Keywords endometriosis - genetics - epigenetics - molecular - signaling - pathway
Schlüsselwörter Endometriose - Genetik - Epigenetik - molekular - Signal - Weg
Introduction
Endometriosis classification
Endometriosis describes a disease characterized by the colonization of endometrium-like
lesions outside the uterine cavity. Ectopic lesions were thought to represent solely
lesions on the peritoneum of the internal genital organs (endometriosis genitalis
externa), but in the meantime a migration of endometrial-like cells into the myometrium
has been also described, hence rendering adenomyosis uteri (= endometriosis genitalis
interna) a distinct disease entity. However, since endometriotic lesions may also
infiltrate deeply into organs (mostly bowel, bladder or ureter) (deep infiltrating
endometriosis) or even spread to the diaphragm or the umbilicus (extragenital endometriosis),
symptoms are often extremely complicated [1 ]. A clinical/intraoperative distinction is made between the following four major
entities of endometriosis depending on localization and extent: superficial, ovarian,
uterine and deep
infiltrating endometriosis. Deep infiltrating endometriotic lesions exceed the surface
(usually the peritoneum) and invade into neighboring tissue or organs with an infiltration
depth of at least 0.5 cm ([Fig. 1 ]) [2 ]
[3 ]. The most widely used clinical/intraoperative classification is the rASRM score,
the revised classification of the American Society for Reproductive Medicine (formerly
the American Fertility Society) [2 ]
[4 ]. The rASRM score describes peritoneal and ovarian endometriosis. Deep endometriosis
is included in the calculation of the numerical value, but no mapping or classification
can be derived from it. To remedy this
deficiency, a German-speaking working group has developed the Enzian classification.
This classifies deep lesions in 3 anatomical levels or compartments (A: rectovaginal
septum/vagina, B: sacrouterine ligament/pelvic wall, C: rectum). #Enzian represents
since 2021 a novel comprehensive classification that included the superficial endometriosis,
ovarian and with the Enzian classification, hence constituting a more rounded classification
system, which, nevertheless, does not incorporate the two major symptoms of endometriosis:
pain and infertility [5 ].
Fig. 1
Deep infiltrating endometriosis-Intraoperative laparoscopic views (Patient collective-Department
of Gynecology, University Hospital of Erlangen).
Symptoms and diagnosis
Diagnosis of endometriosis is based on a detailed medical history, a thorough gynecological
clinical examination including vaginal and rectovaginal or rectal palpation, a transvaginal
and/or even transrectal sonographic evaluation, a renal ultrasonography with a view
to ruling out asymptomatic urinary retention caused by deep infiltrating endometriosis
of the ureter, magnetic resonance imaging, as well as a histological examination [6 ]
[7 ]
[8 ]. The diagnosis of a deep infiltrating endometriosis is mainly clinical – by describing
the clinical symptoms (although not specific), inspection with two-leaf specula and
vaginal and rectal palpation. Vaginal sonography should be performed first as an imaging
measure, not least because of the simultaneous possibility of
identifying ovarian endometriomas. Furthermore, deep rectal infiltration can be easily
diagnosed by an experienced physician. If rectal endometriosis is suspected, an endosonography
and/or colorectoscopy is often automatically arranged. However, endometrial infiltration
of the mucosa is rather rare. A colorectoscopy should be performed in the presence
of intestinal bleeding and whenever a bowel resection is intended in the case of suspected
bowel infestation in order to rule out primary bowel pathologies such as polyps, tumors
or inflammatory bowel diseases [9 ]. [Fig. 2 ] summarizes the possible diagnostic approaches based on the four major endometriosis
entities. Taken altogether, endometriosis genitalis (including vaginal endometriosis)
is mainly associated with dysmenorrhea and dyspareunia, deep infiltrating endometriosis
correlates with dysuria
and dyschezia, while extragenital endometriosis (in organs other than the bladder
or the bowel) requires a symptom-oriented examination [6 ]
[7 ]
[8 ]
[9 ]
[10 ]. Importantly, superficial peritoneal endometriosis might remain obscure until the
performance of a diagnostic laparoscopy.
Fig. 2
Diagnostic approaches based on the four major endometriotic entities.
Current therapeutic standards
Conservative options include medical and complementary procedures, reproductive medicine
measures as well as multimodal pain management models (i.e. heat application, physical
exercises, etc.) and psychotherapy in the wider context of the bio-psycho-social model.
Surgical options include organ-preserving or radical and, if necessary, interdisciplinary
ablation or excision of endometriosis lesions, preferably in certified endometriosis
facilities [11 ]. Established pharmacologic approaches include either analgesics from the group of
non-steroidal anti-inflammatory drugs (NSAIDs) or hormone therapy. NSAIDs pursue a
symptomatic therapeutic approach [12 ]. As non-hormonal options treat purely symptomatically, hormone therapy is generally
used. Established hormonal options include progesterons in the first-line therapy,
as well as oral
contraceptives and Gonadotropin-Releasing Hormone (GnRH) (ant-)agonists in the second-line
therapy. There are no objectifiable differences with regard to the reduction of typical
pain symptoms. There are differences in terms of undesirable side effects, the duration
of possible use and the costs. Drug therapy is only effective while it is being taken,
after which symptoms may recur immediately [13 ]. Progestogens (especially dienogest 2 mg) are the options for first-line therapy.
They produce hypoestrogenism through anovulation. In oral contraceptives, they are
part of a fixed combination of ethinylestradiol or estradiol valerate. When selecting
progestogen, secondary treatment goals such as the treatment of skin blemishes can
also be taken into account. Long-cycle use is more effective than cyclical use in
reducing symptoms typical of endometriosis and should be favored [11 ]
[13 ]
[14 ]
[15 ].
Hallmarks of Endometriosis
Hallmarks of Endometriosis
Over the past years, the scientific community has been able to investigate different
molecular pathways and gain an insight into the (epi-)genetic and/or cellular mechanisms
that seem to play a significant role in the genesis and progression of endometriosis.
Of utmost significance, these pathomechanisms seem to pave new ways in the context
of endometriosis diagnosis (as biomarkers) and therapy (as drug targets). The (epi-)genetic
mechanisms are involved in the immunologic, immunohistochemical, histological, and
biological aberrations of endometriosis [16 ]. Pelvic endometriosis has a complex pathogenesis and pathophysiological features.
Two possible causes of the endometriotic lesions are in situ coelomic metaplasia of
the peritoneal lining and transplantation of endometrial tissue through retrograde
menstruation. In cases of extrapelvic lesions, vascular or lymphatic metastasis most
likely happens
infrequently. Through interacting molecular mechanisms that support cellular adhesion
and proliferation, systemic and localized steroidogenesis, localized inflammatory
response and immune dysregulation, as well as vascularization and innervation, superficial
and deep endometriotic lesions seem to be established and maintained [17 ]. Endometriosis-related signaling pathways included estrogen-2, nuclear factor kappa-light-chain-enhancer
of activated B cells (NF-κB), mitogen-activated protein kinase (MAPK), extracellular-signal
regulated kinase (ERK), phosphatidylinositol 3-kinase (PI3K), protein kinase B (PKB/AKT)
and mechanistic target of rapamycin (mTOR) (PAM), yes-associated protein (YAP), Wnt/β-catenin,
Rho-associated protein kinase (ROCK), transforming growth factor β (TGF-β), vascular
endothelial growth factor (VEGF), nitric oxide (NO), iron, cytokines and chemokines
[18 ]. Despite being a benign condition, endometriosis exhibits malignant traits such
as metastasis, hyperplasia, and cell invasion. This suggests a possible connection
between endometriosis and particular signaling molecules and pathways that influence
the invasion and metastasis of numerous common malignancies. The six biological abilities
that are acquired throughout the multi-step development of human tumors are the hallmarks
of cancer. The defining multiple characteristics provide a framework for understanding
the complexity of neoplastic disease. The ability to maintain proliferative signaling,
avoid growth suppressors, withstand cellular death, permit replicative immortality,
trigger angiogenesis, and initiate invasion and metastasis are a few of them. These
hallmarks are underpinned by inflammation, which supports several hallmark functions,
and genome instability, which produces the genetic diversity that speeds up their
acquisition. Two newer hallmarks of potential generality include reprogramming of
energy metabolism and immune escape [19 ]. In this regard, [Fig. 3 ] summarizes the corresponding hallmarks of endometriosis.
Fig. 3
Hallmarks of endometriosis and their potential targets for therapy. Captions in blue:
completed clinical trials. Captions in green: ongoing clinical trials. Captions in
black: experimental models in vitro/in vivo. Abbreviations: ADAM17 = A Disintegrin
and Metalloprotease 17; COX-2 = Cyclooxygenase 2; E2 = Estrogen 2; EPAC1 = Exchange
Protein Directly Activated by cAMP 1; ERK = Extracellular Signal-regulated Kinase;
FAK = Focal Adhesion Kinase; FGFR2 = Fibroblast Growth Factor Receptor 2; GPR30 =
G Protein-coupled Receptor 30; HIF = Hypoxia-inducible Factor; LXA4 = Lipoxin A4;
MAPK = Mitogen-activated Protein Kinase; METTL3 = Methyltransferase-like 3; miR =
micro RNA; MMP = Matrix Metallopeptidase; NF-κB = Nuclear Factor kappa B; NGF = Nerve
growth factor; NLRP3 = NLR Family Pyrin Domain Containing 3; NO = Nitric Oxide; PGE2
= Prostaglandin E2; PKA = Protein Kinase A; PI3K/Akt/mTOR = Phosphatidylinositol 3-kinase/Protein
Kinase B/mammalian Target of
Rapamycin; PPAR = Peroxisome Proliferator-activated Receptor; PrPC = Cellular Prion
Protein; Rap1 = Ras-associated Protein-1; ROCK = Rho-associated Protein Kinase; S1P
= Sphingosine-1-Phosphate; SERCA = Sarcoplasmic/endoplasmic Reticulum Ca2+-ATPase;
SF = Steroidogenic Factor; SHH = Sonic Hedgehog; SHP-1 = Src Homology Region 2 Domain-containing
Phosphatase-1; sICAM-1 = soluble form of intercellular adhesion molecule-1; SMAD =
Suppressor of Mothers against Decapentaplegic; SRC = Steroid Receptor Coactivator;
TGF = Transforming Growth Factor; TIMP = Tissue Inhibitor of MMP; TNF = Tumor Necrosis
Factor; VEGFR = Vascular Endothelial Growth Factor Receptor; ZEB1 = Zinc Finger E-Box
Binding Homeobox 1
Current (Pre-)Clinical Trials Investigating the Hallmarks of Endometriosis
Current (Pre-)Clinical Trials Investigating the Hallmarks of Endometriosis
In 2016, the kinase signaling pathways in endometriosis were investigated and it was
concluded that the three main pathways to be targeted for treatment purposes are the
IKKβ/NFκB, the MAPK, and the PI3K/AKT/mTOR pathway [20 ]. The literature on medications that specifically target the molecular and signaling
pathways involved in the pathophysiology of endometriosis was thoroughly reviewed.
The discussion included possible therapeutic targets, the molecules upstream and downstream
that exhibit critical aberrant signaling, and the regulatory pathways that facilitate
the expansion and maturation of endometriotic tissues and cells [21 ]. Recently, Shi also examined angiogenesis, lymphangiogenesis, neurogenesis, progesterone
resistance, genetic alterations, estrogen-dependent induction of inflammation, imbalances
in proliferation and
apoptosis, and tissue remodeling in the pathogenesis of endometriosis. Additionally,
the pharmacological mechanisms, constitutive relationships, and potential applications
of each compound were studied as well [22 ]. Based on these works, a thorough search of both the ClinicalTrials.gov and the
European Union Clinical Trials Register was conducted with a view to identifying completed
and ongoing clinical studies investigating the role of the aforementioned pathways
in patients with endometriosis. [Table 1 ] and [Table 2 ] briefly summarize the search results. [Table 3 ] provides a brief overview of the relevant preclinical studies.
Table 1
Completed registered clinical trials on the role of signaling pathways in endometriosis.
Agent
Pathway
Completed Clinical trial
Abbreviations: E2 = Estrogen 2; VEGF = Vascular Endothelial Growth Factor
Relugolix
Triptorelin
Linzagolix
ASP1707
FOR-6219
Leuprolide
Elagolix
Sufugolix
MK-8342B
Progesterone/Estradiol
GnRH-agonist
E2
NCT03204331
NCT03204318
NCT03654274
NCT03232281
NCT03992846
NCT01767090
NCT03709420
NCT02807363
NCT00797225
EudraCT Number: 2004–003829–28
EudraCT Number: 2004–001721–13
EudraCT Number: 2012–002791–14
EudraCT Number: 2012–002449–40
EudraCT Number: 2010–019287–37
EudraCT Number: 2013–003788–67
EudraCT Number: 2015–004326–34
EudraCT Number: 2015–004325–14
EudraCT Number: 2013–000993–32
Ezetimibe
Quinagolide
VEGF
NCT04844996
NCT00625950
EudraCT Number: 2018–000915–26
Anti-TNFα
Cytokines
NCT00604864
Table 2
Ongoing registered clinical trials on the role of signaling pathways in endometriosis.
Agent
Pathway
Clinical trial
Abbreviations: CXCL3 = Chemokine Ligand 3; GnRH = Gonadotropin-Releasing Hormone;
IFN = Interferon; JNK = c-Jun N-terminal kinase; MAPK = Mitogen-Activated Protein
Kinase; MMP = Matrix-Metalloprotease; NF-κB = Nuclear factor kappa-light-chain-enhancer
of activated B cells; STAT3 = Signal transducer and activator of transcription 3;
TNFα = Tumor Necrosis Factor-α; VEGFR = Vascular Endothelial Growth Factor Receptor;
TGF = Transforming Growth Factor
Anastrazole plus GnRH agonist
Hormonal
Phase 4
NCT01769781
Danazol
Hormonal
Phase 4
NCT05697471
Resveratrol
Regulation of antioxidant enzymes, TNFα-mediated cytokines
Phase 4
NCT02475564
Vitamin D3 and fish oil
Anti-inflammatory
Phase 4
NCT02387931
Quinagolide
Dopaminergic, VEGF/VEGFR2
Phase 4
NCT03692403
DLBS1442
Anti-inflammatory, antiangiogenic, and apoptosis-inducing
Phase 3
NCT01942122
Pentoxifylline
VEGFC and VEGFR2
Phase 3
NCT00632697
Cabergoline
Dopaminergic
Phase 2
NCT02542410
Botulinum toxin
Neurotoxic
Phase 2
NCT01553201
Gefapixant
P2X3 receptor antagonist
Phase 2
NCT03654326
Vilaprisan
Selective progesterone receptor modulator
Phase 2
NCT03573336
Epigallocatechin gallate
TGF-β1-stimulated activation of MAPK and Smad pathway, VEGFC-mediated c-JUN, IFN-γ,
CXCL3, and MMP-9 pathway
Phase 2
NCT02832271
Melatonin
Caspase, Radical scavenging activity
Phase 2
NCT03782740
MT-2990
Fully human anti-interleukin-33 monoclonal antibody
Phase 2
NCT03840993
Curcumin
p53/NF-κB, IκKα/β, STAT3, and JNK
Recruiting
NCT03016039
Table 3
Preclinical studies on the role of signaling pathways in endometriosis.
Agent
Pathway
Preclinical study
(cell culture/animal model/tissue sample)
Abbreviations: CBP = CREB binding protein; CDK = Cyclin dependent kinase; COX = Cyclooxygenase;
CXCL3 = Chemokine (C-X-C motif) ligand 3; ER = Estrogen receptor; FOXO = Forkhead
box transcription factors; GnRH = Gonadotropin-releasing hormone; IFN = Interferon;
IκK = Inhibitor of kappa kinase; iNOS = Inducible-NO synthase; JNK = Jun N-terminal
kinase; LATS1 = Large tumor suppressor kinase 1; LHRH = Luteinizing hormone-releasing
hormone; MAPK = Mitogen-activated protein kinase; MMP = Matrix metallopeptidase; mTOR
= Mammalian target of rapamycin; NF-κB = Nuclear factor kappa-light-chain-enhancer
of activated B cells; NK = Natural killer; NO = Nitric oxide; PDTC = Pyrrolidine dithiocarbamate;
PGE = Prostaglandin; PPARγ = Peroxisome proliferator-activated receptor gamma; RAF/MEK/ERK
= Rapidly accelerated fibrosarcoma/mitogen-activated protein kinase/extracellular
signal-regulated kinase; ROCK = Rho-associated, coiled-coil containing kinases; SMAD
= Suppressor of mothers
against decapentaplegic; STAT3 = Signal transducer and activator of transcription
3; TGF = Transforming growth factor; TNFα = Tumor necrosis factor α; TPCK = Tosyl
phenylalanyl chloromethyl ketone; VEGF = Vascular endothelial growth factor; WNT2
= Wnt family member 2; YAP1 = Yes-associated protein 1
Indomethacin
COX-2
Mice model
Celecoxib
COX-2/PGE2, COX-2/VEGF
Primary human endometriotic stromal cells
BAY11-7085
Caspase and apoptotic proteins effects
Primary human endometriotic and endometrial stromal cells
Chloroindazole
E2/ER
Primary human endometriotic stromal cells and mice model
Oxabicycloheptene sulfonate
E2/ER
Primary human endometriotic stromal cells and mice model
Tunicamycin
ER stress
Primary human endometriotic and endometrial stromal cells
Verteporfin
Hypoxia/LATS1/YAP1
Primary human endometriotic stromal cells and mice model
Curcumin
p53/NF-κB, IκKα/β, STAT3, and JNK
Mice model
Genistein
COX-2 and NF-κB/MMP-2/MMP-9
Mice model
Sorafenib
RAF/MEK/ERK and VEGF/VEGFR
Primary human endometriotic stromal cells and mice xenograft model
Vemurafenib
MAPK/ERK
Primary human stromal epithelial; endometriotic/endometrial cells and animals mice
xenograft model
U0126
MAPK/PR
Primary human endometriotic and endometrial stromal cells
Puerarin
MAPK/ERK1/2
Primary human endometriotic stromal cells
PGE2 inhibitors
EGFR/ERK1/2, Akt, B-catenin, NF-κB
Cell line (12Z and 22B), primary human endometriotic and endometrial stromal cells
WIN 55212-2
mTOR/Akt
Primary human stromal/epithelial; endometriotic/endometrial cells and mice xenograft
model
Propofol
p53, p21, Caspase, FOXO, inducing apoptosis
Cell line (CRL-7566)
Dichloroacetate
Metabolic process
Primary human peritoneal mesothelial cells, cell line (SHT290), mice model
MK2206 and chloroquine
Akt/PR, Autophagy modulators regulated autophagy
Primary human stromal/epithelial; endometriotic/endometrial cells and mice xenograft
model
Ginsenoside
NF-κB, E2/ER and PR, NK cells cytotoxicity
Primary human endometriotic and endometrial stromal cells and mice model
Müllerian inhibiting substance
ERK and Beclin1 inducing autophagy, CDK
Cell line (CRL-7566)
C-82
CBP/β-catenin
Primary human endometriotic and endometrial stromal cells
ICG-001
CBP/β-catenin
Primary human endometriotic and endometrial stromal cells and mice model
Metformin
Wnt2/β-catenin, cytokines
Primary human endometriotic and endometrial stromal cells, endometrial epithelial
cell
PKF115-584/ CGP049090
Wnt/β-catenin
Primary human stromal/epithelial; endometriotic/endometrial cells
Fasudil
Rho/ROCK
Primary human endometriotic stromal cells
Heparin
Rho/ROCK
Primary human endometriotic stromal cells
Pazopanib, sunitinib and sorafenib
VEGF/VEGFR
Rat model
Pyrrolidine dithiocarbamate
NF-κB/TNFα/VEGF
Primary human endometriotic and endometrial stromal cells
Pentoxifylline
VEGFC and VEGFR2
Wistar rat model
N-acetylcysteine
Radical scavenging activity/ERK, cytokines
Primary human stromal/epithelial; endometriotic/endometrial cells, mice xenograft
model
Caffeic Acid
Regulation of antioxidant enzymes
Primary human endometriotic and endometrial stromal cells
Crocin
Cytokines
Mice model and cell lines (HUVEC and THP-1)
ISO-1
Cytokines
Mice model
Puerarin
E2/ER
Rat model
Niclosamide
MAPK, Wnt pathway
Mice model
Acai
VEGF/VEGFR, iNOS/NO, COX-2/PGE2
Cell line (J774.G8) and Sprague-Dawley rats
Bortezomib
Proteasome
Wistar rats
TPCK
NF-κB
Endometriosis stromal cells
PDTC
IkappaB
Endometriosis stromal cells and Wistar rats
Thalidomide
IkappaB
Endometriosis stromal cells and Sprague-Dawley rats
Thiazolidinediones
PPARγ
Sprague-Dawley rats
Interleukin 10
DNA binding
Endometriosis stromal cells
Decoy Nucleotides
DNA binding
Endometriosis stromal cells
SB203580
Interleukin 1β
Endometriosis stromal cells
SP600125
JNK
Endometriosis stromal cells
Temsirolimus
mTOR
Endometriotic cells
Discussion and Conclusion
Discussion and Conclusion
A great number of extensive review articles has so far been published on the role
of signaling pathways/molecules in endometriosis [16 ]
[17 ]
[18 ]
[19 ]
[20 ]
[21 ]
[22 ]. Of note, given some shared molecular (genetic) mechanisms, endometriosis seems
to be associated with various risk factors and other disease entities such as migraine,
autoimmunity and chronic pelvic pain [23 ]
[24 ]
[25 ]
[26 ] ([Fig. 4 ]). Chronic pain, for instance, seems to share similar pathomechanisms as endometriosis
in terms of abundance of proinflammatory molecules, angiogenesis and estrogen-dependent
pain meditation [23 ]. Even though endometriosis is not yet officially classified as an autoimmune disease,
there are a number of similarities between the two conditions, including a predominance
of females (and hormones), immunological abnormalities, genetic polymorphisms, as
well as chronicity [24 ]. In the case of migraine, mechanisms associated with sex hormone activities, protein
adhesion,
phosphorylation, inflammation or immune dysregulation seem to play a similar role
as in the pathogenesis of endometriosis [25 ]. Endometriosis is a disease condition encountering gynecologists every single day
in both the outpatient and the clinic routine. Patients seek medical advice either
because of the adverse pain symptoms and/or due to the unfulfilled desire to become
pregnant. Unfortunately, most patients are very disappointed once they learn that
surgery does not grant the end of the disease and that the only possible symptomatic
treatment is hormone-based. In times of targeted treatment therapies and ample possibilities
to investigate and discover novel therapeutic approaches (i.e. inflammation, apoptosis,
angiogenesis, cellular adhesion, etc.), endometriosis represents a profound example
of an understudied disease that to date may only be treated symptomatically. The present
work aims at raising the
awareness of both researchers and clinicians in this context and to highlight the
need of further research in order to establish and launch targeted therapies for the
successful treatment of endometriosis patients. All in all, we herein intended to
summarize the current research status and to point out the field’s novel therapeutic
approaches. However, the considerable side effects of these targeted therapies need
to be further examined and taken into consideration in the context of risk–benefit
calculation.
Fig. 4
Risk factors and associated conditions with endometriosis [14 ]
[17 ]
[23 ]
[24 ]
[25 ]
[26 ].