- 1School of Nursing, University of South China, Hengyang, Hunan, China
- 2Scientific Research Department, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- 3Department of Neonatology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- 4Operating Room, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- 5Center for a Combination of Obstetrics and Gynecology and Reproductive Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- 6Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- 7Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
This review focuses on the field of endometrial cancer. Since 2020, there have been 417,367 new cases of endometrial cancer diagnosed globally and 97,370 deaths reported. Endometrial cancer ranks second in terms of incidence among female genital malignancies and third in terms of mortality among gynecological cancers. The stage, grade, and histological subtype of endometrial cancer were closely correlated with the risk of recurrence and prognosis for survival. Meanwhile, endometrial cancer exhibits significant biological heterogeneity. The complex interactions among the reproductive tract, host cells, and the microbial environment may harbor novel disease mechanisms. In this review, we provide an overview of the epidemiological characteristics, major risk factors, histological and molecular subtypes of endometrial cancer, as well as explore the associations between the female reproductive tract microbiome, immunity, and cancer progression. We also identify the specific roles of different cytokines in the pathophysiology of endometrial cancer. By integrating findings from diverse research fields, this comprehensive review offers an in-depth understanding of the multidimensional nature of endometrial cancer and highlights the significant potential and promising avenues that microbiological factors present for advancing future cancer research and guiding the development of innovative therapeutic strategies.
1 Introduction
Endometrial cancer (EC), a common gynecological malignancy, originates from the epithelial lining of the endometrium. Currently, a total of 417,367 newly diagnosed cases have been reported globally, with staggering death toll of 97,370 in 2020 reached, underscoring its significant impact on public health (1). It ranks as the fourth most prevalent malignancy among women in Europe and North America, trailing only breast, colorectal, and lung cancers, with the highest incidence observed in women aged 65–75 years (2). The elevated incidence of EC in Europe and North America may be correlated with the heightened prevalence of obesity and metabolic syndrome in these regions (3, 4). Furthermore, demographic shifts including population aging (5, 6) and the ongoing decrease in fertility rates have partially attributed to the rise in EC prevalence. Notably, obesity is recognized as an independent risk factor for EC, emphasizing its pivotal role in the etiology of this malignancy.
In China, the incidence of EC ranks second among malignancies of the female reproductive system, while its mortality rate stands as the third highest among gynecological malignancies (7, 8). This highlights the substantial impact of EC on women’s health in China. Since the mid-1970s, survival rates for most common cancers have improved, except for cervical cancer and EC, indirectly indicating a lack of significant therapeutic progress in treating these two types of malignancies. Previously, variations in histomorphological classification of EC, combined with limited reproducibility, imprecise risk stratification, and diverse treatment approaches, have contributed to the deficiencies of current risk stratification systems, thereby resulting in lower accuracy in identifying patients at high risk for disease recurrence or metastasis. However, grounded on the seminal discovery of four molecular subtypes of EC by the Cancer Genome Atlas (TCGA), an optimized assessment of disease staging and prognosis has been facilitated at the molecular level, enabling a more precise and individualized approach to EC patient management. Although the TCGA study findings present a promising new prospect for the clinical management of EC, their widespread implementation in clinical practice remains challenging due to the inherent complexity of the molecular subtyping system. This underscores the necessity for further validation and simplification of these findings to ensure their accessibility and practicality in routine clinical settings.
The intricate pathogenesis of cancer has long presented a major challenge facing the entire human race. So far, there have been many hypotheses about the EC pathogenesis. Among them, the estrogenic hypothesis, which posits the pivotal role of estrogen in EC development, has gradually emerged as the prevailing theory. Nevertheless, advancements in gene sequencing technologies have led to the gradual recognition of the uterus as an organ characterized by low microbial abundance. Given the high biological heterogeneity observed in EC, its diverse stages, grades, and pathological tissue types may exhibit associations with recurrence rates and prognostic survival, potentially revealing an alternative underlying mechanism involving interactions among the genital tract, host cells, and the microbial environment. This insight has inspired our comprehension of the relationship between the EC occurrence and the genital tract microbiome. At present, research on the relationship between the microbiome and the immune system remains in its early stages, and there are significant deficiencies in comprehensive studies linking molecular subtypes with immune microenvironment analysis. Regarding the mechanism by which microbial metabolites regulate cytokines in the body and affect tumor immunosurveillance, there is still controversy (9, 10). Therefore, a dedicated endeavor is warranted to elucidate the causal relationship between the composition and distribution of the genital tract microbiome and the host’s health status, with the aim of further enhancing our comprehension of the intricate interplay between these factors. From a microbial perspective, this review systematically integrates the application of epidemiological patterns, molecular subtype frameworks, and the interactions between the microbiome and immune microenvironment in EC, providing a comprehensive understanding of their roles in disease progression. It primarily elucidates the diagnostic potential of microbial biomarkers, explores the underlying pathogenic mechanisms, and evaluates the feasibility of therapeutic interventions targeting the microbiome.
2 Epidemiology
The epidemiological characteristics of EC exhibit pronounced variations across racial, geographical, and socioeconomic dimensions, with a particularly noteworthy high prevalence in developed countries (11). There was obvious regional heterogeneity in the age-standardized incidence rate (ASIR) of EC (Table 1). The incidence rates of EC varied significantly among different racial groups, exhibiting distinct trends. From 1900 to 2017 in America, the average incidence of EC was estimated at 25.9 per 100,000 among white women, 25.0 per 100,000 among all women, and 21.0 per 100,000 among black women (12). During the period from 2000 to 2015 in the United States, the incidence of EC was the highest among non-Hispanic whites, but the annual percentage change (APC) remained relatively stable at approximately 0.3%. And the incidence rates of EC among non-Hispanic blacks (APC = 2.0%), Hispanics (APC = 2.6%), and Asian/Pacific Islanders (APC = 1.9%) changed significantly and showed a continuous upward trend (13). This indicated the huge differences and inequalities among races and might imply the significant challenges faced by low socioeconomic groups. Based on the aggregated analysis of epidemiological research data in the United States from 2010 to 2019, it was found that the annual increase in EC mortality among African American/Black individuals was 1.9%, whereas for American white women, the average annual increase was 1.6% (14). Further studies have shown that the higher EC mortality in black women compared to white women can be attributed to tumor genomic differences, less access to chemotherapy and radiotherapy, low socioeconomic status, and lack of health insurance (15). Black women are more likely to present with advanced stages and higher grades of EC, as well as a more aggressive histomorphological subtype of the disease (15–17). The disparities in survival outcomes caused by race differences highlight the critical need for early screening and targeted interventions tailored to specific groups. It was worth noting that the median age at diagnosis for EC patients was 63 years old, and women over 50 years old accounted for a predominant majority of approximately 90% of all cases. The age distribution of disease onset showed a distinct pattern. Specifically, only 14% of EC cases were diagnosed prior to menopause, and an even smaller proportion, 5% involved women under the age of 40 (15, 18, 19).
Morbidity, recurrence rate, and mortality of EC exhibited a negative correlation with socio-economic indicators; that is, regions with lower socioeconomic levels tend to have higher EC morbidity and mortality (20). This relationship significantly increases the disease burden in low- and middle-income countries, posing a substantial threat to the health and well-being of the population (21). Significantly, limited accessibility to healthcare resources and a prevalent lack of awareness regarding health maintenance have contributed to an increase in late-stage cancer diagnoses upon initial detection, accompanied by a markedly shorter survival duration for individuals with lower socioeconomic status. This phenomenon underscored the urgent need for heightened attention from all sectors of society and the adoption of effective intervention measures (22, 23).
EC shows significant variations across dimensions such as race, geography and socio-economic factors. The epidemiological differences may be associated with EC and microbiome, potentially involving underlying biological mechanisms.
3 Risk factor
3.1 Metabolic syndrome
3.1.1 Obesity
According to the American Cancer Society, approximately 70% of EC can be attributed to overweight and physical inactivity, with obesity accounting for 57% (24). Obesity is a significant risk factor, and its association with EC is particularly prominent among all gynecological cancers. A robust positive correlation between obesity and EC has been consistently demonstrated (25, 26). As the body mass index (BMI) increased, the relative risk (RR) of developing EC also significantly rised (27). Setiawan et al. reported 10 cohort studies and 14 case-control studies in the meta-analysis to explore the risk factors of endometrial cancer. Among them, 77.7% of EC patients were white women with an average age of 62 years, and the proportion of postmenopausal women was as high as 79.6%. The study population came from the United States, Canada, Europe and Australia. When analyzing the association between BMI and EC, it was found that the RR for overweight individuals (BMI between 25kg/m² and 29.9kg/m²) was approximately 1.5; for first-degree obesity(BMI between 30kg/m² and 34.9kg/m²), the RR increased to 2.5; for second-degree obesity (BMI between 35kg/m² and 39.9kg/m²), the RR significantly increased to 4.5; for third-degree obesity (BMI ≥ 40kg/m²), and the RR markedly rose to 7.1 (28). These findings indicated the influence of BMI on type I tumors was greater than that on type II tumors. In a prospective cohort study conducted in Japan, 36,172 participants aged 40–59 years were enrolled. The results demonstrated that there was no significant association between BMI and the risk of type II EC (hazard ratio[HR] = 133%, 95%CI: 0.74-2.38) (29). Recent data suggested that obesity was driving the age of obesity-related cancers to younger age groups, particularly in EC, where there was a sustained and significant upward trend in age-specific incidence among women aged 50 years and younger (30, 31). Obesity constitutes a chronic inflammatory condition characterized by the establishment of a pro-inflammatory environment, primarily mediated through increased secretion levels of circulating biomarkers such as c-reactive protein, interleukin-6, and tumor necrosis factor-alpha (32). The main mechanism by which obesity promotes EC development was the aromatization of adrenal androgens in the surrounding adipose tissue into estrogen, resulting in a substantial elevation of estrogen levels in the body. These high levels of estrogen persistently stimulated the endometrial tissue, potentially leading to abnormal hyperplasia and even cancer of the endometrial tissue (33–35). For postmenopausal women, they faced dual risks: on the one hand, due to the lack of natural progesterone in the body, obesity exacerbated the excess state of non-antagonistic estrogen; On the other hand, their immune function tends to decline with advancing age (34). The combination of these two factors greatly increased the risk of EC.
3.1.2 Diabetes mellitus
Numerous studies have consistently established a positive correlation between diabetes and the risk of EC(Table 2). The association between diabetes and increased EC risk may involve multiple intricate mechanisms, including obesity, hyperglycemia, hyperinsulinemia, activation of the Insulin-like Growth Factor-1 (IGF-1) pathway, and elevated levels of inflammatory cytokines (36–38). High levels of blood glucose can induce angiogenesis and lead to significant changes in the concentration and types of immune cells as well as inflammatory factors (39). A meta-analysis conducted in 2007 demonstrated that diabetic patients had a twofold increased risk of developing EC compared to individuals without diabetes (40). Despite long-standing controversies regarding the comorbid impact and confounding factors between diabetes, obesity, and EC, comprehensive analyses of four BMI-adjusted cohort studies have confirmed diabetes as an independent risk factor for EC (41–43). Among them, patients with EC who have type 2 diabetes exhibited an augmented risk of mortality (44).
3.1.3 Polycystic ovary syndrome
Polycystic ovary syndrome (PCOS) is a heterogeneous endocrine disorder. Substantial research has demonstrated an elevated risk of EC among women with PCOS (45–47). This heightened risk stemmed from the typical association of PCOS with anovulation, which manifested as a persistent dominance of estrogen levels. This sustained estrogenic stimulation led to prolonged endometrial proliferation, thereby increasing the risk of EC(Figure 1). An early meta-analysis, which encompassed four case-control studies involving a total of 4,056 women, revealed that individuals with PCOS had a nearly three-fold increased risk of developing EC compared to the general population (odds ratio [OR]: 2.70; 95% confidence interval [CI]: 1.00-7.29) (48). The correlation was subsequently demonstrated in a later meta-analysis (49).

Figure 1. Polycystic ovary syndrome is associated with an increased risk of endometrial cancer in women. Patients with polycystic ovary syndrome often have abnormal levels of estrogen. Their follicles are difficult to mature and ovulate, resulting in the accumulation of small follicles in the ovaries and excessive secretion of estrogen. The absence of a luteinizing hormone peak hinders the formation of corpus luteum, leading to an imbalance in estrogen and progesterone secretion. The endometrium undergoes prolonged stimulation by elevated levels of estrogen, leading to sustained proliferation. Without the antagonism of progesterone, the endometrium fails to undergo the normal periodic exfoliation and instead continues to thicken, which may develop into atypical hyperplasia and increase the risk of EC.
It has been confirmed that PCOS is closely related to systemic and local low-grade inflammatory reactions (50). Existing data showed significant changes in the composition of immune cells among PCOS patients, including an increase in the number of macrophages and a decrease in the density of uterine natural killer cells (uNK) in the endometrium, which may affect immune homeostasis and endometrial receptivity (51, 52).
3.2 Other Factors
Tamoxifen is a critical drug widely used in endocrine therapy and for the prevention of hormone receptor-positive breast cancer. Its unique mechanism of action involved exerting anti-estrogenic effects on breast tissue while producing pro-estrogenic effects on the uterus (53). Long-term use of tamoxifen was associated with an increased EC risk, particularly in postmenopausal women. This risk was further exacerbated in patients who already have endometrial lesions, such as abnormal uterine bleeding or endometrial thickening (54–56). It is noteworthy that the majority of tamoxifen-induced EC cases were detectable at an early stage, and thus the overall prognosis was favorable (57). However, with prolonged duration of tamoxifen treatment, the risk of EC also escalates, potentially leading to more severe prognostic outcomes for patients. A study specifically indicated that patients treated with tamoxifen for 2 to 5 years had a relative risk of 2.2 (95% CI: 1.2-3.2) for EC compared to those not receiving tamoxifen treatment, while those treated for at least 5 years faced an even higher risk of 6.9 (95% CI: 2.4-19.4) (58). Another case-control study reported similar findings (59). Compared with patients not receiving tamoxifen therapy, patients treated with tamoxifen exhibited a significantly elevated risk of EC (OR = 2.4; 95%CI = 1.8-3.0). Moreover, the risk of EC increased progressively with longer durations of tamoxifen treatment(P < 0.001). Specifically, patients who received tamoxifen treatment for more than 5 years had an OR of 3.6 (95%CI = 2.6-4.8) (59). Both this study and prior research have consistently demonstrated a significant association between prolonged tamoxifen use (exceeding 5 years) and an increased risk of EC. Close attention should be paid to the condition of endometrial cancer in female patients with negative estrogen receptors during tamoxifen treatment (both premenopausal and postmenopausal) and for at least 5 years after the treatment. In addition, Lynch syndrome is a genetic disorder with an autosomal dominant inheritance mode (60). It is not only closely related to the occurrence of hereditary colorectal cancer (61), but also significantly associated with an increased risk of EC (62, 63). Lynch syndrome is primarily characterized by germline mutations in the DNA mismatch repair (MMR) system genes, including MSH2, MLH1, MSH6, and PMS2 (64). These genes play a crucial role in recognizing and repairing errors occurring during DNA replication. Mutations in MMR system genes resulted in dysregulation of intracellular immunity or abnormal cytokinesis. It interfered with the normal growth and division processes of cells, thereby significantly raising the risk of EC development. Of note, hereditary EC tends to exhibit an earlier onset compared to sporadic EC. That is to say, individuals with Lynch syndrome are at a higher risk of developing EC at a younger age (65–67).
4 Comparative analysis of tissue and molecular typing in endometrial cancer
4.1 Histological grading and typing
In 1973, the histological classification of EC was established by the Fédération Internationale de Gynécologie et d’Obstétrique (FIGO), classifying it into three grades: well-differentiated EC(G1), moderately differentiated EC (G2), and poorly differentiated EC (G3) (68). In 1983, Bokhman proposed a traditional binary model that classified EC into type I and type II (69). Type I EC, known as endometrioid adenocarcinoma, includes G1 and G2 subtypes. This type of cancer is estrogen-dependent and has been associated with obesity and excess estrogen (70, 71). Type II EC, referred to as non-endometrioid adenocarcinoma or non-estrogen-dependent carcinoma, encompasses G3 endometrioid carcinoma and non-endometrioid carcinoma, such as representative uterine papillary serous carcinoma (UPSC), clear cell carcinoma (CCC), and undifferentiated carcinoma (UDC) (72). In 2014, the EC classification by the World Health Organization (WHO) primarily focused on histomorphological features, categorizing it into distinct subtypes including endometrioid carcinoma, mucinous carcinoma, serous carcinoma, clear cell carcinoma, carcinosarcoma, neuroendocrine carcinoma, mixed cell adenocarcinoma, and undifferentiated carcinoma (73). In 2020, an updated version of the classification system for EC was introduced, formally incorporating renal adenocarcinoma, renal adenoid cancer, squamous cell carcinoma (im), and mucus carcinoma. Additionally, neuroendocrine tumor was separated from the original EC chapter and established as a distinct chapter focusing on neuroendocrine tumors of the female genital tract (74).
The WHO classification system is widely regarded as the “gold standard” in the field of EC. However, due to the limited global accessibility of certain molecular techniques, traditional histocytology continues to serve as the fundamental approach for classifying EC and remains the primary guideline for the clinical management of patients (75). In conditions permitting, the WHO’s new classification system emphasizes that immunohistochemical technique is highly recommended to complement histologic diagnosis for distinguishing between endometrial carcinoma and other types of endometrial malignancies, as this approach holds particular significance (76). As we continue to explore the histological aspects of EC, it is expected that future editions of the WHO classification will undergo further advancements.
4.2 Molecular typing
In 2013, the Cancer Genome Atlas (TCGA) research marked a profound shift in cancer classification from the traditional morphological basis to the molecular level. Based on distinct mutational profiles, EC have been classified into four major categories: POLE (DNA polymerase) mutant, microsatellite instability (MSI) high mutant, low copy number (CN-L), and high copy number (CN-H). Among these four types, POLE mutants exhibit the most favorable prognosis, while the CN-H subtype portends the worst (77).
In retrospect, despite the widespread adoption of the traditional binary staging system proposed by Bokhman in 1983 and the histopathological staging introduced by WHO in 2014, both have failed to adequately capture the inherent molecular heterogeneity in malignancies, thereby limiting their utility as accurate references for predicting patient clinical outcomes. Especially in type I EC, the phosphatase and tensin homolog (PTEN) gene mutations are among the most prevalent molecular events (77). PTEN, which acts as a tumor suppressor gene, encodes a protein that crucially regulates cell growth through modulating tyrosine kinase activity. Mutations in this gene lead to uncontrolled cellular proliferation (78). Additionally, Kras proto-oncogene aberrations played a pivotal role in the initiation and progression of type I EC by disrupting the regulatory mechanisms of normal cell division, further fueling tumorigenesis and development (78).
The TCGA classification is a clinically-oriented method that shows better prognostic relevance and optimized inter-observer consistency when compared to traditional morphological classification. The traditional classification of EC provides a specific research framework. However, it exhibits obvious limitations in terms of subtype classification uncertainty, prognostic variability, homogeneity, and the correlation with other factors contributing to endometrial cancer. Molecular classification represents a groundbreaking advancement in the realm of EC classification research, providing an important theoretical basis for personalized treatment strategies. With the deepening comprehension of the four different molecular subtypes of EC identified by TCGA, the related molecular genetic changes and clinicopathological significance have become increasingly important. To streamline operational procedures and enhance clinical feasibility, subsequent research has simplified the TCGA molecular classification by dividing EC into four distinct groups: POLE mutation (POLE mut), Mismatch Repair Deficient (MMRd), Non-Specific Molecular Profile (NSMP), and p53 Abnormality (p53abn). Within this simplified framework, POLE mut signified a favorable prognosis, while MMRd and NSMP indicated intermediate prognoses (79). Conversely, p53abn denoted an unfavorable prognosis (79). The proposed classification method demonstrated the potential to accurately predict the prognosis of EC and enhance the treatment effect.
To sum up, EC is a group of highly heterogeneous tumors, and different types of EC have distinct histological features, molecular patterns, and clinical significance. Therefore, there is an urgent need to develop novel molecular staging strategies based on traditional staging in order to achieve more precise diagnosis and treatment.
5 The human microbiome and cancer
The most prominent hypotheses for EC are obesity and excessive estrogen, while the potential role of reproductive tract microbiome in EC has not received adequate attention (80). As a result, the microbiome is often referred to as the forgotten organ. Indeed, both hormones and inflammation play an extremely important role in the pathogenesis of EC, constituting a complex equilibrium relationship. It is hypothesized that dysbiosis of the microbiota could alter immune and metabolic signaling, thereby exerting influence on various cancer-related features including chronic inflammation, epithelial barrier disruption, changes in cell proliferation and apoptosis, genomic instability, angiogenesis, and metabolic dysregulation (81). Currently, the most widely accepted link between microbial communities and cancer lies in their intricate influence on the host’s immune system. Specifically, microorganisms play a vital role in activating, shaping, and regulating the host’s immune responses. These microbes are capable of stimulating immune reactions. However, in cases of microbial community imbalance (termed dysbiosis), they may trigger the abnormal secretion of a series of pro-inflammatory cytokines or growth factors. These factors, under certain conditions, were believed to contribute to the development of cancer (82).
Existing studies have proven that chronic inflammation caused by the microbiome plays an important role in the occurrence, development, and metastasis of cancer (83–86). For instance, chronic gastritis caused by Helicobacter pylori was closely related to gastric adenocarcinoma (87); human papillomavirus infection made local tissues more susceptible to the invasion of pathogenic microorganisms, thereby inducing inflammatory reactions, which was closely related to cervical cancer (88); chronic inflammation and fibrosis of the liver caused by hepatitis viruses (types B and C) are closely related to liver cancer (89) (Table 3). At the same time, the carcinogenic mechanisms of these infection-associated cancers have been extensively studied, particularly focusing on the impact of viral and bacterial infections as well as resulting inflammation on cell proliferation, cell signaling, and genetic alterations. A rich microbiota has been detected in the reproductive tract of patients with endometrial hyperplasia and cancer, suggesting that these bacteria may play an infectious/inflammatory role in the onset of EC (90).
6 Female reproductive tract microbiome and endometrial cancer
6.1 Origin of the reproductive tract microbiome
During the early 20th century, there was a prevailing belief that the cervicovaginal epithelium exclusively harbored microorganisms, and it was widely held that a healthy uterus should be a sterile cavity (91). However, in the 21st century, particularly since 2007, the advent of next-generation sequencing (NGS) technology has revolutionized the field. It makes it possible to conduct a comprehensive quantitative assessment of the uterine microbiome, which was previously unattainable using traditional culture-based methods (92). The combination of this technological breakthrough and the availability of bacterial genome sequences has facilitated a deeper understanding of uterine microbial composition, surpassing the limitations of culture-based methods (81). Currently, it is widely acknowledged that the female reproductive tract (FRT) harbors an active microbiome. However, a comprehensive understanding of the potential roles that these microbiomes play in fertility and gynecological diseases remains elusive and merits thorough exploration (93). The composition of the lower genital tract microbiome undergoes dynamic changes throughout a woman’s lifespan, influenced by factors such as age, hormonal variations, sexual activities, and hygienic habits. Considering the FRT as an open system, the microbial communities in the upper reproductive tract exhibit diverse colonization pathways, including ascending microbiota from the lower reproductive tract, bacterial transmission through blood circulation, cervical tube abnormalities, retrograde infection of the uterus by abdominal microorganisms, gynecological surgery interventions, and direct drug inoculation methods (94–97). Among these pathways, one of the most certain and critical colonization pathways is the upward migration of bacteria from the vagina.
6.2 Vaginal microbiome and endometrial cancer
In the majority of women in their reproductive years, the microbiome within the lower FRT (including the vagina and cervix) was dominated by various species of Lactobacillus. Studies have shown that Lactobacillus species, including L.iners, L.crispatus, L.gasseri, and L.jensenii, dominate the vaginal microbiome and present five main types of community states (CSTs) (98). In these CSTs, Lactobacillus primarily dominated CST I, II, III and V (99). CST IV stands out due to its significant incorporation of anaerobic bacteria, such as Prevotella, Dialister, Atopobium, Gardnerella, and Megasphaera. This indicated a complex microbiota structure that was skewed towards an anaerobic environment. Given the vast diversity of Lactobacillus species and their varying antimicrobial capabilities, this underscored the significant alterations in composition and distribution of the vaginal microbiome that occur under different disease states (98).
Although the current research on the vaginal microbiome in patients with EC is relatively limited, a recent study has yielded significant new findings. Not only did the vaginal microbiome effectively distinguish between benign gynecological diseases and EC, but it also had the potential to predict cancer grade and histological type. Vaginal microbiome samples were prospectively collected from 61 patients with diverse racial and ethnic backgrounds who underwent hysterectomy (100). These patients were categorized into three groups: a control group comprising individuals with benign gynecological diseases, a low-grade EC group, and a high-grade EC group. The results revealed significant differences in both α-diversity and β-diversity among the three groups, and identified four vaginal CSTs associated with disease grade. Specifically, the benign diseases were predominantly clustered in CST1; low-grade EC exhibited a higher concentration in CST2; and high-grade EC was found in both CST3 and CST4 (P=0.036) (100). Furthermore, some studies have unveiled significant variations in compositions of cervical-vaginal microbiome among different pathological subtypes of EC. For example, in patients with high-grade EC, the abundances of Fusobacterium nucleatum and Prevotella bivia were significantly increased. Among them, Prevotella bivia has been demonstrated to up-regulate pro-inflammatory factors (such as lysosomal associated membrane protein 3 (LAMP3), STAT1, and TAP1), induce the overactive immune response, and lead to carcinogenesis and poor treatment outcomes (101). F. nucleatum can promote tumor growth and metastasis (102, 103). The FadA protein of F.nucleatum was capable of binding to the E-cadherin of host cells, which activated signaling pathways, thereby leading to the expression of oncogenic microRNA miR21 and an increase in pro-inflammatory cytokines such as TNF, IL-6, IL-8, and IL-1β (104–106). Once a tumor forms, F.nucleatum could bind to tumor cells via its Fap2 lectin, thereby leading to its accumulation within the tumor tissue (107). Conversely, low-grade EC patients exhibited significant increases in the abundances of Clostridium spp., Corynebacterium amycolatum, L.gasseri, and Peptoniphilus duerdeni; only Staphylococcus epidermidis exhibited a lower abundance (100). The presence of specific bacterial species, such as vaginal Proteobacteria and Porphyromonas, has been observed to be correlated with a high vaginal pH and the occurrence of EC (9). Another study found that the vaginal and cervical microbial ecology of healthy females was predominantly shaped by several key species, including L.crispatus, L.iners, L.gasseri, and G.vaginalis (108). However, a significant alteration in bacterial frequencies was observed within vaginal samples when focusing on patients with EC (P=0.003) (108). The detection frequencies of two specific bacteria, Mobiluncus curtisii and F.nucleatum, were aberrantly elevated in the vaginal samples of EC patients, with this increase being more pronounced in the vagina compared to the cervix (35.5% vs 17.7%, P=0.009; 38.5% vs 24.0%, P=0.043) (108). Furthermore, the composition of vaginal microbiome exhibited significant differences among different subtypes of EC. Gressel et al. investigated the characteristics of vaginal microbiome in patients with endometrioid carcinoma and uterine serous carcinoma, revealing a substantial decrease in the alpha diversity of vaginal microbiome in patients with uterine serous carcinoma compared to those with endometrioid carcinoma (Chao1 index, P=0.004; Fisher index, P=0.007) (109).
These findings confirmed that the composition and abundance of the vaginal microbiome may serve as robust predictors for EC grading, typing, histological features, and diagnosis, providing a novel microbiological perspective.
6.3 Endometrial microbiome and endometrial cancer
Indeed, the specimen collection process for the endometrial microbiome often requires invasive or somewhat traumatic procedures, necessitating a heightened awareness of potential contamination during sampling. Additionally, the complexity introduced by factors such as the inherent cyclical changes of the endometrium, patient age, and ethnicity should be taken into consideration. These technical challenges and diverse influencing factors have in turn limited the scope of research on the endometrial microbiome. Nevertheless, it is noteworthy that despite these constraints, existing studies focusing on specific populations (e.g., infertility patients, EC patients) or under particular conditions (such as pre- and post- assisted reproductive technology treatments) have robustly demonstrated the crucial role of endometrial microbiome in the initiation and progression of EC. The microbial community of the endometrium exhibited high microbial diversity, yet its overall relative abundance remained relatively low (9, 80, 110). According to the currently available data, it has been observed that the dominant microbial phyla in the uterine primarily consist of Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria (81). Most studies have indicated that the Firmicutes phylum dominates, with Lactobacillus being a significant genus whose importance cannot be overlooked. This recent finding is highly consistent with previous in-depth investigations into the vaginal microbiome (111, 112). Compared to the benign endometrial tissues, the microbial α-diversity in the endometrium of EC patients was significantly elevated (P=0.04). Additionally, there was a notable decrease in Lactobacillus abundance and a significant increase in specific bacterial taxa such as Anaerobic cocci, Porphyromonas, Prevotella, Fusobacterium, Acidobacteria, Firmicutes, Spirochetes, Actinobacteria, and Proteobacteria (9, 113). Significantly, Bacteroides and Faecalibacterium exhibited a strong association with the EC patient cohort. The abundance of Bacteroides increased in the mucosa-associated lymphoid tissues of cancer patients and triggered immune responses, which may be closely related to the poor prognosis of cancer lesions (114–116). Besides, microorganisms belonging to the genera Porphyromonas and Atopobium exhibited a high prevalence in EC samples, whereas they were virtually absent in normal endometrial samples (9). The abundance of Micrococcus on the endometrium of EC patients was significantly higher compared to benign uterine lesions (117). The abundance of Micrococcus was positively correlated with the levels of IL-6 and IL-17 mRNA in endometrial tissues, thereby inducing excessive expression of immune response (117). Further speculation suggested that these abnormal alterations in the reproductive tract microenvironment, particularly the increased microbial diversity and the enrichment of specific pathogens, might contribute to the onset and progression of endometrial inflammation. The persistent inflammatory state of the endometrium, as a crucial risk factor for EC, could facilitate cancer cell proliferation and invasion through various mechanisms, thereby accelerating cancer progression (118, 119). Moreover, notable differences in endometrial microbiome were observed between EC tissues and their adjacent non-cancerous tissues. A study has conclusively demonstrated that, compared to non-cancerous tissues, the endometrial microbial communities within cancer tissues exhibit a significant increase in both α-diversity and evenness (P<0.01), highlighting the intricate relationship between the endometrial microbiome and the initiation and progression of EC (120).
These findings underscored the significance of the endometrial microbiome in the onset, progression, diagnosis, and prognosis of EC. An increase in the diversity of endometrial microbial communities and the enrichment of specific pathogens may both contribute to the initiation and progression of EC.
7 Immunity, endometrial microbiome, and endometrial cancer
7.1 Immune response of the vaginal microbiome in endometrial cancer
The interaction between bacteria and their hosts involves the engagement of pattern recognition receptors (PRRs), including dectin-1 receptors, toll-like receptors (TLRs), and nucleotide-binding oligomerization domain (NOD)-like receptors. These PRRs are widely distributed on both squamous epithelial cells of the vaginal lining and columnar epithelial cells of the FRT. They play a crucial role in facilitating the recognition and sensing of microorganisms by host cells, thereby influencing the survival of these bacteria within the reproductive tract. Essentially, the survival of commensal bacteria in the FRT is intimately linked to the host’s ability to develop immunological tolerance towards them.
In recent years, numerous in vitro and in vivo studies demonstrated that dysbiosis of the vaginal microbiome could disrupt local microhomeostasis and alter immune parameters (including immune cells and cytokines), thereby inducing pro-inflammatory responses. A recent in vitro study found that certain bacteria in the reproductive tract had the ability to directly induce the expression of pro-inflammatory cytokines, such as vaginae and micromonas inducing pro-inflammatory cytokines IL-1α, IL-1β, IL-17α, and TNF-α. Furthermore, they also altered the transcription of CCL13, CCL8, CXCL2, IL22, and IL9 transcripts (121).
Cytokines represent a class of small soluble proteins synthesized by various cells types in response to infection and inflammation. For instance, interleukin-6 (IL-6), interleukin-8 (IL-8), and interleukin-17 (IL-17) are pro-inflammatory cytokines that play a crucial role in inflammation and tumorigenesis. Notably, IL-6, IL-8, and IL-17 have been proven to be associated with EC. There were significant differences in the mRNA expression of these cytokines between women with EC and those with benign uterine lesions in the endometrial microenvironment (117).
Micrococcus, a Gram-positive bacterial genus classified under the phylum Actinomycetota, naturally colonizes the human skin as part of the commensal microbiota. In recent years, research has uncovered potential associations between an increased abundance of Micrococcus and various types of cancer, including cervical cancer (122) and colorectal cancer (123). A recent groundbreaking study has found a positive correlation between the abundance of Micrococcus in the endometrial microbiome and the mRNA expression levels of pro-inflammatory cytokines IL-6 and IL-17. This discovery provides compelling evidence supporting a close relationship between Micrococcus, the immune system, and EC (117).
In addition, the decline in estrogen levels in vaginal epithelial cells, whether resulting from natural menopause, interruption of hormonal therapy, or cytotoxic cancer treatment outcomes, triggered a shift in the vaginal microbiome towards a non-Lactobacillus-dominated microbiome. As a result, there was an increase in the abundance of anaerobic bacteria such as Gardnerella and Atopobium. An overgrowth of facultative or obligate anaerobic bacteria in the FRT can result in microbial dysbiosis, thereby adversely impacting the healthy vaginal environment (122). Macrophage inflammatory protein (MIP-3α), also known as the chemokine CCL20, has the ability to infiltrate various types of immune cells in the tumor microenvironment (124), including dendritic cells (DCs), regulatory T lymphocytes (Tregs), and Th17 helper cells (125). In studies on EC, scholars have observed an upregulation in the expression of CCL20 and have highlighted its ability to accelerate invasion and induce epithelial-mesenchymal transition (EMT) in EC cells (126, 127). Furthermore, the presence of Atopobium in the vagina was associated with the activation of various pro-inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), MIP-3α, and the transcription factor NF-KB (123, 128). Short-chain fatty acids (SCFAs) within the vagina contributed to the development of a pro-inflammatory environment. In cases of vaginal dysbiosis, elevated concentrations of SCFAs increases can enhance the production of IL-8 and TNF-α induced by TLR2 and TLR7 ligands. SCFAs can also facilitate the generation of pro-inflammatory cytokines through partially inducing the production of reactive oxygen species (ROS) (129, 130), thereby exacerbating the inflammatory process and potentially promoting tumor cell proliferation and metastasis, consequently increasing the risk of EC.
7.2 Immune response of the endometrial microbiome in endometrial cancer
In recent years, the investigation of the endometrial microbiome and its intricate interplay with the immune system has garnered significant scholarly attention. The endometrial microbiome maintains a dynamic equilibrium, actively participating in the modulation of immune defense mechanisms in the endometrium. Additionally, their metabolic byproducts exerted influence over crucial physiological processes including endometrial cell proliferation, differentiation, and apoptosis. The maintenance of these functions is crucial to the health of the endometrium. Gynecological cancers originate in the female reproductive organs, and dysbiosis of the microbiota within these organs, particularly imbalances in the endometrial microbiota, can impact the host immune system, thereby increasing the risk of developing EC (Figure 2).

Figure 2. Immune mechanism of endometrial microbiome in the development of endometrial cancer. Dysregulation of the microbial community in the host’s endometrium results in an increase in pathogenic bacteria, which may subsequently induce the release of pro-inflammatory cytokines or toxins. Metabolic products from these pathogens, such as short-chain fatty acids (SCFAs), have the potential to activate the immune system, thereby inducing the release of pro-inflammatory cytokines like TNF-α, IL-1β, IL-6, IL-17, and IL-1α. These pro-inflammatory factors trigger local or systemic inflammatory responses. Prolonged exposure to a pro-inflammatory state within the host’s internal environment can lead to cellular damage. Chronic inflammation and cell proliferation may contribute to abnormal changes and carcinogenesis in endometrial cells. An uncontrolled proliferation of cells ultimately results in abnormal hyperplasia, which may progress into endometrial cancer. Therefore, maintaining microbial homeostasis and preventing the invasion of pathogens are crucial for preserving host health and impeding the onset and progression of cancer. Investigating the relationship between the microbiome and endometrial cancer holds paramount significance in elucidating the pathogenesis of gynecological cancers, particularly endometrial cancer, as well as in developing novel preventive and therapeutic strategies.
8 Roles of diverse cytokines in the pathogenesis of endometrial cancer
8.1 Interleukin-6 and endometrial cancer
IL-6, the pivotal member of the IL-6 cytokine family, acts as a soluble factor that stimulates T cells to secrete and modulate B cell responses (131). The signal transduction of IL-6 relies on a hexameric high-affinity complex composed of IL-6, IL-6 receptor α (IL-6Rα), and glycoprotein 130 (gp130), which serves as the shared signaling chain among IL-6 subfamily receptors, thereby facilitating the proliferation of EC cells (132). The production and expression of IL-6 were mainly influenced by ERK-NF-κB signaling pathway (133–135). Matrix metalloproteinases (MMPs) were intimately linked to cancer progression and promote cancer cell invasion and migration by degrading extracellular matrix. Studies demonstrated that IL-6 was involved in estrogen E2-triggered EC cell migration and invasion, accompanied by upregulated expression of MMP-2. Moreover, apart from MMP-2, IL-6 can exacerbate the invasive capacity of EC cells by inducing the release of MMP-9 (136, 137)(Figure 3).

Figure 3. The role of different cytokines in the pathophysiology of endometrial cancer. Diverse cytokines can not only influence cell proliferation and differentiation but also modulate the immune system and inflammatory response, thereby impacting the development of endometrial cancer (1). Interleukin-1β, interleukin-6 and interleukin-8 can trigger inflammation, promote the progression of chronic inflammation, activate signaling pathways, stimulate cell proliferation and immune cell recruitment, and promote the secretion of matrix metalloproteinases to accelerate cancer cell invasion and metastasis (2).The binding of Chemokines to receptor proteins can form signaling axes, which can activate signaling pathways, enhance tumor cell migration, invasion, and angiogenesis, regulate the tumor microenvironment, and participate in the onset of endometrial cancer (3). Bacteria and their metabolites can act as exogenous stimuli, recognized and bound by receptors on cells, transmitting signals to the nucleus to promote the transcription of specific cytokines, thereby influencing the occurrence and development of endometrial cancer.
8.2 Interleukin-8 and endometrial cancer
IL-8 is a major mediator of inflammation and acts as a chemotactic agent for various immune cells populations (138). Studies have shown that an upregulation in the expression level of IL-8 mRNA in EC patients, suggesting a significant role of this cytokine in promoting neutrophils infiltration into tumor tissue (117, 139). In addition, IL-8 exerts its biological effects mainly by binding to its receptors (such as CXCR1 and CXCR2). The expression level of IL-8 receptors may potentially be higher in EC than in normal endometrial tissues. This high expression may enhance the sensitivity of endometrial cancer cells to IL-8, thus promoting the proliferation, invasion and metastasis of tumor cells (138). Studies in vitro have found that aspergillus albacus can induce NF-κB signaling activity and increase the secretion of IL-8 (140), which may promote the occurrence and development of EC (Figure 3).
8.3 Chemokines and endometrial cancer
Chemokines are a class of secretory proteins produced by cancer cells and leukocytes infiltrating the tumor microenvironment. These chemokines can regulate cellular behavior by interacting with specific G protein-coupled seven-helix chemokine receptors on the cell surface (141). Notably, chemokines not only participate in regulating the aging process and inducing apoptosis of cancer cells, but also play a crucial role in the progression and metastatic cascade of EC. Specifically, they promote epithelial-mesenchymal transition (EMT), which is a biological process that provides key molecular mechanisms supporting cancer cell invasion and metastasis (142). Numerous studies have robustly demonstrated the pivotal roles of various chemokines in the progression of diverse cancer types, including ovarian, breast, and lung cancers (143–146). Consequently, the expression patterns of chemokines in patients with EC have emerged as a focal point of intense research. In particular, the CXCL12 ligand and its receptors, CXCR4 and CXCR7 form an axis contributing to tumor progression and metastatic cascade of EC (147)(Figure 3). CXCL12 exhibited a significant correlation with an unfavorable prognosis across various cancer types and played a pivotal role by activating distinct cellular signaling pathways (141, 147–149). In the context of EC research, the expression level of CXCR4 was significantly upregulated compared to atypical hyperplasia, simple hyperplasia, and normal endometrial cells (150). The data indicated that CXCR4 was detected in up to 69.23% of EC tissue samples (150). Moreover, the expression status of CXCR7 was closely associated with reduced overall survival rates across diverse cancer patient populations (151, 152). Collectively, the intricate CXCL12-CXCR4-CXCR7 signaling axis is emerging as a pivotal predictor for early diagnosis and assessment of unfavorable prognosis in EC. A profound understanding of the molecular mechanisms underpinning this axis is of paramount importance.
However, the precise expression profiles of these chemokines throughout different stages of EC remain undetermined (153). The CCL2-CCR2 signaling pathway plays a pivotal role in the intricate processes of cancer invasion within the tumor microenvironment and lymphatic dissemination (154, 155). A key function of CCL2 is its ability to elicit an invasive phenotype in cancer cells and recruit monocytes to the tumor site, thereby implying that overexpression of CCL2 may have therapeutic implications in gynecological malignancies (156, 157). For instance, the expression of CCL2 served as a biomarker in cervical cancer, while an upregulation of CCR2 expression had been associated with reduced overall survival rates (158). The expression of CCL2 in breast cancer had been identified as an independent risk factor for disease-free survival (DFS) and was significantly correlated with a poor prognosis (159).
8.4 Other cytokines and endometrial cancer
The expression of IL-17A within EC tissues has been recently discovered to exhibit a significant elevation, suggesting its potential as a crucial pro-inflammatory factor that promotes endometrial carcinogenesis (160). IL-17A has been confirmed as a vital CD4 T cell-derived pro-inflammatory cytokine, which significantly contributes to tumor angiogenesis, cell proliferation, and invasion in various solid tumors, including breast and cervical cancers (161–163). Tumor necrosis factor alpha (TNF-α), a potent inflammatory cytokine, not only augments local estrogen synthesis in endometrial cells but also stimulates the proliferation of human endometrial cells through adipocyte paracrine mechanisms (164). This aforementioned observation indirectly underscores a potential causal link between obesity and the development of EC. IL-1ra may play a pro-inflammatory role, thereby implying its potential involvement in the promotion of endometrial cancer development (165–167). However, an experimental study has provided empirical evidence supporting the rationality of IL-1ra’s mechanism in playing a protective role against cancer. The results demonstrated that interleukin-1 receptor antagonist (IL-1ra/IL1RN) was negatively correlated with both the overall risk of EC (r=0.86, 95% CI:0.80-0.93, P=2.23×10−4) and the risk of endometrioid subtype cancer (r=0.85, 95% CI:0.78-0.94, P=7.9×10−4). And a positive correlation was observed between interferon-induced monokine (MIG/CXCL9) and the risk of non-endometrioid endometrial cancer (r=3.73, 95% CI:1.86-7.47, P=2×10−4) (168). The presented compelling evidences establishes a correlation between cytokines and EC, but further verification and in-depth investigation into the underlying mechanisms are still needed.
9 Conclusions
EC is one of the most common gynecological malignancies, and its incidence is increasing year by year. Obesity, diabetes, PCOS, and other related conditions are all significant risk factors for EC. The reproductive tract microbiome plays a crucial role in maintaining local immune homeostasis. Once the balance of the microbiome is destroyed, it may trigger abnormal immune responses and promote the occurrence and development of EC. Cytokines, being pivotal immunomodulatory molecules, not only regulate tumor cell proliferation, apoptosis and migration but also affect immune cells function and angiogenesis within the tumor microenvironment. Due to the biological heterogeneity of EC, reaching a unified consensus on its molecular subtypes remains a great challenge. Given the intricate interactions between the microbiome and EC, the mechanisms by which specific microbiome and its metabolites regulate the immune microenvironment and thus affect tumorigenesis remain to be further elucidated. Combining microbiome data with multi-omics information, including genomics, proteomics, and transcriptomics data, may provide a more precise way to predict the risk of EC onset and recurrence. In the future, it is worthwhile to explore targeted interventions targeting specific microbiota and evaluate their impact on the therapeutic efficacy of EC. EC research is full of opportunities and challenges. We eagerly look forward to further breakthroughs in this area in the coming years.
Author contributions
BL: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Software, Supervision, Validation, Writing – original draft. JT: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Software, Supervision, Validation, Writing – original draft. JL: Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Project administration, Software, Supervision, Validation, Writing – original draft. XW: Software, Writing – original draft. GL: Supervision, Writing – original draft. HL: Supervision, Writing – original draft. TL: Supervision, Writing – original draft. HG: Funding acquisition, Resources, Visualization, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by China Scholarship Council (Grant NO. 202008430075) and an Ottawa Hospital Research Institute-Taggart-Parkes Foundation.
Acknowledgments
We would like to express our deepest gratitude to all those who have contributed to the successful completion of this comprehensive review. We acknowledge the financial support from China Scholarship Council, grant number [No. 202008430075] and Ottawa Hospital Research Institute-Taggart-Parkes Foundation, which has made it possible for us to undertake this research endeavor.
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2021) 71:209–49. doi: 10.3322/caac.21660
2. Siegel RL, Miller KD, and Jemal A. Cancer statistics, 2015. CA Cancer J Clin. (2015) 65:5–29. doi: 10.3322/caac.21254
3. Trabert B, Wentzensen N, Felix AS, Yang HP, Sherman ME, and Brinton LA. Metabolic syndrome and risk of endometrial cancer in the United States: a study in the SEER-medicare linked database. Cancer Epidemiol Biomarkers Prev. (2015) 24:261–7. doi: 10.1158/1055-9965.EPI-14-0923
4. Bjørge T, Häggström C, Ghaderi S, Nagel G, Manjer J, Tretli S, et al. BMI and weight changes and risk of obesity-related cancers: a pooled European cohort study. Int J Epidemiol. (2019) 48:1872–85. doi: 10.1093/ije/dyz188
5. Sheikh MA, Althouse AD, Freese KE, Soisson S, Edwards RP, Welburn S, et al. USA endometrial cancer projections to 2030: should we be concerned? Future Oncol. (2014) 10:2561–8. doi: 10.2217/fon.14.192
6. Wu Y, Sun W, Liu H, and Zhang D. Age at menopause and risk of developing endometrial cancer: A meta-analysis. BioMed Res Int. (2019) 2019:8584130. doi: 10.1155/2019/8584130
7. Morice P, Leary A, Creutzberg C, Abu-Rustum N, and Darai E. Endometrial cancer. Lancet. (2016) 387:1094–108. doi: 10.1016/S0140-6736(15)00130-0
8. Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, et al. Cancer statistics in China, 2015. CA Cancer J Clin. (2016) 66:115–32. doi: 10.3322/caac.21338
9. Walther-António MRS, Chen J, Multinu F, Hokenstad A, Distad TJ, Cheek EH, et al. Potential contribution of the uterine microbiome in the development of endometrial cancer. Genome Med. (2016) 8:122. doi: 10.1186/s13073-016-0368-y
10. Lee JE, Lee S, Lee H, Song YM, Lee K, Han MJ, et al. Association of the vaginal microbiota with human papillomavirus infection in a Korean twin cohort. PloS One. (2013) 8:e63514. doi: 10.1371/journal.pone.0063514
11. Gu B, Shang X, Yan M, Li X, Wang W, Wang Q, et al. Variations in incidence and mortality rates of endometrial cancer at the global, regional, and national levels, 1990-2019. Gynecol Oncol. (2021) 161:573–80. doi: 10.1016/j.ygyno.2021.01.036
12. Lu KH and Broaddus RR. Endometrial cancer. N Engl J Med. (2020) 383:2053–64. doi: 10.1056/NEJMra1514010
13. Clarke MA, Devesa SS, Harvey SV, and Wentzensen N. Hysterectomy-corrected uterine corpus cancer incidence trends and differences in relative survival reveal racial disparities and rising rates of nonendometrioid cancers. J Clin Oncol. (2019) 37:1895–908. doi: 10.1200/JCO.19.00151
14. Giaquinto AN, Miller KD, Tossas KY, Winn RA, Jemal A, and Siegel RL. Cancer statistics for african american/black people 2022. CA Cancer J Clin. (2022) 72:202–29. doi: 10.3322/caac.21718
15. Makker V, MacKay H, Ray-Coquard I, Levine DA, Westin SN, Aoki D, et al. Endometrial cancer. Nat Rev Dis Primers. (2021) 7:88. doi: 10.1038/s41572-021-00324-8
16. Felix AS, Weissfeld JL, Stone RA, Bowser R, Chivukula M, Edwards RP, et al. Factors associated with Type I and Type II endometrial cancer. Cancer Causes Control. (2010) 21:1851–6. doi: 10.1007/s10552-010-9612-8
17. Cirisano FD, Robboy SJ, Dodge RK, Bentley RC, Krigman HR, Synan IS, et al. Epidemiologic and surgicopathologic findings of papillary serous and clear cell endometrial cancers when compared to endometrioid carcinoma. Gynecol Oncol. (1999) 74:385–94. doi: 10.1006/gyno.1999.5505
18. Sorosky JI. Endometrial cancer. Obstet Gynecol. (2012) 120:383–97. doi: 10.1097/AOG.0b013e3182605bf1
19. Cronin KA, Lake AJ, Scott S, Sherman RL, Noone AM, Howlader N, et al. Annual Report to the Nation on the Status of Cancer, part I: National cancer statistics. Cancer. (2018) 124:2785–800. doi: 10.1002/cncr.v124.13
20. Njoku K, Barr CE, Hotchkies L, Quille N, Wan YL, and Crosbie EJ. Impact of socio-economic deprivation on endometrial cancer survival in the North West of England: a prospective database analysis. BJOG. (2021) 128:1215–24. doi: 10.1111/1471-0528.16618
21. Zhang S, Gong TT, Liu FH, Jiang YT, Sun H, Ma XX, et al. Global, regional, and national burden of endometrial cancer, 1990-2017: results from the global burden of disease study, 2017. Front Oncol. (2019) 9:1440. doi: 10.3389/fonc.2019.01440
22. Svanvik T, Marcickiewicz J, Sundfeldt K, Holmberg E, and Strömberg U. Sociodemographic disparities in stage-specific incidences of endometrial cancer: a registry-based study in West Sweden, 1995-2016. Acta Oncol. (2019) 58:845–51. doi: 10.1080/0284186X.2019.1581947
23. Desravines N, Hazimeh D, Desjardins MR, and Wethington SL. Healthcare utilization in women diagnosed with endometrial cancer: A survey-based study. Gynecol Oncol. (2024) 186:1–8. doi: 10.1016/j.ygyno.2024.03.015
24. Calle EE and Kaaks R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer. (2004) 4:579–91. doi: 10.1038/nrc1408
25. Nakamura K, Hongo A, Kodama J, and Hiramatsu Y. Fat accumulation in adipose tissues as a risk factor for the development of endometrial cancer. Oncol Rep. (2011) 26:65–71. doi: 10.3892/or.2011.1259
26. Xu WH, Matthews CE, Xiang YB, Zheng W, Ruan ZX, Cheng JR, et al. Effect of adiposity and fat distribution on endometrial cancer risk in Shanghai women. Am J Epidemiol. (2005) 161:939–47. doi: 10.1093/aje/kwi127
27. Gómez-Banoy N, Ortiz EJ, Jiang CS, Dagher C, Sevilla C, Girshman J, et al. Body mass index and adiposity influence responses to immune checkpoint inhibition in endometrial cancer. J Clin Invest. (2024) 134:e180516. doi: 10.1172/JCI180516
28. Setiawan VW, Yang HP, Pike MC, McCann SE, Yu H, Xiang YB, et al. Type I and II endometrial cancers: have they different risk factors? J Clin Oncol. (2013) 31:2607–18. doi: 10.1200/JCO.2012.48.2596
29. Kawachi A, Shimazu T, Budhathoki S, Sawada N, Yamaji T, Iwasaki M, et al. Association of BMI and height with the risk of endometrial cancer, overall and by histological subtype: a population-based prospective cohort study in Japan. Eur J Cancer Prev. (2019) 28:196–202. doi: 10.1097/CEJ.0000000000000449
30. Koroukian SM, Dong W, and Berger NA. Changes in age distribution of obesity-associated cancers. JAMA Netw Open. (2019) 2:e199261. doi: 10.1001/jamanetworkopen.2019.9261
31. Sung H, Siegel RL, Rosenberg PS, and Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. (2019) 4:e137–47. doi: 10.1016/S2468-2667(18)30267-6
32. Naqvi A, MacKintosh ML, Derbyshire AE, Tsakiroglou AM, Walker TDJ, McVey RJ, et al. The impact of obesity and bariatric surgery on the immune microenvironment of the endometrium. Int J Obes (Lond). (2022) 46:605–12. doi: 10.1038/s41366-021-01027-6
33. Kitson SJ and Crosbie EJ. Optimising endometrial cancer survivorship through lifestyle change. Gynecol Oncol. (2023) 179:A1–2. doi: 10.1016/j.ygyno.2023.11.032
34. Kaaks R, Lukanova A, and Kurzer MS. Obesity, endogenous hormones, and endometrial cancer risk: a synthetic review. Cancer Epidemiol Biomarkers Prev. (2002) 11:1531–43.
35. Marin AG, Filipescu A, and Petca A. The role of obesity in the etiology and carcinogenesis of endometrial cancer. Cureus. (2024) 16:e59219. doi: 10.7759/cureus.59219
36. Saed L, Varse F, Baradaran HR, Moradi Y, Khateri S, Friberg E, et al. The effect of diabetes on the risk of endometrial Cancer: an updated a systematic review and meta-analysis. BMC Cancer. (2019) 19:527. doi: 10.1186/s12885-019-5748-4
37. Shikata K, Ninomiya T, and Kiyohara Y. Diabetes mellitus and cancer risk: review of the epidemiological evidence. Cancer Sci. (2013) 104:9–14. doi: 10.1111/cas.2013.104.issue-1
38. Pliszka M and Szablewski L. Associations between diabetes mellitus and selected cancers. Int J Mol Sci. (2024) 25:7476. doi: 10.3390/ijms25137476
39. Yang T, Qi F, Guo F, Shao M, Song Y, Ren G, et al. An update on chronic complications of diabetes mellitus: from molecular mechanisms to therapeutic strategies with a focus on metabolic memory. Mol Med. (2024) 30:71. doi: 10.1186/s10020-024-00824-9
40. Friberg E, Orsini N, Mantzoros CS, and Wolk A. Diabetes mellitus and risk of endometrial cancer: a meta-analysis. Diabetologia. (2007) 50:1365–74. doi: 10.1007/s00125-007-0681-5
41. Friberg E, Mantzoros CS, and Wolk A. Diabetes and risk of endometrial cancer: a population-based prospective cohort study. Cancer Epidemiol Biomarkers Prev. (2007) 16:276–80. doi: 10.1158/1055-9965.EPI-06-0751
42. Terry P, Baron JA, Weiderpass E, Yuen J, Lichtenstein P, and Nyrén O. Lifestyle and endometrial cancer risk: a cohort study from the Swedish Twin Registry. Int J Cancer. (1999) 82:38–42. doi: 10.1002/(SICI)1097-0215(19990702)82:1<38::AID-IJC8>3.0.CO;2-Q
43. Anderson KE, Anderson E, Mink PJ, Hong CP, Kushi LH, Sellers TA, et al. Diabetes and endometrial cancer in the Iowa women’s health study. Cancer Epidemiol Biomarkers Prev. (2001) 10:611–6.
44. Njoku K, Agnew HJ, and Crosbie EJ. Impact of type 2 diabetes mellitus on endometrial cancer survival: A prospective database analysis. Front Oncol. (2022) 12:899262. doi: 10.3389/fonc.2022.899262
45. Yin W, Falconer H, Yin L, Xu L, and Ye W. Association between polycystic ovary syndrome and cancer risk. JAMA Oncol. (2019) 5:106–7. doi: 10.1001/jamaoncol.2018.5188
46. Fearnley EJ, Marquart L, Spurdle AB, Weinstein P, Webb PM, and Australian Ovarian Cancer Study Group and Australian National Endometrial Cancer Study Group. Polycystic ovary syndrome increases the risk of endometrial cancer in women aged less than 50 years: an Australian case-control study. Cancer Causes Control. (2010) 21:2303–8. doi: 10.1007/s10552-010-9658-7
47. Indhavivadhana S, Rattanachaiyanont M, Wongwananuruk T, Techatraisak K, Rayasawath N, and Dangrat C. Endometrial neoplasia in reproductive-aged Thai women with polycystic ovary syndrome. Int J Gynaecol Obstet. (2018) 142:170–5. doi: 10.1002/ijgo.2018.142.issue-2
48. Chittenden BG, Fullerton G, Maheshwari A, and Bhattacharya S. Polycystic ovary syndrome and the risk of gynaecological cancer: a systematic review. Reprod BioMed Online. (2009) 19:398–405. doi: 10.1016/S1472-6483(10)60175-7
49. Amiri M, Bidhendi-Yarandi R, Fallahzadeh A, Marzban Z, and Ramezani Tehrani F. Risk of endometrial, ovarian, and breast cancers in women with polycystic ovary syndrome: A systematic review and meta-analysis. Int J Reprod Biomed. (2022) 20:893–914. doi: 10.18502/ijrm.v20i11.12357
50. Hu C, Pang B, Ma Z, and Yi H. Immunophenotypic profiles in polycystic ovary syndrome. Mediators Inflamm. (2020) 2020:5894768. doi: 10.1155/2020/5894768
51. Piltonen TT, Chen J, Erikson DW, Spitzer TLB, Barragan F, Rabban JT, et al. Mesenchymal stem/progenitors and other endometrial cell types from women with polycystic ovary syndrome (PCOS) display inflammatory and oncogenic potential. J Clin Endocrinol Metab. (2013) 98:3765–75. doi: 10.1210/jc.2013-1923
52. Oróstica L, Astorga I, Plaza-Parrochia F, Vera C, García V, Carvajal R, et al. Proinflammatory environment and role of TNF-α in endometrial function of obese women having polycystic ovarian syndrome. Int J Obes (Lond). (2016) 40:1715–22. doi: 10.1038/ijo.2016.154
53. Gennari A, André F, Barrios CH, Cortés J, de Azambuja E, DeMichele A, et al. ESMO Clinical Practice Guideline for the diagnosis, staging and treatment of patients with metastatic breast cancer. Ann Oncol. (2021) 32:1475–95. doi: 10.1016/j.annonc.2021.09.019
54. Emons G, Mustea A, and Tempfer C. Tamoxifen and endometrial cancer: A janus-headed drug. Cancers (Basel). (2020) 12:2535. doi: 10.3390/cancers12092535
55. Ghanavati M, Khorshidi Y, Shadnoush M, Akbari ME, Ardehali SH, Chavarri-Guerra Y, et al. Tamoxifen use and risk of endometrial cancer in breast cancer patients: A systematic review and dose-response meta-analysis. Cancer Rep (Hoboken). (2023) 6:e1806. doi: 10.1002/cnr2.1806
56. Lee M, Piao J, and Jeon MJ. Risk factors associated with endometrial pathology in premenopausal breast cancer patients treated with tamoxifen. Yonsei Med J. (2020) 61:317–22. doi: 10.3349/ymj.2020.61.4.317
57. Hoogendoorn WE, Hollema H, van Boven HH, Bergman E, de Leeuw-Mantel G, Platteel I, et al. Prognosis of uterine corpus cancer after tamoxifen treatment for breast cancer. Breast Cancer Res Treat. (2008) 112:99–108. doi: 10.1007/s10549-007-9823-1
58. Bergman L, Beelen ML, Gallee MP, Hollema H, Benraadt J, and van Leeuwen FE. Risk and prognosis of endometrial cancer after tamoxifen for breast cancer. Comprehensive Cancer Centres’ ALERT Group. Assessment of Liver and Endometrial cancer Risk following Tamoxifen. Lancet. (2000) 356:881–7. doi: 10.1016/S0140-6736(00)02677-5
59. Swerdlow AJ, Jones ME, and British Tamoxifen Second Cancer Study Group. Tamoxifen treatment for breast cancer and risk of endometrial cancer: a case-control study. J Natl Cancer Inst. (2005) 97:375–84. doi: 10.1093/jnci/dji057
60. Peltomäki P, Nyström M, Mecklin JP, and Seppälä TT. Lynch syndrome genetics and clinical implications. Gastroenterology. (2023) 164:783–99. doi: 10.1053/j.gastro.2022.08.058
61. Abu-Ghazaleh N, Kaushik V, Gorelik A, Jenkins M, and Macrae F. Worldwide prevalence of Lynch syndrome in patients with colorectal cancer: Systematic review and meta-analysis. Genet Med. (2022) 24:971–85. doi: 10.1016/j.gim.2022.01.014
62. Zhao S, Chen L, Zang Y, Liu W, Liu S, Teng F, et al. Endometrial cancer in Lynch syndrome. Int J Cancer. (2022) 150:7–17. doi: 10.1002/ijc.v150.1
63. Bonadona V, Bonaïti B, Olschwang S, Grandjouan S, Huiart L, Longy M, et al. Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA. (2011) 305:2304–10. doi: 10.1001/jama.2011.743
64. Boland PM, Yurgelun MB, and Boland CR. Recent progress in Lynch syndrome and other familial colorectal cancer syndromes. CA Cancer J Clin. (2018) 68:217–31. doi: 10.3322/caac.21448
65. Lancaster JM, Powell CB, Chen LM, Richardson DL, and SGO Clinical Practice Committee. Society of Gynecologic Oncology statement on risk assessment for inherited gynecologic cancer predispositions. Gynecol Oncol. (2015) 136:3–7. doi: 10.1016/j.ygyno.2014.09.009
66. Ryan NAJ, McMahon R, Tobi S, Snowsill T, Esquibel S, Wallace AJ, et al. The proportion of endometrial tumours associated with Lynch syndrome (PETALS): A prospective cross-sectional study. PloS Med. (2020) 17:e1003263. doi: 10.1371/journal.pmed.1003263
67. Chao X, Li L, Wu M, Ma S, Tan X, Zhong S, et al. Comparison of screening strategies for Lynch syndrome in patients with newly diagnosed endometrial cancer: a prospective cohort study in China. Cancer Commun (Lond). (2019) 39:42. doi: 10.1186/s40880-019-0388-2
68. Kosary CL. FIGO stage, histology, histologic grade, age and race as prognostic factors in determining survival for cancers of the female gynecological system: an analysis of 1973–87 SEER cases of cancers of the endometrium, cervix, ovary, vulva, and vagina. Semin Surg Oncol. (1994) 10:31–46. doi: 10.1002/ssu.2980100107
69. Bokhman JV. Two pathogenetic types of endometrial carcinoma. Gynecol Oncol. (1983) 15:10–7. doi: 10.1016/0090-8258(83)90111-7
70. Eakin CM, Liao CI, Salani R, Cohen JG, Kapp DS, and Chan JK. The association of obesity with type I uterine cancer-is this an oversimplification? Am J Obstet Gynecol. (2022) 227:538–9. doi: 10.1016/j.ajog.2022.05.016
71. Waluyo ST, Tjokroprawiro BA, and Rahaju AS. Estrogen receptor and programmed death ligand-1 expression in type 1 endometrial cancer and its associated clinicopathological characteristics. Cancer Treat Res Commun. (2023) 37:100766. doi: 10.1016/j.ctarc.2023.100766
72. Parkash V, Aisagbonhi O, Riddle N, Siddon A, Panse G, and Fadare O. Recent advances in the classification of gynecological tract tumors: updates from the 5th edition of the world health organization “Blue book. Arch Pathol Lab Med. (2023) 147:1204–16. doi: 10.5858/arpa.2022-0166-RA
73. Lu Z and Chen J. Introduction of WHO classification of tumours of female reproductive organs, fourth edition. Zhonghua Bing Li Xue Za Zhi. (2014) 43:649–50.
74. McCluggage WG, Singh N, and Gilks CB. Key changes to the World Health Organization (WHO) classification of female genital tumours introduced in the 5th edition (2020). Histopathology. (2022) 80:762–78. doi: 10.1111/his.14609
75. Khoury JD, Solary E, Abla O, Akkari Y, Alaggio R, Apperley JF, et al. The 5th edition of the world health organization classification of haematolymphoid tumours: myeloid and histiocytic/dendritic neoplasms. Leukemia. (2022) 36:1703–19. doi: 10.1038/s41375-022-01613-1
76. Cree IA, White VA, Indave BI, and Lokuhetty D. Revising the WHO classification: female genital tract tumours. Histopathology. (2020) 76:151–6. doi: 10.1111/his.13977
77. Köbel M, Nelson GS, McAlpine J, Leon-Castillo A, and Bosse T. The rise of a novel classification system for endometrial carcinoma; integration of molecular subclasses. J Pathol. (2018) 244:538–49. doi: 10.1002/path.2018.244.issue-5
78. Lax SF. Molecular genetic pathways in various types of endometrial carcinoma: from a phenotypical to a molecular-based classification. Virchows Arch. (2004) 444:213–23. doi: 10.1007/s00428-003-0947-3
79. Berek JS, Matias-Guiu X, Creutzberg C, Fotopoulou C, Gaffney D, Kehoe S, et al. FIGO staging of endometrial cancer: 2023. Int J Gynaecol Obstet. (2023) 162:383–94. doi: 10.1002/ijgo.v162.2
80. Walsh DM, Hokenstad AN, Chen J, Sung J, Jenkins GD, Chia N, et al. Postmenopause as a key factor in the composition of the Endometrial Cancer Microbiome (ECbiome). Sci Rep. (2019) 9:19213. doi: 10.1038/s41598-019-55720-8
81. Baker JM, Chase DM, and Herbst-Kralovetz MM. Uterine microbiota: residents, tourists, or invaders? Front Immunol. (2018) 9:208. doi: 10.3389/fimmu.2018.00208
82. Sepich-Poore GD, Zitvogel L, Straussman R, Hasty J, Wargo JA, and Knight R. The microbiome and human cancer. Science. (2021) 371:eabc4552. doi: 10.1126/science.abc4552
83. Cai J, Sun L, and Gonzalez FJ. Gut microbiota-derived bile acids in intestinal immunity, inflammation, and tumorigenesis. Cell Host Microbe. (2022) 30:289–300. doi: 10.1016/j.chom.2022.02.004
84. Hieken TJ, Chen J, Chen B, Johnson S, Hoskin TL, Degnim AC, et al. The breast tissue microbiome, stroma, immune cells and breast cancer. Neoplasia. (2022) 27:100786. doi: 10.1016/j.neo.2022.100786
85. Kostic AD, Chun E, Meyerson M, and Garrett WS. Microbes and inflammation in colorectal cancer. Cancer Immunol Res. (2013) 1:150–7. doi: 10.1158/2326-6066.CIR-13-0101
86. Ilhan ZE, Łaniewski P, Thomas N, Roe DJ, Chase DM, and Herbst-Kralovetz MM. Deciphering the complex interplay between microbiota, HPV, inflammation and cancer through cervicovaginal metabolic profiling. EBioMedicine. (2019) 44:675–90. doi: 10.1016/j.ebiom.2019.04.028
87. Padda J, Khalid K, Cooper AC, and Jean-Charles G. Association between helicobacter pylori and gastric carcinoma. Cureus. (2021) 13:e15165. doi: 10.7759/cureus.15165
88. Balasubramaniam SD, Balakrishnan V, Oon CE, and Kaur G. Key molecular events in cervical cancer development. Medicina (Kaunas). (2019) 55:384. doi: 10.3390/medicina55070384
89. D’souza S, Lau KC, Coffin CS, and Patel TR. Molecular mechanisms of viral hepatitis induced hepatocellular carcinoma. World J Gastroenterol. (2020) 26:5759–83. doi: 10.3748/wjg.v26.i38.5759
90. Chase D, Goulder A, Zenhausern F, Monk B, and Herbst-Kralovetz M. The vaginal and gastrointestinal microbiomes in gynecologic cancers: a review of applications in etiology, symptoms and treatment. Gynecol Oncol. (2015) 138:190–200. doi: 10.1016/j.ygyno.2015.04.036
91. Møller BR, Kristiansen FV, Thorsen P, Frost L, and Mogensen SC. Sterility of the uterine cavity. Acta Obstet Gynecol Scand. (1995) 74:216–9. doi: 10.3109/00016349509008942
92. Balla B, Illés A, Tobiás B, Pikó H, Beke A, Sipos M, et al. The role of the vaginal and endometrial microbiomes in infertility and their impact on pregnancy outcomes in light of recent literature. Int J Mol Sci. (2024) 25:13227. doi: 10.3390/ijms252313227
93. Franasiak JM and Scott RT. Introduction: Microbiome in human reproduction. Fertil Steril. (2015) 104:1341–3. doi: 10.1016/j.fertnstert.2015.10.021
94. Lindheim L, Bashir M, Münzker J, Trummer C, Zachhuber V, Leber B, et al. Alterations in gut microbiome composition and barrier function are associated with reproductive and metabolic defects in women with polycystic ovary syndrome (PCOS): A pilot study. PloS One. (2017) 12:e0168390. doi: 10.1371/journal.pone.0168390
95. Jeon SJ, Cunha F, Vieira-Neto A, Bicalho RC, Lima S, Bicalho ML, et al. Blood as a route of transmission of uterine pathogens from the gut to the uterus in cows. Microbiome. (2017) 5:109. doi: 10.1186/s40168-017-0328-9
96. Ogah CO, Anikwe CC, Ajah LO, Ikeotuonye AC, Lawani OL, Okorochukwu BC, et al. Preoperative vaginal cleansing with chlorhexidine solution in preventing post-cesarean section infections in a low resource setting: A randomized controlled trial. Acta Obstet Gynecol Scand. (2021) 100:694–703. doi: 10.1111/aogs.v100.4
97. Dong M, Dong Y, Bai J, Li H, Ma X, Li B, et al. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol. (2023) 13:1124591. doi: 10.3389/fcimb.2023.1124591
98. Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SSK, McCulle SL, et al. Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci U S A. (2011) 108 Suppl 1:4680–7. doi: 10.1073/pnas.1002611107
99. France MT, Ma B, Gajer P, Brown S, Humphrys MS, Holm JB, et al. VALENCIA: a nearest centroid classification method for vaginal microbial communities based on composition. Microbiome. (2020) 8:166. doi: 10.1186/s40168-020-00934-6
100. Hakimjavadi H, George SH, Taub M, Dodds LV, Sanchez-Covarrubias AP, Huang M, et al. The vaginal microbiome is associated with endometrial cancer grade and histology. Cancer Res Commun. (2022) 2:447–55. doi: 10.1158/2767-9764.CRC-22-0075
101. Lam KC, Vyshenska D, Hu J, Rodrigues RR, Nilsen A, Zielke RA, et al. Transkingdom network reveals bacterial players associated with cervical cancer gene expression program. PeerJ. (2018) 6:e5590. doi: 10.7717/peerj.5590
102. Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science. (2017) 358:1443–8. doi: 10.1126/science.aal5240
103. Zhang T, Li Y, Zhai E, Zhao R, Qian Y, Huang Z, et al. Intratumoral fusobacterium nucleatum recruits tumor-associated neutrophils to promote gastric cancer progression and immune evasion. Cancer Res. (2025) 85:1819–41. doi: 10.1158/0008-5472.CAN-24-2580
104. Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, and Han YW. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe. (2013) 14:195–206. doi: 10.1016/j.chom.2013.07.012
105. Yang Y, Weng W, Peng J, Hong L, Yang L, Toiyama Y, et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating toll-like receptor 4 signaling to nuclear factor-κB, and up-regulating expression of microRNA-21. Gastroenterology. (2017) 152:851–66. doi: 10.1053/j.gastro.2016.11.018
106. Brennan CA and Garrett WS. Fusobacterium nucleatum - symbiont, opportunist and oncobacterium. Nat Rev Microbiol. (2019) 17:156–66. doi: 10.1038/s41579-018-0129-6
107. Abed J, Emgård JEM, Zamir G, Faroja M, Almogy G, Grenov A, et al. Fap2 mediates fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed gal-galNAc. Cell Host Microbe. (2016) 20:215–25. doi: 10.1016/j.chom.2016.07.006
108. Barczyński B, Frąszczak K, Grywalska E, Kotarski J, and Korona-Głowniak I. Vaginal and cervical microbiota composition in patients with endometrial cancer. Int J Mol Sci. (2023) 24:8266. doi: 10.3390/ijms24098266
109. Gressel GM, Usyk M, Frimer M, Kuo DYS, and Burk RD. Characterization of the endometrial, cervicovaginal and anorectal microbiota in post-menopausal women with endometrioid and serous endometrial cancers. PloS One. (2021) 16:e0259188. doi: 10.1371/journal.pone.0259188
110. Moreno I and Simon C. Deciphering the effect of reproductive tract microbiota on human reproduction. Reprod Med Biol. (2019) 18:40–50. doi: 10.1002/rmb2.12249
111. Fang RL, Chen LX, Shu WS, Yao SZ, Wang SW, and Chen YQ. Barcoded sequencing reveals diverse intrauterine microbiomes in patients suffering with endometrial polyps. Am J Transl Res. (2016) 8:1581–92.
112. Moreno I, Codoñer FM, Vilella F, Valbuena D, Martinez-Blanch JF, Jimenez-Almazán J, et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am J Obstet Gynecol. (2016) 215:684–703. doi: 10.1016/j.ajog.2016.09.075
113. Semertzidou A, Whelan E, Smith A, Ng S, Roberts L, Brosens JJ, et al. Microbial signatures and continuum in endometrial cancer and benign patients. Microbiome. (2024) 12:118. doi: 10.1186/s40168-024-01821-0
114. Davenport M, Poles J, Leung JM, Wolff MJ, Abidi WM, Ullman T, et al. Metabolic alterations to the mucosal microbiota in inflammatory bowel disease. Inflammation Bowel Dis. (2014) 20:723–31. doi: 10.1097/MIB.0000000000000011
115. Omar Al-Hassi H, Ng O, and Brookes M. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut. (2018) 67:395. doi: 10.1136/gutjnl-2017-314219
116. Cao Y, Shang F, Jin M, Deng S, Gu J, Mao F, et al. Changes in Bacteroides and the microbiota in patients with obstructed colorectal cancer: retrospective cohort study. BJS Open. (2023) 7:zrad105. doi: 10.1093/bjsopen/zrad105
117. Lu W, He F, Lin Z, Liu S, Tang L, Huang Y, et al. Dysbiosis of the endometrial microbiota and its association with inflammatory cytokines in endometrial cancer. Int J Cancer. (2021) 148:1708–16. doi: 10.1002/ijc.v148.7
118. Łaniewski P, Ilhan ZE, and Herbst-Kralovetz MM. The microbiome and gynaecological cancer development, prevention and therapy. Nat Rev Urol. (2020) 17:232–50. doi: 10.1038/s41585-020-0286-z
119. Zheng HT, Lou MWC, Dugué PA, and Lynch BM. Circulating inflammatory markers and risk of endometrial cancer: A systematic review and meta-analysis. Cancer Epidemiol. (2024) 93:102662. doi: 10.1016/j.canep.2024.102662
120. Wang L, Yang J, Su H, Shi L, Chen B, and Zhang S. Endometrial microbiota from endometrial cancer and paired pericancer tissues in postmenopausal women: differences and clinical relevance. Menopause. (2022) 29:1168–75. doi: 10.1097/GME.0000000000002053
121. Caselli E, Soffritti I, D’Accolti M, Piva I, Greco P, and Bonaccorsi G. Atopobium vaginae And Porphyromonas somerae Induce Proinflammatory Cytokines Expression In Endometrial Cells: A Possible Implication For Endometrial Cancer? Cancer Manag Res. (2019) 11:8571–5. doi: 10.2147/CMAR.S217362
122. Tango CN, Seo SS, Kwon M, Lee DO, Chang HK, and Kim MK. Taxonomic and functional differences in cervical microbiome associated with cervical cancer development. Sci Rep. (2020) 10:9720. doi: 10.1038/s41598-020-66607-4
123. Yazici C, Wolf PG, Kim H, Cross TWL, Vermillion K, Carroll T, et al. Race-dependent association of sulfidogenic bacteria with colorectal cancer. Gut. (2017) 66:1983–94. doi: 10.1136/gutjnl-2016-313321
124. Kapur N, Mir H, Clark Iii CE, Krishnamurti U, Beech DJ, Lillard JW, et al. CCR6 expression in colon cancer is associated with advanced disease and supports epithelial-to-mesenchymal transition. Br J Cancer. (2016) 114:1343–51. doi: 10.1038/bjc.2016.113
125. Chen W, Qin Y, and Liu S. CCL20 signaling in the tumor microenvironment. Adv Exp Med Biol. (2020) 1231:53–65. doi: 10.1007/978-3-030-36667-4_6
126. Liu Y, Wang J, Ni T, Wang L, Wang Y, and Sun X. CCL20 mediates RANK/RANKL-induced epithelial-mesenchymal transition in endometrial cancer cells. Oncotarget. (2016) 7:25328–39. doi: 10.18632/oncotarget.v7i18
127. Yang X, Liao D, Huang Y, Li C, Li Y, Deng Z, et al. CCL20 expression via AKT-ERK1/2-AP1 pathway in mycoplasma pneumoniae infection: implications for EMT and cell migration. J Inflammation Res. (2025) 18:5727–39. doi: 10.2147/JIR.S512408
128. Tosado-Rodríguez E, Mendez LB, Espino AM, Dorta-Estremera S, Aquino EE, Romaguera J, et al. Inflammatory cytokines and a diverse cervicovaginal microbiota associate with cervical dysplasia in a cohort of Hispanics living in Puerto Rico. PloS One. (2023) 18:e0284673. doi: 10.1371/journal.pone.0284673
129. Delgado-Diaz DJ, Tyssen D, Hayward JA, Gugasyan R, Hearps AC, and Tachedjian G. Distinct immune responses elicited from cervicovaginal epithelial cells by lactic acid and short chain fatty acids associated with optimal and non-optimal vaginal microbiota. Front Cell Infect Microbiol. (2019) 9:446. doi: 10.3389/fcimb.2019.00446
130. Amabebe E and Anumba DOC. Female gut and genital tract microbiota-induced crosstalk and differential effects of short-chain fatty acids on immune sequelae. Front Immunol. (2020) 11:2184. doi: 10.3389/fimmu.2020.02184
131. Choy EH, De Benedetti F, Takeuchi T, Hashizume M, John MR, and Kishimoto T. Translating IL-6 biology into effective treatments. Nat Rev Rheumatol. (2020) 16:335–45. doi: 10.1038/s41584-020-0419-z
132. Wang SY, Cai Y, Hu X, Li F, Qian XH, Xia LY, et al. P. gingivalis in oral-prostate axis exacerbates benign prostatic hyperplasia via IL-6/IL-6R pathway. Mil Med Res. (2024) 11:30. doi: 10.1186/s40779-024-00533-8
133. Tu Z, Wei W, Zeng F, Wang W, Zhang Y, Zhang Y, et al. IL-6 up-regulates expression of LIM-domain only protein 4 in psoriatic keratinocytes through activation of the MEK/ERK/NF-κB pathway. Am J Pathol. (2024) 194:708–20. doi: 10.1016/j.ajpath.2024.01.014
134. Choi MK, Kim J, Park HM, Lim CM, Pham TH, Shin HY, et al. The DPA-derivative 11S, 17S-dihydroxy 7,9,13,15,19 (Z,E,Z,E,Z)-docosapentaenoic acid inhibits IL-6 production by inhibiting ROS production and ERK/NF-κB pathway in keratinocytes HaCaT stimulated with a fine dust PM10. Ecotoxicol Environ Saf. (2022) 232:113252. doi: 10.1016/j.ecoenv.2022.113252
135. Kim J, Park HM, Lim CM, Jeon KB, Kim S, Lee J, et al. Specialized pro-resolving mediator 7S MaR1 inhibits IL-6 expression via modulating ROS/p38/ERK/NF-κB pathways in PM10-exposed keratinocytes. BMB Rep. (2024) 57:490–6. doi: 10.5483/BMBRep.2024-0124
136. So KA, Min KJ, Hong JH, and Lee JK. Interleukin-6 expression by interactions between gynecologic cancer cells and human mesenchymal stem cells promotes epithelial-mesenchymal transition. Int J Oncol. (2015) 47:1451–9. doi: 10.3892/ijo.2015.3122
137. Do TV, Kubba LA, Du H, Sturgis CD, and Woodruff TK. Transforming growth factor-beta1, transforming growth factor-beta2, and transforming growth factor-beta3 enhance ovarian cancer metastatic potential by inducing a Smad3-dependent epithelial-to-mesenchymal transition. Mol Cancer Res. (2008) 6:695–705. doi: 10.1158/1541-7786.MCR-07-0294
138. Ewington L, Taylor A, Sriraksa R, Horimoto Y, Lam EWF, and El-Bahrawy MA. The expression of interleukin-8 and interleukin-8 receptors in endometrial carcinoma. Cytokine. (2012) 59:417–22. doi: 10.1016/j.cyto.2012.04.036
139. Roškar L, Pušić M, Roškar I, Kokol M, Pirš B, Smrkolj Š, et al. Models including preoperative plasma levels of angiogenic factors, leptin and IL-8 as potential biomarkers of endometrial cancer. Front Oncol. (2022) 12:972131. doi: 10.3389/fonc.2022.972131
140. Khapuinamai A, Rudraprasad D, Pandey S, Gandhi J, Mishra DK, and Joseph J. Global transcriptomic profiling of innate and adaptive immunity during aspergillus flavus endophthalmitis in a murine model. Invest Ophthalmol Vis Sci. (2024) 65:44. doi: 10.1167/iovs.65.4.44
141. Meng W, Xue S, and Chen Y. The role of CXCL12 in tumor microenvironment. Gene. (2018) 641:105–10. doi: 10.1016/j.gene.2017.10.015
142. Kojiro-Sanada S, Yasuda K, Nishio S, Ogasawara S, Akiba J, Ushijima K, et al. CXCL14-CXCR4 and CXCL12-CXCR4 axes may play important roles in the unique invasion process of endometrioid carcinoma with MELF-pattern myoinvasion. Int J Gynecol Pathol. (2017) 36:530–9. doi: 10.1097/PGP.0000000000000362
143. King J, Mir H, and Singh S. Association of cytokines and chemokines in pathogenesis of breast cancer. Prog Mol Biol Transl Sci. (2017) 151:113–36. doi: 10.1016/bs.pmbts.2017.07.003
144. Huang X, Hao J, Tan YQ, Zhu T, Pandey V, and Lobie PE. CXC chemokine signaling in progression of epithelial ovarian cancer: theranostic perspectives. Int J Mol Sci. (2022) 23:2642. doi: 10.3390/ijms23052642
145. Cheng ZH, Shi YX, Yuan M, Xiong D, Zheng JH, and Zhang ZY. Chemokines and their receptors in lung cancer progression and metastasis. J Zhejiang Univ Sci B. (2016) 17:342–51. doi: 10.1631/jzus.B1500258
146. Goenka A, Khan F, Verma B, Sinha P, Dmello CC, Jogalekar MP, et al. Tumor microenvironment signaling and therapeutics in cancer progression. Cancer Commun (Lond). (2023) 43:525–61. doi: 10.1002/cac2.12416
147. Dobroch J, Bojczuk K, Kołakowski A, Baczewska M, and Knapp P. The exploration of chemokines importance in the pathogenesis and development of endometrial cancer. Molecules. (2022) 27:2041. doi: 10.3390/molecules27072041
148. Liang JJ, Zhu S, Bruggeman R, Zaino RJ, Evans DB, Fleming JB, et al. High levels of expression of human stromal cell-derived factor-1 are associated with worse prognosis in patients with stage II pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomarkers Prev. (2010) 19:2598–604. doi: 10.1158/1055-9965.EPI-10-0405
149. Ottaiano A, Scala S, Normanno N, Botti G, Tatangelo F, Di Mauro A, et al. Prognostic and predictive role of CXC chemokine receptor 4 in metastatic colorectal cancer patients. Appl Immunohistochem Mol Morphol. (2020) 28:755–60. doi: 10.1097/PAI.0000000000000828
150. Liu P, Long P, Huang Y, Sun F, and Wang Z. CXCL12/CXCR4 axis induces proliferation and invasion in human endometrial cancer. Am J Transl Res. (2016) 8:1719–29.
151. Miao Z, Luker KE, Summers BC, Berahovich R, Bhojani MS, Rehemtulla A, et al. CXCR7 (RDC1) promotes breast and lung tumor growth in vivo and is expressed on tumor-associated vasculature. Proc Natl Acad Sci U S A. (2007) 104:15735–40. doi: 10.1073/pnas.0610444104
152. Wang G, Pang Y, Li N, Hui Y, and Jin D. CXCR7 promoted proliferation, migration and invasion in HCC Cells by inactivating Hippo-YAP signaling. Discov Oncol. (2025) 16:561. doi: 10.1007/s12672-025-02324-6
153. Teng F, Tian WY, Wang YM, Zhang YF, Guo F, Zhao J, et al. Cancer-associated fibroblasts promote the progression of endometrial cancer via the SDF-1/CXCR4 axis. J Hematol Oncol. (2016) 9:8. doi: 10.1186/s13045-015-0231-4
154. Yoshimura T. The chemokine MCP-1 (CCL2) in the host interaction with cancer: a foe or ally? Cell Mol Immunol. (2018) 15:335–45. doi: 10.1038/cmi.2017.135
155. Modak RV, de Oliveira Rebola KG, McClatchy J, Mohammadhosseini M, Damnernsawad A, Kurtz SE, et al. Targeting CCL2/CCR2 signaling overcomes MEK inhibitor resistance in acute myeloid leukemia. Clin Cancer Res. (2024) 30:2245–59. doi: 10.1158/1078-0432.CCR-23-2654
156. Peña CG, Nakada Y, Saatcioglu HD, Aloisio GM, Cuevas I, Zhang S, et al. LKB1 loss promotes endometrial cancer progression via CCL2-dependent macrophage recruitment. J Clin Invest. (2015) 125:4063–76. doi: 10.1172/JCI82152
157. Liu W, Wang L, Zhang J, Cheng K, Zheng W, and Ma Z. CC chemokine 2 promotes ovarian cancer progression through the MEK/ERK/MAP3K19 signaling pathway. Int J Mol Sci. (2023) 24:10652. doi: 10.3390/ijms241310652
158. Huang T, Fan Q, Wang Y, Cui Y, Wang Z, Yang L, et al. Schwann cell-derived CCL2 promotes the perineural invasion of cervical cancer. Front Oncol. (2020) 10:19. doi: 10.3389/fonc.2020.00019
159. Li D, Ji H, Niu X, Yin L, Wang Y, Gu Y, et al. Tumor-associated macrophages secrete CC-chemokine ligand 2 and induce tamoxifen resistance by activating PI3K/Akt/mTOR in breast cancer. Cancer Sci. (2020) 111:47–58. doi: 10.1111/cas.v111.1
160. Cheng R, Xue X, and Liu X. Expression of IL17A in endometrial carcinoma and effects of IL17A on biological behaviour in Ishikawa cells. J Int Med Res. (2020) 48:300060520950563. doi: 10.1177/0300060520950563
161. Dawod B, Liu J, Gebremeskel S, Yan C, Sappong A, Johnston B, et al. Myeloid-derived suppressor cell depletion therapy targets IL-17A-expressing mammary carcinomas. Sci Rep. (2020) 10:13343. doi: 10.1038/s41598-020-70231-7
162. Lv Q, Wu K, Liu F, Wu W, Chen Y, and Zhang W. Interleukin−17A and heparanase promote angiogenesis and cell proliferation and invasion in cervical cancer. Int J Oncol. (2018) 53:1809–17. doi: 10.3892/ijo.2018.4503
163. Picard FSR, Lutz V, Brichkina A, Neuhaus F, Ruckenbrod T, Hupfer A, et al. IL-17A-producing CD8+ T cells promote PDAC via induction of inflammatory cancer-associated fibroblasts. Gut. (2023) 72:1510–22. doi: 10.1136/gutjnl-2022-327855
164. Uzan J, Laas E, Alsamad IA, Skalli D, Mansouri D, Haddad B, et al. Supervised clustering of adipokines and hormonal receptors predict prognosis in a population of obese women with type 1 endometrial cancer. Int J Mol Sci. (2017) 18:1055. doi: 10.3390/ijms18051055
165. Arend WP, Malyak M, Guthridge CJ, and Gabay C. Interleukin-1 receptor antagonist: role in biology. Annu Rev Immunol. (1998) 16:27–55. doi: 10.1146/annurev.immunol.16.1.27
166. Lewis AM, Varghese S, Xu H, and Alexander HR. Interleukin-1 and cancer progression: the emerging role of interleukin-1 receptor antagonist as a novel therapeutic agent in cancer treatment. J Transl Med. (2006) 4:48. doi: 10.1186/1479-5876-4-48
167. Elaraj DM, Weinreich DM, Varghese S, Puhlmann M, Hewitt SM, Carroll NM, et al. The role of interleukin 1 in growth and metastasis of human cancer xenografts. Clin Cancer Res. (2006) 12:1088–96. doi: 10.1158/1078-0432.CCR-05-1603
168. Bouras E, Karhunen V, Gill D, Huang J, Haycock PC, Gunter MJ, et al. Circulating inflammatory cytokines and risk of five cancers: a Mendelian randomization analysis. BMC Med. (2022) 20:3. doi: 10.1186/s12916-021-02193-0
169. Massouh N, Jaffa AA, and Jaffa MA. Diabetes and the social, biologic, and behavioral determinants of endometrial cancer in the United States. BMC Cancer. (2024) 24:540. doi: 10.1186/s12885-024-12192-y
170. Esposito G, Bravi F, Serraino D, Parazzini F, Crispo A, Augustin LSA, et al. Diabetes risk reduction diet and endometrial cancer risk. Nutrients. (2021) 13:2630. doi: 10.3390/nu13082630
171. Zabuliene L, Kaceniene A, Steponaviciene L, Linkeviciute-Ulinskiene D, Stukas R, Arlauskas R, et al. Risk of endometrial cancer in women with diabetes: A population-based retrospective cohort study. J Clin Med. (2021) 10:3453. doi: 10.3390/jcm10163453
172. Kim S, Park J, Chen Y, Rowe K, Snyder J, Fraser A, et al. Long-term diabetes risk among endometrial cancer survivors in a population-based cohort study. Gynecol Oncol. (2020) 156:185–93. doi: 10.1016/j.ygyno.2019.10.015
173. Wartko PD, Beck TL, Reed SD, Mueller BA, and Hawes SE. Association of endometrial hyperplasia and cancer with a history of gestational diabetes. Cancer Causes Control. (2017) 28:819–28. doi: 10.1007/s10552-017-0908-9
174. Luo J, Beresford S, Chen C, Chlebowski R, Garcia L, Kuller L, et al. Association between diabetes, diabetes treatment and risk of developing endometrial cancer. Br J Cancer. (2014) 111:1432–9. doi: 10.1038/bjc.2014.407
175. Foulkes WD, Polak P, and Karlić R. Helicobacter pylori, homologous-recombination genes, and gastric cancer. N Engl J Med. (2023) 389:379–80. doi: 10.1056/NEJMoa2211807
176. Wu S, Liu L, Xu H, Zhu Q, and Tan M. The involvement of MALAT1-ALKBH5 signaling axis into proliferation and metastasis of human papillomavirus-positive cervical cancer. Cancer Biol Ther. (2023) 24:2249174. doi: 10.1080/15384047.2023.2249174
177. Guo M, Yao Z, Jiang C, Songyang Z, Gan L, and Xiong Y. Three-dimensional and single-cell sequencing of liver cancer reveals comprehensive host-virus interactions in HBV infection. Front Immunol. (2023) 14:1161522. doi: 10.3389/fimmu.2023.1161522
178. Yuan L, Li S, Chen Q, Xia T, Luo D, Li L, et al. EBV infection-induced GPX4 promotes chemoresistance and tumor progression in nasopharyngeal carcinoma. Cell Death Differ. (2022) 29:1513–27. doi: 10.1038/s41418-022-00939-8
179. Liao C, Li M, Chen X, Tang C, Quan J, Bode AM, et al. Anoikis resistance and immune escape mediated by Epstein-Barr virus-encoded latent membrane protein 1-induced stabilization of PGC-1α promotes invasion and metastasis of nasopharyngeal carcinoma. J Exp Clin Cancer Res. (2023) 42:261. doi: 10.1186/s13046-023-02835-6
180. Li T, Li F, Guo X, Hong C, Yu X, Wu B, et al. Anti-epstein-barr virus BNLF2b for mass screening for nasopharyngeal cancer. N Engl J Med. (2023) 389:808–19. doi: 10.1056/NEJMoa2301496
181. Toyoda K, Yasunaga JI, Shichijo T, Arima Y, Tsujita K, Tanaka A, et al. HTLV-1 bZIP factor-induced reprogramming of lactate metabolism and epigenetic status promote leukemic cell expansion. Blood Cancer Discov. (2023) 4:374–93. doi: 10.1158/2643-3230.BCD-22-0139
182. Goncalves PH, Ziegelbauer J, Uldrick TS, and Yarchoan R. Kaposi sarcoma herpesvirus-associated cancers and related diseases. Curr Opin HIV AIDS. (2017) 12:47–56. doi: 10.1097/COH.0000000000000330
183. Grossmann C, Podgrabinska S, Skobe M, and Ganem D. Activation of NF-kappaB by the latent vFLIP gene of Kaposi’s sarcoma-associated herpesvirus is required for the spindle shape of virus-infected endothelial cells and contributes to their proinflammatory phenotype. J Virol. (2006) 80:7179–85. doi: 10.1128/JVI.01603-05
184. Keller SA, Schattner EJ, and Cesarman E. Inhibition of NF-kappaB induces apoptosis of KSHV-infected primary effusion lymphoma cells. Blood. (2000) 96:2537–42. doi: 10.1182/blood.V96.7.2537.h8002537_2537_2542
185. Hughes DJ, Wood JJ, Jackson BR, Baquero-Pérez B, and Whitehouse A. NEDDylation is essential for Kaposi’s sarcoma-associated herpesvirus latency and lytic reactivation and represents a novel anti-KSHV target. PloS Pathog. (2015) 11:e1004771. doi: 10.1371/journal.ppat.1004771
186. Aoki Y, Jaffe ES, Chang Y, Jones K, Teruya-Feldstein J, Moore PS, et al. Angiogenesis and hematopoiesis induced by Kaposi’s sarcoma-associated herpesvirus-encoded interleukin-6. Blood. (1999) 93:4034–43. doi: 10.1182/blood.V93.12.4034.412k38_4034_4043
187. Mohammed SA, Hetta HF, Zahran AM, Tolba MEM, Attia RAH, Behnsawy HM, et al. T cell subsets, regulatory T, regulatory B cells and proinflammatory cytokine profile in Schistosoma haematobium associated bladder cancer: First report from Upper Egypt. PloS Negl Trop Dis. (2023) 17:e0011258. doi: 10.1371/journal.pntd.0011258
Keywords: endometrial cancer, reproductive tract microbiome, immune response, inflammation, dysbiosis
Citation: Liang B, Tan J, Li J, Wang X, Li G, Li H, Li T and Gao H (2025) Epidemiology, molecular typing, microbiome-immune interactions and treatment strategies of endometrial cancer: a review. Front. Immunol. 16:1595638. doi: 10.3389/fimmu.2025.1595638
Received: 18 March 2025; Accepted: 05 June 2025;
Published: 25 June 2025.
Edited by:
Bastian Czogalla, LMU Munich University Hospital, GermanyReviewed by:
Eswari Dodagatta-Marri, University of California, San Francisco, United StatesZhijun Liu, First Affiliated Hospital of Xi’an Jiaotong University, China
Valerie Hughes, University of Cincinnati, United States
Copyright © 2025 Liang, Tan, Li, Wang, Li, Li, Li and Gao. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Hong Gao, Z2FvaDk4OEBxcS5jb20=
†These authors share first authorship