Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Mod Pathol. Author manuscript; available in PMC 2014 Feb 6.
Published in final edited form as:
PMCID: PMC3915240
NIHMSID: NIHMS536970
PMID: 23765252

Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles

Associated Data

Supplementary Materials

Abstract

Ovarian endometrioid carcinomas and endometrial endometrioid carcinomas share many histological and molecular alterations. These similarities are likely due to a common endometrial epithelial precursor cell of origin, with most ovarian endometrioid carcinomas arising from endometriosis. To directly compare the mutation profiles of two morphologically similar tumor types, we performed select exon capture sequencing on a panel of genes: ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, TP53. We found that PTEN mutations are more frequent in low-grade endometrial endometrioid carcinomas (67.0%) compared to low-grade ovarian endometrioid carcinomas (16.6%) (p<0.0001). In contrast, CTNNB1 mutations are significantly different in low-grade ovarian endometrioid carcinomas (53.3%) compared to low-grade endometrial endometrioid carcinomas (27.5%) (p<0.0057). This difference in CTNNB1 mutation frequency may be reflective of the distinct microenvironments; the epithelial cells lining an endometriotic cyst within the ovary are exposed to a highly oxidative environment that promotes tumorigenesis. Understanding the distinct mutation patterns found in the PI3K and Wnt pathways of ovarian and endometrial endometrioid carcinomas may provide future opportunities for stratifying patients for targeted therapeutics.

Keywords: Endometrial, ovarian, endometrioid, carcinoma, PTEN, CTNNB1, mutations

Introduction

Ovarian and endometrial carcinomas comprise the majority of gynecological carcinomas in developed countries 1. Endometrial carcinoma is the most common with an estimated 287,000 new cases worldwide, however ovarian carcinoma is the most lethal gynecological cancer 2. Both ovarian and endometrial carcinomas can be subclassified into the histological types; endometrioid, serous, clear cell, mixed, carcinosarcomas and mucinous carcinomas. Endometrial endometrioid carcinomas (EECs) occur most frequently, accounting for 70–80% of cases; these tend to be low-grade with good prognosis and are generally cured by hysterectomy 3, 4. The remaining 20–30% of endometrial carcinomas are serous, mixed, clear cell carcinomas, and carcinosarcomas. These subtypes are high-risk tumors that tend to have poor outcomes due to metastasis and ineffective treatment options 5. Endometrial carcinomas have been shown to possess distinct mutations specific to each subtype; EECs are characterized by mutations in PTEN, ARID1A, PIK3CA, PIK3R1, CTNNB1 and KRAS (reviewed in 6).

Ovarian endometrioid carcinomas (OECs), account for only 10% of ovarian carcinomas. The majority of ovarian carcinomas (70–80%) are high-grade serous carcinomas, which similar to endometrial serous carcinomas are aggressive with poor outcomes. The majority of OECs, like EECs, are also low-grade with good prognosis 7. Endometrioid carcinomas of the ovary and endometrium are both described to contain PTEN, PIK3CA, ARID1A, PPP2R1A, and CTNNB1 (β-catenin) mutations 8, 9, however the frequency of mutations differs in these tumor types.

There is accumulating evidence that ovarian endometrioid carcinomas arise from transformed endometriosis 10. Therefore, endometrial and ovarian endometrioid carcinomas evolve from similar precursor endometrial epithelial cells, however the process of molecular pathogenesis is still unclear. The molecular features of OECs and EECs have been characterized in many studies using immunohistochemistry markers and mutational analysis by DNA sequencing (reviewed in 4, 6, 8, 11). Recently, the ability to sequence multiple genes in the same cases using next-generation sequencing technology has allowed the investigation of the mutational landscapes of these tumor types. There have been studies that infer differences in mutation frequencies in OECs and EECs 12, however to the best of our knowledge, no prior studies have directly compared endometrial and ovarian endometrioid mutation frequencies with a uniform technical approach in a large cohort of tumors. We recently published a study using select exon capture coupled with next-generation sequencing to determine the mutation profiles in the various subtypes of endometrial carcinoma 13. We have now also performed the same exon capture sequencing method and applied this to a cohort of ovarian endometrioid carcinomas. In this study, we have directly compared the mutation frequencies of seven specific genes in low-grade endometrial endometrioid carcinomas and low-grade ovarian endometrioid carcinomas.

Materials and Methods

Patient Samples

We obtained frozen tumor tissue from 129 endometrial endometrioid carcinomas and 33 ovarian endometrioid carcinomas, for exon capture sequencing, originating from the OvCaRe Tissue Biobank repository, Vancouver, BC, Canada. Patients provided informed consent, and research ethics was approved, and DNA extracted as previously described 9. An additional 178 endometrial endometrioid carcinoma tumor DNA samples, with grade information available, were obtained from Washington University, St. Louis, Missouri. All samples from both centers have undergone histological review by gynecological pathologists. All subtype and mutational data for all endometrial carcinomas have been previously reported 13. An additional 20 ovarian endometrioid carcinoma cases (18 frozen tumors, 2 Formalin-Fixed Paraffin Embedded (FFPE)) were obtained from the OvCaRe Tissue Biobank repository to test CTNNB1 mutation status, so that a total of 53 OECs were tested for mutation in this gene only, and 33 OEC cases were tested for the other genes in the panel.

Mutation analysis

Genomic DNA (500ng) was used for indexed Illumina library construction, then underwent targeted enrichment using biotinylated RNA capture probes generated from cDNA clones or PCR amplicons representing exons of ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, TP53, BRAF and PPP2R5C and sequenced using the Illumina (GAIIx). All sequencing validation methods and primers used are previously described 13. The two genes BRAF and PPP2R5C were excluded from subsequent analysis due to only one mutation found in BRAF and no mutations in PPP2R5C in the ovarian endometrioid carcinoma cases. To validate the differences in CTNNB1 mutation frequencies, we re-sequenced the hotspot exon 3 of CTNNB1 by Sanger sequencing from all 178 low-grade endometrial endometrioid DNA obtained from Washington University. Additional Sanger sequencing validations for the hotspot exon 3 of CTNNB1 were also performed on 20 low-grade ovarian endometrioid carcinoma cases that were not included in the original select exon capture sequencing set.

Bioinformatics Analysis

Short reads were aligned to the human genome (hg18) using the BWA aligner v0.5.9 14. A Random Forest classifier trained on validated SNVs was used to remove false-positive calls 15. SNVs in the Catalogue of Somatic Mutations in Cancer (COSMIC) 16 were considered to be true positives. All analysis was performed as previously described 13.

Statistical Analysis

Fisher exact tests 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. The Benjamini-Hochberg17 (B-H) method was used to adjust p-values to account for multiple comparisons (R stats package).

Results

To determine the differences in somatic mutation frequencies between endometrial and ovarian endometrioid carcinomas, we used select gene exon capture sequencing of ARID1A, PTEN, PIK3CA, KRAS, CTNNB1, PPP2R1A, and TP53. This was performed using 33 cases of ovarian endometrioid and 307 cases of endometrial endometrioid carcinomas (Supplementary Table 1). Comparison of the mutational frequencies of low-grade (grade 1 and 2) endometrial endometrioid carcinomas to low-grade (grade 1 and 2) ovarian endometrioid carcinomas showed a significant difference for PTEN (adjusted p<0.0007) and CTNNB1 (adjusted p=0.02) mutations (Table 1, Figure 1). PTEN mutations were found in 67.0% of low-grade EECs, while 16.6% of low-grade OECs harbor PTEN mutations. Mutations of CTNNB1 were identified in 53.3% of OECs, and only 27.5% of EECs. This frequency of CTNNB1 mutations in EECs is slightly higher than previously published 13 due to additional validations of mutations using Sanger sequencing and select exon capture sequencing. To further verify the high CTNNB1 mutation frequency found in OECs, we acquired an additional 20 low-grade OEC cases and tested for hotspot CTNNB1 mutations using direct Sanger sequencing. We found that 45% (9 of 20) of these cases contained hotspot CTNNB1 mutations, bringing the overall frequency to 50% (25 of 50) CTNNB1 mutations in low-grade OECs (Supplementary Table 1). The mutation frequencies of PIK3CA, ARID1A, PPP2R1A, and TP53 are not significantly different between low-grade EECs and OECs. There is a trend towards more KRAS mutations in low-grade OEC’s (33.3%) compared to low-grade EECs (18.1%), however this is not significant after multiple comparison adjustments (Table 1).

An external file that holds a picture, illustration, etc.
Object name is nihms536970f1.jpg
Low-grade ovarian and endometrial endometrioid mutation profiles

A. Low-grade endometrial endometrioid carcinomas (EEC) including grade 1 and 2 (n=276). B. Low-grade ovarian endometrioid carcinomas (OEC) including grade 1 and 2 (n=30). Individual columns designate one tumor case, and rows indicate genes. All colored boxes specify a genetic alteration such as missense, truncating, indels, splice site mutations and combinations of these mutations. Grey boxes indicate no mutation were identified by sequencing. These colors are specifically shown in the color legend.

Table 1

Comparison of mutation frequencies in low-grade ovarian endometrioid carcinomas and low-grade endometrial endometrioid carcinomas.

Low-Grade
Ovarian
Endometrioid
(Grade 1 and 2)
(n=30)
Low-Grade
Endometrial
Endometrioid
(Grade 1 and 2)
(n=276)
Fisher Exact
Test (p-value)
Adjusted
p-value*
PTEN5 (16.6%)185 (67.0%)1.08e-070.001
PIK3CA12 (40.0%)107 (38.7%)+11
ARID1A9 (30.0%)129 (46.7%)0.0860.120
KRAS10 (33.3%)50 (18.1%)+0.0550.120
CTNNB116 (53.3%)76 (27.5%)+0.0060.020
PPP2R1A5 (16.6%)19 (6.9%)0.0710.120
TP532 (6.6%)28 (10.1%)11
*p-values are adjusted using the B-H method
+additional mutations have been verified by Sanger sequencing post original publication.

The comparisons of high-grade (grade 3) endometrial endometrioid carcinomas with high-grade ovarian endometrioid carcinomas are limited by the rarity of high-grade OECs (n=3). Each high-grade OEC case harbors different mutations in different genes. Similarly to low-grade EECs, high-grade EECs also have a high frequency of PTEN mutations (87.1%), and a lower frequency of CTNNB1 mutations (19.4%) (Supplementary Table 2).

Most CTNNB1 mutations found in both OEC and EEC involve known phospho-acceptor sites. In OECs, 16 of 25 (64%) contain CTNNB1 mutations that affect serine (S33 and S37) or threonine (T41) amino acid residues, which are phosphorylation targets for glycogen synthase kinase 3-beta (GSK3β). Similarly in low-grade EECs, 43 of 76 (56.6%) of the CTNNB1 mutations are located at serine (S33, S37, S45) and threonine (T41) residues (Supplementary Table 1). Our analysis of all CTNNB1 coding sequences also revealed additional somatic mutations outside the hotspot serine/threonine residues.

Discussion

The distinct molecular abnormalities of endometrial and ovarian carcinoma subtypes will be the basis for subtype specific treatment, and may become an essential component of stratified management strategies. Standard treatment options for ovarian and endometrial carcinomas have not yet changed, but a shift towards subtype specific clinical trials highlights the need to better understand the molecular abnormalities and potential therapeutic targets in the different subtypes 18. The same subtypes at different sites in the gynecological tract, EECs and OECs, have indistinguishable morphology, clinical similarities, and share at least some genetic abnormalities. We have directly compared the mutation frequencies in the same gene set, using the same technology, in low-grade ovarian and endometrial endometrioid carcinomas. This has shown that there are similar mutations patterns but there are also two distinct differences.

Previous literature often refers to endometrial and ovarian endometrioid carcinomas as having similar molecular alterations 19, 20. The same genes are often mutated, however some studies suggest that OECs and EECs have similar frequencies of CTNNB1 12 and PTEN mutations 21, but do not directly compare the mutation frequencies of the two tumor types in the same study. In this study, PTEN mutations are found at higher frequencies in low-grade EEC compared to low-grade OECs, and CTNNB1 mutations at higher frequencies in OECs compared to EECs. One limitation of this study, is the small number of low-grade OECs (n=30) analyzed compared to the number of low-grade EECs (n=276), in part a reflection on the relative rarity of OECs. Previous studies have reported low-grade OECs with CTNNB1 mutation frequencies between 16–54% 2228, with an average frequency of 40–50%. This range of CTNNB1 mutation frequency is due to varying methods of detection and limited exon sequencing. PTEN mutations are reported in only about 20% of OECs 29, 30. All these studies also assessed small cohorts of OECs, where the mutation frequencies for both CTNNB1 and PTEN are similar to the frequencies identified in this study.

Recently, our group 13 and others 3133 have published mutation frequencies in a large number of EECs. Byron et al. report a 19% CTNNB1 mutation frequency from 466 EEC tumors 32. In this study we report a slightly higher CTNNB1 mutation frequency of 27.5% from 276 low-grade EECs. This difference is most probably due to sequencing and analysis methods used, as well as the fact that we screened all exons of all genes for mutations. The current study included 25 EEC tumor samples also analyzed by Byron et al.; there were 66 mutations in CTNNB1 and 46 in KRAS reported in the original paper13, whereas we have now identified 76 CTNNB1 and 50 KRAS mutations, all validated by Sanger sequencing, indicating a low false negative rate in the earlier study. This does not change the conclusions of that study13 but does indicate that there was a slight underestimation of mutation frequencies. As previously shown PTEN mutations are very common in both low-grade (67.0%) and high-grade EECs (87.1%) 13, and is the most frequently mutated gene in this tumor type. In a separate study of 243 EECs, 44% were found to contain PTEN mutations, and 16% CTNNB1 mutations 31, which does confirm the frequency of CTNNB1 mutations, however the PTEN mutation rate is lower than our observations and those reported by others. For example, Urick et al. showed that 78% of EEC’s have PTEN mutations, but did not sequence CTNNB1 33. In conclusion, our study confirms observations from other studies suggesting there are differences in PTEN and CTNNB1 mutation rates in ovarian endometrioid carcinomas and endometrial endometrioid carcinomas. Similarly, ovarian and endometrial serous carcinoma subtypes are morphologically equivalent and were often thought to have similar mutation patterns, with both showing a high frequency of TP53 mutations. However, more detailed sequencing analyses of these tumor types have revealed mutational profile differences. Mutations of PPP2R1A are found at high frequencies in endometrial serous carcinomas but are rarely found in ovarian high-grade serous carcinomas 9, 34. More recent studies have identified high frequencies of other gene mutations (ie. FBXW7, CHD4) 35, 36, in endometrial serous carcinomas but not in ovarian serous carcinomas 37.

The majority of ovarian endometrioid carcinomas are believed to arise from endometriosis 8, 38. Although ovarian and endometrial endometrioid carcinomas may develop from the same cell type, namely the endometrial epithelial cell, these two tumor types are exposed to different microenvironments that may reflect differences in their mutation spectrums. EECs frequently occur in postmenopausal women with unopposed estrogen 39; exposure to high estrogen and low progesterone levels have been found to increase proliferation of endometrial cells and thus increases the risk of tumorigenesis 40. Chromosomal aberrations have been identified at a high frequency in ovarian endometriotic cysts compared to extra-gonadal endometriosis 41. Endometriosis is thought to occur via retrograde menstruation, where endometrial epithelial cells travel from the uterus through the fallopian tubes and can establish as an endometriotic cyst within the ovary 42. This creates a unique microenvironment where menstruation-like blood and necrotic tissue is trapped within the cyst, resulting in high concentrations of iron in a confined space 43. This iron causes oxidative stress, and a hypoxic environment that can in turn lead to DNA damage and mutation accumulation 44, 45. The mutational analysis of a small number of endometriosis lesions has mostly been confined to a subset of genes, PTEN, CTNNB1 and KRAS with only a small number of somatic mutations found in PTEN 29, and KRAS 46. It will be important to determine the malignant transformation pathways of endometriosis to OECs, and in so doing, identify the genetic differences between the precursors of endometrial and ovarian endometrioid carcinomas.

Based on the mutation frequencies found in this study, CTNNB1 mutations in the ovary and PTEN mutations in the endometrium are characteristic features of these diseases. Mutations in CTNNB1 and deregulation of the Wnt pathway are a well-established pathway in cancer signaling first characterized in colorectal cancers 47, 48. As seen in our study, the majority of CTNNB1 mutations affect the GSK3β binding sites for β-catenin, which ultimately influences the ability of GSK3β to phosphorylate β-catenin and signal its degradation. Lack of β-catenin phosphorylation results in an accumulation of protein, which then translocates to the nucleus and interacts with the transcription factors TCF/LEF1 (T-cell factor/lymphoid enhancer factor) to induce gene expression of the cell proliferation genes MYC and cyclin D1, as well as the inflammatory gene COX2 (reviewed in 49). β-catenin is also known to play a role in cell adhesion by cytoplasmic interaction with the tumor suppressor E-cadherin at the cell membrane. Ovarian carcinomas with an accumulation of β-catenin in the nucleus, either due to CTNNB1 mutations or deregulation of other Wnt family member like APC or Axin, is an indicator of good prognosis 23. Similar to the Wnt pathway, PI3K signaling is also a highly studied cancer pathway, and is one of the most commonly altered pathways 50. PTEN acts as a negative regulator of the PI3K pathway by dephosphorylating the signaling lipid molecule PIP3 to PIP2; where PIK3CA (p110α) together with PIK3R1 (p85α) acts to phosphorylate PIP2 to PIP3 allowing PI3K signaling to proceed through AKT and mTOR, and thus activation of multiple cell proliferation and cell survival proteins. Mutations in both PTEN and PIK3CA can act to maintain constitutively activated PI3K signaling (reviewed in 51). These pathways do not occur linearly, and are interactive within their own signal transduction networks as well as with other pathways, which is shown by convergence on GSK3β in both the PI3K and Wnt pathways (Figure 2). Constitutive activation of AKT by PI3K signaling, by PTEN and PI3K mutations, leads to the phosphorylation of GSK3β targeting it for degradation, and thus allowing free β-catenin to translocate to the nucleus 52. Similarly, CTNNB1 mutations can lead to elevated β-catenin activity by abolishing GSK3β’s ability to negatively regulate β-catenin, which accumulates in the nucleus. There are however many other gene regulation events that can result through the activation of PI3K/AKT/mTOR pathway that are not activated through the Wnt pathway. Therefore, in OECs, it may be beneficial to target both the Wnt pathway, to inhibit β-catenin and the PI3K pathway when PI3KCA is mutated. In EECs, PTEN and PIK3CA are both frequently mutated, likely leading to the up-regulation of PI3K signaling, thus targeting the PI3K pathway may be of benefit. Additionally, there is no mutual exclusivity of CTNNB1 and PTEN or PIK3CA mutations in EECs and OECs (Figure 1), indicating that they are not functionally equivalent, therefore when both pathways are mutated; simultaneously targeting the PI3K and Wnt pathways may be appropriate. Careful consideration will be needed when deciding which molecules to target in one or multiple pathways, as well as the specific cellular context.

An external file that holds a picture, illustration, etc.
Object name is nihms536970f2.jpg
PI3K/AKT and WNT signaling pathways

Theses two signaling pathways show convergence on GSK3β and β-catenin. Genetic alterations caused by mutations in both pathways can result in the transcription of cell growth and proliferation genes. Mutations are indicated by black stars and are found in both ovarian and endometrial endometrioid tumors.

Ovarian and endometrial endometrioid carcinomas share obvious histogenic connections and are morphologically similar, however there are genomic differences, as shown by CTNNB1 and PTEN mutation frequencies. The occurrence of these mutations may reflect different environmental niches during oncogenesis, and ultimately point toward different routes of distinct targeted therapeutics.

Supplementary Material

Supp Table 1

Supp Table 2

Acknowledgments

We would like to thank all the women who generously donated the samples used in this study. This work is supported by grants from the British Columbia (BC) Cancer Foundation, the Vancouver General Hospital (VGH)–University of British Columbia Hospital Foundation (to the OvCaRe ovarian cancer research team, Vancouver), and the Canadian Institutes of Health Research (CIHR). Work supported at Washington University in part by RO1CA71754 and P50CA134254 to PJG. We would also like to thank 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 tumor bank.

Footnotes

Disclosure/Conflict of Interest

The authors have no conflict of interest to disclose.

References

1. Jemal A, Bray F, Center MM, et al. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. [PubMed] [Google Scholar]
2. Ferlay J, Shin HR, Bray F, et al. International Agency for Research on Cancer. Lyon, France: IARC CancerBase No. 10; 2010. GLOBOCAN 2008 v1.2, Cancer Incidence and Mortality Worldwide. http://globocan.iarc.fr. [Google Scholar]
3. Catasus L, Gallardo A, Prat J. Molecular genetics of endometrial carcinoma. Diagnostic Histopathology. 2009;15:554–563. [Google Scholar]
4. Samarnthai N, Hall K, Yeh IT. Molecular profiling of endometrial malignancies. Obstet Gynecol Int. 2010;2010:162363. [PMC free article] [PubMed] [Google Scholar]
5. Prat J. Prognostic parameters of endometrial carcinoma. Hum Pathol. 2004;35:649–662. [PubMed] [Google Scholar]
6. O'Hara AJ, Bell DW. The genomics and genetics of endometrial cancer. Advances in Genomics and Genetics. 2012;2:33–47. [PMC free article] [PubMed] [Google Scholar]
7. Chen S, Leitao MM, Tornos C, Soslow RA. Invasion patterns in stage I endometrioid and mucinous ovarian carcinomas: a clinicopathologic analysis emphasizing favorable outcomes in carcinomas without destructive stromal invasion and the occasional malignant course of carcinomas with limited destructive stromal invasion. Mod Pathol. 2005;18:903–911. [PubMed] [Google Scholar]
8. Kurman RJ, Shih Ie M. Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer--shifting the paradigm. Hum Pathol. 2011;42:918–931. [PMC free article] [PubMed] [Google Scholar]
9. McConechy MK, Anglesio MS, Kalloger SE, et al. Subtype-specific mutation of PPP2R1A in endometrial and ovarian carcinomas. J Pathol. 2011;223:567–573. [PubMed] [Google Scholar]
10. Prowse AH, Manek S, Varma R, et al. Molecular genetic evidence that endometriosis is a precursor of ovarian cancer. International Journal of Cancer. 2006;119:556–562. [PubMed] [Google Scholar]
11. Bell DA. Origins and molecular pathology of ovarian cancer. Mod Pathol. 2005;18(Suppl 2):S19–S32. [PubMed] [Google Scholar]
12. Catasus L, Bussaglia E, Rodrguez I, et al. Molecular genetic alterations in endometrioid carcinomas of the ovary: similar frequency of beta-catenin abnormalities but lower rate of microsatellite instability and PTEN alterations than in uterine endometrioid carcinomas. Hum Pathol. 2004;35:1360–1368. [PubMed] [Google Scholar]
13. McConechy MK, Ding J, Cheang MC, et al. Use of mutation profiles to refine the classification of endometrial carcinomas. J Pathol. 2012;228:20–30. [PMC free article] [PubMed] [Google Scholar]
14. Li H, Durbin R. Fast and accurate short read alignment with Burrows- Wheeler transform. Bioinformatics. 2009;25:1754–1760. [PMC free article] [PubMed] [Google Scholar]
15. 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. [PMC free article] [PubMed] [Google Scholar]
16. Forbes SA, Bindal N, Bamford S, et al. COSMIC: mining complete cancer genomes in the Catalogue of Somatic Mutations in Cancer. Nucleic Acids Res. 2011;39:D945–D950. [PMC free article] [PubMed] [Google Scholar]
17. Benjamini Y, Hochberg Y. Controlling the False Discovery Rate - a Practical and Powerful Approach to Multiple Testing. J Roy Stat Soc B Met. 1995;57:289–300. [Google Scholar]
18. Stuart GCE, Kitchener H, Bacon M, et al. 2010 Gynecologic Cancer InterGroup (GCIG) Consensus Statement on Clinical Trials in Ovarian Cancer Report From the Fourth Ovarian Cancer Consensus Conference. Int J Gynecol Cancer. 2011;21:750–755. [PubMed] [Google Scholar]
19. Geyer JT, Lopez-Garcia MA, Sanchez-Estevez C, et al. Pathogenetic pathways in ovarian endometrioid adenocarcinoma: a molecular study of 29 cases. Am J Surg Pathol. 2009;33:1157–1163. [PubMed] [Google Scholar]
20. McCluggage WG. Morphological subtypes of ovarian carcinoma: a review with emphasis on new developments and pathogenesis. Pathology. 2011;43:420–432. [PubMed] [Google Scholar]
21. Shedden KA, Kshirsagar MP, Schwartz DR, et al. Histologic type, organ of origin, and Wnt pathway status: effect on gene expression in ovarian and uterine carcinomas. Clin Cancer Res. 2005;11:2123–2131. [PubMed] [Google Scholar]
22. Palacios J, Gamallo C. Mutations in the beta-catenin gene (CTNNB1) in endometrioid ovarian carcinomas. Cancer Res. 1998;58:1344–1347. [PubMed] [Google Scholar]
23. Gamallo C, Palacios J, Moreno G, et al. beta-catenin expression pattern in stage I and II ovarian carcinomas : relationship with beta-catenin gene mutations, clinicopathological features, and clinical outcome. Am J Pathol. 1999;155:527–536. [PMC free article] [PubMed] [Google Scholar]
24. Wu R, Zhai Y, Fearon ER, Cho KR. Diverse mechanisms of beta-catenin deregulation in ovarian endometrioid adenocarcinomas. Cancer Res. 2001;61:8247–8255. [PubMed] [Google Scholar]
25. Wright K, Wilson P, Morland S, et al. beta-catenin mutation and expression analysis in ovarian cancer: exon 3 mutations and nuclear translocation in 16% of endometrioid tumours. Int J Cancer. 1999;82:625–629. [PubMed] [Google Scholar]
26. Saegusa M, Okayasu I. Frequent nuclear beta-catenin accumulation and associated mutations in endometrioid-type endometrial and ovarian carcinomas with squamous differentiation. J Pathol. 2001;194:59–67. [PubMed] [Google Scholar]
27. Sagae S, Kobayashi K, Nishioka Y, et al. Mutational analysis of beta-catenin gene in Japanese ovarian carcinomas: frequent mutations in endometrioid carcinomas. Jpn J Cancer Res. 1999;90:510–515. [PMC free article] [PubMed] [Google Scholar]
28. Moreno-Bueno G, Gamallo C, Perez-Gallego L, et al. beta-Catenin expression pattern, beta-catenin gene mutations, and microsatellite instability in endometrioid ovarian carcinomas and synchronous endometrial carcinomas. Diagn Mol Pathol. 2001;10:116–122. [PubMed] [Google Scholar]
29. Sato N, Tsunoda H, Nishida M, et al. Loss of Heterozygosity on 10q23.3 and Mutation of the Tumor Suppressor Gene PTEN in Benign Endometrial Cyst of the Ovary: Possible Sequence Progression from Benign Endometrial Cyst to Endometrioid Carcinoma and Clear Cell Carcinoma of the Ovary. Cancer Res. 2000;60:7052–7056. [PubMed] [Google Scholar]
30. Obata K, Morland SJ, Watson RH, et al. Frequent PTEN/MMAC Mutations in Endometrioid but not Serous or Mucinous Epithelial Ovarian Tumors. Cancer Res. 1998;58:2095–2097. [PubMed] [Google Scholar]
31. 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 Discov. 2011;1:170–185. [PMC free article] [PubMed] [Google Scholar]
32. 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. [PMC free article] [PubMed] [Google Scholar]
33. Urick ME, Rudd ML, Godwin AK, et al. PIK3R1 (p85{alpha}) Is Somatically Mutated at High Frequency in Primary Endometrial Cancer. Cancer Res. 2011;71:4061–4067. [PMC free article] [PubMed] [Google Scholar]
34. Shih Ie M, Panuganti PK, Kuo KT, et al. Somatic mutations of PPP2R1A in ovarian and uterine carcinomas. Am J Pathol. 2011;178:1442–1447. [PMC free article] [PubMed] [Google Scholar]
35. Kuhn E, Wu RC, Guan B, et al. Identification of Molecular Pathway Aberrations in Uterine Serous Carcinoma by Genome-wide Analyses. J Natl Cancer Inst. 2012 [PMC free article] [PubMed] [Google Scholar]
36. Gallo ML, O'Hara AJ, Rudd ML, et al. Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat Genet. 2012 [PMC free article] [PubMed] [Google Scholar]
37. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615. [PMC free article] [PubMed] [Google Scholar]
38. Jiang X, Morland SJ, Hitchcock A, Thomas EJ, Campbell IG. Allelotyping of endometriosis with adjacent ovarian carcinoma reveals evidence of a common lineage. Cancer Res. 1998;5:1707–1712. [PubMed] [Google Scholar]
39. Siiteri PK, Schwarz BE, MacDonald PC. Estrogen receptors and the estrone hypothesis in relation to endometrial and breast cancer. Gynecol Oncol. 1974;2:228–238. [PubMed] [Google Scholar]
40. Key TJA, Pike MC. The Dose-Effect Relationship between Unopposed Estrogens and Endometrial Mitotic Rate - Its Central Role in Explaining and Predicting Endometrial Cancer Risk. Brit J Cancer. 1988;57(2):205–212. [PMC free article] [PubMed] [Google Scholar]
41. Korner M, Burckhardt E, Mazzucchelli L. Higher frequency of chromosomal aberrations in ovarian endometriosis compared to extragonadal endometriosis: A possible link to endometrioid adenocarcinoma. Mod Pathol. 2006;19:1615–1623. [PubMed] [Google Scholar]
42. Giudice LC. Endometriosis. New England Journal of Medicine. 2010;362:2389–2398. [PMC free article] [PubMed] [Google Scholar]
43. Yamaguchi K, Mandai M, Toyokuni S, et al. Contents of endometriotic cysts, especially the high concentration of free iron, are a possible cause of carcinogenesis in the cysts through the iron-induced persistent oxidative stress. Clinical Cancer Research. 2008;14:32–40. [PubMed] [Google Scholar]
44. Van Langendonckt A, Casanas-Roux F, Dolmans MM, Donnez J. Potential involvement of hemoglobin and heme in the pathogenesis of peritoneal endometriosis. Fertil Steril. 2002;77:561–570. [PubMed] [Google Scholar]
45. Kobayashi H, Kajiwara H, Kanayama S, et al. Molecular pathogenesis of endometriosis-associated clear cell carcinoma of the ovary (Review) Oncol Rep. 2009;22:233–240. [PubMed] [Google Scholar]
46. Vestergaard AL, Thorup K, Knudsen UB, et al. Oncogenic events associated with endometrial and ovarian cancers are rare in endometriosis. Molecular Human Reproduction. 2011;17:758–761. [PubMed] [Google Scholar]
47. Su LK, Vogelstein B, Kinzler KW. Association of the APC tumor suppressor protein with catenins. Science. 1993;262:1734–1737. [PubMed] [Google Scholar]
48. Rubinfeld B, Souza B, Albert I, et al. Association of the APC gene product with beta-catenin. Science. 1993;262:1731–1734. [PubMed] [Google Scholar]
49. Klaus A, Birchmeier W. Wnt signalling and its impact on development and cancer. Nat Rev Cancer. 2008;8:387–398. [PubMed] [Google Scholar]
50. Samuels Y, Wang Z, Bardelli A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science. 2004;304:554. [PubMed] [Google Scholar]
51. Cully M, You H, Levine AJ, Mak TW. Beyond PTEN mutations: the PI3K pathway as an integrator of multiple inputs during tumorigenesis. Nat Rev Cancer. 2006;6:184–192. [PubMed] [Google Scholar]
52. McCubrey JA, Steelman LS, Chappell WH, et al. Mutations and Deregulation of Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR Cascades. Oncotarget. 2012;3:954–987. [PMC free article] [PubMed] [Google Scholar]
-