Next Article in Journal
An AI-Driven Clinical Decision Support Framework Utilizing Female Sex Hormone Parameters for Surgical Decision Guidance in Uterine Fibroid Management
Previous Article in Journal
Factors Associated with Health-Related Quality of Life in Patients with Benign Prostatic Hyperplasia Undergoing Pharmacological Treatment: A Cross-Sectional Study
Previous Article in Special Issue
Characteristics of LGBTQ+ Patients and Their Care in Comparison with Heterosexual Individuals: What Is Important for the OBGYN?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Ovarian Neoplasms: Subtypes and Therapeutic Options

1
Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
2
Cancer Microenvironment Branch, Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
*
Authors to whom correspondence should be addressed.
Medicina 2025, 61(12), 2246; https://doi.org/10.3390/medicina61122246
Submission received: 4 November 2025 / Revised: 2 December 2025 / Accepted: 14 December 2025 / Published: 18 December 2025

Abstract

The ovary, as the primary organ responsible for reproduction and new life, plays a central role in female development, maturation, and health. Neoplasms arising from the ovary and its associated tissues exhibit substantial heterogeneity in their histopathological and molecular profiles, many of which remain poorly understood. This review aims to summarize recent advances in the understanding of genetic alterations underlying ovarian neoplasms and to explore therapeutic strategies informed by molecular biomarkers and tumor microenvironmental factors. A comprehensive literature search was performed, focusing on genomic alterations, biomarker-guided therapies, and tumor microenvironmental modulation in ovarian cancers. Emphasis was placed on studies addressing lipid mediator pathways and their roles in immune regulation and therapeutic response. Based on diagnostic classifications, recurrent alterations in TP53, MYC, PIK3CA, and KRAS are consistently observed across epithelial and germ cell ovarian tumors, whereas non-epithelial subtypes such as sex cord–stromal tumors (SCSTs) and small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT), are predominantly associated with ARID1A and SMARCA4 mutations, respectively. These findings highlight distinct pathogenic mechanisms linked to specific genetic alterations and reveal potential therapeutic vulnerabilities. Moreover, lipid metabolism has been closely implicated in immune surveillance through STING signaling cascades within innate immune cells, suggesting that lipid mediators and their associated genes may represent promising therapeutic targets in ovarian cancers (OCs). Targeting lipid mediators could be particularly effective in relapsed OCs, as modulating innate immune cells within the tumor microenvironment (TME) may enhance immune surveillance and improve antitumor responses. Integrating genetic and microenvironmental insights offers a promising direction for developing more effective and personalized therapeutic strategies in OC.

1. Introduction

According to recent statistics, 55% of ovarian cancer (OC) patients in the United States are diagnosed with distant metastasis, resulting in a 5-year survival rate of only 31.8% [1]. Similarly, in South Korea, 44.5% of OC patients present with distant metastasis, with a corresponding 5-year survival rate of 44.4% [2,3]. In contrast, patients diagnosed with localized (~90%) or regional (~80%) OC have significantly higher survival rates, highlighting the critical need to understand the tumor microenvironment (TME) in order to improve prognosis in metastatic cases.
Recent technological advancements have enabled a deeper understanding of tumorigenesis, malignancy, and drug resistance at the molecular level. These insights have led to the development of advanced therapeutic strategies, many of which are now being evaluated in clinical trials. However, most of the available data—particularly from multiomics-based approaches—derive from large cohorts, predominantly composed of epithelial OCs (EOCs), leaving other ovarian and related neoplasms underrepresented. Moreover, effective therapeutic options for patients with metastatic disease remain limited.
For this review, we collected literature related to OC subtypes—including pathology, diagnostic features, and genetic alterations, without imposing year limitations—from the 2020 WHO classification, cBioPortal databases, and PubMed. When multiple articles addressed the same subtype, we prioritized more recent publications and, when applicable, studies with larger patient cohorts for citation.
Based on this information, we provide an overview of neoplasms arising from ovarian and related tissues, highlighting the associated genetic markers and their relevance to therapeutic strategies. We also examine key components of the TME that influence immune surveillance, with a particular focus on lipid mediators and related immune cells, to inform future therapeutic applications.

2. OC—Histological Subtypes and Genetic Alterations

2.1. OC—Histological Subtypes

Based on histological classification, neoplasms originating from the ovary and related tissues are categorized into seven major types according to their tissue of origin: epithelial (E), mesenchymal, mixed epithelial and mesenchymal, sex cord–stromal (SCS), germ cell (GC), miscellaneous tumors, and tumor-like lesions (Table 1). Within each category, tumors are further classified by their histological characteristics and degree of malignancy—benign, borderline, or malignant (carcinoma). Epithelial ovarian carcinomas (EOCs), the most common subtype, are further subclassified based on histological and prognostic features into high-grade serous (HGS), low-grade serous (LGS), mucinous (M), endometrioid (E), clear cell (CC), Brenner (B), mesonephric-like, undifferentiated, carcinosarcoma, and mixed carcinomas, as outlined by the World Health Organization (WHO) classification of female genital tumors over the past five decades [4,5,6]. These carcinoma subtypes exhibit significant variability in clinical features and molecular composition, often more so than in cell of origin, leading to distinct therapeutic approaches [7].
In South Korea, epithelial carcinomas comprise approximately 81% of all OCs [2]. Among these, serous carcinoma (SOC) is the most prevalent (44.9%), followed by clear cell carcinoma (CCC, 10.2%), endometrioid carcinoma (EndOC, 9.4%), mucinous carcinoma (MOC, 8.8%), and adenocarcinoma not otherwise specified (NOS, 2.9%) [2,3,8]. Non-epithelial OCs include sex cord–stromal tumors (SCSTs, 5.5%) and germ cell tumors (GCTs, 4.2%). Recent advancements in epidemiologic analyses have enabled more refined identification of OC risk factors [7].
Most EOCs show a 5-year survival rate of 80–100% when localized, and 70–90% when regionally spread [9]. However, survival drops significantly to a variable 16–60% when the cancer has metastasized distantly [9]. Specifically in the United States, the 5-year survival rates for HGSOC, CCC, and MOC are 34.5%, 24.0%, and 16.6%, respectively [10]. Similarly, in Norway, 5-year survival rates for advanced-stage (Stage III–IV) HGSOC and CCC are reported to be under 46% and 31%, respectively [11], underscoring the need for robust preclinical models for advanced OC to inform therapeutic strategies. Furthermore, OCs are usually diagnosed with peritoneal metastasis, so that TME is critical for multifocal therapeutic strategies. Non-epithelial ovarian tumors—comprising SCSTs, GCTs, miscellaneous tumors, and tumor-like lesions—are also listed in Table 1. Among these, SCSTs and GCTs are the most prominent subtypes, though their incidence remains low, estimated at 2.1–3.7 cases per million women. Malignant GCTs are responsible for approximately 80% of malignant ovarian tumors in preadolescent females, whereas SCSTs occur across a broader age range, accounting for up to 5% of ovarian malignancies [12,13]. According to a cohort study conducted across MITO centers, the 5-year survival rates for GCT subtypes were as follows: 100% for immature teratomas, 97.9% for dysgerminomas, 69.9% for yolk sac tumors, and 62.9% for mixed germ cell tumors [14].
Table 1. Neoplasms from ovary and related tissues and markers associated with molecular events or diagnosis.
Table 1. Neoplasms from ovary and related tissues and markers associated with molecular events or diagnosis.
Tumor Types (WHO 2020 Classification)Associated with Biomarkers for DiagnosisRef.
1. Epithelial
1.1. Serous
     Serous cystadenoma, adenofibroma and surface papilloma
     Serous borderline tumor
     Low-grade serous carcinoma

     High-grade serous carcinoma


Polyclonal alteration of KRAS, BRAF

Copy number alterations, BRAF, KRAS
KRAS, BRAF, NRAS, and CDKN2A; gain of 1q or 18p; loss of 1p, 18q, and 22
TP53, PIK3CA, HLTF, POLQ, PIK3CB, MET, ARID1B, NF1, MRE11A, CCNE1, RB1, CDK12, PTEN, TP53BP1, BRCA1, BRCA2. Homologous recombination RNA repair defects, whole-genome duplication, MHC-II expression
WT1, estrogen receptor[4,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30]
[31,32,33,34]
1.2. Mucinous
     Mucinous cystadenoma and adenofibroma
     Mucinous borderline tumor
     Mucinous carcinoma
1.3. Endometrioid tumors
     Endometrioid cystadenoma and adenofibroma
     Endometrioid borderline tumor
     Endometrioid carcinoma
1.4. Clear cell tumors
     Clear cell cystadenoma and adenofibroma
     Clear cell borderline tumor
     Clear cell carcinoma
1.5. Seromucinous tumors
     Seromucinous cystadenoma and adenofibroma
     Seromucinous borderline tumor
1.6. Brenner tumors
     Brenner tumor
     Borderline Brenner tumor
     Malignant Brenner tumor
1.7. Other carcinomas
     Mesonephric-like adenocarcinoma

     Undifferentiated and dedifferentiated carcinoma
     Carcinosarcoma

     Mixed carcinoma



ARID1A
TP53, RNF43, ELF3, GNAS, ERBB3, KLF5



ARID1A, CTNNB1,
CTNNB1, PTEN, POLE, MSI,


AKT
[5,35,36,37,38,39,40]
ARID1A
ARID1A, PTEN, PIK3CA, TP53


[40,41,42,43,44,45,46,47,48,49,50]
ARID1A



MDM2, PIK3CA

NRAS, KRAS; gain of 1q or 18p; loss of 1p, 18q, and 22

GATA3, TTF1, CD10
[34]
[10,32,33,51,52]
KIT, EGFR, HER2, TP53, PTEN, CHD4, BCOR, KRAS, PIK3CA, ARID1A, CTNNB1[17,26,36,53,54,55,56,57,58,59,60,61]
2. Mesenchymal
2.1. Endometrioid stromal sarcoma
     Low-grade endometrioid stromal sarcoma
     High-grade endometrioid stromal sarcoma
2.2. Smooth muscle tumors
     Leiomyoma
     Smooth muscle tumor of uncertain malignant potential
     Leiomyosarcoma
2.3. Ovarian myxoma
JAZF1::SUZ12 fusion CD10[54,62,63]
YWHAE::FAM22 fusionCD10
CD10
3. Mixed epithelial and mesenchymal tumors
     Adenosarcoma
CD10[54,62]
4. Sex cord–stromal tumors
4.1. Pure stromal tumors
     Fibroma
     Thecoma
     Sclerosing stromal tumor
     Microcystic stromal tumor
     Signet ring stromal tumor
     Leydig cell tumor
     Steroid cell tumor
     Fibrosarcoma
4.2. Pure sex cord tumors
     Adult granulosa cell tumor
     Juvenile granulosa cell tumor
     Sertoli cell tumor
     Sex cord tumor with annular tubules
4.3. Mixed sex cord–stromal tumors
     Sertoli–Leydig cell tumor
     Sex cord–stromal tumor, NOS
     Gynandroblastoma
FHL2::GLI2 fusion
CTNNB1, APC
[64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80]
FOXL2
AKT1
STK11
DICER1, FOXL2
DICER1
5. Germ cell tumors
5.1. Teratoma, benign
5.2. Immature teratoma, NOS
5.3. Dysgerminoma
5.4. Yolk sac tumor
5.5. Embryonal carcinoma
Isochromosome 12p
Isochromosome 12p, KIT
Isochromosome 12p

Isochromosome 12p
AFP
Sall4, OCT3/4, LDH
Sall4, AFP
Sall4, OCT3/4, SOX2, β-hCG, AFP
β-hCG
LDH, AFP, β-hCG
[12,13,69,72,81,82,83,84,85,86,87,88,89,90,91,92]
5.6. Choriocarcinoma, NOS
5.7. Mixed germ cell tumor
5.8. Monodermal teratomas and somatic type tumors arising from a dermoid cyst
     Struma ovarii, NOS
     Struma ovarii, malignant
     Strumal carcinoid
     Teratoma with malignant transformation
     Cystic teratoma, NOS
5.9. Germ cell sex cord–stromal tumors
     Gonadoblastoma,
     Mixed germ cell—sex cord–stromal tumor, unclassified
BRAFPLAP, OCT4
OCT3/4
12p amplification
6. Miscellaneous tumors
     Rete cystadenoma, adenoma and adenocarcinoma
     Wolffian tumor
     Solid pseudopapillary tumor
     Small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT)
     Wilms tumor
CTNNB1
SMARCA4
[45,93,94,95,96,97,98,99,100,101,102,103]
7. Tumor-like lesions
     Follicle cyst
     Corpus luteum cyst
     Large solitary luteinized follicle cyst
     Hyperreactio luteinalis
[64,104]
NOS, not otherwise specified; AFP, α-fetoprotein; LDH, lactate dehydrogenase; β-hCG, beta-human chorionic gonadotropin.

2.2. OC-Genetic Alterations Associated with Subtypes

Several genes involved in key molecular events during OC development have been identified, many of which also represent potential targets for therapeutic intervention (Table 1). Subtype-specific genetic alterations were identified and further analyzed in terms of their alteration frequencies across several cohort studies, including those conducted by The Cancer Genome Atlas (TCGA) Research Network [29,105], the Memorial Sloan Kettering Cancer Center (MSKCC) [106,107], and others, as shown in Table 2 [31,97,108]. As expected, TP53 is the most frequently altered gene in HGSOC, with mutations observed in over 90% of cases in each cohort [29,108]. Notably, TP53 alterations are also present in approximately 50% of MOCs [31]. In contrast, LGSOC is characterized by a distinct pattern, with frequent KRAS mutations but a relatively low frequency of TP53 alterations [107]. Data from one of the largest tissue-based cancer patient cohorts, the MSKCC-IMPACT database, include information on GCTs, such as mixed germ cell tumors, yolk sac tumors, choriocarcinomas, teratomas (both mature and immature), and teratomas with malignant transformation. In these tumors, mutation frequencies were reported as follows: TP53 (54%), KRAS (28%), and PTEN (11%) [106]. Interestingly, in one of the miscellaneous tumor types—small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT)—a striking dependence on SMARCA4 alterations has been observed, with up to 92% of cases [97]. SCSTs were associated with FOXL2 and CTNNB1, 68% and 16%, respectively.

3. OC—Current Therapeutic Strategies

Alterations in TP53, MYC, PIK3CA, and KRAS predominated in epithelial and GCTs, whereas ARID1A and SMARCA4 mutations were characteristic of SCST and SCCOHT, respectively, indicating chromosomal instability in non-epithelial OCs (Table 2). Alterations in BRCA1 are frequently observed in OCs (Table 2). A recent study identified an intriguing association between BRCA1 mutation carriers and elevated blood molybdenum levels [109], a trace element linked to mitochondrial biogenesis [110]. However, the majority of genetic alterations are still being investigated for their roles as risk factors, and effective therapeutic strategies to target these alterations remain in early stages of development. Regardless of genetic alterations, standard treatments against OCs include cytoreductive surgery and chemotherapy with platinum drugs. Despite initial responsiveness to cytoreductive surgery and platinum-based chemotherapy, the majority of patients with ovarian cancer ultimately experience disease relapse. Approximately 70% of patients develop recurrent, more aggressive, and chemotherapy-resistant tumors following first-line therapy [111]. Conventional treatment strategies—comprising maximal cytoreductive surgery followed by adjuvant chemotherapy—have succussed only modest improvements over the past five decades, and the 5-year survival rate for patients with advanced or metastatic disease remains below 30% [112]. Even among those receiving optimal standard-of-care regimens, most relapse within a few years, and subsequent lines of therapy yield diminishing responses, culminating in a 5-year overall survival rate of only 30–40% worldwide. The introduction of maintenance therapy with poly(ADP-ribose) polymerase inhibitors (PARPis) has extended progression-free survival (PFS) and, in selected populations, improved long-term overall survival [113,114]. Nevertheless, a substantial proportion of patients either fail to respond initially or acquire resistance during treatment, limiting durable benefit. Such intrinsic and acquired drug resistance remains a major therapeutic obstacle and the principal cause of poor prognosis in ovarian cancer. Overcoming these resistance mechanisms (spanning defects in DNA damage response reprogramming, replication stress tolerance, and TME-mediated immune suppression) represents a critical unmet need for improving patient outcomes.

4. OC—Therapeutic Strategies Targeting TME

4.1. Lipid-Driven Immune Suppression in the Ovarian Cancer TME

4.1.1. Tumor-Intrinsic Lipid-Mediated Immunosuppression

Aberrant lipid metabolism and bioactive lipid mediators are increasingly recognized as key determinants of immune evasion in ovarian cancer. Cyclooxygenase-derived prostaglandin E2 (PGE2), mainly produced in tumors, is a central lipid mediator in tumors and directly disables dendritic cell (DC) function via EP2/EP4 signaling, skewing myeloid cells toward suppressive phenotypes and blunting T-cell priming. Selective EP2/EP4 blockade restores antitumor DC activity in human settings, underscoring the pathway’s translational relevance [115]. Beyond PGE2, arachidonate 5-lipoxygenase (5-LOX)-derived leukotrienes correlate with poor prognosis, enhance invasion, and recruit tumor-promoting macrophages, reinforcing a lipid driven immunosuppressive loop. COX-2/PGE2 signaling also promotes VEGF and metastatic programs that indirectly suppress immune control [116].

4.1.2. Tumor-Extrinsic Lipid-Mediated Immunosuppression

Ovarian tumors grow in adipose-rich peritoneal/omental spaces. Adipocyte-derived lipid moieties make tumors resistant to immune-activating oncolytic virotherapy. Depleting lipids from adipocyte-conditioned medium or blocking fatty acid uptake resensitizes OC models, highlighting how excess fatty acids can keep “cold” tumors. At a tissue scale, high-fat environments reduce intratumoral viral titers and favor therapeutic resistance [117].
Ascites and omental niches provide abundant fatty acids that OC cells rapidly absorb, boosting ATP production, suppressing AMPK, and activating mTOR/TAK1–NF-κB programs that drive aggressiveness that also favor immunosuppression. Targeting this axis (AMPK activation + TAK1 and FASN inhibition) synergistically impairs peritoneal metastases, suggesting a route to relieve lipid conditioned immune dysfunction [118]. OC-derived TGF-β1 converts omental adipocytes into cancer-associate adipocytes (CAAs) through SMAD3/TRIB3, which remodel the extracellular matrix (ECM, collagen/fibronectin) and raise IL-1β/IL-6 changes that support implantation and can further skew myeloid cells toward suppressive states. Pharmacologic blockade of TGF-β/SMAD3 prevents CAA/PMN formation and reduces metastatic burden [119].

4.2. Type I Interferon (IFN) and STING Activation Pathways in OC: Mechanistic Insights and Therapeutic Opportunities

The cGAS–STING pathway acts as a cytosolic DNA-sensing system that connects innate detection to adaptive immune activation. Binding of cytoplasmic DNA activates cGAS, generating cyclic GMP–AMP (cGAMP) that engages STING, leading to TBK1–IRF3 phosphorylation and type I interferon (IFN-I) production. These cytokines drive DC maturation, antigen presentation, and CD8+ T-cell priming. In OCs, persistent genomic instability continuously releases DNA into the cytosol, yet post-translational repression, ER stress, and lipid-mediated suppression often blunt STING signaling. A recent review of gynecologic malignancies reported that cGAS and STING are broadly expressed but functionally silenced in OC, indicating that pharmacologic re-engagement of this pathway could restore antitumor immunity [120]. DNA damage-response (DDR) inhibitors can activate cGAS–STING by generating cytosolic DNA. In BRCA1-deficient OC, PARP inhibition provokes STING-dependent IFN-β secretion, CD8+ T-cell infiltration, and tumor regression; loss of STING or TBK1 abolishes these effects [121]. Moreover, PARP inhibition upregulates PD-L1, CXCL10, and CCL5, explaining synergy with PD-L1 blockade [122]. Similarly, CX-5461, an RNA polymerase I inhibitor, causes nucleolar stress and cytosolic-DNA accumulation that activates STING–IRF3 signaling and type I IFN transcription, suppressing tumor growth [123]. Together, DDR-targeting agents transform immune-cold OC into IFN-rich, T-cell-inflamed microenvironments.
Ovarian tumors employ intrinsic brakes on STING activation. The deubiquitinase, USP35, binds phosphorylated STING and removes activating K63-linked ubiquitin chains, thereby inhibiting TBK1 and IRF3 phosphorylation and suppressing IFN-β expression [124]. Similarly, mitotic disruption can reignite innate sensing: CENPM knockdown triggers cytosolic-DNA accumulation, activates cGAS–STING, and induces pyroptosis, suppressing ovarian-tumor growth [125]. Relieving post-translational inhibition or exploiting mitotic stress thus restores endogenous STING–IFN signaling in OC.
Cofactor ions and metabolic states modulate STING activation. Manganese (Mn2+), a natural cGAS cofactor, enhances enzymatic activity. In the preclinical OC model, Mn2+ strengthened macrophage phagocytosis, CD8+ recruitment, and type I IFN production, reshaping the tumor milieu toward immune stimulation [126]. At the adaptive level, raising T-cell NAD+ downregulates the Golgi transporter SURF4, stabilizing STING. This NAD+–SURF4–STING axis, discovered by Jiacheng Shen et al. [127], enhances T-cell cytotoxicity and cooperates with tumor-cell STING activation. Hence, ionic and metabolic cues can globally reinforce STING-driven antitumor immunity [127].
Merging DDR targeting with nanotechnology can intensify STING activation. Polymer–PARP-inhibitor conjugates co-delivering a USP1 inhibitor (USP1i), which increased DNA damage, caused cytosolic-DNA accumulation, and robustly activated cGAS–STING signaling. This approach elevated IFN-I genes; enhanced CD8+ T-cell infiltration; and, when combined with PD-1 blockade, produced synergistic tumor regression in high-grade serous OC [128]. Such nanoplatforms exemplify next-generation synthetic-lethality immunotherapies that couple DNA-repair inhibition with innate immune activation. Clinically, BRCA1/HRD-positive tumors exhibit strong DDR–STING coupling and respond favorably to PARPi plus PD-L1 blockade [121,122]. Biomarkers such as phospho-STING, ISG signatures (CXCL10 and CCL5), and USP35 expression may help select patients. Because sustained STING activity can pivot toward NF-κB-dominant inflammation and T-cell exhaustion, future strategies should emphasize transient, immunogenic activation combined with checkpoint or metabolic therapy to produce durable, well-tolerated IFN-driven responses.

4.3. DC-Based Immunotherapy and Reprogramming Strategies in OC

The ovarian TME is profoundly immunosuppressive, characterized by lipid mediators, cytokines, and metabolic stress that collectively impair DC maturation and antigen presentation. OCs recruit large numbers of immature myeloid progenitors that differentiate into dysfunctional tumor-associated DCs, driven by β-defensin/CCR6-dependent chemotaxis and VEGF-A-mediated pro-angiogenic programming. These tumor-infiltrating DCs exhibit high PD-L1, CD277 (BTN3A1) expression, and Arginase-1 activity, and they release immunoregulatory cytokines that collectively suppress T-cell priming. Metabolic and stress-response pathways further exacerbate this dysfunction. OC-derived PGE2 and TGF-β jointly upregulate PD-L1 and Arginase-1 activity in DCs, while ER stress-induced activation of XBP1 promotes lipid droplet accumulation and defective cross-presentation. Downregulation of miR-155, a key regulator of antigen-presenting capacity in DC, also contributes to the tolerogenic phenotype [129]. Consequently, OC-associated DCs act not as immune initiators but as facilitators of angiogenesis and immune tolerance.
Recent advances in single-cell and functional analyses have delineated how different DC subsets are selectively modulated in OC. cDC1s (CD103+/XCR1+) are the principal cross-presenting cells responsible for CD8+ T-cell priming; however, their abundance and type I interferon signaling are markedly reduced in ascitic and metastatic lesions. cDC2s (CD11b+/Sirpα+) retain plasticity but frequently acquire a suppressive or Th2-skewing profile under the influence of IL-6, VEGF, and lactic acid. Plasmacytoid DCs (pDCs), though capable of secreting large amounts of type I interferons, often become functionally exhausted in the ovarian TME, expressing high levels of IDO1, PD-L1, and ICOS-L that promote regulatory T-cell (Treg) expansion and correlate with poor prognosis. The lipid mediator PGE2 plays a dominant role in DC suppression by signaling through EP2 and EP4 receptors.
Fibrinogen-like protein 2 (FGL2) has emerged as a potent DC-suppressive factor within OC ascites. FGL2, primarily produced by tumor-infiltrating macrophages/monocytes and DCs, downregulates CD40 and CD86 on CD11+ DCs through FcγRIIB/III-mediated signaling. In Fgl2−/− mice, ovarian tumors displayed enhanced infiltration of cDC1s, CD3+ T cells, and DNAM-1+ NK cells, and they induced expression of co-stimulatory markers (MHC-II, CD86+, and CD40+) in spleen, resulting in reduced tumor burden. These findings identify FGL2 as a non-canonical immune checkpoint that limits DC maturation and adaptive immune activation [130].
DC-based vaccines have long been pursued as a therapeutic modality for OC. Early clinical trials were conducted using monocyte-derived DCs (moDCs) pulsed with HER-2/neu, WT-1, MUC1, or tumor lysates [131]. Contemporary approaches are refining vaccine composition through improved antigen loading, maturation stimuli, and DC subset selection. Reviews by Caro and Zhang emphasize the superior antigen-presenting potential of cDC1s and the promise of fusion-cell and in vivo targeted DC vaccines to enhance cytotoxic T-cell responses [131,132].
A particularly innovative strategy was reported by Zhang, who developed a DC–tumor cell fusion membrane nano-vaccine (FCM-NP). The FCM-NPs, loaded with CpG-ODN adjuvant, promoted DC maturation, and elicited robust CD8+ T-cell responses in multiple OC models, effectively delaying tumor growth and abdominal metastasis [133]. Restoration of type I IFN signaling is a critical determinant of DC immunogenicity. Nanoparticles co-delivering Mn2+ and platinum were shown to activate the cGAS–STING pathway in cancer cells, and they enhanced maturation of DCs (CD80+ CD86+) for potent antitumor immunity in OC models [134]. When combined with DC-targeted strategies such as EP2/EP4 inhibition or fusion-cell vaccination, these approaches hold potential to overcome the immune-exclusion barriers of advanced disease.
Collectively, these studies establish a mechanistic linking lipid-mediated suppression (PGE2–EP2/EP4 axis), immune checkpoints (FGL2 and PD-L1), and metabolic stress (IRE1 alpha-XBP1 pathway) to DC dysfunction in OC. Therapeutic reprogramming of DCs—through nanoparticle-based EP2/EP4 blockade, FGL2 inhibition, STING activation, or next-generation DC vaccines—offers a strategy to convert the ovarian TME from an immunologically “cold” environment into a type I IFN-rich, DC-driven immunogenic niche capable of sustaining durable T-cell-mediated tumor control.

4.4. Tumor-Associated Macrophages (TAMs) in OC: Reprogramming Immunosuppressive Niches Toward Antitumor Immunity

TAMs represent the dominant myeloid population in OC ascites and metastatic implants. These cells exhibit remarkable phenotypic plasticity, polarizing between M1-like (pro-inflammatory) and M2-like (immunoregulatory) states depending on tumor-derived cues. HGSOCs are characterized by a predominance of M2-like macrophages expressing CD163, CD206, ARG1, and IL-10, which foster immune evasion, angiogenesis, and chemotherapy resistance. In contrast, M1-like macrophages—driven by IFN-γ, TNF-α, or STING activation—correlate with improved survival outcomes through antigen presentation and cytotoxic T-cell recruitment. A recent study reported that the tumor ECM directly educates macrophages toward a tissue-remodeling, immunoregulatory phenotype that recapitulates the transcriptional profile of TAMs in patient metastases [135]. Using a decellularized omental metastasis model preserving the ECM’s native structure, the authors found that infiltrating monocytes differentiated into macrophages expressing a M0/M2-like phenotype. These “matrix-educated macrophages (MAMs)” diminished T-cell activation, revealing that ECM composition alone is sufficient to drive TAM polarization toward immune suppression and tissue remodeling [135]. Targeting desmoplastic ECM components (e.g., COL11A1, FN1, VCAN, MXRA5, and SFRP2) can recondition these MAMs, enhancing antitumor immunity. Complement pathways critically shape macrophage polarization within the ovarian TME. Fang identified that loss of KLHDC8A in normal ovarian epithelial cells triggers C5a secretion, which acts through C5aR/p65 NF-κB signaling to polarize macrophages toward a pro-tumoral phenotype [136]. This axis was validated by the reversal of M2 polarization (CD206) upon NF-κB inhibition, demonstrating that C5a/C5aR blockade may restore immune surveillance. Complementary evidence shows that C5aR inhibition synergizes with CXCL9 upregulation to enhance CD8+ T-cell infiltration, linking complement control with adaptive immunity reactivation [137].
Macrophage checkpoint pathways represent another central barrier to antitumor immunity. Liu first showed that CD47, a “don’t eat me” signal overexpressed on OC cells, suppresses macrophage phagocytosis and correlates with poor prognosis [138]. CD47 knockdown or blockade with monoclonal antibodies enhanced macrophage infiltration, increased the phagocytic index, and prolonged survival in xenografted mice [138]. Building on this, Yang developed a bispecific antibody fusion protein that simultaneously targets CD47/SIRPα and CD24/Siglec-10 axes—dual “don’t eat me” checkpoints co-expressed in OCs [139]. In preclinical models, PPAB001 therapy promoted phagocytosis of human OC cell line and enhanced tumor regression in SK-OV-3 xenografts [139]. Furthermore, a phase I clinical trial of the bispecific Fc-fusion protein SL-172154, which simultaneously blocks CD47 and activates antigen-presenting cells (APCs), has been reported [140]. These findings define bispecific macrophage checkpoint inhibitors as a potent next-generation immunotherapy to reprogram TAMs toward antitumor activity. Chemoresistance in OC is partly sustained by macrophage-driven immunosuppression. Li uncovered that the circular RNA circITGB6 promotes cisplatin resistance by driving TAM polarization via a circITGB6–IGF2BP2–FGF9 RNA–protein complex, which stabilizes FGF9 mRNA [141]. Elevated circITGB6 correlated with high CD206+ macrophage infiltration, mediating immunosuppressive TME. Notably, FGF9 deletion or ASO-mediated circITGB6 silencing restored M1 markers (TNF-α and iNOS) and decreased M2 cytokines (IL-10 and ARG1) [141]. Additionally, ongoing clinical trials of immune cell-based therapy are listed in Table 3.
Advances in biomaterial immunotherapy have enabled macrophage-targeted reprogramming. VandenHeuvel et al. introduced a lipid nanoparticle (LNP)-based siRNA system that selectively delivers siSIRPα to TAMs, effectively silencing the CD47–SIRPα checkpoint [148]. This “macrophage checkpoint nano-immunotherapy” reversed carboplatin resistance, and decreased spheroid invasion in 3D OC models. By exploiting the natural phagocytic properties of macrophages, these LNPs represent a scalable strategy to restore innate immune clearance without systemic toxicity.
Host obesity increases TAM density, lowers the pro-to-anti-inflammatory macrophage ratio, and is associated with reduced chemotherapy responsiveness. Strategies that deplete or repolarize M2-biased TAMs may counteract obesity-linked immune suppression and improve outcomes [149].
Collectively, these studies delineate multifaceted TAM reprogramming mechanisms in OC—ranging from ECM-mediated education and complement signaling to checkpoint inhibition and RNA-mediated polarization. Therapeutic integration of ECM remodeling, checkpoint blockade (CD47/CD24 bispecific antibodies), and circITGB6–FGF9 axis suppression could synergize with DC or NK cell-activating platforms, driving a comprehensive myeloid rejuvenation within the ovarian TME.
A phase I clinical trial of a mesothelin–CD40-bispecific antibody has been conducted in solid tumors, but its efficacy in ovarian cancer was found to be limited [150]. In addition, a phase I trial employing the myeloid-targeting antibodies PY159 and PY314 was performed; despite the complementary strategies of PY159 (promoting antitumor immunity) and PY314 (depleting TAMs), the results were largely limited to disease stabilization [151]. These findings highlight the need to understand the complex immunosuppressive mechanisms of the OC TME and to develop strategies for TAM suppression and reprogramming.

4.5. Natural Killer (NK) Cells in OC: Dysfunction, Engineering, and Combination Strategies

Innate cytotoxic NK cells are potent effectors against EOC, yet their function is often curtailed by ascites-borne suppressive cues. High peritoneal TGF-β1 was strongly associated with ascites-induced NK dysfunction, and it reduced progression-free and overall survival. Pharmacologic blockade of TGF-β signaling partially restored NK proliferation and function ex vivo, nominating TGF-β1 as a dominant soluble inhibitor in HGEOC [152]. Cytokine-inducible regulators also rise within the ovarian milieu: CISH, a regulator contributing to exhaustion is enriched in both early and late stages, with CISH levels positively associated with IL-10 and ER-stress marker GRP78 in tumor tissues—linking chronic cytokine/stress exposure to NK exhaustion [153]. Beyond cytokines, tumor iron overload leads to an immunosuppressive peritoneal niche. In a preclinical animal model and patient-derived organoid system, the FDA-approved chelator deferiprone reprogrammed OC cells to produce type I IFN, NKG2D ligand-encoding genes (e.g., Mult1, H60b, and Ulbp1) and drive NK-dependent control of metastatic disease. Mechanistically, iron chelation triggered releasing mtDNA and DNA-damage responses, amplifying IFN signaling and inducing a DC-IL-15-NK axis that accumulated NK cells at tumor sites and improved survival [154]. Cytokine-induced memory-like (CIML) NK cells retain heightened activation after brief cytokine priming. Arming CIML NKs with a mesothelin (MSLN)-targeting CAR (membrane-proximal epitope) produced durable antitumor activity in EOC lines, resisted dysfunction in patient ascites, and prevented metastasis in xenografts—outperforming conventional CAR-NKs [155]. To enable off-the-shelf manufacturing, hESC-derived MSLN CAR-NK cells were generated, and they eliminated human ovarian tumor cells, underscoring feasibility for standardized CAR-NK products [156]. For clinical translation, non-invasive tracking is crucial. HER2-CAR-NK-92 cells co-expressing the human sodium-iodide symporter (NIS) were visualized by PET, while simultaneously reducing tumor burden and prolonging survival in a HER2+ ovarian model—demonstrating that human-derived reporter imaging can monitor NK persistence and biodistribution alongside efficacy [157].
Oncolytic platforms can both arm NK cells and reshape tumor immunogenicity. A vIL-2-encoding oncolytic adenovirus selectively boosted effector NK/CD8+ T cells without expanding Tregs in ex vivo human ovarian co-cultures and improved in vivo control of patient-derived xenografts when combined with adoptive allogeneic NK therapy [158]. These experimental findings align with broader synthesis indicating that combination therapy (oncolytic viruses, targeted antibodies, and checkpoint blockade) can overcome NK deficits and heighten tumor susceptibility to NK cytotoxicity [159].
Clinical trials using NK cells (Table 3) in recurrent OC have been conducted [160,161,162]. In two phase I studies, ex vivo cultured allogeneic NK cells were administered intraperitoneally, but limitations in cell yield and uncertainty regarding in vivo expansion were observed [161,162]. In a phase II study, Hi-Cy/Flu-based lymphodepletion and IL-2 administration were used to promote NK cell expansion in vivo; however, donor NK cell expansion was inhibited by immunosuppressive factors within the TME. As a result, NK cells were only transiently detectable, and they were largely replaced by host T cells by day 14, indicating a failure of sustained expansion. Additionally, IL-2 administration intended to support NK cell proliferation was associated with concurrent expansion of Tregs [160].
Collectively, preclinical models have shown that OC suppresses NK surveillance through TGF-β1, CISH-linked chronic cytokine/stress programs, and metabolic (iron) conditioning. Convergent solutions—type I IFN restoration (iron chelation), precision CD16a engagement, CIML/CAR engineering (including off-the-shelf products), oncolytic-vIL-2 therapy, and real-time imaging—map a practical route to durable NK-mediated control in the peritoneal cavity [152,153,154]. In clinical studies involving patients with recurrent OC, however, NK cell-based therapies demonstrated limited in vivo expansion and persistence. These findings suggest that, to enhance the efficacy of NK cell-based therapies in the clinic, strategies targeting the TME concurrently with NK cells should be considered [160,161,162].

4.6. Cancer-Associated Fibroblasts (CAFs) in Ovarian Cancer Targeting Fibroblast-Driven Oncogenic and Immunosuppressive Pathways

Cancer-associated fibroblasts (CAFs) play key roles within the ovarian cancer microenvironment and have recently emerged as promising therapeutic targets. Multiple studies have reported that CAF-derived growth factors and cytokines directly contribute to OC cell proliferation, invasion, chemoresistance, and the establishment of an immunosuppressive TME [163,164]. Among these factors, CAF-secreted FGF7 promotes OC cell growth and migration and is associated with poor prognosis [164]. In addition, CAF-derived WNT5A drives the maintenance and expansion of cancer stem cells, and inhibition of WNT5A reduces stemness and chemoresistance. These findings indicate that targeting CAF-derived signaling may suppress OC progression and restore responsiveness to anticancer immunotherapies [163].
From an immunotherapy perspective, strategies to target CAFs are also being explored. The fibroblast activation protein-specific 4-1BB ligand fusion protein (FAP-4-1BBL) has shown the ability to enhance intra-tumoral T-cell proliferation and function in preclinical models [165]. Furthermore, early clinical trials of RO7122290, a complex combining FAP binding with 4-1BB co-stimulation, have demonstrated tumor-localized T-cell activation [166]. These results support the concept that CAF-targeted approaches may represent viable immune-enhancing strategies in ovarian cancer.
However, CAFs do not constitute a uniform population; rather, they exhibit substantial heterogeneity, and some CAF subsets can exert tumor-restraining or immunomodulatory effects. Indeed, depletion of specific CAF populations has paradoxically resulted in enhanced metastasis and other unintended consequences, highlighting the potential risks associated with CAF-depletion strategies [167,168]. Moreover, studies directly characterizing the immunoregulatory functions of CAFs in OC remain limited, and a systematic definition of CAF subtypes, cellular origins, and functional markers in this disease is still lacking. Consequently, comprehensive preclinical studies and integrative analyses of CAF heterogeneity are needed to clarify how CAF-derived signals shape immune regulation and to assess the therapeutic potential of targeting CAFs in OC.

5. Discussion

OC is a highly heterogeneous malignancy with diverse histological and molecular subtypes that display distinct clinical behaviors. EOCs constitute the majority, with HGSOC being most common and characterized by TP53 mutations. In contrast, non-epithelial tumors such as SCST and GCT are rarer but harbor unique genetic drivers, including FOXL2, SMARCA4, and KRAS. Despite progress in surgery and chemotherapy, recurrence and drug resistance remain major obstacles. A deeper understanding of subtype-specific molecular alterations and the TME has driven the development of targeted and immune-based therapies. Current therapeutic paradigms are evolving toward multi-modal immune-metabolic approaches that integrate metabolic reprogramming, STING–IFN pathway activation, and myeloid cell (DC and macrophage) reprogramming to convert tumors from “immune-cold” to “immune-active.” Furthermore, engineered NK cells (CAR-NK and CIML-NK) and nanoparticle-based delivery systems enhance immune precision and cytotoxic efficacy. Collectively, these integrated strategies represent a next-generation treatment paradigm aimed at achieving durable immune control and improving long-term survival in OC (Figure 1).

6. Conclusions

Despite advances in cytoreductive surgery, platinum-based chemotherapy, and PARP inhibitor-based maintenance therapy, durable control of ovarian cancer remains limited, reflecting gaps in our understanding of oncogenic drivers and tumor–immune interactions. Future progress will require clearer definition of lineage-specific programs and targeted disruption of metabolic and immune regulatory pathways, particularly those shaping lipid-conditioned stromal niches and suppressing type I IFN–STING signaling. Major translational barriers—including intratumoral heterogeneity, poor drug penetration, and resistance to PARP inhibitors and immunotherapy—highlight the need for integrated multiomics profiling, patient-derived models, and improved delivery platforms. Ultimately, therapeutic strategies combining targeted agents with modulators of innate immunity and TME reprogramming hold promise for advancing next-generation ovarian cancer treatment.

Author Contributions

Conceptualization, C.-S.C. and H.J.Y.; methodology, S.Y.H., A.C., C.-S.C. and H.J.Y.; resources, A.C., S.Y.H., C.-S.C. and H.J.Y.; data curation, H.J.Y.; writing—original draft preparation, S.Y.H., C.-S.C. and H.J.Y.; writing—review and editing, C.-S.C. and H.J.Y.; supervision, C.-S.C. and H.J.Y.; project administration, C.-S.C. and H.J.Y.; funding acquisition, C.-S.C. and H.J.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by National Cancer Center Grants NCC-2410861 (to H.J.Y.), NCC-2410862 (to C.-S.C.), and NCC-2310660 (to C.-S.C.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not appliable.

Data Availability Statement

The data that support the findings of this work are available from the corresponding author upon request.

Acknowledgments

We thank Hyun Sang Cho for his support on this manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sherman, R.L.; Firth, A.U.; Henley, S.J.; Siegel, R.L.; Negoita, S.; Sung, H.; Kohler, B.A.; Anderson, R.N.; Cucinelli, J.; Scott, S.; et al. Annual Report to the Nation on the Status of Cancer, featuring state-level statistics after the onset of the COVID-19 pandemic. Cancer 2025, 131, e35833. [Google Scholar] [CrossRef] [PubMed]
  2. Park, E.H.; Jung, K.W.; Park, N.J.; Kang, M.J.; Yun, E.H.; Kim, H.J.; Kim, J.E.; Kong, H.J.; Choi, K.S.; Yang, H.K.; et al. Cancer Statistics in Korea: Incidence, Mortality, Survival, and Prevalence in 2022. Cancer Res. Treat. 2025, 57, 312–330. [Google Scholar] [CrossRef]
  3. Yun, B.S.; Park, E.H.; Ha, J.; Lee, J.Y.; Lee, K.H.; Lee, T.S.; Lee, K.J.; Kim, Y.J.; Jung, K.W.; Roh, J.W. Incidence and survival of gynecologic cancer including cervical, uterine, ovarian, vaginal, vulvar cancer and gestational trophoblastic neoplasia in Korea, 1999-2019: Korea Central Cancer Registry. Obstet. Gynecol. Sci. 2023, 66, 545–561. [Google Scholar] [CrossRef]
  4. Kobel, M.; Kang, E.Y. The Evolution of Ovarian Carcinoma Subclassification. Cancers 2022, 14, 416. [Google Scholar] [CrossRef]
  5. De Leo, A.; Santini, D.; Ceccarelli, C.; Santandrea, G.; Palicelli, A.; Acquaviva, G.; Chiarucci, F.; Rosini, F.; Ravegnini, G.; Pession, A.; et al. What Is New on Ovarian Carcinoma: Integrated Morphologic and Molecular Analysis Following the New 2020 World Health Organization Classification of Female Genital Tumors. Diagnostics 2021, 11, 697. [Google Scholar] [CrossRef] [PubMed]
  6. WHO. Classification of Tumours Editorial Board. In Female Genital Tumours, 5th ed.; International Agency for Research on Cancer: Lyon, France, 2020. [Google Scholar]
  7. Wang, M.; Bi, Y.; Jin, Y.; Zheng, Z.J. Global Incidence of Ovarian Cancer According to Histologic Subtype: A Population-Based Cancer Registry Study. JCO Glob. Oncol. 2024, 10, e2300393. [Google Scholar] [CrossRef]
  8. Ha, H.I.; Chang, H.K.; Park, S.J.; Lim, J.; Won, Y.J.; Lim, M.C. The incidence and survival of cervical, ovarian, and endometrial cancer in Korea, 1999-2017: Korea Central Cancer Registry. Obstet. Gynecol. Sci. 2021, 64, 444–453. [Google Scholar] [CrossRef]
  9. Matz, M.; Coleman, M.P.; Carreira, H.; Salmeron, D.; Chirlaque, M.D.; Allemani, C.; Group, C.W. Worldwide comparison of ovarian cancer survival: Histological group and stage at diagnosis (CONCORD-2). Gynecol. Oncol. 2017, 144, 396–404. [Google Scholar] [CrossRef]
  10. Peres, L.C.; Cushing-Haugen, K.L.; Kobel, M.; Harris, H.R.; Berchuck, A.; Rossing, M.A.; Schildkraut, J.M.; Doherty, J.A. Invasive Epithelial Ovarian Cancer Survival by Histotype and Disease Stage. J. Natl. Cancer Inst. 2019, 111, 60–68. [Google Scholar] [CrossRef]
  11. Fortner, R.T.; Trewin-Nybraten, C.B.; Paulsen, T.; Langseth, H. Characterization of ovarian cancer survival by histotype and stage: A nationwide study in Norway. Int. J. Cancer 2023, 153, 969–978. [Google Scholar] [CrossRef] [PubMed]
  12. De Maria, F.; Amant, F.; Chiappa, V.; Paolini, B.; Bergamini, A.; Fruscio, R.; Corso, G.; Raspagliesi, F.; Bogani, G. Malignant germ cells tumor of the ovary. J. Gynecol. Oncol. 2025, 36, e108. [Google Scholar] [CrossRef] [PubMed]
  13. Ray-Coquard, I.; Morice, P.; Lorusso, D.; Prat, J.; Oaknin, A.; Pautier, P.; Colombo, N.; Committee, E.G. Non-epithelial ovarian cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, iv1–iv18. [Google Scholar] [CrossRef] [PubMed]
  14. Mangili, G.; Sigismondi, C.; Gadducci, A.; Cormio, G.; Scollo, P.; Tateo, S.; Ferrandina, G.; Greggi, S.; Candiani, M.; Lorusso, D. Outcome and risk factors for recurrence in malignant ovarian germ cell tumors: A MITO-9 retrospective study. Int. J. Gynecol. Cancer 2011, 21, 1414–1421. [Google Scholar] [CrossRef] [PubMed]
  15. Burdett, N.L.; Willis, M.O.; Alsop, K.; Hunt, A.L.; Pandey, A.; Hamilton, P.T.; Abulez, T.; Liu, X.; Hoang, T.; Craig, S.; et al. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer. Nat. Genet. 2023, 55, 437–450. [Google Scholar] [CrossRef]
  16. Burdett, N.L.; Willis, M.O.; Pandey, A.; Twomey, L.; Alaei, S.; Australian Ovarian Cancer Study, G.; Bowtell, D.D.L.; Christie, E.L. Timing of whole genome duplication is associated with tumor-specific MHC-II depletion in serous ovarian cancer. Nat. Commun. 2024, 15, 6069. [Google Scholar] [CrossRef]
  17. Chapel, D.B.; Joseph, N.M.; Krausz, T.; Lastra, R.R. An Ovarian Adenocarcinoma With Combined Low-grade Serous and Mesonephric Morphologies Suggests a Mullerian Origin for Some Mesonephric Carcinomas. Int. J. Gynecol. Pathol. 2018, 37, 448–459. [Google Scholar] [CrossRef] [PubMed]
  18. Cheng, E.J.; Kurman, R.J.; Wang, M.; Oldt, R.; Wang, B.G.; Berman, D.M.; Shih Ie, M. Molecular genetic analysis of ovarian serous cystadenomas. Lab. Investig. 2004, 84, 778–784. [Google Scholar] [CrossRef]
  19. Grisham, R.N.; Iyer, G.; Garg, K.; Delair, D.; Hyman, D.M.; Zhou, Q.; Iasonos, A.; Berger, M.F.; Dao, F.; Spriggs, D.R.; et al. BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer 2013, 119, 548–554. [Google Scholar] [CrossRef]
  20. Huang, H.; Keathley, R.; Kim, U.; Cardenas, H.; Xie, P.; Wei, J.; Lengyel, E.; Nephew, K.P.; Zhao, G.; Fu, Z.; et al. Comparative transcriptomic, epigenomic and immunological analyses identify drivers of disparity in high-grade serous ovarian cancer. NPJ Genom. Med. 2024, 9, 64. [Google Scholar] [CrossRef]
  21. Hunter, S.M.; Anglesio, M.S.; Sharma, R.; Gilks, C.B.; Melnyk, N.; Chiew, Y.E.; deFazio, A.; Australian Ovarian Cancer Study Group; Longacre, T.A.; Huntsman, D.G.; et al. Copy number aberrations in benign serous ovarian tumors: A case for reclassification? Clin. Cancer Res. 2011, 17, 7273–7282. [Google Scholar] [CrossRef]
  22. Hussein, Y.R.; Ducie, J.A.; Arnold, A.G.; Kauff, N.D.; Vargas-Alvarez, H.A.; Sala, E.; Levine, D.A.; Soslow, R.A. Invasion Patterns of Metastatic Extrauterine High-grade Serous Carcinoma With BRCA Germline Mutation and Correlation With Clinical Outcomes. Am. J. Surg. Pathol. 2016, 40, 404–409. [Google Scholar] [CrossRef]
  23. Jones, S.; Wang, T.L.; Kurman, R.J.; Nakayama, K.; Velculescu, V.E.; Vogelstein, B.; Kinzler, K.W.; Papadopoulos, N.; Shih Ie, M. Low-grade serous carcinomas of the ovary contain very few point mutations. J. Pathol. 2012, 226, 413–420. [Google Scholar] [CrossRef] [PubMed]
  24. Kelliher, L.; Yoeli-Bik, R.; Schweizer, L.; Lengyel, E. Molecular changes driving low-grade serous ovarian cancer and implications for treatment. Int. J. Gynecol. Cancer 2024, 34, 1630–1638. [Google Scholar] [CrossRef] [PubMed]
  25. Malpica, A.; Wong, K.K. The molecular pathology of ovarian serous borderline tumors. Ann. Oncol. 2016, 27, i16–i19. [Google Scholar] [CrossRef]
  26. McCluggage, W.G.; Vosmikova, H.; Laco, J. Ovarian Combined Low-grade Serous and Mesonephric-like Adenocarcinoma: Further Evidence for A Mullerian Origin of Mesonephric-like Adenocarcinoma. Int. J. Gynecol. Pathol. 2020, 39, 84–92. [Google Scholar] [CrossRef]
  27. Murali, R.; Selenica, P.; Brown, D.N.; Cheetham, R.K.; Chandramohan, R.; Claros, N.L.; Bouvier, N.; Cheng, D.T.; Soslow, R.A.; Weigelt, B.; et al. Somatic genetic alterations in synchronous and metachronous low-grade serous tumours and high-grade carcinomas of the adnexa. Histopathology 2019, 74, 638–650. [Google Scholar] [CrossRef] [PubMed]
  28. Wong, K.K.; Tsang, Y.T.; Deavers, M.T.; Mok, S.C.; Zu, Z.; Sun, C.; Malpica, A.; Wolf, J.K.; Lu, K.H.; Gershenson, D.M. BRAF mutation is rare in advanced-stage low-grade ovarian serous carcinomas. Am. J. Pathol. 2010, 177, 1611–1617. [Google Scholar] [CrossRef]
  29. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011, 474, 609–615. [Google Scholar] [CrossRef]
  30. Veneziani, A.C.; Gonzalez-Ochoa, E.; Alqaisi, H.; Madariaga, A.; Bhat, G.; Rouzbahman, M.; Sneha, S.; Oza, A.M. Heterogeneity and treatment landscape of ovarian carcinoma. Nat. Rev. Clin. Oncol. 2023, 20, 820–842. [Google Scholar] [CrossRef]
  31. Ryland, G.L.; Hunter, S.M.; Doyle, M.A.; Caramia, F.; Li, J.; Rowley, S.M.; Christie, M.; Allan, P.E.; Stephens, A.N.; Bowtell, D.D.; et al. Mutational landscape of mucinous ovarian carcinoma and its neoplastic precursors. Genome. Med. 2015, 7, 87. [Google Scholar] [CrossRef]
  32. Seidman, J.D.; Khedmati, F. Exploring the histogenesis of ovarian mucinous and transitional cell (Brenner) neoplasms and their relationship with Walthard cell nests: A study of 120 tumors. Arch. Pathol. Lab. Med. 2008, 132, 1753–1760. [Google Scholar] [CrossRef]
  33. Simons, M.; Simmer, F.; Bulten, J.; Ligtenberg, M.J.; Hollema, H.; van Vliet, S.; de Voer, R.M.; Kamping, E.J.; van Essen, D.F.; Ylstra, B.; et al. Two types of primary mucinous ovarian tumors can be distinguished based on their origin. Mod. Pathol. 2020, 33, 722–733. [Google Scholar] [CrossRef] [PubMed]
  34. Wu, C.H.; Mao, T.L.; Vang, R.; Ayhan, A.; Wang, T.L.; Kurman, R.J.; Shih Ie, M. Endocervical-type mucinous borderline tumors are related to endometrioid tumors based on mutation and loss of expression of ARID1A. Int. J. Gynecol. Pathol. 2012, 31, 297–303. [Google Scholar] [CrossRef]
  35. McConechy, M.K.; Ding, J.; Senz, J.; Yang, W.; Melnyk, N.; Tone, A.A.; Prentice, L.M.; Wiegand, K.C.; McAlpine, J.N.; Shah, S.P.; et al. Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles. Mod. Pathol. 2014, 27, 128–134. [Google Scholar] [CrossRef]
  36. Euscher, E.D.; Bassett, R.; Duose, D.Y.; Lan, C.; Wistuba, I.; Ramondetta, L.; Ramalingam, P.; Malpica, A. Mesonephric-like Carcinoma of the Endometrium: A Subset of Endometrial Carcinoma With an Aggressive Behavior. Am. J. Surg. Pathol. 2020, 44, 429–443. [Google Scholar] [CrossRef]
  37. Nagamani, M.; Stuart, C.A.; Doherty, M.G. Increased steroid production by the ovarian stromal tissue of postmenopausal women with endometrial cancer. J. Clin. Endocrinol. Metab. 1992, 74, 172–176. [Google Scholar] [CrossRef]
  38. Noe, M.; Ayhan, A.; Wang, T.L.; Shih, I.M. Independent development of endometrial epithelium and stroma within the same endometriosis. J. Pathol. 2018, 245, 265–269. [Google Scholar] [CrossRef] [PubMed]
  39. Parra-Herran, C.; Lerner-Ellis, J.; Xu, B.; Khalouei, S.; Bassiouny, D.; Cesari, M.; Ismiil, N.; Nofech-Mozes, S. Molecular-based classification algorithm for endometrial carcinoma categorizes ovarian endometrioid carcinoma into prognostically significant groups. Mod. Pathol. 2017, 30, 1748–1759. [Google Scholar] [CrossRef] [PubMed]
  40. Sato, N.; Tsunoda, H.; Nishida, M.; Morishita, Y.; Takimoto, Y.; Kubo, T.; Noguchi, M. Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: Possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary. Cancer Res. 2000, 60, 7052–7056. [Google Scholar] [PubMed]
  41. Bennett, J.A.; Morales-Oyarvide, V.; Campbell, S.; Longacre, T.A.; Oliva, E. Mismatch Repair Protein Expression in Clear Cell Carcinoma of the Ovary: Incidence and Morphologic Associations in 109 Cases. Am. J. Surg. Pathol. 2016, 40, 656–663. [Google Scholar] [CrossRef]
  42. Campbell, I.G.; Russell, S.E.; Choong, D.Y.; Montgomery, K.G.; Ciavarella, M.L.; Hooi, C.S.; Cristiano, B.E.; Pearson, R.B.; Phillips, W.A. Mutation of the PIK3CA gene in ovarian and breast cancer. Cancer Res. 2004, 64, 7678–7681. [Google Scholar] [CrossRef]
  43. Friedlander, M.L.; Russell, K.; Millis, S.; Gatalica, Z.; Bender, R.; Voss, A. Molecular Profiling of Clear Cell Ovarian Cancers: Identifying Potential Treatment Targets for Clinical Trials. Int. J. Gynecol. Cancer 2016, 26, 648–654. [Google Scholar] [CrossRef]
  44. Jones, S.; Wang, T.L.; Shih Ie, M.; Mao, T.L.; Nakayama, K.; Roden, R.; Glas, R.; Slamon, D.; Diaz, L.A., Jr.; Vogelstein, B.; et al. Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 2010, 330, 228–231. [Google Scholar] [CrossRef]
  45. Karnezis, A.N.; Wang, Y.; Ramos, P.; Hendricks, W.P.; Oliva, E.; D’Angelo, E.; Prat, J.; Nucci, M.R.; Nielsen, T.O.; Chow, C.; et al. Dual loss of the SWI/SNF complex ATPases SMARCA4/BRG1 and SMARCA2/BRM is highly sensitive and specific for small cell carcinoma of the ovary, hypercalcaemic type. J. Pathol. 2016, 238, 389–400. [Google Scholar] [CrossRef] [PubMed]
  46. Kuo, K.T.; Mao, T.L.; Jones, S.; Veras, E.; Ayhan, A.; Wang, T.L.; Glas, R.; Slamon, D.; Velculescu, V.E.; Kuman, R.J.; et al. Frequent activating mutations of PIK3CA in ovarian clear cell carcinoma. Am. J. Pathol. 2009, 174, 1597–1601. [Google Scholar] [CrossRef] [PubMed]
  47. Lu, F.I.; Gilks, C.B.; Mulligan, A.M.; Ryan, P.; Allo, G.; Sy, K.; Shaw, P.A.; Pollett, A.; Clarke, B.A. Prevalence of loss of expression of DNA mismatch repair proteins in primary epithelial ovarian tumors. Int. J. Gynecol. Pathol. 2012, 31, 524–531. [Google Scholar] [CrossRef] [PubMed]
  48. Parra-Herran, C.; Bassiouny, D.; Lerner-Ellis, J.; Olkhov-Mitsel, E.; Ismiil, N.; Hogen, L.; Vicus, D.; Nofech-Mozes, S. p53, Mismatch Repair Protein, and POLE Abnormalities in Ovarian Clear Cell Carcinoma: An Outcome-based Clinicopathologic Analysis. Am. J. Surg. Pathol. 2019, 43, 1591–1599. [Google Scholar] [CrossRef]
  49. Takenaka, M.; Kobel, M.; Garsed, D.W.; Fereday, S.; Pandey, A.; Etemadmoghadam, D.; Hendley, J.; Kawabata, A.; Noguchi, D.; Yanaihara, N.; et al. Survival Following Chemotherapy in Ovarian Clear Cell Carcinoma Is Not Associated with Pathological Misclassification of Tumor Histotype. Clin. Cancer Res. 2019, 25, 3962–3973. [Google Scholar] [CrossRef]
  50. Wang, Y.K.; Bashashati, A.; Anglesio, M.S.; Cochrane, D.R.; Grewal, D.S.; Ha, G.; McPherson, A.; Horlings, H.M.; Senz, J.; Prentice, L.M.; et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat. Genet. 2017, 49, 856–865. [Google Scholar] [CrossRef]
  51. Kurman, R.J.; Shih Ie, M. The origin and pathogenesis of epithelial ovarian cancer: A proposed unifying theory. Am. J. Surg. Pathol. 2010, 34, 433–443. [Google Scholar] [CrossRef]
  52. Wang, T.; Huang, W.; Finkelman, B.S.; Zhang, H. Malignant Brenner tumor of the ovary: Pathologic evaluation, molecular insights and clinical management. Hum. Pathol. Rep. 2024, 36, 300739. [Google Scholar] [CrossRef]
  53. Ramphal, K.; Hadfield, M.J.; Bandera, C.M.; Hart, J.; Dizon, D.S. Genomic and Molecular Characteristics of Ovarian Carcinosarcoma. Am. J. Clin. Oncol. 2023, 46, 572–576. [Google Scholar] [CrossRef] [PubMed]
  54. Mbatani, N.; Olawaiye, A.B.; Prat, J. Uterine sarcomas. Int. J. Gynaecol. Obstet. 2018, 143, 51–58. [Google Scholar] [CrossRef] [PubMed]
  55. da Silva, E.M.; Fix, D.J.; Sebastiao, A.P.M.; Selenica, P.; Ferrando, L.; Kim, S.H.; Stylianou, A.; Da Cruz Paula, A.; Pareja, F.; Smith, E.S.; et al. Mesonephric and mesonephric-like carcinomas of the female genital tract: Molecular characterization including cases with mixed histology and matched metastases. Mod. Pathol. 2021, 34, 1570–1587. [Google Scholar] [CrossRef]
  56. Ariyoshi, K.; Kawauchi, S.; Kaku, T.; Nakano, H.; Tsuneyoshi, M. Prognostic factors in ovarian carcinosarcoma: A clinicopathological and immunohistochemical analysis of 23 cases. Histopathology 2000, 37, 427–436. [Google Scholar] [CrossRef]
  57. Dasgupta, S.; Bose, D.; Bhattacharyya, N.K.; Biswas, P.K. Carcinosarcoma of ovary with its various immunohistochemical expression: Report of a rare case. J. Cancer Res. Ther. 2015, 11, 1022. [Google Scholar] [CrossRef]
  58. Jones, S.; Stransky, N.; McCord, C.L.; Cerami, E.; Lagowski, J.; Kelly, D.; Angiuoli, S.V.; Sausen, M.; Kann, L.; Shukla, M.; et al. Genomic analyses of gynaecologic carcinosarcomas reveal frequent mutations in chromatin remodelling genes. Nat. Commun. 2014, 5, 5006. [Google Scholar] [CrossRef]
  59. Sawada, M.; Tsuda, H.; Kimura, M.; Okamoto, S.; Kita, T.; Kasamatsu, T.; Yamada, T.; Kikuchi, Y.; Honjo, H.; Matsubara, O. Different expression patterns of KIT, EGFR, and HER-2 (c-erbB-2) oncoproteins between epithelial and mesenchymal components in uterine carcinosarcoma. Cancer Sci. 2003, 94, 986–991. [Google Scholar] [CrossRef] [PubMed]
  60. Zhao, S.; Bellone, S.; Lopez, S.; Thakral, D.; Schwab, C.; English, D.P.; Black, J.; Cocco, E.; Choi, J.; Zammataro, L.; et al. Mutational landscape of uterine and ovarian carcinosarcomas implicates histone genes in epithelial-mesenchymal transition. Proc. Natl. Acad. Sci. USA 2016, 113, 12238–12243. [Google Scholar] [CrossRef]
  61. Zheng, J.; Tang, C.; Liu, P.; Hao, H. Carcinosarcoma of the ovary: A case report and literature review. Front. Oncol. 2023, 13, 1278300. [Google Scholar] [CrossRef]
  62. Clement, P.B.; Scully, R.E. Uterine tumors with mixed epithelial and mesenchymal elements. Semin. Diagn. Pathol. 1988, 5, 199–222. [Google Scholar]
  63. Liu, F.S.; Kohler, M.F.; Marks, J.R.; Bast, R.C., Jr.; Boyd, J.; Berchuck, A. Mutation and overexpression of the p53 tumor suppressor gene frequently occurs in uterine and ovarian sarcomas. Obstet. Gynecol. 1994, 83, 118–124. [Google Scholar]
  64. McCluggage, W.G.; Singh, N.; Gilks, C.B. Key changes to the World Health Organization (WHO) classification of female genital tumours introduced in the 5th edition (2020). Histopathology 2022, 80, 762–778. [Google Scholar] [CrossRef]
  65. Al-Agha, O.M.; Huwait, H.F.; Chow, C.; Yang, W.; Senz, J.; Kalloger, S.E.; Huntsman, D.G.; Young, R.H.; Gilks, C.B. FOXL2 is a sensitive and specific marker for sex cord-stromal tumors of the ovary. Am. J. Surg. Pathol. 2011, 35, 484–494. [Google Scholar] [CrossRef]
  66. Buza, N.; Wong, S.; Hui, P. FOXL2 Mutation Analysis of Ovarian Sex Cord-Stromal Tumors: Genotype-Phenotype Correlation With Diagnostic Considerations. Int. J. Gynecol. Pathol. 2018, 37, 305–315. [Google Scholar] [CrossRef] [PubMed]
  67. Conlon, N.; Schultheis, A.M.; Piscuoglio, S.; Silva, A.; Guerra, E.; Tornos, C.; Reuter, V.E.; Soslow, R.A.; Young, R.H.; Oliva, E.; et al. A survey of DICER1 hotspot mutations in ovarian and testicular sex cord-stromal tumors. Mod. Pathol. 2015, 28, 1603–1612. [Google Scholar] [CrossRef] [PubMed]
  68. Goulvent, T.; Ray-Coquard, I.; Borel, S.; Haddad, V.; Devouassoux-Shisheboran, M.; Vacher-Lavenu, M.C.; Pujade-Laurraine, E.; Savina, A.; Maillet, D.; Gillet, G.; et al. DICER1 and FOXL2 mutations in ovarian sex cord-stromal tumours: A GINECO Group study. Histopathology 2016, 68, 279–285. [Google Scholar] [CrossRef]
  69. Michal, M.; Vanecek, T.; Sima, R.; Mukensnabl, P.; Hes, O.; Kazakov, D.V.; Matoska, J.; Zuntova, A.; Dvorak, V.; Talerman, A. Mixed germ cell sex cord-stromal tumors of the testis and ovary. Morphological, immunohistochemical, and molecular genetic study of seven cases. Virchows Arch. 2006, 448, 612–622. [Google Scholar] [CrossRef] [PubMed]
  70. de Kock, L.; Terzic, T.; McCluggage, W.G.; Stewart, C.J.R.; Shaw, P.; Foulkes, W.D.; Clarke, B.A. DICER1 Mutations Are Consistently Present in Moderately and Poorly Differentiated Sertoli-Leydig Cell Tumors. Am. J. Surg. Pathol. 2017, 41, 1178–1187. [Google Scholar] [CrossRef]
  71. Devouassoux-Shisheboran, M.; Silver, S.A.; Tavassoli, F.A. Wolffian adnexal tumor, so-called female adnexal tumor of probable Wolffian origin (FATWO): Immunohistochemical evidence in support of a Wolffian origin. Hum. Pathol. 1999, 30, 856–863. [Google Scholar] [CrossRef]
  72. Fumagalli, D.; Jayraj, A.; Olearo, E.; Capasso, I.; Hsu, H.C.; Tzur, Y.; Piedimonte, S.; Jugeli, B.; Santana, B.N.; De Vitis, L.A.; et al. Primary versus interval cytoreductive surgery in patients with rare epithelial or non-epithelial ovarian cancer. Int. J. Gynecol. Cancer 2025, 35, 101664. [Google Scholar] [CrossRef] [PubMed]
  73. Heravi-Moussavi, A.; Anglesio, M.S.; Cheng, S.W.; Senz, J.; Yang, W.; Prentice, L.; Fejes, A.P.; Chow, C.; Tone, A.; Kalloger, S.E.; et al. Recurrent somatic DICER1 mutations in nonepithelial ovarian cancers. N. Engl. J. Med. 2012, 366, 234–242. [Google Scholar] [CrossRef]
  74. Karnezis, A.N.; Wang, Y.; Keul, J.; Tessier-Cloutier, B.; Magrill, J.; Kommoss, S.; Senz, J.; Yang, W.; Proctor, L.; Schmidt, D.; et al. DICER1 and FOXL2 Mutation Status Correlates With Clinicopathologic Features in Ovarian Sertoli-Leydig Cell Tumors. Am. J. Surg. Pathol. 2019, 43, 628–638. [Google Scholar] [CrossRef] [PubMed]
  75. Kim, S.H.; Da Cruz Paula, A.; Basili, T.; Dopeso, H.; Bi, R.; Pareja, F.; da Silva, E.M.; Gularte-Merida, R.; Sun, Z.; Fujisawa, S.; et al. Identification of recurrent FHL2-GLI2 oncogenic fusion in sclerosing stromal tumors of the ovary. Nat. Commun. 2020, 11, 44. [Google Scholar] [CrossRef] [PubMed]
  76. Meurgey, A.; Descotes, F.; Mery-Lamarche, E.; Devouassoux-Shisheboran, M. Lack of mutation of DICER1 and FOXL2 genes in microcystic stromal tumor of the ovary. Virchows Arch. 2017, 470, 225–229. [Google Scholar] [CrossRef]
  77. Schultz, K.A.P.; Harris, A.K.; Finch, M.; Dehner, L.P.; Brown, J.B.; Gershenson, D.M.; Young, R.H.; Field, A.; Yu, W.; Turner, J.; et al. DICER1-related Sertoli-Leydig cell tumor and gynandroblastoma: Clinical and genetic findings from the International Ovarian and Testicular Stromal Tumor Registry. Gynecol. Oncol. 2017, 147, 521–527. [Google Scholar] [CrossRef]
  78. Tchrakian, N.; Oliva, E.; Chong, A.S.; Rivera-Polo, B.; Bennett, J.A.; Nucci, M.R.; Sah, S.; Schoolmeester, J.K.; van der Griend, R.A.; Foulkes, W.D.; et al. Ovarian Signet-ring Stromal Tumor: A Morphologic, Immunohistochemical, and Molecular Study of 7 Cases With Discussion of the Differential Diagnosis. Am. J. Surg. Pathol. 2022, 46, 1599–1610. [Google Scholar] [CrossRef]
  79. Zhang, Y.; Tao, L.; Yin, C.; Wang, W.; Zou, H.; Ren, Y.; Liang, W.; Jiang, J.; Zhang, W.; Jia, W.; et al. Ovarian microcystic stromal tumor with undetermined potential: Case study with molecular analysis and literature review. Hum. Pathol. 2018, 78, 171–176. [Google Scholar] [CrossRef]
  80. Rabban, J.T.; Karnezis, A.N.; Devine, W.P. Practical roles for molecular diagnostic testing in ovarian adult granulosa cell tumour, Sertoli-Leydig cell tumour, microcystic stromal tumour and their mimics. Histopathology 2020, 76, 11–24. [Google Scholar] [CrossRef]
  81. Schmidt, J.; Derr, V.; Heinrich, M.C.; Crum, C.P.; Fletcher, J.A.; Corless, C.L.; Nose, V. BRAF in papillary thyroid carcinoma of ovary (struma ovarii). Am. J. Surg. Pathol. 2007, 31, 1337–1343. [Google Scholar] [CrossRef]
  82. Cools, M.; Looijenga, L.H.; Wolffenbuttel, K.P.; Drop, S.L. Disorders of sex development: Update on the genetic background, terminology and risk for the development of germ cell tumors. World J. Pediatr. 2009, 5, 93–102. [Google Scholar] [CrossRef]
  83. Riopel, M.A.; Spellerberg, A.; Griffin, C.A.; Perlman, E.J. Genetic analysis of ovarian germ cell tumors by comparative genomic hybridization. Cancer Res. 1998, 58, 3105–3110. [Google Scholar] [PubMed]
  84. Stoop, H.; Honecker, F.; van de Geijn, G.J.; Gillis, A.J.; Cools, M.C.; de Boer, M.; Bokemeyer, C.; Wolffenbuttel, K.P.; Drop, S.L.; de Krijger, R.R.; et al. Stem cell factor as a novel diagnostic marker for early malignant germ cells. J. Pathol. 2008, 216, 43–54. [Google Scholar] [CrossRef]
  85. Cheng, L.; Roth, L.M.; Zhang, S.; Wang, M.; Morton, M.J.; Zheng, W.; Abdul Karim, F.W.; Montironi, R.; Lopez-Beltran, A. KIT gene mutation and amplification in dysgerminoma of the ovary. Cancer 2011, 117, 2096–2103. [Google Scholar] [CrossRef] [PubMed]
  86. Cheng, L.; Zhang, S.; Talerman, A.; Roth, L.M. Morphologic, immunohistochemical, and fluorescence in situ hybridization study of ovarian embryonal carcinoma with comparison to solid variant of yolk sac tumor and immature teratoma. Hum. Pathol. 2010, 41, 716–723. [Google Scholar] [CrossRef] [PubMed]
  87. Cossu-Rocca, P.; Zhang, S.; Roth, L.M.; Eble, J.N.; Zheng, W.; Karim, F.W.; Michael, H.; Emerson, R.E.; Jones, T.D.; Hattab, E.M.; et al. Chromosome 12p abnormalities in dysgerminoma of the ovary: A FISH analysis. Mod. Pathol. 2006, 19, 611–615. [Google Scholar] [CrossRef]
  88. Hersmus, R.; Kalfa, N.; de Leeuw, B.; Stoop, H.; Oosterhuis, J.W.; de Krijger, R.; Wolffenbuttel, K.P.; Drop, S.L.; Veitia, R.A.; Fellous, M.; et al. FOXL2 and SOX9 as parameters of female and male gonadal differentiation in patients with various forms of disorders of sex development (DSD). J. Pathol. 2008, 215, 31–38. [Google Scholar] [CrossRef]
  89. Liang, L.; Zhang, Y.; Malpica, A.; Ramalingam, P.; Euscher, E.D.; Fuller, G.N.; Liu, J. Gliomatosis peritonei: A clinicopathologic and immunohistochemical study of 21 cases. Mod. Pathol. 2015, 28, 1613–1620. [Google Scholar] [CrossRef]
  90. Poulos, C.; Cheng, L.; Zhang, S.; Gersell, D.J.; Ulbright, T.M. Analysis of ovarian teratomas for isochromosome 12p: Evidence supporting a dual histogenetic pathway for teratomatous elements. Mod. Pathol. 2006, 19, 766–771. [Google Scholar] [CrossRef]
  91. Snir, O.L.; DeJoseph, M.; Wong, S.; Buza, N.; Hui, P. Frequent homozygosity in both mature and immature ovarian teratomas: A shared genetic basis of tumorigenesis. Mod. Pathol. 2017, 30, 1467–1475. [Google Scholar] [CrossRef]
  92. Witkowski, L.; Mattina, J.; Schonberger, S.; Murray, M.J.; Choong, C.S.; Huntsman, D.G.; Reis-Filho, J.S.; McCluggage, W.G.; Nicholson, J.C.; Coleman, N.; et al. DICER1 hotspot mutations in non-epithelial gonadal tumours. Br. J. Cancer 2013, 109, 2744–2750. [Google Scholar] [CrossRef]
  93. Kupryjanczyk, J.; Dansonka-Mieszkowska, A.; Moes-Sosnowska, J.; Plisiecka-Halasa, J.; Szafron, L.; Podgorska, A.; Rzepecka, I.K.; Konopka, B.; Budzilowska, A.; Rembiszewska, A.; et al. Ovarian small cell carcinoma of hypercalcemic type—Evidence of germline origin and SMARCA4 gene inactivation. a pilot study. Pol. J. Pathol. 2013, 64, 238–246. [Google Scholar] [CrossRef] [PubMed]
  94. Ramachandra, C.A.; Attili, V.S.S.; Dadhich, H.P.; Kumari, A.; Appaji, L.; Giri, G.V.; Mukharjee, G. Extrarenal Wilms’ tumor: A report of two cases and review of literature. J. Indian Assoc. Pediatr. Surg. 2007, 12, 145–147. [Google Scholar] [CrossRef]
  95. Kominami, A.; Fujino, M.; Murakami, H.; Ito, M. beta-catenin mutation in ovarian solid pseudopapillary neoplasm. Pathol. Int. 2014, 64, 460–464. [Google Scholar] [CrossRef]
  96. Singh, K.; Patel, N.; Patil, P.; Paquette, C.; Mathews, C.A.; Lawrence, W.D. Primary Ovarian Solid Pseudopapillary Neoplasm With CTNNB1 c.98C>G (p.S33C) Point Mutation. Int. J. Gynecol. Pathol. 2018, 37, 110–116. [Google Scholar] [CrossRef]
  97. Jelinic, P.; Mueller, J.J.; Olvera, N.; Dao, F.; Scott, S.N.; Shah, R.; Gao, J.; Schultz, N.; Gonen, M.; Soslow, R.A.; et al. Recurrent SMARCA4 mutations in small cell carcinoma of the ovary. Nat. Genet. 2014, 46, 424–426. [Google Scholar] [CrossRef]
  98. Ramos, P.; Karnezis, A.N.; Craig, D.W.; Sekulic, A.; Russell, M.L.; Hendricks, W.P.; Corneveaux, J.J.; Barrett, M.T.; Shumansky, K.; Yang, Y.; et al. Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4. Nat. Genet. 2014, 46, 427–429. [Google Scholar] [CrossRef] [PubMed]
  99. Witkowski, L.; Carrot-Zhang, J.; Albrecht, S.; Fahiminiya, S.; Hamel, N.; Tomiak, E.; Grynspan, D.; Saloustros, E.; Nadaf, J.; Rivera, B.; et al. Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type. Nat. Genet. 2014, 46, 438–443. [Google Scholar] [CrossRef]
  100. Fahiminiya, S.; Witkowski, L.; Nadaf, J.; Carrot-Zhang, J.; Goudie, C.; Hasselblatt, M.; Johann, P.; Kool, M.; Lee, R.S.; Gayden, T.; et al. Molecular analyses reveal close similarities between small cell carcinoma of the ovary, hypercalcemic type and atypical teratoid/rhabdoid tumor. Oncotarget 2016, 7, 1732–1740. [Google Scholar] [CrossRef] [PubMed]
  101. Witkowski, L.; Goudie, C.; Ramos, P.; Boshari, T.; Brunet, J.S.; Karnezis, A.N.; Longy, M.; Knost, J.A.; Saloustros, E.; McCluggage, W.G.; et al. The influence of clinical and genetic factors on patient outcome in small cell carcinoma of the ovary, hypercalcemic type. Gynecol. Oncol. 2016, 141, 454–460. [Google Scholar] [CrossRef]
  102. Holdhof, D.; Johann, P.D.; Spohn, M.; Bockmayr, M.; Safaei, S.; Joshi, P.; Masliah-Planchon, J.; Ho, B.; Andrianteranagna, M.; Bourdeaut, F.; et al. Atypical teratoid/rhabdoid tumors (ATRTs) with SMARCA4 mutation are molecularly distinct from SMARCB1-deficient cases. Acta Neuropathol. 2021, 141, 291–301. [Google Scholar] [CrossRef]
  103. Kommoss, F.K.; Tessier-Cloutier, B.; Witkowski, L.; Forgo, E.; Koelsche, C.; Kobel, M.; Foulkes, W.D.; Lee, C.H.; Kolin, D.L.; von Deimling, A.; et al. Cellular context determines DNA methylation profiles in SWI/SNF-deficient cancers of the gynecologic tract. J. Pathol. 2022, 257, 140–145. [Google Scholar] [CrossRef] [PubMed]
  104. Ben-Rafael, Z.; Bider, D.; Menashe, Y.; Maymon, R.; Zolti, M.; Mashiach, S. Follicular and luteal cysts after treatment with gonadotropin-releasing hormone analog for in vitro fertilization. Fertil. Steril. 1990, 53, 1091–1094. [Google Scholar] [CrossRef] [PubMed]
  105. Bailey, M.H.; Tokheim, C.; Porta-Pardo, E.; Sengupta, S.; Bertrand, D.; Weerasinghe, A.; Colaprico, A.; Wendl, M.C.; Kim, J.; Reardon, B.; et al. Comprehensive Characterization of Cancer Driver Genes and Mutations. Cell 2018, 173, 371–385.e18. [Google Scholar] [CrossRef]
  106. Zehir, A.; Benayed, R.; Shah, R.H.; Syed, A.; Middha, S.; Kim, H.R.; Srinivasan, P.; Gao, J.; Chakravarty, D.; Devlin, S.M.; et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017, 23, 703–713. [Google Scholar] [CrossRef]
  107. Manning-Geist, B.; Gordhandas, S.; Liu, Y.L.; Zhou, Q.; Iasonos, A.; Da Cruz Paula, A.; Mandelker, D.; Long Roche, K.; Zivanovic, O.; Maio, A.; et al. MAPK Pathway Genetic Alterations Are Associated with Prolonged Overall Survival in Low-Grade Serous Ovarian Carcinoma. Clin. Cancer Res. 2022, 28, 4456–4465. [Google Scholar] [CrossRef]
  108. Elkin, R.; Oh, J.H.; Liu, Y.L.; Selenica, P.; Weigelt, B.; Reis-Filho, J.S.; Zamarin, D.; Deasy, J.O.; Norton, L.; Levine, A.J.; et al. Geometric network analysis provides prognostic information in patients with high grade serous carcinoma of the ovary treated with immune checkpoint inhibitors. NPJ Genom. Med. 2021, 6, 99. [Google Scholar] [CrossRef] [PubMed]
  109. Matuszczak, M.; Kiljanczyk, A.; Marciniak, W.; Derkacz, R.; Stempa, K.; Baszuk, P.; Bryskiewicz, M.; Cybulski, C.; Debniak, T.; Jacek, G.; et al. Blood molybdenum level as a marker of cancer risk on BRCA1 carriers. Hered. Cancer Clin. Pract. 2024, 22, 19. [Google Scholar] [CrossRef]
  110. Singh, K.A.; Soukar, J.; Zulkifli, M.; Kersey, A.; Lokhande, G.; Ghosh, S.; Murali, A.; Garza, N.M.; Kaur, H.; Keeney, J.N.; et al. Atomic vacancies of molybdenum disulfide nanoparticles stimulate mitochondrial biogenesis. Nat. Commun. 2024, 15, 8136. [Google Scholar] [CrossRef]
  111. Giornelli, G.H. Management of relapsed ovarian cancer: A review. Springerplus 2016, 5, 1197. [Google Scholar] [CrossRef]
  112. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef]
  113. Ray-Coquard, I.; Leary, A.; Pignata, S.; Cropet, C.; Gonzalez-Martin, A.; Marth, C.; Nagao, S.; Vergote, I.; Colombo, N.; Maenpaa, J.; et al. Olaparib plus bevacizumab first-line maintenance in ovarian cancer: Final overall survival results from the PAOLA-1/ENGOT-ov25 trial. Ann. Oncol. 2023, 34, 681–692. [Google Scholar] [CrossRef]
  114. DiSilvestro, P.; Banerjee, S.; Colombo, N.; Scambia, G.; Kim, B.G.; Oaknin, A.; Friedlander, M.; Lisyanskaya, A.; Floquet, A.; Leary, A.; et al. Overall Survival With Maintenance Olaparib at a 7-Year Follow-Up in Patients With Newly Diagnosed Advanced Ovarian Cancer and a BRCA Mutation: The SOLO1/GOG 3004 Trial. J. Clin. Oncol. 2023, 41, 609–617. [Google Scholar] [CrossRef]
  115. Cuenca-Escalona, J.; Bodder, J.; Subtil, B.; Sanchez-Sanchez, M.; Vidal-Manrique, M.; Sweep, M.W.D.; Fauerbach, J.A.; Cambi, A.; Florez-Grau, G.; de Vries, J.M. EP2/EP4 targeting prevents tumor-derived PGE2-mediated immunosuppression in cDC2s. J. Leukoc. Biol. 2024, 116, 1554–1567. [Google Scholar] [CrossRef]
  116. Chaudhry, S.; Thomas, S.N.; Simmons, G.E., Jr. Targeting lipid metabolism in the treatment of ovarian cancer. Oncotarget 2022, 13, 768–783. [Google Scholar] [CrossRef]
  117. Surendran, A.; Jamalkhah, M.; Poutou, J.; Birtch, R.; Lawson, C.; Dave, J.; Crupi, M.J.F.; Mayer, J.; Taylor, V.; Petryk, J.; et al. Fatty acid transport protein inhibition sensitizes breast and ovarian cancers to oncolytic virus therapy via lipid modulation of the tumor microenvironment. Front. Immunol. 2023, 14, 1099459. [Google Scholar] [CrossRef]
  118. Chen, R.R.; Yung, M.M.H.; Xuan, Y.; Zhan, S.; Leung, L.L.; Liang, R.R.; Leung, T.H.Y.; Yang, H.; Xu, D.; Sharma, R.; et al. Author Correction: Targeting of lipid metabolism with a metabolic inhibitor cocktail eradicates peritoneal metastases in ovarian cancer cells. Commun. Biol. 2025, 8, 294. [Google Scholar] [CrossRef] [PubMed]
  119. He, Z.Y.; Deng, F.; Wei, X.W.; Ma, C.C.; Luo, M.; Zhang, P.; Sang, Y.X.; Liang, X.; Liu, L.; Qin, H.X.; et al. Ovarian cancer treatment with a tumor-targeting and gene expression-controllable lipoplex. Sci. Rep. 2016, 6, 23764. [Google Scholar] [CrossRef] [PubMed]
  120. Zhang, D.; Zhang, B. cGAS/STING signaling pathway in gynecological malignancies: From molecular mechanisms to therapeutic values. Front. Immunol. 2025, 16, 1525736. [Google Scholar] [CrossRef]
  121. Ding, L.; Kim, H.J.; Wang, Q.; Kearns, M.; Jiang, T.; Ohlson, C.E.; Li, B.B.; Xie, S.; Liu, J.F.; Stover, E.H.; et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep. 2018, 25, 2972–2980.e5. [Google Scholar] [CrossRef] [PubMed]
  122. Meng, J.; Peng, J.; Feng, J.; Maurer, J.; Li, X.; Li, Y.; Yao, S.; Chu, R.; Pan, X.; Li, J.; et al. Niraparib exhibits a synergistic anti-tumor effect with PD-L1 blockade by inducing an immune response in ovarian cancer. J. Transl. Med. 2021, 19, 415. [Google Scholar] [CrossRef]
  123. Cornelison, R.; Biswas, K.; Llaneza, D.C.; Harris, A.R.; Sosale, N.G.; Lazzara, M.J.; Landen, C.N. CX-5461 Treatment Leads to Cytosolic DNA-Mediated STING Activation in Ovarian Cancer. Cancers 2021, 13, 5056. [Google Scholar] [CrossRef]
  124. Zhang, J.; Chen, Y.; Chen, X.; Zhang, W.; Zhao, L.; Weng, L.; Tian, H.; Wu, Z.; Tan, X.; Ge, X.; et al. Deubiquitinase USP35 restrains STING-mediated interferon signaling in ovarian cancer. Cell Death Differ. 2021, 28, 139–155. [Google Scholar] [CrossRef]
  125. Xie, W.; Zhang, L.; Shen, J.; Lai, F.; Han, W.; Liu, X. Knockdown of CENPM activates cGAS-STING pathway to inhibit ovarian cancer by promoting pyroptosis. BMC Cancer 2024, 24, 551. [Google Scholar] [CrossRef]
  126. Xu, Y.; Huang, X.; Nie, X.C.; Liu, Y.S.; Zhou, Y.; Niu, J.M. Manganese and IL-12 treatment alters the ovarian tumor microenvironment. Aging 2024, 16, 191–206. [Google Scholar] [CrossRef] [PubMed]
  127. Shen, J.; Xu, F.; Liu, T.; Ye, Y.; Xu, S. NAD(+) Metabolism-Mediated SURF4-STING Axis Enhances T-Cell Anti-Tumor Effects in the Ovarian Cancer Microenvironment. Cell Death Dis. 2025, 16, 640. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, H.; Sun, Y.; Zhang, L.; Shen, M.; Tong, X.; Zhao, D.; Xiao, H.; Li, B. Polymer-PARPi Conjugates Delivering USP1i for Maximizing Synthetic Lethality to Stimulate STING Pathway in High-Grade Serous Ovarian Cancer. Adv. Mater. 2025, e12962. [Google Scholar] [CrossRef] [PubMed]
  129. Chae, C.S.; Teran-Cabanillas, E.; Cubillos-Ruiz, J.R. Dendritic cell rehab: New strategies to unleash therapeutic immunity in ovarian cancer. Cancer Immunol. Immunother. 2017, 66, 969–977. [Google Scholar] [CrossRef]
  130. Galpin, K.J.C.; Rodriguez, G.M.; Maranda, V.; Cook, D.P.; Macdonald, E.; Murshed, H.; Zhao, S.; McCloskey, C.W.; Chruscinski, A.; Levy, G.A.; et al. FGL2 promotes tumour growth and attenuates infiltration of activated immune cells in melanoma and ovarian cancer models. Sci. Rep. 2024, 14, 787. [Google Scholar] [CrossRef]
  131. Zhang, X.; He, T.; Li, Y.; Chen, L.; Liu, H.; Wu, Y.; Guo, H. Dendritic Cell Vaccines in Ovarian Cancer. Front. Immunol. 2020, 11, 613773. [Google Scholar] [CrossRef]
  132. Caro, A.A.; Deschoemaeker, S.; Allonsius, L.; Coosemans, A.; Laoui, D. Dendritic Cell Vaccines: A Promising Approach in the Fight against Ovarian Cancer. Cancers 2022, 14, 4037. [Google Scholar] [CrossRef]
  133. Zhang, L.; Zhao, W.; Huang, J.; Li, F.; Sheng, J.; Song, H.; Chen, Y. Development of a Dendritic Cell/Tumor Cell Fusion Cell Membrane Nano-Vaccine for the Treatment of Ovarian Cancer. Front. Immunol. 2022, 13, 828263. [Google Scholar] [CrossRef]
  134. Zhang, X.; Tang, D.; Xiao, H.; Li, B.; Shang, K.; Zhao, D. Activating the cGAS-STING Pathway by Manganese-Based Nanoparticles Combined with Platinum-Based Nanoparticles for Enhanced Ovarian Cancer Immunotherapy. ACS Nano 2025, 19, 4346–4365. [Google Scholar] [CrossRef] [PubMed]
  135. Puttock, E.H.; Tyler, E.J.; Manni, M.; Maniati, E.; Butterworth, C.; Burger Ramos, M.; Peerani, E.; Hirani, P.; Gauthier, V.; Liu, Y.; et al. Extracellular matrix educates an immunoregulatory tumor macrophage phenotype found in ovarian cancer metastasis. Nat. Commun. 2023, 14, 2514. [Google Scholar] [CrossRef] [PubMed]
  136. Fang, J.; Wang, J.; Zhao, X.; Yang, Y.; Xiao, Y. KLHDC8A knockdown in normal ovarian epithelial cells promoted the polarization of pro-tumoral macrophages via the C5a/C5aR/p65 NFκB signaling pathway. Cell Immunol. 2025, 409–410, 104913. [Google Scholar] [CrossRef] [PubMed]
  137. Luan, X.; Lei, T.; Fang, J.; Liu, X.; Fu, H.; Li, Y.; Chu, W.; Jiang, P.; Tong, C.; Qi, H.; et al. Blockade of C5a receptor unleashes tumor-associated macrophage antitumor response and enhances CXCL9-dependent CD8(+) T cell activity. Mol. Ther. 2024, 32, 469–489. [Google Scholar] [CrossRef]
  138. Liu, R.; Wei, H.; Gao, P.; Yu, H.; Wang, K.; Fu, Z.; Ju, B.; Zhao, M.; Dong, S.; Li, Z.; et al. CD47 promotes ovarian cancer progression by inhibiting macrophage phagocytosis. Oncotarget 2017, 8, 39021–39032. [Google Scholar] [CrossRef]
  139. Yang, Y.; Wu, H.; Yang, Y.; Kang, Y.; He, R.; Zhou, B.; Guo, H.; Zhang, J.; Li, J.; Ge, C.; et al. Dual blockade of CD47 and CD24 signaling using a novel bispecific antibody fusion protein enhances macrophage immunotherapy. Mol. Ther. Oncolytics 2023, 31, 100747. [Google Scholar] [CrossRef]
  140. Lakhani, N.J.; Stewart, D.; Richardson, D.L.; Dockery, L.E.; Van Le, L.; Call, J.; Rangwala, F.; Wang, G.; Ma, B.; Metenou, S.; et al. First-in-human phase I trial of the bispecific CD47 inhibitor and CD40 agonist Fc-fusion protein, SL-172154 in patients with platinum-resistant ovarian cancer. J. Immunother. Cancer 2025, 13, e010565. [Google Scholar] [CrossRef]
  141. Li, H.; Luo, F.; Jiang, X.; Zhang, W.; Xiang, T.; Pan, Q.; Cai, L.; Zhao, J.; Weng, D.; Li, Y.; et al. CircITGB6 promotes ovarian cancer cisplatin resistance by resetting tumor-associated macrophage polarization toward the M2 phenotype. J. Immunother. Cancer 2022, 10, e004029. [Google Scholar] [CrossRef]
  142. Vledder, A.; Zeeburg, H.v.; Brummel, K.; Eerkens, A.L.; Rooij, N.v.; Plat, A.; Rovers, J.; Bruyn, M.d.; Nijman, H. Successful induction of tumor-directed immune responses in high grade serious ovarian carcinoma patients after primary treatment using a whole tumor cell vaccine. J. Clin. Oncol. 2025, 43, 5566. [Google Scholar] [CrossRef]
  143. Koeneman, B.; Schreibelt, G.; Duiveman-de Boer, T.; Bos, K.; van Oorschot, T.; Pots, J.; Scharenborg, N.; de Boer, A.; Hins-de Bree, S.; de Haas, N.; et al. NEOadjuvant Dendritic cell therapy added to first line standard of care in advanced epithelial Ovarian Cancer (NEODOC): Protocol of a first-in-human, exploratory, single-centre phase I/II trial in the Netherlands. BMJ Open 2025, 15, e102184. [Google Scholar] [CrossRef] [PubMed]
  144. Reiss, K.A.; Angelos, M.G.; Dees, E.C.; Yuan, Y.; Ueno, N.T.; Pohlmann, P.R.; Johnson, M.L.; Chao, J.; Shestova, O.; Serody, J.S.; et al. CAR-macrophage therapy for HER2-overexpressing advanced solid tumors: A phase 1 trial. Nat. Med. 2025, 31, 1171–1182. [Google Scholar] [CrossRef]
  145. Li, X.; Wang, X.; Wang, H.; Zuo, D.; Xu, J.; Feng, Y.; Xue, D.; Zhang, L.; Lin, L.; Zhang, J. A clinical study of autologous chimeric antigen receptor macrophage targeting mesothelin shows safety in ovarian cancer therapy. J. Hematol. Oncol. 2024, 17, 116. [Google Scholar] [CrossRef]
  146. Li, J.; Hu, H.; Lian, H.; Yang, S.; Liu, M.; He, J.; Cao, B.; Chen, D.; Hu, Y.; Zhi, C.; et al. NK-92MI Cells Engineered with Anti-claudin-6 Chimeric Antigen Receptors in Immunotherapy for Ovarian Cancer. Int. J. Biol. Sci. 2024, 20, 1578–1601. [Google Scholar] [CrossRef] [PubMed]
  147. Knisely, A.; Rafei, H.; Basar, R.; Banerjee, P.P.; Metwalli, Z.; Lito, K.; Fellman, B.M.; Yuan, Y.; Wolff, R.A.; Morelli, M.P.; et al. Phase I/II study of TROP2 CAR engineered IL15-transduced cord blood-derived NK cells delivered intraperitoneally for the management of platinum resistant ovarian cancer, mesonephric-like adenocarcinoma, and pancreatic cancer. J. Clin. Oncol. 2024, 42, TPS5626. [Google Scholar] [CrossRef]
  148. VandenHeuvel, S.N.; Chau, E.; Mohapatra, A.; Dabbiru, S.; Roy, S.; O’Connell, C.; Kamat, A.; Godin, B.; Raghavan, S.A. Macrophage Checkpoint Nanoimmunotherapy Has the Potential to Reduce Malignant Progression in Bioengineered In Vitro Models of Ovarian Cancer. ACS Appl. Bio. Mater. 2024, 7, 7871–7882. [Google Scholar] [CrossRef]
  149. Liu, Y.; Yang, J.; Hilliard, T.S.; Wang, Z.; Johnson, J.; Wang, W.; Harper, E.I.; Ott, C.; O’Brien, C.; Campbell, L.; et al. Host obesity alters the ovarian tumor immune microenvironment and impacts response to standard of care chemotherapy. J. Exp. Clin. Cancer Res. 2023, 42, 165. [Google Scholar] [CrossRef]
  150. Luke, J.J.; Barlesi, F.; Chung, K.; Tolcher, A.W.; Kelly, K.; Hollebecque, A.; Le Tourneau, C.; Subbiah, V.; Tsai, F.; Kao, S.; et al. Phase I study of ABBV-428, a mesothelin-CD40 bispecific, in patients with advanced solid tumors. J. Immunother. Cancer 2021, 9, e002015. [Google Scholar] [CrossRef]
  151. Yeku, O.O.; Barve, M.; Tan, W.W.; Wang, J.; Patnaik, A.; LoRusso, P.; Richardson, D.L.; Naqash, A.R.; Lynam, S.K.; Fu, S.; et al. Myeloid targeting antibodies PY159 and PY314 for platinum-resistant ovarian cancer. J. Immunother. Cancer 2025, 13, e010959. [Google Scholar] [CrossRef]
  152. Maas, R.J.A.; Hoogstad-van Evert, J.S.; Hagemans, I.M.; Brummelman, J.; van Ens, D.; de Jonge, P.; Hooijmaijers, L.; Mahajan, S.; van der Waart, A.B.; Hermans, C.; et al. Increased peritoneal TGF-beta1 is associated with ascites-induced NK-cell dysfunction and reduced survival in high-grade epithelial ovarian cancer. Front. Immunol. 2024, 15, 1448041. [Google Scholar] [CrossRef] [PubMed]
  153. Acosta, J.C.; Bahr, J.M.; Basu, S.; O’Donnell, J.T.; Barua, A. Expression of CISH, an Inhibitor of NK Cell Function, Increases in Association with Ovarian Cancer Development and Progression. Biomedicines 2023, 11, 299. [Google Scholar] [CrossRef]
  154. Sandoval, T.A.; Salvagno, C.; Chae, C.S.; Awasthi, D.; Giovanelli, P.; Marin Falco, M.; Hwang, S.M.; Teran-Cabanillas, E.; Suominen, L.; Yamazaki, T.; et al. Iron Chelation Therapy Elicits Innate Immune Control of Metastatic Ovarian Cancer. Cancer Discov. 2024, 14, 1901–1921. [Google Scholar] [CrossRef]
  155. Tarannum, M.A.-O.; Dinh, K.A.-O.X.; Vergara, J.A.-O.; Birch, G.; Abdulhamid, Y.A.-O.; Kaplan, I.A.-O.; Ay, O.; Maia, A.A.-O.; Beaver, O.; Sheffer, M.A.-O.; et al. CAR memory-like NK cells targeting the membrane proximal domain of mesothelin demonstrate promising activity in ovarian cancer. Sci. Adv. 2024, 10, eadn0881. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, Y.; Zhang, M.; Shen, X.; Xia, C.; Hu, F.; Huang, D.; Weng, Q.; Zhang, Q.; Liu, L.; Zhu, Y.; et al. Mesothelin CAR-engineered NK cells derived from human embryonic stem cells suppress the progression of human ovarian cancer in animals. Cell Prolif. 2024, 57, e13727. [Google Scholar] [CrossRef]
  157. Shalaby, N.; Xia, Y.; Kelly, J.J.; Sanchez-Pupo, R.; Martinez, F.; Fox, M.S.; Thiessen, J.D.; Hicks, J.W.; Scholl, T.J.; Ronald, J.A. Imaging CAR-NK cells targeted to HER2 ovarian cancer with human sodium-iodide symporter-based positron emission tomography. Eur. J. Nucl. Med. Mol. Imaging 2024, 51, 3176–3190. [Google Scholar] [CrossRef]
  158. Quixabeira, D.C.A.; Pakola, S.; Jirovec, E.; Havunen, R.; Basnet, S.; Santos, J.M.; Kudling, T.V.; Clubb, J.H.A.; Haybout, L.; Arias, V.; et al. Boosting cytotoxicity of adoptive allogeneic NK cell therapy with an oncolytic adenovirus encoding a human vIL-2 cytokine for the treatment of human ovarian cancer. Cancer Gene Ther. 2023, 30, 1679–1690. [Google Scholar] [CrossRef]
  159. Hou, Y.; Zhao, X.; Nie, X. Enhancing the therapeutic efficacy of NK cells in the treatment of ovarian cancer (Review). Oncol. Rep. 2024, 51, 50. [Google Scholar] [CrossRef]
  160. Geller, M.A.; Cooley, S.; Judson, P.L.; Ghebre, R.; Carson, L.F.; Argenta, P.A.; Jonson, A.L.; Panoskaltsis-Mortari, A.; Curtsinger, J.; McKenna, D.; et al. A phase II study of allogeneic natural killer cell therapy to treat patients with recurrent ovarian and breast cancer. Cytotherapy 2011, 13, 98–107. [Google Scholar] [CrossRef]
  161. Hoogstad-van Evert, J.; Bekkers, R.; Ottevanger, N.; Schaap, N.; Hobo, W.; Jansen, J.H.; Massuger, L.; Dolstra, H. Intraperitoneal infusion of ex vivo-cultured allogeneic NK cells in recurrent ovarian carcinoma patients (a phase I study). Medicine 2019, 98, e14290. [Google Scholar] [CrossRef] [PubMed]
  162. Evert, J.S.H.; de Jonge, P.; Zusterzeel, P.L.M.; Hobo, W.; van der Waart, A.B.; Fredrix, H.; Janssen, L.; Wuts, M.; Bosmans, L.; Spijkers, E.; et al. Intraperitoneal infusion of stem cell-derived natural killer cells in recurrent epithelial ovarian cancer patients: Results of the phase 1 INTRO-01 trial. Gynecol. Oncol. 2025, 204, 91–99. [Google Scholar] [CrossRef]
  163. Fang, Y.; Xiao, X.; Wang, J.; Dasari, S.; Pepin, D.; Nephew, K.P.; Zamarin, D.; Mitra, A.K. Cancer associated fibroblasts serve as an ovarian cancer stem cell niche through noncanonical Wnt5a signaling. NPJ Precis. Oncol. 2024, 8, 7. [Google Scholar] [CrossRef] [PubMed]
  164. Feng, S.; Ding, B.; Dai, Z.; Yin, H.; Ding, Y.; Liu, S.; Zhang, K.; Lin, H.; Xiao, Z.; Shen, Y. Cancer-associated fibroblast-secreted FGF7 as an ovarian cancer progression promoter. J. Transl. Med. 2024, 22, 280. [Google Scholar] [CrossRef] [PubMed]
  165. Trub, M.; Uhlenbrock, F.; Claus, C.; Herzig, P.; Thelen, M.; Karanikas, V.; Bacac, M.; Amann, M.; Albrecht, R.; Ferrara-Koller, C.; et al. Fibroblast activation protein-targeted-4-1BB ligand agonist amplifies effector functions of intratumoral T cells in human cancer. J. Immunother. Cancer 2020, 8, e000238. [Google Scholar] [CrossRef] [PubMed]
  166. Melero, I.; Tanos, T.; Bustamante, M.; Sanmamed, M.F.; Calvo, E.; Moreno, I.; Moreno, V.; Hernandez, T.; Martinez Garcia, M.; Rodriguez-Vida, A.; et al. A first-in-human study of the fibroblast activation protein-targeted, 4-1BB agonist RO7122290 in patients with advanced solid tumors. Sci. Transl. Med. 2023, 15, eabp9229. [Google Scholar] [CrossRef]
  167. Li, Y.; Liu, Q.; Jing, X.; Wang, Y.; Jia, X.; Yang, X.; Chen, K. Cancer-Associated Fibroblasts: Heterogeneity, Cancer Pathogenesis, and Therapeutic Targets. MedComm 2025, 6, e70292. [Google Scholar] [CrossRef]
  168. Rupert, J.; Daquinag, A.; Yu, Y.; Dai, Y.; Zhao, Z.; Kolonin, M.G. Depletion of Adipose Stroma-Like Cancer-Associated Fibroblasts Potentiates Pancreatic Cancer Immunotherapy. Cancer Res. Commun. 2025, 5, 5–12. [Google Scholar] [CrossRef]
Figure 1. A simplified diagram of combined molecule-based and immune cell-based therapies aimed at improving clinical outcomes. EOCs, epithelial ovarian carcinomas; GCTs, germ cell tumors; SCSTs, sex cord–stromal tumors; SCCOHT, small-cell carcinoma of the ovary, hypercalcemic type; CAF, cancer-associated fibroblast; VEGF, Vascular Endothelial Growth Factor; Type1 IFN, type I interferon; DC, dendritic cell; PGE2, prostaglandin E2; ER, endoplasmic reticulum; TAMs, tumor-associated macrophages; ECM, extracellular matrix; NK, natural killer; CAR, Chimeric Antigen Receptor. This image was created by BioRender (http://biorender.com/).
Figure 1. A simplified diagram of combined molecule-based and immune cell-based therapies aimed at improving clinical outcomes. EOCs, epithelial ovarian carcinomas; GCTs, germ cell tumors; SCSTs, sex cord–stromal tumors; SCCOHT, small-cell carcinoma of the ovary, hypercalcemic type; CAF, cancer-associated fibroblast; VEGF, Vascular Endothelial Growth Factor; Type1 IFN, type I interferon; DC, dendritic cell; PGE2, prostaglandin E2; ER, endoplasmic reticulum; TAMs, tumor-associated macrophages; ECM, extracellular matrix; NK, natural killer; CAR, Chimeric Antigen Receptor. This image was created by BioRender (http://biorender.com/).
Medicina 61 02246 g001
Table 2. Subtype-specific frequencies of genetic alterations in OCs.
Table 2. Subtype-specific frequencies of genetic alterations in OCs.
HGSOC
(n = 306) [29]
HGSOC
(n = 42)
[108]
HGSOC
(n = 133)
[106]
LGSOC
(n = 119)
[107]
MOC
(n = 9)
[106]
MOC
(n = 31)
[31]
CCC
(n = 24)
[106]
SCST
(n = 19)
[106]
GCT
(n = 46)
[106]
SCCOHT
(n = 12)
[97]
TP5396%98%98%2%89%52%4% 54%
Myc31%11%7%0% 8%
PIK3CA18%4%9%<1%22% 50%
SOX215%0%4%0%
BRCA112%7%2%0%
BRCA212%13%1%<1%
NF112%2%6%2%
KRAS11%11%5%32%67%45%8% 28%
SMARCA411%4% <1% 8% 92%
AKT13%0%<1%0%
PTEN8%4%7%0% 11%
CHD48%
BRAF7%0% 9% 23%
FOXL25%0% 0% 68%
ARID1A2%4%7%<1% 3%83%
CTNNB12%2% 0% 4%16%
APC 4% 0%
POLE2%0% 0% 4%
DICER13%0%0%<1%
BCOR<1%7%4%<1% 4%
Genetic-alteration frequencies (%) are cohort-specific. Cohort size and reference for each dataset are provided in the corresponding column. HGSOC, high-grade serous ovarian carcinoma; LGSOC, low-grade serous ovarian carcinoma; MOC, mucinous ovarian carcinoma; CCC, clear cell carcinoma; SCST, sex cord–stromal tumor; GCT, germ cell tumor; SCCOHT, small-cell carcinoma of the ovary, hypercalcemic type.
Table 3. Ongoing clinical trials on immune cell-based immune therapy on ClinicalTrials.gov.
Table 3. Ongoing clinical trials on immune cell-based immune therapy on ClinicalTrials.gov.
TypesNCT NumberPhaseTitleStatusRef.
DC-based therapyNCT007991102Vaccination of Patients with Ovarian Cancer with Dendritic Cell/Tumor Fusions with Granulocyte Macrophage Colony-Stimulating Factor (GM-CSF) and ImiquimodACTIVE_
NOT_RECRUITING
NCT048345442A Study of Maintenance DCVAC/OvCa After First-Line Chemotherapy Added Standard of CareRECRUITING
NCT047395271Phase 1 Study to Evaluate the Safety, Feasibility and Immunogenicity of an Allogeneic, Cell-Based Vaccine (DCP-001) in High-Grade Serous Ovarian Cancer Patients After Primary TreatmentACTIVE_
NOT_RECRUITING
[142]
NCT057738591/2NEOadjuvant Dendritic Cell Vaccination for Ovarian CancerRECRUITING[143]
NCT059207981/2Vaccine Therapy Plus Pembrolizumab in Treating Advanced Ovarian, Fallopian Tube, or Primary Peritoneal Cavity CancerRECRUITING
NCT059643611/2First-in-Human Interleukin-15-Transpresenting Wilms’ Tumor Protein 1-Targeting Autologous Dendritic Cell Vaccination in Cancer PatientsACTIVE_
NOT_RECRUITING
NCT066390742Folate Receptor Alpha Dendritic Cells or Placebo for the Treatment of Patients with Stage III or IV Ovarian, Fallopian Tube, or Primary Peritoneal Cancer, FAROUT TrialRECRUITING
Macrophage-based therapyNCT01113112-Biobehavioral–Cytokine Interactions in Ovarian CancerACTIVE_
NOT_RECRUITING
NCT046609291CAR-Macrophages for the Treatment of HER2 Overexpressing Solid TumorsACTIVE_
NOT_RECRUITING
[144]
NCT05053750Early_
1
TAME: A Pilot Study of Weekly Paclitaxel, Bevacizumab, and Tumor Associated Macrophage Targeted Therapy (Zoledronic Acid) in Women with Recurrent, Platinum-Resistant, Epithelial Ovarian, Fallopian Tube or Primary Peritoneal CancerACTIVE_
NOT_RECRUITING
NCT054676702Safety and Efficacy of Anti-CD47, ALX148 in Combination with Liposomal Doxorubicin and Pembrolizumab in Recurrent Platinum-Resistant Ovarian CancerRECRUITING
NCT06562647N.A.SY001 Targets Mesothelin in a Single-Arm, Dose-Increasing Setting in Subjects with Advanced Solid TumorsRECRUITING[145]
NCT06887673Early_
1
Lipid Mediators and Cancer: Montelukast, SPM, and AlmondsNOT_YET_RECRUITING
NK cell-based therapyNCT024876932Radiofrequency Ablation Combined with Cytokine-Induced Killer Cells for the Patients with Ovarian CarcinomaACTIVE_
NOT_RECRUITING
NCT054107171CLDN6/GPC3/Mesothelin/AXL-CAR-NK Cell Therapy for Advanced Solid TumorsRECRUITING[146]
NCT059229301/2Study of TROP2 CAR Engineered IL15-Transduced Cord Blood-derived NK Cells Delivered Intraperitoneally for the Management of Platinum Resistant Ovarian Cancer, Mesonephric-Like Adenocarcinoma, and Pancreatic CancerRECRUITING[147]
NCT063958441/2Safety and Efficacy of Intraperitoneal Injection of METR-NK Cells as Neoadjuvant Therapy for Advanced Epithelial Ovarian CancerRECRUITING
NCT063429861Intraperitoneal FT536 in Recurrent Ovarian, Fallopian Tube, and Primary Peritoneal CancerRECRUITING
NCT05856643Early_
1
Single-Arm, Open-Label Clinical Study of SZ011 in the Treatment of Ovarian Epithelial CarcinomaRECRUITING
NCT063214841Intraperitoneal Cytokine-Induced Memory-Like (CIML) Natural Killer (NK) Cells in Recurrent Ovarian CancerRECRUITING
NCT068843451/2METR-NK Cells in Combination with Anti-Angiogenic Neoadjuvant Therapy for Advanced Epithelial Ovarian CancerACTIVE_
NOT_RECRUITING
NCT070965831/2An Exploratory Study on NK Cell-Assisted Prevention of Bone Marrow Suppression During Chemotherapy for Ovarian CancerACTIVE_
NOT_RECRUITING
Information related to ongoing clinical studies was obtained from https://ClinicalTrials.gov and includes only studies that are currently active, not completed.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hong, S.Y.; Cho, A.; Chae, C.-S.; You, H.J. Targeting Ovarian Neoplasms: Subtypes and Therapeutic Options. Medicina 2025, 61, 2246. https://doi.org/10.3390/medicina61122246

AMA Style

Hong SY, Cho A, Chae C-S, You HJ. Targeting Ovarian Neoplasms: Subtypes and Therapeutic Options. Medicina. 2025; 61(12):2246. https://doi.org/10.3390/medicina61122246

Chicago/Turabian Style

Hong, Seon Young, Ahyoung Cho, Chang-Suk Chae, and Hye Jin You. 2025. "Targeting Ovarian Neoplasms: Subtypes and Therapeutic Options" Medicina 61, no. 12: 2246. https://doi.org/10.3390/medicina61122246

APA Style

Hong, S. Y., Cho, A., Chae, C.-S., & You, H. J. (2025). Targeting Ovarian Neoplasms: Subtypes and Therapeutic Options. Medicina, 61(12), 2246. https://doi.org/10.3390/medicina61122246

Article Metrics

Back to TopTop