Skip to main content

Advertisement

Log in

Nutrition and neuroendocrine tumors: An update of the literature

  • Published:
Reviews in Endocrine and Metabolic Disorders Aims and scope Submit manuscript

Abstract

Neuroendocrine tumors (NETs) are a heterogeneous group of neoplasms with worldwide increasing incidence, high prevalence and survival. Both the tumor itself and the systemic therapy may have an impact on patients’ nutrition. Malnutrition negatively impacts on outcome in NETs patients. Moreover, it has been demonstrated that body mass index was a risk factor for NET development and that metabolic syndrome was associated with worse prognosis in these patients. Of note, food could also interact with the metabolism of oral target therapy and antineoplastic agents used for the treatment of progressive NETs. Therefore, the nutritional assessment, based on body composition, and lifestyle modifications should be an integral component of management of the NET patients. The nutrition care plans are an integral part of the multidisciplinary management team for patients with NETs. Nutritionists with expertise in NETs can provide dietary approaches to improve the quality of life and nutritional status during various therapeutic modalities used in patients with NETs. The aim of this review is to critically discuss the importance of nutrition and body composition in patients with NETs.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Dasari A, Shen C, Halperin D, Zhao B, Zhou S, Xu Y, et al. Trends in the incidence, prevalence, and survival outcomes in patients with neuroendocrine tumors in the United States. JAMA Oncol. 2017;3(10):1335–42. https://doi.org/10.1001/jamaoncol.2017.0589.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Boyar Cetinkaya R, Aagnes B, Thiis-Evensen E, Tretli S, Bergestuen DS, Hansen S. Trends in incidence of neuroendocrine neoplasms in Norway: a report of 16,075 cases from 1993 through 2010. Neuroendocrinology. 2017;104(1):1–10. https://doi.org/10.1159/000442207.

    Article  CAS  PubMed  Google Scholar 

  3. Scherubl H, Streller B, Stabenow R, Herbst H, Hopfner M, Schwertner C, et al. Clinically detected gastroenteropancreatic neuroendocrine tumors are on the rise: epidemiological changes in Germany. World J Gastroenterol. 2013;19(47):9012–9. https://doi.org/10.3748/wjg.v19.i47.9012.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Petersenn S, Koch CA. Neuroendocrine neoplasms - still a challenge despite major advances in clinical care with the development of specialized guidelines. Rev Endocr Metab Disord. 2017;18(4):373–8. https://doi.org/10.1007/s11154-018-9442-7.

    Article  PubMed  Google Scholar 

  5. Rindi G, Arnold R, Capella C, Klimstra DS, Klöppel G, Komminoth P, et al. Nomenclature and classification of digestive neuroendocrine tumours. In: WHO Classification of Tumours of the Digestive System. 4th ed. WHO Classification of Tumours, Volume 3. IARC; 2010. ISBN-13 9789283224327.

  6. Travis WD, Brambilla E, Burke AP, Marx A, Nicholson AG. WHO classification of tumours of the lung, pleura, thymus and heart. 4th ed. WHO Classification of Tumours, Volume 7. IARC; 2015. ISBN-139789283224365.

  7. Faggiano A, Ferolla P, Grimaldi F, Campana D, Manzoni M, Davi MV, et al. Natural history of gastro-entero-pancreatic and thoracic neuroendocrine tumors. Data from a large prospective and retrospective Italian epidemiological study: the NET management study. J Endocrinol Investig. 2012;35(9):817–23. https://doi.org/10.3275/8102.

    Article  CAS  Google Scholar 

  8. Anlauf M, Garbrecht N, Bauersfeld J, Schmitt A, Henopp T, Komminoth P, et al. Hereditary neuroendocrine tumors of the gastroenteropancreatic system. Virchows Arch. 2007;451(Suppl 1):S29–38. https://doi.org/10.1007/s00428-007-0450-3.

    Article  PubMed  Google Scholar 

  9. Krauss T, Ferrara AM, Links TP, Wellner U, Bancos I, Kvachenyuk A, et al. Preventive medicine of von Hippel-Lindau disease-associated pancreatic neuroendocrine tumors. Endocr Relat Cancer. 2018;25(9):783–93. https://doi.org/10.1530/ERC-18-0100.

    Article  PubMed  Google Scholar 

  10. Glasker S, Neumann HPH, Koch CA, Vortmeyer AO. Von Hippel-Lindau disease. In: De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM et al, editors. South Dartmouth, MA: Endotext; 2000.

  11. Falconi M, Eriksson B, Kaltsas G, Bartsch DK, Capdevila J, Caplin M, et al. ENETS consensus guidelines update for the management of patients with functional pancreatic neuroendocrine tumors and non-functional pancreatic neuroendocrine tumors. Neuroendocrinology. 2016;103(2):153–71. https://doi.org/10.1159/000443171.

    Article  CAS  PubMed  Google Scholar 

  12. Zandee WT, Kamp K, van Adrichem RC, Feelders RA, de Herder WW. Effect of hormone secretory syndromes on neuroendocrine tumor prognosis. Endocr Relat Cancer. 2017;24(7):R261–R74. https://doi.org/10.1530/ERC-16-0538.

    Article  CAS  PubMed  Google Scholar 

  13. Hallet J, Law CH, Cukier M, Saskin R, Liu N, Singh S. Exploring the rising incidence of neuroendocrine tumors: a population-based analysis of epidemiology, metastatic presentation, and outcomes. Cancer. 2015;121(4):589–97. https://doi.org/10.1002/cncr.29099.

    Article  PubMed  Google Scholar 

  14. Pavel M, O'Toole D, Costa F, Capdevila J, Gross D, Kianmanesh R, et al. ENETS consensus guidelines update for the management of distant metastatic disease of intestinal, pancreatic, bronchial neuroendocrine neoplasms (NEN) and NEN of unknown primary site. Neuroendocrinology. 2016;103(2):172–85. https://doi.org/10.1159/000443167.

    Article  CAS  PubMed  Google Scholar 

  15. Frost M, Lines KE, Thakker RV. Current and emerging therapies for PNETs in patients with or without MEN1. Nat Rev Endocrinol. 2018;14(4):216–27. https://doi.org/10.1038/nrendo.2018.3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Maasberg S, Knappe-Drzikova B, Vonderbeck D, Jann H, Weylandt KH, Grieser C, et al. Malnutrition predicts clinical outcome in patients with neuroendocrine neoplasia. Neuroendocrinology. 2017;104(1):11–25. https://doi.org/10.1159/000442983.

    Article  CAS  PubMed  Google Scholar 

  17. Lis CG, Gupta D, Lammersfeld CA, Markman M, Vashi PG. Role of nutritional status in predicting quality of life outcomes in cancer--a systematic review of the epidemiological literature. Nutr J. 2012;11:27. https://doi.org/10.1186/1475-2891-11-27.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Ekeblad S, Skogseid B, Dunder K, Oberg K, Eriksson B. Prognostic factors and survival in 324 patients with pancreatic endocrine tumor treated at a single institution. Clin Cancer Res. 2008;14(23):7798–803. https://doi.org/10.1158/1078-0432.CCR-08-0734.

    Article  CAS  PubMed  Google Scholar 

  19. Marrache F, Vullierme MP, Roy C, El Assoued Y, Couvelard A, O'Toole D, et al. Arterial phase enhancement and body mass index are predictors of response to chemoembolisation for liver metastases of endocrine tumours. Br J Cancer. 2007;96(1):49–55. https://doi.org/10.1038/sj.bjc.6603526.

    Article  CAS  PubMed  Google Scholar 

  20. Qureshi SA, Burch N, Druce M, Hattersley JG, Khan S, Gopalakrishnan K, et al. Screening for malnutrition in patients with gastro-entero-pancreatic neuroendocrine tumours: a cross-sectional study. BMJ Open. 2016;6(5):e010765. https://doi.org/10.1136/bmjopen-2015-010765.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Byers T, Sedjo RL. Body fatness as a cause of cancer: epidemiologic clues to biologic mechanisms. Endocr Relat Cancer. 2015;22(3):R125–34. https://doi.org/10.1530/ERC-14-0580.

    Article  CAS  PubMed  Google Scholar 

  22. Leoncini E, Carioli G, La Vecchia C, Boccia S, Rindi G. Risk factors for neuroendocrine neoplasms: a systematic review and meta-analysis. Ann Oncol. 2016;27(1):68–81. https://doi.org/10.1093/annonc/mdv505.

    Article  CAS  PubMed  Google Scholar 

  23. Gallo M, Muscogiuri G, Pizza G, Ruggeri RM, Barrea L, Faggiano A, et al. The management of neuroendocrine tumours: a nutritional viewpoint. Crit Rev Food Sci Nutr. 2017:1–12. https://doi.org/10.1080/10408398.2017.1390729.

  24. Glazer E, Stanko K, Ong E, Guerrero M. Decreased inpatient mortality in obese patients with abdominal Nets. Endocr Pract. 2014:1–20. https://doi.org/10.4158/EP14203.OR.

  25. Caan BJ, Cespedes Feliciano EM, Kroenke CH. The importance of body composition in explaining the overweight paradox in cancer-counterpoint. Cancer Res. 2018;78(8):1906–12. https://doi.org/10.1158/0008-5472.CAN-17-3287.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kushi LH, Byers T, Doyle C, Bandera EV, McCullough M, McTiernan A, et al. American Cancer Society guidelines on nutrition and physical activity for cancer prevention: reducing the risk of cancer with healthy food choices and physical activity. CA Cancer J Clin. 2006;56(5):254–81 quiz 313-4.

    Article  PubMed  Google Scholar 

  27. Longo VD, Fontana L. Calorie restriction and cancer prevention: metabolic and molecular mechanisms. Trends Pharmacol Sci. 2010;31(2):89–98. https://doi.org/10.1016/j.tips.2009.11.004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Jin XF, Spampatti MP, Spitzweg C, Auernhammer CJ. Supportive therapy in gastroenteropancreatic neuroendocrine tumors: often forgotten but important. Rev Endocr Metab Disord. 2018. https://doi.org/10.1007/s11154-018-9443-6.

  29. Kushi LH, Doyle C, McCullough M, Rock CL, Demark-Wahnefried W, Bandera EV, et al. American Cancer Society guidelines on nutrition and physical activity for cancer prevention: reducing the risk of cancer with healthy food choices and physical activity. CA Cancer J Clin. 2012;62(1):30–67. https://doi.org/10.3322/caac.20140.

    Article  PubMed  Google Scholar 

  30. Wang X, Ouyang Y, Liu J, Zhu M, Zhao G, Bao W, et al. Fruit and vegetable consumption and mortality from all causes, cardiovascular disease, and cancer: systematic review and dose-response meta-analysis of prospective cohort studies. BMJ. 2014;349:g4490. https://doi.org/10.1136/bmj.g4490.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Turati F, Rossi M, Pelucchi C, Levi F, La Vecchia C. Fruit and vegetables and cancer risk: a review of southern European studies. Br J Nutr. 2015;113(Suppl 2):S102–10. https://doi.org/10.1017/S0007114515000148.

    Article  CAS  PubMed  Google Scholar 

  32. Warner ME. Nutritional concerns for the carcinoid patient: developing nutrition guidelines for persons with carcinoid disease., https://www.carcinoid.org/for-patients/general-information/nutrition/nutritional-concerns-for-the-carcinoid-patient-developing-nutrition-guidelines-for-persons-with-carcinoid-disease/. 2008 update. Accessed 16 May 2018.

  33. Go VL, Srihari P, Kamerman Burns LA. Nutrition and gastroenteropancreatic neuroendocrine tumors. Endocrinol Metab Clin N Am. 2010;39(4):827–37. https://doi.org/10.1016/j.ecl.2010.08.003.

    Article  CAS  Google Scholar 

  34. Promotion. USDoHaHSOoDPaH. 2015–2020 Dietary Guidelines for Americans. https://health.gov/dietaryguidelines/?_ga=2.93506781.1509651194.1526503567-2137365160.1526503567. Accessed 16 May 2018.

  35. Blanchard CM, Courneya KS, Stein K. American Cancer Society’s SCS, II. Cancer survivors’ adherence to lifestyle behavior recommendations and associations with health-related quality of life: results from the American Cancer Society’s SCS-II. J Clin Oncol. 2008;26(13):2198–204. https://doi.org/10.1200/JCO.2007.14.6217.

    Article  PubMed  Google Scholar 

  36. Murphy RA, Mourtzakis M, Chu QS, Reiman T, Mazurak VC. Skeletal muscle depletion is associated with reduced plasma (n-3) fatty acids in non-small cell lung cancer patients. J Nutr. 2010;140(9):1602–6. https://doi.org/10.3945/jn.110.123521.

    Article  CAS  PubMed  Google Scholar 

  37. Hutton JL, Martin L, Field CJ, Wismer WV, Bruera ED, Watanabe SM, et al. Dietary patterns in patients with advanced cancer: implications for anorexia-cachexia therapy. Am J Clin Nutr. 2006;84(5):1163–70. https://doi.org/10.1093/ajcn/84.5.1163.

    Article  CAS  PubMed  Google Scholar 

  38. Prado CM, Lieffers JR, McCargar LJ, Reiman T, Sawyer MB, Martin L, 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. 2008;9(7):629–35. https://doi.org/10.1016/S1470-2045(08)70153-0.

    Article  PubMed  Google Scholar 

  39. Pavel M, Valle JW, Eriksson B, Rinke A, Caplin M, Chen J, et al. ENETS consensus guidelines for the standards of care in neuroendocrine neoplasms: systemic therapy - biotherapy and novel targeted agents. Neuroendocrinology. 2017;105(3):266–80. https://doi.org/10.1159/000471880.

    Article  CAS  PubMed  Google Scholar 

  40. Yao JC, Phan AT, Chang DZ, Wolff RA, Hess K, Gupta S, et al. Efficacy of RAD001 (everolimus) and octreotide LAR in advanced low- to intermediate-grade neuroendocrine tumors: results of a phase II study. J Clin Oncol. 2008;26(26):4311–8. https://doi.org/10.1200/JCO.2008.16.7858.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Yao JC, Phan A, Hoff PM, Chen HX, Charnsangavej C, Yeung SC, et al. Targeting vascular endothelial growth factor in advanced carcinoid tumor: a random assignment phase II study of depot octreotide with bevacizumab and pegylated interferon alpha-2b. J Clin Oncol. 2008;26(8):1316–23. https://doi.org/10.1200/JCO.2007.13.6374.

    Article  CAS  PubMed  Google Scholar 

  42. Vinik AI, Anthony L, Boudreaux JP, Go VL, O'Dorisio TM, Ruszniewski P, et al. Neuroendocrine tumors: a critical appraisal of management strategies. Pancreas. 2010;39(6):801–18. https://doi.org/10.1097/MPA.0b013e3181ea5839.

    Article  PubMed  Google Scholar 

  43. Plockinger U, Wiedenmann B. Diagnosis of non-functioning neuro-endocrine gastro-enteropancreatic tumours. Neuroendocrinology. 2004;80(Suppl 1):35–8. https://doi.org/10.1159/000080739.

    Article  CAS  PubMed  Google Scholar 

  44. Sofi F, Macchi C, Abbate R, Gensini GF, Casini A. Mediterranean diet and health status: an updated meta-analysis and a proposal for a literature-based adherence score. Public Health Nutr. 2014;17(12):2769–82. https://doi.org/10.1017/S1368980013003169.

    Article  PubMed  Google Scholar 

  45. Castello A, Amiano P, Fernandez de Larrea N, Martin V, Alonso MH, Castano-Vinyals G, et al. Low adherence to the western and high adherence to the mediterranean dietary patterns could prevent colorectal cancer. Eur J Nutr. 2018. https://doi.org/10.1007/s00394-018-1674-5.

  46. Castello A, Pollan M, Buijsse B, Ruiz A, Casas AM, Baena-Canada JM, et al. Spanish Mediterranean diet and other dietary patterns and breast cancer risk: case-control EpiGEICAM study. Br J Cancer. 2014;111(7):1454–62. https://doi.org/10.1038/bjc.2014.434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Romaguera D, Gracia-Lavedan E, Molinuevo A, de Batlle J, Mendez M, Moreno V, et al. Adherence to nutrition-based cancer prevention guidelines and breast, prostate and colorectal cancer risk in the MCC-Spain case-control study. Int J Cancer. 2017;141(1):83–93. https://doi.org/10.1002/ijc.30722.

    Article  CAS  PubMed  Google Scholar 

  48. Schwingshackl L, Hoffmann G. Adherence to Mediterranean diet and risk of cancer: a systematic review and meta-analysis of observational studies. Int J Cancer. 2014;135(8):1884–97. https://doi.org/10.1002/ijc.28824.

    Article  CAS  PubMed  Google Scholar 

  49. Rock CL, Doyle C, Demark-Wahnefried W, Meyerhardt J, Courneya KS, Schwartz AL, et al. Nutrition and physical activity guidelines for cancer survivors. CA Cancer J Clin. 2012;62(4):243–74. https://doi.org/10.3322/caac.21142.

    Article  PubMed  Google Scholar 

  50. Jones LW, Demark-Wahnefried W. Diet, exercise, and complementary therapies after primary treatment for cancer. Lancet Oncol. 2006;7(12):1017–26. https://doi.org/10.1016/S1470-2045(06)70976-7.

    Article  PubMed  Google Scholar 

  51. Agnoli C, Sieri S, Ricceri F, Giraudo MT, Masala G, Assedi M, et al. Adherence to a Mediterranean diet and long-term changes in weight and waist circumference in the EPIC-Italy cohort. Nutr Diabetes. 2018;8(1):22. https://doi.org/10.1038/s41387-018-0023-3.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Arnold M, Pandeya N, Byrnes G, Renehan PAG, Stevens GA, Ezzati PM, et al. Global burden of cancer attributable to high body-mass index in 2012: a population-based study. Lancet Oncol. 2015;16(1):36–46. https://doi.org/10.1016/S1470-2045(14)71123-4.

    Article  PubMed  Google Scholar 

  53. Freisling H, Arnold M, Soerjomataram I, O'Doherty MG, Ordonez-Mena JM, Bamia C, et al. Comparison of general obesity and measures of body fat distribution in older adults in relation to cancer risk: meta-analysis of individual participant data of seven prospective cohorts in Europe. Br J Cancer. 2017;116(11):1486–97. https://doi.org/10.1038/bjc.2017.106.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kyrgiou M, Kalliala I, Markozannes G, Gunter MJ, Paraskevaidis E, Gabra H, et al. Adiposity and cancer at major anatomical sites: umbrella review of the literature. BMJ. 2017;356:j477. https://doi.org/10.1136/bmj.j477.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Okorodudu DO, Jumean MF, Montori VM, Romero-Corral A, Somers VK, Erwin PJ, et al. Diagnostic performance of body mass index to identify obesity as defined by body adiposity: a systematic review and meta-analysis. Int J Obes. 2010;34(5):791–9. https://doi.org/10.1038/ijo.2010.5.

    Article  CAS  Google Scholar 

  56. Koch CA, Bornstein SR, Birkenfeld AL. Introduction to Hanefeld symposium: 40+ years of metabolic syndrome. Rev Endocr Metab Disord. 2016;17(1):1–4. https://doi.org/10.1007/s11154-016-9356-1.

    Article  PubMed  Google Scholar 

  57. Melcescu E, Griswold M, Xiang L, Belk S, Montgomery D, Bray M, et al. Prevalence and cardiometabolic associations of the glucocorticoid receptor gene polymorphisms N363S and BclI in obese and non-obese black and white Mississippians. Hormones (Athens). 2012;11(2):166–77.

    Article  Google Scholar 

  58. De Lorenzo A, Bianchi A, Maroni P, Iannarelli A, Di Daniele N, Iacopino L, et al. Adiposity rather than BMI determines metabolic risk. Int J Cardiol. 2013;166(1):111–7. https://doi.org/10.1016/j.ijcard.2011.10.006.

    Article  PubMed  Google Scholar 

  59. Norman K, Pichard C, Lochs H, Pirlich M. Prognostic impact of disease-related malnutrition. Clin Nutr. 2008;27(1):5–15. https://doi.org/10.1016/j.clnu.2007.10.007.

    Article  PubMed  Google Scholar 

  60. Stefanaki C, Peppa M, Boschiero D, Chrousos GP. Healthy overweight/obese youth: early osteosarcopenic obesity features. Eur J Clin Investig. 2016;46(9):767–78. https://doi.org/10.1111/eci.12659.

    Article  CAS  Google Scholar 

  61. Barrea L, Muscogiuri G, Macchia PE, Di Somma C, Falco A, Savanelli MC, et al. Mediterranean diet and phase angle in a sample of adult population: results of a pilot study. Nutrients. 2017;9(2). https://doi.org/10.3390/nu9020151.

    Article  PubMed Central  Google Scholar 

  62. Barbosa-Silva MC, Barros AJ, Wang J, Heymsfield SB, Pierson RN Jr. Bioelectrical impedance analysis: population reference values for phase angle by age and sex. Am J Clin Nutr. 2005;82(1):49–52. https://doi.org/10.1093/ajcn.82.1.49.

    Article  CAS  PubMed  Google Scholar 

  63. Macinnis RJ, English DR, Gertig DM, Hopper JL, Giles GG. Body size and composition and risk of postmenopausal breast cancer. Cancer Epidemiol Biomark Prev. 2004;13(12):2117–25.

    CAS  Google Scholar 

  64. MacInnis RJ, English DR, Hopper JL, Haydon AM, Gertig DM, Giles GG. Body size and composition and colon cancer risk in men. Cancer Epidemiol Biomark Prev. 2004;13(4):553–9.

    Google Scholar 

  65. Norman K, Stobaus N, Pirlich M, Bosy-Westphal A. Bioelectrical phase angle and impedance vector analysis--clinical relevance and applicability of impedance parameters. Clin Nutr. 2012;31(6):854–61. https://doi.org/10.1016/j.clnu.2012.05.008.

    Article  PubMed  Google Scholar 

  66. Paiva SI, Borges LR, Halpern-Silveira D, Assuncao MC, Barros AJ, Gonzalez MC. Standardized phase angle from bioelectrical impedance analysis as prognostic factor for survival in patients with cancer. Support Care Cancer. 2010;19(2):187–92. https://doi.org/10.1007/s00520-009-0798-9.

    Article  PubMed  Google Scholar 

  67. Norman K, Stobaus N, Zocher D, Bosy-Westphal A, Szramek A, Scheufele R, et al. Cutoff percentiles of bioelectrical phase angle predict functionality, quality of life, and mortality in patients with cancer. Am J Clin Nutr. 2010;92(3):612–9. https://doi.org/10.3945/ajcn.2010.29215.

    Article  CAS  PubMed  Google Scholar 

  68. Grundmann O, Yoon SL, Williams JJ. The value of bioelectrical impedance analysis and phase angle in the evaluation of malnutrition and quality of life in cancer patients--a comprehensive review. Eur J Clin Nutr. 2015;69(12):1290–7. https://doi.org/10.1038/ejcn.2015.126.

    Article  CAS  PubMed  Google Scholar 

  69. Ravasco P, Monteiro Grillo I, Camilo M. Cancer wasting and quality of life react to early individualized nutritional counselling! Clin Nutr. 2007;26(1):7–15. https://doi.org/10.1016/j.clnu.2006.10.005.

    Article  PubMed  Google Scholar 

  70. Segal EM, Flood MR, Mancini RS, Whiteman RT, Friedt GA, Kramer AR, et al. Oral chemotherapy food and drug interactions: a comprehensive review of the literature. J Oncol Pract. 2014;10(4):e255–68. https://doi.org/10.1200/JOP.2013.001183.

    Article  PubMed  Google Scholar 

  71. Deng J, Zhu X, Chen Z, Fan CH, Kwan HS, Wong CH, et al. A review of food-drug interactions on Oral drug absorption. Drugs. 2017;77(17):1833–55. https://doi.org/10.1007/s40265-017-0832-z.

    Article  CAS  PubMed  Google Scholar 

  72. Collado-Borrell R, Escudero-Vilaplana V, Romero-Jimenez R, Iglesias-Peinado I, Herranz-Alonso A, Sanjurjo-Saez M. Oral antineoplastic agent interactions with medicinal plants and food: an issue to take into account. J Cancer Res Clin Oncol. 2016;142(11):2319–30. https://doi.org/10.1007/s00432-016-2190-8.

    Article  CAS  PubMed  Google Scholar 

  73. Haefeli WE, Carls A. Drug interactions with phytotherapeutics in oncology. Expert Opin Drug Metab Toxicol. 2014;10(3):359–77. https://doi.org/10.1517/17425255.2014.873786.

    Article  CAS  PubMed  Google Scholar 

  74. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138(1):103–41. https://doi.org/10.1016/j.pharmthera.2012.12.007.

    Article  CAS  PubMed  Google Scholar 

  75. Kovarik JM, Hartmann S, Figueiredo J, Rordorf C, Golor G, Lison A, et al. Effect of food on everolimus absorption: quantification in healthy subjects and a confirmatory screening in patients with renal transplants. Pharmacotherapy. 2002;22(2):154–9.

    Article  CAS  PubMed  Google Scholar 

  76. Hammond LA, Eckardt JR, Baker SD, Eckhardt SG, Dugan M, Forral K, et al. Phase I and pharmacokinetic study of temozolomide on a daily-for-5-days schedule in patients with advanced solid malignancies. J Clin Oncol. 1999;17(8):2604–13. https://doi.org/10.1200/JCO.1999.17.8.2604.

    Article  CAS  PubMed  Google Scholar 

  77. Thomaschewski M, Neeff H, Keck T, Neumann HPH, Strate T, von Dobschuetz E. Is there any role for minimally invasive surgery in NET? Rev Endocr Metab Disord. 2017;18(4):443–57. https://doi.org/10.1007/s11154-017-9436-x.

    Article  CAS  PubMed  Google Scholar 

  78. Angelousi A, Kaltsas G, Koumarianou A, Weickert MO, Grossman A. Chemotherapy in NETs: when and how. Rev Endocr Metab Disord. 2017;18(4):485–97. https://doi.org/10.1007/s11154-017-9432-1.

    Article  PubMed  Google Scholar 

  79. Singer J, Werner F, Koch CA, Bartels M, Aigner T, Lincke T, et al. Ectopic cushing’s syndrome caused by a well differentiated ACTH-secreting neuroendocrine carcinoma of the ileum. Exp Clin Endocrinol Diabetes. 2010;118(8):524–9. https://doi.org/10.1055/s-0029-1243634.

    Article  CAS  PubMed  Google Scholar 

  80. Heber D, Greenway FL, Kaplan LM, Livingston E, Salvador J, Still C, et al. Endocrine and nutritional management of the post-bariatric surgery patient: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2010;95(11):4823–43. https://doi.org/10.1210/jc.2009-2128.

    Article  CAS  PubMed  Google Scholar 

  81. Bays H, Kothari SN, Azagury DE, Morton JM, Nguyen NT, Jones PH, et al. Lipids and bariatric procedures Part 2 of 2: scientific statement from the American Society for Metabolic and Bariatric Surgery (ASMBS), the National Lipid Association (NLA), and Obesity Medicine Association (OMA). Surg Obes Relat Dis. 2016;12(3):468–95. https://doi.org/10.1016/j.soard.2016.01.007.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Antongiulio Faggiano.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Statement of human rights and on the welfare of animals

This article does not contain any studies with human participants or animals performed by any of the authors.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Altieri, B., Barrea, L., Modica, R. et al. Nutrition and neuroendocrine tumors: An update of the literature. Rev Endocr Metab Disord 19, 159–167 (2018). https://doi.org/10.1007/s11154-018-9466-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11154-018-9466-z

Keywords

Navigation