Your new experience awaits. Try the new design now and help us make it even better

MINI REVIEW article

Front. Immunol., 09 July 2025

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1631226

This article is part of the Research TopicThe Role of Metabolic Reprogramming in Tumor TherapyView all 19 articles

Immunosuppressive mechanisms and therapeutic targeting of regulatory T cells in ovarian cancer

  • 1Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
  • 2School of Nursing, Yanbian University, Yanji, Jilin, China
  • 3Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
  • 4Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
  • 5The School of Clinical Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
  • 6Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning, China

Ovarian cancer remains the most lethal gynecologic malignancy, largely due to its late-stage diagnosis and immunosuppressive tumor microenvironment (TME). A key mediator of immune evasion in ovarian cancer is the infiltration and activation of regulatory T cells (Tregs), which suppress antitumor immunity and foster therapeutic resistance. Emerging therapeutic strategies to target Tregs—such as cytokine modulation, checkpoint blockade, metabolic inhibitors, and epigenetic regulators—are critically evaluated for their potential to restore antitumor immunity. This review synthesizes recent advances in understanding how the ovarian TME shapes Treg biology, highlighting mechanisms such as cytokine signaling, chemokine-driven recruitment, metabolic reprogramming, and immune checkpoint interactions, as well as the phenotypic and functional heterogeneity of tumor-infiltrating Tregs, including tissue-resident and follicular subsets, and their clonal expansion in response to tumor antigens. By elucidating the dynamic crosstalk between Tregs and the ovarian TME, this review provides a framework for developing novel immunotherapies to overcome Treg-mediated immunosuppression and improve clinical outcomes.

1 Introduction

Ovarian cancer is one of the most prevalent malignant tumors affecting the female reproductive system (1). Due to its insidious onset and lack of early clinical symptoms, more than 70% of patients are diagnosed at an advanced stage, making it the leading cause of mortality among gynecologic malignancies (2). Investigating the origins and pathogenesis of ovarian cancer is therefore of paramount importance for developing effective strategies for its prevention and treatment. Accumulating evidence indicates that the onset of ovarian cancer is closely associated with impaired anti-tumor immunity in the host (3, 4). Furthermore, tumor immunosurveillance has been shown to correlate with clinical outcomes in ovarian cancer. Immune cells and the tumor immune microenvironment (TIME) actively participate in orchestrating the initiation and progression of ovarian malignancies.

Recent studies have demonstrated substantial immune cell infiltration within the ovarian tumor microenvironment, notably an enrichment of regulatory T cells (Tregs), a suppressive T cell subset. Increased Treg abundance has been strongly linked to immune evasion, poor prognosis, and elevated mortality risk in patients with ovarian cancer (5, 6). The differentiation and effector functions of tumor-infiltrating Tregs are governed by signals derived from the tumor microenvironment, playing a pivotal role in mediating therapeutic resistance and facilitating immune escape. This review aims to summarize recent advances in our understanding of how the ovarian cancer microenvironment regulates Treg cell biology, providing insights into their role in immune suppression and therapeutic resistance.

2 Regulatory T cells

2.1 Differentiation and effector mechanisms of Treg cells

Treg cells represent a subset of CD4+ T cells characterized by the expression of the lineage-defining transcription factor forkhead box protein P3 (FOXP3) (7). These cells exert potent immunosuppressive functions and play essential roles in maintaining immune homeostasis (810). They are critically involved in the regulation of immune tolerance and are implicated in the pathogenesis of autoimmune diseases and cancer (1113). Based on their developmental origin, Treg cells can be broadly classified into thymus-derived regulatory T cells (tTregs) and peripherally induced regulatory T cells (iTregs) (9, 14, 15). tTregs originate from CD4+CD8- thymocytes that exhibit high-affinity recognition of self-antigens (16). Upon T cell receptor (TCR) signaling and activation of the interleukin-2 (IL-2) pathway, FOXP3 expression is induced, conferring suppressive identity (17, 18). In contrast, iTregs differentiate from naive peripheral CD4+ T cells in response to TCR stimulation and exposure to cytokines such as IL-2 and transforming growth factor-β (TGF-β), which collectively induce FOXP3 expression (11, 19, 20). Functionally, Tregs mediate immune regulation by modulating immune tolerance and suppressing the activity of effector T cells, macrophages, dendritic cells, and B cells, primarily via immunosuppressive molecules and cytokines (2125).

Treg cells express high levels of immunoregulatory surface molecules including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), CD39, and T cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) (2630). CTLA-4 on Tregs can bind the co-stimulatory molecules CD80 and CD86 on dendritic cells (DCs), thereby inhibiting antigen presentation (31, 32). In addition, CTLA-4 induces the expression of indoleamine 2,3-dioxygenase (IDO) in DCs, reducing tryptophan availability—a crucial nutrient for effector T cell proliferation and activation—thus dampening T cell responses (33). The ectoenzymes CD39 and CD73 on Tregs catalyze the hydrolysis of extracellular ATP to adenosine, a potent immunosuppressive metabolite that inhibits DC-mediated antigen presentation and T cell proliferation (3437). TIGIT expression on Tregs is associated with their activation state and contributes to suppression by inducing the expression of fibrinogen-like protein 2 (Fgl2), which inhibits the activation of effector T cells and selectively attenuates inflammation mediated by T helper 1 (Th1) and Th17 cells (38). Tregs also exert suppressive functions through the secretion of cytokines such as TGF-β, IL-10, and IL-35, which inhibit both T cell and B cell effector functions (39). Upon activation, Tregs can secrete perforin and granzymes, leading to cytotoxic activity against effector CD4+ and CD8+ T cells (40). Moreover, as T cell proliferation and differentiation are IL-2-dependent processes, the high expression of CD25 on Tregs enables them to outcompete effector T cells for IL-2, resulting in cytokine deprivation that impairs effector T cell responses and contributes to immune suppression within the tumor microenvironment (41).

2.2 Treg cells in the ovarian cancer microenvironment

In patients with ovarian cancer, Treg cells are markedly enriched in both tumor tissues and ascites. These tumor-infiltrating Tregs (Ti-Tregs) can suppress anti-tumor effector responses and promote immune evasion, thereby facilitating tumor progression (42). Recent studies employing high-throughput sequencing have revealed distinct phenotypic and functional traits of Tregs within the ovarian tumor microenvironment. Laumont et al. (43) identified a subset of CD39+CD103+PD-1+ Tregs within tumors, displaying enhanced TCR diversity and a tissue-resident phenotype. Another study revealed that CEACAM1+ Tregs preferentially accumulate in tumor sites, are highly activated, and exhibit strong suppressive capacity. The accumulation of CEACAM1+ Tregs correlates with tumor progression; notably, their depletion enhances tumor-infiltrating lymphocyte (TIL) function and potentiates the therapeutic efficacy of anti-programmed death-1 (PD-1) therapy (44). TCR repertoire sequencing of Tregs in ovarian and other solid tumors has demonstrated considerable clonality and responsiveness to tumor-associated antigens, suggesting that intratumoral Tregs undergo clonal expansion and selection driven by antigen stimulation (45). Furthermore, a distinct subset of follicular regulatory T (Tfr) cells, which express chemokine receptor CXCR5, has been identified within the ovarian tumor microenvironment. Tfr cells, which typically reside in the germinal centers of secondary lymphoid tissues, regulate B cell responses. In ovarian cancer tissues and ascitic fluid, Tfr cells have been found to infiltrate and express high levels of TGFB1 and IL-10. Through IL-10 secretion, Tfr cells suppress the activation and cytotoxic function of CD8+ T cells, thereby contributing to an immunosuppressive microenvironment (46).

3 Mechanisms of Treg regulation by the ovarian cancer microenvironment

3.1 Cytokines regulate Treg biology

Tregs in ovarian cancer commonly express high levels of CD4, CD25, and FOXP3. Their increased presence is associated with immune evasion, lower survival rates, and elevated mortality risk in patients with ovarian cancer (6). Toker et al. (47), utilizing spatial transcriptomics, single-cell RNA sequencing, and TCR sequencing, identified the enrichment and heterogeneity of CD4+ Tregs in immune “cold” ovarian tumors. These findings suggest that Tregs constitute an immunosuppressive tumor microenvironment (TME) and are regulated by multiple factors.

IL-2 signaling is essential for Treg development. Studies show ovarian tumor-derived CD4+CD25+ Tregs exhibit IL-2-dependent Th17 plasticity under CD3/APC stimulation, revealing microenvironmental modulation of Treg function (48). While high-affinity IL-2R inhibition reduces Tregs and tumor progression, it compromises effector T cells and induces autoimmunity. Drerup et al. (49) demonstrated low-affinity IL-2Rβ engagement increases Treg numbers while impairing suppression, improving CD8+/Treg ratios and tumor control, suggesting IL-2Rβ as a therapeutic target. TGF-β signaling similarly regulates Treg biology, with ovarian cancer cells secreting TGF-β to recruit tTregs and induce iTregs (50). TGF-β neutralization inhibits tumor progression, reduces ascites, and enhances CD8+/Treg ratios (51), while TGFBR2 SNPs further implicate this pathway in Treg modulation (52). Additional cytokines contribute to Treg regulation: M2-TAMs secrete TGF-α, IL-6, and IL-10 to maintain Tregs (53, 54), whereas Treg depletion enables IFN-α to stimulate DC-derived IL-6 and antitumor immunity (55) (Figure 1).

Figure 1
Diagram illustrating the TME-mediated induction of immunosuppressive Tregs in ovarian cancer. It includes various elements such as cytokines, chemokines, metabolic regulators, transcription factors, epigenetic modulators, cell-cell interactions, immune checkpoints, surface receptors, and other factors. Arrows and labels indicate interactions and processes influenced by these components, with ovarian cancer depicted at the center.

Figure 1. TME-mediated induction of immunosuppressive Tregs in ovarian cancer.

3.2 Chemokines mediate Treg trafficking and enrichment in the TME

Chemokines mediate Treg trafficking and enrichment in the TME. The CCL28 and CCL22/CCL17–CCR4 signaling axes selectively recruit Tregs into ovarian tumors (56, 57). In hypoxic in vitro ovarian cancer models, CCL28 expression is induced, promoting Treg migration via CCR10 engagement and suppressing host antitumor responses (56). Similarly, TAMs in ovarian tumors and ascites produce abundant CCL22, attracting Tregs and suppressing immune surveillance, whereas CCL22 inhibitors reduce T cell migration (57). Additional chemokines involved in Treg trafficking include CCR6/CCL20, CD11a, and integrins such as CD49d/CD29 (58, 59). Although CCR6 is highly expressed in hepatocellular and colorectal carcinomas and plays a role in autoimmune diseases (60), its contribution in ovarian cancer remains to be elucidated. Inhibition of the CXCR4–CXCL12 axis diminishes intratumoral Tregs and facilitates their conversion to helper T cells, enhancing intratumoral immune responses (61). Idorn et al. (62), using flow cytometry, revealed that FOXP3- Treg infiltration in ovarian cancer correlates with CCR4 expression and enrichment of CCR4+, CCR5+, CXCR3+, and CXCR4+ T cells in both ascitic fluid and peripheral blood, linking this to elevated CCL22, CXCL9/10, and CXCL12 levels (Figure 1).

3.3 Immune checkpoints and surface receptors

Tregs in ovarian cancer highly express PD-1, which upon ligand binding suppresses immune activity. Tumor cells exploit this by upregulating PD-1 on Tregs to facilitate immune tolerance. PD-1 promotes apoptosis of inflammatory T cells while inhibiting apoptosis of Tregs, thereby increasing their proportion and contributing to immune suppression (47, 63). Sato et al. (64) found that CD45RA-FOXP3+ effector Tregs in ascites correlated with elevated PD-1 expression on CD8+ T cells. Compared to primary tumors, peritoneally metastasized ovarian cancers express higher PD-L1 levels, potentially enhancing Treg activation and promoting tumor progression (65). Tregs in ovarian cancer also overexpress TNFR2, a potent immunosuppressive receptor, likely in response to elevated IL-6 in the TME (66). High CCR4 expression in the TME facilitates TNFR2+ Treg accumulation in tumors and ascites (67). Active TNFR2 supports cell growth and modulates proliferation-apoptosis balance (68). TNF signaling enhances FOXP3 expression, maintaining the suppressive Treg phenotype (69). High CD73 expression in ovarian tumors correlates with poor prognosis. The CD73/adenosine axis supports the accumulation of Tregs and M2 macrophages, with Tregs suppressing antitumor immunity via CD73-dependent adenosine production (70). TIGIT, an inhibitory checkpoint molecule, is upregulated on CD4+ Tregs in murine ovarian cancer models. Anti-TIGIT antibody blockade reduces Treg numbers and suppressive function without affecting CD4+, CD8+, or NK cells, thereby improving survival (71). Recent clinical trials on ovarian cancer, including JAVELIN Ovarian 100 (NCT02718417) and JAVELIN Ovarian 200 (NCT02580058), demonstrated that the PD-L1 inhibitor avelumab, either as monotherapy or in combination with chemotherapy, did not significantly improve progression-free survival (PFS) or overall survival (OS) compared to chemotherapy alone (72, 73). Similarly, the KEYNOTE-100 trial evaluating pembrolizumab monotherapy reported a low objective response rate (ORR) (approximately 9.9%) and short duration of response (DOR) in patients with recurrent ovarian cancer (74). These findings suggest that immune checkpoint inhibitor monotherapy has limited efficacy in ovarian cancer, which may be attributed to Treg-mediated immunosuppression within the tumor microenvironment. These results underscore the need for rational combination strategies that concurrently target Tregs and stimulate effector immune responses to overcome the observed clinical resistance to immunotherapy (Figure 1).

3.4 Metabolic regulators

The aberrant metabolic milieu and metabolic byproducts in tumors influence Treg biology. Tregs in ovarian cancer tissues exhibit increased apoptosis, partially induced by adenosine, which also mediates immunosuppression. Maj et al. (36) found that apoptotic Tregs convert ATP to adenosine via CD39 and CD73, releasing adenosine that activates A2A receptors and dampens immune responses. Treg apoptosis is linked to their vulnerability to reactive oxygen species and diminished NRF2 antioxidant signaling, suggesting that oxidative stress-induced cell death enhances their immunosuppressive capacity. IDO also modulates Tregs by metabolizing tryptophan into kynurenine, which binds to aryl hydrocarbon receptors (AHRs) on T cells, skewing the Th17/Treg balance toward Tregs. Kynurenine also binds AHRs on TAMs, creating a feedback loop that upregulates IDO. Ovarian cancer progression is marked by a shift from Th17 dominance to Treg dominance, implicating metabolic byproducts in local immune modulation (75). Tryptophan deprivation induces AHR overexpression, promoting kynurenine uptake, AHR pathway activation, and Treg differentiation (76). Glucose metabolism also influences Treg function. Xu et al. (77) reported that Tregs in ovarian cancer overexpress glucose transporter 1 (GLUT1) and hypoxia-inducible factor 1-alpha (HIF-1α). TLR8 signaling suppresses mTOR activity, thereby modulating glycolysis and suppressing Treg functionality (Figure 1).

3.5 Transcription factors and epigenetic modulators

FOXP3 is the master transcription factor of Tregs. Studies on FOXP3 polymorphisms in ovarian cancer identified SNPs rs3761548A/C and rs5902434del/ATT as associated with epithelial ovarian tumor susceptibility and prognosis. The rs3761548A/C variant confers increased susceptibility, while rs5902434del/ATT is an independent prognostic factor (78). Epigenetic regulators also control Tregs. Class I histone deacetylases (HDACs) maintain Treg function; inhibition of HDACs suppresses Tregs, restores the CD8+/FOXP3+ Treg ratio, and reduces ascites (79). N6-methyladenosine (m6A), a key mRNA modification, regulates immunity via m6A reader proteins. TNF signaling induces expression of YTHDF2 in Tregs, which accelerates degradation of NF-κB repressors, thereby enhancing Treg activation and suppressive function. Conditional YTHDF2 deletion in Tregs impairs their function, increases apoptosis, and limits tumor growth, confirming the role of m6A regulation in tumor Tregs (80, 81) (Figure 1).

3.6 Cell-cell interactions

Tumor-associated DCs can promote Treg induction. Dysfunctional DCs in ovarian tumors facilitate Treg conversion (82). Conrad et al. (83) demonstrated that tumor-infiltrating Tregs overexpress ICOS, whose expansion depends on interactions with ICOS ligand on plasmacytoid DCs, promoting Treg proliferation and suppressive function. Additionally, DCs can induce IDO expression, facilitating Treg differentiation and T cell exhaustion (84). TAMs recruit CCR4+ Tregs via CCL22 secretion, suppressing T cell proliferation and enhancing immune evasion. Myeloid-derived suppressor cells (MDSCs) promote iTreg differentiation and tTreg expansion through CD40-CD40L, IFN-γ, IL-10, and TGF-β pathways (8587). Cancer-associated fibroblasts (CAFs) also contribute to immune suppression. Givel et al. (88) found that the CAF-S1 subset in high-grade serous ovarian cancer (HGSOC) is enriched in mesenchymal tumors and promotes Treg chemotaxis, survival, and differentiation via a miR-141/miR-200a-dependent CXCL12β mechanism.

Theodoraki et al. (89) showed that TLR3 agonists such as Sendai virus (SeV), poly-I:C, and rintatolimod (poly-I:C12U) activate IFN-α and CXCL10 expression, enhancing T cell infiltration. These agonists also stimulate MAVS signaling, inhibiting NF-κB and TNF-α-dependent COX2 activation. The COX2/PGE2 pathway promotes Treg infiltration by inducing IDO, IL-10, CCL22, and CXCL12. Extracellular vesicles also regulate Tregs in ovarian cancer. TAM-derived exosomes enriched in miR-29a-3p and miR-21-5p inhibit STAT3 in CD4+ T cells, promoting Treg differentiation and expression of TGF-β and IL-10, while suppressing Th17 differentiation and TNF-α/IL-6 secretion (90) (Figure 1).

4 Ovarian cancer therapy targeting Treg in the TME

4.1 Inhibiting Treg proliferation and recruitment

Given the pivotal immunosuppressive role of Tregs in ovarian cancer, targeted strategies to disrupt their function have emerged as promising therapeutic approaches. While IL-2 demonstrates clinical efficacy with response rates of 16-20% in cancer patients, its therapeutic potential is limited by the paradoxical expansion of CD4+CD25+FOXP3+ Tregs (91). To overcome this limitation, novel agents have been developed, including Ontak, a fusion protein of IL-2 and diphtheria toxin, which selectively depletes Tregs through inhibition of protein synthesis, and nemvaleukin alfa, an engineered cytokine that preferentially activates CD8+ T cells and NK cells while minimizing Treg expansion (NCT02799095) (92, 93). Alternative strategies focus on blocking key Treg pathways, such as CTLA-4 inhibition with MDX-CTLA-4 which reduces CA125 levels, or CCR4 targeting with mAb2–3 that stimulates IFN-γ while suppressing IL-2-driven Treg proliferation (94, 95). Additionally, immunotherapeutic approaches like the GVAX whole-cell vaccine enhance antitumor immunity through GM-CSF-mediated CTL infiltration and Treg reduction, demonstrating the potential of combinatorial strategies to effectively modulate the immunosuppressive tumor microenvironment in ovarian cancer (96).

4.2 Modulate Treg biosynthesis, differentiation, or suppressive function

Cyclophosphamide, an alkylating agent that disrupts DNA Treatment with MDX-CTLA-4 in ovarian cancer patients replication in rapidly dividing cells, preferentially eliminates highly proliferative Tregs in the tumor milieu at low doses, thereby potentiating antitumor immunity (97). Combined use of cyclophosphamide with intratumoral Bacillus Calmette–Guérin (BCG) vaccination has been shown to diminish Treg frequencies while enhancing CD8+ T cell infiltration (98). Other compounds, including mitoxantrone, the glycolytic inhibitor 2-deoxy-D-glucose (2-DG), and IDO inhibitors, similarly reduce Treg numbers and impair their immunosuppressive activity (99101). Recent findings indicate that anionic liposomal delivery of Toll-like receptor (TLR) antagonists in ovarian cancer leads to a reduction in Treg accumulation, concurrent with elevated T cell infiltration and M1 macrophage polarization within the TME (102).

4.3 Inhibition of tumor angiogenesis

Another therapeutic axis centers on the inhibition of tumor angiogenesis. Vascular endothelial growth factor (VEGF), often overexpressed in the TME, has been implicated in the recruitment of Tregs. Application of anti-VEGF monoclonal antibodies in ovarian cancer has been associated with a reduction in circulating Tregs and a concomitant rise in effector T cell populations (103). Programmed death-1 (PD-1), an immunoinhibitory receptor, facilitates immune escape in tumors (104). OX40, a co-stimulatory receptor expressed on activated T cells and Tregs, belongs to the tumor necrosis factor receptor superfamily and plays a crucial role in T cell activation and expansion. Notably, combinatorial blockade of PD-1 and activation of OX40 signaling induces a robust immunostimulatory response in ovarian cancer murine models, characterized by elevated IFN-γ levels and diminished Treg infiltration (105). Furthermore, CCR4 is highly expressed on Tregs, and chemokines secreted by tumor cells can attract CCR4+ Tregs into the tumor niche (106). Consequently, targeting CCR4 in clinical trials holds potential to provide a new and reliable strategy for immunotherapeutic intervention in ovarian cancer (Table 1).

Table 1
www.frontiersin.org

Table 1. Therapeutic strategies targeting Tregs in ovarian cancer.

5 Conclusion

Ovarian cancer remains a formidable challenge in oncology, with its immunosuppressive tumor microenvironment playing a critical role in disease progression and therapeutic resistance. Regulatory T cells are key mediators of immune evasion, suppressing antitumor responses through multiple mechanisms, including cytokine secretion, metabolic regulation, and immune checkpoint interactions. Despite advances in immunotherapy, clinical trials targeting PD-1/PD-L1 in ovarian cancer have shown limited efficacy, underscoring the need for novel strategies that disrupt Treg-mediated immunosuppression. Emerging approaches such as Treg depletion, inhibition of recruitment signals, metabolic modulation, and combination therapies targeting immune checkpoints hold promise for restoring antitumor immunity. Future research should focus on identifying biomarkers for patient stratification and optimizing combinatorial regimens that simultaneously neutralize Treg suppression while enhancing effector T cell function. Overcoming Treg-driven resistance will be crucial for improving outcomes in ovarian cancer immunotherapy.

Author contributions

JL: Writing – original draft. HH: Writing – original draft. RX: Writing – original draft. RY: Writing – original draft. HW: Writing – original draft. LW: Funding acquisition, Writing – original draft, 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 First Hospital of China Medical University.

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. Roett MA and Evans P. Ovarian cancer: an overview. Am Fam Physician. (2009) 80:609–16.

PubMed Abstract | Google Scholar

2. Renz M, Kehoe S, Kumar L, and Friedlander M. Cancer of the ovary, fallopian tube, and peritoneum: 2021 update. Int J Gynaecol Obstet. (2021) 155 Suppl 1:61–85. doi: 10.1002/ijgo.13878

PubMed Abstract | Crossref Full Text | Google Scholar

3. Müller V, Wöckel A, Lux MP, Janni W, Ettl J, Belleville E, et al. Translational highlights in breast and ovarian cancer 2019 - immunotherapy, DNA repair, PI3K inhibition and CDK4/6 therapy. Geburtshilfe Frauenheilkd. (2019) 79:1309–19. doi: 10.1055/a-1039-4458

PubMed Abstract | Crossref Full Text | Google Scholar

4. Lynam S, Lugade AA, and Odunsi K. Immunotherapy for gynecologic cancer: current applications and future directions. Clin Obstet Gynecol. (2020) 63:48–63. doi: 10.1097/GRF.0000000000000513

PubMed Abstract | Crossref Full Text | Google Scholar

5. Cimino-Mathews A, Lee JS, Nair J, Lee MJ, Yuno A, An D, et al. Clinical outcomes of prexasertib monotherapy in recurrent BRCA wild-type high-grade serous ovarian cancer involve innate and adaptive immune responses. J Immunother Cancer. (2020) 8:e000516. doi: 10.1136/jitc-2019-000516

PubMed Abstract | Crossref Full Text | Google Scholar

6. Kandalaft LE, Dangaj Laniti D, and Coukos G. Immunobiology of high-grade serous ovarian cancer: lessons for clinical translation. Nat Rev Cancer. (2022) 22:640–56. doi: 10.1038/s41568-022-00503-z

PubMed Abstract | Crossref Full Text | Google Scholar

7. Wing JB, Tanaka A, and Sakaguchi S. Human FOXP3(+) regulatory T cell heterogeneity and function in autoimmunity and cancer. Immunity. (2019) 50:302–16. doi: 10.1016/j.immuni.2019.01.020

PubMed Abstract | Crossref Full Text | Google Scholar

8. Bednar KJ, Lee JH, and Ort T. Tregs in autoimmunity: insights into intrinsic brake mechanism driving pathogenesis and immune homeostasis. Front Immunol. (2022) 13:932485. doi: 10.3389/fimmu.2022.932485

PubMed Abstract | Crossref Full Text | Google Scholar

9. Ohkura N and Sakaguchi S. Transcriptional and epigenetic basis of Treg cell development and function: its genetic anomalies or variations in autoimmune diseases. Cell Res. (2020) 30:465–74. doi: 10.1038/s41422-020-0324-7

PubMed Abstract | Crossref Full Text | Google Scholar

10. Ding M, Wang Q, Song Y, Huo K, Chen X, Xiang Z, et al. Advances in Foxp3+ regulatory T cells (Foxp3+ Treg) and key factors in digestive Malignancies. Front Immunol. (2024) 15:1404974. doi: 10.3389/fimmu.2024.1404974

PubMed Abstract | Crossref Full Text | Google Scholar

11. Mikami N and Sakaguchi S. Regulatory T cells in autoimmune kidney diseases and transplantation. Nat Rev Nephrol. (2023) 19:544–57. doi: 10.1038/s41581-023-00733-w

PubMed Abstract | Crossref Full Text | Google Scholar

12. Lu L, Barbi J, and Pan F. The regulation of immune tolerance by FOXP3. Nat Rev Immunol. (2017) 17:703–17. doi: 10.1038/nri.2017.75

PubMed Abstract | Crossref Full Text | Google Scholar

13. Li J, Bai L, Jiang M, Janke L, Zong X, Hao X, et al. A distal Foxp3 enhancer enables interleukin-2 dependent thymic Treg cell lineage commitment for robust immune tolerance. Immunity. (2021) 54:931–46.e11. doi: 10.1016/j.immuni.2021.03.020

PubMed Abstract | Crossref Full Text | Google Scholar

14. Chu KH and Chiang BL. A novel subset of regulatory T cells induced by B cells alleviate the severity of immunological diseases. Clin Rev Allergy Immunol. (2024) 67:73–82. doi: 10.1007/s12016-024-09009-y

PubMed Abstract | Crossref Full Text | Google Scholar

15. Hou P, Lin H, Chen M, Wang R, and Xu T. Inhibition of Lck/Fyn kinase activity promotes the differentiation of induced Treg cells through AKT/mTOR pathway. Int Immunopharmacol. (2024) 134:112237. doi: 10.1016/j.intimp.2024.112237

PubMed Abstract | Crossref Full Text | Google Scholar

16. Chen W and Konkel JE. Development of thymic Foxp3(+) regulatory T cells: TGF-β matters. Eur J Immunol. (2015) 45:958–65. doi: 10.1002/eji.201444999

PubMed Abstract | Crossref Full Text | Google Scholar

17. Su Y, Jiao A, Wang X, and Zhang B. T cells in health and disease. Signal Transduct Target Ther. (2023) 8:235. doi: 10.1038/s41392-023-01471-y

PubMed Abstract | Crossref Full Text | Google Scholar

18. Khalsa JK, Dhar A, Gupta S, Umar D, Arimbasseri GA, Bal V, et al. A role for cell-autocrine interleukin-2 in regulatory T-cell homeostasis. Immunology. (2020) 160:295–309. doi: 10.1111/imm.13194

PubMed Abstract | Crossref Full Text | Google Scholar

19. Zhu H, Wang P, Que W, Zhong L, Li XK, and Du F. Different subpopulations of regulatory T cells in human autoimmune disease, transplantation, and tumor immunity. MedComm (2020). (2022) 3:e137. doi: 10.1002/mco2.137

PubMed Abstract | Crossref Full Text | Google Scholar

20. Nakatsukasa H, Okada M, Lu Q, and Yoshimura A. Induced regulatory T cells: their development, stability, and applications. Trends Immunol. (2016) 37:803–11. doi: 10.1016/j.it.2016.08.012

PubMed Abstract | Crossref Full Text | Google Scholar

21. Iglesias-Escudero M, Arias-González N, and Martínez-Cáceres E. Regulatory cells and the effect of cancer immunotherapy. Mol Cancer. (2023) 22:26. doi: 10.1186/s12943-023-01714-0

PubMed Abstract | Crossref Full Text | Google Scholar

22. Nishikawa H and Koyama S. Mechanisms of regulatory T cell infiltration in tumors: implications for innovative immune precision therapies. J Immunother Cancer. (2021) 9:e002591. doi: 10.1136/jitc-2021-002591

PubMed Abstract | Crossref Full Text | Google Scholar

23. Ganesan R, Singh S, and Grewal IS. Regulatory T cell targeting in cancer: Emerging strategies in immunotherapy. Eur J Immunol. (2021) 51:280–91. doi: 10.1002/eji.202048992

PubMed Abstract | Crossref Full Text | Google Scholar

24. Vilbois S, Xu Y, and Ho PC. Metabolic interplay: tumor macrophages and regulatory T cells. Trends Cancer. (2024) 10:242–55. doi: 10.1016/j.trecan.2023.11.007

PubMed Abstract | Crossref Full Text | Google Scholar

25. Yin W, Guan F, Zeng W, Lee P, Candotti F, James LK, et al. B cell-T cell interplay in immune regulation: A focus on follicular regulatory T and regulatory B cell functions. Front Cell Dev Biol. (2022) 10:991840. doi: 10.3389/fcell.2022.991840

PubMed Abstract | Crossref Full Text | Google Scholar

26. Davies SP, Fiancette R, Wootton GE, Faustini S, Kwok HF, Richardson N, et al. The dual role of TIGIT in regulatory and effector T cells in chronic liver disease. JHEP Rep. (2025) 7:101405. doi: 10.1016/j.jhepr.2025.101405

PubMed Abstract | Crossref Full Text | Google Scholar

27. Blinova VG and Zhdanov DD. Many faces of regulatory T cells: heterogeneity or plasticity? Cells. (2024) 13:959. doi: 10.3390/cells13110959

PubMed Abstract | Crossref Full Text | Google Scholar

28. Tay C, Tanaka A, and Sakaguchi S. Tumor-infiltrating regulatory T cells as targets of cancer immunotherapy. Cancer Cell. (2023) 41:450–65. doi: 10.1016/j.ccell.2023.02.014

PubMed Abstract | Crossref Full Text | Google Scholar

29. Annese T, Tamma R, and Ribatti D. Update in TIGIT immune-checkpoint role in cancer. Front Oncol. (2022) 12:871085. doi: 10.3389/fonc.2022.871085

PubMed Abstract | Crossref Full Text | Google Scholar

30. Liu X, Gu Z, Jiang Z, Zhao S, Song Y, and Yu J. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. biomark Res. (2024) 12:7. doi: 10.1186/s40364-023-00543-z

PubMed Abstract | Crossref Full Text | Google Scholar

31. Zheng Y, Nakamura K, Attridge K, Manzotti C, Schmidt EM, Baker J, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science. (2011) 332:600–3. doi: 10.1126/science.1202947

PubMed Abstract | Crossref Full Text | Google Scholar

32. Bosma DMT, Busselaar J, Battaglia TW, Staal MD, Verbrugge I, and Borst J. PD-1 or CTLA-4 blockade promotes CD86-driven Treg responses upon radiotherapy of lymphocyte-depleted cancer in mice. J Clin Invest. (2024) 134:e171154. doi: 10.1172/JCI171154

PubMed Abstract | Crossref Full Text | Google Scholar

33. Orabona C, Fallarino F, Vacca C, Calcinaro F, Falorni A, Candeloro P, et al. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat Immunol. (2002) 3:1097–101. doi: 10.1038/ni846

PubMed Abstract | Crossref Full Text | Google Scholar

34. Brigida I, Carriglio N, Hernandez RJ, Scaramuzza S, Clavenna D, Sanvito F, et al. Alterations in the adenosine metabolism and CD39/CD73 adenosinergic machinery cause loss of Treg cell function and autoimmunity in ADA-deficient SCID. Blood. (2012) 119:1428–39. doi: 10.1182/blood-2011-07-366781

PubMed Abstract | Crossref Full Text | Google Scholar

35. Chen L, Enk A, Ring S, and Mahnke K. The multifaceted actions of CD73 during development and suppressive actions of regulatory T cells. Front Immunol. (2022) 13:914799. doi: 10.3389/fimmu.2022.914799

PubMed Abstract | Crossref Full Text | Google Scholar

36. Wang W, Crespo J, Zhang H, Wang W, Wei S, Zhao L, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol. (2017) 18:1332–41. doi: 10.1038/ni.3868

PubMed Abstract | Crossref Full Text | Google Scholar

37. Min YK, Jang J, Park H, Lee S, and Lee CH. Single-cell RNA sequencing reveals distinct cellular factors for response to immunotherapy targeting CD73 and PD-1 in colorectal cancer. J Immunother Cancer. (2021) 9:e002503. doi: 10.1136/jitc-2021-002503

PubMed Abstract | Crossref Full Text | Google Scholar

38. Lozano E, Burkett PR, Patel B, Xiao S, Zhu C, Xia J, et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity. (2014) 40:569–81. doi: 10.1016/j.immuni.2014.02.012

PubMed Abstract | Crossref Full Text | Google Scholar

39. Dikiy S and Rudensky AY. Principles of regulatory T cell function. Immunity. (2023) 56:240–55. doi: 10.1016/j.immuni.2023.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

40. Verbsky JW, Barchet W, Colonna M, Atkinson JP, and Ley TJ. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. (2004) 21:589–601. doi: 10.1016/j.immuni.2004.09.002

PubMed Abstract | Crossref Full Text | Google Scholar

41. Togashi Y, Shitara K, and Nishikawa H. Regulatory T cells in cancer immunosuppression - implications for anticancer therapy. Nat Rev Clin Oncol. (2019) 16:356–71. doi: 10.1038/s41571-019-0175-7

PubMed Abstract | Crossref Full Text | Google Scholar

42. Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, and Osada J. Crosstalk of immune cells and platelets in an ovarian cancer microenvironment and their prognostic significance. Int J Mol Sci. (2023) 24:9279. doi: 10.3390/ijms24119279

PubMed Abstract | Crossref Full Text | Google Scholar

43. Wouters MCA, Smazynski J, Gierc NS, Chavez EA, Chong LC, Thornton S, et al. Single-cell profiles and prognostic impact of tumor-infiltrating lymphocytes coexpressing CD39, CD103, and PD-1 in ovarian cancer. Clin Cancer Res. (2021) 27:4089–100. doi: 10.1158/1078-0432.CCR-20-4394

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kang M, Jeon M, Chung Y, Kim AR, Lee YJ, Kim ES, et al. CEACAM1 marks highly suppressive intratumoral regulatory T cells for targeted depletion therapy. Clin Cancer Res. (2023) 29:1794–806. doi: 10.1158/1078-0432.CCR-22-1843

PubMed Abstract | Crossref Full Text | Google Scholar

45. Pasetto A, Jia L, Deniger DC, Stevanović S, Robbins PF, and Rosenberg SA. Tumor-infiltrating human CD4(+) regulatory T cells display a distinct TCR repertoire and exhibit tumor and neoantigen reactivity. Sci Immunol. (2019) 4:eaao4310. doi: 10.1126/sciimmunol.aao4310

PubMed Abstract | Crossref Full Text | Google Scholar

46. Li L, Ma Y, and Xu Y. Follicular regulatory T cells infiltrated the ovarian carcinoma and resulted in CD8 T cell dysfunction dependent on IL-10 pathway. Int Immunopharmacol. (2019) 68:81–7. doi: 10.1016/j.intimp.2018.12.051

PubMed Abstract | Crossref Full Text | Google Scholar

47. Nguyen LT, Stone SC, Yang SYC, Katz SR, Shaw PA, Clarke BA, et al. Regulatory T cells in ovarian cancer are characterized by a highly activated phenotype distinct from that in melanoma. Clin Cancer Res. (2018) 24:5685–96. doi: 10.1158/1078-0432.CCR-18-0554

PubMed Abstract | Crossref Full Text | Google Scholar

48. Deknuydt F, Bioley G, Old LJ, Matsuzaki J, Odunsi K, Ayyoub M, et al. Interleukin 2-mediated conversion of ovarian cancer-associated CD4+ regulatory T cells into proinflammatory interleukin 17-producing helper T cells. J Immunother. (2009) 32:101–8. doi: 10.1097/CJI.0b013e318195b59e

PubMed Abstract | Crossref Full Text | Google Scholar

49. Deng Y, Pandeswara SL, Padrón Á S, Reyes RM, Zhang X, Mendez J, et al. CD122-selective IL2 complexes reduce immunosuppression, promote treg fragility, and sensitize tumor response to PD-L1 blockade. Cancer Res. (2020) 80:5063–75. doi: 10.1158/0008-5472.CAN-20-0002

PubMed Abstract | Crossref Full Text | Google Scholar

50. Liu M, Nixon BG, Kang D, Toure A, Bivona M, Li MO, et al. Foxp3-independent mechanism by which TGF-β controls peripheral T cell tolerance. Proc Natl Acad Sci U.S.A. (2017) 114:E7536–e7544. doi: 10.1073/pnas.1706356114

PubMed Abstract | Crossref Full Text | Google Scholar

51. Meza-Perez S, Katre AA, Goldsberry WN, Randall TD, Norian LA, Birrer MJ, et al. Neutralization of TGFβ Improves tumor immunity and reduces tumor progression in ovarian carcinoma. Mol Cancer Ther. (2021) 20:602–11. doi: 10.1158/1535-7163.MCT-20-0412

PubMed Abstract | Crossref Full Text | Google Scholar

52. Sucheston-Campbell LE, Cannioto R, Chang-Claude J, Modugno F, Dörk T, Hillemanns P, et al. Assessment of variation in immunosuppressive pathway genes reveals TGFBR2 to be associated with risk of clear cell ovarian cancer. Oncotarget. (2016) 7:69097–110. doi: 10.18632/oncotarget.10215

PubMed Abstract | Crossref Full Text | Google Scholar

53. Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. (2004) 10:942–9. doi: 10.1038/nm1093

PubMed Abstract | Crossref Full Text | Google Scholar

54. Noy R and Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. (2014) 41:49–61. doi: 10.1016/j.immuni.2014.06.010

PubMed Abstract | Crossref Full Text | Google Scholar

55. Barnett BB, Pandeswara S, Wall SR, Hurez V, Dao V, Sun L, et al. IFNα Augments clinical efficacy of regulatory T-cell depletion with denileukin diftitox in ovarian cancer. Clin Cancer Res. (2021) 27:3661–73. doi: 10.1158/1078-0432.CCR-20-4594

PubMed Abstract | Crossref Full Text | Google Scholar

56. Peng X, Hagemann IS, Balint K, Barchetti A, Wang LP, Gimotty PA, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and T(reg) cells. Nature. (2011) 475:226–30. doi: 10.1038/nature10169

PubMed Abstract | Crossref Full Text | Google Scholar

57. Treilleux I, Bendriss-Vermare N, Bachelot T, Goddard-Leon S, Arfi V, Biota C, et al. Regulatory T cells recruited through CCL22/CCR4 are selectively activated in lymphoid infiltrates surrounding primary breast tumors and lead to an adverse clinical outcome. Cancer Res. (2009) 69:2000–9. doi: 10.1158/0008-5472.CAN-08-2360

PubMed Abstract | Crossref Full Text | Google Scholar

58. Kunder N, Inaku C, Hagan NB, Gibbons G, Mathey-Andrews N, Anandappa G, et al. Longitudinal intravascular antibody labeling identified regulatory T cell recruitment as a therapeutic target in a mouse model of lung cancer. J Immunol. (2024) 213:906–18. doi: 10.4049/jimmunol.2400268

PubMed Abstract | Crossref Full Text | Google Scholar

59. Yoshitomi H, Hashimoto M, Maeda S, Teradaira S, Sugimoto N, Yamaguchi T, et al. Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. J Exp Med. (2007) 204:2803–12. doi: 10.1084/jem.20071397

PubMed Abstract | Crossref Full Text | Google Scholar

60. Rubie C, Keilholz U, and Ghadjar P. Chemokine/chemokine receptor pair CCL20/CCR6 in human colorectal Malignancy: An overview. World J Gastroenterol. (2016) 22:833–41. doi: 10.3748/wjg.v22.i2.833

PubMed Abstract | Crossref Full Text | Google Scholar

61. Li B, Liang Y, Reeves PM, Qu X, Ran C, Liu Q, et al. Dual blockade of CXCL12-CXCR4 and PD-1-PD-L1 pathways prolongs survival of ovarian tumor-bearing mice by prevention of immunosuppression in the tumor microenvironment. FASEB J. (2019) 33:6596–608. doi: 10.1096/fj.201802067RR

PubMed Abstract | Crossref Full Text | Google Scholar

62. Olsen M, Halldórsdóttir HR, Skadborg SK, Pedersen M, Høgdall C, Høgdall E, et al. Improved migration of tumor ascites lymphocytes to ovarian cancer microenvironment by CXCR2 transduction. Oncoimmunology. (2018) 7:e1412029. doi: 10.1080/2162402X.2017.1412029

PubMed Abstract | Crossref Full Text | Google Scholar

63. Kuchroo JR, Sage PT, Liang D, Francisco LM, Buck J, Thaker YR, et al. PD-1 restraint of regulatory T cell suppressive activity is critical for immune tolerance. J Exp Med. (2021) 218:e20182232. doi: 10.1084/jem.20182232

PubMed Abstract | Crossref Full Text | Google Scholar

64. Matsushita H, Shintani D, Kobayashi Y, Fujieda N, Yabuno A, Nishikawa T, et al. Association between effector-type regulatory T cells and immune checkpoint expression on CD8(+) T cells in Malignant ascites from epithelial ovarian cancer. BMC Cancer. (2022) 22:437. doi: 10.1186/s12885-022-09534-z

PubMed Abstract | Crossref Full Text | Google Scholar

65. Siraj AK, Al-Badawi IA, Tulbah A, Al-Dayel F, and Al-Kuraya KS. Differential expression of PD-L1 between primary and metastatic epithelial ovarian cancer and its clinico-pathological correlation. Sci Rep. (2021) 11:3750. doi: 10.1038/s41598-021-83276-z

PubMed Abstract | Crossref Full Text | Google Scholar

66. Madondo MT, McNally OM, Stephens AN, Quinn MA, and Plebanski M. Interleukin 6 present in inflammatory ascites from advanced epithelial ovarian cancer patients promotes tumor necrosis factor receptor 2-expressing regulatory T cells. Front Immunol. (2017) 8:1482. doi: 10.3389/fimmu.2017.01482

PubMed Abstract | Crossref Full Text | Google Scholar

67. Scalzo-Inguanti K, Madondo M, Hallo J, Flanagan K, Quinn M, Plebanski M, et al. Impaired Th1 immunity in ovarian cancer patients is mediated by TNFR2+ Tregs within the tumor microenvironment. Clin Immunol. (2013) 149:97–110. doi: 10.1016/j.clim.2013.07.003

PubMed Abstract | Crossref Full Text | Google Scholar

68. Butterworth J, Mera T, Okubo Y, Wang L, Baum D, Defusco A, et al. Targeting TNFR2 with antagonistic antibodies inhibits proliferation of ovarian cancer cells and tumor-associated Tregs. Sci Signal. (2017) 10:eaaf8608. doi: 10.1126/scisignal.aaf8608

PubMed Abstract | Crossref Full Text | Google Scholar

69. Wu X, Zhou Q, Howard OM, Netea MG, and Oppenheim JJ. TNFR2 is critical for the stabilization of the CD4+Foxp3+ regulatory T. cell phenotype in the inflammatory environment. J Immunol. (2013) 190:1076–84. doi: 10.4049/jimmunol.1202659

PubMed Abstract | Crossref Full Text | Google Scholar

70. de Leve S, Wirsdörfer F, and Jendrossek V. Targeting the immunomodulatory CD73/adenosine system to improve the therapeutic gain of radiotherapy. Front Immunol. (2019) 10:698. doi: 10.3389/fimmu.2019.00698

PubMed Abstract | Crossref Full Text | Google Scholar

71. Xu Y, Chen Y, and Shan S. TIGIT enhances CD4(+) regulatory T-cell response and mediates immune suppression in a murine ovarian cancer model. Cancer Med. (2020) 9:3584–91. doi: 10.1002/cam4.2976

PubMed Abstract | Crossref Full Text | Google Scholar

72. Colombo N, Oza AM, Fujiwara K, Birrer MJ, Randall L, Poddubskaya EV, et al. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN Ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol. (2021) 22:1275–89. doi: 10.1016/S1470-2045(21)00342-9

PubMed Abstract | Crossref Full Text | Google Scholar

73. Fujiwara K, Ledermann JA, Oza AM, Kristeleit R, Ray-Coquard IL, Richardson GE, et al. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN Ovarian 200): an open-label, three-arm, randomised, phase 3 study. Lancet Oncol. (2021) 22:1034–46. doi: 10.1016/S1470-2045(21)00216-3

PubMed Abstract | Crossref Full Text | Google Scholar

74. Shapira-Frommer R, Santin AD, Lisyanskaya AS, Pignata S, Vergote I, Raspagliesi F, et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann Oncol. (2019) 30:1080–7. doi: 10.1093/annonc/mdz135

PubMed Abstract | Crossref Full Text | Google Scholar

75. Partlová S, Sojka L, Hromádková H, Brtnický T, Fučíková J, Kocián P, et al. Dynamics of T-cell infiltration during the course of ovarian cancer: the gradual shift from a Th17 effector cell response to a predominant infiltration by regulatory T-cells. Int J Cancer. (2013) 132:1070–9. doi: 10.1002/ijc.27759

PubMed Abstract | Crossref Full Text | Google Scholar

76. Holfelder P, Klaessens S, Pilotte L, Stroobant V, Lamy J, Naulaerts S, et al. Tryptophan depletion sensitizes the AHR pathway by increasing AHR expression and GCN2/LAT1-mediated kynurenine uptake, and potentiates induction of regulatory T lymphocytes. J Immunother Cancer. (2023) 11:e006728. doi: 10.1136/jitc-2023-006728

PubMed Abstract | Crossref Full Text | Google Scholar

77. Wu M, Liu S, Shang W, Li R, Xu J, Huang L, et al. Glucose metabolism characteristics and TLR8-mediated metabolic control of CD4(+) Treg cells in ovarian cancer cells microenvironment. Cell Death Dis. (2021) 12:22. doi: 10.1038/s41419-020-03272-5

PubMed Abstract | Crossref Full Text | Google Scholar

78. Xu L, Zhou B, Li Q, You D, Liu C, Song H, et al. Transcription factor FOXP3 gene variants affect epithelial ovarian carcinoma in the Han Chinese population. Int J Clin Exp Pathol. (2018) 11:1684–93.

PubMed Abstract | Google Scholar

79. Goel N, Brooke DJ, Katre AA, Londoño AI, Smith HJ, Randall TD, et al. Class I histone deacetylase inhibition promotes CD8 T cell activation in ovarian cancer. Cancer Med. (2021) 10:709–17. doi: 10.1002/cam4.3337

PubMed Abstract | Crossref Full Text | Google Scholar

80. Dou X, Zheng Z, Ye C, Lu TX, Liang HL, Wang L, et al. YTHDF2/m(6) A/NF-κB axis controls anti-tumor immunity by regulating intratumoral Tregs. EMBO J. (2023) 42:e113126. doi: 10.15252/embj.2022113126

PubMed Abstract | Crossref Full Text | Google Scholar

81. Liu G, Jin H, Chen X, He J, Xiao J, Qin Y, et al. The comprehensive expression and functional analysis of m6A modification “readers” in hepatocellular carcinoma. Aging (Albany NY). (2022) 14:6269–98. doi: 10.18632/aging.204217

PubMed Abstract | Crossref Full Text | Google Scholar

82. Schmitt E, Schuler G, Knop J, and Enk AH. Induction of interleukin 10-producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med. (2000) 192:1213–22. doi: 10.1084/jem.192.9.1213

PubMed Abstract | Crossref Full Text | Google Scholar

83. Gregorio J, Wang YH, Ito T, Meller S, Hanabuchi S, Anderson S, et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. (2012) 72:5240–9. doi: 10.1158/0008-5472.CAN-12-2271

PubMed Abstract | Crossref Full Text | Google Scholar

84. Kobayashi A, Toujima S, Shiro M, Mizoguchi M, Mabuchi Y, Yagi S, et al. Indoleamine 2,3-dioxygenase promotes peritoneal metastasis of ovarian cancer by inducing an immunosuppressive environment. Cancer Sci. (2014) 105:966–73. doi: 10.1111/cas.12445

PubMed Abstract | Crossref Full Text | Google Scholar

85. Gamrekelashvili J, Manns MP, Greten TF, and Korangy F. Plasticity of human Th17 cells and iTregs is orchestrated by different subsets of myeloid cells. Blood. (2011) 117:6532–41. doi: 10.1182/blood-2010-11-317321

PubMed Abstract | Crossref Full Text | Google Scholar

86. Ma G, Weber KJ, Ozao-Choy J, Wang G, Yin B, Divino CM, et al. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer Res. (2010) 70:99–108. doi: 10.1158/0008-5472.CAN-09-1882

PubMed Abstract | Crossref Full Text | Google Scholar

87. Yang Y, Yang J, Zhao X, and Wei X. Tumor microenvironment in ovarian cancer: function and therapeutic strategy. Front Cell Dev Biol. (2020) 8:758. doi: 10.3389/fcell.2020.00758

PubMed Abstract | Crossref Full Text | Google Scholar

88. Kieffer Y, Scholer-Dahirel A, Sirven P, Cardon M, Pelon F, Magagna I, et al. miR200-regulated CXCL12β promotes fibroblast heterogeneity and immunosuppression in ovarian cancers. Nat Commun. (2018) 9:1056. doi: 10.1038/s41467-018-03348-z

PubMed Abstract | Crossref Full Text | Google Scholar

89. Yerneni S, Sarkar SN, Orr B, Muthuswamy R, Voyten J, Modugno F, et al. Helicase-driven activation of NFκB-COX2 pathway mediates the immunosuppressive component of dsRNA-driven inflammation in the human tumor microenvironment. Cancer Res. (2018) 78:4292–302. doi: 10.1158/0008-5472.CAN-17-3985

PubMed Abstract | Crossref Full Text | Google Scholar

90. Li X, Wu X, Zhang T, Zhu Q, Wang X, Wang H, et al. Exosomes released from tumor-associated macrophages transfer miRNAs that induce a Treg/Th17 cell imbalance in epithelial ovarian cancer. Cancer Immunol Res. (2018) 6:1578–92. doi: 10.1158/2326-6066.CIR-17-0479

PubMed Abstract | Crossref Full Text | Google Scholar

91. Kryczek I, Edwards RP, Zou L, Szeliga W, Banerjee M, Cost M, et al. Interleukin-2 administration alters the CD4+FOXP3+ T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res. (2007) 67:7487–94. doi: 10.1158/0008-5472.CAN-07-0565

PubMed Abstract | Crossref Full Text | Google Scholar

92. Moss RW, Kleef R, Szasz MA, and Anderson CC. Exploiting autoimmunity unleashed by low-dose immune checkpoint blockade to treat advanced cancer. Scand J Immunol. (2019) 90:e12821. doi: 10.1111/sji.12821

PubMed Abstract | Crossref Full Text | Google Scholar

93. Muzaffar J, Winer I, Rosen SD, Hoimes CJ, Chauhan A, Spreafico A, et al. Nemvaleukin alfa, a modified interleukin-2 cytokine, as monotherapy and with pembrolizumab in patients with advanced solid tumors (ARTISTRY-1). J Immunother Cancer. (2024) 12:e010143. doi: 10.1136/jitc-2024-010143

PubMed Abstract | Crossref Full Text | Google Scholar

94. Maiorano MFP, Lorusso D, and Maiello E. Ovarian cancer in the era of immune checkpoint inhibitors: state of the art and future perspectives. Cancers (Basel). (2021) 13:4438. doi: 10.3390/cancers13174438

PubMed Abstract | Crossref Full Text | Google Scholar

95. Francois A, McGray AR, Miliotto A, and Odunsi K. Compensatory upregulation of PD-1, LAG-3, and CTLA-4 limits the efficacy of single-agent checkpoint blockade in metastatic ovarian cancer. Oncoimmunology. (2017) 6:e1249561. doi: 10.1080/2162402X.2016.1249561

PubMed Abstract | Crossref Full Text | Google Scholar

96. Duraiswamy J, Freeman GJ, and Coukos G. Therapeutic PD-1 pathway blockade augments with other modalities of immunotherapy T-cell function to prevent immune decline in ovarian cancer. Cancer Res. (2013) 73:6900–12. doi: 10.1158/0008-5472.CAN-13-1550

PubMed Abstract | Crossref Full Text | Google Scholar

97. Zhong Q, Yang Z, Li H, Pan C, Huang L, Ni T, et al. Low-dose cyclophosphamide combined with IL-2 inhibits tumor growth by decreasing regulatory T cells and increasing CD8+ T cells and natural killer cells in mice. Immunobiology. (2022) 227:152212. doi: 10.1016/j.imbio.2022.152212

PubMed Abstract | Crossref Full Text | Google Scholar

98. Nicolas A, Cathelin D, Larmonier N, Ferrand C, Foucher P, Fanton A, et al. Increase of CD4+ CD25+ regulatory T cells in the peripheral blood of patients with metastatic carcinoma: a Phase I clinical trial using cyclophosphamide and immunotherapy to eliminate CD4+ CD25+ T lymphocytes. Clin Exp Immunol. (2007) 150:523–30. doi: 10.1111/j.1365-2249.2007.03521.x

PubMed Abstract | Crossref Full Text | Google Scholar

99. Budhu S, Tchaicha J, Weng CH, Gigoux M, Cohen IJ, Redmond D, et al. Blockade of the AHR restricts a Treg-macrophage suppressive axis induced by L-Kynurenine. Nat Commun. (2020) 11:4011. doi: 10.1038/s41467-020-17750-z

PubMed Abstract | Crossref Full Text | Google Scholar

100. Rong X, Zhang F, Mu X, and Jiang J. Cryo-trojan mesenchymal stem cells as non-living tumor-homing supercarriers for enhanced drug delivery and immune activation in prostate cancer. Mater Today Bio. (2025) 32:101650. doi: 10.1016/j.mtbio.2025.101650

PubMed Abstract | Crossref Full Text | Google Scholar

101. Hakak R, Poopak B, and Majd A. Increased IDO expression and regulatory T cells in acute myeloid leukemia: implications for immune escape and therapeutic targeting. Blood Res. (2024) 59:42. doi: 10.1007/s44313-024-00048-0

PubMed Abstract | Crossref Full Text | Google Scholar

102. Flores L, Ngai HW, Cornejo YR, Haber T, McDonald M, Moreira DF, et al. Large, anionic liposomes enable targeted intraperitoneal delivery of a TLR 7/8 agonist to repolarize ovarian tumors’ Microenvironment. Bioconjug Chem. (2021) 32:1581–92. doi: 10.1021/acs.bioconjchem.1c00139

PubMed Abstract | Crossref Full Text | Google Scholar

103. Wild K, Smits M, Zoldan K, Csernalabics B, Flecken T, Lang J, et al. OX40 stimulation and PD-L1 blockade synergistically augment HBV-specific CD4 T cells in patients with HBeAg-negative infection. J Hepatol. (2019) 70:1103–13. doi: 10.1016/j.jhep.2019.02.016

PubMed Abstract | Crossref Full Text | Google Scholar

104. Kang K, Chen P, Zeng Z, Li G, Xiong W, Yi M, et al. Regulatory mechanisms of PD-1/PD-L1 in cancers. Mol Cancer. (2024) 23:108. doi: 10.1186/s12943-024-02023-w

PubMed Abstract | Crossref Full Text | Google Scholar

105. Almazan TH, Hong EK, Khodadoust MS, Arai S, Weng WK, and Kim YH. Potential association of anti-CCR4 antibody mogamulizumab and graft-vs-host disease in patients with mycosis fungoides and sézary syndrome. JAMA Dermatol. (2018) 154:728–30. doi: 10.1001/jamadermatol.2018.0884

PubMed Abstract | Crossref Full Text | Google Scholar

106. Dhar S, Chakraborty D, Pati S, Bose S, Panda AK, Basak U, et al. FOXP3/HAT1 axis controls Treg infiltration in the tumor microenvironment by inducing CCR4 expression in breast cancer. Front Immunol. (2022) 13:740588. doi: 10.3389/fimmu.2022.740588

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: regulatory T cell, ovarian cancer, immunosuppression, metabolic reprogramming, immune checkpoint blockade

Citation: Li J, Huang H, Xie R, Yang R, Wang H and Wan L (2025) Immunosuppressive mechanisms and therapeutic targeting of regulatory T cells in ovarian cancer. Front. Immunol. 16:1631226. doi: 10.3389/fimmu.2025.1631226

Received: 19 May 2025; Accepted: 25 June 2025;
Published: 09 July 2025.

Edited by:

Lilong Zhang, Renmin Hospital of Wuhan University, China

Reviewed by:

Emily Cassar, RMIT University, Australia

Copyright © 2025 Li, Huang, Xie, Yang, Wang and Wan. 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: Li Wan, MTg5NDE2NDQzOTlAMTYzLmNvbQ==

These authors have contributed equally to this work and share first authorship

Disclaimer: 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.