Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

VOLUME 24 䡠 NUMBER 29 䡠 OCTOBER 10 2006

JOURNAL OF CLINICAL ONCOLOGY B I O L O G Y O F N E O P L A S I A

Molecular and Pathologic Aspects of


Endometrial Carcinogenesis
Jonathan L. Hecht and George L. Mutter
From the Department of Pathology,
A B S T R A C T
Beth Israel Hospital; Department of
Pathology, Brigham and Women’s Endometrial cancer is the most common gynecological malignancy, with 41,000 new cases projected in the
Hospital, Boston, MA. United States for 2006. Two different clinicopathologic subtypes are recognized: the estrogen-related (type I,
Submitted March 21, 2006; accepted endometrioid) and the non– estrogen-related types (type II, nonendometrioid such as papillary serous and clear
June 12, 2006. cell). The morphologic differences in these cancers are mirrored in their molecular genetic profile with type I
Terms in blue are defined in the glossary,
showing defects in DNA-mismatch repair and mutations in PTEN, K-ras, and beta-catenin, and type II showing
found at the end of this article and online
aneuploidy and p53 mutations. This article reviews the genetic aspects of endometrial carcinogenesis and
at www.jco.org. progression. We will define the precursor lesion of type I endometrioid cancer and the role of genetics and
estrogen in its progression.
Authors’ disclosures of potential con-
flicts of interest and author contribu-
J Clin Oncol 24:4783-4791. © 2006 by American Society of Clinical Oncology
tions are found at the end of this
article.

Address reprint requests to Department In the last decade, progress has been greatest in
INTRODUCTION
of Pathology, Beth-Israel Deaconess molecular and histologic resolution of precursors of
Medical Center, 330 Brookline Ave,
Boston, MA 02215; e-mail: jlhecht@ Current concepts of endometrial cancer successfully type I cancer, resulting in a cohesive model of endo-
BIDMC.harvard.edu integrate traditional histopathology with pathoge- metrial carcinogenesis encompassing both genetic
© 2006 by American Society of Clinical netic mechanisms. Endometrial cancers have long and hormonal factors, revised precancer diagnostic
Oncology been classified into two major divisions (types I and criteria, and novel prevention strategies. We will pri-
0732-183X/06/2429-4783/$20.00 II) based on light microscopic appearance, clinical marily focus on these advances and will describe
DOI: 10.1200/JCO.2006.06.7173 behavior, and epidemiology.1 Type I, those with ongoing attempts at genetic substratification within
the endometrioid group.
endometrioid histology, comprise 70% to 80% of
newly diagnosed cases of endometrial cancer in the
United States.2 They are associated with unopposed GENETICS OF TYPE I CANCERS
estrogen exposure and are often preceded by pre-
malignant disease. In contrast, type II endometrial
While most serous (type II) cancers contain muta-
cancers have nonendometrioid histology (usually tions of p53,3 endometrioid (type I) adenocarcino-
papillary serous or clear cell) with an aggressive clin- mas demonstrate larger numbers of genetic changes
ical course. Hormonal risk factors have not been in which the temporal sequence of mutation, and
identified, and there is no readily observed prema- the final combination of defects differ substantially
lignant phase. The morphologic and clinical differ- between individual examples. Common genetic
ences are paralleled by genetic distinctions, in that changes in endometrioid endometrial cancers in-
type I and II cancers carry mutations of independent clude, but are not limited to, microsatellite instabil-
sets of genes (Table 13-8). Most were identified as ity (MSI),11-15 or specific mutation of PTEN,16-21
candidate genes by analogy with other tumor sys- K-ras,12,22-28 and ␤-catenin genes.13,29-31
tems, and were confirmed subsequently to be altered
MSI
in endometrial cancer. Open-ended gene discovery
Approximately 20% of sporadic endometrioid
by comparison of tumor subsets using genome-wide endometrial cancers of all grades demonstrate a mo-
methods such as expression profiling has further lecular phenotype referred to as MSI.11,12,15,32 Mic-
broadened our understanding of relevant genetic rosatellites are short segments of repetitive DNA
basis for these differences.9,10 The result is an en- bases that are scattered throughout the genome; they
hanced understanding of early genetic events, rein- are found predominantly in noncoding DNA. MSI
forcement of the clinicopathologic subgroups is the propensity to develop changes in the number
originally defined by histologic and clinical features, of repeat elements as compared with normal tissue
and development of biomarkers informative in due to DNA repair errors made during replication.
identification of previously unknown or poorly de- MSI is rare (⬍ 5%) in type II endometrial can-
scribed precursor lesions. cers,33,34 where the primary genetic defect is in the

4783
Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Hecht and Mutter

Table 1. Genetic Differences Between Cancers With and Without Endometrioid Histology
Endometrioid Nonendometrioid
Gene Alteration (type 1; %) (type 2; %) Reference

p53 Inactivating mutation 5-10 80-90 3, 8


PTEN Loss of function through deletion 55 11 6
and/or mutation
K-ras Mutational activation 13-26 0-10 3, 7
␤-catenin (CTNNB1) Gain of function mutation 25-38 Rare 7
MLH1 Epigenetic silencing causes 17 5 4, 5
microsatellite instability

p53 gene and genetic instability is manifest globally at the chromo- nism.49,50 The most commonly observed PTEN defect is inactivation
somal rather than microsatellite level. Thus, type II tumors frequently of both alleles to generate a protein null, or complete loss of function,
demonstrate high-order aneuploidy while having an intact mismatch- phenotype. There is some evidence that even a PTEN hemizygous
repair mechanism.34,35 inactivation leading to a protein deficient, rather than null state, may
MSI is due to inactivation of any of a number of intranuclear be functionally significant when combined with abnormalities of
proteins that comprise the mismatch repair system, leading to accu- other genes which converge on its downstream pathway. PTEN acts in
mulation of structural changes in coding and noncoding repetitive opposition to phosphotidylinositol-3-kinase (PIK3CA) to control lev-
elements of many genes.36 MLH1 inactivation, a component of the els of phosphorylated AKT. PIK3CA mutation, seen in 36% of endo-
mismatch repair system, is the most common mechanism in the metrial carcinomas, is most frequent in those tumors which also bear
endometrium and is accomplished by hypermethylation of CpG is- PTEN mutation.51 This is consistent with a cooperative effect between
lands in the gene promoter, a process known as epigenetic silencing.5 these two elements in promoting neoplastic transformation.
This mechanism stands in contrast to colon cancer, where MSI is due PTEN inactivation is a functionally significant in carcinogenesis
to mutations in the MSH2, MLH1, and MSH6 genes.37 Inherited or
as is demonstrated by the high prevalence of endometrial cancer in
somatically acquired mutations of MSH6, another mismatch repair
PTEN knockout mice. Heterozygous PTEN inactivation produces an
element, are also common in patients with MSI endometrial can-
abnormal endometrial phenotype in mice, with 100% of mice develop-
cers.34 A single nucleotide insertion (frameshift) mutation in MSH3
ing hyperplastic lesions, and 20% of animals progressing to endometrial
has been described less frequently in MSI endometrial cancers with
MSI. This deletion occurs in a string of eight consecutive adenosine carcinoma.52 Humans with constitutive germline PTEN mutations may
residues (A8). Because simple repeat sequences are unstable in cells present with the heritable cancer syndrome of Cowden’s disease, which
with MSI, the observed mutation may be secondary to the MSI derived includes high rates of breast, thyroid, and other cancers in conjunction
from defective MLH1 expression.38,39 Since MLH1 is important in with hamartomas of multiple organs.6 There appears to be an in-
repair of short segments (two to four bases), and MSH2:MSH3 serves creased risk of endometrial cancer in women with Cowden’s disease,
to repair larger insertion-deletion mutations, the combined defect of but only small numbers of these patients have been available for study,
the mismatch repair system results in inhibition of both small and and the magnitude of increased risk is modest.
large insertion-deletion mismatch repair.40 PTEN inactivation may be caused by a variety of mechanisms.
MSI, and abnormal methylation of MLH1, is an early event in Mutation, or deletions resulting in loss of heterozygosity (LOH) at
endometrial carcinogenesis that has been described in precancerous chromosome 10q23, are detected in 37% to 61% of cancers.6,53 The
lesions.11,18,41 Once established within a tumor lineage, MSI leads to pattern of PTEN mutation is different in MSI and microsatellite stable
creation of genetic heterogeneity within carcinoma cells of a single cancers. MSI-positive tumors have a higher frequency of deletions
tumor. MSI may specifically target for inactivation those genes which involving ⱖ three base pairs when compared with the MSI-negative
contain susceptible repeat elements, such as transforming growth group. In addition, the mutations observed in MSI tumors only rarely
factor ␤receptor type II, (TGF-␤RII), BAX, insulin-like growth factor involve the polyadenine repeat of exon 8 that is the expected target
II receptor (IGFIIR), and hMSH3, resulting in secondary tumor sub- (containing a microsatellite marker). The mechanism for these MSI
clones with an altered capacity to invade and metastasize.42-46 Indeed, related changes is unknown.40 Promoter methylation has been postu-
screening of MSI endometrial cancers has shown frameshift muta- lated an alternative transcriptional inactivating event, but has been
tions in the coding region repeats of FAS, BAX, CASP5, and IGF-␤RII difficult to prove experimentally because of technical limitations in
genes that are infrequent in microsatellite stable cancers.47,48 quantification of methylation level and assay interference by a co-
PTEN amplified pseudogene.
Inactivation of the PTEN tumor-suppressor gene (formerly The PTEN locus on chromosome 10 may be flanked by other
known as MMAC1) is the most common genetic defect in endometri- tumor suppressor genes that are codeleted with PTEN itself.54,55
oid carcinoma and is seen in up to 83% of tumors that are preceded by The homeodomain containing gene EMX2 is a strong candidate for
a histologically discrete premalignant phase.16 PTEN is a tumor sup- just such a gene.56 Increased native EMX2 expression is associated
pressor gene encoding a lipid phosphatase which acts to maintain G1 with diminished endometrial proliferative activity, a pattern that
arrest and enable apoptosis through an AKT-dependent mecha- might be predicted for a tumor suppressor. Deletion of EMX2,

4784 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Molecular Biology of Endometrial Cancer

with commensurate decline in expression, is seen in some endo- p53 is a nuclear phosphoprotein that induces proliferative arrest
metrial adenocarcinomas. or apoptosis through induction of P21Waf1/Cip1 and hMdm2 in
response to cellular stress. Although more common in serous and
K-ras clear-cell type carcinoma, aberrant accumulation of inactivated p53
K-ras mutations have been identified in 10% to 30% of type I protein is seen in approximately 5% of endometrioid cancers.27 High
endometrial cancers.26,27,57 There is a higher frequency of K-ras mu- protein levels correlate with high grade and stage, but are also an
tations in MSI cancers, and many of these are characterized as meth- independent prognostic factor.70 Unlike serous cancers, p53 trunca-
ylation related GC 3 AT transitions.28 tion mutations are rare in endometrioid tumors.71 In p53 “positive”
endometrioid endometrial tumors, p53 protein accumulation may be
␤-catenin (CTNNB1) secondary to changes in its upstream regulatory proteins rather than
Gain of function mutations in exon 3 of CTNNB1 gene at 3p21 the p53 gene itself.72 Several genes, including MDM2 and p14 ARF,
are seen in 25% to 38% of type I cancers.29-31 These mutations in exon that regulate p53 levels have been shown to cause detectable levels of
3 result in stabilization of the protein, cytoplasmic and nuclear accu- p53 in the absence of p53 mutation, and their dysregulation may be
mulation, and participation in signal transduction and transcriptional associated with adverse clinical outcomes.71-73 Alternatively, nonspe-
activation through the formation of complexes with DNA-binding cific DNA damage such as that induced by irradiation are also known
proteins.58 ␤-catenin is a component of the E-cadherin-catenin unit to induce accumulation of wild-type p53.74
essential for cell differentiation and maintenance of normal tissue Other candidate prognostic markers investigated in endometrial
architecture, and plays an important role in signal transduction. In- adenocarcinoma have differing degrees of evidence supporting clini-
creased nuclear levels of ␤-catenin produce transcriptional activation cal relevance. Many have only been described in in vitro models, and
through the LEF/Tcf pathway.59 The APC protein downregulates others fail to achieve a level of clinical outcome prediction indepen-
␤-catenin levels by cooperating with glycogen synthase kinase 3␤ dent of existing pathologic tumor classification. Expression of
(GSK-3␤), inducing phosphorylation of serine-threonine residues HOXB13, a member of the homeobox (HOX) gene family, is induced
coded in exon 3 of the ␤-catenin gene and its degradation through the by estrogens in endometrial cancer cell lines and confers invasive
ubiquitin-proteasome pathway.60,61 potential.75 High rates of inactivation by promoter methylation of
␤-catenin mutation may represent a pathway to endometrial another homeobox gene HOXA11 correlates with tumor recurrence
carcinogenesis characterized by squamous differentiation and inde- in stage I to II cancers.76 E-cadherin functions to maintain cell-to-cell
pendent of PTEN.60,61 Although MSI, PTEN, and K-ras mutations adhesion and its loss is has been associated with invasive capacity
frequently coexist with each other, these molecular abnormalities are in breast, esophagus and ovarian cancers.77 Inactivation of the E-
not usually seen in tumors with ␤-catenin alterations.58 When abnor- cadherin gene by methylation is present in a subset of cancers and with
mal, ␤-catenin expression changes are usually seen throughout all tumor increasing frequency at higher stage and grade. In addition to MSI,
cells, and ␤-catenin changes are present in some premalignant lesions. inactivation of MLH1 may create a selective growth advantage under
This suggests that ␤-catenin mutation is an early step of endometrial conditions of oxidative stress or cross-linking agents via the deregula-
tumorigenesisthatisclonallyrepresentedin all tumor cells.7,64 However, tion of apoptosis.78 Comparison of expression profiling of endome-
changes in ␤-catenin activity may contribute to later tumor progres- trial cancer cell lines before and after treatment with a demethylation
sion as well.65,66 Based on analogy with colon cancer, several genes agent have revealed tumor suppressor genes that undergo epigenetic
may be potential targets for a dysregulated WNT/␤-catenin pathway. silencing through promoter methylation.79 Reported genes include
For example, in colonic adenocarcinoma, elevated ␤-catenin levels Tazarotene-induced gene-1 (TIG1) and CCAAT/enhancer binding
caused by mutations in CTNNB1 or APC result triggers cyclin-D1 protein-alpha (C/EBPalpha).
gene expression, and subsequently, uncontrolled progression of tu-
mor cells into the cell cycle.67 ␤-catenin might regulate the expression
of the matrix metalloprotease-7, which would have a role in the estab- ROLE OF SEX HORMONES
lishment of the microenvironment necessary for the initiation and
maintenance of growth of the primary tumors and their metastasis.68 Estrogens and progestins act reciprocally on the hormonally respon-
Nuclear localization (activation) of ␤-catenin is also induced by sive endometrial tissue field to modify endometrial cancer risk. Pro-
the WNT signaling pathway, which is abnormal in some MSI tumors. gestins have the ability to abrogate, or “oppose” the biologic effects of
Gene expression profiling of cancers with and without MSI shows two coexisting estrogens through down-regulation of the estrogen recep-
members of the secreted frizzled related protein family (SFRP1 and tor itself. For this reason the biologic effects of admixtures of circulat-
SFRP4) as more often downgraded in MSI cancers.69 These proteins ing progestins and estrogens are dominated by the progestational
are negative regulators of the WNT signaling pathway. Fibroblast component. Women exposed to estrogens without opposing effects of
growth factor 18 (FGF18), a direct target of the WNT pathway, was progestins show a dose and duration dependant 2- to 10-fold in-
upregulated in MSI cancers. creased cancer risk.80-83 The protective effects of progestins are evident
in women using combined oral contraceptives, who have a 0.5 to 0.7
endometrial cancer risk relative to controls.84,85 There are several
OTHER GENES
postulated mechanisms by which sex hormones affect endometrial
cancer risk, and it is likely that all are relevant to varying degrees.
The phenotype and behavior of type I cancers are codetermined by Large-scale expression profiling of endometrial tissue RNAs has
cumulative genetic factors that can include mutations other than enabled comparison of hormonally induced transcriptional changes
mentioned above. in benign endometrium to those seen in carcinoma.86 The profile of

www.jco.org 4785
Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Hecht and Mutter

type 1 endometrioid carcinoma resembles the expression profile seen imparts invasive potential.75 Cables, a cyclin-dependent kinase bind-
in estrogen-driven proliferative endometrium and lacks expression of ing protein that is upregulated by progesterone and down-regulated
those genes induced by progestins. Overall, cancers are characterized by estrogen in benign endometrium, is lost in the majority of endo-
by a loss of gene relative to benign endometrium, accounting for eight metrial cancers. Mice deficient in cables develop hyperplasia and can-
of 10 discriminating changes in RNA abundance. Among the genes cer, and overexpression in cell lines slows proliferation.95
with reduced or lost expression in carcinoma are a small number of
tumor-suppressor genes affected by primary mutation or deletion and
MODEL FOR TYPE I CARCINOGENESIS
a larger number of downstream target genes affected secondarily.
Estrogen promotes cell proliferation and inhibits apoptosis
through a complex downstream cascade of transcriptional changes Type I cancers are frequently preceded by pan-endometrial hormonally
that may include modulation of tumor suppressor function. An ex- induced changes referred to as endometrial hyperplasia, from which a
ample is PTEN expression in normal endometrial glands, which is localized monoclonal population of genetically altered glands emerges
greatly elevated by estrogens and reduced by progestins during hor- as a discrete premalignant lesion, endometrial intraepithelial neopla-
monal fluctuations of the normal menstrual cycle.87 This expression sia (EIN). In the past, distinction between generalized hormonal field
pattern reflects the role of PTEN as regulator of mitosis and enabler of effects and actual premalignant lesions was obscured by imprecise
apoptosis in the estrogen stimulated proliferative phase. Unopposed diagnostic criteria and a nomenclature system that did not accurately
estrogens thus act as positive selection factor for PTEN mutant cells, by segregate lesions by natural history. The common term “hyperplasia”
unmasking the inability of mutant compared with wild-type cells to was applied to all entities, with the assumption that nonatypical hy-
check the proliferative process. In the presence of circulating proges- perplasia and atypical hyperplasia subgroups correspond to benign
tins, the proliferative advantage of PTEN mutant cells is lost, and as a and premalignant. Diagnosis of atypia is itself poorly reproducible96
result, PTEN mutant clones have a tendency to involute.88 The cancer and architectural features were underutilized as relevant discriminators
protective effects of progestins are further mediated by induction of between hormonal field effects and premalignant disease. Improved res-
apoptosis through the increased expression of Bcl-2 and BAX.89 The olution was possible with the advent of polymerase chain reaction–based
ability of progestins to induce endometrial epithelial apoptotic cell clonal assays and relevant biomarkers that facilitated a molecular, rather
death extinguishes after just a few days, but increases dramatically on than purely histopathologic, approach to precancer diagnosis. Careful
withdrawal of progestins.90 The net effect cannot be simply viewed as correlation of genetically ascertained premalignant disease with his-
simple opposition of estrogen and progesterone on a static gland topathologic presentation and clinical cancer outcomes launched the
population, but must also incorporate a dynamic element of changing molecular entity of EIN into a clinically relevant lesion that may be rou-
tissue responsiveness over time. Estrogens may also increase the rate of tinely diagnosed by pathologists and used for therapeutic intervention.
mutagenesis through free radical formation,91 but the magnitude of Somatic mutation of endometrial glandular cells is very common
this effect is minimal compared with the increased likelihood of ran- in normal endometrium, and occurs in advance of any discernable
dom mutation conferred by increases in proliferative activity.92 histopathologic changes. The term “latent precancer” has been ap-
Less specific information is available regarding estrogen influ- plied to this condition to emphasize that endometrial cancer risk is not
ence on cellular differentiation and growth. PAX2 is a paired-box gene necessarily increased without additional genetic change and onset of
known to be overexpressed in cancers of the kidney, prostate, breast, morphologic change. This preclinical phase of disease is recognizable
and ovary.93 PAX2 expression is increased by estrogens in neoplastic only through genetic analysis of mutant cells or identified by biomar-
endometrial epithelium but not in normal endometrium,94 indicating kers (Fig 1), and may persist through several years of cyclical menstrual
that neoplastic tissues have an intrinsically altered estrogen response shedding. During that interval, systemic hormonal factors positively
mechanism. Whether this is a cause or effect of malignant transforma- or negatively select for growth of these mutant cells. Inactivation of
tion is unclear. Endometrial cancer cell line expression of HOXB13, a the PTEN gene through deletion and/or mutation97 and acquisition
member of the HOX gene family can be induced by estrogens and of microsatellite instability33 are two examples of such early genetic

Fig 1. PTEN inactivation is one of the


earliest events in endometrial precancers.
Loss of expression is detectible by immuno-
histochemistry16 in scattered glands of
normal-appearing proliferative endometrium
(A), and carries forward to endometrial intra-
epithelial neoplasia (B) and carcinoma (C). In
each panel, notice the absence of staining in
the neoplastic glands; normal admixed
glands and stroma serve as an internal pos-
itive control.

4786 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Molecular Biology of Endometrial Cancer

events documented in otherwise normal appearing endometrium.


PREMALIGNANT EIN LESIONS CAN BE RECOGNIZED
The mutant cells in these cases participate in normal monthly endo-
BY SPECIFIC HISTOLOGIC CRITERIA
metrial regeneration, and may become widely dispersed by remodel-
ing of the endometrial compartment. More work is needed to define Accurate diagnosis of type 1 endometrial precancers has presented a
the long-term behavior of latent phases of carcinogenesis. major challenge to pathologists. The WHO endometrial hyperplasia
Clinically evident premalignant lesions that can be diagnosed schema captures many precancers in the atypical hyperplasia sub-
by a pathologist arise as localized clonal outgrowths of cytologically group, but is a poorly reproducible system,101-103 which, because of its
and architecturally altered endometrial glands that sequentially vintage, is missing diagnostic elements that have only become clear in
acquire additional somatic mutations leading to cancer (Fig 2). The the last 15 years. For example, the clonal origin of precancers is re-
premalignant lesions are referred to as EIN to distinguish them from flected in an initially localizing topographic distribution and a need to
the diffuse estrogen associated changes of benign endometrial hyper- establish size thresholds for diagnosis, and morphometric studies have
plasia. EIN is a histologically recognizable localized lesion composed precisely quantified diagnostic architectural changes.
of a clonal proliferation of glands and that usually carry one or several The application of computer based morphometry has been success-
of the genetic abnormalities associated with endometrioid carcino- ful at further improving diagnostic reproducibility of precursor diagnosis.
Biopsylesionsthatprogresstocarcinomacanbediscriminatedfromthose
ma.11,98 Markers that have been informative in clonal evaluation of
unlikely to progress using a series of discrete measurable variables.104,105
putative premalignant endometrial lesions include nonrandom
Volume percent stroma (a measure of gland crowding), standard devia-
X-chromosome inactivation (HUMARA assay) and clonal propaga-
tion of the shortest nuclear axis (a measure of nuclear pleomorphism),
tion of altered micosatellites.99,100 Identification of such monoclonal
and gland outer surface density (a measure of branching and folding)
lesions, combined with demonstration of lineage continuity with sub- when combined into a threshold D-score were effective at discriminat-
sequent carcinomas that occur in the same patient, provide a robust ing those endometrial lesions which progress to adenocarcinoma
standard for molecular definition of precancers.98 from those that do not.104,106-108 A recent meta-analysis combining
Comparison of the extent and range of genomic damage between the cumulative outcome prediction experience of the D-score (Fig 3)107
premalignant EIN and malignant carcinoma phases indicates a greater shows that patients with a D-score less than 1 have an overall 89-fold
cumulative mutational load in cancers, a feature that must contribute increased cancer risk than those with D-score more than 1. Even if
to their differing morphology and behavior. For example, while 55% one excludes concurrent cancers, those diagnosed within 12 months
of EIN lesions have demonstrable PTEN inactivating events the pro- of EIN, cancer risk over the next two decades is 45-fold elevated.
portion rises to 83% in those cancers which follow an EIN diagnosis.16 Molecular, histologic, and clinical outcome definitions of prema-
Similarly, for those lesions with microsatellite instability, the burden lignant disease have merged into a unified EIN concept. This was
of altered microsatellite alleles increases between EIN and carcino- possible through a study which showed that those high cancer risk
ma.11,98 Based on similarities between the length of DNA micro- lesions identified through molecular analysis (monoclonal lesions
satellites in adenocarcinoma and associated EIN in hysterectomy with lineage continuity to subsequent carcinoma) and morphometric
specimens, Faquin et al4 constructed philogenetic trees linking clonal analysis (D-Score) are essentially identical.98
foci of EIN to surface cancer and invasive cancer. These findings were
confirmed in a larger series of hysterectomy specimens.98 Although it
SUBJECTIVE (HISTOLOGIC, HUMAN-EYE) CRITERIA
is convenient to label PTEN and microsatellite alterations as “early”
events, these genetic targets may in fact be altered in the latent, prema-
lignant, or malignant disease phases. In order to develop histologic criteria that could be reproducibly
recognized by a practicing pathologist, a “type” collection of EIN

Fig 2. Initial genetic changes, including


PTEN inactivation and microsatellite instabil-
ity may occur in the absence of a change in
histomorphology. Endometrial intraepithelial
neoplasia arises through clonal expansion of
the mutant cells and is characterized by
altered cytology and architecture. The com-
bination and order of additional mutations
leading to invasive carcinoma differs be-
tween patients, but may include changes in
K-ras and ␤-catenin. Nongenetic factors such
as sex hormone exposures may act as pos-
itive (estrogens) or negative (progestins) se-
lectors of mutated clones.

www.jco.org 4787
Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Hecht and Mutter

risk by morphometry alone.110 While all morphometrically low-risk


endometria (D-score ⬎ 1) are consistently recognized by pathologists
as non-EIN, morphometrically high-risk endometria (D-score ⬍ 1),
comprise an admixture of subjectively benign endometria and EIN.
Pathologists are able to recognize a subset of endometria such as
normal secretory endometrium as low risk, whereas these benign
patterns demonstrate morphometric characteristics which mimic
those seen in EIN. Cases that progress to adenocarcinoma are mutu-
ally recognized as high risk by both D-score (D-score ⬍ 1) and sub-
jective (EIN) classification, emphasizing the need for the human
element in diagnosis to ensure diagnostic specificity.

CONCLUSION

For the most part, the dichotomous classification of endometrial can-


cers holds up to genetic analysis and the histologic subtypes are under-
scored by systematic changes in a limited set of genes. Combined
molecular, histomorphometric, and clinical outcome analysis of
premalignant lesions, has provided a clearer multidisciplinary def-
inition of endometrial precancers, known as EIN. Genetic and
Fig 3. Overall cancer free survival of 674 patients with “endometrial hyperpla-
sia” stratified by morphometry into endometrial intraepithelial neoplasia (EIN) or endocrine disease mechanisms have been integrated into a multi-
(benign) non-EIN categories.106 Almost all cancer occurrences occur in patients step model for oncogenesis in which hormonal exposures act as
with EIN, with a relative risk of 89 compared to those without EIN. Carcinomas
following EIN might be considered concurrent when seen within the first year of
selection factors for mutated endometrial cells. Of particular inter-
EIN diagnosis. Excluding these, the longer term risk for carcinoma is 45 times est is the newly defined preclinical phase of “latent” precancers
higher for EIN patients. in which mutant cells capable of responding to the modulating
influences of hormones have been shown to commonly occur in
lesions complete with photomicrographs and associated specialized “normal” endometrium. This is a potentially exciting target for pre-
studies was created.109 Computerized morphometric analysis of these ventative therapy.
and other samples with respect to clinical follow-up have defined Although the model in Figure 2 is appealing, it is incomplete. We
architectural and cytologic features of endometrial precancers; a de- have described the role of several candidate genes active in this pro-
tailed description and examples are available at www.endometrium.org. gression scenario, but not all genes are effective in each case. Clearly,
Application of these criteria has been shown to be reproducible, and there are other primary events that drive progression. These could be
reliable in predicting concurrent cancer and the presence of monoclo- abnormalities of genes elsewhere in the regulatory pathways repre-
nal growth.110-112 sented by the exemplars described here, or perhaps entirely new path-
Pathologists applying EIN diagnostic criteria subjectively, are ways that have yet to be discovered. Finally, the heterogeneity of
able to recognize some lesions as low risk that would be scored as high endometrioid cancers has not been fully explained.

8. Coller HA, Grandori C, Tamayo P, et al: nomas with microsatellite instability. Jpn J Cancer
REFERENCES Expression analysis with oligonucleotide microar- Res 87:141-145, 1996
rays reveals that MYC regulates genes involved in 14. Catasus L, Bussaglia E, Rodrguez I, et al:
1. Bokhman JV: Two pathogenetic types of en- growth, cell cycle, signaling, and adhesion. Proc Natl Molecular genetic alterations in endometrioid carcino-
dometrial carcinoma. Gynecol Oncol 15:10-17, 1983 Acad Sci U S A 97:3260-3265, 2000 mas of the ovary: Similar frequency of beta-catenin
2. American Cancer Society: Cancer Facts and
9. Risinger JI, Maxwell GL, Chandramouli GV, abnormalities but lower rate of microsatellite instability
Figures 2006. Available at http://www.cancer.org/
et al: Microarray analysis reveals distinct gene ex- and PTEN alterations than in uterine endometrioid
downloads/STT/CAFF2006PWSecured.pdf
pression profiles among different histologic types of carcinomas. Hum Pathol 35:1360-1368, 2004
3. Sherman ME, Bur ME, Kurman RJ: p53 in
endometrial cancer. Cancer Res 63:6-11, 2003 15. Risinger JI, Berchuck A, Kohler MF, et al:
endometrial cancer and its putative precursors: Evi-
10. Zorn KK, Bonome T, Gangi L, et al: Gene Genetic instability of microsatellites in endometrial
dence for diverse pathways of tumorigenesis. Hum
expression profiles of serous, endometrioid, and carcinoma. Cancer Res 53:5100-5103, 1993
Pathol 26:1268-1274, 1995
clear cell subtypes of ovarian and endometrial can- 16. Mutter GL, Lin MC, Fitzgerald JT, et al:
4. Faquin WC, Fitzgerald JT, Boynton KA, et al:
Intratumoral genetic heterogeneity and progression cer. Clin Cancer Res 11:6422-6430, 2005 Altered PTEN expression as a diagnostic marker for
of endometrioid type endometrial adenocarcinomas. 11. Mutter GL, Boynton KA, Faquin WC, et al: the earliest endometrial precancers. J Natl Cancer
Gynecol Oncol 78:152-157, 2000 Allelotype mapping of unstable microsatellites es- Inst 92:924-930, 2000
5. Esteller M, Levine R, Baylin SB, et al: MLH1 tablishes direct lineage continuity between endo- 17. Gurin CC, Federici MG, Kang L, et al: Causes
promoter hypermethylation is associated with the metrial precancers and cancer. Cancer Res 56: and consequences of microsatellite instability in
microsatellite instability phenotype in sporadic endo- 4483-4486, 1996 endometrial carcinoma. Cancer Res 59:462-466,
metrial carcinomas. Oncogene 17:2413-2417, 1998 12. Duggan BD, Felix JC, Muderspach LI, et al: 1999
6. Mutter GL: Pten, a protean tumor suppres- Microsatellite instability in sporadic endometrial car- 18. Levine RL, Cargile CB, Blazes MS, et al:
sor. Am J Pathol 158:1895-1898, 2001 cinoma. J Natl Cancer Inst 86:1216-1221, 1994 PTEN mutations and microsatellite instability in
7. Matias-Guiu X, Catasus L, Bussaglia E, et al: 13. Kobayashi K, Matsushima M, Koi S, et al: complex atypical hyperplasia, a precursor lesion to
Molecular pathology of endometrial hyperplasia and Mutational analysis of mismatch repair genes, uterine endometrioid carcinoma. Cancer Res 58:
carcinoma. Hum Pathol 32:569-577, 2001 hMLH1 and hMSH2, in sporadic endometrial carci- 3254-3258, 1998

4788 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Molecular Biology of Endometrial Cancer

19. Maxwell GL, Risinger JI, Gumbs C, et al: 38. Swisher EM, Mutch DG, Herzog TJ, et al: ment of multiple tumor suppressors. Gynecol Oncol
Mutation of the PTEN tumor suppressor gene in Analysis of MSH3 in endometrial cancers with de- 71:391-395, 1998
endometrial hyperplasias. Cancer Res 58:2500- fective DNA mismatch repair. J Soc Gynecol Inves- 55. Peiffer SL, Herzog TJ, Tribune DJ, et al:
2503, 1998 tig 5:210-216, 1998 Allelic loss of sequences from the long arm of
20. Risinger JI, Hayes AK, Berchuck A, et al: 39. Risinger JI, Umar A, Boyd J, et al: Mutation chromosome 10 and replication errors in endome-
PTEN/MMAC1 mutations in endometrial cancers. of MSH3 in endometrial cancer and evidence for its trial cancers. Cancer Res 55:1922-1926, 1995
Cancer Res 57:4736-4738, 1997 functional role in heteroduplex repair. Nat Genet 56. Noonan FC, Mutch DG, Ann Mallon M, et al:
21. Tashiro H, Blazes MS, Wu R, et al: Mutations 14:102-105, 1996 Characterization of the homeodomain gene EMX2:
in PTEN are frequent in endometrial carcinoma but 40. Cohn DE, Basil JB, Venegoni AR, et al: sequence conservation, expression analysis, and a
rare in other common gynecological malignancies. Absence of PTEN repeat tract mutation in endome- search for mutations in endometrial cancers.
Cancer Res 57:3935-3940, 1997 trial cancers with microsatellite instability. Gynecol Genomics 76:37-44, 2001
22. Mutter GL, Wada H, Faquin WC, et al: K-ras Oncol 79:101-106, 2000 57. Sasaki H, Nishii H, Takahashi H, et al: Muta-
mutations appear in the premalignant phase of both 41. Esteller M, Catasus L, Matias-Guiu X, et al: tion of the Ki-ras protooncogene in human endome-
microsatellite stable and unstable endometrial carci- hMLH1 promoter hypermethylation is an early event trial hyperplasia and carcinoma. Cancer Res 53:
nogenesis. Mol Pathol 52:257-262, 1999 in human endometrial tumorigenesis. Am J Pathol 1906-1910, 1993
23. Enomoto T, Inoue M, Perantoni AO, et al: 155:1767-1772, 1999 58. Palacios J, Gamallo C: Mutations in the beta-
K-ras activation in premalignant and malignant epi- 42. Catasus L, Matias-Guiu X, Machin P, et al: catenin gene (CTNNB1) in endometrioid ovarian
thelial lesions of the human uterus. Cancer Res BAX somatic frameshift mutations in endometrioid carcinomas. Cancer Res 58:1344-1347, 1998
51:5308-5314, 1991 adenocarcinomas of the endometrium: Evidence for 59. Morin PJ, Sparks AB, Korinek V, et al: Acti-
24. Fujimoto I, Shimizu Y, Hirai Y, et al: Studies a tumor progression role in endometrial carcinomas vation of beta-catenin-Tcf signaling in colon cancer
on ras oncogene activation in endometrial carci- with microsatellite instability. Lab Invest 78:1439- by mutations in beta-catenin or APC. Science 275:
noma. Gynecol Oncol 48:196-202, 1993 1444, 1998 1787-1790, 1997
25. Sakamoto T, Murase T, Urushibata H, et al: 43. Catasus L, Matias-Guiu X, Machin P, et al: 60. Su LK, Vogelstein B, Kinzler KW: Association
Microsatellite instability and somatic mutations in Frameshift mutations at coding mononucleotide of the APC tumor suppressor protein with catenins.
endometrial carcinomas. Gynecol Oncol 71:53-58, repeat microsatellites in endometrial carcinoma Science 262:1734-1737, 1993
1998 with microsatellite instability. Cancer 88:2290- 61. Rubinfeld B, Albert I, Porfiri E, et al: Binding
26. Swisher EM, Peiffer-Schneider S, Mutch 2297, 2000 of GSK3beta to the APC-beta-catenin complex and
DG, et al: Differences in patterns of TP53 and 44. Ouyang H, Shiwaku HO, Hagiwara H, et al:
regulation of complex assembly. Science 272:1023-
KRAS2 mutations in a large series of endometrial The insulin-like growth factor II receptor gene is
1026, 1996
carcinomas with or without microsatellite instability. mutated in genetically unstable cancers of the en-
62. Irving JA, Catasus L, Gallardo A, et al: Syn-
dometrium, stomach, and colorectum. Cancer Res
Cancer 85:119-126, 1999 chronous endometrioid carcinomas of the uterine
57:1851-1854, 1997
27. Lax SF, Kendall B, Tashiro H, et al: The corpus and ovary: Alterations in the beta-catenin
45. Ouyang H, Furukawa T, Abe T, et al: The BAX
frequency of p53, K-ras mutations, and microsatel- (CTNNB1) pathway are associated with independent
gene, the promoter of apoptosis, is mutated in
lite instability differs in uterine endometrioid and primary tumors and favorable prognosis. Hum
genetically unstable cancers of the colorectum,
serous carcinoma: Evidence of distinct molecular Pathol 36:605-619, 2005
stomach, and endometrium. Clin Cancer Res
genetic pathways. Cancer 88:814-824, 2000 63. Saegusa M, Hashimura M, Kuwata T, et al:
4:1071-1074, 1998
28. Lagarda H, Catasus L, Arguelles R, et al: Upregulation of TCF4 expression as a transcriptional
46. Schwartz S Jr, Yamamoto H, Navarro M, et
K-ras mutations in endometrial carcinomas with target of beta-catenin/p300 complexes during trans-
al: Frameshift mutations at mononucleotide repeats
microsatellite instability. J Pathol 193:193-199, 2001 differentiation of endometrial carcinoma cells. Lab
in caspase-5 and other target genes in endometrial
29. Schlosshauer PW, Pirog EC, Levine RL, et al: Invest 85:768-779, 2005
and gastrointestinal cancer of the microsatellite mu-
Mutational analysis of the CTNNB1 and APC genes 64. Saegusa M, Hashimura M, Yoshida T, et al:
tator phenotype. Cancer Res 59:2995-3002, 1999
in uterine endometrioid carcinoma. Mod Pathol 13: beta- Catenin mutations and aberrant nuclear ex-
47. Vassileva V, Millar A, Briollais L, et al: Apo-
1066-1071, 2000 pression during endometrial tumorigenesis. Br J
ptotic and growth regulatory genes as mutational
30. Mirabelli-Primdahl L, Gryfe R, Kim H, et al: Cancer 84:209-217, 2001
targets in mismatch repair deficient endometrioid
Beta-catenin mutations are specific for colorectal 65. Kim YT, Choi EK, Kim JW, et al: Expression
adenocarcinomas of young patients. Oncol Rep 11:
carcinomas with microsatellite instability but occur of E-cadherin and alpha-, beta-, gamma-catenin pro-
931-937, 2004
in endometrial carcinomas irrespective of mutator 48. Sakaguchi J, Kyo S, Kanaya T, et al: Aberrant teins in endometrial carcinoma. Yonsei Med J 43:
pathway. Cancer Res 59:3346-3351, 1999 expression and mutations of TGF-beta receptor type 701-711, 2002
31. Fukuchi T, Sakamoto M, Tsuda H, et al: II gene in endometrial cancer. Gynecol Oncol 98: 66. Shih HC, Shiozawa T, Miyamoto T, et al:
Beta-catenin mutation in carcinoma of the uterine 427-433, 2005 Immunohistochemical expression of E-cadherin and
endometrium. Cancer Res 58:3526-3528, 1998 49. Kurose K, Zhou XP, Araki T, et al: Frequent beta-catenin in the normal and malignant human
32. Burks RT, Kessis TD, Cho KR, et al: Micro- loss of PTEN expression is linked to elevated phos- endometrium: An inverse correlation between
satellite instability in endometrial carcinoma. Onco- phorylated Akt levels, but not associated with p27 E-cadherin and nuclear beta-catenin expression. An-
gene 9:1163-1166, 1994 and cyclin D1 expression, in primary epithelial ovar- ticancer Res 24:3843-3850, 2004
33. Faquin W, Fitzgerald J, Lin M, et al: Sporadic ian carcinomas. Am J Pathol 158:2097-2106, 2001 67. Shtutman M, Zhurinsky J, Simcha I, et al:
microsatellite instability is specific to neoplastic and 50. Zhu X, Kwon CH, Schlosshauer PW, et al: The cyclin D1 gene is a target of the beta-catenin/
preneoplastic endometrial tissues. Am J Clin Pathol PTEN induces G(1) cell cycle arrest and decreases LEF-1 pathway. Proc Natl Acad Sci U S A 96:5522-
113:576-582, 2000 cyclin D3 levels in endometrial carcinoma cells. 5527, 1999
34. Goodfellow P, Buttin B, Herzog T, et al: Cancer Res 61:4569-4575, 2001 68. Brabletz T, Jung A, Dag S, et al: beta-
Prevalence of defective DNA mismatch repair and 51. Oda K, Stokoe D, Taketani Y, et al: High catenin regulates the expression of the matrix
MSH6 mutation in an unselected series of endome- frequency of coexistent mutations of PIK3CA and metalloproteinase-7 in human colorectal cancer.
trial cancers. Proc Natl Acad Sci U S A 100:5908- PTEN genes in endometrial carcinoma. Cancer Res Am J Pathol 155:1033-1038, 1999
5913, 2003 65:10669-10673, 2005 69. Risinger JI, Maxwell GL, Chandramouli GV,
35. Basil JB, Goodfellow PJ, Rader JS, et al: 52. Stambolic V, Tsao MS, Macpherson D, et al: et al: Gene expression profiling of microsatellite
Clinical significance of microsatellite instability in High incidence of breast and endometrial neoplasia unstable and microsatellite stable endometrial can-
endometrial carcinoma. Cancer 89:1758-1764, 2000 resembling human Cowden syndrome in pten⫹/⫺ cers indicates distinct pathways of aberrant signal-
36. Gryfe R, Kim H, Hsieh ET, et al: Tumor mice. Cancer Res 60:3605-3611, 2000 ing. Cancer Res 65:5031-5037, 2005
microsatellite instability and clinical outcome in 53. Kong D, Suzuki A, Zou TT, et al: PTEN1 is 70. Alkushi A, Lim P, Coldman A, et al: Interpre-
young patients with colorectal cancer. N Engl J Med frequently mutated in primary endometrial carcino- tation of p53 immunoreactivity in endometrial carci-
342:69-77, 2000 mas. Nat Genet 17:143-144, 1997 noma: Establishing a clinically relevant cut-off level.
37. Dovrat S, Figer A, Fidder HH, et al: Muta- 54. Simpkins SB, Peiffer-Schneider S, Mutch Int J Gynecol Pathol 23:129-137, 2004
tional analysis of hMsh6 in Israeli HNPCC and DG, et al: PTEN mutations in endometrial cancers 71. Pijnenborg JM, van de Broek L, Dam de
HNPCC-like Families. Fam Cancer 4:291-294, 2005 with 10q LOH: Additional evidence for the involve- Veen GC, et al: TP53 overexpression in recurrent

www.jco.org 4789
Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Hecht and Mutter

endometrial carcinoma. Gynecol Oncol 100:397- 86. Mutter GL, Baak JP, Fitzgerald JT, et al: 100. Mutter GL, Chaponot ML, Fletcher JA: A
404, 2006 Global expression changes of constitutive and hor- polymerase chain reaction assay for non-random X
72. Soslow RA, Shen PU, Chung MH, et al: monally regulated genes during endometrial neo- chromosome inactivation identifies monoclonal en-
Distinctive p53 and mdm2 immunohistochemical plastic transformation. Gynecol Oncol 83:177-185, dometrial cancers and precancers. Am J Pathol
expression profiles suggest different pathogenetic 2001 146:501-508, 1995
pathways in poorly differentiated endometrial carci- 87. Mutter GL, Lin MC, Fitzgerald JT, et al: 101. Zaino RJ: Endometrial hyperplasia: Is it time
noma. Int J Gynecol Pathol 17:129-134, 1998 Changes in endometrial PTEN expression through- for a quantum leap to a new classification? Int J
73. Schmitz M, Hendricks D, Farley J, et al: p27 out the human menstrual cycle. J Clin Endocrinol Gynecol Pathol 19:314-321, 2000
and cyclin D1 abnormalities in uterine papillary se- Metab 85:2334-2338, 2000 102. Hunter JE, Tritz DE, Howell MG, et al: The
rous carcinoma. Gynecol Oncol 77:439-445, 2000 88. Zheng W, Baker HE, Mutter GL: Involution of prognostic and therapeutic implications of cytologic
74. MacCallum D, Hupp T: Induction of p53 PTEN-null endometrial glands with progestin ther- atypia in patients with endometrial hyperplasia. Gy-
protein as a marker for ionizing radiation exposure in apy. Gynecol Oncol 92:1008-1013, 2004 necol Oncol 55:66-71, 1994
vivo. Methods Mol Biol 113:583-589, 1999 89. Vereide AB, Kaino T, Sager G, et al: Bcl-2, 103. Skov BG, Broholm H, Engel U, et al: Com-
75. Zhao Y, Yamashita T, Ishikawa M: Regulation BAX, and apoptosis in endometrial hyperplasia after parison of the reproducibility of the WHO classifica-
of tumor invasion by HOXB13 gene overexpressed high dose gestagen therapy: A comparison of tions of 1975 and 1994 of endometrial hyperplasia.
in human endometrial cancer. Oncol Rep 13:721- responses in patients treated with intrauterine Int J Gynecol Pathol 16:33-37, 1997
726, 2005 levonorgestrel and systemic medroxyprogesterone. 104. Baak JP, Nauta JJ, Wisse-Brekelmans EC, et
76. Whitcomb BP, Mutch DG, Herzog TJ, et al: Gynecol Oncol 97:740-750, 2005 al: Architectural and nuclear morphometrical fea-
Frequent HOXA11 and THBS2 promoter methyl- 90. Wang S, Pudney J, Song J, et al: Mecha- tures together are more important prognosticators
ation, and a methylator phenotype in endometrial nisms involved in the evolution of progestin resis- in endometrial hyperplasias than nuclear morpho-
adenocarcinoma. Clin Cancer Res 9:2277-2287, tance in human endometrial hyperplasia–precursor metrical features alone. J Pathol 154:335-341, 1988
2003 of endometrial cancer. Gynecol Oncol 88:108-117, 105. Baak JP, Wisse-Brekelmans EC, Fleege JC,
77. Saito T, Nishimura M, Yamasaki H, et al: et al: Assessment of the risk on endometrial cancer
2003
Hypermethylation in promoter region of E-cadherin
91. Burcham PC: Internal hazards: Baseline DNA in hyperplasia, by means of morphological and mor-
gene is associated with tumor dedifferention and
damage by endogenous products of normal metab- phometrical features. Pathol Res Pract 188:856-859,
myometrial invasion in endometrial carcinoma. Can-
olism. Mutat Res 443:11-36, 1999 1992
cer 97:1002-1009, 2003
92. Cairns J: Mutation and cancer: The anteced- 106. Dunton CJ, Baak JP, Palazzo JP, et al: Use of
78. Hardman RA, Afshari CA, Barrett JC: Involve-
ents to our studies of adaptive mutation. Genetics computerized morphometric analyses of endome-
ment of mammalian MLH1 in the apoptotic re-
148:1433-1440, 1998 trial hyperplasias in the prediction of coexistent
sponse to peroxide-induced oxidative stress. Cancer
93. Robson EJ, He SJ, Eccles MR: A PANorama cancer. Am J Obstet Gynecol 174:1518-1521, 1996
Res 61:1392-1397, 2001
of PAX genes in cancer and development. Nat Rev 107. Baak JP, Mutter GL, Robboy S, et al: The
79. Takai N, Kawamata N, Walsh CS, et al:
Cancer 6:52-62, 2006 molecular genetics and morphometry-based endo-
Discovery of epigenetically masked tumor suppres-
94. Wu H, Chen Y, Liang J, et al: Hypomethylation- metrial intraepithelial neoplasia classification system
sor genes in endometrial cancer. Mol Cancer Res
linked activation of PAX2 mediates tamoxifen-stimulated predicts disease progression in endometrial hyper-
3:261-269, 2005
endometrial carcinogenesis. Nature 438:981-987, 2005 plasia more accurately than the 1994 World Health
80. Effects of hormone replacement therapy on
95. Zukerberg LR, DeBernardo RL, Kirley SD, et Organization classification system. Cancer 103:
endometrial histology in postmenopausal women:
The Postmenopausal Estrogen/Progestin Interven- al: Loss of cables, a cyclin-dependent kinase regu- 2304-2312, 2005
tions (PEPI) Trial. The Writing Group for the PEPI latory protein, is associated with the development of 108. Orbo A, Baak JP, Kleivan I, et al: Computer-
Trial. JAMA 275:370-375, 1996 endometrial hyperplasia and endometrial cancer. ised morphometrical analysis in endometrial hyper-
81. Zeleniuch-Jacquotte A, Akhmedkhanov A, Cancer Res 64:202-208, 2004 plasia for the prediction of cancer development: A
Kato I, et al: Postmenopausal endogenous oestro- 96. Zaino R, Trimble C, Silverberg S, et al: Re- long-term retrospective study from northern Nor-
gens and risk of endometrial cancer: Results of a producibility Of The Diagnosis Of Atypical Endome- way. J Clin Pathol 53:697-703, 2000
prospective study. Br J Cancer 84:975-981, 2001 trial Hyperplasia (AEH): A Gynecologic Oncology 109. http://www.endometrium.org/
82. Potischman N, Hoover RN, Brinton LA, et al: Group (GOG) Study. Cancer 106:804-811, 2006 110. Hecht JL, Ince TA, Baak JP, et al: Prediction
Case-control study of endogenous steroid hor- 97. Mutter GL, Ince TA, Baak JP, et al: Molecular of endometrial carcinoma by subjective endometrial
mones and endometrial cancer. J Natl Cancer Inst identification of latent precancers in histologically intraepithelial neoplasia diagnosis. Mod Pathol 18:
88:1127-1135, 1996 normal endometrium. Cancer Res 61:4311-4314, 324-330, 2005
83. Parazzini F, La Vecchia C, Bocciolone L, et al: 2001 111. Silverberg S, Mutter G, Kurman R: Tumors of
The epidemiology of endometrial cancer. Gynecol 98. Mutter GL, Baak JP, Crum CP, et al: Endo- the uterine corpus: Epithelial tumors and related
Oncol 41:1-16, 1991 metrial precancer diagnosis by histopathology, lesions, in: FA T, MR S, eds: WHO Classification of
84. Weiderpass E, Adami HO, Baron JA, et al: clonal analysis, and computerized morphometry. Tumors: Pathology and Genetics of Tumors of the
Use of oral contraceptives and endometrial cancer J Pathol 190:462-469, 2000 Breast and Female Genital Organs, ed. 1 Lyon,
risk (Sweden). Cancer Causes Control 10:277-284, 99. Jovanovic AS, Boynton KA, Mutter GL: Uteri France, IARC Press, 2003, pp 221-232
1999 of women with endometrial carcinoma contain a 112. Mutter GL: Endometrial intraepithelial neo-
85. Grimes DA, Economy KE: Primary preven- histopathological spectrum of monoclonal putative plasia (EIN): will it bring order to chaos? The
tion of gynecologic cancers. Am J Obstet Gynecol precancers, some with microsatellite instability. Endometrial Collaborative Group. Gynecol Oncol
172:227-235, 1995 Cancer Res 56:1917-1921, 1996 76:287-290, 2000

■ ■ ■

Authors’ Disclosures of Potential Conflicts of Interest


The authors indicated no potential conflicts of interest.

Author Contributions

Manuscript writing: Jonathan L. Hecht, George L. Mutter


Final approval of manuscript: Jonathan L. Hecht, George L. Mutter

4790 JOURNAL OF CLINICAL ONCOLOGY


Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.
Molecular Biology of Endometrial Cancer

GLOSSARY

␤-catenin: Originally identified as a component of cell-cell Non-random X-chromosome inactivation (HUMARA


adhesion complexes composed of cadherins and actin, ␤-catenin assay): Tumors arise from a single genetically altered cell. Conse-
has now been shown to be a downstream signaling molecule in quently, detection of monoclonality, a genetic similarity among the
the Wnt signaling pathway. cells in a tumor mass, can be used to distinguish neoplastic from
polyclonal or hyperplastic lesions. Clonality can be established based
EIN (endometrial intraepithelial neoplasia): EIN is a on X-chromosome inactivation analysis. During early embryonic
premalignant lesion of the endometrium composed of a localized development of mammalian female embryos, one of the two
monoclonal population of genetically altered glands. The pres- X-chromosomes in each cell is randomly deactivated by the methyl-
ence of EIN on biopsy indicates a 30% chance of concurrent type ation of deoxycytosine residues. Once established, genetic imprinting
I (endometrioid) endometrial cancer and a 45 fold increased risk retains X-chromosome inactivation through all subsequent cell divi-
for developing cancer 1 or more years later. sions. The cells in a clonal tumor all show inactivation of the same
K-ras: The gene that encodes K-Ras, a protein that is a member copy of the X-chromosome. One common assay detects this inactiva-
of the small GTPase superfamily, in which a single amino acid tion by examining inactivation of the human androgen receptor
substitution results in an activating mutation. Alternative splicing (HUMARA) on the X-chromosome.
gives rise to variants encoding two isoforms that differ in the
C-terminal region. Null: Complete loss of function phenotype. This term is used in refer-
ence to the mutation or inactivation of specific genes, resulting in com-
LOH (loss of heterozygosity): A situation where one plete absence of protein expression.
chromosome has a normal allele of a gene and one chromosome
has a mutant or deleted allele. p53: p53 is a tumor suppressor gene. The normal function of p53 is
to act as a transcriptional activator of genes with a p53-binding site
Mismatch repair: One of four major pathways of DNA re-
and an inhibitor of genes lacking a p53 binding site. Expression of
pair in mammalian cells. Mismatch repair recognizes and cor-
high levels of wild-type p53 is associated with cell cycle arrest and
rects errors in DNA replication leading to single base-pair
apoptosis through induction of P21Waf1/Cip1 and hMdm2. Muta-
mismatches or insertions/deletions in small repetitive tracts of
tions in p53 are seen in many tumors as well as in the Li-Fraumeni–
DNA known as microsatellites.
inherited cancer syndrome.
MSI (microsatellite instability): Microsatellites are re-
peating units of 1-4 DNA base pairs that are distributed widely PTEN (phosphatase and tensin homolog): PTEN is a tumor
throughout the genome and have a high degree of repeat length suppressor gene with a gamut of regulatory activities. The gene product
variation in the population. Their length remains stable with cell is a multifunctional molecule. The predominant activity identified for
division and inheritance so they may be used as molecular mark- PTEN is its lipid phosphatase activity that converts inositol trisphos-
ers of cell lineage, in population genetic studies or paternity test- phates into inositol bisphosphates, thus inhibiting survival and prolifer-
ing. Defects in the genes involved in DNA mismatch repair result ative pathways that are activated by inositol trisphosphates. PTEN acts
in genomic instability that may be detected as MSI, an alteration to maintain arrest in the G1 phase of the cell cycle and enable apoptosis
in the length of the microsatellites from cell to cell. through an AKT-dependent mechanism.

www.jco.org 4791
Information downloaded from jco.ascopubs.org and provided by National Medical Library on April 7, 2007 from
132.64.167.162.
Copyright © 2006 by the American Society of Clinical Oncology. All rights reserved.

You might also like