Elsevier

Gynecologic Oncology

Volume 135, Issue 3, December 2014, Pages 552-559
Gynecologic Oncology

DNA methylation as a biomarker for the detection of hidden carcinoma in endometrial atypical hyperplasia

https://doi.org/10.1016/j.ygyno.2014.10.018Get rights and content

Highlights

  • AJAP1, HS3ST2 and SOX1 methylation analysis is a potential method for detection of endometrial carcinoma hidden in atypical hyperplasia.

  • Testing the methylation status of candidate genes may assist in devising an adequate treatment strategy prior to major surgery.

  • AJAP1, HS3ST2 and SOX1 expression may help to differentiate premalignant endometrium and endometrial carcinoma.

Abstract

Objective

Women with atypical hyperplasia (AH) are often found to have endometrial carcinoma (EC) at hysterectomy. The purpose of this study was to evaluate whether the hypermethylation of specific genes found by methylomic approaches to the study of gynecologic cancers is a biomarker for EC in women with AH.

Methods

We evaluated the methylation of AJAP1, HS3ST2, SOX1, and PTGDR from 61 AH patients undergoing hysterectomy. Endometrial biopsy samples were analyzed by bisulfite conversion and quantitative methylation-specific polymerase chain reaction. A methylation index was used to predict the presence of cancer. To confirm the silencing effects of DNA methylation, immunohistochemical analysis of AJAP1, HS3ST2, and SOX1 was performed using tissue microarray.

Results

Fourteen (23%) patients had EC at hysterectomy. AJAP1, HS3ST2, and SOX1 were highly methylated in the EC patients' biopsy samples (p  0.023). AJAP1, HS3ST2, and SOX1 protein expression was significantly higher in patients with AH only (p  0.038). The predictive value of AJAP1, HS3ST2, and SOX1 methylation for EC was 0.81, 0.72, and 0.70, respectively. Combined testing of both AJAP1 and HS3ST2 methylation had a positive predictive value of 56%, methylation of any one of AJAP1, SOX1, or HS3ST2 had a 100% negative predictive value.

Conclusions

Hypermethylation of AJAP1, HS3ST2, and SOX1 is predictive of EC in AH patients. Testing for methylation of these genes in endometrial biopsy samples may be a hysterectomy-sparing diagnostic tool. Validation of these new genes as biomarkers for AH screening in a larger population-based study is warranted.

Introduction

Endometrial carcinoma (EC) is one of the most common cancers of the female genital tract, although the incidence varies between countries [1]. Prolonged exposure to estrogen promotes the development of endometrial hyperplasia (EH), which leads to atypical hyperplasia (AH); 25–40% of patients with AH subsequently progress to EC. Although AH is the least common type of hyperplasia, it is the type most likely to progress to type 1 EC, which accounts for more than 80% of uterine cancers [2]. EC is usually confined to the inner lining of the uterus, which can be removed by hysterectomy. Unfortunately, even in stage I EC, there is a significant risk of tumor recurrence, distant metastasis, and death [3].

EH is classified into two categories by the World Health Organization: 1) EH, including simple and complex hyperplasia without atypia, and 2) endometrial AH, including simple and complex hyperplasia with atypia [4]. Several studies have shown that cytological atypia, which is the major criterion for the diagnosis of AH and the most reliable indicator of progression from EH to EC, has poor reproducibility [2], [5], [6]. In 12.7–42.6% of cases, EC coexists in patients with a diagnosis of AH [7]. The high rate of unrecognized cancer among women diagnosed preoperatively with AH reflects the fact that the histologic criteria for differentiating AH from some types of EC on dilation and curettage (D&C) are controversial and subject to different interpretations [8], [9], [10].

Because of overlap in the histologic picture of AH and low-grade EC in the limited tissue sample evaluated before major surgery, differentiation on pathologic grounds alone can be very difficult or impossible [5], [10], [11]. Although D&C before hysterectomy is the gold standard method for the diagnosis of endometrial lesions, detection of AH cannot rule out a more severe lesion [12], [13]. EC found at the time of hysterectomy for AH may be associated with deep myometrial (10%) or cervical stroma (5%) invasion [14].

Hysterectomy is the main therapeutic modality for AH. Conservative approaches such as high-dose progestin may be acceptable treatment options in certain situations (e.g., to maintain fertility), but the risks of progression to malignancy and of concurrent EC remain high [14]. At present, there is no established biomarker to differentiate endometrial AH and EC. Such a marker could be hysterectomy sparing for AH patients without EC. Even when EC arising from endometrial premalignant lesions is clearly defined, the possibilities for EC screening are very limited. Reliable determination of the presence or absence of EC would allow for better surgical decisions about hysterectomy and staging. The reassurance of patients given fertility-sparing management for AH may alleviate unnecessary anxiety. There is, therefore, a need to develop new, molecular-based, complementary tools that could improve the pathological diagnosis.

Epigenetic studies have demonstrated that silencing of genes, such as tumor-suppressor genes (TSGs), can act as a mechanism of carcinogenesis [15], [16]. The addition of a methyl group to the cytosine–guanine (CpG) island results in gene silencing. Because epigenetic silencing of TSGs by promoter hypermethylation is observed commonly in human cancers, it is possible that DNA methylation could be used for the early diagnosis of cancer. This concept, and its application in gynecologic cancers, has been gaining acceptance during the past few years, especially in diagnosing and treating cervical cancer (screening and triage) and ovarian cancer (prognosis) [17], [18], [19], [20]. However, similar studies of EC are relatively limited. It is known that the progression of EC involves a multistep process, and both genetic and epigenetic events have been shown to play important roles. Although gene promoter CpG islands epigenetically marked by de novo DNA methylation may serve as biomarkers in EC, they have been rarely studied in AH [21], [22], [23], [24]. Such epigenetic biomarkers could be useful for identifying EC in AH.

Our previous research on the epigenomics of cervical cancer using methylomic approaches identified several candidate genes that are methylated in cervical cancer tissues. Several candidate genes were significantly hypermethylated in CIN3 + lesions [19], [25]. Because the uterine cervix and endometrium both originate from the Müllerian duct system, this close embryologic relationship between the uterine cervix and endometrium may be reflected in adulthood in the form of malignant lesions. We hypothesized that some of the genes hypermethylated in cervical cancer may also be hypermethylated in EC. We initially tested 28 development-related genes. To test further the feasibility of using these new biomarkers in identifying endometrial lesions, we converted the methylation analysis to a quantitative methylation-specific polymerase chain reaction (QMSP) approach and tested its application value. We found that the following several genes were potentially implicated in endometrial carcinogenesis: adherens junction-associated protein 1 (AJAP1), heparan sulfate d-glucosamyl 3-O-sulfotransferase-2 (HS3ST2), sex-determining region Y, box 1 (SOX1), prostanoid receptor gene, prostaglandin D2 receptor (PTGDR), and LIM-homeobox gene 1A (LMX1A). These candidate genes, which could be used for the triage of AH, were validated in The Cancer Genome Atlas (TCGA) EC database. The aim of the present study was to analyze the DNA methylation status of AJAP1, HS3ST2, SOX1, PTGDR, and LMX1A genes as biomarkers for EC diagnosis in patients with endometrial AH. The discovery of reliable epigenetic biomarkers for diagnosis may open a new avenue for the management of AH patients with and without EC.

Section snippets

Patients and clinical samples

Samples of endometrium from patients with endometrioid-type EC (n = 20; 8 G1 cases, 8 G2 cases, 4 G3 cases) and with dysfunctional uterine bleeding (n = 20) were included as cancer and normal controls, respectively. Specimens were obtained from tissue blocks for methylation analysis of the candidate genes. Endometrial biopsy tissues of patients with AH (n = 61) were collected for methylation analysis. All patients underwent hysterectomy within 3 months after endometrial sampling. The clinicopathologic

DNA methylation of AJAP1, PTGDR, HS3ST2, SOX1, and LMX1A in EC

We first examined the methylation status of the five genes AJAP1, PTGDR, HS3ST2, SOX1, and LMX1A from EC patients and compared it with that in samples from normal endometrium. We randomly chose 20 samples of EC and 20 normal controls for analysis. The methylation status of AJAP1, PTGDR, HS3ST2, and SOX1 in the EC and normal controls is shown in Fig. 1. The EC samples showed a higher methylation index for AJAP1, PTGDR, HS3ST2, and SOX1 compared with normal controls (p < 0.0001, p = 0.0002, p = 0.0004,

Discussion

Endometrial carcinogenesis is a multistep process involving a precursor lesion with underlying genetic and epigenetic events. Therefore, molecular diagnostic methods have been proposed as new ancillary tools for the detection of undetected cancers and differential diagnosis of premalignant and malignant lesions. Abnormal patterns of DNA methylation have been recognized in various cancers. An increase in DNA methylation in gene promoter regions often precedes apparent malignant changes, which

Conflict of interest statement

The authors declare that no conflict of interest exists.

Acknowledgments

This study was supported by grants from the Tri-Service General Hospital (TSGH-C101-073, TSGH-C102-074, TSGH-C103-076, TSGH-C103-006-008-S01, TSGH-C103-006-008-S02, and TSGH-C103-006-008S03), NSC-102-2628-B-038-010-MY3, NSC-102-2314-B-016-038 and NSC-102-2314-B-016-043 from the National Science Council, Taiwan, Republic of China, TMUTOP103005-1 and 103TMU-SHH-11 from Taipei Medical University, MOHW103-TDU-B-212-113001 and MOHW103-TDU-PB-211-122025 from Ministry of Health and Welfare.

References (40)

  • G.L. Mutter

    Diagnosis of premalignant endometrial disease

    J Clin Pathol

    (2002)
  • Y.S. Fu et al.

    Pathophysiology and management of endometrial hyperplasia and carcinoma

    West J Med

    (1990)
  • P. Litta et al.

    Atypical endometrial lesions: hysteroscopic resection as an alternative to hysterectomy

    Eur J Gynaecol Oncol

    (2013)
  • K.H. Allison et al.

    Diagnosing endometrial hyperplasia: why is it so difficult to agree?

    Am J Surg Pathol

    (2008)
  • B.S. Kendall et al.

    Reproducibility of the diagnosis of endometrial hyperplasia, atypical hyperplasia, and well-differentiated carcinoma

    Am J Surg Pathol

    (1998)
  • G.L. Mutter

    Histopathology of genetically defined endometrial precancers

    Int J Gynecol Pathol

    (2000)
  • E. Suh-Burgmann et al.

    Complex atypical endometrial hyperplasia: the risk of unrecognized adenocarcinoma and value of preoperative dilation and curettage

    Obstet Gynecol

    (2009)
  • E. Rakha et al.

    Clinical outcome of atypical endometrial hyperplasia diagnosed on an endometrial biopsy: institutional experience and review of literature

    Am J Surg Pathol

    (2012)
  • S. Agrawal et al.

    DNA methylation of tumor suppressor genes in clinical remission predicts the relapse risk in acute myeloid leukemia

    Cancer Res

    (2007)
  • G. Wu et al.

    LOXL1 and LOXL4 are epigenetically silenced and can inhibit ras/extracellular signal-regulated kinase signaling pathway in human bladder cancer

    Cancer Res

    (2007)
  • Cited by (26)

    • Elevated miR-20b-5p expression in peripheral blood mononuclear cells: A novel, independent molecular biomarker of favorable prognosis in chronic lymphocytic leukemia

      2018, Leukemia Research
      Citation Excerpt :

      However, the bootstrapping that was performed in Cox regression strengthens the hypothesis that miR-20b-5p overexpression is a promising molecular biomarker of favorable prognosis in CLL. Therefore, a panel of molecular prognostic biomarkers that have recently emerged − e.g. the human leukocyte antigen (HLA-G) [44], high histone deacetylase (HDAC) activity [45], BCL2-like 12 (BCL2L12) [46], circulating miR-150 [47], and/or SRY-box 1 (SOX1) [48] − could prove even more promising, provided that it integrates miR-20b-5p expression as a new component. In support of this suggestion, it should be added that accumulation of several unfavorable prognostic markers has been proven to deteriorate prognosis in CLL [49].

    • mRNA overexpression of the hypoxia inducible factor 1 alpha subunit gene (HIF1A): An independent predictor of poor overall survival in chronic lymphocytic leukemia

      2017, Leukemia Research
      Citation Excerpt :

      Still, the bootstrap Cox regression analysis performed in the current study strengthens the hypothesis that HIF1A mRNA overexpression is a promising molecular biomarker of unfavorable prognosis in CLL. Taking into consideration that besides HIF1A mRNA several other molecular biomarkers have been suggested during the last decade − including the human leukocyte antigen (HLA-G) [50], high histone deacetylase (HDAC) activity [51], mRNA expression of kallikrein-related peptidase 14 (KLK14) [52] and BCL2-like 12 (BCL2L12) [53], circulating miR-150 [54], as well as the aforementioned SOX1 [47] and soluble TACI [48] – along with the previously drawn conclusion that accumulation of several unfavorable prognostic markers has been proven to deteriorate prognosis in CLL [55], it makes sense to examine the combinatorial value of all these previously suggested putative prognostic biomarkers plus HIF1A mRNA expression in a large multicenter study, in attempt to generate a prognostic score in CLL, similarly to the one that has recently been suggested by the German Chronic Lymphocytic Leukemia Study Group [56]. In conclusion, this is the very first time that the prognostic value of HIF1A mRNA expression is being investigated in CLL patients.

    • Methylation of tumor suppressor gene CDH13 and SHP1 promoters and their epigenetic regulation by the UHRF1/PRMT5 complex in endometrial carcinoma

      2016, Gynecologic Oncology
      Citation Excerpt :

      Thus, early diagnosis or prevention of endometrial carcinoma is the key to reduce incidence of this now deadly cancer and improve survival rate. Molecularly, accumulating evidence suggests that epigenetic changes are the major event in transformation of normal cells to cancers and promoter hypermethylation of tumor suppressor genes is an epigenetic hallmark of human cancer [5–7]. Methylation of normally unmethylated genes, particularly tumor suppressors, has been shown to result in gene silencing and ultimately lead to cancer development [8].

    • Quantitative DNA methylation analysis of selected genes in endometrial carcinogenesis

      2015, Taiwanese Journal of Obstetrics and Gynecology
      Citation Excerpt :

      Abnormal patterns of DNA methylation have been recognized in various cancers. An increase in DNA methylation in gene promoter regions often precedes apparent malignant changes, which suggests that the assessment of DNA methylation could be used for the early diagnosis of cancer [8,12,22]. Our results demonstrate the usefulness of assessing DNA methylation for detecting EC in endometrial hyperplasia.

    • Detection of endometrial cancer via molecular analysis of DNA collected with vaginal tampons

      2015, Gynecologic Oncology
      Citation Excerpt :

      In this study, we validated methylation markers previously identified as hypermethylated in EC compared with BE [18,21–23,29,34,40], and demonstrated detection of these markers in the downstream effluent of the female reproductive tract. One of these markers, HS3ST2, was identified in our original discovery efforts [34] and has recently been validated as an EC biomarker in a cohort of women with endometrial biopsies showing atypical hyperplasia [48]. In addition, we identified a novel gene methylated in EC, HOXA9, which is also highly methylated in the vaginal pool of women with EC.

    View all citing articles on Scopus
    1

    HC Lai and YC Wang contributed equally to this work.

    View full text