Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2025 Apr 19;75(5):436–460. doi: 10.3322/caac.70008

Transforming treatment paradigms: Focus on personalized medicine for high‐grade serous ovarian cancer

Pawel Kordowitzki 1,2,3, Britta Lange 4, Kevin M Elias 5, Marcia C Haigis 2, Sylvia Mechsner 3, Ioana Elena Braicu 3, Jalid Sehouli 3,
PMCID: PMC12432820  PMID: 40252048

Abstract

High‐grade serous ovarian cancer (HGSOC) is the most common and aggressive subtype of ovarian cancer, accounting for approximately 70% of all ovarian cancer cases and contributing significantly to the high mortality rates associated with this disease. Because of the asymptomatic nature of early stage disease, most patients are diagnosed at advanced stages when the cancer has already spread into the abdominal cavity, requiring complex and intensive surgical and chemotherapeutic interventions followed by maintenance therapies. Although a minority of cases are associated with well defined genetic syndromes, specific risk factors and a clear etiology in many cases remain elusive. HGSOC tumors are characterized by a high frequency of somatic gene copy number alterations, often associated with defects in homologous recombination repair of DNA. All attempts to introduce an effective screening for HGSOC to date have been unsuccessful. This review elucidates the complexities surrounding HGSOC and encompasses its etiology, epidemiology, classification, pathogenesis, and the current array of treatment strategies. Understanding molecular underpinnings is crucial for the development of targeted therapies and personalized multimodal treatment approaches in centralized therapeutic structures. This review also examines the importance of the tumor microenvironment. In addition, the authors' objective is to underscore the critical importance of placing the patient's perspective and diversity at the forefront of therapeutic strategies, thereby fostering a genuinely participatory decision‐making process and ultimately improving patient quality of life.

Keywords: diversity, high‐grade serous ovarian cancer, personalized medicine, quality of life, treatment, tumor microenvironment

INTRODUCTION

High‐grade serous ovarian cancer (HGSOC) stands as the most prevalent and deadly subtype among ovarian malignancies, characterized by its aggressive nature and high recurrence rates. HGSOC is one of the most common gynecologic malignancies in women worldwide and, among common gynecologic tumors, carries the highest case lethality. 1 , 2 , 3 According to the American Cancer Society, in 2024, there will be approximately 19,680 new ovarian cancer cases and 12,740 ovarian cancer deaths in the United States. 4 Epithelial ovarian cancer accounts for 90% of cases, with high‐grade serous tumors having the poorest prognosis among the major subtypes. The low survival rates are primarily attributable to diagnoses at more advanced disease stages. 4 Contemporary analyses have provided valuable insights into the molecular drivers and heterogeneity of this disease, paving the way for personalized and multimodal strategies. In addition, we recommend that these approaches be taken from a patient‐oriented perspective, as discussed below in detail. The symptoms and early signs of HGSOC are typically vague and nonspecific, contributing to the challenges in early detection (Figure 1). Common symptoms include abdominal bloating, pelvic pain, early satiety, and urinary urgency or frequency. These symptoms are often mistaken for benign conditions, such as gastrointestinal disorders, leading to diagnostic delays. 5 , 6 As the disease progresses, symptoms become more pronounced and may include weight loss, fatigue, and changes in bowel habits. The insidious onset of symptoms underscores the importance of raising awareness and promoting early diagnostic initiatives to improve outcomes. It is noteworthy that thrombosis, paraneoplastic cerebellar symptoms, and paraneoplastic dermatitis are rare but well described presentations and should not be neglected or misinterpreted.

FIGURE 1.

FIGURE 1

Common presenting symptoms of patients with high‐grade serous ovarian cancer. The symptoms and early signs of high‐grade serous ovarian cancer are typically vague and nonspecific, contributing to the challenges in early detection. Common symptoms include abdominal bloating, pelvic pain, early satiety, and urinary urgency or frequency. These symptoms are often mistaken for benign conditions, such as gastrointestinal disorders, leading to diagnostic delayed diagnosis. As the disease progresses, symptoms become more pronounced and may include weight loss, fatigue, postmenopausal bleeding, and changes in bowel habits. Thrombosis, paraneoplastic cerebellar symptoms, or paraneoplastic dermatitis are rare symptoms. The insidious onset of symptoms underscores the importance of raising awareness and promoting early diagnostic initiatives to improve outcomes. Created with Biorender (License No. EL27ZKGY7G).

This review aims to elucidate the complexities surrounding HGSOC with a focus on its epidemiology, classification and staging, pathogenesis, and the current array of treatment strategies employed to combat this disease. Moreover, we delve into emerging research avenues and prospective advancements that hold the promise of transforming the management and outcomes of HGSOC. This review examines the importance of the tumor microenvironment (TME). In addition, we aim to underscore the critical importance of placing the patient's perspective and diversity at the forefront of therapeutic strategies, thereby fostering a genuinely participatory decision‐making process and ultimately improving patient quality of life (QoL). We advocate that a paradigm shift in centralized therapeutic structures is needed in personalized medicine for patients with HGSOC and should be urgently implemented.

ETIOLOGY AND EPIDEMIOLOGY

The interplay between genetics, environment, and lifestyle factors forms a complex web influencing the incidence and progression of HGSOC. The incidence and prevalence rates of HGSOC exhibit significant variation across diverse populations; however, disparities are well known and particularly striking in the gynecologic oncology care continuum and should be considered when interpreting these data. There is a compelling need to include minority groups in clinical trials, and this issue is broadly recognized by leading scientific societies and organizations, such as the Gynecologic Cancer Intergroup, the American Society of Clinical Oncology, and the World Health Organization. Therefore, we suggest a circumscribed interpretation of HGSOC studies with inadequate representation of the patient pool worldwide (Figure 2). Understanding health care disparities in patients with HGSOC is critical for developing effective prevention and personalized therapeutic approaches. We highlight the importance of diversity in HGSOC because diagnosis and treatment should be available regardless of race/ethnicity, age, gender, socioeconomic status, education, geographic region, sexual identity, disability, or language. 7 , 8 The analysis of epidemiologic data has highlighted significant variations in incidence and survival rates across different demographic groups. In particular, our review substantiates that racial and socioeconomic disparities play a central role in influencing HGSOC outcomes. This reinforces the notion that research approaches must integrate broad determinants of health, encompassing sociodemographic variables, educational level, and behavioral factors that may affect survivorship. Moreover, the clinical and societal implications of embracing a diversity‐informed approach are profound. Incorporating nuanced diversity metrics not only enhances our understanding of disease etiology but also ensures that treatment strategies are equitable and personalized. The emphasis on early detection, particularly among populations at elevated risk, is not merely a clinical imperative but also a matter of social justice. Emphasizing diversity fosters innovation in research methodologies and encourages collaboration among researchers from varied backgrounds. Diverse teams bring unique perspectives, driving creativity and enhancing problem solving (Figure 2). This inclusivity can lead to the discovery of novel biomarkers and treatment responses that may vary by ethnicity and genetic background.

FIGURE 2.

FIGURE 2

Diversity in gynecologic oncology. This scheme depicts the ultimate need to include underrepresented minorities and to promote diversity. Racial inequities are well known and particularly striking in the gynecologic oncology care continuum. There is a strong need to include racial and ethnic minority groups in clinical trials. We call for a circumscribed interpretation of high‐grade serous ovarian cancer studies, with inadequate representation of the patient pool. Understanding health care disparities in gynecologic oncology is critical for developing effective, personalized therapeutic approaches. Diversity in patients with high‐grade serous ovarian cancer and in gynecologic oncology should be respected and will be a benefit for the scientific community, leading to more robust data for publications based on innovative and creative research generated in an excellent team. Created with Biorender (License No. RD27ZKH4P1).

HGSOC is predominantly diagnosed in women aged 55–64 years, with incidence rates peaking for all women in their late 70s. 4 According to GLOBOCAN 2020 data, ovarian cancer, which includes HGSOC, is the seventh most common cancer among women and the eighth leading cause of cancer death worldwide. 9 The incidence and mortality rates of HGSOC exhibit considerable geographic variations. An association between socioeconomic conditions and HGSOC incidence is evident and significantly influences the disease's epidemiology and survival rates. 10 , 11 , 12 These analyses underline the existence of disparities affecting women with HGSOC in different geographic regions.

Environmental and lifestyle factors, obesity, use of hormone‐replacement therapy, and reproductive history are also implicated in the epidemiology of HGSOC. 13 , 14 Lower parity, earlier age of menarche, later age of menopause, higher body mass index, endometriosis, pelvic inflammatory disease, and longer use of hormone‐replacement therapy after menopause all increase HGSOC risk; whereas higher parity, use of hormonal contraception, breastfeeding, and tubal ligation all lower HGSOC risk. Familial aggregation of HGSOC and other cancers, such as breast, pancreatic, melanoma, and colon cancers, in first‐degree relatives underscores the importance of genetic epidemiology in HGSOC management. 15 Unfortunately, genetic testing is often less accessible to minority groups despite equal or high prevalence of mutations and although variants of unknown significance are more common among minority patients when genetic testing is offered. 16 This evidence underpins the rationale for incorporating comprehensive genetic assessments into the clinical pathway to optimize personalized treatment strategies.

PATHOGENESIS AND TUMOR MICROENVIRONMENT

Currently, the general consensus is that the fallopian tube epithelium is the primary origin for most HGSOC tumors. 17 , 18 , 19 HGSOC appears to develop from three precursors in the fallopian tube: secretory cell outgrowths, p53 signatures, and serous tubal intraperitoneal carcinomas (STICs; Figure 3). Secretory cell outgrowths are extended regions of secretory cells in the tubal epithelium without intervening ciliated cells. The p53 signatures are morphologically normal‐appearing regions of epithelium with contiguous stretches of secretory cells expressing mutant p53 identifiable by immunohistochemistry. STICs are noninvasive lesions with cytologic atypia, high proliferative index, and hyperplasia that develop in the distal fallopian tube epithelium and can progress to malignancy either on the tubal surface or in the periphery after they have implanted either on the ovarian surface or peritoneum. 20 , 21 This exfoliative behavior is highly characteristic of HGSOC, and peritoneal metastases generally occur as a series of metastatic events, rather than stepwise outward growth from a single lesion. Notably, so‐called primary peritoneal carcinomas may exhibit STICs as well, making this a potential precursor lesion for this disease. Nuclear atypia, high mitotic activity, and apoptotic bodies are commonly observed in STICs, as are TP53 mutations, which may be nonsense or missense. STICs may be preceded by serous tubal intraepithelial lesions, which may or may not exhibit a mutant p53 signature, are characterized by a low Ki‐67 proliferation index (<10%), and are commonly referred to as dormant. 22 , 23 Studies indicate that incidental STICs are more commonly seen in women at high risk of cancer because of germline BRCA1 and/or BRCA2 mutations. 24 Although STICs are clearly linked with a heightened risk of HGSOC, not all STICs progress to HGSOC, nor do all HGSOCs arise from STICs, even among women with high‐risk factors. 25

FIGURE 3.

FIGURE 3

High‐grade serous ovarian cancer origin and development. This scheme depicts the precursor lesions of HGSOC and the invasion of STICs on the ovarian surface in the distal fallopian tube epithelium. Immunophenotypic classification: the T‐cell inflamed or hot category means that T cells infiltrate deposits (islets) of malignant cells and the surrounding and intervening stroma; whereas the noninflamed category, also known as immune‐desert or cold tumors, lack this feature. The excluded classification means that T cells remain confined to the stroma, and there are no deposits of malignant cells. + indicates positive; HGSOC, high‐grade serous ovarian carcinoma; IGF, insulin‐like growth factor; ROS, reactive oxygen species; SCOUT, secretory cell outgrowth; SEE‐FIM, Sectioning and Extensively Examining the FIMbriated End; STICs, serous tubal intraperitoneal carcinomas; TAM, tumor‐associated macrophage. Created with Biorender (License No. MB27ZKHB14).

Moreover, in patients with HGSOC, we recommend that the tubal resections should be completely examined pathologically by applying the SEE‐FIM (Sectioning and Extensively Examining the FIMbriated End) protocol. Here, the fimbrial funnel is examined in longitudinal sections, and the tube is examined in cross sections. During opportunistic salpingectomy, at least the fimbrial funnel should be completely examined. 26 Therefore, for lesions suspected of STIC, we recommend immunohistochemical examination for p53 and Ki‐67 that can be used to confirm the diagnosis.

The molecular and cellular mechanisms of HGSOC are predominantly driven by genetic mutations and dysregulated cellular signaling pathways. As mentioned above, mutations in the TP53 gene are nearly ubiquitous in HGSOC and are considered a hallmark of the disease. 14 , 19 , 27 TP53 encodes the p53 tumor‐suppressor protein, which plays a critical role in maintaining genomic stability and preventing tumor development. Loss of p53 function caused by TP53 mutations can lead to inappropriate or unregulated cellular proliferation and tumor growth. 19 , 28

Other genetic alterations implicated in HGSOC include mutations in BRCA1, BRCA2, and other genes involved in homologous recombination repair pathways, contributing to genomic instability and oncogenesis. 29 , 30 BRCA1 and BRCA2 mutations loom large in this context, increasing the lifetime risk for ovarian cancer and influencing the tumor's response to therapy. 31 , 32 Beyond these well‐established genetic abnormalities, emerging research has identified epigenetic alterations, including DNA methylation and histone modification, which may contribute to cancer pathogenesis by altering gene expression without changing the DNA sequence. 33 , 34

Importantly, the TME plays a critical role in the pathogenesis of HGSOC (Figure 4). The TME of HGSOC is a highly orchestrated and dynamic ecosystem composed of a diverse array of cellular and acellular components that collectively contribute to tumor progression, metastasis, and therapeutic resistance. 35 Epithelial ovarian cancer cells engage in reciprocal crosstalk with various stromal cell types, including cancer‐associated fibroblasts, immune cells, stromal cells, extracellular matrix, exosomes, and neovascular blood vessels. 36 , 37 , 38 Cancer‐associated fibroblasts (CAFs) are particularly significant because they modulate the TME to support tumor growth and metastasis. 38 The intricate interactions between cancer cells and CAFs play a pivotal role in the progression and therapeutic resistance of HGSOC. In addition, the immune microenvironment, characterized by the presence of tumor‐infiltrating lymphocytes (TILs) and macrophages, influences tumor behavior and patient prognosis. 39 , 40 The above‐mentioned interactions promote the establishment of a favorable niche that supports tumor growth, angiogenesis, immune evasion, and the metastatic dissemination of ovarian cancer cells (Figure 3). CAFs secrete factors that promote angiogenesis, cancer cell proliferation, and survival while also altering the extracellular matrix to facilitate tumor invasion. 41 More precisely, CAFs within the ovarian TME (Figure 4) secrete a plethora of cytokines, chemokines, and extracellular matrix components that foster an immunosuppressive milieu, facilitate the epithelial‐mesenchymal transition of ovarian cancer cells, and prime the metastatic niche in the omentum. 42 , 43 Ovarian cancer cells, in turn, can reprogram stromal cells to adopt a pro‐tumorigenic phenotype, thereby creating a self‐reinforcing cycle of reciprocal signaling that perpetuates disease progression. This reprogramming of stromal cells by ovarian cancer cells is a key mechanism that sustains the TME and facilitates continued tumor growth, invasion, and metastasis. Moreover, tumor cells possess an abnormal extracellular matrix deposition that leads to stiffness, proliferation, and resistance to cell death. 44 The ability of ovarian cancer cells to alter the phenotype of surrounding stromal cells is a critical component of the dynamic interplay between the cancer cells of HGSOC and their microenvironment. 45 Another one of the key factors that contribute to ovarian cancer development and progression is oxidative stress (OS), which can result from an imbalance between the production of reactive oxygen species (ROS) and the ability of the cell to neutralize them. 46 Ovulation‐induced generation of ROS also influences the occurrence of HGSOC. 47 Moreover, OS can trigger the angiogenic switch, a cascade of biologic events that are essential for tumor growth and metastasis. 48

FIGURE 4.

FIGURE 4

The microenvironment of high‐grade serous ovarian cancer. In the tumor microenvironment, cancer‐associated fibroblasts (CAFs) are particularly important because they support tumor growth and metastasis. Tumor cells with increased DNA damage show signs of inflammation upon sensing DNA damage and the upregulation of stimulator of interferon genes (STING) and tumor‐intrinsic type I interferon signaling pathways. BRCA1/BRCA2 and TP53 gene mutations are common. Several mechanisms contribute to immunosuppression within tumors, including the secretion of IL‐10/IL‐6 with inhibition of the PD‐1 receptor and the immunosuppressive actions of myeloid‐derived suppressor cells (MDSCs), tumor‐associated macrophages (TAMs), or CAFs. CAFs indicates cancer‐associated fibroblasts; DC, dendritic cells; PD1, programmed death 1; PD‐L1, programmed death‐ligand 1; Treg, regulatory T cells. Created in Biorender (License No. VN27ZKHGXA).

Mitochondria are a significant source of ROS and are known to be affected by the TME. 49 Mitochondrial dysfunction can lead to further OS, exacerbating the already hostile tumor environment. This interplay between OS, mitochondrial dysfunction, and the TME can have profound implications for cancer development (Figure 4), progression, and resistance to treatment. 50 Interestingly, the chemoresistance of ovarian cancer relies in part on an enhanced antioxidant capacity of the cancer cells. HGSOC cells protect themselves by initiating antioxidant responses, such as upregulating SLC7A11 (solute carrier family 7 member 11), resulting in alterations in cysteine import into cancer cells and preserving redox balance. 51

In HGSOCs, there is a heterogeneous mixture of tumor cells infiltrated by, among others, various immune or stromal cells. Several mechanisms contribute to immunosuppression within tumors, including the inhibition of CD8‐positive T cells by regulatory T cells, secretion of IL‐10/IL‐6 with promotion of the programmed death 1 receptor (Figure 4), and the immunosuppressive actions of myeloid‐derived suppressor cells, tumor‐associated macrophages, and CAFs. 38 Interestingly, among all ovarian cancers, HGSOC is the most likely to show significant CD8‐positive T‐cell infiltration, reported to be a favorable prognostic factor. 45 The presence of intraepithelial TILs in tumor islets has been linked to improved survival in numerous studies, although these are present in less than one half of cases. 52 , 53 It has been demonstrated in HGSOC that CD8‐positive T cells within so‐called inflamed tumors, also known as hot tumors, invade the stroma surrounding malignant cells and express transcriptional signatures of cytotoxic T cells, such as granzyme B (GZMB), GZMA, GZMH, granulysin (GNLY), and interferon γ (IFNG). 45 , 54 , 55 Chemokine circuits precisely regulate the infiltration and retention of intraepithelial CD8‐positive TILs in the TME, whose presence is linked to slower tumor progression and extended survival. 45 However, because HGSOC tends to metastasize as a series of episodic exfoliative events, different molecular changes and pathways are activated at different metastatic tumor sites, giving rise to a heterogeneous immunophenotype. 56

Contemporary studies provide evidence that there is a significant link between mitochondria, apoptosis, induced glutathione inactivation, and platinum resistance in ovarian cancer, suggesting that mitochondrial metabolism could serve as a potential target in cancer precision medicine. 57 Mutations in mitochondrial DNA, such as ND2 F40L, can impact proton transport pathways, affect mitochondrial complex I activity, increase ROS levels, and decrease sensitivity to cisplatin by activating the nuclear factor‐κB signaling pathway and inducing IAP (inhibitor of apoptosis) expression. 58 Moreover, it has been established that mitochondria are involved in the intrinsic apoptotic signaling pathway to control cell death. 59 Therefore, a thorough analysis of mitochondria‐related genes and their connection with apoptosis could enhance understanding of HGSOC progression and platinum resistance, paving the way for developing novel strategies to combat platinum resistance.

DIAGNOSIS, STAGING, AND MOVING TOWARD MOLECULAR CLASSIFICATION

It is our belief that a critical view on the current classification and staging systems commonly used in patients with HGSOC is needed. Ovarian cancers can be categorized by molecular classification and anatomic staging, although, given the heterogeneity of individual tumors, not every patient can be precisely categorized. Keeping this caveat in mind, we suggest using the concept of the five major, pathogenetically independent, histologically and molecularly different groups of ovarian carcinomas, namely: (1) high‐grade serous carcinomas; (2) endometrioid and (3) clear cell carcinomas, which often arise in the ovary and are often associated with endometriosis; (4) mucinous carcinomas; and (5) low‐grade serous carcinomas. An algorithm using molecular markers for the aforementioned five types is provided below (Figure 5).

FIGURE 5.

FIGURE 5

Algorithm for the diagnosis of histologic types. The scheme shows the diagnostic algorithm based on additional immunohistochemistry examinations. − indicates negative; +, positive; WT, wild type.

Traditionally, HGSOCs were classified as ovarian, tubal, or primary peritoneal in origin. However, as noted, most HGSOCs are thought to originate from tubal precursors. HGSOC is now classified as originating from the fallopian tube (Figure 3) when the tube is either affected by STICs, contains mucosal carcinoma, or is largely occupied by HGSOC. 60 , 61 , 62 Consequently, the majority of HGSOCs are now regarded as having an extra ovarian origin, and a significant shift in the International Classification of Diseases for Oncology site codes from C56.1 (ovary) to C57 was introduced by the World Health Organization. 63 Recent advancements in the molecular characterization of epithelial ovarian cancer have further refined these descriptions to include TP53 mutational status and homologous DNA damage repair proficiency.

HGSOC is pathologically staged according to the International Federation of Gynecology and Obstetrics (FIGO) staging system (Table 1). Staging and grading of ovarian cancer, guided by the FIGO system, are fundamental to determining the extent of the disease and tailoring treatment approaches. The FIGO system ranges from stage I (tumor confined to the ovaries) to stage IV (distant metastasis), thus offering a standardized framework for evaluating disease progression. 64 Another classification is provided by the TNM (primary tumor [T], regional lymph nodes [N], and distant metastases [M]) system based on the anatomic spread of malignant tumors (Table 1). An effort to harmonize FIGO with TNM is underway. Moreover, histopathologic features are essential for the accurate diagnosis of HGSOC. 65 , 66 The presence of the aforementioned papillary structures and STICs in the fallopian tubes supports a diagnosis of HGSOC, highlighting the interplay between ovarian and fallopian tube origins. 60 , 61 The Silverberg grading system adds to these descriptions by categorizing tumors based on architectural and nuclear features, providing further granularity that can inform prognosis and therapeutic decisions. 67 Moreover, the Silverberg grading system appears to be preferable for categorizing patients with HGSOC for prognosis and disease management. 68

TABLE 1.

Tumor, lymph node, and metastasis and International Federation of Gynecology and Obstetrics classification systems for ovarian cancer.

TNM FIGO stage Definition
TX Primary tumors cannot be assessed
T0 No evidence of a primary tumor
T1 I Tumor limited to ovaries (one or both) or fallopian tube(s)
T1a IA Tumor limited to one ovary; capsule intact, no tumor on the surface of the ovary or fallopian tube; no malignant cells in ascites or with peritoneal lavage
T1b IB Tumor limited to both ovaries or tubes; capsule intact, no tumor on the surface of both ovaries or tubes; no malignant cells in ascites or peritoneal lavage
T1c IC Tumor limited to one or both ovaries or tubes with one of the following:
T1c1 IC1 Tumor cell dissemination during surgery
T1c2 IC2 Capsule rupture before surgery or tumor on ovarian or tubal surface
T1c3 IC3 or malignant cells in ascites or peritoneal lavage
T2 II Tumor affects one or both ovaries or tubes and spreads throughout the pelvis, below the edge of the pelvis
T2a IIA Spread to and/or implants on the uterus and/or tube(s) and/or ovaries
T2b IIB Spread to other pelvic tissues
T3 and/or N1 III Tumor affecting one or both ovaries or tubes or primary carcinoma of the peritoneum with cytologic or histologic evidence, spread to the peritoneum outside the pelvis, and/or regional lymph node metastases
N1 Only retroperitoneal lymph node metastases
N1a IIIA1i Lymph node metastases ≤10 mm in greatest extent
N1b IIIA1ii Lymph node metastases >10 mm in greatest extent
T3a, every N IIIA2 Microscopic spread beyond the pelvis (above the edge of the pelvis) with or without retroperitoneal lymph node metastases beyond the pelvis
T3b, every N IIIB Macroscopic peritoneal metastases beyond the pelvis, ≤2 cm in greatest extent
T3c every N IIIC Peritoneal metastases beyond the pelvis, ≥N1, 2 cm in greatest extent, and/or regional lymph node metastases, including tumor spread to the liver and spleen capsule without parenchymal involvement of these organs
M1 IV Distant metastases (exclusively peritoneal metastases)
M1a IVA Pleural effusions and positive cytology
M1b IVB Parenchymal metastases and distant metastases in extra‐abdominal organs (including inguinal lymph nodes and lymph nodes outside the abdominal cavity

Note: Metastases to the liver capsule correspond to T3/stage III, liver parenchyma metastases correspond to M1/stage IV according to the German S3 guidelines (S3‐Leitlinie Diagnostik, Therapie und Nachsorge maligner Ovarialtumoren Version 6.0) and Feng 2024. 64

Abbreviations: FIGO, International Federation of Gynecology and Obstetrics; TNM, primary tumor (T), regional lymph nodes (N), and distant metastases (M) system based on the anatomic spread of malignant tumors.

The clear distinction between low‐grade and high‐grade ovarian carcinomas was established in 2014. Although a dualistic classification (type I and II tumors) existed previously, it was the World Health Organization classification of 2014 that standardized this approach. Consequently, patients diagnosed before 2014, particularly those initially classified as grade 2, should be reclassified if and when a recurrence occurs according to these guidelines. For clinicians and specialists, it is important to distinguish high‐grade serous carcinoma from other serous neoplasms, such as low‐grade serous carcinoma and serous borderline tumors, which have different molecular features and clinical outcomes. Low‐grade serous carcinoma is characterized by minimal nuclear atypia and lower mitotic activity (as defined by the Ki‐67 index) compared with high‐grade serous carcinoma. It generally affects younger patients and has a slower rate of progression. In contrast, high‐grade serous carcinoma is marked by pronounced nuclear atypia, a high mitotic index, and a rapid proliferative rate, leading to a more aggressive clinical course. 5 As mentioned above, high‐grade serous carcinomas are characterized in most cases by mutations in the TP53 gene, which manifests on immunohistochemistry either as pronounced overexpression (overexpression pattern) or as the complete absence of staining (null pattern). Low‐grade serous carcinomas, in contrast, typically exhibit wild‐type p53 and more frequently harbor mutations in other genes, such as KRAS or BRAF. In practice, p53 staining is often used to complement morphologic classification or to confirm uncertain cases. Nevertheless, p53 assessment should be considered standard in the pathologic diagnosis of ovarian carcinoma to provide a comprehensive diagnostic framework and to guide optimal therapeutic decisions.

I could no longer cope with the psychological uncertainty. All samples were forwarded to the pathology department for further investigation. Ten days later, the doctor told me that a malignant carcinoma had been diagnosed by the pathologists in both the ovary and the uterus. The doctors initially assumed that it was a single tumor that had already metastasized—but whether the ovary or the uterus was primary was an issue doctors were still trying to clarify in the pathology department at that time.

In addition to the morphologic and anatomic classification systems, The Cancer Genome Atlas described four molecular subtypes of HGSOC based on unsupervised clusters from messenger RNA expression data. 69 These have been called the immunoreactive subtype (chemokine expression), the proliferative subtype (proliferation marker expression), the differentiated subtype (ovarian tumor marker expression), and the mesenchymal subtype (expression of markers suggestive of increased stromal components). The immunoreactive subtype demonstrated the significantly highest progression‐free survival and overall survival, whereas the overall survival rate was lowest in the mesenchymal subtype. 70

A summary of HGSOC characteristics is provided in Table 2. All these points emphasize the importance of specialized gynecologic oncologists and trained pathologists for the review of tissue. Importantly, this should be an expedited process, acknowledging that delays in final diagnosis can be exceptionally stressful for the patient.

TABLE 2.

Summary of high‐grade serous ovarian cancer characteristics.

Subtype specific information High‐grade serous ovarian cancer
Purported cell of origin Fallopian tube
Precursor lesion Serous tubal intraepithelial carcinoma lesions
Inherited predisposition 15%–25%
Molecular features Somatic tumor testing may reveal: Chromosomal instability; copy number alterations; homologous repair defects; alterations of the genes TP53 and CCNE1, PIK3CA amplifications, BRCA1/2 mutations, or other forms of homologous recombination deficiency; microsatellite instability; mismatch repair; HER2 expression; tumor mutational burden; BRAF V600E mutation; folate receptor alpha expression; RET mutations; NTRK gene fusion
Risk factors Germline mutations in BRCA1, BRCA2, or other homologous DNA repair genes or Lynch syndrome; environmental and lifestyle factors; obesity; hormone‐replacement therapy and reproductive history, including parity, age at first birth, lifetime number of ovulatory cycles, and endometriosis; family history of ovarian, breast, or bowel cancer
Potential protective factors Oral contraception treatment, tubal ligation, birth, and breastfeeding; bilateral salpingo‐oophorectomy recommended for patients with a BRCA mutation after childbearing is complete (or not desired), ideally between ages 35 and 40 years for BRCA1 and between ages 40 and 45 years for BRCA2
Platinum‐based chemotherapy Usually sensitive
PARP inhibitor Variably sensitive
Diagnostic clinical and pathologic features

Pelvic examination; imaging tests: Blood tests (CA 125, HE‐4, OVA1; diagnostic laparoscopy; surgery (removing mass to determine whether it is cancerous); biopsy (fine‐needle aspiration/fine‐needle aspiration biopsy of metastatic sites, core biopsy, or paracentesis)

Pathologic features: Solid, papillary, and glandular areas with high mitotic activity and high‐grade atypia; often bilateral and advanced stage at clinical presentation

Diagnostic imaging Pelvic ultrasound, CT chest CT or MRI abdomen/pelvis, PET (in select cases)
Prognostic feature Postoperative residual disease is the most important prognostic factor

Abbreviations: CA 125, cancer antigen 125; CT, computed tomography; FNA, fine‐needle aspiration; HE‐4, human epididymis protein 4; MRI, magnetic resonance imaging; OVA1, blood test from Aspira Women's Health that measures five biomarkers assessing malignancy; PARP, poly(adenosine diphosphate‐ribose) polymerase; PET, positron emission tomography.

TREATMENT STRATEGIES FOR HIGH‐GRADE SEROUS OVARIAN CANCER

The treatment paradigm for HGSOC is evolving rapidly, encompassing a spectrum of therapeutic approaches that aim to extend survival and enhance QoL for patients. Surgical interventions, chemotherapy regimens, and emerging targeted therapies are being used in an expanding repertoire for treatment options. For the prediction of HGSOC therapy success, we suggest that patients should be categorized according to their general health status into the following three groups: (1) prefrail, (2) frail, and (3) nonfrail. Frailty is considered to be a state of elevated vulnerability to adverse outcomes. 71 More specifically, frailty represents a multisystem syndrome of low physiologic reserves combined with a lowered capacity to respond to stressors. In addition, frailty has been reported to affect the prediction of surgical outcomes more than the patient's age. 72 To measure frailty, a respective index has been established, representing a ratio of deficits that are present before surgery to the total number of deficits taken into consideration. 71 Based on this ratio, a patient will receive a frailty index of 0.2 (10 of 50) if, for example, 50 deficits have been considered and 10 were present. In other words, the frailty index principle reflects health deficits, such as diseases, disabilities, or abnormalities of laboratory, radiographic or electrocardiographic outcomes. However, the classification of patients according to their frailty status is not routinely performed. Moreover, we suggest a personalized prehabilitation therapeutic strategy (Figure 6) that is combined with an enhanced recovery after surgery approach to decrease postoperative morbidity. 72 , 73 , 74 , 75 It has been demonstrated that the preoperative quality‐of‐life evaluation can be a predictive marker for severe postoperative complications in patients with ovarian cancer. 75 Importantly, the aforementioned study provided evidence that patients with limited mobility, debilitating symptoms, and cognitive impairment had a higher risk of developing severe postsurgery complications. 75

The doctor gave me very helpful advice, telling me that experience has shown that it is recommended to be honest with children and family about such a diagnosis. I didn’t ask myself how I could deal with this in the future. The situation was too acute, with a 14‐year‐old girl at home. Nevertheless, the question always weighed on me as to whether my daughter might be genetically affected. At work, I was open with my colleagues about my cancer diagnosis and received a great deal of support and encouragement. However, I also felt that many of them were hesitant to remain close to me.

FIGURE 6.

FIGURE 6

Patient management. Scheme showing the proposed personalized prehabilitation therapeutic strategy that is combined with an enhanced recovery after surgery (ERAS) to decrease postoperative morbidity. For the prediction of cancer therapy success, patients should be precategorized according to their general status into the following groups: prefrail, frail, and nonfrail.

PATIENT PREFERENCES IN THE TREATMENT DECISION‐MAKING PROCESS

A key aspect of effective ovarian cancer management is the integration of patient preferences into the treatment decision‐making process. By actively involving patients in discussions about their therapeutic options, clinicians can enhance both adherence to the chosen treatments and patient satisfaction. This approach ensures that medical decisions align not only with clinical evidence but also with the individual values, goals, and circumstances of each patient. Studies, such as those from the North‐Eastern German Society for Gynecologic Oncology (Nord‐Ostdeutsche Gesellschaft fur Gynakologische Onkologie/NOGGO) expression series, highlight the importance of considering patient preferences as a central component of shared decision making. This collaborative process fosters a sense of empowerment among patients, enabling them to take an active role in their care, particularly when faced with the psychological uncertainty before definitive diagnosis. Moreover, aligning treatment strategies with patient priorities has been shown to improve adherence and optimize outcomes, in terms of both QoL and clinical effectiveness. Incorporating patient preferences is not only ethically essential but also is a practical strategy to build trust and strengthen the therapeutic alliance. This ensures that care is both patient‐centered and reflective of the diverse needs and aspirations of those living with ovarian cancer. 76 , 77 HGSOC management should be tailored to the unique priorities and concerns of each patient. By recognizing and addressing individual aspirations, goals, and preferences, care teams can develop personalized treatment plans that optimize not only medical outcomes but also enhance the overall QoL. Fertility‐preserving strategies should be provided to every patient in those with early stage disease and a low risk for recurrence. 78 The quality‐of‐life aspect will be discussed in the management of HGSOC, and understanding the patient's perspective is indispensable because the disease exerts a profound impact on the physical, psychological, and social dimensions of life. Fear, often described as one of the most dominant emotions, significantly influences patient decision making and their overall treatment experience. 79 , 80 This fear is particularly pronounced in the context of ovarian cancer surgery, which is more extensive compared with surgeries for other malignancies. This highlights the need for early integration of psychological and psycho‐oncologic support to help patients navigate the emotional burden associated with their diagnosis and treatment (Figure 7).

FIGURE 7.

FIGURE 7

Summary box I. Key points for shared decision‐making during treatment selection in patients with high‐grade squamous ovarian cancer (HGSOC).

Surgical management

Current standard treatments for HGSOC include an aggressive surgical approach aimed at maximum cytoreduction, which remains indispensable in the therapeutic landscape of HGSOC. 3 , 81 For HGSOCs, primary surgical cytoreduction, including total abdominal hysterectomy, bilateral salpingo‐oophorectomy, omentectomy, and peritoneal biopsies, remains the standard of care. HGSOC surgery is one of the most invasive interventions in oncology. This complexity contributes to heightened anxiety among patients, often described as terrifying. For many patients, these fears extend beyond physical recovery, encompassing worries about body image, independence, and future QoL. Early psycho‐oncologic interventions are critical to addressing these concerns. Providing patients with preoperative counseling and fostering open discussions about their fears can alleviate anxiety and help them feel more prepared for the surgical journey. In early stage HGSOC, most guidelines continue to recommend pelvic and para‐aortic nodal staging because identifying lymph node metastases would upstage the disease from stage I to stage IIIA. This distinction carries significant therapeutic implications because maintenance treatments are generally indicated for stage III disease only. However, the therapeutic value of systematic lymph node dissection in this context remains unclear. Interestingly, in patients with macroscopically normal lymph nodes, lymphadenectomy has no influence on progression‐free survival or overall survival but significantly increases morbidity. 82 Although nodal metastases are prognostic, routine lymphadenectomy does not improve overall survival. 83 Still, in patients with apparent pelvic‐confined disease, systematic pelvic and para‐aortic lymphadenectomy can upstage 15%–20% of patients. 84 Identification of these patients with occult stage IIIC1 or IIIC2 disease can provide important prognostic information as well as select patients for poly(adenosine diphosphate‐ribose) polymerase (PARP)‐inhibitor (PARPi) or intravenous/intraperitoneal chemotherapy who otherwise might not be considered for these more aggressive modalities. 85

For advanced‐stage ovarian cancer, the quantity of residual disease after surgery remains highly prognostic for progression‐free and overall survival. 86 The definition of an optimal cytoreduction has shifted in recent years from no single residual deposit of disease <2 cm, to <1 cm, to no gross residual disease (R0). 87 Achieving an R0 resection in patients with bulky stage IIIC or IV disease may require extensive procedures, such as low anterior resection, ileocectomy, multiple small bowel resections, splenectomy, partial gastrectomy, or diaphragmatic resection. This push for a complete resection creates a natural tension between surgical aggressiveness and surgical morbidity, making the appropriate selection of patients for primary cytoreduction vitally important. This assessment should include consideration of frailty, as noted above. Although a complete resection portends a more favorable clinical outcome, a suboptimal cytoreductive effort exposes a patient to a painful and potentially prolonged recovery without improving her survival. For patients with radiographic stage IIIC disease, diagnostic laparoscopy to assess the intraperitoneal disease burden is highly accurate for predicting the feasibility of an R0 resection and reduces the likelihood of a suboptimal or aborted attempt at cytoreduction. 88 , 89

For patients with disease not amenable to primary cytoreduction, whether because of medical frailty or anatomic localization of disease burden, three randomized trials have indicated that neoadjuvant therapy with three or four cycles of platinum‐based chemotherapy, followed by interval cytoreduction, and then from two to four cycles of adjuvant chemotherapy is a noninferior approach. 90 , 91 , 92 According to the American Society of Clinical Oncology and Society of Gynecologic Oncology consensus, patients who have a high perioperative risk profile or a low likelihood of achieving cytoreduction to <1 cm (ideally to no visible disease) should receive neoadjuvant chemotherapy. 93 Neoadjuvant chemotherapy in this population is associated with lower rates of surgical complications, perioperative mortality, and ostomy formation. 94 Patients with bulky stage IIIC disease not amenable to primary cytoreduction who otherwise have good functional status, respond well to chemotherapy, and are able to achieve an optimal cytoreduction at interval surgery should be evaluated for possible heated intraperitoneal chemotherapy (HIPEC), typically with cisplatin, at the time of interval cytoreduction. 95 , 96 , 97 Nonetheless, wide variation persists among reports in terms of technique, dosing, temperature, and patient selection. Complete cytoreductive surgery remains an essential part of HGSOC therapy; however, in underserved regions, complex and multispecialty procedures may be difficult to implement. Improved surgical training, well staffed operating room support services, safe blood banks, and adequate surgical equipment are prerequisites for high‐quality ovarian cancer care and should be included in advocacy efforts to promote improved outcomes for ovarian cancer globally. Policy changes, such as a separate International Classification of Diseases for Oncology 10th Revision code for HGSOC, might focus these efforts and ensure that policymakers devote sufficient resources to making these critical surgeries available in reference centers. There is a need for a fundamental understanding that enhanced funding and meticulous resource allocation can drive improvements in cytoreduction surgical efficacy, especially in underserved regions. Research has provided strong evidence that increased funding cultivates an environment in which high‐precision technologies and specialized training programs are routinely accessible and the administrative processes become more streamlined. 98 , 99 , 100 The clear association between enhanced financial inputs and improved surgical outcomes underscores the need for more dynamic resource‐allocation models in oncologic surgery. 101 Policymakers must recognize that investing in health care infrastructure and clinical training can yield immediate and measurable benefits.

We advocate for a re‐examination of current budgeting practices within health care institutions. The causal relations between increased funding and improved surgical outcomes are multifaceted. 102 , 103 , 104 Enhanced funding directly translates into better surgical equipment, increased staffing ratios, and more comprehensive training programs. These improvements create a more conducive surgical environment that promotes precision and accountability. 105 , 106

Frontline chemotherapy

Platinum‐based combination regimens have been the first‐line therapy for HGSOC since the 1980s, initially with cisplatin, but the current worldwide standard consists of carboplatin and paclitaxel. The most commonly used regimen is six cycles of a 21‐day cycle with intravenous carboplatin at an area under the curve of 5 or 6 combined with intravenous paclitaxel, both given on cycle day 1. use carboplatin is generally well tolerated as a single agent, studies have examined withholding paclitaxel from older, more frail patients. However, even among more vulnerable populations, the doublet regimen is associated with better progression‐free and overall survival. 107 Alternate platinum doublets, such as carboplatin/liposomal doxorubicin and carboplatin/docetaxel, are noninferior to carboplatin/paclitaxel but have different toxicity profiles (less neuropathy and alopecia, but at the expense of more hematologic toxicity). 108 , 109 Even so, the Gynecologic Oncology Group (GOG) GOG 182‐ICON5 study, one of the largest clinical trials ever performed for ovarian cancer, concluded that the addition of a third cytotoxic agent to carboplatin and paclitaxel offered no additional benefit. 110 Notably, cisplatin is still used as part of the GOG‐172 intravenous/intraperitoneal regimen because intravenous carboplatin is not as effective as intraperitoneal. For intravenous therapy, there are not a lot of compelling reasons to choose cisplatin over carboplatin given the higher rates of neuropathy and renal toxicity as well as nausea from cisplatin. In select cases, one might consider cisplatin because of the lower rates of myelosuppression with cisplatin compared with carboplatin; however, in practice, most oncologists would address this problem with growth factor support.

I underwent a full course of six cycles of chemotherapy (paclitaxel and carboplatin), even though my doctors expressed uncertainty about whether it was the ideal strategy. This made the process feel more difficult and discouraging. I was very concerned about potential side effects, especially given how poorly I recovered from the surgery.

It is noteworthy that the determination of homologous recombination deficiency (HRD) status in patients with HGSOC is commonly taken into consideration to predict response to platinum‐based or PARPi therapy. A recent study demonstrated that BRCA mutational status, in combination with the BRCA1 methylation status, lends the highest predictive power for favorable clinical outcomes, such as high sensitivity to platinum‐based therapeutic strategies, whereas HRD positivity (BRCA unrelated) does not similarly predict platinum response. 111

Several studies have examined dosing variations of the carboplatin/paclitaxel doublet. A large phase 3 study in Japan reported weekly dosing of paclitaxel at 80 mg/m2 combined with 3‐weekly carboplatin led to a substantial improvement in overall survival compared with the standard 3‐weekly regimen. 112 However, this observation was not reproduced in confirmatory studies in the United States and in Europe. 113 , 114 A large, multicenter, phase 3 study in 810 patients with advanced ovarian cancer compared 3‐weekly versus weekly carboplatin‐paclitaxel treatment. The weekly regimen offered similar progression‐free survival but demonstrated a more stable QoL and fewer severe side effects. 115 , 116

Well selected patients may benefit from combining intraperitoneal chemotherapy with intravenous chemotherapy. For these patients, intravenous paclitaxel 135 mg/m2 over 24 hours on cycle day 1, intraperitoneal cisplatin 100 mg/m2 on cycle day 2, and intraperitoneal paclitaxel 60 mg/m2 on cycle day 8 of a 21‐day cycle is prescribed based on the GOG‐172 regimen, in which it resulted in an almost 16‐month improvement in overall survival compared with intravenous cisplatin/paclitaxel. 117 However, in GOG‐172, only 42% of patients completed all six cycles of treatment, highlighting the difficulty of this regimen.

In contrast, GOG‐252, in which all patients received bevacizumab plus either (1) intravenous carboplatin with weekly intravenous paclitaxel; or (2) intraperitoneal carboplatin with weekly intravenous paclitaxel; or (3) a modification of the GOG‐172 regimen with a 3‐hour, cycle day 1 paclitaxel infusion, dose‐reduced intraperitoneal cisplatin 75 mg/m2 on cycle day 2, plus intraperitoneal cisplatin intraperitoneal paclitaxel 60 mg/m2 on cycle day 8, failed to produce an improvement with the intraperitoneal regimens. 118 However, there has yet to be a comparison between the GOG‐172 intravenous/intraperitoneal regimen and the bevacizumab‐containing regimen.

Bevacizumab, a monoclonal antibody targeting the vascular endothelial growth factor receptor, may be added to cytotoxic chemotherapy for stage II–IV disease (Table 1). The ICON7 (EudraCT identifier ISRCTN91273375) and GOG‐218 phase 3 randomized trials both examined adding bevacizumab to standard chemotherapy followed by single‐agent bevacizumab maintenance therapy. 119 , 120 Although neither study demonstrated an overall survival improvement with the addition of bevacizumab, an exploratory analysis suggested a modest benefit for women with a poor prognosis, although that group was not well defined. Bevacizumab is especially useful for symptomatic relief in patients with large‐volume ascites. 121

One outstanding question in the management of advanced HGSOC is whether the addition of bevacizumab to a PARPi regimen is truly mandatory. Although bevacizumab has demonstrated efficacy in combination with chemotherapy and in maintenance settings, definitive data regarding its obligatory role when paired specifically with a PARPi remain inconclusive. Ongoing and future trials are expected to clarify whether this combination offers a significant advantage over PARPi monotherapy, particularly in terms of overall survival and QoL. 122 In the N‐Plus study (ClinicalTrials.gov identifier NCT05460000), which is a prospective, multicenter clinical trial investigating a de‐escalation approach specifically for patients with stage III, HRD‐positive ovarian cancer, early findings suggest that carefully selected HRD‐positive patients may benefit from this tailored approach without compromising oncologic outcomes, potentially improving the overall QoL. 123 Rather than administering a full‐intensity treatment regimen universally, the study explores whether a reduced or modified therapeutic strategy (e.g., fewer chemotherapy cycles or limited surgical intervention) can maintain efficacy while mitigating toxicity. 123

Wide variation in the upfront management of HGSOC remains, reflecting the uncertainty that remains. Whether to initiate therapy with neoadjuvant chemotherapy or primary debulking depends on the frailty of the patient (Figure 6), the expected morbidity of surgical recovery, the feasibility of complete cytoreduction, and the availability of surgical resources. Although, in many centers, most patients may receive neoadjuvant chemotherapy by default, in others, primary cytoreduction remains the preferred approach. When neoadjuvant chemotherapy is given, there are no precise guidelines to indicate whether three, four, or even six cycles before interval cytoreduction are better. In practice, patients are reassessed at regular intervals for physical fitness and radiographic response until it seems that complete cytoreduction will be feasible and well tolerated. Several studies have suggested that administering HIPEC at interval cytoreduction may improve survival, but these results have not been consistent, and it is unclear which patients benefit most from this more intensive approach. There is even less support for HIPEC at primary cytoreduction. Whereas intravenous/intraperitoneal chemotherapy was strongly recommended after the publication of GOG‐172 in 2006, enthusiasm for this approach has waned with the advent of maintenance therapies and the higher resource use required for these treatments. Even so, for select patients with stage IIIC disease, good performance status, and complete primary cytoreduction, the GOG‐172 regimen may be considered at centers experienced in the technique.

Maintenance therapy

After adjuvant chemotherapy, maintenance regimens are determined primarily by genomic profiling (Figure 8). For patients with germline mutations in BRCA1 or BRCA2 or those who undergo somatic tumor profiling suggesting a homologous repair defect, the addition of a PARPi significantly improves progression‐free and overall survival. 124 , 125 , 126 For patients receiving bevacizumab during primary chemotherapy, the addition of olaparib to bevacizumab improves progression‐free and overall survival compared with bevacizumab alone. 127 , 128 For patients without germline mutations in BRCA1 or evidence of a somatic homologous repair defect, PARPi with niraparib was previously a common option, but its use has been tempered given the potential for toxicity and likely modest clinical benefit, notably the finding of no improvement in overall survival. 124 , 129 , 130 , 131 For patients receiving bevacizumab during primary chemotherapy but without a homologous repair defect, bevacizumab alone remains a maintenance option (Figure 8).

FIGURE 8.

FIGURE 8

Summary box II. Key points of maintenance therapy for patients with high‐grade squamous ovarian cancer. HR indicates homologous recombination; HRD, homologous recombination deficiency; PARP, poly(adenosine diphosphate‐ribose) polymerase.

Management of recurrent disease

While primary treatment of HGSOC is highly efficacious at putting patients into clinical remission, most patients with advanced‐stage disease will relapse. Relapse can be diagnosed by radiographic progression, rising cancer antigen 125 (CA 125), or physical examination. Biopsy confirmation may be helpful in cases of diagnostic discordance (e.g., no change in CA 125 but a new radiographic finding) or to obtain fresh tissue for molecular profiling. However, earlier treatment of asymptomatic relapse diagnosed by CA 125 changes alone is not associated with improved survival. 132 Measurement of circulating tumor DNA to assess minimal residual disease is prognostic for relapse, but there are no data to support using circulating tumor DNA to guide initiation of treatment for suspected recurrence. 133

For women who relapse, the first action should be to assess the feasibility of repeat surgical resection. The DESKTOP III and SOC‐1 trials (ClinicalTrials.gov identifier NCT01166737 and NCT01611766, respectively) both established that, for patients with a platinum‐free interval of 6 months or more, good functional status, minimal ascites, a complete cytoreduction at the initial surgery, and a complete secondary cytoreduction followed by chemotherapy improve overall survival compared with chemotherapy alone. 3 , 134 Tertiary cytoreduction may also be beneficial but is usually limited to patients with oligometastatic recurrence and long treatment‐free intervals. 135 , 136

The expected response to a repeat challenge with platinum‐based therapy informs clinical decision making in the recurrent setting. In general, the longer the time interval since completing the last cycle of platinum‐based chemotherapy, the higher the likelihood of a favorable response to a repeat platinum challenge. 137 When relapse occurs more than 6 months from the completion of platinum‐based treatment, tumors are considered platinum‐sensitive. Tumors that recur less than 6 months from treatment are labeled platinum‐resistant, whereas those that progress during therapy or less than 1 month from treatment are called platinum‐refractory. 138

Platinum‐sensitive ovarian cancer is treated with another platinum‐based doublet. Although this could be carboplatin/paclitaxel again, often, residual neuropathy from the initial treatment or a desire to avoid repeat alopecia motivates the selection of an alternative regimen. The most common options are carboplatin/liposomal doxorubicin, carboplatin/gemcitabine, or carboplatin/topotecan. 139 , 140 , 141 The doublets are similar in efficacy. Bevacizumab can be added to standard chemotherapy for well selected patients. 142 For patients receiving bevacizumab with initial treatment, there may be a modest benefit to including bevacizumab again, with treatment and maintenance for subsequent therapy. 115 After a complete or partial response to chemotherapy, patients with germline BRCA1/2 mutations or somatic homologous repair defects should receive maintenance PARPi therapy, especially if it was not used after frontline treatment. 143 , 144 , 145 Patients without homologous repair defects may benefit from niraparib therapy. 143 , 146 For patients progressing after olaparib maintenance therapy, there may be a modest benefit from olaparib rechallenge, as demonstrated in the OReO/ENGOT‐ov38 trial. 147

In our practice, patients are rarely rechallenged with the same maintenance drug but, instead, are usually rotated to the alternate maintenance therapy from what they received in the first‐line setting. For example, if PARPi was given as first‐line maintenance but bevacizumab was not included in first‐line chemotherapy, we use a platinum‐based doublet with bevacizumab for the treatment of platinum‐sensitive recurrence, followed by bevacizumab maintenance. In a recent real‐world, international study, the efficacy of subsequent chemotherapy after PARPi exposure in patients with HGSOC was evaluated. In those patients who progressed on PARPi with a platinum‐free interval of more than 6 months, the platinum‐based chemotherapy rechallenge remained the best option 148 (Figure 9).

FIGURE 9.

FIGURE 9

Summary box III. Key points of recurrence therapy for patients with high‐grade squamous ovarian cancer. ER, estrogen therapy; ET, endocrine therapy; PARP, poly(adenosine diphosphate‐ribose) polymerase; TMB‐H, high tumor mutational burden.

Progression‐free intervals typically shorten with each subsequent round of platinum‐based therapy, such that platinum‐resistant ovarian cancer remains the leading cause of ovarian cancer deaths. 149 Until recently, options for platinum‐resistant HGSOC were limited to cytotoxic agents, such as liposomal doxorubicin, topotecan, gemcitabine, or paclitaxel. Overall response rates were generally poor, with a median progression‐free survival of 3–4 months and an overall survival of about 1 year. 150 Bevacizumab was the first drug to provide modest additional benefits. 151 There was hope that the introduction of immunotherapy would improve outcomes; however, unfortunately, checkpoint inhibitors have shown little effectiveness in platinum‐resistant or platinum‐refractory disease. 152 , 153 However, new antibody–drug conjugates have started to provide new options for subgroups of patients. For example, patients with high folate receptor alpha expression saw a 42.3% objective response rate to mirvetuximab soravtansine‐gynx, whereas patients with high HER‐2 expression using gastric cancer scoring criteria saw a 45% objective response rate to trastuzumab deruxtecan. 154 , 155 These more individualized therapies require high expression of the requisite antibody target. Because these drugs are now being investigated in earlier lines of therapy and in rational drug combinations, testing for these markers is increasingly done with initial diagnosis.

Investigational therapies

Several new treatments for HGSOC remain on the horizon. To overcome PARPi resistance, new small‐molecule inhibitors are being studied that target other members of the DNA repair pathways, such as ATR and WEE1. 156 , 157 , 158 , 159 , 160 , 161 , 162 Because repairing DNA damage requires functional cell cycling checkpoints, other approaches focus on targeting cell cycle checkpoints, such as CHK1 and CHK2. 163 , 164 Despite poor initial results, immunotherapy remains an active area of research as well. Therapeutic vaccines using tumor neoantigens, new combinations of immune checkpoint inhibitors, and adoptive cell therapies, such as chimeric antigen receptor T (CAR‐T) cells or chimeric antigen receptor natural killer cells, are all in early‐stage clinical trials and may provide new modalities with more durable treatment responses. 165 , 166 , 167 , 168 , 169 , 170 , 171 In the context of HGSOC, the molecular landscape, including the expression of various antigens and immune evasion mechanisms, offers a unique yet challenging environment for effective CAR‐T cell therapy. It is within this context that the design of CAR‐T cells must be rigorously evaluated, not only in terms of targeting and killing tumor cells but also regarding their ability to function amid the immunosuppressive signals prevalent in the ovarian cancer niche and side effects. The upregulation of immune checkpoints and their ligands, such as programmed death‐ligand 1, contributes to immune evasion by inhibiting T‐cell activation (Figure 4). This duality of immune regulation and suppression not only complicates the therapeutic environment but also emphasizes the necessity for advanced strategies that can overcome these suppression mechanisms. 172 , 173 New immune checkpoint inhibitors targeting the proteins that regulate immune activation are being investigated for their potential to reinvigorate T‐cell responses against HGSOC cells. 174 This raises the question of how to overcome resistance in T‐cell–inflamed tumors (Figure 1). The T‐cell–inflamed HGSOC tumors, in theory, should be ideal candidates for immune checkpoint blockade because their microenvironment is already favorable to immune attack. However, response rates to immune checkpoint inhibitors in ovarian cancer are very low. 175 For patients with T‐cell–inflamed tumors, adoptive cell therapy using autologous TILs offers a potential treatment option. Briefly, TILs are extracted from the patient's own tumor; then, the patient undergoes lymphodepletion on chemotherapy followed by infusion of the expanded TILs and receives systemic IL‐2 treatment to support further TIL expansion within the body. 45 However, challenges like the immunosuppressive TME and low immunogenicity of HGSOC necessitate combinatorial approaches, such as integrating immunotherapy with other treatment modalities, to enhance efficacy. 176

Emerging research and potential innovations in the treatment of HGSOC are strongly geared toward personalized medicine. 177 Advances in genomics and epigenetics have unveiled novel biomarkers and therapeutic targets, paving the way for precision oncology. 178 Epigenetic therapies, including those targeting DNA methylation and histone modification, show promise in overcoming drug resistance and sensitizing tumors to conventional treatments. 179 , 180 , 181 The integration of comprehensive genetic profiling into clinical practice could revolutionize treatment algorithms, enabling tailored therapeutic regimens that reflect the unique molecular landscape of each tumor. For example, relacorilant, an antiglucocorticoid, is currently under clinical investigation in advanced ovarian cancer, aiming to determine its efficacy in combination with standard chemotherapy. In parallel, Artistry‐7 (ClinicalTrials.gov identifier NCT05092360) is a phase 3 trial evaluating nemvaleukin alfa, a selective IL‐2 pathway agonist designed to expand CD8‐positive T cells while sparing regulatory T cells, together with pembrolizumab in patients with platinum‐resistant advanced ovarian cancer. Both trials build on earlier phase data indicating tolerable safety and encouraging immunologic responses, highlighting the potential of these novel therapeutic strategies. However, the above‐mentioned novel therapies for HGSOC, including targeted therapies and immunotherapies, can lead to various side effects. The efficacy of CAR‐T cell treatment has been accompanied by several significant negative outcomes, including cytokine release syndrome, hemophagocytic lymphohistiocytosis/macrophage activation‐like syndrome, neurologic toxicity, and on‐target off‐tumor toxicity. One of the main challenges in cytokine release syndrome is that the stimulation and expansion of CAR‐T cells leads to an excess production of proinflammatory cytokines, resulting in a systemic inflammatory reaction. This occurs in up to 50%–90% of individuals and is characterized by symptoms ranging from fever and fatigue to more severe issues, such as shock, disseminated intravascular coagulation, multiorgan failure, and mortality. 182 , 183 , 184 Monitoring and managing these adverse effects are crucial to maintaining patient QoL. Patient education on potential side effects is essential for informed decision making. Ongoing research is necessary to better understand the full spectrum of side effects and their impact on treatment outcomes.

QUALITY OF LIFE AND SURVIVORSHIP

It’s not just about survival. I want to live in a way that feels meaningful and balanced—physically, emotionally, and socially.

QoL is emerging as a cornerstone in the management of ovarian cancer, transcending survival to address the broader experiences of patients. QoL must be understood as a multidimensional concept encompassing physical, mental, and social health. For numerous patients, maintaining QoL is pivotal. Beyond its immediate relevance to patient well‐being, QoL is increasingly recognized as a key predictive and prognostic factor. Studies like that by Armbrust et al. have demonstrated that lower preoperative QoL scores can predict higher rates of postoperative complications. Furthermore, QoL holds significant prognostic value, with better scores correlating to improved survival outcomes. These findings highlight the necessity of integrating QoL assessments into routine care and using them to guide both therapeutic decisions and supportive interventions. 185 QoL and supportive care are integral components of the treatment strategy for HGSOC. An important element in HGSOC management is the psychological well‐being of patients because it is a crucial aspect of their overall health care, personalized treatment outcomes, and survival. Cancer, in general, can have a significant impact on a person's mental health, with issues like depression, anxiety, and a need for help with coping skills being common. This is particularly relevant for patients who face unique challenges throughout their disease journey. Notably, a primary psychological concern for patients with ovarian cancer is the fear of recurrence. 186 Many survivors report persistently worrying about their cancer returning, which can lead to heightened anxiety and stress.

It is worth mentioning that a restrictive inclusion of patients into investigations on HGSOC or ovarian cancer, in general, has been widely criticized for inappropriate representation of patients worldwide (Figure 2). Data generated from underrepresented groups and sociodemographic domains, such as race and ethnicity, need to be included in studies to draw robust conclusions regarding QoL as well. Keeping the diversity aspect in mind, therefore, we suggest and encourage more clinical research on ovarian cancer with the inclusion of diverse trial participants in which patients from broad and diverse backgrounds are enrolled to enhance external validity and facilitate the translation of results across all relevant groups. 7 , 8 This will also shed light on essential factors that may affect clinical outcomes.

In the management of ovarian cancer, understanding the patient's perspective is indispensable because the disease exerts a profound impact on the physical, psychological, and social dimensions of life (Figure 10). Fear, often described as one of the most dominant emotions, significantly influences patient decision making and their overall treatment experience. This fear is particularly pronounced in the context of HGSOC surgery, which is more extensive compared with surgeries for other malignancies. This highlights the need for early integration of psychological and psycho‐oncologic support to help patients navigate the emotional burden associated with their diagnosis and treatment (Figure 10).

FIGURE 10.

FIGURE 10

Summary box IV. Key points of a cancer survivorship care plan for patients with high‐grade squamous ovarian cancer. PARP indicates poly(adenosine diphosphate‐ribose) polymerase.

The diagnosis of ovarian cancer deeply affects personal relationships. Many patients experience a strain on their partnerships, compounded by a reluctance to openly share their fears to avoid burdening their loved ones. This emotional restraint can create a cycle of isolation and distance, further complicating the patient's psychological well‐being. In addition, geographic separation from family members, such as children living abroad, adds to the sense of loneliness.

Importantly, the shift in focus from active treatment to long‐term survivorship can be disorienting because patients may feel a loss of support. During this critical transition period, it is essential to monitor the mental health of ovarian cancer survivors and provide appropriate interventions. Nurses play a vital role in addressing the psychosocial needs of patients with ovarian cancer. 187 Furthermore, screening for distress and referring patients to appropriate resources, such as counseling or support groups, can help mitigate the psychological burden of the disease. 188 Understanding the importance of psycho‐oncology, the above‐mentioned molecular underpinnings, the mixed phenotypic nature of HGSOC, and the ethnic diversity of patients with HGSOC is crucial for developing targeted therapies and personalized multimodal treatment approaches in centralized therapeutic structures.

Addressing the physical and psychological toll of HGSOC and its treatment involves multidisciplinary approaches, encompassing pain management, nutritional support, and psychosocial interventions. Palliative care, introduced early in the treatment continuum, plays a crucial role in alleviating symptoms, enhancing life quality, and supporting patients and their families through the complexities of the disease trajectory. 189 , 190 , 191

It is worth noting that the time constraints of cancer care for patients and care partners have been highlighted and termed time toxicity. 192 , 193 , 194 This concept is particularly important in the context of advanced metastatic disease, in which life expectancy is limited and time becomes very precious. 195 Future studies should also focus on the aspect of time toxicity from the patients' perspective, particularly in the context of HGSOC therapy. Time toxicity also refers to the temporal burden that patients experience because of medical interventions, including travel time to appointments, waiting periods, and the duration of treatments, such as infusions. This aspect is particularly relevant for patients with HGSOC, who often require extensive and time‐consuming therapies. Understanding the implications of time toxicity is crucial for developing patient‐centered care approaches and improving the overall treatment experience. By investigating the impact of time toxicity on patients with HGSOC, research can optimize health care delivery to enhance both efficiency and patient satisfaction.

CONCLUSION AND FUTURE RESEARCH DIRECTIONS

HGSOC continues to represent a significant challenge in the realm of gynecologic oncology because of its aggressive nature, often late stage diagnosis, and complex treatment landscape. The multifaceted approach encompassing epidemiology, pathogenesis, diagnosis, and treatment strategies highlights the intricate nature of managing this formidable malignancy. We have explored the incidence, prevalence rates, and risk factors of HGSOC. In addition, survival rates exhibit notable variability. The landscape of HGSOC treatment and research is characterized by an ongoing pursuit of innovation and holistic patient management. Through a deeper understanding of the disease's epidemiologic patterns, TME, and comprehensive treatment strategies, the goal remains to transform HGSOC from a perilous diagnosis to a manageable condition with enhanced survival and QoL outcomes.

Patient‐centered care is fundamental for the effective management of ovarian cancer. The heightened fear associated with ovarian cancer surgery necessitates early psychological and psycho‐oncological support to address emotional and psychological challenges. Moreover, integrating QoL as a multidimensional and actionable measure into routine care ensures that treatment strategies align with the lived experiences and priorities of patients. Because of the complexity of patient perspectives and backgrounds, there is an ultimate need for holistic approaches that encompass physical, emotional, and social dimensions. Future research should continue to explore the predictive and prognostic value of QoL and develop interventions that empower patients throughout their journey.

As research advances and new therapies emerge, the combined efforts of health care providers, researchers, and policymakers will be crucial in overcoming the existing challenges and forging a path toward improved prognoses for patients afflicted with HGSOC. Current treatment paradigms for HGSOC involve a combination of surgical cytoreduction, platinum‐based chemotherapy, and emerging targeted therapies, such as PARPi and angiogenesis inhibitors. We advocate new research to clarify whether the above‐mentioned treatment combination or PARPi alone in HRD and BRCA‐positive patients with more limited disease is more adequate. Despite their efficacy, these therapies are often hampered by significant toxicities and the eventual emergence of chemoresistance. Therefore, we encourage more research on the topic of whether chemotherapy in all patients with HGSOC is recommended or whether surgery and PARPi alone would be more favorable to avoid chemotherapy‐related side effects. Immunotherapy and precision oncology represent promising frontiers, with ongoing research into immune checkpoint inhibitors and personalized treatment plans based on genetic profiling demonstrating the potential to revolutionize patient care. In HRD‐negative patients, we suggest new research experiments investigating the potential for checkpoint inhibitors, for which applications still remain elusive. ADCs are emerging as a very promising direction of future clinical research in HGSOC, with results rarely seen in this deadly disease that is usually diagnosed at an advanced stage and in which relapse and resistance to standard‐of‐care treatment are possible. We encourage research to ascertain whether ADCs should be taken into consideration either as frontline therapy or only in disease relapse cases. Furthermore, supportive care strategies are indispensable for managing the physical and psychological toll of HGSOC, improving patient QoL and overall well‐being. We advocate for patient‐centered communication with explicit emphasis on diversity and cultural sensitivity. We strongly suggest new clinical trials on HGSOC with increased representation of underrepresented groups, including, but not limited to, socioeconomic status, geographic regions, ethnicity, age, gender, physical ability, and sexual orientation.

In summary, the treatment strategies for HGSOC are multifaceted and should respect patient diversity, combining current standards involving surgery and chemotherapy with emerging targeted therapies and immunotherapy. Future directions in research and clinical practice should address the importance of including underrepresented groups, paradigm shifts in personalized medicine, innovative therapeutic approaches, and comprehensive supportive care to improve patient outcomes and QoL.

CONFLICT OF INTEREST STATEMENT

Kevin M. Elias reports a patent with Aspira Women's Health Inc. Marcia C. Haigis reports personal/consulting fees from Alixia and Mitoq outside the submitted work. Ioana Elena Braicu reports support for professional activities from AbbVie, AstraZeneca, GlaxoSmithKline, ImmunoGen Inc., Merck, and Myriad; and travel support from AstraZeneca outside the submitted work. The remaining authors declared no conflicts of interest. Jalid Sehouli reports research activities by Roche Pharma, AstraZeneca, Bayer, Clovis Oncology, GlaxoSmithKline, Lilly, Iqvia, Mural, and MSD; receiving honoraries by GlaxoSmithKline, PharmaMar, AstraZeneca, Clovis Oncology, Bayer, Roche Pharma, Vifor Pharma, Hexal AG, Novartis Pharma, Eisai, Esteve Pharmaceuticals, Incyte Biosciences, Phytolife Nutrition, JenaPharm, Kyowa Kirin, Oncoinvent AS, Daiichi, Medtronic Covidien, AMGEN, AbbVie, Corcept Therapeutics, Gilead Sciences, and Myriad; and consulting activities for Merck /Pfizer, PharmaMar, Clovis Oncology, AstraZeneca, Roche Pharma, GlaxoSmithKline, MSD, Eisai, Novocure, Oncoinvent, Intuitive Surgical, Seagen, Bayer Vital, Mundipharma, Sanofi‐Aventis Deutschland GmbH, Immunogen, Tubulis GmbH, Daiichi Sankyo, Bristol Myers Squibb, Karyopharm Therapeutics, and Corcept Therapeutics.

ACKNOWLEDGMENTS

We thank the patients for providing their perspectives. Figures have been created with Biorender. License numbers are provided in the respective figure legends. Pawel Kordowitzki received an “Excellence Initiative‐Research University” (IDUB) program Mobility Grant from Nicolaus Copernicus University.

Kordowitzki P, Lange B, Elias KM, et al. Transforming treatment paradigms: focus on personalized medicine for high‐grade serous ovarian cancer. CA Cancer J Clin. 2025;75(5):436‐460. doi: 10.3322/caac.70008

REFERENCES

  • 1. Narod S. Can advanced‐stage ovarian cancer be cured? Nat Rev Clin Oncol. 2016;13(4):255‐261. doi: 10.1038/nrclinonc.2015.224 [DOI] [PubMed] [Google Scholar]
  • 2. Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin. 2018;68(4):284‐296. doi: 10.3322/caac.21456 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Harter P, Sehouli J, Vergote I, et al. Randomized trial of cytoreductive surgery for relapsed ovarian cancer. N Engl J Med. 2021;385(23):2123‐2131. Erratum in: N Engl J Med. 2022;386(7):704. doi: 10.1056/NEJMoa2103294 [DOI] [PubMed] [Google Scholar]
  • 4. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin. 2024;74(1):12‐49. doi: 10.3322/caac.21820 [DOI] [PubMed] [Google Scholar]
  • 5. Zamwar UM, Anjankar AP. Aetiology, epidemiology, histopathology, classification, detailed evaluation, and treatment of ovarian cancer. Cureus. 2022;14(10):e30561. doi: 10.7759/cureus.30561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Kwong FLA, Kristunas C, Davenport C, et al. Symptom‐triggered testing detects early stage and low volume resectable advanced stage ovarian cancer. Int J Gynecol Cancer. Printed online September 25, 2024. doi: 10.1136/ijgc-2024-005371 [DOI] [PubMed] [Google Scholar]
  • 7. Barry D, Steinberg JR, Towner M, Barber EL, Simon MA, Roque DR. Enrollment of racial and ethnic minoritized groups in gynecologic oncology clinical trials: a review of the scope of the problem, contributing factors, and strategies to improve inclusion. Clin Obstet Gynecol. 2023;66(1):22‐35. doi: 10.1097/GRF.0000000000000765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Sehouli J, Boer J, Brand AH, et al. How to optimize and evaluate diversity in gynecologic cancer clinical trials: statements from the GCIG Barcelona meeting. Int J Gynecol Cancer. 2024;34(11):1677‐1684. doi: 10.1136/ijgc-2024-005982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209‐249. doi: 10.3322/caac.21660 [DOI] [PubMed] [Google Scholar]
  • 10. Bristow RE, Chang J, Ziogas A, Campos B, Chavez LR, Anton‐Culver H. Sociodemographic disparities in advanced ovarian cancer survival and adherence to treatment guidelines. Obstet Gynecol. 2015;125(4):833‐842. doi: 10.1097/AOG.0000000000000643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Armidie TA, Bandera EV, Johnson CE, et al. Diet and survival in Black women with epithelial ovarian cancer. JAMA Netw Open. 2024;7(10):e2440279. doi: 10.1001/jamanetworkopen.2024.40279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.12. National Cancer Institute; Surveillance, Epidemiology, and End Results (SEER) Program . SEER*StatDatabase: Incidence—SEER Research Plus Data with Delay‐Adjustment, 17 Registries, Malignant Only, November 2022 Submission (2000–2020)—Linked To County Attributes—Total U.S., 1969–2021 Counties, National Cancer Institute, Division of Cancer Control and Population Sciences, Surveillance Research Program; 2023.
  • 13. Trabert B, Tworoger SS, O'Brien KM, et al. The risk of ovarian cancer increases with an increase in the lifetime number of ovulatory cycles: an analysis from the Ovarian Cancer Cohort Consortium (OC3). Cancer Res. 2020;80(5):1210‐1218. doi: 10.1158/0008-5472.CAN-19-2850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Webb PM, Jordan SJ. Global epidemiology of epithelial ovarian cancer. Nat Rev Clin Oncol. 2024;21(5):389‐400. doi: 10.1038/s41571-024-00881-3 [DOI] [PubMed] [Google Scholar]
  • 15. Li W, Li L, Wu M. A family pedigree of malignancies associated with BRCA1 pathogenic variants: a reflection of the state of art in China. Hered Cancer Clin Pract. 2019;17(1):26. doi: 10.1186/s13053-019-0126-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Shingala K, Stavros S, Parag S, et al. Racial disparities in survival outcomes of patients with serous epithelial ovarian cancer: a retrospective cohort analysis. Cureus. 2023;15(1):e34389. doi: 10.7759/cureus.34389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Perets R, Wyant GA, Muto KW, et al. Transformation of the fallopian tube secretory epithelium leads to high‐grade serous ovarian cancer in Brca;Tp53;Pten models. Cancer Cell. 2013;24(6):751‐765. doi: 10.1016/j.ccr.2013.10.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Kim J, Park EY, Kim O, et al. Cell origins of high‐grade serous ovarian cancer. Cancers (Basel). 2018;10(11):433. doi: 10.3390/cancers10110433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lheureux S, Braunstein M, Oza AM. Epithelial ovarian cancer: evolution of management in the era of precision medicine. CA Cancer J Clin. 2019;69(4):280‐304. doi: 10.3322/caac.21559 [DOI] [PubMed] [Google Scholar]
  • 20. Lee Y, Miron A, Drapkin R, et al. A candidate precursor to serous carcinoma that originates in the distal fallopian tube. J Pathol. 2007;211(1):26‐35. doi: 10.1002/path.2091 [DOI] [PubMed] [Google Scholar]
  • 21. Verhaak RG, Tamayo P, Yang JY, et al. Prognostically relevant gene signatures of high‐grade serous ovarian carcinoma. J Clin Invest. 2013;123(1):517‐525. doi: 10.1172/JCI65833 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Visvanathan K, Vang R, Shaw P, et al. Diagnosis of serous tubal intraepithelial carcinoma based on morphologic and immunohistochemical features: a reproducibility study. Am J Surg Pathol. 2011;35(12):1766‐1775. doi: 10.1097/PAS.0b013e31822f58bc [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kuhn E, Kurman RJ, Sehdev AS, Shih IM. Ki‐67 labeling index as an adjunct in the diagnosis of serous tubal intraepithelial carcinoma. Int J Gynecol Pathol. 2012;31(5):416‐422. doi: 10.1097/PGP.0b013e31824cbeb4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Medeiros F, Muto MG, Lee Y, et al. The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome. Am J Surg Pathol. 2006;30(2):230‐236. doi: 10.1097/01.pas.0000180854.28831.77 [DOI] [PubMed] [Google Scholar]
  • 25. Wu RC, Wang P, Lin SF, et al. Genomic landscape and evolutionary trajectories of ovarian cancer precursor lesions. J Pathol. 2019;248(1):41‐50. doi: 10.1002/path.5219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Negri S, Fisch C, de Hullu JA, et al. Diagnosis and management of isolated serous tubal intraepithelial carcinoma: a qualitative focus group study. BJOG. 2024;131(13):1851‐1861. doi: 10.1111/1471-0528.17919 [DOI] [PubMed] [Google Scholar]
  • 27. Shih IM, Wang Y, Wang TL. The origin of ovarian cancer species and precancerous landscape. Am J Pathol. 2021;191(1):26‐39. doi: 10.1016/j.ajpath.2020.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Morice P, Gouy S, Leary A. Mucinous ovarian carcinoma. N Engl J Med. 2019;380(13):1256‐1266. doi: 10.1056/NEJMra1813254 [DOI] [PubMed] [Google Scholar]
  • 29. Fitzgerald RC, Antoniou AC, Fruk L, Rosenfeld N. The future of early cancer detection. Nat Med. 2022;28(4):666‐677. doi: 10.1038/s41591-022-01746-x [DOI] [PubMed] [Google Scholar]
  • 30. Tee XR, Hazard E, Latorre‐Esteves E, et al. Increased TP53 somatic evolution in peritoneal washes of individuals with BRCA1 germline mutations. Gynecol Oncol. 2024;190:18‐27. doi: 10.1016/j.ygyno.2024.07.690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Hughley R, Karlic R, Joshi H, Turnbull C, Foulkes WD, Polak P. Etiologic index—a case‐only measure of BRCA1/2‐associated cancer risk. N Engl J Med. 2020;383(3):286‐288. doi: 10.1056/NEJMc1913988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Valenza C, Mongillo M, Gigli F, et al. Germline BRCA pathogenic variants in patients with ovarian cancer and post‐poly (ADP‐ribose) polymerase inhibitor myeloid neoplasms. ESMO Open. 2024;9(9):103685. doi: 10.1016/j.esmoop.2024.103685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Del Castillo Falconi VM, Díaz‐Chávez J, Torres‐Arciga K, et al. Expression of DNA methyltransferase 3B isoforms is associated with DNA satellite 2 hypomethylation and clinical prognosis in advanced high‐grade serous ovarian carcinoma. Int J Mol Sci. 2022;23(21):12759. doi: 10.3390/ijms232112759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Machino H, Dozen A, Konaka M, et al. Integrative analysis reveals early epigenetic alterations in high‐grade serous ovarian carcinomas. Exp Mol Med. 2023;55(10):2205‐2219. doi: 10.1038/s12276-023-01090-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Vergara D, Merlot B, Lucot JP, et al. Epithelial‐mesenchymal transition in ovarian cancer. Cancer Lett. 2010;291(1):59‐66. doi: 10.1016/j.canlet.2009.09.017 [DOI] [PubMed] [Google Scholar]
  • 36. Lisio MA, Fu L, Goyeneche A, Gao ZH, Telleria C. High‐grade serous ovarian cancer: basic sciences, clinical and therapeutic standpoints. Int J Mol Sci. 2019;20(4):952. doi: 10.3390/ijms20040952 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Wilczyński B, Dąbrowska A, Kulbacka J, Baczyńska D. Chemoresistance and the tumor microenvironment: the critical role of cell‐cell communication. Cell Commun Signal. 2024;22(1):486. doi: 10.1186/s12964-024-01857-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Liu L, Zou C, Shen J, et al. MUL1 identified as mitochondria‐linked biomarker promoting cisplatin resistance in OC cells. Gene. 2024;930:148841. doi: 10.1016/j.gene.2024.148841 [DOI] [PubMed] [Google Scholar]
  • 39. Rohaan MW, Borch TH, van den Berg JH, et al. Tumor‐infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med. 2022;387(23):2113‐2125. doi: 10.1056/NEJMoa2210233 [DOI] [PubMed] [Google Scholar]
  • 40. Leidner R, Sanjuan Silva N, Huang H, et al. Neoantigen T‐cell receptor gene therapy in pancreatic cancer. N Engl J Med. 2022;386(22):2112‐2119. doi: 10.1056/NEJMoa2119662 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Izar B, Tirosh I, Stover EH, et al. A single‐cell landscape of high‐grade serous ovarian cancer. Nat Med. 2020;26(8):1271‐1279. doi: 10.1038/s41591-020-0926-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Motohara T, Motohara T, Masuda K, et al. An evolving story of the metastatic voyage of ovarian cancer cells: cellular and molecular orchestration of the adipose‐rich metastatic microenvironment. Oncogene. 2019;38(16):2885‐2898. doi: 10.1038/s41388-018-0637-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Garg P, Ramisetty SK, Raghu Subbalakshmi A, et al. Gynecological cancer tumor microenvironment: unveiling cellular complexity and therapeutic potential. Biochem Pharmacol. 2024;229:116498. doi: 10.1016/j.bcp.2024.116498 [DOI] [PubMed] [Google Scholar]
  • 44. Sleeboom JJF, van Tienderen GS, Schenke‐Layland K, van der Laan LJW, Khalil AA, Verstegen MMA. The extracellular matrix as hallmark of cancer and metastasis: from biomechanics to therapeutic targets. Sci Transl Med. 2024;16(728):eadg3840. doi: 10.1126/scitranslmed.adg3840 [DOI] [PubMed] [Google Scholar]
  • 45. Kandalaft LE, Dangaj Laniti D, Coukos G. Immunobiology of high‐grade serous ovarian cancer: lessons for clinical translation. Nat Rev Cancer. 2022;22(11):640‐656. doi: 10.1038/s41568-022-00503-z [DOI] [PubMed] [Google Scholar]
  • 46. Xu S, Liu Y, Yang S, et al. FXN targeting induces cell death in ovarian cancer stem‐like cells through PRDX3‐mediated oxidative stress. iScience. 2024;27(8):110506. doi: 10.1016/j.isci.2024.110506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Bhattacharya R, Ghosh A, Mukhopadhyay S. High‐grade serous ovarian carcinoma, the “Achiles' hill” for clinicians and molecular biologists: a molecular insight. Mol Biol Rep. 2023;50(11):9511‐9519. doi: 10.1007/s11033-023-08760-3 [DOI] [PubMed] [Google Scholar]
  • 48. Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3(6):401‐410. doi: 10.1038/nrc1093 [DOI] [PubMed] [Google Scholar]
  • 49. Spinelli JB, Haigis MC. The multifaceted contributions of mitochondria to cellular metabolism. Nat Cell Biol. 2018;20(7):745‐754. doi: 10.1038/s41556-018-0124-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Scharping NE, Menk AV, Moreci RS, et al. The tumor microenvironment represses T cell mitochondrial biogenesis to drive intratumoral T cell metabolic insufficiency and dysfunction. Immunity. 2016;45(3):701‐703. doi: 10.1016/j.immuni.2016.08.009 [DOI] [PubMed] [Google Scholar]
  • 51. Fantone S, Piani F, Olivieri F, et al. Role of SLC7A11/xCT in ovarian cancer. Int J Mol Sci. 2024;25(1):587. doi: 10.3390/ijms25010587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Zhang L, Conejo‐Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348(3):203‐213. doi: 10.1056/NEJMoa020177 [DOI] [PubMed] [Google Scholar]
  • 53. Adams SF, Levine DA, Cadungog MG, et al. Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer. 2009;115(13):2891‐2902. doi: 10.1002/cncr.24317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Hornburg M, Desbois M, Lu S, et al. Single‐cell dissection of cellular components and interactions shaping the tumor immune phenotypes in ovarian cancer. Cancer Cell. 2021;39(7):928‐944.e6. doi: 10.1016/j.ccell.2021.04.004 [DOI] [PubMed] [Google Scholar]
  • 55. Olalekan S, Xie B, Back R, Eckart H, Basu A. Characterizing the tumor microenvironment of metastatic ovarian cancer by single‐cell transcriptomics. Cell Rep. 2021;35(8):109165. doi: 10.1016/j.celrep.2021.109165 [DOI] [PubMed] [Google Scholar]
  • 56. Zhang AW, McPherson A, Milne K, et al. Interfaces of malignant and immunologic clonal dynamics in ovarian cancer. Cell. 2018;173(7):1755‐1769.e22. doi: 10.1016/j.cell.2018.03.073 [DOI] [PubMed] [Google Scholar]
  • 57. Sainero‐Alcolado L, Liaño‐Pons J, Ruiz‐Pérez MV, Arsenian‐Henriksson M. Targeting mitochondrial metabolism for precision medicine in cancer. Cell Death Differ. 2022;29(7):1304‐1317. doi: 10.1038/s41418-022-01022-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Horibe S, Ishikawa K, Nakada K, et al. Mitochondrial DNA mutations are involved in the acquisition of cisplatin resistance in human lung cancer A549 cells. Oncol Rep. 2022;47(2):32. doi: 10.3892/or.2021.8243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Bustos G, Cruz P, Lovy A, Cárdenas C. Endoplasmic reticulum‐mitochondria calcium communication and the regulation of mitochondrial metabolism in cancer: a novel potential target. Front Oncol. 2017;7:199. doi: 10.3389/fonc.2017.00199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Zhang S, Dolgalev I, Zhang T, Ran H, Levine DA, Neel BG. Both fallopian tube and ovarian surface epithelium are cells‐of‐origin for high‐grade serous ovarian carcinoma. Nat Commun. 2019;10(1):5367. doi: 10.1038/s41467-019-13116-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Gjorgoska M, Rižner TL. From fallopian tube epithelium to high‐grade serous ovarian cancer: a single‐cell resolution review of sex steroid hormone signaling. Prog Lipid Res. 2024;11:101302. doi: 10.1016/j.plipres.2024.101302 [DOI] [PubMed] [Google Scholar]
  • 62. Byun JM, Cho HJ, Lee DS, et al. Frequency of serous tubal intraepithelial carcinoma (STIC) in patients with high grade serous ovarian cancer. Taiwan J Obstet Gynecol. 2023;62(1):107‐111. doi: 10.1016/j.tjog.2022.09.006 [DOI] [PubMed] [Google Scholar]
  • 63. World Health Organization (WHO) Classification of Tumours Editorial Board, eds.Female Genital Tumours. WHO Classification of Tumours. 5th ed. Volume 4. International Agency for Research on Cancer; 2020. [Google Scholar]
  • 64. Feng Y, Miao F, Li Y, Li M, Cao Y. Validating the 2023 FIGO staging system: a nomogram for endometrioid endometrial cancer and adenocarcinoma. Cancer Med. 2024;13(10):e7216. doi: 10.1002/cam4.7216 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Darb‐Esfahani S, Kolaschinski I, Trillsch F, et al. Morphology and tumour‐infiltrating lymphocytes in high‐stage, high‐grade serous ovarian carcinoma correlated with long‐term survival. Histopathology. 2018;73(6):1002‐1012. doi: 10.1111/his.13711 [DOI] [PubMed] [Google Scholar]
  • 66. Ahn B, Moon D, Kim HS, et al. Histopathologic image‐based deep learning classifier for predicting platinum‐based treatment responses in high‐grade serous ovarian cancer. Nat Commun. 2024;15(1):4253. doi: 10.1038/s41467-024-48667-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Momeni‐Boroujeni A, Song H, Irshaid L, Strickland S, Parra‐Herran C, Busca A. Outcome‐based validation of confluent/expansile versus infiltrative pattern assessment and growth‐based grading in ovarian mucinous carcinoma. Am J Surg Pathol. 2022;46(9):1250‐1259. doi: 10.1097/PAS.0000000000001895 [DOI] [PubMed] [Google Scholar]
  • 68. Parra‐Herran C, Bassiouny D, Vicus D, et al. FIGO versus Silverberg grading systems in ovarian endometrioid carcinoma: a comparative prognostic analysis. Am J Surg Pathol. 2019;43(2):161‐167. doi: 10.1097/PAS.0000000000001160 [DOI] [PubMed] [Google Scholar]
  • 69. Cancer Genome Atlas Research Network . Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(7353):609‐615. Erratum in: Nature. 2012;490(7419):298. doi: 10.1038/nature10166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Murakami R, Matsumura N, Mandai M, et al. Establishment of a novel histopathological classification of high‐grade serous ovarian carcinoma correlated with prognostically distinct gene expression subtypes. Am J Pathol. 2016;186(5):1103‐1113. doi: 10.1016/j.ajpath.2015.12.029 [DOI] [PubMed] [Google Scholar]
  • 71. Searle SD, Mitnitski A, Gahbauer EA, Gill TM, Rockwood K. A standard procedure for creating a frailty index. BMC Geriatr. 2008;8(1):24. doi: 10.1186/1471-2318-8-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Inci MG, Anders L, Woopen H, et al. Frailty Index for prediction of surgical outcome in ovarian cancer: results of a prospective study. Gynecol Oncol. 2021;161(2):396‐401. doi: 10.1016/j.ygyno.2021.02.012 [DOI] [PubMed] [Google Scholar]
  • 73. Schneider S, Armbrust R, Spies C, du Bois A, Sehouli J. Prehabilitation programs and ERAS protocols in gynecological oncology: a comprehensive review. Arch Gynecol Obstet. 2020;301(2):315‐326. doi: 10.1007/s00404-019-05321-7 [DOI] [PubMed] [Google Scholar]
  • 74. Inci MG, Sehouli J, Schnura E, et al. The KORE‐INNOVATION trial, a prospective controlled multi‐site clinical study to implement and assess the effects of an innovative peri‐operative care pathway for patients with ovarian cancer: rationale, methods and trial design. Int J Gynecol Cancer. 2023;33(8):1304‐1309. doi: 10.1136/ijgc-2023-004531 [DOI] [PubMed] [Google Scholar]
  • 75. Sehouli J, Heise K, Richter R, Woopen H, Anders L, Inci MG. Correction to: Preoperative quality of life as prediction for severe postoperative complications in gynecological cancer surgery: results of a prospective study. Arch Gynecol Obstet. 2022;306(3):925. Erratum for: Arch Gynecol Obstet. 2021;303(4):1057‐1063. doi: 10.1007/s00404-021-06333-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Oskay‐Özcelik G, Alavi S, Richter R, et al. Expression III: patients' expectations and preferences regarding physician‐patient relationship and clinical management‐results of the international NOGGO/ENGOT‐ov4‐GCIG study in 1830 ovarian cancer patients from European countries. Ann Oncol. 2018;29(4):910‐916. doi: 10.1093/annonc/mdy037 [DOI] [PubMed] [Google Scholar]
  • 77. Rohr I, Alavi S, Richter R, et al. Expectations and preferences of patients with primary and relapsed ovarian cancer to maintenance therapy: an NOGGO/ENGOT‐ov22 and GCIG survey (Expression IV). Int J Gynecol Cancer. 2020;30(4):509‐514. doi: 10.1136/ijgc-2019-000892 [DOI] [PubMed] [Google Scholar]
  • 78. Hasenburg A, Plett H, Krämer B, et al. The effect of fertility‐sparing surgery on sexuality and global quality of life in women with malignant ovarian germ cell and sex cord stromal tumors: an analysis of the CORSETT database of the AGO Study Group. Arch Gynecol Obstet. 2021;304(6):1541‐1549. doi: 10.1007/s00404-021-06019-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Frangou E, Bertelli G, Love S, et al. OVPSYCH2: a randomized controlled trial of psychological support versus standard of care following chemotherapy for ovarian cancer. Gynecol Oncol. 2021;162(2):431‐439. doi: 10.1016/j.ygyno.2021.05.024 [DOI] [PubMed] [Google Scholar]
  • 80. Lakhiani A, Cummins C, Kumar S, et al. Analysis of anxiety, depression and fear of progression at 12 months post‐cytoreductive surgery in the SOCQER‐2 (Surgery in Ovarian Cancer‐Quality of Life Evaluation Research) prospective, international, multicentre study. Cancers (Basel). 2023;16(1):75. doi: 10.3390/cancers16010075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Fotopoulou C, Bowen R, Manchanda R, et al. Current practices for the management of advanced high‐grade epithelial ovarian cancer in the UK: OC‐NOW survey (2023). Future Oncol. 2025;22(1):95‐103. doi: 10.1080/14796694.2024.2424153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Harter P, Sehouli J, Lorusso D, et al. A randomized trial of lymphadenectomy in patients with advanced ovarian neoplasms. N Engl J Med. 2019;380(9):822‐832. doi: 10.1056/NEJMoa1808424 [DOI] [PubMed] [Google Scholar]
  • 83. Maggioni A, Benedetti Panici P, Dell'Anna T, et al. Randomised study of systematic lymphadenectomy in patients with epithelial ovarian cancer macroscopically confined to the pelvis. Br J Cancer. 2006;95(6):699‐704. doi: 10.1038/sj.bjc.6603323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Ditto A, Martinelli F, Reato C, et al. Systematic para‐aortic and pelvic lymphadenectomy in early stage epithelial ovarian cancer: a prospective study. Ann Surg Oncol. 2012;19(12):3849‐3855. doi: 10.1245/s10434-012-2439-7 [DOI] [PubMed] [Google Scholar]
  • 85. Timmers PJ, Zwinderman K, Coens C, Vergote I, Trimbos JB. Lymph node sampling and taking of blind biopsies are important elements of the surgical staging of early ovarian cancer. Int J Gynecol Cancer. 2010;20(7):1142‐1147. doi: 10.1111/igc.0b013e3181ef8e03 [DOI] [PubMed] [Google Scholar]
  • 86. Chase DM, Mahajan A, Scott DA, Hawkins N, Kalilani L. The impact of varying levels of residual disease following cytoreductive surgery on survival outcomes in patients with ovarian cancer: a meta‐analysis. BMC Womens Health. 2024;24(1):179. doi: 10.1186/s12905-024-02977-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Bryant A, Hiu S, Kunonga PT, et al. Impact of residual disease as a prognostic factor for survival in women with advanced epithelial ovarian cancer after primary surgery. Cochrane Database Syst Rev. 2022;9(9):CD015048. doi: 10.1002/14651858.CD015048.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Fagotti A, Vizzielli G, De Iaco P, et al. A multicentric trial (Olympia‐MITO 13) on the accuracy of laparoscopy to assess peritoneal spread in ovarian cancer. Am J Obstet Gynecol.2013;209(5):462e1‐462e11. doi: 10.1016/j.ajog.2013.07.016 [DOI] [PubMed] [Google Scholar]
  • 89. Hansen JM, Sood AK, Coleman RL, et al. Concordance of a laparoscopic scoring algorithm with primary surgery findings in advanced stage ovarian cancer. Gynecol Oncol. 2018;151(3):428‐432. doi: 10.1016/j.ygyno.2018.10.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Vergote I, Trope CG, Amant F, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med. 2010;363(10):943‐953. doi: 10.1056/NEJMoa0908806 [DOI] [PubMed] [Google Scholar]
  • 91. Kehoe S, Hook J, Nankivell M, et al. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): an open‐label, randomised, controlled, non‐inferiority trial. Lancet. 2015;386(9990):249‐257. doi: 10.1016/S0140-6736(14)62223-6 [DOI] [PubMed] [Google Scholar]
  • 92. Fagotti A, Ferrandina MG, Vizzielli G, et al. Randomized trial of primary debulking surgery versus neoadjuvant chemotherapy for advanced epithelial ovarian cancer (SCORPION‐NCT01461850). Int J Gynecol Cancer. 2020;30(11):1657‐1664. doi: 10.1136/ijgc-2020-001640 [DOI] [PubMed] [Google Scholar]
  • 93. Wright AA, Bohlke K, Armstrong DK, 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. 2016;143(1):3‐15. doi: 10.1016/j.ygyno.2016.05.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Coleridge SL, Bryant A, Kehoe S, Morrison J. Neoadjuvant chemotherapy before surgery versus surgery followed by chemotherapy for initial treatment in advanced ovarian epithelial cancer. Cochrane Database Syst Rev. 2021;7(7):CD005343. doi: 10.1002/14651858.CD005343.pub6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. van Driel WJ, Koole SN, Sikorska K, et al. Hyperthermic intraperitoneal chemotherapy in ovarian cancer. N Engl J Med. 2018;378(3):230‐240. doi: 10.1056/NEJMoa1708618 [DOI] [PubMed] [Google Scholar]
  • 96. Aronson SL, Lopez‐Yurda M, Koole SN, et al. Cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy in patients with advanced ovarian cancer (OVHIPEC‐1): final survival analysis of a randomised, controlled, phase 3 trial. Lancet Oncol. 2023;24(10):1109‐1118. doi: 10.1016/S1470-2045(23)00396-0 [DOI] [PubMed] [Google Scholar]
  • 97. Lee JY, Lee YJ, Son JH, et al. Hyperthermic intraperitoneal chemotherapy after interval cytoreductive surgery for patients with advanced‐stage ovarian cancer who had received neoadjuvant chemotherapy. JAMA Surg. 2023;158(11):1133‐1140. doi: 10.1001/jamasurg.2023.3944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Abdul‐Aziz S, Bhattarai N, Vale L, Sagoo GS, Mukhopadhyay A. Challenges and opportunities to address the emerging burden of targeted therapies in ovarian cancer. Gynecol Oncol Rep. 2025;57:101680. doi: 10.1016/j.gore.2025.101680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Chandra A, Pius C, Nabeel M, et al. Ovarian cancer: current status and strategies for improving therapeutic outcomes. Cancer Med. 2019;8(16):7018‐7031. doi: 10.1002/cam4.2560 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Kurnit KC, Fleming GF, Lengyel E. Updates and new options in advanced epithelial ovarian cancer treatment. Obstet Gynecol. 2021;137(1):108‐121. doi: 10.1097/AOG.0000000000004173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Bisch SP, Wells T, Gramlich L, et al. Enhanced recovery after surgery (ERAS) in gynecologic oncology: system‐wide implementation and audit leads to improved value and patient outcomes. Gynecol Oncol. 2018;151(1):117‐123. doi: 10.1016/j.ygyno.2018.08.007 [DOI] [PubMed] [Google Scholar]
  • 102. Kwon JS, McAlpine JN, Hanley GE, et al. Costs and benefits of opportunistic salpingectomy as an ovarian cancer prevention strategy. Obstet Gynecol. 2015;125(2):338‐345. doi: 10.1097/AOG.0000000000000630 [DOI] [PubMed] [Google Scholar]
  • 103. Urban RR, He H, Alfonso‐Cristancho R, Hardesty MM, Goff BA. The cost of initial care for Medicare patients with advanced ovarian cancer. J Natl Compr Canc Netw. 2016;14(4):429‐437. doi: 10.6004/jnccn.2016.0049 [DOI] [PubMed] [Google Scholar]
  • 104. Dholakia J, Cohn DE, Straughn JM, Dilley SE. Prehabilitation for medically frail patients undergoing surgery for epithelial ovarian cancer: a cost‐effectiveness analysis. J Gynecol Oncol. 2021;32(6):e92. doi: 10.3802/jgo.2021.32.e92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Temkin SM, Smeltzer MP, Dawkins MD, et al. Improving the quality of care for patients with advanced epithelial ovarian cancer: program components, implementation barriers, and recommendations. Cancer. 2022;128(4):654‐664. doi: 10.1002/cncr.34023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Smye SW, Frangi AF. Interdisciplinary research: shaping the healthcare of the future. Future Healthc J. 2021;8(2):e218‐e223. doi: 10.7861/fhj.2021-0025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Falandry C, Rousseau F, Mouret‐Reynier MA, et al. Efficacy and safety of first‐line single‐agent carboplatin vs carboplatin plus paclitaxel for vulnerable older adult women with ovarian cancer: a GINECO/GCIG randomized clinical trial. JAMA Oncol. 2021;7(6):853‐861. doi: 10.1001/jamaoncol.2021.0696 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Vasey PA, Jayson GC, Gordon A, et al. Phase III randomized trial of docetaxel‐carboplatin versus paclitaxel‐carboplatin as first‐line chemotherapy for ovarian carcinoma. J Natl Cancer Inst. 2004;96(22):1682‐1691. doi: 10.1093/jnci/djh323 [DOI] [PubMed] [Google Scholar]
  • 109. Pignata S, Scambia G, Ferrandina G, et al. Carboplatin plus paclitaxel versus carboplatin plus pegylated liposomal doxorubicin as first‐line treatment for patients with ovarian cancer: the MITO‐2 randomized phase III trial. J Clin Oncol. 2011;29(27):3628‐3635. doi: 10.1200/JCO.2010.33.8566 [DOI] [PubMed] [Google Scholar]
  • 110. Bookman MA, Brady MF, McGuire WP, et al. Evaluation of new platinum‐based treatment regimens in advanced‐stage ovarian cancer: a phase III trial of the Gynecologic Cancer Intergroup. J Clin Oncol. 2009;27(9):1419‐1425. doi: 10.1200/JCO.2008.19.1684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Fiegl H, Schnaiter S, Reimer DU, et al. BRCA loss of function including BRCA1 DNA‐methylation, but not BRCA‐unrelated homologous recombination deficiency, is associated with platinum hypersensitivity in high‐grade ovarian cancer. Clin Epigenetics. 2024;16(1):171. doi: 10.1186/s13148-024-01781-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Katsumata N, Yasuda M, Isonishi S, et al. Long‐term results of dose‐dense paclitaxel and carboplatin versus conventional paclitaxel and carboplatin for treatment of advanced epithelial ovarian, fallopian tube, or primary peritoneal cancer (JGOG 3016): a randomised, controlled, open‐label trial. Lancet Oncol. 2013;14(10):1020‐1026. doi: 10.1016/S1470-2045(13)70363-2 [DOI] [PubMed] [Google Scholar]
  • 113. Chan JK, Brady MF, Penson RT, et al. Weekly vs. every‐3‐week paclitaxel and carboplatin for ovarian cancer. N Engl J Med. 2016;374(8):738‐748. doi: 10.1056/NEJMoa1505067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Clamp AR, James EC, McNeish IA, et al. Weekly dose‐dense chemotherapy in first‐line epithelial ovarian, fallopian tube, or primary peritoneal cancer treatment (ICON8): overall survival results from an open‐label, randomised, controlled, phase 3 trial. Lancet Oncol. 2022;23(7):919‐930. doi: 10.1016/S1470-2045(22)00283-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Pignata S, Lorusso D, Joly F, et al. Carboplatin‐based doublet plus bevacizumab beyond progression versus carboplatin‐based doublet alone in patients with platinum‐sensitive ovarian cancer: a randomised, phase 3 trial. Lancet Oncol. 2021;22(2):267‐276. doi: 10.1016/S1470-2045(20)30637-9 [DOI] [PubMed] [Google Scholar]
  • 116. Pignata S, Scambia G, Katsaros D, et al. Carboplatin plus paclitaxel once a week versus every 3 weeks in patients with advanced ovarian cancer (MITO‐7): a randomised, multicentre, open‐label, phase 3 trial. Lancet Oncol. 2014;15(4):396‐405. doi: 10.1016/S1470-2045(14)70049-X [DOI] [PubMed] [Google Scholar]
  • 117. Armstrong DK, Bundy B, Wenzel L, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med. 2006;354(1):34‐43. doi: 10.1056/NEJMoa052985 [DOI] [PubMed] [Google Scholar]
  • 118. Walker JL, Brady MF, Wenzel L, et al. Randomized trial of intravenous versus intraperitoneal chemotherapy plus bevacizumab in advanced ovarian carcinoma: an NRG Oncology/Gynecologic Oncology Group study. J Clin Oncol. 2019;37(16):1380‐1390. doi: 10.1200/JCO.18.01568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Oza AM, Cook AD, Pfisterer J, 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. 2015;16(8):928‐936. doi: 10.1016/S1470-2045(15)00086-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Tewari KS, Burger RA, Enserro D, et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol. 2019;37(26):2317‐2328. doi: 10.1200/JCO.19.01009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Ferriss JS, Java JJ, Bookman MA, et al. Ascites predicts treatment benefit of bevacizumab in front‐line therapy of advanced epithelial ovarian, fallopian tube and peritoneal cancers: an NRG Oncology/GOG study. Gynecol Oncol. 2015;139(1):17‐22. doi: 10.1016/j.ygyno.2015.07.103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Heitz F, Marth C, Henry S, et al. AGO‐OVAR 28/ENGOT‐ov57. Niraparib alone versus niraparib in combination with bevacizumab in patients with carboplatin‐taxane‐based chemotherapy in advanced ovarian cancer: a multicenter randomized phase III trial. Int J Gynecol Cancer. 2023;33(12):1966‐1969. doi: 10.1136/ijgc-2023-004944 [DOI] [PubMed] [Google Scholar]
  • 123. Braicu EI, Pietzner K, Zocholl D, et al. First trial of chemotherapy de‐escalation in ovarian cancer (OC): a phase III randomized, open label study of niraparib maintenance after carboplatin and paclitaxel in patients with optimally debulked advanced (homologous recombination deficient) HRD high‐grade ovarian cancer in first line therapy (N‐PLUS/NOGGO‐ov53/ENGOT‐ov62) [abstract]. J Clin Oncol. 2023;41(16 suppl):TPS5620. doi: 10.1200/JCO.2023.41.16_suppl.TPS5620 [DOI] [Google Scholar]
  • 124. Gonzalez‐Martin A, Pothuri B, Vergote I, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019;381(25):2391‐2402. doi: 10.1056/NEJMoa1910962 [DOI] [PubMed] [Google Scholar]
  • 125. Banerjee S, Moore KN, Colombo N, et al. Maintenance olaparib for patients with newly diagnosed advanced ovarian cancer and a BRCA mutation (SOLO1/GOG 3004): 5‐year follow‐up of a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Oncol. 2021;22(12):1721‐1731. doi: 10.1016/S1470-2045(21)00531-3 [DOI] [PubMed] [Google Scholar]
  • 126. Monk BJ, Parkinson C, Lim MC, et al. A randomized, phase III trial to evaluate rucaparib monotherapy as maintenance treatment in patients with newly diagnosed ovarian cancer (ATHENA‐MONO/GOG‐3020/ENGOT‐ov45). J Clin Oncol. 2022;40(34):3952‐3964. doi: 10.1200/JCO.22.01003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Ray‐Coquard I, Leary A, Pignata S, et al. Olaparib plus bevacizumab first‐line maintenance in ovarian cancer: final overall survival results from the PAOLA‐1/ENGOT‐ov25 trial. Ann Oncol. 2023;34(8):681‐692. doi: 10.1016/j.annonc.2023.05.005 [DOI] [PubMed] [Google Scholar]
  • 128. Ray‐Coquard I, Pautier P, Pignata S, et al. Olaparib plus bevacizumab as first‐line maintenance in ovarian cancer. N Engl J Med. 2019;381(25):2416‐2428. doi: 10.1056/NEJMoa1911361 [DOI] [PubMed] [Google Scholar]
  • 129. Gulia S, Kannan S, Ghosh J, Rath S, Maheshwari A, Gupta S. Maintenance therapy with a poly(ADP‐ribose) polymerase inhibitor in patients with newly diagnosed advanced epithelial ovarian cancer: individual patient data and trial‐level meta‐analysis. ESMO Open. 2022;7(5):100558. doi: 10.1016/j.esmoop.2022.100558 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Tinker AV, Altman AD, Bernardini MQ, et al. A pan‐Canadian consensus statement on first‐line PARP inhibitor maintenance for advanced, high‐grade serous and endometrioid tubal, ovarian, and primary peritoneal cancers. Curr Oncol. 2022;29(6):4354‐4369. doi: 10.3390/curroncol29060348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Li N, Zhu J, Yin R, et al. Treatment with niraparib maintenance therapy in patients with newly diagnosed advanced ovarian cancer: a phase 3 randomized clinical trial. JAMA Oncol. 2023;9(9):1230‐1237. doi: 10.1001/jamaoncol.2023.2283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Rustin GJ, van der Burg ME, Griffin CL, et al. Early versus delayed treatment of relapsed ovarian cancer (MRC OV05/EORTC 55955): a randomised trial. Lancet. 2010;376(9747):1155‐1163. doi: 10.1016/S0140-6736(10)61268-8 [DOI] [PubMed] [Google Scholar]
  • 133. Taliento C, Morciano G, Nero C, et al. Circulating tumor DNA as a biomarker for predicting progression‐free survival and overall survival in patients with epithelial ovarian cancer: a systematic review and meta‐analysis. Int J Gynecol Cancer. 2024;34(6):906‐918. doi: 10.1136/ijgc-2024-005313 [DOI] [PubMed] [Google Scholar]
  • 134. Shi T, Zhu J, Feng Y, et al. Secondary cytoreduction followed by chemotherapy versus chemotherapy alone in platinum‐sensitive relapsed ovarian cancer (SOC‐1): a multicentre, open‐label, randomised, phase 3 trial. Lancet Oncol. 2021;22(4):439‐449. doi: 10.1016/S1470-2045(21)00006-1 [DOI] [PubMed] [Google Scholar]
  • 135. Falcone F, Scambia G, Benedetti Panici P, et al. Tertiary cytoreductive surgery in recurrent epithelial ovarian cancer: a multicentre MITO retrospective study. Gynecol Oncol. 2017;147(1):66‐72. doi: 10.1016/j.ygyno.2017.07.008 [DOI] [PubMed] [Google Scholar]
  • 136. Manning‐Geist BL, Chi DS, Long Roche K, et al. Tertiary cytoreduction for recurrent ovarian carcinoma: an updated and expanded analysis. Gynecol Oncol. 2021;162(2):345‐352. doi: 10.1016/j.ygyno.2021.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Armstrong DK, Coleman RL, Penson RT. Emerging therapeutic options for platinum‐sensitive ovarian cancer patients. Clin Adv Hematol Oncol. 2011;9(4 suppl 5):1‐16. [PubMed] [Google Scholar]
  • 138. Stuart GC, Kitchener H, Bacon M, et al. 2010 Gynecologic Cancer InterGroup (GCIG) consensus statement on clinical trials in ovarian cancer: report from the Fourth Ovarian Cancer Consensus Conference. Int J Gynecol Cancer. 2010;21(4):750‐755. doi: 10.1097/IGC.0b013e31821b2568 [DOI] [PubMed] [Google Scholar]
  • 139. Pfisterer J, Plante M, Vergote I, et al. Gemcitabine plus carboplatin compared with carboplatin in patients with platinum‐sensitive recurrent ovarian cancer: an intergroup trial of the AGO‐OVAR, the NCIC CTG, and the EORTC GCG. J Clin Oncol. 2006;24(29):4699‐4707. doi: 10.1200/JCO.2006.06.0913 [DOI] [PubMed] [Google Scholar]
  • 140. Wagner U, Marth C, Largillier R, et al. Final overall survival results of phase III GCIG CALYPSO trial of pegylated liposomal doxorubicin and carboplatin vs paclitaxel and carboplatin in platinum‐sensitive ovarian cancer patients. Br J Cancer. 2012;107(4):588‐591. doi: 10.1038/bjc.2012.307 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Sehouli J, Chekerov R, Reinthaller A, et al. Topotecan plus carboplatin versus standard therapy with paclitaxel plus carboplatin (PC) or gemcitabine plus carboplatin (GC) or pegylated liposomal doxorubicin plus carboplatin (PLDC): a randomized phase III trial of the NOGGO‐AGO‐Study Group‐AGO Austria and GEICO‐ENGOT‐GCIG intergroup study (HECTOR). Ann Oncol. 2016;27(12):2236‐2241. doi: 10.1093/annonc/mdw418 [DOI] [PubMed] [Google Scholar]
  • 142. Coleman RL, Brady MF, Herzog TJ, et al. Bevacizumab and paclitaxel‐carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum‐sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG‐0213): a multicentre, open‐label, randomised, phase 3 trial. Lancet Oncol. 2017;18(6):779‐791. doi: 10.1016/S1470-2045(17)30279-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum‐sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375(22):2154‐2164. doi: 10.1056/NEJMoa1611310 [DOI] [PubMed] [Google Scholar]
  • 144. Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet. 2017;390(10106):1949‐1961. doi: 10.1016/S0140-6736(17)32440-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Poveda A, Floquet A, Ledermann JA, et al. Olaparib tablets as maintenance therapy in patients with platinum‐sensitive relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT‐Ov21): a final analysis of a double‐blind, randomised, placebo‐controlled, phase 3 trial. Lancet Oncol. 2021;22(5):620‐631. doi: 10.1016/S1470-2045(21)00073-5 [DOI] [PubMed] [Google Scholar]
  • 146. O'Malley DM, Krivak TC, Kabil N, Munley J, Moore KN. PARP inhibitors in ovarian cancer: a review. Target Oncol. 2023;18(4):471‐503. doi: 10.1007/s11523-023-00970-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Pujade‐Lauraine E, Selle F, Scambia G, et al. Maintenance olaparib rechallenge in patients with platinum‐sensitive relapsed ovarian cancer previously treated with a PARP inhibitor (OReO/ENGOT‐ov38): a phase IIIb trial. Ann Oncol. 2023;34(12):1152‐1164. doi: 10.1016/j.annonc.2023.09.3110 [DOI] [PubMed] [Google Scholar]
  • 148. Xu‐Vuillard A, Guerin‐Charbonnel C, Bocquet F, et al. Efficacy of chemotherapy after progression during or following PARPi exposure in ovarian cancer. ESMO Open. 2024;9(9):103694. doi: 10.1016/j.esmoop.2024.103694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Richardson DL, Eskander RN, O'Malley DM. Advances in ovarian cancer care and unmet treatment needs for patients with platinum resistance: a narrative review. JAMA Oncol. 2023;9(6):851‐859. doi: 10.1001/jamaoncol.2023.0197 [DOI] [PubMed] [Google Scholar]
  • 150. Shoji T, Enomoto T, Abe M, et al. Efficacy and safety of standard of care with/without bevacizumab for platinum‐resistant ovarian/fallopian tube/peritoneal cancer previously treated with bevacizumab: the Japanese Gynecologic Oncology Group study JGOG3023. Cancer Sci. 2022;113(1):240‐250. doi: 10.1111/cas.15185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Pujade‐Lauraine E, Hilpert F, Weber B, et al. Bevacizumab combined with chemotherapy for platinum‐resistant recurrent ovarian cancer: the AURELIA open‐label randomized phase III trial. J Clin Oncol. 2014;32(13):1302‐1308. doi: 10.1200/JCO.2013.51.4489 [DOI] [PubMed] [Google Scholar]
  • 152. Matulonis UA, Shapira‐Frommer R, Santin AD, et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: results from the phase II KEYNOTE‐100 study. Ann Oncol. 2019;30(7):1080‐1087. doi: 10.1093/annonc/mdz135 [DOI] [PubMed] [Google Scholar]
  • 153. Pujade‐Lauraine E, Fujiwara K, Ledermann JA, et al. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum‐resistant or platinum‐refractory ovarian cancer (JAVELIN Ovarian 200): an open‐label, three‐arm, randomised, phase 3 study. Lancet Oncol. 2021;22(7):1034‐1046. doi: 10.1016/S1470-2045(21)00216-3 [DOI] [PubMed] [Google Scholar]
  • 154. Moore KN, Angelergues A, Konecny GE, et al. Mirvetuximab soravtansine in FRalpha‐positive, platinum‐resistant ovarian cancer. N Engl J Med. 2023;389(23):2162‐2174. doi: 10.1056/NEJMoa2309169 [DOI] [PubMed] [Google Scholar]
  • 155. Meric‐Bernstam F, Makker V, Oaknin A, et al. Efficacy and safety of trastuzumab deruxtecan in patients with HER2‐expressing solid tumors: primary results from the DESTINY‐PanTumor02 phase II trial. J Clin Oncol. 2024;42(1):47‐58. doi: 10.1200/JCO.23.02005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Konstantinopoulos PA, Cheng SC, Wahner Hendrickson AE, et al. Berzosertib plus gemcitabine versus gemcitabine alone in platinum‐resistant high‐grade serous ovarian cancer: a multicentre, open‐label, randomised, phase 2 trial. Lancet Oncol. 2020;21(7):957‐968. doi: 10.1016/S1470-2045(20)30180-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Oza AM, Estevez‐Diz M, Grischke EM, et al. A biomarker‐enriched, randomized phase II trial of adavosertib (AZD1775) plus paclitaxel and carboplatin for women with platinum‐sensitive TP53‐mutant ovarian cancer. Clin Cancer Res. 2020;26(18):4767‐4776. doi: 10.1158/1078-0432.CCR-20-0219 [DOI] [PubMed] [Google Scholar]
  • 158. Yap TA, O'Carrigan B, Penney MS, et al. Phase I trial of first‐in‐class ATR inhibitor M6620 (VX‐970) as monotherapy or in combination with carboplatin in patients with advanced solid tumors. J Clin Oncol. 2020;38(27):3195‐3204. doi: 10.1200/JCO.19.02404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Lheureux S, Cristea MC, Bruce JP, et al. Adavosertib plus gemcitabine for platinum‐resistant or platinum‐refractory recurrent ovarian cancer: a double‐blind, randomised, placebo‐controlled, phase 2 trial. Lancet. 2021;397(10271):281‐292. doi: 10.1016/S0140-6736(20)32554-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Shah PD, Wethington SL, Pagan C, et al. Combination ATR and PARP Inhibitor (CAPRI): a phase 2 study of ceralasertib plus olaparib in patients with recurrent, platinum‐resistant epithelial ovarian cancer. Gynecol Oncol. 2021;163(2):246‐253. doi: 10.1016/j.ygyno.2021.08.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Yap TA, Fontana E, Lee EK, et al. Camonsertib in DNA damage response‐deficient advanced solid tumors: phase 1 trial results. Nat Med. 2023;29(6):1400‐1411. doi: 10.1038/s41591-023-02399-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Konstantinopoulos PA, Cheng SC, Lee EK, et al. Randomized phase II study of gemcitabine with or without ATR inhibitor berzosertib in platinum‐resistant ovarian cancer: final overall survival and biomarker analyses. JCO Precis Oncol. 2024;8:e2300635. doi: 10.1200/PO.23.00635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Konstantinopoulos PA, Lee JM, Gao B, et al. A phase 2 study of prexasertib (LY2606368) in platinum resistant or refractory recurrent ovarian cancer. Gynecol Oncol. 2022;167(2):213‐225. doi: 10.1016/j.ygyno.2022.09.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Park SJ, Chang SJ, Suh DH, et al. A phase IA dose‐escalation study of PHI‐101, a new checkpoint kinase 2 inhibitor, for platinum‐resistant recurrent ovarian cancer. BMC Cancer. 2022;22(1):28. doi: 10.1186/s12885-021-09138-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Rocconi RP, Stevens EE, Bottsford‐Miller JN, et al. Proof of principle study of sequential combination atezolizumab and Vigil in relapsed ovarian cancer. Cancer Gene Ther. 2022;29(3‐4):369‐382. doi: 10.1038/s41417-021-00317-5 [DOI] [PubMed] [Google Scholar]
  • 166. Bobisse S, Bianchi V, Tanyi JL, et al. A phase 1 trial of adoptive transfer of vaccine‐primed autologous circulating T cells in ovarian cancer. Nat Cancer. 2023;4(10):1410‐1417. doi: 10.1038/s43018-023-00623-x [DOI] [PubMed] [Google Scholar]
  • 167. Hong DS, Van Tine BA, Biswas S, et al. Autologous T cell therapy for MAGE‐A4(+) solid cancers in HLA‐A*02(+) patients: a phase 1 trial. Nat Med. 2023;29(1):104‐114. doi: 10.1038/s41591-022-02128-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Gitto SB, Ihewulezi CJN, Powell DJ Jr. Adoptive T cell therapy for ovarian cancer. Gynecol Oncol. 2024;186:77‐84. doi: 10.1016/j.ygyno.2024.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Hinchcliff EM, Knisely A, Adjei N, et al. Randomized phase 2 trial of tremelimumab and durvalumab in combination versus sequentially in recurrent platinum‐resistant ovarian cancer. Cancer. 2024;130(7):1061‐1071. doi: 10.1002/cncr.35126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Mirza MR, Tandaric L, Henriksen JR, et al. NSGO‐OV‐UMB1/ENGOT‐OV30: a phase II study of durvalumab in combination with the anti‐CD73 monoclonal antibody oleclumab in patients with relapsed ovarian cancer. Gynecol Oncol. 2024;188:103‐110. doi: 10.1016/j.ygyno.2024.06.017 [DOI] [PubMed] [Google Scholar]
  • 171. Tarannum M, Dinh K, Vergara J, et al. CAR memory‐like NK cells targeting the membrane proximal domain of mesothelin demonstrate promising activity in ovarian cancer. Sci Adv. 2024;10(28):eadn0881. doi: 10.1126/sciadv.adn0881 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Chen B, Liu J. Prospects and challenges of CAR‐T in the treatment of ovarian cancer. Int Immunopharmacol. 2024;133:112112. doi: 10.1016/j.intimp.2024.112112 [DOI] [PubMed] [Google Scholar]
  • 173. Li T, Wang J. Therapeutic effect of dual CAR‐T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer. 2020;20(1):678. doi: 10.1186/s12885-020-07180-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Connor AE, Lyons PM, Kilgallon AM, Simpson JC, Perry AS, Lysaght J. Examining the evidence for immune checkpoint therapy in high‐grade serous ovarian cancer. Heliyon. 2024;10(20):e38888. doi: 10.1016/j.heliyon.2024.e38888 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. McGrail DJ, Pilié PG, Rashid NU, et al. High tumor mutation burden fails to predict immune checkpoint blockade response across all cancer types. Ann Oncol. 2021;32(5):661‐672. doi: 10.1016/j.annonc.2021.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Sun Y, Yin Z, Li S, et al. Losartan rewires the tumor‐immune microenvironment and suppresses IGF‐1 to overcome resistance to chemo‐immunotherapy in ovarian cancer. Br J Cancer. 2024;5(10):1683‐1693. doi: 10.1038/s41416-024-02863-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Armbrust R, Fotopoulou C, Sehouli J. The role of neoadjuvant chemotherapy in advanced ovarian cancer: the right treatment to the right patient—ensuring optimal outcomes in advanced ovarian cancer. BJOG. 2024;131(11):1573‐1574. doi: 10.1111/1471-0528.17707 [DOI] [PubMed] [Google Scholar]
  • 178. Rushton T, Krause HB, Samec T, et al. Characterizing the genomic landscape through the lens of FOLR1 status in low and high grade serous ovarian carcinoma. Gynecol Oncol. 2024;191:80‐85. doi: 10.1016/j.ygyno.2024.09.021 [DOI] [PubMed] [Google Scholar]
  • 179. Feng LY, Yan BB, Huang YZ, Li L. Abnormal methylation characteristics predict chemoresistance and poor prognosis in advanced high‐grade serous ovarian cancer. Clin Epigenetics. 2021;13(1):141. doi: 10.1186/s13148-021-01133-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Oliveira DVNP, Biskup E, O'Rourke CJ, et al. Developing a DNA methylation signature to differentiate high‐grade serous ovarian carcinomas from benign ovarian tumors. Mol Diagn Ther. 2024;28(6):821‐834. doi: 10.1007/s40291-024-00740-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Xue Y, Yin T, Yuan S, et al. CYP1B1 promotes PARPi‐resistance via histone H1.4 interaction and increased chromatin accessibility in ovarian cancer. Drug Resist Updat. 2024;77:101151. doi: 10.1016/j.drup.2024.101151 [DOI] [PubMed] [Google Scholar]
  • 182. Shimabukuro‐Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6(1):56. doi: 10.1186/s40425-018-0343-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183. Cutri‐French C, Nasioudis D, George E, Tanyi JL. CAR‐T cell therapy in ovarian cancer: where are we now? Diagnostics (Basel). 2024;14(8):819. doi: 10.3390/diagnostics14080819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Adkins S. CAR T‐cell therapy: adverse events and management. J Adv Pract Oncol. 2019;10(suppl 3):21‐28. doi: 10.6004/jadpro.2019.10.4.11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Inci MG, Richter R, Heise K, Dukatz R, Woopen H, Sehouli J. Patient‐reported outcomes (PROs) and health‐related quality of life (HR‐QoL) in patients with ovarian cancer: what is different compared to healthy women? Cancers (Basel). 2021;13(4):631. doi: 10.3390/cancers13040631 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Johansen G, Lampic C, Flöter Rådestad A, Dahm‐Kähler P, Rodriguez‐Wallberg KA. Health‐related quality of life, sexual function, psychological health, reproductive concerns and fertility outcome in young women treated with fertility‐sparing surgery for ovarian tumors—a prospective longitudinal multicentre study. Gynecol Oncol. 2024;189:101‐108. doi: 10.1016/j.ygyno.2024.07.004 [DOI] [PubMed] [Google Scholar]
  • 187. Harris E, Yorke J, Law K, Winter‐Roach MB, Taylor S. Advanced ovarian cancer patients' experiences of surgical treatment: a qualitative analysis. Semin Oncol Nurs. 2024;40(4):151679. doi: 10.1016/j.soncn.2024.151679 [DOI] [PubMed] [Google Scholar]
  • 188. Dhanis J, Strijker D, Drager LD, et al. Feasibility of introducing a prehabilitation program into the care of gynecological oncology patients—a single institution experience. Cancers (Basel). 2024;16(5):1013. doi: 10.3390/cancers16051013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Ko E, Lee Y. Symptom burden, palliative care knowledge, and palliative care needs in advanced gynecological cancer patients in Korea. Support Care Cancer. 2024;32(9):582. doi: 10.1007/s00520-024-08772-0 [DOI] [PubMed] [Google Scholar]
  • 190. Mokshagundam S, McGree ME, Tapia AL, et al. Physical and psychological distress amongst patients undergoing neoadjuvant chemotherapy for advanced ovarian cancer. Gynecol Oncol. 2024;190:230‐235. doi: 10.1016/j.ygyno.2024.08.024 [DOI] [PubMed] [Google Scholar]
  • 191. Shrestha S, Sapkota S, Teoh SL, et al. Comprehensive assessment of pain characteristics, quality of life, and pain management in cancer patients: a multi‐center cross‐sectional study. Qual Life Res. 2024;33(10):2755‐2771. doi: 10.1007/s11136-024-03725-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Fundytus A, Prasad V, Booth CM. Has the current oncology value paradigm forgotten patients' time? Too little of a good thing. JAMA Oncol. 2021;7(12):1757‐1758. doi: 10.1001/jamaoncol.2021.3600 [DOI] [PubMed] [Google Scholar]
  • 193. Gupta A, Eisenhauer EA, Booth CM. The time toxicity of cancer treatment. J Clin Oncol. 2022;40(15):1611‐1615. doi: 10.1200/JCO.21.0281 [DOI] [PubMed] [Google Scholar]
  • 194. Johnson WV, Blaes AH, Booth CM, Ganguli I, Gupta A. The unequal burden of time toxicity. Trends Cancer. 2023;9(5):373‐375. doi: 10.1016/j.trecan.2023.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Gupta A. Management of malignant bowel obstruction. Lancet Gastroenterol Hepatol. 2024;9(1):14‐15. doi: 10.1016/S2468-1253(23)00333-3 [DOI] [PubMed] [Google Scholar]

Articles from Ca are provided here courtesy of Wiley

RESOURCES