CC BY-NC-ND 4.0 · Geburtshilfe Frauenheilkd
DOI: 10.1055/a-2613-0489
GebFra Science
Review

Revisiting Genomic Instability, Tumor Microenvironment and Immune Response in High-Grade Serous Ovarian Cancer

Neue Überlegungen zu genomischer Instabilität, Tumormikromilieu und Immunreaktion bei hochgradigen serösen Ovarialkarzinomen
1   Department of Obstetrics and Gynecology, University Hospital Cologne, Cologne, Germany (Ringgold ID: RIN27182)
2   Genome Instability and Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany (Ringgold ID: RIN130368)
,
Sunhwa Baek
1   Department of Obstetrics and Gynecology, University Hospital Cologne, Cologne, Germany (Ringgold ID: RIN27182)
,
Stephanie Panier
2   Genome Instability and Ageing, Max Planck Institute for Biology of Ageing, Cologne, Germany (Ringgold ID: RIN130368)
3   Medical Faculty, Institute for Genome Stability in Ageing and Disease, University of Cologne, Cologne, Germany (Ringgold ID: RIN14309)
› Author Affiliations

Supported by: Else Kröner-Fresenius-Stiftung
Supported by: Federation of European Biochemical Societies FEBS Excellence Award
Supported by: Deutsche Forschungsgemeinschaft FOR5504, FOR5504/1
 

Abstract

High-grade serous tubo-ovarian cancer is the most common and aggressive type of ovarian cancer characterized by extensive genomic instability and marked inter- and intra-patient tumor heterogeneity. Tumor-site specific signaling crosstalk between cancer cells and the tumor microenvironment influences different tumor ecosystems that drive therapy response and disease progression. Cancer cell-intrinsic genomic aberrations further contribute to the diversity of the tumor immune landscape. Homologous recombination deficiency is considered a key oncogenic driver in 50% of the cases underlying distinctive mechanisms of tumor evolution. The heterogenous character of the tumor microenvironment represents a major challenge to identify predictive biomarkers of therapy response and to stratify subgroups amenable to immunotherapies.


#

Zusammenfassung

Das hochgradige seröse Tubo-Ovarialkarzinom ist die häufigste und aggressivste Art des Ovarialkarzinoms. Gekennzeichnet ist diese Tumorart durch genomische Instabilität sowie eine ausgeprägte Tumorheterogenität zwischen verschiedenen Patientinnen und verschiedenen Tumorlokalisationen innerhalb einer Patientin. In unterschiedlichen Tumorlokalisationen beeinflussen spezifische Signalwege zwischen Krebszellen und dem Tumormikromilieu verschiedene Tumorekosysteme, die sich auf das Therapieansprechen und die Krankheitsprogression auswirken. Krebszellenspezifische genomische Defekte tragen zusätzlich zur Diversität der Tumor-Immunantwort bei. Eine homologe Rekombinationsdefizienz gilt in 50% der Fälle als wichtiger onkogener Faktor für unterschiedliche Mechanismen der Tumorevolution. Der heterogene Charakter dieser Tumormikromilieus stellt eine große Herausforderung dar bei der Identifizierung von prädiktiven Biomarkern für das Therapieansprechen und bei der Stratifikation von Untergruppen, die auf eine Immuntherapie ansprechen.


#

Introduction

High-grade serous tubo-ovarian cancer (HGSC) is the most common and aggressive type of ovarian cancer, and the second leading cause of death among gynecological cancers [1]. Intra-abdominal dissemination and non-specific late symptoms are major obstacles to early disease detection leading to 75% of cases diagnosed at advanced stages with a 5-year survival rate of 30% [2]. At the mutational level, HGSC is characterized by chromosomal aberrations, copy number alterations, nearly ubiquitous TP53 mutation, frequent loss of RB1, NF1, and PTEN, and a very low tumor mutational burden (TMB) [3] [4] [5] [6]. Loss of p53 function is a key early event in HGSC tumorigenesis, leading to chromosomal instability. HGSC exhibits extensive genomic rearrangements, including telomeric allelic imbalances, large-scale structural transitions, and loss of heterozygosity, creating a permanent “genomic scar” [7] [8] [9]. Approximately 50% of HGSCs show homologous recombination deficiency (HRD), considered an early oncogenic driver that further provokes chromosomal instability. Indeed, ovarian cancer has the highest HRD score in a pan-cancer analysis [10]. The HRD phenotype results from BRCA1/2 loss-of-function mutations in 30% of cases. In addition, alterations in other genes involved in homologous recombination (HR)-mediated DNA repair (ATM, ATR, BAP1, BARD1, BRIP1, CHEK1, CHEK2, MRE11A, NBN, PALB2, RAD50, RAD51, XRCC2) contribute to the HRD-positive phenotype ([Fig. 1]). Aberrations in non-HR DNA repair pathways are prevalent in distinct ovarian cancer histotypes with different therapeutic implications [11].

Zoom Image
Fig. 1 Homologous recombination repair pathway of double-strand breaks (for a detailed description see info box). Created with Biorender.com [rerif]
Info box

Homologous recombination repair pathway of double-strand breaks

Homologous recombination (HR) plays an essential role in the repair of DNA double-strand breaks (DSBs) and collapsed replication forks thereby preserving genome integrity. HR is a high-fidelity repair mechanism. Alternatively, DSB repair is processed via non-homologous end joining (NHEJ), or other homology-directed ligation-mediated repair such as MMEJ or SSA, that are highly efficient but error-prone mechanisms [12].

Differential expression and post-translational modifications of regulatory factors determine cell-cycle dependent regulation of HR proteins [12] [13]. DSB repair by HR is initiated by 5‘ to 3‘ end resection that depends on CtIP and the MRN nuclease complex producing 3‘ single-strand DNA (ssDNA) overhangs. The ssDNA overhangs are rapidly coated by RPA. CtIP also recruits the nucleases and helicases DNA2 and EXO1 generating long tracts of ssDNA. Cells with a deficiency of a core end resection-regulating gene are more susceptible to chemotherapy.

The DNA-dependent protein kinases ATM and ATR are recruited to DSBs, and phosphorylate a pool of substrates including p53, BRCA1, BRCA2, PALB2, RAD51, H2AX, and cell cycle checkpoint factors (CHK1, CHK2) promoting DNA repair and related processes (transcription, replication, mitosis) [14]. The checkpoint function of ATM plays an important role in tumor suppression. ATM loss renders cancer cells sensitive to chemotherapy and PARPi. ATM further affects HR via 53BP1, an important regulator of DNA repair pathway choice antagonizing end resection. In BRCA1-deficient patients, loss of 53BP1 leads to PARPi resistance due to recovery of end resection [15] [16]. Loss of ATR and CHK1 are synergistically lethal with p53-deficiency, which is the basis for the implementation of ATRi in the treatment of p53-dependent cancers.

BRCA1, BRCA2 and RAD51 are the main HR regulators. Phosphorylated BRCA1 forms complexes with different proteins (BARD1, BRIP1, PALB2) that exert pleiotropic roles in DSB repair, ubiquitination, gene transcription and cell cycle progression [17]. Mutations in BARD1 confer hypersensitivity to inhibitors of topoisomerases, ionizing radiation and PARPi [18]. BRIP1 interacting with BRCA1 suppresses NHEJ-mediated repair and is thus considered a tumor suppressor gene.

The BRCA1-PALB2-BRCA2 complex catalyzes the formation of the RAD51-ssDNA nucleoprotein filament. PALB2 loss is associated with increased risk for ovarian cancer [17]. BAP1 supports recruitment of BRCA1, BRCA2 and RAD51. Loss of BAP1 function results in defective DSB repair accelerating carcinogenesis [19]. Deleterious variants of BRCA2 at RAD51 binding motifs causes hereditary breast and ovarian cancer syndromes [17].

RAD51, the final effector in the HR cascade, initiating homology search for complementary sequences and strand invasion. Synthesis of dsDNA is mediated by RAD51 recombinase and its paralogues RAD51C, RAD51D, RAD51B, XRCC2, XRCC3. The synthesis branch proceeds by engaging the second end of the broken DNA by either annealing to the complementary sequence of the non-invading strand (synthesis-dependent strand annealing, SDSA), or a second invasion event forming a double Holliday junction (dHJ) [20]. Final steps in HR involve (a) dissolution or (b) resolution of the repair branch involving genetic crossover or non-crossover-products [21] [22].

ATM: Ataxia teleangiectasia mutated; ATR: Ataxia Telangiectasia and Rad3-related; BAP1: BRCA1-Associated Protein 1; BARD1: BRCA1-associated RING domain protein 1; BRIP1: BRCA1 interacting DNA helicase 1; CtIP: carboxy-terminal binding protein-interacting protein; MMEJ: microhomology-mediated end joining, also known as alternative end-joining (altEJ); MRN: MRE11-RAD50-NBS1; PALB2: Partner and localizer of BRCA2; RPA: replication protein A; SSA: single-strand annealing; yH2AX: phosphorylated H2AX.

BRCA1/2 mutations disrupt the high-fidelity HR DNA repair pathway. In turn these cells depend on error-prone DNA repair mechanisms such as non-homologous end-joining (NHEJ), polymerase Theta (POLQ)-mediated end-joining (also referred to as alternative end-joining, altEJ), or single-strand annealing (SSA) [23] [24]. HRD imparts sensitivity to DNA-damaging platinum-based chemotherapy, favoring better progression-free and overall survival [10]. It also enables the use of PARP (Poly [ADP-ribose] polymerase) inhibitors as a synthetic lethal treatment by preventing the repair of single-strand DNA breaks. These breaks persist and ultimately convert to double-strand DNA breaks that lead to cell death in HR-deficient cells [25]. Patients with BRCA1/2 mutations who achieved a complete or partial response to first-line platinum-based chemotherapy have shown a significant survival benefit with seven years overall survival of 67% from PARP inhibition (PARPi) with olaparib, as demonstrated in the SOLO1 trial [26]. PARPi implementation delayed disease recurrence and the need for second chemotherapy, and has now shifted to earlier application in the course of the disease as frontline maintenance therapy after surgical debulking and chemotherapy based on the SOLO1, PAOLA-1, ATHENA-MONO and PRIMA trials [27] [28] [29] [30].

Despite advances, many patients experience disease progression or recurrence under standard of care first-line treatment. Molecular markers predicting therapy resistance are under investigation but not yet in clinical use. Of note, heterogenous disease courses are governed by marked inter- and intra-patient and tumor microenvironment heterogeneity, affecting both therapy response and disease progression. Here, we aim to revisit the multifaceted aspects of tumor heterogeneity in HGSC at the level of tumor mutational context and the tumor microenvironment.


#

Mechanisms and Markers of Therapy Resistance in HGSC

Platinum resistance

Patients with platinum-resistant HGSC represent a heterogeneous group. The definition of platinum responsiveness after relapse has been revisited and is now based on the treatment-free interval after platinum therapy (TFIp) [31]. Shorter TFIp (< 6 months) is linked to reduced survival due to limited treatment options [32] [33]. Diverse mechanisms contribute to these variable treatment responses [34] [35] [36]. For example, resistance can arise via clonal selection of quiescent, drug-resistant cells from a pool of genomically heterogenous sub-clones in the chemo-naïve state, secondary mutations, or adaptive epigenetic changes [36] [37]. The latter may represent a transient resistant state induced by potentially druggable targets. Further, reversion mutations in BRCA1/2 or loss of BRCA1 promoter methylation can restore DNA repair capacity.

About 10–20% of patients with HGSC are platinum-refractory, showing no initial response [38]. Even so, validated predictive markers or genomic signatures of platinum resistance remain absent from clinical use [35]. Partial BRCA function correlates with poorer survival and may go undetected in standard BRCA mutation testing [39]. Genes forming other DNA repair pathways that resolve platinum DNA-adducts, for example nucleotide excision repair genes (CDK7, CETN2, LIG3, TFIIH), are often overexpressed in resistant HGSC [40]. PARP4, among the most overexpressed DNA repair genes in platinum-resistant HGSC, has been linked to multidrug resistance, although its role remains unclear [40] [41]. Further investigations into PARP4 as a marker and target for platinum-resistant HGSC are warranted, especially in light of already existing PARP inhibitors.

Copy number analysis of tumors from platinum-resistant patients revealed more frequent amplification of CCNE1, KRAS and PIK3CA compared to platinum-sensitive patients [40] [42]. CCNE1 and CDK2 facilitate cell cycle progression, replication initiation and centrosome duplication [43]. CCNE1 amplification induces replication stress and chromosomal instability [44], and CDK2 inhibition sensitizes CCNE1-amplified ovarian cancer cells to DNA damaging agents [45]. CCNE1 is one of the most common driver gene amplifications [46] and should be further evaluated as a negative predictor of response to chemotherapy and PARPi. High KRAS signaling disrupts G2 and mitotic checkpoint controls, causing chromosomal instability platinum-resistance in vitro [47] [48]. PI3K signaling dysregulation also promotes chemoresistance, and PI3KCA gene amplification may contribute to oncogenic signaling [49].

Tumors overexpressing negative cell cycle regulators (PTEN, APC) show poor chemotherapy responsiveness, possibly due to slower proliferation allowing enhanced DNA repair [40]. Conversely, high levels of proliferation markers (PCNA, MKI67) correlate with better long-term survival [50]. Single-analyte biomarker studies may fail to capture the full range of resistance mechanisms. However, a multi-protein prediction model integrating proteomics, whole genome sequencing, and bulk RNA-sequencing on a cohort of primary chemo-naïve HGSC tumors revealed heterogeneous resistance mechanisms [51]. In this model, enriched pathways included translational and rRNA processing, cell-cycle, metabolism (oxidative phosphorylation and tricarboxylic acid cycle), hypoxia, EMT, and TGFβ, suggesting subtype-specific treatment approaches [51].


#

PARPi resistance

Roughly 50% of patients with a BRCA1/2 mutated (BRCAm) ovarian cancer relapse within seven years following PARPi maintenance therapy [26]. Mechanisms of resistance to PARPi overlap only partly with those for platinum resistance [37] [52]. Clinically relevant relapse mechanisms include reversion mutations in BRCA1, BRCA2, RAD51B, RAD51C and RAD51D, and loss of BRCA1 promoter methylation, all of which restore HR repair [36] [53] [54] [55]. The BRCA1-Δ11q isoform promotes resistance to platinum and PARPi by skipping transcription of the germline mutation and is linked to poor overall survival [56] [57]. Sequence alterations such as MRE11 amplification, which leads to increased DNA end-resection and HR, and TP53BP1 loss can compensate for BRCA1 loss, but may present opportunities for therapeutic targeting [56] [58].

Despite improved patient prognosis with current therapies, post-relapse treatment selection remains critical. Several studies suggest that prior PARPi treatment may reduce subsequent chemotherapy efficacy [59] [60] [61] due to overlapping resistance mechanisms. In the phase III ARIEL4 trial, 7% of patients had BRCA1/2 reversion mutations after platinum-based chemotherapy [62], compared to 26% after PARPi treatment [63]. Reversion mutation impact varies by location within different BRCA1/2 protein domains. Alternative DSB repair pathways like NHEJ and MMEJ, may drive these mutations and could be targeted to prevent or delay PARPi resistance [62] [64]. Other resistance mechanisms under PARPi included upregulation of the multidrug efflux pump gene ABCB1, CCNE1 amplification, and downregulation of SLFN11, a DNA/RNA helicase that contributes to resistance to replication stress-inducing agents [65] [66]. Further molecular analysis is needed to understand post-PARPi chemotherapy response.

Of note, prior PARPi exposure does not always preclude benefit from PARPi retreatment. For example, the OReO trial showed a modest significant improvement of progression-free survival with PARPi rechallenge in both the BRCAm and non-BRCAm cohorts [67]. For this trial, eligible patients had prior partial or complete responses to platinum. Ongoing preclinical and clinical studies are testing novel combinations of PARPi with inhibitors of AKT, PI3K, ATR, CHK1, WEE1, and HDAC, which may provide new options for HGSC patients [68] [69].


#
#

Determinants of the Anti-Tumor Immune Response

While PD-1/PD-L1-targeting immunotherapies show clinical success in many cancer types, their efficacy in unselected ovarian cancers remains limited [70] [71]. This is largely attributed to distinct mutational signatures and heterogenous tumor immune phenotypes, making ovarian cancer particularly resistant to immunotherapy. Although biomarkers like tumor-infiltrating lymphocytes (TILs), PD-L1 expression, high microsatellite instability/defects in mismatch repair and TMB have been established in other cancers, clinical trials have shown their limited predictive value in ovarian cancer [72].

The presence of TILs, especially CD8+ T cells, correlates with improved prognosis in ovarian cancer and long-term survivorship in selected subgroups [73]. HGSC tumors display variable TIL patterns that correspond to known molecular HGSC subtypes: epithelial immune-infiltrated (“inflamed”), stromal restricted immune-infiltrated (“excluded”) and non-infiltrated (“desert”) [74] [75]. However, HGSC classification by gene expression signatures and TIL presence lacks cross-study robustness and clinical use [76] [77] [78]. This is partly due to the diversity within the intra-tumoral CD8+ T cell compartment, including naïve (CD8+PD-1−TOX−TCF1−), activated/predysfunctional (CD8+PD-1+TOX−), exhausted/dysfunctional (CD8+PD-1+TOX+), and heterogenous memory (CD45RA−/CD45RO+) T cells, as well as various transitional states. Moreover, TIL presence does not always indicate anti-tumor reactivity, with many T cells identified as bystanders lacking cytotoxic function [79] [80]. Immune cells residing within the malignant compartment also appear to differ transcriptionally from those in the stromal compartment [80].

The outcome of the anti-tumor immune response is further shaped by spatially distinct tumor microenvironments (TME). Variability in immune cell composition and functional states within the TME complicates the use of PD-L1 expression as a reliable biomarker [76] [77]. Interaction networks between malignant, stromal, and immune cell lineages and signaling pathways guiding T cell infiltration remain incompletely understood but are essential for immunotherapy response and biomarker development. The following sections discuss how spatial immune heterogeneity in HGSC underlies disease progression and therapeutic outcomes.


#

HGSC is Governed by Spatially Distinct Immune Ecosystems

HGSC typically spreads intra-abdominally, leading to multiple tumor sites with distinct TMEs. Comparative analyses of tumors from the ovary, omentum and ascites revealed significant variation in tumor and immune cell composition ([Table 1]). Limited migration of T cell clusters between sites further underscores the non-representative nature of single-site sampling and its implications for treatment planning [81] [82]. Intriguingly, tumor site-specific immune dynamics appear to depend on underlying mutational processes.

Table 1 Comparison of immune changes in different tumor niches.

Tumor niche

Immune characteristics

Primary tumor

  • Chronic antigen exposure leading to dysfunctional CD4+ and CD8+ T cells

  • JAK-STAT signaling

  • PD-L1 upregulation on cancer cells and macrophages suppressing T cell cytotoxicity

  • Immunosuppressive cells

    • Regulatory T cells

    • M2-macrophages

    • Myeloid-derived suppressor cells

Peritoneal implants

  • Reduced antigen recognition

  • Reduced interaction between cancer cells and immune cells

Omental implants

  • High T cell infiltration pattern correlated with pro-inflammatory immune cell infiltrates:

    • M1-like PD-L1+ macrophages

    • CD4+ T cell

    • Tissue resident memory CD8+ T cells

    • Plasmablasts, plasma B cells

  • Low T cell infiltration pattern resembling an immune-excluded phenotype:

    • Cancer-associated fibroblasts

    • Stromal localization of CD4+ and non-exhausted CD8+ T cells

    • Few interactions of T cells and macrophages with PD-L1+ cancer cells

    • Reduced interferon signaling

    • Reduced MHC class I expression

Ascites

  • Lack of chronic antigen exposure

  • Low MHC-I and II expression on cancer cells

  • Increased naive/stem-like (TCF1+LEF1+) CD8+ T cells

  • Increased central memory (IL7R+TCF1+) CD4+ T cells

  • Fibroblast-derived IL-6 signaling

  • Low-anti-tumor immunity

Primary tumor sites

Adnexal tumors, often the primary tumor sites, are enriched in dysfunctional CD4+ and CD8+ T cells, indicating chronic cancer antigen exposure and adoptive immune exhaustion during early carcinogenesis [81] [82]. MHC-mediated antigen presentation activates type I interferon signaling, triggering the JAK-STAT pathway activation and PD-L1 upregulation in cancer cells and macrophages [82]. PD-L1/PD-1 engagement in turn suppresses T cell cytotoxicity as part of an immune regulatory feedback loop. Primary tumor sites also contain immunosuppressive regulatory CD4+CD25+FOXP3+ T cells (Tregs), tumor-associated/M2 macrophages, and myeloid-derived suppressor cells (MDSCs), further reinforcing immune evasion [81] [82]. Tregs, expressing CTLA-4, inhibit cytotoxic T cells [83], and their proximity to CD8+ TILs correlates with poor prognosis [83] [84].


#

Metastatic niches

Tumor cell dissemination to secondary sites is associated with site-specific TME evolution patterns. High CD8+ T cell abundance but reduced antigen recognition were evident in intraperitoneal tumors compared to primary tumors. Bowel metastatic sites showed increased interaction between PD-L1-expressing cancer cells and activated/predysfunctional T cells, along with JAK-STAT signaling [82]. In contrast, ascites samples were enriched in naive/stem-like (TCF1+LEF1+) CD8+ and central memory (IL7R+TCF1+) CD4+ T cells, but contained fewer dysfunctional CD4+ and CD8+ T cells or dendritic cells, indicating limited antigen exposure and low MHC-I/II expression [82]. The ascites microenvironment is further dominated by fibroblast-derived IL-6, activating JAK/STAT signaling and promoting tumor growth and drug resistance [85]. Although ascites appears to be an immune-infiltrated TME, its low antigen presentation suggests reduced anti-tumor reactivity and immune characteristics that are hostile for an adaptive immune response. Ascitic fluid, for example, suppressed the major glucose transporter GLUT1 in CD4+ T cells leading to activation of the unfolded protein response, and ultimately to endoplasmic reticulum (ER) stress. ER stress activates the IRE1α-XBP1 signaling pathway inducing metabolic dysfunction and reduced mitochondrial activity in CD4+ T cells correlating with reduced IFNγ production and reduced T cell infiltration [86]. Additional immunoregulatory metabolites, such as 1-methylnicotinamide (MNA), contribute to intratumoral T cell dysfunction in HGSC, reduced IFNγ production and diminished antitumor activity in vitro [87]. Mature neutrophils in the ascitic TME exert T cell suppression that is initiated by complement pathway signaling (via complement receptor 3 and C1q), and subsequent inhibition of CD8+ effector T cell differentiation [88]. Ovarian cancer clustered into an ‘upregulated complement’ group in TCGA data indicating that complement activation plays a role in ovarian carcinogenesis [89].

The omentum, a common metastatic niche, displays heterogenous T cell infiltration patterns. Highly infiltrated samples show co-infiltration of pro-inflammatory immune cells, including M1-like PD-L1+ macrophages, CD8+ tissue-resident memory T cells (TRM), CD4+ T cells, plasmablasts, and plasma B cell clusters supporting an effective anti-tumor CD8+ TIL response [90]. TRM T cells (CD8+CD103+CD69+CD62L−) can be reactivated by anti-PD-L1 therapy, exert cytotoxic function, and are associated with longer survival [90] [91] [92] [93]. B cells colocalize with CD8+ T cells in tertiary lymphoid structures (TLSs) and enhance immune activation through a robust, prognostically favorable CD8+ TIL response [94] [95] that includes increased chemokine signaling and coordinated antigen presentation between M1 macrophages or dendritic cells and T and B cells [90] [96] [97] [98]. However, TLSs in HGSC are often immature, marginally located, and contain CD8+TIM3+PD-1+ T cells associated with resistance to immune checkpoint inhibition – factors that could partially explain the limited responsiveness of HGSC to anti-PD-1/PD-L1 treatment [95].

Conversely, omental samples with low T cell infiltration exhibit an immune-excluded phenotype, with stromal CD4+ and non-exhausted (CD8+PD-1−GZMB−) CD8+ T cells, reduced interferon signaling, and impaired antigen presentation [81] [82]. These regions are dominated by cancer-associated fibroblasts (CAFs), which regulate extracellular matrix remodeling, immune cell distribution, and intercellular signaling by PDGFB, IL-6, TGFB1/2 and CXCL12-CXCR4 signaling [97] [99] [100] [101] [102] [103]. CAF-rich TMEs correlate with poor disease outcomes, therapy resistance and early relapse [96] [97] [104]. Stroma-dominated tumors respond poorly to neoadjuvant chemotherapy (NACT), whereas immune-rich tumors show better responses, underscoring the contribution of cytotoxic immune cells to successful chemotherapy [102]. Fibroblast-mediated regional barriers generate immune privileged sites that dampen tumor-immune cell engagement, contributing to immunotherapy failure and highlighting that TIL abundance alone is insufficient for predicting immune response in HGSC. Elevated tumor immune dysfunction and exclusion, T cell exclusion, and CAF scores in HGSC further support a high degree of immune escape in this cancer type [105] [106]. Better characterization of common stromal phenotypes may improve HGSC classification. These exemplary differences in TME composition and cellular activity states underscore that the immune TME in ascites samples does not represent solid tumors.


#
#

Mutational Processes are Associated to Distinct HGSC Immunophenotypes

Cancer cell-intrinsic genomic aberrations contribute to the heterogeneity of the tumor immune landscape. While the molecular regulators of tumor immunity in HGSC remain incompletely understood, recent studies highlight how genomic instability influences TME composition and cellular networks with implications for therapy response and resistance.

Copy number alterations, including gene deletions and amplifications, were shown to disrupt immune regulation and promote immune evasion. A cancer-cell intrinsic transcriptional program was linked to T and NK cell infiltration across HGSC samples and tumor sites [80]. Chemokines (CCL5, CXCL10, CXCL9 and CXCL16), oxidative stress genes (GPX3 and SOD2), and immune response genes – such as those involved in antigen presentation (MHCI, CIITA, HLA-A/HLA-B/HLA-C), interferon gamma signaling (IDO1, JAK1, STAT1), and cell adhesion (ICAM1, ITGAV and ITGB2) – were associated with an “immune-infiltrated” phenotype and may serve as future biomarkers for immunotherapy [80]. Conversely, expression of genes involved in Wnt signaling (CTNNB1, FZD3/FZD4/FZD6, FGFR2, WNT7A), epigenetic regulation (DNMT3A, HDAC1/HDAC11/HDAC4/HDAC5), and cell differentiation (BMP7, BMPR1A, ETV4, FGFR1/FGFR2, S100A4, SMAD4) negatively correlated with T and NK cell infiltration, suggesting a link between these pathways and immune evasion [80].

Tumor evolution

The pronounced tumor heterogeneity observed in HGSC stems in part from polyclonal tumor evolution, with subclones diverging after early clonal alterations in TP53, BRCA1/2, PTEN, RB1 and NF1 [56] [107] [108]. Emerging evidence suggests that the current HGSC molecular subtypes may reflect late subclonal events, which has therapeutic implications, as targeted therapies may miss heterogenous, resistant subclones [107] [108]. Subclonal heterogeneity also impacts TME composition through variable expression of signaling molecules that shape local immune networks [109]. Rather than discrete subtypes, HGSC may follow a continuous evolutionary trajectory from early “differentiated” to late “proliferative” tumors [107] [110]. Differentiated tumors exhibit low copy number alterations, near-normal ploidy, high lymphocyte infiltration, fallopian tube gene signatures, and correlate with lower patient age [107]. In contrast, proliferative tumors show high ploidy, frequent genome duplication, extensive amplifications, loss of epithelial features, immune evasion, and correlation to higher patient age [107]. Autopsy samples from end-stage HGSC, often highly aneuploid, display reduced infiltration of CD8+PD-1+ TILs, Tregs, plasma and myeloid cells, indicating a shift towards an immunosuppressive, protumorigenic microenvironment in advanced disease [56].


#

Whole genome doubling

HGSC exhibits one of the highest rates of whole genome doubling (WGD) (40%), correlating with poor overall survival [111]. WGD is associated with decreased immune cell infiltration, IFNγ-signaling, STING and MHC-II expression – all of which are hallmarks of an immunosuppressive TME [111] [112] [113] [114]. MHC-II-expressing ovarian cancer cells generate immune-active hotspots, enhancing IFNγ expression and cellular crosstalk [115]. Such hotspots are linked to improved immunotherapy in other cancers [116] [117]. Histone deacetylase inhibition (e.g. entinostat) has been shown to restore MHC-II expression in ovarian cancer cells and suppressed tumor growth in vitro [118]. As WGD increases TMB and, potentially, immunogenicity, reduced MHC-II expression may allow immune evasion in genetically unstable tumors, providing a potential therapeutic target in HGSC.


#

Tumor mutational burden and neoantigens

Despite wide-spread genomic instability, HGSC generally has low TMB [119] [120]. While DNA repair defects can increase TMB, levels remain low compared to highly immunogenic tumors such as lung cancer and melanoma, and even among gynecological cancers [6] [121]. Unlike cervical cancer, where TMB predicts immune checkpoint inhibitor response, this marker lacks predictive value in ovarian cancers [122] [123].


#

HRD signature and BRCA1/2 mutation status

HRD-positive HGSCs, particularly those with BRCA1/2 mutations, often show higher neoantigen load, lymphoid cell infiltration and improved overall survival ([Table 2]) [124]. BRCA1/2-mutant tumors exhibit elevated inflammatory signaling (e.g. CCL5, CXCL9/10, interferon-related gene expression) [74] [115] [125] [126] and active immune signaling, including PD-L1+ cancer cells engaging with CD8+ and CD4+ T cells [82] [126]. Interactions were also found between CD8+ T cells and antigen-presenting (IBA1+ CD11c+) macrophages, between CD4+ T cells and (CD11c+) antigen presenting cells, and both CD8+ T cells and CD4+ T cells with B cells, indicating a coordinated immune response [126]. However, the immune response varies between BRCA1 and BRCA2 mutations [127], partly due to a dual role of cGAS-STING signaling. This pathway senses cytoplasmic DNA that is released at increased levels by chromosomal instability. It activates interferon signaling and NF-kB responses that can be tumor-suppressive or -promoting depending on extent and duration of STING-activation [128] [129] [130] [131].

Table 2 Immune differences between HRD and non-HRD tumors.

HRD ovarian cancer

Non-HRD ovarian cancer

Increased lymphoid cell infiltration

Immune cell exclusion

Increased interaction of PD-L1+ cancer cells with CD8+ and CD4+ T cells

Low interaction between cancer cells and CD8+ T cells

Increased interaction between CD8+ T cells and APCs

Increased interaction between M2-macrophages and stromal cells

Increased interaction between CD8+ T cells and B cells

Increased expression of TGFβ

Increased expression of CCL5, CXCL9/10, interferon-related genes


#

Non-HRD HGSC and CCNE1 amplification

Non-HRD HGSC typically presents with an immunosuppressive TME, featuring a high-proliferative stroma, immune exclusion, scarce CD8+ T cell-tumor interactions, increased naïve T cells, and predominant engagement of M2-like (IBA1+CD163+CD11c+) macrophages with the stroma [82] [126] [132]. In HRD-negative tumors with CCNE1 amplification, heightened cancer cell-intrinsic TGFβ signaling at metastatic sites suggests a role in driving immune suppression and metastasis [82] [126]. As TGFβ signaling promotes T cell exclusion, CAF activation, and chemoresistance, it represents a promising target to modulate tumor immune composition [101] [133]. In vivo, TGFβ inhibition restores MHC-I expression on ovarian cancer cells [101], increases CD8+ T cell and NK cell activation and infiltration, and induces M1-polarization of TAMs [133]. These effects support ongoing clinical trials exploring TGFβ inhibition in HGSC [134].


#
#

Immunoregulatory Role of Standard-of-Care Treatments and its Combination with Immunotherapies

Standard treatments of HGSC, including cytoreductive surgery, chemotherapy, PARPi, and anti-angiogenesis treatment, modulate the immune TME with potential to impact immunotherapy efficacy ([Table 3]).

Table 3 Influence of standard-of-care treatment on immune environment of ovarian cancer.

Tumor niche

Immune characteristics

Surgery

  • Unfavorable tumor-intrinsic characteristics and immune TME composition promoting tumor metastasis, unfavorable dissemination pattern

  • Sub-optimal tumor removal

  • Increased regulatory T cells

  • Increased TGFβ levels

  • Complete tumor removal

  • Increased CD8+ T cell cytotoxicity

Chemotherapy

  • Reduced regulatory T cells

  • Increased number of CD8+ T cells

  • Increased CD8+ T cell cytotoxicity

    • Granzyme A, perforin 1, IFNγ

  • Increased interaction between antigen presenting cells and T cells

  • Increased interaction between PD-L1+ macrophages and CD8+ T cells

  • Stimulation of B cell differentiation

  • Reduced levels of TNF, IL-6, IL-8

  • Increased expression of MHCI, PD-L1+ on tumor cells

PARP inhibition

  • Increased number of CD8+ and CD4+ T cells

  • Increased activation of cGAS-STING

  • Increased expression of PD-1/PD-L1

VEGF inhibition

  • Endothelial normalization

  • Reduced hypoxia

  • Increased number of CD8+ and CD4+ T cells

  • Increased CD8+ to CD4+FOXP3+ T cell ratio

Surgery

Surgery is a main pillar in HGSC treatment since complete tumor resection has significant prognostic benefits. Ongoing research aims to identify cancer-intrinsic markers predicting the presence of miliary peritoneal metastasis that impede maximal tumor resectability and surgical success [135] [136]. Differences in tumor cell-intrinsic characteristics and immune cell composition impact miliary versus non-miliary dissemination patterns [137] [138]. Incorporating radiomic tumor quantitative analyses, diagnostic biopsies and unfavorable tumor immune characteristics may enhance predictive models.

Further, timing and extent of cytoreductive surgery critically affect patient prognosis, through reduction of tumor cell load and modulation of the immune microenvironment [139] [140] [141]. Elevated TGFβ levels and increased numbers of circulating CD4+ Tregs (CD4+/CD25+/FOXP3+) were observed following sub-optimal tumor removal compared to optimal primary debulking [140]. Primary cytoreductive surgery also enhanced the potential of CD8+ T cells to secrete IFNγ [139]. The favorable prognostic impact of CD8+ T cells was restricted to patients undergoing complete tumor resection and was absent in those with residual disease [142]. Notably, the immunomodulatory benefits of surgery were more pronounced when cytoreduction was performed as primary treatment modality [139].


#

Chemotherapy

Chemotherapy induces immunosuppression due to bone marrow cytotoxicity and has further immunomodulatory effects. NACT consisting of carboplatin/paclitaxel reshapes the immune landscape mostly by reducing CD4+ Tregs, increasing CD8+ T cell counts, and promoting spatial redistribution and interaction of CD8+ T cells with macrophages [139] [143] [144] [145]. Good responders exhibit stronger cytotoxic T cell responses, characterized by coexpression of granzyme A and perforin-1, elevated IFNγ production [143], and higher T cell receptor clonality, suggesting that chemotherapy induces or unmask (neo)antigens in post-NACT patients [146]. This tumor antigen release likely boosts B cell activation and differentiation, as indicated by an increase in class-switched memory B cells [147]. Chemotherapy-induced immunoregulation also involves elevated levels of dendritic cells, PD-L1+ macrophages, and increased antigen presentation signaling [148] [149] [150]. In responders, pro-tumorigenic cytokines such as TNF, IL-6, IL-8 are reduced, while IL-6 among other immune-related genes are more highly expressed in non-responders [143] [148]. Tumor cells upregulate MHC-I and PD-L1 in response to chemotherapy treatment [144] [146] [149] [150]. Overall, these effects promote an orchestrated immune response that could favor immune checkpoint inhibition. However, clinical trial results remain mixed. The phase III JAVELIN Ovarian 100 trial found no benefit from adding anti-PD-L1 (avelumab) therapy to carboplatin and paclitaxel in the first-line setting [151], while the phase II NeoPembrOv trial, which combined pembrolizumab (anti-PD-1) with NACT showed modest improvements in response rate, resectability, and overall survival [152]. Of note, spatial and molecular analyses from NeoPembrOv revealed notable TME changes in responders, including increased CD8+PD-1+ T cell density within tumor islets and reduced distance to tumor cells [153]. A high monocyte signature, low endothelial signature, and high CD8B/FOXP3 expression ratio were associated with better survival and may serve as predictive biomarkers for immunotherapy response [153].


#

PARP inhibition

PARPi modulates the tumor microenvironment by enhancing IFNγ-mediated STING response and upregulating PD-L1 expression, and increasing microvascular density and VEGFA expression in BRCA1-deficient in vivo models [130] [154]. It also induces antitumor immunity through induction of effector CD4+ and CD8+ T cells [154]. In PARPi-resistant BRCA1-deficient HGSC, macrophages shift toward an immunosuppressive, pro-tumorigenic M2 phenotype via STAT3 signaling [155] [156]. Enhancing DNA damage through PARPi activates cGAS/STING and improves anti-PD-1/anti-PD-L1 efficacy, with strong results in preclinical [154] [157] [158] and early phase I/II clinical studies [159] [160]. However, the phase III ANITA/ENGOT-Ov41/GEICO 69-O clinical trial found no added benefit from combining niraparib maintenance treatment and atezolizumab (anti-PD-L1) following platinum-based chemotherapy in patients with recurrent ovarian cancer and a platinum-free interval > 6 months [161].


#

VEGF inhibition

Angiogenic factors like VEGF and TGFβ in the HGSC TME promote tumor progression, ascites formation and immunosuppression [162] [163]. VEGF recruits immunosuppressive Tregs and pro-tumorigenic M2 macrophages, inhibits dendritic cell maturation, and reduces lymphocyte infiltration by inducing endothelial dysfunction, and downregulating cell adhesion molecules and chemokines [163] [164]. The VEGF receptor VEGFR2 was identified as a resistance factor to anti-PD-1 treatment in the NeoPembrOv trial [153]. Bevacizumab (anti-VEGF) normalizes tumor microvasculature, reduces hypoxia-driven signaling [163], and enhances immune infiltration by boosting cytotoxic CD8+ and CD4+ T cells, and increasing the CD8+/CD4+/FOXP3+ T cell ratio, which correlates with improved progression-free survival [165]. Although combining anti-angiogenesis therapy with immune checkpoint inhibition was hypothesized to be synergistic, the phase III clinical trial AGO-OVAR 2.29 failed to demonstrate significant improvements in progression-free and overall survival (ClinicalTrials.gov: NCT03353831, [166]). Similarly, in the ongoing ATALANTE/ENGOT OV-29 phase III clinical trial for platinum-sensitive recurrent ovarian cancer, combining atezolizumab with platinum-based chemotherapy and bevacizumab did not improve progression-free survival. Here, overall survival follow-up data are still pending [167].


#

Triple combination therapy

Triple therapy is under investigation in the first-line setting as well as for maintenance therapy. The KEYLYNK-001 trial (ClinicalTrials.gov: NCT03740165) demonstrated a benefit in progression-free survival for pembrolizumab (anti-PD-1) and olaparib after upfront chemotherapy in newly diagnosed BRCA wildtype patients. Triple maintenance therapy for platinum-sensitive relapsed ovarian cancer was investigated in the MEDIOLA phase II trial. Germline BRCAm patients treated with olaparib and durvalumab (anti-PD-L1) showed excellent overall response rate of 92% [160]. In the non-BRCAm subgroups, triple maintenance therapy with bevacizumab in addition to olaparib and durvalumab showed an overall response rate of 87% compared to 34% without bevacizumab, indicating that especially for patients with non-BRCAm platinum sensitive relapsed ovarian cancer, triple maintenance therapy could be of clinical relevance regardless of previous therapy with bevacizumab. Also, durvalumab and olaparib treatment in combination with bevacizumab maintenance therapy after chemotherapy showed improved progression-free survival compared to bevacizumab only maintenance therapy in newly-diagnosed ovarian cancer patients with non-BRCAm tumors in the phase III DUO-O trial (ClinicalTrials.gov: NCT03737643). The benefit was slightly higher for HRD-positive patients than for HRD-negative patients. Since this trial lacked an olaparib/bevacizumab control arm, individual contributions of PARPi and anti-PD-L1 treatment or a synergistic effect of the triplet maintenance therapy remained inconclusive.


#
#

HRD Score and Predictive Markers of Immunotherapy Revisited

Currently, HRD and BRCA1/2 mutation status are the only validated genomic markers predicting response to platinum-based and PARPi therapy [11] [47]. However, they do not correlate with clinical benefit from immune checkpoint inhibition [168] [169]. Clinical HRD assays measure the “genomic scar” from accumulated mutations, insertions/deletions, and genome rearrangements, but various non-equivalent HRD assays exist [170]. Importantly, HRD scores reflect past, not functional current HR capacity in a tumor [55] [171]. PARPi resistance due to reversion mutations despite high HRD scores yields inconsistent outcomes. Further, HRD scores remained stable after NACT, while mutational and immune signatures changed, further underscoring their limited utility in capturing tumor dynamics or predicting the response to immune checkpoint inhibition [172]. Hence, debate continues on which surrogate best represents functional HRD status. Current candidates are classification of HRD status discriminated by mechanism (genetic/epigenetic), immune-related gene signatures, immunogenomic profiling and immune cell phenotype analysis [69] [72] [108] [173] [174] [175]. For example, antigen presenting cells are key interactors of CD8+PD-1+ T cells, and were more abundant in patients with recurrent ovarian cancer responding to anti-PD-1 therapy. Further validation of clinical application for candidate markers is necessary.


#

Future Prospects in Immunomodulatory Therapy for HGSC

Adoptive immunotherapies, including TIL therapy and genetically engineered chimeric antigen receptor (CAR) immune cells, represent promising future treatment strategies. Targets under investigation in advanced preclinical and phase I clinical trials include Mesothelin (ClinicalTrials.gov: NCT03054298) [176] [177], MUC16 (NCT03907527) [178] [179], HER2 [180], NY-ESO-1 (NCT01567891, NCT03691376, NCT02457650, NCT02869217, NCT03159585), and Claudin 6 (NCT04503278). CD137, a co-stimulatory T cell receptor used in CAR constructs, is being studied for its role in enhancing the efficacy of adoptive T cell therapy [181] [182] [183]. It marks a tumor-reactive CD3+ TIL subset and may serve as a prognostic marker, with potential for better T cell sub-phenotyping [181] [182] [183] [184].

Folate receptor alpha (FRα), targeted by the EMA approved antibody-drug conjugate mirvetuximab soravtansine, is being explored as target of a chimeric costimulatory receptor (CoStAR). TILs engineered to express an extracellular anti-FRα-specific receptor domain and an intracellular CD28/CD40 costimulatory signaling domain showed enhanced T cell effector function, tumor control, and survival in a HGSC patient-derived xenograft model [185]. Preclinical studies include oncolytic adenovirus expressing IL2 in combination with autologous TIL or NK cell injections to reshape the immunosuppressive TME [186] [187]. Whether adoptive cell therapy can overcome tumor heterogeneity and immunosuppressive TME in HGSC remains to be seen in clinical trials. Combinatorial CAR-T cell strategies may enhance tumor cell targeting [188], but improved design of CAR-T cell constructs and administration protocols are needed to reduce off-tumor effects and improve safety profiles. Several ongoing clinical trials are addressing these issues (ClinicalTrials.gov: NCT02498912, NCT05568680).

Understanding T cell metabolism is further critical to identify dysfunctional states during cancer progression and therapy response. CD8+ T cell dysfunction in HGSC has been linked to impaired lipid uptake and metabolic programming due to faulty FABP5 transport, which is mediated by TAGLN2 [189]. Targeting this pathway may restore immune function in ovarian cancer and improve the efficacy of T cell-based immunotherapies. Lastly, dysregulated innate immunological mechanisms are exploited as potential treatment targets since they can dampen immune checkpoint inhibition or adoptive cell therapies [88]. Targeting ER stress by IRE1α ablation in neutrophils in a HGSC mouse model unleashed endogenous and anti-PD-L1-elicited anti-tumor immunity [184]. Complement inhibition to abrogate the suppressive effects of mature neutrophils in the TME represents further therapeutic opportunities, but caution should be taken regarding the complex roles of the complement system in tumor immunity.


#

Conclusion

Disease outcome and treatment success in HGSC are shaped by the interplay between tumor-intrinsic DNA repair capacity, mutational and gene expression signatures, and interactions among tumor, stromal and immune cells within the TME ([Fig. 2]). Recent technological advancements in single-cell analysis and spatial imaging turned the spotlight on cellular diversity, phenotypic states, and the dynamics of immune cell networks that are crucial to the quality of the anti-tumor immune response. Co-existence of multiple distinct TMEs within a patient could explain failure of current immunotherapy approaches. Integration of relative quantity of anatomic tumor location in combination with novel molecular tests might improve the performance of predictive models for immunotherapies. Since TIL density is an insufficient predictive marker for immunotherapies, additional measures of a coordinated immune response including B cells and myeloid lineage cells more faithfully represent susceptibility to adaptive immunotherapy in recent clinical trials.

Zoom Image
Fig. 2 Graphical summary of factors determining tumor heterogeneity in HGSC. Spatial and temporal heterogeneity in HGSC is shaped by the tumor-intrinsic capacity to repair DNA damaged sites, mutational and gene expression signatures that are associated with HR repair gene defects, and crosstalk between tumor, stromal and immune cells in the tumor microenvironment with implications for prognosis and therapy outcome. Created with Biorender.com [rerif]

Targeting the PD-1/PD-L1 engagement between cytotoxic T cells and tumor cells has been central to preclinical and clinical studies. However, immunosuppressive pathways in the TME beyond adaptive immune checkpoints dampen current immunotherapy approaches in HGSC. Innate immune components – such as neutrophils, macrophages, NK cells, and cGAS-STING signaling – significantly affect adaptive immunity. Single immunotherapeutic approaches may be insufficient to reinvigorate anti-tumor immunity in immune-escaped HGSC. Synchronous targeting of innate and adaptive immune regulators may hold promise for future treatment strategies.


#
#

Contributorsʼ Statement

L.R. researched data for the article and wrote the article. All authors contributed substantially to discussion of the content. L.R. and S.P. reviewed and edited the manuscript before submission.

Conflict of Interest

The authors declare that they have no conflict of interest.

  • References

  • 1 Robert Koch-Institut, Zentrum für Krebsregisterdaten. Eierstockkrebs (Ovarialkarzinom) 2022. Accessed October 22, 2024 at: https://www.krebsdaten.de/Krebs/DE/Content/Krebsarten/Eierstockkrebs/eierstockkrebs.html
  • 2 Cancer Statistics Explorer Network. SEER*Explorer: An interactive website for SEER cancer statistics. Accessed December 16, 2024 at: https://seer.cancer.gov/statistics-network/explorer/
  • 3 Ahmed AA, Etemadmoghadam D, Temple J. et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010; 221: 49-56
  • 4 Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011; 474: 609-615
  • 5 Chui MH, Momeni Boroujeni A, Mandelker D. et al. Characterization of TP53-wildtype tubo-ovarian high-grade serous carcinomas: rare exceptions to the binary classification of ovarian serous carcinoma. Mod Pathol 2021; 34: 490-501
  • 6 Contos G, Baca Y, Xiu J. et al. Assessment of immune biomarkers and establishing a triple negative phenotype in gynecologic cancers. Gynecol Oncol 2021; 163: 312-319
  • 7 Birkbak NJ, Wang ZC, Kim JY. et al. Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents. Cancer Discov 2012; 2: 366-375
  • 8 Popova T, Manie E, Rieunier G. et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res 2012; 72: 5454-5462
  • 9 Abkevich V, Timms KM, Hennessy BT. et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br J Cancer 2012; 107: 1776-1782
  • 10 Knijnenburg TA, Wang L, Zimmermann MT. et al. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 2018; 23: 239-254.e6
  • 11 Wang YK, Bashashati A, Anglesio MS. et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat Genet 2017; 49: 856-865
  • 12 Her J, Bunting SF. How cells ensure correct repair of DNA double-strand breaks. J Biol Chem 2018; 293: 10502-10511
  • 13 Klaric JA, Wust S, Panier S. New Faces of old Friends: Emerging new Roles of RNA-Binding Proteins in the DNA Double-Strand Break Response. Front Mol Biosci 2021; 8: 668821
  • 14 Menolfi D, Zha S. ATM, ATR and DNA-PKcs kinases-the lessons from the mouse models: inhibition not equal deletion. Cell Biosci 2020; 10: 8
  • 15 Zhao F, Kim W, Kloeber JA. et al. DNA end resection and its role in DNA replication and DSB repair choice in mammalian cells. Exp Mol Med 2020; 52: 1705-1714
  • 16 Jaspers JE, Kersbergen A, Boon U. et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 2013; 3: 68-81
  • 17 Yamamoto H, Hirasawa A. Homologous Recombination Deficiencies and Hereditary Tumors. Int J Mol Sci 2021; 23: 348
  • 18 Tarsounas M, Sung P. The antitumorigenic roles of BRCA1-BARD1 in DNA repair and replication. Nat Rev Mol Cell Biol 2020; 21: 284-299
  • 19 Kwon J, Lee D, Lee SA. BAP1 as a guardian of genome stability: implications in human cancer. Exp Mol Med 2023; 55: 745-754
  • 20 Elbakry A, Lobrich M. Homologous Recombination Subpathways: A Tangle to Resolve. Front Genet 2021; 12: 723847
  • 21 Li X, Heyer WD. Homologous recombination in DNA repair and DNA damage tolerance. Cell Res 2008; 18: 99-113
  • 22 Sanchez A, Reginato G, Cejka P. Crossover or non-crossover outcomes: tailored processing of homologous recombination intermediates. Curr Opin Genet Dev 2021; 71: 39-47
  • 23 Konstantinopoulos PA, Ceccaldi R, Shapiro GI. et al. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov 2015; 5: 1137-1154
  • 24 Gee ME, Faraahi Z, McCormick A. et al. DNA damage repair in ovarian cancer: unlocking the heterogeneity. J Ovarian Res 2018; 11: 50
  • 25 Lord CJ, Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017; 355: 1152-1158
  • 26 DiSilvestro P, Banerjee S, Colombo N. 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
  • 27 Moore K, Colombo N, Scambia G. et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2018; 379: 2495-2505
  • 28 Monk BJ, Parkinson C, Lim MC. et al. A Randomized, Phase III Trial to Evaluate Rucaparib Monotherapy as Maintenance Treatment in Patients With Newly Diagnosed Ovarian Cancer (ATHENA-MONO/GOG-3020/ENGOT-ov45). J Clin Oncol 2022; 40: 3952-3964
  • 29 Lorusso D, Mouret-Reynier MA, Harter P. et al. Updated progression-free survival and final overall survival with maintenance olaparib plus bevacizumab according to clinical risk in patients with newly diagnosed advanced ovarian cancer in the phase III PAOLA-1/ENGOT-ov25 trial. Int J Gynecol Cancer 2024; 34: 550-558
  • 30 Gonzalez-Martin A, Pothuri B, Vergote I. et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2019; 381: 2391-2402
  • 31 McGee J, Bookman M, Harter P. et al. Fifth Ovarian Cancer Consensus Conference: individualized therapy and patient factors. Ann Oncol 2017; 28: 702-710
  • 32 Gonzalez-Martin A, Harter P, Leary A. et al. Newly diagnosed and relapsed epithelial ovarian cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34: 833-848
  • 33 Baert T, Ferrero A, Sehouli J. et al. The systemic treatment of recurrent ovarian cancer revisited. Ann Oncol 2021; 32: 710-725
  • 34 Galluzzi L, Senovilla L, Vitale I. et al. Molecular mechanisms of cisplatin resistance. Oncogene 2012; 31: 1869-1883
  • 35 Huang D, Savage SR, Calinawan AP. et al. A highly annotated database of genes associated with platinum resistance in cancer. Oncogene 2021; 40: 6395-6405
  • 36 Patch AM, Christie EL, Etemadmoghadam D. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 2015; 521: 489-494
  • 37 Rottenberg S, Disler C, Perego P. The rediscovery of platinum-based cancer therapy. Nat Rev Cancer 2021; 21: 37-50
  • 38 Cooke SL, Brenton JD. Evolution of platinum resistance in high-grade serous ovarian cancer. Lancet Oncol 2011; 12: 1169-1174
  • 39 Maxwell KN, Wubbenhorst B, Wenz BM. et al. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers. Nat Commun 2017; 8: 319
  • 40 Weberpals JI, Pugh TJ, Marco-Casanova P. et al. Tumor genomic, transcriptomic, and immune profiling characterizes differential response to first-line platinum chemotherapy in high grade serous ovarian cancer. Cancer Med 2021; 10: 3045-3058
  • 41 Sung HY, Han J, Chae YJ. et al. Identification of a novel PARP4 gene promoter CpG locus associated with cisplatin chemoresistance. BMB Rep 2023; 56: 347-352
  • 42 Smith P, Bradley T, Gavarro LM. et al. The copy number and mutational landscape of recurrent ovarian high-grade serous carcinoma. Nat Commun 2023; 14: 4387
  • 43 Caldon CE, Musgrove EA. Distinct and redundant functions of cyclin E1 and cyclin E2 in development and cancer. Cell Div 2010; 5: 2
  • 44 Karst AM, Jones PM, Vena N. et al. Cyclin E1 deregulation occurs early in secretory cell transformation to promote formation of fallopian tube-derived high-grade serous ovarian cancers. Cancer Res 2014; 74: 1141-1152
  • 45 Brown VE, Moore SL, Chen M. et al. CDK2 regulates collapsed replication fork repair in CCNE1-amplified ovarian cancer cells via homologous recombination. NAR Cancer 2023; 5: zcad039
  • 46 Kang EY, Weir A, Meagher NS. et al. CCNE1 and survival of patients with tubo-ovarian high-grade serous carcinoma: An Ovarian Tumor Tissue Analysis consortium study. Cancer 2023; 129: 697-713
  • 47 Macintyre G, Goranova TE, De Silva D. et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat Genet 2018; 50: 1262-1270
  • 48 Jamalzadeh S, Dai J, Lavikka K. et al. Genome-wide quantification of copy-number aberration impact on gene expression in ovarian high-grade serous carcinoma. BMC Cancer 2024; 24: 173
  • 49 Rinne N, Christie EL, Ardasheva A. et al. Targeting the PI3K/AKT/mTOR pathway in epithelial ovarian cancer, therapeutic treatment options for platinum-resistant ovarian cancer. Cancer Drug Resist 2021; 4: 573-595
  • 50 Garsed DW, Pandey A, Fereday S. et al. The genomic and immune landscape of long-term survivors of high-grade serous ovarian cancer. Nat Genet 2022; 54: 1853-1864
  • 51 Chowdhury S, Kennedy JJ, Ivey RG. et al. Proteogenomic analysis of chemo-refractory high-grade serous ovarian cancer. Cell 2023; 186: 3476-3498.e35
  • 52 Bouwman P, Jonkers J. Molecular pathways: how can BRCA-mutated tumors become resistant to PARP inhibitors?. Clin Cancer Res 2014; 20: 540-547
  • 53 Kondrashova O, Nguyen M, Shield-Artin K. et al. Secondary Somatic Mutations Restoring RAD51C and RAD51D Associated with Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma. Cancer Discov 2017; 7: 984-998
  • 54 Norquist B, Wurz KA, Pennil CC. et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J Clin Oncol 2011; 29: 3008-3015
  • 55 Swisher EM, Kwan TT, Oza AM. et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat Commun 2021; 12: 2487
  • 56 Burdett NL, Willis MO, Alsop K. et al. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer. Nat Genet 2023; 55: 437-450
  • 57 Wang Y, Bernhardy AJ, Cruz C. et al. The BRCA1-Delta11q Alternative Splice Isoform Bypasses Germline Mutations and Promotes Therapeutic Resistance to PARP Inhibition and Cisplatin. Cancer Res 2016; 76: 2778-2790
  • 58 Zatreanu D, Robinson HMR, Alkhatib O. et al. Poltheta inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance. Nat Commun 2021; 12: 3636
  • 59 Frenel JS, Kim JW, Aryal N. et al. Efficacy of subsequent chemotherapy for patients with BRCA1/2-mutated recurrent epithelial ovarian cancer progressing on olaparib versus placebo maintenance: post-hoc analyses of the SOLO2/ENGOT Ov-21 trial. Ann Oncol 2022; 33: 1021-1028
  • 60 Gadducci A, Cosio S, Landoni F. et al. Response to Chemotherapy and Clinical Outcome of Patients With Recurrent Epithelial Ovarian Cancer After PARP Inhibitor Maintenance Treatment: A Multicenter Retrospective Italian Study. Anticancer Res 2022; 42: 2017-2022
  • 61 Xu-Vuillard A, Guerin-Charbonnel C, Bocquet F. et al. Efficacy of chemotherapy after progression during or following PARPi exposure in ovarian cancer. ESMO Open 2024; 9: 103694
  • 62 Kristeleit R, Lisyanskaya A, Fedenko A. et al. Rucaparib versus standard-of-care chemotherapy in patients with relapsed ovarian cancer and a deleterious BRCA1 or BRCA2 mutation (ARIEL4): an international, open-label, randomised, phase 3 trial. Lancet Oncol 2022; 23: 465-478
  • 63 Tobalina L, Armenia J, Irving E. et al. A meta-analysis of reversion mutations in BRCA genes identifies signatures of DNA end-joining repair mechanisms driving therapy resistance. Ann Oncol 2021; 32: 103-112
  • 64 Pettitt SJ, Frankum JR, Punta M. et al. Clinical BRCA1/2 Reversion Analysis Identifies Hotspot Mutations and Predicted Neoantigens Associated with Therapy Resistance. Cancer Discov 2020; 10: 1475-1488
  • 65 Lheureux S, Oaknin A, Garg S. et al. EVOLVE: A Multicenter Open-Label Single-Arm Clinical and Translational Phase II Trial of Cediranib Plus Olaparib for Ovarian Cancer after PARP Inhibition Progression. Clin Cancer Res 2020; 26: 4206-4215
  • 66 Winkler C, Armenia J, Jones GN. et al. SLFN11 informs on standard of care and novel treatments in a wide range of cancer models. Br J Cancer 2021; 124: 951-962
  • 67 Pujade-Lauraine E, Selle F, Scambia G. et al. Maintenance olaparib rechallenge in patients with platinum-sensitive relapsed ovarian cancer previously treated with a PARP inhibitor (OReO/ENGOT-ov38): a phase IIIb trial. Ann Oncol 2023; 34: 1152-1164
  • 68 Bhamidipati D, Haro-Silerio JI, Yap TA. et al. PARP inhibitors: enhancing efficacy through rational combinations. Br J Cancer 2023; 129: 904-916
  • 69 Yang D, Huang FX, Wei W. et al. Loss of HRD functional phenotype impedes immunotherapy and can be reversed by HDAC inhibitor in ovarian cancer. Int J Biol Sci 2023; 19: 1846-1860
  • 70 Disis ML, Taylor MH, Kelly K. et al. Efficacy and Safety of Avelumab for Patients With Recurrent or Refractory Ovarian Cancer: Phase 1b Results From the JAVELIN Solid Tumor Trial. JAMA Oncol 2019; 5: 393-401
  • 71 Matulonis UA, Shapira-Frommer R, Santin AD. et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann Oncol 2019; 30: 1080-1087
  • 72 Farkkila A, Gulhan DC, Casado J. et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun 2020; 11: 1459
  • 73 Nelson BH, Hamilton P, Phung MT. et al. Immunological and molecular features of the tumor microenvironment of long-term survivors of ovarian cancer. J Clin Invest 2024; 134
  • 74 Zhang AW, McPherson A, Milne K. et al. Interfaces of Malignant and Immunologic Clonal Dynamics in Ovarian Cancer. Cell 2018; 173: 1755-1769.e22
  • 75 Sato E, Olson SH, Ahn J. et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 2005; 102: 18538-18543
  • 76 Waldron L, Haibe-Kains B, Culhane AC. et al. Comparative meta-analysis of prognostic gene signatures for late-stage ovarian cancer. J Natl Cancer Inst 2014; 106: dju049
  • 77 Chen GM, Kannan L, Geistlinger L. et al. Consensus on Molecular Subtypes of High-Grade Serous Ovarian Carcinoma. Clin Cancer Res 2018; 24: 5037-5047
  • 78 Verhaak RG, Tamayo P, Yang JY. et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest 2013; 123: 517-525
  • 79 Scheper W, Kelderman S, Fanchi LF. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2019; 25: 89-94
  • 80 Yeh CY, Aguirre K, Laveroni O. et al. Mapping spatial organization and genetic cell-state regulators to target immune evasion in ovarian cancer. Nat Immunol 2024; 25: 1943-1958
  • 81 Yang B, Li X, Zhang W. et al. Spatial heterogeneity of infiltrating T cells in high-grade serous ovarian cancer revealed by multi-omics analysis. Cell Rep Med 2022; 3: 100856
  • 82 Vazquez-Garcia I, Uhlitz F, Ceglia N. et al. Ovarian cancer mutational processes drive site-specific immune evasion. Nature 2022; 612: 778-786
  • 83 Curiel TJ, Coukos G, Zou L. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10: 942-949
  • 84 Mateiou C, Lokhande L, Diep LH. et al. Spatial tumor immune microenvironment phenotypes in ovarian cancer. NPJ Precis Oncol 2024; 8: 148
  • 85 Izar B, Tirosh I, Stover EH. et al. A single-cell landscape of high-grade serous ovarian cancer. Nat Med 2020; 26: 1271-1279
  • 86 Song M, Sandoval TA, Chae CS. et al. IRE1alpha-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity. Nature 2018; 562: 423-428
  • 87 Kilgour MK, MacPherson S, Zacharias LG. et al. 1-Methylnicotinamide is an immune regulatory metabolite in human ovarian cancer. Sci Adv 2021; 7: eabe1174
  • 88 Singel KL, Emmons TR, Khan ANH. et al. Mature neutrophils suppress T cell immunity in ovarian cancer microenvironment. JCI Insight 2019; 4: e122311
  • 89 Roumenina LT, Daugan MV, Petitprez F. et al. Context-dependent roles of complement in cancer. Nat Rev Cancer 2019; 19: 698-715
  • 90 Olalekan S, Xie B, Back R. et al. Characterizing the tumor microenvironment of metastatic ovarian cancer by single-cell transcriptomics. Cell Rep 2021; 35: 109165
  • 91 Anadon CM, Yu X, Hanggi K. et al. Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell 2022; 40: 545-557.e13
  • 92 Webb JR, Milne K, Watson P. et al. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer. Clin Cancer Res 2014; 20: 434-444
  • 93 Paijens ST, Vledder A, Loiero D. et al. Prognostic image-based quantification of CD8CD103 T cell subsets in high-grade serous ovarian cancer patients. Oncoimmunology 2021; 10: 1935104
  • 94 Kroeger DR, Milne K, Nelson BH. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin Cancer Res 2016; 22: 3005-3015
  • 95 Kasikova L, Rakova J, Hensler M. et al. Tertiary lymphoid structures and B cells determine clinically relevant T cell phenotypes in ovarian cancer. Nat Commun 2024; 15: 2528
  • 96 Xu AM, Haro M, Walts AE. et al. Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma. Sci Adv 2024; 10: eadk8805
  • 97 Olbrecht S, Busschaert P, Qian J. et al. High-grade serous tubo-ovarian cancer refined with single-cell RNA sequencing: specific cell subtypes influence survival and determine molecular subtype classification. Genome Med 2021; 13: 111
  • 98 Ukita M, Hamanishi J, Yoshitomi H. et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight 2022; 7: e157215
  • 99 Schwede M, Waldron L, Mok SC. et al. The Impact of Stroma Admixture on Molecular Subtypes and Prognostic Gene Signatures in Serous Ovarian Cancer. Cancer Epidemiol Biomarkers Prev 2020; 29: 509-519
  • 100 Zhang Q, Wang C, Cliby WA. Cancer-associated stroma significantly contributes to the mesenchymal subtype signature of serous ovarian cancer. Gynecol Oncol 2019; 152: 368-374
  • 101 Desbois M, Udyavar AR, Ryner L. et al. Integrated digital pathology and transcriptome analysis identifies molecular mediators of T-cell exclusion in ovarian cancer. Nat Commun 2020; 11: 5583
  • 102 Stur E, Corvigno S, Xu M. et al. Spatially resolved transcriptomics of high-grade serous ovarian carcinoma. iScience 2022; 25: 103923
  • 103 Zhang L, Cascio S, Mellors JW. et al. Single-cell analysis reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of ovarian cancer. Commun Biol 2024; 7: 20
  • 104 Yang D, Duan MH, Yuan QE. et al. Suppressive stroma-immune prognostic signature impedes immunotherapy in ovarian cancer and can be reversed by PDGFRB inhibitors. J Transl Med 2023; 21: 586
  • 105 Li Y, Tian R, Liu J. et al. Deciphering the immune landscape dominated by cancer-associated fibroblasts to investigate their potential in indicating prognosis and guiding therapeutic regimens in high grade serous ovarian carcinoma. Front Immunol 2022; 13: 940801
  • 106 Yang H, Gu X, Fan R. et al. Deciphering tumor immune microenvironment differences between high-grade serous and endometrioid ovarian cancer to investigate their potential in indicating immunotherapy response. J Ovarian Res 2023; 16: 223
  • 107 Geistlinger L, Oh S, Ramos M. et al. Multiomic Analysis of Subtype Evolution and Heterogeneity in High-Grade Serous Ovarian Carcinoma. Cancer Res 2020; 80: 4335-4345
  • 108 Sveen A, Johannessen B, Klokkerud SM. et al. Evolutionary mode and timing of dissemination of high-grade serous carcinomas. JCI Insight 2024; 9: e170423
  • 109 Denisenko E, de Kock L, Tan A. et al. Spatial transcriptomics reveals discrete tumour microenvironments and autocrine loops within ovarian cancer subclones. Nat Commun 2024; 15: 2860
  • 110 Schwarz RF, Ng CK, Cooke SL. et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med 2015; 12: e1001789
  • 111 Bielski CM, Zehir A, Penson AV. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat Genet 2018; 50: 1189-1195
  • 112 Cheng Z, Mirza H, Ennis DP. et al. The Genomic Landscape of Early-Stage Ovarian High-Grade Serous Carcinoma. Clin Cancer Res 2022; 28: 2911-2922
  • 113 McPherson A, Vazquez-Garcia I, Myers MA. et al. Ongoing genome doubling promotes evolvability and immune dysregulation in ovarian cancer. bioRxiv 2024;
  • 114 Burdett NL, Willis MO, Pandey A. et al. Timing of whole genome duplication is associated with tumor-specific MHC-II depletion in serous ovarian cancer. Nat Commun 2024; 15: 6069
  • 115 Perez-Villatoro F, van Wagensveld L, Shabanova A. et al. Single-cell spatial atlas of high-grade serous ovarian cancer unveils MHC class II as a key driver of spatial tumor ecosystems and clinical outcomes. bioRxiv 2024;
  • 116 Wang XQ, Danenberg E, Huang CS. et al. Spatial predictors of immunotherapy response in triple-negative breast cancer. Nature 2023; 621: 868-876
  • 117 Johnson DB, Estrada MV, Salgado R. et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 2016; 7: 10582
  • 118 Turner TB, Meza-Perez S, Londono A. et al. Epigenetic modifiers upregulate MHC II and impede ovarian cancer tumor growth. Oncotarget 2017; 8: 44159-44170
  • 119 Alexandrov LB, Nik-Zainal S, Wedge DC. et al. Signatures of mutational processes in human cancer. Nature 2013; 500: 415-421
  • 120 Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells. Science 2015; 349: 1483-1489
  • 121 Tian W, Shan B, Zhang Y. et al. Association between DNA damage repair gene somatic mutations and immune-related gene expression in ovarian cancer. Cancer Med 2020; 9: 2190-2200
  • 122 Cristescu R, Aurora-Garg D, Albright A. et al. Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors. J Immunother Cancer 2022; 10: e003091
  • 123 Marcus L, Fashoyin-Aje LA, Donoghue M. et al. FDA Approval Summary: Pembrolizumab for the Treatment of Tumor Mutational Burden-High Solid Tumors. Clin Cancer Res 2021; 27: 4685-4689
  • 124 Strickland KC, Howitt BE, Shukla SA. et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016; 7: 13587-13598
  • 125 Park J, Kim JC, Lee YJ. et al. Unique immune characteristics and differential anti-PD-1-mediated reinvigoration potential of CD8(+) TILs based on BRCA1/2 mutation status in epithelial ovarian cancers. J Immunother Cancer 2024; 12: e009058
  • 126 Launonen IM, Lyytikainen N, Casado J. et al. Single-cell tumor-immune microenvironment of BRCA1/2 mutated high-grade serous ovarian cancer. Nat Commun 2022; 13: 835
  • 127 Przybytkowski E, Davis T, Hosny A. et al. An immune-centric exploration of BRCA1 and BRCA2 germline mutation related breast and ovarian cancers. BMC Cancer 2020; 20: 197
  • 128 Chen M, Linstra R, van Vugt M. Genomic instability, inflammatory signaling and response to cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2022; 1877: 188661
  • 129 Kwon J, Bakhoum SF. The Cytosolic DNA-Sensing cGAS-STING Pathway in Cancer. Cancer Discov 2020; 10: 26-39
  • 130 Bruand M, Barras D, Mina M. et al. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance via STING. Cell Rep 2021; 36: 109412
  • 131 Samstein RM, Krishna C, Ma X. et al. Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy. Nat Cancer 2021; 1: 1188-1203
  • 132 van Wagensveld L, van Baal J, Timmermans M. et al. Homologous Recombination Deficiency and Cyclin E1 Amplification Are Correlated with Immune Cell Infiltration and Survival in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14: 5965
  • 133 Kment J, Newsted D, Young S. et al. Blockade of TGF-beta and PD-L1 by bintrafusp alfa promotes survival in preclinical ovarian cancer models by promoting T effector and NK cell responses. Br J Cancer 2024; 130: 2003-2015
  • 134 Rocconi RP, Grosen EA, Ghamande SA. et al. Gemogenovatucel-T (Vigil) immunotherapy as maintenance in frontline stage III/IV ovarian cancer (VITAL): a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Oncol 2020; 21: 1661-1672
  • 135 Keunecke C, Kulbe H, Dreher F. et al. Predictive biomarker for surgical outcome in patients with advanced primary high-grade serous ovarian cancer. Are we there yet? An analysis of the prospective biobank for ovarian cancer. Gynecol Oncol 2022; 166: 334-343
  • 136 Fotopoulou C, Rockall A, Lu H. et al. Validation analysis of the novel imaging-based prognostic radiomic signature in patients undergoing primary surgery for advanced high-grade serous ovarian cancer (HGSOC). Br J Cancer 2022; 126: 1047-1054
  • 137 Auer K, Bachmayr-Heyda A, Aust S. et al. Peritoneal tumor spread in serous ovarian cancer-epithelial mesenchymal status and outcome. Oncotarget 2015; 6: 17261-17275
  • 138 Auer K, Bachmayr-Heyda A, Sukhbaatar N. et al. Role of the immune system in the peritoneal tumor spread of high grade serous ovarian cancer. Oncotarget 2016; 7: 61336-61354
  • 139 Napoletano C, Bellati F, Landi R. et al. Ovarian cancer cytoreduction induces changes in T cell population subsets reducing immunosuppression. J Cell Mol Med 2010; 14: 2748-2759
  • 140 Nowak M, Glowacka E, Lewkowicz P. et al. Sub-optimal primary surgery leads to unfavorable immunological changes in ovarian cancer patients. Immunobiology 2018; 223: 1-7
  • 141 Torkildsen CF, Austdal M, Jarmund AH. et al. New immune phenotypes for treatment response in high-grade serous ovarian carcinoma patients. Front Immunol 2024; 15: 1394497
  • 142 Wouters MC, Komdeur FL, Workel HH. et al. Treatment Regimen, Surgical Outcome, and T-cell Differentiation Influence Prognostic Benefit of Tumor-Infiltrating Lymphocytes in High-Grade Serous Ovarian Cancer. Clin Cancer Res 2016; 22: 714-724
  • 143 Bohm S, Montfort A, Pearce OM. et al. Neoadjuvant Chemotherapy Modulates the Immune Microenvironment in Metastases of Tubo-Ovarian High-Grade Serous Carcinoma. Clin Cancer Res 2016; 22: 3025-3036
  • 144 Peng J, Hamanishi J, Matsumura N. et al. Chemotherapy Induces Programmed Cell Death-Ligand 1 Overexpression via the Nuclear Factor-kappaB to Foster an Immunosuppressive Tumor Microenvironment in Ovarian Cancer. Cancer Res 2015; 75: 5034-5045
  • 145 Launonen IM, Erkan EP, Niemiec I. et al. Chemotherapy induces myeloid-driven spatial T-cell exhaustion in ovarian cancer. bioRxiv 2024;
  • 146 Jimenez-Sanchez A, Cybulska P, Mager KL. et al. Unraveling tumor-immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy. Nat Genet 2020; 52: 582-593
  • 147 Montfort A, Pearce O, Maniati E. et al. A Strong B-cell Response Is Part of the Immune Landscape in Human High-Grade Serous Ovarian Metastases. Clin Cancer Res 2017; 23: 250-262
  • 148 Lodewijk I, Bernardini A, Suarez-Cabrera C. et al. Genomic landscape and immune-related gene expression profiling of epithelial ovarian cancer after neoadjuvant chemotherapy. NPJ Precis Oncol 2022; 6: 7
  • 149 Mesnage SJL, Auguste A, Genestie C. et al. Neoadjuvant chemotherapy (NACT) increases immune infiltration and programmed death-ligand 1 (PD-L1) expression in epithelial ovarian cancer (EOC). Ann Oncol 2017; 28: 651-657
  • 150 Vanguri R, Benhamida J, Young JH. et al. Understanding the impact of chemotherapy on the immune landscape of high-grade serous ovarian cancer. Gynecol Oncol Rep 2022; 39: 100926
  • 151 Monk BJ, Colombo N, Oza AM. et al. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN Ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22: 1275-1289
  • 152 Ray-Coquard IL, Savoye AM, Schiffler C. et al. Neoadjuvant and adjuvant pembrolizumab in advanced high-grade serous carcinoma: the randomized phase II NeoPembrOV clinical trial. Nat Commun 2024; 15: 5931
  • 153 Le Saux O, Ardin M, Berthet J. et al. Immunomic longitudinal profiling of the NeoPembrOv trial identifies drivers of immunoresistance in high-grade ovarian carcinoma. Nat Commun 2024; 15: 5932
  • 154 Ding L, Kim HJ, Wang Q. et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep 2018; 25: 2972-2980.e5
  • 155 Ding L, Wang Q, Martincuks A. et al. STING agonism overcomes STAT3-mediated immunosuppression and adaptive resistance to PARP inhibition in ovarian cancer. J Immunother Cancer 2023; 11: e005627
  • 156 Martincuks A, Song J, Kohut A. et al. PARP Inhibition Activates STAT3 in Both Tumor and Immune Cells Underlying Therapy Resistance and Immunosuppression In Ovarian Cancer. Front Oncol 2021; 11: 724104
  • 157 Wang Z, Sun K, Xiao Y. et al. Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Sci Rep 2019; 9: 1853
  • 158 Shen J, Zhao W, Ju Z. et al. PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness. Cancer Res 2019; 79: 311-319
  • 159 Konstantinopoulos PA, Waggoner S, Vidal GA. et al. Single-Arm Phases 1 and 2 Trial of Niraparib in Combination With Pembrolizumab in Patients With Recurrent Platinum-Resistant Ovarian Carcinoma. JAMA Oncol 2019; 5: 1141-1149
  • 160 Domchek SM, Postel-Vinay S, Im SA. et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): an open-label, multicentre, phase 1/2, basket study. Lancet Oncol 2020; 21: 1155-1164
  • 161 Gonzalez Martin A, Sanchez Lorenzo L, Colombo N. et al. A phase III, randomized, double blinded trial of platinum based chemotherapy with or without atezolizumab followed by niraparib maintenance with or without atezolizumab in patients with recurrent ovarian, tubal, or peritoneal cancer and platinum treatment free interval of more than 6 months: ENGOT-Ov41/GEICO 69-O/ANITA Trial. Int J Gynecol Cancer 2021; 31: 617-622
  • 162 Monk BJ, Minion LE, Coleman RL. Anti-angiogenic agents in ovarian cancer: past, present, and future. Ann Oncol 2016; 27 (Suppl. 1) i33-i39
  • 163 Garcia-Martinez E, Redondo A, Piulats JM. et al. Are antiangiogenics a good ‘partner’ for immunotherapy in ovarian cancer?. Angiogenesis 2020; 23: 543-557
  • 164 Bouzin C, Brouet A, De Vriese J. et al. Effects of vascular endothelial growth factor on the lymphocyte-endothelium interactions: identification of caveolin-1 and nitric oxide as control points of endothelial cell anergy. J Immunol 2007; 178: 1505-1511
  • 165 Tavira B, Iscar T, Manso L. et al. Analysis of Tumor Microenvironment Changes after Neoadjuvant Chemotherapy with or without Bevacizumab in Advanced Ovarian Cancer (GEICO-89T/MINOVA Study). Clin Cancer Res 2024; 30: 176-186
  • 166 Spigel DR, Cheng Y, Cho BC. et al. ADRIATIC: Durvalumab (D) as consolidation treatment (tx) for patients (pts) with limited-stage small-cell lung cancer (LS-SCLC). J Clin Oncol 2024; 42 (Suppl. 17)
  • 167 Kurtz JE, Pujade-Lauraine E, Oaknin A. et al. Atezolizumab Combined With Bevacizumab and Platinum-Based Therapy for Platinum-Sensitive Ovarian Cancer: Placebo-Controlled Randomized Phase III ATALANTE/ENGOT-ov29 Trial. J Clin Oncol 2023; 41: 4768-4778
  • 168 Liu YL, Selenica P, Zhou Q. et al. BRCA Mutations, Homologous DNA Repair Deficiency, Tumor Mutational Burden, and Response to Immune Checkpoint Inhibition in Recurrent Ovarian Cancer. JCO Precis Oncol 2020; 4: PO.20.00069
  • 169 Landen CN, Molinero L, Hamidi H. et al. Influence of Genomic Landscape on Cancer Immunotherapy for Newly Diagnosed Ovarian Cancer: Biomarker Analyses from the IMagyn050 Randomized Clinical Trial. Clin Cancer Res 2023; 29: 1698-1707
  • 170 Stover EH, Fuh K, Konstantinopoulos PA. et al. Clinical assays for assessment of homologous recombination DNA repair deficiency. Gynecol Oncol 2020; 159: 887-898
  • 171 Patel JN, Braicu I, Timms KM. et al. Characterisation of homologous recombination deficiency in paired primary and recurrent high-grade serous ovarian cancer. Br J Cancer 2018; 119: 1060-1066
  • 172 Takaya H, Nakai H, Sakai K. et al. Intratumor heterogeneity and homologous recombination deficiency of high-grade serous ovarian cancer are associated with prognosis and molecular subtype and change in treatment course. Gynecol Oncol 2020; 156: 415-422
  • 173 Takaya H, Nakai H, Takamatsu S. et al. Homologous recombination deficiency status-based classification of high-grade serous ovarian carcinoma. Sci Rep 2020; 10: 2757
  • 174 Duraiswamy J, Turrini R, Minasyan A. et al. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 2021; 39: 1623-1642 e1620
  • 175 Ledermann JA, Shapira-Frommer R, Santin AD. et al. Molecular determinants of clinical outcomes of pembrolizumab in recurrent ovarian cancer: Exploratory analysis of KEYNOTE-100. Gynecol Oncol 2023; 178: 119-129
  • 176 Haas AR, Tanyi JL, O’Hara MH. et al. Phase I Study of Lentiviral-Transduced Chimeric Antigen Receptor-Modified T Cells Recognizing Mesothelin in Advanced Solid Cancers. Mol Ther 2019; 27: 1919-1929
  • 177 Chen J, Hu J, Gu L. et al. Anti-mesothelin CAR-T immunotherapy in patients with ovarian cancer. Cancer Immunol Immunother 2023; 72: 409-425
  • 178 Crawford A, Haber L, Kelly MP. et al. A Mucin 16 bispecific T cell-engaging antibody for the treatment of ovarian cancer. Sci Transl Med 2019; 11: eaau7534
  • 179 Li T, Wang J. Therapeutic effect of dual CAR-T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer 2020; 20: 678
  • 180 Shalaby N, Xia Y, Kelly JJ. et al. 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
  • 181 Tubridy EA, Eiva MA, Liu F. et al. CD137+ tumor infiltrating lymphocytes predicts ovarian cancer survival. Gynecol Oncol 2024; 184: 74-82
  • 182 Tassi E, Bergamini A, Wignall J. et al. Epithelial ovarian cancer is infiltrated by activated effector T cells co-expressing CD39, PD-1, TIM-3, CD137 and interacting with cancer cells and myeloid cells. Front Immunol 2023; 14: 1212444
  • 183 Amaria R, Knisely A, Vining D. et al. Efficacy and safety of autologous tumor-infiltrating lymphocytes in recurrent or refractory ovarian cancer, colorectal cancer, and pancreatic ductal adenocarcinoma. J Immunother Cancer 2024; 12: e006822
  • 184 Emmanuelli A, Salvagno C, Hwang SM. et al. High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1alpha activity in neutrophils. Oncoimmunology 2024; 13: 2411070
  • 185 Kalaitsidou M, Moon OR, Sykorova M. et al. Signaling via a CD28/CD40 chimeric costimulatory antigen receptor (CoStAR), targeting folate receptor alpha, enhances T cell activity and augments tumor reactivity of tumor infiltrating lymphocytes. Front Immunol 2023; 14: 1256491
  • 186 Quixabeira DCA, Jirovec E, Pakola S. et al. Improving the cytotoxic response of tumor-infiltrating lymphocytes towards advanced stage ovarian cancer with an oncolytic adenovirus expressing a human vIL-2 cytokine. Cancer Gene Ther 2023; 30: 1543-1553
  • 187 Capellero S, Erriquez J, Melano C. et al. Preclinical immunotherapy with Cytokine-Induced Killer lymphocytes against epithelial ovarian cancer. Sci Rep 2020; 10: 6478
  • 188 Banville AC, Wouters MCA, Oberg AL. et al. Co-expression patterns of chimeric antigen receptor (CAR)-T cell target antigens in primary and recurrent ovarian cancer. Gynecol Oncol 2021; 160: 520-529
  • 189 Hwang SM, Awasthi D, Jeong J. et al. Transgelin 2 guards T cell lipid metabolism and antitumour function. Nature 2024; 635: 1010-1018

Correspondence

Dr. Leonie Ratz
Department of Obstetrics and Gynecology, University Hospital Cologne
Kerpener Str. 34
50931 Cologne
Germany   

Publication History

Received: 11 January 2025

Accepted after revision: 11 May 2025

Article published online:
11 June 2025

© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany

  • References

  • 1 Robert Koch-Institut, Zentrum für Krebsregisterdaten. Eierstockkrebs (Ovarialkarzinom) 2022. Accessed October 22, 2024 at: https://www.krebsdaten.de/Krebs/DE/Content/Krebsarten/Eierstockkrebs/eierstockkrebs.html
  • 2 Cancer Statistics Explorer Network. SEER*Explorer: An interactive website for SEER cancer statistics. Accessed December 16, 2024 at: https://seer.cancer.gov/statistics-network/explorer/
  • 3 Ahmed AA, Etemadmoghadam D, Temple J. et al. Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary. J Pathol 2010; 221: 49-56
  • 4 Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 2011; 474: 609-615
  • 5 Chui MH, Momeni Boroujeni A, Mandelker D. et al. Characterization of TP53-wildtype tubo-ovarian high-grade serous carcinomas: rare exceptions to the binary classification of ovarian serous carcinoma. Mod Pathol 2021; 34: 490-501
  • 6 Contos G, Baca Y, Xiu J. et al. Assessment of immune biomarkers and establishing a triple negative phenotype in gynecologic cancers. Gynecol Oncol 2021; 163: 312-319
  • 7 Birkbak NJ, Wang ZC, Kim JY. et al. Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents. Cancer Discov 2012; 2: 366-375
  • 8 Popova T, Manie E, Rieunier G. et al. Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation. Cancer Res 2012; 72: 5454-5462
  • 9 Abkevich V, Timms KM, Hennessy BT. et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. Br J Cancer 2012; 107: 1776-1782
  • 10 Knijnenburg TA, Wang L, Zimmermann MT. et al. Genomic and Molecular Landscape of DNA Damage Repair Deficiency across The Cancer Genome Atlas. Cell Rep 2018; 23: 239-254.e6
  • 11 Wang YK, Bashashati A, Anglesio MS. et al. Genomic consequences of aberrant DNA repair mechanisms stratify ovarian cancer histotypes. Nat Genet 2017; 49: 856-865
  • 12 Her J, Bunting SF. How cells ensure correct repair of DNA double-strand breaks. J Biol Chem 2018; 293: 10502-10511
  • 13 Klaric JA, Wust S, Panier S. New Faces of old Friends: Emerging new Roles of RNA-Binding Proteins in the DNA Double-Strand Break Response. Front Mol Biosci 2021; 8: 668821
  • 14 Menolfi D, Zha S. ATM, ATR and DNA-PKcs kinases-the lessons from the mouse models: inhibition not equal deletion. Cell Biosci 2020; 10: 8
  • 15 Zhao F, Kim W, Kloeber JA. et al. DNA end resection and its role in DNA replication and DSB repair choice in mammalian cells. Exp Mol Med 2020; 52: 1705-1714
  • 16 Jaspers JE, Kersbergen A, Boon U. et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors. Cancer Discov 2013; 3: 68-81
  • 17 Yamamoto H, Hirasawa A. Homologous Recombination Deficiencies and Hereditary Tumors. Int J Mol Sci 2021; 23: 348
  • 18 Tarsounas M, Sung P. The antitumorigenic roles of BRCA1-BARD1 in DNA repair and replication. Nat Rev Mol Cell Biol 2020; 21: 284-299
  • 19 Kwon J, Lee D, Lee SA. BAP1 as a guardian of genome stability: implications in human cancer. Exp Mol Med 2023; 55: 745-754
  • 20 Elbakry A, Lobrich M. Homologous Recombination Subpathways: A Tangle to Resolve. Front Genet 2021; 12: 723847
  • 21 Li X, Heyer WD. Homologous recombination in DNA repair and DNA damage tolerance. Cell Res 2008; 18: 99-113
  • 22 Sanchez A, Reginato G, Cejka P. Crossover or non-crossover outcomes: tailored processing of homologous recombination intermediates. Curr Opin Genet Dev 2021; 71: 39-47
  • 23 Konstantinopoulos PA, Ceccaldi R, Shapiro GI. et al. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov 2015; 5: 1137-1154
  • 24 Gee ME, Faraahi Z, McCormick A. et al. DNA damage repair in ovarian cancer: unlocking the heterogeneity. J Ovarian Res 2018; 11: 50
  • 25 Lord CJ, Ashworth A. PARP inhibitors: Synthetic lethality in the clinic. Science 2017; 355: 1152-1158
  • 26 DiSilvestro P, Banerjee S, Colombo N. 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
  • 27 Moore K, Colombo N, Scambia G. et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2018; 379: 2495-2505
  • 28 Monk BJ, Parkinson C, Lim MC. et al. A Randomized, Phase III Trial to Evaluate Rucaparib Monotherapy as Maintenance Treatment in Patients With Newly Diagnosed Ovarian Cancer (ATHENA-MONO/GOG-3020/ENGOT-ov45). J Clin Oncol 2022; 40: 3952-3964
  • 29 Lorusso D, Mouret-Reynier MA, Harter P. et al. Updated progression-free survival and final overall survival with maintenance olaparib plus bevacizumab according to clinical risk in patients with newly diagnosed advanced ovarian cancer in the phase III PAOLA-1/ENGOT-ov25 trial. Int J Gynecol Cancer 2024; 34: 550-558
  • 30 Gonzalez-Martin A, Pothuri B, Vergote I. et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2019; 381: 2391-2402
  • 31 McGee J, Bookman M, Harter P. et al. Fifth Ovarian Cancer Consensus Conference: individualized therapy and patient factors. Ann Oncol 2017; 28: 702-710
  • 32 Gonzalez-Martin A, Harter P, Leary A. et al. Newly diagnosed and relapsed epithelial ovarian cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023; 34: 833-848
  • 33 Baert T, Ferrero A, Sehouli J. et al. The systemic treatment of recurrent ovarian cancer revisited. Ann Oncol 2021; 32: 710-725
  • 34 Galluzzi L, Senovilla L, Vitale I. et al. Molecular mechanisms of cisplatin resistance. Oncogene 2012; 31: 1869-1883
  • 35 Huang D, Savage SR, Calinawan AP. et al. A highly annotated database of genes associated with platinum resistance in cancer. Oncogene 2021; 40: 6395-6405
  • 36 Patch AM, Christie EL, Etemadmoghadam D. et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature 2015; 521: 489-494
  • 37 Rottenberg S, Disler C, Perego P. The rediscovery of platinum-based cancer therapy. Nat Rev Cancer 2021; 21: 37-50
  • 38 Cooke SL, Brenton JD. Evolution of platinum resistance in high-grade serous ovarian cancer. Lancet Oncol 2011; 12: 1169-1174
  • 39 Maxwell KN, Wubbenhorst B, Wenz BM. et al. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers. Nat Commun 2017; 8: 319
  • 40 Weberpals JI, Pugh TJ, Marco-Casanova P. et al. Tumor genomic, transcriptomic, and immune profiling characterizes differential response to first-line platinum chemotherapy in high grade serous ovarian cancer. Cancer Med 2021; 10: 3045-3058
  • 41 Sung HY, Han J, Chae YJ. et al. Identification of a novel PARP4 gene promoter CpG locus associated with cisplatin chemoresistance. BMB Rep 2023; 56: 347-352
  • 42 Smith P, Bradley T, Gavarro LM. et al. The copy number and mutational landscape of recurrent ovarian high-grade serous carcinoma. Nat Commun 2023; 14: 4387
  • 43 Caldon CE, Musgrove EA. Distinct and redundant functions of cyclin E1 and cyclin E2 in development and cancer. Cell Div 2010; 5: 2
  • 44 Karst AM, Jones PM, Vena N. et al. Cyclin E1 deregulation occurs early in secretory cell transformation to promote formation of fallopian tube-derived high-grade serous ovarian cancers. Cancer Res 2014; 74: 1141-1152
  • 45 Brown VE, Moore SL, Chen M. et al. CDK2 regulates collapsed replication fork repair in CCNE1-amplified ovarian cancer cells via homologous recombination. NAR Cancer 2023; 5: zcad039
  • 46 Kang EY, Weir A, Meagher NS. et al. CCNE1 and survival of patients with tubo-ovarian high-grade serous carcinoma: An Ovarian Tumor Tissue Analysis consortium study. Cancer 2023; 129: 697-713
  • 47 Macintyre G, Goranova TE, De Silva D. et al. Copy number signatures and mutational processes in ovarian carcinoma. Nat Genet 2018; 50: 1262-1270
  • 48 Jamalzadeh S, Dai J, Lavikka K. et al. Genome-wide quantification of copy-number aberration impact on gene expression in ovarian high-grade serous carcinoma. BMC Cancer 2024; 24: 173
  • 49 Rinne N, Christie EL, Ardasheva A. et al. Targeting the PI3K/AKT/mTOR pathway in epithelial ovarian cancer, therapeutic treatment options for platinum-resistant ovarian cancer. Cancer Drug Resist 2021; 4: 573-595
  • 50 Garsed DW, Pandey A, Fereday S. et al. The genomic and immune landscape of long-term survivors of high-grade serous ovarian cancer. Nat Genet 2022; 54: 1853-1864
  • 51 Chowdhury S, Kennedy JJ, Ivey RG. et al. Proteogenomic analysis of chemo-refractory high-grade serous ovarian cancer. Cell 2023; 186: 3476-3498.e35
  • 52 Bouwman P, Jonkers J. Molecular pathways: how can BRCA-mutated tumors become resistant to PARP inhibitors?. Clin Cancer Res 2014; 20: 540-547
  • 53 Kondrashova O, Nguyen M, Shield-Artin K. et al. Secondary Somatic Mutations Restoring RAD51C and RAD51D Associated with Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma. Cancer Discov 2017; 7: 984-998
  • 54 Norquist B, Wurz KA, Pennil CC. et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J Clin Oncol 2011; 29: 3008-3015
  • 55 Swisher EM, Kwan TT, Oza AM. et al. Molecular and clinical determinants of response and resistance to rucaparib for recurrent ovarian cancer treatment in ARIEL2 (Parts 1 and 2). Nat Commun 2021; 12: 2487
  • 56 Burdett NL, Willis MO, Alsop K. et al. Multiomic analysis of homologous recombination-deficient end-stage high-grade serous ovarian cancer. Nat Genet 2023; 55: 437-450
  • 57 Wang Y, Bernhardy AJ, Cruz C. et al. The BRCA1-Delta11q Alternative Splice Isoform Bypasses Germline Mutations and Promotes Therapeutic Resistance to PARP Inhibition and Cisplatin. Cancer Res 2016; 76: 2778-2790
  • 58 Zatreanu D, Robinson HMR, Alkhatib O. et al. Poltheta inhibitors elicit BRCA-gene synthetic lethality and target PARP inhibitor resistance. Nat Commun 2021; 12: 3636
  • 59 Frenel JS, Kim JW, Aryal N. et al. Efficacy of subsequent chemotherapy for patients with BRCA1/2-mutated recurrent epithelial ovarian cancer progressing on olaparib versus placebo maintenance: post-hoc analyses of the SOLO2/ENGOT Ov-21 trial. Ann Oncol 2022; 33: 1021-1028
  • 60 Gadducci A, Cosio S, Landoni F. et al. Response to Chemotherapy and Clinical Outcome of Patients With Recurrent Epithelial Ovarian Cancer After PARP Inhibitor Maintenance Treatment: A Multicenter Retrospective Italian Study. Anticancer Res 2022; 42: 2017-2022
  • 61 Xu-Vuillard A, Guerin-Charbonnel C, Bocquet F. et al. Efficacy of chemotherapy after progression during or following PARPi exposure in ovarian cancer. ESMO Open 2024; 9: 103694
  • 62 Kristeleit R, Lisyanskaya A, Fedenko A. et al. Rucaparib versus standard-of-care chemotherapy in patients with relapsed ovarian cancer and a deleterious BRCA1 or BRCA2 mutation (ARIEL4): an international, open-label, randomised, phase 3 trial. Lancet Oncol 2022; 23: 465-478
  • 63 Tobalina L, Armenia J, Irving E. et al. A meta-analysis of reversion mutations in BRCA genes identifies signatures of DNA end-joining repair mechanisms driving therapy resistance. Ann Oncol 2021; 32: 103-112
  • 64 Pettitt SJ, Frankum JR, Punta M. et al. Clinical BRCA1/2 Reversion Analysis Identifies Hotspot Mutations and Predicted Neoantigens Associated with Therapy Resistance. Cancer Discov 2020; 10: 1475-1488
  • 65 Lheureux S, Oaknin A, Garg S. et al. EVOLVE: A Multicenter Open-Label Single-Arm Clinical and Translational Phase II Trial of Cediranib Plus Olaparib for Ovarian Cancer after PARP Inhibition Progression. Clin Cancer Res 2020; 26: 4206-4215
  • 66 Winkler C, Armenia J, Jones GN. et al. SLFN11 informs on standard of care and novel treatments in a wide range of cancer models. Br J Cancer 2021; 124: 951-962
  • 67 Pujade-Lauraine E, Selle F, Scambia G. et al. Maintenance olaparib rechallenge in patients with platinum-sensitive relapsed ovarian cancer previously treated with a PARP inhibitor (OReO/ENGOT-ov38): a phase IIIb trial. Ann Oncol 2023; 34: 1152-1164
  • 68 Bhamidipati D, Haro-Silerio JI, Yap TA. et al. PARP inhibitors: enhancing efficacy through rational combinations. Br J Cancer 2023; 129: 904-916
  • 69 Yang D, Huang FX, Wei W. et al. Loss of HRD functional phenotype impedes immunotherapy and can be reversed by HDAC inhibitor in ovarian cancer. Int J Biol Sci 2023; 19: 1846-1860
  • 70 Disis ML, Taylor MH, Kelly K. et al. Efficacy and Safety of Avelumab for Patients With Recurrent or Refractory Ovarian Cancer: Phase 1b Results From the JAVELIN Solid Tumor Trial. JAMA Oncol 2019; 5: 393-401
  • 71 Matulonis UA, Shapira-Frommer R, Santin AD. et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE-100 study. Ann Oncol 2019; 30: 1080-1087
  • 72 Farkkila A, Gulhan DC, Casado J. et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun 2020; 11: 1459
  • 73 Nelson BH, Hamilton P, Phung MT. et al. Immunological and molecular features of the tumor microenvironment of long-term survivors of ovarian cancer. J Clin Invest 2024; 134
  • 74 Zhang AW, McPherson A, Milne K. et al. Interfaces of Malignant and Immunologic Clonal Dynamics in Ovarian Cancer. Cell 2018; 173: 1755-1769.e22
  • 75 Sato E, Olson SH, Ahn J. et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A 2005; 102: 18538-18543
  • 76 Waldron L, Haibe-Kains B, Culhane AC. et al. Comparative meta-analysis of prognostic gene signatures for late-stage ovarian cancer. J Natl Cancer Inst 2014; 106: dju049
  • 77 Chen GM, Kannan L, Geistlinger L. et al. Consensus on Molecular Subtypes of High-Grade Serous Ovarian Carcinoma. Clin Cancer Res 2018; 24: 5037-5047
  • 78 Verhaak RG, Tamayo P, Yang JY. et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest 2013; 123: 517-525
  • 79 Scheper W, Kelderman S, Fanchi LF. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat Med 2019; 25: 89-94
  • 80 Yeh CY, Aguirre K, Laveroni O. et al. Mapping spatial organization and genetic cell-state regulators to target immune evasion in ovarian cancer. Nat Immunol 2024; 25: 1943-1958
  • 81 Yang B, Li X, Zhang W. et al. Spatial heterogeneity of infiltrating T cells in high-grade serous ovarian cancer revealed by multi-omics analysis. Cell Rep Med 2022; 3: 100856
  • 82 Vazquez-Garcia I, Uhlitz F, Ceglia N. et al. Ovarian cancer mutational processes drive site-specific immune evasion. Nature 2022; 612: 778-786
  • 83 Curiel TJ, Coukos G, Zou L. et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med 2004; 10: 942-949
  • 84 Mateiou C, Lokhande L, Diep LH. et al. Spatial tumor immune microenvironment phenotypes in ovarian cancer. NPJ Precis Oncol 2024; 8: 148
  • 85 Izar B, Tirosh I, Stover EH. et al. A single-cell landscape of high-grade serous ovarian cancer. Nat Med 2020; 26: 1271-1279
  • 86 Song M, Sandoval TA, Chae CS. et al. IRE1alpha-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity. Nature 2018; 562: 423-428
  • 87 Kilgour MK, MacPherson S, Zacharias LG. et al. 1-Methylnicotinamide is an immune regulatory metabolite in human ovarian cancer. Sci Adv 2021; 7: eabe1174
  • 88 Singel KL, Emmons TR, Khan ANH. et al. Mature neutrophils suppress T cell immunity in ovarian cancer microenvironment. JCI Insight 2019; 4: e122311
  • 89 Roumenina LT, Daugan MV, Petitprez F. et al. Context-dependent roles of complement in cancer. Nat Rev Cancer 2019; 19: 698-715
  • 90 Olalekan S, Xie B, Back R. et al. Characterizing the tumor microenvironment of metastatic ovarian cancer by single-cell transcriptomics. Cell Rep 2021; 35: 109165
  • 91 Anadon CM, Yu X, Hanggi K. et al. Ovarian cancer immunogenicity is governed by a narrow subset of progenitor tissue-resident memory T cells. Cancer Cell 2022; 40: 545-557.e13
  • 92 Webb JR, Milne K, Watson P. et al. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer. Clin Cancer Res 2014; 20: 434-444
  • 93 Paijens ST, Vledder A, Loiero D. et al. Prognostic image-based quantification of CD8CD103 T cell subsets in high-grade serous ovarian cancer patients. Oncoimmunology 2021; 10: 1935104
  • 94 Kroeger DR, Milne K, Nelson BH. Tumor-Infiltrating Plasma Cells Are Associated with Tertiary Lymphoid Structures, Cytolytic T-Cell Responses, and Superior Prognosis in Ovarian Cancer. Clin Cancer Res 2016; 22: 3005-3015
  • 95 Kasikova L, Rakova J, Hensler M. et al. Tertiary lymphoid structures and B cells determine clinically relevant T cell phenotypes in ovarian cancer. Nat Commun 2024; 15: 2528
  • 96 Xu AM, Haro M, Walts AE. et al. Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma. Sci Adv 2024; 10: eadk8805
  • 97 Olbrecht S, Busschaert P, Qian J. et al. High-grade serous tubo-ovarian cancer refined with single-cell RNA sequencing: specific cell subtypes influence survival and determine molecular subtype classification. Genome Med 2021; 13: 111
  • 98 Ukita M, Hamanishi J, Yoshitomi H. et al. CXCL13-producing CD4+ T cells accumulate in the early phase of tertiary lymphoid structures in ovarian cancer. JCI Insight 2022; 7: e157215
  • 99 Schwede M, Waldron L, Mok SC. et al. The Impact of Stroma Admixture on Molecular Subtypes and Prognostic Gene Signatures in Serous Ovarian Cancer. Cancer Epidemiol Biomarkers Prev 2020; 29: 509-519
  • 100 Zhang Q, Wang C, Cliby WA. Cancer-associated stroma significantly contributes to the mesenchymal subtype signature of serous ovarian cancer. Gynecol Oncol 2019; 152: 368-374
  • 101 Desbois M, Udyavar AR, Ryner L. et al. Integrated digital pathology and transcriptome analysis identifies molecular mediators of T-cell exclusion in ovarian cancer. Nat Commun 2020; 11: 5583
  • 102 Stur E, Corvigno S, Xu M. et al. Spatially resolved transcriptomics of high-grade serous ovarian carcinoma. iScience 2022; 25: 103923
  • 103 Zhang L, Cascio S, Mellors JW. et al. Single-cell analysis reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of ovarian cancer. Commun Biol 2024; 7: 20
  • 104 Yang D, Duan MH, Yuan QE. et al. Suppressive stroma-immune prognostic signature impedes immunotherapy in ovarian cancer and can be reversed by PDGFRB inhibitors. J Transl Med 2023; 21: 586
  • 105 Li Y, Tian R, Liu J. et al. Deciphering the immune landscape dominated by cancer-associated fibroblasts to investigate their potential in indicating prognosis and guiding therapeutic regimens in high grade serous ovarian carcinoma. Front Immunol 2022; 13: 940801
  • 106 Yang H, Gu X, Fan R. et al. Deciphering tumor immune microenvironment differences between high-grade serous and endometrioid ovarian cancer to investigate their potential in indicating immunotherapy response. J Ovarian Res 2023; 16: 223
  • 107 Geistlinger L, Oh S, Ramos M. et al. Multiomic Analysis of Subtype Evolution and Heterogeneity in High-Grade Serous Ovarian Carcinoma. Cancer Res 2020; 80: 4335-4345
  • 108 Sveen A, Johannessen B, Klokkerud SM. et al. Evolutionary mode and timing of dissemination of high-grade serous carcinomas. JCI Insight 2024; 9: e170423
  • 109 Denisenko E, de Kock L, Tan A. et al. Spatial transcriptomics reveals discrete tumour microenvironments and autocrine loops within ovarian cancer subclones. Nat Commun 2024; 15: 2860
  • 110 Schwarz RF, Ng CK, Cooke SL. et al. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med 2015; 12: e1001789
  • 111 Bielski CM, Zehir A, Penson AV. et al. Genome doubling shapes the evolution and prognosis of advanced cancers. Nat Genet 2018; 50: 1189-1195
  • 112 Cheng Z, Mirza H, Ennis DP. et al. The Genomic Landscape of Early-Stage Ovarian High-Grade Serous Carcinoma. Clin Cancer Res 2022; 28: 2911-2922
  • 113 McPherson A, Vazquez-Garcia I, Myers MA. et al. Ongoing genome doubling promotes evolvability and immune dysregulation in ovarian cancer. bioRxiv 2024;
  • 114 Burdett NL, Willis MO, Pandey A. et al. Timing of whole genome duplication is associated with tumor-specific MHC-II depletion in serous ovarian cancer. Nat Commun 2024; 15: 6069
  • 115 Perez-Villatoro F, van Wagensveld L, Shabanova A. et al. Single-cell spatial atlas of high-grade serous ovarian cancer unveils MHC class II as a key driver of spatial tumor ecosystems and clinical outcomes. bioRxiv 2024;
  • 116 Wang XQ, Danenberg E, Huang CS. et al. Spatial predictors of immunotherapy response in triple-negative breast cancer. Nature 2023; 621: 868-876
  • 117 Johnson DB, Estrada MV, Salgado R. et al. Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 2016; 7: 10582
  • 118 Turner TB, Meza-Perez S, Londono A. et al. Epigenetic modifiers upregulate MHC II and impede ovarian cancer tumor growth. Oncotarget 2017; 8: 44159-44170
  • 119 Alexandrov LB, Nik-Zainal S, Wedge DC. et al. Signatures of mutational processes in human cancer. Nature 2013; 500: 415-421
  • 120 Martincorena I, Campbell PJ. Somatic mutation in cancer and normal cells. Science 2015; 349: 1483-1489
  • 121 Tian W, Shan B, Zhang Y. et al. Association between DNA damage repair gene somatic mutations and immune-related gene expression in ovarian cancer. Cancer Med 2020; 9: 2190-2200
  • 122 Cristescu R, Aurora-Garg D, Albright A. et al. Tumor mutational burden predicts the efficacy of pembrolizumab monotherapy: a pan-tumor retrospective analysis of participants with advanced solid tumors. J Immunother Cancer 2022; 10: e003091
  • 123 Marcus L, Fashoyin-Aje LA, Donoghue M. et al. FDA Approval Summary: Pembrolizumab for the Treatment of Tumor Mutational Burden-High Solid Tumors. Clin Cancer Res 2021; 27: 4685-4689
  • 124 Strickland KC, Howitt BE, Shukla SA. et al. Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer. Oncotarget 2016; 7: 13587-13598
  • 125 Park J, Kim JC, Lee YJ. et al. Unique immune characteristics and differential anti-PD-1-mediated reinvigoration potential of CD8(+) TILs based on BRCA1/2 mutation status in epithelial ovarian cancers. J Immunother Cancer 2024; 12: e009058
  • 126 Launonen IM, Lyytikainen N, Casado J. et al. Single-cell tumor-immune microenvironment of BRCA1/2 mutated high-grade serous ovarian cancer. Nat Commun 2022; 13: 835
  • 127 Przybytkowski E, Davis T, Hosny A. et al. An immune-centric exploration of BRCA1 and BRCA2 germline mutation related breast and ovarian cancers. BMC Cancer 2020; 20: 197
  • 128 Chen M, Linstra R, van Vugt M. Genomic instability, inflammatory signaling and response to cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2022; 1877: 188661
  • 129 Kwon J, Bakhoum SF. The Cytosolic DNA-Sensing cGAS-STING Pathway in Cancer. Cancer Discov 2020; 10: 26-39
  • 130 Bruand M, Barras D, Mina M. et al. Cell-autonomous inflammation of BRCA1-deficient ovarian cancers drives both tumor-intrinsic immunoreactivity and immune resistance via STING. Cell Rep 2021; 36: 109412
  • 131 Samstein RM, Krishna C, Ma X. et al. Mutations in BRCA1 and BRCA2 differentially affect the tumor microenvironment and response to checkpoint blockade immunotherapy. Nat Cancer 2021; 1: 1188-1203
  • 132 van Wagensveld L, van Baal J, Timmermans M. et al. Homologous Recombination Deficiency and Cyclin E1 Amplification Are Correlated with Immune Cell Infiltration and Survival in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2022; 14: 5965
  • 133 Kment J, Newsted D, Young S. et al. Blockade of TGF-beta and PD-L1 by bintrafusp alfa promotes survival in preclinical ovarian cancer models by promoting T effector and NK cell responses. Br J Cancer 2024; 130: 2003-2015
  • 134 Rocconi RP, Grosen EA, Ghamande SA. et al. Gemogenovatucel-T (Vigil) immunotherapy as maintenance in frontline stage III/IV ovarian cancer (VITAL): a randomised, double-blind, placebo-controlled, phase 2b trial. Lancet Oncol 2020; 21: 1661-1672
  • 135 Keunecke C, Kulbe H, Dreher F. et al. Predictive biomarker for surgical outcome in patients with advanced primary high-grade serous ovarian cancer. Are we there yet? An analysis of the prospective biobank for ovarian cancer. Gynecol Oncol 2022; 166: 334-343
  • 136 Fotopoulou C, Rockall A, Lu H. et al. Validation analysis of the novel imaging-based prognostic radiomic signature in patients undergoing primary surgery for advanced high-grade serous ovarian cancer (HGSOC). Br J Cancer 2022; 126: 1047-1054
  • 137 Auer K, Bachmayr-Heyda A, Aust S. et al. Peritoneal tumor spread in serous ovarian cancer-epithelial mesenchymal status and outcome. Oncotarget 2015; 6: 17261-17275
  • 138 Auer K, Bachmayr-Heyda A, Sukhbaatar N. et al. Role of the immune system in the peritoneal tumor spread of high grade serous ovarian cancer. Oncotarget 2016; 7: 61336-61354
  • 139 Napoletano C, Bellati F, Landi R. et al. Ovarian cancer cytoreduction induces changes in T cell population subsets reducing immunosuppression. J Cell Mol Med 2010; 14: 2748-2759
  • 140 Nowak M, Glowacka E, Lewkowicz P. et al. Sub-optimal primary surgery leads to unfavorable immunological changes in ovarian cancer patients. Immunobiology 2018; 223: 1-7
  • 141 Torkildsen CF, Austdal M, Jarmund AH. et al. New immune phenotypes for treatment response in high-grade serous ovarian carcinoma patients. Front Immunol 2024; 15: 1394497
  • 142 Wouters MC, Komdeur FL, Workel HH. et al. Treatment Regimen, Surgical Outcome, and T-cell Differentiation Influence Prognostic Benefit of Tumor-Infiltrating Lymphocytes in High-Grade Serous Ovarian Cancer. Clin Cancer Res 2016; 22: 714-724
  • 143 Bohm S, Montfort A, Pearce OM. et al. Neoadjuvant Chemotherapy Modulates the Immune Microenvironment in Metastases of Tubo-Ovarian High-Grade Serous Carcinoma. Clin Cancer Res 2016; 22: 3025-3036
  • 144 Peng J, Hamanishi J, Matsumura N. et al. Chemotherapy Induces Programmed Cell Death-Ligand 1 Overexpression via the Nuclear Factor-kappaB to Foster an Immunosuppressive Tumor Microenvironment in Ovarian Cancer. Cancer Res 2015; 75: 5034-5045
  • 145 Launonen IM, Erkan EP, Niemiec I. et al. Chemotherapy induces myeloid-driven spatial T-cell exhaustion in ovarian cancer. bioRxiv 2024;
  • 146 Jimenez-Sanchez A, Cybulska P, Mager KL. et al. Unraveling tumor-immune heterogeneity in advanced ovarian cancer uncovers immunogenic effect of chemotherapy. Nat Genet 2020; 52: 582-593
  • 147 Montfort A, Pearce O, Maniati E. et al. A Strong B-cell Response Is Part of the Immune Landscape in Human High-Grade Serous Ovarian Metastases. Clin Cancer Res 2017; 23: 250-262
  • 148 Lodewijk I, Bernardini A, Suarez-Cabrera C. et al. Genomic landscape and immune-related gene expression profiling of epithelial ovarian cancer after neoadjuvant chemotherapy. NPJ Precis Oncol 2022; 6: 7
  • 149 Mesnage SJL, Auguste A, Genestie C. et al. Neoadjuvant chemotherapy (NACT) increases immune infiltration and programmed death-ligand 1 (PD-L1) expression in epithelial ovarian cancer (EOC). Ann Oncol 2017; 28: 651-657
  • 150 Vanguri R, Benhamida J, Young JH. et al. Understanding the impact of chemotherapy on the immune landscape of high-grade serous ovarian cancer. Gynecol Oncol Rep 2022; 39: 100926
  • 151 Monk BJ, Colombo N, Oza AM. et al. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN Ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22: 1275-1289
  • 152 Ray-Coquard IL, Savoye AM, Schiffler C. et al. Neoadjuvant and adjuvant pembrolizumab in advanced high-grade serous carcinoma: the randomized phase II NeoPembrOV clinical trial. Nat Commun 2024; 15: 5931
  • 153 Le Saux O, Ardin M, Berthet J. et al. Immunomic longitudinal profiling of the NeoPembrOv trial identifies drivers of immunoresistance in high-grade ovarian carcinoma. Nat Commun 2024; 15: 5932
  • 154 Ding L, Kim HJ, Wang Q. et al. PARP Inhibition Elicits STING-Dependent Antitumor Immunity in Brca1-Deficient Ovarian Cancer. Cell Rep 2018; 25: 2972-2980.e5
  • 155 Ding L, Wang Q, Martincuks A. et al. STING agonism overcomes STAT3-mediated immunosuppression and adaptive resistance to PARP inhibition in ovarian cancer. J Immunother Cancer 2023; 11: e005627
  • 156 Martincuks A, Song J, Kohut A. et al. PARP Inhibition Activates STAT3 in Both Tumor and Immune Cells Underlying Therapy Resistance and Immunosuppression In Ovarian Cancer. Front Oncol 2021; 11: 724104
  • 157 Wang Z, Sun K, Xiao Y. et al. Niraparib activates interferon signaling and potentiates anti-PD-1 antibody efficacy in tumor models. Sci Rep 2019; 9: 1853
  • 158 Shen J, Zhao W, Ju Z. et al. PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness. Cancer Res 2019; 79: 311-319
  • 159 Konstantinopoulos PA, Waggoner S, Vidal GA. et al. Single-Arm Phases 1 and 2 Trial of Niraparib in Combination With Pembrolizumab in Patients With Recurrent Platinum-Resistant Ovarian Carcinoma. JAMA Oncol 2019; 5: 1141-1149
  • 160 Domchek SM, Postel-Vinay S, Im SA. et al. Olaparib and durvalumab in patients with germline BRCA-mutated metastatic breast cancer (MEDIOLA): an open-label, multicentre, phase 1/2, basket study. Lancet Oncol 2020; 21: 1155-1164
  • 161 Gonzalez Martin A, Sanchez Lorenzo L, Colombo N. et al. A phase III, randomized, double blinded trial of platinum based chemotherapy with or without atezolizumab followed by niraparib maintenance with or without atezolizumab in patients with recurrent ovarian, tubal, or peritoneal cancer and platinum treatment free interval of more than 6 months: ENGOT-Ov41/GEICO 69-O/ANITA Trial. Int J Gynecol Cancer 2021; 31: 617-622
  • 162 Monk BJ, Minion LE, Coleman RL. Anti-angiogenic agents in ovarian cancer: past, present, and future. Ann Oncol 2016; 27 (Suppl. 1) i33-i39
  • 163 Garcia-Martinez E, Redondo A, Piulats JM. et al. Are antiangiogenics a good ‘partner’ for immunotherapy in ovarian cancer?. Angiogenesis 2020; 23: 543-557
  • 164 Bouzin C, Brouet A, De Vriese J. et al. Effects of vascular endothelial growth factor on the lymphocyte-endothelium interactions: identification of caveolin-1 and nitric oxide as control points of endothelial cell anergy. J Immunol 2007; 178: 1505-1511
  • 165 Tavira B, Iscar T, Manso L. et al. Analysis of Tumor Microenvironment Changes after Neoadjuvant Chemotherapy with or without Bevacizumab in Advanced Ovarian Cancer (GEICO-89T/MINOVA Study). Clin Cancer Res 2024; 30: 176-186
  • 166 Spigel DR, Cheng Y, Cho BC. et al. ADRIATIC: Durvalumab (D) as consolidation treatment (tx) for patients (pts) with limited-stage small-cell lung cancer (LS-SCLC). J Clin Oncol 2024; 42 (Suppl. 17)
  • 167 Kurtz JE, Pujade-Lauraine E, Oaknin A. et al. Atezolizumab Combined With Bevacizumab and Platinum-Based Therapy for Platinum-Sensitive Ovarian Cancer: Placebo-Controlled Randomized Phase III ATALANTE/ENGOT-ov29 Trial. J Clin Oncol 2023; 41: 4768-4778
  • 168 Liu YL, Selenica P, Zhou Q. et al. BRCA Mutations, Homologous DNA Repair Deficiency, Tumor Mutational Burden, and Response to Immune Checkpoint Inhibition in Recurrent Ovarian Cancer. JCO Precis Oncol 2020; 4: PO.20.00069
  • 169 Landen CN, Molinero L, Hamidi H. et al. Influence of Genomic Landscape on Cancer Immunotherapy for Newly Diagnosed Ovarian Cancer: Biomarker Analyses from the IMagyn050 Randomized Clinical Trial. Clin Cancer Res 2023; 29: 1698-1707
  • 170 Stover EH, Fuh K, Konstantinopoulos PA. et al. Clinical assays for assessment of homologous recombination DNA repair deficiency. Gynecol Oncol 2020; 159: 887-898
  • 171 Patel JN, Braicu I, Timms KM. et al. Characterisation of homologous recombination deficiency in paired primary and recurrent high-grade serous ovarian cancer. Br J Cancer 2018; 119: 1060-1066
  • 172 Takaya H, Nakai H, Sakai K. et al. Intratumor heterogeneity and homologous recombination deficiency of high-grade serous ovarian cancer are associated with prognosis and molecular subtype and change in treatment course. Gynecol Oncol 2020; 156: 415-422
  • 173 Takaya H, Nakai H, Takamatsu S. et al. Homologous recombination deficiency status-based classification of high-grade serous ovarian carcinoma. Sci Rep 2020; 10: 2757
  • 174 Duraiswamy J, Turrini R, Minasyan A. et al. Myeloid antigen-presenting cell niches sustain antitumor T cells and license PD-1 blockade via CD28 costimulation. Cancer Cell 2021; 39: 1623-1642 e1620
  • 175 Ledermann JA, Shapira-Frommer R, Santin AD. et al. Molecular determinants of clinical outcomes of pembrolizumab in recurrent ovarian cancer: Exploratory analysis of KEYNOTE-100. Gynecol Oncol 2023; 178: 119-129
  • 176 Haas AR, Tanyi JL, O’Hara MH. et al. Phase I Study of Lentiviral-Transduced Chimeric Antigen Receptor-Modified T Cells Recognizing Mesothelin in Advanced Solid Cancers. Mol Ther 2019; 27: 1919-1929
  • 177 Chen J, Hu J, Gu L. et al. Anti-mesothelin CAR-T immunotherapy in patients with ovarian cancer. Cancer Immunol Immunother 2023; 72: 409-425
  • 178 Crawford A, Haber L, Kelly MP. et al. A Mucin 16 bispecific T cell-engaging antibody for the treatment of ovarian cancer. Sci Transl Med 2019; 11: eaau7534
  • 179 Li T, Wang J. Therapeutic effect of dual CAR-T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer 2020; 20: 678
  • 180 Shalaby N, Xia Y, Kelly JJ. et al. 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
  • 181 Tubridy EA, Eiva MA, Liu F. et al. CD137+ tumor infiltrating lymphocytes predicts ovarian cancer survival. Gynecol Oncol 2024; 184: 74-82
  • 182 Tassi E, Bergamini A, Wignall J. et al. Epithelial ovarian cancer is infiltrated by activated effector T cells co-expressing CD39, PD-1, TIM-3, CD137 and interacting with cancer cells and myeloid cells. Front Immunol 2023; 14: 1212444
  • 183 Amaria R, Knisely A, Vining D. et al. Efficacy and safety of autologous tumor-infiltrating lymphocytes in recurrent or refractory ovarian cancer, colorectal cancer, and pancreatic ductal adenocarcinoma. J Immunother Cancer 2024; 12: e006822
  • 184 Emmanuelli A, Salvagno C, Hwang SM. et al. High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1alpha activity in neutrophils. Oncoimmunology 2024; 13: 2411070
  • 185 Kalaitsidou M, Moon OR, Sykorova M. et al. Signaling via a CD28/CD40 chimeric costimulatory antigen receptor (CoStAR), targeting folate receptor alpha, enhances T cell activity and augments tumor reactivity of tumor infiltrating lymphocytes. Front Immunol 2023; 14: 1256491
  • 186 Quixabeira DCA, Jirovec E, Pakola S. et al. Improving the cytotoxic response of tumor-infiltrating lymphocytes towards advanced stage ovarian cancer with an oncolytic adenovirus expressing a human vIL-2 cytokine. Cancer Gene Ther 2023; 30: 1543-1553
  • 187 Capellero S, Erriquez J, Melano C. et al. Preclinical immunotherapy with Cytokine-Induced Killer lymphocytes against epithelial ovarian cancer. Sci Rep 2020; 10: 6478
  • 188 Banville AC, Wouters MCA, Oberg AL. et al. Co-expression patterns of chimeric antigen receptor (CAR)-T cell target antigens in primary and recurrent ovarian cancer. Gynecol Oncol 2021; 160: 520-529
  • 189 Hwang SM, Awasthi D, Jeong J. et al. Transgelin 2 guards T cell lipid metabolism and antitumour function. Nature 2024; 635: 1010-1018

Zoom Image
Fig. 1 Homologous recombination repair pathway of double-strand breaks (for a detailed description see info box). Created with Biorender.com [rerif]
Zoom Image
Fig. 2 Graphical summary of factors determining tumor heterogeneity in HGSC. Spatial and temporal heterogeneity in HGSC is shaped by the tumor-intrinsic capacity to repair DNA damaged sites, mutational and gene expression signatures that are associated with HR repair gene defects, and crosstalk between tumor, stromal and immune cells in the tumor microenvironment with implications for prognosis and therapy outcome. Created with Biorender.com [rerif]