecological Pathologists. 2010;29:343–350. doi: 10.1097/PGP.0b013e3181cd6552. [DOI] [PubMed] [Google Scholar]
  • 11.Hamilton CA, Cheung MK, Osann K, et al. Uterine papillary serous and clear cell carcinomas predict for poorer survival compared to grade 3 endometrioid corpus cancers. Br J Cancer. 2006;94:642–646. doi: 10.1038/sj.bjc.6603012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Soslow RA. Endometrial carcinomas with ambiguous features. Seminars in Diagnostic Pathology. 2010;27:261–273. doi: 10.1053/j.semdp.2010.09.003. [DOI] [PubMed] [Google Scholar]
  • 13.Sidhu D, Duggan M, Clement P, et al. Interobserver Agreement on High-Grade Endometrial Carcinoma and Correlation with ER, p53 and p16 Expression. Modern Pathology. 2012;25:296A–297A. [Google Scholar]
  • 14.Gilks CB, Oliva E, Soslow R. Poor Interobserver Reproducibility in the Diagnosis of High-Grade Endometrial Carcinoma. USCAP; San Antonio, TX, USA: 2011. [DOI] [PubMed] [Google Scholar]
  • 15.Oda K, Stokoe D, Taketani Y, et al. High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer research. 2005;65:10669–10673. doi: 10.1158/0008-5472.CAN-05-2620. [DOI] [PubMed] [Google Scholar]
  • 16.Konopka B, Paszko Z, Janiec-Jankowska A, et al. Assessment of the quality and frequency of mutations occurrence in PTEN gene in endometrial carcinomas and hyperplasias. Cancer Lett. 2002;178:43–51. doi: 10.1016/s0304-3835(01)00815-1. [DOI] [PubMed] [Google Scholar]
  • 17.Velasco A, Bussaglia E, Pallares J, et al. PIK3CA gene mutations in endometrial carcinoma: correlation with PTEN and K-RAS alterations. Hum Pathol. 2006;37:1465–1472. doi: 10.1016/j.humpath.2006.05.007. [DOI] [PubMed] [Google Scholar]
  • 18.Schlosshauer PW, Pirog EC, Levine RL, et al. Mutational Analysis of the CTNNB1 and APC Genes in Uterine Endometrioid Carcinoma. Modern pathology : an official journal of the United States and Canadian Academy of Pathology, Inc. 2000;13:1066–1071. doi: 10.1038/modpathol.3880196. [DOI] [PubMed] [Google Scholar]
  • 19.Okuda T, Sekizawa A, Purwosunu Y, et al. Genetics of endometrial cancers. Obstet Gynecol Int. 2010;2010:984013. doi: 10.1155/2010/984013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.O'Hara AJ, Bell DW. The genomics and genetics of endometrial cancer. Advances in Genomics and Genetics. 2012;2:33–47. doi: 10.2147/AGG.S28953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pollock PM, Gartside MG, Dejeza LC, et al. Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes. Oncogene. 2007;26:7158–7162. doi: 10.1038/sj.onc.1210529. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Goodfellow PJ, Buttin BM, Herzog TJ, et al. Prevalence of defective DNA mismatch repair and MSH6 mutation in an unselected series of endometrial cancers. Proceedings of the National Academy of Sciences. 2003;100:5908–5913. doi: 10.1073/pnas.1030231100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Urick ME, Rudd ML, Godwin AK, et al. PIK3R1 (p85{alpha}) Is Somatically Mutated at High Frequency in Primary Endometrial Cancer. Cancer research. 2011;71:4061–4067. doi: 10.1158/0008-5472.CAN-11-0549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cheung LW, Hennessy BT, Li J, et al. High Frequency of PIK3R1 and PIK3R2 Mutations in Endometrial Cancer Elucidates a Novel Mechanism for Regulation of PTEN Protein Stability. Cancer discovery. 2011;1:170–185. doi: 10.1158/2159-8290.CD-11-0039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.McConechy MK, Anglesio MS, Kalloger SE, et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. The Journal of pathology. 2011;223:567–573. doi: 10.1002/path.2848. [DOI] [PubMed] [Google Scholar]
  • 26.Shih Ie M, Panuganti PK, Kuo KT, et al. Somatic mutations of PPP2R1A in ovarian and uterine carcinomas. The American journal of pathology. 2011;178:1442–1447. doi: 10.1016/j.ajpath.2011.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Abeln ECA, Smit VTHBM, Wessels JW, et al. Molecular genetic evidence for the conversion hypothesis of the origin of malignant mixed Müllerian tumours. The Journal of pathology. 1997;183:424–431. doi: 10.1002/(SICI)1096-9896(199712)183:4<424::AID-PATH949>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 28.Voss MA, Ganesan R, Ludeman L, et al. Should grade 3 endometrioid endometrial carcinoma be considered a type 2 cancer-A clinical and pathological evaluation. Gynecologic oncology. 2011 doi: 10.1016/j.ygyno.2011.07.030. [DOI] [PubMed] [Google Scholar]
  • 29.Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in endometriosis-associated ovarian carcinomas. The New England journal of medicine. 2010;363:1532–1543. doi: 10.1056/NEJMoa1008433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Morin RD, Mendez-Lago M, Mungall AJ, et al. Frequent mutation of histone-modifying genes in non-Hodgkin lymphoma. Nature. 2011;476:298–303. doi: 10.1038/nature10351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760. doi: 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ding J, Bashashati A, Roth A, et al. Feature-based classifiers for somatic mutation detection in tumour-normal paired sequencing data. Bioinformatics. 2012;28:167–175. doi: 10.1093/bioinformatics/btr629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Forbes SA, Bindal N, Bamford S, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic acids research. 2011;39:D945–950. doi: 10.1093/nar/gkq929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Guan B, Mao TL, Panuganti PK, et al. Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. The American journal of surgical pathology. 2011;35:625–632. doi: 10.1097/PAS.0b013e318212782a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tashiro H, Blazes MS, Wu R, et al. Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer research. 1997;57:3935–3940. [PubMed] [Google Scholar]
  • 36.Sun H, Enomoto T, Fujita M, et al. Mutational analysis of the PTEN gene in endometrial carcinoma and hyperplasia. American journal of clinical pathology. 2001;115:32–38. doi: 10.1309/7JX6-B9U9-3P0R-EQNY. [DOI] [PubMed] [Google Scholar]
  • 37.Darvishian F, Hummer AJ, Thaler HT, et al. Serous endometrial cancers that mimic endometrioid adenocarcinomas: a clinicopathologic and immunohistochemical study of a group of problematic cases. The American journal of surgical pathology. 2004;28:1568–1578. doi: 10.1097/00000478-200412000-00004. [DOI] [PubMed] [Google Scholar]
  • 38.D'Angelo E, Prat J. Pathology of mixed Müllerian tumours. Best Practice &amp; Research Clinical Obstetrics &amp; Gynaecology. 2011;25:705–718. doi: 10.1016/j.bpobgyn.2011.05.010. [DOI] [PubMed] [Google Scholar]
  • 39.Soslow RA, Bissonnette JP, Wilton A, et al. Clinicopathologic analysis of 187 high-grade endometrial carcinomas of different histologic subtypes: similar outcomes belie distinctive biologic differences. Am J Surg Pathol. 2007;31:979–987. doi: 10.1097/PAS.0b013e31802ee494. [DOI] [PubMed] [Google Scholar]
  • 40.Kobel M, Kalloger SE, Baker PM, et al. Diagnosis of ovarian carcinoma cell type is highly reproducible: a transcanadian study. The American journal of surgical pathology. 2010;34:984–993. doi: 10.1097/PAS.0b013e3181e1a3bb. [DOI] [PubMed] [Google Scholar]
  • 41.Anglesio MS, Carey MS, Kobel M, et al. Clear cell carcinoma of the ovary: a report from the first Ovarian Clear Cell Symposium, June 24th, 2010. Gynecol Oncol. 2011;121:407–415. doi: 10.1016/j.ygyno.2011.01.005. [DOI] [PubMed] [Google Scholar]
  • 42.Alkushi A, Clarke BA, Akbari M, et al. Identification of prognostically relevant and reproducible subsets of endometrial adenocarcinoma based on clustering analysis of immunostaining data. Modern Pathology. 2007;20:1156–1165. doi: 10.1038/modpathol.3800950. [DOI] [PubMed] [Google Scholar]
  • 43.Zannoni G, Vellone V, Arena V, et al. Does high-grade endometrioid carcinoma (grade 3 FIGO) belong to type I or type II endometrial cancer? A clinical–pathological and immunohistochemical study. Virchows Archiv. 2010;457:27–34. doi: 10.1007/s00428-010-0939-z. [DOI] [PubMed] [Google Scholar]
  • 44.Crum CP, Lee, Kenneth R, Genest, David R. Adenocarcinoma, carcinosarcoma and other epithelial tumors of the endometrium. In: Christopher P, Crum KRL, editors. Diagnostic gynecologic and obstetric pathology. 1st ed. Saunders/Elsevier; Philadelphia, PA: 2006. p. 1182. [Google Scholar]
  • 45.Amant F, Cadron I, Fuso L, et al. Endometrial carcinosarcomas have a different prognosis and pattern of spread compared to high-risk epithelial endometrial cancer. Gynecologic oncology. 2005;98:274–280. doi: 10.1016/j.ygyno.2005.04.027. [DOI] [PubMed] [Google Scholar]
  • 46.Chiyoda T, Tsuda H, Tanaka H, et al. Expression profiles of carcinosarcoma of the uterine corpus— are these similar to carcinoma or sarcoma? Genes, Chromosomes and Cancer. 2012;51:229–239. doi: 10.1002/gcc.20947. [DOI] [PubMed] [Google Scholar]
  • 47.Murray S, Linardou H, Mountzios G, et al. Low frequency of somatic mutations in uterine sarcomas: a molecular analysis and review of the literature. Mutation research. 2010;686:68–73. doi: 10.1016/j.mrfmmm.2010.01.019. [DOI] [PubMed] [Google Scholar]
  • 48.Byron SA, Gartside M, Powell MA, et al. FGFR2 Point Mutations in 466 Endometrioid Endometrial Tumors: Relationship with MSI, KRAS, PIK3CA, CTNNB1 Mutations and Clinicopathological Features. PLoS ONE. 2012;7:e30801. doi: 10.1371/journal.pone.0030801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chapman PB, Hauschild A, Robert C, et al. Improved Survival with Vemurafenib in Melanoma with BRAF V600E Mutation. New England Journal of Medicine. 2011;364:2507–2516. doi: 10.1056/NEJMoa1103782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Prahallad A, Sun C, Huang S, et al. Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR. Nature. 2012;483:100–103. doi: 10.1038/nature10868. [DOI] [PubMed] [Google Scholar]
  • 51.Corcoran RB, Ebi H, Turke AB, et al. EGFR-Mediated Reactivation of MAPK Signaling Contributes to Insensitivity of BRAF-Mutant Colorectal Cancers to RAF Inhibition with Vemurafenib. Cancer discovery. 2012 doi: 10.1158/2159-8290.CD-11-0341. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Figure 1
Supp Figure 2
Supp Figure 3
Supp Figure 4
Supp Table 1
Supp Table 2
Supp Table Legends
Supplementary Materials and Methods

RESOURCES