Ovarian cancer and malnutrition: a literature review
Review Article

Ovarian cancer and malnutrition: a literature review

Zhuyan Shao1, Liying Fu1, Yi Lu2, Haifei Zhou3, Yuyang Zhu4, Tao Zhu1

1Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou, China; 2Department of Clinical Nutrition, Zhejiang Cancer Hospital, Hangzhou, China; 3Department of Clinical Oncology, Wenzhou Medical University, Wenzhou, China; 4School of The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China

Contributions: (I) Conception and design: Z Shao, T Zhu; (II) Administrative support: L Fu; (III) Provision of study materials or patients: Y Lu; (IV) Collection and assembly of data: H Zhou, Y Zhu; (V) Data analysis and interpretation: Z Shao, T Zhu; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Tao Zhu, MD. Department of Gynecologic Oncology, Zhejiang Cancer Hospital, No. 1 East Banshan Road, Hangzhou 310022, China. Email: zhutao@zjcc.org.cn.

Background and Objective: Among gynecological malignancies, ovarian cancer is the most fatal, with surgery and chemotherapy being the primary treatment modalities. The nutritional status of patients with ovarian cancer undergoing chemotherapy after surgery remains generally poor, with 76.1% classified as severely malnourished according to the Patient-Generated Subjective Global Assessment (PG-SGA), while only 9.0% are well-nourished and do not require nutritional intervention. The high risk of nutritional decline associated with the new treatment modality, poly (ADP ribose) polymerase (PARP) inhibitors, has not received sufficient attention. This review examined the factors contributing to malnutrition in ovarian cancer, the adverse effects of malnutrition on treatment outcomes, the importance of regular nutritional screening and assessment, and potential nutritional interventions.

Methods: A review of the relevant literature was conducted to analyze the prevalence of malnutrition in patients with ovarian cancer, its impact on treatment and prognosis, and the role of nutritional assessment and interventions in improving patient outcomes.

Key Content and Findings: Malnutrition is highly prevalent among patients with ovarian cancer and is associated with worsened treatment side effects, reduced quality of life, and decreased survival rates. The use of PARP inhibitors may pose an additional risk for nutritional decline, but this possibility has not been sufficiently studied. Regular nutritional screening and assessment are essential for the early detection and management of malnutrition. Nutritional interventions have been investigated for their potential to support patients with ovarian cancer through treatment, but further research is needed to determine the most effective strategies.

Conclusions: Malnutrition is a major concern for ovarian cancer patients, especially those receiving chemotherapy or PARP inhibitors. Regular nutritional assessment and timely interventions may improve treatment tolerance and overall prognosis. Further prospective studies and large randomized controlled trials are necessary to establish effective nutritional strategies for high-risk patients.

Keywords: Ovarian cancer; malnutrition; poly (ADP ribose) polymerase inhibitors (PARP inhibitors); nutritional interventions


Submitted Apr 10, 2025. Accepted for publication May 19, 2025. Published online May 27, 2025.

doi: 10.21037/tcr-2025-758


Introduction

Overview of ovarian cancer and malnutrition

Ovarian cancer remains a significant global health issue, being the eighth most common cancer among women and responsible for a considerable number of cancer-related deaths (1). Ovarian cancer accounts for approximately 23% of all gynecological cancer diagnoses but is responsible for 47% of all deaths from gynecological cancer (2). According to World Cancer Research Fund (https://www.wcrf.org/cancer-trends/ovarian-cancer-statistics/), in 2022, there were 324,603 new cases of ovarian cancer and 206,956 related deaths worldwide. Approximately 70% of newly diagnosed patients and 85% of those who experience relapse may have peritoneal metastasis, leading to symptoms such as abdominal pain, bloating, and decreased appetite, which can affect nutritional intake (3). The disease places the body in a high metabolic state, increasing the risk of malnutrition (4).

Malnutrition is a significant concern for patients with ovarian cancer, particularly those undergoing chemotherapy, due to its potential to exacerbate side effects, impair recovery, and worsen overall survival (OS) outcomes. Based on the Subjective Global Assessment (SGA) and Patient-Generated SGA (PG-SGA) scores, 24% of patients with gynecological cancer are classified as malnourished, with the highest prevalence among those with ovarian cancer (67%) (5). The incidence of malnutrition can be as high as 75% in patients with associated bowel obstruction. Furthermore, about 23% of patients are diagnosed with cachexia (6).

Although the relationship between malnutrition and poor clinical outcomes such as complications, prolonged hospital stays, and reduced quality of life (QoL) is well-documented, the mechanisms through which traditional treatments such as chemotherapy, tumor progression, and newer therapies such as PARP inhibitors contribute to nutritional decline have not been completely clarified. Despite the recognized importance of nutritional intervention, it remains under-addressed in clinical settings, and there is limited research on the multifaceted factors contributing to nutritional decline in patients with ovarian cancer.

This review was thus conducted to address the deficiencies in understanding regarding malnutrition in ovarian cancer via a focus on the nutritional challenges these patients face, particularly the underexplored impact of PARP inhibitors. We also evaluated the role of early nutritional assessment and intervention in addressing these challenges and improving patient outcomes. The ultimate goal is to identify more effective strategies for the prevention and management of malnutrition and to optimize the overall therapeutic strategy for patients with ovarian cancer. We present this article in accordance with the Narrative Review reporting checklist (available at https://tcr.amegroups.com/article/view/10.21037/tcr-2025-758/rc).


Methods

The MEDLINE, Embase, Cochrane, and the Cumulative Index to Nursing and Allied Health Literature (CINAHL) databases were searched from inception until November 2024. One author (Z.S.) screened the title and abstracts of the retrieved records for keywords and reviewed the full text of all relevant publications for eligibility. The search strategy was limited to English-language articles, and there were no restrictions on date of publication. The search strategy is described in Table 1.

Table 1

Summary of search strategy

Items Specification
Date of search Until November 2024
Databases and other sources searched MEDLINE, Embase, Cochrane, CINAHL
Search terms used Table S1
Timeframe From database inception to November 2024
Inclusion and exclusion criteria Inclusion: (I) studies evaluating nutrition interventions in women undergoing active treatment for ovarian cancer; (II) published in English language; (III) original clinical trials, systematic reviews, or meta-analyses; (IV) interventions focusing on nutrition alone or combined with supportive care (e.g., exercise)
Exclusion: studies on mixed cancer populations unless they reported specific outcomes for ovarian cancer
Selection process Titles and abstracts were screened by one author (Z.S.). Full texts of eligible studies were reviewed to confirm inclusion
Any additional considerations, if applicable There were no restrictions on publication date or study outcomes due to limited research in this area

CINAHL, the Cumulative Index to Nursing and Allied Health Literature.


Causes of malnutrition in ovarian cancer

Tumor progression-related factors

Patients with ovarian cancer often experience loss of appetite, which may be related to hormones such as leptin and ghrelin secreted by the tumor. The levels and mechanisms of action of these hormones are altered in those with cancer, leading to decreased appetite and reduced food intake (7,8). Moreover, advanced ovarian cancer can cause bowel obstruction and ascites (fluid accumulation in the abdomen), leading to nausea, vomiting, and early satiety, which reduce food intake (9).

Ovarian cancer can cause systemic inflammatory response in the body. Inflammatory factors [such as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β)] secreted by tumor cells and their microenvironment activate signaling pathways such as nuclear factor kappa beta (NF-κB) and signal transducer and activator of transcription 3 (STAT3) (10-12). Activation of these inflammatory factors and signaling pathways can lead to increased protein catabolism, fat decomposition, and decreased appetite, leading to malnutrition (13).

Ovarian cancer places the body in a hypercatabolic state and consumes a large amount of energy and nutrients (such as glucose and amino acids) to support the rapid growth of the tumor (14). This leads to an insufficient supply of energy and nutrients for normal cells. In addition, ovarian cancer can also cause enhanced gluconeogenesis in the liver, leading to an increased demand for glucose in the liver, leading to systemic malnutrition. The substantial consumption of energy by tumors leads to rapid depletion of fat and muscle reserves in patients. These may explain why about 23% of patients with ovarian cancer have cachexia at the time of diagnosis (6).

Treatment-related factors

The conventional treatment for advanced epithelial ovarian cancer is primary debulking surgery (PDS) and postoperative platinum-based adjuvant chemotherapy. If there is tumor involvement or invasion, multiple organs need to be removed to achieve satisfactory cytoreduction, usually including the intestines, which can directly affect nutritional intake and absorption. Epithelial ovarian cancer accounts for 90% of all cases (4).

Nausea and vomiting are common side effects of chemotherapy. A meta-analysis of 1,213 patients with recurrent ovarian cancer found that 58.1% of patients experienced nausea and 31.0% experienced vomiting, which can greatly reduce appetite and food intake. Mucositis and altered taste can make eating uncomfortable or unpalatable (15). Studies have shown that platinum-based chemotherapy regimens, commonly used for ovarian cancer, produce significant gastrointestinal reactions, which can lead to a significant reduction in dietary intake and consequently affect nutritional status (16,17).

Poly (ADP ribose) polymerase (PARP) inhibitors are a newer form of treatment for ovarian cancer, and several large clinical studies have shown the potential to cause decreased appetite, dysgeusia, nausea, and vomiting (18); these are summarized in Table 2 (19-28).

Table 2

Adverse effects of PARP inhibitors

Trial Therapy N PARP inhibitor dose Decreased appetite (%) Dysgeusia (%) Nausea (%) Vomiting (%) Ref.
PARP inhibitor as first-line maintenance therapy in ovarian cancer
   SOLO1 Olaparib vs. placebo 260 vs. 130 300 mg BID 0.4 vs. 0 26 vs. 4 77 vs. 38 40 vs. 15 (19)
   PAOLA1 Bevacizumab + olaparib vs. bevacizumab + placebo 535 vs. 267 300 mg BID 8 vs. 4 8 vs. 1 53 vs. 22 22 vs. 11 (20)
   PRIMA Niraparib vs. placebo 484 vs. 244 300 mg QD 19 vs. 8.2 NA 57.4 vs. 27.5 22.3 vs. 11.9 (21)
   OVARIO Bevacizumab + niraparib 105 200 mg/300 mg QD + bevacizumab 15 mg/kg 12.4 15.2 52.4 18.1 (22)
   VELIA Control group vs. veliparib-combination-only group vs. veliparib-throughout group 371 vs. 376 vs. 377 300 mg BID × 2 weeks, then 400 mg BID 23 vs. 22 vs. 29 18 vs. 15 vs. 20 68 vs. 72 vs. 80 36 vs. 35 vs. 49 (23)
PARP inhibitor in platinum-sensitive, relapsed ovarian cancer therapy
   SOLO2 Olaparib vs. placebo 195 vs. 95 300 mg BID 22 vs. 11 27 vs. 7 76 vs. 33 38 vs. 19 (24)
   NOVA Niraparib vs. placebo 367 vs. 179 300 mg QD 25.3 vs. 14.5 10.1 vs. 3.9 73.6 vs. 35.2 34.3 vs. 16.2 (25)
   NORA Niraparib vs. placebo 177 vs. 88 300 mg QD 17.5 vs. 2.3 NA 53.1 vs. 19.3 32.2 vs. 4.5 (26)
   ARIEL2 Rucaparib 204 600 mg BID 42 43 79 44 (27)
   ARIEL3 Rucaparib vs. placebo 375 vs. 189 600 mg BID 23 vs. 14 NA 75 vs. 37 37 vs. 15 (28)

ARIEL2, a clinical trial evaluating rucaparib in ovarian cancer; ARIEL3, a phase 3 trial of rucaparib maintenance therapy in recurrent ovarian cancer; BID × 2 weeks, twice a day for 2 weeks; BID, twice a day; NA, not available; NORA, a clinical trial related to ovarian cancer (specific details needed); NOVA, a phase 3 trial evaluating niraparib maintenance therapy in ovarian cancer; OVARIO, a clinical trial studying bevacizumab with maintenance therapy in ovarian cancer; PAOLA1, a phase 3 trial assessing olaparib and bevacizumab in ovarian cancer; PARP, poly (ADP-ribose) polymerase (a family of enzymes involved in DNA repair); PRIMA, a phase 3 trial of niraparib as maintenance therapy in ovarian cancer; QD, once a day; SOLO1, a phase 3 trial of olaparib maintenance therapy in newly diagnosed BRCA-mutated ovarian cancer; SOLO2, a phase 3 trial of olaparib in platinum-sensitive recurrent ovarian cancer; VELIA, a phase 3 trial assessing veliparib in combination with chemotherapy for ovarian cancer.

Although several large clinical studies have reported gastrointestinal side effects of PARP inhibitors, there is still a lack of high-quality, prospective studies directly linking PARP inhibitor use to malnutrition in ovarian cancer patients (19-28). Existing evidence primarily focuses on individual symptoms such as nausea, vomiting, or dysgeusia, rather than comprehensive nutritional outcomes. Furthermore, most clinical trials of PARP inhibitors were conducted in relatively homogenous populations, limiting their generalizability to patients from diverse ethnic and socioeconomic backgrounds.

To address this gap, future research should include nutritional endpoints in trials of PARP inhibitors, such as weight change, nutritional risk screening scores, and quality-of-life (QoL) measures. Subgroup analyses by age, comorbidities, and ethnicity would also help clarify how different populations may be disproportionately affected. Multicenter observational studies or randomized trials incorporating nutritional assessments could provide much-needed insight into the long-term nutritional impact of PARP inhibitors.

Psychological and economic factors

The psychological stress and emotional problems caused by cancer and its treatment may also affect patients’ eating behavior and nutritional status. For example, anxiety and depression may lead to a loss of appetite, thereby affecting nutrient intake (29).

Financial difficulties may limit patients’ ability to obtain high-quality food, especially when long-term treatment is required. In addition, economic pressure may also reduce patients’ overall QoL and indirectly impair their nutritional status (30).


Adverse effects of malnutrition in ovarian cancer

Malnutrition, cachexia, and sarcopenia in ovarian cancer

Malnutrition, cachexia, and sarcopenia have highly similar symptoms and often overlap. Malnutrition is a state of energy or nutrient deficiency caused by insufficient intake or utilization disorders, which in turn leads to changes in body composition, decreased physiological and mental functions, and adverse clinical outcomes. Cachexia, characterized by severe weight loss, muscle wasting, and fatigue, is common in patients with cancer. It is driven by a combination of metabolic changes induced by the tumor and inflammatory responses. Sarcopenia is a syndrome caused by progressive, widespread reduction in skeletal muscle mass, decreased muscle strength, and decreased human function, which is associated with an increased risk of adverse outcomes such as decreased motor ability, decreased QoL, and increased mortality.

Clinically, these three phenotypes often coexist and cannot be easily or completely differentiated (31-33). A study has found that 80% of older adult patients with cancer have at least one of these conditions, and 30% have all three (34). Indeed, malnutrition, cachexia, and sarcopenia have both similarities and differences.

Surgical outcomes

Malnutrition in women with ovarian cancer has been related to outcomes from surgery. Multiple studies have consistently shown that R0 resection is the most important factor related to the prognosis of patients with ovarian cancer (12,35). A prospective study enrolling 152 patients with ovarian cancer found that patients with age >65 years, ascites >500 mL, or platinum-resistant ovarian cancer had a significantly increased risk of a Nutritional Risk Score 2002 (NRS-2002) ≥3 (P=0.014, 0.001, and 0.007, respectively). In addition, NRS-2002 <3 was an independent predictor of complete tumor resection (P=0.009) (36).

Skeletal muscle mass reflects nutritional status. In a study that included 75 consecutive patients with advanced-stage ovarian cancer who underwent elective surgery, 26% of patients had sarcopenia. The results indicated that compared with patients without sarcopenia, patients with sarcopenia had a higher incidence of postoperative complications, especially nonsurgical complications (such as pneumonia, urinary tract infection, and cardiac complications). In addition, length of stay and mortality were significantly higher in patients with sarcopenia (37).

When surgery is required to treat intestinal obstruction, a common complication of ovarian cancer, malnutrition (as manifest by weight loss, low serum albumin, or low lymphocyte count) is a factor significantly associated with poor surgical outcomes, lower postoperative survival, and increased rates of postoperative infectious complications (38-40).

Length of hospital stay

Malnutrition is associated with the incidence and length of hospital stay of patients with ovarian cancer. A cohort study enrolled 157 patients with suspected or confirmed gynecologic neoplasms. Malnutrition (PG-SGA score group B or C), low health-related QoL (functional assessment of cancer therapy-general score) and diagnosis of advanced ovarian cancer were found to be the main factors contributing to prolonged hospitalization (41). Another study analyzed the National Surgical Quality Improvement Program database and included 290 patients who underwent surgery for ovarian, uterine, or cervical cancer. The study found that for patients with ovarian cancer, malnutrition [body mass index (BMI) <18.5 kg/m2], as defined by the European Society for Clinical Nutrition (ESPEN), was associated with higher rates of postoperative complications (risk ratio 1.36), reoperation (risk ratio 2.53), and rehospitalization (risk ratio 1.69). In addition, a serum albumin level <3.5 g/dL was also associated with poor postoperative outcomes in patients with ovarian cancer. The malnourished patients with ovarian cancer had higher rates of readmissions, reoperations, and complications (42).

Progression-free survival (PFS) and OS

Malnutrition may promote tumor metastasis and recurrence. Studies have shown that there is an interaction between malnutrition and tumor metastasis, with the greater the degree of malnutrition, the greater the degree of tumor infiltration and metastasis (43-45).

One study investigated the relationship between protein intake after primary treatment and disease recurrence and survival in patients with ovarian cancer. A higher level of protein intake was associated with better PFS [>1–1.5 g/kg body weight: adjusted hazard ratio (HR) =0.69, 95% confidence interval (CI): 0.48–1.00; >1.5 g/kg: adjusted HR =0.61, 95% CI: 0.41–0.90; >20% total energy intake: adjusted HR =0.77, 95% CI: 0.61–0.96]. A higher level of protein intake was associated with better PFS. The study concluded that protein intake should be encouraged after primary treatment in patients with ovarian cancer (45).

Multiple studies have consistently shown that malnutrition is a common complication in patients with ovarian cancer and is associated with shorter OS and PFS. A prospective study enrolling 152 patients with ovarian cancer found that the median OS for patients with NRS-2002 ≥3 was 7 months (95% CI: 0–24 months), while the median OS for patients with NRS-2002 <3 was 46 months (P=0.001) (36). A retrospective study included 213 patients with advanced ovarian cancer and used the Nutritional Risk Index (NRI) to assess nutritional status. The results showed that the 5-year OS rate of patients with moderate-to-severe malnutrition (45.3%) was significantly lower than that of patients with normal-to-mild malnutrition (64.0%) (44). After adjustments were made for known prognostic factors, the relative risk of death in patients with moderate-to-severe malnutrition was 5.8 times that of those with normal malnutrition, and the PFS was shorter in this group than in the normal-to-mild group (median PFS: 15 vs. 28 months; P=0.011).

There is also a complex link between malnutrition and economic factors which deeply affects the medical and public health fields. One study examined 2,873 cases of ovarian cancer, and after adjustment for comorbid conditions, cancer stage, tumor histology, operation status, and lifestyle factors, it was found that in women with early-stage cancer, the HR for death among less educated women was 1.56 (95% CI: 1.11–2.68), which was higher than that for more educated women; moreover, they had a shorter survival time, thus suggesting socioeconomic differences associated with survival (46).

Although current evidence highlights the association between malnutrition and short-term clinical outcomes such as surgical complications, hospital length of stay, and postoperative readmissions, there is a notable paucity of studies focusing on long-term endpoints. These include OS, PFS, and patient-reported QoL measures. Most existing data are derived from retrospective analyses or cross-sectional studies, which may not fully capture the dynamic changes in nutritional status over the disease trajectory.

Therefore, future studies should prioritize long-term, prospective cohort designs or randomized controlled trials (RCTs) to evaluate the sustained impact of nutritional status and interventions. Incorporating validated QoL instruments and objective nutritional biomarkers could offer a more comprehensive understanding of how malnutrition affects survivorship in ovarian cancer. Longitudinal evidence will also support the development of targeted, stage-specific nutritional strategies and health policies.

Economic implications

Malnutrition increases the cost of treatment and thus increases the burden on the health system. Malnourished patients may require additional health care resources, such as additional nutritional support, management of complications, and specialized care. In one study, 39% of patients with ovarian cancer had a high level of financial burden, and 19% reported that ovarian cancer caused financial difficulties for their families (30). Another study examined the association between financial status, based upon family income and expenses, and QoL in Chinese women with recurrent ovarian cancer. The results indicated that low financial status was associated with poorer QoL scores, and patients with low financial status had a higher risk of a worsening QoL (47). Additionally, frequent hospitalizations and treatments result in an overuse of public healthcare resources, which places additional economic pressure on the healthcare system. One study assessed the burden of malnutrition among 70,855,014 patients receiving care in US hospitals and found that the mean length of hospital stay for malnourished patients was about 9–10 days, which equated to a mean total expenditure of USD 105,925–110,389. These findings demonstrate that malnutrition poses significant clinical and economic burden on patients and the healthcare system (48).

Malnutrition may also lead to reduced work capacity, lower earnings, and decreased productivity. Specifically, patients who are undernourished experience a decline in physical and cognitive functions, which in turn affects their ability to work. For low-income patients in particular, the loss of the ability to work means greater financial stress. In one study that included 6,925 patients with ovarian cancer, unemployed patients were more likely to have a high financial burden (P<0.001). The financial burden on patients included indirect costs of care (e.g., lost work), out-of-pocket costs, housing and heating costs, and transportation (49). When a large number of patients have reduced productivity due to malnutrition, this reduces the available workforce in society, adversely affecting the overall economy.

Socioeconomic factors such as education level, income, insurance coverage, and access to cancer centers significantly affect nutritional support and treatment adherence in ovarian cancer patients. While much of the available institutional data stems from China—owing to the country’s large patient population and centralized healthcare reporting systems—similar patterns have been observed globally. For example, a study from Europe also reports disparities in nutritional intervention access based on socioeconomic status, especially among uninsured, rural, or minority populations. A European guideline also emphasize early screening and support in socioeconomically disadvantaged groups (50). These international findings suggest that despite regional differences, socioeconomic inequality remains a common barrier to optimal nutritional care for ovarian cancer patients worldwide.


The impact of nutritional intervention

Many studies and guidelines emphasize that regular nutritional risk screening, timely nutritional assessment, and nutritional therapy are critical components in the comprehensive treatment of ovarian cancer (51,52). The OS of patients with improved nutritional status is significantly better than that of patients with deteriorated nutritional status and is independent of other prognostic factors (44). Therefore, it is important to assess the status of malnutrition in patients with ovarian cancer to provide effective nutritional intervention, reduce nutritional decline during treatment, prevent or control malnutrition, and reduce muscle loss, thereby improving survival.

Methods and tools for nutritional screening of patients with ovarian cancer

To address the challenges of malnutrition in ovarian cancer, a personalized, multidisciplinary, comprehensive, and proactive approach to screening, assessment and treatment is recommended. This can not only improve the nutritional status and psychological state of the patients but also further optimize their QoL and significantly improve their survival expectancy.

Nutrition screening and evaluation constitute the first step of nutritional intervention for patients with cancer. There are several tools available for examining malnutrition and nutritional assessment, the most common assessment tools include NRS-2002 and PG-SGA. In a study that included 442 patients with ovarian cancer, multivariate analysis showed that NRS-2002 predicted the 30-day readmission rates (P<0.05) (53). The PG-SGA has a high sensitivity in predicting nutritional status and is the standard for nutrition assessment among patients with cancer. A gynecological study (25% ovarian cancer) reported that PG-SGA was significantly correlated with subjective and objective parameters, especially in patients with partially normal albumin and skinfold thickness, suggesting a potential risk of malnutrition. Therefore, the study concluded that PG-SGA is more suitable for the nutritional assessment of patients with gynecological malignancies (5). Other nutritional screening and nutritional assessment tools are described in Table 3 (54-67).

Table 3

The nutritional screening and nutritional assessment tools

Tool (acronym), year Features/aspects Patient group Reference
Nutritional screening
   Malnutrition Universal Screening Tool (MUST), 2004 BMI, weight loss, and illness in relation to food intake All inpatients Stratton et al. (54)
   Nutritional Risk Screening 2002 (NRS-2002), 2003 BMI, weight loss, and acute disease score All inpatients Kondrup et al. (55)
   Prognostic Nutritional Index (PNI), 1979 Serum albumin, TSF, TFN, and DH Surgical patients Mullen et al. (56)
   Malnutrition Screening Tool (MST),1999 Data about recent appetite status and weight loss All inpatients Ferguson et al. (57)
   Mini Nutritional Assessment short form (MNA-SF), 2009 First 6 items of 18 MNA Older adult patients Kaiser et al. (58)
   Nutritional Control (CONUT), 2002 Laboratory data (serum albumin, cholesterol, total lymphocytes, and hematocrit) All inpatients de Ulíbarri Pérez et al. (59)
   Short Nutritional Assessment Questionnaire (SNAQ), 2005 Items related to appetite, food timing during day, food preferences, and daily number of meals Elderly patients Kruizenga et al. (60)
   Nutriscore, 2017 MST, tumor location, active treatment Cancer patients Arribas et al. (61)
Nutritional assessment
   Subjective Global Assessment (SGA), 1987 Weight change, dietary intake change, gastrointestinal symptoms, functional capacity, and physical examination Cancer patients, surgery, liver diseases Detsky et al. (62)
   Patient-Generated Subjective Global Assessment (PG-SGA), 1996 Weight change, dietary intake change, gastrointestinal symptoms, functional capacity, and physical examination Cancer patients, surgery, liver diseases Ottery (63)
   Mini-Nutritional Assessment (MNA), 1996 Anthropometry; general evaluation; dietary assessment; subjective assessment Older adult patients Guigoz et al. (64)
   American Society for Parenteral and Enteral Nutrition (ASPEN) criteria for malnutrition, 2012 Insufficient energy intake, weight loss, loss of muscle mass, loss of subcutaneous fat, localized or generalized fluid accumulation, and diminished functional status All patients White et al. (65)
   The European Society of Clinical Nutrition and Metabolism (ESPEN) criteria for malnutrition, 2015 BMI (18.5 kg/m2), or weight loss and reduced BMI, or a low FFMI All patients Cederholm et al. (66)
   Global Leadership Initiative on Malnutrition (GLIM), 2020 Weight loss, BMI, muscle mass, dietary intake change, inflammation All patients de van der Schueren et al. (67)

BMI, body mass index; DH, delayed healing; FFMI, Fat-Free Mass Index; Nutriscore, Nutritional Scoring System; TFN, total lymphocyte count; TSF, triceps skinfold.

Notably, PG-SGA is arguably the most appropriate for ovarian cancer patients. ovarian cancer patients frequently experience cancer-specific symptoms such as nausea, early satiety, and ascites, all of which are systematically evaluated in the PG-SGA. Furthermore, the tool’s dynamic nature makes it suitable for monitoring during prolonged treatment courses, including chemotherapy and PARP inhibitor-based maintenance therapy. Additionally, PG-SGA scores have been associated with critical clinical outcomes such as postoperative recovery and survival, further supporting its utility in this setting. In contrast, NRS-2002 and Global Leadership Initiative on Malnutrition (GLIM), while useful in broader contexts, may lack the necessary specificity or feasibility in routine gynecologic oncology practice. Given these strengths, PG-SGA is particularly well-suited for the ongoing nutritional evaluation of ovarian cancer patients.

Patients with advanced ovarian cancer often have ascites and pleural effusion and should be evaluated for weight correction and preferably for muscle mass. In addition to malnutrition risk examination and related assessment tools, nutrition can be assessed via the area of skeletal muscle through several methods. These include bioelectrical impedance analysis (BIA), dual-energy X-ray absorptiometry (DXA), and computed tomography (CT) imaging. C-reactive protein, lymphocytes, and other inflammation status indicators, such as neutrophil-to-lymphocyte ratio, lymphocyte-to-monocyte ratio, and C-reactive protein-to-albumin ratio, are also highly informative.

Nutrition intervention during treatment for ovarian cancer

Patients at different stages of treatment need timely, comprehensive, and effective interventions to improve appetite and nutrition. This can include daily dietary supplements, nutritional supplementation, and pharmacologic interventions.

Dietary regulation

Nutritional education and dietary guidance are the first choice for nutritional treatment. A study has shown that high vegetable intake is associated with better survival (68). Cruciferous vegetables such as broccoli and cabbage are rich in antioxidants and fiber, which help improve gut health and overall immune function. Additionally, animal products and vitamin-rich dietary patterns have been associated with a reduced risk of ovarian cancer (69). Overall, beneficial diet choices consist of lean meats, fish, and eggs, as well as fruits and vegetables rich in vitamins C and E. Unsaturated fats from natural sources such as fish oil and olive oil are also helpful.

Cancer patients often face inadequate protein intake, which may affect the effectiveness of treatment and speed of recovery. It is recommended that adequate protein supply be ensured via an increased intake of fish, meat, eggs, and soy products (70).

In contrast, it has been shown that intake of red and white meat is positively associated with the risk of ovarian cancer (68). Therefore, reduction in the consumption these types of meat in favor of more plant-based protein sources such as beans and soy products is recommended. Moreover, high-starch dietary patterns have been associated with an increased risk of ovarian cancer (69). Thus, the intake of high-sugar and refined carbohydrates should be limited in favor of whole grains and foods with a low glycemic index.

Nutritional supplementation

When the patient’s oral intake is insufficient, supplementary enteral nutrition (EN) is recommended, and oral nutritional supplementation (ONS) is preferred. When oral feeding plus EN cannot meet the requirement, parenteral nutrition (PN) should be considered, and even total parenteral nutrition (TPN) if necessary.

For patients at risk of malnutrition, early introduction of oral nutritional counseling and recommendation of oral nutritional supplements have been shown to reduce mortality, complications, and shorten hospital stays. Oral feeing early in the perioperative period may stimulate intestinal hormones and increase intestinal motility, which would promote normal recovery of bowel motility and tolerance of oral intake and improve nutritional status. EN is the preferred approach because it is more physiologic, has fewer complications, is less expensive, and is easier to monitor than is PN. EN can increase energy intake and nutritional status while reducing the gastrointestinal toxicity caused by cancer treatment. Branched chain amino acids (BCAAs) appear to reduce hospital stay, reduce morbidity, and improve QoL. However, there is no reduction in mortality for the long-term benefit of the patient (71).

PN in terminal patients has been demonstrated to improve the QoL, energy balance, and body composition while also prolonging survival. Omega-3 fatty acid supplementation provides clear benefits at the biochemical, clinical, and functional levels, while glutamine supplementation appears to enhance the efficacy of chemotherapy and radiotherapy while reducing tissue toxicity and improving outcomes (72).

The use of TPN is controversial. A retrospective cohort study included 55 patients with advanced epithelial ovarian cancer and compared the survival of patients treated with TPN after bowel obstruction with that of patients who did not receive TPN. The results showed that OS was 35 months for patients treated with TPN and 23 months for patients not treated with TPN (P=0.05); moreover, patients receiving TPN after obstruction were more likely to undergo concurrent chemotherapy (64% vs. 26%; P=0.004) (73). However, in another study, 147 patients with ovarian cancer were enrolled, 69 of whom received TPN and 78 conservative treatment. The results showed that patients in the TPN group had a longer time to regain bowel function (5.77 vs. 4.70 days; P<0.001), and longer hospitalization (11.46 vs. 7.14 days; P<0.001) (74). Therefore, the use of TPN needs to be evaluated in the context of the patient’s specific situation.

Pharmacologic interventions

Pharmacotherapy plays a key role in the treatment of cancer-caused anorexia nervosa and has the primary goal of targeting etiologic mechanisms.

Progestogens are capable of targeting the synthesis of inflammatory factors and have been shown to be effective therapeutic options (75). Megestrol acetate (MA) exerts anti-inflammatory effects and is able to reduce the levels of inflammatory factors such as IL-1, IL-6, and TNF-α. In addition, MA can improve appetite in patients cancer by activating proappetite neurons in the hypothalamic region through increasing the levels of neuropeptide Y, an appetite stimulant in the central nervous system (76,77). The efficacy of MA is positively correlated with its dose, and 800 mg is recommended by guidelines (78,79). Indeed, several studies have shown significant improvements in appetite and body weight in patients with cancer following MA administration, particularly with a daily dose of 800 mg and a treatment duration of 12 weeks, with effects observed at higher doses or after a certain duration of treatment (80-83). The third generation of nanocrystal MA oral suspension significantly improves the solubility and dissolution rate of megestrol, enhances the bioadhesion effect, and increases the absorption pathway. This improvement eliminates the need for patients to rely on a high-fat, high-calorie diet and is effective both before and after meals (84,85). Clinical studies have also demonstrated that compared to the regular dosage form, patients with nanocrystal MA oral suspension gain weight significantly faster. Specifically, the mean time to weight gain was shortened from 14 to 3 days, and the average weight gain over 12 weeks was 5.4 kg, which was 1.5 times higher than that achieved by the conventional dosage form (86,87). International guidelines are unanimous in considering MA as the preferred treatment option for cancer-related anorexia cachexia syndrome (CACS) (78,88-90).

Corticosteroids, such as dexamethasone, are not as efficacious as megestrol, although they have been shown to improve appetite (91). In addition, corticosteroid use is associated with side effects such as muscle atrophy, glucose intolerance, and immunosuppression, which may lead to decreased efficacy with long-term treatment. The potential nutritional advantages of long-term use are often outweighed by the risk of their adverse effects (92). Therefore, given the higher frequency of adverse effects, corticosteroids should be applied with caution and limited mainly to the treatment of patients with end-stage tumors (93).

Anamorelin is an orally active, high-affinity, selective ghrelin-receptor agonist. Ghrelin is the natural ligand for the G-protein-coupled ghrelin receptor, and when activated, it exerts anabolic and appetite-stimulating effects. These effects are partly mediated through transient increases in growth hormone and insulin-like growth factor 1 (IGF-1). Several double-blind, randomized phase 2 clinical studies in patients with advanced cancer have shown that anamorelin is effective in increasing lean body mass, body weight, and appetite. However, some subgroups (such as patients with a BMI ≤18.5 kg/m2) showed no improvement in lean body mass (89,94). Additionally, anamorelin may increase IGF-1, which theoretically could promote tumor progression. Therefore, the long-term benefits and safety of anamorelin require further assessment and validation (95).

The research on cannabinoids for the treatment of CACS is limited and inconsistent. Their effectiveness in terms of appetite improvement, weight gain, and QoL enhancement appears to be less than that of other drugs (89,96).

Other therapeutic agents have been studied, including olanzapine (92), androgens or selective androgen receptor modulators (89,90), cyproheptadine, hydralazine sulfate, melatonin, and TNF inhibitors (89,92,96); however, these either provide little benefit or are not supported by a sufficient evidence to justify their routine use. More research is therefore needed to clarify the efficacy and safety of these agents.

Nutritional intervention and results

Accurate evaluation of perioperative nutrition in patients with ovarian cancer is crucial in clinical settings. Preoperative nutritional management is critical to improving the clinical outcomes of patients with ovarian cancer. A study has shown that preoperative nutritional management can optimize patients’ nutritional status and surgical prognosis by providing personalized nutritional intervention, monitoring and evaluation, and increasing physical activity (97). Early postoperative nutritional intervention, including innovative nutritional strategies such as chewing gum, drinking coffee, ketogenic diet, or supplementation with fruit and vegetable juice concentrates, has been shown to reduce hospital stay and improve postoperative intestinal recovery (98). The findings of other studies on perioperative nutritional interventions are provided in Table 4 (99-106).

Table 4

Perioperative nutritional interventions

Perioperative outcome Study ID % ovarian cancer Sample (n) Time of intervention Nutritional intervention vs. comparison Results
LOS Pearl et al., 1998 (99) 32.3% 195 Post-op day 1 EOF: clear liquid diet; TOF: nothing by mouth until return of bowel function Significant reduction in LOS in EOF group (P=0.001)
Cutillo et al., 1999 (100) 48.3% 122 Post-op day 1 EOF: clear-fluid diet to semiliquid fibreless diet within 24 h vs. nasogastric tube decompression with oral feeding delayed until the first passage of flatus Significant reduction in LOS in EOF group (P<0.05)
Celik et al., 2009 (101) 32% 50 2 days pre-op and 7 days post-op IEN vs. standard enteral nutrition Significant reduction in LOS in IEN group (P<0.05)
Minig et al., 2009 (102) 87.5% 40 Post-op day 1 EOF: liquid diet in the post-op day 1 and then regular diet; TOF: nothing by mouth until return of bowel function Significant reduction in LOS in EOF group (P=0.022)
Minig et al., 2009 (103) 58% 143 Post-op day 1 EOF: liquid diet in the post-op day 1 and then regular diet; TOF: nothing by mouth until return of bowel function Significant reduction in LOS in EOF group (P=0.006)
Ertas et al., 2013 (104) 36.9% 149 Post-op day 1 until flatus Chewing-gum 3 times/day vs. control Significant reduction in LOS in gum-chewing group (P<0.001)
Güngördük et al., 2017 (105) 39.5% 114 Post-op day 1 3 cups of caffeinated coffee daily (100 mL at 10:00, 15:00, and 19:00) vs. routine care without coffee consumption Significant reduction in LOS in coffee group (P<0.001)
Intestinal function recovery Pearl et al., 1998 (99) 32.3% 195 Post-op day 1 EOF: clear liquid diet; TOF: nothing by mouth until return of bowel function Time to bowel sounds, initiation of clear liquid and regular diets significantly reduced in EOF group (P<0.05)
Cutillo et al., 1999 (100) 48.3% 122 Post-op day 1 EOF: clear-fluid diet to semiliquid fibreless diet within 24 h vs. nasogastric tube decompression with oral feeding delayed until the first passage of flatus Time to resolution of postoperative ileus, first passage of stool, and time elapsed to a regular diet significantly reduced in EOF group (P<0.001)
Ertas et al., 2013 (104) 36.9% 149 Post-op day 1 until flatus Chewing-gum 3 times/day vs. control Time to flatus, defecation, bowel movement, and diet tolerance significantly reduced in gum-chewing group (P<0.001)
Güngördük et al., 2017 (105) 39.5% 114 Post-op day 1 3 cups of caffeinated coffee daily (100 mL at 10:00, 15:00, and 19:00) vs. routine care without coffee consumption Time to flatus and defecation/time to bowel movement/time to toleration of diet
Complication Pearl et al., 1998 (99) 32.3% 195 Post-op day 1 EOF: clear liquid diet; TOF: nothing by mouth until return of bowel function No difference in vomiting and abdominal distention between groups (P>0.05)
Feng et al., 2008 (106) 18.3% 60 Within 6 hours post-op Semiliquid diet followed by regular diet vs. clear-liquid diet or semiliquid diet to regular diet No difference in vomiting incidence between groups (P>0.05)
Celik et al., 2009 (101) 32% 50 2 days pre-op and 7 days post-op IEN vs. standard enteral nutrition Postoperative complications (wound infections, dehiscence) significantly reduced in IEN group (P<0.05)

EOF, early oral feeding; IEN, immuno-enhancing nutrition; LOS, length of stay; P, probability value (used in statistical significance); post-op, postoperative; pre-op, preoperative; TOF, total oral feeding.

Prehabilitation during neoadjuvant chemotherapy (NACT) has been associated with improved nutritional parameters and postoperative recovery, further supporting the integration of nutritional interventions at various stages of the treatment continuum (107). The BENITA study examined a holistic approach that integrated exercise and nutrition interventions during and after first-line chemotherapy, which had high acceptance and safety among patients (108).

A study has shown that for patients with ovarian cancer undergoing chemotherapy, the use of nutritional risk screening combined with EN support can significantly improve the patient’s body weight and BMI while improving their nutritional status and QoL (97).

Despite the well-established role of nutritional interventions in improving outcomes for ovarian cancer patients, a major limitation in current research is the lack of high-quality RCTs. This gap leads to uncertainty in the clinical decision-making regarding the ideal timing (e.g., preoperative vs. postoperative), delivery route (e.g., oral vs. enteral vs. parenteral), and specific nutrient composition (e.g., protein, omega-3, or immunonutrients). To address this, future research should prioritize well-designed, multicenter RCTs with standardized nutritional endpoints, which would allow for better comparability and integration of evidence into clinical guidelines. Additionally, studies should explore patient subgroups to assess whether different nutritional strategies are needed based on disease stage, treatment regimen, or baseline nutritional status.


Conclusions

Patients with ovarian cancer are often encountered at an advanced stage and are likely to be malnourished, a state which is strongly associated with poor clinical outcomes. Tumor progression, surgery, chemotherapy, and PARP inhibitors can lead to loss of appetite and malnutrition. Early nutritional risk assessment and individualized and comprehensive nutritional management are thus essential for these patients. However, the means to accurately defining malnutrition in patients with ovarian cancer and the optimal timing and regimen selection for nutritional therapy still require further investigation. In the future, high-quality clinical studies are needed to clarify the mechanisms of malnutrition and optimize nutritional treatment strategies to provide comprehensive and integrated treatment for patients with ovarian cancer.


Acknowledgments

None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tcr.amegroups.com/article/view/10.21037/tcr-2025-758/rc

Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-2025-758/prf

Funding: This work was supported by the Zhejiang Provincial Health Science and Technology Program (No. 2022ky625).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-2025-758/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Huang J, Chan WC, Ngai CH, et al. Worldwide Burden, Risk Factors, and Temporal Trends of Ovarian Cancer: A Global Study. Cancers (Basel) 2022;14:2230. [Crossref] [PubMed]
  2. Berek JS, Renz M, Kehoe S, et al. Cancer of the ovary, fallopian tube, and peritoneum: 2021 update. Int J Gynaecol Obstet 2021;155:61-85. [Crossref] [PubMed]
  3. Hennessy BT, Coleman RL, Markman M. Ovarian cancer. Lancet 2009;374:1371-82. [Crossref] [PubMed]
  4. Jelovac D, Armstrong DK. Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin 2011;61:183-203. [Crossref] [PubMed]
  5. Laky B, Janda M, Cleghorn G, et al. Comparison of different nutritional assessments and body-composition measurements in detecting malnutrition among gynecologic cancer patients. Am J Clin Nutr 2008;87:1678-85. [Crossref] [PubMed]
  6. Hilal Z, Rezniczek GA, Klenke R, et al. Nutritional status, cachexia, and anorexia in women with peritoneal metastasis and intraperitoneal chemotherapy: a longitudinal analysis. J Gynecol Oncol 2017;28:e80. [Crossref] [PubMed]
  7. Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009;89:381-410. [Crossref] [PubMed]
  8. Argilés JM, Busquets S, Stemmler B, et al. Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 2014;14:754-62. [Crossref] [PubMed]
  9. von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachexia Sarcopenia Muscle 2010;1:1-5. [Crossref] [PubMed]
  10. Savant SS, Sriramkumar S, O'Hagan HM. The Role of Inflammation and Inflammatory Mediators in the Development, Progression, Metastasis, and Chemoresistance of Epithelial Ovarian Cancer. Cancers (Basel) 2018;10:251. [Crossref] [PubMed]
  11. Poole EM, Lee IM, Ridker PM, et al. A prospective study of circulating C-reactive protein, interleukin-6, and tumor necrosis factor α receptor 2 levels and risk of ovarian cancer. Am J Epidemiol 2013;178:1256-64. [Crossref] [PubMed]
  12. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet 2001;357:539-45. [Crossref] [PubMed]
  13. Argilés JM, López-Soriano FJ, Stemmler B, et al. Cancer-associated cachexia - understanding the tumour macroenvironment and microenvironment to improve management. Nat Rev Clin Oncol 2023;20:250-64. [Crossref] [PubMed]
  14. Pavlova NN, Thompson CB. The Emerging Hallmarks of Cancer Metabolism. Cell Metab 2016;23:27-47. [Crossref] [PubMed]
  15. Woopen H, Richter R, Chekerov R, et al. Prognostic role of chemotherapy-induced nausea and vomiting in recurrent ovarian cancer patients: results of an individual participant data meta-analysis in 1213. Support Care Cancer 2020;28:73-8. [Crossref] [PubMed]
  16. Li Z, Li C, Yang D, et al. Comparing the efficacy and safety of cisplatin and other platinum-based chemotherapies in locally advanced nasopharyngeal carcinoma: a systematic review and meta-analysis. BMC Cancer 2022;22:616. [Crossref] [PubMed]
  17. Choi S, Seo S, Lee JH, et al. Impact of Patient Sex on Adverse Events and Unscheduled Utilization of Medical Services in Cancer Patients Undergoing Adjuvant Chemotherapy: A Multicenter Retrospective Cohort Study. Cancer Res Treat 2024;56:404-13. [Crossref] [PubMed]
  18. Tew WP, Lacchetti C, Ellis A, et al. PARP Inhibitors in the Management of Ovarian Cancer: ASCO Guideline. J Clin Oncol 2020;38:3468-93. [Crossref] [PubMed]
  19. Moore K, Colombo N, Scambia G, et al. Maintenance Olaparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2018;379:2495-505. [Crossref] [PubMed]
  20. Ray-Coquard I, Pautier P, Pignata S, et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med 2019;381:2416-28. [Crossref] [PubMed]
  21. González-Martín A, Pothuri B, Vergote I, et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med 2019;381:2391-402. [Crossref] [PubMed]
  22. Hardesty MM, Krivak TC, Wright GS, et al. OVARIO phase II trial of combination niraparib plus bevacizumab maintenance therapy in advanced ovarian cancer following first-line platinum-based chemotherapy with bevacizumab. Gynecol Oncol 2022;166:219-29. [Crossref] [PubMed]
  23. Coleman RL, Fleming GF, Brady MF, et al. Veliparib with First-Line Chemotherapy and as Maintenance Therapy in Ovarian Cancer. N Engl J Med 2019;381:2403-15. [Crossref] [PubMed]
  24. Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol 2017;18:1274-84. [Crossref] [PubMed]
  25. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med 2016;375:2154-64. [Crossref] [PubMed]
  26. Wu XH, Zhu JQ, Yin RT, et al. Niraparib maintenance therapy in patients with platinum-sensitive recurrent ovarian cancer using an individualized starting dose (NORA): a randomized, double-blind, placebo-controlled phase III trial Ann Oncol 2021;32:512-21. [Crossref] [PubMed]
  27. Swisher EM, Lin KK, Oza AM, et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol 2017;18:75-87. [Crossref] [PubMed]
  28. 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;390:1949-61. [Crossref] [PubMed]
  29. Torosian MH, Daly JM. Nutritional support in the cancer-bearing host. Effects on host and tumor. Cancer 1986;58:1915-29. [Crossref] [PubMed]
  30. Gardy J, Dejardin O, Thobie A, et al. Impact of socioeconomic status on survival in patients with ovarian cancer. Int J Gynecol Cancer 2019;29:792-801. [Crossref] [PubMed]
  31. Meza-Valderrama D, Marco E, Dávalos-Yerovi V, et al. Sarcopenia, Malnutrition, and Cachexia: Adapting Definitions and Terminology of Nutritional Disorders in Older People with Cancer. Nutrients 2021;13:761. [Crossref] [PubMed]
  32. Morton M, Patterson J, Sciuva J, et al. Malnutrition, sarcopenia, and cancer cachexia in gynecologic cancer. Gynecol Oncol 2023;175:142-55. [Crossref] [PubMed]
  33. Miller J, Wells L, Nwulu U, et al. Validated screening tools for the assessment of cachexia, sarcopenia, and malnutrition: a systematic review. Am J Clin Nutr 2018;108:1196-208. [Crossref] [PubMed]
  34. Bullock AF, Patterson MJ, Paton LW, et al. Malnutrition, sarcopenia and cachexia: exploring prevalence, overlap, and perceptions in older adults with cancer. Eur J Clin Nutr 2024;78:486-93. [Crossref] [PubMed]
  35. Škof E, Merlo S, Pilko G, et al. The role of neoadjuvant chemotherapy in patients with advanced (stage IIIC) epithelial ovarian cancer. Radiol Oncol 2016;50:341-6. [Crossref] [PubMed]
  36. Nasser S, Bilir E, Derin X, et al. Pre-Operative Malnutrition in Patients with Ovarian Cancer: What Are the Clinical Implications? Results of a Prospective Study. Cancers (Basel) 2024;16:622. [Crossref] [PubMed]
  37. Can E, Sönmez S, Konal M, et al. Does sarcopenia predict perioperative mortality in patients with advanced ovarian cancer? Eur Rev Med Pharmacol Sci 2022;26:9409-15. [Crossref] [PubMed]
  38. Krebs HB, Goplerud DR. Surgical management of bowel obstruction in advanced ovarian carcinoma. Obstet Gynecol 1983;61:327-30.
  39. DeLong ER, DeLong DM, Clarke-Pearson DL. Comparing the areas under two or more correlated receiver operating characteristic curves: a nonparametric approach. Biometrics 1988;44:837-45.
  40. Donato D, Angelides A, Irani H, et al. Infectious complications after gastrointestinal surgery in patients with ovarian carcinoma and malignant ascites. Gynecol Oncol 1992;44:40-7. [Crossref] [PubMed]
  41. Mattila E, Marttila H, Sahlberg N, et al. Inhibition of receptor tyrosine kinase signalling by small molecule agonist of T-cell protein tyrosine phosphatase. BMC Cancer 2010;10:7. [Crossref] [PubMed]
  42. Goins EC, Weber JM, Truong T, et al. Malnutrition as a risk factor for post-operative morbidity in gynecologic cancer: Analysis using a national surgical outcomes database. Gynecol Oncol 2022;165:309-16. [Crossref] [PubMed]
  43. Benoit L, Boudebza A, Bentivegna E, et al. What is the most pertinent definition of malnutrition in epithelial ovarian cancer to assess morbidity and mortality? Gynecol Oncol 2024;181:12-9. [Crossref] [PubMed]
  44. Yim GW, Eoh KJ, Kim SW, et al. Malnutrition Identified by the Nutritional Risk Index and Poor Prognosis in Advanced Epithelial Ovarian Carcinoma. Nutr Cancer 2016;68:772-9. [Crossref] [PubMed]
  45. Johnston EA, Ibiebele TI, Friedlander ML, et al. Association of Protein Intake with Recurrence and Survival Following Primary Treatment of Ovarian Cancer. Am J Clin Nutr 2023;118:50-8. [Crossref] [PubMed]
  46. Ibfelt EH, Dalton SO, Høgdall C, et al. Do stage of disease, comorbidity or access to treatment explain socioeconomic differences in survival after ovarian cancer? - A cohort study among Danish women diagnosed 2005-2010. Cancer Epidemiol 2015;39:353-9. [Crossref] [PubMed]
  47. Shao Z, Zhu T, Zhang P, et al. Association of financial status and the quality of life in Chinese women with recurrent ovarian cancer. Health Qual Life Outcomes 2017;15:144. [Crossref] [PubMed]
  48. Sulo S, Phillips W, Smith I, et al. Comparing Malnutrition Risk and Burden among Medicare Fee-for-Service (FFS) Versus Medicare Advantage (MA) Patients in US Hospitals. Curr Dev Nutr 2024;8:102513.
  49. Smith AJB, Sharma MH, Powell K, et al. Abstract A001: Financial toxicity and social needs in patients with ovarian cancer: a multi-practice survey. Cancer Res 2024;84:A001.
  50. Arends J, Bachmann P, Baracos V, et al. ESPEN guidelines on nutrition in cancer patients. Clin Nutr 2017;36:11-48. [Crossref] [PubMed]
  51. Reber E, Schönenberger KA, Vasiloglou MF, et al. Nutritional Risk Screening in Cancer Patients: The First Step Toward Better Clinical Outcome. Front Nutr 2021;8:603936. [Crossref] [PubMed]
  52. Baughman C, Norman K, Mukamal K. Adherence to American Cancer Society Nutrition and Physical Activity Guidelines Among Cancer Survivors. JAMA Oncol 2024;10:789-92. [Crossref] [PubMed]
  53. Xing L, Chen R, Qian J, et al. A comparison of three preoperative nutritional assessment methods for predicting ovarian cancer patient prognosis: which is better? Support Care Cancer 2022;30:5221-9. [Crossref] [PubMed]
  54. Stratton RJ, Hackston A, Longmore D, et al. Malnutrition in hospital outpatients and inpatients: prevalence, concurrent validity and ease of use of the 'malnutrition universal screening tool' ('MUST') for adults. Br J Nutr 2004;92:799-808. [Crossref] [PubMed]
  55. Kondrup J, Rasmussen HH, Hamberg O, et al. Nutritional risk screening (NRS 2002): a new method based on an analysis of controlled clinical trials. Clin Nutr 2003;22:321-36. [Crossref] [PubMed]
  56. Mullen JL, Buzby GP, Waldman MT, et al. Prediction of operative morbidity and mortality by preoperative nutritional assessment. Surg Forum 1979;30:80-2.
  57. Ferguson M, Capra S, Bauer J, et al. Development of a valid and reliable malnutrition screening tool for adult acute hospital patients. Nutrition 1999;15:458-64. [Crossref] [PubMed]
  58. Kaiser MJ, Bauer JM, Ramsch C, et al. Validation of the Mini Nutritional Assessment short-form (MNA-SF): a practical tool for identification of nutritional status. J Nutr Health Aging 2009;13:782-8. [Crossref] [PubMed]
  59. de Ulíbarri Pérez JI, González-Madroño Giménez A, González Pérez P, et al. New procedure for the early detection and control of hospital malnutrition. Nutr Hosp 2002;17:179-88.
  60. Kruizenga HM, Seidell JC, de Vet HC, et al. Development and validation of a hospital screening tool for malnutrition: the short nutritional assessment questionnaire (SNAQ). Clin Nutr 2005;24:75-82. [Crossref] [PubMed]
  61. Arribas L, Hurtós L, Sendrós MJ, et al. NUTRISCORE: A new nutritional screening tool for oncological outpatients. Nutrition 2017;33:297-303. [Crossref] [PubMed]
  62. Detsky AS, McLaughlin JR, Baker JP, et al. What is subjective global assessment of nutritional status? JPEN J Parenter Enteral Nutr 1987;11:8-13. [Crossref] [PubMed]
  63. Ottery FD. Definition of standardized nutritional assessment and interventional pathways in oncology. Nutrition 1996;12:S15-9. [Crossref] [PubMed]
  64. Guigoz Y, Vellas B, Garry PJ. Assessing the nutritional status of the elderly: The Mini Nutritional Assessment as part of the geriatric evaluation. Nutr Rev 1996;54:S59-65. [Crossref] [PubMed]
  65. White JV, Guenter P, Jensen G, et al. Consensus statement: Academy of Nutrition and Dietetics and American Society for Parenteral and Enteral Nutrition: characteristics recommended for the identification and documentation of adult malnutrition (undernutrition). JPEN J Parenter Enteral Nutr 2012;36:275-83. [Crossref] [PubMed]
  66. Cederholm T, Bosaeus I, Barazzoni R, et al. Diagnostic criteria for malnutrition - An ESPEN Consensus Statement. Clin Nutr 2015;34:335-40. [Crossref] [PubMed]
  67. de van der Schueren MAE. Global Leadership Initiative on Malnutrition (GLIM): Guidance on validation of the operational criteria for the diagnosis of protein-energy malnutrition in adults. Clin Nutr 2020;39:2872-80. [Crossref] [PubMed]
  68. Nagle CM, Purdie DM, Webb PM, et al. Dietary influences on survival after ovarian cancer. Int J Cancer 2003;106:264-9. [Crossref] [PubMed]
  69. Edefonti V, Decarli A, La Vecchia C, et al. Nutrient dietary patterns and the risk of breast and ovarian cancers. Int J Cancer 2008;122:609-13. [Crossref] [PubMed]
  70. Ravasco P. Nutrition in Cancer Patients. J Clin Med 2019;8:1211. [Crossref] [PubMed]
  71. Paccagnella A, Morassutti I, Rosti G. Nutritional intervention for improving treatment tolerance in cancer patients. Curr Opin Oncol 2011;23:322-30. [Crossref] [PubMed]
  72. Heslin MJ, Brennan MF. Advances in perioperative nutrition: cancer. World J Surg 2000;24:1477-85. [Crossref] [PubMed]
  73. Brard L, Weitzen S, Strubel-Lagan SL, et al. The effect of total parenteral nutrition on the survival of terminally ill ovarian cancer patients. Gynecol Oncol 2006;103:176-80. [Crossref] [PubMed]
  74. Mendivil AA, Rettenmaier MA, Abaid LN, et al. The impact of total parenteral nutrition on postoperative recovery in patients treated for advanced stage ovarian cancer. Arch Gynecol Obstet 2017;295:439-44. [Crossref] [PubMed]
  75. Inui A. Cancer anorexia-cachexia syndrome: current issues in research and management. CA Cancer J Clin 2002;52:72-91. [Crossref] [PubMed]
  76. Goddard LM, Ton AN, Org T, et al. Selective suppression of endothelial cytokine production by progesterone receptor. Vascul Pharmacol 2013;59:36-43. [Crossref] [PubMed]
  77. McCarthy HD, Crowder RE, Dryden S, et al. Megestrol acetate stimulates food and water intake in the rat: effects on regional hypothalamic neuropeptide Y concentrations. Eur J Pharmacol 1994;265:99-102. [Crossref] [PubMed]
  78. Arends J, Strasser F, Gonella S, et al. Cancer cachexia in adult patients: ESMO Clinical Practice Guidelines ESMO Open 2021;6:100092. [Crossref] [PubMed]
  79. Chinese Society of Clinical Oncology (CSCO). Guidelines for Diagnosis and Treatment of Cancer Cachexia (2021 version). Available online: https://www.csco.org.cn/cn/index.aspx
  80. Loprinzi CL, Bernath AM, Schaid DJ, et al. Phase III evaluation of 4 doses of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia. Oncology 1994;51:2-7. [Crossref] [PubMed]
  81. Loprinzi CL, Michalak JC, Schaid DJ, et al. Phase III evaluation of four doses of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia. J Clin Oncol 1993;11:762-7. [Crossref] [PubMed]
  82. Von Roenn JH. Randomized trials of megestrol acetate for AIDS-associated anorexia and cachexia. Oncology 1994;51:19-24. [Crossref] [PubMed]
  83. Loprinzi CL, Ellison NM, Schaid DJ, et al. Controlled trial of megestrol acetate for the treatment of cancer anorexia and cachexia. J Natl Cancer Inst 1990;82:1127-32. [Crossref] [PubMed]
  84. Shah VP, Amidon GL G.L.. Amidon, H. Lennernas, V.P. Shah, and J.R. Crison. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability, Pharm Res 12, 413-420, 1995--backstory of BCS. AAPS J 2014;16:894-8. [Crossref] [PubMed]
  85. Galia E, Nicolaides E, Hörter D, et al. Evaluation of various dissolution media for predicting in vivo performance of class I and II drugs. Pharm Res 1998;15:698-705. [Crossref] [PubMed]
  86. Deschamps B, Musaji N, Gillespie JA. Food effect on the bioavailability of two distinct formulations of megestrol acetate oral suspension. Int J Nanomedicine 2009;4:185-92. [Crossref] [PubMed]
  87. Cilla DD, Gutierrez JL, Femia RA, et al. A Pilot Study Comparing Megestrol Acetate Concentrated Suspension (MA-CS) to Megestrol Acetate Oral Suspension (MA-OS) on Weight Gain and Body Composition in Patients with HIV-Associated Unintended Weight Loss (UWL). Blood 2005;106:1433.
  88. Levy MH, Back A, Benedetti C, et al. NCCN clinical practice guidelines in oncology: palliative care. J Natl Compr Canc Netw 2009;7:436-73. [Crossref] [PubMed]
  89. Roeland EJ, Bohlke K, Baracos VE, et al. Management of Cancer Cachexia: ASCO Guideline. J Clin Oncol 2020;38:2438-53. [Crossref] [PubMed]
  90. Muscaritoli M, Arends J, Bachmann P, et al. ESPEN practical guideline: Clinical Nutrition in cancer. Clin Nutr 2021;40:2898-913. [Crossref] [PubMed]
  91. Lee CM, Kang JH, Go SI. Clinical implication of megestrol acetate in metastatic gastric cancer: a big data analysis from Health Insurance Review and Assessment (HIRA) database. Support Care Cancer 2024;32:249. [Crossref] [PubMed]
  92. Mattox TW. Cancer Cachexia: Cause, Diagnosis, and Treatment. Nutr Clin Pract 2017;32:599-606. [Crossref] [PubMed]
  93. Oakvik J, Ready D. Updates in Cancer-Related Symptom Management of Anorexia and Cachexia Syndrome. Semin Oncol Nurs 2022;38:151254. [Crossref] [PubMed]
  94. Bai Y, Hu Y, Zhao Y, et al. Anamorelin for cancer anorexia-cachexia syndrome: a systematic review and meta-analysis. Support Care Cancer 2017;25:1651-9. [Crossref] [PubMed]
  95. Inui A. Ghrelin: an orexigenic and somatotrophic signal from the stomach. Nat Rev Neurosci 2001;2:551-60. [Crossref] [PubMed]
  96. Advani SM, Advani PG, VonVille HM, et al. Pharmacological management of cachexia in adult cancer patients: a systematic review of clinical trials. BMC Cancer 2018;18:1174. [Crossref] [PubMed]
  97. Mu J, Wu Y, Jiang C, et al. Progress in Applicability of Scoring Systems Based on Nutritional and Inflammatory Parameters for Ovarian Cancer. Front Nutr 2022;9:809091. [Crossref] [PubMed]
  98. Rinninella E, Fagotti A, Cintoni M, et al. Nutritional Interventions to Improve Clinical Outcomes in Ovarian Cancer: A Systematic Review of Randomized Controlled Trials. Nutrients 2019;11:1404. [Crossref] [PubMed]
  99. Pearl ML, Valea FA, Fischer M, et al. A randomized controlled trial of early postoperative feeding in gynecologic oncology patients undergoing intra-abdominal surgery. Obstet Gynecol 1998;92:94-7. [Crossref] [PubMed]
  100. Cutillo G, Maneschi F, Franchi M, et al. Early feeding compared with nasogastric decompression after major oncologic gynecologic surgery: a randomized study. Obstet Gynecol 1999;93:41-5. [Crossref] [PubMed]
  101. Celik JB, Gezginç K, Ozçelik K, et al. The role of immunonutrition in gynecologic oncologic surgery. Eur J Gynaecol Oncol 2009;30:418-21.
  102. Minig L, Biffi R, Zanagnolo V, et al. Early oral versus "traditional" postoperative feeding in gynecologic oncology patients undergoing intestinal resection: a randomized controlled trial. Ann Surg Oncol 2009;16:1660-8. [Crossref] [PubMed]
  103. Minig L, Biffi R, Zanagnolo V, et al. Reduction of postoperative complication rate with the use of early oral feeding in gynecologic oncologic patients undergoing a major surgery: a randomized controlled trial. Ann Surg Oncol 2009;16:3101-10. [Crossref] [PubMed]
  104. Ertas IE, Gungorduk K, Ozdemir A, et al. Influence of gum chewing on postoperative bowel activity after complete staging surgery for gynecological malignancies: a randomized controlled trial. Gynecol Oncol 2013;131:118-22. [Crossref] [PubMed]
  105. Güngördük K, Özdemir İA, Güngördük Ö, et al. Effects of coffee consumption on gut recovery after surgery of gynecological cancer patients: a randomized controlled trial. Am J Obstet Gynecol 2017;216:145.e1-7. [Crossref] [PubMed]
  106. Feng S, Chen L, Wang G, et al. Early oral intake after intra-abdominal gynecological oncology surgery. Cancer Nurs 2008;31:209-13. [Crossref] [PubMed]
  107. Miralpeix E, Sole-Sedeno JM, Rodriguez-Cosmen C, et al. Impact of prehabilitation during neoadjuvant chemotherapy and interval cytoreductive surgery on ovarian cancer patients: a pilot study. World J Surg Oncol 2022;20:46. [Crossref] [PubMed]
  108. Maurer T, Belau MH, von Grundherr J, et al. Randomised controlled trial testing the feasibility of an exercise and nutrition intervention for patients with ovarian cancer during and after first-line chemotherapy (BENITA-study). BMJ Open 2022;12:e054091. [Crossref] [PubMed]
Cite this article as: Shao Z, Fu L, Lu Y, Zhou H, Zhu Y, Zhu T. Ovarian cancer and malnutrition: a literature review. Transl Cancer Res 2025;14(5):3239-3254. doi: 10.21037/tcr-2025-758

Download Citation