Next Article in Journal
Influences of Vitamin B12 Supplementation on Cognition and Homocysteine in Patients with Vitamin B12 Deficiency and Cognitive Impairment
Next Article in Special Issue
Sarcopenia as an Independent Risk Factor for Specific Cancers: A Propensity Score-Matched Asian Population-Based Cohort Study
Previous Article in Journal
Understanding the Relationship between Food Security and Mental Health for Food-Insecure Mothers in Virginia
Previous Article in Special Issue
Plasma Metabolite Profiles of Red Meat, Poultry, and Fish Consumption, and Their Associations with Colorectal Cancer Risk
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Molecular Approach to Understanding the Role of Diet in Cancer-Related Fatigue: Challenges and Future Opportunities

1
Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL 33617, USA
2
Cancer Control and Population Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
3
Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT 84112, USA
4
Department of Population Health Sciences, University of Utah, Salt Lake City, UT 84112, USA
5
Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
6
Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, 69047 Heidelberg, Germany
7
Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76798, USA
8
Department of Physical Medicine and Rehabilitation, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
9
Center for Integrated Research in Cancer and Lifestyle, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
10
Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
Nutrients 2022, 14(7), 1496; https://doi.org/10.3390/nu14071496
Submission received: 24 February 2022 / Revised: 27 March 2022 / Accepted: 31 March 2022 / Published: 2 April 2022
(This article belongs to the Special Issue Nutritional Metabolomics in Cancer Epidemiology)

Abstract

:
Cancer-related fatigue (CRF) is considered one of the most frequent and distressing symptoms for cancer survivors. Despite its high prevalence, factors that predispose, precipitate, and perpetuate CRF are poorly understood. Emerging research focuses on cancer and treatment-related nutritional complications, changes in body composition, and nutritional deficiencies that can compound CRF. Nutritional metabolomics, the novel study of diet-related metabolites in cells, tissues, and biofluids, offers a promising tool to further address these research gaps. In this position paper, we examine CRF risk factors, summarize metabolomics studies of CRF, outline dietary recommendations for the prevention and management of CRF in cancer survivorship, and identify knowledge gaps and challenges in applying nutritional metabolomics to understand dietary contributions to CRF over the cancer survivorship trajectory.

1. Introduction

The American Cancer Society defines a cancer survivor as “anyone who has ever been diagnosed with cancer no matter where they are in the course of their disease” [1]. For many survivors, cancer-related fatigue (CRF) is a “distressing, persistent, subjective sense of physical, emotional, and/or cognitive tiredness or exhaustion related to cancer or cancer treatment that is not proportional to recent activity and interferes with usual functioning” [2], and prevention and interventional strategies are needed to address this growing challenge. CRF is considered one of the most frequent side-effects of cancer treatment [3], prompting the National Cancer Institute (NCI) to identify CRF as one of the top five high-priority cancer research areas [4]. CRF negatively impacts work, social relationships, and daily activities, resulting in a significant impairment in quality of life [5,6,7]. Despite these negative implications, CRF is underreported by cancer survivors and physicians [2,8]. A recent systematic review suggests that over half of all cancer survivors experience CRF, [3] and 96% of patients undergoing chemotherapy experience CRF [9]. Clinical symptoms of CRF include cognitive difficulties, hot flashes, functional decline, and depression, all of which result in a significant reduction in quality of life [10,11]. Although there is no evidence-based, gold-standard treatment for CRF, current interventions include education and counseling, skills training in fatigue management strategies, and specific nonpharmacologic and pharmacologic interventions [2]. Although exercise has been shown to reduce CRF [12,13], many patients are unable to exercise consistently [14,15]. Psychosocial interventions demonstrate benefit [16,17,18], although they tend to be time-intensive, limiting uptake, compliance, maintenance, and disseminability. Pharmacologic interventions have not resulted in substantial decreases in CRF. Paroxetine, sertraline, modafinil, and armodafinil have produced no benefit for CRF in randomized trials [19,20,21,22,23,24]. Evidence is mixed regarding the effects of methylphenidate [25,26,27,28] and uptake has been limited, perhaps because methylphenidate can be habit-forming and is associated with side effects including agitation, insomnia, and loss of appetite [29]. CRF is a complex syndrome, and treatment strategies of CRF may differ depending on the survivor’s clinical status (e.g., under active treatment, post-treatment, end-of-life care). While corticosteroids may provide short-term relief for fatigue and improve quality of life in terminal patients, they may not be suitable for long term use in post-treatment survivors [30]. Additional treatment options for CRF are clearly needed. Nutritional interventions offer a promising strategy for reducing CRF [31], but research is needed to elucidate the potential contributions of diet to CRF development and severity to isolate dietary targets that could help mitigate its devastating impacts.
Nutritional metabolomics, the study of diet-related small molecules, is an emerging field of study. It has recently been leveraged to identify objective biomarkers of dietary exposures, and to identify biochemical pathways underlying complex relationships between diet and health or disease. Nutritional metabolomics also encompasses the metabolic state of an individual based on nutritional status (i.e., the state of a person’s health in terms of the nutrients in their diet) [32,33,34]. This is particularly relevant to cancer survivors, as evidence supports the role of nutritional status in the etiology of CRF [31]. Moreover, levels of circulating pro-inflammatory biomarkers are increased in cancer survivors with fatigue [5]. Thus, specific diets aimed at reducing chronic inflammation (e.g., a Mediterranean diet or other specific dietary patterns shown to reduce circulating inflammatory biomarkers [35]) may mitigate CRF via modulating inflammation, and additionally address nutritional deficiencies. Identifying other biochemical pathways linked with CRF could offer additional targets for dietary change. In all, evidence-based nutritional guidelines for alleviating CRF are critically needed, despite the high prevalence of nutritional complications related to fatigue in cancer survivors [36]. In this position paper, we examine CRF risk factors, summarize metabolomics studies of CRF, outline dietary recommendations for the prevention and management of CRF in cancer survivorship, and identify knowledge gaps and challenges in applying nutritional metabolomics to understand dietary contributions to CRF after a cancer diagnosis.

2. Risk Factors of Cancer-Related Fatigue

Several barriers currently prevent adequate risk assessment and management of CRF: an incomplete understanding of CRF biological mechanisms; the complexity of CRF risk factors; a paucity of effective CRF treatments beyond existing pharmacological interventions; and the existing interventions being time intensive and difficult to implement [37]. Identifying the biological, genetic, and biobehavioral risk factors for CRF will highlight opportunities to target diet as a tool for CRF prevention and management.

2.1. Biological and Genetic Risk Factors

Numerous biological and genetic mechanisms have been linked to CRF. Cancer and its treatment are associated with increased circulating cytokines [38,39], hypothalamic-pituitary-adrenal axis dysfunction [39,40,41], 5-hydroxytryptamine (5-HT) (serotonin) dysfunction [21,39], and altered adenosine triphosphate (ATP) metabolism [39,42], which are hypothesized to contribute to CRF burden. Preliminary longitudinal studies in cancer cohorts support the hypothesis that inflammatory processes involving single nucleotide polymorphisms (SNPs) in inflammation-related genes (e.g., interleukin-1 beta (IL1ß), in [43] terleukin-6 (IL6), and tumor necrosis factor alpha (TNFα)) are associated with increased risk of CRF [44,45,46,47], although the precise mechanisms remain unclear. In a study of long-term survivors of testicular cancer, higher levels of circulating interleukin-1 receptor antagonist (IL-1ra) and C-Reactive protein (CRP) were found in those survivors with chronic CRF, thus lending support to the hypothesis that low-grade inflammatory processed are involved in chronic CRF [43]. Cytokine dysregulation may also contribute to other nutritional risk factors, including anemia, cachexia, anorexia, and infection, all of which further contribute to fatigue [38,48]. Anemia results in decreased oxygen delivery to tissues, resulting in fatigue, dyspnea, and dizziness [49]. Hypoxia-related organ dysfunction is suspected to be related to anemia or hemoglobin dysfunction, which may cause fatigue [39]. Cachexia has been associated with increased levels of interleukins and TNF-α, and abnormalities in energy metabolism in metabolic organs such as the liver, heart, skeletal muscle, adipose tissue, and the gastrointestinal tract [9,50]. Physical conditions such as anorexia, malnutrition, or infections may interact with affective symptoms (e.g., chemical dependency, irritability, and feelings of hopelessness), further contributing to CRF [50]. Other inflammatory processes linked to CRF include cellular immunity marked by leukocyte elevation, the reactivation of latent viruses in an immune-compromised setting, neuroendocrine alteration involving changes in cortisol metabolism, and autonomic nervous system dysregulation [37]. Preliminary findings exploring genetic pathways in CRF provide support for further characterizing inflammatory pathways in the etiology and management of CRF.

2.2. Biobehavioral Risk Factors

Dietary behaviors are integrally tied to biobehavioral risk factors for CRF, including sleep and physical inactivity. Sleep disturbance (e.g., insomnia, sleep apnea, hypersomnia, parasomnias, and problems with circadian rhythm) is prevalent among cancer survivors with estimates ranging from 17–70% [51]. Sleep has a bi-directional relationship with diet and nutrition. In a randomized clinical trial in breast cancer survivors who completed cancer treatment, a three-month fatigue reduction diet characterized by high intakes of fruit, vegetables, whole grains, and omega-3 fatty acids improved fatigue (p = 0.01) and sleep (p = 0.03) compared with the general health curriculum group [52]. Recent research suggests lower levels of calcium, vitamin C, and selenium are associated with disrupted sleep [53], and lower overall diet quality has been associated with a shorter sleep duration [54]. There is also increasing evidence suggesting sleep has an influence on dietary choices. Both cross-sectional and epidemiologic studies have demonstrated that those who sleep less are more likely to consume energy-rich foods (e.g., refined carbohydrates and fats), fewer vegetables, and have irregular meal patterns [55]. In contrast, foods with high availability of tryptophan (e.g., milk, canned tuna, turkey, oats), a precursor to serotonin and melatonin, may be the most helpful in promoting sleep [55]. The exact mechanism linking sleep disturbance, diet, and CRF remains unclear, but preliminary evidence suggests abnormal cortisol levels, pro-inflammatory biomarkers of inflammation and angiogenesis, and circadian rhythm expression play a role [56,57]. The links among sleep disturbance, diet, and CRF suggest potential benefits of diet intervention as a non-toxic treatment strategy for CRF.
In addition to sleep, physical inactivity has been associated with increased CRF [58,59]. Inadequate dietary intake resulting in involuntary muscle wasting and muscle weakness contribute to CRF through altered ATP metabolism. ATP is a key mediator in generating new muscle tissue and assisting in muscle contractions. Thus, decreased ATP production may play a significant role in the development of CRF [59]. Vitamin D represents another potential link between physical activity and CRF. Vitamin D deficiency is prevalent among obese cancer survivors, and is associated with physical function, musculoskeletal pain, and alterations in body composition [60,61,62,63]. While further research on the mechanisms underlying biobehavioral relationships with CRF is needed, it is likely that biobehavioral risk factors have a synergistic effect on fatigue (e.g., sleep disruption contributes to fatigue, and physical inactivity further exacerbates fatigue).

3. Metabolomic Studies of Cancer-Related Fatigue

Studies identifying metabolic signatures of CRF are emerging across cancer populations, bolstered by an improvement in metabolomics technologies that measure up to thousands of metabolites from low volume biospecimens. Untargeted metabolomics investigations provide an unbiased means of discovering metabolites unique to CRF that could be modulated by dietary factors, thus unveiling potential dietary targets to intervene upon for mitigating or preventing CRF. Pathways of metabolic dysfunction that have been associated with increased fatigue include those consistent with a hypometabolic state (e.g., declines in sphingo- and glycosphingolipids, phospholipids, purines, microbiome aromatic amino acid, branch chain amino acids, and lathosterol) [64]. Numerous metabolic pathways have been associated with CRF in different cancer populations. Mechanisms that may lead to dysregulation include the regulation of ATP production and cellular energy, metabolism, and the steroid hormone biosynthesis pathway [65,66,67].
In a recent longitudinal study of cancer survivors with mixed cancer types, metabolites involved in sphingolipid, histidine, cysteine, and methionine metabolism were associated with increased CRF in cancer survivors as compared to cancer-free individuals [66]. A pilot study of post-treatment colorectal cancer survivors found that fatigue was associated with two metabolomic pathways involved in the regulation of ATP production and cellular energy (i.e., glutathione metabolism, and D-glutamine and D-glutamate metabolism) [67]. Fatigue worsens over time following androgen deprivation therapy (ADT) initiation, in a study of prostate cancer survivors receiving ADT, androstenedione, androgen, estrogen, amino acid, and glutathione metabolic pathways discriminated patients receiving ADT from those not receiving the treatment [65]. Among these metabolites, steroid hormone biosynthesis metabolites were most significantly correlated with fatigue severity [65]. A global metabolomics profiling of cerebrospinal fluid (CSF) in acute lymphoblastic leukemia (ALL) survivors identified eight metabolites associated with fatigue (p < 0.05). Fatigue was positively associated with dimethylglycine (amino acid pathway), allantoin (nucleotide pathway), ribitol (carbohydrate pathway), and dimethylmalonic acid (lipid pathway), while fatigue was negatively associated with gamma-glutamylglutamine (peptide pathway), 3-methoxytyrosine (amino acid pathway), asparagine (amino acid pathway), and myoinositol (lipid pathway) [68]. A pilot study of metabolite profiles among breast cancer survivors found that profiles among those with post-chemotherapy fatigue, compared with pre-chemotherapy fatigue, were characterized by higher concentrations of acetyl-l-alanine (amino acid) and indoxyl sulfate (indole) and lower levels of 5-oxo-l-proline (carboxylic acid) [69]. These findings could be useful for elucidating biological mechanisms associated with the development of pain and fatigue [69]. In a longitudinal study of hepatocellular carcinoma survivors, lenvatinib therapy affected the carnitine system by lowering carnitine levels, and carnitine insufficiency was associated with increased fatigue [70]. Thus, metabolites have been associated with fatigue across numerous cancer populations. Further research comparing metabolic signatures unique to cancer survivors with CRF vs. metabolites that may be associated with, but not unique to, CRF (i.e., overlap pathways) vs. metabolites associated with cancer but not CRF, will strengthen our understanding of metabolomic profiles specific to CRF [66] (Figure 1).

4. Dietary Recommendations for Cancer-Related Fatigue

Despite the burden of CRF among cancer survivors, few studies have investigated the role of diet in the development or management of CRF. Indeed, specific dietary guidelines for cancer survivors have been developed for some cancer types, but they focus on dietary relationships with disease recurrence or mortality rather than optimizing diet to prevent or manage cancer and treatment related side-effects [71]. Existing dietary interventions do not directly target CRF, focusing more indirectly on mechanisms tied to CRF, such as inflammation and lean muscle mass maintenance [31]. The few diet interventions provided prior to, during, and after cancer treatment that may influence CRF have previously been summarized [31]. Moreover, while various patient-reported survey measures for CRF are currently used, objective measures of CRF have not been validated. Identifying objective biomarkers of CRF would strengthen CRF outcome assessment in interventional studies [31].
Nutritional status plays an important role as a mediator of fatigue both directly and indirectly through its impact on underlying physiological mechanisms, such as poor nutritional status and micronutrient deficiencies [72]. Specific nutrition-related mechanisms posited to relate to CRF include chemotherapy-induced malnutrition, anemia, treatment-related nutrient malabsorption and altered metabolism (e.g., mitochondrial dysfunction), involuntary weight loss or gain, modification in body composition (e.g., obesity, undernutrition, and sarcopenia), and difficulty meeting protein-energy requirements in the presence of cancer and/or its treatment [72]. Fatigue is also thought to be a result of the dysphagia-malnutrition relationship [73], and chewing problems including loss of teeth further exacerbate malnutrition and fatigue [74,75]. Alterations in body composition due to cancer (e.g., muscle wasting-associated sarcopenia) or changes in dietary intake (e.g., undernutrition) [76] underscore the value of nutritional interventions targeted to counteract muscle decline in the context of fatigue [72].
Current dietary guidelines for cancer survivors recommend diets high in anti-inflammatory foods such as fruits, vegetables, whole grains, lean proteins, and healthy fats (specifically omega-3 and omega-6 fatty acids) and low in saturated fat and processed meat [77,78]. These dietary patterns after cancer diagnosis have been inversely associated with CRF [36,79,80,81,82,83,84]. In a prospective cohort study of colorectal cancer survivors, higher dietary fiber, fruit, and vegetable intakes were longitudinally associated with better physical functioning and less fatigue from six-weeks to 24 months post-treatment [85]. Macronutrients including omega-3 fatty acids, fiber, and protein, which assists in maintaining and building lean muscle mass, have all been associated with improvements in CRF randomized dietary trials [52,80]. For example, studies have shown lower fatigue among breast cancer survivors with higher dietary omega-3 fatty acid intake [52] and those with a higher fiber intake [80]. Additionally, high protein intake has been found to predict a lower CRF burden in advanced-stage cancer patients undergoing chemotherapy [79]. In contrast, in a review of nutritional interventions for CRF, a high intake of pro-inflammatory refined carbohydrates, including sugar and processed grains, has been associated with increased CRF burden [31]. Furthermore, caloric restriction (CR) of 30% without an incurrence of malnutrition was associated with decreased adiposity and inflammation, and improved insulin sensitivity, blood glucose, inflammation and angiogenesis in non-obese humans, suggesting a feasible intervention for reducing CRF [86]. However, chronic CR is contraindicated for many cancer patients at risk for weight loss, cachexia, and immunosuppression, thus intermittent CR or low carbohydrate/ketogenic diets may be more suitable [86].
More recently, the focus has shifted from individual food components to broader recommendations for overall dietary pattern. The World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) diet and lifestyle guidelines now recommend a diet rich in whole grains, vegetables, fruits, and beans, with limited consumption of processed foods, red meat, sugar-sweetened drinks, and alcohol [87]. This dietary pattern largely aligns with the Dietary Guidelines for Americans, and other chronic disease-specific dietary guidelines [88,89,90]. Robust systematic reviews with dose-response meta-analyses contributed to the evidence-base supporting these guidelines. Dietary patterns associated with CRF, specifically, have been reviewed elsewhere. Briefly, anti-inflammatory dietary patterns (e.g., the Mediterranean diet [91,92], ketogenic diet [93,94], macrobiotic diet [80,95], Paleolithic diet [92], and other plant-based diets [80,96]) have been associated with reduced fatigue, possibly by lowering inflammation and improving gut microbial characteristics. Additionally, two recent studies [97,98] identified distinct gut microbial taxa associated with high compared with low fatigue among cancer survivors of multiple cancer types. Dietary patterns are one of the strongest factors influencing the gut microbiome [99]. The gut microbiome significantly influences immune response with the capacity to induce inflammation, which is relevant to the etiology of CRF [100]. Future studies that evaluate how the gut microbiome may mediate the relationship between diet and CRF are warranted.
Indeed, adherence to the WCRF/AICR diet and lifestyle guidelines has been associated with a decreased fatigue burden in several studies [101,102,103]. The alteration of dietary patterns after cancer diagnosis (i.e., increasing consumption of fruits, vegetables, fish, nuts and seeds) could be more important than total energy and/or protein intake prescriptions for reducing CRF [52]. A 2019 systematic review and meta-analysis of nutrition therapy for CRF and quality of life lent further support to the potential benefits of anti-inflammatory dietary patterns (i.e., high intake of fruits, vegetables, fish, nuts and seeds) in reducing CRF, however evidence to determine the optimal nutrition care plan to improve CRF and quality of life is insufficient [104]. A 2020 study among Malay breast cancer survivors found that an increased intake of green leafy and cruciferous vegetables and reduced intake of red meat, sugar, and fried cooking methods were associated with improved emotional function and reduced fatigue [105].
The WCRF/AICR diet and lifestyle guidelines recommend meeting nutritional needs through diet alone, and advise against using high-dose dietary supplements for cancer prevention. The guidelines advise cancer patients to follow the same recommendations as for cancer prevention. Dietary-supplement use in cancer survivors is common [106]; however, evidence supporting its beneficial health effects is limited. Only a handful of studies to date have examined associations between dietary supplement use and fatigue in cancer survivors, and these few studies show mixed results [107,108,109]. In a phase II nationwide study, among cancer survivors who supplemented with fish oil, greater increases in serum omega-3s were associated with greater improvements in fatigue [109]. Among colorectal cancer survivors using Vitamin B6 supplements, higher circulating pyridoxal 5′-phosphate (PLP), the active form of Vitamin B6, was cross-sectionally associated with reduced fatigue and improved physical and social functioning 6 months post-diagnosis [107]. However, a longitudinal study among colorectal cancer survivors showed no differences in fatigue between dietary supplement users (predominately multivitamins) and nonusers [108]. In contrast, the use of oral complementary and alternative medicine (OCAM), defined as taking homeopathy, vitamins/minerals, or herbal/dietary supplements, was associated with more severe CRF in stage I–III breast cancer survivors [110].

5. Challenges of Nutritional Metabolomics in Cancer-Related Fatigue

Nutritional interventions promoting dietary patterns that target CRF-relevant pathways such as inflammation provide a promising therapy for cancer survivors to prevent or manage CRF. However, few interventions have been conducted to date. Observational studies that evaluate dietary relationships with CRF remain limited by challenges of imprecise dietary intake assessment, and few cancer cohorts have implemented dietary assessments longitudinally after cancer diagnosis. Nutritional metabolomics has been employed to identify objective dietary biomarkers to reduce dietary measurement error and biases associated with dietary self-report [111,112,113,114,115,116,117,118,119]. Moreover, studies of nutritional metabolomics in relation to cancer risk and outcomes have provided objective validation of dietary relationships with cancer, and unveiled novel metabolic pathways linking diet to cancer [120].
Dietary intake is multi-dimensional and dynamic. Emerging evidence suggests that cancer survivors change their diet after diagnosis [121], particularly those experiencing physical symptoms [122]. Yet, few studies have characterized dietary intake across the cancer survivorship trajectory (pre-treatment, peri-treatment (0–6 months), early post treatment (6–24 months), and late-post treatment (24–36 months)) in conjunction with measuring cancer-relevant endpoints, including CRF. Future longitudinal studies will contribute immensely to the evidence base on diet intake history and relationships between CRF and dietary changes across survivorship. By leveraging validated food intake biomarkers, nutritional metabolomics offers a means of studying dietary changes among cancer survivors in the absence of self-reported questionnaires, which are difficult to implement in populations experiencing severe illness. Furthermore, nutritional metabolites identified in recent studies demonstrate greater specificity and more robust diet-biomarker associations than prior dietary biomarkers [123]. The utility of food biomarkers in a dietary interventional setting extends as a tool for monitoring compliance to dietary intervention, which is a recognized challenge. Yet, their use in measuring diet in cancer survivor populations is limited.
Nutritional interventions for cancer survivors with CRF represent a new area of research. Given the application of nutritional metabolomics to better understand the role of diet in cancer risk [120], a similar application could be used to understand the role of diet in the etiology and/or management of CRF. Untargeted metabolomics (i.e., identifying all measurable metabolites in a sample) with machine learning approaches could be used to identify groups of metabolites that distinguish CRF from patients without fatigue, and subsequent analyses could identify food combinations or dietary patterns associated with those metabolic profiles. In a similar vein, endogenous metabolites associated with CRF dietary patterns could unveil potential metabolic mediators. Secondly, a priori-defined food biomarkers that have reached an appropriate level of validation [124] could be measured in association with CRF to test specific diet exposure-CRF hypotheses. For example, biomarkers of coffee (quinate, 3-hydroxypyridine sulfate, 1,3-dimethylurate), alcohol (ethyl glucuronide), multivitamins (pantothenate (B5), pyridoxal, alpha-tocopherol) and citrus fruits (proline betaine) have been validated in large-scale cohort studies and subsequently tested in acute dietary intervention or feeding studies [116,125,126,127,128,129,130,131]. Leveraging nutritional metabolomics data derived from other types of biospecimens (e.g., fecal metabolites) may also provide novel insights, such as diet-microbiome interaction in relation to CRF.
Dietary pattern analysis is increasingly recognized as an important method to account for the totality of diet, the complexity of its influence on biochemical pathways, and, ultimately, on health and symptom development [132]. Pre-defined indices of diet quality have been associated with metabolites in several studies, underscoring the endogenous pathways possibly influenced by a certain pattern of eating and, and the specific dietary index components (i.e., foods) that may drive associations with disease outcomes (e.g., the Healthy Eating Index-2015 (HEI-2015), scoring adherence to the Dietary Guidelines for Americans [133,134,135] and the Alternative Mediterranean Diet (aMED) [136,137,138,139]). Empirical, hypothesis-oriented dietary patterns are developed using reduced rank regression and dimensionality reduction approaches. They are characterized by groups of specific foods that, when consumed together, have the strongest association with specific biomarkers. Such dietary patterns have been developed and validated in different populations to explain variations in inflammatory and insulinemic biomarkers [35,140]. A similar approach could be used to develop an empirical dietary index for CRF, following the identification of validated CRF biomarkers [35,140,141,142]. A next step would be testing such a dietary pattern in dietary interventions for the prevention or management of CRF in cancer patients.

6. Conclusions

CRF is prevalent and debilitating for cancer survivors. In the last decade, there has been a surge in research focused on identifying CRF risk factors and potential interventions to target these etiological factors [143]. Studies examining links between CRF and dietary inflammation largely support the hypothesis that inflammatory processes contribute to fatigue during and after cancer treatment [5]. Currently, our understanding and management of CRF is limited by unclear etiological mechanisms, a dearth of evidence-based guidelines for the nutritional management of CRF, and weak epidemiologic evidence for associations between CRF and dietary factors. The application of untargeted nutritional metabolomics with machine learning approaches could identify groups of metabolites that distinguish survivors with CRF from survivors without fatigue. Subsequent analytic approaches could identify food combinations and dietary patterns associated with unique CRF metabolic profiles, thus informing dietary intervention research to specifically target CRF. Harnessing metabolomics to examine the role of dietary biomarkers in CRF could facilitate an objective dietary assessment, mitigate imprecision related to self-reported diet, and identify mechanisms underlying the association of diet with CRF. A nutritional metabolomics approach to understanding the etiology of CRF could in turn strengthen evidence-based dietary guidelines for cancer survivors and optimize CRF interventions to reduce symptom burden.

Author Contributions

Conceptualization: S.L.C. and M.C.P.; writing—original draft preparation, S.L.C., M.C.P., H.S.L.J. and J.C.F.; review and editing, S.L.C., M.C.P., L.M.G., J.O., B.G., L.G., A.R.P., A.G.S., H.S.L.J. and J.C.F.; visualization, L.M.G.; funding acquisition, J.C.F. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the National Cancer Institute NR018762 (principal investigators Jane C. Figueiredo and Heather S. L. Jim); a T32CA090314 (multiple principal investigators Thomas Brandon and Susan Vadaparampil); and Jennifer Ose was supported by National Institutes of Health (NIH), U01CA206110, R01CA207371, R01189184, and R01CA254108.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

Heather S. L. Jim is a consultant for RedHill BioPharma, Janssen Scientific Affairs, and Merck.

References

  1. American Cancer Society. Survivorship: During and After Treatment. Available online: https://www.cancer.org/treatment/survivorship-during-and-after-treatment.html (accessed on 17 March 2022).
  2. Berger, A.M.; Mooney, K.; Alvarez-Perez, A.; Breitbart, W.S.; Carpenter, K.M.; Cella, D.; Cleeland, C.; Dotan, E.; Eisenberger, M.A.; Escalante, C.P.; et al. Cancer-Related Fatigue, Version 2.2015. J. Natl. Compr. Cancer Netw. 2015, 13, 1012–1039. [Google Scholar] [CrossRef] [PubMed]
  3. Ma, Y.; He, B.; Jiang, M.; Yang, Y.; Wang, C.; Huang, C.; Han, L. Prevalence and risk factors of cancer-related fatigue: A systematic review and meta-analysis. Int. J. Nurs. Stud. 2020, 111, 103707. [Google Scholar] [CrossRef] [PubMed]
  4. Barsevick, A.M.; Irwin, M.; Hinds, P.; Miller, A.; Berger, A.; Jacobsen, P.; Ancoli-Israel, S.; Reeve, B.B.; Mustian, K.; O’Mara, A.; et al. Recommendations for High-Priority Research on Cancer-Related Fatigue in Children and Adults. JNCI J. Natl. Cancer Inst. 2013, 105, 1432–1440. [Google Scholar] [CrossRef] [PubMed]
  5. Bower, J.E. Cancer-related fatigue: Links with inflammation in cancer patients and survivors. Brain Behav. Immun. 2007, 21, 863–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Curt, G.A.; Breitbart, W.; Cella, D.; Groopman, J.E.; Horning, S.J.; Itri, L.M.; Johnson, D.H.; Miaskowski, C.; Scherr, S.L.; Portenoy, R.K.; et al. Impact of Cancer-Related Fatigue on the Lives of Patients: New Findings From the Fatigue Coalition. Oncologist 2000, 5, 353–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Bower, J.E.; Ganz, P.A.; Desmond, K.A.; Rowland, J.H.; Meyerowitz, B.E.; Belin, T.R. Fatigue in Breast Cancer Survivors: Occurrence, Correlates, and Impact on Quality of Life. J. Clin. Oncol. 2000, 18, 743–753. [Google Scholar] [CrossRef] [PubMed]
  8. Vogelzang, N.J.; Breitbart, W.; Cella, D.; A Curt, G.; E Groopman, J.; Horning, S.J.; Itri, L.M.; Johnson, D.H.; Scherr, S.L.; Portenoy, R.K. Patient, caregiver, and oncologist perceptions of cancer-related fatigue: Results of a tripart assessment survey. The Fatigue Coalition. Semin. Hematol. 1997, 34, 4–12. [Google Scholar] [PubMed]
  9. Stasi, R.; Abriani, L.; Beccaglia, P.; Terzoli, E.; Amadori, S. Cancer-related fatigue: Evolving concepts in evaluation and treatment. Cancer 2003, 98, 1786–1801. [Google Scholar] [CrossRef] [PubMed]
  10. Palesh, O.; Scheiber, C.; Kesler, S.; Mustian, K.; Koopman, C.; Schapira, L. Management of side effects during and post-treatment in breast cancer survivors. Breast J. 2017, 24, 167–175. [Google Scholar] [CrossRef] [PubMed]
  11. Ahlberg, K.; Ekman, T.; Gaston-Johansson, F.; Mock, V. Assessment and management of cancer-related fatigue in adults. Lancet 2003, 362, 640–650. [Google Scholar] [CrossRef]
  12. Kessels, E.; Husson, O.; Van der Feltz-Cornelis, C.M. The effect of exercise on cancer-related fatigue in cancer survivors: A systematic review and meta-analysis. Neuropsychiatr. Dis. Treat. 2018, 14, 479–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Meneses-Echávez, J.F.; González-Jiménez, E.; Ramírez-Vélez, R. Effects of supervised exercise on cancer-related fatigue in breast cancer survivors: A systematic review and meta-analysis. BMC Cancer 2015, 15, 77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. van Waart, H.; van Harten, W.H.; Buffart, L.M.; Sonke, G.S.; Stuiver, M.M.; Aaronson, N.K. Why do patients choose (not) to participate in an exercise trial during adjuvant chemotherapy for breast cancer? Psycho-Oncology 2016, 25, 964–970. [Google Scholar] [CrossRef] [PubMed]
  15. Blaney, J.M.; Lowe-Strong, A.; Rankin-Watt, J.; Campbell, A.; Gracey, J.H. Cancer survivors’ exercise barriers, facilitators and preferences in the context of fatigue, quality of life and physical activity participation: A questionnaire-survey. Psycho-Oncology 2013, 22, 186–194. [Google Scholar] [CrossRef]
  16. Goedendorp, M.M.; Gielissen, M.F.; Verhagen, C.A.; Bleijenberg, G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database Syst. Rev. 2009, 2009, CD006953. [Google Scholar] [CrossRef] [Green Version]
  17. Duijts, S.F.A.; Faber, M.M.; Oldenburg, H.S.A.; Van Beurden, M.; Aaronson, N.K. Effectiveness of behavioral techniques and physical exercise on psychosocial functioning and health-related quality of life in breast cancer patients and survivors-a meta-analysis. Psycho-Oncology 2011, 20, 115–126. [Google Scholar] [CrossRef]
  18. Jacobsen, P.B.; Donovan, K.A.; Vadaparampil, S.T.; Small, B.J. Systematic review and meta-analysis of psychological and activity-based interventions for cancer-related fatigue. Health Psychol. 2007, 26, 660–667. [Google Scholar] [CrossRef] [Green Version]
  19. Orrow, G.R.; Hickok, J.T.; Roscoe, J.A.; Raubertas, R.F.; Andrews, P.L.; Flynn, P.J.; Hynes, H.E.; Banerjee, T.K.; Kirshner, J.J.; King, D.K. Differential Effects of Paroxetine on Fatigue and Depression: A Randomized, Double-Blind Trial From the University of Rochester Cancer Center Community Clinical Oncology Program. J. Clin. Oncol. 2003, 21, 4635–4641. [Google Scholar]
  20. Stockler, M.R.; O’Connell, R.; Nowak, A.K.; Goldstein, D.; Turner, J.; Wilcken, N.R.; Wyld, D.; Abdi, E.A.; Glasgow, A.; Beale, P.J.; et al. Effect of sertraline on symptoms and survival in patients with advanced cancer, but without major depression: A placebo-controlled double-blind randomised trial. Lancet Oncol. 2007, 8, 603–612. [Google Scholar] [CrossRef]
  21. Roscoe, J.A.; Morrow, G.R.; Hickok, J.T.; Mustian, K.M.; Griggs, J.J.; Matteson, S.E.; Bushunow, P.; Qazi, R.; Smith, B. Effect of paroxetine hydrochloride (Paxil®) on fatigue and depression in breast cancer patients receiving chemotherapy. Breast Cancer Res. Treat. 2005, 89, 243–249. [Google Scholar] [CrossRef]
  22. Berenson, J.R.; Yellin, O.; Shamasunder, H.K.; Chen, C.-S.; Charu, V.; Woliver, T.B.; Sanani, S.; Schlutz, M.; Nassir, Y.; Swift, R.A.; et al. A phase 3 trial of armodafinil for the treatment of cancer-related fatigue for patients with multiple myeloma. Support. Care Cancer 2014, 23, 1503–1512. [Google Scholar] [CrossRef] [PubMed]
  23. Jean-Pierre, P.; Morrow, G.R.; Roscoe, J.A.; Heckler, C.; Mohile, S.; Janelsins, M.; Peppone, L.; Hemstad, A.; Esparaz, B.T.; Hopkins, J.O. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: A University of Rochester Cancer Center Community Clinical Oncology Program Research base study. Cancer 2010, 116, 3513–3520. [Google Scholar] [PubMed]
  24. Spathis, A.; Fife, K.; Blackhall, F.; Dutton, S.; Bahadori, R.; Wharton, R.; O’Brien, M.; Stone, P.; Benepal, T.; Bates, N.; et al. Modafinil for the Treatment of Fatigue in Lung Cancer: Results of a Placebo-Controlled, Double-Blind, Randomized Trial. J. Clin. Oncol. 2014, 32, 1882–1888. [Google Scholar] [CrossRef] [PubMed]
  25. Qu, D.; Zhang, Z.; Yu, X.; Zhao, J.; Qiu, F.; Huang, J. Psychotropic drugs for the management of cancer-related fatigue: A systematic review and meta-analysis. Eur. J. Cancer Care 2016, 25, 970–979. [Google Scholar] [CrossRef] [PubMed]
  26. Gong, S.; Sheng, P.; Jin, H.; He, H.; Qi, E.; Chen, W.; Dong, Y.; Hou, L. Effect of Methylphenidate in Patients with Cancer-Related Fatigue: A Systematic Review and Meta-Analysis. PLoS ONE 2014, 9, e84391. [Google Scholar] [CrossRef] [PubMed]
  27. Bruera, E.; Yennurajalingam, S.; Palmer, J.L.; Perez-Cruz, P.E.; Frisbee-Hume, S.; Allo, J.A.; Williams, J.L.; Cohen, M.Z. Methylphenidate and/or a Nursing Telephone Intervention for Fatigue in Patients With Advanced Cancer: A Randomized, Placebo-Controlled, Phase II Trial. J. Clin. Oncol. 2013, 31, 2421–2427. [Google Scholar] [CrossRef] [Green Version]
  28. Moraska, A.R.; Sood, A.; Dakhil, S.R.; Sloan, J.A.; Barton, D.; Atherton, P.J.; Suh, J.J.; Griffin, P.C.; Johnson, D.B.; Ali, A.; et al. Phase III, Randomized, Double-Blind, Placebo-Controlled Study of Long-Acting Methylphenidate for Cancer-Related Fatigue: North Central Cancer Treatment Group NCCTG-N05C7 Trial. J. Clin. Oncol. 2010, 28, 3673–3679. [Google Scholar] [CrossRef] [Green Version]
  29. Breitbart, W.; Alici, Y. Psychostimulants for Cancer-Related Fatigue. J. Natl. Compr. Cancer Netw. 2010, 8, 933–942. [Google Scholar] [CrossRef] [Green Version]
  30. Begley, S.; Rose, K.; O’Connor, M. The use of corticosteroids in reducing cancer-related fatigue: Assessing the evidence for clinical practice. Int. J. Palliat. Nurs. 2016, 22, 5–9. [Google Scholar] [CrossRef] [PubMed]
  31. Inglis, J.E.; Lin, P.-J.; Kerns, S.L.; Kleckner, I.R.; Kleckner, A.S.; A Castillo, D.; Mustian, K.M.; Peppone, L.J. Nutritional Interventions for Treating Cancer-Related Fatigue: A Qualitative Review. Nutr. Cancer 2019, 71, 21–40. [Google Scholar] [CrossRef]
  32. Maruvada, P.; Lampe, J.W.; Wishart, D.S.; Barupal, D.; Chester, D.N.; Dodd, D.; Feunang, Y.D.; Dorrestein, P.C.; O Dragsted, L.; Draper, J.; et al. Perspective: Dietary Biomarkers of Intake and Exposure—Exploration with Omics Approaches. Adv. Nutr. Int. Rev. J. 2019, 11, 200–215. [Google Scholar] [CrossRef] [PubMed]
  33. Harmon, B.E.; Boushey, C.J.; Shvetsov, Y.B.; Ettienne, R.; Reedy, J.; Wilkens, L.R.; Le Marchand, L.; Henderson, B.E.; Kolonel, L.N. Associations of key diet-quality indexes with mortality in the Multiethnic Cohort: The Dietary Patterns Methods Project. Am. J. Clin. Nutr. 2015, 101, 587–597. [Google Scholar] [CrossRef] [Green Version]
  34. National Cancer Institute. Metabolomics. Available online: https://dceg.cancer.gov/research/how-we-study/metabolomics (accessed on 24 February 2022).
  35. Tabung, F.K.; Smith-Warner, S.A.; Chavarro, J.E.; Fung, T.T.; Hu, F.B.; Willett, W.C.; Giovannucci, E.L. An Empirical Dietary Inflammatory Pattern Score Enhances Prediction of Circulating Inflammatory Biomarkers in Adults. J. Nutr. 2017, 147, 1567–1577. [Google Scholar] [CrossRef] [Green Version]
  36. Baguley, B.J.; Bolam, K.A.; Wright, O.R.L.; Skinner, T.L. The Effect of Nutrition Therapy and Exercise on Cancer-Related Fatigue and Quality of Life in Men with Prostate Cancer: A Systematic Review. Nutrients 2017, 9, 1003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Bower, J.E. Cancer-related fatigue—Mechanisms, risk factors, and treatments. Nat. Rev. Clin. Oncol. 2014, 11, 597–609. [Google Scholar] [CrossRef]
  38. Barsevick, A.; Geneqol, C.; Frost, M.; Zwinderman, A.; Hall, P.; Halyard, M. I’m so tired: Biological and genetic mechanisms of cancer-related fatigue. Qual. Life Res. 2010, 19, 1419–1427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Ryan, J.L.; Carroll, J.K.; Ryan, E.P.; Mustian, K.M.; Fiscella, K.; Morrow, G.R. Mechanisms of Cancer-Related Fatigue. Oncologist 2007, 12, 22–34. [Google Scholar] [CrossRef] [Green Version]
  40. Bender, C.M.; Sereika, S.M.; Brufsky, A.M.; Ryan, C.; Vogel, V.G.; Rastogi, P.; Cohen, S.; Casillo, F.E.; Berga, S.L. Memory impairments with adjuvant anastrozole versus tamoxifen in women with early-stage breast cancer. Menopause 2007, 14, 995–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Bower, J.E.; Ganz, P.; Aziz, N. Altered Cortisol Response to Psychologic Stress in Breast Cancer Survivors with Persistent Fatigue. Psychosom. Med. 2005, 67, 277–280. [Google Scholar] [CrossRef] [Green Version]
  42. Agteresch, H.J.; Dagnelie, P.C.; Van Der Gaast, A.; Stijnen, T.; Wilson, J.H.P. Randomized Clinical Trial of Adenosine 5′-Triphosphate in Patients With Advanced Non-Small-Cell Lung Cancer. J. Natl. Cancer Inst. 2000, 92, 321–328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Orre, I.J.; Murison, R.; Dahl, A.A.; Ueland, T.; Aukrust, P.; Fosså, S.D. Levels of circulating interleukin-1 receptor antagonist and C-reactive protein in long-term survivors of testicular cancer with chronic cancer-related fatigue. Brain Behav. Immun. 2009, 23, 868–874. [Google Scholar] [CrossRef] [PubMed]
  44. Jim, H.S.; Park, J.Y.; Permuth-Wey, J.; Rincon, M.A.; Phillips, K.M.; Small, B.J.; Jacobsen, P.B. Genetic predictors of fatigue in prostate cancer patients treated with androgen deprivation therapy: Preliminary findings. Brain Behav. Immun. 2012, 26, 1030–1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Miaskowski, C.; Dodd, M.; Lee, K.; West, C.; Paul, S.M.; Cooper, B.A.; Wara, W.; Swift, P.S.; Dunn, L.B.; Aouizerat, B.E. Preliminary Evidence of an Association Between a Functional Interleukin-6 Polymorphism and Fatigue and Sleep Disturbance in Oncology Patients and Their Family Caregivers. J. Pain Symptom Manag. 2010, 40, 531–544. [Google Scholar] [CrossRef]
  46. Aouizerat, B.E.; Dodd, M.; Lee, K.; West, C.; Paul, S.M.; Cooper, B.A.; Wara, W.; Swift, P.; Dunn, L.B.; Miaskowski, C. Preliminary Evidence of a Genetic Association Between Tumor Necrosis Factor Alpha and the Severity of Sleep Disturbance and Morning Fatigue. Biol. Res. Nurs. 2009, 11, 27–41. [Google Scholar] [CrossRef] [PubMed]
  47. Collado-Hidalgo, A.; Bower, J.E.; Ganz, P.A.; Irwin, M.; Cole, S.W. Cytokine gene polymorphisms and fatigue in breast cancer survivors: Early findings. Brain Behav. Immun. 2008, 22, 1197–1200. [Google Scholar] [CrossRef] [Green Version]
  48. Kurzrock, R. The role of cytokines in cancer-related fatigue. Cancer 2001, 92, 1684–1688. [Google Scholar] [CrossRef]
  49. Cella, D. Quality of life and clinical decisions in chemotherapy-induced anemia. Oncologist 2006, 20, 25–28. [Google Scholar]
  50. Wang, X.S. Pathophysiology of Cancer-Related Fatigue. Clin. J. Oncol. Nurs. 2008, 12, 11–20. [Google Scholar] [CrossRef] [PubMed]
  51. Davis, M.P.; Goforth, H.W. Long-term and Short-term Effects of Insomnia in Cancer and Effective Interventions. Cancer J. 2014, 20, 330–344. [Google Scholar] [CrossRef] [PubMed]
  52. Zick, S.M.; Colacino, J.; Cornellier, M.L.; Khabir, T.; Surnow, K.; Djuric, Z. Fatigue reduction diet in breast cancer survivors: A pilot randomized clinical trial. Breast Cancer Res. Treat. 2017, 161, 299–310. [Google Scholar] [CrossRef] [PubMed]
  53. Grandner, M.A.; Jackson, N.; Gerstner, J.; Knutson, K. Sleep symptoms associated with intake of specific dietary nutrients. J. Sleep Res. 2014, 23, 22–34. [Google Scholar] [CrossRef] [Green Version]
  54. Stern, J.H.; Grant, A.S.; Thomson, C.A.; Tinker, L.; Hale, L.; Brennan, K.M.; Woods, N.F.; Chen, Z. Short sleep duration is associated with decreased serum leptin, increased energy intake and decreased diet quality in postmenopausal women. Obesity 2014, 22, E55–E61. [Google Scholar] [CrossRef] [PubMed]
  55. Peuhkuri, K.; Sihvola, N.; Korpela, R. Diet promotes sleep duration and quality. Nutr. Res. 2012, 32, 309–319. [Google Scholar] [CrossRef] [PubMed]
  56. Palesh, O.; Zeitzer, J.M.; Conrad, A.; Giese-Davis, J.; Mustian, K.M.; Popek, V.; Nga, K.; Spiegel, D. Vagal regulation, cortisol, and sleep disruption in women with metastatic breast cancer. J. Clin. Sleep Med. 2008, 4, 441–449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Himbert, C.; Ose, J.; Lin, T.; Warby, C.A.; Gigic, B.; Steindorf, K.; Schrotz-King, P.; Abbenhardt-Martin, C.; Zielske, L.; Boehm, J.; et al. Inflammation- and angiogenesis-related biomarkers are correlated with cancer-related fatigue in colorectal cancer patients: Results from the ColoCare Study. Eur. J. Cancer Care 2019, 28, e13055. [Google Scholar] [CrossRef] [PubMed]
  58. Winters-Stone, K.M.; Bennett, J.A.; Nail, L.; Schwartz, A. Strength, Physical Activity, and Age Predict Fatigue in Older Breast Cancer Survivors. Oncol. Nurs. Forum 2008, 35, 815–821. [Google Scholar] [CrossRef] [PubMed]
  59. Mustian, K.M.; Peppone, L.J.; Palesh, O.G.; Janelsins, M.C.; Mohile, S.G.; Purnell, J.Q.; Darling, T.V. Exercise and Cancer-related Fatigue. US Oncol. 2009, 5, 20–23. [Google Scholar] [CrossRef] [Green Version]
  60. Peppone, L.J.; Rickles, A.S.; Janelsins, M.C.; Insalaco, M.R.; Skinner, K.A. The Association Between Breast Cancer Prognostic Indicators and Serum 25-OH Vitamin D Levels. Ann. Surg. Oncol. 2012, 19, 2590–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Khan, Q.J.; Kimler, B.F.; Reddy, P.S.; Sharma, P.; Klemp, J.R.; Nydegger, J.L.; Yeh, H.-W.; Fabian, C.J. Randomized trial of vitamin D3 to prevent worsening of musculoskeletal symptoms in women with breast cancer receiving adjuvant letrozole. The VITAL trial. Breast Cancer Res. Treat. 2017, 166, 491–500. [Google Scholar] [CrossRef] [PubMed]
  62. Armstrong, T.; Walters, E.; Varshney, S.; Johnson, C. Deficiencies of micronutrients, altered bowel function, and quality of life during late follow-up after pancreaticoduodenectomy for malignancy. Pancreatology 2002, 2, 528–534. [Google Scholar] [CrossRef] [PubMed]
  63. Khan, Q.J.; Reddy, P.S.; Kimler, B.F.; Sharma, P.; Baxa, S.E.; O’Dea, A.P.; Klemp, J.R.; Fabian, C.J. Effect of vitamin D supplementation on serum 25-hydroxy vitamin D levels, joint pain, and fatigue in women starting adjuvant letrozole treatment for breast cancer. Breast Cancer Res. Treat. 2009, 119, 111–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Naviaux, R.K.; Naviaux, J.C.; Li, K.; Bright, A.T.; Alaynick, W.A.; Wang, L.; Baxter, A.; Nathan, N.; Anderson, W.; Gordon, E. Metabolic features of chronic fatigue syndrome. Proc. Natl. Acad. Sci. USA 2016, 113, E5472–E5480. [Google Scholar] [CrossRef] [Green Version]
  65. Feng, L.R.; Barb, J.J.; Allen, H.; Regan, J.; Saligan, L. Steroid Hormone Biosynthesis Metabolism Is Associated with Fatigue Related to Androgen Deprivation Therapy for Prostate Cancer. Front. Cell Dev. Biol. 2021, 9, 642307. [Google Scholar] [CrossRef]
  66. Feng, L.R.; Barb, J.J.; Regan, J.; Saligan, L.N. Plasma metabolomic profile associated with fatigue in cancer patients. Cancer Med. 2021, 10, 1623–1633. [Google Scholar] [CrossRef]
  67. Chou, Y.-J.; Kober, K.M.; Kuo, C.-H.; Yeh, K.-H.; Kuo, T.-C.; Tseng, Y.J.; Miaskowski, C.; Liang, J.-T.; Shun, S.-C. A Pilot Study of Metabolomic Pathways Associated With Fatigue in Survivors of Colorectal Cancer. Biol. Res. Nurs. 2021, 23, 42–49. [Google Scholar] [CrossRef] [PubMed]
  68. Brown, A.L.; Sok, P.; Taylor, O.; Woodhouse, J.P.; Bernhardt, M.B.; Raghubar, K.P.; Kahalley, L.S.; Lupo, P.J.; Hockenberry, M.J.; Scheurer, M.E. Cerebrospinal Fluid Metabolomic Profiles Associated With Fatigue During Treatment for Pediatric Acute Lymphoblastic Leukemia. J. Pain Symptom Manag. 2021, 61, 464–473. [Google Scholar] [CrossRef]
  69. Lyon, D.E.; Starkweather, A.; Yao, Y.; Garrett, T.; Kelly, D.L.; Menzies, V.; Dereziński, P.; Datta, S.; Kumar, S.; Jackson-Cook, C. Pilot Study of Metabolomics and Psychoneurological Symptoms in Women with Early Stage Breast Cancer. Biol. Res. Nurs. 2017, 20, 227–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Okubo, H.; Ando, H.; Ishizuka, K.; Kitagawa, R.; Okubo, S.; Saito, H.; Kokubu, S.; Miyazaki, A.; Ikejima, K.; Shiina, S.; et al. Carnitine insufficiency is associated with fatigue during lenvatinib treatment in patients with hepatocellular carcinoma. PLoS ONE 2020, 15, e0229772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. American Institute for Cancer Research. Cancer Survivors. Available online: https://www.wcrf.org/dietandcancer/cancer-survivors/ (accessed on 2 February 2022).
  72. Azzolino, D.; Arosio, B.; Marzetti, E.; Calvani, R.; Cesari, M. Nutritional Status as a Mediator of Fatigue and Its Underlying Mechanisms in Older People. Nutrients 2020, 12, 444. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Ney, D.M.; Weiss, J.M.; Kind, A.J.H.; Robbins, J. Senescent Swallowing: Impact, Strategies, and Interventions. Nutr. Clin. Pract. 2009, 24, 395–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Kossioni, A.E. The Association of Poor Oral Health Parameters with Malnutrition in Older Adults: A Review Considering the Potential Implications for Cognitive Impairment. Nutrients 2018, 10, 1709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Avlund, K.; Schultz-Larsen, K.; Christiansen, N.; Holm-Pedersen, P. Number of Teeth and Fatigue in Older Adults. J. Am. Geriatr. Soc. 2011, 59, 1459–1464. [Google Scholar] [CrossRef] [PubMed]
  76. Elia, M. The Malnutrition Advisory Group consensus guidelines for the detection and management of malnutrition in the community. Nutr. Bull. 2001, 26, 81–83. [Google Scholar] [CrossRef]
  77. Robien, K.; Demark-Wahnefried, W.; Rock, C.L. Evidence-Based Nutrition Guidelines for Cancer Survivors: Current Guidelines, Knowledge Gaps, and Future Research Directions. J. Am. Diet. Assoc. 2011, 111, 368–375. [Google Scholar] [CrossRef]
  78. Rock, C.L.; Doyle, C.; Demark-Wahnefried, W.; Meyerhardt, J.; Courneya, K.S.; Schwartz, A.L.; Bandera, E.V.; Hamilton, K.K.; Grant, B.; McCullough, M.; et al. Nutrition and physical activity guidelines for cancer survivors. CA A Cancer J. Clin. 2012, 62, 242–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Stobäus, N.; Müller, M.J.; Küpferling, S.; Schulzke, J.-D.; Norman, K. Low Recent Protein Intake Predicts Cancer-Related Fatigue and Increased Mortality in Patients with Advanced Tumor Disease Undergoing Chemotherapy. Nutr. Cancer 2015, 67, 818–824. [Google Scholar] [CrossRef] [PubMed]
  80. Guest, D.D.; Evans, E.M.; Rogers, L.Q. Diet components associated with perceived fatigue in breast cancer survivors. Eur. J. Cancer Care 2013, 22, 51–59. [Google Scholar] [CrossRef] [PubMed]
  81. Alfano, C.M.; Imayama, I.; Neuhouser, M.L.; Kiecolt-Glaser, J.K.; Smith, A.W.; Meeske, K.; McTiernan, A.; Bernstein, L.; Baumgartner, K.B.; Ulrich, C.M.; et al. Fatigue, inflammation, and ω-3 and ω-6 fatty acid intake among breast cancer survivors. J. Clin. Oncol. 2012, 30, 1280–1287. [Google Scholar] [CrossRef] [PubMed]
  82. Zick, S.M.; Sen, A.; Han-Markey, T.L.; Harris, R.E. Examination of the Association of Diet and Persistent Cancer-Related Fatigue: A Pilot Study. Oncol. Nurs. Forum 2013, 40, E41–E49. [Google Scholar] [CrossRef] [Green Version]
  83. George, S.M.; Alfano, C.M.; Neuhouser, M.L.; Smith, A.W.; Baumgartner, R.N.; Baumgartner, K.B.; Bernstein, L.; Ballard-Barbash, R. Better postdiagnosis diet quality is associated with less cancer-related fatigue in breast cancer survivors. J. Cancer Surviv. 2014, 8, 680–687. [Google Scholar] [CrossRef] [PubMed]
  84. Huang, X.; Zhang, Q.; Kang, X.; Song, Y.; Zhao, W. Factors associated with cancer-related fatigue in breast cancer patients undergoing endocrine therapy in an urban setting: A cross-sectional study. BMC Cancer 2010, 10, 453. [Google Scholar] [CrossRef] [Green Version]
  85. Kenkhuis, M.F.; van Duinjoven, F.J.B.; van Roekel, E.H.; Breedveld-Peters, J.J.L.; Bruekink, S.O.; Janssen-Heijen, M.L.; Keulen, E.T.P.; Mols, F.; Weijenber, M.P.; Bours, M.J.L. Longitudinal associations of fiber, vegetable, and fruit intake with quality of life and fatigue in colorectal cancer survivors up to 24 months post-treatment. Am. J. Clin. Nutr. 2022, 3, 822–832. [Google Scholar] [CrossRef]
  86. O’Flanagan, C.H.; Smith, L.A.; McDonell, S.; Hursting, S.D. When less may be more: Calorie restriction and response to cancer therapy. BMC Med. 2017, 15, 106. [Google Scholar] [CrossRef] [PubMed]
  87. Shams-White, M.M.; Romaguera, D.; Mitrou, P.; Reedy, J.; Bender, A.; Brockton, N.T. Further Guidance in Implementing the Standardized 2018 World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) Score. Cancer Epidemiol. Biomark. Prev. 2020, 29, 889–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. U.S. Department of Agriculture; U.S. Department of Health and Human Services. Dietary Guidelines for Americans, 2020–2025. Available online: www.DietaryGuidelines.gov (accessed on 24 February 2022).
  89. American Diabetes Association. Available online: https://www.diabetesfoodhub.org/articles/what-is-the-diabetes-plate-method.html#:~:text=The%20Diabetes%20Plate%20Method%20is,you%20need%20is%20a%20plate! (accessed on 24 February 2022).
  90. Lichtenstein, A.H.; Appel, L.J.; Vadiveloo, M.; Hu, F.B.; Kris-Etherton, P.M.; Rebholz, C.M.; Sacks, F.M.; Thorndike, A.N.; Van Horn, L.; Wylie-Rosett, J.; et al. 2021 Dietary Guidance to Improve Cardiovascular Health: A Scientific Statement From the American Heart Association. Circulation 2021, 144, e472–e487. [Google Scholar] [CrossRef] [PubMed]
  91. Haslam, A.; Robb, S.W.; Hébert, J.R.; Huang, H.; Ebell, M.H. Greater adherence to a Mediterranean diet is associated with lower prevalence of colorectal adenomas in men of all races. Nutr. Res. 2017, 48, 76–84. [Google Scholar] [CrossRef] [PubMed]
  92. A Whalen, K.; McCullough, M.L.; Flanders, W.D.; Hartman, T.J.; Judd, S.; Bostick, R.M. Paleolithic and Mediterranean Diet Pattern Scores Are Inversely Associated with Biomarkers of Inflammation and Oxidative Balance in Adults. J. Nutr. 2016, 146, 1217–1226. [Google Scholar] [CrossRef]
  93. Masino, S.A.; Ruskin, D.N. Ketogenic Diets and Pain. J. Child Neurol. 2013, 28, 993–1001. [Google Scholar] [CrossRef] [Green Version]
  94. Cohen, C.W.; Fontaine, K.R.; Arend, R.C.; Soleymani, T.; Gower, B.A. Favorable Effects of a Ketogenic Diet on Physical Function, Perceived Energy, and Food Cravings in Women with Ovarian or Endometrial Cancer: A Randomized, Controlled Trial. Nutrients 2018, 10, 1187. [Google Scholar] [CrossRef] [Green Version]
  95. Harmon, B.E.; Carter, M.; Hurley, T.G.; Shivappa, N.; Teas, J.; Hebert, J.R. Nutrient Composition and Anti-inflammatory Potential of a Prescribed Macrobiotic Diet. Nutr. Cancer 2015, 67, 933–940. [Google Scholar] [CrossRef] [Green Version]
  96. Clarys, P.; Deliens, T.; Huybrechts, I.; Deriemaeker, P.; Vanaelst, B.; De Keyzer, W.; Hebbelinck, M.; Mullie, P. Comparison of Nutritional Quality of the Vegan, Vegetarian, Semi-Vegetarian, Pesco-Vegetarian and Omnivorous Diet. Nutrients 2014, 6, 1318–1332. [Google Scholar] [CrossRef] [PubMed]
  97. Xiao, C.; Fedirko, V.; Beitler, J.; Bai, J.; Peng, G.; Zhou, C.; Gu, J.; Zhao, H.; Lin, I.-H.; Chico, C.E.; et al. The role of the gut microbiome in cancer-related fatigue: Pilot study on epigenetic mechanisms. Support. Care Cancer 2021, 29, 3173–3182. [Google Scholar] [CrossRef] [PubMed]
  98. Hajjar, J.; Mendoza, T.; Zhang, L.; Fu, S.; Piha-Paul, S.A.; Hong, D.S.; Janku, F.; Karp, D.D.; Ballhausen, A.; Gong, J.; et al. Associations between the gut microbiome and fatigue in cancer patients. Sci. Rep. 2021, 11, 5847. [Google Scholar] [CrossRef] [PubMed]
  99. Leeming, E.R.; Johnson, A.J.; Spector, T.D.; Le Roy, C.I. Effect of Diet on the Gut Microbiota: Rethinking Intervention Duration. Nutrients 2019, 11, 2862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Belkaid, Y.; Hand, T.W. Role of the Microbiota in Immunity and Inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
  101. Breedveld-Peters, J.J.L.; Koole, J.L.; Müller-Schulte, E.; van der Linden, B.W.A.; Windhausen, C.; Bours, M.J.L.; van Roekel, E.H.; Weijenberg, M.P. Colorectal cancers survivors’ adherence to lifestyle recommendations and cross-sectional associations with health-related quality of life. Br. J. Nutr. 2018, 120, 188–197. [Google Scholar] [CrossRef]
  102. Kenkhuis, M.-F.; Mols, F.; van Roekel, E.H.; Breedveld-Peters, J.J.L.; Breukink, S.O.; Janssen-Heijnen, M.L.G.; Keulen, E.T.P.; van Duijnhoven, F.J.B.; Weijenberg, M.P.; Bours, M.J.L. Longitudinal Associations of Adherence to the World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) Lifestyle Recommendations with Quality of Life and Symptoms in Colorectal Cancer Survivors up to 24 Months Post-Treatment. Cancers 2022, 14, 417. [Google Scholar] [CrossRef]
  103. Lei, Y.-Y.; Ho, S.C.; Cheng, A.; Kwok, C.; Lee, C.-K.I.; Cheung, K.L.; Lee, R.; Loong, H.H.; He, Y.-Q.; Yeo, W. Adherence to the World Cancer Research Fund/American Institute for Cancer Research Guideline Is Associated with Better Health-Related Quality of Life among Chinese Patients with Breast Cancer. J. Natl. Compr. Cancer Netw. 2018, 16, 275–285. [Google Scholar] [CrossRef]
  104. Baguley, B.J.; Skinner, T.L.; Wright, O. Nutrition therapy for the management of cancer-related fatigue and quality of life: A systematic review and meta-analysis. Br. J. Nutr. 2019, 122, 527–541. [Google Scholar] [CrossRef]
  105. Zainordin, N.H.; Talib, R.A.; Shahril, M.R.; Sulaiman, S.; Karim, N.A. Dietary Changes and Its Impact on Quality of Life among Malay Breast and Gynaecological Cancer Survivors in Malaysia. Asian Pac. J. Cancer Prev. 2020, 21, 3689–3696. [Google Scholar] [CrossRef]
  106. Song, S.; Youn, J.; Lee, Y.J.; Kang, M.; Hyun, T.; Song, Y.; Lee, J.E. Dietary supplement use among cancer survivors and the general population: A nation-wide cross-sectional study. BMC Cancer 2017, 17, 891. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Koole, J.L.; Bours, M.J.L.; Geijsen, A.J.M.R.; Gigic, B.; Ulvik, A.; E Kok, D.; Brezina, S.; Ose, J.; Baierl, A.; Böhm, J.; et al. Circulating B-vitamin biomarkers and B-vitamin supplement use in relation to quality of life in patients with colorectal cancer: Results from the FOCUS consortium. Am. J. Clin. Nutr. 2021, 113, 1468–1481. [Google Scholar] [CrossRef] [PubMed]
  108. Koole, J.L.; Bours, M.J.; Breedveld-Peters, J.J.; van Roekel, E.H.; Breukink, S.O.; Janssen-Heijnen, M.L.; Vogelaar, F.J.; Aquarius, M.; Keulen, E.; Stoot, J.; et al. Is dietary supplement use longitudinally associated with fatigue in stage I–III colorectal cancer survivors? Clin. Nutr. 2020, 39, 234–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Kleckner, A.S.; Culakova, E.; Kleckner, I.R.; Belcher, E.K.; Demark-Wahnefried, W.; Parker, E.A.; Padula, G.D.A.; Ontko, M.; Janelsins, M.C.; Mustian, K.M.; et al. Nutritional Status Predicts Fatty Acid Uptake from Fish and Soybean Oil Supplements for Treatment of Cancer-Related Fatigue: Results from a Phase II Nationwide Study. Nutrients 2021, 14, 184. [Google Scholar] [CrossRef] [PubMed]
  110. Lapidari, P.; Djehal, N.; Havas, J.; Gbenou, A.; Martin, E.; Charles, C.; Dauchy, S.; Pistilli, B.; Cadeau, C.; Bertaut, A.; et al. Determinants of use of oral complementary-alternative medicine among women with early breast cancer: A focus on cancer-related fatigue. Breast Cancer Res. Treat. 2021, 190, 517–529. [Google Scholar] [CrossRef]
  111. Cheung, W.; Keski-Rahkonen, P.; Assi, N.; Ferrari, P.; Freisling, H.; Rinaldi, S.; Slimani, N.; Zamora-Ros, R.; Rundle, M.; Frost, G.; et al. A metabolomic study of biomarkers of meat and fish intake. Am. J. Clin. Nutr. 2017, 105, 600–608. [Google Scholar] [CrossRef]
  112. Edmands, W.M.; Ferrari, P.; Rothwell, J.; Rinaldi, S.; Slimani, N.; Barupal, D.; Biessy, C.; Jenab, M.; Clavel-Chapelon, F.; Fagherazzi, G.; et al. Polyphenol metabolome in human urine and its association with intake of polyphenol-rich foods across European countries. Am. J. Clin. Nutr. 2015, 102, 905–913. [Google Scholar] [CrossRef] [Green Version]
  113. Guertin, K.A.; Loftfield, E.; Boca, S.M.; Sampson, J.N.; Moore, S.C.; Xiao, Q.; Huang, W.-Y.; Xiong, X.; Freedman, N.D.; Cross, A.J.; et al. Serum biomarkers of habitual coffee consumption may provide insight into the mechanism underlying the association between coffee consumption and colorectal cancer. Am. J. Clin. Nutr. 2015, 101, 1000–1011. [Google Scholar] [CrossRef]
  114. Playdon, M.C.; Moore, S.C.; Derkach, A.; Reedy, J.; Subar, A.F.; Sampson, J.N.; Albanes, D.; Gu, F.; Kontto, J.; Lassale, C.; et al. Identifying biomarkers of dietary patterns by using metabolomics. Am. J. Clin. Nutr. 2017, 105, 450–465. [Google Scholar] [CrossRef] [Green Version]
  115. Playdon, M.C.; Sampson, J.N.; Cross, A.J.; Sinha, R.; Guertin, K.; A Moy, K.; Rothman, N.; Irwin, M.L.; Mayne, S.T.; Stolzenberg-Solomon, R.; et al. Comparing metabolite profiles of habitual diet in serum and urine. Am. J. Clin. Nutr. 2016, 104, 776–789. [Google Scholar] [CrossRef] [Green Version]
  116. Scalbert, A.; Brennan, L.; Manach, C.; Andres-Lacueva, C.; O Dragsted, L.; Draper, J.; Rappaport, S.M.; van der Hooft, J.J.; Wishart, D.S. The food metabolome: A window over dietary exposure. Am. J. Clin. Nutr. 2014, 99, 1286–1308. [Google Scholar] [CrossRef] [Green Version]
  117. Schmidt, J.A.; Rinaldi, S.; Ferrari, P.; Carayol, M.; Achaintre, D.; Scalbert, A.; Cross, A.J.; Gunter, M.J.; Fensom, G.K.; Appleby, P.N.; et al. Metabolic profiles of male meat eaters, fish eaters, vegetarians, and vegans from the EPIC-Oxford cohort. Am. J. Clin. Nutr. 2015, 102, 1518–1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Pallister, T.; Jennings, A.; Mohney, R.P.; Yarand, D.; Mangino, M.; Cassidy, A.; MacGregor, A.; Spector, T.D.; Menni, C. Characterizing Blood Metabolomics Profiles Associated with Self-Reported Food Intakes in Female Twins. PLoS ONE 2016, 11, e0158568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Playdon, M.C.; Ziegler, R.G.; Sampson, J.N.; Stolzenberg-Solomon, R.; Thompson, H.J.; Irwin, M.L.; Mayne, S.T.; Hoover, R.N.; Moore, S. Nutritional metabolomics and breast cancer risk in a prospective study. Am. J. Clin. Nutr. 2017, 106, 637–649. [Google Scholar] [CrossRef]
  120. McGee, E.E.; Kiblawi, R.; Playdon, M.C.; Eliassen, A.H. Nutritional Metabolomics in Cancer Epidemiology: Current Trends, Challenges, and Future Directions. Curr. Nutr. Rep. 2019, 8, 187–201. [Google Scholar] [CrossRef] [PubMed]
  121. Sun, Y.; Bao, W.; Liu, B.; Caan, B.J.; Lane, D.S.; Millen, A.E.; Simon, M.S.; Thomson, C.A.; Tinker, L.F.; Van Horn, L.V.; et al. Changes in Overall Diet Quality in Relation to Survival in Postmenopausal Women with Breast Cancer: Results from the Women’s Health Initiative. J. Acad. Nutr. Diet. 2018, 118, 1855–1863. [Google Scholar] [CrossRef] [PubMed]
  122. Trudel-Fitzgerald, C.; Tworoger, S.S.; Poole, E.M.; Zhang, X.; Giovannucci, E.L.; Meyerhardt, J.A.; Kubzansky, L.D. Psychological symptoms and subsequent healthy lifestyle after a colorectal cancer diagnosis. Health Psychol. 2018, 37, 207–217. [Google Scholar] [CrossRef] [PubMed]
  123. Playdon, M.C.; Lampe, J.W.; Tinker, L.F.; Prentice, R.; Hayden, K.M.; Van Horn, L.; Sampson, J.; Stolzenberg-Solomon, R.; Moore, S.C. Objective biomarkers of usual diet: A metabolomics analysis of weighed food intake. Am. J. Clin. Nutr. 2016, 104, 776–789. [Google Scholar] [CrossRef] [Green Version]
  124. Dragsted, L.O.; Gao, Q.; Scalbert, A.; Vergères, G.; Kolehmainen, M.; Manach, C.; Brennan, L.; Afman, L.A.; Wishart, D.S.; Lacueva, C.A.; et al. Validation of biomarkers of food intake—Critical assessment of candidate biomarkers. Genes Nutr. 2018, 13, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Brouwer-Brolsma, E.M.; Brennan, L.; Drevon, C.A.; van Kranen, H.; Manach, C.; Dragsted, L.O.; Roche, H.M.; Andres-Lacueva, C.; Bakker, S.J.L.; Bouwman, J.; et al. Combining traditional dietary assessment methods with novel metabolomics techniques: Present efforts by the Food Biomarker Alliance. Proc. Nutr. Soc. 2017, 76, 619–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Dragsted, L.O.; Gao, Q.; Praticò, G.; Manach, C.; Wishart, D.S.; Scalbert, A.; Feskens, E.J.M. Dietary and health biomarkers—Time for an update. Genes Nutr. 2017, 12, 24. [Google Scholar] [CrossRef] [Green Version]
  127. Ulaszewska, M.; Garcia-Aloy, M.; Vázquez-Manjarrez, N.; Soria-Florido, M.T.; Llorach, R.; Mattivi, F.; Manach, C. Food intake biomarkers for berries and grapes. Genes Nutr. 2020, 15, 17. [Google Scholar] [CrossRef]
  128. Vázquez-Manjarrez, N.; Ulaszewska, M.; Garcia-Aloy, M.; Mattivi, F.; Praticò, G.; Dragsted, L.O.; Manach, C. Biomarkers of intake for tropical fruits. Genes Nutr. 2020, 15, 11. [Google Scholar] [CrossRef] [PubMed]
  129. Brouwer-Brolsma, E.M.; Brandl, B.; Buso, M.E.C.; Skurk, T.; Manach, C. Food intake biomarkers for green leafy vegetables, bulb vegetables, and stem vegetables: A review. Genes Nutr. 2020, 15, 7. [Google Scholar] [CrossRef] [PubMed]
  130. Giesbertz, P.; Brandl, B.; Lee, Y.-M.; Hauner, H.; Daniel, H.; Skurk, T. Specificity, Dose Dependency, and Kinetics of Markers of Chicken and Beef Intake Using Targeted Quantitative LC-MS/MS: A Human Intervention Trial. Mol. Nutr. Food Res. 2020, 64, e1900921. [Google Scholar] [CrossRef]
  131. Ulaszewska, M.; Vázquez-Manjarrez, N.; Garcia-Aloy, M.; Llorach, R.; Mattivi, F.; Dragsted, L.O.; Praticò, G.; Manach, C. Food intake biomarkers for apple, pear, and stone fruit. Genes Nutr. 2018, 13, 29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Reedy, J.; Subar, A.F.; George, S.M.; Krebs-Smith, S.M. Extending Methods in Dietary Patterns Research. Nutrients 2018, 10, 571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Kennedy, E.T.; Ohls, J.; Carlson, S.; Fleming, K. The Healthy Eating Index: Design and applications. J. Am. Diet. Assoc. 1995, 95, 1103–1108. [Google Scholar] [CrossRef]
  134. Guenther, P.M.; Kirkpatrick, S.; Reedy, J.; Krebs-Smith, S.M.; Buckman, D.W.; Dodd, K.W.; Casavale, K.O.; Carroll, R.J. The Healthy Eating Index-2010 Is a Valid and Reliable Measure of Diet Quality According to the 2010 Dietary Guidelines for Americans. J. Nutr. 2014, 144, 399–407. [Google Scholar] [CrossRef] [Green Version]
  135. Guenther, P.M.; Casavale, K.O.; Reedy, J.; Kirkpatrick, S.I.; Hiza, H.A.B.; Kuczynski, K.J.; Kahle, L.L.; Krebs-Smith, S.M. Update of the healthy eating index: HEI-2010. J. Acad. Nutr. Diet. 2013, 113, 569–580. [Google Scholar] [CrossRef] [Green Version]
  136. Trichopoulou, A.; Costacou, T.; Bamia, C.; Trichopoulos, D. Adherence to a Mediterranean Diet and Survival in a Greek Population. N. Engl. J. Med. 2003, 348, 2599–2608. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Trichopoulou, A.; Kouris-Blazos, A.; Wahlqvist, M.L.; Gnardellis, C.; Lagiou, P.; Polychronopoulos, E.; Vassilakou, T.; Lipworth, L.; Trichopoulos, D. Diet and overall survival in elderly people. BMJ 1995, 311, 1457–1460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kim, E.H.J.; Willett, W.C.; Fung, T.; Rosner, B.; Holmes, M.D. Diet Quality Indices and Postmenopausal Breast Cancer Survival. Nutr. Cancer 2011, 63, 381–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Fung, T.T.; McCullough, M.L.; Newby, P.; Manson, J.E.; Meigs, J.B.; Rifai, N.; Willett, W.C.; Hu, F.B. Diet-quality scores and plasma concentrations of markers of inflammation and endothelial dysfunction. Am. J. Clin. Nutr. 2015, 82, 163–173. [Google Scholar] [CrossRef]
  140. Abung, F.K.; Giovannucci, E.L.; Giulianini, F.; Liang, L.; Chandler, P.; Balasubramanian, R.; E Manson, J.; Feliciano, E.M.C.; Hayden, K.M.; Van Horn, L.; et al. An Empirical Dietary Inflammatory Pattern Score Is Associated with Circulating Inflammatory Biomarkers in a Multi-Ethnic Population of Postmenopausal Women in the United States. J. Nutr. 2018, 148, 771–780. [Google Scholar] [CrossRef] [PubMed]
  141. Fu, B.C.; Tabung, F.K.; Pernar, C.H.; Wang, W.; Gonzalez-Feliciano, A.G.; Chowdhury-Paulino, I.M.; Clinton, S.K.; Folefac, E.; Song, M.; Kibel, A.S.; et al. Insulinemic and Inflammatory Dietary Patterns and Risk of Prostate Cancer. Eur. Urol. 2021, 79, 405–412. [Google Scholar] [CrossRef] [PubMed]
  142. Tabung, F.K.; Wang, W.; Fung, T.T.; Hu, F.B.; Smith-Warner, S.A.; Chavarro, J.E.; Fuchs, C.S.; Willett, W.C.; Giovannucci, E.L. Development and validation of empirical indices to assess the insulinaemic potential of diet and lifestyle. Br. J. Nutr. 2016, 116, 1787–1798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Pritlove, C.; Capone, G.; Kita, H.; Gladman, S.; Maganti, M.; Jones, J.M. Cooking for Vitality: Pilot Study of an Innovative Culinary Nutrition Intervention for Cancer-Related Fatigue in Cancer Survivors. Nutrients 2020, 12, 2760. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nutritional Metabolomics in Cancer Survivorship.
Figure 1. Nutritional Metabolomics in Cancer Survivorship.
Nutrients 14 01496 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Crowder, S.L.; Playdon, M.C.; Gudenkauf, L.M.; Ose, J.; Gigic, B.; Greathouse, L.; Peoples, A.R.; Sleight, A.G.; Jim, H.S.L.; Figueiredo, J.C. A Molecular Approach to Understanding the Role of Diet in Cancer-Related Fatigue: Challenges and Future Opportunities. Nutrients 2022, 14, 1496. https://doi.org/10.3390/nu14071496

AMA Style

Crowder SL, Playdon MC, Gudenkauf LM, Ose J, Gigic B, Greathouse L, Peoples AR, Sleight AG, Jim HSL, Figueiredo JC. A Molecular Approach to Understanding the Role of Diet in Cancer-Related Fatigue: Challenges and Future Opportunities. Nutrients. 2022; 14(7):1496. https://doi.org/10.3390/nu14071496

Chicago/Turabian Style

Crowder, Sylvia L., Mary C. Playdon, Lisa M. Gudenkauf, Jennifer Ose, Biljana Gigic, Leigh Greathouse, Anita R. Peoples, Alix G. Sleight, Heather S. L. Jim, and Jane C. Figueiredo. 2022. "A Molecular Approach to Understanding the Role of Diet in Cancer-Related Fatigue: Challenges and Future Opportunities" Nutrients 14, no. 7: 1496. https://doi.org/10.3390/nu14071496

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop