1. Introduction
Despite notable advances in cancer detection and treatment, completely eradicating the burden of cancer remains an unlikely outcome. A rational strategy for cancer control involves prioritizing primary prevention while also advancing new therapies and improving early detection. Promoting healthy lifestyle choices is central to effective primary prevention and lowering cancer risk. This includes adopting a predominantly plant-based diet, sustaining healthy body weight over time, engaging in consistent physical activity, avoiding or minimizing tobacco and alcohol use, and reducing exposure to radiation and carcinogenic substances. These preventive behaviors are expected to significantly lower the incidence of several cancers, such as melanoma, breast, colorectal, esophageal, liver, and head and neck cancers [
1].
Specific lifestyle recommendations for preventing melanoma include consistent sunscreen use, wearing protective clothing, seeking shade, limiting sun exposure during peak hours (10 a.m. to 2 p.m.), and avoiding indoor tanning [
2]. The role of carotenoid intake, particularly β-carotene and vitamin A, in cancer prevention has been explored, with findings indicating a notable inverse relationship between their consumption and cancer risk, including lung cancer [
3]. Around 90% of lung cancer cases are attributed to smoking, and the combined use of tobacco and alcohol further elevates the risk of developing cancer. Globally, the use of smokeless tobacco is linked to approximately 400,000 cases of oral cancer, accounting for 4% of all cancers [
4]. Therefore, tobacco cessation and alcohol moderation are likely to play a critical role in reducing cancer incidence.
There is a well-established connection between dietary habits, obesity, metabolic syndrome, and the development of various cancers. These factors are estimated to contribute to about 30–35% of cancer-related deaths, suggesting that many of these could be preventable through dietary interventions. Such approaches include fasting, fasting-mimicking diets, ketogenic diets, and calorie restriction [
5].
Emerging research also highlights the synergistic potential of combining energy restriction —whether dietary or pharmacological— with regular physical activity to promote healthy aging and reduce cancer risk. These combined strategies have been shown to modulate key biological pathways involved in carcinogenesis, including inflammation and metabolic dysregulation [
6].
This narrative review focused on lifestyle-based interventions aimed at improving cancer prevention and reducing associated risks, while also enhancing the overall health of cancer patients. Additionally, it explores the therapeutic potential of specific dietary strategies, nutritional supplements, and complementary and alternative medicine in cancer care.
2. Materials and Methods
This narrative review was conducted through a non-systematic search of the scientific literature in the electronic databases PubMed and Scopus, with additional manual search in Google Scholar to identify studies not indexed in the primary databases. The search was performed on 11 August 2025 and focused on peer-reviewed articles and reviews published within the last 25 years. The following Boolean string was used: ("cancer" OR "neoplasm" OR "tumor" OR "malignancy" OR "oncology" OR "cancer prevention" OR "cancer management") AND ("lifestyle" OR "lifestyle medicine" OR "behavioral interventions" OR "lifestyle interventions" OR "diet" OR "dietary pattern" OR "nutritional interventions" OR "nutrition" OR "physical activity" OR "exercise" OR "body weight" OR "obesity" OR "overweight" OR "micronutrients" OR "vitamins" OR "vitamin D" OR "vitamin B" OR "vitamin C" OR "vitamin E" OR "retinol" OR "beta-carotene" OR "carotenoids" OR "antioxidants" OR "dietary supplements" OR "nutraceuticals") AND ("complementary therapies" OR "complementary and alternative medicine" OR "CAM" OR "integrative oncology" OR "integrative medicine") AND ("carcinogen" OR "carcinogens" OR "tobacco" OR "smoking" OR "alcohol" OR "radiation") AND ("cancer risk" OR "cancer recurrence" OR "cancer outcomes" OR "survivorship") AND ("fasting" OR "intermittent fasting" OR "time-restricted feeding" OR "fasting-mimicking diet" OR "caloric restriction" OR "ketogenic diet") AND ("melanoma" OR "breast cancer" OR "colorectal cancer" OR "esophageal cancer" OR "liver cancer" OR "head and neck cancer" OR "pancreatic cancer" OR "prostate cancer" OR "lung cancer" OR "endometrial cancer"). Given the narrative design, no formal risk-of-bias appraisal was conducted; however, priority was given to recent systematic reviews, randomized trials, and studies in high-impact journals, with triangulation across multiple sources when available.
2.1. Inclusion criteria
Studies were included if they met the following criteria:
Peer-reviewed articles, systematic or narrative reviews, and meta-analyses.
Published between 2001 and 2025.
Focused on human subjects.
Addressed at least one of the following themes: lifestyle interventions, dietary strategies, physical activity, obesity, complementary or alternative medicine, carcinogen exposure, or specific cancer types related to lifestyle factors.
2.2. Exclusion criteria
Studies were excluded if they:
Were not written in English.
Were published before 2001 unless deemed highly relevant.
Focused exclusively on pediatric or veterinary populations.
Reported preliminary data without peer review (e.g., preprints).
Lacked relevance to the scope of lifestyle-based cancer prevention or management.
3. Intermittent and Periodic Energy Restriction (IF/FMD)
Fasting, traditionally associated with religious practices, involves the intentional avoidance of all food or certain types of food for a specified duration. In oncology, prolonged fasting—lasting several weeks or months—may not be advisable for cancer patients, as it can lead to unintended weight loss, which is often counterproductive. However, shorter-term fasting protocols, implemented over several weeks or months and consisting of fasting periods ranging from 12 to 72 hours followed by unrestricted food intake during specific eating windows, may be more feasible and suitable for individuals undergoing cancer treatment [
7].
Intermittent fasting (IF), which involves alternating cycles of eating and fasting and typically includes calorie-restricted intake one to three days per week, is attracting growing global interest, particularly in oncology. This is due to its potential to modulate nutrient metabolism and energy homeostasis, support overall health, and possibly influence the development and progression of disease. A related but milder approach is the fasting-mimicking diet (FMD), which entails consuming a low-calorie, low-protein, plant-based diet for three to five days during each chemotherapy cycle [
8].
The following sections will explore how nutrition influences cancer-related molecular pathways and how fasting regimens may affect tumor growth and treatment response.
3.1. Mechanistic Rationale: Cancer-Related Signaling and Nutritional Modulation
Cancer cells display a variety of traits that drive their malignant behavior. These include accelerated growth, impaired or lost tumor suppressor function, the ability to invade and spread to distant tissues and organs, stimulation of new blood vessel formation (angiogenesis), resistance to programmed cell death (apoptosis) and immune surveillance, as well as alterations in metabolic processes. The availability of nutrients such as sugars and amino acids influences several of these hallmarks, partly by modulating key signaling pathways, including insulin-like growth factor-1 (IGF-1), Ras, protein kinase A (PKA), and the PI3K–AKT pathway [
9]. Persistent activation of these pathways can also result from mutations in oncogenes. To meet increased energy and biosynthetic needs, these signaling cascades often promote glycolysis, leading to elevated levels of glucose transporters and glycolytic enzymes in cancer cells. As a result, many tumor types become dependent on increased glucose intake or the supply of specific amino acids [
10].
Elevated blood glucose levels (hyperglycemia) have been associated with shorter progression-free and overall survival in cancer patients [
11]. This underlines the importance of altered metabolic demands and glucose metabolism in cancer progression and patient outcomes, thereby supporting the potential role of nutritional strategies in regulating cancer cell growth and viability. In this context, fasting has demonstrated benefits for long-term health by decelerating the aging process, extending lifespan, reducing inflammation and oxidative damage, promoting cellular regeneration, and enhancing cardiovascular and cognitive functions [
12]. Fasting also shows promise as a complementary strategy for cancer prevention and treatment by improving the efficacy and tolerability of anticancer therapies. Additionally, it may improve the quality of life in cancer patients through several adaptive biological responses triggered by the fasting state [
13].
3.2. Fasting Regimens and Tumor Biology
Recent research has shown that altering the energy metabolism of cancer cells can suppress tumor development and enhance the immune system’s capacity to combat tumors. For this purpose, there are several fasting regimes that need to be known; an overview is presented in
Table 1.
Intermittent fasting (IF) has been found to decrease the adverse effects associated with chemotherapy and radiotherapy, while simultaneously increasing cancer cells’ sensitivity to these standard treatments. As a result, IF is gaining recognition as a potentially valuable strategy in clinical oncology [
14,
15].
Implementing fasting or a fasting-mimicking diet (FMD) can lead to reduced levels of blood glucose and insulin-like growth factor-1 (IGF-1). These metabolic changes affect several intracellular signaling pathways, such as PI3K-Akt, Ras, and the mammalian target of rapamycin (mTOR), thereby inhibiting cell growth and proliferation [
16]. This is particularly significant because cancer cells are highly dependent on glucose metabolism, which makes them more vulnerable to chemotherapy when FMD is applied. This mechanism is referred to as differential stress sensitization (DSS), and it contributes to enhancing the therapeutic impact of anticancer treatments [
21].
Fasting also activates a biological process known as differential stress resistance (DSR), through which healthy cells shift their focus toward protection and internal repair, potentially reducing the damage caused by chemotherapy. Recent preclinical findings suggest that fasting or FMD not only protects normal cells but may also support effector T-cell–mediated cytotoxicity, thereby strengthening the immune response against tumors [
17].
Under fasting or FMD conditions, reduced insulin, leptin and IGF-1 levels down-modulate PI3K–Akt, Ras–PKA and mTOR–S6K signaling, decrease glucose transporter (GLUT) activity, and promote a metabolic shift toward oxidative phosphorylation (OxPhos) with increased reactive oxygen species (ROS) generation. These changes activate transcription factors such as FOXO, EGR-1 and p53 in normal tissues, enhancing stress-response and repair pathways that support treatment tolerance [
21].
3.3. Preclinical Evidence on Fasting
The benefits of incorporating fasting into cancer treatment have been strongly supported by preclinical research. Among these are the protection of healthy cells from the harmful side effects of chemotherapy, increased sensitivity of tumor cells to chemotherapy, and enhanced intratumoral infiltration of CD8+ T cells induced by chemotherapy, which collectively contribute to slowing disease progression [
22]. In colon cancer mouse models, short-term fasting (STF) causes deficiencies in glucose and amino acids, triggering an anti-Warburg effect. This is marked by increased oxygen consumption without efficient ATP production, leading to elevated oxidative stress and programmed cell death (apoptosis) [
23].
Recent mouse studies have demonstrated that intermittent fasting cycles—periods of 12 to 72 hours without food followed by refeeding—can have positive impacts on lifespan, age-associated diseases, physiological health markers, stress resistance, metabolic balance, and tissue regeneration [
24,
25]. In xenograft mouse models of breast cancer, melanoma, and neuroblastoma, combining fasting with high-dose chemotherapy helped protect healthy cells from oxidative stress, DNA damage, and treatment-related endometrial hyperplasia [
22]. Remarkably, findings from the same study suggest that two separate 48-hour fasting cycles alone were as effective as two rounds of chemotherapy in curbing tumor progression [
22].
These effects are thought to result from reductions in blood glucose, insulin, IGF-1, and inflammatory markers, along with increases in IGF-1-binding proteins (IGFBPs) and ketone bodies [
26]. Altogether, these findings underscore the potential of non-pharmacological dietary strategies, when used alongside conventional treatments, to significantly improve survival outcomes in certain types of cancer.
3.4. Clinical and Translational Evidence on Fasting
To date, only a few small-scale clinical trials have explored the impact of various intermittent fasting protocols used alongside chemotherapy, focusing on their effects on cancer progression and prognosis through the evaluation of metabolic and hormonal parameters. These clinical investigations have confirmed that integrating fasting into chemotherapy regimens is both safe and generally well-tolerated. Moreover, fasting may help reduce the toxicity commonly associated with chemotherapy [
16,
27,
28,
29].
One clinical study examined the safety, immune-modulating, and metabolic outcomes of a five-day, cyclic fasting-mimicking diet (FMD) administered in combination with standard cancer therapies. In a cohort of 101 patients, the FMD was shown to be feasible and well-tolerated, leading to consistent reductions in blood glucose and growth factor levels. These metabolic shifts resemble those seen in preclinical studies and are believed to contribute to the tumor-suppressive properties of fasting and FMD. Additionally, the FMD alters systemic and tumor-specific immune responses, activating multiple immune mechanisms with antitumor effects [
30].
In a broader clinical context, over 300 cancer patients have undergone FMD cycles alongside other therapies, and the occurrence of fasting-related adverse effects has been notably low [
27,
30,
31]. Furthermore, clinical trials have revealed several beneficial outcomes of FMD in terms of cancer-related biological markers, including improved insulin sensitivity, reduced blood glucose, insulin, and leptin levels, and lower systemic inflammation [
21].
Ongoing research is now focusing on developing short-term, controlled, and adaptable fasting protocols as cost-effective and powerful adjuncts to conventional cancer treatments. Preclinical models suggest that fasting implemented around chemotherapy or radiation sessions may reduce treatment-related side effects while enhancing therapeutic efficacy. Current human trials aim to evaluate safety, tolerability, and biological mechanisms underlying this promising strategy. In underweight or sarcopenic patients, fasting-like regimens should be avoided unless embedded in a supervised protocol with weight and albumin monitoring.
4. Ketogenic Strategies in Oncologic Metabolism (LC/KD)
A ketogenic diet (KD) is characterized by an extremely low carbohydrate (LC) intake and is not primarily designed around calorie restrictions or fasting protocols. Typically, its macronutrient composition includes approximately 90% of caloric intake from fats, while carbohydrates and proteins account for only about 2% and 8%, respectively. These ratios align with the conventional KD formulations, typically represented by a fat-to-carbohydrate ratio of 4:1 and a fat-to-protein ratio of 3:1 [
32].
KD is known to effectively stimulate the production of ketone bodies, such as β-hydroxybutyrate and acetoacetate, which can help suppress appetite and maintain low plasma glucose levels in non-cancer individuals [
33]. The consistent high intake of fatty acids combined with minimal carbohydrate consumption drives a metabolic shift from glucose-based to fatty acid-based energy production. This increase in fatty acid oxidation leads to elevated concentrations of ketone bodies in the bloodstream [
34].
The rationale behind KD in oncology stems from the Warburg effect—a metabolic hallmark of cancer cells where energy is primarily generated through glycolysis and lactate fermentation, even in the presence of oxygen. Cancer cells often exhibit increased glucose uptake to support their rapid growth and proliferation. Therefore, reducing carbohydrate intake through KD can help lower blood glucose, insulin, and IGF-1 levels. As many cancer cells depend on glucose for ATP production, carbohydrate restriction can starve tumor cells while allowing normal cells to rely on ketones for energy [
35].
Among ketone bodies, β-hydroxybutyrate has been shown to possess anti-inflammatory effects. It achieves this by downregulating interleukin-1β (IL-1β) expression in bone marrow-derived macrophages and promoting the expression of mitochondrial uncoupling protein 2, thereby protecting against oxidative stress and supporting increased longevity [
36]. Additionally, reduced levels of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), have been observed with KD, suggesting its potential to enhance cancer treatment outcomes through its anti-inflammatory actions [
37].
Evidence also points to KD's role in hindering colon cancer progression by increasing oxidative stress within tumors. This process suppresses matrix metalloproteinase-9 (MMP-9) expression and promotes the conversion of tumor-associated macrophages (TAMs) from the M2 to the M1 phenotype, both of which are associated with anti-tumor activity [
38]. MMPs are crucial contributors to tumor progression, metastasis, angiogenesis, and the degradation of the extracellular matrix [
39].
The potential of low-carbohydrate diets like KD in cancer management is currently under investigation, particularly because of cancer cells’ elevated glucose demands compared to normal cells. While preclinical studies in cellular and animal models have shown promising outcomes for KD in cancer prevention and therapy, clinical data on its safety and effectiveness remain limited and preliminary [
40].
4.1. Preclinical Evidence on Ketogenic Diet
Multiple preclinical studies conducted on mouse models have highlighted the antitumor potential of ketogenic diets (KDs) when used as an adjunct to chemotherapy or radiotherapy. These studies have reported significant inhibitory effects against various types of cancer, including colorectal, gastric, prostate, head and neck, brain, and thyroid cancers [
40].
In a recent investigation, the effectiveness of KDs in suppressing tumor development was demonstrated in autochthonous animal models of colorectal cancer (CRC). The antineoplastic effects of KDs were primarily attributed to the ketone body β-hydroxybutyrate (BHB), which was shown to inhibit the proliferation of colonic crypt cells and markedly reduce the progression of intestinal tumors. Further analysis suggested that BHB mediates its tumor-suppressive action via activation of the hydroxycarboxylic acid receptor 2 (HCAR2) and the transcriptional regulator homeodomain-only protein homeobox (HOPX), ultimately leading to gene expression changes and suppression of cell growth [
41].
Additionally, both BHB and acetone have been identified as modulators of N-methyl-D-aspartate (NMDA) signaling, a mechanism of physiological relevance given the widespread expression of NMDA receptors across various cancer types. Through these mechanisms, the ketogenic diet influences cellular redox balance by altering the ratio between nicotinamide adenine dinucleotide (NAD
+) and its reduced form (NADH), thereby modulating oxidative phosphorylation (OxPhos) and enhancing the production of reactive oxygen species (ROS) within tumor cells. This redox stress can promote apoptosis and sensitize tumors to cytotoxic therapies, while normal cells are generally protected through improved mitochondrial efficiency and adaptive metabolic responses [
42,
43].
Collectively, these preclinical findings suggest that ketogenic interventions may exert context-dependent anticancer effects through the regulation of oxidative stress and metabolic signaling pathways involving HCAR2, NMDA, and ROS.
4.2. Clinical and Translational Evidence on Ketogenic Diet
Several clinical trials have examined the effects of the ketogenic diet (KD) when used alongside chemotherapy, radiotherapy, or antiangiogenic therapies in cancer treatment, producing varied outcomes. Evidence from case reports and small-scale trials suggests that KD is generally safe and well-tolerated by cancer patients. For example, a four-week pilot study involving ten individuals with various types of cancer evaluated the safety and feasibility of KD. The findings showed that patients who experienced stable disease or partial remission had, on average, BHB levels three times higher than those whose disease progressed [
44].
Similarly, Schmidt et al. conducted a pilot trial with 16 advanced-stage cancer patients, where 7 participants received a KD supplemented with oil-protein shakes. The study reported a statistically significant average weight reduction without significant changes in blood lipid or cholesterol levels. Some improvements in quality of life were also noted, and no serious adverse events occurred [
45].
In a retrospective study of 53 patients undergoing treatment for high-grade glioma, six followed a KD composed of 77% fat, 8% carbohydrates, and 15% protein. These patients exhibited a substantial reduction in average blood glucose levels, dropping from 142.5 mg/dl before the diet to 84 mg/dl, compared to 122 mg/dl in patients on a standard diet [
46].
Another randomized controlled trial involving breast cancer patients evaluated the effects of a medium-chain triglyceride (MCT)-based KD. Results showed reductions in fasting blood glucose, body weight, BMI, and body fat percentage, alongside increased blood ketone concentrations, indicating positive metabolic shifts in the intervention group [
46].
Some studies also reported significant reductions in insulin levels among KD participants and an inverse relationship between BHB concentrations and IGF-1 levels [
47]. A growing body of literature on adult cancer patients (aged 18 and over) with various malignancies—including glioblastoma, breast, liver, lung, pancreatic, colorectal, and head and neck cancers—has shown encouraging outcomes. These include improved survival, extended progression-free survival, better responses to conventional therapies, and enhanced quality of life [
48].
Despite these promising findings, the limited scope and scale of existing studies underscore the need for more extensive research. Larger, controlled trials are necessary to thoroughly evaluate KD’s efficacy in cancer treatment and to explore its potential in combination with other therapeutic strategies. KD feasibility hinges on adherence and safety checks; cap unintended weight loss (>5% over a cycle) and discontinue if fasting glucose persistently <70 mg/dL or BHB >4 mmol/L in symptomatic patients.
5. Vitamin D, Carotenoids and Signaling Axes
Dietary supplements refer to ingestible products, such as vitamins, minerals, herbal extracts, and other compounds, that are taken to enhance overall health and complement the regular diet. These supplements are widely used across the globe and encompass a diverse category of consumables that differ from standard foods and pharmaceutical drugs [
49].
5.1. Vitamins and Signaling
Numerous dietary elements, including specific vitamins, have been shown to interfere with key signaling pathways and molecular mechanisms involved in the progression of cancer. Vitamins such as B and D have been extensively studied over the years for their potential to lower cancer risk and mortality [
50]. Researchers have concentrated on the preventive role of vitamin D against various cancer types. In the liver, both vitamin D2 and D3 are converted into the circulating form, 25-hydroxyvitamin D (25(OH)D or calcidiol), which is subsequently transformed into the biologically active form, 1,25-dihydroxyvitamin D (1,25(OH)
2D or calcitriol), within the kidneys [49, 51, 52]. Numerous studies in vitro have demonstrated that vitamin D can inhibit the proliferation and induce apoptosis of various cancer cells, thereby impeding tumor development [
53]. Similarly, B vitamins and their derivatives are vital to human physiology and have been recognized as important modulators of cancer risk [
54]. Despite consistent observational links, results from high-dose supplementation trials are heterogeneous, supporting a ‘target-to-sufficiency’ rather than a pharmacologic approach.
5.1.1. Breast Cancer
Vitamin D plays a vital role in regulating the function of multiple organs, including the mammary glands. Its influence on mammary gland development is primarily mediated through the vitamin D receptor (VDR), which facilitates the vitamin’s anti-proliferative, pro-apoptotic, and differentiation-inducing effects [
55]. The pioneering work of Abe et al. and Colston et al. first identified the anti-cancer properties of vitamin D [
56,
57]. A range of clinical and observational studies have since supported vitamin D's role in modulating both the initiation and progression of cancer [
58,
59,
60]. These findings underscore the involvement of VDR and associated proteins such as CYP27B1 and CYP24A1 in the development of breast cancer (BC) [
61].
In particular, studies have indicated that individuals with triple-negative breast cancer (TNBC) exhibit the lowest vitamin D levels among all breast cancer subtypes, suggesting a possible protective function of vitamin D against TNBC [
61]. Moreover, Bauer et al. conducted a meta-analysis of nine studies involving 5,206 patients and 6,450 controls, revealing a nonlinear inverse relationship between plasma vitamin D levels and breast cancer risk in postmenopausal women [
61,
62]. Additional in vitro and in vivo research has demonstrated that 1,25(OH)
2D
3 and its analogues impact key cancer-related processes in breast tissue, including cell proliferation, apoptosis, differentiation, epithelial–mesenchymal transition, autophagy, metabolism, and the population of cancer stem cells [
63].
Similarly, in a prostate cancer-related in vitro study comparing the MCF-7 breast cancer cell line with the non-tumorigenic MCF10A line, treatment with thiamine (1–2 mg/mL for 24 hours) significantly reduced proliferation in the MCF-7 cells only. This indicates a potential link between elevated levels of vitamin B12 and holohaptocorrin with increased cancer risk. Furthermore, a nested case–control study found a positive association between higher blood vitamin B12 levels and the overall risk of developing lung cancer [
54].
5.1.2. Colorectal Cancer
Vitamin D has been extensively investigated for its potential protective role against colorectal cancer (CRC) [
15,
16,
17,
18,
19,
20]. Higher intake of vitamin D has been associated with a reduced risk of early-onset CRC and its precancerous lesions. Several mechanisms have been proposed to explain this inverse relationship, including vitamin D’s ability to inhibit cell proliferation, migration, invasion, and angiogenesis in colon cancer cells, as well as its regulatory effects on intestinal immune cells [
64]. Furthermore, colorectal cancer patients with elevated pre-diagnostic levels of 25-hydroxyvitamin D [25(OH)D] have demonstrated a lower risk of cancer-related mortality [
65].
The COLON study found that patients with vitamin D levels exceeding 50 nmol/L, combined with higher magnesium intake (above the median), experienced reduced mortality rates from CRC [
66,
67]. In a randomized trial led by Ng et al., serum 25(OH)D levels were measured in 515 patients undergoing chemotherapy for stage 4 CRC. The findings revealed that vitamin D deficiency (defined as <20–30 ng/mL) was highly prevalent in this population [
66,
68]. More recently, a large cohort study by Yuan et al. involving 1,041 patients with previously untreated metastatic or advanced CRC confirmed a widespread deficiency in vitamin D (less than 20–30 ng/mL) among this group [66, 69].
5.1.3. Lung Cancer
Over the past few decades, abnormalities in vitamin D levels or disruptions in vitamin D receptor (VDR) function have been linked to the development of various cancers, including lung cancer [
53]. Epidemiological studies have highlighted the protective role of adequate vitamin D levels in lung cancer prevention. Several investigations have identified a correlation between serum vitamin D levels and the risk of developing lung cancer [
53].
One study demonstrated that higher circulating vitamin D concentrations are associated with lower mortality rates and improved survival outcomes in lung cancer patients [
70]. Additionally, a cohort study in Finland observed an inverse association between vitamin D levels and lung cancer incidence, particularly among women and younger individuals [
53,
71].
Genetic variations in vitamin D signaling pathways may also influence lung cancer susceptibility and the effectiveness of therapeutic responses [
53,
72]. Vitamin D has been shown to upregulate the expression of p53, a well-known tumor suppressor gene, while downregulating the expression of the pro-apoptotic gene Bcl-2 in A549 human lung carcinoma cells [
53,
72].
Furthermore, vitamin D therapy has been reported to enhance the VDR-mediated suppression of the epidermal growth factor receptor (EGFR) gene. Studies have found that vitamin D significantly influences EGFR signaling in lung cancer cell lines, suggesting that vitamin D/VDR-based therapies could be a promising strategy for targeting lung cancer through EGFR modulation [
53,
73,
74]. In addition, a nested case–control (NCC) study found a positive association between elevated blood levels of vitamin B12 and an increased overall risk of lung cancer [
54].
5.1.4. Ovarian Cancer
Numerous studies examining sun exposure have identified an inverse relationship between vitamin D levels and the risk of ovarian cancer. Experimental research using cell lines, human xenograft models, and mouse models has provided compelling evidence that vitamin D contributes to reduced cell proliferation, increased apoptosis, and inhibited tumor progression in ovarian cancer. Additionally, Mendelian randomization studies have shown that genetically lower levels of 25-hydroxyvitamin D [25(OH)D] are significantly associated with an elevated risk of ovarian cancer, thereby reinforcing the potential protective role of vitamin D [
51,
75].
An Australian cohort study found that higher serum levels of 25D3 at diagnosis were associated with longer survival among women with invasive ovarian cancer [
76]. Similarly, Mendelian randomization research conducted in European populations reported that genetically decreased 25D3 levels were inversely associated with ovarian cancer risk [
77]. Evidence from genome-wide association studies (GWAS) also supports a protective role, suggesting that elevated 25D3 concentrations may lower the risk of epithelial ovarian cancer (EOC) [
65,
78].
In a case–control study involving 1,631 women with EOC, higher 25(OH)D levels were linked to improved survival outcomes [
79]. Additional studies have explored variants in the vitamin D receptor (VDR) gene, which modulates the activity of 1,25-dihydroxyvitamin D, and have suggested that these genetic differences may influence both the risk of developing ovarian cancer and survival rates among patients [
80,
81,
82,
83,
84].
To deepen our understanding of vitamin D’s anti-cancer effects and to tailor VDR agonist therapies to individual patients, further research is needed. This should include integrated “omics” approaches—such as epigenomics, metabolomics, genomics, proteomics, and transcriptomics—to better predict individual sensitivities or susceptibilities to vitamin D-based interventions [
50,
85].
5.2. Carotenoids and Retinoid-Related Pathways
Carotenoids, which are antioxidant compounds found in fruits and vegetables, possess immuno-regulatory and anti-mutagenic properties. Numerous studies have reported associations between the intake of various carotenoids and a reduced risk of multiple cancer types. While these associations do not confirm direct causality, they highlight important patterns that suggest a protective effect against cancer development. Beyond their pigment-related aesthetic roles, carotenoids are recognized for their potent antioxidant functions and their role in the biosynthesis of essential nutrients such as vitamin A [
86,
87].
Due to their antioxidant properties, specific carotenoids help mitigate the damaging effects of reactive oxygen species (ROS), which are implicated in the onset of various diseases including cancer, cardiovascular and neurodegenerative disorders, and aging. A broad range of scientific studies have also documented the role of carotenoids in regulating apoptosis, gene expression, angiogenesis, and immune function [
87,
88]. Furthermore, dietary intake of carotenoids is believed to influence cellular pathways associated with antioxidant defense mechanisms [
89].
5.2.1. Head and Neck Cancer
Head and neck cancer (HNC), which encompasses oral cavities, laryngeal, and pharyngeal cancers, has been linked to carotenoid intake due to compounds’ antioxidant, anti-mutagenic, and immune-modulating effects. Although the exact biological mechanisms by which carotenoids may influence HNC risk remain to be fully elucidated, numerous studies have highlighted a potential protective role [
88].
Two meta-analyses conducted by Leoncini et al. examined the association between dietary carotenoid intake and HNC risk [
86,
90]. These analyses reviewed data from sixteen epidemiological studies and found that β-carotene consumption was associated with a significant reduction in HNC risk, including a 46% lower risk of oropharyngeal cancer (OPC) and a 57% lower risk of laryngeal cancer (LC). Similarly, β-cryptoxanthin and lycopene were associated with a 59% and 50% decrease in LC risk, respectively. Moreover, α-carotene, β-cryptoxanthin, and lycopene intake correlated with a 26% reduced risk of oral cavity (OCC) and pharyngeal (PC) cancers [
86,
88].
In another pooled analysis of ten case–control studies conducted across North America, Japan, and Europe, involving 18,207 patients and 12,248 controls, researchers reported that higher overall carotenoid consumption was associated with a 39% reduction in the risk of OCC and PC, as well as a 39% lower risk of LC [
90].
5.2.2. Breast Cancer
Research indicates that carotenoids may play a significant protective role against prostate and breast cancers. In a recent in vitro study, treatment with 1 μM of β-carotene was shown to suppress the expression of anti-apoptotic proteins such as PARP and Bcl-2. Similarly, studies using the MCF-7 breast cancer cell line demonstrated that lycopene concentrations of 2 μM or higher reduced cell proliferation and enhanced apoptosis, accompanied by increased mRNA expression of p53 and Bax—two key regulators of cell death [
91,
92].
A meta-analysis conducted by Fulan Hu and colleagues examined the relationship between carotenoid intake and breast cancer risk, including dose–response patterns [
93]. Their findings indicated that higher dietary intake of α-carotene was significantly associated with a reduced risk of breast cancer, though the associations involving dietary and total β-carotene intake remain inconclusive and require further exploration [
93].
Another large case–control study involving 5,707 women with invasive breast cancer and 6,389 controls—comprising both pre- and postmenopausal women from Wisconsin, Massachusetts, and New Hampshire—investigated the links between vegetable, fruit, vitamin, and carotenoid consumption and breast cancer risk. The study found that high carotenoid intake was associated with a decreased breast cancer risk in premenopausal women, while no such protective effect was observed in postmenopausal participants, particularly among smokers [
94]. Overall, these findings support the potential of carotenoids to inhibit breast cancer cell growth [
91,
92].
5.2.3. Colorectal Cancer
Numerous epidemiological and clinical studies have linked the risk of colorectal cancer (CRC) to both dietary and serum levels of carotenoids and retinol. In a cell culture study, lycopene at concentrations of 2 μM or higher significantly inhibited the proliferation of HT-29 human colorectal cancer cells. This antiproliferative effect is thought to involve suppression of the PI3K/Akt signaling pathway [
95]. Additionally, lycopene has been shown to increase the expression of p21 and PCNA, while reducing levels of pro-inflammatory markers such as PGE2, COX-2, and phosphorylated ERK1/2 in animal models. Plasma concentrations of matrix metalloproteinase-9 (MMP-9) have also been inversely correlated with dietary lycopene intake [
96].
A meta-analysis conducted by Xiaoyong Han and colleagues reviewed 22 studies examining the relationship between colorectal cancer risk and serum or dietary levels of carotenoids and retinol [
97]. Their findings confirmed that higher levels of β-cryptoxanthin, lycopene, α-carotene, and β-carotene were significantly associated with a reduced risk of CRC. These results support the potential role of lycopene and other carotenoids in inhibiting tumor development and progression in colorectal cancer [
96].
5.2.4. Lung Cancer
Carotenoids help protect the lungs from early stages of cancer development through their antioxidant properties, which are obtained at varying dietary concentrations. Multiple studies have reported a linear dose–response relationship between β-carotene intake and lung cancer risk, indicating a 2% reduction in risk for every additional milligram of β-carotene consumed daily. Likewise, a 1% decrease in lung cancer risk has been observed for every additional 10 μg of β-cryptoxanthin intake per day, and a 3% reduction for each extra milligram of lycopene consumed daily [
98].
A recent case–control study conducted in Hawaii also reported an inverse association between lung cancer risk and the consumption of vegetables rich in carotenoids, suggesting a potential protective role of carotenoids against lung cancer development [
99]. Furthermore, a random-effects meta-analysis by Gallicchio and colleagues—encompassing six clinical trials and 25 observational studies—found an overall inverse association between carotenoid intake and lung cancer risk. However, the observed reduction in risk was generally modest and did not reach statistical significance [
98]. Beta-carotene supplementation in smokers has shown harm in legacy trials, underscoring that food-based carotenoids should not be conflated with high-dose single-nutrient pills.
5.2.5. Prostate Cancer
Due to their antioxidant properties, carotenoids may help reduce tumor-promoting inflammation. Among them, α-carotene, β-carotene, and lycopene have been extensively studied in relation to prostate cancer. Over seventy studies have investigated their associations, consistently finding that higher dietary intake and blood concentrations of lycopene are linked to a decreased risk of developing prostate cancer. Supporting this, two cell culture studies have shown that lycopene treatment significantly reduces cell proliferation and impairs cell cycle progression in prostate cancer cells [
91,
100].
Additionally, a nested case–control study examined the relationship between prostate cancer risk and plasma levels of carotenoids such as cis-lycopene, trans-lutein, β-trans-carotene, and dihydrolycopene. The findings indicated that, particularly among African American men, trans β-carotene isomers were positively associated with increased prostate cancer risk, regardless of smoking habits or multivitamin use [
101].
Furthermore, a separate case–control study involving 65 prostate cancer patients and 132 control subjects assessed the influence of plasma levels of lycopene and other carotenoids and vitamins, including retinol and α- and γ-tocopherols. The study revealed a significant inverse association between prostate cancer and plasma levels of lycopene and zeaxanthin, while β-cryptoxanthin and lutein showed borderline associations with reduced risk [
102].
6. Physical Activity and Sedentary Behavior
At present, smoking remains the leading modifiable risk factor for cancer. However, physical inactivity is rapidly emerging as the second-most significant preventable contributor to cancer development [
103]. According to the World Health Organization (WHO), more than 1.4 billion adults—over a quarter of the global population—do not engage in sufficient physical activity [
104]. With obesity now affecting individuals at younger ages, people are increasingly exposed to related health risks throughout their lives. High-income countries report rates of physical inactivity that are approximately twice as high as those in low-income nations. The global trend toward reduced physical activity has been steadily rising, posing negative consequences for environmental sustainability, economic productivity, quality of life, and the overall health of communities [
105,
106].
Regular engagement in physical activity is widely recognized as a key preventive measure against non-communicable diseases such as cardiovascular disease, hypertension, type 2 diabetes, stroke, and numerous types of cancer [
6,
107,
108,
109,
110,
111,
112,
113,
114]. For decades, epidemiological research has consistently linked higher levels of physical activity with a lower risk of several common cancers. In 2012, it was estimated that the elevated body mass index (BMI) was responsible for 3.6% of all new cancer cases globally [
115].
6.1. Prevention: Physical Activity and Cancer Risk
Any bodily movement driven by skeletal muscles that leads to energy expenditure qualifies as physical activity. While exercise is a subset of physical activity, it encompasses a wide range of intensities and forms, including occupational tasks, household chores, commuting, and recreational pursuits like walking, cycling, sports, and active play [
116]. Engaging in physical activity positively influences the body’s metabolic processes, making it more resistant to various types of cancer. The nature and intensity of the activity can impact metabolism and energy balance, thereby influencing how much food and nutrients can be consumed without leading to fat accumulation. When caloric intake exceeds energy expenditure, it results in a positive energy balance, promoting weight gain and increased body fatness [
116].
Although physical activity is often emphasized for weight management, it also offers direct cancer-protective benefits through several biological mechanisms. These include modulation of the immune system [
117,
118,
119], reduction in circulating estrogen [
120], decreased oxidative stress [
121], improved insulin sensitivity [
122], suppression of inflammation [
123], favorable epigenetic changes [
124], reduced telomere shortening [
125], and enhanced DNA repair [
118]—all of which are associated with the initiation and progression of cancer.
Independent of its effects on obesity, physical activity has been inversely associated with colon and breast cancer, as demonstrated in a 2020 Mendelian randomization study using UK Biobank data. This inverse relationship was found to be independent of BMI [
126].
Moreover, strength training, which builds skeletal muscle mass, is linked to lower risks of kidney, bladder, and colorectal cancers [
127,
128]. Aerobic exercise also improves glucose metabolism and insulin sensitivity, potentially reducing the risk of cancers associated with insulin resistance, such as colon, liver, pancreatic, and endometrial cancers [
129,
130]. While not all cancers are equally affected by physical activity, lifestyle interventions, including regular exercise, have shown potential to lower overall cancer risk [
131]. For example, intense domestic activities like heavy housework may offer protective benefits against breast cancer. Exercising for three to five hours per week has been associated with a reduced risk of developing cancer [
132].
One study found that individuals who exercised had a 15–20% lower risk of breast cancer and a 24% reduced risk of colorectal cancer [
133]. Exercise has been shown to reduce the risk of breast cancer in postmenopausal women and may help protect them from autonomic nervous system dysfunction [
134,
135]. Among 126 studies on exercise and colorectal cancer (CRC), more active participants had a 19% lower risk of CRC [
136]. Similarly, a meta-analysis on stomach cancer, encompassing 10 cohort and 12 case–control studies, reported a 19% risk reduction among those who exercised more frequently [
137].
Given that obesity is a known risk factor for endometrial cancer, exercise-induced weight loss plays a protective role. According to a review of 33 studies, women who exercised regularly had a 20% lower risk of endometrial cancer [
138]. A similar review on esophageal cancer found that those with the highest levels of physical activity had a 21% lower risk compared to the least active individuals [
139]. In a large-scale study involving over 1 million participants, leisure-time exercise was associated with a 13% lower risk of bladder cancer and a 23% lower risk of kidney cancer [
140]. A review of 25 studies found that while physical activity had little effect on lung cancer risk in non-smokers, it did lower the risk among smokers [
141].
6.2. Physical Activity During and After Therapy
Beyond its role in cancer prevention, exercise has been shown to reduce the adverse effects associated with cancer treatments. Therapies such as chemotherapy and targeted treatments often produce side effects including fatigue, cognitive decline, depression, muscle and bone loss, and cardiac toxicity. These side effects can negatively impact treatment outcomes and overall quality of life [
131]. In some cases, patients are unable to complete or tolerate therapy due to the severity of these side effects, potentially altering the progression of the disease. However, substantial evidence indicates that engaging in physical activity can help alleviate many of these treatment-related complications.
Exercise training influences both cancer-specific and physiological as well as psychological outcomes associated with treatment [
107,
108,
109,
110,
111]. Not only can exercise reduce the side effects of therapy, but it may also enhance treatment efficacy, making it a valuable addition to the therapeutic process [
142]. While some healthcare professionals express concern about whether physical activity could influence survival or prognosis, current research suggests that exercise may not significantly affect survival rates for cancers such as gastric cancer, non-Hodgkin lymphoma, and several others [
143].
Exercise can also be leveraged as a form of cancer-suppressive therapy by disrupting cancer metabolism, particularly anaerobic glycolysis. The impact of physical activity on tumor metabolism depends on its frequency, duration, intensity, and type. By decreasing vasoconstriction in hypoxic environments, exercise improves tumor perfusion and reduces hypoxia, making the cancer microenvironment less aggressive and more susceptible to treatment [
144,
145].
Additionally, regular physical activity helps counteract immunosenescence, the aging of the immune system, which is marked by decreased natural killer (NK) cell activity, chronic inflammation, impaired antigen presentation by dendritic and monocyte cells, and a reduction in naïve T cells capable of targeting emerging cancer cells. These changes contribute to the heightened cancer risk in older adults [
146]. Exercise has been shown to slow immune aging by enhancing immune cell function [
124], thereby playing a protective role against immune-related malignancies.
In summary, robust evidence supports that physical activity reduces cancer risk, curbs tumor growth and progression, mitigates the side effects of cancer treatments, improves treatment adherence, and enhances patient quality of life. A pragmatic mix of aerobic plus resistance exercise counters treatment-related sarcopenia and supports dose intensity, even when total weekly volume is modest.
7. Adiposity and Metabolic Health
A body with overweight or obesity (defined as excessive fat accumulation) contains a higher proportion of fat compared to lean tissues such as muscle and bone. Carrying excess weight at unhealthy levels increases the risk of developing certain types of cancer and raises the likelihood of recurrence after treatment. According to the World Health Organization, in 2016, over 340 million children and adolescents and 1.9 billion adults were living with overweight or obesity, a number projected to rise further [
147]. The global cancer burden is expected to continue growing due to ongoing trends of reduced physical activity and increased body fat, particularly in the context of an aging global population. If these trends persist, overweight and obesity may soon surpass smoking as the leading preventable cause of cancer.
7.1. Obesity and Cancer: Epidemiology and Mechanisms
Since the early 20th century, the relationship between body fat and cancer has been explored in scientific literature, with more focused discussions emerging since 1987 [
148]. As the prevalence of overweight and obesity continues to rise, so does the incidence of cancers associated with excess body fat. This trend is expected to significantly increase the cost of future cancer treatments and place a growing financial burden on healthcare systems due to the long-term management of related comorbidities.
Maintaining a healthy weight is one of the most effective strategies for cancer prevention. Importantly, excess body fat during childhood has been linked to an increased risk of developing various diseases, including cancer, later in life. Hidayat et al. identified associations between early-life adiposity and the development of eight different types of cancer in adulthood [
149]. Moreover, weight gain in adulthood is associated with a higher risk of overweight, obesity, and postmenopausal breast cancer, with a 6% increased risk for every 5 kg gained [
150]. Despite growing evidence of the link between obesity and cancer in adults, the impact of weight gain throughout adulthood is often underestimated.
Body fatness contributes to cancer risk through several biological mechanisms. Obesity is associated with elevated fasting insulin levels and increased inflammatory mediators, both of which promote cell proliferation (Hallmark: sustained proliferative signaling) [
151]. Additionally, obesity can impair apoptosis, the process by which damaged cells are eliminated, thereby allowing abnormal cells to survive (Hallmark: resisting cell death) [
152]. Cancer cells can also spread through the bloodstream or lymphatic system (Hallmark: activating invasion and metastasis). These endocrine and metabolic disruptions, particularly involving insulin and inflammation, highlight the complex relationship between obesity and cancer development.
7.2. Clinical weight-loss strategies in oncology
Combined dietary energy restriction and progressive exercise represent first-line, scalable interventions to reduce adiposity, improve insulin resistance, and lower low-grade inflammation, mechanisms tightly linked to carcinogenesis and poorer outcomes [
6,
122,
123]. Structured programs should favor protein-adequate, minimally processed diets and supervised training tailored to treatment phase and fatigue [
107].
7.3. Survivorship and relapse risk
Post-treatment weight gain is common and associated with higher recurrence and mortality in several cancers [
131,
150]. Survivorship care plans should embed long-term weight management with behavioral support, periodic monitoring, and referral pathways to exercise oncology services [
131].
8. Carcinogenic Exposures: Tobacco and Ultraviolet (UV) Radiation
8.1. Tobacco as a Carcinogen
Tobacco and tobacco smoke contain a complex mixture of over 9,500 chemical substances, many of which have been identified by regulatory authorities as harmful to human health. In 2012, the U.S. Food and Drug Administration (FDA) published a list of hazardous or potentially hazardous constituents found in both tobacco smoke and unburned tobacco, identifying 79 of them as carcinogenic [
153]. Tobacco smoke increases cancer risk primarily due to its content of carcinogens such as nitrosamines, acrylamides, polycyclic aromatic hydrocarbons (PAHs), cadmium, and volatile organic compounds (VOCs) [
154,
155].
There is a well-established causal link between smoking and lung cancer risk [
154]. Extensive experimental and epidemiological evidence confirms that smoking is a major risk factor for several cancers, including those of the kidney, bladder, gastrointestinal tract, head and neck, colorectum, esophagus, pancreas, stomach, liver, cervix, and myeloid leukemia [
153,
154,
156,
157,
158].
Animal studies have shown that several carcinogens in tobacco smoke can induce mammary tumors, and human research has demonstrated that various tobacco-derived compounds can reach breast tissue. Furthermore, the absorption and metabolic activation of carcinogens implicated in breast cancer—such as 4-aminobiphenyl and PAHs—are significantly higher in smokers than in non-smokers [
157].
Head and neck squamous cell carcinoma (HNSCC) is a cancer type strongly associated with tobacco use. The risk of developing HNSCC is nearly ten times higher in smokers compared to non-smokers, and approximately 70–80% of newly diagnosed cases are linked to the combined use of tobacco and alcohol [
158].
The carcinogenic effects of tobacco smoke are largely mediated by reactive oxygen species (ROS) and reactive nitrogen species (RNS), which damage key macromolecules such as DNA, lipids, and proteins. Scientific evidence indicates that smoking-induced oxidative stress plays a central role in both cancer development and inflammation. This oxidative stress triggers inflammatory responses, which in turn generate additional ROS, perpetuating a cycle of molecular damage that may initiate carcinogenesis [
159].
8.2. Non-Ionizing Radiation as a Carcinogen
Human exposure to radiofrequency electromagnetic fields (RF-EMF) primarily arises from the use of personal electronic devices—such as mobile phones, Bluetooth headsets, cordless phones, and amateur radios—as well as from occupational and environmental sources. Occupational exposure includes equipment like induction heaters, high-frequency dielectric devices, and high-powered pulsed radars, while environmental sources involve medical technologies, broadcast antennas, and mobile phone base stations. Among the general population, the highest levels of exposure typically come from handheld devices used in close proximity to the body, particularly mobile phones. Additionally, numerous occupational groups are regularly exposed to RF radiation, including radar operators, military and security personnel using walkie-talkies, technicians servicing broadcast antennas, plastic welders, workers involved in RF-based testing or drying processes, and physiotherapists using diathermy equipment [
160].
Stang et al. conducted both hospital- and population-based case–control studies investigating the development of uveal melanoma in relation to occupational RF radiation exposure and reported an increased risk among individuals exposed to radiofrequency-emitting devices, such as radios and mobile phones [
161]. Similarly, Karipidis et al. carried out a case–control study across five major population centers in Victoria, Australia, identifying a potential link between occupational RF exposure and glioma risk [
162].
Ultraviolet (UV) radiation, a non-visible component of the electromagnetic spectrum with shorter wavelengths than visible light, is also recognized as carcinogenic. While sunlight is the primary source of UV radiation, certain industrial activities—such as electric arc welding—can also emit significant UV levels. The International Commission on Non-Ionizing Radiation Protection (ICNIRP) has established a safety threshold for artificial UV exposure to the eyes and unprotected skin, recommending a limit of 30 J/m
2 over an 8-hour period [
163]. UV radiation is a well-known risk factor for basal cell carcinoma (BCC), and prolonged occupational exposure to sunlight is believed to increase the risk of developing this form of skin cancer. A total of 19 case–control and five cohort studies have reported a significant association between occupational UV exposure and BCC incidence [
164].
From a public-health perspective, UV avoidance and photoprotection remain the most actionable radiation-related strategies for cancer risk reduction in the general population.
9. Complementary and Mind–Body Approaches
The term complementary and alternative medicine (CAM) refers to a wide array of medical and health-related practices and products that fall outside the scope of conventional medicine. Among individuals with cancer, CAM is often used alongside standard treatments to help alleviate stress and anxiety related to the disease and its therapies, as well as to manage side effects such as nausea, pain, and fatigue. Common CAM approaches in oncology include herbal medicine, acupuncture, mind–body practices—such as meditation, yoga, and Tai Chi—nutritional supplements, energy-based therapies, and massage [
165,
166,
167,
168].
The use of CAM among cancer patients has been steadily increasing. It is estimated that between 33% and 47% of patients worldwide incorporate complementary, alternative, or integrative therapies into their care [
169]. One of the most burdensome symptoms experienced by cancer patients is cancer-related fatigue (CRF), which significantly impairs quality of life by affecting daily functioning, social engagement, work performance, leisure activities, and interpersonal relationships. CRF can also hinder adherence to prescribed cancer treatments.
As a result, many patients turn to CAM in an effort to regain a sense of well-being and better manage their symptoms [
170,
171]. While some of these therapies may offer symptom relief and contribute to improved quality of life, their therapeutic efficacy remains variable. Further research is needed to determine how best to integrate these modalities into evidence-based cancer care. Given heterogeneity in quality and potential drug–herb interactions, CAM use should be discussed with the oncology team and, where possible, evaluated within clinical protocols.
10. Limitations
Since this is a narrative review, a systematic study selection protocol was not followed (no protocol registration, no predefined eligibility with dual independent screening, and no formal risk-of-bias assessment). This may introduce a selection bias, as the inclusion of articles was partially based on the authors’ criteria. However, this approach allowed for a deeper and more critical discussion of the available evidence, facilitating the formulation of practical recommendations based on the most relevant literature. Nonetheless, the non-systematic search strategy may have led to the omission of relevant studies, particularly those not indexed in the selected databases and to susceptibility to publication bias. The English-language focus may also have excluded pertinent non-English studies, and heterogeneity across study designs and populations limits generalizability. Some emerging or less-studied interventions may have been underrepresented due to the broad scope of the review. Accordingly, the findings should be interpreted as hypothesis-generating rather than causal. Future systematic reviews and meta-analyses are warranted to validate and expand upon the insights presented here.
11. Conclusions and Future Perspectives
Modifiable lifestyle factors such as physical activity, body weight regulation, dietary habits, tobacco and alcohol use, and exposure to environmental carcinogens play a decisive role in both the prevention and management of cancer. A growing body of epidemiological and mechanistic evidence confirms that adopting healthy behaviors significantly reduces cancer incidence, improves treatment tolerability, and enhances long-term survivorship outcomes. In particular, regular physical activity emerges as a key modulator of cancer risk and prognosis by influencing multiple biological pathways, including hormonal regulation, immune function, oxidative stress, and inflammation.
Excess body fat, increasingly prevalent across all age groups, is strongly associated with higher cancer risk and poorer post-diagnosis outcomes. The obesity epidemic, driven by sedentary lifestyles and high-calorie diets, poses a major public health challenge that may soon surpass tobacco as the leading preventable cause of cancer. Weight management through a combination of physical activity and dietary intervention should therefore be prioritized in cancer control strategies.
While the harmful effects of tobacco and radiation exposure are well established, the ongoing dissemination of new technologies and changing occupational environments calls for continued surveillance and updated safety standards. Population-wide interventions, such as taxation policies on tobacco and ultra-processed foods, stricter workplace protections, and educational campaigns, remain essential to mitigate carcinogenic exposures and promote sustainable prevention.
In addition to conventional medical approaches, the integration of evidence-based complementary and alternative medicine (CAM) into oncology care is gaining traction. CAM interventions—such as mind–body therapies, acupuncture, and herbal medicine—may help manage symptoms like fatigue and anxiety, although their effectiveness remains variable and often underexplored. Rigorous clinical and translational studies are still needed to establish safety profiles, mechanisms of action, and best practices for integrating CAM into evidence-based oncology.
From a clinical and public health standpoint, lifestyle interventions remain the most accessible, cost-effective, and impactful tools for cancer prevention and control. Future research should refine individualized prevention strategies, define dose–response relationships for diet and physical activity, and explore the interplay between lifestyle and genetic predisposition. Moreover, creating supportive environments through urban planning that promotes active living and school- or community-based health education will be critical to sustaining behavioral change across the lifespan.
Ultimately, lifestyle medicine should be regarded not as an adjunct but as a core pillar of modern oncology, integrating nutrition, exercise, and behavioral interventions within standard care pathways to improve outcomes and quality of life. Implementation priorities include embedding exercise oncology and nutrition services in cancer centers, developing pragmatic home- and community-based programs, and ensuring equitable access across healthcare systems.
Author Contributions
Conceptualization, G.G. and F.F.; methodology, G.G. and A.P.; software, G.G. and A.P.; resources, S.C. and F.F.; writing—original draft preparation, G.G. and A.P.; writing—review and editing, S.C. and F.F.; supervision, G.G., S.C. and F.F. All authors have read and agreed to the published version of the manuscript.
Funding
This research received no external funding.
Institutional Review Board Statement
Not applicable.
Informed Consent Statement
Not applicable.
Data Availability Statement
Not applicable.
Conflicts of Interest
The authors declare no conflicts of interest.
References
- LoConte, N. K., Gershenwald, J. E., Thomson, C. A., Crane, T. E., Harmon, G. E., & Rechis, R. (2018). Lifestyle modifications and policy implications for primary and secondary cancer prevention: Diet, exercise, sun safety, and alcohol reduction. American Society of Clinical Oncology Educational Book, 38, 88–100. [CrossRef]
- Tripp, M. K., Watson, M., Balk, S. J., Swetter, S. M., & Gershenwald, J. E. (2016). State of the science on prevention and screening to reduce melanoma incidence and mortality: The time is now. CA: A Cancer Journal for Clinicians, 66(6), 460–480.
- Sun, Y., Wu, J., Yoon, H.-S., Buchowski, M. S., Cai, H., Deppen, S. A., et al. (2022). Associations of dietary intakes of carotenoids and vitamin A with lung cancer risk in a low-income population in the southeastern United States. Cancers, 14(20), 5159.
- Anand, P., Kunnumakara, A. B., Sundaram, C., Harikumar, K. B., Tharakan, S. T., Lai, O. S., et al. (2008). Cancer is a preventable disease that requires major lifestyle changes. Pharmaceutical Research, 25, 2097–2116.
- Vernieri, C., Nichetti, F., Raimondi, A., Pusceddu, S., Platania, M., Berrino, F., et al. (2018). Diet and supplements in cancer prevention and treatment: Clinical evidences and future perspectives. Critical Reviews in Oncology/Hematology, 123, 57–73.
- Cagigas, M. L., De Ciutiis, I., Masedunskas, A., & Fontana, L. (2025). Dietary and pharmacological energy restriction and exercise for healthspan extension. Trends in Endocrinology and Metabolism, 36(6), 521–545.
- Doyle, C., Kushi, L. H., Byers, T., Courneya, K. S., Demark-Wahnefried, W., Grant, B., et al. (2006). Nutrition and physical activity during and after cancer treatment: An American Cancer Society guide for informed choices. CA: A Cancer Journal for Clinicians, 56(6), 323–353.
- Brandhorst, S., Choi, I. Y., Wei, M., Cheng, C. W., Sedrakyan, S., Navarrete, G., et al. (2015). A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metabolism, 22(1), 86–99.
- Levine, M. E., Suarez, J. A., Brandhorst, S., Balasubramanian, P., Cheng, C.-W., Madia, F., et al. (2014). Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metabolism, 19(3), 407–417.
- Schiliro, C., & Firestein, B. L. (2021). Mechanisms of metabolic reprogramming in cancer cells supporting enhanced growth and proliferation. Cells, 10(5), 1056.
- Lin, X., Xiao, Z., Chen, T., Liang, S. H., & Guo, H. (2020). Glucose metabolism on tumor plasticity, diagnosis, and treatment. Frontiers in Oncology, 10, 317.
- Mattson, M. P., Longo, V. D., & Harvie, M. (2017). Impact of intermittent fasting on health and disease processes. Ageing Research Reviews, 39, 46–58.
- Tiwari, S., Sapkota, N., & Han, Z. (2022). Effect of fasting on cancer: A narrative review of scientific evidence. Cancer Science, 113(10), 3291–3302.
- Patterson, R. E., & Sears, D. D. (2017). Metabolic effects of intermittent fasting. Annual Review of Nutrition, 37.
- Longo, V. D., & Mattson, M. P. (2014). Fasting: Molecular mechanisms and clinical applications. Cell Metabolism, 19(2), 181–192.
- Safdie, F. M., Dorff, T., Quinn, D., Fontana, L., Wei, M., Lee, C., et al. (2009). Fasting and cancer treatment in humans: A case series report. Aging, 1(12), 988.
- Brandhorst, S. (2021). Fasting and fasting-mimicking diets for chemotherapy augmentation. Geroscience, 43(3), 1201–1216.
- Longo, V. D., Di Tano, M., Mattson, M. P., & Guidi, N. (2021). Intermittent and periodic fasting, longevity and disease. Nature Aging, 1(1), 47–59.
- Turbitt, W. J., Demark-Wahnefried, W., Peterson, C. M., & Norian, L. A. (2019). Targeting glucose metabolism to enhance immunotherapy: Emerging evidence on intermittent fasting and calorie restriction mimetics. Frontiers in Immunology, 1402.
- Wei, M., Brandhorst, S., Shelehchi, M., Mirzaei, H., Cheng, C. W., Budniak, J., et al. (2019). Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Science Translational Medicine, 9(377), eaai8700.
- Blaževitš, O., Di Tano, M., & Longo, V. D. (2023). Fasting and fasting mimicking diets in cancer prevention and therapy. Trends in Cancer, 9(3), 212–222.
- Lee, C., Raffaghello, L., Brandhorst, S., Safdie, F. M., Bianchi, G., Martin-Montalvo, A., et al. (2012). Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Science Translational Medicine, 4(124), 124ra27.
- Bianchi, G., Martella, R., Ravera, S., Marini, C., Capitanio, S., Orengo, A., et al. (2015). Fasting induces anti-Warburg effect that increases respiration but reduces ATP-synthesis to promote apoptosis in colon cancer models. Oncotarget, 6(14), 11806.
- De Cabo, R., & Mattson, M. P. (2019). Effects of intermittent fasting on health, aging, and disease. The New England Journal of Medicine, 381(26), 2541–2551.
- Longo, V. D., & Cortellino, S. (2020). Fasting, dietary restriction, and immunosenescence. Journal of Allergy and Clinical Immunology, 146(5), 1002–1004.
- Vernieri, C., Casola, S., Foiani, M., Pietrantonio, F., de Braud, F., & Longo, V. (2016). Targeting cancer metabolism: Dietary and pharmacologic interventions anticancer metabolic therapies. Cancer Discovery, 6(12), 1315–1333.
- de Groot, S., Vreeswijk, M. P., Welters, M. J., Gravesteijn, G., Boei, J. J., Jochems, A., et al. (2015). The effects of short-term fasting on tolerance to (neo)adjuvant chemotherapy in HER2-negative breast cancer patients: A randomized pilot study. BMC Cancer, 15(1), 1–9.
- Dorff, T. B., Groshen, S., Garcia, A., Shah, M., Tsao-Wei, D., Pham, H., et al. (2016). Safety and feasibility of fasting in combination with platinum-based chemotherapy. BMC Cancer, 16(1), 1–9.
- Bauersfeld, S. P., Kessler, C. S., Wischnewsky, M., Jaensch, A., Steckhan, N., Stange, R., et al. (2018). The effects of short-term fasting on quality of life and tolerance to chemotherapy in patients with breast and ovarian cancer: A randomized cross-over pilot study. BMC Cancer, 18, 1–10.
- Vernieri, C., Fucà, G., Ligorio, F., Huber, V., Vingiani, A., Iannelli, F., et al. (2022). Fasting-mimicking diet is safe and reshapes metabolism and antitumor immunity in patients with cancer. Cancer Discovery, 12(1), 90–107.
- de Groot, S., Lugtenberg, R. T., Cohen, D., Welters, M. J., Ehsan, I., Vreeswijk, M. P., et al. (2020). Fasting mimicking diet as an adjunct to neoadjuvant chemotherapy for breast cancer in the multicentre randomized phase 2 DIRECT trial. Nature Communications, 11(1), 3083.
- Allen, B. G., Bhatia, S. K., Anderson, C. M., Eichenberger-Gilmore, J. M., Sibenaller, Z. A., Mapuskar, K. A., et al. (2014). Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biology, 2, 963–970.
- Yancy, W. S., Jr., Olsen, M. K., Guyton, J. R., Bakst, R. P., & Westman, E. C. (2004). A low-carbohydrate, ketogenic diet versus a low-fat diet to treat obesity and hyperlipidemia: A randomized, controlled trial. Annals of Internal Medicine, 140(10), 769–777.
- Dhamija, R., Eckert, S., & Wirrell, E. (2013). Ketogenic diet. Canadian Journal of Neurological Sciences, 40(2), 158–167.
- Choi, J.-W., & Hua, T. N. (2021). Impact of lifestyle behaviors on cancer risk and prevention. Journal of Lifestyle Medicine, 11(1), 1.
- Shimazu, T., Hirschey, M. D., Newman, J., He, W., Shirakawa, K., Le Moan, N., et al. (2013). Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science, 339(6116), 211–214.
- Talib, W. H., Mahmod, A. I., Kamal, A., Rashid, H. M., Alashqar, A. M. D., Khater, S., et al. (2021). Ketogenic diet in cancer prevention and therapy: Molecular targets and therapeutic opportunities. Current Issues in Molecular Biology, 43(2), 558–589.
- Zhang, N., Liu, C., Jin, L., Zhang, R., Wang, T., Wang, Q., et al. (2020). Ketogenic diet elicits antitumor properties through inducing oxidative stress, inhibiting MMP-9 expression, and rebalancing M1/M2 tumor-associated macrophage phenotype in a mouse model of colon cancer. Journal of Agricultural and Food Chemistry, 68(40), 11182–11196.
- Said, A. H., Raufman, J. P., & Xie, G. (2014). The role of matrix metalloproteinases in colorectal cancer. Cancers, 6(1), 366–375.
- Elisia, I., & Krystal, G. (2021). The pros and cons of low carbohydrate and ketogenic diets in the prevention and treatment of cancer. Frontiers in Nutrition, 8, 634845.
- Dmitrieva-Posocco, O., Wong, A. C., Lundgren, P., Golos, A. M., Descamps, H. C., Dohnalová, L., et al. (2022). β-Hydroxybutyrate suppresses colorectal cancer. Nature, 605(7908), 160–165.
- Pflanz, N. C., Daszkowski, A. W., James, K. A., & Mihic, S. J. (2019). Ketone body modulation of ligand-gated ion channels. Neuropharmacology, 148, 21–30.
- Mundi, M. S., Mohamed Elfadil, O., Patel, I., Patel, J., & Hurt, R. T. (2021). Ketogenic diet and cancer: Fad or fabulous? Journal of Parenteral and Enteral Nutrition, 45(S2), S26–S32.
- Fine, E. J., Segal-Isaacson, C., Feinman, R. D., Herszkopf, S., Romano, M. C., Tomuta, N., et al. (2012). Targeting insulin inhibition as a metabolic therapy in advanced cancer: A pilot safety and feasibility dietary trial in 10 patients. Nutrition, 28(10), 1028–1035.
- Schmidt, M., Pfetzer, N., Schwab, M., Strauss, I., & Kammerer, U. (2011). Effects of a ketogenic diet on the quality of life in 16 patients with advanced cancer: A pilot trial. Nutrition & Metabolism (London), 8(1), 54.
- Khodabakhshi, A., Akbari, M. E., Mirzaei, H. R., Mehrad-Majd, H., Kalamian, M., & Davoodi, S. H. (2020). Feasibility, safety, and beneficial effects of MCT-based ketogenic diet for breast cancer treatment: A randomized controlled trial study. Nutrition and Cancer, 72(4), 627–634.
- Plotti, F., Terranova, C., Luvero, D., Bartolone, M., Messina, G., Feole, L., et al. (2020). Diet and chemotherapy: The effects of fasting and ketogenic diet on cancer treatment. Chemotherapy, 65(3–4), 77–84.
- Lane, J., Brown, N. I., Williams, S., Plaisance, E. P., & Fontaine, K. R. (2021). Ketogenic diet for cancer: Critical assessment and research recommendations. Nutrients, 13(10).
- Bonetti, G., Herbst, K. L., Donato, K, Dhuli, K., Kiani, A. K., Aquilanti, B., et al. (2022). Dietary supplements for obesity. Journal of Preventive Medicine and Hygiene, 63(2 Suppl 3), E160–E168.
- Munoz, A., & Grant, W. B. (2022). Vitamin D and cancer: An historical overview of the epidemiology and mechanisms. Nutrients, 14(7).
- L’Esperance, K., Datta, G. D., Qureshi, S., & Koushik, A. (2020). Vitamin D exposure and ovarian cancer risk and prognosis. International Journal of Environmental Research and Public Health, 17(4).
- Kiani, A. K., Medori, M. C., Dhuli, K., Donato, K., Caruso, P., Fioretti, F., et al. (2022). Clinical assessment for diet prescription. Journal of Preventive Medicine and Hygiene, 63(2 Suppl 3), E102–E124.
- Wang, W., Hu, W., Xue, S., Chen, Q., Jiang, Y., Zhang, H., et al. (2021). Vitamin D and lung cancer; association, prevention, and treatment. Nutrition and Cancer, 73(11–12), 2188–2200.
- Peterson, C. T., Rodionov, D. A., Osterman, A. L., & Peterson, S. N. (2020). B vitamins and their role in immune regulation and cancer. Nutrients, 12(11).
- Hossain, S. , Beydoun, M. A., Beydoun, H. A., Chen, X., Zonderman, A. B., & Wood, R. J. (2019). Vitamin D and breast cancer: A systematic review and meta-analysis of observational studies. Clinical Nutrition ESPEN, 30, 170–185.
- Abe, E. , Miyaura, C., Sakagami, H., Takeda, M., Konno, K., Yamazaki, T., et al. (1981). Differentiation of mouse myeloid leukemia cells induced by 1 alpha,25-dihydroxyvitamin D3. Proceedings of the National Academy of Sciences of the United States of America, 78(8), 4990–4994.
- Colston, K. , Colston, M. J., & Feldman, D. (1981). 1,25-dihydroxyvitamin D3 and malignant melanoma: The presence of receptors and inhibition of cell growth in culture. Endocrinology, 108(3), 1083–1086.
- Welsh, J. (2018). Vitamin D and breast cancer: Past and present. Journal of Steroid Biochemistry and Molecular Biology, 177, 15–20.
- Narvaez, C. J. , Matthews, D., LaPorta, E., Simmons, K. M., Beaudin, S., & Welsh, J. (2014). The impact of vitamin D in breast cancer: Genomics, pathways, metabolism. Frontiers in Physiology, 5, 213.
- Feldman, D., Krishnan, A. V., Swami, S., Giovannucci, E., & Feldman, B. J. (2014). The role of vitamin D in reducing cancer risk and progression. Nature Reviews Cancer, 14(5), 342–357.
- Blasiak, J. , Pawlowska, E., Chojnacki, J., Szczepanska, J., Fila, M., & Chojnacki, C. (2020). Vitamin D in triple-negative and BRCA1-deficient breast cancer—Implications for pathogenesis and therapy. International Journal of Molecular Sciences, 21(10).
- Bauer, S. R. , Hankinson, S. E., Bertone-Johnson, E. R., & Ding, E. L. (2013). Plasma vitamin D levels, menopause, and risk of breast cancer: Dose–response meta-analysis of prospective studies. Medicine (Baltimore), 92(3), 123–131.
- Vanhevel, J. , Verlinden, L., Doms, S., Wildiers, H., & Verstuyf, A. (2022). The role of vitamin D in breast cancer risk and progression. Endocrine-Related Cancer, 29(2), R33–R55.
- Kim, H. , Lipsyc-Sharf, M., Zong, X., Wang, X., Hur, J., Song, M., et al. (2021). Total vitamin D intake and risks of early-onset colorectal cancer and precursors. Gastroenterology, 161(4), 1208–1217.
- Guo, H. , Guo, J., Xie, W., Yuan, L, & Sheng, X. (2018). The role of vitamin D in ovarian cancer: Epidemiology, molecular mechanism and prevention. Journal of Ovarian Research, 11(1), 71.
- Na, S. Y. , Kim, K. B., Lim, Y. J., & Song, H. J. (2022). Vitamin D and colorectal cancer: Current perspectives and future directions. Journal of Cancer Prevention, 27(3), 147–156.
- Wesselink, E. , Kok, D. E., Bours, M. J., De Wilt, J. H., Van Baar, H., Van Zutphen, M., et al. (2020). Vitamin D, magnesium, calcium, and their interaction in relation to colorectal cancer recurrence and all-cause mortality. American Journal of Clinical Nutrition, 111(5), 1007–1017.
- Ng, K. , Sargent, D. J., Goldberg, R. M., Meyerhardt, J. A., Green, E. M., Pitot, H. C., et al. (2011). Vitamin D status in patients with stage IV colorectal cancer: Findings from intergroup trial N9741. Journal of Clinical Oncology, 29(12), 1599.
- Yuan, C. , Sato, K., Hollis, B. W., Zhang, S., Niedzwiecki, D., Ou, F.-S., et al. (2019). Plasma 25-hydroxyvitamin D levels and survival in patients with advanced or metastatic colorectal cancer: Findings from CALGB/SWOG 80405 (Alliance). Clinical Cancer Research, 25(24), 7497–7505.
- Pilz, S. , Tomaschitz, A., Obermayer-Pietsch, B., Dobnig, H., & Pieber, T. R. (2009). Epidemiology of vitamin D insufficiency and cancer mortality. Anticancer Research, 29(9), 3699–3704.
- Kilkkinen, A. , Knekt, P., Heliövaara, M., Rissanen, H., Marniemi, J., Hakulinen, T., et al. (2008). Vitamin D status and the risk of lung cancer: A cohort study in Finland. Cancer Epidemiology, Biomarkers & Prevention, 17(11), 3274–3278.
- Maj, E., Filip-Psurska, B., Switalska, M., Kutner, A., & Wietrzyk, J. (2015). Vitamin D analogs potentiate the antitumor effect of imatinib mesylate in a human A549 lung tumor model. International Journal of Molecular Sciences, 16(11), 27191–27207.
- Verone-Boyle, A. R. , Shoemaker, S., Attwood, K., Morrison, C. D., Makowski, A. J., Battaglia, S., et al. (2016). Diet-derived 25-hydroxyvitamin D3 activates vitamin D receptor target gene expression and suppresses EGFR mutant non-small cell lung cancer growth in vitro and in vivo. Oncotarget, 7(1), 995.
- McGaffin, K. R. , & Chrysogelos, S. A. (2005). Identification and characterization of a response element in the EGFR promoter that mediates transcriptional repression by 1,25-dihydroxyvitamin D3 in breast cancer cells. Journal of Molecular Endocrinology, 35(1), 117–133.
- Qin, B. , Moorman, P. G., Alberg, A. J., Barnholtz-Sloan, J. S., Bondy, M., Cote, M. L., et al. (2016). Dairy, calcium, vitamin D and ovarian cancer risk in African-American women. British Journal of Cancer, 115(9), 1122–1130.
- Garland, C. F., Mohr, S. B., Gorham, E. D., Grant, W. B., & Garland, F. C. (2006). Role of ultraviolet B irradiance and vitamin D in prevention of ovarian cancer. American Journal of Preventive Medicine, 31(6), 512–514.
- Boscoe, F. P. , & Schymura, M. J. (2006). Solar ultraviolet-B exposure and cancer incidence and mortality in the United States, 1993–2002. BMC Cancer, 6, 264.
- Tran, B., Jordan, S. J., Lucas, R., Webb, P. M., & Neale, R. (2012). Association between ambient ultraviolet radiation and risk of epithelial ovarian cancer. Cancer Prevention Research (Philadelphia), 5(11), 1330–1336.
- Webb, P. M. , de Fazio, A., Protani, M. M., Ibiebele, T. I., Nagle, C. M., Brand, A. H., et al. (2015). Circulating 25-hydroxyvitamin D and survival in women with ovarian cancer. American Journal of Clinical Nutrition, 102(1), 109–114.
- Laczmanski, L. , Lwow, F., Osina, A., Kepska, M., Laczmanska, I., & Witkiewicz, W. (2017). Association of the vitamin D receptor FokI gene polymorphism with sex- and non-sex-associated cancers: A meta-analysis. Tumour Biology, 39(10), 1010428317727164.
- Christakos, S. , Dhawan, P., Verstuyf, A., Verlinden, L., & Carmeliet, G. (2016). Vitamin D: Metabolism, molecular mechanism of action, and pleiotropic effects. Physiological Reviews, 96(1), 365–408.
- Gnagnarella, P. , Raimondi, S., Aristarco, V., Johansson, H. A., Bellerba, F., Corso, F., et al. (2020). Vitamin D receptor polymorphisms and cancer. In Sunlight, Vit D, Skin Cancer (pp. 53–114).
- Laczmanski, L. , Lwow, F., Osina, A., Kepska, M., Laczmanska, I., & Witkiewicz, W. (2017). Association of the vitamin D receptor FokI gene polymorphism with sex- and non-sex-associated cancers: A meta-analysis. Tumor Biology, 39(10), 1010428317727164.
- Mostowska, A. , Sajdak, S., Pawlik, P., Lianeri, M., & Jagodzinski, P. P. (2016). Polymorphic variants in the vitamin D pathway genes and the risk of ovarian cancer among non-carriers of BRCA1/BRCA2 mutations. Oncology Letters, 11(2), 1181–1188.
- Carlberg, C., & Haq, A. (2018). The concept of the personal vitamin D response index. Journal of Steroid Biochemistry and Molecular Biology, 175, 12–17.
- Leoncini, E. , Nedovic, D, Panic, N., Pastorino, R., Edefonti, V., & Boccia, S. (2015). Carotenoid intake from natural sources and head and neck cancer: A systematic review and meta-analysis of epidemiological studies. Cancer Epidemiology, Biomarkers & Prevention, 24(7), 1003–1011.
- Chew, B. P., & Park, J. S. (2004). Carotenoid action on the immune response. Journal of Nutrition, 134(1), 257S–S261.
- Brewczynski, A. , Jablonska, B., Kentnowski, M., Mrowiec, S., Skladowski, K., & Rutkowski, T. (2021). The association between carotenoids and head and neck cancer risk. Nutrients, 14(1).
- Serebryakov, P. V., Pankova, V. B., Fedina, I. N., & Rushkevich, O. P. (2021). Coverage of issues of professional malignant neoplasms of the respiratory tract in modern clinical guidelines of the Ministry of Health of Russia. Vestnik Otorinolaringologii, 86(5), 48–53.
- Leoncini, E. , Edefonti, V., Hashibe, M., Parpinel, M., Cadoni, G., Ferraroni, M., et al. (2016). Carotenoid intake and head and neck cancer: A pooled analysis in the International Head and Neck Cancer Epidemiology Consortium. European Journal of Epidemiology, 31(4), 369–383.
- Rowles, J. L. , 3rd, & Erdman, J. W., Jr. (2020). Carotenoids and their role in cancer prevention. Biochimica et Biophysica Acta (BBA) – Molecular and Cell Biology of Lipids, 1865(11), 158613.
- Peng, S. J. , Li, J., Zhou, Y., Tuo, M., Qin, X. X., Yu, Q., et al. (2017). In vitro effects and mechanisms of lycopene in MCF-7 human breast cancer cells. Genetics and Molecular Research, 16(2).
- Hu, F. , Wang, Y. B., Zhang, W., Liang, J., Lin, C., Li, D., et al. (2012). Carotenoids and breast cancer risk: A meta-analysis and meta-regression. Breast Cancer Research and Treatment, 131, 239–253.
- Mignone, L. I. , Giovannucci, E., Newcomb, P. A., Titus-Ernstoff, L., Trentham-Dietz, A., Hampton, J. M., et al. (2009). Dietary carotenoids and the risk of invasive breast cancer. International Journal of Cancer, 124(12), 2929–2937.
- Tang, F. Y. , Shih, C. J., Cheng, L. H., Ho, H. J., & Chen, H. J. (2008). Lycopene inhibits growth of human colon cancer cells via suppression of the Akt signaling pathway. Molecular Nutrition & Food Research, 52(6), 646–654.
- Tang, F. Y., Pai, M. H., & Wang, X. D. (2011). Consumption of lycopene inhibits the growth and progression of colon cancer in a mouse xenograft model. Journal of Agricultural and Food Chemistry, 59(16), 9011–9021.
- Han, X. , Zhao, R., Zhang, G., Jiao, Y, Wang, Y., Wang, D., et al. (2022). Association of retinol and carotenoids content in diet and serum with risk for colorectal cancer: A meta-analysis. Frontiers in Nutrition, 9, 918777.
- Gallicchio, L. , Boyd, K., Matanoski, G., Tao, X. G., Chen, L., Lam, T. K., et al. (2008). Carotenoids and the risk of developing lung cancer: A systematic review. American Journal of Clinical Nutrition, 88(2), 372–383.
- Le Marchand, L. , Hankin, J. H., Kolonel, L. N., Beecher, G. R., Wilkens, L. R., & Zhao, L. P. (1993). Intake of specific carotenoids and lung cancer risk. Cancer Epidemiology, Biomarkers & Prevention (A Publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology), 2(3), 183–187.
- Rafi, M. M., Kanakasabai, S., Reyes, M. D., & Bright, J. J. (2013). Lycopene modulates growth and survival associated genes in prostate cancer. Journal of Nutritional Biochemistry, 24(10), 1724–1734.
- Lee, S. A. , Cai, Q., Franke, A. A., Steinwandel, M., Wu, J., Wen, W., et al. (2021). Associations of subtype and isomeric plasma carotenoids with prostate cancer risk in low-income African and European Americans. Cancer Epidemiology, Biomarkers & Prevention, 30(10), 1846–1857.
- Lu, Q.-Y. , Hung, J.-C., Heber, D., Go, V. L. W., Reuter, V. E., Cordon-Cardo, C., et al. (2001). Inverse associations between plasma lycopene and other carotenoids and prostate cancer. Cancer Epidemiology, Biomarkers & Prevention, 10(7), 749–756.
- Anderson, A. S. , Renehan, A. G., Saxton, J. M., Bell, J., Cade, J., Cross, A. J., et al. (2021). Cancer prevention through weight control—Where are we in 2020? British Journal of Cancer, 124(6), 1049–1056.
- Guthold, R. , Stevens, G. A., Riley, L. M., & Bull, F. C. (2018). Worldwide trends in insufficient physical activity from 2001 to 2016: A pooled analysis of 358 population-based surveys with 1.9 million participants. The Lancet Global Health, 6(10), e1077–e1086.
- Bull, F. C., Al-Ansari, S. S., Biddle, S., Borodulin, K., Buman, M. P., Cardon, G., et al. (2020). World Health Organization 2020 guidelines on physical activity and sedentary behaviour. British Journal of Sports Medicine, 54(24), 1451–1462.
- World Health Organization. (2022). Global status report on physical activity 2022. Geneva: World Health Organization. Licence: CC BY-NC-SA 3.0 IGO. Disponibile su: https://iris.who.int/bitstream/handle/10665/363607/9789240059153-eng.pdf.
- Cataldi, S. , Greco, G., Mauro, M., & Fischetti, F. (2021). Effect of exercise on cancer-related fatigue: A systematic review. Journal of Human Sport and Exercise, 16, 476–492.
- Cataldi, S. , Amato, A., Messina, G., Iovane, A., Greco, G., Guarini, A., Proia, P., & Fischetti, F. (2020). Effects of combined exercise on psychological and physiological variables in cancer patients: A pilot study. Acta Medica Mediterranea, 36, 1105–1113.
- Cataldi, S., Latino, F., Greco, G., & Fischetti, F. (2019). Multilateral training improves physical fitness and fatigue perception in cancer patients. Journal of Human Sport and Exercise, 14, S910–S920.
- Fischetti, F., Greco, G., Cataldi, S., Minoia, C., Loseto, G., & Guarini, A. (2019). Effects of physical exercise intervention on psychological and physical fitness in lymphoma patients. Medicina, 55(7), 379.
- Pepe, I., Petrelli, A., Fischetti, F., Minoia, C., Morsanuto, S., Talaba, L., Cataldi, S., & Greco, G. (2025). Nonregular physical activity and handgrip strength as indicators of fatigue and psychological distress in cancer survivors. Current Oncology, 32(5), 289.
- Poli, L. , Petrelli, A., Fischetti, F., Morsanuto, S., Talaba, L., Cataldi, S., & Greco, G. (2025). The effects of multicomponent training on clinical, functional, and psychological outcomes in cardiovascular disease: A narrative review. Medicina, 61(5), 822.
- Poli, L. , Mazić, S., Ciccone, M. M., Cataldi, S., Fischetti, F., & Greco, G. (2025). A 10-week multicomponent outdoor exercise program improves hemodynamic parameters and physical fitness in cardiovascular disease adult and elderly patients. Sport Sciences for Health, 21, 239–249.
- Poli, L. , Greco, G., Cataldi, S., Ciccone, M. M., De Giosa, A., & Fischetti, F. (2024). Multicomponent versus aerobic exercise intervention: Effects on hemodynamic, physical fitness and quality of life in adult and elderly cardiovascular disease patients: A randomized controlled study. Heliyon, 10(16), e36200.
- Arnold, M. , Pandeya, N., Byrnes, G., Renehan, A. G., Stevens, G. A., Ezzati, M., et al. (2015). Global burden of cancer attributable to high body-mass index in 2012: A population-based study. The Lancet Oncology, 16(1), 36–46.
- World Cancer Research Fund/American Institute for Cancer Research. (2018). Diet, nutrition, physical activity and cancer: A global perspective. Continuous Update Project Expert Report 2018. Disponibile su: https://www.wcrf.org/wp-content/uploads/2024/11/Summary-of-Third-Expert-Report-2018.pdf.
- McTiernan, A. (2008). Mechanisms linking physical activity with cancer. Nature Reviews Cancer, 8(3), 205–211.
- Friedenreich, C. M., Neilson, H. K., & Lynch, B. M. (2010). State of the epidemiological evidence on physical activity and cancer prevention. European Journal of Cancer, 46(14), 2593–2604.
- Hojman, P. (2017). Exercise protects from cancer through regulation of immune function and inflammation. Biochemical Society Transactions, 45(4), 905–911.
- Oh, H. , Arem, H., Matthews, C. E., Wentzensen, N., Reding, K. W., Brinton, L. A., et al. (2017). Sitting, physical activity, and serum estrogen metabolism in postmenopausal women: The Women’s Health Initiative Observational Study. British Journal of Cancer, 117(7), 1070–1078.
- de Sousa, C. V. , Sales, M. M., Rosa, T. S., Lewis, J. E., de Andrade, R. V., & Simões, H. G. (2017). The antioxidant effect of exercise: A systematic review and meta-analysis. Sports Medicine, 47, 277–293.
- Gunter, M. J. , Xie, X., Xue, X., Kabat, G. C., Rohan, T. E., Wassertheil-Smoller, S., et al. (2015). Breast cancer risk in metabolically healthy but overweight postmenopausal women. Cancer Research, 75(2), 270–274.
- Clemente-Suárez, V. J. , Martín-Rodríguez, A., Redondo-Flórez, L., Ruisoto, P., Navarro-Jiménez, E., Ramos-Campo, D. J., et al. (2023). Metabolic health, mitochondrial fitness, physical activity, and cancer. Cancers, 15(3), 814.
- Ferioli, M., Zauli, G., Maiorano, P., Milani, D., Mirandola, P., & Neri, L. M. (2019). Role of physical exercise in the regulation of epigenetic mechanisms in inflammation, cancer, neurodegenerative diseases, and aging process. Journal of Cellular Physiology, 234(9), 14852–14864.
- Nomikos, N. N. , Nikolaidis, P. T., Sousa, C. V., Papalois, A. E., Rosemann, T., & Knechtle, B. (2018). Exercise, telomeres, and cancer: “The exercise–telomere hypothesis.” Frontiers in Physiology, 9, 1798.
- Papadimitriou, N. , Dimou, N., Tsilidis, K. K., Banbury, B., Martin, R. M., Lewis, S. J., et al. (2020). Physical activity and risks of breast and colorectal cancer: A Mendelian randomisation analysis. Nature Communications, 11(1), 597.
- Mazzilli, K. M. , Matthews, C. E., Salerno, E. A., & Moore, S. C. (2019). Weight training and risk of 10 common types of cancer. Medicine & Science in Sports & Exercise, 51(9), 1845.
- Rezende, L. F. , Lee, D. H., Keum, N., Wu, K, Eluf-Neto, J., Tabung, F. K., et al. (2020). Resistance training and total and site-specific cancer risk: A prospective cohort study of 33,787 US men. British Journal of Cancer, 123(4), 666–672.
- Bird, S. R. , & Hawley, J. A. (2017). Update on the effects of physical activity on insulin sensitivity in humans. BMJ Open Sport & Exercise Medicine, 2(1), e000143.
- Inoue, M. , & Tsugane, S. (2012). Insulin resistance and cancer: Epidemiological evidence. Endocrine-Related Cancer, 19(5), F1–F8.
- Wang, Q., & Zhou, W. (2021). Roles and molecular mechanisms of physical exercise in cancer prevention and treatment. Journal of Sport and Health Science, 10(2), 201–210.
- Magné, N. , Melis, A., Chargari, C., Castadot, P., Guichard, J.-B., & Barani, D., et al. (2011). Recommendations for a lifestyle which could prevent breast cancer and its relapse: Physical activity and dietetic aspects. Critical Reviews in Oncology/Hematology, 80(3), 450–459.
- Desnoyers, A. , Riesco, E., Fülöp, T., & Pavic, M. (2016). Physical activity and cancer: Update and literature review. La Revue de médecine interne, 37(6), 399–405.
- Gonçalves, A. K. , Florêncio, G. L. D., de Atayde Silva, M. J. M., Cobucci, R. N., Giraldo, P. C., & Cote, N. M. (2014). Effects of physical activity on breast cancer prevention: A systematic review. Journal of Physical Activity and Health, 11(2), 445–454.
- Reis, A. D. , Garcia, J. B. S., Diniz, R. R., Silva-Filho, A. C., Dias, C. J., Leite, R. D., & Mostarda, C. (2017). Effect of exercise training and detraining in autonomic modulation and cardiorespiratory fitness in breast cancer survivors. Journal of Sports Medicine and Physical Fitness, 57(7–8), 1062–1068.
- Liu, L. , Shi, Y., Li, T., Qin, Q., Yin, J., Pang, S., et al. (2016). Leisure time physical activity and cancer risk: Evaluation of the WHO’s recommendation based on 126 high-quality epidemiological studies. British Journal of Sports Medicine, 50(6), 372–378.
- Psaltopoulou, T. , Ntanasis-Stathopoulos, I., Tzanninis, I.-G., Kantzanou, M., Georgiadou, D., & Sergentanis, T. N. (2016). Physical activity and gastric cancer risk: A systematic review and meta-analysis. Clinical Journal of Sport Medicine, 26(6), 445–464.
- Schmid, D. , Behrens, G., Keimling, M., Jochem, C., Ricci, C., & Leitzmann, M. (2015). A systematic review and meta-analysis of physical activity and endometrial cancer risk. European Journal of Epidemiology, 30, 397–412.
- Behrens, G. , Jochem, C., Keimling, M., Ricci, C., Schmid, D., & Leitzmann, M. F. (2014). The association between physical activity and gastroesophageal cancer: Systematic review and meta-analysis. European Journal of Epidemiology, 29, 151–170.
- Moore, S. C. , Lee, I.-M., Weiderpass, E., Campbell, P. T., Sampson, J. N., Kitahara, C. M., et al. (2016). Association of leisure-time physical activity with risk of 26 types of cancer in 1.44 million adults. JAMA Internal Medicine, 176(6), 816–825.
- Schmid, D. , Ricci, C., Behrens, G., & Leitzmann, M. F. (2016). Does smoking influence the physical activity and lung cancer relation? A systematic review and meta-analysis. European Journal of Epidemiology, 31, 1173–1190.
- Christensen, J. F., Simonsen, C., & Hojman, P. (2011). Exercise training in cancer control and treatment. Comprehensive Physiology, 9(1), 165–205.
- Patel, A. V. , Friedenreich, C. M., Moore, S. C., Hayes, S. C., Silver, J. K., Campbell, K. L., et al. (2019). American College of Sports Medicine roundtable report on physical activity, sedentary behavior, and cancer prevention and control. Medicine & Science in Sports & Exercise, 51(11), 2391.
- Koelwyn, G. J. , Quail, D. F., Zhang, X., White, R. M., & Jones, L. W. (2017). Exercise-dependent regulation of the tumour microenvironment. Nature Reviews Cancer, 17(10), 620–632.
- McCullough, D. J. , Stabley, J. N., Siemann, D. W., & Behnke, B. J. (2014). Modulation of blood flow, hypoxia, and vascular function in orthotopic prostate tumors during exercise. JNCI: Journal of the National Cancer Institute, 106(4).
- Bigley, A. B. , Spielmann, G., LaVoy, E. C., & Simpson, R. J. (2013). Can exercise-related improvements in immunity influence cancer prevention and prognosis in the elderly? Maturitas, 76(1), 51–56.
- World Health Organization. (2020). Obesity and overweight: Fact sheet. Geneva: World Health Organization. Disponibile su: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight.
- Simopoulos, A. P. (1987). Obesity and carcinogenesis: Historical perspective. American Journal of Clinical Nutrition, 45(1), 271–276.
- Hidayat, K. , Du, X., & Shi, B. M. (2018). Body fatness at a young age and risks of eight types of cancer: Systematic review and meta-analysis of observational studies. Obesity Reviews, 19(10), 1385–1394.
- World Cancer Research Fund. (2023). Diet, nutrition, physical activity and cancer: A global perspective. Body fatness and weight gain and the risk of cancer. Available at: https://www.wcrf.org/wp-content/uploads/2024/10/Body-fatness-and-weight-gain_0.pdf.
- Ellulu, M. S. , Patimah, I., Khaza’ai, H., Rahmat, A., & Abed, Y. (2017). Obesity and inflammation: The linking mechanism and the complications. Archives of Medical Science, 13(4), 851–863.
- Harris, B. H. L. , Macaulay, V. M., Harris, D. A., Klenerman, P., Karpe, F., Lord, S. R., et al. (2022). Obesity: A perfect storm for carcinogenesis. Cancer and Metastasis Reviews, 41(3), 491–515.
- Li, Y. , & Hecht, S. S. (2022). Carcinogenic components of tobacco and tobacco smoke: A 2022 update. Food and Chemical Toxicology, 165, 113179.
- Larsson, S. C. , Carter, P., Kar, S., Vithayathil, M., Mason, A. M., Michaelsson, K., et al. (2020). Smoking, alcohol consumption, and cancer: A Mendelian randomisation study in UK Biobank and international genetic consortia participants. PLOS Medicine, 17(7), e1003178.
- Secretan, B. , Straif, K., Baan, R., Grosse, Y., El Ghissassi, F., Bouvard, V., et al. (2009). A review of human carcinogens—Part E: Tobacco, areca nut, alcohol, coal smoke, and salted fish. The Lancet Oncology, 10(11), 1033–1034.
- Wei, P.-L. , Chang, Y.-J., Ho, Y.-S., Lee, C.-H., Yang, Y.-Y., An, J., et al. (2009). Tobacco-specific carcinogen enhances colon cancer cell migration through α7-nicotinic acetylcholine receptor. Annals of Surgery, 249(6), 978–985.
- Hecht, S. S. (2002). Tobacco smoke carcinogens and breast cancer. Environmental and Molecular Mutagenesis, 39(2–3), 119–126.
- Jethwa, A. R. , & Khariwala, S. S. (2017). Tobacco-related carcinogenesis in head and neck cancer. Cancer and Metastasis Reviews, 36, 411–423.
- Caliri, A. W. , Tommasi, S., & Besaratinia, A. (2021). Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. Mutation Research/Reviews in Mutation Research, 787, 108365.
- International Agency for Research on Cancer (IARC). (2013). Non-ionizing radiation, Part 2: Radiofrequency electromagnetic fields. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, 102(Pt 2), 1–460. https://www.iarc.who.int/wp-content/uploads/2018/07/Monographs102.pdf.
- Stang, A., Anastassiou, G., Ahrens, W., Bromen, K., Bornfeld, N., & Jöckel, K. H. (2001). The possible role of radiofrequency radiation in the development of uveal melanoma. Epidemiology, 12(1), 7–12.
- Karipidis, K. K. , Benke, G., Sim, M. R., Kauppinen, T., & Giles, G. (2007). Occupational exposure to ionizing and non-ionizing radiation and risk of glioma. Occupational Medicine (London), 57(7), 518–524.
- Gupta, S. , Sharma, R. S., & Singh, R. (2022). Non-ionizing radiation as possible carcinogen. International Journal of Environmental Health Research, 32(4), 916–940.
- Bauer, A. , Diepgen, T. L., & Schmitt, J. (2011). Is occupational solar ultraviolet irradiation a relevant risk factor for basal cell carcinoma? A systematic review and meta-analysis of the epidemiological literature. British Journal of Dermatology, 165(3), 612–625.
- National Cancer Institute. (2023). Complementary and alternative medicine. https://www.cancer.gov/about-cancer/treatment/cam.
- Carlson, L. E. , Zelinski, E., Toivonen, K., Flynn, M., Qureshi, M., Piedalue, K.-A., & Grant, R. (2017). Mind–body therapies in cancer: What is the latest evidence? Current Oncology Reports, 19, 67.
- Cramer, H. , Lauche, R., Langhorst, J., Dobos, G., & Ward, L. (2022). Mind–body practices for cancer-related symptoms management: An overview of systematic reviews. Supportive Care in Cancer, 30, 12345–12356.
- Deleemans, J. M. , Mather, H., Spiropoulos, A., Toivonen, K., Baydoun, M., & Carlson, L. E. (2023). Recent progress in mind–body therapies in cancer care. Current Oncology Reports, 25, 293–307.
- Horneber, M. , Bueschel, G., Dennert, G., Less, D., Ritter, E., & Zwahlen, M. (2012). How many cancer patients use complementary and alternative medicine: A systematic review and meta-analysis. Integrative Cancer Therapies, 11(3), 187–203.
- Frenkel, M. , Slater, R., Sapire, K., & Sierpina, V. (2018). Complementary and integrative medicine in lung cancer: Questions and challenges. Journal of Alternative and Complementary Medicine, 24(9–10), 862–871.
- David, A. , Hausner, D., & Frenkel, M. (2021). Cancer-related fatigue—Is there a role for complementary and integrative medicine? Current Oncology Reports, 23(12), 145.
Table 1.
Fasting regimens: definitions and practical notes.
Table 1.
Fasting regimens: definitions and practical notes.
| Fasting regimen |
Definition |
Reference |
| Fasting |
Complete caloric abstention or exclusion of selected foods for a defined interval |
[14] |
| Intermittent fasting |
Alternation of eating and energy-restriction/water-only intervals on 1–3 days per week |
[14,15] |
| Periodic fasting |
Severe energy restriction or water-only phases of ~48 h (up to a week in some protocols) |
[18] |
| Short-term fasting |
Time-limited abstention (≈12–72 h), e.g., alternate-day patterns |
[19] |
Fasting mimicking diet |
Brief, hypocaloric, low-protein, plant-forward cycles aligned to treatment sessions |
[17,20] |
|
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).