Metabolic consequences of perioperative oral carbohydrates in breast cancer patients - an explorative study

Tone Hoel Lende, Marie Austdal, Tone Frost Bathen, Anne Elin Varhaugvik, Ivar Skaland, Einar Gudlaugsson, Nina G Egeland, Siri Lunde, Lars A Akslen, Kristin Jonsdottir, Emiel A M Janssen, Håvard Søiland, Jan P A Baak, Tone Hoel Lende, Marie Austdal, Tone Frost Bathen, Anne Elin Varhaugvik, Ivar Skaland, Einar Gudlaugsson, Nina G Egeland, Siri Lunde, Lars A Akslen, Kristin Jonsdottir, Emiel A M Janssen, Håvard Søiland, Jan P A Baak

Abstract

Background: The metabolic consequences of preoperative carbohydrate load in breast cancer patients are not known. The present explorative study investigated the systemic and tumor metabolic changes after preoperative per-oral carbohydrate load and their influence on tumor characteristics and survival.

Methods: The study setting was on university hospital level with primary and secondary care functions in south-west Norway. Serum and tumor tissue were sampled from a population-based cohort of 60 patients with operable breast cancer who were randomized to either per-oral carbohydrate load (preOp™; n = 25) or standard pre-operative fasting (n = 35) before surgery. Magnetic resonance (MR) metabolomics was performed on serum samples from all patients and high-resolution magic angle spinning (HR-MAS) MR analysis on 13 tumor samples available from the fasting group and 16 tumor samples from the carbohydrate group.

Results: Fourteen of 28 metabolites were differently expressed between fasting and carbohydrate groups. Partial least squares discriminant analysis showed a significant difference in the metabolic profile between the fasting and carbohydrate groups, compatible with the endocrine effects of insulin (i.e., increased serum-lactate and pyruvate and decreased ketone bodies and amino acids in the carbohydrate group). Among ER-positive tumors (n = 18), glutathione was significantly elevated in the carbohydrate group compared to the fasting group (p = 0.002), with a positive correlation between preoperative S-insulin levels and the glutathione content in tumors (r = 0.680; p = 0.002). In all tumors (n = 29), glutamate was increased in tumors with high proliferation (t-test; p = 0.009), independent of intervention group. Moreover, there was a positive correlation between tumor size and proliferation markers in the carbohydrate group only. Patients with ER-positive / T2 tumors and high tumor glutathione (≥1.09), high S-lactate (≥56.9), and high S-pyruvate (≥12.5) had inferior clinical outcomes regarding relapse-free survival, breast cancer-specific survival, and overall survival. Moreover, Integrated Pathway Analysis (IPA) in serum revealed activation of five major anabolic metabolic networks contributing to proliferation and growth.

Conclusions: Preoperative carbohydrate load increases systemic levels of lactate and pyruvate and tumor levels of glutathione and glutamate in ER-positive patients. These biological changes may contribute to the inferior clinical outcomes observed in luminal T2 breast cancer patients.

Trial of registration: ClinicalTrials.gov; NCT03886389. Retrospectively registered March 22, 2019.

Keywords: Breast cancer; Carbohydrate load; Clinical outcome; Fasting state; Insulin; Insulin c-peptide; Ketonic bodies; Proliferation; S-lactate; S-pyruvate; Tumor glutamate; Tumor glutathione.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
Flowchart of study participants
Fig. 2
Fig. 2
Partial Least Square Discriminant Analysis (PLS-DA) in serum. a Scores plot showing serum samples from the fasting group (green) and carbohydrate group (red). The carbohydrate and fasting groups have significantly different metabolic profiles as evidenced by permutation testing. b Variable Importance in Projection (VIP) scores showing the top 14 metabolites contributing to differences between the groups. The right column indicates increased (red) or decreased (green) metabolite in the indicated group
Fig. 3
Fig. 3
Correlation between serum metabolic profile and serum insulin, insulin C-peptide, and IGFBP3. Samples from carbohydrate-fed patients are shown in red, while samples from fasting patients are shown in blue. Metabolites are colored according to their variable importance in projection (VIP) score and labeled when VIP≥1. a Measured insulin vs. predicted insulin levels based on metabolic profile (cross-validated measurements). b Metabolites versus regression coefficient for insulin. Increased S-glucose, S-lactate, and decreased S-Leucine are important to prediction of serum insulin from the metabolic profile. c Measured insulin C peptide vs. predicted insulin C-peptide levels. d Regression weight plot showing metabolites versus the regression coefficient for insulin C-peptide. Increased S-Glucose, S-Lactate, and decreased S-Leucine are important to prediction of serum insulin C-peptide from the metabolic profile. e Measured Insulin Growth Factor Binding Protein 3 (IGFBP3) vs. predicted IGFBP3 based on metabolic profile. f Regression weight plot showing metabolites versus the regression coefficient for IGFBP3. Increased S-Acetone, S-Glycoproteins, and S-Leucine are important to prediction of serum IGFBP3 from the metabolic profile
Fig. 4
Fig. 4
a Principal Component Analysis (PCA) of tumor metabolites. No grouping of fasting vs carbohydrate groups observed. b Glutathione levels in ER positive tumors. c ROC curve for classification into carbohydrate or fasting group by glutathione concentration in ER-positive tumors. AUC= 0.894; 95%CI=0.0.687-1.000, P=0.002
Fig. 5
Fig. 5
Survival analyses for Tumor-Glutathione, Serum-lactate and Serum-pyruvate. a-c Relapse Free Survival (RFS); d-f Breast Cancer Survival (BCSS); g-i Overall Survival (OS)
Fig. 6
Fig. 6
Pathway analyses in serum metabolites. a Metabolite Set Enrichment Analysis of serum metabolism. Significantly enriched pathways are annotated in the pathway network. The circle size denotes significance of the pathway, and lines denote at least 25% shared metabolites in the pathways. b Ingenuity pathway analysis (IPA) bar chart showing the top 5 functions enriched in the dataset. c IPA pathway network showing the metabolites connected to four microRNAs found to be involved in tamoxifen resistance. Metabolites in green are downregulated in carbohydrate-fed patients, while metabolites in red are upregulated. MicroRNAs are colored purple. d IPA Function plot showing metabolites involved in organismal growth. Orange arrows indicate activation, while blue arrows indicate inhibition

References

    1. Lundqvist A, Andersson E, Ahlberg I, Nilbert M, Gerdtham U. Socioeconomic inequalities in breast cancer incidence and mortality in Europe-a systematic review and meta-analysis. Eur J Pub Health. 2016;26(5):804–813. doi: 10.1093/eurpub/ckw070.
    1. Yap Yoon-Sim, Lu Yen-Shen, Tamura Kenji, Lee Jeong Eon, Ko Eun Young, Park Yeon Hee, Cao A-Yong, Lin Ching-Hung, Toi Masakazu, Wu Jiong, Lee Soo-Chin. Insights Into Breast Cancer in the East vs the West. JAMA Oncology. 2019;5(10):1489. doi: 10.1001/jamaoncol.2019.0620.
    1. Sun YS, Zhao Z, Yang ZN, Xu F, Lu HJ, Zhu ZY, Shi W, Jiang J, Yao PP, Zhu HP. Risk factors and preventions of breast Cancer. Int J Biol Sci. 2017;13(11):1387–1397. doi: 10.7150/ijbs.21635.
    1. Baum M, Demicheli R, Hrushesky W, Retsky M. Does surgery unfavourably perturb the "natural history" of early breast cancer by accelerating the appearance of distant metastases? Eur J Cancer. 2005;41(4):508–515. doi: 10.1016/j.ejca.2004.09.031.
    1. Pukazhendhi G, Gluck S. Circulating tumor cells in breast cancer. J Carcinog. 2014;13:8. doi: 10.4103/1477-3163.135578.
    1. Price TT, Burness ML, Sivan A, Warner MJ, Cheng R, Lee CH, Olivere L, Comatas K, Magnani J, Kim Lyerly H, et al. Dormant breast cancer micrometastases reside in specific bone marrow niches that regulate their transit to and from bone. Sci Transl Med. 2016;8(340):340–373. doi: 10.1126/scitranslmed.aad4059.
    1. Wangchinda P, Ithimakin S. Factors that predict recurrence later than 5 years after initial treatment in operable breast cancer. World J Surg Oncol. 2016;14(1):223. doi: 10.1186/s12957-016-0988-0.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Zhao Hua, Shen Jie, Moore Steven C., Ye Yuanqing, Wu Xifeng, Esteva Francisco J., Tripathy Debasish, Chow Wong-Ho. Breast cancer risk in relation to plasma metabolites among Hispanic and African American women. Breast Cancer Research and Treatment. 2019;176(3):687–696. doi: 10.1007/s10549-019-05165-4.
    1. Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324(5930):1029–1033. doi: 10.1126/science.1160809.
    1. Warburg OPK, Negelein E. Ueber den stoffwechhsel der tumoren. Biochem Z. 1924;152(1):319–344.
    1. Fernandez-de-Cossio-Diaz J, Vazquez A. Limits of aerobic metabolism in cancer cells. Sci Rep. 2017;7(1):13488. doi: 10.1038/s41598-017-14071-y.
    1. Tran Q, Lee H, Park J, Kim SH, Park J. Targeting Cancer metabolism - revisiting the Warburg effects. Toxicol Res. 2016;32(3):177–193. doi: 10.5487/TR.2016.32.3.177.
    1. Hart CD, Tenori L, Luchinat C, Di Leo A. Metabolomics in breast Cancer: current status and perspectives. Adv Exp Med Biol. 2016;882:217–234. doi: 10.1007/978-3-319-22909-6_9.
    1. Ljungqvist O. ERAS--enhanced recovery after surgery: moving evidence-based perioperative care to practice. JPEN J Parenter Enteral Nutr. 2014;38(5):559–566. doi: 10.1177/0148607114523451.
    1. Lende TH, Austdal M, Varhaugvik AE, Skaland I, Gudlaugsson E, Kvaløy JT, Akslen LA, Søiland H, Janssen EAM, Baak JPA. Influence of pre-operative oral carbohydrate loading vs. standard fasting procedure on tumor proliferation and clinical outcome in breast cancer patients — a randomized trial. BMC Cancer. 2019;19(1):1076. doi: 10.1186/s12885-019-6275-z.
    1. Austdal M, Tangeras LH, Skrastad RB, Salvesen K, Austgulen R, Iversen AC, Bathen TF. First trimester urine and serum metabolomics for prediction of preeclampsia and gestational hypertension: a prospective screening study. Int J Mol Sci. 2015;16(9):21520–21538. doi: 10.3390/ijms160921520.
    1. Giskeodegard GF, Madssen TS, Euceda LR, Tessem MB, Moestue SA, Bathen TF. NMR-based metabolomics of biofluids in cancer. NMR Biomed. 2018:e3927. 10.1002/nbm.3927.
    1. Euceda LR, Hill DK, Stokke E, Hatem R, El Botty R, Bièche I, Marangoni E, Bathen TF, Moestue SA. Metabolic response to Everolimus in patient-derived triple-negative breast Cancer Xenografts. J Proteome Res. 2017;16(5):1868–1879. doi: 10.1021/acs.jproteome.6b00918.
    1. Eilers PH. Parametric time warping. Anal Chem. 2004;76(2):404–411. doi: 10.1021/ac034800e.
    1. Sitter B, Sonnewald U, Spraul M, Fjosne HE, Gribbestad IS. High-resolution magic angle spinning MRS of breast cancer tissue. NMR Biomed. 2002;15(5):327–337. doi: 10.1002/nbm.775.
    1. Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Ann Stat. 2001;29(4):1165–1188. doi: 10.1214/aos/1013699998.
    1. R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. .
    1. pls: Partial Least Squares and Principal Component Regression. R package version 2.7–1.
    1. Xia J., Mandal R., Sinelnikov I. V., Broadhurst D., Wishart D. S. MetaboAnalyst 2.0--a comprehensive server for metabolomic data analysis. Nucleic Acids Research. 2012;40(W1):W127–W133. doi: 10.1093/nar/gks374.
    1. Mehmood TLK, Snipen L, Sæbø S. A review of variable selection methods in partial least squares regression. Chemom Intell Lab Syst. 2012;118:62–69. doi: 10.1016/j.chemolab.2012.07.010.
    1. Egeland NG, Lunde S, Jonsdottir K, Lende TH, Cronin-Fenton D, Gilje B, Janssen EA, Søiland H. The role of MicroRNAs as predictors of response to Tamoxifen treatment in breast Cancer patients. Int J Mol Sci. 2015;16(10):24243–24275. doi: 10.3390/ijms161024243.
    1. Shaham O, Wei R, Wang TJ, Ricciardi C, Lewis GD, Vasan RS, Carr SA, Thadhani R, Gerszten RE, Mootha VK. Metabolic profiling of the human response to a glucose challenge reveals distinct axes of insulin sensitivity. Mol Syst Biol. 2008;4:214. doi: 10.1038/msb.2008.50.
    1. Hou Y, Zhou M, Xie J, Chao P, Feng Q, Wu J. High glucose levels promote the proliferation of breast cancer cells through GTPases. Breast Cancer (Dove Med Press) 2017;9:429–436.
    1. Tang FY, Pai MH, Chiang EP. Consumption of high-fat diet induces tumor progression and epithelial-mesenchymal transition of colorectal cancer in a mouse xenograft model. J Nutr Biochem. 2012;23(10):1302–1313. doi: 10.1016/j.jnutbio.2011.07.011.
    1. Passarella S. Schurr a: l-lactate transport and metabolism in mitochondria of Hep G2 cells-the Cori cycle revisited. Front Oncol. 2018;8:120. doi: 10.3389/fonc.2018.00120.
    1. Scott MJ, Fawcett WJ. Oral carbohydrate preload drink for major surgery - the first steps from famine to feast. Anaesthesia. 2014;69(12):1308–1313. doi: 10.1111/anae.12921.
    1. Atkins RPVK, Constantin-Teodosiu D, Lobo DN, Greenhaff PL. Rates of skeletal muscle mitochondrial ATP production are reduced during elective abdominal surgery in humans. J Am Coll Surg. 2011;2013:S59. doi: 10.1016/j.jamcollsurg.2011.06.130.
    1. Varadan KKAR, Dimitru CT, Blackshaw E, Perkins AC, Greenhaff PL, Lobo DN. Gastrointestinal surgery mediated increases in gut permeability and expression of IL6 and PDK4 mRNAs in quadriceps muscle may underpin the post-operative increase in whole-body insulin resistance in humans. J Am Coll Surg. 2011;2013:S53. doi: 10.1016/j.jamcollsurg.2011.06.114.
    1. Prando R, Cheli V, Buzzo P, Melga P, Ansaldi E, Accoto S. Blood lactate behavior after glucose load in diabetes mellitus. Acta Diabetol Lat. 1988;25(3):247–256. doi: 10.1007/BF02624820.
    1. Moore MC, Davis SN, Mann SL, Cherrington AD. Acute fructose administration improves oral glucose tolerance in adults with type 2 diabetes. Diabetes Care. 2001;24(11):1882–1887. doi: 10.2337/diacare.24.11.1882.
    1. Hui S, Ghergurovich JM, Morscher RJ, Jang C, Teng X, Lu W, Esparza LA, Reya T, Le Z, Yanxiang Guo J, et al. Glucose feeds the TCA cycle via circulating lactate. Nature. 2017;551(7678):115–118. doi: 10.1038/nature24057.
    1. Faubert B, Li KY, Cai L, Hensley CT, Kim J, Zacharias LG, Yang C, Do QN, Doucette S, Burguete D, et al. Lactate metabolism in human lung tumors. Cell. 2017;171(2):358–371. doi: 10.1016/j.cell.2017.09.019.
    1. Papavasiliou P, Fisher T, Kuhn J, Nemunaitis J, Lamont J. Circulating tumor cells in patients undergoing surgery for hepatic metastases from colorectal cancer. Proc (Baylor Univ Med Cent) 2010;23(1):11–14. doi: 10.1080/08998280.2010.11928572.
    1. Braun S, Vogl FD, Naume B, Janni W, Osborne MP, Coombes RC, Schlimok G, Diel IJ, Gerber B, Gebauer G, et al. A pooled analysis of bone marrow micrometastasis in breast cancer. N Engl J Med. 2005;353(8):793–802. doi: 10.1056/NEJMoa050434.
    1. de Boer M, van Dijck JA, Bult P, Borm GF, Tjan-Heijnen VC. Breast cancer prognosis and occult lymph node metastases, isolated tumor cells, and micrometastases. J Natl Cancer Inst. 2010;102(6):410–425. doi: 10.1093/jnci/djq008.
    1. Bidard FC, Hajage D, Bachelot T, Delaloge S, Brain E, Campone M, Cottu P, Beuzeboc P, Rolland E, Mathiot C, Pierga JY. Assessment of circulating tumor cells and serum markers for progression-free survival prediction in metastatic breast cancer: a prospective observational study. Breast Cancer Res. 2012;14(1):R29. doi: 10.1186/bcr3114.
    1. Liu D, Wang D, Wu C, Zhang L, Mei Q, Hu G, Long G, Sun W. Prognostic significance of serum lactate dehydrogenase in patients with breast cancer: a meta-analysis. Cancer Manag Res. 2019;11:3611–3619. doi: 10.2147/CMAR.S199260.
    1. Liu L, He Y, Ge G, Li L, Zhou P, Zhu Y, Tang H, Huang Y, Li W, Zhang L. Lactate dehydrogenase and creatine kinase as poor prognostic factors in lung cancer: a retrospective observational study. PLoS One. 2017;12(8):e0182168. doi: 10.1371/journal.pone.0182168.
    1. Choi JS, Yoon D, Koo JS, Kim S, Park VY, Kim EK, Kim S, Kim MJ. Magnetic resonance metabolic profiling of estrogen receptor-positive breast cancer: correlation with currently used molecular markers. Oncotarget. 2017;8(38):63405–63416. doi: 10.18632/oncotarget.18822.
    1. Dornier E, Rabas N, Mitchell L, Novo D, Dhayade S, Marco S, Mackay G, Sumpton D, Pallares M, Nixon C, et al. Glutaminolysis drives membrane trafficking to promote invasiveness of breast cancer cells. Nat Commun. 2017;8(1):2255. doi: 10.1038/s41467-017-02101-2.
    1. Knox WE, Horowitz ML, Friedell GH. The proportionality of glutaminase content to growth rate and morphology of rat neoplasms. Cancer Res. 1969;29(3):669–680.
    1. Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 2006;43(2):143–181. doi: 10.1080/10408360500523878.
    1. Franco R, Cidlowski JA. Apoptosis and glutathione: beyond an antioxidant. Cell Death Differ. 2009;16(10):1303–1314. doi: 10.1038/cdd.2009.107.
    1. Lien EC, Lyssiotis CA, Juvekar A, Hu H, Asara JM, Cantley LC, Toker A. Glutathione biosynthesis is a metabolic vulnerability in PI (3) K/Akt-driven breast cancer. Nat Cell Biol. 2016;18(5):572–578. doi: 10.1038/ncb3341.
    1. DeBerardinis RJ, Cheng T. Q's next: the diverse functions of glutamine in metabolism, cell biology and cancer. Oncogene. 2010;29(3):313–324. doi: 10.1038/onc.2009.358.
    1. Fack F, Espedal H, Keunen O, Golebiewska A, Obad N, Harter PN, Mittelbronn M, Bahr O, Weyerbrock A, Stuhr L, et al. Bevacizumab treatment induces metabolic adaptation toward anaerobic metabolism in glioblastomas. Acta Neuropathol. 2015;129(1):115–131. doi: 10.1007/s00401-014-1352-5.
    1. Liberti MV, Locasale JW. The Warburg effect: how does it benefit Cancer cells? Trends Biochem Sci. 2016;41(3):211–218. doi: 10.1016/j.tibs.2015.12.001.
    1. Lee SY, Ju MK, Jeon HM, Lee YJ, Kim CH, Park HG, Han SI, Kang HS. Oncogenic metabolism acts as a prerequisite step for induction of Cancer metastasis and Cancer stem cell phenotype. Oxidative Med Cell Longev. 2018;2018:1027453.
    1. Klauber-DeMore N, Van Zee KJ, Linkov I, Borgen PI, Gerald WL. Biological behavior of human breast cancer micrometastases. Clin Cancer Res. 2001;7(8):2434–2439.
    1. Engstrom MJ, Opdahl S, Hagen AI, Romundstad PR, Akslen LA, Haugen OA, Vatten LJ, Bofin AM. Molecular subtypes, histopathological grade and survival in a historic cohort of breast cancer patients. Breast Cancer Res Treat. 2013;140(3):463–473. doi: 10.1007/s10549-013-2647-2.
    1. Saunier E, Antonio S, Regazzetti A, Auzeil N, Laprevote O, Shay JW, Coumoul X, Barouki R, Benelli C, Huc L, Bortoli S. Resveratrol reverses the Warburg effect by targeting the pyruvate dehydrogenase complex in colon cancer cells. Sci Rep. 2017;7(1):6945. doi: 10.1038/s41598-017-07006-0.
    1. Blanquer-Rossello MD, Hernandez-Lopez R, Roca P, Oliver J, Valle A. Resveratrol induces mitochondrial respiration and apoptosis in SW620 colon cancer cells. Biochim Biophys Acta Gen Subj. 2017;1861(2):431–440. doi: 10.1016/j.bbagen.2016.10.009.
    1. Weber DD, Aminazdeh-Gohari S, Kofler B. Ketogenic diet in cancer therapy. Aging (Albany NY) 2018;10(2):164–165. doi: 10.18632/aging.101382.
    1. Tan-Shalaby J. Ketogenic diets and Cancer: emerging evidence. Fed Pract. 2017;34(Suppl 1):37S–42S.
    1. Khodabakhshi A, Akbari ME, Mirzaei HR, Mehrad-Majd H, Kalamian M, Davoodi SH. Feasibility, safety, and beneficial effects of MCT-based Ketogenic diet for breast Cancer treatment: a randomized controlled trial study. Nutr Cancer. 2019:1–8. [Epub ahead of print].
    1. Licha D, Vidali S, Aminzadeh-Gohari S, Alka O, Breitkreuz L, Kohlbacher O, Reischl RJ, Feichtinger RG, Kofler B, Huber CG. Untargeted metabolomics reveals molecular effects of Ketogenic diet on healthy and tumor Xenograft mouse models. Int J Mol Sci. 2019;20(16):E3873. doi: 10.3390/ijms20163873.
    1. Paoli A, Rubini A, Volek JS, Grimaldi KA. Beyond weight loss: a review of the therapeutic uses of very-low-carbohydrate (ketogenic) diets. Eur J Clin Nutr. 2013;67(8):789–796. doi: 10.1038/ejcn.2013.116.
    1. McTiernan A, Friedenreich CM, Katzmarzyk PT, Powell KE, Macko R, Buchner D, Pescatello LS, Bloodgood B, Tennant B, Vaux-Bjerke A, et al. Physical activity in Cancer prevention and survival: a systematic review. Med Sci Sports Exerc. 2019;51(6):1252–1261. doi: 10.1249/MSS.0000000000001937.
    1. Irwin ML, Smith AW, McTiernan A, Ballard-Barbash R, Cronin K, Gilliland FD, Baumgartner RN, Baumgartner KB, Bernstein L. Influence of pre- and postdiagnosis physical activity on mortality in breast cancer survivors: the health, eating, activity, and lifestyle study. J Clin Oncol. 2008;26(24):3958–3964. doi: 10.1200/JCO.2007.15.9822.
    1. Smith AJ, Phipps WR, Thomas W, Schmitz KH, Kurzer MS. The effects of aerobic exercise on estrogen metabolism in healthy premenopausal women. Cancer Epidemiol Biomark Prev. 2013;22(5):756–764. doi: 10.1158/1055-9965.EPI-12-1325.
    1. Yager JD, Davidson NE. Estrogen carcinogenesis in breast Cancer. N Engl J Med. 2006;354(3):270–282. doi: 10.1056/NEJMra050776.
    1. Donaldson MS. Nutrition and cancer: a review of the evidence for an anti-cancer diet. Nutr J. 2004;3:19. doi: 10.1186/1475-2891-3-19.
    1. Thomas F, Rome S, Mery F, Dawson E, Montagne J, Biro PA, Beckmann C, Renaud F, Poulin R, Raymond M, Ujvari B. Changes in diet associated with cancer: an evolutionary perspective. Evol Appl. 2017;10(7):651–657. doi: 10.1111/eva.12465.
    1. Chlebowski RT, Pettinger M, Stefanick ML, Howard BV, Mossavar-Rahmani Y, McTiernan A. Insulin, physical activity, and caloric intake in postmenopausal women: breast cancer implications. J Clin Oncol. 2004;22(22):4507–4513. doi: 10.1200/JCO.2004.04.119.
    1. Mattson MP, Longo VD, Harvie M. Impact of intermittent fasting on health and disease processes. Ageing Res Rev. 2017;39:46–58. doi: 10.1016/j.arr.2016.10.005.
    1. Harvie MN, Howell T. Could intermittent energy restriction and intermittent fasting reduce rates of Cancer in obese, overweight, and Normal-weight subjects? A summary of evidence. Adv Nutr. 2016;7(4):690–705. doi: 10.3945/an.115.011767.
    1. Longo VD, Panda S. Fasting, circadian rhythms, and time-restricted feeding in healthy lifespan. Cell Metab. 2016;23(6):1048–1059. doi: 10.1016/j.cmet.2016.06.001.
    1. Lee C, Raffaghello L, Brandhorst S, Safdie FM, Bianchi G, Martin-Montalvo A, Pistoia V, Wei M, Hwang S, Merlino A, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4(124):ra127. doi: 10.1126/scitranslmed.3003293.
    1. Poff AM, Ari C, Arnold P, Seyfried TN, D'Agostino DP. Ketone supplementation decreases tumor cell viability and prolongs survival of mice with metastatic cancer. Int J Cancer. 2014;135(7):1711–1720. doi: 10.1002/ijc.28809.
    1. Xu R, Rai A, Chen M, Suwakulsiri W, Greening DW, Simpson RJ. Extracellular vesicles in cancer - implications for future improvements in cancer care. Nat Rev Clin Oncol. 2018;15(10):617–638. doi: 10.1038/s41571-018-0036-9.
    1. Melo SA, Sugimoto H, O'Connell JT, Kato N, Villanueva A, Vidal A, Qiu L, Vitkin E, Perelman LT, Melo CA, et al. Cancer exosomes perform cell-independent microRNA biogenesis and promote tumorigenesis. Cancer Cell. 2014;26(5):707–721. doi: 10.1016/j.ccell.2014.09.005.
    1. Chatterjee Sayantani, Lee Ling Y., Kawahara Rebeca, Abrahams Jodie L., Adamczyk Barbara, Anugraham Merrina, Ashwood Christopher, Sumer‐Bayraktar Zeynep, Briggs Matthew T., Chik Jenny H. L., Everest‐Dass Arun, Förster Sarah, Hinneburg Hannes, Leite Katia R. M., Loke Ian, Möginger Uwe, Moh Edward S. X., Nakano Miyako, Recuero Saulo, Sethi Manveen K., Srougi Miguel, Stavenhagen Kathrin, Venkatakrishnan Vignesh, Wongtrakul‐Kish Katherine, Diestel Simone, Hoffmann Peter, Karlsson Niclas G., Kolarich Daniel, Molloy Mark P., Muders Michael H., Oehler Martin K., Packer Nicolle H., Palmisano Giuseppe, Thaysen‐Andersen Morten. Protein Paucimannosylation Is an Enriched N ‐Glycosylation Signature of Human Cancers. PROTEOMICS. 2019;19(21-22):1900010. doi: 10.1002/pmic.201900010.
    1. de-Freitas-Junior JCM, Andrade-da-Costa J, Silva MC, Pinho SS. Glycans as Regulatory Elements of the Insulin/IGF System: Impact in Cancer Progression. Int J Mol Sci. 2017;18(9):E1921. doi: 10.3390/ijms18091921.
    1. Lu J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. 2019;38(1–2):157–164. doi: 10.1007/s10555-019-09794-5.
    1. Dasgupta S, Rajapakshe K, Zhu B, Nikolai BC, Yi P, Putluri N, Choi JM, Jung SY, Coarfa C, Westbrook TF, et al. Metabolic enzyme PFKFB4 activates transcriptional coactivator SRC-3 to drive breast cancer. Nature. 2018;556(7700):249–254. doi: 10.1038/s41586-018-0018-1.

Source: PubMed

3
購読する