
Treatment of ovarian cancer: From the past to the new era (Review)
- Authors:
- Published online on: June 3, 2025 https://doi.org/10.3892/ol.2025.15130
- Article Number: 384
-
Copyright: © Alrosan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
Abstract
Introduction
Ovarian cancer is the seventh most common type of cancer in female patients (1–4) and the third most common gynecological cancer, following cervical and uterine cancer (5). Although ovarian cancer ranks eighth as a cause of cancer-associated mortality in female patients globally (3,6), it is the most lethal among gynecological tumors (2,3,6,7). Ovarian cancer has a lifetime risk of 1.3% (6) with ~314,000 new cases and ~207,000 mortalities globally each year, based on data from 2020 (8,9). In the United States, >22,000 new cases of ovarian cancer are diagnosed each year, with 14,000 deaths attributed to the disease (1,10). Ovarian cancer is uncommon in young patients, especially those <30 years old; however, the risk rises with age, with a sharp increase in patients ≥50 years old (2). There are also geographical variations, with the highest prevalence in the United States and northern Europe, and the lowest prevalence in Japan (3). Notably, ethnicity also has an impact on the prevalence of ovarian cancer and the incidence of mortality (1). The highest incidence is observed in Caucasian patients with ~12 cases per 100,000 individuals, and the lowest incidence is observed in African-American patients with ~0.4 cases per 100,000 individuals (3).
Symptoms
The term ‘silent killer’ has been given to ovarian cancer due to its high mortality rate, which is attributed to the inapparent tumor growth, delayed onset of symptoms and inadequate screening, often leading to an advanced-stage diagnosis in the first instance (5). Furthermore, ovarian cancer is generally associated with non-specific symptoms. The symptoms are common and include abdominal bloating, abdominal pain, frequent urination, early satiety and changes in bowel habits. However, because these symptoms are often mild or seem ordinary, patients may not seek medical attention, which can lead to delays in diagnosis (2). Ovarian cancer is uncommon, and a general physician may only encounter a case every 5 years (11). This rarity, combined with the commonality of the symptoms, creates notable challenges for both physicians and patients. For example, physicians frequently misinterpret these symptoms, attributing them to conditions such as irritable bowel syndrome, stress or gastritis. At the same time, patients may not recognize the seriousness of these symptoms, further contributing to delays in diagnosis (12). As such, the overlap of ovarian cancer symptoms with more common conditions, along with a lack of awareness, often results in misdiagnosis and delayed treatment.
Risk factors
Momenimovahed et al (5) have classified the risk factors that are associated with ovarian cancer into three main categories: Predisposing, protective and controversial. The predisposing factors include age (more common in patients aged >65 years), menstrual-associated factors, endometriosis, family history of breast or ovarian cancer, BRCA gene mutations (Table I), Lynch syndrome and lower socioeconomic status.
Age is a key predisposing factor for ovarian cancer, with a higher incidence in women >65 years and a median diagnosis age of 63 years. Early-onset ovarian cancer, occurring between 18 and 30 years, accounts for <5% of cases and is usually diagnosed at a localized stage. By contrast, late-onset ovarian cancer is often identified at advanced stages with metastasis, indicating a more favorable prognosis for early-onset cases (13).
A thorough assessment of family history is key for identifying hereditary risks, primarily in cases associated with hereditary breast and ovarian cancer and Lynch syndrome. This evaluation includes analyzing cancer diagnoses in first- and second-degree relatives, particularly those with ovarian, breast, prostate or pancreatic cancer, as well as early-onset or bilateral malignancy, which may indicate pathogenic variants in BRCA1, BRCA2 or mismatch repair genes. The genes that are highly associated with ovarian cancer are listed in Table I. Understanding familial cancer patterns aids risk stratification, enabling personalized interventions such as enhanced surveillance, prophylactic surgery, targeted therapies [such as poly(ADP-ribose) polymerase (PARP) inhibitors] and genetic counseling (14).
Protective factors include multiparity, advanced maternal age at first childbirth, contraceptive use and lactation, which are associated with a reduced risk of ovarian cancer (5). Factors whose associations with ovarian cancer remain inconsistent or debated, include age at menarche and menopause, pregnancy characteristics, pelvic inflammatory disease, hormone replacement therapy, infertility treatment, dietary and nutritional patterns, obesity, physical activity, and the consumption of alcohol, caffeine and tobacco (5).
Etiology
The development of ovarian cancer remains to be elucidated and determining the exact pathophysiology is hampered by the heterogeneous nature of the disease, which includes a variety of histological types with distinct behaviors and features. Several hypotheses have been proposed to explain the etiology of ovarian cancer, including the incessant ovulation (repeated ovulatory cycles cause damage to the ovarian epithelium, increasing cancer risk), inflammation (highlights the role of chronic pelvic inflammation in stimulating carcinogenesis; androgen/progesterone and gonadotropin hypotheses, propose that hormonal imbalances may stimulate abnormal cellular growth (15). The tubal origin hypothesis suggests that many high-grade serous ovarian cancers may actually originate from the epithelial cells of the fallopian tubes (16) . However, a definitive scientific consensus has yet to be established.
Types of ovarian cancer
Primary ovarian malignancies are classified in three primary forms (epithelial, germ cell, and sex cord). Of all ovarian malignancies, ~95% are epithelial cancers. Epithelial cancers are hypothesized to originate from the single-cell layer covering the ovary (2). According to histology, serous [high-grade serous ovarian carcinoma (HGSOC) or low-grade serous ovarian carcinoma (LGSOC)], clear-cell, endometrioid and mucinous tumors are the four most prevalent forms of ovarian cancer, with HGSOC being the most common, accounting for 70–80% of all epithelial subtypes (2). The unique biology and responses to different treatments allow further stratification of the subtypes (17). The remaining 5% of all ovarian cancers consist of sex and germ cell cancers. Germ cell tumors are rare, accounting for 3% of cases of ovarian cancer, and they are most commonly diagnosed in younger patients, typically between the ages of 10 and 30 years. Sex cord-stromal malignancies are the rarest ovarian neoplasms, accounting for <2% of primary ovarian tumors. They are typically benign and frequently identified early during development (2).
Staging and diagnosis
Ovarian cancer is characterized by a low 5-year survival rate (1). The 5-year survival rate for patients with early-stage ovarian cancer (stages I and II) is 70%, but this drops to <20% in patients with advanced-stage cancer (stages III and IV) (7,16). The International Federation of Gynecology and Obstetrics (FIGO) was the first association to develop a staging system for gynecological cancer. The current FIGO staging system combines ovarian, fallopian tube and peritoneum cancer classifications, and considers findings obtained primarily through surgical exploration (Table II) (18).
![]() | Table II.International Federation of Gynecology and Obstetrics staging classification for cancer of the ovary, fallopian tube and peritoneum (18). |
The understanding of ovarian cancer biology has advanced and is now based on histological characteristics and the molecular phenotype (19). The first step in diagnosis is to obtain a thorough medical history from the patient to determine any family history of ovarian and other types of cancer (18). If ovarian cancer is detected early in the development process, the chances of successful treatment are higher (20). Screening tests must be performed multiple times to ensure early, cost-effective treatment while minimizing unnecessary interventions (21). Due to its role in oncogenesis and metastasis, the cancer antigen-125 (CA-125) test has been widely used for ovarian cancer screening and differentiation from benign conditions, making it a key focus in developing antitumor strategies (18,19).
While >90% of patients with advanced-stage cancer exhibit elevated CA-125 levels in the blood, 50–60% of patients with stage I ovarian cancer exhibit elevated CA-125 levels (6,16). A potential explanation for the low serum concentration of CA-125 in patients with partial epithelial ovarian cancer (EOC) may be the development of circulating immune complexes, which may interfere with detection (19). However, CA-125 testing has limitations. Studies have reported false positives, with ~1% of healthy individuals showing elevated CA-125 levels (>35 U/ml) and 5% of patients with benign conditions also exhibiting increased levels (20,22). Furthermore, CA-125 levels vary due to factors such as differences in ethnicity, pregnancy, the menstrual cycle (particularly the follicular phase), aging and menopause (20,22). This raises questions regarding the application of static cut-off points for CA-125 (20) and makes it an unreliable indicator of ovarian cancer.
Due to its low sensitivity and specificity, research shows that a single CA-125 measurement is insufficient for efficient screening. To improve specificity and early-stage disease detection, an approach with two phases that tracks changes in CA-125 over time and applies transvaginal ultrasound for abnormal increases can be adopted (20). It has previously been demonstrated that the Risk of Ovarian Cancer Algorithm, which combines age and serial CA-125 measurements, can increase the identification of cancer during the early stages (20,23). However, these actions are not effective in decreasing ovarian cancer-associated mortality and are expensive (20). The multivariate index assay, which has been reported to have a lower specificity of 40% and a higher sensitivity of 94% compared with CA-125 assessment alone, incorporates five different markers [CA-125, transferrin, transthyretin (prealbumin), apolipoprotein AI and β-2 microglobulin] to generate a score to assess the likelihood of ovarian cancer in patients with a pelvic mass (19).
In addition to CA-125, >110 other possible protein biomarkers have been assessed separately and in combination. Other potential biomarkers include CA15.3, transthyretin, human epididymal protein 4 (HE4) and CA72.4 (6,16). HE4 is a 124-amino acid glycosylated protein that is elevated in the serum of 60–75% of patients with ovarian cancer and can identify a small percentage of cases that are not detected based on CA-125. Other markers and techniques have been shown to improve ovarian cancer detection, such as the presence of autoantibodies, circulating tumor DNA (ctDNA), microRNAs (miRs/miRNAs), DNA methylation, fallopian tube cytology and tumor DNA detection in cervical screening tests (6,16). Furthermore, it has been demonstrated that membrane-spanning mucin (MUC16) glycoprotein CA-125 is a highly glycosylated protein with a molecular weight of ~5 MDa (16). The ovarian cancer cell surface is the site of cleavage of the extracellular domain of MUC16, which releases CA-125 into the pericellular space and the blood, where it can be detected using an immunoassay (16). Extensive research has demonstrated the association between ovarian cancer and the MUC16 biomarker (17,18).
Furthermore, novel imaging techniques have been assessed for the detection of ovarian cancer, including magnetic resonance imaging (MRI) and relaxometry, superconducting quantum interference device technology (SQUID), microbubbles and light-induced endogenous fluorescence (autofluorescence) (6,16). Although it is more expensive and less widely available, MRI combined with relaxometry improves tissue characterization and offers high-resolution, radiation-free imaging. The use of SQUID is constrained by its technical complexity and requirement for cryogenic equipment, despite the fact that it provides ultra-sensitive detection of magnetic signals from cancer-related biomarkers (24). Although it might be limited in deep pelvic regions, microbubble-enhanced ultrasonography increases visualization of tumor vasculature and aids in early identification in a real-time and cost-effective manner. Autofluorescence imaging uses variations in natural tissue fluorescence to detect cancers. It is quick and non-invasive, but its specificity is low (25).
Treatment of ovarian cancer
Comorbidities, previous therapy and the specific biology of the disease serve a role in guiding treatment decisions. As 90% of cases of early-stage ovarian cancer are curable, this highlights the importance of early detection and prompt specialist treatment. However, most patients are diagnosed at a later stage, when the effectiveness of targeted agents, such as chemotherapy and surgery, is limited (26). Since choices made during the surgical and medical phases of the disease may impact the prognosis, controlling the cancer and decreasing the symptoms are the primary goals of treatment (27). Furthermore, although the current standard of care for the treatment of ovarian cancer is primary debulking surgery followed by systemic chemotherapy (28,29), the age at presentation, performance status (PS) and stage at presentation are prognostic factors that influence the therapeutic recommendations (30).
Surgery
Surgical procedures serve a key role in the management of ovarian cancer, functioning as both a diagnostic and therapeutic approach (31). The clinical stage of the disease, histology, specific biology and clinical characteristics of the patient determine the extent of the surgery. Surgery is primarily responsible for cytoreduction and cancer staging. Cytoreductive surgery, which includes primary, interval, second-look and secondary cytoreductive surgery, can be performed at various stages during the course of the treatment (26). The goal of surgeries is to eradicate all macroscopic illnesses or leave no residual illnesses, a state known as R0 (27).
There is a clear association between improved survival results for women with ovarian cancer and complete cytoreduction (R0), which is the removal of all visible tumor burden. After surgery, individuals who have no residual disease have a longer overall survival (OS) and progression-free survival (PFS) than individuals who still have tumor tissue (32). According to Wright et al (33), optimal cytoreduction was associated with a median OS time of up to 60 months in cases of advanced ovarian cancer; however, sub-optimal debulking considerably lowered the median OS time to <30 months. These findings underscore the critical importance of achieving complete cytoreduction to enhance patient prognosis.
Beyond its direct therapeutic benefits, surgical intervention also enables the collection of tissue samples for histopathological evaluation and molecular profiling, which are crucial for guiding personalized treatment approaches. For example, BRCA-mutated patients may benefit from PARP inhibitors as part of their treatment regimen (34,35). Furthermore, the combination of surgery with systemic therapies, including chemotherapy and targeted therapies, maximizes treatment efficacy and improves patient outcomes. Given its pivotal role in disease management, surgical intervention represents a necessary component of comprehensive ovarian cancer treatment (36,37).
Of patients with ovarian cancer, ~35% are diagnosed at early stages (FIGO stage I–II) (38,39). The typical course of treatment entails extensive surgical staging to diagnose the condition and determine the extent of the illness (38). In the initial stages, the surgery needs to be staged (or stratified) and the following protocols should be performed: Peritoneal lavage, total hysterectomy with bilateral salpingo-oophorectomy, biopsy of any suspicious areas; resection of any adhesions adjacent to the tumor, infracolic omentectomy and random biopsy of the uterine fundus, bladder peritoneum, right and left pelvic walls, ovarian fossae, right and left colic canals and both hemidiaphragms. Additionally, pelvic lymphadenectomy, along with sampling or dissection of para-aortic and paracaval lymph nodes, should be performed. Adjuvant chemotherapy should be used if necessary, and the surgery should yield sufficient information for prognostication and staging (27).
In advanced stages, the objective of EOC treatment is to eradicate all visible macroscopic disease, as this has been associated with a higher OS and longer time without disease (27,40). Cytoreductive surgery and platinum-based chemotherapy are the most commonly used treatments for advanced ovarian cancer. Since primary cytoreductive surgery is the gold standard for patients with advanced ovarian cancer and as this surgical outcome is associated with improved OS, the aim of the procedure is to stage the tumor and cytoreduce its volume to the point where there is no gross residual disease >1 cm of tumor (27,41,42). An alternative to primary cytoreductive surgery for patients deemed unsuitable due to insufficient physical fitness or incapacity to resect disease to <1 cm is interval debulking surgery combined with platinum-based neoadjuvant chemotherapy (41).
Chemotherapy
Chemotherapy is a mainstay in the management of ovarian cancer, showing notable effectiveness in different stages and clinical settings (43). Platinum-based agents and taxanes are combined in a standard first-line regimen, resulting in marked response rates, and extending OS and PFS, especially in advanced stages of the disease (37,44). Chemotherapy is important in cases of recurrence; recurrence of platinum-sensitive tumors typically responds to treatment with platinum-based regimens, while non-platinum agents are used in platinum-resistant cancer to mitigate disease progression (45). Furthermore, the integration of chemotherapy with novel therapeutic strategies, such as PARP inhibitors and anti-angiogenic agents, has enhanced its efficacy, particularly in patients with specific genetic profiles such as BRCA mutations (46,47). Clinical trials have demonstrated that these combinations not only improve response rates but also prolong PFS and, in some cases, OS. For instance, the addition of bevacizumab to chemotherapy was associated with a median PFS time improvement of several months in advanced ovarian cancer (48), while PARP inhibitors as maintenance therapy post-chemotherapy reduced the risk of recurrence by up to 70% in BRCA-mutated patients (46). These advancements underscore the evolving role of chemotherapy as a foundational treatment that synergizes with targeted therapies to optimize outcomes for patients with ovarian cancer.
Platinum-based compounds are considered to be the most effective chemotherapeutic drugs in ovarian cancer (28,49). Platinum chemotherapy compounds have been used since the mid-1970s. Cisplatin was the first platinum-based drug, but it had several undesirable side effects, such as nephrotoxicity, neurotoxicity, ototoxicity, gastrointestinal tract problems and allergic reactions. Consequently, the development of second-generation platinum led to the 1989 launch of carboplatin, which is equally as effective as cisplatin but has fewer severe side effects, especially regarding nephrotoxicity (50). The guidance on the use of platinum-based chemotherapy for relapsed EOC has changed over time, becoming a limited and occasionally variable time-based approach. If a relapse occurs >6 months after the conclusion of the previous platinum-based treatment, the patient is deemed ‘platinum-sensitive’ and eligible for further platinum-based chemotherapy. If the gap is <6 months, the patients are considered ‘platinum-resistant’ and not suitable for platinum-based treatment (44). In the latter case, non-platinum regimens are typically offered. Single-agent non-platinum-based chemotherapy, such as weekly administration of paclitaxel, pegylated liposomal doxorubicin or topotecan, is typically offered to patients who are not eligible for additional platinum-based chemotherapy (44,49). Oral etoposide, tamoxifen, gemcitabine and treosulfan are also potential non-platinum alternatives; however, there is a limited probability of these medications being effective because of their decreased cytotoxic potency and the development of resistance mechanisms, including enhanced DNA repair and increased drug efflux, these agents show limited efficacy (51). Moreover, their lack of molecular specificity and weakened activity against aggressive tumor subtypes result in lower response rates compared with platinum-based chemotherapy (52).
Adjuvant chemotherapy using carboplatin (area under the curve, 5–6) and paclitaxel (175 mg/m2) are administered following cytoreductive surgery in accordance with established protocols (53). Typically, 6–8 cycles are given every 21 days (30). While there is some disagreement regarding the optimal number of chemotherapy cycles, to the best of our knowledge, there is no evidence that >6 cycles of postoperative combination chemotherapy improve outcomes for patients with advanced ovarian cancer (28,54). It is advised to start chemotherapy as soon as possible following surgery, usually within 2–4 weeks; longer wait times are associated with worse results (30). Notably, compared with single-agent platinum-based regimens, a combination of platinum-based drugs (containing paclitaxel, gemcitabine or pegylated liposomal doxorubicin) is associated with longer PFS and OS (51). The toxicity profile should be taken into consideration when choosing therapeutic agent combinations (55).
Despite high initial response rates (~70%) with chemotherapy and surgery, recurrence is a notable concern (56). In the 10 years following diagnosis, 80–85% of patients with advanced ovarian cancer experience a relapse (55). The need for additional therapy and their performance status should be considered before beginning treatment for recurrent disease. The next stage is to determine whether platinum is the best option; severe side effects, which include fatigue, arthralgia and neurotoxicity, of chemotherapeutic treatments for ovarian cancer impede the quality of life (55). Thus, investigation of the disease mechanisms requires an understanding of the biology of heterogeneous ovarian cancers. Intrinsic signaling pathways, angiogenesis, hormone receptors and immunological factors are among the possible therapeutic targets being investigated for the treatment of ovarian cancer (56). When chemotherapy is combined with targeted treatments such as bevacizumab and PARP inhibitors, patients with homologous recombination (HR) deficiency (HRD) or BRCA mutations (BRCAms) exhibit improved results once compared with chemotherapy alone (57).
Resistance
Certain patients experience relapses following chemotherapeutic treatments as a result of developing drug resistance mechanisms (58). In ovarian cancer, the initial treatment with carboplatin and paclitaxel as first-line chemotherapy has shown an enhanced complete response rate compared with single agents or platinum based regimens (50). However, recurrence rates are 70–80%, particularly for patients with advanced-stage cancer (50). Notably, patients who receive neoadjuvant carboplatin therapy before surgery are more likely to exhibit platinum resistance. Matsuo et al demonstrated a markedly elevated incidence of carboplatin resistance among patients who receive neoadjuvant therapy (33.3%) compared with those undergoing primary cytoreductive surgery (9.2%) (59). Similarly, Rauh-Hain et al identified a substantially higher prevalence of carboplatin resistance in patients who underwent neoadjuvant therapy (88.8%) compared with those subjected to primary cytoreductive surgery (55.3%) (60).
The molecular diversity of tumor cells contributes to variations in signaling pathways, involving the activation of oncogenes, inactivation of tumor suppressors and the presence of pro-survival genetic mutations. Consequently, resistance to standard chemotherapy regimens is a hurdle in managing the disease (61) and treating patients effectively (56). Thus, understanding of resistance mechanisms is important for the development of novel therapeutic approaches. There are two primary types of resistance: Intrinsic and acquired (extrinsic) resistance. Nevertheless, accurate discrimination between these forms is difficult (34). Intrinsic resistance pertains to the inherent capability of cancer cells to withstand treatment owing to pre-existing characteristics present before their initial exposure to therapeutic agents. Cell attributes associated with intrinsic chemo-resistance include the capacity to decrease drug uptake, increase drug efflux and elevate the activity of detoxification enzymes such as cytochrome P450 or glutathione (GSH) transferases. Conversely, acquired chemoresistance can emerge from genetic and epigenetic alterations that enable cancer cells to adapt to the effects induced by chemotherapy, such as stress, DNA damage and apoptosis (22).
In ovarian cancer, a subset of patients possess germline mutations in BRCA1 and/or BRCA2, which are key constituents of the HR pathway, essential for repairing DNA double-strand breaks. Consequently, BRCAms impair the capacity to rectify DNA damage via HR, potentially accounting for the heightened sensitivity of this cancer subtype to platinum-based chemotherapeutic agents (62). Conversely, the p53 protein, which serves a key role in governing the cell cycle, is sensitive to DNA damage incurred during replication, resulting in G1 arrest and/or apoptosis, thereby inhibiting the generation of defective cells (63). Mutation of the gene responsible for p53 expression in human cancer can result in the loss of p53 function. This enables uncontrolled cell proliferation and confers resistance to agents inducing DNA damage. Consequently, a potential avenue for addressing chemotherapy resistance involves reactivating mutant p53 (64).
Ovarian cancer resistance to chemotherapy is markedly influenced by abnormal transmembrane transport, which includes decreased drug influx and increased drug efflux, leading to decreased intracellular drug concentrations and treatment failure. Platinum-resistant ovarian cancer exhibits diminished drug transport-associated gene and transmembrane transporter expression, resulting in inadequate intracellular platinum accumulation (51,65). miRNAs serve a key role in regulating these transporters by binding to the 3′-untranslated region (UTR) of target genes, thereby modulating their transcription and contributing to drug resistance (66). The solute carrier (SLC) superfamily transporters, such as SLC31A1 and SLC22A1/2/3, are responsible for drug influx. The transport of cisplatin, carboplatin and oxaliplatin by SLC31A1 aids intracellular platinum accumulation (67). Patients with ovarian cancer and low SLC22A2 expression are more likely to develop resistance to platinum drugs as they are unable to absorb as much of the drug (68). While evidence has demonstrated that dysregulated expression of miRNAs and target genes serves a critical role in the initiation, proliferation, survival and chemoresistance of ovarian cancer, the understanding of how miRNAs contribute to the disease pathology remains limited (69,70). Studies focusing on how miRNAs regulate the pathology of ovarian cancer account for <4% of the total published research, highlighting the need for further investigation (69).
Key efflux transporters include ATP-binding cassette subfamily B member 1 (ABCB1), G member 2 and C. miRNAs, such as miR-27a, miR-451, and miR-298, directly bind to the 3′-UTRs) of ABC transporter mRNAs, thereby inhibiting their translation or promoting mRNA degradation by influencing the expression of genes that encode nuclear receptors, transcription factors and signaling molecules associated with ABC transporters. ABCB1 is the only efflux transporter reported to exhibit elevated expression in resistant ovarian cancer cells, while the expression of other ABC transporters is markedly decreased (71). P-glycoprotein, an ATP-dependent drug efflux pump, is encoded by ABCB1 and upregulated in resistant ovarian cancer cell lines, making it a key factor in resistance to paclitaxel, doxorubicin, sorafenib and PARP inhibitors (72,73). miRNAs, including miR-130a/b, miR-186 and miR-495, bind to the 3′-UTR of ABCB1 to degrade the mRNA or limit translation. Although the exact regulatory mechanism is unknown, upregulated ABCB1 expression decreases miR-21-5p expression (68). In patients with HGSOC who undergo chemotherapy or targeted treatment, a whole-genome study identified that an increase in ABCB1 expression is associated with the transcriptional fusion of ABCB1 and SLC25A40 (74).
Beyond transport mechanisms, drug resistance also results from drug inactivation by metallothionein (MT) and GSH (Table III). These thiol-containing proteins bind platinum-based drugs, rendering them inactive, allowing for drug resistance beyond transport mechanisms. Short hairpin RNA targeting MT reverses the well-established resistance mechanism by decreasing MT binding to cisplatin (68,75). The GSH S-platinum complex formed by GSH and cisplatin lowers intracellular platinum levels (76). This platinum inactivation mechanism is catalyzed by GSH S-transferase and associated with platinum resistance in ovarian cancer (68,77).
Novel treatment modalities
Targeted therapy and immunotherapy
Targeted therapy for ovarian cancer utilizes treatments that specifically target the pathways essential for the progression of the disease. By targeting specific proteins, these treatments minimize the adverse effects of cytotoxic treatment on healthy cells. Patients with recurrent disease are typically the first to be assessed for targeted therapy (78). If these treatments demonstrate potential in clinical studies focusing on recurrent diseases, they may be considered a primary treatment option for further investigation. In previous years, targeted therapies, such as PARP inhibitors, antiangiogenic medications and MAPK inhibitors, have been acknowledged as notable advancements in treating ovarian cancer (79,80).
Targeted therapies have revolutionized the treatment of ovarian cancer by focusing on specific biological pathways that drive tumor development and resistance. Bevacizumab and PARP inhibitors are two of the most commonly used targeted treatments (81). Bevacizumab, a VEGF inhibitor, prevents the formation of new blood vessels, decreasing blood supply and slowing tumor growth (82). Bevacizumab can be administered alongside chemotherapy during initial treatment or as maintenance therapy, either alone or in combination with olaparib, a PARP inhibitor. PARP inhibitors block PARP protein, which serves a key role in DNA repair in cancer cells (46,47). Initially developed for patients with mutations in BRCA1 or BRCA2, the use of PARP inhibitors has expanded to patients with other types of DNA repair deficiencies (such as RAD51C (RAD51 Paralog C)/RAD51D (RAD51 Paralog D), ATR (Ataxia Telangiectasia and Rad3-related protein), CHEK1 (Checkpoint Kinase 1)/CHEK2 (Checkpoint Kinase 2), BARD1 (BRCA1 Associated RING Domain 1), BRIP1 (BRCA1 Interacting Protein C-terminal Helicase 1), ATM (Ataxia Telangiectasia Mutated) and PALB2 (Partner and Localizer of BRCA2)) (83). PARP inhibitors approved for use as ovarian cancer treatment are primarily used as maintenance therapy following chemotherapy for advanced ovarian cancer, decreasing the risk of recurrence and tumor progression (84). Previous research has evaluated their effectiveness in broader patient populations, including those with inherited mutations in DNA repair genes, such as partner and localizer of BRCA2 (PALB2), BRCA1-interacting protein C-terminal helicase 1 (BRIP1), RAD51 recombinase paralog C (RAD51C) and RAD51 recombinase paralog D (RAD51D) (85). Additionally, patients without inherited mutations, but with acquired tumor biomarker mutations in DNA repair genes may also benefit from PARP inhibitors (86). There is growing interest in combining PARP inhibitors with immunotherapy or other targeted agents to enhance treatment outcomes (87,88).
Other targeted therapies have been developed for ovarian cancer, particularly for recurrent disease or cases where chemotherapy is ineffective. Mirvetuximab soravtansine (MIRV)-gynx is approved for recurrent ovarian cancer positive for folate receptor α (FRα) (89). Larotrectinib is used to treat metastatic ovarian cancer or cases that cannot be surgically removed and have progressed despite prior treatment, especially if the tumor harbors a neurotrophic receptor tyrosine kinase gene fusion (90). Selpercatinib is prescribed for ovarian cancer with a RET gene fusion, as identified by tumor biomarker testing (91). The integration of these therapies emphasizes the importance of biomarker testing, which can identify patients most likely to respond to precision medicine approaches. Ongoing research continues to explore novel targeted therapies and combination treatment strategies aimed at improving survival and clinical outcomes for patients with ovarian cancer (92–94).
PARP inhibitors
In 2014, the United States Food and Drug Administration (FDA) and European Medication Agency authorized the use of PARP inhibitors for the treatment of ovarian cancer (95). These drugs target EOC that cannot repair DNA through HR, which is key for fixing double-stranded DNA breaks (80,95). Mutations in BRCA1/2 induce HR repair (HRR) pathway deficiencies in tumor cells, which prevent DNA double-stranded breaks from being repaired. PARP inhibitors prevent DNA damage repair in these cells, inducing apoptosis by synthetic lethality (96). Synthetic lethality describes a phenomenon where the presence of a mutated gene, such as one involved in DNA repair, combined with the functional loss or inhibition of another gene or its product, leads to a synergistic effect that induces cellular toxicity and cell death (97).
The FDA has approved olaparib as a maintenance treatment for patients with advanced ovarian cancer with a BRCAm who respond well to initial platinum-based chemotherapy (98). Notably, olaparib increased PFS compared with a placebo following a median follow-up of ~41 months. Furthermore, after 7 years of monitoring, a notable improvement in OS was noted in the SOLO1 (trial no. NCT01844986) clinical study (99).
A previous study compared the efficacy of maintenance niraparib with a placebo in patients with advanced ovarian cancer (100,101). In the PRIMA study (trial no. NCT02655016), 733 patients with newly diagnosed advanced ovarian cancer were randomly assigned to receive either maintenance niraparib or a placebo for up to 36 months, or until disease progression. After 3.5 years of follow-up, the improvement in progression-free survival (PFS) with niraparib was significant, confirming PFS as the primary and durable outcome of the trial (101,102). As a first line of maintenance treatment, 384 patients with advanced ovarian cancer were randomized to receive niraparib [individualized starting dose (ISD)] or a placebo in the PRIME trial (NCT03709316). Following a median observation period of 27.5 months, there was a marked increase in PFS with the niraparib (ISD) regimen compared with the placebo (100).
Similarly, the ATHENA-MONO trial (trial no. NCT03522246) compared maintenance rucaparib with a placebo (103). Rucaparib maintenance therapy markedly increased the median PFS compared with the placebo in the HRD-positive patient group after a median follow-up of ~26 months (103,104). Initial research demonstrated that administration of rucaparib resulted in improved PFS outcomes in patients with BRCAm, non-BRCAm/loss of heterozygosity-high cancer and malignancies that test negative for HRD compared with a placebo (103). Preservation therapy with PARP inhibitors is effective when administered initially to patients with BRCAm or tumors exhibiting HRD (103).
While the PARP inhibitor veliparib is in the advanced stages of clinical testing, the FDA has approved four PARP inhibitors to date: Olaparib, rucaparib, niraparib and talazoparib (105). Talazoparib may exert its therapeutic effect by blocking PARP enzyme activity, which leads to PARP1/2 being trapped on damaged DNA (thereby inhibiting DNA repair) (106). The clinical efficacy of olaparib, an oral PARP inhibitor, or cediranib, an oral VEGF inhibitor, in conjunction with durvalumab, has been evaluated in a phase I dose-escalation trial (107). After determining that chemotherapy with avelumab exhibits antitumor activity and acceptable safety, the JAVELIN OVARIAN PARP100 trial (trial no. NCT03642132) proceeded with maintenance treatment combining the two drugs (108). The selective PARP inhibitor saruparib markedly inhibited tumor growth in preclinical models of breast, ovarian, pancreatic and prostate cancer with HRD mutations (109,110). Notably, saruparib exhibited lower toxicity compared with other PARP inhibitors, allowing administration at higher doses (111).
Chemotherapeutic drugs, such as carboplatin and paclitaxel, along with angiogenesis inhibitors, such as bevacizumab, have demonstrated synergistic effects with PARP inhibitors (35,46). Clinical trials evaluate these combinations to optimize treatment strategies (112–114). Although PARP inhibitors have markedly improved the treatment of ovarian cancer, particularly in patients with BRCAm and HRD, their use is hindered. A major challenge is drug resistance (Table III). Adverse effects, including hematological toxicity (anemia, neutropenia and thrombocytopenia) and gastrointestinal problems (nausea, vomiting and diarrhea), further limit treatment adherence (115,116). Furthermore, the high cost of PARP inhibitors restricts access, particularly in low-resource settings, making affordability a concern (117). Another serious risk is the potential development of secondary malignancy during and after treatment with PARP inhibitors (118). To address these challenges, research is exploring combination therapy, novel biomarkers for patient selection and strategies to overcome resistance and enhance PARP inhibitor efficacy (119–121).
Angiogenesis inhibitors
Tumor growth and angiogenesis are facilitated by the production of VEGF (122). Drugs that hinder angiogenesis and the formation of blood vessels are an alternative approach for managing ovarian cancer. As the number of cancerous cells increases in a tumor, the cells experience hypoxia, leading to angiogenesis, which is the formation of new blood vessels (123). VEGF and its receptor enhance endothelial cell proliferation and movement (124). Angiogenesis is involved in initiating and advancing ovarian cancer (123). Bevacizumab is a monoclonal antibody that targets VEGF-A (124). Phase II trials show improvement in PFS in recurrent ovarian cancer treated with bevacizumab, either alone or in combination with other agents (47,125) Subsequent phase III studies have investigated bevacizumab in both initial treatment and recurrent cases of ovarian cancer, regardless of platinum sensitivity (44,126). Improvements in PFS have been noted in two clinical trials, ICON7 (trial no. NCT00483782) (126,127) and GOG218 (trial no. NCT00262847) (126), when using bevacizumab in the primary treatment and maintenance therapy especially for patients at high risk for progression. The outcomes of bevacizumab are moderate and increased the likelihood of intestinal perforation (128). Therefore, bevacizumab is not commonly used as the initial therapeutic option. Furthermore, bevacizumab has been applied in clinical trials, such as OCEANS and AURELIA (trial no. NCT00976911), to extend PFS in platinum-sensitive and platinum-resistant cases of recurrent ovarian cancer when combined with chemotherapy. This suggests that patients with notable ascites or high angiogenic activity are the most likely to benefit from this strategy (44). Notably, increased EGFR expression is associated with a negative prognosis in ovarian cancer (129). Administering carboplatin alongside cetuximab enhanced results in 9 of 26 patients with ovarian or primary peritoneal cancer who were EGFR-positive (130). However, a second round of patient selection was not performed due to the poor response rate.
Although angiogenesis inhibitors have demonstrated promising efficacy in ovarian cancer treatment, patients exhibit adverse effects such as hypertension, proteinuria and gastrointestinal perforation, necessitating careful patient selection (131,132). Therapeutic strategies that combine angiogenesis inhibitors with PARP inhibitors or immunotherapy have been explored to improve clinical outcomes and overcome resistance mechanisms (96).
MAPK inhibitors and miRNAs
MAPK inhibitors are promising therapeutic options for LGSOC (133). LGSOC is frequently driven by mutations in the MAPK signaling pathway, including in BRAF and KRAS. These genetic alterations result in constitutive activity, promoting tumor growth and resistance to conventional chemotherapy (134,135). MAPK pathway inhibitors, particularly MEK inhibitors such as trametinib and binimetinib, have notable efficacy in increasing PFS in patients with LGSOC, as demonstrated in clinical trials such as GOG-281 and MILO (trial no. NCT02101788) (134). In patients with a BRAF V600E mutation or non-mutated ovarian cancer with limited therapeutic options, it is advisable to target MAPK pathways, such as p38 and JNK pathways (136), as this approach is more effective and manageable regarding toxicity. A previous study reported that the upregulation of p38 and JNK was directly associated with the development of resistance to olaparib in ovarian cancer (137). Combining p38 and JNK inhibitors induced notable antitumor effects in both laboratory studies and animal experiments (138), emphasizing the therapeutic potential of MAPK inhibitors in addressing olaparib-resistant human ovarian cancer (139). While MAPK inhibitors show notable potential in ovarian cancer treatment, challenges persist, including the emergence of resistance through pathway reactivation or alternative signaling mechanisms, as well as adverse effects such as rash, diarrhea and fatigue (140). Additional research on combination therapies and biomarker-driven approaches is required to improve their effectiveness and therapeutic value in multiple subtypes of ovarian cancer.
Several miRNAs are dysregulated in numerous types of cancer, and this dysregulated expression is associated with resistance to chemotherapy (141). miR-139-5p serves a crucial role in ovarian cancer, and its expression levels are decreased in ovarian cancer tissues from cisplatin-resistant patients (142). As such, upregulation of miR-139-5p can impede proliferation, decrease resistance to cisplatin and enhance apoptosis in ovarian cancer cells. Furthermore, the combined use of miR-139-5p and MAPK inhibitors decreases cisplatin resistance in ovarian cancer (143). Thus, upregulating miR-139-5p expression may be a promising treatment approach for ovarian cancer (144).
Immunotherapy
Immunotherapy, which is designed to encourage the immune system to identify and eradicate cancerous cells, has become a promising therapeutic option for ovarian cancer; immunotherapeutic strategies, such as immune checkpoint inhibitors (ICIs) and customized vaccinations, improve outcomes in certain patients with ovarian cancer (145).
Dendritic cells are specialized antigen-presenting cells that are essential for initiating and guiding the development of numerous subsets of CD4+ T cells. They activate immune cells to fight against invading pathogens or cancer cells, and the presence of tumors hinders their function (146). TGF-b is a protein released by tumor cells that impedes the ability of cytotoxic CD8 T lymphocytes to eradicate cancer cells (147). Programmed death ligand 1 (PD-L1) is an immunosuppressive ligand, which is produced by tumor cells. PD-L1 induces immunological tolerance by inhibiting T cells through binding to their receptor, programmed cell death protein 1 (PD-1). Additionally, PD-L1 inhibits IL-2 release by interacting with PD-1, inducing T-cell immunity. PD-L1 expression on monocytes in blood samples of patients with ovarian cancer is associated with a poor prognosis (148,149). Antigen-presenting cells have another immunological checkpoint known as cytotoxic T lymphocyte-associated protein 4 (CTLA-4). CTLA-4 binds to CD80, a co-stimulatory factor, and prevents T-cell activation, resulting in cell cycle arrest (150).
Anti-PD-1 and anti-CTLA-4 ICI therapies were first authorized for use in 2011 (such as ipilimumab) to treat malignant melanoma and non-small and renal cell carcinoma. Official authorization for other anti-PD1 therapies, such as nivolumab use was obtained in 2014 (151). In a phase I trial (trial no. NCT00729664), 17 patients with ovarian cancer were treated with a PD-L1 blocking antibody (BMS-936559); 1 patient exhibited a partial response, while disease stability was reported in 2 patients (152). A total of 10% of patients with platinum-resistant ovarian cancer exhibited a sustained complete response in a phase II trial (trial no. UMIN000005714) utilizing nivolumab (anti-PD-1) (153). In the KEYNOTE-028 multicohort phase Ib trial (trial no. NCT02054806), patients with platinum-resistant ovarian cancer treated with pembrolizumab achieved an objective response rate of 11.5%, while 23% of patients experienced stable disease (154). Ipilimumab, a CTLA-4 blocker, was given as a monotherapy in a phase II trial including patients with platinum-sensitive ovarian cancer (trial no. NCT01611558) (155); 95% of the patients did not survive the induction phase due to disease progression, medication toxicity, mortality or undiscovered factors.
Due to the limited efficacy of immunotherapies that target the PD-1/PD-L1 pathway in ovarian cancer, there is interest in combination treatments that target additional immune checkpoints, such as the T cell immunoreceptor with Ig and ITIM domains (TIGIT)/CD155/DNAX accessory molecule-1 (DNAM-1) pathway. This dual blockade approach may boost T cell and natural killer (NK) cell activity against cancer cells, improve tumor antigen expression and overcome immunosuppression in the tumor microenvironment (156,157). However, more research is required to understand the mechanisms and synergistic benefits of targeting both the PD-1/PD-L1 and TIGIT/CD155/DNAM-1 pathways in ovarian cancer (156). Although immunotherapy has potential in the treatment of ovarian cancer, individual outcomes can vary, and certain patients may not experience notable improvements. Immunotherapy may also result in immune-associated side effects, such as autoimmune responses and inflammation (158).
Combination therapy
Researchers have investigated various combination treatment approaches to enhance clinical outcomes (26,159). Integrating multiple treatment modalities effectively treats ovarian cancer by enhancing the efficacy of each approach (160). These approaches utilize the increasing accessibility of therapeutic drugs and comprehension of the disease biology. The combinations target multiple cancer pathways simultaneously by utilizing DNA-damaging medications, targeted therapy impacting signaling pathways and immunotherapies. Immunotherapy is effective in patients with hypercalcemic small cell carcinoma with high PD-L1 expression and severe ovarian cancer due to their active immune environment (161). A review of 15 clinical trials, involving 945 patients with advanced ovarian cancer, found that PD-1/PD-L1 inhibitors achieve an overall response rate (ORR) of 19%. These inhibitors were significantly more effective when combined with chemotherapy (36% ORR) compared to when used alone (9% ORR) (162). Additionally, patients with platinum-sensitive ovarian cancer responded better to these inhibitors (31% ORR) than those with platinum-resistant disease (19%) ORR (162). Checkpoint-blocking medications have not been successful in treating ovarian cancer despite their efficacy in solid tumors such as melanoma, lung cancer and renal cell carcinoma (163).
It has been demonstrated that elevated intracellular enzyme indoleamine 2,3 dioxygenase (IDO) levels suppress the immunological response (164). Due to its toxic nature, IDO converts tryptophan into kynurenine, enhancing regulatory T cell levels and reducing NK cell levels (165). This results in a weakened immune response, allowing cancer cells to evade immune detection and continue to proliferate. In a phase I trial (trial no. NCT01191216), 41% of patients with various metastatic solid tumors achieved disease stability. By comparison, 18% had a partial response when given a combination of docetaxel and the IDO inhibitor indoximod (146,166). Phase II research on using an IDO1 inhibitor + tamoxifen to treat recurrent EOC and primary peritoneal and fallopian tube carcinoma was discontinued because there was no notable difference in responses between the treatment and control groups (167).
Combining ICIs with cytotoxic medications is a rational approach to enhance tumor immunogenicity and improve the effectiveness of ICIs. For example, a phase II trial investigated the efficacy of combining nivolumab with bevacizumab in recurrent ovarian cancer (168). The combination demonstrated clinical effectiveness, with an overall response rate (ORR) of 28.9% and a PFS of 8.1 months. Atezolizumab is being evaluated in various cancer types in ongoing phase III trials that combine it with chemotherapy and/or bevacizumab. OS and investigator-assessed PFS are co-primary outcomes in the IMagyn050 study (NCT03038100) (169,170). Evaluation of atezolizumab effectiveness in combination with platinum-based chemotherapy with concurrent and maintenance bevacizumab is the primary goal of the ATALANTE study (NCT02891824) (170).
Additionally, ICIs have been studied in combination with PARP inhibitors, as reported in the TOPACIO/KEYNOTE-162 trial (trial no. NCT02657889). Niraparib + pembrolizumab achieved a 25% ORR and 68% disease control rate (DCR) in patients with platinum-resistant recurrent ovarian cancer, with patients with BRCAm showing higher responses (ORR, 45%; DCR, 73%). In platinum-sensitive recurrent ovarian cancer, adding atezolizumab to carboplatin and niraparib maintenance does not improve PFS, regardless of BRCA status or PD-L1 expression (171). The DUO-O trial (NCT03737643) demonstrated that triplet therapy (Durvalumab with chemotherapy and Bevacizumab, followed by maintenance Durvalumab, Bevacizumab and Olaparib) extended the median PFS by 5 months overall and 14.3 months in HRD-positive patients compared with bevacizumab alone, offering notable benefits for non-BRCAm ovarian cancer (172). Furthermore, ICIs have been studied with antibody-drug conjugates in the FORWARD II trial (NCT02606305). MIRV combined with pembrolizumab had promising efficacy in 14 patients with FRα-platinum-resistant recurrent ovarian cancer, achieving a 43% ORR, a median duration of response of 6.9 months and a median PFS of 5.2 months, with no severe adverse events (145).
The FDA has approved olaparib in combination with bevacizumab as a maintenance treatment for patients with advanced ovarian cancer who show improvement after receiving first-line platinum-based chemotherapy and whose tumors are positive for HRD (173). The primary endpoint of investigator-assessed PFS was markedly longer with olaparib + bevacizumab compared with the placebo + bevacizumab after a median follow-up of 22.9 months. The combination of olaparib and bevacizumab resulted in a markedly improved PFS for patients with BRCAm tumors and tumors positive for HRD compared with bevacizumab alone. The combined group receiving olaparib and bevacizumab had a median OS of 56.5 months, while the placebo + bevacizumab group had a median OS of 51.6 months, according to a randomized controlled trial (174). Patients whose tumors were positive for HRD or for BRCAm had the longest median PFS and the highest rates of PFS at 18 months (175,176). Furthermore, OVARIO (phase II), has examined the use of niraparib with bevacizumab as a first-line maintenance regimen for patients with recently diagnosed advanced ovarian cancer demonstrated promising progression-free survival (PFS) outcomes. The safety profile was consistent with the established adverse effect patterns of niraparib and bevacizumab when administered as monotherapies. In the triplet combination of niraparib, dostarlimab and bevacizumab, the OPAL-A trial (NCT03574779) observed limited ORR in patients with platinum-resistant ovarian cancer, although the median PFS of 7.9 months and OS of 22.1 months were favorable compared with historical data. Notably, the majority of responders (85.7%) were bevacizumab-naïve. Exploratory biomarker analysis from paired pre- and post-treatment samples indicated immune activation, warranting further investigation into whether these biomarkers predict the clinical efficacy of triplet therapy (177).
Combining therapeutic modalities can improve the efficacy of ovarian cancer treatment, but it also makes treatment more complicated and raises the possibility of adverse effects, such as immunological, hematological and gastrointestinal toxicity (178).
Nanoparticles
The domain of cancer nanomedicine is experiencing notable growth, with a range of nanoparticle systems investigated through various targeting strategies, suggesting potential for reshaping cancer therapeutics (179,180). Nanomedicine may confer notable advantages over traditional chemotherapeutic agents (181). Nanotechnology-based therapeutics are associated with improved efficacy, decreased toxicity experienced by healthy tissues and improved patient adherence (182). Furthermore, the encapsulation of drugs within nanocarriers offers control over pharmacokinetic properties, including drug release kinetics, prolonged circulation half-life and interaction with healthy tissues (58,180). As such, numerous materials, including carbon- and metal-based nanomaterials, liposomal formulations, cubosomes, lipid and polymeric nanoparticles, micelles (179,182), as well as viral and cell membrane-coated nanoconstructs (179), have been investigated.
Notably, investigations have examined the application of nanomaterials in the encapsulation and concurrent delivery of not only pharmaceutical agents but also imaging agents and genetic material, as well as in the recognition of neoplastic cells through receptor-specific binding mechanisms (179,183). Additionally, these nanostructures yield synergistic effects by amalgamating imaging techniques, such as Ultrasound, Computed Tomography (CT), Magnetic Resonance Imaging (MRI), Positron Emission Tomography (PET), fluorescence Imaging, and photoacoustic Imaging, with one or multiple therapeutic approaches, such as chemotherapy, photodynamic and photothermal therapy, radiotherapy, and immunotherapy (179).
Two principal tumor targeting strategies, passive and active targeting, have been investigated (180,182). The aberrant vascular architecture resulting from rapid tumor vascularization, coupled with inadequate lymphatic drainage, facilitates the enhanced permeability and retention (EPR) effect, which is key for the enrichment of proliferating malignant tumors (180–182). Nevertheless, passive targeting is associated with non-discriminatory accumulation in both healthy and diseased tissue, akin to conventional chemotherapeutic regimens (181). Additionally, the EPR effect presents challenges, as macromolecules or nanoparticles must evade reticuloendothelial system clearance and renal filtration to infiltrate tumor tissue (184). Furthermore, to exploit the EPR effect, a drug must remain in circulation for ≥6 h to accumulate in neoplastic tissues (180). By contrast, active targeting strategies leverage the strong binding affinity of targeting ligands or agents to tumor cell surfaces, facilitating receptor-mediated endocytosis (180,181). Targeted nanocarriers offer advantages over non-targeted counterparts by enhancing efficacy at the delivery site while mitigating potential adverse effects (181). Several active targeting ligands, including folate receptors, monoclonal antibodies, nucleic acids and polypeptides, have been employed to modify nanocarriers, thereby promoting cell uptake (185).
Patients with ovarian cancer often initially respond favorably to conventional therapeutic approaches, but develop resistance over time (182,186). An avenue to enhance the effectiveness and specificity of chemotherapeutic agents involves nanotechnology-based formulations, encompassing encapsulated, conjugated or entrapped/loaded forms within nanocarriers or drug delivery vectors (58,182). The integration of nanotechnology in ovarian cancer management has garnered increasing attention due to its promising attributes in molecular imaging, tumor targeting and drug delivery (187,188). Furthermore, the use of nanotechnology in ovarian cancer extends beyond the delivery of therapeutic agents, including the incorporation of imaging and diagnostic materials (189), rendering such systems ‘theranostic’ nanotechnology (190,191). Various types of nanoparticles have been employed in ovarian cancer therapeutics to facilitate the delivery of drugs, including liposomes, nanoparticles, micelles, dendrimers and polymers (187,189). Notably, certain formulations loaded with chemotherapeutic agents have gained approval from the FDA for ovarian cancer treatment (58). Examples of approved nanoparticles include Doxil®, Genexol-PM® (192) and Abraxane® (190).
Several clinical studies have incorporated drugs into nanoparticles for the treatment of ovarian cancer. Although the majority of studies focused on applying nanoparticles with chemotherapeutic agents, primarily paclitaxel (trial nos. NCT03304210, NCT00499252, NCT00825201, NCT00666991 and NCT00989131), docetaxel (trial no. NCT03742713) and doxorubicin (trial no. NCT01489371), other clinical studies demonstrated the effectiveness of applying nanoparticles with other treatment options such as PARP inhibitors [olaparib (trial no. NCT04669002)] or angiogenesis inhibitors [bevacizumab (trial no. NCT01652079)]. Furthermore, the application of nanoparticles in ovarian cancer extends to studies of the application of combination therapy with nanoparticles such as irinotecan with bevacizumab (trial no. NCT04753216), sargramostim with paclitaxel (trial no. NCT00466960) (193), apatinib and paclitaxel (trial no. NCT03942068), and lapatinib and paclitaxel (trial no. NCT00313599) (186).
Although nanoparticles have potential, there are drawbacks, including toxicity, issues with biocompatibility and immunological reactions that can cause accumulation in organs (194). Concerns regarding scalability, quality control and environmental effects are associated with the complicated and expensive production process (195). Numerous types of treatment based on nanoparticles are in the experimental stage and have had minimal clinical success despite continuous research (196,197).
Genetic testing and gene therapy
HRR pathway germline or somatic mutations are responsible for 20–30% of ovarian cancer cases (198). BRCA1/2, RAD51C, RAD51D, BRIP1, PALB2 and BRCA1 associated RING domain 1 are key proteins in the HRR pathway. According to recommendations by National Comprehensive Cancer Network (NCCN) (9,199,200), American Society of Clinical Oncology and Society of Gynecologic Oncology, genetic testing is recommended for all newly diagnosed cases of EOC (200). Furthermore, BRCA1/2 mutation testing is crucial because it can inform the potential efficacy of PARP inhibitors (9,200). Despite the clinical advantages of genetic testing in ovarian cancer treatment, such as detection of hereditary cancer syndromes, guiding treatment decisions, facilitating risk assessment, early intervention and preventive measures for patients and their family members, it remains underused due to insufficient awareness among clinicians and patients, financial and insurance-related barriers, and limited availability of genetic counseling resources (9). Genetic testing for hereditary ovarian cancer includes whole-exome/genome sequencing, multigene panels or single-gene tests, with next-generation sequencing enabling high-throughput analysis (201). Variant interpretation follows American College of Medical Genetics and Genomics guidelines, with in silico tools, databases such as ClinVar and tumor testing used to differentiate germline from somatic mutations (202). A key part of the genetic testing process is genetic counseling, particularly for individuals with hereditary cancer syndrome. Professional genetic counselors or adequately qualified oncologists should provide counseling, in accordance with regional regulatory requirements (9).
The process of correcting a mutated gene to treat an underlying disorder is known as gene therapy (203). Several gene therapy approaches have been investigated in preclinical studies focused on the management of ovarian cancer (96,204): These strategies include the replacement of tumor suppressor genes to reestablish cellular regulation (such as TP53), oncogene inhibition (such as EGFR), suicide gene therapy involving the introduction of toxin-encoding genes (such as herpes simplex virus thymidine kinase), genetic immunopotentiation to enhance the immune response against tumor cells (such as IL-12A/B), antiangiogenic gene therapy (such as collagen type XVIII α1 chain), methods to restore pharmacological sensitivity (such as survivin) and cancer virotherapy (such as vesicular stomatitis virus). Notably, several of these approaches have been tested in clinical trials; although they showed promising results, most of them are in phase I (96,204).
Hormone receptor modulators
It is well established that estrogen stimulates the proliferation of ovarian cancer cells (205–207). Estrogen signaling, which is mediated by estrogen receptor (ER)α and ERβ and their various isoforms, is further amplified by G protein-coupled ER 1 (208). Both in vitro and in vivo studies have demonstrated that estrogen, through its interaction with ERα, influences cell motility and survival by promoting ovarian cancer cell proliferation and migration and triggering epithelial-mesenchymal transition (205,209,210). Several clinical studies have demonstrated that EOCs, which express ERα, respond well to hormonal therapy such as tamoxifen and aromatase inhibitors (such as letrozole) (211–213). When binding to ER, tamoxifen competes with estrogen, but aromatase inhibitors work by preventing the synthesis of estrogen (96).
Patients with platinum-resistant and recurrent ovarian cancer may consider hormonal therapy as an alternative treatment option according to the 2019 guidelines from the European Society for Medical Oncology-European Society of Gynecological Oncology (214) and the 2021 guidelines from NCCN (version 2.2021) (215). To the best of our knowledge, the clinical effectiveness of hormonal therapy in the treatment of ovarian cancer has not been systematically evaluated in large-scale clinical trials, in spite of these recommendations. Notable trials have assessed fulvestrant, an ER degrader (trial no. NCT00617188), tamoxifen (trial nos. NCT02728622 and NCT00041080), arzoxifene, an ER modulator (trial no. NCT00003670) and mifepristone, a progesterone receptor modulator (trial nos. NCT00459290 and NCT02046421) (96).
Hyperthermic intraperitoneal chemotherapy (HIPEC)
For patients undergoing surgery, adjuvant treatment options such as intraperitoneal and HIPEC therapy should be considered (216). Following cytoreductive surgery, HIPEC is the administration of chemotherapeutic agents directly into the peritoneal cavity to improve patient outcomes through more efficient removal of residual disease. The sensitivity of the tumor to treatment is increased by the hyperthermic environment, which also improves chemotherapeutic drug penetration at the peritoneal surface (96). Notably, when several ovarian cancer treatments, such as carboplatin, paclitaxel and docetaxel, are administered intraperitoneally compared with intravenously, their effective drug concentration in the abdominal cavity is increased several fold and their clearance from the peritoneal cavity is notably slower (34). Although intraperitoneal therapy has been demonstrated to markedly improve the OS (216), other studies have found that paclitaxel and cisplatin intraperitoneal therapy do not extend OS or PFS in patients with stage III ovarian cancer (217,218).
HIPEC is effective in treating gastric and colorectal cancer and primary peritoneal carcinomatosis (92). Uncertainties exist regarding patient selection, drug delivery protocol, treatment timing, chemotherapeutic regimen and possibility of complications (96). The current NCCN guidelines state that patients with ovarian cancer with peritoneal carcinomatosis (FIGO stage III) who show improvement or stable disease following neoadjuvant chemotherapy should be considered for HIPEC (96). Additionally, intraperitoneal injection application has extended to include the administration of low-dose bevacizumab after the drainage of malignant ascites and has shown efficacy and a tolerable safety profile, especially in symptomatic patients with chemotherapy-resistant ovarian, fallopian tube or primary peritoneal cancer (219).
FRα-targeting drugs
The metabolism of folate is key for cellular functions such as DNA synthesis, methylation and repair (220). Folate and its derivatives enter cells via endocytosis, which is aided by the transmembrane glycoprotein FRα (221). In most cases, FRα expression is limited to specific tissue, such as the kidney, retina, lung, choroid plexus and placenta. FRα is markedly upregulated in several types of cancer, such as those that impact the ovaries, breast, lung and endometrium (222,223). Due to the capacity to enter the cell post-ligand binding and its selective upregulation, FRα is a desirable target for cancer drug delivery schemes (96). FRα is typically upregulated in ovarian carcinoma, while this receptor is absent in normal ovarian epithelium. Notably, the levels of soluble FRα (sFRα) in circulation are associated with tumor FRα expression, disease progression and treatment outcomes in patients with EOC. As a result, sFRα shows improved diagnostic accuracy compared with serum CA-125 levels, suggesting it may be a useful biomarker for early EOC detection (224).
Approaches that target FRα have become increasingly attractive in the treatment for ovarian cancer (96,225,226). Antibody-drug conjugates are a specialized class of drugs to deliver chemotherapeutics to tumor sites in a targeted and selective manner. A common antibody-drug conjugate that targets FRα, MIRV, combines an anti-FRα antibody with DM4, a strong tubulin-targeting agent. This compound functions by binding FRα and allowing for the targeted delivery of DM4 to tumor cells, which optimizes the balance of beneficial to side effects (225). Treatment of platinum-resistant EOC using MIRV has been evaluated (225). Following encouraging results from the phase III trial SORAYA (trial no. NCT04296890), the FDA granted accelerated approval in 2022 for use in patients with FRα-positive, platinum-resistant EOC who have previously received systemic anticancer therapy (225,227). The humanized monoclonal antibody farletuzumab is another treatment strategy involving FRα. Preclinical research has demonstrated that farletuzumab may hinder FRα-expressing ovarian cancer cell proliferation (228,229). However, clinical trials evaluating farletuzumab in combination with other therapies for platinum-sensitive EOC (trial nos. NCT00318370 and NCT02289950) have yielded conflicting outcomes (96,226).
STRO-002 is an innovative antibody-drug conjugate targeting FRα, currently under clinical investigation for ovarian and endometrial cancer. Preclinical studies have demonstrated that a single dose of STRO-002 markedly inhibited tumor growth in FRα-expressing xenograft and patient-derived models, with enhanced efficacy when combined with carboplatin or bevacizumab (230,231). These findings underscore its potential as a promising therapeutic option for FRα-expressing cancer, including ovarian, endometrial and non-small cell lung cancer (226).
Treatment options for ovarian cancer either in the initial stages or if recurrence occurs are presented in Fig. 1.
Drug repurposing
Drug repurposing is the process of finding novel therapeutic uses for approved medications outside of their initial indications (Table IV) (232). For example, vitamin D and its analogs, are being studied for the treatment of ovarian cancer (233–237). These steroid-like compounds have demonstrated antitumor activity in preclinical models, in addition to their typical physiological roles. In particular, they suppress cell proliferation and the potential for metastasis while causing tumor cell differentiation and apoptosis (238). As a result, synthetic vitamin D analogs, designed to mitigate the risk of hypercalcemia, have been developed for targeting malignant disease, such as breast, colorectal and prostate cancer. By contrast, the effect of vitamin D and its analogs on ovarian cancer remains unclear (96,239). Vitamin D-based treatment may improve the effectiveness of PARP inhibitors and chemotherapeutics (240). The active form of vitamin D, calcitriol, has been shown to inhibit PARP1 activity in both cell-free and cellular assays. This suggests that vitamin D supplementation may enhance the efficacy of pharmacologic PARP1 inhibitors through a synergistic inhibitory effect (240). Furthermore, combining vitamin D with immunotherapy may be advantageous due to its immunomodulatory effects (241,242). To the best of our knowledge, there are few clinical studies that have evaluated the effectiveness of vitamin D-based treatment for ovarian cancer (96,239).
Coagulation-targeting approaches
Venous thromboembolism (VTE), which has an incidence rate of 10–30%, is a common diagnosis in patients with ovarian tumors (243). For patients with cancer, this thrombotic event is the second most common cause of mortality. Notably, most patients with cancer show signs of hypercoagulability even in the absence of VTE (244). Within the tumor microenvironment, cancer cells produce tissue factor (TF) independently and promote TF production by normal cells. The pro-tumorigenic functions of TF include tumor cell proliferation, maintenance of cancer stemness, angiogenesis, immune evasion and metastasis through both clotting-dependent and -independent mechanisms (245–247). In several tumor types, including ovarian cancer, upregulation of TF is associated with a poor prognosis (96).
The FDA recently approved tisotumab vedotin (Tivdak™), a human antibody-drug conjugate specific to TF and associated with the tubulin-targeting agent monomethyl auristatin E, for the treatment of recurrent or metastatic cervical cancer (248). In patients with platinum-resistant ovarian cancer, the drug exhibits a favorable safety profile and notable antitumor activity, according to the phase I/II innovaTV-201 trial (trial no. NCT02001623) (249). These findings support the continued investigation of tisotumab vedotin in this patient cohort.
Key clinical trials for ovarian cancer treatment are listed in Table V.
Future perspective and directions
Despite notable advancements in targeted therapy, ICIs and biomarker-driven treatment, several gaps remain in the treatment of ovarian cancer. These gaps include underserved patient populations, the role of precision medicine and the integration of comprehensive molecular profiling in clinical decision-making.
Underserved patient populations
While biomarker-driven treatment strategies have improved outcomes for certain groups, disparities persist, particularly for ethnic minorities, elderly patients and those with rare ovarian cancer subtypes. Clinical trials have predominantly included Caucasian populations, leading to limited data on Black, Hispanic and Asian patients (102,113). Studies have indicated that African American and Hispanic patients experience higher mortality rates and lower enrollment in clinical trials (250), which may limit access to novel therapies such as PARP inhibitors and immunotherapy (251). Additionally, several clinical trials exclude older patients (aged ≥70 years) or those with multiple comorbidities, despite the fact that ovarian cancer predominantly affects postmenopausal patients (252–254). Accordingly, trial findings may not be generalizable to the broader patient population. This is revealed by the limited number of trials, such as ROSiA, ENGOT-OV16/NOVA, and GOG-182 trials, that were conducted on patients aged 70 or more (252–254). The impact of aggressive treatments, such as PARP inhibitors, ICIs and combination regimens, on older or frail patients requires further investigation.
Another underserved subgroup is patients with rare ovarian cancer subtypes, such as LGSOC, clear cell and mucinous ovarian cancer. Most phase III clinical trials focus on HGSOC, which represents the majority of cases, while rarer subtypes exhibit distinct molecular alterations that may render standard therapies less effective (46,113,126,255,256). For example, LGSOC often harbors KRAS or BRAF mutations, suggesting that MEK inhibitors may be a more effective approach compared with traditional platinum-based chemotherapy (134). However, these alternatives remain underexplored, emphasizing the need for histology-specific clinical trials.
Role of precision medicine
While precision medicine has transformed ovarian cancer treatment, current biomarker-based strategies remain incomplete. Presently, treatment decisions are primarily guided by BRCAm status and HRD testing, but these biomarkers do not fully capture the complexity of ovarian cancer biology. A large proportion of HRD-negative tumors respond to PARP inhibitors, suggesting that improved stratification tools are needed to identify the true PARP-sensitive patient population. Additionally, certain HRD-positive tumors exhibit intrinsic resistance to PARP inhibitors, highlighting the limitations of genomic testing alone (257).
Beyond HRD testing, the role of other emerging biomarkers, such as tumor mutation burden (TMB), microsatellite instability (MSI) and PD-L1 expression, is uncertain in ovarian cancer. While high TMB and MSI are used as predictive biomarkers for checkpoint inhibitor therapy in several types of cancer, their utility in ovarian cancer has not been well established (258,259). Similarly, PD-L1 expression, a key biomarker for ICIs in lung and breast cancer, has shown limited predictive value in ovarian cancer trials (260–262). The lack of validated predictive biomarkers for immunotherapy is a major limitation, contributing to the low success of ICIs in ovarian cancer compared with other solid tumors.
Another gap in precision medicine is the reliance on genomic HRD assays, which assess DNA repair deficiencies at a static point in time, but may not accurately predict treatment response. Some researchers argue that functional HRD testing, which directly measures the ability of a tumor to repair DNA damage, may be a more reliable biomarker for PARP inhibitor sensitivity (263,264). As some HRD-negative tumors benefit from PARP inhibitors, the development of more comprehensive functional assays may improve patient selection and maximize treatment efficacy (265).
Tailoring treatment using comprehensive molecular profiling
The integration of genomic, transcriptomic and proteomic data may transform ovarian cancer treatment by identifying novel drug targets and guiding therapy selection (266). However, challenges remain in implementing comprehensive molecular profiling in routine clinical practice. A notable limitation is the lack of real-time, actionable molecular data. Most genomic profiling methods provide retrospective insight, but real-time molecular testing is necessary to dynamically adjust treatment based on tumor evolution (267). The integration of liquid biopsy, which analyzes ctDNA or circulating tumor cells, may offer a minimally invasive way to track treatment response and resistance mechanisms in real-time (268).
Another challenge is the integration of multi-omic data to create a holistic view of tumor biology. Several targeted therapies focus on a single pathway, yet ovarian cancer is heterogeneous, and adaptive resistance mechanisms often emerge. For example, while PARP inhibitors target DNA repair defects, resistance can develop through secondary BRCA reversion mutation or upregulation of drug efflux transporters. The ability to combine genomic, epigenomic, transcriptomic and proteomic insight may facilitate more precise, patient-specific treatment strategies (269). Additionally, targetable mutations in TP53, cyclin E1, KRAS and PI3K/AKT pathways remain underexplored in ovarian cancer, highlighting the need for novel drug development beyond BRCA/PARP inhibitors.
Conclusion
Targeted therapies have replaced surgery and chemotherapy as the mainstays of ovarian cancer treatment, notably improving patient outcomes. However, optimal patient selection, resistance mechanisms and the long-term effectiveness of these medicines are key concerns. Although targeted treatments provide improved control of ovarian cancer, issues with cost-effectiveness, accessibility and treatment-associated toxicity exist in practical application. Furthermore, due to the possibility of medication resistance and secondary malignancy, it is still unclear how long the response lasts and how it affects long-term survival. Future studies should concentrate on improving biomarker-driven strategies for treatment, identifying combination treatments to overcome resistance and developing affordable models for greater accessibility globally.
Acknowledgements
Not applicable.
Funding
Funding: No funding was received.
Availability of data and materials
Not applicable.
Authors' contributions
KA designed and performed the narrative review, and drafted and proofread the article critically. AZA helped in writing the section on poly(ADP-ribose) polymerase inhibitors, immunotherapy, angiogenesis inhibitors and targeted therapy, and critically revised the manuscript. GBH helped in writing the combination therapy, MEK inhibitors and microRNAs sections, and revised the article critically for intellectual content. AFA contributed to writing of the section on hyperthermic intraperitoneal chemotherapy. OSG, AA, SM and MJA revised the manuscript. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.
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
Reid BM, Permuth JB and Sellers TA: Epidemiology of ovarian cancer: A review. Cancer Biol Med. 14:9–32. 2017. View Article : Google Scholar : PubMed/NCBI | |
Stewart C, Ralyea C and Lockwood S: Ovarian cancer: An integrated review. Semin Oncol Nurs. 35:151–156. 2019. View Article : Google Scholar : PubMed/NCBI | |
Gaona-Luviano P, Medina-Gaona LA and Magaña-Pérez K: Epidemiology of ovarian cancer. Chin Clin Oncol. 9:472020. View Article : Google Scholar : PubMed/NCBI | |
Khanlarkhani N, Azizi E, Amidi F, Khodarahmian M, Salehi E, Pazhohan A, Farhood B, Mortezae K, Goradel NH and Nashtaei MS: Metabolic risk factors of ovarian cancer: A review. JBRA Assist Reprod. 26:335–347. 2022.PubMed/NCBI | |
Momenimovahed Z, Tiznobaik A, Taheri S and Salehiniya H: Ovarian cancer in the world: Epidemiology and risk factors. Int J Womens Health. 11:287–299. 2019. View Article : Google Scholar : PubMed/NCBI | |
Nebgen DR, Lu KH and Bast RC: Novel approaches to ovarian cancer screening. Curr Oncol Rep. 21:752019. View Article : Google Scholar : PubMed/NCBI | |
Huang J, Chan WC, Ngai CH, Lok V, Zhang L, Lucero-Prisno DE III, Xu W, Zheng ZJ, Elcarte E, Withers M, et al: Worldwide burden, risk factors, and temporal trends of ovarian cancer: A global study. Cancers (Basel). 14:22302022. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Sambasivan S: Epithelial ovarian cancer: Review article. Cancer Treat Res Commun. 33:1006292022. View Article : Google Scholar : PubMed/NCBI | |
Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar : PubMed/NCBI | |
Gajjar K, Ogden G, Mujahid MI and Razvi K: Symptoms and risk factors of ovarian cancer: A survey in primary care. ISRN Obstet Gynecol. 2012:7541972012. View Article : Google Scholar : PubMed/NCBI | |
Goff B: Symptoms associated with ovarian cancer. Clin Obstet Gynecol. 55:36–42. 2012. View Article : Google Scholar : PubMed/NCBI | |
Horackova K, Janatova M, Kleiblova P, Kleibl Z and Soukupova J: Early-onset ovarian cancer <30 years: What do we know about its genetic predisposition? Int J Mol Sci. 24:170202023. View Article : Google Scholar : PubMed/NCBI | |
Norquist BM, Harrell MI, Brady MF, Walsh T, Lee MK, Gulsuner S, Bernards SS, Casadei S, Yi Q, Burger RA, et al: Inherited mutations in women with ovarian carcinoma. JAMA Oncol. 2:482–490. 2016. View Article : Google Scholar : PubMed/NCBI | |
Choi JH, Wong AST, Huang HF and Leung PCK: Gonadotropins and ovarian cancer. Endocr Rev. 28:440–461. 2007. View Article : Google Scholar : PubMed/NCBI | |
Elias KM, Guo J and Bast RC Jr: Early detection of ovarian cancer. Hematol Oncol Clin North Am. 32:903–914. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zamwar UM and Anjankar AP: Aetiology, epidemiology, histopathology, classification, detailed evaluation, and treatment of ovarian cancer. Cureus. 14:e305612022.PubMed/NCBI | |
Berek JS, Renz M, Kehoe S, Kumar L and Friedlander M: Cancer of the ovary, fallopian tube, and peritoneum: 2021 update. Int J Gynecol Obstet. 155 (Suppl 1):S61–S85. 2021. View Article : Google Scholar | |
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 | |
Gupta KK, Gupta VK and Naumann RW: Ovarian cancer: Screening and future directions. Int J Gynecol Cancer. 29:195–200. 2019. View Article : Google Scholar : PubMed/NCBI | |
Jacobs IJ, Menon U, Ryan A, Gentry-Maharaj A, Burnell M, Kalsi JK, Amso NN, Apostolidou S, Benjamin E, Cruickshank D, et al: Ovarian cancer screening and mortality in the UK collaborative trial of ovarian cancer screening (UKCTOCS): A randomised controlled trial. Lancet. 387:945–956. 2016. View Article : Google Scholar : PubMed/NCBI | |
Hu X, Zhang J and Cao Y: Factors associated with serum CA125 level in women without ovarian cancer in the United States: A population-based study. BMC Cancer. 22:5442022. View Article : Google Scholar : PubMed/NCBI | |
Skates SJ, Menon U, MacDonald N, Rosenthal AN, Oram DH, Knapp RC and Jacobs IJ: Calculation of the risk of ovarian cancer from serial CA-125 values for preclinical detection in postmenopausal women. J Clin Oncol. 21 (10 Suppl):206s–210s. 2003. View Article : Google Scholar : PubMed/NCBI | |
Adolphi NL, Butler KS, Lovato DM, Tessier TE, Trujillo JE, Hathaway HJ, Fegan DL, Monson TC, Stevens TE, Huber DL, et al: Imaging of Her2-targeted magnetic nanoparticles for breast cancer detection: Comparison of SQUID-detected magnetic relaxometry and MRI. Contrast Media Mol Imaging. 7:308–319. 2012. View Article : Google Scholar : PubMed/NCBI | |
Wan W, Liu H, Zou J, Xie T, Zhang G, Ying W and Zou X: The optimization and application of photodynamic diagnosis and autofluorescence imaging in tumor diagnosis and guided surgery: Current status and future prospects. Front Oncol. 14:15034042025. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Hernandez-Lopez LA and Elizalde-Mendez A: How far should we go in optimal cytoreductive surgery for ovarian cancer? Chin Clin Oncol. 9:702020. View Article : Google Scholar : PubMed/NCBI | |
Haghighat S: New treatment of advanced ovarian cancer: A literature review. J Obstet Gynecol Cancer Res. 4:131–134. 2019. View Article : Google Scholar | |
Lukanović D, Kobal B and Černe K: Ovarian cancer: Treatment and resistance to pharmacotherapy. Reprod Med. 3:127–140. 2022. View Article : Google Scholar | |
Orr B and Edwards RP: Diagnosis and treatment of ovarian cancer. Hematol Oncol Clin North Am. 32:943–964. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ghirardi V, Fagotti A, Ansaloni L, Valle M, Roviello F, Sorrentino L, Accarpio F, Baiocchi G, Piccini L, De Simone M, et al: Diagnostic and therapeutic pathway of advanced ovarian cancer with peritoneal metastases. Cancers (Basel). 15:4072023. View Article : Google Scholar : PubMed/NCBI | |
Chang SJ, Hodeib M, Chang J and Bristow RE: Survival impact of complete cytoreduction to no gross residual disease for advanced-stage ovarian cancer: A meta-analysis. Gynecol Oncol. 130:493–498. 2013. View Article : Google Scholar : PubMed/NCBI | |
Wright AA, Bohlke K, Armstrong DK, Bookman MA, Cliby WA, Coleman RL, Dizon DS, Kash JJ, Meyer LA, Moore KN, et al: Neoadjuvant chemotherapy for newly diagnosed, advanced ovarian cancer: Society of gynecologic oncology and American society of clinical oncology clinical practice guideline. Gynecol Oncol. 143:3–15. 2016. View Article : Google Scholar : PubMed/NCBI | |
Armstrong DK and Walker JL: Role of intraperitoneal therapy in the initial management of ovarian cancer. J Clin Oncol. 37:2416–2419. 2019. View Article : Google Scholar : PubMed/NCBI | |
Matulonis UA, Sood AK, Fallowfield L, Howitt BE, Sehouli J and Karlan BY: Ovarian cancer. Nat Rev Dis Primer. 2:160612016. View Article : Google Scholar : PubMed/NCBI | |
Vergote I, Tropé CG, Amant F, Kristensen GB, Ehlen T, Johnson N, Verheijen RHM, van der Burg MEL, Lacave A, Panici PB, et al: Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 363:943–953. 2010. View Article : Google Scholar : PubMed/NCBI | |
du Bois A, Reuss A, Pujade-Lauraine E, Harter P, Ray-Coquard I and Pfisterer J: Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: By the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d'Investigateurs Nationaux Pour les Etudes des Cancers de l'Ovaire (GINECO). Cancer. 115:1234–1244. 2009. View Article : Google Scholar : PubMed/NCBI | |
Gallotta V, Certelli C, Oliva R, Rosati A, Federico A, Loverro M, Lodoli C, Foschi N, Lathouras K, Fagotti A and Scambia G: Robotic surgery in ovarian cancer. Best Pract Res Clin Obstet Gynaecol. 90:1023912023. View Article : Google Scholar : PubMed/NCBI | |
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2022. CA Cancer J Clin. 72:7–33. 2022. View Article : Google Scholar : PubMed/NCBI | |
Finch L and Chi DS: An overview of the current debate between using minimally invasive surgery versus laparotomy for interval cytoreductive surgery in epithelial ovarian cancer. J Gynecol Oncol. 34:e842023. View Article : Google Scholar : PubMed/NCBI | |
Kumar A and Cliby WA: Advanced ovarian cancer: Weighing the risks and benefits of surgery. Clin Obstet Gynecol. 63:74–79. 2019. View Article : Google Scholar | |
Ramirez PT: Standardizing ovarian cancer surgery and peri-operative care: A European society of gynecological oncology (ESGO) consensus statement. Int J Gynecol Cancer. 31:1207–1208. 2021. View Article : Google Scholar : PubMed/NCBI | |
McGuire WP III and Markman M: Primary ovarian cancer chemotherapy: Current standards of care. Br J Cancer. 89 (Suppl 3):S3–S8. 2003. View Article : Google Scholar : PubMed/NCBI | |
Pujade-Lauraine E, Hilpert F, Weber B, Reuss A, Poveda A, Kristensen G, Sorio R, Vergote I, Witteveen P, Bamias A, et al: Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: The AURELIA open-label randomized phase III trial. J Clin Oncol. 32:1302–1308. 2014. View Article : Google Scholar : PubMed/NCBI | |
Mikuła-Pietrasik J, Witucka A, Pakuła M, Uruski P, Begier-Krasińska B, Niklas A, Tykarski A and Książek K: Comprehensive review on how platinum- and taxane-based chemotherapy of ovarian cancer affects biology of normal cells. Cell Mol Life Sci. 76:681–697. 2019. View Article : Google Scholar : PubMed/NCBI | |
Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, Lisyanskaya A, Floquet A, Leary A, Sonke GS, et al: Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 379:2495–2505. 2018. View Article : Google Scholar : PubMed/NCBI | |
Mirza MR, Lundqvist EA, Birrer MJ, dePont Christensen R, Nyvang GB, Malander S, Anttila M, Werner TL, Lund B, Lindahl G, et al: Niraparib plus bevacizumab versus niraparib alone for platinum-sensitive recurrent ovarian cancer (NSGO-AVANOVA2/ENGOT-ov24): A randomised, phase 2, superiority trial. Lancet Oncol. 20:1409–1419. 2019. View Article : Google Scholar : PubMed/NCBI | |
Oza AM, Cook AD, Pfisterer J, Embleton A, Ledermann JA, Pujade-Lauraine E, Kristensen G, Carey MS, Beale P, Cervantes A, et al: Standard chemotherapy with or without bevacizumab for women with newly diagnosed ovarian cancer (ICON7): Overall survival results of a phase 3 randomised trial. Lancet Oncol. 16:928–936. 2015. View Article : Google Scholar : PubMed/NCBI | |
Baert T, Ferrero A, Sehouli J, O'Donnell DM, González-Martín A, Joly F, van der Velden J, Blecharz P, Tan DSP, Querleu D, et al: The systemic treatment of recurrent ovarian cancer revisited. Ann Oncol. 32:710–725. 2021. View Article : Google Scholar : PubMed/NCBI | |
Dasari S and Tchounwou PB: Cisplatin in cancer therapy: Molecular mechanisms of action. Eur J Pharmacol. 740:364–378. 2014. View Article : Google Scholar : PubMed/NCBI | |
Galluzzi L, Senovilla L, Vitale I, Michels J, Martins I, Kepp O, Castedo M and Kroemer G: Molecular mechanisms of cisplatin resistance. Oncogene. 31:1869–1883. 2012. View Article : Google Scholar : PubMed/NCBI | |
Bowtell DD, Böhm S, Ahmed AA, Aspuria PJ, Bast RC, Beral V, Berek JS, Birrer MJ, Blagden S, Bookman MA, et al: Rethinking ovarian cancer II: Reducing mortality from high-grade serous ovarian cancer. Nat Rev Cancer. 15:668–679. 2015. View Article : Google Scholar : PubMed/NCBI | |
Vergote I, Denys H, Greve JD, Gennigens C, de Vijver V, Kerger J, Vuylsteke P and Baurain JF: Treatment algorithm in patients with ovarian cancer. Facts Views Vis Obgyn. 12:227–239. 2020.PubMed/NCBI | |
Coada CA, Dondi G, Ravegnini G, Di Costanzo S, Tesei M, Fiuzzi E, Di Stanislao M, Giunchi S, Zamagni C, Bovicelli A, et al: Optimal number of neoadjuvant chemotherapy cycles prior to interval debulking surgery in advanced epithelial ovarian cancer: A systematic review and meta-analysis of progression-free survival and overall survival. J Gynecol Oncol. 34:e822023. View Article : Google Scholar : PubMed/NCBI | |
Redondo A, Guerra E, Manso L, Martin-Lorente C, Martinez-Garcia J, Perez-Fidalgo JA, Varela MQ, Rubio MJ, Barretina-Ginesta MP and Gonzalez-Martin A: SEOM clinical guideline in ovarian cancer (2020). Clin Transl Oncol. 23:961–968. 2021. View Article : Google Scholar : PubMed/NCBI | |
Akter S, Rahman MA, Hasan MN, Akhter H, Noor P, Islam R, Shin Y, Rahman MDH, Gazi MS, Huda MN, et al: Recent advances in ovarian cancer: Therapeutic strategies, potential biomarkers, and technological improvements. Cells. 11:6502022. View Article : Google Scholar : PubMed/NCBI | |
Faraoni I and Graziani G: Role of BRCA mutations in cancer treatment with Poly(ADP-ribose) polymerase (PARP) inhibitors. Cancers (Basel). 10:4872018. View Article : Google Scholar : PubMed/NCBI | |
Levit SL and Tang C: Polymeric nanoparticle delivery of combination therapy with synergistic effects in ovarian cancer. Nanomaterials (Basel). 11:10482021. View Article : Google Scholar : PubMed/NCBI | |
Matsuo K, Eno ML, Im DD and Rosenshein NB: Chemotherapy time interval and development of platinum and taxane resistance in ovarian, fallopian, and peritoneal carcinomas. Arch Gynecol Obstet. 281:325–328. 2010. View Article : Google Scholar : PubMed/NCBI | |
Rauh-Hain JA, Nitschmann CC, Worley MJ Jr, Bradford LS, Berkowitz RS, Schorge JO, Campos SM, del Carmen MG and Horowitz NS: Platinum resistance after neoadjuvant chemotherapy compared to primary surgery in patients with advanced epithelial ovarian carcinoma. Gynecol Oncol. 129:63–68. 2025. View Article : Google Scholar | |
Pokhriyal R, Hariprasad R, Kumar L and Hariprasad G: Chemotherapy resistance in advanced ovarian cancer patients. Biomark Cancer. 11:1179299X19860812019. View Article : Google Scholar : PubMed/NCBI | |
Basourakos SP, Li L, Aparicio AM, Corn PG, Kim J and Thompson TC: Combination platinum-based and DNA damage response-targeting cancer therapy: Evolution and future directions. Curr Med Chem. 24:1586–1606. 2017. View Article : Google Scholar : PubMed/NCBI | |
Luqmani YA: Mechanisms of drug resistance in cancer chemotherapy. Med Princ Pract. 14 (Suppl 1):S35–S48. 2005. View Article : Google Scholar : PubMed/NCBI | |
Bykov VJN, Eriksson SE, Bianchi J and Wiman KG: Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer. 18:89–102. 2018. View Article : Google Scholar : PubMed/NCBI | |
Yang L, Xie HJ, Li YY, Wang X, Liu XX and Mai J: Molecular mechanisms of platinum-based chemotherapy resistance in ovarian cancer (Review). Oncol Rep. 47:822022. View Article : Google Scholar : PubMed/NCBI | |
Mihanfar A, Fattahi A and Nejabati HR: MicroRNA-mediated drug resistance in ovarian cancer. J Cell Physiol. 234:3180–3191. 2019. View Article : Google Scholar : PubMed/NCBI | |
Puris E, Fricker G and Gynther M: The role of solute carrier transporters in efficient anticancer drug delivery and therapy. Pharmaceutics. 15:3642023. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Wang X, Zhu X, Zhong L, Jiang Q, Wang Y, Tang Q, Li Q, Zhang C, Wang H and Zou D: Drug resistance in ovarian cancer: from mechanism to clinical trial. Mol Cancer. 23:662024. View Article : Google Scholar : PubMed/NCBI | |
Alshamrani AA: Roles of microRNAs in ovarian cancer tumorigenesis: Two decades later, what have we learned? Front Oncol. 10:10842020. View Article : Google Scholar : PubMed/NCBI | |
Nguyen VHL, Yue C, Du KY, Salem M, O'Brien J and Peng C: The role of microRNAs in epithelial ovarian cancer metastasis. Int J Mol Sci. 21:70932020. View Article : Google Scholar : PubMed/NCBI | |
Vaidyanathan A, Sawers L, Gannon AL, Chakravarty P, Scott AL, Bray SE, Ferguson MJ and Smith G: ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. Br J Cancer. 115:431–441. 2016. View Article : Google Scholar : PubMed/NCBI | |
Rottenberg S, Jaspers JE, Kersbergen A, van Der Burg E, Nygren AOH, Zander SAL, Derksen PW, de Bruin M, Zevenhoven J, Lau A, et al: High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proc Natl Acad Sci USA. 105:17079–17084. 2008. View Article : Google Scholar : PubMed/NCBI | |
Zhang B, Kang Z, Zhang J, Kang Y, Liang L, Liu Y, Liu Y and Wang Q: Simultaneous binding mechanism of multiple substrates for multidrug resistance transporter P-glycoprotein. Phys Chem Chem Phys. 23:4530–4543. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kazmierczak D, Jopek K, Sterzynska K, Nowicki M, Rucinski M and Januchowski R: The profile of MicroRNA expression and potential role in the regulation of drug-resistant genes in cisplatin- and paclitaxel-resistant ovarian cancer cell lines. Int J Mol Sci. 23:5262022. View Article : Google Scholar : PubMed/NCBI | |
Lee JH, Chae JW, Kim JK, Kim HJ, Chung JY and Kim YH: Inhibition of cisplatin-resistance by RNA interference targeting metallothionein using reducible oligo-peptoplex. J Control Release. 215:82–90. 2015. View Article : Google Scholar : PubMed/NCBI | |
Boušová I and Skálová L: Inhibition and induction of glutathione S-transferases by flavonoids: Possible pharmacological and toxicological consequences. Drug Metab Rev. 44:267–286. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Xie S, Zhou L, Tang X, Guan X, Deng M, Zheng H, Wang Y, Lu R and Guo L: Up-regulation of GSTT1 in serous ovarian cancer associated with resistance to TAXOL/carboplatin. J Ovarian Res. 14:1222021. View Article : Google Scholar : PubMed/NCBI | |
Tagawa T, Morgan R, Yen Y and Mortimer J: Ovarian cancer: Opportunity for targeted therapy. J Oncol. 2012:6824802012. View Article : Google Scholar : PubMed/NCBI | |
Duan P, Fan L, Gao Q, Silwal M, Ren M, Shen Y and Qu W: Targeted therapy of ovarian cancer with angiogenesis inhibitors. Curr Drug Targets. 18:1171–1178. 2017. View Article : Google Scholar : PubMed/NCBI | |
Pulla P, Lakshmanan K, Byran G, Rajagopal K, Krishnamurthy PT and Palati DJ: A review on recent PARP inhibitor advancements in cancer therapy. Curr Enzyme Inhib. 18: View Article : Google Scholar : 2022. | |
Suh YJ, Lee B, Kim K, Jeong Y, Choi HY, Hwang SO and Kim YB: Bevacizumab versus PARP-inhibitors in women with newly diagnosed ovarian cancer: A network meta-analysis. BMC Cancer. 22:3462022. View Article : Google Scholar : PubMed/NCBI | |
Ferrara N, Hillan KJ and Novotny W: Bevacizumab (Avastin), a humanized anti-VEGF monoclonal antibody for cancer therapy. Biochem Biophys Res Commun. 333:328–335. 2005. View Article : Google Scholar : PubMed/NCBI | |
Chen A: PARP inhibitors: Its role in treatment of cancer. Chin J Cancer. 30:463–471. 2011. View Article : Google Scholar : PubMed/NCBI | |
O'Malley DM, Krivak TC, Kabil N, Munley J and Moore KN: PARP inhibitors in ovarian cancer: A review. Target Oncol. 18:471–503. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sato K, Koyasu M, Nomura S, Sato Y, Kita M, Ashihara Y, Adachi Y, Ohno S, Iwase T, Kitagawa D, et al: Mutation status of RAD51C, PALB2 and BRIP1 in 100 Japanese familial breast cancer cases without BRCA1 and BRCA2 mutations. Cancer Sci. 108:2287–2294. 2017. View Article : Google Scholar : PubMed/NCBI | |
Lee JM, Ledermann JA and Kohn EC: PARP Inhibitors for BRCA1/2 mutation-associated and BRCA-like malignancies. Ann Oncol. 25:32–40. 2014. View Article : Google Scholar : PubMed/NCBI | |
Domchek SM, Postel-Vinay S, Im SA, Park YH, Delord JP, Italiano A, Alexandre J, You B, Bastian S, Krebs MG, 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. 21:1155–1164. 2020. View Article : Google Scholar : PubMed/NCBI | |
Drew Y, Kaufman B, Banerjee S, Lortholary A, Hong SH, Park YH, Zimmermann S, Roxburgh P, Ferguson M, Alvarez RH, et al: Phase II study of olaparib + durvalumab (MEDIOLA): Updated results in germline BRCA-mutated platinum-sensitive relapsed (PSR) ovarian cancer (OC). Ann Oncol. 30:v485–v486. 2019. View Article : Google Scholar | |
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 | |
Orbach D, Carton M, Khadir SK, Feuilly M, Kurtinecz M, Phil D, Vokuhl C, Koscielniak E, Pierron G, Lemelle L and Sparber-Sauer M: Therapeutic benefit of larotrectinib over the historical standard of care in patients with locally advanced or metastatic infantile fibrosarcoma (EPI VITRAKVI study). ESMO Open. 9:1030062024. View Article : Google Scholar : PubMed/NCBI | |
Gouda MA and Subbiah V: Precision oncology with selective RET inhibitor selpercatinib in RET-rearranged cancers. Ther Adv Med Oncol. 15:175883592311770152023. View Article : Google Scholar : PubMed/NCBI | |
Dinkins K, Barton W, Wheeler L, Smith HJ, Mythreye K and Arend RC: Targeted therapy in high grade serous ovarian cancer: A literature review. Gynecol Oncol Rep. 54:1014502024. View Article : Google Scholar : PubMed/NCBI | |
Luvero D, Angioli R, Celoro F, Plotti F, Terranova C, Guzzo F, Cundari GB, Liparulo F, Verdone C and Montera R: Tailored treatment strategies in first line therapy for ovarian cancer patients: A critical review of the literature. Pharmaceuticals (Basel). 17:7782024. View Article : Google Scholar : PubMed/NCBI | |
Hillmann J, Maass N, Bauerschlag DO and Flörkemeier I: Promising new drugs and therapeutic approaches for treatment of ovarian cancer-targeting the hallmarks of cancer. BMC Med. 23:102025. View Article : Google Scholar : PubMed/NCBI | |
Zeimet A, Wieser V, Knoll K, Reimer D and Marth C: PARP inhibitors in the treatment of ovarian cancer. Memo Mag Eur Med Oncol. 13:198–201. 2020. | |
Tavares V, Marques IS, de Melo IG, Assis J, Pereira D and Medeiros R: Paradigm shift: A comprehensive review of ovarian cancer management in an era of advancements. Int J Mol Sci. 25:18452024. View Article : Google Scholar : PubMed/NCBI | |
Kolesnichenko M and Scheidereit C: Synthetic lethality by PARP inhibitors: New mechanism uncovered based on unresolved transcription-replication conflicts. Signal Transduct Target Ther. 9:1792024. View Article : Google Scholar : PubMed/NCBI | |
Ledermann JA and Pujade-Lauraine E: Olaparib as maintenance treatment for patients with platinum-sensitive relapsed ovarian cancer. Ther Adv Med Oncol. 11:17588359198497532019. View Article : Google Scholar : PubMed/NCBI | |
Bradley W, Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, Lisyanskaya A, Floquet A, Leary A, et al: Maintenance olaparib for patients with newly diagnosed, advanced ovarian cancer and a BRCA mutation: 5-year follow-up from SOLO1. Gynecol Oncol. 162:S25–S26. 2021. View Article : Google Scholar | |
Li N, Zhu J, Yin R, Wang J, Pan L, Kong B, Zheng H, Liu J, Wu X, Wang L, et al: Efficacy and safety of niraparib as maintenance treatment in patients with newly diagnosed advanced ovarian cancer using an individualized starting dose (PRIME Study): A randomized, double-blind, placebo-controlled, phase 3 trial (LBA 5). Gynecol Oncol. 166:S50–S51. 2022. View Article : Google Scholar | |
Tyszka M and Stec R: Niraparib maintenance in newly diagnosed advanced ovarian cancer-review and case series. Oncol Clin Pract. 19:62022. | |
O'Cearbhaill R, Perez-Fidalgo JA, Monk B, Tusquets I, McCormick C, Fuentes J, Moore RG, Vulsteke C, Shahin MS, Forget F, et al: Efficacy of niraparib by time of surgery and postoperative residual disease status: A post hoc analysis of patients in the PRIMA/ENGOT-OV26/GOG-3012 study. Gynecol Oncol. 166:36–43. 2022. View Article : Google Scholar : PubMed/NCBI | |
Monk B, Parkinson C, Lim M, O'Malley D, Oaknin A, Wilson M, Coleman RL, Lorusso D, Bessette P, Ghamande S, 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. 381:JCO.22.01003. 2022. | |
Ledermann J, Oza A, 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 | |
Mirza MR, Pignata S and Ledermann JA: Latest clinical evidence and further development of PARP inhibitors in ovarian cancer. Ann Oncol. 29:1366–1376. 2018. View Article : Google Scholar : PubMed/NCBI | |
Boussios S, Abson C, Moschetta M, Rassy E, Karathanasi A, Bhat T, Ghumman F, Sheriff M and Pavlidis N: Poly (ADP-Ribose) polymerase inhibitors: Talazoparib in ovarian cancer and beyond. Drugs R D. 20:55–73. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zimmer A, Nichols E, Cimino-Mathews A, Peer C, Cao L, Lee MJ, Kohn EC, Annunziata CM, Lipkowitz S, Trepel JB, et al: A phase I study of the PD-L1 inhibitor, durvalumab, in combination with a PARP inhibitor, olaparib, and a VEGFR1-3 inhibitor, cediranib, in recurrent women's cancers with biomarker analyses. J Immunother Cancer. 7:1972019. View Article : Google Scholar : PubMed/NCBI | |
Eskander R, Ledermann J, Birrer M, Fujiwara K, Gaillard S, Richardson G, Wei C, Baig MA, Zohren F and Monk BJ: JAVELIN ovarian PARP 100 study design: Phase III trial of avelumab + chemotherapy followed by avelumab + talazoparib maintenance in previously untreated epithelial ovarian cancer. J Clin Oncol. 37:TPS9. 2019. View Article : Google Scholar | |
Herencia-Ropero A, Llop-Guevara A, Staniszewska AD, Domènech-Vivó J, García-Galea E, Moles-Fernández A, Pedretti F, Domènech H, Rodríguez O, Guzmán M, et al: The PARP1 selective inhibitor saruparib (AZD5305) elicits potent and durable antitumor activity in patient-derived BRCA1/2-associated cancer models. Genome Med. 16:1072024. View Article : Google Scholar : PubMed/NCBI | |
Muzzana M, Broggini M and Damia G: The landscape of PARP inhibitors in solid cancers. Onco Targets Ther. 18:297–317. 2025. View Article : Google Scholar : PubMed/NCBI | |
American Association for Cancer Research (AACR), . Next-generation PARP inhibitor demonstrates clinical benefit in patients with homologous recombination repair-deficient breast cancer [Internet]. AACR, Philadelphia. 2024.https://www.aacr.org/about-the-aacr/newsroom/news-releases/next-generation-parp-inhibitor-demonstrates-clinical-benefit-in-patients-with-homologous-recombination-repair-deficient-breast-cancer/February 22–2025 | |
MERCK, . LYNPARZA® (olaparib) Phase 3 PAOLA-1 trial significantly increased progression-free survival as first-line maintenance treatment with bevacizumab for newly-diagnosed advanced ovarian cancer [Internet]. Merck & Co., Inc., Rahway, NJ. 2019.https://www.merck.com/news/lynparza-olaparib-phase-3-paola-1-trial-significantly-increased-progression-free-survival-as-first-line- maintenance-treatment-with-bevacizumab-for-newly-diagnosed- advanced-ovarian-cancer/May 4–2025 | |
Ray-Coquard I, Leary A, Pignata S, Cropet C, González-Martín A, Marth C, Nagao S, Vergote I, Colombo N, Mäenpää J, et al: Olaparib plus bevacizumab first-line maintenance in ovarian cancer: Final overall survival results from the PAOLA-1/ENGOT-ov25 trial. Ann Oncol. 34:681–692. 2023. View Article : Google Scholar : PubMed/NCBI | |
Paclitaxel and Carboplatin and Veliparib in Ovarian Cancer and Ovarian Neoplasm-Clinical Trials Registry-ICH GCP [Internet], . https://ichgcp.net/clinical-trials-registry/NCT02470585?utm_source=chatgpt.comMay 4–2025 | |
Mirza MR, Benigno B, Dørum A, Mahner S, Bessette P, Barceló IB, Berton-Rigaud D, Ledermann JA, Rimel BJ, Herrstedt J, et al: Long-term safety in patients with recurrent ovarian cancer treated with niraparib versus placebo: Results from the phase III ENGOT-OV16/NOVA trial. Gynecol Oncol. 159:442–448. 2020. View Article : Google Scholar : PubMed/NCBI | |
Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, Mortimer P, Swaisland H, Lau A, O'Connor MJ, et al: Inhibition of poly(ADP-Ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med. 361:123–134. 2009. View Article : Google Scholar : PubMed/NCBI | |
Chan VKY, Yang R, Wong ICK and Li X: Cost-effectiveness of poly ADP-ribose polymerase inhibitors in cancer treatment: A systematic review. Front Pharmacol. 13:8911492022. View Article : Google Scholar : PubMed/NCBI | |
European Society for Medical Oncology (ESMO), . Elucidating a risk of developing second primary malignancy during and after treatment with PARP inhibitors [Internet]. ESMO; Lugano: 2021, https://www.esmo.org/oncology-news/elucidating-a-risk-of-developing-second-primary-malignancy-during-and-after-treatment-with-parp-inhibitorsFebruary 11–2025 | |
Desai C, Pathak A, Limaye S, Maniar V and Joshi A: A review on mechanisms of resistance to PARP inhibitors. Indian J Cancer. 59 (Suppl):S119–S129. 2022. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Yang Y, Jin D, Zhang Z, Shen K, Yang J, Chen H, Zhao X, Yang L and Lu H: PARP inhibitor resistance in breast and gynecological cancer: Resistance mechanisms and combination therapy strategies. Front Pharmacol. 13:9676332022. View Article : Google Scholar : PubMed/NCBI | |
Dana-Farber Cancer Institute, . Combination ATR and PARP Inhibitor (CAPRI) trial with AZD6738 and olaparib in recurrent ovarian cancer. Dana-Farber Cancer Institute, Inc.; Boston MA: https://www.dana-farber.org/clinical-trials/20-320May 4–2025 | |
Yang Y, Yang Y, Yang J, Zhao X and Wei X: Tumor microenvironment in ovarian cancer: Function and therapeutic strategy. Front Cell Dev Biol. 8:7582020. View Article : Google Scholar : PubMed/NCBI | |
Mei C, Gong W, Wang X, Lv Y, Zhang Y, Wu S and Zhu C: Anti-angiogenic therapy in ovarian cancer: Current understandings and prospects of precision medicine. Front Pharmacol. 14:11477172023. View Article : Google Scholar : PubMed/NCBI | |
Park SA, Jeong MS, Ha KT and Jang SB: Structure and function of vascular endothelial growth factor and its receptor system. BMB Rep. 51:73–78. 2018. View Article : Google Scholar : PubMed/NCBI | |
Burger RA, Sill MW, Monk BJ, Greer BE and Sorosky JI: Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: A gynecologic oncology group study. J Clin Oncol. 25:5165–5171. 2007. View Article : Google Scholar : PubMed/NCBI | |
Perren T, Swart AM, Pfisterer J, Ledermann JA, Lortholary A, Kristensen G, Carey MS, Beale P, Cervantes A and Oza A: ICON7: A phase iii randomised gynaecologic cancer intergroup trial of concurrent bevacizumab and chemotherapy followed by maintenance bevacizumab, versus chemotherapy alone in women with newly diagnosed epithelial ovarian (EOC), primary peritoneal (PPC) or fallopian tube cancer (FTC). Ann Oncol. 21:2010.PubMed/NCBI | |
Achtari C, Fink D, Günthert AR, Huober J, Pestalozzi B, Petignat P, von Moos R and Sessa C: Bevacizumab in the primary treatment of epithelial ovarian cancer-some comments on the latest results. chweizer Krebs-Bulletin = Bulletin Suisse du Cancer. 35:pp35–38. 2011. | |
Sfakianos GP, Numnum TM, Halverson CB, Panjeti D, Kendrick JE IV and Straughn JM Jr: The risk of gastrointestinal perforation and/or fistula in patients with recurrent ovarian cancer receiving bevacizumab compared to standard chemotherapy: A retrospective cohort study. Gynecol Oncol. 114:424–426. 2009. View Article : Google Scholar : PubMed/NCBI | |
Psyrri A, Kassar M, Yu Z, Bamias A, Weinberger PM, Markakis S, Kowalski D, Camp RL, Rimm DL and Dimopoulos MA: Effect of epidermal growth factor receptor expression level on survival in patients with epithelial ovarian cancer. Clin Cancer Res. 11:8637–8643. 2005. View Article : Google Scholar : PubMed/NCBI | |
Konner J, Schilder RJ, DeRosa FA, Gerst SR, Tew WP, Sabbatini PJ, Hensley ML, Spriggs DR and Aghajanian CA: A phase II study of cetuximab/paclitaxel/carboplatin for the initial treatment of advanced-stage ovarian, primary peritoneal, or fallopian tube cancer. Gynecol Oncol. 110:140–145. 2008. View Article : Google Scholar : PubMed/NCBI | |
Liang XJ and Shen J: Adverse events risk associated with angiogenesis inhibitors addition to therapy in ovarian cancer: A meta-analysis of randomized controlled trials. Eur Rev Med Pharmacol Sci. 20:2701–2790. 2016.PubMed/NCBI | |
Watson N and Al-Samkari H: Thrombotic and bleeding risk of angiogenesis inhibitors in patients with and without malignancy. J Thromb Haemost. 19:1852–1863. 2021. View Article : Google Scholar : PubMed/NCBI | |
Moujaber T, Balleine R, Gao B, Madsen I, Harnett P and DeFazio A: New therapeutic opportunities for low-grade serous ovarian cancer. Endocr Relat Cancer. 29:R1–R16. 2021. View Article : Google Scholar : PubMed/NCBI | |
Gershenson DM, Miller A, Brady WE, Paul J, Carty K, Rodgers W, Millan D, Coleman RL, Moore KN, Banerjee S, et al: Trametinib versus standard of care in patients with recurrent low-grade serous ovarian cancer (GOG 281/LOGS): An international, randomised, open-label, multicentre, phase 2/3 trial. Lancet. 399:541–553. 2022. View Article : Google Scholar : PubMed/NCBI | |
Grisham RN, Iyer G, Garg K, DeLair D, Hyman DM, Zhou Q, Iasonos A, Berger MF, Dao F, Spriggs DR, et al: BRAF Mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer. Cancer. 119:548–554. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hendrikse C, Theelen P, van der Ploeg P, Westgeest H, Boere I, 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 : PubMed/NCBI | |
Chen X, Chen Y, Lin X, Su S, Hou X, Zhang Q and Tian Y: The drug combination of SB202190 and SP600125 significantly inhibit the growth and metastasis of olaparib-resistant ovarian cancer cell. Curr Pharm Biotechnol. 19:506–513. 2018. View Article : Google Scholar : PubMed/NCBI | |
Cicenas J, Zalyte E, Rimkus A, Dapkus D, Noreika R and Urbonavicius S: JNK, p38, ERK, and SGK1 inhibitors in cancer. Cancers (Basel). 10:12017. View Article : Google Scholar : PubMed/NCBI | |
Santiago-O'Farrill J, Essien S, Figueroa M, Pang L, Amaravadi R, Lu Z and Bast RC: Abstract 3317: Autophagy protects ovarian cancer cells from olaparib-induced toxicity. Cancer Res. 77:33172017. View Article : Google Scholar | |
Welsh SJ and Corrie PG: Management of BRAF and MEK inhibitor toxicities in patients with metastatic melanoma. Ther Adv Med Oncol. 7:122–136. 2015. View Article : Google Scholar : PubMed/NCBI | |
Rupaimoole R and Slack FJ: MicroRNA therapeutics: Towards a new era for the management of cancer and other diseases. Nat Rev Drug Discov. 16:203–222. 2017. View Article : Google Scholar : PubMed/NCBI | |
Langhe R: microRNA and ovarian cancer. Adv Exp Med Biol. 889:119–151. 2015. View Article : Google Scholar : PubMed/NCBI | |
Chen Y, Cao XY, Li YN, Qiu YY, Li YN, Li W and Wang H: Reversal of cisplatin resistance by microRNA-139-5p-independent RNF2 downregulation and MAPK inhibition in ovarian cancer. Am J Physiol Cell Physiol. 315:C225–C235. 2018. View Article : Google Scholar : PubMed/NCBI | |
Wu Y, Wang T, Xia L and Zhang M: LncRNA WDFY3-AS2 promotes cisplatin resistance and the cancer stem cell in ovarian cancer by regulating hsa-miR-139-5p/SDC4 axis. Cancer Cell Int. 21:2842021. View Article : Google Scholar : PubMed/NCBI | |
Chen K, Wang J, Yang M, Deng S and Sun L: Immunotherapy in recurrent ovarian cancer. Biomedicines. 13:1682025. View Article : Google Scholar : PubMed/NCBI | |
Turner T, Buchsbaum D, Straughn J, Randall T and Arend R: Ovarian cancer and the immune system-The role of targeted therapies. Gynecol Oncol. 142:349–356. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Zhang C, Yue D and Dong J: Tumor specific TGF-β insensitive CD8 + T cells augments the antitumor effect through inhibition of epithelial-mesenchymal transition in CD 105 + renal carcinoma stem cells. 2024. View Article : Google Scholar | |
Han Y, Liu D and Li L: PD-1/PD-L1 pathway: Current researches in cancer. Am J Cancer Res. 10:727–742. 2020.PubMed/NCBI | |
Zhu J, Yan L and Wang Q: Efficacy of PD-1/PD-L1 inhibitors in ovarian cancer: A single-arm meta-analysis. J Ovarian Res. 14:1122021. View Article : Google Scholar : PubMed/NCBI | |
Conway J, Kofman E, Mo S, Elmarakeby H and Van Allen E: Genomics of response to immune checkpoint therapies for cancer: Implications for precision medicine. Genome Med. 10:932018. View Article : Google Scholar : PubMed/NCBI | |
Hargadon KM, Johnson CE and Williams CJ: Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int Immunopharmacol. 62:29–39. 2018. View Article : Google Scholar : PubMed/NCBI | |
Brahmer J, Tykodi S, Chow L, Hwu WJ, Topalian S, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al: Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 366:2455–2465. 2012. View Article : Google Scholar : PubMed/NCBI | |
Inayama Y, Hamanishi J, Matsumura N, Murakami R, Abiko K, Yamaguchi K, Baba T, Horie K, Konishi I and Mandai M: Antitumor effect of nivolumab on subsequent chemotherapy for platinum-resistant ovarian cancer. Oncologist. 23:1382–1384. 2018. View Article : Google Scholar : PubMed/NCBI | |
Varga A, Piha-Paul S, Ott P, Mehnert J, Berton-Rigaud D, Morosky A, Zhao GQ, Rangwala RA and Matei D: Pembrolizumab in patients (pts) with PD-L1-positive (PD-L1 +) advanced ovarian cancer: Updated analysis of KEYNOTE-028. J Clin Oncol. 35:55132017. View Article : Google Scholar | |
Demircan N, Boussios S, Tasci T and Öztürk MA: Current and future immunotherapy approaches in ovarian cancer. Ann Transl Med. 8:17142020. View Article : Google Scholar : PubMed/NCBI | |
Pawłowska A, Skiba W, Suszczyk D, Kuryło W, Jakubowicz-Gil J, Paduch R and Wertel I: The dual blockade of the TIGIT and PD-1/PD-L1 pathway as a new hope for ovarian cancer patients. Cancers (Basel). 14:57572023. View Article : Google Scholar | |
Chauvin JM and Zarour HM: TIGIT in cancer immunotherapy. J Immunother Cancer. 8:e0009572020. View Article : Google Scholar : PubMed/NCBI | |
Postow MA, Sidlow R and Hellmann MD: Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 378:158–168. 2018. View Article : Google Scholar : PubMed/NCBI | |
Sanmamed MF and Chen L: A paradigm shift in cancer immunotherapy: From enhancement to normalization. Cell. 175:313–326. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liao T, Li L and Wang L: Bevacizumab combined with chemotherapy for ovarian cancer: A protocol for systematic review and meta-analysis. Medicine (Baltimore). 100:e283762021. View Article : Google Scholar : PubMed/NCBI | |
Tischkowitz M, Huang S, Banerjee S, Hague J, Hendricks W, Huntsman D, Lang JD, Orlando KA, Oza AM, Pautier P, et al: Small-cell carcinoma of the ovary, hypercalcemic type-genetics, new treatment targets, and current management guidelines. Clin Cancer Res. 26:3908–3917. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zhu J, Yan L and Wang Q: Efficacy of PD-1/PD-L1 inhibitors in ovarian cancer: A single-arm meta-analysis. J Ovarian Res. 14:1122021. View Article : Google Scholar : PubMed/NCBI | |
Pawłowska A, Rekowska A, Kuryło W, Pańczyszyn A, Kotarski J and Wertel I: Current understanding on why ovarian cancer is resistant to immune checkpoint inhibitors. Int J Mol Sci. 24:108592023. View Article : Google Scholar : PubMed/NCBI | |
Wu H, Gong J and Liu Y: Indoleamine 2, 3-dioxygenase regulation of immune response (Review). Mol Med Rep. 17:4867–4873. 2018.PubMed/NCBI | |
Liu X, Newton RC, Friedman SM and Scherle PA: Indoleamine 2,3-dioxygenase, an emerging target for anti-cancer therapy. Curr Cancer Drug Targets. 9:938–952. 2009. View Article : Google Scholar : PubMed/NCBI | |
Rogala E, Nowicka A, Bednarek W, Barczyński B, Piekarczyk W, Klimek K, Zakrzewski M and Kotarski J: Evaluation of the expression of the immunosuppressive enzyme-indoleamine 2,3-dioxygenase in ovarian cancer tissue. Menopause Review/Przegląd Menopauzalny. 3:223–227. 2013.(In Polish). | |
Safdarian A, Farhangnia P and Rezaei N: Indoleamine 2,3-Dioxygenase (IDO) and cancerous cells. 2023.1–23 | |
Liu JF, Herold C, Luo W, Penson R, Horowitz N, Konstantinopoulos P, Castro C, Curtis J, Matulonis UA, Cannistra S and Dizon DS: 937PD - A phase II trial of combination nivolumab and bevacizumab in recurrent ovarian cancer. Ann Oncol. 29:viii334–viii335. 2018. View Article : Google Scholar | |
Moore K, Bookman M, Sehouli J, Miller A, Anderson C, Scambia G, Myers T, Taskiran C, Robison K, Mäenpää J, et al: Atezolizumab, bevacizumab, and chemotherapy for newly diagnosed stage III or IV ovarian cancer: Placebo-controlled randomized phase III trial (IMagyn050/GOG 3015/ENGOT-OV39). J Clin Oncol. 39:1842–1855. 2021. View Article : Google Scholar : PubMed/NCBI | |
Kurtz JE, Pujade-Lauraine E, Oaknin A, Belin L, Leitner K, Cibula D, Denys H, Rosengarten O, Rodrigues M, de Gregorio N, 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. 41:4768–4778. 2023. View Article : Google Scholar : PubMed/NCBI | |
González-Martín A, Rubio MJ, Heitz F, Christensen RD, Colombo N, Van Gorp T, Romeo M, Ray-Coquard I, Gaba L, Leary A, et al: Atezolizumab combined with platinum and maintenance niraparib for recurrent ovarian cancer with a platinum-free interval >6 months: ENGOT-OV41/GEICO 69-O/ANITA phase III trial. J Clin Oncol. 42:4294–4304. 2024. View Article : Google Scholar : PubMed/NCBI | |
Okamoto A, Kim JW, Yin R, Trillsch F, Reuss A, Aghajanian C, Rubio-Pérez MJ, Vardar MA, Scambia G, Floquet A, et al: A randomized Phase III trial of durvalumab with chemotherapy and bevacizumab, followed by maintenance durvalumab, bevacizumab and olaparib in newly diagnosed advanced ovarian cancer (DUO-O): Updated trial endpoint and inclusion of China cohort (329). Gynecol Oncol. 166 (Suppl 1):S1702022. View Article : Google Scholar | |
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 | |
Harter P, Mouret-Reynier MA, Pignata S, Cropet C, González-Martín A, Bogner G, Fujiwara K, Vergote I, Colombo N, Nøttrup TJ, et al: Efficacy of 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. Gynecol Oncol. 164:254–264. 2022. View Article : Google Scholar : PubMed/NCBI | |
Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Sevelda P, Fujiwara K, Vergote IB, Colombo N, Maenpaa J, et al: LBA2_PRPhase III PAOLA-1/ENGOT-ov25 trial: Olaparib plus bevacizumab (bev) as maintenance therapy in patients (pts) with newly diagnosed, advanced ovarian cancer (OC) treated with platinum-based chemotherapy (PCh) plus bev. Ann Oncol. 30:2019. View Article : Google Scholar | |
Hardesty MM, Krivak TC, Wright GS, Hamilton E, Fleming EL, Belotte J, Keeton EK, Wang P, Gupta D, Clements A, et al: OVARIO phase II trial of combination niraparib plus bevacizumab maintenance therapy in advanced ovarian cancer following first-line platinum-based chemotherapy with bevacizumab. Gynecol Oncol. 166:219–229. 2022. View Article : Google Scholar : PubMed/NCBI | |
Liu JF, Gaillard S, Hendrickson AE, Yeku O, Diver E, Jackson CG, Arend R, Ratner E, Samnotra V, Gupta D, et al: Niraparib, dostarlimab, and bevacizumab as combination therapy in pretreated, advanced platinum-resistant ovarian cancer: Findings from cohort A of the OPAL phase II trial. JCO Precis Oncol. 8:e23006932024. View Article : Google Scholar : PubMed/NCBI | |
Bukowska B, Gajek A and Marczak A: Two drugs are better than one. A short history of combined therapy of ovarian cancer. Contemp Oncol (Pozn). 5:350–353. 2015.PubMed/NCBI | |
Niculescu AG and Grumezescu AM: Novel tumor-targeting nanoparticles for cancer treatment-a review. Int J Mol Sci. 23:52532022. View Article : Google Scholar : PubMed/NCBI | |
Jani RK and Krupa G: Active targeting of nanoparticles: An innovative technology for drug delivery in cancer therapeutics. J Drug Deliv Ther. 9:408–415. 2019. View Article : Google Scholar | |
Wakaskar RR: Passive and active targeting in tumor microenvironment. Int J Drug Dev Res. 9:22017. | |
Yallapu MM, Jaggi M and Chauhan SC: Scope of nanotechnology in ovarian cancer therapeutics. J Ovarian Res. 3:192010. View Article : Google Scholar : PubMed/NCBI | |
Liu H, Zhu X, Wei Y, Song C and Wang Y: Recent advances in targeted gene silencing and cancer therapy by nanoparticle-based delivery systems. Biomed Pharmacother. 157:1140652023. View Article : Google Scholar : PubMed/NCBI | |
Blanco E, Shen H and Ferrari M: Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 33:941–951. 2015. View Article : Google Scholar : PubMed/NCBI | |
Guo X, Guo N, Zhao J and Cai Y: Active targeting co-delivery system based on hollow mesoporous silica nanoparticles for antitumor therapy in ovarian cancer stem-like cells. Oncol Rep. 38:1442–1450. 2017. View Article : Google Scholar : PubMed/NCBI | |
Madej M, Kurowska N and Strzalka-Mrozik B: Polymeric nanoparticles-tools in a drug delivery system in selected cancer therapies. Appl Sci. 12:94792022. View Article : Google Scholar | |
Hascicek C and Gun O: Nano drug delivery systems for ovarian cancer therapy. Integr Cancer Sci Ther. 4:1–4. 2017.PubMed/NCBI | |
Saripilli R and Sharma DK: Nanotechnology-based drug delivery system for the diagnosis and treatment of ovarian cancer. Discov Oncol. 16:4222025. View Article : Google Scholar : PubMed/NCBI | |
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 | |
Engelberth SA, Hempel N and Bergkvist M: Development of nanoscale approaches for ovarian cancer therapeutics and diagnostics. Crit Rev Oncog. 19:281–315. 2014. View Article : Google Scholar : PubMed/NCBI | |
Zhao J, Tan W, Zheng J, Su Y and Cui M: Aptamer nanomaterials for ovarian cancer target theranostics. Front Bioeng Biotechnol. 10:8844052022. View Article : Google Scholar : PubMed/NCBI | |
Di Lorenzo G, Ricci G, Severini GM, Romano F and Biffi S: Imaging and therapy of ovarian cancer: clinical application of nanoparticles and future perspectives. Theranostics. 8:4279–4294. 2018. View Article : Google Scholar : PubMed/NCBI | |
Search for: Ovarian Cancer, Other terms: Nanoparticles, Completed studies | Card Results | ClinicalTrials.gov [Internet]. [cited 2024 Jun 29]. Available from:, . https://clinicaltrials.gov/search?cond=Ovarian%20Cancer&term=Nanoparticles&aggFilters=status:com&page=2 | |
Gavas S, Quazi S and Karpiński TM: Nanoparticles for cancer therapy: Current progress and challenges. Nanoscale Res Lett. 16:1732021. View Article : Google Scholar : PubMed/NCBI | |
Kumah EA, Fopa RD, Harati S, Boadu P, Zohoori FV and Pak T: Human and environmental impacts of nanoparticles: A scoping review of the current literature. BMC Public Health. 23:10592023. View Article : Google Scholar : PubMed/NCBI | |
Pandey RP, Vidic J, Mukherjee R and Chang CM: Experimental methods for the biological evaluation of nanoparticle-based drug delivery risks. Pharmaceutics. 15:6122023. View Article : Google Scholar : PubMed/NCBI | |
Zhang J, Ding H, Zhang F, Xu Y, Liang W and Huang L: New trends in diagnosing and treating ovarian cancer using nanotechnology. Front Bioeng Biotechnol. 11:11609852023. View Article : Google Scholar : PubMed/NCBI | |
Wilson MK, Pujade-Lauraine E, Aoki D, Mirza MR, Lorusso D, Oza AM, du Bois A, Vergote I, Reuss A, Bacon M, et al: Fifth ovarian cancer consensus conference of the gynecologic cancer intergroup: Recurrent disease. Ann Oncol. 28:727–732. 2017. View Article : Google Scholar : PubMed/NCBI | |
Daly MB, Pal T, Berry MP, Buys SS, Dickson P, Domchek SM, Elkhanany A, Friedman S, Goggins M, Hutton ML, et al: Genetic/Familial high-risk assessment: Breast, ovarian, and pancreatic, Version 2.2021, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 19:77–102. 2021. View Article : Google Scholar : PubMed/NCBI | |
Konstantinopoulos PA, Norquist B, Lacchetti C, Armstrong D, Grisham RN, Goodfellow PJ, Kohn EC, Levine DA, Liu JF, Lu KH, et al: Germline and somatic tumor testing in epithelial ovarian cancer: ASCO guideline. J Clin Oncol. 38:1222–1245. 2020. View Article : Google Scholar : PubMed/NCBI | |
Zelli V, Compagnoni C, Cannita K, Capelli R, Capalbo C, Di Vito Nolfi M, Alesse E, Zazzeroni F and Tessitore A: Applications of next generation sequencing to the analysis of familial breast/ovarian cancer. High Throughput. 9:12020. View Article : Google Scholar : PubMed/NCBI | |
Richards S, Aziz N, Bale S, Bick D, Das S, Gastier-Foster J, Grody WW, Hegde M, Lyon E, Spector E, et al: Standards and guidelines for the interpretation of sequence variants: A joint consensus recommendation of the American College of medical genetics and genomics and the association for molecular pathology. Genet Med. 17:405–424. 2015. View Article : Google Scholar : PubMed/NCBI | |
Das SK, Menezes ME, Bhatia S, Wang X, Emdad L, Sarkar D and Fisher PB: Gene therapies for cancer: Strategies, challenges and successes. J Cell Physiol. 230:259–271. 2015. View Article : Google Scholar : PubMed/NCBI | |
Áyen Á, Martínez YJ, Marchal JA and Boulaiz H: Recent progress in gene therapy for ovarian cancer. Int J Mol Sci. 19:19302018. View Article : Google Scholar : PubMed/NCBI | |
Li S, Jiang K, Li J, Hao X, Chu W, Luo C, Zhu Y, Xie R and Chen B: Estrogen enhances the proliferation and migration of ovarian cancer cells by activating transient receptor potential channel C3. J Ovarian Res. 13:202020. View Article : Google Scholar : PubMed/NCBI | |
Cunat S, Hoffmann P and Pujol P: Estrogens and epithelial ovarian cancer. Gynecol Oncol. 94:25–32. 2004. View Article : Google Scholar : PubMed/NCBI | |
Kozieł MJ and Piastowska-Ciesielska AW: Estrogens, estrogen receptors and tumor microenvironment in ovarian cancer. Int J Mol Sci. 24:146732023. View Article : Google Scholar : PubMed/NCBI | |
Zhang M, Xu H, Zhang Y, Li Z, Meng W, Xia J, Le W, Meng K and Guo Y: Research progress of estrogen receptor in ovarian cancer. Clin Exp Obstet Gynecol. 50:1992023. View Article : Google Scholar | |
Jeon SY, Hwang KA and Choi KC: Effect of steroid hormones, estrogen and progesterone, on epithelial mesenchymal transition in ovarian cancer development. J Steroid Biochem Mol Biol. 158:1–8. 2016. View Article : Google Scholar : PubMed/NCBI | |
Park SH, Cheung LWT, Wong AST and Leung PCK: Estrogen regulates snail and slug in the down-regulation of e-cadherin and induces metastatic potential of ovarian cancer cells through estrogen receptor alpha. Mol Endocrinol. 22:2085–2098. 2008. View Article : Google Scholar : PubMed/NCBI | |
Smyth JF, Gourley C, Walker G, MacKean MJ, Stevenson A, Williams ARW, Nafussi AA, Rye T, Rye R, Stewart M, et al: Antiestrogen therapy is active in selected ovarian cancer cases: the use of letrozole in estrogen receptor-positive patients. Clin Cancer Res. 13:3617–3622. 2007. View Article : Google Scholar : PubMed/NCBI | |
Gershenson DM, Cobb LP and Sun CC: Endocrine therapy in the management of low-grade serous ovarian/peritoneal carcinoma: Mounting evidence for therelative efficacy of tamoxifen and aromatase inhibitors. Gynecol Oncol. 159:601–603. 2020. View Article : Google Scholar : PubMed/NCBI | |
George A, McLachlan J, Tunariu N, Pepa CD, Migali C, Gore M, Kaye S and Banerjee S: The role of hormonal therapy in patients with relapsed high-grade ovarian carcinoma: A retrospective series of tamoxifen and letrozole. BMC Cancer. 17:4562017. View Article : Google Scholar : PubMed/NCBI | |
Colombo N, Sessa C, du Bois A, Ledermann J, McCluggage WG, McNeish I, Morice P, Pignata S, Ray-Coquard I, Vergote I, et al: ESMO-ESGO consensus conference recommendations on ovarian cancer: Pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann Oncol. 30:672–705. 2019. View Article : Google Scholar : PubMed/NCBI | |
National Comprehensive Cancer Network (NCCN), . Guidelines Detail. NCCN; Plymouth Meeting, PA: 2025, https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1455May 3–2025 | |
Lopresti ML, Bandera CA and Miner TJ: New approaches to improving survival after neoadjuvant chemotherapy: The role of intraperitoneal therapy and heated intraperitoneal chemotherapy in ovarian cancer. Am Soc Clin Oncol Educ Book. 39:19–23. 2019. View Article : Google Scholar : PubMed/NCBI | |
Murphy M, Martin G, Mahmoudjafari Z, Bivona C, Grauer D and Henry D: Intraperitoneal paclitaxel and cisplatin compared with dose-dense paclitaxel and carboplatin for patients with stage III ovarian cancer. J Oncol Pharm Pract. 26:1566–1574. 2020. View Article : Google Scholar : PubMed/NCBI | |
Cascales-Campos P, López-López V, Gil J, Arévalo-Pérez J, Nieto A, Barceló F, Gil E and Parrilla P: Hyperthermic intraperitoneal chemotherapy with paclitaxel or cisplatin in patients with stage III-C/IV ovarian cancer. Is there any difference? Surg Oncol. 25:164–170. 2016.PubMed/NCBI | |
Sjoquist KM, Espinoza D, Mileshkin L, Ananda S, Shannon C, Yip S, Goh J, Bowtell D, Harrison M and Friedlander ML: REZOLVE (ANZGOG-1101): A phase 2 trial of intraperitoneal bevacizumab to treat symptomatic ascites in patients with chemotherapy-resistant, epithelial ovarian cancer. Gynecol Oncol. 161:374–381. 2021. View Article : Google Scholar : PubMed/NCBI | |
Virdi S and Jadavji NM: The impact of maternal folates on brain development and function after birth. Metabolites. 12:8762022. View Article : Google Scholar : PubMed/NCBI | |
Gonzalez T, Muminovic M, Nano O and Vulfovich M: Folate receptor alpha-a novel approach to cancer therapy. Int J Mol Sci. 25:10462024. View Article : Google Scholar : PubMed/NCBI | |
Elnakat H and Ratnam M: Distribution, functionality and gene regulation of folate receptor isoforms: Implications in targeted therapy. Adv Drug Deliv Rev. 56:1067–1084. 2004. View Article : Google Scholar : PubMed/NCBI | |
Mai J, Wu L, Yang L, Sun T, Liu X, Yin R, Jiang Y, Li J and Li Q: Therapeutic strategies targeting folate receptor α for ovarian cancer. Front Immunol. 14:12545322023. View Article : Google Scholar : PubMed/NCBI | |
Kurosaki A, Hasegawa K, Kato T, Abe K, Hanaoka T, Miyara A, O'Shannessy DJ, Somers EB, Yasuda M, Sekino T and Fujiwara K: Serum folate receptor alpha as a biomarker for ovarian cancer: Implications for diagnosis, prognosis and predicting its local tumor expression. Int J Cancer. 138:1994–2002. 2016. View Article : Google Scholar : PubMed/NCBI | |
Moore KN, Martin LP, O'Malley DM, Matulonis UA, Konner JA, Vergote I, Ponte JF and Birrer MJ: A review of mirvetuximab soravtansine in the treatment of platinum-resistant ovarian cancer. Future Oncol. 14:123–136. 2018. View Article : Google Scholar : PubMed/NCBI | |
Li X, Zhou S, Abrahams CL, Krimm S, Smith J, Bajjuri K, Stephenson HT, Henningsen R, Hanson J, Heibeck TH, et al: Discovery of STRO-002, a novel homogeneous ADC targeting folate receptor alpha, for the treatment of ovarian and endometrial cancers. Mol Cancer Ther. 22:155–167. 2023. View Article : Google Scholar : PubMed/NCBI | |
Matulonis UA, Oaknin A, Pignata S, Denys H, Colombo N, Van Gorp T, Konner JA, Romeo M, Harter P, Murphy CG, et al: Mirvetuximab soravtansine (MIRV) in patients with platinum-resistant ovarian cancer with high folate receptor alpha (FRα) expression: Characterization of antitumor activity in the SORAYA study. J Clin Oncol. 40 (16_suppl):S55122022. View Article : Google Scholar | |
Jelovac D and Armstrong DK: Role of farletuzumab in epithelial ovarian carcinoma. Curr Pharm Des. 18:3812–3815. 2012. View Article : Google Scholar : PubMed/NCBI | |
Fayoud AM, Darwish MY, Nada EA, Helal AA, Mohamed NS, Elrashedy AA and Abd-ElGawad M: Efficacy and safety of farletuzumab in ovarian cancer: A systematic review and single-arm meta-analysis. Cureus. 16:e735032024.PubMed/NCBI | |
Abrahams C, Krimm S, Li X, Zhou S, Hanson J, Masikat MR, Bajjuri K, Heibeck T, Kothari D, Yu A, et al: Abstract NT-090: Preclinical activity and safety of STRO-002, a novel adc targeting folate receptor alpha for ovarian and endometrial cancer. Clin Cancer Res. 25 (22_Suppl):NT–090. 2019. View Article : Google Scholar | |
Naumann RW, Braiteh FS, Martin LP, Hamilton EP, Diaz JP, Diab S, Schilder RJ, Moroney JW, Uyar D, O'Malley DM, et al: Phase 1 dose-escalation study of STRO-002, an antifolate receptor alpha (FRα) antibody drug conjugate (ADC), in patients with advanced, progressive platinum-resistant/refractory epithelial ovarian cancer (EOC). J Clin Oncol. 39 (15_suppl):S55502021. View Article : Google Scholar | |
Ashburn TT and Thor KB: Drug repositioning: identifying and developing new uses for existing drugs. Nat Rev Drug Discov. 3:673–683. 2004. View Article : Google Scholar : PubMed/NCBI | |
Kim JH, Park WH, Suh DH, Kim K, No JH and Kim YB: Calcitriol combined with platinum-based chemotherapy suppresses growth and expression of vascular endothelial growth factor of SKOV-3 ovarian cancer cells. Anticancer Res. 41:2945–2952. 2021. View Article : Google Scholar : PubMed/NCBI | |
Srivastava AK, Rizvi A, Cui T, Han C, Banerjee A, Naseem I, Zheng Y, Wani AA and Wang QE: Depleting ovarian cancer stem cells with calcitriol. Oncotarget. 9:14481–14491. 2018. View Article : Google Scholar : PubMed/NCBI | |
Kuittinen T, Rovio P, Luukkaala T, Laurila M, Grénman S, Kallioniemi A and Mäenpää J: Paclitaxel, carboplatin and 1,25-D3 inhibit proliferation of ovarian cancer cells in vitro. Anticancer Res. 40:3129–313. 2020. View Article : Google Scholar : PubMed/NCBI | |
Wang L, Zhou S and Guo B: Vitamin D suppresses ovarian cancer growth and invasion by targeting long non-coding RNA CCAT2. Int J Mol Sci. 21:23342020. View Article : Google Scholar : PubMed/NCBI | |
Dovnik A and Dovnik NF: Vitamin D and ovarian cancer: Systematic review of the literature with a focus on molecular mechanisms. Cells. 9:3352020. View Article : Google Scholar : PubMed/NCBI | |
Duffy MJ, Murray A, Synnott NC, O'Donovan N and Crown J: Vitamin D analogues: Potential use in cancer treatment. Crit Rev Oncol Hematol. 112:190–197. 2017. View Article : Google Scholar : PubMed/NCBI | |
Piatek K, Schepelmann M and Kallay E: The effect of vitamin D and its analogs in ovarian cancer. Nutrients. 14:38672022. View Article : Google Scholar : PubMed/NCBI | |
Rizvi A and Naseem I: Causing DNA damage and stopping DNA repair-Vitamin D supplementation with Poly(ADP-ribose) polymerase 1 (PARP1) inhibitors may cause selective cell death of cancer cells: A novel therapeutic paradigm utilizing elevated copper levels within the tumour. Med Hypotheses. 144:1102782020. View Article : Google Scholar : PubMed/NCBI | |
Ghaseminejad-Raeini A, Ghaderi A, Sharafi A, Nematollahi-Sani B, Moossavi M, Derakhshani A and Sarab GA: Immunomodulatory actions of vitamin D in various immune-related disorders: A comprehensive review. Front Immunol. 14:9504652023. View Article : Google Scholar : PubMed/NCBI | |
Munteanu C, Mârza SM and Papuc I: The immunomodulatory effects of vitamins in cancer. Front Immunol. 15:14643292024. View Article : Google Scholar : PubMed/NCBI | |
Glassman D, Bateman NW, Lee S, Zhao L, Yao J, Tan Y, Ivan C, Rangel KM, Zhang J, Conrads KA, et al: Molecular correlates of venous thromboembolism (VTE) in ovarian cancer. Cancers (Basel). 14:14962022. View Article : Google Scholar : PubMed/NCBI | |
Skorda A, Bay ML, Hautaniemi S, Lahtinen A and Kallunki T: Kinase inhibitors in the treatment of ovarian cancer: Current state and future promises. Cancers (Basel). 14:62572022. View Article : Google Scholar : PubMed/NCBI | |
Kasthuri RS, Taubman MB and Mackman N: Role of tissue factor in cancer. J Clin Oncol. 27:4834–4838. 2009. View Article : Google Scholar : PubMed/NCBI | |
Versteeg HH, Spek CA, Peppelenbosch MP and Richel DJ: Tissue factor and cancer metastasis: The role of intracellular and extracellular signaling pathways. Mol Med. 10:6–11. 2004. View Article : Google Scholar : PubMed/NCBI | |
Ruf W, Yokota N and Schaffner F: Tissue factor in cancer progression and angiogenesis. Thromb Res. 125:S36–S38. 2010. View Article : Google Scholar : PubMed/NCBI | |
Coleman RL, Lorusso D, Gennigens C, González-Martín A, Randall L, Cibula D, Lund B, Woelber L, Pignata S, Forget F, et al: Efficacy and safety of tisotumab vedotin in previously treated recurrent or metastatic cervical cancer (innovaTV 204/GOG-3023/ENGOT-cx6): A multicentre, open-label, single-arm, phase 2 study. Lancet Oncol. 22:609–619. 2021. View Article : Google Scholar : PubMed/NCBI | |
de Bono JS, Concin N, Hong DS, Thistlethwaite FC, Machiels JP, Arkenau HT, Plummer R, Jones RH, Nielsen D, Windfeld K, et al: Tisotumab vedotin in patients with advanced or metastatic solid tumours (InnovaTV 201): A first-in-human, multicentre, phase 1–2 trial. Lancet Oncol. 20:383–393. 2019. View Article : Google Scholar : PubMed/NCBI | |
Arter ZL, Desmond D, Berenberg JL, Killeen JL, Bunch K and Merritt MA: Epithelial ovarian cancer survival by race and ethnicity in an equal-access healthcare population. Br J Cancer. 130:108–113. 2024. View Article : Google Scholar : PubMed/NCBI | |
Wagar MK, Mojdehbakhsh RP, Godecker A, Rice LW and Barroilhet L: Racial and ethnic enrollment disparities in clinical trials of poly(ADP-ribose) polymerase inhibitors for gynecologic cancers. Gynecol Oncol. 165:49–52. 2022. View Article : Google Scholar : PubMed/NCBI | |
Fabbro M, Moore KN, Dørum A, Tinker AV, Mahner S, Bover I, Banerjee S, Tognon G, Goffin F, Shapira-Frommer R, et al: Efficacy and safety of niraparib as maintenance treatment in older patients (≥70 years) with recurrent ovarian cancer: Results from the ENGOT-OV16/NOVA trial. Gynecol Oncol. 152:560–567. 2019. View Article : Google Scholar : PubMed/NCBI | |
Selle F, Colombo N, Korach J, Mendiola C, Cardona A, Ghazi Y and Oza AM: Safety and efficacy of extended bevacizumab therapy in elderly (≥70 Years) versus younger patients treated for newly diagnosed ovarian cancer in the international ROSiA study. Int J Gynecol Cancer. 28:729–737. 2018. View Article : Google Scholar : PubMed/NCBI | |
Freyer G, Tew WP and Moore KN: Treatment and trials: Ovarian cancer in older women. Am Soc Clin Oncol Educ Book. pp227–235. 2013. View Article : Google Scholar : PubMed/NCBI | |
Coleman RL, Fleming GF, Brady MF, Swisher EM, Steffensen KD, Friedlander M, Okamoto A, Moore KN, Efrat Ben-Baruch N, Werner TL, et al: Veliparib with first-line chemotherapy and as maintenance therapy in ovarian cancer. N Engl J Med. 381:2403–2415. 2019. View Article : Google Scholar : PubMed/NCBI | |
González-Martín A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, McCormick C, Lorusso D, Hoskins P, Freyer G, et al: Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 381:2391–2402. 2019. View Article : Google Scholar : PubMed/NCBI | |
Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, Sun J, Konecny GE, Coleman RL, Tinker AV, O'Malley DM, et al: Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): An international, multicentre, open-label, phase 2 trial. Lancet Oncol. 18:75–87. 2017. View Article : Google Scholar : PubMed/NCBI | |
Sha D, Jin Z, Budczies J, Kluck K, Stenzinger A and Sinicrope FA: Tumor mutational burden as a predictive biomarker in solid tumors. Cancer Discov. 10:1808–1825. 2020. View Article : Google Scholar : PubMed/NCBI | |
Fan S, Gao X, Qin Q, Li H, Yuan Z and Zhao S: Association between tumor mutation burden and immune infiltration in ovarian cancer. Int Immunopharmacol. 89:1071262020. View Article : Google Scholar : PubMed/NCBI | |
Matulonis UA, Shapira-Frommer R, Santin AD, Lisyanskaya AS, Pignata S, Vergote I, Raspagliesi F, Sonke GS, Birrer M, Provencher DM, 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. 30:1080–1087. 2019. View Article : Google Scholar : PubMed/NCBI | |
Lee M, Samstein RM, Valero C, Chan TA and Morris LGT: Tumor mutational burden as a predictive biomarker for checkpoint inhibitor immunotherapy. Hum Vaccines Immunother. 16:112–115. 2020. View Article : Google Scholar : PubMed/NCBI | |
Klempner SJ, Fabrizio D, Bane S, Reinhart M, Peoples T, Ali SM, Sokol ES, Frampton G, Schrock AB, Anhorn R and Reddy P: Tumor mutational burden as a predictive biomarker for response to immune checkpoint inhibitors: A review of current evidence. Oncologist. 25:e147–e159. 2020. View Article : Google Scholar : PubMed/NCBI | |
Lee CY, Cheng WF, Lin PH, Chen YL, Huang SH, Lei KH, Chang KY, Ko MY and Chi P: An activity-based functional test for identifying homologous recombination deficiencies across cancer types in real time. Cell Rep Med. 4:1012472023. View Article : Google Scholar : PubMed/NCBI | |
Korsholm LM, Kjeldsen M, Perino L, Mariani L, Nyvang GB, Kristensen E, Bagger FO, Mirza MR and Rossing M: Combining homologous recombination-deficient testing and functional RAD51 analysis enhances the prediction of Poly(ADP-Ribose) polymerase inhibitor sensitivity. JCO Precis Oncol. 8:e23004832024. View Article : Google Scholar : PubMed/NCBI | |
Pettitt SJ, Krastev DB, Brandsma I, Dréan A, Song F, Aleksandrov R, Harrell MI, Menon M, Brough R, Campbell J, et al: Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance. Nat Commun. 9:18492018. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Liu T, Zhang Z, Payne SH, Zhang B, McDermott JE, Zhou JY, Petyuk VA, Chen L, Ray D, et al: Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 166:755–765. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tannock IF and Hickman JA: Limits to personalized cancer medicine. N Engl J Med. 375:1289–1294. 2016. View Article : Google Scholar : PubMed/NCBI | |
Wan JCM, Massie C, Garcia-Corbacho J, Mouliere F, Brenton JD, Caldas C, Pacey S, Baird R and Rosenfeld N: Liquid biopsies come of age: Towards implementation of circulating tumour DNA. Nat Rev. 17:223–238. 2017. View Article : Google Scholar : PubMed/NCBI | |
Mateo J, Lord CJ, Serra V, Tutt A, Balmaña J, Castroviejo-Bermejo M, Cruz C, Oaknin A, Kaye SB and de Bono JS: A decade of clinical development of PARP inhibitors in perspective. Ann Oncol. 30:1437–1447. 2019. View Article : Google Scholar : PubMed/NCBI | |
McAlpine JN, Porter H, Köbel M, Nelson BH, Prentice LM, Kalloger SE, Senz J, Milne K, Ding J, Shah SP, et al: BRCA1 and BRCA2 mutations correlate with TP53 abnormalities and presence of immune cell infiltrates in ovarian high-grade serous carcinoma. Mod Pathol. 25:740–750. 2012. View Article : Google Scholar : PubMed/NCBI | |
Scully R and Livingston DM: In search of the tumour-suppressor functions of BRCA1 and BRCA2. Nature. 408:429–432. 2000. View Article : Google Scholar : PubMed/NCBI | |
Venkitaraman AR: Cancer suppression by the chromosome custodians, BRCA1 and BRCA2. Science. 343:1470–1475. 2014. View Article : Google Scholar : PubMed/NCBI | |
Davies AA, Masson JY, McIlwraith MJ, Stasiak AZ, Stasiak A, Venkitaraman AR and West SC: Role of BRCA2 in control of the RAD51 recombination and DNA repair protein. Mol Cell. 7:273–282. 2001. View Article : Google Scholar : PubMed/NCBI | |
Genome Atlas Research Network, . Integrated genomic analyses of ovarian carcinoma. Nature. 474:609–615. 2011. View Article : Google Scholar : PubMed/NCBI | |
Walsh T, Casadei S, Lee MK, Pennil CC, Nord AS, Thornton AM, Roeb W, Agnew KJ, Stray SM, Wickramanayake A, et al: Mutations in 12 genes for inherited ovarian, fallopian tube, and peritoneal carcinoma identified by massively parallel sequencing. Proc Natl Acad Sci USA. 108:18032–18037. 2011. View Article : Google Scholar : PubMed/NCBI | |
Kandoth C, McLellan MD, Vandin F, Ye K, Niu B, Lu C, Xie M, Zhang Q, McMichael JF, Wyczalkowski MA, et al: Mutational landscape and significance across 12 major cancer types. Nature. 502:333–339. 2013. View Article : Google Scholar : PubMed/NCBI | |
Levine AJ: p53, the cellular gatekeeper for growth and division. Cell. 88:323–331. 1997. View Article : Google Scholar : PubMed/NCBI | |
Hoadley KA, Yau C, Wolf DM, Cherniack AD, Tamborero D, Ng S, Leiserson MDM, Niu B, McLellan MD, Uzunangelov V, et al: Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell. 158:929–944. 2014. View Article : Google Scholar : PubMed/NCBI | |
Haupt Y, Maya R, Kazaz A and Oren M: Mdm2 promotes the rapid degradation of p53. Nature. 387:296–299. 1997. View Article : Google Scholar : PubMed/NCBI | |
Vaz F, Hanenberg H, Schuster B, Barker K, Wiek C, Erven V, Neveling K, Endt D, Kesterton I, Autore F, et al: Mutation of the RAD51C gene in a Fanconi anemia-like disorder. Nat Genet. 42:406–409. 2010. View Article : Google Scholar : PubMed/NCBI | |
Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, Karlan BY, Taniguchi T and Swisher EM: Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J Clin Oncol. 29:3008–3015. 2011. View Article : Google Scholar : PubMed/NCBI | |
Guan B, Mao TL, Panuganti PK, Kuhn E, Kurman RJ, Maeda D, Chen E, Jeng YM, Wang TL and Shih IM: Mutation and loss of expression of ARID1A in uterine low-grade endometrioid carcinoma. Am J Surg Pathol. 35:625–632. 2011. View Article : Google Scholar : PubMed/NCBI | |
Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, et al: ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med. 363:1532–1543. 2010. View Article : Google Scholar : PubMed/NCBI | |
Samartzis EP, Gutsche K, Dedes KJ, Fink D, Stucki M and Imesch P: Loss of ARID1A expression sensitizes cancer cells to PI3K- and AKT-inhibition. Oncotarget. 5:5295–5303. 2014. View Article : Google Scholar : PubMed/NCBI | |
Li J, Yen C, Liaw D, Podsypanina K, Bose S, Wang SI, Puc J, Miliaresis C, Rodgers L, McCombie R, et al: PTEN, a putative protein tyrosine phosphatase gene mutated in human brain, breast, and prostate cancer. Science. 275:1943–1947. 1997. View Article : Google Scholar : PubMed/NCBI | |
Song MS, Salmena L and Pandolfi PP: The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 13:283–296. 2012. View Article : Google Scholar : PubMed/NCBI | |
Obata K, Morland SJ, Watson RH, Hitchcock A, Chenevix-Trench G, Thomas EJ and Campbell IG: Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors. Cancer Res. 58:2095–2097. 1998.PubMed/NCBI | |
Bos JL: Ras oncogenes in human cancer: A review. Cancer Res. 49:4682–4689. 1989.PubMed/NCBI | |
Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, et al: Mutations of the BRAF gene in human cancer. Nature. 417:949–954. 2002. View Article : Google Scholar : PubMed/NCBI | |
Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 3:11–22. 2003. View Article : Google Scholar : PubMed/NCBI | |
Garnett MJ and Marais R: Guilty as charged: B-RAF is a human oncogene. Cancer Cell. 6:313–319. 2004. View Article : Google Scholar : PubMed/NCBI | |
Singer G, Oldt R, Cohen Y, Wang BG, Sidransky D, Kurman RJ and Shih IM: Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma. JNCI. J Natl Cancer Inst. 95:484–486. 2003. View Article : Google Scholar : PubMed/NCBI | |
Kamb A, Gruis NA, Weaver-Feldhaus J, Liu Q, Harshman K, Tavtigian SV, Stockert E, Day RS III, Johnson BE and Skolnick MH: A cell cycle regulator potentially involved in genesis of many tumor types. Science. 264:436–440. 1994. View Article : Google Scholar : PubMed/NCBI | |
Sherr CJ: The INK4a/ARF network in tumour suppression. Nat Rev Mol Cell Biol. 2:731–737. 2001. View Article : Google Scholar : PubMed/NCBI | |
Kim WY and Sharpless NE: The regulation of INK4/ARF in cancer and aging. Cell. 127:265–275. 2006. View Article : Google Scholar : PubMed/NCBI | |
Pomerantz J, Schreiber-Agus N, Liégeois NJ, Silverman A, Alland L, Chin L, Potes J, Chen K, Orlow I, Lee HW, et al: The Ink4a tumor suppressor gene product, p19Arf, interacts with MDM2 and neutralizes MDM2's inhibition of p53. Cell. 92:713–723. 2025. View Article : Google Scholar : PubMed/NCBI | |
Zhao R, Choi BY, Lee MH, Bode AM and Dong Z: Implications of genetic and epigenetic alterations of CDKN2A (p16(INK4a)) in cancer. EBioMedicine. 8:30–39. 2016. View Article : Google Scholar : PubMed/NCBI | |
Reed AL, Califano J, Cairns P, Westra WH, Jones RM, Koch W, Ahrendt S, Eby Y, Sewell D, Nawroz H, et al: High frequency of p16 (CDKN2/MTS-1/INK4A) inactivation in head and neck squamous cell carcinoma. Cancer Res. 56:3630–3633. 1996.PubMed/NCBI | |
Cawthon RM, Weiss R, Xu GF, Viskochil D, Culver M, Stevens J, Robertson M, Dunn D, Gesteland R and O'Connell P: A major segment of the neurofibromatosis type 1 gene: cDNA sequence, genomic structure, and point mutations. Cell. 62:193–201. 1990. View Article : Google Scholar : PubMed/NCBI | |
Xu G, O'Connell P, Viskochil D, Cawthon R, Robertson M, Culver M, Dunn D, Stevens J, Gesteland R and White R: The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell. 62:599–608. 1990. View Article : Google Scholar : PubMed/NCBI | |
Dasgupta B and Gutmann DH: Neurofibromatosis 1: Closing the GAP between mice and men. Curr Opin Genet Dev. 13:20–27. 2003. View Article : Google Scholar : PubMed/NCBI | |
Ballester R, Marchuk D, Boguski M, Saulino A, Letcher R, Wigler M and Collins F: The NF1 locus encodes a protein functionally related to mammalian GAP and yeast IRA proteins. Cell. 63:851–859. 1990. View Article : Google Scholar : PubMed/NCBI | |
Cancer Genome Atlas Research Network, ; Kandoth C, Schultz N, Cherniack AD, Akbani R, Liu Y, Shen H, Robertson AG, Pashtan I, Shen R, et al: Integrated genomic characterization of endometrial carcinoma. Nature. 497:67–73. 2013. View Article : Google Scholar : PubMed/NCBI | |
Ratner N and Miller SJ: A RASopathy gene commonly mutated in cancer: The neurofibromatosis type 1 tumour suppressor. Nat Rev Cancer. 15:290–301. 2015. View Article : Google Scholar : PubMed/NCBI | |
Ortiz M, Wabel E, Mitchell K and Horibata S: Mechanisms of chemotherapy resistance in ovarian cancer. Cancer Drug Resist. 5:304–316. 2022.PubMed/NCBI | |
National Library of Medicine (NIH), . ClinicalTrials.gov. https://clinicaltrials.gov/NIH; Bethesda, MD: 2025 March 15–2025 |