Open Access

Ovarian cancer: Diagnosis and treatment strategies (Review)

  • Authors:
    • Xuejiao Li
    • Zhuocheng Li
    • Huiling Ma
    • Xinwei Li
    • Hongxiao Zhai
    • Xixi Li
    • Xiaofei Cheng
    • Xiaohui Zhao
    • Zhilong Zhao
    • Zhenhua Hao
  • View Affiliations

  • Published online on: July 18, 2024     https://doi.org/10.3892/ol.2024.14574
  • Article Number: 441
  • Copyright: © Li et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is a malignant tumor that seriously endangers health. Early ovarian cancer symptoms are frequently challenging to detect, resulting in a large proportion of patients reaching an advanced stage when diagnosed. Conventional diagnosis relies heavily on serum biomarkers and pathological examination, but their sensitivity and specificity require improvement. Targeted therapy inhibits tumor growth by targeting certain characteristics of tumor cells, such as signaling pathways and gene mutations. However, the effectiveness of targeted therapy varies among individuals due to differences in their unique biological characteristics and requires individualized strategies. Immunotherapy is a promising treatment for ovarian cancer due to its long‑lasting antitumor effect. Nevertheless, issues such as variable efficacy, immune‑associated adverse effects and drug resistance remain to be resolved. The present review discusses the diagnostic strategies, rationale, treatment strategies and prospects of targeted therapy and immunotherapy for ovarian cancer.

Introduction

Ovarian cancer is a major malignancy of the female reproductive system and currently the eighth most frequently diagnosed cancer globally (1). Due to the concealed location of ovarian cancer within the body and the absence of reliable early screening techniques, coupled with the subtle nature of initial symptoms, most patients are typically diagnosed with advanced-stage disease upon presentation. However, developments in liquid biopsy and imaging technology are expected to improve the early diagnostic rate of ovarian cancer and provide greater treatment opportunities. Ovarian cancer frequently exhibits primary or secondary drug resistance, rendering treatment challenging and resulting in 5-year survival rates of only 30–45% (2). Deepening biomedical research has resulted in targeted therapy and immunotherapy gradually becoming new options for the treatment of ovarian cancer. Targeted therapies achieve antitumor effects by specifically interfering with signaling pathways critical to tumor cell growth and proliferation. Currently, targeted ovarian cancer therapy mainly includes anti-angiogenesis therapy and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors. Immunotherapy, notably immune checkpoint inhibitors (ICI) and adoptive T-cell therapy, has demonstrated notable efficacy in the treatment of melanoma (3), non-small cell lung cancer (4) and various other malignancies, offering distinct therapeutic benefits. However, the value of immune therapy for the treatment of ovarian cancer (5) has not yet been realized, as it is still at the research stage. Researchers have recently begun to focus on the issue of drug resistance (6) and integrate targeted therapy and immunotherapy with other treatments to improve the survival and quality of life (QOL) of patients with ovarian cancer. The present review focuses on the most recent research progress in ovarian cancer diagnosis, targeted therapy and immunotherapy to provide clinicians with the latest available research results and treatment directions.

Diagnosis

Pathological classification and molecular characteristics

Ovarian cancer is a heterogeneous disease with varying histological, morphological and molecular characteristics. Understanding these molecular features can be used to guide ovarian cancer diagnosis, prognosis and treatment strategies. Ovarian cancers are categorized according to their origin and histological characteristics, with the predominant type being epithelial ovarian cancer (7), constituting ~90% of diagnosed cases. Epithelial ovarian cancer has subtypes including high-grade serous carcinoma (HGSOC) comprising 70% of cases and low-grade serous carcinoma (LGSOC) comprising 3% of cases, along with endometrioid, clear cell, mucinous and other subtypes. Other less common types of ovarian cancer include those that originate from primitive germ cells, known as germ cell tumors, and ovarian stroma cells, known as sex cord-stromal tumors. Although these common subtypes are derived from epithelial tissue, their clinical presentation, molecular features and prognosis differ markedly. Although HGSOC and LGSOC are both derived from the serous epithelium, HGSOC is more invasive than LGSOC and often accompanied by tumor protein p53 (TP53) gene mutations (8), whereas LGSOC is mainly associated with KRAS and BRAF mutations (9). By contrast, endometrioid ovarian cancer shares molecular features with endometrial cancer, such as PTEN and catenin β1 gene mutations; clear-cell ovarian cancer often resembles renal cell carcinoma and is characterized by AR-rich interaction domain 1A and phosphatidylinositol-4, 5-bisphosphonate 3-kinase catalytic subunit α gene mutations; and mucinous ovarian cancer has mucin-secreting characteristics and is mainly associated with KRAS mutations (10,11). Personalized treatment strategies may be required for different ovarian cancer subtypes according to their specific molecular characteristics.

Serum biomarkers

Currently, the ovarian cancer molecular markers routinely detected in clinical practice are cancer antigen 125 (CA125) (12), carcinoembryonic antigen (CEA) (13), human epididymis protein 4 (HE4) (14), carbohydrate antigen 19-9 (CA19-9) (15), mesothelin (16), α-fetoprotein (17), PARP (18), FRα (19), TP53 (20), HRD (21), BRCA1/2 (22) and AXL (23) (Table I); however, the detection of only one of these markers often provides an inaccurate result (24). CA125 is the most frequently used biomarker in the diagnosis of ovarian cancer (25), while HE4 was introduced as a biomarker more recently. CA125 has been reported to exhibit low sensitivity during the early stages of ovarian cancer, during menstruation or in patients with endometriosis, as these are also associated with elevated CA125 levels in some cases. However, the analysis of HE4 in combination with CA125 can be used to improve diagnostic accuracy (24). A clinical investigation involving 458 participants, in which electrochemiluminescence immunoassays were used to evaluate the serum levels of HE4, CA125, CA19-9 and CEA, demonstrated that the joint assessment of HE4 and CA125 yielded the most optimal outcomes. This combination exhibited a sensitivity of 80.10% and specificity of 69.08%. Notably, increasing the number of tumor markers to three or four did not increase diagnostic accuracy beyond the efficacy achieved by the HE4 and CA125 combination (26).

Table I.

Predictive and prognostic capabilities of common tumor biomarkers and their clinical effectiveness.

Table I.

Predictive and prognostic capabilities of common tumor biomarkers and their clinical effectiveness.

First author, yearBiomarker Predictive/prognostic biomarkerSensitivity and specificityClinical effectiveness(Refs.)
Samborski, 2022CA125PredictiveLow sensitivity and specificityOften used for the detection and efficacy evaluation of patients with epithelial ovarian tumors (serous tumors)(12)
Turner, 2021CEAPredictive and prognosticHigh sensitivity and low specificityNot specific to malignant tumors; mainly used as an auxiliary in diagnosis; valuable in the judgement of prognosis and treatment evaluation(13)
Anastasi, 2023HE4PredictiveLow sensitivity and high specificityFDA approved as a marker for monitoring disease progression and recurrence; important for differentiating pelvic masses, benign and malignant tumors(14)
Ali, 2022CA19-9Predictive-Highly expressed in mucinous ovarian cancer and gastroin-testinal metastatic ovarian cancer(15)
Lv, 2019MesothelinPredictive and prognosticHigh sensitivity and low specificityNovel tumor marker for ovarian cancer; candidate for targeted therapy(16)
Rath, 2022AFPPredictive-Mostly used in liver cancer diagnosis; can also be increased in ovarian immature teratoma and endoembryonic sinus tumor(17)
Wu, 2023PARPPredictive and prognostic-Benefits patients with recurrent platinum-sensitive ovarian cancer, regardless of BRCA mutation(18)
Dilawari, 2023FRαPredictive-FDA-approved for FRα-positive, platinum-resistant epithelial ovarian cancer(19)
Borcoman, 2023TP53Predictive and prognostic-Can be used for tumor diagnosis, treatment evaluation, and prognosis; targeted intervention against TP53 can significantly inhibit tumor growth and improve survival(20)
Magliacane, 2022HRDPredictive-Positive HRD is a clinical predictor of PARPi sensitivity(21)
Grafodatskaya, 2022BRCA1/2Predictive and prognostic-Used to assess prognostic outcomes, develop treatment options and predict response to PARPi treatment(22)
Umemura, 2020AXLPrognostic-Upregulation indicates a poor prognosis and is also a potential therapeutic target(23)

[i] CA125, cancer antigen 12; HE4, human epididymis protein 4; FDA, Food and Drug Administration; CEA, carcinoembryonic antigen; AFP, α-fetoprotein; CA19-9, carbohydrate antigen 19-9; FRα, folate receptor α; TP53, tumor protein 53; HRD, homologous recombination deficiency; PARP, poly (adenosine diphosphate-ribose) polymerase; PARPi, PARP inhibitor.

Nanotechnology has recently demonstrated great potential for the diagnosis of ovarian cancer due to its high accuracy and low invasiveness. However, the cost and technical requirements are high, which might limit its clinical applications (27). In addition, autoantibodies such as anti-TP53 and anti-New York esophageal squamous cell carcinoma 1 (NY-ESO-1) have been found to have diagnostic value, although their sensitivity and specificity require further study. Specifically, it was reported that a combination of tripartite motif containing 21, NY-ESO-1, TP53 and paired box 8 exhibited 67% sensitivity and 94% specificity for HGSOC (28). Other new markers and detection methods are also under investigation, including the CytoSaLPs scoring tool (29), circ-DENN domain containing 4 C circular RNA (30) and lysophosphatidic acid (31). In addition, a blood extracellular vesicle microRNA (miRNA) microarray has been developed, which has been demonstrated to enhance the specificity of early ovarian cancer diagnosis, reduce false positives and exhibit clear clinical utility in distinguishing malignant ovarian tumors (32).

Pathological examination

Histopathological examination is key in the diagnosis of ovarian cancer. The microscopic observation of tissue samples enables pathologists to accurately diagnose cancer and observe its biological characteristics. The precise diagnosis of ovarian cancer, particularly when the ovarian tumor is large, relies on the histopathological findings of the resected specimen. Fine-needle aspiration is a commonly used biopsy technique for ovarian cancer; however, the amount of tissue obtained may be insufficient to enable a definite diagnosis in some cases (33). The currently emerging diagnostic method is liquid biopsy, which is non-invasive, low-trauma and low-risk, and enables continuous sampling (34). Circulating tumor DNA (ctDNA) is an important element of the liquid biopsy. ctDNA has been demonstrated to have an improved diagnostic performance compared with conventional CA125 for early cancer detection (35). For example, TP53 mutant ctDNA is a promising tumor-specific biomarker for tracking the treatment response of HGSOC. Notably, it exhibits greater sensitivity compared with CA125, with a superior ability to monitor disease progression and therapeutic efficacy (36). In addition, a meta-analysis demonstrated that the mutation spectrum of a solid tumor and its ctDNA are consistent, which indicates the potential application of the ctDNA in the diagnosis of ovarian cancer (37). However, this field is currently only in clinical development. Therefore, the available methods for the early diagnosis of ovarian cancer are limited in sensitivity and specificity, and the ability to evaluate the multifactorial processes associated with chemoresistance development is restricted (38). Immunohistochemical testing is another important method used in the diagnosis of ovarian cancer. It can be used to evaluate the proteins expressed by tumor cells, thereby providing information about the tumor origin, degree of differentiation and prognosis (39). A recent cross-sectional study determined that ovarian masses exhibiting a high neutrophil-to-lymphocyte ratio and elevated tumor marker levels, are likely to be malignant; however, substantial clinical data are required to validate the diagnostic accuracy of these indicators (40).

Genetic diagnosis

Gene expression profiling is useful in ovarian cancer, as it can provide deeper insights into the molecular mechanisms underlying this multifaceted condition. Microarray technology (41) or RNA sequencing (4244) can be used analyze the expression levels of thousands of gene simultaneously, and are able to identify the different molecular signatures associated with ovarian cancer subtypes (45). Furthermore, gene expression profiling can be used to detect persistently overexpressed or downregulated genes in ovarian cancer, thereby revealing potential therapeutic targets. Nevertheless, gene expression profiling has limitations, such as high cost and impracticality in small-sample experiments (46). Gene expression profiling can also be used to monitor treatment response and detect resistance mechanisms (47). This information may be used to guide personalized treatment strategies.

Next-generation sequencing (NGS) technology offers the capability to sequence vast amounts of nucleotides quickly and at a cost that is significantly lower than that of traditional methods, such as Sanger sequencing. Despite its widespread use, the scientific community has made no clear recommendations regarding the use of NGS in oncology (48). NGS is a high-throughput sequencing technology that can comprehensively evaluate various types of genomic variations, including point mutations, copy number variations and gene fusions. NGS has successfully identified gene mutations that are suitable for use as therapeutic targets and provided insights into the molecular mechanisms of disease development and progression (49). For example, a study indicated that holliday junction recognition protein (HJURP) accelerates tumor progression and increases resistance to chemotherapy in patients with ovarian cancer, suggesting that HJURP is potentially a novel treatment target. This was supported by in vitro experiments, which suggested that HJURP silencing used in conjunction with cisplatin and AZD1775 offers a potential strategy for the management of ovarian cancer (50). NGS technologies such as whole-exome or whole-genome sequencing may be used to identify prognostic and predictive biomarkers associated with ovarian cancer and promote the development of personalized medicine. In addition, the combined detection of miRNA-205, HE4 and CA125 in exosomes extracted from the plasma of patients with ovarian cancer, using healthy controls as a reference, yielded an area under the curve of 0.951, 100% sensitivity and 86.1% specificity in the early detection of ovarian cancer (51).

Epigenetic factors are important in the occurrence, development and prognosis of ovarian cancer. Epigenetic studies have focused on non-coding changes, such as DNA methylation (52,53), histone modifications (54) and non-coding RNAs (55), that might lead to the silencing of tumor-suppressor genes or the overexpression of oncogenes. For example, in a study that aimed to develop a novel panel of methylation-specific genes for use in a TaqMan-based qPCR assay, promoter methylation of high homeobox A9 (HOXA9) and hypermethylation in cancer 1 (HIC1) was detected ≥80% of ovarian cancer tissues, but no hypermethylation was found in the serum of matched cancer-free women. Thus, the study confirmed the excellent performance of combined HOXA9 and HIC1 methylation analysis in the screening of ovarian cancer samples (56).

Targeted therapy

Targeted therapy for ovarian cancer involves the specific binding of targeted drugs to molecular markers on the surface of tumor cells, effectively blocking their signaling pathways and inhibiting the proliferation and survival of the cells. Various targeted drugs are currently employed in the treatment of ovarian cancer, including PARP (5761), vascular endothelial growth factor (VEGF) (62), tyrosine kinase inhibitors (TKIs) and FRα (63) as listed in Table II. The introduction of these drugs has improved the survival rates and QOL of patients with ovarian cancer, and also expanded the therapeutic options available to physicians. With ongoing advancements in biomarker research, more effective targeted drugs are likely to be developed, further enhancing the treatment landscape for ovarian cancer.

Table II.

Overview of clinical trials and therapeutic targets for ovarian cancer therapies.

Table II.

Overview of clinical trials and therapeutic targets for ovarian cancer therapies.

First author, yearTargeted drugClinical trialTargetDate to market, yyyy/mm/ddCompanyCondition(Refs.)
Washington, 2019 Lynparza/olaparibNCT01844986PARP2018/12/19AstraZenecaMaintenance therapy for advanced ovarian cancer with BRCA1/2 mutation and platinum-sensitive recurrent ovarian cancer after response to chemotherapy(57)
Shirley, 2019 Rubraca/rucaparibNCT02855944PARP2018/06/13Clovis OncologyAdvanced BRCA-mutated ovarian cancer with receipt of ≥2 prior chemotherapy regiments(58)
Essel, 2018 Zejula/niraparibNCT02655016PARP2017/03/27Glaxo Smith KlineUsed in combination with some chemotherapy agents to treat platinum-resistant recurrent ovarian cancer(59)
Lee, 2021 AiRuiYi/fuzuloparibNCT03509636PARP2020/12/11Jiangsu Hengrui Pharmaceuticals Co., Ltd.Treatment after second-line chemotherapy for platinum-sensitive recurrent ovarian cancer with BRCA1/2 mutation, and maintenance therapy after effective chemotherapy for platinum-sensitive recurrent cancer(60)
Luo, 2022 Baihuize/pamiparibNCT03333915PARP2021/05/07BeiGene, Ltd.Recurrent ovarian cancer with germline BRCA1/2 mutation after prior second-line or higher chemotherapy(61)
Guan, 2018 Avastin/bevacizumabNCT00262847 and NCT00483782VEGF2018/06/13Genentech, Inc.Combined with chemotherapeutics, such as paclitaxel and topotecan, to treat platinum-resistant recurrent ovarian cancer(62)
Matulonis, 2023 Elahere/Mirvetuximab soravtansine-gynxNCT04209855FRα2022/11/14ImmunoGenPlatinum-resistant, FRα-positive epithelial ovarian cancer prior to 1–3 line systemic therapy(63)

[i] GOG, Gynecologic Oncology Group; VEGF, vascular endothelial growth factor; FRα, folate receptor α.

PARP inhibitors

PARP inhibitors function via the mechanism of targeted synthetic lethality. PARP inhibitors inhibit the repair of DNA damage in tumor cells and promote apoptosis, thereby enhancing the curative effect of radiotherapy and platinum drug-based chemotherapy (18). In a number of clinical trials, PARP inhibitors significantly prolonged the progression-free survival (PFS) of patients with ovarian cancer in first-line maintenance therapy as well as in platinum-sensitive relapsed maintenance therapy. Therefore, the use of PARP inhibitors as a maintenance therapy has been introduced for ovarian cancer (6466). Additionally, treatments with PARP inhibitors have shown encouraging outcomes in various solid tumors with BRCA1/2 mutations, including HER2-negative breast cancer, metastatic pancreatic cancer and prostate cancer resistant to conventional therapies (67). At present, the US Food and Drug Administration has sanctioned the use of three PARP inhibitors, namely olaparib, niraparib and rucaparib, for the management of ovarian cancer.

Olaparib, developed by AstraZeneca, has been endorsed for initial maintenance therapy in ovarian cancer and as a maintenance treatment for recurrent ovarian cancer that responds to platinum-based chemotherapy. The SOLO2/ENGOT-Ov21 phase III trial used olaparib as platinum-sensitive maintenance therapy in patients with recurrent ovarian cancer (68). The results indicated that olaparib exerted an obvious curative effect on patients with recurrent ovarian cancer and BRCA1/2 mutation compared with placebo, where the patients experienced an increase in median overall survival (OS) of 12.9 months compared with placebo-treated controls. However, in August 2022, based on extensive clinical data, the manufacturer of olaparib voluntarily withdrew the indication of this drug as a single agent for the treatment of relapsed advanced ovarian cancer with BRCA mutation after third-line or more prior chemotherapy. Nakazawa et al (69) reported that patients with platinum-sensitive recurrent ovarian cancer with disease progression after olaparib maintenance therapy had a very poor response to subsequent platinum-based chemotherapy. Nevertheless, the continued use of olaparib following platinum-based chemotherapy remains the preferred approach for individuals newly diagnosed with BRCA-mutated advanced ovarian cancer, as well as for those with platinum-responsive recurrent disease. However, further studies are required to support the use of this drug in posterior-line therapy, and more findings are expected.

Rucaparib is the second PARP inhibitor, after olaparib, to be FDA-approved for the treatment of ovarian cancer. In 2018, the FDA approved rucaparib for use in platinum chemotherapy-sensitive recurrent epithelial ovarian cancer and fallopian tube carcinoma, or as maintenance therapy in primary peritoneal carcinoma. Evaluations of participants in two phase I/II studies and a phase II clinical trial have shown that rucaparib is effective as a third-line therapy for ovarian cancer with BRCA mutations, with an overall response rate (ORR) of 80% in patients who underwent treatment in the phase II trial (70). In addition, the phase III ARIEL3 clinical trial used rucaparib in the maintenance treatment of platinum-sensitive ovarian cancer, and the results revealed that the treatment significantly improved the PFS of the patients (71). These trial results indicate that rucaparib is a promising treatment option that may extend the lifespan of the patient and reduce adverse reactions, such as aminotransferase levels, inflammation and elevated serum creatinine.

Niraparib has received authorization for use as a maintenance treatment in adult individuals with recurrent epithelial ovarian cancer, cancer of the fallopian tube or primary peritoneal cancer that is sensitive to platinum, following initial platinum-based chemotherapy. The NOVA trial demonstrated the efficacy of niraparib maintenance chemotherapy in patients with a partial response to platinum-based therapy, with and without a germline BRCA mutation (gBRCAm), as their PFS was longer than that of the patients treated with placebo (72). In patients achieving a complete response, niraparib was associated with greater PFS improvement relative to placebo in patients with a gBRCAm compared with those without. Niraparib exhibited controllable tolerance during adverse drug reaction monitoring, and dose reduction was possible to limit the possibility of adverse drug reactions (73). However, data on the cost-effectiveness of niraparib as a maintenance therapy are limited, particularly due to the lack of mature OS data. Furthermore, it is unclear whether the advantage of niraparib over placebo in PFS will lead to a confirmed improvement in OS (73,74).

In addition to the use of PARP inhibitors as monotherapy, other ongoing clinical trials (7577) are focusing on their combinations with other agents, including platinum-based chemotherapy, anti-angiogenic agents and ICIs, with specific examples being carboplatin and bevacizumab. Patient-derived organoids (PDOs) have been recognized as an effective tumor model for the screening of PARP inhibitors and for addressing drug resistance in ovarian cancer, with patients experiencing improved results when treated according to recommendations derived from these models (78,79). However, the use of PDOs faces challenges, such as ensuring high-quality biopsies and navigating the intricacies of the tumor microenvironment. Consequently, the identification of PARP inhibitors that more accurately meet patient requirements necessitates further investigation and study.

Generally, PARP inhibitors demonstrate positive efficacy in the first- and second-line maintenance treatment of ovarian cancer, but their use in post-line treatment requires further study. A subgroup analysis demonstrated that patients with BRCA mutations or with homologous recombination deficiency (HRD) benefited more than those without BRCA mutations or HRD, respectively (66). Thus, genetic testing can guide accurate treatment, and follow-up requires further research.

VEGF inhibitors

VEGF is important in new blood vessel formation, tumor growth and metastasis in ovarian cancer, as it stimulates endothelial cell proliferation, division and migration, and increases vascular permeability (80,81). VEGF prevents tumor neovascularization by hindering the interaction between VEGF and its receptor, VEGFR, thereby contributing to its anticancer activity. Currently, the most studied targeted therapy is bevacizumab, a recombinant humanized monoclonal antibody targeting VEGF, which has been shown to prolong median PFS effectively when added to chemotherapy (81). Two randomized phase III trials, namely Gynecologic Oncology Group (GOG)-0218 and ICON7, resulted in the inclusion of upfront bevacizumab in the National Comprehensive Cancer Network (NCCN) ovarian cancer guidelines (82). The first-line approval was based on the results of the pivotal GOG-0218 study, which demonstrated that adding bevacizumab to carboplatin and paclitaxel chemotherapy significantly prolonged the median PFS by 3.8 months and that this combination did not reduce the QOL of the patients (80,83). Following this rationale, the European Medicines Agency sanctioned the use of bevacizumab with platinum and paclitaxel as an initial chemotherapy combination for ovarian cancer (62). The ICON7 trial used a lower bevacizumab dose and shorter maintenance period. This significantly improved the median PFS by 2.7 months, with a greater benefit for patients with high-risk disease. This finding might be explained by the greater demand for angiogenesis in the patients with a high risk of progression, causing them to be more susceptible to antiangiogenic therapies (62,82). Studies have also shown that additional treatment with bevacizumab significantly prolonged PFS, but not OS, in patients regardless of their resistance or sensitivity to platinum-based chemotherapy (81,84). In short, bevacizumab significantly improved the PFS and ORR of patients with platinum-resistant ovarian cancer. However, the inclusion of bevacizumab in chemotherapy regimens should be tailored to the specific circumstances of patients with platinum-sensitive ovarian cancer, with its addition being contingent upon the clinical status of the patient. The use of bevacizumab in maintenance therapy should be based on treatment with this agent during first- or second-line chemotherapy and the HRD and BRCA status of the patient. Although the GOG-0218 and ICON7 clinical trials reported that bevacizumab prolonged the PFS by several months, it did not effectively extend OS. Therefore, bevacizumab does not currently fulfill the necessary criteria for being an optimal treatment option for patients with advanced ovarian cancer.

TKIs

In addition to bevacizumab, certain TKIs also serve as anti-angiogenic drugs targeting VEGF and VEGFR, and may be added to the treatment regimen according to the condition of the patient.

VEGFR signaling pathways are typically inhibited by TKIs that target receptor tyrosine kinases (RTKs). RTKs are typically imbalanced in most cases of HGSOC (85) and frequently excessively activated in tumors, which can lead to recurrence, progression and metastasis. Therefore, RTKs have long been considered a therapeutic target for endometrial ovarian cancer. The AGO-OVAR 12, ICON6 and AGO-OVAR 16 phase III trials used nintedanib, cediranib and pazopanib, respectively, to target TKIs (8688). Numerous studies have reported that these drugs are more effective when used in combination therapies, and exhibit little activity as single drugs (8789). Pazopanib targets VEGFR and platelet-derived growth factor receptor, which regulate tumor cell growth, metabolism and angiogenesis, and demonstrates good activity when used as a single agent. However, the AGO-OVAR 16 study found that although pazopanib prolonged PFS, it did not improve the median OS significantly, and had a reduced effect on prolonging PFS in East Asian patients compared with patients of other ethnicities (88). Based on these results, the US FDA did not approve pazopanib for the treatment of advanced ovarian cancer, and the NCCN guidelines do not recommend pazopanib as a maintenance therapy after initial ovarian cancer treatment. However, this drug can be used for the treatment of platinum-resistant recurrent ovarian cancer.

Cediranib exerts good antitumor effects in recurrent ovarian cancer; the ICON6 study reported that cediranib chemotherapy combined with maintenance therapy extended the PFS by 2.3 months compared with simple chemotherapy and did not reduce the QOL of patients (87). Additionally, the anti-angiogenic properties of anlotinib have been reported to be superior to those of sunitinib, sorafenib and nintedanib (84). Notably, individuals receiving only anlotinib had a PFS period of 7.7 months, demonstrating that the efficacy of anlotinib was comparable to that of bevacizumab. Additionally, anlotinib is associated with a reduced rate of toxic reactions compared with sunitinib, especially in terms of hematological toxicities (90). Therefore, anlotinib appears to be a potential anti-angiogenic agent that is safe to use in ovarian cancer.

Overall, the TKIs nintedanib, cediranib and pazopanib effectively prolong the PFS but not the OS of patients with ovarian cancer, and are not used in routine clinical practice. Therefore, it is urgently necessary for more clinical trials to evaluate the effectiveness of TKIs and discover more effective combination therapies. Furthermore, anlotinib may be a potential treatment option with low toxicity for patients with ovarian cancer that is resistant or refractory to platinum-based therapies. However, few studies of anlotinib have been performed, and its mechanisms of action have not been fully elucidated. Thus, further trials are required to verify its specific efficacy.

Epidermal growth factor inhibitors

The epidermal growth factor receptor (EGFR), a member of the ErbB group of cell surface receptors, is highly expressed in 30–70% of ovarian cancers, a condition that is often associated with a less favorable outcome (85). Current evidence (91,92) suggests that single-agent anti-EGFR biotherapy with EGFR TKIs or anti-EGFR antibodies, either as maintenance therapy after first-line chemotherapy or combined with chemotherapy, has little effect on the survival of patients with recurrent cancer. New-generation EGFR inhibitors interfere with the interaction of multiple ErbB family members. These include lapatinib, an EGFR and ErbR-2 dual inhibitor, which is currently undergoing a GOG phase II study in the US for the treatment of advanced ovarian cancer (93).

Activation of PI3K-AKT-mTOR signaling and other pathogenic pathways

The PI3K-AKT-mTOR signaling pathway is involved in various cellular processes, including regulation of the cell cycle, the production of proteins that precede apoptosis, the regulation of angiogenesis, and the promotion of cancer cell invasion and metastasis. In addition, mutations and hyperactivation of this pathway are observed in the majority of patients with ovarian cancer. Therefore, targeting this signaling pathway is important to inhibit cancer cell proliferation and apoptosis (9496). In addition to its involvement in ovarian cancer, the PI3K-AKT-mTOR pathway prevents oocyte maturation in ovarian follicular dysplasia (94). In an in vitro study, the dual PI3K/mTOR inhibitor PKI-402 significantly reduced cell proliferation and cell viability compared that achieved with single PI3K or mTOR inhibitors. By reducing the activity of this survival signaling pathway, PKI-402 may have the potential to suppress tumor formation, migration and invasion, and overcome chemotherapy and radiotherapy resistance (97). However, although this dual mTOR/PI3K inhibitor yielded good antitumor effects, it nevertheless exerts toxic and adverse effects on normal cells. Therefore, much additional research is necessary to develop improved mTOR/PI3K dual inhibitors and single PI3K/AKT/mTOR inhibitors.

The RAS-RAF-MEK-ERK pathway, a key MAPK pathway, is involved in cell proliferation, differentiation and development, growth factor signaling, and the apoptosis pathway. Clinical studies have explored the use of MAPK inhibitors both as monotherapies and in combination therapies, focusing on the selective inhibition of proteins within the MAPK pathway, known as vertical inhibition, or targeting multiple signaling pathways to reduce their activity (98,99). There is considerable evidence that MEK inhibitors are only potentially curative in patients with LGSOC, while dual inhibition of this MAPK pathway using RAF and MEK inhibitors has greater potential (98,99). Additionally, the simultaneous inhibition of MEK and PARP has been shown to result in increased DNA damage, potentially leading to cell death. Specifically, MEK inhibition augments the effectiveness of the PARP inhibitor, with an extended duration of effect and broadened therapeutic potential (18,100).

The folate receptor (FR)-mediated signaling pathway is involved in tumor division and migration, and the inhibition of FRα has been demonstrated to have direct anticancer activity. Therefore, FRα is a novel therapeutic target in ovarian cancer research. The employment of FRα antibody-drug conjugates (ADCs) and small-molecule drug conjugates, which increase the targeted delivery of anticancer treatments directly to the tumor site, has improved the outlook of patients with cancer exhibiting high levels of FRα expression (101). In November 2022, the US FDA approved mirvetuximab soravtansine, the first-ever FAα target in the ADC field. This ADC has been approved used as a systemic treatment for adult patients with platinum-resistant epithelial ovarian cancer, fallopian tube carcinoma or primary peritoneal carcinoma (102).

A phase II trial of mirvetuximab soravtansine for platinum-resistant ovarian cancer, conducted as a single-arm study, demonstrated uniform antitumor effects, along with favorable tolerability and safety profiles in patients with FRa-high tumors (63). In summary, mirvetuximab soravtansine is a promising treatment for patients who have ovarian cancer with high FRα expression levels, and may stimulate the development of new targeted therapy methods for the further improvement of prognosis and survival.

Immunotherapy

Cancer vaccines

Tumors escape the immune system through various mechanisms. It is generally thought that epithelial ovarian cancer is immunogenic (103). Ovarian cancer immunotherapy using cancer vaccines is a treatment strategy that aims to stimulate the immune system of the patient to recognize and attack cancer cells, thereby killing tumor cells and inhibiting their proliferation. A number of cancer vaccines have been developed based on the delivery of selected tumor-specific antibodies, including tumor antigens, tumor cells, peptides and genetic vaccines. However, various factors influence the generation of an autoimmune response. Dendritic cell (DC) vaccines increase DC uptake and the presentation of tumor-associated antigens and so are a particularly attractive option for immunotherapy (104). Human conventional type 1 DCs have the ability to activate natural killer (NK) and NK T cells (105). Numerous clinical trials have investigated the treatment of ovarian cancer using therapeutic vaccines, and indicated that a single application of a vaccine does not have an obvious effect (106), possibly due to the tumor interfering with the effectiveness of the vaccine. Therefore, it may be necessary to use a vaccine in combination with other therapies (107). Furthermore, recent studies have demonstrated that novel ovarian cancer vaccines, such as a formulation comprising cancer antigen and adjuvants (108), and cancer stem cell vaccines (109), have potential as a direction for further investigation. However, the limitations of these studies are that they were only conducted in mouse models and cannot be directly generalized to humans.

ICIs

Evasion of the immune response presents a significant obstacle in the treatment of ovarian cancer. At present, the ICIs showing the most potential target cytotoxic T-lymphocyte-associated protein 4 (CTLA-4). This protein is expressed on the surface of activated lymphocytes and resembles the protein CD28; however, while CD28 promotes T-cell activation, CTLA-4 suppresses the immune response. Anti-CTLA-4 treatment is associated with some adverse reactions, include fatigue, mild infusion reaction and inflammatory skin reaction (110). However, data on anti-CTLA-4 as a single-agent therapy for ovarian cancer are limited, and anti-CTLA-4 agents are being explored in combination with other ICIs, such as anti-programmed death 1 (PD-1) and anti-PD-1 ligand (PD-L1) agents, in an attempt to improve efficacy (111). Furthermore, the use of anti-CTLA-4 drugs with other treatments such as chemotherapy, targeted therapy or cancer vaccines has potential in the treatment of ovarian cancer (112).

Immune cells, especially activated T lymphocytes, express PD-1 and PD-L1 on the cell surface. As interaction between PD-1 and PD-L1 impedes the activity in the target cell, blocking the signal transduction between the two proteins effectively alters tumor activity. Currently, several commonly used PD-1/PD-L1 inhibitors are undergoing clinical trials in ovarian cancer (113,114). In a clinical trial, patients with uncommon serous tumors resistant to PD-1/PD-L1 inhibitors demonstrated significantly better responses compared to those with other, typically more responsive, ovarian cancer subtypes (115). Another trial observed that poor prognosis was significantly associated with the tumor cell PD-L1 level in ovarian cancer (90). Overall, PD-1/PD-L1 inhibitors have found to be effective in a smaller proportion of patients with ovarian cancer than in patients with other tumors.

ICIs have been combined with other treatments for ovarian cancer (116119). However, the incidence and severity of adverse events are much higher with combined treatment than with single treatments (120,121). There are various clinical situations with unmet clinical demands, particularly for recurrent platinum-resistant ovarian cancer. Furthermore, there is evidence that the gut microbiome influences the efficacy of ICIs, with different bacterial strains varying in their influence on immune regulation (122).

Adoptive cell therapy

Cellular immunotherapy, also known as adoptive cell therapy, is a personalized immunotherapy. It involves the ex vivo expansion and manipulation of lymphocytes from the patient, and their reinfusion into the patient to promote an immune response to cancer. This novel method is an potential cure with multiple strategies (123,124), such as the use of tumor-infiltrating lymphocytes, NK cells or modified immune components, such as chimeric antigen receptors (CARs) and engineered T cell receptors, for adoptive cell transfer. Continuous advances in genomics and immune engineering technology have facilitated the development and use of CAR-T cells; however, they have several challenges, including limited activity in solid tumors, off-target effects, tumor antigen escape, ovarian tumor heterogeneity and immunosuppressed tumor cells (125,126). T-cell receptor therapy is an alternative cell-based therapy that has potential as a future option for the treatment of ovarian cancer. However, it is major histocompatibility complex (MHC)-restricted, as it relies on the presence of MHC molecules to identify antigenic targets and activate T-cell function (127). Based on data from the Cancer Genome Atlas, CD47 is amplified in HGSOC. The amplification of CD47 enhances its ‘don't eat me’ signal, which protects cancer cells from being phagocytized by macrophages. Therefore, targeting bone marrow immune checkpoints might also be an attractive therapy option (128).

Conclusions

Despite remarkable progress in the diagnosis, targeted therapy and immunotherapy of ovarian cancer, early detection and accurate diagnosis are the key to successful treatment. The emergence of novel biomarkers combined with artificial intelligence-based imaging technologies is expected to revolutionize diagnostic capabilities. With the advent of personalized precision medicine, targeted therapy enables a personalized and precise treatment regimen by selectively focusing on the molecular alterations driving ovarian cancer progression. However, it currently does not alter OS, highlighting the need for more effective maintenance therapies. Immunotherapy is a promising means of harnessing the immune system to fight the cancer. Recent advances in ICIs, adoptive T-cell therapy, oncolytic viruses, DCs, cancer vaccines and cytokines have provided promising prospects for the current and future treatment of ovarian cancer. Future strategies may involve the design of therapies that limit the production of inflammatory cytokines while preserving antitumor efficacy. This approach could significantly mitigate the side effects typically associated with conventional treatments.

Despite these major achievements, challenges remain in the optimization of treatment strategies and overcoming drug resistance mechanisms in ovarian cancer. As research continues into the complex tumor microenvironment and tumor-host interactions, a complete understanding of the molecular and immunological underpinnings of ovarian cancer is critical. Furthermore, a single approach is not generally advantageous in the treatment of ovarian cancer, and future research efforts that focus on combining targeted therapy with immunotherapy and selecting the appropriate combination therapy according to the ovarian cancer type are recommended. Given the complex relationship between the response to different combination therapies and treatment effects, additional biomarkers might be necessary for use in response prediction. Furthermore, it is necessary to develop uniform standards for combination drugs in the treatment of ovarian cancer. Despite novel therapies offering new hope, their high costs and uncertain efficacy limit their acceptability and widespread use. Moreover, the recurrence rate of ovarian cancer remains high, and the options for post-recurrence treatment are limited, which presents a significant challenge for current treatment protocols. Therefore, future research focusing on the identification of new therapeutic targets and the development of more effective strategies for post-recurrence treatment is necessary. In terms of future directions, improvements in personalized treatment and precision medicine are likely to be crucial. Tailoring treatment plans that are optimally suited for each patient, based on an analysis of the genetic and molecular profiles of the tumor, may be key to improving the therapeutic outcomes and QOL of patients with ovarian cancer.

With the knowledge and insights presented in the present review, it is expected that the scientific community will progress greatly in the advancement of ovarian cancer diagnosis, treatment and management. This may ultimately herald a new era of precision medicine and improved patient care.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Henan Science and Technology Research Project (grant no. 232102310318) and the Excellent Young Teachers Training Program of Sanquan College of Xinxiang Medical University (grant nos. SQ2021YQJH02 and SQ2022YQJH01).

Availability of data and materials

Not applicable.

Authors' contributions

XJL and ZCL performed the literature search and drafted the manuscript. HLM and XWL were primarily responsible for writing the section on targeted therapy. HXZ, XXL, XFC, XHZ, ZLZ and ZHH were major contributors to revision of the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Freimund AE, Beach JA, Christie EL and Bowtell DDL: Mechanisms of drug resistance in high-grade serous ovarian cancer. Hematol Oncol Clin North Am. 32:983–996. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Borch TH, Andersen R, Ellebaek E, Met Ö, Donia M and Svane IM: Future role for adoptive T-cell therapy in checkpoint inhibitor-resistant metastatic melanoma. J Immunother Cancer. 8:e0006682020. View Article : Google Scholar : PubMed/NCBI

4 

Zhang W, Shi F, Kong Y, Li Y, Sheng C, Wang S and Wang Q: Association of PTPRT mutations with immune checkpoint inhibitors response and outcome in melanoma and non-small cell lung cancer. Cancer Med. 11:676–691. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Wang M, Herbst RS and Boshoff C: Toward personalized treatment approaches for non-small-cell lung cancer. Nat Med. 27:1345–1356. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Soberanis Pina P and Lheureux S: Overcoming PARP inhibitor resistance in ovarian cancer. Int J Gynecol Cancer. 33:364–376. 2023. View Article : Google Scholar : PubMed/NCBI

7 

Cheasley D, Nigam A, Zethoven M, Hunter S, Etemadmoghadam D, Semple T, Allan P, Carey MS, Fernandez ML, Dawson A, et al: Genomic analysis of low-grade serous ovarian carcinoma to identify key drivers and therapeutic vulnerabilities. J Pathol. 253:41–54. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Flaum N, Crosbie EJ, Edmondson RJ, Smith MJ and Evans DG: Epithelial ovarian cancer risk: A review of the current genetic landscape. Clin Genet. 97:54–63. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Garziera M, Cecchin E, Canzonieri V, Sorio R, Giorda G, Scalone S, De Mattia E, Roncato R, Gagno S, Poletto E, et al: Identification of novel somatic TP53 mutations in patients with high-grade serous ovarian cancer (HGSOC) using next-generation sequencing (NGS). Int J Mol Sci. 19:15102018. View Article : Google Scholar : PubMed/NCBI

10 

Bolton KL, Chen D, Corona de la Fuente R, Fu Z, Murali R, Köbel M, Tazi Y, Cunningham JM, Chan ICC, Wiley BJ, et al: Molecular subclasses of clear cell ovarian carcinoma and their impact on disease behavior and outcomes. Clin Cancer Res. 28:4947–4956. 2022. View Article : Google Scholar : PubMed/NCBI

11 

Zhang X, Wei X, Bai G, Huang X, Hu S, Mao H and Liu P: Identification of three potential prognostic genes in platinum-resistant ovarian cancer via integrated bioinformatics analysis. Cancer Manag Res. 13:8629–8646. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Samborski A, Miller MC, Blackman A, MacLaughlan-David S, Jackson A, Lambert-Messerlian G, Rowswell-Turner R and Moore RG: HE4 and CA125 serum biomarker monitoring in women with epithelial ovarian cancer. Tumour Biol. 44:205–213. 2022. View Article : Google Scholar : PubMed/NCBI

13 

Turner MA, Lwin TM, Amirfakhri S, Nishino H, Hoffman RM, Yazaki PJ and Bouvet M: The use of fluorescent anti-CEA antibodies to label, resect and treat cancers: A review. Biomolecules. 11:18192021. View Article : Google Scholar : PubMed/NCBI

14 

Anastasi E, Farina A, Granato T, Colaiacovo F, Pucci B, Tartaglione S and Angeloni A: Recent insight about HE4 role in ovarian cancer oncogenesis. Int J Mol Sci. 24:104792023. View Article : Google Scholar : PubMed/NCBI

15 

Ali FT, Soliman RM, Hassan NS, Ibrahim AM, El-Gizawy MM, Mandoh AAY and Ibrahim EA: Sensitivity and specificity of microRNA-204, CA125, and CA19.9 as biomarkers for diagnosis of ovarian cancer. PLoS One. 17:e02723082022. View Article : Google Scholar : PubMed/NCBI

16 

Lv J and Li P: Mesothelin as a biomarker for targeted therapy. Biomark Res. 7:182019. View Article : Google Scholar : PubMed/NCBI

17 

Rath A, Mitra S, Adhya AK and Majumdar SKD: Immature teratoma with very high AFP levels. Eur J Obstet Gynecol Reprod Biol X. 16:1001702022.PubMed/NCBI

18 

Wu Y, Xu S, Cheng S, Yang J and Wang Y: Clinical application of PARP inhibitors in ovarian cancer: From molecular mechanisms to the current status. J Ovarian Res. 16:62023. View Article : Google Scholar : PubMed/NCBI

19 

Dilawari A, Shah M, Ison G, Gittleman H, Fiero MH, Shah A, Hamed SS, Qiu J, Yu J, Manheng W, et al: FDA approval summary: Mirvetuximab soravtansine-gynx for FRα-positive, platinum-resistant ovarian cancer. Clin Cancer Res. 29:3835–3840. 2023. View Article : Google Scholar : PubMed/NCBI

20 

Borcoman E, Santana Dos Santos E, Genestie C, Pautier P, Lacroix L, Caputo SM, Cabaret O, Guillaud-Bataille M, Michels J, Auguste A, et al: Combined tumor-based BRCA1/2 and TP53 mutation testing in ovarian cancer. Int J Mol Sci. 24:115702023. View Article : Google Scholar : PubMed/NCBI

21 

Magliacane G, Brunetto E, Calzavara S, Bergamini A, Pipitone GB, Marra G, Redegalli M, Grassini G, Rabaiotti E, Taccagni G, et al: Locally Performed HRD testing for ovarian cancer? Yes, we can! Cancers (Basel). 15:422022.PubMed/NCBI

22 

Grafodatskaya D, O'Rielly DD, Bedard K, Butcher DT, Howlett CJ, Lytwyn A, McCready E, Parboosingh J, Spriggs EL, Vaags AK and Stockley TL: Practice guidelines for BRCA1/2 tumour testing in ovarian cancer. J Med Genet. 59:727–736. 2022. View Article : Google Scholar : PubMed/NCBI

23 

Umemura S, Sowa Y, Iizumi Y, Kitawaki J and Sakai T: Synergistic effect of the inhibitors of RAF/MEK and AXL on KRAS-mutated ovarian cancer cells with high AXL expression. Cancer Sci. 111:2052–2061. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Dochez V, Caillon H, Vaucel E, Dimet J, Winer N and Ducarme G: Biomarkers and algorithms for diagnosis of ovarian cancer: CA125, HE4, RMI and ROMA, a review. J Ovarian Res. 12:282019. View Article : Google Scholar : PubMed/NCBI

25 

Zhang M, Cheng S, Jin Y, Zhao Y and Wang Y: Roles of CA125 in diagnosis, prediction, and oncogenesis of ovarian cancer. Biochim Biophys Acta Rev Cancer. 1875:1885032021. View Article : Google Scholar : PubMed/NCBI

26 

Chen F, Shen J, Wang J, Cai P and Huang Y: Clinical analysis of four serum tumor markers in 458 patients with ovarian tumors: Diagnostic value of the combined use of HE4, CA125, CA19-9, and CEA in ovarian tumors. Cancer Manag Res. 10:1313–1318. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Barani M, Bilal M, Sabir F, Rahdar A and Kyzas GZ: Nanotechnology in ovarian cancer: Diagnosis and treatment. Life Sci. 266:1189142021. View Article : Google Scholar : PubMed/NCBI

28 

Hurley LC, Levin NK, Chatterjee M, Coles J, Muszkat S, Howarth Z, Dyson G and Tainsky MA: Evaluation of paraneoplastic antigens reveals TRIM21 autoantibodies as biomarker for early detection of ovarian cancer in combination with autoantibodies to NY-ESO-1 and TP53. Cancer Biomark. 27:407–421. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Lekka S, Psomiadou V, Panoskaltsis T, Tsouma E, Novkovic N, Trihia H, Tzaida O, Korfias D, Giannakas P, Iavazzo C, et al: CytoSaLPs score: A promising new tool for the detection and screening of extrauterine high grade serous carcinoma. BMC Cancer. 23:1572023. View Article : Google Scholar : PubMed/NCBI

30 

Liu S, Yuan L, Li J, Liu Y, Wang H and Ren X: circDENND4C, a novel serum marker for epithelial ovarian cancer, acts as a tumor suppressor by downregulating miR-200b/c. Ann Med. 55:908–919. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Chae CS, Sandoval TA, Hwang SM, Park ES, Giovanelli P, Awasthi D, Salvagno C, Emmanuelli A, Tan C, Chaudhary V, et al: Tumor-derived lysophosphatidic acid blunts protective type I interferon responses in ovarian cancer. Cancer Discov. 12:1904–1921. 2022. View Article : Google Scholar : PubMed/NCBI

32 

Xu M, Zhang D, Xu X, Zhang J and Jiang G: Application of blood extracellular vesicle miRNA in early diagnosis of ovarian cancer. Chinese Patent Application Publication CN117802228A. Filed. September 30–2022.

33 

Bansal A, Srinivasan R, Rohilla M, Sundaram A, Rai B, Rajwanshi A, Suri V, Saha SC, Gupta N, Gupta P and Dey P: Morphologic and immunocytochemical features of high-grade serous carcinoma of ovary in ascitic fluid effusion and fine-needle aspiration cytology. Am J Clin Pathol. 154:103–114. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Asante DB, Calapre L, Ziman M, Meniawy TM and Gray ES: Liquid biopsy in ovarian cancer using circulating tumor DNA and cells: Ready for prime time? Cancer Lett. 468:59–71. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Campos-Carrillo A, Weitzel JN, Sahoo P, Rockne R, Mokhnatkin JV, Murtaza M, Gray SW, Goetz L, Goel A, Schork N and Slavin TP: Circulating tumor DNA as an early cancer detection tool. Pharmacol Ther. 207:1074582020. View Article : Google Scholar : PubMed/NCBI

36 

Kim YM, Lee SW, Lee YJ, Lee HY, Lee JE and Choi EK: Prospective study of the efficacy and utility of TP53 mutations in circulating tumor DNA as a non-invasive biomarker of treatment response monitoring in patients with high-grade serous ovarian carcinoma. J Gynecol Oncol. 30:e322019. View Article : Google Scholar : PubMed/NCBI

37 

Lu Y and Li L: The prognostic value of circulating tumor DNA in ovarian cancer: A meta-analysis. Technol Cancer Res Treat. 20:153303382110437842021. View Article : Google Scholar : PubMed/NCBI

38 

Zhu JW, Charkhchi P and Akbari MR: Potential clinical utility of liquid biopsies in ovarian cancer. Mol Cancer. 21:1142022. View Article : Google Scholar : PubMed/NCBI

39 

Buza N: Immunohistochemistry in gynecologic carcinomas: Practical update with diagnostic and clinical considerations based on the 2020 WHO classification of tumors. Semin Diagn Pathol. 39:58–77. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Hosseini MS, Amiri F, Rezapour M, Ashraf Ganjoie T, Farzaneh F, Arab M, Talayeh M, Beheshti Rooy R and Hadi F: Evaluation of the cutoff point and diagnostic value of the neutrophil-to-lymphocyte ratio in predicting ovarian cancer compared to pathological findings. Asian Pac J Cancer Prev. 25:971–976. 2024. View Article : Google Scholar : PubMed/NCBI

41 

Wang L: Prognostic effect of programmed death-ligand 1 (PD-L1) in ovarian cancer: A systematic review, meta-analysis and bioinformatics study. J Ovarian Res. 12:372019. View Article : Google Scholar : PubMed/NCBI

42 

Olbrecht S, Busschaert P, Qian J, Vanderstichele A, Loverix L, Van Gorp T, Van Nieuwenhuysen E, Han S, Van den Broeck A, Coosemans A, 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. 13:1112021. View Article : Google Scholar : PubMed/NCBI

43 

Liu C, Zhang Y, Li X and Wang D: Ovarian cancer-specific dysregulated genes with prognostic significance: scRNA-Seq with bulk RNA-Seq data and experimental validation. Ann N Y Acad Sci. 1512:154–173. 2022. View Article : Google Scholar : PubMed/NCBI

44 

Nagasawa S, Ikeda K, Horie-Inoue K, Sato S, Takeda S, Hasegawa K and Inoue S: Identification of novel mutations of ovarian cancer-related genes from RNA-sequencing data for Japanese epithelial ovarian cancer patients. Endocr J. 67:219–229. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Shih AJ, Menzin A, Whyte J, Lovecchio J, Liew A, Khalili H, Bhuiya T, Gregersen PK and Lee AT: Identification of grade and origin specific cell populations in serous epithelial ovarian cancer by single cell RNA-seq. PLoS One. 13:e02067852018. View Article : Google Scholar : PubMed/NCBI

46 

Wang J, Dean DC, Hornicek FJ, Shi H and Duan Z: RNA sequencing (RNA-Seq) and its application in ovarian cancer. Gynecol Oncol. 152:194–201. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Huang D, Chowdhury S, Wang H, Savage SR, Ivey RG, Kennedy JJ, Whiteaker JR, Lin C, Hou X, Oberg AL, et al: Multiomic analysis identifies CPT1A as a potential therapeutic target in platinum-refractory, high-grade serous ovarian cancer. Cell Rep Med. 2:1004712021. View Article : Google Scholar : PubMed/NCBI

48 

Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, Normanno N, Scarpa A, Robson M, Meric-Bernstam F, et al: Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: A report from the ESMO precision medicine working group. Ann Oncol. 31:1491–1505. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Huhtaniemi I, Hovatta O, La Marca A, Livera G, Monniaux D, Persani L, Heddar A, Jarzabek K, Laisk-Podar T, Salumets A, et al: Advances in the molecular pathophysiology, genetics, and treatment of primary ovarian insufficiency. Trends Endocrinol Metab. 29:400–419. 2018. View Article : Google Scholar : PubMed/NCBI

50 

Dou Z, Qiu C, Zhang X, Yao S, Zhao C, Wang Z, Chu R, Chen J, Chen Z, Li R, et al: HJURP promotes malignant progression and mediates sensitivity to cisplatin and WEE1-inhibitor in serous ovarian cancer. Int J Biol Sci. 18:1188–1210. 2022. View Article : Google Scholar : PubMed/NCBI

51 

Zhu Z, Chen Z, Wang M, Zhang M, Chen Y, Yang X, Zhou C, Liu Y, Hong L and Zhang L: Detection of plasma exosomal miRNA-205 as a biomarker for early diagnosis and an adjuvant indicator of ovarian cancer staging. J Ovarian Res. 15:272022. View Article : Google Scholar : PubMed/NCBI

52 

Gong G, Lin T and Yuan Y: Integrated analysis of gene expression and DNA methylation profiles in ovarian cancer. J Ovarian Res. 13:302020. View Article : Google Scholar : PubMed/NCBI

53 

Chen D, Wu Y, Tilley RD and Gooding JJ: Rapid and ultrasensitive electrochemical detection of DNA methylation for ovarian cancer diagnosis. Biosens Bioelectron. 206:1141262022. View Article : Google Scholar : PubMed/NCBI

54 

Hooda J, Novak M, Salomon MP, Matsuba C, Ramos RI, MacDuffie E, Song M, Hirsch MS, Lester J, Parkash V, et al: Early loss of histone H2B monoubiquitylation alters chromatin accessibility and activates key immune pathways that facilitate progression of ovarian cancer. Cancer Res. 79:760–772. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Peng B, Li J, Yan Y, Liu Y, Liang Q, Liu W, Thakur A, Zhang K, Xu Z, Wang J and Zhang F: Non-coding RNAs: The recently accentuated molecules in the regulation of cell autophagy for ovarian cancer pathogenesis and therapeutic response. Front Pharmacol. 14:11620452023. View Article : Google Scholar : PubMed/NCBI

56 

Singh A, Gupta S, Badarukhiya JA and Sachan M: Detection of aberrant methylation of HOXA9 and HIC1 through multiplex MethyLight assay in serum DNA for the early detection of epithelial ovarian cancer. Int J Cancer. 147:1740–1752. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Washington CR, Richardson DL and Moore KN: Olaparib in the treatment of ovarian cancer. Future Oncol. 15:3435–3449. 2019. View Article : Google Scholar : PubMed/NCBI

58 

Shirley M: Rucaparib: A review in ovarian cancer. Target Oncol. 14:237–246. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Essel KG and Moore KN: Niraparib for the treatment of ovarian cancer. Expert Rev Anticancer Ther. 18:727–733. 2018. View Article : Google Scholar : PubMed/NCBI

60 

Lee A: Fuzuloparib: First approval. Drugs. 81:1221–1226. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Luo L and Keyomarsi K: PARP inhibitors as single agents and in combination therapy: The most promising treatment strategies in clinical trials for BRCA-mutant ovarian and triple-negative breast cancers. Expert Opin Investig Drugs. 31:607–631. 2022. View Article : Google Scholar : PubMed/NCBI

62 

Guan LY and Lu Y: New developments in molecular targeted therapy of ovarian cancer. Discov Med. 26:219–229. 2018.PubMed/NCBI

63 

Matulonis UA, Lorusso D, Oaknin A, Pignata S, Dean A, Denys H, Colombo N, Van Gorp T, Konner JA, Marin MR, et al: Efficacy and safety of mirvetuximab soravtansine in patients with platinum-resistant ovarian cancer with high folate receptor alpha expression: Results from the SORAYA study. J Clin Oncol. 41:2436–2445. 2023. View Article : Google Scholar : PubMed/NCBI

64 

Smith M and Pothuri B: Appropriate selection of PARP inhibitors in ovarian cancer. Curr Treat Options Oncol. 23:887–903. 2022. View Article : Google Scholar : PubMed/NCBI

65 

Lee CK, Friedlander ML, Tjokrowidjaja A, Ledermann JA, Coleman RL, Mirza MR, Matulonis UA, Pujade-Lauraine E, Bloomfield R, Goble S, et al: Molecular and clinical predictors of improvement in progression-free survival with maintenance PARP inhibitor therapy in women with platinum-sensitive, recurrent ovarian cancer: A meta-analysis. Cancer. 127:2432–2441. 2021. View Article : Google Scholar : PubMed/NCBI

66 

Mirza MR, Coleman RL, González-Martín A, Moore KN, Colombo N, Ray-Coquard I and Pignata S: The forefront of ovarian cancer therapy: update on PARP inhibitors. Ann Oncol. 31:1148–1159. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Lheureux S, Braunstein M and Oza AM: Epithelial ovarian cancer: Evolution of management in the era of precision medicine. CA Cancer J Clin. 69:280–304. 2019. View Article : Google Scholar : PubMed/NCBI

68 

Poveda A, Floquet A, Ledermann JA, Asher R, Penson RT, Oza AM, Korach J, Huzarski T, Pignata S, Friedlander M, et al: Olaparib tablets as maintenance therapy in patients with platinum-sensitive relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): A final analysis of a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 22:620–631. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Nakazawa H, Nagao S, Narita M, Shibutani T, Jimi T, Yano H, Kitai M, Shiozaki T and Yamaguchi S: Effect of prior olaparib maintenance therapy for platinum sensitive recurrent ovarian cancer on response to subsequent platinum-based chemotherapy. J Obstet Gynaecol Res. 48:1248–1254. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Moore DC, Ringley JT and Patel J: Rucaparib: A Poly(ADP-Ribose) polymerase inhibitor for BRCA-mutated relapsed ovarian cancer. J Pharm Pract. 32:219–224. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Ledermann JA, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, Colombo N, Weberpals JI, Clamp AR, Scambia G, et al: Rucaparib for patients with platinum-sensitive, recurrent ovarian carcinoma (ARIEL3): Post-progression outcomes and updated safety results from a randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 21:710–722. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Del Campo JM, Matulonis UA, Malander S, Provencher D, Mahner S, Follana P, Waters J, Berek JS, Woie K, Oza AM, et al: Niraparib maintenance therapy in patients with recurrent ovarian cancer after a partial response to the last platinum-based chemotherapy in the ENGOT-OV16/NOVA trial. J Clin Oncol. 37:2968–2973. 2019. View Article : Google Scholar : PubMed/NCBI

73 

Lee A: Niraparib: A review in first-line maintenance therapy in advanced ovarian cancer. Target Oncol. 16:839–845. 2021. View Article : Google Scholar : PubMed/NCBI

74 

No authors listed. Niraparib for ovarian cancer. Aust Prescr. 44:208–209. 2021. View Article : Google Scholar : PubMed/NCBI

75 

Gupta S, Nag S, Aggarwal S, Rauthan A and Warrier N: Maintenance therapy for recurrent epithelial ovarian cancer: Current therapies and future perspectives-a review. J Ovarian Res. 12:1032019. View Article : Google Scholar : PubMed/NCBI

76 

Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Berger R, Fujiwara K, Vergote I, Colombo N, Mäenpää J, et al: Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 381:2416–2428. 2019. View Article : Google Scholar : PubMed/NCBI

77 

Boussios S, Karathanasi A, Cooke D, Neille C, Sadauskaite A, Moschetta M, Zakynthinakis-Kyriakou N and Pavlidis N: PARP inhibitors in ovarian cancer: The route to ‘Ithaca’. Diagnostics (Basel). 9:552019. View Article : Google Scholar : PubMed/NCBI

78 

Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ Jr, Feltmate CM, Nucci MR, Swisher EM, Nguyen H, et al: Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discov. 8:1404–1421. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Kopper O, de Witte CJ, Lõhmussaar K, Valle-Inclan JE, Hami N, Kester L, Balgobind AV, Korving J, Proost N, Begthel H, et al: An organoid platform for ovarian cancer captures intra- and interpatient heterogeneity. Nat Med. 25:838–849. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R and Chinot OL: Bevacizumab (Avastin®) in cancer treatment: A review of 15 years of clinical experience and future outlook. Cancer Treat Rev. 86:1020172020. View Article : Google Scholar : PubMed/NCBI

81 

Nakai H and Matsumura N: The roles and limitations of bevacizumab in the treatment of ovarian cancer. Int J Clin Oncol. 27:1120–1126. 2022. View Article : Google Scholar : PubMed/NCBI

82 

O'Malley DM: New Therapies for ovarian cancer. J Natl Compr Canc Netw. 17:619–621. 2019.

83 

Tewari KS, Burger RA, Enserro D, Norquist BM, Swisher EM, Brady MF, Bookman MA, Fleming GF, Huang H, Homesley HD, et al: Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol. 37:2317–2328. 2019. View Article : Google Scholar

84 

Cui Q, Hu Y, Ma D and Liu H: A retrospective observational study of anlotinib in patients with platinum-resistant or platinum-refractory epithelial ovarian cancer. Drug Des Devel Ther. 15:339–347. 2021. View Article : Google Scholar

85 

Wei Y, Erfani S, Schweer D, de Gouvea R, Qadir J, Shi J, Cheng K, Wu D, Craven R, Wu Y, et al: Targeting receptor tyrosine kinases in ovarian cancer: Genomic dysregulation, clinical evaluation of inhibitors, and potential for combinatorial therapies. Mol Ther Oncolytics. 28:293–306. 2023. View Article : Google Scholar

86 

Ray-Coquard I, Cibula D, Mirza MR, Reuss A, Ricci C, Colombo N, Koch H, Goffin F, González-Martin A, Ottevanger PB, et al: Final results from GCIG/ENGOT/AGO-OVAR 12, a randomised placebo-controlled phase III trial of nintedanib combined with chemotherapy for newly diagnosed advanced ovarian cancer. Int J Cancer. 146:439–448. 2020. View Article : Google Scholar

87 

Ledermann JA, Embleton-Thirsk AC, Perren TJ, Jayson GC, Rustin GJS, Kaye SB, Hirte H, Oza A, Vaughan M, Friedlander M, et al: Cediranib in addition to chemotherapy for women with relapsed platinum-sensitive ovarian cancer (ICON6): Overall survival results of a phase III randomised trial. ESMO Open. 6:1000432021. View Article : Google Scholar

88 

Vergote I, du Bois A, Floquet A, Rau J, Kim JW, Del Campo JM, Friedlander M, Pignata S, Fujiwara K, Colombo N, et al: Overall survival results of AGO-OVAR16: A phase 3 study of maintenance pazopanib versus placebo in women who have not progressed after first-line chemotherapy for advanced ovarian cancer. Gynecol Oncol. 155:186–191. 2019. View Article : Google Scholar

89 

Kuroki L and Guntupalli SR: Treatment of epithelial ovarian cancer. BMJ. 371:m37732020. View Article : Google Scholar

90 

Zhou AP, Bai Y, Song Y, Luo H, Ren XB, Wang X, Shi B, Fu C, Cheng Y, Liu J, et al: Anlotinib versus sunitinib as first-line treatment for metastatic renal cell carcinoma: A randomized phase II clinical trial. Oncologist. 24:e702–e708. 2019. View Article : Google Scholar

91 

Morrison J, Thoma C, Goodall RJ, Lyons TJ, Gaitskell K, Wiggans AJ and Bryant A: Epidermal growth factor receptor blockers for the treatment of ovarian cancer. Cochrane Database Syst Rev. 10:CD0079272018.

92 

Chilimoniuk Z, Rocka A, Stefaniak M, Tomczyk Ż, Jasielska F, Madras D and Filip A: Molecular methods for increasing the effectiveness of ovarian cancer treatment: A systematic review. Future Oncol. 18:1627–1650. 2022. View Article : Google Scholar

93 

Voigtlaender M, Schneider-Merck T and Trepel M: Lapatinib. Recent Results Cancer Res. 211:19–44. 2018. View Article : Google Scholar

94 

Ediriweera MK, Tennekoon KH and Samarakoon SR: Role of the PI3K/AKT/mTOR signaling pathway in ovarian cancer: Biological and therapeutic significance. Semin Cancer Biol. 59:147–160. 2019. View Article : Google Scholar

95 

Ren M, Zhao H, Gao Y, Chen Q, Zhao X and Yue W: NUF2 promotes tumorigenesis by interacting with HNRNPA2B1 via PI3K/AKT/mTOR pathway in ovarian cancer. J Ovarian Res. 16:172023. View Article : Google Scholar

96 

Siegel RL, Miller KD, Wagle NS and Jemal A: Cancer statistics, 2023. CA Cancer J Clin. 73:17–48. 2023. View Article : Google Scholar

97 

Hu X, Xia M, Wang J, Yu H, Chai J, Zhang Z, Sun Y, Su J and Sun L: Dual PI3K/mTOR inhibitor PKI-402 suppresses the growth of ovarian cancer cells by degradation of Mcl-1 through autophagy. Biomed Pharmacother. 129:1103972020. View Article : Google Scholar

98 

Hendrikse CSE, Theelen PMM, van der Ploeg P, Westgeest HM, Boere IA, Thijs AMJ, Ottevanger PB, van de Stolpe A, Lambrechts S, Bekkers RLM and Piek JMJ: The potential of RAS/RAF/MEK/ERK (MAPK) signaling pathway inhibitors in ovarian cancer: A systematic review and meta-analysis. Gynecol Oncol. 171:83–94. 2023. View Article : Google Scholar

99 

Degirmenci U, Wang M and Hu J: Targeting aberrant RAS/RAF/MEK/ERK signaling for cancer therapy. Cells. 9:1982020. View Article : Google Scholar

100 

Vena F, Jia R, Esfandiari A, Garcia-Gomez JJ, Rodriguez-Justo M, Ma J, Syed S, Crowley L, Elenbaas B, Goodstal S, et al: MEK inhibition leads to BRCA2 downregulation and sensitization to DNA damaging agents in pancreas and ovarian cancer models. Oncotarget. 9:11592–11603. 2018. View Article : Google Scholar

101 

Scaranti M, Cojocaru E, Banerjee S and Banerji U: Exploiting the folate receptor α in oncology. Nat Rev Clin Oncol. 17:349–359. 2020. View Article : Google Scholar

102 

Heo YA: Mirvetuximab soravtansine: First approval. Drugs. 83:265–273. 2023. View Article : Google Scholar

103 

Rodriguez GM, Yakubovich E and Vanderhyden BC: Unveiling the immunogenicity of ovarian tumors as the crucial catalyst for therapeutic success. Cancers (Basel). 15:56942023. View Article : Google Scholar

104 

Guo Q, Yang Q, Li J, Liu G, Nikoulin I and Jia S: Advanced clinical trials of dendritic cell vaccines in ovarian cancer. J Investig Med. 68:1223–1227. 2020. View Article : Google Scholar

105 

Calmeiro J, Carrascal MA, Tavares AR, Ferreira DA, Gomes C, Falcão A, Cruz MT and Neves BM: Dendritic cell vaccines for cancer immunotherapy: The role of human conventional type 1 dendritic cells. Pharmaceutics. 12:1582020. View Article : Google Scholar

106 

Dafni U, Martín-Lluesma S, Balint K, Tsourti Z, Vervita K, Chenal J, Coukos G, Zaman K, Sarivalasis A and Kandalaft LE: Efficacy of cancer vaccines in selected gynaecological breast and ovarian cancers: A 20-year systematic review and meta-analysis. Eur J Cancer. 142:63–82. 2021. View Article : Google Scholar

107 

Manning-Geist BL, Gnjatic S, Aghajanian C, Konner J, Kim SH, Sarasohn D, Soldan K, Tew WP, Sarlis NJ, Zamarin D, et al: Phase I study of a multivalent WT1 peptide vaccine (galinpepimut-S) in combination with nivolumab in patients with WT1-expressing ovarian cancer in second or third remission. Cancers (Basel). 15:14582023. View Article : Google Scholar

108 

Zhao Z, Ortega-Rivera OA, Chung YH, Simms A and Steinmetz NF: A co-formulated vaccine of irradiated cancer cells and cowpea mosaic virus improves ovarian cancer rejection. J Mater Chem B. 11:5429–5441. 2023. View Article : Google Scholar

109 

Xu H, Zhao F, Wu D, Zhang Y, Bao X, Shi F, Cai Y and Dou J: Eliciting effective tumor immunity against ovarian cancer by cancer stem cell vaccination. Biomed Pharmacother. 161:1145472023. View Article : Google Scholar

110 

Liu X, Shi Y, Zhang D, Zhou Q, Liu J, Chen M, Xu Y, Zhao J, Zhong W and Wang M: Risk factors for immune-related adverse events: What have we learned and what lies ahead? Biomark Res. 9:792021. View Article : Google Scholar

111 

Zamarin D, Burger RA, Sill MW, Powell DJ Jr, Lankes HA, Feldman MD, Zivanovic O, Gunderson C, Ko E, Mathews C, et al: Randomized phase II trial of nivolumab versus nivolumab and ipilimumab for recurrent or persistent ovarian cancer: An NRG oncology study. J Clin Oncol. 38:1814–1823. 2020. View Article : Google Scholar

112 

Peyraud F and Italiano A: Combined PARP inhibition and immune checkpoint therapy in solid tumors. Cancers (Basel). 12:15022020. View Article : Google Scholar

113 

Disis ML, Taylor MH, Kelly K, Beck JT, Gordon M, Moore KM, Patel MR, Chaves J, Park H, Mita AC, 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. 5:393–401. 2019. View Article : Google Scholar

114 

Xin Yu J, Hodge JP, Oliva C, Neftelinov ST, Hubbard-Lucey VM and Tang J: Trends in clinical development for PD-1/PD-L1 inhibitors. Nat Rev Drug Discov. 19:163–164. 2020. View Article : Google Scholar

115 

Komura N, Mabuchi S, Shimura K, Yokoi E, Kozasa K, Kuroda H, Takahashi R, Sasano T, Kawano M, Matsumoto Y, et al: The role of myeloid-derived suppressor cells in increasing cancer stem-like cells and promoting PD-L1 expression in epithelial ovarian cancer. Cancer Immunol Immunother. 69:2477–2499. 2020. View Article : Google Scholar

116 

Lee JY, Kim JW, Lim MC, Kim S, Kim HS, Choi CH, Yi JY, Park SY and Kim BG; KGOG investigators, : A phase II study of neoadjuvant chemotherapy plus durvalumab and tremelimumab in advanced-stage ovarian cancer: A Korean gynecologic oncology group study (KGOG 3046), TRU-D. J Gynecol Oncol. 30:e1122019. View Article : Google Scholar

117 

Shen J, Zhao W, Ju Z, Wang L, Peng Y, Labrie M, Yap TA, Mills GB and Peng G: PARPi triggers the STING-dependent immune response and enhances the therapeutic efficacy of immune checkpoint blockade independent of BRCAness. Cancer Res. 79:311–319. 2019. View Article : Google Scholar

118 

Brewer M, Angioli R, Scambia G, Lorusso D, Terranova C, Panici PB, Raspagliesi F, Scollo P, Plotti F, Ferrandina G, et al: Front-line chemo-immunotherapy with carboplatin-paclitaxel using oregovomab indirect immunization in advanced ovarian cancer: A randomized phase II study. Gynecol Oncol. 156:523–529. 2020. View Article : Google Scholar

119 

Liu J, Gaillard S, Hendrickson AW, Moroney J, Yeku O, Diver E, Gunderson C, Arend R, Ratner E, Samnotra V, et al: An open-label phase II study of dostarlimab (TSR-042), bevacizumab (bev), and niraparib combination in patients (pts) with platinum-resistant ovarian cancer (PROC): Cohort A of the OPAL trial. Gynecol Oncol. 162 (Suppl 1):S17–S18. 2021. View Article : Google Scholar

120 

Marin-Acevedo JA, Kimbrough EO and Lou Y: Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol. 14:452021. View Article : Google Scholar

121 

Ochoa de Olza M, Navarro Rodrigo B, Zimmermann S and Coukos G: Turning up the heat on non-immunoreactive tumours: Opportunities for clinical development. Lancet Oncol. 21:e419–e430. 2020. View Article : Google Scholar

122 

Kim E, Ahn H and Park H: A review on the role of gut microbiota in immune checkpoint blockade therapy for cancer. Mamm Genome. 32:223–231. 2021. View Article : Google Scholar

123 

Dafni U, Michielin O, Lluesma SM, Tsourti Z, Polydoropoulou V, Karlis D, Besser MJ, Haanen J, Svane IM, Ohashi PS, et al: Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: A systematic review and meta-analysis. Ann Oncol. 30:1902–1913. 2019. View Article : Google Scholar

124 

Yan W, Hu H and Tang B: Advances Of chimeric antigen receptor T cell therapy in ovarian cancer. Onco Targets Ther. 12:8015–8022. 2019. View Article : Google Scholar

125 

Fucà G, Reppel L, Landoni E, Savoldo B and Dotti G: Enhancing chimeric antigen receptor T-cell efficacy in solid tumors. Clin Cancer Res. 26:2444–2451. 2020. View Article : Google Scholar

126 

Zhang XW, Wu YS, Xu TM and Cui MH: CAR-T cells in the treatment of ovarian cancer: A promising cell therapy. Biomolecules. 13:4652023. View Article : Google Scholar

127 

Wu JWY, Dand S, Doig L, Papenfuss AT, Scott CL, Ho G and Ooi JD: T-cell receptor therapy in the treatment of ovarian cancer: A mini review. Front Immunol. 12:6725022021. View Article : Google Scholar

128 

Le Saux O, Ray-Coquard I and Labidi-Galy SI: Challenges for immunotherapy for the treatment of platinum resistant ovarian cancer. Semin Cancer Biol. 77:127–143. 2021. View Article : Google Scholar

Related Articles

Journal Cover

September-2024
Volume 28 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li X, Li Z, Ma H, Li X, Zhai H, Li X, Cheng X, Zhao X, Zhao Z, Hao Z, Hao Z, et al: Ovarian cancer: Diagnosis and treatment strategies (Review). Oncol Lett 28: 441, 2024.
APA
Li, X., Li, Z., Ma, H., Li, X., Zhai, H., Li, X. ... Hao, Z. (2024). Ovarian cancer: Diagnosis and treatment strategies (Review). Oncology Letters, 28, 441. https://doi.org/10.3892/ol.2024.14574
MLA
Li, X., Li, Z., Ma, H., Li, X., Zhai, H., Li, X., Cheng, X., Zhao, X., Zhao, Z., Hao, Z."Ovarian cancer: Diagnosis and treatment strategies (Review)". Oncology Letters 28.3 (2024): 441.
Chicago
Li, X., Li, Z., Ma, H., Li, X., Zhai, H., Li, X., Cheng, X., Zhao, X., Zhao, Z., Hao, Z."Ovarian cancer: Diagnosis and treatment strategies (Review)". Oncology Letters 28, no. 3 (2024): 441. https://doi.org/10.3892/ol.2024.14574