Skip to main content
Intended for healthcare professionals

Abstract

The complex cellular mechanisms and inter-related pathways of cancer proliferation, evasion, and metastasis remain an emerging field of research. Over the last several decades, nutritional research has prominent role in identifying emerging adjuvant therapies in our fight against cancer. Nutritional and dietary interventions are being explored to improve the morbidity and mortality for cancer patients worldwide. In this review, we examine several dietary interventions and their proposed mechanisms against cancer as well as identifying limitations in the currently available literature. This review provides a comprehensive review of the cancer metabolism, dietary interventions used during cancer treatment, anti metabolic drugs, and their impact on nutritional deficiencies along with a critical review of the following diets: caloric restriction, intermittent fasting, ketogenic diet, Mediterranean diet, Japanese diet, and vegan diet.

Introduction to cancer and nutrition

Cancer is the leading cause of death worldwide. In 2018 alone, over 1.7 million people were diagnosed with cancer and over 600,000 deaths have resulted from this disease.1 From 1999 to 2015, the overall incidence of cancer decreased by 2.2% in men and remained stable in women, yet the mortality rate for both has only decreased slightly to 1.8% and 1.4% respectively.2 These modest improvements in outcomes could be attributed to improvements in cancer therapies and detection; however, it is tempting to speculate that greater improvements could be achieved if the therapeutic effects of diet on cancer are considered.
The last decade seems to have witnessed a surge of interest in the role of diet and its effects on cancer metabolism. This interest is at least partly ignited by the rapid increase in the prevalence of obesity in the United States. Between 2015 and 2016, the National Health and Nutrition Examination Survey demonstrated 39.8% of adults and 18.5% of youth were obese.3 A prospective study of over 900,000 adults in the United States showed a significant proportional increase between obesity and mortality risk from multiple cancers, including of the esophagus, colon and rectum, liver, gallbladder, pancreas, kidney, prostate, breast, uterus, cervix, and ovary.4 With the high prevalence of obesity in the United States, many investigators have sought to investigate the effects of excessive nutritional intake on the outcomes of cancer patients.
In general, cancer therapies target the hypermetabolic state of cancer cells to primarily destroying rapidly dividing tumor cells. However, rapidly dividing normal cells are also affected by anti-neoplastic therapies. As our understanding of tumor proliferation and apoptosis within individual cancers expands, investigators have been making significant strides to create targeted immunotherapies that will eliminate the bystander effect seen with traditional chemotherapy such as neutropenia, mucositis, renal failure, and cardiac dysfunction. The first example of immunotherapy was developed by Milstein and Kohler in 1970. Then, in 1997, rituximab, a monoclonal CD20 antibody, became available as a targeted treatment for non-Hodgkin’s lymphoma and has since become a widely utilized therapeutic option due to its short-lived side effects that can be managed medically for 3–6 months.5 While targeted therapy remains the goal of oncologists and patients alike, investigators are interested in how lifestyle modifications, such as dietary pattern, can impact tumor physiology and clinical outcomes. The advent of personalized medicine, precision cancer medicine, or “theranostics” further supports a departure from broad algorithmic interventions and instead focuses on the matrix of a specific patient’s oncogenomic, pharmacogenomic, epigenetic, historical, and lifestyle characteristics. Nutritional status and diet are increasingly seen in this conception as significant matrix variables in predicting risk for disease, and response to intervention.
The role of diet in cancer metabolism is certainly an area of popular interest. The American Institute for Cancer Research and the World Cancer Research Fund estimates that 30%–40% of cancers can be prevented by healthy dietary regimens, improved physical activity, and maintenance of appropriate body weight.6 While “prevention” is likely an overstatement, reduction of risk does appear to be supported by the evidence. It has been shown in epidemiological studies of breast cancer, prostate cancer, and colon cancer that migration to different countries influences overall risk of the development of these cancers, leading to hypotheses that changes in dietary habits may alter cancer risk. Kolonel et al.7 demonstrated in 1980 that first-generation Japanese women had a threefold increase in breast cancer compared to Japanese woman living in Japan. It has been postulated that this difference may be partially explained by the switch from a primarily plant-based diet to a high-fat, high-sugar diet. Similarly, the mortality rate from stomach cancer in European migrants to Australia decreased in parallel with the length of time migrants stayed in Australia, but risk of colorectal cancer increased proportionally to the length of stay.8 Prostate cancer studies have shown similar results where certain lifestyle factors are associated with the progression of malignancy.9 These epidemiologic studies suggest that changes in lifestyle and dietary influences play a role in determining the risk of various cancers.
At the time of cancer diagnosis, many patients inquire about how lifestyle modifications can slow their tumor progression. Calorie restriction (CR) is a well-established dietary intervention for preventing cancer and increasing lifespan in experimental animal models.10 In a prostate cancer xenograft mouse model, CR alone was shown to decrease final tumor weight, plasma insulin, and insulin-like growth factor (IGF)-1 levels and increase apoptosis, overall suggesting that decreasing caloric intake may reduce tumor proliferation.11 In human studies, a 15% caloric reduction over 4 years demonstrated a sustained reduction in plasma growth factors and hormones, which have been associated with increased risk of cancer.10,12 The CALERIE study revealed that it was feasible for patients to comply with a 25% CR intervention over a 2-year period and later showed a reduction in markers of oxidative stress, which may inhibit cancer proliferation.13,14 Another diet of interest is the vegan diet, which has been shown to decrease tumor markers and inhibit tumor cell growth in prostate cancer studies.15 Optimization of dietary regimens has thus gained attention as a lifestyle modification that individuals may undertake to alter their risk of the development of malignancies.
Certain malignancies have well-documented associations with lifestyle habits, such as lung cancer with smoking and mesothelioma with asbestos exposure.16,17 Although primary prevention of tumorigenesis is a shared goal by patients and oncologists alike, the majority of cancers do not have clear risk factors, thus posing challenges in educating patients about the proper exposures to avoid. With regard to lifestyle modifications and cancer risk, the general dietary guidelines published by the American Cancer Society advocate for the avoidance of excess weight gain, consumption of a primarily vegetarian-based diet, and limited intake of alcohol, red meat, and processed foods.18,19 Here, we present a review of the expansive literature on the interactions between nutrition, diet, and the course of malignant neoplasms.

Cancer metabolism

Cachexia is an integral component of involuntary weight loss in the setting of disease-associated wasting. It is further and still broadly defined as an inflammatory-associated wasting of protein, particularly skeletal muscle, and loss of energy stores. Cytokines and hormones, such as leptin, insulin, several interleukins, and growth factors, elicited in disease and as principal mediators of inflammation may in effect tilt physiology toward catabolic breakdown of tissue presumably in service of mobilizing critical nutrients for central nervous system and aspects of immune function.20
Cachexia has been shown to be associated with cancer-related mortality; however, no consistently effective therapies have been developed to prevent or hamper its progression.20 Even for patients who are able to eat—appetite suppression or anorexia is a common cachexia symptom—and efforts to improved nutrition often offer little respite.21 It is interesting to speculate that underreporting of cachexia as a “contributing cause of death” has perennially affected the primacy placed on research and funding of this enormously pervasive process.
As a special case, cancer-induced cachexia is described as a multifactorial metabolic disorder seen in 50% of cancer patients.22 The exact mechanisms remain unclear, but it is thought to be typified by an increase in energy expenditure, hepatic gluconeogenesis, fat lipolysis, and skeletal muscle proteolysis leading to progressive weight loss throughout therapy.23,24 Further complicating this phenomenon, as noted previously, is that this auto-catabolic process is not reversed by total parental nutrition or supplements.25 It is important to note that the relationship between tumor phenotype to host genotype likely results in heterogeneity in the degree of cachexia between individuals with the same malignancy.
Even prior to diagnosis, most cancer patients will experience a significant degree of catabolism leading to both muscle and adipose depletion, in contrast to the predominant wasting of adipose tissue in anorexic patients.26 This difference may be due to the change in resting energy expenditure (REE), which is defined as the amount of energy expended by a person at rest. A recent meta-analysis of 27 studies comparing 1453 cancer patients to 1145 control patients shows that, on average, the REE of cancer patients was 9.66% higher than that of controls.27 It is important to note that different malignancies are known to have varying degree of metabolism, and one study found marked REE variation among different cancer groups (p < 0.001).27 For example, Burkitt’s lymphoma has a doubling time of 24 h,28 while Hodgkin’s lymphoma has a doubling time of 29 days.29 Therefore, the specific type of cancer may have large effects on the overall metabolism within the patient.
It was first described in 1927 by Otto Warburg et al.30 that tumors cells demonstrate an aberrant metabolic pathway that contributes to excessive catabolism. In normal tissues via oxidative phosphorylation, one molecule of glucose is able to produce 36 adenosine triphosphate (ATP) molecules, which are the intracellular energy used to drive cellular processing. Cells also have an alternative pathway known as glycolysis, which utilizes one molecule of glucose to produce just two ATP molecules. In a phenomenon known as the Warburg effect, tumor cells selectively upregulate key regulators of glycolysis, such as hypoxia-induced factor-1α (HIF-1α).31 HIF-1α drives three main processes: (1) increased expression of glucose transporter-1 (GLUT-1) leading to increased glucose availability for glycolysis;32 (2) increased pyruvate dehydrogenase kinase production leading to increased conversion of lactate to pyruvate and decreased production of acetyl CoA necessary for the Krebs cycle (produces one ATP and three nicotinamide adenine dinucleotide (NADH));33 and (3) increased lactate dehydrogenase production leading to increased conversion of lactate to pyruvate, which serves as the primary substrate for gluconeogenesis in the liver.31 Increased lactate acid production via glycolysis is also released systemically, where it undergoes gluconeogenesis in an ATP-consumptive process known as the futile lactate–glucose shunt, or Cori cycle.34 Overall, these processes promote an overall energy deficient state, leading to lipolysis and proteolysis and eventually cachexia. A recent in vivo study with human pancreatic cancer cells in athymic mice reveals that the Warburg effect was present in the complex system of a live animal, thus further supporting these proposed mechanisms of cancer-induced cachexia.24 After 90 years since the Warburg effect was proposed, it remains unclear if the Warburg effect acts independently or if physiologic interactions in response to the Warburg effect drive cancer-induced cachexia.
Systemic inflammation that occurs during tumor proliferation is thought to contribute to cachexia.35 Elevation in pro-inflammatory markers, such as C-reactive protein and fibrinogen, has been positively correlated with the degree of muscle wasting in cancer and chronic disease.36,37 Cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1(IL-1), and interleukin-6 (IL-6), contribute to persistent inflammation and increase rates of gluconeogenesis, lipolysis, and proteolysis.38 TNF-α also stimulates the expression of uncoupling proteins 2 and 3 (UCP2/3) in cachectic skeletal muscle, due to UCP’s mediated proton leakage, and decreases the coupling of respiration to ADP phosphorylation, thereby generating heat instead of ATP from brown fat.37,39 In fact, IL-6 levels have been found to be elevated in cachexic patients when compared with patients who maintained their weight during therapy.37 These mechanisms have been replicated in animal studies, where IL-6 blockade in mice leads to attenuation of protein degradation40 and TNF-α blockade decreases catabolism in rat models.41,42
The role of neuropeptides in inducing anorexia during a catabolic state is emerging as a major contributing factor to cancer-induced cachexia. There appears to be an imbalance between appetite-stimulating molecules, such as neuropeptide P (NYP), melanin-concentrating hormone, orexins, endogenous opioids, and cannabinoids compared with appetite suppressant molecules such as serotonin, peptide YY, cholecystokinin, leptin, and insulin.43 The role of neuropeptide Y is of special consideration as it is produced primarily in the hypothalamus and has a robust feeding stimulatory effect. Overstimulation with NYP leads to obesity in rats, in addition to decreased energy expenditure and reduced brown fat thermogenesis.43 However, NYP injection into rats with sarcomas showed a reduction in feeding behavior, and a separate study revealed that rats with malignancies demonstrated a reduced release of NYP, suggesting that tumors may induce neurohormonal changes that decrease feeding activity and compound weight loss in cancer patients.4446
Another hormone of interest is leptin, a protein primarily produced in white and brown adipocytes that is responsible for peripheral signaling to reduce appetite and increase energy expenditure.47 While decreased food intake normally suppresses leptin production, anorexia has been associated with increased leptin levels in adipose tissue and plasma.48,49 Multiple authors have reported low or undetectable circulating leptin levels in cancer patients, suggesting a degree of leptin dysregulation.50 Elucidating the mechanisms behind neurohormonal dysregulation of leptin may provide insight into further therapeutic options in the future.

Anti-metabolic drugs and their impact on nutritional deficiencies

Cancer chemotherapy regimens target rapidly dividing cells by either inhibiting DNA or protein synthesis or restricting essential micronutrients, thus leading to cellular death. Further, studies show that between 30% and 90% of patients have inadequate dietary regimens.51,52 Chemotherapy, along with poor nutritional intake during treatment, is a great concern for patients and physicians. Chemotherapies that affect the nutritional status of patients include anti-metabolites, such folate, purine, and pyrimidine analogs, in addition to platinum-based drugs. Due to the broad mechanism of action of these drugs, many organ systems are adversely affected.
Methotrexate is an anti-metabolite folate analog that is used in standard therapy protocols for leukemia, lymphoma, and osteosarcoma.5355 Methotrexate leads to cellular death by acting as a dihydrofolate inhibitor, which is responsible for the conversion of dihydrofolate to tetrahydrofolate. Inhibition of tetrahydrofolate formation reduces the availability of one-carbon fragments necessary for the production of purines and the conversion of deoxyuridylate to thymidylate for DNA synthesis and cell reproduction. Although rapidly dividing malignant cells are most affected by inhibition of DNA synthesis, other rapidly proliferating cells are affected as well, such as hematopoietic and epithelial cells, thus leading to myelosuppression and mucositis.56 Due to these side effects, a leucovorin “rescue” is performed 24 h after methotrexate is administered. Leucovorin is a derivative of tetrahydrofolic acid, which does not require dihydrofolate reductase activity and thus “rescues” the healthy cells by allowing for de novo DNA synthesis to resume.57 Although methotrexate is associated with favorable mortality outcomes in combination with other chemotherapies, it is difficult to predict one’s toxicity after treatment. Therefore, dose reductions are often necessary due to severe side effects from inhibition of DNA synthesis and cellular proliferation.
5-Fluorouracil (5-FU) is a competitive inhibitor of thymidylate synthase, which blocks thymidine synthesis and inhibits DNA and RNA replication.58 5-FU is widely used in colorectal, breast, and head and neck cancers.58 Interestingly, increased intracellular folate levels increase thymidylate synthase levels, thereby enhancing the inhibitory effects of 5-FU. This interaction between metabolites suggests that optimal nutrition can improve chemotherapy effects in specific cancer groups.59,60 Gemcitabine has a similar mechanism of action with 5-FU; however, it is a prodrug that, when phosphorylated, interrupts DNA synthesis and inhibits DNA repair, leading to cellular death.56 In pancreatic cancer, it has been shown that intravenous omega-3 fatty acids, along with gemcitabine, improve quality of life scores with regard to chemotherapy-induced side effects.61 The relationship between targeted supplementation and chemotherapy optimization suggests that optimized nutrition and reduction of chemotherapy effects could be dependent on specific treatment regimens.
Platinum-based chemotherapy drugs such as cisplatin, carboplatin, and oxaliplatin are used in the treatment of cancers such as testicular, ovarian, and lung cancer, in addition to osteosarcoma and neuroblastoma.62 After the platinum-based drug is incorporated into the cell using copper transporters, it binds at the N7 position of guanine, causing cross-linking between adjacent guanine. This leads to failed DNA repair mechanisms and eventually cellular apoptosis.63 Two studies in patients with solid tumors showed that selenium supplementation, when given during cisplatin therapy, reduced myelosuppression and nephrotoxicity, suggesting that optimal levels of selenium could aid in the toxicity profile related to platinum-based therapies. It was not reported whether mortality or relapse rates were affected by this type of supplementation, and thus, more studies are warranted to elucidate these data (Table 1).64,65
Table 1. Chemotherapy and nutritional deficiencies.
Chemotherapy Mechanism of chemotherapy Type of malignancy Nutritional deficiencies Nutritional supplementation
Methotrexate Antifolate metabolite Leukemia
Lymphoma
Osteosarcoma
Folic acid Folic acid
5-Fluorouracil
Gemcitabine
Competitive inhibitor of thymidylate synthase Colorectal
Breast
Head and neck
Pancreatic
Intravenous omega-3 (with gemcitabine; single-study evidence)
Cisplatin
Carboplatin
Oxaliplatin
Cross-linking between adjacent guanine Testicular
Ovarian
Lung
Osteosarcoma
Neuroblastoma
  Selenium (with cisplatin)

Dietary approaches

Western diet

Environmental and lifestyle factors, including diet, are hypothesized to influence risk of malignancy.66 In the United States, the vast majority of Americans consume a Western diet, which consists of a high intake of fats, processed meat, dairy, and carbohydrates and low intakes of fiber.67 This composition has been associated with broadly negative impacts on glycemic load, fatty acid composition, micronutrient and macronutrient composition, sodium intake, and fiber content, all of which have been suggested to contribute to tumorigenesis.68 Since the diet is a modifiable risk factor in many of the diseases we see today, the interaction between how diet may modify cancer risk is an important area of emerging research.
It is postulated that the shift to the Western diet among many countries may be a contributing factor to the apparent increase in chronic diseases, including hypertension, coronary artery disease, hyperlipidemia, and osteoporosis.68 Over the last few decades, the Western diet has been epidemiologically associated with an increased incidence of many cancers, including prostate cancer,67 breast cancer,69 colorectal cancer,70 among many others.71 Specifically, one study concluded that a Western diet showed an overall odds ratio of colon cancer risk of 1.88 when compared with individual who consumed higher intakes fruits, vegetables, and grains combined with a lower intake of red/processed meats.72 Another study found that those who followed a Western diet had an increased risk of prostate cancer, with an odds ratio of 1.34 compared with those who consumed healthier diets of fish, poultry, and whole grains.67 These large cohort studies suggest that dietary pattern has a substantial impact on risk of developing various malignancies. Despite large samples, hypotheses should be tempered by the clinical reality that there are many antecedent and intervening variables that may confound conclusions.
Nevertheless, specific dietary regimens have gained increased interest as potentially effective adjuvant interventions to traditional cancer therapies. Here, we will discuss some of the more prominent dietary approaches, the evidence behind the respective elements, and the principal limitations.

CR

Introduction

CR is defined as a reduction in dietary intake by approximately 30%, to improve metabolic profile, without causing malnutrition.73,74 It has been postulated to be a potentially powerful dietary intervention that could be integrated into a patient’s cancer therapy due to results showing an increased life span and decreased risk of chronic and age-related diseases, including cancer, type II diabetes, and cardiovascular disease.75 The mechanism behind CR stems from the concept that excessive adiposity from obesity can increase levels of oxidative stress, inflammation, and growth factors, which all may have promotive effects on tumorgenesis.76 Thus, the negative net energy resulting from CR may lead to decreased inflammation and growth factor production, thus suppressing the neoplastic activity of the tumor cells. As such, the utility of CR is gaining momentum as a powerful option to improve the efficacy and augment the response of current anti-neoplastic therapies.

Proposed mechanism

CR has been shown to exert its effects on tumor progression through altered levels of various growth factors. The most well-studied growth factor is IGF-1, which upregulated by growth hormone and acts with other anabolic hormones to upregulate energy metabolism and proliferation.77 In murine models, CR has been shown to reduce IGF-1 by about 30%–40% in mice.78 Conversely, mice with growth hormone deficiency are shown to have low IGF-1 levels associated with delayed incidence of lung adenocarcinoma.79 Interestingly, in a cohort of Laron dwarfs, who are individuals with an autosomal recessive mutation in growth hormone receptor and are also IGF-1 deficient, none of the patients with dwarfism had a history of cancer while 9%–24% of family members developed a wide variety of malignancies.80 In stark contrast, patients with acromegaly, who have chronically elevated IGF-1 levels, have been shown to have an increased risk of colon cancer.81 Furthering this theory is that infusion of growth hormone or IGF-1 in mice reverses the protective effects of CR against mononuclear cell leukemia development and progression.10,82 IGF-1 has a role in the proliferation profile of several malginancies, and CR may be a preventive and/or complementary approach for reducing a known tumor-stimulating growth factor.
IGF-1 is primarily involved in the regulation of two major pathways that coordinate cellular proliferation and growth: Ras/MAPK (mitogen-activated protein kinase) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT; Figure 1). Activation of Ras/MAPK promotes transcription factors involved in cellular growth, while activation of PI3K/AKT decreases apoptosis.83 CR has been shown to decrease serum levels of IGF-1, thereby causing downregulation of both the Ras/MAPK and PI3K/AKT pathways, which inhibit both tumor growth and development.73 In addition, CR has been shown to improve insulin sensitivity via downregulation of IGF-1 levels and improve glucose tolerance.84 Thus, CR may promote insulin sensitivity leading to decreased serum blood glucose and decreased IGF-1 levels, which may mitigate one of the mechanisms driving cancer cell proliferation.
Figure 1. Dietary effect on proliferation pathways associated with cancer. Dietary interventions have been shown to affect the complex interaction between proliferation and apoptotic pathways. IGF-1, a growth factor that acts as an anabolic hormone to upregulate energy metabolism and proliferation, has been thoroughly studied in numerous cancer models. Activation of Ras/MAPK promotes transcription factors involved in cellular growth. Conversely, the PI3K/AKT pathway decreases apoptosis via mTOR inhibition of autophagy. Caloric restriction, intermittent fasting, and the ketogenic diet have been shown to decrease IGF-1 levels, thereby inhibiting both Ras/MAPK and PI3K/AKT proliferation pathways. Caloric restriction and intermittent fasting have also been shown to directly inhibit the mTOR pathway, thereby relieving the inhibition of the autophagy pathway to result in apoptosis. Conversely, intermittent fasting has been shown to activate AMPK, which inhibits mTOR from halting autophagy in rapidly growing cancer cells. Finally, the ketogenic diet inhibits the mTOR and IGF-1 pathways and promotes ketosis, which reduces the availability of glucose and leads to the reliance on the HMP shunt over oxidative phosphorylation. The deprivation of glucose in the ketogenic diet creates an environment where non-malignant cells are able to utilize ketones to survive during ketosis, while cancer cells are “starved” of an energy source stemming from a lack of glucose availability.
MAPK: mitogen-activated protein kinase; PI3K: phosphatidylinositol 3-kinase; AKT: protein kinase B; HMP: hexose monophosphate; IGF: insulin-like growth factor; mTOR: mammalian target of rapamycin; AMPK: AMP-activated protein kinase; AMP: adenosine monophosphate.
The numbers indicate references with additional information about the dietary interaction and metabolic pathway interactions.
The utility of CR may be further enhanced when complemented with local cytotoxic therapy. Targeting the primary tumor with local radiation, in combination with CR, is hypothesized to alter the molecular profile of the tumor.85 Indeed, mice receiving an IGF-1R inhibitor, to replicate CR, along with radiation therapy, required a longer period to develop breast cancer metastasis to the lung and had increased overall survival.85 A report shows that chloroquine, in combination with cell starvations, induces a greater degree of growth arrest and cell death compared with either treatment used in isolation in melanoma, glioma, and fibrosarcoma cells.86 This synergistic potential of CR is an especially powerful tool that may better the outcomes of patients receiving standard therapies.
Another growth factor associated with obesity is leptin, a hormone produced by white adipose tissue that activates cell proliferation and affects angiogenesis, carcinogenesis, and cytokine production.87 CR has been shown to decrease leptin release while increasing serum levels of adiponectin, another growth hormone found to improve insulin sensitivity and promote anti-inflammatory and anti-proliferative properties.88 As stated previously, leptin may have proliferative and angiogenic effects to promote cancer progression. Since CR may decrease serum leptin levels,89 this concept has been applied to breast cancers, where it has be shown that leptin may have pro-carcinogenic effects via modulation of signaling pathways responsible for tumor proliferation.90 Further, a meta-analysis found that elevated leptin levels may play a significant role in the metastasis of breast cancer.91 Since leptin receptors have been found to be overexpressed in breast tumors compared to normal breast tissue,92 leptin inhibition has gained attention as a powerful new target in breast cancer therapy.93 Overall, CR may be utilized as an adjuvant in patients with cancer due to its effects on serum leptin levels.
CR also targets the mechanism of autophagy to inhibit tumor proliferation. Autophagy is a catabolic process of self-eating in response to starvation and stress which in theory could increase tumor cell death, thus inhibiting tumor proliferation.94 It is utilized by non-cancerous cells to promote the recycling of cellular components required to maintain homeostatic function. Stressors, such as exposure to metals, hypoxia, sepsis, and chemotherapy, induce oxidative damage that may harm organelles, especially mitochondria; thus, autophagy provides the mechanism to degrade defective organelles, remove misfolded proteins, and digest non-critical cellular components as an alternative energy source to repair critical proteins.94 It has been found that CR may further promote autophagy and assist with maintenance of homeostasis in the exposure to stress.95 The balance of autophagy is under the influence of the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) axis.73 IGF-1 has been shown to activate mTOR, which promotes cellular proliferation and inhibits autophagy, while AMPK inhibits mTOR. CR has been shown to activate AMPK, thereby inducing systemic autophagic activity.73 While the role of autophagy in benign cells is well established, the function of autophagy in cancerous cells is unclear. An upregulation of autophagy in surrounding stromal cells, in the context of decreased autophagy in cancerous cells, has been proposed to be both tumor suppressive and promotive.94,96 Conversely, some propose that established cancer cells instead may upregulate autophagy during CR, thus promoting evasion of TNF-α-induced apoptosis and cellular survival.73 Further research behind the underlying effects on the proliferation pathways is essential to better understand how CR-induced autophagy contributes to tumorigenesis.
Angiogenesis and inflammation have been shown to be related to tumor proliferation, and CR has been shown to module these processes to exert anti-neoplastic effects. CR has been shown to decrease expression of plasminogen activator inhibitor 1 (PAI-1)97 and vascular endothelial growth factor (VEGF)74,98 which are cytokines that promote angiogenesis, thereby allowing for metastatic spread of tumor cells. CR also decreases levels of chronic inflammation by reducing absolute quantity of adipose tissue.99 When adipocytes are present in excess, they risk hypoxia and necrosis from insufficient perfusion.100 Free fatty acids may escape from necrotic tissue and release inflammatory cytokines such as nuclear factor kappa B (NF-κB), a transcription factor that induces gene expression for cellular proliferation and metastasis, or COX-2, which increases production of inflammatory lipid metabolites.101 CR has shown to be a potent inhibitory modulator of tumorigenesis via inhibition of angiogenesis and inflammation.

Limitations

It is important to consider the patient’s baseline nutritional status, stage of therapy, and recommendations from the primary oncologist and registered dietitian prior to incorporating any dietary intervention. One of the primary concerns with CR is the risk of cachexia. Extreme cachexia has been associated with alterations between the patient’s immune system and metabolic state.73 Since CR requires a decrease in caloric intake, patients who have already experienced significant weight loss due to therapy, vomiting, diarrhea, or anorexia may not be ideal candidates to incorporate CR into their overall treatment plan. In addition, CR may require several months for effects to be observed in patients,102 thus potentially exacerbating further weight loss in patients who may already be significantly underweight and at risk for malnourishment. In addition, some physicians may advise against CR as certain chemotherapy regimens are already associated with patient-driven CR due to nausea and anorexia. As a result, the practicality of CR has been difficult to assess since oncology patients are generally advised to increase food intake to combat therapy-induced weight loss.103 Therefore, other dietary interventions, such as intermittent fasting (IF), fasting-mimicking diets, or ketogenic diets (KDs), which have lower potential for weight loss, have been explored as alternative forms of dietary interventions in cancer therapy.
Due to the concerns surrounding extreme weight loss in at-risk patients, CR mimetics have been proposed as an alternative to provide the beneficial effects of CR without inducing cachexia. CR mimetics are supplements or medications that take advantage of the beneficial effects described during CR without dietary restriction.73 For example, metformin is a common type 2 diabetes medication which activates the AMPK (5’ AMPK) pathway and increases insulin sensitivity in a similar fashion to the proposed CR mechanism described above that has been linked to anti-neoplastic activity.104 Metformin is a relatively safe medication; however, common side effects include hypoglycemia. Another drug of interest is rapamycin, an antibiotic and immunosuppressive drug that inhibits mTOR, which has been shown to delay cancer progression, extend life span, and increase the sensitivity of certain cancers to chemotherapy and radiation therapy.105 Common side effects of rapamycin include hypertriglyceridemia, increased risk of infection, diabetes-like symptoms, and hypertension.106 Given these promising results, a wide variety of CR mimetics are currently under development to affect tumor metabolism and sensitize tumors to standard and emerging cancer therapies.73 Of note, side effect profiles should be considered prior to starting any medications as their risk may vary widely based on the patient’s medical history.

IF

Introduction

IF has been used as a dietary regimen for acute and chronic diseases worldwide.103 IF is a dietary regimen that includes the complete cessation of caloric intake for a period of time, generally from 16 up to 120 h, followed by a refeeding period. During the refeeding period, a person is not restricted to certain food groups or amount of food they can consume.103 In murine models, IF has been shown to improve the efficacy of chemotherapy regimens used in breast cancer, melanoma, neuroblastoma, pancreatic cancer, and colorectal cancer.107 Interestingly, IF has been shown to protect healthy cells against chemotherapy damage and stress resistance as a “protective effect,” therefore decreasing off-target side effects of standard chemotherapy regimens.107,108 Therefore, the utility of IF has emerged as a possible adjuvant dietary therapy to decrease tumor proliferation and improve therapeutic tolerance.

Proposed mechanisms

The primary benefit of IF is based on the physiological mechanisms that occur during starvation. During first 10 h of starvation, the body will utilize consumed food and stored glycogen for energy primarily in the liver. Upon depletion of liver glycogen stores, muscle and adipose tissues are broken down to supply energy through an alternative pathway. Amino acids are produced via breakdown of muscle and fatty acids via breakdown of adipose tissue. Fatty acids can further be broken down into ketone bodies, which can be used as an energy source by most tissues except the brain.109 Clearly, acute periods of starvation induce a drastic change in the metabolic state of the body and has shown to play a role in cancer models.
During fasting, where there is a cessation of caloric intake, normal cells will reallocate energy away from anabolic and regenerative pathways toward maintenance pathways due to resource limitation, which may limit the growth potential of cancerous cells.107 This overall decrease in anabolism is thought to lead to a protective state known as a phenomenon termed differential stress resistance (DSR).110,111 Unfortunately, neoplastic cells are incapable of inhibition of oncogenes, such as Ras/Raf/MAPK, phosphatase and tensin homolog (PTEN), and PI3K, which are the primary drivers of malignant proliferation cellular profiles (Figure 1).107,111 Together, IF may be a mechanism of protecting the healthy cells from transformation into malignant cells and resistance against treatment-related toxicity.
The differential stress response is manifested through several molecular mechanisms. IF has been reported in literature to delay tumor progression and increase effectiveness of chemotherapy against a variety of cancers, including melanoma, glioma, and breast cancer.107 This sensitization of tumor cells to chemotherapy is thought to be related to the decreased circulation of growth factors and changes in plasma levels of nutrients that sensitize cells to toxic therapy.112 Specifically, IF has been shown to decrease insulin and growth hormone levels, which lead to decreased IGF-1 levels thus reducing the activation of the Ras/MAPK and PI3K pathways.113,114 In addition, IF has been shown to activate AMPK (5’ AMPK), which inhibits cellular proliferation and protein synthesis, activates glycolysis and fatty acid oxidation, and induces autophagy.115,116 IF has also been shown to increase activation of caspase-3, which is a crucial mediator of programmed cellular death that is selectively upregulated in malignant cells.107 IF has multiple molecular mechanisms that appear to have a complex role in cancer metabolism and further studies will be needed to elucidate key pathways.
Murine and human studies have shown that IF may reduce the toxicities from chemotherapy and improve clinical outcomes. Specifically, a case series found that fasting before and after chemotherapy can selectively enhance toxicity of chemotherapy drugs to various malignancies, including the breast, esophagus, and prostate.117 One study found that fasting cycles combined with cyclophosphamide reduced breast tumor size to less than half of what was seen with fasting or chemotherapy alone. This same study also saw that combination of low serum glucose with chemotherapy promoted a 20-fold increase in DNA damage in breast and melanoma cancer cells, greater than what was seen with each treatment in isolation.107 Another study shows that starvation of mice for 24 h before and after chemotherapy selectively sensitizes cancer cell lines to oxidative stress from doxorubicin and cyclophosphamide.108 Thus, multiple studies have shown that there may be a synergistic effect between IF and specific chemotherapies, suggesting that particular cancer types may benefit more from adjuvant dietary interventions such as IF.
Severe side effects from chemotherapy can limit a patient’s ability to receive the full course of their treatment regimen; thus, some investigators have sought to study the potential ways that IF could decrease the severity of these side effect profiles. In a murine model, mice that underwent IF for 48–60 h had lower rates of toxicity to etoposide than mice that were allowed to eat ad libitum.108,118 In two human cohorts, IF was well tolerated as well and showed significant decrease in chemotherapy-related side effects, such as weakness, fatigue, and nausea,117 while another study showed less gastrointestinal (GI) upset from therapy.119 Together, these studies suggest that IF may improve side effect profiles, allowing the individual to better tolerate a complete chemotherapy regimen without delays in care or dose reduction, thereby potentially promoting the effectiveness of their therapy.

Limitations

The main concern about IF is the potential for significant weight loss. Many cancer patients experience nausea, vomiting, diarrhea, loss of appetite, or depression which can lead to malnutrition in this already at-risk population.119 Studies have suggested that IF may need to be extended to at least 24 h and potentially up to 48 h, to see the protective effects described above.103 In contrast to an overall reduction in caloric intake which is used in CR, IF has the potential to lead to malnourishment, given the strict caloric limitations.73,103 With that in mind, the extended duration of IF may not be possible for some patients for various reasons, including baseline weight and nutritional status prior to diagnosis, particular cancer diagnosis, current chemotherapy regimens being used, and their individual risk for nutritional deficiencies. Therefore, the cost/benefit of IF should be assessed on an individual basis with the treating oncologist, registered dietitian, and patient to determine if IF is safe and feasible. Overall, preliminary results suggest that IF is well tolerated by most patients but the risk of severe weight loss cannot be ignored.

KD

Introduction

The KD is a high-fat, moderate-protein, and low-carbohydrate diet. Recently, the KD has gained popularity as a method to promote weight loss and reduce risk for various chronic diseases,120 including metabolic diseases such as GLUT-1 deficiency and pyruvate dehydrogenase complex deficiency.121 Most notably, the KD has been used for over 80 years as an effective adjuvant therapy for children suffering from refractory epilepsy122,123 and, more recently, it is being explored with diabetes and cancer.124126
The foundation of the KD is an emphasis on higher fat consumption with moderate to low protein content and very low carbohydrate intake, with a ratio of fat:carbohydrate and protein around 3:1 or 4:1 in order to drastically decrease circulating glucose, which is thought to be the primary energy source of the rapid dividing malignant cells.126 This dramatic shift away from glucose as the primary energy supply to ketosis which is the breakdown of adipose tissue via fatty acid oxidation leading to ketone body formation is known as dietary ketosis.127 Ketosis has been hypothesized to inhibit tumor proliferation while still providing sufficient energy for peripheral tissue.126,127 Moreover, CR and KD have been found to dramatically decrease tumor cell proliferation through the inhibition of the IGF-1 pathway (Figure 1);128,129 however, the degree of weight loss may potentiate cancer-related cachexia.103,119 This has led to the KD being explored as an alternative dietary regimen that shifts the metabolic processes away from tumor growth while avoiding CR and prolonged periods of fasting.

Proposed mechanism

The proposed effectiveness of the KD relies on the many metabolic differences between normal cells and cancer cells, especially in the metabolism of glucose. As a review, under normal oxygen-rich conditions, glucose is broken down to pyruvate via glycolysis, which is then converted to acetyl CoA. Acetyl CoA enters the mitochondria to initiate the citric acid cycle and the electron transport chain in a process known as oxidative phosphorylation to produce energy for the body in the form of adenosine triphosphate or ATP (36 ATP per one glucose molecule). This process is driven by the availability of glucose and is the most efficient mechanism of energy production in the body. Mitochondria with preserved function tightly regulate cellular energy production via these processes, and the majority of ATP is produced through oxidative phosphorylation. This is the primary mechanism by which all cells in our body receive energy for growth and metabolism and cancer cells are no exception. It is has been suggested that tumor cells may have mitochondrial DNA mutations that drive cells to utilize the oxidative phosphorylation pathway, leading to an increase in reactive oxygen species production and oxidative stress as a subsequent by-product of aerobic metabolism.130
In the absence of oxygen, pyruvate is instead converted to lactate for energy production via non-oxidative pathways such as ketosis, which produces far less ATP than oxidative phosphorylation (two ATPs per one glucose molecule). ATP is essential for driving the metabolic machinery in all cells, especially for malignant cells. Since malignant cells require more ATP, it has been postulated that if one can decrease excess glucose intake, then the glycolytic pathways will prevail and essentially “starve” the malignant cells, leading to cellular death. This has led to the idea that a KD may aid in “starving” malignant, thus reducing or preventing cellular proliferation pathways.
Malignant cells are thought to utilize a unique metabolic pattern termed the Warburg effect, where anaerobic glycolysis is preferably utilized instead of oxidative phosphorylation for ATP production, regardless of oxygen status.131 As described above, a diet low in carbohydrates will result in an initial increased rate of glucose metabolism forcing the cells to utilize glycolysis as their primary energy source. This is thought to be beneficial to the patient as malignant cells are rapidly dividing and thus require glucose to more efficiently produce energy to drive their anabolic metabolism. Some studies have shown that glucose uptake and lactate release by tumors is 30- to 43-fold that of non-malignant cells, suggesting that tumor cells will utilize both pathways, including ketosis, to compensate for lack of glucose availability for oxidative phosphorylation (Figure 1).126,132 In addition, to address the lack of glucose, malignant cells rely heavily on the hexose phosphate pathway (HMP shunt),133 which converts glucose 6-phosphate, an intermediate in glycolysis, to the cofactors nicotinamide adenine dinucleotide phosphate (NADPH) and ribose 5-phosphate to detoxify organic peroxides and mitigate oxidative damage.130
The KD seeks to shift the metabolism toward ketosis to limit anabolism, therefore inhibiting growth and proliferation pathways in malignant cells. When glucose is limited, the body is forced to utilize stored adipose tissue instead of glucose for ATP production. After prolonged periods of glucose restriction, the body preferentially enters ketosis where fat is metabolized via fatty acid oxidation to produce ketone bodies including β-hydroxybutyrate, acetone, and acetoacetate. Ketone bodies are then converted to acetyl CoA for use in the citric acid cycle for ATP production.126 This is an important underlying principal in the KD, where glucose availability is limited, creating an environment where non-malignant cells are able to utilize ketones to survive during ketosis, while cancer cells are “starved” of an energy source due to a lack of glucose availability.
Thus, the KD may selectively increase oxidative stress in cancer cells through two mechanisms. First, malignant cells are dependent on glucose metabolism for the production of glucose 6-phosphate, which is converted to detoxifying cofactors via HMP shunt. Without glucose, the source of cofactors to reduce reactive oxygen species is eliminated, thereby inducing oxidative stress in malignant cells.126 Second, forced fatty acid oxidation in the KD provides energy that is primarily utilized by non-malignant cells since cancerous cells are poor metabolizers of ketones. Together, the KD decreases the substrate for the most efficient energy production in the cell via a significant reduction in glucose availability while concurrently promoting oxidative stress in malignant cells by removing the source for detoxifying cofactors.
There are a wide variety of studies that illustrate the possible anti-neoplastic effects of the KD.134,135 A review article evaluated the effects of KD on tumor growth and survival time in animal models. The majority of the papers revealed a beneficial effect seen across a variety of tumor types including prostate, gastric, neuroblastoma, and lung.136 More specifically, in a colon carcinoma murine model, mice fed with a ketogenic formula showed cancer suppression compared with mice fed with regular diets. Interestingly, there was a significant negative correlation between blood ketone concentration and tumor weight. Adverse effects were not noted. The investigators postulated that elevated blood ketone levels may have anti-tumor effects by promoting maintenance of body weight and muscle mass leading to a reduction of inflammation.137 Another mouse model with neuroblastoma xenografts shows a significant decrease in neoplastic growth when exposed to low-dose chemotherapy in combination with a KD diet.138 Despite the evidence of anti-neoplastic activity of the KD in animal models, the data in humans are lacking (Table 1).135

Limitations

Despite the promising effects of the KD, this regimen risks potential side effects including lethargy, nausea, and vomiting, in addition to hypoglycemia.126 These side effects are not universal but could limit one’s ability to adhere to the KD. In addition, the high fat content of KD may not be well tolerated by some individuals, with some studies showing long-term increases in serum total cholesterol139 along with renal damage.140 In addition, some patients experience progressive bone mineral content loss; however, this is likely multifactorial, given the use of steroids and various chemotherapies, in addition to gender discrepancies, that may contribute to bone loss.141 Although some propose that the KD can lead to weight loss,129,142 more recent data suggest that the KDs may actually prevent cachexia and maintain body weight.137 Regardless, more studies are needed to further illustrate the utility and adverse effects of the KD (Table 2).
Table 2. Comparison of dietary interventions.
Dietary intervention Benefits Limitations
Western diet None Low nutritional density
Associated with prostate, breast, and colorectal cancer
Associated with chronic diseases
Caloric Restriction Reduction in oxidative stress, inflammation, and growth factors (i.e. IGF-1 and Ras/MAPK)
Improved insulin sensitivity and glucose tolerance
Decreased leptin levels
Promotes autophagy
Decreased angiogenesis
Excessive weight loss
Risk of cachexia
Risk of malnutrition
Intermittent fasting Associated with improved chemotherapy-associated side effects
Improved insulin sensitivity and glucose tolerance
Decreased growth factors (i.e. IGF-1 and Ras/MAPK)
Decrease anabolic metabolism (termed differential stress resistance)
Increased AMPK
Excessive weight loss
Risk of cachexia
Risk of malnutrition
Ketogenic diet Increased ketosis
Decreased inflammation and growth factors (i.e. IGF-1)
Inhibition of tumorigenesis
Utilization of Warburg effect
Selective increased oxidative stress in cancer cells
Weight loss
Hypoglycemia, nausea, vomiting, and lethargy
Increase in serum cholesterol
Progressive bone loss
IGF-1: insulin-like growth factor-1; MAPK: mitogen-activated protein kinase; AMPK: 5’ adenosine monophosphate–activated protein kinase.

Other dietary regimens under exploration

Mediterranean diet

The Mediterranean diet (MedD) has gained attention as a healthy diet to reduce the risk of cancer. Schwingshackl and Hoffmann143 provide a comprehensive review on the proposed beneficial effects of the MedD on overall risk of cancer. The MedD is common in countries bordering the Mediterranean Sea. It is defined by a high consumption of plant-based foods, whole grain products, vegetables, fruits, nuts, and legumes along with a regular intake of fish and seafood, while eggs and red/processed meats are limited. Alcohol consumption is moderate, with a preference for red wine, and olive oil serves as a predominant form of fat.143
The supposed protective effects of the MedD are not due to any single dietary component but rather the cumulative pattern. Higher fruit and vegetable consumption affects inflammation, redox reactions, and various metabolic processes that may exert anti-cancer effects and promote healthy weight management.144 Whole grains contain phytic acids and fiber that bind and neutralize carcinogens in the GI tract.145 Olive oil contains polyphenols, which contribute anti-inflammatory and anti-oxidant effects.146 The role of dairy products is uncertain and may increase risk of prostate cancer,147 but calcium and protein in dairy may also illicit anti-neoplastic effects.148 The MedD reduces intake of red meats, which are pro-inflammatory and pro-oxidative.149 The benefits of alcohol are controversial since the evidence with moderate consumption and cancer risk is mixed.150,151 Evidently, the MedD is a complex diet with numerous components that may exert individual effects on cancer protection and cancer risk.
Overall, there was an observed inverse association between adherence to MedD and cancer mortality. The strongest results were seen in colorectal cancer,143 with smaller associations seen in cancers of the breast, gastric, liver, gallbladder, head and neck, and prostate.143,152 While results are promising, exact interpretation is difficult since there is no clear-cut definition of an MedD since it varies from region to region based on ethnic, cultural, religious, and economic factors. Again, this is because the MedD is not a discrete regimen, but rather a collection of food practices seen in populations that border the Mediterranean Sea.153 As such, future studies might focus on identifying a more analytically illuminating definition of the MedD that may help delineate its putative beneficial effects.

Japanese diet

Individuals from Japan have a lower incidence of various cancers, especially those termed fat-related cancers of the colon, breast, prostate, and ovary.154 Inhabitants of Okinawa, who historically consume significantly fewer calories than those living on the main islands, have lower death rates from cancer and other chronic conditions compared with Japanese living in the main lands.155 For Japanese individuals who moved from Japan to Hawaii, the incidence of stomach cancer decreased while the rates of breast, colon, and prostate cancer increased in just one generation.156 In the last 40 years, the incidence of colon cancer has increased 9.4 times for males and 4.7 times for females as Japanese have adopted a more Western dietary pattern.157
The lower incidence of malignancies is thought to stem from the makeup of the traditional Japanese diet, which is characterized by a low intake of fats and oils, along with a low intake ratio of n-6 polyunsaturated fatty acids (PUFAs) versus n-3 PUFAs.154 In addition, other components of the Japanese diet, such as higher intake of vegetables and dietary fiber, may contribute to the lower cancer rates.158 Of interest is miso soup, which contains wakame, a vegetable rich in fucoxanthin that may have protective effects against gastric cancer.159 Japanese individuals also demonstrate a higher intake of green tea, which is rich in flavonoids and may have anti-oxidant effects.160 Soy products contain isoflavones and saponins that may have similar beneficial effects.161 Together, these individual components of the Japanese diet are hypothesized to establish the protective roles of this dietary regimen in cancer risk.
A precise characterization of a Japanese diet is difficult to achieve, and just why it is associated with lower incidence of malignancies has been puzzling. Regardless, further studies are warranted to elucidate possible mechanisms behind its apparent protective effects.

Vegan diet

A vegetarian diet removes any meat, poultry, and fish from one’s diet. A vegan diet further removes all animal products, including dairy or eggs.162 Overall, vegan diets are high in fiber, magnesium, folic acid, iron, and vitamin C and E, and low in overall calories, saturated fats, zinc, calcium, vitamin D, and vitamin B12.163 The vegan diet has been associated with a wide variety of health benefits, including lower risk of cardiovascular disease, obesity, and type II diabetes.164 Of special interest in this review is the lower risk of cancer in patients who follow a vegan diet.
There are several proposed mechanisms for the protective anti-neoplastic effects of a plant-based diet. The body mass index (BMI) of individuals who follow a vegan diet is much lower;163 thus, the risk of obesity-related cancers, such as colorectal, breast, and prostate cancers, is decreased. In addition, vegan diets consist of a higher proportion of fruits and vegetables, which contain phytochemicals and anti-oxidants, in addition to fiber, flavonoids, and vitamin C, which are all proposed to have protective effects against the development of malignancy.165167 Of important consideration is the increased intake of phytochemicals, which are abundant in plants, and the decreased intake of red/processed meats. In animal models and in vitro studies, phytochemicals have been shown to induce apoptosis, arrest cell growth, and decrease angiogenic potential, though beneficial effects have not been shown to be consistent across studies.168,169 Red meat has been strongly associated with the development of colorectal cancer,170 in addition to malignancies of the esophagus, liver, and lung.149 Another characteristic of a vegan diet includes high consumption of legumes, which may decrease risk of prostate cancer.171 A differentiating factor between the vegetarian diet and vegan diet is the absence of eggs in the latter, which have been associated with an increased risk of pancreatic cancer.172 Evidently, the cumulative effects of the vegan diet, due to food items that are consumed in abundance and food items that are avoided, contribute to the potentially anti-cancer properties of this dietary regimen.
Overall, following a vegan diet appears to provide a variety of health benefits. Although the literature is deficient in clinical trials that assess the relationship between plant-based diets and cancers, data thus far suggest that vegan and vegetarian diets are nonetheless protective against cancer, with a net 10%–12% reduction in overall cancer risk.173 As such, further research is warranted to explore the utility of a vegan diet as a lifestyle modification for cancer prevention.

Conclusion

Expert nutrition groups have issued clinical guidelines for nutritional treatment of cancer patients. These guidelines state that patients should undergo nutrition screening and assessment and receive early nutrition intervention to improve outcomes. However, it appears that there has been an absence of innovative or clinically meaningful techniques. So, where are we really? With unprecedented sophistication in molecular biology, investigational oncology, and computational power, clinical medicine in general and oncologic nutrition in particular have failed to achieve meaningful progress for patients. A casual survey indicates virtually every major “comprehensive cancer center” has a page on cancer nutrition support. The American Cancer Society’s “Nutrition for People with Cancer” website page declares, “Nutrition is an important part of cancer treatment. Eating the right kinds of foods during and after treatment can help you feel better and stay stronger.” While the internet material is, of course, intended for patients, its quixotic but superficial character speaks volumes. Upon review of the website information and firsthand assessment of clinical reality, it may be reasonably concluded that there is a strong conventional wisdom that the pathology of cancers can result in malnutrition, that cancer treatments often result in malnutrition, and that chronic undernutrition and overnutrition can have an impact on either accelerating or reducing cancer risk or tumor burden (Table 2).

Declaration of conflicting interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) received no financial support for the research, authorship, and/or publication of this article.

ORCID iDs

References

1. National Cancer Statistics Annual Report to the Nation, 2018, https://seer.cancer.gov/report_to_nation/statistics.html
2. Cronin KA, Lake AJ, Scott S, et al. Annual report to the nation on the status of cancer, Part I: national cancer statistics. Cancer 2018; 124(13): 2785–2800, http://doi.wiley.com/10.1002/cncr.31551
3. Hales CM, Carroll MD, Fryar CD, et al. Prevalence of obesity among adults and youth: United States, 2015-2016 key findings data from the national health and nutrition examination survey, 2015, https://www.cdc.gov/nchs/data/databriefs/db288_table.pdf#1
4. Calle EE, Rodriguez C, Walker-Thurmond K, et al. Overweight, Obesity, and Mortality from Cancer in a Prospectively Studied Cohort of U.S. Adults. N Engl J Med 2003; 348: 1625–1638.
5. Kohler GMC. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975; 256: 495–497, https://www.nature.com/articles/256495a0.pdf
7. Kolonel LN, Nomura AM, Hinds MW, et al. Role of diet in cancer incidence in Hawaii. Cancer Res 1983; 43(5 Suppl): 2397s–2402s, http://www.ncbi.nlm.nih.gov/pubmed/6831463
8. McMichael AJ, McCall MG, Hartshorne JM, et al. Patterns of gastro-intestinal cancer in European migrants to Australia: the role of dietary change. Int J Cancer 1980; 25(4): 431–437, http://www.ncbi.nlm.nih.gov/pubmed/7372370
9. Peisch SF, Van Blarigan EL, Chan JM, et al. Prostate cancer progression and mortality: a review of diet and lifestyle factors. World J Urol 2017; 35(6): 867–874, http://www.ncbi.nlm.nih.gov/pubmed/27518576
10. Longo VD, Fontana L. Calorie restriction and cancer prevention: metabolic and molecular mechanisms. Trends Pharmacol Sci 2010; 31: 89–98.
11. Galet C, Gray A, Said JW, et al. Effects of calorie restriction and IGF-1 receptor blockade on the progression of 22Rv1 prostate cancer xenografts. Int J Mol Sci 2013; 14: 14, www.mdpi.com/journal/ijmsArticle
12. Fontana L, Partridge L, Longo VD. Extending healthy life span—from yeast to humans. Science 2010; 328(5976): 321–326, http://www.ncbi.nlm.nih.gov/pubmed/20395504
13. Redman LM, Heilbronn LK, Martin CK, et al. Metabolic and behavioral compensations in response to caloric restriction: implications for the maintenance of weight loss. PLoS One 2009; 4(2): e4377, http://www.ncbi.nlm.nih.gov/pubmed/19198647
14. Gill JG, Piskounova E, Morrison SJ. Cancer, oxidative stress, and metastasis. Cold Spring Harb Symp Quant Biol 2016; 81: 163–175, http://www.ncbi.nlm.nih.gov/pubmed/28082378
15. Ornish D, Weidner G, Fair WR, et al. Intensive lifestyle changes may affect the progression of prostate cancer, 2005, https://www.ornish.com/wp-content/uploads/Intensive_Lifestyle_Changes_and_Prostate_Cancer.pdf
16. IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Tobacco smoke and involuntary smoking. IARC Monogr Eval Carcinog Risks Hum 2004; 83: 1–1438, http://www.ncbi.nlm.nih.gov/pubmed/15285078
17. Xu R, Barg FK, Emmett EA, et al. Association between mesothelioma and non-occupational asbestos exposure: systematic review and meta-analysis. Environ Heal 2018; 17(1): 90, http://www.ncbi.nlm.nih.gov/pubmed/30567579
18. Kushi LH, Doyle C, McCullough M, et al. American cancer society guidelines on nutrition and physical activity for cancer prevention. CA Cancer J Clin 2012; 62(1): 30–67, http://www.ncbi.nlm.nih.gov/pubmed/22237782
19. Bayo J, Molina R, Pérez J, et al. SEOM clinical guidelines to primary prevention of cancer (2018). Clin Transl Oncol 2019; 21: 106–113, https://doi.org/10.1007/s12094-018-02016-4
20. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: An international consensus. Lancet Oncol 2011; 12: 489–495.
21. Tackling the Conundrum of Cachexia in Cancer National Cancer Institute, 2011, https://www.cancer.gov/about-cancer/treatment/research/cachexia
22. Tisdale MJ. Mechanisms of cancer cachexia. Physiol Rev 2009; 89: 381–410.
23. Tayek JA. A review of cancer cachexia and abnormal glucose metabolism in humans with cancer. J Am Coll Nutr 1992; 11(4): 445–456, http://www.ncbi.nlm.nih.gov/pubmed/1506607
24. Wang F, Liu H, Hu L, et al. The Warburg effect in human pancreatic cancer cells triggers cachexia in athymic mice carrying the cancer cells. BMC Cancer 2018; 18(360): 1–12, https://doi.org/10.1186/s12885-018-4271-3
25. Evans WK, Makuch R, Clamon GH, et al. Limited impact of total parenteral nutrition on nutritional status during treatment for small cell lung cancer. Cancer Res 1985; 45(7): 3347–3353, http://www.ncbi.nlm.nih.gov/pubmed/2988769
26. Moley JF, Aamodt R, Rumble W, et al. Body cell mass in cancer-bearing and anorexic patients. J Parenter Enter Nutr 1987; 11(3): 219–222, http://www.ncbi.nlm.nih.gov/pubmed/3474427
27. Nguyen TYV, Batterham MJ, Edwards C. Comparison of resting energy expenditure between cancer subjects and healthy controls: a meta-analysis. Nutr Cancer 2016; 68(3): 374–387, http://www.ncbi.nlm.nih.gov/pubmed/27007947
28. Blum KA, Lozanski G, Byrd JC. Adult Burkitt leukemia and lymphoma. Blood 2004; 104: 3009–3020.
29. Roesch K, Hasenclever D, Scholz M. Modelling lymphoma therapy and outcome. Bull Math Biol 2014; 76(2): 401–430, http://www.ncbi.nlm.nih.gov/pubmed/24338592
30. Warburg O, Wind F, Negelein E. The metabolism of tumors in the body. J Gen Physiol 1927; 8(6): 519–530, http://www.ncbi.nlm.nih.gov/pubmed/19872213
31. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de novo protein synthesis binds to the human erythropoietin gene enhancer at a site required for transcriptional activation. Mol Cell Biol 1992; 12(12): 5447–5454, http://www.ncbi.nlm.nih.gov/pubmed/1448077
32. Bui T, Thompson CB. Cancer’s sweet tooth. Cancer Cell 2006; 9(6): 419–420, http://www.ncbi.nlm.nih.gov/pubmed/16766260
33. Kim J, Tchernyshyov I, Semenza GL, et al. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab 2006; 3(3): 177–185, http://www.ncbi.nlm.nih.gov/pubmed/16517405
34. Waterhouse C, Keilson J. Cori cycle activity in man. J Clin Invest 1969; 48(12): 2359–2366, http://www.ncbi.nlm.nih.gov/pubmed/5355347
35. Straub RH, Cutolo M, Buttgereit F, et al. Review: energy regulation and neuroendocrine-immune control in chronic inflammatory diseases. J Intern Med 2010; 267(6): 543–560, http://www.ncbi.nlm.nih.gov/pubmed/20210843
36. Pepys MB, Hirschfield GM, Tennent GA, et al. Targeting C-reactive protein for the treatment of cardiovascular disease. Nature 2006; 440(7088): 1217–1221, http://www.nature.com/articles/nature04672
37. Fearon KC, Barber MD, Falconer JS, et al. Pancreatic cancer as a model: inflammatory mediators, acute-phase response, and cancer cachexia. World J Surg 1999; 23(6): 584–588, http://www.ncbi.nlm.nih.gov/pubmed/10227928
38. Tijerina AJ. The biochemical basis of metabolism in cancer cachexia. Dimens Crit Care Nurs 2004; 23(6): 237–243, http://www.ncbi.nlm.nih.gov/pubmed/15586034
39. Giordano A, Calvani M, Petillo O, et al. Skeletal muscle metabolism in physiology and in cancer disease. J Cell Biochem 2003; 90(1): 170–186, http://www.ncbi.nlm.nih.gov/pubmed/12938166
40. Narsale AA, Carson JA. Role of interleukin-6 in cachexia. Curr Opin Support Palliat Care 2014; 8(4): 321–327, http://www.ncbi.nlm.nih.gov/pubmed/25319274
41. Torelli GF, Meguid MM, Moldawer LL, et al. Use of recombinant human soluble TNF receptor in anorectic tumor-bearing rats. Am J Physiol Integr Comp Physiol 1999; 277(3): R850–R855, http://www.ncbi.nlm.nih.gov/pubmed/10484503
42. Tisdale MJ. Biology of cachexia. J Natl Cancer Inst 1997; 89(23): 1763–1773, http://www.ncbi.nlm.nih.gov/pubmed/9392617
43. Patra SK, Arora S. Integrative role of neuropeptides and cytokines in cancer anorexia–cachexia syndrome. Clin Chim Acta 2012; 413(13–14): 1025–1034, https://www.sciencedirect.com/science/article/pii/S0009898111006723?via%3Dihub
44. McCarthy HD, McKibbin PE, Perkins AV, et al. Alterations in hypothalamic NPY and CRF in anorexic tumor-bearing rats. Am J Physiol 1993; 264(4 Pt 1): E638–E643, http://www.ncbi.nlm.nih.gov/pubmed/8476042
45. Chance WT, Balasubramaniam A, Dayal R, et al. Hypothalamic concentration and release of neuropeptide Y into microdialysates is reduced in anorectic tumor-bearing rats. Life Sci 1994; 54(24): 1869–1874, https://www.sciencedirect.com/science/article/pii/0024320594901449
46. Chance WT, Balasubramaniam A, Thompson H, et al. Assessment of feeding response of tumor-bearing rats to hypothalamic injection and infusion of neuropeptide Y. Peptides 1996; 17(5): 797–801, https://www.sciencedirect.com/science/article/pii/0196978196001088
47. Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature 1998; 395(6704): 763–770, http://www.nature.com/articles/27376
48. Sarraf P, Frederich RC, Turner EM, et al. Multiple cytokines and acute inflammation raise mouse leptin levels: potential role in inflammatory anorexia. J Exp Med 1997; 185(1): 171–175, http://www.ncbi.nlm.nih.gov/pubmed/8996253
49. Finck BN, Kelley KW, Dantzer R, et al. In Vivo and in Vitro Evidence for the Involvement of Tumor Necrosis Factor-α in the Induction of Leptin by Lipopolysaccharide 1. Endocrinology 1998; 139(5): 2278–2283, https://academic.oup.com/endo/article-lookup/doi/10.1210/endo.139.5.6012
50. Ramos EJB, Suzuki S, Marks D, et al. Cancer anorexia-cachexia syndrome: cytokines and neuropeptides. Curr Opin Clin Nutr Metab Care 2004; 7(4): 427–434, http://www.ncbi.nlm.nih.gov/pubmed/15192446
51. Tong H, Isenring E, Yates P. The prevalence of nutrition impact symptoms and their relationship to quality of life and clinical outcomes in medical oncology patients. Support Care Cancer 2009; 17(1): 83–90, http://www.ncbi.nlm.nih.gov/pubmed/18551322
52. Fearon KC, Voss AC, Hustead DS, et al. Definition of cancer cachexia: effect of weight loss, reduced food intake, and systemic inflammation on functional status and prognosis. Am J Clin Nutr 2006; 83(6): 1345–1350, http://www.ncbi.nlm.nih.gov/pubmed/16762946
53. Fuchs N, Bielack SS, Epler D, et al. Long-term results of the co-operative German-Austrian-Swiss osteosarcoma study group’s protocol COSS-86 of intensive multidrug chemotherapy and surgery for osteosarcoma of the limbs. Ann Oncol Off J Eur Soc Med Oncol 1998; 9(8): 893–899, http://www.ncbi.nlm.nih.gov/pubmed/9789613
54. Ferreri AJM, Reni M, Dell’Oro S, et al. Treatment with high-dose methotrexate, vincristine and procarbazine, without intrathecal chemotherapy, followed by consolidation radiotherapy for primary central nervous system lymphoma in immunocompetent patients combined. Oncology 2001; 60(2): 134–140, http://www.ncbi.nlm.nih.gov/pubmed/11244328
55. Farber S, Diamond LK, Mercer RD, et al. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid (Aminopterin). N Engl J Med 1948; 238(23): 787–793, http://www.ncbi.nlm.nih.gov/pubmed/18860765
56. Kaye SB. New antimetabolites in cancer chemotherapy and their clinical impact. British Joumal of Cancer 1998, http://images.biomedsearch.com/9717984/brjcancer00009-0006.pdf?AWSAccessKeyId=AKIAIBOKHYOLP4MBMRGQ&Expires=1549929600&Signature=8xSuXBSQutSx1C7zk7I9HhDtJkI%3D
57. Marina N and Children’s oncology group appendix for AOST0331. A randomized trial of the European and American osteosarcoma study group to optimize treatment strategies for resectable osteosarcoma based on histological response to pre-operative chemotherapy (IND# 12697). A Phase III Intergroup Study, 2005, https://members.childrensoncologygroup.org
58. Longley DB, Harkin DP, Johnston PG. 5-Fluorouracil: mechanisms of action and clinical strategies. Nat Rev Cancer 2003; 3(5): 330–338, http://www.nature.com/articles/nrc1074
59. Nadal JC, Van Groeningen CJ, Pinedo HM, et al. In vivo potentiation of 5-fluorouracil by leucovorin in murine colon carcinoma. Biomed Pharmacother 1988; 42(6): 387–393, http://www.ncbi.nlm.nih.gov/pubmed/3064823
60. Wright JE, Dreyfuss A, el-Magharbel I, et al. Selective expansion of 5,10-methylenetetrahydrofolate pools and modulation of 5-fluorouracil antitumor activity by leucovorin in vivo. Cancer Res 1989; 49(10): 2592–2596, http://www.ncbi.nlm.nih.gov/pubmed/2785434
61. Arshad A, Isherwood J, Mann C, et al. Intravenous ω-3 fatty acids plus gemcitabine. J Parenter Enter Nutr 2017; 41(3): 398–403, http://www.ncbi.nlm.nih.gov/pubmed/26220200
62. Basourakos SP, Li L, Aparicio AM, et al. Combination platinum-based and DNA damage response-targeting cancer therapy: evolution and future directions. Curr Med Chem 2017; 24(15): 1586–1606, http://www.ncbi.nlm.nih.gov/pubmed/27978798
63. Reed E. Platinum-DNA adduct, nucleotide excision repair and platinum based anti-cancer chemotherapy. Cancer Treat Rev 1998; 24(5): 331–344, http://www.ncbi.nlm.nih.gov/pubmed/9861196
64. Hu YJ, Chen Y, Zhang YQ, et al. The protective role of selenium on the toxicity of cisplatin-contained chemotherapy regimen in cancer patients. Biol Trace Elem Res 1997; 56(3): 331–341, http://www.ncbi.nlm.nih.gov/pubmed/9197929
65. Sieja K, Talerczyk M. Selenium as an element in the treatment of ovarian cancer in women receiving chemotherapy. Gynecol Oncol 2004; 93(2): 320–327, http://www.ncbi.nlm.nih.gov/pubmed/15099940
66. Boffetta P, Nyberg F. Contribution of environmental factors to cancer risk. Br Med Bull 2003; 68: 71–94.
67. Fabiani R, Minelli L, Bertarelli G, et al. A western dietary pattern increases prostate cancer risk: a systematic review and meta-analysis. Nutrients 2016; 8(10): 626.
68. Cordain L, Eaton SB, Sebastian A, et al. Origins and evolution of the Western diet: health implications for the 21st century. Am J Clin Nutr 2005; 81: 341–354.
69. Xiao Y, Xia J, Li L, et al. Associations between dietary patterns and the risk of breast cancer: a systematic review and meta-analysis of observational studies. Breast Cancer Res 2019; 21: 16.
70. Moss A, Nalankilli K. The association between diet and colorectal cancer risk: moving beyond generalizations. Gastroenterology 2017; 152(8): 1821–1823.
71. Key TJ, Allen NE, Spencer EA, et al. The effect of diet on risk of cancer. Lancet 2002; 360: 861–868.
72. Tayyem RF, Bawadi HA, Shehadah I, et al. Dietary patterns and colorectal cancer. Clin Nutr 2017; 36: 848–852.
73. O’Flanagan CH, Smith LA, McDonell SB, et al. When less may be more: calorie restriction and response to cancer therapy. BMC Med 2017; 15(1): 106–109.
74. Hursting SD, Dunlap SM, Ford NA, et al. Calorie restriction and cancer prevention: a mechanistic perspective. Cancer Metab 2013; 1(1): 10.
75. Hursting SD, Smith SM, Lashinger LM, et al. Calories and carcinogenesis: lessons learned from 30 years of calorie restriction research. Carcinogenesis 2010; 31: 83–89.
76. Sugimura T. Nutrition and dietary carcinogens. Carcinogenesis 2000; 21: 387–395.
77. Laron Z. Insulin-like growth factor 1 (IGF-1): a growth hormone. J Clin Pathol—Mol Pathol 2001; 54: 311–316.
78. Hursting SD, Slaga TJ, Fischer SM, et al. Mechanism-based cancer prevention approaches: targets, examples, and the use of transgenic mice. J Natl Cancer Inst 1999; 91: 215–225.
79. Ikeno Y, Bronson RT, Hubbard GB, et al. Delayed occurrence of fatal neoplastic diseases in Ames dwarf mice: correlation to extended longevity. J Gerontol A Biol Sci Med Sci 2003; 58(4): 291–296.
80. Shevah O, Laron Z. Patients with congenital deficiency of IGF-I seem protected from the development of malignancies: a preliminary report. Growth Horm IGF Res 2007; 17(1): 54–57.
81. Ron E, Gridley G, Hrubec Z, et al. Acromegaly and gastrointestinal cancer. Cancer 1991; 68: 1673–1677.
82. Hursting SD, Switzer BR, French JE, et al. The growth hormone: insulin-like growth factor 1 axis is a mediator of diet restriction-induced inhibition of mononuclear cell leukemia in Fischer rats. Cancer Res 1993; 53(12): 2750–2757.
83. Chitnis MM, Yuen JSP, Protheroe AS, et al. The type 1 insulin-like growth factor receptor pathway. Clinical Cancer Res 2008; 14: 6364–6370.
84. Johnson ML, Distelmaier K, Lanza IR, et al. Mechanism by which caloric restriction improves insulin sensitivity in sedentary obese adults. Diabetes 2016; 65(1): 74–84.
85. Simone BA, Dan T, Palagani A, et al. Caloric restriction coupled with radiation decreases metastatic burden in triple negative breast cancer. Cell Cycle 2016; 15(17): 2265–2274, https://doi.org/10.1080/15384101.2016.1160982
86. Harhaji-Trajkovic L, Arsikin K, Kravic-Stevovic T, et al. Chloroquine-mediated lysosomal dysfunction enhances the anticancer effect of nutrient deprivation. Pharm Res 2012; 29(8): 2249–2263.
87. Gautron L, Elmquist JK. Sixteen years and counting: an update on leptin in energy balance. J Clin Invest 2011; 121: 2087–2093.
88. Dalamaga M, Diakopoulos KN, Mantzoros CS. The role of adiponectin in cancer: a review of current evidence. Endocr Rev 2012; 33(4): 547–594.
89. Rogozina OP, Bonorden MJL, Seppanen CN, et al. Effect of chronic and intermittent calorie restriction on serum adiponectin and leptin and mammary tumorigenesis. Cancer Prev Res (Phila) 2011; 4(4): 568–581.
90. Jarde T, Perrier S, Vasson MP, et al. Molecular mechanisms of leptin and adiponectin in breast cancer. Eur J Cancer 2011; 47(1): 33–43.
91. Gu L, Wang C, Di Cao C, et al. Association of serum leptin with breast cancer: a meta-analysis. Medicine (Baltimore) 2019; 98: e14094.
92. Tessitore L, Vizio B, Pesola D, et al. Adipocyte expression and circulating levels of leptin increase in both gynaecological and breast cancer patients. Int J Oncol 2004; 24(6): 1529–1535.
93. Yom CK, Lee KM, Han W, et al. Leptin as a potential target for estrogen receptor-positive breast cancer. J Breast Cancer 2013; 16(2): 138–145.
94. Galluzzi L, Pietrocola F, Bravo-San Pedro JM, et al. Autophagy in malignant transformation and cancer progression. EMBO J 2015; 34(7): 856–880.
95. van Niekerk G, Hattingh SM, Engelbrecht AM. Enhanced therapeutic efficacy in cancer patients by short-term fasting: the autophagy connection. Front Oncol 2016; 6: 242–247.
96. White E, Mehnert JM, Chan CS. Autophagy, metabolism, and cancer HHS public access. Clin Cancer Res 2015; 21: 5037–5046.
97. Iwaki T, Urano T, Umemura K. PAI-1, progress in understanding the clinical problem and its aetiology. Br J Haematol 2012; 157: 291–298.
98. Saharinen P, Eklund L, Pulkki K, et al. VEGF and angiopoietin signaling in tumor angiogenesis and metastasis. Trends Mol Med 2011; 17: 347–362.
99. Prete AD, Allavena P, Santoro G, et al. Molecular pathways in cancer-related inflammation. Biochem Medica 2011; 21: 264–275.
100. Divella R, De Luca R, Abbate I, et al. Obesity and cancer: the role of adipose tissue and adipo-cytokines-induced chronic inflammation. J Cancer 2016; 7: 2346–2359.
101. O’Rourke RW. Inflammation in obesity-related diseases. Surgery 2009; 145: 255–259.
102. Redman LM, Ravussin E. Caloric restriction in humans: impact on physiological, psychological, and behavioral outcomes. Antioxid Redox Signal 2011; 14: 275–287.
103. Sun L, Li Y-J, Yang X, et al. Effect of fasting therapy in chemotherapy-protection and tumor-suppression: a systematic review. Transl Cancer Res 2017; 6(2): 354–365.
104. Pollak MN. Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov 2012; 2: 778–790.
105. Zhou Y, Zhao RH, Tseng KF, et al. Sirolimus induces apoptosis and reverses multidrug resistance in human osteosarcoma cells in vitro via increasing microRNA-34b expression. Acta Pharmacol Sin 2016; 37(4): 519–529.
106. Blättler SM, Cunningham JT, Verdeguer F, et al. Yin Yang 1 deficiency in skeletal muscle protects against rapamycin-induced diabetic-like symptoms through activation of insulin/IGF signaling. Cell Metab 2012; 15(4): 505–517, http://www.ncbi.nlm.nih.gov/pubmed/22482732
107. Lee C, Raffaghello L, Brandhorst S, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med 2012; 4(124): 124ra27.
108. Raffaghello L, Lee C, Safdie FM, et al. Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci U S A 2008; 105(24): 8215–8220.
109. Naveed S, Aslam M, Ahmad A. Starvation based differential chemotherapy: a novel approach for cancer treatment. Oman Med J 2014; 29(6): 391–398.
110. Raffaghello L, Safdie F, Bianchi G, et al. Fasting and differential chemotherapy protection in patients. Cell Cycle 2010; 9(22): 4474–4476.
111. Buono R, Longo VD. Starvation, Stress Resistance, and Cancer. Trends Endocrinol Metab 2018; 29(4): 271–280.
112. Di Biase S, Longo VD. Fasting-induced differential stress sensitization in cancer treatment. Mol Cell Oncol 2016; 3(3): e1117701.
113. Henning PC, Scofield DE, Rarick KR, et al. Effects of acute caloric restriction compared to caloric balance on the temporal response of the IGF-I system. Metabolism 2013; 62(2): 179–187, http://www.ncbi.nlm.nih.gov/pubmed/22906764
114. Beauloye V, Willems B, de Coninck V, et al. Impairment of liver GH receptor signaling by fasting. Endocrinology 2002; 143(3): 792–800, http://www.ncbi.nlm.nih.gov/pubmed/11861499
115. Mihaylova MM, Shaw RJ. The AMPK signalling pathway coordinates cell growth, autophagy and metabolism. Nat Cell Biol 2011; 13(9): 1016–1023.
116. Pietrocola F, Pol J, Vacchelli E, et al. Caloric restriction mimetics enhance anticancer immunosurveillance. Cancer Cell 2016; 30(1): 147–160.
117. Safdie FM, Dorff T, Quinn D, et al. Fasting and cancer treatment in humans: a case series report. Aging (Albany NY) 2009; 1(12): 988–1007.
118. Kawaguchi T, Takemura G, Kanamori H, et al. Prior starvation mitigates acute doxorubicin cardiotoxicity through restoration of autophagy in affected cardiomyocytes. Cardiovasc Res 2012; 96(3): 456–465.
119. Caccialanza R, Cereda E, De Lorenzo F, et al. To fast, or not to fast before chemotherapy, that is the question. BMC Cancer 2018; 18(1): 10–13.
120. Weber DD, Aminazdeh-Gohari S, Kofler B. Ketogenic diet in cancer therapy. Aging 2018; 10: 164–165.
121. Vidali S, Aminzadeh S, Lambert B, et al. Mitochondria: the ketogenic diet—a metabolism-based therapy. Int J Biochem Cell Biol 2015; 63: 55–59.
122. Gasior M, Rogawski MA, Hartman AL. Neuroprotective and disease-modifying effects of the ketogenic diet. Behav Pharmacol 2006; 17: 431–439.
123. Neal EG, Chaffe H, Schwartz RH, et al. The ketogenic diet for the treatment of childhood epilepsy: a randomised controlled trial. Lancet Neurol 2008; 7(6): 500–506.
124. Evangeliou A, Vlachonikolis I, Mihailidou H, et al. Application of a ketogenic diet in children with autistic behavior: pilot study. J Child Neurol 2003; 18: 113–118.
125. Mavropoulos JC, Yancy WS, Hepburn J, et al. The effects of a low-carbohydrate, ketogenic diet on the polycystic ovary syndrome: a pilot study. Nutr Metab 2005; 2: 35.
126. Allen BG, Bhatia SK, Anderson CM, et al. Ketogenic diets as an adjuvant cancer therapy: history and potential mechanism. Redox Biol 2014; 2: 963–970.
127. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 2009; 324(5930): 1029–1033.
128. Lv M, Zhu X, Wang H, et al. Roles of caloric restriction, ketogenic diet and intermittent fasting during initiation, progression and metastasis of cancer in animal models: a systematic review and meta-analysis. PLoS ONE 2014; 9(12): e115147.
129. Klement RJ, Sweeney RA. Impact of a ketogenic diet intervention during radiotherapy on body composition: I. Initial clinical experience with six prospectively studied patients. BMC Res Notes 2016; 9: 143.
130. Aykin-Burns N, Ahmad IM, Zhu Y, et al. Increased levels of superoxide and H2O2 mediate the differential susceptibility of cancer cells versus normal cells to glucose deprivation. Biochem J 2009; 9: 29–37.
131. Warburg O. On the origin of cancer cells. Science 1956; 123: 309–314.
132. Rigo P, Paulus P, Kaschten BJ, et al. Oncological applications of positron emission tomography with fluorine-18 fluorodeoxyglucose. Eur J Nucl Med 1996; 23: 1641–1674.
133. Boros LG, Lee PWN, Brandes JL, et al. Nonoxidative pentose phosphate pathways and their direct role in ribose synthesis in tumors: is cancer a disease of cellular glucose metabolism. Med Hypotheses 1998; 50: 55–59.
134. Nebeling LC, Lerner E. Implementing a ketogenic diet based on medium-chain triglyceride oil in pediatric patients with cancer. J Am Diet Assoc 1995; 95(6): 693–697.
135. Klement RJ. Beneficial effects of ketogenic diets for cancer patients: a realist review with focus on evidence and confirmation. Med Oncol 2017; 34(8): 132–115.
136. Khodadadi S, Sobhani N, Mirshekar S, et al. Tumor cells growth and survival time with the ketogenic diet in animal models: a systematic review. Int J Prev Med 2017; 8: 35.
137. Nakamura K, Tonouchi H, Sasayama A, et al. A ketogenic formula prevents tumor progression and cancer cachexia by attenuating systemic inflammation in colon 26 tumor-bearing mice. Nutrients 2018; 10(2): E206.
138. Morscher RJ, Aminzadeh-Gohari S, Feichtinger R, et al. Inhibition of neuroblastoma tumor growth by ketogenic diet and/or calorie restriction in a CD1-nu mouse model. PLoS ONE 2015; 10(6): e0129802.
139. Mosek A, Natour H, Neufeld MY, et al. Ketogenic diet treatment in adults with refractory epilepsy: a prospective pilot study. Seizure 2009; 18(1): 30–33.
140. Westerterp-Plantenga MS, Nieuwenhuizen A, Tomé D, et al. Dietary protein, weight loss, and weight maintenance. Annu Rev Nutr 2009; 29: 21–41.
141. Bergqvist AGC, Schall JI, Stallings VA, et al. Progressive bone mineral content loss in children with intractable epilepsy treated with the ketogenic diet. Am J Clin Nutr 2008; 88(6): 1678–1684.
142. Chung H-Y, Park YK. Rationale, feasibility and acceptability of ketogenic diet for cancer treatment. J Cancer Prev 2017; 22(3): 127–134.
143. Schwingshackl L, Hoffmann G. Adherence to Mediterranean diet and risk of cancer: an updated systematic review and meta-analysis of observational studies. Cancer Med 2015; 4(12): 1933–1947.
144. Boeing H, Bechthold A, Bub A, et al. Critical review: vegetables and fruit in the prevention of chronic diseases. Eur J Nutr 2012; 51: 637–663.
145. Fardet A. New hypotheses for the health-protective mechanisms of whole-grain cereals: what is beyond fibre. Nutr Res Rev 2010; 23(1): 65–134.
146. Schwingshackl L, Lampousi AM, Portillo MP, et al. Olive oil in the prevention and management of type 2 diabetes mellitus: a systematic review and meta-analysis of cohort studies and intervention trials. Nutr Diabetes 2017; 7(4): e262.
147. Aune D, Navarro Rosenblatt DA, Chan DS, et al. Dairy products, calcium, and prostate cancer risk: a systematic review and meta-analysis of cohort studies. Am J Clin Nutr 2015; 101(1): 87–117.
148. Vieira AR, Abar L, Chan DSM, et al. Foods and beverages and colorectal cancer risk: a systematic review and meta-analysis of cohort studies, an update of the evidence of the WCRF-AICR Continuous Update Project. Ann Oncol 2017; 28: 1788–1802.
149. Cross AJ, Leitzmann MF, Gail MH, et al. A prospective study of red and processed meat intake in relation to cancer risk. PLoS Med 2007; 4(12): e325.
150. Lachenmeier DW, Rehm J. Moderate red wine drinking does not help cut women’s breast cancer risk. J Women Health 2012; 21: 469–470.
151. Lee JE, Hunter DJ, Spiegelman D, et al. Alcohol intake and renal cell cancer in a pooled analysis of 12 prospective studies. J Natl Cancer Inst 2007; 99(10): 801–810.
152. Praud D, Bertuccio P, Bosetti C, et al. Adherence to the Mediterranean diet and gastric cancer risk in Italy. Int J Cancer 2014; 134: 2935–2941.
153. Simopoulos AP. The Mediterranean Diets: what is so special about the diet of Greece? The scientific evidence. J Nutr 2001; 131(11 Suppl): 3065S–3073S.
154. Tokudome S, Nagaya T, Okuyama H, et al. Japanese versus Mediterranean diets and cancer. Asian Pac J Cancer Prev 2000; 1(1): 61–66.
155. Kagawa Y. Impact of westernization on the nutrition of Japanese: changes in physique, cancer, longevity and centenarians. Prev Med 1978; 7(2): 205–217.
156. Kolonel LN, Altshuler D, Henderson BE. The multiethnic cohort study: exploring genes, lifestyle and cancer risk. Nat Rev Cancer 2004; 4: 519–527.
157. Minami Y, Nishino Y, Tsubono Y, et al. Increase of colon and rectal cancer incidence rates in Japan: trends in incidence rates in Miyagi Prefecture, 1959-1997. J Epidemiol 2006; 16(6): 240–248.
158. Wakai K, Date C, Fukui M, et al. Dietary fiber and risk of colorectal cancer in the Japan collaborative cohort study. Cancer Epidemiol Biomarkers Prev 2007; 16(4): 668–675.
159. Hirayama T. Relationship of soybean paste soup intake to gastric cancer risk. Nutr Cancer 1981; 3: 223–233.
160. Fujiki H, Watanabe T, Sueoka E, et al. Cancer prevention with green tea and its principal constituent, EGCG: from early investigations to current focus on human cancer stem cells. Mol Cells 2018; 41: 73–82.
161. Soldati L, Di Renzo L, Jirillo E, et al. The influence of diet on anti-cancer immune responsiveness. J Transl Med 2018; 16: 75.
162. Key TJ, Appleby PN, Rosell MS. Health effects of vegetarian and vegan diets. Proc Nutr Soc 2006; 65(1): 35–41.
163. Davey GK, Spencer EA, Appleby PN, et al. EPIC–Oxford: lifestyle characteristics and nutrient intakes in a cohort of 33 883 meat-eaters and 31 546 non meat-eaters in the UK. Public Health Nutr 2003; 6(3): 259–269.
164. American Dietetic Association; Dietitians of Canada. Position of the American Dietetic Association/Dietitians of Canada: vegetarian diets. J Am Diet Assoc 2003; 103: 748–765.
165. Liu RH. Potential synergy of phytochemicals in cancer prevention: mechanism of action. J Nutr 2004; 134(12 Suppl.): 3479S–3485S.
166. Food nutrition, physical activity, the prevention of cancer: a global perspective. Washington, DC: AICR, 2008.
167. Craig WJ. Health effects of vegan diets. Am J Clin Nutr 2009; 89: 1627S–1633S.
168. Vissers MCM, Das AB. Potential mechanisms of action for vitamin C in cancer: reviewing the evidence. Front Physiol 2018; 9: 809.
169. Wang H, Oo Khor T, Shu L, et al. Plants vs. cancer: a review on natural phytochemicals in preventing and treating cancers and their druggability. Anticancer Agents Med Chem 2012; 12: 1281–1305.
170. Aykan NF. Red meat and colorectal cancer. Oncol Rev 2015; 9: 288.
171. Li J, Mao Q. Legume intake and risk of prostate cancer: a meta-analysis of prospective cohort studies. Oncotarget 2017; 8(27): 44776–44784.
172. Chan JM, Wang F, Holly EA. Pancreatic cancer, animal protein and dietary fat in a population-based study, San Francisco Bay Area, California. Cancer Causes Control 2007; 18(10): 1153–1167.
173. Lanou AJ, Svenson B. Reduced cancer risk in vegetarians: an analysis of recent reports. Cancer Manag Res 2010; 3: 1–8.

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: June 1, 2020
Issue published: January-December 2020

Keywords

  1. Diet
  2. oncology
  3. caloric restriction
  4. ketogenic diet
  5. intermittent fasting

Rights and permissions

© The Author(s) 2020.
Creative Commons License (CC BY-NC 4.0)
This article is distributed under the terms of the Creative Commons Attribution-NonCommercial 4.0 License (https://creativecommons.org/licenses/by-nc/4.0/) which permits non-commercial use, reproduction and distribution of the work without further permission provided the original work is attributed as specified on the SAGE and Open Access page (https://us.sagepub.com/en-us/nam/open-access-at-sage).
Request permissions for this article.
PubMed: 32537159

Authors

Affiliations

Ashley Gray
Division of Pediatric Hematology/Oncology, Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
Brian N Dang
David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
Theodore B Moore
Division of Pediatric Hematology/Oncology, Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
Roger Clemens
Pharmacology & Pharmaceutical Sciences, USC School of Pharmacy, International Center for Regulatory Science, Los Angeles, CA, USA
Peter Pressman
Polyscience Consulting & Director of Nutrition and Public Health, The Daedalus Foundation, San Clemente, CA, USA

Notes

Ashley Gray, Division of Pediatric Hematology/Oncology, Mattel Children’s Hospital, David Geffen School of Medicine, University of California, Los Angeles, 10833 Le Conte Ave., MDCC A2-410, Los Angeles, CA 90095, USA. Email: [email protected]

Metrics and citations

Metrics

Journals metrics

This article was published in SAGE Open Medicine.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 7805

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 21 view articles Opens in new tab

Crossref: 21

  1. Weight loss and response to chemotherapy in pediatric patients with os...
    Go to citation Crossref Google Scholar
  2. Game-theoretical description of the go-or-grow dichotomy in tumor deve...
    Go to citation Crossref Google Scholar
  3. Dietary approaches for controlling cancer by limiting the Warburg effe...
    Go to citation Crossref Google Scholar
  4. Ketogenic Diet and Multiple Health Outcomes: An Umbrella Review of Met...
    Go to citation Crossref Google Scholar
  5. Ketogenic diet and metastasis: A critical review of the literature and...
    Go to citation Crossref Google Scholar
  6. The beneficial and adverse effects of autophagic response to caloric r...
    Go to citation Crossref Google Scholar
  7. Survival pattern of colorectal cancer in Sub-Saharan Africa: A systema...
    Go to citation Crossref Google Scholar
  8. Do Cancer and Cancer Treatments Accelerate Aging?
    Go to citation Crossref Google Scholar
  9. Critical Review on Fatty Acid-Based Food and Nutraceuticals as Support...
    Go to citation Crossref Google Scholar
  10. Avenues of research in dietary interventions to target tumor metabolis...
    Go to citation Crossref Google Scholar
  11. Mitigation of Iron Irradiation-Induced Genotoxicity and Genomic Instab...
    Go to citation Crossref Google Scholar
  12. Metabolic reprogramming in prostate cancer
    Go to citation Crossref Google Scholar
  13. Ketogenic Diet for Cancer: Critical Assessment and Research Recommenda...
    Go to citation Crossref Google Scholar
  14. Ketogenic Diet in Cancer Prevention and Therapy: Molecular Targets and...
    Go to citation Crossref Google Scholar
  15. Ketogenic Diets in Pancreatic Cancer and Associated Cachexia: Cellular...
    Go to citation Crossref Google Scholar
  16. Beneficial Diets and Pancreatic Cancer: Molecular Mechanisms and Clini...
    Go to citation Crossref Google Scholar
  17. Magnesium: Biochemistry, Nutrition, Detection, and Social Impact of Di...
    Go to citation Crossref Google Scholar
  18. Methanolic Fenugreek Seed Extract Induces p53-Dependent Mitotic Catast...
    Go to citation Crossref Google Scholar
  19. Progress on the Mechanism for Aspirin’s Anti-tumor Effects
    Go to citation Crossref Google Scholar
  20. High-Fat Diet and Feeding Regime Impairs Number, Phenotype, and Cytoto...
    Go to citation Crossref Google Scholar
  21. Pediatric hematopoietic cell transplantation: Longitudinal trends in b...
    Go to citation Crossref Google Scholar

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:


Alternatively, view purchase options below:

Access journal content via a DeepDyve subscription or find out more about this option.