Open Access

Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review)

  • Authors:
    • Wenwen Zhang
    • Kang Li
    • Aiwen Jian
    • Guanran Zhang
    • Xiaoli Zhang
  • View Affiliations

  • Published online on: December 23, 2024     https://doi.org/10.3892/mmr.2024.13422
  • Article Number: 57
  • Copyright: © Zhang 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 (EM) is a chronic inflammatory disease that is one of the most common causes of gynecological systemic lesions in women before menopause. The most representative histological feature of EM is that the endometrium appears outside of the uterine cavity, often in the ovary. Although it is generally accepted that the epithelial and stromal cells of the ectopic endometrium are not malignant, they still have numerous similarities to malignant tumors, including considerable changes to the immune microenvironment (immune monitoring disorder), the creation of a specific hormone environment, high levels of oxidative stress, chronic inflammation and abnormal immune cell regulation. The pathogenesis of EM is not fully understood, which makes it difficult to identify specific biomarkers and potential therapeutic targets for early disease diagnosis and effective treatment. However, considerable progress has been made in this field over the past few decades. The purpose of the present review is to summarize the confirmed and potential biomarkers for EM, and to identify potential therapeutic targets based on changes in immunological factors (including natural killer cells, macrophages, the complement system, miRNA and P‑selectin) in the ectopic endometrial tissue. It is hoped that this work can be used as the basis for identifying accurate diagnostic markers for EM and developing personalized immune‑targeted therapy.

Introduction

Endometriosis (EM) is a common disease of the female reproductive system in which endometrial tissue exists outside of the uterus. Current estimates suggest that the total number of women diagnosed with endometriosis worldwide is as high as 190 million (1,2). These ectopic endometrial tissues are usually found in the ovary, ovarian fossa, uterosacral ligament, and both the anterior and posterior compartments of the pelvis (35). Although EM is recognized as benign cell proliferation, it has characteristics similar to malignant tumors, such as progressive and invasive growth, genetic instability, excessive proliferation, estrogen-dependent growth and a tendency to metastasize (6). Studies over the past few decades have shown that there is a correlation between EM and susceptibility to a variety of malignancies, including endometrioid carcinoma, clear cell carcinoma and low-grade serous ovarian cancer (7,8). It has also been reported that multifocal EM often presents with clonal growth and an increased mutation load, which are similar characteristics to cancer (9). The ectopic epithelial cells of patients with advanced EM even show signs of atypical hyperplasia. Typical changes of EM, as reported in the studies by Czernobilsky and Morris (10), and LeGrenade and Silverberg (11), which are used as diagnostic criteria in most studies (6), include three features: i) Enlarged hyperchromatic or morbid nuclei with moderate to considerable pleomorphism; ii) increased nuclear:cytoplasmic ratio; and iii) crowding, stratification or tufting of cells. This indicates that EM may be a transitional form between a benign and malignant lesion.

A delayed clinical diagnosis of EM is common (12), which may lead to disease-associated deterioration, a poor prognosis and an increased recurrence rate. Patients with at least one of the following symptoms may be candidates for an EM diagnosis: i) Dysmenorrhea that affects daily activities and life; ii) chronic pelvic pain and pain during or after intercourse; iii) gastrointestinal symptoms associated with the menstrual cycle (especially painful bowel movements); iv) urinary symptoms associated with the menstrual cycle (particularly hematuria or painful urination); and v) infertility in combination with at least one of the aforementioned symptoms.

The actual prevalence of EM among adult women remains unknown (13). The prevalence of EM in infertile women is 1.5–5%, and the prevalence of EM in women who undergo sterilization can range from 2–68% (14). For women who suffer from pelvic pain, the rate of identifying EM lesions during laparoscopy can range from 15 to 75% (15). EM invading other organs is often accompanied by specific symptoms, such as frequent bowel movements, constipation, hematochezia, painful bowel movements or bowel cramps, in the setting of intestinal EM (16). Other ancillary examinations, including ultrasound, MRI, cystoscopy, enteroscopy, transintestinal ultrasound and biopsy are frequently used in the clinical diagnosis of EM (1619). No specific biomarker is currently capable of diagnosing EM.

The general purpose of EM therapy is to reduce and eliminate lesions and pain, improve and promote fertility, and reduce and avoid recurrence. Treatments should strictly follow the following principles: i) Clinical problem-oriented, patient-centered, comprehensive long-term management according to different age stages; ii) empirical drug therapy should be started as early as possible based on the clinical diagnosis; iii) the timing of surgery should be standardized and attention should be paid to the protection of ovarian function and fertility to maximize the benefits of surgery; iv) after conservative surgery, long-term drug management and comprehensive treatment should be used to prevent recurrence; and v) regular review is recommended. Patients with considerable risk factors for malignant transformation should receive additional attention to avoid a missed or delayed diagnosis.

Medical and surgical treatments are both common in the clinical management of EM (17). Long-term management of EM should maximize the efficacy of drug therapies by suppressing the activity and differentiation of lymphocytes, forming an in vivo hypoestrogenic environment and relieving pain (17,20). Five main types of drugs are included in the common medical management of EM: Non-steroidal anti-inflammatory drugs, progestins, combined oral contraceptives (COCs), gonadotropin-releasing hormone agonists (GnRH-a) and traditional Chinese medicine (2123). Surgical treatment is recommended for patients who are infertile, who have adnexal cysts with a diameter >4 cm and who are unresponsive to medical treatment (24). Different types of surgery are carried out according to the preoperative evaluation and personal needs of the patient. Lesion resection (or conservative surgery), which is mainly conducted laparoscopically, preserves reproductive function (17,25). Hysterectomy is suitable for patients with severe symptoms or those at a high risk of recurrence, who have no reproductive requirements but wish to preserve their ovarian endocrine function. Hysterectomy and bilateral adnexectomy are recommended for patients with severe symptoms, a high risk of recurrence, no reproductive requirements and who are unresponsive to drug therapies (25). Since EM is prone to relapse and has a considerable impact on female fertility (26), preserving the reproductive ability and endocrine function of the ovaries and uterus, and preventing disease recurrence should be the top priority of EM management. However, a more thorough understanding and interpretation of the pathogenesis and etiology of EM is required to make the most of current medical treatments and to innovate new techniques for achieving an improved outcome.

The origin and pathogenesis of EM remains unclear. At present, the most commonly accepted theory is Sampson's retrograde menstruation theory, in which menstrual debris may be transferred to the peritoneal cavity through the reverse peristalsis of the fallopian tube (27,28). However, it is argued that retrograde menstruation is widespread in healthy women, and that retrograde menstruation alone does not necessarily lead to EM (29).

Etiological study of EM shows that it is a multifactorial disease. Pathological studies have shown that the immune microenvironment in the ectopic endometrium is considerably altered. Researchers found that there were considerable abnormalities in the immune surveillance system of ectopic endometrial tissue, which permits its implantation into the peritoneal cavity without clearance by immune tissue (3032). Ectopic endometrial tissue not only promotes an oxidative stress response and chronic inflammation in the ectopic areas, but also promotes the aggregation and activation of macrophages, thus inducing the production and release of the growth factors, angiogenic factors and inflammatory cytokines secreted by macrophages. This may also be the reason why EM effects fusion of the spermatocyte and oocyte, embryo implantation and embryo development, resulting in reproductive disorders (3335).

The ectopic endometrium also has an abnormal inflammatory hormone environment that is characterized by local estrogen levels that are increased several-fold when compared to that of the peripheral blood. This results in a series of cellular and cytokine responses that include cell proliferation and the release of various immune and inflammatory factors, such as tumor necrosis factor-α (TNF-α), transforming growth factor (TGF)-β1, interleukin (IL)-1, IL-6, IL-8 and IL-10 (36). Possible subsequent clinical outcomes include an acute inflammatory response, pain (including dysmenorrhea, chronic pelvic pain and dyspareunia), gastrointestinal symptoms (painful bowel movements) and urinary symptoms (hematuria) that are associated with the menstrual cycle, as well as EM-associated infertility (35,3740) (Fig. 1).

Researchers therefore hypothesize that EM is not only a gynecological disease, but also a chronic inflammatory systemic disease that is associated with immunity. Findings that support this include increases in non-specific inflammatory markers, such as CA-125 and CRP, and the presence of antinuclear antibodies in the patient's peripheral blood (4043). Immune cells and their products are typically able to detect and eliminate abnormal cells (44). Considerable changes have been found in the regulation of various immune cells in patients with EM, including downregulation of the cytotoxicity of natural killer (NK) cells, infiltration and activation of macrophages, infiltration and dysfunction of T and B lymphocytes, activation of polyclonal B lymphocytes, impaired apoptosis, dysfunction of Th1 and B cells, and translocation of T regulatory cells (4548). This abnormal immune cell regulation provides various targets for EM therapies. The inhibition of NK cells and the abnormal activation of macrophages are considered key factors in the progression of EM, and therefore potential targets for EM immunotherapy (4951). In addition to the requirement for a favorable immune environment for the survival of ectopic endometrial tissue, hypoxia stress and adhesiveness are two additional obstacles for the successful implantation of ectopic endometrial tissue. In a previous study, increased levels of hypoxia inducible factor-1α (HIF-1α) were observed in ectopic endometrial tissues compared with those in normal endometrium (52). HIF-1α is considered the best biomarker for tissue hypoxia and has an important role in the hypoxic response to ectopic tissues, including cell adhesion, angiogenesis and cell proliferation (53). As predicted, the inhibition of HIF-1α production in a mouse model of EM induced by suturing slices of uterus to intestinal mesenteric vessels could inhibit EM progression (54). HIF-1α could therefore function as the molecular target of EM therapy. Previous studies have shown that hypoxia promotes the release of angiogenic factors, such as vascular endothelial growth factor (VEGF-A), and inflammatory cytokines, such as IL-1β, TNF, TGF-β and IL-8 (5557). Some responses to hypoxia interact with and regulate the activity of certain types of immune cells. For example, hypoxia-induced TGF-β elevation in the peritoneal fluid of patients with EM was found to be associated with the suppression of NK cells (58). Activation of macrophages in the peritoneal fluid in response to hypoxia was also found to be associated with and possibly contributed to the reduced cytotoxicity of NK cells in patients with EM (59). The programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) pathway was also found to be involved in the immune tolerance that contributes to the pathogenesis of EM (60). These results suggest that not only are NK cells potential therapeutic targets for EM, but that immune checkpoint blocking to avoid NK cell immunosuppression can be used to investigate alternative methods for treating EM.

The normal endometrium contains various immune cells that change in distribution and number throughout the menstrual cycle (61). The normal periodic changes in immune cells are dysregulated in EM, considerably impacting both the composition and function of immune cells. Lymphocytes and macrophages are the main components in the lesion microenvironment. Compared with healthy women, women with EM have a considerably increased proportion of peritoneal macrophages in the peritoneal fluid, which contributes to the proliferation and survival of ectopic endometrial cells (62). The levels of the main components of potential biomarkers in the peritoneal fluid are also increased in the setting of EM, including phosphatidylcholine, phenylalanine isoleucine, glycidyl deproteinization, placental protein 14, midkine, IL-8 and osteoprotegerin (59,60). Changes in the composition and proportion of certain molecules in the peritoneal fluid may lead to impaired T cell and NK cell cytotoxicity (47,63). The aforementioned changes in the immune environment help to establish an immunosuppressive microenvironment that is conducive to the proliferation and invasion of epithelial cells and stromal cells into the ectopic endometrial tissue, and supports angiogenesis in the ectopic microenvironment (64,65).

The treatment for EM includes surgical resection and hormone therapy, both of which have advantages and disadvantages. Surgical treatment can remove the ectopic cyst and identify the location of the lesion. However, surgical resection without long-term medical treatment has a high EM recurrence rate and a decline in ovarian function. Medical therapy can slow the progression of EM to a certain extent, delay the need for surgery and avoid surgical complications. However, it cannot clarify the nature of the lesion or effectively reduce the lesion size. As specific targeted immunotherapy is usually not universal and there is not enough experimental validation, it may be potentially effective in this domain and deserves more attention (66,67).

NK cells

Function and regulation of NK cells

NK cells are large granular lymphocytes that are characterized by CD56+, CD16+/− and CD57+/− expression, and positivity for natural cytotoxicity trigger receptor 1 (NCR1), otherwise known as CD335, but not CD3 or surface T cell receptor (68,69).

NK cells can be divided by their expression level of CD56 into cd56bright and cd56dim, which have increased and reduced expression levels of CD56, respectively. Cd56bright NK cells produce more abundant cytokines, while cd56dim NK cells have increased cytotoxicity and increased expression levels of FC γ receptor III (fcgr3), also known as CD16 (70,71). NK cells spontaneously recognize and eliminate infected, ectopic, tumorigenic and stress responsive cells so as to automatically monitor for viral infections, ectopic tissue and malignant cells (7274). Repeated exposure to the same target results in increased accumulation of NK cells and the production of a specific recall response by NK cells that is characterized by the enhancement of the functional activity of NK cells against the target (7577). The binding of inhibitory killer cell immunoglobulin-like receptor (KIR) on NK cells with MHC class 1 or human leukocyte antigen (HLA)-1 (KIR/HLA-1) inhibits the activation of NK cells and allow for further intrinsic interactions. KIR therefore has an important role in distinguishing autologous cells from diseased and foreign cells to avoid non-selective killing of autologous healthy cells (78,79). The maturation and cytotoxic function of NK cells is based on the interactions between KIR and autologous MHC molecules, which is called licensing (80). Once licensed and functionally mature, NK cells are inhibited by inhibitory receptors that bind to the autologous MHC. Cells without MHC class 1 expression will be eliminated by activated NK cells (81). If NK cells are not stimulated by interaction with autologous MHC they may lose their normal function (82). However, the inhibition of KIRs and MHC is not absolute in mature NK cells and can also be eliminated or offset by a much stronger active stimulator. Other MHC receptors are also involved in NK cell cytotoxicity regulation against target cells, such as leukocyte immunoglobulin-like receptor subfamily B (LILRB) and natural killer group protein 2 (NKG2) (8385).

Among the receptors that activate NK cells, FCGR3, which is expressed in almost all NK cells, is key for antibody dependent cytotoxicity. FCGR3 expression levels alone are sufficient to induce interferon γ and TNF, making it is one of the most effective activating receptors of NK cells. Other receptors that can activate NK cell cytotoxicity include NCRs, which are divided into NCR1, NCR2 and NCR3 (or NKp46, NKp44 and NKp30, respectively) (86).

In addition to licensing through the interaction between receptors on the surface of NK cells and the autoantibodies of the body, exposure to cytokines is also essential to activate the cytotoxicity of immature NK cells and promote cytokine secretion (87). IL-2 and IL-15 secretion by macrophages can activate and trigger the maturation of NK cells and promote their proliferation (8890). Simultaneous exposure to IL-12 and IL-18 is not only able to activate NK cells, but can promote IFN-γ secretion (91,92). Conversely, increased interferon secretion can also enhance the cytotoxicity of NK cells, such as the anti-tumor response of NK cells that is mediated by the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway activation (93). Activated NK cells can induce apoptosis by releasing cytolytic particles against target cells. They can also by cytotoxic to target cells through a Fas L-mediated mechanism with the help of the CD95 receptors on the surface of target cells (94,95).

NK cells in EM

Several reports have shown that NK cells in patients with EM have reduced abilities both in clearing out of ectopic endometrium and in participating in local and systemic immunity, which creates a favorable environment for the survival and growth of ectopic endometrial tissue (9698). NK cytotoxicity decreases not only in ectopic endometrial tissue, but also in the peripheral blood and peritoneal fluid (99).

Decreased NK cytotoxicity is currently debated, as to date, studies on this topic have not adequately shown this cytotoxicity to occur (48,100,101). It has been speculated that the upregulation of NK cell inhibitory receptors and the downregulation of stimulatory receptors in ectopic endometrium may be caused by cytokines such as IL-2, IFNs and TGF-β. This indicates that cytokine therapy targeting NK cell inhibitory or stimulatory receptors is feasible (102,103). Most studies show that the proportion of NK cells decreases in patients with EM. However, there are also studies showing that the proportion of NK cells in patients with EM increases when compared with that in healthy controls (104,105). These results suggest that the decreased NK cytotoxicity in patients with EM is not the result of decreased NK cell infiltration, but rather the abnormal expression level of NK cell activation receptor and/or inhibition receptor. However, only the upregulation of NK cell inhibitory receptors is supported by current research, while the regulation of stimulatory receptors remains unclear due to the lack of studies and considerable results.

Overexpression of inhibitory receptors is considered vital for modulating immune evasion and maintaining immune tolerance in EM. Increased levels of HLA-1 in the glandular and stromal cells of endometrial tissue was observed in the study by Vernet-Tomas Mdel et al (106), which may result in increased resistance to NK cytolysis in patients with EM (106). The case-control study by Wu et al (107) finds that the levels of KIR in the peritoneal NK cells of patients with EM also increased (including NKB1 and EB6), thus further reducing the cytotoxicity of NK cells (107). Overexpression of other inhibitory receptors and their ligands were observed in patients with EM, including the inhibitory receptors KIR2DL1, CD94/NKG2a and LILRB1 on peritoneal NK cells, and their separate endogenous ligands HLA-C, HLA-E and HLA-G (108110). The altered presence and distinct combined presence of different KIR genes contributes to a unique genetic background of patients with EM. It should be noted that not all KIR/HLA binding promotes the development of EM. For example, receptor KIR2DS5 in combination with its ligand HLA-C C2 has a protective effect against EM (111).

Decreased expression levels of NKG2D, a stimulatory receptor of NK cells, was also reported in patients with EM (112). It is plausible that the downregulation of NKG2D is the result of increased levels of TGF-β and ectopic endometrial tissue-derived IL-15 (113,114). However, the ligands for NKG2D, including MHC class-I chain-associated proteins (MIC)A and B that were upregulated in the peritoneal fluid of patients with EM (115), paradoxically exert an inhibitory effect on NK cells (116,117). The precise mechanism behind how the regulation of NK cell stimulatory receptors influences the pathogenesis of EM requires further verification and examination.

IL-12 can regulate the immune recognition of NK cells in the endometrium. IL-12 is composed of two heterologous polypeptide chains, p40 and p35. A study found that the concentration of IL-12 in patients with EM is similar compared with that of healthy controls. However, increased levels of free p40 in the peritoneal fluid of these patients indicated that overexpression of the p40 subunit alone could reduce the cytotoxicity induced by IL-12 (118). The ratio of FCGR3-negative NK cells to FCGR3-positive NK cells in the peritoneal cavity of patients with EM was increased (119). Based on these findings, the increased expression levels of inhibitory receptors and ligands has a key role in the decline of NK cytotoxicity.

Platelets also regulate the function of NK cells (111). Platelets release TGF-β during retrograde menstruation in patients with EM, thereby suppressing the expression level of a stimulatory receptor of NK cells, NKG2D and reducing their cytotoxicity (Fig. 2) (120123).

NK cell-associated therapeutic targets for EM

Studies on the changes and mechanisms of NK cells in patients with EM provide a solid experimental basis for the targeted treatment of EM. The present focus of immunotherapy targeting NK cells is to restore their cytotoxicity. To reach this goal, three possible aspects should be considered: i) Blocking inhibitory receptors; ii) anti-inhibitory or stimulatory cytokine therapy; and iii) immune checkpoint therapy.

KIR2DL1, LILRB1/2 and CD94/NKG2a are inhibitory receptors that are overexpressed in the NK cells of women with EM. The ability to interfere with the binding of the inhibitory receptors and their ligands to improve the cytotoxicity of NK cells has been tested by multiple studies (124,125). Disruption of the inhibitory receptors on NK cells contributing to enhanced cytotoxicity was found in a human cell model treated with 5-aza-2′-deoxycytidine reported by Binyamin et al (124) in 2008. The study observed increased NK cell cytotoxicity when the inhibitor KIR2DL1 was applied to the NK cells of a healthy woman. It was also observed that blocking KIR2DL1 can enhance the effects of rituximab (an anti-CD20 monoclonal antibody known to recruit the immune system to attack and kill B cells) by increasing the cytotoxicity of NK cells (124,126). The study by Andre et al (125) targeted NKG2A on NK cells with monalizumab combined with cetuximab (an EGFR inhibitor) in patients with head and neck carcinoma, resulting in increased NK cell cytotoxicity (125). Whilst neither of these studies were based on EM, this blocking of inhibitory receptors therapy may be a promising treatment for EM and has been shown to be effective against some malignancies (125,127); however, further studies on their role in EM are required to make conclusive statements.

Previous studies tried to identify cytokines that affect the regulation of NK cell activity as new targets for immunotherapy. Some ILs (such as IL-2 and IL-12) and IFNs are NK cell stimulative cytokines. Intraperitoneal injection of IL-2 in surgically implanted EM rat models was found to be capable of recruiting leukocytes into EM lesions and reducing lesion size (128,129). However, existing studies on the therapeutic effects of IL-2 on EM are based in animal models, therefore, further studies and analysis are needed on the applicability to human patients. IL-12 is another important NK cell stimulative cytokine. Researchers pretreated NK cells with an IL-12 heterodimer to reduce the ratio of free p40 to IL-12 and enhance the cytotoxicity of NK cells in ectopic endometrial tissue, resulting in suppressed development of ectopic endometrial tissue. IL-12 is therefore considered a potential specific target for correcting the increase in free p40 levels in patients with EM (118). Type I IFNs, which include IFN-α2b, IFN-β1a and type II IFN (IFN-γ), can activate NK cells and enhance their cytotoxicity. However, to the best of our knowledge, no work has studied the feasibility of using the NK cell activating effects of IFNs to treat EM. The study by Dicitore et al (130) showed that IFN-β1a is superior to IFN-α2b at inhibiting the proliferation and migratory activities of endometrial stromal cells.

A case-control study by Wu et al (131) found that treatment with GnRH-a could restore the damaged immune function of the peritoneal fluid in patients with EM, and proposed their hypothesis according to their findings (131). In this study, women with EM who used GnRH-a long-term were found to have increased levels of the CD3CD69+ subpopulation of peripheral blood mononuclear cells and the CD3+CD69+/CD3+CD24+ subpopulation of activated T cells. It was suggested that the increased level of activated T cells induced by GnRH-a secreted increased levels of IL-2 and IFN-γ, which led to restoration of NK cell activity in the peritoneal fluid.

TGF-β suppresses the cytotoxicity of NK cells and the function of other immune cells (132,133). TGF-β secretion is upregulated in women with EM and is considered important to EM pathogenesis (134,135). Anti-TGF-β therapies are currently being evaluated clinically as treatments for malignancies and other diseases, such as diabetes. However, the results are generally unsatisfactory, reporting non-responsiveness and potential systematic side effects (136,137). There are also concerns that anti-TGF-β therapies would cause systematic suppression and result in severe systematic side effects due to the important role that TGF-β has in multiple vital signaling pathways, such as cell proliferation and differentiation. In vitro and in vivo studies are needed to further study anti-TGF-β therapy for EM.

Other immunotherapies may restore NK cell function in EM, most of which are based on immunotherapy models of other diseases. In 2004, the study by Clayton et al (138) first proposed the possibility of using Mycobacterium to restore NK cell activity in EM (138). However, the hypothesis was only supported by in vitro studies and further verification is required.

Immune checkpoint blocks (ICBs) are also new immunotherapies that researchers are currently evaluating. PD-1/PD-L1 pathway-associated inhibitors are a type of checkpoint blocking therapy. Previous studies have reported increased PD-1 expression levels in the peripheral blood cells of patients with EM, and increased PD-L1 expression levels in both the ectopic and non-ectopic endometrial tissues of patients with EM (139,140). This indicates that the peripheral tolerance caused by PD-1/PD-L1-induced T cell suppression may contribute to the immune abnormalities noted in EM. ICB therapy using PD-1/PD-L1 inhibitors is a promising treatment for preventing immune tolerance to EM (141143). However, studies have reported that PD-1/PD-L1 inhibitor treatment can lead to adverse reactions in a variety of tissues and organs throughout the body. The use of ICBs in the treatment of EM should therefore be carefully selected, and inhibitors with the strongest specificity for EM should be utilized (144,145). In addition to ICB therapy, the use of genetically modified NK cells, such as chimeric antigen receptor (CAR)-NK cells, in tumor immunotherapy has also attracted increased attention (146148). However, engineering a CAR-NK structure requires a biomarker specifically expressed on the surface of ectopic endometrial cells, which, to the best of our knowledge, has not yet been discovered.

Although the aforementioned targets for the treatment of EM have been supported in theory by in vitro and animal experiments, limited clinical trials have been reported.

Obstacles and future prospects

The exact mechanism behind how immunosuppression in ectopic endometrial tissue and its environment damages the cytotoxicity of NK cells is unclear, which makes it difficult to identify an appropriate immunotherapy target. Three main specific inhibitory NK cell receptor families have been identified: KIR, LILRB and NKG2. To the best of our knowledge, there are no reports on the inhibition of these receptor/ligand interactions. Cytokine therapy and the upregulation of associated activated receptors also requires further research. The possibility of utilizing immunotherapy in the treatment of EM needs further analysis due to the lack of tissue/cell specificity, which results in systemic side effects. It requires investigation on the epigenic differences between the ectopic and eutopic cells to develop treatments with increased specificity.

It should be noted that whether enhanced NK cell cytotoxicity is associated with abortion is debatable (149,150). Further examinations and analyses are needed before NK cell treatment can enter clinical research.

Macrophages

Phenotypes and function

Macrophages are a late differentiation cell type of the mononuclear-phagocyte system, which have an important role in both the non-specific and specific immune response. Macrophages were previously considered to be solely derived from blood monocytes, which are widely distributed and participate in the innate immunity of the body (151). This notion has changed due to the discovery of macrophages derived from and residing in specific tissues without the participation of circulating monocytes (152). Macrophages can be polarized into different directions based on the effects of different microenvironments and stimulating factors. Based on the surface markers of polarized macrophages and their functions, polarized macrophages can be categorized into two types: Classically activated macrophages (M1) and alternatively activated macrophages (M2) (153,154). M1 has a pro-inflammatory effect on the early stages of inflammation, phagocytizes and digests foreign pathogens, secretes pro-inflammatory factors, activates the T cell-dependent immune response and promotes the Th1 immune response. M2 can promote tissue repair and wound healing, regulates the Th2 immune response and contributes to disease recovery during the later stage of inflammation, which results in an anti-inflammatory effect (153,154).

There are two types of macrophages in the female pelvis: Endometrial and peritoneal. Endometrial macrophages (eMs) are involved in triggering and regulating the process of endometrial breakdown, and the subsequent repair of the endometrial functional layer by facilitating cell proliferation and angiogenesis (155,156). eMs function in the following three ways: i) Production and release of VEGF to promote angiogenesis; ii) participation in triggering and controlling the shedding process; and iii) facilitating and rebuilding the functional layer (157). Peritoneal macrophages (pMs) are distributed in ectopic endometrial tissues outside of the reproductive tract. The increased macrophages in patients with EM are mainly pMs. pMs have an immune monitoring role on the peritoneal surface. pMs can be classified into resident pMs and monocyte-derived pMs of bone marrow origin (158,159).

Based on the differences in MHCII and F4/80 expression levels, pMs can be divided into two phenotypes: Big, tissue-resident pMs and small, monocyte-derived pMs. Both types of macrophages can be either polarized into M1, which is pro-inflammatory, or M2, which is anti-inflammatory, depending on the stimulation of pathogen-associated molecular patterns (51). The pro-inflammatory M1 phenotype of pMs, similar to the classification of helper T cells, is activated mainly through the activation of IFN-γ, LPS, TNF-α or a combination of the three. The anti-inflammatory M2 phenotype of pM is mainly activated by IL-1, IL-10 and IL-13. The polarization of pMs produces corresponding molecular markers, which allows researchers to detect the regulation of macrophages (160162). Another type of macrophage, tumor-associated macrophage, has a role in the nutrition and angiogenesis of patients with EM and endometrial cancer (Fig. 3) (64).

Macrophages in EM

Although the number and activation of pMs in patients with EM are increased, the phagocytic capacity of these pMs is still unable to remove the ectopic endometrial tissue debris. pMs obtained from women with EM show a reduced capacity for phagocytosis due to the decreased expression level and activity of matrix metalloproteinase-9, which is regulated by prostaglandin E2 (PGE2) and is the enzyme that is necessary in the degradation of the extracellular matrix (163165).

Ectopic endometrial tissue is in a hormonal environment that contains abnormal concentrations of estrogen and androgens. The secretion of C-C motif chemokine ligand 2 (CCL2) by endometrial stromal cells is upregulated in the ectopic milieu, which has been confirmed to be mediated by estrogen (166). CCL2 mediates the polarization of macrophages to M2 instead of M1 (167). The abnormal EM environment also promotes the elevation of distinct anti-inflammatory phenotypes of macrophages, forming an immunosuppressive microenvironment by stimulating the proliferation of epithelial and stromal cells in endometriotic foci, and promoting angiogenesis (168). However, chronic inflammation is still observed in the lesion microenvironment. It could be possible that the upregulation of M2 is compensatory, induced by persistent inflammation and tissue repair. According to the macrophage depletion study by Bacci et al (169), anti-inflammatory M2 induced by macrophage colony-stimulating factor and IL-10 is considered to be of importance to the growth and development of ectopic EM tissue, while pro-inflammatory M1 induced by IFN-γ is capable of eliminating the ectopic tissue (169). The phenotypic plasticity of pMs makes it possible to investigate potential therapeutic targets for EM based on the suppression of the M2 phenotype in pMs or the activation of the M1 phenotype. The suppression of M2 polarization has already been proposed as chemical therapy for colon tumors, such as by using ovatodiolide to prevent the polarization of M2 tumor-associated macrophages (170).

Estrogen receptors on macrophages can be classified into surface receptors and nuclear receptors. Estrogen nuclear receptors can be divided into ER-α (ER1) and ER-β (ER2). ER2 promotes inflammation and disease progression by increasing the production of inflammatory cytokines, including IL-1β and IL-6 (171,172). IL-6 mediates the recruitment of monocytes and their differentiation into macrophages, which contributes to the increased macrophage infiltration into the EM lesions. ER2 also inhibits apoptosis by interacting with the NLRP3 sensor, caspase 1 and apoptosis signal regulated kinase-1 (173). However, chloroindazole, an ER2 ligand developed in 2015, can suppress inflammation and angiogenesis within the EM lesion, thereby suppressing EM progression (174). These data indicate that the activation of ER2 can also be anti-inflammatory and can serve as a possible target for EM treatment.

ER1 has two main roles. Firstly, ER1 promotes the secretion of pro-inflammatory cytokines, such as IFN-1, contributing to the inflammatory response (175,176). Secondly, ER1 activation also inhibits the NF-κB pathway, which limits the extent of the inflammation (177,178).

Upregulation of ER2 expression levels and downregulation of ER1 expression levels in macrophages and endometrial stromal cells will result in an extremely low ratio of ER1:ER2. There are controversies on whether the influence the ER1 deficiency and ER2 overexpression have an inflammatory or anti-inflammatory effect on the EM environment due to the opposing findings of previous studies (172,179). Despite these controversies, the consensus is that the dysregulation of estrogen and ERs contributes to inflammation in EM. The regulation of inflammatory pathways and immune cells by ERs and estrogen in endometriotic stromal cells will be discussed in further detail below.

Estrogen receptors on the cell surface are G protein-coupled ERs (GPERs), which are expressed on the surface of macrophages in this hormonal environment (180). GPERs are seven transmembrane-spanning receptors that bind to estrogen and mediate rapid non-genomic signaling pathways, such as the mitogen-activated protein kinases (MAPK) pathway and the phosphatidylinositide-3-kinases/Akt (PI3K/Akt) pathways, which can be initiated within seconds and rapidly induce a physiological response in target cells (181). GPER expression levels in macrophages within ectopic endometrium are increased, suggesting that this abnormality may be important to the regulation of the macrophage immune response (182). The roles of GPERs in EM have been gradually discovered and reported, making them a promising target for EM therapy. Activation of GPERs by their agonist G-1 can inhibit the secretion of TNF-α and IL-6 that was induced by LPS, resulting in an anti-inflammatory effect on GPER-expressing macrophages (183).

Continuously elevated estrogen levels also lead to the synthesis and secretion of inflammatory cytokines by macrophages, such as IL-1, IL-6 and TNF-α, which trigger a series of pro-inflammatory responses (173,184). Macrophages have a two-way response to estrogen: Upregulation of pro-inflammatory cytokines is induced by comparatively low concentrations of estrogen and inhibited by increased concentrations of estrogen (185). It has been hypothesized that the function of macrophages in ectopic endometrial tissue may be estrogen-dependent, and that estrogen may regulate the immune response through the GPERs and ERs of macrophages in ectopic endometrial tissue (182,186). These observations indicate that whether these estrogen-dependent events have a pro- or anti-inflammatory role on macrophages in EM depends on the types of ERs and the local concentration of estrogen within the lesion.

The chronic inflammatory environment in ectopic endometrial tissue triggers the secretion of inflammatory cytokines, such as IL-1β, IL-17A and TNF-α (187). Increased inflammatory cytokines can lead to the abnormal activation of the mTOR/PI3K signaling pathway, and the abnormal activation and proliferation of keratinocytes (188). Whether the same inference can be made on macrophages in EM requires further study. In addition, inflammatory cytokine IL-6 in inflamed tissue acts as a superior coordinator of protein synthesis capacity and cell growth rate by stimulating the translation of c-Myc mRNA, an oncogenic transcription factor that activates transcription via all three nuclear RNA polymerases. RNA polymerase I-associated transcription factors are recruited to rDNA by IL-6 when quiescent cells are stimulated to re-enter the cell cycle. Stimulated c-Myc mRNA will therefore eventually lead to an upregulation of rRNA transcription and enhanced proliferation of macrophages (189).

Macrophages are also involved in the induction of the immunosuppressive peritoneal environment in EM. IL-8 secreted by macrophages increases the expression level of Fas ligand (FasL) in endometrial cells, and the binding of FasL to Fas on T cells triggers apoptosis (190). It has been reported that the mRNA level of IL-8 in the peripheral blood and peritoneal fluid of patients with EM is considerably increased. The expression level of FasL on endometrial cells is also increased, which contributes to the formation of the immunosuppressed and immune tolerant microenvironment that is conducive to the adhesion of ectopic cells (191). Serum IL-8 levels have the potential to be an early indicator of EM (192). However, there seems to be little research assessing relevant immunotherapeutic targeting of Fas/FasL for EM. Other immunosuppressive cytokines are also secreted by macrophages in ectopic endometrial tissues, including IL-10 and TGF-β, leading to the inhibition of NK cells in the peritoneal cavity (Fig. 4) (59).

Similar to malignant tumors, the expression level of oncogenes and tumor suppressor genes in EM is considerably altered. Studies have shown that c-Myc, a recognized oncogene, is overexpressed in most patients with EM. It has been proposed that c-Myc also participates in the pathogenesis of EM (189,193).

In addition to the aforementioned inflammatory cytokines that have important roles in the pathogenesis of EM, Tie2-expressing macrophages in ectopic endometrial tissue inhibit endothelial cell apoptosis by preventing the caspase-3 activation of neovascular endothelial cells. This may also be used as a potential therapeutic target for EM (194).

EM is characterized by considerably increased levels of macrophage migration inhibitory factor (MIF), a multipotent protein that has a range of immune regulatory functions and is a key upstream regulator of both the non-specific and specific immune responses. During the onset of premenstrual syndrome, patients with EM have elevated MIF levels in the normal endometrium, early ectopic endometrium, peritoneal fluid and systemic circulation (195198). Studies have shown that MIF and its specific inhibitors can be used not only to improve the accuracy of EM diagnosis, but also to develop new therapeutic strategies against EM. The study by Seeber et al (199) found that the combined use of MIF and factors such as CA-125, monocyte chemoattractant protein 1 and leptin, can improve the accuracy of EM diagnosis to 93% (199). The concentration of macrophages in the normal endometrium of patients with EM is considerably reduced when compared with that of healthy controls, which predisposes patients to a poor prognosis (200).

It has been suggested that abnormal macrophage regulation may also be associated with the various clinical features of EM. For example, the increased concentration of pMs may disturb normal fertilization and lead to infertility in women with EM (201). Decreased insulin-like growth factor-1 (IGF-1) production by macrophages may also be associated with pelvic pain associated with EM (202).

In conclusion, patients with EM have increased macrophage levels and activation mediated by the mTOR/PI3K signaling pathway, c-Myc oncogene expression levels, and its resulting ribosome biogenesis in EM lesions and peritoneal fluid. Macrophages that co-exist with ectopic EM cells in vitro are immunosuppressive and macrophages in the peritoneal fluid of women with EM exist as a mix of both pro-inflammatory and anti-inflammatory cells. Anti-inflammatory macrophages in peritoneal fluid, which are M2, promote the development of EM lesions, while pro-inflammatory macrophages, which are M1, are antagonistic. Increased concentrations of estrogen in the ectopic EM microenvironment promote re-polarization from M1 to M2, which further contributes to the growth of the lesion.

Macrophage-associated therapeutic targets

Several theories for immune-associated therapies targeting the upregulation of pMs in EM have been proposed. The main hypotheses regarding the upregulation of pMs in the peritoneal fluid of patients with EM include the estrogen dependency theory, the mTOR/PI3K signaling pathway theory, overexpression of the oncogene c-Myc, ribosome biogenesis and the overexpression of MIF. Several potential medication therapies for managing these etiologies have been proposed.

Regarding the estrogen dependency theory, estrogen replacement and other associated therapies are being investigated and introduced into clinical management. The estrogen replacement therapy is currently the most commonly used method for treating EM that can achieve complete ovarian suppression, and has been in use since it was first reported in 1948 (203). Low-doses of combined estrogen and progestin or progestin alone can effectively relieve the clinical symptoms of pelvic pain caused by EM, and can also reduce the adverse effects of low estrogen that are induced by GnRH agonists (204). However, GnRH therapy can lead to several clinical side effects, including increased follicular development (205).

In addition to the estrogen-dependent theory, the mTOR/PI3K signaling pathway theory is also important, where mTOR/PI3K functions as the upstream regulator of ribosome biogenesis and plays a key role in protein synthesis (206). The mTOR/PI3K inhibitor GSK2126458 and the RNA polymerase 1 inhibitors CX5461 and BMH21 have been developed, all of which have shown very good therapeutic effects in a mouse model of human EM (206).

In terms of the MIF theory, the study by Khoufache et al (207) showed that the specific antagonist of MIF (ISO-1) can effectively reduce the growth and progression of ectopic endometrium, indicating that this agent has good clinical potential (207). The research and development of other associated drugs is still in the experimental stage and requires additional attention.

Complement system

Complement system in EM

The complement system is an indispensable part of the innate immune response and is involved in the identification and elimination of pathogens and abnormal cells, such as apoptotic and necrotic cells (208210). The complement system recognizes and tags pathogens and altered or transformed self-cells, thereby activating the inflammatory response and modulating the adaptive immune response, and ultimately leading to the lysis of target cells or pathogens (211). The complement system is a functionally complex system that can trigger a severe immune response or inflammatory process (211). This system may be harmful to the body when it is excessively or abnormally activated in conditions such as inflammation or tissue damage (212215). The complement system has a considerable role in peritoneal inflammation, which is associated with the early stages of EM (216).

The complement system is formed from a group of small proteins that demonstrate enzymatic activity after activation and exist in the serum and tissue fluid of healthy individuals and animals. The components of the complement system are extremely complex and variable. The study by Aslan et al (217) reported that 23 out of 84 immune response genes were upregulated in patients with EM, two of them considerably so. Some of these differentiating molecules were later confirmed to be members of the complement system (217).

Upregulated members of complement system in patients with EM

In a previous study, most components of the complement system that are associated with EM were found to be upregulated in patients with EM (218). To the best of our knowledge, a limited number of complement components were found to be decreased. Such decreased components included mannose/mannan binding lectin-associated serine protease-1 (MASP-1), and several remain controversial (219).

In a previous study, the quantities of C1q and C1INH in the peritoneal fluid of patients with EM at various stages were considerably increased compared with those in normal controls (220). Moreover, increased levels of C1q and C1INH were found in the peritoneal fluid of early stage EM (220). These results suggest that immunoglobulins participate in the initiation of the classical pathway in ectopic endometrial tissue, especially during early EM (220). Furthermore, C1-associated genes (including C1QA, C1QB, C1R and C1S) and C2 genes were increased in ectopic endometrial tissues compared with those in healthy controls (221). C3 is usually expressed in the ectopic tissue of patients with EM despite its regular expression levels in the glandular epithelium of normal endometrium (222,223). The overall C3 levels in the peritoneal fluid and peripheral blood of patients with EM were increased compared with those of normal controls, especially C3c and C3b (219,224227).

A growing body of evidence has reported that the C3 levels in the serum of patients with EM were considerably upregulated, in particular among patients with mild EM, when compared with that in healthy subjects and patients with severe EM (228231). Other studies found that the C3 levels in the eutopic endometrium of patients with EM were also considerably increased under the influence of ectopic endometrial tissue (228,232). Elevated iC3b in the peritoneal fluid of patients with EM may negatively regulate NK cell activity via the iC3b/CR3 signaling pathway, thereby downregulating NK cell cytotoxicity (219,233).

The levels of other members of the complement system, such as C5, C6, C7 and C8A, were also upregulated in the ectopic endometrial tissue of patients with EM (217,221). C6 levels were considerably increased in patients with early stage EM compared with those in their healthy counterparts (234). C7 was also upregulated in ectopic endometrial tissue (221).

The expression levels of complement factor (CF)B, CFD, CFH and CFI in the complement system are upregulated in ectopic EM tissue, while the expression level of MASP-1 is downregulated (Fig. 5).

Controversies

There are controversial theories about the relationship between C4 and EM. Studies have shown that the concentration of C4 in the peripheral blood and peritoneal secretions of women with EM are increased compared with those of normal controls (219,224,225). C4a was considerably decreased in the peritoneal fluid of patients with peritoneal and ovarian EM (218,235). The C4A/B gene expression level was upregulated in ectopic endometrial tissues, while complement component 4 binding protein α (C4BPA) expression level was reduced (221).

Two studies have examined the membrane attack complex (MAC; also known as SC5b-9) in patients with EM. One study found that the MAC levels in the peritoneal fluid and peripheral blood of patients with EM were increased. The concentrations of terminal complex were also increased in patients with advanced EM (219). However, another study found no considerable difference in the MAC level in the peritoneal fluid in patients with EM compared with that in normal controls (236,237).

The relationship between mannose-binding lectin (MBL) and EM is also controversial. Some studies have shown that equivalent MBL levels exist between normal controls and patients with EM (238,239), while the study by Sikora et al (220) observed an increased level of MBL in the peritoneal fluid of patients with EM. Furthermore, the concentration of MBL in patients with early EM was increased compared with that in patients with late EM (218,220,236,237,240).

Potential therapeutic targets associated with the complement system

Complement C3 inhibitors can interrupt the inflammatory cascade at its earliest stage and reduce the production of iC3b, which weakens NK cytotoxicity. The blockade of C5a and C3a to induce macrophage activation, C1q inducing the transformation of macrophages to the M2 phenotype and angiogenesis in EM lesions via complement immune therapy could all be promising targets for EM treatment (241,220).

Sex steroid hormones

Sex steroid hormone regulation and immune alteration in EM

Estrogen and progesterone are two key sex hormones that are closely associated with the occurrence and progression of EM. As previously discussed, estrogen mainly exerts its functions by interacting with ER and inducing an inflammatory environment. ER2 is associated with the inhibition of the inflammatory response. Increased ER2 activity can promote cell survival by inhibiting TNF1-mediated apoptosis, participating in growth factor signaling and promoting epithelial mesenchymal transition (242).

The close relationship between sex hormones such as estrogen and progesterone and the immune system has been frequently demonstrated. Estrogen can induce the activation of the immune response and immune cells through nuclear receptors. The dysregulation of estrogen and progesterone signaling in EM are termed estrogen dominance and progesterone resistance (243,244).

The binding of progesterone to progesterone receptor (PR) in epithelial and stromal cells inhibits epithelial cell proliferation and promotes decidualization (245). These effects of progesterone are achieved by the integration of the response through two functionally different subtypes of PR: PR-A and PR-B. These two subtypes share the same gene but have separate promoters, which makes their structure and function distinct from one another (246). PR-A is recognized as the initial driver of uterine PR function, while PR-B is key to progesterone-induced morphogenesis during pregnancy, and mainly improves progesterone reactivity by maintaining an appropriate ratio to PR-A (247).

Estrogen and its two nuclear receptor subtypes have already been briefly introduced in the preceding sections. Estrogen promotion of epithelial cell proliferation and endometrial stromal decidualization is also mediated by the binding of estrogen and its receptors. The two receptor subtypes, ER1 and ER2, are transcribed by different genes (248).

ER1 is expressed in most cells of the immune system, while ER2 is limited to certain cell types of some immune organs, such as lymphocytes in human lymph nodes, bone marrow and thymus. Therefore, ER1 has a stronger impact on the immune system than ER2 (220). Both ER subtypes are expressed in the endometrium, with the expression levels of ER1 outnumbering those of ER2 (249). ER1 also has a more important role in promoting the proliferation of endometrial epithelial cells, implantation and fertilization than ER2 (221). The response induced by estrogen binding to ER1 may be mediated by slower genomic signaling pathways such as the IGF-1-PI3K/AKT pathway (250). As aforementioned, ER2 is associated with the inhibition of the inflammatory response. Increased ER2 activity can promote cell survival by inhibiting TNF1-mediated apoptosis, participating in growth factor signaling and promoting epithelial mesenchymal transition (242).

In addition to the two aforementioned types of sex steroid hormones, the abnormal elevation of prostaglandins (another hormone that induces an inflammatory response) is also involved in the pathophysiological changes of EM. Increased PGE2 is detected in both the eutopic and ectopic endometrial tissues of patients with EM. PGE2 participates in the direct and indirect induction of pain through positive feedback with estradiol (E2). In this positive feedback loop, E2 activates cyclooxygenase II (COX2) to promote the production of PGE2, and the upregulation of PGE2 level in turn promotes the expression of steroidogenesis-associated genes and aromatase, thereby increasing E2 production (251). Sensitivity to IL-1β is important in the regulation of COX2, which contributes to the maintenance of sex hormone-associated inflammation in EM lesions (165).

Endometriotic changes of sex steroid hormone

Abnormal regulation of sex hormone nuclear receptors in patients with EM has been reported (252). The main dysregulations of sex steroid hormones in EM can be classified into two types: Estrogen dominance and progesterone resistance.

Estrogen dominance refers to estrogen-induced cell proliferation and inflammation. The estrogen response is primarily triggered by ER1 and ER2. These two receptors have different behaviors, and thus the expression level and ratio of these two receptors are important to determine effects of estrogen on EM. The ratio of ER1:ER2 is decreased in ectopic endometrial tissue of the ovary. This decreased ratio is caused by the upregulation of ER2 and the downregulation of ER1 due to changes in the methylation level of their promoters (249). Decreased methylation of the ER2 promoter leads to increased expression levels of ER2, while increased methylation of the ER1 promoter leads to decreased ER1 expression levels (251,253). In addition to epigenetic changes, crosstalk between ER1, ER2 and PRs has also been found to be important. ER2 directly downregulates the expression of ER1 by binding to the promotor region of ER1 (254). The downregulation of ER1 contributes to the reduction of PR and further promotes the development of EM and infertility (255).

ER2 upregulation in EM can activate a variety of proliferation- and inflammation-associated signaling pathways, such as the COX2-PGE2 feedback loop, which may be the main reason for increased lesion survival, cell proliferation and inflammation (249). Other research has found that ER2 can interact with inflammatory factors to regulate apoptosis and the inflammatory response, which is also associated with the pathogenesis of EM (172).

The other sex steroid hormone dysregulation in EM is progesterone resistance, in which normal and ectopic endometrial tissues in patients with EM do not respond to progesterone (256). Little is known about the mechanism behind progesterone resistance. Several studies have suggested that the downregulation of progesterone receptors may be a potential contributor to progesterone resistance, but this remains controversial (257,258).

Hormone therapies and therapeutic targets

Studies have shown that the hormonal treatment of EM is feasible (249,259). Current hormone therapies include GnRH agonists, aromatase inhibitors, COCs containing progesterone and E2, progestin-based therapies and androgen therapy (25,260,261). Hormone therapy aims to inhibit lesion growth or control pelvic pain by reducing the estrogen response and promoting the progesterone response (25,262). However, hormone therapy interferes with ovarian function, homeostasis and individual immunity, resulting in numerous side effects that include weight gain, androgen effects, reduced bone density, infertility and other adverse effects (262). Clinically, hormonal therapies are currently the most effective drugs for the treatment of EM (1,260). COCs are generally composed of a specific proportion of estrogens and progestogens that can inhibit steroid production in the ovary to treat chronic pelvic pain caused by ectopic endometrium (263). Although COCs are effective against the post-operative recurrence of EM, the estrogen contained in COCs still carries a risk of aggravating progestin resistance. Progestogen-based hormone therapy is another ideal treatment for EM. Medroxyprogesterone acetate (MPA) has been effective at reducing the pain caused by EM and reversing the decreased bone mineral density caused by low estrogen levels (264). A decrease in ER and an increase in PR in endometrial tissue can be found in patients with EM using MPA and other progestin-based drugs (265,266). GnRH agonists are second-line drugs after hormone therapy, which can reduce the production of estrogen, weaken E2 dominance and downregulate pituitary function through negative feedback (261). Although GnRH agonists are effective at alleviating the pain caused by EM, their side effects, such as bone loss, greatly limit their clinical use. As aforementioned, there are still several obstacles that block the treatment of EM with hormone therapy, including unresponsiveness to progesterone caused by progesterone resistance and the adverse effects of hypoestrogenemia.

P-selectin

The inflammatory and coagulation systems are the two main host defense systems. The coagulation system can be triggered by the inflammatory system (267,268). Inflammation is regulated by coagulation. P-selectin is a platelet adhesion molecule, whose expression levels are regulated by protein kinase C. Studies have found that its expression level is abnormal in patients with EM (269271). The study by Guo et al (272) reported that platelet aggregation was induced by P-selectin in ectopic endometrium, which promoted the proliferation and progression of the cell cycle for endometriotic stromal cells (272). Studies have found that P-selectin is also involved in leukocyte adhesion and inflammation (273,274). P-selectin is therefore considered a potential immune-associated therapeutic target. P-selectin can be targeted and blocked in several ways. For example, inclucumab is a highly specific recombinant human monoclonal antibody against P-selectin, and has been in clinical trials for the treatment of myocardial injury (275). The Fc fragment of recombinant P-selectin has also been tested in a mouse model of human EM, where it was effective without signs of bleeding complications (272). However, to the best of our knowledge, there are no reports of the clinical use of P-selectin antagonists or antibodies to treat EM.

Future perspectives

The treatment of EM still primarily uses hormone-regulating drugs, such as progesterone-based therapy, GnRH agonists, aromatase and COX inhibitors, and COC. Although clinical trials have shown the effectiveness of hormone therapy, patients still focus on its moderate or severe adverse effects, such as osteoporosis and sexual function inhibition. Studies have tested the effects of various compounds on EM, hoping that these compounds have therapeutic effects on EM without side effects. In the present review, several compounds are discussed that have been shown to be capable of improving symptoms of EM in animal experiments, clinical trials or both. Dienogest is a derivative of 19-nortestosterone, which serves the dual roles of anti-ovulation and anti-proliferation against endometrial cells, which can effectively relieve EM symptoms without the side effects of estrogen and androgens (276). Clinical randomized controlled trials at different stages have been carried out in Europe and Japan, whose results show that Dienogest is superior to other progesterone drugs in terms of efficacy, safety, receptor selectivity and tolerance in the treatment of EM (277,278). Although Dienogest has numerous advantages over other hormone drugs, severe bleeding seems to be a potential serious side effect.

Beyond hormone-associated therapy, researchers are also committed to studying and developing drugs targeting other immune-associated factors to treat of EM. Since the first anti-complement drug eculizumab (anti-C5 antibody) was approved by the US Food and Drug Administration for the treatment of paroxysmal nocturnal hemoglobinuria in 2007, more complement pathway blocking drugs have been fully developed (279).

In addition to studying drugs targeting immune system factors, researchers are also studying the utility of ribosome biosynthesis (associated with macrophage proliferation) in the treatment of EM. Chang et al (206) tested the potential of using ribosome biogenesis inhibitors targeting mTOR/PI3K and RNA polymerase I as an alternative to the treatment of EM in an animal study in 2022. The results showed that the ribosome biogenesis inhibitor could inhibit inflammation, reduce neutrophils in the peritoneal fluid and relieve pain in the treatment of an EM mouse model, which confirmed its therapeutic potential (206).

In previous decades, there has been a push towards efforts to find other potential targets for the treatment of EM. It has become a consensus that local and systemic changes to immune cells and immune-associated factors are important to the pathogenesis and development of EM. More attention should be paid to the development of drugs that target the components of the immune system. To the best of our knowledge, numerous side effects can be avoided by immunotherapy, which should be the direction of future research on EM treatment. Immunotherapy targeting NK cells and macrophages is in the preclinical trial stage, which may inspire other researchers to seek improved immune-associated solutions.

Conclusion

Ectopic endometrial tissue in patients with EM is a clone of ectopic proliferating endometrial cells in the immune microenvironment of the inflammatory response, which is characterized by increased estrogen pro-inflammatory cytokine levels and alterations to the immune cell infiltration spectrum. The pathogenesis of EM remains unclear, and it is relatively difficult to find and select a satisfactory treatment for this disease. To the best of our knowledge, there is no treatment plan that can completely cure EM. At present, the treatment of EM is mainly symptomatic, and includes reducing pain, avoiding infertility and delaying recurrence as much as possible. Compared with the inefficiency of medical symptomatic treatment, laparoscopic surgery is still the first choice for patients with EM of childbearing age due to its high postoperative pregnancy rate. However, as with all surgeries, conservative surgery in patients with EM may only require partial ovariectomy. Hysterectomy is occasionally required, but there is a risk of over-operation or premature ovarian failure. Hormone therapy for EM is not ideal and is usually accompanied by side effects. Thus, although there are limited studies on the clinical application and evaluation of immune targeted therapy and personalized therapy for EM, it is still necessary to further investigate this area.

The present review discussed the role of five major factors (NK cells, macrophages, the complement system, sex steroid hormones and P-selectin), and summarized their functions, regulation and association with EM. Several potential therapeutic targets for EM have also been summarized, whether they are in the hypothetical stage or established by animal experiments. It is hoped that through the present review, more attention can be given to EM and its potential therapeutic targets, further advancing EM treatment methods.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant no. 81672861) and the Science and Technology Development Plan of Shandong Province (grant no. 2017GSF218029).

Availability of data and materials

Not applicable.

Author's contributions

WZ, KL and XZ designed the research, WZ and KL collected and analyzed the reference articles, and wrote the draft. GZ and AJ designed the figures, XZ and GZ critically revised the manuscript. All authors contributed to manuscript revision, and 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 

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

2 

Taylor HS, Kotlyar AM and Flores VA: Endometriosis is a chronic systemic disease: Clinical challenges and novel innovations. Lancet. 397:839–582. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Williams EA: Endometriosis in Clinical-practice-surgical aspects. Irish J Med Sci. 152:14–17. 1983. View Article : Google Scholar : PubMed/NCBI

4 

Van Gorp T, Amant F, Neven P, Vergote I and Moerman P: Endometriosis and the development of malignant tumours of the pelvis. A review of literature. Best Pract Res Clin Obstet Gynaecol. 18:349–371. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Indrielle-Kelly T, Fruhauf F, Burgetova A, Fanta M and Fischerova D: Diagnosis of endometriosis 3rd Part-ultrasound diagnosis of deep endometriosis. Ceska Gynekol. 84:269–275. 2019.PubMed/NCBI

6 

Fukunaga M, Nomura K, Ishikawa E and Ushigome S: Ovarian atypical endometriosis: Its close association with malignant epithelial tumours. Histopathology. 30:249–255. 1997. View Article : Google Scholar : PubMed/NCBI

7 

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. View Article : Google Scholar : PubMed/NCBI

8 

He Y, Cao B and Huang Y: Effect of endometriosis on prognosis of ovarian clear cell carcinoma: A 10-year retrospective study. Front Oncol. 14:14383092024. View Article : Google Scholar : PubMed/NCBI

9 

Anglesio MS, Bashashati A, Wang YK, Senz J, Ha G, Yang W, Aniba MR, Prentice LM, Farahani H, Li Chang H, et al: Multifocal endometriotic lesions associated with cancer are clonal and carry a high mutation burden. J Pathol. 236:201–209. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Czernobilsky B and Morris WJ: A histologic study of ovarian endometriosis with emphasis on hyperplastic and atypical changes. Obstet Gynecol. 53:318–323. 1979.PubMed/NCBI

11 

LaGrenade A and Silverberg SG: Ovarian tumors associated with atypical endometriosis. Hum Pathol. 19:1080–1084. 1988. View Article : Google Scholar : PubMed/NCBI

12 

Nouri B, Hashemi SH, J Ghadimi D, Roshandel S and Akhlaghdoust M: Machine Learning-based detection of endometriosis: A retrospective study in a population of iranian female patients. Int J Fertil Steril. 18:362–366. 2024.PubMed/NCBI

13 

Moradi Y, Shams-Beyranvand M, Khateri S, Gharahjeh S, Tehrani S, Varse F, Tiyuri A and Najmi Z: A systematic review on the prevalence of endometriosis in women. Indian J Med Res. 154:446–454. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Mafra F, Catto M, Bianco B, Barbosa CP and Christofolini D: Association of Wnt4 polymorphisms with endometriosis in infertile patients. J Assist Reprod Genet. 32:1359–1364. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Santulli P, Bourdon M, Presse M, Gayet V, Marcellin L, Prunet C, de Ziegler D and Chapron C: Endometriosis-Related infertility: Assisted reproductive technology has no adverse impact on pain or quality-of-life scores. Fertil Steril. 105:978–987.e4. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Ferrero S, Stabilini C, Barra F, Clarizia R, Roviglione G and Ceccaroni M: Bowel resection for intestinal endometriosis. Best Pract Res Clin Obstet Gynaecol. 71:114–128. 2021. View Article : Google Scholar : PubMed/NCBI

17 

Chapron C, Marcellin L, Borghese B and Santulli P: Rethinking mechanisms, diagnosis and management of endometriosis. Nat Rev Endocrinol. 15:666–682. 2019. View Article : Google Scholar : PubMed/NCBI

18 

Ferrero S, Barra F, Scala C and Condous G: Ultrasonography for bowel endometriosis. Best Pract Res Clin Obstet Gynaecol. 71:38–50. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Geng JH and Lee YC: Bladder endometriosis. N Engl J Med. 381:e432019. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Brown J, Crawford TJ, Allen C, Hopewell S and Prentice A: Nonsteroidal anti-inflammatory drugs for pain in women with endometriosis. Cochrane Database Syst Rev. 1:CD0047532017.PubMed/NCBI

22 

Streuli I, de Ziegler D, Santulli P, Marcellin L, Borghese B, Batteux F and Chapron C: An update on the pharmacological management of endometriosis. Expert Opin Pharmacother. 14:291–305. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Wu Y, Liu Y, Jia H, Luo C and Chen H: Treatment of endometriosis with dienogest in combination with traditional Chinese medicine: A systematic review and meta-analysis. Front Surg. 9:9924902022. View Article : Google Scholar : PubMed/NCBI

24 

Chinese Medical Association Obstetrics, Gynecology Branch, Endometriosis Professional Committee and Chinese Medical Association Obstetrics and Gynecology Branch Endometriosis Collaborative Group, . Long-term management of endometriosis: Chinese expert consensus. Chin J Obstet Gynecol. 53:836–841. 2018.(In Chinese).

25 

Endometriosis Collaborative Group of the Obstetrics and Gynecology Branch of the Chinese Medical Association, . Guidelines for the diagnosis and treatment of endometriosis. Chin J Obstet Gynecol. 3:161–169. 2015.

26 

Vercellini P, Viganò P, Somigliana E and Fedele L: Endometriosis: Pathogenesis and treatment. Nat Rev Endocrinol. 10:261–275. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Barbieri RL: Stenosis of the external cervical os: An association with endometriosis in women with chronic pelvic pain. Fertil Steril. 70:571–573. 1998. View Article : Google Scholar : PubMed/NCBI

28 

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

29 

Halme J, Hammond MG, Hulka JF, Raj SG and Talbert LM: Retrograde menstruation in healthy women and in patients with endometriosis. Obstet Gynecol. 64:151–154. 1984.PubMed/NCBI

30 

Herington JL, Bruner-Tran KL, Lucas JA and Osteen KG: Immune interactions in endometriosis. Expert Rev Clin Immunol. 7:611–626. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Kyama CM, Debrock S, Mwenda JM and D'Hooghe TM: Potential involvement of the immune system in the development of endometriosis. Reprod Biol Endocrinol. 1:1232003. View Article : Google Scholar : PubMed/NCBI

32 

Christodoulakos G, Augoulea A, Lambrinoudaki I, Sioulas V and Creatsas G: Pathogenesis of endometriosis: The role of defective ‘immunosurveillance’. Eur J Contracept Reprod Health Care. 12:194–202. 2007. View Article : Google Scholar : PubMed/NCBI

33 

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. View Article : Google Scholar : PubMed/NCBI

34 

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

35 

Tanbo T and Fedorcsak P: Endometriosis-associated infertility: Aspects of pathophysiological mechanisms and treatment options. Acta Obstet Gynecol Scand. 96:659–667. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Gibson DA, Simitsidellis I, Collins F and Saunders PTK: Androgens, oestrogens and endometrium: A fine balance between perfection and pathology. J Endocrinol. 246:R75–R93. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Mohammed Rasheed HA and Hamid P: Inflammation to infertility: Panoramic view on endometriosis. Cureus. 12:e115162020.PubMed/NCBI

38 

Halis G and Arici A: Endometriosis and Inflammation in Infertility. Ann N Y Acad Sci. 1034:300–315. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Kang YJ, Jeung IC, Park A, Park YJ, Jung H, Kim TD, Lee HG, Choi I and Yoon SR: An increased level of IL-6 suppresses NK cell activity in peritoneal fluid of patients with endometriosis via regulation of SHP-2 expression. Hum Reprod. 29:2176–2189. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Bedaiwy MA, Falcone T, Sharma RK, Goldberg JM, Attaran M, Nelson DR and Agarwal A: Prediction of endometriosis with serum and peritoneal fluid markers: A prospective controlled trial. Hum Reprod. 17:426–431. 2002. View Article : Google Scholar : PubMed/NCBI

41 

Vilas Boas L, Bezerra Sobrinho C, Rahal D, Augusto Capellari C, Skare T and Nisihara R: Antinuclear antibodies in patients with endometriosis: A cross-sectional study in 94 patients. Hum Immunol. 83:70–73. 2022. View Article : Google Scholar : PubMed/NCBI

42 

Dias JA Jr, de Oliveira RM and Abrao MS: Antinuclear antibodies and endometriosis. Int J Gynaecol Obstet. 93:262–263. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Malinowski A, Szpakowski M, Wilczynski J, Banasik M and Puchala B: Occurrence of antinuclear antibodies in women with endometriosis and unexplained infertility. Ginekol Pol. 66:420–424. 1995.(In Polish). PubMed/NCBI

44 

Swann JB and Smyth MJ: Immune surveillance of tumors. J Clin Invest. 117:1137–1146. 2007. View Article : Google Scholar : PubMed/NCBI

45 

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

46 

Podgaec S, Abrao MS, Dias JA Jr, Rizzo LV, de Oliveira RM and Baracat EC: Endometriosis: An inflammatory disease with a Th2 immune response component. Hum Reprod. 22:1373–1379. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Olkowska-Truchanowicz J, Bocian K, Maksym RB, Bialoszewska A, Wlodarczyk D, Baranowski W, Ząbek J, Korczak-Kowalska G and Malejczyk J: CD4+ CD25+ FOXP3+ regulatory T cells in peripheral blood and peritoneal fluid of patients with endometriosis. Hum Reprod. 28:119–124. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Tanaka E, Sendo F, Kawagoe S and Hiroi M: Decreased natural killer cell activity in women with endometriosis. Gynecol Obstet Invest. 34:27–30. 1992. View Article : Google Scholar : PubMed/NCBI

49 

Thiruchelvam U, Wingfield M and O'Farrelly C: Natural killer cells: Key players in endometriosis. Am J Reprod Immunol. 74:291–301. 2015. View Article : Google Scholar : PubMed/NCBI

50 

Sciezynska A, Komorowski M, Soszynska M and Malejczyk J: Nk cells as potential targets for immunotherapy in endometriosis. J Clin Med. 8:172019. View Article : Google Scholar

51 

Artemova D, Vishnyakova P, Khashchenko E, Elchaninov A, Sukhikh G and Fatkhudinov T: Endometriosis and cancer: Exploring the role of macrophages. Int J Mol Sci. 22:51962021. View Article : Google Scholar : PubMed/NCBI

52 

Wu MH, Chen KF, Lin SC, Lgu CW and Tsai SJ: Aberrant expression of leptin in human endometriotic stromal cells is induced by elevated levels of hypoxia inducible factor-1alpha. Am J Pathol. 170:590–598. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Semenza GL: Hif-1 and mechanisms of hypoxia sensing. Curr Opin Cell Biol. 13:167–171. 2001. View Article : Google Scholar : PubMed/NCBI

54 

Becker CM, Rohwer N, Funakoshi T, Cramer T, Bernhardt W, Birsner A, Folkman J and D'Amato RJ: 2-Methoxyestradiol inhibits hypoxia-inducible factor-1{alpha} and suppresses growth of lesions in a mouse model of endometriosis. Am J Pathol. 172:534–544. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Hsiao KY, Chang N, Lin SC, Li YH and Wu MH: Inhibition of dual specificity Phosphatase-2 by hypoxia promotes interleukin-8-mediated angiogenesis in endometriosis. Hum Reprod. 29:2747–2755. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Hsiao KY, Chang N, Tsai JL, Lin SC, Tsai SJ and Wu MH: Hypoxia-inhibited DUSP2 expression promotes IL-6/STAT3 signaling in endometriosis. Am J Reprod Immunol. 78:2017.doi: 10.1111/aji.12690. View Article : Google Scholar : PubMed/NCBI

57 

Sharkey AM, Day K, McPherson A, Malik S, Licence D, Smith SK and Charnock-Jones DS: Vascular endothelial growth factor expression in human endometrium is regulated by hypoxia. J Clin Endocrinol Metab. 85:402–409. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Kupker W, Schultze-Mosgau A and Diedrich K: Paracrine changes in the peritoneal environment of women with endometriosis. Hum Reprod Update. 4:719–723. 1998. View Article : Google Scholar : PubMed/NCBI

59 

Yang HL, Zhou WJ, Chang KK, Mei J, Huang LQ, Wang MY, Meng Y, Ha SY, Li DJ and Li MQ: The crosstalk between endometrial stromal cells and macrophages impairs cytotoxicity of NK cells in endometriosis by secreting IL-10 and TGF-β. Reproduction. 154:815–825. 2017. View Article : Google Scholar : PubMed/NCBI

60 

El Hafny-Rahbi B, Brodaczewska K, Collet G, Majewska A, Klimkiewicz K, Delalande A, Grillon C and Kieda C: Tumour angiogenesis normalized by Myo-inositol trispyrophosphate alleviates hypoxia in the microenvironment and promotes antitumor immune response. J Cell Mol Med. 25:3284–3299. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Thiruchelvam U, Dransfield I, Saunders PT and Critchley HO: The importance of the macrophage within the human endometrium. J Leukoc Biol. 93:217–225. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Mehedintu C, Plotogea MN, Ionescu S and Antonovici M: Endometriosis still a challenge. J Med Life. 7:349–357. 2014.PubMed/NCBI

63 

Matarese G, De Placido G, Nikas Y and Alviggi C: Pathogenesis of endometriosis: Natural immunity dysfunction or autoimmune disease? Trends Mol Med. 9:223–228. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Hogg C, Horne AW and Greaves E: Endometriosis-associated macrophages: Origin, phenotype, and function. Front Endocrinol (Lausanne). 11:72020. View Article : Google Scholar : PubMed/NCBI

65 

Capobianco A and Rovere-Querini P: Endometriosis, a disease of the macrophage. Front Immunol. 4:92013. View Article : Google Scholar : PubMed/NCBI

66 

Colette S and Donnez J: Are aromatase inhibitors effective in endometriosis treatment? Expert Opin Investig Drugs. 20:917–931. 2011. View Article : Google Scholar : PubMed/NCBI

67 

Check JH: Chronic pelvic pain syndromes-Traditional and novel therapies: Part I surgical therapy. Clin Exp Obstet Gynecol. 38:10–13. 2011.PubMed/NCBI

68 

Sivori S, Vitale M, Morelli L, Sanseverino L, Augugliaro R, Bottino C, Moretta L and Moretta A: P46, a novel natural killer cell-specific surface molecule that mediates cell activation. J Exp Med. 186:1129–1136. 1997. View Article : Google Scholar : PubMed/NCBI

69 

Freud AG, Zhao S, Wei S, Gitana GM, Molina-Kirsch HF, Atwater SK and Natkunam Y: Expression of the activating receptor, NKp46 (CD335), in human natural killer and T-cell neoplasia. Am J Clin Pathol. 140:853–866. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Cooper MA, Fehniger TA and Caligiuri MA: The biology of human natural killer-cell subsets. Trends Immunol. 22:633–640. 2001. View Article : Google Scholar : PubMed/NCBI

71 

Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, Carson WE and Caligiuri MA: Human natural killer cells: A unique innate immunoregulatory role for the CD56(bright) subset. Blood. 97:3146–3151. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Hamerman JA, Ogasawara K and Lanier LL: NK cells in innate immunity. Curr Opin Immunol. 17:29–35. 2005. View Article : Google Scholar : PubMed/NCBI

73 

Vivier E, Tomasello E, Baratin M, Walzer T and Ugolini S: Functions of natural killer cells. Nat Immunol. 9:503–510. 2008. View Article : Google Scholar : PubMed/NCBI

74 

Freud AG, Mundy-Bosse BL, Yu J and Caligiuri MA: The broad spectrum of human natural killer cell diversity. Immunity. 47:820–833. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Reeves RK, Li H, Jost S, Blass E, Li H, Schafer JL, Varner V, Manickam C, Eslamizar L, Altfeld M, et al: Antigen-specific NK cell memory in rhesus macaques. Nat Immunol. 16:927–932. 2015. View Article : Google Scholar : PubMed/NCBI

76 

Hammer Q, Ruckert T and Romagnani C: Natural killer cell specificity for viral infections. Nat Immunol. 19:800–808. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Nikzad R, Angelo LS, Aviles-Padilla K, Le DT, Singh VK, Bimler L, Vukmanovic-Stejic M, Vendrame E, Ranganath T, Simpson L, et al: Human natural killer cells mediate adaptive immunity to viral antigens. Sci Immunol. 4:eaat81162019. View Article : Google Scholar : PubMed/NCBI

78 

Morvan MG and Lanier LL: Nk cells and cancer: You can teach innate cells new tricks. Nat Rev Cancer. 16:7–19. 2016. View Article : Google Scholar : PubMed/NCBI

79 

Chiossone L, Dumas PY, Vienne M and Vivier E: Natural killer cells and other innate lymphoid cells in cancer. Nat Rev Immunol. 18:671–688. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Raulet DH and Vance RE: Self-tolerance of natural killer cells. Nat Rev Immunol. 6:520–531. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Karre K, Ljunggren HG, Piontek G and Kiessling R: Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature. 319:675–678. 1986. View Article : Google Scholar : PubMed/NCBI

82 

Viant C, Fenis A, Chicanne G, Payrastre B, Ugolini S and Vivier E: SHP-1-mediated inhibitory signals promote responsiveness and anti-Tumour functions of natural killer cells. Nat Commun. 5:51082014. View Article : Google Scholar : PubMed/NCBI

83 

van der Touw W, Chen HM, Pan PY and Chen SH: LILRB Receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol Immunother. 66:1079–1087. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Stojanovic A, Correia MP and Cerwenka A: The NKG2D/NKG2DL axis in the crosstalk between lymphoid and myeloid cells in health and disease. Front Immunol. 9:8272018. View Article : Google Scholar : PubMed/NCBI

85 

Zingoni A, Molfetta R, Fionda C, Soriani A, Paolini R, Cippitelli M, Cerboni C and Santoni A: NKG2D and its ligands: ‘One for all, all for one’. Front Immunol. 9:4762018. View Article : Google Scholar : PubMed/NCBI

86 

Ferlazzo G, Thomas D, Lin SL, Goodman K, Morandi B, Muller WA, Moretta A and Münz C: The abundant NK cells in human secondary lymphoid tissues require activation to express killer cell Ig-like receptors and become cytolytic. J Immunol. 172:1455–1462. 2004. View Article : Google Scholar : PubMed/NCBI

87 

Bryceson YT, March ME, Ljunggren HG and Long EO: Synergy among receptors on resting NK cells for the activation of natural cytotoxicity and cytokine secretion. Blood. 107:159–166. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Masztalerz A, Van Rooijen N, Den Otter W and Everse LA: Mechanisms of Macrophage cytotoxicity in IL-2 and IL-12 mediated tumour regression. Cancer Immunol Immunother. 52:235–242. 2003. View Article : Google Scholar : PubMed/NCBI

89 

Mukherjee S, Jensen H, Stewart W, Stewart D, Ray WC, Chen SY, Nolan GP, Lanier LL and Das J: In silico modeling identifies CD45 as a regulator of IL-2 synergy in the NKG2D-mediated activation of immature human NK cells. Sci Signal. 10:eaai90622017. View Article : Google Scholar : PubMed/NCBI

90 

Fehniger TA and Caligiuri MA: Interleukin 15: Biology and relevance to human disease. Blood. 97:14–32. 2001. View Article : Google Scholar : PubMed/NCBI

91 

Kasaian MT, Whitters MJ, Carter LL, Lowe LD, Jussif JM, Deng B, Johnson KA, Witek JS, Senices M, Konz RF, et al: Il-21 limits NK cell responses and promotes Antigen-specific T cell activation: A mediator of the transition from innate to adaptive immunity. Immunity. 16:559–569. 2002. View Article : Google Scholar : PubMed/NCBI

92 

Tarannum M and Romee R: Cytokine-induced memory-like natural killer cells for cancer immunotherapy. Stem Cell Res Ther. 12:5922021. View Article : Google Scholar : PubMed/NCBI

93 

Marcus A, Mao AJ, Lensink-Vasan M, Wang L, Vance RE and Raulet DH: Tumor-derived cGAMP triggers a STING-mediated interferon response in non-tumor cells to activate the NK cell response. Immunity. 49:754–763.e4. 2018. View Article : Google Scholar : PubMed/NCBI

94 

Lettau M, Paulsen M, Schmidt H and Janssen O: Insights into the molecular regulation of fasl (CD178) biology. Eur J Cell Biol. 90:456–466. 2011. View Article : Google Scholar : PubMed/NCBI

95 

Prager I and Watzl C: Mechanisms of natural killer Cell-mediated cellular cytotoxicity. J Leukoc Biol. 105:1319–1329. 2019. View Article : Google Scholar : PubMed/NCBI

96 

Gazvani R and Templeton A: Peritoneal environment, cytokines and angiogenesis in the pathophysiology of endometriosis. Reproduction. 123:217–226. 2002. View Article : Google Scholar : PubMed/NCBI

97 

Ulukus M and Arici A: Immunology of endometriosis. Minerva Ginecol. 57:237–248. 2005.PubMed/NCBI

98 

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

99 

Oosterlynck DJ, Meuleman C, Waer M, Vandeputte M and Koninckx PR: The natural killer activity of peritoneal fluid lymphocytes is decreased in women with endometriosis. Fertil Steril. 58:290–295. 1992. View Article : Google Scholar : PubMed/NCBI

100 

Maeda N, Izumiya C, Kusum T, Masumoto T, Yamashita C, Yamamoto Y, Oguri H and Fukaya T: Killer inhibitory receptor CD158a overexpression among natural killer cells in women with endometriosis is undiminished by laparoscopic surgery and gonadotropin releasing hormone agonist treatment. Am J Reprod Immunol. 51:364–372. 2004. View Article : Google Scholar : PubMed/NCBI

101 

Vigano P, Vercellini P, Di Blasio AM, Colombo A, Candiani GB and Vignali M: Deficient antiendometrium Lymphocyte-mediated cytotoxicity in patients with endometriosis. Fertil Steril. 56:894–899. 1991. View Article : Google Scholar : PubMed/NCBI

102 

Lotze MT and Rosenberg SA: Results of clinical trials with the administration of interleukin 2 and adoptive immunotherapy with activated cells in patients with cancer. Immunobiology. 172:420–437. 1986. View Article : Google Scholar : PubMed/NCBI

103 

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

104 

Dias JA Jr, Podgaec S, de Oliveira RM, Carnevale Marin ML, Baracat EC and Abrao MS: Patients with endometriosis of the rectosigmoid have a higher percentage of natural killer cells in peripheral blood. J Minim Invasive Gynecol. 19:317–324. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Oosterlynck DJ, Meuleman C, Lacquet FA, Waer M and Koninckx PR: Flow cytometry analysis of lymphocyte subpopulations in peritoneal fluid of women with endometriosis. Am J Reprod Immunol. 31:25–31. 1994. View Article : Google Scholar : PubMed/NCBI

106 

Vernet-Tomas Mdel M, Perez-Ares CT, Verdu N, Molinero JL, Fernandez-Figueras MT and Carreras R: The endometria of patients with endometriosis show higher expression of Class I human leukocyte antigen than the endometria of healthy women. Fertil Steril. 85:78–83. 2006. View Article : Google Scholar : PubMed/NCBI

107 

Wu MY, Yang JH, Chao KH, Hwang JL, Yang YS and Ho HN: Increase in the expression of killer cell inhibitory receptors on peritoneal natural killer cells in women with endometriosis. Fertil Steril. 74:1187–1191. 2000. View Article : Google Scholar : PubMed/NCBI

108 

Matsuoka S, Maeda N, Izumiya C, Yamashita C, Nishimori Y and Fukaya T: Expression of inhibitory-motif killer immunoglobulin-like receptor, KIR2DL1, is increased in natural killer cells from women with pelvic endometriosis. Am J Reprod Immunol. 53:249–254. 2005. View Article : Google Scholar : PubMed/NCBI

109 

Maeda N, Izumiya C, Oguri H, Kusume T, Yamamoto Y and Fukaya T: Aberrant expression of intercellular adhesion Molecule-1 and killer inhibitory receptors induces immune tolerance in women with pelvic endometriosis. Fertil Steril. 77:679–683. 2002. View Article : Google Scholar : PubMed/NCBI

110 

Galandrini R, Porpora MG, Stoppacciaro A, Micucci F, Capuano C, Tassi I, Di Felice A, Benedetti-Panici P and Santoni A: Increased frequency of human leukocyte Antigen-E inhibitory receptor Cd94/Nkg2a-Expressing peritoneal natural killer cells in patients with endometriosis. Fertil Steril. 89 (5 Suppl):S1490–S1496. 2008. View Article : Google Scholar : PubMed/NCBI

111 

Nowak I, Ploski R, Barcz E, Dziunycz P, Kaminski P, Kostrzewa G, Milewski Ł, Roszkowski PI, Senitzer D, Malejczyk J and Kuśnierczyk P: KIR2DS5 in the presence of HLA-C C2 protects against endometriosis. Immunogenetics. 67:203–209. 2015. View Article : Google Scholar : PubMed/NCBI

112 

Guo SW, Du Y and Liu X: Platelet-derived Tgf-Beta1 mediates the Down-modulation of Nkg2d expression and may be responsible for impaired natural Killer (Nk) cytotoxicity in women with endometriosis. Hum Reprod. 31:1462–1474. 2016. View Article : Google Scholar : PubMed/NCBI

113 

Bellelis P, Frediani Barbeiro D, Gueuvoghlanian-Silva BY, Kalil J, Abrao MS and Podgaec S: Interleukin-15 and Interleukin-7 Are the major cytokines to maintain endometriosis. Gynecol Obstet Invest. 84:435–444. 2019. View Article : Google Scholar : PubMed/NCBI

114 

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. View Article : Google Scholar : PubMed/NCBI

115 

Gonzalez-Foruria I, Santulli P, Chouzenoux S, Carmona F, Batteux F and Chapron C: Soluble ligands for the NKG2D receptor are released during endometriosis and correlate with disease Severity. PLoS One. 10:e01199612015. View Article : Google Scholar : PubMed/NCBI

116 

Salih HR, Rammensee HG and Steinle A: Cutting edge: Down-regulation of mica on human tumors by proteolytic shedding. J Immunol. 169:4098–4102. 2002. View Article : Google Scholar : PubMed/NCBI

117 

Salih HR, Goehlsdorf D and Steinle A: Release of micb molecules by tumor cells: Mechanism and soluble micb in sera of cancer patients. Hum Immunol. 67:188–195. 2006. View Article : Google Scholar : PubMed/NCBI

118 

Mazzeo D, Vigano P, Di Blasio AM, Sinigaglia F, Vignali M and Panina-Bordignon P: Interleukin-12 and Its Free P40 subunit regulate immune recognition of endometrial cells: Potential role in endometriosis. J Clin Endocrinol Metab. 83:911–916. 1998. View Article : Google Scholar : PubMed/NCBI

119 

Mei J, Zhou WJ, Zhu XY, Lu H, Wu K, Yang HL, Fu Q, Wei CY, Chang KK, Jin LP, et al: Suppression of Autophagy and HCK Signaling Promotes PTGS2high FCGR3 NK cell differentiation triggered by ectopic endometrial stromal cells. Autophagy. 14:1376–1397. 2018. View Article : Google Scholar : PubMed/NCBI

120 

Palumbo JS, Talmage KE, Massari JV, La Jeunesse CM, Flick MJ, Kombrinck KW, Hu Z, Barney KA and Degen JL: Tumor cell-associated tissue factor and circulating hemostatic factors cooperate to increase metastatic potential through natural killer cell-dependent and-independent mechanisms. Blood. 110:133–141. 2007. View Article : Google Scholar : PubMed/NCBI

121 

Nieswandt B, Hafner M, Echtenacher B and Mannel DN: Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 59:1295–1300. 1999.PubMed/NCBI

122 

Kopp HG, Placke T and Salih HR: Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 69:7775–7783. 2009. View Article : Google Scholar : PubMed/NCBI

123 

Zhang Q, Ding D, Liu X and Guo SW: Activated platelets induce estrogen receptor beta expression in endometriotic stromal cells. Gynecol Obstet Invest. 80:187–192. 2015. View Article : Google Scholar : PubMed/NCBI

124 

Binyamin L, Alpaugh RK, Hughes TL, Lutz CT, Campbell KS and Weiner LM: Blocking NK cell inhibitory self-recognition promotes antibody-dependent cellular cytotoxicity in a model of anti-lymphoma therapy. J Immunol. 180:6392–6401. 2008. View Article : Google Scholar : PubMed/NCBI

125 

Andre P, Denis C, Soulas C, Bourbon-Caillet C, Lopez J, Arnoux T, Bléry M, Bonnafous C, Gauthier L, Morel A, et al: Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell. 175:1731–1743.e13. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Chan EY, Yap DY, Colucci M, Ma AL, Parekh RS and Tullus K: Use of rituximab in childhood idiopathic nephrotic syndrome. Clin J Am Soc Nephrol. 18:533–548. 2023. View Article : Google Scholar : PubMed/NCBI

127 

Sivori S, Della Chiesa M, Carlomagno S, Quatrini L, Munari E, Vacca P, Tumino N, Mariotti FR, Mingari MC, Pende D and Moretta L: Inhibitory receptors and checkpoints in human NK cells, implications for the immunotherapy of cancer. Front Immunol. 11:21562020. View Article : Google Scholar : PubMed/NCBI

128 

Velasco I, Quereda F, Bermejo R, Campos A and Acien P: Intraperitoneal recombinant Interleukin-2 activates leukocytes in rat endometriosis. J Reprod Immunol. 74:124–132. 2007. View Article : Google Scholar : PubMed/NCBI

129 

Quereda F, Bermejo R, Velasco I, Campos A and Acien P: The effect of intraperitoneal Interleukin-2 on surgically induced endometriosis in rats. Eur J Obstet Gynecol Reprod Biol. 136:243–248. 2008. View Article : Google Scholar : PubMed/NCBI

130 

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

131 

Wu MY, Chao KH, Chen SU, Chen HF, Yang YS, Huang SC and Ho HN: The suppression of peritoneal cellular immunity in women with endometriosis could be restored after gonadotropin releasing hormone agonist treatment. Am J Reprod Immunol. 35:510–516. 1996. View Article : Google Scholar : PubMed/NCBI

132 

Regis S, Dondero A, Caliendo F, Bottino C and Castriconi R: NK cell function regulation by TGF-β-induced epigenetic mechanisms. Front Immunol. 11:3112020. View Article : Google Scholar : PubMed/NCBI

133 

Yoshimura A, Wakabayashi Y and Mori T: Cellular and molecular basis for the regulation of inflammation by TGF-beta. J Biochem. 147:781–792. 2010. View Article : Google Scholar : PubMed/NCBI

134 

Young VJ, Brown JK, Saunders PT, Duncan WC and Horne AW: The peritoneum is both a source and target of TGF-β in women with endometriosis. PLoS One. 9:e1067732014. View Article : Google Scholar : PubMed/NCBI

135 

Zhang M, Xu T, Tong D, Li S, Yu X, Liu B, Jiang L and Liu K: Research advances in endometriosis-related signaling pathways: A review. Biomed Pharmacother. 164:1149092023. View Article : Google Scholar : PubMed/NCBI

136 

Hawinkels LJ and Ten Dijke P: Exploring Anti-TGF-β therapies in cancer and fibrosis. Growth Factors. 29:140–152. 2011. View Article : Google Scholar : PubMed/NCBI

137 

Voelker J, Berg PH, Sheetz M, Duffin K, Shen T, Moser B, Greene T, Blumenthal SS, Rychlik I, Yagil Y, et al: Anti-TGF-β1 antibody therapy in patients with diabetic nephropathy. J Am Soc Nephrol. 28:953–962. 2017. View Article : Google Scholar : PubMed/NCBI

138 

Clayton RD, Duffy SR, Wilkinson N, Garry R and Jackson AM: Increase in peripheral blood mononuclear cell (PBMC)- and CD56+ cell-mediated killing of endometrial stromal cells by mycobacteria; a possible role in endometriosis immunotherapy? Hum Reprod. 19:1886–1893. 2004. View Article : Google Scholar : PubMed/NCBI

139 

Oksasoglu B, Hepokur C, Misir S, Yildiz C, Sonmez G and Yanik A: Determination of PD-1 expression in peripheral blood cells in patients with endometriosis. Gynecol Endocrinol. 37:157–161. 2021. View Article : Google Scholar : PubMed/NCBI

140 

Wu L, Lv C, Su Y, Li C, Zhang H, Zhao X and Li M: Expression of programmed death-1 (PD-1) and Its Ligand PD-L1 is upregulated in endometriosis and promoted by 17beta-estradiol. Gynecol Endocrinol. 35:251–256. 2019. View Article : Google Scholar : PubMed/NCBI

141 

Chen Z, Yang Y, Liu LL and Lundqvist A: Strategies to augment natural killer (NK) cell activity against solid tumors. Cancers (Basel). 11:10402019. View Article : Google Scholar : PubMed/NCBI

142 

Memon H and Patel BM: Immune checkpoint inhibitors in non-small cell lung cancer: A Bird's eye view. Life Sci. 233:1167132019. View Article : Google Scholar : PubMed/NCBI

143 

Giannopoulos K: Targeting immune signaling checkpoints in acute myeloid leukemia. J Clin Med. 8:2362019. View Article : Google Scholar : PubMed/NCBI

144 

Hofmann L, Forschner A, Loquai C, Goldinger SM, Zimmer L, Ugurel S, Schmidgen MI, Gutzmer R, Utikal JS, Göppner D, et al: Cutaneous, gastrointestinal, hepatic, endocrine, and renal side-effects of Anti-PD-1 therapy. Eur J Cancer. 60:190–209. 2016. View Article : Google Scholar : PubMed/NCBI

145 

Sosa A, Lopez Cadena E, Simon Olive C, Karachaliou N and Rosell R: Clinical assessment of immune-related adverse events. Ther Adv Med Oncol. 10:17588359187646282018. View Article : Google Scholar : PubMed/NCBI

146 

Afolabi LO, Adeshakin AO, Sani MM, Bi J and Wan X: Genetic reprogramming for NK cell cancer immunotherapy with CRISPR/Cas9. Immunology. 158:63–69. 2019. View Article : Google Scholar : PubMed/NCBI

147 

Wang L, Dou M, Ma Q, Yao R and Liu J: Chimeric antigen receptor (CAR)-modified NK cells against cancer: Opportunities and challenges. Int Immunopharmacol. 74:1056952019. View Article : Google Scholar : PubMed/NCBI

148 

Kloess S, Kretschmer A, Stahl L, Fricke S and Koehl U: CAR-expressing natural killer cells for cancer retargeting. Transfus Med Hemother. 46:4–13. 2019. View Article : Google Scholar : PubMed/NCBI

149 

Seshadri S and Sunkara SK: Natural killer cells in female infertility and recurrent miscarriage: A systematic review and meta-analysis. Hum Reprod Update. 20:429–438. 2014. View Article : Google Scholar : PubMed/NCBI

150 

Hadinedoushan H, Mirahmadian M and Aflatounian A: Increased natural killer cell cytotoxicity and Il-2 production in recurrent spontaneous abortion. Am J Reprod Immunol. 58:409–414. 2007. View Article : Google Scholar : PubMed/NCBI

151 

Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D, et al: Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 38:792–804. 2013. View Article : Google Scholar : PubMed/NCBI

152 

Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F and Rodewald HR: Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 518:547–551. 2015. View Article : Google Scholar : PubMed/NCBI

153 

Gordon S and Martinez FO: Alternative activation of macrophages: Mechanism and functions. Immunity. 32:593–604. 2010. View Article : Google Scholar : PubMed/NCBI

154 

Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M, Vaisberg E, Kastelein R, Kolk A, de Waal-Malefyt R and Ottenhoff TH: Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci USA. 101:4560–4565. 2004. View Article : Google Scholar : PubMed/NCBI

155 

Bonatz G, Hansmann ML, Buchholz F, Mettler L, Radzun HJ and Semm K: Macrophage- and lymphocyte-subtypes in the endometrium during different phases of the ovarian cycle. Int J Gynaecol Obstet. 37:29–36. 1992. View Article : Google Scholar : PubMed/NCBI

156 

Vallve-Juanico J, Santamaria X, Vo KC, Houshdaran S and Giudice LC: Macrophages display proinflammatory phenotypes in the eutopic endometrium of women with endometriosis with relevance to an infectious etiology of the disease. Fertil Steril. 112:1118–1128. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Maybin JA and Critchley HO: Menstrual physiology: Implications for endometrial pathology and beyond. Hum Reprod Update. 21:748–761. 2015. View Article : Google Scholar : PubMed/NCBI

158 

Bain CC and Jenkins SJ: The biology of serous cavity macrophages. Cell Immunol. 330:126–135. 2018. View Article : Google Scholar : PubMed/NCBI

159 

Hudson QJ, Ashjaei K, Perricos A, Kuessel L, Husslein H, Wenzl R and Yotova I: Endometriosis patients show an increased M2 response in the peritoneal CD14+low/CD68+low macrophage subpopulation coupled with an increase in the T-helper 2 and T-regulatory cells. Reprod Sci. 27:1920–1931. 2020. View Article : Google Scholar : PubMed/NCBI

160 

Roszer T: Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediators Inflamm. 2015:8164602015. View Article : Google Scholar : PubMed/NCBI

161 

Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A and Locati M: The Chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 25:677–686. 2004. View Article : Google Scholar : PubMed/NCBI

162 

Bardi GT, Smith MA and Hood JL: Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine. 105:63–72. 2018. View Article : Google Scholar : PubMed/NCBI

163 

Simpson JL, Elias S, Malinak LR and Buttram VC Jr: Heritable aspects of Endometriosis. I. Genetic studies. Am J Obstet Gynecol. 137:327–331. 1980. View Article : Google Scholar : PubMed/NCBI

164 

Dmowski WP, Gebel H and Braun DP: Decreased apoptosis and sensitivity to macrophage mediated cytolysis of endometrial cells in endometriosis. Hum Reprod Update. 4:696–701. 1998. View Article : Google Scholar : PubMed/NCBI

165 

Wu MH, Shoji Y, Wu MC, Chuang PC, Lin CC, Huang MF and Tsai SJ: Suppression of matrix metalloproteinase-9 by prostaglandin E(2) in peritoneal macrophage is associated with severity of endometriosis. Am J Pathol. 167:1061–1069. 2005. View Article : Google Scholar : PubMed/NCBI

166 

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

167 

Kwon MJ, Shin HY, Cui Y, Kim H, Thi AH, Choi JY, Kim EY, Hwang DH and Kim BG: CCL2 mediates neuron-macrophage interactions to drive proregenerative macrophage activation following preconditioning injury. J Neurosci. 35:15934–15947. 2015. View Article : Google Scholar : PubMed/NCBI

168 

Johan MZ, Ingman WV, Robertson SA and Hull ML: Macrophages infiltrating endometriosis-like lesions exhibit progressive phenotype Changes in a heterologous mouse model. J Reprod Immunol. 132:1–8. 2019. View Article : Google Scholar : PubMed/NCBI

169 

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. View Article : Google Scholar : PubMed/NCBI

170 

Huang YJ, Yang CK, Wei PL, Huynh TT, Whang-Peng J, Meng TC, Hsiao M, Tzeng YM, Wu AT and Yen Y: Ovatodiolide suppresses colon tumorigenesis and prevents polarization of M2 Tumor-associated macrophages through YAP oncogenic pathways. J Hematol Oncol. 10:602017. View Article : Google Scholar : PubMed/NCBI

171 

Monsivais D, Dyson MT, Yin P, Coon JS, Navarro A, Feng G, Malpani SS, Ono M, Ercan CM, Wei JJ, et al: ERβ- and prostaglandin E2-regulated pathways integrate cell proliferation via Ras-like and estrogen-regulated growth inhibitor in endometriosis. Mol Endocrinol. 28:1304–1315. 2014. View Article : Google Scholar : PubMed/NCBI

172 

Han SJ, Jung SY, Wu SP, Hawkins SM, Park MJ, Kyo S, Qin J, Lydon JP, Tsai SY, Tsai MJ, et al: Estrogen receptor β modulates apoptosis complexes and the inflammasome to drive the pathogenesis of endometriosis. Cell. 163:960–974. 2015. View Article : Google Scholar : PubMed/NCBI

173 

Stanojevic S, Curuvija I, Blagojevic V, Petrovic R, Prijic I and Vujic V: The involvement of estrogen receptors α and β in the in vitro effects of 17β-estradiol on secretory profile of peritoneal macrophages from naturally menopausal female and middle-aged male rats. Exp Gerontol. 113:86–94. 2018. View Article : Google Scholar : PubMed/NCBI

174 

Zhao Y, Gong P, Chen Y, Nwachukwu JC, Srinivasan S, Ko C, Bagchi MK, Taylor RN, Korach KS, Nettles KW, et al: Dual suppression of estrogenic and inflammatory activities for targeting of endometriosis. Sci Transl Med. 7:271ra92015. View Article : Google Scholar : PubMed/NCBI

175 

Panchanathan R, Shen H, Zhang X, Ho SM and Choubey D: Mutually positive regulatory feedback loop between interferons and estrogen receptor-alpha in mice: Implications for sex bias in autoimmunity. PLoS One. 5:e108682010. View Article : Google Scholar : PubMed/NCBI

176 

Smith S, Ni Gabhann J, McCarthy E, Coffey B, Mahony R, Byrne JC, Stacey K, Ball E, Bell A, Cunnane G, et al: Estrogen receptor α regulates tripartite motif-containing protein 21 expression, contributing to dysregulated cytokine production in systemic lupus erythematosus. Arthritis Rheumatol. 66:163–172. 2014. View Article : Google Scholar : PubMed/NCBI

177 

Feldman I, Feldman GM, Mobarak C, Dunkelberg JC and Leslie KK: Identification of proteins within the nuclear factor-kappa B transcriptional complex including estrogen receptor-alpha. Am J Obstet Gynecol. 196:394.e1–13. 2007. View Article : Google Scholar : PubMed/NCBI

178 

Kalaitzidis D and Gilmore TD: Transcription factor cross-talk: The estrogen receptor and NF-kappaB. Trends Endocrinol Metab. 16:46–52. 2005. View Article : Google Scholar : PubMed/NCBI

179 

Greaves E, Temp J, Esnal-Zufiurre A, Mechsner S, Horne AW and Saunders PT: Estradiol is a critical mediator of macrophage-nerve cross talk in peritoneal endometriosis. Am J Pathol. 185:2286–2297. 2015. View Article : Google Scholar : PubMed/NCBI

180 

Gaudet HM, Cheng SB, Christensen EM and Filardo EJ: The G-protein coupled estrogen receptor, gper: The inside and inside-out story. Mol Cell Endocrinol. 418:207–219. 2015. View Article : Google Scholar : PubMed/NCBI

181 

Revankar CM, Cimino DF, Sklar LA, Arterburn JB and Prossnitz ER: A Transmembrane intracellular estrogen receptor mediates rapid cell signaling. Science. 307:1625–1630. 2005. View Article : Google Scholar : PubMed/NCBI

182 

Heublein S, Lenhard M, Vrekoussis T, Schoepfer J, Kuhn C, Friese K, Makrigiannakis A, Mayr D and Jeschke U: The G-protein-coupled estrogen receptor (GPER) is expressed in normal human ovaries and is upregulated in ovarian endometriosis and pelvic inflammatory disease involving the ovary. Reprod Sci. 19:1197–1204. 2012. View Article : Google Scholar : PubMed/NCBI

183 

Okamoto M, Suzuki T, Mizukami Y and Ikeda T: The Membrane-type estrogen receptor G-protein-coupled estrogen receptor suppresses lipopolysaccharide-induced interleukin 6 via inhibition of nuclear factor-kappa B pathway in murine macrophage cells. Anim Sci J. 88:1870–1879. 2017. View Article : Google Scholar : PubMed/NCBI

184 

Calippe B, Douin-Echinard V, Delpy L, Laffargue M, Lelu K, Krust A, Pipy B, Bayard F, Arnal JF, Guéry JC and Gourdy P: 17Beta-estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor alpha signaling in macrophages in vivo. J Immunol. 185:1169–1176. 2010. View Article : Google Scholar : PubMed/NCBI

185 

Zhang YH, He M, Wang Y and Liao AH: Modulators of the balance between M1 and M2 macrophages during pregnancy. Front Immunol. 8:1202017.PubMed/NCBI

186 

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

187 

Hirata T, Osuga Y, Takamura M, Kodama A, Hirota Y, Koga K, Yoshino O, Harada M, Takemura Y, Yano T and Taketani Y: Recruitment of CCR6-expressing Th17 cells by CCL 20 secreted from IL-1 beta-, TNF-alpha-, and IL-17A-stimulated endometriotic stromal cells. Endocrinology. 151:5468–5476. 2010. View Article : Google Scholar : PubMed/NCBI

188 

Buerger C, Shirsath N, Lang V, Berard A, Diehl S, Kaufmann R, Boehncke WH and Wolf P: Inflammation dependent mTORC1 signaling interferes with the switch from keratinocyte proliferation to differentiation. PLoS One. 12:e01808532017. View Article : Google Scholar : PubMed/NCBI

189 

Schenken RS, Johnson JV and Riehl RM: C-myc protooncogene polypeptide expression in endometriosis. Am J Obstet Gynecol. 164:1031–1037. 1991. View Article : Google Scholar : PubMed/NCBI

190 

Selam B, Kayisli UA, Garcia-Velasco JA, Akbas GE and Arici A: Regulation of Fas ligand expression by IL-8 in human endometrium. J Clin Endocrinol Metab. 87:3921–3927. 2002. View Article : Google Scholar : PubMed/NCBI

191 

Gazvani MR, Christmas S, Quenby S, Kirwan J, Johnson PM and Kingsland CR: Peritoneal fluid concentrations of interleukin-8 in women with endometriosis: Relationship to stage of disease. Human Reproduction. 13:1957–1961. 1998. View Article : Google Scholar : PubMed/NCBI

192 

Borrelli GM, Abrao MS and Mechsner S: Can chemokines be used as biomarkers for endometriosis? A systematic review. Hum Reprod. 29:253–266. 2014. View Article : Google Scholar : PubMed/NCBI

193 

Schneider J, Jimenez E, Rodriguez F and del Tanago JG: c-myc, c-erb-B2, nm23 and p53 expression in human endometriosis. Oncol Rep. 5:49–52. 1998.PubMed/NCBI

194 

Capobianco A, Monno A, Cottone L, Venneri MA, Biziato D, Di Puppo F, Ferrari S, De Palma M, Manfredi AA and Rovere-Querini P: Proangiogenic Tie2(+) macrophages infiltrate human and murine endometriotic lesions and dictate their growth in a mouse model of the disease. Am J Pathol. 179:2651–2659. 2011. View Article : Google Scholar : PubMed/NCBI

195 

Kats R, Collette T, Metz CN and Akoum A: Marked elevation of macrophage migration inhibitory factor in the peritoneal fluid of women with endometriosis. Fertil Steril. 78:69–76. 2002. View Article : Google Scholar : PubMed/NCBI

196 

Akoum A, Metz CN, Al-Akoum M and Kats R: Macrophage migration inhibitory factor expression in the intrauterine endometrium of women with endometriosis varies with disease stage, infertility status, and pelvic pain. Fertil Steril. 85:1379–1385. 2006. View Article : Google Scholar : PubMed/NCBI

197 

Morin M, Bellehumeur C, Therriault MJ, Metz C, Maheux R and Akoum A: Elevated levels of macrophage migration inhibitory factor in the peripheral blood of women with endometriosis. Fertil Steril. 83:865–872. 2005. View Article : Google Scholar : PubMed/NCBI

198 

Kats R, Metz CN and Akoum A: Macrophage migration inhibitory factor is markedly expressed in active and early-stage endometriotic lesions. J Clin Endocrinol Metab. 87:883–889. 2002. View Article : Google Scholar : PubMed/NCBI

199 

Seeber B, Sammel MD, Fan X, Gerton GL, Shaunik A, Chittams J and Barnhart KT: Panel of markers can accurately predict endometriosis in a subset of patients. Fertil Steril. 89:1073–1081. 2008. View Article : Google Scholar : PubMed/NCBI

200 

Zinovkin DA, Pranjol MZI, Bilsky IA and Zmushko VA: Tumor-associated T-lymphocytes and macrophages are decreased in endometrioid endometrial carcinoma with melf-pattern stromal changes. Cancer Microenviron. 11:107–114. 2018. View Article : Google Scholar : PubMed/NCBI

201 

Jha P, Farooq A, Agarwal N and Buckshee K: In vitro sperm phagocytosis by human peritoneal macrophages in endometriosis-associated infertility. Am J Reprod Immunol. 36:235–237. 1996. View Article : Google Scholar : PubMed/NCBI

202 

Forster R, Sarginson A, Velichkova A, Hogg C, Dorning A, Horne AW, Saunders PTK and Greaves E: Macrophage-derived insulin-like growth Factor-1 is a key neurotrophic and nerve-sensitizing factor in pain associated with endometriosis. FASEB J. 33:11210–11222. 2019. View Article : Google Scholar : PubMed/NCBI

203 

Bushnell LF: Endometriosis and estrogen therapy. Am J Obstet Gynecol. 55:9151948. View Article : Google Scholar

204 

Barbieri RL: Endometriosis and the estrogen threshold theory. Relation to surgical and medical treatment. J Reprod Med. 43 (3 Suppl):S287–S292. 1998.

205 

Howell R, Dowsett M, King N and Edmonds DK: Endocrine effects of gnrh analogue with low-dose hormone replacement therapy in women with endometriosis. Clin Endocrinol (Oxf). 43:609–615. 1995. View Article : Google Scholar : PubMed/NCBI

206 

Chang CY, Chiang AJ, Yan MJ, Lai MT, Su YY, Huang HY, Chang CY, Li YH, Li PF, Chen CM, et al: Ribosome biogenesis serves as a therapeutic target for treating endometriosis and the associated complications. Biomedicines. 10:1852022. View Article : Google Scholar : PubMed/NCBI

207 

Khoufache K, Bazin S, Girard K, Guillemette J, Roy MC, Verreault JP, Al-Abed Y, Foster W and Akoum A: Macrophage migration inhibitory factor antagonist blocks the development of endometriosis in vivo. PLoS One. 7:e372642012. View Article : Google Scholar : PubMed/NCBI

208 

Daha MR: Role of complement in innate immunity and infections. Crit Rev Immunol. 30:47–52. 2010. View Article : Google Scholar : PubMed/NCBI

209 

Conigliaro P, Triggianese P, Ballanti E, Perricone C, Perricone R and Chimenti MS: Complement, infection, and autoimmunity. Curr Opin Rheumatol. 31:532–541. 2019. View Article : Google Scholar : PubMed/NCBI

210 

Trouw LA, Blom AM and Gasque P: Role of complement and complement regulators in the removal of apoptotic cells. Mol Immunol. 45:1199–1207. 2008. View Article : Google Scholar : PubMed/NCBI

211 

Hajishengallis G, Reis ES, Mastellos DC, Ricklin D and Lambris JD: Novel mechanisms and functions of complement. Nat Immunol. 18:1288–1298. 2017. View Article : Google Scholar : PubMed/NCBI

212 

Lubbers R, van Essen MF, van Kooten C and Trouw LA: Production of complement components by cells of the immune system. Clin Exp Immunol. 188:183–194. 2017. View Article : Google Scholar : PubMed/NCBI

213 

Merle NS, Church SE, Fremeaux-Bacchi V and Roumenina LT: Complement system part I-molecular mechanisms of activation and regulation. Front Immunol. 6:2622015. View Article : Google Scholar : PubMed/NCBI

214 

Merle NS, Noe R, Halbwachs-Mecarelli L, Fremeaux-Bacchi V and Roumenina LT: Complement system part II: Role in immunity. Front Immunol. 6:2572015. View Article : Google Scholar : PubMed/NCBI

215 

Ricklin D, Reis ES and Lambris JD: Complement in disease: A defence system turning offensive. Nat Rev Nephrol. 12:383–401. 2016. View Article : Google Scholar : PubMed/NCBI

216 

Zhang T, De Carolis C, Man GCW and Wang CC: The Link between Immunity, autoimmunity and endometriosis: A literature update. Autoimmunity Revi. 17:945–955. 2018. View Article : Google Scholar : PubMed/NCBI

217 

Aslan C, Ak H, Askar N, Ozkaya AB, Ergenoglu AM, Yeniel AO, Akdemir A and Aydin HH: Overexpression of complement C5 in endometriosis. Clin Biochem. 47:496–498. 2014. View Article : Google Scholar : PubMed/NCBI

218 

Rahal D, Andrade F and Nisihara R: Insights into the role of complement system in the pathophysiology of endometriosis. Immunol Lett. 231:43–48. 2021. View Article : Google Scholar : PubMed/NCBI

219 

Kabut J, Kondera-Anasz Z, Sikora J and Mielczarek-Palacz A: Levels of complement components iC3b, C3c, C4, and SC5b-9 in peritoneal fluid and serum of infertile women with endometriosis. Fertil Steril. 88:1298–1303. 2007. View Article : Google Scholar : PubMed/NCBI

220 

Sikora J, Wroblewska-Czech A, Smycz-Kubanska M, Mielczarek-Palacz A, Cygal A, Witek A and Kondera-Anasz Z: The role of complement components C1q, MBL and C1 inhibitor in pathogenesis of endometriosis. Arch Gynecol Obstet. 297:1495–1501. 2018. View Article : Google Scholar : PubMed/NCBI

221 

Ahn SH, Khalaj K, Young SL, Lessey BA, Koti M and Tayade C: Immune-inflammation gene signatures in endometriosis patients. Fertil Steril. 106:1420–1431.e7. 2016. View Article : Google Scholar : PubMed/NCBI

222 

Weed JC and Arquembourg PC: Endometriosis: Can it produce an autoimmune response resulting in infertility? Clin Obstet Gynecol. 23:885–893. 1980. View Article : Google Scholar : PubMed/NCBI

223 

Isaacson KB, Coutifaris C, Garcia CR and Lyttle CR: Production and secretion of complement component-3 by endometriotic tissue. J Clin Endocrinol Metab. 69:1003–1009. 1989. View Article : Google Scholar : PubMed/NCBI

224 

Badawy SZA, Cuenca V, Stitzel A, Jacobs RDB and Tomar RH: Autoimmune phenomena in infertile patients with endometriosis. Obstet Gynecol. 63:271–275. 1984.PubMed/NCBI

225 

Badawy SZA, Cuenca V, Marshall L, Munchback R, Rinas AC and Coble DA: Cellular-components in peritoneal-fluid in infertile patients with and without endometriosis. Fertil Steril. 42:704–708. 1984. View Article : Google Scholar : PubMed/NCBI

226 

Tao XJ, Sayegh RA and Isaacson KB: Increased expression of complement component 3 in human ectopic endometrium compared with the matched eutopic endometrium. Fertil Steril. 68:460–467. 1997. View Article : Google Scholar : PubMed/NCBI

227 

Sayegh RA, Tao XJ, Awwad JT and Isaacson KB: Localization of the expression of complement component 3 in the human endometrium by in situ hybridization. J Clin Endocrinol Metab. 81:1641–1649. 1996. View Article : Google Scholar : PubMed/NCBI

228 

Steele RW, Dmowski WP and Marmer DJ: Immunological aspects of human endometriosis. Am J Reprod Immunol (1980). 6:33–36. 1984. View Article : Google Scholar : PubMed/NCBI

229 

Meek SC, Hodge DD and Musich JR: Autoimmunity in infertile patients with endometriosis. Am J Obstet Gynecol. 158:1365–1373. 1988. View Article : Google Scholar : PubMed/NCBI

230 

Isaacson KB, Galman M, Coutifaris C and Lyttle CR: Endometrial synthesis and secretion of complement component-3 by patients with and without endometriosis. Fertil Steril. 53:836–841. 1990. View Article : Google Scholar : PubMed/NCBI

231 

Hasan A, Rahim A, Afzal M, Naveed AK, Ayub S and Jahan S: Serum albumin and C3 complement levels in endometriosis. J Coll Physicians Surg Pak. 29:702–705. 2019. View Article : Google Scholar : PubMed/NCBI

232 

Bischof P, Planasbasset D, Campana A and Meisser A: Investigations on the cell type responsible for the endometrial secretion of complement component-3 (C3). Human Reprod. 9:1652–1659. 1994. View Article : Google Scholar : PubMed/NCBI

233 

Liu CF, Min XY, Wang N, Wang JX, Ma N, Dong X, Zhang B, Wu W, Li ZF, Zhou W and Li K: Complement receptor 3 Has negative impact on tumor surveillance through suppression of natural killer cell function. Front Immunol. 8:16022017. View Article : Google Scholar : PubMed/NCBI

234 

Suryawanshi S, Huang X, Elishaev E, Budiu RA, Zhang L, Kim S, Donnellan N, Mantia-Smaldone G, Ma T, Tseng G, et al: Complement pathway is frequently altered in endometriosis and endometriosis-associated ovarian cancer. Clin Cancer Res. 20:6163–6174. 2014. View Article : Google Scholar : PubMed/NCBI

235 

Woelfler MM, Meinhold-Heerlein IM, Soehngen L, Rath W, Knuechel R, Neulen J, Maass N and Henkel C: Two-dimensional gel electrophoresis in peritoneal fluid samples identifies differential protein regulation in patients suffering from peritoneal or ovarian endometriosis. Fertil Steril. 95:2764–2768. 2011. View Article : Google Scholar : PubMed/NCBI

236 

Riley CF, Moen MH and Videm V: Inflammatory markers in endometriosis: Reduced peritoneal neutrophil response in minimal endometriosis. Acta Obstet Gynecol Scand. 86:877–881. 2007. View Article : Google Scholar : PubMed/NCBI

237 

Kruse C, Steffensen R, Nielsen HJ and Jensenius JC: Mannan-binding lectin polymorphisms and serum levels in patients with endometriosis. Eur J Obstet Gynecol Reprod Biol. 181:256–258. 2014. View Article : Google Scholar : PubMed/NCBI

238 

Kruse C, Steffensen R, Nielsen HJ and Jensenius JC: Mannan-binding lectin polymorphisms and serum levels in patients with endometriosis. Eur J Obstet Gynecol Reprod Biol. 181:256–258. 2014. View Article : Google Scholar : PubMed/NCBI

239 

Kavoussi SK, Mueller MD and Lebovic DI: Expression of mannose-binding lectin in the peritoneal fluid of women with and without endometriosis. Fertil Steril. 85:1526–1528. 2006. View Article : Google Scholar : PubMed/NCBI

240 

Ozerkan K, Oral B and Uncu G: Mannose-binding lectin levels in endometriosis. Fertil Steril. 94:775–776. 2010. View Article : Google Scholar : PubMed/NCBI

241 

Karadadas E, Hortu I, Ak H, Ergenoglu AM, Karadadas N and Aydin HH: Evaluation of complement system proteins C3a, C5a and C6 in patients of endometriosis. Clin Biochem. 81:15–19. 2020. View Article : Google Scholar : PubMed/NCBI

242 

Stefkovich ML, Arao Y, Hamilton KJ and Korach KS: Experimental models for evaluating non-genomic estrogen signaling. Steroids. 133:34–37. 2018. View Article : Google Scholar : PubMed/NCBI

243 

Al-Sabbagh M, Lam EW and Brosens JJ: Mechanisms of endometrial progesterone resistance. Mol Cell Endocrinol. 358:208–215. 2012. View Article : Google Scholar : PubMed/NCBI

244 

Han SJ and O'Malley BW: The dynamics of nuclear receptors and nuclear receptor coregulators in the pathogenesis of endometriosis. Hum Reprod Update. 20:467–484. 2014. View Article : Google Scholar : PubMed/NCBI

245 

Patel BG, Rudnicki M, Yu J, Shu Y and Taylor RN: Progesterone resistance in endometriosis: Origins, consequences and interventions. Acta Obstet Gynecol Scand. 96:623–632. 2017. View Article : Google Scholar : PubMed/NCBI

246 

Kastner P, Krust A, Turcotte B, Stropp U, Tora L, Gronemeyer H and Chambon P: Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J. 9:1603–1614. 1990. View Article : Google Scholar : PubMed/NCBI

247 

Patel B, Elguero S, Thakore S, Dahoud W, Bedaiwy M and Mesiano S: Role of Nuclear progesterone receptor isoforms in uterine pathophysiology. Hum Reprod Update. 21:155–173. 2015. View Article : Google Scholar : PubMed/NCBI

248 

Vasquez YM and DeMayo FJ: Role of nuclear receptors in blastocyst implantation. Semin Cell Dev Biol. 24:724–735. 2013. View Article : Google Scholar : PubMed/NCBI

249 

Chantalat E, Valera MC, Vaysse C, Noirrit E, Rusidze M, Weyl A, Vergriete K, Buscail E, Lluel P, Fontaine C, et al: Estrogen receptors and endometriosis. Int J Mol Sci. 21:28152020. View Article : Google Scholar : PubMed/NCBI

250 

Wang Y, Zhu L, Kuokkanen S and Pollard JW: Activation of protein synthesis in mouse uterine epithelial cells by estradiol-17β is mediated by a PKC-ERK1/2-mTOR signaling pathway. Proc Natl Acad Sci USA. 112:E1382–E1391. 2015.PubMed/NCBI

251 

Reis FM, Petraglia F and Taylor RN: Endometriosis: Hormone regulation and clinical consequences of chemotaxis and apoptosis. Hum Reprod Update. 19:406–418. 2013. View Article : Google Scholar : PubMed/NCBI

252 

Yilmaz BD and Bulun SE: Endometriosis and nuclear receptors. Hum Reprod Update. 25:473–485. 2019. View Article : Google Scholar : PubMed/NCBI

253 

Xue Q, Lin Z, Cheng YH, Huang CC, Marsh E, Yin P, Milad MP, Confino E, Reierstad S, Innes J and Bulun SE: Promoter methylation regulates estrogen receptor 2 in human endometrium and endometriosis. Biol Reprod. 77:681–687. 2007. View Article : Google Scholar : PubMed/NCBI

254 

Trukhacheva E, Lin Z, Reierstad S, Cheng YH, Milad M and Bulun SE: Estrogen receptor (ER) beta regulates eralpha expression in stromal cells derived from ovarian endometriosis. J Clin Endocrinol Metab. 94:615–622. 2009. View Article : Google Scholar : PubMed/NCBI

255 

Bulun SE, Yilmaz BD, Sison C, Miyazaki K, Bernardi L, Liu S, Kohlmeier A, Yin P, Milad M and Wei J: Endometriosis. Endocr Rev. 40:1048–1079. 2019. View Article : Google Scholar : PubMed/NCBI

256 

Burney RO, Talbi S, Hamilton AE, Vo KC, Nyegaard M, Nezhat CR, Lessey BA and Giudice LC: Gene expression analysis of endometrium reveals progesterone resistance and candidate susceptibility genes in women with endometriosis. Endocrinology. 148:3814–3826. 2007. View Article : Google Scholar : PubMed/NCBI

257 

Flores VA, Vanhie A, Dang T and Taylor HS: Progesterone receptor status predicts response to progestin therapy in endometriosis. J Clin Endocrinol Metab. 103:4561–4568. 2018. View Article : Google Scholar : PubMed/NCBI

258 

Broi MGD, Rocha CVJ, Meola J, Martins WP, Carvalho FM, Ferriani RA and Navarro PA: Expression of PGR, HBEGF, ITGAV, ITGB3 and SPP1 genes in eutopic endometrium of infertile women with endometriosis during the implantation window: A pilot study. JBRA Assist Reprod. 21:196–202. 2017. View Article : Google Scholar : PubMed/NCBI

259 

Strowitzki T, Marr J, Gerlinger C, Faustmann T and Seitz C: Dienogest is as effective as leuprolide acetate in treating the painful symptoms of endometriosis: A 24-Week, randomized, multicentre, open-label trial. Hum Reprod. 25:633–641. 2010. View Article : Google Scholar : PubMed/NCBI

260 

Saunders PTK and Horne AW: Endometriosis: Etiology, pathobiology, and therapeutic prospects. Cell. 184:2807–2824. 2021. View Article : Google Scholar : PubMed/NCBI

261 

Ferrero S, Evangelisti G and Barra F: Current and emerging treatment options for endometriosis. Expert Opin Pharmacother. 19:1109–1125. 2018. View Article : Google Scholar : PubMed/NCBI

262 

Capezzuoli T, Rossi M, La Torre F, Vannuccini S and Petraglia F: Hormonal drugs for the treatment of endometriosis. Curr Opin Pharmacol. 67:1023112022. View Article : Google Scholar : PubMed/NCBI

263 

Dunselman GA, Vermeulen N, Becker C, Calhaz-Jorge C, D'Hooghe T, De Bie B, Heikinheimo O, Horne AW, Kiesel L, Nap A, et al: ESHRE guideline: Management of women with endometriosis. Hum Reprod. 29:400–412. 2014. View Article : Google Scholar : PubMed/NCBI

264 

Crosignani PG, Luciano A, Ray A and Bergqvist A: Subcutaneous depot medroxyprogesterone acetate versus leuprolide acetate in the treatment of endometriosis-associated pain. Hum Reprod. 21:248–256. 2006. View Article : Google Scholar : PubMed/NCBI

265 

Selak V, Farquhar C, Prentice A and Singla A: Danazol for pelvic pain associated with endometriosis. Cochrane Database Syst Rev. CD0000682007.PubMed/NCBI

266 

Strowitzki T, Faustmann T, Gerlinger C and Seitz C: Dienogest in the treatment of endometriosis-associated pelvic pain: A 12-week, randomized, double-blind, placebo-controlled study. Eur J Obstet Gynecol Reprod Biol. 151:193–198. 2010. View Article : Google Scholar : PubMed/NCBI

267 

Petaja J: Inflammation and coagulation. An overview. Thromb Res. 127 (Suppl 2):S34–S37. 2011. View Article : Google Scholar : PubMed/NCBI

268 

Demetz G and Ott I: The interface between inflammation and coagulation in cardiovascular disease. Int J Inflam. 2012:8603012012.PubMed/NCBI

269 

Lessey BA, Castelbaum AJ, Sawin SW, Buck CA, Schinnar R, Bilker W and Strom BL: Aberrant integrin expression in the endometrium of women with endometriosis. J Clin Endocrinol Metab. 79:643–649. 1994. View Article : Google Scholar : PubMed/NCBI

270 

Regidor PA, Vogel C, Regidor M, Schindler AE and Winterhager E: Expression pattern of integrin adhesion molecules in endometriosis and human endometrium. Hum Reprod Update. 4:710–718. 1998. View Article : Google Scholar : PubMed/NCBI

271 

Libersan D and Merhi Y: Platelet P-selectin expression: Requirement for protein kinase C, but not protein tyrosine kinase or phosphoinositide 3-kinase. Thromb Haemost. 89:1016–1023. 2003. View Article : Google Scholar : PubMed/NCBI

272 

Guo SW, Ding D, Geng JG, Wang L and Liu X: P-selectin as a potential therapeutic target for endometriosis. Fertil Steril. 103:990–1000.e8. 2015. View Article : Google Scholar : PubMed/NCBI

273 

Lorant DE, Patel KD, McIntyre TM, McEver RP, Prescott SM and Zimmerman GA: Coexpression of Gmp-140 and paf by endothelium stimulated by histamine or thrombin: A juxtacrine system for adhesion and activation of neutrophils. J Cell Biol. 115:223–234. 1991. View Article : Google Scholar : PubMed/NCBI

274 

Lefer DJ: Pharmacology of selectin inhibitors in ischemia/reperfusion states. Annu Rev Pharmacol Toxicol. 40:283–294. 2000. View Article : Google Scholar : PubMed/NCBI

275 

Kling D, Stucki C, Kronenberg S, Tuerck D, Rheaume E, Tardif JC, Gaudreault J and Schmitt C: Pharmacological control of platelet-leukocyte interactions by the human anti-P-selectin antibody inclacumab-preclinical and clinical studies. Thromb Res. 131:401–410. 2013. View Article : Google Scholar : PubMed/NCBI

276 

Harada T and Taniguchi F: Dienogest: A new therapeutic agent for the treatment of endometriosis. Womens Health (Lond). 6:27–35. 2010. View Article : Google Scholar : PubMed/NCBI

277 

Kaminski K, Fiegler P, Marr J and Moore C: Treatment of endometriosis with dienogest: Preliminary report. Ginekol Pol. 72:299–304. 2001.(In Polish). PubMed/NCBI

278 

Harada T, Momoeda M, Taketani Y, Aso T, Fukunaga M, Hagino H and Terakawa N: Dienogest is as effective as intranasal buserelin acetate for the relief of pain symptoms associated with endometriosis-a randomized, double-blind, multicenter, controlled trial. Fertil Steril. 91:675–681. 2009. View Article : Google Scholar : PubMed/NCBI

279 

Rother RP, Rollins SA, Mojcik CF, Brodsky RA and Bell L: Discovery and development of the complement inhibitor eculizumab for the treatment of paroxysmal nocturnal hemoglobinuria. Nat Biotechnol. 25:1256–1264. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2025
Volume 31 Issue 3

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang W, Li K, Jian A, Zhang G and Zhang X: Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review). Mol Med Rep 31: 57, 2025.
APA
Zhang, W., Li, K., Jian, A., Zhang, G., & Zhang, X. (2025). Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review). Molecular Medicine Reports, 31, 57. https://doi.org/10.3892/mmr.2024.13422
MLA
Zhang, W., Li, K., Jian, A., Zhang, G., Zhang, X."Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review)". Molecular Medicine Reports 31.3 (2025): 57.
Chicago
Zhang, W., Li, K., Jian, A., Zhang, G., Zhang, X."Prospects for potential therapy targeting immune‑associated factors in endometriosis (Review)". Molecular Medicine Reports 31, no. 3 (2025): 57. https://doi.org/10.3892/mmr.2024.13422