Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Sep 7.
Published in final edited form as: Recent Results Cancer Res. 2016;207:241–266. doi: 10.1007/978-3-319-42118-6_12

Fasting and Caloric Restriction in Cancer Prevention and Treatment

Sebastian Brandhorst 1, Valter D Longo 2,3
PMCID: PMC7476366  NIHMSID: NIHMS1620321  PMID: 27557543

Abstract

Cancer is the second leading cause of death in the USA and among the leading major diseases in the world. It is anticipated to continue to increase because of the growth of the aging population and prevalence of risk factors such as obesity, smoking, and/or poor dietary habits. Cancer treatment has remained relatively similar during the past 30 years with chemotherapy and/or radiotherapy in combination with surgery remaining the standard therapies although novel therapies are slowly replacing or complementing the standard ones. According to the American Cancer Society, the dietary recommendation for cancer patients receiving chemotherapy is to increase calorie and protein intake. In addition, there are no clear guidelines on the type of nutrition that could have a major impact on cancer incidence. Yet, various forms of reduced caloric intake such as calorie restriction (CR) or fasting demonstrate a wide range of beneficial effects able to help prevent malignancies and increase the efficacy of cancer therapies. Whereas chronic CR provides both beneficial and detrimental effects as well as major compliance challenges, periodic fasting (PF), fasting-mimicking diets (FMDs), and dietary restriction (DR) without a reduction in calories are emerging as interventions with the potential to be widely used to prevent and treat cancer. Here, we review preclinical and preliminary clinical studies on dietary restriction and fasting and their role in inducing cellular protection and chemotherapy resistance.

1. Introduction

In 2012, an estimated 14.1 million new cases of cancer occurred and approximately 8.2 million people died of cancer worldwide (Torre et al. 2015), although the exact number can only be estimated because incidence rates and treatment modalities for some parts of the world are not fully established. In the USA, one out of every four deaths was estimated to be due to cancer in 2012 (Siegel et al. 2012). The worldwide number of deaths is projected to increase to 13.2 million by 2030 due to the expected increase in the elderly population, as well as the adoption to cancer-causing behaviors (e.g., cigarette smoking) (Brawley 2011). Notably up to 35 % of all cancer deaths worldwide have been reported to be avoidable through adjustments to lifestyle and environmental factors, such as physical activity and dietary habits (Danaei et al. 2005). However, this estimate does not take into account the more recent advances in dietary interventions to affect aging and age-related diseases, which are likely to cause a major increase in the percentage of preventable cancers (Levine et al. 2014).

Compared to the improvements in the prevention and treatment of heart disease, cancer treatment and mortality have remained relatively unchanged over the past 30 years (Jemal et al. 2008). Chemotherapy and/or radiotherapy in combination with surgical removal of the tumor mass remain the standard therapies. Chemotherapy improves the survival rates of cancer patients but causes damage to normal tissues and leads to significant side effects such as emotional distress, myelo-suppression, fatigue, vomiting, diarrhea, and even death (Love et al. 1989; Partridge et al. 2001). Despite newly designed drugs that target specific cancer markers, cytotoxic drugs will have accompanying side effects unless novel and complimentary interventions or strategies are adopted. In the following sections, we discuss preclinical and preliminary clinical results on how specific forms of dietary restriction (DR) and periodic fasting (PF; 24 h or more) can have significant impact on the prevention and treatment of cancer while minimizing the adverse effects and limitations associated with chronic calorie restriction (CR).

The effects of CR on cancer prevention were first described more than 100 years ago (Moreschi 1909) and was followed by a large body of work demonstrating its tumor preventive effects in various animal models. However, recent studies in both mice and monkeys indicate that the effects of CR on longevity are either limited or absent and, in mice, CR can even reduce longevity (Colman et al. 2009; Mattison et al. 2012; Goodrick et al. 1990; Liao et al. 2010). The role of CR in cancer treatment is even more problematic considering that it would be extremely difficult to chronically restrict calorie intake in cancer patients. PF instead has been demonstrated to be effective in reducing pre-neoplastic lesions (Grasl-Kraupp et al. 1994) and in both cancer prevention and treatment, particularly in combination with cytotoxic drugs (Grasl-Kraupp et al. 1994; Brandhorst et al. 2015; Lee et al. 2012). PF protects normal cells/tissues from chemotherapy-induced toxicity (Lee et al. 2010; Raffaghello et al. 2008) while simultaneously increasing its therapeutic efficacy on a wide variety of malignant cells (Lee et al. 2012; Safdie et al. 2012). Newly designed dietary compositions aimed at inducing fasting-like effects that allow nourishment are slowly emerging as potential therapies to delay aging-associated diseases such as cancer (Brandhorst et al. 2015). These fasting-mimicking diets (FMD) and PF also promote multisystem regeneration and rejuvenation by promoting the self-renewal of stem cells and the generation of white blood cells (Brandhorst et al. 2015; Cheng et al. 2014). Thus, novel and periodic forms of extreme DR as well as targeted reductions in specific macronutrients, and a combination of both, are likely to replace the original balanced and chronic restriction of all calorie sources.

2. Conserved Role of Nutrient-Sensing Signaling Pathways in Life span and Stress Resistance

Fluctuations in food availability force most organisms into periods of drastically reduced caloric intake or even starvation. Whereas a sufficient nutrient supply enables growth and proliferation, periods of very low food availability, or the lack of specific macromolecules, activate alternative metabolic modes allowing organisms to remain protected against damage that could negatively affect fitness (Madia et al. 2009; Harrison and Archer 1989). High cellular growth rates and high stress protection rarely coexist, indicating that cells invest energy in either one or the other. In fact, when nutrients are abundant, the activation of nutrient-sensing signaling cascades promotes cellular growth while when they are scarce the down-regulation of these signaling pathways blocks cellular proliferation and activates stress resistance transcription factors which negatively regulate these pro-aging pathways (Fontana et al. 2010; Longo 1999; Guarente and Kenyon 2000; Kenyon 2001; Longo and Finch 2003). Over the last 20 years, diet-based approaches have been combined with genetic approaches to identify the genes and pathways that mediate nutrient-dependent effects on longevity and health span.

The most studied form of diet-based antiaging interventions is CR, which commonly describes a 20–40 % reduction in calorie intake. DR instead can also refer to restrictions of particular macromolecular components of the diet, without affecting daily calorie intake (e.g., low protein compensated with high fat). Alternate-day fasting (ADF), also often described as intermittent fasting (IF), is another well-studied and beneficial form of DR discussed in detail elsewhere. Finally, PF is distinct from ADF/IF as meal frequency is interrupted and caloric intake drastically reduced for periods generally ranging from 48 h to 3 weeks. In addition to CR, some forms of IF and PF have been shown to promote stress resistance and longevity in model organisms ranging from unicellular yeast to mammals, indicating that the molecular mechanisms responsible for the protective effects of CR have likely evolved billions of years ago and are partially conserved in many species (Fontana et al. 2010; Longo and Mattson 2014).

In the prokaryote E. coli, lack of glucose or nitrogen (comparable to protein restriction in mammals) increases the resistance to high levels of hydrogen peroxide (Jenkins et al. 1988). The complete lack of nutrients also extends the longevity of bacteria, which can be reversed by adding to the medium various nutrients except for acetate, a carbon source associated with starvation conditions (Gonidakis et al. 2010). In the yeast S. cerevisiae, cells switched from standard growth medium to water display a twofold increase in chronological life span and a major increase in resistance against oxidative insults and heat stress (Lee et al. 2012; Fabrizio and Longo 2003). Reducing the concentration of glucose from 2 to 0.5 % in the growth media can have similar, although less efficient effects on stress resistance (Longo et al. 1997; Wei et al. 2008). PF in yeast, which is implemented by switching yeast cells back and forth from nutrient-rich medium to water every 48 h, extends both medium and maximum longevity and increases the number of yeast cells surviving hydrogen peroxide treatment by more than 100-fold (Brandhorst et al. 2015). Although the precise mechanisms of DR-dependent life span extension in yeast are not yet fully understood, they include the down-regulation of the amino acid response Tor-S6K (Sch9) pathway and of the glucose-responsive Ras/adenylate cyclase(AC)/PKA pathway, and the activation of the serine threonine kinase Rim15 which increases stress resistance against oxidants, genotoxins, and heat shock through genes including superoxide dismutases and heat-shock proteins controlled by the transcription factors Msn2, Msn4, and Gis1 (Madia et al. 2009; Longo and Finch 2003; Fabrizio et al. 2003; Fabrizio et al. 2001). Deletion of these transcription factors reverses the protective effects of glucose restriction, demonstrating their requirement for maximum protection (Wei et al. 2008). Surprisingly, the deletion of Rim15, or of its downstream stress response transcription factors Msn2/4 and Gis1, does not prevent the life span effects of PF (Brandhorst et al. 2015). These results indicate that PF can protect simple organisms from toxins and aging by mechanisms that are in part independent of conserved pro-longevity transcription factors.

The protective effects of food restriction/starvation on the unicellular E. coli and S. cerevisiae are also observed in multicellular organisms. In the fruit fly D. melanogaster, dilution or reduction of food have been shown to extend life span, although intermittent food deprivation does not (Grandison et al. 2009; Piper and Partridge 2007). The fasting-induced protection against oxidative stress in flies is mediated by the repression of translation (consistent with energy diversion from cellular growth to protection) through increased expression of d4E-BP downstream of the PI3K/Akt/dFOXO3 pathway (Tettweiler et al. 2005; Villa-Cuesta et al. 2010). Of note is that moderate DR does not protect flies against the oxidative damage caused by re-oxygenation injury whereas a more stringent DR does (Vigne and Frelin 2007). Mutations in the insulin receptor substrate chico extend life span in D. melanogaster although its role in stress resistance remains unclear (Clancy et al. 2001; Giannakou and Partridge 2007). In the nematode C. elegans, CR and fasting, achieved by feeding either reduced amounts or no bacteria, also increase life span (Smith et al. 2008). Life span extension in C. elegans requires AMPK, a regulator of cellular glucose uptake and β-oxidation of fatty acids, as well as the stress resistance transcription factor DAF-16, analogous to Msn2/4 and Gis1 in yeast and FOXOs in D. melanogaster and mammals (Greer et al. 2007). Fasting every other day also increases oxidative stress resistance in C. elegans and increases life span by up to 56 % via modulation of the RHEB-1 and TOR signaling pathway, which are linked to DAF-16 (Weinkove et al. 2006; Honjoh et al. 2009). Conversely, excessive glucose shortens the life span of C. elegans, in part by decreasing DAF-16 activity (Lee et al. 2009). In adult C. elegans, mutations in age-1 (PI3K homolog) and daf-2 (insulin/IGF-1 receptor homolog) result in a 65–100 % life span extension by decreasing AKT-1/AKT-2 signaling, and by activating DAF-16, which is also associated with resistance to oxidative and ER stress (Johnson 1990; Paradis et al. 1999; Hsu et al. 2003; Henis-Korenblit et al. 2010).

Orthologues of the genes regulating life span and stress resistance in S. cerevisiae and C. elegans downstream of the growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis regulate stress resistance and/or life span in mice (Coschigano et al. 2003; Holzenberger et al. 2003; Bonkowski et al. 2009; Selman et al. 2009; Brown-Borg 2009). In laboratory mice, mutations in the insulin-GH/IGF-1 axis increase life span by up to 150 % and conversely, mice that overexpress GH have a shortened life span (Brown-Borg et al. 1996; Murakami 2006; Bartke et al. 2002). In agreement with the findings in lower eukaryotes, cultured cells derived from long-lived mice with deficiencies in the GH/IGF-1 axis have higher resistance against H2O2-induced oxidative stress, UV, genotoxins, as well as heat- and cadmium-induced stress (Murakami 2006; Salmon et al. 2005). Vice versa, activities of the antioxidant enzymes superoxide dismutases and catalase decrease in murine hepatocytes exposed to GH or IGF-1 and in transgenic mice overexpressing GH (Brown-Borg and Rakoczy 2000; Brown-Borg et al. 2002). IGF-1 sensitizes primary neurons to oxidative stress through Ras/Erk-dependent mechanisms (Li et al. 2008), and experiments in rat primary glia and mouse fibroblasts suggest that IGF-1 exposure sensitizes these cell types against oxidative damage and chemotherapeutic drugs (Lee et al. 2010). In addition, IGF-1 attenuates the cellular stress response and the expression of stress response proteins HSP72 and heme-oxygenase in rats (Sharma et al. 2000).

In mammals, various forms of partial or complete food deprivation have been investigated that range from daily 20–40 % CR, intermittent fasting (IF, including alternate-day fasting, ADF), and PF. The multisystemic benefits of CR on rodent life span have been extensively studied and are beyond the scope of this review. Generally, CR increases rodent life span by up to 60 % and delays the occurrence of many chronic diseases including cancer and improves stress resistance (Koubova and Guarente 2003; Mattson 2005; Mattson and Wan 2005). However, there is variability in the effects of CR and some genetic backgrounds do not experience a life span extension and can even display reduced longevity (Liao et al. 2010; Sohal and Forster 2014), likely because of trade-offs between effects on aging and effects on systems which may benefit from a higher calorie intake and/or fat level (Rikke et al. 2010). In mice, the longevity effects of CR involve reduced activity of the GHR/IGF-1 pathways since CR does not further extend the life span of GH signaling-deficient mice (Bonkowski et al. 2009; Arum et al. 2009). CR is associated with reduced oxidative stress and cellular proliferation (Youngman 1993; Sohal and Weindruch 1996) while enhancing DNA repair processes and autophagy (Weraarchakul et al. 1989; Cuervo et al. 2005; Wohlgemuth et al. 2007). CR effectively reduces the levels of plasma insulin, cholesterol, triglycerides, growth factors such as IGF-1, and inflammatory cytokines (Mahoney et al. 2006; Matsuzaki et al. 2001). CR also elevates plasma high-density lipoprotein levels, resulting in a reduced risk for atherosclerosis, diabetes, and obesity (Paoletti et al. 2006; Larson-Meyer et al. 2008). In a mouse model of DR-induced stress resistance, restriction of sulfur amino acids increased hydrogen sulfide production and the protection from oxidative damage induced by hepatic reperfusion injury (Hine et al. 2015). Of note, a severe restriction of dietary protein can extend the life span of rodents by up to 20 %, independently of the caloric intake (Pamplona and Barja 2006). Reduced levels of serum IGF-1 in rats and mice fed with protein-restricted diets might explain the beneficial effects on longevity (Sonntag et al. 1999; Brandhorst et al. 2013). Reducing protein intake and IGF-1 signaling also significantly reduces the incidence and progression of breast cancer and melanoma in murine models of tumor progression (Levine et al. 2014).

In rodents, the most studied fasting method has been IF which promotes protection against multiple diseases and causes a major reduction in the incidence of lymphomas (Goodrick et al. 1990; Mattson and Wan 2005; Descamps et al. 2005; Mattson 2012). The effects of alternate-day fasting on longevity in rodents depend on the species and age at diet initiation and can range from negative effects to as much as an 80 % life span extension (Goodrick et al. 1990; Arum et al. 2009; Goodrick et al. 1982). Similar to CR, IF increases the survivorship and improves insulin sensitivity of male wild-type mice, but fails to affect either parameter in GHR-KO mice (Arum et al. 2009). The major differences between IF and PF in mice are the duration and frequency of the fast. IF cycles usually last 24 h and are separated by one day of normal food intake (alternate-day fasting), whereas PF cycles last 2 or more days and are at least 1 week apart to allow for regaining of normal weight (Longo and Mattson 2014). Additional differences include the molecular changes caused by these fasting regimes on a variety of growth factors and metabolic markers, since IF causes more frequent but less pronounced changes than PF. Although less studied than IF, PF has been shown to have potent effects on cellular protection. PF for 48–60 h prior to etoposide induced lethal oxidative stress causes a significant increase in survival in three different mouse strains (Raffaghello et al. 2008). Similarly, 72 h of food deprivation before exposure to lethal doses of the chemotherapeutic drug doxorubicin, which causes oxidative stress-induced cardiotoxicity, protects CD-1 mice (Lee et al. 2010). Following ischemic reperfusion, which is associated with the production of reactive oxygen species in the affected tissue, fasting demonstrates protective effects in the mouse kidney and liver, rat brain as well as human liver against reperfusion injury (Mitchell et al. 2009; van Ginhoven et al. 2009; Verweij et al. 2011). Traumatic brain injury followed by fasting has neuroprotective effects, reduces oxidative damage, and improves cognitive function (Davis et al. 2008).

Despite its advantages, the extreme nature of prolonged water-only fasting could cause adverse effects, such as the exacerbation of previous malnourishments and dysfunctions. These concerns are now being addressed through the implementation of newly designed dietary interventions aimed to induce PF-like effects while minimizing the risk of adverse effects and the burden of complete food restriction. In C57Bl/6 mice, a 4-day-long diet that mimics the effects offasting (fasting-mimicking diet, FMD) has been shown to lower blood glucose levels by 40 % and IGF-1 by 45 %, while increasing ketone bodies ninefold and IGFBP-1, which inhibits IGF-1, eightfold by the end of the FMD. The FMD extends health span and longevity, promotes hippocampal neurogenesis, lowers visceral fat, reduces skin lesions, rejuvenates the immune system, and retards bone mineral density loss in old mice (Brandhorst et al. 2015). The FMD started at middle age reduces tumor incidence, delays cancer onset, and causes a major reduction in the number of lesions, which may reflect a general switch from malignant to benign tumors.

Together, results from the range of studies and organisms described above indicate that enhanced stress resistance is a highly conserved phenotype of starved and long-lived organisms, which is in part mediated by reduced signaling through the GH and IGF-1 axis.

3. The Physiological Response to Fasting in Mammals

Mammals undergo distinct metabolic changes when deprived of food that are different from that of CR (Wang et al. 2006; Lee and Longo 2011). The post-absorptive phase, lasting for 10 or more hours after food ingestion involves the utilization of glycogen as the main stored energy source. Following the depletion of the hepatic glycogen storage, amino acids serve as gluconeogenic substrates while the brain consumes most of the remaining glucose. After about 72 h of fasting, serum glucose levels can reach 50–60 mg/dL in a healthy person, but return to normal levels within 30 min after oral administration of 100 g glucose (Unger et al. 1963). Glycerol and fatty acids are released from the adipose tissue and as a result, acetoacetate, β-hydroxybutyrate and acetone become the main carbon sources within a few days of fasting. During the last phase of prolonged food deprivation, the fat storage is eventually exhausted and muscles are being degraded to allow gluconeogenesis. In rats, this occurs after 4–5 days but does not result in a major increase in glucose levels (Wang et al. 2006). For humans, it is estimated that a 70-kg person can obtain basal caloric requirements from fat reserves for up to 2–3 months of fasting (Cahill and Owen 1968; Cahill et al. 1968), leading to an initially rapid weight loss that subsequently tapers off. During the first week of fasting, an average of 0.9 kg/day is lost, followed by a 0.3-kg/day weight loss by the third week of fasting, resulting in an approximately 20 % body weight loss after 30–35 days of fasting (Kerndt et al. 1982). Thus, PF is generally feasible and tolerable, but may be accompanied by side effects, such as headaches, light-headedness, nausea, weakness, edema, anemia, and amenorrhea (Bloom 1959; Runcie and Thomson 1970; Drenick and Smith 1964).

In accordance with the systemic effects of fasting, a plethora of extrinsic and intrinsic growth factors is affected, including mediators of evolutionarily conserved pathways that promote stress sensitivity and aging, such as IGF-1 (Fontana et al. 2010; Longo and Mattson 2014). IGF-1 levels decrease by up to 65 % following 120 h of PF in humans despite increased GH secretion (Ketelslegers et al. 1995; Underwood et al. 1994; Moller and Jorgensen 2009; Maccario et al. 2001) due to the increase in IGFBP-1, which decreases IGF-1 bioavailability and prevents the feedback inhibition of GH secretion by IGF-1 (Zapf et al. 1995). In humans, GH levels eventually decrease after 3–10 days of fasting and level off (Merimee and Fineberg 1974; Palmblad et al. 1977). In mice, PF (24–72 h) decreases IGF-1 level by 70 % and is associated with a 11-fold increase in IGFBP-1 (Lee et al. 2010; Tannenbaum et al. 1979; Frystyk et al. 1999), reduced protein synthesis, reduced AKT activity, reduced mTOR/S6K, increased 4E-BP1 activity, and increased FOXO-1, FOXO-3, and FOXO-4 (Sans et al. 2004; Imae et al. 2003).

Starving mice or mammalian cells triggers autophagy in various tissues, which is regulated by several genes that also regulate aging and stress resistance, such as AMPK, mTOR, and sirtuins (Kroemer et al. 2010; Marino et al. 2010; Morselli et al. 2009). Oncogenic signaling, e.g., through PI3K and Akt, has been shown to inhibit autophagy, whereas tumor suppressors such as PTEN and TSC2 can trigger autophagy (Morselli et al. 2009). Autophagy is elevated in many cancer cells and increases the resistance of cancer cells to chemotherapy (Morselli et al. 2009). Increased autophagy has been hypothesized to be mediated by ammonia as a by-product of glutaminolysis occurring in the mitochondria of malignant cells undergoing the Warburg effect to supply biosynthetic precursors required for their high proliferation rate (Vander Heiden et al. 2009). Therefore, autophagy triggered by fasting may be beneficial to normal cells but detrimental to malignant cells.

4. DR, Fasting and Cancer Treatment

Tumors arise over time from the combination of DNA damage and mutations associated with changes in the environmental niche surrounding precancerous cells (Hanahan and Weinberg 2011). Not surprisingly, aging is the major risk factor for the development of many cancer types (DePinho 2000; Campisi et al. 2011). Studies on caloric intake that delay organismal aging emphasize the preventive role of CR in cancer establishment. Over a century ago, the first direct relationship between chronic CR and the prevention of tumor transplantation in mice was demonstrated (Moreschi 1909). Subsequently, a multitude of studies has established that CR reduces the progression of spontaneous or induced tumors in various animal models (Tannenbaum 1996; Tannenbaum 1940; Berrigan et al. 2002; Hursting et al. 1994). Further, CR has also been shown to reduce the cancer incidence in rodents. Roy Walford and Richard Weindruch reported that CR beginning either at the time of weaning or at 12 months of age increased life span, delayed immunologic aging in laboratory mice, and reduced spontaneous cancer incidence by more than 50 % (Weindruch and Walford 1982; Weindruch et al. 1986). In a multitude of auto- and xenograft tumor models, CR causes potent antigrowth effects, although some cell lines remain resistant. Resistance to CR has been associated with mutations that cause the constitutive activation of the phosphatidylinositol-3-kinase (PI3K) pathway; substitution of the mutant allele of PI3K with wild-type PI3K reestablishes a DR-sensitive cancer cell (Kalaany and Sabatini 2009).

Support for the potential application of CR in humans arose from a 20-year longitudinal adult-onset 30 % CR study in rhesus monkeys which decreased cancer incidence by 50 % and reduced mortality (Colman et al. 2009). These findings, however, have to be analyzed carefully as a CR regimen implemented in young and older age rhesus monkeys at the National Institute on Aging (NIA) has not improved survival outcomes (Mattison et al. 2012). Whether or not CR has the potential to reduce cancer incidence in humans remains largely unclear, although CR can reduce clinical markers associated with cancer if it also involves protein restriction (Longo and Fontana 2010). Despite data indicating benefits in cancer prevention at least in certain mouse genetic backgrounds, the use of CR in therapy is problematic in part because it would be extremely difficult to chronically restrict cancer patients even if a benefit was demonstrated. Further, chronic CR merely delays the progression of the disease (Mukherjee et al. 2004; Bonorden et al. 2009), and its effectiveness might be reduced to a subset of CR-responsive cancers (Kalaany and Sabatini 2009). In addition, in mice chronic CR is associated with weight loss, delayed wound healing, and impaired immune function, all of which may impose a significant risk to cancer patients receiving chemotherapy, surgery or immunity-based treatments, or who are at risk for losing weight and becoming frail and cachectic (Fontana et al. 2010; Kristan 2008; Reed et al. 1996; Kim and Demetri 1996).

Data suggest that PF (mice for 48–72 h, humans 4–5 days) may induce metabolic changes which delay the growth of damaged cells, without the negative side effects and limitations associated with chronic CR. In rats, fasting for 8 days or 40 % CR for 3 months, reduces pre-neoplastic liver masses by 20–30 %. Putative pre-neoplastic liver foci have significantly lowered DNA replications but increased apoptosis during PF, resulting in the reduced number and volume of putative pre-neoplastic liver foci by 85 % throughout the following 17 months (Grasl-Kraupp et al. 1994). However, in contrast to the protective effect of chronic CR on carcinogenesis (Tagliaferro et al. 1996), fasting followed by refeeding sustained tumor initiation in the rat liver enhances the growth of aberrant crypt foci in the colorectal mucosa and mammary tumors even by otherwise non-initiating carcinogen doses (Premoselli et al. 1998; Sesca et al. 1998; Tessitore et al. 1999). Given that the refeeding phase is associated with increased cellular proliferation in the liver and colorectal epithelium (Brandhorst et al. 2015; Cuervo et al. 2005), exposure to carcinogens during this period might explain these effects. Therefore, it is of importance to consider that normal food intake following PF should be initiated once the half-life of potential carcinogens indicates its expiration. Despite these caveats, PF has been shown to have potent protective effects in cancer treatment, in part due its impact on host metabolism. PF decreases blood glucose in mice by up to 75 % whereas long-term CR or IF cause a 15 % reduction (Longo and Mattson 2014; Lee and Longo 2011); this seems of particular importance given the glucose dependent phenotype of many cancer cells (Vander Heiden et al. 2009; Warburg 1956). PF reduces the pro-proliferative growth factor IGF-1 by up to 75 % (Lee et al. 2010; Underwood et al. 1994), while chronic CR causes a 25 % IGF-1 reduction in mice (Barger et al. 2008) and has no impact on IGF-1 levels in humans unless combined with protein restriction (Fontana et al. 2008). Furthermore, chronic DR with protein restriction only causes a 30 % reduction in IGF-1 for humans (Fontana et al. 2008).

5. Differential Stress Resistance by Fasting

As described above, PF inactivates partially conserved pro-proliferative pathways and increases resistance to oxidative and other stresses in lower eukaryotes, whereas the constitutive activation of analogous pathways play central roles in promoting cancer. Ras and Akt function in signal transduction pathways that are frequently found in a constitutively activated form in cancer cells (Kinzler and Vogelstein 1996; Hanahan and Weinberg 2000; Medema and Bos 1993). The connection between (de-regulated) cellular proliferation and stress resistance provides the theoretical foundation for a fasting-induced differential protection of normal cells from chemotoxicity (differential stress resistance, DSR). DSR is based on the hypothesis that in response to fasting, normal cells enter a stress-resistant state (Raffaghello et al. 2008; Longo et al. 1997) characterized by reduced/lack of cell divisions and the utilization of metabolites generated from the breakdown of fats, proteins and organelles. Self-sufficiency in growth signals enabled by gain-of-function mutations in oncogenes (e.g., Ras, Akt, mTOR) grants constitutive activation of proliferation pathways and together with loss-of-function mutations in tumor-suppressor genes (e.g., Rb, p53, PTEN) allows cancer cells to disregard antiproliferative signals such as those occurring during fasting, thereby prohibiting malignant cells to enter a nondividing and protected state (Hanahan and Weinberg 2000). The inability of cancer cells to respond to antigrowth signals or to grow in the absence of growth factors, a well-established hallmark of cancer, prevents the protective effect of fasting regardless of the type of cancer or oncogene mutations (Lee and Longo 2011). DSR therefore presents a method to preferentially target malignant cells with chemotherapy and other drugs (Raffaghello et al. 2008) while increasing the resistance against cytotoxic stressors through cell detoxification systems in normal dividing and/or nondividing cells. In fact, fasting induces the protection against lethal doses of the chemotherapeutic drug etoposide in A/J, CD-1, and athymic nude mice (Raffaghello et al. 2008). In a different set of experiments, CD-1 mice were protected from high-dose doxorubicin, known to cause cardiotoxicity (Lee et al. 2010). Analogously, transgenic mice with a conditional liver-specific IGF-1 gene deletion (LID), which results in a 70–80 % reduction in circulating IGF-1 levels, show enhanced protection to cytotoxic chemotherapy drugs including cyclophosphamide, doxorubicin, and 5-FU, although they are not protected against the topoisomerase inhibitor etoposide (Lee et al. 2010). Studies in mice demonstrate that restoring IGF-1 to normal levels during fasting reverses the protection against lethal doses of doxorubicin (Lee et al. 2010). Similarly, three days of fasting protects FabplCre;Apc15lox/+ mice, which spontaneously develop intestinal tumors, against the side effects of a high dose of irinotecan whereas ad libitum-fed mice experience weight loss, reduced activity, ruffled coat, hunched-back posture, diarrhea, and leukopenia (Huisman et al. 2015). Fasting reduces the delayed-type chemotherapy-induced nausea and vomiting in cancer-bearing dogs receiving doxorubicin (Withers et al. 2014). The protective effect of fasting can promote potent changes even to the stem cell population: PF represses the immunosuppression and mortality caused by cyclophosphamide through signal transduction changes in long-term hematopoietic stem cells and niche cells that promote stress resistance, self-renewal, and lineage-balanced regeneration (Cheng et al. 2014), in agreement with preliminary data on the protection of lymphocytes from chemotoxicity in fasting patients (Safdie et al. 2009). Fasting has been shown to also promote regenerative effects in both the blood and other systems including the nervous system, muscle and liver indicating that, in addition to protecting against the toxicity of cancer drugs, it can stimulate the generation of healthy cells and tissue damaged by the therapy (Brandhorst et al. 2015; Cheng et al. 2014).

6. Differential Stress Sensitization of Cancer Cells by Fasting

Malignant cells are generated in a high nourishment environment and, therefore, thrive in environments resembling those in which they have evolved. Cancer cells generally prefer high levels of glucose to rely on glycolysis more than on oxidative phosphorylation (Warburg effect), which provides energy and biosynthetic precursors essential for proliferation (Vander Heiden et al. 2009). Glycolysis allows the metabolism of glucose-6-phosphate in the pentose shunt pathway, which provides substrates essential for nucleotide synthesis and NADPH production. The Warburg effect may restrict cytochrome c-mediated apoptosis, thereby favoring tumor cell survival and apoptosis evasion through decreased respiration (Vaughn and Deshmukh 2008; Ruckenstuhl et al. 2009). However, glucose alone does not supply all the building blocks required for cellular proliferation and cancer cells therefore depend on amino acids, particularly glutamine, as a nitrogen source (Vander Heiden et al. 2009). Glutamine, the circulating amino acid with the highest concentration in humans, plays a crucial part in the uptake of essential amino acids and can support NADPH production, making it necessary for lipid and nucleotide biosynthesis (Nicklin et al. 2009). Similarly to the effects of CR, protein/amino acid restriction have been demonstrated to increase longevity and to delay the onset of many aging-related diseases, including cancer (Mirzaei et al. 2014). The major reduction in circulating glucose and amino acids during fasting is a significant disadvantage to tumor cells that usually experience an almost unlimited supply through the bloodstream.

The same mutations that cause tumor cells to remain locked in a pro-growth mode render them also sensitive to alterations in the cellular environment. In evolutionary biology, it is well established that most acquired mutations become disadvantageous. The mutations generated in cancer cells are also mostly deleterious although such negative effects may not be observable until the cancer cells are in an environment that exposes that detrimental effect. For example, a mutation that requires high levels of glutamine would not be problematic to cancer cells until the concentration of glutamine becomes limited. In S. cerevisiae, expression of the oncogene-like RAS2val19 not only reverses the (water-) starvation induced protection against hydrogen peroxide and menadione, the constitutive activation of Ras sensitizes yeast cells compared to wild-type cells, a phenomenon called differential stress sensitization (DSS) (Lee et al. 2012).

In vitro models of fasting (by reducing glucose and/or serum availability in the growth medium) sensitize murine, rat and human glioblastoma multiforme cells, but not primary mixed glia, to temozolomide chemotherapy (Safdie et al. 2012). Similarly, fasting sensitizes 15 of 17 human and rodent cancer cell lines to the chemotherapeutic agents’ doxorubicin and/or cyclophosphamide (Lee et al. 2012), while serum starvation alone is sufficient to induce sensitization to cisplatin in human mesothelioma and lung carcinoma cells (Shi et al. 2012). In a metastatic mouse neuroblastoma model, fasting for 48 h followed by a single administration of high-dose chemotherapy, but not either treatment alone, successfully improves survival limited by both drug toxicity and metastases and results in long-term cancer-free survival (Lee et al. 2012). In subcutaneous mouse models of melanoma and breast cancer, fasting cycles are as effective as chemotherapy alone while the combination of both treatment modalities significantly improves treatment efficacy (Lee et al. 2012). Similarly, fasting prior to gemcitabine injection significant decreases the progression of pancreatic cancer tumors by more than 40 %, in part through increased gemcitabine uptake of malignant cells (D’Aronzo et al. 2015). PF in combination with cisplatin for three weeks reduces the progression of mesothelioma by more than 60 % compared to the untreated control and a complete remission was observed in 60 % of the combination-treated mice (Shi et al. 2012). 48 h of PF also sensitizes both subcutaneous and intracranial glioma models to radio- and chemotherapy (Safdie et al. 2012), an effect not mimicked by dietary protein restriction alone (Brandhorst et al. 2013). 48 h of starvation, with or without oxaliplatin, reduces the progression of CT26 colorectal tumors through the down-regulation of aerobic glycolysis and glutaminolysis, while increasing oxidative phosphorylation in complex I and II of the mitochondrial electron transport chain, thereby resulting in reduced ATP production, increased oxidative stress, and apoptosis (Bianchi et al. 2015). In murine 4T1 breast cancer cells, PF increases the phosphorylation of the stress-sensitizing Akt and S6 kinases and is associated with increased oxidative stress, caspase-3 cleavage, DNA damage, and subsequent apoptosis (Lee et al. 2012), whereas the PF-induced activation of the ATM/Chk2/p53 signaling cascade is AMPK dependent and sensitizes mesothelioma cells to cisplatin (Shi et al. 2012). In breast cancer and melanoma cells fasting causes the sumoylation of the specialized DNA polymerase REV1 by SUMO2/3, resulting in the relief of REV1’s inhibition of p53 and enhancing p53’s effects on pro-apoptotic gene expression and apoptosis (Shim et al. 2015).

Many oncogenes are tyrosine kinases and thus provide a target for cancer treatment. PF potentiates the growth inhibiting efficacy of commonly administered tyrosine kinase inhibitors, such as erlotinib, gefitinib, lapatinib, crizotinib, and regorafenib, in in vitro and xenograft models by inhibiting the MAPK signaling and E2F-dependent inhibition of transcription (Caffa et al. 2015). In a non-small cell lung cancer xenograft model, subcutaneous tumor growth was effectively reduced by crizotinib or fasting whereas the combination of fasting with crizotinib was the most efficient treatment option. Similar results have been demonstrated in a colorectal cancer xenograft model for the combination of fasting with regorafenib (Caffa et al. 2015). These results demonstrate the dependence of malignant cells on steady levels of glucose, amino acids, and growth factors and indicate that reducing these factors may protect the organism while reducing tumor progression, particularly in combination with chemotherapy, by generating a challenging environment for the survival of the cancer cell.

Different molecule classes, including antisense RNA, monoclonal antibodies, and dominant negative IGF-1R gene variants have been employed toward this aim (Bahr and Groner 2004), and at least 12 different IGF-1R targeting compounds, including small antagonistic molecules and antibodies, have entered clinical trials (Gualberto and Pollak 2009). These agents have been able to reverse the transformed phenotype in several rodent and human cancer cell lines and, in analogy to fasting, sensitized cancer cells to conventional chemotherapeutic treatment and irradiation (Bahr and Groner 2004). However, IGF-1R blockade did not yield positive results in human trials, possibly because it also interferes with the positive function of IGF-1 in the proliferation of normal cells including stem and immune cells. In contrast, fasting promotes reduced levels of glucose and IGF-1 which return to the normal range after refeeding therefore generating an ideal environment to negatively affect the growth and survival of cancer cells but not that of normal cells.

7. Clinical Efficacy of Fasting

Several studies indicate that fasting has the potential to prevent and treat diseases and promote health in humans. For example, a water-only fast lasting 10–14 days followed by a low-fat, low-sodium vegan-based refeeding period (approximately 6–7 days on average) reduces systolic blood pressure points more than twofold compared to a combined vegan low-fat, low-salt diet and exercise (Goldhamer et al. 2001; Goldhamer 2002). Consuming a very-low-calorie diet of 350 kcal/day (an almost fasting-like approach) has been considered safe in a large cohort with over 2000 participants with chronic diseases, providing evidence that the very-low-calorie diet may protect against several diseases (Michalsen et al. 2005). Additionally, in a randomized clinical trial to evaluate the effects of 2 days of IF (500 kcal/day) a week on weight loss and metabolic disease risk markers in young overweight women found that both chronic CR and IF are equally efficient in reducing biomarkers associated with disease risk (Harvie et al. 2011). Fasting has also been proposed to protect patients from ischemic reperfusion damage following surgery, in which oxidative stress is largely responsible for the damage (Mitchell et al. 2009; van Ginhoven et al. 2009).

Although no randomized clinical trials are (yet) available to evaluate the effect of CR, IF or PF in cancer prevention, preclinical and clinical research supports the potential application in prevention and treatment of human cancers. PF, or fasting-like dietary regimen, can have pronounced effects on IGF-1, insulin, glucose, IGFBP1, and ketone body levels, thereby generating a protective environment for normal cells while creating a metabolic environment that does not favor precancerous and/or cancer cells (Barger et al. 2008; Mercken et al. 2013). In a pilot clinical trial, three monthly FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote health span (Brandhorst et al. 2015). Although not significant, the percentage of mesenchymal stem and progenitor cells in the peripheral blood mono-nucleated cell population showed a trend to increase at the end of FMD, in line with the results obtained in mice (Brandhorst et al. 2015). In these study participants, fasting blood glucose and IGF-1 levels were significantly reduced and remained lower than baseline levels even after resuming their normal diet following the final FMD cycle. Elevated circulating IGF-1 is associated with increased risk of developing certain malignancies (Giovannucci et al. 2003; Smith et al. 2000). In a population-based study of over 6000 American adults, respondents of 50–65 years that reported high protein intake had the highest circulating IGF-1 levels and experienced a 75 % increase in overall mortality and a fourfold increase in cancer death risk during the following 18 years compared to the low-protein/low-IGF-1 cohort (Levine et al. 2014). However, it is important to note that high protein intake was associated with reduced cancer and overall mortality in respondents over 65, indicating that high protein intake may be beneficial in older adults or at least that older adults reporting a low-protein diet are malnourished, frail, and sick (Levine et al. 2014; Dickinson et al. 2014). Although severe IGF-1 deficiency caused by growth hormone receptor deficiency (GHRD) known as Laron’s syndrome leads to growth defects in humans, individuals with GHRD rarely develop cancer (Guevara-Aguirre et al. 2011; Steuerman et al. 2011).

In cancer treatment, preliminary studies of 10 patients with a variety of malignancies that voluntarily fasted for up to 180 h in combination with their prescribed chemotherapy indicate a reduction in common chemotherapy-associated side effects such as vomiting, diarrhea, fatigue and weakness, and in the cases where cancer progression could be followed, there was no evidence that fasting protected tumors or interfered with chemotherapy efficacy (Safdie et al. 2009). Analogous to the effects of fasting on the immune system in mice, the results from a phase I clinical trial indicate that 72 but not 24 h of PF in combination with chemotherapy were associated with normal lymphocyte counts and maintenance of a normal lineage balance in white blood cells (Cheng et al. 2014; Safdie et al. 2009). In a pilot study, 7 out of 13 women diagnosed with HER2-negative stage II/III breast cancer were randomized to fast 24 h before and 24 h after receiving neo-adjuvant (docetaxel/doxorubicin/cyclophosphamide) chemotherapy (de Groot et al. 2015). Fasting was well tolerated and protected from the chemotherapy-dependent reduction in erythrocyte and thrombocyte counts and possibly DNA damage in healthy cells compared to the non-fasted group. The study observed no changes in leukocyte or neutrophil counts which may be associated with a pegfilgrastim-induced production of white blood cells. Notably, the use of the antiemetic drug dexamethasone may explain the observed glucose increase despite 24 h of starvation and likely attenuated some metabolic benefits of starvation, e.g., a more prominent reduction in IGF-1 (de Groot et al. 2015). However, 48 h of fasting may not be sufficient to optimize the differential stress resistance or sensitization of normal and malignant cells. A number of clinical trials are now addressing the effects of fasting or fasting-mimicking diets on humans and the diet-induced protection of patients from the side effects of chemotherapy while sensitizing cancer cells to the treatment (Table 1).

Table 1.

Clinical trials using dietary restriction and fasting in patients with cancer

Identifier Title Purpose Intervention Location
NCT01175837 “Short-term fasting before chemotherapy in treating patients with cancer” Assess the safety and feasibility of short-term fasting prior to administration of chemotherapy Short-term fasting, 24–48 h Mayo Clinic, Rochester, Minnesota, USA
NCT00936364 “Short-term fasting: impact on toxicity” Reducing side effects in patients receiving gemcitabine hydrochloride and cisplatin for advanced solid tumors Short-term fasting, 24–72 h University of Southern California, Los Angeles, California, USA
NCT01304251 “Effects of short-term fasting on tolerance to chemotherapy” Determine the effect of short-term fasting on tolerance to adjuvant chemotherapy in breast cancer patients Short-term fasting, 24 h before and after chemotherapy Leiden University Hospital, Leiden, Netherlands
NCT01954836 “Short-term fasting during chemotherapy in patients with gynecological cancer—a randomized controlled cross-over trial” Assessments of adverse effects, quality of life and laboratory values Short-term fasting, 60–72 h Charite University, Berlin, Germany
NCT01802346 “Controlled low calorie diet in reducing side effects and increasing response to chemotherapy in patients with breast or prostate cancer” Assessments of a low-calorie diet in reducing side effects and increasing response to chemotherapy in patients with breast or prostate cancer Low-calorie diet 3 days prior to and 1 day after chemotherapy University of Southern California, Los Angeles, California, USA
NCT02126449 “Dietary restriction as an adjunct to neoadjuvant chemotherapy for HER2 negative breast cancer” Evaluate the impact of a FMD on tolerance to and efficacy of neoadjuvant chemotherapy in women with stage II or III breast cancer FMD 3 days prior to and 1 day after chemotherapy Leiden University Hospital, Leiden, Netherlands

8. Conclusions

CR has been known to have major health benefits in the vast majority of laboratory animal models. However, chronic CR is not feasible for the great majority of subjects and it often causes detrimental effects, possibly by negatively affecting the immune system, wound healing, and other important functions. PF but particularly higher calorie fasting-mimicking diets have the potential to promote the beneficial effects of CR, while reducing or eliminating adverse effects, and minimizing the burden of chronic restriction or of diets requiring drastic changes several times a week or every other day. The constitutive activation of nutrient signaling pathways which promotes the growth of cancerous cells might also be their Achilles’ heel since they allow the use of fasting to promote the protection of normal cells and organs and sensitization of cancer cells, thus generating a wide-acting, consistent, and inexpensive strategy to increase therapeutic index.

Taken together, these results indicate that PF and FMD have the potential to play an important complementary role in medicine by promoting disease prevention, enhancing disease treatment, delaying the aging process, and stimulating stem cell-based regeneration.

Contributor Information

Sebastian Brandhorst, Department of Biological Sciences, School of Gerontology, Longevity Institute, University of Southern California, Los Angeles, CA, USA.

Valter D. Longo, Department of Biological Sciences, School of Gerontology, Longevity Institute, University of Southern California, Los Angeles, CA, USA IFOM, FIRC Institute of Molecular Oncology, Milan, Italy.

References

  1. Arum O, Bonkowski MS, Rocha JS, Bartke A (2009) The growth hormone receptor gene-disrupted mouse fails to respond to an intermittent fasting diet. Aging cell 8(6):756–760. doi: 10.1111/j.1474-9726.2009.00520.x ACE520 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Bahr C, Groner B (2004) The insulin like growth factor-1 receptor (IGF-1R) as a drug target: novel approaches to cancer therapy. Growth Hormone IGF Res Off J Growth Hormone Res Soc Int IGF Res Soc 14(4):287–295. doi: 10.1016/j.ghir.2004.02.004 [DOI] [PubMed] [Google Scholar]
  3. Barger JL, Kayo T, Vann JM, Arias EB, Wang J, Hacker TA, Wang Y, Raederstorff D, Morrow JD, Leeuwenburgh C, Allison DB, Saupe KW, Cartee GD, Weindruch R, Prolla TA (2008) A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS One 3(6):e2264. doi: 10.1371/journal.pone.0002264 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Bartke A, Chandrashekar V, Bailey B, Zaczek D, Turyn D (2002) Consequences of growth hormone (GH) overexpression and GH resistance. Neuropeptides 36(2–3):201–208. S0143417902908899 [pii] [DOI] [PubMed] [Google Scholar]
  5. Berrigan D, Perkins SN, Haines DC, Hursting SD (2002) Adult-onset calorie restriction and fasting delay spontaneous tumorigenesis in p53-deficient mice. Carcinogenesis 23(5):817–822 [DOI] [PubMed] [Google Scholar]
  6. Bianchi G, Martella R, Ravera S, Marini C, Capitanio S, Orengo A, Emionite L, Lavarello C, Amaro A, Petretto A, Pfeffer U, Sambuceti G, Pistoia V, Raffaghello L, Longo VD (2015) Fasting induces anti-Warburg effect that increases respiration but reduces ATP-synthesis to promote apoptosis in colon cancer models. Oncotarget 6(14):11806–11819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Huisman SA, Bijman-Lagcher W, JN IJ, Smits R, de Bruin RW (2015) Fasting protects against the side effects of irinotecan but preserves its anti-tumor effect in Apc15lox mutant mice. Cell Cycle 14(14):2333–2339. doi: 10.1080/15384101.2015.1044170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bloom WL (1959) Fasting as an introduction to the treatment of obesity. Metabolism 8(3):214–220 [PubMed] [Google Scholar]
  9. Bonkowski MS, Dominici FP, Arum O, Rocha JS, Al Regaiey KA, Westbrook R, Spong A, Panici J, Masternak MM, Kopchick JJ, Bartke A (2009) Disruption of growth hormone receptor prevents calorie restriction from improving insulin action and longevity. PLoS One 4 (2):e4567. doi: 10.1371/journal.pone.0004567 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  10. Bonorden MJ, Rogozina OP, Kluczny CM, Grossmann ME, Grambsch PL, Grande JP, Perkins S, Lokshin A, Cleary MP (2009) Intermittent calorie restriction delays prostate tumor detection and increases survival time in TRAMP mice. Nutr Cancer 61(2):265–275 doi: 10.1080/01635580802419798 908921174 [pii] [DOI] [PubMed] [Google Scholar]
  11. Brandhorst S, Wei M, Hwang S, Morgan TE, Longo VD (2013) Short-term calorie and protein restriction provide partial protection from chemotoxicity but do not delay glioma progression. Exp Gerontol doi: 10.1016/j.exger.2013.02.016 S0531–5565(13)00047–8 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Brandhorst S, Choi IY, Wei M, Cheng CW, Sedrakyan S, Navarrete G, Dubeau L, Yap LP, Park R, Vinciguerra M, Di Biase S, Mirzaei H, Mirisola MG, Childress P, Ji L, Groshen S, Penna F, Odetti P, Perin L, Conti PS, Ikeno Y, Kennedy BK, Cohen P, Morgan TE, Dorff TB, Longo VD (2015) A periodic diet that mimics fasting promotes multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab 22(1):86–99. doi: 10.1016/j.cmet.2015.05.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Brawley OW (2011) Avoidable cancer deaths globally. CA Cancer J Clin 61(2):67–68. caac.20108 [pii] [DOI] [PubMed] [Google Scholar]
  14. Brown-Borg HM (2009) Hormonal control of aging in rodents: the somatotropic axis. Mol Cell Endocrinol 299(1):64–71. doi: 10.1016/j.mce.2008.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Brown-Borg HM, Rakoczy SG (2000) Catalase expression in delayed and premature aging mouse models. Exp Gerontol 35(2):199–212. S0531–5565(00)00079–6 [pii] [DOI] [PubMed] [Google Scholar]
  16. Brown-Borg HM, Borg KE, Meliska CJ, Bartke A (1996) Dwarf mice and the ageing process. Nature 384(6604):33. doi: 10.1038/384033a0 [DOI] [PubMed] [Google Scholar]
  17. Brown-Borg HM, Rakoczy SG, Romanick MA, Kennedy MA (2002) Effects of growth hormone and insulin-like growth factor-1 on hepatocyte antioxidative enzymes. Exp Biol Med (Maywood) 227(2):94–104 [DOI] [PubMed] [Google Scholar]
  18. Caffa I, D’Agostino V, Damonte P, Soncini D, Cea M, Monacelli F, Odetti P, Ballestrero A, Provenzani A, Longo VD, Nencioni A (2015) Fasting potentiates the anticancer activity of tyrosine kinase inhibitors by strengthening MAPK signaling inhibition. Oncotarget 6 (14):11820–11832 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cahill GF Jr, Owen OE (1968) Starvation and survival. Trans Am Clin Climatol Assoc 79:13–20 [PMC free article] [PubMed] [Google Scholar]
  20. Cahill GJ Jr, Owen OE, Morgan AP (1968) The consumption of fuels during prolonged starvation. Adv Enzyme Regul 6:143–150 [DOI] [PubMed] [Google Scholar]
  21. Campisi J, Andersen JK, Kapahi P, Melov S (2011) Cellular senescence: a link between cancer and age-related degenerative disease? Semin Cancer Biol 21(6):354–359. doi: 10.1016/j.semcancer.2011.09.001 S1044–579X(11)00050–2 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cheng C-W, Adams Gregor B, Perin L, Wei M, Zhou X, Lam Ben S, Da Sacco S, Mirisola M, Quinn David I, Dorff Tanya B, Kopchick John J, Longo Valter D (2014) Prolonged fasting reduces IGF-1/PKA to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression. Cell Stem Cell 14(6):810–823. doi: 10.1016/j.stem.2014.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Clancy DJ, Gems D, Harshman LG, Oldham S, Stocker H, Hafen E, Leevers SJ, Partridge L (2001) Extension of life-span by loss of CHICO, a Drosophila insulin receptor substrate protein. Science 292(5514):104–106. doi: 10.1126/science.1057991292/5514/104 [pii] [DOI] [PubMed] [Google Scholar]
  24. Colman RJ, Anderson RM, Johnson SC, Kastman EK, Kosmatka KJ, Beasley TM, Allison DB, Cruzen C, Simmons HA, Kemnitz JW, Weindruch R (2009) Caloric restriction delays disease onset and mortality in rhesus monkeys. Science 325(5937): 201–204. 325/5937/201 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Coschigano KT, Holland AN, Riders ME, List EO, Flyvbjerg A, Kopchick JJ (2003) Deletion, but not antagonism, of the mouse growth hormone receptor results in severely decreased body weights, insulin, and insulin-like growth factor I levels and increased life span. Endocrinology 144(9):3799–3810 [DOI] [PubMed] [Google Scholar]
  26. Cuervo AM, Bergamini E, Brunk UT, Droge W, Ffrench M, Terman A (2005) Autophagy and aging: the importance of maintaining “clean” cells. Autophagy 1(3):131–140. 2017 [pii] [DOI] [PubMed] [Google Scholar]
  27. Danaei G, Vander Hoorn S, Lopez AD, Murray CJ, Ezzati M (2005) Causes of cancer in the world: comparative risk assessment of nine behavioural and environmental risk factors. Lancet 366(9499):1784–1793. S0140–6736(05)67725–2 [pii] [DOI] [PubMed] [Google Scholar]
  28. D’Aronzo M, Vinciguerra M, Mazza T, Panebianco C, Saracino C, Pereira SP, Graziano P, Pazienza V (2015) Fasting cycles potentiate the efficacy of gemcitabine treatment in in vitro and in vivo pancreatic cancer models. Oncotarget 6(21):18545–18557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Davis LM, Pauly JR, Readnower RD, Rho JM, Sullivan PG (2008) Fasting is neuroprotective following traumatic brain injury. J Neurosci Res 86(8):1812–1822. doi: 10.1002/jnr.21628 [DOI] [PubMed] [Google Scholar]
  30. de Groot S, Vreeswijk MP, Welters MJ, Gravesteijn G, Boei JJ, Jochems A, Houtsma D, Putter H, van der Hoeven JJ, Nortier JW, Pijl H, Kroep JR (2015) The effects of short-term fasting on tolerance to (neo) adjuvant chemotherapy in HER2-negative breast cancer patients: a randomized pilot study. BMC Cancer 15:652. doi: 10.1186/s12885-015-1663-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. DePinho RA (2000) The age of cancer. Nature 408(6809):248–254. doi: 10.1038/35041694 [DOI] [PubMed] [Google Scholar]
  32. Descamps O, Riondel J, Ducros V, Roussel AM (2005) Mitochondrial production of reactive oxygen species and incidence of age-associated lymphoma in OF1 mice: effect of alternate-day fasting. Mech Ageing Dev 126(11):1185–1191. doi: 10.1016/j.mad.2005.06.007 S0047–6374 (05)00147–8 [pii] [DOI] [PubMed] [Google Scholar]
  33. Dickinson JM, Gundermann DM, Walker DK, Reidy PT, Borack MS, Drummond MJ, Arora M, Volpi E, Rasmussen BB (2014) Leucine-enriched amino acid ingestion after resistance exercise prolongs myofibrillar protein synthesis and amino acid transporter expression in older men. J Nutr 144(11):1694–1702. doi: 10.3945/jn.114.198671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Drenick EJ, Smith R (1964) Weight reduction by prolonged starvation. Practical management. Postgrad Med 36:A95–100 [DOI] [PubMed] [Google Scholar]
  35. Fabrizio P, Longo VD (2003) The chronological life span of Saccharomyces cerevisiae. Aging Cell 2(2):73–81 [DOI] [PubMed] [Google Scholar]
  36. Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD (2001) Regulation of longevity and stress resistance by Sch9 in yeast. Science 292(5515):288–290. doi: 10.1126/science.1059497 1059497 [pii] [DOI] [PubMed] [Google Scholar]
  37. Fabrizio P, Liou LL, Moy VN, Diaspro A, Valentine JS, Gralla EB, Longo VD (2003) SOD2 functions downstream of Sch9 to extend longevity in yeast. Genetics 163(1):35–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Fontana L, Weiss EP, Villareal DT, Klein S, Holloszy JO (2008) Long-term effects of calorie or protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell 7 (5):681–687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Fontana L, Partridge L, Longo VD (2010) Extending healthy life span–from yeast to humans. Science 328(5976):321–326. doi: 10.1126/science.1172539328/5976/321 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Frystyk J, Delhanty PJ, Skjaerbaek C, Baxter RC (1999) Changes in the circulating IGF system during short-term fasting and refeeding in rats. Am J Physiol 277(2 Pt 1):E245–E252 [DOI] [PubMed] [Google Scholar]
  41. Giannakou ME, Partridge L (2007) Role of insulin-like signalling in Drosophila lifespan. Trends Biochem Sci 32(4):180–188 doi: 10.1016/j.tibs.2007.02.007 S0968–0004(07)00064–3 [pii] [DOI] [PubMed] [Google Scholar]
  42. Giovannucci E, Pollak M, Liu Y, Platz EA, Majeed N, Rimm EB, Willett WC (2003) Nutritional predictors of insulin-like growth factor I and their relationships to cancer in men. Cancer Epidemiol Biomarkers Prev 12(2):84–89 [PubMed] [Google Scholar]
  43. Goldhamer AC (2002) Initial cost of care results in medically supervised water-only fasting for treating high blood pressure and diabetes. J Altern Complement Med 8(6):696–697. doi: 10.1089/10755530260511694 [DOI] [PubMed] [Google Scholar]
  44. Goldhamer A, Lisle D, Parpia B, Anderson SV, Campbell TC (2001) Medically supervised water-only fasting in the treatment of hypertension. J Manipulative Physiol Ther 24(5):335–339. doi: 10.1067/mmt.2001.115263 S0161–4754(01)85575–5 [pii] [DOI] [PubMed] [Google Scholar]
  45. Gonidakis S, Finkel SE, Longo VD (2010) Genome-wide screen identifies Escherichia coli TCA-cycle-related mutants with extended chronological lifespan dependent on acetate metabolism and the hypoxia-inducible transcription factor ArcA. Aging Cell 9(5):868–881. doi: 10.1111/j.1474-9726.2010.00618.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider NL (1982) Effects of intermittent feeding upon growth and life span in rats. Gerontology 28(4):233–241 [DOI] [PubMed] [Google Scholar]
  47. Goodrick CL, Ingram DK, Reynolds MA, Freeman JR, Cider N (1990) Effects of intermittent feeding upon body weight and lifespan in inbred mice: interaction of genotype and age. Mech Ageing Dev 55(1):69–87. 0047–6374(90)90107-Q [pii] [DOI] [PubMed] [Google Scholar]
  48. Grandison RC, Wong R, Bass TM, Partridge L, Piper MD (2009) Effect of a standardised dietary restriction protocol on multiple laboratory strains of Drosophila melanogaster. PLoS One 4(1): e4067. doi: 10.1371/journal.pone.0004067 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Grasl-Kraupp B, Bursch W, Ruttkay-Nedecky B, Wagner A, Lauer B, Schulte-Hermann R (1994) Food restriction eliminates preneoplastic cells through apoptosis and antagonizes carcinogenesis in rat liver. Proc Natl Acad Sci USA 91(21):9995–9999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Greer EL, Dowlatshahi D, Banko MR, Villen J, Hoang K, Blanchard D, Gygi SP, Brunet A (2007) An AMPK-FOXO pathway mediates longevity induced by a novel method of dietary restriction in C. elegans. Current Biol CB 17(19):1646–1656. doi: 10.1016/j.cub.2007.08.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Gualberto A, Pollak M (2009) Emerging role of insulin-like growth factor receptor inhibitors in oncology: early clinical trial results and future directions. Oncogene 28(34):3009–3021. doi: 10.1038/onc.2009.172 [DOI] [PubMed] [Google Scholar]
  52. Guarente L, Kenyon C (2000) Genetic pathways that regulate ageing in model organisms. Nature 408(6809):255–262. doi: 10.1038/35041700 [DOI] [PubMed] [Google Scholar]
  53. Guevara-Aguirre J, Balasubramanian P, Guevara-Aguirre M, Wei M, Madia F, Cheng CW, Hwang D, Martin-Montalvo A, Saavedra J, Ingles S, de Cabo R, Cohen P, Longo VD (2011) Growth hormone receptor deficiency is associated with a major reduction in pro-aging signaling, cancer, and diabetes in humans. Sci Transl Med 3(70):70ra13 doi: 10.1126/scitranslmed.3001845 3/70/70ra13 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100(1):57–70. S0092–8674(00) 81683–9 [pii] [DOI] [PubMed] [Google Scholar]
  55. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144(5):646–674. doi: 10.1016/j.cell.2011.02.013 S0092–8674(11)00127–9 [pii] [DOI] [PubMed] [Google Scholar]
  56. Harrison DE, Archer JR (1989) Natural selection for extended longevity from food restriction. Growth Dev Aging GDA 53(1–2):3. [PubMed] [Google Scholar]
  57. Harvie MN, Pegington M, Mattson MP, Frystyk J, Dillon B, Evans G, Cuzick J, Jebb SA, Martin B, Cutler RG, Son TG, Maudsley S, Carlson OD, Egan JM, Flyvbjerg A, Howell A (2011) The effects of intermittent or continuous energy restriction on weight loss and metabolic disease risk markers: a randomized trial in young overweight women. Int J Obes 35(5):714–727. doi: 10.1038/ijo.2010.171 ijo2010171 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Henis-Korenblit S, Zhang P, Hansen M, McCormick M, Lee SJ, Cary M, Kenyon C (2010)Insulin/IGF-1 signaling mutants reprogram ER stress response regulators to promote longevity. Proc Natl Acad Sci USA 107(21):9730–9735. doi: 10.1073/pnas.1002575107 1002575107 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hine C, Harputlugil E, Zhang Y, Ruckenstuhl C, Lee BC, Brace L, Longchamp A, Trevino-Villarreal JH, Mejia P, Ozaki CK, Wang R, Gladyshev VN, Madeo F, Mair WB, Mitchell JR (2015) Endogenous hydrogen sulfide production is essential for dietary restriction benefits. Cell 160(1–2):132–144. doi: 10.1016/j.cell.2014.11.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Holzenberger M, Dupont J, Ducos B, Leneuve P, Geloen A, Even PC, Cervera P, Le Bouc Y (2003) IGF-1 receptor regulates lifespan and resistance to oxidative stress in mice. Nature 421 (6919):182–187. doi: 10.1038/nature01298 nature01298 [pii] [DOI] [PubMed] [Google Scholar]
  61. Honjoh S, Yamamoto T, Uno M, Nishida E (2009) Signalling through RHEB-1 mediates intermittent fasting-induced longevity in C. elegans. Nature 457(7230):726–730. doi: 10.1038/nature07583 [DOI] [PubMed] [Google Scholar]
  62. Hsu AL, Murphy CT, Kenyon C (2003) Regulation of aging and age-related disease by DAF-16 and heat-shock factor. Science 300(5622):1142–1145. doi: 10.1126/science.1083701 300/5622/1142 [pii] [DOI] [PubMed] [Google Scholar]
  63. Hursting SD, Perkins SN, Phang JM (1994) Calorie restriction delays spontaneous tumorigenesis in p53-knockout transgenic mice. Proc Natl Acad Sci USA 91(15):7036–7040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Imae M, Fu Z, Yoshida A, Noguchi T, Kato H (2003) Nutritional and hormonal factors control the gene expression of FoxOs, the mammalian homologues of DAF-16. J Mol Endocrinol 30 (2):253–262 [DOI] [PubMed] [Google Scholar]
  65. Jemal A, Siegel R, Ward E, Hao Y, Xu J, Murray T, Thun MJ (2008) Cancer statistics, 2008. CA Cancer J Clin 58 (2):71–96. CA.2007.0010 [pii] [DOI] [PubMed] [Google Scholar]
  66. Jenkins DE, Schultz JE, Matin A (1988) Starvation-induced cross protection against heat or H2O2 challenge in Escherichia coli. J Bacteriol 170(9):3910–3914 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Johnson TE (1990) Increased life-span of age-1 mutants in Caenorhabditis elegans and lower Gompertz rate of aging. Science 249(4971):908–912 [DOI] [PubMed] [Google Scholar]
  68. Kalaany NY, Sabatini DM (2009) Tumours with PI3K activation are resistant to dietary restriction. Nature 458(7239):725–731. doi: 10.1038/nature07782 nature07782 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Kenyon C (2001) A conserved regulatory system for aging. Cell 105(2):165–168. S0092–8674(01) 00306–3 [pii] [DOI] [PubMed] [Google Scholar]
  70. Kerndt PR, Naughton JL, Driscoll CE, Loxterkamp DA (1982) Fasting: the history, pathophysiology and complications. West J Med 137(5):379–399 [PMC free article] [PubMed] [Google Scholar]
  71. Ketelslegers JM, Maiter D, Maes M, Underwood LE, Thissen JP (1995) Nutritional regulation of insulin-like growth factor-I. Metabolism 44(10 Suppl 4):50–57 [DOI] [PubMed] [Google Scholar]
  72. Kim SK, Demetri GD (1996) Chemotherapy and neutropenia. Hematol Oncol Clin North Am 10 (2):377–395 [DOI] [PubMed] [Google Scholar]
  73. Kinzler KW, Vogelstein B (1996) Lessons from hereditary colorectal cancer. Cell 87(2):159–170. S0092–8674(00)81333–1 [pii] [DOI] [PubMed] [Google Scholar]
  74. Koubova J, Guarente L (2003) How does calorie restriction work? Genes Dev 17(3):313–321. doi: 10.1101/gad.1052903 [DOI] [PubMed] [Google Scholar]
  75. Kristan DM (2008) Calorie restriction and susceptibility to intact pathogens. Age (Dordr) 30(2–3):147–156. doi: 10.1007/s11357-008-9056-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kroemer G, Marino G, Levine B (2010) Autophagy and the integrated stress response. Mol Cell 40 (2):280–293. doi: 10.1016/j.molcel.2010.09.023 S1097–2765(10)00751–3 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Larson-Meyer DE, Newcomer BR, Heilbronn LK, Volaufova J, Smith SR, Alfonso AJ, Lefevre M, Rood JC, Williamson DA, Ravussin E (2008) Effect of 6-month calorie restriction and exercise on serum and liver lipids and markers of liver function. Obesity (Silver Spring) 16 (6):1355–1362. doi: 10.1038/oby.2008.201 oby2008201 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Lee C, Longo VD (2011) Fasting vs dietary restriction in cellular protection and cancer treatment: from model organisms to patients. Oncogene 30(30):3305–3316. doi: 10.1038/onc.2011.91 onc201191 [pii] [DOI] [PubMed] [Google Scholar]
  79. Lee SJ, Murphy CT, Kenyon C (2009) Glucose shortens the life span of C. elegans by downregulating DAF-16/FOXO activity and aquaporin gene expression. Cell Metab 10 (5):379–391. doi: 10.1016/j.cmet.2009.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lee C, Safdie FM, Raffaghello L, Wei M, Madia F, Parrella E, Hwang D, Cohen P, Bianchi G, Longo VD (2010) Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res 70(4):1564–1572. doi: 10.1158/0008-5472.CAN-09-3228 0008–5472.CAN-09–3228 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G, Martin-Montalvo A, Pistoia V, Wei M, Hwang S, Merlino A, Emionite L, de Cabo R, Longo VD (2012) Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. doi: 10.1126/scitranslmed.3003293 scitranslmed.3003293 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Levine ME, Suarez JA, Brandhorst S, Balasubramanian P, Cheng CW, Madia F, Fontana L, Mirisola MG, Guevara-Aguirre J, Wan J, Passarino G, Kennedy BK, Wei M, Cohen P, Crimmins EM, Longo VD (2014) Low protein intake is associated with a major reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older population. Cell Metab 19(3):407–417. doi: 10.1016/j.cmet.2014.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Li Y, Xu W, McBurney MW, Longo VD (2008) SirT1 inhibition reduces IGF-I/IRS-2/Ras/ERK1/2 signaling and protects neurons. Cell Metab 8(1):38–48. doi: 10.1016/j.cmet.2008.05.004 S1550–4131(08)00148–4 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Liao CY, Rikke BA, Johnson TE, Diaz V, Nelson JF (2010) Genetic variation in the murine lifespan response to dietary restriction: from life extension to life shortening. Aging Cell 9 (1):92–95. doi: 10.1111/j.1474-9726.2009.00533.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Longo VD (1999) Mutations in signal transduction proteins increase stress resistance and longevity in yeast, nematodes, fruit flies, and mammalian neuronal cells. Neurobiol Aging 20 (5):479–486. S0197–4580(99)00089–5 [pii] [DOI] [PubMed] [Google Scholar]
  86. Longo VD, Finch CE (2003) Evolutionary medicine: from dwarf model systems to healthy centenarians? Science 299(5611):1342–1346. doi: 10.1126/science.1077991 299/5611/1342 [pii] [DOI] [PubMed] [Google Scholar]
  87. Longo VD, Fontana L (2010) Calorie restriction and cancer prevention: metabolic and molecular mechanisms. Trends Pharmacol Sci 31(2):89–98. doi: 10.1016/j.tips.2009.11.004 S0165–6147 (09)00202–8 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Longo VD, Mattson MP (2014) Fasting: molecular mechanisms and clinical applications. Cell Metab 19(2):181–192. doi: 10.1016/j.cmet.2013.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Longo VD, Ellerby LM, Bredesen DE, Valentine JS, Gralla EB (1997) Human Bcl-2 reverses survival defects in yeast lacking superoxide dismutase and delays death of wild-type yeast. J Cell Biol 137(7):1581–1588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Love RR, Leventhal H, Easterling DV, Nerenz DR (1989) Side effects and emotional distress during cancer chemotherapy. Cancer 63(3):604–612 [DOI] [PubMed] [Google Scholar]
  91. Maccario M, Aimaretti G, Grottoli S, Gauna C, Tassone F, Corneli G, Rossetto R, Wu Z, Strasburger CJ, Ghigo E (2001) Effects of 36 hour fasting on GH/IGF-I axis and metabolic parameters in patients with simple obesity. comparison with normal subjects and hypopituitary patients with severe GH deficiency. Int J Obes Relat Metab Disord 25(8):1233–1239. doi: 10.1038/sj.ijo.0801671 [DOI] [PubMed] [Google Scholar]
  92. Madia F, Wei M, Yuan V, Hu J, Gattazzo C, Pham P, Goodman MF, Longo VD (2009) Oncogene homologue Sch9 promotes age-dependent mutations by a superoxide and Rev1/Polzeta-dependent mechanism. J Cell Biol 186(4):509–523. doi: 10.1083/jcb.200906011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Mahoney LB, Denny CA, Seyfried TN (2006) Caloric restriction in C57BL/6J mice mimics therapeutic fasting in humans. Lipids Health Dis 5:13. doi: 10.1186/1476-511X-5-13 1476–511X-5–13 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Marino G, Madeo F, Kroemer G (2010) Autophagy for tissue homeostasis and neuroprotection. Curr Opin Cell Biol 23(2):198–206 doi: 10.1016/j.ceb.2010.10.001 S0955–0674(10)00172–9 [pii] [DOI] [PubMed] [Google Scholar]
  95. Matsuzaki J, Kuwamura M, Yamaji R, Inui H, Nakano Y (2001) Inflammatory responses to lipopolysaccharide are suppressed in 40 % energy-restricted mice. J Nutr 131(8):2139–2144 [DOI] [PubMed] [Google Scholar]
  96. Mattison JA, Roth GS, Beasley TM, Tilmont EM, Handy AM, Herbert RL, Longo DL, Allison DB, Young JE, Bryant M, Barnard D, Ward WF, Qi W, Ingram DK, de Cabo R (2012) Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study. Nature 489(7415):318–321. doi: 10.1038/nature11432 nature11432 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Mattson MP (2005) Energy intake, meal frequency, and health: a neurobiological perspective. Annu Rev Nutr 25:237–260. doi: 10.1146/annurev.nutr.25.050304.092526 [DOI] [PubMed] [Google Scholar]
  98. Mattson MP (2012) Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab 16(6):706–722. doi: 10.1016/j.cmet.2012.08.012 S1550–4131 (12)00402–0 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Mattson MP, Wan R (2005) Beneficial effects of intermittent fasting and caloric restriction on the cardiovascular and cerebrovascular systems. J Nutr Biochem 16(3):129–137. doi: 10.1016/j.jnutbio.2004.12.007 S0955–2863(04)00261-X [pii] [DOI] [PubMed] [Google Scholar]
  100. Medema RH, Bos JL (1993) The role of p21ras in receptor tyrosine kinase signaling. Crit Rev Oncog 4(6):615–661 [PubMed] [Google Scholar]
  101. Mercken EM, Crosby SD, Lamming DW, JeBailey L, Krzysik-Walker S, Villareal DT, Capri M, Franceschi C, Zhang Y, Becker K, Sabatini DM, de Cabo R, Fontana L (2013) Calorie restriction in humans inhibits the PI3 K/AKT pathway and induces a younger transcription profile. Aging Cell 12(4):645–651. doi: 10.1111/acel.12088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Merimee TJ, Fineberg SE (1974) Growth hormone secretion in starvation: a reassessment. J Clin Endocrinol Metab 39(2):385–386. doi: 10.1210/jcem-39-2-385 [DOI] [PubMed] [Google Scholar]
  103. Michalsen A, Hoffmann B, Moebus S, Backer M, Langhorst J, Dobos GJ (2005) Incorporation of fasting therapy in an integrative medicine ward: evaluation of outcome, safety, and effects on lifestyle adherence in a large prospective cohort study. J Altern Complement Med 11(4):601–607. doi: 10.1089/acm.2005.11.601 [DOI] [PubMed] [Google Scholar]
  104. Mirzaei H, Suarez JA, Longo VD (2014) Protein and amino acid restriction, aging and disease: from yeast to humans. Trends Endocrinol Metab TEM 25(11):558–566. doi: 10.1016/j.tem.2014.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Mitchell JR, Verweij M, Brand K, van de Ven M, Goemaere N, van den Engel S, Chu T, Forrer F, Muller C, de Jong M, van IW, Jn IJ, Hoeijmakers JH, de Bruin RW (2009) Short-term dietary restriction and fasting precondition against ischemia reperfusion injury in mice. Aging Cell 9 (1):40–53. doi: 10.1111/j.1474-9726.2009.00532.x ACE532[pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Moller N, Jorgensen JO (2009) Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev 30(2):152–177. doi: 10.1210/er.2008-0027 er.2008–0027 [pii] [DOI] [PubMed] [Google Scholar]
  107. Moreschi (1909) Beziehungen zwischen Ernährung und Tumorwachstum. Z Immunitätsforsch Orig (2): 651–675 [Google Scholar]
  108. Morselli E, Galluzzi L, Kepp O, Criollo A, Maiuri MC, Tavernarakis N, Madeo F, Kroemer G (2009a) Autophagy mediates pharmacological lifespan extension by spermidine and resveratrol. Aging (Albany NY) 1(12):961–970 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Morselli E, Galluzzi L, Kepp O, Vicencio JM, Criollo A, Maiuri MC, Kroemer G (2009) Anti- and pro-tumor functions of autophagy. Biochimica et Biophysica Acta 1793(9):1524–1532. doi: 10.1016/j.bbamcr.2009.01.006 S0167–4889(09)00024-X [pii] [DOI] [PubMed] [Google Scholar]
  110. Mukherjee P, Abate LE, Seyfried TN (2004) Antiangiogenic and proapoptotic effects of dietary restriction on experimental mouse and human brain tumors. Clin Cancer Res 10(16):5622–5629. doi: 10.1158/1078-0432.CCR-04-0308 10/16/5622 [pii] [DOI] [PubMed] [Google Scholar]
  111. Murakami S (2006) Stress resistance in long-lived mouse models. Exp Gerontol 41(10):1014–1019. doi: 10.1016/j.exger.2006.06.061 S0531–5565(06)00236–1 [pii] [DOI] [PubMed] [Google Scholar]
  112. Nicklin P, Bergman P, Zhang B, Triantafellow E, Wang H, Nyfeler B, Yang H, Hild M, Kung C, Wilson C, Myer VE, MacKeigan JP, Porter JA, Wang YK, Cantley LC, Finan PM, Murphy LO (2009) Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136 (3):521–534. doi: 10.1016/j.cell.2008.11.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Palmblad J, Levi L, Burger A, Melander A, Westgren U, von Schenck H, Skude G (1977) Effects of total energy withdrawal (fasting) on thelevels of growth hormone, thyrotropin, cortisol, adrenaline, noradrenaline, T4, T3, and rT3 in healthy males. Acta Medica Scandinavica 201(1–2):15–22 [DOI] [PubMed] [Google Scholar]
  114. Pamplona R, Barja G (2006) Mitochondrial oxidative stress, aging and caloric restriction: the protein and methionine connection. Biochimica et biophysica acta 1757(5–6):496–508. doi: 10.1016/j.bbabio.2006.01.009 S0005–2728(06)00022–3 [pii] [DOI] [PubMed] [Google Scholar]
  115. Paoletti R, Bolego C, Poli A, Cignarella A (2006) Metabolic syndrome, inflammation and atherosclerosis. Vasc Health Risk Manag 2(2):145–152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Paradis S, Ailion M, Toker A, Thomas JH, Ruvkun G (1999) A PDK1 homolog is necessary and sufficient to transduce AGE-1 PI3 kinase signals that regulate diapause in Caenorhabditis elegans. Genes Dev 13(11):1438–1452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Partridge AH, Burstein HJ, Winer EP (2001) Side effects of chemotherapy and combined chemohormonal therapy in women with early-stage breast cancer. J Natl Cancer Inst Monogr 30:135–142 [DOI] [PubMed] [Google Scholar]
  118. Piper MD, Partridge L (2007) Dietary restriction in Drosophila: delayed aging or experimental artefact? PLoS Genet 3(4):e57. doi: 10.1371/journal.pgen.0030057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Premoselli F, Sesca E, Binasco V, Caderni G, Tessitore L (1998) Fasting/re-feeding before initiation enhances the growth of aberrant crypt foci induced by azoxymethane in rat colon and rectum. Int J Cancer 77(2):286–294. doi: [DOI] [PubMed] [Google Scholar]
  120. Raffaghello L, Lee C, Safdie FM, Wei M, Madia F, Bianchi G, Longo VD (2008) Starvation-dependent differential stress resistance protects normal but not cancer cells against high-dose chemotherapy. Proc Natl Acad Sci USA 105(24):8215–8220. doi: 10.1073/pnas.0708100105 0708100105 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Reed MJ, Penn PE, Li Y, Birnbaum R, Vernon RB, Johnson TS, Pendergrass WR, Sage EH, Abrass IB, Wolf NS (1996) Enhanced cell proliferation and biosynthesis mediate improved wound repair in refed, caloric-restricted mice. Mech Ageing Dev 89(1):21–43. 004763749601737X [pii] [DOI] [PubMed] [Google Scholar]
  122. Rikke BA, Liao CY, McQueen MB, Nelson JF, Johnson TE (2010) Genetic dissection of dietary restriction in mice supports the metabolic efficiency model of life extension. Exp Gerontol 45 (9):691–701. doi: 10.1016/j.exger.2010.04.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Ruckenstuhl C, Buttner S, Carmona-Gutierrez D, Eisenberg T, Kroemer G, Sigrist SJ, Frohlich KU, Madeo F (2009) The Warburg effect suppresses oxidative stress induced apoptosis in a yeast model for cancer. PLoS One 4(2):e4592. doi: 10.1371/journal.pone.0004592 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Runcie J, Thomson TJ (1970) Prolonged starvation—a dangerous procedure? Br Med J 3 (5720):432–435 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Safdie FM, Dorff T, Quinn D, Fontana L, Wei M, Lee C, Cohen P, Longo VD (2009) Fasting and cancer treatment in humans: a case series report. Aging (Albany NY) 1(12):988–1007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Safdie F, Brandhorst S, Wei M, Wang W, Lee C, Hwang S, Conti PS, Chen TC, Longo VD (2012) Fasting enhances the response of glioma to chemo- and radiotherapy. PloS One 7(9):e44603. doi: 10.1371/journal.pone.0044603 PONE-D-12–00123 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Salmon AB, Murakami S, Bartke A, Kopchick J, Yasumura K, Miller RA (2005) Fibroblast cell lines from young adult mice of long-lived mutant strains are resistant to multiple forms of stress. Am J Physiol Endocrinol Metab 289(1):E23–E29. doi: 10.1152/ajpendo.00575.2004 00575.2004 [pii] [DOI] [PubMed] [Google Scholar]
  128. Sans MD, Lee SH, D’Alecy LG, Williams JA (2004) Feeding activates protein synthesis in mouse pancreas at the translational level without increase in mRNA. Am J Physiol Gastrointest Liver Physiol 287(3):G667–G675. doi: 10.1152/ajpgi.00505.2003 [DOI] [PubMed] [Google Scholar]
  129. Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury AI, Claret M, Al-Qassab H, Carmignac D, Ramadani F, Woods A, Robinson IC, Schuster E, Batterham RL, Kozma SC, Thomas G, Carling D, Okkenhaug K, Thornton JM, Partridge L, Gems D, Withers DJ (2009) Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science 326 (5949):140–144 doi: 10.1126/science.1177221326/5949/140 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Sesca E, Premoselli F, Binasco V, Bollito E, Tessitore L (1998) Fasting-refeeding stimulates the development of mammary tumors induced by 7,12-dimethylbenz[a]anthracene. Nutr Cancer 30 (1):25–30. doi: 10.1080/01635589809514636 [DOI] [PubMed] [Google Scholar]
  131. Sharma HS, Nyberg F, Gordh T, Alm P, Westman J (2000) Neurotrophic factors influence upregulation of constitutive isoform of heme oxygenase and cellular stress response in the spinal cord following trauma. An experimental study using immunohistochemistry in the rat. Amino Acids 19(1):351–361 [PubMed] [Google Scholar]
  132. Shi Y, Felley-Bosco E, Marti TM, Orlowski K, Pruschy M, Stahel RA (2012) Starvation-induced activation of ATM/Chk2/p53 signaling sensitizes cancer cells to cisplatin. BMC Cancer 12:571 doi: 10.1186/1471-2407-12-5711471-2407-12-571 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Shim HS, Wei M, Brandhorst S, Longo VD (2015) Starvation promotes REV1 SUMOylation and p53-dependent sensitization of melanoma and breast cancer cells. Cancer Res 75(6):1056–1067. doi: 10.1158/0008-5472.CAN-14-2249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Siegel R, Naishadham D, Jemal A (2012) Cancer statistics, 2012. CA Cancer J Clin 62(1):10–29. doi: 10.3322/caac.20138 [DOI] [PubMed] [Google Scholar]
  135. Smith GD, Gunnell D, Holly J (2000) Cancer and insulin-like growth factor-I. A potential mechanism linking the environment with cancer risk. Bmj 321(7265):847–848 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Smith ED, Kaeberlein TL, Lydum BT, Sager J, Welton KL, Kennedy BK, Kaeberlein M (2008) Age- and calorie-independent life span extension from dietary restriction by bacterial deprivation in Caenorhabditis elegans. BMC Dev Biol 8:49. doi: 10.1186/1471-213X-8-49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Sohal RS, Forster MJ (2014) Caloric restriction and the aging process: a critique. Free Radic Biol Med 73:366–382. doi: 10.1016/j.freeradbiomed.2014.05.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Sohal RS, Weindruch R (1996) Oxidative stress, caloric restriction, and aging. Science 273 (5271):59–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Sonntag WE, Lynch CD, Cefalu WT, Ingram RL, Bennett SA, Thornton PL, Khan AS (1999) Pleiotropic effects of growth hormone and insulin-like growth factor (IGF)-1 on biological aging: inferences from moderate caloric-restricted animals. J Gerontol Ser A Biol Sci Med Sci 54(12):B521–B538 [DOI] [PubMed] [Google Scholar]
  140. Steuerman R, Shevah O, Laron Z (2011) Congenital IGF1 deficiency tends to confer protection against post-natal development of malignancies. Eur J Endocrinol/ Eur Fed Endocr Soc 164 (4):485–489. doi: 10.1530/EJE-10-0859 [DOI] [PubMed] [Google Scholar]
  141. Tagliaferro AR, Ronan AM, Meeker LD, Thompson HJ, Scott AL, Sinha D (1996) Cyclic food restriction alters substrate utilization and abolishes protection from mammary carcinogenesis female rats. J Nutr 126(5):1398–1405 [DOI] [PubMed] [Google Scholar]
  142. Tannenbaum A (1940) The initiation and growth of tumours. Introduction. 1. Effects of underfeeding. Am J Cancer 38:335–350 [Google Scholar]
  143. Tannenbaum A (1996) The dependence of tumor formation on the composition of the calorie-restricted diet as well as on the degree of restriction. 1945. Nutrition 12(9):653–654. S0899900796001797 [pii] [PubMed] [Google Scholar]
  144. Tannenbaum GS, Rorstad O, Brazeau P (1979) Effects of prolonged food deprivation on the ultradian growth hormone rhythm and immunoreactive somatostatin tissue levels in the rat. Endocrinology 104(6):1733–1738 [DOI] [PubMed] [Google Scholar]
  145. Tessitore L, Tomasi C, Greco M (1999) Fasting-induced apoptosis in rat liver is blocked by cycloheximide. Eur J Cell Biol 78(8):573–579 doi: 10.1016/S0171-9335(99)80023-5 S0171–9335(99)80023–5 [pii] [DOI] [PubMed] [Google Scholar]
  146. Tettweiler G, Miron M, Jenkins M, Sonenberg N, Lasko PF (2005) Starvation and oxidative stress resistance in Drosophila are mediated through the eIF4E-binding protein, d4E-BP. Genes Dev 19(16):1840–1843. doi: 10.1101/gad.1311805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A (2015) Global cancer statistics, 2012. CA Cancer J Clin 65(2):87–108. doi: 10.3322/caac.21262 [DOI] [PubMed] [Google Scholar]
  148. Underwood LE, Thissen JP, Lemozy S, Ketelslegers JM, Clemmons DR (1994) Hormonal and nutritional regulation of IGF-I and its binding proteins. Horm Res 42(4–5):145–151 [DOI] [PubMed] [Google Scholar]
  149. Unger RH, Eisentraut AM, Madison LL (1963) The effects of total starvation upon the levels of circulating glucagon and insulin in man. J Clin Investig 42:1031–1039. doi: 10.1172/JCI104788 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. van Ginhoven TM, Mitchell JR, Verweij M, Hoeijmakers JH, Ijzermans JN, de Bruin RW (2009) The use of preoperative nutritional interventions to protect against hepatic ischemia-reperfusion injury. Liver Transpl 15(10):1183–1191. doi: 10.1002/lt.21871 [DOI] [PubMed] [Google Scholar]
  151. Vander Heiden MG, Cantley LC, Thompson CB (2009) Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324(5930):1029–1033. doi: 10.1126/science.1160809324/5930/1029 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Vaughn AE, Deshmukh M (2008) Glucose metabolism inhibits apoptosis in neurons and cancer cells by redox inactivation of cytochrome c. Nat Cell Biol 10(12):1477–1483. doi: 10.1038/ncb1807 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Verweij M, van Ginhoven TM, Mitchell JR, Sluiter W, van den Engel S, Roest HP, Torabi E, Ijzermans JN, Hoeijmakers JH, de Bruin RW (2011) Preoperative fasting protects mice against hepatic ischemia/reperfusion injury: mechanisms and effects on liver regeneration. Liver Transpl 17(6):695–704. doi: 10.1002/lt.22243 [DOI] [PubMed] [Google Scholar]
  154. Vigne P, Frelin C (2007) Diet dependent longevity and hypoxic tolerance of adult Drosophila melanogaster. Mech Ageing Dev 128(5–6):401–406. doi: 10.1016/j.mad.2007.05.008 [DOI] [PubMed] [Google Scholar]
  155. Villa-Cuesta E, Sage BT, Tatar M (2010) A role for Drosophila dFoxO and dFoxO 5’UTR internal ribosomal entry sites during fasting. PLoS One 5(7):e11521. doi: 10.1371/journal.pone.0011521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Wang T, Hung CC, Randall DJ (2006) The comparative physiology of food deprivation: from feast to famine. Annu Rev Physiol 68:223–251. doi: 10.1146/annurev.physiol.68.040104.105739 [DOI] [PubMed] [Google Scholar]
  157. Warburg O (1956) On respiratory impairment in cancer cells. Science 124(3215):269–270 [PubMed] [Google Scholar]
  158. Wei M, Fabrizio P, Hu J, Ge H, Cheng C, Li L, Longo VD (2008) Life span extension by calorie restriction depends on Rim15 and transcription factors downstream of Ras/PKA, Tor, and Sch9. PLoS Genet 4(1):e13 doi: 10.1371/journal.pgen.004001307-PLGE-RA-0807 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Weindruch R, Walford RL (1982) Dietary restriction in mice beginning at 1 year of age: effect on life-span and spontaneous cancer incidence. Science 215(4538):1415–1418 [DOI] [PubMed] [Google Scholar]
  160. Weindruch R, Walford RL, Fligiel S, Guthrie D (1986) The retardation of aging in mice by dietary restriction: longevity, cancer, immunity and lifetime energy intake. J Nutr 116(4):641–654 [DOI] [PubMed] [Google Scholar]
  161. Weinkove D, Halstead JR, Gems D, Divecha N (2006) Long-term starvation and ageing induce AGE-1/PI 3-kinase-dependent translocation of DAF-16/FOXO to the cytoplasm. BMC Biol 4:1. doi: 10.1186/1741-7007-4-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Weraarchakul N, Strong R, Wood WG, Richardson A (1989) The effect of aging and dietary restriction on DNA repair. Exp Cell Res 181(1):197–204. 0014–4827(89)90193–6 [pii] [DOI] [PubMed] [Google Scholar]
  163. Withers SS, Kass PH, Rodriguez CO Jr, Skorupski KA, O’Brien D, Guerrero TA, Sein KD, Rebhun RB (2014) Fasting reduces the incidence of delayed-type vomiting associated with doxorubicin treatment in dogs with lymphoma. Transl Oncol. doi: 10.1016/j.tranon.2014.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Wohlgemuth SE, Julian D, Akin DE, Fried J, Toscano K, Leeuwenburgh C, Dunn WA Jr (2007) Autophagy in the heart and liver during normal aging and calorie restriction. Rejuvenation Res 10(3):281–292. doi: 10.1089/rej.2006.0535 [DOI] [PubMed] [Google Scholar]
  165. Youngman LD (1993) Protein restriction (PR) and caloric restriction (CR) compared: effects on DNA damage, carcinogenesis, and oxidative damage. Mutat Res 295(4–6):165–179 [DOI] [PubMed] [Google Scholar]
  166. Zapf J, Hauri C, Futo E, Hussain M, Rutishauser J, Maack CA, Froesch ER (1995) Intravenously injected insulin-like growth factor (IGF) I/IGF binding protein-3 complex exerts insulin-like effects in hypophysectomized, but not in normal rats. J Clin Investig 95(1):179–186. doi: 10.1172/JCI117636 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES