Obesity, a prevalent and multifactorial disease, is linked to a range of metabolic abnormalities, including insulin resistance, dyslipidemia, and chronic inflammation. These imbalances not only contribute to cardiometabolic diseases but also play a significant role in cancer pathogenesis. The rising prevalence of obesity underscores the need to investigate dietary strategies for effective weight management for individuals with overweight or obesity and cancer. This European Society for the Study of Obesity (EASO) position statement aimed to summarize current evidence on the role of obesity in cancer and to provide insights on the major nutritional interventions, including the Mediterranean diet (MedDiet), the ketogenic diet (KD), and the intermittent fasting (IF), that should be adopted to manage individuals with overweight or obesity and cancer. The MedDiet, characterized by high consumption of plant-based foods and moderate intake of olive oil, fish, and nuts, has been associated with a reduced cancer risk. The KD and the IF are emerging dietary interventions with potential benefits for weight loss and metabolic health. KD, by inducing ketosis, and IF, through periodic fasting cycles, may offer anticancer effects by modifying tumor metabolism and improving insulin sensitivity. Despite the promising results, current evidence on these dietary approaches in cancer management in individuals with overweight or obesity is limited and inconsistent, with challenges including variability in adherence and the need for personalized dietary plans.

Obesity, a chronic and multifactorial disease, has reached epidemic proportions globally, significantly impacting public health. Its association with various metabolic abnormalities, including insulin resistance, dyslipidemia, and chronic inflammation, is well documented [1]. These metabolic imbalances are not only linked to the development of cardiovascular diseases and type 2 diabetes but also play a critical role in the pathogenesis of several types of cancer [2, 3]. With rising obesity rates, there is an urgent need to explore effective dietary strategies for weight management in individuals with cancer and cancer prevention in individuals with overweight or obesity.

Given the complex relationship between diet, obesity, and cancer, there is a growing interest in understanding how nutritional interventions can be integrated into cancer prevention and treatment strategies [4]. Numerous clinical trials are currently underway to assess the effectiveness of different nutritional interventions in cancer patients, particularly those with obesity-related cancer.

Adopting a balanced diet rich in plant-based foods has been associated with a reduced risk of developing various types of cancer [5]. The Mediterranean Diet (MedDiet), in particular, characterized by a high intake of fruits, vegetables, legumes, whole grains, and moderate consumption of extra virgin olive oil, fish, and nuts, has been correlated with a decreased risk of several cancers [6]. This dietary model not only promotes a healthy body weight but also provides nutrients and bioactive compounds that may have synergistic protective effects against cancer development [6].

Among other dietary interventions, the ketogenic diet (KD) has recently garnered significant attention. Initially developed to manage drug-resistant epilepsy in children, KD has shown potential benefits in promoting weight loss and improving metabolic profiles in individuals with obesity [7]. By drastically reducing carbohydrate intake, KD induces a state of ketosis, where ketone bodies become the primary energy source, potentially exerting anticancer effects by altering tumor metabolism and reducing insulin levels [8].

Intermittent fasting (IF) is another dietary approach that has gained popularity for obesity management [9]. IF involves periodic cycles of fasting and eating, which can improve insulin sensitivity, reduce inflammation, and promote weight loss [9]. Variants of IF have shown promise in improving metabolic health and a potential role in the prevention and treatment of cancer [10].

However, current evidence on the role of dietary interventions like KD and IF in cancer management is limited and inconsistent. Challenges include the lack of standardized nutritional guidelines, variability in dietary adherence, and the need for personalized dietary plans based on individual metabolic and genetic profiles. Therefore, well-designed clinical trials and comprehensive nutritional assessments are essential to establishing the efficacy and safety of these dietary interventions in cancer care.

This position statement of the European Society for the Study of Obesity (EASO) aimed to provide an overview of the current evidence on the role of obesity in cancer and the major nutritional interventions in the treatment of obesity-related cancers, highlighting ongoing clinical trials and potential mechanisms through which these diets may influence management and prognosis. Then, it aimed to provide clinical insights that should be adopted in the medical nutritional management of individuals with overweight or obesity and cancer.

Several epidemiological studies have shown an association between obesity and cancer in terms of increased incidence, risk of progression, and relapse [11‒13]. Experimental and clinical data provided a possible cause-and-effect relationship, but further studies are needed to prove a causality process linking obesity and cancer [14]. The Continuous Update Project (CUP) database of the American Institute for Cancer Research (AICR) is an ongoing program that analyzes global research on how diet, nutrition, and physical activity affect cancer risk and survival and provides a continuous update of 17 different types of cancer: endometrial, ovarian, esophageal, stomach, postmenopausal breast cancer, liver, colorectal, kidney, gallbladder, pancreatic, prostate, bladder, skin, lung, mouth, pharynx, and larynx cancer [15]. Among these types of tumors, in the present study, we will consider the main obesity-related cancers: endometrial, ovarian, esophageal, stomach, postmenopausal breast cancer, liver, colorectal, kidney, gallbladder, pancreatic, and prostate.

Obesity and Cancer: Epidemiology and Relative Risk

Breast cancer is the most common cancer in women worldwide and is associated with a greater risk of developing in postmenopausal women with obesity, especially for BMI >35.0 kg/m2 [16]. The relative risk (RR) was 1.40 (CI: 1.31–1.49) with a 10 kg/m2 increase in BMI [17] or 1.12 [95% confidence intervals (CIs) 1.08–1.16] with a 5 kg/m2 increase in BMI [12]. It is worthy to note that obesity, both before and after 30 years of age, decreases the risk of premenopausal breast cancer and especially the risk of all estrogen receptor/progesterone receptor/human epidermal growth factor receptor 2 (HER2)-defined subtypes of breast cancer [18, 19]. In this context, obesity is inversely associated with progesterone receptor and estrogen receptor-positive premenopausal breast cancer [20]. In contrast, obesity is a risk factor for postmenopausal hormone receptor-positive breast cancer [16, 21]. Among women, endometrial cancer is more strongly associated with obesity than any other cancer type [22]. The RR was 2.89 (95% CI: 2.62–3.18) with a 10 kg/m2 increase in BMI [17] or 1.59 (95% CI: 1.50–1.68) with a 5 kg/m2 increase in BMI [12]. Compared with women with a normal weight, the RR of mortality for women with moderate obesity (BMI 35–39.99 kg/m2) is 2.53, and for severe obesity (≥40 kg/m2) is 6.25 [11]. Moreover, obesity is also associated with a worse outcome [23]. Patients with obesity show more complications related to surgery, radiation, and chemotherapy, and both systemic chemotherapy and endocrine therapy are less effective [24]. Although the association is not as strong as with endometrial cancer, obesity is also related to the risk of developing ovarian cancer [25]. The RR was 1.14 (95% CI: 1.03–1.27) with a 10 kg/m2 increase in BMI or 1.03 (95% CI: 0.99–1.08) with a 5 kg/m2 increase in BMI [12]. The visceral fat seems more linked to the development of ovarian cancer than BMI per se [26]. There are several risk factors for esophageal adenocarcinoma (EAC) and gastroesophageal junction cancer. The RR for EAC occurrence is 1.52 (95% CI: 1.33–1.74) for each 5 kg/m2 increase in BMI in men and 1.51 in females [12]. Obesity is not linked to EAC only due to an anatomic matter since abdominal obesity is associated with Barrett’s esophagus even after adjusting for the presence of reflux. A multicenter study demonstrated an association of visceral adiposity, measured by computed tomography, with esophageal and junctional adenocarcinomas [27]. In Western countries, a large amount of hepatocellular carcinoma (HCC) developed from metabolic dysfunction-associated steatotic liver disease, which is in turn closely associated with obesity and type 2 diabetes mellitus [28]. Therefore, obesity was related to increased cirrhosis, HCC, and risk of death, particularly for male patients with moderate obesity (RR 4.52 [95% CI: 2.94–6.94]) [11]. Over 40% of kidney (renal cell) cancer (RCC) seems to be associated with obesity assessed based on BMI. The RR was 1.24 (95% CI: 1.15–1.34) for men and 1.34 (95% CI: 1.25–1.43) for women with a 5 kg/m2 increase in BMI [12]. The RR of the highest BMI category versus normal BMI was 1.8 (95% CI: 1.7–1.9), according to a strong body of evidence [13]. A large, prospective study revealed that weight gain until 50 years of age strongly correlated with the incidence of RCC [29]. The Metabolic Syndrome and Cancer Project (Me-Can) showed that increased levels of BMI, blood pressure, glucose, and triglycerides were associated with increased risk of RCC among men, while high BMI was most important in women [30]. The relationship between obesity and colorectal cancer (CRC) development has been evaluated in several epidemiological studies [11‒13]. The RR was 1.24 (95% CI: 1.20–1.28) for men and 1.09 (95% CI: 1.05–1.13) for women with a 5 kg/m2 increase in BMI, and the association was stronger in men than in women [12, 31]. The RR for CRC was higher in patients with obesity in comparison to patients with BMI <25 kg/m2 [32]. In a large meta-analysis, higher BMI was significantly associated with more favorable CRC outcomes, even though higher BMI/obesity is an established determinant for the development of CRC [33]. The impact of obesity on the incidence of gallbladder cancer is more pronounced in women (men 11%, RR 1.09 [95% CI: 0.99–1.21], women 42%, RR 1.59 [95% CI: 1.02–2.47]) and the RR of death from gallbladder cancer for a BMI 30.0–34.99 kg/m2 was 2.13 for women and 1.76 for men [11]. Beyond molecular mechanisms, obesity may act indirectly by increasing the risk of gallstones [34]. The RR for pancreatic cancer occurrence was 1.16 (95% CI: 1.05–1.28) for each 5 kg/m2 increase in BMI in men and 1.10 (95% CI: 1.02–1.09) in women [35] with an increased mortality in female with BMI >40 kg/m2 (female RR 2.76 [1.74–4.36]). The relationship between BMI and prostate cancer has also been much discussed [12, 36]. Cancer-related mortality for male patients with moderate obesity showed a RR of 1.34 (95% CI: 0.98–1.83) [11]. While a high BMI was positively related with nonmetastatic high-grade prostate cancer, it was inversely related with the risk of low-grade tumor [37].

In summary, obesity significantly increases the risk for various cancers, especially in postmenopausal women. It is strongly associated with breast, endometrial, colorectal, and kidney cancers, among others. The presence of visceral fat plays a critical role in cancer development, particularly in ovarian and esophageal cancers. Additionally, obesity influences cancer prognosis, often leading to poorer outcomes and complicating treatment efficacy. The relationship between obesity and cancer is complex, with some cancers showing differing risk profiles depending on factors like fat distribution and tumor type.

Obesity and Cancer: Mechanisms

The underlying mechanisms explaining the association between obesity and cancer consider adipose tissue and adiponiche (i.e., adipose tissue stem niche) as the main drivers. During the expansion of adipose tissue, the adiponiche become dysfunctional, contributing to the development of obesity and metabolic complications [38‒41]. Hypoxia occurs when the rapid expansion of adipose tissue has not been supported by an adequate capillary network [42]. Hypoxia induces pro-angiogenic factors, such as vascular endothelial growth factor and hypoxia-inducible factor 1α, that have been implicated in breast cancer biology [43]. Moreover, hypoxia stimulates adipose tissue to produce other proinflammatory cytokines, such as tumor necrosis factor α (TNF-α), that enhance fibrosis, a process involved in carcinogenesis. The dysfunctional adipose tissue induces also a dysfunctional immune system with increased proinflammatory M1-polarized adipose tissue macrophages, reduced levels of regulatory T cells, eosinophils and their anti-inflammatory interleukin (IL)-4 and IL-13, and M2-polarized anti-inflammatory adipose tissue macrophages [41]. Furthermore, the inflammasomes, activated by fatty acids and high glucose levels, induce inflammatory IL-1β and IL-18, promoting an inflammatory microenvironment surrounding cancer cells, in particular in CRC [44, 45]. Adipose stromal/stem cells are influenced by microenvironment and cell-to-cell interactions to support physiological adipose tissue expansion [41, 42]. An increase of adipose stromal/stem cells, present in obesity, was also described on CRC and seems to sustain tumor growth [46]. Adipocytes are the most important cells of the adipose tissue implicated in the modulation of the microenvironment and thus of the tumor growth through paracrine, autocrine, and endocrine ways. Adipocyte-derived factors include adipokines, extracellular matrix components, hormonal factors, growth factors, and inflammatory cytokines [47]. In mice, loss of adiponectin enhances the development of colitis-associated CRC [48], while adiponectin released from perinephric adipose tissue may impact RCC aggressiveness [30]. On the other hand, leptin induces TNF-α and IL-6 and, in an in vitro study, stimulated lipid droplets, increasing cell proliferation [49]. The insulin growth factor (IGF) pathway, which is upregulated in obesity, demonstrated a crucial role in the development of EAC. Studies have also found associations between leptin/adiponectin and Barrett’s esophagus presence and progression to EAC. Moreover, peritumoral adipose tissue in patients affected by EAC showed increased adipocyte size and inflammation markers, high levels of leptin, increased angiogenesis, and was associated with lymph node invasion. The leptin receptor G2548A mutation is statistically significantly associated with an increased risk of prostate cancer. Moreover, patients with high-volume prostate cancer had higher serum leptin levels [37]. The chronic low-grade inflammation characterizes not only the visceral and subcutaneous adipose tissue but also the breast fat, and it is associated with activation of signaling pathways, including LKB1/AMPK, p53, hypoxia-inducible factor 1α, and PKM2. This produces the increased levels of aromatase, which usually limit estrogen biosynthesis, causing the production of estrogens, particularly estradiol, determining breast cancer growth and progression [50‒53]. Interestingly, it was found an involvement in creatine synthesis by peritumoral adipocytes in the development of breast cancers in obese mice [54]. Creatine is implicated in T-cell development and macrophage polarization [55] and is transported into the cancer cells, producing cell growth and larger tumors, possibly due to an increased cell energy supply [54]. This is consistent with the observation that mitochondrial creatine kinase is stabilized in HER2+ breast cancer, increasing cytosolic phosphocreatine and thus cytosolic energy supply [56]. It was recently demonstrated that casein kinase 2 covers a pivotal role in obesity, stimulating adipocyte hypertrophy/hyperplasia and adipogenesis, resulting in the adipose tissue expansion [57]. Casein kinase 2, usually overexpressed in cancer, contributes to the regulation of aberrant lipid metabolism in malignant cells, leading to upregulation of protein kinase B (AKT), Wnt, and extracellular signal-regulated kinase and to downregulation of tumor suppressor molecules [58, 59]. The involvement of microbiota dysregulation in obesity and related diseases, such as cancer, was described [38]. Short-chain fatty acids (SCFAs) produced by bacteria are decreased in obesity, with a reduction in triglyceride hydrolysis, free fatty acid oxidation, beige adipogenesis, and mitochondrial biogenesis [60, 61]. Indeed, microbiome-derived peptides modulate immune cell activity, and leakage of lipopolysaccharides activates Toll-like receptor 4 and nuclear factorkB, leading to inflammatory-induced carcinogenesis in the CRC and HCC [62, 63]. In summary, the adipose organ and cancer crosstalk, throughout several mechanisms that adipose organ and tumor share, determines the role of obesity in cancer development, progression, invasiveness, and chemoresistance.

Excess adipose tissue is known to be an independent risk factor for cancer. Adopting a healthier lifestyle can lead to a lower body weight; however, an overall healthy dietary pattern may also contribute to cancer prevention. A healthy dietary pattern includes a high intake of vegetables, pulses, berries, fruits, whole grains, nuts, seafood, and fish, low-fat dairy, and a low intake of red meat and processed meat [64]. Furthermore, a healthy dietary pattern is low in sugary foods and beverages, as well as in ultraprocessed starch and fats. The energy intake in balance with energy expenditure is one of the components in a healthy diet. A diet rich in various bioactive compounds, such as in the MedDiet, may have a favorable impact on obesity and cancer risk. The best healthy dietary strategy can be sustained over a long period, leading to improved health and weight outcomes for people living with obesity [65]. Registered dietitians can provide medical nutrition therapy to support people in adopting individualized, realistic, and sustainable healthy eating patterns as a part of a multidisciplinary obesity care plan [66]. It should be recognized that the modern food environment with unlimited access to highly palatable, energy dense foods often challenges dietary patterns and weight management.

Fruits, vegetables, and pulses are crucial components of a healthy diet and recommended by the World Health Organization (WHO) [67] and the World Cancer Research Fund (WCRF) for cancer prevention [68]. Intake of more than 400 g per day of fruits, berries, and non-starchy vegetables has health benefits and may reduce the cancer risk. Fruits and vegetables are rich in dietary fiber, as well as in a variety of vitamins, minerals, antioxidants, and bioactive compounds. Antioxidants and phytochemicals such as alkaloids, flavonoids, polyphenols, vitamin C, and E may have potential benefits in cancer prevention in reducing inflammation and acting as an anticarcinogen in scavengingfree radicals [69]. It is recommended that people consume a diverse range of colorful vegetables, fruits, and berries to maximize their nutrient intake because the content of these nutrients can vary significantly. Pulses, such as beans and lentils, are also essential sources of dietary fiber, proteins, and other nutrients. Many vegetables have a high content of water and dietary fiber, contributing to the volume and low energy density in a meal and may facilitate weight management, thereby reducing the risk of cancer.

Foods rich in whole grains are also important for cancer prevention, especially for reducing the risk of CRC. Examples of whole grain-rich foods include unprocessed maize, oats, rye, barley, millet, and wheat. When whole grains are processed, the nutrient-dense bran and germ are removed, which reduces the amount of fiber and other beneficial nutrients.

Furthermore, a high intake of dietary fiber (25–29 g) in people with or without chronic disease has been associated with a lower body weight and lower risk of CRC, breast cancer, a total cancer, and cancer mortality in a large systematic review and meta-analysis [70]. The dose-response findings were similar for dietary fiber as well as for whole grains and the risk reduction of cancer. The high fiber content in whole grains, fruits, and vegetables promotes a feeling of fullness and satiety and may therefore facilitate weight management, which is beneficial in cancer prevention. The recommended total daily fiber intake is at least 30 g, according to the WCRF [68].

High consumption of red and processed meat may increase the risk of CRC [68]. Red meat includes, e.g., pork, beef, and lamb, whereas white meat includes poultry, such as chicken and turkey. Processed meat is preserved by smoking, salting, curing, or adding chemical preservatives, such as bacon and sausages. For cancer prevention, it is recommended to limit the consumption of processed meat as much as possible, and the intake of red meat should be a maximum of 350–500 g per week in cooked weight [68]. However, meat also provides valuable nutrients such as iron, zinc, vitamin B12, and protein and can be part of a healthy diet. It is particularly important to consider when providing medical nutrition therapy after metabolic and bariatric surgery.

Alcohol consumption increases the risk of various types of cancer, including breast cancer, mouth and pharynx and larynx cancers, CRC, stomach cancer, liver cancer, kidney cancer, and esophageal cancer, according to the WCRF [68]. There is no established minimum safe level of alcohol consumption, and it is recommended that alcohol intake be kept to a minimum to reduce the risk of cancer. Alcohol has a high energy content (7 kcal/g) and can therefore contribute to increased energy intake and challenges in weight management [68].

There are recommendations that people should not use dietary supplements for cancer prevention [68]. However, people living with obesity may sometimes require dietary supplementation with vitamins and minerals such as vitamin D, vitamin B12, calcium, and iron to meet their nutritional needs. This is the case, for example, after metabolic and bariatric surgery since the intake and absorption of these nutrients may be reduced [68].

The intake of sugar-sweetened drinks should be restricted because these beverages can contribute to challenges in weight management [68]. Increased body weight is associated with cancer. Drinking an adequate amount of water helps to maintain a sufficient level of hydration and is the optimal beverage choice. The shift from sugar-sweetened drinks to beverages with artificial sweeteners may help reduce energy intake in people who consume sugar-sweetened beverages. A systematic review and meta-analysis of 17 randomized controlled trials reported that changing sugar-sweetened drinks to artificially sweetened beverages may contribute to a small improvement in weight status [71]. Nevertheless, the WHO has recently recommended that artificial sweeteners should not be used to control body weight owing to the limited evidence, primarily from observational studies [72]. Instead, the focus should be on enhancing overall healthy eating patterns and decreasing free sugar intake. The artificial sweetener aspartame was recently classified as “possible carcinogenic to humans” (group 2B) by the International Agency for Research on Cancer (IARC) [73]. The classification was based on limited evidence, and group 2B is the lowest evidence category for carcinogens. The WHO has not changed the acceptable daily intake of aspartame (40 mg/kg body weight/day) based on the new classification. The European Food Safety Authority's (EFSA) evaluation of aspartame safety and acceptable daily intake recommendations aligns with the conclusion of the WHO [74].

Mediterranean Diet

Historically, the MedDiet has been initially proposed as a healthy pattern diet [75] that combines certain foods and nutrients that interact synergically between each other, and has been revealed to be associated with a reduction in the risk of cardiovascular diseases and overall mortality [76]. Subsequently, several epidemiological studies have shown a strong correlation between a higher adherence to the MedDiet and a lower incidence (i.e., new onset and prevention) of several forms of cancer (colorectal, breast, stomach, pancreas, prostate, and lung cancer) [77‒79]. On the other hand, the period during cancer (the active phase), or in other words, immediately after diagnosis and during treatment, remains critical and crucial [80] since important health-related outcomes occur due to the nature of the cancer by itself or as an adverse consequence of treatment (e.g., chemotherapy, radiotherapy, etc.) [80]. Most commonly, weight loss and cachexia occur in the course of cancer treatment; however, with the aim of preventing or treating malnutrition in this population, strong nutritional evidence for adults with cancer is available, and guidelines have been established [81]. Interestingly, in certain cancers and/or treatments, different kinds of changes are observed, such as weight gain [82], increased body fat mass and its central visceral deposition [83], as well as a reduction in muscle mass and strength, leading to sarcopenic obesity in this population (that is, with cancer and obesity). This unavoidably increases the development of cardiometabolic risk factors (metabolic syndrome/obesity, hyperglycemia/diabetes, dyslipidemia, and hypertension) [84, 85], as well therapy-induced inflammation [86]. In this scenario, the treatment of patients with cancer becomes extremely challenging [87], especially as excess body weight (i.e., obesity) represents one of the main factors for a poorer prognostic outcome for certain cancers (chemotherapy resistance, rate of remission and recurrence, survivorship and mortality, etc.) [87]. In this case, evidence for dietary interventions during cancer treatment is very limited [88], as no clear nutritional indications are available in this population (cancer and obesity). Additionally, during this particular period, namely, immediately after diagnosis, as well as during cancer treatment, in the past few years, emerging data have proposed the MedDiet to have the potential to improve the clinical and supportive-care outcomes in adults with cancer [89]. However, the evidence available regarding the effects of the MedDiet in patients with active cancer has not been extensively evaluated, especially in people with obesity [90].

With this aim in mind, some recent randomized controlled trials have been conducted on people with cancer and obesity (mean BMI≈30 kg/m2) [91‒97]. In general, according to the available data, it can be summarized, as the MedDiet in these studies was delivered over a mean period of 6 months, and it appeared to be safe and feasible in people with cancer (breast, lung, and prostate) while coexisting with obesity, due to no/very few adverse events, as well as high adherence and low attrition (i.e., dropout) rates [91‒97]. Moreover, the dietary composition and patterns used in these studies were mainly calorie-restricted [91, 94], rich in fibers (whole grains, legumes, fruit, and vegetables) [91, 95, 98], and monounsaturated fatty acids (olive oil), as well as low in saturated fatty acids [91, 95, 97, 98], and were revealed to have beneficial effects in terms of health indicators such as anthropometric variables [body weight, waist circumference (WC)] [91‒93, 95‒97], body composition (fat and lean mass) [91, 95, 97], and biomarker indicators (inflammation, glycemic and lipid profiles) [91‒93, 95, 96, 98, 99], as well as other cancer outcomes (i.e., quality of life) [91, 93‒96, 99].

For instance, the MedDiet approach in people with cancer (mainly breast but also prostate cancer) and obesity appears to be associated with a significant reduction in body weight [91, 95‒97], WC [95, 97], and total body fat [91, 95, 97], but, in some cases, it may lead to a reduction in lean mass (LM) [91, 100], when compared with patients who were in “usual care” during which they were not on a clear dietary intervention, resulting in being more prone to regaining weight with significantly increased BMI [97] and WC [92] or, in the best scenario, no changes were detected in the anthropometric variables and body composition compartments in these arms (i.e., no increase) [95]. Moreover, in patients with breast, lung, and prostate cancer, the MedDiet was found to be associated with improvements in certain biomarkers, such as those related to lipid and glycemic profiles, as well as inflammation. Thus, the MedDiet has been related to a reduction in blood triglycerides [96], glycemia [97, 98], and IL-8 [91], and the advanced lung cancer inflammation index (ALI) [98], as well as an increase in high-density lipoprotein cholesterol levels [95‒97]. On the other hand, the “usual care” arms reported no ameliorative changes in these biomarkers, or in some studies, led to a deterioration of some of these parameters, such as an increase of total cholesterol and low-density lipoprotein cholesterol [101]. Moreover, the MedDiet appeared to improve patients’ general condition in terms of global health and quality of life [95], reduction of stress [94] and cancer-related fatigue [93], even in the absence of significant changes in body weight status (i.e., reduction). Last but not least, very recent data deriving from a small cohort study (i.e., not a randomized controlled trial) of patients with advanced melanoma, revealed that higher adherence to the MedDiet was associated with a higher probability of response to treatment with immune checkpoint blockade (ICB) [102]. ICB is a relatively new and highly successful treatment against severe cancers used in immunotherapy to enhance the immune system [103].

The mechanisms behind the beneficial impact of MedDiet on both cancer (i.e., during the active phase) and obesity outcomes are complex and still not fully understood [104]. However, we speculate that some nutrients, specifically polyphenols (e.g., olive oil, red wine, nuts and seeds), fibers (e.g., whole grains, fruits, and vegetables), and omega-3 fatty acids (e.g., fish), that are a significant part of the MedDiet, may explain and could be behind these benefits.

For example, several preclinical investigations showed the positive effect of polyphenols (i.e., curcumin) in limiting weight gain [105] and the decrease of fat storage [106], and for these two properties, it has been proposed as an anti-obesity strategy; however, clinical studies are still limited [107]. In terms of cancer, the dietary polyphenols (flavonoids, phenolic acids) are known to prevent cancer (i.e., lung, gastric, colorectal, breast, and prostate), through antioxidant and anti-inflammatory mechanisms, as well as the modulation of multiple molecular events involved in carcinogenesis [108]. In addition to that, some early clinical investigations found that natural polyphenols can be considered as potential candidates for anticancer therapy by acting on premalignant forms (i.e., familial adenomatous polyposis [curcumin and quercetin]) [109], or as a coadjutant to traditional cancer treatments [110, 111], for example, by enhancing the efficacy of radiotherapy in breast cancer, overcoming chemotherapy resistance in pancreas cancer (epigallocatechin gallate [curcumin]), or reducing the recurrence rate (colon rectal cancer [flavonoid]) [112].

Secondly, the MedDiet is characterized by a high intake of fibers, which seems to act on the intestinal microbiota, modulating its “composition” and “activity” in terms of the production of metabolites regulating immune function and inflammatory pathways [113]. In particular, a high-fiber diet, such as the MedDiet, seems to increase the number of intestinal bacterial species responsible for the production of SCFAs (acetate, propionate, and butyrate) [114, 115], important for the proper functioning of the immune system and the prevention of inflammatory diseases [115]. Recently, the same SCFAs were proposed to have a protective effect against obesity by increasing appetite control and satiety, as well as energy expenditure; however, such a relationship needs further confirmation [116, 117]. In terms of cancer, SCFAs seem to induce cell apoptosis and suppress cell proliferation and metastasis in several types of cancers (lung, breast, bladder, gastric cancer), but mainly CRC [118]. Moreover, they reduce anticancer drug resistance, such as that associated with anticancer drug chemotherapy [118].

Finally, the MedDiet ensures a high intake of omega-3 polyunsaturated fatty acids (PUFAs). While the anti-inflammatory and triglyceride-reducing properties of long-chain PUFAs are well established [119], the anti-obesity effect in humans remains under debate since published research suggests that even if omega-3 PUFAs may not be helpful in losing body weight, they are able to limit weight regain in the longer term, and for this reason they can be useful during the weight loss maintenance phase [120]. In terms of cancer, epidemiological studies link high dietary PUFAs intake with a reduced incidence of cancer (colorectal, breast) [121, 122]. Moreover, in breast cancer, the role of eicosapentaenoic acid and docosahexaenoic acid supplementation as an adjunctive measures has been explored in reducing chemotherapy-associated weight gain and muscle loss (sarcopenic obesity) [122, 123]. In the same direction, dietary PUFAs may improve the efficacy and tolerability of cancer chemotherapy drugs (in CRC) [121]. Finally, higher intakes of eicosapentaenoic acid and docosahexaenoic acid from dietary sources were reported to be associated with a reduction in breast cancer recurrence and improved overall mortality in early-stage breast cancer [123, 124].

Recommendations

The MedDiet is ideal for almost all cancer patients, particularly during treatment phases, as it helps manage weight, cachexia, and improves quality of life (Fig. 1). It is also beneficial for those at high risk of metabolic complications, including metabolic syndrome, dyslipidemia, and hypertension. However, it should be avoided by those with severe allergies to key MedDiet components (such as nuts) and adjusted for patients undergoing specific cancer treatments based on their guidelines. Individuals with gastrointestinal issues affecting nutrient absorption may need a modified version of the diet. The MedDiet should consist of 45–55% of daily calories from complex carbohydrates (whole grains, legumes, fruits, vegetables), 15–25% from lean proteins (fish, poultry, legumes, nuts), and 25–35% from healthy fats (olive oil, nuts), with a focus on limiting saturated fats. Aim for a daily fiber intake of 25–35 g from high-fiber foods to support overall health and manage both obesity and cancer outcomes.

Fig. 1.

Favorable mechanisms of the Mediterranean diet in the patient with obesity and cancer.

Fig. 1.

Favorable mechanisms of the Mediterranean diet in the patient with obesity and cancer.

Close modal

Ketogenic Diet

KD is a dietary pattern characterized by high fat intake, moderate-to-low protein consumption, and very low-carbohydrate intake (<50 g) [8]. Initially, the KD was employed to treat drug-resistant epilepsy in children, but it has now gained widespread acceptance for obesity management. Due to the connection between obesity, diet, and cancer, there is a growing interest in using the KD as an adjuvant therapy in cancer treatment [125, 126]. Preclinical and some clinical trials have shown that the KD can be cost-effective and relatively well tolerated, with the potential to improve metabolic abnormalities, reduce inflammation, limit tumor growth, and protect healthy cells from the damage caused by chemotherapy and radiation [126].

With a diet that limits intake of carbohydrate, glucose availability for glycolysis is limited, preventing the formation of pyruvate and subsequent conversion to lactate to generate ATP [127]. Lipid and ketone bodies become the predominant source of energy and require the cell to use mitochondria, which may be dysfunctional in cancer cells as noted above. This, combined with the limited availability of glucose for the pentose phosphate pathway and the generation of NADPH, leads to oxidative stress in cancer cells compared with normal cells [127]. Evidence of this oxidative stress was demonstrated in mouse models of neuroblastoma through measurement of activation of energy sensor adenosine monophosphate (AMP)-activated protein kinase (AMPK) [128]. In response to KD, they found higher activation of AMPK in tumor tissue but not in normal tissue [128]. It is important to note that a few mitochondrial enzymes are key in the metabolism of ketone bodies for energy, and tumor cells tend to express these enzymes in varying amounts. Zhang et al. [129] demonstrated that expression of genes encoding the two ketolytic enzymes 3-hydroxybutyrate dehydrogenase 1 (BDH1) and succinyl-CoA:3-oxoacid CoA transferase 1 in tumor cells correlated with response to KD.

In addition to these metabolic benefits, KD, perhaps through the action of ketone bodies, can exert several other benefits, including modulation of signaling molecules and gene expression, as well as reduction in inflammation [126]. β-hydroxybutyrate and acetone have been noted to modulate the signaling of N-methyl-D-aspartate, which is physiologically relevant as N-methyl-D-aspartate receptor expression has been observed in various types of cancers [130]. Similarly, hydroxy-carboxylic acid receptor 2 is activated by β-hydroxybutyrate [131]. Hydroxy-carboxylic acid receptor 2 is described as a tumor suppressor and activates specific macrophages that have neuroprotective effects [131]. KD and ketone bodies have also been associated with anti-inflammatory effects through reduction in cytokines such as TNF-α, IL-1, and IL-6 [126]. A recent study in a mouse model of colon cancer noted that a KD was associated with lower tumor weight, as well as plasma IL-6 levels [132]. Additionally, blood ketone body concentrations were also negatively correlated with tumor weight [132]. KD also reduces inflammation through suppression of NLRP3 inflammasome, which is a multiprotein complex that controls the activation of caspase-1 and subsequent release of proinflammatory cytokines [133]. β-hydroxybutyrate was shown to inhibit assembly of NLRP3 inflammasome [133].

Despite these theoretical benefits, clinical trials with KD are quite limited [134]. A recent review of clinical trials regarding use of KD in cancer noted that of the approximately 30 trials, the vast majority were case reports or pilot/feasibility studies, with most focusing on the tolerability of KD in this population as well as its impact on body weight, glucose, and other metabolic parameters [126]. As an example, a clinical trial retrospectively evaluated 53 patients undergoing treatment for high-grade glioma who had adequate glucose values recorded [135]. Six of the 53 patients were on KD during treatment with a micronutrient composition of 77% fat, 8% carbohydrate, and 15% protein. They noted that the mean blood glucose for patients on a standard diet was 122 mg/dL, whereas that of patients on a KD was 84 mg/dL and decreased from 142.5 mg/dL prior to initiation of KD [135]. Another clinical trial randomized 73 women with ovarian or endometrial cancer to either a KD (energy from fat, protein, and carbohydrate of 70:25:5) or a diet recommended by the American Cancer Society [136]. After 12 weeks, KD resulted in lower overall and central fat mass. There was also a 21.2% reduction in visceral fat mass with KD compared with 4.6% with the American Cancer Society diet, along with a more significant reduction in insulin, C-peptide levels, glucose, and IGF-1 levels [136]. KD also showed more significant improvement in physical function scores, reports of fatigue, as well as cravings for starchy foods and fast-food fat [137].

Another randomized clinical trial was designed to enroll three cohorts, including patients with breast, rectal, and head and neck cancer, and included two interventions: ketogenic breakfast with a ketogenic drink and whole food KD [138]. However, ketogenic breakfast drink was not tolerated by many, thus limiting the data to individuals with nonmetastatic breast cancer comparing either whole-food KD (n = 29) or standard diet (n = 30). The authors noted that the KD was well tolerated and resulted in body weight loss of 0.4 kg per week, as well as fat mass. Although fat-free mass and skeletal muscle mass declined initially, they were subsequently preserved. Insulin and IGF-1 levels also decreased more in the KD group [138].

The potential contraindications of KD in patients with cancer come from in vitro studies and some early-stage in vivo evidence [139, 140]. In a preclinical model of tuberous sclerosis complex, a rare genetic disorder, the growth of renal lesions in Eker rats (Tsc2+/−) was evaluated subjected to ad libitum prolonged feeding of KD for 4, 6, and 8 months [139]. The authors demonstrated that, especially in its long-term usage, KD leads to excessive growth of renal tumors by recruiting ERK1/2 and mTOR, which are related to oleic acid accumulation and the overproduction of growth hormone. The authors also reported the exhaustion of the initial adaptive upregulation of some protective proteins such as Nrf2, p53, and 8-oxoguanine glycosylase, suggesting that KD may be contraindicated in patients with tuberous sclerosis complex [139]. KD may also be contraindicated in cancer patients with the BRAF V600E mutation [140]. In a xenograft mouse model, the ketone body acetoacetate selectively enhanced BRAF V600E mutant-dependent MEK1 activation in human tumors, promoting the growth of human melanoma cells expressing this mutation [140].

Additionally, KD can be deficient in selenium, vitamin D, zinc, and other vitamins and minerals [8]. These potential adverse side effects can be prevented or corrected if KD is prescribed and monitored by a qualified nutritionist who selects appropriate patients and ensures that KD is well formulated and adjusted in food choices and supplements with necessary vitamins and minerals [8]. In conclusion, data on the safety and efficacy of KD in cancer patients remain relatively sparse and well designed, and rigorous clinical trials are urgently needed to evaluate the efficacy and safety of KD in the cancer setting.

Recommendations

The KD is suitable for patients with obesity and cancer who seek to manage weight and metabolic abnormalities while undergoing treatment (Fig. 2). It may be beneficial for those needing to improve glucose regulation and reduce inflammation. However, KD may not be appropriate for patients with certain genetic mutations or conditions, such as tuberous sclerosis complex or BRAF V600E mutation, due to potential adverse effects. It may also result in deficiencies of essential nutrients, which can be managed with careful planning and supplementation by a qualified nutritionist. KD should consist of about 70–80% of daily calories from fat, 15–25% from protein, and 5–10% from carbohydrates. However, there are different ketogenic approaches, and the correct macronutrient composition may vary on a case-by-case basis. Regular monitoring and adjustment are necessary to ensure nutritional adequacy and safety.

Fig. 2.

Favorable mechanisms of the ketogenic diet in the patient with obesity and cancer.

Fig. 2.

Favorable mechanisms of the ketogenic diet in the patient with obesity and cancer.

Close modal

Intermittent Fasting

In recent decades, there has been a growing interest in IF as a strategy for managing obesity [141]. IF involves periodic episodes of minimal to no calorie consumption. The variations of IF include complete 24-h fasting every other day or fasting on one or two nonconsecutive days per week, commonly known as the 6:1 and 5:2 diets, respectively. Many IF programs recommend a minimal caloric intake (e.g., 500 kcal daily) during fasting periods, allowing unlimited consumption of calorie-free beverages. Another approach, time-restricted feeding (TRF), limits calorie intake to a specific window of time each day, typically between 4 and 12 h. TRF may also involve calorie restriction during the non-fasting period, potentially offering additional benefits, such as improvements in circadian rhythm [141].

Emerging evidence suggests that fasting could play a key role in cancer treatment by fostering conditions that limit cancer cells’ adaptability, survival, and growth [10]. Fasting could increase the efficacy of cancer treatments and limit adverse events. Yet, we lack an integrated mechanistic model for how these two complicated systems interact, limiting our ability to understand, prevent, and treat cancer using fasting [10].

Clinical studies have been carried out to measure the relevance of various modalities of IF on metabolic and hormonal endpoints correlated to cancer development and prognosis, given their practicality and the beneficial weight reduction effect in individuals with obesity [142]. IF has been demonstrated to improve cancer risk variables in several short-term randomized clinical studies, including lowered levels of glucose, insulin, leptin, and higher adiponectin, which are linked to cancer etiology [142]. TRF has been reported to cause substantial alterations in biochemical indicators linked to weight, including insulin sensitivity and oxidative stress, in small and underpowered investigations [143, 144]. Nonetheless, in one study (n = 116) in individuals with obesity on TRF, no significant change in weight, fasting insulin, or fasting glucose level was reported [145]. Furthermore, evidence shows that, unlike rats, people require weight loss to optimize metabolic health [146]. Whether weight reduction has a causal function in lowering cancer risk and improving prognosis without considerable food composition changes remains a critical but unresolved issue [146].

A few studies have used IF in cancer patients as one of the regimens [147‒150]. In one clinical trial of glioma with 25 patients, a significant decrease in HbA1c, insulin, and fat was observed, while lean body weight and ketone bodies (in the brain) were increased with a well-tolerated ketogenic IF diet [150]. Another study on 23 overweight premenopausal women with a high risk of breast cancer reported that 1 month of IF resulted in a 4.8% weight loss, 8% fat loss, and an improvement in insulin resistance [149]. Furthermore, a study that enrolled 13 stage II breast cancer women receiving (neo) adjuvant chemotherapy suggested that IF was well tolerated, with a lower rate of withdrawals from adverse events [147]. In addition, fasting for a brief time was observed to minimize hematologic damage in women receiving chemotherapy, with the fasting group having considerably increased erythrocyte and thrombocyte levels following treatment. Following chemotherapeutic treatments, women in the control group exhibited higher DNA damage indicators and lowered circulating IGF-1 levels than those who had fasted and had lower IGF-1 levels [147]. In another clinical study of diverse tumor types, fasting for 24, 48, or 72 h before treatment was determined to be possible and safe, with only minor adverse effects, such as weariness, headache, and dizziness [148]. Patients fasting for 48–72 h before the chemotherapeutic treatment had a nonsignificant tendency towards a lower incidence of neutropenia and neuropathy than those who fasted for 24 h before treatment. In peripheral blood mononuclear cells, markers of DNA damage increased in all groups, although to a lesser extent in the extended fasting group [148].

Despite initial concerns about weight loss in patients with cancer receiving chemotherapy, no trials have revealed substantial loss of lean body mass or malnutrition due to IF [151]. IF was found safe and tolerable in an observational study of 28 children with type 1 diabetes with diverse cancers undergoing treatment during Ramadan [152]. In a case series, IF before and after chemotherapy was reported to be secure and well received in 10 patients with a variety of cancers [153]. Finally, in another clinical trial, 25 patients with head or neck cancer undergoing chemotherapy were assigned to either a short-term fasting diet followed by a standard caloric diet or a standard caloric diet followed by a short-term fasting diet [154]. Those patients fasted for 36 h before treatment and 24 h thereafter, having a total of 350 calories per day. Within 8 days of chemotherapy, no substantial weight loss was recorded, although there was an improvement in quality of life and weariness. Such benefits, in contrast, were not seen while having a regular diet [154].

Despite the limited clinical evidence currently available, several ongoing clinical trials are exploring the potential benefits of IF in various advanced cancer scenarios [146]. However, data on the specific nutritional interventions applied in these clinical trials remain incomplete and inconsistent. This includes details on nutritional guidelines, meal quality, and specific foods included in the diets. Comprehensive and standardized information on these aspects is critical to assessing the efficacy and feasibility of IF as a therapeutic strategy in cancer treatment. Further rigorous and well-designed clinical trials, especially in subjects with obesity, are needed to provide conclusive evidence on the role of IF in cancer treatment and survival.

Recommendations

IF may benefit cancer patients, such as those with glioma or early-stage breast cancer, by showing improvements in biomarkers like HbA1c and insulin resistance and potentially reducing side effects during chemotherapy (Fig. 3). Possible minor side effects include fatigue, headache, and dizziness. While IF generally does not cause significant weight loss or malnutrition, ongoing monitoring is essential. IF typically involves minimal caloric intake (e.g., 500 kcal/day) during fasting periods with unlimited calorie-free beverages. TRF limits calorie intake to specific hours, but detailed macronutrient composition varies and should be adjusted to individual needs.

Fig. 3.

Favorable mechanisms of the IF in the patient with obesity and cancer.

Fig. 3.

Favorable mechanisms of the IF in the patient with obesity and cancer.

Close modal

Micronutrient Deficiencies in Subjects with Obesity and Cancer and Recommended Supplementation

Certain micronutrient deficiencies have been implicated in the development of obesity and are frequently present in individuals with obesity, including vitamins (A, B, C, D, E, folic acid) and minerals (iron, zinc, magnesium, selenium, copper, iodine, calcium, and phosphorus) [155]. Potential mechanisms linking such micronutrient deficiencies with obesity involve dysregulation of carbohydrate and fat metabolism, dysfunction of pancreatic β-cell, and abnormalities in the insulin-signaling cascade [155]. Since most micronutrients serve as enzyme co-factors in the body, thus regulating several metabolic processes, their deficiencies can lead to the development of severe health problems (apart from obesity), including cardiovascular diseases, immune dysfunction, osteoporosis, defective antioxidant defense mechanisms, neurodegenerative disorders, and cancer [155].

Focusing on cancer, it has been recognized that nutritional status can greatly influence cancer development and progression [156], as well as anticancer treatment efficacy: both inadequate diet and cachexia can be present and negatively affect clinical outcomes [1]. Malnutrition is the main cause of inadequate micronutrient status in cancer patients since macronutrients serve as “the natural carriers” of micronutrients [157]. However, apart from malnutrition, it is important to consider that cancer patients (especially with obesity) may require higher amounts of micronutrients due to their disease-related pathophysiological, inflammatory, and metabolic changes. Furthermore, the side effects of radiotherapy or chemotherapy (e.g., loss of appetite, aversion to certain foods, alterations in taste, diarrhea, or vomiting) can contribute to micronutrient deficiencies [157]. On the other hand, the presence of micronutrient deficiency can adversely impact disease prognosis, the effectiveness of cytoreductive therapy, and the risk of complications (such as depression, fatigue, delayed wound healing, and impaired immunocompetence) [157]. Indeed, there has been some evidence on the potential beneficial effects of certain vitamins (A, B, C, D, E, K) and other micronutrients (copper, zinc, selenium, and magnesium) on cancer prevention and treatment via their antioxidant, anti-inflammatory, and antiapoptotic properties [158].

Based on the above associations, individuals with obesity (with or without cancer) should be screened for micronutrient deficiencies [158]. In the case of individuals with obesity and cancer, micronutrient supplementation should be considered in clinical practice, focusing on improving the outcomes of their standard therapy as well as potentially minimizing the adverse effects of both cancer and anticancer treatment. Indeed, there are studies reporting the beneficial effects of the supplementation of certain micronutrients (mainly those with immuno-stabilizing and antioxidant properties, such as selenium, vitamin C, and vitamin D) in cancer patients, as mentioned above. Administration of micronutrients with little storage or reserve capacity (such as folic acid, vitamin C, vitamin B12) is also clinically important [158].

The AICR does not recommend the use of supplements for cancer protection [159]. The WCRF/AICR Third Expert Report on Diet, Nutrition, Physical Activity, and Cancer discusses the evidence on micronutrient intake and cancer risk [160]. For example, although there is strong evidence that consuming calcium supplements may help to protect against CRC, there is limited data that dairy products might increase prostate cancer risk; thus, no recommendations have been made [160]. Also, there is limited-suggestive evidence that multivitamin supplements and vitamin D may decrease the risk of colon cancer, whereas there is limited-inconclusive evidence that folic acid, vitamin A, vitamin B6, vitamin E, and selenium intake can reduce colon cancer risk [160]. Overall, this WCRF/AICR report does not recommend high-dose supplements for cancer prevention but rather suggests that all attempts should be made to obtain the needed nutrients through dietary sources before prescribing supplements [160].

Healthcare professionals must collaborate to identify individuals at risk of micronutrient deficiencies, manage imbalances, and promote evidence-based nutrition guidelines, emphasizing diverse diets and cautious supplementation [161]. According to the AICR, daily multivitamin/mineral supplement intake should be within the range of the recommended daily allowance (RDA) [157]. The RDA for several micronutrients can be found in ref [161]. Therefore, in the presence of micronutrient deficiencies that cannot be covered by diet (e.g., due to malnutrition or adverse events from the anticancer therapy), individuals with obesity and cancer may take a multivitamin/mineral supplement based on the RDA.

Certain recommendations for clinical practice could be made, especially for vitamins D and C, as well as selenium. For example, it is recommended to monitor vitamin D status in all cancer patients and treat any deficiencies by adequate vitamin D supplementation, i.e., 40–60 IU vitamin D/kg/day with a 25(OH)D target value of 100–150 nmol/L (40–60 ng/mL) [157]. Vitamin D deficiencies are particularly frequent in cancer patients with poor nutritional status, those treated with bisphosphonates, aromatase inhibitors, and chemotherapy regimens (e.g., taxanes, anthracyclines, and monoclonal antibodies), as well as in cases of muscular disorders, cancer-related anemia, cachexia, and fatigue.

The Physician Data Query (PDQ) cancer information summaries for health professionals can provide evidence-based data regarding the use of high-dose vitamin C in cancer patients [162]. In this context, intravenous ascorbate administration has been reported to rarely exert adverse events (e.g., hemolysis in patients with glucose-6-phosphate dehydrogenase deficiency(G6PD) and oxalate nephropathy). Thus, G6PD laboratory testing should always be assessed before a vitamin C infusion. Both parenteral or oral administration of vitamin C supplements could be useful in the presence of poor nutritional condition, fatigue, cachexia, or delayed postoperative wound healing [157]. However, the FDA has not approved the use of high-dose vitamin C as a treatment for cancer [162].

Regarding selenium, efforts should be made to achieve the optimal serum selenium level of 130–150 µg/L. However, patients with serum selenium concentrations of ≥122 µg/L should not be supplemented with selenium [157]. It should be noted that serum selenium may reduce the toxicity of chemotherapy and radiotherapy, and thus certain centers administer sodium selenite (i.e., the preferred selenium salt in oncology) as a pre-medication before chemotherapy (e.g., 1 mg sodium selenite in 100 mL 0.9% NaCl) [157].

There is limited evidence that l-carnitine may beneficially affect the cancer anorexia-cachexia syndrome. Oral supplementation of l-carnitine may be helpful in malnourished cancer patients, as well as in patients on chemotherapy regimens that induce carnitine deficiency (e.g., ifosfamide and cisplatin) or peripheral neuropathy (e.g., taxanes) [157].

According to the PDQ cancer information summaries, the administration of vitamin A, vitamin C, vitamin E, a multivitamin (containing folic acid, vitamin B6, and vitamin B12) or beta-carotene did not affect breast cancer incidence or progression [163]. Similarly, beta-carotene or vitamin E supplementation did not protect against lung cancer [164], whereas beta-carotene, vitamin E, vitamin C, and selenium intake did not affect the progression or regression of gastric cancer [165]. In contrast, a previous large randomized placebo-controlled trial (i.e., SELECT; n = 34,887 healthy men) of vitamin E (400 IU/d of all-rac-α-tocopheryl acetate) and selenium (200 μg/d from l-selenomethionine) supplementation not only failed to decrease prostate cancer incidence but also reported an increased risk of prostate cancer with vitamin E administration [166]. Obviously, there is still a need for further research in this field.

Interestingly, several studies reported an increased dietary supplement intake among cancer patients. For example, among 2,772 US cancer survivors and 31,310 individuals without cancer from the NHANES 2003–2016, cancer patients reported a higher prevalence of any dietary supplement use (70.4 vs. 51.2%), multivitamin/mineral supplement intake (48.9 vs. 36.6%), and supplement use of 11 individual vitamins and 8 minerals compared with individuals without cancer [167]. The most frequently used supplements for individual vitamins and minerals were vitamin C, D, E, and B12, as well as calcium. However, cancer patients reported lower amounts of nutrient intake from foods than individuals without cancer [167]. It should also be noted that nearly half of the cancer patients used dietary supplements without previously consulting their healthcare providers, thus highlighting the need for discussion on dietary supplement use between physicians and cancer patients in daily practice during clinical visits [167]. Similarly, another cross-sectional survey among 1,217 ambulatory cancer patients found that 47.2% of them reported using dietary supplements: calcium and magnesium supplementation was most often reported, followed by botanical and herbal supplements, multivitamins, vitamin D, and the vitamin B group [168]. Of note, 41.5% of the supplement users started the use after cancer diagnosis, whereas 37.1% of them had used the supplements regularly and 19.2% sporadically before cancer diagnosis [168]. A previous systematic review evaluating dietary supplement use among patients with cancer (n = 65 studies) reported that the majority of the studied supplements (for example, vitamins, omega-3 fatty acids, and botanicals) were found to be safe [169].

Based on all the above data, a complementary oncological treatment should include nutrition therapy and laboratory-validated supplementation of micronutrients. In this context, clinicians should discuss openly with individuals with obesity and cancer about nutritional supplementation since several patients may be already on supplements without the knowledge of their physicians [157]. Nutritional supplementation should be tailored and individualized based on each patient’s diet, genetics, cancer type, and treatment [157]. There is still a need for further evidence to establish global recommendations on micronutrient intake in individuals with obesity and cancer. Additionally, the clinical use of vitamins and minerals supplementation to prevent cancer development remains to be elucidated.

Recommendations

Priority should be given to obtaining micronutrients from food sources rather than supplements. Daily supplements, if needed, should stay within RDAs, and high-dose supplements are discouraged. Monitoring and treating vitamin D deficiency is advised, along with maintaining optimal selenium levels, particularly in cancer patients. Specific supplements like sodium selenite and l-carnitine may be considered in targeted cases, while high-dose vitamin C lacks FDA approval as a cancer treatment. During chemotherapy or radiotherapy, it is recommended to avoid antioxidant supplements in doses above tolerable limits.

The adoption of a healthy, balanced, diet rich in plant-based foods has been related to a reduced risk of developing various types of cancer [5]. In particular, a high intake of dietary fiber, fruits, vegetables, pulses, and whole grains, as well as a minimum consumption of alcohol and processed meat, with total restriction of sugar-sweetened drinks, are crucial for cancer prevention and treatment [6]. Although no clear nutritional indications are available in individuals with obesity and cancer, the MedDiet has been correlated with a decreased risk of several cancers [6, 170]. Furthermore, there is a growing interest in the use of IF or KD as an adjuvant therapy in cancer treatment, but current evidence is scarce; thus, no recommendations can be made [171].

On another issue, malnutrition, along with the side effects of radiotherapy or chemotherapy can lead to inadequate micronutrient status in cancer patients [172]. Furthermore, cancer patients (especially in the presence of obesity) may require higher amounts of micronutrients due to their disease-related inflammatory, pathophysiological, and metabolic changes [173]. On the other hand, micronutrient deficiencies may adversely affect cancer prognosis and the efficacy of cytoreductive therapy [172]. Having said that, there has been some evidence on the potential beneficial effects of certain vitamins (A, B, C, D, E, K) and other micronutrients (copper, zinc, selenium, magnesium) on cancer prevention and treatment via their antiapoptotic, anti-inflammatory, and antioxidant properties. Administration of micronutrients with little reserve or storage capacity (such as folic acid and vitamin B12) is also clinically significant.

Overall, individuals with obesity and cancer should be screened for micronutrient deficiencies. Although the AICR does not recommend supplement use for cancer protection, if micronutrient deficiencies exist and cannot be corrected by diet (e.g., due to adverse events from the anticancer therapy or malnutrition), individuals with obesity and cancer may be administered a multivitamin/mineral supplement based on the RDA [159]. Certain recommendations for the supplementation of vitamins D and C, as well as selenium have been made. In clinical practice, a complementary oncological team should include registered dieticians that can provide medical nutrition therapy and laboratory-validated supplementation of micronutrients.

Not applicable. As this is a review article, no patient data were collected.

The authors have no conflicts of interest to declare.

This study was not supported by any sponsor or funder.

G.M. conceived of the presented idea. L.B., S.B., M.E.G., N.K., L.T., and L.V. wrote the draft of the manuscript. G.M., A.C., L.B., V.Y., and M.H. reviewed the manuscript.

Not applicable. This review relies solely on publicly available data and does not include any original patient data.

1.
Zhang
X
,
Ha
S
,
Lau
HC
,
Yu
J
.
Excess body weight: novel insights into its roles in obesity comorbidities
.
Semin Cancer Biol
.
2023
;
92
:
16
27
.
2.
Iyengar
NM
,
Gucalp
A
,
Dannenberg
AJ
,
Hudis
CA
.
Obesity and cancer mechanisms: tumor microenvironment and inflammation
.
J Clin Oncol
.
2016
;
34
(
35
):
4270
6
.
3.
Miro
C
,
Docimo
A
,
Barrea
L
,
Verde
L
,
Cernea
S
,
Sojat
AS
, et al
.
Time for obesity-related cancer: the role of the circadian rhythm in cancer pathogenesis and treatment
.
Semin Cancer Biol
.
2023
;
91
:
99
109
.
4.
Otsuka
K
,
Nishiyama
H
,
Kuriki
D
,
Kawada
N
,
Ochiya
T
.
Connecting the dots in the associations between diet, obesity, cancer, and microRNAs
.
Semin Cancer Biol
.
2023
;
93
:
52
69
.
5.
Hardt
L
,
Mahamat-Saleh
Y
,
Aune
D
,
Schlesinger
S
.
Plant-based diets and cancer prognosis: a review of recent research
.
Curr Nutr Rep
.
2022
;
11
(
4
):
695
716
.
6.
Muscogiuri
G
,
Verde
L
,
Sulu
C
,
Katsiki
N
,
Hassapidou
M
,
Frias-Toral
E
, et al
.
Mediterranean diet and obesity-related disorders: what is the evidence
.
Curr Obes Rep
.
2022
;
11
(
4
):
287
304
.
7.
Muscogiuri
G
,
El Ghoch
M
,
Colao
A
,
Hassapidou
M
,
Yumuk
V
,
Busetto
L
, et al
.
European guidelines for obesity management in adults with a very low-calorie ketogenic diet: a systematic review and meta-analysis
.
Obes Facts
.
2021
;
14
(
2
):
222
45
.
8.
Barrea
L
,
Caprio
M
,
Tuccinardi
D
,
Moriconi
E
,
Di Renzo
L
,
Muscogiuri
G
, et al
.
Could ketogenic diet “starve” cancer? Emerging evidence
.
Crit Rev Food Sci Nutr
.
2022
;
62
(
7
):
1800
21
.
9.
Patikorn
C
,
Roubal
K
,
Veettil
SK
,
Chandran
V
,
Pham
T
,
Lee
YY
, et al
.
Intermittent fasting and obesity-related health outcomes: an umbrella review of meta-analyses of randomized clinical trials
.
JAMA Netw Open
.
2021
;
4
(
12
):
e2139558
.
10.
Clifton
KK
,
Ma
CX
,
Fontana
L
,
Peterson
LL
.
Intermittent fasting in the prevention and treatment of cancer
.
CA Cancer J Clin
.
2021
;
71
(
6
):
527
46
.
11.
Calle
EE
,
Rodriguez
C
,
Walker-Thurmond
K
,
Thun
MJ
.
Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults
.
N Engl J Med
.
2003
;
348
(
17
):
1625
38
.
12.
Renehan
AG
,
Tyson
M
,
Egger
M
,
Heller
RF
,
Zwahlen
M
.
Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies
.
Lancet
.
2008
;
371
(
9612
):
569
78
.
13.
O’Sullivan
J
,
Lysaght
J
,
Donohoe
CL
,
Reynolds
JV
.
Obesity and gastrointestinal cancer: the interrelationship of adipose and tumour microenvironments
.
Nat Rev Gastroenterol Hepatol
.
2018
;
15
(
11
):
699
714
.
14.
Trevellin
E
,
Bettini
S
,
Pilatone
A
,
Vettor
R
,
Milan
G
.
Obesity, the adipose organ and cancer in humans: association or causation
.
Biomedicines
.
2023
;
11
(
5
):
1319
.
15.
American Institute for cancer research (AICR) - the continuous update Project (CUP)
. Available from: https://wwwaicrorg/research/the-continuous-update-project (accessed on 24th May 2024).
16.
Neuhouser
ML
,
Aragaki
AK
,
Prentice
RL
,
Manson
JE
,
Chlebowski
R
,
Carty
CL
, et al
.
Overweight, obesity, and postmenopausal invasive breast cancer risk: a secondary analysis of the women’s health initiative randomized clinical trials
.
JAMA Oncol
.
2015
;
1
(
5
):
611
21
.
17.
Reeves
GK
,
Pirie
K
,
Beral
V
,
Green
J
,
Spencer
E
,
Bull
D
, et al
.
Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study
.
BMJ
.
2007
;
335
(
7630
):
1134
.
18.
Borgo
C
,
Ruzzene
M
.
Protein kinase CK2 inhibition as a pharmacological strategy
.
Adv Protein Chem Struct Biol
.
2021
;
124
:
23
46
.
19.
Ma
H
,
Ursin
G
,
Xu
X
,
Lee
E
,
Togawa
K
,
Malone
KE
, et al
.
Body mass index at age 18 years and recent body mass index in relation to risk of breast cancer overall and ER/PR/HER2-defined subtypes in white women and African-American women: a pooled analysis
.
Breast Cancer Res
.
2018
;
20
(
1
):
5
.
20.
Munsell
MF
,
Sprague
BL
,
Berry
DA
,
Chisholm
G
,
Trentham-Dietz
A
.
Body mass index and breast cancer risk according to postmenopausal estrogen-progestin use and hormone receptor status
.
Epidemiol Rev
.
2014
;
36
(
1
):
114
36
.
21.
Picon-Ruiz
M
,
Morata-Tarifa
C
,
Valle-Goffin
JJ
,
Friedman
ER
,
Slingerland
JM
.
Obesity and adverse breast cancer risk and outcome: mechanistic insights and strategies for intervention
.
CA Cancer J Clin
.
2017
;
67
(
5
):
378
97
.
22.
World Cancer Research Fund/American Institute for Cancer Research: Continuous Update Project Report Food Nutr Phys Activity, Prev Endometrial Cancer
. Available from: https://wwwwcrforg/diet-activity-and-cancer/cancer-types/endometrial-cancer/ (accessed on 24th May, 2024).
23.
von Gruenigen
VE
,
Tian
C
,
Frasure
H
,
Waggoner
S
,
Keys
H
,
Barakat
RR
.
Treatment effects, disease recurrence, and survival in obese women with early endometrial carcinoma: a Gynecologic Oncology Group study
.
Cancer
.
2006
;
107
(
12
):
2786
91
.
24.
Lee
K
,
Kruper
L
,
Dieli-Conwright
CM
,
Mortimer
JE
.
The impact of obesity on breast cancer diagnosis and treatment
.
Curr Oncol Rep
.
2019
;
21
(
5
):
41
.
25.
World cancer research Fund/American Institute for cancer research: continuous update Project report: food, nutrition, physical activity, and the prevention of ovarian cancer
. Available from: https://wwwwcrforg/diet-activity-and-cancer/cancer-types/ovarian-cancer/ (accessed on 24th Jan, 2024).
26.
Delort
L
,
Kwiatkowski
F
,
Chalabi
N
,
Satih
S
,
Bignon
YJ
,
Bernard-Gallon
DJ
.
Central adiposity as a major risk factor of ovarian cancer
.
Anticancer Res
.
2009
;
29
(
12
):
5229
34
.
27.
Beddy
P
,
Howard
J
,
McMahon
C
,
Knox
M
,
de Blacam
C
,
Ravi
N
, et al
.
Association of visceral adiposity with oesophageal and junctional adenocarcinomas
.
Br J Surg
.
2010
;
97
(
7
):
1028
34
.
28.
Rinella
ME
,
Lazarus
JV
,
Ratziu
V
,
Francque
SM
,
Sanyal
AJ
,
Kanwal
F
, et al
.
A multisociety Delphi consensus statement on new fatty liver disease nomenclature
.
Hepatology
.
2023
;
78
(
6
):
1966
86
.
29.
Adams
KF
,
Leitzmann
MF
,
Albanes
D
,
Kipnis
V
,
Moore
SC
,
Schatzkin
A
, et al
.
Body size and renal cell cancer incidence in a large US cohort study
.
Am J Epidemiol
.
2008
;
168
(
3
):
268
77
.
30.
Haggstrom
C
,
Rapp
K
,
Stocks
T
,
Manjer
J
,
Bjorge
T
,
Ulmer
H
, et al
.
Metabolic factors associated with risk of renal cell carcinoma
.
PLoS One
.
2013
;
8
(
2
):
e57475
.
31.
Larsson
SC
,
Wolk
A
.
Obesity and colon and rectal cancer risk: a meta-analysis of prospective studies
.
Am J Clin Nutr
.
2007
;
86
(
3
):
556
65
.
32.
Moghaddam
AA
,
Woodward
M
,
Huxley
R
.
Obesity and risk of colorectal cancer: a meta-analysis of 31 studies with 70,000 events
.
Cancer Epidemiol Biomarkers Prev
.
2007
;
16
(
12
):
2533
47
.
33.
Li
Y
,
Li
C
,
Wu
G
,
Yang
W
,
Wang
X
,
Duan
L
, et al
.
The obesity paradox in patients with colorectal cancer: a systematic review and meta-analysis
.
Nutr Rev
.
2022
;
80
(
7
):
1755
68
.
34.
Camilleri
M
,
Malhi
H
,
Acosta
A
.
Gastrointestinal complications of obesity
.
Gastroenterology
.
2017
;
152
(
7
):
1656
70
.
35.
Larsson
SC
,
Orsini
N
,
Wolk
A
.
Body mass index and pancreatic cancer risk: a meta-analysis of prospective studies
.
Int J Cancer
.
2007
;
120
(
9
):
1993
8
.
36.
Bonn
SE
,
Sjolander
A
,
Tillander
A
,
Wiklund
F
,
Gronberg
H
,
Balter
K
.
Body mass index in relation to serum prostate-specific antigen levels and prostate cancer risk
.
Int J Cancer
.
2016
;
139
(
1
):
50
7
.
37.
Harrison
S
,
Tilling
K
,
Turner
EL
,
Martin
RM
,
Lennon
R
,
Lane
JA
, et al
.
Systematic review and meta-analysis of the associations between body mass index, prostate cancer, advanced prostate cancer, and prostate-specific antigen
.
Cancer Causes Control
.
2020
;
31
(
5
):
431
49
.
38.
Marzullo
P
,
Bettini
S
,
Menafra
D
,
Aprano
S
,
Muscogiuri
G
,
Barrea
L
, et al
.
Spot-light on microbiota in obesity and cancer
.
Int J Obes
.
2021
;
45
(
11
):
2291
9
.
39.
Ghaben
AL
,
Scherer
PE
.
Adipogenesis and metabolic health
.
Nat Rev Mol Cell Biol
.
2019
;
20
(
4
):
242
58
.
40.
Calejman
CM
,
Doxsey
WG
,
Fazakerley
DJ
,
Guertin
DA
.
Integrating adipocyte insulin signaling and metabolism in the multi-omics era
.
Trends Biochem Sci
.
2022
;
47
(
6
):
531
46
.
41.
Milan
G
,
Conci
S
,
Sanna
M
,
Favaretto
F
,
Bettini
S
,
Vettor
R
.
ASCs and their role in obesity and metabolic diseases
.
Trends Endocrinol Metab
.
2021
;
32
(
12
):
994
1006
.
42.
Belligoli
A
,
Compagnin
C
,
Sanna
M
,
Favaretto
F
,
Fabris
R
,
Busetto
L
, et al
.
Characterization of subcutaneous and omental adipose tissue in patients with obesity and with different degrees of glucose impairment
.
Sci Rep
.
2019
;
9
(
1
):
11333
.
43.
Semenza
GL
.
Hypoxia-inducible factors: mediators of cancer progression and targets for cancer therapy
.
Trends Pharmacol Sci
.
2012
;
33
(
4
):
207
14
.
44.
Stienstra
R
,
Joosten
LA
,
Koenen
T
,
van Tits
B
,
van Diepen
JA
,
van den Berg
SA
, et al
.
The inflammasome-mediated caspase-1 activation controls adipocyte differentiation and insulin sensitivity
.
Cell Metab
.
2010
;
12
(
6
):
593
605
.
45.
Keshavarz Shahbaz
S
,
Koushki
K
,
Ayati
SH
,
Bland
AR
,
Bezsonov
EE
,
Sahebkar
A
.
Inflammasomes and colorectal cancer
.
Cells
.
2021
;
10
(
9
):
2172
.
46.
Bunnell
BA
,
Martin
EC
,
Matossian
MD
,
Brock
CK
,
Nguyen
K
,
Collins-Burow
B
, et al
.
The effect of obesity on adipose-derived stromal cells and adipose tissue and their impact on cancer
.
Cancer Metastasis Rev
.
2022
;
41
(
3
):
549
73
.
47.
Zhang
Z
,
Scherer
PE
.
Adipose tissue: the dysfunctional adipocyte: a cancer cell’s best friend
.
Nat Rev Endocrinol
.
2018
;
14
(
3
):
132
4
.
48.
Mutoh
M
,
Teraoka
N
,
Takasu
S
,
Takahashi
M
,
Onuma
K
,
Yamamoto
M
, et al
.
Loss of adiponectin promotes intestinal carcinogenesis in Min and wild-type mice
.
Gastroenterology
.
2011
;
140
(
7
):
2000
8.e20082
.
49.
Fazolini
NP
,
Cruz
AL
,
Werneck
MB
,
Viola
JP
,
Maya-Monteiro
CM
,
Bozza
PT
.
Leptin activation of mTOR pathway in intestinal epithelial cell triggers lipid droplet formation, cytokine production and increased cell proliferation
.
Cell Cycle
.
2015
;
14
(
16
):
2667
76
.
50.
Rose
DP
,
Gracheck
PJ
,
Vona-Davis
L
.
The interactions of obesity, inflammation and insulin resistance in breast cancer
.
Cancers
.
2015
;
7
(
4
):
2147
68
.
51.
Subbaramaiah
K
,
Howe
LR
,
Bhardwaj
P
,
Du
B
,
Gravaghi
C
,
Yantiss
RK
, et al
.
Obesity is associated with inflammation and elevated aromatase expression in the mouse mammary gland
.
Cancer Prev Res
.
2011
;
4
(
3
):
329
46
.
52.
Zahid
H
,
Simpson
ER
,
Brown
KA
.
Inflammation, dysregulated metabolism and aromatase in obesity and breast cancer
.
Curr Opin Pharmacol
.
2016
;
31
:
90
6
.
53.
Ando
S
,
Catalano
S
.
The multifactorial role of leptin in driving the breast cancer microenvironment
.
Nat Rev Endocrinol
.
2011
;
8
(
5
):
263
75
.
54.
Maguire
OA
,
Ackerman
SE
,
Szwed
SK
,
Maganti
AV
,
Marchildon
F
,
Huang
X
, et al
.
Creatine-mediated crosstalk between adipocytes and cancer cells regulates obesity-driven breast cancer
.
Cell Metab
.
2021
;
33
(
3
):
499
512 e6
.
55.
Kazak
L
,
Cohen
P
.
Creatine metabolism: energy homeostasis, immunity and cancer biology
.
Nat Rev Endocrinol
.
2020
;
16
(
8
):
421
36
.
56.
Kurmi
K
,
Hitosugi
S
,
Yu
J
,
Boakye-Agyeman
F
,
Wiese
EK
,
Larson
TR
, et al
.
Tyrosine phosphorylation of mitochondrial creatine kinase 1 enhances a druggable tumor energy shuttle pathway
.
Cell Metab
.
2018
;
28
(
6
):
833
47.e8
.
57.
Borgo
C
,
Milan
G
,
Favaretto
F
,
Stasi
F
,
Fabris
R
,
Salizzato
V
, et al
.
CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity
.
Sci Rep
.
2017
;
7
(
1
):
17569
.
58.
Husain
K
,
Williamson
TT
,
Nelson
N
,
Ghansah
T
.
Protein kinase 2 (CK2): a potential regulator of immune cell development and function in cancer
.
Immunol Med
.
2021
;
44
(
3
):
159
74
.
59.
Guerra
B
,
Issinger
OG
.
Role of protein kinase CK2 in aberrant lipid metabolism in cancer
.
Pharmaceuticals
.
2020
;
13
(
10
):
292
.
60.
Lu
Y
,
Fan
C
,
Li
P
,
Lu
Y
,
Chang
X
,
Qi
K
.
Short chain fatty acids prevent high-fat-diet-induced obesity in mice by regulating G protein-coupled receptors and gut microbiota
.
Sci Rep
.
2016
;
6
:
37589
.
61.
Singh
N
,
Gurav
A
,
Sivaprakasam
S
,
Brady
E
,
Padia
R
,
Shi
H
, et al
.
Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis
.
Immunity
.
2014
;
40
(
1
):
128
39
.
62.
Cani
PD
,
Jordan
BF
.
Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer
.
Nat Rev Gastroenterol Hepatol
.
2018
;
15
(
11
):
671
82
.
63.
Pradere
JP
,
Dapito
DH
,
Schwabe
RF
.
The yin and yang of toll-like receptors in cancer
.
Oncogene
.
2014
;
33
(
27
):
3485
95
.
64.
World Health Organization
.
Healthy diet
.
2020
. [updated April, 20 2020. Available from: https://www.who.int/news-room/fact-sheets/detail/healthy-diet
65.
Hassapidou
M
,
Vlassopoulos
A
,
Kalliostra
M
,
Govers
E
,
Mulrooney
H
,
Ells
L
, et al
.
European association for the study of obesity position statement on medical nutrition therapy for the management of overweight and obesity in adults developed in collaboration with the European federation of the associations of dietitians
.
Obes Facts
.
2023
;
16
(
1
):
11
28
.
66.
Morgan-Bathke
M
,
Raynor
HA
,
Baxter
SD
,
Halliday
TM
,
Lynch
A
,
Malik
N
, et al
.
Medical nutrition therapy interventions provided by dietitians for adult overweight and obesity management: an academy of nutrition and dietetics evidence-based practice guideline
.
J Acad Nutr Diet
.
2023
;
123
(
3
):
520
45.e10
.
67.
World Health Organization
.
Cancer, prevention
.
Geneva
:
World Health Organization
. Available from: https://www.who.int/health-topics/cancer#tab=tab_2
68.
World Cancer Recearch Fund International (WCRF), American Institute for Cancer Research
.
Continous Update Project. Recommendations and public health and policy impications
.
London
:
World Cancer Recearch Fund International (WCRF)
;
2018
.
69.
Rosell
M
,
Fadnes
LT
.
Vegetables, fruits, and berries: a scoping review for Nordic Nutrition Recommendations 2023
.
Food Nutr Res
.
2024
;
68
.
70.
Reynolds
A
,
Mann
J
,
Cummings
J
,
Winter
N
,
Mete
E
,
Te Morenga
L
.
Carbohydrate quality and human health: a series of systematic reviews and meta-analyses
.
Lancet
.
2019
;
393
(
10170
):
434
45
.
71.
McGlynn
ND
,
Khan
TA
,
Wang
L
,
Zhang
R
,
Chiavaroli
L
,
Au-Yeung
F
, et al
.
Association of low- and No-calorie sweetened beverages as a replacement for sugar-sweetened beverages with body weight and cardiometabolic risk: a systematic review and meta-analysis
.
JAMA Netw Open
.
2022
;
5
(
3
):
e222092
.
72.
World Health Organization
.
Use of non-sugar sweeteners: WHO guideline
.
Geneva
:
World Health Organization
;
2023
.
73.
World Health Organization
.
Summary of findings of the evaluation of aspartame at the international agency for research on cancer (IARC) monographs programme’s 134th meeting, and the joint FAO/WHO Expert committee on food additives (JECFA) 96th meeting
.
Geneva
:
World Health Organization
;
2023
.
74.
EFSA Panel on Food Additives and Nutrient Sources added to Food ANS
.
Scientific opinion on the re-evaluation of aspartame (E951) as a food additive
.
EFSA J
.
2013
;
11
(
12
):
3496
.
75.
Rishor-Olney
CR
,
Hinson
MR
.
StatPearls [internet]. Treasure island (FL)
.
StatPearls Publishing
;
2024
.
76.
Tosti
V
,
Bertozzi
B
,
Fontana
L
.
Health benefits of the Mediterranean diet: metabolic and molecular mechanisms
.
J Gerontol
.
2018
;
73
(
3
):
318
26
.
77.
Mentella
MC
,
Scaldaferri
F
,
Ricci
C
,
Gasbarrini
A
,
Miggiano
G
.
Cancer and mediterranean diet: a review
.
Nutrients
.
2019
;
11
(
9
):
2059
.
78.
Morze
J
,
Danielewicz
A
,
Przybyłowicz
K
,
Zeng
H
,
Hoffmann
G
,
Schwingshackl
L
.
An updated systematic review and meta-analysis on adherence to mediterranean diet and risk of cancer
.
Eur J Nutr
.
2021
;
60
(
3
):
1561
86
.
79.
Barak
Y
,
Fridman
D
.
Impact of mediterranean diet on cancer: focused literature review
.
Cancer Genomics Proteomics
.
2017
;
14
(
6
):
403
8
.
80.
Bae
KR
,
Cho
J
.
Changes after cancer diagnosis and return to work: experience of Korean cancer patients
.
BMC Cancer
.
2021
;
21
(
1
):
86
.
81.
Arends
J
,
Bachmann
P
,
Baracos
V
,
Barthelemy
N
,
Bertz
H
,
Bozzetti
F
, et al
.
ESPEN guidelines on nutrition in cancer patients
.
Clin Nutr
.
2017
;
36
(
1
):
11
48
.
82.
Wen
H
,
Deng
G
,
Shi
X
,
Liu
Z
,
Lin
A
,
Cheng
Q
, et al
.
Body mass index, weight change, and cancer prognosis: a meta-analysis and systematic review of 73 cohort studies
.
ESMO Open
.
2024
;
9
(
3
):
102241
.
83.
Haseen
F
,
Murray
LJ
,
Cardwell
CR
,
O'Sullivan
JM
,
Cantwell
MM
.
The effect of androgen deprivation therapy on body composition in men with prostate cancer: systematic review and meta-analysis
.
J Cancer Surviv
.
2010
;
4
(
2
):
128
39
.
84.
Zullig
LL
,
Sung
AD
,
Khouri
MG
,
Jazowski
S
,
Shah
NP
,
Sitlinger
A
, et al
.
Cardiometabolic comorbidities in cancer survivors: jacc: CardioOncology state-of-the-art review
.
JACC CardioOncol
.
2022
;
4
(
2
):
149
65
.
85.
Guha
A
,
Gong
Y
,
DeRemer
D
,
Owusu-Guha
J
,
Dent
SF
,
Cheng
RK
, et al
.
Cardiometabolic consequences of targeted anticancer therapies
.
J Cardiovasc Pharmacol
.
2022
;
80
(
4
):
515
21
.
86.
Du
Y
,
Carranza
Z
,
Luan
Y
,
Busman-Sahay
K
,
Wolf
S
,
Campbell
SP
, et al
.
Evidence of cancer therapy-induced chronic inflammation in the ovary across multiple species: a potential cause of persistent tissue damage and follicle depletion
.
J Reprod Immunol
.
2022
;
150
:
103491
.
87.
LeVee
A
,
Mortimer
J
.
The challenges of treating patients with breast cancer and obesity
.
Cancers
.
2023
;
15
(
9
):
2526
.
88.
Ligibel
JA
,
Bohlke
K
,
May
AM
,
Clinton
SK
,
Demark-Wahnefried
W
,
Gilchrist
SC
, et al
.
Exercise, diet, and weight management during cancer treatment: ASCO guideline
.
J Clin Oncol
.
2022
;
40
(
22
):
2491
507
.
89.
Villarini
A
,
Pasanisi
P
,
Raimondi
M
,
Gargano
G
,
Bruno
E
,
Morelli
D
, et al
.
Preventing weight gain during adjuvant chemotherapy for breast cancer: a dietary intervention study
.
Breast Cancer Res Treat
.
2012
;
135
(
2
):
581
9
.
90.
Rubino
R
,
Iuliucci
MR
,
Gatani
S
,
Piscosquito
A
,
D’Ambrosio
B
,
Ingenito
C
, et al
.
Mediterranean diet as a supportive intervention in cancer patients: current evidence and future directions
.
Curr Oncol
.
2022
;
29
(
10
):
7579
82
.
91.
Baguley
BJ
,
Skinner
TL
,
Jenkins
DG
,
Wright
ORL
.
Mediterranean-style dietary pattern improves cancer-related fatigue and quality of life in men with prostate cancer treated with androgen deprivation therapy: a pilot randomised control trial
.
Clin Nutr
.
2021
;
40
(
1
):
245
54
.
92.
Braakhuis
A
,
Campion
P
,
Bishop
K
.
The effects of dietary nutrition education on weight and health biomarkers in breast cancer survivors
.
Med Sci
.
2017
;
5
(
2
):
12
.
93.
Kleckner
AS
,
Reschke
JE
,
Kleckner
IR
,
Magnuson
A
,
Amitrano
AM
,
Culakova
E
, et al
.
The effects of a mediterranean diet intervention on cancer-related fatigue for patients undergoing chemotherapy: a pilot randomized controlled trial
.
Cancers
.
2022
;
14
(
17
):
4202
.
94.
Long Parma
DA
,
Reynolds
GL
,
Munoz
E
,
Ramirez
AG
.
Effect of an antiinflammatory dietary intervention on quality of life among breast cancer survivors
.
Support Care Cancer
.
2022
;
30
(
7
):
5903
10
.
95.
Papandreou
P
,
Gioxari
A
,
Nimee
F
,
Skouroliakou
M
.
Application of clinical decision support system to assist breast cancer patients with lifestyle modifications during the COVID-19 pandemic: a randomised controlled trial
.
Nutrients
.
2021
;
13
(
6
):
2115
.
96.
Ruiz-Vozmediano
J
,
Löhnchen
S
,
Jurado
L
,
Recio
R
,
Rodríguez-Carrillo
A
,
López
M
, et al
.
Influence of a multidisciplinary program of diet, exercise, and mindfulness on the quality of life of stage iia-iib breast cancer survivors
.
Integr Cancer Ther
.
2020
;
19
:
1534735420924757
11
.
97.
Skouroliakou
M
,
Grosomanidis
D
,
Massara
P
,
Kostara
C
,
Papandreou
P
,
Ntountaniotis
D
, et al
.
Serum antioxidant capacity, biochemical profile and body composition of breast cancer survivors in a randomized Mediterranean dietary intervention study
.
Eur J Nutr
.
2018
;
57
(
6
):
2133
45
.
98.
Gioxari
A
,
Tzanos
D
,
Kostara
C
,
Papandreou
P
,
Mountzios
G
,
Skouroliakou
M
.
Mediterranean diet implementation to protect against advanced lung cancer index (ALI) rise: study design and preliminary results of a randomised controlled trial
.
Int J Environ Res Public Health
.
2021
;
18
(
7
):
3700
.
99.
Bagheri
A
,
Asoudeh
F
,
Rezaei
S
,
Babaei
M
,
Esmaillzadeh
A
.
The effect of mediterranean diet on body composition, inflammatory factors, and nutritional status in patients with cachexia induced by colorectal cancer: a randomized clinical trial
.
Integr Cancer Ther
.
2023
;
22
:
15347354231195322
.
100.
Harvie
M
,
Pegington
M
,
McMullan
D
,
Bundred
N
,
Livingstone
K
,
Campbell
A
, et al
.
The effectiveness of home versus community-based weight control programmes initiated soon after breast cancer diagnosis: a randomised controlled trial
.
Br J Cancer
.
2019
;
121
(
6
):
443
54
.
101.
Alimperti
A
,
Alikari
V
,
Tsironi
M
,
Rojas Gil
AP
,
Papageorgiou
D
,
Kolovos
P
, et al
.
Lipid disturbances in breast cancer patients during chemotherapy
.
Nurs Rep
.
2023
;
13
(
4
):
1500
10
.
102.
Bolte
LA
,
Lee
KA
,
Björk
JR
,
Leeming
ER
,
Campmans-Kuijpers
MJE
,
de Haan
JJ
, et al
.
Association of a mediterranean diet with outcomes for patients treated with immune checkpoint blockade for advanced melanoma
.
JAMA Oncol
.
2023
;
9
(
5
):
705
9
.
103.
Sun
Q
,
Hong
Z
,
Zhang
C
,
Wang
L
,
Han
Z
,
Ma
D
.
Immune checkpoint therapy for solid tumours: clinical dilemmas and future trends
.
Signal Transduct Target Ther
.
2023
;
8
(
1
):
320
.
104.
Mittelman
SD
.
The role of diet in cancer prevention and chemotherapy efficacy
.
Annu Rev Nutr
.
2020
;
40
:
273
97
.
105.
Boccellino
M
,
D’Angelo
S
.
Anti-obesity effects of polyphenol intake: current status and future possibilities
.
Int J Mol Sci
.
21
(
16
):
5642
.
106.
Kim
JH
,
Kim
OK
,
Yoon
HG
,
Park
J
,
You
Y
,
Kim
K
, et al
.
Anti-obesity effect of extract from fermented Curcuma longa L. through regulation of adipogenesis and lipolysis pathway in high-fat diet-induced obese rats
.
Food Nutr Res
.
2016
;
60
:
30428
.
107.
Mohammadi
A
,
Sahebkar
A
,
Iranshahi
M
,
Amini
M
,
Khojasteh
R
,
Ghayour-Mobarhan
M
, et al
.
Effects of supplementation with curcuminoids on dyslipidemia in obese patients: a randomized crossover trial
.
Phytother Res
.
2013
;
27
(
3
):
374
9
.
108.
Zhou
Y
,
Zheng
J
,
Li
Y
,
Xu
DP
,
Li
S
,
Chen
YM
, et al
.
Natural polyphenols for prevention and treatment of cancer
.
Nutrients
.
2016
;
8
(
8
):
515
.
109.
Cruz-Correa
M
,
Shoskes
DA
,
Sanchez
P
,
Zhao
R
,
Hylind
LM
,
Wexner
SD
, et al
.
Combination treatment with curcumin and quercetin of adenomas in familial adenomatous polyposis
.
Clin Gastroenterol Hepatol
.
2006
;
4
(
8
):
1035
8
.
110.
Zhang
G
,
Wang
Y
,
Zhang
Y
,
Wan
X
,
Li
J
,
Liu
K
, et al
.
Anti-cancer activities of tea epigallocatechin-3-gallate in breast cancer patients under radiotherapy
.
Curr Mol Med
.
2012
;
12
(
2
):
163
76
.
111.
Maleki Dana
P
,
Sadoughi
F
,
Asemi
Z
,
Yousefi
B
.
The role of polyphenols in overcoming cancer drug resistance: a comprehensive review
.
Cell Mol Biol Lett
.
2022
;
27
(
1
):
1
.
112.
Hoensch
H
,
Groh
B
,
Edler
L
,
Kirch
W
.
Prospective cohort comparison of flavonoid treatment in patients with resected colorectal cancer to prevent recurrence
.
World J Gastroenterol
.
2008
;
14
(
14
):
2187
93
.
113.
Cronin
P
,
Joyce
SA
,
O’Toole
PW
,
O’Connor
EM
.
Dietary fibre modulates the gut microbiota
.
Nutrients
.
2021
;
13
(
5
):
1655
.
114.
Tan
J
,
McKenzie
C
,
Potamitis
M
,
Thorburn
AN
,
Mackay
CR
,
Macia
L
.
The role of short-chain fatty acids in health and disease
.
Adv Immunol
.
2014
;
121
:
91
119
.
115.
Thorburn
AN
,
Macia
L
,
Mackay
CR
.
Diet, metabolites, and “western-lifestyle” inflammatory diseases
.
Immunity
.
2014
;
40
(
6
):
833
42
.
116.
Anachad
O
,
Taouil
A
,
Taha
W
,
Bennis
F
,
Chegdani
F
.
The implication of short-chain fatty acids in obesity and diabetes
.
Microbiol Insights
.
2023
;
16
:
11786361231162720
.
117.
Barrea
L
,
Muscogiuri
G
,
Annunziata
G
,
Laudisio
D
,
Pugliese
G
,
Salzano
C
, et al
.
From gut microbiota dysfunction to obesity: could short-chain fatty acids stop this dangerous course
.
Hormones
.
2019
;
18
(
3
):
245
50
.
118.
Son
MY
,
Cho
HS
.
Anticancer effects of gut microbiota-derived short-chain fatty acids in cancers
.
J Microbiol Biotechnol
.
2023
;
33
(
7
):
849
56
.
119.
Liu
Q
.
Triglyceride-lowering and anti-inflammatory mechanisms of omega-3 polyunsaturated fatty acids for atherosclerotic cardiovascular risk reduction
.
J Clin Lipidol
.
2021
;
15
(
4
):
556
68
.
120.
Albracht-Schulte
K
,
Kalupahana
NS
,
Ramalingam
L
,
Wang
S
,
Rahman
SM
,
Robert-McComb
J
, et al
.
Omega-3 fatty acids in obesity and metabolic syndrome: a mechanistic update
.
J Nutr Biochem
.
2018
;
58
:
1
16
.
121.
Cockbain
AJ
,
Toogood
GJ
,
Hull
MA
.
Omega-3 polyunsaturated fatty acids for the treatment and prevention of colorectal cancer
.
Gut
.
2012
;
61
(
1
):
135
49
.
122.
Theinel
MH
,
Nucci
MP
,
Alves
AH
,
Dias
OFM
,
Mamani
JB
,
Garrigós
MM
, et al
.
The effects of omega-3 polyunsaturated fatty acids on breast cancer as a preventive measure or as an adjunct to conventional treatments
.
Nutrients
.
2023
;
15
(
6
):
1310
.
123.
Jing
K
,
Wu
T
,
Lim
K
.
Omega-3 polyunsaturated fatty acids and cancer
.
Anticancer Agents Med Chem
.
2013
;
13
(
8
):
1162
77
.
124.
Patterson
RE
,
Flatt
SW
,
Newman
VA
,
Natarajan
L
,
Rock
CL
,
Thomson
CA
, et al
.
Marine fatty acid intake is associated with breast cancer prognosis
.
J Nutr
.
2011
;
141
(
2
):
201
6
.
125.
Camajani
E
,
Feraco
A
,
Verde
L
,
Moriconi
E
,
Marchetti
M
,
Colao
A
, et al
.
Ketogenic diet as a possible non-pharmacological therapy in main endocrine diseases of the female reproductive system: a practical guide for nutritionists
.
Curr Obes Rep
.
2023
;
12
(
3
):
231
49
.
126.
Weber
DD
,
Aminzadeh-Gohari
S
,
Tulipan
J
,
Catalano
L
,
Feichtinger
RG
,
Kofler
B
.
Ketogenic diet in the treatment of cancer: where do we stand
.
Mol Metab
.
2020
;
33
:
102
21
.
127.
Allen
BG
,
Bhatia
SK
,
Anderson
CM
,
Eichenberger-Gilmore
JM
,
Sibenaller
ZA
,
Mapuskar
KA
, et al
.
Ketogenic diets as an adjuvant cancer therapy: history and potential mechanism
.
Redox Biol
.
2014
;
2
:
963
70
.
128.
Aminzadeh-Gohari
S
,
Feichtinger
RG
,
Vidali
S
,
Locker
F
,
Rutherford
T
,
O'Donnel
M
, et al
.
A ketogenic diet supplemented with medium-chain triglycerides enhances the anti-tumor and anti-angiogenic efficacy of chemotherapy on neuroblastoma xenografts in a CD1-nu mouse model
.
Oncotarget
.
2017
;
8
(
39
):
64728
44
.
129.
Zhang
J
,
Jia
PP
,
Liu
QL
,
Cong
MH
,
Gao
Y
,
Shi
HP
, et al
.
Low ketolytic enzyme levels in tumors predict ketogenic diet responses in cancer cell lines in vitro and in vivo
.
J Lipid Res
.
2018
;
59
(
4
):
625
34
.
130.
Pflanz
NC
,
Daszkowski
AW
,
James
KA
,
Mihic
SJ
.
Ketone body modulation of ligand-gated ion channels
.
Neuropharmacology
.
2019
;
148
:
21
30
.
131.
Trotta
MC
,
Maisto
R
,
Guida
F
,
Boccella
S
,
Luongo
L
,
Balta
C
, et al
.
The activation of retinal HCA2 receptors by systemic beta-hydroxybutyrate inhibits diabetic retinal damage through reduction of endoplasmic reticulum stress and the NLRP3 inflammasome
.
PLoS One
.
2019
;
14
(
1
):
e0211005
.
132.
Nakamura
K
,
Tonouchi
H
,
Sasayama
A
,
Ashida
K
.
A ketogenic formula prevents tumor progression and cancer cachexia by attenuating systemic inflammation in colon 26 tumor-bearing mice
.
Nutrients
.
2018
;
10
(
2
):
206
.
133.
Youm
YH
,
Nguyen
KY
,
Grant
RW
,
Goldberg
EL
,
Bodogai
M
,
Kim
D
, et al
.
The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease
.
Nat Med
.
2015
;
21
(
3
):
263
9
.
134.
Klement
RJ
,
Brehm
N
,
Sweeney
RA
.
Ketogenic diets in medical oncology: a systematic review with focus on clinical outcomes
.
Med Oncol
.
2020
;
37
(
2
):
14
.
135.
Champ
CE
,
Palmer
JD
,
Volek
JS
,
Werner-Wasik
M
,
Andrews
DW
,
Evans
JJ
, et al
.
Targeting metabolism with a ketogenic diet during the treatment of glioblastoma multiforme
.
J Neuro Oncol
.
2014
;
117
(
1
):
125
31
.
136.
Cohen
CW
,
Fontaine
KR
,
Arend
RC
,
Alvarez
RD
,
Leath
CA
III
,
Huh
WK
, et al
.
A ketogenic diet reduces central obesity and serum insulin in women with ovarian or endometrial cancer
.
J Nutr
.
2018
;
148
(
8
):
1253
60
.
137.
Cohen
CW
,
Fontaine
KR
,
Arend
RC
,
Soleymani
T
,
Gower
BA
.
Favorable effects of a ketogenic diet on physical function, perceived energy, and food cravings in women with ovarian or endometrial cancer: a randomized, controlled trial
.
Nutrients
.
2018
;
10
(
9
):
1187
.
138.
Klement
RJ
,
Champ
CE
,
Kammerer
U
,
Koebrunner
PS
,
Krage
K
,
Schafer
G
, et al
.
Impact of a ketogenic diet intervention during radiotherapy on body composition: III-final results of the KETOCOMP study for breast cancer patients
.
Breast Cancer Res
.
2020
;
22
(
1
):
94
.
139.
Liskiewicz
AD
,
Kasprowska
D
,
Wojakowska
A
,
Polanski
K
,
Lewin-Kowalik
J
,
Kotulska
K
, et al
.
Long-term high fat ketogenic diet promotes renal tumor growth in a rat model of tuberous sclerosis
.
Sci Rep
.
2016
;
6
:
21807
.
140.
Xia
S
,
Lin
R
,
Jin
L
,
Zhao
L
,
Kang
HB
,
Pan
Y
, et al
.
Prevention of dietary-fat-fueled ketogenesis attenuates BRAF V600E tumor growth
.
Cell Metab
.
2017
;
25
(
2
):
358
73
.
141.
Annunziata
G
,
Capo
X
,
Muscogiuri
G
,
Colao
A
,
Barrea
L
.
Intermittent fasting: a new trend or a valid approach for the treatment of obesity
.
Minerva Endocrinol
.
2023
;
48
(
4
):
367
70
.
142.
Cho
Y
,
Hong
N
,
Kim
KW
,
Cho
SJ
,
Lee
M
,
Lee
YH
, et al
.
The effectiveness of intermittent fasting to reduce body mass index and glucose metabolism: a systematic review and meta-analysis
.
J Clin Med
.
2019
;
8
(
10
):
1645
.
143.
Hutchison
AT
,
Regmi
P
,
Manoogian
ENC
,
Fleischer
JG
,
Wittert
GA
,
Panda
S
, et al
.
Time-restricted feeding improves glucose tolerance in men at risk for type 2 diabetes: a randomized crossover trial
.
Obesity
.
2019
;
27
(
5
):
724
32
.
144.
Jamshed
H
,
Beyl
RA
,
Della Manna
DL
,
Yang
ES
,
Ravussin
E
,
Peterson
CM
.
Early time-restricted feeding improves 24-hour glucose levels and affects markers of the circadian clock, aging, and autophagy in humans
.
Nutrients
.
2019
;
11
(
6
):
1234
.
145.
Lowe
DA
,
Wu
N
,
Rohdin-Bibby
L
,
Moore
AH
,
Kelly
N
,
Liu
YE
, et al
.
Effects of time-restricted eating on weight loss and other metabolic parameters in women and men with overweight and obesity: the TREAT randomized clinical trial
.
JAMA Intern Med
.
2020
;
180
(
11
):
1491
9
.
146.
Tiwari
S
,
Sapkota
N
,
Han
Z
.
Effect of fasting on cancer: a narrative review of scientific evidence
.
Cancer Sci
.
2022
;
113
(
10
):
3291
302
.
147.
de Groot
S
,
Vreeswijk
MP
,
Welters
MJ
,
Gravesteijn
G
,
Boei
JJ
,
Jochems
A
, et al
.
The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: a randomized pilot study
.
BMC Cancer
.
2015
;
15
:
652
.
148.
Dorff
TB
,
Groshen
S
,
Garcia
A
,
Shah
M
,
Tsao-Wei
D
,
Pham
H
, et al
.
Safety and feasibility of fasting in combination with platinum-based chemotherapy
.
BMC Cancer
.
2016
;
16
:
360
.
149.
Harvie
MN
,
Sims
AH
,
Pegington
M
,
Spence
K
,
Mitchell
A
,
Vaughan
AA
, et al
.
Intermittent energy restriction induces changes in breast gene expression and systemic metabolism
.
Breast Cancer Res
.
2016
;
18
(
1
):
57
.
150.
Schreck
KC
,
Hsu
FC
,
Berrington
A
,
Henry-Barron
B
,
Vizthum
D
,
Blair
L
, et al
.
Feasibility and biological activity of a ketogenic/intermittent-fasting diet in patients with glioma
.
Neurology
.
2021
;
97
(
9
):
e953
63
.
151.
Nencioni
A
,
Caffa
I
,
Cortellino
S
,
Longo
VD
.
Fasting and cancer: molecular mechanisms and clinical application
.
Nat Rev Cancer
.
2018
;
18
(
11
):
707
19
.
152.
El-Hawary
A
,
Salem
N
,
Elsharkawy
A
,
Metwali
A
,
Wafa
A
,
Chalaby
N
, et al
.
Safety and metabolic impact of Ramadan fasting in children and adolescents with type 1 diabetes
.
J Pediatr Endocrinol Metab
.
2016
;
29
(
5
):
533
41
.
153.
Safdie
FM
,
Dorff
T
,
Quinn
D
,
Fontana
L
,
Wei
M
,
Lee
C
, et al
.
Fasting and cancer treatment in humans: a case series report
.
Aging
.
2009
;
1
(
12
):
988
1007
.
154.
Patterson
JM
,
Fay
M
,
Exley
C
,
McColl
E
,
Breckons
M
,
Deary
V
.
Feasibility and acceptability of combining cognitive behavioural therapy techniques with swallowing therapy in head and neck cancer dysphagia
.
BMC Cancer
.
2018
;
18
(
1
):
1
.
155.
Vrieling
F
,
Stienstra
R
.
Obesity and dysregulated innate immune responses: impact of micronutrient deficiencies
.
Trends Immunol
.
2023
;
44
(
3
):
217
30
.
156.
Chen
KL
,
Jung
P
,
Kulkoyluoglu-Cotul
E
,
Liguori
C
,
Lumibao
J
,
Mazewski
C
, et al
.
Impact of diet and nutrition on cancer hallmarks
.
J Cancer Prev Curr Res
.
2017
;
7
(
4
):
240
.
157.
Grober
U
,
Holzhauer
P
,
Kisters
K
,
Holick
MF
,
Adamietz
IA
.
Micronutrients in oncological intervention
.
Nutrients
.
2016
;
8
(
3
):
163
.
158.
Fagbohun
OF
,
Gillies
CR
,
Murphy
KPJ
,
Rupasinghe
HPV
.
Role of antioxidant vitamins and other micronutrients on regulations of specific genes and signaling pathways in the prevention and treatment of cancer
.
Int J Mol Sci
.
2023
;
24
(
7
):
6092
.
159.
Research TAIfC
.
Supplements and Cancer
. Available from: https://www.aicr.org/cancer-prevention/healthy-eating/supplements-nutrients/
160.
Clinton
SK
,
Giovannucci
EL
,
Hursting
SD
.
The World cancer research Fund/American Institute for cancer research Third Expert report on diet, nutrition, physical activity, and cancer: impact and future directions
.
J Nutr
.
2020
;
150
(
4
):
663
71
.
161.
Espinosa-Salas
S
,
Gonzalez-Arias
M
.
Nutrition: micronutrient intake, imbalances, and interventions. StatPearls. Treasure island (FL) with ineligible companies
. In:
Disclosure: mauricio Gonzalez-Arias declares no relevant financial relationships with ineligible companies
.
2024
.
162.
Intravenous Vitamin C (PDQ(R))
.
Health professional version. PDQ cancer information summaries
.
2002
Bethesda (MD).
163.
Breast cancer prevention (PDQ(R)): health professional version. PDQ cancer information summaries
.
Bethesda (MD)
;
2002
.
164.
Lung cancer prevention (PDQ(R)): health professional version. PDQ cancer information summaries
.
2002
Bethesda (MD).
165.
Stomach (gastric) cancer prevention (PDQ(R)): health professional version. PDQ cancer information summaries
.
2002
Bethesda (MD).
166.
Klein
EA
,
Thompson
IM
Jr
,
Tangen
CM
,
Crowley
JJ
,
Lucia
MS
,
Goodman
PJ
, et al
.
Vitamin E and the risk of prostate cancer: the selenium and vitamin E cancer prevention trial (SELECT)
.
JAMA
.
2011
;
306
(
14
):
1549
56
.
167.
Du
M
,
Luo
H
,
Blumberg
JB
,
Rogers
G
,
Chen
F
,
Ruan
M
, et al
.
Dietary supplement use among adult cancer survivors in the United States
.
J Nutr
.
2020
;
150
(
6
):
1499
508
.
168.
Tank
M
,
Franz
K
,
Cereda
E
,
Norman
K
.
Dietary supplement use in ambulatory cancer patients: a survey on prevalence, motivation and attitudes
.
J Cancer Res Clin Oncol
.
2021
;
147
(
7
):
1917
25
.
169.
Zeng
Z
,
Mishuk
AU
,
Qian
J
.
Safety of dietary supplements use among patients with cancer: a systematic review
.
Crit Rev Oncol Hematol
.
2020
;
152
:
103013
.
170.
Muscogiuri
G
,
Barrea
L
,
Cantone
MC
,
Guarnotta
V
,
Mazzilli
R
,
Verde
L
, et al
.
Neuroendocrine tumors: a comprehensive review on nutritional approaches
.
Cancers
.
2022
;
14
(
18
):
4402
.
171.
Kucuk
AN
,
Ciftci
S
.
The role of intermittent fasting and the ketogenic diet in cancer disease: can they replace the Mediterranean diet
.
Eur J Cancer Prev
.
2023
;
32
(
6
):
533
43
.
172.
Arends
J
.
Malnutrition in cancer patients: causes, consequences and treatment options
.
Eur J Surg Oncol
.
2024
;
50
(
5
):
107074
.
173.
Kobylinska
M
,
Antosik
K
,
Decyk
A
,
Kurowska
K
.
Malnutrition in obesity: is it possible
.
Obes Facts
.
2022
;
15
(
1
):
19
25
.