You are on page 1of 18

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/221981422

Epigenetics and genetics in endometrial cancer: New carcinogenic


mechanisms and relationship with clinical practice

Article  in  Epigenomics · April 2012


DOI: 10.2217/epi.12.13 · Source: PubMed

CITATIONS READS

33 147

8 authors, including:

Banno Kouji Megumi Yanokura


Keio University Keio University
204 PUBLICATIONS   2,374 CITATIONS    71 PUBLICATIONS   1,283 CITATIONS   

SEE PROFILE SEE PROFILE

Yusuke Kobayashi Nobuyuki Susumu


Keio University Keio University
114 PUBLICATIONS   1,219 CITATIONS    184 PUBLICATIONS   3,242 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Gynecological Oncology Team References View project

Uterine transplantation View project

All content following this page was uploaded by Megumi Yanokura on 07 May 2014.

The user has requested enhancement of the downloaded file.


Review
For reprint orders, please contact: reprints@futuremedicine.com

Epigenetics and genetics in endometrial cancer:


new carcinogenic mechanisms and relationship with
clinical practice
Endometrial cancer is the seventh most common cancer worldwide among females. An increased incidence
and a younger age of patients are also predicted to occur, and therefore elucidation of the pathological
mechanisms is important. However, several aspects of the mechanism of carcinogenesis in the endometrium
remain unclear. Associations with genetic mutations of cancer-related genes have been shown, but these
do not provide a complete explanation. Therefore, epigenetic mechanisms have been examined. Silencing
of genes by DNA hypermethylation, hereditary epimutation of DNA mismatch repair genes and regulation
of gene expression by miRNAs may underlie carcinogenesis in endometrial cancer. New therapies include
targeting epigenetic changes using histone deacetylase inhibitors. Some cases of endometrial cancer may
also be hereditary. Thus, patients with Lynch syndrome which is a hereditary disease, have a higher risk
for developing endometrial cancer than the general population. Identification of such disease-related
genes may contribute to early detection and prevention of endometrial cancer.

KEYWORDS: DNA hypermethylation n DNA mismatch repair n endometrial cancer Kouji Banno*, Iori Kisu,
n epimutation n HDAC inhibitor n hMLH1 n lower uterine segment n Lynch syndrome
n miRNA n MMR
Megumi Yanokura,
Kenta Masuda,
Arisa Ueki,
New carcinogenic mechanisms in by small noncoding RNAs, called miRNAs, Yusuke Kobayashi,
endometrial cancer without a change in the methylation level of Nobuyuki Susumu
Endometrial cancer is the seventh most com- the gene itself has also been shown.
& Daisuke Aoki
mon cancer worldwide among females. In Japan, Most cases of type I endometrial cancer
Department of Obstetrics &
the westernization of lifestyle has increased the result from the malignant transformation after Gynecology, School of Medicine,
number of patients with endometrial cancer, changes from a normal endometrium to atypical Keio University, Shinanomachi 35
Shinjuku-ku, Tokyo 160–8582, Japan
and this disease now accounts for approximately endometrial hyperplasia, a precancerous lesion. *Author for correspondence:
40% of cancers of the uterus. A younger age of This process is thought to be due to accumu- Tel.: +81 3 3353 1211
onset is also predicted, and therefore it is impor- lation of mutations in cancer-related genes, Fax: +81 3 3226 1667
kbanno@sc.itc.keio.ac.jp
tant to elucidate the pathogenesis of the cancer including oncogenes, tumor suppressor genes,
and develop a treatment for it. However, beyond and DNA MMR genes. The cancer thus devel-
the involvement of estrogen, the mechanism of ops in a multistage process, which is referred
carcinogenesis in the endometrium remains to as multistage carcinogenesis. By contrast, if
unclear. Advances in molecular biology of the aberrant cancer-related genes are already present
disease have revealed associations with varia- in germ cells, the lifetime risk for endometrial
tions and mutations of cancer-related genes, but cancer increases, and approximately 5% of cases
these do not provide a complete explanation for of endometrial cancer are caused by a genetic
endometrial carcinogenesis. Therefore, in recent predisposition. Lynch syndrome (hereditary
years, there has been a focus on epigenetic mech- nonpolyposis colorectal cancer), Cowden syn-
anisms, which involve the regulation of gene drome (CS) and Peutz–Jeghers syndrome (PJS)
expression by modulation of chromatin without are hereditary diseases with an increased risk
changes in DNA sequences. Breakdown of the for endometrial cancer, with Lynch syndrome
DNA mismatch repair (MMR) mechanism by being the most frequent disease associated with
aberrant methylation is particularly important hereditary endometrial cancer. Lynch syndrome
for development of type 1 endometrial cancer, is also associated with the aberrant expression
and changes in expression of genes such as of DNA MMR genes, while CS is linked to
hMLH1 and hMSH2 may be involved in this the aberrant expression of PTEN. These results
mechanism. Epimutation, histone deacetylation are important in understanding the carcino-
and hypermethylation may all be important epi- genic mechanism and biological characteris-
genetic carcinogenic mechanisms in endome- tics involved in the development of sporadic
trial cancer, and regulation of gene expression endometrial cancers. part of

10.2217/EPI.12.13 © 2012 Future Medicine Ltd Epigenomics (2012) 4(2), 147–162 ISSN 1750-1911 147
Review Banno, Kisu, Yanokura et al.

Cancer & epigenetics same DNA allows nucleosome formation and


Epigenetics refers to information other than the blocks transcription. Anticancer genes such as
DNA base sequence that is directly stored at the hMLH1, CDKN2A and CDH1 (E‑cadherin) are
time of somatic cell division. Epigenetic mecha- inactivated by DNA methylation of the promoter
nisms of regulation of gene expression include of CpG islands, which suggests that aberrant
DNA methylation, histone modification and DNA methylation may cause carcinogenesis.
the effects of polycomb group proteins. In DNA
methylation in vertebrates, methyl groups are DNA hypermethylation in
added to cytosine bases at CpG sites (a cytosine endometrial cancer
base followed by a guanine base in the DNA Among the epigenetic mechanisms, DNA methy­
sequence) by enzymatic transfer of the methyl lation has been most widely studied. Altered
group from S-adenosyl-l-methionine by DNA DNA methylation has been linked to tumorous
methyltransferases. There are two types of DNA lesions [1,2] and hypermethylation is commonly
methylation: maintenance methylation and associated with the downregulation of gene
de novo methylation. In maintenance methyla- expression. Analysis of methylation in micro-
tion, the methylation pattern is maintained in a scopic amounts of DNA is possible at a high sen-
new DNA strand at the same CpG as that in the sitivity using the PCR. However, aberrant DNA
template DNA. Thus, in DNA replication asso- methylation also occurs in noncancerous cells
ciated with cell division, the methylation pat- and attention should be paid to whether truly
tern in the template DNA is copied into a new cancer-specific DNA methylation is detected.
DNA to maintain the pattern in descendants. Some studies have tried to detect methylation
By contrast, in de novo methylation, CpG sites of genes specific to particular cancers for diag-
are newly methylated in differentiation, aging or nosis. In cancer screening, genes with aberrant
tumorigenesis. DNA methyltransferases, includ- methylation are detected in clinical specimens
ing DNMT1, DNMT2, DNMT3A, DNMT3B mixed with normal cells. Methylation-specific
and DNMT3L, are divided into those associated PCR, in which bisulfite treatment and PCR
with maintenance methylation and those that are combined, can be used to detect aberrantly
produce de novo methylation. DNA methylation methylated genes with high accuracy from a
of CpG islands (regions with a high density of small amount of DNA from cancer cells. In a
CpG sites) in promoters upstream of the start 2009 review, Muraki et al. reported gene silenc-
of gene transcription plays a critical role in gene ing due to hypermethylation of hMLH1 and
expression. When the DNA in this region is not APC, E‑cadherin, CHFR, CASP8, TGF‑bRII,
methylated, a nucleosome does not form and p73, HOXA11 and COMT in endometrial cancer
transcription occurs, while methylation of the [3] . Since this review, further studies have shown
hypermethylation of gene promoters linked to
Table 1. Highly methylated genes in endometrial cancer. reduced expression of SPRY2 [4] , RASSF1A [5] ,
Gene Function Ref. GPR54 [6] , CDH1 [7] and RSK4 [8] through a
hMLH1 DNA mismatch repair [3] similar silencing mechanism (Table 1) . Among
these genes, aberrant methylation is detected
APC Wnt signaling molecule [3,10]
most frequently in hMLH1 in endometrial can-
E‑cadherin Cell–cell adhesion [3] cer and is considered to be an important event
CHFR M phase checkpoint gene [3,11,12] in the early stage of endometrial carcinogenesis;
[3]
however, the mechanism of aberrant methyla-
CASP8 Caspase
tion remains unclear. hMLH1 encodes the DNA
TGF‑bRII Action to INK4a genes [3]
MMR protein and it has been proposed that a
P73 p53 family [3] cascade of malignant transformation occurs in
HOXA11 Proliferation and differentiation of human endometrium [3] which microsatellite instability (MSI) induced
[3]
by the downregulation of hMLH1 due to aber-
COMT Estrogen-converting enzyme
rant methylation induces mutations in hMSH6
Sprouty2 FGF and RAS-MAPK pathway inhibition [4]
and other cancer-related genes such as TGF‑bII,
RASSF1A Gene suppressor gene for RAS-MAPK pathway [5] BAX and PTEN [9] .
GPR54 Endogenous receptor of Kisspeptin, a suppressor of cancer [6] Hypermethylation of the APC promoter is
metastasis not found in the normal endometrium or in
CDH1 Promotor of E‑cadherin [7] endometrial hyperplasia, but is detected in atypi-
cal hyperplasia and early endometrial cancer.
RSK4 Tumor suppressor gene targeting ERK signaling pathway [8]
Interestingly, the frequency of hypermethylation

148 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
in the APC promoter is reduced with progression hypermethylation of CDH1 reduced E‑cadherin
of endometrial cancer, which led Ignatov et al. expression in endometrial cancer, with resulting
to suggest that this hypermethylation may be an effects on clinical and pathological progression
important event in early malignant transforma- and 5‑year survival rates. Hypermethylation of
tion of the endometrium [10] . Satoh et al. linked CDH1 is common in undifferentiated cancers,
hypermethylation to the response of tumors to leading to the suggestion that this mechanism
taxane drugs [11] and Wang et al. found that may be more closely associated with carcino-
reduced expression of CHFR by hypermethyla- genesis than invasive capacity [7] . RSK4 is a
tion improves the response of both stomach and substrate of ERK that inhibits transcriptional
endometrial cancers to paclitaxel [12] . SPRY2 activity of some receptor tyrosine kinases for
is an antagonist regulator of receptor tyrosine certain targets. RSK4 is also associated with the
kinases in the FGF and RAS‑MAPK pathways FGFR2/RAS/ERK signaling pathway and is a
and a tumor suppressor gene involved in cell tumor suppressor. Dewdney et al. showed that
proliferation, differentiation and angiogenesis. expression of RSK4 is reduced by hypermethyla-
Parts of the FGF and RAS‑MAPK pathways are tion in colon, breast and kidney cancer, as well as
changed in endometrial cancer and expression in endometrial cancer, but the tumor inhibitory
of SPRY2 is reduced due to hypermethylation action of RSK4 in the endometrium is currently
in several types of cancer [13–15] . Velasco et al. unclear [8] .
found that SPRY2 in the normal endometrium
is expressed in accord with the menstrual cycle Epimutation & malignant
and suggested that SPRY2 contributes to growth transformation of the endometrium
of glandular structures. Furthermore, SPRY2 Epimutation is a term that refers to the epi­
expression is extremely low in advanced inva- genetic silencing of a gene for which expression is
sive cancers and other types of endometrial can- normally not suppressed, or epigenetic activation
cer, other than endometrioid adenocarcinoma, of a gene for which expression is normally sup-
which indicates that SPRY2 may play a role in pressed [16,17] . Many studies of malignant trans-
suppression of endometrial cancer by regulating formation of the endometrium and epimutation
the MAPK pathway [4] . of genes have been conducted and have mainly
RASSF1A is a tumor suppressor that is a focused on three genes: hMLH1, hMSH2 and
negative regulator in the RAS‑MAPK pathway EPCAM. hMLH1 is a DNA MMR gene. MMR
and, along with SPRY2, has reduced expression deficiency is especially prevalent in endometrial
in various cancers. In a comparison of normal cancer and is found in approximately 20–30%
and cancerous endometrial tissues, Pallares of cases. hMSH2, hMSH6 and PMS2 are also
et al. showed that the RASSF1A promoter was DNA MMR genes and hMLH1 and hMSH2
hypermethylated and that reduced expression of are thought to be involved in endometrial can-
RASSF1A was prevalent in endometrial cancer cer, similarly to colon cancer [18] . Since Kondo
with MSI, especially in advanced cancers. This et al. first showed that epigenetic inhibition of
led to the suggestion that RASSF1A participates hMLH1 expression is more frequent than that of
in cell proliferation and apoptosis by regulating hMSH2 in endometrial cancer [19] , most studies
the MAPK pathway, and has effects on malig- have examined hMLH1 and this gene has been
nant transformation of the endometrium [5] . found to be a tumor suppressor that has reduced
GPR54 is an endogenous receptor of kiss­peptin expression in various cancers.
(KISS1), a suppressor of cancer metastasis, and In endometrial cancer, inactivation by
has reduced expression in some cancers, but methylation of hMLH1 is found in approxi-
an unclear status in endometrial cancer. Kang mately 30% of cases. Inactivation of hMLH1
et al. found high survival rates in cases with high is thought to be an important step in the early
GPR54 expression and showed that expression stage of malignant transformation of the uterus.
of GPR54 is epigenetically regulated. Functional Banno et al. detected aberrant methylation of
recovery of GPR54 was possible with treatment hMLH1 in 40.4% of cases of endometrial can-
with 5-aza-2-deoxycytidine (5‑aza‑dC), a drug cer, and similar aberrant methylation of APC,
that causes DNA hypomethylation, and sub- E‑cadherin and CHFR in 22.0, 14.0 and 12.8%,
sequent expression of GPR54 and activation respectively. In cases with aberrant methylation
of a downstream response pathway involv- of hMLH1 and E‑cadherin, protein expression
ing m ­ etastin-10 were effective for inhibiting was significantly reduced (hMLH1: p < 0.01,
metastasis of endometrial cancer [6] . CDH1 is E‑cadherin: p < 0.05) [20] . Aberrant methylation
a promoter of E‑cadherin. Yi et al. found that of hMLH1 was also detected in 14.3% of cases

future science group www.futuremedicine.com 149


Review Banno, Kisu, Yanokura et al.

of atypical endometrial hyperplasia. However, hMSH2 promoter [26] . Ligtenberg et al. showed
in the normal endometrium, none of the four that epimutation of EPCAM itself is not involved
cancer-related genes were aberrantly methylated. in development of endometrial cancer, but a
On the other hand, mutation of hMLH1 is espe- mutation to the 3´ side of EPCAM upstream and
cially common in cases with multiple primary close to hMSH2 epigenetically silences hMSH2,
cancers such as Lynch syndrome (hereditary l­eading to occurrence of endometrial cancer [26] .
nonpolyposis colorectal cancer) and epimutation
of the germline may be present even if mutation Endometrial cancer & aberrant
of hMLH1 itself does not occur [21] . Hitchins methylation of miRNAs
et al. suggested that epimutation may be inher- miRNAs are short noncoding RNAs of approxi-
ited through the mother, even though germline mately 18–25 bases that regulate expression of
epimutation is thought not to be inherited, and genes. miRNAs have been found to be down-
found that during egg formation an epigenetic regulated by methylation of DNA in various
error is essentially solved by demethylation, cancers, and these miRNAs are referred to as
but methylation may remain in a few cases. t umor-suppressor miRNAs (TS-miRNAs)
­
However, this type of heredity epimutation [27,28] . Identified TS-miRNAs include miR‑124,
occurs with lower frequency than that of germ- miR‑126, miR‑137 and miR‑491 [27–31] .
line sequence variants [22] . Goel et al. showed Using a microarray assay, Huang et al. found
that epimutation of hMLH1 could occur in the that SOX4 genes were highly expressed in
allele from the father, in contrast to the under- endometrial cancer cells. In database ana­lysis,
standing that epimutation of hMLH1 occurred SOX4 was identified as a target for miR‑129­-2,
only in the allele derived from the mother [23] . and a reporter assay showed that miR‑129-2
In two cases in which epimutation of hMLH1 was a negative regulator of SOX4. Expression of
occurred de novo, the allele was derived from the miR‑129-2 was verified in 117 cases of endome-
father, rather than the mother, in one of the cases trial cancer with increased expression of SOX4,
[23] . These cases suggest that a new epimutation and in 68% of the patients SOX4 expression
may occur after fertilization, rather than during was silenced by methylation. Histone acetyla-
egg formation without d­emethylation, and that tion and DNA demethylation increased the
the epimutation can be inherited. expression of miR‑129-2, reduced SOX4 expres-
An inherited germline epimutation of hMSH2 sion, and suppressed proliferation of cancer
was reported in 2006 by Chan et al. [24] in a cells. Furthermore, hypermethylation of the
family line of three patients (brothers and sisters) miR‑129-2 gene was statistically related to MSI
with colon or endometrial cancer with onset at and the methylation status of hMLH1. Thus,
an early age. Genetic mutation of hMSH2 was previously it was thought that a cancer gene
not present, but protein deficiency was recog- could only be activated by hypo­methylation of
nized and MSI was shown, with epimutation of a promoter, but this study proved that hyper­
hMSH2 inherited from the mother. The same methylation of a promoter can also directly
epimutation was inherited by three children of ­activate a cancer gene [32] .
the three patients, indicating that not only a Expression of miR‑152 is also inhibited by
DNA sequence mutation but also an epimuta- DNA hypermethylation [33] . Tsuruta et al. iden-
tion can be inherited over multiple generations. tified miR‑152 as a candidate TS-miRNA in
Sequencing shows that the epimutation does not endometrial cancer through DNA methylation
occur in all cells and that methylation mosaics screening and expression screening of endome-
exist. Different degrees of methylation occur trial cancer cells. Methylation of miR‑152 is
based on the two-hit theory and may be a cause altered in acute lymphocytic leukemia and
of heritability of diseases. miR‑152 expression is changed in digestive sys-
The EPCAM gene codes for an epithelial cell tem cancers and cholangiocellular cancer [34–36] .
adhesion molecule and is overexpressed in most Tsuruta et al. found a high frequency of methyla-
cancers. There is a diversity of opinion on the tion and downregulation of miR‑152 and showed
effects of EPCAM on carcinogenesis. EPCAM that expression of miR‑152 can be recovered
is a homophilic intercellular adhesion molecule by 5‑aza‑dC. Methylation of miR‑152 is com-
that may prevent metastasis, but conversely may pletely consistent with expression, and hyper­
also promote metastasis of cancers by preventing methylation in the promoter region of miR‑152
intercellular adhesion mediated by E‑cadherin reduces expression. DNMT1 is a well-known tar-
[25] . In endometrial cancer, EPCAM deficiency get of miR‑152, and E2F3, MET and Rictor have
is also involved in hypermethylation of the been identified as additional targets by Tsuruta

150 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
et al. [33] E2F3 is an E2F family transcriptional CHFR is also an ubiquitin ligase with substrates
inhibitor and may be a cancer gene [37] . MET including Aurora‑A and polo-like-kinase 1.
is a cell surface receptor for hepatocyte growth When microtubule stress occurs, CHFR is
factor and a known cancer gene [38] . Rictor is a activated, decomposes Aurora‑A and polo-like-
component of mTORC2 (mTOR complex 2), kinase 1, and arrests the cell cycle [45,46] . The
which directly regulates phosphorylation of Akt fork-head associated domain may be associated
and is important in cancer cells [39,40] . Activation with phosphorylated proteins and is important
of mTORC2-Akt signaling contributes to malig- for checkpoint control, but further ana­lysis is
nant transformation of the endometrium, but required to establish the molecules that interact
the action of Rictor in carcinogenesis is unclear. with CHFR.
However, silencing of miR‑152 appears to lead to CHFR is methylated at high rates in cancer
activation of multiple targets, and miR‑152 itself [43] and this has been proposed to have the fol-
and E2F3, MET and Rictor may be new targets lowing relationship with the mitotic index. In
for treatment of endometrial cancer [33] . normally functioning cells without detectable
CHFR methylation, cell division is stopped in the
Application of aberrant DNA G2/M phases by administration of docetaxel, an
methylation in treatment of anticancer agent, and the mitotic index is low. By
endometrial cancer contrast, in cells in which expression of CHFR
Epigenetic patterns in cancers can be restored or is reduced by methylation, a high mitotic index
partially improved by pharmacological action, is found after administration of docetaxel; thus,
unlike irreversible genetic changes. Anticancer the expression level of CHFR and the mitotic
agents can produce epigenetic changes and index are inversely correlated [47] . In some cells,
recovery of expression of inactivated cancer- the checkpoint mechanism of action in response
related genes due to methylation has been to mitotic stress involves transition of cyclin B1
attempted using methylation inhibitors. Lubbert to the nucleus, which is unaffected by CHFR
et al. obtained a response rate of 60% using methylation. In addition, if cells in which CHFR
5‑aza‑dC in patients with myelodysplastic syn- is methylated are treated with 5‑aza‑dC, a meth-
drome (MDS) [41] . In cases that were respon- ylation inhibitor, the checkpoint mechanism
sive to 5‑aza‑dC, expression of p15INK4A was is restored and the mitotic index is reduced.
detected following demethylation. Issa et al. also Methylation of CHFR and sensitivity to micro-
obtained a response rate of 60% with repeated tubule inhibitors (taxanes) are promising mark-
administration of a low dose of 5‑aza‑dC in cases ers in endometrial cancer and in stomach and
of acute myelocytic leukemia [42] . However, a cervical cancer, and the methylation index may
problem with this approach is that demethyla- become a useful marker for prediction of the
tion is not sequence specific and removal of bio- effects of anticancer drugs [48,49] .
logically important methyl groups or activation
of oncogenes that are inactivated by methylation Histone deacetylase & histone
may occur. Therefore, demethylating agents are deacetylase inhibitors in endometrial
being developed that can, for example, demeth- cancer
ylate a DNA sequence bound by a particular The nucleosome, a unit of chromatin, consists
transcription factor. of a core tetramer of histone pairs of H3–H4
Utilization of epigenetic aberrations as mark- dimers that form an octamer with two histone
ers of sensitivity to anticancer drugs is also being H2A–H2B dimers, with approximately 147 bp
examined. Satoh et al. found that aberrant meth- of DNA wrapping around the octamer about
ylation of CHFR, an M phase checkpoint gene, 1.75‑times. The crystal structure of the nucleo-
is correlated with the sensitivity of tumors to some shows that the N‑terminus of histones pro-
microtubule inhibitors [43] . This finding sug- trudes to sites outside the DNA. The N‑terminal
gests that treatment may be selected based on the domains of the histone core proteins (histone
specific methylation properties of tumor cells. tails) undergo post-translational modifications,
CHFR is likely to be a marker for the response of including acetylation, methylation, ubiquiti-
endometrial cancer to anticancer drugs. CHFR nation and phosphorylation. Combinations of
includes a fork-head associated domain and a histone modifications define the status of acti-
RING finger domain, and was first identified vation of a chromosome, and therefore this is
as a homolog of the yeast DMA1 gene [44] . If considered to be a kind of genetic code. Histone
mitotic stress occurs during M phase, CHFR acetylation removes positive charges of lysine
delays the progress from early to late prophase. residues and decreases electrostatic interactions

future science group www.futuremedicine.com 151


Review Banno, Kisu, Yanokura et al.

with the negatively charged bases of DNA. This HDAC inhibitors elevate acetylation of his-
reduces the affinity of the histone for the DNA tones and enhance transcriptional activity, lead-
and leads to conversion of heterochromatin to ing to antitumor effects through induction of
euchromatin, allowing cofactors to interact with apoptosis, cell cycle arrest, antiangiogenesis,
DNA and increase transcriptional activity. and cell differentiation. HDAC inhibitors can
Histone acetylation and deacetylation are increase expression of p21WAE1, which, along with
catalyzed by histone acetyl transferase and his- p27KIP1, is a cyclin-dependent kinase inhibitor
tone deacetylase (HDAC) enzymes, respectively. that decreases the kinase activity of the cyclin-
Histone acetylation activates binding of tran- dependent kinase complex, leading to arrest of
scription factors and enhances transcriptional the cell cycle in the G0/G1 phase [54] . HDAC
activation, while deacetylation inhibits transcrip- inhibitors elevate acetylation of histones in the
tion. The 18 known HDACs are classified into p21WAF1 promoter through selective inhibition of
classes I–IV based on their amino acid sequence particular HDACs, which restores production of
homology at sites of enzyme activity [50,51] . Class I cyclin D1 and D2 and inhibits cell proliferation
enzymes (HDAC 1, 2, 3 and 8) are homologs [55] . Many basic studies have shown effects of
of yeast Rpd3 that are present mainly in nuclei HDAC inhibitors on endometrial cancer cells.
and are constitutively expressed in many cells HDAC inhibitors can also reduce bc1–2 expres-
and tissues. Class II enzymes (HDAC 4, 5, 6, sion, thereby blocking inhibition of apoptosis
7, 9 and 10) are homologs of yeast Hda1 and in endometrial cancer cells, bind to b‑catenin,
are mainly present in the brain, myocardium and increase the expression of E‑cadherin, a
and skeletal muscles. Class II is further classi- tumor suppressor gene. HDAC inhibitors can
fied into classes IIa (HDAC 4, 5, 7 and 9) and also inhibit the expression of angiogenic growth
IIb (HDAC 6 and 10). Class III enzymes are factors such as HIF‑1a, VEGF, VEGF receptor,
Sir2 homologs and include SIRT 1, 2, 3, 4, 5, endothelial NO synthase, IL‑2 and IL‑8; induce
6 and 7. Class IV currently includes HDAC11 antiangiogenic factors such as p53 and the von
alone. The relationship between HDAC expres- Hippel-Lindau tumor suppressor; and inhibit
sion and endometrial malignant transformation angiogenesis [56] . As noted above, many tran-
remains unclear. Class I HDAC expression in scription factors and coregulators in transcrip-
the endometrium and in endometrial cancer tion are regulated by acetylation. Thus, HDAC
has recently been shown for the first time [52,53] . inhibitors can alter gene transcription by regulat-
HDAC expression occurs throughout the men- ing the extent of acetylation of histones for tar-
strual cycle, but HDAC3 expression decreases get genes including ACTR, cMyb, E2F1, EKLF,
from menstrual days 6–10, suggesting that FEN, GATA, HNF‑4, HSP90, Ku70, NF‑kB,
HDAC expression is involved in endometrial dif- PCNA, p53, RB, Runx, SF1, Sp3, STAT, TFIIE,
ferentiation [52] . Endometrial cancer with strong TCF 1 and YY1 [57] . A mechanism through
class I HDAC expression may have a poor prog- which HDAC inhibitors induce differentiation
nosis [53] and it is of note that many studies have in endometrial cancer cells via glycodelin has
shown antitumor effects of HDAC ­inhibitors on also been described [58] .
cancer, including e­ ndometrial cancer. Clinical studies of HDAC inhibitors are cur-
rently ongoing in solid cancers and hemato-
Table 2. Effect of histone deacetylase inhibitors in endometrial logic tumors. The inhibitors are classified into
cancer cells. hydroxamic acids, short-chain fatty acids, benza-
mides, cyclic tetrapeptides and sulfon­amide ani-
Group Substance HDAC inhibited ED50 (M) Ref. lides, based on their chemical structure. HDAC
Hydroxamate TSA Class I, II 5.2 × 10 -8
[59,60] inhibitors in clinical trials in endometrial can-
SAHA Class I, II, IV 7.8–31.0 × 10 -7 [61–63] cer, the HDAC isoforms that are inhibited, and
CBHA Class I 1.8–2.5 × 10 -6 [64] the preliminary results of the clinical trials are
Scriptaid Class I 9.0 × 10 -6 [65]
shown in Table 2 . Trichostatin A (TSA) was iso-
Oxamflatin Class I 2.5 × 10 -7 [67]
lated from Actinomyces media as a differentia-
Short-chain fatty acid NaB Class I, IIa 8.3 × 10 -4 [66] tion-inducer for mouse erythroblastic leukemia
Valproate Class I, IIa 4.1 × 10 -4 [63]
cells and was found to have HDAC inhibitory
Benzamides MS‑275 Class I, II 5.0–22.0 × 10 -7 [67] activity. In vitro, TSA has anti-tumor activity
M344 Class I, II 2.3 × 10 -6 [68] as an HDAC inhibitor and promotes cell dif-
Cyclic tetrapeptides Apicidin Class I, II 1.0 × 10 -6 [69] ferentiation. However, in vivo, the use of TSA
CBHA: m‑carboxycinnamic acid bishydroxamic; ED50 : 50% effective dose; HDAC: Histone is restricted due to severe adverse reactions.
deacetylase; NaB: Sodium butyrate; SAHA: Suberoylanilide hydroxamic acid; TSA: Trichostatin A. Dowdy et al. showed that TSA and paclitaxel

152 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
in combination inhibit the growth of Ark2 and classified into epoxyketones (e.g., HC‑toxin and
KLE endometrial cancer cells, with findings of trapoxin) and nonepoxyketones (e.g., apicidin,
increased acetylation of tubulin and microtubule depsipeptide and FK228). Ueda et al. showed
stabilization [59,60] . that apicidin arrested the cell cycle and promoted
The most advanced HDAC inhibitor clini- gene expression associated with apoptosis induc-
cally is suberoylanilide hydroxamic acid, which tion [69] . Oxamflatin is a sulfonamide derivative
is currently in Phase I and II clinical trials that induces morphological recovery of K‑ras-
[61,62] . In contrast to the adverse effects of TSA, transformed NIH3T3 cells and of malignant
suberoylanilide hydroxamic acid has a high tol- transformation induced by oncogenes such as
erability and good efficacy at a low dose, and v‑src and v‑fos. Jiang et al. conducted a study of
can be administered orally or intravenously [63] . oxamflatin in Ark2 endometrial cancer cells and
m‑carboxycinnamic acid bishydroxamic is a syn- found that the tumor cells decreased by 78 and
thetic HDAC inhibitor that inhibits cell prolif- 95% at 4 days after treatment at doses of 0.25
eration and induces cell differentiation in vitro at and 0.75 µM, respectively. Thus, many HDAC
micromolar concentrations. Takai et al. showed inhibitors have useful properties in vitro, but
that normal endometrial cells survived after their clinical application as anti-tumor agents
m‑carboxycinnamic acid bishydroxamic was in endometrial cancer remains to be established.
administered at a dose inhibiting proliferation
of endometrial cancer cells [64] . Scriptaid is a Endometrial cancer as a
HDAC inhibitor in the hydroxamic acid class hereditary tumor
that targets the TGF‑b pathway, including Tumors with aberrant accumulation within fam-
SMD4, and activates tumor suppressor genes ilies and with a strong genetic background are
[65] . Sodium butyrate (NaB) is endogenous in referred to as hereditary tumors. Most heredi-
the human colon and may have efficacy in pre- tary tumors develop after a single gene associated
cancerous conditions and for tumorous lesions. with cancer susceptibility undergoes a germline
NaB was shown to be a HDAC inhibitor in vitro mutation. Tumor genes, ­tumor-suppressor genes
in 1978, and Terao found that NaB inhibits and DNA MMR genes have been shown to
proliferation and promotes differentiation of be disease-related genes in hereditary tumors.
endometrial cancer cells. However, the half life Tumor genes are activated and result in malig-
of NaB is only 5 min and the concentration rap- nant transformation when one of a pair of genes
idly decreases after intravenous administration; is mutated. By contrast, tumor suppressor genes
therefore, a dose of 400 mg/kg/day is required and DNA MMR genes result in malignant
and clinical use is difficult [66] . Valproic acid transformation when both of a pair of genes are
(VPA), a short-chain fatty acid, is an HDAC inactivated. In hereditary tumors, a mutation of
inhibitor that is used for treatment of epilepsy either tumor suppressor gene may be inherited
in clinical practice, and therefore many stud- from a parent; therefore, a tumor results due to
ies of VPA have been conducted. VPA is a safe malignant transformation once the other allele
drug with less cell toxicity than HDAC inhibi- mutates. Some cases of endometrial cancer
tors such as TSA and has clinical potential due develop due to hereditary predisposition, with
to its long half-life [63] . a high risk in several syndromes. Elucidation of
MS‑275 is a benzamide that induces expres- aberrant genes specific to these syndromes has
sion of the cyclin-dependent kinase inhibi- helped to clarify the carcinogenic mechanism of
tor p21WAF1 and gelsolin, and regulates the cell hereditary endometrial cancer.
cycle. MS‑275 has anticancer effects in vivo and
in vitro, and clinical trials are ongoing in solid Endometrial cancer & Lynch
cancers and hematologic tumors. Jiang et al. syndrome
showed that the number of Ark2 endometrial Lynch syndrome is a typical familial tumor
cancer cells decreased by approximately 60% with autosomal dominant inheritance. Female
after 4‑day administration of 0.5 µM MS‑275 patients with Lynch syndrome complicate with
[67] . M344 is a synthetic amide with a structure endometrial cancer at a high incidence. In the
similar to TSA. Jung et al. showed that M344 revised 1999 Amsterdam II Criteria (ACII)
had activity as a HDAC inhibitor and promoted endometrial cancer was included as a cancer sim-
cell differentiation [68] . Apicidin is a cyclic ilar to colon, small intestine, ureteral and kid-
tetrapeptide with antiprotozoal activity for api- ney cancers (Box 1) [70] . The prevalence of Lynch
complexan parasites and has been shown to be syndrome is 0.9–2.7% [71] , and approximately
a HDAC inhibitor. Such cyclic tetrapeptides are 2.3% of cases of endometrial cancer occur due

future science group www.futuremedicine.com 153


Review Banno, Kisu, Yanokura et al.

to Lynch syndrome [72] . The lifetime risk of was also found in group C. Stage III and IV
endometrial cancer is 40–60% in females with advanced cancer was more common in groups B
aberrant genes associated with Lynch syndrome and C compared with group A [79] . Thus, cases
(Table 3) [73–75] . of MSI-positive sporadic endometrial cancer due
Aberrant MMR genes are involved in carcino- to MLH1 promoter methylation had less differ-
genesis of endometrial cancer. Cloning has been entiation and most were advanced cancer; by
performed in six MMR genes (MSH2, MLH1, contrast, Lynch syndrome-related endometrial
MLH3, MSH6, PMS1 and PMS2) and most cancer due to MLH1 or MSH2 gene mutation
mutations occur in PMS2 and MSH2, in par- was clinico­pathologically similar to endo­metrial
ticular, and in MLH1 and MSH6 (Table 4) [76] . cancer with onset at an early age. However,
In endometrial cancer, MSH2 and MSH6 are Lynch syndrome-related endometrial cancer had
strongly associated with carcinogenesis [77,78] . a variety of histo­logical types, including nonen-
If aberration occurs in these MMR genes, mis- dometrioid adenocarcinoma, and had different
match bases cannot be correctly repaired, result- characteristics from sporadic endometrial can-
ing in DNA strands of different length. This cer. In the study by Broaddus et al., most patients
phenomenon occurs in microsatellite regions of with Lynch syndrome had a MSH2 mutation;
the human genome and is referred to as MSI. consequently, the clinicopathological character­
Most short-tandem repeats such as micro­ istics of patients with a MLH1 mutation is
satellites exists in noncoding regions and have unknown. The onset age of sporadic endome-
no relation to production of aberrant proteins trial cancer was 60 years old, whereas that of
with mutations. However, short-tandem repeats Lynch syndrome-related endometrial cancer was
can occur in regions coding genes for which only 48 years old [80] . However, the prognosis
mutation is involved in carcinogenesis, includ- of Lynch syndrome-related endometrial cancer
ing BAX, which is related to apoptosis induction, did not differ from that of sporadic endometrial
and IGF2R, which is associated with inhibition cancer [81] .
of cell proliferation.
Broaddus et al. examined the clinico­ Hereditary endometrial cancer not
pathological characteristics of 50 patients with meeting the ACII
Lynch syndrome-related endometrial cancer Definitive diagnosis of Lynch syndrome is per-
(group A), 42 patients with sporadic endo­ formed by a MMR genetic test in germ cells, but
metrial cancer who were aged <50 years old ACII are used clinically. However, the results
and had no aberrant mutation (group B), and of germline MMR gene mutations show that
26 patients with loss of MLH protein expression some patients with a mutation do not meet ACII.
and MSI-positive sporadic endometrial cancer Hampel et al. conducted a genetic ana­lysis of
due to MLH promoter methylation (group C). MMR in 543 patients with endometrial cancer
The mean ages in the three groups were 46.8, and detected a gene mutation in 10 (1.8%) of
39.9 and 61.1 years old, respectively, with a sig- these cases (one in MLH1, two in MSH2 and six
nificant difference between groups A and C. in MSH6 ), of which seven did not meet ACII
There was slightly more frequent occurrence [82] . ACII does not include Lynch syndrome-
of nonendometrioid adenocarcinoma (clear cell related cancers such as gastric, breast and ovar-
adenocarcinoma, serous adenocarcinoma and ian cancers, and therefore these criteria alone
carcinosarcoma) in group A (14%). Regarding cannot detect all cases of hereditary endometrial
differentiation, G2 and G3 cells were frequently cancer.
detected in group C and vascular invasion The Japanese working group on familial
endometrial cancer conducted a genetic ana­
lysis in patients who were suspected to have
Box 1. The Amsterdam II Criteria.
Lynch syndrome, although they did not meet
ƒƒ There should be at least three relatives with colorectal cancer or with an ACII. Lynch syndrome-related tumors (colon,
hereditary nonpolyposis colorectal cancer-associated cancer: cancer of the endometrial, small intestinal, ureteral, renal
endometrium, small bowel, ureter or renal pelvis
pelvic, gastric, breast and ovarian cancers) were
ƒƒ One relative should be a first degree relative of the other two found in at least two families (including first-
ƒƒ At least two successive generations should be affected degree relative probands). A genetic ana­lysis was
ƒƒ At least one tumor should be diagnosed before the age of 50 years performed in 120 patients, including 57 with
ƒƒ Familial adenomatous polyposis should be excluded in the colorectal cancer endometrial cancer in whom the age of onset of
case if any at least one tumor was ≤50 years old (group A),
ƒƒ Tumors should be verified by histopathological examination 48 with endometrial cancer and simultaneous

154 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
or metachronous complication with a Lynch Table 3. Risks of patients with hereditary diseases developing
syndrome-related tumor (group B), and 15 endometrial cancer throughout life.
with endometrial cancer who met the criteria
Disease Risk throughout life (%) Ref.
for groups A and B (group A + B). Peripheral
blood leukocyte-derived DNA was obtained Lynch syndrome 42 (diseased until 70 years old) [74]

and germline mutations in three MMR genes 61 (MSH2 mutation) [75]


42 (MLH1 mutation) [75]
(MLH1, MSH2 and MSH6 ) were analyzed by
direct sequence ana­lysis. Gene mutations were Cowden syndrome 5–10 [100]

found in 18 (15%) of the 120 patients: nine Peutz–Jeghers syndrome 9 [110]


(15.8%) in group A, four (8.3%) in group B,
and five (33.3%) in group A + B. Of 18 patients and colon cancers at <50 years old, those with
with genetic mutations, five (27.8%) had a endometrial and ovarian cancers who simulta-
mutation in MLH1, four (22.2%) in MSH2 neously or metachronously complicated with
and nine (50%) in MSH6. Of nine patients a Lynch syndrome-related tumor, those with
with a mutation in MSH6, eight had mutations endometrial and colon cancers with at least two
mainly in exons 4–6 and most were frameshift first- or second-degree relatives who developed
mutations. On the other hand, in patients Lynch syndrome-related tumor, and those with a
with double cancer who had genetic mutations first or second-degree relative who met the above
(group B: 4, group A + B: 5), five and one of criteria had more than a 5–10% probability of
six patients with double cancer including colon a hereditary predisposition for endometrial and
cancer had mutations in MLH1 and MSH2, colon cancers; therefore, making evaluation of
respectively. By contrast, all three patients with hereditary risks useful [85] .
double cancer including breast or ovarian can- MSI ana­lysis and immunohistochemical stain-
cer had mutations in MSH6. All of these dou- ing are useful in screening to identify subjects
ble cancers developed simultaneously with or for a mutation test for Lynch syndrome. The
before development of endometrial cancer [83,84] . NCI recommends five satellite markers (BAT25,
These results suggest that there may be many BAT26, D2S123, D5S346 and D173250) for
cases of Lynch syndrome-related endometrial MSI ana­lysis [86] . A case with an allelic shift in at
cancer and that genetic analyses for MMR least two markers is defined as MSI-high and that
genes are useful in patients with endometrial with only one marker as MSI-low. However, most
or double cancer showing a familial cluster of MSI reflects inactivity due to epigenetic changes
cancer. Furthermore, the MSH6 gene appears in MMR genes, and particularly MLH1 promoter
to be strongly associated with carcinogenesis of methylation, which occurs in sporadic endome-
endometrial cancer. trial cancer at a rate of approximately 20% [87] .
Therefore, the value of MSI as screening for
Screening & surveillance for Lynch Lynch syndrome is not always high in patients
syndrome-related endometrial with endometrial cancer. On the other hand,
cancer immunohistochemical staining may be effective
In general, hereditary tumors tend to have for identifying aberrant MMR genes [88] .
juvenile onset, multiple organ double cancer, Patients with Lynch syndrome and persons
and bilateral occurrence. The guidelines of the from a family with Lynch syndrome have a high
Society of Gynecologic Oncologists Education risk of other cancers, and therefore appropriate
Committee are useful for specific diagnosis of screening and preventive measures for cancer
Lynch syndrome [85] . These guidelines suggest are needed. Lindor et al. proposed screening for
that patients with endometrial and colon cancers
who meet ACII, those who simultaneously or Table 4. Mismatch repair genes related to Lynch syndrome.
metachronously develop endometrial and colon Gene Chromosomal cDNA size Number of Gene size
cancers at <50 years old, those who simultane- position (kb) exons (kb)
ously or metachronously develop ovarian and MSH2 2p21 2.8 16 73
colon cancers at <50 years old, and those with
MLH1 3p21–p23 2.3 19 58–100
a first- or second-degree relative with aber-
rant MMR genes have a 20–25% probability MLH3 14q24.3 4.3 12 37
for a hereditary predisposition to endometrial MSH6 2p21 4.2 10 20
and colon cancers. Therefore, hereditary risks PMS1 2q31 2.8 12 93
should be evaluated in these patients. It has been
shown that patients diagnosed with endometrial PMS2 7q22 2.6 15 16

future science group www.futuremedicine.com 155


Review Banno, Kisu, Yanokura et al.

females with Lynch syndrome using colonos- Endometrial cancer & CS


copy (once every 1–2 years from 20–25 years CS is a rare disease with autosomal dominant
old), endometrial sampling (once a year from inheritance and has characteristics of multiple
30–35 years old), transvaginal ultrasonography hamartomas that occur in various tissues. The
(once a year from 30–35 years old), urine cytol- prevalence of CS is estimated to be one per
ogy (once every 1–2 years from 25–35 years old), 200,000–250,000 population [97] . Patients with
and history taking and physical examination CS have increased risks for malignant tumors,
including genetic counseling and review (every and particularly breast, thyroid and endome-
year from 21 years old), with hysterectomy plus trial cancers. Onset of CS is thought to involve
adnexectomy to be considered if a patient does PTEN [98] and approximately 80% of patients
not want a child [89] . However, the effective- with CS have a PTEN mutation [99] . The life-
ness of these surveillance methods remains time risk for endometrial cancer is 5–10% in
unclear [90] . patients with CS [100] , compared with 2–4% in
the general population (Table 3) . PTEN codes for
Carcinoma of the lower uterine a protein with tyrosine kinase activity and acts
segment & Lynch syndrome as a tumor suppressor gene. PTEN is also a phos-
Endometrial cancer that occurs in the lower phatidylinositol phosphatase that regulates PI3K
uterine body is referred to as carcinoma of the activity and inhibits activation of Akt via PI3K.
lower uterine segment (LUS) and has recently Akt is a serine/threonine kinase that activates
been shown to be a hereditary cancer [91] . or inactivates downstream factors by phosphor-
Carcinoma of the LUS accounts for 3–8% of ylating serine/threonine residues, with resulting
cases of endometrial cancer [92–94] ; however, it transmission of signals related to cell growth,
has characteristics of advanced muscle invasion survival, differentiation and glucose metabo-
and juvenile onset in comparison with non-LUS lism [101] . Akt is related to cell survival through
carcinoma [92,93,95] . Westin et al. conducted a inactivation of proapoptotic executioner caspases
large-scale comparative study in 35 patients and transcription factors related to apoptosis-
with carcinoma of the LUS and 974 with non- inducing factors. Therefore, if PTEN regulation
LUS carcinoma. The mean ages were 54.2 and of Akt activation is lost, the PI3K‑Akt pathway is
62.9 years old in the LUS and non-LUS cases, activated and leads to malignant transformation
respectively, with a significant difference in of endometrial cells [102,103] . Screening criteria
the age of onset. The younger age of onset of for endometrial cancer in patients with CS are
carcinoma of the LUS was suggested to indi- still under discussion [104–106] . In this context,
cate a hereditary predisposition. Five of the it is of note that a recent study showed a rela-
35 patients with carcinoma of the LUS (14.2%) tionship of adolescent-onset endometrial cancer
met the ACII criteria. All of these patients had with CS [107] .
a MSH2 mutation, were highly MSI-positive,
and showed loss of MSH2 and MSH6 proteins. Endometrial cancer & PJS
Four patients (11.4%) did not meet ACII, but PJS is characterized by multiple hamartomatous
had loss of MSH2 and MSH6 proteins, and polyps in the gastrointestinal tract and muco-
one patient (2.8%) did not meet ACII, but had cutaneous pigmentation. Patients with PJS
loss of MSH2 and MSH6 proteins. Based on have a higher risk of developing a malignant
these results, ten (29%) of the 35 patients were tumor in the gastrointestinal tract and other
concluded to have carcinoma of the LUS associ- organs compared with the general population.
ated with Lynch syndrome. In comparison with LKB1/STK11 has been identified as a disease-
the approximately 2.3% incidence of Lynch syn- related gene with autosomal dominant inher-
drome in all cases of endometrial cancer [72] , the itance, and a LKB1 gene mutation is found in
incidence of Lynch syndrome in cases of carci- 80–94% of patients with PJS [108] . The incidence
noma of the LUS was extremely high, which sug- of PJS is estimated to be one per 50,000–250,000
gests that carcinoma of the LUS is a characteris- population [109] . Patients with PJS are at risk for
tic phenotype of endometrial cancer that occurs gynecological cancers and have a 9% lifetime
in patients with Lynch syndrome. Westin et al. risk of developing endometrial cancer [110] . LKB1
suggested that the sensitivity to DNA MMR was codes for a serine/threonine kinase and has
high due to specificity of the endometrial duct functions including regulation of glucose, lipid
and interstitium in the LUS, but the reason why metabolism, cell proliferation and cell polarity.
Lynch syndrome is associated with carcinoma of A relationship of LKB1 downregulation with
the LUS remains unclear [93,96] . endometrial carcinogenesis has been found using

156 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
knockout mice, with the suggestion that LKB1 also been developed, but are not used in clini-
inactivation correlates with endometrial malig- cal practice due to cell toxicity. Many HDAC
nant pheno­t ypes and that LKB1 downregulation inhibitors (i.e., anticancer agents targeting aber-
may be a prognostic factor in endometrial cancer rant acetylation) have also been developed and
[111] . Screening for endometrial cancer similar improved drugs with focused targeting may be
to that used in Lynch syndrome can be used useful in clinical practice.
in patients with PJS; however, it has been sug- Lynch syndrome, CS and PJS are hereditary
gested that specific surveillance is unnecessary diseases associated with endometrial cancer, and
based on the relatively low risk of development Lynch syndrome is particularly important with
of endometrial cancer (Table 3) [110] . Cases of mul- regard to the prevalence and risk for develop-
tiple cancer including endometrial cancer have ing endometrial cancer. In most patients with
also been reported in patients with PJS [112] . Lynch syndrome, endometrial cancer is likely
to be a sentinel cancer; therefore, it is impor-
Conclusion & future perspective tant to suspect Lynch syndrome if juvenile
In 2007, endometrial cancer was newly diag- endometrial cancer is diagnosed. ACII crite-
nosed in 226,000 women worldwide. It is the ria are used for diagnosis of Lynch syndrome,
cause of death of more than 7000 people annu- but genetic ana­lysis should also be performed
ally in the USA alone. In Japan, the number in high-risk patients with familial accumula-
of patients with this cancer is increasing and tion, even though they do not meet diagnos-
a younger age of onset is also predicted, which tic criteria. For patients with a hereditary pre­
emphasizes the need to find more effective disposition, screening and preventive measures
therapies based on improved understanding of are recommended, but their efficacy is currently
the pathogenesis. As for many other cancers, unknown. Screening for endometrial cancer
development of endometrial cancer cannot will also be important in patients with CS and
be fully explained by genetic mutations alone PJS, but the risk for developing endometrial
and is likely to involve epigenetic changes. In cancer is lower than that in Lynch syndrome.
endometrial cancer, mutations of MMR genes Investigation of the extent to which survival is
are important and regulation of these genes has improved by current screening and preventive
been widely examined. Expression of hMLH1, measures and establishment of new diagnostic
hMSH2 and EPCAM is suppressed by pro- criteria and screening methods are required. A
moter hypermethylation, which inhibits direct relationship with disease-related genes in carci-
or indirect DNA MMR and contributes to noma of the LUS and Lynch syndrome has been
development of endometrial cancer type 1 and suggested. Clarification of this relationship and
development of endometrial cancer in Lynch discovery of factors associated with the onset
syndrome. Reduced expression of genes such of hereditary endometrial cancer will require
as APC, E‑cadherin, CHFR, SPRY2, RASSF1A, large-scale global studies.
GPR54, CDH1 and RSK4 through a similar Next-generation sequencing has provided
mechanism of hypermethylation has also been genome-wide analyses of genetics and epi­
found in endometrial cancer. Suppression genetics, and consequently, genome-wide
of gene expression by miRNAs also occurs association studies have attracted attention.
in endometrial cancer and expression of the Genome-wide analyses of lung cancer and
miRNA itself may be increased or decreased melanoma [113,114] have identified mutations
by promoter methylation, based on differ- in at least 10,000 somatic cells and it is likely
ences between normal and cancerous endo­ that dozens of missense mutations are present
metrial tissue, and may contribute to malignant in protein-coding genes. Genome-wide com-
­transformation of the endometrium. parison has shown that mutation from guanine
More knowledge of the mechanism of malig- to thymine is common in pulmonary adeno-
nant transformation of the endometrium is likely carcinoma, whereas mutation from cytosine
to reveal new therapeutic targets and lead to the to thymine is significantly more frequent in
discovery of new drugs. This is especially true melanoma. Genome-wide analyses are pro-
for miRNA targeting, since siRNA modulation moting development of catalogs of genomic
of miRNAs has been established in vitro and aberrations in all cancers, and large-scale can-
a similar approach in vivo may prevent pro- cer genome projects including The Cancer
gression of carcinogenesis caused by silencing Genome Atlas (TCGA [201]) in the USA and
of miRNA. Drugs that cause gene demethyla- the International Cancer Genome Consortium
tion (DNA methyltransferase inhibitors) have [202] , including research groups from 11 nations,

future science group www.futuremedicine.com 157


Review Banno, Kisu, Yanokura et al.

have been started [115–117] . TCGA started with Financial & competing interests disclosure
studies in lung and ovarian cancer and glio­ The authors gratefully acknowledge grant support from the
blastoma, and analyses are currently ongoing in Japan Society for the Promotion of Science (JSPS) through
at least 20 kinds of cancer, including endome- a Grant-in-Aid for Scientific Research (KAKENHI); a
trial cancer. However, a genome-wide study Grant-in-Aid for Scientific Research (C) (22591866) and
of methylation in endometrial cancer has not a Grant-in-Aid for Young Scientists (B) (21791573); the
yet been performed, excluding one study that Ichiro Kanehara Foundation; and the Keio University
used an array-based approach [118] . However, Medical Science Fund through a Research Grant for Life
data from 373 genome-wide studies of methyla- Sciences and Medicine. The authors have no other relevant
tion in endometrial cancer are currently reg- affiliations or financial involvement with any organization
istered in TCGA and this number continues or entity with a financial interest in or financial conflict
to increase. Thus, genome-wide gene mutation with the subject matter or materials d­ iscussed in the
and methylation analyses are likely to produce ­manuscript apart from those disclosed.
further breakthroughs in understanding of the No writing assistance was utilized in the production of
­carcinogenic mechanism of ­endometrial cancer. this manuscript.

Executive summary
Carcinogenic mechanism in endometrial cancer
ƒƒ Endometrial cancer is the seventh most common cancer worldwide among females and accounts for approximately 40% of cancers of
the uterus in Japan.
ƒƒ In addition to the involvement of hormones such as estrogen, epigenetic and genetic aberrations are important for carcinogenesis of
type I endometrial cancer.
Epigenetics in endometrial cancer
ƒƒ hMLH1, APC, E‑cadherin, CHFR, SPRY2, RASSF1A, GPR54, CDH1 and RSK4 are genes that are hypermethylated in endometrial cancer.
Aberrant promoter methylation of hMLH1 occurs in approximately 40% of cases and is an early-stage event in the malignant
transformation of the endometrium.
ƒƒ Epimutation of DNA mismatch repair genes hMLH1 and hMSH2 is a new mechanism of carcinogenesis in endometrial cancer.
ƒƒ miR‑152 is a tumor suppressor-type miRNA that shows decreased expression due to aberrant methylation in endometrial cancer. This
miRNA inhibits cell proliferation of endometrial cancer in vivo and in vitro.
ƒƒ Aberrant methylation of CHFR is a potential biomarker for sensitivity of endometrial cancer to treatment with taxanes.
ƒƒ Histone deacetylase inhibitors are potential new drugs for the treatment of endometrial cancer.
Endometrial cancer as a hereditary tumor
ƒƒ Some cases of endometrial cancer are hereditary tumors. Patients with Lynch syndrome (hereditary nonpolyposis colorectal cancer),
Cowden syndrome and Peutz–Jeghers syndrome, which are hereditary diseases, have a higher risk for endometrial cancer than the
general population. MSH6, a mismatch repair gene, PTEN and LKB1/STK11 have been identified as disease-related genes in these
hereditary conditions.
ƒƒ Lynch syndrome is found in approximately 2% of all types of endometrial cancer.
ƒƒ The lifetime risk of endometrial cancer is 40–60% in females with Lynch syndrome. Therefore, screening for endometrial cancer is
recommended for these patients and their relatives. The Amsterdam II Criteria are used for diagnosis of Lynch syndrome, but some
cases of Lynch syndrome-related endometrial cancer do not meet these criteria. Microsatellite instability and genetic analyses of
mismatch repair genes are useful in these cases.
ƒƒ Endometrial cancer that specifically occurs in the lower uterine body is referred to as carcinoma of the lower uterine segment and has a
hereditary predisposition. Carcinoma of the lower uterine segment is related to Lynch syndrome; therefore, it is a characteristic
phenotype of endometrial cancer that occurs in patients with Lynch syndrome.
ƒƒ Elucidation of disease-related genes in hereditary endometrial cancer and understanding of the carcinogenic mechanism will contribute
to early detection and prevention of endometrial cancer.

References 3 Muraki Y, Banno K, Yanokura M et al. sprouty 2 in endometrial carcinoma. Hum.


Papers of special note have been highlighted as: Epigenetic DNA hypermethylation: clinical Pathol. 42 (2), 185–193 (2011).
n of interest applications in endometrial cancer (Review). 5 Pallares J, Velasco A, Eritja N et al. Promoter
nn of considerable interest Oncol. Rep. 22(5), 967–972 (2009). hypermethylation and reduced expression of
1 Esteller M. Epigenetics in cancer. N. Engl. nn Recent comprehensive review of RASSF1A are frequent molecular alterations
J. Med. 358(11), 1148–1159 (2008). endometrial cancer epigenetic DNA of endometrial carcinoma. Mod. Pathol.
hypermethylation. 21(6), 691–699 (2008).
2 Wajed SA, Laird PW, DeMeester TR. DNA
methylation: an alternative pathway to cancer. 4 Velasco A, Pallares J, Santacana M et al. 6 Kang HS, Baba T, Mandai M et al. GPR54 is
Ann. Surg. 234 (1), 10–20 (2001). Promoter hypermethylation and expression of a target for suppression of metastasis in

158 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
endometrial cancer. Mol. Cancer Ther. 10 (4), cancer with microsatellite instability. Int. 31 Nakano H, Miyazawa T, Kinoshita K,
580–590 (2011). J. Oncol. 17(3), 535–541 (2000). Yamada Y, Yoshida T. Functional screening
7 Yi TZ, Guo J, Zhou L et al. Prognostic value 20 Banno K, Yanokura M, Kawaguchi M et al. identifies a microRNA, miR‑491 that induces
of E‑cadherin expression and CDH1 promoter Epigenetic inactivation of the CHFR gene in apoptosis by targeting Bcl‑X(L) in colorectal
methylation in patients with endometrial cervical cancer contributes to sensitivity to cancer cells. Int. J. Cancer 127(5), 1072–1080
carcinoma. Cancer Invest. 29(1), 86–92 taxanes. Int. J. Oncol. 31(4), 713–720 (2010).
(2011). (2007). 32 Huang YW, Liu JC, Deatherage DE et al.
8 Dewdney SB, Rimel BJ, Thaker PH et al. 21 Suter CM, Martin DI, Ward RL. Germline Epigenetic repression of microRNA-129-2
Aberrant methylation of the X‑linked epimutation of MLH1 in individuals with leads to overexpression of SOX4 oncogene in
ribosomal S6 kinase RPS6KA6 (RSK4) in multiple cancers. Nat. Genet. 36(5), 497–501 endometrial cancer. Cancer Res. 69(23),
endometrial cancers. Clin. Cancer Res. 17(8), (2004). 9038–9046 (2009).
2120–2129 (2011). 22 Hitchins MP, Wong JJ, Suthers G et al. 33 Tsuruta T, Kozaki KI, Uesugi A et al.
9 Kawaguchi M, Banno K, Yanokura M et al. Inheritance of a cancer-associated MLH1 miR‑152 is a tumor suppressor microRNA
Analysis of candidate target genes for germ-line epimutation. N. Engl. J. Med. that is silenced by DNA hypermethylation in
mononucleotide repeat mutation in 356(7), 697–705 (2007). endometrial cancer. Cancer Res. 71(20),
microsatellite instability-high (MSI‑H) 6450–6462 (2011).
23 Goel A, Nguyen TP, Leung HC et al. De novo
endometrial cancer. Int. J. Oncol. 35(5), constitutional MLH1 epimutations confer n Striking report demonstrating the tumor
977–982 (2009). early-onset colorectal cancer in two new suprressor effect of miR‑152 against
10 Ignatov A, Bischoff J, Ignatov T et al. APC sporadic Lynch syndrome cases, with endometrial cancer in vivo and in vitro.
promoter hypermethylation is an early event derivation of the epimutation on the paternal 34 Stumpel DJ, Schotte D, Lange-Turenhout EA
in endometrial tumorigenesis. Cancer Sci. allele in one. Int. J. Cancer 128(4), 869–878 et al. Hypermethylation of specific
101(2), 321–327 (2010). (2011). microRNA genes in MLL-rearranged infant
11 Satoh A, Toyota M, Itoh F et al. Epigenetic 24 Chan TL, Yuen ST, Kong CK et al. Heritable acute lymphoblastic leukemia: major matters
inactivation of CHFR and sensitivity to germline epimutation of MSH2 in a family at a micro scale. Leukemia 25(3), 429–439
microtubule inhibitors in gastric cancer. with hereditary nonpolyposis colorectal (2011).
Cancer Res. 63(24), 8606–8613 (2003). cancer. Nat. Genet. 38(10), 1178–1183 35 Chen Y, Song Y, Wang Z et al. Altered
12 Wang X, Yang Y, Xu C et al. CHFR (2006). expression of miR‑148a and miR‑152 in
suppression by hypermethylation sensitizes n First report of epimutation of the hMSH2 gastrointestinal cancers and its clinical
endometrial cancer cells to paclitaxel. Int. gene in Lynch syndrome. significance. J. Gastrointest. Surg. 14(7),
J. Gynecol. Cancer 21(6), 996–1003 (2011). 1170–1179 (2010).
25 van der Gun BT, Melchers LJ, Ruiters MH,
13 Lo TL, Yusoff P, Fong CW et al. The ras/ de Leij LF, McLaughlin PM, Rots MG. 36 Braconi C, Huang N, Patel T. MicroRNA-
mitogen-activated protein kinase pathway EpCAM in carcinogenesis: the good, the bad dependent regulation of DNA
inhibitor and likely tumor suppressor or the ugly. Carcinogenesis 31(11), 1913–1921 methyltransferase‑1 and tumor suppressor
proteins, sprouty 1 and sprouty 2 are (2010). gene expression by interleukin-6 in human
deregulated in breast cancer. Cancer Res. malignant cholangiocytes. Hepatology 51(3),
26 Ligtenberg MJ, Kuiper RP, Chan TL et al.
64(17), 6127–6136 (2004). 881–890 (2010).
Heritable somatic methylation and
14 Sutterluty H, Mayer CE, Setinek U et al. inactivation of MSH2 in families with Lynch 37 Chen HZ, Tsai SY, Leone G. Emerging roles
Down-regulation of Sprouty2 in non-small syndrome due to deletion of the 3´ exons of of E2Fs in cancer: an exit from cell cycle
cell lung cancer contributes to tumor TACSTD1. Nat. Genet. 41(1), 112–117 control. Nat. Rev. Cancer 9(11), 785–797
malignancy via extracellular signal-regulated (2009). (2009).
kinase pathway-dependent and -independent 38 Trusolino L, Bertotti A, Comoglio PM. MET
27 Kozaki K, Imoto I, Mogi S, Omura K,
mechanisms. Mol. Cancer Res. 5(5), 509–520 signalling: principles and functions in
Inazawa J. Exploration of tumor-suppressive
(2007). development, organ regeneration and cancer.
microRNAs silenced by DNA
15 Fong CW, Chua MS, McKie AB et al. hypermethylation in oral cancer. Cancer Res. Nat. Rev. Mol. Cell Biol. 11(12), 834–848
Sprouty 2, an inhibitor of mitogen-activated 68(7), 2094–2105 (2008). (2010).
protein kinase signaling, is down-regulated in 39 Roulin D, Cerantola Y, Dormond-Meuwly A,
28 Furuta M, Kozaki KI, Tanaka S, Arii S,
hepatocellular carcinoma. Cancer Res. 66(4), Demartines N, Dormond O. Targeting
Imoto I, Inazawa J. miR‑124 and miR‑203
2048–2058 (2006). mTORC2 inhibits colon cancer cell
are epigenetically silenced tumor-suppressive
16 Holliday R. The inheritance of epigenetic microRNAs in hepatocellular carcinoma. proliferation in vitro and tumor formation
defects. Science 238(4824), 163–170 (1987). Carcinogenesis 31(5), 766–776 (2010). in vivo. Mol. Cancer 9, 57 (2010).
17 Schofield PN, Joyce JA, Lam WK et al. 29 Pierson J, Hostager B, Fan R, Vibhakar R. 40 Guertin DA, Stevens DM, Saitoh M et al.
Genomic imprinting and cancer; new Regulation of cyclin dependent kinase 6 by mTOR complex 2 is required for the
paradigms in the genetics of neoplasia. microRNA 124 in medulloblastoma. development of prostate cancer induced by
Toxicol. Lett. 120(1–3), 151–160 (2001). J. Neurooncol. 90(1), 1–7 (2008). Pten loss in mice. Cancer Cell 15(2), 148–159
(2009).
18 Peltomäki P. Lynch syndrome genes. Fam. 30 Saito Y, Friedman JM, Chihara Y, Egger G,
Cancer 4(3), 227–232 (2005). Chuang JC, Liang G. Epigenetic therapy 41 Dakalakis M, Nguyen TT, Nguen C et al.
upregulates the tumor suppressor Demethylation of a hypermethylated P15/
19 Kondo E, Furukawa T, Yoshinaga K et al.
microRNA‑126 and its host gene EGFL7 in INK4B gene in patients with myelodysplastic
Not hMSH2 but hMLH1 is frequently
human cancer cells. Biochem. Biophys. Res. syndrome by 5‑Aza-2´‑deoxy cytidine
silenced by hypermethylation in endometrial
Commun. 379(3), 726–731 (2009). (decitabine) treatment. Blood 100(8),
cancer but rarely silenced in pancreatic
2957–2964 (2002).

future science group www.futuremedicine.com 159


Review Banno, Kisu, Yanokura et al.

42 Issa JP, Garcia-Manero G, Giles FJ. Phase 1 carcinoma cells: histone deacetylase 65 Takai N, Ueda T, Nishida M, Nasu, K,
study of low-dose prolonged exposure inhibitors exhibit antiproliferative activity Narahara H. A novel histone deacetylase
schedules of the hypomethylating agent and potently induce apoptosis. Cancer inhibitor, Scriptaid, induces growth
5‑aza-2´‑deoxycytidine(decitabine) in 101(12), 2760–2770 (2004). inhibition, cell cycle arrest and apoptosis in
hematopoietic malignancies. Blood 103(5), 55 Gui CY, Ngo L, Xu WS, Richon VM, Marks human endometrial cancer and ovarian
1635–1640 (2004). PA. Histone deacetylase (HDAC) inhibitor cancer cells. Int. J. Mol. Med. 17(2), 323–329
43 Satoh A, Toyota M, Itoh F. Epigenetic activation of p21WAF1 involves changes in (2006).
inactivation of CHFR and sensitivity to promoter-associated proteins, including 66 Terao Y, Nishida J, Horiuchi S et al. Sodium
microtubule inhibitors in gastric cancer. HDAC1. Proc. Natl Acad. Sci. USA 101(5), butyrate induces growth arrest and
Cancer Res. 63(24), 8606–8613 (2003). 1241–1246 (2004). senescence-like phenotypes in gynecologic
44 Scolnick DM, Halazonetis TD. Chfr defines 56 Marchion D, Munster P. Development of cancer cells. Int. J. Cancer 94(2), 257–267
amitotic stress checkpoint that delays entry histone deacetylase inhibitors for cancer (2001).
into metaphase. Nature 406(6794), 430–435 treatment. Expert Rev. Anticancer Ther. 7(4), 67 Jiang S, Dowdy SC, Meng XW et al. Histone
(2000). 583–598 (2007). deacetylase inhibitors induce apoptosis in
45 Kang D, Chen J, Wong J. The checkpoint 57 Takai N, Narahara H. Human endometrial both type I and type II endometrial cancer
protein Chfr is a ligase that ubiquitinates Plk1 and ovarian cancer cells: histone deacetylase cells. Gynecol. Oncol. 105(2), 493–500
and inhibits Cdc2 at the G2 to M transition. inhibitors exhibit antiproliferative activity, (2007).
J. Cell Biol. 156(2), 249–259 (2002). potently induce cell cycle arrest, and stimulate 68 Jung M, Brosch G, Kölle D, Scherf H,
46 Yu X, Minter-Dykhouse K, Malureanu L. apoptosis. Curr. Med. Chem. 14(24), Gerhäuser C, Loidl P. Amide analogues of
Chfr is required for tumor suppression and 2548–2553 (2007). trichostatin A as inhibitors of histone
Aurora A regulation. Nat. Genet. 37(4), n Recent review highlighting the histone deacetylase and inducers of terminal cell
401–406 (2005). differentiation. J. Med. Chem. 42(22),
deacetylase inhibitors to endometrial cancer.
4669–4679 (1999).
47 Toyota M, Sasaki Y, Satoh A. Epigenetic 58 Uchida H, Maruyama H, Ngashima T, Asada
inactivation of CHFR in human tumors. 69 Ueda T, Takai N, Nishida M, Nasu K,
H, Yoshimura Y. Histone deacetylase
Proc. Natl Acad. Sci. USA 100(13), Narahara H. Apicidin, a novel histone
inhibitors induce differentiation of human
7818–7823 (2003). deacetylase inhibitor, has profound
endometrial adenocarcinoma cells through
anti-growth activity in human endometrial
n Highly significant finding about the CHFR up-regulation of glycoderin. Endocrinology
and ovarian cancer cells. Int. J. Mol. Med.
gene, relevant to the biomarker for 146(129), 5365–5373 (2005).
19(2), 301–308 (2007).
chemosinsitivity to taxane. 59 Sandor V, Robbins AR, Robey R, Myers T.
70 Vasen HF, Watson P, Mecklin JP, Lynch HT.
48 Ogi K, Toyota M, Mita H. Small interfering
FR901228 causes mitotic arrest but does not
New clinical criteria for hereditary
RNA-induced CHFR silencing sinsitizes oral alter microtubule polymerization. Anticancer
nonpolyposis colorectal cancer (HNPCC,
squamous cell cancer cells to microtubule Drugs 11(6), 445–454 (2011).
Lynch syndrome) proposed by the
inhibitors. Cancer Biol. Ther. 4(7), 773–780 60 Dowdy SC, Jiang S, Zhou XC et al. Histone International Collaborative group on
(2005). deacetylase inhibitors and paclitaxel cause HNPCC. Gastroenterology 116(6), 1453–1456
49 Koga Y, Kitajima Y, Miyoshi A. The
synergistic effects on apoptosis and (1999).
significance of aberrant CHFR methylation microtubule stabilization in papillary serous
endometrial cancer cells. Mol. Cancer Ther.
n First report of the revised clinical criteria for
for clinical response to microtubule inhibitors Lynch syndrome.
in gastric cancer. J. Gastroenterol. 41(2), 5(11), 2767–2776 (2006).
61 Camacho LH, Olson J, Tong WP, Young 71 Boland CR, Shike M. Report from the
133–139 (2006).
CW, Spriggs DR, Malkin MG. Phase I dose Jerusalem workshop on Lynch syndrome-
50 Jenuwein T, Allis CD. Translating the
escalation clinical trial of phenylbutyrate hereditary nonpolyposis colorectal cancer.
histone code. Science 293(5532), 1074–1080 Gastroenterology 138(7), 2197.e1–2197.e7
(2001). sodium administered twice daily to patients
with advanced solid tumors. Invest. New (2010).
51 Bhalla KN. Epigenetic and chromatin
Drugs 25(2), 131–138 (2007). 72 Hampel H, Panescu J, Lockman J et al.
modifiers as targeted therapy of hematologic Comment on: screening for Lynch syndrome
62 Modesitt SC, Sill M, Hoffman JS, Bender
malignancies. J. Clin. Oncol. 23(17), (hereditary nonpolyposis colorectal cancer)
3971–3993 (2005). DP. A Phase II study of vorinostat in the
treatment of persistent or recurrent epithelial among endometrial cancer patients. Cancer
52 Krusche CA, Vloet AJ, Classen-Linke I, von Res. 67(19), 9603 (2007).
ovarian or primary peritoneal carcinoma:
Rango U, Beier HM, Alfer J. Class I histone a Gynecologic Oncology Group study. 73 Meyer LA, Broaddus RR, Lu KH.
deacetylase expression in the human cyclic Gynecol. Oncol. 109(2), 182–186 (2008). Endometrial cancer and Lynch syndrome:
endometrium and endometrial clinical and pathologic considerations. Cancer
63 Takai N, Desmond JC, Kumagai T et al.
adenocarcinomas. Hum. Reprod. 22(11), Control 16(1), 14–22 (2009).
2956–2966 (2007). Histone deacetylase inhibitors have a
profound antigrowth activity in endometrial 74 Dunlop MG, Farrington SM, Carothers AD
53 Weichert W, Denkert C, Noske A et al.
cancer cells. Clin. Cancer Res. 10(3), et al. Cancer risk associated with germline
Expression of class I histone deacetylases 1141–1149 (2004). DNA mismatch repair gene mutations. Hum.
indicates poor prognosis in endometrioid Mol. Genet. 6(1), 105–110 (1997).
64 Takai N, Ueda T, Nishida M et al. CBHA is a
subtypes of ovarian and endometrial
carcinomas. Neoplasia 10(9), 1021–1027 family of hybrid polar compounds that inhibit 75 Vasen HF, Wijnen JT, Menko FH et al.

(2008). histone deacetylase, and induces growth Cancer risk in families with hereditary
inhibition, cell cycle arrest and apoptosis in nonpolyposis colorectal cancer diagnosed by
54 Takai N, Kawamata N, Gui D, Said JW,
human endometrial and ovarian cancer cells. mutation ana­lysis. Gastroenterology 110(4),
Miyakawa I, Koeffler HP. Human ovarian Oncology 70(2), 97–105 (2006). 1020–1027 (1996).

160 Epigenomics (2012) 4(2) future science group


Epigenetics & genetics in endometrial cancer Review
76 Peltomaki P, Vasen H. Mutations associated colorectal cancer. Cancer Res. 58(22), genotype–phenotype correlations. Eur.
with HNPCC predisposition – update of 5248–5257 (1998). J. Hum. Genet. 7(3), 267–273 (1997).
ICG-HNPCC/INSiGHT mutation database. 87 Esteller M, Levine R, Baylin SB, Ellenson 98 Nelen MR, Padberg GW, Peeters EA et al.
Dis. Markers 20(4–5), 269–276 (2004). LH, Herman JG. MLH1 promoter Localization of the gene for Cowden disease
77 Schweizer P, Moisio AL, Kuismanen SA et al. hypermethylation is associated with the to chromosome 10q22–23. Nat. Genet. 13(1),
Lack of MSH2 and MSH6 characterizes microsatellite instability phenotype in 114–116 (1996).
endometrial but not colon carcinomas in sporadic endometrial carcinomas. Oncogene 99 Liaw D, Marsh DJ, Li J et al. Germline
hereditary nonpolyposis colorectal cancer. 17(18), 2413–2417 (1998). mutations of the PTEN gene in Cowden
Cancer Res. 61(7), 2813–2815 (2001). 88 Resnick KE, Hampel H, Fishel R, Cohn DE. disease, an inherited breast and thyroid cancer
78 Banno K, Susumu N, Hirao T et al. Current and emerging trends in Lynch syndrome. Nat. Genet. 16(1), 64–67 (1997).
Identification of germline MSH2 gene syndrome identification in women with 100 Salem OS, Steck WD. Cowden’s disease
mutations in endometrial cancer not endometrial cancer. Gynecol. Oncol. 114(1), (multiple hamartoma and neoplasia
fulfilling the new clinical criteria for 128–134 (2009). syndrome). A case report and review of the
hereditary nonpolyposis coloretcal cancer. 89 Lindor NM, Petersen GM, Hadley DW et al. English literature. J. Am. Acad. Dermatol.
Cancer Genet. Cytogenet. 146(1), 58–65 Recommendations for the care of individuals 8(5), 686–696 (1983).
(2003). with an inherited predisposition to Lynch 101 Higuchi M, Onishi K, Kikuchi C, Gotoh Y.
79 Broaddus RR, Lynch HT, Chen LM et al. syndrome: a systematic review. JAMA Scaffolding function of PAK in the
Pathologic features of endometrial carcinoma 296(12), 1507–1517 (2006). PDK1‑Akt pathway. Nat. Cell Biol. 10(1),
associated with HNPCC: a comparison with 90 Vasen HF, Moslein G, Alonso A et al. 1356–1364 (2008).
sporadic endometrial carcinoma. Cancer Guidelines for the clinical management of 102 Black D, Bogomolniy F, Robson ME, Pffit K,
106(1), 87–94 (2006). Lynch syndrome (hereditary non-polyposis Barakat RR, Boyd J. Evaluation of germline
80 Vasen HF, Watson P, Mecklin JP et al. The cancer). J. Med. Genet. 44(6), 353–362 PTEN mutations in endometrial cancer
epidemiology of endometrial cancer in (2007). patients. Gynecol. Oncol. 96(1), 1–24 (2005).
hereditary nonpolyposis colorectal cancer. 91 Ollikainen M, Abdel-Rahman WM, Moisio 103 Stambolic V, Tsao MS, Macpherson D,
Anticancer Res. 14(4B), 1675–1678 (1994). AL et al. Molecular ana­lysis of familial Suzuki A, Chapman WB, Mak TW. High
81 Boks DE, Trujillo AP, Voogd AC, Morreau endometrial carcinoma: a manifestation of incidence of breast and endometrial neoplasia
H, Kenter GG, Vasen HF. Survival ana­lysis hereditary nonpolyposis colorectal cancer or a resembling human Cowden syndrome in
of endometrial carcinoma associated with separate syndrome? J. Clin. Oncol. 23(21), PTEN +/- mice. Cancer Res. 60(13),
hereditary nonpolyposis colorectal cancer. 4609–4616 (2005). 3605–3611 (2000).
Int. J. Cancer 102(2), 198–200 (2002). 92 Jacques SM, Qureshi F, Ramirez NC, 104 Sardinoux M, Raingeard I, Bessis D, Coupier
82 Hampel H, Frankel W, Panescu J et al. Malviya VK, Lawrence WD. Tumors of the I, Renard E, Bringer J. Cowden syndrome, or
Screening for Lynch syndrome (hereditary uterine isthmus: Clinicopathologic features multiple hamartomatous tumor syndrome, in
nonpolyposis colorectal cancer) among and immunohistochemical characterization of clinical endocrinology. Ann. Endocrinol.
endometrial cancer patients. Cancer Res. p53 expression and hormone receptors. Int. 71(4), 264–273 (2010).
66(15), 7810–7817 (2006). J. Gynecol. Pathol. 16(1), 38–44 (1997).
105 Farooq A, Walker LJ, Bowling J, Audisio RA.
83 Hirai Y, Banno K, Suzuki M et al. Molecular 93 Westin S, Lacour R, Urbauer D et al. Cowden syndrome. Cancer Treat. Rev. 36(8),
epidemiological and mutational ana­lysis of Carcinoma of the lower uterine segment: 577–583 (2010).
DNA mismatch repair (MMR) genes in a newly described association with Lynch
106 Gustafson S, Zbuk KM, Scacheri C, Eng C.
endometrial cancer patients with HNPCC- syndrome. J. Clin. Oncol. 26(36), 5965–5971
Cowden syndrome. Semin. Oncol. 34(5),
associated familial predisposition to cancer. (2008).
428–434 (2007).
Cancer Sci. 99(9), 1715–1719 (2008). n Striking report demonstrating the
107 Schmeler KM, Daniels MS, Brandt AC, Lu
nn Notable report demonstrating the association between cancer of the lower
KH. Endometrial cancer in an adolescent:
responsibility of hMSH6 in Lynch uterine segment and Lynch syndrome.
a possible manifestation of Cowden syndrome.
syndrome-associated endometrial cancer. 94 Masuda K, Banno K, Yanokura M et al. Obstet. Gynecol. 114(2), 477–479 (2009).
84 Banno K, Yanokura M, Kobayashi Y et al. Carcinoma of the lower uterine segment
108 Beggs AD, Latchford AR, Vasen HF et al.
Endometrial cancer as a familial tumor: (LUS): clinicopathological characteristics and
Peutz–Jeghers syndrome: a systematic review
pathology and molecular carcinogenesis association with lynch syndrome. Curr.
and recommendations for management. Gut
(review). Curr. Genomics 10(2), 127–132 Genomics 12(1), 25–29 (2011).
59(7), 975–986 (2010).
(2009). 95 Hachisuga T, Kaku T, Enjoji M. Carcinoma
109 Scully RE. Sex cord tumor with annular
85 Lancaster JM, Powell CB, Kauff ND et al. of the lower uterine segment.
tubules a distinctive ovarian tumor of the
Society of Gynecologic Oncologists Clinicopathologic ana­lysis of 12 cases. Int.
Peutz–Jeghers syndrome. Cancer 25(5),
Education Committee statement on risk J. Gynecol. Pathol. 8(1), 26–35 (1989).
1107–1121 (1970).
assessment for inherited gynecologic cancer 96 Watanabe Y, Nakajima H, Nozaki K et al.
predispositions. Gynecol. Oncol. 107(2), 110 Giardiello FM, Brensinger JD, Tersmette AC
Clinicopathologic and immunohistochemical
159–162 (2007). et al. Very high risk of cancer in familial
features and microsatellite status of
Peutz–Jeghers syndrome. Gastroenterology
86 Boland CR, Thibodeau SN, Hamilton SR endometrial cancer of the uterine isthmus. Int.
119(6), 1447–1453 (2000).
et al. A National Cancer Institute Workshop J. Gynecol. Pathol. 20(4), 368–373 (2001).
on Microsatellite Instability for cancer 111 Conteras CM, Gurumurthy S, Haynie JM
97 Nelen MR, Kremer H, Konings IB et al.
detection and familial predisposition: et al. Loss of Lkb1 provokes highly invasive
Novel PTEN mutations in patients with
development of international criteria for the endometrial adenocarcinomas. Cancer Res.
Cowden disease: absence of clear
determination of microsatellite instability in 68(3), 759–766 (2008).

future science group www.futuremedicine.com 161


Review Banno, Kisu, Yanokura et al.

112 Kondi-Pafiti A, Bakalianou K, Iavazzo C, signatures of tobacco exposure. Nature genome projects. Nature 464(7291) 993–998
Dastamani C, Hasiakos D, Liapis A. 463(7278), 184–190 (2009). (2010).
Endometrial carcinoma and ovarian sex cord 115 Cancer Genome Atlas Research Network. 118 Takai N, Narahara H. Array-based approach
tumor with annular tubules in a patients with comprehensive genomic characterization for the identification of epigenetic silenced
history of Peutz-Jegers syndrome and multiple defines human glioblastoma genes and core tumor suppressor genes. Curr. Genomics 9(1),
malignancies. Eur. J. Gynaecol. Oncol. 32(49), pathways. Nature 455(7216), 1061–1068 22–24 (2008).
452–454 (2011). (2008).
113 Pleasance ED, Cheetham RK, Stephense PJ 116 Cancer Genome Atlas Research Network.
et al. A comprehensive catalogue of somatic Integrated genomic ana­lysis of ovarian „„Websites
mutations from a human cancer genome. carcinoma. Nature 474(7353), 609–615 201 The Cancer Genome Atlas.
Nature 463(7278), 191–196 (2009). (2011) http://cancergenome.nih.gov
114 Pleasance ED, Stephens PJ, O’Meara S et al. A 117 The International Cancer Genome 202 International Cancer Genome Consortium.
small-cell lung cancer genome with complex Consortium. International network of cancer www.icgc.org

162 Epigenomics (2012) 4(2) future science group


Reproduced with permission of the copyright owner. Further reproduction prohibited without permission.

View publication stats

You might also like