Open Access

Molecular mechanisms underlying the inhibition of cell migration and invasion in endometriosis: Advances in pharmacological research (Review)

  • Authors:
    • Ni Wei
    • Haibin Guan
    • Yanfen Zhang
    • Jianping Shi
    • Jiannan Ma
    • Ruiweng Shi
    • Xiao Qi
    • Zhiheng Dong
    • Rongwei Zhao
  • View Affiliations

  • Published online on: July 8, 2025     https://doi.org/10.3892/br.2025.2030
  • Article Number: 152
  • Copyright: © Wei et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Endometriosis (EMS) is a prevalent gynecological disease afflicting reproductive‑age women, characterized by the presence of endometrial tissue outside the uterus. The clinical manifestations of this condition include pelvic pain, dyspareunia, and, in severe cases, infertility. The pathology of EMS is similar to that of malignant tumors in terms of implantation, invasion and metastasis, which complicates diagnosis and treatment. Ectopic endometrium has the potential to invade the ovaries, the uterosacral ligament, the peritoneum, and the vaginal wall. Its core mechanism involves the dynamic regulation of cell migration and invasion, which is an important process affecting the development and distant metastasis of EMS. Therefore, understanding the underlying mechanisms of its formation and development is crucial for its treatment. At present, research on inhibitors targeting such mechanisms remains in the exploratory stage and has yet to be translated into clinical applications. The present study aimed to methodically review the molecular regulatory network of cell migration and invasion in EMS, encompassing the interactions between relevant cells, the functions of key signaling pathways, and regulatory factors. The authors focused on chemically synthesized drugs, plant extracts, Chinese medicine, potential targets of action and mechanisms. The present review offers novel research concepts and theoretical foundations that may facilitate the identification of novel therapeutic targets for EMS. These targets could be implemented in clinical settings to effectively mitigate metastasis and recurrence rates, thereby enhancing our comprehension of the intricacies of the disease, and guiding the exploration of more efficacious therapeutic avenues.

1. Introduction

Endometriosis (EMS) is a chronic inflammatory gynecological disease characterized by the invasive growth of ectopic endometrial tissue outside the uterine cavity. The clinical manifestations of EMS include severe dysmenorrhea, menstrual abnormalities and fertility disorders (1). Epidemiologic data (1,2), show that ~30-50% of patients with EMS have fertility disorders, while 50-80% of cases of pelvic pain are associated with the disease, which has a global prevalence of more than 176 million. However, the absence of specific biomarkers and the presence of non-specific clinical symptoms in the early stages of the disease often result in delayed or misdiagnosis (3), which can lead to suboptimal treatment outcomes and a recurrent course of pain.

In 1986, Sanfilippo et al (4) proposed that the primary causative factor of EMS is reversal of menstrual blood flow, and the disease is now regarded as systemic, rather than one that is primarily confined to the pelvis (1). The classification of EMS is based on its location and severity, with three distinct forms: superficial, ovarian and deep infiltrative (5), as shown in Fig. 1. The heterogeneity of EMS across different subtypes is a significant factor contributing to the challenges associated with the development of effective medication. Furthermore, the development of EMS may be influenced by a variety of factors, including geographic location (6), ethnicity (7), genetics (8), lifestyle and dietary habits (9).

The etiology of endometriotic lesions remains a subject of considerable debate, with several theories proposing potential mechanisms, including retrograde menstruation, cellular chemotaxis, involvement of stem cells, blood or lymphatic dissemination, and the theory of embryogenesis introduced by Burney and Giudice (10). Among the various mechanisms, cells may require the capacity to migrate, invade, and adapt to new microenvironments, and studies have demonstrated that cell migration and invasion are of paramount importance in this context (11-13). However, it is imperative to acknowledge the interplay between cells associated with EMS, such as endometrial stromal cells (ESCs), endometrial epithelial cells (EECs), immune cells and stem cells, among others (14-18). Studies (19,20) have confirmed the migration, invasion, epithelial-mesenchymal transition (EMT) and inflammation of the relevant cells through in vitro experimentation, thereby providing a foundation to predict the therapeutic effect.

Another study (21) elaborated on the pathogenesis of related molecules in EMS by regulating the proliferation and migratory activity of ESCs. Estrogen signaling plays a central regulatory role in the remodeling of the disease microenvironment. For instance, 17β-estradiol influences lesion progression by modulating the expression levels of inflammatory factors (22). The signaling pathways associated with migration and invasion of EMS cells, as well as their upstream and downstream regulators, constitute a large and complex transduction system. Abnormalities in these pathways and their interactions can lead to abnormal proliferation, apoptosis, migration, invasion, angiogenesis, immune system, and inflammatory responses in ectopic endometrial tissues, resulting in rapid proliferation of lesions. In this process, matrix metalloproteinases (MMPs) (23), adhesion molecules (24), small-molecule proteins (25) and hormone receptor systems (26) play important roles. The process of cell migration and invasion is influenced by the dysregulation of downstream molecules, such as MMPs and adhesion molecules, due to localized inflammation and immune responses within the cellular microenvironment.

The prevailing treatment for EMS is primarily surgical resection, followed by the postoperative administration of gonadotropin-releasing hormone agonist (GnRH-a) (27). However, it is improbable that pharmacologic therapy will fully attenuate the cells. The primary adverse effects associated with prolonged GnRH-a administration are perimenopausal symptoms and hypoestrogenic-induced osteoporosis (28), and some patients may also encounter severe bone pain.

With the advent of molecular targeted therapy and gene editing technology, precision diagnostic systems and gene therapy (29) based on blood microRNA (miRNA or miR) biomarkers (30) have gradually become a research focus. miR-193b-5p and miR-374b-5p can inhibit endometrial cell migration in ex vivo experiments (31). KLF6 is a tumor suppressor gene (32), and overexpression thereof significantly inhibits ECESC proliferation, migration and invasion, as well as induces cell apoptosis in eutopic ESCs (EuESCs) and ectopic ESCs (EESCs) knockdown and overexpression assays (33).

Current research has focused on the exploration of the mechanisms of natural drug active ingredients. For example, a recent study by Meng et al (34) investigated the anti-EMS mechanism of the classic Chinese herbal medicine formula Juan-Tong-Yin. It was found that the formula could improve the inflammatory microenvironment by enhancing endoplasmic reticulum stress and autophagy, and decreasing the migration and invasion of ESCs to treat EMS (35).

The molecular mechanisms associated with EMS cell migration and invasion, as well as drug studies, are crucial for clinical treatment. It was therefore aimed to review the mechanisms of action and risks of these drugs to develop more effective treatments for EMS.

2. Mechanisms of EMS cell migration and invasion

Although a number of studies have been devoted to unraveling the pathogenesis and treatment of EMS, its pathophysiology and molecular mechanisms are still not fully understood. Cell migration and invasion are important factors in the pathogenesis thereof and are important processes that promote the formation and growth of ectopic endometrial tissues, with mechanisms involving the regulation of cell adhesion molecules, MMPs, extracellular matrix (ECM) remodeling, the hypoxic microenvironment and mesenchymal transition, among other factors.

Anti-cell migration and invasion mechanism

The development and progression of EMS is dependent on the migration and invasion of endometriotic cells (36), as shown in Table I (37-40). An increasing number of researchers are judging the effectiveness of intervention components by studying the migration and invasion abilities of EMS cell models, and through continuous development, the research direction is gradually moving toward non-invasive biomarker screening, the establishment of in vivo animal models, the study of related signaling pathways, and the prospective treatment with targeted drugs.

Table I

Inhibitory effects on cell migration and invasion across different tumor types.

Table I

Inhibitory effects on cell migration and invasion across different tumor types.

First author/s, yearTumorMechanismImpactTarget(Refs.)
Mao et al, 2019CCCSN6 promotes the migration and invasion of CC cells by inhibiting autophagic degradation of Cathepsin LHighly lethal and aggressiveCSN6(37)
Gao et al, 2020OCMiR-26a inhibits ovarian cancer cell proliferation, migration and invasion by targeting TCF12Survival and poor prognosisTCF12(38)
Geng et al, 2021ECMiR-29a-3p inhibits EC cell proliferation, migration and invasion by targeting VEGFA/CD C42/PAK1Tumor metastasis, invasion and poor prognosismiR-29a-3p(39)

[i] OC, ovarian cancer; EC, endometrial cancer; CC, cervical cancer; miR, microRNA.

Cellular characterization of EMS. The pathological evolution of EMS is closely related to cell migration, adhesion, invasive behavior, dynamic remodeling of the cytoskeleton, microenvironmental remodeling, and the EMT process, which involves interactions between multiple cell types. Among them, ESCs, as core effector cells, can be ectopically implanted and initiate lesion formation via the transepithelial reflux pathway. ESCs from patients with EMS present a unique pathological phenotype when interacting with peritoneal mesothelial cells (PMCs). This includes the upregulation of adhesion molecule expression to enhance the anchoring ability of the cells, the aberrant mesenchymal marker expression driving EMT progression, invasive pseudopod formation, and increased secretion of matrix degrading enzymes, as well as more invasive and motile ESCs promoted by PMCs (14,15). ESCs from patients with EMS exhibit significant changes in mechanical properties, such as reduced cell deformability and stiffness, which makes these cells susceptible to migration and spreading in the microenvironment (41).

Additionally, diseased ESCs often show myofibroblast-like transformation accompanied by α-SMA overexpression and increased collagen secretion, and their pro-fibrotic features have molecular similarities with tumor microenvironment (TME) remodeling (42). As shown in Fig. 2, the peritoneal cavity immune network exhibits multicellular synergistic features, with macrophages amplifying local inflammation through CCL2/CXCL1-mediated neutrophil recruitment, which promotes the proliferation and migration of EESCs (16,17). NK cells secrete IFN-γ to regulate Th1/Th2 balance, and dendritic cells activate T-cell immune responses through MHC-II-mediated antigen presentation (43-46). Mast cells are also involved in the immune and inflammatory response therein through recruitment and differentiation, secretion of pro-inflammatory mediators (47,48). A recent study (49) identified a novel class of ‘Pale cells’, which lack bridging granule structures and have a translucent cytoplasm, and may be involved in migration and invasion, but the lack of in vitro culture models prevents further investigation.

Menstrual blood-derived stem cells exhibit abnormal homing ability and paracrine dysfunction in patients with EMS, and exhibit different properties in patients with EMS, which may be biomarkers for early diagnosis and treatment (18). Taken together, the cellular heterogeneity and multicellular interaction network of EMS provide key entry points to analyze the pathogenesis and develop targeted intervention strategies.

PI3K/Akt/p-mTOR pathway. The ability of EMS cells to migrate and invade is a central pathological feature driving lesion development, progression and ectopic dissemination. The molecular mechanism involves a complex cascade of abnormalities in the function of the resident endometrial stem cells, interactions of multiple molecular networks, and dynamic regulation of the microenvironment. Cellular and molecular studies have shown that a variety of proteins, genes, and compounds can regulate the process of cell migration and invasion by activating specific signaling pathways. As a heterodimer with serine/threonine kinase activity, phosphatidylinositol 3-kinase (PI3K) plays a key role in the regulation of cell migration and invasion by collaborating with protein kinase B to form a classical signal transduction axis. A typical example is DJ-1 protein, which promotes endometrial cell proliferation, migration, and angiogenesis by activating the PI3K/Akt/p-mTOR signaling cascade, suggesting its potential value as a common therapeutic target for EMS and adenomyosis (50).

Immunoglobulin superfamily member 8 (IGSF8), known as EWI-2, inhibits endometrial cell proliferation, invasion and filamentous pseudopod formation by negatively regulating the Akt signaling pathway (51,52). Zheng and Yang (53) demonstrated by protein blotting analysis that knockdown of EWI-2 could significantly enhance the level of Akt phosphorylation, which promotes the migratory invasion ability of EECs, revealing a critical negative regulatory role of the EWI-2-PI3K/Akt axis in EMS. These gene-level-based studies may provide new directions and further ideas for subsequent research strategies.

In addition, the plant-derived flavonoid 3,6-dihydroxyflavone exerts anticancer effects by inhibiting pathological processes such as malignant transformation and invasion of tumor cells (54,55). This compound significantly reduces the migratory invasive ability of ESCs by inhibiting the expression of key effector molecules of the Notch signaling pathway (56). In vitro assays have demonstrated that the PI3K/Akt pathway agonist 740 Y-P effectively reverses the effects of GATA-binding protein gene silencing on the proliferation, migration and death of EESCs (57). A mechanistic analysis has also revealed that PGRN overexpression significantly elevates the p-Akt/Akt ratio, while the PI3K inhibitor LY294002 completely blocks the promotion of PGRN on ESC proliferation and invasion (58). Further studies are needed to develop antibody therapy to block EMS and improve targeting efficiency.

Wnt/β-catenin and NF-κB pathways. The wingless-type mouse mammary tumor virus integration site family (Wnt) signaling axis plays a critical role in regulating EMS cell migration and invasion. The silencing of the multidrug resistance protein 4 gene can contribute to the progression of endometrial lesions by inducing aberrant activation of Wnt/β-catenin signaling (59). The downstream effector molecules of this pathway, MMP-2/-9 and cyclin D1, are significantly overexpressed in EECs and ESCs of patients with EMS (60).

The MMP family, a group of key effector molecules, has also been shown to regulate cell migration, invasion and angiogenesis through the modulation of growth factor/cytokine dynamics. Abnormally high expression of MMPs is positively correlated with the pathological progression of EMS (61). Furthermore, T-cell factor/β-catenin (Tcf/β-catenin) complexes can transcriptionally inhibit MMP-9 activity and restore cell invasion ability to normal endothelial levels, suggesting their potential value as therapeutic targets for EMS (62). However, the field of EMS research has yet to dedicate significant resources to this topic.

NF-κB is a class of key proteins that can significantly drive the cell proliferation process while effectively inhibiting the apoptosis phenomenon in endometrium and endometriotic cells. In 2018, Li et al (63) identified an oncogene, programmed cell death 4, which inhibits cell proliferation by suppressing autophagy and the NF-κB/MMP2/MMP9 signaling pathway, thereby inhibiting endometrial cell proliferation, migration and invasion. This jointly inhibits disease progression at the transcriptional and translational levels.

Effect of the cellular microenvironment. Hypoxic microenvironment

The hypoxic microenvironment is an important factor affecting EMS cell proliferation and invasion, promoting ESC migration and invasion by upregulating autophagy. Autophagy is an intracellular process of protein and organelle degradation that is mediated through lysosomes, with the primary function of maintaining stability of the intracellular environment (64). In the physiological state, the process is cytoprotective; however, excessive activation or dysfunction of autophagy can lead to organelle depletion and induce programmed death (65-67).

Preliminary experimental studies have demonstrated that treatment with hypoxia results in enhanced migration of EECs, concurrently inducing autophagy, thereby activating the EMT process (68). Mechanistic elucidation (69) has revealed that hypoxia promotes the formation of autophagic vesicles by stabilizing hypoxia-inducible factor-1α (HIF-1α), which drives the invasive phenotype of EESCs. Paeonol intervention inhibits this process in a dose-dependent manner, downregulating the LC3-II/LC3-I ratio, and upregulating p62 expression. This ultimately restores the migratory capacity of EESCs to normoxic levels. The HIF-1α/autophagy signaling axis amplifies the invasive potential of EMS foci through a positive feedback loop (70).

In a recent study, a network pharmacology in vitro and in vivo model demonstrated that the Chinese herbal compound ‘Luoshi Neiyi prescription’ significantly inhibited endometrial stromal cell adhesion and invasion by targeting the HIF1A/EZH2/ANTXR2 signaling axis (71). Hypoxia promotes EEC invasion (68), and the initial events that are critical in the formation of EMS lesions are initiated by cellular responses to hypoxic conditions, which subsequently activate the autophagy cascade effect. This activation drives the process of EMT while enhancing the invasive potential of endometriotic cells (72).

Furthermore, the hypoxic microenvironment plays a pivotal role in the migration and invasion of tumor cells, with the process being regulated by carbonic anhydrase IX (CAIX)-mediated pH dynamics (73). The functional significance of CAIX in tumor cell migration and invasion has been well-established; however, its potential applications in the context of EMS cell migration and invasion remain to be elucidated. Consequently, the concerted targeting of the hypoxia-autophagy-EMT regulatory network may emerge as a novel therapeutic strategy for the treatment of EMS.

EMT. EMT is a complex event that drives the transformation of polarized epithelial cells from adherent cells to motile mesenchymal cells. This process involves immune cells and stromal cells, and EMT plays a crucial role in migration and invasion in EMS (74). During the process thereof, cells undergo a series of significant changes, including the loss of cell polarity; attachment to the basement membrane; enhanced invasion, migration, anti-apoptosis; and ECM degradation; as well as a decrease in cell adhesion (75). Aberrant activation of the oncogenic signaling pathway, the presence of hypoxia, and the interactions with stromal cells are crucial factors that contribute to the process of EMT. This leads to a decrease in intercellular adhesion and an enhancement in migratory and invasive phenotypes (76).

EMT is more active in non-endometriotic tissues compared with ectopic endometriotic tissues (77). The novel findings of Liu et al (68) suggest that hypoxia-induced activation of autophagy cascades is central to the invasive phenotypic profile of EMT and endometriotic implant cells. They also found that the interaction between endometriotic implants and the surrounding peritoneal microenvironment may influence the endometrial microenvironment. These interactions exert a significant influence on the development of peritoneal EMS, and it was sought to explore the classification of EMT for the purpose of facilitating a more precise diagnosis and treatment of peritoneal EMS.

EMS and malignant neoplasms, including ovarian cancer (OC) and breast cancer (BC), exhibit significant similarities at the level of molecular mechanisms, such as autophagy regulation, EMT processes and ECM remodeling. The transition from an epithelial to a mesenchymal phenotype is associated with systemic dissemination of cells from the primary site, which significantly enhances the migration and invasion of tumor cells (78). Transmembrane protein 176B can effectively block the EMT process by inhibiting the activation of the Wnt/β-catenin signaling pathway, which inhibits OC cell migration, invasion and adhesion (79). Complementary evidence based on ex vivo and in vivo experiments has shown that lysine acetyltransferase 2B gene silencing enhances autophagic activity through activation of the TGF-β/Smad3/7 signaling pathway, which drives the EMT-dependent proliferative and invasive phenotypes of epithelial OC (80).

In endometrial cancer (EC) studies, kinesin family member 15 knockdown significantly inhibited the EMT process by suppressing Wnt/β-catenin signaling, thereby reducing tumor cell proliferation, cloning, migration and invasive capacity (81). A study based on preclinical models and pre- and post-treatment biopsy samples from patients with EC confirmed that systemic application of an anti-netrin-1 antibody significantly reduced the expression of EMT signature markers in tumor cells and increased chemosensitivity (82). Although the molecular mechanisms by which EMT regulates the malignant phenotype of tumors have been thoroughly analyzed, clinical interventions targeting the EMT process are still extremely limited, and the clinical trial conducted by Cassier et al (82) is the first to systematically evaluate antitumor strategies targeting the EMT signature, providing an important paradigm for translational medicine research.

Mechanisms related to cervical cancer (CC) have shown that EFEMP2 gene silencing can significantly inhibit cancer cell proliferation and invasion by synergistically downregulating MMP-1/3/10/13 expression, blocking EMT progression, and inhibiting the Raf/MEK/ERK signaling pathway (83). Conversely, EFEMP2 overexpression promoted tumor progression by inducing EMT and activating the Raf/MEK/ERK signaling pathway (83). Similarly, the metastatic process of BC was found to be closely associated with EMT-mediated dysregulation of MMP-9/13 expression (84). As demonstrated in Fig. 3, a comprehensive review of the existing literature revealed that the mechanisms of EMT associated with EMS and the female reproduction-related cancers (OC, EC, CC and BC) were analogous. A comparative analysis of the EMT features of EMS and cancer revealed numerous commonalities, particularly with regard to MMPs.

Future studies should systematically elucidate the multidimensional mechanisms by which the hypoxic microenvironment regulates the progression of EMS, with a focus on resolving the synergistic network of hypoxia-induced autophagy and EMT. It is recommended that the well-established EMT regulation strategies be integrated into tumor invasion research, and that a ‘hypoxia-autophagy-EMT’ cascade model be constructed by targeting the HIF-1α/PI3K/Akt signaling axis to intervene at the key nodes of EMT (for example, Snail, Twist1 and ZEB1). This research direction will facilitate the elucidation of the molecular foundations that govern the acquisition of invasive properties by ectopic endothelial cells. Moreover, this will provide a theoretical framework for the development of multi-targeted therapeutic strategies that are based on the inhibition of EMT.

Molecular mechanisms associated with cell migration and invasion in EMS

Abnormalities in the molecular mechanisms associated with cell migration and invasion result in persistent growth and inflammatory responses of ectopic endometrial tissue. The MMP family plays an important role in cell migration and invasion. In addition, RNA molecules are involved in the adhesion and migration of ectopic endometrial cells to the surrounding tissues. Further research is needed to investigate the role of cytokines, inhibitors, proteins and genes in this process. However, due to the intricate pathogenesis of EMS, as illustrated in Fig. 4, the research timeline concerning molecules implicated in the design of EMS migration and invasion, the prevailing treatment for EMS as a first-line drug therapy and the lack of molecularly-targeted drugs available for clinical application, make it necessary to consider the feasibility and effectiveness of molecular drugs.

MMPs associated with EMS cell migration and invasion. MMPs are a family of at least 15 secreted and membrane-bound zinc endopeptidases that degrade all components of the ECM, including fibrillar and nonfibrillar collagens, fibronectin, laminin, and basement membrane glycoproteins. MMPs are imperative for a range of invasive processes implicated in angiogenesis and tumor metastasis (85). Among these, MMP-2 and MMP-9 are key effector molecules, whose expression levels are significantly and positively correlated with the ectopic invasive ability of EMS, and the progress of the MMP family in EMS-associated migration and invasion, as shown in Table II (86-93).

Table II

MMPs associated with endometriosis progression: Mechanisms regulating cell migration and invasion.

Table II

MMPs associated with endometriosis progression: Mechanisms regulating cell migration and invasion.

First author/s, yearTypes of MMPMachineRoleReported elsewhere(Refs.)
Sharpe-Timms et al, 1998TIMP-1Concentrations in peritoneal fluid and serum are lower in patients with endometriosis than in normal womenDiagnosing endometriosis and monitoring treatment outcomesCC, EC(86)
Wenzl et al, 1998MMP-2Endometriotic tissues express proteases capable of invading surrounding tissuesLocalization in the uterine endometrium and ectopic implantsOC, EC, CC, BC(87)
Cox et al, 2001; Lv et al, 2015MMP-3Ectopic endometrial MMP-3 may be involved in the process of invasion and tissue remodeling that occurs in the pathogenesis of endometriosis. miR-93 inhibits the proliferation, migration and invasion of endometrial stromal cells by combining MMP3 and VEGFADifferential regulation of matrix metalloproteinase-3 gene expression in endometriotic lesions in comparison with endometrium, inhibition of endometriosis migrationOC, CC(88,89)
Chung et al, 2002TIMP-2Expression levels are associated with endometriosis aggressivenessExplain the aggressive factors leading to endometriosisEC, CC(90)
Collette et al, 2006; Lin et al, 2021MMP-9MMP-9/TIMP-1 imbalance is involved in tissue Invasion and Ectopic Development in patients with endometriosis. Cytokine IL-33 acts on stromal cells to promote cell invasion and MMP-9 expression in endometriosisSupporting an important role for MMP-9 in the ability of endometrial tissue to invade and develop into host tissue. Providing new strategies for the prevention and treatment of endometriosisOC, EC, CC, BC(91,92)
Gaetje et al, 2007MT5-MMPMT5-MMP expression is significantly elevated in human endometrium and endometriosis lesionsInfluence endometriosis invasion (93)

[i] TIMP, tissue inhibitors of metalloproteinase; MT5-MMP, membrane type 5 metalloproteinase; OC, ovarian cancer; EC, endometrial cancer; CC, cervical cancer; BC, breast cancer.

Since the first discovery of stromelysin in endometrial tissues in 1993 by Rodgers et al (94) and the confirmation of MMP properties and pathological functions, the centrality of MMPs in the regulation of tissue remodeling and invasion in EC has been repeatedly substantiated. It has been demonstrated that L-33 enhances the invasive capacity of human ovarian endometriotic stromal cells by activating MMP-9 expression (92). Furthermore, miRNA-34a-5p exerts an inhibitory effect on the transcriptional activity of the MMP-2 gene by directly binding to its 3'-untranslated region, thereby significantly curtailing the invasive migratory capacity and stemness maintenance function of ESCs (23).

MMP-9 significantly enhances the invasive ability of endometriotic cells through degradation of basement membrane type IV collagen and activation of precursor growth factors, which are involved in the pathologic process of EMS. Tissue inhibitors of metalloproteinases (TIMPs), as endogenous regulatory proteins, can maintain the dynamic balance of degradation and synthesis of ECM through specific binding to the active site of MMPs (85). This suggests that targeted modulation of MMP-9 expression or enzymatic activity may be a novel therapeutic strategy to inhibit cell migration and invasion. However, the translational application of TIMPs is severely limited due to their short half-life in vivo and systemic toxicity.

Imbalance of the MMPs/TIMPs ratio is not only a central feature of invasive metastasis in EMS, but also exists in benign diseases such as malignant tumors. Epidemiologic studies have shown that patients with EMS have an elevated risk of developing OC compared with the normal population (95,96). OC is a lethal gynecologic malignancy, and has a unique pattern of transcavitary metastasis, resulting in extensive implantation of cancer cells on the peritoneal and omental surfaces (97). EMS and OC show remarkable similarity in cell migration invasion mechanisms, and MMP14 (MT1-MMP), MMP2, and MMP9 can be activated through the synergistic degradation of ECM components such as laminin/fibronectin, activation of EMT, and upregulation of related transcription factors, which together drive the malignant dissemination of OC cells (97).

Clinicopathological analyses have shown that the expression levels of MMPs are generally upregulated in OC tissues compared with normal ovarian tissues (98). Conversely, inhibition of MMP activity reduces the progression of cell invasion in vitro and metastasis in vivo (99). Based on this, researchers have developed machine learning models integrating markers such as MMP-2/-3/-11/-26, HE4 and CA125, which have an area under the receiver operating characteristic curve of 0.97 for the diagnosis of OC (100).

Mechanistic studies have revealed that enhancing zeste homolog 2 (EZH2), a histone lysine N-methyltransferase, could enhance OC cell invasiveness by inhibiting TIMP2 expression to reduce and promote MMP9 transcription. The covalent EZH2 inhibitor SKLB-03220 reverses this effect and restores the TIMP2/MMP9 ratio to physiological levels (101). In addition, the invasive and metastatic process of BC, a highly prevalent malignancy in women, is also significantly dependent on MMP2/MMP9-mediated ECM remodeling (102).

High expression of MMP-2, together with low expression of TIMP2, suggests an increased risk of local and distant metastasis in EC, and both molecules have the potential to be key markers for human gynecological cancer cell lines (98). Furthermore, MMP9 may be a favorable marker for the adjuvant diagnosis of early EC (103).

In the pathological process of CC, MMP-2 drives malignant progression through a dual mechanism, directly degrading type IV collagen to disrupt basement membrane integrity; and regulating the MMP cascade and the dynamic balance of ECM components (cytokines, chemokines and growth factor receptors) (104). Shukla et al (104) compared patients with CC with healthy individuals and found that elevated serum MMP-2 levels were positively correlated with higher CC stage, and MMP-2 expression increased progressively with the grade of cervical intraepithelial neoplasia, peaking at the stage of CC. In a CC-related study, Schröpfer et al (98) demonstrated that CC cells (HeLa/Caski/SiHa) showed heterogeneity in MMP expression profiles by multicellular lineage analysis, with MMP-1/-11/-13/-15/-17/-24/-28 being commonly expressed in all three lines. Additionally, high expression of MMP-9 was associated with poor prognosis in CC, leading to a study conducted to inhibit the proliferation of human CC cells by reducing the expression of MMP-9(105).

TIMPs and MMPs co-regulate the ECM remodeling process and are closely related to tumor growth, invasion and metastasis. They play a key role in regulating the activities of various enzymes in the ECM. The regulatory network of MMPs has universal biological significance in malignant invasive phenotypes, and TIMPs, as endogenous inhibitors of MMPs, are involved in the entire process of tumor growth and metastasis regulation by modulating the ECM enzyme activity network. MMP-9 overexpression in the TME can promote ECM remodeling and metastatic foci formation through degradation of basement membrane proteoglycan and activation of integrin signaling (106). As shown in Fig. 3, the interaction between MMPs and EMT is highly conserved in EMS and gynecological malignancies, and its synergistic regulatory network provides a molecular basis for the development of cross-disease therapeutic strategies.

RNAs associated with EMS cell migration and invasion. RNA molecules play a key role in the regulation of cellular functions, including miRNAs and long non-coding RNAs (lncRNAs). These RNA molecules regulate gene expression through a variety of molecular mechanisms, which affect cellular physiological and pathological processes. Specific RNA molecules are involved in the regulation of cell migration and invasion in EMS, as shown in Table III (31,89,107-114).

Table III

Differentially expressed RNAs associated with cell migration and invasion in endometriosis.

Table III

Differentially expressed RNAs associated with cell migration and invasion in endometriosis.

First author/s, yearNameMechanismExperimental modelRole(Refs.)
Lv et al, 2015miR-93Inhibits the proliferation, migration and invasionESCsTherapeutic target(89)
Frisendahl et al, 2024miR-193b-5p and miR-374b-5pInhibits migrationThe human immortalized epithelial endometriotic cell line (12Z) and the endometrial stromal cell line (HESC)Explain the pathogenesis(31)
Wan et al, 2022hsa-miR-340-5pRestrains migration, invasiveness and EMTESCsTherapeutic target(107)
Zhang et al, 2022miR-30cDiminishes the invasion and migrationEndometrial epithelial cellsAttenuating the tumor-like behaviors(108)
Ntzeros et al, 2025miR-1229-5pMiR-1229-5p overexpression promotes migration, invasion, and inhibits apoptosisESCs and Ishikawa cellsTherapeutic target(109)
Hagh et al, 2024miR-2233p and miR-200aLow expressionSecretory phase endometriumDiagnostic biomarkers(110)
Wang et al, 2025miR-375-3pSuppresses cell proliferation, migration, and invasionEndometriosis cell line, hEM15ABiomarker for EMs(111)
Zhang et al, 2024miR-145Estrogen receptor α mediates stromal cell migrationStromal cellTherapeutic target(112)
Cui et al, 2022DHRS4Inhibits EC-ESC proliferation, migration and invasion, but promotes apoptosisEctopic ESCsTherapeutic target(113)
Bao et al, 2022HOTAIRInhibits the invasion and migration abilityESCsMechanism study(114)

[i] ESCs, endometriosis stromal cells; miR, microRNA.

miR-93 is a miRNA that belongs to the class of non-coding RNAs. By integrating phenotypic analysis of clinical EMS samples, in vitro cellular experiments, and data from previous studies, miR-93 downregulation has been shown to promote aberrant proliferation, migration, and invasion of ESCs, suggesting a central role in the development and prognosis of EMS (89). A variety of miRNAs may serve as potential diagnostic markers and therapeutic targets for EMS. miR-193b-5p and miR-374b-5p show aberrant expression in the disease state and inhibited endometrial cell migration in an in vitro model (31).

miR-340-5p significantly reduces ESC proliferation and invasion by targeting MAP3K2 and inhibiting the MAPK/ERK signaling pathway, reducing the migratory ability, invasiveness, and EMT process of ESCs (107). However, the miR-93 regulatory mechanism identified by Lv et al (89) has not yet reached clinical translation. A recent study (108) demonstrated the potential of miR-30c to inhibit the invasive migration of EMS cells. miR-1229-5p shows disease severity-related upregulation of expression in ectopic ovarian endothelium, which promotes migratory invasion and inhibits apoptosis of ESCs and Ishikawa cells by targeting STMN1(115). Additionally, a clinicopathologic study targeting the epithelial marker E-CADHERIN and the EMT regulator miR-200b have revealed specific gene expression patterns in the in situ endometrium of ovarian EMS (109).

EMT is a process by which epithelial cells acquire mesenchymal properties that contribute to endometriotic cell migration and invasion. By contrast, miR-200a and miR-223-3p expression are significantly reduced in secretory phase endometriotic lesions (110). MiR-375-3p predicts disease severity by targeting NOX4, and its overexpression effectively inhibits cell proliferation, migration and invasion (111). Mechanistic studies further demonstrated that miR-182 inactivates the NF-κB pathway by directly targeting RelA, thereby inhibiting ESC proliferation, migration, invasion and the EMT process, providing a theoretical basis for the development of therapeutic strategies based on the NF-κB pathway (20). Estrogen receptor α-mediated upregulation of miR-145 promotes stromal cell migration by inhibiting the downstream target gene CITED2(112). These findings systematically reveal the multidimensional regulatory network of miRNAs in EMS cell migration and invasion, but the precise molecular mechanisms still need to be fully analyzed.

lncRNAs are non-protein transcripts that regulate various physiological responses associated with human diseases. Their gene regulatory functions have received increasing attention, but they are still understudied in the field of EMS (116). Two landmark studies have demonstrated that DHRS4-AS1 inhibits cell proliferation and migration and induces apoptosis by regulating miR-139-5p to inhibit cell proliferation and migration and induce apoptosis (113). HOX antisense intergenic RNA regulates cell invasion and migration through the miR-519b-3p/PRRG4 axis (114). Nevertheless, the overall framework and molecular mechanisms of the role of lncRNAs in EMS remain largely unexplored.

Actins associated with cell invasion and migration. A recent review systematically summarized 13 actins and their binding proteins associated with the pathological process of EMS (115). Among these, proteins such as Talin (TLN) and Tensin (TNS) play key roles in the regulation of cell migration, adhesion and invasion (117). Talin-1 promotes the invasion, migration and adhesion of a wide range of malignant tumor cells. However, the precise functional characteristics of this receptor in the context of EMS remain to be fully elucidated. Dysregulation of Talin-1 expression results in a substantial downregulation of N-cadherin, MMP-2, and integrin β3 expression, accompanied by an increase in E-cadherin levels. Talin-1 exhibits abnormally elevated expression levels in both ectopic and ectopic endometrial tissues of patients with EMS when compared with in situ endometrial tissues from healthy patients. Its targeted knockdown exhibits a substantial inhibitory effect on the adherence, invasion and migration capabilities of ESCs (24). A series of studies (118-120) have revealed that TLN regulates the inflammatory microenvironment through integrin-mediated T-cell signaling cascade responses, which in turn inhibit the adhesion and migration ability of endometrial cells.

Tensin-1 (TNS1) is an actin cytoskeletal interplay protein that plays a regulatory role in various biological processes, including cell adhesion, migration, proliferation and differentiation. A clinical study (121) analyzed endometrial tissue and serum samples from patients in untreated and GnRH-a-treated groups, and found that the mRNA and protein expression levels of TNS1 in ectopic endometrial tissues were significantly reduced after GnRH-a intervention. Immunohistochemical results demonstrated that TNS1 exhibited strong positive expression in the epithelial and stromal cells of ectopic tissues in the untreated group. However, treatment significantly weakened the intensity of its expression, and the serum TNS1 concentration decreased by 53%. This finding confirms that TNS1 can be used as a dynamic marker for monitoring the response to GnRH-a treatment and highlights its potential as a molecular target for therapeutic intervention in EMS.

Invasion and migration-related integrins. EECs from patients with EMS exhibit a distinctly differentiated integrin expression profile compared with normal endometrial tissue. Integrin αvβ3 demonstrates significant overexpression in ectopic endometrial tissues, exhibiting considerably higher expression levels compared with normal endometrial tissues. This characteristic may contribute to the enhancement of adhesion, migration and invasion capabilities in ectopic cells (122). A positive correlation has also been shown between integrin expression levels and disease severity, as well as heterogeneous expression patterns in different endometrial cell subpopulations (123).

Integrin isoforms manifest distinctive expression characteristics in EMS. The co-overexpression of integrin αvβ3 and integrin β1 is pronounced in ectopic endometrial cells. Integrin αvβ3 plays a pivotal role in the adhesion, anchoring, migration, diffusion and invasion of ectopic cells by regulating the ECM signaling network (124). Conversely, elevated expression of integrin β1 is closely associated with the chronic inflammatory state of the focal microenvironment. This heightened expression has the potential to exacerbate the pathological process by enhancing cell-cell interactions and pro-inflammatory signaling (125).

At the molecular level, estrogen signaling regulates integrin expression levels through epigenetic modifications, suggesting a potential bridging role for the hormone-integrin axis in the development of EMS (126). These findings systematically elucidate the biological nature of the disease, as reflected by the heterogeneity of integrin expression. They also highlight the translational potential of integrins as novel therapeutic targets, providing innovative strategies to improve the clinical prognosis of patients through precise intervention of integrin-mediated cell-matrix interaction.

Other relevant small molecules and proteins. Small-molecule inhibitors have important regulatory functions in modulating pathological processes such as cell proliferation, migration, invasion, angiogenesis and metastasis (127). Typical examples are the FoxM1 inhibitor thiostrepton. Treatment with FoxM1 significantly reduces nasopharyngeal carcinoma cell survival and effectively blocks tumor cell proliferation, migration and invasion by inhibiting FoxM1 expression (128).

In the field of EMS research, small-molecule interventional strategies have also demonstrated therapeutic potential. Yu et al (129) demonstrated that the knockdown of Chemokine-Like Receptor 1 (CMKLR1) or the use of its specific antagonist, α-Naphthoyl ethyl-trimethyl-ammonium iodide (α-NETA), significantly inhibited the migration and invasiveness of endometriotic stromal cells. The capacity of stromal cells to migrate and invade in patients with EMS has been demonstrated to be associated with the progression of mesenchymal-epithelial transition (MET). In vivo experiments have substantiated the efficacy of α-NETA in mitigating disease progression by targeting the chemokine/CMKLR1 axis. In a recent study (130), researchers examined the effects of a selective NF-κB inhibitor, namely, dihydroxy-methyl-epoxide quinimicin (DHMEQ), which was optimized using a natural compound design. This inhibitor effectively suppressed the migratory and invasive behavior of human ESCs (HESCs) at non-toxic concentrations. This observation suggests that DHMEQ could serve as a novel therapeutic agent for targeting inflammatory pathways in medical treatments.

Periostin, a member of the ECM protein family, plays a pivotal role in tissue repair, cell adhesion and signaling. Periostin stimulation significantly enhances the migratory invasion and adhesion of EMS cells through the activation of the integrin-linked kinase (ILK) 1/Akt signaling pathway (131). This suggests that it may act as an agonist in the development of EMS (132). However, the clinical translational value of periostin remains to be systematically evaluated due to the lack of in vivo experimental validation. Osteopontin, an integrin-binding glycoprotein, has also been shown to play a role in the adhesion and migration of ectopic foci by regulating cell-matrix interactions (25).

In addition, lipoprotein A4 (LXA4) has been shown to inhibit EMS cell migration and invasion by regulating the activity of autophagy. Li et al (133) found that LXA4 significantly reduced the migratory invasive ability of EMS cells by inhibiting NF-κB signaling-mediated autophagy activity. Furthermore, the reactivation of autophagy induced by rapamycin reversed this effect, thereby revealing a bidirectional regulatory role of autophagy modulation in LXA4 treatment. A previous study (60) demonstrated that the expression of T-cadherin significantly inhibited the invasive and migratory activity of ESCs. Furthermore, the expression level of T-cadherin in ectopic endometrium was significantly lower than that in the ectopic endometrium. These findings suggest that T-cadherin may be a novel therapeutic target for EMS.

Estrogen. The pathological features of EMS, a disease dependent on estrogen, are closely related to the regulation of the menstrual cycle. As demonstrated in Fig. 5, the proliferative, secretory and regressive changes of the normal endometrium are synergistically regulated by estrogen and progesterone. The two secretory peaks of estrogen (especially the secretory phase) can significantly drive the abnormal proliferation of the in-situ endometrium. Ectopic foci exhibit a high degree of dependence on estrogen signaling during the processes of migration, invasion and adaptation to the new microenvironment.

There is also a positive correlation between growth activity and estrogen levels. The action of estrogen is facilitated by the estrogen receptors, Erα and Erβ (134). Estrogen can significantly reduce the migratory invasive ability of cells by inhibiting ERβ expression (22). Subsequent experiments corroborated the finding that estradiol induces EMT in EECs and promotes disease progression through activation of the β-catenin/Snail signaling axis. Reverse genetics experiments have demonstrated that β-catenin knockdown completely blocks the estrogen-induced Snail-dependent EMT process (26). In addition, estrogen upregulates nicotinamide N-methyltransferase expression in ESCs in a dose-dependent manner, which regulates ESC proliferation and enhances migratory invasiveness (19).

Wakatsuki et al (135) determined that the estrogen receptor modulator estetrol inhibits disease progression through downregulation of migration-related signaling pathways. ERβ-mediated pathological processes may engage in molecular interactions with the innate immune anti-inflammatory effects regulated by NLRC5(136). In light of these mechanisms, contemporary therapeutic strategies are oriented toward the systematic reduction of circulating estrogen levels or the localized targeting thereof.

3. Drug studies on inhibitors of EMS cell migration and invasion

The current state of the field has resulted in the identification of various inhibitors of cell migration and invasion. These inhibitors can be categorized into several groups, including synthetic drugs and drug analogs, natural extracts, small-molecule complex inhibitors, Chinese medicine and targeted drugs. Among these inhibitors, synthetic drugs and drug analogs refer to the adjuvant therapeutic aspects that are widely used in clinical practice for the treatment of invasive diseases. Therefore, their safety, efficacy, side effects and pharmacological therapies have been demonstrated. However, the targets of action of this class of drugs are complex and diverse, and the mechanisms are more complex and cannot be explained comprehensively, especially in the treatment of cancer (137). As illustrated in Fig. 6, the mechanism of action of various pharmaceutical agents and their respective targets is concisely delineated.

Synthetic drugs, drug analogs and natural plant extracts

Natural plant extracts have become an important choice for the treatment of gynecological diseases due to their multicomponent properties and low side effect advantages. However, their complex chemical composition and mechanism of action have led to significant heterogeneity in clinical applications. More than 40 formulations and 20 compounds and monomers have been used for gynecological disease interventions, mainly targeting pathological aspects such as apoptosis, invasion, migration, oxidative stress and immunomodulation (138).

Synthetic compounds. Imatinib is a tyrosine kinase inhibitor that has been approved for the clinical treatment of two distinct conditions: chronic granulocytic leukemia and gastrointestinal mesenchymal stromal tumor. This pharmaceutical agent exhibited a substantial inhibitory effect on the proliferation of lesions and the capacity of human embryonic stem cells to invade in a three-dimensional type I collagen matrix in a mouse model of experimental EMS (139). This effect was achieved by impeding the interaction of colony-stimulating factor-1 with its receptor c-FMS. A comprehensive review of the available clinical data set (140) confirmed the antitumor efficacy of Imatinib, and in vitro experimentation validated the inhibitory effect on cell migration and invasion.

Pyrrolidine dithiocarbamate (PDTC) functions as a sulfhydryl-modified NF-κB inhibitor, exhibiting both antioxidant and anti-inflammatory properties (141,142). PDTC possesses the capability to induce apoptosis and to significantly inhibit adhesion, migration and invasive activity of endometriotic cells through a multi-targeted modulation process (143). This finding serves to underscore the therapeutic potential of PDTC.

β-Sitosterol, a tetracyclic triterpenoid phytosterol, is found in a variety of vegetable oils, nuts and medicinal plants (144,145). This compound exerts anticancer effects by inhibiting the proliferation and migration of malignant cells, inducing apoptosis, and interfering with metabolic pathways. A recent study expanded the application of this technique and confirmed its effectiveness in inhibiting the migration and proliferation of endometrial cells (146).

Kaempferol, a flavonoid, has garnered attention for its antioxidant, anti-inflammatory and anticancer properties (147-149). Network pharmacological predictions and in vitro validation suggest that the anti-EMS mechanism may involve the regulation of the PI3K pathway (150). This pathway inhibits cell migration and invasion by affecting the expression of phosphatase and tensin homolog (PTEN) and MMP-9.

Chamomile lactone, a sesquiterpene lactone, has been utilized in a variety of in vivo and ex vivo experiments, and studies have demonstrated its favorable anti-inflammatory, analgesic and anticancer properties (151-155). In addition to its inhibitory effect on the proliferation and migration of CC cells (156), a recent study confirmed its ability to reverse the EMT process in immortalized epithelial endometriotic cell line 12Z and to inhibit the invasive migratory properties of endometrial tissue implants (157).

Metformin, a well-established hypoglycemic agent, has recently emerged as a potential regulator of reproductive function and a treatment for EMS (158,159). Metformin enhances fertility by improving endometrial tolerance; however, its clinical use must be critically evaluated due to its glucose-lowering properties, which may trigger the risk of hypoglycemia in patients with normal glucose levels.

Plant extracts. Emodin (1,3,8-trihydroxy-6-methylanthraquinone), a hydroxyanthraquinone natural product, is widely found in a variety of medicinal plants (160). Emodin possesses immunosuppressive, antibacterial, anti-inflammatory and antitumor activities (161,162). However, its application in the treatment of EMS has not been fully exploited due to its physicochemical properties, such as low polarity and bioavailability.

A previous study demonstrated that rhodopsin can inhibit the migration and invasion of HESCs by targeting ILK to induce MET (163). Rhodopsin also exhibits significant antineoplastic properties, particularly against cancerous cells, while demonstrating minimal toxicity toward normal cells (164). Consequently, rhodopsin is currently regarded as a promising pharmaceutical agent with the potential to impede cell migration and invasion. Nevertheless, further refinement is necessary for its application in a clinical setting, including the optimal dosage, the ideal timing of administration, and the precise target of action.

Ginsenoside Rg3, a tetracyclic triterpenoid active ingredient of ginseng, possesses a variety of pharmacological properties, including antitumor, immunomodulatory and antiangiogenic properties (165-167). The multifunctional liposomes of Rg3 developed by Zhu et al (168) significantly enhanced the inhibition rate of paclitaxel-resistant cancer cells, providing an innovative direction in the treatment of drug-resistant tumors. In the context of EMS research, the administration of Rg3 has been observed to markedly diminish the fibrosis and migration potential of HESCs, a phenomenon that may be associated with the regulation of microRNA-27b-3p (169). However, despite the significant antitumor effects of ginsenoside Rg3, its clinical application is still subject to some limitations. For instance, the specific molecular targets of this agent have not been thoroughly investigated, which may impede its widespread clinical application.

Berberine (BBR), a quaternary alkaloid, is found in a variety of medicinal plants, including Rhizoma Coptidis, and has been shown to possess anti-inflammatory, anticancer and metabolic regulatory properties (170-175). BBR exerts its inhibitory effects on the proliferation, invasion and migration phenotypes of HESCs by decreasing the expression of miRNA-429. However, the overexpression of miR-429 has been shown to reverse these effects (176,177).

In vitro studies have demonstrated that β-elemene, a constituent of turmeric extract, exhibits significant inhibitory effects on the proliferation and migration of endometriotic cells (178). Taraxerol has also been shown to induce apoptosis and to inhibit ectopic cell proliferation and migration by blocking the PI3K/Akt/mTOR signaling pathway (179). Conversely, amygdalin has been shown to impede HESC proliferation, migration and invasion by modulating the Wnt/β-catenin signaling axis (35). These natural extracts are predominantly in the experimental research stage, and their clinical translation necessitates substantial sample data and systematic pharmacodynamic validation, as evidenced in Table IV (35,139,143,146,150,157,163,169,178,179).

Molecular inhibitors and melatonin

The role of interleukin-1β (IL-1β) in EMS is primarily characterized by its promotion of EESC proliferation, migration and invasion. IL-1β may act as a positive regulator affecting the progression of EMS, acting through multiple mechanisms. For instance, a group study (180) found that LXA4, as revealed by in vitro experiments, may inhibit the progression of EMS cell migration and invasion in part by decreasing or increasing the action of IL-1β. A previous study reported the use of a small-molecule antagonist of the Tcf/β-catenin complex, which inhibited MMP-9 activity in endometriotic epithelial and stromal cells, thereby suppressing cell proliferation, migration and invasion (181). Additionally, a robust correlation has been observed between cell migration and invasion and MMP-9. Consequently, a small-molecule antagonist of the Tcf/β-catenin complex may impede migration and invasion by hindering the Wnt/β-catenin pathway. However, the potential utilization of this approach in the management of EMS remains to be elucidated through further investigation.

Fibrinogen α-chain (FGA) plays a role in EMS and is a cell adhesion molecule containing two arginine-glycyl-aspartate (RGD) sequences that bind to integrins. A strong correlation between elevated levels of FGA and tumor progression and metastasis has been demonstrated. For instance, FGA has been identified as a promising biomarker for BC (182). Furthermore, the migration and invasion of the endometrial stromal cell line hEM15A can be substantially hindered by the inhibition of FGA (183). A team of researchers conducted a study to analyze the effect of FGA on the biological behavior of EuESCs (184). The findings indicated that the knockdown of FGA led to a suppression in the migratory and invasive capabilities of EuESCs. Additionally, the study revealed alterations in the distribution and morphology of the cytoskeletal filaments, which represents a novel observation that suggests the potential for FGA inhibitors to be utilized as therapeutic agents for the treatment of EMS.

Melatonin, an amine hormone secreted by the pineal gland of the brain, is chemically known as N-acetyl-5-methoxytryptamine and belongs to the indole heterocyclic class of compounds, which is a small-molecule neuroendocrine hormone. Melatonin has an inhibitory effect on tumor cells and complements conventional cancer treatment modalities. Melatonin also significantly reduces tumor load in OC by inhibiting the invasion and migration of OC stem cells (185-187). In addition, in 2018, Qi et al (188) discovered that melatonin could impede the migration, invasion and EMT of normal and endometriotic EECs. Previous studies have identified the potential anticancer activity of melatonin, as well as its ability to inhibit tumor cell growth (189), invasiveness (190) and angiogenesis (191). This finding warrants further investigation through internal and external experiments to verify its clinical relevance. The exploration of the anticancer properties of melatonin represents a novel and promising research direction that merits continued attention (192).

Traditional Chinese medicines (TCMs)

With the advent of molecular biology and network pharmacology, the therapeutic strategies of TCM for EMS have been the subject of new insights and advances. Juan-tong-yin induces endoplasmic reticulum stress through activation of the unfolded protein response, which has been demonstrated to significantly enhance the level of autophagy in ESCs (34). This process has been linked not only to endoplasmic reticulum stress-mediated apoptotic mechanisms, but also to the effective promotion of programmed endometrial cell death in patients with EMS. Furthermore, it has been observed to significantly inhibit the migratory invasive ability of ESCs. Autophagy, a core mechanism of cellular homeostasis regulation, has the potential to remodel the intracellular environment through the Juan-Tong-Yin-mediated lysosomal degradation pathway. This may intervene in the pathological process of EMs.

The c-Jun N-terminal kinase (JNK) signaling pathway is a key signaling pathway in the stress response, whose activation promotes apoptosis. In the context of stress signaling regulation, the Jiangpingni formula has been demonstrated to induce ectopic ESC apoptosis and inhibit their abnormal proliferation by specifically activating the JNK signaling pathway. This provides a novel paradigm for therapies targeting the cell death pathway (193).

Conversely, the Yushén Huǒyào Formula has been shown through multi-omics analysis to modulate the tumor necrosis factor (TNF) signaling pathway, thereby significantly improving the inflammatory microenvironment of EMS lesions (194). A study found that Luoshi Neiyi prescription is capable of inhibiting the adhesion, migration and invasion of ESCs (71). This effect was significantly superior to that of danazol. This conclusion was reached through the implementation of network and serum pharmacology combined with in vitro and ex vivo experiments. A recent study also revealed that Hua Yu Xiao Zheng Decoction exerts antioxidant and anti-aging effects by inhibiting the PI3K/Akt signaling pathway (195). Concurrently, it effectively blocks the proliferation and migration activity of ectopic ESCs

Chinese herbs such as Juan-Tong-Yin, Jiang Ping Ni formula and Yu Shen Huo Yao formula affect the proliferation, migration and invasion of endometriotic cells through different mechanisms, including enhancement of autophagy, activation of JNK signaling pathway to induce apoptosis, as well as modulation of TNF signaling pathway and inhibition of hypoxia-inducible factors. The studies thereof suggest novel approaches and strategies for the treatment of EMS and demonstrate the therapeutic potential of TCM.

Relevant therapeutic targets

To improve the prognosis for early screening and diagnosis of EMS, the discovery of more specific predictive targets is essential. A number of specific targets have been identified through analysis of clinical samples or validation through in vivo and in vitro experiments. Significant gene expression changes and cytokine alterations present in the endometrium of patients with EMS, which may enhance the formation of EMS lesions and provide new therapeutic targets, are shown in Table V (33,136,196-203). Drugs aimed at these targets may have therapeutic benefits, but further clinical validation is needed.

Table V

Targets associated with endometriosis cell migration and invasion.

Table V

Targets associated with endometriosis cell migration and invasion.

First author/s, yearTargetDefinitionStudy methodologyImpactLimitationsRole(Refs.)
Miller et al, 2013ADAMsThe principal enzymes for shedding RTK ectodomainsIn vitro cell experimental proteomicsReduces cell migrationSmall sample size and lack of long-term prognosis studiesFuture use as therapeutic target(196)
Shi et al, 2024KLF6A transcription factorIn vitro two-cell experiment validation tInhibits the proliferation, migration and angiogenesisLack of in vivo experimental validationTargeted therapy for EMS(33)
Zanon et al, 2024Ang IICardiovascular active peptideIn vivo and in vitro experimental validationRegulates hESC proliferation and migration through tPA-uPA/uPAR pathway activationSmall clinical samplesTreatment(197)
Chen et al, 2023AGPAT4Integral to the AGPAT familyMulti-omics study of clinical tissue samplesThe siRNA-mediated silencing of AGPAT4 resulted in a marked inhibition of interstitial cell proliferation, migration and invasionNo further molecular interactions and functional roles for AGPAT4Potential for biomarkers and therapeutic targets(198)
Zheng et al, 2023HIF1ANHypoxia-inducible factor 1 subunit alpha inhibitorIn vivo and in vitro experimental validationmiR-429 targets HIF1AN in HESCsInvestigations are needed to clarify the role of the miR-429/HIF1AN axis in EM in the futureTreatment(199)
Zhang et al, 2025FZD7Transmembrane receptor proteinEndometrial stromal cells (ESCs) and human ectopic endometrial stromal cell line hEM15ARegulates endometriotic cell proliferation, invasion, migration, and angiogenesis via ferroptosisLack of in vivo experimental validationTreatment(200)
Lv et al, 2025METTL14RNA methyltransferasehuman ESC (HEM15A)Inhibits he activation of the MEK/ERK pathway and ESC proliferation, migration, and invasionVarious drugs and plasmids were used to treat the cells.Treatment(201)
Tang et al, 2024RNF43Ubiquitin ligasePrimary ectopic ESCsNegatively affects viability and migration of ESCsNo further molecular interactions and functional roles for RNF43Treatment(202)
Vu et al, 2024APOBEC3BDNA editing12Z immortalized human endometriotic cell lineInhibits cell proliferation, invasion and migrationLack of clinical researchTherapeutic target(203)
Guo et al, 2024NLRC5NOD-like receptors familyHuman ESCsInhibits migrationSmall clinical samplesTreatment(136)

[i] ESCs, endometrial stromal cells; ADAMS, A Disintegrin and Metalloproteinases; RTK, receptor tyrosine kinase; KLF6, Krüppel-like Factor 6; Ang II, angiotensin II; FZD7, Frizzled-7; METTL14, methyltransferase-like 14; RNF43, RNF43, ring finger protein 43; APOBEC3B, Apolipoprotein-B mRNA-editing complex 3B; NLRC5, NOD-like receptors (NLRs) family CARD domain-containing 5.

4. Clinical applications

There are few clinical applications for drugs that inhibit EMS cell migration and invasion. Currently, the most commonly used drugs are hormonal therapies, including oral contraceptives (estrogen-progestin preparations), progestin preparations (containing progestin derivatives), androgens (danazol) and GnRH agonists and antagonists (204). Oral contraceptives are still not as effective as expected in improving pain (205). Progestogens, although cheaper than other drugs, have side effects such as progressive uterine bleeding, weight gain, breast discomfort, headache, nausea and mood changes (206). Furthermore, progesterone derivatives usually have longer treatment cycles (207).

Danazol, a derivative of the hormone testosterone, is also used to treat EMS by reducing pituitary secretion of follicle-stimulating hormone and luteinizing hormone by inhibiting GnRH secretion, but there are adverse effects and poor compliance (207,208). Pseudopausal therapy drugs include GnRH-a and danazol, which lower the estrogen levels in patients with EMS. These elicit temporary menopause, so that the ectopic endothelium atrophies and achieves the therapeutic purpose (Fig. 6). Therapy can be achieved by regulating TNS, TNL and integrins to inhibit EMS cell migration and invasion (24,121,209). TNS, TNL and integrins are the main molecules affecting the migration and invasion of EMS cells. Pseudopausal therapy not only controls the disease progression from the level of lowering estrogens but also inhibits the proliferation from the cellular molecular level.

Compounds, natural extracts, molecular inhibitors, TCM and related targets can inhibit migration and invasion in EMS cells, although most studies are in the experimental research phase. Anexelekto (Axl), a member of the receptor tyrosine kinase family Tyro3, Axl and Mertk. Axl is phosphorylated and activated by binding to its natural ligand, with growth arrest activated by phosphorylation through binding to its natural ligand, growth-stopping-specific protein 6 (Gas6). Axl plays an important role in tumor cell growth, migration, invasion and immunosuppression (210-212). Small-molecule tyrosine kinase inhibitors and monoclonal antibodies (mAb) are the main Axl inhibitors, and there have been numerous clinical trials for use thereof in patients with cancer (213). This has not yet been applied to the treatment of EMS. However, the rational and effective use of these antibodies is expected to enhance the therapeutic effect (214).

The study of pathogenesis of EMS and related inhibitors remains a complex area of research. Certain agents that have been used to inhibit cell migration and invasion in other tumors, including flavonoids (215), curcumin (216), rhodopsin (164) and ginsenosides (169), must be tested for toxicologic and pharmacokinetic properties before they can be formally applied in the clinic. Thus, one of the major obstacles to research to translate preclinical findings into clinical practice is the lack of robust in vivo models to accurately summarize the complex pathophysiology of EMS.

Current animal models, such as the mouse xenograft model or the rat autograft model (217), provide valuable insights but often do not fully mimic human disease progression due to differences in immune responses and hormonal regulation. Other studies have identified endometrial-like organs (218) and xenografts between homozygous rats and mice (219). This is critical for the development of more representative in vivo systems to validate the efficacy and safety of potential inhibitors prior to conducting clinical trials.

Another major obstacle lies in optimizing drug delivery to effectively target endometriotic lesions. Systemic delivery of drugs may result in off-target effects and inadequate site-of-action concentrations, especially given the unique microenvironment of endometriotic lesions, for example, hypoxia and ECM remodeling (68,72). Novel delivery systems, for example, a cRGD-modified liposome nanodrug (cRGD-LP-ART), have been synthesized, offering promising solutions by improving drug stability, bioavailability and specificity (220). Similarly, topical administration via intrauterine devices can circumvent systemic exposure and improve outcomes in patients with deep invasive EMS (221).

5. Summary

EMS is a common benign gynecologic disease, but the invasive and metastatic nature thereof leads to complex and limited treatment. Cell migration and invasion are important pathological processes in EMS, involving complex signaling pathways and multiple influencing factors, which not only increase the complexity of targeted therapies, but also increase the difficulty of treatment. In the past, a solution has not been found, and there are still some challenges and limitations. There is also no completely eradicable clinical treatment at present.

According to the present review (Fig. 7), numerous researchers have found that cell migration and invasion can be inhibited by regulating small molecules and other mechanisms that involve the activation and regulation of signaling pathways. Although these treatments have shown some efficacy, they may have adverse effects in terms of reproductive function and embryotoxicity. Therefore, a comprehensive understanding of the signaling networks associated with EMS cell migration and invasion, the integration of multiple pathways and targets, and the development of new plant extracts to inhibit lesions are all therapeutic approaches that will provide new directions for future research.

In conclusion, the rational use of drugs targeting the migration and invasion of endometriotic cells is expected to address the problems of misdiagnosis, delayed diagnosis and reduced fertility in patients with EMS. Follow-up studies need to focus on the following three issues. First, to establish a dynamic balance of efficacy-toxicity evaluation systems and clarify the quantitative relationship between the dose threshold and the threshold of adverse effects in the therapeutic window. Second, to construct a female physiological simulation model integrating neuroendocrine cycle regulation, emotional stress responses, and the gastrointestinal metabolism microenvironment. This model must focus on the dynamic expression of estrogen receptors, the feedback mechanism of the hypothalamus-pituitary axis, the interaction between the 5-HTergic system and hypothalamic-pituitary-adrenal axis, the kinetics of substance transport in intestinal and hepatic axes, and the metabolism of bacterial flora and other key elements. Third, the development of targeted intervention strategies, including specific modulation of signaling pathways based on single-cell multi-omics resolution or the combination of multiple therapeutic approaches to improve therapeutic efficacy. In addition, future research should prioritize improving in vivo models, exploring innovative drug delivery platforms, and fostering interdisciplinary collaborations to accelerate the transition from bench to bedside. By addressing these translational challenges, the relevance of the findings to real-world treatment strategies will be strengthened.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the ‘Trinity’ college students innovation and entrepreneurship cultivation program of Inner Mongolia Medical University (grant no. SWYT2022010), the Nature Science Foundation of Inner Mongolia Autonomous Region (grant nos. 2023LHMS08001 and 2022MS08060), the Unite program of Inner Mongolia Medical University (grant no. YKD2023LH060), the National Natural Science Foundation of China (grant no. 82160703), the Program for Young Talents of Science and Technology in Universities of Inner Mongolia Autonomous Region (grant no. NJYT23114), the Health Science and Technology Program of Inner Mongolia Health Commission (grant nos. 202202158 and 202201337), the General program of Inner Mongolia Medical University (grant no. YKD2022MS045), the Science and Technology Program of the Joint Fund of Scientific Research for the Public Hospitals of Inner Mongolia Academy of Medical Sciences (grant nos. 2024GLLH0371 and 2024GLLH0290).

Availability of data and materials

Not applicable.

Authors' contributions

NW, RZ and ZD designed and wrote the review. HG, YZ, JS, JM, RS and XQ revised and provided comments during all stages of writing the manuscript. All authors contributed for the final version of the manuscript. All authors read and approved the final version of the 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.

References

1 

Taylor HS, Kotlyar AM and Flores VA: Endometriosis is a chronic systemic disease: Clinical challenges and novel innovations. Lancet. 397:839–852. 2021.PubMed/NCBI View Article : Google Scholar

2 

Peiris AN, Chaljub E and Medlock D: Endometriosis. JAMA. 320(2608)2018.PubMed/NCBI View Article : Google Scholar

3 

Greene R, Stratton P, Cleary SD, Ballweg ML and Sinaii N: Diagnostic experience among 4,334 women reporting surgically diagnosed endometriosis. Fertil Steril. 91:32–39. 2009.PubMed/NCBI View Article : Google Scholar

4 

Sanfilippo JS, Wakim NG, Schikler KN and Yussman MA: Endometriosis in association with uterine anomaly. Am J Obstet Gynecol. 154:39–43. 1986.PubMed/NCBI View Article : Google Scholar

5 

Imperiale L, Nisolle M, Noël JC and Fastrez M: Three types of endometriosis: Pathogenesis, diagnosis and treatment. state of the art. J Clin Med. 12(994)2023.PubMed/NCBI View Article : Google Scholar

6 

As-Sanie S, Shafrir AL, Halvorson L, Chawla R, Hughes R and Merz M: The Burden of pelvic pain associated with endometriosis among women in selected European countries and the United States: A restricted systematic review. J Minim Invasive Gynecol. 31:653–666.e5. 2024.PubMed/NCBI View Article : Google Scholar

7 

Katon JG, Plowden TC and Marsh EE: Racial disparities in uterine fibroids and endometriosis: A systematic review and application of social, structural, and political context. Fertil Steril. 119:355–363. 2023.PubMed/NCBI View Article : Google Scholar

8 

Koninckx PR, Ussia A, Adamyan L, Wattiez A, Gomel V and Martin DC: Pathogenesis of endometriosis: The genetic/epigenetic theory. Fertil Steril. 111:327–340. 2019.PubMed/NCBI View Article : Google Scholar

9 

van Haaps AP, Wijbers JV, Schreurs AMF, Vlek S, Tuynman J, De Bie B, de Vogel AL, van Wely M and Mijatovic V: The effect of dietary interventions on pain and quality of life in women diagnosed with endometriosis: A prospective study with control group. Hum Reprod. 38:2433–2446. 2023.PubMed/NCBI View Article : Google Scholar

10 

Burney RO and Giudice LC: Pathogenesis and pathophysiology of endometriosis. Fertil Steril. 98:511–519. 2012.PubMed/NCBI View Article : Google Scholar

11 

Jensen JR and Coddington CC III: Evolving spectrum: The pathogenesis of endometriosis. Clin Obstet Gynecol. 53:379–388. 2010.PubMed/NCBI View Article : Google Scholar

12 

Maignien C, Santulli P, Chouzenoux S, Gonzalez-Foruria I, Marcellin L, Doridot L, Jeljeli M, Grange P, Reis FM, Chapron C and Batteux F: Reduced α-2,6 sialylation regulates cell migration in endometriosis. Hum Reprod. 34:479–490. 2019.PubMed/NCBI View Article : Google Scholar

13 

Hapangama DK, Raju RS, Valentijn AJ, Barraclough D, Hart A, Turner MA, Platt-Higgins A, Barraclough R and Rudland PS: Aberrant expression of metastasis-inducing proteins in ectopic and matched eutopic endometrium of women with endometriosis: Implications for the pathogenesis of endometriosis. Hum Reprod. 27:394–407. 2012.PubMed/NCBI View Article : Google Scholar

14 

Lin LL, Makwana S, Chen M, Wang CM, Gillette LH, Huang TH, Burney RO, Nicholson BJ and Kirma NB: Cellular junction and mesenchymal factors delineate an endometriosis-specific response of endometrial stromal cells to the mesothelium. Mol Cell Endocrinol. 539(111481)2022.PubMed/NCBI View Article : Google Scholar

15 

Chen CW, Chavez JB, Kumar R, Go VA, Pant A, Jain A, Polusani SR, Hart MJ, Robinson RD, Gaczynska M, et al: Hypersensitive intercellular responses of endometrial stromal cells drive invasion in endometriosis. Elife. 13(e94778)2024.PubMed/NCBI View Article : Google Scholar

16 

Wang X, Zheng Q, Sun M, Liu L, Zhang H and Ying W: Signatures of necroptosis-related genes as diagnostic markers of endometriosis and their correlation with immune infiltration. BMC Womens Health. 23(535)2023.PubMed/NCBI View Article : Google Scholar

17 

Dai F, Li J and Liu Y: Phosphatase and tensin homolog deficiency induces M2 macrophage polarization by promoting glycolytic activity in endometrial stromal cells. Biol Reprod. 112:640–650. 2025.PubMed/NCBI View Article : Google Scholar

18 

Cordeiro MR, Carvalhos CA and Figueiredo-Dias M: The emerging role of menstrual-blood-derived stem cells in endometriosis. Biomedicines. 11(39)2022.PubMed/NCBI View Article : Google Scholar

19 

Hou S, Xu H, Lei S and Zhao D: Overexpressed nicotinamide N-methyltransferase in endometrial stromal cells induced by macrophages and estradiol contributes to cell proliferation in endometriosis. Cell Death Discov. 10(463)2024.PubMed/NCBI View Article : Google Scholar

20 

Wu M and Zhang Y: MiR-182 inhibits proliferation, migration, invasion and inflammation of endometrial stromal cells through deactivation of NF-κB signaling pathway in endometriosis. Mol Cell Biochem. 476:1575–1588. 2021.PubMed/NCBI View Article : Google Scholar

21 

Sarsenova M, Stepanjuk A, Saare M, Kasvandik S, Soplepmann P, Mikeltadze I, Götte M, Salumets A and Peters M: Carboxypeptidase inhibitor LXN expression in endometrial tissue is menstrual cycle phase-dependent and is upregulated in endometriotic lesions. Genes (Basel). 15(1086)2024.PubMed/NCBI View Article : Google Scholar

22 

Peng Y, Xiong W, He H, Liu H, Fu T, Long X, Li X, Dai X, Xu Y, Zhang L and Liu Y: Estradiol promotes endometriosis progression via the eRβ/QKI/circSMAD2 Axis. Curr Pharm Biotechnol: Feb 20, 2025 (Epub ahead of print).

23 

Begum Y, Pandit A, Shukla D, Gupta R, DasMahapatra P, Srivastava AK and Swarnakar S: Suppression of endometriosis by miRNA-34a via inhibition of matrix metalloproteinase-2: An alternative pathway to impede invasion. Noncoding RNA Res. 12:92–101. 2025.PubMed/NCBI View Article : Google Scholar

24 

Tang X, Li Q, Li L and Jiang J: Expression of Talin-1 in endometriosis and its possible role in pathogenesis. Reprod Biol Endocrinol. 19(42)2021.PubMed/NCBI View Article : Google Scholar

25 

D'Amico F, Skarmoutsou E, Quaderno G, Malaponte G, La Corte C, Scibilia G, D'Agate G, Scollo P, Fraggetta F, Spandidos DA and Mazzarino MC: Expression and localisation of osteopontin and prominin-1 (CD133) in patients with endometriosis. Int J Mol Med. 31:1011–1016. 2013.PubMed/NCBI View Article : Google Scholar

26 

Xiong W, Zhang L, Liu H, Li N, Du Y, He H, Zhang Z and Liu Y: E(2) -mediated EMT by activation of β-catenin/Snail signalling during the development of ovarian endometriosis. J Cell Mol Med. 23:8035–8045. 2019.PubMed/NCBI View Article : Google Scholar

27 

Sesti F, Pietropolli A, Capozzolo T, Broccoli P, Pierangeli S, Bollea MR and Piccione E: Hormonal suppression treatment or dietary therapy versus placebo in the control of painful symptoms after conservative surgery for endometriosis stage III-IV. A randomized comparative trial. Fertil Steril. 88:1541–1547. 2007.PubMed/NCBI View Article : Google Scholar

28 

Qin Z, Dong Z, Liu J, Zhong A, Bao M, Wang H, Yu H, Zhang S, Zhang W, Shen L, et al: A preliminary study on the effects of black cohosh preparations on bone metabolism of rat models with GnRH-a-induced peri-menopausal symptoms. Front Endocrinol (Lausanne). 13(854345)2022.PubMed/NCBI View Article : Google Scholar

29 

Houshdaran S, Oke AB, Fung JC, Vo KC, Nezhat C and Giudice LC: Steroid hormones regulate genome-wide epigenetic programming and gene transcription in human endometrial cells with marked aberrancies in endometriosis. PLoS Genet. 16(e1008601)2020.PubMed/NCBI View Article : Google Scholar

30 

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.PubMed/NCBI View Article : Google Scholar

31 

Frisendahl C, Tang Y, Boggavarapu NR, Peters M, Lalitkumar PG, Piltonen TT, Arffman RK, Salumets A, Götte M, Korsching E and Gemzell-Danielsson K: miR-193b-5p and miR-374b-5p are aberrantly expressed in endometriosis and suppress endometrial cell migration in vitro. Biomolecules. 14(1400)2024.PubMed/NCBI View Article : Google Scholar

32 

Rodríguez E, Aburjania N, Priedigkeit NM, DiFeo A and Martignetti JA: Nucleo-cytoplasmic localization domains regulate Krüppel-like factor 6 (KLF6) protein stability and tumor suppressor function. PLoS One. 5(e12639)2010.PubMed/NCBI View Article : Google Scholar

33 

Shi J, Jing W, He Y and Huang Y: Decreased expression of KLF6 in ectopic endometrial stromal cells contributes to endometriosis progression by targeting CTNNB1. Cell Signal. 120(111230)2024.PubMed/NCBI View Article : Google Scholar

34 

Meng F, Li J, Dong K, Bai R, Liu Q, Lu S, Liu Y, Wu D, Jiang C and Li W: Juan-tong-yin potentially impacts endometriosis pathophysiology by enhancing autophagy of endometrial stromal cells via unfolded protein reaction-triggered endoplasmic reticulum stress. J Ethnopharmacol. 325(117859)2024.PubMed/NCBI View Article : Google Scholar

35 

Yu M, Yang L, Pei Y and Xu M: Amygdalin inhibits endometrial stromal cell proliferation, migration, and invasion in endometriosis mice via inhibiting Wnt/β-catenin signaling. J Mol Histol. 56(11)2024.PubMed/NCBI View Article : Google Scholar

36 

Kao AP, Wang KH, Chang CC, Lee JN, Long CY, Chen HS, Tsai CF, Hsieh TH and Tsai EM: Comparative study of human eutopic and ectopic endometrial mesenchymal stem cells and the development of an in vivo endometriotic invasion model. Fertil Steril. 95:1308–1315.e1. 2011.PubMed/NCBI View Article : Google Scholar

37 

Mao Z, Sang MM, Chen C, Zhu WT, Gong YS and Pei DS: CSN6 promotes the migration and invasion of cervical cancer cells by inhibiting autophagic degradation of cathepsin L. Int J Biol Sci. 15:1310–1324. 2019.PubMed/NCBI View Article : Google Scholar

38 

Gao S, Bian T, Su M, Liu Y and Zhang Y: miR-26a inhibits ovarian cancer cell proliferation, migration and invasion by targeting TCF12. Oncol Rep. 43:368–374. 2020.PubMed/NCBI View Article : Google Scholar

39 

Geng A, Luo L, Ren F, Zhang L, Zhou H and Gao X: miR-29a-3p inhibits endometrial cancer cell proliferation, migration and invasion by targeting VEGFA/CD C42/PAK1. BMC Cancer. 21(843)2021.PubMed/NCBI View Article : Google Scholar

40 

Cong Y, Cui Y, Zhu S, Cao J, Zou H, Martin TA, Qiao G, Jiang W and Yu Z: Tim-3 promotes cell aggressiveness and paclitaxel resistance through NF-κB/STAT3 signalling pathway in breast cancer cells. Chin J Cancer Res. 32:564–579. 2020.PubMed/NCBI View Article : Google Scholar

41 

Altayyeb A, Othman E, Khashbah M, Esmaeel A, El-Mokhtar M, Lambalk C, Mijatovic V and Abdelgawad M: Characterization of mechanical signature of eutopic endometrial stromal cells of endometriosis patients. Reprod Sci. 27:364–374. 2020.PubMed/NCBI View Article : Google Scholar

42 

Maurya VK, Szwarc MM, Fernandez-Valdivia R, Lonard DM, Yong S, Joshi N, Fazleabas AT and Lydon JP: Early growth response 1 transcription factor is essential for the pathogenic properties of human endometriotic epithelial cells. Reproduction. 164:41–54. 2022.PubMed/NCBI View Article : Google Scholar

43 

Chen SM, Liu YK, Ma XQ, Wei CY, Li MQ and Zhu XY: Creatine promotes endometriosis progression by inducing M2 polarization of peritoneal macrophages. Reproduction. 169(e240278)2025.PubMed/NCBI View Article : Google Scholar

44 

Bacci M, Capobianco A, Monno A, Cottone L, Di Puppo F, Camisa B, Mariani M, Brignole C, Ponzoni M, Ferrari S, et al: Macrophages are alternatively activated in patients with endometriosis and required for growth and vascularization of lesions in a mouse model of disease. Am J Pathol. 175:547–556. 2009.PubMed/NCBI View Article : Google Scholar

45 

Yu JJ, Sun HT, Zhang ZF, Shi RX, Liu LB, Shang WQ, Wei CY, Chang KK, Shao J, Wang MY and Li MQ: IL15 promotes growth and invasion of endometrial stromal cells and inhibits killing activity of NK cells in endometriosis. Reproduction. 152:151–160. 2016.PubMed/NCBI View Article : Google Scholar

46 

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

47 

Makoui MH, Fekri S, Makoui RH, Ansari N and Esmaeilzadeh A: The role of mast cells in the development and advancement of endometriosis. Am J Reprod Immunol. 93(e70019)2025.PubMed/NCBI View Article : Google Scholar

48 

McCallion A, Nasirzadeh Y, Lingegowda H, Miller JE, Khalaj K, Ahn S, Monsanto SP, Bidarimath M, Sisnett DJ, Craig AW, et al: Estrogen mediates inflammatory role of mast cells in endometriosis pathophysiology. Front Immunol. 13(961599)2022.PubMed/NCBI View Article : Google Scholar

49 

Tian J, Hoffmann V, Ibrahim MG, Hansen U, Schüring AN, Velho RV, Mechsner S and Götte M: Characterization of E-Cadherin, SSEA-1, MSI-1, and SOX-2 expression and their association with pale cells in adenomyosis. Biomolecules. 14(1355)2024.PubMed/NCBI View Article : Google Scholar

50 

Guo J, Gao J, Yu X, Luo H, Xiong X and Huang O: Expression of DJ-1 and mTOR in eutopic and ectopic endometria of patients with endometriosis and adenomyosis. Gynecol Obstet Invest. 79:195–200. 2015.PubMed/NCBI View Article : Google Scholar

51 

Gentilini D, Busacca M, Di Francesco S, Vignali M, Viganò P and Di Blasio AM: PI3K/Akt and ERK1/2 signalling pathways are involved in endometrial cell migration induced by 17beta-estradiol and growth factors. Mol Hum Reprod. 13:317–322. 2007.PubMed/NCBI View Article : Google Scholar

52 

Guzeloglu Kayisli O, Kayisli UA, Luleci G and Arici A: In vivo and in vitro regulation of Akt activation in human endometrial cells is estrogen dependent. Biol Reprod. 71:714–721. 2004.PubMed/NCBI View Article : Google Scholar

53 

Zheng T and Yang J: Differential expression of EWI-2 in endometriosis, its functional role and underlying molecular mechanisms. J Obstet Gynaecol Res. 43:1180–1188. 2017.PubMed/NCBI View Article : Google Scholar

54 

Chen J, Chang H, Peng X, Gu Y, Yi L, Zhang Q, Zhu J and Mi M: 3,6-dihydroxyflavone suppresses the epithelial-mesenchymal transition in breast cancer cells by inhibiting the Notch signaling pathway. Sci Rep. 6(28858)2016.PubMed/NCBI View Article : Google Scholar

55 

Yamaguchi M, Murata T, Shoji M and Weitzmann MN: The flavonoid p-hydroxycinnamic acid mediates anticancer effects on MDA-MB-231 human breast cancer cells in vitro: Implications for suppression of bone metastases. Int J Oncol. 47:1563–1571. 2015.PubMed/NCBI View Article : Google Scholar

56 

Yu MM and Zhou QM: 3,6-dihydroxyflavone suppresses the epithelial-mesenchymal transition, migration and invasion in endometrial stromal cells by inhibiting the Notch signaling pathway. Eur Rev Med Pharmacol Sci. 22:4009–4017. 2018.PubMed/NCBI View Article : Google Scholar

57 

Du X, Yang H, Kang X, Fu C and Yang T: Blocking GATA6 alleviates pyroptosis and inhibits abdominal wall endometriosis lesion growth through inactivating the PI3K/AKT pathway. Cell Biochem Biophys. 83:1757–1770. 2025.PubMed/NCBI View Article : Google Scholar

58 

Qin R, Zheng F, Qin W, Wang J, Ma N, Tian W, Li J, Liao M and Qin A: Progranulin promotes proliferation, migration and invasion via the PI3K/Akt signalling pathway in a model of endometriosis. Reprod Biomed Online. 46:425–435. 2023.PubMed/NCBI View Article : Google Scholar

59 

Chen JJ, Xiao ZJ, Meng X, Wang Y, Yu MK, Huang WQ, Sun X, Chen H, Duan YG, Jiang X, et al: MRP4 sustains Wnt/β-catenin signaling for pregnancy, endometriosis and endometrial cancer. Theranostics. 9:5049–5064. 2019.PubMed/NCBI View Article : Google Scholar

60 

Lu Q, Huang Y, Wu J, Guan Y, Du M, Wang F, Liu Z, Zhu Y, Gong G, Hou H, et al: T-cadherin inhibits invasion and migration of endometrial stromal cells in endometriosis. Hum Reprod. 35:145–156. 2020.PubMed/NCBI View Article : Google Scholar

61 

Matsuzaki S, Botchorishvili R, Pouly JL and Canis M: Targeting the Wnt/β-catenin pathway in endometriosis: A potentially effective approach for treatment and prevention. Mol Cell Ther. 2(36)2014.PubMed/NCBI View Article : Google Scholar

62 

Lepourcelet M, Chen YN, France DS, Wang H, Crews P, Petersen F, Bruseo C, Wood AW and Shivdasani RA: Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell. 5:91–102. 2004.PubMed/NCBI View Article : Google Scholar

63 

Li Y, Wang X, Wang X, Wan L, Liu Y, Shi Y, Zhang L, Fang Z and Wei Z: PDCD4 suppresses proliferation, migration, and invasion of endometrial cells by inhibiting autophagy and NF-κB/MMP2/MMP9 signal pathway. Biol Reprod. 99:360–372. 2018.PubMed/NCBI View Article : Google Scholar

64 

Dikic I and Elazar Z: Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 19:349–364. 2018.PubMed/NCBI View Article : Google Scholar

65 

Jung S, Jeong H and Yu SW: Autophagy as a decisive process for cell death. Exp Mol Med. 52:921–930. 2020.PubMed/NCBI View Article : Google Scholar

66 

Doherty J and Baehrecke EH: Life, death and autophagy. Nat Cell Biol. 20:1110–1117. 2018.PubMed/NCBI View Article : Google Scholar

67 

Cooper KF: Till death do us part: The marriage of autophagy and apoptosis. Oxid Med Cell Longev. 2018(4701275)2018.PubMed/NCBI View Article : Google Scholar

68 

Liu H, Du Y, Zhang Z, Lv L, Xiong W, Zhang L, Li N, He H, Li Q and Liu Y: Autophagy contributes to hypoxia-induced epithelial to mesenchymal transition of endometrial epithelial cells in endometriosis. Biol Reprod. 99:968–981. 2018.PubMed/NCBI View Article : Google Scholar

69 

Pang C, Wu Z, Xu X, Yang W, Wang X and Qi Y: Paeonol alleviates migration and invasion of endometrial stromal cells by reducing HIF-1α-regulated autophagy in endometriosis. Front Biosci (Landmark Ed). 26:485–495. 2021.PubMed/NCBI View Article : Google Scholar

70 

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.PubMed/NCBI View Article : Google Scholar

71 

Wu L, Lin S, Hu Y, Jing S, Sun B, Chen X, Jia J, Zeng C and Pei F: Potential mechanism of Luoshi Neiyi prescription in endometriosis based on serum pharmacochemistry and network pharmacology. Front Pharmacol. 15(1395160)2024.PubMed/NCBI View Article : Google Scholar

72 

Foster WG: Hypoxia-induced autophagy, epithelial to mesenchymal transition, and invasion in the pathophysiology of endometriosis: A perspective. Biol Reprod. 99:905–906. 2018.PubMed/NCBI View Article : Google Scholar

73 

McDonald PC, Swayampakula M and Dedhar S: Coordinated regulation of metabolic transporters and migration/invasion by carbonic anhydrase IX. Metabolites. 8(20)2018.PubMed/NCBI View Article : Google Scholar

74 

Quan Q, Wu J, Yu M and Tang J: Immune micro-environment and drug analysis of peritoneal endometriosis based on epithelial-mesenchymal transition classification. Front Endocrinol (Lausanne). 13(1035158)2022.PubMed/NCBI View Article : Google Scholar

75 

Acloque H, Thiery JP and Nieto MA: The physiology and pathology of the EMT. Meeting on the epithelial-mesenchymal transition. EMBO Rep. 9:322–326. 2008.PubMed/NCBI View Article : Google Scholar

76 

Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP and Sethi G: Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev. 43:1141–1200. 2023.PubMed/NCBI View Article : Google Scholar

77 

Matsuzaki S and Darcha C: Epithelial to mesenchymal transition-like and mesenchymal to epithelial transition-like processes might be involved in the pathogenesis of pelvic endometriosis. Hum Reprod. 27:712–721. 2012.PubMed/NCBI View Article : Google Scholar

78 

Das V, Bhattacharya S, Chikkaputtaiah C, Hazra S and Pal M: The basics of epithelial-mesenchymal transition (EMT): A study from a structure, dynamics, and functional perspective. J Cell Physiol. 234:14535–14555. 2019.PubMed/NCBI View Article : Google Scholar

79 

Yan L, Song Z, Yi L, Tian C, Zhang R, Qin X, Wang X, Ren S, Ma X, Wang X, et al: TMEM176B inhibits ovarian cancer progression by regulating EMT via the Wnt/β-catenin signaling pathway. J Transl Med. 23(350)2025.PubMed/NCBI View Article : Google Scholar

80 

Yao Y, Niu Y, Zhou H and Yong M: KAT2B inhibits proliferation and invasion via inactivating TGF-β/Smad3 pathway-medicated autophagy and EMT in epithelial ovarian cancer. Sci Rep. 15(3417)2025.PubMed/NCBI View Article : Google Scholar

81 

Huang J, Sun X, Diao G, Li R, Guo J and Han J: KIF15 knockdown inhibits the development of endometrial cancer by suppressing epithelial-mesenchymal transition and stemness through Wnt/β-catenin signaling. Environ Toxicol. 38:1824–1834. 2023.PubMed/NCBI View Article : Google Scholar

82 

Cassier PA, Navaridas R, Bellina M, Rama N, Ducarouge B, Hernandez-Vargas H, Delord JP, Lengrand J, Paradisi A, Fattet L, et al: Netrin-1 blockade inhibits tumour growth and EMT features in endometrial cancer. Nature. 620:409–416. 2023.PubMed/NCBI View Article : Google Scholar

83 

Li N, Ji GX and Yang ZY: EFEMP2 increases the invasion ability of cervical cancer cells by promoting EMT via the Raf/MEK/ERK signaling pathway. Neoplasma. 69:1185–1197. 2022.PubMed/NCBI View Article : Google Scholar

84 

Zhu Y, Yin WF, Yu P, Zhang C, Sun MH, Kong LY and Yang L: Meso-Hannokinol inhibits breast cancer bone metastasis via the ROS/JNK/ZEB1 axis. Phytother Res. 37:2262–2279. 2023.PubMed/NCBI View Article : Google Scholar

85 

Wojtowicz-Praga SM, Dickson RB and Hawkins MJ: Matrix metalloproteinase inhibitors. Invest New Drugs. 15:61–75. 1997.PubMed/NCBI View Article : Google Scholar

86 

Sharpe-Timms KL, Keisler LW, McIntush EW and Keisler DH: Tissue inhibitor of metalloproteinase-1 concentrations are attenuated in peritoneal fluid and sera of women with endometriosis and restored in sera by gonadotropin-releasing hormone agonist therapy. Fertil Steril. 69:1128–1134. 1998.PubMed/NCBI View Article : Google Scholar

87 

Wenzl RJ and Heinzl H: Localization of matrix metalloproteinase-2 in uterine endometrium and ectopic implants. Gynecol Obstet Invest. 45:253–257. 1998.PubMed/NCBI View Article : Google Scholar

88 

Cox KE, Piva M and Sharpe-Timms KL: Differential regulation of matrix metalloproteinase-3 gene expression in endometriotic lesions compared with endometrium. Biol Reprod. 65:1297–1303. 2001.PubMed/NCBI View Article : Google Scholar

89 

Lv X, Chen P and Liu W: Down regulation of MiR-93 contributes to endometriosis through targeting MMP3 and VEGFA. Am J Cancer Res. 5:1706–1717. 2015.PubMed/NCBI

90 

Chung HW, Lee JY, Moon HS, Hur SE, Park MH, Wen Y and Polan ML: Matrix metalloproteinase-2, membranous type 1 matrix metalloproteinase, and tissue inhibitor of metalloproteinase-2 expression in ectopic and eutopic endometrium. Fertil Steril. 78:787–795. 2002.PubMed/NCBI View Article : Google Scholar

91 

Collette T, Maheux R, Mailloux J and Akoum A: Increased expression of matrix metalloproteinase-9 in the eutopic endometrial tissue of women with endometriosis. Hum Reprod. 21:3059–3067. 2006.PubMed/NCBI View Article : Google Scholar

92 

Lin TC, Wang KH, Chuang KH, Kao AP and Kuo TC: Interleukin-33 promotes invasiveness of human ovarian endometriotic stromal cells through the ST2/MAPK/MMP-9 pathway activated by 17β-estradiol. Taiwan J Obstet Gynecol. 60:658–664. 2021.PubMed/NCBI View Article : Google Scholar

93 

Gaetje R, Holtrich U, Engels K, Kourtis K, Cikrit E, Kissler S, Rody A, Karn T and Kaufmann M: Expression of membrane-type 5 matrix metalloproteinase in human endometrium and endometriosis. Gynecol Endocrinol. 23:567–573. 2007.PubMed/NCBI View Article : Google Scholar

94 

Rodgers WH, Osteen KG, Matrisian LM, Navre M, Giudice LC and Gorstein F: Expression and localization of matrilysin, a matrix metalloproteinase, in human endometrium during the reproductive cycle. Am J Obstet Gynecol. 168 (1 Pt 1):253–260. 1993.PubMed/NCBI View Article : Google Scholar

95 

Pearce CL, Templeman C, Rossing MA, Lee A, Near AM, Webb PM, Nagle CM, Doherty JA, Cushing-Haugen KL, Wicklund KG, et al: Association between endometriosis and risk of histological subtypes of ovarian cancer: A pooled analysis of case-control studies. Lancet Oncol. 13:385–394. 2012.PubMed/NCBI View Article : Google Scholar

96 

Barnard ME, Farland LV, Yan B, Wang J, Trabert B, Doherty JA, Meeks HD, Madsen M, Guinto E, Collin LJ, et al: Endometriosis typology and ovarian cancer risk. JAMA. 332:482–489. 2024.PubMed/NCBI View Article : Google Scholar

97 

Yousefi M, Dehghani S, Nosrati R, Ghanei M, Salmaninejad A, Rajaie S, Hasanzadeh M and Pasdar A: Current insights into the metastasis of epithelial ovarian cancer-hopes and hurdles. Cell Oncol (Dordr). 43:515–538. 2020.PubMed/NCBI View Article : Google Scholar

98 

Schröpfer A, Kammerer U, Kapp M, Dietl J, Feix S and Anacker J: Expression pattern of matrix metalloproteinases in human gynecological cancer cell lines. BMC Cancer. 10(553)2010.PubMed/NCBI View Article : Google Scholar

99 

Cho-Clark M, Larco DO, Zahn BR, Mani SK and Wu TJ: GnRH-(1-5) activates matrix metallopeptidase-9 to release epidermal growth factor and promote cellular invasion. Mol Cell Endocrinol. 415:114–125. 2015.PubMed/NCBI View Article : Google Scholar

100 

Kicman A, Gacuta E, Kulesza M, Będkowska EG, Marecki R, Klank-Sokołowska E, Knapp P, Niczyporuk M and Ławicki S: Diagnostic utility of selected matrix metalloproteinases (MMP-2, MMP-3, MMP-11, MMP-26), HE4, CA125 and ROMA algorithm in diagnosis of ovarian cancer. Int J Mol Sci. 25(6265)2024.PubMed/NCBI View Article : Google Scholar

101 

Shi L, Zhang Q, Zhu S, Tang Q, Chen X, Lan R, Wang N and Zhu Y: Pharmacological inhibition of EZH2 using a covalent inhibitor suppresses human ovarian cancer cell migration and invasion. Mol Cell Biochem. 479:831–841. 2024.PubMed/NCBI View Article : Google Scholar

102 

Li H, Qiu Z, Li F and Wang C: The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 14:5865–5870. 2017.PubMed/NCBI View Article : Google Scholar

103 

Ünüvar S, Melekoğlu R, Yüce H, Çelik NZ, Okumuş EB, Toprak S, Tanbek K, Yaşar Ş, Doğan A, Türkmen NB, et al: Diagnostic utility of lipocalin 2 and metalloproteinase 9 levels in early-stage endometrial cancer. Cancer Biomark. 41(18758592241290951)2024.PubMed/NCBI View Article : Google Scholar

104 

Shukla S, Qureshi S, Singh U and Khattri S: A study of matrix metalloproteinase-2 and interleukin-18 in preinvasive and invasive lesions of cancer cervix. J Midlife Health. 11:236–239. 2020.PubMed/NCBI View Article : Google Scholar

105 

Chauhan R, Malhotra L, Gupta A, Dagar G, Mendiratta M, Masoodi T, Hashem S, Al Marzooqi S, Das D, Uddin S, et al: Bergenin inhibits growth of human cervical cancer cells by decreasing Galectin-3 and MMP-9 expression. Sci Rep. 14(15287)2024.PubMed/NCBI View Article : Google Scholar

106 

Kochumon S, Al-Sayyar A, Jacob T, Bahman F, Akhter N, Wilson A, Sindhu S, Hannun YA, Ahmad R and Al-Mulla F: TGF-β and TNF-α interaction promotes the expression of MMP-9 through H3K36 dimethylation: Implications in breast cancer metastasis. Front Immunol. 15(1430187)2024.PubMed/NCBI View Article : Google Scholar

107 

Wan Y, Huang J, Song Y, Gu C, Kong J, Zuo L and Chen J: hsa-miR-340-5p inhibits epithelial-mesenchymal transition in endometriosis by targeting MAP3K2 and inactivating MAPK/ERK signaling. Open Med (Wars). 17:566–576. 2022.PubMed/NCBI View Article : Google Scholar

108 

Zhang M, Wang X, Xia X, Fang X, Zhang T and Huang F: Endometrial epithelial cells-derived exosomes deliver microRNA-30c to block the BCL9/Wnt/CD44 signaling and inhibit cell invasion and migration in ovarian endometriosis. Cell Death Discov. 8(151)2022.PubMed/NCBI View Article : Google Scholar

109 

Ntzeros K, Voros C, Mavrogianni D, Kathopoulis N, Kypriotis K, Varthaliti A, Darlas M, Douligeris A and Protopapas A: Expression of E-CADHERIN and miR-200b in Different Forms of Endometriosis. Biomedicines. 13(524)2025.PubMed/NCBI View Article : Google Scholar

110 

Hagh YN, Ahmadifard M, Esmaelzadeh S, Abbaszadeh S and Shokrzadeh N: Decreased expression of miR-200a and miR-223-3p in endometriosis during the secretory phase of menstrual cycle: Insights from a case-control study on molecular biomarkers and disease-related infertility. Int J Reprod Biomed. 22:1003–1014. 2025.PubMed/NCBI View Article : Google Scholar

111 

Wang J, Li J, Han H, Wang C, Shi T and Yang X: miR-375-3p predicts the severity of endometriosis and regulates cellular progression by targeting NOX4. Mol Cell Probes. 79(101999)2025.PubMed/NCBI View Article : Google Scholar

112 

Zhang Z, Qin Y, Huang J, Wang Y, Zeng L, Wang Y, Zhuyun F and Wang L: Oestrogen promotes the progression of adenomyosis by inhibiting CITED2 through miR-145. Reprod Biomed Online. 49(104108)2024.PubMed/NCBI View Article : Google Scholar

113 

Cui X, Zhou S and Lin Y: Long non-coding RNA DHRS4 antisense RNA 1 inhibits ectopic endometrial cell proliferation, migration, and invasion in endometriosis by regulating microRNA-139-5p expression. Bioengineered. 13:9792–9804. 2022.PubMed/NCBI View Article : Google Scholar

114 

Bao Q, Zheng Q, Wang S, Tang W and Zhang B: LncRNA HOTAIR regulates cell invasion and migration in endometriosis through miR-519b-3p/PRRG4 pathway. Front Oncol. 12(953055)2022.PubMed/NCBI View Article : Google Scholar

115 

Liu L, Wang L, Hao N, Du N, Li Y and Kang S: miRNA-1229-5p promotes migration and invasion and suppresses apoptosis of endometrial cells via the STMN1/p38 MAPK axis in endometriosis. Gene. 950(149385)2025.PubMed/NCBI View Article : Google Scholar

116 

Hudson QJ, Proestling K, Perricos A, Kuessel L, Husslein H, Wenzl R and Yotova I: The role of long non-coding RNAs in endometriosis. Int J Mol Sci. 22(11425)2021.PubMed/NCBI View Article : Google Scholar

117 

Arendt W, Kleszczyński K, Gagat M and Izdebska M: Endometriosis and cytoskeletal remodeling: The functional role of actin-binding proteins. Cells. 14(360)2025.PubMed/NCBI View Article : Google Scholar

118 

Ma J and Jiang J: ATG8 inhibited endometriosis formation by regulating Treg cells differentiation via integrin α4β1 and Talin-1 interaction. Reprod Biomed Online. 48(103646)2024.PubMed/NCBI View Article : Google Scholar

119 

Knez J, Kovačič B and Goropevšek A: The role of regulatory T-cells in the development of endometriosis. Hum Reprod: May 19, 2024 (Epub ahead of print).

120 

Sun H, Lagarrigue F and Ginsberg MH: The connection between Rap1 and Talin1 in the activation of integrins in blood cells. Front Cell Dev Biol. 10(908622)2022.PubMed/NCBI View Article : Google Scholar

121 

Rahmawati E, Yang WV, Lei YP, Maurya PK, Chen HW and Tzeng CR: Gonadotropin-releasing hormone agonist induces downregulation of tensin 1 in women with endometriosis. Acta Obstet Gynecol Scand. 98:222–231. 2019.PubMed/NCBI View Article : Google Scholar

122 

Zhang J, Wang L, Li C, Zhang H, Li R and Li M: Letrozole promotes the expression of integrin αvβ3 and HOXA10 in endometrium of endometriosis. Syst Biol Reprod Med. 68:121–128. 2022.PubMed/NCBI View Article : Google Scholar

123 

Duan R, Wang Y, Lin A, Lian L, Cao H, Gu W, Li T and Sun Q: Expression of nm23-H1, p53, and integrin β1 in endometriosis and their clinical significance. Int J Clin Exp Pathol. 13:1024–1029. 2020.PubMed/NCBI

124 

Gao X, Shao W, Wang J, Gao H, Zhang X, Xia C, Li M and Liu S: Integrin β3 enhances glycolysis and increases lactate production in endometriosis. J Reprod Immunol. 165(104312)2024.PubMed/NCBI View Article : Google Scholar

125 

Rosa-E-Silva ACJS, Mamillapalli R, Rosa-E-Silva JC, Ucar A, Schwartz J and Taylor HS: Uterine administration of C-X-C motif chemokine ligand 12 increases the pregnancy rates in mice with induced endometriosis. F S Sci. 4:65–73. 2023.PubMed/NCBI View Article : Google Scholar

126 

Harden S, Tan TY, Ku CW, Zhou J, Chen Q, Chan JKY, Brosens J and Lee YH: Peritoneal autoantibody profiling identifies p53 as an autoantibody target in endometriosis. Fertil Steril. 120:176–187. 2023.PubMed/NCBI View Article : Google Scholar

127 

Raghuwanshi S and Gartel AL: Small-molecule inhibitors targeting FOXM1: Current challenges and future perspectives in cancer treatments. Biochim Biophys Acta Rev Cancer. 1878(189015)2023.PubMed/NCBI View Article : Google Scholar

128 

Yu C, Chen L, Yie L, Wei L, Wen T, Liu Y and Chen H: Targeting FoxM1 inhibits proliferation, invasion and migration of nasopharyngeal carcinoma through the epithelial-to-mesenchymal transition pathway. Oncol Rep. 33:2402–2410. 2015.PubMed/NCBI View Article : Google Scholar

129 

Yu M, Yang Y, Zhao H, Li M, Chen J, Wang B, Xiao T, Huang C, Zhao H, Zhou W and Zhang JV: Targeting the chemerin/CMKLR1 axis by small molecule antagonist α-NETA mitigates endometriosis progression. Front Pharmacol. 13(985618)2022.PubMed/NCBI View Article : Google Scholar

130 

Lin Y, Kojima S, Ishikawa A, Matsushita H, Takeuchi Y, Mori Y, Ma J, Takeuchi K, Umezawa K and Wakatsuki A: Inhibition of MLCK-mediated migration and invasion in human endometriosis stromal cells by NF-κB inhibitor DHMEQ. Mol Med Rep. 28(141)2023.PubMed/NCBI View Article : Google Scholar

131 

Xu X, Zheng Q, Zhang Z, Zhang X, Liu R and Liu P: Periostin enhances migration, invasion, and adhesion of human endometrial stromal cells through integrin-linked kinase 1/Akt signaling pathway. Reprod Sci. 22:1098–1106. 2015.PubMed/NCBI View Article : Google Scholar

132 

Zheng QM, Lu JJ, Zhao J, Wei X, Wang L and Liu PS: Periostin facilitates the epithelial-mesenchymal transition of endometrial epithelial cells through ILK-Akt signaling pathway. Biomed Res Int. 2016(9842619)2016.PubMed/NCBI View Article : Google Scholar

133 

Li YH, Geng YY, Liu L, Chen CY and Gao Y: Lipoxin A4 inhibits the invasion and migration of endometrial stromal cells by down-regulating NF-κB signaling-mediated autophagy. Zhonghua Fu Chan Ke Za Zhi. 53:547–553. 2018.PubMed/NCBI View Article : Google Scholar : (In Chinese).

134 

Marquardt RM, Kim TH, Shin JH and Jeong JW: Progesterone and Estrogen Signaling in the Endometrium: What Goes Wrong in Endometriosis? Int J Mol Sci. 20(3822)2019.PubMed/NCBI View Article : Google Scholar

135 

Wakatsuki A, Lin Y, Kojima S, Matsushita H, Takeuchi K and Umezawa K: Inhibitory effects of estetrol on the invasion and migration of immortalized human endometrial stromal cells. Endocr J. 71:199–206. 2024.PubMed/NCBI View Article : Google Scholar

136 

Guo B, Zhu H, Xiao C, Zhang J, Liu X, Fang Y, Wei B, Zhang J, Cao Y and Zhan L: NLRC5 exerts anti-endometriosis effects through inhibiting ERβ-mediated inflammatory response. BMC Med. 22(351)2024.PubMed/NCBI View Article : Google Scholar

137 

Wang S, Long S, Deng Z and Wu W: Positive role of Chinese herbal medicine in cancer immune regulation. Am J Chin Med. 48:1577–1592. 2020.PubMed/NCBI View Article : Google Scholar

138 

He Q, Wan S, Jiang M, Li W, Zhang Y, Zhang L, Wu M, Lin J, Zou L and Hu Y: Exploring the therapeutic potential of tonic Chinese herbal medicine for gynecological disorders: An updated review. J Ethnopharmacol. 329(118144)2024.PubMed/NCBI View Article : Google Scholar

139 

Griffith JS, Binkley PA, Kirma NB, Schenken RS, Witz CA and Tekmal RR: Imatinib decreases endometrial stromal cell transmesothial migration and proliferation in the extracellular matrix of modeled peritoneum. Fertil Steril. 94:2531–2535. 2010.PubMed/NCBI View Article : Google Scholar

140 

Yao Z, Zhang J, Zhang B, Liang G, Chen X, Yao F, Xu X, Wu H, He Q, Ding L and Yang B: Imatinib prevents lung cancer metastasis by inhibiting M2-like polarization of macrophages. Pharmacol Res. 133:121–131. 2018.PubMed/NCBI View Article : Google Scholar

141 

Ferraz CR, Calixto-Campos C, Manchope MF, Casagrande R, Clissa PB, Baldo C and Verri WA Jr: Jararhagin-induced mechanical hyperalgesia depends on TNF-α, IL-1β and NFκB in mice. Toxicon. 103:119–128. 2015.PubMed/NCBI View Article : Google Scholar

142 

Pinho-Ribeiro FA, Fattori V, Zarpelon AC, Borghi SM, Staurengo-Ferrari L, Carvalho TT, Alves-Filho JC, Cunha FQ, Cunha TM, Casagrande R and Verri WA Jr: Pyrrolidine dithiocarbamate inhibits superoxide anion-induced pain and inflammation in the paw skin and spinal cord by targeting NF-κB and oxidative stress. Inflammopharmacology. 24:97–107. 2016.PubMed/NCBI View Article : Google Scholar

143 

Zhang JJ, Xu ZM, Zhang CM, Dai HY, Ji XQ, Wang XF and Li C: Pyrrolidine dithiocarbamate inhibits nuclear factor-κB pathway activation, and regulates adhesion, migration, invasion and apoptosis of endometriotic stromal cells. Mol Hum Reprod. 17:175–181. 2011.PubMed/NCBI View Article : Google Scholar

144 

Babu S and Jayaraman S: An update on β-sitosterol: A potential herbal nutraceutical for diabetic management. Biomed Pharmacother. 131(110702)2020.PubMed/NCBI View Article : Google Scholar

145 

Khan Z, Nath N, Rauf A, Emran TB, Mitra S, Islam F, Chandran D, Barua J, Khandaker MU, Idris AM, et al: Multifunctional roles and pharmacological potential of β-sitosterol: Emerging evidence toward clinical applications. Chem Biol Interact. 365(110117)2022.PubMed/NCBI View Article : Google Scholar

146 

Wen Y, Pang L, Fan L, Zhou Y, Li R, Zhao T and Zhang M: β-Sitosterol inhibits the proliferation of endometrial cells via regulating Smad7-Mediated TGF-β/Smads signaling pathway. Cell J. 25:554–563. 2023.PubMed/NCBI View Article : Google Scholar

147 

Periferakis A, Periferakis K, Badarau IA, Petran EM, Popa DC, Caruntu A, Costache RS, Scheau C, Caruntu C and Costache DO: Kaempferol: Antimicrobial properties, sources, clinical, and traditional applications. Int J Mol Sci. 23(15054)2022.PubMed/NCBI View Article : Google Scholar

148 

Imran M, Salehi B, Sharifi-Rad J, Aslam Gondal T, Saeed F, Imran A, Shahbaz M, Tsouh Fokou PV, Umair Arshad M, Khan H, et al: Kaempferol: A key emphasis to its anticancer potential. Molecules. 24(2277)2019.PubMed/NCBI View Article : Google Scholar

149 

Jantas D, Malarz J, Le TN and Stojakowska A: Neuroprotective properties of kempferol derivatives from maesa membranacea against oxidative stress-induced cell damage: An association with cathepsin D inhibition and PI3K/Akt activation. Int J Mol Sci. 22(10363)2021.PubMed/NCBI View Article : Google Scholar

150 

Zhao J, Wang J, Liu J, Li S, Liu P and Zhang X: Effect and mechanisms of kaempferol against endometriosis based on network pharmacology and in vitro experiments. BMC Complement Med Ther. 22(254)2022.PubMed/NCBI View Article : Google Scholar

151 

Ren Y, Li Y, Lv J, Guo X, Zhang J, Zhou D, Zhang Z, Xue Z, Yang G, Xi Q, et al: Parthenolide regulates oxidative stress-induced mitophagy and suppresses apoptosis through p53 signaling pathway in C2C12 myoblasts. J Cell Biochem. 120:15695–15708. 2019.PubMed/NCBI View Article : Google Scholar

152 

Freund RRA, Gobrecht P, Fischer D and Arndt HD: Advances in chemistry and bioactivity of parthenolide. Nat Prod Rep. 37:541–565. 2020.PubMed/NCBI View Article : Google Scholar

153 

Jafari N, Nazeri S and Enferadi ST: Parthenolide reduces metastasis by inhibition of vimentin expression and induces apoptosis by suppression elongation factor α - 1 expression. Phytomedicine. 41:67–73. 2018.PubMed/NCBI View Article : Google Scholar

154 

Mathema VB, Koh YS, Thakuri BC and Sillanpää M: Parthenolide, a sesquiterpene lactone, expresses multiple anti-cancer and anti-inflammatory activities. Inflammation. 35:560–565. 2012.PubMed/NCBI View Article : Google Scholar

155 

Wang M and Li Q: Parthenolide could become a promising and stable drug with anti-inflammatory effects. Nat Prod Res. 29:1092–1101. 2015.PubMed/NCBI View Article : Google Scholar

156 

Huang L, Liu F, Liu X, Niu L, Sun L, Fang F, Ma K and Hu P: Parthenolide inhibits the proliferation and migration of cervical cancer cells via FAK/GSK3β pathway. Cancer Chemother Pharmacol. 93:203–213. 2024.PubMed/NCBI View Article : Google Scholar

157 

Kabil SL, Rashed HE, Mohamed NM and Elwany NE: Parthenolide repressed endometriosis induced surgically in rats: Role of PTEN/PI3Kinase/AKT/GSK-3β/β-catenin signaling in inhibition of epithelial mesenchymal transition. Life Sci. 331(122037)2023.PubMed/NCBI View Article : Google Scholar

158 

Cheng J, Li C, Ying Y, Lv J, Qu X, McGowan E, Lin Y and Zhu X: Metformin alleviates endometriosis and potentiates endometrial receptivity via decreasing VEGF and MMP9 and increasing leukemia inhibitor factor and HOXA10. Front Pharmacol. 13(750208)2022.PubMed/NCBI View Article : Google Scholar

159 

Neto AC, Botelho M, Rodrigues AR, Lamas S, Araújo B, Guimarães JT, Gouveia AM, Almeida H and Neves D: Metformin reverses infertility in a mouse model of endometriosis: Unveiling disease pathways and implications for future clinical approaches. Reprod Biomed Online. 50(104474)2025.PubMed/NCBI View Article : Google Scholar

160 

Semwal RB, Semwal DK, Combrinck S and Viljoen A: Emodin-A natural anthraquinone derivative with diverse pharmacological activities. Phytochemistry. 190(112854)2021.PubMed/NCBI View Article : Google Scholar

161 

Demirezer LO, Kuruüzüm-Uz A, Bergere I, Schiewe HJ and Zeeck A: The structures of antioxidant and cytotoxic agents from natural source: Anthraquinones and tannins from roots of Rumex patientia. Phytochemistry. 58:1213–1217. 2001.PubMed/NCBI View Article : Google Scholar

162 

Zhang Q, Chen WW, Sun X, Qian D, Tang DD, Zhang LL, Li MY, Wang LY, Wu CJ and Peng W: The versatile emodin: A natural easily acquired anthraquinone possesses promising anticancer properties against a variety of cancers. Int J Biol Sci. 18:3498–3527. 2022.PubMed/NCBI View Article : Google Scholar

163 

Zheng Q, Xu Y, Lu J, Zhao J, Wei X and Liu P: Emodin inhibits migration and invasion of human endometrial stromal cells by facilitating the mesenchymal-epithelial transition through targeting ILK. Reprod Sci. 23:1526–1535. 2016.PubMed/NCBI View Article : Google Scholar

164 

Cui Y, Chen LJ, Huang T, Ying JQ and Li J: The pharmacology, toxicology and therapeutic potential of anthraquinone derivative emodin. Chin J Nat Med. 18:425–435. 2020.PubMed/NCBI View Article : Google Scholar

165 

Liu X, Mi X, Wang Z, Zhang M, Hou J, Jiang S, Wang Y, Chen C and Li W: Ginsenoside Rg3 promotes regression from hepatic fibrosis through reducing inflammation-mediated autophagy signaling pathway. Cell Death Dis. 11(454)2020.PubMed/NCBI View Article : Google Scholar

166 

Nakhjavani M, Smith E, Townsend AR, Price TJ and Hardingham JE: Anti-angiogenic properties of ginsenoside Rg3. Molecules. 25(4905)2020.PubMed/NCBI View Article : Google Scholar

167 

Ren B, Feng J, Yang N, Guo Y, Chen C and Qin Q: Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-κB pathway. Int Immunopharmacol. 98(107841)2021.PubMed/NCBI View Article : Google Scholar

168 

Zhu Y, Wang A, Zhang S, Kim J, Xia J, Zhang F, Wang D, Wang Q and Wang J: Paclitaxel-loaded ginsenoside Rg3 liposomes for drug-resistant cancer therapy by dual targeting of the tumor microenvironment and cancer cells. J Adv Res. 49:159–173. 2023.PubMed/NCBI View Article : Google Scholar

169 

Kim MK, Lee SK, Park JH, Lee JH, Yun BH, Park JH, Seo SK, Cho S and Choi YS: Ginsenoside Rg3 decreases fibrotic and invasive nature of endometriosis by modulating miRNA-27b: In vitro and in vivo studies. Sci Rep. 7(17670)2017.PubMed/NCBI View Article : Google Scholar

170 

Vlavcheski F, O'Neill EJ, Gagacev F and Tsiani E: Effects of berberine against pancreatitis and pancreatic cancer. Molecules. 27(8630)2022.PubMed/NCBI View Article : Google Scholar

171 

Habtemariam S: Berberine pharmacology and the gut microbiota: A hidden therapeutic link. Pharmacol Res. 155(104722)2020.PubMed/NCBI View Article : Google Scholar

172 

Feng X, Sureda A, Jafari S, Memariani Z, Tewari D, Annunziata G, Barrea L, Hassan STS, Šmejkal K, Malaník M, et al: Berberine in cardiovascular and metabolic diseases: From mechanisms to therapeutics. Theranostics. 9:1923–1951. 2019.PubMed/NCBI View Article : Google Scholar

173 

Wang K, Feng X, Chai L, Cao S and Qiu F: The metabolism of berberine and its contribution to the pharmacological effects. Drug Metab Rev. 49:139–157. 2017.PubMed/NCBI View Article : Google Scholar

174 

Chen Y, Li K, Zhao H, Hao Z, Yang Y, Gao M and Zhao D: Integrated lipidomics and network pharmacology analysis to reveal the mechanisms of berberine in the treatment of hyperlipidemia. J Transl Med. 20(412)2022.PubMed/NCBI View Article : Google Scholar

175 

Hu S, Wang J, Liu E, Zhang X, Xiang J, Li W, Wei P, Zeng J, Zhang Y and Ma X: Protective effect of berberine in diabetic nephropathy: A systematic review and meta-analysis revealing the mechanism of action. Pharmacol Res. 185(106481)2022.PubMed/NCBI View Article : Google Scholar

176 

Braicu OL, Budisan L, Buiga R, Jurj A, Achimas-Cadariu P, Pop LA, Braicu C, Irimie A and Berindan-Neagoe I: miRNA expression profiling in formalin-fixed paraffin-embedded endometriosis and ovarian cancer samples. Onco Targets Ther. 10:4225–4238. 2017.PubMed/NCBI View Article : Google Scholar

177 

Gu Y and Zhou Z: Berberine inhibits the proliferation, invasion and migration of endometrial stromal cells by downregulating miR-429. Mol Med Rep. 23(416)2021.PubMed/NCBI View Article : Google Scholar

178 

Fu Z, Liu H, Kuang Y, Yang J, Luo M, Cao L and Zheng W: β-elemene, a sesquiterpene constituent from Curcuma phaeocaulis inhibits the development of endometriosis by inducing ferroptosis via the MAPK and STAT3 signaling pathways. J Ethnopharmacol. 341(119344)2025.PubMed/NCBI View Article : Google Scholar

179 

Liu Y, Cao H, Zheng S and Zhuang Y: Unveiling the therapeutic mechanisms of taraxasterol from dandelion in endometriosis: Network pharmacology and cellular insights. Biochem Biophys Res Commun. 742(151079)2025.PubMed/NCBI View Article : Google Scholar

180 

Wu RF, Yang HM, Zhou WD, Zhang LR, Bai JB, Lin DC, Ng TW, Dai SJ, Chen QH and Chen QX: Effect of interleukin-1β and lipoxin A(4) in human endometriotic stromal cells: Proteomic analysis. J Obstet Gynaecol Res. 43:308–319. 2017.PubMed/NCBI View Article : Google Scholar

181 

Matsuzaki S and Darcha C: In vitro effects of a small-molecule antagonist of the Tcf/ß-catenin complex on endometrial and endometriotic cells of patients with endometriosis. PLoS One. 8(e61690)2013.PubMed/NCBI View Article : Google Scholar

182 

van Winden AW, van den Broek I, Gast MC, Engwegen JY, Sparidans RW, van Dulken EJ, Depla AC, Cats A, Schellens JH, Peeters PH, et al: Serum degradome markers for the detection of breast cancer. J Proteome Res. 9:3781–3788. 2010.PubMed/NCBI View Article : Google Scholar

183 

Chen Y, Li H, Cheng HY, Rui-Qiong M, Ye X, Cui H, Hong-Lan Z and Chang XH: Fibrinogen alpha chain is up-regulated and affects the pathogenesis of endometriosis. Reprod Biomed Online. 39:893–904. 2019.PubMed/NCBI View Article : Google Scholar

184 

Li H, Ma RQ, Cheng HY, Ye X, Zhu HL and Chang XH: Fibrinogen alpha chain promotes the migration and invasion of human endometrial stromal cells in endometriosis through focal adhesion kinase/protein kinase B/matrix metallopeptidase 2 pathway†. Biol Reprod. 103:779–790. 2020.PubMed/NCBI View Article : Google Scholar

185 

Akbarzadeh M, Movassaghpour AA, Ghanbari H, Kheirandish M, Fathi Maroufi N, Rahbarghazi R, Nouri M and Samadi N: The potential therapeutic effect of melatonin on human ovarian cancer by inhibition of invasion and migration of cancer stem cells. Sci Rep. 7(17062)2017.PubMed/NCBI View Article : Google Scholar

186 

Bu S, Wang Q, Sun J, Li X, Gu T and Lai D: Melatonin suppresses chronic restraint stress-mediated metastasis of epithelial ovarian cancer via NE/AKT/beta-catenin/SLUG axis. Cell Death Dis. 11(644)2020.PubMed/NCBI View Article : Google Scholar

187 

El-Sokkary GH, Ismail IA and Saber SH: Melatonin inhibits breast cancer cell invasion through modulating DJ-1/KLF17/ID-1 signaling pathway. J Cell Biochem. 120:3945–3957. 2019.PubMed/NCBI View Article : Google Scholar

188 

Qi S, Yan L, Liu Z, Mu YL, Li M, Zhao X, Chen ZJ and Zhang H: Melatonin inhibits 17β-estradiol-induced migration, invasion and epithelial-mesenchymal transition in normal and endometriotic endometrial epithelial cells. Reprod Biol Endocrinol. 16(62)2018.PubMed/NCBI View Article : Google Scholar

189 

Bhattacharya S, Patel KK, Dehari D, Agrawal AK and Singh S: Melatonin and its ubiquitous anticancer effects. Mol Cell Biochem. 462:133–155. 2019.PubMed/NCBI View Article : Google Scholar

190 

Reiter RJ, Rosales-Corral SA, Tan DX, Acuna-Castroviejo D, Qin L, Yang SF and Xu K: Melatonin, a full service anti-cancer agent: Inhibition of initiation, progression and metastasis. Int J Mol Sci. 18(843)2017.PubMed/NCBI View Article : Google Scholar

191 

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 View Article : Google Scholar

192 

Kwon MR, Park JS, Ko EJ, Park J, Ju EJ, Shin SH, Son GW, Lee HW, Park YY, Kang MH, et al: Ibulocydine inhibits migration and invasion of TNBC cells via MMP-9 regulation. Int J Mol Sci. 25(6123)2024.PubMed/NCBI View Article : Google Scholar

193 

Liang RN, Li PS, Zou Y, Liu YL, Jiang Z, Liu Z, Fan P, Xu L, Peng JH and Sun XY: Ping-Chong-Jiang-Ni formula induces apoptosis and inhibits proliferation of human ectopic endometrial stromal cells in endometriosis via the activation of JNK signaling pathway. Evid Based Complement Alternat Med. 2017(6489427)2017.PubMed/NCBI View Article : Google Scholar

194 

Huang J, Zhang X, Wang J, Gu C, Zhang Y, Hu G and Chen J: Mechanism of Yushenhuoxue prescription in treating endometriosis based on network pharmacology and the effect on the TNF pathway. Heliyon. 9(e20283)2023.PubMed/NCBI View Article : Google Scholar

195 

Xu Y, Li Y, Zhang J and Cai P: Hua Yu Xiao Zheng decoction induces ectopic endometrial stromal cell senescence via inhibiting the PI3K/AKT signaling. Tissue Cell. 93(102763)2025.PubMed/NCBI View Article : Google Scholar

196 

Miller MA, Meyer AS, Beste MT, Lasisi Z, Reddy S, Jeng KW, Chen CH, Han J, Isaacson K, Griffith LG and Lauffenburger DA: ADAM-10 and -17 regulate endometriotic cell migration via concerted ligand and receptor shedding feedback on kinase signaling. Proc Natl Acad Sci USA. 110:E2074–E2083. 2013.PubMed/NCBI View Article : Google Scholar

197 

Zanon P, Terraciano PB, Quandt L, Palma Kuhl C, Pandolfi Passos E and Berger M: Angiotensin II-AT1 receptor signalling regulates the plasminogen-plasmin system in human stromal endometrial cells increasing extracellular matrix degradation, cell migration and inducing a proinflammatory profile. Biochem Pharmacol. 225(116280)2024.PubMed/NCBI View Article : Google Scholar

198 

Chen J, Shen L, Wu T and Yang Y: Unraveling the significance of AGPAT4 for the pathogenesis of endometriosis via a multi-omics approach. Hum Genet. 143:1163–1174. 2024.PubMed/NCBI View Article : Google Scholar

199 

Zheng R, Liu Y, Lei Y and Yue Y: Upregulated microRNA-429 confers endometrial stromal cell dysfunction by targeting HIF1AN and regulating the HIF1A/VEGF pathway. Open Med (Wars). 18(20230775)2023.PubMed/NCBI View Article : Google Scholar

200 

Zhang Y and Yang H: Silencing of FZD7 inhibits endometriotic cell viability, migration, and angiogenesis by promoting ferroptosis. Cell Biochem Biophys. 83:2471–2480. 2025.PubMed/NCBI View Article : Google Scholar

201 

Lv X and Li F: METTL14 promotes proliferation, migration, and invasion in endometriotic stromal cell growth by activating the ZEB1/MEK/ERK pathway. Gynecol Obstet Invest. 90:42–54. 2025.PubMed/NCBI View Article : Google Scholar

202 

Tang Y, Lu X and Lin K, Li J, Yuan M and Lin K: m6A methylation of RNF43 inhibits the progression of endometriosis through regulating oxidative phosphorylation via NDUFS1. J Cell Physiol. 239(e31367)2024.PubMed/NCBI View Article : Google Scholar

203 

Vu TH, Nakamura K, Shigeyasu K, Kashino C, Okamoto K, Kubo K, Kamada Y and Masuyama H: Apolipoprotein-B mRNA-editing complex 3B could be a new potential therapeutic target in endometriosis. Sci Rep. 14(24968)2024.PubMed/NCBI View Article : Google Scholar

204 

Practice bulletin no. 114. Management of endometriosis. Obstet Gynecol. 116:223–236. 2010.

205 

Vlahos N, Vlachos A, Triantafyllidou O, Vitoratos N and Creatsas G: Continuous versus cyclic use of oral contraceptives after surgery for symptomatic endometriosis: A prospective cohort study. Fertil Steril. 100:1337–1342. 2013.PubMed/NCBI View Article : Google Scholar

206 

Jewson M, Purohit P and Lumsden MA: Progesterone and abnormal uterine bleeding/menstrual disorders. Best Pract Res Clin Obstet Gynaecol. 69:62–73. 2020.PubMed/NCBI View Article : Google Scholar

207 

Rafique S and Decherney AH: Medical management of endometriosis. Clin Obstet Gynecol. 60:485–496. 2017.PubMed/NCBI View Article : Google Scholar

208 

Resta C, Moustogiannis A, Chatzinikita E, Malligiannis Ntalianis D, Malligiannis Ntalianis K, Philippou A, Koutsilieris M and Vlahos N: Gonadotropin-releasing hormone (GnRH)/GnRH receptors and their role in the treatment of endometriosis. Cureus. 15(e38136)2023.PubMed/NCBI View Article : Google Scholar

209 

Surrey ES, Katz-Jaffe M, Kondapalli LV, Gustofson RL and Schoolcraft WB: GnRH agonist administration prior to embryo transfer in freeze-all cycles of patients with endometriosis or aberrant endometrial integrin expression. Reprod Biomed Online. 35:145–151. 2017.PubMed/NCBI View Article : Google Scholar

210 

Leconet W, Chentouf M, du Manoir S, Chevalier C, Sirvent A, Aït-Arsa I, Busson M, Jarlier M, Radosevic-Robin N, Theillet C, et al: Therapeutic activity of Anti-AXL antibody against triple-negative breast cancer patient-derived xenografts and metastasis. Clin Cancer Res. 23:2806–2816. 2017.PubMed/NCBI View Article : Google Scholar

211 

Duan Y, Luo L, Qiao C, Li X, Wang J, Liu H, Zhou T, Shen B, Lv M and Feng J: A novel human anti-AXL monoclonal antibody attenuates tumour cell migration. Scand J Immunol. 90(e12777)2019.PubMed/NCBI View Article : Google Scholar

212 

Colavito SA: AXL as a target in breast cancer therapy. J Oncol. 2020(5291952)2020.PubMed/NCBI View Article : Google Scholar

213 

Davis JD, Bravo Padros M, Conrado DJ, Ganguly S, Guan X, Hassan HE, Hazra A, Irvin SC, Jayachandran P, Kosloski MP, et al: Subcutaneous administration of monoclonal antibodies: Pharmacology, delivery, immunogenicity, and learnings from applications to clinical development. Clin Pharmacol Ther. 115:422–439. 2024.PubMed/NCBI View Article : Google Scholar

214 

Ye X, Li Y, Stawicki S, Couto S, Eastham-Anderson J, Kallop D, Weimer R, Wu Y and Pei L: An anti-Axl monoclonal antibody attenuates xenograft tumor growth and enhances the effect of multiple anticancer therapies. Oncogene. 29:5254–5264. 2010.PubMed/NCBI View Article : Google Scholar

215 

Netcharoensirisuk P, Abrahamian C, Tang R, Chen CC, Rosato AS, Beyers W, Chao YK, Filippini A, Di Pietro S, Bartel K, et al: Flavonoids increase melanin production and reduce proliferation, migration and invasion of melanoma cells by blocking endolysosomal/melanosomal TPC2. Sci Rep. 11(8515)2021.PubMed/NCBI View Article : Google Scholar

216 

Li M, Guo T, Lin J, Huang X, Ke Q, Wu Y, Fang C and Hu C: Curcumin inhibits the invasion and metastasis of triple negative breast cancer via Hedgehog/Gli1 signaling pathway. J Ethnopharmacol. 283(114689)2022.PubMed/NCBI View Article : Google Scholar

217 

Yoo JY, Kim TH, Shin JH, Marquardt RM, Müller U, Fazleabas AT, Young SL, Lessey BA, Yoon HG and Jeong JW: Loss of MIG-6 results in endometrial progesterone resistance via ERBB2. Nat Commun. 13(1101)2022.PubMed/NCBI View Article : Google Scholar

218 

Jiang Y, Palomares AR, Munoz P, Nalvarte I, Acharya G, Inzunza J, Varshney M and Rodriguez-Wallberg KA: Proof-of-concept for long-term human endometrial epithelial organoids in modeling menstrual cycle responses. Cells. 13(1811)2024.PubMed/NCBI View Article : Google Scholar

219 

Abdolmaleki A, Jalili C, Mansouri K and Bakhtiari M: New rat to mouse xenograft transplantation of endometrium as a model of human endometriosis. Animal Model Exp Med. 4:268–277. 2021.PubMed/NCBI View Article : Google Scholar

220 

Ma J, Liao Z, Li J, Li X, Guo H, Zhong Q, Huang J, Shuai X and Chen S: A cRGD-modified liposome for targeted delivery of artesunate to inhibit angiogenesis in endometriosis. Biomater Sci. 13:1045–1058. 2025.PubMed/NCBI View Article : Google Scholar

221 

Abhang A and Burgess DJ: Recent advancements and future applications of intrauterine drug delivery systems. Expert Opin Drug Deliv. 22:841–856. 2025.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

September-2025
Volume 23 Issue 3

Print ISSN: 2049-9434
Online ISSN:2049-9442

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wei N, Guan H, Zhang Y, Shi J, Ma J, Shi R, Qi X, Dong Z and Zhao R: Molecular mechanisms underlying the inhibition of cell migration and invasion in endometriosis: Advances in pharmacological research (Review). Biomed Rep 23: 152, 2025.
APA
Wei, N., Guan, H., Zhang, Y., Shi, J., Ma, J., Shi, R. ... Zhao, R. (2025). Molecular mechanisms underlying the inhibition of cell migration and invasion in endometriosis: Advances in pharmacological research (Review). Biomedical Reports, 23, 152. https://doi.org/10.3892/br.2025.2030
MLA
Wei, N., Guan, H., Zhang, Y., Shi, J., Ma, J., Shi, R., Qi, X., Dong, Z., Zhao, R."Molecular mechanisms underlying the inhibition of cell migration and invasion in endometriosis: Advances in pharmacological research (Review)". Biomedical Reports 23.3 (2025): 152.
Chicago
Wei, N., Guan, H., Zhang, Y., Shi, J., Ma, J., Shi, R., Qi, X., Dong, Z., Zhao, R."Molecular mechanisms underlying the inhibition of cell migration and invasion in endometriosis: Advances in pharmacological research (Review)". Biomedical Reports 23, no. 3 (2025): 152. https://doi.org/10.3892/br.2025.2030