2,012
Views
67
CrossRef citations to date
0
Altmetric
Reviews

Nutritional Interventions for Treating Cancer-Related Fatigue: A Qualitative Review

ORCID Icon, , , , , , & show all
Pages 21-40 | Received 30 May 2018, Accepted 06 Aug 2018, Published online: 26 Jan 2019

References

  • Peoples AR, Roscoe JA, Block RC, Heckler CE, Ryan JL, et al.: Nausea and disturbed sleep as predictors of cancer-related fatigue in breast cancer patients: a multicenter NCORP study. Support Care Cancer 25, 1271–1278, 2017. doi: 10.1007/s00520-016-3520-8
  • Bower JE: Cancer-related fatigue–mechanisms, risk factors, and treatments. Nat Rev Clin Oncol 11, 597–609, 2014. doi: 10.1038/nrclinonc.2014.127
  • Palesh O, Scheiber C, Kesler S, Mustian K, Koopman C, et al.: Management of side effects during and post-treatment in breast cancer survivors. Breast J 24, 167–175, 2017. doi: 10.1111/tbj.12862
  • Borneman T: Assessment and management of cancer-related fatigue. J Hospice Palliative Nurs 15, 77–86, 2013. doi: 10.1097/NJH.0b013e318286dc19
  • Mortimer JE, Waliany S, Dieli-Conwright CM, Patel SK, Hurria A, et al.: Objective physical and mental markers of self-reported fatigue in women undergoing (neo)adjuvant chemotherapy for early-stage breast cancer. Cancer 123, 1810–1816, 2017. doi: 10.1002/cncr.30426
  • Vieira ML, Fonseca FL, Costa LG, Beltrame RL, Chaves CM, et al.: Supplementation with selenium can influence nausea, fatigue, physical, renal, and liver function of children and adolescents with cancer. J Med Food 18, 109–117, 2015. doi: 10.1089/jmf.2014.0030
  • Chandwani KD, Ryan JL, Peppone LJ, Janelsins MM, Sprod LK, et al.: Cancer-related stress and complementary and alternative medicine: a review. Evid Based Complement Alternat Med 2012, 979213, 2012. doi: 10.1155/2012/979213
  • Palesh O, Scheiber C, Kesler S, Mustian K, Koopman C, et al.: Management of side effects during and post-treatment in breast cancer survivors. Breast J 24, 167–175, 2018. doi: 10.1111/tbj.12862
  • Campbell A, Stevinson C, and Crank H: The BASES Expert Statement on exercise and cancer survivorship. J Sports Sci 30, 949–952, 2012. doi: 10.1080/02640414.2012.671953
  • Jankowski CM, and Matthews EE: Exercise guidelines for adults with cancer: a vital role in survivorship. Clin J Oncol Nurs 15, 683–686, 2011. doi: 10.1188/11.Cjon.683-686
  • Beckett DM and Wold LE: Tumour-induced myopathy occurs in both skeletal and cardiac muscle of tumour-bearing mice. 2nd International Cancer Fatigue Symposium: Putting the Pieces Together, Montreal, Quebec. Curr Oncol 17, 2010.
  • Bower JE, Ganz PA, Aziz N, and Fahey JL: Fatigue and proinflammatory cytokine activity in breast cancer survivors. Psychosom Med 64, 604–611, 2002.
  • Bower JE, Ganz PA, Aziz N, Fahey JL, and Cole SW: T-cell homeostasis in breast cancer survivors with persistent fatigue. J Natl Cancer Inst 95, 1165–1168, 2003.
  • Collado-Hidalgo A, Bower JE, Ganz PA, Cole SW, and Irwin MR: Inflammatory biomarkers for persistent fatigue in breast cancer survivors. Clin Cancer Res 12, 2759–2766, 2006. doi: 10.1158/1078-0432.ccr-05-2398
  • Segerstrom SC and Miller GE: Psychological stress and the human immune system: a meta-analytic study of 30 years of inquiry. Psychol Bull 130, 601–630, 2004. doi: 10.1037/0033-2909.130.4.601
  • National Cancer Institute: Nutrition in Cancer Care (PDQ®)–Health Professional Version. Bethesda, MD: National Cancer Institute, 2017.
  • Zietarska M, Krawczyk-Lipiec J, Kraj L, Zaucha R, and Malgorzewicz S: Chemotherapy-related toxicity, nutritional status and quality of life in precachectic oncologic patients with, or without, high protein nutritional support. A prospective, randomized study. Nutrients 9, 2017. doi: 10.3390/nu9101108
  • Demark-Wahnefried W, Campbell KL, and Hayes SC: Weight management and its role in breast cancer rehabilitation. Cancer 118, 2277–2287, 2012. doi: 10.1002/cncr.27466
  • Bossola M: Nutritional interventions in head and neck cancer patients undergoing chemoradiotherapy: a narrative review. Nutrients 7, 265–276, 2015. doi: 10.3390/nu7010265
  • Kilgour RD, Vigano A, Trutschnigg B, Lucar E, Borod M, et al.: Handgrip strength predicts survival and is associated with markers of clinical and functional outcomes in advanced cancer patients. Support Care Cancer 21, 3261–3270, 2013. doi: 10.1007/s00520-013-1894-4
  • Kilgour RD, Vigano A, Trutschnigg B, Hornby L, Lucar E, et al.: Cancer-related fatigue: the impact of skeletal muscle mass and strength in patients with advanced cancer. J Cachexia Sarcopenia Muscle 1, 177–185, 2010. doi: 10.1007/s13539-010-0016-0
  • Coronha AL, Camilo ME, and Ravasco P: [The relevance of body composition in cancer patients: what is the evidence?]. Acta Med Port 24 Suppl 4, 769–778, 2011.
  • Carneiro IP, Mazurak VC, and Prado CM: Clinical implications of sarcopenic obesity in cancer. Curr Oncol Rep 18, 62, 2016. doi: 10.1007/s11912-016-0546-5
  • Prado CM, Cushen SJ, Orsso CE, and Ryan AM: Sarcopenia and cachexia in the era of obesity: clinical and nutritional impact. Proc Nutr Soc 75, 188–198, 2016. doi: 10.1017/s0029665115004279
  • Nyrop KA, Deal AM, Lee JT, Muss HB, Choi SK, et al.: Weight gain in hormone receptor-positive (HR+) early-stage breast cancer: is it menopausal status or something else? Breast Cancer Res Treat, 167, 235–248, 2017. doi: 10.1007/s10549-017-4501-4
  • Makari-Judson G, Braun B, Jerry DJ, and Mertens WC: Weight gain following breast cancer diagnosis: implication and proposed mechanisms. World J Clin Oncol 5, 272–282, 2014. doi: 10.5306/wjco.v5.i3.272
  • Sheean PM, Hoskins K, and Stolley M: Body composition changes in females treated for breast cancer: a review of the evidence. Breast Cancer Res Treat 135, 663–680, 2012. doi: 10.1007/s10549-012-2200-8
  • Finkelstein EA, Chen H, Prabhu M, Trogdon JG, and Corso PS: The relationship between obesity and injuries among U.S. adults. Am J Health Promot 21, 460–468, 2007.
  • George SM, McTiernan A, Villasenor A, Alfano CM, Irwin ML, et al.: Disentangling the body weight-bone mineral density association among breast cancer survivors: an examination of the independent roles of lean mass and fat mass. BMC Cancer 13, 497, 2013. doi: 10.1186/1471-2407-13-497
  • Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, et al.: Prevalence and clinical implications of sarcopenic obesity in patients with solid tumours of the respiratory and gastrointestinal tracts: a population-based study. Lancet Oncol 9, 629–635, 2008. doi: 10.1016/s1470-2045(08)70153-0
  • Ryan AM, Power DG, Daly L, Cushen SJ, Ni Bhuachalla E, et al.: Cancer-associated malnutrition, cachexia and sarcopenia: the skeleton in the hospital closet 40 years later. Proc Nutr Soc 75, 199–211, 2016. doi: 10.1017/s002966511500419x
  • Baguley BJ, Bolam KA, Wright ORL, and Skinner TL: The effect of nutrition therapy and exercise on cancer-related fatigue and quality of life in men with prostate cancer: a systematic review. Nutrients 9, 2017. doi: 10.3390/nu9091003
  • de Vries YC, van den Berg M, de Vries JHM, Boesveldt S, de Kruif J, et al.: Differences in dietary intake during chemotherapy in breast cancer patients compared to women without cancer. Support Care Cancer 25, 2581–2591, 2017. doi: 10.1007/s00520-017-3668-x
  • Malihi Z, Kandiah M, Chan YM, Esfandbod M, Vakili M, et al.: The effect of dietary intake changes on nutritional status in acute leukaemia patients after first induction chemotherapy. Eur J Cancer Care (Engl) 24, 542–552, 2015. doi: 10.1111/ecc.12262
  • Hutton JL, Martin L, Field CJ, Wismer WV, Bruera ED, et al.: Dietary patterns in patients with advanced cancer: implications for anorexia-cachexia therapy. Am J Clin Nutr 84, 1163–1170, 2006.
  • Ryan AM, Power DG, Daly L, Cushen SJ, Bhuachalla EN, et al.: Cancer-associated malnutrition, cachexia and sarcopenia: the skeleton in the hospital closet 40 years later. Proc Nutr Soc 75, 199–211, 2016. doi: 10.1017/s002966511500419x
  • Stobaus N, Muller MJ, Kupferling S, Schulzke JD, and Norman K: Low recent protein intake predicts cancer-related fatigue and increased mortality in patients with advanced tumor disease undergoing chemotherapy. Nutr Cancer 67, 818–824, 2015. doi: 10.1080/01635581.2015.1040520
  • Gramignano G, Lusso MR, Madeddu C, Massa E, Serpe R, et al.: Efficacy of l-carnitine administration on fatigue, nutritional status, oxidative stress, and related quality of life in 12 advanced cancer patients undergoing anticancer therapy. Nutrition 22, 136–145, 2006. doi: 10.1016/j.nut.2005.06.003
  • Lima A, Oliveira J, Saude F, Mota J, and Ferreira RB: Proteins in soy might have a higher role in cancer prevention than previously expected: soybean protein fractions are more effective MMP-9 inhibitors than non-protein fractions, even in cooked seeds. Nutrients 9, 2017. doi: 10.3390/nu9111224
  • Kucuk O: Soy foods, isoflavones, and breast cancer. Cancer 123, 1901–1903, 2017. doi: 10.1002/cncr.30614
  • Lesinski GB, Reville PK, Mace TA, Young GS, Ahn-Jarvis J, et al.: Consumption of soy isoflavone enriched bread in men with prostate cancer is associated with reduced proinflammatory cytokines and immunosuppressive cells. Cancer Prev Res (Phila) 8, 1036–1044, 2015. doi: 10.1158/1940-6207.CAPR-14-0464
  • Ahn-Jarvis JH, Riedl KM, Schwartz SJ, and Vodovotz Y: Design and selection of soy breads used for evaluating isoflavone bioavailability in clinical trials. J Agric Food Chem 61, 3111–3120, 2013. doi: 10.1021/jf304699k
  • Hashimoto R, Sakai A, Murayama M, Ochi A, Abe T, et al.: Effects of dietary soy protein on skeletal muscle volume and strength in humans with various physical activities. J Med Invest 62, 177–183, 2015. doi: 10.2152/jmi.62.177
  • Witard OC, Wardle SL, Macnaughton LS, Hodgson AB, and Tipton KD: Protein considerations for optimising skeletal muscle mass in healthy young and older adults. Nutrients 8, 181, 2016. doi: 10.3390/nu8040181
  • Verreijen AM, Verlaan S, Engberink MF, Swinkels S, de Vogel-van den Bosch J, et al.: A high whey protein-, leucine-, and vitamin D-enriched supplement preserves muscle mass during intentional weight loss in obese older adults: a double-blind randomized controlled trial. Am J Clin Nutr 101, 279–286, 2015. doi: 10.3945/ajcn.114.090290
  • Devries MC and Phillips SM: Supplemental protein in support of muscle mass and health: advantage whey. J Food Sci 80 Suppl 1, A8–A15, 2015. doi: 10.1111/1750-3841.12802
  • Witard OC, Jackman SR, Breen L, Smith K, Selby A, et al.: Myofibrillar muscle protein synthesis rates subsequent to a meal in response to increasing doses of whey protein at rest and after resistance exercise. Am J Clin Nutr 99, 86–95, 2014. doi: 10.3945/ajcn.112.055517
  • Marx W, Teleni L, Opie RS, Kelly J, Marshall S, et al.: Efficacy and effectiveness of carnitine supplementation for cancer-related fatigue: a systematic literature review and meta-analysis. Nutrients 9, 2017. doi: 10.3390/nu9111224
  • Mantovani G, Maccio A, Madeddu C, Serpe R, Massa E, et al.: Randomized phase III clinical trial of five different arms of treatment in 332 patients with cancer cachexia. Oncologist 15, 200–211, 2010. doi: 10.1634/theoncologist.2009-0153
  • Winter SC, Szabo-Aczel S, Curry CJ, Hutchinson HT, Hogue R, et al.: Plasma carnitine deficiency. Clinical observations in 51 pediatric patients. Am J Dis Child 141, 660–665, 1987.
  • Endo K, Tsuji A, Kondo S, Wakisaka N, Murono S, et al.: Carnitine is associated with fatigue following chemoradiotherapy for head and neck cancer. Acta Otolaryngol 135, 846–852, 2015. doi: 10.3109/00016489.2015.1030769
  • Zick SM, Colacino J, Cornellier M, Khabir T, Surnow K, et al.: Fatigue reduction diet in breast cancer survivors: a pilot randomized clinical trial. Breast Cancer Res Treat 161, 299–310, 2017. doi: 10.1007/s10549-016-4070-y
  • Moura-Assis A, Afonso MS, de Oliveira V, Morari J, Dos Santos GA, et al.: Flaxseed oil rich in omega-3 protects aorta against inflammation and endoplasmic reticulum stress partially mediated by GPR120 receptor in obese, diabetic and dyslipidemic mice models. J Nutr Biochem 53, 9–19, 2017. doi: 10.1016/j.jnutbio.2017.09.015
  • Rodriguez-Cruz M, Cruz-Guzman ODR, Almeida-Becerril T, Solis-Serna AD, Atilano-Miguel S, et al.: Potential therapeutic impact of omega-3 long chain-polyunsaturated fatty acids on inflammation markers in Duchenne muscular dystrophy: a double-blind, controlled randomized trial. Clin Nutr, 2017. doi: 10.1016/j.clnu.2017.09.011
  • Alfano CM, Imayama I, Neuhouser ML, Kiecolt-Glaser JK, Smith AW, et al.: Fatigue, inflammation, and omega-3 and omega-6 fatty acid intake among breast cancer survivors. J Clin Oncol 30, 1280–1287, 2012. doi: 10.1200/jco.2011.36.4109
  • Navarro-Xavier RA, de Barros KV, de Andrade IS, Palomino Z, Casarini DE, et al.: Protective effect of soybean oil- or fish oil-rich diets on allergic airway inflammation. J Inflamm Res 9, 79–89, 2016. doi: 10.2147/jir.s102221
  • Guest DD, Evans EM, and Rogers LQ: Diet components associated with perceived fatigue in breast cancer survivors. Eur J Cancer Care (Engl) 22, 51–59, 2013. doi: 10.1111/j.1365-2354.2012.01368.x
  • Di Sebastiano KM and Mourtzakis M: The role of dietary fat throughout the prostate cancer trajectory. Nutrients 6, 6095–6109, 2014. doi: 10.3390/nu6126095
  • Lin S, Li Y, Zamyatnin Jr AA, Werner J, and Bazhin AV: Reactive oxygen species and colorectal cancer. J Cell Physiol 233, 5119–5132, 2017. doi: 10.1002/jcp.26356
  • Valdearcos M, Robblee MM, Benjamin DI, Nomura DK, Xu AW, et al.: Microglia dictate the impact of saturated fat consumption on hypothalamic inflammation and neuronal function. Cell Rep 9, 2124–2138, 2014. doi: 10.1016/j.celrep.2014.11.018
  • Gupta S, Knight AG, Gupta S, Keller JN, and Bruce-Keller AJ: Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J Neurochem 120, 1060–1071, 2012. doi: 10.1111/j.1471-4159.2012.07660.x
  • Yamada S, Kamada N, Amiya T, Nakamoto N, Nakaoka T, et al.: Gut microbiota-mediated generation of saturated fatty acids elicits inflammation in the liver in murine high-fat diet-induced steatohepatitis. BMC Gastroenterol 17, 136, 2017. doi: 10.1186/s12876-017-0689-3
  • Fritsche KL: The science of fatty acids and inflammation. Adv Nutr 6, 293s–301s, 2015. doi: 10.3945/an.114.006940
  • Zheng JS, Sharp SJ, Imamura F, Koulman A, Schulze MB, et al.: Association between plasma phospholipid saturated fatty acids and metabolic markers of lipid, hepatic, inflammation and glycaemic pathways in eight European countries: a cross-sectional analysis in the EPIC-InterAct study. BMC Med 15, 203, 2017. doi: 10.1186/s12916-017-0968-4
  • Gao Y, Bielohuby M, Fleming T, Grabner GF, Foppen E, et al.: Dietary sugars, not lipids, drive hypothalamic inflammation. Mol Metab 6, 897–908, 2017. doi: 10.1016/j.molmet.2017.06.008
  • Nilsson J, Ericsson M, Joibari MM, Anderson F, Carlsson L, et al.: A low-carbohydrate high-fat diet decreases lean mass and impairs cardiac function in pair-fed female C57BL/6J mice. Nutr Metab (Lond) 13, 79, 2016. doi: 10.1186/s12986-016-0132-8
  • Ruiz-Nunez B, Dijck-Brouwer DA, and Muskiet FA: The relation of saturated fatty acids with low-grade inflammation and cardiovascular disease. J Nutr Biochem 36, 1–20, 2016. doi: 10.1016/j.jnutbio.2015.12.007
  • Hernandez TL, Van Pelt RE, Anderson MA, Reece MS, Reynolds RM, et al.: Women with gestational diabetes mellitus randomized to a higher-complex carbohydrate/low-fat diet manifest lower adipose tissue insulin resistance, inflammation, glucose, and free fatty acids: a pilot study. Diabetes Care 39, 39–42, 2016. doi: 10.2337/dc15-0515
  • Alves BC, Silva TR, and Spritzer PM: Sedentary lifestyle and high-carbohydrate intake are associated with low-grade chronic inflammation in post-menopause: a cross-sectional study. Rev Bras Ginecol Obstet 38, 317–324, 2016. doi: 10.1055/s-0036-1584582
  • Hu T, Yao L, Reynolds K, Whelton PK, Niu T, et al.: The effects of a low-carbohydrate diet vs. a low-fat diet on novel cardiovascular risk factors: a randomized controlled trial. Nutrients 7, 7978–7994, 2015. doi: 10.3390/nu7095377
  • Okumatsu K, Tsujimoto T, Seki A, Yamauchi T, Yamauchi H, et al.: Effect of a combined diet plus exercise intervention on weight loss, physical fitness, and cancer-related fatigue among Japanese women with breast cancer. J Clin Oncol 35, e21658, 2017.
  • Kaluza J, Harris H, Wallin A, Linden A, and Wolk A: Dietary fiber intake and risk of chronic obstructive pulmonary disease: a prospective cohort study of men. Epidemiology 29, 254–260, 2018. doi: 10.1097/ede.0000000000000750
  • Ma Y, Griffith JA, Chasan-Taber L, Olendzki BC, and Jackson E, et al.: Association between dietary fiber and serum C-reactive protein. Am J Clin Nutr 83, 760–766, 2006.
  • Tonstad S, Malik N, and Haddad E: A high-fibre bean-rich diet versus a low-carbohydrate diet for obesity. J Hum Nutr Diet 27 Suppl 2, 109–116, 2014. doi: 10.1111/jhn.12118
  • Sylvetsky AC, Edelstein SL, Walford G, Boyko EJ, Horton ES, et al.: A high-carbohydrate, high-fiber, low-fat diet results in weight loss among adults at high risk of type 2 diabetes. J Nutr 147, 2060–2066, 2017. doi: 10.3945/jn.117.252395
  • Grober U, Holzhauer P, Kisters K, Holick MF, and Adamietz IA: Micronutrients in oncological intervention. Nutrients 8, 163, 2016. doi: 10.3390/nu8030163
  • Bilariki K, Anagnostou E, Masse V, Elie C, Grill J, et al.: Low bone mineral density and high incidences of fractures and vitamin D deficiency in 52 pediatric cancer survivors. Horm Res Paediatr 74, 319–327, 2010. doi: 10.1159/000313378
  • van Dijk M, Dijk FJ, Hartog A, van Norren K, Verlaan S, et al.: Reduced dietary intake of micronutrients with antioxidant properties negatively impacts muscle health in aged mice. J Cachexia Sarcopenia Muscle 9, 146–159, 2018. doi: 10.1002/jcsm.12237
  • Mantovani G, Maccio A, Madeddu C, Gramignano G, Lusso MR, et al.: A phase II study with antioxidants, both in the diet and supplemented, pharmaconutritional support, progestagen, and anti-cyclooxygenase-2 showing efficacy and safety in patients with cancer-related anorexia/cachexia and oxidative stress. Cancer Epidemiol Biomarkers Prev 15, 1030–1034, 2006. doi: 10.1158/1055-9965.epi-05-0538
  • Chapple IL, Griffiths HR, Milward MR, Ling MR, and Grant MM: Antioxidant micronutrients and oxidative stress biomarkers. Methods Mol Biol 1537, 61–77, 2017. doi: 10.1007/978-1-4939-6685-1_4
  • Mantovani G, Maccio A, Madeddu C, Gramignano G, Lusso MR, et al.: A phase II study with antioxidants, both in the diet and supplemented, pharmaconutritional support, progestagen, and anti-cyclooxygenase-2 showing efficacy and safety in patients with cancer-related anorexia/cachexia and oxidative stress. Cancer Epidemiol Biomarkers Prevent 15, 1030–1034, 2006. doi: 10.1158/1055-9965.Epi-05-0538
  • Mayland CR, Bennett MI, and Allan K: Vitamin C deficiency in cancer patients. Palliat Med 19, 17–20, 2005. doi: 10.1191/0269216305pm970oa
  • Dreizen S, McCredie KB, Keating MJ, and Andersson BS: Nutritional deficiencies in patients receiving cancer chemotherapy. Postgrad Med 87, 163–167, 170, 1990.
  • Petzel MQB and Hoffman L: Nutrition implications for long-term survivors of pancreatic cancer surgery [Formula: see text]. Nutr Clin Pract 32, 588–5z98, 2017. doi: 10.1177/0884533617722929
  • Silay K, Akinci S, Silay YS, Guney T, Ulas A, et al.: Hospitalization risk according to geriatric assessment and laboratory parameters in elderly hematologic cancer patients. Asian Pac J Cancer Prev 16, 783–786, 2015.
  • National Cancer Institute. Percent of New Cancers by Age Group: All Cancer Sites. Bethesda, MD, 2017.
  • Ludwig H, Muldur E, Endler G, and Hubl W: Prevalence of iron deficiency across different tumors and its association with poor performance status, disease status and anemia. Ann Oncol 24, 1886–1892, 2013. doi: 10.1093/annonc/mdt118
  • Jun JH, Yoo JE, Lee JA, Kim YS, Sunwoo S, et al.: Anemia after gastrectomy in long-term survivors of gastric cancer: a retrospective cohort study. Int J Surg 28, 162–168, 2016. doi: 10.1016/j.ijsu.2016.02.084
  • Vadhan-Raj S, Dahl NV, Bernard K, Li Z, and Strauss WE: Efficacy and safety of IV ferumoxytol for iron deficiency anemia in patients with cancer. J Blood Med 8, 199–209, 2017. doi: 10.2147/jbm.s138474
  • Kim A, Rivera S, Shprung D, Limbrick D, Gabayan V, et al.: Mouse models of anemia of cancer. PLoS One 9, e93283, 2014. doi: 10.1371/journal.pone.0093283
  • Bennett CL, Silver SM, Djulbegovic B, Samaras AT, Blau CA, et al.: Venous thromboembolism and mortality associated with recombinant erythropoietin and darbepoetin administration for the treatment of cancer-associated anemia. JAMA 299, 914–924, 2008. doi: 10.1001/jama.299.8.914
  • Vadhan-Raj S, Strauss W, Ford D, Bernard K, Boccia R, et al.: Efficacy and safety of IV ferumoxytol for adults with iron deficiency anemia previously unresponsive to or unable to tolerate oral iron. Am J Hematol 89, 7–12, 2014. doi: 10.1002/ajh.23582
  • Kim YT, Kim SW, Yoon BS, Cho HJ, Nahm EJ, et al.: Effect of intravenously administered iron sucrose on the prevention of anemia in the cervical cancer patients treated with concurrent chemoradiotherapy. Gynecol Oncol 105, 199–204, 2007. doi: 10.1016/j.ygyno.2006.11.014
  • Nagao T and Hirokawa M: Diagnosis and treatment of macrocytic anemias in adults. J Gen Fam Med 18, 200–204, 2017. doi: 10.1002/jgf2.31
  • Hu Y, Kim HI, Hyung WJ, Song KJ, Lee JH, et al.: Vitamin B(12) deficiency after gastrectomy for gastric cancer: an analysis of clinical patterns and risk factors. Ann Surg 258, 970–975, 2013. doi: 10.1097/sla.0000000000000214
  • Kim HI, Hyung WJ, Song KJ, Choi SH, Kim CB, et al.: Oral vitamin B12 replacement: an effective treatment for vitamin B12 deficiency after total gastrectomy in gastric cancer patients. Ann Surg Oncol 18, 3711–3717, 2011. doi: 10.1245/s10434-011-1764-6
  • Armstrong T, Walters E, Varshney S, and Johnson CD: Deficiencies of micronutrients, altered bowel function, and quality of life during late follow-up after pancreaticoduodenectomy for malignancy. Pancreatology 2, 528–534, 2002. doi: 10.1159/000066095
  • Peppone LJ, Rickles AS, Janelsins MC, Insalaco MR, and Skinner KA: The association between breast cancer prognostic indicators and serum 25-OH vitamin D levels. Ann Surg Oncol 19, 2590–2599, 2012. doi: 10.1245/s10434-012-2297-3
  • Alco G, Igdem S, Dincer M, Ozmen V, Saglam S, et al.: Vitamin D levels in patients with breast cancer: importance of dressing style. Asian Pac J Cancer Prev 15, 1357–1362, 2014.
  • Gröber U, Kisters K, and Adamietz IA: Vitamin D in oncology: update 2015. Medizinische Monatsschrift fur Pharmazeuten 38, 512–516, 2015.
  • Khan QJ, Kimler BF, Reddy PS, Sharma P, Klemp JR, et al.: 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 166, 491–500, 2017. doi: 10.1007/s10549-017-4429-8
  • Mager DR, Carroll MW, Wine E, Siminoski K, MacDonald K, et al.: Vitamin D status and risk for sarcopenia in youth with inflammatory bowel diseases. Eur J Clin Nutr 72, 623–626, 2018. doi: 10.1038/s41430-018-0105-2
  • Lee JH, Kim S, Kim MK, Yun BH, Cho S, et al.: Relationships between 25(OH)D concentration, sarcopenia and HOMA-IR in postmenopausal Korean women. Climacteric, 21, 40-46, 2017. doi: 10.1080/13697137.2017.1395410
  • Kaneko I, Miyamoto KI, and Nikawa T: [Update on recent progress in vitamin D research. Vitamin D and muscle tissues.]. Clin Calcium 27, 1571–1578, 2017.
  • Woo J: Nutritional interventions in sarcopenia: where do we stand? Curr Opin Clin Nutr Metab Care 21, 19–23, 2017. doi: 10.1097/mco.0000000000000432
  • Tukaj S, Bieber K, Witte M, Ghorbanalipoor S, Schmidt E, et al.: Calcitriol treatment ameliorates inflammation and blistering in mouse models of epidermolysis bullosa acquisita. J Invest Dermatol 138, 301–309, 2018. doi: 10.1016/j.jid.2017.09.009
  • Missel M, Hansen M, Jackson R, Siemsen M, and Schonau MN: Re-embodying eating after surgery for esophageal cancer: patients’ lived experiences of participating in an education and counseling nutritional intervention. J Clin Nurs 27, 1420–1430, 2018. doi: 10.1111/jocn.14297
  • Roemmler-Zehrer J, Geigenberger V, Stormann S, Losa M, Crippa V, et al.: Food intake regulating hormones in adult craniopharyngioma patients. Eur J Endocrinol 170, 627–635, 2014. doi: 10.1530/eje-13-0832
  • Ruskin DN, Kawamura M, and Masino SA: Reduced pain and inflammation in juvenile and adult rats fed a ketogenic diet. PLoS One 4, e8349, 2009. doi: 10.1371/journal.pone.0008349
  • Masino SA and Ruskin DN: Ketogenic diets and pain. J Child Neurol 28, 993–1001, 2013. doi: 10.1177/0883073813487595
  • Chung HY and Park YK: Rationale, feasibility and acceptability of ketogenic diet for cancer treatment. J Cancer Prev 22, 127–134, 2017. doi: 10.15430/jcp.2017.22.3.127
  • Kosinski C and Jornayvaz FR: Effects of ketogenic diets on cardiovascular risk factors: evidence from animal and human studies. Nutrients 9, 2017. doi: 10.3390/nu9050517
  • Haslam A, Robb SW, Hebert JR, Huang H, and Ebell MH: Greater adherence to a Mediterranean diet is associated with lower prevalence of colorectal adenomas in men of all races. Nutr Res 48, 76–84, 2017. doi: 10.1016/j.nutres.2017.10.003
  • Scoditti E, Calabriso N, Massaro M, Pellegrino M, Storelli C, et al.: Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: a potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch Biochem Biophys 527, 81–89, 2012. doi: 10.1016/j.abb.2012.05.003
  • Tosti V, Bertozzi B, and Fontana L: The Mediterranean diet: metabolic and molecular mechanisms. J Gerontol A Biol Sci Med Sci 73, 318-326, 2017. doi: 10.1093/gerona/glx227
  • Hoy MK and Goldman JD: What we eat in America, NHANES 2009-2010. Food Surveys Res Group Dietary Data Brief 12, 1-6, 2014.
  • Lerman RH: The macrobiotic diet in chronic disease. Nutr Clin Pract 25, 621–626, 2010. doi: 10.1177/0884533610385704
  • Kushi LH, Cunningham JE, Hebert JR, Lerman RH, Bandera EV, et al.: The macrobiotic diet in cancer. J Nutr 131, 3056S–3064S, 2001.
  • Harmon BE, Carter M, Hurley TG, Shivappa N, Teas J, et al.: Nutrient composition and anti-inflammatory potential of a prescribed macrobiotic diet. Nutr Cancer 67, 933–940, 2015. doi: 10.1080/01635581.2015.1055369
  • Swedish Council on Health Technology A: SBU systematic review summaries. In: Osteoporosis - Prevention, Diagnosis and Treatment: A Systematic Review. Swedish Council on Health Technology Assessment (SBU) Copyright (c) 2003 by the Swedish Council on Health Technology Assessment (ed.). Stockholm: SBU Yellow Report No. 165/1+2, 2003.
  • Dai Z and Koh WP: B-vitamins and bone health–a review of the current evidence. Nutrients 7, 3322–3346, 2015. doi: 10.3390/nu7053322
  • Maritess C, Small S, and Waltz-Hill M: Alternative nutrition therapies in cancer patients. Semin Oncol Nurs 21, 173–176, 2005. doi: 10.1016/j.soncn.2005.04.005
  • Whalen KA, McCullough ML, Flanders WD, Hartman TJ, Judd S, et al.: Paleolithic and mediterranean diet pattern scores are inversely associated with biomarkers of inflammation and oxidative balance in adults. J Nutr 146, 1217–1226, 2016. doi: 10.3945/jn.115.224048
  • Whalen KA, Judd S, McCullough ML, Flanders WD, Hartman TJ, et al.: Paleolithic and Mediterranean diet pattern scores are inversely associated with all-cause and cause-specific mortality in adults. J Nutr 147, 612–620, 2017. doi: 10.3945/jn.116.241919
  • Hoffman R: Can the Paleolithic diet meet the nutritional needs of older people? Maturitas 95, 63–64, 2017. doi: 10.1016/j.maturitas.2016.09.005
  • Tantamango-Bartley Y, Jaceldo-Siegl K, Fan J, and Fraser G: Vegetarian diets and the incidence of cancer in a low-risk population. Cancer Epidemiol Biomarkers Prev 22, 286–294, 2013. doi: 10.1158/1055-9965.epi-12-1060
  • Melina V, Craig W, and Levin S: Position of the Academy of Nutrition and Dietetics: vegetarian diets. J Acad Nutr Diet 116, 1970–1980, 2016. doi: 10.1016/j.jand.2016.09.025
  • Elorinne AL, Alfthan G, Erlund I, Kivimaki H, Paju A, et al.: Food and nutrient intake and nutritional status of Finnish vegans and non-vegetarians. PLoS One 11, e0148235, 2016. doi: 10.1371/journal.pone.0148235
  • Clarys P, Deliens T, Huybrechts I, Deriemaeker P, Vanaelst B, et al.: Comparison of nutritional quality of the vegan, vegetarian, semi-vegetarian, pesco-vegetarian and omnivorous diet. Nutrients 6, 1318–1332, 2014. doi: 10.3390/nu6031318
  • Davey GK, Spencer EA, Appleby PN, Allen NE, Knox KH, et al.: EPIC-Oxford: lifestyle characteristics and nutrient intakes in a cohort of 33 883 meat-eaters and 31 546 non meat-eaters in the UK. Public Health Nutr 6, 259–269, 2003. doi: 10.1079/phn2002430
  • Le LT and Sabate J: Beyond meatless, the health effects of vegan diets: findings from the Adventist cohorts. Nutrients 6, 2131–2147, 2014. doi: 10.3390/nu6062131
  • Bours MJ, Beijer S, Winkels RM, van Duijnhoven FJ, Mols F, et al.: Dietary changes and dietary supplement use, and underlying motives for these habits reported by colorectal cancer survivors of the Patient Reported Outcomes Following Initial Treatment and Long-Term Evaluation of Survivorship (PROFILES) registry. Br J Nutr 114, 286–296, 2015. doi: 10.1017/s0007114515001798
  • Public. IoMUCotUoCaAMbtA: Dietary supplements. In: Complementary and Alternative Medicine in the United States. Washington (DC): National Academies Press (US), 2005, pp. 1–3.
  • Majeed F, Malik FZ, Ahmed Z, Afreen A, Afzal MN, et al.: Ginseng phytochemicals as therapeutics in oncology: recent perspectives. Biomed Pharmacother 100, 52–63, 2018. doi: 10.1016/j.biopha.2018.01.155
  • Barton DL, Liu H, Dakhil SR, Linquist B, Sloan JA, et al.: Wisconsin Ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst 105, 1230–1238, 2013. doi: 10.1093/jnci/djt181
  • Viale PH: Can ginseng alleviate cancer-related fatigue? J Adv Pract Oncol 4, 392–393, 2013.
  • Park HJ, Shim HS, Kim JY, Kim JY, Park SK, et al.: Ginseng purified dry extract, BST204, improved cancer chemotherapy-related fatigue and toxicity in mice. Evid Based Complement Alternat Med 2015, 197459, 2015. doi: 10.1155/2015/197459
  • Yennurajalingam S, Tannir NM, Williams JL, Lu Z, Hess KR, et al.: A double-blind, randomized, placebo-controlled trial of Panax ginseng for cancer-related fatigue in patients with advanced cancer. J Natl Compr Canc Netw 15, 1111–1120, 2017. doi: 10.6004/jnccn.2017.0149
  • Barton DL, Soori GS, Bauer BA, Sloan JA, Johnson PA, et al.: Pilot study of Panax quinquefolius (American ginseng) to improve cancer-related fatigue: a randomized, double-blind, dose-finding evaluation: NCCTG trial N03CA. Support Care Cancer 18, 179–187, 2010. doi: 10.1007/s00520-009-0642-2
  • Lima NDS, Teixeira L, Gambero A, and Ribeiro ML: Guarana (Paullinia cupana) stimulates mitochondrial biogenesis in mice fed high-fat diet. Nutrients 10, 2018. doi: 10.3390/nu10020165
  • Lima NDS, Numata EP, Mesquita LMS, Dias PH, Vilegas W, et al.: Modulatory effects of guarana (Paullinia cupana) on adipogenesis. Nutrients 9, 2017. doi: 10.3390/nu9060635
  • Schimpl FC, da Silva JF, Goncalves JF, and Mazzafera P: Guarana: revisiting a highly caffeinated plant from the Amazon. J Ethnopharmacol 150, 14–31, 2013. doi: 10.1016/j.jep.2013.08.023
  • Moustakas D, Mezzio M, Rodriguez BR, Constable MA, Mulligan ME, et al.: Guarana provides additional stimulation over caffeine alone in the planarian model. PLoS One 10, e0123310, 2015. doi: 10.1371/journal.pone.0123310
  • de Oliveira Campos MP, Riechelmann R, Martins LC, Hassan BJ, Casa FB, et al.: Guarana (Paullinia cupana) improves fatigue in breast cancer patients undergoing systemic chemotherapy. J Altern Complement Med 17, 505–512, 2011. doi: 10.1089/acm.2010.0571
  • da Costa Miranda V, Trufelli DC, Santos J, Campos MP, Nobuo M, et al.: Effectiveness of guarana (Paullinia cupana) for postradiation fatigue and depression: results of a pilot double-blind randomized study. J Altern Complement Med 15, 431–433, 2009. doi: 10.1089/acm.2008.0324
  • Sette CVM, Ribas de Alcantara BB, Schoueri JHM, Cruz FM, Cubero DIG, et al.: Purified dry Paullinia cupana (PC-18) extract for chemotherapy-induced fatigue: results of two double-blind randomized clinical trials. J Diet Suppl, 15, 673–683, 2017. doi: 10.1080/19390211.2017.1384781
  • Basile A, Ferrara L, Pezzo MD, Mele G, Sorbo S, et al.: Antibacterial and antioxidant activities of ethanol extract from Paullinia cupana Mart. J Ethnopharmacol 102, 32–36, 2005. doi: 10.1016/j.jep.2005.05.038
  • Leite RP, Wada RS, Monteiro JC, Predes FS, and Dolder H: Protective effect of Guarana (Paullinia cupana var. sorbilis) pre-treatment on cadmium-induced damages in adult Wistar testis. Biol Trace Elem Res 141, 262–274, 2011. doi: 10.1007/s12011-010-8729-7
  • Yonekura L, Martins CA, Sampaio GR, Monteiro MP, Cesar LA, et al.: Bioavailability of catechins from guarana (Paullinia cupana) and its effect on antioxidant enzymes and other oxidative stress markers in healthy human subjects. Food Funct 7, 2970–2978, 2016. doi: 10.1039/c6fo00513f
  • Mattei R, Dias RF, Espinola EB, Carlini EA, and Barros SB: Guarana (Paullinia cupana): toxic behavioral effects in laboratory animals and antioxidants activity in vitro. J Ethnopharmacol 60, 111–116, 1998.
  • Alkhatib A, Seijo M, Larumbe E, and Naclerio F: Acute effectiveness of a “fat-loss” product on substrate utilization, perception of hunger, mood state and rate of perceived exertion at rest and during exercise. J Int Soc Sports Nutr 12, 44, 2015. doi: 10.1186/s12970-015-0105-8
  • Zeidan-Chulia F, Gelain DP, Kolling EA, Rybarczyk-Filho JL, Ambrosi P, et al.: Major components of energy drinks (caffeine, taurine, and guarana) exert cytotoxic effects on human neuronal SH-SY5Y cells by decreasing reactive oxygen species production. Oxid Med Cell Longev 2013, 791795, 2013. doi: 10.1155/2013/791795
  • Oliveira SS, Del Giglio AB, Lerner TG, Zanellato RM, Tiemi L, et al.: Paullinia cupana for control of hot flashes in breast cancer patients: a pilot study. Einstein (Sao Paulo) 11, 435–438, 2013.
  • Wu YS, Ho SY, Nan FH, and Chen SN: Ganoderma lucidum beta 1,3/1,6 glucan as an immunomodulator in inflammation induced by a high-cholesterol diet. BMC Complement Altern Med 16, 500, 2016. doi: 10.1186/s12906-016-1476-3
  • Pan K, Jiang Q, Liu G, Miao X, and Zhong D: Optimization extraction of Ganoderma lucidum polysaccharides and its immunity and antioxidant activities. Int J Biol Macromol 55, 301–316, 2013. doi: 10.1016/j.ijbiomac.2013.01.022
  • Chi B, Wang S, Bi S, Qin W, Wu D, et al.: Effects of ganoderic acid A on lipopolysaccharide-induced proinflammatory cytokine release from primary mouse microglia cultures. Exp Ther Med 15, 847–853, 2018. doi: 10.3892/etm.2017.5472
  • Wang J, Cao B, Zhao H, and Feng J: Emerging roles of Ganoderma lucidum in anti-aging. Aging Dis 8, 691–707, 2017. doi: 10.14336/ad.2017.0410
  • Sohretoglu D and Huang S: Ganoderma lucidum polysaccharides as an anti-cancer agent. Anticancer Agents Med Chem 18, 667–674, 2017. doi: 10.2174/1871520617666171113121246
  • Liu CM, Kao CL, Tseng YT, Lo YC, and Chen CY: Ginger phytochemicals inhibit cell growth and modulate drug resistance factors in docetaxel resistant prostate cancer cell. Molecules 22, 2017. doi: 10.3390/molecules22091477
  • Pashaei-Asl R, Pashaei-Asl F, Mostafa Gharabaghi P, Khodadadi K, Ebrahimi M, et al.: The inhibitory effect of ginger extract on ovarian cancer cell line; application of systems biology. Adv Pharm Bull 7, 241–249, 2017. doi: 10.15171/apb.2017.029
  • Funk JL, Frye JB, Oyarzo JN, Chen J, Zhang H, et al.: Anti-inflammatory effects of the essential oils of ginger (Zingiber officinale Roscoe) in experimental rheumatoid arthritis. PharmaNutrition 4, 123–131, 2016. doi: 10.1016/j.phanu.2016.02.004
  • Bolognesi G, Belcaro G, Feragalli B, Cornelli U, Cotellese R, et al.: Movardol® (N-acetylglucosamine, Boswellia serrata, ginger) supplementation in the management of knee osteoarthritis: preliminary results from a 6-month registry study. Eur Rev Med Pharmacol Sci 20, 5198–5204, 2016.
  • Abolaji AO, Ojo M, Afolabi TT, Arowoogun MD, Nwawolor D, et al.: Protective properties of 6-gingerol-rich fraction from Zingiber officinale (Ginger) on chlorpyrifos-induced oxidative damage and inflammation in the brain, ovary and uterus of rats. Chem Biol Interact 270, 15–23, 2017. doi: 10.1016/j.cbi.2017.03.017
  • Lee S, Jerng UM, Liu Y, Kang JW, Nam D, et al.: The effectiveness and safety of moxibustion for treating cancer-related fatigue: a systematic review and meta-analyses. Support Care Cancer 22, 1429–1440, 2014. doi: 10.1007/s00520-014-2161-z
  • Marx W, McCarthy AL, Ried K, McKavanagh D, Vitetta L, et al.: The effect of a standardized ginger extract on chemotherapy-induced nausea-related quality of life in patients undergoing moderately or highly emetogenic chemotherapy: a double blind, randomized, placebo controlled trial. Nutrients 9, 2017. doi: 10.3390/nu9080867
  • Marx W, McCarthy AL, Ried K, Vitetta L, McKavanagh D, et al.: Can ginger ameliorate chemotherapy-induced nausea? Protocol of a randomized double blind, placebo-controlled trial. BMC Complement Altern Med 14, 134, 2014. doi: 10.1186/1472-6882-14-134
  • Cai Z, Liu J, Bian H, and Cai J: Astragaloside IV ameliorates necrotizing enterocolitis by attenuating oxidative stress and suppressing inflammation via the vitamin D3-upregulated protein 1/NF-kappaB signaling pathway. Exp Ther Med 12, 2702–2708, 2016. doi: 10.3892/etm.2016.3629
  • Zhang WJ and Frei B: Astragaloside IV inhibits NF- kappa B activation and inflammatory gene expression in LPS-treated mice. Mediators Inflamm 2015, 274314, 2015. doi: 10.1155/2015/274314
  • Guo Z, Xu HY, Xu L, Wang SS, and Zhang XM: In vivo and in vitro immunomodulatory and anti-inflammatory effects of total flavonoids of Astragalus. Afr J Tradit Complement Altern Med 13, 60–73, 2016. doi: 10.21010/ajtcam.v13i4.10
  • Zhang G, Zhou SM, Zheng SJ, Liu FY, and Gao YQ: Astragalus on the anti-fatigue effect in hypoxic mice. Int J Clin Exp Med 8, 14030–14035, 2015.
  • Huang YF, Lu L, Zhu DJ, Wang M, Yin Y, et al.: Effects of Astragalus polysaccharides on dysfunction of mitochondrial dynamics induced by oxidative stress. Oxid Med Cell Longev 2016, 9573291, 2016. doi: 10.1155/2016/9573291
  • Liu CH, Tsai CH, Li TC, Yang YW, Huang WS, et al.: Effects of the traditional Chinese herb Astragalus membranaceus in patients with poststroke fatigue: a double-blind, randomized, controlled preliminary study. J Ethnopharmacol 194, 954–962, 2016. doi: 10.1016/j.jep.2016.10.058
  • Lin RD, Mao YW, Leu SJ, Huang CY, and Lee MH: The immuno-regulatory effects of Schisandra chinensis and its constituents on human monocytic leukemia cells. Molecules 16, 4836–4849, 2011. doi: 10.3390/molecules16064836
  • Kang S, Lee KP, Park SJ, Noh DY, Kim JM, et al.: Identification of a novel anti-inflammatory compound, alpha-cubebenoate from Schisandra chinensis. J Ethnopharmacol 153, 242–249, 2014. doi: 10.1016/j.jep.2014.02.027
  • Chi A, Zhang Y, Kang Y, and Shen Z: Metabolic mechanism of a polysaccharide from Schisandra chinensis to relieve chronic fatigue syndrome. Int J Biol Macromol 93, 322–332, 2016. doi: 10.1016/j.ijbiomac.2016.08.042
  • Kang YS, Han MH, Hong SH, Park C, Hwang HJ, et al.: Anti-inflammatory effects of Schisandra chinensis (Turcz.) baill fruit through the inactivation of nuclear factor-kappab and mitogen-activated protein kinases signaling pathways in lipopolysaccharide-stimulated murine macrophages. J Cancer Prev 19, 279–287, 2014. doi: 10.15430/jcp.2014.19.4.279
  • Yuan XX, Yang LP, Yang ZL, Xiao WL, Sun HD, et al.: Effect of nigranoic acid on Ca(2)(+) influx and its downstream signal mechanism in NGF-differentiated PC12 cells. J Ethnopharmacol 153, 725–731, 2014. doi: 10.1016/j.jep.2014.03.038
  • Alok S, Jain SK, Verma A, Kumar M, Mahor A, et al.: Herbal antioxidant in clinical practice: a review. Asian Pacific J Tropical Biomed 4, 78–84, 2014. doi: 10.1016/S2221-1691(14)60213-6
  • Zou S, Fang L, and Lee MH: Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol Rep (Oxf) 6, 1–12, 2018. doi: 10.1093/gastro/gox031
  • Inglis JE and Ilich JZ: The microbiome and osteosarcopenic obesity in older individuals in long-term care facilities. Curr Osteoporos Rep 13, 358–362, 2015. doi: 10.1007/s11914-015-0287-7
  • Marx TL and Mehta AE: Polycystic ovary syndrome: pathogenesis and treatment over the short and long term. Cleveland Clinic J Med 70, 31–45, 2003.
  • Lakhan SE, Kirchgessner A: Gut inflammation in chronic fatigue syndrome. Nutr Metab (Lond) 7, 79, 2010. doi: 10.1186/1743-7075-7-79
  • Fremont M, Coomans D, Massart S, and De Meirleir K: High-throughput 16S rRNA gene sequencing reveals alterations of intestinal microbiota in myalgic encephalomyelitis/chronic fatigue syndrome patients. Anaerobe 22, 50–56, 2013. doi: 10.1016/j.anaerobe.2013.06.002
  • Sheedy JR, Wettenhall RE, Scanlon D, Gooley PR, Lewis DP, et al.: Increased d-lactic acid intestinal bacteria in patients with chronic fatigue syndrome. In Vivo 23, 621–628, 2009.
  • Logan AC, Venket Rao A, and Irani D: Chronic fatigue syndrome: lactic acid bacteria may be of therapeutic value. Med Hypotheses 60, 915–923, 2003.
  • Dulal S, Keku TO: Gut microbiome and colorectal adenomas. Cancer J 20, 225–231, 2014. doi: 10.1097/ppo.0000000000000050
  • Groeger D, O’Mahony L, Murphy EF, Bourke JF, Dinan TG, et al.: Bifidobacterium infantis 35624 modulates host inflammatory processes beyond the gut. Gut Microbes 4, 325–339, 2013. doi: 10.4161/gmic.25487
  • Hamad A, Fragkos KC, and Forbes A: A systematic review and meta-analysis of probiotics for the management of radiation induced bowel disease. Clin Nutr 32, 353–360, 2013. doi: 10.1016/j.clnu.2013.02.004
  • Liu MM, Li ST, Shu Y, and Zhan HQ: Probiotics for prevention of radiation-induced diarrhea: a meta-analysis of randomized controlled trials. PLoS One 12, e0178870, 2017. doi: 10.1371/journal.pone.0178870
  • Hauer-Jensen M, Denham JW, and Andreyev HJ: Radiation enteropathy–pathogenesis, treatment and prevention. Nat Rev Gastroenterol Hepatol 11, 470–479, 2014. doi: 10.1038/nrgastro.2014.46
  • Packey CD and Ciorba MA: Microbial influences on the small intestinal response to radiation injury. Curr Opin Gastroenterol 26, 88–94, 2010. doi: 10.1097/MOG.0b013e3283361927
  • Delia P, Sansotta G, Donato V, Frosina P, Messina G, et al.: Use of probiotics for prevention of radiation-induced diarrhea. World J Gastroenterol 13, 912–915, 2007.
  • Mego M, Chovanec J, Vochyanova-Andrezalova I, Konkolovsky P, Mikulova M, et al.: Prevention of irinotecan induced diarrhea by probiotics: a randomized double blind, placebo controlled pilot study. Complement Ther Med 23, 356–362, 2015. doi: 10.1016/j.ctim.2015.03.008
  • Sonis ST: The pathobiology of mucositis. Nat Rev Cancer 4, 277–284, 2004. doi: 10.1038/nrc1318
  • van Vliet MJ, Harmsen HJ, de Bont ES, and Tissing WJ: The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog 6, e1000879, 2010. doi: 10.1371/journal.ppat.1000879
  • Lee JY, Chu SH, Jeon JY, Lee MK, Park JH, et al.: Effects of 12 weeks of probiotic supplementation on quality of life in colorectal cancer survivors: a double-blind, randomized, placebo-controlled trial. Dig Liver Dis 46, 1126–1132, 2014. doi: 10.1016/j.dld.2014.09.004
  • Kleckner IR, Dunne RF, Asare M, Cole C, Fleming F, et al.: Exercise for toxicity management in cancer – a narrative review. Oncol Hematol Rev 14, 28–37, 2018.
  • Mustian KM, Alfano CM, Heckler C, Kleckner AS, Kleckner IR, et al.: Comparison of pharmaceutical, psychological, and exercise treatments for cancer-related fatigue: a meta-analysis. JAMA Oncol 3, 961–968, 2017. doi: 10.1001/jamaoncol.2016.6914
  • Lin PJ, Peppone LJ, Janelsins MC, Mohile SG, Kamen CS, et al.: Yoga for the management of cancer treatment-related toxicities. Curr Oncol Rep 20, 5, 2018. doi: 10.1007/s11912-018-0657-2

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.