ArticlePDF AvailableLiterature Review

Abstract and Figures

Endometriosis is the leading cause of morbidity among premenopausal women affecting about 1 in 10 females. The features shared by endometriosis and cancer include the ability to evade apoptosis, the stem cell-like ability and angiogenic potential. As such characteristics are encoded by the cell's genetic constitution, acquired mutations are responsible for the malignant transformation of endometriosis. Indeed, a number of tumour-suppressor genes and proto-oncogenes, such as protein 53 (P53) and B-cell lymphoma 2 (BCL-2) respectively, are mutated and as a result differentially expressed between endometriotic and malignant tissue associated with endometriosis. Moreover, cytokines and macrophages, both of which are inflammatory mediators have been implicated in the transformation process. The angiogenic properties possessed by cancer arising from endometriosis signifies a bad prognosis, while the stem cell-like activity possessed by both endometriosis and cancer has been attributed to the effect of oestrogen. A number of differences between endometriosis and cancer are found at the molecular level. Considering the link between these two pathologies, the three components which fuel the malignant transformation of endometriosis can be embodied in the endometriosis-induced carcinoma (EIC) triangle which shows the intricate relationship between endocrinologic, immunologic and genetic components.
Content may be subject to copyright.

Gynecological Endocrinology
2012
28
8
577
581
© 2012 Informa UK, Ltd.
10.3109/09513590.2011.650761
0951-3590
1473-0766
Gynecological Endocrinology, 2012; 28(8): 577–581
© 2012 Informa UK, Ltd.
ISSN 0951-3590 print/ISSN 1473-0766 online
DOI: 10.3109/09513590.2011.650761
Endometriosis is the leading cause of morbidity among premeno-
pausal women affecting about 1 in 10 females. The features
shared by endometriosis and cancer include the ability to evade
apoptosis, the stem cell-like ability and angiogenic potential.
As such characteristics are encoded by the cell’s genetic consti-
tution, acquired mutations are responsible for the malignant
transformation of endometriosis. Indeed, a number of tumour-
suppressor genes and proto-oncogenes, such as protein 53
(P53) and B-cell lymphoma 2 (BCL-2) respectively, are mutated
and as a result differentially expressed between endometriotic
and malignant tissue associated with endometriosis. Moreover,
cytokines and macrophages, both of which are inflammatory
mediators have been implicated in the transformation process.
The angiogenic properties possessed by cancer arising from
endometriosis signifies a bad prognosis, while the stem cell-like
activity possessed by both endometriosis and cancer has been
attributed to the effect of oestrogen. A number of differences
between endometriosis and cancer are found at the molecular
level. Considering the link between these two pathologies, the
three components which fuel the malignant transformation of
endometriosis can be embodied in the endometriosis-induced
carcinoma (EIC) triangle which shows the intricate relationship
between endocrinologic, immunologic and genetic components.
Keywords: Apoptosis, cancer, cytokines, endometriosis
Abbreviations: AE, atypical endometriosis; ARID-1A, AT-rich
interactive domain 1A; BCL-2, B-cell lymphoma 2; bFGF, basic
fibroblast growth factor; EAOC, endometriosis-associated
ovarian carcinoma; GALT, galactose-1-phosphate uridyl
transferase; hMLH1, human MutL homolog 1; HNF-1β,
Hepatocyte nuclear factor-1β; HUMARA, human androgen
receptor gene; IL-1, interleukin-1; IL-1β, interleukin-1β; JNK,
c-Jun N-terminal kinase; LOH, loss of heterozygosity; MMR,
mismatch repair; mRNA, messenger RNA; MSI, microsatellite
instability; NF-κB, nuclear factor-κb; OC, ovarian cancer; P53,
protein 53; PGE-2, prostaglandin E2; PGK-1, phosphoglycerate
kinase 1; PTEN, phosphatase and tensin homolog; RANK,
receptor activator of nuclear factor ĸB; ROS, reactive oxygen
species; TAM, tumour-associated macrophage; TGF-β,
transforming growth factor-β; VEGF, vascular endothelial
growth factor
Introduction
Endometriosis is a benign gynaecological pathology aecting about
1 in 10 females during their reproductive years [1]. is condition
shares a number of features with cancer [2] such as its stem cell-like
activity [3], the ability to evade apoptosis [4] and to induce angiogen-
esis [5,6], as well as the intrinsic self-regulation of its rate of prolifera-
tion [7,8]. Such properties can be explained through the molecular
signals between the cells which regulate the intracellular signalling
cascades. is allows the population of ectopic cells to survive
within its new tissue environment [9]. Is it possible that a change
in the genetic code of the endometriotic cells during the course of
the benign disease can modify the gene expression pattern of such
cells rendering them capable of metastatic behaviour. Indeed, the
malignant transformation of endometriosis has been the subject of
a lot of studies whereby the risk of developing certain cancers, most
notably ovarian cancer (OC), was found to be signicantly increased
in women having a history of endometriosis [7,10–18]. is review
seeks to outline the link between the molecular properties of endo-
metriosis and cancer and identify possible candidate signals respon-
sible for the malignant transformation of endometriosis. Moreover,
according to a study by Banz et al. [2], two distinct sets of genes
were found, one of which includes genes which show dierential
expression in endometriosis and endometriosis-associated ovarian
cancer (EAOC), but not in OC and benign ovaries whilst the other
consists of genes which are dierently expressed in endometriosis-
associated ovarian cancer (EAOC) and OC but not in endometriosis
and benign ovaries. e genes which are dierentially expressed in
endometriosis and EAOC as opposed to OC and benign ovaries,
may be possible candidates for predisposing to the development of
EAOCs.
Apoptosis in endometriosis and cancer
In apoptosis, there are two major apoptotic regulatory proteins
which dier in their level of expression between benign and
malignant tissue: B-cell lymphoma 2 protein (BCL-2) and protein
53 (P53) [4]. BCL-2 is one of a diverse class of regulatory proteins,
the majority of which are antiapoptotic [19]. P53 is a regulatory
protein which allows the repair of minor insults which occur in
the process of DNA replication and which signals apoptosis in
case of major DNA damage [20]. BCL-2 levels are upregulated in
endometriotic tissues with about one in four endometriotic cysts
expressing BCL-2 [4]. e expression of BCL-2 in ovarian carci-
noma is higher than that for the ovarian endometriotic cysts [4].
Moreover, according to the same study, areas of the carcinoma
which showed up as being malignant had a higher expression than
those areas which showed up as being benign [4]. is suggests
that upregulation of BCL-2 in endometriotic tissue may be one of
the factors that promotes malignant transformation. e level of
BCL-2 expression in the serous papillary ovarian carcinoma is also
highly upregulated in comparison to the ovarian endometriotic
GYNECOLOGICAL CANCER
Molecular links between endometriosis and cancer
Joel Pollacco1, Keith Sacco1, Mark Portelli1, Pierre Schembri-Wismayer1 & Jean Calleja-Agius1,2
1 Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Msida, MSD 2080, Malta and 2Department
of Obstetrics and Gynaecology, Mater Dei Hospital, Msida, MSD 2090, Malta
Correspondence: Dr Jean Calleja-Agius, Department of Anatomy, Faculty of Medicine and Surgery, University of Malta, Msida, MSD 2080, Malta.
Tel: +356 21693041. Fax: +356 21319527. E-mail: jean.calleja-agius@um.edu.mt
578 J. Pollacco et al.
Gynecological Endocrinology
cyst [4]. Benign endometriotic cysts do not overexpress P53,
while the expression of P53 is upregulated in the carcinomatous
tissues [4]. Ovarian carcinoma was shown to exhibit a signicantly
higher level of protein expression than the benign counterpart of
the same histologic sub-type [4]. P53 has been documented in the
case of an ovarian yolk-sac tumour associated with endometrioid
adenocarcinoma and endometriosis [21]. Another study has
shown that expression of BCL-2 was downregulated while that
of P53 was upregulated in the case of ovarian epithelial tumours
[22]. e same pattern of results was obtained in another study
[18], whereby all the tumours examined also tested positive for
KI-67, which was used as a marker for proliferation [18]. It is not
only the level of expression of BCL-2 and P53 which is altered
in the malignant transformation of endometriosis, but certain
studies suggest that the proteins themselves are structurally modi-
ed as a result of mutations sustained within the specic gene loci
coding for such proteins [22,23]. is may in turn account for the
upregulation of P53 in ovarian endometrioid carcinomas arising
in endometriosis [24].
The role of macrophages
Macrophages play a key role in inammatory processes.
Previous research has shown that macrophages contribute to
tumorigenesis through the chronic inammatory processes
which they mediate [25]. Endometriosis induces a localized
inammatory reaction in the immediate vicinity of the area
of the peritoneum or viscera, where the endometriotic cells
inltrate [26]. e macrophages produce a wide range of cytok-
ines such as interleukin-1β (IL-1β) which in turn encourage
the progression of tumorigenesis [9]. IL-1β is one of the most
potent mediators of inammation, and it promotes tumorige-
nicity by stimulating the production of a diverse array of cytok-
ines, chemokines and intercellular communication mediators
which also lead to angiogenesis [9,27]. IL-1β is one of the two
members of the interleukin-1(IL-1) class family, which act as
agonists of the IL-1 receptor [27].
Macrophages also produce an array of cytotoxic reactive
oxygen species (ROS) in their function to destroy pathogenic
material [28]. e production of cytotoxic ROS is also contrib-
uted through the Fenton reaction [29] by the high levels of free
iron ions found in the endometriotic cysts due to the repeated
degradation of ectopic menstrual blood [30]. ese free radicals
scavenge the cells’ macromolecules including DNA and proteins
[31], causing structural DNA damage by oxidation of the DNA
bases [32]. ey also act indirectly through specic molecular
pathways. Although this has not been substantiated to the endo-
metrial tissue, in breast cancer it has been found that nuclear
factor-κb (NF-ĸB) and AP molecular pathway activation by ROS
are actually mediated by convergence of the molecular signals
received from activation of receptor activator of nuclear factor
ĸB (RANK) and transforming growth factor-β (TGF-β) recep-
tors [33]. ese transcription factors promote the expression of
proteins which increase the cells’ angiogenic and proliferative
potential [34]. Indirect free-radical DNA damage can also be due
to adduct formation between the nucleic acid bases and the lipid
peroxides [32].
In normal physiologic responses to tissue injury, macrophages
function in extensive remodelling of the injured tissue area
leading to complete resolution [28]. In order to carry out these
functions, macrophages harbour a set of catabolic enzymes. In
particular, the matrix metalloproteinases are presumably able
to induce the secretion of a large amount of growth factors such
as vascular endothelial growth factor (VEGF) and basic bro-
blast growth factor (bFGF) from the underlying extracellular
matrix [35] so as to activate the trophic eect of the local tissues
in an attempt to replace the damaged tissue. It is hypothesized
that there is continuous cell to cell communication between the
macrophages and the premalignant cells in the chronic inam-
ma to ry st ate [36]. e same genes which impart the macrophage
with the ability to degrade extracellular matrix material are
induced in the stromal cells [28]. is increased invasiveness
occurs via activation of two molecular pathways, the NF-κB
and c-Jun N-terminal kinase (JNK) pathways respectively,
through the action of tumour necrosis factor α (TNF-α) [36].
Having lost their intrinsic ability to regulate the rate of cell divi-
sion through repeated insults to their genome, these stromal
cells can progress to malignancy [28]. Such macrophages are
known as tissue-activated macrophages (TAMs). Indeed, this
not only applies to the endometriosis-carcinoma sequence but
also applies to the malignant transformation of other benign
counterparts of endometriosis [28]. e TAMs are known to
be involved in antagonizing T-cell mediated immunity [25]
either by producing immunosuppressive substances such as
indoleamine dioxygenase [37] or by inhibiting dendritic cell
maturation and thus antigen presentation [25].
Angiogenesis in endometriosis and cancer
VEGF has been detected in almost all samples of EAOC. This is
in contrast to the detection of VEGF in only 12.5% of atypical
endometriosis (AE) specimens [38]. This indicates that VEGF
plays a major critical role in the survival of the tumour cells.
Hence, the increase in level of VEGF expression by the endo-
metriotic tissue may be an indication of transformation to
malignancy [38]. Ovarian cancers with a higher VEGF expres-
sion were shown to have a significantly worse prognosis [5].
Moreover, in one study, the presence of increasingly high levels
of VEGF in tissues corresponded to the absence of T cells in the
immediate vicinity of the tumour [39]. Subcutaneous tumours
implanted in immunodeficient mice stop growing when VEGF
function was blocked using a complementary antibody [6].
This suggests that VEGF provides adequate vascular perfu-
sion to new populations of cells which emerge in a body tissue
[6]. Besides the role of VEGF as an angiogenic factor, it has
been proposed that VEGF functions as a growth factor for the
tumour, independent of angiogenesis [5].
Stem cell-like activity in endometriosis and cancer
e limit to a cell’s replicative ability lies in the short repeti-
tive DNA sequences found at the ends of chromosomes termed
telomeres. e progressive shortening of the telomeres which
occurs as a cell passes from one cell cycle to the next leads
to its senescence and eventually to its death [32]. However,
endometrial cells are induced to express telomerase enzyme
by oestrogen [3]. is may be the factor which imparts the
endometrial cells with increased proliferative ability that allows
them to replace the functional layers of the endometrium
during the menstrual cycle [40]. In endometriosis, the ability of
self-renewal of the endometrial tissue is somewhat increased,
and this allows them to proliferate in such a way as to form
macroscopic lesions outside the uterus [41]. Since telomerase
reactivation and prolonged proliferation are features of cancer
cells, the upregulation of telomerase enzyme may be another
factor which contributes to the malignant transformation of
endometriosis.
Endometriosis and cancer 579
©  Informa UK, Ltd.
Genomic differences between endometrial cells, endometriosis
and cancer
A number of comparisons have been described between the
genetic constitution of endometriotic cells and clear cell or
endometrioid ovarian carcinomatous cells in terms of the gene
expression patterns [24,42,43]. e c-erbB-2 ge ne i s mo re h ighl y
expressed in endometriosis adjacent to ovarian carcinoma than
in endometriotic tissue without any adjacent malignant tissue
[24]. However, many dierences in terms of gene expression
patterns have been found. A number of dierent polymor-
phisms are found at the codon 72 of the p53 gene in women
with endometriosis [44]. is is one of the dierences in the
genetic constitution between wild-type endometrial cells and
endometriotic cells. Loss of heterozygosity (LOH) at the 10q23.3
locus of the genome in ovarian endometrioid carcinomas,
clear-cell ovarian carcinomas and endometriotic cysts has been
documented [42] with the LOH prevailing to the highest extent
in endometriotic cysts. A series of mutations at the same locus
have also been detected in a signicant amount of carcinomas
of the ovary. Moreover, some of the carcinomas associated with
endometriosis also displayed LOH at the 10q23.3 gene locus
[42]. LOH has also been found in a number of genetic loci such
as on the chromosome arms 9p, 11q and 22q [45]. In another
study [43], LOH was shown to occur at 17 dierent genetic loci
in 9 out of 17 cases of endometriosis coincident with carci-
noma. e LOH was present in both the malignant tissue and
the adjacent benign endometriotic tissue.
Mutations have been found in the AT-rich interactive domain
1A (ARID-1A) in less than half of samples of ovarian clear-cell
carcinomas and endometrioid carcinomas [46]. e ARID-1A
gene codes for BAF250, a tumour-suppressor protein. In the same
study, these mutations have been linked to the loss of expression of
BAF250. e fact that such a mutation is found in only the two types
of cancer, which can arise from endometriosis means that such a
change in endometriotic cysts may give rise to such cancers [46].
e human MutL homolog 1 (hMLH1) gene is known to
be hypermethylated in Stage IV endometriosis not associ-
ated with carcinoma and AE in association with clear-cell or
endometrioid ovarian carcinoma [47]. e hypermethylation
of the tumour-suppressor P16 gene together with an increase
in the level of tumour markers is also associated with stage IV
endometriosis [47]. hMLH1 itself also functions as a tumour-
suppressor gene since it encodes an enzyme involved in the
mismatch repair (MMR) system. e MMR system is one of
the intrinsic mechanisms a cell possesses which proofread the
DNA for any errors in base-pairing aer DNA replication [32].
Consequently it has been shown that the loss of expression of
the hMLH1 protein in endometrioid cancers with microsatellite
instability (MSI) is largely due to gene silencing of the hMLH1
gene through hypermethylation.
Alternatively, the presence of MSI in cells which do not have
hypermethylation or any abnormal methylation patterns at the
hMLH1 locus have been attributed to the occurrence of somatic
mutations at the respective microsatellite regions [48]. Since MSI
is so tightly associated with endometrioid carcinomas and is
largely caused by inactivation of the hMLH1 gene locus [48,49],
it follows that hMLH1 gene inactivation could be an early marker
of the malignant conversion of endometriotic lesions to endo-
metrioid carcinomas [47].
e expression of a particularly modied gene for the
progesterone receptor, the PROGINS gene is associated with
an increased risk of developing OC [50]. Hence, subsequent
somatic mutations at the gene locus which codes for proges-
terone receptor in the course of the rapid cell cycling which
occurs in endometriosis may predispose a woman to onco-
genesis. To assess the monoclonality of carcinomas arising
from endometriosis, the methylation of various gene loci on
the X-chromosome such as human androgen receptor gene
(HUMARA) [51] and phosphoglycerate kinase 1 (PGK-1) [52]
can be used as markers. Endometriotic lesions have been shown
to be polyclonal [52]. e monoclonality of cancers arising
from endometriosis means that the cancer has arisen from the
clonal expansion of a single cell and hence supports the notion
of EAOCs, as being cancers arising from the clonal expansion
of a single endometriotic cell.
Possible mechanisms responsible for the malignant
transformation of endometriotic lesions
Malignant cells exploit the cells’ intrinsic mechanisms to regulate
the length of its lifespan and its metabolism to their own advantage.
As oxygen, heat and fuel constitute the re triangle, we propose
the endometriosis-induced carcinoma (EIC) triangle (Figure 1).
e three factors which directly contribute to endometriosis-
induced oncogenesis are (1) alterations within the endometriotic
cells’ genome, (2) endocrinological factors and (3) immunological
factors. e relationship between inammation, genetic inuences
and cancer has been described by Balkwill et al. [25], as the genetic
inuences being “the fuel that feeds the ame” and inammation
being the “fuel that feeds the ame.However, in endometriosis,
there is also the endocrinological component which interacts with
the genetic inuences, in addition to the immunological factors. In
eect, the high local levels of oestrogen in the ovarian endometri-
otic foci, due to the upregulation of aromatase cause an increase
in the level of prostaglandin E2 (PGE-2) [53], which in turn
suppresses normal immunologic responses such as phagocytosis
[54] and leads to diminished apoptosis [55]. e increased levels
of local oestrogens increases the level of expression of angiogenic
Figure 1. e endometriosis-induced carcinoma (EIC) triangle. bcl-2, B-cell
lymphoma 2; hMLH1, human MutL homolog 1; PTEN, phosphatase and
tensin homolog; p16, protein 16; p53, protein 53; TNF-α, tumour necrosis
factor α; TSGs, tumour-suppressor genes.
580 J. Pollacco et al.
Gynecological Endocrinology
factors such as VEGF [56] and may contribute to the increased
expression of telomerase [32].
In our view, there is a continuum between the physiologically
normal endometrium, endometriosis and eventually carcinoma,
which explains the dierent characteristics exhibited by the
pathological tissue obtained from dierent women suering from
endometriosis. Endometriosis lies in the middle of the spectrum
ranging from the physiologically normal endometrium at one
end, to carcinoma at the other extreme. us, even endometriosis
itself is a consequence of the three components of the EIC triangle
to a certain extent. e EIC triangle being proposed is the gateway
for progression to malignancy from the benign endometriotic
state. e three components are closely interconnected to each
other at the molecular level such that the more they contribute to
the oncogenic process, the greater the risk for a woman to develop
neoplasia early on in her life. e genetic component of the EIC
triangle would be somatic loss of function mutations in tumour-
suppressor genes such as phosphatase and tensin homolog (PTEN),
P53, hMLH1 genes and BCL-2. e endocrinologic component
would be the role of oestrogen in determining the potential of
the endometriotic tissue to become cancerous by inducing the
expression of telomerase amongst others. e immunologic
component of the triad making up the EIC triangle would be the
consequences of endometriosis as an inammatory process, espe-
cially the role played by macrophages.
Considering this scenario, it should be possible to halt the
progression of endometriosis to carcinoma by breaking up the
EIC triangle. is could be done by antagonizing the endocrino-
logic or immunologic eects contributing to the triangle such as
by antagonizing the action of TAMs. Indeed, the use of methods
by which the action of inammatory mediators is inhibited to halt
tumorigenesis or cancer progression has already been described
in the literature [25]. Moreover, the components which are to
be therapeutically targeted in such a case must be specic to the
endometriosis-carcinoma sequence so as to avoid adverse eects.
Details as to how this can be actualized within the clinical setting
are not yet currently available, and further studies in this prom-
ising eld of research are required.
Conclusion
Despite the molecular links between the two, endometriosis and
malignancy, endometriosis remains a benign gynaecological condi-
tion having a major impact on women’s health [57], while cancer
claims the lives of many women annually [58], with OC being the
second most lethal gynaecological malignancy in 2008 in the UK
[59]. A knowledge of the divergences and convergencies of these
two diseases may shed light on innovative therapy in this respect.
Declaration of Interest: e authors declare no conict of interest.
References
1. Germeyer A, Giudice LC. How does endometriosis cause infertility?
In: Tulandi T, Redwine D, editors. Endometriosis Advances and
Controversies. New York: Marcel Dekker Inc; 2004. pp 151.
2. Banz C, Ungethuem U, Kuban RJ, Diedrich K, Lengyel E, Hornung D.
e molecular signature of endometriosis-associated endometrioid
ovarian cancer diers signicantly from endometriosis-independent
endometrioid ovarian cancer. Fertil Steril 2010;94:1212–1217.
3. Wang Z, Kyo S, Maida Y, Takakura M, Tanaka M, Yatabe N, Kanaya
T, et al. Tamoxifen regulates human telomerase reverse transcriptase
(hTERT) gene expression dierently in breast and endometrial cancer
cells. Oncogene 2002;21:3517–3524.
4. Nezhat F, Cohen C, Rahaman J, Gretz H, Cole P, Kalir T. Comparative
immunohistochemical studies of bcl-2 and p53 proteins in benign and
malignant ovarian endometriotic cysts. Cancer 2002;94:2935–2940.
5. Shen GH, Ghazizadeh M, Kawanami O, Shimizu H, Jin E, Araki
T, Sugisaki Y. Prognostic signicance of vascular endothelial
growth factor expression in human ovarian carcinoma. Br J Cancer
2000;83:196–203.
6. Mesiano S, Ferrara N, Jae RB. Role of vascular endothelial
growth factor in ovarian cancer: inhibition of ascites formation by
immunoneutralization. Am J Pathol 1998;153:1249–1256.
7. Varma R, Rollason T, Gupta JK, Maher ER. Endometriosis and the
neoplastic process. Reproduction 2004;127:293–304.
8. Prowse AH, Manek S, Varma R, Liu J, Godwin AK, Maher ER, Tomlinson
IP, Kennedy SH. Molecular genetic evidence that endometriosis is a
precursor of ovarian cancer. Int J Cancer 2006;119:556–562.
9. Gazvani R, Templeton A. Peritoneal environment, cytokines and
angiogenesis in the pathophysiology of endometriosis. Reproduction
2002;123:217–226.
10. Melin A, Sparén P, Persson I, Bergqvist A. Endometriosis and the
risk of cancer with special emphasis on ovarian cancer. Hum Reprod
2006;21:1237–1242.
11. Aris A. Endometriosis-associated ovarian cancer: A ten-year cohort
study of women living in the Estrie Region of Quebec, Canada. J
Ovarian Res 2010; 3:2.
12. Kusaka M, Mikuni M, Nishiya M. A case of high-grade endometrial
stromal sarcoma arising from endometriosis in the cul-de-sac. Int J
Gynecol Cancer 2006;16:895–899.
13. Leng J, Lang J, Guo L, Li H, Liu Z. Carcinosarcoma arising from
atypical endometriosis in a cesarean section scar. Int J Gynecol Cancer
2006;16:432–435.
14. Garavan F, Grainger R, Jeers M. Endometrioid carcinoma of the
urinary bladder complicating vesical Mullerianosis: a case report and
review of the literature. Virchows Arch 2004;444:587–589.
15. Jimenez RE, Tiguert R, Hurley P, An T, Grignon DJ, Lawrence D, Triest
J. Unilateral hydronephrosis resulting from intraluminal obstruction
of the ureter by adenosquamous endometrioid carcinoma arising from
disseminated endometriosis. Urology 2000;56:331.
16. Salerno MG, Masciullo V, Naldini A, Zannoni GF, Vellone V, Scambia
G. Endometrioid adenocarcinoma with squamous dierentiation
arising from ureteral endometriosis in a patient with no history of
gonadal endometriosis. Gynecol Oncol 2005;99:749–752.
17. Park HM, Lee SS, Eom DW, Kang GH, Yi SW, Sohn WS. Endometrioid
adenocarcinoma arising from endometriosis of the uterine cervix: a
case report. J Korean Med Sci 2009;24:767–771.
18. Han AC, Hovenden S, Rosenblum NG, Salazar H. Adenocarcinoma
arising in extragonadal endometriosis: an immunohistochemical study.
Cancer 1998;83:1163–1169.
19. Paglieri LJ, Pinkoski MJ, Green DR., Apoptosis signalling: A means to
an end: Chapter 331. In: Bradshaw RA, Dennis EA, editors-in-chief.
Handbook of Cell Signalling, Vol. 3. United States of America: Elsevier
Science; 2003. pp 431–439.
20. Underwood JCE, Cross SS. General and systematic pathology 5th ed.
China: Elsevier Limited; 2009. pp 248.
21. Horiuchi A, Osada R, Nakayama K, Toki T, Nikaido T, Fujii S.
Ovarian yolk sac tumor with endometrioid carcinoma arising from
endometriosis in a postmenopausal woman, with special reference to
expression of alpha-fetoprotein, sex steroid receptors, and p53. Gynecol
Oncol 1998;70:295–299.
22. Chan WY, Cheung KK, Schorge JO, Huang LW, Welch WR, Bell DA,
Berkowitz RS, Mok SC. Bcl-2 and p53 protein expression, apoptosis,
and p53 mutation in human epithelial ovarian cancers. Am J Pathol
2000;156:409–417.
23. Akahane T, Sekizawa A, Purwosunu Y, Nagatsuka M, Okai T. e role
of p53 mutation in the carcinomas arising from endometriosis. Int J
Gynecol Pathol 2007;26:345–351.
24. Prefumo F, Venturini PL, Fulcheri E. Analysis of p53 and c-erbB-2
expression in ovarian endometrioid carcinomas arising in
endometriosis. Int J Gynecol Pathol 2003;22:83–88.
25. Balkwill, F., Charles, K. A. and Mantovani, A. 2007. e Links between
Inammation and Cancer. e Cancer Handbook.
26. Ness RB, Cottreau C. Possible role of ovarian epithelial inammation in
ovarian cancer. J Natl Cancer Inst 1999;91:1459–1467.
27. Song X, Voronov E, Dvorkin T, Fima E, Cagnano E, Benharroch
D, Shendler Y, et al. Dierential eects of IL-1 alpha and IL-1
beta on tumorigenicity patterns and invasiveness. J Immunol
2003;171:6448–6456.
Endometriosis and cancer 581
©  Informa UK, Ltd.
28. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell
migration, invasion, and metastasis. Cell 2006;124:263–266.
29. Fenton HJH. Oxidation of tartaric acid in presence of iron. J Chem Soc
1894;65:899–810.
30. Yamaguchi K, Mandai M, Toyokuni S, Hamanishi J, Higuchi T,
Takakura K, Fujii S. Contents of endometriotic cysts, especially the
high concentration of free iron, are a possible cause of carcinogenesis
in the cysts through the iron-induced persistent oxidative stress. Clin
Cancer Res 2008;14:32–40.
31. Stricker TP, Kumar V. Neoplasia: Chapter 7. In: Kumar V, Abbas AK,
Fausto N, Aster JC, editors. Robbins and Cotran Pathologic Basis of
Disease. 8th ed. China: Elsevier Saunders; 2010. pp 310.
32. Edenburg HJ. DNA replication, recombination and repair. Chapter
4. In: Devlin TM, editor. Textbook of Biochemistry With Clinical
Correlations. United States of America: Wiley Liss; 2006. pp 152–153,
159, 159–160 and 164–165.
33. Benz CC, Yau C. Ageing, oxidative stress and cancer: paradigms in
parallax. Nat Rev Cancer 2008;8:875–879.
34. Zhou Y, Yau C, Gray JW, Chew K, Dairkee SH, Moore DH, Eppenberger
U, et al. Enhanced NF kappa B and AP-1 transcriptional activity
associated with antiestrogen resistant breast cancer. BMC Cancer
2007;7:59.
35. Coussens LM, Werb Z. Matrix metalloproteinases and the development
of cancer. Chem Biol 1996;3:895–904.
36. Hagemann T, Wilson J, Kulbe H, Li NF, Leinster DA, Charles K, Klemm
F, et al. Macrophages induce invasiveness of epithelial cancer cells via
NF-kappa B and JNK. J Immunol 2005;175:1197–1205.
37. Bronte V, Serani P, Mazzoni A, Segal DM, Zanovello P. L-arginine
metabolism in myeloid cells controls T-lymphocyte functions. Trends
Immunol 2003;24:302–306.
38. Del Carmen MG, Smith Sehdev AE, Fader AN, Zahurak ML,
Richardson M, Fruehauf JP, Montz FJ, Bristow RE. Endometriosis-
associated ovarian carcinoma: dierential expression of vascular
endothelial growth factor and estrogen/progesterone receptors. Cancer
2003;98:1658–1663.
39. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M,
Regnani G, Makrigiannakis A, et al. Intratumoral T cells, recurrence, and
survival in epithelial ovarian cancer. N Engl J Med 2003;348:203–213.
40. Yokoyama Y, Takahashi Y, Morishita S, Hashimoto M, Niwa K, Tamaya
T. Telomerase activity in the human endometrium throughout the
menstrual cycle. Mol Hum Reprod 1998;4:173–177.
41. Kim CM, Oh YJ, Cho SH, Chung DJ, Hwang JY, Park KH, Cho DJ,
et al. Increased telomerase activity and human telomerase reverse
transcriptase mRNA expression in the endometrium of patients with
endometriosis. Hum Reprod 2007;22:843–849.
42. Sato N, Tsunoda H, Nishida M, Morishita Y, Takimoto Y, Kubo T,
Noguchi M. Loss of heterozygosity on 10q23.3 and mutation of
the tumor suppressor gene PTEN in benign endometrial cyst of the
ovary: possible sequence progression from benign endometrial cyst to
endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer
Res 2000;60:7052–7056.
43. Jiang X, Morland SJ, Hitchcock A, omas EJ, Campbell IG. Allelotyping
of endometriosis with adjacent ovarian carcinoma reveals evidence of a
common lineage. Cancer Res 1998;58:1707–1712.
44. Hsieh YY, Lin CS. P53 codon 11, 72, and 248 gene polymorphisms in
endometriosis. Int J Biol Sci 2006;2:188–193.
45. Jiang X, Hitchcock A, Bryan EJ, Watson RH, Engleeld P, omas EJ,
Campbell IG. Microsatellite analysis of endometriosis reveals loss of
heterozygosity at candidate ovarian tumor suppressor gene loci. Cancer
Res 1996;56:3534–3539.
46. Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, et al.
ARID1A mutations in endometriosis-associated ovarian carcinomas.
N Engl J Med 2010;363:1532–1543.
47. Martini M, Ciccarone M, Garganese G, Maggiore C, Evangelista A,
Rahimi S, Zannoni G, et al. Possible involvement of hMLH1, p16(INK4a)
and PTEN in the malignant transformation of endometriosis. Int J
Cancer 2002;102:398–406.
48. Esteller M, Catasus L, Matias-Guiu X, Mutter GL, Prat J, Baylin SB,
Herman JG. hMLH1 promoter hypermethylation is an early event in
human endometrial tumorigenesis. Am J Pathol 1999;155:1767–1772.
49. Esteller M, Levine R, Baylin SB, Ellenson LH, Herman JG. MLH1
promoter hypermethylation is associated with the microsatellite
instability phenotype in sporadic endometrial carcinomas. Oncogene
1998;17:2413–2417.
50. Agoulnik IU, Tong XW, Fischer DC, Körner K, Atkinson NE, Edwards
DP, Headon DR, et al. A germline variation in the progesterone receptor
gene increases transcriptional activity and may modify ovarian cancer
risk. J Clin Endocrinol Metab 2004;89:6340–6347.
51. Jang SJ, Mao L. Methylation patterns in human androgen receptor gene
and clonality analysis. Cancer Res 2000;60:864–866.
52. Mayr D, Amann G, Siefert C, Diebold J, Anderegg B. Does endometriosis
really have premalignant potential? A clonal analysis of laser-microdissected
tissue. FASEB J 2003;17:693–695. DOI: 10.1096/.02-0562e.
53. Attar E, Bulun SE. Aromatase and other steroidogenic genes in
endometriosis: translational aspects. Hum Reprod Update 2006;12:49–56.
54. Dmowski WP, Gebel HM, Braun DP. e role of cell-mediated immunity
in pathogenesis of endometriosis. Acta Obstet Gynecol Scand Suppl
1994;159:7–14.
55. Sacco K, Portelli M, Pollacco J, Schembri-Wismayer P, Calleja-Agius J.
e role of prostaglandin E(2) in endometriosis. Gynecol Endocrinol
2011. [Epub Ahead of Print].
56. Lin YJ, Lai MD, Lei HY, Wing LY. Neutrophils and macrophages
promote angiogenesis in the early stage of endometriosis in a mouse
model. Endocrinology 2006;147:1278–1286.
57. Koninckx RF. Epidemiology of endometriosis. In: Tulandi T, Redwine
D, editors. Endometriosis Advances and Controversies. New York:
Marcel Dekker Inc; 2004. pp 16.
58. Ricciardelli C, Oehler MK. Diverse molecular pathways in ovarian
cancer and their clinical signicance. Maturitas 2009;62:270–275.
59. Cancer Research UK [Internet]. Ovarian Cancer UK Incidence
Statistics, Cancer Research UK. Cited Jul 8, 2011. Available from:
http://info.cancerresearchuk.org/cancerstats/types/ovary/incidence/
Copyright of Gynecological Endocrinology is the property of Taylor & Francis Ltd and its content may not be
copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written
permission. However, users may print, download, or email articles for individual use.
... Endometriosis and endometrial cancer share common features in terms of molecular pathogenesis, overgrowth, and resistance to apoptosis (35)(36)(37). Surprisingly however, the effects of Fuling on endometrial cancer have not previously been assessed. Work here evaluated treatments with a broad range of Fuling doses in two grades of EC cell lines. ...
... Ongoing research is defining possible mechanisms of action. Common molecular and genetic features between endometriosis and cancer (35,37) prompted our hypothesis that Fuling might have beneficial effects in endometrial cancer, an idea that has not to our knowledge been tested previously. The major outcome of this study was the demonstration that Fuling inhibits invasiveness in both high and low grades of EC cells, and that this effect was achieved at doses estimated to be comparable to those being used in clinical testing and traditional practice. ...
Article
Full-text available
The Traditional Chinese medicine, Guizhi Fuling (here called Fuling), has been confirmed in meta-analysis studies to reduce recurrence of endometriosis and improve pregnancy outcomes; however, the possible use of Fuling as a fertility-preserving treatment in endometrial cancer has not previously been tested. Results here are the first to demonstrate dose-dependent inhibition of cell motility by Fuling in two endometrial cancer cell lines, classified as Grade I which is responsive to progesterone treatment, and Grade III (MFE-280) which is resistant. The major outcome of this study was the novel demonstration that Fuling (30-80 µg/ml) significantly inhibits invasiveness in both high and low grades of EC cells, achieving 70-80% block of trans-barrier migration without cytotoxicity. This effective dose range is estimated to be comparable to that used in human clinical trials and traditional practice. Results here further show that clinically relevant doses of Fuling override the motility-promoting effects of estradiol in endometrial cancer cell lines. Medroxyprogesterone acetate has to date been the standard therapy to treat metastatic or inoperable endometrial cancers; however, success rates are low with high rates of recurrence, due in part to acquired resistance to medroxyprogesterone acetate therapy. The discovery here that Fuling appears to control the spread of treatment-resistant advanced cancers is an exciting prospect.
... In our study, women suffering from endometriosis showed increased serum miR-30c-5p expression. Molecular links exist between cancer and endometriosis, because the main cells involved in both pathologies are able to evade the apoptosis process, share characteristics with stem cells regarding division capacity and standout for a high angiogenic potential [94]. Endometriosis is a benign gynecological condition, although there are studies that relate it to an increased risk of some types of cancer such as ovarian cancer, breast cancer or endometrial cancer [95]. ...
Article
Full-text available
The diagnosis of endometriosis by laparoscopy is delayed until advanced stages. In recent years, microRNAs have emerged as novel biomarkers for different diseases. These molecules are small non-coding RNA sequences involved in the regulation of gene expression and can be detected in peripheral blood. Our aim was to identify candidate serum microRNAs associated with endometriosis and their role as minimally invasive biomarkers. Serum samples were obtained from 159 women, of whom 77 were diagnosed with endometriosis by laparoscopy and 82 were healthy women. First, a preliminary study identified 29 differentially expressed microRNAs between the two study groups. Next, nine of the differentially expressed microRNAs in the preliminary analysis were evaluated in a new cohort of 67 women with endometriosis and 72 healthy women. Upon validation by quantitative real-time PCR technique, the circulating level of miR-30c-5p was significantly higher in the endometriosis group compared with the healthy women group. The area under the curve value of miR-30c-5p was 0.8437, demonstrating its diagnostic potential even when serum samples registered an acceptable limit of hemolysis. Dysregulation of this microRNA was associated with molecular pathways related to cancer and neuronal processes. We concluded that miR-30c-5p is a potential minimally invasive biomarker of endometriosis, with higher expression in the group of women with endometriosis diagnosed by laparoscopy.
... Nevertheless, several theories have been proposed, including retrograde menstruation, dissemination of endometrial cells through the blood or lymphatic system, and coelomic dysplasia [6]. On the other hand, endometriosis also shares characteristics with malignant tissue, such as tissue invasion, induction of angiogenesis, increased proliferative capacity, 2 of 10 ability to evade apoptosis, and ability to form local and distant foci [7,8]. Several studies have established that a history of endometriosis is associated with an increased risk of both endometrioid and clear cell ovarian cancer [9]. ...
Article
Full-text available
Simple Summary Previous studies have shown that women with endometriosis have an increased risk of ovarian cancer. However, it is unclear whether endometriosis is associated with a risk of developing endometrial cancer. Therefore, this study was designed to retrospectively assess the clinicopathological relationship between endometrial cancer and endometriosis using the medical records of patients with endometrial cancer who underwent surgery at our institution. Abstract Endometriosis is known to be associated with an increased risk of endometrioid and clear cell ovarian cancer. However, the association between endometriosis and endometrial cancer is controversial. Therefore, we retrospectively analyzed the medical records of women with endometrial cancer who had undergone surgery at our institution to evaluate the clinicopathological relationship between endometrial cancer and endometriosis. The study included 720 women pathologically diagnosed with endometrial cancer at our hospital between 2000 and 2020. The participants were allocated to two groups of patients with endometrial cancer: patients with endometriosis (n = 101) and patients without endometriosis (n = 619). Endometrial cancer patients with endometriosis were significantly younger (median age 54.0 vs. 58.0; p = 0.002). In addition, endometrial cancer patients with endometriosis had fewer pregnancies and deliveries (median pregnancy 1.58 vs. 1.99; p = 0.019, median delivery 1.25 vs. 1.56; p = 0.012). The percentage of patients classified as stage IA was significantly higher in those with endometrial cancer with endometriosis (68.3% vs. 56.4%; p = 0.029). In the analysis of synchronous ovarian cancer, the percentage of dual primary cancer was higher in patients with endometriosis (14.9% vs. 1.6%; p < 0.001). The association of young-onset early-stage endometrial cancer with endometriosis is an important finding that cannot be ignored clinically.
... The risk of malignant transformation of premenopausal endometriosis is around 1% occurring most commonly in ovarian lesions [14]. Furthermore, patients with endometriosis have an Society reported a position statement regarding to managing menopause in women with previous history of endometriosis [16]. ...
... Other features shared by endometriosis and cancer are the ability to evade apoptosis, the stem cell-like ability, and angiogenic potential. 204 Endometriotic cells have all the characteristics of neoplastic cells, viz., uncontrolled proliferation, invasion, and metastasis at the macro level, whereas aneuploidy, copy number variations (20q13.33 duplication), and mutations in epithelial component as well as epigenomic changes in stromal component at the molecular level and still not categorized as neoplastic condition, at least low grade locally invasive category. ...
Article
Full-text available
Genetics and genomics play a role in the causation of various human diseases. A large number of human reproductive disorders also arise as a result of genetic and genomic abnormalities. Reproductive disorders associated with predominantly genetics and genomic abnormalities are infertility, early pregnancy loss, congenital malformations, difference or disorder of sex development and reproductive cancers. The genetic etiology of human reproductive disorders is increasing with improved molecular biology techniques such as DNA microarray and next-generation sequencing.
... Stage II tumors typically invade into cervical stromal connective tissue, but without invading beyond the uterus, while stage III tumors may invade local or regional structures beyond the uterus including the lymph nodes. While adenomyosis and endometriosis are not as aggressive as endometrial cancers, these two pathological tissues share several cellular characteristics with malignant endometrial cancer tissue including invasion, migration and augmented cell survival [6,7]. Further, adenomyosis and endometriosis are often comorbid with one another [8], and both diseases have been associated with endometrial cancer [9]. ...
Article
Full-text available
Adenomyosis is defined as the development of endometrial epithelial glands and stroma within the myometrial layer of the uterus. These “ectopic” lesions share many cellular characteristics with endometriotic epithelial cells as well as endometrial adenocarcinoma cells, including enhanced proliferation, migration, invasion and progesterone resistance. We recently reported that the 60S acidic ribosomal protein P1, RPLP1, is up-regulated in endometriotic epithelial cells and lesion tissue where it plays a role in cell survival. To evaluate if a similar pattern of expression and function for RPLP1 exists in adenomyosis and endometrial cancer, we examined RPLP1 expression in adenomyosis and endometrial cancer tissue specimens and assessed its function in vitro using well-characterized cell lines. A total of 12 control endometrial biopsies and 20 eutopic endometrial and matched adenomyosis biopsies as well as 103 endometrial adenocarcinoma biopsies were evaluated for RPLP1 localization by immunohistochemistry. Endometrial adenocarcinoma cell lines, Ishikawa, HEC1A, HEC1B and AN3 were evaluated for RPLP1 protein and transcript expression, while in vitro function was evaluated by knocking down RPLP1 expression and assessing cell survival and migration. RPLP1 protein was up-regulated in eutopic epithelia as well as in adenomyosis lesions compared to eutopic endometria from control subjects. RPLP1 was also significantly up-regulated in endometrial adenocarcinoma tissue. Knockdown of RPLP1 in endometrial adenocarcinoma cell lines was associated with reduced cell survival and migration. RPLP1 expression is up-regulated in eutopic and ectopic adenomyotic epithelia as well as in the epithelia of endometrial cancer specimens. In vitro studies support an essential role for RPLP1 in mediating cell survival and migration, processes which are all involved in pathophysiology associated with both diseases.
... Endometriosis is a benign disease that holds a series of malignant characteristics, including apoptosis resistance, metastasis, invasion, and angiogenesis [136]. Angiogenesis is defined as the growth of new blood vessels. ...
Article
Endometriosis is a common condition in women of reproductive age, but its current interventions are unsatisfactory. Recent research discovered a dysregulation of the sphingosine 1-phosphate (S1P) signaling pathway in endometriosis and showed a positive outcome by targeting it. The S1P axis participates in a series of fundamental pathophysiological processes. This narrative review is trying to expound the reported and putative (due to limited reports in this area for now) interactions between the S1P axis and endometriosis in those pathophysiological processes, to provide some perspectives for future research. In short, S1P signaling pathway is highly activated in the endometriotic lesion. The S1P concentration has a surge in the endometriotic cyst fluid and the peritoneal fluid, with the downstream dysregulation of its receptors. The S1P axis plays an essential role in the migration and activation of the immune cells, fibrosis, angiogenesis, pain-related hyperalgesia, and innervation. S1P receptor (S1PR) modulators showed an impressive therapeutic effect by targeting the different S1P receptors in the endometriosis model, and many other conditions resemble endometriosis. And several of them already got approval for clinical application in many diseases, which means a drug repurposing direction and a rapid clinical translation for endometriosis treatments.
... Among them, the p53 gene is a kind of pathogenic gene that is strongly linked to tumor incidence, and its unique single nucleotide polymorphism is linked to the prevalence of female endometriosis. [10,11] (see Figure 1). Terefore, many doctors and researchers have carried out clinical research on the problem and made research progress to varying degrees [12,13]. ...
Article
Full-text available
Background: The P53 gene is critical to the onset and progression of cancers. Currently, relevant study findings indicate that the p53 gene may have a strong association with the risk of endometriosis, but these findings have not been united. To gather more statistically meaningful clinical data, we used meta-analysis to examine the relationship between the rs1042522 single nucleotide polymorphism of the tumor suppressor gene p53 and the incidence of endometriosis. Methods: Through a comprehensive literature survey of PubMed, MEDLINE, EMBASE, Springer, and Web of Science literature databases, we obtained a clinical control case study on the relationship between p53 gene polymorphism and the prevalence of female endometriosis and finally traced the relevant references included. The quality of the literature included in this study was evaluated, and Revman5.3 was used to complete the meta-analysis. Results: This research includes eight publications. The total number of cases in the study group was 1551, whereas the total number of cases in the control group was 1440. The findings of the sensitivity analyses of each omitted piece of the literature revealed no significant difference. The results of the meta-analysis showed that there were significant differences in the GG gene frequency (OR = 0.56, 95%CI (0.38, 0.92), P = 0.003), allele G (OR = 2.46, 95%CI (1.41,4.29), P = 0.002), and allele C (OR = 0.62, 95%CI (0.46, 0.84), P = 0.002) between the study group and the control group (P < 0.01), but there was no significant difference in the GC gene frequency (OR = 1.17, 95%CI (1.01,1.36), P = 0.03), and the CC gene frequency (OR = 1.25, 95%CI (0.85,1.82), P = 0.26) (P > 0.01). Conclusion: Our study results show that there is a significant correlation between the single nucleotide of the p53 gene and the incidence rate of female endometriosis, in which the decrease of the GG gene frequency and the increase of allele C are likely to increase the risk of such diseases.
Article
Endometriosis consists of ectopic endometrial epithelial cells (EEECs) and ectopic endometrial stromal cells (EESCs) mixed with heterogeneous stromal cells. To address how endometriosis‐constituting cells are different from normal endometrium and among endometriosis subtypes and how their molecular signatures are related to phenotypic manifestations, we analyzed ovarian endometrial cyst (OEC), superficial peritoneal endometriosis (SPE), and deep infiltrating endometriosis (DIE) from 12 patients using single‐cell RNA‐sequencing (scRNA‐seq). We identified 11 cell clusters, including EEEC, EESC, fibroblasts, inflammatory/immune, endothelial, mesothelial, and Schwann cells. For hormonal signatures, EESCs, but not EEECs, showed high estrogen signatures (estrogen response scores and HOXA downregulation) and low progesterone signatures ( DKK1 downregulation) compared to normal endometrium. In EEECs, we found MUC5B ⁺ TFF3 low cells enriched in endometriosis. In lymphoid cells, evidence for both immune activation (high cytotoxicity in NK) and exhaustion (high checkpoint genes in NKT and cytotoxic T) was identified in endometriosis. Signatures and subpopulations of macrophages were remarkably different among endometriosis subtypes with increased monocyte‐derived macrophages and IL1B expression in DIE. The scRNA‐seq predicted NRG1 (macrophage)‐ ERBB3 (Schwann cell) interaction in endometriosis, expressions of which were validated by immunohistochemistry. Myofibroblast subpopulations differed according to the location (OECs from fibroblasts and SPE/DIEs from mesothelial cells and fibroblasts). Endometriosis endothelial cells displayed proinflammation, angiogenesis, and leaky permeability signatures that were enhanced in DIE. Collectively, our study revealed that (1) many cell types—endometrial, lymphoid, macrophage, fibroblast, and endothelial cells—are altered in endometriosis; (2) endometriosis cells show estrogen responsiveness, immunologic cytotoxicity and exhaustion, and proinflammation signatures that are different in endometriosis subtypes; and (3) novel endometriosis‐specific findings of MUC5B + EEECs, mesothelial cell‐derived myofibroblasts, and NRG1‐ERBB3 interaction may underlie the pathogenesis of endometriosis. Our results may help extend pathologic insights, dissect aggressive diseases, and discover therapeutic targets in endometriosis. © 2023 The Pathological Society of Great Britain and Ireland.
Article
The influence of vascular endothelial growth factor (VEGF) expression and microvessel density (MVD) on prognosis and the relationship between VEGF expression and MVD in ovarian carcinoma are not well defined. We studied VEGF expression in parallel with MVD by immunohistochemistry in 94 ovarian tumours (64 malignant, 13 borderline, and 17 benign) and correlated the results with the clinicopathologic prognostic factors of the disease to clarify their significance in this disease. Assessment of VEGF mRNA isoforms by RT-PCR was also performed. Of the malignant, borderline, and benign ovarian tumours respectively, two (3%), four (31%) and 16 (94%) were negative, 31 (48%), seven (54%) and one (6%) had low expressions. and 31 (48%), two (15%) and none (0%) had high expressions of VEGF. There were significant associations between the VEGF expression and disease stage (P=0.002), histologic grade (P=0.0004), and patient outcome (P=0.0002). MVD did not correlate significantly with the clinicopathologic parameters. Likewise, no correlation was found between MVD and VEGF expression. The survival of patients with high VEGF expression was significantly worse than that of patients with low and negative VEGF expression (P=0.0004). Multivariate analysis revealed that disease stage and VEGF expression were significant and independent prognostic indicators of overall survival time (P=0.008 and P=0.006 respectively). These findings suggest that in conjunction with the established clinicopathologic prognostic parameters of ovarian carcinoma, VEGF expression may enhance the predictability of patients at high risk for tumour progression who are potential candidates for further aggressive therapy. (C) 2000 Cancer Research Campaign.
Chapter
Apoptosis is defined by its morphological features of membrane blebbing, cellular shrinkage, and chromosomal condensation. Endonuclease activation that results in a characteristic 200-bp nucleosomal DNA ladder is a common feature of apoptosis, although not definitive. Intense research has indicated that the cellular events leading to apoptosis are complex and varied, often depending on the cell type and stimulus utilized. However, several aspects of these death pathways are common among various stimuli and cell types. Programmed cell death is a fundamental biological process of all multicellular organisms and plays important roles in tissue homeostasis, host defense, development, metamorphosis, and morphogenesis. In animals, programmed cell death occurs via apoptosis, a morphologically defined form of cell death that has a number of biochemical features. In addition to its physiological roles, apoptosis contributes to several pathological conditions, such as cancer, AIDS, aging, and cardiovascular, neurodegenerative, and autoinmiune diseases. The mechanisms that govern a cellular decision to live or die are complex and tightly regulated by a plethora of molecules with distinct roles in the signaling process. Generally, cell death occurs after an initial apoptotic signal spurs a cascade of subsequent events from which a cell cannot recover.
Article
In a total of 41 endometrial tissue samples, the relationship between telomerase activity and proliferating cell nuclear antigen (PCNA) labelling index was studied. In samples of endometrium from the proliferative phase of the menstrual cycle, telomerase activity was found in 15 out of 17 cases (88%). Two samples from the early proliferative phase showed negative telomerase activity and a low PCNA labelling index. However, three out of 16 samples of early secretory phase endometrium showed telomerase activity and a PCNA labelling index. In mid- to late secretory phase endometrium, in menopausal endometrium and in decidualized endometrium induced by progesterone neither telomerase activity nor PCNA labelling was found. These results suggest that telomerase activity of the endometrium may be correlated with the proliferative potential of the epithelial cells and that its activity may be regulated by oestrogen.
Article
BACKGROUND Malignant transformation is an infrequent but reported complication of endometriosis. Previous reports of these cases have been limited to clinicopathologic studies based on routine histologic examination of these tumors, whereas, to the authors' knowledge, characterization of these lesions based on immunophenotype and hormone receptor and oncoprotein expression has not been described.METHODS Using commercially available monoclonal antibodies, the authors studied three recent cases of adenocarcinoma arising in extragonadal endometriosis using paraffin immunohistochemistry. Proteins examined included different cytokeratin (CK) subtypes, as well as hormone receptor status, proliferation rate, and oncoprotein expression.RESULTSAll three cases presented clinically and macroscopically as colonic masses, and the tumors expressed an endometrial CK phenotype (CK7+, CK20-). In contrast, the adjacent benign colonic epithelium expressed the expected opposite phenotype (CK7-, CK20+). Estrogen receptor (ER) and progesterone receptor (PR) were expressed in one of the three tumors. Interestingly, in the ER/PR negative tumors, receptor expression was present in areas of benign endometriosis adjacent to malignancy, suggesting a loss of receptor expression with malignant transformation. The tumors also were examined for proliferation by Ki-67, and the expression of oncoproteins c-erb B-2, p53, cyclin D1, and bcl-2. All cases of malignancy had a high proliferation rate as measured by Ki-67, which was in contrast to areas of benign endometriosis which had a low proliferation rate. Of the other oncoproteins only p53 protein was detected at a significant level in all three cases. Cyclin D1 was overexpressed in two of the three cases. c-erb B2 and bcl-2 overexpression was not detected.CONCLUSIONS The results of the current study 1) show the utility of CK subtypes in confirming endometrioid phenotype in tumors arising in extragonadal endometriosis with colonic involvement and 2) suggest that loss of hormone receptor expression and p53 oncoprotein abnormalities may be involved as mechanisms in malignant transformation in extragonadal endometriosis. Cancer 1998;83:1163-1169. © 1998 American Cancer Society.