Back to Journals » Journal of Inflammation Research » Volume 18
Inflammation and Immune Escape in Ovarian Cancer: Pathways and Therapeutic Opportunities
Authors Liu C , Yin Q , Wu Z , Li W, Huang J, Chen B, Yang Y, Zheng X, Zeng L, Wang J
Received 29 October 2024
Accepted for publication 6 January 2025
Published 21 January 2025 Volume 2025:18 Pages 895—909
DOI https://doi.org/10.2147/JIR.S503479
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 2
Editor who approved publication: Professor Ning Quan
Chunyan Liu,1,* Qinan Yin,2,3,* Zhaoying Wu,1 Wenhui Li,1 Jun Huang,1 Bo Chen,1 Yanjun Yang,1 Xuewei Zheng,3 Li Zeng,3 Jingjing Wang3
1Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, Beijing, People’s Republic of China; 2Department of Radiation Oncology, First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, People’s Republic of China; 3Precision Medicine Laboratory, School of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Chunyan Liu, Department of Obstetrics and Gynecology, China-Japan Friendship Hospital, No. 2 Yinghua Dongjie, Chaoyang District, Beijing, 100029, People’s Republic of China, Email [email protected]
Abstract: Ovarian cancer (OC) remains one of the most lethal gynecological malignancies, largely due to its late-stage diagnosis and high recurrence rates. Chronic inflammation is a critical driver of OC progression, contributing to immune evasion, tumor growth, and metastasis. Inflammatory cytokines, including IL-6, TNF-α, and IL-8, as well as key signaling pathways such as nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3), are upregulated in OC, promoting a tumor-promoting environment. The tumor microenvironment (TME) is characterized by immune cells like tumor-associated macrophages (TAMs) and regulatory T cells (Tregs), which suppress anti-tumor immune responses, facilitating immune evasion. Furthermore, OC cells utilize immune checkpoint pathways, including PD-1/PD-L1, to inhibit cytotoxic T cell activity. Targeting these inflammatory and immune evasion mechanisms offers promising therapeutic strategies. COX-2 inhibitors, Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway blockers, and NF-kB inhibitors have shown potential in preclinical studies, while immune checkpoint inhibitors targeting PD-1/PD-L1 and CTLA-4 have been explored with mixed results in OC. Additionally, emerging research on the microbiome and inflammation-related biomarkers, such as microRNAs (miRNAs) and exosomes, points to new opportunities for early detection and precision medicine. Future approaches to OC treatment must focus on personalized strategies that target the inflammatory TME, integrating anti-inflammatory therapies with immunotherapy to enhance patient outcomes. Continued research into the interplay between inflammation and immune evasion in OC is essential for developing effective, long-lasting treatments.
Keywords: ovarian cancer, inflammation-driven mechanisms, evasion of immune response, therapeutic strategies
Introduction
Ovarian cancer (OC) is one of the most lethal gynecological malignancies, with a high mortality rate due to its late-stage diagnosis and frequent recurrence. It is one of the most common cancers among women worldwide, and despite advances in surgery and chemotherapy, the overall five-year survival rate remains less than 50%.1,2 Over the last decade, cost-effective strategies for early detection and prevention of OC have been explored. However, OC remains one of the most financially burdensome cancers to treat, with the average initial treatment cost reaching approximately USD 80,000 in the first year and escalating to USD 100,000 or more in the final year of care.3 Most OC patients are diagnosed at an advanced stage, when the disease has already spread beyond the ovaries.4
This delayed diagnosis is primarily due to the asymptomatic nature of early-stage ovarian cancer and the lack of reliable screening methods.5 Currently, CA125 and HE4 are the only approved biomarkers for use in epithelial ovarian cancer (EOC), but they are insufficient for early detection. To address these limitations, multivariate index (MVI) assays, such as the Risk of Malignancy Algorithm (ROMA), have been developed. ROMA integrates menopausal status, CA125, and HE4 levels to improve diagnostic accuracy, particularly during the pre-surgical evaluation of adnexal masses. Additionally, microRNAs (miRNAs) show promising potential in various aspects of EOC prediction. However, further work is needed to establish their characterization and clinical utility as biomarkers.6 Even though treatments such as surgery and platinum-based chemotherapy can initially be effective, most patients eventually develop resistance to treatment, leading to disease recurrence and poor outcomes.7 Among the key mechanisms driving OC progression, the PI3K pathway is frequently upregulated in EOC and plays a pivotal role in chemoresistance and the preservation of genomic stability. This pathway is critical for regulating DNA replication and cell cycle progression. However, inhibition of PI3K can disrupt these processes, leading to genomic instability and mitotic catastrophe. Specifically, reduced activity of the spindle assembly checkpoint protein Aurora kinase B may result in increased occurrence of lagging chromosomes during prometaphase, contributing to mitotic failure and cell death.8 These clinical challenges highlight the urgent need for a deeper understanding of the underlying mechanisms driving OC progression and for the development of new therapeutic strategies.
Chronic inflammation has been increasingly recognized as a key factor in the development and progression of many cancers, including OC.9 Inflammation is a complex biological response to harmful stimuli, such as infection or tissue damage, and is characterized by the activation of immune cells, the release of pro-inflammatory cytokines, and the recruitment of additional immune cells to the site of injury.10 While acute inflammation is a protective response that facilitates tissue repair and resolution of infection, chronic inflammation can lead to tissue damage, genomic instability, and the promotion of cancerous growth.11 The chronic inflammation has been linked to several risk factors, including endometriosis, pelvic inflammatory disease, and obesity in OC.12 These conditions create a pro-inflammatory environment that promotes tumorigenesis through the sustained activation of inflammatory pathways and immune cell infiltration into the tumor microenvironment (TME).13
The TME in OC is a complex and dynamic system that plays a critical role in disease progression. It is composed of cancer cells, stromal cells, immune cells, and various soluble factors such as cytokines and chemokines.14 Chronic inflammation within the TME creates conditions that promote the survival, proliferation, and dissemination of cancer cells.11 Pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interleukin-8 (IL-8) are frequently upregulated in OC and have been shown to promote tumor growth and metastasis.15 These cytokines activate key signaling pathways such as the nuclear factor kappa B (NF-kB) and signal transducer and activator of transcription 3 (STAT3) pathways, which are known to drive cell survival, proliferation, and resistance to apoptosis.16,17 Moreover, chronic inflammation can lead to the recruitment of TAMs and other immunosuppressive cell types, which further exacerbate tumor progression by promoting immune evasion.18
One of the hallmark features of cancer is its ability to evade the immune system, and chronic inflammation plays a pivotal role in facilitating this immune evasion.13 In OC, tumor cells exploit the inflammatory microenvironment to escape immune surveillance by downregulating the expression of major histocompatibility complex (MHC) molecules, which are critical for antigen presentation to T cells.19 This prevents the immune system from recognizing and eliminating cancer cells.19 In addition, OC cells often overexpress immune checkpoint molecules such as programmed death-ligand 1 (PD-L1), which binds to programmed death-1 (PD-1) receptors on T cells, leading to the suppression of T cell activity and the inhibition of anti-tumor immune responses.20 This immune checkpoint pathway has become a major target for cancer immunotherapy, and several clinical trials are currently investigating the efficacy of PD-1/PD-L1 inhibitors in OC.21
Another important mechanism by which inflammation promotes immune evasion in OC is through the recruitment of immunosuppressive cells such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs).22 These cells secrete anti-inflammatory cytokines such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), which suppress the activation and function of cytotoxic T cells.23 This creates an immunosuppressive environment that allows OC cells to thrive and metastasize. The presence of high levels of Tregs and MDSCs in OC has been associated with poor prognosis and resistance to immunotherapy.24 Therefore, targeting these immunosuppressive cells and their associated signaling pathways may represent a promising therapeutic strategy for overcoming immune evasion in OC.
Given the critical role of inflammation in OC progression and immune evasion, there has been growing interest in developing anti-inflammatory therapies as potential treatments for this disease. Several preclinical and clinical studies have investigated the use of nonsteroidal anti-inflammatory drugs (NSAIDs), cyclooxygenase-2 (COX-2) inhibitors, and corticosteroids in OC.25–27 COX-2 is an enzyme that is upregulated in many cancers, including OC, and is involved in the production of pro-inflammatory prostaglandins that promote tumor growth and metastasis.28 COX-2 inhibitors have shown promise in reducing tumor growth and enhancing the efficacy of chemotherapy in preclinical models of OC.29 However, the clinical benefits of these agents in OC patients have been limited, and further research is needed to determine their potential as part of combination therapy regimens.30
In addition to traditional anti-inflammatory drugs, targeted therapies that inhibit key inflammatory signaling pathways such as NF-kB and STAT3 are also being explored. These pathways are frequently dysregulated in OC and play a central role in mediating the effects of chronic inflammation on tumor growth and immune evasion.31 Inhibitors of these pathways have shown efficacy in preclinical models, but their clinical utility in OC remains to be fully established. Furthermore, the combination of anti-inflammatory therapies with immunotherapies, such as immune checkpoint inhibitors, represents a promising approach for enhancing anti-tumor immune responses and improving patient outcomes.
Recent studies have highlighted the pivotal role of the microbiome in ovarian cancer, demonstrating that oncobiosis—characterized by microbial dysbiosis and increased inflammatory potential—may contribute to ovarian cancer initiation and progression.32 Additionally, the cervicovaginal and gut microbiota have shown potential as biomarkers and therapeutic targets, opening new avenues for early diagnosis and innovative treatment strategies.33 However, significant gaps remain in understanding the causal relationships between microbiome dysbiosis and ovarian cancer, the precise mechanisms driving inflammation-mediated carcinogenesis, and the validation of microbiome-based diagnostics and therapies in large-scale clinical settings.
Here, this review focuses on exploring the intricate inflammatory pathways and immune evasion mechanisms involved in OC. We also highlight potential therapeutic targets and relevant biomarkers that could lead to improved treatments and outcomes for patients.
Overview of OC Subtypes
OC is not a single disease but a group of tumors that originate from different tissues within the ovary, each with its distinct biology, prognosis, and treatment approaches. The three major subtypes of OC are epithelial ovarian cancer (EOC), germ cell tumors, and stromal tumors.34
EOC is by far the most common subtype, accounting for more than 90% of OC cases.4 Among the EOC subtypes, high-grade serous ovarian carcinoma (HGSOC) is the most aggressive and prevalent.35 Genomic analysis, such as The Cancer Genome Atlas (TCGA), has revealed that HGSOC is characterized by widespread TP53 mutations and chromosomal instability, which contribute to rapid disease progression and resistance to therapy.36 These genetic alterations, particularly mutations in BRCA1/2, are also associated with defects in DNA repair mechanisms, such as homologous recombination deficiency, making these tumors initially sensitive to platinum-based chemotherapy.37 BRCA1/2 germline mutations, which are found in 6–15% of women diagnosed with EOC, are the strongest known genetic risk factors for the disease. Importantly, BRCA1/2 carriers exhibit better responses to platinum-based chemotherapies compared to non-carriers, resulting in improved survival outcomes despite often being diagnosed at a later stage and higher grade.38 However, resistance to chemotherapy often develops over time, leading to treatment failure and poor survival outcomes.
Germ cell tumors account for less than 5% of OC cases and typically affect younger women. These tumors arise from the reproductive cells of the ovary and include types such as dysgerminomas and teratomas.39 Studies demonstrated that germ cell tumors are highly responsive to platinum-based chemotherapy, with excellent survival rates even in advanced cases.40 Although ovarian germ cell tumors are uncommon in postmenopausal women, they should be considered in patients presenting with an ovarian mass and elevated serum alpha-fetoprotein (AFP) levels. Recognizing this diagnostic challenge is critical for ensuring timely and accurate diagnosis in this unique patient population.41 The germ cell tumours are also characterized by faster rate of growth, unilateral localisation (95% of cases), and early-stage disease (60–70%).42 Additionally, studies also underscored the importance of early detection and appropriate chemotherapy regimens for this subtype, which can achieve cure rates exceeding 90% for localized tumors.39
Stromal tumors, though rare, originate from the connective tissue within the ovary.43 These tumors include granulosa cell tumors, which are often hormonally active and can lead to symptoms like abnormal uterine bleeding due to estrogen production.44 It has been emphasized the need for long-term monitoring of patients with stromal tumors, as these tumors tend to recur years after initial treatment.45 Although stromal tumors generally have a better prognosis than epithelial subtypes, the risk of late recurrence warrants prolonged surveillance. In assessing stromal tumors, miRNAs such as miR-202-3p and miR-513c-5p are significantly higher compared to germ cell tumors, with miR-202-3p being proposed as a specific marker for sex-cord stromal tumors. This miRNA is expressed in Sertoli and granulosa cells and is correlated with the expression of the transcription factor FOXL2. Furthermore, studies have identified recurrent somatic mutations in FOXL2 as a hallmark of granulosa cell tumors of the ovary, reinforcing its diagnostic and prognostic significance.46
Pathophysiology of OC and Inflammation-Driven Mechanisms
TME in OC
The TME plays an essential role in OC development and progression. It is a dynamic system comprising cancer cells, immune cells, stromal cells, endothelial cells, and extracellular matrix (ECM) components. This complex network not only supports tumor growth but also shapes the immune response, promoting immune evasion and metastasis.47
One of the most significant components of the TME in OC is the TAM. TAMs are immune cells that are recruited to the tumor site, where they often adopt a pro-tumorigenic M2-like phenotype. These cells are central to promoting tumor growth, angiogenesis, and immunosuppression.48 A study by Hagemann et al demonstrated that high levels of TAMs in OC tissues correlate with poor patient prognosis.49 TAMs secrete a range of cytokines, such as IL-6 and TNF-α, which enhance tumor cell proliferation and survival, while also facilitating the remodeling of the ECM to support metastasis.50
T cells are also critical components of the immune landscape in OC. While cytotoxic T cells can directly kill tumor cells and are generally associated with improved patient outcomes, their activity is often suppressed within the TME. This suppression is largely mediated by Tregs, which dampen the anti-tumor immune response. Curiel et al found that the recruitment of Tregs to ovarian tumors was associated with reduced survival rates.51 The presence of Tregs creates an immunosuppressive microenvironment that inhibits the activation of cytotoxic T cells, allowing the tumor to evade immune surveillance.52 On the other hand, a high infiltration of cytotoxic T cells is linked to better outcomes, as these cells can directly target and kill cancer cells.53
Natural killer (NK) cells also play a role in the early immune response to tumors. However, NK cell function is often inhibited by factors secreted by the tumor, which downregulate activating receptors on NK cells in OC.54 This suppression of NK cell activity allows tumor cells to evade early immune detection and continue growing unchecked.
Inflammation and Pathways in OC
Chronic inflammation is a well-established driver of cancer progression, and OC is no exception. A variety of inflammatory mediators, including cytokines and chemokines, are upregulated in the OC microenvironment, promoting tumor growth, immune evasion, and metastasis.
IL-6
IL-6 is one of the most studied cytokines in OC and plays a crucial role in activating the STAT3 pathway. IL-6 is elevated in the OC microenvironment and drives the expression of genes involved in cancer cell survival, proliferation, and immune evasion.55–57 It has been shown that high levels of IL-6 in OC patients were associated with poor survival, suggesting that IL-6 could be a valuable therapeutic target.58 IL-6 stimulates the activation of STAT3, which in turn promotes the expression of anti-apoptotic and pro-survival genes, further supporting tumor growth and resistance to chemotherapy.9 The STAT3 pathway is frequently activated in OC, primarily through IL-6 signaling.59 Persistent activation of STAT3 promotes tumor growth, immune suppression, and the recruitment of immunosuppressive cells such as TAMs and Tregs.60 It has been demonstrated that blocking STAT3 activation significantly reduced tumor growth and enhanced anti-tumor immune responses in OC models.61 Targeting STAT3 has emerged as a promising therapeutic approach, particularly in combination with other treatments that modulate the immune response.
TNF-α
TNF-α is another key pro-inflammatory cytokine in OC. It has been demonstrated that TNF-α is essential for creating a pro-inflammatory microenvironment that supports tumor growth and metastasis.62 Inhibiting TNF-α has been shown to reduce tumor growth in preclinical models, making it a potential target for therapeutic intervention.63 It is well known that TNF- α contributes to tumor progression by activating the NF-kB pathway. NF-kB is a transcription factor that regulates the expression of genes involved in inflammation, cell survival, and angiogenesis. TNF-α induces the activation of NF-kB, which promotes tumor cell survival and proliferation.64 In OC, chronic activation of NF-kB promotes tumor cell survival and resistance to apoptosis, which contributes to the aggressiveness of the disease. Recent study showed that NF-kB activation in OC cells is associated with chemotherapy resistance, particularly to platinum-based agents like cisplatin.65 This finding highlights the potential of targeting NF-kB as a strategy to overcome drug resistance in OC.
IL-8
IL-8 plays a key role in promoting angiogenesis and metastasis in OC. IL-8 is upregulated in response to hypoxic conditions within the TME, and its expression is associated with increased vascularization and tumor spread.66 Studies found that elevated levels of IL-8 in OC patients were correlated with poor outcomes, highlighting the potential of targeting IL-8 in anti-cancer therapies.63,67–69
CXCL12
Chemokines such as CXCL12 and their receptors are also critical in regulating immune cell migration and metastasis. The CXCL12/CXCR4 axis is particularly important in OC, where it promotes the migration of cancer cells to the omentum, a common metastatic site.70 A report highlighted the importance of targeting the CXCL12/CXCR4 signaling axis to prevent OC metastasis.71
COX-2 and Prostaglandins
Chronic inflammation plays a pivotal role in the development and progression of OC, and the enzyme COX-2 is central to this process. COX-2 is overexpressed in many ovarian cancers, where it catalyzes the production of prostaglandin E2 (PGE2), a key mediator of inflammation.72 PGE2 promotes the recruitment of Tregs and TAMs to the tumor microenvironment, both of which contribute to immune evasion.73
In addition to its role in immune suppression, PGE2 has been implicated in promoting angiogenesis, the process by which tumors develop new blood vessels to support their growth.74 By inhibiting the activity of NK cells and cytotoxic T cells, PGE2 creates an immunosuppressive environment that allows OC cells to evade immune detection and destruction.75
COX-2 inhibitors, such as NSAIDs, are being explored as potential adjuvants in OC therapy76 These inhibitors have shown promise in preclinical studies by reducing the production of PGE2, thereby disrupting the immunosuppressive tumor environment and enhancing anti-tumor immunity.77 Inflammatory mediators and their roles in OC progression has been summarized in Table 1.
![]() |
Table 1 Inflammatory Mediators and Their Roles in OC Progression |
Chronic Inflammation Driving Genomic Instability in OC
Chronic inflammation not only promotes immune evasion but also contributes to genomic instability, which drives the progression of OC. Inflammatory cytokines such as IL-6, TNF-α, and IL-1β induce the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) within the tumor microenvironment.78,79 These reactive molecules cause oxidative stress, which leads to DNA damage and increases the likelihood of mutations.78,79
Over time, this chronic inflammation-driven DNA damage fosters genomic instability, which is a hallmark of aggressive tumor behavior. Tumors with high levels of genomic instability tend to exhibit more mutations, making them more resistant to standard therapies such as chemotherapy. Furthermore, inflammation-induced genomic instability contributes to tumor heterogeneity, making it more difficult to target the tumor with precision therapies.
Key inflammatory pathways, such as the NF-kB and STAT3 signaling pathways, play central roles in mediating the effects of chronic inflammation in OC. Targeting NF-kB and STAT3 with specific inhibitors has shown promise in preclinical studies as a strategy to reduce chronic inflammation, limit DNA damage, and improve therapeutic outcomes in OC.31
Immune Evasion in OC
OC is notorious for its ability to evade immune detection and leverage chronic inflammation to support its growth, survival, and progression. Immune evasion allows tumor cells to escape immune surveillance, while inflammation provides a microenvironment conducive to malignancy. This combination is one of the critical reasons for the high mortality rate associated with OC. By understanding the mechanisms involved, researchers and clinicians can identify therapeutic targets and develop strategies to overcome inflammation and immune suppression.
TAMs and Their Pro-Tumor Role
TAMs play a critical role in shaping the immune landscape of OC. TAMs typically exhibit an M2-like, pro-tumorigenic phenotype. These macrophages promote tissue remodeling, angiogenesis, and immune suppression. Instead of attacking cancer cells, M2-like TAMs secrete cytokines such as IL-6 and TNF-α, which facilitate tumor growth by promoting angiogenesis and creating an immunosuppressive microenvironment.18
In OC, TAMs are commonly recruited in large numbers, and their presence correlates with poor prognosis.80 Their role in suppressing anti-tumor immune responses is significant, as they hinder the activation and function of cytotoxic T cells and NK cells, which are essential for identifying and eliminating cancer cells. Moreover, TAMs release immunosuppressive molecules like TGF-β and IL-10, which dampen the immune response, allowing the tumor to evade immune detection.81
Efforts to target TAMs as part of cancer therapy have gained momentum in recent years. One approach is to block the recruitment of TAMs to the TME. Another strategy is to reprogram TAMs from a tumor-promoting M2 phenotype to a tumor-suppressing M1 phenotype, which supports anti-tumor immune responses and inhibits tumor growth.82
Tregs and Immune Suppression
Tregs are another critical player in the immune evasion strategies of OC. Tregs function to maintain immune homeostasis by preventing excessive immune responses, but in the context of cancer, they suppress anti-tumor immunity. In OC, tumors actively recruit Tregs to the TME, where they suppress the activity of effector T cells, including cytotoxic T cells, which are responsible for attacking and killing tumor cells.83
High levels of Tregs within the OC microenvironment are associated with poor patient outcomes, as they hinder the ability of the immune system to mount an effective anti-tumor response.84 Tregs suppress immune function through the production of anti-inflammatory cytokines such as IL-10 and TGF-β, which inhibit the proliferation and activity of cytotoxic T cells.85 They also express immune checkpoint molecules such as CTLA-4, which blocks the activation of effector T cells, further contributing to immune suppression.86
Given their crucial role in immune evasion, strategies targeting Tregs have been explored as potential cancer therapies. For instance, depleting Tregs or inhibiting their suppressive functions may restore anti-tumor immune responses and enhance the efficacy of immunotherapies like immune checkpoint inhibitors.87
Tumor Cell Downregulation of MHC Molecules to Evade Antigen Presentation
One of the hallmark mechanisms by which OC cells evade immune detection is by downregulating major histocompatibility complex (MHC) molecules, particularly MHC class I. MHC class I molecules are critical for presenting tumor antigens to cytotoxic T cells, allowing them to recognize and eliminate cancer cells.88 Without adequate MHC class I expression, cancer cells become invisible to cytotoxic T cells, preventing immune-mediated destruction.89
Additionally, OC cells frequently overexpress immune checkpoint proteins such as PD-L1, which binds to the PD-1 receptor on T cells, further inhibiting their function and promoting immune evasion.20 The combination of reduced antigen presentation through MHC downregulation and increased immune suppression via checkpoint pathways allows OC to thrive in the presence of an otherwise functional immune system.
Efforts to restore MHC class I expression and block immune checkpoints have shown promise in preclinical and clinical studies. Immune checkpoint inhibitors, such as anti-PD-1 and anti-PD-L1 therapies, have demonstrated efficacy in enhancing T cell function and improving immune-mediated tumor clearance.90 Recent therapeutic strategies have explored combining PARP inhibitors with immunotherapies, such as anti-CTLA-4 and PD-1/PD-L1, particularly in BRCA1/2-mutated or homologous recombination (HR)-deficient tumors. These tumors display a higher neo-antigen load compared to HR-proficient cancers, leading to more robust anti-tumor immune responses. Additionally, BRCA deficiency has been shown to activate a STING-dependent innate immune response, inducing type I interferons and pro-inflammatory cytokines, which further enhances immune-mediated tumor suppression. Clinical models have also demonstrated that PARP inhibition can inactivate GSK3 and upregulate PD-L1 in a dose-dependent manner, suppressing T-cell activation and promoting enhanced cancer cell apoptosis. These findings highlight the potential of combining PARP inhibitors with immune checkpoint inhibitors to optimize therapeutic outcomes in ovarian cancer.91 Immune Evasion Mechanisms in OC is summarized in Table 2.
![]() |
Table 2 Immune Evasion Mechanisms in OC |
Diagnostic and Prognostic Biomarkers
Inflammatory Markers as Diagnostic Tools
The detection of systemic inflammation is a common feature in OC patients, and several inflammatory markers have been proposed as potential diagnostic and prognostic tools. C-reactive protein (CRP), a well-established acute-phase protein, has gained attention due to its role in inflammation. CRP levels increase in response to pro-inflammatory cytokines, particularly IL-6, and can reflect tumor burden and the extent of inflammation in OC patients.92 Numerous studies have linked elevated CRP levels to poor prognosis, increased tumor stage, and the presence of metastasis in OC patients. In addition to CRP, IL-6 serves as both a diagnostic and prognostic biomarker. Elevated serum levels of IL-6 correlate with advanced-stage OC and poor survival rates.93
The relationship between chronic inflammation and OC has also led to the use of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) as prognostic indicators. A high NLR is indicative of an inflammatory response driven by neutrophils, which are recruited to the tumor site, while a decrease in lymphocytes signals immune suppression. Several studies have demonstrated that a high NLR and PLR are associated with decreased survival in OC patients.94
Role of Immune Checkpoint Molecules in Prognosis
Immune checkpoint molecules, such as PD-1 and PD-L1, are central to immune evasion in OC. The interaction between PD-1, expressed on T cells, and its ligand PD-L1, expressed on tumor cells and immune cells within the TME, inhibits T cell activation and allows the tumor to evade immune detection. PD-L1 expression has been linked to poor prognosis in OC, with higher levels of PD-L1 expression correlating with shorter overall survival and progression-free survival.20 Furthermore, tumors with high PD-L1 expression tend to have a more immunosuppressive microenvironment, characterized by increased Treg infiltration, and reduced cytotoxic T cells activity.95,96
The prognostic value of PD-L1 expression has led to the exploration of immune checkpoint inhibitors, such as anti-PD-1/PD-L1 antibodies, as therapeutic agents. The response to checkpoint inhibitors appears to be better in tumors with higher PD-L1 expression and higher levels of TILs in OC patients, which suggests that these markers could be useful in predicting treatment response.95,97
Molecular Markers for Treatment Response
Biomarkers Predicting Response to Immunotherapy
With the advent of immunotherapy, including immune checkpoint inhibitors and adoptive T cell therapy, the search for biomarkers that predict treatment response has intensified. Tumor mutational burden (TMB) has emerged as a promising biomarker for predicting response to immunotherapy in several cancers, including OC.98 High TMB is associated with an increased number of neoantigens, which makes the tumor more recognizable by the immune system.99 This, in turn, enhances the efficacy of immunotherapies that rely on reactivating the immune system, such as anti-PD-1/PD-L1 antibodies.
Another crucial biomarker is the presence of TILs. OCs with a higher density of TILs tend to respond better to immunotherapy because TILs represent an active anti-tumor immune response. Studies have shown that the presence of CD8+ T cells within the tumor correlates with improved survival and a better response to immunotherapy.100 Conversely, tumors with low levels of TILs or high levels of immunosuppressive cells, such as Tregs and TAMs, are often resistant to immunotherapy.
Role of Inflammation-Related miRNAs and Exosomes in Diagnosis and Prognosis
In recent years, miRNAs and exosomes have emerged as promising biomarkers in OC. MiRNAs are small non-coding RNAs that regulate gene expression and are involved in numerous cellular processes, including inflammation, immune response, and cancer progression.101 Exosomal miR-21-5p promotes ovarian cancer progression by enhancing cell proliferation, migration, and invasion, while inhibiting apoptosis through the regulation of CDK6, making it a potential therapeutic target.102 ROS-induced downregulation of exosomal miR-155-5p promotes tumor growth by enhancing immunosuppressive macrophage activity and PD-L1 expression, while targeting miR-155-5p restores anti-tumor immunity, offering a novel therapeutic strategy for ovarian cancer.103 MiR-146a has also been found to inhibit ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8+ T cell infiltration.104
Exosomes, which are small vesicles secreted by cells, play a significant role in cell-cell communication within the TME. Tumor-derived exosomes carry proteins, lipids, and nucleic acids, including miRNAs, that influence immune cell function and promote tumor progression.105 Exosomes have been shown to modulate the immune system by suppressing T cell activity, promoting the recruitment of TAMs, and enhancing the immunosuppressive properties of Tregs.106 Inflammation-related exosomal miRNAs, such as those involved in the NF-kB and STAT3 pathways, have been explored as diagnostic and prognostic biomarkers in OC.107 The ability to isolate and analyze exosomal miRNAs from blood samples offers a non-invasive method for early detection and monitoring of OC.
The potential of miRNAs and exosomes as biomarkers extends beyond diagnosis. They are also being investigated as predictors of treatment response, particularly in the context of chemotherapy and immunotherapy. For instance, patients with high levels of specific miRNAs, such as miR-21, have been found to be resistant to platinum-based chemotherapy, which is the standard treatment for OC.108 Similarly, exosomal PD-L1 levels are being explored as a potential biomarker for predicting response to immune checkpoint inhibitors.109
Inflammation-associated biomarkers are essential tools for improving the diagnosis, prognosis, and treatment of OC. Markers such as CRP, IL-6, PD-L1, and inflammation-related miRNAs provide valuable insights into the disease’s progression and its response to therapy. As our understanding of the role of inflammation in OC deepens, these biomarkers will likely play an increasingly important role in guiding personalized treatment approaches. The development of non-invasive methods for detecting biomarkers, such as exosomal miRNAs, will also enhance early detection and monitoring, ultimately improving outcomes for OC patients. Inflammation-associated biomarkers in OC are summarized in Table 3. The chronic inflammation, immune response, tumor growth, and metastasis in OC is illustrated in Figure 1.
![]() |
Table 3 Inflammation-Associated Biomarkers in OC |
Current and Emerging Therapeutic Strategies Targeting Inflammation in OC
Current therapeutic strategies in OC target inflammation, a key driver of cancer progression, immune evasion, and metastasis. Anti-inflammatory treatments, such as COX-2 inhibitors, block prostaglandin synthesis to reduce immune suppression and enhance chemotherapy. Targeting key inflammatory pathways like JAK/STAT and NF-kB also holds potential, with inhibitors disrupting tumor growth and survival. Immunotherapies, including immune checkpoint inhibitors (PD-1, CTLA-4) and CAR-T cell therapy, show promise but are often limited by the tumor’s immunosuppressive environment. Emerging combination therapies integrating anti-inflammatory agents, chemotherapy, and immunotherapy aim to enhance patient outcomes. Lifestyle interventions, including diet and exercise, are also explored to reduce inflammation and improve prognosis.
Challenges and Future Directions in Targeting Inflammation in OC
OC presents significant challenges due to the dual role of inflammation in both promoting and inhibiting tumor progression. Chronic inflammation within the TME can enhance immune evasion, angiogenesis, and metastasis through the actions of cytokines like IL-6 and TNF-α, while also activating immune responses such as cytotoxic T cells that target cancer cells. Personalized therapies that disrupt tumor-promoting inflammation while enhancing anti-tumor immunity are critical. Current anti-inflammatory therapies face limitations, including toxicity and resistance. Targeting inflammatory pathways like NF-kB and JAK/STAT with inhibitors can suppress immune responses and lead to complications such as infections. Emerging areas of research focus on the role of the microbiome in regulating inflammation, which could lead to novel therapies such as probiotics to modulate the TME. Additionally, biomarkers like circulating miRNAs show promise for early detection of OC and tailoring treatments to individual patients. Long-term research is necessary to understand the full impact of immunotherapy on inflammatory pathways, as these therapies can modulate inflammation, potentially influencing recurrence, and resistance.
Conclusion
In OC, inflammatory pathways such as NF-kB, JAK/STAT, and COX-2 play a central role in tumor progression, immune evasion, and resistance to therapy. Targeting these pathways has significant therapeutic implications, as it allows for the reduction of inflammation-driven tumor growth while potentially enhancing the effectiveness of immunotherapy and chemotherapy. COX-2 inhibitors, JAK/STAT pathway blockers, and NF-kB inhibitors represent key areas where anti-inflammatory treatments may provide benefit, especially when combined with immune checkpoint inhibitors. However, these strategies are not without challenges, as toxicity and resistance can limit their effectiveness.
Integrating findings on inflammation-driven pathways and immune evasion mechanisms into clinical practice holds great potential for transforming OC management.110 For instance, targeting key pathways such as NF-κB and JAK/STAT through inhibitors has demonstrated preclinical efficacy in reducing tumor-promoting inflammation and overcoming chemoresistance.111 Additionally, combining these approaches with immune checkpoint inhibitors (eg, PD-1/PD-L1 blockade) can synergistically enhance anti-tumor immune responses.112 Recent advancements in microbiome research further provide novel therapeutic opportunities, with strategies like microbiota modulation showing promise in reshaping the tumor microenvironment. Future research should focus on translating these findings into personalized treatment regimens and conducting large-scale clinical trials to validate these integrated approaches.113 Such efforts could significantly improve patient survival and quality of life, paving the way for more effective and durable therapeutic strategies.
The future of OC therapy lies in continued research into the complex interplay between inflammation and tumor biology. Investigating the role of the microbiome, novel biomarkers, and emerging combination therapies is essential to improving patient outcomes. Moreover, developing personalized approaches that target inflammation while preserving immune responses will be critical in integrating these therapies into clinical practice.
The integration of inflammation-targeting therapies into standard treatment regimens offers the potential to enhance survival and quality of life for OC patients. Continued innovation and clinical trials are crucial to refining these treatments and achieving long-term success.
Author Contributions
All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas. They took part in drafting, revising, or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.
Funding
There is no funding to report.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Coburn SB, Bray F, Sherman ME, Trabert B. International patterns and trends in ovarian cancer incidence, overall and by histologic subtype. Int J Cancer. 2017;140(11):2451–2460. doi:10.1002/ijc.30676
2. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136(5):E359–86. doi:10.1002/ijc.29210
3. Ghose A, Bolina A, Mahajan I, et al. Hereditary ovarian cancer: towards a cost-effective prevention strategy. Int J Environ Res Public Health. 2022;19(19). doi:10.3390/ijerph191912057
4. Arora T, Mullangi S, Vadakekut ES, Lekkala MR. Epithelial ovarian cancer. StatPearls; 2024.
5. Xiao L, Li H, Jin Y. Automated early ovarian cancer detection system based on bioinformatics. Sci Rep. 2024;14(1):22887. doi:10.1038/s41598-024-71863-9
6. Ghose A, McCann L, Makker S, et al. Diagnostic biomarkers in ovarian cancer: advances beyond CA125 and HE4. Ther Adv Med Oncol. 2024;16:17588359241233225. doi:10.1177/17588359241233225
7. Dragomir R, Sas I, Saftescu S, et al. Treatment experience and predictive factors associated with response in platinum-resistant recurrent ovarian cancer: a retrospective single-institution study. J Clin Med. 2021;10(16). doi:10.3390/jcm10163596
8. Aliyuda F, Moschetta M, Ghose A, et al. Advances in ovarian cancer treatment beyond PARP inhibitors. Curr Cancer Drug Targets. 2023;23(6):433–446. doi:10.2174/1568009623666230209121732
9. Savant SS, Sriramkumar S, O’Hagan HM. The role of inflammation and inflammatory mediators in the development, progression, metastasis, and chemoresistance of epithelial ovarian cancer. Cancers. 2018;10(8). doi:10.3390/cancers10080251
10. Chen L, Deng H, Cui H, et al. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget. 2018;9(6):7204–7218. doi:10.18632/oncotarget.23208
11. Zhao H, Wu L, Yan G, et al. Inflammation and tumor progression: signaling pathways and targeted intervention. Signal Transduct Target Ther. 2021;6(1):263. doi:10.1038/s41392-021-00658-5
12. Huang JY, Yang SF, Wu PJ, Wang CH, Tang CH, Wang PH. Different influences of endometriosis and pelvic inflammatory disease on the occurrence of ovarian cancer. Int J Environ Res Public Health. 2021;18(16). doi:10.3390/ijerph18168754
13. Hu A, Sun L, Lin H, Liao Y, Yang H, Mao Y. Harnessing innate immune pathways for therapeutic advancement in cancer. Signal Transduct Target Ther. 2024;9(1):68. doi:10.1038/s41392-024-01765-9
14. Baghban R, Roshangar L, Jahanban-Esfahlan R, et al. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun Signal. 2020;18(1):59. doi:10.1186/s12964-020-0530-4
15. Pawlik W, Pawlik J, Kozlowski M, et al. The clinical importance of IL-6, IL-8, and TNF-alpha in patients with ovarian carcinoma and benign cystic lesions. Diagnostics. 2021;11(9). doi:10.3390/diagnostics11091625
16. Devanaboyina M, Kaur J, Whiteley E, et al. NF-kappaB signaling in tumor pathways focusing on breast and ovarian cancer. Oncol Rev. 2022;16:10568. doi:10.3389/or.2022.10568
17. Tosic I, Frank DA. STAT3 as a mediator of oncogenic cellular metabolism: pathogenic and therapeutic implications. Neoplasia. 2021;23(12):1167–1178. doi:10.1016/j.neo.2021.10.003
18. Huang R, Kang T, Chen S. The role of tumor-associated macrophages in tumor immune evasion. J Cancer Res Clin Oncol. 2024;150(5):238. doi:10.1007/s00432-024-05777-4
19. Rodriguez GM, Yakubovich E, Vanderhyden BC. Unveiling the immunogenicity of ovarian tumors as the crucial catalyst for therapeutic success. Cancers. 2023;15(23). doi:10.3390/cancers15235694
20. Dumitru A, Dobrica EC, Croitoru A, Cretoiu SM, Gaspar BS. Focus on PD-1/PD-L1 as a therapeutic target in ovarian cancer. Int J Mol Sci. 2022;23(20). doi:10.3390/ijms232012067
21. Chen Y, Liu X, Hu Y, Xia L. Efficacy and safety of PD-1/PD-L1 immune checkpoint inhibitors in the treatment of recurrent ovarian cancer: a systematic review and meta-analysis. Medicine. 2024;103(18):e38019. doi:10.1097/MD.0000000000038019
22. Haist M, Stege H, Grabbe S, Bros M. The functional crosstalk between myeloid-derived suppressor cells and regulatory T cells within the immunosuppressive tumor microenvironment. Cancers. 2021;13(2). doi:10.3390/cancers13020210
23. Sanjabi S, Zenewicz LA, Kamanaka M, Flavell RA. Anti-inflammatory and pro-inflammatory roles of TGF-beta, IL-10, and IL-22 in immunity and autoimmunity. Curr Opin Pharmacol. 2009;9(4):447–453. doi:10.1016/j.coph.2009.04.008
24. Okla K. Myeloid-derived suppressor cells (MDSCs) in ovarian cancer-looking back and forward. Cells. 2023;12(14). doi:10.3390/cells12141912
25. Meric JB, Rottey S, Olaussen K, et al. Cyclooxygenase-2 as a target for anticancer drug development. Crit Rev Oncol Hematol. 2006;59(1):51–64. doi:10.1016/j.critrevonc.2006.01.003
26. Prizment AE, Folsom AR, Anderson KE. Nonsteroidal anti-inflammatory drugs and risk for ovarian and endometrial cancers in the Iowa Women’s Health Study. Cancer Epidemiol Biomarkers Prev. 2010;19(2):435–442. doi:10.1158/1055-9965.EPI-09-0976
27. Sun H, Zhang X, Sun D, et al. COX-2 expression in ovarian cancer: an updated meta-analysis. Oncotarget. 2017;8(50):88152–88162. doi:10.18632/oncotarget.21538
28. Desai SJ, Prickril B, Rasooly A. Mechanisms of phytonutrient modulation of cyclooxygenase-2 (COX-2) and inflammation related to cancer. Nutr Cancer. 2018;70(3):350–375. doi:10.1080/01635581.2018.1446091
29. Suri A, Sheng X, Schuler KM, et al. The effect of celecoxib on tumor growth in ovarian cancer cells and a genetically engineered mouse model of serous ovarian cancer. Oncotarget. 2016;7(26):39582–39594. doi:10.18632/oncotarget.8659
30. Lee JM, Minasian L, Kohn EC. New strategies in ovarian cancer treatment. Cancer. 2019;125(Suppl 24):4623–4629. doi:10.1002/cncr.32544
31. Wu CJ, Sundararajan V, Sheu BC, Huang RY, Wei LH. Activation of STAT3 and STAT5 signaling in epithelial ovarian cancer progression: mechanism and therapeutic opportunity. Cancers. 2019;12(1). doi:10.3390/cancers12010024
32. Choi SY, Choi JH. Ovarian cancer and the microbiome: connecting the dots for early diagnosis and therapeutic innovations—a review. Medicina. 2024;60(3). doi:10.3390/medicina60030516
33. Sipos A, Ujlaki G, Miko E, et al. The role of the microbiome in ovarian cancer: mechanistic insights into oncobiosis and to bacterial metabolite signaling. Mol Med. 2021;27(1):33. doi:10.1186/s10020-021-00295-2
34. Reid BM, Permuth JB, Sellers TA. Epidemiology of ovarian cancer: a review. Cancer Biol Med. 2017;14(1):9–32. doi:10.20892/j.issn.2095-3941.2016.0084
35. Lupia M, Melocchi V, Bizzaro F, et al. Integrated molecular profiling of patient-derived ovarian cancer models identifies clinically relevant signatures and tumor vulnerabilities. Int J Cancer. 2022;151(2):240–254. doi:10.1002/ijc.33983
36. Punzon-Jimenez P, Lago V, Domingo S, Simon C, Mas A. Molecular management of high-grade serous ovarian carcinoma. Int J Mol Sci. 2022;23(22). doi:10.3390/ijms232213777
37. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D’Andrea AD. Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov. 2015;5(11):1137–1154. doi:10.1158/2159-8290.CD-15-0714
38. Shah S, Cheung A, Kutka M, Sheriff M, Boussios S. Epithelial ovarian cancer: providing evidence of predisposition genes. Int J Environ Res Public Health. 2022;19(13). doi:10.3390/ijerph19138113
39. Vasta FM, Dellino M, Bergamini A, et al. Reproductive outcomes and fertility preservation strategies in women with malignant ovarian germ cell tumors after fertility sparing surgery. Biomedicines. 2020;8(12). doi:10.3390/biomedicines8120554
40. Gershenson DM. Update on malignant ovarian germ cell tumors. Cancer. 1993;71(4 Suppl):1581–1590. doi:10.1002/cncr.2820710425
41. Boussios S, Attygalle A, Hazell S, et al. Malignant ovarian germ cell tumors in postmenopausal patients: the Royal Marsden experience and literature review. Anticancer Res. 2015;35(12):6713–6722.
42. Saani I, Raj N, Sood R, et al. Clinical challenges in the management of malignant ovarian germ cell tumours. Int J Environ Res Public Health. 2023;20(12). doi:10.3390/ijerph20126089
43. Roth LM, Czernobilsky B. Perspectives on pure ovarian stromal neoplasms and tumor-like proliferations of the ovarian stroma. Am J Surg Pathol. 2011;35(3):e15–33. doi:10.1097/PAS.0b013e31820acb89
44. Birgersson M, Indukuri R, Antonson P, Nalvarte I, Archer A, Williams C. ERbeta in granulosa cell tumors and its clinical potential. Endocrinology. 2023;164(6). doi:10.1210/endocr/bqad063
45. Colombo N, Parma G, Zanagnolo V, Insinga A. Management of ovarian stromal cell tumors. J Clin Oncol. 2007;25(20):2944–2951. doi:10.1200/JCO.2007.11.1005
46. Boussios S, Moschetta M, Zarkavelis G, Papadaki A, Kefas A, Tatsi K. Ovarian sex-cord stromal tumours and small cell tumours: pathological, genetic and management aspects. Crit Rev Oncol Hematol. 2017;120:43–51. doi:10.1016/j.critrevonc.2017.10.007
47. Baghy K, Ladanyi A, Reszegi A, Kovalszky I. Insights into the tumor microenvironment-components, functions and therapeutics. Int J Mol Sci. 2023;24(24). doi:10.3390/ijms242417536
48. Colvin EK. Tumor-associated macrophages contribute to tumor progression in ovarian cancer. Front Oncol. 2014;4:137. doi:10.3389/fonc.2014.00137
49. Hagemann T, Wilson J, Burke F, et al. Ovarian cancer cells polarize macrophages toward a tumor-associated phenotype. J Immunol. 2006;176(8):5023–5032. doi:10.4049/jimmunol.176.8.5023
50. Nowak M, Klink M. The role of tumor-associated macrophages in the progression and chemoresistance of ovarian cancer. Cells. 2020;9(5). doi:10.3390/cells9051299
51. Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med. 2004;10(9):942–949. doi:10.1038/nm1093
52. Dwivedi M, Tiwari S, Kemp EH, Begum R. Implications of regulatory T cells in anti-cancer immunity: from pathogenesis to therapeutics. Heliyon. 2022;8(8):e10450. doi:10.1016/j.heliyon.2022.e10450
53. Santoiemma PP, Powell DJ. Tumor infiltrating lymphocytes in ovarian cancer. Cancer Biol Ther. 2015;16(6):807–820. doi:10.1080/15384047.2015.1040960
54. Pesce S, Tabellini G, Cantoni C, et al. B7-H6-mediated downregulation of NKp30 in NK cells contributes to ovarian carcinoma immune escape. Oncoimmunology. 2015;4(4):e1001224. doi:10.1080/2162402X.2014.1001224
55. Guo Y, Nemeth J, O’Brien C, et al. Effects of siltuximab on the IL-6-induced signaling pathway in ovarian cancer. Clin Cancer Res. 2010;16(23):5759–5769. doi:10.1158/1078-0432.CCR-10-1095
56. Gong T, Zhang P, Jia L, Pan Y. Suppression of ovarian cancer by low-intensity ultrasound through depletion of IL-6/STAT3 inflammatory pathway-maintained cancer stemness. Biochem Biophys Res Commun. 2020;526(3):820–826. doi:10.1016/j.bbrc.2020.03.136
57. Zhang T, Yang J, Sun Y, et al. Interleukin-6 and hypoxia synergistically promote EMT-mediated invasion in epithelial ovarian cancer via the IL-6/STAT3/HIF-1alpha feedback loop. Anal Cell Pathol. 2023;2023:8334881. doi:10.1155/2023/8334881
58. Lane D, Matte I, Rancourt C, Piche A. Prognostic significance of IL-6 and IL-8 ascites levels in ovarian cancer patients. BMC Cancer. 2011;11:210. doi:10.1186/1471-2407-11-210
59. Xu J, Lin H, Wu G, Zhu M, Li M. IL-6/STAT3 is a promising therapeutic target for hepatocellular carcinoma. Front Oncol. 2021;11:760971. doi:10.3389/fonc.2021.760971
60. Rebe C, Vegran F, Berger H, Ghiringhelli F. STAT3 activation: a key factor in tumor immunoescape. JAKSTAT. 2013;2(1):e23010. doi:10.4161/jkst.23010
61. Wen W, Wu J, Liu L, et al. Synergistic anti-tumor effect of combined inhibition of EGFR and JAK/STAT3 pathways in human ovarian cancer. Mol Cancer. 2015;14:100. doi:10.1186/s12943-015-0366-5
62. Charles KA, Kulbe H, Soper R, et al. The tumor-promoting actions of TNF-alpha involve TNFR1 and IL-17 in ovarian cancer in mice and humans. J Clin Invest. 2009;119(10):3011–3023. doi:10.1172/JCI39065
63. Thaklaewphan P, Ruttanapattanakul J, Monkaew S, et al. Kaempferia parviflora extract inhibits TNF-alpha-induced release of MCP-1 in ovarian cancer cells through the suppression of NF-kappaB signaling. Biomed Pharmacother. 2021;141:111911. doi:10.1016/j.biopha.2021.111911
64. Lawrence T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb Perspect Biol. 2009;1(6):a001651. doi:10.1101/cshperspect.a001651
65. Kan Y, Liu J, Li F. High expression of nuclear transcription factor-kappaB is associated with cisplatin resistance and prognosis for ovarian cancer. Cancer Manag Res. 2020;12:8241–8252. doi:10.2147/CMAR.S265531
66. Wysoczynski M, Shin DM, Kucia M, Ratajczak MZ. Selective upregulation of interleukin-8 by human rhabdomyosarcomas in response to hypoxia: therapeutic implications. Int J Cancer. 2010;126(2):371–381. doi:10.1002/ijc.24732
67. Lokshin AE, Winans M, Landsittel D, et al. Circulating IL-8 and anti-IL-8 autoantibody in patients with ovarian cancer. Gynecol Oncol. 2006;102(2):244–251. doi:10.1016/j.ygyno.2005.12.011
68. Kassim SK, El-Salahy EM, Fayed ST, et al. Vascular endothelial growth factor and interleukin-8 are associated with poor prognosis in epithelial ovarian cancer patients. Clin Biochem. 2004;37(5):363–369. doi:10.1016/j.clinbiochem.2004.01.014
69. Mayerhofer K, Bodner K, Bodner-Adler B, et al. Interleukin-8 serum level shift in patients with ovarian carcinoma undergoing paclitaxel-containing chemotherapy. Cancer. 2001;91(2):388–393.
70. Guo Q, Gao BL, Zhang XJ, et al. CXCL12-CXCR4 axis promotes proliferation, migration, invasion, and metastasis of ovarian cancer. Oncol Res. 2014;22(5–6):247–258. doi:10.3727/096504015X14343704124430
71. Luo N, Chen DD, Liu L, Li L, Cheng ZP. CXCL12 promotes human ovarian cancer cell invasion through suppressing ARHGAP10 expression. Biochem Biophys Res Commun. 2019;518(3):416–422. doi:10.1016/j.bbrc.2019.07.098
72. Sahu A, Raza K, Pradhan D, Jain AK, Verma S. Cyclooxygenase-2 as a therapeutic target against human breast cancer: a comprehensive review. WIREs Mech Dis. 2023;15(3):e1596. doi:10.1002/wsbm.1596
73. Thumkeo D, Punyawatthananukool S, Prasongtanakij S, et al. PGE(2)-EP2/EP4 signaling elicits immunosuppression by driving the mregDC-Treg axis in inflammatory tumor microenvironment. Cell Rep. 2022;39(10):110914. doi:10.1016/j.celrep.2022.110914
74. Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the tumor microenvironment, a convoluted affair mediated by EP receptors 2 and 4. Pharmacol Rev. 2024;76(3):388–413. doi:10.1124/pharmrev.123.000901
75. Drakes ML, Stiff PJ. Regulation of ovarian cancer prognosis by immune cells in the tumor microenvironment. Cancers. 2018;10(9). doi:10.3390/cancers10090302
76. Duncan K, Uwimpuhwe H, Czibere A, et al. NSAIDs induce apoptosis in nonproliferating ovarian cancer cells and inhibit tumor growth in vivo. IUBMB Life. 2012;64(7):636–643. doi:10.1002/iub.1035
77. Finetti F, Travelli C, Ercoli J, Colombo G, Buoso E, Trabalzini L. Prostaglandin E2 and cancer: insight into tumor progression and immunity. Biology. 2020;9(12). doi:10.3390/biology9120434
78. Landskron G, De la Fuente M, Thuwajit P, Thuwajit C, Hermoso MA. Chronic inflammation and cytokines in the tumor microenvironment. J Immunol Res. 2014;2014:149185. doi:10.1155/2014/149185
79. Saed GM, Diamond MP, Fletcher NM. Updates of the role of oxidative stress in the pathogenesis of ovarian cancer. Gynecol Oncol. 2017;145(3):595–602. doi:10.1016/j.ygyno.2017.02.033
80. Schweer D, McAtee A, Neupane K, Richards C, Ueland F, Kolesar J. Tumor-associated macrophages and ovarian cancer: implications for therapy. Cancers. 2022;14(9). doi:10.3390/cancers14092220
81. Yang YL, Yang F, Huang ZQ, et al. T cells, NK cells, and tumor-associated macrophages in cancer immunotherapy and the current state of the art of drug delivery systems. Front Immunol. 2023;14:1199173. doi:10.3389/fimmu.2023.1199173
82. Pan Y, Yu Y, Wang X, Zhang T. Tumor-associated macrophages in tumor immunity. Front Immunol. 2020;11:583084. doi:10.3389/fimmu.2020.583084
83. Oparaugo NC, Ouyang K, Nguyen NPN, Nelson AM, Agak GW. Human regulatory T cells: understanding the role of tregs in select autoimmune skin diseases and post-transplant nonmelanoma skin cancers. Int J Mol Sci. 2023;24(2). doi:10.3390/ijms24021527
84. Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol Cancer. 2020;19(1):116. doi:10.1186/s12943-020-01234-1
85. Sojka DK, Huang YH, Fowell DJ. Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunology. 2008;124(1):13–22. doi:10.1111/j.1365-2567.2008.02813.x
86. Watanabe T, Ishino T, Ueda Y, et al. Activated CTLA-4-independent immunosuppression of Treg cells disturbs CTLA-4 blockade-mediated antitumor immunity. Cancer Sci. 2023;114(5):1859–1870. doi:10.1111/cas.15756
87. Cassar E, Kartikasari AER, Plebanski M. Regulatory T cells in ovarian carcinogenesis and future therapeutic opportunities. Cancers. 2022;14(22). doi:10.3390/cancers14225488
88. Taylor BC, Balko JM. Mechanisms of MHC-I downregulation and role in immunotherapy response. Front Immunol. 2022;13:844866. doi:10.3389/fimmu.2022.844866
89. Dhatchinamoorthy K, Colbert JD, Rock KL. Cancer immune evasion through loss of MHC class I antigen presentation. Front Immunol. 2021;12:636568. doi:10.3389/fimmu.2021.636568
90. Alsaab HO, Sau S, Alzhrani R, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561. doi:10.3389/fphar.2017.00561
91. Revythis A, Limbu A, Mikropoulos C, et al. Recent insights into PARP and immuno-checkpoint inhibitors in epithelial ovarian cancer. Int J Environ Res Public Health. 2022;19(14). doi:10.3390/ijerph19148577
92. Poole EM, Lee IM, Ridker PM, Buring JE, Hankinson SE, Tworoger SS. A prospective study of circulating C-reactive protein, interleukin-6, and tumor necrosis factor alpha receptor 2 levels and risk of ovarian cancer. Am J Epidemiol. 2013;178(8):1256–1264. doi:10.1093/aje/kwt098
93. Szulc-Kielbik I, Kielbik M, Nowak M, Klink M. The implication of IL-6 in the invasiveness and chemoresistance of ovarian cancer cells. Systematic review of its potential role as a biomarker in ovarian cancer patients. Biochim Biophys Acta Rev Cancer. 2021;1876(2):188639. doi:10.1016/j.bbcan.2021.188639
94. Lu C, Zhou L, Ouyang J, Yang H. Prognostic value of lymphocyte-to-monocyte ratio in ovarian cancer: a meta-analysis. Medicine. 2019;98(24):e15876. doi:10.1097/MD.0000000000015876
95. Webb JR, Milne K, Kroeger DR, Nelson BH. PD-L1 expression is associated with tumor-infiltrating T cells and favorable prognosis in high-grade serous ovarian cancer. Gynecol Oncol. 2016;141(2):293–302. doi:10.1016/j.ygyno.2016.03.008
96. Appleton KM, Elrod AK, Lassahn KA, Shuford S, Holmes LM, DesRochers TM. PD-1/PD-L1 checkpoint inhibitors in combination with olaparib display antitumor activity in ovarian cancer patient-derived three-dimensional spheroid cultures. Cancer Immunol Immunother. 2021;70(3):843–856. doi:10.1007/s00262-021-02849-z
97. Bekos C, Pils D, Dekan S, et al. PD-1 and PD-L1 expression on TILs in peritoneal metastases compared to ovarian tumor tissues and its associations with clinical outcome. Sci Rep. 2021;11(1):6400. doi:10.1038/s41598-021-85966-0
98. Cui M, Xia Q, Zhang X, et al. Development and validation of a tumor mutation burden-related immune prognostic signature for ovarian cancers. Front Genet. 2021;12:688207. doi:10.3389/fgene.2021.688207
99. Shao C, Li G, Huang L, et al. Prevalence of high tumor mutational burden and association with survival in patients with less common solid tumors. JAMA Netw Open. 2020;3(10):e2025109. doi:10.1001/jamanetworkopen.2020.25109
100. Guo N, Yang A, Farooq FB, et al. CD8 + T cell infiltration is associated with improved survival and negatively correlates with hypoxia in clear cell ovarian cancer. Sci Rep. 2023;13(1):6530. doi:10.1038/s41598-023-30655-3
101. Raisch J, Darfeuille-Michaud A, Nguyen HT. Role of microRNAs in the immune system, inflammation and cancer. World J Gastroenterol. 2013;19(20):2985–2996. doi:10.3748/wjg.v19.i20.2985
102. Cao J, Zhang Y, Mu J, Yang D, Gu X, Zhang J. Exosomal miR-21-5p contributes to ovarian cancer progression by regulating CDK6. Hum Cell. 2021;34(4):1185–1196. doi:10.1007/s13577-021-00522-2
103. Li X, Wang S, Mu W, et al. Reactive oxygen species reprogram macrophages to suppress antitumor immune response through the exosomal miR-155-5p/PD-L1 pathway. J Exp Clin Cancer Res. 2022;41(1):41. doi:10.1186/s13046-022-02244-1
104. Chen R, Coleborn E, Bhavsar C, et al. miR-146a inhibits ovarian tumor growth in vivo via targeting immunosuppressive neutrophils and enhancing CD8(+) T cell infiltration. Mol Ther Oncolytics. 2023;31:100725. doi:10.1016/j.omto.2023.09.001
105. Zhou Y, Zhang Y, Gong H, Luo S, Cui Y. The role of exosomes and their applications in cancer. Int J Mol Sci. 2021;22(22). doi:10.3390/ijms222212204
106. Xie QH, Zheng JQ, Ding JY, et al. Exosome-mediated immunosuppression in tumor microenvironments. Cells. 2022;11(12). doi:10.3390/cells11121946
107. Li Y, Tan J, Miao Y, Zhang Q. MicroRNA in extracellular vesicles regulates inflammation through macrophages under hypoxia. Cell Death Discov. 2021;7(1):285. doi:10.1038/s41420-021-00670-2
108. Wang Y, Chen G, Dai F, et al. miR-21 induces chemoresistance in ovarian cancer cells via mediating the expression and interaction of CD44v6 and P-gp. Onco Targets Ther. 2021;14:325–336. doi:10.2147/OTT.S286639
109. Liu Y, Li X, Zhang T, Liu G. The roles of exosomes in ovarian cancer chemo-resistance. J Cancer. 2023;14(11):2128–2144. doi:10.7150/jca.84930
110. Hu X, Bian C, Zhao X, Yi T. Efficacy evaluation of multi-immunotherapy in ovarian cancer: from bench to bed. Front Immunol. 2022;13:1034903. doi:10.3389/fimmu.2022.1034903
111. Yunianto I, Currie M, Chitcholtan K, Sykes P. Potential drug repurposing of ruxolitinib to inhibit the JAK/STAT pathway for the treatment of patients with epithelial ovarian cancer. J Obstet Gynaecol Res. 2023;49(11):2563–2574. doi:10.1111/jog.15761
112. Khatoon E, Parama D, Kumar A, et al. Targeting PD-1/PD-L1 axis as new horizon for ovarian cancer therapy. Life Sci. 2022;306:120827. doi:10.1016/j.lfs.2022.120827
113. Zhang M, Mo J, Huang W, Bao Y, Luo X, Yuan L. The ovarian cancer-associated microbiome contributes to the tumor’s inflammatory microenvironment. Front Cell Infect Microbiol. 2024;14:1440742. doi:10.3389/fcimb.2024.1440742
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 3.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.