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:

The immunological and metabolic landscape in primary and metastatic liver cancer

Abstract

The liver is the sixth most common site of primary cancer in humans, and generally arises in a background of cirrhosis and inflammation. Moreover, the liver is frequently colonized by metastases from cancers of other organs (particularly the colon) because of its anatomical location and organization, as well as its unique metabolic and immunosuppressive environment. In this Review, we discuss how the hepatic microenvironment adapts to pathologies characterized by chronic inflammation and metabolic alterations. We illustrate how these immunological or metabolic changes alter immunosurveillance and thus hinder or promote the development of primary liver cancer. In addition, we describe how inflammatory and metabolic niches affect the spreading of cancer metastases into or within the liver. Finally, we review the current therapeutic options in this context and the resulting challenges that must be surmounted.

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 immune microenvironment of the healthy, inflamed and cancerous liver.
Fig. 2: The metabolic microenvironment of the healthy and inflamed liver.
Fig. 3: Liver cell plasticity and oncogenic transformation.
Fig. 4: The immunometabolic microenvironment of cancerous liver.
Fig. 5: The prometastatic and antimetastatic microenvironment of the liver.

Similar content being viewed by others

References

  1. Llovet, J. M. et al. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 7, 6 (2021). This article provides the latest progress in the pathogenic mechanisms of HCC and its therapy.

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  3. Keenan, B. P., Fong, L. & Kelley, R. K. Immunotherapy in hepatocellular carcinoma: the complex interface between inflammation, fibrosis, and the immune response. J. Immunother. Cancer 7, 1–13 (2019).

    Article  Google Scholar 

  4. Kanwal, F. et al. Risk of hepatocellular cancer in patients with non-alcoholic fatty liver disease. Gastroenterology 155, 1828–1837.e2 (2018).

    Article  PubMed  Google Scholar 

  5. Protzer, U., Maini, M. K. & Knolle, P. A. Living in the liver: hepatic infections. Nat. Rev. Immunol. 12, 201–213 (2012).

    Article  CAS  PubMed  Google Scholar 

  6. European Association for the Study of the Liver. EASL clinical practice guidelines: management of hepatocellular carcinoma. J. Hepatol. 69, 182–236 (2018).

    Article  Google Scholar 

  7. Carrat, F. et al. Clinical outcomes in patients with chronic hepatitis C after direct-acting antiviral treatment: a prospective cohort study. Lancet 393, 1453–1464 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Anstee, Q. M., Reeves, H. L., Kotsiliti, E., Govaere, O. & Heikenwalder, M. From NASH to HCC: current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 16, 411–428 (2019).

    Article  PubMed  Google Scholar 

  9. Steeg, P. S. Tumor metastasis: mechanistic insights and clinical challenges. Nat. Med. 12, 895–904 (2006).

    Article  CAS  PubMed  Google Scholar 

  10. Milette, S., Sicklick, J. K., Lowy, A. M. & Brodt, P. Molecular pathways: targeting the microenvironment of liver metastases. Clin. Cancer Res. 23, 6390–6399 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. de Ridder, J. et al. Incidence and origin of histologically confirmed liver metastases: an explorative case-study of 23,154 patients. Oncotarget 7, 55368–55376 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 70, 7–30 (2020).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Llovet, J. M., Montal, R. & Villanueva, A. Randomized trials and endpoints in advanced HCC: role of PFS as a surrogate of survival. J. Hepatol. 70, 1262–1277 (2019).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. Nault, J. C., Cheng, A. L., Sangro, B. & Llovet, J. M. Milestones in the pathogenesis and management of primary liver cancer. J. Hepatol. 72, 209–214 (2020). This paper summarizes important events in the medical history of primary liver cancer and discusses the most current discoveries.

    Article  PubMed  Google Scholar 

  17. Rizvi, S., Khan, S. A., Hallemeier, C. L., Kelley, R. K. & Gores, G. J. Cholangiocarcinoma — evolving concepts and therapeutic strategies. Nat. Rev. Clin. Oncol. 15, 95–111 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Vogel, A. et al. Hepatocellular carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 29, iv238–iv255 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Mizukoshi, E. & Kaneko, S. Immune cell therapy for hepatocellular carcinoma. J. Hematol. Oncol. 12, 52 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Kudo, M. Combination cancer immunotherapy in hepatocellular carcinoma. Liver Cancer 7, 20–27 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Heymann, F. et al. Liver inflammation abrogates immunological tolerance induced by Kupffer cells. Hepatology 62, 279–291 (2015).

    Article  CAS  PubMed  Google Scholar 

  22. Jenne, C. N. & Kubes, P. Immune surveillance by the liver. Nat. Immunol. 14, 996–1006 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. Knolle, P. A. & Thimme, R. Hepatic immune regulation and its involvement in viral hepatitis infection. Gastroenterology 146, 1193–1207 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Winau, F. et al. Ito cells are liver-resident antigen-presenting cells for activating T cell responses. Immunity 26, 117–129 (2007).

    Article  CAS  PubMed  Google Scholar 

  25. Thomson, A. W. & Knolle, P. A. Antigen-presenting cell function in the tolerogenic liver environment. Nat. Rev. Immunol. 10, 753–766 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Thomson, A. W., Vionnet, J. & Sanchez-Fueyo, A. Understanding, predicting and achieving liver transplant tolerance: from bench to bedside. Nat. Rev. Gastroenterol. Hepatol. 17, 719–739 (2020).

    Article  CAS  PubMed  Google Scholar 

  27. Krueger, P. D., Kim, T. S., Sung, S. S., Braciale, T. J. & Hahn, Y. S. Liver-resident CD103+ dendritic cells prime antiviral CD8+ T cells in situ. J. Immunol. 194, 3213–3222 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Deczkowska, A. et al. XCR1+ type 1 conventional dendritic cells drive liver pathology in non-alcoholic steatohepatitis. Nat. Med. 27, 1043–1054 (2021).

    Article  CAS  PubMed  Google Scholar 

  29. Peng, H. et al. Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J. Clin. Invest. 123, 1444–1456 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Paust, S. et al. Critical role for the chemokine receptor CXCR6 in NK cell-mediated antigen-specific memory of haptens and viruses. Nat. Immunol. 11, 1127–1135 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lassen, M. G., Lukens, J. R., Dolina, J. S., Brown, M. G. & Hahn, Y. S. Intrahepatic IL-10 maintains NKG2A+Ly49 liver NK cells in a functionally hyporesponsive state. J. Immunol. 184, 2693–2701 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. 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).

    Article  CAS  PubMed  Google Scholar 

  33. Gabrilovich, D. I. & Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 9, 162–174 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Curiel, T. J. Tregs and rethinking cancer immunotherapy. J. Clin. Invest. 117, 1167–1174 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Diehl, L. et al. Tolerogenic maturation of liver sinusoidal endothelial cells promotes B7-homolog 1-dependent CD8+ T cell tolerance. Hepatology 47, 296–305 (2008).

    Article  CAS  PubMed  Google Scholar 

  36. Limmer, A. et al. Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance. Nat. Med. 6, 1348–1354 (2000).

    Article  CAS  PubMed  Google Scholar 

  37. Aizarani, N. et al. A human liver cell atlas reveals heterogeneity and epithelial progenitors. Nature 572, 199–204 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Peng, W. C. et al. Inflammatory cytokine TNFα promotes the long-term expansion of primary hepatocytes in 3D culture. Cell 175, 1607–1619.e15 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Berndt, N. et al. Functional consequences of metabolic zonation in murine livers: insights for an old story. Hepatology 73, 795–810 (2021).

    Article  CAS  PubMed  Google Scholar 

  40. Halpern, K. B. et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 542, 352–356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Droin, C. et al. Space–time logic of liver gene expression at sub-lobular scale. Nat. Metab. 3, 43–58 (2021). This article reveals how liver function is compartmentalized spatiotemporally at the sub-lobular scale.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Planas-Paz, L. et al. The RSPO–LGR4/5–ZNRF3/RNF43 module controls liver zonation and size. Nat. Cell Biol. 18, 467–479 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Inverso, D. et al. A spatial vascular transcriptomic, proteomic, and phosphoproteomic atlas unveils an angiocrine Tie–Wnt signaling axis in the liver. Dev. Cell 56, 1677–1693.e10 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Keegan, A., Martini, R. & Batey, R. Ethanol-related liver injury in the rat: a model of steatosis, inflammation and pericentral fibrosis. J. Hepatol. 23, 591–600 (1995).

    Article  CAS  PubMed  Google Scholar 

  45. Hall, Z. et al. Lipid zonation and phospholipid remodeling in nonalcoholic fatty liver disease. Hepatology 65, 1165–1180 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Michelotti, G. A., Machado, M. V. & Diehl, A. M. NAFLD, NASH and liver cancer. Nat. Rev. Gastroenterol. Hepatol. 10, 656–665 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Lackner, C. Hepatocellular ballooning in nonalcoholic steatohepatitis: the pathologist’s perspective. Expert Rev. Gastroenterol. Hepatol. 5, 223–231 (2011).

    Article  PubMed  Google Scholar 

  48. Malehmir, M. et al. Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. 25, 641–655 (2019). This paper provides a rationale for antiplatelet therapy in reverting NASH and preventing NASH-to-HCC transition.

    Article  CAS  PubMed  Google Scholar 

  49. 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).

    Article  CAS  PubMed  Google Scholar 

  50. Kietzmann, T. Liver zonation in health and disease: hypoxia and hypoxia-inducible transcription factors as concert masters. Int. J. Mol. Sci. 20, 2347 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  51. Kudo, Y. et al. PKCλ/ι loss induces autophagy, oxidative phosphorylation, and NRF2 to promote liver cancer progression. Cancer Cell 38, 247–262.e11 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Wilson, G. K., Tennant, D. A. & McKeating, J. A. Hypoxia inducible factors in liver disease and hepatocellular carcinoma: current understanding and future directions. J. Hepatol. 61, 1397–1406 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. Wing, P. A. C. et al. Hypoxia inducible factors regulate hepatitis B virus replication by activating the basal core promoter. J. Hepatol. 75, 64–73 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Moreau, M. et al. Hepatitis C viral proteins perturb metabolic liver zonation. J. Hepatol. 62, 278–285 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Gautheron, J. et al. A positive feedback loop between RIP3 and JNK controls non-alcoholic steatohepatitis. EMBO Mol. Med. 6, 1062–1074 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. 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.e10 (2017). This article illustrates diverging mechanistic links of caspase 8 to cancer biology in the liver.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Holze, C. et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat. Immunol. 19, 130–140 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Seehawer, M. et al. Necroptosis microenvironment directs lineage commitment in liver cancer. Nature 562, 69–75 (2018). This article sheds light on lineage commitment during liver carcinogenesis and elucidates the molecular basis for common liver-damaging risk factors that result in either HCC or CCA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Faubion, W. A. et al. Toxic bile salts induce rodent hepatocyte apoptosis via direct activation of Fas. J. Clin. Invest. 103, 137–145 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Brenner, C., Galluzzi, L., Kepp, O. & Kroemer, G. Decoding cell death signals in liver inflammation. J. Hepatol. 59, 583–594 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Asselah, T. et al. In vivo hepatic endoplasmic reticulum stress in patients with chronic hepatitis C. J. Pathol. 221, 264–274 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Merquiol, E. et al. HCV causes chronic endoplasmic reticulum stress leading to adaptation and interference with the unfolded protein response. PLoS ONE 6, e24660 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Roos, W. P., Thomas, A. D. & Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 16, 20–33 (2016).

    Article  CAS  PubMed  Google Scholar 

  64. Brumatti, G., Salmanidis, M. & Ekert, P. G. Crossing paths: interactions between the cell death machinery and growth factor survival signals. Cell Mol. Life Sci. 67, 1619–1630 (2010).

    Article  CAS  PubMed  Google Scholar 

  65. Farazi, P. A. & DePinho, R. A. Hepatocellular carcinoma pathogenesis: from genes to environment. Nat. Rev. Cancer 6, 674–687 (2006).

    Article  CAS  PubMed  Google Scholar 

  66. Roychowdhury, S., McMullen, M. R., Pisano, S. G., Liu, X. & Nagy, L. E. Absence of receptor interacting protein kinase 3 prevents ethanol-induced liver injury. Hepatology 57, 1773–1783 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Linkermann, A., Stockwell, B. R., Krautwald, S. & Anders, H. J. Regulated cell death and inflammation: an auto-amplification loop causes organ failure. Nat. Rev. Immunol. 14, 759–767 (2014).

    Article  CAS  PubMed  Google Scholar 

  68. Newton, K. et al. Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis. Nature 574, 428–431 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Upton, J. W., Kaiser, W. J. & Mocarski, E. S. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 11, 290–297 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li, J. et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Murphy, J. M. et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 (2013).

    Article  CAS  PubMed  Google Scholar 

  72. Malhi, H., Gores, G. J. & Lemasters, J. J. Apoptosis and necrosis in the liver: a tale of two deaths? Hepatology 43, S31–S44 (2006).

    Article  CAS  PubMed  Google Scholar 

  73. Caesar, R. et al. Gut-derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut 61, 1701–1707 (2012).

    Article  CAS  PubMed  Google Scholar 

  74. Sutti, S. et al. Adaptive immune responses triggered by oxidative stress contribute to hepatic inflammation in NASH. Hepatology 59, 886–897 (2014).

    Article  CAS  PubMed  Google Scholar 

  75. Sutti, S. & Albano, E. Adaptive immunity: an emerging player in the progression of NAFLD. Nat. Rev. Gastroenterol. Hepatol. 17, 81–92 (2020).

    Article  CAS  PubMed  Google Scholar 

  76. Wang, H. et al. TNF-α/IFN-γ profile of HBV-specific CD4 T cells is associated with liver damage and viral clearance in chronic HBV infection. J. Hepatol. 72, 45–56 (2020).

    Article  CAS  PubMed  Google Scholar 

  77. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, cancer. Cell 140, 883–899 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kopp, J. L., Grompe, M. & Sander, M. Stem cells versus plasticity in liver and pancreas regeneration. Nat. Cell Biol. 18, 238–245 (2016).

    Article  PubMed  CAS  Google Scholar 

  79. Yanger, K. et al. Robust cellular reprogramming occurs spontaneously during liver regeneration. Genes Dev. 27, 719–724 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Sakurai, T. et al. Hepatocyte necrosis induced by oxidative stress and IL-1α release mediate carcinogen-induced compensatory proliferation and liver tumorigenesis. Cancer Cell 14, 156–165 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Flecken, T. et al. Immunodominance and functional alterations of tumor-associated antigen-specific CD8+ T-cell responses in hepatocellular carcinoma. Hepatology 59, 1415–1426 (2014).

    Article  CAS  PubMed  Google Scholar 

  82. Macdonald, R. Lifespan of liver cells — autoradiographic study using tritiated thymidine in normal, cirrhotic, and partially hepatectomized rats. Arch. Intern. Med. 107, 335–343 (1961).

    Article  CAS  PubMed  Google Scholar 

  83. Karin, M. & Clevers, H. Reparative inflammation takes charge of tissue regeneration. Nature 529, 307–315 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Pikarsky, E. et al. NF-κB functions as a tumour promoter in inflammation-associated cancer. Nature 431, 461–466 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Finkin, S. et al. Ectopic lymphoid structures function as microniches for tumor progenitor cells in hepatocellular carcinoma. Nat. Immunol. 16, 1235–1244 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Takai, A. et al. A novel mouse model of hepatocarcinogenesis triggered by AID causing deleterious p53 mutations. Oncogene 28, 469–478 (2009).

    Article  CAS  PubMed  Google Scholar 

  89. Marrogi, A. J. et al. Oxidative stress and p53 mutations in the carcinogenesis of iron overload-associated hepatocellular carcinoma. J. Natl Cancer Inst. 93, 1652–1655 (2001).

    Article  CAS  PubMed  Google Scholar 

  90. Elinav, E. et al. Inflammation-induced cancer: crosstalk between tumours, immune cells and microorganisms. Nat. Rev. Cancer 13, 759–771 (2013).

    Article  CAS  PubMed  Google Scholar 

  91. Stauffer, J. K., Scarzello, A. J., Jiang, Q. & Wiltrout, R. H. Chronic inflammation, immune escape, and oncogenesis in the liver: a unique neighborhood for novel intersections. Hepatology 56, 1567–1574 (2012).

    Article  CAS  PubMed  Google Scholar 

  92. Grohmann, M. et al. Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC. Cell 175, 1289–1306.e20 (2018). This article demonstrates that obesity-related hepatic oxidative stress might contribute independently to the aetiology of NASH, fibrosis and HCC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Xue, R. et al. Genomic and transcriptomic profiling of combined hepatocellular and intrahepatic cholangiocarcinoma reveals distinct molecular subtypes. Cancer Cell 35, 932–947.e8 (2019). This paper demonstrates that combined and mixed forms of HCC–CCA exhibit distinct clinical and molecular characteristics and identifies Nestin as a potential marker for diagnosis of HCC–CCA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Mu, X. et al. Hepatocellular carcinoma originates from hepatocytes and not from the progenitor/biliary compartment. J. Clin. Invest. 125, 3891–3903 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Sia, D., Villanueva, A., Friedman, S. L. & Llovet, J. M. Liver cancer cell of origin, molecular class, and effects on patient prognosis. Gastroenterology 152, 745–761 (2017).

    Article  CAS  PubMed  Google Scholar 

  96. Ko, S., Russell, J. O., Molina, L. M. & Monga, S. P. Liver progenitors and adult cell plasticity in hepatic injury and repair: knowns and unknowns. Annu. Rev. Pathol. 15, 23–50 (2020).

    Article  CAS  PubMed  Google Scholar 

  97. Marquardt, J. U., Andersen, J. B. & Thorgeirsson, S. S. Functional and genetic deconstruction of the cellular origin in liver cancer. Nat. Rev. Cancer 15, 653–667 (2015).

    Article  CAS  PubMed  Google Scholar 

  98. Okabe, H. et al. Wnt signaling regulates hepatobiliary repair following cholestatic liver injury in mice. Hepatology 64, 1652–1666 (2016).

    Article  CAS  PubMed  Google Scholar 

  99. Hyun, J. et al. Dysregulated activation of fetal liver programme in acute liver failure. Gut 68, 1076–1087 (2019).

    Article  CAS  PubMed  Google Scholar 

  100. Komuta, M. et al. Histological diversity in cholangiocellular carcinoma reflects the different cholangiocyte phenotypes. Hepatology 55, 1876–1888 (2012).

    Article  CAS  PubMed  Google Scholar 

  101. Banales, J. M. et al. Expert consensus document: cholangiocarcinoma: current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat. Rev. Gastroenterol. Hepatol. 13, 261–280 (2016).

    Article  PubMed  Google Scholar 

  102. Komuta, M. et al. Clinicopathological study on cholangiolocellular carcinoma suggesting hepatic progenitor cell origin. Hepatology 47, 1544–1556 (2008).

    Article  CAS  PubMed  Google Scholar 

  103. Brunt, E. et al. cHCC-CCA: consensus terminology for primary liver carcinomas with both hepatocytic and cholangiocytic differentation. Hepatology 68, 113–126 (2018).

    Article  PubMed  Google Scholar 

  104. Satriano, L., Lewinska, M., Rodrigues, P. M., Banales, J. M. & Andersen, J. B. Metabolic rearrangements in primary liver cancers: cause and consequences. Nat. Rev. Gastroenterol. Hepatol. 16, 748–766 (2019). This review discusses the impact of metabolic liver disturbances and the effects on primary liver cancer.

    Article  CAS  PubMed  Google Scholar 

  105. Yamashita, T. et al. Activation of lipogenic pathway correlates with cell proliferation and poor prognosis in hepatocellular carcinoma. J. Hepatol. 50, 100–110 (2009).

    Article  CAS  PubMed  Google Scholar 

  106. Wang, M. et al. Dysregulated fatty acid metabolism in hepatocellular carcinoma. Hepat. Oncol. 3, 241–251 (2016).

    Article  PubMed  Google Scholar 

  107. Xia, S., Pan, Y., Liang, Y., Xu, J. & Cai, X. The microenvironmental and metabolic aspects of sorafenib resistance in hepatocellular carcinoma. EBioMedicine 51, 102610 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Ma, M. K. F. et al. Stearoyl-CoA desaturase regulates sorafenib resistance via modulation of ER stress-induced differentiation. J. Hepatol. 67, 979–990 (2017).

    Article  CAS  PubMed  Google Scholar 

  109. Rudalska, R. et al. LXRα activation and Raf inhibition trigger lethal lipotoxicity in liver cancer. Nat. Cancer 2, 201–217 (2021).

    Article  PubMed  Google Scholar 

  110. Lally, J. S. V. et al. Inhibition of acetyl-CoA carboxylase by phosphorylation or the inhibitor ND-654 suppresses lipogenesis and hepatocellular carcinoma. Cell Metab. 29, 174–182.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  111. Saha, S. K. et al. Mutant IDH inhibits HNF-4α to block hepatocyte differentiation and promote biliary cancer. Nature 513, 110–114 (2014). This article establishes a functional relationship between IDH mutations and CCA pathogenesis, and provides a novel genetically engineered mouse model of IDH-driven malignancy.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Saha, S. K. et al. Isocitrate dehydrogenase mutations confer dasatinib hypersensitivity and SRC dependence in intrahepatic cholangiocarcinoma. Cancer Discov. 6, 727–739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wu, E. M. et al. Gender differences in hepatocellular cancer: disparities in nonalcoholic fatty liver disease/steatohepatitis and liver transplantation. Hepatoma Res. 4, 66 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Manieri, E. et al. Adiponectin accounts for gender differences in hepatocellular carcinoma incidence. J. Exp. Med. 216, 1108–1119 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Ma, C. et al. NAFLD causes selective CD4+ T lymphocyte loss and promotes hepatocarcinogenesis. Nature 531, 253–257 (2016). The findings from this article, for the first time, connect lipid dysregulation with decreased antitumour surveillance in CD4+ T cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Zhang, Q. et al. Fatty acid oxidation contributes to IL-1β secretion in M2 macrophages and promotes macrophage-mediated tumor cell migration. Mol. Immunol. 94, 27–35 (2018).

    Article  PubMed  CAS  Google Scholar 

  117. Pacella, I. et al. Fatty acid metabolism complements glycolysis in the selective regulatory T cell expansion during tumor growth. Proc. Natl Acad. Sci. USA 115, E6546–E6555 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Li, T.-E. et al. PKM2 drives hepatocellular carcinoma progression by inducing immunosuppressive microenvironment. Front. Immunol. 11, 2722 (2020).

    Article  Google Scholar 

  119. Chen, D. P. et al. Glycolytic activation of peritumoral monocytes fosters immune privilege via the PFKFB3–PD-L1 axis in human hepatocellular carcinoma. J. Hepatol. 71, 333–343 (2019).

    Article  CAS  PubMed  Google Scholar 

  120. Klein, G. Cancer, apoptosis, and nonimmune surveillance. Cell Death Differ. 11, 13–17 (2004).

    Article  CAS  PubMed  Google Scholar 

  121. Dhar, D. et al. Liver cancer initiation requires p53 inhibition by CD44-enhanced growth factor signaling. Cancer Cell 33, 1061–1077.e6 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Tschaharganeh, D. F. et al. p53-Dependent Nestin regulation links tumor suppression to cellular plasticity in liver cancer. Cell 158, 579–592 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  124. Mizukoshi, E. et al. Comparative analysis of various tumor-associated antigen-specific T-cell responses in patients with hepatocellular carcinoma. Hepatology 53, 1206–1216 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Endig, J. et al. Dual role of the adaptive immune system in liver injury and hepatocellular carcinoma development. Cancer Cell 30, 308–323 (2016).

    Article  CAS  PubMed  Google Scholar 

  126. Fu, J. et al. Impairment of CD4+ cytotoxic T cells predicts poor survival and high recurrence rates in patients with hepatocellular carcinoma. Hepatology 58, 139–149 (2013).

    Article  CAS  PubMed  Google Scholar 

  127. Garnelo, M. et al. Interaction between tumour-infiltrating B cells and T cells controls the progression of hepatocellular carcinoma. Gut 66, 342–351 (2017).

    Article  CAS  PubMed  Google Scholar 

  128. Chen, K. J. et al. Selective recruitment of regulatory T cell through CCR6–CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. PLoS ONE 6, e24671 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. 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).

    Article  PubMed  Google Scholar 

  130. Gallimore, A. M. & Simon, A. K. Positive and negative influences of regulatory T cells on tumour immunity. Oncogene 27, 5886–5893 (2008).

    Article  CAS  PubMed  Google Scholar 

  131. Ghiringhelli, F., Menard, C., Martin, F. & Zitvogel, L. The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression. Immunol. Rev. 214, 229–238 (2006).

    Article  CAS  PubMed  Google Scholar 

  132. Sun, Y. et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 184, 404–421.e16 (2021). This paper uses single-cell analysis of primary and recurrent HCC tumours to produce deep insights into immune evasion mechanisms.

    Article  CAS  PubMed  Google Scholar 

  133. Dudek, M. et al. Auto-aggressive CXCR6+ CD8 T cells cause liver immune pathology in NASH. Nature 592, 444–449 (2021). This article reveals that the mechanisms behind auto-aggression by CD8+ T cells are separate from those of antigen-specific killing by CD8+ T cells.

    Article  CAS  PubMed  Google Scholar 

  134. Heinrich, B. et al. Steatohepatitis impairs T-cell-directed immunotherapies against liver tumors in mice. Gastroenterology 160, 331–345.e6 (2021).

    Article  CAS  PubMed  Google Scholar 

  135. Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, 450–456 (2021). This paper supports stratifying patients with HCC according to their underlying aetiology, before using immunotherapy as a main or adjuvant treatment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Albertsson, P. A. et al. NK cells and the tumour microenvironment: implications for NK-cell function and anti-tumour activity. Trends Immunol. 24, 603–609 (2003).

    Article  CAS  PubMed  Google Scholar 

  137. Bricard, G. et al. Enrichment of human CD4+ Vα24/Vβ11 invariant NKT cells in intrahepatic malignant tumors. J. Immunol. 182, 5140–5151 (2009).

    Article  CAS  PubMed  Google Scholar 

  138. Crowe, N. Y. et al. Differential antitumor immunity mediated by NKT cell subsets in vivo. J. Exp. Med. 202, 1279–1288 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Pommier, A. et al. Inflammatory monocytes are potent antitumor effectors controlled by regulatory CD4+ T cells. Proc. Natl Acad. Sci. USA 110, 13085–13090 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Lee, J. W. et al. Hepatocytes direct the formation of a pro-metastatic niche in the liver. Nature 567, 249–252 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Condeelis, J. & Pollard, J. W. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124, 263–266 (2006).

    Article  CAS  PubMed  Google Scholar 

  142. Noy, R. & Pollard, J. W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ramachandran, P. et al. Resolving the fibrotic niche of human liver cirrhosis at single-cell level. Nature 575, 512–518 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Budhu, A. et al. Prediction of venous metastases, recurrence, and prognosis in hepatocellular carcinoma based on a unique immune response signature of the liver microenvironment. Cancer Cell 10, 99–111 (2006).

    Article  CAS  PubMed  Google Scholar 

  145. Zhu, X. D. et al. High expression of macrophage colony-stimulating factor in peritumoral liver tissue is associated with poor survival after curative resection of hepatocellular carcinoma. J. Clin. Oncol. 26, 2707–2716 (2008).

    Article  PubMed  Google Scholar 

  146. Ciner, A. T., Jones, K., Muschel, R. J. & Brodt, P. The unique immune microenvironment of liver metastases: challenges and opportunities. Semin. Cancer Biol. 71, 143–156 (2021).

    Article  CAS  PubMed  Google Scholar 

  147. Yu, J. et al. Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination. Nat. Med. 27, 152–164 (2021). This paper shows that hepatic metastases utilize host peripheral tolerance mechanisms to promote CD8+ T cell depletion.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Fidler, I. J. Timeline — the pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat. Rev. Cancer 3, 453–458 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  150. Clark, A. M., Ma, B., Taylor, D. L., Griffith, L. & Wells, A. Liver metastases: microenvironments and ex-vivo models. Exp. Biol. Med. 241, 1639–1652 (2016).

    Article  CAS  Google Scholar 

  151. Benedicto, A., Romayor, I. & Arteta, B. Role of liver ICAM-1 in metastasis. Oncol. Lett. 14, 3883–3892 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  152. Khatib, A. M. et al. Rapid induction of cytokine and E-selectin expression in the liver in response to metastatic tumor cells. Cancer Res. 59, 1356–1361 (1999).

    CAS  PubMed  Google Scholar 

  153. Brodt, P. Role of the microenvironment in liver metastasis: from pre- to prometastatic niches. Clin. Cancer Res. 22, 5971–5982 (2016).

    Article  CAS  PubMed  Google Scholar 

  154. Wohlfeil, S. A. et al. Hepatic endothelial notch activation protects against liver metastasis by regulating endothelial-tumor cell adhesion independent of angiocrine signaling. Cancer Res. 79, 598–610 (2019).

    Article  CAS  PubMed  Google Scholar 

  155. Mendoza, L. et al. Hydrogen peroxide mediates vascular cell adhesion molecule-1 expression from interleukin-18-activated hepatic sinusoidal endothelium: implications for circulating cancer cell arrest in the murine liver. Hepatology 34, 298–310 (2001).

    Article  CAS  PubMed  Google Scholar 

  156. Hu, C. T. et al. MIF, secreted by human hepatic sinusoidal endothelial cells, promotes chemotaxis and outgrowth of colorectal cancer in liver prometastasis. Oncotarget 6, 22410–22423 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  157. Ou, J. et al. Endothelial cell-derived fibronectin extra domain A promotes colorectal cancer metastasis via inducing epithelial–mesenchymal transition. Carcinogenesis 35, 1661–1670 (2014).

    Article  CAS  PubMed  Google Scholar 

  158. Huang, J., Pan, C., Hu, H., Zheng, S. & Ding, L. Osteopontin-enhanced hepatic metastasis of colorectal cancer cells. PLoS ONE 7, e47901 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Tabaries, S. et al. Claudin-2 promotes breast cancer liver metastasis by facilitating tumor cell interactions with hepatocytes. Mol. Cell Biol. 32, 2979–2991 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  160. Shimizu, S. et al. Ultrastructure of early phase hepatic metastasis of human colon carcinoma cells with special reference to desmosomal junctions with hepatocytes. Pathol. Int. 50, 953–959 (2000).

    Article  CAS  PubMed  Google Scholar 

  161. Yoshioka, T. et al. Significance of integrin αvβ5 and erbB3 in enhanced cell migration and liver metastasis of colon carcinomas stimulated by hepatocyte-derived heregulin. Cancer Sci. 101, 2011–2018 (2010).

    Article  CAS  PubMed  Google Scholar 

  162. Bu, P. et al. Aldolase B-mediated fructose metabolism drives metabolic reprogramming of colon cancer liver metastasis. Cell Metab. 27, 1249–1262.e4 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Li, Y. et al. Hepatic lipids promote liver metastasis. JCI Insight 5, e136215 (2020).

    Article  PubMed Central  Google Scholar 

  164. Wen, S. W., Ager, E. I. & Christophi, C. Bimodal role of Kupffer cells during colorectal cancer liver metastasis. Cancer Biol. Ther. 14, 606–613 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Costa-Silva, B. et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 17, 816–826 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Shao, Y. et al. Colorectal cancer-derived small extracellular vesicles establish an inflammatory premetastatic niche in liver metastasis. Carcinogenesis 39, 1368–1379 (2018).

    Article  CAS  PubMed  Google Scholar 

  168. Bhattacharjee, S. et al. Tumor restriction by type I collagen opposes tumor-promoting effects of cancer-associated fibroblasts. J. Clin. Invest. 131, e146987 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  169. Iredale, J. P., Thompson, A. & Henderson, N. C. Extracellular matrix degradation in liver fibrosis: biochemistry and regulation. Biochim. Biophys. Acta 1832, 876–883 (2013).

    Article  CAS  PubMed  Google Scholar 

  170. Friedman, S. L. Seminars in medicine of the Beth Israel Hospital, Boston. The cellular basis of hepatic fibrosis. Mechanisms and treatment strategies. N. Engl. J. Med. 328, 1828–1835 (1993).

    Article  CAS  PubMed  Google Scholar 

  171. Eveno, C. et al. Proof of prometastatic niche induction by hepatic stellate cells. J. Surg. Res. 194, 496–504 (2015).

    Article  PubMed  Google Scholar 

  172. Cox, D., Brennan, M. & Moran, N. Integrins as therapeutic targets: lessons and opportunities. Nat. Rev. Drug Discov. 9, 804–820 (2010).

    Article  CAS  PubMed  Google Scholar 

  173. Zhang, D. Y. & Friedman, S. L. Fibrosis-dependent mechanisms of hepatocarcinogenesis. Hepatology 56, 769–775 (2012).

    Article  CAS  PubMed  Google Scholar 

  174. Shen, Y. et al. Reduction of liver metastasis stiffness improves response to bevacizumab in metastatic colorectal cancer. Cancer Cell 37, 800–817.e7 (2020).

    Article  CAS  PubMed  Google Scholar 

  175. Zhao, W. et al. Hepatic stellate cells promote tumor progression by enhancement of immunosuppressive cells in an orthotopic liver tumor mouse model. Lab. Invest. 94, 182–191 (2014).

    Article  CAS  PubMed  Google Scholar 

  176. Hsu, B. E. et al. Immature low-density neutrophils exhibit metabolic flexibility that facilitates breast cancer liver metastasis. Cell Rep. 27, 3902–3915.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  177. Gordon-Weeks, A. N. et al. Neutrophils promote hepatic metastasis growth through fibroblast growth factor 2-dependent angiogenesis in mice. Hepatology 65, 1920–1935 (2017).

    Article  CAS  PubMed  Google Scholar 

  178. Haemmerle, M., Stone, R. L., Menter, D. G., Afshar-Kharghan, V. & Sood, A. K. The platelet lifeline to cancer: challenges and opportunities. Cancer Cell 33, 965–983 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Verheul, H. M. et al. Vascular endothelial growth factor trap blocks tumor growth, metastasis formation, and vascular leakage in an orthotopic murine renal cell cancer model. Clin. Cancer Res. 13, 4201–4208 (2007).

    Article  CAS  PubMed  Google Scholar 

  180. Gervaz, P. et al. Angiogenesis of liver metastases: role of sinusoidal endothelial cells. Dis. Colon Rectum 43, 980–986 (2000).

    Article  CAS  PubMed  Google Scholar 

  181. Kimura, Y. et al. The innate immune receptor Dectin-2 mediates the phagocytosis of cancer cells by Kupffer cells for the suppression of liver metastasis. Proc. Natl Acad. Sci. USA 113, 14097–14102 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Timmers, M. et al. Interactions between rat colon carcinoma cells and Kupffer cells during the onset of hepatic metastasis. Int. J. Cancer 112, 793–802 (2004).

    Article  CAS  PubMed  Google Scholar 

  183. Molgora, M. et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature 551, 110–114 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  184. Takeda, K. et al. Involvement of tumor necrosis factor-related apoptosis-inducing ligand in surveillance of tumor metastasis by liver natural killer cells. Nat. Med. 7, 94–100 (2001).

    Article  CAS  PubMed  Google Scholar 

  185. Ballas, Z. K., Buchta, C. M., Rosean, T. R., Heusel, J. W. & Shey, M. R. Role of NK cell subsets in organ-specific murine melanoma metastasis. PLoS ONE 8, e65599 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Ducimetière, L. et al. Conventional NK cells and tissue-resident ILC1s join forces to control liver metastasis. Proc. Natl Acad. Sci. USA 118, e2026271118 (2021).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  187. Tauriello, D. V. F. et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554, 538–543 (2018).

    Article  CAS  PubMed  Google Scholar 

  188. Lee, J. H. et al. Adjuvant immunotherapy with autologous cytokine-induced killer cells for hepatocellular carcinoma. Gastroenterology 148, 1383–1391.e6 (2015).

    Article  CAS  PubMed  Google Scholar 

  189. Lee, J.-H., Oh, S.-Y., Kim, J. Y. & Nishida, N. Cancer immunotherapy for hepatocellular carcinoma. Hepatoma Res. 4, 51 (2018).

    Article  CAS  Google Scholar 

  190. Zhang, Z. et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Crowther, M. D. et al. Genome-wide CRISPR–Cas9 screening reveals ubiquitous T cell cancer targeting via the monomorphic MHC class I-related protein MR1. Nat. Immunol. 21, 178–185 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Sangro, B. et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J. Hepatol. 59, 81–88 (2013).

    Article  CAS  PubMed  Google Scholar 

  193. Duffy, A. G. et al. Tremelimumab in combination with ablation in patients with advanced hepatocellular carcinoma. J. Hepatol. 66, 545–551 (2017).

    Article  CAS  PubMed  Google Scholar 

  194. Finn, R. S. et al. Atezolizumab plus bevacizumab in unresectable hepatocellular carcinoma. N. Engl. J. Med. 382, 1894–1905 (2020). This article reports an improvement of overall survival and progression-free survival in patients with advanced unresectable HCC who were treated with atezolizumab and bevacizumab when compared with sorafenib.

    Article  CAS  PubMed  Google Scholar 

  195. Llovet, J. M. et al. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2, 16018 (2016).

    Article  PubMed  Google Scholar 

  196. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Yarchoan, M. et al. Recent developments and therapeutic strategies against hepatocellular carcinoma. Cancer Res. 79, 4326–4330 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Harding, J. J. et al. Prospective genotyping of hepatocellular carcinoma: clinical implications of next-generation sequencing for matching patients to targeted and immune therapies. Clin. Cancer Res. 25, 2116–2126 (2019).

    Article  CAS  PubMed  Google Scholar 

  199. Jin, H. et al. A powerful drug combination strategy targeting glutamine addiction for the treatment of human liver cancer. eLife 9, e56749 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Rizvi, S. & Gores, G. J. Pathogenesis, diagnosis, and management of cholangiocarcinoma. Gastroenterology 145, 1215–1229 (2013).

    Article  CAS  PubMed  Google Scholar 

  201. Rothwell, P. M. et al. Effects of aspirin on risks of vascular events and cancer according to bodyweight and dose: analysis of individual patient data from randomised trials. Lancet 392, 387–399 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Simon, T. G. et al. Association of aspirin with hepatocellular carcinoma and liver-related mortality. N. Engl. J. Med. 382, 1018–1028 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Xiong, X. et al. Landscape of intercellular crosstalk in healthy and NASH liver revealed by single-cell secretome gene analysis. Mol. Cell 75, 644–660.e5 (2019). This article reports unprecedented insights into intercellular interaction and reprogramming of liver cells in health and disease.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Gola, A. et al. Commensal-driven immune zonation of the liver promotes host defence. Nature 589, 131–136 (2021). This paper explains the link between the localization of hepatic immune cells and host protection.

    Article  CAS  PubMed  Google Scholar 

  205. Manco, R. & Itzkovitz, S. Liver zonation. J. Hepatol. 74, 466–468 (2021).

    Article  CAS  PubMed  Google Scholar 

  206. Krenkel, O. et al. Myeloid cells in liver and bone marrow acquire a functionally distinct inflammatory phenotype during obesity-related steatohepatitis. Gut 69, 551–563 (2020).

    Article  CAS  PubMed  Google Scholar 

  207. MacParland, S. A. et al. Single cell RNA sequencing of human liver reveals distinct intrahepatic macrophage populations. Nat. Commun. 9, 4383 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  208. Dobie, R. et al. Single-cell transcriptomics uncovers zonation of function in the mesenchyme during liver fibrosis. Cell Rep. 29, 1832–1847.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Verfaillie, T. et al. PERK is required at the ER–mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death Differ. 19, 1880–1891 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Liu, J. et al. Endoplasmic reticulum stress modulates liver inflammatory immune response in the pathogenesis of liver ischemia and reperfusion injury. Transplantation 94, 211–217 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Ren, F. et al. Endoplasmic reticulum stress-activated glycogen synthase kinase 3β aggravates liver inflammation and hepatotoxicity in mice with acute liver failure. Inflammation 38, 1151–1165 (2015).

    Article  CAS  PubMed  Google Scholar 

  212. Wei, C. et al. Tumor microenvironment regulation by the endoplasmic reticulum stress transmission mediator Golgi protein 73 in mice. Hepatology 70, 851–870 (2019).

    Article  CAS  PubMed  Google Scholar 

  213. Enzan, H. et al. α-Smooth muscle actin-positive perisinusoidal stromal cells in human hepatocellular carcinoma. Hepatology 19, 895–903 (1994).

    CAS  PubMed  Google Scholar 

  214. Seitz, H. K. & Stickel, F. Risk factors and mechanisms of hepatocarcinogenesis with special emphasis on alcohol and oxidative stress. Biol. Chem. 387, 349–360 (2006).

    Article  CAS  PubMed  Google Scholar 

  215. Heindryckx, F., Colle, I. & Van Vlierberghe, H. Experimental mouse models for hepatocellular carcinoma research. Int. J. Exp. Pathol. 90, 367–386 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Carloni, V., Luong, T. V. & Rombouts, K. Hepatic stellate cells and extracellular matrix in hepatocellular carcinoma: more complicated than ever. Liver Int. 34, 834–843 (2014).

    Article  PubMed  Google Scholar 

  217. Baglieri, J., Brenner, D. A. & Kisseleva, T. The role of fibrosis and liver-associated fibroblasts in the pathogenesis of hepatocellular carcinoma. Int. J. Mol. Sci. 20, 1723 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  218. Rosenberg, W. M. et al. Serum markers detect the presence of liver fibrosis: a cohort study. Gastroenterology 127, 1704–1713 (2004).

    Article  PubMed  Google Scholar 

  219. Wu, Y., Qiao, X., Qiao, S. & Yu, L. Targeting integrins in hepatocellular carcinoma. Expert. Opin. Ther. Targets 15, 421–437 (2011).

    Article  CAS  PubMed  Google Scholar 

  220. Dhar, D., Baglieri, J., Kisseleva, T. & Brenner, D. A. Mechanisms of liver fibrosis and its role in liver cancer. Exp. Biol. Med. 245, 96–108 (2020).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank S. Gallage for his critical proofreading of the manuscript. M.H. was supported by an European Research Council (ERC) Consolidator grant (HepatoMetaboPath), SFBTR179 project ID 272983813, SFB/TR 209 project ID 314905040, SFBTR1335 project ID 360372040, SFB 1479 (Project ID: 441891347), the Wilhelm Sander-Stiftung, the Rainer Hoenig Stiftung, a Horizon 2020 grant (Hepcar), Research Foundation Flanders (FWO) under grant 30826052 (EOS Convention MODEL-IDI), Deutsche Krebshilfe projects 70113166 and 70113167, German-Israeli Cooperation in Cancer Research (DKFZ-MOST) and the Helmholtz-Gemeinschaft, Zukunftsthema ‘Immunology and Inflammation’ (ZT-0027).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to all aspects of the article.

Corresponding author

Correspondence to Mathias Heikenwalder.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Cancer thanks J. Fan, J. Moscat and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Necro-inflammation

A pathological condition characterized by cell death (necrosis) that triggers the activation of the immune system, thereby sustaining a local inflammatory response.

Liver sinusoids

Specific capillaries characterized by a particular distribution of endothelial cells presenting a typical fenestration that enables arterial and venous blood to mix.

Steatosis

Abnormal cytosolic accumulation of lipids in more than 5% of the total hepatocyte content in the liver.

Nonalcoholic fatty liver disease

(NAFLD). Metabolic disease of the liver related to abnormal accumulation of lipids, encompassing a wide spectrum of pathologies from simple fatty liver to nonalcoholic steatohepatitis (characterized by hepatic inflammatory infiltrate (steatohepatitis)), fibrosis and cirrhosis.

Natural killer T cells

A heterogeneous population of T cells that share characteristics of classical T cells and natural killer cells. Their classical function relates to antibacterial activity; upon activation, they produce large amounts of interferon-γ (IFNγ), IL-4 and many other cytokines.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, X., Ramadori, P., Pfister, D. et al. The immunological and metabolic landscape in primary and metastatic liver cancer. Nat Rev Cancer 21, 541–557 (2021). https://doi.org/10.1038/s41568-021-00383-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-021-00383-9

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