Open Access

Angiogenesis signaling in endometriosis: Molecules, diagnosis and treatment (Review)

  • Authors:
    • Caixia Bo
    • Yunfei Wang
  • View Affiliations

  • Published online on: January 18, 2024     https://doi.org/10.3892/mmr.2024.13167
  • Article Number: 43
  • Copyright: © Bo et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY_NC 4.0].

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endometriosis (EM) is one of the most common diseases among women of reproductive age. The etiology and pathogenesis of EM remain unclear and therefore there is a lack of effective treatment measures, which affects physical and mental health, as well as the quality of life of patients with EM. Angiogenesis has become a hotspot for research on the pathogenesis of EM; the role of angiogenesis‑related serological markers and anti‑angiogenic therapy in the diagnosis and treatment of EM is promising for early diagnosis and treatment of EM. Angiogenesis in EM is subject to complex regulation by hormones, immunity and associated cytokines. Therefore, novel targets for angiogenesis therapy are also being discovered and developed. The present review summarized the pathological mechanisms of angiogenesis and the value of relevant markers in pathogenesis and diagnosis of EM, along with the status of research on anti‑angiogenic drugs in the treatment of EM. The role of angiogenesis in EM provides an important reference for treatment and diagnosis, but there is no uniform non‑invasive diagnostic marker and proven strategy for anti‑angiogenesis.

Introduction

Endometriosis (EM) is an estrogen-dependent disorder characterized by the presence of endometrioid epithelium and glands outside the endometrium and myometrium (1). It affects 10–15% of women of reproductive age worldwide and is one of the most frequently occurring diseases among this group (1). The main clinical manifestation of EM is pain, including typical but non-specific pain, such as dysmenorrhea and chronic pelvic pain, and ~1/3 of patients have infertility, with advanced EM potentially leading to gynecological malignancy, such as ovarian cancer (2,3). The lack of specific clinical manifestations of EM makes it difficult to differentiate from conditions characterized by chronic pelvic pain in clinical practice, which makes the diagnosis and treatment of EM challenging (3). This results in psychological burden to patients and affects physical and mental health, as well as their quality of life (4,5).

The pathophysiology of EM disease is not thoroughly understood. The potential pathogenesis of EM is hypothesized to involve retrograde menstruation, uterine stem cells and somatic epithelium (1). Among them, the theory of retrograde menstruation is the leading theory of the pathogenesis of EM (6). However, development of EM is associated with immune alterations and a proinflammatory peritoneal milieu, which determines the progression to EM (7). It is noteworthy that the formation and growth of ectopic lesions depends on nutrient support, with neovascularization serving an important role in the development of endothelial ectopic lesions (2,8). Vascular permeability is notably increased in ectopic lesions of EM and a large number of new blood vessels appear (2). This indicates that angiogenesis is a key link in the pathogenesis of EM, which represents a potential target for development of future diagnostic and therapeutic strategies. Recent study of angiogenic mechanisms in EM has revealed the importance and potential role of angiogenesis-related pathways and markers of EM for diagnosis and treatment (912). That has provided a new direction for the discovery of novel targets of anti-angiogenic action and therapy and novel lines of thought and strategies for clinical treatment of EM.

The present review aimed to summarize advances in the investigation of EM angiogenesis, including availability of non-invasive molecular markers for the diagnosis of EM, for the potential clinical value of anti-angiogenic therapy as an alternative to non-hormonal therapies for treatment of EM.

Search methods

Literature searches were performed in PubMed (pubmed.ncbi.nlm.nih.gov/) and ISI Web of Knowledge (webofscience.com) for English articles with the key words ‘endometriosis’, ‘endometriotic lesions’, ‘angiogenesis’, ‘vascularization’, ‘anti-angiogenic’, ‘anti-angiogenesis’, ‘anti-angiogenic therapy’ and ‘diagnosis’. The search included animal and human studies focusing on EM angiogenesis, diagnosis and treatment, and no restriction was set regarding the publication date.

Angiogenesis in EM

Angiogenesis is defined as generating new blood vessels from the germination of pre-existing vessels. This is initiated by vascular endothelial growth factor (VEGF), which activates resting endothelial cells in microvasculature to release matrix metalloproteinase (MMP), thereby mediating degradation of the vascular basal membrane so that endothelial cells migrate into the surrounding tissue; additionally, tubular branches of endothelial cells form vascular rings and novel basement membranes, ultimately leading to new vessel formation (13). Angiogenesis is key for normal physiological processes by spontaneous endometrial thickening and development, serving an important role in the menstrual cycle (14). In patients with EM, the endometrium forces menstrual blood to the peritoneal cavity leading to induction of oxidative stress and immune responses, releasing related inflammatory factors and cytokines. This results in disrupted homeostasis of the peritoneal microenvironment and a relative increase in pro-angiogenic factors, which leads to neovascularization of ectopic endometrial vessels and establishment of the microvascular network (2,15). EM lesions develop in a complex and dynamic environment that is regulated by a number of molecules, cytokines and associated signaling pathways (Table I) (16).

Table I.

Expression of pro-angiogenic factors in intimal tissue, peritoneal fluid or serum/plasma of patients with EM.

Table I.

Expression of pro-angiogenic factors in intimal tissue, peritoneal fluid or serum/plasma of patients with EM.

FactorSourceTest sample(Refs.)
VEGFEndometrial glandular epithelial and stromal cells, macrophages, neutrophilsIntimal tissue, serum, peritoneal fluid(10,21,24,26,50)
FGFStromal cells, macrophagesIntimal tissue(10)
AngEndothelial cellsSerum(27)
HIF-1αStromal cellsSerum(24)
IL-1βMacrophages, monocytesPeritoneal fluid(20)
Serum(26,27)
IL-6Macrophages, mast cells, lymphocytesPeritoneal fluid(21,23)
Serum(104)
IL-8Endothelial cells, macrophages, neutrophilsPeritoneal fluid(22)
Serum(22,26,27)
IL-10Plasma cells, dendritic cells, NK cellsSerum(26)
IL-17Lymphocytes, neutrophilsPeritoneal fluid(23)
Serum(23)
TNF-αMacrophages, NK cellsIntimal tissue(25)
Serum(26,27)
MMIFLymphocytes, macrophagesSerum(24)
COX-2MacrophagesSerum(26)

[i] VEGF, vascular endothelial growth factor; FGF, fibroblast growth factor; Ang, angiopoietin; NK, natural killer; HIF-1α, hypoxia-inducible factor-1α; IL, interleukin; TNF-α, tumor necrosis factor-α; MMIF, macrophage migration inhibitory factor; COX-2, cyclooxygenase 2.

Role of inflammatory cells in angiogenesis progression in EM

At present, although the etiology of EM is unclear, immune dysfunction has been cited as a key contributing factor in the growth of ectopic lesions of endometrial debris following retrograde menstruation (17,18). Density of immunocompetent cells such as T lymphocytes, natural killer cells, dendritic cells (DCs), macrophages and neutrophils is markedly increased in EM lesions and the peritoneal cavity (Fig. 1) (17,19). Abnormal alterations in inflammatory and immune cells cause alterations in cytokines in the peritoneal fluid of patients with EM. Compared with healthy patients, the expression of interleukin (IL) −1β, −6, −8, −10 and −17, tumor necrosis factor-α (TNF-α) and macrophage migration inhibitory factor is notably elevated in the peritoneal cavity or serum of patients with EM (2027). These cytokines are not only involved in the chronic inflammatory response to EM, but also promote the implantation and growth of ectopic lesions by upregulating expression of cyclooxygenase 2 (COX-2) and/or VEGF.

Previous studies have suggested that enhanced concentration of neutrophils in the peritoneal fluid of patients with EM may be the result of elevated concentrations of potent neutrophil chemokines, such as IL-8, in plasma and peritoneal fluid (28,29). Moreover, neutrophils secrete cytokines such as VEGF and IL-8 and −17 to promote the proliferation and invasion of endometriotic cells and angiogenesis (30). In this process, estrogen is involved in neutrophil activation, and it was found that in vitro culture in the presence of IL-6, TNF-α, lipopolysaccharide (LPS) and estrogen-enhanced VEGF release from isolated peritoneal macrophages and neutrophils (31). Conversely, inhibition of estrogen signaling activity decreases the number of neutrophils and expression of proinflammatory cytokines, thereby inhibiting the progression of EM (32).

Immune dysfunction characterized by hyperactive peritoneal macrophages with altered phagocytic ability represents a key point in EM angiogenesis (33,34). Macrophages are hypothesized to be one of the sources of VEGF and fibroblast growth factor (FGF), which promote the growth of diseased blood vessels in EM and accelerate the process of EM development (24). Previous studies have found that accumulated macrophages are recruited to the ectopic environment, primarily using the alternatively activated macrophage (M2) phenotype, which promotes and enhances proliferation and clonogenic capacity of endometrial stromal cells (ESCs) (3336). Additionally, M2 cells produce a variety of stimulating factors to induce peritoneal inflammation, thus forming a complex loop of regulatory mechanisms to maintain the altered peritoneal microenvironment of EM to promote immune escape, adhesion and angiogenesis of the ectopic endothelium (31,37). By contrast, nanovesicles derived from M1 macrophages directly or indirectly inhibit migration and invasion of endometrial mesenchymal stromal cells (E-MSCs) to decrease formation of blood vessels (35).

Contrary to macrophages, the role of DCs in pathogenesis of EM is not yet clear. Suen et al (38) showed that IL-10 secreted by plasma cell-like DCs enhances ESC migration and promotes angiogenesis via the secretion of pro-angiogenic factors, leading to the growth of endometriotic lesions. Furthermore, DCs may promote EM angiogenesis by secreting cytokines such as IL-6 and −12 and transforming growth factor-β (TGF-β) (39).

Regulatory T cells and helper T 17 (Th17) cells are essential for immune defense and immune homeostasis (18). Imbalance in expression of Th2 and Th1 cells is one of the essential promoters of ectopic endothelial growth (40). Th17 cells produce IL-17, a strong proinflammatory mediator that stimulates expression of angiogenic and proinflammatory cytokines, and promotes angiogenesis in the ectopic endothelium (30,41). In vitro results suggest that IL-17 may be a stimulus that induces pathogenic polarization of macrophages towards the M2 phenotype (42). This suggests a synergistic role of inflammation and the Th17 immune response.

Molecules involved in angiogenesis in EM

Previous studies have found numerous similarities between EM angiogenesis and tumor pathologic angiogenesis (8,43). Ectopic endometrial tissue initially has a degree of hypoxia comparable with that of cells in growing tumor centers (16,44).

Hypoxia is one of the most potent stimuli for upregulation of angiogenic growth factors; it prevents proteasomal degradation of hypoxia-inducible factor-1α (HIF-1α) (44,45). HIF-1α is required for oxygen-regulated transcriptional activation of genes encoding VEGF to enhance hypoxia-induced angiogenesis. HIF-1α enters the nucleus to form hypoxia response elements and binds the promoter region of the VEGF gene, which in turn upregulates VEGF expression and promotes angiogenesis (9). In the hypoxic microenvironment of the peritoneal cavity, HIF-1α downregulates expression of chicken ovalbumin upstream of the promoter transcription factor II, which leads to elevated angiopoietin expression and promotes neovascularization of ectopic lesions in EM (46). Additionally, HIF-1α is involved in immune regulation and inflammatory responses and it can upregulate expression of IL-8 and COX-2, thus initiating IL-8- and COX-2-mediated angiogenic mechanisms and indirectly promoting ectopic endothelial neovascularization (47). The involvement of HIF-1α in regulating disease is complex and HIF-1α is also involved in cellular autophagy. HIF-1α promotes mesenchymal cell migration and invasion in EM via upregulation of autophagy (45). Reactive oxygen species (ROS) are a key element in stabilizing HIF-1α. The release of ROS and the expression of VEGF are enhanced under hypoxic conditions, which forms a positive feedback mechanism between ROS and VEGF in enhancing angiogenesis (48). As a consequence, HIF-1α is an important regulator that promotes angiogenesis and is involved in angiogenesis in EM via the complex regulation of multiple pathways.

VEGF is an important pro-angiogenic factor that induces vascular endothelial cell neogenesis and cell proliferation by binding to vascular endothelial cells, increases vascular permeability and promotes blood vessel formation (43,49). VEGF protein family includes VEGF-A, -B and -C, virus-encoded VEGF-D and placental growth factor, with VEGF-A playing pivotal roles in regulation of angiogenesis (43). The VEGF signaling pathway regulates activity of multiple kinases by binding to cell surface tyrosine kinase receptors, VEGF receptor (R)-1, VEGFR-2, and VEGFR-3, resulting in different biological effects that ultimately result in angiogenesis (43). In recent years, expression of VEGF has been studied in different experimental EM models and tissue samples from patients with EM (10,5052). Numerous researchers have found that increased expression of VEGF in the serum and peritoneal fluid of patients with EM can serve as an auxiliary diagnostic indicator of EM (9,10,50). Li et al (10) found that angiogenic factors released from ESCs may be influenced by expression of fibrinogen α chain (FGA) and that the expression of pro-angiogenic factors, such as VEGF, platelet-derived growth factor (PDGF), FGF and MMP-2 and −9, is reduced in FGA-knockdown hEM15A cells. FGA may activate the VEGFA/VEGFR-2/FAK signaling pathway in EM and promote angiogenesis by regulating expression of VEGF-A and MMPs in EM ESCs. In addition, VEGF-C expression is upregulated in EM cells, and it has been demonstrated that VEGF-C enhances lymphangiogenic capacity of lymphatic endothelial cells via extracellular vesicle transport in an autograft mouse model of EM, whereas blocking the VEGF-C signaling pathway attenuates development of local chronic inflammation and EM (50). The expression of VEGF in EM is regulated by expression of multiple factors and related pathways and plays a vital regulatory role in the process of neovascularization in EM.

The stimulation of endometrial and endometriotic cells leads to activation of different intracellular pathways and associated signaling molecules. Nuclear factor-κB (NF-κB) is a transcription factor that mediates inflammatory signaling pathways and is a driver of inflammation and plays a crucial role in the development and regulation of inflammation (2). NF-κB is expressed in trace amounts in normal endometrium, but it is expressed at high levels in EM, suggesting that activated NF-κB serves an important role in regulating the development of ectopic endometrium (26,53). Gou et al (33) showed that estrogen receptor β (ERβ) regulates production of C-C motif chemokine ligand 2 through activation of the NF-κB signaling pathway in B cells, thereby recruiting macrophages to ectopic lesions in ESCs to promote pathogenesis. Additionally, ERβ stimulates expression of genes associated with the unfolded protein response in normal endometrium, inhibits the IL-6/JAK/STAT3 signaling pathway and suppresses the TNFα/NF-κB signaling pathway, leading to endometrial dysfunction associated with EM (54). In addition, oxidative stress is associated with development of EM and is a key inducer of the NF-κB signaling pathway in EM cells, extracellular high mobility group box-1 (HMGB-1), a prototypical molecule of damage-associated molecular patterns, inducing an inflammatory response via HMGB-1/TLR4/NF-κB axis (2,53). Excessive production of ROS in the pelvis of patients with EM is an important inducer of NF-κB-mediated chronic inflammatory responses, and NF-κB activation is primarily regulated by ROS, regulating the expression of cytokines, such as IL-10, in EM by activating the NF-κB pathway (26). Oxidative damage to DNA in EM increases DNA fragmentation and activates NF-κB signaling to promote transcription and expression of inflammatory factors such as TNF-α, and IL-1, −8 and −6 (26,55), while NF-κB is activated by TNF-α, IL-1β and LPS (56), thus creating positive feedback to promote alteration of the abdominal inflammatory microenvironment and the development of ectopic endothelial lesions. Additionally, the NF-κB signaling pathway decreases expression of antioxidant enzymes by activating the nitric oxide synthase (NOS)/NO signaling pathway, which is involved in oxidative stress (53).

In addition to VEGF, other factors have been shown to be involved in angiogenesis in EM. COX-2 serves as a rate-limiting enzyme for prostaglandins (PGs), induces the production of PGE2 and E2, which increase VEGF expression (57). COX-2 is rapidly upregulated in response to proinflammatory and pathogenic stimuli, and it is able to induce VEGF synthesis upon stimulation by cytokines (26,57) Furthermore, COX-2 has been shown to serve a vital role in the pathological process of diseases such as endometrial carcinoma and endometrial carcinoma (53,58). COX-2, which is either not expressed or expressed at low levels in normal endometrium, is aberrantly expressed in patients with EM and may regulate VEGF to serve a role in the process of disease development (54,57). MMPs play a key role in various pathophysiological processes, such as adhesion and invasion of ectopic endothelium (13,59). Furthermore, COX-2 can regulate angiogenesis via regulation of MMP-2 activity in EM by the COX-2/PGE2/phosphorylated AKT axis (60). Blocking the expression of COX-2 and/or AKT inhibits MMP-2 activity and endothelial tube formation and inhibition of MMP-2 and COX-2 notably decreases the number of lesions in a mouse model of EM (60).

In conclusion, the pathogenesis of EM is complex and its angiogenesis is driven by a variety of cytokines mediating pro- and anti-angiogenic effects, such as hormonal, immune and inflammatory oxidative stress. These cytokines interact with each other to form a complex network of regulatory mechanisms and are involved in development of neovascularization in EM via multiple cellular pathways (Fig. 2). Further studies on pathogenesis of EM will contribute to identification of biomarkers or biomarker groups to improve and the diagnostic process in a non-invasive manner.

New non-invasive biomarkers in EM

At present, reliable laboratory biomarkers for EM pathology are difficult to obtain and there are no uniform international standards (1). The gold standard for diagnosis of EM remains the pathological findings following surgical treatment (61,62). There is often a 6–12-year hiatus between onset and diagnosis of EM, with the delay being caused by multiple factors, including economic deprivation and uneven distribution of health services (63,64). Reliable biomarkers found in the biological fluids of affected patients may be an expedient diagnostic tool for EM, which would also facilitate an objective assessment of treatment efficacy.

Although lacking both specificity and sensitivity for this pathology, the most representative glycoproteins used as biomarkers for EM include cancer antigen CA-125 and CA-199, which have similar specificity and reflect the severity of disease (65,66). However, in total laparoscopic ectopic lesion removal, high CA-199 expression is markedly associated with a high postoperative recurrence rate; monitoring the expression of CA-199 may help predict the progression of EM (67).

Vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) are members of the integrin adhesion protein family, expressed by endothelial and other cells, and are involved in inflammatory responses, immunity, tumor immune escape and other processes (68,69), potentially due to involvement of the NF-κB pathway in TNF-α-induced upregulation of ICAM-1 and VCAM-1 in vascular endothelial cells (70,71). Kuessel et al (72) showed that the mRNA levels of both VCAM-1 and ICAM-1 are high in normal peritoneal samples of patients with EM and that serum soluble VCAM-1 (sVCAM-1) levels are not influenced by the entity of the lesion, the stage of menstrual cycle or the severity of the disease. The aforementioned study further indicated that the specificity and sensitivity of serum VCAM-1 for diagnosis of EM is 80 and 84%, respectively. It is noteworthy that the diagnostic specificity and sensitivity of sVCAM-1/sICAM-1 ratio, which further improved this predictive performance, were 86.7 and 90.3%, respectively. In addition, elevated levels of sICAM-1 may impair natural killer cell activity, which in turn accelerates the progression of EM (70).

VEGF, one of the most potent pro-angiogenic factors, is expressed at elevated levels in patients with EM and may be a potential biological molecule for the diagnosis of EM (10,50). A previous study demonstrated that degree of VEGF elevation is associated with the stage of EM, with additional marked elevation in advanced stages (24).

Serum microRNA (miRNA/miR) is a non-coding RNA consisting of 21–23 nucleotides widely found in eukaryotes, which bind exosomes or to specific protein complexes to protect against endogenous RNase degradation, playing an active role in the regulation of gene expression and cell cycle (73,74). Several studies (75,76) have shown that multiple miRNAs regulate angiogenesis and inflammatory responses in the ectopic endometrium by regulating expression of molecules such as VEGF, MMPs, IL and TGF-β1. Consequently, dysregulation of their expression serves an essential role in the pathogenesis of EM (75,77). Compared with conventional ultrasound, miRNAs have the advantages of improved stability and specificity, simpler detection methods and lower detection costs, they are becoming potential molecular indicators for non-invasive identification of EM (Table II).

Table II.

Studies reporting the sensitivity and specificity of dysregulated miRNAs in EM.

Table II.

Studies reporting the sensitivity and specificity of dysregulated miRNAs in EM.

miRNASensitivity, %Specificity, %Alteration(Refs.)
miRNA-125b-5p100.096.0(74)
miRNA-34a-5p79.049.0(95)
miRNA-12296.594.1(86)
miRNA-14171.970.8(94)
miRNA-185-5p81.290.0(78)
miRNA-199a100.0100.0(86)
miRNAs-199b-3p96.080.0(99)
miRNA-200a90.662.5(94)
miRNA-200c100.096.0(95)
miRNA-224-5p84.080.0(99)
miRNA-34290.091.2(79)
miRNA-451a85.484.6(84)
miRNA-361392.761.0(79)
let-7d83.3100.0(80)
miRNA-199a, miRNA-122, miRNA-145 and miRNA-542-3p93.296.0-(98)
miRNAs-199b-3p, miRNA-224-5p and let-7d-3p96.0100.0-(99)
miRNA-155, miRNA574-3p and miRNA139-3p83.051.0-(97)

[i] miRNA, microRNA.

An increasing number of studies have been conducted to detect expression of one or more specific miRNAs in plasma by reverse transcription-quantitative PCR to investigate their diagnostic value in EM and it has been hypothesized that the miR-125b, −451 and let-7 families may serve as potential circulating biomarkers for EM (7880). A previous study showed that serum miR-125b-5p has a sensitivity and specificity of 100 and 96%, respectively, for the diagnosis of EM (74). In patients with EM, upregulation of miR-125b-5p and downregulation of let-7b-5p expression may be associated with an increase in cytokines such as TNF-α and IL-1β and −6 (81,82). These cytokines are hypothesized to be important components involved in angiogenesis (31,39). Migration inhibitory factor (MIF) is a pleiotropic inflammatory cytokine with upstream regulatory roles in immunity and has been shown to be involved in the development of EM. miRNA-451, which is hypothesized to target MIF, is a mitogenic cytokine that creates a proliferative and angiogenic phenotype for growth of ectopic endothelium (76). Compared with the healthy population, miRNA-451 expression is decreased in patients with endometrial co-infertility, suggesting that miRNA-451 expression levels are negatively associated with co-infertility in patients with EM and may be used as a marker for disease prognosis (83,84). In addition, elevated miR-145-5p expression levels in EM are associated with upregulation of VEGF-A and decreased levels of epidermal growth factor 2, phosphatase and tensin homolog and C-X-C chemokine receptor type 4 (75).

MiR-199a is another possible suitable biomarker for EM. miR-199a downregulates the expression of VEGF-A in ESCs under hypoxic conditions and partially attenuates angiogenesis in hematopoietic stem cells under hypoxic conditions by inhibiting the HIF-1α/VEGF-A pathway (85). Maged et al (86) showed that serum miR-199a and −122 have high sensitivity and specificity and miR-199a has 100% sensitivity and specificity for the diagnosis of EM. Another study showed that VEGF-A is a direct and functional target of miR-199a-5p and elevated miR-199a-5p expression inhibits the proliferation and angiogenesis of MSCs, as confirmed in vitro experiments and EM female C57BL/6j mouse models (87). Notably, HIF-1 upregulates miR-20a; miR-20a not only increases the expression of the VEGF-A angiogenic gene, but also downregulates anti-angiogenic thrombospondin 1 to promote angiogenesis (76,8890). However, miR-20 as a biomarker for the diagnosis of EM remains to be investigated. In addition, dysregulation of miRNA-202-3p (91), miRNA-126 (92), miRNA-200 (75,9395) and other genes mediate VEGF expression to promote angiogenesis and invasion of ectopic endothelium by promoting cell proliferation and angiogenesis. The aforementioned studies suggest that epigenetic abnormalities and dysregulation of miRNAs may play a key role in the formation of EM by affecting different physiological processes, which may also be beneficial for further studies on mechanisms of EM.

With the progress of miRNA research, diagnostic value of miRNAs has been further confirmed in the clinical setting. Moustafa et al (79) first demonstrated in a prospective study that plasma miRNAs accurately distinguish EM from other gynecological diseases, and miR-125b-5p, −150-5p, −342-3p and 451a are notably higher in patients with EM, while miR-3613-5p and let-7b are expressed at lower levels, miRNA expression levels are not markedly associated with menstrual cycle or hormonal drug use. To improve the diagnostic value of miRNA, co-diagnostic methods have been applied (9699).

Results of several studies remain controversial and even contradictory due to the irreproducible nature of the studies (12,100). At present, no single or groups of miRNA biomarkers has sufficient specificity and sensitivity in the diagnosis of EM (101). Future studies need to adopt uniform and standardized methods to clarify the value of miRNA as a biological marker for early diagnosis and prognosis prediction of EM and for clinical translation and application.

The Internet of Medical Thing (IoMT) is the most emerging era of the Internet of Things (102,103). IoMT can generate large amounts of data without human intervention through connected smart medical devices, allowing healthcare professionals to facilitate disease detection by learning complex models and extracting meaningful information from large amounts of data (103). Based on this, researchers have trained on pulmonary embolism detection datasets and obtained satisfactory results in terms of sensitivity and specificity (102). To the best of our knowledge, however, its application in EM has not been reported yet. EM is a chronic inflammatory disease and using IoMT to establish a safe and effective system may contribute to the examination and treatment of the disease.

Numerous studies reveal (69,104,105) the importance and effectiveness of extracting various putative biomarkers from the biological fluids of affected patients. Nevertheless, due to the large heterogeneity between different studies, there is no uniform conclusion and further studies with larger sample sizes are needed to identify sensitive and specific indicators for transition from the laboratory to the clinic. As vascular mechanisms of ectopic endometrium are refined, newer and more accurate diagnostic markers for EM are expected to be discovered.

Anti-angiogenic treatment in EM

The current medical treatment for EM focuses on either hormonal modulation to induce a low estrogen state or surgical treatment to remove the ectopic lesions. However, even if the ectopic lesions are removed by surgery, certain patients will still have recurrence and even face the possibility of reoperation (106). Nevertheless, targeted therapeutic strategies based on anti-angiogenic therapy have been demonstrated in oncology and ophthalmology and have been widely used in clinical practice (8,43). As aforementioned, angiogenesis serves a key role in the development of EM and is essential for survival of ectopic endothelial implants. This shows that inhibition of ectopic endothelial angiogenesis is an important method to treat EM and if the mechanism can be regulated or inhibited or its pathway expression blocked, ectopic endothelial angiogenesis can be inhibited, improving the symptoms of patients and having value for clinical treatment. Currently, the primary anti-angiogenic drugs used in targeted therapy include anti-VEGF antibodies, VEGFR tyrosine kinase and COX-2 inhibitors and dopamine agonists (D2-ags; Table III).

Table III.

Research progress of anti-angiogenic drugs in EM.

Table III.

Research progress of anti-angiogenic drugs in EM.

Class of drugDrugExperimental designMechanism of actionOutcome(Refs.)
Anti-VEGF antibodyBevacizumabRat modelAnti-VEGFSignificant decrease in lesion area(52)
RanibizumabRat modelAnti-VEGFSignificant decrease in lesion area(51)
Tyrosine kinaseSorafenibMouse modelInhibition of VEGFDecreased lesion implantation(109)
inhibitorSunitinibMouse modelPromotes maturation of peritoneal fluid MDSCs and suppresses immunosuppressive functionSignificant decrease in the size and weight of EM lesions(108)
PazopanibRat modelInhibits VEGF and CD117 expressionDecreased lesion implantation(109)
Dopamine receptor agonistCabergolineClinical trialInactivates VEGFR-2 to exert anti-angiogenic effectsSignificant decrease in size of endometriomas(112)
QuinagolideIn vitroInhibition of AKT signaling pathwaySignificant inhibition of ectopic E-MSC(110)
COX-2 InhibitorCelecoxibBALB/c mouse modelInhibition of COX-2 signaling pathwayDecreased lesion implant volume and vascular density(114)
ParecoxibC57BL/6 mouse modelInhibition of COX-2 signaling pathwayDecreased volume of diseased implant volume(117)
PPARγ activatorRosiglitazoneBALB/c mouse and rat modelActivation of PPARγ signaling pathwayInhibited lesion growth, cell proliferation and vascularization; increased apoptosis(114,115)
Angiotensin II receptor blockerTelmisartanC57BL/6 mouse modelCombined blockade of AT1R and activation of PPARγSignificant inhibition of the growth of lesions and decreased density of blood vessels(116,117)
NF-κB inhibitorBAY11-7085In vitroInhibition of NF-κB signaling pathway and expression of anti-apoptotic proteinsInhibition of the viability of ectopic endothelial cells(118)
Pyrrolidine dithiocarbamateIn vitroInhibition of NF-κB signaling pathwayInhibition of COX-2 expression, decreased PGE2 production, inhibition of EM cell proliferation, angiogenesis and inflammatory response(119)
OtherGinsenoside Rg3In vitroRegulation of apoptosis and angiogenesis via nuclear factor/NF-κB signaling pathwaySignificantly decreased levels of VEGF(120)
CurcuminIn vitroInhibition of NF-κB pathway activationDecreased secretion of chemokines and cytokines(121,122)
NobiletinC57BL/6 mouse modelInhibition of NF-κB pathway activationSignificantly decreased lesion size and pain in EM mice(123)
DienogestClinical trialInhibition of NF-κB pathwayEffective decrease in endometrial tumor size and EM-associated pain(124)
ImmunomodulatorEtanerceptWistar Albino rat modelDecreased expression and activity of serum VEGF, IL-6 and TNF-αEffective decrease in the volume of ectopic lesions(125)
RapamycinMouse modelInhibition of VEGF expressionSignificant decrease in the volume of ectopic lesions(126)
Interleukin (IFN)In vitroInterference with the cell cycleInhibition of ESC cell proliferation and migration(128)
AntioxidantMelatoninClinical trialAntioxidationImproved patient sleep quality and decreased pain associated with EM(129,131)
Vitamins E and CClinical trialAntioxidationDecreased inflammatory markers in peritoneal fluid and chronic pelvic pain(130)
ResveratrolClinical trialAntioxidationDecreased expression levels of VEGF and TNF-α(25)
Iron death inducerErastinC57BL/6 mouse modelInduces ectopic endometrial stromal cell deathSignificant decrease in ectopic lesions(133)

[i] VEGF, vascular endothelial growth factor; MDSC, myeloid-derived suppressor cells; EM, endometriosis; VEGFR, VEGF receptor; COX-2, cyclooxygenase 2; E-MSC, endometrial mesenchymal stromal cell; PPARγ, peroxisome proliferator-activated receptor γ; AT1R, angiotensin II type 1 receptor; TNF-α, tumor necrosis factor-α.

Anti-VEGF and its receptor signaling pathway

Since VEGF is the most potent angiogenic factor and has an important role in EM (24), VEGF and its signaling pathway are considered to be the most effective targets for anti-angiogenic therapy, and in treatment of kidney, lung, rectal and cervical cancer (13,107). Bevacizumab and ranibizumab are monoclonal antibodies that bind and selectively neutralize VEGF activity by inhibiting binding of VEGF to VEGFR, primarily VEGFR2 or kinase insertion domain receptor, thereby reducing angiogenesis; additionally, both antibodies are effective against EM (51,52). Blocking VEGF signaling and inhibiting tyrosine kinase activity are potential targets for anti-angiogenic therapy (107109). Research on VEGFR tyrosine kinase inhibitors has been ongoing and tyrosine kinase inhibitors such as sorafenib and sunitinib have been shown to be effective against EM (108,109). In comparing the effects of pazopanib, sorafenib and sunitinib on the VEGF/VEGFR protein kinase pathway and their role in EM, Yildiz et al (109) noted that pazopanib is more effective than control and other treatments, reducing EM lesions by ≥45%, but sorafenib exhibited improved modulation of VEGF. Anlotinib is a novel oral multitarget tyrosine kinase inhibitor developed independently in China that can effectively inhibit VEGFR, platelet-derived growth factor receptor, FGFR and c-Kit and exert antitumor angiogenesis effects and has been applied in gynecological tumors such as ovarian cancer; to the best of our knowledge however, no studies have reported on its application in EM (107).

D2-ags

E-MSCs express D2, with notably higher levels of ectopic endometrial expression relative to normal endometrial tissue (110). D2-ags decrease tumor size by targeting abnormal angiogenesis in pathological tissue, showing that D2-ags could be used to treat EM (111,112). Cabergoline, a dopamine agonist, exerts anti-angiogenic effects by inducing endocytosis of VEGFR-2 in endothelial cells, leading to decreased and inactivated VEGF-VEGFR-2 binding (112). D2-ags are as powerful as standard anti-angiogenic compounds in interfering with angiogenesis and lesion size (111,113). In addition, in a prospective clinical trial, D2-ags were shown to be superior to luteinizing hormone-releasing hormone agonists in decreasing size of endometrial tumors, with fewer drug side effects, easier administration and lower price (112). Quinagolide is a non-ergot-derived D2-ag that has been reported to reduce or eliminate peritoneal EM lesions in patients with EM (110). Another study found notable inhibition of E-MSCs by quinagolide, which decreases invasion and endothelial differentiation via the AKT signaling pathway, further supporting the theoretical basis for the use of this drug in treatment of EM (110). The role of quinagolide in EM is currently undergoing phase 2 clinical trials (trial nos. NCT03749109 and NCT03692403), which are expected to provide a novel treatment strategy for the future treatment of EM (110).

COX-2 inhibitor

Celecoxib, a potent COX-2 inhibitor, has been shown to inhibit EM lesions by markedly decreasing the size of ectopic lesions and vascular density in a study exploring the effects of celecoxib and rosiglitazone on implantation and growth of EM-like lesions in mice with EM (114). Additionally, treatment with peroxisome proliferator-activated receptor γ (PPARγ) activator rosiglitazone has also been shown to inhibit the growth of EM implants, and the combination of celecoxib and rosiglitazone is more potent than single application of celecoxib in the inhibition of ectopic EM lesions (114,115). Telmisartan, a partial agonist of PPARγ, also blocks angiotensin II type 1 receptor (AT1R); combined blockade of AT1R and activation of PPARγ markedly inhibits angiogenesis and growth in EM-like lesions in mice (116). In addition, Nenicu et al (117) found that combination of telmisartan and parecoxib in the treatment of endometrioid lesions increases the rate of lesion regression and notably improves therapeutic effects compared with treatment with telmisartan alone. This suggests that the combination of ≥2 drugs may achieve more desirable therapeutic effects than a single drug against angiogenesis.

NF-κB inhibitor

NF-κB, a key factor in signaling, can regulate growth of ectopic endothelium through multiple pathways (53). Therefore, NF-κB is considered an important target for anti-angiogenic therapy. BAY11-7085, an NF-κB inhibitor, inhibits the viability of EM cells and induces apoptosis in endometriotic cyst stromal cells by inhibiting anti-apoptotic proteins in an in vitro experimental study; its effect on normal endometrial cells is not notable, but, to the best of our knowledge, clinical studies for its treatment have not been reported (118). Pyrrolidine dithiocarbamate is another potent NF-κB inhibitor that inhibits NF-κB signaling in EM cells, suppresses COX-2 expression, decreases PGE2 production and suppresses EM cell proliferation, angiogenesis and inflammatory responses (119). In addition, natural extracts such as ginsenoside Rg3 and curcumin decrease EM by regulating apoptosis and inhibiting VEGF expression and angiogenesis via the NF-κB signaling pathway (120122). The low risk of adverse effects of these natural products indicates their potential clinical value in the treatment of EM. It has been demonstrated that the inhibitory effect of nobiletin on EM is achieved by inhibiting activation of the NF-κB pathway, which markedly decreases the regulation of the expression of angiogenic factors, including VEGF and E-cadherin, in ectopic endometrium (123). Dienogest is an artificially developed, fourth-generation progestin with strong anti-proliferative effects on EM implants, as well as anti-angiogenic and anti-inflammatory properties (124). Its mechanism of action may be associated with inhibition of the NF-κB pathway, which is effective in decreasing the size of endometriomas and EM-related pain and has a good safety and tolerability profile, with improved clinical efficacy in young patients with endometriomas who have not yet had children (124). Moreover, 2-cyano-3,12-dioxooleana1,9-dien-28-oic acid effectively inhibit EM by increasing ectopic cell apoptosis and decreasing angiogenesis by modulating Nrf2 and NF-κB pathways and has anti-fibrotic, anti-inflammatory and antioxidant effects in EM (55).

Immunomodulators

Numerous researchers have noted involvement of immune inflammatory factors in ectopic endothelial vascularization (18,27,104). Therefore, modulation of the immune system may also serve a role in EM treatment. Etanercept, a fusion protein consisting of the human recombinant soluble TNF receptor 2 bound to human Fc antibody subunits, is effective in decreasing the volume of ectopic lesions in rats by decreasing serum VEGF and IL-6 levels, and TNF-α activity (125). An experiment investigating the effect of rapamycin (RAPA) on EM lesions in severe combined immunodeficient mice found that RAPA inhibits the growth of EM lesions, potentially by suppressing expression of VEGF in the lesions and thus angiogenesis (126). In addition, similar pharmacological efficacy of interferons (IFNs) and pentoxifylline for treatment of EM has been observed, IFN-β 1a has a notably stronger in vitro inhibitory effect on ESC proliferation and migration than IFN-α 2b, while the efficacy and safety of pentoxifylline in treatment of EM has been reported, but there is insufficient evidence to support its effectiveness in the treatment of infertility and pain in EM (127,128). The data from the aforementioned studies provide a theoretical basis for future clinical trials.

Antioxidant therapy

With in-depth research on development and targets of EM, the important role of oxidative stress in EM has become increasingly recognized (2,53). In parallel, studies have demonstrated that antioxidants inhibit the development of ectopic endothelium and achieve efficacy in the treatment of EM (25,129131). The effects of antioxidant melatonin and vitamins E and C on EM are all demonstrated in clinical randomized placebo-controlled trials; they notably improve the reduction of chronic pelvic pain and decrease expression of inflammatory markers in peritoneal fluid in patients with EM (129132). Resveratrol is a naturally occurring synthetic polyphenolic compound with antitumor, anti-inflammatory, antioxidant and anti-angiogenic properties (25). In a randomized controlled clinical trial study, resveratrol was found to decrease angiogenesis and inflammation in endometrial tissue of patients with EM by downregulating expression of VEGF and TNF-α (25). Although melatonin, resveratrol and combined vitamin C and E supplements have shown good results in treatment of EM, their use requires further investigation. Recently, Li et al (133) demonstrated that the iron death inducer erastin could induce ectopic endometrial stromal cell death in C57BL/6 female mouse model of EM, and the ectopic lesions are reduced after treatment with erastin, suggesting that erastin may be a potential drug for treatment of EM, which provides a new idea for the targeted treatment of EM.

In conclusion, anti-angiogenic therapy has become a focal point of medical research and has an improved effect in the treatment of EM, which may provide a potential novel treatment for EM (Fig. 3). However, most of the drug application research results are at the stage of in vitro and animal experiments; although some drugs are at the stage of clinical experiments, there is still lack of drug safety and efficacy assessment. Therefore, more clinical trials are needed to assess the value of these drugs clinically.

Conclusions

Although EM is a benign lesion, the mechanism of its angiogenesis has similarities with pathologic angiogenesis mediating tumor and metastasis, which is complicated by multiple factors and mechanisms. The improvement of understanding of the EM angiogenic network regulation system has brought new hope for its diagnosis and treatment. However, the majority of studies are still at the laboratory stage, and to the best of our knowledge, there are no satisfactory clinical trials due to differences in research data and non-reproducibility, making it challenging to apply non-invasive biomarkers and anti-angiogenic drugs in the clinic.

In addition, clinical evidence for the efficacy of anti-angiogenic treatment strategies in EM is lacking. Anti-angiogenic therapy may adversely affect normal physiological angiogenesis such as ovulation and wound healing (134,135). Research is required to elucidate the interactions between factors in the angiogenic microenvironment to discover more effective targets for drug therapy. In addition, clinical trials are needed to evaluate the therapeutic value of these serum markers and anti-vascular drugs and to strengthen multi-targeted combined diagnosis and treatment to inhibit angiogenesis.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation of China (grant no. 81502255), Shandong Provincial Traditional Chinese Medicine Science and Technology Development Project (grant no. M-2022242) and the Key R&D Program of Jining (grant nos. 2020YXNS026 and 2022YXNS007).

Availability of data and materials

Not applicable.

Authors' contributions

CB conceived the study, performed the literature review, wrote the manuscript and constructed figures. YW conceived and supervised the study and wrote and edited the manuscript. Both authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

AT1R

angiotensin II type 1 receptor

COX-2

cyclooxygenase 2

DC

dendritic cell

D2-ag

dopamine receptor agonist

E-MSC

endometrial mesenchymal stromal cell

EM

endometriosis

ESC

endometrial stromal cell

ERβ

estrogen receptor β

FGA

fibrinogen α chain

FGF

fibroblast growth factor

Th17

helper T 17

HIF-1α

hypoxia-inducible factor-1α

ICAM-1

intercellular adhesion molecule-1

LPS

lipopolysaccharide

MMP

matrix metalloproteinase

NF-κB

nuclear factor-κB

PPARγ

peroxisome proliferator-activated receptor γ

PDGF

platelet-derived growth factor

PGE2

prostaglandin E2

RAPA

rapamycin

ROS

reactive oxygen species

TNF-α

tumor necrosis factor-α

TFG-β

transforming growth factor-β

sVCAM-1

soluble vascular cell adhesion molecule-1

VEGF

vascular endothelial growth factor

References

1 

Smolarz B, Szyłło K and Romanowicz H: Endometriosis: Epidemiology, classification, pathogenesis, treatment and genetics (Review of Literature). Int J Mol Sci. 22:105542021. View Article : Google Scholar : PubMed/NCBI

2 

Samimi M, Pourhanifeh MH, Mehdizadehkashi A, Eftekhar T and Asemi Z: The role of inflammation, oxidative stress, angiogenesis, and apoptosis in the pathophysiology of endometriosis: Basic science and new insights based on gene expression. J Cell Physiol. 234:19384–19392. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Zondervan KT, Becker CM and Missmer SA: Endometriosis. N Engl J Med. 382:1244–1256. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Brasil DL, Montagna E, Trevisan CM, La Rosa VL, Laganà AS, Barbosa CP, Bianco B and Zaia V: Psychological stress levels in women with endometriosis: Systematic review and meta-analysis of observational studies. Minerva Med. 111:90–102. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Sullivan-Myers C, Sherman KA, Beath AP, Duckworth TJ and Cooper MJW: Delineating sociodemographic, medical and quality of life factors associated with psychological distress in individuals with endometriosis. Hum Reprod. 36:2170–2180. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Wang Y, Nicholes K and Shih IM: The origin and pathogenesis of endometriosis. Annu Rev Pathol. 15:71–95. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Laganà AS, Vitale SG, Salmeri FM, Triolo O, Ban Frangež H, Vrtačnik-Bokal E, Stojanovska L, Apostolopoulos V, Granese R and Sofo V: Unus pro omnibus, omnes pro uno: A novel, evidence-based, unifying theory for the pathogenesis of endometriosis. Med Hypotheses. 103:10–20. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Dudley AC and Griffioen AW: Pathological angiogenesis: Mechanisms and therapeutic strategies. Angiogenesis. 26:313–347. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Ben Dhaou C, Mandi K, Frye M, Acheampong A, Radi A, De Becker B, Antoine M, Baeyens N, Wittamer V and Parmentier M: Chemerin regulates normal angiogenesis and hypoxia-driven neovascularization. Angiogenesis. 25:159–179. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Li H, Cai E, Cheng H, Ye X, Ma R, Zhu H and Chang X: FGA Controls VEGFA secretion to promote angiogenesis by activating the VEGFR2-FAK signalling pathway. Front Endocrinol (Lausanne). 13:7918602022. View Article : Google Scholar : PubMed/NCBI

11 

Tan Y, Flynn WF, Sivajothi S, Luo D, Bozal SB, Davé M, Luciano AA, Robson P, Luciano DE and Courtois ET: Single-cell analysis of endometriosis reveals a coordinated transcriptional programme driving immunotolerance and angiogenesis across eutopic and ectopic tissues. Nat Cell Biol. 24:1306–1318. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Hon JX, Wahab NA, Karim AKA, Mokhtar NM and Mokhtar MH: MicroRNAs in Endometriosis: Insights into inflammation and progesterone resistance. Int J Mol Sci. 24:150012023. View Article : Google Scholar : PubMed/NCBI

13 

Potente M, Gerhardt H and Carmeliet P: Basic and therapeutic aspects of angiogenesis. Cell. 146:873–887. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Cha J, Sun X and Dey SK: Mechanisms of implantation: Strategies for successful pregnancy. Nat Med. 18:1754–1767. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Tarokh M, Ghaffari Novin M, Poordast T, Tavana Z, Nazarian H, Norouzian M and Gharesi-Fard B: Serum and peritoneal fluid cytokine profiles in infertile women with endometriosis. Iran J Immunol. 16:151–162. 2019.PubMed/NCBI

16 

Laschke MW and Menger MD: Basic mechanisms of vascularization in endometriosis and their clinical implications. Hum Reprod Update. 24:207–224. 2018. View Article : Google Scholar : PubMed/NCBI

17 

Symons LK, Miller JE, Kay VR, Marks RM, Liblik K, Koti M and Tayade C: The immunopathophysiology of endometriosis. Trends Mol Med. 24:748–762. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Chen S, Liu Y, Zhong Z, Wei C, Liu Y and Zhu X: Peritoneal immune microenvironment of endometriosis: Role and therapeutic perspectives. Front Immunol. 14:11346632023. View Article : Google Scholar : PubMed/NCBI

19 

Guo F, He Y, Fan Y, Du Z, Sun H, Feng Z, Zhang G and Xiong T: G-CSF and IL-6 may be involved in formation of endometriosis lesions by increasing the expression of angiogenic factors in neutrophils. Mol Hum Reprod. 27:gaab0642021. View Article : Google Scholar : PubMed/NCBI

20 

Sikora J, Mielczarek-Palacz A and Kondera-Anasz Z: Association of the precursor of interleukin-1β and peritoneal inflammation-role in pathogenesis of endometriosis. J Clin Lab Anal. 30:831–837. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Li C, Zhao HL, Li YJ, Zhang YY, Liu HY, Feng FZ and Yan H: The expression and significance of leukemia inhibitory factor, interleukin-6 and vascular endothelial growth factor in Chinese patients with endometriosis. Arch Gynecol Obstet. 304:163–170. 2021. View Article : Google Scholar : PubMed/NCBI

22 

Barcz E, Rózewska ES, Kaminski P, Demkow U, Bobrowska K and Marianowski L: Angiogenic activity and IL-8 concentrations in peritoneal fluid and sera in endometriosis. Int J Gynaecol Obstet. 79:229–235. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Sikora J, Smycz-Kubańska M, Mielczarek-Palacz A, Bednarek I and Kondera-Anasz Z: The involvement of multifunctional TGF-β and related cytokines in pathogenesis of endometriosis. Immunol Lett. 201:31–37. 2018. View Article : Google Scholar : PubMed/NCBI

24 

Zhang F, Liu XL, Wang W, Dong HL, Xia YF, Ruan LP and Liu LP: Expression of MMIF, HIF-1α and VEGF in serum and endometrial tissues of patients with endometriosis. Curr Med Sci. 38:499–504. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Khodarahmian M, Amidi F, Moini A, Kashani L, Salahi E, Danaii-Mehrabad S, Nashtaei MS, Mojtahedi MF, Esfandyari S and Sobhani A: A randomized exploratory trial to assess the effects of resveratrol on VEGF and TNF-α 2 expression in endometriosis women. J Reprod Immunol. 143:1032482021. View Article : Google Scholar : PubMed/NCBI

26 

Nanda A K T, Banerjee P, Dutta M, Wangdi T, Sharma P, Chaudhury K and Jana SK: Cytokines, angiogenesis, and extracellular matrix degradation are augmented by oxidative stress in endometriosis. Ann Lab Med. 40:390–397. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Singh AK, Dutta M, Chattopadhyay R, Chakravarty B and Chaudhury K: Intrafollicular interleukin-8, interleukin-12, and adrenomedullin are the promising prognostic markers of oocyte and embryo quality in women with endometriosis. J Assist Reprod Genet. 33:1363–1372. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Monsanto SP, Edwards AK, Zhou J, Nagarkatti P, Nagarkatti M, Young SL, Lessey BA and Tayade C: Surgical removal of endometriotic lesions alters local and systemic proinflammatory cytokines in endometriosis patients. Fertil Steril. 105:968–977. e52016. View Article : Google Scholar : PubMed/NCBI

29 

Vazgiourakis VM, Zervou MI, Papageorgiou L, Chaniotis D, Spandidos DA, Vlachakis D, Eliopoulos E and Goulielmos GN: Association of endometriosis with cardiovascular disease: Genetic aspects (Review). Int J Mol Med. 51:292023. View Article : Google Scholar : PubMed/NCBI

30 

Ahn SH, Edwards AK, Singh SS, Young SL, Lessey BA and Tayade C: IL-17A contributes to the pathogenesis of endometriosis by triggering proinflammatory cytokines and angiogenic growth factors. J Immunol. 195:2591–2600. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Lin YJ, Lai MD, Lei HY and Wing LY: Neutrophils and macrophages promote angiogenesis in the early stage of endometriosis in a mouse model. Endocrinology. 147:1278–1286. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Yan WK, Liu YN, Song SS, Kang JW, Zhang Y, Lu L, Wei SW, Xu QX, Zhang WQ, Liu XZ, et al: Zearalenone affects the growth of endometriosis via estrogen signaling and inflammatory pathways. Ecotoxicol Environ Saf. 241:1138262022. View Article : Google Scholar : PubMed/NCBI

33 

Gou Y, Li X, Li P, Zhang H, Xu T, Wang H, Wang B, Ma X, Jiang X and Zhang Z: Estrogen receptor β upregulates CCL2 via NF-κB signaling in endometriotic stromal cells and recruits macrophages to promote the pathogenesis of endometriosis. Hum Reprod. 34:646–658. 2019. View Article : Google Scholar : PubMed/NCBI

34 

Laganà AS, Salmeri FM, Ban Frangež H, Ghezzi F, Vrtačnik-Bokal E and Granese R: Evaluation of M1 and M2 macrophages in ovarian endometriomas from women affected by endometriosis at different stages of the disease. Gynecol Endocrinol. 36:441–444. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Li Q, Yuan M, Jiao X, Huang Y, Li J, Li D, Ji M and Wang G: M1 macrophage-derived nanovesicles repolarize M2 macrophages for inhibiting the development of endometriosis. Front Immunol. 12:7077842021. View Article : Google Scholar : PubMed/NCBI

36 

Vallvé-Juanico J, Houshdaran S and Giudice LC: The endometrial immune environment of women with endometriosis. Hum Reprod Update. 25:564–591. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Gao X, Gao H, Shao W, Wang J, Li M and Liu S: The extracellular vesicle-macrophage regulatory axis: A novel pathogenesis for endometriosis. Biomolecules. 13:13762023. View Article : Google Scholar : PubMed/NCBI

38 

Suen JL, Chang Y, Shiu YS, Hsu CY, Sharma P, Chiu CC, Chen YJ, Hour TC and Tsai EM: IL-10 from plasmacytoid dendritic cells promotes angiogenesis in the early stage of endometriosis. J Pathol. 249:485–497. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Fainaru O, Adini A, Benny O, Adini I, Short S, Bazinet L, Nakai K, Pravda E, Hornstein MD, D'Amato RJ and Folkman J: Dendritic cells support angiogenesis and promote lesion growth in a murine model of endometriosis. FASEB J. 22:522–529. 2008. View Article : Google Scholar : PubMed/NCBI

40 

Talaat RM, Mohamed SF, Bassyouni IH and Raouf AA: Th1/Th2/Th17/Treg cytokine imbalance in systemic lupus erythematosus (SLE) patients: Correlation with disease activity. Cytokine. 72:146–153. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Kang YJ, Cho HJ, Lee Y, Park A, Kim MJ, Jeung IC, Jung YW, Jung H, Choi I, Lee HG and Yoon SR: IL-17A and Th17 cells contribute to endometrial cell survival by inhibiting apoptosis and NK cell mediated cytotoxicity of endometrial cells via ERK1/2 pathway. Immune Netw. 23:e142023. View Article : Google Scholar : PubMed/NCBI

42 

Miller JE, Ahn SH, Marks RM, Monsanto SP, Fazleabas AT, Koti M and Tayade C: IL-17A modulates peritoneal macrophage recruitment and M2 polarization in endometriosis. Front Immunol. 11:1082020. View Article : Google Scholar : PubMed/NCBI

43 

Apte RS, Chen DS and Ferrara N: VEGF in signaling and disease: Beyond discovery and development. Cell. 176:1248–1264. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Wu MH, Hsiao KY and Tsai SJ: Hypoxia: The force of endometriosis. J Obstet Gynaecol Res. 45:532–541. 2019. View Article : Google Scholar : PubMed/NCBI

45 

Liu H, Zhang Z, Xiong W, Zhang L, Xiong Y, Li N, He H, Du Y and Liu Y: Hypoxia-inducible factor-1α promotes endometrial stromal cells migration and invasion by upregulating autophagy in endometriosis. Reproduction. 153:809–820. 2017. View Article : Google Scholar : PubMed/NCBI

46 

Fu JL, Hsiao KY, Lee HC, Li WN, Chang N, Wu MH and Tsai SJ: Suppression of COUP-TFII upregulates angiogenin and promotes angiogenesis in endometriosis. Hum Reprod. 33:1517–1527. 2018. View Article : Google Scholar : PubMed/NCBI

47 

Zhou Y, Jin Y, Wang Y and Wu R: Hypoxia activates the unfolded protein response signaling network: An adaptive mechanism for endometriosis. Front Endocrinol (Lausanne). 13:9455782022. View Article : Google Scholar : PubMed/NCBI

48 

Cheng J, Yang HL, Gu CJ, Liu YK, Shao J, Zhu R, He YY, Zhu XY and Li MQ: Melatonin restricts the viability and angiogenesis of vascular endothelial cells by suppressing HIF-1α/ROS/VEGF. Int J Mol Med. 43:945–955. 2019.PubMed/NCBI

49 

Ferrara N and Adamis AP: Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 15:385–403. 2016. View Article : Google Scholar : PubMed/NCBI

50 

Li WN, Hsiao KY, Wang CA, Chang N, Hsu PL, Sun CH, Wu SR, Wu MH and Tsai SJ: Extracellular vesicle-associated VEGF-C promotes lymphangiogenesis and immune cells infiltration in endometriosis. Proc Natl Acad Sci USA. 117:25859–25868. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Ureyen Ozdemir E, Adali E, Islimye Taskin M, Yavasoglu A, Aktug H, Oltulu F and Inceboz U: Effects of ranibizumab and zoledronic acid on endometriosis in a rat model. Arch Gynecol Obstet. 305:267–274. 2022. View Article : Google Scholar : PubMed/NCBI

52 

Zani ACT, Valerio FP, Meola J, da Silva AR, Nogueira AA, Candido-Dos-Reis FJ, Poli-Neto OB and Rosa-E-Silva JC: Impact of bevacizumab on experimentally induced endometriotic lesions: Angiogenesis, invasion, apoptosis, and cell proliferation. Reprod Sci. 27:1943–1950. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Liu Y, Wang J and Zhang X: An update on the multifaceted role of NF-kappaB in endometriosis. Int J Biol Sci. 18:4400–4413. 2022. View Article : Google Scholar : PubMed/NCBI

54 

Shamloo N, Taghavi N, Yazdani F, Azimian P and Ahmadi S: Evaluation of VEGF expression correlates with COX-2 expression in pleomorphic adenoma, mucoepidermoid carcinoma and adenoid cystic carcinoma. Dent Res J (Isfahan). 17:100–106. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Siracusa R, D'Amico R, Cordaro M, Peritore AF, Genovese T, Gugliandolo E, Crupi R, Impellizzeri D, Cuzzocrea S, Fusco R and Di Paola R: The Methyl Ester of 2-Cyano-3,12-Dioxooleana-1,9-Dien-28-Oic acid reduces endometrial lesions development by modulating the NFkB and Nrf2 pathways. Int J Mol Sci. 22:39912021. View Article : Google Scholar : PubMed/NCBI

56 

Santulli P, Marcellin L, Tosti C, Chouzenoux S, Cerles O, Borghese B, Batteux F and Chapron C: MAP kinases and the inflammatory signaling cascade as targets for the treatment of endometriosis? Expert Opin Ther Targets. 19:1465–1483. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Lai ZZ, Yang HL, Ha SY, Chang KK, Mei J, Zhou WJ, Qiu XM, Wang XQ, Zhu R, Li DJ and Li MQ: Cyclooxygenase-2 in Endometriosis. Int J Biol Sci. 15:2783–2797. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Hashemi Goradel N, Najafi M, Salehi E, Farhood B and Mortezaee K: Cyclooxygenase-2 in cancer: A review. J Cell Physiol. 234:5683–5699. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Ke J, Ye J, Li M and Zhu Z: The role of matrix metalloproteinases in endometriosis: A potential target. Biomolecules. 11:17392021. View Article : Google Scholar : PubMed/NCBI

60 

Jana S, Chatterjee K, Ray AK, DasMahapatra P and Swarnakar S: Regulation of matrix metalloproteinase-2 activity by COX-2-PGE2-pAKT axis promotes angiogenesis in endometriosis. PLoS One. 11:e01635402016. View Article : Google Scholar : PubMed/NCBI

61 

Horne AW, Daniels J, Hummelshoj L, Cox E and Cooper KG: Surgical removal of superficial peritoneal endometriosis for managing women with chronic pelvic pain: time for a rethink? BJOG. 126:1414–1416. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Kiesel L and Sourouni M: Diagnosis of endometriosis in the 21st century. Climacteric. 22:296–302. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Kotowska M, Urbaniak J, Falęcki WJ, Łazarewicz P, Masiak M and Szymusik I: Awareness of endometriosis symptoms-A cross sectional survey among polish women. Int J Environ Res Public Health. 18:99192021. View Article : Google Scholar : PubMed/NCBI

64 

Ghai V, Jan H, Shakir F, Haines P and Kent A: Diagnostic delay for superficial and deep endometriosis in the United Kingdom. J Obstet Gynaecol. 40:83–89. 2020. View Article : Google Scholar : PubMed/NCBI

65 

Rokhgireh S, Mehdizadeh Kashi A, Chaichian S, Delbandi AA, Allahqoli L, Ahmadi-Pishkuhi M, Khodaverdi S and Alkatout I: The diagnostic accuracy of combined Enolase/Cr, CA125, and CA19-9 in the detection of endometriosis. Biomed Res Int. 2020:52082792020. View Article : Google Scholar : PubMed/NCBI

66 

Harada T, Kubota T and Aso T: Usefulness of CA19-9 versus CA125 for the diagnosis of endometriosis. Fertil Steril. 78:733–739. 2002. View Article : Google Scholar : PubMed/NCBI

67 

Zhang X, Nie D, Zhang L and Liu X: Study on diagnostic values and pathological conditions of serum HGF and CA199 in endometriosis. Am J Transl Res. 13:2849–2857. 2021.PubMed/NCBI

68 

Anastasiu CV, Moga MA, Elena Neculau A, Bălan A, Scârneciu I, Dragomir RM, Dull AM and Chicea LM: Biomarkers for the noninvasive diagnosis of endometriosis: State of the art and future perspectives. Int J Mol Sci. 21:17502020. View Article : Google Scholar : PubMed/NCBI

69 

Moein Mahini S, Younesi M, Mortazavi G, Samare-Najaf M, Karim Azadbakht M and Jamali N: Non-invasive diagnosis of endometriosis: Immunologic and genetic markers. Clin Chim Acta. 538:70–86. 2023. View Article : Google Scholar : PubMed/NCBI

70 

Kim KH, Park JK, Choi YW, Kim YH, Lee EN, Lee JR, Kim HS, Baek SY, Kim BS, Lee KS and Yoon S: Hexane extract of aged black garlic reduces cell proliferation and attenuates the expression of ICAM-1 and VCAM-1 in TNF-α-activated human endometrial stromal cells. Int J Mol Med. 32:67–78. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Fukaya T, Sugawara J, Yoshida H, Murakami T and Yajima A: Intercellular adhesion molecule-1 and hepatocyte growth factor in human endometriosis: Original investigation and a review of literature. Gynecol Obstet Invest. 47 (Suppl 1):S11–S16; discussion 16–17. 1999. View Article : Google Scholar

72 

Kuessel L, Wenzl R, Proestling K, Balendran S, Pateisky P, Yotova I, Yerlikaya G, Streubel B and Husslein H: Soluble VCAM-1/soluble ICAM-1 ratio is a promising biomarker for diagnosing endometriosis. Hum Reprod. 32:770–779. 2017.PubMed/NCBI

73 

O'Brien J, Hayder H, Zayed Y and Peng C: Overview of MicroRNA biogenesis, mechanisms of actions, and circulation. Front Endocrinol (Lausanne). 9:4022018. View Article : Google Scholar : PubMed/NCBI

74 

Cosar E, Mamillapalli R, Ersoy GS, Cho S, Seifer B and Taylor HS: Serum microRNAs as diagnostic markers of endometriosis: A comprehensive array-based analysis. Fertil Steril. 106:402–409. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Yang RQ, Teng H, Xu XH, Liu SY, Wang YH, Guo FJ and Liu XJ: Microarray analysis of microRNA deregulation and angiogenesis-related proteins in endometriosis. Genet Mol Res. 15:2016.

76 

Zubrzycka A, Migdalska-Sęk M, Jędrzejczyk S and Brzeziańska-Lasota E: Circulating miRNAs related to epithelial-mesenchymal transitions (EMT) as the new molecular markers in endometriosis. Curr Issues Mol Biol. 43:900–916. 2021. View Article : Google Scholar : PubMed/NCBI

77 

Quintero-Fabián S, Arreola R, Becerril-Villanueva E, Torres-Romero JC, Arana-Argáez V, Lara-Riegos J, Ramírez-Camacho MA and Alvarez-Sánchez ME: Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol. 9:13702019. View Article : Google Scholar : PubMed/NCBI

78 

Hossein Razi M, Eftekhar M, Ghasemi N, Hasan Sheikhha M and Dehghani Firoozabadi A: Expression levels of circulatory mir-185-5p, vascular endothelial growth factor, and platelet-derived growth factor target genes in endometriosis. Int J Reprod Biomed. 18:347–358. 2020.PubMed/NCBI

79 

Moustafa S, Burn M, Mamillapalli R, Nematian S, Flores V and Taylor HS: Accurate diagnosis of endometriosis using serum microRNAs. Am J Obstet Gynecol. 223:557.e1–557.e11. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Cho S, Mutlu L, Grechukhina O and Taylor HS: Circulating microRNAs as potential biomarkers for endometriosis. Fertil Steril. 103:1252–1260.e1. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Sheikhvatan M, Chaichian S and Moazzami B: A systematic review and bioinformatics study on genes and micro-RNAs involving the transformation of endometriosis into ovarian cancer. Microrna. 9:101–111. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Nematian SE, Mamillapalli R, Kadakia TS, Majidi Zolbin M, Moustafa S and Taylor HS: Systemic inflammation induced by microRNAs: Endometriosis-Derived alterations in circulating microRNA 125b-5p and Let-7b-5p regulate macrophage cytokine production. J Clin Endocrinol Metab. 103:64–74. 2018. View Article : Google Scholar : PubMed/NCBI

83 

Wang L, Zhang J, Sun H, Ji X and Zhang S: Effect of miR-451 on IVF/ICSI-ET outcome in patient with endometriosis and infertility. Am J Transl Res. 13:13051–13058. 2021.PubMed/NCBI

84 

Nothnick WB, Falcone T, Joshi N, Fazleabas AT and Graham A: Serum miR-451a levels are significantly elevated in women with endometriosis and recapitulated in baboons (Papio anubis) with experimentally-induced disease. Reprod Sci. 24:1195–1202. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Dai L, Lou W, Zhu J, Zhou X and Di W: MiR-199a inhibits the angiogenic potential of endometrial stromal cells under hypoxia by targeting HIF-1α/VEGF pathway. Int J Clin Exp Pathol. 8:4735–4744. 2015.PubMed/NCBI

86 

Maged AM, Deeb WS, El Amir A, Zaki SS, El Sawah H, Al Mohamady M, Metwally AA and Katta MA: Diagnostic accuracy of serum miR-122 and miR-199a in women with endometriosis. Int J Gynaecol Obstet. 141:14–19. 2018. View Article : Google Scholar : PubMed/NCBI

87 

Hsu CY, Hsieh TH, Tsai CF, Tsai HP, Chen HS, Chang Y, Chuang HY, Lee JN, Hsu YL and Tsai EM: miRNA-199a-5p regulates VEGFA in endometrial mesenchymal stem cells and contributes to the pathogenesis of endometriosis. J Pathol. 232:330–343. 2014. View Article : Google Scholar : PubMed/NCBI

88 

Lin SC, Wang CC, Wu MH, Yang SH, Li YH and Tsai SJ: Hypoxia-induced microRNA-20a expression increases ERK phosphorylation and angiogenic gene expression in endometriotic stromal cells. J Clin Endocrinol Metab. 97:E1515–1523. 2012. View Article : Google Scholar : PubMed/NCBI

89 

Lei Z, Li B, Yang Z, Fang H, Zhang GM, Feng ZH and Huang B: Regulation of HIF-1alpha and VEGF by miR-20b tunes tumor cells to adapt to the alteration of oxygen concentration. PLoS One. 4:e76292009. View Article : Google Scholar : PubMed/NCBI

90 

Ramón LA, Braza-Boïls A, Gilabert-Estellés J, Gilabert J, España F, Chirivella M and Estellés A: microRNAs expression in endometriosis and their relation to angiogenic factors. Hum Reprod. 26:1082–1090. 2011. View Article : Google Scholar : PubMed/NCBI

91 

Marí-Alexandre J, García-Oms J, Barceló-Molina M, Gilabert-Aguilar J, Estellés A, Braza-Boíls A and Gilabert-Estellés J: MicroRNAs and angiogenesis in endometriosis. Thromb Res. 135 (Suppl 1):S38–S40. 2015. View Article : Google Scholar : PubMed/NCBI

92 

van Solingen C, Seghers L, Bijkerk R, Duijs JM, Roeten MK, van Oeveren-Rietdijk AM, Baelde HJ, Monge M, Vos JB, de Boer HC, et al: Antagomir-mediated silencing of endothelial cell specific microRNA-126 impairs ischemia-induced angiogenesis. J Cell Mol Med. 13((8A)): 1577–1585. 2009. View Article : Google Scholar : PubMed/NCBI

93 

Panda H, Pelakh L, Chuang TD, Luo X, Bukulmez O and Chegini N: Endometrial miR-200c is altered during transformation into cancerous states and targets the expression of ZEBs, VEGFA, FLT1, IKKβ, KLF9, and FBLN5. Reprod Sci. 19:786–796. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Rekker K, Saare M, Roost AM, Kaart T, Sõritsa D, Karro H, Sõritsa A, Simón C, Salumets A and Peters M: Circulating miR-200-family micro-RNAs have altered plasma levels in patients with endometriosis and vary with blood collection time. Fertil Steril. 104:938–946.e2. 2015. View Article : Google Scholar : PubMed/NCBI

95 

Misir S, Hepokur C, Oksasoglu B, Yildiz C, Yanik A and Aliyazicioglu Y: Circulating serum miR-200c and miR-34a-5p as diagnostic biomarkers for endometriosis. J Gynecol Obstet Hum Reprod. 50:1020922021. View Article : Google Scholar : PubMed/NCBI

96 

Papari E, Noruzinia M, Kashani L and Foster WG: Identification of candidate microRNA markers of endometriosis with the use of next-generation sequencing and quantitative real-time polymerase chain reaction. Fertil Steril. 113:1232–1241. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Nisenblat V, Sharkey DJ, Wang Z, Evans SF, Healey M, Ohlsson Teague EMC, Print CG, Robertson SA and Hull ML: Plasma miRNAs display limited potential as diagnostic tools for endometriosis. J Clin Endocrinol Metab. 104:1999–2022. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Wang WT, Zhao YN, Han BW, Hong SJ and Chen YQ: Circulating microRNAs identified in a genome-wide serum microRNA expression analysis as noninvasive biomarkers for endometriosis. J Clin Endocrinol Metab. 98:281–289. 2013. View Article : Google Scholar : PubMed/NCBI

99 

Zafari N, Tarafdari AM, Izadi P, Noruzinia M, Yekaninejad MS, Bahramy A and Mohebalian A: A Panel of Plasma miRNAs 199b-3p, 224-5p and Let-7d-3p as non-invasive diagnostic biomarkers for endometriosis. Reprod Sci. 28:991–999. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Marí-Alexandre J, Carcelén AP, Agababyan C, Moreno-Manuel A, García-Oms J, Calabuig-Fariñas S and Gilabert-Estellés J: Interplay Between MicroRNAs and oxidative stress in ovarian conditions with a focus on ovarian cancer and endometriosis. Int J Mol Sci. 20:53222019. View Article : Google Scholar : PubMed/NCBI

101 

Zhou S, Huang C, Wang W and Liu J: MiR-370-3p inhibits the development of human endometriosis by downregulating EDN1 expression in endometrial stromal cells. Cell Biol Int. 45:1183–1190. 2021. View Article : Google Scholar : PubMed/NCBI

102 

Khan M, Shah PM, Khan IA, Islam SU, Ahmad Z, Khan F and Lee Y: IoMT-Enabled Computer-Aided Diagnosis of pulmonary embolism from computed tomography scans using deep learning. Sensors (Basel). 23:14712023. View Article : Google Scholar : PubMed/NCBI

103 

Mohd Aman AH, Hassan WH, Sameen S, Attarbashi ZS, Alizadeh M and Latiff LA: IoMT amid COVID-19 pandemic: Application, architecture, technology, and security. J Netw Comput Appl. 174:1028862021. View Article : Google Scholar : PubMed/NCBI

104 

Wang F, Wang H, Jin D and Zhang Y: Serum miR-17, IL-4, and IL-6 levels for diagnosis of endometriosis. Medicine (Baltimore). 97:e108532018. View Article : Google Scholar : PubMed/NCBI

105 

Menzhinskaya IV, Pavlovich SV, Melkumyan AG, Chuprynin VD, Yarotskaya EL and Sukhikh GT: Potential significance of serum autoantibodies to endometrial antigens, α-Enolase and hormones in non-invasive diagnosis and pathogenesis of endometriosis. Int J Mol Sci. 24:155782023. View Article : Google Scholar : PubMed/NCBI

106 

Mathiasen M, Egekvist AG, Kesmodel US, Knudsen UB and Seyer-Hansen M: Similar evolution of pain symptoms and quality of life in women with and without endometriosis undergoing assisted reproductive technology (ART). Acta Obstet Gynecol Scand. 98:77–85. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Gao Y, Liu P and Shi R: Anlotinib as a molecular targeted therapy for tumors. Oncol Lett. 20:1001–1014. 2020. View Article : Google Scholar : PubMed/NCBI

108 

He Y, Hung SW, Liang B, Zhang R, Gao Y, Chu CY, Zhang T, Xu H, Chung JPW and Wang CC: Receptor tyrosine kinase inhibitor sunitinib as novel immunotherapy to inhibit myeloid-derived suppressor cells for treatment of endometriosis. Front Immunol. 12:6412062021. View Article : Google Scholar : PubMed/NCBI

109 

Yildiz C, Kacan T, Akkar OB, Karakus S, Kacan SB, Ozer H and Cetin A: Effects of pazopanib, sunitinib, and sorafenib, Anti-VEGF agents, on the growth of experimental endometriosis in rats. Reprod Sci. 22:1445–1451. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Iampietro C, Brossa A, Canosa S, Tritta S, Croston GE, Reinheimer TM, Bonelli F, Carosso AR, Gennarelli G, Cosma S, et al: Quinagolide treatment reduces invasive and angiogenic properties of endometrial mesenchymal stromal cells. Int J Mol Sci. 23:17752022. View Article : Google Scholar : PubMed/NCBI

111 

Pellicer N, Galliano D, Herraiz S, Bagger YZ, Arce JC and Pellicer A: Use of dopamine agonists to target angiogenesis in women with endometriosis. Hum Reprod. 36:850–858. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Hamid AM, Madkour WA, Moawad A, Elzaher MA and Roberts MP: Does cabergoline help in decreasing endometrioma size compared to LHRH agonist? A prospective randomized study. Arch Gynecol Obstet. 290:677–682. 2014. View Article : Google Scholar : PubMed/NCBI

113 

Mendoza-Torreblanca JG, Cárdenas-Rodríguez N, Carro-Rodríguez J, Contreras-García IJ, Garciadiego-Cázares D, Ortega-Cuellar D, Martínez-López V, Alfaro-Rodríguez A, Evia-Ramírez AN, Ignacio-Mejía I, et al: Antiangiogenic effect of dopamine and dopaminergic agonists as an adjuvant therapeutic option in the treatment of cancer, endometriosis, and osteoarthritis. Int J Mol Sci. 24:101992023. View Article : Google Scholar : PubMed/NCBI

114 

Olivares C, Ricci A, Bilotas M, Barañao RI and Meresman G: The inhibitory effect of celecoxib and rosiglitazone on experimental endometriosis. Fertil Steril. 96:428–433. 2011. View Article : Google Scholar : PubMed/NCBI

115 

Zhang S, Zhuang L, Liu Q, Yu X, Min Q, Chen M and Chen Q: Rosiglitazone affects the progression of surgically-induced endometriosis in a rat model. Mol Med Rep. 23:352021.PubMed/NCBI

116 

Nenicu A, Körbel C, Gu Y, Menger MD and Laschke MW: Combined blockade of angiotensin II type 1 receptor and activation of peroxisome proliferator-activated receptor-γ by telmisartan effectively inhibits vascularization and growth of murine endometriosis-like lesions. Hum Reprod. 29:1011–1024. 2014. View Article : Google Scholar : PubMed/NCBI

117 

Nenicu A, Gu Y, Körbel C, Menger MD and Laschke MW: Combination therapy with telmisartan and parecoxib induces regression of endometriotic lesions. Br J Pharmacol. 174:2623–2635. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Nasu K, Nishida M, Ueda T, Yuge A, Takai N and Narahara H: Application of the nuclear factor-kappaB inhibitor BAY 11-7085 for the treatment of endometriosis: An in vitro study. Am J Physiol Endocrinol Metab. 293:E16–E23. 2007. View Article : Google Scholar : PubMed/NCBI

119 

Zhang JJ, Xu ZM, Chang H, Zhang CM, Dai HY, Ji XQ, Li C and Wang XF: Pyrrolidine dithiocarbamate attenuates nuclear factor-ĸB activation, cyclooxygenase-2 expression and prostaglandin E2 production in human endometriotic epithelial cells. Gynecol Obstet Invest. 72:163–168. 2011. View Article : Google Scholar : PubMed/NCBI

120 

Huang R, Chen S, Zhao M, Li Z and Zhu L: Ginsenoside Rg3 attenuates endometriosis by inhibiting the viability of human ectopic endometrial stromal cells through the nuclear factor-kappaB signaling pathway. J Gynecol Obstet Hum Reprod. 49:1016422020. View Article : Google Scholar : PubMed/NCBI

121 

Vallée A and Lecarpentier Y: Curcumin and Endometriosis. Int J Mol Sci. 21:E24402020. View Article : Google Scholar

122 

Chowdhury I, Banerjee S, Driss A, Xu W, Mehrabi S, Nezhat C, Sidell N, Taylor RN and Thompson WE: Curcumin attenuates proangiogenic and proinflammatory factors in human eutopic endometrial stromal cells through the NF-κB signaling pathway. J Cell Physiol. 234:6298–6312. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Wei X and Shao X: Nobiletin alleviates endometriosis via down-regulating NF-κB activity in endometriosis mouse model. Biosci Rep. 38:BSR201804702018. View Article : Google Scholar : PubMed/NCBI

124 

Uludag SZ, Demirtas E, Sahin Y and Aygen EM: Dienogest reduces endometrioma volume and endometriosis-related pain symptoms. J Obstet Gynaecol. 41:1246–1251. 2021. View Article : Google Scholar : PubMed/NCBI

125 

Keleş CD, Vural B, Filiz S, Vural F, Gacar G, Eraldemir FC and Kurnaz S: The effects of etanercept and cabergoline on endometriotic implants, uterus and ovaries in rat endometriosis model. J Reprod Immunol. 146:1033402021. View Article : Google Scholar : PubMed/NCBI

126 

Ren XU, Wang Y, Xu G and Dai L: Effect of rapamycin on endometriosis in mice. Exp Ther Med. 12:101–106. 2016. View Article : Google Scholar : PubMed/NCBI

127 

Grammatis AL, Georgiou EX and Becker CM: Pentoxifylline for the treatment of endometriosis-associated pain and infertility. Cochrane Database Syst Rev. 8:CD0076772021.PubMed/NCBI

128 

Dicitore A, Castiglioni S, Saronni D, Gentilini D, Borghi MO, Stabile S, Vignali M, Di Blasio AM, Persani L and Vitale G: Effects of human recombinant type I IFNs (IFN-α2b and IFN-β1a) on growth and migration of primary endometrial stromal cells from women with deeply infiltrating endometriosis: A preliminary study. Eur J Obstet Gynecol Reprod Biol. 230:192–198. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Söderman L, Böttiger Y, Edlund M, Järnbert-Pettersson H and Marions L: Adjuvant use of melatonin for pain management in endometriosis-associated pelvic pain-A randomized double-blinded, placebo-controlled trial. PLoS One. 18:e02861822023. View Article : Google Scholar : PubMed/NCBI

130 

Vašková J, Klepcová Z, Špaková I, Urdzík P, Štofilová J, Bertková I, Kľoc M and Rabajdová M: The importance of natural antioxidants in female reproduction. Antioxidants (Basel). 12:9072023. View Article : Google Scholar : PubMed/NCBI

131 

Schwertner A, Conceição Dos Santos CC, Costa GD, Deitos A, de Souza A, de Souza IC, Torres IL, da Cunha Filho JS and Caumo W: Efficacy of melatonin in the treatment of endometriosis: A phase II, randomized, double-blind, placebo-controlled trial. Pain. 154:874–881. 2013. View Article : Google Scholar : PubMed/NCBI

132 

Santanam N, Kavtaradze N, Murphy A, Dominguez C and Parthasarathy S: Antioxidant supplementation reduces endometriosis-related pelvic pain in humans. Transl Res. 161:189–195. 2013. View Article : Google Scholar : PubMed/NCBI

133 

Li Y, Zeng X, Lu D, Yin M, Shan M and Gao Y: Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 36:951–964. 2021. View Article : Google Scholar : PubMed/NCBI

134 

Bouquet de Joliniere J, Fruscalzo A, Khomsi F, Stochino Loi E, Cherbanyk F, Ayoubi JM and Feki A: Antiangiogenic therapy as a new strategy in the treatment of endometriosis? The first case report. Front Surg. 8:7916862021. View Article : Google Scholar : PubMed/NCBI

135 

Bodnar RJ: Anti-Angiogenic Drugs: Involvement in cutaneous side effects and wound-healing complication. Adv Wound Care (New Rochelle). 3:635–646. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2024
Volume 29 Issue 3

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bo C and Bo C: Angiogenesis signaling in endometriosis: Molecules, diagnosis and treatment (Review). Mol Med Rep 29: 43, 2024
APA
Bo, C., & Bo, C. (2024). Angiogenesis signaling in endometriosis: Molecules, diagnosis and treatment (Review). Molecular Medicine Reports, 29, 43. https://doi.org/10.3892/mmr.2024.13167
MLA
Bo, C., Wang, Y."Angiogenesis signaling in endometriosis: Molecules, diagnosis and treatment (Review)". Molecular Medicine Reports 29.3 (2024): 43.
Chicago
Bo, C., Wang, Y."Angiogenesis signaling in endometriosis: Molecules, diagnosis and treatment (Review)". Molecular Medicine Reports 29, no. 3 (2024): 43. https://doi.org/10.3892/mmr.2024.13167