bs_bs_banner

doi:10.1111/jog.12442

J. Obstet. Gynaecol. Res. Vol. 40, No. 8: 1957–1967, August 2014

Carcinogenic mechanisms of endometrial cancer: Involvement of genetics and epigenetics Kouji Banno, Megumi Yanokura, Miho Iida, Kenta Masuda and Daisuke Aoki Department of Obstetrics and Gynecology, School of Medicine, Keio University, Tokyo, Japan

Abstract Endometrial cancer is increasing worldwide and the number of patients with this disease is likely to continue to grow, including younger patients. Many endometrial cancers show estrogen-dependent proliferation, but the carcinogenic mechanisms are unknown or not completely explained beyond mutations of single oncogenes and tumor suppressor genes. Possible carcinogenic mechanisms include imbalance between endometrial proliferation by unopposed estrogen and the mismatch repair (MMR) system; hypermethylation of the MMR gene hMLH1; mutation of PTEN, β-catenin and K-ras genes in type I endometrial cancer and of HER-2/neu and p53 genes in type II endometrial cancer; hypermethylation of SPRY2, RASSF1A, RSK4, CHFR and CDH1; and methylation of tumor suppressor microRNAs, including miR-124, miR-126, miR-137, miR-491, miR-129-2 and miR-152. Thus, it is likely that the carcinogenic mechanisms of endometrial cancer involve both genetic and epigenetic changes. Mutations and methylation of MMR genes induce various oncogenic changes that cause carcinogenesis, and both MMR mutation in germ cells and methylation patterns may be inherited over generations and cause familial tumorigenesis. Determination of the detailed carcinogenic mechanisms will be useful for prevention and diagnosis of endometrial cancer, risk assessment, and development of new treatment strategies targeting MMR genes. Key words: DNA hypermethylation, DNA mismatch repair, endometrial cancer, epigenetics, genetics.

Introduction A total of 288 000 patients were newly diagnosed with endometrial cancer worldwide in 2008.1 More than 4000 women died from endometrial cancer in the USA in 2011.2 In Japan, the annual morbidity increased from 48 in the 20–30 years in 1975 to 478 in 2005.3 The annual mortality per 100 000 population in Japan increased from 0.4 in 1975 to 3.2 in 2012 and the total morbidity increased from 229 to 2092.4 The incidence of endometrial cancer is likely to continue to increase based on these recent trends. Discovering the causes of the increase and establishment of prophylactic measures and new therapeutic strategies requires an improved understanding of the carcinogenic mechanisms of endometrial cancer.

Environmental factors, including estrogen, an abnormal mismatch repair (MMR) system, genetic abnormalities, and aberrant methylation of DNA and microRNA, are currently proposed as major mechanisms of carcinogenesis in endometrial cancer. Endometrial cancer is defined as type I or II based on clinicopathological properties. Type I endometrial cancer more commonly develops in premenopausal or perimenopausal women and occurs in an estrogendependent manner via atypical endometrial hyperplasia. The tumor is positive for the estrogen receptor and progesterone receptor, shows well-differentiated endometrioid adenocarcinoma, has a lower frequency of lymph node metastasis, shows little muscular invasion, and often has a relatively favorable prognosis. In contrast, type II endometrial cancer tends to develop in

Received: February 18 2014. Accepted: March 3 2014. Reprint request to: Dr Kouji Banno, Department of Obstetrics and Gynecology, School of Medicine, Keio University, Shinanomachi 35 Shinjuku-ku, Tokyo 160-8582, Japan. Email: [email protected]

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1957

K. Banno et al.

postmenopausal women in an estrogen-independent manner, and is thought to be due to de novo carcinogenesis that develops directly from the normal endometrium, rather than via endometrial hyperplasia or undiagnosed precancerous lesions. The tissue type is specific, including extremely poorly differentiated endometrioid adenocarcinoma and serous adenocarcinoma, and the prognosis is often poor. This review focuses on the mechanisms of carcinogenesis in endometrial cancer that have recently emerged.

Carcinogenic Mechanisms Involving Estrogen Estrogen is a steroid hormone that promotes the development of female genitalia, including the endometrium, vagina, vulva and mammary gland. Estrogen passes through the cell membrane and binds to estrogen receptor (ER) in the cytoplasm. ER forms dimers and regulates gene expression via estrogen response elements in promoter regions of target genes. ER has ligand- and DNA-binding domains, and ligandindependent activation function (AF)-1 and liganddependent AF-2 transcriptional activation domains.5 The balance of transcriptional activation domains varies among tissues, with dominance of AF-2 in mammary gland cells and AF-1 in endometrial cells.6,7 Miyamoto et al.8 suggested that mismatch repair (MMR) deficiency was the most important abnormality in early-stage endometrial cancer, and examined the correlation between MMR and estrogen. Expression of hMLH1 and hMSH2, which are important MMR proteins, was examined by immunostaining and showed a strong positive correlation with blood estrogen. MMR activity in endometrial epithelial cells in vitro also showed a dose-dependent increase with higher estrogen levels. This suggests that cancer is unlikely to occur in a high estrogen environment because increased cell growth is paralleled by increased MMR activity. In contrast, hMLH1 and hMSH2 were absent or had extremely low expression at estrogen levels ranging from 20 to 60 pg/mL, but some cell growth still occurred. Therefore, cells dividing in a low-estrogen environment are more likely to accumulate genetic errors due to low repair activity and may be at high risk for carcinogenesis. Based on these results, Miyamoto et al.8 suggested that the incidence of growth-induced genetic errors should be low in young women with high estrogen levels and sufficient repair activity of MMR proteins, making carcinogenesis unlikely. In

1958

Low estrogen

Relatively low

High estrogen

(E2 < 15 pg/mL)

estrogen

(E2 > 80 pg/mL)

(E2 20–60pg/mL) CP = MMR activity

CP > MMR activity

CP < MMR activity

Non-carcinogenic

Carcinogenic

Non-carcinogenic

Cancer window

Figure 1 Hypothesis of endometrial carcinogenesis based on the estrogen environment and mismatch repair (MMR) activity. CP, cell proliferation.

older women with lower estrogen but an atrophic endometrium, carcinogenesis would also be unlikely because of the absence of cell growth. However, under perimenopausal conditions, the carcinogenic risk would be increased because sufficient estrogen is present to promote cell division, but MMR activity is low. This intermediate status was defined as the cancer window (Fig. 1).

Carcinogenic Mechanisms of Abnormal Mismatch Repair System The mismatch repair (MMR) system is responsible for repairing base mismatches that arise during DNA replication. Typical MMR proteins include hMLH1, hMSH2, hPMS2, hMSH3 and hMSH6. Genes encoding these proteins are called MMR genes and aberrations in these genes prevent correct repair of mismatched bases, resulting in DNA strands with different lengths. This phenomenon occurs in microsatellite regions of the human genome and is referred to as microsatellite instability (MSI). Microsatellites or short tandem repeats (STR) are repeating sequences of one to five base pairs of DNA, such as CA and CAG. Some STR occur in regions encoding phosphatase and tensin homolog deleted on chromosome ten (PTEN), a lipid phosphatase that is a tumor suppressor gene; TGF-βR2 and IGF2R, which are associated with inhibition of cell proliferation; K-ras, which is involved in cell proliferation; and BAX, which is related to apoptosis induction. Therefore, MSI is implicated in carcinogenesis.9 Aberrations in MMR genes are involved in carcinogenesis of type I endometrial cancer. These aberrations are caused by epigenetic changes independent of the DNA sequence, that is, gene inactivation by aberrant

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

Carcinogenesis of endometrial cancer

hypermethylation of promoter regions. Such inactivation of MMR genes permits accumulation of gene mutations and leads to carcinogenesis. In endometrial cancer, carcinogenesis most frequently involves aberrant methylation of hMLH1 and mutation of hMLH1 is detected in 30% of cases. Mutations of hMLH1 are also found in atypical endometrial hyperplasia, which suggests that hMLH1 is implicated in the early stage of carcinogenesis.10,11 Muraki et al.12 reported aberrant hMLH1 hypermethylation in 40.4% of patients with endometrial cancer and found significantly reduced hMLH1 protein levels in these patients (P < 0.01). However, none of the four cancer-related genes were aberrantly methylated in the normal endometrium. MMR genes are also causative genes in Lynch syndrome (hereditary nonpolyposis colorectal cancer). Lynch syndrome is a typical familial tumor with autosomal dominant inheritance. Female patients with Lynch syndrome complicate with endometrial cancer at a high incidence. In the revised 1999 Amsterdam II Criteria (AC II), endometrial cancer was included as a cancer with similar features to colon, small intestine, ureteral and kidney cancers.13 The prevalence of Lynch syndrome is 0.9–2.7%14 and approximately 2.3% of cases of endometrial cancer occur due to Lynch syndrome.15 The lifetime risk of endometrial cancer is 28–60% in women with aberrant genes associated with Lynch syndrome.16 hMLH1, hMSH2 and hMSH6 mutations are particularly important in families of patients with Lynch syndrome. Most mutations occur in hMLH1 and hMSH2, whereas hMHS6 mutations are important in tumorigenesis in patients with endometrial cancer.17,18 Kawaguchi et al.19 proposed a possible new cascade in which hMSH6 mutation is induced by

silencing of hMLH1 due to aberrant DNA hypermethylation in endometrial cancer. Westin et al.20 showed that the incidence of Lynch syndrome was 1.8% in endometrial cancer in total and 9% in endometrial cancer in women aged less than 50 years old, but 29% in cases with lower uterine segment cancer (LUS) and germ cell mutation of hMSH2. These results suggest a correlation between endometrial cancer of the uterine isthmus and Lynch syndrome. Masuda et al.21 also found germ cell mutations of hMLH1 in 1.4% of patients with LUS. Based on these findings, Lynch syndrome may be clinically predictive of the onset site of endometrial cancer.

Carcinogenic Mechanisms of Gene Mutation Several gene mutations have emerged as candidates for roles in carcinogenesis of type I and II endometrial cancer (Fig. 2), based on observation of the mutation in endometrial hyperplasia and at least a similar incidence of mutation in endometrial cancer. Different genes are involved in carcinogenesis of the two types of endometrial cancer. Gene mutations found in type I endometrial cancer include those in PTEN, β-catenin and K-ras. PTEN is a tumor suppressor gene on chromosome 10 and has been identified as a disease gene in three autosomal dominant disorders (Cowden disease, LhermitteDuclos disease and Bannayan-Zonana syndrome). PTEN inactivation is also found in malignant melanoma, brain tumors, and endometrial, ovarian, thyroid, breast and prostate cancers. PTEN protein induces

PTEN mutation hMLH1 methylation hMSH6 mutation

PTEN mutation K-ras mutation b-catenin mutation

p53 mutation

Type I Normal endometrium

Type II

Figure 2 Gene mutations in the carcinogenesis of endometrial cancer.

Normal endometrium Atrophic endometrium

Atypical endometrial hyperplasia

Endometrioid cancer (low-grade)

Endometrioid cancer (high-grade)

p53 mutation HER-2/neu mutation Non-endometrioid cancer

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1959

K. Banno et al.

apoptosis and carcinogenesis occurs in cells with PTEN mutation due to avoidance of apoptosis. PTEN mutations have been detected in 20–33% of cases of atypical endometrial hyperplasia and 33–50% of cases of endometrial cancer;22–24 thus, PTEN appears to be involved in the early stage of carcinogenesis, which is a pattern that differs from that in late-onset cancer, including rectal cancer. Furthermore, in endometrial cancer, PTEN mutations are frequently detected in cases with aberrant methylation in promoter regions of hMLH1 that cause inactivation of the MMR gene; therefore, PTEN is thought to be a target of MMR genes.25 β-catenin (CTNNB1) mutations are found in 20–40% of cases of type I endometrial cancer.26–28 β-catenin is a component of E-cadherin, which has an important role in cell adhesion and is involved in the Wnt signaling pathway that regulates cell proliferation and differentiation. β-catenin degradation is prevented by mutations and the transcription levels of target genes of β-catenin increase. These mutations are also detected in atypical endometrial hyperplasia; therefore, β-catenin mutations are implicated in the early stage of carcinogenesis.29 The K-ras oncogene encodes a protein of 21 kDa that has a signaling function from activated membrane receptors in the MAPK pathway. If mutations occur, K-ras continuously functions as activated Ras and excessive signaling causes cell proliferation and induces carcinogenesis.30 K-ras mutations have been detected in 6–16% of cases of endometrial hyperplasia31 and 10–31% cases of endometrial cancer.32,33 Tsuda et al.34 showed that the incidence of K-ras mutation was significantly higher in tumors with invasive proliferation (P < 0.002) and that the incidence of mutation in well-differentiated (Grade 1) tumors was significantly higher than that in moderately (Grade 2) and poorly differentiated (Grade 3) tumors (P < 0.025). These results suggest that K-ras is involved in two stages of carcinogenesis: a shift from endometrial hyperplasia to endometrial cancer and invasive proliferation of well-differentiated tumor cells. Lagarda et al.35 found that the incidence of K-ras mutation was significantly higher in MSI-positive endometrial cancer and was related to aberrant methylation of MMR genes. Mutations in type II endometrial cancer are thought to be linked to the oncogene HER-2/neu and tumor suppressor gene p53. HER-2/neu is a tyrosine kinase membrane receptor in the epidermal growth factor (EGF) receptor family. Mutations of this gene are also found in breast and ovarian cancers. HER-2/neu expression in endometrial cancer has a strong inverse corre-

1960

lation with differentiation.36 However, the incidence of gene amplification differs from 14% to 63% in all cancers37–40 and overexpression of the protein ranges from 9% to 74%.41,42 A p53 gene mutation is the most frequent mutation in human cancer. Normal p53 regulates cell proliferation, apoptosis induction and DNA repair. Point mutations in p53 are found in 90% of cases of type II endometrial cancer, but in only 10–20% of cases of Grade 3 type I endometrial cancer. The incidence is low in endometrial hyperplasia and type I endometrial cancer of other grades.43,44 Feng et al.45 showed that p53 gene mutations occurred only at sites with positive p53 protein expression in endometrioid adenocarcinoma, which were poorly differentiated regions of cancer tissues. p53 is also implicated in the early stage of carcinogenesis of serous adenocarcinoma. Zheng et al.46,47 found strong positive p53 immunostaining in normal endometria and suggested that this ‘p53 signature’ reflected potential serous adenocarcinoma lesions. The p53 signature is found frequently in normal endometria adjacent to serous adenocarcinoma, but rarely detected in other normal endometria or tissues adjacent to endometrioid adenocarcinoma. Based on these findings, Zheng et al. proposed a carcinogenic model in which genetic changes occur prior to morphological changes. Atypical epithelium develops features similar to serous adenocarcinoma and covers endometrial cortical layers, but is not invasive. This state is referred to as serous endometrial intraepithelial carcinoma (EIC) and finally progresses to serous adenocarcinoma. RB and cyclin may also be involved in carcinogenesis of endometrial cancer. RB was the first gene to be identified as a disease gene in retinoblastoma in children. Non-phosphorylated RB protein inhibits cell proliferation in the G0 and early G1 phases. After phosphorylation by the complex of cyclin and cyclin-dependent kinase (CDK), pRB releases the transcription factor E2F, which then increases DNA polymerase activity and promotes cell proliferation. RB gene mutations have been found in small cell lung, bladder and esophageal cancers. In endometrial cancer, loss of heterozygosity (LOH) was found in 18% of RB genes and pRB downregulation was consistent with LOH.48 The incidence of mutations increased with advancement of the clinical stage.49 Cyclin is a protein that controls the cell cycle in cooperation with CDK and is overexpressed in endometrial cancer. Shih et al.50 showed that expression of cyclin A was an independent poor prognostic factor. Overexpression of cyclin D1 is induced by mutations in sites with ubiquitin degradation in the same

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

Carcinogenesis of endometrial cancer

gene.51 Cables, an inhibitor of CDK2 that negatively regulates cell proliferation, was recently indicated to be involved in onset of endometrial cancer through a relation with cyclin. Endometrial hyperplasia and well-differentiated endometrial cancer occur in Cablesknockout mice and Cables is downregulated in human endometrial cancer, regardless of the tissue type, which implicates Cables mutation in the onset of endometrial cancer.52,53

Carcinogenic Mechanisms of Epigenetic Changes Epigenetic regulation of gene expression includes effects of DNA methylation, histone modification and Polycomb group proteins.54 DNA methylation is imprinted at the time of cell division and has been widely studied in mammals. Genomic DNA methylation in vertebrates is based on addition of a methyl group to a cytosine base at a CpG sequence by DNA methyltransferase. This includes methylation of a CpG in a new DNA strand to maintain the methylation pattern found in the template DNA strand, and de novo methylation of a CpG that was not previously methylated. Maintenance methylation permits inheritance of DNA methylation patterns, while de novo methylation creates new methylation patterns in cell development and differentiation, aging and tumorigenesis processes. DNA methylation in CpG islands, which are regions dense with CpG sites, in promoters upstream of gene transcription start sites is critical in gene expression.55 If the DNA in this region is not methylated, a nucleosome does not form and transcription occurs, while methylation of the same DNA allows nucleosome formation and blocks transcription.56,57 Many tumor suppressor genes in cancer cells are inactivated by aberrant DNA methylation in promoter CpG islands, which suggests that aberrant DNA methylation may cause carcinogenesis similarly to gene mutations.58 MMR gene methylation is particularly important and, as described above, Muraki et al.12 detected aberrant methylation of hMLH1 in 40.4% of patients with endometrial cancer. Inactivation of MMR genes that repair mismatches induces MSI in many tumor suppressor genes, including PTEN, TGF-βR2, IGF2R and BAX, and contributes to carcinogenesis. For example, TGF-βR2 encodes receptors of TGF-β, a cytokine that inhibits epithelial cell proliferation. Sakaguchi et al.59 showed downregulation of TGF-βR2 in endometrial cancer and suggested that the major

cause was hMLH1 methylation and that TGF-βR2 was a target gene of MMR genes. PTEN and K-ras mutations are found in cases with aberrant methylation of the hMLH1 promoter region and MSI-positive cases, suggesting that PTEN and K-ras are also MMR target genes.25,35 In addition to hMLH1, genes inactivated by DNA methylation in endometrial cancer include SPRY2 (Sprouty2), Ras association domain family 1 isoform A (RASSF1A), ribosomal 56 kinase4 (RSK4), adenomatous polyposis coil (APC), checkpoint with FHA and RING (CHFR), p73, caspase-8 (CASP8), G-protein coupled receptor 54 (GPR54), cadherin 1 (CDH1), homeobox A11 (HOXA11) and catechol-O-methyltransferase (COMT).12,60–67 SPRY2 is an antagonist of the fibroblast growth factor (FGF) receptor, and inhibits cell proliferation and differentiation and angiogenesis by inhibiting the RAS-MAPK pathway downstream of the FGF receptor. Velasco et al.60 found that SPRY2 expression depended on the menstrual cycle in normal endometria and proposed involvement of SPRY2 in development of glandular structures. SPRY2 expression is extremely low in highly invasive cancer other than endometrioid adenocarcinoma.60 RASSF1A is also a tumor suppressor gene that negatively regulates the RAS-MAPK pathway. Pallarés et al.61 found aberrant hypermethylation of RASSF1A promoters and downregulation of RASSF1A in advanced endometrial cancer associated with MSI. RSK4 is a tumor suppressor gene in the FGFR2/RAS/ERK pathways that inhibits cell proliferation. Dewdney et al.62 showed that RSK4 expression was downregulated by methylation in atypical endometrial cancer (and particularly in high-grade endometrial cancer), as well as in rectal, breast and kidney cancers. APC is also a tumor suppressor gene and APC protein induces degradation of β-catenin, a Wnt-signaling factor. Aberrant APC methylation is found in endometrial hyperplasia and early endometrial cancer. Ignatov et al.68 showed that the incidence of APC methylation decreased with progression of endometrial cancer, which suggests that aberrant APC methylation may be an important marker of early carcinogenesis of endometrial cancer. CHFR is an M phase checkpoint gene that regulates progression of the cell cycle. Satoh et al.69 and Wang et al.70 showed that CHFR downregulation by aberrant hypermethylation increases the paclitaxel sensitivity of gastric and endometrial cancers. These findings suggest that examination of CHFR expression could form the basis of personalized cancer treatment. p73 is a homolog of the tumor suppressor gene p53 that

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1961

K. Banno et al.

regulates DNA repair, cell growth arrest and apoptosis, similar to p53. CASP8 is an apoptosis-related gene involved in cell death via Fas ligands.71 Both p73 and CASP8 have been found to be methylated in endometrial cancer.63 GPR54 is a gene encoding endogenous receptors of kisspeptin (KISS1), a cancer metastasis suppressor. Kang et al.64 found significantly higher survival in patients with endometrial cancer with high GPR54 expression (P < 0.05) and showed that the expression was epigenetically regulated by methylation. Yi et al.65 showed that CDH1, a promoter of E-cadherin involved in cell adhesion, was methylated in endometrial cancer, and the consequent downregulation of E-cadherin had effects on both cancer progression in clinical pathology and 5-year survival rates. These findings suggest that aberrant methylation of GPR54 and CDH1 promotes invasion and metastasis of cancer cells and worsens the prognosis of endometrial cancer. HOXA11 is involved in proliferation and differentiation of the endometrium. Whitcomb et al.66 showed that methylation of the HOXA11 promoter was more frequent in recurrent endometrial cancer than in primary cases. COMT is an enzyme that degrades catechol estrogen. Sasaki et al.67 found that methylation of the COMT promoter selectively inactivated membrane-bound COMT and was implicated in carcinogenic mechanisms of endometrial cancer via estrogen. Overall organization of gene methylation can be described using the concept of the CpG island methylator phenotype (CIMP). CpG island methylation in colon cancer is found genome-wide or in specific regions. Toyota et al.72 proposed classification of the cancer type based on CIMP. Thus, cancer with genomewide methylation is classified as CIMP-positive because of breakdown of regulation of methylation. Weisenberger et al.73 suggested that CIMP could be a new tumor marker. In endometrial cancer, Zhang et al.74 examined the methylation status of five genes (p14, p16, ER, COX-2 and RASSF1A) and found CIMPpositive cancer tissues and adjacent normal endometrial tissues. These findings suggest that CIMP could be a marker for early carcinogenesis in endometrial cancer.

Epimutation and Inheritance Epimutation refers to epigenetic changes in germ cells that inhibit transcription of genes for which expression

1962

is not usually inhibited or activate genes that are usually inhibited.75,76 Typical epigenetic changes include DNA methylation and histone acetylation. Epimutation may be the first stage or a direct cause of carcinogenesis. Development of endometrial cancer may involve epimutation of MMR genes, including hMLH1 and hMSH2. Kondo et al.77 showed that epigenetic silencing of hMLH1 was more frequent than that of hMSH2. hMLH1 mutation is particularly found in multiple primary neoplasms, including Lynch syndrome; however, epimutation may exist in germ cells without mutation of hMLH1 itself.78 Hitchins et al.79 suggested that epigenetic errors can be resolved by demethylation during oogenesis, but that small amounts of methylation remain; consequently, epimutation is maternally inherited. However, the frequency of inheritance is lower than that of variants in germ cell lines. Goel et al.80 described a case of hMLH1 epimutation in the paternal allele, which suggests that new epimutation can also occur after fertilization and is inherited. In 2006, Chan et al.81 showed hMSH2 epimutation in germ cell lines of a family including three parents who developed colon or endometrial cancer in young adulthood. hMSH2 mutation did not exist, but protein deficiency was found and MSI was shown to be associated with epimutation of maternally inherited hMSH2. Inheritance of the same epimutation from three parents to three children suggested that not only DNA sequence mutation but also epimutation may be inherited through multiple generations. Also, epimutation did not exist in all cells and methylation levels differed among tissues. This mosaic status of methylation may be the first hit of the two-hit theory of onset and suggests new genetic mechanisms that do not comply with Mendel’s laws. Methylation levels were the highest in the rectal mucosa and colon cancer tissues, and the lowest in leukocytes, leading to the suggestion that epimutation may be overlooked in common gene mutation assays using leukocytes.81 The EPCAM gene encodes epithelial cell adhesion molecules and is overexpressed in most cancers. There are various opinions on the role of EPCAM in carcinogenesis. EPCAM is a homophilic intracellular adhesion molecule that may promote metastasis of cancer cells by inhibiting intracellular adhesion due to E-cadherin.82 Ligtenberg et al.83 showed that epigenetic mutation in the 3′-upstream region of EPCAM inactivated hMSH2 and was involved in carcinogenesis of endometrial cancer.

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

Carcinogenesis of endometrial cancer

Aberrant DNA hypermethylation

Estrogen

hMLH1 hMSH6 hMSH2

Reduction of TS-miRNA expression miR-152, miR-129-2

EPCAM

Mismatch repair deficiency

APC RASSF1A RSK4 p53 p73 GPR54 CHFR CDH1 CASP8 Inactivation of tumor suppressor genes

Microsatellite instability

DNMT1 E2F3 MET Rictor

SOX4

Activation of oncogenes

PTEN, TGF-βR2, IGF2R, K-ras, BAX ?

Mutation of tumor-associated genes

Normal endometrium

Atypical endometrial hyperplasia

Endometrial cancer (well-differentiated)

?

?

Endometrial cancer (poorly differentiated)

Figure 3 Carcinogenic mechanisms caused by aberrant DNA hypermethylation in type I endometrial cancer.

Carcinogenic Mechanisms Involving micro-RNA microRNAs (miRNAs) are short noncoding RNAs of 18–25 base pairs that regulate gene expression. miRNAs that inhibit DNA methylation in cancers are referred to as tumor suppressor miRNAs (TSmiRNA), and include miR-124, miR-126, miR-137 and miR-491.84–88 Huang et al.89 showed that miR-129-2 functions as a TS-miRNA through negative regulation of SRY-related high-mobility group box 4 (SOX4), an oncogene that is overexpressed in endometrial cancer. Methylation of miR-129-2 was found in 68% of 117 patients with endometrial cancer with elevated SOX4 expression. Methylation of miR-129-2 is also related to MSI and hypermethylated hMLH1. Therefore, oncogene activation may be caused by methylation of a miRNA that has an inhibitory action on oncogene

expression, in addition to direct promoter demethylation. Tsuruta et al.90 similarly showed that expression of miR-152 is reduced by aberrant DNA methylation and can be recovered by the demethylating action of 5-aza-dC. Screening of methylation and expression showed that miR-152 is also a TS-miRNA in endometrial cancer. miR-152 methylation levels are also changed in acute lymphoblastic leukemia, gastrointestinal cancer and cholangiocarcinoma.91–93 DNA methyltransferase 1 (DNMT1) is a well-known target of miR-152; and E2F3, MET and Rictor have been identified as new targets. miR-152 inhibits expression of all of these genes. E2F3 is an E2F family transcriptional inhibitor and may be an oncogene;94 MET is a cell surface receptor for hepatocyte growth factor and a known oncogene;95 and Rictor is part of the mTOR complex 2 (mTORC2) and is important for cancer cell proliferation.96,97

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1963

K. Banno et al.

Conclusion In this review, we summarized new findings on the carcinogenic mechanisms of endometrial cancer. Carcinogenesis cannot be completely explained by endometrial proliferation due to estrogen and a single gene mutation. However, the core carcinogenic mechanisms of type I endometrial cancer are DNA methylation (an epigenetic change) and subsequent breakdown of the MMR system (Fig. 3). These actions cause oncogene mutation, inactivation of tumor suppressor genes, and oncogene activation via TS-miRNA silencing, and contribute to chaotic cell proliferation, that is, carcinogenesis. Methylation patterns of MMR genes may be inherited over generations and may cause familial tumorigenesis, including Lynch syndrome, while estrogen may control both cell proliferation and MMR activity. However, the carcinogenic mechanisms remain largely unknown, particularly with regard to de novo carcinogenesis of type II endometrial cancer. Improved diagnosis, risk assessment, and new treatment strategies targeting MMR genes will require establishment of the details of these mechanisms in endometrial cancer.

4.

5.

6.

7.

8.

9.

10.

Acknowledgments The authors gratefully acknowledge grant support from the Japan Society for the Promotion of Science (JSPS) through a Grant-in-Aid for Scientific Research (KAKENHI), a Grant-in-Aid for Scientific Research (C) (22591866), and a Grant-in-Aid for Young Scientists (B) (24791718); the Medical Research Encouragement Prize of The Japan Medical Association; and the Keio Gijyuku Academic Development Fund.

Disclosure

11.

12.

13.

14.

None disclosed. 15.

References 1. World Cancer Research Fund International. Cancer facts and figures: Endometrial cancer rates. 2013. [Cited on 16 Nov 2013.] Available from URL: http://www.wcrf.org/cancer _statistics/data_specific_cancers/endometrial_cancer _statistics.php 2. Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 2011; 61: 212– 236. 3. Matsuda T, Marugame T, Kamo K, Katanoda K, Ajiki W, Sobue T, Japan Cancer Surveillance Research Group. Cancer

1964

16.

17.

18.

incidence and incidence rates in Japan in 2006: based on data from 15 population-based cancer registries in the monitoring of cancer incidence in Japan (MCIJ) project. Jpn J Clin Oncol 2012; 42: 139–147. Center for Cancer Control and Information Services. Vital Statistics Japan (Ministry of Health, Labour and Welfare). 2013. [Cited on 16 Nov 2013.] Available from URL: http:// ganjoho.jp/professional/statistics/statistics.html Metzger D, Ali S, Bornert JM, Chambon P. Characterization of the amino-terminal transcriptional activation function of the human estrogen receptor in animal and yeast cells. J Biol Chem 1995; 270: 9535–9542. McLnerney EM, Katzenellenbogen BS. Different regions in activation function-1 of the human estrogen receptor required for antiestrogen- and estradiol-dependent transcription activation. J Biol Chem 1996; 271: 24172–24178. Sakamoto T, Eguchi H, Omoto Y, Ayabe T, Mori H, Hayasi S. Estrogen receptor-mediated effects of tamoxifen on human endometrial cancer cells. Mol Cell Endocrinol 2002; 192: 93–104. Miyamoto T, Shiozawa T, Kashima H et al. Estrogen up-regulates mismatch repair activity in normal and malignant endometrial glandular cells. Endocrinology 2006; 147: 4863–4870. Hecht JL, Mutter GL. Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol 2006; 24: 4783– 4791. Kanaya T, Kyo S, Maida Y et al. Frequent hypermethylation of MLH1 promoter in normal endometrium of patients with endometrial cancers. Oncogene 2003; 22: 2352– 2360. Banno K, Yanokura M, Susumu N et al. Relationship of the aberrant DNA hypermethylation of cancer-related genes with carcinogenesis of endometrial cancer. Oncol Rep 2006; 16: 1189–1196. Muraki Y, Banno K, Yanokura M et al. Epigenetic DNA hypermethylation: Clinical applications in endometrial cancer. Oncol Rep 2009; 22: 967–972. Vasen HF, Watson P, Mecklin JP, Lynch HT. New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology 1999; 116: 1453–1456. Boland CR, Shike M. Report from the Jerusalem workshop on Lynch syndrome-hereditary nonpolyposis colorectal cancer. Gastroenterology 2010; 138: 2191–2197. Hampel H, Panescu J, Lockman J et al. Comment on: Screening for Lynch syndrome (hereditary nonpolyposis colorectal cancer) among endometrial cancer patients. Cancer Res 2007; 67: 9603. Meyer LA, Broaddus RR, Lu KH. Endometrial cancer and Lynch syndrome: clinical and pathologic considerations. Cancer Control 2009; 16: 14–22. Peltomäki P, Vasen H. Mutations associated with HNPCC predisposition: Update of ICG-HNPCC/INSiGHT mutation database. Dis Markers 2004; 20: 269–276. Schweizer P, Moisio AL, Kuismanen SA et al. Lack of MSH2 and MSH6 characterizes endometrial but not colon carcinomas in hereditary nonpolyposis colorectal cancer. Cancer Res 2001; 61: 2813–2815.

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

Carcinogenesis of endometrial cancer

19. Kawaguchi M, Banno K, Yanokura M et al. Analysis of candidate target genes for mononucleotide repeat mutation in microsatellite instability-high (MSI-H) endometrial cancer. Int J Oncol 2009; 35: 977–982. 20. Westin SN, Lacour RA, Urbauer DL et al. Carcinoma of the lower uterine segment: A newly described association with Lynch syndrome. J Clin Oncol 2008; 26: 5965–5971. 21. Masuda K, Banno K, Hirasawa A et al. Relationship of lower uterine segment cancer with Lynch syndrome: A novel case with an hMLH1 germline mutation. Oncol Rep 2012; 28: 1537– 1543. 22. Risinger JI, Hayes AK, Berchuck A, Barrett JC. PTEN/ MMAC1 mutations in endometrial cancers. Cancer Res 1997; 57: 4736–4738. 23. Tashiro H, Blazes MS, Wu R et al. Mutations in PTEN are frequent in endometrial carcinoma but rare in other common gynecological malignancies. Cancer Res 1997; 57: 3935–3940. 24. Kurose K, Bando K, Fukino K, Sugisaki Y, Araki T, Emi M. Somatic mutations of the PTEN/MMAC1 gene in fifteen Japanese endometrial cancers: Evidence for inactivation of both alleles. Jpn J Cancer Res 1998; 89: 842–848. 25. Kanaya T, Kyo S, Sakaguchi J et al. Association of mismatch repair deficiency with PTEN frameshift mutations in endometrial cancers and the precursors in a Japanese population. Am J Clin Pathol 2005; 124: 89–96. 26. Schlosshauer PW, Pirog EC, Levine RL, Ellenson LH. Mutational analysis of the CTNNB1 and APC genes in uterine endometrioid carcinoma. Mod Pathol 2000; 13: 1066– 1071. 27. Mirabelli-Primdahl L, Gryfe R, Kim H et al. Beta-catenin mutations are specific for colorectal carcinomas with microsatellite instability but occur in endometrial carcinomas irrespective of mutator pathway. Cancer Res 1999; 59: 3346– 3351. 28. Fukuchi T, Sakamoto M, Tsuda H, Maruyama K, Nozawa S, Hirohashi S. Beta-catenin mutation in carcinoma of the uterine endometrium. Cancer Res 1998; 58: 3526–3528. 29. Saegusa M, Hashimura M, Yoshida T, Okayasu I. β-Catenin mutations and aberrant nuclear expression during endometrial tumorigenesis. Br J Cancer 2001; 84: 209–217. 30. Matias-Guiu X, Catasus L, Bussaglia E et al. Molecular pathology of endometrial hyperplasia and carcinoma. Hum Pathol 2001; 32: 569–577. 31. Sasaki H, Nishii H, Takahashi H et al. Mutation of the Ki-ras protooncogene in human endometrial hyperplasia and carcinoma. Cancer Res 1993; 53: 1906–1910. 32. Enomoto T, Fujita M, Inoue M et al. Alterations of the p53 tumor suppressor gene and its association with activation of the c-K-ras-2 protooncogene in premalignant and malignant lesions of the human uterine endometrium. Cancer Res 1993; 53: 1883–1888. 33. Caduff RF, Johnston CM, Frank TS. Mutations of the Ki-ras oncogene in carcinoma of the endometrium. Am J Pathol 1995; 146: 182–188. 34. Tsuda H, Jiko K, Yajima M et al. Frequent occurrence of c-Kiras gene mutations in well differentiated endometrial adenocarcinoma showing infiltrative local growth with fibrosing stromal response. Int J Gynecol Pathol 1995; 14: 255–259. 35. Lagarda H, Catasus L, Arguelles R, Matias-Guiu X, Prat J. K-ras mutations in endometrial carcinomas with microsatellite instability. J Pathol 2001; 193: 193–199.

36. Halperin R, Zehavi S, Habler L, Hadas E, Bukovsky I, Schneider D. Comparative immunohistochemical study of endometrioid and serous papillary carcinoma of endometrium. Eur J Gynaecol Oncol 2001; 22: 122–126. 37. Esteller M, García A, Martínez i Palones JM, Cabero A, Reventós J. Detection of c-erbB-2/neu and fibroblast growth factor-3/INT-2 but not epidermal growth factor receptor gene amplification in endometrial cancer by differential polymerase chain reaction. Cancer 1995; 75: 2139–2146. 38. Saffari B, Jones LA, El-Naggar A, Felix JC, George J, Press MF. Amplification and overexpression of HER-2/neu (c-erbB2) in endometrial cancers: Correlation with overall survival. Cancer Res 1995; 55: 5693–5698. 39. Coronado PJ, Vidart JA, Lopez-Asenjo JA et al. P53 overexpression predicts endometrial carcinoma recurrence better than HER-2/neu overexpression. Eur J Obstet Gynecol Reprod Biol 2001; 98: 103–108. 40. Manavi M, Bauer M, Baghestanian M et al. Oncogenic potential of c-erbB-2 and its association with c-K-ras in premalignant and malignant lesions of the human uterine endometrium. Tumour Biol 2001; 22: 299–309. 41. Khalifa MA, Mannel RS, Haraway SD, Walker J, Min KW. Expression of EGFR, HER-2/neu, P53, and PCNA in endometrioid, serous papillary, and clear cell endometrial adenocarcinomas. Gynecol Oncol 1994; 53: 84–92. 42. Cianciulli AM, Guadagni F, Marzano R et al. HER-2/neu oncogene amplification and chromosome 17 aneusomy in endometrial carcinoma: Correlation with oncoprotein expression and conventional pathological parameters. J Exp Clin Cancer Res 2003; 22: 265–271. 43. Tashiro H, Isacson C, Levine R, Kurman RJ, Cho KR, Hedrick L. p53 gene mutations are common in uterine serous carcinoma and occur early in their pathogenesis. Am J Pathol 1997; 150: 177–185. 44. Sherman ME, Bur ME, Kurman RJ. p53 in endometrial cancer and its putative precursors: Evidence for diverse pathways of tumorigenesis. Hum Pathol 1995; 26: 1268–1274. 45. Feng YZ, Shiozawa T, Horiuchi A et al. Intratumoral heterogeneous expression of p53 correlates with p53 mutation, Ki-67, and cyclin A expression in endometrioid-type endometrial adenocarcinomas. Virchows Arch 2005; 447: 816–822. 46. Zheng W, Xiang L, Fadare O, Kong B. A proposed model for endometrial serous carcinogenesis. Am J Surg Pathol 2011; 35: e1–e14. 47. Zhang X, Liang SX, Jia L et al. Molecular identification of ‘latent precancers’ for endometrial serous carcinoma in benign-appearing endometrium. Am J Pathol 2009; 174: 2000– 2006. 48. Semczuk A, Marzec B, Roessner A, Jakowicki JA, Wojcierowski J, Schneider-Stock R. Loss of heterozygosity of the retinoblastoma gene is correlated with the altered pRb expression in human endometrial cancer. Virchows Arch 2002; 441: 577–583. 49. Niemann TH, Yilmaz AG, McGaughy VR, Vaccarello L. Retinoblastoma protein expression in endometrial hyperplasia and carcinoma. Gynecol Oncol 1997; 65: 232–236. 50. Shih HC, Shiozawa T, Kato K et al. Immunohistochemical expression of cyclins, cyclin-dependent kinases, tumorsuppressor gene products, Ki-67, and sex steroid receptors in endometrial carcinoma: Positive staining for cyclin A as a poor prognostic indicator. Hum Pathol 2003; 34: 471–478.

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1965

K. Banno et al.

51. Moreno-Bueno G, Rodríguez-Perales S, Sánchez-Estévez C et al. Cyclin D1 gene (CCND1) mutations in endometrial cancer. Oncogene 2003; 22: 6115–6118. 52. DeBernardo RL, Littell RD, Luo H et al. Defining the extent of cables loss in endometrial cancer subtypes and its effectiveness as an inhibitor of cell proliferation in malignant endometrial cells in vitro and in vivo. Cancer Biol Ther 2005; 4: 103–107. 53. Zukerberg LR, DeBernardo RL, Kirley SD et al. Loss of cables, a cyclin-dependent kinase regulatory protein, is associated with the development of endometrial hyperplasia and endometrial cancer. Cancer Res 2004; 64: 202–208. 54. Goldberg AD, Allis CD, Bernstein E. Epigenetics: A landscape takes shape. Cell 2007; 128: 635–638. 55. Ushijima T. Detection and interpretation of altered methylation patterns in cancer cells. Nat Rev Cancer 2005; 5: 223–231. 56. Gal-Yam EN, Jeong S, Tanay A, Egger G, Lee AS, Jones PA. Constitutive nucleosome depletion and ordered factor assembly at the GRP78 promoter revealed by single molecule footprinting. PLoS Genet 2006; 2: 160. 57. Appanah R, Dickerson DR, Goyal P, Groudine M, Lorincz MC. An unmethylated 3′ promoter-proximal region is required for efficient transcription initiation. PLoS Genet 2007; 3: 27. 58. Jones PA, Baylin SB. The epigenomics of cancer. Cell 2007; 128: 683–692. 59. Sakaguchi J, Kyo S, Kanaya T et al. Aberrant expression and mutations of TGF-beta receptor type II gene in endometrial cancer. Gynecol Oncol 2005; 98: 427–433. 60. Velasco A, Pallares J, Santacana M et al. Promoter hypermethylation and expression of sprouty 2 in endometrial carcinoma. Hum Pathol 2011; 42: 185–193. 61. Pallarés J, Velasco A, Eritja N et al. Promoter hypermethylation and reduced expression of RASSF1A are frequent molecular alterations of endometrial carcinoma. Mod Pathol 2008; 21: 691–699. 62. Dewdney SB, Rimel BJ, Thaker PH et al. Aberrant methylation of the X-linked ribosomal S6 kinase RPS6KA6 (RSK4) in endometrial cancers. Clin Cancer Res 2011; 17: 2120– 2129. 63. Yang HJ, Liu VW, Wang Y, Tsang PC, Ngan HY. Differential DNA methylation profiles in gynecological cancers and correlation with clinico-pathological data. BMC Cancer 2006; 6: 212. 64. Kang HS, Baba T, Mandai M et al. GPR54 is a target for suppression of metastasis in endometrial cancer. Mol Cancer Ther 2011; 10: 580–590. 65. Yi TZ, Guo J, Zhou L et al. Prognostic value of E-cadherin expression and CDH1 promoter methylation in patients with endometrial carcinoma. Cancer Invest 2011; 29: 86–92. 66. Whitcomb BP, Mutch DG, Herzog TJ, Rader JS, Gibb RK, Goodfellow PJ. Frequent HOXA11 and THBS2 promoter methylation, and a methylator phenotype in endometrial adenocarcinoma. Clin Cancer Res 2003; 9: 2277–2287. 67. Sasaki M, Kaneuchi M, Sakuragi N, Dahiya R. Multiple promoters of catechol-O-methyltransferase gene are selectively inactivated by CpG hypermethylation in endometrial cancer. Cancer Res 2003; 63: 3101–3106. 68. Ignatov A, Bischoff J, Ignatov T et al. APC promoter hypermethylation is an early event in endometrial tumorigenesis. Cancer Sci 2010; 101: 321–327.

1966

69. Satoh A, Toyota M, Itoh F et al. Epigenetic inactivation of CHFR and sensitivity to microtubule inhibitors in gastric cancer. Cancer Res 2003; 63: 8606–8613. 70. Wang X, Yang Y, Xu C et al. CHFR suppression by hypermethylation sensitizes endometrial cancer cells to paclitaxel. Int J Gynecol Cancer 2011; 21: 996–1003. 71. Di Paola M, Loverro G, Caringella AM, Cormioselvaggi GL. Receptorial and mitochondrial apoptotic pathways in normal and neoplastic human endometrium. Int J Gynecol Cancer 2005; 15: 523–528. 72. Toyota M, Ahuja N, Ohe-Toyota M, Herman JG, Baylin SB, Issa JP. CpG island methylator phenotype in colorectal cancer. Proc Natl Acad Sci U S A 1999; 96: 8681– 8686. 73. Weisenberger DJ, Siegmund KD, Campan M et al. CpG island methylator phenotype underlies sporadic microsatellite instability and is tightly associated with BRAF mutation in colorectal cancer. Nat Genet 2006; 38: 787–793. 74. Zhang QY, Yi DQ, Zhou L, Zhang DH, Zhou TM. Status and significance of CpG island methylator phenotype in endometrial cancer. Gynecol Obstet Invest 2011; 72: 183–191. 75. Holliday R. The inheritance of epigenetic defects. Science 1987; 238: 163–170. 76. Schofield PN, Joyce JA, Lam WK et al. Genomic imprinting and cancer; new paradigms in the genetics of neoplasia. Toxicol Lett 2001; 120: 151–160. 77. Kondo E, Furukawa T, Yoshinaga K et al. Not hMSH2 but hMLH1 is frequently silenced by hypermethylation in endometrial cancer but rarely silenced in pancreatic cancer with microsatellite instability. Int J Oncol 2000; 17: 535–541. 78. Suter CM, Martin DI, Ward RL. Germline epimutation of MLH1 in individuals with multiple cancers. Nat Genet 2004; 36: 497–501. 79. Hitchins MP, Wong JJ, Suthers G et al. Inheritance of a cancerassociated MLH1 germ-line epimutation. N Engl J Med 2007; 356: 697–705. 80. Goel A, Nguyen TP, Leung HC et al. De novo constitutional MLH1 epimutations confer early-onset colorectal cancer in two new sporadic Lynch syndrome cases, with derivation of the epimutation on the paternal allele in one. Int J Cancer 2011; 128: 869–878. 81. Chan TL, Yuen ST, Kong CK et al. Heritable germline epimutation of MSH2 in a family with hereditary nonpolyposis colorectal cancer. Nat Genet 2006; 38: 1178– 1183. 82. van der Gun BT, Melchers LJ, Ruiters MH, de Leij LF, McLaughlin PM, Rots MG. EpCAM in carcinogenesis: The good, the bad or the ugly. Carcinogenesis 2010; 31: 1913– 1921. 83. Ligtenberg MJ, Kuiper RP, Chan TL et al. Heritable somatic methylation and inactivation of MSH2 in families with Lynch syndrome due to deletion of the 3′ exons of TACSTD1. Nat Genet 2008; 41: 112–117. 84. Kozaki K, Imoto I, Mogi S, Omura K, Inazawa J. Exploration of tumor-suppressive microRNAs silenced by DNA hypermethylation in oral cancer. Cancer Res 2008; 68: 2094– 2105. 85. Furuta M, Kozaki KI, Tanaka S, Arii S, Imoto I, Inazawa J. miR-124 and miR-203 are epigenetically silenced tumorsuppressive microRNAs in hepatocellular carcinoma. Carcinogenesis 2010; 31: 766–776.

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

Carcinogenesis of endometrial cancer

86. Pierson J, Hostager B, Fan R, Vibhakar R. Regulation of cyclin dependent kinase 6 by microRNA 124 in medulloblastoma. J Neurooncol 2008; 90: 1–7. 87. Saito Y, Friedman JM, Chihara Y, Egger G, Chuang JC, Liang G. Epigenetic therapy upregulates the tumor suppressor microRNA-126 and its host gene EGFL7 in human cancer cells. Biochem Biophys Res Commun 2009; 379: 726– 731. 88. Nakano H, Miyazawa T, Kinoshita K, Yamada Y, Yoshida T. Functional screening identifies a microRNA, miR-491 that induces apoptosis by targeting Bcl-X(L) in colorectal cancer cells. Int J Cancer 2010; 127: 1072–1080. 89. Huang YW, Liu JC, Deatherage DE et al. Epigenetic repression of microRNA-129-2 leads to overexpression of SOX4 oncogene in endometrial cancer. Cancer Res 2009; 69: 9038– 9046. 90. Tsuruta T, Kozaki K, Uesugi A et al. miR-152 is a tumor suppressor microRNA that is silenced by DNA hypermethylation in endometrial cancer. Cancer Res 2011; 71: 6450–6462. 91. Stumpel DJ, Schotte D, Lange-Turenhout EA et al. Hypermethylation of specific microRNA genes in

92.

93.

94.

95.

96.

97.

MLL-rearranged infant acute lymphoblastic leukemia: Major matters at a micro scale. Leukemia 2011; 25: 429–439. Chen Y, Song Y, Wang Z et al. Altered expression of MiR-148a and MiR-152 in gastrointestinal cancers and its clinical significance. J Gastrointest Surg 2010; 14: 1170–1179. Braconi C, Huang N, Patel T. MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology 2010; 51: 881–890. Chen HZ, Tsai SY, Leone G. Emerging roles of E2Fs in cancer: An exit from cell cycle control. Nat Rev Cancer 2009; 9: 785– 797. Trusolino L, Bertotti A, Comoglio PM. MET signalling: Principles and functions in development, organ regeneration and cancer. Nat Rev Mol Cell Biol 2010; 11: 834–848. Roulin D, Cerantola Y, Dormond-Meuwly A, Demartines N, Dormond O. Targeting mTORC2 inhibits colon cancer cell proliferation in vitro and tumor formation in vivo. Mol Cancer 2010; 9: 57. Guertin DA, Stevens DM, Saitoh M et al. mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell 2009; 15: 148–159.

© 2014 The Authors Journal of Obstetrics and Gynaecology Research © 2014 Japan Society of Obstetrics and Gynecology

1967

Carcinogenic mechanisms of endometrial cancer: involvement of genetics and epigenetics.

Endometrial cancer is increasing worldwide and the number of patients with this disease is likely to continue to grow, including younger patients. Man...
237KB Sizes 0 Downloads 6 Views