Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions

Abstract

Excess adiposity is a risk factor for several cancers of the gastrointestinal system, specifically oesophageal adenocarcinoma and colorectal, small intestine, pancreatic, liver, gallbladder and stomach cancers. With the increasing prevalence of obesity in nearly all regions of the world, this relationship could represent a growing source of cancers of the digestive system. Experimental and molecular epidemiological studies indicate important roles for alterations in insulin signalling, adipose tissue-derived inflammation and sex hormone pathways in mediating the association between adiposity and gastrointestinal cancer. The intestinal microbiome, gut hormones and non alcoholic fatty liver disease (NAFLD) also have possible roles. However, important gaps remain in our knowledge. For instance, our understanding of how adiposity throughout the life course is related to the risk of gastrointestinal cancer development and of how obesity influences gastrointestinal cancer prognosis and survival is limited. Nonetheless, the increasing use of state-of-the-art analytical methods (such as omics technologies, Mendelian randomization and MRI) in large-scale epidemiological studies offers exciting opportunities to advance our understanding of the complex relationship between adiposity and gastrointestinal cancers. Here, we examine the epidemiology of associations between obesity and gastrointestinal cancer, explore potential mechanisms underlying these relationships and highlight important unanswered research questions.

Key points

  • A large body of epidemiological evidence supports a causal relationship between excess adiposity and elevated risk of developing gastrointestinal cancers.

  • With the rising prevalence of obesity worldwide, this relationship could represent a growing source of cancers of the gastrointestinal system.

  • Experimental and molecular epidemiological studies indicate important roles for alterations in insulin signalling, adipose tissue-derived inflammation and sex hormone pathways in mediating the association between adiposity and gastrointestinal cancer.

  • Emerging evidence suggests that the gut microbiome, gut hormones, and non alcoholic fatty liver disease might also have important roles in mediating the obesity-gastrointestinal cancer relationship.

  • The use of state-of-the-art analytical methods (omics technologies, Mendelian randomization, MRI and molecular pathological epidemiology) in large-scale epidemiological studies will probably offer novel insights into the adiposity-gastrointestinal cancer relationship.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Worldwide estimated incidence of selected gastrointestinal cancers and obesity prevalence in adult men.

Adapted from ref.8, CC-BY-4.0.

Fig. 2: Worldwide estimated incidence of selected gastrointestinal cancers and obesity prevalence in adult women.

Adapted from ref.8, CC-BY-4.0.

Fig. 3: Major established mechanisms linking adiposity and gastrointestinal cancers.

Similar content being viewed by others

References

  1. WCRF-AICR. Diet, nutrition, physical activity and colorectal cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/CUP%20Colorectal%20Report_2017_Digital.pdf (2017).

  2. WCRF-AICR. Diet, nutrition, physical activity and liver cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/Liver-Cancer-2015-Report.pdf (2015).

  3. WCRF-AICR. Diet, nutrition, physical activity and oesophageal cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/Oesophageal-cancer-report.pdf (2016).

  4. WCRF-AICR. Food, nutrition, physical activity, and the prevention of pancreatic cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/Pancreatic-Cancer-2012-Report.pdf (2012).

  5. WCRF-AICR. Diet, nutrition, physical activity and stomach cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/Stomach-Cancer-2016-Report.pdf (2016).

  6. Lauby-Secretan, B. et al. Body fatness and cancer — viewpoint of the IARC Working Group. N. Engl. J. Med. 375, 794–798 (2016).

    Google Scholar 

  7. WCRF-AICR. Diet, nutrition, physical activity and gallbladder cancer. Continuous Update Project http://www.wcrf.org/sites/default/files/Gallbladder-Cancer-2015-Report.pdf (2015).

  8. NCD Risk Factor Collaboration. Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19.2 million participants. Lancet 387, 1377–1396 (2016).

    Google Scholar 

  9. Lin, K. J., Cheung, W. Y., Lai, J. Y.-C. & Giovannucci, E. L. The effect of estrogen versus combined estrogen-progestogen therapy on the risk of colorectal cancer. Int. J. Cancer 130, 419–430 (2012).

    CAS  PubMed  Google Scholar 

  10. Terry, P. D., Miller, A. B. & Rohan, T. E. Obesity and colorectal cancer risk in women. Gut 51, 191–194 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Pischon, T. et al. Body size and risk of colon and rectal cancer in the European Prospective Investigation Into Cancer and Nutrition (EPIC). J. Natl Cancer Inst. 98, 920–931 (2006).

    PubMed  Google Scholar 

  12. Adams, K. F. et al. Body mass and colorectal cancer risk in the NIH–AARP cohort. Am. J. Epidemiol. 166, 36–45 (2007).

    PubMed  Google Scholar 

  13. Keimling, M. et al. Comparison of associations of body mass index, abdominal adiposity, and risk of colorectal cancer in a large prospective cohort study. Cancer Epidemiol. Biomarkers Prev. 22, 1383–1394 (2013).

    PubMed  Google Scholar 

  14. Ortega, L. S. et al. A prospective investigation of body size, body fat composition and colorectal cancer risk in the UK Biobank. Sci. Rep. 7, 17807 (2017).

    PubMed  PubMed Central  Google Scholar 

  15. Davey Smith, G. & Hemani, G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum. Mol. Genet. 23, R89–R98 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Sheehan, N. A., Didelez, V., Burton, P. R. & Tobin, M. D. Mendelian randomisation and causal inference in observational epidemiology. PLOS Med. 5, e177 (2008).

    Google Scholar 

  17. Jarvis, D. et al. Mendelian randomisation analysis strongly implicates adiposity with risk of developing colorectal cancer. Br. J. Cancer 115, 266–272 (2016).

    PubMed  PubMed Central  Google Scholar 

  18. Campbell, P. T. et al. Case–control study of overweight, obesity, and colorectal cancer risk, overall and by tumor microsatellite instability status. J. Natl Cancer Inst. 102, 391–400 (2010).

    PubMed  PubMed Central  Google Scholar 

  19. Hughes, L. A. E. et al. Body size and risk for colorectal cancers showing BRAF mutations or microsatellite instability: a pooled analysis. Int. J. Epidemiol. 41, 1060–1072 (2012).

    PubMed  Google Scholar 

  20. Morikawa, T. et al. Prospective analysis of body mass index, physical activity, and colorectal cancer risk associated with β-catenin (CTNNB1) status. Cancer Res. 73, 1600–1610 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Kuchiba, A. et al. Body mass index and risk of colorectal cancer according to fatty acid synthase expression in the Nurses’ Health Study. J. Natl Cancer Inst. 104, 415–420 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, X. et al. Early life body fatness and risk of colorectal cancer in U. S. women and men — results from two large cohort studies. Cancer Epidemiol. Biomarkers Prev. 24, 690–697 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Arnold, M. et al. Duration of adulthood overweight, obesity, and cancer risk in the Women’s Health Initiative: a longitudinal study from the United States. PLOS Med. 13, e1002081 (2016).

    Google Scholar 

  24. Fedirko, V. et al. Pre-diagnostic anthropometry and survival after colorectal cancer diagnosis in Western European populations. Int. J. Cancer 135, 1949–1960 (2014).

    CAS  PubMed  Google Scholar 

  25. Campbell, P. T. et al. Impact of body mass index on survival after colorectal cancer diagnosis: The Cancer Prevention Study-II Nutrition Cohort. J. Clin. Oncol. 30, 42–52 (2012).

    PubMed  Google Scholar 

  26. Caan, B. J. et al. Explaining the obesity paradox: the association between body composition and colorectal cancer survival (C-SCANS Study). Cancer Epidemiol. Biomarkers Prev. 26, 1008–1015 (2017).

    PubMed  PubMed Central  Google Scholar 

  27. Cross, A. J., Hollenbeck, A. R. & Park, Y. A large prospective study of risk factors for adenocarcinomas and malignant carcinoid tumors of the small intestine. Cancer Causes Control 24, 1737–1746 (2013).

    PubMed  PubMed Central  Google Scholar 

  28. Lu, Y. et al. Comparison of abdominal adiposity and overall obesity in relation to risk of small intestinal cancer in a European Prospective Cohort. Cancer Causes Control 27, 919–927 (2016).

    PubMed  PubMed Central  Google Scholar 

  29. Aune, D. et al. Body mass index, abdominal fatness and pancreatic cancer risk: a systematic review and non-linear dose–response meta-analysis of prospective studies. Ann. Oncol. 23, 843–852 (2012).

    CAS  PubMed  Google Scholar 

  30. Carreras-Torres, R. et al. The role of obesity, type 2 diabetes, and metabolic factors in pancreatic cancer: a Mendelian randomization study. J. Natl Cancer Inst. 109, djx012 (2017).

    PubMed Central  Google Scholar 

  31. Nogueira, L., Stolzenberg-Solomon, R., Gamborg, M., Sørensen, T. I. & Baker, J. L. Childhood body mass index and risk of adult pancreatic cancer. Curr. Dev. Nutr. 1, e001362 (2017).

    Google Scholar 

  32. Stolzenberg-Solomon, R. Z., Schairer, C., Moore, S., Hollenbeck, A. & Silverman, D. T. Lifetime adiposity and risk of pancreatic cancer in the NIH-AARP Diet and Health Study cohort. Am. J. Clin. Nutr. 98, 1057–1065 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Pang, Y. et al. Young adulthood and adulthood adiposity in relation to incidence of pancreatic cancer: a prospective study of 0.5 million Chinese adults and a meta-analysis. J. Epidemiol. Commun. Health 71, 1059–1067 (2017).

    Google Scholar 

  34. Yuan, C. et al. Prediagnostic body mass index and pancreatic cancer survival. J. Clin. Oncol. 31, 4229–4234 (2013).

    PubMed  PubMed Central  Google Scholar 

  35. Shi, Y.-Q. et al. Effect of body mass index on overall survival of pancreatic cancer: a meta-analysis. Medicine 95, e3305 (2016).

    Google Scholar 

  36. Campbell, P. T. et al. Body mass index, waist circumference, diabetes, and risk of liver cancer for U. S. adults. Cancer Res. 76, 6076–6083 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Schlesinger, S. et al. Abdominal obesity, weight gain during adulthood and risk of liver and biliary tract cancer in a European cohort. Int. J. Cancer 132, 645–657 (2013).

    CAS  PubMed  Google Scholar 

  38. Marrero, J. A. et al. Alcohol, tobacco and obesity are synergistic risk factors for hepatocellular carcinoma. J. Hepatol. 42, 218–224 (2005).

    CAS  PubMed  Google Scholar 

  39. Venook, A. P., Papandreou, C., Furuse, J. & Ladrón de Guevara, L. The incidence and epidemiology of hepatocellular carcinoma: a global and regional perspective. Oncology 15, 5–13 (2010).

    Google Scholar 

  40. Yang, B. et al. Adiposity across the adult life course and incidence of primary liver cancer: the NIH-AARP cohort. Int. J. Cancer 141, 271–278 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Campbell, P. T. et al. Body Size Indicators and Risk of Gallbladder Cancer: Pooled Analysis of Individual-Level Data from 19 Prospective Cohort Studies. Cancer Epidemiol. Biomarkers amp; Prevention 26, 597–606 (2017).

    CAS  Google Scholar 

  42. Petrick, J. L. et al. Body weight trajectories and risk of oesophageal and gastric cardia adenocarcinomas: a pooled analysis of NIH-AARP and PLCO Studies. Br. J. Cancer 116, 951–959 (2017).

    PubMed  PubMed Central  Google Scholar 

  43. Steffen, A. et al. General and abdominal obesity and risk of esophageal and gastric adenocarcinoma in the European Prospective Investigation into Cancer and Nutrition. Int. J. Cancer 137, 646–657 (2015).

    CAS  PubMed  Google Scholar 

  44. Thrift, A. P. et al. Obesity and Risk of esophageal adenocarcinoma and Barrett’s esophagus: a mendelian randomization study. J. Natl Cancer Inst. 106, dju252 (2014).

    PubMed  PubMed Central  Google Scholar 

  45. Song, M. et al. Trajectory of body shape across the lifespan and cancer risk. Int. J. Cancer 138, 2383–2395 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Song, H. et al. Body mass index change during adulthood and risk of oesophageal squamous-cell carcinoma in a Japanese population: the Japan Public Health (JPHC)-based prospective study. Br. J. Cancer 117, 1715–1722 (2017).

    PubMed  PubMed Central  Google Scholar 

  47. Bezemer, I. D. et al. C-peptide, IGF-I, sex-steroid hormones and adiposity: a cross-sectional study in healthy women within the European Prospective Investigation into Cancer and Nutrition (EPIC). Cancer Causes Control 16, 561–572 (2005).

    PubMed  Google Scholar 

  48. Pollak, M. N., Schernhammer, E. S. & Hankinson, S. E. Insulin-like growth factors and neoplasia. Nat. Rev. Cancer 4, 505–518 (2004).

    CAS  PubMed  Google Scholar 

  49. Giorgino, F. et al. Overexpression of insulin receptors in fibroblast and ovary cells induces a ligand-mediated transformed phenotype. Mol. Endocrinol. 5, 452–459 (1991).

    CAS  PubMed  Google Scholar 

  50. Ish-Shalom, D. et al. Mitogenic properties of insulin and insulin analogues mediated by the insulin receptor. Diabetologia 40 (Suppl. 2), S25–S31 (1997).

    CAS  PubMed  Google Scholar 

  51. Tran, T. T. et al. Hyperinsulinemia, but not other factors associated with insulin resistance, acutely enhances colorectal epithelial proliferation in vivo. Endocrinology 147, 1830–1837 (2006).

    CAS  PubMed  Google Scholar 

  52. Chettouh, H. et al. Mitogenic Insulin receptor-A is overexpressed in human hepatocellular carcinoma due to EGFR-mediated dysregulation of RNA splicing factors. Cancer Res. 73, 3974–3986 (2013).

    CAS  PubMed  Google Scholar 

  53. Kiunga, G. A. et al. Elevated insulin receptor protein expression in experimentally induced colonic tumors. Cancer Lett. 211, 145–153 (2004).

    CAS  PubMed  Google Scholar 

  54. Santoro, M. A. et al. Reduced insulin-like growth factor I receptor and altered insulin receptor isoform mRNAs in normal mucosa predict colorectal adenoma risk. Cancer Epidemiol. Biomarkers Prev. 23, 2093–2100 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Spector, S. A. et al. Human insulin receptor and insulin signaling proteins in hepatic disease. J. Surg. Res. 83, 32–35 (1999).

    CAS  PubMed  Google Scholar 

  56. Belfiore, A. et al. Insulin receptor isoforms in physiology and disease: an updated view. Endocr. Rev. 38, 379–431 (2017).

    PubMed  PubMed Central  Google Scholar 

  57. Schoen, R. E. et al. Increased blood glucose and insulin, body size, and incident colorectal cancer. J. Natl Cancer Inst. 91, 1147–1154 (1999).

    CAS  PubMed  Google Scholar 

  58. Limburg, P. J. et al. Insulin, glucose, insulin resistance and incident colorectal cancer in male smokers. Clin. Gastroenterol. Hepatol. 4, 1514–1521 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Gunter, M. J. et al. Insulin, insulin-like growth factor-I, endogenous estradiol, and risk of colorectal cancer in postmenopausal women. Cancer Res. 68, 329–337 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Palmqvist, R. et al. Plasma insulin, IGF-binding proteins-1 and -2 and risk of colorectal cancer: a prospective study in Northern Sweden. Int. J. Cancer 107, 89–93 (2003).

    CAS  PubMed  Google Scholar 

  61. Saydah, S. H. et al. Association of markers of insulin and glucose control with subsequent colorectal cancer risk. Cancer Epidemiol. Biomarkers Prev. 12, 412–418 (2003).

    CAS  PubMed  Google Scholar 

  62. Kaaks, R. et al. Serum C-peptide, insulin-like growth factor (IGF)-I, IGF-binding proteins, and colorectal cancer risk in women. J. Natl Cancer Inst. 92, 1592–1600 (2000).

    CAS  PubMed  Google Scholar 

  63. Otani, T., Iwasaki, M., Sasazuki, S., Inoue, M. & Tsugane, S. Plasma C-peptide, insulin-like growth factor-I, insulin-like growth factor binding proteins and risk of colorectal cancer in a nested case-control study: the Japan public health center-based prospective study. Int. J. Cancer 120, 2007–2012 (2007).

    CAS  PubMed  Google Scholar 

  64. Jenab, M. et al. Serum C-peptide, IGFBP-1 and IGFBP-2 and risk of colon and rectal cancers in the European Prospective Investigation into Cancer and Nutrition. Int. J. Cancer 121, 368–376 (2007).

    CAS  PubMed  Google Scholar 

  65. Wei, E. K. et al. A prospective study of C-peptide, insulin-like growth factor-I, insulin-like growth factor binding protein-1, and the risk of colorectal cancer in women. Cancer Epidemiol. Biomarkers Prev. 14, 850–855 (2005).

    CAS  PubMed  Google Scholar 

  66. Chen, L. et al. Circulating C-peptide level is a predictive factor for colorectal neoplasia: evidence from the meta-analysis of prospective studies. Cancer Causes Control 24, 1837–1847 (2013).

    CAS  PubMed  Google Scholar 

  67. Murphy, N. et al. A nested case-control study of metabolically defined body size phenotypes and risk of colorectal cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC). PLOS Med. 13, e1001988 (2016).

    Google Scholar 

  68. Grote, V. A. et al. Diabetes mellitus, glycated haemoglobin and C-peptide levels in relation to pancreatic cancer risk: a study within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Diabetologia 54, 3037–3046 (2011).

    CAS  PubMed  Google Scholar 

  69. Wolpin, B. M. et al. Hyperglycemia, insulin resistance, impaired pancreatic β-cell function, and risk of pancreatic cancer. J. Natl Cancer Inst. 105, 1027–1035 (2013).

    PubMed  PubMed Central  Google Scholar 

  70. Aleksandrova, K. et al. Inflammatory and metabolic biomarkers and risk of liver and biliary tract cancer. Hepatology 60, 858–871 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Hidaka, A. et al. Plasma insulin, C-peptide and blood glucose and the risk of gastric cancer: the Japan Public Health Center-based prospective study. Int. J. Cancer 136, 1402–1410 (2015).

    CAS  PubMed  Google Scholar 

  72. Valentinis, B. & Baserga, R. IGF-I receptor signalling in transformation and differentiation. Mol. Pathol. 54, 133–137 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Rinaldi, S. et al. Serum levels of IGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies. Int. J. Cancer 126, 1702–1715 (2010).

    CAS  PubMed  Google Scholar 

  74. Douglas, J. B. et al. Serum IGF-I, IGF-II, IGFBP-3, and IGF-I/IGFBP-3 molar ratio and risk of pancreatic cancer in the prostate, lung, colorectal, and ovarian cancer screening trial. Cancer Epidemiol. Biomarkers Prev. 19, 2298–2306 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Rohrmann, S. et al. Concentrations of IGF-I and IGFBP-3 and pancreatic cancer risk in the European Prospective Investigation into Cancer and Nutrition. Br. J. Cancer 106, 1004–1010 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Lukanova, A. et al. Prediagnostic plasma testosterone, sex hormone-binding globulin, IGF-I and hepatocellular carcinoma: etiological factors or risk markers? Int. J. Cancer 134, 164–173 (2014).

    PubMed  Google Scholar 

  77. Endogenous Hormones and Breast Cancer Collaborative Group. Insulin-like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: pooled individual data analysis of 17 prospective studies. Lancet Oncol. 11, 530–542 (2010).

    Google Scholar 

  78. Tannenbaum, G., Guyda, H. & Posner, B. Insulin-like growth factors: a role in growth hormone negative feedback and body weight regulation via brain. Science 220, 77–79 (1983).

    CAS  PubMed  Google Scholar 

  79. Allen, N. E. et al. Lifestyle determinants of serum insulin-like growth-factor-I (IGF-I), C-peptide and hormone binding protein levels in British women. Cancer Causes Control 14, 65–74 (2003).

    PubMed  Google Scholar 

  80. Lee, K.-W. et al. Rapid apoptosis induction by IGFBP-3 involves an insulin-like growth factor-independent nucleomitochondrial translocation of RXRα/Nur77. J. Biol. Chem. 280, 16942–16948 (2005).

    CAS  PubMed  Google Scholar 

  81. Baxter, R. C. Insulin-like growth factor binding protein-3 (IGFBP-3): novel ligands mediate unexpected functions. J. Cell Commun. Signal. 7, 179–189 (2013).

    PubMed  PubMed Central  Google Scholar 

  82. Crusz, S. M. & Balkwill, F. R. Inflammation and cancer: advances and new agents. Nat. Rev. Clin. Oncol. 12, 584 (2015).

    CAS  PubMed  Google Scholar 

  83. Zhou, B. et al. C-Reactive protein, interleukin-6 and the risk of colorectal cancer: a meta-analysis. Cancer Causes Control 25, 1397–1405 (2014).

    PubMed  Google Scholar 

  84. Nimptsch, K. et al. Association of CRP genetic variants with blood concentrations of C-reactive protein and colorectal cancer risk. Int. J. Cancer 136, 1181–1192 (2015).

    CAS  PubMed  Google Scholar 

  85. Kant, P. & Hull, M. A. Excess body weight and obesity[mdash]the link with gastrointestinal and hepatobiliary cancer. Nat. Rev. Gastroenterol. Hepatol. 8, 224–238 (2011).

    CAS  PubMed  Google Scholar 

  86. Vaiopoulos, A. G., Marinou, K., Christodoulides, C. & Koutsilieris, M. The role of adiponectin in human vascular physiology. Int. J. Cardiol. 155, 188–193 (2012).

    PubMed  Google Scholar 

  87. Wei, E. K., Giovannucci, E., Fuchs, C. S., Willett, W. C. & Mantzoros, C. S. Low Plasma adiponectin levels and risk of colorectal cancer in men: a prospective study. J. Natl Cancer Inst. 97, 1688–1694 (2005).

    CAS  PubMed  Google Scholar 

  88. Aleksandrova, K. et al. Leptin and soluble leptin receptor in risk of colorectal cancer in the European Prospective Investigation into Cancer and Nutrition Cohort. Cancer Res. 72, 5328–5337 (2012).

    CAS  PubMed  Google Scholar 

  89. Stattin, P. et al. Obesity and colon cancer: does leptin provide a link? Int. J. Cancer 109, 149–152 (2004).

    CAS  PubMed  Google Scholar 

  90. Ho, G. Y. F. et al. Adipokines linking obesity with colorectal cancer risk in postmenopausal women. Cancer Res. 72, 3029–3037 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Bao, Y. et al. A prospective study of plasma adiponectin and pancreatic cancer risk in five US cohorts. J. Natl Cancer Inst. 105, 95–103 (2013).

    CAS  PubMed  Google Scholar 

  92. Bao, Y. et al. Inflammatory plasma markers and pancreatic cancer risk: a prospective study of five U. S. cohorts. Cancer Epidemiol. Biomarkers Prev. 22, 855–861 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Ohishi, W. et al. Serum interleukin-6 associated with hepatocellular carcinoma risk: a nested case–control study. Int. J. Cancer 134, 154–163 (2014).

    PubMed  Google Scholar 

  94. Endogenous Hormones and Breast Cancer Collaborative Group. Circulating sex hormones and breast cancer risk factors in postmenopausal women: reanalysis of 13 studies. Br. J. Cancer 105, 709–722 (2011).

    Google Scholar 

  95. Wallace, I. R., McKinley, M. C., Bell, P. M. & Hunter, S. J. Sex hormone binding globulin and insulin resistance. Clin. Endocrinol. 78, 321–329 (2013).

    CAS  Google Scholar 

  96. Hartman, J. et al. Tumor repressive functions of estrogen receptor β in SW480 colon cancer cells. Cancer Res. 69, 6100–6106 (2009).

    CAS  PubMed  Google Scholar 

  97. Konduri, S. & Schwarz, R. E. Estrogen receptor β/α ratio predicts response of pancreatic cancer cells to estrogens and phytoestrogens. J. Surg. Res. 140, 55–66 (2007).

    CAS  PubMed  Google Scholar 

  98. Waliszewski, P. et al. Molecular study of sex steroid receptor gene expression in human colon and in colorectal carcinomas. J. Surg. Oncol. 64, 3–11 (1997).

    CAS  PubMed  Google Scholar 

  99. Castiglione, F. et al. Expression of estrogen receptor beta in colon cancer progression. Diagn. Mol. Pathol. 17, 231–236 (2008).

    CAS  PubMed  Google Scholar 

  100. Murphy, N. et al. A prospective evaluation of endogenous sex hormone levels and colorectal cancer risk in postmenopausal women. J. Natl Cancer Inst. 107, djv210 (2015).

    PubMed  PubMed Central  Google Scholar 

  101. Clendenen, T. V. et al. Postmenopausal levels of endogenous sex hormones and risk of colorectal cancer. Cancer Epidemiol. Biomarkers Prev. 18, 275–281 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Lin, J. H. et al. Association between sex hormones and colorectal cancer risk in men and women. Clin. Gastroenterol. Hepatol. 11, 419–424 (2013).

    CAS  PubMed  Google Scholar 

  103. Hampel, H., Abraham, N. S. & El-Serag, H. B. Meta-analysis: obesity and the risk for gastroesophageal reflux disease and its complications. Ann. Internal Med. 143, 199–211 (2005).

    Google Scholar 

  104. Friedenberg, F. K., Xanthopoulos, M., Foster, G. D. & Richter, J. E. The association between gastroesophageal reflux disease and obesity. Am. J. Gastroenterol. 103, 2111 (2008).

    PubMed  Google Scholar 

  105. Lagergren, J., Bergström, R., Lindgren, A. & Nyrén, O. Symptomatic gastroesophageal reflux as a risk factor for esophageal adenocarcinoma. N. Engl. J. Med. 340, 825–831 (1999).

    CAS  PubMed  Google Scholar 

  106. Rubenstein, J. H. & Taylor, J. B. Meta-analysis: the association of oesophageal adenocarcinoma with symptoms of gastro-oesophageal reflux. Aliment. Pharmacol. Ther. 32, 1222–1227 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Derakhshan, M. H. et al. Combination of gastric atrophy, reflux symptoms and histological subtype indicates two distinct aetiologies of gastric cardia cancer. Gut 57, 298–305 (2008).

    CAS  PubMed  Google Scholar 

  108. Figueroa, J. D. et al. Cigarette smoking, body mass index, gastro-esophageal reflux disease, and non-steroidal anti-inflammatory drug use and risk of subtypes of esophageal and gastric cancers by P53 overexpression. Cancer Causes Control 20, 361–368 (2009).

    PubMed  Google Scholar 

  109. Karimi, P., Islami, F., Anandasabapathy, S., Freedman, N. D. & Kamangar, F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol. Biomarkers Prev. 23, 700–713 (2014).

    PubMed  PubMed Central  Google Scholar 

  110. Turnbaugh, P. J. et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 1027–1031 (2006).

    PubMed  Google Scholar 

  111. Turnbaugh, P. J. et al. A core gut microbiome in obese and lean twins. Nature 457, 480–484 (2009).

    CAS  PubMed  Google Scholar 

  112. Boulangé, C. L., Neves, A. L., Chilloux, J., Nicholson, J. K. & Dumas, M.-E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 8, 42 (2016).

    PubMed  PubMed Central  Google Scholar 

  113. Cani, P. D. et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 57, 1470–1481 (2008).

    CAS  PubMed  Google Scholar 

  114. Higgins, S. C., Gueorguiev, M. & Korbonits, M. Ghrelin, the peripheral hunger hormone. Ann. Med. 39, 116–136 (2007).

    CAS  PubMed  Google Scholar 

  115. Baatar, D., Patel, K. & Taub, D. D. The effects of ghrelin on inflammation and the immune system. Mol. Cell. Endocrinol. 340, 44–58 (2011).

    CAS  PubMed  Google Scholar 

  116. Shiiya, T. et al. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J. Clin. Endocrinol. Metab. 87, 240–244 (2002).

    CAS  PubMed  Google Scholar 

  117. Murphy, G. et al. The relationship between serum ghrelin and the risk of gastric and esophagogastric junctional adenocarcinomas. J. Natl Cancer Inst. 103, 1123–1129 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Murphy, G. et al. Serum ghrelin is associated with risk of colorectal adenocarcinomas in the ATBC study. Gut https://doi.org/10.1136/gutjnl-2016-313157 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Sundkvist, A., Myte, R., Palmqvist, R., Harlid, S. & Van Guelpen, B. Plasma ghrelin is probably not a useful biomarker for risk prediction or early detection of colorectal cancer. Gut https://doi.org/10.1136/gutjnl-2018-316110 (2018).

    Article  PubMed  Google Scholar 

  120. Ruhl, C. E. & Everhart, J. E. Determinants of the association of overweight with elevated serum alanine aminotransferase activity in the United States. Gastroenterology 124, 71–79 (2003).

    CAS  PubMed  Google Scholar 

  121. Fabbrini, E., Sullivan, S. & Klein, S. Obesity and Nonalcoholic fatty liver disease: biochemical, metabolic and clinical implications. Hepatology 51, 679–689 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Michelotti, G. A., Machado, M. V. & Diehl, A. M. NAFLD, NASH and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 10, 656 (2013).

    CAS  Google Scholar 

  123. Renehan, A. G., Zwahlen, M. & Egger, M. Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat. Rev. Cancer 15, 484–498 (2015).

    CAS  PubMed  Google Scholar 

  124. Kim, G.-A. et al. Association between non-alcoholic fatty liver disease and cancer incidence rate. J. Hepatol. 68, 140–146 (2018).

    Google Scholar 

  125. Wong, V. W.-S. et al. High prevalence of colorectal neoplasm in patients with non-alcoholic steatohepatitis. Gut 60, 829–836 (2011).

    PubMed  Google Scholar 

  126. Pearson-Stuttard, J. et al. Worldwide burden of cancer attributable to diabetes and high body mass index: a comparative risk assessment. Lancet Diabetes Endocrinol. 6, 95–104 (2017).

    PubMed  Google Scholar 

  127. Arnold, M. et al. Global patterns and trends in colorectal cancer incidence and mortality. Gut 66, https://doi.org/10.1136/gutjnl-2015-310912 (2016).

    PubMed  Google Scholar 

  128. Würtz, P. et al. Metabolic signatures of adiposity in young adults: Mendelian randomization analysis and effects of weight change. PLOS Med. 11, e1001765 (2014).

    Google Scholar 

  129. Stepien, M. et al. Alteration of amino acid and biogenic amine metabolism in hepatobiliary cancers: findings from a prospective cohort study. Int. J. Cancer 138, 348–360 (2016).

    CAS  PubMed  Google Scholar 

  130. Keum, N., Lee, D. H., Kim, R., Greenwood, D. C. & Giovannucci, E. L. Visceral adiposity and colorectal adenomas: dose-response meta-analysis of observational studies. Ann. Oncol. 26, 1101–1109 (2015).

    CAS  PubMed  Google Scholar 

  131. UK-Biobank. Protocol for a large-scale prospective epidemiological resource. UK Biobank http://www.ukbiobank.ac.uk/wp-content/uploads/2011/11/UK-Biobank-Protocol.pdf (2010).

  132. Allen, N. et al. UK Biobank: Current status and what it means for epidemiology. Health Policy Technol. 1, 123–126 (2012).

    Google Scholar 

  133. German National Cohort Consortium. The German National Cohort: aims, study design and organization. Eur. J. Epidemiol. 29, 371–382 (2014).

    Google Scholar 

  134. Ogino, S. et al. The role of molecular pathological epidemiology in the study of neoplastic and non-neoplastic diseases in the era of precision medicine. Epidemiology 27, 602–611 (2016).

    PubMed  PubMed Central  Google Scholar 

  135. Ogino, S., Chan, A. T., Fuchs, C. S. & Giovannucci, E. Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut 60, 397–411 (2011).

    PubMed  Google Scholar 

  136. Colussi, D., Brandi, G., Bazzoli, F. & Ricciardiello, L. Molecular Pathways involved in colorectal cancer: implications for disease behavior and prevention. Int. J. Mol. Sci. 14, 16365 (2013).

    PubMed  PubMed Central  Google Scholar 

  137. Nishihara, R. et al. Molecular pathological epidemiology gives clues to paradoxical findings. Eur. J. Epidemiol. 30, 1129–1135 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Holmes, M. V., Ala-Korpela, M. & Smith, G. D. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat. Rev. Cardiol. 14, 577 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Bowden, J., Davey Smith, G. & Burgess, S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int. J. Epidemiol. 44, 512–525 (2015).

    PubMed  PubMed Central  Google Scholar 

  140. Bowden, J., Davey Smith, G., Haycock, P. C. & Burgess, S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet. Epidemiol. 40, 304–314 (2016).

    PubMed  PubMed Central  Google Scholar 

  141. O’Doherty, M. G., Freedman, N. D., Hollenbeck, A. R., Schatzkin, A. & Abnet, C. C. A prospective cohort study of obesity and risk of oesophageal and gastric adenocarcinoma in the NIH–AARP Diet and Health Study. Gut 61, 1261–1268 (2012).

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

M.J.G. and N.M. researched data for the article. All authors made substantial contributions to discussion of content, wrote the article and reviewed and/or edited the manuscript before submission.

Corresponding author

Correspondence to Marc J. Gunter.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Murphy, N., Jenab, M. & Gunter, M.J. Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol 15, 659–670 (2018). https://doi.org/10.1038/s41575-018-0038-1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-018-0038-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer