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:

From NASH to HCC: current concepts and future challenges

Abstract

Caloric excess and sedentary lifestyle have led to a global epidemic of obesity and metabolic syndrome. The hepatic consequence of metabolic syndrome and obesity, nonalcoholic fatty liver disease (NAFLD), is estimated to affect up to one-third of the adult population in many developed and developing countries. This spectrum of liver disease ranges from simple steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. Owing to the high prevalence of NAFLD, especially in industrialized countries but also worldwide, and the consequent burden of progressive liver disease, there is mounting epidemiological evidence that NAFLD has rapidly become a leading aetiology underlying many cases of hepatocellular carcinoma (HCC). In this Review, we discuss NAFLD-associated HCC, including its epidemiology, the key features of the hepatic NAFLD microenvironment (for instance, adaptive and innate immune responses) that promote hepatocarcinogenesis and the management of HCC in patients with obesity and associated metabolic comorbidities. The challenges and future directions of research will also be discussed, including clinically relevant biomarkers for early detection, treatment stratification and monitoring as well as approaches to therapies for both prevention and treatment in those at risk or presenting with NAFLD-associated HCC.

Key points

  • Nonalcoholic fatty liver disease (NAFLD) is a spectrum of chronic liver disease that ranges from simple steatosis to nonalcoholic steatohepatitis (NASH) and is strongly associated with metabolic syndrome.

  • NAFLD dramatically increases the prevalence of hepatocellular carcinoma (HCC) development; however, the increased HCC risk of patients with NAFLD is often misdiagnosed.

  • The degree of fibrosis is considered the strongest predictive factor for correlating the progression of NAFLD with life-threating complications.

  • Several factors contribute to the development of NAFLD or NASH and subsequent HCC development; these factors include genetic and environmental modifiers such as diet or lifestyle.

  • The pathogenesis of NAFLD-associated HCC is a complex landscape composed of mechanisms involved in immune and inflammatory responses, DNA damage, oxidative stress and autophagy.

  • Currently, the diagnosis of NAFLD-associated HCC depends on imaging, whereas proper HCC staging is necessary for the evaluation of prognosis.

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: The sequential pathophysiological states of NAFLD and HCC.
Fig. 2: Environmental and gut-derived factors in NASH pathogenesis and the increased risk of liver tumorigenesis.
Fig. 3: Metabolic reprogramming and tumorigenesis induced by adaptive and innate chronic inflammation of the liver.
Fig. 4: Staging and treatment options for patients with HCC.
Fig. 5: Different treatment options for NASH-associated liver cancer development.

Similar content being viewed by others

References

  1. Swinburn, B. A. et al. The global obesity pandemic: shaped by global drivers and local environments. Lancet 378, 804–814 (2011).

    PubMed  Google Scholar 

  2. World Health Organization. Obesity and overweight. WHO https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (2017).

  3. Van Gaal, L. F., Mertens, I. L. & De Block, C. E. Mechanisms linking obesity with cardiovascular disease. Nature 444, 875–880 (2006).

    PubMed  Google Scholar 

  4. Adams, L. A., Anstee, Q. M., Tilg, H. & Targher, G. Non-alcoholic fatty liver disease and its relationship with cardiovascular disease and other extrahepatic diseases. Gut 66, 1138–1153 (2017).

    PubMed  Google Scholar 

  5. Younossi, Z. et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 15, 11–20 (2018).

    PubMed  Google Scholar 

  6. Bhaskaran, K. et al. Body-mass index and risk of 22 specific cancers: a population-based cohort study of 5.24 million UK adults. Lancet 384, 755–765 (2014).

    PubMed  PubMed Central  Google Scholar 

  7. Gupta, A. et al. Obesity is independently associated with increased risk of hepatocellular cancer-related mortality: a systematic review and meta-analysis. Am. J. Clin. Oncol. 41, 874–881 (2018).

    PubMed  PubMed Central  Google Scholar 

  8. Kanwal, F. et al. Risk of hepatocellular cancer in patients with non-alcoholic fatty liver disease. Gastroenterology 155, 1828–1837 (2018).

    PubMed  Google Scholar 

  9. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD) & European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 64, 1388–1402 (2016).

    Google Scholar 

  10. Burt, A. D., Lackner, C. & Tiniakos, D. G. Diagnosis and assessment of NAFLD: definitions and histopathological classification. Semin. Liver Dis. 35, 207–220 (2015).

    PubMed  Google Scholar 

  11. Chalasani, N. et al. The diagnosis and management of non-alcoholic fatty liver disease: practice Guideline by the American Association for the Study of Liver Diseases, American College of Gastroenterology, and the American Gastroenterological Association. Hepatology 55, 2005–2023 (2012).

    PubMed  Google Scholar 

  12. Younossi, Z. M. et al. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64, 73–84 (2016).

    PubMed  Google Scholar 

  13. Anstee, Q. M., Targher, G. & Day, C. P. Progression of NAFLD to diabetes mellitus, cardiovascular disease or cirrhosis. Nat. Rev. Gastroenterol. Hepatol. 10, 330–344 (2013).

    CAS  PubMed  Google Scholar 

  14. Dixon, J. B., Bhathal, P. S. & O’Brien, P. E. Nonalcoholic fatty liver disease: predictors of nonalcoholic steatohepatitis and liver fibrosis in the severely obese. Gastroenterology 121, 91–100 (2001).

    CAS  PubMed  Google Scholar 

  15. Estes, C., Razavi, H., Loomba, R., Younossi, Z. & Sanyal, A. J. Modeling the epidemic of nonalcoholic fatty liver disease demonstrates an exponential increase in burden of disease. Hepatology 67, 123–133 (2018).

    CAS  PubMed  Google Scholar 

  16. Estes, C. et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J. Hepatol. 69, 896–904 (2018).

    PubMed  Google Scholar 

  17. Hardy, T., Oakley, F., Anstee, Q. M. & Day, C. P. Nonalcoholic fatty liver disease: pathogenesis and disease spectrum. Annu. Rev. Pathol. 11, 451–496 (2016).

    CAS  PubMed  Google Scholar 

  18. Day, C. P. & James, O. F. Steatohepatitis: a tale of two “hits”? Gastroenterology 114, 842–845 (1998).

    CAS  PubMed  Google Scholar 

  19. Day, C. P. & Saksena, S. Non-alcoholic steatohepatitis: definitions and pathogenesis. J. Gastroenterol. Hepatol. 17 (Suppl. 3), S377–S384 (2002).

    PubMed  Google Scholar 

  20. Buzzetti, E., Pinzani, M. & Tsochatzis, E. A. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism 65, 1038–1048 (2016).

    CAS  PubMed  Google Scholar 

  21. Anstee, Q. M. & Day, C. P. The genetics of nonalcoholic fatty liver disease: spotlight on PNPLA3 and TM6SF2. Semin. Liver Dis. 35, 270–290 (2015).

    CAS  PubMed  Google Scholar 

  22. Utzschneider, K. M. & Kahn, S. E. Review: the role of insulin resistance in nonalcoholic fatty liver disease. J. Clin. Endocrinol. Metab. 91, 4753–4761 (2006).

    CAS  PubMed  Google Scholar 

  23. Krenkel, O. et al. Therapeutic inhibition of inflammatory monocyte recruitment reduces steatohepatitis and liver fibrosis. Hepatology 67, 1270–1283 (2018).

    CAS  PubMed  Google Scholar 

  24. Gadd, V. L. et al. The portal inflammatory infiltrate and ductular reaction in human nonalcoholic fatty liver disease. Hepatology 59, 1393–1405 (2014).

    PubMed  Google Scholar 

  25. Moylan, C. A. et al. Hepatic gene expression profiles differentiate presymptomatic patients with mild versus severe nonalcoholic fatty liver disease. Hepatology 59, 471–482 (2014).

    CAS  PubMed  Google Scholar 

  26. Boyle, M., Masson, S. & Anstee, Q. M. The bidirectional impacts of alcohol consumption and the metabolic syndrome: cofactors for progressive fatty liver disease. J. Hepatol. 68, 251–267 (2018).

    PubMed  Google Scholar 

  27. Hart, C. L., Morrison, D. S., Batty, G. D., Mitchell, R. J. & Smith, G. D. Effect of body mass index and alcohol consumption on liver disease: analysis of data from two prospective cohort studies. BMJ 340, c1240 (2010).

    PubMed  PubMed Central  Google Scholar 

  28. White, D. L., Kanwal, F. & El-Serag, H. B. Association between nonalcoholic fatty liver disease and risk for hepatocellular cancer, based on systematic review. Clin. Gastroenterol. Hepatol. 10, 1342–1359 (2012).

    PubMed  PubMed Central  Google Scholar 

  29. Wong, V. W. et al. Disease progression of non-alcoholic fatty liver disease: a prospective study with paired liver biopsies at 3 years. Gut 59, 969–974 (2010).

    PubMed  Google Scholar 

  30. McPherson, S. et al. Evidence of NAFLD progression from steatosis to fibrosing-steatohepatitis using paired biopsies: implications for prognosis and clinical management. J. Hepatol. 62, 1148–1155 (2015).

    PubMed  Google Scholar 

  31. Pais, R. et al. A systematic review of follow-up biopsies reveals disease progression in patients with non-alcoholic fatty liver. J. Hepatol. 59, 550–556 (2013).

    CAS  PubMed  Google Scholar 

  32. Singh, S. et al. Fibrosis progression in nonalcoholic fatty liver versus nonalcoholic steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Clin. Gastroenterol. Hepatol. 13, 643–654 (2015).

    PubMed  Google Scholar 

  33. Prashanth, M. et al. Prevalence of nonalcoholic fatty liver disease in patients with type 2 diabetes mellitus. J. Assoc. Physicians India 57, 205–210 (2009).

    CAS  PubMed  Google Scholar 

  34. Wolf, M. J. et al. Metabolic activation of intrahepatic CD8+T cells and NKT cells causes nonalcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014).

    CAS  PubMed  Google Scholar 

  35. Ringelhan, M., Pfister, D., O’Connor, T., Pikarsky, E. & Heikenwalder, M. The immunology of hepatocellular carcinoma. Nat. Immunol. 19, 222–232 (2018).

    CAS  PubMed  Google Scholar 

  36. Younossi, Z. M. et al. Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology 53, 1874–1882 (2011).

    PubMed  Google Scholar 

  37. Ekstedt, M. et al. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology 61, 1547–1554 (2015).

    CAS  PubMed  Google Scholar 

  38. Angulo, P. et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology 149, 389–397 (2015).

    PubMed  Google Scholar 

  39. Dulai, P. S. et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 65, 1557–1565 (2017).

    CAS  PubMed  Google Scholar 

  40. Vilar-Gomez, E. et al. Fibrosis severity as a determinant of cause-specific mortality in patients with advanced nonalcoholic fatty liver disease. Gastroenterology 155, 443–457 (2018).

    PubMed  Google Scholar 

  41. Jemal, A. et al. Global cancer statistics. CA Cancer J. Clin. 61, 69–90 (2011).

    PubMed  Google Scholar 

  42. Seyda Seydel, G. et al. Economic growth leads to increase of obesity and associated hepatocellular carcinoma in developing countries. Ann. Hepatol. 15, 662–672 (2016).

    CAS  PubMed  Google Scholar 

  43. Powell, E. E. et al. The natural history of nonalcoholic steatohepatitis: a follow-up study of forty-two patients for up to 21 years. Hepatology 11, 74–80 (1990).

    PubMed  Google Scholar 

  44. Ascha, M. S. et al. The incidence and risk factors of hepatocellular carcinoma in patients with nonalcoholic steatohepatitis. Hepatology 51, 1972–1978 (2010).

    PubMed  Google Scholar 

  45. Sanyal, A., Poklepovic, A., Moyneur, E. & Barghout, V. Population-based risk factors and resource utilization for HCC: US perspective. Curr. Med. Res. Opin. 26, 2183–2191 (2010).

    CAS  PubMed  Google Scholar 

  46. Baffy, G., Brunt, E. M. & Caldwell, S. H. Hepatocellular carcinoma in non-alcoholic fatty liver disease: an emerging menace. J. Hepatol. 56, 1384–1391 (2012).

    PubMed  Google Scholar 

  47. Wong, R. J., Cheung, R. & Ahmed, A. Nonalcoholic steatohepatitis is the most rapidly growing indication for liver transplantation in patients with hepatocellular carcinoma in the US. Hepatology 59, 2188–2195 (2014).

    PubMed  Google Scholar 

  48. Kim, G. A. et al. Association between non-alcoholic fatty liver disease and cancer incidence rate. J. Hepatol. 68, 140–146 (2017).

    Google Scholar 

  49. Kawamura, Y. et al. Large-scale long-term follow-up study of Japanese patients with non-alcoholic Fatty liver disease for the onset of hepatocellular carcinoma. Am. J. Gastroenterol. 107, 253–261 (2012).

    CAS  PubMed  Google Scholar 

  50. Adams, L. A. et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology 129, 113–121 (2005).

    PubMed  Google Scholar 

  51. Petrick, J. L. et al. International trends in liver cancer incidence, overall and by histologic subtype, 1978–2007. Int. J. Cancer 139, 1534–1545 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Bray, F. et al. Cancer Incidence in Five Continents Vol. XI (International Agency for Research on Cancer, 2017).

  53. Dyson, J. et al. Hepatocellular cancer: the impact of obesity, type 2 diabetes and a multidisciplinary team. J. Hepatol. 60, 110–117 (2014).

    PubMed  Google Scholar 

  54. Pais, R. et al. Temporal trends, clinical patterns and outcomes of NAFLD-related HCC in patients undergoing liver resection over a 20-year period. Aliment. Pharmacol. Ther. 46, 856–863 (2017).

    CAS  PubMed  Google Scholar 

  55. Piscaglia, F. et al. Clinical patterns of hepatocellular carcinoma in nonalcoholic fatty liver disease: a multicenter prospective study. Hepatology 63, 827–838 (2016).

    PubMed  Google Scholar 

  56. Ertle, J. et al. Non-alcoholic fatty liver disease progresses to hepatocellular carcinoma in the absence of apparent cirrhosis. Int. J. Cancer 128, 2436–2443 (2011).

    CAS  PubMed  Google Scholar 

  57. Yasui, K. et al. Characteristics of patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. Clin. Gastroenterol. Hepatol. 9, 428–433 (2011).

    PubMed  Google Scholar 

  58. Reeves, H. L., Zaki, M. Y. & Day, C. P. Hepatocellular carcinoma in obesity, type 2 diabetes, and NAFLD. Dig. Dis. Sci. 61, 1234–1245 (2016).

    CAS  PubMed  Google Scholar 

  59. Baffy, G. Hepatocellular carcinoma in obesity: finding a needle in the haystack? Adv. Exp. Med. Biol. 1061, 63–77 (2018).

    CAS  PubMed  Google Scholar 

  60. Mantovani, A. & Targher, G. Type 2 diabetes mellitus and risk of hepatocellular carcinoma: spotlight on nonalcoholic fatty liver disease. Ann. Transl Med. 5, 270 (2017).

    PubMed  PubMed Central  Google Scholar 

  61. Margini, C. & Dufour, J. F. The story of HCC in NAFLD: from epidemiology, across pathogenesis, to prevention and treatment. Liver Int. 36, 317–324 (2016).

    CAS  PubMed  Google Scholar 

  62. Teufel, A. et al. Comparison of gene expression patterns between mouse models of nonalcoholic fatty liver disease and liver tissues from patients. Gastroenterology 151, 513–525 (2016).

    CAS  PubMed  Google Scholar 

  63. Kaur, J. A comprehensive review on metabolic syndrome. Cardiol. Res. Pract. 2014, 943162 (2014).

    PubMed  PubMed Central  Google Scholar 

  64. Must, A. et al. The disease burden associated with overweight and obesity. JAMA 282, 1523–1529 (1999).

    CAS  PubMed  Google Scholar 

  65. Romeo, S. et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 40, 1461–1465 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Valenti, L. et al. Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease. Hepatology 51, 1209–1217 (2010).

    CAS  PubMed  Google Scholar 

  67. Kozlitina, J. et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nat. Genet. 46, 352–356 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Dongiovanni, P. et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 61, 506–514 (2015).

    CAS  PubMed  Google Scholar 

  69. Liu, Y. L. et al. TM6SF2 rs58542926 influences hepatic fibrosis progression in patients with non-alcoholic fatty liver disease. Nat. Commun. 5, 4309 (2014).

    CAS  PubMed  Google Scholar 

  70. Anstee, Q. M., Seth, D. & Day, C. P. Genetic factors that affect risk of alcoholic and nonalcoholic fatty liver disease. Gastroenterology 150, 1728–1744 (2016).

    PubMed  Google Scholar 

  71. BasuRay, S., Smagris, E., Cohen, J. C. & Hobbs, H. H. The PNPLA3 variant associated with fatty liver disease (I148M) accumulates on lipid droplets by evading ubiquitylation. Hepatology 66, 1111–1124 (2017).

    CAS  PubMed  Google Scholar 

  72. Ehrhardt, N. et al. Hepatic Tm6sf2 overexpression affects cellular ApoB-trafficking, plasma lipid levels, hepatic steatosis and atherosclerosis. Hum. Mol. Genet. 26, 2719–2731 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Liu, Y. L. et al. Carriage of the PNPLA3 rs738409 C>G polymorphism confers an increased risk of non-alcoholic fatty liver disease associated hepatocellular carcinoma. J. Hepatol. 61, 75–81 (2014).

    CAS  PubMed  Google Scholar 

  74. Singal, A. G. et al. The effect of PNPLA3 on fibrosis progression and development of hepatocellular carcinoma: a meta-analysis. Am. J. Gastroenterol. 109, 325–334 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Seko, Y. et al. Development of hepatocellular carcinoma in Japanese patients with biopsy-proven non-alcoholic fatty liver disease: association between PNPLA3 genotype and hepatocarcinogenesis/fibrosis progression. Hepatol. Res. 47, 1083–1092 (2017).

    CAS  PubMed  Google Scholar 

  76. Donati, B. et al. MBOAT7 rs641738 variant and hepatocellular carcinoma in non-cirrhotic individuals. Sci. Rep. 7, 4492 (2017).

    PubMed  PubMed Central  Google Scholar 

  77. Stickel, F. et al. Genetic variants in PNPLA3 and TM6SF2 predispose to the development of hepatocellular carcinoma in individuals with alcohol-related cirrhosis. Am. J. Gastroenterol. 113, 1475–1483 (2018).

    CAS  PubMed  Google Scholar 

  78. Zucman-Rossi, J., Villanueva, A., Nault, J. C. & Llovet, J. M. Genetic landscape and biomarkers of hepatocellular carcinoma. Gastroenterology 149, 1226–1239 (2015).

    CAS  PubMed  Google Scholar 

  79. Kechagias, S. et al. Fast-food-based hyper-alimentation can induce rapid and profound elevation of serum alanine aminotransferase in healthy subjects. Gut 57, 649–654 (2008).

    CAS  PubMed  Google Scholar 

  80. Leslie, T. et al. Survey of health status, nutrition and geography of food selection of chronic liver disease patients. Ann. Hepatol. 13, 533–540 (2014).

    PubMed  Google Scholar 

  81. Valtueña, S. et al. Dietary glycemic index and liver steatosis. Am. J. Clin. Nutr. 84, 136–142 (2006).

    PubMed  Google Scholar 

  82. Rietman, A., Sluik, D., Feskens, E. J. M., Kok, F. J. & Mensink, M. Associations between dietary factors and markers of NAFLD in a general Dutch adult population. Eur. J. Clin. Nutr. 72, 117–123 (2018).

    CAS  PubMed  Google Scholar 

  83. Jensen, T. et al. Fructose and sugar: a major mediator of non-alcoholic fatty liver disease. J. Hepatol. 68, 1063–1075 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Lanaspa, M. A. et al. High salt intake causes leptin resistance and obesity in mice by stimulating endogenous fructose production and metabolism. Proc. Natl Acad. Sci. USA 115, 3138–3143 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. McCarthy, E. M. & Rinella, M. E. The role of diet and nutrient composition in nonalcoholic fatty liver disease. J. Acad. Nutr. Diet. 112, 401–409 (2012).

    CAS  PubMed  Google Scholar 

  86. Gerber, L. et al. Non-alcoholic fatty liver disease (NAFLD) is associated with low level of physical activity: a population-based study. Aliment. Pharmacol. Ther. 36, 772–781 (2012).

    CAS  PubMed  Google Scholar 

  87. Hallsworth, K. et al. Non-alcoholic fatty liver disease is associated with higher levels of. Frontline Gastroenterol. 6, 44–51 (2015).

    PubMed  Google Scholar 

  88. Sunny, N. E., Bril, F. & Cusi, K. Mitochondrial adaptation in nonalcoholic fatty liver disease: novel mechanisms and treatment strategies. Trends Endocrinol. Metab. 28, 250–260 (2017).

    CAS  PubMed  Google Scholar 

  89. Nakagawa, H. et al. Lipid metabolic reprogramming in hepatocellular carcinoma. Cancers (Basel) 10, 447 (2018).

    Google Scholar 

  90. Nishida, N. et al. Unique features associated with hepatic oxidative DNA damage and DNA methylation in non-alcoholic fatty liver disease. J. Gastroenterol. Hepatol. 31, 1646–1653 (2016).

    CAS  PubMed  Google Scholar 

  91. Seki, S. et al. In situ detection of lipid peroxidation and oxidative DNA damage in non-alcoholic fatty liver diseases. J. Hepatol. 37, 56–62 (2002).

    CAS  PubMed  Google Scholar 

  92. Tanaka, S. et al. Increased hepatic oxidative DNA damage in patients with nonalcoholic steatohepatitis who develop hepatocellular carcinoma. J. Gastroenterol. 48, 1249–1258 (2013).

    CAS  PubMed  Google Scholar 

  93. Begriche, K., Massart, J., Robin, M. A., Bonnet, F. & Fromenty, B. Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology 58, 1497–1507 (2013).

    CAS  PubMed  Google Scholar 

  94. Masarone, M. et al. Role of oxidative stress in pathophysiology of nonalcoholic fatty liver disease. Oxid. Med. Cell. Longev. 2018, 9547613 (2018).

    PubMed  PubMed Central  Google Scholar 

  95. Perla, F. M., Prelati, M., Lavorato, M., Visicchio, D. & Anania, C. The role of lipid and lipoprotein metabolism in non-alcoholic fatty liver disease. Children (Basel) 4, (E46 (2017).

    Google Scholar 

  96. Wiseman, H. & Halliwell, B. Damage to DNA by reactive oxygen and nitrogen species: role in inflammatory disease and progression to cancer. Biochem. J. 313, 17–29 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Kuper, H., Adami, H. O. & Trichopoulos, D. Infections as a major preventable cause of human cancer. J. Intern. Med. 248, 171–183 (2000).

    CAS  PubMed  Google Scholar 

  98. Ohnishi, S. et al. DNA damage in inflammation-related carcinogenesis and cancer stem cells. Oxid. Med. Cell. Longev. 2013, 387014 (2013).

    PubMed  PubMed Central  Google Scholar 

  99. Wilson, C. L. et al. NFkappaB1 is a suppressor of neutrophil-driven hepatocellular carcinoma. Nat. Commun. 6, 6818 (2015).

    CAS  PubMed  Google Scholar 

  100. Yuan, D. et al. Kupffer cell-derived Tnf triggers cholangiocellular tumorigenesis through JNK due to chronic mitochondrial dysfunction and ROS. Cancer Cell 31, 771–789 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Canli, Ö. et al. Myeloid cell-derived reactive oxygen species induce epithelial mutagenesis. Cancer Cell 32, 869–883 (2017).

    CAS  PubMed  Google Scholar 

  102. Teoh, N. C. et al. Defective DNA strand break repair causes chromosomal instability and accelerates liver carcinogenesis in mice. Hepatology 47, 2078–2088 (2008).

    CAS  PubMed  Google Scholar 

  103. Delire, B. & Stärkel, P. The Ras/MAPK pathway and hepatocarcinoma: pathogenesis and therapeutic implications. Eur. J. Clin. Invest. 45, 609–623 (2015).

    CAS  PubMed  Google Scholar 

  104. Theurillat, J. P. et al. URI is an oncogene amplified in ovarian cancer cells and is required for their survival. Cancer Cell 19, 317–332 (2011).

    CAS  PubMed  Google Scholar 

  105. Tummala, K. S. et al. Inhibition of de novo NAD+ synthesis by oncogenic URI causes liver tumorigenesis through DNA damage. Cancer Cell 26, 826–839 (2014).

    CAS  PubMed  Google Scholar 

  106. Gomes, A. L. et al. Metabolic inflammation-associated IL-17A causes non-alcoholic steatohepatitis and hepatocellular carcinoma. Cancer Cell 30, 161–175 (2016).

    CAS  PubMed  Google Scholar 

  107. Boege, Y. et al. A dual role of caspase-8 in triggering and sensing proliferation-associated DNA damage, a key determinant of liver cancer development. Cancer Cell 32, 342–359 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Lin, Z. et al. Prognostic value of DNA repair based stratification of hepatocellular carcinoma. Sci. Rep. 6, 25999 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Daugherity, E. K. et al. The DNA damage checkpoint protein ATM promotes hepatocellular apoptosis and fibrosis in a mouse model of non-alcoholic fatty liver disease. Cell Cycle 11, 1918–1928 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. McKinnon, P. J. ATM and the molecular pathogenesis of ataxia telangiectasia. Annu. Rev. Pathol. 7, 303–321 (2012).

    CAS  PubMed  Google Scholar 

  111. Ditch, S. & Paull, T. T. The ATM protein kinase and cellular redox signaling: beyond the DNA damage response. Trends Biochem. Sci. 37, 15–22 (2012).

    CAS  PubMed  Google Scholar 

  112. Guo, Z., Kozlov, S., Lavin, M. F., Person, M. D. & Paull, T. T. ATM activation by oxidative stress. Science 330, 517–521 (2010).

    CAS  PubMed  Google Scholar 

  113. Gao, D. et al. Oxidative DNA damage and DNA repair enzyme expression are inversely related in murine models of fatty liver disease. Am. J. Physiol. Gastrointest. Liver Physiol. 287, G1070–1077 (2004).

    CAS  PubMed  Google Scholar 

  114. Schults, M. A. et al. Decreased nucleotide excision repair in steatotic livers associates with myeloperoxidase-immunoreactivity. Mutat. Res. 736, 75–81 (2012).

    CAS  PubMed  Google Scholar 

  115. Collis, S. J., DeWeese, T. L., Jeggo, P. A. & Parker, A. R. The life and death of DNA-PK. Oncogene 24, 949–961 (2005).

    CAS  PubMed  Google Scholar 

  116. Wong, R. H. et al. A role of DNA-PK for the metabolic gene regulation in response to insulin. Cell 136, 1056–1072 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Fautrel, A. et al. Overexpression of the two nucleotide excision repair genes ERCC1 and XPC in human hepatocellular carcinoma. J. Hepatol. 43, 288–293 (2005).

    CAS  PubMed  Google Scholar 

  118. Cornell, L. et al. DNA-PK-A candidate driver of hepatocarcinogenesis and tissue biomarker that predicts response to treatment and survival. Clin. Cancer Res. 21, 925–933 (2015).

    CAS  PubMed  Google Scholar 

  119. Evert, M. et al. Deregulation of DNA-dependent protein kinase catalytic subunit contributes to human hepatocarcinogenesis development and has a putative prognostic value. Br. J. Cancer 109, 2654–2664 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Pascale, R. M. et al. DNA-PKcs: a promising therapeutic target in human hepatocellular carcinoma? DNA Repair (Amst.) 47, 12–20 (2016).

    CAS  Google Scholar 

  121. Pfeifer, U. Inverted diurnal rhythm of cellular autophagy in liver cells of rats fed a single daily meal. Virchows Arch. B Cell Pathol. 10, 1–3 (1972).

    CAS  PubMed  Google Scholar 

  122. Khaminets, A., Behl, C. & Dikic, I. Ubiquitin-dependent and independent signals in selective autophagy. Trends Cell Biol. 26, 6–16 (2016).

    CAS  PubMed  Google Scholar 

  123. Kim, J., Kundu, M., Viollet, B. & Guan, K. L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, 132–141 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Taniguchi, K., Yamachika, S., He, F. & Karin, M. p62/SQSTM1-Dr. Jekyll and Mr. Hyde that prevents oxidative stress but promotes liver cancer. FEBS Lett. 590, 2375–2397 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Ichimura, Y. et al. Phosphorylation of p62 activates the Keap1-Nrf2 pathway during selective autophagy. Mol. Cell 51, 618–631 (2013).

    CAS  PubMed  Google Scholar 

  126. Taguchi, K., Motohashi, H. & Yamamoto, M. Molecular mechanisms of the Keap1-Nrf2 pathway in stress response and cancer evolution. Genes Cells 16, 123–140 (2011).

    CAS  PubMed  Google Scholar 

  127. Blommaart, E. F., Krause, U., Schellens, J. P., Vreeling-Sindelarova, H. & Meijer, A. J. The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes. Eur. J. Biochem. 243, 240–246 (1997).

    CAS  PubMed  Google Scholar 

  128. Singh, R. et al. Autophagy regulates lipid metabolism. Nature 458, 1131–1135 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Tanaka, S. et al. Rubicon inhibits autophagy and accelerates hepatocyte apoptosis and lipid accumulation in nonalcoholic fatty liver disease in mice. Hepatology 64, 1994–2014 (2016).

    CAS  PubMed  Google Scholar 

  130. Ramos-Gomez, M. et al. Sensitivity to carcinogenesis is increased and chemoprotective efficacy of enzyme inducers is lost in nrf2 transcription factor-deficient mice. Proc. Natl Acad. Sci. USA 98, 3410–3415 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Guichard, C. et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 44, 694–698 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Totoki, Y. et al. Trans-ancestry mutational landscape of hepatocellular carcinoma genomes. Nat. Genet. 46, 1267–1273 (2014).

    CAS  PubMed  Google Scholar 

  133. Schulze, K. et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat. Genet. 47, 505–511 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Zavattari, P. et al. Nrf2, but not beta-catenin, mutation represents an early event in rat hepatocarcinogenesis. Hepatology 62, 851–862 (2015).

    CAS  PubMed  Google Scholar 

  135. Petrelli, A. et al. MicroRNA/gene profiling unveils early molecular changes and nuclear factor erythroid related factor 2 (NRF2) activation in a rat model recapitulating human hepatocellular carcinoma (HCC). Hepatology 59, 228–241 (2014).

    CAS  PubMed  Google Scholar 

  136. Kim, H. et al. Human hepatocellular carcinomas with “Stemness”-related marker expression: keratin 19 expression and a poor prognosis. Hepatology 54, 1707–1717 (2011).

    CAS  PubMed  Google Scholar 

  137. Govaere, O. et al. Keratin 19: a key role player in the invasion of human hepatocellular carcinomas. Gut 63, 674–685 (2014).

    CAS  PubMed  Google Scholar 

  138. Govaere, O. et al. Laminin-332 sustains chemoresistance and quiescence as part of the human hepatic cancer stem cell niche. J. Hepatol. 64, 609–617 (2016).

    CAS  PubMed  Google Scholar 

  139. Umemura, A. et al. p62, upregulated during preneoplasia, induces hepatocellular carcinogenesis by maintaining survival of stressed HCC-initiating cells. Cancer Cell 29, 935–948 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Xu, L. Z. et al. p62/SQSTM1 enhances breast cancer stem-like properties by stabilizing MYC mRNA. Oncogene 36, 304–317 (2017).

    CAS  PubMed  Google Scholar 

  141. Yu, L. et al. Regulation of an ATG7-beclin 1 program of autophagic cell death by caspase-8. Science 304, 1500–1502 (2004).

    CAS  PubMed  Google Scholar 

  142. Takamura, A. et al. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 25, 795–800 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Nakagawa, H. et al. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 26, 331–343 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Puri, P. et al. Activation and dysregulation of the unfolded protein response in nonalcoholic fatty liver disease. Gastroenterology 134, 568–576 (2008).

    CAS  PubMed  Google Scholar 

  145. Ley, R. E. et al. Obesity alters gut microbial ecology. Proc. Natl Acad. Sci. USA 102, 11070–11075 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Ley, R. E., Turnbaugh, P. J., Klein, S. & Gordon, J. I. Microbial ecology: human gut microbes associated with obesity. Nature 444, 1022–1023 (2006).

    CAS  PubMed  Google Scholar 

  147. Turnbaugh, P. J., Bäckhed, F., Fulton, L. & Gordon, J. I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3, 213–223 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Le Chatelier, E. et al. Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541–546 (2013).

    PubMed  Google Scholar 

  149. Murphy, E. F. et al. Divergent metabolic outcomes arising from targeted manipulation of the gut microbiota in diet-induced obesity. Gut 62, 220–226 (2013).

    PubMed  Google Scholar 

  150. Karlsson, F. H. et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99–103 (2013).

    CAS  PubMed  Google Scholar 

  151. Qin, J. et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55–60 (2012).

    CAS  PubMed  Google Scholar 

  152. Vrieze, A. et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143, 913–916 (2012).

    CAS  PubMed  Google Scholar 

  153. Larsen, N. et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLOS ONE 5, e9085 (2010).

    PubMed  PubMed Central  Google Scholar 

  154. Boursier, J. & Diehl, A. M. Implication of gut microbiota in nonalcoholic fatty liver disease. PLOS Pathog. 11, e1004559 (2015).

    PubMed  PubMed Central  Google Scholar 

  155. Zhu, L. et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57, 601–609 (2013).

    CAS  PubMed  Google Scholar 

  156. Mouzaki, M. et al. Bile acids and dysbiosis in non-alcoholic fatty liver disease. PLOS ONE 11, e0151829 (2016).

    PubMed  PubMed Central  Google Scholar 

  157. Llopis, M. et al. Intestinal microbiota contributes to individual susceptibility to alcoholic liver disease. Gut 65, 830–839 (2016).

    CAS  PubMed  Google Scholar 

  158. Le Roy, T. et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 62, 1787–1794 (2013).

    PubMed  Google Scholar 

  159. Turner, J. R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 9, 799–809 (2009).

    CAS  PubMed  Google Scholar 

  160. Miele, L. et al. Increased intestinal permeability and tight junction alterations in nonalcoholic fatty liver disease. Hepatology 49, 1877–1887 (2009).

    CAS  PubMed  Google Scholar 

  161. Volynets, V. et al. Nutrition, intestinal permeability, and blood ethanol levels are altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig. Dis. Sci. 57, 1932–1941 (2012).

    CAS  PubMed  Google Scholar 

  162. Luther, J. et al. Hepatic injury in nonalcoholic steatohepatitis contributes to altered intestinal permeability. Cell. Mol. Gastroenterol. Hepatol. 1, 222–232 (2015).

    PubMed  PubMed Central  Google Scholar 

  163. Kolodziejczyk, A. A., Zheng, D., Shibolet, O. & Elinav, E. The role of the microbiome in NAFLD and NASH. EMBO Mol. Med. 11, e9302 (2018).

    PubMed Central  Google Scholar 

  164. Gäbele, E. et al. DSS induced colitis increases portal LPS levels and enhances hepatic inflammation and fibrogenesis in experimental NASH. J. Hepatol. 55, 1391–1399 (2011).

    PubMed  Google Scholar 

  165. Chavez-Talavera, O., Tailleux, A., Lefebvre, P. & Staels, B. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 152, 1679–1694 (2017).

    CAS  PubMed  Google Scholar 

  166. Swann, J. R. et al. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc. Natl Acad. Sci. USA 108 (Suppl. 1), 4523–4530 (2011).

    CAS  PubMed  Google Scholar 

  167. Sayin, S. I. et al. Gut microbiota regulates bile acid metabolism by reducing the levels of tauro-beta-muricholic acid, a naturally occurring FXR antagonist. Cell Metab. 17, 225–235 (2013).

    CAS  PubMed  Google Scholar 

  168. Lefebvre, P., Cariou, B., Lien, F., Kuipers, F. & Staels, B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol. Rev. 89, 147–191 (2009).

    CAS  PubMed  Google Scholar 

  169. Wahlström, A. et al. Induction of farnesoid X receptor signaling in germ-free mice colonized with a human microbiota. J. Lipid Res. 58, 412–419 (2017).

    PubMed  PubMed Central  Google Scholar 

  170. Sinal, C. J. et al. Targeted disruption of the nuclear receptor FXR/BAR impairs bile acid and lipid homeostasis. Cell 102, 731–744 (2000).

    CAS  PubMed  Google Scholar 

  171. Seyer, P. et al. Hepatic glucose sensing is required to preserve β cell glucose competence. J. Clin. Invest. 123, 1662–1676 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Claudel, T., Staels, B. & Kuipers, F. The Farnesoid X receptor: a molecular link between bile acid and lipid and glucose metabolism. Arterioscler. Thromb. Vasc. Biol. 25, 2020–2030 (2005).

    CAS  PubMed  Google Scholar 

  173. Matsubara, T., Li, F. & Gonzalez, F. J. FXR signaling in the enterohepatic system. Mol. Cell Endocrinol. 368, 17–29 (2013).

    CAS  PubMed  Google Scholar 

  174. Li, G. & Guo, G. L. Farnesoid X receptor, the bile acid sensing nuclear receptor, in liver regeneration. Acta Pharm. Sin. B 5, 93–98 (2015).

    PubMed  PubMed Central  Google Scholar 

  175. Zhu, Y., Liu, H., Zhang, M. & Guo, G. L. Fatty liver diseases, bile acids, and FXR. Acta Pharm. Sin. B 6, 409–412 (2016).

    PubMed  PubMed Central  Google Scholar 

  176. Li, G. & Guo, G. L. Role of class II nuclear receptors in liver carcinogenesis. Anticancer Agents Med. Chem. 11, 529–542 (2011).

    PubMed  Google Scholar 

  177. Wagner, M., Zollner, G. & Trauner, M. Nuclear receptors in liver disease. Hepatology 53, 1023–1034 (2011).

    CAS  PubMed  Google Scholar 

  178. Armstrong, L. E. & Guo, G. L. Role of FXR in liver inflammation during nonalcoholic steatohepatitis. Curr. Pharmacol. Rep. 3, 92–100 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Forman, B. M. et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 81, 687–693 (1995).

    CAS  PubMed  Google Scholar 

  180. Li, F. et al. Microbiome remodelling leads to inhibition of intestinal farnesoid X receptor signalling and decreased obesity. Nat. Commun. 4, 2384 (2013).

    PubMed  Google Scholar 

  181. Jiang, C. et al. Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction. Nat. Commun. 6, 10166 (2015).

    CAS  PubMed  Google Scholar 

  182. Jiang, C. et al. Intestinal farnesoid X receptor signaling promotes nonalcoholic fatty liver disease. J. Clin. Invest. 125, 386–402 (2015).

    PubMed  Google Scholar 

  183. Parséus, A. et al. Microbiota-induced obesity requires farnesoid X receptor. Gut 66, 429–437 (2017).

    PubMed  Google Scholar 

  184. Tremaroli, V. et al. Roux-en-Y gastric bypass and vertical banded gastroplasty induce long-term changes on the human gut microbiome contributing to fat mass regulation. Cell Metab. 22, 228–238 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Ryan, K. K. et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature 509, 183–188 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Massafra, V. & van Mil, S. W. C. Farnesoid X receptor: a “homeostat” for hepatic nutrient metabolism. Biochim. Biophys. Acta 1864, 45–59 (2018).

    CAS  Google Scholar 

  187. Stanimirov, B., Stankov, K. & Mikov, M. Pleiotropic functions of bile acids mediated by the farnesoid X receptor. Acta Gastroenterol. Belg. 75, 389–398 (2012).

    CAS  PubMed  Google Scholar 

  188. Mudaliar, S. et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology 145, 574–582 (2013).

    CAS  PubMed  Google Scholar 

  189. Prawitt, J. et al. Farnesoid X receptor deficiency improves glucose homeostasis in mouse models of obesity. Diabetes 60, 1861–1871 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Zhang, Y. et al. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc. Natl Acad. Sci. USA 103, 1006–1011 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  191. Fang, S. et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nat. Med. 21, 159–165 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Watanabe, M. et al. Lowering bile acid pool size with a synthetic farnesoid X receptor (FXR) agonist induces obesity and diabetes through reduced energy expenditure. J. Biol. Chem. 286, 26913–26920 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Fiorucci, S., Zampella, A. & Distrutti, E. Development of FXR, PXR and CAR agonists and antagonists for treatment of liver disorders. Curr. Top. Med. Chem. 12, 605–624 (2012).

    CAS  PubMed  Google Scholar 

  194. Wang, X. et al. Bile acid receptors and liver cancer. Curr. Pathobiol Rep. 1, 29–35 (2013).

    PubMed  Google Scholar 

  195. Chen, T. et al. Serum and urine metabolite profiling reveals potential biomarkers of human hepatocellular carcinoma. Mol. Cell Proteomics https://doi.org/10.1074/mcp.M110.004945 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  196. Jansen, P. L. Endogenous bile acids as carcinogens. J. Hepatol. 47, 434–435 (2007).

    CAS  PubMed  Google Scholar 

  197. Chiang, J. Y. L. Bile acid metabolism and signaling in liver disease and therapy. Liver Res. 1, 3–9 (2017).

    PubMed  PubMed Central  Google Scholar 

  198. Knisely, A. S. et al. Hepatocellular carcinoma in ten children under five years of age with bile salt export pump deficiency. Hepatology 44, 478–486 (2006).

    CAS  PubMed  Google Scholar 

  199. Yang, F. et al. Spontaneous development of liver tumors in the absence of the bile acid receptor farnesoid X receptor. Cancer Res. 67, 863–867 (2007).

    CAS  PubMed  Google Scholar 

  200. Degirolamo, C. et al. Prevention of spontaneous hepatocarcinogenesis in farnesoid X receptor-null mice by intestinal-specific farnesoid X receptor reactivation. Hepatology 61, 161–170 (2015).

    CAS  PubMed  Google Scholar 

  201. Calle, E. E. Obesity and cancer. BMJ 335, 1107–1108 (2007).

    PubMed  PubMed Central  Google Scholar 

  202. Tuncman, G. et al. Functional in vivo interactions between JNK1 and JNK2 isoforms in obesity and insulin resistance. Proc. Natl Acad. Sci. USA 103, 10741–10746 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Park, J., Morley, T. S., Kim, M., Clegg, D. J. & Scherer, P. E. Obesity and cancer—mechanisms underlying tumour progression and recurrence. Nat. Rev. Endocrinol. 10, 455–465 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  204. Park, E. J. et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140, 197–208 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Rakhra, K. et al. CD4+ T cells contribute to the remodeling of the microenvironment required for sustained tumor regression upon oncogene inactivation. Cancer Cell 18, 485–498 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  206. Kang, T. W. et al. Senescence surveillance of pre-malignant hepatocytes limits liver cancer development. Nature 479, 547–551 (2011).

    CAS  PubMed  Google Scholar 

  207. Ma, C. et al. NAFLD causes selective CD4+ T lymphocyte loss and promotes hepatocarcinogenesis. Nature 531, 253–257 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Fu, J. et al. Increased regulatory T cells correlate with CD8 T cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 132, 2328–2339 (2007).

    PubMed  Google Scholar 

  209. Gao, Q. et al. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. J. Clin. Oncol. 25, 2586–2593 (2007).

    PubMed  Google Scholar 

  210. Hoechst, B. et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology 50, 799–807 (2009).

    CAS  PubMed  Google Scholar 

  211. Kubes, P. & Mehal, W. Z. Sterile inflammation in the liver. Gastroenterology 143, 1158–1172 (2012).

    CAS  PubMed  Google Scholar 

  212. Davis, B. K., Wen, H. & Ting, J. P. The inflammasome NLRs in immunity, inflammation, and associated diseases. Annu. Rev. Immunol. 29, 707–735 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Arrese, M., Cabrera, D., Kalergis, A. M. & Feldstein, A. E. Innate Immunity and Inflammation in NAFLD/NASH. Dig. Dis. Sci. 61, 1294–1303 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Feldstein, A. E. Novel insights into the pathophysiology of nonalcoholic fatty liver disease. Semin. Liver Dis. 30, 391–401 (2010).

    CAS  PubMed  Google Scholar 

  215. Peverill, W., Powell, L. W. & Skoien, R. Evolving concepts in the pathogenesis of NASH: beyond steatosis and inflammation. Int. J. Mol. Sci. 15, 8591–8638 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Lanthier, N. Targeting Kupffer cells in non-alcoholic fatty liver disease/non-alcoholic steatohepatitis: why and how? World J. Hepatol. 7, 2184–2188 (2015).

    PubMed  PubMed Central  Google Scholar 

  217. Reid, D. T. et al. Kupffer cells undergo fundamental changes during the development of experimental NASH and are critical in initiating liver damage and inflammation. PLOS ONE 11, e0159524 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Bieghs, V. & Trautwein, C. The innate immune response during liver inflammation and metabolic disease. Trends Immunol. 34, 446–452 (2013).

    CAS  PubMed  Google Scholar 

  219. Miura, K. et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology 139, 323–334 (2010).

    CAS  PubMed  Google Scholar 

  220. Wu, J. et al. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res. 72, 3977–3986 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Ju, C. & Tacke, F. Hepatic macrophages in homeostasis and liver diseases: from pathogenesis to novel therapeutic strategies. Cell. Mol. Immunol. 13, 316–327 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Malehmir, M. et al. Platelet GPIba is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. (in the press).

  223. Kurien, B. T. & Scofield, R. H. Autoimmunity and oxidatively modified autoantigens. Autoimmun. Rev. 7, 567–573 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Nobili, V. et al. Oxidative stress parameters in paediatric non-alcoholic fatty liver disease. Int. J. Mol. Med. 26, 471–476 (2010).

    CAS  PubMed  Google Scholar 

  225. DeFuria, J. et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T cell function and an inflammatory cytokine profile. Proc. Natl Acad. Sci. USA 110, 5133–5138 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. McPherson, S., Henderson, E., Burt, A. D., Day, C. P. & Anstee, Q. M. Serum immunoglobulin levels predict fibrosis in patients with non-alcoholic fatty liver disease. J. Hepatol. 60, 1055–1062 (2014).

    CAS  PubMed  Google Scholar 

  227. Shalapour, S. et al. Inflammation-induced IgA+cells dismantle anti-liver cancer immunity. Nature 551, 340–345 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 169, 361–371 (2017).

    CAS  PubMed  Google Scholar 

  229. Matter, M. S., Decaens, T., Andersen, J. B. & Thorgeirsson, S. S. Targeting the mTOR pathway in hepatocellular carcinoma: current state and future trends. J. Hepatol. 60, 855–865 (2014).

    CAS  PubMed  Google Scholar 

  230. Guri, Y., Nordmann, T. M. & Roszik, J. mTOR at the transmitting and receiving ends in tumor immunity. Front. Immunol. 9, 578 (2018).

    PubMed  PubMed Central  Google Scholar 

  231. Hagiwara, A. et al. Hepatic mTORC2 activates glycolysis and lipogenesis through Akt, glucokinase, and SREBP1c. Cell Metab. 15, 725–738 (2012).

    CAS  PubMed  Google Scholar 

  232. Guri, Y. et al. mTORC2 promotes tumorigenesis via lipid synthesis. Cancer Cell 32, 807–823 (2017).

    CAS  PubMed  Google Scholar 

  233. European Association for the Study of the Liver & European Organisation for Research and Treatment of Cancer. EASL-EORTC clinical practice guidelines: management of hepatocellular carcinoma. J. Hepatol. 56, 908–943 (2012).

    Google Scholar 

  234. Heimbach, J. K. et al. Aasld guidelines for the treatment of hepatocellular carcinoma. Hepatology 67, 358–380 (2017).

    Google Scholar 

  235. Taylor, E. J., Jones, R. L., Guthrie, J. A. & Rowe, I. A. Modeling the benefits and harms of surveillance for hepatocellular carcinoma: Information to support informed choices. Hepatology 66, 1546–1555 (2017).

    PubMed  Google Scholar 

  236. Singal, A. G. et al. Failure rates in the hepatocellular carcinoma surveillance process. Cancer Prev. Res. (Phila.) 5, 1124–1130 (2012).

    Google Scholar 

  237. Mittal, S. et al. Temporal trends of nonalcoholic fatty liver disease-related hepatocellular carcinoma in the veteran affairs population. Clin. Gastroenterol. Hepatol. 13, 594–601 (2015).

    PubMed  Google Scholar 

  238. Della Corte, C. & Colombo, M. Surveillance for hepatocellular carcinoma. Semin. Oncol. 39, 384–398 (2012).

    PubMed  Google Scholar 

  239. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD) & European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. Diabetologia 59, 1121–1140 (2016).

    Google Scholar 

  240. Stickel, F. & Hellerbrand, C. Non-alcoholic fatty liver disease as a risk factor for hepatocellular carcinoma: mechanisms and implications. Gut 59, 1303–1307 (2010).

    CAS  PubMed  Google Scholar 

  241. Morling, J. R. et al. Clinically significant chronic liver disease in people with type 2 diabetes: the Edinburgh type 2 diabetes study. QJM 109, 249–256 (2016).

    CAS  PubMed  Google Scholar 

  242. Wen, C. P. et al. Hepatocellular carcinoma risk prediction model for the general population: the predictive power of transaminases. J. Natl Cancer Inst. 104, 1599–1611 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Berhane, S. et al. Role of the GALAD and BALAD-2 serologic models in diagnosis of hepatocellular carcinoma and prediction of survival in patients. Clin. Gastroenterol. Hepatol. 14, 875–886 (2016).

    CAS  PubMed  Google Scholar 

  244. Hwang, A. et al. Supervised learning reveals circulating biomarker levels diagnostic of hepatocellular carcinoma in a clinically relevant model of non-alcoholic steatohepatitis; an OAD to NASH. PLOS ONE 13, e0198937 (2018).

    PubMed  PubMed Central  Google Scholar 

  245. Ng, C. K. Y., Di Costanzo, G. G., Terracciano, L. M. & Piscuoglio, S. Circulating cell-free DNA in hepatocellular carcinoma: current insights and outlook. Front. Med. (Lausanne) 5, 78 (2018).

    Google Scholar 

  246. Zhang, Y. J. et al. Predicting hepatocellular carcinoma by detection of aberrant promoter methylation in serum DNA. Clin. Cancer Res. 13, 2378–2384 CCR-06-1900 (2007).

    CAS  PubMed  Google Scholar 

  247. Tangkijvanich, P. et al. Serum LINE-1 hypomethylation as a potential prognostic marker for hepatocellular carcinoma. Clin. Chim. Acta 379, 127–133 (2007).

    CAS  PubMed  Google Scholar 

  248. Xu, R. H. et al. Circulating tumour DNA methylation markers for diagnosis and prognosis of hepatocellular carcinoma. Nat. Mater. 16, 1155–1161 (2017).

    CAS  PubMed  Google Scholar 

  249. Yáñez-Mó, M. et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 4, 27066 (2015).

    PubMed  Google Scholar 

  250. Hirsova, P. et al. Extracellular vesicles in liver pathobiology: small particles with big impact. Hepatology 64, 2219–2233 (2016).

    PubMed  Google Scholar 

  251. Arbelaiz, A. et al. Serum extracellular vesicles contain protein biomarkers for primary sclerosing cholangitis and cholangiocarcinoma. Hepatology 66, 1125–1143 (2017).

    CAS  PubMed  Google Scholar 

  252. Torbenson, M. & Schirmacher, P. Liver cancer biopsy - back to the future?! Hepatology 61, 431–433 (2015).

    PubMed  Google Scholar 

  253. Sherman, M. & Bruix, J. Biopsy for liver cancer: How to balance research needs with evidence-based clinical practice. Hepatology 61, 433–437 (2015).

    PubMed  Google Scholar 

  254. Friemel, J. et al. Intratumor heterogeneity in hepatocellular carcinoma. Clin. Cancer Res. 21, 1951–1961 (2015).

    CAS  PubMed  Google Scholar 

  255. Bruix, J. et al. Prognostic factors and predictors of sorafenib benefit in patients with hepatocellular carcinoma: analysis of two phase III studies. J. Hepatol. 67, 999–1008 (2017).

    CAS  PubMed  Google Scholar 

  256. Llovet, J. M. et al. Sorafenib in advanced hepatocellular carcinoma. N. Engl. J. Med. 359, 378–390 (2008).

    CAS  PubMed  Google Scholar 

  257. Cheng, A. L. et al. Efficacy and safety of sorafenib in patients in the Asia-Pacific region with advanced hepatocellular carcinoma: a phase III randomised, double-blind, placebo-controlled trial. Lancet Oncol. 10, 25–34 (2009).

    CAS  PubMed  Google Scholar 

  258. Mehnert, J. M. et al. The challenge for development of valuable immuno-oncology biomarkers. Clin. Cancer Res. 23, 4970–4979 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Pantel, K. & Alix-Panabieres, C. Liquid biopsy in 2016: circulating tumour cells and cell-free DNA in gastrointestinal cancer. Nat. Rev. Gastroenterol. Hepatol. 14, 73–74 (2017).

    CAS  PubMed  Google Scholar 

  260. Ogle, L. F. et al. Imagestream detection and characterisation of circulating tumour cells - A liquid biopsy for hepatocellular carcinoma? J. Hepatol. 65, 305–313 (2016).

    CAS  PubMed  Google Scholar 

  261. Sun, Y. F. et al. Circulating stem cell-like epithelial cell adhesion molecule-positive tumor cells indicate poor prognosis of hepatocellular carcinoma after curative resection. Hepatology 57, 1458–1468 (2013).

    CAS  PubMed  Google Scholar 

  262. Li, J. et al. Detection of circulating tumor cells in hepatocellular carcinoma using antibodies against asialoglycoprotein receptor, carbamoyl phosphate synthetase 1 and pan-cytokeratin. PLOS ONE 9, e96185 (2014).

    PubMed  PubMed Central  Google Scholar 

  263. Kalinich, M. et al. An RNA-based signature enables high specificity detection of circulating tumor cells in hepatocellular carcinoma. Proc. Natl Acad. Sci. USA 114, 1123–1128 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Morris, K. L. et al. Circulating biomarkers in hepatocellular carcinoma. Cancer Chemother. Pharmacol. 74, 323–332 (2014).

    CAS  PubMed  Google Scholar 

  265. Sun, Y. F. et al. Circulating tumor cells from different vascular sites exhibit spatial heterogeneity in epithelial and mesenchymal composition and distinct clinical significance in hepatocellular carcinoma. Clin. Cancer Res. 24, 547–559 (2018).

    CAS  PubMed  Google Scholar 

  266. Li, J. et al. pERK/pAkt phenotyping in circulating tumor cells as a biomarker for sorafenib efficacy in patients with advanced hepatocellular carcinoma. Oncotarget 7, 2646–2659 (2016).

    PubMed  Google Scholar 

  267. Lorentzen, A. et al. Single cell polarity in liquid phase facilitates tumour metastasis. Nat. Commun. 9, 887 (2018).

    PubMed  PubMed Central  Google Scholar 

  268. Prentis, J. M. et al. Submaximal cardiopulmonary exercise testing predicts 90-day survival after liver transplantation. Liver Transpl. 18, 152–159 (2012).

    PubMed  Google Scholar 

  269. Kolly, P. et al. Assessment of the Hong Kong Liver Cancer Staging System in Europe. Liver Int. 36, 911–917 (2016).

    PubMed  Google Scholar 

  270. Kadalayil, L. et al. A simple prognostic scoring system for patients receiving transarterial embolisation for hepatocellular cancer. Ann. Oncol. 24, 2565–2570 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  271. Sieghart, W. et al. The ART of decision making: retreatment with transarterial chemoembolization in patients with hepatocellular carcinoma. Hepatology 57, 2261–2273 (2013).

    CAS  PubMed  Google Scholar 

  272. Varela, M. et al. Chemoembolization of hepatocellular carcinoma with drug eluting beads: efficacy and doxorubicin pharmacokinetics. J. Hepatol. 46, 474–481 (2007).

    CAS  PubMed  Google Scholar 

  273. Vilgrain, V. et al. Efficacy and safety of selective internal radiotherapy with yttrium-90 resin microspheres compared with sorafenib in locally advanced and inoperable hepatocellular carcinoma (SARAH): an open-label randomised controlled phase 3 trial. Lancet Oncol. 18, 1624–1636 (2017).

    CAS  PubMed  Google Scholar 

  274. Kirstein, M. M. et al. Patterns and challenges of treatment sequencing in patients with hepatocellular carcinoma: experience from a German referral center. J. Gastroenterol. Hepatol. 32, 1730–1738 (2017).

    CAS  PubMed  Google Scholar 

  275. Pardo, F. et al. The Post-SIR-Spheres Surgery Study (P4S): retrospective analysis of safety following hepatic resection or transplantation in patients previously treated with selective internal radiation therapy with Yttrium-90 resin microspheres. Ann. Surg. Oncol. 24, 2465–2473 (2017).

    PubMed  Google Scholar 

  276. Moir, J. A. et al. Selective internal radiation therapy for liver malignancies. Br. J. Surg. 102, 1533–1540 (2015).

    CAS  PubMed  Google Scholar 

  277. Kudo, M. Lenvatinib in advanced hepatocellular carcinoma. Liver Cancer 6, 253–263 (2017).

    PubMed  PubMed Central  Google Scholar 

  278. Bruix, J. et al. Regorafenib for patients with hepatocellular carcinoma who progressed on sorafenib treatment (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 389, 56–66 (2017).

    CAS  PubMed  Google Scholar 

  279. Abou-Alfa, G. K. et al. Cabozantinib in patients with advanced and progressing hepatocellular carcinoma. N. Engl. J. Med. 379, 54–63 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  280. Kudo, M. Immune checkpoint blockade in hepatocellular carcinoma. Liver Cancer 4, 201–207 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  281. El-Khoueiry, A. B. et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 389, 2492–2502 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  282. Raoul, J. L. et al. Systemic therapy for intermediate and advanced hepatocellular carcinoma: Sorafenib and beyond. Cancer Treat. Rev. 68, 16–24 (2018).

    CAS  PubMed  Google Scholar 

  283. Vilar-Gomez, E. et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology 149, 367–378 (2015).

    PubMed  Google Scholar 

  284. Mishra, S. I. et al. Exercise interventions on health-related quality of life for cancer survivors. Cochrane Database Syst. Rev. 8, CD007566 (2012).

    Google Scholar 

  285. Mishra, S. I. et al. Exercise interventions on health-related quality of life for people with cancer during active treatment. Cochrane Database Syst. Rev. 8, CD008465 (2012).

    Google Scholar 

  286. Gustafson, M. P. et al. A systems biology approach to investigating the influence of exercise and fitness on the composition of leukocytes in peripheral blood. J. Immunother. Cancer 5, 30 (2017).

    PubMed  PubMed Central  Google Scholar 

  287. Idorn, M. & Thor Straten, P. Exercise: a new role for an old tool. Mol. Cell Oncol. 3, e1163005 (2016).

    PubMed  PubMed Central  Google Scholar 

  288. Koelwyn, G. J., Quail, D. F., Zhang, X., White, R. M. & Jones, L. W. Exercise-dependent regulation of the tumour microenvironment. Nat. Rev. Cancer 17, 620–632 (2017).

    PubMed  Google Scholar 

  289. McCuskey, R. S. et al. Hepatic microvascular dysfunction during evolution of dietary steatohepatitis in mice. Hepatology 40, 386–393 (2004).

    PubMed  Google Scholar 

  290. Iyer, S., Upadhyay, P. K., Majumdar, S. S. & Nagarajan, P. Animal models correlating immune cells for the development of NAFLD/NASH. J. Clin. Exp. Hepatol. 5, 239–245 (2015).

    PubMed  PubMed Central  Google Scholar 

  291. Denda, A. et al. Development of hepatocellular adenomas and carcinomas associated with fibrosis in C57BL/6J male mice given a choline-deficient, L-amino acid-defined diet. Jpn J. Cancer Res. 93, 125–132 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  292. Matsumoto, M. et al. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int. J. Exp. Pathol. 94, 93–103 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  293. Ikawa-Yoshida, A. et al. Hepatocellular carcinoma in a mouse model fed a choline-deficient, L-amino acid-defined, high-fat diet. Int. J. Exp. Pathol. 98, 221–233 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  294. Sun, B. & Karin, M. Obesity, inflammation, and liver cancer. J. Hepatol. 56, 704–713 (2012).

    CAS  PubMed  Google Scholar 

  295. Charlton, M. et al. Fast food diet mouse: novel small animal model of NASH with ballooning, progressive fibrosis, and high physiological fidelity to the human condition. Am. J. Physiol. Gastrointest. Liver Physiol. 301, G825–G834 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  296. Asgharpour, A. et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J. Hepatol. 65, 579–588 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  297. Wang, Y., Ausman, L. M., Greenberg, A. S., Russell, R. M. & Wang, X. D. Nonalcoholic steatohepatitis induced by a high-fat diet promotes diethylnitrosamine-initiated early hepatocarcinogenesis in rats. Int. J. Cancer 124, 540–546 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  298. Tsuchida, T. et al. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J. Hepatol. 69, 385–395 (2018).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Q.M.A., O.G. and H.L.R. are members of the EPoS (Elucidating Pathways of Steatohepatitis) consortium funded by the Horizon 2020 Framework Programme of the European Union under Grant Agreement 634413 and the Newcastle National Institute for Health Research Biomedical Research Centre. At the Northern Institute for Cancer Research, H.L.R. is supported by the Bobby Robson Foundation, Cancer Research UK (CRUK) Newcastle Experimental Cancer Medicine Centre award C9380/A18084 and CRUK programme grant C18342/A23390. This work was supported by the European Union’s Horizon 2020 Research and Innovation Programme (no. 667273/HEPCAR) to M.H.; a European Research Council Consolidator grant ‘HepatoMetaboPath’; the MOST (Ministry of Science and Technology) programme; Research Foundation Flanders (FWO) under grant 30826052 (EOS Convention MODEL-IDI); the Sonderforschungsbereiche (SFB) Transregio (TR) SFB/TR179, 209 and SFB1335 to M.H.; Graduiertenkolleg (GRK482) to E.K. and M.H.; and the German Cancer Research Center (DKFZ)–MOST cooperation program to M.H.

Reviewer information

Nature Reviews Gastroenterology & Hepatology thanks M. Machado and the other anonymous reviewer(s), for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Quentin M. Anstee or Mathias Heikenwalder.

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

Anstee, Q.M., Reeves, H.L., Kotsiliti, E. et al. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol 16, 411–428 (2019). https://doi.org/10.1038/s41575-019-0145-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-019-0145-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing