- 1Xinxiang Medical University, Xinxiang, China
- 2The Fourth Clinical College, Xinxiang Medical University, Xinxiang, China
- 3Henan Medical Key Laboratory for Immunology and Targeted Therapy, Xinxiang, China
- 4School of Basic Medicine, Xuzhou Medical University, Xuzhou, China
- 5School of Basic Medicine, Xinxiang Medical University, Xinxiang, China
Endometrial carcinoma (EC) represents one of the most prevalent malignancies within the female reproductive system. The frequency of its occurrence is on the rise annually, and patients diagnosed at advanced stages face a less favorable prognosis. Recent studies have highlighted the significant influence of the tumor immune microenvironment (TME) on the initiation, progression, metastasis, and therapeutic resistance of endometrial cancer. The TME encompasses various components such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM). These elements contribute to an immunosuppressive milieu by secreting cytokines, extracellular vesicles (EVs), and engaging immune checkpoint pathways like PD-1/PD-L1, thereby supporting tumor immune evasion and resistance to treatment. This review synthesizes current understanding of the EC-TME, focusing on the distinct roles and interactions of its key constituents within the context of EC biology. Furthermore, we explore the rationale and progress for novel therapeutic strategies targeting the TME, such as immune checkpoint inhibitors, combination therapies, and nano delivery systems leveraging EVs, aiming to provide insights for improving EC patient outcomes.
1 Introduction
Endometrial cancer (Endometrial Cancer, EC) is a common malignant tumor in gynecology. The global incidence and mortality rates of EC have been increasing persistently, and it has become the tumor with the highest incidence in the female reproductive system in some developed countries, especially with a high prevalence among postmenopausal individuals (1). Alarmingly, epidemiological shifts reveal a marked increase in incidence among younger women and specific ethnic groups, such as women under 50 in Puerto Rico, highlighting the heterogeneous nature of EC risk and distribution (2). The insidious onset of EC, often with non-specific or absent early symptoms, contributes to a high proportion of patients presenting with advanced-stage disease, which severely compromises therapeutic efficacy and prognosis (3).
The crucial challenges influencing the prognosis of EC are centered on the high recurrence rate (20 - 30%) and treatment resistance, which is particularly prominent in advanced cases (4). Recent research has disclosed that the dynamic regulation of the tumor microenvironment (Tumor Microenvironment, TME) plays a core role herein. The TME is composed of tumor cells, immune cells (such as TAMs, TILs), stromal components, and cytokines, etc., and drives tumor progression through multiple mechanisms: Immunosuppressive cells (M2 macrophages, Tregs) weaken the anti-tumor immune response; Remodeling of the extracellular matrix promotes invasion and metastasis; Factors such as TGF-β and TNF-α mediate immune escape and chemotherapy resistance (5, 6). Crucially, the unique composition and signaling within the EC-TME not only dictates disease aggressiveness but also unveils novel targets for therapeutic intervention, such as immune checkpoint blockade, offering hope for improving patient survival (4).
In recent years, as research on the Tumor Microenvironment (TME) has deepened continuously, targeted therapeutic strategies aimed at immune cells such as Tumor-Associated Macrophages (TAMs), Myeloid-Derived Suppressor Cells (MDSCs), and Cancer-Associated Fibroblasts (CAFs), along with stromal components, have gradually emerged as a research focus. For example, reshaping the immune microenvironment and enhancing the anti-tumor immune response are expected by reprogramming the polarization state of TAMs, inhibiting the functions of MDSCs, or regulating the activities of CAFs. Furthermore, extracellular vesicles (EVs), as crucial mediators of intercellular communication and by carrying bioactive molecules such as miRNAs and proteins, mediate immune suppression and chemotherapy resistance, thereby offering new therapeutic targets. Nevertheless, despite certain advancements in existing research, the complexity and heterogeneity of the TME still present substantial challenges for treatment.
This review comprehensively examines the current landscape of the EC-TME. We detail the critical cellular players – TAMs, MDSCs, and CAFs – and non-cellular components, particularly EVs, emphasizing their EC-specific functions and interplay. We then critically evaluate evolving therapeutic strategies designed to target these elements, including the integration of single-cell sequencing insights to understand TME heterogeneity. Finally, we discuss future research directions and clinical trial design considerations, aiming to provide a foundation for advancing precision immunotherapy in endometrial cancer.
2 The components and dynamic interactions of the tumor microenvironment in endometrial cancer
2.1 Immune cell clusters
In the tumor immune microenvironment (TME) of endometrial cancer, immune cell populations play a pivotal role. These cells modulate tumor growth, invasion, and immune evasion through the secretion of cytokines, chemokines, and metabolic products. The following sections detail the principal immune cells and their functions:
2.1.1 Tumor-associated macrophages
TAMs constitute one of the most abundant immune cell types within the TME and are primarily classified into M1 (anti-tumor) and M2 (pro-tumor) phenotypes (7, 8). Within the EC-TME, M2-polarized TAMs typically predominate. These cells secrete immunosuppressive cytokines (e.g., IL-10, TGF-β), directly inhibiting CD8+ T cell activity and cytotoxic function (9–11). Concurrently, M2 TAMs produce potent pro-angiogenic factors like VEGF (12), fostering the development of new blood vessels that supply nutrients and oxygen essential for tumor growth and metastasis (13). Clinically, a high density of M2 TAMs within EC tumors is strongly correlated with increased tumor grade, stage, lymphovascular space invasion, and reduced overall survival, establishing them as key contributors to EC aggressiveness and poor prognosis (14). Consequently, strategies aimed at modulating TAM polarization (e.g., promoting M1 or repolarizing M2 to M1) represent a promising therapeutic approach to reinvigorate anti-tumor immunity in EC (15).
2.1.2 Myeloid-derived suppressor cells
MDSCs (Myeloid-Derived Suppressor Cells) constitute a heterogeneous population of myeloid cells characterized by their immunosuppressive properties. This category includes mononuclear MDSCs (M-MDSCs) and polymorphonuclear MDSCs (PMN-MDSCs). MDSCs suppress T cell proliferation through the secretion of arginase and reactive oxygen species (ROS), thereby dampening the anti-tumor immune response (14, 16). The accumulation of MDSCs in the peripheral blood and tumor tissue of EC patients is significantly associated with advanced disease stage, metastatic spread, and inferior survival outcomes (17, 18). Studies specifically in EC have demonstrated that MDSC abundance disrupts the balance of tumor-infiltrating immune cells, creating an environment permissive for tumor progression and resistance to immunotherapy (19). Therefore, targeting MDSCs to alleviate immunosuppression is a rational strategy to enhance treatment efficacy in EC (20).
2.1.3 Cancer-associated fibroblasts
Cancer-associated fibroblasts (CAFs) play a pivotal role in the tumor microenvironment, originating from diverse cell types including epithelial cells, endothelial cells, and mesenchymal stem cells (MSCs). The transformation of these cells into CAFs is mediated by various mechanisms. Epithelial cells undergo epithelial-mesenchymal transition (EMT), which is regulated by multiple signaling pathways within the tumor microenvironment (21). Endothelial cells, particularly during tumor angiogenesis, contribute to CAF formation by secreting cytokines and growth factors that promote CAF activation and function (22). MSCs can also be induced to differentiate into CAFs through signals within the tumor microenvironment, resulting in CAFs with potent immunosuppressive properties that facilitate tumor immune evasion (23).
CAFs remodel the extracellular matrix (ECM) by secreting cytokines such as FGF and IL-6, as well as growth factors, thereby enhancing tumor invasiveness and metastatic potential (24, 25). In EC, CAFs secrete a plethora of growth factors (e.g., FGF, HGF) and cytokines (e.g., IL-6, TGF-β). These factors directly stimulate EC tumor cell proliferation and survival while also recruiting and activating immunosuppressive cells like TAMs and MDSCs, thereby reinforcing the immunosuppressive niche (26). Critically, CAFs in EC contribute significantly to chemoresistance. They secrete factors like glutathione (GSH) that protect tumor cells from oxidative stress induced by platinum-based drugs and can alter drug availability or metabolism (27). The activation state and specific markers of CAF subpopulations in EC are emerging as potential prognostic indicators and therapeutic targets (28) (Figure 1).

Figure 1. Schematic representation of key cellular interactions in the EC tumor microenvironment. Created in BioRender. yilin, w. (2025) https://BioRender.com/kgamfy8.
2.2 Extracellular vesicles and signaling networks in cellular communication
Extracellular vesicles (EVs) are small membrane-bound structures secreted by cells and are ubiquitously present in various body fluids, including blood, urine, and lymph. Based on their cellular origin and biological properties, EVs can be categorized into three primary types: exosomes, microvesicles, and apoptotic bodies (29). EVs play a crucial role in intercellular communication by transporting diverse bioactive molecules, such as miRNAs, proteins, and lipids, thereby modulating the function and behavior of recipient cells (30). Within the EC-TME, EVs play multifaceted and significant roles in driving tumor progression and therapy resistance (31, 32).
2.2.1 EV cargo in EC pathogenesis
EVs derived from EC tumor cells and stromal cells carry specific molecules that directly influence tumor behavior. For instance, EVs can transport oncogenic miRNAs (e.g., miR-223) that target tumor suppressor genes or pathways involved in apoptosis, enhancing tumor cell survival and proliferation (33). They also carry proteins like P-glycoprotein (P-gp), a drug efflux pump, contributing directly to multidrug resistance (MDR) by reducing intracellular chemotherapeutic drug concentrations (34).
2.2.2 EVs in immunosuppression
EVs are potent mediators of immune suppression within the EC-TME. They can deliver immunosuppressive ligands (e.g., PD-L1), cytokines (e.g., TGF-β, IL-10), and regulatory miRNAs directly to immune cells like T cells and NK cells, inhibiting their activation, proliferation, and cytotoxic functions (4). EV-mediated transfer of specific miRNAs can also reprogram immune cell phenotypes, for example, promoting the differentiation of monocytes into M2 macrophages (35, 36).
2.2.3 EVs in angiogenesis and metastasis
EVs facilitate EC progression by promoting angiogenesis and invasion. They transport pro-angiogenic factors (e.g., VEGF, FGF) that stimulate endothelial cell proliferation and new vessel formation. Furthermore, EVs carry matrix-remodeling enzymes (e.g., MMPs, uPA) that degrade the basement membrane and ECM, clearing a path for tumor cell migration and invasion into surrounding tissues and vasculature (37). EVs also prepare distant metastatic niches by influencing the phenotype of cells at potential secondary sites.
The critical role of EVs in mediating communication between EC tumor cells and the various TME components positions them as attractive targets for novel diagnostics and therapeutics aimed at disrupting these pro-tumorigenic networks.
3 The dual roles of tumor-associated macrophages in endometrial cancer
TAMs are not merely passive bystanders but active participants in EC pathogenesis, exhibiting complex and context-dependent functions primarily dictated by their polarization state.
3.1 Phenotypic polarization and functionality
TAMs in EC display significant heterogeneity, existing along a spectrum between classically activated M1 (anti-tumor) and alternatively activated M2 (pro-tumor) phenotypes (3). M1 TAMs, typically induced by microbial products or Th1 cytokines (e.g., IFN-γ), secrete pro-inflammatory cytokines (TNF-α, IL-12, IL-1β), generate reactive oxygen and nitrogen species (ROS/RNS), and promote antigen presentation, thereby stimulating anti-tumor immune responses. In contrast, M2 TAMs, polarized by Th2 cytokines (e.g., IL-4, IL-13, IL-10), secrete immunosuppressive factors (IL-10, TGF-β, PGE2), express scavenger receptors, and produce pro-angiogenic factors (VEGF, FGF, PDGF), actively supporting tumor growth, angiogenesis, tissue remodeling, and immune evasion (38) Metabolic reprogramming underpins the functional divergence between M1 and M2 TAMs. M1 TAMs utilize glycolysis and the pentose phosphate pathway, converting arginine to nitric oxide (NO) via inducible nitric oxide synthase (iNOS), which has cytotoxic effects. M2 TAMs, however, rely on oxidative phosphorylation and fatty acid oxidation, metabolizing arginine to ornithine and polyamines via arginase-1 (ARG1), promoting cell proliferation and tissue repair (39). In the endometrial cancer microenvironment, lactic acid and hypoxic conditions are critical factors that induce the polarization of TAMs toward the M2 phenotype. Lactic acid inhibits mitochondrial function in TAMs, promoting their polarization to the M2 type and establishing a positive feedback loop of immunosuppression (40). This polarization process not only enhances the immunosuppressive capabilities of TAMs but also further weakens the anti-tumor immune response, ultimately facilitating tumor progression (41).
3.2 The mechanism of treatment resistance
The predominance of M2 TAMs within the EC-TME directly contributes to resistance against both chemotherapy and immunotherapy through several interconnected mechanisms (42):
3.2.1 Immune checkpoint modulation
M2 TAMs frequently express high levels of immune checkpoint ligands, particularly PD-L1. Interaction of PD-L1 on TAMs with PD-1 on T cells delivers inhibitory signals that dampen T cell activation, proliferation, cytokine production, and cytotoxic capacity, effectively blunting anti-tumor immune responses (43, 44). The polarization state influences this; M2 TAMs generally exhibit higher PD-L1 expression and stronger immunosuppressive capacity via this pathway compared to M1 TAMs (45). TAMs located in hypoxic tumor regions or near blood vessels are often hotspots for PD-L1 expression and T cell suppression.
3.2.2 Activation of pro-survival pathways
TAMs secrete a multitude of factors (e.g., EGF, FGF, IL-6) that activate key pro-survival and proliferative signaling pathways in EC tumor cells, such as PI3K/Akt/mTOR and JAK/STAT pathways (46). Activation of PI3K/Akt signaling promotes tumor cell survival, proliferation, and metabolism, while also conferring resistance to apoptosis induced by chemotherapeutic agents. TAM-derived factors can also activate the NF-κB pathway in tumor cells, promoting inflammation, cell survival, and inhibition of apoptosis (47, 48). Collectively, these signals enhance tumor cell resilience to therapy.
3.2.3 Sustaining an immunosuppressive niche
Beyond direct effects on tumor cells and T cells, M2 TAMs contribute to a broader immunosuppressive environment by secreting cytokines (IL-10, TGF-β) that inhibit dendritic cell maturation, promote Treg differentiation and function, and recruit additional immunosuppressive cells like MDSCs. They also produce enzymes (e.g., IDO) that deplete tryptophan, further suppressing T cell function.
Targeting TAMs, particularly strategies to deplete M2 TAMs, block their recruitment, or reprogram them towards an M1 phenotype, is therefore a critical component of overcoming therapy resistance in EC.
4 The synergistic cooperation between MDSCs and CAFs
4.1 The immunosuppressive functionality of myeloid-derived suppressor cells
MDSCs are potent suppressors of anti-tumor immunity in EC, utilizing diverse mechanisms to inhibit effector immune cell function and promote a tolerant microenvironment (49):
4.1.1 Cytokine-mediated tumor promotion and immune suppression
MDSCs secrete significant amounts of cytokines like IL-6 and TGF-β within the EC-TME. IL-6 acts as a potent growth factor for EC tumor cells, activating the JAK/STAT3 pathway, which promotes proliferation, survival, and invasion (50, 51). Furthermore, IL-6 contributes to immunosuppression by promoting the polarization of monocytes towards M2 TAMs and inhibiting dendritic cell function (52).
TGF-β is a master regulator with pleiotropic effects. It directly suppresses the activation and function of T cells, NK cells, and macrophages. Crucially, TGF-β is a potent inducer of epithelial-mesenchymal transition (EMT) in EC tumor cells, enhancing their invasive and metastatic potential. It also stimulates CAF activation and ECM production (53).
4.1.2 Inhibition of lymphocyte function via soluble mediators
MDSCs employ enzymatic pathways to directly inhibit T and NK cell function. High expression of ARG1 depletes extracellular L-arginine, an essential amino acid for T cell function. L-arginine starvation leads to downregulation of the T cell receptor CD3ζ chain, impairing T cell receptor signaling and proliferation (54, 55). MDSCs also produce inducible nitric oxide synthase (iNOS), generating high levels of nitric oxide (NO). At elevated concentrations, NO induces T cell apoptosis, inhibits T cell proliferation and cytokine secretion, and disrupts NK cell cytotoxicity (54, 55). Elevated levels of ARG1 and iNOS activity in the EC-TME correlate strongly with disease progression and poor prognosis, underscoring their functional importance (56). MDSCs can also produce ROS, which further damages T cells and contributes to T cell anergy (57).
Through these multifaceted mechanisms, MDSCs establish a robust immunosuppressive network in EC, enabling tumor evasion and progression (58).
4.2 Therapy resistance mediated by CAFs
CAFs contribute significantly to EC progression and treatment failure through several key mechanisms (23, 59).
4.2.1 Metabolic protection and chemoresistance
CAFs protect EC tumor cells from chemotherapy-induced cytotoxicity. They achieve this partly by secreting high levels of glutathione (GSH), a major cellular antioxidant. GSH neutralizes reactive oxygen species (ROS) generated by chemotherapeutic agents like platinum drugs, reducing oxidative stress and apoptosis in tumor cells (60). Furthermore, GSH can directly conjugate with platinum drugs (e.g., cisplatin), forming less reactive and more easily excreted complexes, effectively reducing the intracellular concentration of active drug (61). CAFs may also modulate the availability of other metabolites, like cysteine, which can influence drug uptake and detoxification pathways in tumor cells.
4.2.2 Targetable pro-tumorigenic signaling
CAFs express high levels of fibroblast activation protein (FAP) and secrete substantial amounts of IL-6. FAP, a cell surface serine protease, is involved in ECM remodeling and promotes tumor growth and immune evasion. IL-6, as mentioned previously, stimulates tumor cell proliferation and survival via JAK/STAT3 signaling and contributes to immunosuppression. Preclinical studies in EC models suggest that targeting FAP (e.g., with inhibitory antibodies or small molecules) or blocking IL-6 signaling can inhibit CAF activity, reduce tumor growth, and sensitize tumors to chemotherapy (62, 63).
4.2.3 ECM remodeling and physical barrier
CAFs are the primary architects of the desmoplastic reaction in EC, characterized by excessive deposition and cross-linking of collagen and other ECM proteins. This leads to increased stromal stiffness (64). Increased matrix stiffness physically impeders drug diffusion into the tumor parenchyma, reducing drug delivery to cancer cells. Moreover, stiffness activates mechanosensitive signaling pathways (e.g., YAP/TAZ, FAK/Src) in both tumor cells and CAFs themselves, promoting tumor cell survival, proliferation, stemness, EMT, and resistance to therapy (65). CAF-derived EVs can also carry enzymes like LOX that cross-link collagen, further increasing stiffness and promoting metastatic behavior (66).
4.2.4 Paracrine signaling and survival cues
Beyond metabolites and ECM, CAFs secrete a wide array of growth factors (HGF, FGF, IGF), cytokines (IL-6, IL-8, CXCL12), and exosomes that deliver pro-survival signals directly to EC tumor cells. These signals activate PI3K/Akt, MAPK, and JAK/STAT pathways, counteracting the pro-apoptotic effects of chemotherapy and targeted therapies (67).
The profound impact of CAFs on chemoresistance and tumor progression highlights them as essential targets for improving EC treatment outcomes (68).
5 Therapeutic strategies aimed at the tumor microenvironment
The complexity of the EC-TME necessitates multi-faceted therapeutic approaches. Strategies targeting key immunosuppressive cells and pathways are under active development.
5.1 Immune checkpoint inhibitors
Immune checkpoint inhibitors (ICIs) have emerged as a critical therapeutic modality for various malignancies, demonstrating particularly significant efficacy in endometrial cancer (EC). PD-1/PD-L1 inhibitors restore T-cell anti-tumor activity by blocking the interaction between PD-1 and its ligand PD-L1 (69). The efficacy of PD-1/PD-L1 inhibitors in EC is strongly linked to the molecular subtype. Patients with mismatch repair-deficient (dMMR) or microsatellite instability-high (MSI-H) tumors, which typically have a higher tumor mutational burden (TMB) and greater immune cell infiltration, exhibit significantly higher objective response rates (ORR: 40-60%) compared to those with mismatch repair-proficient (pMMR) or microsatellite-stable (MSS) tumors, where response rates are generally low (70). This highlights the critical need for biomarker-driven patient selection.
Combination strategies are essential to overcome resistance in MSS tumors and enhance efficacy overall. Combining ICIs with agents targeting other TME components is a major focus. For example, combining ICIs with therapies that deplete or inhibit MDSCs (e.g., PDE5 inhibitors, ARG1/iNOS inhibitors, CXCR2 antagonists) has shown promise in preclinical EC models and early clinical trials, demonstrating enhanced T cell activation and improved anti-tumor responses (71, 72). This approach aims to alleviate the immunosuppressive brake applied by MDSCs, allowing unleashed T cells to effectively attack the tumor (73).
5.2 Rational combination therapies
Given the interconnected and redundant immunosuppressive pathways within the EC-TME, rationally designed combination therapies targeting multiple nodes simultaneously offer the greatest potential for success.
5.2.1 ICI + anti-angiogenic agents
Combining ICIs with anti-angiogenic drugs (e.g., Bevacizumab - anti-VEGF antibody, Tyrosine Kinase Inhibitors - TKIs) addresses two key aspects. Anti-angiogenics normalize the aberrant tumor vasculature, reducing hypoxia, improving drug delivery, and facilitating enhanced T cell infiltration into the tumor. VEGF itself has immunosuppressive effects, inhibiting dendritic cell maturation and promoting Treg and MDSC accumulation. Blocking VEGF can thus reverse this immunosuppression and synergize with ICIs. Clinical trials in EC (e.g., NCT03526432) are evaluating such combinations (70).
5.2.2 Targeting key immunosuppressive cells
TAMs: Strategies include CSF-1R inhibitors to block M2 TAM recruitment/survival and TLR agonists (e.g., TLR4, TLR9) to repolarize TAMs towards an M1 state (74, 75).
MDSCs: Pharmacological inhibitors targeting MDSC development (e.g., all-trans retinoic acid - ATRA), function (e.g., ARG1/iNOS inhibitors), or recruitment (e.g., CXCR1/2 inhibitors) are being explored. Combining these with ICIs aims to relieve MDSC-mediated suppression of T cells (72, 76).
CAFs: Approaches include neutralizing key CAF-derived factors (e.g., anti-IL-6, anti-TGF-β antibodies), inhibiting FAP enzymatic activity, or targeting specific CAF subpopulations identified via single-cell analysis. The goal is to disrupt CAF-tumor cell crosstalk, reduce ECM stiffness, and reverse chemoresistance (25).
5.2.3 Disrupting EV-mediated communication
Strategies focus on inhibiting EV biogenesis, release, or uptake (e.g., using neutral sphingomyelinase inhibitors, heparin derivatives, or specific receptor blockers) to interrupt the transfer of pro-tumorigenic and immunosuppressive cargoes between cells in the EC-TME (77, 78).
These multi-targeted combination approaches represent a paradigm shift, aiming to systematically remodel the immunosuppressive EC-TME into a state conducive to effective anti-tumor immunity.
5.3 Extracellular vesicle based nanodelivery systems
Exploiting the inherent properties of EVs offers a novel and promising therapeutic avenue (79). EVs possess natural biocompatibility, low immunogenicity, and the intrinsic ability to cross biological barriers, making them attractive candidates as drug delivery vehicles. They can be engineered to encapsulate various therapeutic agents, including small molecule chemotherapeutics, siRNAs, miRNAs, and proteins (80). For EC therapy, engineered EVs can be designed to specifically target TME components. For instance, EVs loaded with siRNA targeting key immunosuppressive genes (e.g., Arg1, Pd-l1, Il10) in TAMs or MDSCs, and decorated with surface ligands that bind receptors enriched on these cells, can deliver their cargo directly to the intended targets, silencing gene expression and reversing immunosuppression (81). Similarly, EVs loaded with chemotherapeutic agents can be targeted to EC tumor cells or CAFs, potentially overcoming drug resistance mechanisms associated with poor tumor penetration or efflux pumps (82). This EV-based nanodelivery strategy holds the potential to significantly enhance therapeutic efficacy while minimizing off-target systemic side effects, representing a cutting-edge approach for EC treatment.
6 Conclusion and future perspectives
The tumor immune microenvironment of endometrial carcinoma is a dynamic and highly heterogeneous ecosystem, playing a central and complex role in disease progression, metastasis, and response to therapy. Key cellular components, including immunosuppressive TAMs, MDSCs, and CAFs, alongside non-cellular factors like EVs and the remodeled ECM, engage in intricate cross-talk to establish a profoundly immunosuppressive milieu. This environment facilitates immune evasion and confers resistance to chemotherapy, radiotherapy, and even immunotherapy. While ICIs have shown remarkable success in the dMMR/MSI-H subset of EC patients, the majority with pMMR/MSS tumors derive limited benefit, underscoring the urgent need for novel therapeutic strategies.
6.1 Future research directions and clinical translation
Decoding Heterogeneity with Single-Cell Technologies: The application of single-cell RNA sequencing, spatial transcriptomics, and proteomics to EC patient samples is paramount. These technologies will resolve the true diversity of cell states within the TME (distinct TAM, MDSC, CAF subpopulations, exhausted T cell subsets), elucidate their spatial organization, define specific EC-TME signatures, and identify novel cell-type-specific therapeutic vulnerabilities and biomarkers predictive of treatment response.
6.2 Advanced biomarker integration
Moving beyond PD-L1 IHC and MSI status, future clinical trials must systematically incorporate and validate multiplexed biomarker panels. This includes assessing tumor mutational burden (TMB), specific immune gene expression signatures (e.g., interferon-gamma), the composition and functional state of the TME (e.g., ratios of CD8+ T cells to Tregs/MDSCs, M1/M2 TAM balance), and circulating biomarkers (e.g., EV cargo, cytokine levels). Such integration is essential for refining patient stratification and enabling truly personalized therapy.
6.3 Rational multi-target combination therapies
Overcoming the redundancy and complexity of the EC-TME will require intelligent combination strategies. Future efforts should focus on rationally combining ICIs with agents targeting the dominant immunosuppressive mechanisms in specific EC molecular subtypes or TME contexts (e.g., ICI + TAM modulator + MDSC inhibitor; ICI + CAF-targeting agent + anti-angiogenic). Preclinical models, including patient-derived organoids and orthotopic models incorporating human immune cells, are crucial for prioritizing the most promising combinations.
6.4 Leveraging EVs for therapy and diagnosis
Research into EV biology in EC must expand. This includes fully characterizing the EV cargo (miRNA, protein, lipid) specific to different cell types within the EC-TME and different disease states, understanding their precise roles in mediating communication, and developing robust methods for EV isolation and engineering. EV-based therapeutics (as delivery vehicles) and diagnostics (liquid biopsies) hold immense potential for improving EC management.
6.5 Innovative clinical trial design
Future clinical trials need to be biomarker-driven from the outset. Adaptive trial designs (e.g., basket, umbrella trials) that allow for patient selection based on molecular/TME profiling and enable evaluation of multiple targeted combinations within a single framework are essential. Incorporating deep correlative science, including serial biopsies and multi-omics analyses, will provide critical insights into mechanisms of response and resistance.
In conclusion, advancing the treatment of endometrial cancer demands a holistic understanding of the intricate and patient-specific interactions within the TME. By leveraging cutting-edge technologies to dissect TME complexity, developing sophisticated multi-targeted therapeutic strategies, and implementing biomarker-guided clinical trials, we can move towards more effective and personalized therapies, ultimately improving survival and quality of life for EC patients. Collaborative efforts across basic science, translational research, and clinical oncology are key to unlocking the potential of TME-targeted therapies in endometrial cancer.
Author contributions
YW: Writing – original draft, Writing – review & editing, Data curation, Formal Analysis, Investigation. NL: Writing – original draft, Writing – review & editing. XG: Investigation, Writing – review & editing. RH: Conceptualization, Writing – review & editing. JB: Supervision, Writing – review & editing. JZ: Methodology, Writing – review & editing. QW: Methodology, Supervision, 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 the Natural Science Foundation of Henan Province (Project No.: 252300421399) and the Jointly Built Project of Henan Medical Science and Technology Research Plan (Project No.: LHGJ20230896).
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 Generative AI was used in the creation of this manuscript. Disclosure During the preparation of this work the author(s) used NetEase Cloud AI in order to improve readability. After using this tool/service, the author(s) reviewed and edited the content as needed. The author(s) take(s) full responsibility for the content of the publication.
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. Zhang C, Wang M, and Wu Y. Features of the immunosuppressive tumor microenvironment in endometrial cancer based on molecular subtype. Front Oncol. (2023) 13:1278863. doi: 10.3389/fonc.2023.1278863
2. Rosario-Santos A, Torres-Cintrón CR, López-Rexach AG, Gonzalez-Carcache P, Tortolero-Luna G, and Umpierre S. A comparative analysis of endometrial cancer disparities in incidence, mortality, and survival between women living in Puerto Rico, Non-Hispanic Blacks, Non-Hispanic Whites, and US Hispanics between 2000-2018. Gynecol Oncol Rep. (2023) 49:101275. doi: 10.1016/j.gore.2023.101275
3. Garg P, Ramisetty SK, Raghu Subbalakshmi A, Krishna BM, Pareek S, Mohanty A, et al. Gynecological cancer tumor Microenvironment: Unveiling cellular complexity and therapeutic potential. Biochem Pharmacol. (2024) 229:116498. doi: 10.1016/j.bcp.2024.116498
4. Lee EK and Liu JF. Uterine serous carcinoma and uterine carcinosarcoma: molecular features, clinical advances, and emerging therapies. Clin Adv Hematol Oncol. (2024) 22:301–10. doi: 10.25270/ahc.2024.04.001
5. Chlebowski RT, Aragaki AK, Pan K, Haque R, Rohan TE, Song M, et al. Menopausal hormone therapy and ovarian and endometrial cancers: long-term follow-up of the women’s health initiative randomized trials. J Clin Oncol. (2024) 42:3537–49. doi: 10.1200/JCO.23.01918
6. Gupta YH, Khanom A, and Acton SE. Control of dendritic cell function within the tumour microenvironment. Front Immunol. (2022) 13:733800. doi: 10.3389/fimmu.2022.733800
7. Le Bras A. Tracking tumor-associated macrophages. Lab Anim (NY). (2025) 54:38. doi: 10.1038/s41684-025-01511-w
8. Li M, He L, Zhu J, Zhang P, and Liang S. Targeting tumor-associated macrophages for cancer treatment. Cell Biosci. (2022) 12:85. doi: 10.1186/s13578-022-00823-5
9. Malekghasemi S, Majidi J, Baghbanzadeh A, Abdolalizadeh J, Baradaran B, and Aghebati-Maleki L. Tumor-associated macrophages: protumoral macrophages in inflammatory tumor microenvironment. Adv Pharm Bull. (2020) 10:556–65. doi: 10.34172/apb.2020.066
10. Guo F, Gao Y, Zhou P, Wang H, Ma Z, Wang X, et al. Single-cell analysis reveals that TCF7L2 facilitates the progression of ccRCC via tumor-associated macrophages. Cell Signal. (2024) 124:111453. doi: 10.1016/j.cellsig.2024.111453
11. Shobaki N, Sato Y, Suzuki Y, Okabe N, and Harashima H. Manipulating the function of tumor-associated macrophages by siRNA-loaded lipid nanoparticles for cancer immunotherapy. J Control Release. (2020) 325:235–48. doi: 10.1016/j.jconrel.2020.07.001
12. Okikawa S, Morine Y, Saito Y, Yamada S, Tokuda K, Teraoku H, et al. Inhibition of the VEGF signaling pathway attenuates tumor−associated macrophage activity in liver cancer. Oncol Rep. (2022) 47:71. doi: 10.3892/or.2022.8282
13. Pan Y, Yu Y, Wang X, and Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol. (2020) 11:583084. doi: 10.3389/fimmu.2020.583084
14. Bhardwaj V and Ansell SM. Modulation of T-cell function by myeloid-derived suppressor cells in hematological Malignancies. Front Cell Dev Biol. (2023) 11:1129343. doi: 10.3389/fcell.2023.1129343
15. Li S, Sheng J, Zhang D, and Qin H. Targeting tumor-associated macrophages to reverse antitumor drug resistance. Aging (Albany NY). (2024) 16:10165–96. doi: 10.18632/aging.205858
16. Yokota S, Yonezawa T, Momoi Y, and Maeda S. Myeloid derived suppressor cells in peripheral blood can be a prognostic factor in canine transitional cell carcinoma. Vet Immunol Immunopathol. (2024) 269:110716. doi: 10.1016/j.vetimm.2024.110716
17. Zhang W, Li X, Jiang M, Ji C, Chen G, Zhang Q, et al. SOCS3 deficiency-dependent autophagy repression promotes the survival of early-stage myeloid-derived suppressor cells in breast cancer by activating the Wnt/mTOR pathway. J Leukoc Biol. (2023) 113:445–60. doi: 10.1093/jleuko/qiad020
18. Asaka S, Verma N, Yen TT, Hicks JL, Nonogaki H, Shen YA, et al. Association of glutaminase expression with immune-suppressive tumor microenvironment, clinicopathologic features, and clinical outcomes in endometrial cancer. Int J Gynecol Cancer. (2024) 34:1737–44. doi: 10.1136/ijgc-2024-005920
19. Mise Y, Hamanishi J, Daikoku T, Takamatsu S, Miyamoto T, Taki M, et al. Immunosuppressive tumor microenvironment in uterine serous carcinoma via CCL7 signal with myeloid-derived suppressor cells. Carcinogenesis. (2022) 43:647–58. doi: 10.1093/carcin/bgac032
20. Zhang K, Zakeri A, Alban T, Dong J, Ta HM, Zalavadia AH, et al. VISTA promotes the metabolism and differentiation of myeloid-derived suppressor cells by STAT3 and polyamine-dependent mechanisms. Cell Rep. (2024) 43:113661. doi: 10.1016/j.celrep.2023.113661
21. Helms EJ, Berry MW, Chaw RC, DuFort CC, Sun D, Onate MK, et al. Mesenchymal lineage heterogeneity underlies nonredundant functions of pancreatic cancer-associated fibroblasts. Cancer Discov. (2022) 12:484–501. doi: 10.1158/2159-8290.CD-21-0601
22. Li M, Wu B, Li L, Lv C, and Tian Y. Reprogramming of cancer-associated fibroblasts combined with immune checkpoint inhibitors: A potential therapeutic strategy for cancers. Biochim Biophys Acta Rev Cancer. (2023) 1878:188945. doi: 10.1016/j.bbcan.2023.188945
23. Aden D, Zaheer S, Ahluwalia H, and Ranga S. Cancer-associated fibroblasts: Is it a key to an intricate lock of tumorigenesis? Cell Biol Int. (2023) 47:859–93.
24. Zhao Z, Li T, Yuan Y, and Zhu Y. What is new in cancer-associated fibroblast biomarkers? Cell Commun Signal. (2023) 21:96.
25. Tokhanbigli S, Haghi M, Dua K, and Oliver BGG. Cancer-associated fibroblast cell surface markers as potential biomarkers or therapeutic targets in lung cancer. Cancer Drug Resist. (2024) 7:32. doi: 10.20517/cdr.2024.55
26. Hu C, Zhang Y, Wu C, and Huang Q. Heterogeneity of cancer-associated fibroblasts in head and neck squamous cell carcinoma: opportunities and challenges. Cell Death Discov. (2023) 9:124. doi: 10.1038/s41420-023-01428-8
27. Zhao Y, Shen M, Wu L, Yang H, Yao Y, Yang Q, et al. Stromal cells in the tumor microenvironment: accomplices of tumor progression? Cell Death Dis. (2023) 14:587.
28. Sulaiman R, De P, Aske JC, Lin X, Dale A, Gaster K, et al. A CAF-based two-cell hybrid co-culture model to test drug resistance in endometrial cancers. Biomedicines. (2023) 11:1326. doi: 10.3390/biomedicines11051326
29. Du S, Guan Y, Xie A, Yan Z, Gao S, Li W, et al. Extracellular vesicles: a rising star for therapeutics and drug delivery. J Nanobiotechnology. (2023) 21:231. doi: 10.1186/s12951-023-01973-5
30. Marostica G, Gelibter S, Gironi M, Nigro A, and Furlan R. Extracellular vesicles in neuroinflammation. Front Cell Dev Biol. (2020) 8:623039. doi: 10.3389/fcell.2020.623039
31. Swatler J, Dudka W, and Piwocka K. Isolation and characterization of extracellular vesicles from cell culture conditioned medium for immunological studies. Curr Protoc Immunol. (2020) 129:e96. doi: 10.1002/cpim.v129.1
32. Upadhya D and Shetty AK. MISEV2023 provides an updated and key reference for researchers studying the basic biology and applications of extracellular vesicles. Stem Cells Transl Med. (2024) 13:848–50. doi: 10.1093/stcltm/szae052
33. Abu-Rustum N, Yashar C, Arend R, Barber E, Bradley K, Brooks R, et al. Uterine neoplasms, version 1.2023, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. (2023) 21:181–209. doi: 10.6004/jnccn.2023.0006
34. Wang Y, Zheng YT, Zhang L, Cao XQ, Lin Z, Liu HY, et al. Undifferentiated endometrial carcinoma diagnosed during perimenopausal hormone therapy: a case report and literature review. Front Oncol. (2024) 14:1440246. doi: 10.3389/fonc.2024.1440246
35. Zheng L, Chang R, Liang B, Wang Y, Zhu Y, Jia Z, et al. Overcoming drug resistance through extracellular vesicle-based drug delivery system in cancer treatment. Cancer Drug Resist. (2024) 7:50. doi: 10.20517/cdr.2024.107
36. Biondi A, Vacante M, Catania R, and Sangiorgio G. Extracellular vesicles and immune system function: exploring novel approaches to colorectal cancer immunotherapy. Biomedicines. (2024) 12:1473. doi: 10.3390/biomedicines12071473
37. Yao H, Huang C, Zou J, Liang W, Zhao Y, Yang K, et al. Extracellular vesicle-packaged lncRNA from cancer-associated fibroblasts promotes immune evasion by downregulating HLA-A in pancreatic cancer. J Extracell Vesicles. (2024) 13:e12484. doi: 10.1002/jev2.12484
38. Li K, Xie T, Li Y, and Huang X. LncRNAs act as modulators of macrophages within the tumor microenvironment. Carcinogenesis. (2024) 45:363–77. doi: 10.1093/carcin/bgae021
39. Fernando V, Zheng X, Sharma V, and Furuta S. Reprogramming of breast tumor-associated macrophages with modulation of arginine metabolism. bioRxiv. (2023), 2023.08.22.554238. doi: 10.1101/2023.08.22.554238
40. Fang X, Zhao P, Gao S, Liu D, Zhang S, Shan M, et al. Lactate induces tumor-associated macrophage polarization independent of mitochondrial pyruvate carrier-mediated metabolism. Int J Biol Macromol. (2023) 237:123810. doi: 10.1016/j.ijbiomac.2023.123810
41. Liu H, Yao M, and Ren J. Codonopsis pilosula-derived glycopeptide dCP1 promotes the polarization of tumor-associated macrophage from M2-like to M1 phenotype. Cancer Immunol Immunother. (2024) 73:128. doi: 10.1007/s00262-024-03694-6
43. Wang L, Guo W, Guo Z, Yu J, Tan J, Simons DL, et al. PD-L1-expressing tumor-associated macrophages are immunostimulatory and associate with good clinical outcome in human breast cancer. Cell Rep Med. (2024) 5:101420. doi: 10.1016/j.xcrm.2024.101420
44. Li Z, Duan D, Li L, Peng D, Ming Y, Ni R, et al. Tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for hepatocellular carcinoma: recent research progress. Front Pharmacol. (2024) 15:1382256. doi: 10.3389/fphar.2024.1382256
45. Zhang H, Liu L, Liu J, Dang P, Hu S, Yuan W, et al. Roles of tumor-associated macrophages in anti-PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer. (2023) 22:58. doi: 10.1186/s12943-023-01725-x
46. Zhang B, Zhang H, and Qin Y. A primer on the role of TP53 mutation and targeted therapy in endometrial cancer. Front Biosci (Landmark Ed). (2025) 30:25447. doi: 10.31083/FBL25447
47. Fan L, Xiao H, Ren J, Hou Y, Cai J, Wu W, et al. Newcastle disease virus induces clathrin-mediated endocytosis to establish infection through the activation of PI3K/AKT signaling pathway by VEGFR2. J Virol. (2024) 98:e0132224. doi: 10.1128/jvi.01322-24
48. Luecke S, Guo X, Sheu KM, Singh A, Lowe SC, Han M, et al. Dynamical and combinatorial coding by MAPK p38 and NFκB in the inflammatory response of macrophages. Mol Syst Biol. (2024) 20:898–932. doi: 10.1038/s44320-024-00047-4
49. Mabuchi S and Sasano T. Myeloid-derived suppressor cells as therapeutic targets in uterine cervical and endometrial cancers. Cells. (2021) 10:1073. doi: 10.3390/cells10051073
50. Zaporowska-Stachowiak I, Springer M, Stachowiak K, Oduah M, Sopata M, Wieczorowska-Tobis K, et al. Interleukin-6 family of cytokines in cancers. J Interferon Cytokine Res. (2024) 44:45–59. doi: 10.1089/jir.2023.0103
51. Zhang Y, Lv P, Li Y, Zhang Y, Cheng C, Hao H, et al. Inflammatory cytokine interleukin-6 (IL-6) promotes the proangiogenic ability of adipose stem cells from obese subjects via the IL-6 signaling pathway. Curr Stem Cell Res Ther. (2023) 18:93–104. doi: 10.2174/1574888X17666220429103935
52. Liu Z, Choksi S, Kwon HJ, Jiao D, Liu C, and Liu ZG. Tumor necroptosis-mediated shedding of cell surface proteins promotes metastasis of breast cancer by suppressing anti-tumor immunity. Breast Cancer Res. (2023) 25:10. doi: 10.1186/s13058-023-01604-9
53. Dash S, Sahu AK, Srivastava A, Chowdhury R, and Mukherjee S. Exploring the extensive crosstalk between the antagonistic cytokines- TGF-β and TNF-α in regulating cancer pathogenesis. Cytokine. (2021) 138:155348. doi: 10.1016/j.cyto.2020.155348
54. Douglass MS, Zhang Y, Kaplowitz MR, and Fike CD. L-citrulline increases arginase II protein levels and arginase activity in hypoxic piglet pulmonary artery endothelial cells. Pulm Circ. (2021) 11:20458940211006289. doi: 10.1177/20458940211006289
55. Tange Y, Sunakawa R, and Yoshitake S. Renal replacement therapy removes a large number of nitric oxide donors responsible for the nitrate-nitrite-nitric oxide pathway. Int J Artif Organs. (2023) 46:129–34. doi: 10.1177/03913988231157427
56. Tengbom J, Cederström S, Verouhis D, Böhm F, Eriksson P, Folkersen L, et al. Arginase 1 is upregulated at admission in patients with ST-elevation myocardial infarction. J Intern Med. (2021) 290:1061–70. doi: 10.1111/joim.v290.5
57. Damodharan SN, Walker KL, Forsberg MH, McDowell KA, Bouchlaka MN, Drier DA, et al. Analysis of ex vivo expanded and activated clinical-grade human NK cells after cryopreservation. Cytotherapy. (2020) 22:450–7. doi: 10.1016/j.jcyt.2020.05.001
58. Yokoi E, Mabuchi S, Komura N, Shimura K, Kuroda H, Kozasa K, et al. The role of myeloid-derived suppressor cells in endometrial cancer displaying systemic inflammatory response: clinical and preclinical investigations. Oncoimmunology. (2019) 8:e1662708. doi: 10.1080/2162402X.2019.1662708
59. Houthuijzen JM, de Bruijn R, van der Burg E, Drenth AP, Wientjens E, Filipovic T, et al. CD26-negative and CD26-positive tissue-resident fibroblasts contribute to functionally distinct CAF subpopulations in breast cancer. Nat Commun. (2023) 14:183. doi: 10.1038/s41467-023-35793-w
60. Zhong T, Yu J, Pan Y, Zhang N, Qi Y, and Huang Y. Recent advances of platinum-based anticancer complexes in combinational multimodal therapy. Adv Healthc Mater. (2023) 12:e2300253. doi: 10.1002/adhm.202300253
61. Su S, Chen Y, Zhang P, Ma R, Zhang W, Liu J, et al. The role of Platinum(IV)-based antitumor drugs and the anticancer immune response in medicinal inorganic chemistry. A systematic review from 2017 to 2022. Eur J Med Chem. (2022) 243:114680. doi: 10.1016/j.ejmech.2022.114680
62. Kuno I, Yoshida H, Kohno T, Ochiai A, and Kato T. Endometrial cancer arising after complete remission of uterine Malignant lymphoma: A case report and mutation analysis. Gynecol Oncol Rep. (2019) 28:50–3. doi: 10.1016/j.gore.2019.02.007
63. Ishizaka A, Taguchi A, Tsuruga T, Maruyama M, Kawata A, Miyamoto Y, et al. Endometrial cancer with concomitant endometriosis is highly associated with ovarian endometrioid carcinoma: a retrospective cohort study. BMC Womens Health. (2022) 22:332. doi: 10.1186/s12905-022-01917-5
64. Wright K, Ly T, Kriet M, Czirok A, and Thomas SM. Cancer-associated fibroblasts: master tumor microenvironment modifiers. Cancers (Basel). (2023) 15:1899. doi: 10.3390/cancers15061899
65. Axemaker H, Plesselova S, Calar K, Jorgensen M, Wollman J, and de la Puente P. Reprogramming of normal fibroblasts into ovarian cancer-associated fibroblasts via non-vesicular paracrine signaling induces an activated fibroblast phenotype. Biochim Biophys Acta Mol Cell Res. (2024) 1871:119801. doi: 10.1016/j.bbamcr.2024.119801
66. Liu X, Li J, Yang X, Li X, Kong J, Qi D, et al. Carcinoma-associated fibroblast-derived lysyl oxidase-rich extracellular vesicles mediate collagen crosslinking and promote epithelial-mesenchymal transition via p-FAK/p-paxillin/YAP signaling. Int J Oral Sci. (2023) 15:32. doi: 10.1038/s41368-023-00236-1
67. Ren B, Li X, Zhang Z, Tai S, and Yu S. Exosomes: a significant medium for regulating drug resistance through cargo delivery. Front Mol Biosci. (2024) 11:1379822. doi: 10.3389/fmolb.2024.1379822
68. Tashireva LA, Larionova IV, Ermak NA, Maltseva AA, Livanos EI, Kalinchuk AY, et al. Predicting immunotherapy efficacy in endometrial cancer: focus on the tumor microenvironment. Front Immunol. (2024) 15:1523518. doi: 10.3389/fimmu.2024.1523518
69. Albertí-Valls M, Olave S, Olomí A, Macià A, and Eritja N. Advances in immunotherapy for endometrial cancer: insights into MMR status and tumor microenvironment. Cancers (Basel). (2024) 16:3918. doi: 10.3390/cancers16233918
70. Atkins MB, Ascierto PA, Feltquate D, Gulley JL, Johnson DB, Khushalani NI, et al. Society for Immunotherapy of Cancer (SITC) consensus definitions for resistance to combinations of immune checkpoint inhibitors with targeted therapies. J Immunother Cancer. (2023) 11:e005923. doi: 10.1136/jitc-2022-005923
71. Li ZZ, He JY, Wu Q, Liu B, and Bu LL. Recent advances in targeting myeloid-derived suppressor cells and their applications to radiotherapy. Int Rev Cell Mol Biol. (2023) 378:233–64. doi: 10.1016/bs.ircmb.2023.03.007
72. Ren R, Xiong C, Ma R, Wang Y, Yue T, Yu J, et al. The recent progress of myeloid-derived suppressor cell and its targeted therapies in cancers. MedComm (2020). (2023) 4:e323. doi: 10.1002/mco2.v4.4
73. Chen H, Molberg K, Carrick K, Niu S, Rivera Colon G, Gwin K, et al. Expression and prognostic significance of LAG-3, TIGIT, VISTA, and IDO1 in endometrial serous carcinoma. Mod Pathol. (2024) 37:100532. doi: 10.1016/j.modpat.2024.100532
74. Yadav S, Gowda S, and Agrawal-Rajput R. CSF-1R blockade to alleviate azithromycin mediated immunosuppression in a mouse model of intracellular infection. Int Immunopharmacol. (2024) 143:113477. doi: 10.1016/j.intimp.2024.113477
75. Tarale P and Alam MM. Colony-stimulating factor 1 receptor signaling in the central nervous system and the potential of its pharmacological inhibitors to halt the progression of neurological disorders. Inflammopharmacology. (2022) 30:821–42. doi: 10.1007/s10787-022-00958-4
76. Abdalsalam NMF, Ibrahim A, Saliu MA, Liu TM, Wan X, and Yan D. MDSC: a new potential breakthrough in CAR-T therapy for solid tumors. Cell Commun Signal. (2024) 22:612. doi: 10.1186/s12964-024-01995-y
77. Romenskaja D, Jonavičė U, and Pivoriūnas A. Extracellular vesicles promote autophagy in human microglia through lipid raft-dependent mechanisms. FEBS J. (2024) 291:3706–22. doi: 10.1111/febs.v291.16
78. Mi C, Chen W, Zhang Y, Yang Y, Zhao J, Xu Z, et al. BaP/BPDE suppresses human trophoblast cell migration/invasion and induces unexplained miscarriage by up-regulating a novel lnc-HZ11 in extracellular vesicles: An intercellular study. Environ Int. (2024) 188:108750. doi: 10.1016/j.envint.2024.108750
79. Zhan C, Jin Y, Xu X, Shao J, and Jin C. Antitumor therapy for breast cancer: Focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med. (2023) 12:11049–72. doi: 10.1002/cam4.v12.10
80. Zhang J, Su N, Liu W, Li M, Zheng H, Li B, et al. An effective cell-penetrating peptide-based loading method to extracellular vesicles and enhancement in cellular delivery of drugs. Anal Bioanal Chem. (2025) 417. doi: 10.1007/s00216-025-05742-1
81. Lu G, Wang X, Cheng M, Wang S, and Ma K. The multifaceted mechanisms of ellagic acid in the treatment of tumors: State-of-the-art. BioMed Pharmacother. (2023) 165:115132. doi: 10.1016/j.biopha.2023.115132
Keywords: endometrial carcinoma, tumor immune microenvironment, immune evasion, treatment resistance, targeted therapy, immunotherapy
Citation: Wang Y, Liu N, Guo X, Han R, Bai J, Zhong J and Wang Q (2025) The immune microenvironment in endometrial carcinoma: mechanisms and therapeutic targeting. Front. Immunol. 16:1586315. doi: 10.3389/fimmu.2025.1586315
Received: 02 March 2025; Accepted: 27 June 2025;
Published: 17 July 2025.
Edited by:
Claudine Kieda, Military Institute of Medicine (Poland), PolandReviewed by:
Pengpeng Qu, Tianjin Central Hospital for Gynecology and Obstetrics, ChinaMaria Laura Zenclussen, National Scientific and Technical Research Council (CONICET), Argentina
Jing Hao, Shandong University, China
Copyright © 2025 Wang, Liu, Guo, Han, Bai, Zhong and Wang. 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: Qianqing Wang, d2FuZ3ExMTExQDEyNi5jb20=