10.1007@978 981 13 3438 23

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Endometrial Hyperplasia:

Diagnosis and Management


3
Bijal M. Patel

Abbreviations NETA Norethisterone acetate


OCs Oral contraceptives
AUB Abnormal uterine bleeding PCNA Proliferating cell nuclear antigen
BMI Body mass index PCOS Polycystic ovarian syndrome
BSO Bilateral salpingo-oophorectomy PMB Postmenopausal bleeding
CT Computerized tomography PR Progesterone receptor
D&C Dilatation and curettage RCOG Royal College of Obstetricians and
DMPA Depot medroxyprogesterone acetate Gynaecologists
EB Endometrial biopsy SERM Selective estrogen receptor modulator
EC Endometrial carcinoma SNPs Single nucleotide polymorphisms
EGF Epithelial growth factor TAH Total abdominal hysterectomy
EH Endometrial hyperplasia TNF R1 Tumor necrosis factor receptor 1
EIN Endometrial intraepithelial neoplasia TNF-α Tumor necrosis factor-α
ER Estrogen receptors TVS Transvaginal sonography
ERT Estrogen replacement therapy USG Ultrasonography
ET Endometrial thickness WHO World Health Organization
GnRH Gonadotropin-releasing hormone
H&E Hematoxylin and eosin
HRT Hormone replacement therapy
IGF-1 Insulin-like growth factor-1 3.1 Introduction
IL-1β Interleukin-1β
IUD Intrauterine device Endometrial hyperplasia (EH) is a spectrum of
LNG Levonorgestrel morphological changes ranging from a slightly
MA Megestrol acetate disordered pattern seen in the late proliferative
MPA Medroxyprogesterone acetate phase of the menstrual cycle to the irregular pro-
MRI Magnetic resonance imaging liferation of the endometrial glands with an
MSI Microsatellite instability increase in gland-to-stroma ratio leading to thick-
ening of the endometrium [1]. It is further classi-
fied on the basis of the complexity of endometrial
glands and any cytological atypia [2]. In fact, EH
B. M. Patel (*)
Department of Gynecologic Oncology, is the only known direct precursor of endometrial
Gujarat Cancer and Research Institute, Ahmedabad, carcinoma (EC). These lesions range from
Gujarat, India anovulatory endometrium to monoclonal
­
© Springer Nature Singapore Pte Ltd. 2019 25
S. Mehta, A. Singla (eds.), Preventive Oncology for the Gynecologist,
https://doi.org/10.1007/978-981-13-3438-2_3
26 B. M. Patel

p­ recancers. Menstrual cycle involves a complex 3.2 Etiology and Risk Factors
interaction of estrogen and progesterone hor-
mones which affects the endometrial lining. A relative excess of estrogen, unopposed by pro-
Factors like hormonal balance, molecular mecha- gesterone, whether it is exogenous or endogenous
nisms, age, environment, and so forth maintain is thought to be one of the primary etiological
the fine equilibrium of the endometrium, and any factors in both EH and EC. Estrogen stimulates
disturbance leading to chronic estrogen stimula- endometrial proliferation by binding to estrogen
tion may lead to several endometrial abnormali- receptors (ER) in the nuclei of endometrial cells.
ties [1, 3, 4]. In 1900, Cullen described an Known risk factors for EH reflect this etiology [1,
etiologic correlation between EH and EC. In 2, 7, 12–14] (Table 3.1).
1932, Taylor and also in 1936, Novak and Yui Hormone replacement therapy (HRT) and
supported this observation. In 1947, Gusberg obesity are considered as reversible risk factors.
focused his attention on the role of estrogenic Postmenopausal women treated with estrogen
stimulation as a factor that caused EH and EC replacement therapy (ERT) without progestins
[5]. Speert (1952) introduced the term “adenoma- are at increased risk of EH. The risk of EH
tous atypical hyperplasia” for the premalignant increases by tenfold with each decade of use of
hyperplasias. EH has been classified by various ERT [1, 2, 15]. Obese women (body mass index
systems over the last 50 years. In 1982, Kurman [BMI] > 30 kg/m2) have a nearly fourfold increase
and Norris first published the study describing in the incidence of atypical EH due to excessive
reproducible criteria for differentiating EH from peripheral conversion of androgens in adipose
well-differentiated EC [6]. The estimated inci- tissue to estrogen coupled with erratic anovula-
dence of EH in developed countries is 200,000 tory cycles [16, 17].
new cases per year [1, 7]. EH incidence without
atypia and with atypia peaks in the early post-
menopausal years and early 60s, respectively [8].
Prospective studies are difficult to conduct to Table 3.1 Risk factors responsible for development of
EH [1, 2, 7, 12–14]
determine the malignant potential of these lesions
for several reasons. The obstacle to long-term Category of
risk factor Risk factor
surveillance is that the condition is usually
Nonmodifiable Increasing age (age >35 years)
detected in symptomatic women who therefore Caucasian
require treatment so that follow-up to determine Family history of endometrium,
the natural history of the various histological sub- ovarian, breast, or colon cancer
types is difficult. There is also difficulty in accu- Lifestyle Smoking
Menstrual Early menarche and/or late
rately differentiating between the atypical
status menopause, postmenopausal
hyperplasia and well-differentiated EC [9]. Reproductive Nulliparity, infertility
Diagnosis of EH is important because it may events
cause abnormal uterine bleeding (AUB), indica- Comorbidity Obesity, DM (type II), metabolic
tive of anovulation and infertility, associated with syndrome, insulin resistance, HT,
PCOS (anovulation), Lynch syndrome
estrogen-producing ovarian tumors, and precede
(HNPCC), estrogen-secreting ovarian
or occur simultaneously with EC [10]. Sensitive tumors (e.g., ovarian granulosa cell
and accurate diagnosis can reduce the likelihood tumors), androgen-secreting tumors of
of development of invasive EC [11]. Regression the adrenal cortex
of hyperplasia to normal endometrium represents Drug-induced Unopposed ERT/prolonged HRT,
long-term tamoxifen therapy
the key to conservative treatment for prevention Others Immunosuppression, infection
of the development of EC [1]. Genetic MSI, PTEN, K-ras, β-catenin,
A brief overview of the development of a cur- mutations PIK3CA, SNPs
rent understanding of EH will serve to under- Cytokine TNF-α, PCNA, EGF, Fas, TNF-R1,
stand their diagnosis and management. system IGF-1, NF-κB, IL-22
3 Endometrial Hyperplasia: Diagnosis and Management 27

Tamoxifen, which is a selective estrogen recep- EC in hysterectomy specimens of up to 50% of


tor modulator (SERM), is used to treat ERα- women with atypia [2, 5]. Dilatation and curet-
positive primary and advanced breast cancers. It tage (D&C) or hysteroscopy-guided biopsy may
leads to EH, development of endometrial polyps, not always be completely representative of the
abnormal vaginal bleeding, and EC due to its entire endometrium. Small foci of malignancy
estrogenic effect on the endometrium [1, 17]. left in situ at the first biopsy might have already
In addition to estrogenic stimulation of the been present in the endometrium. In this situa-
endometrium, other elements such as immuno- tion, the term “progression to carcinoma” is less
suppression and infection may also be involved appropriate than “association with cancer/coexis-
in the development of EH [13]. tent carcinoma” [19].
Genetic alterations like microsatellite instabil- EC with concomitant EH are thought to be
ity (MSI), PTEN mutations, K-ras mutation, beta- associated with less aggressive disease, associ-
catenin mutation, PIK3CA mutation, functional ated with lower grade and stage, significantly
single nucleotide polymorphisms (SNPs), and so lower recurrence risk, and higher 5-year survival
forth are observed in endometrial lesions [1, 14]. rates [2]. The strongest predictors of concurrent
Inflammation in the endometrium disturbs the EC among women with EH are older age, obesity,
balanced cytokine system which leads to most diabetes mellitus, and complex hyperplasia [4].
cases of EH. Inflammation causes decrease in Immunohistochemical staining of complex atypi-
tumor necrosis factor-α (TNF-α), proliferating cell cal hyperplasia for PTEN, MIB-1, and p53
nuclear antigen (PCNA), and epithelial growth fac- improves the prediction of coexistent EC [10, 20].
tor (EGF) mRNA and increased production of Fas
mRNA and IGF-1 receptor (IGF-1R). In glandular
cystic hyperplasia, decreased expression of tumor 3.3.2 Progression to Carcinoma
necrosis factor receptor 1 (TNF R1), interleukin-1β
(IL-1β), and IL-12 genes is found. The expression The natural history of EH is difficult to define,
of the insulin-­like growth factor-1 (IGF-1) gene is but key factors defining the risk for progression
reduced only in adenomatous hyperplasia [1]. to carcinoma are the presence and severity of
Risk factors for EH and EC differ in relation to cytological atypia and architectural crowding [1,
reproductive factors. Parity is found to be protec- 18, 21, 22]. Simple hyperplasia represents the
tive for EC but not for EH [14]. Long duration of lowest risk of cancer progression, and the major-
oral contraceptive use has some protective effect ity spontaneously regress [1]. Among 18% of
[14, 15]. persistent lesions, there are 8% and 3%, rates of
progression to simple atypical hyperplasia and
complex atypical hyperplasia and only 1% prog-
3.3 Risk of Endometrial ress to EC. Complex hyperplasia is reported to
Carcinoma have an intermediate risk of progression with
22% persistent and 4% progression to EC, with a
EH is a pathologically diversified lesion which mean duration to progression of approximately
encompasses histological subtle and spontane- 10 years. Therefore, both simple hyperplasia and
ously reversible proliferative lesions to emerging complex hyperplasia are not recognized as pre-
EC. Women with atypical hyperplasia may have neoplastic forms [2].
coexistent EC or may progress to carcinoma [18]. Another study reported progression to EC in
1%, 3%, 8%, and 29% of patients with simple
hyperplasia, complex hyperplasia, simple atypi-
3.3.1 Coexistent Carcinoma cal hyperplasia, and complex atypical hyperpla-
sia, respectively [23].
EC is found more frequently in women with The lower risks of progression to EC in
cytological atypia. Recent studies have shown women with EH without atypia can help in
28 B. M. Patel

decision-­making for conservative management, complex atypical hyperplasia. The latest and
whereas the higher risks of atypical hyperplasia fourth classification published in 2014 is the most
progressing to EC required consideration of commonly recognized system with the reduction
aggressive approaches [19]. Meticulous knowl- to two categories only, and it reflects a new con-
edge of rates of progression risks for EH to EC cept of molecular genetic changes. Hyperplasia
encourages better clinical management of EH without atypia (Figs. 3.2 and 3.3) expresses no
[18, 21–24]. associated genetic changes. They represent
Thresholds for distinguishing precursors (e.g., benign changes and disappear after the endocrine
atypical hyperplasia or EIN) from carcinoma in background returns to normal. Conversely, in
biopsies vary among pathologists and by the clas- atypical hyperplasia (Figs. 3.4 and 3.5), expres-
sification system, but all of these lesions warrant sions of cellular and genetic changes typical of
close follow-up. A bigger challenge may be how EC (Figs. 3.6 and 3.7) are found. The diagnosis
to evaluate and manage the larger group of of EIN in the WHO 2014 classification is inter-
women with less severe abnormalities [22]. changeable with atypical hyperplasia [5]. This
new classification constitutes an important sim-
plification for clinical practice particularly with
3.4 Classification concern to management options: hyperplasia
without atypia can be treated usually conserva-
Among several histological classification sys- tively, while treatment of atypical hyperplasia/
tems proposed for EH since 1963, two prominent EIN is usually total abdominal hysterectomy
classifications are commonly used for EH at (TAH) [1, 7, 25].
present: the World Health Organization (WHO)
classification and the endometrial intraepithelial Gross manifestations of EH are highly differ-
neoplasia (EIN) classification (Fig. 3.1). ent. Changes in endometrium range from dif-
fusely thickened (5–10 mm or greater) to vaguely
WHO Classification: It was established in 1994 nodular, tan, and soft without hemorrhage or
and classified EH in four categories, simple necrosis. EH may be focal or multifocal in rela-
hyperplasia without atypia, complex hyperplasia tion to cycling endometrium and polyp or on the
without atypia, simple atypical hyperplasia, and diffusely thin endometrium [1, 26–28].

1994 WHO classification 2014 WHO classification EIN classification

Category Category Histopathological features Category

Simple hyperplasia
without atypia Variability in gland size and
shape
Hyperplasia without atypia Endometrial hyperplasia
Cystic glands
Complex hyperplasia Increased
without atypia gland to stroma ratio

Simple atypical hyperplasia Architectural irregularity


of gland
Endometrial intraepithelial
Atypical hyperplasia / EIN Glandular crowding with
neoplasia
little intervening stroma
Complex atypical hyperplasia Nuclear atypia

Fig. 3.1 Schematic representation of 1994 WHO classification, 2014 WHO classification, and EIN classification [7]
3 Endometrial Hyperplasia: Diagnosis and Management 29

Fig. 3.2 Hyperplasia


without atypia (low
power 10×): Increased
gland-to-stroma ratio is
evident. Glands are
mildly crowded and
dilated

Fig. 3.3 Hyperplasia


without atypia (high
power 40×): Glands are
lined by columnar
epithelium and do not
show any atypia

Fig. 3.4 Hyperplasia


with atypia (low power
10×): Crowded back to
back glands are evident
with little intervening
stroma
30 B. M. Patel

Fig. 3.5 Hyperplasia


with atypia (high
power). Cells show loss
of polarity. Nuclei are
enlarged with irregular
nuclear membrane,
coarse chromatin, and
prominent nucleoli.
Atypical mitoses are
evident

Fig. 3.6 Endometrial


carcinoma (low power
10×): Tumor nests
invade the myometrium

Fig. 3.7 Endometrial


carcinoma (high power
40×): Tumor cells with
glandular confluence,
complex architecture,
and lack of intervening
stroma. Individual cells
show high N/C ratio,
pleomorphism,
prominent nucleoli, and
atypical mitosis
3 Endometrial Hyperplasia: Diagnosis and Management 31

EIN Classification It is an alternative classifica- activity (such as decidua basalis or polyps) can be
tion which is based on molecular, morphometric, confused with both the WHO (i.e., as EH) and EIN
and morphologic data and has been proposed by (i.e., as EIN) classifications. Due to sampling
the International Endometrial Collaborative Group errors with endometrial biopsy (EB) specimens
in 2000. It is developed to improve prediction of and the uncertain natural history of endometrial
clinical outcome, improve inter-observer reproduc- precursors, it is difficult for any classification to
ibility, and reduce subjective bias inherent to 1994 have both high sensitivity and specificity [22].
WHO classification [7, 13, 29]. In this, nonatypical
anovulatory or prolonged estrogen-exposed endo-
metrium is classified as EH. The criteria for diag-
nosis of EIN are as follows [11, 22, 27, 30]. 3.5 Clinical Presentation

1. Size of the lesion is at least 1 mm Women with EH are diagnosed by EB performed


2. Glandular volume is more than the stromal for AUB including menorrhagia, intermenstrual
volume bleeding, irregular bleeding on HRT or tamoxi-
3. Changes in cytology in relation to fen, and postmenopausal bleeding (PMB).
background Consequently, it is the most common symptom
4. Exclusion of benign mimics like endometrium for EH. It is common in perimenopausal, in early
in secretory phase, effect of exogenous estro- postmenopausal, or with increasing age in pre-
gen and polyps and malignancy menopausal women. EH accounts for approxi-
mately 15% of women with PMB [2, 12]. In
The EIN classification can be assigned using asymptomatic women, EH is detected acciden-
computer-assisted morphometric analysis, which tally, when workup is done for prolonged HRT
provides a D-score or diagnostic criteria that can be use or cervical cytology demonstrating endome-
applied to standard hematoxylin and eosin (H&E) trial/abnormal glandular cells. The age at presen-
stained slides subjectively by pathologist [22, 27, tation relies on the source of excess estrogen. EH
28]. Prior to the formation of EIN classification, it secondary to anovulation at menarche is uncom-
was believed that unopposed estrogenic stimulation mon and easily reversible [3, 4].
will lead to EH. The EIN classification suggests that
the initial event in EH is a genetic alteration, and
eventually it separates two events: mutational acti- 3.6 Diagnostic and Surveillance
vation and estrogenic stimulation [7]. The EIN sys- Methods
tem has not gained widespread acceptance, most
likely due to cost and/or lack of experience with the Management of EIN requires its accurate diagnosis
computerized D-scoring component [3, 4]. and exclusion of coexistent carcinoma [11].
Women with symptoms suspicious for EH are eval-
Comparison Between WHO and EIN uated at first with physical examination. Diagnosis
Classification: Although EIN classification cate- of EH by cytology is generally unsatisfactory [10].
gories do not correspond directly to particular cat-
egories in the WHO 1994 classification, there is Histopathological Diagnosis: Diagnosis of EH
some distinguishable overlap [7, 25]. The EIN needs histological confirmation of the endome-
classification may better arrange the distorted cel- trial tissue obtained by different techniques which
lular architecture and nuclear characteristics, but include office EB, D&C, and directed EB by hys-
adequate comparative studies are lacking. The teroscopy. Other investigations include transvagi-
WHO classification is more widely used. The risk nal sonography (TVS) and saline ­ infusion
of progression to endometrial carcinoma using the sonography [2, 31]. Apart from diagnosis, endo-
WHO and EIN classification was found to be simi- metrial sampling is also required in monitoring
lar [4, 11, 22]. Lesions that cause glandular over- regression, persistence, or progression [13].
32 B. M. Patel

Office EB technique is usually performed


using the several commercially available devices
(e.g., Pipelle, Vabra aspirator, Gyno Sampler)
and is convenient and safe and has high accuracy
for diagnosing EH or EC. It has replaced the pro-
cedure of D&C for the diagnosis of EH or EC [3,
32]. Despite a negative biopsy result, 2% of
women will still have EH [2, 3, 13]. Mass lesions
that encroach on the uterine cavity, for example,
polyps or uterine fibroids, may divert Pipelle,
which is flexible, preventing sufficient evaluation
of the endometrial cavity. In this situation, EB
may be performed by a rigid curette during D&C
[11]. Office EB can cause some discomfort, and
in approximately 8% of patients, it is not possible
to perform due to stenotic cervical os [20]. Both
Pipelle and D&C are blind sampling techniques
and are not representative of the entire endome- Fig. 3.9 Endometrial hyperplasia on hysteroscopy
trial cavity [2]. If hysterectomy is planned for the
woman for any reason, then the method of sam-
pling is less important. The accuracy of D&C
compared with Pipelle EB in diagnosing a EH,
and excluding concurrent EC, is not clear. Both
have been reported to have equal rates of cancer
detection in patients with AUB [2, 11]. In one
meta-analysis, EB with Pipelle was found to be
superior to other sampling techniques in the
detection of EC and atypical hyperplasia [32].
Diagnostic hysteroscopy is used to examine the
entire surface of the endometrium and biopsy sus-

Fig. 3.10 Hysteroscopy demonstrating endometrial


polyp in the background of atrophic endometrium in
woman on tamoxifen

picious or focal lesions under direct visualization


(Figs. 3.8, 3.9, and 3.10). It is especially important
when an outpatient sampling fails or is inconclu-
sive, if abnormal bleeding persists or if intrauter-
ine structural abnormalities such as polyps or other
discrete focal lesions are suspected on TVS [2, 13,
33]. Office hysteroscopy is conducted in the out-
patient setting using miniature hysteroscope and
Fig. 3.8 Normal endometrium on hysteroscopy without the requirement of anesthesia or vaginal
3 Endometrial Hyperplasia: Diagnosis and Management 33

instrumentation. Hysteroscopy is more precise in


detecting than excluding endometrial lesions and
has a higher precision for EC than EH [13].
Hysteroscopy with targeted biopsy or D&C
compared to hysteroscopy performed alone has
very good sensitivity and specificity for detecting
EH. The combination of hysteroscopy with D&C
or EB is considered to be a superior diagnostic
tool compared with hysteroscopy, D&C, or EB
performed alone [2].
Practical aspects of diagnosis: Problem in
reproducibility of detection of atypical hyperpla-
Fig. 3.11 Endometrial hyperplasia as imaged by trans-
sia: Diagnostic reproducibility is limited by the vaginal sonography in postmenopausal woman.
size of the lesion, inadequate sample, poor quality Endometrial thickness 13.6 mm
of fixation, processing, and staining of tissues [5,
6]. Current diagnostic strategy should include
evaluation of sample adequacy, as is recommended
for evaluating cervical cytology [5, 9, 11]. There is
need to apply multiple diagnostic criteria, and due
to imperfect sampling, only few of them may be
present in a given specimen. In short, sample or
lesion size may be a factor responsible for inaccu-
rate assessment of risk [11]. It is important to
remember that atypical hyperplasia is frequently
associated with concomitant adenocarcinoma and
low level of reproducibility associated with the
diagnosis of atypical hyperplasia in EB [6].
Awareness of the artefactual changes generated Fig. 3.12 Thickened endometrium as imaged by transab-
by EB is necessary to ensure that EH is not overdi- dominal sonography in unmarried patient on tamoxifen
agnosed [15]. When the woman is on exogenous (longitudinal view). Endometrial thickness 27 mm
progestin, it is better to perform EB after with-
drawal of hormones. If EB specimen is compro- Ultrasonography: TVS, a noninvasive diagnos-
mised by sampling errors or regenerative epithelial tic method, is of proven value for evaluation of
changes, either instant additional EB is done or endometrial thickness (ET) and contour as a part
regular follow-up with repeat EB is done after of investigation for PMB [2, 34] (Figs. 3.11 and
6 months to detect the presence of pathology in the 3.12). It can detect an irregularity of the endome-
endometrium. Pathologists must develop their own trium or an abnormal double-layer ET measure-
approach to differentiate between proliferative ment. These findings will guide the clinician to
endometrium and EH especially in women with determine which women should undergo EB with
anovulation or unopposed estrogen exposure [29]. PMB [13, 33]. Meta-analysis of 5892 symptom-
An important issue is the availability of a atic women with PMB in 35 published studies
pathologist experienced in gynecological pathol- demonstrated that an ET of 5 mm or more identi-
ogy. As mentioned, there are a range of abnor- fied 95% of those with EH and EC. In contrast,
malities seen in hyperplastic endometrium, and among them, a woman with an ET of less than
differentiation between them can be quite diffi- 4 mm only had a 1% probability of EC. This
cult. Only those with frequent exposure to such ­cutoff did not differ remarkably between women
specimens are likely to be skilled in interpreting with and without HRT [2, 9, 11, 34]. However,
these lesions [9]. other systematic reviews have recommended a
34 B. M. Patel

cutoff of 3 mm or 4 mm to rule out EC. By using evaluating use of CT scan for follow-up of a
this cutoff value, the probability of diagnosis of woman with EH treated conservatively. CT scan
EC is reduced to less than 1%. A larger cutoff is an expensive test and is not routinely advo-
value has been recommended for women on HRT cated because of the radiation exposure associ-
or tamoxifen presenting with AUB or asymptom- ated with it. Diffusion-weighted MRI has the
atic woman with thickened endometrium on TVS future potential to diagnose EH and other endo-
[13]. ET more than 1 cm on TVS in postmeno- metrial lesions. It may be a useful imaging for
pausal women is associated with an increased follow-up of a woman with atypical hyperplasia
risk of EC [34]. Overall, assessment of endome- as a predictor for malignant change, but more
trial thickness on TVS is of value in mainly post- evidence is needed [13].
menopausal women as there are no cutoff values
for endometrial thickness in premenopausal Immunohistochemical Biomarkers: Prediction
women in whom normal ET can be similar to that of cases which will progress to EC can increase
with EC [11]. The role of ultrasonography (USG) with the use of several immunohistochemical
in premenopausal women is restricted to identi- biomarkers. In some instances, such as for
fying structural abnormalities, as there appears to women with hereditary nonpolyposis colon can-
be an overlap between normal ET and that caused cer, biomarker may have a role in diagnosis. To
by endometrial disease. Royal College of date, not a single biomarker is found to merit its
Obstetricians and Gynaecologists (RCOG) guid- clinical utility [2, 11, 13].
ance recommends TVS for women with polycys-
tic ovarian syndrome (PCOS) or AUB. In a
woman with PCOS, possibility of EH is less 3.7 Differential Diagnosis
likely if ET is less than 7 mm [13]. It is recom-
mended that both TVS and saline infusion sonog- The differential diagnosis of EH includes other
raphy should be performed simultaneously with conditions that present with AUB. Confirmation
EB [2]. Occasionally a palpable adnexal mass of the source of bleeding is the most important
with solid features on USG should raise the pos- step in evaluation of women with AUB, and
sibility of a coexistent granulosa cell tumor. It is bleeding from any other part of the genital tract,
responsible for EH in 40% of cases due to exces- anus, or rectum should be excluded. In women
sive estrogen production [12, 13]. with abnormal cervical cytology, the differential
diagnosis includes benign endometrial and cervi-
To summarize, a woman with risk factors for cal neoplasia [4].
EC and with ET more than 5 mm after meno- Even at the level of histopathology, it has to be
pause should undergo EB. However, routine differentiated from atrophic or weakly prolifera-
screening for women at high risk of EH has not tive endometrium with the architecture of
proven successful or cost-effective except for all hyperplasia, endometrial metaplasia, and EH
­
Lynch syndrome mutation carriers where annual with superimposed secretory changes well-­
EB is recommended by National Comprehensive differentiated adenocarcinoma [15, 20, 35].
Cancer Network (NCCN) guidelines. An inade-
quate EB is an indication for further investiga-
tion. Even if an office EB is adequate and reported 3.8 Management Options
as negative, additional evaluation with TVS and/
or D&C/hysteroscopy should be done if symp- The choice of management for the woman with
toms persist or recur [2, 13]. EH is determined mainly by woman’s age, health,
Computerized tomography (CT) and desire for fertility, as well as the risk factor for
diffusion-­weighted magnetic resonance imaging progression to EC and the type of EH [1, 9, 33,
(MRI) in the diagnosis and management of EH 36]. Nuclear atypia is the main factor, and older
are not commonly used. There are no studies age, obesity, and ovulatory dysfunction are
3 Endometrial Hyperplasia: Diagnosis and Management 35

a­ dditional risk factors in determining the risk for ment, who developed EH, should be reviewed
coexistent or progression to EC [4, 37]. EH with- regarding continuation or change of medicine in
out atypia is usually treated with hormone ther- conjunction with her treating oncologist. A base-
apy. Younger women who desire fertility can be line USG is indicated which also helps to rule out
treated by medical management, regardless of the estrogen-secreting granulosa cell tumor of the
type of EH [1]. However, perimenopausal and ovary [1, 37].
postmenopausal women having EH with atypia However, the woman should be informed that
or symptomatic women having EH without the treatment with progestogen has higher rates
atypia are treated by surgical management unless of regression of EH compared with observation
contraindicated. Regardless of the type of EH, alone. When a woman with EH fails to regress
the woman with risk factors for recurrence or following observation alone for 12 months, pro-
progression to EC requires active management gestogen treatment is required. Observation
and surveillance [1, 9, 30]. alone in the symptomatic woman with AUB is
All management options should be accompa- rarely advised [1, 12, 13]. In view of a high spon-
nied by removal of the exogenous or endogenous taneous regression rate and a very low rate of
source of estrogen exposure as it is the main fac- progression to more severe disease, it is uncertain
tor contributing to EH. This may be done by dis- whether medical management is appropriate for
continuation of ERT without progestin, treatment all women with EH without atypia [1].
of ovulatory dysfunction (e.g., PCOS or hyperp-
rolactinemia), weight loss in obese women, or
even by removal of estrogen-producing tumor 3.8.2 Medical Management
(e.g., granulosa cell tumor) [37].
Hormonal therapy used in the management of EH
includes progestins, selective estrogen receptor
3.8.1 Observation modulators, aromatase inhibitors, sulfatase
inhibitors, gonadotropin-releasing hormone
In the woman with EH without atypia, spontane- (GnRH) antagonists, synthetic androgen (dan-
ous regression usually occurs during follow-up, azol), metformin, and protein-tyrosine kinases
and the risk of progression to EC is less than 5% inhibitor (genistein) [1, 11] (Table 3.2).
over 20 years [1]. Observation alone can be con-
sidered if the risk for coexistent or progression to 3.8.2.1 Progestin Therapy
EC is low and reversible risk factors are identified Progestins, which are synthetic progestogens
and treated. The slow progression of EH without mimicking natural progesterone, are used most
atypia to EC offers a window of opportunity to frequently to induce regression of endometrial
manage these reversible risk factors [1, 12]. hyperplasia in women with EH without atypia,
Anovulatory cycles can lead to EH especially those who desire fertility, those who refuse sur-
in a woman with PCOS or in a premenopausal gery, or those who have contraindications to sur-
woman. Once a woman with PCOS resumes ovu- gery due to significant medical comorbidities [1,
lation or perimenopausal woman reaches meno- 11, 19]. Therapeutic aims of progestin therapy are
pause, regression of EH occurs. A detailed history complete regression of EH, return to normal endo-
regarding the use of exogenous hormones (e.g., metrial function, and the prevention of EC [11].
long-term use of HRT, unopposed ERT without Progesterone brings about secretory changes
progestin) should be elicited. The indication and in the normal endometrium. It produces these
type of HRT should be reviewed especially in effects by increasing the catabolism of estrogen
relation to dose and mode of administration. receptors and increasing the activity of enzymes
Change in the dosage of HRT or discontinuation 17-b-hydroxysteroid dehydrogenase and sulfo-
is often sufficient to induce regression of EH transferase thereby decreasing estrogen levels [1,
without atypia. The woman on tamoxifen treat- 19]. It causes apoptosis leading to decreased
36 B. M. Patel

Table 3.2 Drugs used in medical management of endometrial hyperplasia


Type of therapy Routes of administration
Progestin therapy
 Megestrol acetate Oral
 Medroxyprogesterone acetate Oral/injection
 Norethisterone acetate Oral
 Micronized progesterone Oral/vaginal pessary
 Levonorgestrel Oral/implant/intrauterine insert (IUD)
Therapy other than progestin
 Aromatase inhibitor Oral
 Metformin Oral
 GnRH therapy Injection/implant/nasal spray
 Danazol Oral
 Genistein Oral
Combination therapy
 Megestrol acetate + metformin Oral
 LNG-IUD + metformin IUD + oral
 LNG-IUD + GnRh agonist IUD + injection/implant/nasal spray

glandularity and inhibits angiogenesis in the Table 3.3 Different types of progestin therapy for treat-
myometrium. Eventually, this causes sloughing ment of endometrial hyperplasia [1, 11, 37]
and thinning of the endometrium [1, 11]. Type of progestin Dose
Contraindications to progestin therapy include Megestrol acetate 40–200 mg daily in
current or past history of thromboembolic disor- divided doses or
10 mg × 14 days/month
ders/stroke, severe liver dysfunction, known or sus-
Medroxyprogesterone 10–20 mg daily or
pected malignancy of progesterone receptor acetate 10–20 mg × 14 days/
(PR)-positive breast cancer, vaginal bleeding of month
unknown etiology, pregnancy, and known allergic Depot 150 mg intramuscularly
reaction to progestins [37]. Various routes of admin- medroxyprogesterone every 3 months
acetate
istration can be used – oral, intramuscular, and vagi-
Micronized vaginal 100–200 mg daily or
nal routes or through intrauterine devices [1]. progesterone 100–200 mg × 14 days/
Unfortunately, optimal progestin regimen and month
duration have not been investigated, and posttreat- Levonorgestrel IUD 20 μg/day × 1–5 years
ment long-term follow-up has not been reported Norethisterone acetate 15 mg daily
adequately [20, 38]. Sixty-one percent of women
on estrogen-only replacement therapy and atypical
hyperplasia responded to progestin therapy and progesterone-like and antigonadotropic
were cured [1]. The response to progestin therapy effects [1]. It is considered a “chemotherapeu-
usually begins at 10 weeks and is complete by tic agent” but best classified as a strong pro-
6 months. Cyclic progestin has a low regression rate gestin. Dosages vary between 160 and
for EH, compared to continuous oral progestin or 320 mg/day. At these doses, the beneficial
LNG-IUD [12, 13, 39]. Prognostic factors include effects on endometrium are maximum with
low gland-to-stroma ratio, low mitotic activity, loss minimal effects on blood glucose levels or
of cytologic atypia and other changes in histology, lipid profile [1].
cytoplasm or architecture [11]. Progestins used and b. Medroxyprogesterone acetate (MPA)—It is a
their dosages are shown in Table 3.3. synthetic steroidal progestin commonly used
in hormone replacement therapy in postmeno-
a. Megestrol acetate (MA)—It is a steroidal pro- pausal women in whom its use helps in
gestin and is effective in EH because of its ­prevention of EH. The dose commonly given
3 Endometrial Hyperplasia: Diagnosis and Management 37

is 10 mg/day continuously for 6 weeks or low systemic levels of progestin. Therefore, it


2 weeks/month for 3 months (safer and more has an effect on the endometrium several
acceptable than continuous therapy). If the times stronger without causing side effects
response is not complete, the therapy can be such as breast tenderness, mood changes, and
continued for the next 3 months [1]. weight gain. In addition to higher efficacy, it
c. Norethindrone acetate/norethisterone acetate offers long-acting contraception, does not
(NETA)—It is a synthetic, orally active steroi- require daily dosing, and is better tolerated
dal progestin with antiandrogenic and anties- when compared to oral progestins [11, 39,
trogenic effects. It has been shown to reduce 41]. Other limitations of LNG-IUD are a 1 in
EH in postmenopausal women on HRT [1, 1000 uterine perforation risk and invasive
37]. The recommended dosage is 15 mg/day. placement in the uterus [39]. There is no sig-
d. Micronized progesterone—It is a relatively nificant difference in rates of irregular vaginal
weak progesterone and usually not recom- bleeding with the LNg 52/5 compared with
mended for first-line treatment of EH. In doses oral progestins [37].
of 200–300 mg daily, it is reserved only for Oral progestins are preferred over LNG-­
women who are at low risk for progression and IUD in women who refuse or cannot tolerate
cannot tolerate stronger synthetic progestins or an IUD because of side effects (e.g., dysmen-
refuse levonorgestrel intrauterine device orrhea), women with uterine factors that make
(LNG-IUD). Till date, there are no studies on placement or retention of an IUD difficult
the use of vaginal micronized progesterone for (e.g., severe distortion of the uterine cavity
the treatment of EH. Theoretically, it can be due to fibroids or congenital anomaly), and
used as maintenance treatment, as high endo- women who plan to conceive as soon as a
metrial concentrations may be gained due to complete therapeutic response is achieved.
local effects [37]. Progestins are contraindicated in pregnancy,
e. Levonorgestrel (LNG)—It is a second-­ and the patient can stop an oral medication
generation progestin (synthetic progestogen) without requiring a clinician to remove the
and the intrauterine device containing LNG is device, as with an IUD [37].
an attractive option for managing EH. It f. Progestin injections and implants—Depot
releases a constant amount of LNG inside the medroxyprogesterone acetate (DMPA) is a
uterus and effectively opposes the estrogenic long-acting progestin, provides contraception,
effect [1, 11, 40]. The LNG 52/5 starts with an and requires only four injections per year. It
initial dose of 20 mcg/day, and by 5 years the has not been well studied for the treatment of
daily dose is approximately 10 mcg/day [37]. EH. In one study, the intramuscular DMPA
The LNG-IUD initially results in irregular (150 mg every 3 months) was found to be
bleeding as with other progestin-only therapy, more successful than NETA (15 mg daily for
but eventually, most women become amenor- 14 days/cycle) in the treatment of nonatypical
rheic or have light tolerable bleeding. For the EH. Regarding side effects, nausea and breast
best outcomes, medical treatment of EH discomfort were more with NETA, while
should require LNG-IUD use for up to 5 years amenorrhea was more with DMPA [37].
[12, 13, 39]. LNG-IUD is also available in Common side effects of progestins include
lower daily doses (13.5, 17.5, and 18.6 mcg/ weight gain, headache, nausea, vomiting,
day) and varies from 3- to 5-year formula- menstrual irregularities, and sometimes hyper-
tions, but these have not been studied in tension and depression. The incidence of
women with EH to determine whether the venous thrombosis and embolism may also be
lower progestin dose is as effective as the LNg slightly increased [1, 9, 20]. In addition to sys-
52/5 [37]. temic side effects, oral progestins are associ-
Comparison of oral progestins with LNG-­ ated with poor compliance that may limit its
IUD—LNG-IUD has high intrauterine but overall efficacy [41]. Bothersome side effects
38 B. M. Patel

may require an adjustment in dose or a switch especially helpful in obese women in whom it
to a different progestin therapy. For women on helps to decrease weight and thereby causes
systemic progestins, change over to the LNG-­ decrease in peripheral conversion of androgen
IUD can be considered [37]. to estrogen and better response to progestins
Almost 12–53% women with EH fail to [1, 38, 41]. Metformin is now being studied in
respond to progestin therapy [1, 11]. Response combination with LNG-IUD and MA [1, 39].
to progestins depends not only on patient’s age c. GnRH therapy—GnRH agonists inhibit estro-
and the type and grade of hyperplasia but also gen by inhibiting the hypothalamic pituitary
the number of PR and activity of co-activators ovarian axis thereby exerting antiproliferative
and co-repressors, insulin resistance, and effect on the endometrial cells. Women with
altered activity of TGF-α and EGFR in endo- EH, both with and without atypia, can be given
metrial glandular cells [1]. Rarely, resistance to GnRH at a dose of 1 ampule/3.75 mg intra-
progestin therapy could be a result of paracrine muscularly monthly for 6 months [1, 9]. In a
effects. The histologic response of the glands of study using GnRH and tibolone (a synthetic
atypical EH/EIN is strongly coupled to the steroid with both estrogenic and progestagenic
decidual response in the stroma, so the possibil- effects) for treatment of EH, it was seen that
ity of a paracrine effect is convincing [11]. though a complete response was seen in all
Hence, regular follow-up and EB are recom- patients, recurrence occurred within 2 years in
mended for patients while on progestin therapy 19% after cessation of therapy [1]. In another
[1]. Recently, the role of HE4 as a novel tissue study, GnRH agonists and LNG-IUD were
marker for predicting therapy response and used in combination with a release rate of 19.5
progestin resistance in EH has been studied and mcg/day for 5 years (Mirena; LNg52/5) to suc-
proved to be effective in it [42]. cessfully treat 24 premenopausal women with
either atypical EH or early-stage EC [37].
3.8.2.2 Therapy Other than Progestins d. Danazol—It is a synthetic androgen which
a. Ovulation induction—In the reproductive-age causes atrophy of the endometrium through its
group, ovulation induction done with clomi- ability to produce a hypoestrogenic and
phene or aromatase inhibitors will lead to cor- hypoandrogenic state [1, 9, 36]. It has been
pus luteum formation, exposure to endogenous proven to be effective in treatment of EH with
progesterone, and resolution of EH in some a relapse rate of approximately 8 to 9% [1, 9].
women. Pregnancy is highly unexpected in the The side effects of oral danazol (weight gain,
setting of ongoing EH. Careful surveillance is acne, hirsutism, and muscle cramps) are the
needed to confirm EH regression. This limiting factors in its use for EH which can be
approach is recommended for women with EH curtailed to some extent by using danazol-­
without atypia who desire pregnancy [37]. containing IUD [1, 37].
b. Metformin—EH is associated with obesity, e. Genistein—It is an isoflavonoid extracted
metabolic syndrome, PCOS, insulin resis- from soy products. It decreases estrogen level
tance, and type II diabetes which directly have by inhibiting protein-tyrosine kinases and
a mitogen effect on the endometrium. topoisomerase-II. It has still not been estab-
Metformin (N,N-dimethylbiguanide) is a big- lished for management of EH till the time
uanide and decreases gluconeogenesis in the more clinical trials are conducted [1].
liver which in turn decreases insulin resistance
[41]. Long-term progestin therapy causes
decreased level of progesterone receptors. 3.8.3 Surgical Management
Metformin induces PR expression in endome-
trial cells which helps to overcome resistance Currently, surgical options include hysterectomy
to progestin therapy [1]. Metformin is also with or without bilateral salpingo-oophorectomy
3 Endometrial Hyperplasia: Diagnosis and Management 39

(BSO) by abdominal, vaginal, and minimally 3.9 Management Algorithm


invasive procedures (such as laparoscopic or According to the Type
robotic approach). Total extrafascial hysterec- of Endometrial Hyperplasia
tomy is the procedure of choice providing a (Fig. 3.13)
definitive assessment of a coexistent EC and
effectively treating EH. Supracervical hysterec- 3.9.1 Woman with EH Without
tomy is unacceptable due the potential for local Atypia
extension of endometrial neoplasia into the cer-
vix and hence risk of leaving behind residual dis- Premenopausal women—The initial step in the
ease [11, 13]. Morcellation of the uterus should management of EH is the identification of risk
be avoided due to risk of dissemination of coex- factors and removal of the exogenous and endog-
istent EC [13]. Disadvantages of vaginal hyster- enous source of unopposed estrogen. The pre-
ectomy include technical difficulty in removing menopausal woman with EH without atypia can
the ovaries and comprehensive surgical staging, be managed with observation or low-dose pro-
if required, is not feasible [11]. During the hys- gestins for 3–6 months. Hysterectomy can be
terectomy, gross inspection in routine and frozen considered in women aged >40 years [1, 37].
section especially in high-risk cases should be Options for progestin therapy are oral proges-
performed to evaluate for EC [37]. Discrepancy tins, LNG-IUD, or combined estrogen and pro-
between the frozen-section interpretation of gestin oral contraceptives (OCs). Choice of
endometrial tissue and the final diagnosis based therapy is also according to the need for contra-
on permanent section is problematic. Despite ception of patient since many oral progestins do
preoperative endometrial sampling and intraop- not provide it [37].
erative evaluation, some women with atypical If there is a good response after progestin ther-
EH will have EC detected only on final pathol- apy, i.e., menstrual pattern has normalized, annual
ogy evaluation [13, 37]. Regardless of surgical EB is advised [1, 17]. EB can be performed even
approach, women with high-risk factors should with IUD in place [1, 33]. Some authors prefer
be explained regarding the need for additional waiting for a withdrawal bleed before EB, while
staging surgery if an EC is identified. Following others perform EB while the patient is on proges-
a total hysterectomy, if there is no EC in the tin therapy. The rationale behind this dilemma is
specimen, no further surveillance for EH is the decidual reaction that occurs with progestin
­necessary [11]. therapy making it more difficult to interpret
For women undergoing hysterectomy as treat- pathologic findings [29, 37]. If the bleeding pat-
ment for atypical EH, the choice of bilateral BSO tern does not normalize, high dose of progestins
should be decided after weighing the risk of pre- can be considered after performing repeat EB. If
mature menopause and potential risks of oopho- office EB demonstrates abnormal report, D&C or
rectomy versus the risk of a second surgery if EC hysteroscopy-directed biopsy is mandatory to rule
is found postoperatively. Some women may prefer out coexistent EC or more severe EH. It was
to undergo bilateral salpingectomy alone instead reported that the median time to complete
of oophorectomy, for possible prevention of ovar- response of EH to progestin was 6 months [1].
ian, fallopian tubal, or peritoneal cancer (level 3 or The best plan for follow-up is undetermined.
4 evidence) [11, 13]. Endometrial ablation using Follow-up schedules should be individualized. In
thermal or electric cautery device is not recom- women with high risk for EH relapse, persistence,
mended for the treatment of atypical EH/EIN. No or progression, long-term annual follow-up is rec-
methods are available to confirm the completeness ommended [12, 13]. If repeat EB shows atypical
of ablation. Moreover, because of subsequent EH or persistent EH without atypia despite high
adhesions, the cavity may be partly inaccessible progestins or if symptoms persist, hysterectomy
for surveillance after ablation [11, 13]. can be considered [1, 13].
40 B. M. Patel

Abnormal Uterine Bleeding

Clinical Examination
TVS
EB(Office /D&C/Hysteroscopy)

EH without atypia EH with atypia/EIN

Pre Post
Menopausal Menopausal Pre Menopausal / Post Desires Fertility
Menopausal

High dose
Address Hysterectomy Contraindication Hysterectomy progestins
risk factors to surgery/ Refuses
surgery
No response Good response
Observation Low dose Hysterectomy
progestins (if age > 40 years) Low dose
progestins Hysterectomy Annual EB
No response Good response
No response

High dose Annual EB


progestins High dose
progestins

No response
No response
Hysterectomy
Hysterectomy

Fig. 3.13 Management algorithm for endometrial hyperplasia

Postmenopausal women—Hysterectomy is 3.9.2 Women with Atypical


usually recommended in the postmenopausal Hyperplasia/EIN
woman with EH without atypia, especially with
risk factors for EC or with contraindications to Women with atypical hyperplasia have a high
progestin therapy. If there are contraindications risk of coexistent EC or progression to
to surgery or patient refuses surgery, progestin EC. Hysterectomy is curative and the treatment
alone is recommended. OCs are not preferable as of choice in majority of women who have com-
these women do not need contraception and also pleted childbearing, do not desire preservation of
to avoid unnecessary exposure to estrogen [1, their fertility, or do not respond to hormone
37]. If regression to normal endometrium does ­therapy [11, 13]. If diagnosis of atypical EH is
not occur after 6 months of progestin therapy, the made by office EB, further evaluation with D&C/
patient may be treated with higher dose of oral hysteroscopy is recommended to exclude
progestin or combination of LNG-IUD and oral ­coexistent EC [37].
progestin after excluding atypical EH or coexis- Women who desire fertility or have contrain-
tent EC by EB (D&C/hysteroscopy). In the set- dication to surgery—Women with atypical EH
ting of persistent EH, hysterectomy is the who desire fertility or have contraindication to
standard treatment approach [1, 37]. surgery can be treated with progestin therapy.
3 Endometrial Hyperplasia: Diagnosis and Management 41

Women should be extensively counseled regard- Management of endometrial hyperplasia with


ing the risks including a lack of response or ongoing tamoxifen use for breast cancer treat-
even progression or coexistent EC during hor- ment: Women should be informed about increased
monal therapy. These discussions and patient risk of EH and EC with tamoxifen use and coun-
understanding should be reflected in informed seled for prompt reporting of AUB. In the pres-
consent. Compliance with medical therapy and ence of EH, use of tamoxifen is reassessed in
adequate and regular follow-up with EB are a consultation with medical oncologist and alterna-
prerequisite for medical management [13, 33]. tive drug is sought. Aromatase inhibitors are
Management is planned after reviewing clinical studied as an alternative treatment option without
evaluation, histology, and imaging in multidis- increasing the risk of EH and EC. Prophylactic
ciplinary meeting. LNG-IUD is preferred, as it progestin therapy is not recommended in women
is easy to comply with, is well tolerated, and has on tamoxifen [13].
a regression rate to normal endometrium in
about 90% of cases. Among oral progestins,
megestrol acetate is preferred for management 3.10 Prevention of Endometrial
of atypical EH in dose of 80 to 160 mg twice per Hyperplasia
day. It is more potent than MPA. Median time
for regression of atypical EH to normal endo- In women with AUB, it is appropriate to evaluate
metrium on progestin therapy is usually 6 to woman past their fourth decade of life. Women
9 months. If no EH is detected on subsequent with an intact uterus should never be prescribed
EB, a woman can be allowed to consider for ERT as this increases the risk of EC. For women
natural conception. A decision for assisted on exogenous estrogens, addition of progestin
reproduction immediately after stopping of pro- may prevent EH. Another preventive measure is
gestogen treatment should be made with consul- periodic treatment with a progestin to produce
tation of a multidisciplinary team weighing scheduled withdrawal bleeding in the amenor-
between risks of disease progression and fertil- rhoeic or hypermenorrheic perimenopausal
ity prospects. In women with atypical EH, hys- women with fluctuating levels of estrogen [27].
terectomy should be recommended once fertility
Key Points
is no longer required because of the high rates
of EH recurrence and the potential for EH pro-• A thorough history (age at menarche and
menopause, parity, history of infertility, his-
gression [13]. If there is persistent atypical EH
or coexistent EC on subsequent EB, hysterec- tory of dose and duration of HRT and tamoxi-
tomy is recommended regardless of fertility fen therapy, and family history of uterine or
issue [13, 36]. colon cancer) and complete pelvic examina-
tion are the key aspects for evaluation of EH.
• The classifications commonly used for endo-
3.9.3 Special Issues metrial hyperplasia are WHO and EIN
classification.
Use of HRT in women with endometrial hyper- • In the woman with EH without atypia, sponta-
plasia: In women with EH on a sequential HRT, neous regression usually occurs during fol-
change to continuous combined HRT or LNG-­ low-­up and the risk of progression to EC is
IUD is recommended. In women with EH on less than 5% over 20 years.
continuous combined HRT, review of need of • The presence of cellular atypia carries highest
HRT or replacement with LNG-IUD is recom- chance of persistence, recurrence, and pro-
mended [13]. gression to EC or coexistent EC.
42 B. M. Patel

• Routine screening for women at high risk of 10. Townsend DE, Marrow CP. Tumors of the endome-
trium. In: Paul Morrow C, Curtin JP, editors. Synopsis
EH has not proven to be successful or cost-­ of gynecologic oncology. New York: Churchill
effective except for Lynch syndrome mutation Livingstone; 1975. p. 151–5.
carriers. 11. Trimble CL, Method M, Leitao M, Lu K, Ioffe O,
• Histological examination by an experienced Hampton M, et al. Management of endometrial pre-
cancers. Obstet Gynecol. 2012;120(5):1160–75.
gynecological pathologist is essential to dis- 12. Naeem S, Duncan T. Management of endome-
tinguish between different types of EH that trial hyperplasia. Norfolk and Norwich University
are managed differently. Hospitals 2016. 1–7. http://www.nnuh.nhs.uk/publi-
• In younger women with a desire to retain fer- cation/download/endometrial-hyperplasia-manage-
ment_g40_v2/&ved=0ahUKEwijtcjpzLHYAhWMQ
tility, medical treatment with progestins is o8KHQwuDbMQFgg2MAA&usg=AOvVaw2hjRYN
appropriate, while in older women especially HoS-4cGpZG1Xajmv
with atypical hyperplasia, hysterectomy is the 13. Royal College of Obstetricians and Gynaecologists
treatment of choice. (RCOG) with the British Society for Gynaecological
Endoscopy (BSGE). Management of Endometrial
Hyperplasia. Green-top Guideline No. 67. RCOG/BSGE
joint guideline. London; 2016. Cited 29 Mar 2016.
14. Ricci E, Moroni S, Parazzini F, Surace M, Benzi G,
References Salerio B, et al. Risk factors for endometrial hyper-
plasia: results from a case-control study. Int J Gynecol
1. Chandra V, Kim JJ, Benbrook DM, Dwivedi A, Rai Cancer. 2002;12:257–60.
R. Therapeutic options for management of endome- 15. Wells M. Hyperplasias of the endometrium. In:
trial hyperplasia. J Gynecol Oncol. 2016;27(1):1–25. Gershenson DM, McGuire WP, Gore M, Quinn MA,
2. Palmer JE, Perunovic B, Tidy JA. Review endometrial Gillian T, editors. Gynecologic cancer: controver-
hyperplasia. Obstet Gynecol. 2008;10:211–6. sies in management. Missouri: Elsevier Churchill
3. Landrum LM, Zuna RE, Walker JL. Endometrial Livingstone; 2004. p. 249–57.
hyperplasia, estrogen therapy, and the prevention of 16. Wise MR, Jordan V, Lagas A, Showell M, Wong N,
endometrial cancer. In: DiSaia PJ, Creasman WT, Lensen S, et al. Obesity and endometrial hyperplasia and
editors. Clinical gynecologic oncology. Philadelphia: cancer in premenopausal women: a systematic review.
Mosby Elsevier; 2007. p. 121–8. Am J Obstet Gynecol. 2016;214:689.e1–689.e17.
4. Classification and diagnosis of endometrial hyper- 17. van der Meer ACL, Hanna LS. Development of endo-
plasia [Internet]. Uptodate.com. 2017. https://www. metrioid adenocarcinoma despite Levonorgestrel-­
uptodate.com/contents/classification-and-diagno- releasing intrauterine system: a case report with
sis-of-endometrial-hyperplasia?search=reed%20 discussion and review of the RCOG/BSGE guideline
s d , u r b a n % 2 0 r. % 2 0 r. % 2 0 c l a s s i fi c a t i o n % 2 0 on the management of endometrial hyperplasia. Clin
and%20diagnosis%20of%20endometrial%20- Obes. 2017;7:54–7.
hyperplasia&source=search_result&selectedTitle=1~ 18. Lacey JV Jr, Ioffe OB, Ronnett BM, Rush BB,
150&usage_type=default&display_rank=1. Cited19 Richesson DA, Chatterjee N, et al. Endometrial carci-
Dec 2017 noma risk among women diagnosed with endometrial
5. Trimble CL, Kauderer J, Zaino R, Silverberg S, Lim hyperplasia: the 34-year experience in a large health
PC, Burke JJ II, et al. Concurrent endometrial carci- plan. Br J Cancer. 2008;98:45–53.
noma in women with a biopsy diagnosis of atypical 19. Horn LC, Schnurrbusch U, Bilek K, Hentschel B,
endometrial hyperplasia. Cancer. 2006;106(4):812–9. Einenkel J. Risk of progression in complex and atypi-
6. Zaino RJ, Kauderer J, Trimble CL, Silverberg SG, cal endometrial hyperplasia: clinicopathologic analy-
Curtin JP, Lim PC, Gallup DG. Reproducibility of sis in cases with and without progestogen treatment.
the diagnosis of atypical endometrial hyperplasia. Int J Gynecol Cancer. 2004;14:348–53.
Cancer. 2006;106(4):804–11. 20. Hacker NF, Friedlander ML. Uterine cancer. In: Berek
7. Sanderson PA, Critchley HOD, Williams ARW, JS, Hacker NF, editors. Berek & Hacker’s gyneco-
Arends MJ, Saunders PTK. New concepts for an old logic oncology. Philadelphia: Wolters Kluwer; 2015.
problem: the diagnosis of endometrial hyperplasia. p. 390–434.
Hum Reprod Update. 2017;23(2):1–23. 21. Lacey JV Jr, Sherman ME, Rush BB, Ronnett BM,
8. Reed SD, Newton KM, Clinton WL, Epplein M, Ioffe OB, Duggan MA, Glass AG, Richesson DA,
Garcia R, Allison K, et al. Incidence of endometrial Chatterjee N, Langholz B. Absolute risk of endo-
hyperplasia. Am J Obstet Gynecol. 2009;200:678. metrial carcinoma during 20-year follow-up among
e1–6. women with endometrial hyperplasia. J Clin Oncol.
9. Marsden DE, Hacker NF. Optimal management of 2010;28(5):788–92.
endometrial hyperplasia. Best Pract Res Clin Obstet 22. Lacey JV Jr, Mutter GL, Nucci MR, Ronnett BM,
Gynaecol. 2001;15(3):393–405. Ioffe OB, Rush BB, et al. Risk of subsequent endo-
3 Endometrial Hyperplasia: Diagnosis and Management 43

metrial carcinoma associated with endometrial the endometrium in women with postmenopausal
intraepithelial neoplasia classification of endometrial bleeding-A Nordic multicenter study. Am J Obset
biopsies. Cancer. 2008;113(8):2073–81. Gynecol. 1995;172(5):1488–94.
23. Kurman RJ, Kaminski PF, Norris HJ. The behav- 35. Christopherson WM, Gray LA. Premalignant lesions
ior of endometrial hyperplasia: a long-term study of the endometrium: endometrial hyperplasia and ade-
of “untreated” hyperplasia in 170 patients. Cancer. nocarcinoma in situ. In: Coppelson M, Monaghan JM,
1985;56:403–12. Morrow CP, Tattersall MHN, editors. Gynecologic
24. Kurman RJ, Norris HJ. Evaluation of criteria for distin- oncology: fundamental principles and clinical prac-
guishing atypical endometrial hyperplasia from well-­ tice (Vol 1 & 2). New York: Churchill Livingstone;
differentiated carcinoma. Cancer. 1982;49:2547–59. 1992. p. 731–45.
25. Emons G, Beckmann M, Schmidt D, Mallmann P. New 36. Trope C, Lindahl B. Premalignant lesions of the
WHO classification of endometrial hyperplasias. endometrium: clinical features and management. In:
Geburtshilfe Frauenheilkunde. 2015;75(2):135–6. Coppelson M, Monaghan JM, Morrow CP, Tattersall
26. Rao S, Sundaram S, Narasimhan R. Biological behav- MHN, editors. Gynecologic oncology: fundamental
iour of preneoplastic conditions of the endometrium: principles and clinical practice (Vol 1 & 2). New York:
a retrospective 16-year study in South India. Indian J Churchill Livingstone; 1992. p. 747–51.
Med Paediatr Oncol. 2009;30(4):131–5. 37. Management of endometrial hyperplasia [Internet].
27. Trope C, Alektiar K, Sabbatini P, Zaino R. Corpus: epi- Uptodate.com. 2017. https://www.uptodate.
thelial tumor. In: William JH, Carlos AP, Robert CY, com/contents/management-of-endometrial-
Richard B, Maurie M, Marcus R, editors. Principles hyperplasia?search=reed%20sd,urban%20r.%20
and practice of gynecologic oncology. Philadelphia: r.%20classification%20and%20diagnosis%20of%20
Lippincott Williams & Wilkins; 2005. p. 823–72. endometrial%20hyperplasia&source=search_result&
28. McMeekin DS, Yashar C, Campos S, Zaino selectedTitle=2~150&usage_type=default&display_
RJ. Corpus: epithelial tumors. In: Barakat R, Berchuck rank=2. Cited 19 Dec 2017.
A, Markman M, Randal ME, editors. Principles and 38. Gunderson CC, Fader AN, Carson KA, Bristow
practice of gynecologic oncology. Philadelphia: RE. Oncologic and reproductive outcomes with pro-
Wolters Kluwer; 2013. p. 661–714. gestin therapy in women with endometrial hyperpla-
29. Mutter GL, The Endometrial Collaborative Group. sia and grade 1 adenocarcinoma: a systematic review.
Endometrial Intraepithelial Neoplasia (EIN): will it Gynecol Oncol. 2012;125:477–82.
bring order to chaos? Gynecol Oncol. 2000;76:287–90. 39. Nwanodi O. Progestin intrauterine devices and met-
30. Raychaudhuri G, Bandyopadhyay A, Sarkar D, formin: endometrial hyperplasia and early stage
Mandal S, Mondal S, Mitra PK. Endometrial hyper- endometrial cancer medical management. Healthcare.
plasia: a clinicopathological study in a Tertiary Care 2017;5(30):1–10.
Hospital. J Obstet Gynecol India. 2013;63(6):394–8. 40. Wildemeersch D, Dhont M. Treatment of nonatypi-
31. Memon MJ, Gupta Y. Endometrial Hyperplasia: a 3— cal and atypical endometrial hyperplasia with a
years study at a rural based hospital. Schol J App Med levonorgestrel-­
releasing intrauterine system. Am J
Sci. 2016;4(10B):3707–10. Obstet Gynecol. 2003;188:1297–8.
32. Dijkhuizen FPHLJ, Mol BWJ, Brolmann HAM, 41. Kim MK, Seong SJ. Conservative treatment for
Heintz APM. The accuracy of endometrial sampling atypical endometrial hyperplasia: what is the most
in the diagnosis of patients with endometrial carci- effective therapeutic method? J Gynecol Oncol.
noma and hyperplasia. Cancer. 2000;89(8):1765–72. 2014;25(3):164–5.
33. Iglesias DA, Huang M, Soliman PT, Djordjevic B, Lu 42. Orbo A, Arnes M, Lysa LM, Borgfelt C, Straume
KH. Endometrial hyperplasia and cancer. In: Karlan B. HE4 is a novel tissue marker for therapy
BY, Bristow RE, Andrew LJ, editors. Gynecologic response and progestin resistance in medium- and
oncology clinical practice & surgical atlas. New York: low-risk endometrial hyperplasia. Br J Cancer.
McGraw Hill; 2012. p. 250–326. 2016;115:725–30.
34. Karlsson B, Gransberg S, Wiklan M, Pekka Y, Torvid
K, Marsal K, et al. Transvaginal ultrasonography of

You might also like